+ All Categories
Home > Documents > Regulatory Challenges in Bioassay Practices

Regulatory Challenges in Bioassay Practices

Date post: 11-Feb-2016
Category:
Upload: nibaw
View: 37 times
Download: 0 times
Share this document with a friend
Description:
Regulatory Challenges in Bioassay Practices. Tim Schofield Managing Director & Head of Non-Clinical Services Arlenda, Inc. tim.schofield.arlenda.com Presented at the 35 th Annual Midwest Biopharmaceutical Statistics May 22, 2012, Ball State University, Muncie, Indiana. Outline. - PowerPoint PPT Presentation
Popular Tags:
23
Regulatory Challenges in Bioassay Practices Tim Schofield Managing Director & Head of Non- Clinical Services Arlenda, Inc. tim.schofield.arlenda.com Presented at the 35 th Annual Midwest Biopharmaceutical Statistics May 22, 2012, Ball State University, Muncie, Indiana
Transcript
Page 1: Regulatory Challenges in Bioassay Practices

Regulatory Challenges in Bioassay Practices

Tim SchofieldManaging Director & Head of Non-Clinical Services

Arlenda, Inc.

tim.schofield.arlenda.com

Presented at the 35th Annual Midwest Biopharmaceutical Statistics

May 22, 2012, Ball State University, Muncie, Indiana

Page 2: Regulatory Challenges in Bioassay Practices

Outline

What is a bioassay?

Bioassay guidelines

Regional differences in bioassay practices

US viewpoint on bioassay practices

QbD for analytical methods

Page 3: Regulatory Challenges in Bioassay Practices

What is a bioassay?ICH definition and requirements

Bioassay (Biological Assay) – ICH Q6B (paraphrased) Definition: The measure of the biological activity using a suitably

quantitative biological assay (also called potency assay or bioassay), based on the attribute of the product which is linked to the relevant biological properties.

A valid biological assay to measure activity should be provided by the manufacturer. Examples of procedures used to measure biological activity include:

• Animal-based biological assays, which measure an organism's biological response to the product

• Cell culture-based biological assays, which measure biochemical or physiological response at the cellular level

• Biochemical assays, which measure biological activities such as enzymatic reaction rates or biological responses induced by immunological interactions

Page 4: Regulatory Challenges in Bioassay Practices

Potency definition Specific ability or capacity of the product, as indicated by

appropriate laboratory tests or by adequately controlled clinical data obtained through the administration of the product in the manner intended, to effect a given result. [21 CFR §600.3 (s)]

Tests for potency Tests for potency shall consist of either in vitro or in vivo tests, or

both, which have been specifically designed for each product so as to indicate its potency in a manner adequate to satisfy the interpretation of potency given by the definition in § 600.3 (s) of this chapter. [21 CFR § 610.10]

What is a bioassay?FDA requirements

Page 5: Regulatory Challenges in Bioassay Practices

Measurement of activity rather than mass Specific to the mechanism of action

Usually reported as relative potency to a reference standard

Highly variable 10% to 50% RSD (versus 1% to 2% RSD for chromatographic assays)

Biology rather than chemistry

Resource intensive Time as well as materials

Usually performed on samples in a complex matrix Therapeutic proteins purified from cell culture

Vaccines produced in living systems

What is a bioassay?Distinguishing properties

Page 6: Regulatory Challenges in Bioassay Practices

Bioassay guidelines

Originally USP <111> and EP 5.3 <111> was split into two chapters, USP <1032> Design and

Development of Biological Assays and USP <1034> Analysis of Biological Assays

<1033> Biological Assay Validation added to the suite

“Roadmap” chapter (to include glossary)

6

Page 7: Regulatory Challenges in Bioassay Practices

Bioassay guidelines (cont.)

All but chapter <111> are above 1000 and therefore “informational” Not intended as enforceable (as chapters below 1000)

However, the chapters provide a set of guiding principles which might be considered by regulators in their reviews

Chapter <111> is left to support monographs which reference it USP is working towards addressing product-specific references

to prepare it for further revision

7

Page 8: Regulatory Challenges in Bioassay Practices

Regional differences in bioassay practices

Assessing “linearity” and similarity “Linearity” is the goodness-of-fit to the processing model

Similarity is the equivalence of the bioassay model parameters

• Parallelism in parallel line analysis

• Equivalence of asymptotes and Hill coefficient in 4 parameter logistic regression

• Equivalence of intercepts in slope ratio analysis

Parallel line/curve versus slope ratio analysis For some vaccines the US requires parallel line analysis while the EU requires slope

ratio analysis

Design and performance characteristics are different for the two approaches

Clinical specifications versus process consistency Growing expectation in the US that potency specifications should be supported by

clinical studies

Page 9: Regulatory Challenges in Bioassay Practices

Regional differences in bioassay practicesAssessing “linearity” and similarity

Significance testing versus equivalence testing

Laboratory A

-1.2

-0.8

-0.4

0

0.4

0.8

0.5 1 1.5 2 2.5

Log10 Concentration

Log1

0 R

espo

nse

Standard DataTest DataStandard LineTest Line

Laboratory B

-1.2

-0.8

-0.4

0

0.4

0.8

0.5 1 1.5 2 2.5

Log10 ConcentrationLo

g10

Res

pons

e

Standard DataTest DataStandard LineTest Line

p = 0.02 (p < 0.05, i.e., significantly different)

Conclude nonparallel!

Penalized for good assay performance

p = 0.08 (p > 0.05, i.e., not significantly different)

Conclude parallel!

Rewarded for poor assay performance

Page 10: Regulatory Challenges in Bioassay Practices

Paradox: significance tests reward poor work and penalize good work The greater the precision in the data,

the more likely you will fail the significance test

Solution – use an equivalence test Determine an acceptable range in a

metric related to “linearity” or similarity (LAL,UAL)

Demonstrate (TOST) that there’s acceptable similarity

• CI includes 0 = no evidence of a difference

• CI within (LAL,UAL) = similar

a: no evidence of a differencein slopes; however, possibly outsideacceptance limit (not similar)

b: no evidence of a difference in slopes; but inside acceptance limit (similar)

c: a difference in slopes; however, within acceptance limit (similar)

a b c

0

a b c

LALLAL

Note: Rewarded for good work

N=6 N=8N=4

10

Regional differences in bioassay practicesAssessing “linearity” and similarity (cont.)

UAL

Page 11: Regulatory Challenges in Bioassay Practices

Hurdles to an equivalence approach European regulation adheres to EP 5.3 The revision of EP 5.3 does not allow for other approaches Some points of view

• “Statistical significance is scientifically important”• The significance approach can be moderated by an examination of the results

• Use of historical variability

• Moderation of significance level

• May lead to subjectivity

• “Calibration” to the equivalence approach• Engineer significance approach to duplicate equivalence approach

• More straight forward to apply equivalence approach

Establishment of an equivalence margin

11

Regional differences in bioassay practicesAssessing “linearity” and similarity (cont.)

Page 12: Regulatory Challenges in Bioassay Practices

Approaches for assigning an equivalence margin – operational approaches Approach 1 – based on “process capability” of the bioassay

• Addresses only the producer’s risk• There is no penalty for a poorly designed bioassay

Approach 2 – based on “process capability” using a tolerance interval on the confidence interval

• Penalizes poor assay runs

12

Regional differences in bioassay practicesAssessing “linearity” and similarity (cont.)

Diffe

renc

e of

Slo

pes

Approach 1

Diffe

renc

e of

Slo

pes

Approach 2 -

-

Page 13: Regulatory Challenges in Bioassay Practices

Approaches for assigning an equivalence margin – quality approaches Approach 3 – based on discrimination between “good” and “bad”

behavior• Protects both producer’s and consumer’s risk

• However, how do you define/generate “bad” behavior

• Some sensitivity to clinical correspondence to “bad” behavior Approach 4 – ad hoc limits

• Based on product or assay knowledge

• Should consider potential impact to product quality

13

Regional differences in bioassay practicesAssessing “linearity” and similarity (cont.)

Page 14: Regulatory Challenges in Bioassay Practices

► There are currently regional differences in the accepted processing of some vaccines (flu)

– EU requires slope ratio while North America requires parallel line analysis

► In slope ratio analysis variability of RP is impacted by increased variability of influential regression points Slope is influenced by extreme

points

► Simulation results True relative potency equal to

2.00 Impact of increase in assay

%RSD n=100 simulated assays per

condition

0

2

4

6

8

10

12

14

0 1 2 3 4 5

Slope Ratio Analysis

Standard

Test

Assay%RSD

1% 1.99 (4%) 1.99 (2%)10% 2.06 (45%) 2.02 (19%)20% 1.95 (159%) 2.21 (48%)

Slope Ratio Parallel LineEstimated RP (%RSD)

• Little impact on relative potency determination with increase in assay %RSD

• Dramatic increase in RP %RSD for slope ratio assay• Assay design should be adapted to parallel line

approach (geometric doses)

Regional differences in bioassay practicesBioassay design and data processing (cont.)

Page 15: Regulatory Challenges in Bioassay Practices

US viewpoint on bioassay practicesValidation of assay format

Some US regulators believe that bioassay validation should be a verification of the procedure for obtaining a “reportable value” Groupings in time have different variability characteristics than the sum

of the variance components

Emphasis on product release ignores other uses

Some merit to this if the validation is not designed to address the issue of short term versus long term variability

• Replicate the bioassay under the same set of ruggedness conditions

2.8

3

3.2

3.4

3.6

3.8

4

4.2

0 3 6 9 12 15 18 21 24Time (Month)

Regression

Page 16: Regulatory Challenges in Bioassay Practices

0

50

100

150

200

1 10 100 1000

Res

pons

e

Concentration

Dilutional Linearity Risk of truncation error and range

Retest rules have the potential to lead to truncation bias in the reportable value

• e.g., retest when a measurement is outside the “quantifiable range” of the bioassay

Potential solutions

• Assign a value to the low/high result (e.g., ½ the LLOQ in clinical assays)

• Demonstrate a range which supports low/high potency samples (without retest)

• Retest the series using an adjusted dilution scheme

Managing variabilityBeyond random and systematic variability (cont.)

16

Test

Avg

Retest

Lot 1

Lot 5

Lot 2

Lot 3

Lot 4

Page 17: Regulatory Challenges in Bioassay Practices

US viewpoint on bioassay practicesBioassay characterization

USP allows for “Use of validation results for bioassay characterization” Use of variance components to

adapt bioassay format

• Numbers of runs (assays) and replicates to efficiently manage bioassay variability

• Identify potentially significant sources of variability

• Update technique or training

• Replicate over significant factors

% 1e100yVariabilit ormatF knˆ

kˆ 2

Replicate2Run

Format variability for different combinations of number of runs (k) and number of minimal sets within run (n)

Number of Runs (k)

Reps (n) 1 2 3 6

1 7.2% 5.1% 4.1% 2.9%

2 6.4% 4.5% 3.6% 2.6%

3 6.0% 4.2% 3.4% 2.4%

6 5.7% 4.0% 3.3% 2.3%

ComponentVariance Estimate

Var(Media Lot) 0.0000Var(Analyst) 0.0014

Var(Analyst*Media LOt) 0.0000

Var(Run(Analyst*Media Lot)) 0.0019Var(Error) 0.0022

Page 18: Regulatory Challenges in Bioassay Practices

QbD for analytical methods

Industry has begun to recognize that analytical methods generate a product – measurements

Like pharmaceutical products, measurements should have adequate quality to meet their intended use – decision making

The fundamental goals of product development are: Safety and efficacy (hitting the clinical target)

Variance reduction

The fundamental goals of analytical development are: Accuracy (hitting the analytical target)

Variance reduction

18

Page 19: Regulatory Challenges in Bioassay Practices

QbD for analytical methods (cont.)

Many of the concepts associated with QbD for pharmaceutical products translate to concepts related to analytical methods

Process Concept Analytical Counterpart

Target Product Profile (TPP)•Target clinical performance, manufacturing, and commercial requirements

Analytical Target profile (ATP)•Target analytical performance, testing laboratory, and customer requirements

Critical Quality Attributes(CQAs)•Potency•Aggregation•Purity

Performance attributes (validation parameters)•Precision•Sensitivity•Accuracy

Specifications (acceptance criteria)•80% to 125% potency•Purity > 95%

Acceptance criteria•%GCV < 10%•LLOQ > 1 ng/mL

Critical process parameters•pH, time, temperature

Critical assay parameters•pH, time, temperature

Process control strategy•Comparability protocols•Tech transfer

Assay control strategy•Comparability protocols•Method transfer

Continuous verification•Continuous review and updating of process knowledge

Continuous verification•Continuous review and updating of analytical knowledge 19

Page 20: Regulatory Challenges in Bioassay Practices

QbD for analytical methods (cont.)

The bioassay should be fit for its intended use throughout the bioassay lifecycle Should perform adequately to support decisions

Decisions are made day-to-day using bioassays During development

• Which formulation provides the best stability?

• Does a particular process step impact potency?

• What is the self-life of the product?

During manufacture

• Should a manufactured lot be released to the market?

• Should a process change be implemented?

• Has a manufactured lot maintained potency over it shelf-life?

• Can a new potency standard be used in the bioassay?

20

Page 21: Regulatory Challenges in Bioassay Practices

QbD for analytical methods (cont.)

All decision are made with risk, and risk is costly Risks during development

• Risk of deciding “the process” is suboptimal when it is fit– producers risk – results in excessive development costs or program failure

• Risk of deciding the process is fit when “the process” is flawed – consumers risk – results in excess downstream costs to fix the problems

• Risks during manufacture

• Risk of failing satisfactory product –regulatory burden and lost revenues

• Risk of passing unsatisfactory product – potential risk to the “customer”

Decision risk can be managed in several ways Use sound scientific reasoning and/or experience to guide decisions

When decisions are made on the basis of empirical evidence, decision risk is associated with the strength of the evidence

21

Page 22: Regulatory Challenges in Bioassay Practices

QbD for analytical methods (cont.)

Statistical opportunities – supporting evidence based development and control Statistical thinking

• Understanding variability

• Managing variability

• Communicating uncertainty

Bioassay models and analyses

Bioassay optimization

Bioassay maintenance

• Statistical process control

• Standard qualification/calibration

• Method transfer

• Method comparison – in vivo to in vitro

22

Page 23: Regulatory Challenges in Bioassay Practices

Summary

Bioassays are utilized throughout the biological product lifecycle to make key development and quality decisions

Statistical approaches facilitate decision making and help mitigate the risks of bioassay variability

Industry and regulatory statisticians should work together to support bioassay development, and should help promote best practices in implementation


Recommended