principles, practice and guided evolution of biologics

24
1 Principles, Practice and Guided Evolution of Biologics Manufacturing Platforms David Beattie R&D Director, Biotech Process Solutions EMD Millipore, a division of Merck KGaA CMC Forum Europe 2011 Barcelona

Upload: others

Post on 26-Jan-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

1

Principles, Practice and Guided Evolution of Biologics Manufacturing Platforms

David BeattieR&D Director, Biotech Process Solutions

EMD Millipore, a division of Merck KGaA

CMC Forum Europe 2011Barcelona

2

What is a Platform?

“A method, equipment, procedure or work practice that may be applied across multiple products under development or manufacture.”

Examples:• An expression system Chinese Hamster Ovary (CHO) cells

• A screening system high throughput based on robotics

• An analytical method imaged capillary electrophoresis

• A drug product formulation citrate + sucrose + Tween

• A mode of cell culture perfusion culture

• A purification unit operation affinity chromatography

• A complete process comprising multiple operations Platform DSP

3

What is a Platform?

“A method, equipment, procedure or work practice that may be applied across multiple products under development or manufacture.”

mAb AmAb BmAb CmAb DmAb E

A single company making a variety of related drugs

4

What is a Platform?

“A method, equipment, procedure or work practice that may be applied across multiple products under development or manufacture.”

Flu AFlu BFlu CFlu DFlu E

Flu AFlu BFlu CFlu DFlu E

Flu AFlu BFlu CFlu DFlu E

An industry making a variety of related drugs

5

What are the Benefits?

• Development effort and overall costs reduced

• Time to clinic and therefore to market reduced

• Process improvements leveraged over many products

• Economies of scale for equipment, components and raw materials

• Failure rates during GMP manufacturing reduced over time due to accumulated process experience

• Procedures for in-process and batch release testing become routine,implying risk of errors reduced

• Faster turnaround in multiproduct facilities

• Submissions of INDs/IMPDs during early stage development are expected to be facilitated more readily

• Platform processes are suitable for a modular validation approach, which in turn leads to reduced efforts and costs

• Overall benefit for industry, health care system and patient

6

Definition of PlatformBaseline purification and analysis strategy for future mAb products

Standardized materials and methods

IPC testing matrix and methods with minimum specifications

Purpose of PlatformReduce the time required for DSP development activities

Minimize material demand lead times

Maximize the utilization of process improvements

Establish a standard process for benchmarking to industry expectations

Merck Serono DSP Platform Project

7

Unit Op Identification

Unit operations selected from historical success and common practicesPolish

Chromatography

Protein AChromatography

Capture

Low pHhold

Anion ExchangeChromatography

Bulk DrugSubstanceFormulation

Nanofiltration

UF/DF

To achieve optimal versatility, the platform consists of a primary and alternativeapproach for each operation

Bulk DrugSubstanceFormulation

Flexibility!

8

Platform Process Control

Standardized IPC testing matrix

Promote reliable data comparison

Establish expectations for impurity removal for each operation

Establish generic specifications for drug substance

Analytical Method

IPC Monitoring

BDS Specification

PROA

LOW pH

AEX

POL

VRF

UFDF

BDS

Conc. (rPA) X X X N/A

Conc. (A280nm) X X X X X X X Product Specific

Purity (SEC) X X X X X X X >95% Monomer

Purity (PAGE) X X X X X X X >95% Product Bands

Isotypes (ICE) X X X X Compare to RS

HCP (ELISA) X X X X X X X <50ppm

DNA (qPCR) X X X X X <10pg/mg

FLrPA (ELISA) X X X X <5ppm

Bioactivity X 60-140% of RS

9

Case Study: Anti-CD19

The first application of the generic process

Primary chosen for Capture, Low pH and Virus Reduction FiltrationAlternative chosen for Polish for removal of aggregate to meet purity specificationAlternative chosen for UFDF based on yield, placed before VRF due to high NaCl

cHA

MabSelect Xtra

Low pHhold

SartobindQCharged

Membrane

Bulk DrugSubstance

Formulation

CPV

30KdRC

Process Recovery

Product Purity

Residual Aggregate Process Impurities

>65% >99.5% <0.5%All meet spec

or <LOQ

Repeated for other mAbs Success! (?)

FIMProcess

Development

cGMP API prod.

DP prod.

Pre-clin supply APIrelease

Tox material

Clone selected

Supportive stability studies

Formulation selection

DP release

cDNA available

Pre-MCB tested

Clone generation

Process check

Pre-clinical formulation

Analytical dev

Analytical check

regulatory documentation

MCB available

Stability studies

IND IMPD

Formulation check

Year 2Year 2

Aug OctSep NovJunApr JulMay FebDec MarJan Aug OctSepJunApr JulMay

Q2 Q3 Q4 Q1 Q2 Q3

Year 1Year 1

Feb MarJan

Q1

Nov Dec

Q4

Ph 1

Production

Benefits not realized in Isolation

FIMProcess

Development

cGMP API prod.

DP prod.

Pre-clin supply APIrelease

Tox material

Clone selected

Supportive stability studies

Formulation selection

DP release

cDNA available

Pre-MCB tested

Clone generation

Process check

Pre-clinical formulation

Analytical dev

Analytical check

regulatory documentation

MCB available

Stability studies

IND IMPD

Formulation check

Year 2Year 2

Aug OctSep NovJunApr JulMay FebDec MarJan Aug OctSepJunApr JulMay

Q2 Q3 Q4 Q1 Q2 Q3

Year 1Year 1

Feb MarJan

Q1

Nov Dec

Q4

Ph 1 Production

And Release

Cell Line

Generation

Analytical

Development

Formulation

Development

Benefits not realized in Isolation

30+ mosto FIM !

Lesson Learned: full benefit only from “Total Chain” Platform

Bulk DrugSubstanceFormulation

Purification Platform

Several sorting rounds

Cloning

Sorted cells growth

Staining and sorting highest fluorescing

cells

Stable pool

Several sorting rounds

CloningCloning

Sorted cells growthSorted cells growth

Staining and sorting highest fluorescing

cells

Staining and sorting highest fluorescing

cells

Staining and sorting highest fluorescing

cells

Stable pool

Cell Line Generation Platform

Analytical Platform

Formulation Platform

18 mosto FIM !

Benefits not realized in Isolation

Early Phase Development replaced by Platform Fit and Adaptation

Involve all the Stakeholders

Hydroxyapatite: calcium-phosphate crystal with a zero point charge at neutral pH Extremely sensitive to acidic conditions

Extremely powerful for polishing mAbs !

In non-Platform process, column degradation occurs after cycling

Visible loss of resin and bed cracking

At lab scale, up to 50 cycles run without issue

But, rapid and limiting increase in back-pressure only at production scale

Lesson Learned—manufacturing colleagues need a strong voice in setting Platform and non-Platform information can be relevant

Cycle 1 Cycle 20 Cycle 50

0.0

0.5

1.0

1.5

2.0

2.5

3.0

2 4 6 8 10 12 13 14 15 16 17 18 19 20 21

Batch No

Col

umn

inle

t pre

ssur

e (B

ar)

load

elution

14

Using the Platform to create a Viral Clearance Knowledge Base

Development History and Prior Process Knowledge are core to QbDJust as we use accumulated performance knowledge from multiple

projects to design the Platform, we also look to accumulated virus clearance data to optimize the effectiveness

0

5

10

15

20

25

30

5 5.5 6 6.5 7 7.5 8 8.5 9

pH

Con

duct

ivity

(mS/

cm) MLV> 4

MLV<4MVM>4MVM<4SV40>4SV40<4Reo3>4Reo3<4

Clearance factors in function of the pH and the conductivityObjectives:Improve design space using viral clearance as response factorAmortize clearance data over many projectsReduce validation workload during early clinical phasesEfficient use of data accumulated on generic platform process(es)

Reconcile two apparently contradictory needs of process development: reducing cost and duration of development while simultaneously increasing process understanding

15

Platform Evolution

“A platform process should not be regarded as a static, long-lasting procedure. In contrast, it should be reviewed within agreed life-cycle periods and undergo changes by controlled implementation of improvements and uptake of innovations.”

Moreover, “Processes of the Future” can and should be based on the learnings from the Platform

more Knowledge Management

mAb Chromatography Steps at 2 g/L

Protein A Virus Hold Cation Exchange

Hold Tank Anion Exchange

Hold Tank

Input12,0002 g/L

2,850 L8.2 g/L

3,280 L7.1 g/L

1,530 L13.7 g/L

3,060 L6.9 g/L

3,060 L6.9 g/L

Output2,850 L8.2 g/L

3,280 L7.1 g/L

1,530 L13.7 g/L

3,060 L6.9 g/L

3,060 L6.9 g/L

VirusUF/DF

Plant Design: 5,000L Tank 5,000L Tank 5,000L Tank

Platform Evolution: Process Compression

Protein A Virus Hold Cation Exchange

Hold Tank Anion Exchange

Hold Tank

Input12,0005 g/L

7,200 L8 g/L

8,300 L7 g/L

4,580 L11 g/L

9,160 L5.7g/L

9,160 L5.7g/L

Output7,200 L

8 g/L8,300 L

7 g/L4,580 L11 g/L

9,160 L5.7g/L

9,160 L5.7g/L

VirusUF/DF

Plant Design: 5,000L Tank 5,000L Tank 5,000L Tank

mAb Chromatography Steps at 5 g/L

Platform Evolution: Process Compression

What if you could remove intermediate hold tanks by direct loading of elution steps?

Protein A Direct loading of Pro A

elution and Virus Hold

on CEX media column

Cation Exchange Direct

loading of CIEX elution

on to membraneadsorber

Anion Exchange

Hold Tank

Input12,0005 g/L

7,200 L8 g/L

4,800 L10 g/L

4,800 L10 g/L

Output

7,200 L8 g/L

4,580 L11 g/L

4,800 L10 g/L

VirusUF/DF

Plant Design: 5,000L Tank

Platform Evolution: Process Compression

PoolHCP(ppm)

Leached Protein A

(ppm)

Aggregate (%)

Yield(%)

Feed (MAb spike/non-expressing CHO)

295,000 NA NA

Protein A Pool (ProSep Ultra Plus)

140 12 0.5 98

Cation Exchange Pool(Eshmuno S)

64 < 2* 0.3 83

Anion Exchange Pool(ChromaSorb)

< 3* < 2* 0.3 96

Overall Connected Process

< 3* < 2* 0.3 78

Overall Control Process (with intermediate holds)

< 3* < 2* 0.4 86

Performance Results from Direct Elution Connected Process

Platform Evolution: Process Compression

Comparable performance for Connected Process

Chromatography Step

Column Volumes (CV)

Bufferon-column

inactivation

Virus Conc.(Log)

on-column inactivation

BufferTraditional

Process

Virus Conc.(Log)

TraditionalProcess

CIEX EQ 450mM Sodium Acetate,

25mM NaCl, pH 5.4

50mM Sodium Acetate, 25mM

NaCl, pH 5.4

Load6

(load density = 50mg/mL)

Protein A Pool 6.5 Protein A Pool 6.5

CIEX Wash 350mM Sodium Acetate,

150mM NaCl,pH 3.5

≤ 2.450mM Sodium Acetate, 25mM

NaCl, pH 5.4

≤ 2.7

Low pH Hold

0.5(Total step time =

0.5hr, for on-column

inactivation)

50mM Sodium Acetate, 150mM NaCl,

pH 3.5≤ 2.8 Not performed N/A

CEX Elution 6*50mM Sodium Acetate,

w/NaCl,pH 5.4

≤ 2.250mM Sodium

Acetate, w/NaCl,pH 5.4

2.6

Post Elution 3EQ and EQ w / 1M NaCl

Pooled≤ 2.2

EQ and EQ w/ 1M NaCl Pooled

3.5

Platform Evolution: Process Compression

Virus Clearance Results from Direct Elution Connected Process

Below LOD for Wash, Hold, Elution and Strip

0

5

10

15

20

25

MAb A (1

1.4 g/L

)

MAb B (5

.2 g/L

)

MAb C Lot

A (18 g

/L)

MAb C Lot

B (18 g

/L)

MAb D Lot

A (26 g

/L)

MAb D Lot

A (13 g

/L)

MAb D Lot

B (15 g

/L)

MAb E Lo

t A (1

4 g/L)

MAb E Lo

t A (7

g/L)

MAb F (6

g/L)

MAb G (6

g/L)

MAb H (4

.5 g/L)

MAb I (4

.5 g/L

)

MAb K (1

0 g/L)

MAb L (1

0 g/L)

MAb M (6

g/L)

MAb N (2

g/L)

MAb O (2

g/L)

MAb Q (1

5 g/L)

MAb R (8

g/L)

MAb S (1

4 g/L)

MAb T (5

g/L)

MAb U (1

2 g/L)

MAb X (1

7.6 g/L

)

MAb Y (2

2.5 g/L

)

MAb Z (1

g/L)

Mas

s C

apac

ity (k

g/m

2)

Avg. 6.5 kg/m2 Mass Capacity over Protein concentrations 1-26 g/LClearly not robust !

Evolution: Improving Robustness

Evaluation of Virus Removal Fitration for multiple mAbs

0

2000

4000

6000

8000

10000

12000

14000

16000

MAb1MAb2MAb3MAb4MAb5MAb6MAb7MAb8MAb9MAb1

0MAb1

1MAb1

2MAb1

3MAb1

4

MAb

Mas

s th

roug

hput

(g/m

2)

Vpro alone

V-Pro with CEX shield filter

Evolution: Improving Robustness

Used Platform Knowledge to use a Cation Exchange shield filter to remove aggregates that were primary limiter of capacity

23

Where are We Now?

• Development effort and overall costs reduced

• Time to clinic and therefore to market reduced

• Process improvements leveraged over many products

• Economies of scale for equipment, components and raw materials

• Failure rates during GMP manufacturing reduced over time due to accumulated process experience

• Procedures for in-process and batch release testing become routine,implying risk of errors reduced

• Faster turnaround in multiproduct facilities

• Submissions of INDs/IMPDs during early stage development are expected to be facilitated more readily

• Platform processes are suitable for a modular validation approach, which in turn leads to reduced efforts and costs

• Overall benefit for industry, health care system and patient

Acknowledgements

EBE Concept Paper Team

inc. Enda Moran and Piers Alin

Pascal Valax

Jim Nolan

Neil Soice

Richard Pearce