Unlock the Potential of Autoantibody Biomarkers
eBook
Published: December 4, 2024
Credit: Sengenics
Genetic biomarkers are fixed from birth and offer a static view of what might happen. However, they miss real-time changes in the body as it responds to disease, lifestyle, environmental factors or treatments.
In contrast, autoantibodies provide real-time insights into the body’s current biological state, capturing the immune system’s response to active disease processes.
This eBook explores the transformative role of autoantibodies in precision medicine, especially for complex diseases.
Download this eBook to discover:
- Advances in early detection and personalized therapies for cancer
- Insights into immune regulation with anti-cytokine autoantibodies
- Best practices for autoantibody profiling with protein microarrays
eBook
Beyond Genetics:
Autoantibodies as BiomarkersBeyond Genetics:
Autoantibodies as Biomarkers
Table of Contents
21
Citrullination in Disease:
The Role of Autoantibodies
Examine how profiling autoantibodies
against citrullinated proteins offers new diagnostic and therapeutic insights, especially
in conditions involving chronic inflammation.
05
Autoantibodies: Transforming
Precision Medicine
Get a comprehensive overview of what autoantibodies are and why they hold so much
promise as biomarkers.
14
Anti-Cytokine Autoantibodies
Discover how anti-cytokine autoantibodies regulate immune responses in health
and disease—and how these insights are
advancing precision medicine.
03
Introduction
Understand how autoantibodies address the
limitations of genetic biomarkers, enabling
more personalized and responsive care.
26
Profiling Autoantibodies
with Precision
Learn how protein microarrays facilitate
detailed autoantibody profiling and why
choosing the right microarray is crucial for
generating reliable, high-quality data.
09
Autoantibodies in Cancer
Explore how autoantibody profiling advances early detection, prognosis, and personalized cancer therapies.
Page 2Introduction
Genetic Biomarkers:
Limited by Static Information
Genetic biomarkers, such as DNA mutations, polymorphisms, and other inherited genetic variations,
have long been used to assess disease risk, susceptibility, and inherited traits. While these biomarkers
provide valuable insights into a person’s genetic predisposition to certain diseases, they offer only a static
snapshot of an individual’s biology. Genetic markers
reveal what might happen based on a person’s genetic code but don’t capture the real-time changes occurring in the body as it responds to disease, lifestyle
factors, environmental influences, or treatments.
This limitation means that genetic biomarkers alone
cannot account for the dynamic nature of diseases—
especially complex, heterogeneous conditions like
cancer, autoimmune disorders, and neurodegenerative diseases, where disease progression is influenced
by a constantly shifting biological environment.
Because genetic biomarkers are fixed from birth and
remain constant throughout life, they offer limited
value in monitoring disease progression or treatment
response in real time.
Page 3Autoantibodies: Dynamic, Real-Time
and Historical Indicators of Disease
Autoantibodies, one of the most underexplored
classes of biomarkers, are produced by the immune
system in response to disease-driven changes, targeting the body’s own proteins. Unlike genetic biomarkers, they provide real-time insights into the body’s
current biological state, capturing the immune system’s response to active disease processes.
This eBook explores the expanding role of autoantibodies in precision medicine, particularly as biomarkers for complex diseases like cancer, autoimmune
disorders, neurodegenerative conditions, and chronic
inflammation. Through in-depth explanations and
real-world case studies, each chapter reveals how autoantibody profiling enables earlier diagnosis, more
personalized treatment, and the discovery of new
therapeutic targets.
By harnessing autoantibodies as dynamic biomarkers,
we can move beyond genetic predisposition and gain
a real-time view of disease progression, opening up
new possibilities for targeted, individualized care.
In the following chapters, we invite you to delve into
the science, applications, and future potential of
autoantibodies as biomarkers in precision medicine.
Join us on this journey into the immune system’s hidden messages and discover how they hold the key to
unlocking a healthier, more personalized future for all.
Table of ContentsAutoantibodies:
Transforming Precision Medicine
Page 5
Introduction
AAbs (AAbs) are antibodies produced by the immune
system that mistakenly target the body’s own proteins, often as a response to cellular changes caused
by disease. Traditionally, AAbs were primarily associated with autoimmune diseases, where the immune
system attacks healthy tissues, as seen in conditions
like rheumatoid arthritis and lupus. In these contexts,
AAbs served as key diagnostic markers, helping clinicians identify and monitor autoimmune disorders.
However, recent research has shown that AAbs are
not limited to autoimmune diseases; they are also
found in various other conditions, including cancer,
neurodegenerative disorders, and chronic inflammatory diseases. Their presence across such a wide
range of diseases has revealed their potential as valuable biomarkers for early detection, patient stratification, and monitoring treatment response.
AAbs Reflect Pathological States
Most antibodies are elicited through a limited set of
mechanisms, all of which are associated with abnormal or disease states rather than health. These mechanisms that cause self-proteins to become autoantigenic include:
• Altered protein levels: Significant and rapid
changes in protein concentration
• Protein sequence variation: Genetic mutations or
abnormal mRNA processing
• Ectopic expression: Protein appearing in the
wrong cellular compartment or at an inappropriate time
• Aberrant post-translational modifications: Changes caused by abnormal enzyme activation or
oxidative damage
• Molecular mimicry: An AAb initially produced in
response to a foreign antigen that also recognizes
a self-protein
AAb Biomarkers in Disease
Antibody generation is linked to the formation of TDP-43
protein aggregates that occurs in amylotrophic lateral
sclerosis (ALS) (1).
Tumor-associated proteins in cancer elicit an autoimmune
response (2,3).
Antibody biomarkers of lupus in the blood samples of
military personnel were detected up to nine years before
their diagnosis (4).
Antibodies have shown significant potential in predicting
disease outcomes (5-7).
Researchers identified and validated a signature of 13
antibodies predictive of a lower five-year survival in nonsmall cell lung cancer (8).AUTOANTIBODIES: TRANSFORMING PRECISION MEDICINE
Predictive
Level correlates with
process intensity
Manifest Early
Produced early in protective and pathogenic processes
Accessible
Multiple peripheral
sample types
Specific
Highly specific and
stable antibody-antigen binding
System-wide
Circulate system-wide,
enabling minimal
sample collection
Abundant
High concentration
in minimal sample
volume
Reflect Disease
Produced by immune
system only to molecular changes
Stable
Easily transported,
stored, and handled
• Abnormal protein folding or aggregation: Misfolding or aggregation during cellular stress or
toxicity that overwhelms normal protein degradation processes
• Aberrant proteolysis: Abnormal activity of proteolytic enzymes in inflammatory microenvironments
Various mechanisms can lead to the formation or exposure of new antibody binding sites, known as neoepitopes, that are typically not accessible for binding
under normal in vivo conditions (Figure 1).
Examples of AAbs associated with various diseases
can be found in the light blue box on the previous
page.
AAbs Have Ideal
Biomarker Characteristics
Antibodies—including AAbs—are direct indicators of
disease, often appearing before symptoms and remaining detectable throughout the course of illness.
This makes antibody profiling a powerful tool for
identifying disease-associated proteins. In contrast,
protein profiling alone cannot distinguish between
proteins directly linked to disease or those indirectly
affected.
Key characteristics of antibodies are listed in the gray
hexagons to the right.
Figure 1. Neoepitopes formed during pathological processes can elicit the production of AAbs
Page 6Concluding Remarks
AAb biomarkers hold the potential to
transform patient care. They can reveal
disease-associated proteins and protein
pathways, providing valuable insights for
unraveling the complexities of diseases and
aiding in the development of new medications. They can help facilitate early detection, subtype diseases more accurately,
predict who will (or won’t) respond to treatments, predict the likelihood of immune-related adverse events, and improve patient
stratification for clinical trials.
By analyzing the vast array of AAbs through
a process called immunoprofiling, scientists
can gain a clear understanding of a patient’s
disease state, offering guidance in the complex landscape of chronic illnesses.
AUTOANTIBODIES:
TRANSFORMING PRECISION MEDICINE
Jaeyun Sung, PhD
Senior Associate Consultant
and Assistant Professor,
Mayo Clinic
ON-DEMAND WEBINAR
Serum Autoantibodies Differentiate Rheumatoid
Arthritis Subgroups
Patients with rheumatoid arthritis (RA) can be categorized as either anti-citrullinated protein antibody-positive (ACPA+) or negative (ACPA-). In
this webinar, Dr. Sung presents his research exploring a broad range of
serological autoantibodies to uncover immunological differences between
these RA subgroups using data from ACPA+RA patients, ACPA- RA patients, and matched healthy controls.
See also “Citrulination in Disease: The Role of Autoantibodies.”
Watch Now
Page 7
In many disorders and pathologies
affecting tissue structure, neoepitopes trigger the production of
AAbs, contributing to disease pathology directly or systemically.
Profiling these AAbs is vital for
detecting and monitoring disease
activity and treatment outcomes.
Allan Stensballe, PhD
Associate Professor,
Aalborg UniversityAUTOANTIBODIES: TRANSFORMING PRECISION MEDICINE
2003 Oct 16;349(16):1526-33. doi: 10.1056/NEJMoa021933. PMID:
14561795.
5. Bizzaro N. Autoantibodies as predictors of disease: the clinical and
experimental evidence. Autoimmun Rev. 2007 Jun;6(6):325-33. doi:
10.1016/j.autrev.2007.01.006. Epub 2007 Jan 30. PMID: 17537376.
6. Kathrikolly T, Nair SN, Mathew A, Saxena PPU, Nair S. Can serum
autoantibodies be a potential early detection biomarker for breast
cancer in women? A diagnostic test accuracy review and meta-analysis. Syst Rev. 2022 Oct 9;11(1):215. doi: 10.1186/s13643-022-
02088-y. PMID: 36210467; PMCID: PMC9549667.
7. Zaenker P, Ziman MR. Serologic autoantibodies as diagnostic cancer
biomarkers--a review. Cancer Epidemiol Biomarkers Prev. 2013
Dec;22(12):2161-81. doi: 10.1158/1055-9965.EPI-13-0621. Epub
2013 Sep 20. PMID: 24057574.
8. Patel AJ, Tan TM, Richter AG, Naidu B, Blackburn JM, Middleton
GW. A highly predictive autoantibody-based biomarker panel for
prognosis in early-stage NSCLC with potential therapeutic implications. Br J Cancer. 2022 Feb;126(2):238-246. doi: 10.1038/
s41416-021-01572-x. Epub 2021 Nov 2. PMID: 34728792; PMCID:
PMC8770460.
References
1. Conti E, Sala G, Diamanti S, Casati M, Lunetta C, Gerardi F, Tarlarini
C, Mosca L, Riva N, Falzone Y, Filippi M, Appollonio I, Ferrarese C,
Tremolizzo L. Serum naturally occurring anti-TDP-43 auto-antibodies
are increased in amyotrophic lateral sclerosis. Sci Rep. 2021 Jan
21;11(1):1978. doi: 10.1038/s41598-021-81599-5. PMID: 33479441;
PMCID: PMC7820419.
2. Aziz F, Smith M, M Blackburn J. Autoantibody-Based Diagnostic
Biomarkers: Technological Approaches to Discovery and Validation
[Internet]. Autoantibodies and Cytokines. IntechOpen; 2019. Available from: http://dx.doi.org/10.5772/intechopen.75200
3. Sexauer D, Gray E, Zaenker P. Tumour- associated autoantibodies
as prognostic cancer biomarkers- a review. Autoimmun Rev. 2022
Apr;21(4):103041. doi: 10.1016/j.autrev.2022.103041. Epub 2022
Jan 12. PMID: 35032685.
4. Arbuckle MR, McClain MT, Rubertone MV, Scofield RH, Dennis
GJ, James JA, Harley JB. Development of autoantibodies before
the clinical onset of systemic lupus erythematosus. N Engl J Med.
Table of ContentsPage 9
Introduction
Biomarkers are essential for diagnosing, monitoring,
and treating diseases. However, developing biomarker signatures with high sensitivity and specificity is
particularly challenging for heterogeneous diseases
like cancer.
Recent advancements in immunology suggest that
autoantibodies (AAbs), or antibodies that target
self-molecules, can serve as precise and reliable
biomarkers in cancer (1). This chapter explores how
cancer can induce AAb production and how AAbs
provide valuable insights into disease mechanisms
and therapeutic targets, aiding in early diagnosis,
predicting treatment response, and guiding drug
development.
AAbs: A Paradigm Shift in Cancer
Biomarker Discovery
Cancer processes can induce AAb production by
forming or exposing new binding sites on proteins,
known as neoepitopes, through cellular changes or
therapeutic pressures (Figure 1). Changes in AAb
profiles can reflect both the malignant transformation continuum and subsequent disease progression,
offering detailed insights into the disease’s location,
Autoantibodies in Cancer
Figure 2. Timeline of AAb production before and after cancer diagnosisAUTOANTIBODIES IN CANCER
nature, and timing. Importantly, AAbs may have a
pathogenic or protective role in cancer progression.
Early Detection and Predictive Power of
AAbs in Cancer
AAbs generated during carcinogenesis provide
a valuable opportunity for earlier diagnosis,
enabling intervention before symptoms appear
(Table 1, Figure 2) (2). For example, AAbs targeting p53, the most commonly mutated protein in
cancer, were detected on average 3.5 years prior
to diagnosis, with a positive predictive value of
0.76 for subsequent malignancy (3). AAbs in lung
cancer patients are present up to 5 years prior to
diagnosis (4). In fact, an AAb-based assay, EarlyCDT®-Lung test, has been approved for clinical use
as a complementary diagnostic method (5). AAb
profiling also helped guide the protein signature
that is now utilized in Videssa® Breast, a CLIA-certified blood-based assay to help diagnose early-stage breast cancer following an abnormal
mammogram (6).
Earlier cancer detection provides a window of
opportunity during which interventions are more
able to effectively modify disease to improve survival rates. For public health systems, early diagnosis can reduce the long-term burden of cancer
treatment, both in terms of healthcare costs and
patient quality of life. Additionally, early-stage
treatments are generally less resource-intensive,
leading to better allocation of healthcare resources and increased accessibility to care.
AAb profiling also provides valuable information
beyond their use as early diagnostic biomarkers. For instance, a study discovered a signature
of 13 AAbs predictive of poor survival rates in
patients with resected non-small cell lung cancer
(7). This signature was validated in an independent cohort, achieving a sensitivity of 84% and
specificity of 74%. Another study identified AAb
signatures predictive of outcomes of melanoma
Page 10
To my surprise, AAbs can also predict
response to treatment. It was a very
clear and very clean result.
Iman Osman, MD
Associate Dean for Clinical
Research Strategy,
Rudolf L. Baer Professor of
Dermatology,
Professor of Depts of
Medicine (Oncology) and
Urology,
Director of the NYU
Melanoma SPORE,
Director of the Interdisciplinary Melanoma Cooperative Group (IMCG),
New York University (NYU),
Grossman School of
Medicine
Jessica da Gama
Duarte, PhD
Senior Research Fellow,
Olivia Newton-John
Cancer Research Institute
AAbs in cancer provide critical
insights into how strongly and extensively the immune system recognizes
and responds to the disease. Additionally, they reveal specific proteins
that the immune system targets,
offering personalized insights that
could lead to new treatment options.AUTOANTIBODIES IN CANCER
patients treated with immune checkpoint inhibitors
(8). Interestingly, different AAb profiles were observed
for toxicity (i.e., immune-related adverse events) and
response between non-Hispanic whites and underrepresented minorities.
Impact of Cancer Therapies on AAb Levels
Common cancer treatments like chemotherapy and
radiotherapy induce massive cell death and release
tumor-associated proteins, which can trigger AAb
production. AAb levels also frequently increase in
response to next-generation cancer therapies that
stimulate the immune system, such as immune checkpoint inhibitors.
Role of AAbs in Cancer Vaccine and
Drug Development
AAbs play a significant role in rational cancer vaccine
and drug development (Table 1). They reveal which
cancer-associated autoantigens are targeted in vivo
by the patient’s immune system. In other words, AAb
profiling can pinpoint which autoantigens are immunodominant, elicit B-cell memory, contribute to paraneoplastic syndromes, or stimulate the production of
protective antibodies that slow disease progression.
Such autoantigens could be explored as targets for
vaccines, chimeric antigen receptor T-cell (CAR-T)
therapy, and antibody-drug conjugates. Moreover,
Application Description
Understand disease mechanisms Discover AAb targets that are often associated with disease
Identify biomarkers Diagnose, stratify patients, and subtype disease. Predict patient prognosis and treatment outcomes.
Guide vaccine and drug development Identify potential therapeutic targets. Map epitope spreading. Determine
B-cell specificities.
Table 1. Examples of AAb profiling applications in cancer
Page 11
PROTEIN MICROARRAY
Profile Autoantibodies to
Cancer Antigens
Transform your cancer research with
i-Ome® Cancer, the largest cancer-focused protein microarray in the world.
• Profile AAbs against 500+ meticulously selected protein antigens,
each chosen for their relevance in
cancer
• Discover biologically relevant biomarkers with correctly folded proteins, guaranteed by proprietary
KREX® technology
• Analyze two isotypes (IgG, IgA, or
IgM) in a single, powerful assay
Contact UsAkshay Patel, MD
Specialist Registrar in Thoracic Surgery; Clinical Lecturer, Institute of Immunology and Immunotherapy,
University of Birmingham
ON-DEMAND WEBINAR
B-Cell Repertoire in Determining
Responses to Checkpoint Blockade
in NSCLC
Immunotherapy advancements, particularly immune
checkpoint inhibitors (ICIs) like anti-PD-1 and anti-CTLA-4, are transforming treatment for advanced nonsmall cell lung cancer (NSCLC). This webinar explores
how B-cell biology impacts patient responses and
immune-related adverse events (IRAEs) and highlights
how Sengenics protein microarrays have identified
novel autoantibodies that could predict responses to
ICIs, paving the way for more personalized approaches to NSCLC treatment.
Watch Now
Professor Gary Middleton
Professor of Medical Oncology,
University of Birmingham
AAb profiling aids in identifying B-cell specificities for
chimeric autoantibody receptor T-cell (CAAR-T) therapy, useful in managing cancer-related immune-related
adverse events (irAEs) or paraneoplastic disease.
AAb profiling can map the immune-targeted diversification elicited by vaccines and drugs. For instance,
the HER-2/neu peptide vaccine elicits the generation
of AAbs that target endogenous HER-2/neu. Through
a process called epitope spreading, a patient’s immunoreactivity can spread to the p53 protein. Epitope
spreading is also relevant in certain therapies, enhancing efficacy by stimulating the immune system to
target more than just the original protein target.
Conclusion
Genetic testing alone does not accurately reflect the
dynamic, real-time biological changes and molecular
heterogeneity that occur during cancer. AAb profiling
bridges this gap by providing a direct view of the immune system’s ongoing response to each individual’s
evolving molecular landscape. Highly complementary
to other omics datasets, AAb profiling is a key component in the precision medicine toolkit, transforming
cancer profiles into actionable clinical insights and
ultimately improving patient outcomes.
AUTOANTIBODIES IN CANCER
Page 12AUTOANTIBODIES IN CANCER
References
1. Wu, J., Li, X., Song, W., Fang, Y., Yu, L., Liu, S., Churilov, L.
P., & Zhang, F. (2017). The roles and applications of autoantibodies in progression, diagnosis, treatment and prognosis
of human malignant tumours. Autoimmunity reviews, 16(12),
1270–1281. https://doi.org/10.1016/j.autrev.2017.10.012
2. de Jonge, H., Iamele, L., Maggi, M., Pessino, G., & Scotti, C.
(2021). Anti-Cancer Auto-Antibodies: Roles, Applications and
Open Issues. Cancers, 13(4), 813. https://doi.org/10.3390/
cancers13040813
3. Li, Y., Karjalainen, A., Koskinen, H., Hemminki, K., Vainio, H.,
Shnaidman, M., Ying, Z., Pukkala, E., & Brandt-Rauf, P. W.
(2005). p53 autoantibodies predict subsequent development
of cancer. International journal of cancer, 114(1), 157–160.
https://doi.org/10.1002/ijc.20715
4. Zhong, L., Coe, S. P., Stromberg, A. J., Khattar, N. H., Jett,
J. R., & Hirschowitz, E. A. (2006). Profiling tumor-associated
antibodies for early detection of non-small cell lung cancer.
Journal of thoracic oncology : official publication of the
International Association for the Study of Lung Cancer, 1(6),
513–519.
5. Chapman, C. J., Healey, G. F., Murray, A., Boyle, P., Robertson, C., Peek, L. J., Allen, J., Thorpe, A. J., Hamilton-Fairley,
G., Parsy-Kowalska, C. B., MacDonald, I. K., Jewell, W.,
Maddison, P., & Robertson, J. F. (2012). EarlyCDT®-Lung test:
improved clinical utility through additional autoantibody
assays. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 33(5),
1319–1326. https://doi.org/10.1007/s13277-012-0379-2
6. Anderson, K. S., Sibani, S., Wallstrom, G., Qiu, J., Mendoza,
E. A., Raphael, J., Hainsworth, E., Montor, W. R., Wong, J.,
Park, J. G., Lokko, N., Logvinenko, T., Ramachandran, N.,
Godwin, A. K., Marks, J., Engstrom, P., & Labaer, J. (2011).
Protein microarray signature of autoantibody biomarkers for
the early detection of breast cancer. Journal of proteome
research, 10(1), 85–96. https://doi.org/10.1021/pr100686b
7. Patel AJ, Tan TM, Richter AG, Naidu B, Blackburn JM, Middleton GW. A highly predictive autoantibody-based biomarker panel for prognosis in early-stage NSCLC with potential
therapeutic implications. Br J Cancer. 2022 Feb;126(2):238-
246. doi: 10.1038/s41416-021-01572-x. Epub 2021 Nov 2.
PMID: 34728792; PMCID: PMC8770460.
8. Ibrahim, M., Angulo, P., Fa’ak, F., Abdel-Wahab, N., Diab, A.,
Mehnert, J.M., Weber, J.S., Lund, A.W., Schober, M., Zhong,
J., & Osman, I. (2023). Determinants of racial disparities in
immune-related adverse events (irAE) with checkpoint inhibition (ICI) in melanoma. Journal of Clinical Oncology.
9. Wang, B., Fields, L., & Li, L. (2023). Recent advances in
characterization of citrullination and its implication in human
disease research: From method development to network
integration. Proteomics, 23(21-22), e2200286. https://doi.
org/10.1002/pmic.202200286
10. Arseniy E. Yuzhalin; Citrullination in Cancer. Cancer Res 1
April 2019; 79 (7): 1274–1284. https://doi.org/10.1158/0008-
5472.CAN-18-2797
Table of ContentsAnti-Cytokine Autoantibodies
Page 14
Introduction
Anti-cytokine autoantibodies (ACAAs) are naturally
occurring or elicited antibodies that target cytokines—key proteins that mediate and regulate immune responses (Table 2). While cytokines generally
direct and modulate immune activity, ACAAs often inhibit or potentiate these effects, influencing immune
regulation in both health and disease. Although
less common, ACAAs can also directly drive disease
processes. This white paper delves into the role of
ACAAs across various disease contexts and explores
their potential use in predicting treatment outcomes,
classifying disease severity, guiding vaccine development, and other therapeutic applications.
ACAAs in Health
Low levels of ACAAs are relatively common in the
general population. A Danish study of nearly 9,000
healthy blood donors found that 86% of participants
had at least one detectable ACAA, although the
prevalence varied significantly depending on the
cytokine target (2). Anti-IL-6 ACAAs were the most
frequent, present in 65% of participants, while antiGM-CSF ACAAs were found in just 10%. Interestingly,
the study also found that the cumulative presence
of multiple ACAAs correlated with several indicators
of immune function, including self-reported health
scores and the frequency of antibiotic prescriptions,
supporting the belief that ACAAs influence overall
immune health and resilience (3).
ACAAs could function as natural regulators, balancing the immune response and reducing the risk
of excessive inflammation without compromising
the body’s ability to fight infections. By neutralizing
pro-inflammatory cytokines, they may help prevent
harmful immune over-activation like cytokine storms.
These storms trigger excessive cytokine production, leading to severe inflammation across multiple
organs and systems, which can potentially result in
multi-organ failure and death.
ACAAs in Disease
ACAAs were initially discovered in patients with
thymoma-associated autoimmune diseases but are
now known to be present in healthy individuals and
a variety of other conditions (4). These include autoimmune diseases like rheumatoid arthritis, systemic
lupus erythematosus, and psoriasis; immunodeficiencies; and infectious diseases. In these contexts,
ACAAs may play a causative or associative role,
potentially contributing to disease by inhibiting cytokines crucial for immune defense. They can also be
linked to specific subtypes within disease categories.
For instance, ACAAs can increase susceptibility to
infections (5). Anti-IFNγ antibodies are strongly associated with disseminated non-tuberculous mycobacterial (NTM) infections. Anti-IL-6 ACAAs have been
linked to severe staphylococcal and streptococcal
infections while anti-IL-17/IL-22 antibodies are tied to
chronic mucosal candidiasis. Anti-GM-CSF antibodies
have been implicated in opportunistic infections suchCytokine Drug Type Brand Name Disease Area
G-CSF Protein Neupogen, Neulasta neutropenia
GM-CSF Protein Leukine neutropenia
IFN-α Protein Intron A, Roferon-A
chronic hepatitis B, chronic hepatitis C, hairy cell leukemia, Kaposi’s
sarcoma, malignant melanoma, follicular lymphoma, chronic myelogenous leukemia (CML)
IFN-β Protein Avonex, Betaseron,
Rebif, Plegridy multiple sclerosis (MS)
IFN-γ Protein Actimmune chronic granulomatous disease (CGD), severe malignant osteopetrosis
IL-1R Small molecule Kineret rheumatoid arthritis, neonatal-onset multisystem inflammatory disease
(NOMID)
IL-2 Protein Proleukin metastatic renal cell carcinoma, metastatic melanoma
IL-5 Antibody Nucala, Cinqair, Fasenra severe eosinophilic asthma, eosinophilic granulomatosis with polyangiitis (EGPA)
IL-6R Antibody Actemra, Kevzara rheumatoid arthritis, juvenile idiopathic arthritis, giant cell arteritis,
cytokine release syndrome
IL-11 Protein Neumega severe thrombocytopenia
IL-12/23 1 Antibody Stelara, Tremfya, Om voh - psoriasis, psoriatic arthritis, Crohn’s disease, ulcerative colitis
IL-13 Antibody Adbry atopic dermatitis
IL-17A Antibody Cosentyx, Taltz, Siliq psoriasis, psoriatic arthritis, ankylosing spondylitis, axial spondyloarthritis (axSpA), hidradenitis suppurativa (HS)
IL-17A/IL-17F 2 Antibody Bimzelx psoriatic arthritis, axSpA, ankylosing spondylitis
IL-23 Antibody Tremfya, Ilumya, Skyrizi psoriasis, psoriatic arthritis, Crohn’s disease
TNF Antibody Remicade, Humira,
Cimzia, Simponi
rheumatoid arthritis, Crohn’s disease, ulcerative colitis, ankylosing
spondylitis, psoriasis, psoriatic arthritis
TNFR Receptor fusion
protein Enbrel rheumatoid arthritis, psoriasis, ankylosing spondylitis, juvenile id pathic arthritis ioTable 2. Cytokine-based therapies approved by the U.S. FDA
Cytokine-Based Therapies
The crucial role of cytokines in disease is evident through the numerous cytokine-based therapies approved
by the U.S. Food and Drug Administration (FDA). To date, at least 39 therapies that mimic or target cytokines
or their receptors have been approved for clinical use (Table 2) (1).
1 Drugs target a shared subunit between IL-12 and IL-23.
2 Drug targets IL-17A and IL-17F separately or as a heterodimer.
ANTI-CYTOKINE AUTOANTIBODIES
as those caused by Nocardia spp. and Cryptococcus
spp., as they impair macrophage function, which
is essential for combating these pathogens. In at
least one patient, anti-GM-CSF autoantibodies were
detected 10 years prior to developing Nocardia spp.
infection (6).ANTI-CYTOKINE AUTOANTIBODIES
GM-CSF also plays a key role in the pathophysiology of pulmonary alveolar proteinosis (PAP), which
is a lung condition characterized by the buildup of
surfactant in the alveoli. The titer of circulating antiGM-CSF autoantibodies may help predict how well a
patient will response to PAP treatment with subcutaneous recombinant human GM-CSF (7).
During the COVID-19 pandemic, the discovery of
anti-IFN-α and IFN-ω ACAAs in some patients offered
critical insights into why certain individuals experienced more severe outcomes (8). These antibodies
impair the body’s antiviral defense by neutralizing key
interferons essential for early viral response, thereby
increasing susceptibility to severe COVID-19. Interestingly, the frequency of these ACAAs was associated with increased age and male sex in patients with
critical COVID-19 (p = 3x10-6 and p = 0.003, respectively) (9).
Another study found that recovered COVID-19
patients with higher autoantibody titers targeting a
subset of cytokines known as chemokines—specifically CCL21, CXCL13, and CXCL16—were less likely
to experience long COVID-19 symptoms one year
after infection (10). This suggests that certain ACAAs
may be linked to the progression and outcome of the
disease.
In systemic lupus erythematosus (SLE), disease severity is linked to elevated levels of IFN-α and ACAAs
targeting BAFF, a cytokine involved in B-cell activation (5). On the other hand, ACAAs that neutralize
IFN-α and TNF have been associated with reduced
lupus severity, suggesting that ACAAs may have
therapeutic potential in diseases driven by cytokine
activity.
Beyond these established associations, emerging
research suggests that ACAAs may be more widespread, with implications for conditions like cancer,
cardiovascular disease, and neurological disorders.
It is also worth mentioning that ACAAs may also arise
as unintended consequences of severe infections or
tissue damage, contributing to immune dysregulation. As such, they could serve as early indicators of
immune imbalance and dysfunction.
Expanding Role of Anti-Cytokine
Antibodies (ACAs) in Disease
Treatment and Therapeutics
The therapeutic potential of ACAs is being explored
in various disease contexts (Table 2). In oncology,
ACAs have garnered attention for their possible roles
in treating cancer-related cachexia (CRC) and improving the efficacy of monoclonal antibody (mAb)
therapies. Among the most promising approaches
for treatment of CRC-associated cachexia is a combination of anti-IL-1A antibodies and thalidomide,
reducing the inflammatory burden associated with the
condition (11).
Additionally, researchers are investigating a new class
of therapies called immunocytokines (12-14). Immunocytokines are antibody-cytokine fusion proteins or
therapeutic ACAs (such as those listed in Table 2) that
are coupled to antibodies targeting cancer-specific or
cancer-associated tumor antigens. These are designed to address the challenges posed by the tumor
micro-environment in solid tumors, which often suppresses immune activity and diminishes mAb efficacy,
yet remains sensitive to pro-inflammatory cytokines.
Traditional cytokine therapies have been limited by
off-target toxicity and the short half-life of cytokines
when administered alone. Immunocytokines offer a
solution by delivering cytokines directly to the tumor
site, extending their half-life, and reducing systemic
toxicity. These fusion proteins can also bridge local
cytotoxic immune cells, such as macrophages and
natural killer (NK) cells, with tumor cells, enhancing
the immune system’s ability to target and destroy cancer cells.
Several immunocytokines are currently in clinical trials
(Table 3), demonstrating promise for future cancer
treatment strategies by optimizing immune activation
Page 16ANTI-CYTOKINE AUTOANTIBODIES
at the tumor site without the widespread side effects
of systemic cytokine therapy (13,14).
Experimental models suggest that ACAAs may play
a protective role in certain cardiovascular diseases
by modulating inflammatory responses. For instance,
anti-IL-17 antibodies have been shown to reduce inflammation in murine models of autoimmune myocarditis, a condition driven by an autoimmune response
against cardiac myosin (15). In Kawasaki Disease, a
meta-analysis of mAb studies revealed that while anti-TNF did not lower the incidence of coronary artery
aneurysms, it did reduce resistance to treatment with
intravenous immune globulin (16).
Additionally, romilkimab, a bispecific ACA that neutralizes IL-4 and IL-13, has shown promise in treating
patients with systemic sclerosis (17). Tocilizumab
(TCZ), an mAb targeting the IL-6R and blocking its
interaction with IL-6, is used to treat Takayasu arteritis, giant cell arteritis, and other inflammatory diseases, including Castleman disease, idiopathic juvenile
arthritis, and rheumatoid arthritis.
In neurological disorders, murine models have
demonstrated that ACAs can reduce the severity of
experimental autoimmune encephalitis (EAE), which
is a model for multiple sclerosis (MS)(18). Additionally, anti-cytokine mAbs targeting IL-1β and IL-6 have
Page 17
Cytokine Target Antigen Name Cancer Type Phase
IFN-α2B CD38 Modakafusp alfa
(TAK-573) multiple myeloma I, II
IL-2 GD2 Hu14.18-IL-2 neuroblastoma, melanoma, sarcoma, solid childhood
tumors I, II
IL-2 EpCAM huKS-IL2 SCLC, prostate, ovarian, breast, bladder, kidney, lung, solid tumors I, II
IL-2 CD20 DI-Leu16-IL2 B cell lymphoma I, II
IL-2 Tenascin-C F16-IL2 breast, AML, solid tumors, MCC II
IL-2 variant CEA CEA-IL2v (RG7813) solid tumors I
IL-2 variant FAP FAP-IL12v solid tumors, RCC, melanoma, pancreatic, breast,
HNC, esophageal, cervical I, II
IL-2 variant PD-1 RG6279, IBI363,
IAP0971 solid tumors I
IL-2, IL-12,
TNF EDB
L19-IL2, L19-TNF,
BC1-IL-12 (AS1409),
L19-IL-12
melanoma, RCC, NSCLC, solid tumors, pancreatic,
colorectal, DLBCL, glioblastoma, sarcoma, glioma I, II, III
IL-2LT, IL-12 Histone/DNA structures NHS-IL12 NSCLC, solid tumors, pancreatic, urogenital, bladder,
NHL, Kaposi sarcoma, melanoma I, II
IL-15 PD-L1 KD033, SIM0237,
IGM-7354 solid tumors I
IL-21 PD-1 AMG256 solid tumors I
Table 3. Clinical trials using immunocytokines
AML = acute myeloid leukemia, DLBCL = diffuse large B-cell lymphoma, HNC = head and neck cancer, MCC = Merkel cell carcinoma,
NSCLC = non-small cell lung cancer, RCC = renal cell carcinomaANTI-CYTOKINE AUTOANTIBODIES
shown promise in reducing brain inflammation and
preventing blood-brain barrier permeability in fetal
ischemia-reperfusion injury ovine models (19). These
findings suggest a potential therapeutic role for
ACAs in neuroinflammatory and neurodegenerative
conditions.
ACAs that target TNF, IL-12, and IL-23 are in clinical
use to treat inflammatory bowel disease (IBD), such
as Crohn’s disease and ulcerative colitis (1). These
mAbs bind to their respective pro-inflammatory
cytokine, inhibiting their function, which helps reduce
inflammation and alleviate pain.
Additional conditions treated with FDA-approved
mAbs targeting cytokines or cytokine receptors are
listed in Table 2.
ACAAs in Disease Prevention
Leveraging ACAAs through anti-cytokine vaccination before disease onset has demonstrated promise
in preclinical animal studies. For instance, vaccines
targeting IL-17 have been effective in reducing the
severity of collagen-induced arthritis (CIA) and EAE
(20). Similarly, an anti-IL-6 vaccine protected mice
from CIA, while an anti-IL-18 vaccine helped reduce
the severity of SLE and prevented renal damage (21).
Anti-cytokine vaccination also shows potential in other conditions, including cachexia, antibody-induced
arthritis, Leishmaniasis, atherosclerosis, and collagen
antibody-induced arthritis (CAIA).
However, clinical trials are needed to evaluate the
long-term safety, efficacy, and potential side effects
of anti-cytokine vaccination in humans. If successful, these vaccines could provide a new, targeted
approach for managing autoimmune diseases and
reducing the need for lifelong immunosuppressive
therapies.
PROTEIN MICROARRAY
Profile ACAAs and ACAs
Sengenics’s comprehensive protein
library of 2,000+ human proteins includes cytokines, chemokines, cytokine
and chemokine receptors, antimicrobial
peptides, cytotoxic effectors, and various
other immune effectors and modulators
(Table 6).
Download Protein List
Conclusion
ACAAs play a dual role, acting as natural immune
regulators while also contributing to pathology by
impairing critical cytokine functions. Numerous studies highlight their potential as biomarkers for predicting disease severity, progression, and treatment
response. Monoclonal ACAs are being explored as
therapeutic tools in diseases like cancer, cardiovascular disease, and neurological disorders, offering
targeted immune modulation. Continued research
into ACAAs and ACAs, especially through precision
antibody profiling with Sengenics protein microarrays, holds transformative potential for treating
immune-mediated diseases and paving the way for
personalized, more precise interventions.
Page 18ANTI-CYTOKINE AUTOANTIBODIES
References
1. U.S. Food and Drug Administration. (2024, October 24).
Drugs@FDA: FDA-Approved Drugs. https://www.accessdata.
fda.gov/scripts/cder/daf/index.cfm
2. von Stemann, J. H., Rigas, A. S., Thørner, L. W., Rasmussen, D. G. K., Pedersen, O. B., Rostgaard, K., Erikstrup, C.,
Ullum, H., & Hansen, M. B. (2017). Prevalence and correlation
of cytokine-specific autoantibodies with epidemiological
factors and C-reactive protein in 8,972 healthy individuals:
Results from the Danish Blood Donor Study. PloS one, 12(6),
e0179981. https://doi.org/10.1371/journal.pone.0179981
3. von Stemann, J. H., Pedersen, O. B. V., Hjalgrim, H., Erikstrup, C., Ullum, H., Dowsett, J., Thørner, L. W., Larsen, M.
A. H., Sørensen, E., Hansen, M. B., & Ostrowski, S. R. (2022).
IL-6 Autoantibodies Predict Lower Platelet Counts and Altered Plasma Cytokine Profiles in Healthy Blood Donors: Results From the Danish Blood Donor Study. Frontiers in medicine, 9, 914262. https://doi.org/10.3389/fmed.2022.914262
4. Arts, R. J. W., Janssen, N. A. F., & van de Veerdonk, F. L.
(2023). Anticytokine Autoantibodies in Infectious Diseases: A
Practical Overview. International journal of molecular sciences, 25(1), 515. https://doi.org/10.3390/ijms25010515
5. Vincent, T., Plawecki, M., Goulabchand, R., Guilpain, P., &
Eliaou, J. F. (2015). Emerging clinical phenotypes associated
with anti-cytokine autoantibodies. Autoimmunity reviews,
14(6), 528–535. https://doi.org/10.1016/j.autrev.2015.01.015
6. Rosen, L. B., Rocha Pereira, N., Figueiredo, C., Fiske, L. C.,
Ressner, R. A., Hong, J. C., Gregg, K. S., Henry, T. L., Pak,
K. J., Baumgarten, K. L., Seoane, L., Garcia-Diaz, J., Olivier,
K. N., Zelazny, A. M., Holland, S. M., & Browne, S. K. (2015).
Nocardia-induced granulocyte macrophage colony-stimulating factor is neutralized by autoantibodies in disseminated/
extrapulmonary nocardiosis. Clinical infectious diseases :
an official publication of the Infectious Diseases Society of
America, 60(7), 1017–1025. https://doi.org/10.1093/cid/
ciu968
7. Bonfield, T. L., Kavuru, M. S., & Thomassen, M. J. (2002).
Anti-GM-CSF titer predicts response to GM-CSF therapy
in pulmonary alveolar proteinosis. Clinical immunology
(Orlando, Fla.), 105(3), 342–350. https://doi.org/10.1006/
clim.2002.5301
8. Wang, X., Tang, Q., Li, H., Jiang, H., Xu, J., Bergquist, R., &
Qin, Z. (2023). Autoantibodies against type I interferons in
COVID-19 infection: A systematic review and meta-analysis.
International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases,
130, 147–152. https://doi.org/10.1016/j.ijid.2023.03.011
9. Bastard, P., Gervais, A., Le Voyer, T., Rosain, J., Philippot, Q.,
Manry, J., Michailidis, E., Hoffmann, H. H., Eto, S., Garcia-Prat, M., Bizien, L., Parra-Martínez, A., Yang, R., Haljasmä-
gi, L., Migaud, M., Särekannu, K., Maslovskaja, J., de Prost,
N., Tandjaoui-Lambiotte, Y., Luyt, C. E., … Casanova, J. L.
(2021). Autoantibodies neutralizing type I IFNs are present in
~4% of uninfected individuals over 70 years old and account
for ~20% of COVID-19 deaths. Science immunology, 6(62),
eabl4340. https://doi.org/10.1126/sciimmunol.abl4340
10. Muri, J., Cecchinato, V., Cavalli, A., Shanbhag, A. A., Matkovic, M., Biggiogero, M., Maida, P. A., Moritz, J., Toscano,
C., Ghovehoud, E., Furlan, R., Barbic, F., Voza, A., De Nadai,
G., Cervia, C., Zurbuchen, Y., Taeschler, P., Murray, L. A.,
Danelon-Sargenti, G., Moro, S., … Robbiani, D. F. (2023).
Autoantibodies against chemokines post-SARS-CoV-2 infection correlate with disease course. Nature immunology, 24(4),
604–611. https://doi.org/10.1038/s41590-023-01445-w
11. Prado, B. L., & Qian, Y. (2019). Anti-cytokines in the treatment
of cancer cachexia. Annals of palliative medicine, 8(1), 67–79.
https://doi.org/10.21037/apm.2018.07.06
12. Gout, D. Y., Groen, L. S., & van Egmond, M. (2022). The
present and future of immunocytokines for cancer treatment.
Cellular and molecular life sciences : CMLS, 79(10), 509.
https://doi.org/10.1007/s00018-022-04514-9
13. Shi, W., Liu, N., & Lu, H. (2024). Advancements and challenges in immunocytokines: A new arsenal against cancer.
Acta Pharmaceutica Sinica B. https://doi.org/10.1016/j.
apsb.2024.07.024
14. Runbeck, E., Crescioli, S., Karagiannis, S. N., & Papa, S.
(2021). Utilizing Immunocytokines for Cancer Therapy.
Antibodies (Basel, Switzerland), 10(1), 10. https://doi.
org/10.3390/antib10010010
15. Fan, Y., Weifeng, W., Yuluan, Y., Qing, K., Yu, P., & Yanlan,
H. (2011). Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of coxsackievirus b3-induced
viral myocarditis reduces myocardium inflammation. Virology
journal, 8, 17. https://doi.org/10.1186/1743-422X-8-17
16. Nomura, O., Fukuda, S., Ota, E., Ono, H., Ishiguro, A., &
Kobayashi, T. (2021). Monoclonal antibody and anti-cytokine
biologics for Kawasaki disease: A systematic review and
meta-analysis. Seminars in arthritis and rheumatism, 51(5),
1045–1056. https://doi.org/10.1016/j.semarthrit.2021.07.020
Page 19ANTI-CYTOKINE AUTOANTIBODIES
17. Shima Y. (2021). Cytokines Involved in the Pathogenesis
of SSc and Problems in the Development of Anti-Cytokine Therapy. Cells, 10(5), 1104. https://doi.org/10.3390/
cells10051104
18. Palle, P., Monaghan, K. L., Milne, S. M., & Wan, E. C. K.
(2017). Cytokine Signaling in Multiple Sclerosis and Its Therapeutic Applications. Medical sciences (Basel, Switzerland),
5(4), 23. https://doi.org/10.3390/medsci5040023
19. Patra, A., Chen, X., Sadowska, G. B., Zhang, J., Lim, Y. P.,
Padbury, J. F., Banks, W. A., & Stonestreet, B. S. (2017). Neutralizing anti-interleukin-1β antibodies reduce ischemia-related interleukin-1β transport across the blood-brain barrier
in fetal sheep. Neuroscience, 346, 113–125. https://doi.
org/10.1016/j.neuroscience.2016.12.051
20. Wraith D. C. (2006). Anti-cytokine vaccines and the immunotherapy of autoimmune diseases. European journal of
immunology, 36(11), 2844–2848. https://doi.org/10.1002/
eji.200636760
21. Delavallée, L., Duvallet, E., Semerano, L., Assier, E., & Boissier, M. C. (2010). Anti-cytokine vaccination in autoimmune
diseases. Swiss medical weekly, 140, w13108. https://doi.
org/10.4414/smw.2010.13108
Table of ContentsCitrullination in Disease:
The Role of Autoantibodies
Introduction
The dynamic landscape of proteomics is profoundly
influenced by post-translational modifications (PTMs),
which modulate protein structure, function, stability,
localization, and enzymatic activity. Among these
modifications, citrullination has attracted significant
research interest across various diseases due to its
involvement in processes related to chronic inflammation and its diagnostic relevance in autoimmune
disorders (1).
Citrullination, a specific PTM, entails the enzymatic
conversion of arginine residues into citrulline by the
family of peptidylarginine deiminases (PADs) (Figure
3). This process, which is still not fully understood in
the context of standard cellular activities, predominantly transpires under conditions of cellular stress.
These conditions are often accompanied by inflammation, autophagy, and biological processes that
increase calcium levels required for citrullination such
as apoptosis, necrosis, and oxidative stress.
Profiling anti-citrullinated protein autoantibodies, or
ACPAs adds valuable insight into the role of citrullination in disease. ACPAs are well-established biomarkers in autoimmune diseases like rheumatoid arthritis,
where they are detected years before clinical symptoms appear and correlate with disease severity and
progression. Beyond autoimmune disorders, recent
studies have suggested that citrullinated proteins
may also play a role in cancer, neurodegenerative diseases, and chronic inflammatory conditions, making
ACPAs a potential tool for early diagnosis and patient
stratification in these contexts as well.
By incorporating ACPA profiling, researchers can not
only deepen their understanding of citrullination in
various disease mechanisms but also identify new
diagnostic and therapeutic targets across a range
Figure 3. The citrullination process
Page 21of conditions. This approach highlights the broader
value of autoantibody profiling in precision medicine,
particularly for complex diseases driven by inflammatory and stress-related pathways.
Citrullination in the Context of
Complex Diseases
Recent investigations have shed light on the extensive role of citrullination within the etiology of
complex diseases and its association with the innate
immune system (Table 4). In rheumatoid arthritis (RA),
ACPAs are detectable years before symptoms appear, serving as diagnostic markers in 70% of cases
(2-4). They also correlate with disease prognosis (1).
The elevated levels of PAD enzymes observed in
various carcinomas hint at the potential diagnostic
utility of citrullinated proteins in oncology. PAD4, for
example, has been detected in the blood of patients
with breast, lung, colon, ovarian and prostate cancers
(5,6).
Additionally, links between citrullination and neurodegenerative diseases have been explored. In
patients with Alzheimer’s disease, researchers have
discovered citrullinated beta-amyloid protein in the
brain (7). A meta-analysis of blood metabolites from
dementia patients showed a significant increase in
citrulline levels (8). Additionally, a recent structural
analysis identified a potential citrullination site on an
arginine residue in TDP-43 protein from patients with
frontotemporal lobar degeneration (9). Proteins with
disordered tertiary structures, such as arginine, are
known to undergo citrullination readily (1). The presence of citrullinated proteins in neurological conditions opens avenues for novel diagnostic approaches.
The opportunity to develop new diagnostic methods
using autoantibodies against citrullinated proteins
has yet to be fully investigated.
Challenges and Advances in
Citrullination Research
The study of citrullination, characterized by its low
abundance as a PTM, necessitates highly sensitive
detection techniques. While traditional methods like
ELISA and western blotting offer insights into the
distribution of citrullinated proteins and PADs, they
are limited by low throughput and semi-quantitative
analysis.
Mass spectrometry (MS) and protein microarrays have
emerged as powerful tools for the high-throughput,
sensitive profiling of citrullination, each presenting
distinct advantages and limitations in the context of
sample processing and analytical specificity.
MS offers diverse methodologies to detect citrullination, tailored to specific sample requirements and
investigative goals. Predominantly, a bottom-up proteomics approach using tandem MS is utilized for the
direct identification of citrullinated proteins.
Page 22
CITRULLINATION IN DISEASE
Table 4. Proteins commonly citrullinated in disease
Tissue PAD Proteins Disease
Connective PAD2, PAD4
Fibrinogen, Vimentin,
Fibrin Collagen Type
II, Enolase
Rheumatoid
Arthritis
Tumorous PAD2, PAD4 p53, p21, p300,
ETS Like-1, Histone Cancer
White Matter PAD2 Myelin Basic Protein Multiple Sclerosis
Central Nervous
System PAD2, PAD4
Vimentin, Myelin
Basic Protein, Glial
Fibrillary Acidic
Protein
Alzheimer’s
Disease
Skin PAD1, PAD3 Filaggrin Psoriasis
Eye PAD2 Myelin Basic Protein GlaucomaCITRULLINATION IN DISEASE
Page 23
While a very sensitive approach, MS demands skilled
technicians, involves multiple procedural steps susceptible to errors, and necessitates the use of expensive high-resolution mass spectrometers (10,11).
A notable challenge with MS is that it is difficult to
distinguish between citrullination and the deamidation of glutamine or asparagine.
Protein microarrays present an alternative for the
high-throughput and sensitive detection of citrullination, employed via direct or indirect methods. The
direct method involves the application of labeled
PAD enzymes to an array, enabling the screening of
hundreds to thousands of immobilized proteins to
identify those undergoing citrullination. This approach has led to the discovery of several new citrullinated protein substrates, predominantly involved
in glycolysis, although these findings have yet to be
directly linked to disease-specific citrullination (12).
Indirectly, protein arrays are used to detect autoantibodies against citrullinated proteins. This method has broad applications, including early disease
detection, disease subtyping, patient stratification,
and the identification of new therapeutic targets and
pathways. It also provides insights into the immune
system’s response to this PTM.
As an example of studying citrullination using the indirect approach, sera from both anti-cyclic citrullinated peptide (CCP) positive and negative patients with
diverse pathologies were analyzed using a high-density protein array. Researchers identified 844 autoantibodies, many previously undiscovered, differentiating
between patient groups. This indicates potential for
further stratification of RA patients and suggests that
anti-CCP negative patients might be incorrectly diagnosed through conventional methods (13).
The analysis of autoantibody biomarkers via protein arrays offers significant advantages in studying
citrullination’s role in disease. Antibodies, typically
analyzed in serum, represent a complete spectrum of
circulating antibodies, are stable, and can be present long before disease symptoms manifest. This
technique requires minimal sample preparation and
training. Additionally, analyzing multiple antibody
isotypes concurrently (e.g., IgG and IgA) can provide
comprehensive information on the immune response,
enhancing the accuracy of disease detection and
monitoring treatment efficacy (14).
Implications for Disease Diagnosis
and Therapeutic Development
Citrullinated antigens are increasingly recognized for
their potential in therapeutic applications. The specificity of citrullination to diseased and autophagic
tissues makes citrullinated proteins attractive targets
for therapies aimed at reducing inflammation while
preserving healthy cells. This approach is particularly
relevant for conditions like cancer and RA.
CITRULLINATION ASSAY
Profile Autoantibodies to
Citrullinated Proteins
Sengenics offers a unique citrullination
assay, designed to study ACPAs, with our
functional protein arrays.
Contact UsCITRULLINATION IN DISEASE
For instance, vimentin, which becomes highly citrullinated in metastatic epithelial tumors but remains
unmodified in normal tissue, has been explored as
a therapeutic target. Studies in mice with melanoma
have demonstrated that immunization with citrullinated vimentin significantly enhances survival rates compared to controls receiving placebo. Importantly, this
strategy inflicts minimal damage on healthy tissues,
underscoring the precision of targeting citrullinated
proteins for disease treatment (15).
Sonoma Biotherapeutics recently shared promising preclinical findings for SBT-77-7101, a chimeric
antigen receptor (CAR) T-cell therapy designed to
identify and target citrullinated proteins in patients
with RA, aiming to alleviate pain and inflammation.
These preclinical studies successfully demonstrated the CAR T-cells’ ability to recognize citrullinated
proteins, directly addressing the inflammation at its
source. With clinical trials slated to start in early 2024
(16), this innovative approach highlights the potential
of using CAR T-cells against citrullinated proteins as a
more targeted and potentially less side-effect-prone
treatment compared to existing monoclonal antibody
and CAR T-cell therapies. This strategy opens up new
possibilities for creating highly specific treatments by
leveraging the identification of citrullinated proteins.
Conclusion
The exploration of citrullination has unveiled its close
association with a broad spectrum of diseases, particularly in conditions characterized by chronic inflammation and immune dysregulation. The sensitivity of
current methodologies, including mass spectrometry
and protein microarrays, has significantly advanced
our ability to detect and analyze citrullinated proteins,
thereby enhancing our understanding of their role
in disease. The potential diagnostic and therapeutic
applications arising from this research highlight a
promising approach for the development of targeted
treatments and early detection strategies. Moreover,
the specificity of citrullination to diseased tissues
offers a unique biomarker for distinguishing diseased
from healthy states, promising more precise and less
invasive diagnostic tools.
Page 24CITRULLINATION IN DISEASE
References
1. Gudmann, N. S., Hansen, N. U., Jensen, A. C., Karsdal, M.
A., & Siebuhr, A. S. (2015). Biological relevance of citrullinations: diagnostic, prognostic and therapeutic options.
Autoimmunity, 48(2), 73-79. https://doi.org/10.3109/089169
34.2014.962024
2. Rantapää-Dahlqvist, S., de Jong, B. A., Berglin, E., Hallmans,
G., Wadell, G., Stenlund, H., Sundin, U., & van Venrooij, W.
J. (2003). Antibodies against cyclic citrullinated peptide and
IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum, 48(10), 2741-2749. https://doi.
org/10.1002/art.11223
3. Harada, K., Carr, S. M., Shrestha, A., & La Thangue, N. B.
(2023). Citrullination and the protein code: crosstalk between post-translational modifications in cancer. Philos Trans
R Soc Lond B Biol Sci, 378(1890), 20220243. https://doi.
org/10.1098/rstb.2022.0243
4. Schellekens, G. A., de Jong, B. A., van den Hoogen, F. H.,
van de Putte, L. B., & van Venrooij, W. J. (1998). Citrulline
is an essential constituent of antigenic determinants recognized by rheumatoid arthritis-specific autoantibodies. J Clin
Invest, 101(1), 273-281. https://doi.org/10.1172/JCI1316
5. Chang, X., & Han, J. (2006). Expression of peptidylarginine deiminase type 4 (PAD4) in various tumors. Molecular
Carcinogenesis, 45(3), 183-196. https://doi.org/https://doi.
org/10.1002/mc.20169
6. Lastwika, K. J., Kunihiro, A., Solan, J. L., Zhang, Y., Taverne, L. R., Shelley, D., Rho, J. H., Randolph, T. W., Li, C. I.,
Grogan, E. L., Massion, P. P., Fitzpatrick, A. L., MacPherson,
D., Houghton, A. M., & Lampe, P. D. (2023). Posttranslational modifications induce autoantibodies with risk prediction
capability in patients with small cell lung cancer. Sci Transl
Med, 15(678), eadd8469. https://doi.org/10.1126/scitranslmed.add8469
7. Mukherjee, S., Perez, K. A., Dubois, C., Nisbet, R. M., Li, Q.
X., Varghese, S., Jin, L., Birchall, I., Streltsov, V. A., Vella, L.
J., McLean, C., Barham, K. J., Roberts, B. R., & Masters, C.
L. (2021). Citrullination of Amyloid-b Peptides in Alzheimer’s
Disease. ACS Chem Neurosci, 12(19), 3719-3732. https://
doi.org/10.1021/acschemneuro.1c00474
8. Zinellu, A., Tommasi, S., Sedda, S., & Mangoni, A. A. (2023).
Circulating arginine metabolites in Alzheimer’s disease and
vascular dementia: A systematic review and meta-analysis.
Ageing Research Reviews, 92, 102139. https://doi.org/
https://doi.org/10.1016/j.arr.2023.102139
9. Arseni, D., Chen, R., Murzin, A. G., Peak-Chew, S. Y., Garringer, H. J., Newell, K. L., Kametani, F., Robinson, A. C.,
Vidal, R., Ghetti, B., Hasegawa, M., & Ryskeldi-Falcon, B.
(2023). TDP-43 forms amyloid filaments with a distinct fold in
type A FTLD-TDP. Nature, 620(7975), 898-903. https://doi.
org/10.1038/s41586-023-06405-w
10. De Ceuleneer, M., Van Steendam, K., Dhaenens, M., &
Deforce, D. (2012). In vivo relevance of citrullinated proteins
and the challenges in their detection. Proteomics, 12(6), 752-
760. https://doi.org/10.1002/pmic.201100478
11. Rebak, A. S., Hendriks, I. A., & Nielsen, M. L. (2023). Characterizing citrullination by mass spectrometry-based proteomics. Philos Trans R Soc Lond B Biol Sci, 378(1890), 20220237.
https://doi.org/10.1098/rstb.2022.0237
12. Thomas, M. A., Kim, S. Y., Curran, A. M., Smith, B., Antiochos, B., Na, C. H., & Darrah, E. (2023). An unbiased proteomic analysis of PAD4 in human monocytes: novel substrates, binding partners and subcellular localizations. Philos
Trans R Soc Lond B Biol Sci, 378(1890), 20220477. https://
doi.org/10.1098/rstb.2022.0477
13. Poulsen, T. B. G., Damgaard, D., Jorgensen, M. M., Senolt,
L., Blackburn, J. M., Nielsen, C. H., & Stensballe, A. (2021).
Identification of potential autoantigens in anti-CCP-positive
and anti-CCP-negative rheumatoid arthritis using citrulline-specific protein arrays. Sci Rep, 11(1), 17300. https://doi.
org/10.1038/s41598-021-96675-z
14. Sieghart, D., Konrad, C., Swiniarski, S., Haslacher, H., Aletaha, D., & Steiner, G. (2023). The diagnostic and prognostic
value of IgG and IgA anti-citrullinated protein antibodies in
patients with early rheumatoid arthritis [Brief Research Report]. Frontiers in Immunology, 13. https://doi.org/10.3389/
fimmu.2022.1096866
15. Brentville, V. A., Metheringham, R. L., Gunn, B., Symonds,
P., Daniels, I., Gijon, M., Cook, K., Xue, W., & Durrant, L.
G. (2016). Citrullinated Vimentin Presented on MHC-II in
Tumor Cells Is a Target for CD4+ T-Cell–Mediated Antitumor Immunity. Cancer Research, 76(3), 548-560. https://doi.
org/10.1158/0008-5472.Can-15-1085
16. Charmsaz, S., Tracy, J., Whalen, E., Bui, J., van der Vuurst de
Vries, A., Malmstrom, V., & Blake, M. (2023, 11/12/2023). Detection of Citrullinated Proteins Recognized by a Novel Chimeric Antigen Receptor TregTherapy in Both Synovial Fluid
and Serum from Patients with Rheumatoid Arthritis. American
College of Rheumatology Convergence 2023, San Diego.
Table of ContentsWHITE PAPER
Address Key Challenges in Vaccine Development with Antibody Profiling
Explore how antibody profiling can guide the development of safer, more effective vaccines.
Read Now
Page 26
Profiling Autoantibodies
with Precision
Introduction
Protein microarrays provide a fast and cost-effective
way to profile autoantibodies (AAbs) against hundreds to thousands of antigens at once. This makes
them a powerful tool for studying the immune system, advancing both biomedical research and diagnostic capabilities.
Profiling antibodies and AAbs enables the discovery
of targetable and exploitable biomarkers: protein
antigens, pathogenic antibodies, and protective
antibodies. These insights advance precision medicine for improved patient outcomes. Here are some
applications of protein microarrays:
• Vaccine and Drug Development: Validate target specificity and efficacy while mapping immune-targeted diversification to reveal how the
immune system responds to different antigens
over time.
• Patient Stratification and Subtyping: Identify subtypes within heterogeneous diseases and select
patients for clinical trials who are most likely to
benefit, reducing variability and increasing statistical power.
• Response Prediction: Predict patient outcomes
to treatments, enabling personalized plans and
minimizing the risk of immune-related adverse
reactions.
• Understanding Disease Mechanisms: Discover
disease-associated proteins and AAbs to uncover
therapeutic targets, reveal early diagnostic biomarkers, and track disease progression.
Most protein microarrays use peptides or proteins
that are misfolded, denatured, or fragmented, resulting in the loss of conformational epitopes—specific
three-dimensional (3D) structures recognized by 90%
of AAbs (1,2). In additiona, new, non-native epitopes
that are not typically present in vivo are exposed,
leading to non-specific binding and increased false
positives and negatives.
To ensure precise antibody-antigen interactions and
obtain reliable data, it is crucial to use correctly folded proteins. Selecting the right protein microarray is
therefore key to achieving high-quality results.PROFILING AUTOANTIBODIES WITH PRECISION
Visualizing Immune Interactions with
Protein Microarrays
Protein microarrays are miniaturized immunoassays
that display a large number of antigens—such as
proteins, peptides, or fragments—on a solid surface,
usually a glass slide, in an organized, addressable format. To perform an assay, a test sample, often serum
rich in host antibodies, is applied to the array (Figure
4). Antibodies in the sample bind specifically to target
antigens on the array, creating antibody-antigen complexes.
Following antibody binding, a fluorescently-tagged
secondary antibody specific to the host species of the
sample is added. This secondary antibody binds to
the primary antibodies attached to the proteins, antibodies, enabling indirect visualization and detailed
immunoprofiling.
The fluorescence intensity is proportional to the
quantity of antibodies, whereas the fluorescence location on the array facilitates the identification of the
targeted antigen. Figure 4. Some protein microarrays can simultaneously detect
two antibody isotypes using fluorescently labeled antibodies.
For Research Use Only. Not for use in diagnostic procedures. Page 27
Antibody Subclasses Primary Function Serum Level (g/L)
IgA IgA1-2 Pathogen neutralization, anti-inflammatory, mucosal 0.6 - 4
IgD None Upper aerodigestive immunity, B-cell development,
immune regulation 0 - 0.14
IgE None Tumor surveillance, anti-venom defense,
anti-parasitic defense, type 1 hypersensitivity Trace
IgG IgG1-4 Circulating and tissue immunity 7 - 15
IgM None Immune surveillance, acute response
(e.g., agglutination, complement activation) 0.6 - 3
Table 5. Antibody isotypes: Primary functions and serum reference ranges in adults
Depending on the array, multiple antibody isotypes
(i.e., IgG, IgM, IgA, IgE) with unique effector functions may be analyzed simultaneously (Table 5). This
capability is made possible with the use of secondary
antibodies that bind to specific isotypes and are labeled with different fluorophores. Measuring various
isotypes offers a more comprehensive view of disease, delivering valuable insights into the timing and
localization of the immune response (3).PROFILING AUTOANTIBODIES WITH PRECISION
For Research Use Only. Not for use in diagnostic procedures. Page 28
Correct Protein Folding with
KREX® Technology
Sengenics’s proprietary KREX technology ensures
that only correctly folded proteins are used for AAb
profiling (Figure 5). Each protein is tagged with a
small 10 kDa subunit of biotin carboxyl carrier protein
(BCCP), which acts as a folding marker. If a protein is
misfolded or fragmented, the BCCP misfolds as well,
concealing its biotinylation site and preventing it from
attaching to the streptavidin-coated array surface.
This allows only properly folded proteins to be immobilized for AAb profiling, while misfolded proteins are
washed away, removing them from further analysis.
Unlike other immobilization tags, such as glutathione
S-transferase (GST) or maltose-binding protein (MBP),
BCCP uniquely supports the retention of correctly
folded proteins. Additionally, the non-denaturing surface of Sengenics microarrays preserves the three-dimensional protein structures essential for accurate
AAb binding (4).
Finally, recombinant human proteins on Sengenics arrays are expressed in insect cells, which more closely
replicate protein processing of mammals compared
to bacteria or yeast (4,5). For instance, protein expression in the commonly employed bacterial system,
Escherichia coli, frequently results in insoluble, poorly
folded proteins. Yeast expression systems, such as
Saccharomyces cerevisiae and Pichia pastoris, do
not mimic glycosylation patterns of mammalian cells
well, and the harsh cell lysis conditions often lead to
denatured or fragmented proteins. Therefore, the use
of insect cells aligns more closely with mammalian
systems, significantly improving the functional expression of human proteins.
Other Arrays
High % of false positives
High background noise
Sengenics Arrays
True hits
High specificity
Figure 6. Sengenics micoarrays reveal distinct and specific hits
while data from other arrays are characterized by a high number
of non-specific hits contributing to elevated background noise.
RFU = relative fluorescence units
Figure 5. KREX technology for precise antibody profilingPROFILING AUTOANTIBODIES WITH PRECISION
Key Features of KREX Technology
Native Protein Structure: Full-length proteins with
intact epitopes, ensuring exceptional specificity and a
low false discovery rate (FDR) < 1% (Figure 6)
High Sensitivity and Wide Linear Range: Picomolar
(pM) sensitivity and a linear range spanning over 4
logs
Highly Reproducible: Mean intra-array CV < 10% and
an inter-batch Pearson correlation (R2) > 0.95 (Figure
7)
Versatile and Scalable: Ideal for various applications
and study sizes, consuming minimal sample (< 50 μL)
without compromising on quality
Dual Isotype Analysis: Simultaneous insights into two
antibody isotypes (e.g., IgG and IgA or IgM)
Multiple Measurements: Three or four replicates per
antigen enable more robust and reliable data
Expertly Curated Library
Sengenics’s extensive library of over 2000 human proteins has been carefully curated by immunologists for
their roles as established and potential autoantigens.
In selecting these proteins, considerations included
their relevance in various diseases, biological functions, expression in specific tissues and compartments, and interactions with immune cells from both
the innate and adaptive immune systems.
This library covers a wide range of protein functional
classes and subcellular locations, providing comprehensive representation of critical disease-related
pathways and potential drug targets (Figure 8). It
includes essential immune regulators and modulators
such as cytokines, chemokines, and their receptors
(Table 6; see also the chapter on “Anti-Cytokine Autoantibodies”).
For Research Use Only. Not for use in diagnostic procedures. Page 29
Figure 8. Protein functional classes and disease categories represented by protein antigens in the Sengenics library.
Protein Functional Classes
Disease Categories
Kinase / Phosphatase
Scaffold / Adaptor
Transcription factor
Transmembrane signal receptor
Chromatin / Chromatin associated
Ribosomal
Ubiquitin
Chaperone
GTPase
Protease / Protease inhibitor
Other
Neuronal
Dermatologic
Hepatorenal
Hematologic
Immune
Endocrine
Cardiovascular
Gastrointestinal
Respiratory
Reproductive
Other Arrays
High CVs
Sengenics Arrays
Low CVs
Figure 7. Sengenics microarrays have a lower per-protein coefficients of variation percentage (CV%) than other arrays. Data
represents all proteins and the same 50 serum samples.PROFILING AUTOANTIBODIES WITH PRECISION
Importantly, human antigens are also highly relevant
in infectious disease research due to mechanisms
like molecular mimicry and epitope spreading, where
pathogens trigger immune responses that cross-react
with human proteins. Additionally, tissue damage and
inflammation from infections can directly elicit AAbs.
These factors broaden the platform’s applications,
making it valuable not only for autoimmune and cancer research but also for understanding host-pathogen interactions and immune responses in infectious
diseases.
Flexible, End-to-End Solutions
Sengenics offers both ready-to-use panels and fully
customizable options for antibody and AAb profiling,
tailored to specific protein targets (Table 7). This flexibility allows researchers to select or create arrays that
best meet their research objectives, whether they are
investigating specific diseases, identifying biomarkers, or studying immune responses. Sample analysis
is supported by a global network of certified service
providers, ensuring accessibility and consistent quality regardless of location.
At every stage, Sengenics provides comprehensive
support—from initial experimental design to advanced bioinformatics analysis—helping researchers maximize the value of their data. Detailed data
reports, complete with figures and statistical analyses,
are easily accessible through i-Ome® AI, a user-friendly, open-source data analysis platform from Sengenics. This platform streamlines data interpretation,
enabling researchers to quickly extract actionable
insights.
For Research Use Only. Not for use in diagnostic procedures. Page 30
Cytokines Chemokines Receptors
Activin A (INHBA)
Amphiregulin (AREG)
APRIL (TNFSF13)
G-CSF (CSF3)
IFNA2
IFNB2
IFNW1
IFNG
IL1A
IL1β
IL3
IL5
IL6
IL7
IL8 (CXCL8)
IL10
IL11
IL12A
IL12B
IL13
IL15
IL17A
IL17F
IL18
IL19
IL20
IL21
IL22
IL23 (IL23A+IL12B)
IL24
IL26
IL27
IL31
IL32
IL34
IL35
IL36
IL37
IL39 (IL23A+EBI3)
IL40 (C17orf99)
M-CSF (CSF2)
TNF
TSLP
CCL1
CCL2
CCL3
CCL7
CCL8
CCL13
CCL16
CCL17
CCL19
CCL20
CCL21
CCL22
CCL25
CCL27
CCL28
CXCL6
CXCL8
CXCL9
CXCL10
CXCL11
CXCL12
CXCL13
CXCL16
CXCL17
CX3CL1
Cytokine
Receptors
ACVR2A
ACVR2B
CSF3R
CXCR2
IL13RA1
IL13RA2
IL21R
IL6ST
Chemokine
Receptors
CCR5
CXCR2
CXCR4
CXCR6
Table 6. Cytokines, chemokines, and their receptors included in
the Sengenics protein libraryPROFILING AUTOANTIBODIES WITH PRECISION
Conclusion
Protein microarrays are a powerful tool for profiling
antibodies and AAbs, with applications that span biomarker discovery, disease diagnostics, and therapeutic development. However, to generate high-quality,
reliable data, it is essential to preserve the conformational epitopes recognized by the vast majority of
humoral antibodies. Sengenics microarrays address
this need through their proprietary KREX technology, ensuring that only correctly folded, functional
proteins are displayed for precise antibody-antigen
interactions.
By combining cutting-edge protein folding technology, a comprehensive selection of disease-relevant
protein antigens, and robust analytical support, Sengenics microarrays empower researchers to achieve
deeper insights into disease initiation and progression, ultimately driving advancements in personalized
medicine and targeted treatments.
References
1. Barlow DJ, Edwards MS, Thornton JM. Continuous and
discontinuous protein antigenic determinants. Nature. 1986
Aug 21-27;322(6081):747-8. doi: 10.1038/322747a0. PMID:
2427953.
2. Van Regenmortel MHV (1996). Mapping Epitope Structure
and Activity: From One-Dimensional Prediction to Four-Dimensional Description of Antigenic Specificity. Methods
(San Diego, Calif.), 9(3), 465–472. https://doi.org/10.1006/
meth.1996.0054
3. Janeway CA Jr, Travers P, Walport M, et al. Immunobiology:
The Immune System in Health and Disease. 5th edition. New
York: Garland Science; 2001.
4. Aziz, F., Smith, M., & M Blackburn, J. (2019). Autoantibody-Based Diagnostic Biomarkers: Technological Approaches to Discovery and Validation. IntechOpen. doi: 10.5772/
intechopen.75200
5. Duarte, J. S., J; Mulder, N; Blackburn, J. (2013). Protein
Functional Microarrays: Design, Use and Bioinformatic Analysis in Cancer Biomarker Discovery and Quantitation. In X.
Wang (Ed.), Bioinformatics of Human Proteomics (pp. 39-74).
Springer Science+Business Media Dordrecht.
For Research Use Only. Not for use in diagnostic procedures.
Microarray Protein
Number
Protein Panel
i-Ome® Discovery 1800+ Protein antigens representing a myriad of protein functional classes and disease categories for comprehensive biological and immunoproteomic insights
i-Ome® Cancer 500+
Cancer-associated proteins, encompassing tissue and pathway relevance, therapeutic targets, cytokines and chemokines, cancer-driver proteins, prognostic indicators,
cancer-testis antigens, B-cell and AAb targets, and ectopic expression
Autoimmune Disease
Autoantigen Panel 100+ Clinically relevant protein antigens linked to a wide array of autoimmune diseases, such as systemic lupus erythematosus , rheumatoid arthritis, Sjogren’s, and diabetes
OncoRex p53 Cancer 100+ variants p53 wildtype and mutant variants, enabling precise screening of therapeutic AAbs
and protein-binding compounds for cancer
Custom Project-specific Select from over 2000 proteins in our library or request your own
Table 7. Sengenics protein microarrays
Table of Contents© 2024 Sengenics Corporation LLC. All rights reserved. All trademarks are the property of Sengenics, LLC or
their respective owners.
All information in this document may change without notice and does not constitute any warranties, representations, or recommendations unless explicitly stated. Sengenics products and assay methods are covered by
several patents and patent applications: https://sengenics.com/about-us/company-overview/patents/
Sengenics Corporation LLC. Registered in Delaware, USA no. 5739583
Sengenics Corporation Pte Ltd. Registered in Singapore no. 201734100D
2536, vs1.1, 2024-11-08
About Us
Sengenics is a functional proteomics company that is committed to advancing precision medicine by empowering researchers with biologically relevant and actionable immunoproteomic insights across a broad spectrum of diseases. At the heart
of its mission, Sengenics offers advanced, high-throughput tools using proprietary
technology to precisely detect autoantibody biomarkers and protein interactions
for basic, translational, and clinical research. Its robust tools have been leveraged
by top pharmaceutical companies and leading research institutions to enhance
disease understanding and streamline the biomarker pipeline.
Connect with Us
enquiries@sengenics.com | sengenics.com
For Research Use Only. Not for use in diagnostic procedures.
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version