Вы находитесь на странице: 1из 73

Volume III

ROADMAP TO GMP COMPLIANCE

A division of UBM Americas


A Roadmap to GMP
COMpliance PART 3
Quality Risk Management: State of the IndustryPart 1. Has the Industry Realized
the Full Value of ICH Q9? | Anne Greene, Kelly Waldron, and Nuala Calnan........................................................... 4

Mergers & Acquisitions: Managing GxP Compliance Through Integration and


Organizational Change | Eldon Henson and Whitney Sandberg........................................................................... 12

Structured GMP Audits | Carol Brandt . ............................................................................................................. 18

Moments of Truth: Creating a Quality Culture by Making Good Manufacturing


Practice a Lifestyle in Your Company | David Markovitz . .................................................................................. 23

The FDA Compliance Program Guidance for GLPs - General | Steven S. Kuwarahara ........................................... 26

Quality and Compliance in the Asian Pharmaceutical Industry | David W. Vincent .............................................. 30

Integration of Risk Management Principles into the Quality System: Risk-based


Impact Assessment | Kelly Waldron................................................................................................................... 36

Laboratory Self-Audits | Paul Pluta and Jerry Lanese......................................................................................... 43

Good Manufacturing Practice and Stable Systems - Part One: Reducing Stress
with the Power of Data - Know When to Act and When Not to Act | David Markovitz . ......................................... 54

Implementing an Effective GLP Program at the Contract Laboratory |


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, and N. Knight ............................ 57

GXP Talk: Questions 66 & 67 | Jerry Lanese and Tim Fields............................................................................... 65

GXP Talk: Questions 68 & 69 | Jerry Lanese and Tim Fields............................................................................... 67

GXP Talk: Questions 70 | Jerry Lanese and Tim Fields . ..................................................................................... 69

GXP Talk: Questions 71 & 72 | Jerry Lanese and Tim Fields................................................................................ 71


2017 UBM, LLC All rights reserved.
Reproduction in whole or part is prohibited without prior written permission of the publisher. GMPQ7003
Anne Greene, Kelly Waldron, Nuala Calnan

Quality Risk Management: State


of the IndustryPart 1. Has the
Industry Realized the Full Value
of ICH Q9?
Anne Greene, Kelly Waldron, Nuala Calnan

ABSTRACT
This paper is the first in a series that explores the state of implementa-
tion of Quality Risk Management (QRM) in the pharmaceutical and
biopharmaceutical industries. A brief history of QRM in these indus-
tries with regard to ICH Q9 Quality Risk Management is offered, and
the high-level risk maturity model proposed by the Parenteral Drug
Association (PDA) is described. A comprehensive review of US and
Irish recall data and cGMP-related compliance/enforcement actions was
performed to determine whether those data provide insight into the
maturity of implementation of QRM in these industries and whether
the anticipated benefits of QRM have been realized. Results of this
analysis show that maturity in QRM is lagging and that the benefits of
QRM in terms of improved product quality and patient safety have not
yet been fully harnessed.

INTRODUCTION
the challenge ahead is significant the pharmaceutical community
has arrived at a cross-road; one path goes towards the desired state and
the other maintains the current state. The path towards the desired
state is unfamiliar to many while the current state provides the comfort
of predictability. [We hope] the pharmaceutical community will choose
to move towards the desired state. Food and Drug Administration,
2004 (1) .
Over a decade ago, a paradigm shift in the regulatory environment
surrounding the supply and manufacture of medicines emergeda
transition from traditional rule-based compliance to a modern risk-
based view of compliance. The traditional approach was characterized
by rigid conformity with regulatory statutes that defined the minimum
requirements necessary to ensure medicines were produced with the
appropriate quality, whereas the risk-based view tasked each manu-
facturer with the responsibility to develop a deep product and process
understanding and tailor their quality management systems through a
scientifically sound risk management framework. In the United States,
the initial mobilization towards a risk-based model was championed by
the Food and Drug Administration (FDA) through an initiative known
as Pharmaceutical Current Good Manufacturing Practices (cGMPs) for
the 21st Century. This initiative, announced in August 2002, sought
to enhance and modernize the regulation of pharmaceutical manu-
facturing and product quality by re-envisioning the underpinnings of

4 Special edition: A Roadmap to GMP Compliance Part 3


Anne Greene, Kelly Waldron, Nuala Calnan

compliance through the lens of science and risk. (1) and assurance of medicinal product quality. Q9 notes
that the use of QRM principles and practices has
The final report on this initiative, issued in Sep- the potential to yield multiple benefits for patients,
tember 2004, summarized the key accomplishments regulators, and manufacturers alike. Such benefits
since the 2002 announcement, including the estab- include:
lishment of risk-based inspection planning, change Better assurance of product quality through pro-
review, and dispute resolution within the Agency, active identification and mitigation of potential
among other deliverables. Most notably, the report risks
expressed FDAs desire to improve harmonization Improved compliance by enabling an under-
of regulation through collaboration and knowledge standing of the relationship between regulatory
sharing with sister organizations throughout the requirements and the unique concerns of the
world. (1) Working groups in the International Con- product or process
ference on Harmonization of the Technical Require- More informed and consistent decisions relative
ments for Registration of Pharmaceuticals (ICH) were to product quality (for realized/reactive risks)
tasked with the creation of a risk- and science-based and quality system process design
quality systems model, later to materialize as a triad Potential reduction in the level or frequency of
of guidelines known as ICH Q8 Pharmaceutical De- regulatory oversight through improved commu-
velopment, ICH Q9 Quality Risk Management, and nication and higher confidence in quality system
ICH Q10 Pharmaceutical Quality System. (2, 3, 4) effectiveness (3)
While the final report looked towards the future with This multi-faceted value proposition was both
a sense of optimistic pragmatism, it also acknowl- ambitious and attractive, and the guidance as a whole
edged that further work must be done in order to offered significant insight into the execution and
realize this vision. iterative nature of QRM.
The movement towards a science- and risk-based To further characterize QRM application (as well
approach to cGMPs was advanced with the finaliza- as the concepts laid out in ICH Q8 and Q10), and to
tion of ICH Q9 in 2005, Q10 in 2008, and Q8(R2) in accelerate the learning curve relative to these guid-
2009. ICH Q8 described drug development concepts ances, the Parental Drug Association (PDA) launched
commonly known as Quality by Design (QbD) in the Paradigm Change in Manufacturing Operations
which an intimate scientific understanding of the program in 2008 (5). One of several outputs of this
product and its clinical utility is used, along with risk program was a comprehensive technical report aimed
management techniques, to define critical aspects of at providing a detailed review of how QRM could be
the product and the enabling manufacturing process- implemented throughout the product lifecycle and
es. (2) ICH Q9 focused on defining a lifecycle process supporting quality system. The technical report de-
for quality risk management (QRM), offering a list of scribed the notion of a QRM maturity model, depict-
risk management tools that had seen success when ing the general transition towards maturity associated
applied in other industries and describing potential with QRM program deployment. This QRM maturity
applications of QRM within pharmaceutical develop- model illustrated the expected growth phases as a
ment, manufacturing, and control systems. (3) While manufacturer gained comfort and confidence with
the concepts set forth in Q8 and Q9 showed signifi- QRM:
cant theoretical promise, industry stumbled with the Maturity Phase 1: No quality risk management
application of these guidelines in the absence of an activities performed
overarching framework within which to apply them. Maturity Phase 2: Informal quality risk manage-
The concepts were finally synthesized in ICH Q10, ment in place
which established a quality systems structure to be Maturity Phase 3: Mostly retrospective and/or
applied throughout the product lifecycle, enabled by corrective quality risk management in use
knowledge management and QRM. (4) The remainder Maturity Phase 4: Prospective, preventive quality
of this paper will focus on QRM as described in ICH risk management in use
Q9 and its application in the quality system per ICH Maturity Phase 5: Quality risk management inte-
Q10. grated throughout manufacturing operations and
quality systems (adapted from PDA Technical
ICH Q9 introduced the concept of risk manage- Report No. 54, reference 5)
ment, which had been successfully applied in other The QRM maturity model provided general direction
industries such as aerospace, nuclear energy, and regarding the focus of a firm as they progress towards
finance, to the pharmaceutical and biotechnology a robust QRM programostensibly, with each suc-
arena by focusing on its application in the definition cessive phase of maturity, a QRM program grows

Special edition: A Roadmap to GMP Compliance Part 3 5


Anne Greene, Kelly Waldron, Nuala Calnan

more efficient, effective, and pervasive throughout not categorized according to criticality and, there-
operations and quality systems (5). While the model fore, may not delineate the gravity of noncompliance
can serve as a useful indicator of the maturity of concerns as identified during inspections; in addi-
implementation at a given firm without measurement tion, access to individual Form 483s are not readily
against the model, it does not answer the question available from FDA. Warning letters represent the
of whether we have achieved the value proposition principal means of achieving prompt voluntary com-
offered in ICH Q9. Twelve years after the announce- pliance with applicable regulations and are typically
ment of the 21st century cGMP initiative and almost issued for significant violations of related statutes or,
a decade following the inception of ICH Q9 and these in many cases, inadequate responses or commitments
questions remain: from violative firms following the issuance of a Form
Have we realized the full benefits of QRM? 483 (9). Warning letters are posted to a public access
Are we making better quality products? database and have the potential to offer a rich source
Are we more compliant? of information regarding noncompliance with cGMPs.
This paper describes research undertaken by the Various cross checks (e.g., database searches by year,
Pharmaceutical Regulatory Science Team at the Dub- company name, warning letter category, product
lin Institute of Technology (DIT) in an effort to find type, and keywords) were performed throughout the
the answers. research period to assure the collected data were both
valid and comprehensive.
RESEARCH METHODOLOGY Data from the EU, particularly in the area of inspec-
The research sought to provide a systematic review of tional observations, proved more difficult to retrieve,
US and Irish product recall and compliance enforce- despite allowances offered by the Freedom of Infor-
ment data spanning the 2006 through 2013 time mation Act. As a result, the research focused on Irish
period. This timeframe was selected based on the recall and quality defect data ascertained through
potential to identify trends in product quality and publicly available annual reports written by the
cGMP compliance following the publication of ICH Health Products Regulatory Authority (HPRA; for-
Q9 in 2005, through the most current period in merly the Irish Medicines Board) as well as research
which a comparable data set (i.e. a complete year of assistance for this project provided directly by the
data) was available. HPRA (10, 11). As the worlds largest drug exporter
Data from the US was sourced from the weekly and the hub of the Dublin Institute of Technologys
enforcement report and warning letter databases Pharmaceutical Regulatory Science Team (of which
available through the Food and Drug Administration the authors are part), Ireland is an attractive source
website (6, 7). Drug recall data was reviewed in an ef- for European data (12).
fort to characterize product quality, and by extension All data were categorized and analyzed as discussed
patient safety, over time. In the US, drug recalls are in the Results and Analysis section of this paper.
divided as individual events, each assigned a unique
recall tracking number and associated classification RESULTS AND ANALYSIS
level based on risk to patient or consumer. While Procedure Development and Understanding
most recalls are separated based on product presenta- (Identify and Study Potential Analytical Vari-
tion and dosage form, in certain instances (e.g., where ables)
all products manufactured by a single firm in a given One of the potential benefits of QRM is improved
time period were subject to recall) a single tracking product quality and resultant patient safety; indeed,
number was assigned to a portfolio of products. The one of the primary principles of ICH Q9 is that QRM
data reviewed and presented in this paper retains activities should . . . ultimately link to the protection
the separation and classification as assigned by FDA, of the patient (3). One indicator of trends in this area
including subsequent corrections for previously re- is the number of recalls that may result from inad-
ported data (e.g. expansion of recalled lots). equate quality products reaching the marketplace.
Warning letters issued by FDA for cGMP-related Improvements in product quality, such as the ability
concerns were selected based on the insight they may to meet specifications prior to drug product release,
provide into the industrys state of compliance over should therefore, manifest as a reducing trend in the
time. FDA is charged with protecting public health number of recall events over time. Figures 1 and 2
and as such has several levels of compliance enforce- illustrate the number of quality-related recall events
ment options available to facilitate this mandate (8). initiated from 2006 through 2013 in the US and Irish
Compliance observations noted during inspections markets, respectively.
are summarized on a Form 483. Unlike the practice As shown in Figures 1 and 2, an increasing trend is
in the European Union (EU), these observations are apparent. Further analysis of US data reveals that in

6 Special edition: A Roadmap to GMP Compliance Part 3


Anne Greene, Kelly Waldron, Nuala Calnan

peak recall years (2009 and 2011), seemingly isolated


2500 issues resulted in multiple recall events. For example,
Number of Quality- related

Penicillium spp. cross-contamination at a single firm


2000 (Aidapak Services) led to 1,021 recall events in No-
Recall Events

1500 vember 2011 while cGMP deviations in June and July


2009 led to 1,107 recall events from another manu-
1000 facturer (Advantage Dose LLC). While variables such
500 as improved detection of quality defects or increases
in volume of drug products on the US and Irish mar-
0 2006 2007 2008 2009 2010 2011 2012 2013 kets cannot be ruled out as contributing factors for
the trends seen, the data indicate that recall events
Year are increasingly common.
This led to examination of how the application of
Figure 1: Total Number of Quality-Related Drug Recall Events in the US,
2006 through 2013 (6)
QRM may have influenced these outcomes. Table I
listed the top three categories of recall events for each
year included in the research:

Table 1: Top Three US and Irish Recall Categories, 2006 through 2013 (6)
Number of Quality- related

200 * cGMP deviations is a classification for recall


events given by FDA. Information regarding the im-
150 pact of such deviations, or the specific nature of the
Recall

deviation, was not readily available from the Agency.


100 Annex II of ICH Q9 highlights potential applica-
tions of QRM that, when employed appropriately,
50 could be used to avoid these types of recalls.

0 2006 2007 2008 2009 2010 2011 2012 2013 For example, OOS release specification events
Year might have been prevented through the application
of QRM:
Figure 2: Total Number of Quality-Related Medicinal Product Recalls in
Ireland, 2006 through 2013 (10)

Special edition: A Roadmap to GMP Compliance Part 3 7


Anne Greene, Kelly Waldron, Nuala Calnan

to establish appropriate specifications, identify in warning letters may refine the interpretation of
critical process parameters and establish manu- these data; however, such information is not readily
facturing controls (e.g., using information from available from FDA.
pharmaceutical development studies regarding the A review of warning letters through the lens of QRM
clinical significance of quality attributes and the reveals an increasing trend of citations against the
ability to control them during processing) and to QRM programs and practices themselves, as shown
decrease variability of quality attributes [to] reduce in Figure 4. This would indicate that many applica-
product and material defects [and to] reduce manu- tions of QRM do not inspire confidence that the
facturing defects. (3) manufacturing site has understood the current guid-
Similarly, QRM could have minimized instances ance and regulations in a meaningful way within the
of cross contamination, lack of sterility assurance / context of their individual operations.
sterility failure, and microbial contamination of non- While further research within industry is warranted
sterile products if used: to identify and benchmark effective QRM programs
to determine appropriate zones when designing
buildings and facilities, e.g., [to] minimize contami-
nation, prevent mix-ups, and [to determine the need
for] dedicated or segregated facilities / equipment 60
(3).

Total Number of cgmp- related


50
Though it is not clear in the context of these spe-
cific recall events whether QRM was ineffective or 40

Warning Letters
whether it was simply not used at all, the data dem-
onstrate that product quality (and potential patient 30
exposure to defective product) has not improved
20
since the inception of ICH Q9.
10
State of the Industry: cGMP Compliance
Another potential benefit of QRM implementa- 0
2006 2007 2008 2009 2010 2011 2012 2013
tion is reduced regulatory oversight, which may be
achieved through demonstration of quality system Year
effectiveness in the form of a robust QRM program Figure 3: Total Number of US FDA cGMP-related Warning Letters, 2006
(3, 4). When firms fully embrace the principles through 2013 (7)
and practices of QRM, incidences of breaches of
GMP, product quality deficiencies, and risks to
patient safety should reduce and compliance status
should improve. This should allow manufacturers
Number of cgmp- related Warning Letters with one

to demonstrate the enhanced effectiveness of their


pharmaceutical quality systems through the use of
or more Quality Risk management citations

meaningful measures of quality or quality metrics. 12


Improved quality outcomes for the products and site
in regard to numbers of deviations, CAPAs, custom- 10
er complaints, or evidence of a proactive self-inspec-
tion program would also confirm their increased 8
understanding of how to apply current guidance and
regulations in a meaningful way within the context 6
of their individual operations. 4
Figure 3 illustrates the overall number of cGMP-
related warning letters issued by FDA from 2006 2
through 2013. The increased numbers of compliance
enforcement actions since 2006 indicates that cGMP 0
compliance has not improved since the inception of 2006 2007 2008 2009 2010 2011 2012 2013
ICH Q9. Year
We acknowledge that further analysis of variables
such as the total number of GMP inspections con- Figure 4: US FDA cGMP-related Warning Letters with one or more QRM
ducted in each calendar year and the proportion of citations, 2006 through 2013 (7)
inspection observations (Form 483s) that resulted

8 Special edition: A Roadmap to GMP Compliance Part 3


Anne Greene, Kelly Waldron, Nuala Calnan

that have not been subjects of warning letters, these


data confirm a gap between regulatory expectations
and current industry practice. This conclusion is re-
inforced with a review of the nature of the QRM cita-
tions. Figure 5 illustrates whether each individual ci-
tation indicates either an absence (i.e., failure to apply
QRM where warranted based on an individual event
or circumstance) or misapplication (i.e., inappropriate
use of QRM principles or faulty conclusions drawn as 18.2%
a result of QRM implementation). For example: Misapplied
Absent QRm: we note that your response
includes a commitment to retrain personnel,
revise procedures, and use of premade agar plates
to address [deficiencies in aseptic processing 81.8%
techniques]. Your response is inadequate because Absent
your firm failed to conduct a comprehensive risk
assessment of these poor aseptic process activi-
ties, and the inadequate environmental monitor-
ing program, to evaluate their impact on product
quality (13).
misapplied QRm: We are concernedwith
your risk assessment, which suggests that the
failure of these products to meet acceptance cri-
teria for defects during the 100% inspection has Figure 5: Type of Quality Risk Management Deficiency in US FDA
no bearing on the quality of the released units. Warning Letters Issued between 2006 and 2013 (7)
Please provide detailed information regarding
how you reached your conclusion (14).
An absence of QRM was cited in the overwhelming
majority of instances. This is striking considering 1.8%
that ICH Q9 has been available for nearly a decade. 3.6%
It would be reasonable to expect that sufficient time 5.5%
has elapsed to allow industry to overcome some of
the initial inertia inherent in any paradigm shift, even
one of the magnitude of transitioning from rule-based
to risk-based compliance.
Further analysis with respect to the QRM lifecycle
stage that was cited in the warning letter is similarly
informative. Figure 6 shows the proportion of cita-
tions for the various sections of the lifecycle: risk 89.1%
assessment, risk control, risk review, risk communi-
cation, or risk management as a whole.
The majority of citations indicate that risk assess-
ment, the first phase of QRM, was implicated.
Because QRM is an iterative process beginning with
a robust and science-based risk assessment, citations
in this area of the lifecycle are particularly concerning Risk Management
since it is unlikely that the remaining phases would
prove effective if built upon a faulty or absent risk as- Risk Assessment
sessment. No citations were given for the risk review Risk Control
portion of the lifecycle; if firms struggle to initiate
the QRM lifecycle and successfully complete a risk Risk Communication
assessment, it is likely that the risk review phase was
not reached, thus explaining the absence of citations
for this lifecycle phase. These data support the hy- Figure 6: Quality Risk Management Deficiency by Lifecycle Stage in US
pothesis that certain firms within industry are still in FDA Warning Letters Issued between 2006 and 2013 (7)

Special edition: A Roadmap to GMP Compliance Part 3 9


Anne Greene, Kelly Waldron, Nuala Calnan

the early phases of QRM maturity (i.e., no quality


risk management or informal quality risk manage-
ment) with challenges centered on the initial risk
assessment phase of the QRM lifecycle.
Another indicator as to the expected level of QRM
maturity from the perspective of FDA is whether
warning letter citations include reference to a focus
on reactive or prospective QRM implementation.
While it is broadly acknowledged that risk manage- 30.9%
ment applied in response to a realized issue (i.e., Prospective
reactive) can be helpful to get to true root cause and
plan and appropriate remediation strategy, most risk
69.1%
management practitioners will assert that the full Reactive
value of QRM is achieved through proactive anticipa-
tion and mitigation of potential risks to ensure those
issues do not materialize (3, 5, 15). The PDA QRM
maturity model positioned reactive risk management
as a characteristic of a moderately mature program,
with more effective programs transitioning towards
a prospective and fully integrated focus (5). When
QRM-related warning letter citations are classified
by emphasis (i.e., prospective or reactive; Figure 7),
Figure 7: Quality Risk Management Deficiency Emphasis in US FDA
it is evident that industry shortcomings are primarily Warning Letters Issued between 2006 and 2013 (7)
focused on reactive QRM during the prior eight-year
period. Ireland, excerpts from HPRA inspectional observa-
Examples of this emphasis are evident in the follow- tions similarly indicate a gap in risk maturity. For
ing excerpts: example:
Please provide a copy of your investigation [sur- With regard to the usage of the flexible isola-
rounding breach of data integrity through the tor / barrier for the dispensing of [material X] in
deletion of critical analytical data and backdating [room Y], there was no formal risk assessment
records], along with your risk assessment regard- documented assessing the impact of the intro-
ing the extent and impact of the missing data on duction of the flexible isolator on pre-existing
the quality of all finished drug products released activities in the room. (emphasis proactive, risk
for distribution (16). assessment [11])
your firm failed to investigate numerous cus- Following a risk assessment exercise that had
tomer complaints for several lots of [product] been performed in 2011 on the use of diaphragm
concerning cracked vials your firms response pumps at the site following a diaphragm pump
failed to include a risk assessment for the product failure issue that had occurred at a sister site,
currently on the market (17). appropriate actions had not been taken to ensure
your firm failed to conduct and document that the controls on which the risk had been
a verification under actual conditions of use of deemed acceptable in (Site) were effective
[multiple] laboratory test methods [in your (emphasis proactive, risk control [11])
response] please provide a risk assessment Without focused effort on behalf of industry, this dis-
for possible impurities present in [lots of API parity may intensify as the FDA, European Medicines
released to market] (18). Agency (EMA), and its member states continue along
In these instances FDA is calling for the application the current path towards an increasingly risk-focused
of QRM in situations where an impact assessment regulatory model (19, 20).
might have been used traditionally, i.e., where the
full breadth and gravity of a cGMP-related event or CONCLUSIONS
circumstance must be determined. This trend implies This paper summarizes research designed to char-
that FDA expects industry to have mastered a moder- acterize the current state of pharmaceutical and
ate (reactive/corrective) level of QRM maturity; there- biotechnology industries with respect to the adop-
fore, a gap between the current state of industry (no to tion of Quality Risk Management as per ICH Q9. The
low maturity) and FDA expectations is apparent. research supports the hypotheses that the full value
While a comparable data set was not available from of QRM with respect to product quality and patient

10 Special edition: A Roadmap to GMP Compliance Part 3


Anne Greene, Kelly Waldron, Nuala Calnan

safety has not yet been realized. In addition, industry 12. Looney, W. Irelands Celtic Tiger: Back on the Hunt.
appears to be lagging behind regulatory expectations Pharmaceutical Executive. September 1, 2014. http://www.
with respect to QRM maturity, indicating that cur- pharmexec.com/pharmexec/Europe/Irelands-Celtic-Tiger-
rent approaches to QRM require significant improve- Back-on-the-Hunt/ArticleStandard/Article/detail/853390.
ment. While causality has yet to be established, fur- Retrieved September 22, 2014.
ther research in this area as well as the development 13. FDA, Warning Letter issued to Hemofarm A.D., June 20,
of more robust roadmap towards maturity may offer 2012. http://www.fda.gov/ICECI/EnforcementActions/Warn-
additional insight into what is necessary to achieve ingLetters/2012/ucm310959.htm. Retrieved June 2, 2014.
these goals. The next paper in this series will explore 14. FDA, Warning Letter issued to Jubilant HollisterStier General
this line of inquiry. Partnership, February 20, 2013. http://www.fda.gov/ICECI/
EnforcementActions/WarningLetters/2013/ucm341683.htm.
REFERENCES Retrieved June 2, 2014.
1. FDA, Pharmaceutical cGMPs for the 21st Century A Risk- 15. Vesper, James L. Risk Assessment and Risk Management in the
based Approach. Final Report. September 2004. Pharmaceutical Industry: Clear and Simple. Davis Healthcare
2. ICH Q8 (R2), Pharmaceutical Development. Publishing International LLC, River Grove, IL. 2006.
3. ICH Q9, Quality Risk Management. 16. FDA, Warning Letter issued to RPG Life Sciences Limited,
4. ICH Q10, Pharmaceutical Quality System. May 28, 2013. http://www.fda.gov/ICECI/EnforcementAc-
5. PDA Technical Report No. 54. Implementation of Qual- tions/WarningLetters/2013/ucm355294.htm. Retrieved June
ity Risk Management for Pharmaceutical and Biotechnology 2, 2014.
Manufacturing Operations. 2012. 17. FDA, Warning Letter issued to Bracco Diagnostics Inc., Janu-
6. FDA, Enforcement Reports, http://www.fda.gov/Safety/ ary 31, 2012. http://www.fda.gov/ICECI/EnforcementActions/
Recalls/EnforcementReports/default.htm, accessed July 3 WarningLetters/2012/ucm375606.htm. Retrieved June 21,
through August 2, 2014. 2014.
7. FDA, Warning Letters, http://www.fda.gov/ICECI/Enforce- 18. FDA, Warning Letter issued to Ercros S.A., June 20, 2012.
mentActions/WarningLetters/default.htm, accessed June 1 http://www.fda.gov/ICECI/EnforcementActions/WarningLet-
through July 1, 2014. ters/2012/ucm310963.htm. Retrieved June 18, 2014.
8. FDA, About FDA. http://www.fda.gov/AboutFDA/Transpar- 19. FDA, Strategic Priorities 2014 2018. Draft for Public
ency/Basics/ucm192695.htm. Retrieved August 21, 2014. Comment. http://www.google.com/url?url=http://www.fda.
9. FDA, Regulatory Procedures Manual, Chapter 4 Advisory gov/downloads/AboutFDA/ReportsManualsForms/Reports/
Actions. 2011. UCM403191.pdf&rct=j&frm=1&q=&esrc=s&sa=U&ei=dF
10. HPRA (formerly IMB), Annual Reports, 2006 through 2009. bzU9_ZKMvioASmnIG4Cg&ved=0CBQQFjAA&usg=AFQj
http://www.hpra.ie/homepage/about-us/publications-forms/ CNEvMy6uqINSw2C8tJ7rqhjIHzNclg. Retrieved August 10,
corporate-and-policy-documents. Retrieved June 23, 2014. 2014.
11. Extracted inspectional observations (anonymous) specifi- 20. EMA, Road map to 2015. December 16, 2010. http://www.
cally related to observed QRM deficiencies provided by Dr. K. ema.europa.eu/docs/en_GB/document_library/Report/2011/01/
ODonnell from HPRA on August 8, 2014. WC500101373.pdf. Retrieved September 13, 2014.

Special edition: A Roadmap to GMP Compliance Part 3 11


Eldon Henson and Whitney Sandberg

Mergers & Acquisitions: Managing


GxP Compliance Through
Integration and Organizational
Change
Eldon Henson and Whitney Sandberg

ExEcutivE Summary
In todays world of record-breaking merger and acquisition activity,
management of change, especially relating to GXP compliance is essen-
tial. Many do this poorly with a reported failure rate of 70%. However,
the risks associated with integration can be significantly reduced with
proper planning and execution. This article outlines those key ele-
ments that can drive successful integration activities:
Planning
The Integration Roadmap
The Integration Playbook
Value Capture
Risk Remediation
Human Capital, and
GXP considerations
By developing a plan and working that plan, you can significantly re-
duce the risks and hurdles that can threaten the overall value expected
by these transaction.

iNtrODuctiON
According to Price Waterhouse and Coopers, the life science sector
experienced records levels of merger and acquisition activity and 2015
with over $400B in closed deals1. Higher numbers are expected for
2016. However, did you realize that, according to a number of sources,
most mergers are doomed from the beginning? It is estimated that
approximately 70% of mergers fail to achieve the overall value antici-
pated2.
Because of the challenges of mergers and the poor success rate
experienced, an entire culture and practice of integration management
has arisen designed to manage toward successful integration. Firms
create integration processes, information technology solutions, check-
lists, workplans, and every other tool to ensure that integration is not
doomed. Many have become so process oriented that they actually
believe just follow the process and you will not fail will guarantee
success. Others believe that keeping integration simple is the key.
Despite these formal approaches to integration, our experience is that
integrations are like snowflakes every integration is different. Thus,

12 Special edition: A Roadmap to GMP Compliance Part 3


Eldon Henson and Whitney Sandberg

you need to achieve that difficult balance between integration phases as follows:
formality and flexibility; processes and relationships; 1. PreClose to Close Phase
systems and people. 2. Interim Recognition Phase
McKinsey & Company estimates that those who do 3. Full integration Phase
integration well achieve as much as 6 12% higher 4. Business Continuity.
total shareholder return that those that do integra- Figure 1 below describes the duration of each phase,
tion poorly3. Similarly, in FDA-regulated industries, respective goals, critical activities, and deliverables.
there are a number of examples of GXP compliance
issues that have arisen because important compliance Planning in the Pre-Close Phase
elements have fallen through the cracks during the Perhaps the most important thing we have learned
integration process. in our own integration effort is that involvement by
Integration is a journey that should result in high the integration team early in the process is critical.
GXP performance, a reliable supply chain, outstand- In fact, we believe the best integration results occur
ing product quality, and fair product costs. Anything when the actual integration team is involved in the
less diminishes the return expected and harms the due diligence process. By doing so, those individuals
business case that actually justified the acquisition/ charged with executing integration activities already
merger. understand the deal model, the key risks, the key is-
In this paper, we outline some of the key success sues to be addressed, and the critical success factors.
drivers that can guide you to a successful integration. They will have an excellent understanding of the
By identifying and managing these key drivers, you business from their review of due diligence docu-
can predict better outcomes and higher shareholder mentation and, from a GXP perspective, they will
returns for these critical activities. know what potential compliance issues or concerns
are on the horizon. The integration team can begin
Planning, Planning, Planning planning key activities for the subsequent phases
Despite the need to remain flexible and treat each during due diligence and have an initial plan ready to
integration as a unique event, you must not neglect the execute on the day of closing. When integration team
critical role that planning plays in a successful integra- members are not involved in the due diligence phase,
tion. The integration roadmap is comprised of four you risk delaying integration activities (possibly, by

Figure 1: INTEGRATION ROADMAP.

Special edition: A Roadmap to GMP Compliance Part 3 13


Eldon Henson and Whitney Sandberg

several months) or you risk missing critical compli- Critical activities during this phase include:
ance activities that must be addressed. Systems requirements and alignment Some sys-
Critical activities during this phase include: tems integrations may require extended time or
Talent management Assessment and retention funding to fully integrate. Thus, early identifica-
of key talent is an activity that must begin early tion and planning is essential. Identifying these
in the planning process. In most cases, mergers activities and beginning the planning process
and acquisitions pose significant organizational should occur during this phase.
anxiety. As a result, good individuals often now Regulatory/compliance requirements The key
become more aggressive toward career options. success factor for many mergers or acquisitions
Identifying key talent and communicating future in GXP industries is remediation of compliance
roles and incentives to ensure retention can make concerns or navigation through regulatory pro-
or break the integration process. cesses. Identifying these early may define overall
Governance and/or organizational structure As project success.
quickly as possible, it is important to outline how Talent assessment and organizational design
the new organization will fit into the new organi- - From a business perspective, you must also
zation in total. Creating a vision of the new orga- determine how the acquisition will fit into the
nization can aid both the integration team and existing organization. For example, will the
impacted individuals see their role and direction new entity operate as a separate entity or will it
toward the eventual merged team. be incorporated into an existing business group
Knowledge exchange Early in the process, (e.g., division or commercial function)? Under-
preferably during due diligence, you begin standing this and the key business drivers will
accumulating needed knowledge that might be determine the level of integration required before
critical in the success of the integration. For and after the deal close date.
example, when an acquisition is made of early Development of full integration plan - Identifying
drug development products, key individuals may the team members from each side of the deal to
hold much of the knowledge needed for eventual drive the integration process is also a key activity
product approval and manufacturing. Identify- executed during this phase. Selection of indi-
ing this knowledge and capturing it is critical vidual team members is critical as it possible for
early in the planning process. an integration process to fail simply because of
Communication You cannot over-communicate poor relationships between parties. This team
to both parties during the planning phase for should develop the specific actions, target dates
integration. Helping individuals and teams for completion, and individuals responsible for
understand what is occurring and how they fit the integration that will define and drive success.
into the new organization is critical to early buy-
in and participation. Planning in the Full Integration Phase
Value capture and/or business synergies Begin Once the preliminary integration principles and activi-
early to identify the key value drivers for the ties have been outlined and the deal has closed, you
merger/acquisition. Knowing how the new progress to the full integration phase. By this time,
organization will drive key shareholder value you understand the key business drivers and how val-
can guide future integration activities that may ue of the acquisition will be captured. In this phase,
ensure success. you are executing the integration activities outlined in
the interim integration phase. The integration team
Planning in the Interim Integration Phase is performing and the business is shifting to the new
A number of key activities are essential during the in- normal operating mode.
terim integration phase (that period between the deal Critical activities during this phase include:
close and full integration). The first objective must be Execution of the full integration plan At the full
to do no harm to the existing business. In many inte- integration phase, the primary activity is to simply
gration cases, failure occurs because the purchaser execute the plan already developed. Processes to
attempts to consolidate too much too quickly that the monitor and guide progress (e.g., typical project
key business drivers are lost. For acquisitions with management oversight activities) will be required.
complex supply chains, for example, care must be Functional area policy/procedure alignment It
taken to ensure business continuity through existing may be necessary during full integration to begin
and revised contracts, relationships, and business aligning policies and procedures to reflect the
processes. A failure to plan in these areas is a plan to new normal for the organization.
fail. Systems integration Full integration of systems

14 Special edition: A Roadmap to GMP Compliance Part 3


Eldon Henson and Whitney Sandberg

(e.g., SAP for example) may take months or years. activities or technologies (e.g., biologics, CMO-man-
Monitoring these activities and ensuring processes ufactured, solid dosage forms, etc.). The playbook
exist to drive progress are essential. includes:
Implementation of organizational design At Stage gates (stopping points at which an assess-
this point, final implementation of the new orga- ment of progress occurs, solicitation of additional
nizational design should occur. Involvement resources, and modifications of the plan, as
of Human Resources personnel should have needed)
occurred earlier to ensure that key talent are Role owners and responsibilities for key activities
retained. Core teams, members, and responsibilities
Checklists
Planning for Business Continuity Documentation requirements
Finally, as the primary integration activities are be- Risks
ing completed, planning for the ongoing business and Process maps
business continuity must occur. In this phase, you Performance objectives (a necessary component
are executing new contracts with suppliers, working for ensuring accountability for key deliverables
to enhance business flexibility, removing redundan- AND to monitor progress toward full value cap-
cies, and improving both operational and financial ture)
performance. It is during this phase that you begin However, as occurs in sports, the playbook only out-
realizing the value the drove the deal business case. lines what we know and the capabilities we have. Ev-
Critical activities during this phase include: ery integration process is unique, thus, flexibility and
Routine operation per business plan At some the ability to shift activities, resources, and priorities
point after full integration, the merged/acquired is essential for a successful integration.
entity should be under all the same normal busi-
ness processes that govern other businesses. valuE caPturE
However, some activities, such as metrics, may Organizations often struggle to maximize the po-
take a full year planning cycle to fully imple- tential value during the integration because work is
ment. not planned around key value drivers. A complete
Identification of synergies/redundancies By this evaluation of the supply chain, operations, quality
phase, you should understand what synergies network, and corporate or strategic initiatives is nec-
may be possible through the new organization. essary to fully realize synergies that either maintain
Working to align activities and eliminate redun- or enhance the value of the merger/acquisition.
dancies should now be ongoing. Process maps can often be useful tools for identify-
Continuous improvement As with any activity, ing potential synergies. Lets look at an example. In
continuous improvement should be an ongoing one merger, the classical distribution process was
success driver. Execution of improvement activi- used by the acquiring company. That is, the supply
ties and identification of new opportunities can chain bundled shipments for numerous customers for
drive further value from the merger/acquisition. regular shipment to a distribution partner. How-
In short, planning for integration must involve ever, the acquired firm shipped product directly to
more than the deal close. It must include the busi- the customers every three months from an on-site
ness case for the acquisition, the basic business im- distribution center. By looking at potential synergies,
peratives that provide the expected value, the actual the combined company was able to include the new
execution of the integration to realize that value, products in their bundled shipments to reduce distri-
and the ongoing continuity of the business after it bution handling and shipment costs with no adverse
has been normalized into the operation. A failure to impact to customers or service.
properly plan in any of these stages can hamper the Capturing the total composite value of a merger/ac-
overall success of the integration. quisition requires collaboration, openness to change,
and a realization that change is required. By truly
iNtEgratiON PlaybOOk seeking the best practice across the new combined
Most companies that have been successful in integra- company, the elimination of redundancies and a
tion activities have developed an integration playbook culture of continuous improvement can come quicker
that guides their efforts. A playbook for integration and be done more effectively.
is essentially the same thing as the playbook used
by sports teams. It is a plan that includes repeatable HumaN caPital riSkS: culturE iS kEy
end-to-end processes and specific merger/acquisition Integration is not risk-free. Perhaps the most signifi-

Special edition: A Roadmap to GMP Compliance Part 3 15


Eldon Henson and Whitney Sandberg

cant risk (and, reason for the failure of integration made. Identification and remediation of existing
efforts) is a failure to properly consider and integrate compliance issues can be costly, delay product intro-
competing or diverse cultures. The result is a loss of ductions, or hamper customer service/satisfaction.
human capital, either through undesirable attrition Realizing the ultimate value of the deal can hinge on
or disengagement. the avoidance of costly GXP issues.
So, what are the key factors to fully retain the So, what are some key considerations for manag-
benefit of the human capital resulting from a merger ing GXP compliance risks?
or acquisition? Successful firms consider the fol- Encourage open discussions at the earliest stage
lowing 5 key elements for blending cultures during possible regarding existing or potential GXP
integration: concerns the earlier you know about these
Understanding the values both firms have in risks, the better opportunity to prepare resourc-
common es and financing to correct them
Understanding the differences in values that Share compliance enhancement plans across
drive each entities compliance is one area that should
Collaborating to establish combined values facilitate an early success regarding collabora-
Identifying the right leaders to drive the com- tion and synergies
bined/new culture Encourage cross-functional and cross-company
Engaging the new workforce around the com- learning often, the new acquired company has
bined/new culture technologies that are new to the acquiring com-
For these steps to be successful, engagement from pany take advantage of these as collaborative
both parties is required. Establishing a new culture learning opportunities
cannot be a one-sided effort. It also cannot be a top- Share GXP expertise often, experts from one
down effort. Someone once said, entity can provide immediate benefits to the
Culture is defined as the worst behavior that the other
leader allows to occur in an organization. Ensure that emerging GXP issues (such as data
Culture is not what you say, but what you do. integrity, metrics, etc.) are discussed, shared,
Thus, it is imperative that you ensure that actions to and fully engaged as early in the process as pos-
back-up the desired culture are created, nurtured, sible
and uncompromisingly enforced. GXP compliance is one that should foster sig-
Following are a few learnings we have identified nificant common ground and common interests
that define in the early days of an integration what between companies. This should allow early and
kind of culture is likely to emerge: deep collaboration to help drive compliance up and
How is the leadership of the acquiring firm risks down.
viewed? Is that leadership viewed as trustwor-
thy, open, and forthcoming? cONcluSiONS
Is there a strategic vision and strategy that The failure rate for firms to realize the expected value
makes sense in the newly combined company? from mergers and acquisitions is staggering. Howev-
Can each employee, especially for the acquired er, we must not resign ourselves to that fate. It is pos-
firm, see how they fit into the newly combined sible to realize or exceed the value of these transac-
firm? tions, but the appropriate approach must be exerted.
Does the acquiring firms actions back up their Realizing that a merger or acquisition can be a time
words around compliance, quality, and safety? of significant change for quality and manufacturing
What objectively proves this commitment? organizations, developing a plan to properly integrate
Are people held accountable for results? Is per- the acquired organization into the new combined
formance valued over behaviors? organization is essential. By creating a roadmap with
Are employees rewarded for creative thinking? specific planning activities in each phase of integra-
Are employees reprimanded for taking risks? tion, you can significant reduce the risks and hurdles
Open and frank discussions around these ques- to value capture that have plagued so many others.
tions can set the stage for productive collaboration Integration activities also do not come risk-free.
as the integration progresses. Identifying and remediating these risks early in
the integration process can help avoid pitfalls that
cOmmENtS ON gxP cOmPliaNcE increase costs, impart delays, or impact business con-
Throughout the integration process for GXP prod- tinuity after the deal has closed. In every integration,
ucts/processes (from due diligence through full consideration for people and relationships can be the
integration), considerations for compliance must be difference between success and failure.

16 Special edition: A Roadmap to GMP Compliance Part 3


Eldon Henson and Whitney Sandberg

GXP considerations must be included in any rEFErENcES


merger or acquisition impacting healthcare industries. 1. Drone, Dimitri, and James Woods. Pharmaceutical and life
Identifying opportunities and including compliance sciences. PwC. N.p., Feb. 2016. Web. Sept. 2016.
enhancement into the planning and execution plan 2. McMorris, Esther. Why do up to 90% of Mergers and Ac-
can set the tone early for a smooth and collaborative quisitions Fail? Business Review Europe - European Business
integration. Magazine & Website - European Business News. N.p., 28 Jan.
Integration of mergers and acquisitions can be 2015. Web. 05 Aug. 2016
successful. Hopefully, we have outlined many key 3. Doherty, Rebecca, Oliver Engert, and Andy West. How the
considerations that can drive value for your future Best Acquirers Excel at Integration. McKinsey & Company.
initiatives. N.p., Jan. 2016. Web. 14 Sept. 2016.

Special edition: A Roadmap to GMP Compliance Part 3 17


Carol Brandt

Structured GMP Audits


Carol Brandt

ABSTRACT
Although Good Manufacturing Practices (GMP) audits are common
both for internal and external customers, a structured approach to
planning and execution can result in a successful well-executed audit.
There are a variety of audit types (GMP baseline, GMP risk-based, due
diligence, product-specific, procedures, area-specific, etc.) that may be
scheduled for a variety of reasons (preventative or remedial, Regulatory
Agency findings, inspection readiness, product or process failures, com-
plaints, deviations, routine assessments of systems, etc.). The proper
planning, management, facilitation and execution of an audit are criti-
cal to a successful audit. A complete and concise report of the exercise
provides maximum benefit to the customer.

INTRODUCTION
GMP audits are an important activity in a site compliance program.
An organization that initiates a GMP audit has clear objectives and
expectations for the audit.A significant amount of time, personnel
effort, and cost is expended in a GMP audit.Audits that are properly
planned, structured, and executed should yield maximum benefit to
the organization.A successful audit will result in the appropriate level of
coverage, clear direction, guidance, communication, controls, appropri-
ate auditor skills for the respective site areas, clear observations, good
recommendations, and customer satisfaction that audit objectives and
expectations are met.
A properly planned audit should be carefully structured to comprise
several defined activities.These include:
Customer communication. Customer objectives set the objectives,
tone, focus, and technical scope for the audit.
Auditor selection. Competent auditors with appropriate expertise
(technical, regulations, industry practices, etc.) must be selected.
Multiple auditors may be required to successfully audit widely
divergent technical areas.
Audit planning. Early communication and sharing of any manufac-
turing, facility, and customer information.A proposed agenda and
audit focus is determined.
Audit preparation. The proposed agenda is reviewed with the
customer to finalize specific audit activities.Site documents to be
reviewed in the audit are requested.
Audit execution. The actual audit is performed as planned and pre-
pared in prior discussions.
Audit report. Results of the audit are communicated to the cus-

18 Special edition: a roadmap to gmp compliance part 3


Carol Brandt

is external, ensure a Confidential Disclosure Agree-


ment (CDA) is in place before the final audit planning
since a customer may be reluctant to disclose known
internal issues.

It is critical to understand the basis for the audit.


For examples, audits may be routine, voluntary, prior
to merger or purchase, product or process failures/
focus, deviations, complaints, Regulatory-Agency
driven, and other types. Pre-audit discussions must
include participation from customer compliance or
Quality Assurance (QA) contacts. It is fairly common
for initial customer contacts to be with executive
management or with operations management repre-
senting the site who may not have a strong Quality/
Compliance background. These individuals may not
be able to clearly express any compliance-related
concerns and focus for the audit.
Mock audits may be conducted to prepare and
make ready a site for a future regulatory agency audit.
These mock audits require a trust-based relation-
ship between the customer and auditors to ensure
all potential risk areas are addressed without bias or
prejudice. The customer is provided with a complete
report that identifies areas of compliance risk before
they become investigation findings or present a risk
to product safety at the completion of the mock audit.
A firm date and time should be set as well as the
duration of the audit and number of auditors.Specific
and special needs of the customer should be identi-
fied, such as internal resource issues when multiple
systems are audited concurrently.

AUDIT TypeS AND AppROACh


General pharmaceutical Good Manufacturing
Practices (GMP) audits, risk-based assessments, due
diligence, and baseline audits contain many of the
same requirements for audits. These audits may vary
in length of time, depth of the audit, number of audi-
tors, and perhaps focus. They may be limited to a
specific product, area, or procedures. There are many
different approaches to performing audits. One of the
more common approaches is to take a systems ap-
proach in assessing the six systems described in the
Figure 1: Structured Audit Approach FDA Code of Federal Regulations, Good Manufac-
turing Practices:
tomer. Quality Systems
Figure 1 describes the sequence of activities in a Facilities and Equipment System
structured GMP audit. Materials Controls System
Production Controls System
CUSTOmeR COmmUNICATION Packaging and Labeling System
The customer requesting an audit may be a client, Laboratory Controls System
internal department, or external group or firm. Sub-systems would include any requirements listed
Understanding the customer reasons and concerns in the CFR within the above system categories. For
must set the tone for the focus of the audit.If client example, a facilities and equipment system audit

Special edition: a roadmap to gmp compliance part 3 19


Carol Brandt

might assess the following sub-systems: Physical fa- AUDIT plANNINg


cility space, maintenance and repair function, utilities Early communication and sharing of any manufac-
qualification, temperature/humidity controls, equip- turing, facility, and customer information with the
ment qualification, cleaning validation, and other auditors is a key to the success of the audit. Plan a
related systems. conference call with the auditors ahead of the audit, for
Audits should be conducted for compliance with which customer background has been provided in ad-
21 CFR Parts 210/211/11, ICH (International Confer- vance of the call. Assign a lead auditor to provide audit
ence on Harmonisation) Q10, Pharmaceutical Quality guidance and cohesiveness of the audit team, answer
Systems; ICH Q9, Quality Risk Management; ICH auditor questions, report concerns, and act as primary
Q8, Pharmaceutical Development; and any relevant communicator between the customer and the team. A
US FDA Guidances such as process validation, stabil- draft agenda should be reviewed with the auditors for
ity, Active Pharmaceutical Ingredients (APIs), aseptic the opportunity to identify additional areas of con-
manufacturing, and so on. Additional requirements cern or focus to be assessed. Timing should also be
for compliance with any international regulatory reviewed with the auditors to ensure they are comfort-
agencies should be clarified with the customer. able with the time allotted to each area. Preliminary
responsibilities should be assigned and discussed.
AUDITOR SeleCTION It may be beneficial to mix responsibilities across
Auditors should be subject matter experts (SME) systems based on technical qualifications. This should
with focused areas of expertise. It is rare to identify be clearly delineated, documented and shared with the
someone who has the technical qualifications and auditors to ensure the responsibilities are understood
can successfully audit all of the systems listed above and the sub-system assessments are fully covered.
with a comprehensive knowledge of the require-
ments, industry practices, and regulatory guidance AUDIT pRepARATION
recommendations. This expertise should extend to Audit preparation based on above plans is the next
the customer products manufactured oral sold dose, phase of the structured audit.Once finalized, the
API, sterile products, OTC (over-the-counter), etc. proposed agenda is provided and reviewed with the
Also extremely important are the personal commu- customer for comment. The agenda should include a
nication skills of the auditors. An audit is not based brief overview presentation of the site and products
on opinion or preference, but on compliance with by the customer and a tour of the facilities to fully
the regulations and guidance documents. An audit acclimate the auditors. Each audit day should end
should ultimately result in knowledge transfer from with a status meeting with auditors and appropri-
the SME to the customer in a partnering, helpful, and ate site personnel. The schedule should include time
non-threatening fashion. for auditor preparation prior to end-of-day wrap-up
With the more common audit addressing the six discussions. The end-of-day discussions present the
systems, and the most common risk-based audit most significant findings and observations from the
being conducted in 3-5 days, the best results for ap- day to the customer.
propriate SME coverage are by assignment of at least A brief list of documents for the customer to
three auditors. All of the auditors should be experts provide to the auditors ahead of the audit and during
in the regulations and their applications, and have the audit should be developed. This will allow the
years of auditing experience. For any audit includ- auditors to come prepared and allow the customer to
ing a laboratory, one of the three auditors must be begin retrieving documents. One of the most help-
a chemist. The remaining five systems should be ful documents is an organizational chart to identify
divided among the auditors based on their qualifica- responsible heads of the systems groups. Past inspec-
tions, capabilities and experience. All must have GMP tion reports and responses can be beneficial to under-
expertise and working knowledge of their specific standing the areas of highest compliance risk. Each
expertise and applications. auditor must devote a few hours of time ahead of the
Oral solid dosage and aseptic manufacturing may audit to prepare and review for the audit.
require specific skills and expertise in production The agenda is finalized and distributed to the
and facilities. An auditor with technical microbiology primary customer contacts and auditors prior to the
background should be engaged to conduct the micro- actual audit day. Final dates, location, times and con-
biology lab audit for aseptic processing. tact information are distributed to all involved.
Once auditors are selected and audit dates are con- Detailed checklists should be prepared which
firmed, copies of the respective auditor CVs/resumes follow the GMP requirement sections in the CFR to
and auditor contact information are provided to the assess the quality system sub-systems. These may be
customer. tailored to focus on potentially problematic areas for

20 Special edition: a roadmap to gmp compliance part 3


Carol Brandt

the customer if applicable. For example, if commit- procedures (SOPs) is necessary to understand the
ments have been made in the past two years to a customers requirements. If not compliant with regu-
Regulatory Agency, it may be beneficial to include a latory requirements or agency guidances, this should
review of the status of those commitments for veri- be noted as a finding. Evidence of the execution of
fication of completion. Checklists should be further the SOPs must also be reviewed to ensure the SOP is
tailored for the audit type. Shorter risk-based and being followed in practice. Any SOPs, records, logs,
due diligence audits will focus on the areas of highest other documents reviewed as supporting evidence
potential compliance risk. This requires the preparer must also be identified. While conducting the audit,
to have a full understanding of the customer compli- ensure that documented evidence of any findings are
ance status, as well as knowledge of industry compli- reviewed and references noted. Findings related to
ance trends and regulatory agency key focus points. observations must be referenced by document title,
A well-structured checklist ensures that all scheduled date, area, and book and/or page number such that
sub-systems are assessed and there is appropriate the customer is able to locate and correct the observa-
coverage of the topics defined by the audit type and tion as applicable. Too many audits draw conclusions
focus. about the compliance of a system without looking
beyond the SOPs to the actual execution of the SOPs
AUDIT exeCUTION in practice.
Actual performance of the audit as planned and pre- Thorough audits cannot be conducted solely from
pared is then initiated.It can be beneficial for the au- a review of documentation unless such limited focus
ditors to meet in person ahead of the audit to discuss is specifically requested. Plant walk-throughs can
any concerns and additional questions they might uncover a host of problems not evident unless the
have. If external auditors, they should arrive on auditor is actually present on the plant floor or in the
site together and plan enough time to sign onto the laboratory. Physical maintenance, operational cross-
site in advance of the scheduled start time. Internet contamination risks, people flow and gowning, materi-
connections should be available to them when they al and product flow, and storage issues may be readily
arrive and set-up time should be allowed to minimize identified but only if the auditor devotes sufficient
disruptions when the audit starts. time to the actual operations area. Discussions with
Unless the customer prefers to lead the opening and observing activities of operators and analysts can
meeting, the lead auditor should be prepared to initi- also identify processes which are not represented in an
ate the audit. This should include brief introductions SOP or activities which are not compliant.
from auditors and customers. The auditors should A daily wrap-up meeting each day is recommended
focus on relevant background, expertise, and areas to review significant findings of the day. The meeting
each will be involved in assessing. A review of the ensures that the customer understands the nature of
agenda and responsibilities with all involved should the findings.It also provides the customer an oppor-
be performed. tunity to refute any findings with additional informa-
The auditors should provide additional lists of tion that might not have been originally available to
documents required for review in their relevant areas the auditors.
to their customer contact at the beginning of each
audit day. The status of each of the documents re- AUDIT RepORT
quested, when requested, and when received should The audit report provides finding of the audit team
be documented. All site procedures should be readily for review and initiation of remediation by the cus-
available to all of the auditors on day one of the audit. tomer. The lead auditor should obtain copies of the
Rapid retrieval of information is an important factor facility/product overview presented by the customer
in Regulatory Agency audits. at the beginning of the audit. This may be sum-
The auditors must maintain a list of documents marized in the report to include a description of the
reviewed by title, number, version, and effective date facility, types of products manufactured, previous
for listing in the final report. It is recommended that regulatory agency inspection dates, and any other
the auditors address higher compliance risk sub- pertinent information audit information.
systems first in case more detailed review is required Each finding should be accompanied by document/
requiring more the originally allotted time. Time record evidence of the inadequacy or non-compliance
constraints may then preclude auditing of other areas associated with the observation as applicable which
as previously planned. The lead auditor should be require a review of associated records. The finding
kept informed of the status of such constraints if sub- must be stated as a finding -- not a recommendation
systems must be re-assigned during the audit. -- and based on clear, referenced evidence. Some
A review of the audited area standard operating customers prefer to have the regulatory reference pro-

Special edition: a roadmap to gmp compliance part 3 21


Carol Brandt

vided in the report as well. Auditors should take care CONClUSION


not to provide a personal opinion. It may be that the Consistent, well-planned, and executed GMP audits
typical industry practice is to implement a process in not only provide the auditors with the necessary
a certain way, but if the customer process is different structure to ensure all required areas are assessed,
and compliant, it is simply a personal opinion and but also benefit the customer for the same reasons.
not a requirement.If industry practice differs from From one audit to the next, even if the focus is ad-
the manner in which the customer performs a task, it justed, similar materials may be used with the same
might be noted by the auditor as a consideration but approach. Standardizing and structuring the man-
not cited as a regulatory finding. ner in which the audit is planned and executed can
Findings should be identified individually and ensure consistent, successful, and beneficial audits
categorized is a risk manner (minor, major, critical or and reports.
low, medium, high) depending on the customers re-
quirements. Each finding should be accompanied by ReFeReNCeS
a recommendation for action to be taken to mitigate 1. Code of Federal Regulations, Title 21 Food and Drugs (Govern-
the finding. The more detail that can be provided in ment Printing Office, Washington, DC), Part 210..
the recommendations relative to actions required, the 2. Code of Federal Regulations, Title 21 Food and Drugs (Govern-
more benefit to the customer, and the easier it is for ment Printing Office, Washington, DC), Part 211..
the customer to generate a corrective action plan from 3. ICH (International Conference on Harmonisation of Technical
the report details. Requirements for Registration of Pharmaceuticals for Human
The report is combined from respective auditor Use), ICH Q10 Pharmaceutical Quality System, June, 2008..
inputs and reviewed to ensure findings are based 4. ICH (International Conference on Harmonisation of Technical
solely on regulatory requirements and not on audi- Requirements for Registration of Pharmaceuticals for Human
tor opinions. Any inconsistencies are reconciled and Use), ICH Q9 Quality Risk Management, November, 2005..
overlaps combined. For example, a review of the 5. ICH (International Conference on Harmonisation of Technical
deviation management process in the quality system Requirements for Registration of Pharmaceuticals for Human
may result in the conclusion that the system might Use), ICH Q8 Pharmaceutical Development, November,
not be fully compliant based on the SOP and records 2007.
reviewed, while a sampling of selected production 6. FDA, Guidance for Industry Process Validation: General
system-specific deviation records may have been Principles and Practices, (Jan, 2011)..
properly reported and investigated. 7. FDA, Guidance for Industry Drug Stability Guidelines, (Dec,
Sub-systems that could not be addressed due 2008).
to time constraints but were listed on the original 8. ICH (International Conference on Harmonisation of Technical
agenda should be identified in the report. Requirements for Registration of Pharmaceuticals for Human
Since many audit reports can be quite lengthy Use), ICH Q7 Good Manufacturing Practice Guide for Active
based on the areas covered and number of findings, Pharmaceutical Ingredients, November, 2000..
an executive summary is beneficial to customer man- 9. FDA, Guidance for Industry Sterile Drug Products Produced
agement who may require a high level brief reporting by Aseptic Processing Current Good Manufacturing Prac-
of the critical findings. tice, (Sept, 2004).

22 Special edition: a roadmap to gmp compliance part 3


Anne Greene, Kelly Waldron, Nuala Calnan

Moments of Truth: Creating


a Quality Culture by Making
Good Manufacturing Practice a
Lifestyle in Your Company
David Markovitz

The late management consultant, educator, and author Peter Drucker


said, Culture eats strategy. Culture equates to the habits of the people
who work in the organizationhow they behave on a daily basis.
The late quality guru W. Edwards Deming said, People work in the
system, management works on the system.
So, if we want to create a culture where quality is paramount, then
management must create a system that encourages and rewards people
to do the right thingall the time. Thus, we can have a culture of qual-
ity.
Consider a Gallup survey published in June 2013 (1):
Seven out of 10 workers have checked out at work or are ac-
tively disengaged, according to a recent Gallup survey. The survey
classifies three types of employees among the 100 million people
in America who hold full-time jobs. The first is actively engaged,
which represents about 30 million workers. The second type of
worker is not engaged, which accounts for 50 million. These
employees are going through the motions at work. The third type,
labeled actively disengaged, hates going to work. These workers --
about 20 million -- undermine their companies with their attitude,
according to the report.
Here is an example of how one company is addressing the issue of
not engaged and disengaged workers. In April 2014 Amazon chief Jeff
Bezos, in a letter to the company shareholders, revealed what has to be
the most counterintuitive personnel policy in corporate America today:
If an employee isnt happy working at the online retail giant, they can
earn up to $5,000 just for quitting.
The goal is to encourage folks to take a moment and think about
what they really want, Bezos explains. In the long-run, an employee
staying somewhere they dont want to be isnt healthy for the employee
or the company. (2).
Amazon is giving the rest of us an interesting viewpoint of business.
This action is sending a message to everyone working at Amazon, or
who may be looking to work at Amazon, that they want you on the
team only if you are willing to be actively engaged in your work. The
hiring and selection process is highlighted here as a process critical to
the success of the company.

Making a Difference
Those people who are actively engaged in their work are not only work-
ing for a paycheckthey are working to make a difference.

Special Edition: a roadmap to gmp compliance Part 3 23


Anne Greene, Kelly Waldron, Nuala Calnan

The industries regulated by the US Food and Drug the quality of the products, services, processes, pro-
Administration have an advantage over most other cedures, and policies that guide them in their work.
industries in the Making a Difference department. In a company where GMP is a Lifestyle, people
For the most part, these industries are contributing are more apt to make the right decision when faced
to improving the quality of life and, in many cases, with a Moment of Truth.
preserving life itself. Consider an example from a Warning Letter issued
In our good manufacturing practice (GMP) work- by FDA (3):
shops, we always state that working in one of the In January 2010, your firm identified an insect
FDA-regulated industries is a privilege. Certainly its floating in a waste container in an aseptic filling
a privilege to work in an industry with an honorable room. During your root cause analysis, you stated
goal of improving the quality of life. However, along that the most likely cause was the stopper supplier;
with that privilege comes an awesome responsibility, however, you did not conduct an audit of the supplier
the responsibility to do the right thing, all the time. until April 2011.
Thats where creating a culture of quality, or what Here was a Moment of Truth. Even though the
we call Making GMP a Lifestyle, comes into play. It thinking was that the problem was with the supplier,
starts with top management. Key questions that must the firm waited 14 months to conduct an audit.In a
be asked include: company where GMP is practiced as a lifestyle, the
Does top management view improving the qual- audit would have taken place as soon as the supplier
ity of life as their primary goal or does top man- was suspected to be the source of the problem.
agement view profits as their primary goal? Next, consider two examples from a manufacturing
How does top management communicate their operation. Keep in mind that manufacturing opera-
goals? tions are schedule driven. There is pressure to keep to
Certainly profits are critical to any organization. the schedule.
One couldnt produce products or provide services Example One: Joe works as a Compounder. He is
that improve the quality of life without sustained preparing a batch of product by combining ingre-
revenue. dients as a manufacturing document dictates.
Yet, which company do you think has the best Joe has gathered all the right components and
chance of creating a culture of quality? ingredients together and begins the compound-
Company A, where top management states that ing process. He opens one container that has the
our goal is to improve the quality of life for our quality control (QC) approval label affixed and
customers, often called patients, and when we do is within its expiration date. However, once he
that well, well make money. opens it, he smells an unfamiliar odor. He says
Company B, where top management states that to a fellow worker, This smells different than it
we produce products and services to improve usually does. Here is now a Moment of Truth.
the quality of life for our customers, often called Does Joe proceed, seeing that the QC approval
patients, so that we can make money. label is in order and the material is still within
Most people in the workplace are not in upper the expiration date? Or should Joe stop and alert
management. They work in the system created by his supervisor? In a company where GMP is
those in these higher management positions. Most practiced as a lifestyle, Joe stops and alerts his
people who want to be actively engaged in their work supervisor. Something may be wrong with that
ask themselves, How am I making a difference? container of raw material.
Both (high quality products/services and healthy Example Two: Jill works as a Filling Operator.
profits) are important to the success of a business, She is preparing the filling line for filling a batch
but the difference is often in how it is communicated. of product. When she turns on one of the pieces
Creating a culture where GMP is a Lifestyle begins of equipment, she says to herself, Hmm, this
with communication of the purpose of the company. sounds different than it normally sounds. Here
The behavior of those in top management must is now a Moment of Truth. Does Jill proceed with
be consistent with the message they communicate. starting the filling operation,oOr should she stop
People learn at a very young age that actions are more and alert her supervisor? In a company where
powerful than words. GMP is practiced as a lifestyle, Jill stops and
alerts her supervisor. There may be something
Moments of Truth wrong with that particular piece of equipment.
Every person in a company makes decisions every Heres an example from an FDA 483 Observation
day. Some people make bigger decisions than oth- report (4):
ers, but all employees all make decisions that impact Bad cartons had been handwritten in black ink

24 Special Edition: A Roadmap to GMP Compliance Part 3


Anne Greene, Kelly Waldron, Nuala Calnan

on the cardboard boxes. The cartons were in unre- very best products and services to our customers.
stricted status in their computer inventory system. It is those companies who have the highest per-
Here was a Moment of Truth. Someone determined centage of personnel that are actively engaged that
that something was wrong with those cartons and will outperform their competition. Management
took action to mark them, but they did not follow needs to provide a work environment that encourages
up by having their status changed in their inventory people to be actively engaged in their work. The com-
system nor did they remove the cartons from the mitment to do the right thing at all levels will assure
production area, as evidenced by the FDA inspector customers that they will consistently receive safe,
discovering them. pure, and effective products.

Making GMP a Lifestyle REFERENCES


Review FDA Warning Letters and 483 Observations 1. R. Lopez, Most workers hate their jobs or have checked out,
reports in preparing for your GMP training sessions. Gallup says, LA Times, 2013, available here.
Identify how each observation is related to a Moment 2. Amazon Offers Employees $5,000 to Quit, ABC News, 2014,
of Truth. These often reveal the subtleties and nu- available here..
ances of complying with GMP. 3. FDA, Warning Letter Issued to APP Pharmaceuticals, LLC
Use these examples in GMP training sessions to 2/22/12, Inspections, Compliance, Enforcement, and Criminal
generate dialogue and discussion about complying Investigations, 2012, available here..
with the intent of the regulations and a company 4. FDA, FDA 483 Issued to McNeil Consumer Healthcare, Elec-
policies and procedures. The goal should be to obtain tronic Reading Room, 2010, available here.
everyones commitment to quality and providing the

Special Edition: a roadmap to gmp compliance Part 3 25


Steven S. Kuwahara

The FDA Compliance Program


Guidance for GLPs - General
Steven S. Kuwahara

ABSTRACT
This article provides an overview of the US Food and Drug Admin-
istrations good laboratory practice guidelines found in the compliance
program guidance manuals.

Making a Difference
The enforcement of good laboratory practices (GLPs) is often left to the
BioResearch Monitoring (BiMo) program of the US Food and Drug Ad-
ministrations Center for Drugs Evaluation and Research (CDER). BiMo
inspections have a reputation of being very thorough and conducted
by knowledgeable investigators. It is thus valuable to have some insight
into the guidelines that these investigators are trained to follow. These
guidelines may be found in the compliance program guidance manuals
(CPGMs) that are available online at the FDA website (1).
The CPGMs are internal FDA documents that provide guidance and
instructions for FDA investigators who conduct inspections. For the
worker dealing with the GLP guideline, the most interesting CPGM is
the one specifically covering the GLPs. This document, usually referred
to as CPGM 7348.808, covers the inspection of studies that were con-
ducted under GLP rules (2). As such, it is useful for the GLP worker to
review it to be prepared for an inspection.
CPGM 7348.808 is in Chapter 48 covering bioresearch monitoring
and was issued on February 21, 2001. This 37-page document is mostly
devoted to administrative matters that will concern FDA personnel, but
not the laboratory worker. Certain sections, however, will be of interest,
and shall be discussed here.

BIMO INSPECTION PROGRAM


Although the BiMo inspection program is identified as being a CDER
program, it is clear from the title page and the background statement
of the CPGM document that it applies to the full range of products cov-
ered by the GLP, and therefore covers products regulated by all of the
FDA centers, including foods and veterinary drugs. In fact the docu-
ments background statement says:
The FFDCA (Federal Food Drug and Cosmetics Act) and Public
Health Service Act require that sponsors of FDA-regulated products
submit evidence of their products safety in research and/or marketing
applications. These products include food and color additives, animal
drugs, human drugs and biological products, human medical devices,
diagnostic products, and electronic products. FDA uses these data to
answer questions regarding:

26 Special Edition: A Roadmap to GMP Compliance Part 3


Steven S. Kuwahara

The toxicity profile of the article application that would be an Investigational New
The observed no adverse effect dose level in the Drug (IND) application for a small molecule or bio-
test system logical drug and an Investigational Device Exemption
The risks associated with clinical studies involv- (IDE) application for a device. In turn, this supports
ing humans or animals the idea that in the sequence of drug or device de-
The potential teratogenic, carcinogenic, or other velopment, the GLP regulations come into play well
adverse effects of the article before clinical trials begin.
The level of use that can be approved The FDA centers or the headquarters of the Office
The importance of nonclinical laboratory studies to of Regulatory Affairs initiate the inspections. They
FDAs public health decisions demands that they be are conducted without prior notification when com-
conducted according to scientifically sound protocols mercial laboratories are inspected, but university and
and with meticulous attention to quality (2). government laboratories will receive a notification
This statement makes it clear that conclusions letter from the BiMo program coordinator, at least
stated above and other possible adverse effects of a for the initial inspection. There is a provision that
product are, at least initially, dependent upon good inspections of government or university laboratories
GLP studies. In the past, some of these studies have should be done as soon as possible after the labora-
been conducted with a complete or partial disregard tory has received the letter.
of GLP requirements. This behavior was partially
justified by the lack of close scrutiny of these stud- BiMo Inspections
ies. There was a belief that only human trials could The document describes the two types of inspec-
show the true nature of any lack of safety, and non- tions:
clinical studies were only valuable for finding major 1. Surveillance Inspections
adverse properties. The idea that it was unethical to Surveillance inspections are periodic, routine
ask human volunteers to consume a poorly tested determinations of a laboratorys compliance with
product did not appear to be a major concern. This GLP regulations. These inspections include a facil-
has changed with the increasing sophistication of the ity inspection and audits of on-going and/or recently
public that has produced greater demands for a high completed studies
level of product safety. 2. Directed Inspections
With the pressure on FDA to approve new prod- Directed inspections are assigned to achieve a spe-
ucts more rapidly, their reliance on good GLP-regu- cific purpose, such as:
lated studies has increased proportionally. This will a. Verifying the reliability, integrity, and compli-
also increase with the advent of biosimilars where the ance of critical safety studies being reviewed in
importance of GLP studies will rise in proportion to support of pending applications.
the reduction of human clinical trials. This will mean b. Investigating issues involving potentially
that the quality of the GLP studies will become more unreliable safety data and/or violative conditions
important to FDA reviewers and to manufacturers, brought to FDAs attention.
who will use them to gauge the toxicity, pharmaco- c. Re-inspecting laboratories previously classified
kinetics, and pharmacodynamics of new products OAI [Official Action Indicated] (usually within
especially in relation to a reference product. 6 months after the firm responds to a Warning
Letter).
BiMo Objectives d. Verifying the results from third party audits
The objectives of the BiMo GLP inspection program or sponsor audits submitted to FDA for consider-
are given in section II.A of the document: ation in determining whether to accept or reject
To verify the quality and integrity of data submit- questionable or suspect studies (2).
ted in a research or marketing application Surveillance inspections are the routine inspec-
To inspect (approximately every two years) tions that are conducted on establishments known to
nonclinical laboratories conducting safety stud- the agency. The problem here is that there are many
ies that are intended to support applications for laboratories, especially contract testing laborato-
research or marketing of regulated products ries, that only do GLP-regulated work when a client
To audit safety studies and determine the degree requires it. Unlike the established GLP laboratories,
of compliance with GLP regulations (2). these organizations often have untrained workers
As with the GLP regulations themselves, these and supervisors who only have a hazy idea of what
objectives are aimed at verifying the information sup- GLP is. These laboratories only seem to come to the
porting the idea that the products are safe. The first agencys attention when mentioned in an applica-
objective directs attention to the data in a research tion, causing them to become the target of a directed

Special Edition: a roadmap to gmp compliance Part 3 27


Steven S. Kuwahara

inspection. For example, it would be relatively easy for the study


The inspection team should be composed of at director to unduly influence the actions of the head of
least two people. The team leader will be a field in- the IACUC or the QAU. This could occur if the study
vestigator, and the second person will be a field ana- director and the head of the IACUC or QAU appear
lyst or headquarters person who serves as a scientific to be independent, if the study director were a senior
or technical support for the team leader. This usually member of the test facilitys management while either
results in having experienced investigators including the head of the QAU or the IACUC were relatively
at least one person chosen for knowledge about the junior employees of the test facility. These situations
technical or scientific issues that will be encountered. have been seen in hospital laboratories when the
Part III of the document covers the inspections study director was a senior physician or surgeon and
themselves and the general instructions tell the inves- the head of the QAU, IACUC, or the IRB was a junior
tigators to obtain copies of three documents. These, physician or surgeon.
along with the instructions to the investigators, are as
follows: Floor Plan
Organization ChartIf the facility maintains an The floor plan is important for judging the degree
organization chart, obtain a current version of of isolation of each study and the potential for cross
the chart for use during the inspection and sub- contamination of test samples. In some company lab-
mit it in the EIR. oratories, routine potency and safety tests conducted
Facility Floor-plan DiagramObtain a diagram under GMP rules may coexist with other studies that
of the facility. The diagram may identify areas are being conducted under GLP rules or under no
that are not used for GLP activities. If it does not, rules at all. Also GLP rules require the physical sepa-
request that appropriate facility personnel iden- ration of studies, animal species, test articles, feed,
tify any areas that are not used for GLP activities. and animal wastes. In some testing laboratories, the
Use during the inspection and submit it in the use of single sample preparation rooms has led to the
EIR. cross contamination of all samples processed in the
Master Schedule SheetObtain a copy of the room, while in others the absence of sample prepara-
firms master schedule sheet for all studies listed tion areas has led to the contamination of the whole
since the last GLP inspection or last two years laboratory, including the technicians.
and select studies as defined in 21 CFR 58.3(d). The master schedule allows the investigator to
If the inspection is the first inspection of the draw conclusions about the workload in the labora-
facility, review the entire master schedule. If tory and, by cross checking against the organization
studies are identified as non-GLP, determine the chart, the adequacy of staffing. The potential for
nature of several studies to verify the accuracy of cross contamination among test articles may also be
this designation. See 21 CFR 58.1 and 58.3(d). assessed, as well as the adequacy of study separation
In contract laboratories determine who decides if a in the light of the floor plan. It is fairly easy to use the
study is a GLP study (2). organization chart, floor plan, and master schedule
The experienced auditor will note that items A and to see which areas are used for each study and which
B are often requested when conducting establishment employee works on each study. Also the ability of the
audits. However, in the GLP context they, and item QAU to monitor the studies can be similarly assessed.
C, have certain meanings that were defined in the These three documents are important tools in the
regulations. When the term facility is used in GLP management of the test facility, and the absence of
work, it is important to remember that it refers to the even one of them should raise questions about the
test facility and not the whole establishment in which ability to manage the test facility. It is important to
it may be located. Similarly, the master schedule sheet have the test facility management maintain up-to-
only refers to the schedule of GLP-regulated studies date versions of the organization chart and floor plan,
and not all of the activities of the firm. and the QAU must maintain a current copy of the
The organization chart will be of interest because master schedule sheet.
it will show the reporting relationships among test
facility management, members of the institutional Inspections
animal care and use committee (IACUC), person- What will the investigators inspect? The obvious
nel actually conducting a given study (including the answer is that they will inspect the operations of the
study director), and the members of the quality assur- GLP test facility, but in GLP work there are specific
ance unit (QAU) for a study. These reporting relation- studies that are conducted by the test facility, so the
ships are important when considering the potential next point of interest is how are the studies selected
for conflicts of interest regarding specific studies. for inspection? The answer is found in section III.A.5,

28 Special Edition: A Roadmap to GMP Compliance Part 3


Steven S. Kuwahara

which reads: This information should be used to schedule


5. Identification of Studies inspections of on-going laboratory operations, as
a. Directed InspectionsInspection assignments well as equipment and facilities associated with
will identify studies to be audited. the study. If there are no activities underway in
b. Surveillance inspectionsInspection assign- a given area for the study selected, evaluate the
ments may identify one or more studies to be area based on on-going activities.
audited. If the assignment does not identify a 3) Completed StudiesThe data audit should
study for coverage, or if the referenced study is be carried out as outlined in Part III, D. If pos-
not suitable to assess all portions of current GLP sible, accompany laboratory personnel when they
compliance, the investigator will select studies retrieve the study data to assess the adequacy of
as necessary to evaluate all areas of laboratory data retention, storage, and retrieval as described
operations. When additional studies are selected, in Part III, C 10 (2).
first priority should be given to FDA studies for The studies of interest are those that are critical to
submission to the assigning Center. NOTE: Stud- the assessment of the safety of a product and the ones
ies performed for submission to other govern- that cover a wide range of test facility activities.
ment agencies, e.g., Environmental Protection
Agency, National Toxicology Program, National CONCLUSION
Cancer Institute, etc., will not be audited without There are many interesting and important points in
authorization from the Bioresearch Monitoring CPGM 7348.808 that cover the inspection of studies
Program Coordinator (HFC-230). conducted under GLP rules. Personnel working in a
However, this authorization is not necessary to GLP environment should review CPGM 7348.808 in
briefly look at one of these studies to assess the ongo- preparation for an inspection.
ing operations of a portion of the facility.
1) Criteria for selecting on-going and completed REFERENCES
studies 1. http://www.fda.gov/ICECI/ComogramManual/default.
Safety studies conducted on FDA-regulated prod- htm#bimoplianceManuals/C.....
ucts that have been initiated or completed since 2. CPGM 7348.808, Good Laboratory Practices (Non-clinical
the last GLP inspection. Laboratories), February 21, 2001. (Note: the letters preceding
Safety studies that encompass the full scope of the numbers are sometimes given as CP or CGM) GXP
laboratory operations.
Studies that are significant to safety assessment, ARTICLE ACRONYM LISTING
e.g. carcinogenicity, reproductive toxicity, chronic BiMo BioResearch Monitoring
toxicity studies. CDER Center for Drugs Evaluation and Research
Studies in several species of animals. CPGMs Compliance Program Guidance Manuals
The investigator is encouraged to contact the Cen- GLP Good Laboratory Practice
ter for guidance on study selection. IACUC Institutional Animal Care and Use Committee
2) Ongoing StudiesObtain a copy of the study IND Investigational New Drug
protocol and determine the schedule of activi- IDE Investigational Device Exemption
ties that will be underway during the inspection. QAU Quality Assurance Unit

Special Edition: a roadmap to gmp compliance Part 3 29


David W. Vincent

Quality and Compliance in the


Asian Pharmaceutical Industry
David W. Vincent

Note: This piece is an update to a commentary written in 2011 for the


Journal of Validation Technology entitled Quality and Safety of Asian Phar-
maceutical ProductsEU and US Regulatory Agency Activities and New
Chinese GMP Regulations.

Introduction
The Asian pharmaceutical manufacturing industry is becoming more
complex, with some Asian countries moving more aggressively than
others to supply their to regulated markets such as the US and Europe.
Emerging markets, poised to account for about 45% of the worlds
gross domestic profit by 2018, are projected to represent the majority of
growth in pharmaceutical sales in the next five years (1).
According to a recent report, in 2008, the global active pharmaceuti-
cal ingredients (APIs) market (including both captive and merchant)
worth was $91 billion; it increased to $113 billion (USD) in 2012. From
2008 to 2012, the global API market increased at an average annual
growth rate of 5.6%, though it is less compared to an annual growth
rate of 7.2% from 2004 to 2008. The market, which stood at $113 bil-
lion in 2012, is expected to grow at a compound annual growth rate of
around 8% during 20122017 (1).
Up to 40% of finished drugs used by US patients are manufactured
abroad, and 80% of the active ingredients and bulk chemicals used in
US drugs come from foreign countries (1).
Currently, Asia leads the world in the manufacturing of APIs. While
the US and European pharmaceutical industries are experiencing little
or no growth in API manufacturing, Asian producers are seeing high
growth rates, both from domestic demand and export sales. According
various records, Chinas API producers generated estimated revenues of
$4.4 billion in 2005 and are projected to make sales of $10.3 billion in
2013. Indias API producers had sales of about $2 billion in 2005 and
$20.8 billion in 2013; they are forecast to grow to $35.1 billion by 2016.
Japan accounts for over 50% of pharmaceutical sales in Asia-Pacific, fol-
lowed by China, which is a distant second with 19% (2).
Chinese pharmaceutical companies are primarily oriented towards
supplying their own domestic market. Thus, they tend to place less
emphasis on external good manufacturing practices (GMP) compliance.
Indian API manufacturers, on the other hand, are focused on export
sales to highly regulated global markets. They have thusly developed
considerable expertise in complying with global GMPs and supplying
documentation to foreign regulatory agencies for drug master files.
With rising costs of pharmaceutical products and new low-cost com-
petitors, manufacturing efficiency is becoming more important to Asian

30 Special Edition: A Roadmap to GMP Compliance Part 3


David W. Vincent

API manufacturers. They are looking to regulated- ing failures that can result in product seizures and
markets for growth, and that means compliance to recalls. Yet, theses documents are not intended to cre-
GMPs. The issue of cost and safety for the producers ate new requirements for pharmaceutical manufac-
of API manufacturing equipment is also a significant turing that go beyond those established in the current
driver for growth in this industry. regulations, nor are the publications intended to be a
In recent years, more manufacturing facilities APIs guide for the conduct of FDA and EU inspections.
have relocated to Asia. Approximately 80% of the In Asia, Japan is currently the leader in under-
APIs used in the US and the European Union (EU) to standing the regulatory and compliance requirements
manufacture finished pharmaceutical products come required to produce a high-quality pharmaceutical
from Asia, with China and India accounting for much product. However, the pharmaceutical sector in India
of the supply. However, there is a major concern has been one of the fastest growing sectors of its
with foreign drug manufacturers producing certain economy over the past several years, and it is fore-
proprietary drug products in Asia; this is due to weak casted to continue growing. Similar to China, India is
intellectual property laws. now one of the most important healthcare markets in
In addition, some Asian countries make it difficult the world, having its own domestic market as well as
for small foreign support companies (e.g., validation, a huge pool of clients for exports. India is considered
quality, laboratory, and technical consultant services) to be ahead of China in terms of its control of quality
to establish local businesses and to enter the mar- over pharmaceutical manufacturing. Its approach to
ket. Due to this, many larger foreign and local API quality is much different from the China API manu-
and pharmaceutical companies have to import this facturers, with focus on long-term commitment to
knowledge based services at much higher cost then quality and compliance. Quality is considered to be
could be provided by local businesses. a part of the Indian business model and investment
These, plus other issues, are limiting factors that into strong quality strategy will, in the long-term,
have prevented some foreign companies from relocat- make them a successful player in the global pharma-
ing to certain Asian countries. ceutical markets. Additional Asian markets such as
Taiwan and Singapore are committed to long-term
Quality and Safety Issues investment into the Global API markets. Hopefully,
Asian API and drug manufacturers can better un- other Asian markets will follow suit and commit to
derstand how they can achieve regulatory compli- investing into a strong quality strategy, but the pres-
ance and avoid production stoppages and recalls by sure to maximize profits by minimizing quality will
following new guidance issued by the US Food and always be challenge for the API industry.
Drug Administration and the European regulatory
agencies. In addition, Asian API industry must also Quality and Safety Issues
understand that education and training is a key busi- There has been an accelerating rate of change within
ness investment that in the long run, will produce the industry over the last 20 years, with massive glo-
a safe and effective product. If most Asian countries balization occurring driven by the pressure to drive
focus on developing a strong quality and compliance drug-manufacturing costs down.
philosophy in the long-term, their business goals for US and EU manufacturers supplied 90% of API
financial success will be achieved needs 20 years ago; now, they are supplying less
The international regulating bodies have pub- than 20%. At the same time, the number of generic
lished various guidances, (e.g., Eudralex Part 2 Basic medicines accounted for less than 5% 20 years ago
Requirements for Active Substances used as Starting whereas now they account for over 50%, with much
Materials (3), Pharmaceutical GMPs for the 21ST of the API coming from Asia.
Century A Risk-Based Approach Final Report (4), The above chart shows the cost structure dif-
and Quality Systems Approaches to Pharmaceutical ferential between European-US manufacturers and
Current Good Manufacturing Practice (GMP) Regula- manufacturers in India and China. Key to numerical
tions) (5) in order to help manufacturers maintain superscripts: 1) includes cost of goods sold without
consistent high quality and improve efficiency prod- cost for raw material, 2) assessment of direct person-
uct. FDA has also published a new process validation nel cost reduction from approximately 60 to 6 total
guidance document titled Process Validation: General salary dollars (tsd)/FTE (1=$1.25), indirect person-
Principles and Practices (6). nel cost reduction factor of two, 3) assessment of
The aim of these documents is to help both US direct personnel cost reduction from approximately
and international manufacturers produce drugs more 6 to 3 (tsd)/FTE (7).
efficiently, which should help lower costs and prevent Consumers take it for granted that the medicines
shortages of critical medicines due to manufactur- they buy are manufactured to the highest standards

Special Edition: a roadmap to gmp compliance Part 3 31


David W. Vincent

Figure 1: Assessment of API manufacturing costs by region (in $ mil., basis 2004)

and expect the regulatory authorities to ensure that faced with a daunting task to tackle the escalating
this remains the case. Manufacturing plants have problem of inspecting all API and pharmaceutical
to meet strict standards to ensure the products they manufacturing sites in Asia.
make meet the requirements of the GMPs and are Records indicate that an estimated 7% of all for-
not contaminated by poor manufacturing practices. eign API facilities are inspected by FDA each year,
However, in reality, there is a high probability that compared to 97% of US facilities inspected every
the drugs on the shelves of European and United two years. Most pharmaceutical companies in the US
States pharmacies will contain APIs that were made have expressed that this comparison is unfair given
in Asian manufacturing facilities that have never been that the level of controls, deterrents, or sanctions on
inspected, and the new regulations due to come into foreign facilities are not of the same standards.
force in the future will still not solve this problem. There are almost 7,000 foreign firms that import
Compliance with GMP regulations requires that pharmaceutical products into the US listed on the
batches of an API remain of consistently high quality FDA database. The number of inspections performed
throughout the entire manufacturing process. Any on these firms by FDA had been criticized to be un-
changes to the process can only be implemented after representative of the number of products imported.
the impact on quality has been assessed, and pro- For example, in 2007, 13 out of 714 Chinese import-
cess change records must be kept so that all actions ers of pharmaceutical products into the US were in-
can be traced, if necessary. However, without GMP spected by FDA whereas 65 out of 410 were inspected
or proper understanding of GMPs, the consistency in India (8).
and the quality of the batches cannot be assured. All Furthermore, in 2007, warning letters were issued
process changes should be assessed, regardless of its by FDA for GMP non-compliance to two Chinese
effect on product quality, and the lack of deficient or pharmaceutical manufacturers, resulting in denial of
non-conformance records makes investigations nearly entry of products into the US. This was considered
impossible. This means that a number of problems to reflect a high proportion. From 2008 to 2013,
are more likely to occur without being noticed, such FDA has inspected 785 establishments in China.
as changes in impurity profile, cross contamina- Out of the 785 site inspections, 260 inspections were
tion with other APIs because of ineffective cleaning, conducted by the Center for Drug Evaluation and
or altered crystal forms or particle sizes, which can Research (CDER), 228 inspections were conducted
impact bioavailability. by the Center for Devices and Radiological Health
(CDRH), 42 inspections were conducted by the Cen-
EMEA and FDA Approaches to GMP Inspections ter for Veterinary Medicine (CVM), and 255 inspec-
Performed in Asia tions were conducted by the Center for Food Safety
European Medicines Agency (EMEA) and FDA are and Applied Nutrition (CFSAN) (8).

32 Special Edition: A Roadmap to GMP Compliance Part 3


David W. Vincent

Although FDA uses a risk-based process to develop terfeits of their products, they will take action. Last
a prioritized list of foreign establishments for inspec- year China cracked down when it found companies
tions to monitor the quality of marketed drugs, few using gelatin from scrap leather that contained toxic
are completed in a given year. This prioritized list chemicals to produce drug capsules. It sent 7 people
was used to select foreign establishments for in- to prison and closed dozens of production facilities.
spection in fiscal year 2014. According to the FDA It will take about a year to fill out the staff in
website, for fiscal year 2013, actual unique count of China, Hickey told Bloomberg. Some earlier hires
foreign inspections includes 67 Office of International moved on when they couldnt get visas. The FDA can
Programs (OIP) inspections (17 for China and 50 for still send U.S.-based personnel, as it has for years.
India) (9). Hickey hedged when asked if having more inspectors
FDA is supposed to inspect all facilities that supply there will lead to more actions. One thing we have
the US with APIs, regardless of location. This means seen with our inspections overseas in India, and here
that there are numerous facilities selling APIs to in China, is that over time, inspectors are better able
the US, compared to the thousands of uninspected to identify specific problems in the firms. (10).
plants whose products reach the European market. It The foreign inspection process involves unique
also leads to the complex situation of European API situations that are not encountered domestically. For
manufacturers selling their ingredients for use in the example, FDA relies on staff that inspects domes-
US, while they are left unable to sell them in Europe tic establishments to volunteer for foreign inspec-
because they are undercut by lower cost and unin- tions, and unlike domestic inspections to monitor
spected Asian manufacturers. This unfair, one-sided the quality of a marketed drug, FDA does not arrive
condition puts the European API manufacturers at an unannounced at a foreign establishment. It also lacks
unfair disadvantage. the flexibility to easily extend foreign inspections if
FDA now performs GMP inspections on selected problems are encountered, due to the need to adhere
pharmaceutical enterprises, regardless of location. to an itinerary that typically involves multiple inspec-
However, there has been recent criticism of this ap- tions in the same country. Finally, language barriers
proach, citing that the number of pharmaceutical can make foreign inspections more difficult than
enterprises FDA has been able to inspect is not nearly domestic ones. FDA does not generally provide trans-
representative of the total number of foreign suppliers lators to its inspection teams. Instead, they may have
exporting pharmaceutical ingredients to the US. to rely on an English-speaking representative of the
FDA has started to establish more affiliates in the foreign establishment being inspected, rather than an
main exporting countries. It recently opened affili- independent translator. Most Chinese API producers
ates in Chinese cities such as Beijing, Shanghai, and know this is an issue and they sometimes exploit this
Guangzhou to strengthen its global presence. FDA weakness by redirecting the inspection process.
inspections in exporting countries are expected to
increase, primarily in China and India. By opening Bloomberg Article (11)
these offices and relocating approximately 13 em- More recently (Apr 3, 2014), Anna Edney of Bloom-
ployees in order to certify inspections of U.S.-bound berg wrote the following concerns regarding product
Chinese exports, FDA quality concerns became more safety in China in a piece entitled Drug Quality in
apparent. As FiercePharma reports: China Still Poses Risks for U.S. Market. The article
The FDA is adding 19 people to the staff of 9 it discusses the quality issues in Chinese manufactur-
already has there, but 9 of those are food inspectors, ing, a study on overseas manufacturing of Lipitor, and
according to Bloomberg. The FDA opened an office the international staffing situation at FDA. Readers
there in 2008 after being hammered for lax oversight are recommended to read the article here.
when tainted Chinese heparin led to dozens of deaths
in the U.S. The FDA has been trying to boost its pres- FDA Reduces Domestic Inspections
ence in China for more than a year, but the Chinese Furthermore, as FDANews reports, FDA is scaling
government blocked visas of FDA employees assigned back the number of routine quality inspections it
there. U.S. Vice President Joe Biden was able to get plans to conduct in the U.S. each year by 40 percent
a final commitment for the expanded FDA presence in favor of conducting more inspections overseas. The
during a visit to Beijing last week shift is part of a broader agency push to improve the
While China is believed to be understaffed and quality of drugs imported into the U.S. (12).
unable to keep on top of its burgeoning drug produc- Based on FDAs new strategy, which necessitated a
tion industry, Western drugmakers say it is making reduction in domestic inspection in response to great-
more of an effort. When drugmakers provide Chinese er perceived risks abroad, difficult decisions had to
authorities with evidence of companies making coun- be made due to budget restraints. This does not mean

Special Edition: a roadmap to gmp compliance Part 3 33


David W. Vincent

that domestic drug manufacturing should adopt sometimes under pressure to accept a vendor based
lower level of GMP standards, rather that the FDA on cost and efficiency, rather than quality and full un-
has shifted some of the onus onto the manufacturers derstanding of GMPs. This is one of the reasons there
themselves. From the authors personnel experience, should be some discussion on the development of a
this strategy for foreign inspections should develop global harmonization for second and third-party GMP
interesting results auditing standards for the pharmaceutical industries.
This recommendation is based on the lack of re-
Major Areas of GMP Concerns sources within the regulatory agencies to perform the
According to statistical data based on foreign inspec- inspection necessary to safeguard the public health.
tion observations, the following are the top 15 major Furthermore, another suggested resolution to the cur-
areas of GMP deficiencies for 2008 to 2014 (2): rent problem would be to have the regulatory agencies
Quality system elements/procedures documenta- certify or approve consultants or consultant compa-
tion nies to act on their behalf to perform foreign inspec-
Design and maintenance of premises tion as per standard auditing practices. This would
Design and maintenance of equipment minimize the risk of potential adulterated products
Process validation and materials from entering foreign markets. Unfor-
Procedures and facilities sampling tunately, this suggestion may not be viable because of
Manufacturing documentation issue (falsification) current FDA policies on foreign inspections.
Potential for microbiological contamination One of the major concerns for some of these drug
Specification and testing documentation manufacturers is their inability to produce a sterile
Facilities and equipment status labeling drug product according FDA GMPs. With the ever-in-
Environmental monitoring creasing pressure to be competitive and keep the cost
Supplier and contractor audit and technical agree- of drug production low, there is real temptation to cut
ments corners. The culture of strict inherence to the GMPs is
Equipment validation relative new concept in Asia. With past issues related
Hygiene/clothing to adulterated drug products from foreign countries
Duties of key personnel entering the US, there must be stricter regulatory
Potential for chemical/physical contamination. oversight and controls for final filled sterile drugs
Energy conservation (Many of the API producers entering the US market.
in China consider it acceptable to shutdown their
clean utilities at night and restarting them in the Conclusion
morning without considering of the impact on the Asian pharmaceutical and API suppliers need solu-
environmental conditions of these systems [e.g., tions that will enable them to ensure regulatory
bioburden]). compliance and address the global concerns on qual-
These above-listed deficiencies are an indication of ity and authenticity. Over the past few years, there
lack of understanding or commitment to the GMPs. arose issues that have created a need for a harmonized
These types of GMP deficiencies cause concern among approach to GMP audits of pharmaceutical and API
regulatory agencies regarding the import of APIs and suppliers in Asia. Asian markets need to develop ef-
other drug-related materials utilized in the production fective quality strategies to comply with the various
of finished drug products. Without proper inspec- GMP standards required in API and pharmaceutical
tion or oversight of foreign companies, the potential production. This has been, and will remain for some
of adulterated products entering the US or European time, a significant concern for the pharmaceutical
markets is an accident waiting to happen. industry in Asia. This article is not to be intended
While the finished drug manufacturers are ul- as a criticism of the Asian API and pharmaceutical
timately responsible for the quality and safety of markets, but rather a source of constructive ideas to
the drugs they produce, this should not alleviate promote further growth of the industry.
or remove the oversight by the regulatory agencies.
There should be strong harmonization and guidance REFERENCES
between the regulatory agencies and finished drug 1. Deloitte, Opportunities in Chinas Pharmaceuticals Market,
manufacturing in regards to oversight and inspection Deloitte Analysis.
of foreign imports of APIs. To date, there have been 2. US Food and Drug Administration and European Medicines
no guidance documents related to the standardization Agency websites.
of auditing of APIs or other drug materials used in fin- 3. Eudralex, Part 2 Basic Requirements for Active Substances used
ished drug products. As a result, auditing standards as Starting Materials (Brussels, BE, 2010).
are left up to the finished drug manufacturers who are 4. FDA, Pharmaceutical GMP for the 21st Century A Risk-

34 Special Edition: A Roadmap to GMP Compliance Part 3


David W. Vincent

Based Approach Final Report (Rockville, MD, May 2007). 8. C.H. Yin Lau, GMP Trends in Asia, Life Science Auditing.
5. FDA, Quality Systems Approaches to Pharmaceutical Current 9. Number of routine inspections completed by investigators
Good Manufacturing Practice (GMP) Regulations (Rockville, based in-country in the month, available here.
MD, Sept. 2006). 10. E. Palmer, Vice president clears way for FDA to add inspec-
6. FDA, Process Validation: General Principles and Practices tors in China, FiercePharma, 2013, available here.
(Rockville, MD, Jan. 2011). 11. A. Edney, Drug Quality in China Still Poses Risks for U.S.
7. C. Paddison, C. White, and C. Cruickshank,China and India: Market, Bloomberg, 2014, available here.
Outsourcing Beyond the Comfort Zone, Pharmaceutical 12. FDA to Slash U.S. GMP Inspections by 40 Percent,
Technology, 2005. FDANews, available here.

Special Edition: a roadmap to gmp compliance Part 3 35


Kelly Waldron

Integration of Risk Management


Principles into the Quality
System: Risk-based Impact
Assessment
Kelly Waldron

Introduction
As the principles of quality risk management have become increasingly
pervasive in the biopharmaceutical and medical device industries, a
point of saturation has been reached with respect to the risk toolkit.
Standard risk assessment techniques such as hazard operability analy-
sis (HAZOP), hazard analysis and critical control points (HACCP), and
failure modes and effects analysis and its derivatives (FMEA, FMECA,
FMEDA) have proven effective in increasing understanding of the gen-
eral level of risk inherent in products and processes and have allowed
the industry to realize tangible gains through the reduction of unac-
ceptable risks and the development of more targeted, robust preventive
actions. However, in seeking to apply quality risk management as an
enabler of the overarching quality system (1, 2), this standard toolkit
reveals some inherent limitations; specifically, none of these tools were
designed for use in a GxP environment (3). As a result, when using
such template tools to make risk-based decisions within a given quality
system element, the user is left to interpret the output of the risk tool in
a rather subjective way: what does the level of risk mean with respect
to the quality of products or the safety of patients? Since answering this
question often forces one to invoke concepts and make decisions based
on information extrinsic to the risk assessment, the validity of the risk
tool in a specific GxP context may be called into question. Perhaps the
best path forward, therefore, is not to use existing tools in a novel way
but rather to develop new risk tools to meet specific GxP objectives.
This paper will focus on one such customized approacha risk-based
impact assessment (RBIA) designed to determine the acceptability of
potential impact scenarios that may arise from a process deviation.

Laying the Foundation


The general process and goals of deviation investigation are famil-
iar to each of us. Domestic and international regulations require any
deviation from written, approved procedures to be investigated and
explained (4, 5). Such an investigation commonly includes the determi-
nation of the root cause of the deviation, proposal of appropriate correc-
tive and preventive action (CAPA), and the determination of the impact
of the deviation on product quality. While a variety of root cause analy-
sis tools are available to help satisfy the first goal of the investigation,
few, if any, practical tools exist to assist with the impact assessment.
In the authors experience, deviation impact is typically determined

36 Special Edition: A Roadmap to GMP Compliance Part 3


Kelly Waldron

by building a logical argument stemming from the re- impact could be on a portion of a batch, the whole
sults of the investigation. Since these sorts of impact batch, other batches linked in space or time with the
assessments are often the product of a single assessor, deviation, or implicated equipment or facilities. These
the robustness of such arguments vary widely in ac- potential impact scenarios will then be ranked inde-
cordance with individual experience and potentially pendently through the risk analysis process.
undisclosed bias, subjectivity, or assumptions intro-
duced by the assessor. Within the scope of any indi- Risk Analysis
vidual deviation, the strength of such a narrative and Risk is commonly defined as the combination
the resultant product disposition decision are judged of the probability of occurrence of harm and the
based on their own merits. However, when multiple severity of that harm (1, 6). In order to capture these
impact assessments are viewed together as represen- dimensions, many risk tools, including RBIA, employ
tative of the quality system output, inconsistencies the terms likelihood and severity within the risk
can bubble to the surface. This need not be the case. analysis process. The advent of International Organi-
Replacing or supplementing the current status quo zation for Standardization (ISO) 31000 introduced a
for deviation impact assessments with a common fresh perspective of risk by defining it as the effect of
RBIA tool can increase the consistency of the process uncertainty on objectives (7). In this way, ISO 31000
within the quality system and reduce the subjectivity encourages to consider not only the likelihood and
and bias inherent within such assessments. consequences of a risk, but also the level of certainty
surrounding those aspects. ICH Q9 echoes this
Risk-based Impact Assessment Tool Design sentiment by noting the evaluation of risk to quality
Since the risk-based impact assessment tool is a type should be based on scientific knowledge (1). In
of risk assessment, it follows that the methodology a GxP environment, scientific knowledge can take
should mirror the quality risk management lifecycle. many forms, such as direct testing through appropri-
ICH Q9 Quality Risk Management separates a risk ate design of experiment (DOE), awareness of the ex-
assessment into three component parts: risk identifi- istence and effectiveness of good manufacturing prac-
cation, risk analysis (including risk estimation), and tice (GMP) controls through documented evidence
risk evaluation (1). The sections that follow explore and monitoring, and inductive reasoning based on
each of these components in detail as they relate to sound scientific principles. Since the relative strength
the RBIA tool of the scientific knowledge can vary, and may also be
interpreted in different ways (3), explicit disclosure
Risk Identification of such information is often necessary to preserve the
An important distinction within the RBIA tool is one integrity of the assessment. RBIA directly addresses
common to the quality risk management field: the the question of uncertainty, ambiguity, and subjec-
difference between a hazard and the resultant harm. tivity by including a confidence factor. Likelihood,
ICH Q9 defines a hazard as a potential source of confidence, and severity and their relationship to the
harm and harm as the damage to health, includ- overarching RBIA tool will be discussed in detail in
ing the damage that can occur from loss of product the sections to follow.
quality or availability (1). When placed into the
context of a deviation, the deviation event itself can Risk Estimation
be considered the hazard whereas the harm is the po- To guide the assessment and ensure consistency of
tential impact(s) that may result from that deviation. risk estimation, pre-defined rating criteria should be
For example, a deviation may be raised to address a established for each of the RBIA dimensionslikeli-
loss of high-efficiency particulate air (HEPA) filtration hood, confidence, and severity. It is important to note
during batch manufacturing (the hazard), which in that any such criteria should be assessed prior to
turn may result in microbial or particulate contami- beginning the risk estimation process. The establish-
nation of product, equipment, or facilities (the harm). ment of clear, objective risk-rating criteria that are
While different risk tools address each of these directly tailored to the task at hand will prove invalu-
concepts in different manners, the scope of concern able as the exercise progresses.
with respect to an RBIA is clearly the harmthat is, The first step of risk estimation is to determine the
seeking to understand the risk associated with the likelihood of each identified impact scenario. Since
potential harm to patient safety, product quality, and likelihood is a ubiquitous concept in the risk do-
GxP compliance. The risk identification portion of main, it will not be belabored here. It is important to
the RBIA process should, therefore, focus on defin- reinforce that, within the scope of RBIA, the likeli-
ing all potential impact scenarios within the scope hood rating is directly related to the potential impact
of concern of a specific deviation event, whether the statement(s) outlined during the risk identification

Special Edition: a roadmap to gmp compliance Part 3 37


Kelly Waldron

Rating Criteria

Remote The potential impact is unlikely to have occurred.

Average The potential impact is moderately likely to have occurred.

Likely The potential impact is likely to have occurred.


Table I: Example of Likelihood Rating Scale and Criteria.

Rating Simple Criteria Detailed Criteria

Supporting data was gathered through targeted (DOE) in con-


ditions mirroring those present in the deviation event (e.g.,
using identical buffer formulations, actual product, etc.).
All supplemental controls in place at the time of the deviation
High degree of confi- event were evaluated and the effectiveness confirmed (e.g.,
dence in the likelihood multiple redundant controls in place to prevent occurrence of
High rating assigned. Presence the impact).
of potential impact (nearly) Samples taken to investigate the product quality were rep-
confirmed/refuted through resentative of the entire batch with the appropriate level of
the investigation process. statistical confidence (e.g., multiple aliquots taken from a
homogeneous bulk solution).
Applicable assays used to test samples were validated with
the appropriate level of precision, reproducibility, and robust-
ness to detect the potential impact.

Supporting data gathered through experiments performed


using conditions was comparable, though not identical, to
those present in the deviation event (e.g., using similar buffer
class, representative product, etc.)
Moderate degree of con- The majority of supplemental controls in place at the time of
fidence in the likelihood the deviation event were confirmed as effective; some con-
rating assigned. Presence/ trols were not evaluated or the effectiveness at the time of the
Medium absence of potential impact event could not be confirmed.
can be estimated based on Samples taken to investigate the product quality represented
the results of the investiga- a statistical cross-section only or may not provide assurance
tion of the entirety of the batch (e.g., discrete vials from a filled
lot).
Applicable assays used to test samples were validated with
levels of variability, precision, or robustness that may hinder
the detection of the potential impact, or the impact can be
detected through manual visual inspection only.

Low level of confidence Supporting data gathered from external literature may not
in the likelihood rating represent the conditions of the actual deviation event, or no
assigned. Presence/absence supporting data is available.
Low of potential impact could The majority of supplemental controls in place at the time of
neither be confidently the deviation event was not evaluated or could not be con-
confirmed nor estimated firmed as effective.
based on the results of the Potential impact cannot be directly observed, tested, or
investigation. detected.
Table II: Example of Confidence Rating Scale and Criteria.

38 Special Edition: A Roadmap to GMP Compliance Part 3


Kelly Waldron

process. Likelihood answers the question, How The multiple dimensions of confidence should be
likely is it that this potential impact has occurred? contained within a confidence rating scale with asso-
An example of rating criteria for likelihood is illus- ciated criteria. The level of detail associated with this
trated in Table I. scale may vary based on the complexity of the issue
Once each potential impact has been assigned a at hand and the resulting necessity to specify tangible
likelihood rating, the risk estimation process pro- inputs, as illustrated in Table II.
ceeds to the confidence dimension. The inclusion of a The purpose of the confidence factor is to temper
confidence factor in the RBIA tool links the potential the likelihood rating to account for the strength of
impact(s) directly to the deviation investigation and available evidence in support of the actual probability
explicitly addresses any sources of subjectivity, ambi- of occurrence such that situations wherein limited
guity, or uncertainty that may jeopardize the integrity supporting data around the presence or absence of
of the assessment. Confidence represents a measure a given impact scenario force the assignment of a
of the detectability of each potential impact combined more conservative risk estimation. This objective is
with an assessment of the preventive controls that achieved through the use of a preliminary risk matrix
would have mitigated its occurrence. The confidence as shown in Figure 1. To use the preliminary risk
rating therefore answers the question, How sure are matrix, simply find the intersection of the applicable
we with respect to the likelihood rating assigned to likelihood and confidence ratings to arrive at the
this potential impact scenario? To achieve this ob- preliminary risk level denoted as L1, L2, or L3.
jective, the confidence rating should account for: An analysis of the preliminary risk matrix design
The quantity and quality of data available to sup- illustrates the relationship between the likelihood and
port the likelihood rating confidence ratings. Where the confidence in a given
The depth and rigor of the deviation investigation likelihood rating is high or medium, the matrix indi-
and the resultant evaluation of the existence and cates a customary risk level commensurate with that
effectiveness of risk-mitigating controls probability rating (i.e., L1 for remote, L2 for average,
The relative representativeness of sampling and L3 for likely). However, where the confidence is
schemes used to support likelihood conclusions low, indicating a lack of sufficient scientific evidence
Any inherent weaknesses or limitations associ- to support the likelihood rating, the matrix assigns
ated with impact detection methods used to sup- a more conservative rating (i.e., low confidence in a
port likelihood conclusions remote likelihood becomes an L2, and so on). This
Any other assumptions or circumstances that preliminary risk level can be considered a calibrated
may improve or detract from the validity of con- likelihood rating that is then used, along with sever-
clusions related to product or system impact, as ity, to determine the overall risk.
appropriate. The final step in risk estimation is the assignment
Confidence of a severity rating to each potential impact. Since po-
High Low tential impact scenarios can vary significantly based
Medium
on the type of impact, a bit of discussion around
the structure of severity rating criteria is warranted.
In recent years, regulatory agencies have begun to
Likely

L3 L3 L3 develop defect classifications that directly address the


relative level of patient risk. For example, US Food
and Drug Administration defines recalls associated
with dangerous or defective products that predict-
ably could cause serious health problems or death
Average

as Class I (8), while the European Medicines Agency


L2 L2 L3 (EMA) describes Class I defects as those that are
potentially life threatening (9); Class II defects are
those that may affect the patient in a less significant
way (such as a transient illness or manageable adverse
event); Class III defects are unlikely to impact patient
health. In all cases, the defect classification reflects
Remote

L1 L1 L2 the relative severity of the impact scenario that may


result from product administration; these concepts
should be preserved during the establishment of
severity criteria within the RBIA tool.
Figure 1: Preliminary Risk Matrix A helpful way to classify such defects is to explore

Special Edition: a roadmap to gmp compliance Part 3 39


Kelly Waldron

the relationship between a given potential impact and business climate and product profile. For example,
the products design space; that is, the combination life-saving products for which there is currently insuf-
of and relationships between process parameters, ficient available inventory may warrant a higher risk
equipment components, and product attributes tolerance than a quality-of-life product with several
that collectively define product quality (10, 11). For direct competitors since the impact to patient due to
example, if a deviation is related to a critical process lack of product availability may be more significant
parameter (CPP), there is a strong potential that there for life-saving drugs than for other types of medici-
may have been an impact to a defined critical quality nal products. Therefore, it is recommended that each
attribute (CQA) of the product, which in turns calls manufacturer establish a unique risk tolerance level
product efficacy and patient safety into question (11). for each product based on the business environment
An example of severity criteria, considering design at the time of the assessment (e.g., available inven-
space relationships (for product-type impacts), good tory, comparability of competition, etc.). Any such risk
engineering practices (for equipment and facility-type tolerance must be developed considering the potential
impacts), GxP compliance, and regulatory expecta- impact to the patient rather than to business concerns
tions is detailed in Table III. such as loss of revenue.

Rating Simple Criteria Detailed Criteria


Product Equipment/Facility

Minor Insignificant or negligible Class III defects: Potential Potential impact is not
impact impact is not related to a related to a critical aspect
CQA. (CA) of equipment or
facility and does not
significantly affect GxP
compliance status.

Moderate Moderately significant Class II defects: Potential Potential impact is related


impact impact related to a CQA, to a CA but is of a nature
however, is of a nature or or magnitude unlikely to
magnitude that is unlikely affect critical or key process
to affect product efficacy or parameters. Moderate
patient safety. impact to GxP compliance
status.

Critical Significant impact Class I defects: Potential Potential impact is related


impact related to a CQA and to a CA and is of a nature
is of a nature or magnitude of magnitude likely to affect
likely to affect product CPPs. Significant impact to
efficacy or patient safety. GxP compliance status.

Table 3: Example of Severity Rating Scale and Criteria.

Once the likelihood, confidence, and severity


ratings have been determined and appropriately justi- Within the realm of RBIA, risk tolerance is captured
fied, for each potential impact scenario, the risk as- in a final risk matrix that marries the severity rating
sessment will progress into the risk evaluation phase for a given potential impact scenario with the associ-
where decisions are made regarding risk acceptability ated preliminary risk level. An example is provided
and next steps. in Figure 2. In the event a different risk tolerance is
expected for product-type impact scenarios than for
Risk Evaluation equipment or facility-type impact scenarios (for ex-
Any discussion of risk evaluation should begin with ample, situations wherein one is willing to accept less
a consideration of the relationship between risk toler- risk for equipment impact than for product impact
ance and risk acceptability (7). Decisions surrounding due to responsibility to protect patients), separate
what levels of risk are considered acceptable should matrices should be developed. The actions to be taken
be made following a thorough review of the current based on the acceptability of the risk should be de-

40 Special Edition: A Roadmap to GMP Compliance Part 3


Kelly Waldron

Risk Actions
Acceptability
(Figure 2) Product Equipment/Facility

Product acceptable for Risk is acceptable: No remedial action necessary prior to further
Acceptable
release. processing.

Product not Risk is not acceptable: Immediate remediation/CAPA required


Unacceptable
acceptable for release. before further processing can proceed.

Table 4: Example of Risk Acceptability and Actions Table.

Severit
Minor Moderate Critical
(from the Preliminary Risk Matrix, Figure 1)

Acceptable Unacceptable Unacceptable


Preliminary Risk Level

Acceptable Unacceptable Unacceptable

Acceptable Acceptable Acceptable

Figure 2: Final Risk Matrix

fined (see Table IV for an example) to ensure objectiv- ment principles into their quality systems, the need
ity of decision-making. for customized risk tools becomes apparent. Any such
Since it is expected that there will be several potential custom tools, such as the risk-based impact assess-
impact scenarios for a given deviation event, decisions ment tool described in this article, should be designed
should be made based on the highest risk presented. to directly address GxP concerns and link general risk
For example, when there are two acceptable product- principles to the specific quality system element in
type risks and one unacceptable risk, the more which the tool will be used.
conservative action (discarding of the product) should The RBIA tool expands upon foundational risk
be taken. Employing this practice will ensure that assessment methodologies to facilitate risk-based
patients, or future batches of product, are not exposed decision-making for product and process impact stem-
to any unacceptable level of risk. ming from deviation events. RBIA includes a measure
of confidence in addition to the standard likelihood
Conclusion x severity dimensions to assure compliance with the
As biopharmaceutical and medical device companies primary tenets of risk managementthat the risk
seek to achieve complete integration of risk manage- assessment is founded in the best available scientific

Special Edition: a roadmap to gmp compliance Part 3 41


Kelly Waldron

knowledge, explicitly addresses any sources of un- Solution designed to facilitate compliance with the risk-based
certainty, and ultimately seeks to protect the patient Qualification, Validation, & Change Control GMP require-
(1, 7). Use of a discrete confidence factor within the ments of the EU, Unpublished doctoral dissertation, Dublin
risk tool has several benefits: investigation teams Institute of Technology, Dublin, Ireland, pp. 15, 212-216,
are encouraged to provide exhaustive supporting December 2007.
evidence to improve the confidence ranking, the 4. Code of Federal Regulations, Title 21, Food and Drugs
design of the risk tool improves the robustness of (Government Printing Office, Washington, DC), Part 211,
investigations and the validity of conclusions drawn, subpart F Current Good Manufacturing Practices for Fin-
underlying assumptions are explicitly recognized ished Pharmaceuticals - Production and process controls.
and confirmed or refuted, and patients are protected Section 211.100 Written procedures; deviations.
by assuring the assignment of appropriately conser- 5. EU Guide to Good Manufacturing Practices, Volume 4, Part
vative risk estimations where such supporting data II, Basic requirements for active substances used as starting
is limited. materials. Section 2.16, p. 8.
The implementation of the risk-based impact 6. ISO/IEC Guide 51, Safety aspects Guidelines for their
assessment methodology fosters consistency and inclusion in standards (1999).
improved investigations through a standardized 7. ISO 31000, Risk management principles and guidelines
decision-making process ensuring that the principles (2009).
of quality risk management are integrated into the 8. Code of Federal Regulations, Title 21, Food and Drugs (Gov-
overarching quality system. Repeated use of RBIA ernment Printing Office, Washington, DC), Part 7, subpart
and similar custom-designed tools can elevate risk A Enforcement Policies General provisions. Section 7.3
from a resource-intensive rarity to the new normal Definitions.
the way we do business in a GxP environment. 9. EMEA, Standard Operating Procedure - Public, Dealing
with reports of suspected defective medicinal products.
REFERENCES SOP/INS/2018. Oct 5, 2012. p. 15.
1. ICH, Q9 Quality Risk Management. 10. ICH, Q8 Pharmaceutical Development.
2. ICH, Q10 Pharmaceutical Quality System. 11. ICH, Q11 Development and Manufacture of Drug Substances
3. K. ODonnell The development of a Quality Risk Management (Chemical Entities and Biotechnological/Biological Entities).

42 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

Laboratory Self-Audits
Paul L. Pluta and Jerry Lanese

Audit 101 provides useful and practical information that addresses various
topics associated with audits. Common-sense suggestions with application to
all audits internal and external, regulatory and non-regu-latory, domestic
and international are provided. examples of actual occurrences are de-
scribed whenever possible. Audit 101 has previously discussed the following
First impressions -- the Plant tour (1)
Preparation, Preparation, and More Preparation (2).
reader comments, questions, suggestions for discussion topics, and actual
audit experiences are needed to help us fulfill the column objective. Please
send your comments and suggestions to column coordinator Paul Pluta at
paul.pluta@comcast.net.

iNtrOduCtiON
every organization must become comfortable with being audited.
regulatory audits by global agen-cies, international Organization for
Standardization (iSO) registrar certification audits, external custom-er
audits, and internal quality assurance (QA) audits happen frequently.
Some may come at a moments notice while others are planned weeks
in advance. the key to a success is to make sure your area is always
prepared for an audit. A functioning labora-tory must function in a state

every laboratory must become comfortable with being audited.


A self-audit conducted internally is an excel-lent tool for the lab
to prepare itself for any audit, iden-tify problems, test execution,
and simulate an actual audit experience. Conducting a rigorous
self-audit that simulates an actual audit also helps to establish
a culture that supports an on-going state of prepared-ness in
the laboratory. Successful laboratories oper-ate in this state at
all times. this discussion provides a guide to the self-audit of
a laboratory. Laboratory systems independent of an actual test
result are reviewed beginning with the laboratory organizational
structure through sample collection, analytical testing, and re-
cord retention. Areas of interest are identified including specific
function, procedures, practices, records, and staff compliance
with procedures. topics discussed include the laboratory audit
process, rel-evant audit documents and references, laboratory
procedure availability, document requests by auditors, and
audit training for personnel. An approach to gap identification,
analysis, and prioritization of mitigation projects is presented.
A case study is given describ-ing an actual regulatory audit of
an analytical quality control (QC) laboratory and its supporting
research and development (r&d) laboratory.

Special Edition: a roadmap to gmp compliance Part 3 43


Paul L. Pluta and Jerry Lanese

of readiness on a daily basis. However, audits and in- when self-audits of laboratory operations are com-
spections of the labo-ratory are often not anticipated. pleted and gaps are identified, risks are evalu-ated.
Audits that start in a distant sites conference room the remedial actions required to mitigate these risks
usually lead to the manufacturing floor and ultimate- are then prioritized and addressed.
ly involve one or more laboratories at the site. A case study demonstrating an alternate ap-proach
Organizations often have a designated internal to the laboratory audit is also presented. this case
function whose sole purpose is to conduct inter-nal study follows an actual test result back-wards
audits. these may be very rigorous audits; the person- through the lab, starting with the result. either of
nel conducting such audits are usually very well ex- these approaches, (incoming sample re-ceipt through
perienced. these audits are stressful events for the lab final results or final result through incoming sample),
since the results may be communi-cated to high-level may be utilized in a self-audit. A combination of
personnel in the organization. to prepare for these methods, or use of both approaches at different times,
internal audits as well as the aforementioned regula- may serve to best uncover area weaknesses.
tory, iSO, and other audits, self-audits conducted by
the laboratory personnel are recommended. this is an Approach to the Self-Audit
excellent tool for the lab to prepare itself for any au- Areas of interest are identified within each of the ele-
dit, identify prob-lems, test execution, and simulate ments of the laboratory operation. the auditor should
the actual audit experience. investigate the following in each area and re-spective
Conducting a rigorous self-audit or that simu- activities:
lates an actual audit also helps to establish a culture Function of the specific element or activity
that supports an ongoing state of preparedness in Appropriate procedures are in place
the laboratory. the following discussion provides a Procedures accurately reflect the practice
guide to the self-audit of a laboratory. Audits may be records are complete, accurate, and maintained
conducted in different ways. this approach ad-dresses Staff is compliant with procedures
the laboratory systems independent of an actual test
result. it begins by identifying the major elements of LABOrAtOrY OVerView
a laboratory operation listed according to a typical An overview of the laboratory operation is the first
sequence of activities. it then identi-fies areas to as- area for review in the self-audit. Areas for review in
sess to determine the readiness of the laboratory for this element address the general goals and objectives,
an outside audit. the elements of a laboratory opera- operations, function, and management of the labora-
tion are listed below begin-ning with the laboratory tory. this type of information is requested when the
organizational structure through the sample collec- auditor first arrives at the laboratory. information
tion, testing, and record retention: re-garding the type of testing conducted, qualifica-
Laboratory overview tions of personnel and management, organizational
Sample collection chart, responsibilities, and other descriptive informa-
Sample receipt in the lab tion must be readily available. documentation and
Sample handling and storage in advance before re-cords must be provided when requested. the areas
testing to assess here are:
Sample testing type of testing conducted in the laboratory
instrument qualification Areas and units that submit samples to the labo-
instrument maintenance and calibration ratory
test method and sampling procedure develop- Organization of the laboratory, including respon-
ment sibilities and accountabilities
test method validation Qualifications of laboratory personnel
Laboratory technical training Qualification of laboratory management
data treatment education, training and experience of labora-tory
results verification, review and approval management
results reporting Processes which monitor laboratory performance
investigations Laboratory work outsourced to other areas
record compilation. within the company or outsourced to an out-side
record storage, retention and retrieval laboratory
topics including the laboratory audit process, Prior audit or inspection observations
rel-evant audit documents and references, laboratory
procedures, document requests by auditors, and au- SAMPLe COLLeCtiON
dit training for personnel are also discussed. the areas for review in this element address the

44 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

source of samples for testing. what areas submit SAMPLe HANdLiNG ANd StOrAGe iN AdVANCe OF
samples, how are samples collected, required spe-cial teStiNG
training for sampling personnel, etc. Samples may be the areas for review in this element address the han-
submitted from various areas in the orga-nization. dling and storage of samples before they are actually
Samples may also be submitted at vari-ous stages of tested by the laboratory. these areas are:
manufacturing. Some samples may require immedi- Sample storage while samples are awaiting test-
ate testing, while others may able to be staged within ing
the specific response time for laboratory efficiency. Provisions for special storage conditions such as
each of these considerations may lead to a specific refrigeration, light protection, or other require-
line of questions including specific requirements, ments
specific handling, and so on. the sampling activity is Qualification of sample storage area
often not within the con-trol of the laboratory. if the identification and prioritization of samples that
laboratory cannot assure the quality and integrity of require immediate testing
the samples received for testing, testing results will Security of storage area
be suspect because the sampling process lacks appro- Specific goals for completion of testing
priate controls. the specific areas to assess are: Procedures for sample handling and storage in
Areas or units that submit samples to the lab for the laboratory
testing
Qualifications of individuals who actually SAMPLe teStiNG
remove the samples from the manufacturing pro- the areas for review and assessment in this element
cess, stability area, or other source of samples address the actual testing of samples:
Specialized personnel training required for sam- reference standard program and controls
pling reagent controls
Specialized equipment or facilities required for Control of chromatographic columns
sampling Preparation control of stock reagents
the qualifications of individuals who deliver System suitability requirements for test methods
samples to the laboratory Procedures for handling deviations to analytical
the technical input by the laboratory into proper methods or other laboratory procedures
sampling, packaging, and labeling of the samples Procedures for handling changes to test meth-
Sample labels ods or other laboratory procedures
Procedures for sampling, packaging, and label- test procedures
ing, and submission of samples to the laboratory test-specific work sheets
Qualification of analysts to perform tests
the performance of required tests
SAMPLe reCeiPt iN tHe LAB
the areas for review in this element address specific iNStruMeNt QuALiFiCAtiON
activities associated with the receipt of samples by the areas for review in this element address the ca-
the laboratory from the sample delivery person. Are pability and readiness of laboratory instrumentation
all samples simply dropped into a receipt box or are for testing. All instruments and equipment must be
there specific receipt procedures under laboratory qualified according to their united States Pharma-
control? important areas to evaluate are: copeia (uSP) (or equivalent) instrument categoriza-
Sampling receiving areas security and integrity tion (3). Laboratory management must be mindful
Sample receiving process of the intended use of the instruments as well as
Sample receiving records the instrument manufacturers recommendations
Storage of samples immediately upon receipt and re-garding the equipment use. Changes to laboratory
environmental controls instruments should be evaluated through a change
Security of receipt area control system:
Qualification of the sample receiving area Procedures related to instrument qualification
examination and evaluation of samples upon responsibility for instrument and equipment
receipt, including records of the activity qualification
Process for logging samples into a laboratory Status of laboratory instrument and equipment
information management system (LiMS) com- qualification
puter system for tracking Change control as it applies to instruments and
Validation of the LiMS system. equipments

Special Edition: a roadmap to gmp compliance Part 3 45


Paul L. Pluta and Jerry Lanese

iNStruMeNt MAiNteNANCe ANd CALiBrAtiON LABOrAtOrY teCHNiCAL trAiNiNG


the areas for review in this element address main- the training of laboratory personnel and associated
tenance and calibration activities associated with training records are always requested in an audit.
instruments and other equipment. instrument cali- Areas that should be reviewed include:
bration is significant routine maintenance activ-ity for The laboratory technical training program
analytical instruments. instruments must be calibrat- Process for determination of appropriate train-ing
ed or user standardized for actual use. the following for each analyst
must be assessed: Evidence of analyst qualifications
instrument maintenance procedures Retraining
Laboratory instrument calibration program Maintenance of training records and responsi-
instrument calibration procedures bility, and current records
Calibration records
instrument logbooks dAtA treAtMeNt
the areas for review in this element address raw
teSt MetHOd ANd SAMPLiNG PrOCedure deVeL- data recording, calculations, and other treatment of
OPMeNt data to determine test results. the laboratory analysts
the areas for review in this element address the de- who perform the analytical testing record all data,
velopment of test methods by Analytical research and calculations, and compile instrument documenta-
design (r&d) or other technical support functions: tion and associated information. Calculations may be
Responsibilities for the development of test meth- done with calculators, spreadsheets, or other com-
ods, including sampling used in the laboratory puter software. if computer systems and soft-ware are
Evidence that specific samples for testing are rep- involved, these must be validated. the spe-cific areas
resentative of the material received of data treatment to be reviewed are:
Availability of method development reports in the Procedures for recording data in worksheets or
lab when needed for problem solving notebooks, labeling of instrument printouts,
Transfer of methods from other labs to this labo- entry into computer systems, calculation, and
ratory result reporting
Procedures defining test method and sampling Procedures covering averaging and significant
procedure development figures
Procedures for good recordkeeping practices
(good documentation practices)
teSt MetHOd VALidAtiON Software and spreadsheets used to perform cal-
the areas for review in this element address test culations, spreadsheet development standards,
method validation and associated activities. test evidence of validation
method validation is a key area in the laboratory Change control
audit. industry and regulators focus on analytical
method validation. Any type of test method (physi- reSuLtS VeriFiCAtiON ANd APPrOVAL
cal, biological, microbiological, chemical) must have the areas for review in this element address the
documented evidence that demonstrates the test final review and approval of raw data, calculations,
performs as intended. test methods must be ap-pro- data treatment, and other associated activities by
priately validated if they are used for any test-ing re- super-vision prior to release of results to the appro-
quired by the good manufacturing practices (GMPs). priate organizations. Laboratory records are signed
Changes to validated test methods must be evaluated and dated by the analyst to affirm performance of the
through a change control system. the fol-lowing as- testing. they are then reviewed and approved by lab-
pects of the test method validation must be evaluated: oratory supervision. All records are signed and dated
Responsibilities for test method validation by laboratory supervision affirming performance of
The test method validation process the testing, approval of the test results, and com-
Status of the validation of all test methods pliance with internal procedures and regulations.
Evidence that test methods perform as intended througout this process, there should be heightened
The system for changing test methods emphasis on the assemente of:
The involvement of development subject matter Responsibilities of analysts and reviewers
experts in evaluation of changes Procedures covering verification, approval and
Location and availability of test method valida- release of raw data, calculations, final reports to
tion documentation appropriate areas, and signature/date require-
ments for analysts and supervisors

46 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

Responsibilities for approval of all data and cal- test documentation may be stored electronically or
culations prior to reporting results in hardcopy depending on the organization. Stor-
age must be secure. Access to test records must be
reSuLtS rePOrtiNG restricted to qualified personnel. if storage is by
the areas for review in this element address the electronic systems, these systems must be vali-dated.
report of approved results to the sample originator data and test information must be quickly and easily
or the unit using the data. Sites may have electron-ic retrievable. reviewing the following is a necessity:
systems into which test results are entered and are Procedures for record storage, retention and
ultimately transferred for final lot disposition. Other retrieval
sites may have paper systems that accom-plish the Security and environment of the record storage
same objective. Sites may also have mixed systems, areas
(e.g., electronic systems) for certain sam-ples and Qualifications of individuals responsible for
paper systems for special samples. Spe-cifically, in record storage
results reporting, the following should be reviewed:
The process for reporting results to quality or tHe LABOrAtOrY Audit PrOCeSS
sample originators actual laboratory audits may be conducted in vari-
Validation of any computerized system used in ous ways. the above elements outline a start to fin-
the sample reporting process ish approach to the audit structured according to the
Timeliness of results measured against labora- business process of the lab. it establishs the labo-ra-
tory turnaround goals tory organization infrastructure through the test-ing
Procedures for reporting of results to the storage and maintenance of the records. Audits
Assurance of appropriate testing and complete may also be conducted in the reverse order, (i.e.,
testing following a test result from finish to start.). in this
approach, the audit follows a reported test result from
iNVeStiGAtiONS the laboratory report to data generation. the audit
the areas for review in this element address perfor- then follows to testing through sample receipt and
mance of investigations when aberrant test results original sampling by the sample originator. the at-
are generated. test data may fail specifications or may tached case study is an example of this latter ap-pro-
vary significantly from historical norms. this data ach and represents a realistic audit experience. use of
requires investigation by the laboratory to de-termine both approaches at different times will chal-lenge the
the causes for the aberrant laboratory re-sults. there laboratory system and expose personnel to a variety
should be focus on the reviewing the following: of audit circumstances.
Procedures for handling and investigating out-of- Other documents that may be used as the basis
specification or out-of-trend laboratory results for internal audits include the uS Food and drug
Procedures for laboratory investigations Association Compliance Program Guidance Manual
Evidence of typical laboratory investigations #7356.002 (4), FdA Compliance Program Guidance
Retention of laboratory investigation reports Manual #7345.848 (5), and PiC/S Aide Memoire: in-
Corrective and preventive actions resulting from spection of Pharmaceutical Quality Control Labora-
laboratory investigations tories (6) and periodic review of FdA warning Let-ters
(7) are also helpful. FdA observations provide insight
reCOrd COMPiLAtiON into current regulatory emphases and ideas for ad-
After testing is completed, data verified, results calcu- ditional areas or questions in a self-audit.
lated, reviewed, and approved; and results re-ported
to quality or the originator, all records are compiled FdA Compliance Program Guidance Manual
in a standardized format. Areas for review in #7356.002
this element include:The process for reporting this document was written to provide Center for
results to quality or sample originators drug evaluation and research (Cder) investigators
Procedures covering the format for completed an overview of areas to be reviewed during a drug
laboratory records manufacturing inspection. it identifies the six basic
Procedures for compilation of records systems within a quality system including the labo-
evidence of completed records ratory control system. this guidance identifies areas
within each system which the investigators should re-
reCOrd StOrAGe ANd retrieVAL view. it also directs that the firm should have writ-ten
the areas for review in this element address the and approved procedures for each of the areas and
storage and retrieval of laboratory data. data and records of these procedure results. the investi-gators

Special Edition: a roadmap to gmp compliance Part 3 47


Paul L. Pluta and Jerry Lanese

are also directed to verify that personnel are follow- Pattern of failure to follow an adequate OOS pro-
ing the procedures. the areas listed for the laboratory cedure
control system are: Pattern of failure to retain raw data
Training/qualification of personnel Lack of stability-indicating methods
Adequacy of staffing for laboratory operations Pattern of failure to follow stability programs
Adequacy of equipment and facility for intend-ed
use FdA Compliance Program Guidance Manual
Calibration and maintenance programs for ana- #7345.848
lytical instruments and equipment this document provides Center of Biologics evalu-
Validation and security of computerized or auto- ation and research (CBer) investigators an overview
mated processes of areas to be reviewed during a biolog-ical manu-
Reference standards source, purity and assay, and facturing inspection. it identifies the same six basic
tests to establish equivalency to current official systems within a quality system and critical elements.
reference standards as appropriate Standard operating proce-dures (SOPs) and train-
System suitability checks on chromatographic ing records are highlight-ed. it also identifies areas
systems such as gas chromotography or high per- within the laboratory control system that should
formance liquid chromotography be reviewed, as well as observations that could be
Specifications, standards, and representative reported. these are similar to the areas identified in
sampling plans 7356.002 and include:
Adherence to written methods of analysis Written procedures and control system for labo-
Validation/verification of analytical methods ratory operations
Control system for implementing changes in Calibration and maintenance programs for ana-
laboratory operations lytical instruments and equipment
Required testing performed on the correct sam- Adherence, validation/verification to the writ-ten
ples methods of analysis
Documented investigation into any unexpected Testing and release for distribution
discrepancy Specifications, standards, and representative
Complete analytical records from all tests and sampling plans
summaries of results Stability testing program, including demonstra-
Quality and retention of raw data such as chro- tion of stability indicating capability of the test
matograms and spectra methods
Correlation of result summaries to raw data; Special testing requirements
presence of unused data Adequate reserve samples and documentation of
Adherence to an adequate out of specification reserve sample examination
(OOS) procedure which includes timely com- Required testing is performed on the correct
pletion of the investigation samples
Adequate reserve samples; documentation of Laboratory records
reserve sample examination
Stability testing program, including demon- PiC/S Aide-Memoire
stration of stability indicating capability of test this document provides guidance for GMP inspec-
methods tors for training and preparation of inspections. it
the guidance also identifies example laboratory contains nine tables addressing general subjects and
and control system problems. these include the fol- items to be investigated during GMP inspections. the
lowing: general subjects addressed in this document in-clude
Pattern of failure to establish/follow a control sys- the following:
tem for implementing changes in the labora-tory General information
operations Quality assurance system
Pattern of failure to document investigation of documentation system
discrepancies Personnel
Lack of validation of computerized and/or auto- Premises and equipment
mated processes Materials and supplies
Pattern of inadequate sampling practices Sampling and samples
Lack of validated analytical methods testing
Pattern of failure to follow approved analytical results and release of test results
procedures An extensive list of specific questions for each of

48 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

the above sections is listed in PiC/S Aide-Memoire. nal audi-tors. Practice will help to minimize their
the above provides a different approach to a labora- anxiety and fears.
tory audit with different focus.
Supplements addressing GMP inspection of riSK eVALuAtiON OF ideNtiFied GAPS
chemical, physical, and microbiological testing Gaps in procedures, processes, records, and perfor-
labo-ratories are also provided, also including an mance will be identified after conducting a rigorous
exten-sive list of very specific questions. self-audit. these gaps must then be evaluated and
the quality assurance system section of this corrective actions initiated. Highest risk gaps must
docu-ment asks the following questions regarding be prioritized for rapid response. tables i, ii, and iii
self inspection: illustrate the results of a representative self-audit
How is it, and by whom performed? us-ing the activity format discussed. table i provides
How is it reported? a representative list of audit observations from a
How are corrective measures implemented? self-audit using the format of the above discussion.
is the schedule available and adhered to? table ii demonstrates a risk evaluation of observa-
tions. table ii provides a prioritized list of project
OtHer CONSiderAtiONS assign-ments addressing gaps with target dates and
documents requested by Auditors person-nel responsibilities.
whenever documentation is requested by an audi-
tor, it must be quickly retrieved within a reasonable CASe StudY: LABOrAtOrY Audit
time (for example, 30 minutes). if r&d reports or A regulatory auditor team was conducting a new
other documents or records generated by another tab-let product pre-approval inspection at the
unit are not readily available through an electronic product manufacturing site. the audit was being
report system, hardcopies or relevant reports should conducted in the designated conference room in the
be obtained from the originating unit and stored manu-facturing plant. the regulatory agency auditor,
within the laboratory area to facilitate timely retriev- site compliance officer, and QA manager were pres-
al. excessive time delays in retrieval of documents ent in the conference room.
are not acceptable. the process validation report for the new product
was requested by the auditors. the process valida-
Laboratory Procedures tion report was retrieved from the validation library.
Auditors always request the approved laboratory the auditors reviewed the report. the auditor then
procedures and test methods. these are usually selected the content uniformity test results in the
available through an electronic system. in the nor- validation report. the report provided the sampling,
mal operation of the laboratory, analysts may print a test procedure, test lab location, acceptance criteria,
copy for their immediate personal use. this intro- table of results, description of data treatment, and
duces the potential for using an out-dated procedure a statement that the data met acceptance criteria
when revisions have occurred. Laboratory person- re-quirements.
nel must be trained to always access the current the auditors then requested to visit the labora-to-
procedure when a procedure is needed. there should ry where test data were generated. the testing labo-
be a system that prevents the holding and extended ratory was approximately 30 minutes away from the
use of printed copies. if printed copies are used in manufacturing plant. the laboratory su-pervisor was
performing testing, they should be destroyed after immediately contacted and informed that the audi-
completion of the activity. tors would be visiting his lab in ap-proximately 30
minutes to review content uni-formity testing from
Audit training for Personnel the process validation. the regulatory audit team,
Aside from the technical training for personnel on compliance officer, and QA manager then departed
laboratory and testing procedures for the actual per- for the lab with the process validation report.
formance of sample testing, laboratory personnel After arriving at the laboratory and brief intro-
should also be trained on proper interactions with ductions of personnel, the lead auditor selected a
auditors. this includes topics such as audit protocol, single test result reported in the process validation
listening carefully to questions, asking for clarifica- report. the specified result was determined by an
tion if requests are unclear, being calm, and phrases HPLC test method. He asked to see the following
to avoid. internal audits should include analyst laboratory documentation associated with the test
in-teraction to provide practice for handling audits result:
and responding to auditor questions. Laboratory The analytical test method procedure associ-
analysts will be nervous when dealing with exter- ated with the sample

Special Edition: a roadmap to gmp compliance Part 3 49


Paul L. Pluta and Jerry Lanese

Table 1: Laboratory self-audit


The analytical method validation report Standard storage, handling, and source of supply
The analytical method development repor Reagents used in the test procedures
After the above review, the auditors then focused on Reagent preparation procedures including expi-
the specific test result. they requested the following: ration dating
Hplc chromatogram associated with the sample Instrument qualification report
Sample preparatory equipment associated with the Calibration procedure
test method Calibration history
Actual equipment on which sample was tested Name of laboratory analyst
Standard used in the test procedure Resume of laboratory analyst

50 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

Table 2: Risk evaluation of identified gaps

Training records of laboratory analyst were then requested from the r&d lab manager:
Name of supervisor who approved data and calcu- General overview of r&d laboratory function
lations Organizational chart for laboratory
Resume of supervisor Analytical r&d report for the content uniform test
Training records of supervisor method
Procedure for sample receipt and storage Raw data for testing reported in the analytical r&d
Sample receipt area report (this data was recorded in a labo-ratory
Sample receipt record notebook. Consistency between the raw data and
Sample storage area the analytical r&d report for repre-sentative data
Procedure for data treatment was verified.)
Procedure for data and report storage Procedure describing r&d documentation prac-
Data and report storage facility tices
Procedure describing report security area Data security and storage policy
All laboratory documentation requested was re- Equipment and instrumentation used in the devel-
trieved and reviewed. results were consistent with the opment process, including instrument mainte-
results reported in the process validation report. nance and calibration logs
After looking at the analytical method develop-ment Standards and reagents used in the develop-ment
report which was written by an analytical r&d scien- process, including expiration dating of materials.
tist, the auditors requested to visit the r&d lab where Resume and qualifications for r&d scientist who
analytical method development work was conducted. developed the test method
the r&d area was contacted. the r&d manager was Resume and qualifications for r&d manager
informed that the auditors would be visiting the labora- All auditor questions were successfully answered
tory in connection with the content uniformity method and requested documents provided. No inconsisten-
development for the new product. After approximately cies between reports and original data was found.
a fifteen minute walk between site buildings, the audit this case describes a typical progression of a regula-
team arrived at the analytical r&d lab. the following tory audit. the audit started with a reported test result.

Special Edition: a roadmap to gmp compliance Part 3 51


Paul L. Pluta and Jerry Lanese

Table III: Risk mitigation goals, responsibilites, and target dates


this result was then tracked to laboratory raw data. checked, reagent dating was checked, personnel train-
Activities associated with the test result were then ing was re-viewed, etc.
reviewed. these included laboratory test procedures, All questions and requests from by the auditor were
supporting equipment and instrument information, successfully answered. All documents were quickly
miscellaneous laboratory systems, and review of train- retrieved. the audit of the quality control (QC) and
ing of personnel associated with the original test result. r&d laboratories required one full day of review. No
the audit then continued to the r&d area the developed deficiencies were cited by the auditor. the auditors then
the test method of inter-est. r&d reports were reviewed, returned to the manufacturing area to continue the
equipment was reviewed, calibration records were audit.

52 Special Edition: A Roadmap to GMP Compliance Part 3


Paul L. Pluta and Jerry Lanese

FiNAL tHOuGHtS Pluta. Audit 101. Preparation, Preparation, and More Prepa-
Successful laboratories operate in a constant state of ration. Journal of GXP Compliance, Volume 15, #4, Autumn
compliance to internal procedures and regula-tions 2011.
and they are continually prepared for an au-dit. there 3. uSP <1058> Analytical instrument Qualification. www.usp.
is no getting ready for an audit. Suc-cessful labora- org.
tories are always ready. in reality, regulatory auditors 4. FdA. Compliance Program Guidance Manual: Program,
may arrive unannounced at a firm at any time and #7456.002. implementation date 2-1-2002.
proceed to initiate an audit. Successful laboratories 5. FdA Compliance Program Guidance Manual: Program
understand this reality and operate appropriately. 7345.848. implementation date 10-1-2010
Conducting self-audits on a regular basis is a good 6. PiC/S. Aide-Memoire. inspection of Pharmaceutical Quality
way to assess laboratory performance, im-prove Control Laboratories. September 25, 2007.
readiness for an audit, and expose personnel to an 7. FdA. www.FdA.gov/iCeCi/enforcementActions/warnin-glet-
audit experience. using different approaches and ters/default.htm
styles as described above and in the case study is
desirable. Knowledge of regulatory requirements, ABOut tHe AutHOrS
recommendations, and expectations as described in Paul L. Pluta, Phd, is Column Coordinator and
official documentation is mandatory. Periodic review editor-in-Chief, Journal of GXP Compliance and
of regulatory FdA warning Letters is also helpful. Journal of Validation technol-ogy. Contact dr. Pluta
Audits are a way of life. A good laboratory and its with comments or questions on this discussion at
personnel are always prepared. paul.pluta@comcast.net.
Jerry Lanese, Phd, is an independent consultant
REFERENCES specializing in quality systems and laboratory con-
1. Pluta, Paul L., editor. Audit 101. First impressions -- the trols. He is a member of the editorial Board of Journal
Plant tour. Journal of GXP Compliance, Volume 15 # 1, of GXP Compliance. Contact dr. Lanese at jerry@
winter, 2011. lanesegroup.com
2. Graham, Ballard, richard Poska, robert ertmann, and Paul L.

Special Edition: a roadmap to gmp compliance Part 3 53


David Markovitz

Good Manufacturing Practice


and Stable Systems - Part One:
Reducing Stress with the Power
of Data - Know When to Act and
When Not to Act
David Markovitz

Good Manufacturing Practice (GMP) is a quality system designed to help


manufacturers produce quality products that are safe, pure, and effective. In
theory, this is easy to do. In practice, however, there are several challenges.
Manufacturing operations are comprised of multiple systems and processes.
Effectively managing these systems and processes is the key to success
One of the biggest challenges is knowing when to act and when not to act in
managing our systems and processes. The late W. Edwards Deming (1900-
1993) cited two mistakes people make when managing systems and processes.

Mistake Number One


To react to an outcome as if it came from a special cause of variation,
when actually it came from common causes of variation.

Mistake Number Two


To treat an outcome as if it came from common causes of variation,
when actually it came from a special cause.
So what did Deming mean by these two mistakes. Lets first examine
Mistake Number One.

Understanding Mistake Number One


When all the variation within a system is due to common causes of
variation, we call that a stable system. We may be getting the desired
results or we may not be getting the desired results. The results are
predictable within a certain range. The system is what it is. It does what
it does. All the variation is built into the system.
When we react as if something special is happening, we almost al-
ways make things worse. Deming called this tampering. We take action
when we shouldnt take action.
Heres an example. Consider your commute to work. This is a com-
mute most of us make five days a week, week after week. Lets also
consider that your commute includes about ten miles of surface street
driving and about ten miles of driving on a freeway or turnpike.
Your commute time likely varies from day to day, but not by much,
perhaps five to ten minutes or so. What accounts for the variation? Sev-
eral factors contribute to the variation.

54 Special Edition: A Roadmap to GMP Compliance Part 3


David Markovitz

Among them are: How to avoid Mistake Number One


The time you leave home. (Lets assume that you When dealing with a stable system the only way to
leave home about the same time every day) improve the results is to work to improve the system.
The route you take (Likely to be the same route Identify the inputs and process variables and experi-
every day) ment until you find what works better.
The signal lights along the route, and whether you Deming called this the Plan-Do-Study-Act cycle.
catch them red or green. (Over time, you will get Plan a change. Try it out on a small and manageable
about 50% red lights and about 50% green lights) scale, study the results, and, if favorable, put them
The volume of traffic. (Likely to be about the same into action. If the initial results are not favorable, run
every day) through the Plan-Do-Study-Act cycle again and again
Your personal driving habits. (Whether you until you learn what works best.
change lanes frequently or rush from one red light Rewarding people for things over which they have
to the next) no control and punishing people for things over which
The weather (Most of the time the weather on any they have no control is tampering. It will make things
given day is similar to the previous da worse. This leads to confusion among people and mo-
Based upon these factors, we make a prediction rale usually suffers as a result of these actions. Think
each day on how long it takes to get to work, and of how many Employee of the Month programs are just
hence what time to leave home. rewarding people who were lucky that month. They
What if on Monday you arrive at work at 7:55 am? just got more green lights that month. And the next
And on Tuesday you arrive at 8:05 am? Wednesday at month they are back in the pack, no longer singled
7:59 AM, Thursday at 7:58 am, and on Friday at 8:02 out for good performance, or worse, punished for poor
am? performance.
Do you believe that you deserved a Good Driver People work in the system that was designed by
Award for Mondays performance? After all, it was management. Managements job is to improve the
your best performance for the week. And do you system over time. Continually working to have stable
believe that your award should be taken back when on systems, and optimize those systems so they are
Tuesday you turned in your worst performance? constantly getting better in relationship to all the other
Of course not. The variation is built into the sys- systems that they interact with. You may one day reach
tem. Half the time you will arrive before 8:00 am and the point where additional improvement is not worth
half the time you will arrive after 8:00 am. cost of implementation. Then your responsibility is
Do you think it may be wise to buy a faster car? to continue to monitor the system to maintain the
Of course not. That would be a waste of money and improved level of performance.
acting when we shouldnt. How can you identify whether a system is stable?
How about getting a different driver? Hire someone Easy - Walter Shewhart figured it out in the early
else to drive you to work, perhaps a former race car 1900s. He invented Statistical Process Control (SPC)
driver. Again, a waste of money and acting when we charts. Effectively applying SPC charts helps you
shouldnt. identifying the difference between common causes of
How often have people tried to fix a system by buy- variation and special causes of variation.
ing new equipment (a faster car)? These charts can help you determine when to act
How often have people tried to fix a system by and when not to act. Research and experience has
changing the operator/worker (a different driver)? found that the key to applying SPC charts effectively
Sports teams do this all the time - they fire the lies in knowing what to measure and how frequently
coach or manager and hire someone else expecting to measure it. Preparing the charts and interpreting
different results. It works about half the time - about the charts is the easy part. Knowing what to measure
what you would expect if you have a basic under- is the challenge.
standing of statistics and probability.
The system of commuting to work is what it is - it Understanding Mistake Number Two
does what it does. Lets now turn our attention to Mistake Number Two.
If the results are not meeting your needs (specifica- Heres where something unusual occurs (a special
tions), then the only thing you can do is to work to cause of variation) and we miss it, or ignore it. Special
improve the system. causes, if left alone, can become part of the system over
Perhaps try a different route, or leave at a significant time. Most systems are guided by the laws of nature.
different time (with traffic in most places that would They dont usually get better by themselves, they tend
mean leaving earlier), or even move closer to work. to degrade (in physics we call this entropy), and tend
to get worse over time. When special causes of varia-

Special Edition: a roadmap to gmp compliance Part 3 55


David Markovitz

tion exist, our system is no longer stable. That is - we How you can use this information to make
can no longer predict the results to be within a certain better decisions
range. ONE - Start by characterizing your systems and pro-
Consider our commute to work example and use cesses. Each system and process has inputs, outputs,
the same factors identified above. and process variables. Identify the inputs and then
For several weeks your arrival time at work has put measures into place to manage and control these.
ranged from 7:55 am to 8:05 am. Its a stable system. Identify the system and process variables. Select what
But one day you arrive at 8:30 am. Here its fair to ask to measure and how frequently to measure these. Put
What happened? What was different in your com- measures into place to manage and control these.
mute today? By managing your inputs and system and process
Possible answers include: variables you can more accurately predict outcomes.
Car had a flat tire, and it took 30 minutes to Predictable outcomes and results lead to better and
change to the spare tire. more consistent product quality and leads to less
Big traffic accident on the freeway or turnpike and stress in the workplace - always a good thing.
you were stuck in traffic an extra 30 minutes get-
ting to the next exit. TWO - Learn how to apply SPC in your systems and
The day care provider called in sick and it took an processes. Its not hard; it just takes some knowledge
extra 30 minutes to find alternative care for your and diligence to apply it effectively. Properly applied,
children. SPC can help make the invisible visible.
There are numerous possibilities here.
These are all special causes of variation. But lets THREE - Stop asking who and start asking why. So
say that you start having flat tires at the rate of one often when things go wrong, management is quick to
per week. The action to take here is to buy new tires. ask Who is responsible for this? Its easy to blame
Or big traffic accidents are occurring frequently. The people, and harder to examine our systems. It is far
action is to take alternate routes. better to ask the people closest to the system or pro-
cess, Why do you think this happened?
How to avoid Mistake Number Two Remember people work in the system. Manage-
Sometimes our systems get worse over time because ment works on the system. And those working in the
we are missing the signals that SPC charts can pro- system may have more knowledge of how the system
vide. When special causes occur, specific signals can works than those outside the system can see. Their
be found in interpreting SPC charts. These signals knowledge, combined with the knowledge gained
dont tell you what happened, they just tell you when from the effective use of SPC charts, can help you
to look. You have to be smart enough to know what to make better decisions and avoid making the two
look for and where to look. And then smart enough to mistakes.
know what to do to eliminate the special causes from
the system. Or if the special cause is providing better REFERENCES
results, your job is then to find a way to incorporate 1. The New Economics for Industry, Government, Education;
this into your system so it becomes part of a stable Deming, W. Edwards; MIT Center for Advanced Engineering
system with predictable results. Study; 1994.

56 Special Edition: A Roadmap to GMP Compliance Part 3


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

Implementing an Effective
GLP Program at the Contract
Laboratory | IVT
Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

ABSTRACT
This discussion provides an overview of a GLP program at a contract
laboratory. The objective of the GLP program is generation of analyti-
cal data in support of a GLP study. Understanding what is required
to complete a single study leads to understanding what quality sys-
tems must be in place in order to complete multiple and simultane-
ous studies. Desired qualities of a GLP study include transparency,
traceability, completeness, accuracy, precision, clarity, integrity, and
retrieveability. Each will render the study easier to audit and minimize
non-compliance. Administrative tools recommended for a GLP program
include pre-printed workbooks and logbooks, standardized formats for
result reporting, study-specifics, and peripheral records; study num-
bers, study files, and client codes; checklists, and other tools. A typical
study workflow is presented. This basic workflow may be scaled for
application to multiple simultaneous studies. Other useful information
provided includes an experimental workbook template, instrument
logbook template, information captured on controlled forms, GLP study
requirements, some essential SOPs, and key quality subsystems to GLP.
Elements of a quality systems approach to auditing GLP studies are
discussed. Having and following good SOPs is the key to implementing
the program described. Training personnel on SOPs and revisions, and
dealing with deviations and change is also vital. The concepts dis-
cussed herein are critical to developing a compliant GLP program that
will improve, become self-correcting, be stable and efficient.

INTRODUCTION
Implementing a GLP program in a contract lab such as ours at NSF
Health Sciences is a large undertaking. It does not really help if the
scale of the operation is relatively small: Small labs and large labs are
held to the same regulatory standards. Good quality systems, once
in place, should be scale-able. While it is true that smaller typi-
cally means fewer instruments and scientists to be qualified, it is also
true that small and large CROs and big pharmaceutical companies all
continually face risk vs. resources challenges. One must determine the
right way to do something and how to pay for it. For the purpose of
the present discussion, the something is generating analytical data to
support a GLP study, while the right way is that way or ways that are
in compliance with the GLPs and have a high probability of remaining
in compliance during growth and change (1). The goal of this discus-
sion is to provide useful, hands-on tools that have been tested in our

Special Edition: A Roadmap to GMP Compliance Part 3 57


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

laboratory over the past 15 years for implementing an study leads to understanding what quality systems
effective GLP program. must be in place in order to complete multiple and
simultaneous studies. Before we step through the
BACKGROUND flow of work, we point out some of the important
Many scientists approaching Good Laboratory qualities that a GLP study must possess. This will
Practice (GLP) for the first time are surprised to find help the reader understand why certain steps are
that the subject matter lives up to its name. It is all placed in the workflow. These include: transparency,
about having and following good practices in the traceability, completeness, accuracy, precision, clarity,
laboratory. For example, the importance of keeping integrity, and others such as being readily retrievable.
a scientific record that is good enough to permit the Each will render the study easier to audit. One test of
reconstruction of the experiment at a later date and a good study is that it must be easy to audit and the
the importance of scientific honesty are at the heart audit not identify any GLP non-compliances. Such
of good scientific research and GLP. Keeping good attributes can be built-in by thinking about how the
lab records on the calibration, cleaning, maintenance, GLP program will be administered. We believe good
and performance of lab instruments is part of this, as administration is a key to effective management.
is keeping a lab notebook or workbook that includes Some simple examples of administrative tools fol-
results from all attempts made in an experiment or low:
study both successful and unsuccessful. That this
record has the qualities of being original, attributable Workbooks and Logbooks
to the author, contemporaneous, accurate, and legible We use pre-printed laboratory workbooks. Work-
is emphasized as part of a subset of good documen- books are designed to permit the capture of infor-
tation practices. These and similar ideas have been mation deemed essential for later reconstruction
taught in the undergraduate university curriculum of the experiment. The flow of information follows
for a long time. The 1952 text by Harvard Professor chronologically the steps taken in the lab to obtain
E.B. Wilson, Jr. entitled Introduction to Scientific the data. The design is pre-approved by lab manage-
Research remains an excellent resource (2). Still, the ment and Quality Assurance and changes to design
largest part of the burden of educating scientists in are controlled. Workbooks are easy to use and to au-
this area falls on the companies that use their talents. dit. Information appears time and again in the same
This discussion focuses more on the how than on location in the workbook, regardless of the particular
the why of GLP. Readers will find an introduction analyst, analysis, or study. Workbooks are controlled
to some of the early motivation in referenced litera- documents and are printed by the Document Control
ture (3). Unit (DCU) only. Table I provides a list of typical in-
formation recorded in the workbook. The same logic
COMPLETING A SINGLE STUDY applies to the various laboratory logbooks (Table II).
Understanding what is required to complete a single

Table 1: Experimental workbook template


Analyst name Study number & date Document number
Test method number or protocol
Objective Test specifications/ranges
number
Test article information
Instrument set-up and parameters Standards preparation
(ID #, lot #, description, etc.)
Instrument list (ID #, date of Reference standard information Reagents/solutions
last calibration, next calibration (name, lot, expiration date, purity, (name, lot, expiration date, purity,
due date) manufacturer) manufacturer)
Solution preparation, diluents and
Sample preparation (weights,
mobile phases (lot #, manufacturer Calculations (with units and
volumes, dilution factors and
and expiration date of each reagent proper significant figures)
final concentrations)
used)
Reporting results (with units)

58 Special Edition: A Roadmap to GMP Compliance Part 3


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

Table II: Instrument logbook template

Instrument ID number Manufacturer Model & serial number


Instrument custodian Instrument location/ lab number Qualification/Calibration status
Table of Contents: running list
Date of IQ
in chronological order of all
Details of IQ
Instrument type/description instrument events (e.g. repairs,
Reference the SOP (with version
cleaning, preventative maintenance,
number) used in performing IQ
calibration, etc.)
Date of last calibration
Date when next calibration is Date of PM
Details of OQ
due Details of the PM
Reference the SOP (with version
Details of PQ Reference the SOP (with version
number) used in performing OQ
Reference the SOP (with version number) used in performing PM
number) used in performing PQ
KEY: IQ/OQ/PQ, PM stand
Details of instrument repairs and for Installation/Operational/
Log of Instrument failures
remedial actions Performance Qualification and
Preventative Maintenance

Reporting Results Study-Specific and Peripheral Records


Each data table in the GLP study final report in- A typical study generates a large number of records.
cludes reference to the lab workbook containing the An old joke is that GLP stands for generate lots of
original recording of the data. Each lab workbook paper. Some of the records are identified in Tables
also contains a summary table of the results. We I-IV. Study recreation depends upon records that are
call this section of the workbook Reported Results study-specific and those that are not. For example,
and it includes all results derived from that part of the analyst enters the instrument number into the
the experimental study that is represented by the workbook (a study-specific document) and this links
particular workbook. When the results are numeri- it to the instrument logbook (a document used for
cal (as is typical) they are given with final units and multiple studies). Likewise, the analytical test method
rounded to the proper number of significant figures. document number is entered into the workbook and
The auditor then can easily work backwards and this links to the approved written procedure. The
forwards between the GLP report and the raw data. integrity of the study depends in part on the strength
In addition, since we use a different lab workbook of the links between the study-specific record and the
for each attempt at completing the analysis, coupling record of events occurring simultaneously in the lab
the lab workbooks to the GLP study final report in that impact upon the quality of the study. In Table III,
this way adds integrity and helps to ensure transpar- we identify some of these peripheral records.
ency, traceability, and completeness in our reporting
practices.

Table III: Some records peripheral to the study


Lab Operations Training Metrology
Test article (sample) log Master signature list Master equipment list
Reference standards log Organizational chart Instrument logs
Employee training binder
(including training on SOPs, System suitability and instrument
Instrument use logs
methods, protocols, quality failure logs.
agreements and other)
Workbook log Employee job description Environmental chamber logs
Change control log Employee curriculum vitae

Special Edition: A Roadmap to GMP Compliance Part 3 59


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

Table IV: Useful information captured on controlled forms

Test Article/Sample
Study Transmittal Form Kick-off Meeting Checklist
Transmittal Form
Date of transmittal Client code Meeting date
Name of client-coordinator Test article/sample description Study director
Study director contact info Number of units Study number & client code
Study sponsor contact info Lot number Study personnel in attendance
Check qualifications of all study
Test article identification Container-closure system
personnel
Review study protocol & quality
Listing of tests to be done Test article (sample) ID number
agreement (if any)
Client code Tests to be performed Review test methods
Handling and storage Review test article handling and
Study number
conditions storage and MSDS data
Study title and objective Test specifications or ranges Review reference materials used
Is there a quality agreement in Client, client-coordinator and
Review results reporting requirements
place (Y/N)? QA signatures and dates
Date the study was forwarded to Check qualification of equipment &
the DCU instruments

Study Number, Study File and Client Code where they can aid in process execution.
GLP documentation has four simultaneous goals:
Peripheral Records and the Snap-Shot-in-Time
Capture in the study record all the information Idea
required to reconstruct the study at a later date At any time the company should be able to demon-
Contain an audit of the study to that study strate from the written recordthat at the time of the
Facilitate rapid recovery of the complete study particular analysis or test, the following were true:
record and
Protect client confidentiality. A ll analysts were qualified to perform the analysis
The first goal is reached primarily by designing a and followed a validated or verified test method
workbook that is self-contained while also linking using a qualified/calibrated instrument
to all the supporting study records. The second is Test and control articles and reference standards
reached primarily by segregating lab work by study were properly handled and stored at all times
i.e. by study number. The third is reached primar- Workbook was used to record experimental data
ily by creating for each study a study file. The study Instrument performed properly throughout the
file is maintained by the DCU following an SOP that analysis
states what must be filed and when. The fourth is A nalyst did not deviate from approved procedures
reached primarily by identifying clients by an as- and
signed code that is unintelligible to outsiders. If significant changes were made having the poten-
tial to impact the quality or integrity of the study
Checklists (e.g., the study director was replaced), then these
Checklists greatly simplify study conduct. For ex- were logged in the Change Control Log and were
ample, in order to start experimental work, the study evaluated and approved by the appropriate unit.
director makes readiness checks using the Kick-off The reader following the work-flow (Figure 1)
Meeting checklist (Table IV). This document ensures through a single GLP study should keep in mind
participating scientists understand the study protocol that since it is a single study, all documents can be
requirements and are qualified to perform the tasks one-time-use. Likewise, procedures such as test
they are being asked to perform. Checklists should procedures, calibrations, and unit lab operations can
reflect good planning and be placed in the workflow be written into the study protocol and approved since

60 Special Edition: A Roadmap to GMP Compliance Part 3


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

they are not needed again. The company can set aside a limited number of instruments, equipment, and per-
sonnel to be qualified those required to support the study. All of the experimental preparations and results
can be recorded contemporaneously in a single lab notebook. For a one-time single study, gaining control over
the study documents is a fairly trivial matter.
Even for a single study, however, the lab must meet a large number of GLP regulatory requirements. Those
that are most relevant to the present article are paraphrased and listed in Table V with reference to the cor-
responding requirement found in Title 21 of the Code of Federal Regulations, Part 58 (21CFR, Part 58, for
short). They were put into place by Congress in the late 1970s to ensure the Food and Drug Administration
that results from nonclinical laboratory studies reported to the agency through new drug applications were
valid and accurately reflect study conduct.

Table V: Some GLP study requirements (with reference to 21 CFR, Part 58)
The study must at the start be identified as a GLP study, meaning that the work is to
1
be conducted in compliance with the GLPs.
2 A study director must be assigned by Management. 58.31
3 The study director must write a study protocol. 58.120
The protocol must clearly indicate the objectives and all methods for the conduct of the
4 58.120
study.
5 The study must be placed onto a Master Schedule. 58.35
Management must ensure that an independent Quality Assurance Unit (QAU) is in
place to monitor the study, to report findings back regularly to the study director and
6 study director Management, to review the study final report to ensure that it accurately 58.31 58.35
describes the findings and conduct of the study and to write the Quality Assurance
Statement.
7 The study director must ensure that the lab is prepared to start the study.
The lab must have an area clearly designated for GLP test article (sample) receipt and
8 58.47
ensure that the test article cannot be confused with other substances e.g. regents.
Test articles, control articles and reference standards must be properly stored. It is
9 58.107
typical to dedicate a space that is under environmental control and has limited access.
Participating scientists must be qualified to play their role, which must be clearly
10 58.29
defined.
11 Written and approved procedures (e.g. SOPs) must be in place and followed. 58.81
Any and all deviations from written approved procedures (e.g. SOP deviations) must be 58.31 58.33
12
approved by the study director and documented in the raw data (e.g. workbook). 58.35
All raw data collected in the conduct of the study must be recorded following good
13 58.130
documentation practices and be readily retrievable.
Equipment and instrumentation must be shown to be fit for its intended use (e.g.
14 calibrated or standardized or qualified) and there must be a written record of this 58.63
activity.
A responsible person e.g. metrologist must be assigned to the inspection, cleaning,
15 58.63
maintenance, qualification of each instrument or piece of equipment.
16 Instrument failures must be investigated and repairs documented. 58.63
17 All reagents and solutions must be properly labeled. 58.83
18 QAU must perform at least one audit of the experimental work in progress. 58.35
19 A final report must be prepared, signed and dated by the study director. 58.185
20 The study must be archived in accordance with the GLPs. 58.190

Special Edition: A Roadmap to GMP Compliance Part 3 61


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

COMPLETING SIMULTANEOUS, MULTIPLE STUDIES DCU, and Archivist


Resource sharing is key to scaling-up to perform mul- Templates for study final reports, protocols, work-
tiple simultaneous studies. Scale-up is facilitated by books and logbooks and
copying procedures that are common to many studies Standardized forms to capture information in sup-
from study-specific documents such as the study pro- port of the study other than raw data.
tocol and pasting them into templates under docu- Table VI contains a list of some of the essential
ment control for use in other studies. This involves SOPs that must be in place and Table IV is a list of
creating the following: useful information that is captured on standardized
Workbooks and logbooks forms. The importance of having an SOP on Defini-
Standard Operating Procedures (SOPs) that de- tion and Preservation of Raw Data cannot be under-
scribe how to perform daily lab unit operations estimated. It will establish what must be recorded as
A nalytical test procedures to be used by multiple raw data and how. Any and all documents used in
studies recording raw data (e.g. workbooks and logbooks)
SOPs that contain the procedures to be followed must be under control by the DCU.
by the Quality Assurance Unit (QAU), Metrologist,

Table VI: Some essential SOPs


General lab operation Metrology Quality Assurance
Standard Operating Procedure:
Master Plan for Instrument Quality Assurance Unit Activities and
Administration, Distribution,
and Equipment Qualification Responsibilities
Maintenance and Training on SOPs
Management Commitment to The Master Instrument
Qualification of QAU auditors
Quality (Equipment) List
Use of Instrument Numbers,
Training and Qualification of
Labels, and Responsible Master Schedule
Personnel
Persons
Responsibilities of the Study Creation and Use of
GLP Study Protocol Review
Director Instrument Logbooks
Writing GLP Study Protocols Installation Qualification (IQ) Monitoring In-house GLP studies
Writing GLP Study Final Reports Calibration Schedule Audit of GLP Study Records
Receiving, Logging, Storing,
Operational and Performance
Testing, Archiving and Disposing of Audit of the GLP Study Final Report
Qualification (OQ and PQ)
GLP samples or test articles
OQ, PQ and Preventative
Definition and Preservation of Raw Reporting QAU audit results to Study
Maintenance on Temperature-
Data Directors
controlled Storage Areas
Use of Lab Workbooks, Logbooks, Writing the Quality Assurance
Analytical Balance
and Instrument Use Logs Statement
Document Control Lab Automatic Dishwasher Quality Systems Audit Schedule
Archiving Temp and Humidity Sensor Hosting Regulatory Visitors
Change Control Instrument Change Control Handling Complaints
Investigations
Metrology Investigations QAU File Maintenance and Archiving
(suspect data and OOR data)
Deviations Qualification by a Vendor Vendor Qualification
Corrective and Preventative Actions
Lab Fume Hoods Quality Agreements
(CAPA)

62 Special Edition: A Roadmap to GMP Compliance Part 3


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

Templates, standardized forms, and the company them into the workbook logbook. Participating sci-
organizational (org) chart should be controlled by entists log out the workbook they need and proceed
the DCU. Regarding the later, a format that identifies to perform experimental work as per the appropriate
each employee by job title and function is helpful. For study protocol, SOPs and written test methods, while
example, an employee might be identified on the org recording raw data contemporaneously in the work-
chart as Scientist II and reference standards coordina- book (Table I). The QAU will perform at least one
tor. An auditor can review the org chart, single-out audit of the experimental work while it is in prog-
this employee, and find in the employees training ress. QAU uses an audit checklist and a standard-
binder the following: a job description that includes ized report form to report audit findings back to the
the roles of reference standards coordinator and Sci- study director and the study directors management.
entist II, a curriculum vitae that shows the employee Workbooks are subject to peer-review then submit-
is qualified to serve as reference standards coordi- ted to the QAU for review. The QAU will release
nator, and Scientist II and documentation of all the the workbook after all corrections have been made,
other relevant GLP training (e.g. training on SOPs) i.e., all related Corrective and Preventative Actions
that the employee has received to date. An outside au- (CAPA)) are closed-out. The study director writes the
ditor will review the org chart as part of their effort to study final report, which includes all of the data (i.e.,
determine that the company has adequate resources good, bad and ugly). The study director references in
(e.g. space, qualified lab personnel, instrumentation, the report the study protocol, all workbooks used in
qualified QAU personnel, study director) to conduct the study and all investigation reports (e.g. suspect
all of the GLP studies that are on the Master Sched- data investigation) generated by the study. The study
ule. Keeping a master signature list that includes each director attaches the compliance statement to the
employees printed name, signature, and the initials report that testifies to the fact that the study was con-
they will use on GLP documents is an excellent idea. ducted under GLP and lists out any and all significant
Similarly, keeping an updated master equipment list non-compliances. The QAU reviews the study final
is expected of the lab. report to ensure that the report accurately reflects the
Figure 1 provides the workflow through a study conduct of the study. The QAU writes the Quality As-
in brief. What follows is a fairly detailed look at the surance Statement, listing out each phase of the study
study workflow (starting with receipt of the study that was audited, the date of each audit and the date
and ending with study archival) with references along that audit findings were reported to the study director
the way to procedures and forms that are helpful. and the study directors Management. The study final
The Study Transmittal Form (Table IV) is used report, compliance statement, QAU audit checklist
to identify the GLP study, test article, sponsor, and and QA Statement are all template-based, with the
study director and provide information that is re- template and all its revisions controlled through one
quired for placing the study onto the Master Schedule or more SOPs. The study director signs and dates the
and for creation of the study file by the DCU. By writ- study final report and submits it and all other study
ing the study protocol, the study director establishes materials to the archives. An exact copy of the study
control over the study. The protocol and its template final report is issued to the sponsor.
are controlled through the DCU. Once the protocol
is signed by the study director, the QAU places the THE QUALITY SYSTEMS APPROACH
study onto the Master Schedule. The Kick-off Meet- The QAU will acquire data on the performance of
ing checklist (Table IV) is used by the study direc- personnel, instrumentation, equipment, and facil-
tor to ensure that the lab is ready to start the study. ity from multiple studies over a period of time.
Test articles are received in the designated area by Examples include the number of CAPAs identified
the sample-coordinator following an SOP (listed in with an individual and the number of instrument
Table VI under the title Receiving, Logging, Storing, failures associated with an instrument. On a higher
Testing, Archiving and Disposing of GLP Samples level, the QAU collects data on how well the various
or Test Articles) and using the Test Article/Sample functions are performing. An example is the use of
Transmittal form (Table IV). The later form prompts workbooks: Are the workbook templates being ap-
both sender and receiver to record vital informa- propriately revised over time? Are workbooks being
tion about the test article. Test articles (and control used properly? Logged properly? Controlled properly?
articles) are logged into storage under those condi- On still a higher level, the QAU collects data on the
tions specified on the Test Article/Sample Transmittal various systems in place to control the functions,
form. Their condition upon receipt is noted and any e.g., the DCU, Training, Metrology, Vendor Quali-
chain-of-custody paperwork is filed in the study file. fication, Change Control, and Archiving systems.
The DCU prints the workbooks for the study and logs Are investigations conducted in accordance with the

Special Edition: A Roadmap to GMP Compliance Part 3 63


Richard C. Wedlich, Agata E. Libera, Joseph M. Fransen, C. Tellarini, T. Heath, N. Knight

investigations SOP? Are personnel being trained on VII. They operate in cafeteria-like fashion, ensuring
SOP revisions? Is Metrology on top of the calibration/ that the resources are available in order to meet the
qualification schedule? Are changes to the facility air- various requirements (on personnel, procedures, in-
handling system receiving the proper review and ap- strumentation, equipment, available lab space, proper
proval before being made? We refer to these systems storage conditions, data recording and retrieval, and
as quality systems, although in the larger scheme, others) for conducting the study under GLP. The per-
Good Laboratory Practice is the quality system (4). All formance of each quality system is continually moni-
of these data are developed by the QAU and reported tored by the QAU, who reports back to management,
to management with the aim of identifying and cor- who in turn, assures that corrective and preventative
recting negative or disruptive trends. Some key qual- actions are taken when needed. The QAU will follow-
ity systems used in a GLP program are listed in Table up on the CAPA to ensure that it was effective.

Table VII: Key quality systems (sub-systems) to GLP


Written procedures (SOPs) Test article (sample) receipt LIMS*
Metrology Reference standards Training
Quality Assurance Unit (QAU) Deviations and planned changes Complaints
Vendor qualification Change Control Investigations
Document Control (DCU) & Corrective & Preventative Actions
Continuous improvement
Archiving (CAPA)

*Laboratory Information Management System

out a laboratory program, complete with the support-


IMPLEMENTATION ing functions of document control, records retention
Having and following good SOPs is the key. Closely and quality assurance and backed by a committed
related is the job of training personnel on SOPs management. When installed and implemented, it
and revisions and of dealing with deviations and will become self-correcting and compliant with the
change. The former is handled through the training GLPs. In this way, a quality culture will become a
program, while the latter are handled through the reality in the organization.
CAPA and Change Control programs, respectively.
It is important to understand that implementing REFERENCES
is an iterative, learning process for the lab. Discrep- Code of Federal Regulations, Title 21, Good laboratory
ancies (gaps), redundancies and inconsistencies in practice for non-clinical laboratory studies (Govern-
the program, along with other shortcomings reveal ment Printing Office, Washington, DC), Part 58.
themselves upon implementation. It is a goal for the E.B. Wilson Jr., Introduction to Scientific Research,
process, through continual improvement, to con- McGraw-Hill Publishers, New York, NY, 1952.
verge on what we can call absolute compliance. The J.P. Seiler, Good Laboratory Practice, Springer, New
adequately designed program will improve, become York, NY, 2nd ed., 2005.
self-correcting and at some point highly-stable FDA, Draft Guidance for Industry: Quality Systems
and efficient. The poorly designed program will not Approach to Current Good Manufacturing Practice Regu-
get there. It will grow into a beast, becoming a con- lations (Rockville, MD, Sept., 2004).
stant drain on resources, resulting in lower quality
and a decreased potential for the lab to ever perform
quality work. The companys leadership will reflect
the will, drive, and commitment to implement the
GLP program.

SUMMARY
This discussion has provided detail in describing a
real GLP program so that the reader relatively new
to GLP can get a firm grip on the subject. Having the
correct, clearly stated, and understood goal, one maps

64 Special Edition: A Roadmap to GMP Compliance Part 3


Jerry Lanese and Timothy J. Fields

GXP Talk: Questions 66 & 67 |


IVT
Jerry Lanese and Timothy J. Fields, editors

A Column for Discussion of Issues Identified by Readers of the Journal of GXP


Compliance.
This column is one of a continuing series in which representatives of the in-
dustry and the regulatory agency have an opportunity to ask questions about,
and express opinions on, current GXP issues. The Editors welcome your ques-
tions and your opinions on the questions.

QUESTION 66
If we receive a raw material from a supplier and it does not meet our
specifications should the event be captured in our deviation system, the
suppliers deviation system, or tracked in some separate system?

ANSWER
The failure of an incoming raw material component should be entered
into a defined system that is established to log the failure, manage the
investigation and determination of the cause of the failure and recom-
mend corrective and/or preventive actions. This might be a system
committed only to managing component failure investigations. In most
organizations, it would be the site deviation system designed to log all
deviations and manage the investigation, determination of corrective
and/or preventive actions and the CAPAs. It is entirely up to the orga-
nization how it manages the follow-up to a component failure. There
must be a system, however, for timely determination of the cause and
follow-up, the process must be defined (there should be a procedure or
procedures), the procedure(s) must be followed and records must be
completed, reviewed, and approved by the quality unit and retained.
In the specific case of a raw material failure, it is very likely that the
supplier will be involved. The failure also will be entered into the sup-
pliers system, and the supplier will be responsible for a significant part
of the investigation and follow-up. General supplier responsibilities
in the case of a component failure should be discussed in the Quality
Agreement.

QUESTION 67
The most cited FDA observation is not following procedures. How does
an organization eliminate or justify human error such as not following
procedures?

ANSWER
Yes, this is a common problem with many potential underlying reasons

Special Edition: A Roadmap to GMP Compliance Part 3 65


Jerry Lanese and Timothy J. Fields

that must be explored before providing an answer. also welcome your questions for responses from read-
One potential reason for not following a procedure ers. Please submit your responses to the above or new
is that the procedure is unclear or lacks sufficient questions for consideration to Jerry Lanese or Tim
detail regarding how to perform a task. If the tasks to Fields. You may also to any previous questions via the
be executed are not clearly defined, the person may comments section below.
deviate from the SOP to complete the task. Another
reason for not following the procedure may be due to
training. Often on the job training is provided by a
more experienced person, who may have developed
shortcuts over the years and who then, in turn,
teaches these shortcuts to the new person. These
shortcuts may deviate from the actual SOP. Getting
the people who perform the tasks to write the SOP,
easily solves the above examples. Their input helps
ensure the SOP is written in a way that they under-
stand it and in the manner in which they actually
perform the tasks. They will feel a sense of owner-
ship which will encourage them to follow the SOP.
Another reason for not following SOPs is that
people get into a hurry, so again, they take shortcuts
or simply fail to perform steps as required by the
SOP. A common example of this is the failure to
document the performance of steps. People are hur-
rying to execute tasks and simply forget to document
that the tasks were completed as described in the
SOP. Ensuring that personnel have adequate time to
perform tasks or that adequate resources are available
can help address this issue.
Often SOPs are revised, and the people perform-
ing the tasks are not advised of the changes. They
continue, therefore, to follow the old SOP and are
unaware of the changes. This can be solved by
ensuring that personnel are advised and trained on
new or revised SOPS prior to performing the tasks.
Inadequate training is often an underlying cause of
failure to follow an SOP.
The last reason people do not follow SOPs is lack of
concern, i.e. they are just there for the paycheck. The
only way to resolve this problem is termination.
Mistakes happen; however, the goal is to eliminate
systemic errors that may be caused by some of the
reasons cited above.

Two new questions for comment.

QUESTION 68
The GMPs require that non-sterile products be free of
objectionable organism. What is an objectionable
organism?

QUESTION 69
Do we need to validate test methods for raw materi-
als, intermediates and in-process materials? If yes,
which of the characteristics should be evaluated?
Let us know your opinion on these questions. We

66 Special Edition: A Roadmap to GMP Compliance Part 3


Jerry Lanese and Timothy J. Fields

GXP Talk: Questions 68 & 69 |


IVT
Jerry Lanese and Timothy J. Fields, editors

A Column for Discussion of Issues Identified by Readers of the Journal of GXP


Compliance.
This column is one of a continuing series in which representatives of the in-
dustry and the regulatory agency have an opportunity to ask questions about,
and express opinions on, current GXP issues. The Editors welcome your ques-
tions and your opinions on the questions.

QUESTION 68
The GMPs require that non-sterile products be free of objectionable
organism. What is an objectionable organism?

ANSWER
21CFR Part 211.113(a) states: Appropriate written procedures, de-
signed to prevent objectionable microorganisms in drug products not
required to be sterile, shall be established and followed. This is further
supported by21CFR 211.165(b): There shall be appropriate laboratory
testing, as necessary, of each batch of drug product required to be free
of objectionable microorganisms. However, as is the case with many
cGMP requirements, the determination of what this means is left to the
firm. The definition of an objectionable organism is dependent on the
product or material in question. For example, the definition of an ob-
jectionable organism for an oral drug product may be different than the
definition of an objectionable organism in a topical product. The defini-
tion must consider the effect of the organism on the product as well
as the end users safety. If an organism can affect the product physico-
chemical or biological properties, it should be considered as objection-
able as it may alter the product beyond its defined quality, efficacy, or
purity requirements. Obviously any microorganism that can impact the
safety of the end user or that is considered to be pathogenic must be
considered as objectionable. A risk assessment should be performed to
determine if an organism should be considered as objectionable. The
risk assessment should include subject matter experts in microbiology
and medical personnel that can evaluate the potential risk to the end
user. As such organisms are encountered, a list should be prepared and
maintained of these organisms. As indicated in 21CFR 211.165(b), the
product should be tested for such organisms where necessary. If such
organisms are detected in the environment through the environmental
monitoring program, in water, or other raw materials, appropriate con-
trols should be implemented to eliminate the source.

Special Edition: A Roadmap to GMP Compliance Part 3 67


Jerry Lanese and Timothy J. Fields

QUESTION 69 Limit of Detection


Do we need to validate test methods for raw materi- Limit of Quantitation.
als, intermediates, and in-process materials? If yes,
which of the characteristics should be evaluated? These references also provide guidance as to which
characteristics should be studied for each type of
ANSWER method.
Yes. The requirement for test method validation It is appropriate to note that a USP Expert Panel
is found in 211.165(e), a subparagraph of 211.165, has recently published a stimulus article on the
Testing and Release for Distribution. From this, one lifecycle approach to test method validation. In this
might interpret that a laboratory is only required article, it is stated that ICH Q2(R1) and USP <1225>
to validate product release methods. However, the were originally intended to provide guidance but
GMPs were written a long time ago and the industry have come to be interpreted as mandatory practice.
has changed. It is current industry practice and FDA The article proposes the implementation of a guid-
expectation that all test methods used in the produc- ance that encourages method development chemists
tion and control of a pharmaceutical product are apply good science to understand and control sources
validated. This includes methods used for the testing of variability as they develop and validate a method.
of components such as starting materials and packag-
ing, in-process materials, and finished products. Two new questions for comment.
Whether or not required, demonstrating and docu-
menting that a test method performs as intended is QUESTION 70
good science and good business. Biological products are more variable than small
Both USP <1225> Validation of Compendial Meth- molecule products. How can one validate the process
ods and ICH Q2(R1) categorize test methods into as required by cGMP?
types of methods and list seven characteristics that QUESTION 71
should be considered in the validation of a chemical What is the regulatory requirement for process im-
test method. These include: provement in the pharmaceutical industry?
We welcome your opinion on these questions.
Specificity Also, let us know if you have any questions to ask
Accuracy of the industry. Please submit your questions or
Precision responses to Jerry Lanese or Tim Fields. You may also
Linearity replay to any previous questions.
Range

68 Special Edition: A Roadmap to GMP Compliance Part 3


Jerry Lanese and Timothy J. Fields

GXP Talk: Question 70 | IVT


Jerry Lanese and Timothy J. Fields, editors

GXP Talk provides a forum for compliance practitioners to address


issues identified by the readers of the Journal of GXP Compliance. GXP
Talk is the longest-running continuing series in the Journal of GXP Com-
pliance. This series was introduced in April, 2006. Previous discussions
addressed a wide range of compliance activities covering essentially all
sections of the US GMPs. Responses to questions and associated opinions
have been contributed by representatives from multiple pharmaceutical
industry and regulatory agencies.
Readers are invited to participate and contribute questions, answers,
and discussion for this series please share your successful practices
with others. This column succeeds when we are able to address cur-
rent GXP issues submitted by interested readers. Please contact column
coordinators Jerry Lanese at jerry@lanesegroup.com or Tim Fields at
timfields213@comcast.net with comments or submissions for publication.
We welcome your questions and your opinions on questions.

QUESTION 70

What are the Regulatory Requirement and Expectations for Process


Improvements in the Pharmaceutical Industry?

ANSWER
21CFR211.180, GMP for Finished Pharmaceuticals (1) states that re-
cords for each product must be reviewed at least annually to determine
the need for changes to drug product specifications or manufacturing
control procedures. This is known as the annual product review and
the intent is that the firm should identify and implement product and
process improvements. The GMPs are regulations -- An annual prod-
uct review must be conducted.
In addition to the above legal requirements, more recent guidance
documents support the concepts of continual improvement. Although
these are not regulations, they do express the regulatory expectations.
Regulations are interpreted and enforced consistent with these guid-
ances. As regulatory agency investigators become more familiar with
these guidances, expectations for ongoing improvement initiatives will
increase.
ICH Q8(R2), Pharmaceutical Development (2), discusses product life-
cycle management and continual improvement. Companies have op-
portunities to evaluate approaches to improve product quality. Process
performance should be monitored by trend analysis to confirm predict-

Special Edition: A Roadmap to GMP Compliance Part 3 69


Jerry Lanese and Timothy J. Fields

ed quality attributes. Changes in the design space 3. ICH Q10, Pharmaceutical Quality System, June
may occur based on additional process knowledge. 2008.
ICH Q10, Pharmaceutical Quality System (3), states 4. Yu, Lawrence X, Gregory Amidon, Mansoor A.
that one of the objectives of a Pharmaceutical Quality Khan, Stephen W. Hoag, James Polli, G.K.Raju, and
System is to facilitate continual improvement and Janet Woodcock. Understanding Quality by Design.
provides guidance for the continual Improvement of AAPS J, 2014 Jul; 16(4): 771-783, accessed 10-4-15.
process performance and product quality through- 5. Guidance for the Industry: Process Validation,
out the product lifecycle. General Principles and Practices; FDA, January, 2011.
The Quality by Design (QbD) initiative, first identi- 6. http://ec.europa.eu/health/files/eudralex/vol-4/
fied by Juran and later applied to pharmaceutical vol4-chap1_2013-01_en.pdf, accessed 10-3-15.
manufacturing, was the basis for focus on continual
improvement as described in subsequent ICH and Future Questions and Discussion Topics
FDA guidances. QbD provides a systematic approach The following are questions submitted by readers for
to product development emphasizing process under- discussion in future issues of GXP Talk. Comments
standing. Process understanding coupled with actual on any of the following as well as new questions for
experience and capability analysis should identify discussion are invited. Further discussions on previ-
process improvements. Five phases in continual ous questions (link) are also invited. Submit ques-
improvement activities have been identified. Con- tions, comments, or topics for discussion to column
tinual improvement is applicable to new products and coordinators Jerry Lanese at jerry@lanesegroup.
legacy products with historical manufacturing experi- com or Tim Fields at timfields213@comcast.net. We
ence. A recent paper by Yu et. al. (4) overviews the welcome your input this column succeeds because
QbD process and provides numerous references. of reader input!
The FDA Guidance on Process Validation (5)
suggests that data gathered during the commercial Pending Questions
manufacturing stage of the validation lifecycle might The following are questions that have been received
suggest ways to improve and/or optimize the process and will be answered in the future. We are interested
by altering some aspect of the process or product, in your input as we formulate the responses.
such as the operating conditions (ranges and set- 1. Biotech process validation. Biological products are
points), process controls, component, or in-process more variable than small molecule products. How
material characteristics. can one validate the process as required by cGMPs?
The revised European Union GMPs (6) also include 2. Raw materials. If a raw material does not meet
an expectation for continuous improvement. Al- specifications, should it be captured in the site de-
though the requirement for continual improvement viation, the suppliers deviation system, or another
in the US GMPs, which are now more than 35 years deviation system? Who should investigate this devia-
old, is a matter of interpretation, it is a regulatory tion -- the firm or the supplier?
requirement in the EU. It is clear from the above 3. Manufacturing yield. What are requirements for
regulations, guidances, and associated papers that documenting manufacturing process yield?
contemporary quality systems in the pharmaceutical 4. Personnel training. What are regulatory require-
industry must have a program of continual improve- ments and expectations for personnel training in
ment. pharmaceutical manufacturing and associated activi-
ties?
References 5. What disciplines are required to approve change
1. Current Good Manufacturing Practice for Finished controls?
Pharmaceuticals; 21CFR 211.180(e). 6. How frequently should data be trended?
2. ICH Q8(R2), Pharmaceutical Development. Au-
gust, 2009.

70 Special Edition: A Roadmap to GMP Compliance Part 3


Jerry Lanese and Timothy J. Fields

GXP Talk: Questions 71 & 72 |


IVT
Jerry Lanese and Timothy J. Fields, editors

GXP Talk provides a forum for compliance practitioners to address


issues identified by the readers of the Journal of GXP Compliance. GXP
Talk is the longest-running continuing series in the Journal of GXP Com-
pliance. This series was introduced in April, 2006. Previous discussions
addressed a wide range of compliance activities covering essentially all
sections of the US GMPs. Responses to questions and associated opinions
have been contributed by representatives from multiple pharmaceutical
industry and regulatory agencies.
Readers are invited to participate and contribute questions, answers,
and discussion for this series please share your successful practices
with others. This column succeeds when we are able to address cur-
rent GXP issues submitted by interested readers. Please contact column
coordinators Jerry Lanese at jerry@lanesegroup.com or Tim Fields at
timfields213@comcast.net with comments or submissions for publica-
tion. We welcome your questions and your opinions on questions.

QUESTION 71
Biological products are more variable than small molecule products.
So, how can one validate the biological process, as required by the
FDA?

ANSWER
The U.S. cGMPS, as codified in 21CFR 210-211, do not distinguish
between requirements for large molecules (i.e., Biologics) and small
molecule products. 21CFR 211.110(a) requires that control proce-
dures be established to monitor the output and validate the perfor-
mance of those manufacturing processes that may be responsible for
causing variability in the characteristics of in-process materials and the
drug product. Certainly chemical reactions as used in small molecule
manufacturing processes are well defined and easily controlled relative
to a biological system. However, the same basic validation principles
apply to biologics as are applicable to small molecules: Understand the
sources of variation, detect the presence and degree of variation, under-
stand the impact of variation on the process and ultimately on product
attributes, control the variation in a manner commensurate with the
risk it represents to the process and product.
The FDA Guidance Document: Process Validation: General Princi-
ples and Practices published in 2011 also states that the process valida-
tion principles included in the guidance apply to biologics. This should
be expected since the purpose of validating a process is to ensure that

Special Edition: A Roadmap to GMP Compliance Part 3 71


Jerry Lanese and Timothy J. Fields

a safe product is consistently produced that has the Risk assessments should be considered in deter-
efficacy and quality attributes that it is intended to mining critical process parameters and their control.
have. Likewise the application of risk assessment tools as
The guidance documents breaks process validation part of evaluating changes or deviations are critical
into three major phases: for ensuring the process remains in a state of control.
Stage 1 Process Design: The commercial manu- The key to validating a process whether it is a bio-
facturing process is defined during this stage based logical process or a chemical process is to identify the
on knowledge gained through development and critical process parameters and understand the affect
scale-up activities. they have on the process and control their variability.
Stage 2 Process Qualification: During this stage,
the process design is evaluated to determine if the QUESTION 72
process is capable of reproducible commercial manu- If a raw material does not meet specifications should
facturing. it be captured in the site deviation, the suppliers
Stage 3 Continued Process Verification: Ongoing deviation system or another deviation system? Who
assurance is gained during routine production that should investigate this deviation, the firm or the sup-
the process remains in a state of control. plier?
Lets explore some examples of applying these
principles to biologicals. ANSWER
Stage 1 - Process Design: one example of ap- This is a very timely question because the FDA, and
plying process design concepts to biologicals is in other regulatory agencies, are scrutinizing deviations
purification steps such as chromatography. Small and deviation investigations. It is clear from pub-
scale studies can be performed to define the optimal lished observations that the regulators expect that all
conditions for purification to ensure good yields with deviations will be investigated, the cause identified,
high quality product. Conditions such as resin to be and appropriate corrective and/or preventive actions
used, pH, salt and detergent concentrations can all be implemented and verified within a reasonable time.
developed at small scale and then used to establish The paragraph cited in most FDA observations re-
processing parameters for full scale. Similar pro- lating to inadequate investigations is 21CFR211.194,
cess design studies can be used to establish the best Production Record Review, which states: All drug
growth conditions for upstream processing. product production and control records, including those for
Stage 2 Process Qualification: continuing with packaging and labeling, shall be reviewed and approved
the purification theme, the parameters defined at by the quality control unit to determine compliance with
small scale can be implemented and tested at large all established, approved written procedures before a batch
scale using confirmatory studies or batches. At this is released or distributed. Any unexplained discrepancy
stage it is critical to understand the impact of critical or the failure of any of its components to meet any of its
process parameters on the process. For example, specifications shall be thoroughly investigated, whether or
what happens if the pH is too high or too low, of not the batch has already been distributed.
what happens if the salt concentrations are too high It is clear that the regulation requires an investiga-
or too low. These parameters should be defined at the tion of any deviation from the expected. Since this
small scale and confirmed at full scale. Such experi- deviation was identified at the site of the final prod-
ments are too costly to perform at the full scale. uct production it should be captured in the deviation
Stage 3 Continued Process Verification: pro- system of that site and that site is responsible for
cesses should be continuously monitored to evaluate assuring a timely investigation of the deviation. Cor-
the robustness or the process. Deviations should be rective and/or preventive actions are also expected.
investigated to provide valuable data that may allow Which organization is responsible for conducting
extending operating ranges. However, it is critical the deviation investigation is dependent upon site
that when extending operating ranges that one has policies and procedures and agreements between the
clear understanding of the impact of the changes. supplier and the site. These agreements are gener-
Likewise when evaluating changes to a process, it ally spelled out in the quality agreement between the
is important to fully understand the impact on the product production site and the raw material supplier
process. For example, changes to operating param- and include communications between parties in the
eters for a bioreactor may increase yield, but may lead event of a raw material failure, responsibilities for
to lower purity or other by-products. Going back to failure investigations, and the time within which any
the process knowledge and understanding is critical investigation and follow-up actions are to be com-
for ensuring that the process remains in a state of pleted. Product production site deviation procedures
control. and time requirements should be consistent with the

72 Special Edition: A Roadmap to GMP Compliance Part 3


Jerry Lanese and Timothy J. Fields

quality agreement with the raw material supplier and Whether or not a process for the investigation of
provide additional requirements for actions taken a failure of a component is identified in agreements
internally. with the supplier, the product production site is
In the case sited, it is very likely that any cor- responsible for assuring the completion of an investi-
rective action or preventive action identified in the gation and appropriate follow-up actions in a timely
investigation will occur at the raw material suppliers manner.
site. This will complicate the product production
sites responsibility for assuring a thorough investiga- We want to encourage our readers to ask questions
tion and completion of corrective and/or preventive and comment on the answers. One of the purposes
actions. This deviation does raise questions about of this column is to generate a dialog within the FDA
the consistent quality of the incoming component. regulated industries. Please submit your questions
Identification of the cause of the problem and process or comments to Jerry Lanese or Tim Fields. You are
improvements by the supplier are important for the encouraged to comment on any previous questions or
maintenance of the quality of the incoming raw mate- answers.
rial, and therefore, the quality of the final product.

Special Edition: A Roadmap to GMP Compliance Part 3 73

Вам также может понравиться