Anda di halaman 1dari 7

GLOBAL BIOANALYTICAL SERVICES

WHITE PAPER

Implications of Immunogenicity in Drug Development


Michele Gunsior, PhD, Covance Laboratories, Chantilly, VA
efficacy. The latter is particularly prominent when immune
responses abrogate efficacy of lifesaving therapies, [Worobec,
2005]. The importance of immunogenicity to patient safety and
drug efficacy has resulted in a number of guidance documents
prepared by the regulatory community, the most recent of which
is FDAs Draft Guidance for Industry on Assay Development
for Immunogenicity Testing of Therapeutic Proteins released
December 2009.

Introduction

Biotherapeutics emerged in the 1970s as viable alternatives to


small molecule drugs when the advent of recombinant DNA
technology allowed for cost-efficient, large-scale manufacture of
proteins in cultured cells under controlled conditions. In 1982,
recombinant human insulin, developed by Genentech and Eli
Lilly, was the first biotechnology therapy to be approved by the
FDA. Since then the biotechnology industry has reached more
than 50 billion dollars in annual revenue, and continued growth
is expected as pharmaceutical companies shift more resources
Immune Response
into large molecule drug development, or make high-value
The principal function of an immune system is to protect
acquisitions of biologics companies. There are more than 200
the body against infection. For higher organisms this is
biotechnology products currently on the market with 400-plus
accomplished with the innate (immediate, non-specific) and
additional therapies in clinical trials targeting diseases such as
adaptive (specific, antibody-mediated) immune response. The
cancer, Alzheimers, rheumatoid arthritis, multiple sclerosis,
immune system must distinguish the organisms own cells, tissues
and HIV/AIDS. In 2014, it is predicted
and proteins from non-self antigens,
that 50% of the top 100 drug sales will
although in autoimmune disorders such
Biologics have the potential to induce an
be biologics. This is a significant increase
as rheumatoid arthritis and systemic
antibody-mediated immune response that
from 28% in 2008 and 11% in 2000
lupus erythematosus the immune system
could produce severe consequences for
[EvaluatePharma 2009].
humansthus, immunogenicity testing is
targets self tissues and proteins. Most
an important part of developing
non-self antigens will elicit an antibodylarge-molecule
therapies.
mediated immune response, termed
In contrast to chemically synthesized
immunogenicity, in animals and humans.
drugs, biotherapeutics such as proteins,
This adaptive response to a particular
peptides, and antibodies generally possess
antigen is heterogeneous; no two biological systems react in the
large and complex structures often modified by glycosylation
same manner, and the response of a single system will change
or pegylation, and may be non-human in origin. These
over time. The immune response is used advantageously in
characteristics greatly increase the potential to induce an
vaccinations, however, a primary concern in the development
antibody-mediated immune response over their small-molecule
of non-vaccine therapeutics is understanding, measuring, and
counterparts, and in consequence may cause severe illness
mitigating an unwanted, drug-induced antibody response.
in humans. Real-world examples of therapeutics that have
caused serious health consequences for healthy volunteers and
patients include erythropoietin [Casadevall, 2002], factor VIII
The myriad of factors that cause immunogenicity within a
[Lavigne-Lissalde, 2005], and insulin therapy. Immunogenicity
particular system can be categorized as drug-related or
testing is thus an important part of developing a large-molecule
patient-related. Some examples of drug-related factors
drug for eventual FDA licensure and marketing. Worobec and
are related to protein structure such as alterations due to
Rosenberg state, the paramount concerns are safety and
pegylation, glycosylation, and oxidation of the molecule. While
Page 1

pegylation is often used to increase the bioavailability of


population who lacks Factor VIII protein, and thus will label
drug and reduce immunogenicity, anti-PEG-antibodies have
the drug therapy non-self and xenobiotic in origin.
been detected and in some cases shown to be associated with
changes to pharmacokinetics (PK) data and loss of efficacy
Impact of Immune Response
[Armstrong, 2007]. Structural characteristics of a protein
Since a critical part of the drug development process is to
such as repetitive amino acid sequences
accurately assess drug exposure to
(whether in secondary or tertiary
determine bioavailability in the test
structure) and the tendency to form
The extent, duration, and severity of the
species, it is important to understand
aggregates can affect immunogenicity
immune response can have a wide range
how the presence of anti-drug
rates as the immune system is primed
of clinical consequences from little or no
antibodies (ADA, also anti-therapeutic
to respond to aggregates and other
effect, to severe and life-threatening illness.
antibodies, ATA) may impact this
patterned, repetitive sequences.
assessment. The antibodies produced
Additionally, a particular drug meant
as a result of drug challenge may be
to mimic an endogenous counterpart may differ slightly
sustaining, clearing, or neutralizing, all of which can affect the
in sequence, causing the immune system to recognize the
pharmacokinetic and pharmacodynamic properties of a the drug.
therapeutic as non-self. A second drug-related factor is
Sustaining antibodies prolong the drug half-life by protecting
related to manufacturing process and drug formulation.
it from normal clearance; in contrast, a clearing antibody more
For example, drug production in eukaryotic or prokaryotic
quickly decreases drug concentration below therapeutic levels
systems may introduce immunogenic foreign proteins into the
or removes drug from circulation. Binding antibodies may not
product. Furthermore, the formulation itself may increase the
clear a drug, but can interfere with pharmacokinetic analysis
propensity towards aggregation as well as contain excipients or
by preventing measurement of the therapeutic. Neutralizing
impurities that induce an immune response.
antibodies abrogate a drugs therapeutic effect and potentially

worsen the patients disease if not also inducing serious sideeffects. The extent, duration, and severity of the response can
have a wide range of clinical consequences from little or no
effect, to severe and life-threatening illness. The most wellknown example of severe illness is pure red cell aplasia in which
neutralizing antibodies targeted against erythropoietin cross
react with the endogenous protein. Finally, the presence of ADA
can lead to other clinically important adverse events such as
hypersensitivity reactions, anaphylaxis, serum sickness, and severe
infusion reactions.

Other drug-related factors include the route of administration,


dose level, and frequency of dosing. It has been generally
accepted that subcutaneous (SC) exposure is more
immunogenic than either intramuscular (IM) or intravenous
(IV) dosing. However, anecdotal evidence presented at
a recent conference on immunogenicity (Boris Gorovits,
Immunogenicity Summit, October 2009) indicates that this
type of ranking is not necessarily supported by the data. A
paper exercise evaluating data on 20 biotherapeutics of various
classes indicated that in many cases, the rate of immunogenicity
by IV exposure was equal to SC exposure. Given that many
exceptions exist, specific testing of individual products is
encouraged [Koren, 2008; Ponce, 2009]. Dose and frequency
of drug administration effects are assessed more generally, as
these are not truly independent of other factors. A single dose
is usually not expected to produce a robust or well-developed
immune response compared with multiple doses over time.
However, frequent and high concentration dosing regimens is
one mechanism used to induce drug tolerance in patients.

The early immune response typically comprised of low affinity,


low concentration IgM molecules, is of limited magnitude, and
peaks within 7-14 days. As the immune response matures, often
with repeated drug (antigen) exposure, isotype switching leads
to a more robust response consisting largely of IgG antibodies.
The presence of IgG4 indicates a well-entrenched immune
response, with high affinity antibodies that are more likely to
neutralize the effect of drug in higher concentrations.
Understanding the antibody response to drug exposure is
necessary to evaluating the significance of that response.
Determining the magnitude of response is important in
understanding and ultimately determining the safety and
efficacy of a particular drug which may include:
Response duration - whether sustained or transient
Occurrence of adverse events such as allergic reactions
Evaluating the production of neutralizing antibodies
Correlating change in PK/TK profiles

Patient or host-related factors, such as genetic disposition,


immune status, disease state, and the presence of
co-medications play a role in the immunogenicity rate. For
example, a healthy individual will have a different immune
response to a given drug than that of the targeted disease state
population. For example, Factor VIII drug therapy is unlikely
to induce the same response in healthy subjects whose immune
system is more likely to see the drug as self than the disease

Page 2

The challenge then becomes developing sensitive and specific


immunogenicity assays to determine the presence, specificity,
magnitude, and neutralizing potential of ADA.

levels of true positives. The screening cutpoint is determined by


a statistical evaluation of a drug-nave population representative
of the study samples. Any false positives should then be
distinguished from true positive (reactive) samples through a
competitive assay, termed confirmation assay, with free drug.
The specificity cut point is determined during evaluation of
the screening cut point by incubating the same nave samples
(or, alternately, nave samples spiked with low positive control)
with an excess of drug and evaluating the reduction in signal
of spiked versus unspiked samples. A detailed explanation for
determining both the screening and confirmatory cutpoints can
be found in Shankar, et al.

Methodology for Immunogenicity


Testing and Analysis
A typical immunogenicity analysis follows a tiered approach
that includes three types of analysis (Figure 1): an initial screen,
in which study samples are assessed for the presence of ADA, a
confirmation assay, which determines whether putatively positive
samples react specifically with free drug (versus nonspecific
reactivity), and a titer evaluation that provides a relative level
of ADA. When assessment of neutralizing antibody potential
is required, a fourth type of assay, most often cell-based, is
implemented to determine whether ADAs in confirmed positive
samples specifically prevent drug from binding to its target.

Screening
Assay

Negative

Positive

Confirmatory
Assay

Positive

Immunogenicity assessments can be made using a number of


analytical platforms such as:
Enzyme-linked Immunosorbent Assay (ELISA)
Radioimmunoassay (RIA)
Electrochemiluminescence (ECL)
Surface Plasmon Resonance (SPR)

Characterization:
Neutralizing
Titer
Domain Specificity
Ab Class

Each of the platforms has advantages and disadvantages; a


well-thought out development plan will take into consideration
these practicalities in designing the immunoassay. For example,
ECL assays are often the platform of choice as a homogenous
bridging assay. ECL eliminates multiple wash steps and increases
the ability to detect low affinity antibodies as well as various
isotypes. For further characterization of the immune response,
SPR, such as the Biacore system, may be used to determine the
specific antibody isotype or binding characteristics.

Negative

Figure 1. Tiered approach for immunogenicity sample analysis. The screen


assay to determine whether samples are positive or negative for the
presence of ADA is performed first, typically followed by a confirmatory
analysis, then further characterization such as ADA titer and drugneutralizing potential.

An inadequately designed ADA assay can hinder the


ADA assays are considered qualitative (or sometimes quasiappropriate assessment of clinical findings and delay
quantitative) because the positive control does not accurately
product development or marketing. The ability to correlate
reflect the heterogeneity of immune system response, or,
immunogenicity and clinical events therefore relies upon a
usually, originate from the species of
properly developed and validated assay.
interest. The positive control is often a
The approach to immunogenicity testing
An inadequately designed ADA assay
monoclonal antibody or polyclonal sera
has undergone significant changes in
can hinder the appropriate assessment
derived from hyper-immunization of
recent years. For example, Shankar et al.,
of clinical findings and delay product
a particular species, and will vary from
(2008) published a more concrete plan for
development or marketing.
the study sample in terms of affinity,
developing and validating reliable testing
avidity, specificity, and isotype. Given
methods than was previously considered.
the
inherent
dissimilarity
between positive control and study
Specifically they expanded upon previous industry opinions
sample, the use of mass units in defining the level of response
[Mire-Sluis, 2004] by recommending a more statistical and
is of limited value, and not recommended by the FDA [U.S.
objective approach in evaluating certain method parameters
Dept. Health and Human Services, 2009]. The purpose of
such as cutpoint and confirmatory cut-off limits.
a positive control is therefore to demonstrate the method
The screening cut point is defined as the level at and above
functionality and monitor assay performance. The ideal positive
which a sample is considered positive (or reactive) for ADA
control is derived from the species of interest, which is usually
and below which a sample is considered negative. A risk-based
not possible for human subjects. If the positive control is from
approach requires a cutpoint selection that leads to a 5% false
a different species, it should be diluted into the matrix of
positive rate as it is more appropriate for a certain number of
study samples. In method design and execution, low, high, and
samples be considered false positive than false negative. The
negative controls are used to monitor assay performance. A high
false positive samples can then be proven non-reactive in the
positive control with signal at the upper end of linear response
confirmatory assay ensuring that the method can detect low
is useful in monitoring method performance over time, reagent
Page 3

qualifications, and troubleshooting, or when a method has a


demonstrable hook effect. In contrast, it is the low positive
control that ensures reliable assay performance and provides
confidence that the assay can detect low levels of ADA.

clinical immunogenicity rate was only 12% [Ponce, 2009]. As


many large molecule drugs are human in origin, it is expected
that the animal species will recognize these as non-self proteins
and mount an immune response. The development of ADA and
magnitude of the response can have clear impacts on the ability
to interpret study findings. As discussed earlier, antibodies
can enhance the clearance of drug (reducing drug exposure
and limiting efficacy, as well as altering PK and PD profiles),
sustain bioavailability which may result in increased toxicity
or increased efficacy, or neutralize the drugs pharmacological
effect which can decrease efficacy and toxicity. Thus the main
purpose of evaluating immunogenicity in animals is to help
interpret the PK and PD properties of the drug, as well as any
toxicity findings, or more simply, ADA + PK + PD + toxicity =
immunogenicity assessment. As a result, careful consideration
should be made when selecting appropriate animal models.

Both drug quantitation and ADA determination assays are


subject to interference from matrix components. For drug
quantitation assays this includes the presence of ADA; ADA
assays are similarly prone to interference from free drug.
Antibody drugs in particular have long half-lives, and ADA
may stabilize drugs thereby prolonging the half-life. Effort is
made to minimize these interferences as much as possible: time
points for ADA analysis are typically chosen when the drug is
in the elimination phase, which may be months following drug
challenge, or the assay format may include an acid-dissociation
step to remove or decrease antibody:drug complexes. The affect
of drug on antibody detection, e.g. tolerance, in an analytical
Accurate assessment of ADA in study samples is impacted by
method is evaluated by incubating increasing levels of drug
several factors, including the concentration of drug in matrix
with the low positive control. The lowest concentration of drug
and the choice of positive controls. The effect of high drug
that prevents detection of ADA is termed the drug tolerance;
concentration can sometimes be minimized by the assay format,
drug tolerance depends, like sensitivity, upon the particular
or by the use of techniques such as acid dissociation to decrease
positive control chosen. It is therefore suggested that ADA
drug:ADA complexes. The other limitations of an ADA
method validations are designed to appropriately assess the
method are those imposed by the choice of positive control,
drug tolerance over the range of possible drug concentrations
which will not adequately represent the heterogeneity of the
that may be seen in test samples. Caution is required, however,
study samples. A biological system mounts a polyclonal immune
when interpreting results of study samples with measurable
response in which antibodies produced are of differing affinity
quantities of drug; since drug tolerance is dependent upon the
and avidity. Excellent method
chosen positive control which
drug tolerance and sensitivity
itself is not representative of all
The main purpose of evaluating immunogenicity in animals
may be illustrated with the use
possible immune responses the
is to help interpret the PK and PD properties of the drug,
of a high affinity monoclonal
assessment of ADA (positive or
as well as any toxicity findings:
antibody control, but this is
negative) and the corresponding
ADA + PK + PD + toxicity = immunogenicity assessment.
not reflective of the biological
level (titer) may not be accurate.
response, which likely contains
Similarly, since ADA may
many high affinity antibodies and a host of low affinity/avidity
interfere with quantitation of drug, the concentration of drug
antibodies. The drug tolerance and sensitivity obtained from
obtained from a particular sample may not be a true reflection
method validation should not be applied to study samples,
of the actual level.
or used cautiously with careful consideration of the type of
positive control and its similarity to the biological response; very
Preclinical Implications of
different sensitivities and drug tolerance results will be obtained
Immunogenicity
with a polyclonal preparation versus a single high-affinity
Until recently, the focus of regulatory agencies and opinion
monoclonal antibody control.
papers has largely been the design and application of
immunogenicity studies on human subjects, but an important
Immunogenicity assessment in animals is best used to interpret
part of any drug development program are preclinical
study findings and assess the validity of toxicology studies.
toxicology studies in which the effects of drug are evaluated
During protocol development, blood samples should be
in animal models. The particular choice of animal will depend
collected for any study in which animals are exposed to drug
on the drug characteristics and physiological function so as to
for greater than seven days. In an effort to manage sample
better understand the mechanism of action and effect in vivo as
analysis, Ponce, et al. (2009) provided a decision tree that guides
well as to assess toxicity. However, animal models are generally
the investigator on whether ADA analyses will meaningfully
poor predictors of immunogenicity in humans. For example, the
contribute to data interpretation. For example, if suitable PK
recombinant human monoclonal antibody Humira produced
assays and PD biomarkers are available, and no change in
a 100% induction of ADA in cynomolgus monkeys yet the
profile is seen, then an ADA screen may not be warranted. If
Page 4

however, changes are observed, then at minimum the screening


assay should be performed, followed by titer if shown positive
for ADA. Though a tiered approach (screen, confirmation, titer)
is recommended for clinical sample analysis, confirmation is
generally not needed in animal studies, nor is the fourth tier of
neutralizing antibody assessment.

Risk
Level

Drug
Characteristics

Examples

Consequences

Low

Not structurally
identical to
endogenous
protein

Enzymes

Infusion site
reactions

Antibody drugs

Mild allergic
reactions

Non agonistic

Clinical Implications of
Immunogenicity

Medium Partially or
completely
identical to
endogenous
protein

It is widely understood that the induction of immunogenicity


in animal models is not predictive of a potential for antibody
formation in humans [ICH S6 Guidance] and thus the
immunogenicity rate in a particular animal model does not
necessarily preclude the initiation of clinical studies. An
approach that takes into account a number of factors including
the causes and consequences of immunogenicity is applied to
any new biotherapeutic under consideration for marketing and
licensure; proper risk assessment considers the probability of
generating an immune response and the consequences of such a
response [Rosenberg; Koren, 2008]. The risk is then categorized
as low, medium, or high and the bioanalytical testing strategy is
defined accordingly [Shankar, 2007].

Loss of efficacy

Replacement
therapy as
Factor VIII
Antibody drugs

Endogenous
counterpart is
either missing or
redundant

Same as low
risk
Overstimulation
of endogenous
mechanism
Immune
complex
formation

or
Not structurally
identical to
endogenous
protein/agonistic
High

Partially or
completely
identical to
endogenous
protein

Erythropoeitin
GM-CSF

Same as
medium risk
Neutralization of
endogenous
counterpart

To appraise the probability of an immune response, the causes


Endogenous
Autoimmune
of immunogenicity discussed under the Immune Response
counterpart is
deficiency
section above are considered along with other factors such as:
not redundant
Presence of drug conjugates (as to a toxin)
Exposure of neoantigenic epitopes upon drug denaturation
The type of risk inherent to a particular drug dictates the
or fragmentation
testing and sampling strategy. Low and medium risk drugs
Natural tolerance or use of immunosuppressive drugs
should have ADA samples taken frequently in Phase I
Length of drug treatment
and Phase II trials to help understand the human immune
Homology of drug product to endogenous counterpart
response to the drug; testing may be less frequent in Phase
Redundancy of an endogenous
III. By contrast, a high risk drug, one
counterpart
with potential to cross react with and
Mechanism of action
Proper risk assessment considers the
neutralize an endogenous counterpart,
probability of generating an immune
might require frequent sampling and
response and the consequences of
Following assessment of immunogenic
real-time ADA analysis (including
such a response.
potential, consideration of the possible
neutralization potential) at all stages of
clinical sequelae, as well as whether the
clinical development, or sequential patient
target indication is a life-threatening
dosing (rather than a cohort design) to better mitigate risks.
disease is needed to develop a risk-management plan. In mildAt a minimum, all immunogenicity assessments during clinical
to-moderate responses to ADA, the patient may experience
trials should include screen, confirmatory, and titer analysis.
loss of drug efficacy, or allergic hypersensitivity reactions such
The timing for implementation of a neutralizing antibody
as an infusion-site reaction. More serious consequences include
assay, and type of format required, will vary with drug product
overstimulation of endogeneous mechanisms such as that
and associated risk to the patient. A cell-based assay for a
following treatment with TGN1412 [Medicines and Healthcare
monoclonal antibody designated as low risk may be of limited
Products Regulatory Agency, 2006], anaphylactic shock, or
value (while taking considerable time and effort to develop and
cross reactivity with, and neutralization of endogenous protein
validate).
leading to an autoimmune deficient syndrome (Table 1).
Approaches to attenuate immunogenicity and minimize
patient risk include drug redesign (to remove epitopes of

Table 1. Risk assessment for clinical immunogenicity.

Page 5

high immunogenic potential), change in dose and/or dosing


procedure (IV administration versus inhalation), and exclusion
of sensitive subjects (a patient with known allergies to a similar
product could be prescreened with a skin test, or be administered
drug in a clinical setting where severe allergic reactions can be
readily treated). Once immunogenicity is observed, the clinician
must decide the best path forward for a patient. If ADA inhibits
the function of a life-saving therapy, it may be necessary to
institute a tolerizing regimen to inure the immune system
against the therapeutic. These decisions are made on a case-bycase basis with consideration of all available information.

production of a sound risk-assessment strategy and a fit-forpurpose approach in testing of preclinical and clinical samples.
Whatever the application, properly designed and validated
immunoanalytical methods provide confidence in the data used
for these purposes.

References
Armstrong, J.K., Hempel, G., Koling, S., Chan, L.S., Fisher,
T., Meiselman, H.J., and Garratty, G. Antibody Against
Poly(Ethylene Glycol) Adversely Affects PEG-Asparaginase
Therapy in Acute Lymphoblastic Leukemia Patients. Cancer.
2007, 110(1) 103-111.

Follow-on Biologics

Casadevall, N., Nataf, J., Viron, B., Kolta, A., Kiladjian, J.J.,
Martin-Dupont, P., Michaud, P., Papo, T., Ugo, V., Teyssandier,
I., Varet, B., and Mayeux, P. Pure Red-Cell Aplasia with
Antierythropoietin Antibodies in Patients Treated with
Recombinant Erythropoietin. New Engl. J. Med. 2002, 346(7),
469-475.

Follow-on biologics, or biosimilars, are large-molecule


therapeutics similar to a licensed drug, but manufactured and
marketed by a third-party following patent expiration of the
innovator molecule. While there is currently no mechanism
for approval and licensure of biosimilars in the US, the EU
and Japan both have a legal, regulated process that includes
assessment of immunogenicity between the innovator drug and
follow-on biologic at all stages of the development process to
ensure comparable quality, safety, and efficacy.

EvaluatePharma, Biotech set to dominate drug industry


growth. 2009. (http://www.evaluatepharma.com/Universal/
View.aspx?type=Story&id=188700&sectionID=&isEPVantage
=yes)

Paramount in assessment of immunogenicity is that the


methods show comparable sensitivity and specificity. Key
questions to consider when developing a single method that
shows equal sensitivity and specificity to both the innovator
and biosimilar drug products [U.S. Dept. Health and Human
Services, 2009] include the following:
What is best the assay format and positive control to use?
If a bridging assay is employed, should the capture and
detection reagents be innovator molecule or biosimilar?
Should the positive control be one generated from the
reference product or follow-on biologic, or perhaps some
combination of both?
If a single method can not be developed for
immunogenicity comparison, then do the separate methods
show equivalent sensitivity and specificity?

de Galan, B.E., Simsek, S., Tack, C.J., and Heine, R.J. Efficacy
and safety of inhaled insulin in the treatment of diabetes
mellitus. Neth. J. Med. 2006, 64(9), 319-325.
Koren, E., Smith, H.W., Shores, E., Shankar, G., Finco-Kent,
D., Rup, B., Barrett, Y.C., Devanarayan, V., Gorovits, B.,
Gupta, S., Parish, T., Quarmby, V., Moxness, M., Swanson,
S., Taniguchi, G., Zuckerman, L.A., Stebbins, C.C., and
Mire-Sluis, A. Recommendations on risk-based strategies
for detection and characterization of antibodies against
biotechnology products. J. Immunol. Methods. 2008, 333, 1-9.
Lavigne-Lissalde, G., Schved, J. Granier, C., and Villard, S.
Anti-factor VIII antibodies: a 2005 update. Thromb. Haemost.
2005, 94(4), 760-769.

Answers to these and other questions are important to


distinguish between the effects of different manufacturing
processes, purity of biosimilar versus originator therapeutic, or
biosimilar itself from differences in assay performance.

Medicines and Healthcare Products Regulatory Agency,


Investigations into Adverse Incidents during Clinical
Trials of TGN1412. 2006. (http://www.circare.org/foia5/
clinicaltrialsuspension_interimreport.pdf )

Conclusion
In contrast to new chemical entities, biotherapeutic drugs are
potentially immunogenic, and, in rare cases, treatment with
such products can lead to severe and devastating illnesses in
humans. Therefore, the importance in understanding how
immunogenicity affects drug exposure, efficacy, and toxicity
at all stages of the drug development process can not be
overstated. The complexity of the immune response necessitates

Mire-Sluis, A.R., Barrett, Y.C., Devanarayan, V., Koren, E.,


Liu, H., Maia, M., Parish, T., Scott, G., Shankar, G., Shores,
E., Swanson, S.J., Taniguchi, G., Wierda, D., and Zuckerman,
L.A. Recommendations for the Design and Optimization
of Immunoassays used in the Detection of Host Antibodies
Against Biotechnology Products, J. Immunol. Methods. 2004,
289(1-2), 1-16.
Page 6

Ponce, R., Abad, L., Amaravadi, L., Gelzleichter, T., Gore,


E., Green, J., Gupta, S., Herzyk, D., Hurst, C., Ivens, I.A.,
Kawabata, T., Maier, C., Mounho, B., Rup, B., Shankar, G.,
Smith, H., Thomas, P., and Wierda, D. Immunogenicity of
biologically-derived therapeutics: Assessment and interpretation
of nonclinical safety studies. Reg. Tox. Pharm. 2009, 52(2),
164-182.

U.S. Dept. of Health and Human Services, Food and Drug


Administration, Guidance for Industry: Assay Development
for Immunogenicity Testing of Therapeutic Proteins, Draft
Guidance. FDA-2009-D-0539, 2009.
Worobec, A., and Rosenberg, A.S. A Risk-Based Approach to
Immunogenicity Concerns of Therapeutic Protein Products,
Part 1: Considering Consequences of the Immune Response to
a Protein. BioPharm International. 2005, 17, 22-26.

Proceedings of the International Conference on Harmonization


(ICH) of Technical Requirements for Registration of
Pharmaceuticals for Human Use. Tripartite guideline S6
Preclinical Safety Evaluation of Biotechnology-Derived
Pharmaceuticals. 1997. (http://www.ich.org/LOB/media/
MEDIA503.pdf )
Rosenberg, A.S., and Worobec, A. A Risk-Based Approach to
Immunogenicity Concerns of Therapeutic Protein Products,
Part 2: Considering Host-Specific and Product-Specific Factors
Impacting Immunogenicity. BioPharm International. 2004, 17,
34-42.
Rosenberg, A.S., and Worobec, A. A Risk-Based Approach to
Immunogenicity Concerns of Therapeutic Protein Products,
Part 3: Effects of Manufacturing Changes in Immunogenicity
and the Utility of Animal Immunogenicity Studies. BioPharm
International. 2005, 17, 32-36.
Shankar, G., Pendley, C., and Stein, K.E. A risk-based
bioanalytical strategy for the assessment of antibody immune
responses against biological drugs. Nat. Biotech. 2007, 25(5),
555-561.
Shankar, G., Devanarayan, V., Amaravadi, L., Barrent, Y.C.,
Bowsher, R., Finco-Kent, D., Fiscella, M., Gorovits, B.,
Kirschner, S., Moxness, M., Parish, T., Quarmby, V., Smith, H.,
Smith, W., Zuckerman, L.A., and Koren, E. Recommendations
for the validation of immunoassays used for detection of host
antibodies against biotechnology products. J. Pharm. Biomed.
Anal. 2008, 48(5), 1267-1281.

Covance is an independent, publicly held company with headquarters in Princeton, New Jersey, USA.
Covance is the marketing name for Covance Inc. and its subsidiaries around the world.
The Americas +1.888.COVANCE (+1.888.268.2623) +1.609.419.2240
Europe/Africa +800.2682.2682 +44.1423.500888
Asia Pacific +800.6568.3000 +65.6.5677333
www.covance.com
CSBIO007-1210

Page 7

Anda mungkin juga menyukai