SlideShare a Scribd company logo
1 of 53
PROCESS CHANGES AS
PART OF THE
LIFECYCLE OF A
PROCESS
Dagmar Meissner
January 2011
Outline
• Defining process change
• Assessing and implementing proposed
process changes
• Reducing the frequency of process
changes
• What went wrong? (Case studies)
• Conclusions
2
(Charles Darwin, 1809 – 1882)
“It is not the strongest of the species that survives, nor
the most intelligent that survives. It is the one that is the
most adaptable to change.”
3
DEFINING
PROCESS CHANGE
4
Drug Development Lifecycle
Discovery
Process Transfer
and Design Process Scale-Up Process Scale-up #2
and prelim. optimization and Optimization
Preparation for Commercialization
Clinical Trials
PreClinical Studies CommercializationPhase IIIPhase IIPhase I
Research
Process Development
Manufacturing
Drug Development
Willingness to change a process
depends on the stage of the product
6
PD/preclin. Phase I/II Phase III Commercial Scale
Flexibility
To Change
Changes
• Corrective Action
• Out of spec product (OOS)
• Scale-up/design issues
• Continuous Improvement
• Evolutionary process improvements
• New production platform
• Technology transfer from another company
• Improved process quality (cost)
• New administration method/packaging
• Innovation
• New technologies, scientific advances
7
ASSESSMENT AND
IMPLEMENTATION
8
Typical Protein Manufacturing Process
4
Fermentor
Capture Column Purification 1 Purification 2
Clarification
UF
Sterile filtration
Bulk Product
Cell Bank
E-14
Inocculation Train
Typical Process Changes
10
Cell System
Expression System
Cell Banking System
Drug Product
Excipients
Specification
Test methods
Drug Substance
Specification
Test methods
Purification/Downstream
Processing
Changes to purification steps
Changes to Scale (increased titers)
Production Bioreactor
Cell Growth and Harvesting
Scale-up
Production Process Analytical/Formulation/Packaging
Packaging
Container/closure system
Administration method
Formulation
It walks like a duck
It quacks like a duck
It is a duck
……… but is it the same duck???
Did the product change?
 identity  strength  quality  purity
How did it change and is it relevant?
11
Effect of Process Changes
9
Process Change
(Cell line, fermentation,
downstream processing)
Drug Substance/Drug Product
Chemical/biochemical Properties In vivo properties
Product Attributes to Verify
• Chemical Biochemical Properties
• Impurities
• Clearance (DNA, viral, HCP)
• Molecular weight
• Folding/disulfide bonds
• Glycosylation
• Aggregation
• Activity/Bioassay
• Physical Characterization
• Stability
• Etc.
• In vivo Properties
• Pharmacokinetics
• Safety
• Efficacy
13
Evaluate Proposed Changes
Decision if the proposed change can/should
be implemented
• Teams in place to evaluate proposed changes
and impact on product
• Implemented Decision Tree
• Risk Management
• Communication critical
“That natural selection generally acts with extreme slowness I
fully admit.” (Charles Darwin)
14
Creating a Process for a Drug Substance
(DS) is an Interdisciplinary Approach
The same principle applies to the evaluation of a proposed process change
ProcessDevelopment
QualityAssurance
Facilities/Engineering
Analytical
Validation
Manufacturing
PROCESS
Decision Process
16
Implementing Change
• Change control in place
• Realistic timelines and resource allocation
• Know the agency and their expectations
• Communication
• Good science
“The degree of regulatory flexibility is predicated
on the level of relevant scientific knowledge
provided.”
(ICH 8 guidelines)
17
Basis for Success
• Crossfunctional team
• Project Management
• Alignment of
objectives
• Effective
communication
• Early involvement of
critical group members
• Clear process in place
for evaluating and
implementing a
proposed change
PROCESS
Manufacturing
Analytical
Process
Development
Quality
Assurance
Facilities
Engineering
Validation
REDUCING
FREQUENCY OF
CHANGES
19
How to reduce frequency for changes
• Invest time upfront
• Analytical methods early in place
• Realistic release specifications
• Scientific understanding of process
• Proper characterization of product
• Process development and scale-up driven by science and
engineering rather than deadlines
• Determine key points where implementation is sensible (e.g.
before Phase III start), and combine multiple changes into
one submission in order to reduce cost
• Carefully evaluate the necessity for change
• Clear PRODUCT specifications
• Dosage form and size
• Concentration
• Administration method
20
Available Tools
• Analytical Methods
• Risk analysis (part of QbD program)
• Process Analytical Technology (PAT)
21
Analytical Method Development
• On the critical path for process development
• Should be done early, however, is often
neglected
• Proper characterization essential for process
optimization, technology transfer, scale-up
• Some informal analytical methods may be
developed within PD
• Qualified method in terms of accuracy,
reproducibility, linearity important for successful
data analysis
A Quick Word on Specifications
• Specifications should be as wide as reasonably
possible given the stage of the development
• Scaled up process may need wider specifications
than bench scale
• Don’t pick best case product for preclinical safety
studies but “realistic case”
• Caution to set specifications too early and too
tight
• Continue to gather data FIO with the goal of
setting some specifications later in time
From the FDA Tool Box: QbD
Quality by Design (QbD)
“[…] The demonstration of greater
understanding of pharmaceutical and
manufacturing sciences can create a basis for
flexible regulatory approaches “
(from ICH Q8 (R2))
24
Risk Analysis
• Large component of QbD
• Goal: identify and prioritize weaknesses of
process in terms of process parameters
• Outcome:
• List of process parameters that are considered
high risk;
• Ability to improve process robustness;
• Reduction of OOS and associated corrective
actions
25
Risk Analysis
• FMEA (Failure Mode and Effect Analysis)
• Team Approach
• Performed at every unit operation step
• Critical analysis of operating parameters
• Evaluation of risk of failure/process upsets and
potential impact
• Detection (D)
• Occurrence (O)
• Severity (S)
• Based on historical data from small and large
scale if available (data mining)
Risk Analysis
Occurrence Detection Impact/Severity
Very high Remote Very high
High Unlikely High
Rare Moderate Moderate
Remote Likely Minor
High None
IncreasedRiskNumber
Assign qualitative values for each parameter (Scale 1-10):
Risk Analysis (cont’d)
Outcome:
A means to prioritize the level of risk
associated with a specific step or
process/material parameter
Risk Priority Number, RPN
RPN = D x O x S
(Detection x Occurrence x Severity)
FMEA - Example
Item
No. Process Step
Operating
Parameter
Potential
Failure Modes
OccurrenceRating
DetectionRating
SeverityRanking
Risk
Priority
Number
Additional Controls
and/or Comments or
Risk Remediation
1 Column Load
Product
concentration
Titer too high in
load 4 4 4 64
Expand design space for
load concentration
2 Elution pH
Failed pH probe
solution prep
error 6 6 6 216
3 Elution conductivity
Failed probe;
solution prep
error 2 5 7 70
Consider in-line
conductivity
sensor/alarm; expand
design space
4 Elution Temperature
Faulty temp
control
5 Equilibration
Equilibration
volume
Equil stopped too
early; operator
error, flow cell
error
6
FMEA: Prioritization of Parameters
0
50
100
150
200
250
300
RPN
Outcome of FMEA
• Increased understanding of process
• Critical quality attributes
• CPP (Critical process parameters)
• CMA (Critical material attributes)
• Safe operating ranges
• Partial understanding of design space
• Prioritized punch list of equipment/process
issues
• Prioritized list of operating parameters to
expand design space
Process Analytical Technology
“PAT is a system for designing, analyzing,
and controlling manufacturing through timely
measurements of critical quality and
performance attributes of raw and in-process
materials and processes with the goal of
ensuring final product quality.”
www.fda.gov/AboutFDA/CentersOffices/CDER/ucm088828.html
 "real time“ quality assurance
32
PAT – Example 1
In-line buffer dilution with pH and conductivity control for
chromatography step (isocratic or gradient)
33
Conc. Acid
Salt solution
Recirc pump
WFI
cond pH
Waste
Chromatography
skid
Buffer Composition Control by Mass
Flow versus PAT
Courtesy of: Michael Li, Asahi Kasei BioProcess
PAT Example: chromatography
Gradient control for the separation of trypsinogen and ribonuclease A
using IEX chromatography using mass flow control versus PAT based
on pH and conductivity
Courtesy of: Michael Li, Asahi Kasei BioProcess
Chromatogram
using Mass
Flow
Chromatogram
using PAT
PAT – Example 2
• Continuous monitoring system for a protein
refolding step
• Goal: Consistency in product quality and
process performance across batches
• Protein: Recombinant human endothelial growth
factor (rhVEGF)
• Based on understanding of oxygen needs during
the reaction and its impact on quality
• Monitoring %DO maintaining constant oxygen
transfer KLa across scales 3L, 2,000L, 15,000L
Protein Refolding Using PAT
Courtesy of: Shelly Pizarro, Genentech
Experimental Setup:
Control of O2/N2 sparging based on DO profile in reactor at constant kLa
:
Protein Refolding Using PAT
Courtesy of: Shelly Pizarro, Genentech
Refold reaction:
• Reduction of monomers over time
• Creation of main peak (rhVEGF)
• After 3-6 hrs oxidized and misfolded
species appear
• Process Analytical Tool: DO
 Stop reaction at specific point in DO
profile rather than just time to minimize
misfolds
PAT Summary
• PAT increases robustness of process
by controlling key parameters directly
• Reduced frequency of OOS
• Increased process understanding
• It comes at an increased cost
(development time, equipment cost,
validation)
39
WHAT WENT WRONG?
Case Studies (n=3)
40
483s – Case Study I
AMAG Pharmaceuticals, Inc.
• Engineering modified wiring connections on
the same cooling relay which services the
Clean Room complex . This change caused
the relative humidity in the Clean Room
complex to exceed the established
specification […]. Therefore, Production and
Quality Control approved the relative humidity
specification change to XX %. A change
control was not executed to scientifically
evaluate how this specification change would
affect operations and quality of the Clean
Room […].
41
483s – Case Study II
• Applied Laboratories Inc.
• Your change control procedure […] was found
to lack provisions for complete documentation
of the nature and approval of production and
process changes, (FDA-483 #14) as required by
21 CFR 820.70(b).
42
Case Study III
Nature Biotechnology 26, 592 (2008)
(by George Mack)
• FDA balks at Myozyme scale-up
• Genzyme ran into a snag in April [2008] when the
US Food and Drug Administration (FDA) rejected
its application to produce Myozyme
(alglucosidase alfa, rhGAA) in its 2,000–liter-scale
facility under the same approval authorization
given for its 160-liter-scale plant. The FDA says
the carbohydrate structure of the products
manufactured at each scale differs and thus the
2,000-liter product requires a new biologic license
application.
43
Myozyme becomes Lumizyme after
biologics scale-up
• In February 2009, Genzyme was scheduled to
launch the same biologic under two different
names in the US after the FDA decided the
drug produced at 2000L was considerably
different to the 160L version.
• Approval for Lumizyme was gained in May
2010 (after additional delays due to mfg
issues)
 15 month delay
44
To Conclude
• Don’t be afraid of change, it is part of evolution
• Carefully evaluate and plan a process change
• Implement strategies for risk management
• Have procedures in place (documentation, validation,
additional preclinical or clinical studies, etc)
• Work with FDA
• Use scientific knowledge, historical data, experimental
design, and common sense
FDA and Industry have a common objective to ensure that high quality
pharmaceutical products continue to be available to the public.
(from: PAT workgroup presentation)
45
Thank you!
Special Thanks to:
• ISPE
• Michael Li (Asahi Kasei Bioprocess)
• Henriette Kuehne (Amylin)
• Mayank Patel (PAXVAX)
• Jörg Thömmes (Biogen IDEC)
• Shelley Pizarro (Genentech)
References
• ICH Q8R2 guidelines (2008) - http://www.ich.org/cache/compo/276-254-1.html
• ICH Q9 guidelines (2005) - http://www.ich.org/cache/compo/276-254-1.html
• Maximizing Uptime for Mission-Critical Manufacturing Units; By Gary
Shamshoian, P.E., Genentech, Inc., and Don Nurisso, P.E., EYP Mission Critical;
http://www.pharmamanufacturing.com/articles/2007/008.html;
• Change within design space is not a regulatory change: Genentech official.
http://www.thefreelibrary.com/Change+within+design+space+is+not+a+regulatory+
change%3a+Genentech+...-a0174973741
• Use PAT to Dilute Buer from Concentrate with a Linear pH Gradient for
Downstream Bioprocessing. Michael Li, Hiroyuki Yabe, Shree Jariwala, and
Tomo Miyabayashi Asahi Kasei Bioprocess; Poster Presentation at BPI conference
9/2010
• Biomanufacturing process analytical technology (PAT) application for
downstream processing: Using dissolved oxygen as an indicator of product
quality for a protein refolding reaction. Shelly A. Pizarro, Rachel Dinges, Rachel
Adams, Ailen Sanchez, Charles Winter Biotechnology and Bioengineering
Volume 104, Issue 2, pages 340–351, 1 October 2009
48
and prioritizand prioritiz
Backup slides
49
Defining Operating Ranges
For a Single Operating Parameter
Design Space
51
Experimental Data
Design Space
Approved Process
Modeling used to support
Design Space Characterization
Looking at all critical process parameters and material attributes
PAT – Example 1
Courtesy of: Michael Li, Asahi Kasei BioProcess
Interrelation between Quality Attributes
and Process & Materials
Critical Quality
Attributes
Inputs Outputs
Material Attributes
Process
Parameters
MA1
MA2
CPP1
CPP2
CQA1
CQA3
CQA2

More Related Content

What's hot

Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...
Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...
Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...ijtsrd
 
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...Waters Corporation
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validationSUBHASISH DAS
 
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATION
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATIONCONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATION
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATIONArul Packiadhas
 
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...Stephan O. Krause, PhD
 
Handling of Refernce Standards_Dr.A.Amsavel
Handling of Refernce Standards_Dr.A.Amsavel Handling of Refernce Standards_Dr.A.Amsavel
Handling of Refernce Standards_Dr.A.Amsavel Dr. Amsavel A
 
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13Stephan O. Krause, PhD
 
The Role of Process Characterization in Process Validation
The Role of Process Characterization in Process ValidationThe Role of Process Characterization in Process Validation
The Role of Process Characterization in Process ValidationDavid Goodrich
 
ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationSACHIN C P
 
Qby d presentation-20jun12ppt
Qby d presentation-20jun12pptQby d presentation-20jun12ppt
Qby d presentation-20jun12pptabubacker siddiq
 
validation of analytical method used in cleaning
validation of analytical method used in cleaningvalidation of analytical method used in cleaning
validation of analytical method used in cleaningKUNDLAJAYALAKSHMI
 
Rtrt regulatory perspectiv on real time release testing 27-oct_2011
Rtrt regulatory perspectiv on real time release testing 27-oct_2011Rtrt regulatory perspectiv on real time release testing 27-oct_2011
Rtrt regulatory perspectiv on real time release testing 27-oct_2011lastrajl
 
The Analytical Method Transfer Process SK-Sep 2013
The Analytical Method Transfer Process  SK-Sep 2013The Analytical Method Transfer Process  SK-Sep 2013
The Analytical Method Transfer Process SK-Sep 2013Stephan O. Krause, PhD
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instrumentsFaris ameen
 
Strategies of assuring quality of laboratory results
Strategies of assuring quality of laboratory resultsStrategies of assuring quality of laboratory results
Strategies of assuring quality of laboratory resultsGodwin 'Damilare Emeka
 

What's hot (20)

Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...
Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...
Analytical Quality by Design Concise Review on Approach to Enhanced Analytica...
 
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...
Using Fusion QbD as an Analytical Quality by Design Software for Method Devel...
 
Validation 2
Validation 2Validation 2
Validation 2
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATION
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATIONCONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATION
CONCEPT & TYPE OF VALIDATION OF GOVERNMENT REGULATION
 
Process analytical chemistry
Process analytical chemistryProcess analytical chemistry
Process analytical chemistry
 
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...
FDA (invited) Presentation - Specifications and Analytical Method Lifecycle f...
 
instrument validation
instrument validationinstrument validation
instrument validation
 
Handling of Refernce Standards_Dr.A.Amsavel
Handling of Refernce Standards_Dr.A.Amsavel Handling of Refernce Standards_Dr.A.Amsavel
Handling of Refernce Standards_Dr.A.Amsavel
 
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13
Risk-based Analytical Method Validation and Maintenance Strategies SK-Sep13
 
ICH Q11 Slide Summary
ICH Q11 Slide SummaryICH Q11 Slide Summary
ICH Q11 Slide Summary
 
The Role of Process Characterization in Process Validation
The Role of Process Characterization in Process ValidationThe Role of Process Characterization in Process Validation
The Role of Process Characterization in Process Validation
 
ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validation
 
Qby d presentation-20jun12ppt
Qby d presentation-20jun12pptQby d presentation-20jun12ppt
Qby d presentation-20jun12ppt
 
validation of analytical method used in cleaning
validation of analytical method used in cleaningvalidation of analytical method used in cleaning
validation of analytical method used in cleaning
 
Rtrt regulatory perspectiv on real time release testing 27-oct_2011
Rtrt regulatory perspectiv on real time release testing 27-oct_2011Rtrt regulatory perspectiv on real time release testing 27-oct_2011
Rtrt regulatory perspectiv on real time release testing 27-oct_2011
 
The Analytical Method Transfer Process SK-Sep 2013
The Analytical Method Transfer Process  SK-Sep 2013The Analytical Method Transfer Process  SK-Sep 2013
The Analytical Method Transfer Process SK-Sep 2013
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
 
AMV final 20Jun17
AMV final 20Jun17AMV final 20Jun17
AMV final 20Jun17
 
Strategies of assuring quality of laboratory results
Strategies of assuring quality of laboratory resultsStrategies of assuring quality of laboratory results
Strategies of assuring quality of laboratory results
 

Similar to ISPE 2011 presentation Dagmar Meissner - 13Jan2011

Cmc biologics pathway_draft8
Cmc biologics pathway_draft8Cmc biologics pathway_draft8
Cmc biologics pathway_draft8enarke
 
The CMC Journey in the Regulation of Biologics
The CMC Journey in the Regulation of BiologicsThe CMC Journey in the Regulation of Biologics
The CMC Journey in the Regulation of Biologicsenarke
 
Quality by Design
Quality by DesignQuality by Design
Quality by DesignTeny Thomas
 
3980 s1 10_gardner
3980 s1 10_gardner3980 s1 10_gardner
3980 s1 10_gardnerVIJAY SINGH
 
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTS
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTSPHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTS
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTSMd. Saddam Nawaz
 
Understanding and Controlling Bioprocess Variation | Parker domnick hunter
Understanding and Controlling Bioprocess Variation | Parker domnick hunterUnderstanding and Controlling Bioprocess Variation | Parker domnick hunter
Understanding and Controlling Bioprocess Variation | Parker domnick hunterParker Hannifin Corporation
 
Quality by Design and Process Analytical Technology
Quality by Design and Process Analytical TechnologyQuality by Design and Process Analytical Technology
Quality by Design and Process Analytical TechnologyMANIKANDAN V
 
archive_presentations_march2004_ICH-Q5E-handout.ppt
archive_presentations_march2004_ICH-Q5E-handout.pptarchive_presentations_march2004_ICH-Q5E-handout.ppt
archive_presentations_march2004_ICH-Q5E-handout.pptRajakumari Rajendran
 
Process Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyProcess Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyMilliporeSigma
 
Process Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyProcess Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyMerck Life Sciences
 
LectureItechnology transfer6thJanuary2022 (1).pptx
LectureItechnology transfer6thJanuary2022 (1).pptxLectureItechnology transfer6thJanuary2022 (1).pptx
LectureItechnology transfer6thJanuary2022 (1).pptxSohailSheikh62
 
Quality by design for Pharmaceutical Industries: An introduction
Quality by design for Pharmaceutical Industries: An introductionQuality by design for Pharmaceutical Industries: An introduction
Quality by design for Pharmaceutical Industries: An introductionCovello Luca
 
From Screening to QC: Development Considerations for Octet Methods
From Screening to QC: Development Considerations for Octet MethodsFrom Screening to QC: Development Considerations for Octet Methods
From Screening to QC: Development Considerations for Octet MethodsKBI Biopharma
 
validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...Sharath Hns
 

Similar to ISPE 2011 presentation Dagmar Meissner - 13Jan2011 (20)

Cmc biologics pathway_draft8
Cmc biologics pathway_draft8Cmc biologics pathway_draft8
Cmc biologics pathway_draft8
 
The CMC Journey in the Regulation of Biologics
The CMC Journey in the Regulation of BiologicsThe CMC Journey in the Regulation of Biologics
The CMC Journey in the Regulation of Biologics
 
Quality-by-Design by chattar
Quality-by-Design by chattarQuality-by-Design by chattar
Quality-by-Design by chattar
 
Quality by Design
Quality by DesignQuality by Design
Quality by Design
 
3980 s1 10_gardner
3980 s1 10_gardner3980 s1 10_gardner
3980 s1 10_gardner
 
Qbd1
Qbd1Qbd1
Qbd1
 
Qbd1
Qbd1Qbd1
Qbd1
 
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTS
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTSPHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTS
PHARMACEUTICAL PROCESS VALIDATION CURRENT REGULATORY ASPECTS
 
Understanding and Controlling Bioprocess Variation | Parker domnick hunter
Understanding and Controlling Bioprocess Variation | Parker domnick hunterUnderstanding and Controlling Bioprocess Variation | Parker domnick hunter
Understanding and Controlling Bioprocess Variation | Parker domnick hunter
 
Quality by Design and Process Analytical Technology
Quality by Design and Process Analytical TechnologyQuality by Design and Process Analytical Technology
Quality by Design and Process Analytical Technology
 
archive_presentations_march2004_ICH-Q5E-handout.ppt
archive_presentations_march2004_ICH-Q5E-handout.pptarchive_presentations_march2004_ICH-Q5E-handout.ppt
archive_presentations_march2004_ICH-Q5E-handout.ppt
 
Process Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyProcess Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene Therapy
 
Process Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene TherapyProcess Development for Cell Therapy and Viral Gene Therapy
Process Development for Cell Therapy and Viral Gene Therapy
 
Quality by Design
Quality by DesignQuality by Design
Quality by Design
 
DCN Company Profile 2015
DCN Company Profile 2015DCN Company Profile 2015
DCN Company Profile 2015
 
LectureItechnology transfer6thJanuary2022 (1).pptx
LectureItechnology transfer6thJanuary2022 (1).pptxLectureItechnology transfer6thJanuary2022 (1).pptx
LectureItechnology transfer6thJanuary2022 (1).pptx
 
Quality by design for Pharmaceutical Industries: An introduction
Quality by design for Pharmaceutical Industries: An introductionQuality by design for Pharmaceutical Industries: An introduction
Quality by design for Pharmaceutical Industries: An introduction
 
From Screening to QC: Development Considerations for Octet Methods
From Screening to QC: Development Considerations for Octet MethodsFrom Screening to QC: Development Considerations for Octet Methods
From Screening to QC: Development Considerations for Octet Methods
 
Petition of the Day
Petition of the DayPetition of the Day
Petition of the Day
 
validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...
 

ISPE 2011 presentation Dagmar Meissner - 13Jan2011

  • 1. PROCESS CHANGES AS PART OF THE LIFECYCLE OF A PROCESS Dagmar Meissner January 2011
  • 2. Outline • Defining process change • Assessing and implementing proposed process changes • Reducing the frequency of process changes • What went wrong? (Case studies) • Conclusions 2
  • 3. (Charles Darwin, 1809 – 1882) “It is not the strongest of the species that survives, nor the most intelligent that survives. It is the one that is the most adaptable to change.” 3
  • 5. Drug Development Lifecycle Discovery Process Transfer and Design Process Scale-Up Process Scale-up #2 and prelim. optimization and Optimization Preparation for Commercialization Clinical Trials PreClinical Studies CommercializationPhase IIIPhase IIPhase I Research Process Development Manufacturing Drug Development
  • 6. Willingness to change a process depends on the stage of the product 6 PD/preclin. Phase I/II Phase III Commercial Scale Flexibility To Change
  • 7. Changes • Corrective Action • Out of spec product (OOS) • Scale-up/design issues • Continuous Improvement • Evolutionary process improvements • New production platform • Technology transfer from another company • Improved process quality (cost) • New administration method/packaging • Innovation • New technologies, scientific advances 7
  • 9. Typical Protein Manufacturing Process 4 Fermentor Capture Column Purification 1 Purification 2 Clarification UF Sterile filtration Bulk Product Cell Bank E-14 Inocculation Train
  • 10. Typical Process Changes 10 Cell System Expression System Cell Banking System Drug Product Excipients Specification Test methods Drug Substance Specification Test methods Purification/Downstream Processing Changes to purification steps Changes to Scale (increased titers) Production Bioreactor Cell Growth and Harvesting Scale-up Production Process Analytical/Formulation/Packaging Packaging Container/closure system Administration method Formulation
  • 11. It walks like a duck It quacks like a duck It is a duck ……… but is it the same duck??? Did the product change?  identity  strength  quality  purity How did it change and is it relevant? 11
  • 12. Effect of Process Changes 9 Process Change (Cell line, fermentation, downstream processing) Drug Substance/Drug Product Chemical/biochemical Properties In vivo properties
  • 13. Product Attributes to Verify • Chemical Biochemical Properties • Impurities • Clearance (DNA, viral, HCP) • Molecular weight • Folding/disulfide bonds • Glycosylation • Aggregation • Activity/Bioassay • Physical Characterization • Stability • Etc. • In vivo Properties • Pharmacokinetics • Safety • Efficacy 13
  • 14. Evaluate Proposed Changes Decision if the proposed change can/should be implemented • Teams in place to evaluate proposed changes and impact on product • Implemented Decision Tree • Risk Management • Communication critical “That natural selection generally acts with extreme slowness I fully admit.” (Charles Darwin) 14
  • 15. Creating a Process for a Drug Substance (DS) is an Interdisciplinary Approach The same principle applies to the evaluation of a proposed process change ProcessDevelopment QualityAssurance Facilities/Engineering Analytical Validation Manufacturing PROCESS
  • 17. Implementing Change • Change control in place • Realistic timelines and resource allocation • Know the agency and their expectations • Communication • Good science “The degree of regulatory flexibility is predicated on the level of relevant scientific knowledge provided.” (ICH 8 guidelines) 17
  • 18. Basis for Success • Crossfunctional team • Project Management • Alignment of objectives • Effective communication • Early involvement of critical group members • Clear process in place for evaluating and implementing a proposed change PROCESS Manufacturing Analytical Process Development Quality Assurance Facilities Engineering Validation
  • 20. How to reduce frequency for changes • Invest time upfront • Analytical methods early in place • Realistic release specifications • Scientific understanding of process • Proper characterization of product • Process development and scale-up driven by science and engineering rather than deadlines • Determine key points where implementation is sensible (e.g. before Phase III start), and combine multiple changes into one submission in order to reduce cost • Carefully evaluate the necessity for change • Clear PRODUCT specifications • Dosage form and size • Concentration • Administration method 20
  • 21. Available Tools • Analytical Methods • Risk analysis (part of QbD program) • Process Analytical Technology (PAT) 21
  • 22. Analytical Method Development • On the critical path for process development • Should be done early, however, is often neglected • Proper characterization essential for process optimization, technology transfer, scale-up • Some informal analytical methods may be developed within PD • Qualified method in terms of accuracy, reproducibility, linearity important for successful data analysis
  • 23. A Quick Word on Specifications • Specifications should be as wide as reasonably possible given the stage of the development • Scaled up process may need wider specifications than bench scale • Don’t pick best case product for preclinical safety studies but “realistic case” • Caution to set specifications too early and too tight • Continue to gather data FIO with the goal of setting some specifications later in time
  • 24. From the FDA Tool Box: QbD Quality by Design (QbD) “[…] The demonstration of greater understanding of pharmaceutical and manufacturing sciences can create a basis for flexible regulatory approaches “ (from ICH Q8 (R2)) 24
  • 25. Risk Analysis • Large component of QbD • Goal: identify and prioritize weaknesses of process in terms of process parameters • Outcome: • List of process parameters that are considered high risk; • Ability to improve process robustness; • Reduction of OOS and associated corrective actions 25
  • 26. Risk Analysis • FMEA (Failure Mode and Effect Analysis) • Team Approach • Performed at every unit operation step • Critical analysis of operating parameters • Evaluation of risk of failure/process upsets and potential impact • Detection (D) • Occurrence (O) • Severity (S) • Based on historical data from small and large scale if available (data mining)
  • 27. Risk Analysis Occurrence Detection Impact/Severity Very high Remote Very high High Unlikely High Rare Moderate Moderate Remote Likely Minor High None IncreasedRiskNumber Assign qualitative values for each parameter (Scale 1-10):
  • 28. Risk Analysis (cont’d) Outcome: A means to prioritize the level of risk associated with a specific step or process/material parameter Risk Priority Number, RPN RPN = D x O x S (Detection x Occurrence x Severity)
  • 29. FMEA - Example Item No. Process Step Operating Parameter Potential Failure Modes OccurrenceRating DetectionRating SeverityRanking Risk Priority Number Additional Controls and/or Comments or Risk Remediation 1 Column Load Product concentration Titer too high in load 4 4 4 64 Expand design space for load concentration 2 Elution pH Failed pH probe solution prep error 6 6 6 216 3 Elution conductivity Failed probe; solution prep error 2 5 7 70 Consider in-line conductivity sensor/alarm; expand design space 4 Elution Temperature Faulty temp control 5 Equilibration Equilibration volume Equil stopped too early; operator error, flow cell error 6
  • 30. FMEA: Prioritization of Parameters 0 50 100 150 200 250 300 RPN
  • 31. Outcome of FMEA • Increased understanding of process • Critical quality attributes • CPP (Critical process parameters) • CMA (Critical material attributes) • Safe operating ranges • Partial understanding of design space • Prioritized punch list of equipment/process issues • Prioritized list of operating parameters to expand design space
  • 32. Process Analytical Technology “PAT is a system for designing, analyzing, and controlling manufacturing through timely measurements of critical quality and performance attributes of raw and in-process materials and processes with the goal of ensuring final product quality.” www.fda.gov/AboutFDA/CentersOffices/CDER/ucm088828.html  "real time“ quality assurance 32
  • 33. PAT – Example 1 In-line buffer dilution with pH and conductivity control for chromatography step (isocratic or gradient) 33 Conc. Acid Salt solution Recirc pump WFI cond pH Waste Chromatography skid
  • 34. Buffer Composition Control by Mass Flow versus PAT Courtesy of: Michael Li, Asahi Kasei BioProcess
  • 35. PAT Example: chromatography Gradient control for the separation of trypsinogen and ribonuclease A using IEX chromatography using mass flow control versus PAT based on pH and conductivity Courtesy of: Michael Li, Asahi Kasei BioProcess Chromatogram using Mass Flow Chromatogram using PAT
  • 36. PAT – Example 2 • Continuous monitoring system for a protein refolding step • Goal: Consistency in product quality and process performance across batches • Protein: Recombinant human endothelial growth factor (rhVEGF) • Based on understanding of oxygen needs during the reaction and its impact on quality • Monitoring %DO maintaining constant oxygen transfer KLa across scales 3L, 2,000L, 15,000L
  • 37. Protein Refolding Using PAT Courtesy of: Shelly Pizarro, Genentech Experimental Setup: Control of O2/N2 sparging based on DO profile in reactor at constant kLa :
  • 38. Protein Refolding Using PAT Courtesy of: Shelly Pizarro, Genentech Refold reaction: • Reduction of monomers over time • Creation of main peak (rhVEGF) • After 3-6 hrs oxidized and misfolded species appear • Process Analytical Tool: DO  Stop reaction at specific point in DO profile rather than just time to minimize misfolds
  • 39. PAT Summary • PAT increases robustness of process by controlling key parameters directly • Reduced frequency of OOS • Increased process understanding • It comes at an increased cost (development time, equipment cost, validation) 39
  • 40. WHAT WENT WRONG? Case Studies (n=3) 40
  • 41. 483s – Case Study I AMAG Pharmaceuticals, Inc. • Engineering modified wiring connections on the same cooling relay which services the Clean Room complex . This change caused the relative humidity in the Clean Room complex to exceed the established specification […]. Therefore, Production and Quality Control approved the relative humidity specification change to XX %. A change control was not executed to scientifically evaluate how this specification change would affect operations and quality of the Clean Room […]. 41
  • 42. 483s – Case Study II • Applied Laboratories Inc. • Your change control procedure […] was found to lack provisions for complete documentation of the nature and approval of production and process changes, (FDA-483 #14) as required by 21 CFR 820.70(b). 42
  • 43. Case Study III Nature Biotechnology 26, 592 (2008) (by George Mack) • FDA balks at Myozyme scale-up • Genzyme ran into a snag in April [2008] when the US Food and Drug Administration (FDA) rejected its application to produce Myozyme (alglucosidase alfa, rhGAA) in its 2,000–liter-scale facility under the same approval authorization given for its 160-liter-scale plant. The FDA says the carbohydrate structure of the products manufactured at each scale differs and thus the 2,000-liter product requires a new biologic license application. 43
  • 44. Myozyme becomes Lumizyme after biologics scale-up • In February 2009, Genzyme was scheduled to launch the same biologic under two different names in the US after the FDA decided the drug produced at 2000L was considerably different to the 160L version. • Approval for Lumizyme was gained in May 2010 (after additional delays due to mfg issues)  15 month delay 44
  • 45. To Conclude • Don’t be afraid of change, it is part of evolution • Carefully evaluate and plan a process change • Implement strategies for risk management • Have procedures in place (documentation, validation, additional preclinical or clinical studies, etc) • Work with FDA • Use scientific knowledge, historical data, experimental design, and common sense FDA and Industry have a common objective to ensure that high quality pharmaceutical products continue to be available to the public. (from: PAT workgroup presentation) 45
  • 46. Thank you! Special Thanks to: • ISPE • Michael Li (Asahi Kasei Bioprocess) • Henriette Kuehne (Amylin) • Mayank Patel (PAXVAX) • Jörg Thömmes (Biogen IDEC) • Shelley Pizarro (Genentech)
  • 47. References • ICH Q8R2 guidelines (2008) - http://www.ich.org/cache/compo/276-254-1.html • ICH Q9 guidelines (2005) - http://www.ich.org/cache/compo/276-254-1.html • Maximizing Uptime for Mission-Critical Manufacturing Units; By Gary Shamshoian, P.E., Genentech, Inc., and Don Nurisso, P.E., EYP Mission Critical; http://www.pharmamanufacturing.com/articles/2007/008.html; • Change within design space is not a regulatory change: Genentech official. http://www.thefreelibrary.com/Change+within+design+space+is+not+a+regulatory+ change%3a+Genentech+...-a0174973741 • Use PAT to Dilute Buer from Concentrate with a Linear pH Gradient for Downstream Bioprocessing. Michael Li, Hiroyuki Yabe, Shree Jariwala, and Tomo Miyabayashi Asahi Kasei Bioprocess; Poster Presentation at BPI conference 9/2010 • Biomanufacturing process analytical technology (PAT) application for downstream processing: Using dissolved oxygen as an indicator of product quality for a protein refolding reaction. Shelly A. Pizarro, Rachel Dinges, Rachel Adams, Ailen Sanchez, Charles Winter Biotechnology and Bioengineering Volume 104, Issue 2, pages 340–351, 1 October 2009
  • 50. Defining Operating Ranges For a Single Operating Parameter
  • 51. Design Space 51 Experimental Data Design Space Approved Process Modeling used to support Design Space Characterization Looking at all critical process parameters and material attributes
  • 52. PAT – Example 1 Courtesy of: Michael Li, Asahi Kasei BioProcess
  • 53. Interrelation between Quality Attributes and Process & Materials Critical Quality Attributes Inputs Outputs Material Attributes Process Parameters MA1 MA2 CPP1 CPP2 CQA1 CQA3 CQA2