You are on page 1of 7

Continuing Education Course: Clinical Pathology of Biotherapeutics

Toxicologic Pathology
2018, Vol. 46(8) 1013-1019

Immunogenicity and Immune Complex ª The Author(s) 2018


Article reuse guidelines:
sagepub.com/journals-permissions
Disease in Preclinical Safety Studies DOI: 10.1177/0192623318797070
journals.sagepub.com/home/tpx

John L. Vahle1

Abstract
This article summarizes a continuing education presentation on immunogenicity that was part of a continuing education course
entitled, “Clinical Pathology of Biotherapeutics.” Immunogenicity of a biotherapeutic can have diverse impacts including altered
systemic exposure and pharmacologic responses and, in a fraction of the cases, safety concerns including cross-reactive neu-
tralization of endogenous proteins or sequela related to immune complex disease (ICD). In most cases, immune complexes are
readily cleared from circulation; however, based on physiochemical properties, insoluble complexes form, activate complement,
and deposit in tissues. Using published information and personal experience, a set of repeat-dose monkey toxicity studies with
manifestations suggestive of ICD was reviewed to summarize the spectrum of clinical and pathology findings. The most common
live-phase observation linked to ICD was an acute postdosing reaction following multiple dose administrations characterized by
generalized collapse and attributed to acute complement activation. Less common live-phase observations were related to syn-
dromes such as a consumptive coagulopathy or a protein losing nephropathy. The most common histologic change attributed to
ICD was multi-organ vascular/perivascular inflammation followed by glomerulonephritis. The presentation concluded with a
description of the challenges in assessing the relevance of immunogenicity-related reaction in monkey to human clinical use.

Keywords
immunogenicity, toxicity, immune complex disease, nonhuman primate, biotherapeutics

Introduction Rojko et al. 2014; Mease, Kimzey, and Lansita 2017; De Groot
and Scott 2007; Krishna and Nadler 2016) and served as an
This article summarizes one of the four presentations at a important basis for the presentation.
Continuing Education Session held in association with the
Society of Toxicologic Pathology’s Thirty-seventh Annual
Symposium. The Continuing Education Session focused on the Immunogenicity: An Introduction
clinical pathology evaluation of biotherapeutics in the context
For the purposes of this review, immunogenicity was defined
of nonclinical safety studies. The primary objective of this
as the ability of a therapeutic agent to induce an immune
article was to review the potential impacts of immunogenicity
response. Immunogenicity is most frequently a concern for
on the nonclinical safety assessment of biotherapeutics.
protein-based therapeutics (hereafter referred to as biothera-
Following a brief introduction to the concepts of immunogeni-
peutics) and can occur in both animal and human settings
city including mechanisms of formation of antidrug antibodies
(Krishna and Nadler 2016; Ponce et al. 2009). The antibody
(ADAs), the remainder of the presentation focused on the
that is generated in response to a biotherapeutic has been
various manifestation of immunogenicity in nonclinical safety
variably referred to as an ADA, antitherapeutic antibody,
studies. Particular focus was placed on the development of primate antihuman antibody, or a human antihuman antibody,
immune complex disease (ICD) in nonhuman primates admi- and the general response has been referred to as an anti-
nistered monoclonal antibody-based constructs. The review antibody response. The potential for immunogenicity to
concluded by addressing some of the challenges in understand- limit the use of biotherapeutics, particularly monoclonal
ing the relevance of the immunogenicity findings in animals to
the safety of these agents in human clinical trials. This review
1
did not provide a detailed analysis of mechanisms of immuno- Lilly Research Laboratories, Indianapolis, Indiana, USA
genicity or immune complex formation and did not review
Corresponding Author:
acute (type 1) hypersensitivity reactions. Several excellent
John L. Vahle, Eli Lilly and Company, Lilly Corporate Center, Indianapolis,
reviews on nonclinical and clinical immunogenicity were high- IN 46285, USA.
lighted for the audience (Leach et al. 2014; Ponce et al. 2009; Email: jvahle@lilly.com
1014 Toxicologic Pathology 46(8)

Figure 1. The types of impacts that an immunogenic response to a biotherapeutic can have on a toxicity study. In some cases, very little impact is
noted as there is not an appreciable impact on systemic exposure or pharmacologic or toxicologic effect. At the other end of the spectrum are
those cases where immunogenicity can have substantial impact including morbidity or mortality, including but not limited to hypersensitivity
reactions such as immune complex disease.

antibodies, was an early concern for the field (Kuus-Reichel mechanisms (De Groot and Scott 2007), much of the focus
et al. 1994), and while immunogenicity remains an important regarding predicting immunogenicity is centered on identifying
topic, a wide array of monoclonal antibody and related protein T-cell epitopes that drive T-cell help and antibody production.
constructs has proven to be highly effective therapeutics for a
variety of disease states. In the clinical setting, immunogeni-
city can cause a variety of effects including loss of systemic Potential Impact of Immunogenicity
exposure, loss of the desired therapeutic response, and less on Animal Toxicity Studies
frequently safety concerns such as infusion reactions, cross- Similar to the clinical setting, immunogenicity can have a vari-
reactive neutralization of endogenous proteins that mediate ety of impacts in animal models including the toxicity studies
critical biologic functions, and ICD. When immunogenicity used to characterize the nonclinical safety profile of the
issues arise late in the clinical development process, they are biotherapeutic (Figure 1). Because the biotherapeutic is a
extremely costly to drug developers. As such, methods to human or humanized protein, the frequency and severity of the
predict and mitigate against clinical immunogenicity are immunogenic response can be greater in the animal model than
active areas of investigation (Yin et al. 2015). in the human clinical setting (Leach et al. 2014; Ponce et al.
It is important to recognize that many factors can affect 2009). Despite the increased “foreignness” for the human pro-
immunogenicity. Product-specific factors that affect immuno- tein in animals, in many cases, there may be no appreciable
genic potential often focus on the protein sequence of the con- immunogenicity in the animal model. While robust data are not
struct; however, in many cases, the formulation, aggregates, available on the frequency of immunogenicity in nonclinical
impurities, and therapeutic target play important roles. toxicity studies, pathologists frequently encounter studies of
Treatment-specific factors such as the dose, route, and fre- biotherapeutics where there is essentially no impact of immu-
quency of administration also influence immunogenic poten- nogenicity on the overall study outcome. In these cases, sys-
tial. Patient-specific factors such as the disease state, temic exposures are maintained at adequate levels to assess
concurrent illness or medications, and genetic background are toxicity, there is no loss of intended pharmacologic effects, and
also important and underscore how challenging it can be to no sequela related to hypersensitivity or ICD.
predict immunogenic potential. While the production of ADAs Antibody-mediated changes in the pharmacokinetic profile
can occur via both T-cell-dependent and T-cell-independent are one of the more common sequela of immunogenicity in
Vahle 1015

toxicity studies. Antibodies that bind to the biotherapeutic and influence the physiochemical properties of immune complexes
increase their clearance from systemic circulation are referred and dramatically effect clearance and deposition of immune
to as “clearing” antibodies and can markedly reduce systemic complexes. Lattice size is one of the key factors, and the large
exposure and interfere with the ability to assess toxicity in lattices can form when there is antigen: Antibody equivalence
either a single animal or entire dose groups. Various study is often insoluble and can lead to tissue deposition. Antigen and
designs can be considered in an effort to “dose through” immu- antibody ratios are not the sole determinant, as
nogenicity including increasing administered dose or altering the concentration, valence, and charge of both antigen and
dose frequency or route of administrations (Ponce et al. 2009). antibody can influence immune complex formation and clear-
While less common, there are clear examples where antibodies ance. While these are useful concepts to understand, multiple
to the therapeutic protein have decreased systemic clearance factors can influence immune complex formation, clearance,
resulting in increased systemic exposure (“sustaining anti- and deposition, and it is not possible to predict the potential for
bodies”). In these cases, prolonged circulation time may a given biotherapeutic at a given dose and dose frequency to
increase pharmacologic activity and complicate assessments induce ICD in animals.
of toxicity. In addition to altering systemic exposure, the anti- Immune complex formation leading to systemic and tissue
body to the biotherapeutic can bind at or near the target-binding effects is generally considered a type III hypersensitivity reac-
epitope and inhibit the biotherapeutics ability to bind to the tion in the classical Gel-Coombs classification scheme (Leach
desired target. These antibodies are referred to as “neutralizing et al. 2014); however, the pathophysiology is much more com-
antibodies.” Both neutralizing and clearing antibodies, by plex than simple intravascular deposition of insoluble immune
reducing the pharmacologic effects of the biotherapeutic, can complexes. As summarized by Krishna and Nadler (2016),
compromise the ability to assess the toxicity profile of the three key interrelated mechanisms contribute the reaction: (1)
therapeutic. To aid in the overall interpretation of the toxicity deposition of immune complexes in vasculature and sites of
study, it is important for the study pathologist to work with hyperfiltration leading to inflammation, (2) activation of com-
colleagues in toxicokinetics and immunology to assure an plement by the immune complex, and (3) cross-linking of FcgR
appropriate assay strategy is in place. These assays can include and complement receptor on the surface of a variety of immune
the measurement of the active circulating biotherapeutic, cells leading the release of inflammatory mediators and che-
detection of circulating ADA levels, and measurement of phar- motactive substances. Complement activation plays a key role
macodynamic end points in either the periphery or tissues. in both the systemic and the tissue effects noted in ICD, and
The biologic significance of immunogenicity is heightened key mediators are the generation of the anaphylatoxins C3a,
when the ADA neutralizes not only the administered biother- C4a, and C5a (Krishna and Nadler 2016).
apeutic but also the biologic function of a critical endogenous
protein (cross-reactive antibodies). For example, thrombocyto-
Findings Attributed to ICD in Nonhuman
penia and pancytopenia have been reported in patients follow-
ing the administration of recombinant human thrombopoietin
Primate Toxicity Studies
(Basser et al. 2002; Li et al. 2001). Clearly, in both animal and To summarize the range of live-phase, laboratory, and mor-
human settings, the development of cross-reactive antibodies to phologic findings that have been associated with ICD follow-
endogenous proteins can have life-threatening consequences. ing the administration of biotherapeutics to nonhuman
Finally, as discussed in more detail below, the formation of primates in toxicity studies, a group of 23 repeat-dose nonhu-
immune complexes and sequela can have major impact on the man primate studies was reviewed for this presentation. The
interpretation of toxicity studies. level of detail available for each study varied greatly as the
information was derived from publications, workshop presen-
tations, personal communications, and personal experience.
ICD—Overview of Mechanisms Key publications that either review a series of cases or present
A thorough review of the mechanisms of immune complex detailed results from a single-case experience include (Rojko
formation, clearance, measurement, and pathophysiology was et al. 2014), (Heyen et al. 2014), Husar et al. ( 2017), and
outside the scope of the presentation. The audience was Kronenberg et al. (2017). From the information available
referred to reviews of the topic by Rojko et al. (2014), an across the studies reviewed, key study design elements, live
investigation of anti-infliximab immune complexes by Rojas phase-findings, laboratory findings, and anatomic pathology
et al. (2005), and methods to measure circulating immune com- findings were summarized.
plexes by Pierog et al. (2015). Key concepts discussed include Of the 23 studies examined, 22 used different biotherapeu-
the fact that in health, levels of circulating immune complexes tics (two studies were reported for one of the biotherapeutics),
are relatively low due to two primary clearance mechanisms. and as expected, these were primarily monoclonal antibody-
One mechanism consists of clearance via the FcgRIIa receptor based biotherapeutics. In many cases, only general descriptions
on platelets, monocytes, and macrophages and is present across of the biotherapeutic were disclosed, so it was not possible to
rodent and primates. The other mechanism, which is specific to identify clear trends; however, ICD was reported in studies of
primates, involves shuttling of immune complexes via the com- both monospecific and bispecific antibodies and occurred
plement receptor 1on red blood cells. A variety of factors across various isotypes of antibody (IgG1, IgG2, and IgG4).
1016 Toxicologic Pathology 46(8)

There were insufficient data to determine whether a certain and, in a single case, d-dimers were analyzed and reported as
type of antibody construct was more prone to developing ICD. increased in concentration. Another syndrome that is likely
In terms of methods of dose administration, manifestations of associated with these acute postdose reactions included a sub-
ICD were reported across both intravenous and subcutaneous set of animals that had acute collapse postdosing, and the only
administrations and in studies using weekly, twice-weekly, and histologic change identified was acute thrombosis. The level
once-monthly dose administrations. Although the administered of detail reported in these studies did not allow a determina-
dose on a milligram per kilogram basis was not reported for tion whether these animals also had laboratory data that would
many studies, most studies reported the dose level (low, mid- have been consistent with a consumptive coagulopathy and
dle, and high) at which ICD was reported. Manifestations of disseminated intravascular coagulation. The live-phase obser-
ICD were observed across all dose levels and in various dose- vations described above occurred shortly postdose. This is in
level combinations. While it was difficult to assess the true contrast to the other important live-phase syndrome, nephro-
dose response in all the studies due to limited individual animal tic syndrome, in which there was a gradual onset of clinical
reporting, ICD did not generally present in a dose-responsive signs and laboratory findings. These animals were often first
manner. The incidence of ICD within a given study also varied detected based on live-phase observations of dependent
widely, and in 36% of the studies, only a single animal was edema, decreased body weight, and decreased food consump-
affected, whereas in 64% of the studies, multiple animals were tion and/or laboratory data of decreased serum albumins and
affected. increased urinary proteins. In each of these cases, histologic
In 13 of the 23 studies (57%), findings attributable to ICD examination revealed glomerular disease attributed to
were reported during the live phase. While the clinical signs immune complex deposition.
and laboratory findings reported were quite varied, for the Rojka et al. (2014) provided a thorough review of the his-
purposes of this Continuing Education Session, live-phase tologic findings associated with ICD as well as the immuno-
findings were grouped into broad syndromes. The most com- histochemical localization of various immune component with
mon presentation or syndrome was designated an acute post- tissues. In the current series of studies, the general pattern of
dose reaction. These reactions occurred shortly after dose histologic changes was consistent with that reported by Rojko
administration and most frequently occurred following the et al. Two of the 23 studies described clinical signs or labora-
fourth or fifth dose administration. Clinical signs reported in tory findings that were attributed to ICD and acute complement
nonhuman primates included hypoactivity, retching, emesis, activation but that did not have histologic lesions indicative of
excessive salivation, altered respiration, pallor, hypothermia, ICD. In the remaining 21 studies, in which histologic changes
and pruritus. As discussed in Rojko et al. (2014), these may be were attributed to ICD, perivascular and/or vascular inflamma-
a form of a generalized type III hypersensitivity reaction asso- tion was the most common histologic change occurring in 49%
ciated with acute complement activation. The clinical course of the studies. Within a given study, the individual animal
for these animals varied widely with some showing sponta- incidence of this change varied, and within an animal, the
neous recovery, other requiring veterinary intervention with number of tissues affected was also highly variable ranging
supportive case, and in other cases progressing to morbidity from a single vessel affected to widespread multi-organ invol-
requiring early euthanasia. While the type and timing of labora- vement. Consistent with the sites of predilection for immune
tory data obtained in these animals was variable, the more complex deposition, common sites were small to medium ves-
common observations included transient decreases in neutro- sels in the serosa or mesentery, tortuous vessels in the urogen-
phil and platelet counts, increased fibrinogen and triglyceride ital tract, sites of hyperfiltration (choroid plexus, glomerulus),
concentrations, and decreases in albumin concentration. Com- and the aortic root and coronary artery of the heart. In some
plement data were reported in five of the 23 studies, and while cases, immunohistochemistry for immune components was
the specific complement components analyzed varied, used to confirm a diagnosis of immune complex vasculitis.
increases in C3a, C4a, C4d, and SC5b-9 were reported. Cyto- Glomerulonephritis was less common than vasculitis in this
kine determinations were only reported in two of the 23 studies, series of cases and was reported in 16% of those studies in
and increased MCP1, Il-10, and IL6 were reported in one of which histologic lesions attributable to immune complex
these studies. occurred. The morphology of immune complex glomerulone-
Although likely part of a continuum associated with acute phritis has been well described previously, and in this case
complement activation, there were two additional acute live- series, key histologic findings reported were glomerular hyper-
phase syndromes identified in this case series. A subgroup of cellularity with hypersegmentation, increased mesangial
animals developed a consumptive coagulopathy. While many matrix, thickened peripheral capillary loops, and, in more
of these animals had clinical signs similar to those of the post- severe cases, synechia. Additional diagnostic methods were
dose reaction described above, additional live-phase observa- used in several of the studies and included localization of
tions included failure to clot at the phlebotomy site and/or immune components within the lesion and electron microscopy
petechial hemorrhage. While the laboratory data collected var- to identify electron-dense deposits and further characterize the
ied according to the specific study, laboratory findings reported glomerular change. Other changes reported but not described in
in these animals included reductions in platelet counts, pro- detail included fibrin thrombi in vessels not associated with
longed coagulation times, decreased fibrinogen concentration, vascular/perivascular inflammation and an increase in the size
Vahle 1017

Figure 2. Various end points and approaches study teams can consider in developing a weight-of-evidence assessment regarding immune
complex disease in animal toxicity studies. The author emphasized a case-by-case approach to determining which end points are most appropriate
for a given animal or study.

and/or number of Kupffer cells in the liver. In more advanced study personnel to determine whether different animals are
cases with severe vasculitis, a variety of secondary changes having similar presentations.
were reported including edema, hemorrhage, and necrosis of The routine end points included in standard repeat-dose
adjacent tissue. toxicity studies (live-phase observation, hematology, clinical
chemistry, urinalysis, coagulation panels, anatomic pathol-
ogy, and toxicokinetic data) provide a strong database and
Diagnostic Approach to ICD in some cases may be sufficient to provide a high index of
The author provided a brief overview of the diagnostic tools suspicion that immunogenicity and ICD are present. Other
that can be used to investigate findings that may be attributable assays that are often critical in the overall weight-of-
to ICD (Figure 2). For many of the laboratory alterations that evidence assessment include ADA analysis, assessment of
may be of interest such as detection of acute phase responses or pharmacodynamic markers to assess whether active drug is
acute complement activation, sampling timing is critical. As present, detection of circulating immune complex levels,
such, during the study design phase, the study team can con- complement and cytokine determinations, and an expanded
sider a sample banking strategy where the appropriate matrix panel of acute phase reactants.
(serum and/or plasma) is banked both prestudy (to establish With respect to tissue-based end points, immunohistochem-
individual animal baseline values) and at various intervals both ical or immunofluorescent detection of immune components in
pre- and postdose during the study. In those cases where there the tissue of interest can be helpful and in some cases can
are unexpected events and prospective samples were not provide a definitive diagnosis of ICD. Methods are available
obtained, residual samples from toxicokinetic or routine clin- to detect not only various immunoglobulin and complement
ical pathology samples may be available. An advantage of the components from the monkey (e.g., monkey IgG, IgM, C3) but
toxicokinetic samples, if available and in the appropriate also to localize the therapeutic antibody within the tissue of
matrix, is that they are typically obtained at multiple intervals interest. Other morphologic approaches that may be warranted
postdose. In addition to laboratory data, having timely and include electron microscopy and special stains (e.g., Periodic
consistent recording of live-phase observations is important Acid Schiff [PAS] on thin sections) to better characterize glo-
in those situations where one or more animals develop clinical merular disease.
signs postdose administration. The live-phase observer should One of the most important aspects of the diagnostic or inves-
strive to use consistent terminology across the study to allow tigative approach is, for the study team and the study
1018 Toxicologic Pathology 46(8)

pathologists in particular, to bring a strong diagnostic skill set morbidity and lacked a consistent correlation to altered sys-
and differential diagnostic approach to the study. While it can temic exposure might warrant additional analysis such as asses-
be tempting to default to a diagnosis of ICD for any postdose sing complement activation or attempting to localize immune
reaction or vascular change that occurs in a monkey that has components in the tissue.
developed an immunogenic response, the study team needs to
carefully rule out, as best as possible, that the changes are not
either (a) directly attributable to the test article or (b) a sponta-
Challenges in Assessing Human Relevance
neous event unassociated with the test article. Another impor- The presentation concluded with a brief discussion on how to
tant challenge for the study team to deal with is the fact that in assess the clinical relevance of immunogenicity-related find-
many cases, not all of the individual end points will correlate ings in animal toxicity studies. To frame the discussion, three
for a given animal. Because no single diagnostic test has per- divergent approaches were reviewed. On one end of the spec-
fect specificity and ICD manifests in a variety of ways, these trum was an approach that considers animal immunogenicity-
cases often remain diagnostic and interpretive challenges. A related findings as not relevant to humans, since an immune
relatively straightforward case might present with multiple epi- response to a human or humanized protein is expected to be
sodes of acute postdose reactions, significant loss of systemic greater in animals than in humans. In this approach, the animal
exposure, high titers of ADA, postdose complement activation, findings may not be used in the determination of a study no-
and histologic evidence of vasculitis and glomerulonephritis. In observed-adverse-effect level and summary clinical regulatory,
the studies summarized above, this type of “perfect con- and clinical documents might not discuss the animal findings in
cordance” was relatively rare. For example, at the high doses detail. A downside of this approach is that it may be overly
administered in some toxicity studies in an effort to overcome definitive in using language such as “not relevant” and may not
immunogenicity (“dosing through strategy” [Ponce et al. provide the appropriate degree of transparency and interpreta-
2009]), despite considerable immunogenicity and accelerated tion of this complex phenomenon. At the other end of the
clearance of the biotherapeutic, systemic exposure may not be spectrum is an approach that suggests since humans can
dramatically impacted. In addition, assays for ADA may not be develop immune responses to human or humanized protein
positive due to the design of the assay with respect to drug therapeutics and since ICD can occur in humans, the animal
tolerance. Although not clearly reported in the case series findings are a key safety finding and should be used to deter-
examined, in the author’s experience, there is often not a direct mine human dose levels and drive clinical safety monitoring
correlation between ADA titer and the presence and/or severity strategies. A downside of this approach is that it does not
of ICD. Some biotherapeutics can induce high ADA levels with incorporate sufficient perspective regarding the generally poor
marked reduction in systemic exposure and did not result in any predictive value animals have with respect to immunogenicity
evidence of ICD in the toxicity study. in general, and it may create greater clinical and/or regulatory
Given these challenges, it can be difficult to determine the concern for the findings than is warranted.
appropriate diagnostic approach for any given study. Labora- The author suggested an intermediate approach that indicates
tories and sponsors appear to vary widely in the intensity in that while animal studies are often not predictive of immuno-
which they investigate these findings. The author encouraged a genicity and sequelae related to immunogenicity in humans, the
case-by-case determination and suggested the study team con- animal findings should be carefully considered as part of the
sider a variety of factors in determining what level of evidence overall safety assessment. For example, if there is an on-target
is warranted. Key questions for consideration are: mechanistic basis for enhanced immunogenicity in animals, the
findings might be considered more predictive for humans. In
 What is the severity of the findings? addition, for novel constructs that may be considered more
 Did the findings result in morbidity or mortality? “foreign” to both animals and human and for which there is
 What are the incidence and dose responsiveness of the limited clinical experience, additional caution might be war-
findings across the study? ranted. One important point to keep in mind with this approach
 What is the overall anatomic distribution of the findings is that because immunogenicity does not generally occur in a
(e.g., single vessel vs. widespread vasculitis)? dose-responsive manner, the animal findings should be consid-
 What is the overall weight of evidence with the existing ered more in terms of hazard identification, and it is likely not
data? appropriate to set clinical dose ranges based on immunogenicity-
 What additional data are more likely to be informative? related findings. In both the study and clinical regulatory
documents describing immunogenicity-related findings, it is
For example, in a study where a single animal presents with important to distinguish between immunogenicity-related find-
a minimal to slight vascular lesion and the systemic exposure ing, which have questionable translation for human, and direct
was markedly reduced, it may be warranted to indicate in the on-target pharmacologic and toxicologic effects, which are more
study report that the finding was potentially due to ICD rather likely translatable to the human setting. In the author’s preferred
than representing a direct test-article effect and not to pursue approach, these direct on-target effects are most appropriate for
further evaluation. In contrast, a study with a high incidence of setting the clinical dose range. Ultimately, the goal is to provide
postdose reactions and vascular disease that resulted in clinicians and regulators the transparency and perspective on the
Vahle 1019

immunogenicity-related findings while providing clear descrip- References


tions on the nature and dose response of direct on-target effects. Basser, R. L., O’Flaherty, E., Green, M., Edmonds, M., Nichol, J., Menchaca,
The field currently lacks robust data to either definitively D. M., Cohen, B., and Begley, C. G. (2002). Development of pancytopenia
confirm or refute the statement that ICD in animals does not with neutralizing antibodies to thrombopoietin after multicycle chemother-
apy supported by megakaryocyte growth and development factor. Blood
predict for similar effects in man. Certainly, there are examples
99, 2599–602.
in summary basis of approval documents where the findings in De Groot, A. S., and Scott, D. W. (2007). Immunogenicity of protein thera-
nonhuman primates did not predict for an increased immuno- peutics. Trends Immunol 28, 482–90.
genicity or immune complex in humans. In an important paper Heyen, J. R., Rojko, J., Evans, M., Brown, T. P., Bobrowski, W. F., Vitsky, A.,
discussing both animal and human data, Hussar et al. (2017) Dalton, S., et al. (2014). Characterization, biomarkers, and reversibility of a
monoclonal antibody-induced immune complex disease in cynomolgus
describe a high incidence and severity of ICD in monkeys
monkeys (Macaca fascicularis). Toxicol Pathol 42, 765–73.
administered an anti-CD20 monoclonal antibody; however, the Husar, E., Solonets, M., Kuhlmann, O., Schick, E., Piper-Lepoutre, H., Singer,
accumulated clinical data have not identified an increased risk T., and Tyagi, G. (2017). Hypersensitivity reactions to obinutuzumab in
of events attributable to ICD. cynomolgus monkeys and relevance to humans. Toxicol Pathol 45, 676–86.
The author closed the presentation by emphasizing to the Krishna, M., and Nadler, S. G. (2016). Immunogenicity to biotherapeutics—
The role of anti-drug immune complexes. Front Immunol 7, 1–13.
audience that immunogenicity in nonclinical safety study is
Kronenberg, S., Husar, E., Schubert, C., Freichel, C., Emrich, T., Lechmann,
challenging to predict, and the sequelae associated with M., Giusti, A. M., and Regenass, F. (2017). Comparative assessment of
immunogenicity are diverse. Study teams mostly keep an immune complex-mediated hypersensitivity reactions with biotherapeutics
open mind and use a weight-of-evidence approach when in the non-human primate: Critical parameters, safety and lessons for future
describing and interpreting findings that may be attributable studies. Regul Toxicol Pharmacol 88, 125–37.
Kuus-Reichel, K., Grauer, L. S., Karavodin, L. M., Knott, C., Krusemeier, M.,
to immunogenicity. and Kay, N. E. (1994). Will immunogenicity limit the use, efficacy, and
future development of therapeutic monoclonal antibodies? Clin Diagn Lab
Acknowledgments Immunol 1, 365–72.
The author greatly benefited from collaborative discussions with Leach, M. W., Rottman, J. B., Hock, M. B., Finco, D., Rojko, J. L., and Beyer,
pathology, toxicology and immunology colleagues over the years J. C. (2014). Immunogenicity/hypersensitivity of biologics. Toxicol Pathol
including Nancy Everds, William Siska, Charley Dean, Sven Kronen- 42, 293–300.
berg, Frank Brenan, Michael Leach, Jennifer Rojko, Rafael Ponce, Li, J., Yang, C., Xia, Y., Bertino, A., Glaspy, J., Roberts, M., and Kuter, D. J.
(2001). Thrombocytopenia caused by the development of antibodies to
Mark Evans, Heather Dale, Tara Arndt, Jamie Blackbourne, and Mer-
thrombopoietin. Blood 98, 3241–48.
edith Steeves. These discussions greatly aided the preparation of the
Mease, K. M., Kimzey, A. L., and Lansita, J. A. (2017). Biomarkers for non-
continuing education presentation upon which this article was based. clinical infusion reactions in marketed biotherapeutics and considerations
for study design. Curr Opin Toxicol 4, 1–15.
Author Contributions Pierog, P., Krishna, M., Yamniuk, A., Chauhan, A., and DeSilva, B. (2015).
The author (JV) contributed to conception or design; data acquisition, Detection of drug specific circulating immune complexes from in vivo
analysis, or interpretation; drafting the manuscript; and critically cynomolgus monkey serum samples. J Immunol Methods 416, 124–36.
revising the manuscript. The author gave final approval and agreed Ponce, R., Abad, L., Amaravadi, L., Gelzleichter, T., Gore, E., Green, J.,
Gupta, S., et al. (2009). Immunogenicity of biologically-derived therapeu-
to be accountable for all aspects of work in ensuring that questions
tics: Assessment and interpretation of nonclinical safety studies. Regul
relating to the accuracy or integrity of any part of the work are appro- Toxicol Pharmacol 54, 164–82.
priately investigated and resolved. Rojas, J. R., Taylor, R. P., Cunningham, M. R., Rutkoski, T. J., Vennarini, J.,
Jang, H., Graham, M. A., et al. (2005). Formation, distribution, and elim-
Declaration of Conflicting Interests ination of infliximab and anti-infliximab immune complexes in cynomol-
The author(s) declared no potential, real, or perceived conflicts of gus monkeys. J Pharmacol Exp Ther 313, 578–85.
interest with respect to the research, authorship, and/or publication Rojko, J. L., Evans, M. G., Price, S. A., Han, B., Waine, G., DeWitte, M.,
of this article. Haynes, J., et al. (2014). Formation, clearance, deposition, pathogenicity,
and identification of biopharmaceutical-related immune complexes:
Review and case studies. Toxicol Pathol 42, 725–64.
Funding
Yin, L., Chen, X., Vicini, P., Rup, B., and Hickling, T. P. (2015). Therapeutic
The author(s) received no financial support for the research, author- outcomes, assessments, risk factors and mitigation efforts of immunogeni-
ship, and/or publication of this article. city of therapeutic protein products. Cell Immunol 295, 118–26.

You might also like