12.11.2012 Views

Vaccine

Vaccine

Vaccine

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

USP Bioassay Workshop<br />

August 12-13, 2009<br />

USP Headquarters<br />

Quality Standards for Medicines, Supplements, and Food Ingredients throughout the World


Regulatory perspective on<br />

vaccine potency assays<br />

USP’s 2 nd Bioassay Workshop<br />

August 13, 2009<br />

Phil Krause, MD<br />

FDA/CBER


Potency<br />

Specific ability or capacity of the product,<br />

as indicated by appropriate laboratory<br />

tests or by adequately controlled clinical<br />

data obtained through the administration<br />

of the product in the manner intended, to<br />

effect a given result. -- [21 CFR §600.3<br />

(s)]<br />

2


Tests for Potency<br />

Tests for potency shall consist of either in<br />

vitro or in vivo tests, or both, which have<br />

been specifically designed for each<br />

product so as to indicate its potency in a<br />

manner adequate to satisfy the<br />

interpretation of potency given by the<br />

definition in § 600.3 (s) of this chapter. --<br />

[21 CFR § 610.10]<br />

3


Why do we do a potency assay?<br />

• In development, to:<br />

– Assure that safe potencies are not exceeded in clinical trials<br />

– Obtain information that will support licensure‐including<br />

correlation of potency with clinical response<br />

• After licensure, to assure that lots behave similarly to<br />

those tested in the clinical trials that supported licensure<br />

– The potency should not be below the lowest potency believed to<br />

be efficacious<br />

– The potency should not exceed the highest potency believed to<br />

be safe<br />

• The potency assay thus provides a “bridge” between<br />

licensed material and the clinical trials


What does a potency assay tell us?<br />

• The assay estimates the mean potency value for a lot<br />

• There are two sources of variability in the measured<br />

potency of an individual vial from the same vaccine<br />

lot– manufacturing variability and assay uncertainty<br />

• Thus, we can never know the actual potency in an<br />

individual vial that is given to a vaccine recipient<br />

• We can know the characteristics of the lot of vaccine<br />

from which that vial came (we routinely estimate the<br />

mean potency)


Necessary attributes for<br />

potency assays<br />

• Predictive of clinical benefit<br />

• Possess characteristics that are amenable to<br />

validation<br />

• Precision sufficient to meet goal of potency<br />

assays, i.e., provide assurance that vaccine<br />

is safe and effective throughout the dating<br />

period<br />

– Includes for use in stability studies<br />

– Includes for use in the “bridge” between marketed<br />

and clinical trial materials<br />

• Stability indicating<br />

6


Choosing “the right thing”<br />

Every worker in biology<br />

must know the temptation<br />

to adopt a method<br />

because it measures<br />

something with<br />

reasonable precision,<br />

without waiting for<br />

conclusive evidence that<br />

what it measures is the<br />

right thing<br />

- Sir Henry Dale<br />

Nobel Laureate in Medicine, 1936<br />

7


Not everything that<br />

counts can be<br />

counted, and not<br />

everything that can<br />

be counted counts.<br />

‐Albert Einstein


Building the Bridge<br />

Clinical Lots Assay Results<br />

Potency Assay Bridge<br />

During clinical development, it is necessary to<br />

establish a quantitative link between product efficacy<br />

and laboratory measured parameters? What are the<br />

relevant parameters to use? How do we choose the<br />

“right thing.”<br />

9


Building the Bridge<br />

Clinical Lots Marketed Lots<br />

Potency Assay<br />

Bridge<br />

An assay, or set of assays, is used to measure the<br />

comparability of a given lot of product to those lots of<br />

product that were used in the clinical trials to establish<br />

efficacy. Were the relevant parameters established?<br />

10


Building the Bridge<br />

• Establishing a bridge between clinical outcomes<br />

and laboratory test parameters is enabled by<br />

– Knowledge of disease pathogenesis and mechanisms of<br />

mediation<br />

– Existence of surrogate markers<br />

– Characterization of products (antigens and antibodies<br />

for vaccines)<br />

– Existence of appropriate animal models<br />

– Data from clinical trials<br />

11


Relationship between potency and<br />

surrogate markers<br />

• Potency refers to the ability to effect a clinical<br />

outcome<br />

• Surrogate markers, in clinical subjects, predict<br />

actual clinical outcomes, and imply an<br />

understanding of the mechanism of action<br />

• Often, the best potency assays measure attributes<br />

of the product that are likely to induce a response<br />

measured by a surrogate marker<br />

12


Surrogate Markers<br />

“surrogate endpoint . . . is reasonably likely, based<br />

on epidemiologic, therapeutic, pathophysiologic, or<br />

other evidence, to predict clinical benefit or on the<br />

basis of an effect on a clinical endpoint other than<br />

survival or irreversible morbidity.”<br />

[21 CFR 314.510 (Subpart H) Accelerated approval]<br />

13


Building the Bridge<br />

• In the absence of an existing laboratory<br />

surrogate of protection, establishing the<br />

ability to accurately assess potency, i.e.,<br />

develop the potency assay, will rest on<br />

clinical results with drug preparations of<br />

varying measured quality.<br />

14


Development of a potency test:<br />

whole-cell pertussis vaccine<br />

1940’s: Potency test<br />

for whole-cell<br />

pertussis vaccine<br />

Kendrick & Pittman<br />

Mouse protection<br />

from a lethal<br />

intracerebral<br />

infection<br />

15


Development of a potency test:<br />

whole-cell pertussis vaccine<br />

1950’s: UK MRC<br />

trials showed a<br />

correlation of the<br />

MP test with<br />

vaccine efficacy<br />

Became the world<br />

wide accepted<br />

potency test for<br />

whole-cell pertussis<br />

vaccine<br />

16


• Cell culture systems:<br />

Potency bioassays<br />

– For biologicals, potency assays often detect the ability of the<br />

product to achieve a desired effect in a cell culture system<br />

• Animal models.<br />

– often more difficult to use<br />

– are time-consuming<br />

– frequently have greater variability than in vitro assays<br />

– may have greater biological relevance<br />

• These differences suggest the value of attempting to<br />

identify in vitro predictors of efficacy and safety early in<br />

product development.<br />

17


Potency Assays and <strong>Vaccine</strong>s: A Few<br />

Examples<br />

• Number of plaque forming units (e.g., mumps, measles,<br />

rubella, smallpox)<br />

• Number of colony forming units (e.g., S. typhi, TY21a)<br />

• Chemical and Physical chemical characterization (e.g.,<br />

polysaccharide and polysaccharide‐protein conjugate<br />

vaccines)<br />

• Serological response in animals (e.g., diphtheria)<br />

• Animal protection against challenge (e.g., rabies, anthrax)<br />

18


Situations where potency assays are<br />

particularly difficult to develop<br />

• Mechanism of action or surrogates of protection not<br />

well understood<br />

• Multi‐component products, where there is a<br />

significant interaction among components<br />

• Absent definitive tests that predict efficacy, product<br />

evaluation is based on the demonstration that the<br />

newly manufactured lots are comparable to those<br />

shown in clinical studies to be safe and effective<br />

19


Potency and product life cycle<br />

• Each progressive phase of development is based upon<br />

an ability to relate their respective measures of potency.<br />

• Doses used in Phase 1 studies must be capable of<br />

being related to those used in pre-clinical studies<br />

• In turn, doses used in Phase 2 and Phase 3 studies<br />

must be capable of being related both to each other and<br />

to material used in Phase 1.<br />

• After licensure, potency assays are used to help assure<br />

that marketed product contains a quantity of active<br />

ingredient similar to that shown to be safe and effective<br />

in clinical trials.<br />

20


Potency and the product life‐cycle<br />

(additional points)<br />

• As clinical development progresses, the<br />

measure or measurement of potency may<br />

change.<br />

• At the time a product is first developed, it will<br />

likely not be known what in vivo or in vitro<br />

characteristics are likely to correlate with<br />

clinical benefit. This understanding will evolve<br />

during the course of clinical development.<br />

21


Potency and the product life‐cycle<br />

(additional points)<br />

• The need to understand the performance of assays over<br />

time and through clinical development points to the need<br />

for maintaining stable reference preparations against<br />

which potency results may be standardized<br />

• There is also considerable value in retaining (under<br />

conditions of high stability) samples of material used<br />

during the clinical development process in order to<br />

bridge between clinical studies, for example, if the retesting<br />

of potency in updated assays becomes<br />

necessary.<br />

22


Setting Specifications on Potency Assays:<br />

• How much do we need?<br />

Two Questions<br />

• How certain do we need to be of that value?<br />

23


U.L.<br />

Potency<br />

L.L.<br />

Calculation of vaccine minimum<br />

release potency<br />

•For vaccines, we usually set a minimum release spec to<br />

assure mean potency exceeds a clinically acceptable lower<br />

limit (L.L.) throughout the shelf life<br />

Release spec<br />

} Error term<br />

Time<br />

Expiry<br />

} Expected stability loss<br />

based on stability studies<br />

(linear regression, when<br />

feasible)


U.L.<br />

Potency<br />

L.L.<br />

Calculation of minimum release<br />

}<br />

storage<br />

Release spec<br />

Error term<br />

shipping<br />

storage in clinic<br />

potency<br />

reconstitution<br />

Time<br />

Expiry<br />

} Expected stability loss<br />

should encompass all<br />

anticipated conditions


U.L.<br />

Potency<br />

L.L.<br />

Calculation of minimum release<br />

LRL<br />

} Error term<br />

potency (LRL)<br />

• Error term should account for:<br />

• Variability in potency assay<br />

• Error associated with stability modeling<br />

Time<br />

Expiry<br />

} Expected stability loss


U.L.<br />

Potency<br />

L.L.<br />

Calculation of minimum release<br />

LRL<br />

} Error term<br />

potency (LRL)<br />

•Goal of calculation:<br />

•A lot with a potency test revealing a mean<br />

potency at the minimum release specification<br />

LRL has a 95% chance of retaining a mean<br />

potency at or above L.L. at the time of expiry<br />

Time<br />

Expiry<br />

} Expected stability loss


U.L.<br />

Potency<br />

L.L.<br />

Calculation of minimum release<br />

LRL<br />

} Error term<br />

potency (LRL)<br />

•If variability in potency assay is too great, then it<br />

may not be feasible to manufacture product such<br />

that potency can be assured to remain between LL<br />

and UL (highest clinically acceptable dose)<br />

throughout shelf life<br />

Time<br />

Expiry<br />

} Expected stability loss


U.L.<br />

Potency<br />

L.L.<br />

Release potency window<br />

} Error term<br />

URL<br />

LRL<br />

} Error term<br />

(LRL –URL)<br />

•If product cannot be produced such that it an be<br />

released at potency between LRL and URL , then<br />

there is no consistently manufacturable product<br />

Time<br />

Expiry<br />

} Expected stability loss


When is this easier?<br />

• Assay precision is high<br />

– When assay precision is high, it is easier to<br />

construct a release model due to improved<br />

understanding of release potencies and stability<br />

– Must be careful that assay is measuring the right<br />

thing<br />

• Manufacturing variability is low<br />

• Therapeutic index is wide<br />

– At some point, somebody may be nonetheless<br />

tempted to push limits!


Strategies to reduce effect of<br />

variability in potency assays<br />

• Multiple replicates<br />

– Permits greater confidence in result<br />

• Standardization (to house or international standard)<br />

– Normalize results to a standard of known potency<br />

– Reduces impact of inter‐run assay variability, but not<br />

within‐run assay variability<br />

– Need some way to assure potency value for standard is<br />

and remains accurate<br />

31


A single assay doesn’t always<br />

suffice<br />

• Complex mechanism of action or complex<br />

characterization<br />

• Consider multiple assays, both bioassays and<br />

physical chemical assays<br />

– Example: one assay could address upper safety limit, while<br />

a different assay addresses lower limit<br />

– Bioassay to indicate range of potency or minimum value<br />

and chemical assay to provide quantity<br />

• Need to beware of multiplicity issues<br />

32


Stability‐indicating assays<br />

• Identify clinically meaningful degradation in<br />

product<br />

• Implies that the assay predicts clinical benefit<br />

not only at the beginning of the dating period,<br />

but also at the end<br />

• Supportive data can often be obtained in<br />

accelerated or forced degradation studies


Necessary attributes for potency<br />

assays<br />

• Predictive of clinical benefit<br />

• Possess characteristics that are amenable<br />

to validation<br />

• Precision sufficient to meet goal of<br />

potency assays, i.e., provide assurance<br />

that vaccine is safe and effective<br />

throughout the dating period<br />

• Stability indicating<br />

34


Influenza Potency Assays<br />

Mark S. Galinski<br />

<strong>Vaccine</strong> Analytical Sciences<br />

MedImmune<br />

13-Aug-2009


� Influenza Background<br />

� Drift and shift of virus antigenicity<br />

� Commercial <strong>Vaccine</strong>s<br />

Outline<br />

� Potency Assay Traditional Inactivated Influenza <strong>Vaccine</strong>s<br />

� Single Radial Immunodiffusion Assay<br />

� Potency Assay Live Attenuated Influenza <strong>Vaccine</strong><br />

� Fluorescent Focus Assay<br />

� FFA Automation<br />

� Isocyte for Foci Enumeration


Fields Virology, 5 th ed, 2007<br />

Influenza Virus – Many Reservoirs<br />

– Many Subtypes<br />

� Influenza A, B, & C (serotypes) can infect humans<br />

� Seasonal influenza vaccines contain A and B serotype proteins<br />

� Influenza A has (16) HA and (9) NA subtypes


� HA Hemagglutinin<br />

� Receptor Binding Protein<br />

Fields Virology, 5 th ed, 2007<br />

Influenza A HA and NA subtypes<br />

� NA Neuraminidase<br />

� Receptor destroying activity


Antigenic Drift<br />

Annual Influenza Epidemics<br />

� HA undergoes amino acid changes at sites targeted by antibodies (genetic<br />

mutation) to generate a “current” circulating strain<br />

� Immunity to the “current” circulating strain is reduced (incomplete<br />

protection)<br />

� New strain emerges<br />

> Selection for variant strain to spread due to incomplete protection


Antigenic Shift<br />

Pandemic Influenza<br />

Belshe, R.B. The Origins of Pandemic Influenza –Lessons from the 1918 Virus. The New<br />

England Journal of Medicine 353:2209‐2211, 2005.


Seasonal Influenza <strong>Vaccine</strong>s<br />

FDA-Approved Trivalent <strong>Vaccine</strong>s<br />

Manufacturer <strong>Vaccine</strong> Type<br />

CSL Limited Inactivated, split<br />

GlaxoSmithKline Biologicals Inactivated, split<br />

ID Biomedical Corp of Quebec Inactivated, split<br />

Novartis <strong>Vaccine</strong>s and Diagnostics Ltd Inactivated, split<br />

Sanofi Pasteur, Inc Inactivated, split<br />

MedImmune, LLC Live, intranasal


Embryonated Eggs H1N1<br />

� Traditional inactivated vaccine (split)<br />

Influenza Manufacturing<br />

Drug Substance<br />

� Each strain is harvested from allantoic fluids, clarified, centrifuged,<br />

inactivated, membrane containing surface antigens HA and NA purified<br />

(split using detergent) from the internal viral proteins, and sterile filtered<br />

� Live attenuated influenza vaccine<br />

H3N2<br />

� Each strain is harvested from allantoic fluids, clarified, centrifuged, sterile<br />

filtered<br />

B


Influenza <strong>Vaccine</strong> Antigen Components<br />

� Hemagglutinin (HA) and Neuraminidase (NA) are the<br />

immunologically relevant proteins associated with vaccine<br />

efficacy<br />

Neuraminidase<br />

NA<br />

Matrix<br />

M<br />

Nucleocapsid<br />

N<br />

Hemagglutinin<br />

HA<br />

vRNA<br />

Membrane<br />

Polymerase<br />

Complex<br />

PA, PB1, PB2


� Traditional inactivated vaccine (split)<br />

� Blend of H1N1, H3N2 and B<br />

� Parenteral administration<br />

� Virus components directly antigenic<br />

Influenza <strong>Vaccine</strong>s<br />

Drug Product<br />

� Live attenuated influenza vaccine<br />

� Blend of H1N1, H3N2 and B<br />

� Intranasal administration<br />

� Virus components require<br />

replication to present antigens to<br />

immune system


Inactivated <strong>Vaccine</strong> Potency<br />

SRID/SRD<br />

� Inactivated influenza vaccine dosage based on clinical studies<br />

following re-emergence of influenza H1N1 in the late 1970s<br />

� Naive subjects (no pre-existing immunity) required two doses<br />

� Subjects with some degree of pre-existing immunity needed only a<br />

single dose<br />

� Potency of inactivated split vaccines measures HA content per<br />

dose using a single radial immunodiffusion (SRID/SRD) assay<br />

� 15 mcg/0.5 mL dose for each virus component<br />

� HA content correlated with antibody titers using whole-virus or split<br />

vaccine<br />

� Dose claim balanced maximum immune response in individual with<br />

maximizing population coverage and minimization of adverse reactions


Seasonal Influenza <strong>Vaccine</strong>s<br />

Potency Measurements<br />

� Potency measurements require reference immunoreagents<br />

� Type/Subtype matched antisera<br />

� Type/Subtype matched antigen (virus) HA reference standard<br />

� SRID assay features<br />

� Antiserum is incorporated into agarose matrix<br />

� Samples are solubilized with detergent and allowed to diffuse<br />

into the agarose<br />

� A zone of immunoprecipitin forms where the antibody-antigen<br />

complex is no longer soluble


Single Radial Immunodiffusion Assay<br />

SRD/SRID<br />

� HA potency measured by assessing immunoprecipitin diameter<br />

� Requires reference virus and reference antiserum (CDC, NIBSC)<br />

Ag Dilution<br />

Reference<br />

<strong>Vaccine</strong> (1) 1x<br />

<strong>Vaccine</strong> (2) 4x<br />

Concentrated Virus<br />

Antiserum in<br />

Agarose<br />

1:1 1:2 1:4 1:8<br />

Adapted from Wood et al., 1977 Develop, biol. Standard., 39:193-200<br />

R<br />

V 1<br />

V 2<br />

C<br />

1:1 1:2 1:4 1:8 1:1 1:2 1:4 1:8<br />

4x 2x 1x<br />

R<br />

V 1<br />

V 2<br />

C


Live Attenuated Influenza <strong>Vaccine</strong><br />

Influenza vaccine live, intranasal<br />

� Potency of live attenuated vaccine<br />

measures virus infectivity per dose<br />

using immunostaining of infected MDCK<br />

cells (fluorescent focus units)<br />

� 6.5-7.5 log 10 FFU/0.2 mL dose for each<br />

virus component


Fluorescent Focus Assay (FFA)<br />

� Potency of live attenuated vaccine measures virus infectivity<br />

per dose using immunostaining of infected MDCK cells<br />

(fluorescent focus units)<br />

� 6.5-7.5 log 10 FFU/0.2 mL dose for each virus component<br />

� FFA assay features<br />

� Quantitative assay enumerates influenza-infected cells following<br />

immunostaining<br />

� HA-specific primary antibodies followed by detection with a<br />

fluorescent-labeled secondary antibody (e.g. Alexa Fluor 488)<br />

� Primary antibodies specifically bind to one of each of the vaccine<br />

components<br />

> A/H1N1, A/H3N2 and B strain


Fix cells<br />

Stain with 1 °<br />

Antibody<br />

Infect 96-well MDCK cell monolayer<br />

with serial dilutions of virus sample<br />

Incubate 17-20 hrs<br />

Stain with 2 °<br />

Antibody<br />

Fluorescent Focus Assay<br />

Enumerate fluorescent foci


� FFA Validated<br />

FFA Validated Release Assay<br />

Drug Substance & Drug Product Release<br />

� Accuracy, precision, LOD, LOQ, specificity, linearity/range,<br />

ruggedness, robustness, system suitability<br />

� Potency is a quantitative measure of infectivity assessed at a<br />

specific time range post-infection<br />

� Precision for FFA is better than for a TCID 50 quantal assay<br />

� Typically 0.1 – 0.15 log 10 FFU<br />

� Typically >0.3 log 10 TCID 50<br />

� Accuracy/specificity requires critical immunoreagents<br />

� Potency testing of trivalent drug product needs to distinguish<br />

between each vaccine component


FFA Focus Enumeration<br />

� Manual counting of foci does not use the entire well<br />

� Scan and count across a portion (e.g. ~33%) of the well<br />

� Region of interest (ROI) dependent on optical magnification


FFA Focus Enumeration<br />

� Foci counting can be challenging<br />

� Counting range 15-200 foci<br />

� Analyst fatigue and ergonomics<br />

� Photobleaching reduces signal to<br />

noise for dimly stained foci<br />

� Sample/plate throughput<br />

� Automation assessed as a<br />

technical improvement


The IsoCyte<br />

Blueshift Biotechnologies<br />

MDS Analytical Technologies<br />

� Single or Dual Laser<br />

� Laser lines: 405, 440, 488, 532, or<br />

635 nm excitation<br />

� 4-color detection simultaneously<br />

� 2-color polarization<br />

� light scatter image<br />

� Accepts 6 – 1536-well plates and<br />

microscope slides<br />

� High speed laser scanning<br />

fluorimeter & scatterometer<br />

� Capablity to combine with stacker


Isocyte Technology<br />

� Laser is focused on a confined detection region at bottom of well<br />

� Depth of field allows large objects to be visualized in 3D<br />

� Fluorescence is collected by PMTs and digitized<br />

� Laser has very short dwell time - does not cause photobleaching<br />

PMT 1<br />

PMT 3<br />

Collection<br />

lens<br />

Dichroic mirror<br />

Laser<br />

Sample<br />

Scan lens<br />

Scanning mirror<br />

Emission filters<br />

PMT 2<br />

PMT 4


Implementation in QC Lab<br />

Technical & Regulatory Considerations<br />

� 21CFR Part 11 compliant data collection and control<br />

� Demonstrate equivalency to current manual method<br />

� Instrumentation needs to duplicate an analyst (human)<br />

� Art of staining and manual counting can influence comparability<br />

� Analyst trained to enumerate foci that an instrument finds<br />

problematic<br />

� Instruments can easily enumerate more foci more rapidly than<br />

any analyst<br />

� Extend enumeration capabilities (foci numbers & ROI)<br />

� Rectangle (e.g. 33% of well); 200 max foci<br />

� Circular (e.g.~80% of well); up to ~1,800 max foci (sample<br />

dependent)<br />

� PQ and assay validation


Implementation in QC Lab<br />

Technical & Regulatory Considerations<br />

� 21CFR Part 11 compliant data collection and control<br />

� Password Protection Procedure<br />

� User Account Procedure<br />

� User Modes of Operation<br />

� Audit Trail Management Procedure<br />

� System Calibration Procedure<br />

� Data Protection


Art of staining and manual counting can<br />

influence comparability<br />

FITC<br />

Nonspecific Staining<br />

Alexa 488<br />

Specific Staining


Manual<br />

160<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Foci Number Comparison<br />

y = 1.0715x<br />

R 2 = 0.9636<br />

0 50 100 150<br />

ISOCYTE<br />

Demonstrate equivalency<br />

Instrumentation Needs to Duplicate an Analyst<br />

Manual<br />

10.0<br />

9.0<br />

8.0<br />

7.0<br />

6.0<br />

Titer Comparison<br />

y = 1.0042x - 0.0032<br />

R 2 = 0.9915<br />

6.0 7.0 8.0 9.0 10.0<br />

ISOCYTE


Extend Enumeration Capabilities<br />

(Foci Numbers & ROI)<br />

� Circular (e.g.~80% of well); >500 foci<br />

� Rectangle (e.g. 48% of well); 200 max foci<br />

Manual Rec ROI<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

H1N1<br />

y = 1.4308x<br />

R 2 = 0.9998<br />

y = 1.0156x<br />

R 2 = 0.9898<br />

0<br />

0<br />

0 100 200 300 400 500 600<br />

Isocyte Rec ROI<br />

Manual vs Isocyte Rec vs Rec Isocyte Rec vs Circular<br />

Linear (Manual vs Isocyte Rec vs Rec) Linear (Isocyte Rec vs Circular)<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

Isocyte Cirular ROI


Extend Enumeration Capabilities<br />

(Foci Numbers & ROI)<br />

� Circular (e.g.~80% of well); >500 foci<br />

� Rectangle (e.g. 48% of well); 200 max foci<br />

Potency Isocyte (FFU/mL)<br />

4.0<br />

3.8<br />

3.6<br />

3.4<br />

3.2<br />

3.0<br />

2.8<br />

2.6<br />

2.4<br />

2.2<br />

H1N1<br />

2.0<br />

2.0 2.2 2.4 2.6 2.8 3.0 3.2 3.4 3.6 3.8 4.0<br />

Potency Manual (FFU/mL)<br />

Iso-Rec Iso-Cir


Potency Isocyte (FFU/mL)<br />

4.0<br />

3.8<br />

3.6<br />

3.4<br />

3.2<br />

3.0<br />

2.8<br />

2.6<br />

2.4<br />

2.2<br />

Extend Enumeration Capabilities<br />

(Foci Numbers & ROI)<br />

H3N2<br />

� Circular (e.g.~80% of well); >500 foci<br />

� Rectangle (e.g. 48% of well); 200 max foci<br />

2.0<br />

2.0 2.2 2.4 2.6 2.8 3.0 3.2 3.4 3.6 3.8 4.0<br />

Potency Manual (FFU/mL)<br />

Iso-Rec Iso-Cir<br />

Manual Rec ROI<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

H3N2<br />

y = 1.4708x<br />

R 2 = 0.9999<br />

y = 0.9378x<br />

R 2 = 0.9973<br />

0<br />

0<br />

0 100 200 300 400 500 600<br />

Isocyte Rec ROI<br />

Manual vs Isocye Rec vs Rec Isocye Rec vs Circular<br />

Linear (Manual vs Isocye Rec vs Rec) Linear (Isocye Rec vs Circular)<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

Isocyte Cirular ROI


Manual Rec ROI<br />

800<br />

700<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

y = 0.9625x<br />

R 2 = 0.9998<br />

Extend Enumeration Capabilities<br />

(Foci Numbers & ROI)<br />

B<br />

y = 1.4478x<br />

R 2 = 0.9997<br />

0<br />

0<br />

0 100 200 300 400 500 600<br />

Isocyte Rec ROI<br />

Manual vs Isocyte Rec vs Rec Isocyte REc vs Circular<br />

Linear (Manual vs Isocyte Rec vs Rec) Linear (Isocyte REc vs Circular)<br />

� Circular (e.g.~80% of well); >500 foci<br />

� Rectangle (e.g. 48% of well); 200 max foci<br />

Potency Isocyte (FFU/mL)<br />

4.0<br />

3.8<br />

3.6<br />

3.4<br />

800<br />

3.2<br />

700 3.0<br />

2.8<br />

600<br />

2.6<br />

500 2.4<br />

2.2<br />

400<br />

2.0<br />

300 2.0 2.5 3.0 3.5 4.0<br />

200<br />

100<br />

Isocyte Cirular ROI<br />

B<br />

Potency Manual (FFU/mL)<br />

Iso-Rec Iso-Cir


� PQ and assay validation<br />

� Regulatory Approval<br />

Implementation in QC


Summary<br />

� Influenza potency measurements assure that seasonal trivalent<br />

vaccine products will meet their intended clinical dose effect<br />

� The virus surface glycoproteins (HA, NA) are the<br />

immunologically relevant antigenic components of influenza<br />

vaccines<br />

� SRID assay is the standard methods for release of traditional<br />

inactivated influenza vaccines<br />

� FFA assay is used to measure the infectivity of live attenuated<br />

influenza vaccines<br />

� Influenza potency assays are critically dependent upon the<br />

immunoreagents and reference standards for an accurate<br />

assignment of potency


Influenza <strong>Vaccine</strong> Potency<br />

Determination and Pandemic<br />

Preparedness<br />

The BARDA/IED Perspective<br />

for<br />

USP 2 nd Bioassay Workshop August 13, 2009<br />

Armen Donabedian, PhD<br />

Senior Program Manager, <strong>Vaccine</strong>s Advanced Development<br />

HHS/BARDA/IED


From BARDA/IED to the<br />

USP Workshop<br />

Let’s work together to improve or<br />

replace the Single Radial<br />

Immunodiffusion (SRID) assay and<br />

facilitate seasonal and pandemic<br />

influenza preparedness


What is BARDA/IED?<br />

� Biomedical Advanced Research & Development Authority<br />

(BARDA) established by PAHPA in Jun. 07 in HHS/ASPR<br />

� Implements USG strategies & policies for MCMs from the Public<br />

Health Emergency Medical Countermeasure (MCM) Enterprise<br />

(PHEMCE) within the office of the ASPR<br />

� Coordinates integrated product portfolio approach to planning and<br />

executing research, development and acquisition of public health<br />

emergency MCMs<br />

� Supports MCMs for CBRN Threats, Pandemic Influenza, and<br />

Emerging Diseases<br />

– Advanced development<br />

– Stockpile procurement & establishment<br />

– Manufacturing infrastructure building<br />

– Science and Technology Platforms


Immediate Office<br />

of the ASPR<br />

Chem/Bio<br />

Rad/Nuc<br />

(CBRN)<br />

Biomedical<br />

Advanced Research<br />

& Development<br />

Authority<br />

ASPR/BARDA/IED Organization<br />

Office of the Secretary of HHS<br />

Regulatory/Quality<br />

Affairs<br />

Influenza &<br />

Emerging Diseases<br />

ASPR<br />

Strategic Science<br />

& Technology<br />

Policy, Planning &<br />

Requirements<br />

Office of Medicine,<br />

Science,<br />

& Public Health<br />

Computer<br />

Modeling<br />

Resources &<br />

Program Operations<br />

Office of<br />

Preparedness &<br />

Emergency<br />

Operations<br />

Acquisitions<br />

Management


� <strong>Vaccine</strong>s<br />

U.S. Pan Flu MCM Strategic<br />

Current & Possible New Policy<br />

Goals<br />

– Goal #1: Establish and maintain a dynamic pre-pandemic influenza vaccine stockpile<br />

available for 20 M persons (2 doses/person) or more persons depending on vaccine mfg.<br />

capacity & results of dose-sparing adjuvant studies and prime-boost immunization studies:<br />

H5N1 vaccine stockpiles<br />

– Goal #2: Provide pandemic vaccine to all U.S. citizens within 6 months of a pandemic<br />

declaration: pandemic vaccine (600 M doses)<br />

� Antivirals<br />

– Goal #1: Provide influenza antiviral drug stockpiles for pandemic treatment of 25% of U.S.<br />

population (75 M treatment courses) and federal share of antivirals for outbreak<br />

prophylactic usage as a community mitigation measure as shared responsibility<br />

– Goal #2: Provide influenza antiviral drug stockpiles for strategic limited containment at<br />

onset of pandemic (6 M treatment courses)<br />

� Diagnostics<br />

– Goal #1: Develop new high-throughput laboratory, point-of-care (POC), and home detection<br />

influenza diagnostics for pandemic influenza virus detection<br />

� Other Countermeasures<br />

– Goal #1:Develop and acquire other MCMs including syringes/needles, masks/respirators,<br />

ventilators, antibiotics, & other supplies (Pneumococcal & Streptococcal <strong>Vaccine</strong>s?)<br />

National Strategy for Pandemic Influenza (Nov 2005) and HHS Pandemic Influenza Plan (Nov 2005) www.pandemicflu.gov


32 contracts &<br />

2 grants<br />

totaling $6.0 B<br />

Advanced<br />

Development<br />

Stockpile<br />

Acquisitions<br />

Infrastructure<br />

Building<br />

BARDA Pan Flu Integrated<br />

Program Portfolio Approach<br />

<strong>Vaccine</strong>s Antivirals Diagnostics/<br />

Respiratory<br />

Devices<br />

Cell-based<br />

Antigen-sparing<br />

Next Generation<br />

Recombinant<br />

H5N1 Pre-Pandemic<br />

<strong>Vaccine</strong> Stockpiles<br />

& H1N1 Purchases<br />

Retrofit Existing Mfg<br />

Facilities<br />

Build New Cellbased<br />

Mfg Facilities<br />

Egg-based Supply<br />

Peramivir<br />

More to Come<br />

Tamiflu & Relenza<br />

Federal Stockpiles<br />

State Stockpiles<br />

AV MedKits<br />

Diagnostics<br />

Point of Care<br />

Clinical Lab<br />

Ventilators<br />

Next Generation<br />

Masks &<br />

Respirators


Pandemic Influenza<br />

Medical Countermeasure<br />

Supply-Demand Supply Demand Gap Closure<br />

Reduce Demand: Pre-Pandemic <strong>Vaccine</strong>s, Community Mitigation, Antivirals, <strong>Vaccine</strong>s, Masks<br />

Increase Capacity: Ventilators, Oxygen, Antivirals, Pandemic <strong>Vaccine</strong>s, Masks<br />

Pre-Pandemic<br />

<strong>Vaccine</strong>s<br />

Demand for<br />

Healthcare Services<br />

Recombinant<br />

<strong>Vaccine</strong>s<br />

Egg- & Cellbased<br />

<strong>Vaccine</strong>s<br />

Increase Supplies of Critical Materiel<br />

Current Healthcare Capacity


Influenza Potency Assay<br />

Time Line Considerations<br />

SRID potency assay reagent availability is a key element<br />

8-12 weeks est. during a pandemic emergency - Actual for 2009 H1N1 was 13 weeks<br />

(Seed strains available at end of April – first reagents available at the end of July)<br />

Seasonal SRID potency assay reagents typically available in 4 months<br />

Pre-Pandemic<br />

<strong>Vaccine</strong>s<br />

Demand for<br />

Healthcare Services<br />

Recombinant<br />

<strong>Vaccine</strong>s<br />

12<br />

weeks<br />

20<br />

weeks<br />

Egg- & Cellbased<br />

<strong>Vaccine</strong>s<br />

Increase Supplies of Critical Materiel<br />

Current Healthcare Capacity


Ant<br />

Assay<br />

Based on Bull World Health Organ. 1975; 52(2): 223–231. 223 231.<br />

Influenza <strong>Vaccine</strong> Single Radial<br />

Immunodiffusion<br />

Ab<br />

http://www.gbiosciences.com/EducationalUploads/EducationalProductsImages<br />

http://www.gbiosciences.com/EducationalUploads/EducationalProductsImages<br />

PAGE<br />

Protein Assay<br />

Calibration<br />

Densitometer<br />

http://www.gmi-inc.com/BioTechLab/Bio2520Rad<br />

http://www.gmi inc.com/BioTechLab/Bio2520Rad


� The Problem<br />

Influenza <strong>Vaccine</strong> Potency<br />

Determination and Pandemic<br />

Preparedness<br />

– Preparation and calibration of SRID potency reagents delays<br />

production, formulation, clinical study and/or release of influenza<br />

vaccine<br />

� Considerations for replacement<br />

– SRID is the influenza vaccine stability indicating assay<br />

– The immunological component of the assay is considered to be<br />

essential<br />

� One or more of the three vaccine strains (H1, H3, B) change annually<br />

� compelling evidence of manufacturing consistency is difficult to establish<br />

– Irony is that the antigen standard is measured using physio-chemical<br />

methods only


� Proposed Partial Solution<br />

Influenza <strong>Vaccine</strong> Potency<br />

Determination and Pandemic<br />

Preparedness<br />

– Accelerate and improve accuracy of reagent calibration<br />

– LC-MS/MS<br />

Quantification of Influenza Virus Hemagglutinins in Complex<br />

Mixtures Using Isotope Dilution Tandem Mass Spectrometry<br />

<strong>Vaccine</strong> 26, 2008, p2510<br />

Tracie. L. Williams, Leah Luna, Zhu Guo, Nancy J. Cox, James L.<br />

Pirkle, Ruben O. Donis, John R. Barr<br />

National Center for Environmental Health<br />

&<br />

National Center for Infectious Diseases


Isotope Dilution LC-MS/MS<br />

� Unique conserved sequences were identified<br />

– Apply to 97+% of H1, H3, H5 and B influenza strains<br />

� The peptides are used as a stoichiometric representative of the<br />

specific protein they are derived from<br />

� A known amount (concentration essential) of labeled C 13 and N 15<br />

labeled peptide is spiked into the sample (e.g. 6 -10 daltons<br />

heavier)<br />

� The endogenous target peptide is released by detergent<br />

treatment and trypsin cleavage (completeness essential)<br />

� Quantification is performed by comparing the peak area of the<br />

labeled peptide with that of the endogenous target


� LC-MS/MS<br />

Isotope Dilution LC-MS/MS<br />

– Long established principle used for years in reference labs<br />

– Replaces carbon-12 with carbon-13 to create an internal standard<br />

with nearly identical chemical properties but different mass<br />

� Accuracy traces to NIST standard reference materials for amino<br />

acids<br />

– Amino acid analysis using ninhydrin with UV detection did not show<br />

agreement among labs<br />

– CDC developed a MS amino acid analysis method using NIST<br />

accuracy standards<br />

– CDC analyses of NIST aa estimates 149.85 (CV 2.20%) pmoles<br />

compared with an expected value of 150 pmoles


� Compares well with SRID<br />

Isotope Dilution LC-MS/MS<br />

Accuracy Precision LOD Time to establish<br />

SRID 10% 10% 10 ug One month<br />

LC-MS/MS<br />

?<br />

A priori* 5% 1 ug Immediate<br />

* 24.38, 16.52 and 38.8 ug HA/dose for the H1, H3 and B strains.<br />

– Each strain in influenza vaccine is formulated to 15 ug/dose based on the<br />

SRID assay


BARDA/IED LC-MS/MS LC MS/MS<br />

Proposed Agenda<br />

� Collaboration between CDC and CFSAN for method transfer<br />

– Review of method and qualification data<br />

– Lab evaluation<br />

� Instruments, data collection systems, personnel and special techniques<br />

� Reagents – identify a source for peptides<br />

� Development of acceptance criteria<br />

� Further clarification of the method procedure if necessary<br />

� Further qualification/validation of assay parameters such as sample<br />

storage or sample preparation<br />

� Procedure to confirm complete digestion of each new strain<br />

– Transfer Protocol<br />

� collect data and write report


BARDA/IED LC-MS/MS LC MS/MS<br />

Proposed Agenda<br />

� Collaboration between CBER and CFSAN for comparative<br />

studies<br />

– Implementation proposal (examples)<br />

� Preparation of standards for testing using current methods and LC-<br />

MS/MS<br />

� Comparison of past (H5) antigen standard values with LC-MS/MS<br />

� Comparison of (H5) vaccine and intermediates SRID values with LC-<br />

MS/MS<br />

� Evaluate alternative approaches to measure potency.<br />

– Immunoprecipitation<br />

– Protein mass mapping<br />

– The sialic acid - HA binding concept (NCIRD/CDC)<br />

– Other approaches


Development Funding Opportunity<br />

Science and Technology Platforms<br />

Applied to MCM Development<br />

Broad Agency Announcement (BAA)<br />

BARDA-BAA-09-100-SOL-0010<br />

Solicitation Posted July 8, 2009<br />

www.MedicalCounterMeasures.gov


Purpose of SSTD BAA<br />

Promote Innovations in Product Development<br />

Improvements, Testing, and Manufacturing<br />

� Platform technologies to promote:<br />

– Improvements in product safety, efficacy, stability, and ease of<br />

use<br />

– Efficient and cost-effective technologies associated with<br />

process development, testing, and manufacturing<br />

– Technologies relevant to targets of interest<br />

� Generate validated data sets for comparability purposes<br />

with products already in late stages of development<br />

� Integration with ongoing regulatory activities<br />

� Complement and support CBRN and IED portfolio efforts


SSTD BAA: Areas of<br />

Interest<br />

1. Technologies to Accelerate Evaluation of Candidate<br />

<strong>Vaccine</strong>s and Therapeutics<br />

2. Formulation Chemistry, Protein Stabilization, and<br />

<strong>Vaccine</strong> Delivery Technologies<br />

3. Innovative Methods in Bioprocess Development and<br />

Manufacturing<br />

4. Methods and Technologies to Advance Development<br />

of Diagnostic Tests for Rapid Diagnosis of Human<br />

Infections


1. Anthrax <strong>Vaccine</strong>s and Anti-Toxins<br />

SSTD BAA: Targets of Interest<br />

2. Pandemic Flu <strong>Vaccine</strong>s and Antivirals<br />

3. Botulinum Anti-Toxins<br />

4. Diagnostic Platforms Applicable to a Range of<br />

Pathogens (Bacterial and Viral)<br />

– Category A Bioagents<br />

– Emerging Infectious Diseases


SSTD BAA:<br />

Contact Information<br />

Michael Younkins<br />

Contracting Officer<br />

DHHS, BARDA<br />

Email: Michael.Younkins@hhs.gov<br />

www.MedicalCountermeasures.gov


� Federally-sponsored conferences<br />

� Funding opportunities<br />

� Resource programs<br />

� Regulatory guidance<br />

� Federal strategies and reports<br />

Contact Info<br />

BARDA:<br />

URL: www.hhs.gov/aspr/barda/<br />

E-Mail: BARDA@hhs.gov<br />

� Upcoming Events<br />

� Acquisitions<br />

� BioShield<br />

� Influenza Programs


Assessment of Neutralizing Antibodies<br />

to Biological Therapeutics<br />

Susan L. Kirshner, Ph.D<br />

Office of Biotechnology Products<br />

Division of Therapeutic Proteins


• Introduction:<br />

Outline<br />

– Immunogenicity concerns for therapeutic<br />

proteins<br />

– Definitions<br />

• Evaluating Immunogenicity<br />

• Neutralizing assays


Introduction


Concerns for Antibodies in the Clinic<br />

Clinical Concern Clinical Outcome<br />

Safety<br />

Efficacy<br />

Pharmacokinetics<br />

None<br />

•Neutralize activity of endogenous<br />

counterpart with unique function<br />

causing<br />

deficiency syndrome<br />

• Hypersensitivity Enhancing or decreasing reactionsefficacy<br />

by<br />

extending or decreasing half life.<br />

• Enhancing or decreasing efficacy by<br />

changing biodistribution.<br />

• Antibody production may dictate<br />

changes<br />

in dosing level due to PK changes.<br />

• Despite generation of antibodies, no<br />

discernable impact


There are cases in which immune responses<br />

to therapeutic proteins have had devastating<br />

consequences for healthy volunteers and<br />

patients.<br />

• rDNA Human MGDF<br />

• Erythropoietin<br />

• Glucocerebrosidase<br />

• a-glucosidase(Pompe’s)<br />

• Factor VIII<br />

• Insulin


What is immunogenicity?


Definitions<br />

• Antigen – any substance that can be recognized<br />

by the adaptive immune system (Janeway et al.<br />

Immunobiology, 6th Ed. Pg. 2)<br />

• Epitope – structure recognized by an antibody or<br />

T cell antigen receptor<br />

– Conformational – the epitope is recognized by its 3D<br />

structure. It may be composed of continuous or<br />

discontinuous protein sequences.<br />

– Linear – the epitope is recognized primarily based on<br />

its amino acid sequence. T cell epitopes are<br />

continuous, B cell epitopes can be continuous.


• Adaptive immunity – antigen specific<br />

immune response that evolves over time<br />

leading to increased efficiency in pathogen<br />

recognition and the establishment of<br />

immunological memory to a pathogen.<br />

Memory is established by the presence of<br />

long lived populations of antigen specific T<br />

and B cells. This generally involves either<br />

prolonged or multiple exposures to a<br />

pathogen or to pathogen epitopes (e.g.<br />

vaccines).


Antibody structure


Binding vs Neutralizing Antibodies<br />

Cell<br />

Activation<br />

Receptor<br />

Therapeutic<br />

Protein


-Bind anywhere on<br />

the molecule<br />

-May or may not<br />

inhibit protein<br />

activity in a<br />

bioassay<br />

-Detected through<br />

non-biologically<br />

based assay (e.g.<br />

ELISA, ECL, SPR)<br />

Binding Antibodies (BAbs)


Neutralizing Antibodies (NAbs)<br />

Cell<br />

Activation<br />

-NAbs are a subset if BAbs<br />

-Binding inhibits<br />

receptor/ligand interactions<br />

-Inhibit protein activity in a<br />

bioassay<br />

-Detected using a bioassay


Clinical Significance<br />

• In a patient both BAbs and NAbs can lead<br />

to loss of efficacy and/or negatively impact<br />

safety, therefore both may be clinically<br />

important<br />

• NAbs may be more effective in directly<br />

impacting efficacy<br />

– IL-2 and IFN-b higher titers of NAbs are<br />

associated with loss of efficacy


Evaluating Immunogenicity


Evaluating Immunogenicity: A Tiered<br />

Approach<br />

Sensitive screening immunoassay<br />

Negative<br />

Negative<br />

Negative<br />

Reactive<br />

Confirmatory assay<br />

(e.g. cold competition)<br />

Reactive<br />

Neutralizing<br />

Bioassay<br />

Positive<br />

IgG<br />

IgM<br />

IgE<br />

IgA


NAb Assays<br />

• The presence of NAbs should be<br />

assessed using a biologically based assay<br />

unless the MOA of the therapeutic has<br />

been shown to be cell independent (e.g.<br />

an enzymatic activity that occurs in serum)<br />

• The bioassay should be relevant to the<br />

MOA of the therapeutic<br />

• More than one assay may be required for<br />

a protein with multiple functional domains<br />

that are relevant to its therapeutic activity.


Potency Assays used in NAb<br />

Assays<br />

• Most frequently the assay used to assess<br />

potency for drug release is adapted to be used<br />

in the NAb assay. Therefore any format found to<br />

be appropriate for release is generally<br />

appropriate for the NAb assay. This includes<br />

assays with early (e.g., cAMP induction),<br />

intermediate (e.g. transcription) and late (e.g.<br />

proliferation, cytokine release) endpoints<br />

• Some cell lines do not adapt well to a matrix that<br />

includes human serum. Therefore the NAb<br />

assay may be based on a potency assay that is<br />

different from the release assay. The assay<br />

should still reflect the drugs purported MOA.


NAb Assay Formats<br />

• Most assays are based on a fixed amount of drug<br />

and cells and test articles at a single fixed dilution.<br />

Samples will be assessed as positive or negative if<br />

they inhibit the response beyond the cut-point.<br />

Alternatively the test article may be assessed at<br />

multiple dilutions with the titer reported as the lowest<br />

dilution giving a response below the cut-point.<br />

• For fixed dose assays it is recommended that cell<br />

lines be stimulated to 40% - 70% of their maximum<br />

response in the linear portion of the dose response<br />

curve. This dose is generally established during<br />

development. Dose response curves should be<br />

provided to the Agency to support the dose<br />

selection.


NAb Assay Formats<br />

• Some assays are based on shifts in EC50<br />

based on complete dose response curves<br />

in the presence and absence of test sera.


NAb Assay Formats: Reporting Results<br />

• It is recommended that assay results be reported as<br />

+/- or in titer<br />

• Results may be reported as units of drug inhibited<br />

per volume. It should be understood that this<br />

method of reporting results is quasi-quantitative and<br />

does not necessarily reflect the amount of drug<br />

inhibited in vivo. This method of reporting results is<br />

discouraged because the results are frequently over<br />

interpreted.<br />

• Assay results should not be reported in mass units<br />

based on back calculation to a standard curve<br />

because it is unlikely that dilutional parallelism exists<br />

between the Ab reference standard and Abs in the<br />

test serum for all test articles.


NAb Assays<br />

• The incidence of NAbs is reported in section 6.2<br />

of the label (PLR format). Therefore it is critical<br />

to have a robust validated assay.<br />

• Assay validation:<br />

– Cut-point<br />

–Precision<br />

– Selectivity – matrix interference (patient sera, on<br />

board drug)<br />

– Robustness – sensitivity to changes in operating<br />

parameters (e.g., incubation time/temperature)<br />

– Reproducibility – differences between laboratories.<br />

This is only necessary if the assay will be performed<br />

in more than one laboratory


NAb Assay Sensitivity<br />

• Assay sensitivity is generally assessed by<br />

determining the smallest amount of<br />

neutralizing Ab that tests positive in the NAb<br />

assay. Results are usually reported in mass<br />

units per volume.<br />

• Sensitivity is generally assessed as part of<br />

the validation exercise but may be assessed<br />

outside of the validation exercise.<br />

Regardless, results should be reported to the<br />

Agency.


NAb Assay Sensitivity<br />

• The assessment of assay sensitivity is<br />

dependent on the positive control Ab used<br />

• The development of positive control NAbs can<br />

be very difficult<br />

• If polyclonal Abs are used as a positive control<br />

then it is likely that only a very small portion of<br />

the Ab population will be neutralizing. This will<br />

result in an apparent lack of assay sensitivity.<br />

• Therefore we do not have a recommended<br />

target for assay sensitivity


NAb Assay Sensitivity<br />

• The determination of whether an assay<br />

has appropriate sensitivity is assessed<br />

based on overall assay performance in the<br />

validation exercise, results of the<br />

sensitivity assessment and information<br />

gained during assay development


Specificity<br />

• Due to the complexity of cell based NAb assays<br />

it is usually impossible to validate the specificity<br />

of the Abs<br />

• Because the Ab response is usually shown to be<br />

drug specific in the binding and confirmatory<br />

assays the specificity of the assay is frequently<br />

not confirmed or validated as with BAbs<br />

• Some Sponsors immunodeplete NAb positive<br />

serum samples to confirm that positive<br />

responses are Ab dependent


Selectivity<br />

• It is critical to assess the performance of the cell<br />

line in target population matrix because cell lines<br />

may respond to serum factors other than the<br />

API. Such responses can confound data<br />

interpretation<br />

• Incorporation of Ab against serum factors has<br />

been successfully used to overcome this<br />

problem.<br />

• On-board drug in the serum can also impact<br />

assay performance<br />

• Samples should be obtained at trough drug<br />

levels to mitigate this problem. Samples<br />

obtained after a sufficient washout period<br />

(generally ~5 half lives) may also be needed


Summary<br />

• Patients should be monitored for the presence of<br />

NAbs because they can negatively impact drug<br />

safety and efficacy<br />

• NAb assays should be biologically based and<br />

reflect the purported MOA of the drug<br />

• Results of assay validation exercises as well as<br />

critical development data (e.g. dose/response<br />

curves) and the assay SOP should be provided<br />

to the Agency for licensure.<br />

• NAb incidence (and titer when known/relevant)<br />

is reported in the product label

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!