SlideShare a Scribd company logo
1 of 8
Download to read offline
whitepaper 
Biomanufacturing supply 
chain optimization 
Balancing risk against flexibility and just-in-time production 
© Bioproduction Group. All Rights Reserved.
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
Biomanufacturing SUPPLY CHAIN OPTIMIZATION 
Balancing risk against flexibility and just-in-time production 
INTR ODUCTION 
The biopharmaceutical supply chain presents unique challenges for supply chain 
planners and their supporting technologies, primarily because of the need for very 
high supply reliability. High costs and the life-preserving nature of biopharmaceutical 
products mean that planners must avoid outages and ensure supply continuity. Such 
imperatives make it difficult to meet the traditional goals for planners: flexible supply 
chains that carry minimal inventory and manufacture ‘just-in-time’ to meet demand. 
In this whitepaper, we discuss ways of modeling biomanufacturing networks that 
explicitly account for supply chain risks. Capturing the likelihood and effect of 
risks is a critical first step in determining how much inventory is needed to buffer 
against adverse events, and therefore the level of safety stocks that are required. 
In some cases, supply chain resilience and flexibility are not mutually exclusive, 
and with careful planning, a network can be constructed that optimizes both. 
© Bioproduction Group. All Rights Reserved. 1 
“My boss tells me that ‘no patient 
shall go without’. But what does 
that mean in reality? Nothing can 
be totally certain – we need to 
be able to quantify that risk.” 
Senior Supply Chain Planner, Large 
Biopharmaceutical Manufacturer 
Current Issues in the Biopharmaceutical Supply Chain 
Biopharmaceutical supply chains represent a unique challenge for planners because 
process variability, as well as contamination and other adverse events, need to be 
explicitly considered in supply planning. The threat of the known unknown – risks 
that cannot be accurately predicted – must also be incorporated into plans. Without 
characterization of these issues, traditional inventory models produce results that 
can appear to be optimal, but actually expose the business to unacceptably high 
levels of risk. We outline some of the key factors that must be considered below.
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
1. Cell Growth Times and Yield Variability 
Current cell lines are relatively slow-growing and require very tightly controlled 
growing conditions in terms of temperature, pH, and growth medium. Up to 30 
days is required to express sufficient material for a single batch. The effect of 
this long growth phase is that manufacturers have focused on increasingly large 
batch sizes and multiple serial production trains to ensure sufficient production 
quantities. It also means the time required to react to adverse conditions is long. 
In addition, current cell lines are extremely sensitive to growing conditions and 
contamination by other cells such as bacteria or viruses. Cell growth is exponential 
(since in a well-designed process it is only constrained by the doubling time of the 
cell), but this growth pattern means that small variations in growth are magnified 
over and over again. The result is highly variable output quantity and quality of 30% 
or more (Shah, 2004). This variability is also highly auto-correlated, so that many 
successive batches in a manufacturing campaign may be affected. This is a particular 
problem for the biopharmaceutical industry where a small number of batches 
are typically produced, and the time to react to manufacturing issues is long. 
2. Contamination and Reject Rates 
Biological-based manufacturing also requires exceptionally stringent levels of 
cleanliness since cells grow in conditions well suited for many other bacteria 
and viruses. Even with completely clean equipment, biological contamination 
can be introduced from other sources including the feed stock (media), 
water, operators, detection mechanisms, air handling systems, etc. 
Once biological contamination is present, it can be extremely difficult to remove. 
Typical biological protection mechanisms such as a protective thin film formation 
(seen, for example, in slime) or spores (a reproductive mechanism designed 
for survival in unfavorable conditions) are very difficult to chemically treat and 
usually need to be physically removed from pipes (e.g. with an abrasive slurry). 
The focus for biopharmaceutical manufacturers has therefore been to reduce bio-burden 
(biological contamination) as much as possible using mechanisms like HTST 
pasteurization of media, and to introduce purification steps that will filter out any 
contamination. Specific types of bio-burden will render conditions unsuitable for the 
fermentation process to continue; testing systems are designed to detect this and abort 
the batch as early as possible. Even with such systems in place, contamination events 
are common – as seen in the 2009 contamination event in Genzyme’s Allston facility. 
3. Testing and Quality Assurance 
Product and process-based testing puts a considerable burden on supply 
chain cycle times and throughput. Hundreds of tests are done on each batch 
as it progresses through production to ensure efficacy and safety of the final 
product. A number of tests are done in-line and are relatively quick to complete, 
while some (e.g. bioassays) can take up to 2 months to give results. 
© Bioproduction Group. All Rights Reserved. 2 
“What’s especially troubling 
about the Allston facility event 
is that it could happen to any 
biomanufacturer, at any time. 
Genzyme was just unlucky.” 
Engineer, Cell Culture Operations Group
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
Target release cycle 
time of 70 days. 
Average cycle time 
85 days, standard 
deviation 30 days 
No target release 
cycle time. Average 
time 60 days, 
standard deviation 
of 70 days 
100-120 
80% of batches 
take less than 
120 days 
120-140 
140-160 
180-200 
160-180 
RELEASE TIME (days) 
0-20 
20-40 
40-60 
60-80 
80-100 
220-240 
220-240 
240+ 
0.5 
0.45 
0.4 
0.35 
0.3 
0.25 
0.2 
0.15 
0.1 
0.05 
0 
PERCENT OF VALUE (%) 
Some batches 
require more 
than 1/2 a year 
Company 1 
Company 2 
© Bioproduction Group. All Rights Reserved. 3 
FIGURE 1: 
QA/QC Release Cycle Times 
Overall biotechnology supply chain cycle times are currently between 41 days and 3 
years; the value-added time (time when the material is actively produced) is between 
0.3% and 5% of this (Shah, 2005). The single greatest cause for these very long cycle 
times is the need to test batches at each stage of production. It is not uncommon 
for batches to be delayed for 6 months or longer, if an irregularity is found. 
In aggregate, the prolonged uncertainty of testing results and consequent delays 
of production result in large stock levels of intermediate materials; stock levels 
typically range from 30% to 90% of annual demand quantity (Shah, 2005), an 
order of magnitude higher than traditional pharmaceutical products manufactured 
using chemical processes. This is problematic because these intermediate 
products can tie up millions (or hundreds of millions) of dollars in inventory. 
4. Process-based Regulation 
Since quality testing of the final product cannot provide a complete assurance 
of product efficacy and safety, regulatory authorities around the world have 
adopted a process-based approach to regulation. Agencies such as the 
Food and Drug Administration (FDA) require licensure of the entire process 
surrounding the production of biotechnology product. This means that any 
changes to plant or process design must be certified in each country in which 
it is sold, a process which may take up to 3 years for major projects. 
Process-based regulation has an important effect on innovation in the industry, 
since it provides a large disincentive for companies to change their processes. 
Many companies have pursued a ‘license and leave’ approach to drug manufacture 
where no significant changes to a plant are permitted after the facility has been 
licensed to produce a product. This means that technological changes that 
may have a dramatic impact on manufacturing cost, speed or even product 
safety are not pursued since they would require re-licensure. Examples can be 
seen in the slow adoption of continuous fermentation and purification systems, 
disposables, and in-line testing systems. This is unlike other high-tech industries 
such as the semiconductor industry, where technological innovation provides 
competitive advantage and the only disincentive to innovate is cost-based.
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
2 MONTH OUTAGE 
3 MO. 6 MO. 3 MO. 2 MO. 1 MO. 1 MO. 
RESTART 
CAMPAIGN JAN 2009 JUL 2009 JAN 2010 JUL 2010 
4 MONTH OUTAGE 
6 MO. 3 MO. 
3 MO. + 3 MO. 1 MO. 1 MO. 
PRODUCT 1 
PRODUCT 2 
PRODUCT 3 
CONTAMINATION 
REORDER CAMPAIGNS, 
USE BACKUP FACILITY JAN 2009 JUL 2009 JAN 2010 JUL 2010 
BACKUP FACILITY B 
6 MONTH OUTAGE 
6 MO. 
2 MO. 
© Bioproduction Group. All Rights Reserved. 4 
FIGURE 2: 
Recovery strategy for 3 different 
con tamination severities 
5. Process Design for Manufacturability 
Most production facilities in existence are currently multi-product capable in the 
sense that the basic site infrastructure allows for the manufacture of a range of 
products. At least in principle, this means that new drugs can be produced in existing 
facilities. In practice, however, it is quite difficult for facilities to change production 
modes because of the sensitivity of culture growth and purification to hundreds 
of production parameters. Almost all existing biopharmaceutical production is 
‘product specific,’ that is, the facility is designed around the specific drug compound 
being produced in order to maximize the titer and yield of that cell. This leads to 
a highly irregular supply chain design in firms with more than one product. 
One of the growing issues in process design is the need to match the capacity 
of each of the major production steps. Fermentation, or cell growth, has 
increased titers more quickly than the ability of purification processes to 
manage the additional material produced. This evolution of titer has created 
bottlenecks in recovery operations and the need to make significant changes 
in downstream processing to accommodate these higher titers. 
Case Study: Contamination of a Bulk Biomanufacturing Facility 
One of the critical issues in supply chain planning is the need to model the effect 
of adverse events like contamination. Bioproduction Group was asked by a large 
biomanufacturer with a number of facilities to evaluate the likelihood and impact 
of a contamination event in their facilities similar to Genzyme’s 2008/2009 events. 
The team determined early on that it was not possible to ignore or ‘average out’ 
the above issues and instead constructed a high fidelity, high accuracy facility-network 
model using Bio-G’s patented Simulation System technology. Using data 
from plant managers and engineers, the model was designed to accurately depict 
the production process and constraints of all the facilities in the network. 
2 MO. BACKUP FACILITY A 
2 MO. BACKUP FACILITY B 
1 MO. + 3 MO. 1 MO. 1 MO. 
REORDER CAMPAIGNS, 
USE TWO BACKUP 
JAN 2009 JUL 2009 JAN 2010 JUL 2010 FACILITIES 
Through discussions with subject matter experts and key members of the 
scheduling department, Bio-G determined several possible reactions to a facility 
contamination to prevent product shortages. “When our client says ‘no patient shall 
go without treatment,’ it is not corporate rhetoric: they mean it,” says Principal David 
Zhang. “There is an intelligent way to utilize available network resources to keep 
production flowing while avoiding unnecessary disruption to the supply chain.”
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
600 
400 
© Bioproduction Group. All Rights Reserved. 5 
FIGURE 3: 
INVENTORY RISK PROFILES ANALYZED 
BY THE SIMULATION SYSTEM 
Bio-G’s strategic model was used to evaluate a number of possible contamination 
scenarios, depending on the severity of the contamination event. Low severity (2 
month recovery time), medium severity (4 month recovery) and high severity (6 
month recovery) were each considered to determine the range of effects on the 
supply chain network. The diagram above shows the range of possible responses 
to adverse events, depending on the event’s severity. Backup facilities were used in 
more severe contamination cases to ensure stockout would not occur. The Simulation 
System aided planners in quickly evaluating the best response in each case. 
2 MONTH OUTAGE 
4 MONTH OUTAGE 
6 MONTH OUTAGE (BACKUP) 
BASELINE (NO OUTAGE) 
6 MONTH (NO BACKUP) 
RELEASED INVENTORY LEVEL (KG) 
INVENTORY LEVELS 
TIME (HOURS) 
500 
300 
100 
0 2000 4000 6000 8000 10000 12000 14000 
200 
0 
Bio-G’s strategic model provided a decision support tool that allowed for the 
quantification and mitigation of risk for strategic decision-making. The results 
showed that stockouts would occur with some of the response strategies being 
considered by the manufacturer, leading to policies and accurate inventory 
setting to prevent these from occurring. The tool was then used by the client to 
determine the best responses amongst all possible scenarios, increasing confidence 
that the manufacturer had sufficient agility to respond to adverse events. 
Moving towards Lean and Just-In-Time 
One of the most critical questions facing current supply chain planners today is 
how to optimize their available capacity over the short and medium term (1-5 
years) while also considering the impact of many of the key variables mentioned 
above. New product versions, for example, bring higher titers which move 
bottlenecks in the network. New technologies – disposables, HTST, modular skids, 
etc. – are being adopted, but their impact on the supply chain is not yet clear. 
While SAP and other traditional inventory planning toolsets have been applied 
to this problem, they have been found in practice to grossly underestimate the 
level of variability in the network and overestimate the feasibility of supply plans. 
This is due to the fact that such toolsets are inherently deterministic, using a 
single number to represent each parameter in the scenario rather than allowing a 
range of possible values. Such systems are also inherently ‘push’-based, and are 
inflexible to changing supply and demand conditions (Hopp & Spearman, 2000). 
Bio-G’s approach to this problem is different. Instead of relying on single-point 
estimates, the software gives planners the ability to generate and 
explore ranges of values. This means the software produces results that 
explicitly account for variability, risk and its effect in the supply chain – 
analysis that is desperately needed in the biopharmaceutical industry. 
“Bio-G’s unique software 
plays a role in keeping U.S. 
biomanufacturing capacity 
globally competitive as well as 
ensuring lower costs for patients.” 
Industry Analyst, Biopharmaceutical/ 
Pharmaceutical Sector
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
40 
35 
30 
25 
20 
15 
10 
© Bioproduction Group. All Rights Reserved. 6 
FIGURE 4: 
New protein-free formulation 
at 60% lower observed 
titers at the pilot scale 
FIGURE 5: 
INTRODUCING A NEW PRODUCT 
VARIENT DOUBLES the number 
of SKU’S IN THE SUPPLY CHAIN 
Case Study: Examining the effect of a new product 
variant on supply chain reliability 
Bioproduction Group was asked by a large biopharmaceutical manufacturer to 
implement a toolset to allow them more robust analysis of the effect of a new 
product introduction. The new product used media that was not derived from 
animal-based products, meaning a lower risk of supply outage due to raw material 
contamination. However, the new product was exhibiting significantly lower harvest 
titers than expected, leading them to re-evaluate manufacturing capacity. 
In addition, the biomanufacturer’s supply chain was highly fragmented by the 
need for a number of product variants for dosage, capping type and market. The 
introduction of this additional new product variant required the manufacturer 
to smoothly transition from the existing product to the new, while allowing 
countries to purchase the old product variant until the new was approved. Such 
an approach caused an explosion of product variants that the supply chain was 
required to handle, with an uncertain effect on inventory levels and risk. 
CAPPING TYPE 
PRODUCT DOSAGE 
FINAL MARKET 
NEW PRODUCT 
(NO ANIMAL-DERIVED 
PEPTONES) 
= 60 PRODUCT 
VARIANTS 
+ 
+ 
+ 
TITER (grams/liter) 
1.0-1.2 
1.2-1.4 
1.4-1.6 
1.6-1.8 
1.8-2.0 
2.0-2.2 
2.2-2.4 
2.4-2.6 
2.6-2.8 
2.8-3.0 
3.0-3.2 
5 
0 
LIKELIHOOD (%) 
Existing Product 
New Protien Free Product
BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 
2010 2011 2012 
LAUNCH I N 2 MA RK ETS LAU NCH IN 4 MARKETS LAU NCH IN ALL MARKE TS 
IN VE NTO RY FALLS 
BELOW TARG ETS, 
MANUFACTURING 
AT CA PACIT Y 
© Bioproduction Group. All Rights Reserved. 7 
FEE DBACK 
Please provide your feedback at http://www. 
zoomerang.com/Survey/WEB22AYVM3VLFC 
FURTHER READING 
Shah N (2004) Pharmaceutical supply 
chains: key issues and strategies 
for optimization. Computers and 
Chemical Engineering 28: 929-941 
Hopp and Spearman (2000) 
Factory Physics, McGraw-Hill 
MORE INFORMATION 
BIOPRODUCTION GROUP 
CONTACT@BIO-G.COM 
WWW .BIO-G.COM 
Bioproduction Group’s approach was to build a high speed supply chain 
simulator that could evaluate the capacity of each of the manufacturing facilities 
used by the manufacturer, along with hundreds of rules and constraints on 
the manufacturing process. The result was a model that was both a highly 
accurate representation of the manufacturing network, and one that could 
easily be altered by planners to perform sophisticated what-if analysis. 
The approach allowed the biomanufacturer to confirm that a world-wide introduction of 
the new product posed an unacceptably high risk to their supply chain. Using data from 
over 10,000 years of simulated time and hundreds of automatically generated scenarios, 
the Simulation System suggested a staggered product introduction that focused on 
markets that were likely to approve the product quickly, while leaving countries like 
Japan and Canada (with long and highly variable approval times) for a later date. 
The figure above shows the effect of implementing the staggered production 
introduction strategy vs. the original strategy where the firm launched simultaneously 
in all markets. The difference between the two strategies is not visible to the supply 
chain organization until nearly 9 months after launch when inventory starts falling 
below target levels and manufacturing reacts by shifting to higher capacity. But 
this capacity increase is not sufficient to bring inventory levels back to target values 
for nearly 2 years after launch, posing an unacceptable risk to the business. 
In this graph, the use of a simple yet powerful aggregated metric ‘Supply Chain Risk’ 
based on a Value at Risk or VaR calculation, allows the business to see with one 
view real comparisons between supply scenarios. The ability to perform this analysis 
allowed the business to bring concrete analytics to their supply chains, providing a 
‘virtual supply chain’ that analysts could quickly alter to evaluate what-if scenarios. 
Conclusions 
Biomanufacturing Supply Chains represent unique challenges for planners: the need for 
high service levels in a heavily constrained industry with long lead times, high levels of 
regulation, and increasing focus on inventory and cost reduction. Bioproduction Group’s 
Simulation System provides the means to model such complex supply chains in an easy-to- 
use, powerful framework. The patented technology provides a 100-fold increase 
in speed over existing simulation toolsets, allowing planners to perform sophisticated 
design-of-experiment and what-if optimization easily and quickly. This approach has 
been used to quantify millions of dollars in direct cost savings to biomanufacturers, 
while aiding risk avoidance and increasing supply reliability to customers. 
“The ability to rapidly and 
confidently model and analyze 
a complex supply chain could 
have enormous impact on a firms’ 
ability to remain competitive and 
to continuously improve high 
technology production processes.” 
National Science Foundation Evaluation 
of Bio-G Technology, 2010 
Staggered Product Introduction Strategy 
SUPPLY CHAIN RISK 
ORIGINAL STRATEGY STAGGERED INTRODUCTION S TRATEGY 
FIGURE 6: 
VAR CALCULATION FOR THE 
MANUFACTURER’s original supply 
chain strategy, contrasted 
against a staggered product 
introduction strategy

More Related Content

What's hot

Scale up fermentation process (H.M.Moavia Atique)
Scale up fermentation process (H.M.Moavia Atique)Scale up fermentation process (H.M.Moavia Atique)
Scale up fermentation process (H.M.Moavia Atique)moavia Atiq
 
Beyond Bioprocessing 4.0: The Convergence of IT, OT and Processing Technolog...
Beyond Bioprocessing 4.0:  The Convergence of IT, OT and Processing Technolog...Beyond Bioprocessing 4.0:  The Convergence of IT, OT and Processing Technolog...
Beyond Bioprocessing 4.0: The Convergence of IT, OT and Processing Technolog...Merck Life Sciences
 
Article: On the Cusp of Curing Disease
Article: On the Cusp of Curing DiseaseArticle: On the Cusp of Curing Disease
Article: On the Cusp of Curing DiseaseMilliporeSigma
 
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...Challenges using Multiple Single-use Systems: Functionality versus Extractabl...
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...Merck Life Sciences
 
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination Risks
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination RisksUpstream Viral Safety: A Holistic Approach to Mitigating Contamination Risks
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination RisksMilliporeSigma
 
Strategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlStrategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlMerck Life Sciences
 
Strategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlStrategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlMilliporeSigma
 
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...MilliporeSigma
 
The Emprove® Program: Introduction of New Portfolio Additions
The Emprove® Program: Introduction of New Portfolio AdditionsThe Emprove® Program: Introduction of New Portfolio Additions
The Emprove® Program: Introduction of New Portfolio AdditionsMerck Life Sciences
 
Sartorius Solutions for Cell & Gene based Therapies
Sartorius Solutions for Cell & Gene based TherapiesSartorius Solutions for Cell & Gene based Therapies
Sartorius Solutions for Cell & Gene based TherapiesRaymond Goyco, III
 
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...MilliporeSigma
 
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
 
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter Validation
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter ValidationEU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter Validation
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter ValidationMilliporeSigma
 
Quality by Design Principles Applied to Sterilizing Filtration by Michael Payne
Quality by Design Principles Applied to Sterilizing Filtration by Michael PayneQuality by Design Principles Applied to Sterilizing Filtration by Michael Payne
Quality by Design Principles Applied to Sterilizing Filtration by Michael PayneMilliporeSigma
 
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...Merck Life Sciences
 
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...MilliporeSigma
 
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...MilliporeSigma
 

What's hot (18)

Scale up fermentation process (H.M.Moavia Atique)
Scale up fermentation process (H.M.Moavia Atique)Scale up fermentation process (H.M.Moavia Atique)
Scale up fermentation process (H.M.Moavia Atique)
 
Beyond Bioprocessing 4.0: The Convergence of IT, OT and Processing Technolog...
Beyond Bioprocessing 4.0:  The Convergence of IT, OT and Processing Technolog...Beyond Bioprocessing 4.0:  The Convergence of IT, OT and Processing Technolog...
Beyond Bioprocessing 4.0: The Convergence of IT, OT and Processing Technolog...
 
Article: On the Cusp of Curing Disease
Article: On the Cusp of Curing DiseaseArticle: On the Cusp of Curing Disease
Article: On the Cusp of Curing Disease
 
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...Challenges using Multiple Single-use Systems: Functionality versus Extractabl...
Challenges using Multiple Single-use Systems: Functionality versus Extractabl...
 
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination Risks
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination RisksUpstream Viral Safety: A Holistic Approach to Mitigating Contamination Risks
Upstream Viral Safety: A Holistic Approach to Mitigating Contamination Risks
 
Strategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlStrategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic Control
 
14-6-Monge
14-6-Monge14-6-Monge
14-6-Monge
 
Strategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic ControlStrategies for Effective Bioburden and Aseptic Control
Strategies for Effective Bioburden and Aseptic Control
 
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...
Upcoming USP 665 - Level of Characterization of Single-Use Systems Today and ...
 
The Emprove® Program: Introduction of New Portfolio Additions
The Emprove® Program: Introduction of New Portfolio AdditionsThe Emprove® Program: Introduction of New Portfolio Additions
The Emprove® Program: Introduction of New Portfolio Additions
 
Sartorius Solutions for Cell & Gene based Therapies
Sartorius Solutions for Cell & Gene based TherapiesSartorius Solutions for Cell & Gene based Therapies
Sartorius Solutions for Cell & Gene based Therapies
 
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...
Employing Innovative Platform Manufacturing and Biosafety Testing for your Ge...
 
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
 
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter Validation
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter ValidationEU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter Validation
EU GMP Annex 1 Draft: Implications on Sterilizing Grade Filter Validation
 
Quality by Design Principles Applied to Sterilizing Filtration by Michael Payne
Quality by Design Principles Applied to Sterilizing Filtration by Michael PayneQuality by Design Principles Applied to Sterilizing Filtration by Michael Payne
Quality by Design Principles Applied to Sterilizing Filtration by Michael Payne
 
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...
The Role of BPOG Extractables Data in the Effective Adoption of Single-Use Sy...
 
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...
EU GMP Annex 1 – Implications on Filtration and Single Use Technology by Soma...
 
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...
Identifying Appropriate-Quality Pharmaceutical Raw Materials in an Evolving R...
 

Similar to Biomanufacturing supply chain optimization

Raw Material White Paper
Raw Material White PaperRaw Material White Paper
Raw Material White PaperMason Williams
 
Raw Material White Paper
Raw Material White PaperRaw Material White Paper
Raw Material White PaperMason Williams
 
Bio g - ManufacturingIintelligence webinar GBX
Bio g - ManufacturingIintelligence webinar GBXBio g - ManufacturingIintelligence webinar GBX
Bio g - ManufacturingIintelligence webinar GBXGBX Summits
 
Modelling
ModellingModelling
Modellingskarri
 
Scale up of industrial microbial processes
Scale up of industrial microbial processes Scale up of industrial microbial processes
Scale up of industrial microbial processes Shubham Vats
 
industrial microbial culture (1).pdf
industrial microbial culture (1).pdfindustrial microbial culture (1).pdf
industrial microbial culture (1).pdfRupajitBhattacharjee1
 
Bio g defense in depth presentation
Bio g defense in depth presentationBio g defense in depth presentation
Bio g defense in depth presentationGBX Summits
 
The Future of Bioprocessing – What you need to know.
The Future of Bioprocessing – What you need to know.The Future of Bioprocessing – What you need to know.
The Future of Bioprocessing – What you need to know.Merck Life Sciences
 
Nichols-Scott-Kite-Pharma-2023.pdf
Nichols-Scott-Kite-Pharma-2023.pdfNichols-Scott-Kite-Pharma-2023.pdf
Nichols-Scott-Kite-Pharma-2023.pdfRichardShi22
 
Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing PnuVax
 
Cleanrooms for Vaccines.pdf
Cleanrooms for Vaccines.pdfCleanrooms for Vaccines.pdf
Cleanrooms for Vaccines.pdfSadafAli74
 
Integrated utilization of high-throughput bioreactors & high-throughput analy...
Integrated utilization of high-throughput bioreactors & high-throughput analy...Integrated utilization of high-throughput bioreactors & high-throughput analy...
Integrated utilization of high-throughput bioreactors & high-throughput analy...KBI Biopharma
 
The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingThe Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingMutyala Naidu G
 
IVT Presentation Batch vs Continuous - 45min_REV3
IVT Presentation Batch vs Continuous - 45min_REV3IVT Presentation Batch vs Continuous - 45min_REV3
IVT Presentation Batch vs Continuous - 45min_REV3Eric Sipe
 
SMi Group's Pharmaceutical Microbiology USA 2017 conference
SMi Group's Pharmaceutical Microbiology USA 2017 conferenceSMi Group's Pharmaceutical Microbiology USA 2017 conference
SMi Group's Pharmaceutical Microbiology USA 2017 conferenceDale Butler
 
Industrial Bioprocessing webinar.pptx
Industrial Bioprocessing webinar.pptxIndustrial Bioprocessing webinar.pptx
Industrial Bioprocessing webinar.pptxShubham Chinchulkar
 

Similar to Biomanufacturing supply chain optimization (20)

Raw Material White Paper
Raw Material White PaperRaw Material White Paper
Raw Material White Paper
 
Raw Material White Paper
Raw Material White PaperRaw Material White Paper
Raw Material White Paper
 
Bio g - ManufacturingIintelligence webinar GBX
Bio g - ManufacturingIintelligence webinar GBXBio g - ManufacturingIintelligence webinar GBX
Bio g - ManufacturingIintelligence webinar GBX
 
Modelling
ModellingModelling
Modelling
 
Scale up of industrial microbial processes
Scale up of industrial microbial processes Scale up of industrial microbial processes
Scale up of industrial microbial processes
 
industrial microbial culture (1).pdf
industrial microbial culture (1).pdfindustrial microbial culture (1).pdf
industrial microbial culture (1).pdf
 
PBIO Investor Presentation
PBIO Investor PresentationPBIO Investor Presentation
PBIO Investor Presentation
 
Bio g defense in depth presentation
Bio g defense in depth presentationBio g defense in depth presentation
Bio g defense in depth presentation
 
Pbio 8 8-2013
Pbio 8 8-2013Pbio 8 8-2013
Pbio 8 8-2013
 
The Future of Bioprocessing – What you need to know.
The Future of Bioprocessing – What you need to know.The Future of Bioprocessing – What you need to know.
The Future of Bioprocessing – What you need to know.
 
Nichols-Scott-Kite-Pharma-2023.pdf
Nichols-Scott-Kite-Pharma-2023.pdfNichols-Scott-Kite-Pharma-2023.pdf
Nichols-Scott-Kite-Pharma-2023.pdf
 
Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing
 
PMG Newsletter (Volume 2. Issue 30).pdf
PMG Newsletter (Volume 2. Issue 30).pdfPMG Newsletter (Volume 2. Issue 30).pdf
PMG Newsletter (Volume 2. Issue 30).pdf
 
Campaign production aspects
Campaign production aspectsCampaign production aspects
Campaign production aspects
 
Cleanrooms for Vaccines.pdf
Cleanrooms for Vaccines.pdfCleanrooms for Vaccines.pdf
Cleanrooms for Vaccines.pdf
 
Integrated utilization of high-throughput bioreactors & high-throughput analy...
Integrated utilization of high-throughput bioreactors & high-throughput analy...Integrated utilization of high-throughput bioreactors & high-throughput analy...
Integrated utilization of high-throughput bioreactors & high-throughput analy...
 
The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingThe Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
 
IVT Presentation Batch vs Continuous - 45min_REV3
IVT Presentation Batch vs Continuous - 45min_REV3IVT Presentation Batch vs Continuous - 45min_REV3
IVT Presentation Batch vs Continuous - 45min_REV3
 
SMi Group's Pharmaceutical Microbiology USA 2017 conference
SMi Group's Pharmaceutical Microbiology USA 2017 conferenceSMi Group's Pharmaceutical Microbiology USA 2017 conference
SMi Group's Pharmaceutical Microbiology USA 2017 conference
 
Industrial Bioprocessing webinar.pptx
Industrial Bioprocessing webinar.pptxIndustrial Bioprocessing webinar.pptx
Industrial Bioprocessing webinar.pptx
 

More from GBX Summits

A Healthy Dose of Chips R1
A Healthy Dose of Chips  R1A Healthy Dose of Chips  R1
A Healthy Dose of Chips R1GBX Summits
 
Address The Innovation Imperative!
Address The Innovation Imperative!Address The Innovation Imperative!
Address The Innovation Imperative!GBX Summits
 
Maintenance and calibrations
Maintenance and calibrationsMaintenance and calibrations
Maintenance and calibrationsGBX Summits
 
Innovation in biopharmaceutical production
Innovation in biopharmaceutical productionInnovation in biopharmaceutical production
Innovation in biopharmaceutical productionGBX Summits
 
Bio g strategic capacity assessment
Bio g strategic capacity assessmentBio g strategic capacity assessment
Bio g strategic capacity assessmentGBX Summits
 
Capacity planning in biomanufacturing
Capacity planning in biomanufacturingCapacity planning in biomanufacturing
Capacity planning in biomanufacturingGBX Summits
 
Biomanufacturing flexibility
Biomanufacturing flexibilityBiomanufacturing flexibility
Biomanufacturing flexibilityGBX Summits
 
Biomanufacturing debottlenecking and optimization
Biomanufacturing debottlenecking and optimizationBiomanufacturing debottlenecking and optimization
Biomanufacturing debottlenecking and optimizationGBX Summits
 
Real time scheduling
Real time schedulingReal time scheduling
Real time schedulingGBX Summits
 
Real time modeling system
Real time modeling systemReal time modeling system
Real time modeling systemGBX Summits
 
Titer capacity analysis
Titer capacity analysisTiter capacity analysis
Titer capacity analysisGBX Summits
 
Retrofitting a new process to an existing facility
Retrofitting a new process to an existing facilityRetrofitting a new process to an existing facility
Retrofitting a new process to an existing facilityGBX Summits
 
Prioritizing waste reduction projects
Prioritizing waste reduction projectsPrioritizing waste reduction projects
Prioritizing waste reduction projectsGBX Summits
 
Optimizing bulk campaign lengths
Optimizing bulk campaign lengthsOptimizing bulk campaign lengths
Optimizing bulk campaign lengthsGBX Summits
 
Increasing labor effectiveness
Increasing labor effectivenessIncreasing labor effectiveness
Increasing labor effectivenessGBX Summits
 
Evaluating the effect of raw materials shortages
Evaluating the effect of raw materials shortagesEvaluating the effect of raw materials shortages
Evaluating the effect of raw materials shortagesGBX Summits
 
Evaluating perfusion technologies
Evaluating perfusion technologiesEvaluating perfusion technologies
Evaluating perfusion technologiesGBX Summits
 
Enabling higher process titers at a manufacturing facility
Enabling higher process titers at a manufacturing facilityEnabling higher process titers at a manufacturing facility
Enabling higher process titers at a manufacturing facilityGBX Summits
 
Debottlenecking a bulk manufacturing plant
Debottlenecking a bulk manufacturing plantDebottlenecking a bulk manufacturing plant
Debottlenecking a bulk manufacturing plantGBX Summits
 

More from GBX Summits (20)

A Healthy Dose of Chips R1
A Healthy Dose of Chips  R1A Healthy Dose of Chips  R1
A Healthy Dose of Chips R1
 
Address The Innovation Imperative!
Address The Innovation Imperative!Address The Innovation Imperative!
Address The Innovation Imperative!
 
Maintenance and calibrations
Maintenance and calibrationsMaintenance and calibrations
Maintenance and calibrations
 
Innovation in biopharmaceutical production
Innovation in biopharmaceutical productionInnovation in biopharmaceutical production
Innovation in biopharmaceutical production
 
Bio g strategic capacity assessment
Bio g strategic capacity assessmentBio g strategic capacity assessment
Bio g strategic capacity assessment
 
Capacity planning in biomanufacturing
Capacity planning in biomanufacturingCapacity planning in biomanufacturing
Capacity planning in biomanufacturing
 
Biomanufacturing flexibility
Biomanufacturing flexibilityBiomanufacturing flexibility
Biomanufacturing flexibility
 
Biomanufacturing debottlenecking and optimization
Biomanufacturing debottlenecking and optimizationBiomanufacturing debottlenecking and optimization
Biomanufacturing debottlenecking and optimization
 
Real time scheduling
Real time schedulingReal time scheduling
Real time scheduling
 
Real time modeling system
Real time modeling systemReal time modeling system
Real time modeling system
 
Crosswalk
CrosswalkCrosswalk
Crosswalk
 
Titer capacity analysis
Titer capacity analysisTiter capacity analysis
Titer capacity analysis
 
Retrofitting a new process to an existing facility
Retrofitting a new process to an existing facilityRetrofitting a new process to an existing facility
Retrofitting a new process to an existing facility
 
Prioritizing waste reduction projects
Prioritizing waste reduction projectsPrioritizing waste reduction projects
Prioritizing waste reduction projects
 
Optimizing bulk campaign lengths
Optimizing bulk campaign lengthsOptimizing bulk campaign lengths
Optimizing bulk campaign lengths
 
Increasing labor effectiveness
Increasing labor effectivenessIncreasing labor effectiveness
Increasing labor effectiveness
 
Evaluating the effect of raw materials shortages
Evaluating the effect of raw materials shortagesEvaluating the effect of raw materials shortages
Evaluating the effect of raw materials shortages
 
Evaluating perfusion technologies
Evaluating perfusion technologiesEvaluating perfusion technologies
Evaluating perfusion technologies
 
Enabling higher process titers at a manufacturing facility
Enabling higher process titers at a manufacturing facilityEnabling higher process titers at a manufacturing facility
Enabling higher process titers at a manufacturing facility
 
Debottlenecking a bulk manufacturing plant
Debottlenecking a bulk manufacturing plantDebottlenecking a bulk manufacturing plant
Debottlenecking a bulk manufacturing plant
 

Biomanufacturing supply chain optimization

  • 1. whitepaper Biomanufacturing supply chain optimization Balancing risk against flexibility and just-in-time production © Bioproduction Group. All Rights Reserved.
  • 2. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION Biomanufacturing SUPPLY CHAIN OPTIMIZATION Balancing risk against flexibility and just-in-time production INTR ODUCTION The biopharmaceutical supply chain presents unique challenges for supply chain planners and their supporting technologies, primarily because of the need for very high supply reliability. High costs and the life-preserving nature of biopharmaceutical products mean that planners must avoid outages and ensure supply continuity. Such imperatives make it difficult to meet the traditional goals for planners: flexible supply chains that carry minimal inventory and manufacture ‘just-in-time’ to meet demand. In this whitepaper, we discuss ways of modeling biomanufacturing networks that explicitly account for supply chain risks. Capturing the likelihood and effect of risks is a critical first step in determining how much inventory is needed to buffer against adverse events, and therefore the level of safety stocks that are required. In some cases, supply chain resilience and flexibility are not mutually exclusive, and with careful planning, a network can be constructed that optimizes both. © Bioproduction Group. All Rights Reserved. 1 “My boss tells me that ‘no patient shall go without’. But what does that mean in reality? Nothing can be totally certain – we need to be able to quantify that risk.” Senior Supply Chain Planner, Large Biopharmaceutical Manufacturer Current Issues in the Biopharmaceutical Supply Chain Biopharmaceutical supply chains represent a unique challenge for planners because process variability, as well as contamination and other adverse events, need to be explicitly considered in supply planning. The threat of the known unknown – risks that cannot be accurately predicted – must also be incorporated into plans. Without characterization of these issues, traditional inventory models produce results that can appear to be optimal, but actually expose the business to unacceptably high levels of risk. We outline some of the key factors that must be considered below.
  • 3. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 1. Cell Growth Times and Yield Variability Current cell lines are relatively slow-growing and require very tightly controlled growing conditions in terms of temperature, pH, and growth medium. Up to 30 days is required to express sufficient material for a single batch. The effect of this long growth phase is that manufacturers have focused on increasingly large batch sizes and multiple serial production trains to ensure sufficient production quantities. It also means the time required to react to adverse conditions is long. In addition, current cell lines are extremely sensitive to growing conditions and contamination by other cells such as bacteria or viruses. Cell growth is exponential (since in a well-designed process it is only constrained by the doubling time of the cell), but this growth pattern means that small variations in growth are magnified over and over again. The result is highly variable output quantity and quality of 30% or more (Shah, 2004). This variability is also highly auto-correlated, so that many successive batches in a manufacturing campaign may be affected. This is a particular problem for the biopharmaceutical industry where a small number of batches are typically produced, and the time to react to manufacturing issues is long. 2. Contamination and Reject Rates Biological-based manufacturing also requires exceptionally stringent levels of cleanliness since cells grow in conditions well suited for many other bacteria and viruses. Even with completely clean equipment, biological contamination can be introduced from other sources including the feed stock (media), water, operators, detection mechanisms, air handling systems, etc. Once biological contamination is present, it can be extremely difficult to remove. Typical biological protection mechanisms such as a protective thin film formation (seen, for example, in slime) or spores (a reproductive mechanism designed for survival in unfavorable conditions) are very difficult to chemically treat and usually need to be physically removed from pipes (e.g. with an abrasive slurry). The focus for biopharmaceutical manufacturers has therefore been to reduce bio-burden (biological contamination) as much as possible using mechanisms like HTST pasteurization of media, and to introduce purification steps that will filter out any contamination. Specific types of bio-burden will render conditions unsuitable for the fermentation process to continue; testing systems are designed to detect this and abort the batch as early as possible. Even with such systems in place, contamination events are common – as seen in the 2009 contamination event in Genzyme’s Allston facility. 3. Testing and Quality Assurance Product and process-based testing puts a considerable burden on supply chain cycle times and throughput. Hundreds of tests are done on each batch as it progresses through production to ensure efficacy and safety of the final product. A number of tests are done in-line and are relatively quick to complete, while some (e.g. bioassays) can take up to 2 months to give results. © Bioproduction Group. All Rights Reserved. 2 “What’s especially troubling about the Allston facility event is that it could happen to any biomanufacturer, at any time. Genzyme was just unlucky.” Engineer, Cell Culture Operations Group
  • 4. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION Target release cycle time of 70 days. Average cycle time 85 days, standard deviation 30 days No target release cycle time. Average time 60 days, standard deviation of 70 days 100-120 80% of batches take less than 120 days 120-140 140-160 180-200 160-180 RELEASE TIME (days) 0-20 20-40 40-60 60-80 80-100 220-240 220-240 240+ 0.5 0.45 0.4 0.35 0.3 0.25 0.2 0.15 0.1 0.05 0 PERCENT OF VALUE (%) Some batches require more than 1/2 a year Company 1 Company 2 © Bioproduction Group. All Rights Reserved. 3 FIGURE 1: QA/QC Release Cycle Times Overall biotechnology supply chain cycle times are currently between 41 days and 3 years; the value-added time (time when the material is actively produced) is between 0.3% and 5% of this (Shah, 2005). The single greatest cause for these very long cycle times is the need to test batches at each stage of production. It is not uncommon for batches to be delayed for 6 months or longer, if an irregularity is found. In aggregate, the prolonged uncertainty of testing results and consequent delays of production result in large stock levels of intermediate materials; stock levels typically range from 30% to 90% of annual demand quantity (Shah, 2005), an order of magnitude higher than traditional pharmaceutical products manufactured using chemical processes. This is problematic because these intermediate products can tie up millions (or hundreds of millions) of dollars in inventory. 4. Process-based Regulation Since quality testing of the final product cannot provide a complete assurance of product efficacy and safety, regulatory authorities around the world have adopted a process-based approach to regulation. Agencies such as the Food and Drug Administration (FDA) require licensure of the entire process surrounding the production of biotechnology product. This means that any changes to plant or process design must be certified in each country in which it is sold, a process which may take up to 3 years for major projects. Process-based regulation has an important effect on innovation in the industry, since it provides a large disincentive for companies to change their processes. Many companies have pursued a ‘license and leave’ approach to drug manufacture where no significant changes to a plant are permitted after the facility has been licensed to produce a product. This means that technological changes that may have a dramatic impact on manufacturing cost, speed or even product safety are not pursued since they would require re-licensure. Examples can be seen in the slow adoption of continuous fermentation and purification systems, disposables, and in-line testing systems. This is unlike other high-tech industries such as the semiconductor industry, where technological innovation provides competitive advantage and the only disincentive to innovate is cost-based.
  • 5. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 2 MONTH OUTAGE 3 MO. 6 MO. 3 MO. 2 MO. 1 MO. 1 MO. RESTART CAMPAIGN JAN 2009 JUL 2009 JAN 2010 JUL 2010 4 MONTH OUTAGE 6 MO. 3 MO. 3 MO. + 3 MO. 1 MO. 1 MO. PRODUCT 1 PRODUCT 2 PRODUCT 3 CONTAMINATION REORDER CAMPAIGNS, USE BACKUP FACILITY JAN 2009 JUL 2009 JAN 2010 JUL 2010 BACKUP FACILITY B 6 MONTH OUTAGE 6 MO. 2 MO. © Bioproduction Group. All Rights Reserved. 4 FIGURE 2: Recovery strategy for 3 different con tamination severities 5. Process Design for Manufacturability Most production facilities in existence are currently multi-product capable in the sense that the basic site infrastructure allows for the manufacture of a range of products. At least in principle, this means that new drugs can be produced in existing facilities. In practice, however, it is quite difficult for facilities to change production modes because of the sensitivity of culture growth and purification to hundreds of production parameters. Almost all existing biopharmaceutical production is ‘product specific,’ that is, the facility is designed around the specific drug compound being produced in order to maximize the titer and yield of that cell. This leads to a highly irregular supply chain design in firms with more than one product. One of the growing issues in process design is the need to match the capacity of each of the major production steps. Fermentation, or cell growth, has increased titers more quickly than the ability of purification processes to manage the additional material produced. This evolution of titer has created bottlenecks in recovery operations and the need to make significant changes in downstream processing to accommodate these higher titers. Case Study: Contamination of a Bulk Biomanufacturing Facility One of the critical issues in supply chain planning is the need to model the effect of adverse events like contamination. Bioproduction Group was asked by a large biomanufacturer with a number of facilities to evaluate the likelihood and impact of a contamination event in their facilities similar to Genzyme’s 2008/2009 events. The team determined early on that it was not possible to ignore or ‘average out’ the above issues and instead constructed a high fidelity, high accuracy facility-network model using Bio-G’s patented Simulation System technology. Using data from plant managers and engineers, the model was designed to accurately depict the production process and constraints of all the facilities in the network. 2 MO. BACKUP FACILITY A 2 MO. BACKUP FACILITY B 1 MO. + 3 MO. 1 MO. 1 MO. REORDER CAMPAIGNS, USE TWO BACKUP JAN 2009 JUL 2009 JAN 2010 JUL 2010 FACILITIES Through discussions with subject matter experts and key members of the scheduling department, Bio-G determined several possible reactions to a facility contamination to prevent product shortages. “When our client says ‘no patient shall go without treatment,’ it is not corporate rhetoric: they mean it,” says Principal David Zhang. “There is an intelligent way to utilize available network resources to keep production flowing while avoiding unnecessary disruption to the supply chain.”
  • 6. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 600 400 © Bioproduction Group. All Rights Reserved. 5 FIGURE 3: INVENTORY RISK PROFILES ANALYZED BY THE SIMULATION SYSTEM Bio-G’s strategic model was used to evaluate a number of possible contamination scenarios, depending on the severity of the contamination event. Low severity (2 month recovery time), medium severity (4 month recovery) and high severity (6 month recovery) were each considered to determine the range of effects on the supply chain network. The diagram above shows the range of possible responses to adverse events, depending on the event’s severity. Backup facilities were used in more severe contamination cases to ensure stockout would not occur. The Simulation System aided planners in quickly evaluating the best response in each case. 2 MONTH OUTAGE 4 MONTH OUTAGE 6 MONTH OUTAGE (BACKUP) BASELINE (NO OUTAGE) 6 MONTH (NO BACKUP) RELEASED INVENTORY LEVEL (KG) INVENTORY LEVELS TIME (HOURS) 500 300 100 0 2000 4000 6000 8000 10000 12000 14000 200 0 Bio-G’s strategic model provided a decision support tool that allowed for the quantification and mitigation of risk for strategic decision-making. The results showed that stockouts would occur with some of the response strategies being considered by the manufacturer, leading to policies and accurate inventory setting to prevent these from occurring. The tool was then used by the client to determine the best responses amongst all possible scenarios, increasing confidence that the manufacturer had sufficient agility to respond to adverse events. Moving towards Lean and Just-In-Time One of the most critical questions facing current supply chain planners today is how to optimize their available capacity over the short and medium term (1-5 years) while also considering the impact of many of the key variables mentioned above. New product versions, for example, bring higher titers which move bottlenecks in the network. New technologies – disposables, HTST, modular skids, etc. – are being adopted, but their impact on the supply chain is not yet clear. While SAP and other traditional inventory planning toolsets have been applied to this problem, they have been found in practice to grossly underestimate the level of variability in the network and overestimate the feasibility of supply plans. This is due to the fact that such toolsets are inherently deterministic, using a single number to represent each parameter in the scenario rather than allowing a range of possible values. Such systems are also inherently ‘push’-based, and are inflexible to changing supply and demand conditions (Hopp & Spearman, 2000). Bio-G’s approach to this problem is different. Instead of relying on single-point estimates, the software gives planners the ability to generate and explore ranges of values. This means the software produces results that explicitly account for variability, risk and its effect in the supply chain – analysis that is desperately needed in the biopharmaceutical industry. “Bio-G’s unique software plays a role in keeping U.S. biomanufacturing capacity globally competitive as well as ensuring lower costs for patients.” Industry Analyst, Biopharmaceutical/ Pharmaceutical Sector
  • 7. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 40 35 30 25 20 15 10 © Bioproduction Group. All Rights Reserved. 6 FIGURE 4: New protein-free formulation at 60% lower observed titers at the pilot scale FIGURE 5: INTRODUCING A NEW PRODUCT VARIENT DOUBLES the number of SKU’S IN THE SUPPLY CHAIN Case Study: Examining the effect of a new product variant on supply chain reliability Bioproduction Group was asked by a large biopharmaceutical manufacturer to implement a toolset to allow them more robust analysis of the effect of a new product introduction. The new product used media that was not derived from animal-based products, meaning a lower risk of supply outage due to raw material contamination. However, the new product was exhibiting significantly lower harvest titers than expected, leading them to re-evaluate manufacturing capacity. In addition, the biomanufacturer’s supply chain was highly fragmented by the need for a number of product variants for dosage, capping type and market. The introduction of this additional new product variant required the manufacturer to smoothly transition from the existing product to the new, while allowing countries to purchase the old product variant until the new was approved. Such an approach caused an explosion of product variants that the supply chain was required to handle, with an uncertain effect on inventory levels and risk. CAPPING TYPE PRODUCT DOSAGE FINAL MARKET NEW PRODUCT (NO ANIMAL-DERIVED PEPTONES) = 60 PRODUCT VARIANTS + + + TITER (grams/liter) 1.0-1.2 1.2-1.4 1.4-1.6 1.6-1.8 1.8-2.0 2.0-2.2 2.2-2.4 2.4-2.6 2.6-2.8 2.8-3.0 3.0-3.2 5 0 LIKELIHOOD (%) Existing Product New Protien Free Product
  • 8. BIOMANUFACTURING SUPPLY CHAIN OPTIMIZATION 2010 2011 2012 LAUNCH I N 2 MA RK ETS LAU NCH IN 4 MARKETS LAU NCH IN ALL MARKE TS IN VE NTO RY FALLS BELOW TARG ETS, MANUFACTURING AT CA PACIT Y © Bioproduction Group. All Rights Reserved. 7 FEE DBACK Please provide your feedback at http://www. zoomerang.com/Survey/WEB22AYVM3VLFC FURTHER READING Shah N (2004) Pharmaceutical supply chains: key issues and strategies for optimization. Computers and Chemical Engineering 28: 929-941 Hopp and Spearman (2000) Factory Physics, McGraw-Hill MORE INFORMATION BIOPRODUCTION GROUP CONTACT@BIO-G.COM WWW .BIO-G.COM Bioproduction Group’s approach was to build a high speed supply chain simulator that could evaluate the capacity of each of the manufacturing facilities used by the manufacturer, along with hundreds of rules and constraints on the manufacturing process. The result was a model that was both a highly accurate representation of the manufacturing network, and one that could easily be altered by planners to perform sophisticated what-if analysis. The approach allowed the biomanufacturer to confirm that a world-wide introduction of the new product posed an unacceptably high risk to their supply chain. Using data from over 10,000 years of simulated time and hundreds of automatically generated scenarios, the Simulation System suggested a staggered product introduction that focused on markets that were likely to approve the product quickly, while leaving countries like Japan and Canada (with long and highly variable approval times) for a later date. The figure above shows the effect of implementing the staggered production introduction strategy vs. the original strategy where the firm launched simultaneously in all markets. The difference between the two strategies is not visible to the supply chain organization until nearly 9 months after launch when inventory starts falling below target levels and manufacturing reacts by shifting to higher capacity. But this capacity increase is not sufficient to bring inventory levels back to target values for nearly 2 years after launch, posing an unacceptable risk to the business. In this graph, the use of a simple yet powerful aggregated metric ‘Supply Chain Risk’ based on a Value at Risk or VaR calculation, allows the business to see with one view real comparisons between supply scenarios. The ability to perform this analysis allowed the business to bring concrete analytics to their supply chains, providing a ‘virtual supply chain’ that analysts could quickly alter to evaluate what-if scenarios. Conclusions Biomanufacturing Supply Chains represent unique challenges for planners: the need for high service levels in a heavily constrained industry with long lead times, high levels of regulation, and increasing focus on inventory and cost reduction. Bioproduction Group’s Simulation System provides the means to model such complex supply chains in an easy-to- use, powerful framework. The patented technology provides a 100-fold increase in speed over existing simulation toolsets, allowing planners to perform sophisticated design-of-experiment and what-if optimization easily and quickly. This approach has been used to quantify millions of dollars in direct cost savings to biomanufacturers, while aiding risk avoidance and increasing supply reliability to customers. “The ability to rapidly and confidently model and analyze a complex supply chain could have enormous impact on a firms’ ability to remain competitive and to continuously improve high technology production processes.” National Science Foundation Evaluation of Bio-G Technology, 2010 Staggered Product Introduction Strategy SUPPLY CHAIN RISK ORIGINAL STRATEGY STAGGERED INTRODUCTION S TRATEGY FIGURE 6: VAR CALCULATION FOR THE MANUFACTURER’s original supply chain strategy, contrasted against a staggered product introduction strategy