single-use technology implementation for biologics and ...download.xuebalib.com/oe3uz0mvt8z.pdf ·...

21
721 Biopharmaceutical Processing. https://doi.org/10.1016/B978-0-08-100623-8.00035-9 © 2018 Elsevier Ltd. All rights reserved. Single-Use Technology Implementation For Biologics and Vaccines Production David J. Pollard*, Alain Pralong * Merck & Co., Inc., Kenilworth, NJ, United States, Pharma-Consulting ENABLE GmbH, Solothurn, Switzerland 35.1 SUMMARY The continued success and subsequent expansion of biologics and vaccines is met with significant industry pressures. Drug candidate pipelines are increasing with a range of heterogeneous products containing new modalities such as mAbs, bio- specifics, fusion proteins, and nanobodies. This diverse portfolio carries a greater range of product demands (kg/year). For biologics, the demand now considers a range from 50 to > 500 kg/year compared to the traditional mAb demand of ~250 kg/ year. In addition, there is increasing uncertainty to the prediction of new pipeline molecule demands (kg/year). The industry cost pressures continue to mount, including evolving reimbursement strategy, global competition, and loss of drug exclu- sivity. Meanwhile, the need for widening global patient access to life-saving drugs must also be addressed. Therefore, the industry needs production platforms that are lower cost, flexible, and agile to meet these rapidly changing demands.SUT can support this challenge by providing a simpler, faster, and lower-cost route to production capacity, when compared to the con- ventional stainless-steel equipment and facility design. Cost advantages with SUT can be expected both on capital investment and operational expenses. The use of presterilized, gamma-irradiated SUT eliminates the need for clean-and-steam in-place operations, while supporting sustainability. This simplifies both the design of a facility infrastructure and the complexity of each unit operation equipment skid. The engineering and construction costs are reduced and the time from project start to production readiness can shrink up to 50% with predesigned modular facility layouts [1]. The improvement in cell line expression titers and the ability to run more concentrated (intensified) processes has allowed the reduction of manufacturing scale from the traditional 10,000 L stainless facilities to volumes to 2000 L and below. This provides additional cost savings from reduced capital, translating to lower depreciation and smaller facility footprints. The elimination of cleaning and steam- ing in place allows a faster turnaround between batches, providing an increased facility throughput. This supports simpler and lower-cost operational routines that can lead to lower labor and utility/material expenses. These factors culminate into a major advantage for SUT in terms of an optimized cash flow or net present value (NPV). A number of biopharma companies have invested in modular hybrid SUT facilities for clinical (Biogen Idec, WuXi App Tec, Patheon, Rentschler, and FujiFilm) and commercial production (Shire, Amgen, Pfizer, CMC Biologics, and WuXi AppTec). These currently incorporate single- use bioreactors for upstream and predominately rely on stainless-steel reusable technologies for purification. In the coming years, it is anticipated this will change as single-use purification technologies gain maturity [2,3]. This combination of modular facility and SUT provides flexibility to meet the new drug modalities and variable pro- duction demands. A particular SUT can be brought into the facility when required in a plug-and-play approach. End-users are developing a toolbox of flexible single-use platform solutions including batch processing, semi, and fully continuous operations [4]. The lower-cost modular facilities provide a solution to allow expansion around the globe, to support widen- ing patient access to critical drugs. Despite these significant advantages, a number of challenges to single-use implemen- tation remain. These include the lack of regulatory guidance and standardized approaches for equipment qualification. SUT remains difficult to integrate in the plug-and-play approach. The end-user reliance on supplier supply chains carries significant risk. The single-use component manifolds (disposable bags, filters, and tubing sets) need standardized design and testing approaches for key issues including particulates and extractables/leachables. This chapter sets out to provide guidance on the following topics: (1) the decision process and economic cost considerations, (2) advice on process optimization to best prepare for single-use operation, Chapter 35

Upload: others

Post on 07-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

721Biopharmaceutical Processing. https://doi.org/10.1016/B978-0-08-100623-8.00035-9© 2018 Elsevier Ltd. All rights reserved.

Single-Use Technology Implementation For Biologics and Vaccines ProductionDavid J. Pollard*, Alain Pralong†

*Merck & Co., Inc., Kenilworth, NJ, United States, †Pharma-Consulting ENABLE GmbH, Solothurn, Switzerland

35.1 SUMMARY

The continued success and subsequent expansion of biologics and vaccines is met with significant industry pressures. Drug candidate pipelines are increasing with a range of heterogeneous products containing new modalities such as mAbs, bio-specifics, fusion proteins, and nanobodies. This diverse portfolio carries a greater range of product demands (kg/year). For biologics, the demand now considers a range from 50 to > 500 kg/year compared to the traditional mAb demand of ~250 kg/year. In addition, there is increasing uncertainty to the prediction of new pipeline molecule demands (kg/year). The industry cost pressures continue to mount, including evolving reimbursement strategy, global competition, and loss of drug exclu-sivity. Meanwhile, the need for widening global patient access to life-saving drugs must also be addressed. Therefore, the industry needs production platforms that are lower cost, flexible, and agile to meet these rapidly changing demands.SUT can support this challenge by providing a simpler, faster, and lower-cost route to production capacity, when compared to the con-ventional stainless-steel equipment and facility design. Cost advantages with SUT can be expected both on capital investment and operational expenses. The use of presterilized, gamma-irradiated SUT eliminates the need for clean-and-steam in-place operations, while supporting sustainability. This simplifies both the design of a facility infrastructure and the complexity of each unit operation equipment skid. The engineering and construction costs are reduced and the time from project start to production readiness can shrink up to 50% with predesigned modular facility layouts [1]. The improvement in cell line expression titers and the ability to run more concentrated (intensified) processes has allowed the reduction of manufacturing scale from the traditional 10,000 L stainless facilities to volumes to 2000 L and below. This provides additional cost savings from reduced capital, translating to lower depreciation and smaller facility footprints. The elimination of cleaning and steam-ing in place allows a faster turnaround between batches, providing an increased facility throughput. This supports simpler and lower-cost operational routines that can lead to lower labor and utility/material expenses. These factors culminate into a major advantage for SUT in terms of an optimized cash flow or net present value (NPV). A number of biopharma companies have invested in modular hybrid SUT facilities for clinical (Biogen Idec, WuXi App Tec, Patheon, Rentschler, and FujiFilm) and commercial production (Shire, Amgen, Pfizer, CMC Biologics, and WuXi AppTec). These currently incorporate single-use bioreactors for upstream and predominately rely on stainless-steel reusable technologies for purification. In the coming years, it is anticipated this will change as single-use purification technologies gain maturity [2,3].

This combination of modular facility and SUT provides flexibility to meet the new drug modalities and variable pro-duction demands. A particular SUT can be brought into the facility when required in a plug-and-play approach. End-users are developing a toolbox of flexible single-use platform solutions including batch processing, semi, and fully continuous operations [4]. The lower-cost modular facilities provide a solution to allow expansion around the globe, to support widen-ing patient access to critical drugs. Despite these significant advantages, a number of challenges to single-use implemen-tation remain. These include the lack of regulatory guidance and standardized approaches for equipment qualification. SUT remains difficult to integrate in the plug-and-play approach. The end-user reliance on supplier supply chains carries significant risk. The single-use component manifolds (disposable bags, filters, and tubing sets) need standardized design and testing approaches for key issues including particulates and extractables/leachables.

This chapter sets out to provide guidance on the following topics:

(1) the decision process and economic cost considerations,(2) advice on process optimization to best prepare for single-use operation,

Chapter 35

Page 2: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

722 SECTION | VI Industrial Process Design

(3) defining aspects of single-use component supply and regulations, and(4) discussion of potential future developments.

35.2 BENEFITS OF IMPLEMENTING SINGLE-USE TECHNOLOGY

35.2.1 Impact of Single-Use Technology on Biopharmaceutical Manufacturing

The benefits of disposable single-use technology (SUT) are outlined in Table 35.1 and have been extensively discussed in recent reviews [5–7]. There are now single-use technology options for the majority of unit operations required for manu-facturing monoclonal antibodies (mAb) or vaccines. Further details of these technologies, such as disposable tubing bag component manifolds, bioreactors, depth filters, disposable columns, and disposable purification membranes can be found in Chapter 18. The use of single-use technology, such as a disposable bioreactor, eliminates the need for clean-and-steam in place as the end-user purchases a presterilized gamma-irradiated bag, directly from the supplier. This simplifies the bioreac-tor skid as up to 70% of the automation costs are associated with clean-and-steam in place. This reduced footprint provides a lower capital solution and reduced capital depreciation. The turnaround time between batches is shortened, enabling more batches per year and increased facility utilization.

These benefits of single-use come at a time when the industry faces significant competitive challenges. These include evolving reimbursement environment, global competition, personalized medicine, lost drug exclusivity, biosimilar and ex-panding drug pipelines [8]. Single-use technology provides a tool to innovate the biopharma workflows and commoditize bioprocess manufacturing. For example, rapid drug development and clinical production is enabled by single-use [9,10]. In addition, commercial facilities can be now designed with single-use technology up to 2000 L scale using high titer pro-cesses (>5 g/L). A transition from fixed to variable cost is underway with the shift from large 20,000 L scale stainless-steel facilities to a new paradigm of modular facility design with SUT (Fig. 35.1). The single-use technology enables unit opera-tions to be connected so as to eliminate open processing. This closed processing allows the clean air handling classification requirements to be reduced and affords an open ballroom design (see Chapter 45) with reduced footprint. Closed SUT pro-cessing also has the potential to enable concurrent multiple product processing [1,11,12]. The open plan layout allows the facility to be easily adapted to these rapidly changing needs. The short construction of the modular facilities within [13–15] months allows a quick response to rapidly changing market demands by scaling out with additional modules.

SUT provides a lower cost and flexible manufacturing capacity that can be easily adapted for the expanding modalities of biologics. This demand can now vary from low (<50 kg/year) to large supply (>300 kg/year), and will require a toolbox of platforms, including integrated and continuous processing enabled by disposable technology (Fig. 35.2).

This flexibility is gained from the ability to easily move single-use technology equipment skids in and out of a facil-ity process floor in a “plug-and-play” approach. This flexibility is not easily feasible in a conventional, larger scale rigid structured stainless-steel facility.

35.2.2 Economic Cost Analysis of Single-Use Technology

The foundation of the decision to consider single-use technology implementation is the economic cost benefit provided by the improved speed and flexibility. This economic process cost analysis should be completed as early as possible in the decision- making process. When evaluating the cost benefit of a single-use option for a particular unit operation, it is

TABLE 35.1 A Summary of the Potential Benefits of Applying Single-Use Technology (SUT) to Bioprocessing

Financial Drivers Process Factors Key Influences to Operating Costs

Increased flexibility Ability to rapidly influence facility utilization.Potential for up to 50% faster implementation and installation cost reduction

Production rate change (product kg/year)

Higher equipment and facility utilization Increased number of batches per year

Lower fixed costsLower capital cost

Eliminating clean and steam in place infrastructure lowers capital cost of the processing equipment and facility infrastructure

Cost of SU componentsMaintenance cost differenceCost of purified water and steamValidation costPersonnel requirements

Page 3: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 723

Future state

Range ofmodalities &batch sizes

Flexible, lowcost agilecapacity

Toolbox ofintegratedsingle useplatforms

Real timerelease

Responsiveshort lead

time supplychain

Variable costsfocus

Current state

Mab focusRigid batch size

Stainless steelfed batch

SeparateDS & DP

ConventionalQC

Long lead timesupply chain

Fixed costsfocus

COGs $highfacility capital >

$600M

COGs $lowfacility capital

$50M

FIG. 35.1 Transition from traditional stainless steel to lower cost modular facilities enabled by disposable single-use technologies. The modular facility has inventory warehouse, media/buffer makeup, buffer/media storage linked next to the process open facility (light blue designated area). This will enable a flexible solution to changing modalities and demand. In addition, it will enable a close synchronization of drug substance production integrated with drug product supply to meet patient drug demands with shortened lead times and reduce inventory costs.

Adaptive process control

Predictive MDVA models

Real time release testingQC shop floor

Consistent quality assurance

PAT tools

Synchronized supply chain

On demand productionPortable capacity for

Local or regional options

Automated continuous processing

Flexible facilityPlug-and-playtoolbox

Multiproduct

Expansionvia scale out

Single use closedprocessing

Moldedparts

0–8

–6

–4

–2

0

t[1]

2

4

20 40 60 80 100 120 140 160 180 200

Log Hour

220 240 260 280 300 320 340 360

SIMCA 130 - 8/28/

FIG. 35.2 An example vision of the modular flexible facility of the future enabled by single-use technology for CHO mAb manufacturing. The open ballroom design provides a flexible processing area to support multiple single-use platform configurations such as fed-batch, semicontinuous, and fully automated continuous processing. The processing area is supported by media/buffer makeup, process analytical technology (PAT), single-use operations, and single-use bag manifold tubing systems engineering.

Page 4: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

724 SECTION | VI Industrial Process Design

important to consider the potential impact on the previous and next steps of the process. For example, replacing a stainless-steel fed-batch process at 3 g/L product titer with an intensified higher titer concentration 20–30 g/L process will have cost implications for the resizing of the purification. The cost implications can be analyzed by using a total cost of ownership model that encompasses end-to-end process manufacturing costs. A useful tool to manage this cost workflow is the BioSolve process cost analysis tool [10] that provides a total cost of ownership including facility construction costs, capital, and op-erating costs with depreciation. Typically, in this modeling software, the new single-use process is compared against a fixed cost stainless-steel process. An example of the cost benefit obtained with a single-use bioreactor compared to stainless-steel can be seen in Fig. 35.3. This outlines the range of parameters that must be included in the cost assessment. The analysis determined that despite the additional cost from disposable consumables, SUT was 40% cost favorable over stainless steel.

Completing this process cost modeling exercise generates a good understanding of the cost sensitivities. Establishing this understanding early in the decision-making process can help define the process development targets needed to over-come potential bottlenecks. Identifying the upstream titer target where single-use becomes cost favorable is one such example. Whilst the output of the cost analysis should include capital cost and cost of goods (COGs), it should also encompass the net present cost (NPC) which is an end-to-end cost assessment that includes capital, depreciation and over-head associated with the facility for a defined lifetime (typically 15–20 years). This allows for delays in expenditures and properly accounts for the time value of money [16]. If no profits are accounted during NPV calculation (because of a lack of sales revenue), the negative NPV is interpreted as the net present cost (NPC). This allows for the evaluation of different manufacturing approaches and technologies across the product lifecycle while accounting for the initial capital investment, operating costs, and the amount of product generated per year or per batch [16]. So, the option with the lowest NPC value provides the most favorable financial option.

An example of such an approach is shown in Fig. 35.4 which visually summarizes the impact of the technology platform options to the net present cost (NPC) for a standard CHO cell culture mAb process. This approach assessed the initial tech-nology suitability for driving financial benefits to a stainless-steel approach. Visualizing NPC as a function of production rate has proven to be an effective way to summarize and compare different process platform configurations as it shows low and high facility rates in the same curve [16]. The traditional stainless-steel (6× 15 kL bioreactor) facility has the high-est multimillion NPC cost with a capital facility build cost of over $600 M. The facility contains six stainless steel 15 kL

100

50

Co

st s

avin

gs

(%)

Stainless

Operatinglabor

Media

Capital

Single Use

DisposablesInstallation

0

–50

–100

–150

–200

–250

Cap

ital c

ost

Inst

alla

tion

CIP

/SIP

Dis

posa

bles

Ope

ratin

g la

bor

Med

ia

Mai

nten

ance

Ann

ual c

hang

e ov

er

Util

ities

Ove

rhea

d

Lead

tim

e

Tota

l cos

t

Capital cost Installation CIP/SIP

Disposables Operating laborMedia

Maintenance Annual change over Utilities

Overhead

FIG. 35.3 An example of total cost analysis comparing stainless-steel to single-use bioreactor for mAb production at 2000 L scale with fed-batch process (3 g/L titer).

Page 5: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 725

bioreactors along with a stainless-steel purification process including centrifugation for cell removal, depth filtration for particulate removal and three chromatography steps for purification. The single-use process contained 6× 2000 L single-use bioreactors and purification was performed using prepacked chromatography columns (60 cm). Process steps included single-use mixers <2000 L and storage bags <3000 L. The analysis showed that a 6× 2000 L single-use fed-batch facility can manufacture up to 500 kg/year using a 3 g/L cell culture titer. To go beyond 500 kg, new additional facility capacity needs to be brought online. This is shown by the incremental step increase in NPC from the additional of another 6× 2000 L SUB facility. This expansion then takes the capacity to over 1000 kg/year from the two facilities, each with 6× 2000 L SUBs. This additional facility capital cost is smaller than for the first due to the capacity being added later in the lifetime of the product reducing the impact of inflation. The significant shift of NPC between the conventional stainless-steel facil-ity and the SU facility equates to multimillion dollar savings. The majority of these savings come from the lower capital investment and capital deferral for additional facilities. The next generation technology options such as higher titer 10 g/L and continuous processing provide the most significant cost reductions, shown by the lowest NPC values. The continu-ous benefits come from high titer production 2 g/L day with the benefits of rapid continuous operations and a streamlined workforce [4,13–15]. Higher throughput improvements (3–4× faster) are supported by resin reduction from multicolumn chromatography, single pass TFF, novel membranes and the elimination of intermediate hold steps.

35.3 DESIGNING AND IMPLEMENTING A SINGLE-USE TECHNOLOGY PROCESS

35.3.1 Creating a Single-Use Technology Based Process

Historically, scientists and process engineers have developed manufacturing processes based on their knowledge and technical preferences [17]. Key features of these processes have been captured in user requirement specifications (URS). Engineering teams use this information to design processes and facilities to the required scale. This traditional approach led to laboratory grade processes being scaled up and registered for routine manufacturing. This resulted in a high level of customization within dedicated facilities resulting in significant process inefficiencies and high-risk exposure through single-sourced equipment. Economic pressure, along with major regulatory and GMP compliance issues, have driven the development and adoption of broader platform process designs and equipment. In the case of monoclonal antibody (mAb) manufacturing, a generic process has been defined as presented and described in the mAb case study [18]. Today, the major equipment and solution providers offer traditional process technologies that fit into the generic mAb manufacturing

1000

700

600N

et p

rese

nt

cost

($M

M)

500

400

300

100

00

200

800

900

200 400

6x15kL SS, Fed batch/batch, 3g/L6x2kL SU, FB N-1 FB N/batch, 3g/L4x2kL SU, perfusion/continuous, 2g/L6x2kl SU, perf N-1 Int N/batch, 10g/L

600 1000 1200 1400 1600800

Capacity (kg/year)

Stainless steel 6x 15kL facility

Single use hybrid fed batch 6x 2kL facility

Single use intensified fed batch(10g/L) 6x 2kL facility

Single use perfusion/continuous4x 2kL facility

Capital cost of second facility

First facility variableoperating cost

Capital cost of first facility

FIG. 35.4 Comparison of CHO mAb production with novel single-use processes compared to conventional stainless-steel. NPC: net present cost: rec-ognizes total cost of ownership approach encompassing capital cost, facility construction, depreciation and operating expenditure for a brownfield facility assuming 20 years of operation. The lowest NPC is the most desired process technology platform.

Page 6: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

726 SECTION | VI Industrial Process Design

platform process at various scales. This eliminated the need for the majority of customizations typical of the past. However, although the advent of single-use technology has afforded many benefits, it has primed a return to the in-house designed solutions and customization increasing the overall risk-exposure of SUT manufacturing operations. This fact, along with recurrent supplier reliability issues, is hindering the faster and wider adoption of single-use technology into production op-erations. These issues, until resolved, will continue to support the traditional stainless-steel based approach to be less risky [19]. Single-use limitations inherent to single-use technologies such as scale, connectivity, flow, pressure, temperature and pH limitations, respectively, require a structured process design and equipment assessment/selection workflow. It is this structured approach that ensures right-first-time success with favorable economics. The approaches to overcoming the tech-nological and supply-continuity risk-associated hurdles of single-use technology are discussed in the following sections.

35.3.2 Basis of Process Design and Equipment Selection

The design of a manufacturing process begins in an optimal way when the end-goal is clearly defined. Building on the understanding of the end-state permits comparison of different options and final selection of the most appropriate solution. Process development should have a line of sight to the facility that will ultimately receive the process. This is key for a suc-cessful process fit into the final facility. Table 35.2 summarizes key questions relevant for optimal process design.

35.3.3 Regulatory Requirements for SUT Implementation

The basic regulatory principles valid for SUT use in the biopharmaceutical industry remain unchanged, as regulatory agen-cies have not issued any new major guidance for SUT:

● Single-use components have to be identified within a process flow.● Single-use components have to be assessed in a risk-based approach (e.g., failure mode effect analysis (FMEA), process

hazard analysis (PHA) in the framework of the intended use and conditions.● Suppliers of single-use components have to be audited and assessed with regard to compliance with relevant regulatory

requirements (e.g., change control, validation) and supply reliability.● Single-use components have to be released and maintained through change control.● Single-use components have to be qualified (IQ, OQ) in combination with hardware/equipment used for operation

where applicable.● Single-use components have to be qualified in the frame of a process qualification (PQ) and validation (PV).

35.3.4 Process Architecture and the Control Strategy for Maintaining Product Quality

As indicated in the quote, “All roads lead to Rome,” various options exist to manufacture a mAb or vaccines. However, dif-ferences in process architecture may impact not only capital expenditure (CAPEX) and operational expenditure (OPEX),

TABLE 35.2 An Example of Eight Key Questions Relating to Process Design and Comparability Between Possible Process Architecture Solutions

Relevant Questions for Process Design

1. Are estimates of commercial production volume demand available, and is the single-use approach the most cost favorable?

2. Are cell line productivities known, and is a cell line improvement expected?

3. Does the active pharmaceutical ingredient have known critical physical-chemical characteristics and critical product attributes?

4. Does the preparation of the active pharmaceutical ingredient require any critical physical-chemical process sensitivities?

5. Are all unit operations available in single-use technology and at the desired scales from lab to commercial production?

6. Can adaptation of production capacities be achieved through a scale-out approach by increasing the number of identical production trains, and is this cost favorable?

7. Will the process be operated in a mono- or multiproduct facility?

8. Can customization be restricted to an absolute minimum?

Page 7: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 727

but also overall risk-exposure with regard to manufacturability, process robustness, and compliance. For example, a mAb can be manufactured by two approaches using Chinese Hamster Ovary (CHO) cell culture as illustrated in Fig. 35.5. The first option (Fig.  35.5A) uses a traditional approach with predominantly stainless steel processing and open handling manipulations using a biosafety or laminar flow hood. The second, improved approach (Fig. 35.5B) uses predominantly single-use equipment with fully closed fluid transfers between unit operations via welded or connected tubing. This SUT architecture eliminates three process steps to reach the identical production vessel volume, and the elimination of open handling steps performed by an operator significantly reduces potential contamination risks.

Understanding the TPP dictates the design and development of capability of the manufacturing process and has been translated into corresponding regulatory guidelines in the EU [20] and the US FDA (CFR 21 Part 210, [21]). The overall strategy for maintaining product quality and control via a risk & science-based approach is defined in Fig. 35.6. The TPP is structured into quality attributes that are differentiated into critical and noncritical attributes. For critical quality at-tributes, a FMEA and a PHA are executed to assess the link between process parameters and quality attributes with the objective of distinguishing between critical and noncritical process parameters. The type and magnitude of impact of a critical process parameter on a CQA is determined in design of experiment (DOE) studies. The outcome of the DOE studies

(A)

Batch

batch

Fed

Batch

fed

Batch

t = 5min

t = 15min

t = 2,4days

t = 3days

t = 2,8days

t = 4,5days

vi = 1 mL

vcdi = 10×106 cells.mL–1

vcdi = 10×106 cells.mL–1

vcdi = 0.5×106 cells.mL–1

vcdi = 0.5×106 cells.mL–1

vcdi = 0.5×106 cells.mL–1

vcdi = 0.5×106 cells.mL–1

vcdi = 0.5×106

cells.mL–1

vcdf = 5×106

cells.mL–1

vcdf = 2,5×106 cells.mL–1

vcd = 60×106 cells.mL–1

vcd = 60×106 cells.mL–1

vcdf = 2,5×106 cells.mL–1

vcdi = 0.6×106 cells.mL–1

vcdi = 0.5×106 cells.mL–1

vcdi = 0,5×106 cells.mL–1

vcdi = 2,76×106 cells.mL–1

vcdf = 25×106 cells.mL–1

vcdf = 2,5×106 cells.mL–1

vcdf = 3×106 cells.mL–1

vcdf = 2,8×106 cells.mL–1

vcdf = 5×106 cells.mL–1

vcdf = 3×106 cells.mL–1

vcdi = 0,5×106 cells.mL–1

vcdi = 2,8×106 cells.mL–1

vcdf = 25×106 cells.mL–1

vcdf = 2,8×106 cells.mL–1

vi = 20 mL

vi = 100 mL

vi = 500 mL

vi = 5 L

vi = 50 L

vi = 10 L

vi = 100 mL

vi = 50 L

vi = 300 L

t = 2,7j

t = 2,7j

t = 3,8j

t = 3,8j

t = 3j

t = 2,8j

t = 4,54j

(B)

Ampoule 1 mL

STEP 1ThawingT = 37ºC

STEP 1ThawingT=37ºC

STEP 2Wave 20/50

tv=50Lwv=10LT=37ºC

STEP 3Wave 20/50

tv=50Lwv=25LT=37ºC

STEP 3Wave 20/50

tv=50Lwv=25LT=37ºC

STEP 2T-Flask

wv = 20 mLT = 37ºC

STEP 3Roller Bottle 490 cm2

wv = 100 mLT = 37ºC

STEP 4Roller Bottle 850 cm2

wv = 250 mLT = 37ºC

STEP 4Roller Bottle 850 cm2

wv = 250 mLT = 37ºC

STEP 5Bioreactor BIOSTAT Cplus

tv = 13Lwv = 5LT = 37ºC

STEP 6Bioreactor BIOSTAT D-50

tv = 66Lwv = 50LT=37ºC

STEP 6Bioreactor BIOSTAT D-500

tv=660Lwv = 307L

T=37ºC

STEP 4Bioreactor XDR-500

tv=650Lwv = 300 L

T=37ºC

STEP 8Bioreactor BIOSTAT D-500

tv=660Lwv = 300L to 500L

T=37ºCc=4 g/L

Frozen Wave bag100 mL

STEP 5Bioreactor XDR-500

tv=650Lwv = 300L to 500L

T=37ºCc = 4g/L

FIG. 35.5 A comparison of two different upstream process architectures potentially feasible for manufacturing of a desired mAb using CHO cell cul-tivation. (A) A process architecture consisting of open handling steps performed under a laminar flow hood by an operator using single-use vessels and a sequence of stainless-steel bioreactors of increasing volume. (B) A process architecture consisting only of single-use technologies. The acronyms are: vcdi, initial viable cell density; vcdf, final viable cell density; vi, inoculum volume; tv, total volume; wv, working volume; c, final MAb concentration; and T, temperature.

Page 8: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

728 SECTION | VI Industrial Process Design

permits definition of a process design space defined by a normal operating range (NOR), alarm limits, action limits, and a knowledge design space, respectively. Based on the size of the NOR and the alarm and action limits, a control strategy can be developed. Process analytical technologies (PAT) can also be implemented to control and mitigate risk during process execution. This approach for risk- and science-based process execution enables active control of the manufacturing param-eters to ensure fulfilling the TPP instead of passively operating a process at setpoints unrelated to CQAs. Table 35.3 lists examples of the key quality attributes for a mAb.

35.3.5 End User Expectations of SUT Suppliers

Manufacturers of biopharmaceutical drugs are expected to ensure reliable supply of efficacious, safe, and affordable drugs. Inspections by regulatory authorities assess the capability of a biopharmaceutical manufacturer to comply with those ex-pectations. Noncompliance results in enforcement actions instigated by regulatory authorities, and multiple examples have occurred since the introduction of the first biopharmaceuticals [22,23]. Embedding single-use technologies into commer-cial manufacturing processes has primed scrutiny by regulatory authorities, resulting in enforcement actions. The end-user is reliant upon the single-use supplier to provide the documented evidence to support qualification and validation of the single-use technologies they provide. The end-user performs regular audits of the supplier operations to ensure quality re-quirements and regulatory expectations are maintained. As a consequence, and beyond just ensuring a reliable supply chain, multiple parameters have to be considered for successful embedding and utilization of single-use technologies. Table 35.4 lists parameters defining expectations end-users have from suppliers. It is the end-user expectation that the polymeric, product contact materials will meet the appropriate regulatory requirements, including US Pharmacopeia (USP) and the European Pharmacopeia (EP). It is expected that the suppliers will have evaluated and characterized the materials and will share this data.

Research and review of data and knowledge

Existing informationand literature Clinical studies

In vitro and in vivononclinicalresearch

Product understanding Process understanding

Critical QualityAtributes

Critical ProcessParameters

TPPsQAs to be takeninto considerationfor designing theprocess toguarantee quality,efficacy, and safetyof the drug

Atributes nothaving a negativeimpact on quality,efficacy, and safetyof the drug

CQAs

NonCriticalQuality Atributes

RiskAnalysisFMEA

CPPs DOEs

Definition ofprocess designspaceNon-Critical

Process Parameters

Production at scale

Risk management

Control strategy

Continuous verification

Testing

Process control

Definition ofprocess designspace

FIG. 35.6 This figure shows how the TPPs, Critical Quality Attributes (CQAs), and Critical Process Parameters (CPPs) are interconnected to define a control strategy permitting optimal risk management. Quality Attributes (QAs) are assessed with regard to their impact on drug quality, efficacy, and safety, respectively, to identify the Critical Quality Attributes. The identified CQAs are further assessed in a Failure Mode Effect Analysis (FMEA) to identify the associated Critical Process Parameters (CPPs) which are verified in Design of Experiment (DOE) studies. The identification of CQAs, as-sociated CPPs, and the result of DOE studies permits the definition of a control strategy for monitoring and controlling the production process at scale.

Page 9: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 729

35.3.6 Extractable & Leachables of SUT

Plastic materials release chemical components under certain conditions. This is not new knowledge: baby bottles were found to release bisphenol A (BPA) and polyvinyl-chloride (PVC) and plasticizers such as di(2-ethylhexyl)phthalate (DEHP). The purpose of extractable studies is to characterize single-use materials with regard to the components they release under exaggerated exposition conditions after sterilization as a function of pH and polarity of different solvents [24]. Execution of extractable studies is a key characterization step of single-use materials that determines their suitability for use in biopharmaceutical applications. In this way, extractable studies provide a chemical fingerprint of the single-use component under exaggerated conditions.

The purpose of leachable studies is to characterize single-use materials with regard to the components they release under real process conditions normally used to produce the product. These are considerably less harsh than the conditions used during extractable studies. Leachable studies identify a subset of the releasable components identified in extractable studies. Furthermore, leachable studies determine if cross-reactions occur between leachable components and the active pharmaceutical ingredient (API). Leachable studies serve to determine the total load of chemical components released by single-use components present in the final drug product and administered to the patient together with the API [24].

TABLE 35.3 An Example of Are the Typical Key Parameters of a TPP (Target Product Profile) for Manufacturing of a Specific mAb

Parameter Example Target Product Profile

Drug product Excipient formulationProtein quantity per vialProtein concentration per vialVial volumeVial materialWay of administrationDosePharmacokinetic behaviorStabilitySolubility

Liquid600 mg30 mg/mL20 mLGlassIntravenous4 mg/kg/weekHalf-life of several daysStable at 2–8°CSoluble at high concentrations

Active pharmaceutical ingredient (API) or bulk drug substance

Integrity of mAbAppearancepHActivityPurityOsmolalityProduct qualityHost cell proteinResidual DNAProtein A leachateSterility

Folded correctly, presence of two light and two heavy chainsClear>5.5 and <6.5Binding of specific antigenMonomer >95%250–350 mOsmo/kgGlycan, fucose specifications<1.5 ng/mg<17 pg/mg<20 ng/mgSterile

TABLE 35.4 Listed Are the Quality Attributes of a Typical mAb Segregated in Three Areas of Variants of the mAb Produced, the Purity of the Drug Substance and the Final Drug Product

Quality attributes MAb Variants Purity of Drug Substance Final Drug Product

Quality attributes AggregationConformationC-terminal lysineDi-sulfide bondsDeaminated isoformsFragmentationGlycosilationThioether bondsOxidation

EndotoxinViral contaminationDNAHost cell proteinsProtein AAntibioticsCell cultivation media componentsPurification buffer components

Particulate matterColorOsmolaritypHMAb concentrationVolume

Page 10: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

730 SECTION | VI Industrial Process Design

35.3.7 Particulate Matter With SUT

Drug products administered to patients must meet limits for the presence of particulate matter [25–28]. The biopharmaceu-tical industry distinguishes between visible and subvisible particulate matter. Contamination of drug product with particu-late matter can be relatively well controlled by application of appropriate filtration steps within the process architecture. Furthermore, visual inspection of the drug product after filling is routinely carried out. Particulate control is not possible in processes that do not allow appropriate filtration steps such as live viral vaccines or after adsorption to alum, for example. Process architecture and technology choices have to reduce the potential of particulate matter contamination of the drug product to a minimum. For these latter applications, compliance with this regulatory demand requires knowledge of the manufacturing conditions of single-use components, their conditions of use in the real process, and how to decide on the appropriate process architecture and technology choices. Single-use components for drug product have to be manufactured under conditions that reduce generation of particulate matter and keep contamination of the final product to a minimum.

Ensuring sterility of the drug product requires knowledge of the manufacturing and sterilization conditions of single-use components and appropriate tests for leaks at the manufacturer/assembler site. It requires knowledge of their conditions of use in the real process, and of how to decide on the appropriate process architecture and technology choices with regard to ensuring maintenance of integrity and impact management.

35.3.8 Standards for Single-Use Technology

The single-use community has been working to generate guidance and draft standards for SUT implementation (Table 35.5; [31,32]). Groups such as the BioPhorum Operations Group (BPOG) disposable working group in collaboration with the supplier organization Bioprocess Systems Alliance (BPSA) and Single-Use Technology Assessment Program (SUTAP,

TABLE 35.5 Example of End-User Expectations of Suppliers for Qualification Attributes for SUT: Polymeric Product Contact Materials [24,25,29,30]

Qualification Package Sections Details

1 Biocompatibility USP 87 biological reactivity tests, in vitroUSP 88 biological reactivity tests, in vivo

2 Mechanical properties Tensile strength, elongation at break, seal strength, leak test

3 Gas/vapor transmission ASTM D3985: oxygenASTM F1249: water vapor

4 Compendial testing for plastics (USP <661>)

5 Compendial testing for EVA E.P.3.1.7.: EVA for containers and tubing (required only if material is EVA)

6 Animal origin control E.P.5.2.8. on TSE-BSE

7 TOC analysis

8 pH/conductivity If product is sensitive to pH

9 Extractables and leachables Supplied by vendor

10 Chemical compatibility Supplied by vendor

11 Protein adsorption studies

12 Endotoxin testing

13 Sterilization validation When user requires sterility assurance

14 Container closure integrity (CCI) Film-to-film weld integrity, port-to-film weld integrity, port-to-tubingConnection integrity, tubing-to-connector integrityOverall bag integrity (e.g., absence of pinholes)

15 Particulates USP <788>, EP 2.9.19 For downstream applications (e.g., form/fill)

16 Calibration of embedded instrumentation For embedded single use sensors

Page 11: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 731

http://www.sutap.org) are working to generate aligned guidance documents. This will then be drafted into single-use technology standard via a recognized consensus standards body such as the American Society for Testing and Material (ASTM). The creation of standards, requiring alignment across end-users and suppliers has shown to be a long drawn-out process (nine years so far for one standard). The newly formed group SUTAP is working to accelerate the drafting and the approval process of the standards. The group is providing dedicated writing resources to progress the preparation of the standards as defined in Table 35.6 and shepherd these through the ASTM balloting consensus process. It has become clear, since the creation of SUTAP, that a comprehensive framework of standards, test methods and specifications is required to cover all aspects associated with single-use technology.

Experience has shown that use of different extractable testing protocols across suppliers has added complexity to the point of hindering SUT implementation. The complexity makes the total extractables assessment difficult when a single-use bioreactor bag contains components from multiple suppliers. Harmonized testing methods used by the industry would help to alleviate this issue [33]. The BPOG, BPSA, and ASTM are working to create a framework of guidances, test methods, and specifications to simplify this activity for straightforward SUT implementation.

35.3.9 Securing the Single-Use Technology Supply Chain and Change Control

A key area of attention is SUT selection and sourcing. While advancements in single-use technology have taken place, it is still a maturing technology which requires continued improvements. As the new technologies develop, it is recommended to have a backup (dual) source approach with functional similarity to minimize the risk if supply chain interruptions occur. Technical comparison evaluations with relevant drug manufacturing process streams should be completed using multiple supplier offerings before a purchase is made. For example, a recent end-user comparison of SUBs from a range of suppliers showed a range of operational issues due to differing design-related approaches [34]. The selection criteria should not only include the technical performance and cost assessment but also the ability of the supplier to support supply chain risk, qual-ity/validation strategy, and sterility assurance. Building a strong collaborative partnership with the supplier is also highly important. Traditionally for stainless steel facilities, the end user controlled the cleaning/sterilization, change control, and integrating testing of its stainless steel equipment, while only outsourcing polymeric components such as O-rings, gaskets, filter cartridges, and valve diaphragms. Manufacturing with SUT requires the end-user to entrust the supplier to support the capability and quality of the SU component. This is complicated by a multitiered network of suppliers back to the resin supplier used in the bag film. As an example, there are over two hundred single use components for a typical single use bioreactor bag.

It is the supplier's responsibility to notify end-users of specific component changes and composition changes. This is a cumbersome process hampered by inconsistent expectations for both the supplier and the end-user. The BPOG and BPSA are working to align the notification strategy and the current issues have been well defined [35]. They are now working to define an improved streamlined approach for implementation. The suppliers are working to improve transparency of changes all the way back to the resin suppliers. This will work to avoid previous issues such as the leaching of the cyto-toxic compound bis(2,4-di-tert-butylphenyl) phosphate from Wave bioreactor bags that caused inhibitory impact to CHO cell growth [36]. This generated a significant supply chain burden for end-users who needed to urgently seek alternative bioreactor bag supplies, as well as requiring significant resources to complete the investigation. This experience stresses the importance that end-users consider using a dual source approach to single-use technology to minimize the risk of supply chain unreliability [37].

35.3.10 Single-Use Technology Reliability and Improvement

It is recommended that end-user groups dedicate engineering teams to assess component engineering for design and de-ployment of single-use systems and their manifold components [38]. The engineering team defines optimal designs to minimize the number of different SUT manifold designs while maximizing manifold component strength and integrity. The team also supports the supply chain management activities. A number of end-user companies have begun to move away from expensive custom design single-use manifolds to a library of standardized building blocks that can be shared across a global network [38]. This consolidated approach eases the supply chain risk and reduces reliance on complex customized expensive SU components and manifolds. These are sourced from multiple suppliers so the building blocks are functionally compatible. An industry-aligned catalog of SUT, with input from end users and suppliers, is being compiled [39]. Some suppliers such as GE have pursued similar strategies as shown by the ReadyCircuit approach.

End-users are still experiencing unacceptable bioreactor failure rates due to a range of issues such as seam failures, film punctures, and handling errors. Additional work is needed to improve robustness, but also training and transport packaging

Page 12: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

732 SEC

TIO

N |

VI Industrial Process D

esign

TABLE 35.6 Efforts to Generate SUT Standards in the SU Community

ASME-BPE ASTM E55 BPOG BPSA ELSIE PDA PQRI USP SUTAP

Extractable & Leachable (E&L)

X X X X X TR66 X X X

SUT supply chain X X TR66

Change notification

X X TR66

Change control X X TR66

Particulates in SUT X X X TR66 X

SUT system integrity

X X TR27 X

Connectors X TR66

SUT design verification

X X TR66

Applications X TR66

General Definitions TR66

Modified from J.D. Vogel, M. Eustis, The single use watering hole. Bioprocess. Int. 13(1) (2015) 2–12.

Page 13: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 733

improvements are needed. New films continue to be developed with improved strength in both the film and welds [40]. Once the ASTM single-use technology standards framework is established, the SUTAP group proposes implementation of a single-use assessment program to classify the quality of SU components from the suppliers. This classification would be analogous to the automotive star safety-rating program to simplify the risk assessment of end-users with regard to choos-ing the appropriate single-use technology for any given application [32]. This will provide assessment and classification of single-use components following scientific and risk based testing procedures. This will enable accredited comparative data.

Connectivity (connections between single use systems, components, tubing and bags) improvements are needed to en-hance workflow efficiency as current connection practices are limited to tube welding or one use only aseptic connections. A cost-effective connector that enables multiple on/off/on connections is required. Given the concerns of supply chain reliability, end-users would like standard single-use designs to enable the disposables to be interchangeable between the suppliers. This could be done through cooperation and collaboration with the supplier groups, such as industrial forums, BPOG disposable end-user working, and BPSA supplier groups. Integrating bio-analytical PAT sensors into the single-use bioreactors will improve workflow efficiency and will lead to better understood, robust processes. Improvements of true single-use sensors for pH, dO2, dCO2, and capacitance with defined strategies for calibration and qualification are needed.

Whilst the overall use of single-use enabled processes has shown to provide positive environment life cycle assessment over existing stainless steel, end-users must be aware of the increased disposable and waste packaging that SUT creates. The BPSA has begun a sustainability initiative in 2017 to provide best practices for plastics disposable, recycling and re-duction in SUT packaging. This has been spurred by multiple end-user and supplier collaborations that have demonstrated the feasibility of multiple approaches. These include the feasibility of plastic waste collection and reprocessing to second-ary products such as carpet tiles, the incineration of waste plastics with energy cogeneration and packaging recycle and reuse programs to minimize land fill. A number of end-user companies are applying sustainability design into their process and facility concepts.

35.3.11 Biosafety Applications

The industry can benefit from aligned guidance on the application of single-use technology for vaccine production for biosafety levels above the general use (BSL 1) to higher levels (BSL 2, 2+, 3+). This would highlight the need for widen-ing implementation of smart automated solutions to minimize manual operations, closing the process, and segregating the operator from the process. Developing passive pressure relief systems for single-use would also provide a final level of security in the unlikely event of full automation system failure. Secondary containment systems for the complete volume of the bioreactor is a key requirement and systems have been created with negative pressure control to protect the opera-tor from any aerosol release [41]. An improvement in single-use leak detection systems would help to accelerate process implementations.

35.4 CASE STUDIES OF NEXT GENERATION PROCESSES ENABLED BY SUT

The continued efforts to improve and update processes have multiple drivers including cost reduction, and ensuring supply while assuring product quality. The potential levels of improvements are ranging from simple troubleshooting through debottlenecking to developing a new state-of-the-art process. Single-use technologies can support improving process architectures and increase process efficiency. SUT can be successfully integrated independently of the age of the process architecture in use. The following examples will provide case studies of how the process architecture of a technologically outdated manufacturing process could be modernized and be fully compliant to meet today's regula-tory requirements. These considerations are critical when anticipating to innovate an existing process with single-use technology.

35.4.1 Vaccine Manufacturing

In contrast to modern biologics, most of today's administered vaccines have been developed since the 1930s and the early 1990s of the last century [42]. It is therefore not surprising that these products are produced using manually intensive and low technology manufacturing processes, not in line with today's expectations and requirements. The vaccine industry's use of outdated manufacturing processes and technologies to produce products with impeccable safety records in a highly- regulated environment has created a high threshold of risk adversity to any process change [43,44]. Furthermore, since these mature products and processes have never been characterized with today's analytical capabilities, product knowledge and

Page 14: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

734 SECTION | VI Industrial Process Design

process understanding are insufficient to enable adoption of new technologies, modernization, and improvement through compliant change control procedures. Given the importance of vaccines today, the biopharmaceutical industry together with the regulatory authorities have to rethink how to enable change to state-of-the-art technologies for old manufacturing processes. This must be done in a framework that takes into account the scientific and financial implications without com-promising product efficacy and patient safety [45].

In contrast to the well-established platform mAb manufacturing processes, the vaccine industry lacks established platforms. This resulted from the long development history and the wide variety of vaccine families and technologies (live viral attenuated or inactivated vaccines; subunit vaccines, polysaccharide vaccines, conjugated, etc.). It is the absence of product knowledge and process understanding that limits change in the biopharmaceutical industry. Clinical trials are of-ten required to justify the process change due to the absence of sufficient product knowledge and process understanding. In this way, the weaker the product knowledge and process understanding (the technical bridge), the more likely clinical trials will be necessary to validate the process change (the clinical bridge). This interrelationship is visualized in Fig. 35.7 showing the interdependencies between product knowledge, process understanding, clinical bridge, and technical bridge, respectively.

Understanding this interdependency helps define the appropriate scientific development strategy to assess the impact of the change. Investment in the product and process understanding enables moving the “cursor” as far as possible toward the technical bridge and thus limiting the need for extensive and costly clinical trials. Furthermore, the execution of thorough scientific characterization work can form the basis and provide the relevant data for future process changes in addition to future troubleshooting and problem solving activities [46].

It is absolutely clear that increasing the level of product knowledge and process understanding is a work-intensive and time-consuming process, and that these nonnovel work packages are not very appealing to scientific personnel. Recent experience, however, has proven that this investment can result in massive benefits with regard to overall process robustness and performance, reduction of waste streams and lead time, and an increase in productivity per batch by 50%, respectively [46,47].

Case Study 1: A Second-Generation Live Viral Vaccine Manufacturing Process Based on Modern SUTIn this case study, the impact of transitioning from a traditional open manufacturing process (that required several hundred manual aseptic connections) to a new single-use closed process is evaluated. The live viral vaccines available today are typi-cally produced using a multistep adherent cell cultivation process typically performed in T-flasks and cell factories or roller bottles. A typical generic live viral vaccine manufacturing process architecture using cell factories is shown in Fig. 35.8. When using T-flasks or roller bottles, scale-up requires significant open manual operations in a biosafety cabinet (ISO-5, class 100 environment) until sufficient cell mass is created to inoculate a forty-tray cell factory scale (CF-40). These cell factories can provide 25,280 cm2 of growth surface and be manipulated on the process floor, outside of the Biosafety Cabinet, either individually using a manual handler or as bundles of 4× CF-40s on a rack using robotized equipment. Preparation of CF-40s as well as connection of tubing manifolds for fluid transfer during cultivation is carried out inside a mobile laminar flow hood using aseptic connectors or tube welding. The production step is carried out typically in batches of several CF-40 racks. Furthermore, given the fact that some live viral vaccines cannot be sterile filtered at the point of fill (as a consequence of the virus size), the entire manufacturing process has to be carried out aseptically by personnel-intensive production steps executed in a two-shift setup in a grade B air classification environment. Taken together, the use

Level of product knowledge and process understanding

Clinical bridgeTechnical bridge

FIG. 35.7 Example of the interdependency between technical bridge, clinical bridge and the level of product knowledge and process understanding. The vertical blue cursor can be positioned based on the level of product knowledge and process understanding for the case being considered. It becomes obvious that increasing the level of product knowledge and process understanding (moving the cursor to the left darker green) via thorough scientific work enables reliance on the technical data set (technical bridge) and reduces the requirement for clinical trials (clinical bridge) to assess impact of the process change.

Page 15: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 735

of CF-40s, the requirement for a fully aseptic process and the need for several hundred sterile connections significantly increases the COGs.

This live viral vaccine manufacturing processes reflects the state-of-the-art technologies used at the time of their devel-opment during the 1960s and 1970s. It requires major labor-intensive efforts to execute these in full GMP compliance with many operator manipulations. Addressing the risks associated with the current process architecture, new, two state-of-the-art

Step

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

Culture 1: CF10 Culture 2: CF10 Culture 3: CF40

Infection 1:CF10

Infection 2:CF10

Infection 3:CF40

Current process architecture

Vial thaw Vial thaw Vial thaw

T175

T175

CF10

CF10

CF10

Centrifugation

Centrifugation

Sonication

Centrifugation

Isolation ofsupernatant

Centrifugation

Isolation ofsupernatant

Storage of drugsubstance

Infection 1: Xpansion MBP 200

Harvest

Decantingsupernatant and

resuspensionof pellet

Decantingsupernatant and

resuspensionof pellet

Second generationprocess architecture

option 1

T175

T175

Xpansion MPB 200

Culture 1: Xpansion MBP 200

Harvest

Centrifugation

Decanting supernatant andresuspension of pellet

Centrifugation

Decanting supernatant andresuspension of pellet

Sonication

Centrifugation

Isolation of supernatant

Centrifugation

Isolation of supernatant

Storage of drug substance Storage of drug substance

Second generationprocess architecture

option 2

Infection 1: iCELLis 500

T175

T175

Xpansion MPB 200

Culture 1: iCELLis 500

Harvest

Centrifugation

Decanting supernatant andresuspension of pllet

Centrifugation

Sonication

Centrifugation

Isolation of supernatant

Isolation of supernatant

Centrifugation

Decanting supernatant andrususpension of pllet

FIG. 35.8 Live viral vaccine production: comparison of current processes to potential second-generation manufacturing process options. The red and green frames identify which type of atmosphere control is required to carry out the step in full GMP compliance. Red frames designate ISO 5 require-ments, while green frames designate ISO 7 requirements. For the current process architecture, the infection 3 step is designated in red as CF40s have to be manually connected to media, buffer, and inoculum for operation, although the incubation step could be carried out in an ISO 7 environment. The significant difference in overall requirement of ISO 5 grade atmosphere normally present in a biosafety cabinet or under a mobile laminar flow hood shows why the current process is carried out in a Grade B (ISO 6) environment, while the new process architectures can be carried out in Grade C (ISO 7) environments equipped with ISO 5 (Grade A) bio-safety cabinets. The figure outlines the benefit offered by the second-generation options in eliminating the need of multiple virus infection steps.

Page 16: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

736 SECTION | VI Industrial Process Design

process architecture options that incorporate the latest generation SUT (at two different scales) are shown in Fig. 35.8. These second-generation manufacturing processes are entirely closed and require only manipulation in a Biosafety Cabinet until larger scale, closed Xpansion and/or iCELLis systems (PALL) can be inoculated [48–52]. From then on, the entire upstream manufacturing process is closed. In addition to reducing the number of sterile connections that have to be established, the new process architectures require minimal manipulation by operators, only one-shift operation and only a grade C (ISO-7, class 10,000) environment with the exception of the initial semiopen cell cultivation steps. Furthermore, a very welcomed side-effect of these second-generation process architectures is that need for multiple virus infection steps is eliminated. These new process architectures enable therefore significantly increased productivity while reducing effort. To simplify the transi-tion to a second-generation process, the cell substrates, the cell cultivation medium, and the virus seeds are not changed.

35.4.2 Monoclonal Antibody Production

Today, mAbs are manufactured using a generic platform process, and utilization of single-use components has become relevant with regard to limiting CAPEX spending, trade-offs between scale-up and build-out, shortening construction/qualification timelines, and applicability to multiproduct facilities, respectively [53]. Over the last two decades, major ef-forts have been invested to eliminate bottlenecks in mAb manufacturing processes that were mainly limited by upstream productivities. Manufacturing of required quantities was met by building large scale traditional stainless-steel facilities, comprising multibioreactor trains (up to 8× 25,000 L production vessels with corresponding inoculum trains). These large facilities typically cost multiple hundred million USD in capital and operational expenditure over their entire utilization cycle. Development of high productivity cell lines has driven productivities from milligrams to multiple grams per liter. This evolution has primed review of manufacturing strategies towards smaller scales (typically 15,000 down to 2000 L and below) permitting utilization of cost saving single-use technologies as outlined earlier.

Case Study 2: The Impact of Changing From Resin-Based Column Chromatography to Membrane-Based PurificationThe major bottlenecks in the downstream process of capacity and throughput have relied on large expensive batch col-umns. Prepacked columns can support clinical production needs but struggle to fore fill production capacity for single column batch processing. In this case study, we outline the impact of changing from resin-based column chromatography to membrane-based chromatography. The pore diffusion mass transfer limitations of resin beads result in long column bed heights, slow linear velocities (100–150 cm/h) and dramatically oversized columns [54]. Membrane absorption provides a high throughput approach due to the convective mass transport with minimal pore diffusion. In addition, lower bed-height-to-diameter ratios can be achieved with smaller pressure drops than column chromatography. This enables flow rates of the order of 1–10 membrane volumes per minute, which in turn could enable processing of volumes using a relatively small device. The multilayered porous membrane systems are amenable to fully single-use formats such as, capsules, or car-tridges that have the potential to simplify operations. A variety of functionalized membrane systems are now commercially available including anion exchange (AEX). This is a key polishing step effective for scavenging trace amounts of impuri-ties using flow through mode [55]. An example of the improved throughput and loading provided by membrane processes is shown in Table 35.7, which compares conventional resins to novel membrane systems. This includes the membrane hydrogel (Natrix HD-Q) that is a porous polyacrylamide functionalized 3D hydrogel supported on an inert web backbone [2]. This is different than the conventional membranes in which the ligands are attached to supporting matrixes. The inter-connected pore structure provides a large and highly accessible surface area for protein binding and high permeability. The performance evaluation shown in Table 35.7 shows that mAb loadings up to 10,000 g/L can be achieved with acceptable impurity removal [2]. This provides up to 60× greater loading capacity than traditional resin systems. The residence time is up to 40× faster than resin processes providing significantly faster loading. This results in a reduction of necessary injec-tions and subsequently a reduction in buffer usage, up to 40×, when compared to resin chromatography [16]. This combina-tion of factors reduces cost and the faster processing enables increase a facility output by 4–5 batches/year for a traditional fed-batch CHO mAb process [16]. The suppliers have shown capability to produce the membrane systems in convenient single-use configurations using class 6 compliant materials with acceptable extractable/leachable profiles. These disposable membrane alternatives to resin chromatography are now part of many end-user's tool box for purification polishing options.

Case Study 3: Developing Next Generation Single Use Automated Continuous BioprocessingIn this approach, a low volume disposable perfusion bioreactor (<500 L scale) is integrally connected to each of the sub-sequent unit operations to create a closed, single-use connected continuous end-to-end process from the bioreactor to

Page 17: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 737

UF/DF concentration step, as shown in Fig. 35.9. The continuous process is controlled by an overarching control system that controls the individual methods of each unit operation. The flow rate of the perfusion bioreactor can be adjusted to vary the concentration of product produced by the bioreactor which is then purified through the subsequent continuous purifica-tion process. This allows responsive adjustments to the product production rate (kg/day) to adjust to any changes of patient market demand in a “supply-on-demand” approach. Process analytical technology (PAT) including adaptive process control and product attribute control will ensure product quality into this continuous format. The process analytical tools will also enable real-time release of the product and significantly reduce the QC burden. It is hoped that the traditional assay set of over 25 offline release assays can be potentially replaced by the majority of real-time analysis on the process floor. This will speed QC and QA from multiple weeks to a few days. Therefore, the benefits from this approach come from multiple aspects: the use of multicolumn chromatography technology to use buffer and resin use, novel purification membranes ap-plied to increase throughput [2,16], and smaller capital equipment leading to smaller facility size facility and lower capital equipment cost. The automated processing and real-time analytics leads to reduced labor and inventory costs while provid-ing faster lead-time in getting drugs to the patient.

The feasibility of this single-use automated continuous processing approach have recently been demonstrated at lab scale for greater than 50 days [4,53]. The proof of concept included a perfusion (Titer > 1 g/L day at 1 vvd) integrated with continuous chromatography using disposable multicolumn chromatography (BioSMB, Pall), a viral pH inactivation flow

TABLE 35.7 Comparison of AEX Polishing Membrane to Resin Performance for a Multiple mAbs Using a Range of Loadings

AEX mAb Loading (g/L)Residence Time HCP (ppm) DNA (ppm)

Res Protein A (ppm)

ResinPoros HQ

mAb 1a 100 4 min 15 <LOQ <LOQ

MembraneMustang Q

mAb 1a 1000 6 s 18 <LOQ 1

MembraneNatrix HD-Q

mAb 1a 1000 3 s 22 0.003 1

MembraneNatrix HD-Q

mAb 2b 3000 6 s 18 2.0 <LOQ

MembraneNatrix HD-Q

mAb 3c 10,000 6 s 23 0.03 <LOQ

Feed streams:a Feed = HCP 2300 ppm, feed DNA 2 ppm.b Feed = HCP 100 ppm, DNA 2 ppm.c Feed = HCP 110 ppm, DNA 0.03 ppm.Modified from H. Ying, M. Brower, D. Pollard, D. Kanani, R. Jacquemart, B. Kachuik, J. Stout, Advective hydrogel membrane chromatography for mAb purification, Biotechnol. Prog. 31(4) (2015) 974–982.

Current state : sequential batch processing Fixed drug outputFed Batch,2000 – 20,000 scaleConventional QC releaseLarge inventory

Future process: continuous processing

Adjustable drug outputResponsive to demand Real time QC releaseSmall inventory

Higher productivityReduced buffer

Shorter lead timesReduced cost of inventory

storage

FIG. 35.9 Potential benefits of continuous bioprocessing compared to traditional sequential batch processing for CHO mAb production.

Page 18: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

738 SECTION | VI Industrial Process Design

through loop, and polishing membrane hydrogel steps, respectively. The automation process control used an overarching control system (Delta V) providing supervisory control to each individual unit operation that utilized the existing local con-trol of the supplier software. Automated actuator systems allow active fluid management control between subsequent unit operations that integrate with surge bags to prime unit operations and enable product accumulation during any process in-terruptions. Redundant bioburden filtration steps are activated when single-use pressure sensors detect fouling buildup [53].

In the facility of the future for biologics, it is anticipated that flexible single-use technologies will provide a toolbox for a variety of process architectures such as fed-batch, perfusion, and semi/fully continuous processing to meet the demand of a particular molecule modality. Furthermore, integration of drug substance production with adaptive drug product manufac-turing filling systems, enabled by single-use, has the potential to further reduce manufacturing cycle times.

35.5 FUTURE STATE SUMMARY

Efforts to ease the implementation of single-use technology will clearly enable a wider and faster expansion of lower cost flexible and sustainable processes. One of the key foundational efforts is the generation of SUT industry-aligned standards. There are an encouraging effort and enthusiasm from end-users and suppliers to drive the standards to comple-tion in the coming years. This will have significant impact for the bioprocessing industry in defining expectations of SUT implementation for everyone including suppliers, end-users, and regulators. This effort will ultimately lead to benefits to patients. The adoption of standard equipment such as industry-harmonized single-use manifold designs will continue to expand. This should allow the industry to move to automated manifold production with molding technology. This will drive down costs, reduce lead times and potentially lead to stronger, more robust manifold systems with fewer failures. It is hoped this focus will expand to other single-use technologies such as standard mixing and bioreactor bags designs. Suppliers will continue to improve their supply chain transparency and robustness. This includes implementing the im-proved change control procedures that are harmonized between suppliers and end-users. The implementation of integrated (connected) processing and automated continuous processing enabled by disposable technology will continue to expand into commercial production using modular facilities of the future. This will be key as we move towards an era of more responsive and sustainable manufacturing that can react to rapid changes in biologics and vaccines demand and drive to low inventory practices. In the coming years, it is expected that real-time release of biologics will be implemented that will be enabled by improved single-use sensors, online analytics and parametric models. This will allow more robust processes through improved understanding and product attribute control. The combination of all these efforts will lead to faster adoption of SUT and continue to further lower the costs of bioprocessing that will ultimately lead to wider access of life saving drugs to patients. This will need continued effort to sustain and enhance the close collaboration between technology providers and end-users.

ACKNOWLEDGMENTSThe authors wish to acknowledge Günter Jagschies for providing us the opportunity to prepare a comprehensive overview on the benefits, ap-proaches, challenges, and needs associated with single-use technology for the new Process Handbook. In addition, the authors wish to thank the Merck Single Use Network for assistance with this chapter. This includes Mark Petrich, Jeff Johnson, Dave Moyle, Mark Brower, Chris Smalley, Sabrina Restrepo, and Chris Gross.

REFERENCES [1] H. Levine, R. Stock, H. Hummel, S. Dana Jones, Efficient flexible facilities for the 21st century, Bioprocess Int. 5 (2012) 10–15. [2] H. Ying, M. Brower, D. Pollard, D. Kanani, R. Jacquemart, B. Kachuik, J. Stout, Advective Hydrogel Membrane chromatography for mAb purifica-

tion, Biotechnol. Prog. 31 (4) (2015) 974–982. [3] M. Bisschops, M. Brower, The impact of continuous multichrom chromatography on biomanufacturing efficiency, Pharm. Bioprocess. 1 (4) (2013)

361–372. [4] M. Brower, Y. Hou, D. Pollard, Monoclonal antibody continuous processing enabled by single use, in: G. Subramanian (Ed.), Continuous Processing

in Pharmaceutical Manufacturing, Wiley VCH, Weinheim, 2015, pp. 255–296. [5] A. Seegar, D. Estappe, Production costs in biotech, in: R. Eibl, D. Eibl (Eds.), Single Use Technology in Biopharmaceutical Manufacture, Wiley

VCH, Weinheim, 2011, pp. 351–361. [6] J. Pollock, S.V. Ho, S.S. Farid, Fed-batch and perfusion culture processes: economic, environmental, and operational feasibility under uncertainty,

Biotechnol. Bioeng. 110 (1) (2013) 206–219. [7] A. Pralong, P. Galliher, When the process becomes the product: single use technology and the next bio manufacturing paradigm, Biopharm Int. 26

(4) (2013) 30–34.

Page 19: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

Single-Use Technology Implementation For Biologics and Vaccines Production Chapter | 35 739

[8] K.I. Kaitin, J.A. DiMasi, Pharmaceutical innovation in the 21st century: new drug approvals in the first decade, 2000–2009, Clin. Pharmacol. Ther. 89 (2) (2011) 183–188.

[9] K. Alvin, J. Ly, R. Condon, G. Keil, X. Mou, J. Ye, D. Pollard, Technology improvements to accelerate process development of biologics, Am. Pharm. Rev. 10 (2013) 1–9.

[10] R. Bareither, M. Goldfeld, C. Kistler, A. Tait, N. Bargh, R. Oakeshott, K. O’Neill, L. Hoshan, D. Pollard, Automated disposable small scale bioreac-tor for high throughput process development: implementation of the 24-bioreactor array, Pharm. Bioprocess. 3 (3) (2015) 185–197.

[11] A. Luitjens, J. Lewis, A. Pralong, Single use biotechnologies and modular manufacturing environments invite paradigm shifts in bioprocess develop-ment and biopharmaceutical manufacturing, in: G. Subramanian (Ed.), Biopharmaceutical Production Technology, Wiley VCH, Weinheim, 2012, pp. 817–857.

[12] A. Pralong, H.L. Levine, J. Lilja, A. Gassvik, H. Hummel, Paradigm shift for vaccine manufacturing facilities: the next generation of flexible modu-lar facilities, Eng. Life Sci. 13 (3) (2014) 244–253.

[13] V. Warikoo, R. Godawat, K. Brower, S. Jain, D. Cummings, E. Simons, T. Johnson, J. Walther, C. Hwang, W. Zhou, K. Konstantinov, Integrated continuous production of recombinant therapeutic proteins, Biotechnol. Bioeng. 109 (12) (2012) 3018–3029.

[14] J. Walther, C. Hwang, K. Konstantinov, R. Godawat, Y. Abe, A. Sinclair, The business impact of an integrated continuous biomanufacturing platform for recombinant protein production, J. Biotechnol. 213 (2015) 3–12.

[15] R. Hernandez, Continuous manufacturing: a changing processing paradigm, Biopharm Int. 28 (4) (2015) 25–29. [16] D. Pollard, M. Brower, Y. Abe, A. Lopes, A. Sinclair, Standardised economic cost modeling for next generation mAb production, Bioprocess Int. 14

(8) (2017) 14–23. [17] G.P. Pisano, Knowledge, integration, and the locus of learning: an empirical analysis of process development, Strateg. Manag. J. 15 (1994) 85–100. [18] CMC Biotech Working Group, A-Mab: A Case Study in Bioprocess Development, 2009. Available from: www.ispe.org/pqli/a-mab-case-study-

version-2.1, (12 June 2014). [19] C. Challener, Securing the single-use supply chain, Pharm. Technol. 38 (12) (2014) 24–26. [20] European Commission, EudraLex—Good Manufacturing Practice (GMP) Guidelines, vol. 4, 2003. Available from: http://ec.europa.eu/health/

documents/eudralex/vol-4_en, (10 March 2017). [21] FDA, April 01 2016. CFR Title 21 Part  210, Available from: http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.

cfm?CFRPart=210&showFR=1 (10 March 2017). [22] FiercePharma, Pharma manufacturing consent decrees: 16 black holes, Available from: http://www.fiercepharma.com/special-report/

pharma- manufacturing-consent-decrees-16-black-holes (25 July 2016). [23] Food and Drug Administration, 2014. Warning letter to Amgen Inc. issued on 1/27/14, Available from: http://www.fda.gov/iceci/enforcementactions/

warningletters/2014/ucm385288.htm (11 March 2015). [24] A.  Mire-Sluis, Extractables and leachables, Bioprocess Int. (2011). 01 February, Available from: http://www.bioprocessintl.com/business/

cmc-forums/extractables-and-leachables- 311844/, (11 April 2016). [25] USP, 2011. (788) Particulate matter in injections, Available from: www.usp.org (13 March 2014). [26] USP, 2014. (790) Visible particulates in injections, Available from: www.usp.org (13 June 2015). [27] USP, 2015. (1790) Visual inspection of injections, Available from: www.usp.org (16 September 2015). [28] J.G. Shabushnig, Visible particles: regulatory and compendial requirements, 2014. Available from: https://www.pda.org/docs/default-source/website-

document-library/chapters/presentations/ireland/visible-particles—regulatory-and-compendial-requirements. pdf?sfvrsn=6, (12 September 2014).[29] M. Furbush, M. Felo, Polymeric films for single use applications, Pharm. Process 29 (8) (2014) 14–16.[30] P. Daniels, Building blocks for single use bioreactor components, Bioprocess Int. 12 (9) (2014) 18–23. [31] J.D. Vogel, M. Eustis, The single use watering hole, Bioprocess Int. 13 (1) (2015) 2–12. [32] A. Pralong, Standards for single use technology- a must for success, Biopharm. Asia 5 (2014) 8–20. [33] E. Mahajan, T. Ray-Chaudhuri, J.D. Vogel, Standardization of single use components extractable studies for industry, Pharm. Eng. 32 (3) (2012) 1–3. [34] R. Krishnan, H. Chen, Overview of single use technologies for biologics production, Am. Pharm. Rev. 15 (3) (2012) 15–19. [35] T. White, K. Ott, Management, notification and documentation of single use systems change orders: challenges and opportunities, Bioprocess Int.

10 (2015) 33–39. [36] M. Hammond, L. Marghitoiu, H. Lee, L. Perez, G. Rogers, Y. Nashed-Sameul, A cytotoxic leachable compound from single use bioprocess equip-

ment that causes poor cell growth performance, Biotech. Prog. 30 (2) (2014) 332–337. T.E. Stone, An overview of risk assessment strategies for extractables and leachables, BioPharm. Intl. 25(1) (2012), 39–45.

[37] J. Briggs, N. Chandarana, Exploring options of dual sourcing of single use components, Bioprocess Int. 12 (11) (2014) 26–29. [38] M. Petrich, Design and deployment strategy for single use components and assemblies, Am. Pharm. Rev. 2 (2013). Available From: http://www.

americanpharmaceuticalreview.com/Featured-Articles/151947-Design-and-Deployment-Strategy-for-Single-Use-Components-and-Assemblies/. (15 April 2014).

[39] D. Wolton, L. Heaven, S. McFeaters, M. Kodilkar, Standardization of disposables design: the path forward for a potential game changer, Bioprocess Int. 2 (2015). Available From: http://www.bioprocessintl.com/category/manufacturing/page/7/. (12 January 2016).

[40] E. Vachette, C. Fenge, J.-M. Cappia, L. Delaunay, G. Greller, B. Magali, Robust and convient single use processing: superior strength and flexibility of flexsafe bag, Bioprocess Int. 12 (5) (2014) 23–25.

[41] P.M. Galliher, G.L. Hodge, M. Fisher, Environmental containment systems, 2010. Available from: https://www.google.com/patents/US7819934, (13 March 2012).

[42] S.A. Plotkin, W.A. Orenstein, P.A. Offit, Vaccines, sixth ed., Elsevier, New York, NY, 2013.

Page 20: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

740 SECTION | VI Industrial Process Design

[43] P.A. Offit, The Cutter Incident: How America’s First Polio Vaccine Led to the Growing Vaccine Crisis, Yale University Press, New Haven, CT, 2007. [44] European Center for Disease Prevention and Control, 03.02.2016. Updated risk assessment on shortage of acellular pertussis-containing vac-

cines affecting some EU/EEA member states, Available from: http://ecdc.europa.eu/en/press/news/_layouts/forms/News_DispForm.aspx? ID=1352&List=8db7286c-fe2d-476c-9133-18ff4cb1b568&Source=http%3A%2F%2Fecdc.europa.eu%2Fen%2FPages%2Fhome.aspx (13 March 2015).

[45] Center for Disease Control & Prevention, 07.12.2015, Current Vaccine Shortages & Delays, Available from: http://www.cdc.gov/vaccines/vac-gen/shortages/(12 March 2016).

[46] A. Pralong, Biosimilar vaccines—a potential pathway Curr. Chromatography (in press). [47] A. Luitjens, E. van Corven, Affordable vaccines for developing nations: facts or fiction? Pharm. Bioprocess. 1 (2) (2013) 129–131. [48] Pall Corporation, Xpansion® Multiplate Bioreactor System, Available from: http://www.pall.com/main/biopharmaceuticals/product.

page?lid=hw7uq21i (16 June 2016). [49] Pall Corporation, iCELLis® Bioreactor, Available from: http://www.pall.com/main/biopharmaceuticals/product.page?lid=hw7uq21l (16 June

2016). [50] J.C. Drugmand, N. Vertommen, S. Knowles, F. Debras, J. Castillo, Linear Scalability of Virus Production in the Integrity iCELLis Disposable Fixed-

Bed Bioreactor from Bench-Scale to Industrial Scale, Available from: http://www.pall.com/pdfs/Biopharmaceuticals/iCELLis-Poster_Industry%20Scale_A4_Printer-File.pdf (16 June 2016).

[51] A. Onraedt, P. Lefebvre, M. Kremer, Virus Production From Bench Scale To Industrial Scale: New Technologies That Intensify, Simplify Virus Manufacturing And Increase Overall Process Efficiency, Available from: http://www.pall.com/pdfs/Biopharmaceuticals/15-6201_iCellis_Virus_POS.pdf (16 June 2016).

[52] K. Lehmann, S. Dubois, N. Havelange, S. Beyer, K. Lipinski, CASE STUDY: Fixed-bed, Disposable Bioreactor Technology (iCELLisTM) for Virus/Vaccine Production, Available from: http://www.pall.com/pdfs/Biopharmaceuticals/Case-Study-Fixed-bed-Disposable-Bioreactor-Technology-iCELLis-for-Virus-Vaccine-Production.pdf (16 June 2016).

[53] D.J. Pollard, D. Richardson, M. Brower, Progress towards automated single use continuous monoclonal antibody manufacturing via the protein refinery operations lab, in: G. Subramanian (Ed.), Continuous Processing in Pharmaceutical Manufacturing, Wiley VCH, Weinheim, 2016, pp. 255–296.

[54] U. Gottschalk, Disposables in downstream processing, Adv. Biochem. Eng. Biotechnol. 115 (2009) 171–183. [55] A.A. Shukla, B. Hubbrard, T. Tressel, S. Guhan, D. Low, Downstream processing of monoclonal antibodies, J. Chromatogr. B 848 (2007) 28–39.

Page 21: Single-Use Technology Implementation For Biologics and ...download.xuebalib.com/oe3Uz0mVT8Z.pdf · year. In addition, there is increasing uncertainty to the prediction of new pipeline

本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP

图书馆。

图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具