16
CHAPTER 23 Autoantibodies and Biomarker Discovery Ji Qiu, Karen S. Anderson Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA OUTLINE Introduction 363 History of the Detection of Autoantibodies 364 Immunouorescent Assays 364 Progressive Purication of Antigens 364 Targeted Detection of Autoantibodies 364 Proteomics Methods for the Detection of Autoantibodies 365 Phage Display of cDNA Libraries 365 Cellular Fractionation and Immunoblotting 369 Protein Microarrays 369 Peptide and Peptoid Arrays 370 Detection of Post-Translational Modications 371 Association of Autoantibodies with Disease States 372 Autoimmune Disorders 372 Type 1 Diabetes 372 Neurologic Disorders 372 Cancer 373 Challenges and Future Development 374 References 374 INTRODUCTION The immune system plays a fundamental role in the maintenance of homeostasis. Dysregula- tion of immune responses is observed in multiple pathogenic states, including autoimmu- nity, diabetes, neurologic disorders, and cancer. The mechanisms by which the immune system contributes to the pathogenesis of these diseases are being uncovered. Because the effectiveness of the immune system to ght infections relies on both adaptability and memory, there are clear genetic and environmental components to the depth and specicity of the immune response to a given antigen. B cells produce antibodies with target specicities, embedding genetic vari- ability both in the genomic rearrangement of the VDJ gene segments and the acquisition of somatic hypermutations in the variable regions, which leads to at least 10 11 potential unique anti- body structures. Antibodies that target self- proteins are called autoantibodies to distinguish Proteomic and Metabolomic Approaches to Biomarker Discovery http://dx.doi.org/10.1016/B978-0-12-394446-7.00023-6 Copyright Ó 2013 Elsevier Inc. All rights reserved. 363

Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

  • Upload
    ji

  • View
    214

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

C H A

P T E R

23

Autoantibodies and Biomarker DiscoveryJi Qiu, Karen S. Anderson

Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA

Ph

O U T L I N E

Introduction 36

3

roteomttp://

History of the Detection of Autoantibodies

364

ic andx.do

Immunofluorescent Assays

364 Progressive Purification of Antigens 364 Targeted Detection of Autoantibodies 364

Proteomics Methods for the Detectionof Autoantibodies 365

Phage Display of cDNA Libraries

365 Cellular Fractionation and Immunoblotting 369 Protein Microarrays 369 Peptide and Peptoid Arrays 370

d Metabolomic Approaches to Biomarker Discoveryi.org/10.1016/B978-0-12-394446-7.00023-6 363

Detection of Post-Translational Modifications

371

Association of Autoantibodies with DiseaseStates 372

Autoimmune Disorders

372 Type 1 Diabetes 372 Neurologic Disorders 372 Cancer 373

Challenges and Future Development 374

References 374

INTRODUCTION

The immune system plays a fundamental rolein the maintenance of homeostasis. Dysregula-tion of immune responses is observed inmultiple pathogenic states, including autoimmu-nity, diabetes, neurologic disorders, and cancer.The mechanisms by which the immune systemcontributes to the pathogenesis of these diseasesare being uncovered. Because the effectiveness ofthe immune system to fight infections relies on

both adaptability and memory, there are cleargenetic and environmental components to thedepth and specificity of the immune responseto a given antigen. B cells produce antibodieswith target specificities, embedding genetic vari-ability both in the genomic rearrangement of theVDJ gene segments and the acquisition ofsomatic hypermutations in the variable regions,which leads to at least 1011 potential unique anti-body structures. Antibodies that target self-proteins are called autoantibodies to distinguish

Copyright � 2013 Elsevier Inc. All rights reserved.

Page 2: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY364

them from antibodies produced by our immunesystem when encountering foreign antigens.Genomic sequencing has identified approxi-mately 20,000 unique open reading frames inthe human genome as source antigenic struc-tures. Tissue-specific splice variation and poly-morphisms add significant variation to thecomplexity of self-antigens. As a result, ourunderstanding of immune responses requiresa systems immunology proteomics approachthat can produce thousands of protein structuresto identify antibody signatures that are associ-ated with disease. In this chapter, we focus onthe application of diverse proteomic technolo-gies for the discovery of autoantigens.

History of the Detectionof Autoantibodies

Immunofluorescent Assays

The development of techniques for the detec-tion of autoantibodies has focused on diseasesassociated with autoimmunity, where a directrelation of autoantibody titer to disease patho-genesis has been established. The presence ofdisease-specific antibodies in the sera of patientswith autoimmune diseases was first describedmore than half a century ago.1e6 Initial studiesof sera from patients with systemic lupus ery-thematosus (SLE) led to the discovery of themajor class of autoantibodies, antinuclear anti-bodies (ANAs).7,8 ANAs were initially detectedwith immunofluorescent assays (IFA) usingcultured cells probed with patient sera,9e13 butidentification of the specific targets of theseANAs has been limited. Molecular characteriza-tion of autoantigens is required to transformthese cell-based semiquantitative assays intoreproducible, quantitative assays to improvediagnosis and disease monitoring. Determiningthe identities of these autoantigens will furtherour understanding of the underlying mecha-nisms of disease pathogenesis in autoimmunityand devise therapeutics for diseasemanagement.

Progressive Purification of Antigens

Identification of autoantigens has paralleledthe technological development in immuno-assays over the past six decades, from crudecellular fractions14,15 to chromatographicpeaks,16 gel bands,17e20 and the identificationof protein structures. One of the first autoanti-bodies identified was rheumatoid factor (RF),an antibody against the Fc domain of humanimmunoglobulins. Targets of ANAs were identi-fied using crude characterizations of antigens incellular/nuclear extracts by immunodiffusion,probing with sera from patients with Sjogren’ssyndrome and SLE. This work led to thediscovery of several well-known antigens suchas Ro/SSA, La/SSB, Sm, RNP, Scl-70, and Jo-1,which are intracellular antigens complexedwith nucleic acids.21 Development of proteinelectrophoresis, immunoblotting, and immuno-precipitation techniques provided tools to refineour knowledge of autoantigens to the level ofmolecular weight. It was not until the adventof molecular biology that the exact autoantigenscould be identified at the molecular level.

Targeted Detection of Autoantibodies

Initially, candidate autoantigens were chosenbased on prior experimental and/or theoreticalknowledgeof theirpotential roles indiseasedevel-opment. Inparticular, oncogenes thatare critical tocancer development and overexpressed in cancerswere selected for discovery of autoantibodies.These oncogenes were purified as recombinantproteins from E. coli and tested in ELISA orWestern blot for antibodies in the sera of cancerpatients. This hypothesis-driven approach ledto the identification of autoantibodies againstseveral oncogenes in cancers. For example, anti-c-myc autoantibodies were identified by Westernblot in 25 out of 44 sera from patients with colo-rectal cancer, but in 8 out 46 normal donors.22

Autoantibodies against other oncogenes such asras or HER-2/neu have also been reported.23,24

However, the discovery of autoantibody

Page 3: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

PROTEOMICS METHODS FOR THE DETECTION OF AUTOANTIBODIES 365

biomarkers in diseases using this approach hasbeen limited to selected proteins. Because the rela-tionship between protein structure, proteinexpression, and antigenicity is not well under-stood, there is a need for broad-based discoveryof autoantibodies without selection bias.

PROTEOMICS METHODS FOR THEDETECTION OF AUTOANTIBODIES

Since the beginning of the 21st century, thedevelopment and application of numerous pro-teomics technologies has enabled high-throughput protein expression for autoantigendetection. Based on the source and type ofantigen repertoires, these technologies aredivided into four categories. Antigens havebeen expressed as whole proteins from cDNAlibraries, separated and isolated from cell lysatesand displayed in protein array format, orpeptides can be displayed in microarray format(see Figure 1). Each method displays uniquestructural determinants and contributes to ouroverall understanding of structural immunoge-nicity (Tables 1 and 2).

Phage Display of cDNA Libraries

After the development of immunofluorescentassays, serological identification of recombi-nantly expressed clones (SEREX), initiallydescribed in 1995, represents one of the earliestdevelopmentof this approach that revolutionizedthe discovery of autoantigens.25 In this strategy,cDNA libraries are generated from diseasetissues, cell lines, or testis tissue and the candidateantigens are expressed either directly in E. coli orusing a lambda phage expression system. Colo-nies or phage plaques containing expressedproteins are then replicated onto nitrocellulosemembranes and probed with patient sera(Figure 1, top). Positive clones are sequenced toidentify the autoantigens. This approachwasfirstapplied to cancer but has been widely used for

other diseases.26e30 The autoantigen NY-ESO-1was first identified in esophageal cancer patientsera and was later detected in the sera of patientswith many different cancers. These findings ledto clinical trials targeting NY-ESO-1 for cancerimmunotherapy.31 One key advantage of SEREXis that the cDNA library expression reflects theexpression levels, mutations, and splice varia-tions that are inherent to the specific tissuesfrom which the library is derived. However,SEREX identification of autoantigens has severallimitations. Full-length cDNAs are rarely clonedinto the expression vectors and truncated expres-sion limits coverage. Many of the autoantibodiesthat have been identified target frame shift poly-peptide products, which have yet not beenproven to be expressed at the protein level in therelevant tissue. E. coli expression machinery haspoor expression of high molecular weightproteins or proteins with multiple domains andlacks chaperones to facilitate proper folding.This limits the amount, type, and conformationsof proteins presented for screening. Plaque-based screening is also not amenable to automa-tion and quantitative analysis.

Several recent advances with phage-basedexpression have resulted in the identification ofmultiple new autoantigens and have facilitatedautomation. Lysates of individual E. coli colonieshave been spotted in microarray format for sero-logic screening.32 The use of filamentous T7 orM13 phage for protein expression results insuperior throughput compared with lambdaphage due to compatibility with biopanning.33,34

More recently, solution-based phage display hasbeen developed to further identify autoantigensexpressed in amore native format. Phage displayis compatible with rapid biopanning for negativeand positive selection and has been integratedwith microarray platforms to improve high-throughputmultiplexing and quantitation. Theseadvances have led to the identification of auto-antibody biomarkers in several cancers suchas ovary and prostate.33e35 In summary, theprimary advantages of phage-based expression

Page 4: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

Pooled sera from patients

Cell lysate Multidimensional fractionation in solution or gel

Print fractions on multiple slides

Probe with sera

200 40060080010001200m/z

Identify proteins in informative fractions

Probe library display for recognized antigens

Protein arrays (left) or bead arrays (right) with candidate antigens

Probe with sera Determine sensitivity and specificity of antigens

cDNA library screening and phage display:

Cell fractionation:

Protein microarray:

Print peptide arrays

Peptide display:

Blot 2D gels

Display peptides by phage Probe with sera Determine sensitivity and specificity of peptides

Display protein by phage Biopanning for informative antigens

Phage array screening

FIGURE 1 Methods of antigen identification. In traditional library screening (top row), cDNA libraries from target tissueare expressed by phage, blotted on a membrane or panned in solution, and probed with patient sera. Confirmation ofsensitivity and specificity requires recombinant expression of protein for ELISA analysis. Cell fractionation (second row)involved separation of cell lysates by two-dimensional or three-dimensional separations in solution phase or in gel phase,followed by probing with patient sera. Positive fractions are identified by mass spectrometry and confirmed with ELISA.Protein microarrays (third row) can be printed using recombinant proteins or proteins translated in situ, on slides or on beadarrays. Because the arrays are addressable, no further antigen identification is required. Similarly, peptides (bottom) can beprinted on slides or displayed by T7 phage for rapid serum screening. (Modified from reference #109, reprinted with permission.)

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY366

Page 5: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

TABLE 1 Comparison of Different Technologies for Autoantibody Discovery

Antigen sources Throughput Multiplexity Target identificationSampleconsumption

cDNA expression

Phage plaque Low. Rounds of plaquescreening.

Medium. Many plaquescan be screened on onemembrane lift.

Easy. DNAsequencing.

High

Phage display& microarray

Medium. Screening onspotted phages.

High Easy. DNAsequencing.

High for panning.Low for arrays.

Bacterial expression& microarray

High. Bacterial coloniesare spotted on glassslides.

High Easy. DNAsequencing.

Low

Cell lysates

2D Western Low. 2D SDS-PAGE andWestern blot.

High. Hundreds ofspots on each gel.

Medium-hard. Massspec of positive spots.

Medium

Natural protein array Medium. Thousands offractions obtained fromliquid phase separationof cell lysates.

High Medium-hard. Massspec of positivefractions.

Low

Recombinant proteins

Purified proteins High High Easy Low

In vitro expressionmixtures

High High Easy Low

On-chip synthesizedproteins

High High Easy Low

Synthetic peptides

Synthetic peptides/peptoids on arrays

High High Easy for spatiallyaddressed arrays.NMR analysis maybe neededfor peptoid arrays.

Low

Synthetic humanpeptidome & phagedisplay

Low. Rounds of panningand NextGen sequencing.

High Hard. Sophisticatedanalysis of NextGensequencing data.

Medium

Microbial surfacedisplay

Low. Rounds ofpanning.

High Easy. DNAsequencing.

Medium

PROTEOMICS METHODS FOR THE DETECTION OF AUTOANTIBODIES 367

systems are the ability to rapidly convert a targetlibrary for screening low numbers of serafor autoantigen discovery without specificequipment. However, limitations such as the

overrepresentation of abundant proteins, frameshifts, and lower throughput have limitedthe ultimate translation of these biomarkers intoclinical practice.

Page 6: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

TABLE 2 Comparison of Different Technologies for Autoantibody Discovery

Antigen sources Proteome coverage

Neoantigens fromalternative splicing/mutation/translocation

Post-translationalmodifications

cDNA expression

Phage plaque Low. Limited by bacterialexpression and minimalcoverage of low abundanceproteins. Truncations andframe shifts are prevalent.

Yes No

Phage display & microarray Low. Same as above. Yes No

Bacterial expression &microarray

Medium. Limited bybacterial expression andminimal coverage of lowabundance proteins.

Yes No

Cell lysates

2D Western Medium. Only proteinscompatible with 2D gel.

Yes Yes

Natural protein array Medium. Only proteinscompatible withmultidimensional column-based separation.

Yes Yes

Recombinant proteins

Purified proteins High. Limited byORFeome.

No if without priorknowledge and intentionto include a particularneoantigen.

No

In vitro expressionmixtures

High. Limited byORFeome.

No No

On-chip synthesized proteins High No No in the standard format.Possible to study certainPTMs.

Synthetic peptides

Synthetic peptides/peptoidson arrays

N/A Maybe. If a syntheticpeptide happens to havethe same sequence.

No

Synthetic human peptidome &phage display

High No No

Microbial surface display Low to medium Maybe No

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY368

Page 7: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

PROTEOMICS METHODS FOR THE DETECTION OF AUTOANTIBODIES 369

Cellular Fractionation andImmunoblotting

A primary concern with the use of phagedisplay, as well as solution-based display, isthat proteins are not expressed in native formand post-translational modifications are not rep-resented. An alternative approach uses relevanttissue lysates as the source of candidate antigens,which are then probed with patient sera.Initially, autoantigens were observed directly incell lysates using immunodiffusion, but thecomplexity of protein content within lysateslimited this approach until the innovation oftechniques in protein separation by columnchromatography and gel electrophoresis per-mitted sufficient separation of protein content.Currently, lysate preparation requires two-dimensional and three-dimensional proteinseparation coupled with target identification bymass spectrometry.

Two-dimensional immunoblotting (2D-Western) is well established for the assessmentof antibody reactivity and has been applied toautoantigen discovery. Two-dimensional gelelectrophoresis separates proteins in complexbiological samples based on isoelectric pointand molecular weight. Lysate preparation andgel composition can be customized to improvethe separation of specific protein classes.After addition of sera, immunogenic spots areexcised and sequenced for protein identificationby mass spectrometry.36,37 Because immunoblot-ting favors the detection of denatured epitopes,solution-based methods of antigen detectionwere developed. Multidimensional separationin liquid phase by reverse phase/ion exchangechromatography and chromatofocusing inconjunction with arraying thousands of proteinfactions prepared from cell/tissue lysates resultsin native protein arrays that contain relevantpost-translational modifications (PTMs).38 Forboth 2D-Western and native protein arrays, theautoantigens can be prepared with relative easewithout prior cloning and purification. The

antigens are displayed in the physiologicallyrelevant form that initially elicited the humoralimmune response. However, proteins in cell/tissue lysates exist in varying abundance thatdiffers by orders of magnitude, and the dynamicrange of analysis is limited. The signals from lowabundance proteins can be buried by high abun-dant housekeeping proteins. The identificationof target antigens within a specific spot or frac-tion requires deconvolution by mass spectrom-etry and often requires further targetedseparation to confirm the identity of the anti-gens. Translation of these targets to clinicalbiomarker assays is also a major challenge;high-throughput serologic validation assaysrequire recombinant protein production thatmay not display the antigenic PTMs.

Protein Microarrays

The development of protein microarrays hasnow enabled the simultaneous analysis of thou-sands of different proteins on the footprint ofa microscopic slide or in bead-array format.With the development of high-throughputprotein production, it is now possible to produceproteins that are limited only by ORFeomecontent. In the near term, protein arrays display-ing the entire human proteome will soon beavailable, although protein arrays that reflectthe genetic variability of tumors and the splicevariation of tissues will take longer to develop.One natural advantage of protein microarraysis the rapid, multiplexed measurement of anti-body levels in sera.

There are three primary methods forproducing protein microarrays. First, proteinsare expressed in model organisms such asE. coli, yeast, or insect cells, purified, and printedon the arrays.39e44 In vitro protein production isa challenging process that can be hindered byprotein purity, batch-to-batch variation, andlimited stability of purified protein products.The second approach is to use printed antibodyarrays to detect antigen/antibody complexes

Page 8: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY370

within sera, which is limited to antigens thatare present in sera and the availability of captureantibodies for array printing. We, and others,have developed in vitro transcription/translationmethods for expression and capture of targettagged antigens onto an array format: nucleicacid programmable protein arrays (NAPPA);see Figure 2. These arrays have the advantage ofreal-time protein production for serologic detec-tion. However, protein arrays in general havelimited PTMs and are limited by variable proteinexpression, folding, and reproducibility.45e48

A variation on spotting purified proteins is tospot mixtures of proteins that are produced inmultiwell plates using PCR amplified genes andin vitro expression system.42,49,50

Peptide and Peptoid Arrays

One way to bypass the challenges of full-length protein expression is to display peptidesrather than proteins. Peptides may be synthe-sized in vitro as random peptides, peptide-likestructures (peptoids) or as peptides representingthe entire potential human ORFeome. Peptidesmay be also be generated in vivo using phage,E. coli, or yeast, cloned in fusion with a surfaceprotein, and displayed for biopanning.51e53

With the development of massively parallelpeptide synthesis, tens of thousands of peptidescan now be synthesized, immobilized, and

DNA

Add cexpr

sys

FIGURE 2 Serologic detection ofantibodies using programmableprotein arrays. Left: full-lengthtagged cDNAs are printed on slides,and detected with picogreen. Middle:tagged antigens are expressed usingmammalian cell lysate, captured in situand protein expression is confirmedusing antitag antibodies. Right: arraysare probed with sera, and boundimmunoglobulin is detected.

screened with sera for antigenicity.54e56 Peptidearrays can also be generated in vitro using on-chip synthesis technology, similar to oligonucle-otide Affymetrix arrays.57 Peptides spanning theproteome are first identified bioinformatically.DNA sequences encoding these peptides aresynthesized on-chip similar to DNA oligonucle-otide arrays and expressed on the arrays usingin vitro T7-based expression systems. In compar-ison to peptide-printed arrays, longer peptidescan be generated by in vitro synthesis. As analternative, the synthesized DNAs encoding thepeptide libraries can be displayed on the surfaceof phage coat proteins for biopanning.

Onekey feature of antigen/peptidedisplay forantibody detection is that they lead to identifica-tion of the naturally occurring cognate antigen.This identification is critical for understandingthe pathogenesis of the disease. As a practicalalternative to developing biomarkers that arespecific for diseases, display of peptide mimo-topes or peptoids can be used to detect specificantibodies in sera of patients with the disease.Peptoids are similar to peptides but presentchemically synthesized diverse structuralepitopes for antibody selection. Peptoid microar-rays displaying approximately 15,000 peptoidshave been successfully used for the identificationof autoantibodies in Alzheimer’s disease.58

Although the initiating natural antigenic stim-ulus cannot be identified from the mimotopes,

- GST ControlCase

Add sera

Detect IgG

α

ell-freeessiontem

Page 9: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

PROTEOMICS METHODS FOR THE DETECTION OF AUTOANTIBODIES 371

the peptoids themselves can be developed as clin-ically useful probes for identifying selective auto-antibody biomarkers that correlate with disease.

Detection of Post-TranslationalModifications

Most proteins undergo PTMs that are criticalfor their biological functions. Aberrant modifica-tions occur in disease states that may elicithumoral immune responses. Due to the hightissue specificity of PTMs, these antibodies may

TABLE 3 Examples of Autoantibodies Against Post-Transl

Post-translationalmodifications Example protein(s)

Carbamylation Filaggrin

Citrullination Filaggrin/fibrin/vimentin/collagen/a-enolase

Acetylation DEK

Phosphorylation SR proteins

La/SSB

Protease cleavage U1-70K

Calreticulin

Pso27

Aspartyl/isoaspartylisomerization

snRNP

Methylation SmD1, SmD3

Sulfilimine bond Collage IV

Deamidation Transglutaminase

Oxidation LDL

b2 glycoprotein 1

Glycosylation MUC1

Annexin I

Mutation p53

Alternative splicing isoforms CML66

Lengsin

be very disease- and tissue- specific. Many anti-bodies against post-translationally modifiedproteins have been identified in different dis-eases36,59e76(Table 3). The majority of these anti-bodies have been detected using cellularfractionation technologies that rely on lysatesfrom disease cell lines or tissues that containnatively modified proteins. Modified proteinsor peptides from small sets of target proteins ofinterest can then be displayed usingmicroarrays.However, current technology cannot systemati-cally mimic PTM structure at the proteome level.

ationally Modified Proteins

Example disease(s) References

Rheumatoid arthritis 68

Rheumatoid arthritis 74

Juvenile idiopathic arthritis 67

Systemic lupus erythematosus 70

Systemic lupus erythematosus 73

Systemic lupus erythematosus 65

Cancer 63

Psoriasis 62

Systemic lupus erythematosus 66

Systemic lupus erythematosus 59

Goodpasteur’s disease 71

Celiac disease 60

Atherosclerosis 64

Antiphospholipid syndrome 61

Cancer 75

Cancer 36

Cancer 72

Cancer 76

Cancer 69

Page 10: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY372

ASSOCIATION OFAUTOANTIBODIES WITH DISEASE

STATES

Autoimmune Disorders

Thefirst autoantibodies identifiedwere associ-ated with autoimmune rheumatic diseases.21,77

Autoantibodies not only indicate the presenceof disease; in some instances, they are pathogeniccauses of inflammation. Even when the autoanti-bodies are not pathogenic, they may bebiomarkers for antigens that stimulate CD4þ

cellular immune responses. Traditional IFAmethods to detect antinuclear antibodies,together with ELISA or radioimmunoassays, areused for the clinical measurement of these anti-bodies. Several of the antibodies that have beenidentified in autoimmune disorders target post-translational modifications (Tables 3 and 4).Anticitrullinated antibodies (ACA) have beenidentified that are very specific to adult RA,and distinct antibodies to citrullinated proteinsmay be involved in the pathogenesis of thedisease. Clinical testing for ACA using cyclic-citrullinated-peptides detects RA sera witha sensitivity of up to 80% to 90% at 98%specificity.

Type 1 Diabetes

Type 1 diabetes (T1D) is an autoimmunedisease that is caused by autoimmune destruc-tion of insulin-secretion islet cells in early child-hood. Like many autoimmune diseases, bothgenetic and environmental factors are thoughtto play an important role in disease etiology.Several autoantibodies, including anti-GAD65and anti-IA2, have been identified in T1D. Usinggenomics, the ZnT8 antigen was recently identi-fied.78 The presence of these antibodies in seraindicates the transition from genetic risk toimmunological risk. Both the titer and thecombined frequency of these autoantibodies arepredictive of T1D development in high-risk

populations. The five year risk for T1Dapproaches 50% in subjects with detectable anti-bodies against all three antigens. Several clinicaltrials targeting these antigens are ongoing. High-throughput proteomics are being used to iden-tify novel T1D antigens. However, as the threeknown autoantibodies in T1D have excellentperformance as clinical biomarkers, the identifi-cation of new targets may have limited utilityfor disease diagnosis or risk prediction. Newlydiscovered antigens may still uncover mecha-nisms of the etiology of T1D for therapeuticdevelopment.

Neurologic Disorders

A number of neurologic disorders haveevidence of immune-mediated pathogenesis.Functional autoantibodies to the acetylcholinereceptor are pathogenic in myasthenia gravis(MG), as are calcium-channel autoantibodies inLambert-Eaton myasthenic syndrome. Paraneo-plastic neuropathies, primarily associated withsmall cell lung cancer and ovarian cancer,are mediated by cross-reactive autoantibodiestargeting neuronal proteins, such as anti-Hu/ANNA-1, anti-Ri/ANNA-2, and anti-Yo/PCA1.79 However, emerging evidencesuggests that other neurologic disorders, suchas multiple sclerosis, Alzheimer’s dementia,and Parkinson’s disease, may be mediated inpart by B-cell mediated autoimmunity. Multiplesclerosis (MS) is an autoimmune disease causedby autoreactive T cells targeting myelin sheaths.Recent studies have identified autoantibodiesthat catalyze site-specific destruction of myelinbasic protein in MS patients80 and cross-reactwith the LMP-1 protein of Epstein-Barr virus.81

Multiple other autoantibody targets, such asPGAM1,82 mtHSP70,37 and contactin-2,83 havebeen identified in the sera of patients with MSusing proteomic-based approaches. Alzheimer’sdementia is a progressive neurologic diseasemarked by accumulation of beta-amyloid andtau protein in the central nervous system.

Page 11: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

TABLE 4 Examples of Autoantibodies Identified using Proteomic Technologies

Antigen Sources Example antigens Example diseases References

cDNA expression

Phage plaque/display SSX-2PC326 and othersIFI16 and othersFive-phage peptide

MelanomaMoyamoya diseaseSjogren’s syndromeColorectal carcinoma

29283027

Phage display & microarray A panel of 22 peptidesUbiquilin 1

Prostate cancerLung

3335

Cell lysates

2D Western Annexin I and IImtHSP70

Lung cancerMultiple sclerosis

3637

Natural protein array LAMR1 Lung cancer 38

Recombinant proteins

Purified proteins RALBP1 and othersKelch-like protein 12 and othersRPS20 and othersPanel of antigensA panel of 10 antigens

Ovarian cancerPrimary biliary cirrhosisAutoimmune hepatitisRheumatoid arthritisAlzheimer’s disease

40394341,4442

In vitro expression mixtures Various Infectious disease(Burkholderia pseudomallei)

50

On-chip synthesized proteins A panel of 28 antigensPanel of antigens

Breast cancerankylosing spondylitis

4548

Synthetic peptides

Synthetic peptides/peptoidson arrays

A panel of peptidesSeveral peptoidsGlycopeptides derived frommucins

Alzheimer’s diseaseAlzheimer’s diseaseColorectal cancer

565855

Synthetic human peptidome &phage display

Various Paraneoplastic neurologicaldisorder

57

Microbial surface display HSP60Plasminogen binding protein(PBP)GRP78

AtherosclerosisAutoimmune pancreatitisProstate cancer

535152

ASSOCIATION OF AUTOANTIBODIES WITH DISEASE STATES 373

Although Alzheimer’s is not considered animmune-mediated disorder, autoantibodies topeptoid libraries have been specifically identi-fied in the sera of Alzheimer’s patients, whichmay be biomarkers for disease diagnosis.58

Cancer

Cancer is the second most common cause ofdeath in the United States. In 2012, there will bean estimated 1.6 million new cases of cancer andover 500,000 deaths (http://www.cancer.org),

Page 12: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY374

with a total cost over $200 billion.84 Although theestimated five-year overall survival after a cancerdiagnosis is 67%, there are few FDA-approvedbiomarkers for early detection and monitoringfor disease recurrence, which could have signifi-cant impact on clinical outcome. There is alsoa need for biomarkers that identify biologicsubtypes of cancer and to predict responses toa rising number of immune-targeted thera-peutics, such as ipilimumab85 and sipuleucel-T.86

There is growing scientific evidence thatimmune dysregulation functions the pathogen-esis of cancer paradoxically, both contributingto carcinogenesis through chronic inflamma-tion87 as well as controlling cancer progressionwith direct immune surveillance of tumor anti-gens.88e90 Production of cytokines such as IL-6and CSF-1 by tumor cells promotes thedevelopment of immunosuppressive tumor-associated macrophages (TAMs).91,92 In modelsystems, TAMs have been shown to directlypromote tumor progression and metastasis.93e96

In contrast, tumor-infiltrating lymphocytes(TILs) are associated with improved survivalfor lymphoma,97 colorectal,98,99 breast,100 esoph-ageal,89 and ovarian cancers.101 In particular, Bcell signatures15,16 and intratumoral B cell folli-cles102 have been identified in multiple can-cers103e105 and are associated with improvedclinical outcome.106 These data suggest thatcancers can induce an active, specific antibodyresponse to tumor-derived antigens.

There have been multiple proteomics-basedapproaches to identify autoantibody biomarkersthat distinguish sera from cancer patients andsera from healthy controls (Table 4). Specificautoantibody signatures have been identifiedin the sera of patients from multiple cancertypes, often corresponding to overexpressedtumor antigens. These autoantibodies are beingdeveloped as potential biomarkers for earlycancer diagnosis, such as Annexin 1, 14-3-3theta, and LAMR1 in lung cancer,38 TA90 inmelanoma,107 and p53 in various cancers.108

Although promising, these biomarkers awaitvalidation in blinded, multicenter phase IIIbiomarker trials.

CHALLENGES AND FUTUREDEVELOPMENT

Over the past decades, proteomic technolo-gies have greatly contributed to the discoveryof novel autoantibody biomarkers. Emergingmethods enable the discovery of autoantigensat the scale of proteomes. To this end, the fieldof proteomics has expanded our understandingof the repertoire of antigens that can elicit auto-antibodies in different diseases. Emerging datafrom genomics and transcriptomics continuesto broaden the scope of potential immunogenicstructures to include mutated antigens andsplice variants that may have significant disease-and tissue specificity. In addition, proteomicmethods that can systematically display PTMsare likely to identify numerous other autoanti-body targets that are associated with disease.The discovery and validation of these antigenswill improve our knowledge of the structuralcontent that elicits antibody immune responses,and to understand the underlying mechanismsof autoimmunity. These advances have potentialclinical implications for both biomarker develop-ment and immune-targeted therapeutics.

References1. Ball J. Serum factor in rheumatoid arthritis aggluti-

nating sensitized sheep red cells. Lancet 1950;2(6637):520e4.

2. Hargraves MM, Richmond H, Morton R. Presentationof two bone marrow elements; the tart cell and theL.E. cell. Proc Staff Meet Mayo Clin 1948;23(2):25e8.

3. Waaler E. On the occurrence of a factor in humanserum activating the specific agglutintion of sheepblood corpuscles. Acta Path Micro Im A 2007;115(5):422e38.

4. Lachmann PJ. A two-stage indirect L.E. cell test.Immunology 1961;4:142e52.

Page 13: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

REFERENCES 375

5. Holborow EJ, Weir DM, Johnson GD. A serum factorin lupus erythematosus with affinity for tissue nuclei.BMJ 1957;2(5047):732e4.

6. Holman HR, Kunkel HG. Affinity between the lupuserythematosus serum factor and cell nuclei andnucleoprotein. Science 1957;126(3265):162e3.

7. Fernandez-Madrid F, Mattioli M. Antinuclear anti-bodies (ANA): immunologic and clinical significance.Semin Arthritis Rheum 1976;6(2):83e124.

8. Tan EM. Autoantibodies to nuclear antigens (ANA):their immunobiology and medicine. Adv Immunol1982;33:167e240.

9. Friou GJ, Finch SC, Detre KD. Interaction of nuclei andglobulin from lupus erythematosis serum demon-strated with fluorescent antibody. J Immunol 1958;80(4):324e9.

10. Notsu K, Note S, Nabeya N, Kuno S, Sakurami T.Antinuclear antibodies in childhood diabetics. Endo-crinol Jpn 1983;30(4):469e73.

11. Lendrum R, Walker G, Cudworth AG, et al. Islet-cellantibodies in diabetes mellitus. Lancet 1976;2(7998):1273e6.

12. Lendrum R, Walker G, Gamble DR. Islet-cell anti-bodies in juvenile diabetes mellitus of recent onset.Lancet 1975;1(7912):880e2.

13. BottazzoGF, Florin-ChristensenA,DoniachD. Islet-cellantibodies in diabetes mellitus with autoimmune poly-endocrine deficiencies. Lancet 1974;2(7892):1279e83.

14. Mattioli M, Reichlin M. Heterogeneity of RNA proteinantigens reactive with sera of patients with systemiclupus erythematosus. Description of a cytoplasmicnonribosomalantigen.Arthritis Rheum1974;17(4):421e9.

15. Sharp GC, Irvin WS, May CM, et al. Association ofantibodies to ribonucleoprotein and Sm antigens withmixed connective-tissue disease, systematic lupuserythematosus and other rheumatic diseases. N Engl JMed 1976;295(21):1149e54.

16. Clark G, Reichlin M, Tomasi Jr TB. Characterization ofa soluble cytoplasmic antigen reactive with sera frompatients with systemic lupus erythematosus. J Immu-nol 1969;102(1):117e22.

17. Herrera-Esparza R, Provost TT, Diaz LA. Molecularcharacterization of Ro(SSA) and La(SSB) proteins.J Rheumatol 1986;13(2):327e30.

18. Baekkeskov S, Landin M, Kristensen JK, et al. Anti-bodies to a 64,000 Mr human islet cell antigen precedethe clinical onset of insulin-dependent diabetes. J ClinInvest 1987;79(3):926e34.

19. Baekkeskov S, Nielsen JH, Marner B, Bilde T,Ludvigsson J, Lernmark A. Autoantibodies in newlydiagnosed diabetic children immunoprecipitatehuman pancreatic islet cell proteins. Nature 1982;298(5870):167e9.

20. Christie MR, Hollands JA, Brown TJ, Michelsen BK,Delovitch TL. Detection of pancreatic islet 64,000 M(r)autoantigens in insulin-dependentdiabetesdistinct fromglutamate decarboxylase. J Clin Invest 1993;92(1):240e8.

21. Routsias JG, Tzioufas AG, Moutsopoulos HM. Theclinical value of intracellular autoantigens B-cellepitopes in systemic rheumatic diseases. Clin ChimActa 2004;340(1-2):1e25.

22. Ben-Mahrez K, Sorokine I, Thierry D, et al. Circulatingantibodies against c-myc oncogene product in sera ofcolorectal cancer patients. Int J Cancer 1990;46(1):35e8.

23. Takahashi M, Chen W, Byrd DR, et al. Antibody to rasproteins in patients with colon cancer. Clin Cancer Res1995;1(10):1071e7.

24. Disis ML, Pupa SM, Gralow JR, Dittadi R, Menard S,Cheever MA. High-titer HER-2/neu protein-specificantibody can be detected in patients with early-stagebreast cancer. J Clin Oncol 1997;15(11):3363e7.

25. Sahin U, Tureci O, Schmitt H, et al. Human neoplasmselicit multiple specific immune responses in theautologous host. Proc Natl Acad Sci USA 1995;92(25):11810e3.

26. Krebs P, Kurrer M, Sahin U, Tureci O, Ludewig B.Autoimmunity seen through the SEREX-scope.Autoimmun Rev 2003;2(6):339e45.

27. Chang W, Wu L, Cao F, et al. Development of auto-antibody signatures as biomarkers for early detectionof colorectal carcinoma. Clin Cancer Res 2011;17(17):5715e24.

28. Kim J, Kim SK, Wang KC, Kim HY, Jeoung D. SEREXidentification of the autoantibodies that are prevalentin the cerebrospinal fluid of patients with moyamoyadisease. Biotechnol Lett 2004;26(7):585e8.

29. Tureci O, Sahin U, Schobert I, et al. The SSX-2 gene,which is involved in the t(X;18) translocation of syno-vial sarcomas, codes for the human tumor antigenHOM-MEL-40. Cancer Res 1996;56(20):4766e72.

30. Uchida K, Akita Y, Matsuo K, et al. Identification ofspecific autoantigens in Sjogren’s syndrome bySEREX. Immunology 2005;116(1):53e63.

31. Chen YT, Gure AO, Tsang S, et al. Identification ofmultiple cancer/testis antigens by allogeneic antibodyscreening of a melanoma cell line library. Proc NatlAcad Sci USA 1998;95(12):6919e23.

32. WeinerH,FaupelT,BussowK.Proteinarrays fromcDNAexpression libraries.Methods Mol Biol 2004;264:1e13.

33. Wang X, Yu J, Sreekumar A, et al. Autoantibody signa-tures in prostate cancer. N Engl J Med 2005;353(12):1224e35.

34. Chatterjee M, Mohapatra S, Ionan A, et al. Diagnosticmarkers of ovarian cancer by high-throughput antigencloning and detection on arrays. Cancer Res 2006;66(2):1181e90.

Page 14: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY376

35. Chen G, Wang X, Yu J, et al. Autoantibody profilesreveal ubiquilin 1 as a humoral immune responsetarget in lung adenocarcinoma. Cancer Res 2007;67(7):3461e7.

36. Brichory FM, Misek DE, Yim AM, et al. An immuneresponse manifested by the common occurrence ofannexins I and II autoantibodies and high circulatinglevels of IL-6 in lung cancer. Proc Natl Acad Sci USA2001;98(17):9824e9.

37. Sakurai T, Kimura A, Yamada M, et al. Identificationof antibodies as biological markers in serum frommultiple sclerosis patients by immunoproteomicapproach. J Neuroimmunol 2011;233(1-2):175e80.

38. Qiu J, Choi G, Li L, et al. Occurrence of autoantibodiesto annexin I, 14-3-3 theta and LAMR1 in prediagnosticlung cancer sera. J Clin Oncol 2008;26(31):5060e6.

39. Hu CJ, Song G, Huang W, et al. Identification of newautoantigens for primary biliary cirrhosis usinghuman proteome microarrays. Mol Cell Proteomics2012;11(9):669e80.

40. Hudson ME, Pozdnyakova I, Haines K, Mor G,Snyder M. Identification of differentially expressedproteins in ovarian cancer using high-density proteinmicroarrays. Proc Natl Acad Sci USA 2007;104(44):17494e9.

41. Hueber W, Kidd BA, Tomooka BH, et al. Antigenmicroarray profiling of autoantibodies in rheumatoidarthritis. Arthritis Rheum 2005;52(9):2645e55.

42. Nagele E, Han M, Demarshall C, Belinka B, Nagele R.Diagnosis of Alzheimer’s disease based on disease-specific autoantibody profiles in human sera. PLoSONE 2011;6(8):e23112.

43. Song Q, Liu G, Hu S, et al. Novel autoimmunehepatitis-specific autoantigens identified using proteinmicroarray technology. J Proteome Res 2010;9(1):30e9.

44. Chandra PE, Sokolove J, Hipp BG, et al. Novelmultiplex technology for diagnostic characterizationof rheumatoid arthritis. Arthritis Res Ther 2011;13(3):R102.

45. Anderson KS, Sibani S, Wallstrom G, et al. Proteinmicroarray signature of autoantibody biomarkers forthe early detection of breast cancer. J Proteome Res2011;10(1):85e96.

46. Ramachandran N, Hainsworth E, Bhullar B, et al. Self-assembling proteinmicroarrays. Science 2004;305(5680):86e90.

47. Ramachandran N, Raphael JV, Hainsworth E, et al.Next-generation high-density self-assembling func-tional protein arrays. Nat Methods 2008;5(6):535e8.

48. Wright C, Sibani S, Trudgian D, et al. Detection ofmultiple autoantibodies in patients with ankylosingspondylitis using nucleic acid programmable proteinarrays. Mol Cell Proteomics 2012;11(2):M9.00384.

49. Davies DH, Liang X, Hernandez JE, et al. Profiling thehumoral immune response to infection by using pro-teome microarrays: high-throughput vaccine anddiagnostic antigen discovery. Proc Natl Acad Sci USA2005;102(3):547e52.

50. Felgner PL, Kayala MA, Vigil A, et al. A Burkholderiapseudomallei protein microarray reveals serodiag-nostic and cross-reactive antigens. Proc Natl Acad SciUSA 2009;106(32):13499e504.

51. Frulloni L, Lunardi C, Simone R, et al. Identificationof a novel antibody associated with autoimmunepancreatitis. N Engl J Med 2009;361(22):2135e42.

52. Mintz PJ, Kim J, Do KA, et al. Fingerprinting thecirculating repertoire of antibodies from cancerpatients. Nat Biotechnol 2003;21(1):57e63.

53. Perschinka H, Wellenzohn B, Parson W, et al. Identi-fication of atherosclerosis-associated conformationalheat shock protein 60 epitopes by phage display andstructural alignment. Atherosclerosis 2007;194(1):79e87.

54. Halperin RF, Stafford P, Johnston SA. Exploring anti-body recognition of sequence space through random-sequence peptide microarrays. Mol Cell Proteomics2011;10(3):M110.000786.

55. Pedersen JW,BlixtO,Bennett EP, et al. Seromicprofilingof colorectal cancer patients with novel glycopeptidemicroarray. Int J Cancer 2011;128(8):1860e71.

56. Restrepo L, Stafford P, Magee DM, Johnston SA.Application of immunosignatures to the assessment ofAlzheimer’s disease. Ann Neurol 2011;70(2):286e95.

57. Larman HB, Zhao Z, Laserson U, et al. Autoantigendiscovery with a synthetic human peptidome. NatBiotechnol 2011;29(6):535e41.

58. Reddy MM, Wilson R, Wilson J, et al. Identification ofcandidate IgG biomarkers for Alzheimer’s disease viacombinatorial library screening. Cell 2011;144(1):132e42.

59. Brahms H, Raymackers J, Union A, de Keyser F,Meheus L, Luhrmann R. The C-terminal RG dipeptiderepeats of the spliceosomal Sm proteins D1 and D3contain symmetrical dimethylarginines, which forma major B-cell epitope for anti-Sm autoantibodies.J Biol Chem 2000;275(22):17122e9.

60. Fleckenstein B, Molberg O, Qiao SW, et al. Gliadin Tcell epitope selection by tissue transglutaminase inceliac disease. Role of enzyme specificity and pHinfluence on the transamidation versus deamidationprocess. J Biol Chem 2002;277(37):34109e16.

61. Ioannou Y, Zhang JY, Qi M, et al. Novel assays ofthrombogenic pathogenicity in the antiphospholipidsyndrome based on the detection of molecular oxida-tive modification of the major autoantigen beta2-glycoprotein I. Arthritis Rheum 2011;63(9):2774e82.

Page 15: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

REFERENCES 377

62. Iversen OJ, Lysvand H, Hagen L. The autoantigen Psop27: a post-translational modification of SCCA mole-cules. Autoimmunity 2011;44(3):229e34.

63. Le Naour F, Brichory F, Misek DE, Brechot C,Hanash SM, Beretta L. A distinct repertoire of auto-antibodies in hepatocellular carcinoma identified byproteomic analysis. Mol Cell Proteomics 2002;1(3):197e203.

64. Maggi E, Finardi G, Poli M, et al. Specificity of auto-antibodies against oxidized LDL as an additionalmarker for atherosclerotic risk. Coron Artery Dis 1993;4(12):1119e22.

65. Malmegrim KC, Pruijn GJ, van Venrooij WJ. The fateof the U1 snRNP autoantigen during apoptosis:implications for systemic autoimmunity. Isr Med AssocJ 2002;4(9):706e12.

66. Mamula MJ, Gee RJ, Elliott JI, et al. Isoaspartyl post-translational modification triggers autoimmuneresponses to self-proteins. J Biol Chem 1999;274(32):22321e7.

67. Mor-Vaknin N, Kappes F, Dick AE, et al. DEK in thesynovium of patients with juvenile idiopathic arthritis:characterization of DEK antibodies and post-translational modification of the DEK autoantigen.Arthritis Rheum 2011;63(2):556e67.

68. Mydel P, Wang Z, Brisslert M, et al. Carbamylation-dependent activation of T cells: a novel mechanism inthe pathogenesis of autoimmune arthritis. J Immunol2010;184(12):6882e90.

69. Nakatsugawa M, Hirohashi Y, Torigoe T, et al. Novelspliced form of a lens protein as a novel lung cancerantigen, Lengsin splicing variant 4. Cancer Sci 2009;100(8):1485e93.

70. Neugebauer KM, Merrill JT, Wener MH, Lahita RG,Roth MB. SR proteins are autoantigens in patientswith systemic lupus erythematosus. Importance ofphosphoepitopes. Arthritis Rheum 2000;43(8):1768e78.

71. Pedchenko V, Vanacore R, Hudson B. Goodpasture’sdisease: molecular architecture of the autoantigenprovides clues to etiology and pathogenesis. Curr OpinNephrol Hypertens 2011;20(3):290e6.

72. Soussi T. p53 Antibodies in the sera of patients withvarious types of cancer: a review. Cancer Res 2000;60(7):1777e88.

73. Terzoglou AG, Routsias JG, Avrameas S,Moutsopoulos HM, Tzioufas AG. Preferential recog-nition of the phosphorylated major linear B-cellepitope of La/SSB 349-368 aa by anti-La/SSB auto-antibodies from patients with systemic autoimmunediseases. Clin Exp Immunol 2006;144(3):432e9.

74. van Venrooij WJ, van Beers JJ, Pruijn GJ. Anti-CCPantibodies: the past, the present and the future. NatRev Rheumatology 2011;7(7):391e8.

75. Wandall HH, Blixt O, Tarp MA, et al. Cancer bio-markers defined by autoantibody signatures to aberrantO-glycopeptide epitopes.CancerRes 2010;70(4):1306e13.

76. Yan Y, Phan L, Yang F, et al. A novel mechanism ofalternative promoter and splicing regulates theepitope generation of tumor antigen CML66-L.J Immunol 2004;172(1):651e60.

77. Lernmark A. Autoimmune diseases: are markersready for prediction? J Clin Invest 2001;108(8):1091e6.

78. Wenzlau JM, Juhl K, Yu L, et al. The cation effluxtransporter ZnT8 (Slc30A8) is a major autoantigen inhuman type 1 diabetes. Proc Natl Acad Sci USA 2007;104(43):17040e5.

79. Koike H, Tanaka F, Sobue G. Paraneoplastic neurop-athy: wide-ranging clinicopathological manifestations.Curr Opin Neurol 2011;24(5):504e10.

80. Ponomarenko NA, Durova OM, Vorobiev II , et al.Autoantibodies to myelin basic protein catalyze site-specific degradation of their antigen. Proc Natl AcadSci USA 2006;103(2):281e6.

81. Gabibov AG, Belogurov Jr AA, Lomakin YA, et al.Combinatorial antibody library from multiple scle-rosis patients reveals antibodies that cross-react withmyelin basic protein and EBV antigen. FASEB J 2011;25(12):4211e21.

82. Kimura A, Sakurai T, Koumura A, et al. High preva-lence of autoantibodies against phosphoglyceratemutase 1 in patients with autoimmune central nervoussystem diseases. J Neuroimmunol 2010;219(1-2):105e8.

83. Derfuss T, Parikh K, Velhin S, et al. Contactin-2/TAG-1-directed autoimmunity is identified in multiplesclerosispatients andmediates graymatterpathology inanimals. Proc Natl Acad Sci USA 2009;106(20):8302e7.

84. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ.Cancer statistics, 2009. CA Cancer J Clin 2009;59(4):225e49.

85. Sondak VK, Smalley KS, Kudchadkar R, Grippon S,Kirkpatrick P. Ipilimumab. Nat Rev Drug Discov 2011;10(6):411e2.

86. Higano CS, Small EJ, Schellhammer P, et al.Sipuleucel-T. Nat Rev Drug Discov 2010;9(7):513e4.

87. Grivennikov SI, Greten FR, Karin M. Immunity,inflammation, and cancer. Cell 2010;140(6):883e99.

88. Bei R, Masuelli L, Palumbo C, Modesti M, Modesti A.A common repertoire of autoantibodies is shared bycancer and autoimmune disease patients: Inflamma-tion in their induction and impact on tumor growth.Cancer Lett 2009;281(1):8e23.

89. Rauser S, Langer R, Tschernitz S, et al. High numberof CD45ROþ tumor infiltrating lymphocytes is anindependent prognostic factor in non-metastasized(stage IeIIA) esophageal adenocarcinoma. BMCCancer 2010;10:608.

Page 16: Proteomic and Metabolomic Approaches to Biomarker Discovery || Autoantibodies and Biomarker Discovery

23. AUTOANTIBODIES AND BIOMARKER DISCOVERY378

90. Pages F, Galon J, Dieu-Nosjean MC, Tartour E, Sautes-Fridman C, Fridman WH. Immune infiltration inhuman tumors: a prognostic factor that should not beignored. Oncogene 2010;29(8):1093e102.

91. Menetrier-Caux C, Montmain G, Dieu MC, et al.Inhibition of the differentiation of dendritic cells fromCD34(þ) progenitors by tumor cells: role ofinterleukin-6 and macrophage colony-stimulatingfactor. Blood 1998;92(12):4778e91.

92. Dranoff G. Cytokines in cancer pathogenesis andcancer therapy. Nat Rev Cancer 2004;4(1):11e22.

93. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression ofmammarytumors to malignancy. J Exp Med 2001;193(6):727e40.

94. Pollard JW. Tumour-educated macrophages promotetumour progression and metastasis. Nat Rev Cancer2004;4(1):71e8.

95. Zou W. Immunosuppressive networks in the tumourenvironment and their therapeutic relevance. Nat RevCancer 2005;5(4):263e74.

96. Zou W. Regulatory T cells, tumour immunity andimmunotherapy. Nat Rev Immunol 2006;6(4):295e307.

97. Dave SS, Wright G, Tan B, et al. Prediction of survivalin follicular lymphoma based on molecular features oftumor-infiltrating immune cells. N Engl J Med 2004;351(21):2159e69.

98. Galon J, Costes A, Sanchez-Cabo F, et al. Type,density, and location of immune cells within humancolorectal tumors predict clinical outcome. Science2006;313(5795):1960e4.

99. Pages F, Berger A, Camus M, et al. Effector memory Tcells, early metastasis, and survival in colorectalcancer. N Engl J Med 2005;353(25):2654e66.

100. Schmidt M, Bohm D, von Torne C, et al. The humoralimmune system has a key prognostic impact in node-

negative breast cancer. Cancer Res 2008;68(13):5405e13.

101. Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intra-tumoral T cells, recurrence, and survival in epithelialovarian cancer. N Engl J Med 2003;348(3):203e13.

102. Dieu-Nosjean MC, Antoine M, Danel C, et al. Long-term survival for patients with non-small-cell lungcancer with intratumoral lymphoid structures. J ClinOncol 2008;26(27):4410e7.

103. Milne K, Kobel M, Kalloger SE, et al. Systematicanalysis of immune infiltrates in high-grade serousovarian cancer reveals CD20, FoxP3 and TIA-1 aspositive prognostic factors. PLoS ONE 2009;4(7):e6412.

104. Nelson BH. CD20þ B cells: the other tumor-infiltratinglymphocytes. J Immunol 2010;185(9):4977e82.

105. Willis SN, Mallozzi SS, Rodig SJ, et al. The microen-vironment of germ cell tumors harbors a prominentantigen-driven humoral response. J Immunol 2009;182(5):3310e7.

106. Bianchini G, Qi Y, Alvarez RH, et al. Molecularanatomy of breast cancer stroma and its prognosticvalue in estrogen receptor-positive and -negativecancers. J Clin Oncol 2010;28(28):4316e23.

107. Kelley MC, Gupta RK, Hsueh EC, Yee R, Stern S,Morton DL. Tumor-associated antigen TA90 immunecomplex assay predicts recurrence and survival aftersurgical treatment of stage IeIII melanoma. J ClinOncol 2001;19(4):1176e82.

108. Anderson KS, Wong J, Vitonis A, et al. p53 autoanti-bodies as potential detection and prognosticbiomarkers in serous ovarian cancer. Cancer EpidemiolBiomarkers Prev 2010;19(3):859e68.

109. Anderson KS, LaBaer J. The sentinel within: exploitingthe immune system for cancer biomarkers. J ProteomeRes 2005;4(4):1123e33.