34
Richard D. Braatz Novartis-MIT Center for Continuous Manufacturing MIT Center for Biomedical Innovation Department of Chemical Engineering Controlling Pharmaceutical Quality

Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Richard D. Braatz

Novartis-MIT Center for Continuous Manufacturing

MIT Center for Biomedical Innovation

Department of Chemical Engineering

Controlling Pharmaceutical Quality

Page 2: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

2 | Novartis/MIT, All Rights Reserved

Background on QbD approach

Process development involves:

• Define the target product profile

• Identify the critical quality attributes (CQAs)

• Select an appropriate manufacturing strategy

• Implement a control strategy

This talk is on controlling quality in integrated pharmaceutical and biologics manufacturing

Page 3: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

3 | Novartis/MIT, All Rights Reserved

Why Integrated Manufacturing?

Reduce contact between

biology/chemistry & personnel

Continuous operation has

the potential to

• Increase product quality

• Increase yields

• Enable new drug product

formulations (e.g., thin films)

• Reduce scale-up risks

• Reduce footprint

Page 4: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

4 | Novartis/MIT, All Rights Reserved

Outline

Control Systems for Integrated Continuous Operations

Relationship with Design Space

Application to Biologics Manufacturing

Page 5: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

5 | Novartis/MIT, All Rights Reserved

Integrated Control Strategy

for Continuous Manufacturing

Tight integration of continuous operations can result in disturbances propagating downstream, unless their effects are suppressed by an integrated control strategy

The strategy must optimize the overall plant operation instead of only isolated units (i.e., need plantwide control)

D2

D1

R2

R1LT LT

M1 M2 S1

C1 C2

W1

P1

P2

P3

P4

P5

P9

P7 P8

P10LT

LT LT

C3 C4

P18 P19

P6

LT

P11

P13

P14

P17

P20

P12

P15

P16

LT LT

P22

P24

S3

S4 W2

M3 M4

M5

S5S6 E1 CS

CAT

A

B

S1

S2

S3

S1

PU1

S1

PU2

PU4

PU3

S1

S1

S1

C

D

E

S1

EX1

EX2

S2

FP

s1

s2

s3

s4

s5

s6

s7

s10

s9

s8

s11 s12s13

s14s15

s16

s18

s19

s20

s17

s21

s22

s23

s24

s25

s26s27 s28 s29

s30

s31

s32

s33

s35

s34

s36

s37

s38

s40 s41

s43

s45

s46

s47s39

s44

Q

LT

P21

P21S1

CT FT

FT

CT

DT

CT

CT

CT

FT

DT CT

STTCFT CT

s42

FT

Page 6: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

6 | Novartis/MIT, All Rights Reserved

Plantwide Control of Continuous Manufacturing

Challenges

• Many connected unit operations

• Very fast to slow processes

• Multi-purpose plants with short development time

• Alignment with regulatory requirements

Approach adapted from the chemical industry

• Employs systematic and modular design of plantwide control strategies for continuous manufacturing facilities

• Experimentally demonstrated on continuous pilot plant

R. Lakerveld, B. Benyahia, P.L. Heider, H. Zhang, A. Wolfe, C. Testa, S. Ogden, D.R. Hersey, S. Mascia, J.M.B. Evans, R.D.

Braatz, and P.I. Barton. The application of an automated control strategy for an integrated continuous pharmaceutical pilot plant.

Organic Process Research & Development, in press.

D2

D1

R2

R1LT LT

M1 M2 S1

C1 C2

W1

P1

P2

P3

P4

P5

P9

P7 P8

P10LT

LT LT

C3 C4

P18 P19

P6

LT

P11

P13

P14

P17

P20

P12

P15

P16

LT LT

P22

P24

S3

S4 W2

M3 M4

M5

S5S6 E1 CS

CAT

A

B

S1

S2

S3

S1

PU1

S1

PU2

PU4

PU3

S1

S1

S1

C

D

E

S1

EX1

EX2

S2

FP

s1

s2

s3

s4

s5

s6

s7

s10

s9

s8

s11 s12s13

s14s15

s16

s18

s19

s20

s17

s21

s22

s23

s24

s25

s26s27 s28 s29

s30

s31

s32

s33

s35

s34

s36

s37

s38

s40 s41

s43

s45

s46

s47s39

s44

Q

LT

P21

P21S1

CT FT

FT

CT

DT

CT

CT

CT

FT

DT CT

STTCFT CT

s42

FT

Page 7: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

7 | Novartis/MIT, All Rights Reserved

A continuous pilot plant

S. Mascia, P.L. Heider, H. Zhang, R. Lakerveld, B. Benyahia, P.I. Barton, R.D. Braatz, C.L. Cooney, J.M.B. Evans, T.F. Jamison,

K.F. Jensen, A.S. Myerson, and B.L. Trout. End-to-end continuous manufacturing of pharmaceuticals: Integrated synthesis,

purification, and final dosage formation. Angewandte Chemie, 52(47):12359-12363, 2013

Page 8: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

8 | Novartis/MIT, All Rights Reserved

D2

D1

R2

R1LC LC

M1 M2 S1

C1 C2

W1

LC LC

C3 C4

LC

LCLC

S3

S4W2

M3 M4

M5

S5S6 E1 CS

CAT

A

B

S2

S1

S3

S1

PU1

PU2

PU4

PU3

S1

S1

S1

C

D

E

S1

EX1

EX2

FP

LC

S1

FCsp

FT

FCsp

spsp

sp

FCsp

CCFT

CTFC

FCsp

TC

sp

spsp

FCsp

DCsp

sp

S1

RC CCsp

DC

LC

sp

1

CATA B I

1 2 BPI C I P

2I E API

A continuous pilot plant

• First-principles dynamic models were built for each unit operation (UO) as they were developed

• Models were validated and then placed into a plant-wide simulation

• Plant simulation used to design UO & plantwide control strategy

Page 9: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

9 | Novartis/MIT, All Rights Reserved

Model-based Design of a Plant-wide Control Strategy

2. Optimizing control

objectives Level 1 – Total I/O

Level 4 – Detailed

Control tasks

(CQA, CPP)1

Level 2 – Intermediates

1. Stabilizing control

objectives (local)

Structure of plant-wide

control strategy

Level 3 – Recycles

Control tasks

(CQA, CPP)2

Control tasks

(CQA, CPP)3

Control tasks classified into

optimizing and stabilizing

Hierarchical decomposition

• Reduces complexity

• Exploits separation of time scales

Page 10: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

10 | Novartis/MIT, All Rights Reserved

Parametric Sensitivities Used to Evaluate Relationships Between CPPs and CQAs

0 0 0, , ,ft t t x t t x - State variables

- Input variables

- Output variables (CQAs)

for all feedback control loops

0

1( ) ( ) ( ) ,

( ) ( )

t

MV p D

I

SP CV

du t K t d

dt

t y y t

( ) ( ), ( ), , ,

( ) ( ), ( ), ,

( ), ( ), , 0,

dx t f x t u t p t

dt

y t g x t u t p t

h x t u t p t

( )

( )

( )

x t

u t

y t

p - Parameters (PPs)

Use sensitivities (Si,j) to identify

causal relations CPPs-CQAs:

• Direction and order of magnitude

• Guide selection of automated control loops

• Determined from process simulation (could use DOE)

,i

i j

j

yS

p

Page 11: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

11 | Novartis/MIT, All Rights Reserved

Example Sensitivity Results: Level 1: Total Inputs & Outputs

Page 12: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

12 | Novartis/MIT, All Rights Reserved

Sensitivities Used to Evaluate Dynamic I/O Relationships, to Assess Controllability and Disturbance Propagation

0

0.2

0.4

0.6

0.8

1

0 50 100 150

No

rmal

ize

d S

en

siti

vity

Time / h

API dosage

Impurity content

Model-based sensitivity of two final product quality variables

with respect to feed flow rate of reactant

Page 13: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

13 | Novartis/MIT, All Rights Reserved

D2

D1

R2

R1LC LC

M1 M2 S1

C1 C2

W1

LC LC

C3 C4

LC

LCLC

S3

S4W2

M3 M4

M5

S5S6 E1 CS

CAT

A

B

S2

S1

S3

S1

PU1

PU2

PU4

PU3

S1

S1

S1

C

D

E

S1

EX1

EX2

FP

LC

S1

FCsp

FT

FCsp

spsp

sp

FCsp

CCFT

CTFC

FCsp

TC

sp

spsp

FCsp

DCsp

sp

S1

RC CCsp

DC

LC

sp

1

CATA B I

1 2 BPI C I P

2I E API

A continuous pilot plant

Met all purity

specs in Summer

2012

Page 14: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

14 | Novartis/MIT, All Rights Reserved

Outline

Control Systems for Integrated Continuous Operations

Relationship with Design Space

Application to Biologics Manufacturing

Page 15: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

15 | Novartis/MIT, All Rights Reserved

Comparing Design Space and Feedback Control (both are consistent with QbD principles)

Design-space methods:

• Control strategy based on operation

within a fixed parameter space

• Applicable to each continuous

process unit operation

• More complicated to apply to an

entire continuous pharmaceutical

manufacturing plant

Feedback methods:

• Control strategy based on

feedback to a “parameter space”

• Easier to scale up

• Design space does not need to

be exhaustively validated a priori

• Necessary for continuous

manufacturing

Page 16: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

16 | Novartis/MIT, All Rights Reserved

There are Multiple Strategies for Ensuring That a Particular CQA Spec is Satisfied

1. Direct measurement of the CQA

2. Prediction of the CQA based on a first-principles

model that is fed measurements of related variables

and is running in parallel with operations

3. Prediction of the CQA based on an empirical or semi-

empirical model (e.g., response surface map, PLS

model) that is fed measurements of other variables

4. Operation of the CPPs to lie within a design space, that

is, some specified set shown in offline studies to provide

assurance

A.S. Myerson, M. Krumme, M. Nasr, H. Thomas, and R.D. Braatz. Control systems engineering in

continuous pharmaceutical manufacturing. Organic Process Research & Development, in press.

Can use

in closed-

loop

feedback

control

strategy

Open-

loop

strategy

The smaller the number, the easier to justify real-time release

Page 17: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

17 | Novartis/MIT, All Rights Reserved

Outline

Control Systems for Integrated Continuous Operations

Relationship with Design Space

Application to Biologics Manufacturing

Page 18: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Product manufacturing:

Allston, MA USA

Product QC: Haverhill, UK

Fill/finish: Waterford, IE

Cerezyme patients distributed worldwide

Manufacturing Biologic Drugs Today

18

Page 19: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Towards Biomanufacturing on Demand (BioMOD)

Design Requirements Patient

19

BioMOD capabilities

• Enable flexible methodologies for genetic engineering/modification of microbial strains to synthesize multiple and wide-ranging protein-based therapeutics

• Develop flexible & portable device platforms for manufacturing multiple biologics with high purity, efficacy, and potency, at the point-of-care, in short timeframes (<24 hours), when specific needs arise

• Include end-to-end manufacturing chain (including downstream processing) within a microfluidics-based platform

• Focus on currently approved therapeutics by FDA (i.e., no drug discovery)

Page 20: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Column Tanks

1 2

Downstream

Affinity Chromatography

Sterile Media

Yeast Inoculum

Crude Product Hold Tanks

Upstream

1 2 1 2

Polishing Membrane #1 Tanks

Polishing Membrane #1

Polishing Membrane #2

Waste Waste Waste

Polishing Membrane #2 Tanks

UF/DF Membrane

Waste

Concentration

Identity

Purity

pH, DO, T

Potency

Safety

Sterility

Analytics

Purified Product for Quality Testing

Multivariate

Model

OK for Release

Not OK

Final

Product

On-line Reactor Control

Perfusion

Fill

Elute

Real-Time Process

Data

Integrated and Scalable Cyto-Technology

(InSCyT) Biomanufacturing Platform

20

Page 21: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Rationale for Pichia pastoris as

a Microbial Host for Biosimilar Products

Advantages from a regulatory perspective

• Many products on market or in late-stage development

(including one Phase I target)

• Reduced risk for viral contamination

• Human-like post-translational modifications (folding, glycosylation, etc.)

Technical benefits

• Genetically stable organism

• High-density cultivation (culture volume >70% biomass)

• High yields of secreted proteins (up to ~15 g/L)

• Limited host cell protein (HCP) profile (eases burden on downstream)

• Amenable to lyophilization

21

Page 22: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Column Tanks

1 2

Downstream

Affinity Chromatography

Sterile Media

Yeast Inoculum

Crude Product Hold Tanks

Upstream

1 2 1 2

Polishing Membrane #1 Tanks

Polishing Membrane #1

Polishing Membrane #2

Waste Waste Waste

Polishing Membrane #2 Tanks

UF/DF Membrane

Waste

Concentration

Identity

Purity

pH, DO, T

Potency

Safety

Sterility

Analytics

Purified Product for Quality Testing

Multivariate

Model

OK for Release

Not OK

Final

Product

On-line Reactor Control

Perfusion

Fill

Elute

Real-Time Process

Data

Integrated and Scalable Cyto-Technology

(InSCyT) Biomanufacturing Platform

On-line quality

analytics

22

Page 23: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Recall Quality-by-Design Approach

Process development involves:

• Define the target

product profile

• Identify the

critical quality

attributes (CQAs)

• Select an

appropriate

manufacturing

strategy

• Implement a

control strategy

23

Page 24: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Plant-wide Control Approach

• Characteristics of InSCyT

– Many connected unit operations

– Many discrete operations

– Multi-product plant

– Alignment with regulatory

requirements

• QbD approach adapted from chemical industry

– Employing systematic and modular design of plantwide

control strategies for production-scale manufacturing facilities

– Using numerical algorithms that can handle discrete operations

and multiple products

Column Tanks 1 2

Downstream

Affinity Chromatography

Sterile Media

Yeast Inoculum

Crude Product Hold Tanks

Upstream

1 2 1 2

Polishing Membrane #1 Tanks

Polishing Membrane #1

Polishing Membrane #2

Waste Waste Waste

Polishing Membrane #2 Tanks

UF/DF Membrane

Waste

Concentration

Identity

Purity

pH, DO, T

Potency

Safety

Sterility

Analytics

Purified Product for Quality Testing

Multivariate

Model

OK for Release

Not OK

Final Product

On-line Reactor Control

Perfusion

Fill

Elute

Real-Time Process

Data

24 R. Lakerveld, B. Benyahia, P.L. Heider, H. Zhang, A. Wolfe, C. Testa, S. Ogden, D.R. Hersey, S. Mascia, J.M.B. Evans, R.D. Braatz, and P.I. Barton. The application of an automated control strategy for an integrated continuous pharmaceutical pilot plant. Organic Process Research & Development, in press.

Page 25: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Application to Biologic Drug Production

• Build first-principles

dynamic models for

each unit operation (UO)

• Design control system

for each UO to meet

“local” material attributes

• Evaluate performance in

simulations and propose

design modifications as needed

• Implement and verify the control system for each UO

• Design and verify plantwide control system to ensure

that the CQAs are met 25

Page 26: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Application to Biologic Drug Production

• Build first-principles

dynamic models for

each unit operation (UO)

• Design control system

for each UO to meet

“local” material attributes

• Evaluate performance in

simulations and propose

design modifications as needed

• Implement and verify the control system for each UO

• Design and verify plantwide control system to ensure

that the CQAs are met 26

Page 27: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Column Tanks

1 2

Downstream

Affinity Chromatography

Sterile Media

Yeast Inoculum

Crude Product Hold Tanks

Upstream

1 2 1 2

Polishing Membrane #1 Tanks

Polishing Membrane #1

Polishing Membrane #2

Waste Waste Waste

Polishing Membrane #2 Tanks

UF/DF Membrane

Waste

Concentration

Identity

Purity

pH, DO, T

Potency

Safety

Sterility

Analytics

Purified Product for Quality Testing

Multivariate

Model

OK for Release

Not OK

Final

Product

On-line Reactor Control

Perfusion

Fill

Elute

Real-Time Process

Data

Integrated and Scalable Cyto-Technology

(InSCyT) Biomanufacturing Platform

27

Page 28: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Local “UO” Control for Bioreactor Unit Operations

28

Page 29: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

29

Local “UO” Control: Microscale Controlled Cell Culture

K.S. Lee et al., Lab on a Chip, 11, 1730-1739 (2011)

Page 30: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

OD

600 (

Cell C

on

ce

ntr

ati

on

)

POD 3

POD 2

POD 1

POD 0

Outgrowth Transition Production

Time, h

Reproducible Microbioreactor Cultivations

30

Page 31: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

K.S. Lee et al., Lab on a Chip, 11, 1730-1739 (2011)

Local “UO” Control: Microscale Controlled Cell Culture

31

Page 32: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Application to Biologic Drug Production

• Build first-principles

dynamic models for

each unit operation (UO)

• Design control system

for each UO to meet

“local” material attributes

• Evaluate performance in

simulations and propose

design modifications as needed

• Implement and verify the control system for each UO

• Design and verify plantwide control system to ensure

that the CQAs are met 32

Page 33: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Acknowledgments (Novartis-MIT)

Paul I. Barton

Bernhardt L. Trout

Allan T. Myerson

Klavs F. Jensen

Timothy F. Jamison

Richard Lakerveld (Delft)

Brahim Benyahia (Loughborough)

33

Page 34: Controlling Pharmaceutical Quality - PQRI · M1 M2 S1 C1 C2 W1 P1 P2 P3 P4 P5 P9 P7 P8 P10 LT LT LT C3 C4 P18 P19 P6 LT P11 P13 P14 P17 P20 P12 P15 P16 LT LT P22 P24 S3 S4 W2 M3 M4

Acknowledgments (Bioman)

Chris Love

Rajeev Ram

Anthony Sinskey

Timothy Lu

Michael Strano

Jongyoon Han

James Leung

GK Raju

Stacy Springs

Paul Barone

Kerry Love

Philippe Mouriere

Nate Cosper

William Hancock

Lisa Bradbury

Ralf Kuriyel

Russell Newton

Susan Dexter

Steve Cramer

Pankaj Karande

34