You are on page 1of 10

Biochemical Engineering Journal 49 (2010) 435–444

Contents lists available at ScienceDirect

Biochemical Engineering Journal


journal homepage: www.elsevier.com/locate/bej

Kaizen for improvement of rapid protein production for early reagent


protein quantities
Beth Junker
PO Box 2000, RY 810-126, Merck Research Laboratories, Rahway, NJ, United States

a r t i c l e i n f o a b s t r a c t

Article history: A kaizen improvement effort was undertaken focusing on the workflows of an in-house workgroup whose
Received 16 November 2009 aim was the rapid production of small amounts of protein reagents. The kaizen’s goal was to reduce
Received in revised form 6 February 2010 process lead times to accommodate more deliveries per year and more complex protein types. Business
Accepted 9 February 2010
requirements were established, and baseline performance was evaluated against projected needs. Then
potential areas for change were identified, selected, and implemented. Improvements were quantified
based on established metrics as well as customer input. Overall in-house group capacity was raised 11%
Keywords:
from 1.1 to 1.2 deliveries per person per week at the same time that the percentage of non-platform
Kaizen
Improvement
(more difficult) requests was increased to nearly 50% from under 10%. In-house group lead times from
Reagent protein request to shipping for platform (less difficult) purification deliveries were improved by 30% from 11.1
Transient transfection to 7.7 days.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction from a cell cultivation might be stored at 4 ◦ C for extended times


awaiting purification personnel availability). Often, timing conflicts
A rapid protein production group is a critical element of the with pipeline development projects led to inconsistent lead times.
biological research program portfolio at a company or other In addition, success rates were lower for some steps which required
institution. The impact of such a group to the early phases of further development and closer adherence to procedures.
the basic research process is challenging to measure since it is Consistent lead times were highly desired since deliveries
multi-dimensional: (1) increasing product candidate probability- typically were on the critical path for lead identification and, partic-
of-success (POS) by providing necessary protein reagents for testing ularly, lead optimization studies, or for developing immunoassays
efficacy, (2) decreasing early phase product candidate lead iden- to analyze samples from these studies. Certain basic research
tification and, especially, lead optimization cycle times, and (3) experiments required reservations and advance planning (e.g.,
providing related proteins for additional in vitro or in vivo testing allocating animals and/or personnel). Consequently, a dedicated
to resolve past technical impasse points. A typical antibody pro- rapid protein production group was formed to address these
gram might require 2–3 g of the target molecule over 4–6 deliveries, requirements using internal and outsourced resources. The in-
plus about 6–8 different similar reagent antibodies (1–3 deliver- house (internal) portion of this workgroup was formed in early
ies each, depending on titer) for immunoassay development for a 2007. It became fully functional with dedicated on-board cell
total of ∼15 deliveries. The basic research scientists receiving these cultivation/purification staff and capital equipment by mid-2007,
deliveries were very anxious to be viewed as collaborators and not including completion of the necessary training and technology
customers. Consequently, they were willing to alter their actions transfer activities.
to realize productivity improvements. Transparency of communi- The rapid protein production group became the single source
cation both ways was considered critical to build trust. for growth and purification of reagents, covering both in-house
Initially, rapid protein production tasks were spread across three and outsourcing for 10–100 mg and 100–1000 mg protein quan-
distinct departments and up to 15 individuals from many different tities. An immediate gap was identified since the business process
managerial groups. Specific requests were filled as time permit- for rapid reagent protein requests was not widely known, clear,
ted, often based only loosely on prioritized needs. This set up led or consistent. A single point of contact was selected to coordi-
to delivery delays and handoff mismatches (e.g., harvested broth nate requests and handle scheduling; then, key stakeholders and
sponsors were identified. Ad hoc request emails were replaced
with a manual, but structured, request submission process cen-
tered on a user request form. A decision then was made whether
E-mail address: beth junker@merck.com. to conduct the work in-house or at a vendor to manage over-

1369-703X/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.bej.2010.02.007
436 B. Junker / Biochemical Engineering Journal 49 (2010) 435–444

flow, typically based on technology requirements and timing. antibodies for ELISA assays) more predictably prior to advancing
Request status was also tracked manually. Regular joint meet- projects from basic research into development. Basic research cus-
ings with one point of contact for each customer group were held tomer goals were to obtain protein reagents for further in vivo
to prioritize requests, usually within rather than across customer studies, typically comparing several proteins at one time. Con-
areas. sequently, some basic research requests encompassed multiple
It soon became evident that new outsourcing relationships proteins and, since subsequent experiments could not commence
were required to improve consistency, specifically to replace low until all proteins were delivered, efficient parallel as well as serial
performing vendors where supply gaps existed. Eventually, the processing was important.
vendor list was expanded to encompass most request types, Customers were almost always the same as suppliers. Depend-
unless vendor use was restricted by a material disclosure agree- ing on the project, researchers supplied DNA, cells, or culture broth,
ment. Some technical process improvements were implemented and available information about protein purity requirements and
to improve outsourcing success and simplify technology transfer. other desired protein testing. Individual tasks depended on the spe-
For example, a revised plasmid fermentation process to obtain cific project, but at a high level, the general process was to generate
transient transfection DNA was developed using a dissolved oxy- broth, purify broth, assay product, and then deliver protein (Fig. 1).
gen (DO)-based, rather than carbon evolution rate (CER)-based, Deliveries included the protein (often in a concentrated form), an
feeding strategy. This change translated into a widely transfer- associated analysis sheet, and a brief description of key process
able process to a greater number of vendors since no off-gas mass steps.
spectrometer analyzer (an expensive piece of equipment) was Two broad categories of deliveries were established based on
required. the amount of protein product (primarily considering typical IgG
General methods demonstrated by other organizations to delivery amounts): 10–100 mg and 100–1000 mg. Typically <10 mg
increase task efficiency include centralization of the func- requests were handled by a separate workgroup. In 2007, delivery
tion, adoption of platform technology, institution of rational- quantities ranged from 0.5 to 2 g, generated from broth volumes
ized/standardized workflows, and information sharing. Past efforts ranging from 70 mL to 25 L and broth titers from 3 mg/L to 4.5 g/L.
to improve productivity for rapid delivery applications using lean This wide range of titers and volumes necessitated purification pro-
techniques have effectively utilized these concepts. In one exam- cessing flexibility and efficient workflows.
ple, Pfizer switched from end-driven metrics, such as number The variety of protein products handled expanded as customers
of batches and number of deviations, to methodologies utilizing interacted with the in-house group and recognized that requests for
capacity models based on retrospective data and timelines [1]. other proteins besides IgGs were possible. These included: IgG anti-
Full team meetings permitted everyone to evaluate the entire pro- bodies (types 1–4), IgM antibodies, N-terminal flag-tag proteins,
cess. Precursor steps (i.e., equipment and material readiness) were human Fc receptor proteins (FcRn), His-tag proteins, Fc-fusion pro-
determined to cause the most variability and occupy the most teins, and other conjugate proteins. The associated purification
unexpected time. Subsequently, lead time was improved by imple- “soups” also varied, including animal/insect cell cultivation broth,
menting measures to ensure that each project started on time and animal sera, ascites fluid, and bacterial/yeast cell pastes.
project load variations (range of 5–10 projects per week) were There were two major types of cell culture platform requests: (1)
reduced by implementing workload leveling. In a second exam- Stable cell line (i.e., largely hybridoma with some CHO) cultivations
ple, Glaxo-Smith-Kline (GSK) employed value stream mapping to were passaged until stable growth was obtained. For hybridomas,
remove or rearrange several steps to reduce cycle time for aliquot- this required a T-flask seed train consisting of four stages start-
ting solid samples to assay-ready plates for screening [2]. In a ing from a frozen cryovial. Production cultivations employed a
third example, analogous streamlining efforts were recently pub- batch process using commercial media supplemented with 10 vol%
lished by the Swedish Royal Institute of Technology for 1–10 mg serum. Typically, half the culture volume was inoculated then
scale protein deliveries [3]. Although some efficiency approaches diluted with an equal volume of fresh media at day 2 or 3, followed
were implemented similarly to those used by GSK, it was primar- by harvest at day 7. (2) Transient transfections were conducted
ily the adoption of heavy automation that increased output from using HEK-293 cells (passaged using a rolling seed split twice per
10 to 288 recombinant proteins per week. These examples also week), EBNA (Epstein Barr Virus Nuclear Antigen) transient expres-
demonstrated that one of the most effective strategies to reduce sion technology, a low-cost and efficient transfection reagent (PEI,
lead times is cellular manufacturing, for example establishing a polyethyleneimine), purified heavy and light chain plasmid DNA,
dedicated group within an organization to perform start to finish and fetal bovine serum (FBS) to final concentration of 5 vol% [5].
processing [4]. HEK cells were grown on a proprietary media, using a 7-day, fed-
A kaizen is a focused effort to improve productivity and/or work batch process with feeds on days 3 and 5. Plasmid DNA was grown
environment, typically conducted within a workgroup over a con- in E. coli and purified separately by external groups, typically in
centrated period of a few days. The project goal for this kaizen quantities sufficient for repeat transient transfections. Generally,
effort was to reduce process lead times for rapid protein deliveries transient transfection deliveries were executed in 4–5 versus 8–10
by the in-house dedicated workgroup, both to accommodate more weeks for the alternative stable CHO cell pools, using a single plas-
deliveries per year and more complex types of proteins. mid DNA construct. In 2007, about half of the product candidate
and related molecule deliveries and most of the assay reagent
deliveries were executable by combining multiple disposable shake
2. Key background elements flask cultivations; the remaining deliveries were conducted at the
20 L disposable Wave bioreactor scale (GE/Wave Biotech, Somerset,
Suppliers, inputs, process steps, outputs, and customers NJ).
(SIPOC) were identified. Customers of the rapid protein delivery There were several types of purification platforms and method-
group included analytical scientists in the drug metabolism and ologies: (1) Protein A-based platform purification for IgG and
immunoassay departments, basic research scientists in the thera- Fc-fusion proteins was the most common, and consisted of centrifu-
peutic protein and vaccine departments, and, to a smaller extent, gation, concentration, Protein A chromatography, concentration,
basic research customers in fundamental research areas such as buffer exchange, and delivery. Often, protein-containing sera and
structural biology. Analytical customer goals were to obtain pro- ascites proceeded directly to the Protein A step. Owing to expense,
tein reagents required for assay development (e.g., anti-protein Protein A columns typically were cleaned and reused several
B. Junker / Biochemical Engineering Journal 49 (2010) 435–444 437

Fig. 1. Current state value stream map for purification of transient transfection broth containing a monoclonal antibody being purified by Protein A. All times reported in
minutes. Bold font used to indicate revised times based on implementation of straightforward Kaizen improvements. C/T, cycle time; C/O, changeover time; W/T, wait time.

dozen times. (2) Non-platform but standard methods generally production study for the kaizen activity since most in-house group
were based on a Ni-nitrilotriacetic acid (NTA) protocol aimed at members were thoroughly familiar with it. [Outsourcing work was
purifying affinity tags (e.g., his-tag) or polyclonal IgGs from sera. not emphasized in this kaizen effort.] There was also substantial
These methods consisted of centrifugation, sterile filtration, Ni-NTA subject matter expert (SME) experience and generally good agree-
chromatography, concentration, buffer exchange, and delivery. ment with task touch times/elapsed times, resource needs, and
Disposable resins were used to avoid cleaning steps. (3) Several success rates. The entire process from request through delivery
non-platform methods were implemented which required sub- was broken into a few high level steps: Requests were received,
stantial development but, after repeated execution, were expected a decision was made regarding in-house or outsourcing, requests
to become platform methods. For example, IgM purification was were added to the production schedule, and then starting materials
accomplished using a POROS 50A column (which unfortunately did and protocols transferred by the customer. Next followed plasmid
not work for all IgMs) or GE2-mercaptopyridine chromatography. DNA fermentation, plasmid DNA purification, transient transfec-
(4) In 2008, completely new procedures such as highly challenging tion, protein purification, analysis, and finally delivery. Of these
purification of soluble and insoluble his-tag intracellular proteins steps, it soon became evident that protein purification was the
from E. coli were undertaken with only a 30–40% success rate. clear and obvious bottleneck, necessitating deeper analysis. Thus,
Typical steps were thawing of frozen cell paste, homogenization protein purification was selected to be the scope of the kaizen.
or microfluidization, then centrifugation. If the protein was solu- The kaizen approach undertaken was to develop requirements
ble, then further steps included a pellet wash, centrifugation pellet (i.e., critical to quality attributes), quantify baseline performance,
solubilization, Ni-NTA chromatography, endotoxin removal, and a develop a process flow diagram, identify potential areas for
concentration/buffer exchange. If the protein was insoluble, then improvement, then implement and quantify the new performance
refolding was attempted. For many deliveries of types (1)–(4), level.
impurities were assessed by size exclusion chromatography (SEC)
and subsequent size purification was undertaken using ultrafiltra-
tion. 3.1. Business and customer requirements

Requirements were developed through a variety of methods:


3. Kaizen approach informal feedback, semi-annual oversight meetings with stake-
holders and customer representatives, and an annual customer
Transient transfection and its associated Protein A-based purifi- survey. [This kaizen was conducted in 2008 based on 2007 sur-
cation for an IgG antibody was selected as the typical protein vey results.] The over-riding (highest weighted) concerns were
438 B. Junker / Biochemical Engineering Journal 49 (2010) 435–444

workflow effectiveness and capacity for rapid protein production. • Step 3. Supernatant collected, then pumped through disposable
Key minor (lower weighted) concerns were prioritization across microfilter assembly to separate remaining cells. Filtrate col-
diverse customer groups, alignment with portfolio prioritization lected in a sterile, disposable bag.
shifts, transparent scheduling, appropriate split between in-house • Step 4. Filtrate concentrated using previously assembled and
and outsourcing, customer education about the range of group cleaned ultrafiltration unit. Waste collected into plastic drum.
capabilities, expansion of routine capabilities (i.e., broadening of Retentate collected in disposable bottle, along with two buffer
the protein types that could be delivered), turnaround time par- rinses of ultrafilter to recover hold up. Samples taken and ana-
ticularly for emergency requests, cross-functional team work, and lyzed for protein mass balance. [Closure varied from 50% to 95%.]
communication of interim status during deliveries. • Step 5. Retentate microfiltered into another sterile, disposable
bottle and stored cold (typically over a weekend) to await purifi-
3.2. Kaizen goals cation personnel availability.
• Step 6. Chromatography system cleaned and lines primed. Col-
Consequently, the specific kaizen goals were to improve (1) umn equilibrated, loaded, washed, and eluted to obtain low
lead time from request to delivery and (2) deliveries per month pH Protein A product. System cleaned and column regenerated.
per person, thus satisfying the requirements of more projects Chromatography steps operated in manual mode but equipment
per year and more complex protein types in a timely manner. capable of automated operation.
• Step 7. Base added to obtain quenched Protein A product (QPAP)
Key supporting goals were to optimize and hopefully reduce step
times and resources associated with the cell culture, purification, at neutral pH. [About 10% of the time, this step is critical, unpre-
and analytical stages. The strategy adopted was to investigate dictable, and often troublesome owing to precipitation.] QPAP
and reduce inefficiencies (waste) in areas such as reprocessing microfiltered, collected into sterile, disposable bottle, then sam-
(owing to contamination or unacceptable purification outcomes), pled/assayed for protein. If target protein concentration or buffer
changeovers, set up requirements, and clean up procedures. The required, QPAP is further concentrated and/or buffer exchanged.
expected business benefits were primarily (1) decreased discovery [This required calculations.]
• Step 8. Samples taken and analyzed for endotoxin (if protein
cycle time which improved basic research lead identification and
lead optimization times, and (2) resource efficiency which avoided to be used for animal studies), purity, final concentration, or
assignment of additional staff or outsourcing funds to this support Western blot (if cannot conduct Protein A assay). Analysis sheet
activity. Although the first benefit was difficult to quantify, the sec- completed and e-mailed to customer. Delivery sheet completed
ond benefit was quantifiable using metrics for capacity and lead which described process steps, and recorded final protein con-
time. centration, purity, and endotoxin (if required). Shipping sheet
Demand forecast data from key customers were used to estab- (i.e., bill of laden per state Department of Transportation reg-
lish the desired future state. The forecast was first adjusted for ulations) completed indicating handling requirements (such as
planned outsourcing to obtain the in-house group delivery tar- temperature). Product, shipping sheet, and delivery sheet trans-
get, which was both significantly higher than the prior year and ferred to site shipping office. [Almost all proteins delivered
included substantially more complicated deliveries. This target required off-site shipment.]
next was matched to available manpower by incorporating protein
complexity ratings for the different delivery categories (Table 1). 3.4. Identification of wastes and improvement solutions
Depending upon request volume and complexity, performance
data indicated that about 62.8 ± 8.5 total deliveries/quarter were After completion of the process flow diagram, the team
achievable with 37.2 ± 5.6 deliveries/quarter (about 60%) con- evaluated it for the presence of the eight common wastes:
ducted by the in-house group. Using an estimated available work transportation (global movement of material), inventory (excess
time of 40 weeks and 3 purification staff (Table 1), this translated storage, accumulated work), motion (excess in-process move-
into a customer demand rate of about 3.7 deliveries per week and ment), waiting (lags in information, inspection, or approval),
a maximum average time per delivery to meet this demand (takt over-production (making more or doing more than is neces-
time) of 0.8 day. sary), over-processing (self-created or unnecessary work including
over-inspection), defects (quality flaws), and underutilized tal-
3.3. Process flow diagram ent (idle or misused manpower or capabilities). To better derive
probable root causes of these wastes (Table 2), the wastes were
A next level, current state, flow diagram of the bottleneck pro- categorized using the common root cause and effect (“5M, 1E”)
tein purification steps after delivery of cell culture broth through elements of man (organization, training, assignments), methods
to receipt of isolated protein by the customer was constructed (procedures, policies), materials (process inputs), machines (equip-
(Fig. 1). To facilitate construction, designated team members ment), measurements (data, assays, analytical standards), and
(SMEs) arrived prepared with a draft flow chart for their assigned environment (fixed constraints). Next, the team brainstormed
steps and time estimates for a 20 L scale cell culture purification. and prioritized (based on their impact on the kaizen goals and
These cycle (elapsed) time (C/T), changeover (clean up) time (C/O), effort to implement) ideas for solutions to reduce the identified
and wait time (W/T) estimates were added to obtain the lead (total) waste.
time (L/T) (Fig. 1). Identified wastes and proposed higher priority solutions are
Key process steps are listed below (Fig. 1): listed for each element below:
Man:
• Step 1. Sample taken of cell cultivation and analyzed. Broth Waste: The waste of talent was evidenced by an imbalance
delivered for purification in sterile, disposable bag. Batch sheet between assigned fermentation and purification staff as well as
template printed and information recorded as steps executed. an overall lack of cross-training. Solutions: Staff were reallocated
• Step 2. Broth subdivided into centrifuge bottles and from fermentation to purification (fermentation staff moved from
weighed/reweighed for balanced volumes. Weights recorded 40% in 2007 to 12% in 2008) especially as more fermentation
and bottles centrifuged (sub steps: spin, decant supernatant, tasks were outsourced. Complete process/analytical cross-training
clean bottles). to balance workloads was implemented, taking advantage of less
B. Junker / Biochemical Engineering Journal 49 (2010) 435–444 439

Table 1
Example workload projection for 2008 and gap calculation for purification tasks.

Attribute Platform Non-platform Total Gap

10–100 mg scale 100–1000 mg scale Standard method Method development

Prior year (2007)


Projected group goal 50 25 0 0 75 N/A
(5 staff plus $1 MM outsourcing
budget)
Actual total delivery 146 50 0 0 196 N/A

Current year (2008)


Projected group goal 100 75 0 0 175 N/A
(4 staff plus $1 MM outsourcing
budget)
Customer projections at end of prior 113 38 30 5 186 11
year (2007)
Customer projections 6 months later 131 36 52 12 231 56
(mid-2008)
Outsourcing projection 47 15 6 0 68 N/A
(based on $1 MM budget)
Excluding 39 fermentations for
plasmid DNA
Projected in-house delivery target 84 21 46 40 163 N/A
Projected deliveries/staff-week 2.5 1 0.7 0.3 4.5 N/A
(based on purification task complexity
factor)
Projected required staff-weeks 34 21 74 40 169 49
Projected required purification staff 0.85 0.53 1.8 1.0 4.2 1.2

Notes: (1) Platform: Protein A IgG antibody purification of CHO, HEK or hybridoma broth. (2) Non-platform: standard method, IgM purification with metal affinity chromatog-
raphy. (3) Non-platform: method development, E. coli or other protein source requiring scale up or further development of a basic research laboratory method. (4) Estimate:
40–120 staff-weeks/year; 3 staff for purification and 1 staff for fermentation available.

busy times owing to uneven current state process scheduling, to 20%. In addition, Protein A loading/elution flowrates, binding con-
increase throughput by 50%. These changes permitted increased ditions, and column reuse conditions were optimized to decrease
responsiveness to customer demand and created a shared sense of projected cycle time by 10%
process ownership among in-house group members. Waste: Over-processing also was evident owing to cell cul-
ture broth protein titer variations. Solutions: Appropriately sized
Measurement:
columns and filters were selected based on titer, requiring less
Waste: Over-processing waste became evident in the amount of
time and buffer usage. Disposable centrifugal ultrafiltration devices
analytical data being collected, all of which was not always required
provided flexibility for processing varying QPAP volumes. When
based on customer feedback. Solutions: Standard practice became
implemented along with optimized procedures and conditions for
to measure titer (via HPLC) or aggregation (via size exclusion chro-
final product concentration and buffer exchange, processing time
matography, SEC) for antibodies which were the majority of the
was estimated to decrease by 10%
protein projects. For other proteins, typically SDS PAGE and total
Waste: The waste of waiting emerged since the current work-
protein (by UV spec) were performed. Thus by reducing endotoxin
flow did not plan adequately for complexity. Solutions: Three
assays and avoiding other unnecessary assays, there was a 25%
changes were implemented: (1) Time was scheduled to re-run
reduction in assay efforts.
additional batches for failures owing to lack of expression, oper-
Waste: The waste of waiting was evident when steps waited
ational, or mechanical issues. The time added was based on the
for assay results. Solutions: This waste was minimized by running
2007 value of 83% for the first-time-through success rate. (2) The
sample assays overnight when possible and/or running titer and
entire process schedule was optimized to eliminate the weekend
SEC assays concurrently.
hold step after cell culture broth harvest and in some cases to
Waste: Over-processing also was evident when measurements
ship “at risk” before final assay confirmation was available. A stan-
recorded in batch records were not typically needed for future ref-
dard unit schedule was developed with typical steps occurring on
erence (e.g., recording weights after subdivision into centrifuge
specific weekdays and unattended cell cultivation occurring over
bottles, recording analytical results in a multiple places). Solutions:
weekends. Step overlaps (i.e., beginning the subsequent step while
This waste was resolved by eliminating unnecessary recording and
some material was still being processed in the prior step) were
by combining the analysis and delivery sheets based on customer
implemented. (3) Planning for and segregation of complexity was
input that one sheet was sufficient.
instituted by assigning one person to less variable tasks, and one or
Methods (process): more staff to more variable (typically higher complexity) tasks to
Waste: The waste of over-processing was evident since initial minimize uncertainty for the majority of deliveries. In this way, less
downstream procedures were based on the purification process frequent, uncertain cycle time, non-platform proteins were seg-
used to prepare clinical material. Solutions: These procedures were regated from the higher frequency, shorter cycle time, platform
soon adapted to meet non-clinical material requirements for use Protein A-based purification deliveries.
in in vitro assays and animal in vivo studies. Improved bioburden- Waste: The waste of defects was evident, particularly for non-
reducing harvest and purification operations (such as sterile platform tasks where proven methods were not always available.
filtration prior to holding periods and use of pre-sterilized dis- Solution: As methodologies were developed, work was standard-
posables) reduced endotoxin accumulation potential. This change ized (e.g., Ni-NTA affinity chromatography for his-tagged proteins)
largely eliminated the anion exchange chromatography step nec- and batch sheet templates were updated to reduce learning curves
essary for endotoxin removal, reducing processing time by about and improve success rates. Batch sheets were serialized and num-
440 B. Junker / Biochemical Engineering Journal 49 (2010) 435–444

Table 2
Overall summary of kaizen straightforward or significant time/resource improvements. Where possible, benefits estimated in % resource/time allocation to that task.

Critical to quality attributes Waste types Root causes Solutions Benefit

Reduced lead time Inventory Unclear prioritization Frequent weekly prioritization Timely delivery of high priority
criteria meetings requests
Changing basic research
priorities
Greater capacity/throughput Waiting Purification equipment Added chromatography system Increased number of parallel
(parallel deliveries) Talent and personnel bottleneck Switched fermentation FTEs to projects (1 machine/FTE), 50%
purification Raised purification & analytical
Cross-trained between staff interchangeability, 50%
purification & analytical staff
Waiting Manual operation of Acta Automated method Permitted off-hour
Talent purifier implementation for many steps chromatography unit
operation, 15%

Improved workflow effectiveness Talent Lack of use of disposables Implemented reliable Reduced time spent on
Defects Robustness of disposable disposables in processing cleaning & sterilization, 10%
equipment
Over-processing Clinical production Streamlined procedures to be Optimized steps (i.e., Protein
procedures utilized for suitable for less rigorous A), 10%
purification animal study and assay
development requirements
Over-production Unnecessary assays not Standardized on key assays and Reduced assay number, 25%
required by customer perform others by request only
Waiting End of workday assay Ran assays overnight or Reduced assay cycle time, 20%
interruptions concurrently after
Analytical personnel cross-training other staff
bottleneck
Over-processing Recording data not Combined analysis & delivery Single delivery sheet, 20%
needed for future sheets Reduced paperwork, 5%
reference Revise batch sheets to
streamline recording
requirements
Over-processing Wide variation in protein Used variable size filters & Efficient handling of wide
titers and processing columns/disposable range of proteins and
volumes ultra-filters requested amounts, 10%
Improved concentration & Optimized final processing
buffer exchange steps steps, 10%
Motion Suboptimal lab set up Executed 5S lab improvement Improved lab set up, 5%
activity
Achieved protein amount, quality, Defects Lack of proven methods Updated batch sheet templates Readily available prior results
and timing Serialized completed batch and best practices
sheets
Waiting Mix of non-platform & Segregation of high from low Predictable delivery for a
platform proteins complexity proteins majority of simpler requests,
Weekend processing Slotted schedule 10%
interruptions implementation with Decreased lead time, 40%
predefined “units” based on
complexity and assigned task
days
Defects High Implemented low bioburden Omitted one column step
bioburden/unacceptable processing procedures (anion exchange) for many
endotoxin levels for deliveries, 20%
some customers
Optimized in-house outsourcing Defects Low vendor success Identification of suitable and Ability to handle peak
splits rate/insufficient sufficient number of vendors workflows without staffing
technical ability internally
Over-processing Process required Re-develop process with More vendors met selection
expensive control inexpensive process criteria
instrumentation monitoring strategy Outsourcing of routine plasmid
production tasks to liberate
in-house resources

bers linked to delivery sheets to facilitate future reference for repeat hours, thus decreasing step cycle time and the degree of manual
or similar non-platform projects. operation. An additional chromatography system was acquired to
achieve one unit per purification staff, resulting in a 50% output
Machines:
increase during periods of peak operation, reducing changeover,
Waste: The wastes of talent and waiting were found in the
and permitting full downstream parallel processing.
manual operation of the chromatography systems (ACTA, GE, Pis-
cataway, NJ) and sharing of machines which required changeover Materials:
between two different purification procedures. Solution: Auto- Waste: The waste of defects was expressed in the form of four
mated ACTA methods [6] were created and implemented for specific rework points. In the cultivation stage, there was contam-
cleaning, equilibration, and regeneration steps, and eventually ination caused by faulty disposable equipment (e.g., vent filters on
column loading (column elution was considered too risky). This cultivation bags) and low titers owing to lack of process develop-
automation permitted many chromatography tasks to occur off- ment necessitating additional cultivations. In the purification stage,
B. Junker / Biochemical Engineering Journal 49 (2010) 435–444 441

there was insufficient protein recovery owing to low step yields and fits. The improvements cited fell into two classes. The first class
bioburden growth which raised endotoxin to unacceptable levels represented straightforward changes which were implementable
for some customers. Solutions: Improvements included retrofit of without any technical development or added resources. These
disposable equipment to improve reliability, installation of a target short-term changes had an impressive aggregate impact. A 3-fold
minimum culture titer cut-off for efficient downstream isolation, wait time (W/T) reduction from 13.9 to 4.4 days was realized by
and introduction of improved aseptic handling. optimizing the processing schedule and streamlining assay timing
Waste: The waste of talent was evident by the time required to to avoid weekend/evening interruptions (Table 3 and Fig. 1). W/T
clean equipment and prepare buffers. Solutions: In lieu of weighing was further impacted 2.5-fold (from 4.4 to 1.8 days) by streamlin-
buffer components, reconstituting, and filter-sterilizing, large vol- ing assay and documentation requirements (Table 3). There was a
ume sterile buffers were ordered already prepared from a vendor. smaller impact on cycle time (C/T), reducing it by 15%, by subdivid-
Disposable and pre-sterilized supplies were used to avoid clean- ing broth into centrifugation bottles using variable speed pumps
ing and sterilization. Where possible, remaining non-disposable and by combining analytical and delivery sheets (∼0.5 h each)
equipment was cleaned, assembled, and sterilized by laboratory (Fig. 1). Process cycle efficiency, the ratio of cycle time (value added
service personnel. Best practices were implemented for cleaning time) and lead time (value and non-value added time) improved to
equipment (e.g., 2 L centrifuge bottles which were too expensive at 35.1% (Table 3) which compares favorably to target value of 25% for
∼$50/bottle to be single use) to easily remove animal and microbial a lean business process [8]. Uptime for equipment was uniformly
cell pellets. estimated as greater than >95% for all steps so improvements in
Waste: The waste of motion was apparent since occupied lab- this area were not felt necessary.
oratory areas were not cleared of unneeded equipment from past The second class of improvements comprised long-term
occupants nor set up optimally for the required workflow. Unnec- changes that required significant time or resource investments.
essary energy was spent collecting equipment and transporting The cumulative impact of these changes also was substantial. Esti-
materials/buffers to and from different laboratories. Solutions: Sev- mates of cycle time decreases were 20% from downstream process
eral action items were identified as part of a group 5S activity (sort, simplification (i.e., improved aseptic harvest operations to achieve
set in order, shine, standardize, sustain; [7]) conducted in paral- low endotoxin and avoid the AEX step) and 10% from optimiz-
lel to this kaizen. These included floor markings for items such as ing disposable centrifugal ultrafiltration procedures and shortening
carts and biohazard bins, signs for buffer identification, station- concentration/buffer exchange times. Freed personnel time was
ary (or if possible mobile) hanging buffers, wheeled dollies for estimated at 10–15% from implementation of chromatography
drums for easy mobility, shelf labels, removal of unused machines automation (specifically, automated equilibration, loading, wash-
to liberate valuable laboratory bench top space, rearrangement of ing, and regeneration steps). Direct throughput increased by 50%
existing machines, creation of a material receipt area for supplies, from set up of a third chromatography system. All of the prioritized
and removal of old piping interfering with laboratory space access. short and long-term improvements – whether impacting step cycle
time, personnel time, wait time, or throughput – were able to be
Environment:
fully implemented by the kaizen team members alone.
Waste: The waste of transportation (long distances) was evident
in outsourcing to lower cost, emerging market rather than domes-
3.5. Metrics
tic vendors which also would eliminate customs. Yet, financial
incentives were a significant factor in outsourcing decision-making.
Delivery metrics were calculated from performance data and
Solutions: Both the low-cost and domestic objectives were met in
tracked monthly, then summarized and circulated to team mem-
only one case, specifically employing a local university to gen-
bers and customers via monthly reports.
erate cell pastes requiring purification. Training in domestic and
Initially, the goal was set at 80% of deliveries completed within
international shipping requirements lowered rework of shipping
the 8-week target from receipt of starting materials to delivery.
documentation. Optimal customer handoffs reduced reworking of
Between 2007 and 2008, the in-house group improved on lead
buffer exchanges or cassette transfers. Delivery timing also was
times for transient transfections but that improvement was off-
improved by coordinating with mid-day deadlines of the internal
set by a decrease in meeting lead times for hybridomas (Table 4).
shipping department.
Interestingly, this same trend was evident in the outsourced perfor-
Waste: The waste of inventory emerged owing to frequently
mance suggesting that the proteins being produced by hybridomas
changing project priorities and generally unclear prioritization
were more challenging in 2008. The delivery percentage achieving
criteria. Solutions: When projects were accelerated/decelerated
the target lead time was lower for the high complexity bacterial
as basic research efforts progressed, accumulated requests were
paste purifications in 2008. Despite the greater number of deliv-
reprioritized. Sometimes this reprioritization was not immediately
eries and their overall higher complexity in 2008, overall delivery
implementable since processing already was underway. In these
percentages meeting or exceeding target lead times were fairly con-
cases, processing was advanced to a suitable hold point. Frequent
sistent between 2007 and 2008. Table 5 illustrates the percentage
weekly meetings and other contact with stakeholders were crit-
of deliveries meeting the target of 50% of the requested amount. The
ical to rapidly understanding changing priorities. In some cases,
decrease in deliveries at target from 2007 to 2008 likely reflected
requestors were able to prioritize. For other cases, trial priori-
the 2008 practice of confirming with the customer that a rework
tization mechanisms were considered beyond simply satisfying
was necessary before an additional batch was automatically under-
the “squeakiest wheel” or the request received first. These alter-
taken.
natives included pre-allocated resources according to customer
The customer survey, implemented in 2007, was repeated in
sector, preference for lead optimization (where speed was critical)
2008 (Table 6). As a result of the 2007 customer survey, ∼80% of
rather than lead identification work, or alignment with the basic
the time only about 25% of the material delivered was used. Conse-
research program dashboard prioritization. The preferred solution,
quently, there was seldom a requirement for rework to meet target
however, was to size the rapid protein production effort sufficiently
delivery amounts. In most cases, at least some material delivered
to avoid frequent conflicts.
was used directly after receipt. However, most material was not
Overall: used suggesting that lead time was more important than delivery
Table 2 summarizes the link among critical to quality attributes, amount. Customers noted a positive impact of some type on their
category of observed waste, root causes, solutions, and bene- efforts for about 67% of all deliveries which was sustainable over 2
442 B. Junker / Biochemical Engineering Journal 49 (2010) 435–444

Table 3
Current and post-kaizen state value stream parameters after kaizen identifying straightforward improvements (see
Fig. 1 for value stream map).

Segment Current (days) Current (without off-time) (days) Post-kaizen (days)

Cycle time, C/T 1.5 1.5 1.3


Changeover time, C/O 0.6 0.6 0.6
Wait time, W/T 13.9 4.4 1.8
Lead (total) time 16.0 6.5 3.7
Process cycle efficiency, PCE 9.4% 23.1% 35.1%

Note: Calculations assume 7.5 h per day. Off-time is weekends and holidays. PCE is cycle time (value added time)
divided by lead time (value and non-value added time).

Table 4
Achievement of target lead times (goal of ≤8 weeks from receipt of starting materials to delivery) for in-house deliveries (goal >80%).

Category 2007 2008

No. of deliveries Percent ≤8 weeks No. of deliveries Percent ≤8 weeks

Transient transfection 38 74 27 89
Hybridoma 11 82 17 53
Purification from 65 98 82 98
customer-supplied cell
culture broth
Bacterial paste purification 0 N/A 12 42
Overall 114 88 138 86

Notes: (1) 2008 external vendor performance for transient transfection (n = 59) was 76% and for hybridoma (n = 15) was 6%, with hybridoma lower likely owing to vendor
performing additional batches to meet required delivery amount. (2) 2007 external vendor performance for transient transfection (n = 4) was 0% and for hybridoma (n = 11)
was 64%, with transient lower likely to unsuccessful vendor selection (vendors were switched subsequently). (3) Target lead time goal was the same for in-house and external
vendor deliveries.

Table 5
Achievement of target protein amount by in-house deliveries (goal >50% meet target amount).

Category 2007 2008

No. of deliveries Percent ≥ request No. of deliveries Percent ≥ request

Transient transfection 38 68 27 50
Hybridoma/stable cell line 11 64 17 38
Purification from customer-supplied cell culture broth 65 No request of amount made 82 No request of amount made
Bacterial paste purification 0 N/A 12 18

Notes: (1) 2007 external vendor performance for transient transfection (n = 4) was 50% and for hybridoma (n = 11) was 91%, with transient lower than expected likely to
unsuccessful vendor selection (vendors were switched subsequently in 2008). (2) 2008 external vendor performance for transient transfection (n = 59) was 88% and for
hybridoma (n = 16) was 94%, higher than 2007 likely owing to vendor performing additional batches to meet required delivery amount. (3) Target protein amount goal was
the same for in-house and external vendor deliveries.

Table 6
Customer feedback survey results [Units of % respondents].

Timeliness of material use (%) Amount of material used (%) Impact of material used (%)

Time after receipt to first use 2007 2008 % utilized 2007 2008 Study results 2007 2008

0–2 weeks 68 51 <25% 50 51 Project going forward 50 64


2–4 weeks 16 21 ∼25% 31 17 Project milestone achieved 19 3
4–6 weeks 5 8 ∼50% 1 16 Project dropped entirely 0 2
6+ weeks 8 11 ∼75% 12 7 Project switched to different candidate molecule 6 3
Not used 3 9 ∼100% 6 9 Project on hold or impact unknown 25 28

years. Based on this information, customers were encouraged not only broths, requiring no cell culture and conducted in-house.
to request more material than was required, and customers were Often, multiple protein purifications of small cell culture volumes
consulted before rework steps were undertaken if yields fell short. were requested together. Interestingly, less than 10% of the 2007
In 2008, two survey questions were added to rate turnaround time in-house deliveries were classified as non-platform proteins requir-
(56% excellent, 28% good, 10% fair, 6% needs improvement) and ing more complex processing, but these deliveries utilized about
material quantity received (48% excellent, 37% good, 8% fair, 7% 60% of the staff. In contrast, in 2008 the total annual delivery goal
needs improvement). This feedback showed good or excellent per- was set at 175 (3.5/week) initially, rising to ∼230 only 6 months
formance ∼85% of the time, suggesting few detrimental effects of later (Table 1). Moreover, complexity shifted dramatically with up
the implemented kaizen improvements. to 50% of in-house deliveries classified as highly complex, non-
platform deliveries (i.e., soluble and insoluble proteins expressed
3.6. Kaizen impact in E. coli). There were associated substantial challenges obtaining
advance protocol information, and often front runs were required
In 2007, although the total annual delivery goal was estimated to develop the protocol and in-process assays.
conservatively at 75, actual deliveries were substantially higher at Despite this significant complexity increase, deliveries per
196 (Table 1). About 60–65% of the deliveries were purification- purification staff per month increased dramatically from 2007 to
B. Junker / Biochemical Engineering Journal 49 (2010) 435–444 443

Table 7
Purification task metrics.

Measure Pre-kaizen (2007) Post-kaizen (2008)

Total deliveries 110 in 10 months 114 in 8 months


Projected estimated annual deliveries 132 171
#/month 11 ± 4 14 ± 5
Range of deliveries/month 6–19 2–22
No. of staff 3 3.5
Deliveries/week/staff (40 weeks per year) 1.10 1.22
Purity (%) 95.1 ± 7.5 N/A
Lead time (days) 12.1 ± 7.8 20.8 ± 22.6
Lead time platform only (days) 11.1 ± 5.9 7.7 ± 3.5

Platform only process capability (Cpk )


(normal distribution, AD = 1.14)
14 days Not able to be 0.87
21 days calculated 1.82

AD, Anderson–Darling normality test coefficient.

2008 (Table 7). Moreover, the kaizen permitted the in-house group 4. Future improvements
to exceed its 2007 deliveries by 1.3-fold, despite expanding support
of high complexity requests. Concurrently, the in-house group size The rapid reagent protein production group has impacted sev-
was reduced by 25% and outsourcing was increased by 40% (roughly eral critical go/no-go decisions (specifically which basic research
equivalent to 1 staff plus materials) for no net resource gain. As projects advance forward and which projects are placed on hold
shown by Table 7, in-house downstream deliveries increased 12% or discontinued). It has supplied various antibodies for evalua-
from 1.1 to 1.2 per staff per week. Platform delivery lead time tion, as well as reagents for release and clinical pharmacokinetic
decreased by 30% from a mean of 11.1 to 7.7 days and a standard assays to support fast timelines to achieve first-in-human clini-
deviation (variation) decline of 40% from 5.9 to 3.5 days (99% con- cal milestones. Although this particular group was located within
fidence level). In 2008, the process capability, Cpk (i.e., the ability a large biopharmaceutical company, its reagent supply function
of process to meet customer requirements; [7]) for the platform is key to most organizations engaging in biological research and
purification delivery lead times that were the primary focus of the development – just scaled by the size of the specific effort. Future
kaizen effort was calculated to be 0.87 (2.6 sigma or 135,000 fail- processing improvements for platform deliveries largely depend
ures per million opportunities) for 14 days and 1.8 (5.4 sigma or 50 on the suitability of alternate technologies, such as membrane
failures per million opportunities) for 21 days. chromatography for low titer and low volume processing or low-
Fig. 2 shows a control chart of individual lead times for in- cost, disposable, tangential ultrafiltration membrane units. Future
house purification deliveries in three sections: pre-kaizen in 2007, improvements clearly now are required for non-platform deliv-
immediately post-kaizen in the first part of 2008, and then later in eries and depend on substantial investments to develop suitable
2008 when proteins of greater complexity (i.e., non-platform pro- approaches.
teins requiring significant method development) were undertaken. Moving forward, effectively leveraging external vendor capa-
Mean lead time decreased substantially as a result of the kaizen, yet bilities likely will remain critical. It is desirable to fully develop
later increased as a result of added complexity. This trend suggested external capabilities for protein deliveries, preferably in low-
further actions were necessary to reduce variation and lead times cost geographies to control cost structure without compromising
for this updated assortment of rapid protein reagent requests. These quality, timeliness, or intellectual property. One possibility is to
actions might include a subsequent kaizen to identify and mitigate restructure the in-house group to supplement vendor supplies only
gaps in the group’s current workflow. when necessary. The current state outsourcing timeline included
1–2 weeks to ship starting materials plus an average of 5.9 weeks
(n = 40) for the low-cost, emerging market vendor to deliver within
the 8-week target. Expedited deliveries of 2.5 weeks after start-
ing material receipt were possible if the vendor did not perform a
front run to confirm performance. [This front run reduced vendor
uncertainty since contracts were based on protein mass delivered
and not production runs.] Additional reductions in vendor price
and lead time were possible using 6–12 month request forecasts.
These forecasts were more straightforward for analytical reagents,
but also are possible based on basic research discovery cycle time
estimates.
Information technology support also is expected to be crucial
to achieve further improvement. Web-based submission requests
(with multiple types of submission forms to suit widely differing
customers) and progress tracking tools were two specific elements
identified. These web-based tools link to a database permitting
access to interim and milestone status metrics, including a dash-
board overview of on/off track deliveries. Web-based tools are
Fig. 2. Control chart of in-house purification delivery lead times: 2007 pre-kaizen, somewhat limited to routine requests (i.e., similar to capabilities
2008a post-kaizen, 2008b added complexity (post-kaizen). Mean: X̄; LB: lower already undertaken). Unusual requests, which are highly likely as
bound; LCL: lower control limit (−3 standard deviations); UCL: upper control limit
new discovery platforms are adopted, obviously require direct con-
(+3 standard deviations). Red points indicate times that are outside the calculated
control limits. (For interpretation of the references to color in this figure legend, the versation before becoming standardized options. Such interaction
reader is referred to the web version of the article.) also benefits joint and pro-active suitability assessments of pro-
444 B. Junker / Biochemical Engineering Journal 49 (2010) 435–444

posed protein expression systems to ensure submitted requests [2] M. Allen, M.J. Wigglesworth, Innovation leading the way: application of lean
yield reasonable expression levels [9]. manufacturing to sample management, Journal of Biomolecular Screening 14
(5) (2009) 515–522.
[3] H. Tegel, J. Steen, A. Konrad, H. Nidin, K. Pettersson, M. Stenvall, S. Tourle,
Acknowledgements U. Wrethagen, L. Xu, L. Yderland, M. Uhlen, S. Hober, J. Ottosson, High
throughput protein production—lessons from scaling up from 10 to 288
recombinant proteins per week, Biotechnology Journal 4 (1) (2009) 51–
This paper represents the efforts of past members of the rapid 57.
protein production group: Neal Connors, Jih-Han Hsieh, Jessica [4] R. Suri, Quick Response manufacturing, Productivity Press, New York, 1998.
Okonskowski, Tim St Clair, Kathleen McLaughlin, Angie Sun, David [5] J. Ye, V. Kober, M. Tellers, Z. Naji, P. Salmon, J.F. Markusen, High-level protein
expression in scalable CHO transient transfection, Biotechnology and Bioengi-
Lee, Diane Vesey, Hari Pujar. The enthusiastic training and facil- neering 103 (3) (2009) 542–551.
itation contributions of Ed Hankler (New Jersey Manufacturing [6] D.V. Camper, R.E. Viola, Fully automated protein purification, Analytical Bio-
Extension Program, NJMEP) are also gratefully acknowledged. chemistry 393 (2009) 176–181.
[7] B.H. Junker, Applying operational excellence concepts to biopharmaceutical pro-
cessing, BioPharm International 21 (11) (2008) 54–60.
References [8] http://www.isixsigma.com/dictionary/Process Cycle Efficiency PCE-691.htm.
[9] W.H. Brondyk, Selecting an appropriate method for expressing a recombinant
[1] A. Drakulich, Pfizer offers example of lean design application for clinical sup- protein Methods in Enzymology, vol. 463, Elsevier, New York, 2009, pp. 131–
ply manufacturing. PharmTech.com, August 2, 2007, http://license.icopyright. 147.
net/user/viewFreeUse.act?fuid=Mjg4OTk0MA%3D%#D.

You might also like