You are on page 1of 44

Bio-manufacturing and Facility of the Future:

benefits and challenges of recent innovations


Berthold Boedeker
Bayer Pharma AG; Biologics - Biotech Development

9th Bioinnovation Leaders Summit


Berlin, Feb. 2016

Page 1
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 2
Bayer provides solutions based on
innovations

In all areas of our business, we invent, develop and market new molecules which influence
the biochemical processes in living organisms.

Page 3
Our Life Science businesses
hold leadership positions
Life Sciences
Pharmaceuticals Consumer Health Crop Science1

!  Strong in research and !  No. 2 with leadership positions !  Crop Protection: no. 2 with a
development in key categories highly diversified R&D portfolio
!  Leadership positions in core (dermatology, gastrointestinal !  Seeds: no. 7 but with leading
therapeutic areas, e.g. in disease) and strong brand positions in canola, cotton,
recognition vegetables and rice
cardiology, ophthalmology,
women‘s health & certain !  Strong geographic footprint !  Animal Health: no. 3 in the
segments of oncology companion animal market
!  Focused on consumer-centric (CAP)2
!  Successful market launches, innovation
e.g. Xarelto, Eylea, Xofigo

FTE3 ≈38,000 ≈11,700 ≈26,800

Unique and diversified portfolio mitigates risks


1 Incl. Animal Health (will report as a business unit directly to Liam Condon)
2 Companion animal products
3 Expected headcount on January 1, 2016

Page 4
Best-Selling Pharmaceutical Products
3rd Quarter First Nine First Nine Change
[ € million ] 2015 Months 2014 Months 2015 Fx adj.
% %
Xarelto™ 1,163 1,602 +37.7 +37.1
Eylea™ 540 874 +61.9 +57.1
Kogenate™ 808 869 +7.5 +0.7
Mirena™ product family 594 742 +24.9 +9.8
Nexavar™ 571 661 +15.8 +6.1
Betaferon™ / Betaseron™ 629 634 +0.8 -9.2
YAZ™ / Yasmin™ / Yasminelle™ 570 538 -5.6 -5.0
Adalat™ 435 481 +10.6 +0.8
Aspirin™ Cardio 356 393 +10.4 +2.3
Glucobay™ 310 381 +22.9 +4.1
Avalox™ / Avelox™ 285 294 +3.2 -3.5
Stivarga™ 161 236 +46.6 +29.3
Xofigo™ 128 188 +46.9 +27.5
Total 6,861 8,189 +19.4 +12.0
Proportion of Pharmaceuticals sales 78% 80%
Fx & p adj. = currency- and portfolio-adjusted

Page 5
Drug Discovery - Biologics
Biologics
Research
Development

Elberfeld, Wuppertal

!  Monoclonal antibody
process development
and clinical manufacture
!  Batch-fed fermentation
!  Microbial fermentation
!  Antibody drug conjugate
production

Berkeley, California

!  Process development
and clinical manufacture
of hemophilia pipeline
!  Perfusion-based
fermentation
!  Production cell line and
MCB generation

Page 6
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 7
Biologics Landscape
•  Access to medicine
•  Health Care costs / reimbursement
•  Personalized medicine
-  From blockbuster to specifically patient designed biologics
•  Regional production set-up
•  Biosimilars
-  Opportunities
-  Regulation
-  Pricing
"  Consequences for industry
"  CoGs pressure
"  Fast product turnover in flexible multi-product plants
Page 8
Technology drivers for cell culture industry
•  High titer cell line

•  Chemically defined media

•  Fast and efficient PD

•  Robust production processes

•  Disposable technology

•  Closed systems operation

•  Modular plant

•  Ball-room plant

•  Continuous processing
Page 9
New products after replacement proteins
and mAbs
Complex and specifically designed molecules
-  Bispecific / multispecific
-  Sort half life molecules
-  ADCs / RIA
-  Cancer immunotherapies
-  Active site molecules
will need new / modified production technologies

Cell therapies

Renaissance of gene therapy?

Page 10
Status Quo in Commercial Manufacturing of
Biologics from Mammalian Cell Culture

•  85 – 90 % of all products are produces in fed-batch culture, most in


large facilities (“steel temples”) in a complex GMP infrastructure with
high degree of segregation and automation

•  10 – 15 % are produced in perfusion culture at high cell density by


cell retention using a similar GMP and segregation environment,
which represent a continuous upstream operation followed by
classical batch purification

Page 11
New Trends in Mammalian Production
•  Smaller fermenter volumes needed because of personalized medicine
and high production titers
•  Use of disposables instead of hard-piped equipment
•  Development of upstream and downstream completely or functionally
closed systems based mainly on disposables, which should reduce
environmental segregation and simplify facility design and operation
(ballroom plant concept)
•  Desire to produces several products in parallel (several products at a
time) in addition to the current campaign mode (one product at a time
per suite)
•  Regional production set-up needs facilities which are faster and
inexpensive to built

Page 12
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 13
Impact of Disposables on Protein Production
from Mammalian Cells
•  Complete production processing for biologics can be done in disposables, except:
-  Centrifugation
-  Chromatography skids
-  Large UF / DF

•  The following unit operations are available:


-  Mixing / holding / distribution of media and buffers
-  Seed expansion and production fermentation
-  Cell removal by depth filters
-  Chromatography columns
-  UF / DF/ virus filtration

However, different vendors with different hook-up and connection systems often result in
custom made (expensive) solutions

Page 14
Benefits of disposables
•  Simplification of processing by replacing highly controlled, hard-piped
equipment and utilities by single-use based, stand-alone equipment
connected through flexible tubings

Main advantages of single use:


-  plant design and construction: easier, faster, less expensive, less complex
-  construction in a „lab-like“ infrastructure possible
-  plant qualification/valiation: faster, less effort
-  plant operation: overall cost-efficient, main savings in utilities, water, steam,
CIP, SIP, etc., less depriciation
-  faster product change
-  lower COGs
-  fast and low risk transfer to different sites (emerging markets)

Page 15
General Limitations in Disposable Use
•  Different units from different vendors
-  Interchangeable connections missing
-  Lack of “standardization” among supplier

•  2nd supplier concept


•  Validation packages
-  Extractables, leachables
•  Quality oversight of disposable vendors

•  Regulatory support files


•  Routine production measures
(avoid human errors)
Page 16
Risk Mitigation Strategies Using Disposables
•  Work with vendors on improving quality standards
-  pressure testing of bags
-  in depth inspection for particles , etc.
-  in depth quality control audits
-  visualization of complete manufacturing process for dsiposables

•  Initiative by industry for joint vendors standardization

•  Implement 2nd supplier concept


-  easy for hold bags, filters, catridges, etc
-  difficult and time consuming for single use bioreactors

Page 17
Example of a Current Disposable-based
Fed-Batch Process
Cell Culture: ~ 35 days

Cell Sep:
Dead-End Filtration

Seed-Train Expansion
1 mL cryo-vial/Shaker Flasks Clarified
~ 14 days 200 L BIOSTAT® STR Harvest
~ 4 days
1000 L BIOSTAT® STR
14 - 18 days

Page 18
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 19
New Trends in Mammalian Production Plants

Upstream and downstream completely or functionally closed systems


based mainly on disposables

Produce several products in parallel (several products at a time) in


addition to the current campaign mode (one product at a time per suite)

Reduction in HVAC/seggregation requirements (ballroom concept)

Reduction/avoidance in hard-piping (SIP/CIP)

Faster and inexpensive to build

Based on ballroom design ?

Page 20
Single Use versus Steel based Plant Design
using the current Containment Concepts (1)
Same seggregation/airlocks/room classification/gowning/dedicated equipment and
personell concept as currently state of the art
Simplification of processing by replacing highly controlled, hard-piped equipment and
utilities by single-use based, stand-alone equipment connected through felxible
tubings

Main advantages of single use:


- plant design and construction: easier, faster, less expensive, less complex
- construction in a „lab-like“ infrastructure possible
- plant qualification/valiation: faster, less effort-
- plant operation: overall cost-efficient, main savings in utilities, water,
steam, CIP, SIP, etc., less depriciation
- faster product change
- lower COGs

Page 21
Single Use versus Steel based Plant Design
using the current Containment Concepts (2)
Main disadvantages of single use:
- limited in size of operation units (bags)
- labor intensive
- manual operations
- potential for operator failures
- dependency on bag vendor quality
- more waste - inactivation followed by incineration

Current new plants are often constructed as mixed mode plant combining
disposable and hard-piped processing steps

Page 22
Ballroom Plant Design Concept (1)

Represents innovative concept to enable parallel processing of different


products in the same low classification containment without upstream and
downstream seggregation

Concept addressed in the following paper:


Simon Chalk et.al., „Challenging the Cleanroom Paradigm for
Biopharmaceutical Manufacturing of Bulk Drug Substances“, BioPharm
International, Aug. 1, 2011.

Based on the key assumptions that technological advances including single use
sytems have continuously reduced the risk of environmental impact on
processing. Most steps can be securely performed closed or functionally closed.
The few remaining open processing steps have to be addressed independently
(i.e. portable laminar flow hood, isolator technology)

Page 23
Ballroom Plant Design Concept (2)
Basic thinking is that in a closed or functionally closed system, the process stream is
isolated from the environment

Remaining open operations (cell expansion, column packing, powder additions) have to be
addressed separetely, i.e. in small areas with classical containment set up

Potential breach of the closed system is the major risk, which has to be addressed:
- prove no contamination or cross-contamination
- intense microbial monitoring

Maintaining the closed system status has to be addresse by a risk based approach with
appropiate risk mitigation strategies considering each process step or operation

Page 24
Ballroom Plant Design Concept (3)

The following risks were addressed and mitigation strategies provided using
detailed failure mode and effects analysis tools:
- temporary breakable connections
- open manipulation in process stream
- charging raw materials during media or solution prep
- equipment prep
- cleaning or maintenance
- in-process sampling
- unexpected breach of a closed system element

There are indeed first facilities, which were designed, built and qualified
according to the concept of using risk-mitigated closed systems, which have
much lower containment /room classification requirements and are used for at
least clinical production
Page 25
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 26
Scheme for Continuous Perfusion Culture
with External Retention Device

Page 27
Production of recombinant Factor VIII
90 - 210kD
90kD portion
Heavy Chain
Heavy Chain
A1 A2
6 glycos. B
sites C2 C1 A3

80kD Light Chain B-region


19 glycosylation
sites

Structure 2332 Amino Acids


23 Cysteins

Product / year 150 g (1 Billion units)


Assays / batch 400+

Employees Manufacturing: 700


Quality Control: 300

WFI / year 20 Million Liters

Sales 2008 848 Million Euro

Page 28
Long Term Continuous Fermentation of
rec FVIII
100 100
Viability
Cell concentration [106 vc/mL]

10 Cell concentration
10

Viability [%]
production of unstable
protein
1 1
q/V = 10 /d

Dr. Konstantinov, Bayer Corp.,


Dechema 2002, Frankfurt

0 20 40 60 80 100 120 140


Time t [d]
Page 29
Perfusion Culture Features
•  High volumetric throughput (perfusion 1 – 15 fermenter volumes /
day)
•  Low residence time of product in the fermenter – low impact of “-ase”
activities, degradation
•  Physiological steady state conditions
-  Adjusted by specific perfusion rate
•  Small scale fermenters for commercial production

"  Product types


-  Low titer
-  Productivity correlated to cell growth
-  Fragile, fast degrading proteins
-  toxic
Page 30
Continuous Bio-Manufacturing

•  Currently hot topic in the industry

•  Advocated by regulatory authorities in the context of lean, low costs


and well controlled production

•  Well established for chemical compounds

•  Goal is to define a continuous process using perfusion technology


combined with continuous filtration and chromatography operations

Page 31
Differences between perfusion and
continuous processing

Perfusion Continuous Processing


•  Batch-wise harvest collection •  Completely continuous operation
without harvest collection
•  One or several harvest batches
are then combined to one DSP •  Cell removal is either done
batch
-  by the cell retention system or
-  by continuous depth filtration
•  Each product batch represents a using 2 filters per line (1 in
certain time interval of perfusion use, the other ready for use)
fermentation

Page 32
Mostly used cwell retention system:
ATF Perfusion System from Refine Technol.
(presented at the Biomanufacturing Summit, San Diego, 2013

Page 33
Features of the ATF System

•  Scalable
•  Operation in dual mode
-  One unit in use, the other prepared to be used

•  Perfusion rates of 1 – 3 fermenter volumes per day

•  Complete cell retention avoiding cell clarification step

•  Accumulation of dead cells and debris – cell bleed needed

Page 34
Options for Continuous Downstream
Processing

•  Continuous chromatography using a battery of small scale columns


operated sequentially
•  Continuous operation of membrane absorbers in bind / elute mode as
alternative to column chromatography
•  Continuous operation of membrane absorbers in flow-through mode

Status:
•  Currently established for continuous operaton up to capture step
Protein A for mAbs
•  Several units commercially available

Page 35
Options and Limits of Continuous
Processing
Pros Cons

•  Small investment in equipment •  Complex operation


and facility •  Technical feasibility not
•  Large scale GMP production in a established yet
lab-like environment •  Batch definition ?
•  Easy scale-up by adding same •  Potential product quality issues
size units by long term fermentation
•  Easy transfer to other sites •  Increased validation effort
(decentralized production)

Page 36
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 37
Comparison of a fed-batch facility with a
disposable facility using continuous processing
Cell culture pilot plant in Wuppertal Biofacility of the future
Purpose Purpose
•  Produce material for phase 3 clinical trials •  Production for market
Design Design
•  Stainless steel equipment •  100 % S.U. process equipment
•  Functionally closed processing •  Closed processing
•  Fed-batch fermentation •  Continuous processing
•  Operations are separated in different rooms •  Ballroom production
Building Concept Building Concept
•  5 levels •  2 levels
•  ~ 5000 m2 total area •  ~ 1200 m2 total area
•  ~ 1400 m2 cleanroom (class D and C) •  ~ 360 m2 cleanroom (class D and C)

Page 38
Design Principle:
“Ball Room” Production

Ball room includes:


•  All process units
•  All media and buffer
containers
•  All media and buffer
preparation tanks

… but does not include:


•  Seed lab
•  Bulk filling room (post viral
area)

page23
Layout 1st floor –
Production Level

Cleanroom classification
Black
Class E
Class D
Class C

Flows
Personnel
Material
Product
Waste

Pag 40
Agenda   New Bayer
  Status Quo of biologics manufacturing
  Disposables – benefits and limitations
  Facility of the Future / modern plant design
  Continuous processing
  Comparison of a standard fed-batch steel versus a continuous
processing based disposable facility
  Conclusion and outlook

Page 41
Summary and Conclusions

Single-use technologies are maturing allowing to produce most cell culture process steps in
disposables instead of hard-piped systems
•  Simpler operation in a lab-like environment
•  Issues: 2nd supplier, standardization and regulatory support files
Disposable based flexible facilities with functionally closed operation units are developing
into an alternative or supplement to the standard hard-piped based steel plants:
•  For lower volume products
•  Faster to build, smaller foot print, less complex in Engineering, simpler to qualify
and validate, lower in costs, easier to operate, lower COGs
•  Similar containment and seggregation concept compared to classical plants
Single –use technologies and continuous processing further reduce the footprint of flexible
ballroom plants with less or no seggregation and containment (facility-of-the-future):
•  Different products at a time, no seggregation upstream/downstream
•  Issue: How to handle steps, which still need (functionally) closed systems?
•  Regulatory acceptance/complexity of continuous operations

Page 42
Thank you!

Page 43
Content area and guides 5.92

5.00

0.00

1.30
Please restrict your content to this area

7.60

Page 44

You might also like