Next Article in Journal
Co-Calibrating Physical and Psychological Outcomes and Consumer Wearable Activity Outcomes in Older Adults: An Evaluation of the coQoL Method
Next Article in Special Issue
From Proteomics to Personalized Medicine: The Importance of Isoflavone Dose and Estrogen Receptor Status in Breast Cancer Cells
Previous Article in Journal
Altered Interoceptive Perception and the Effects of Interoceptive Analgesia in Musculoskeletal, Primary, and Neuropathic Chronic Pain Conditions
Previous Article in Special Issue
Effects of Growth Hormone Treatment and Rehabilitation in Incomplete Chronic Traumatic Spinal Cord Injury: Insight from Proteome Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Contribution of Multiplex Immunoassays to Rheumatoid Arthritis Management: From Biomarker Discovery to Personalized Medicine

by
Carlos M. Laborde
1,
Patricia Castro-Santos
2 and
Roberto Díaz-Peña
2,*
1
Dreamgenics, Inc., 33010 Oviedo, Spain
2
Faculty of Health Sciences, Universidad Autónoma de Chile, Talca 3460000, Chile
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2020, 10(4), 202; https://doi.org/10.3390/jpm10040202
Submission received: 7 October 2020 / Revised: 27 October 2020 / Accepted: 28 October 2020 / Published: 30 October 2020
(This article belongs to the Special Issue Proteomics and Personalized Medicine)

Abstract

:
Rheumatoid arthritis (RA) is a multifactorial, inflammatory and progressive autoimmune disease that affects approximately 1% of the population worldwide. RA primarily involves the joints and causes local inflammation and cartilage destruction. Immediate and effective therapies are crucial to control inflammation and prevent deterioration, functional disability and unfavourable progression in RA patients. Thus, early diagnosis is critical to prevent joint damage and physical disability, increasing the chance of achieving remission. A large number of biomarkers have been investigated in RA, although only a few have made it through the discovery and validation phases and reached the clinic. The single biomarker approach mostly used in clinical laboratories is not sufficiently accurate due to its low sensitivity and specificity. Multiplex immunoassays could provide a more complete picture of the disease and the pathways involved. In this review, we discuss the latest proposed protein biomarkers and the advantages of using protein panels for the clinical management of RA. Simultaneous analysis of multiple proteins could yield biomarker signatures of RA subtypes to enable patients to benefit from personalized medicine.

1. Introduction

Rheumatoid arthritis (RA) is a multifactorial, progressive, systemic inflammatory autoimmune disease that affects approximately 1% of the population worldwide and primarily involves the joints, resulting in local inflammation and cartilage destruction [1]. Due to long-term disease complications, RA is associated with reduced life expectancy and constitutes a major cause of chronic incapacity.
Early RA diagnosis is essential for the improvement of prognosis and quality of life. In 2010, the American College of Rheumatology (ACR) and the European League Against Rheumatism (EULAR) established new RA classification criteria [2], which have been widely accepted by rheumatologists, that focus on features at an earlier disease stage. In addition to clinical criteria, they include two immunological markers: anti-citrullinated protein autoantibodies (ACPA) and rheumatoid factor (RF). RA can be classified as one of two forms: “early RA”, which affects patients in the first few years after clinical diagnosis, and “established RA”, which affects all RA patients with a disease duration greater than 1–2 years irrespective of the presence of clinical or radiological joint damage; however, there is no consensus as to the denomination of forms of RA [3]. The reality is that RA symptoms develop gradually. In the “preclinical phase” of development, there are relevant changes in the biomarkers ACPA and RF and another biomarker, anti-carbamylated protein antibodies (anti-CarP) [4]. The great interest in ACPA and RF stems from the fact that their expression may precede the clinical appearance of RA by 10 years or more, allowing for improved predictive accuracy when both ACPAs and RF are used simultaneously [5]. However, while progress has been made regarding the diagnosis of early RA, there is still a need for novel biomarkers to improve RA diagnosis. Treatment initiated at an early stage of the disease is more likely to be successful. Once RA is established, generally there will be damage to the joints and the occurrence of diverse comorbidities with complications as a consequence of chronic inflammation [6]. Early diagnosis and treatment prevent radiologic progression of RA and result in a higher rate of remission than delayed treatment [7,8]. The combination of multiple biomarkers improves the ability to represent complex pathophysiological conditions, which is a prerequisite for personalized treatment. In this review, we discuss the latest proposed biomarkers and the advantages of using panels of protein biomarkers in the clinical management of RA.

2. Rheumatoid Arthritis: A Complex Multifactorial Disease

A diagnostic phenotype of RA does not exist. RA is a disease in which the typical signs, such as pain and fatigue, occur before the onset of joint inflammation [9] and comorbidities may develop over time. The consequence is a heterogeneous disease with a variable clinical spectrum, although RA has been considered a relatively homogeneous clinical syndrome for more than 50 years. Like most rheumatic diseases, RA is a multifactorial entity in which genetic variability, environmental factors and random events interact to trigger activation of pathological pathways [10].

2.1. Genetic Factors

Rheumatoid arthritis-related autoimmunity initially develops years prior to the first joint symptoms in the genetically predisposed population as a result of a complex process that has not been elucidated for the most part. Moreover, these processes trigger different mechanisms that may be responsible for the different symptoms. This finding suggests that the mechanisms may differ between different genetic and/or immunologic subgroups of RA. Genetic susceptibility to RA is evident in familial clustering and monozygotic twin studies, and the heritability of RA has been estimated to be approximately 60% [11]. An improved understanding of the genetic basis of RA is required to develop a more personalized approach for disease treatment. The human “major histocompatibility complex, MHC” (HLA) region, located on chromosome 6, is by far the largest source of genetic susceptibility to RA. The region consists of genes encoding molecules that play a key role in the immune system by modulating responses to invading pathogens. Approximately one-third of genetic risk is associated with the HLA locus [11], specifically with the HLA-DRB1 shared epitope (SE), which encodes a common amino acid sequence associated with RA susceptibility and progression (70QRRAA74, 70QKRAA74, or 70RRRAA74) [12]. HLA-DR loci encode HLA class II molecules (DR, DQ and DP) that function as receptors for processed peptides derived predominantly from membrane and extracellular proteins. HLA-DR expression occurs on the surface of antigen-presenting cells (APCs) (including dendritic cells, macrophages and B cells) and is essential for displaying peptides to CD4+ T cells, inducing their activation. The presence of an SE suggests that the HLA alleles bind the same antigen, suggesting the existence of an arthritogenic self-peptide or molecular mimic of foreign antigens that shapes the T-cell-antigen repertoire. However, this arthritogenic peptide has not yet been identified.
The pathogenesis of RA has a polygenic basis, and genetic variation can be explained by RA risk alleles within the non-HLA locus. In the last few years, our understanding of the genetic susceptibility and pathogenic mechanisms underlying RA has been greatly improved by the increase in the power of genome-wide association studies (GWAS). These studies have uncovered over 100 genetic loci associated with an increased risk of RA, establishing 98 genes that could potentially contribute to the onset of RA [13]. HLA and some non-HLA genes have been linked to the development of antibodies against citrullinated proteins, allowing differentiation between ACPA-seropositive and ACPA-seronegative RA [13,14]. However, analyses of ACPA-negative RA over the years have been challenging. Indeed, it has been suggested that seronegative RA may be heterogeneous with regard to genetics and related pathogenic mechanisms [15].

2.2. Environmental Factors

There are other factors that provide additional complexity and need to be considered. In Table 1, we show the most important of these factors. Smoking is the most common environmental risk factor for RA, increasing the risk of disease by 1.3- and 2.0-fold for women and men, respectively [16]. This finding has led to a model of pathogenesis in which smoking exposure may lead to the presence of citrullinated proteins in the lungs, which is followed by the development of ACPAs in susceptible individuals [17]. Considering that it likely reflects the action of environmental exposure, the presence of ACPA in an unaffected individual is clearly a risk factor for the future development of RA. Exposure to air pollution has been investigated, providing variable results [18]. There seems to be an increased risk of RA development in individuals living close to roads with heavy traffic [19]. Additionally, occupational exposure to mineral oils and silica has been associated with an increased risk of developing RA [20,21].
Although the disease can develop at any age, the typical clinical presentation of RA first occurs between 20–40 years of age, affecting women more frequently than men at a 2–3:1 ratio [1]. A hormonal influence on the onset of the disease in women has been suggested. In fact, pregnancy is in itself a known risk factor for the development of RA [22,23]. However, the mechanism by which gender influences susceptibility to RA remains unclear. Additionally, the role of diet and exercise requires more study before the proposal of relevant recommendations [18]. What seems clear is that most studies have demonstrated an increased risk of RA in individuals with obesity [24]. In addition, studies of bacterial communities in the intestine and the lung have revealed the microbiome as an emerging area of interest in RA [25,26], but no distinct features of the gut or lung microbiomes have been observed prior to the development of RA so far. With regard to bacterial infections and the risk of RA, more tangible results have been obtained for periodontitis-associated bacteria [27]. Other potential risk factors include several physical and psychosocial aspects [28], the most prominent of which is socio-economic status [29].

3. Serum Biomarkers in Rheumatoid Arthritis

Early diagnosis and immediate, effective therapy are crucial to gain control of inflammation and prevent deterioration, functional disability and unfavourable progression in RA patients. Much of the current pharmacotherapy is implemented on a “trial-and-error” basis [30]. All of the RA-associated symptoms, in addition to having serious medical consequences for the individual patient, also economically affect the healthcare system and society as a whole. To carry out personalized medicine for RA, clinical practice requires the use of biomarkers to ensure early diagnosis, accurate stratification and the high efficacy of treatment. A very large number of biomarkers have been investigated in RA, although only a few have made it past the discovery and validation phase into clinical use [31]. RF and ACPA are currently used as biomarkers for diagnostics and can predominantly be detected in the serum and synovial fluid (SF) of RA patients [32] (Table 2). RF has moderate specificity because it can also be found in other connective tissue diseases and in some chronic infectious diseases [33]. ACPA is more specific, and the presence of ACPA is an independent risk factor for developing RA in patients with undifferentiated arthritis [34]. However, in reality, RA constitutes two clinical RA subgroups, ACPA-positive and ACPA-negative RA, that differ with respect to genetic background, predisposing environmental factors and clinical progression/remission [35]. The pathogenic significance of ACPA in RA may be the result of its multiple biological activities [36]. The production of ACPA disrupts immune tolerance and is dependent on the occurrence of both genetic and environmental factors. In a hypothetical model, environmental factors could contribute to stimulating innate immunity. Apoptosis and/or necrosis of some cells could cause citrullination of certain proteins in the lungs (due to the increase in the activity of peptidyl-arginine deiminase (PAD) enzymes). Some of these modified proteins bind specifically to human leukocyte antigen (HLA)-DR molecules on antigen-presenting cells (APCs), resulting in high titres of ACPA. Citrullination of proteins in the joints due to infection, trauma, exercise, etc., could lead to immune complex formation between the modified proteins and ACPA, which could further bind to Fc receptors on the surface of synovial macrophages, contributing to the perpetuation of inflammation. The erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) are used as markers of inflammation and are significant indicators of disease severity [37]. However, these markers only reflect the level of inflammation, and their use is limited.
In Table 2, selected promising biomarkers are presented. Anti-CarP antibodies have been identified in both ACPA-positive and ACPA-negative RA patients in sera and are also linked to more severe disease [38]. Carbamylation is a post-translational modification induced by the presence of cyanate in which the enzyme lysine carbamyltransferase catalyses the carbamylation of a lysine residue into homocitrulline. Compared to ACPA and RF, anti-CarP antibodies show lower sensitivity for the diagnosis of RA [44]. However, a triple-positive autoantibody profile (RF, ACPAs and anti-CarP) provided promising results that might help to identify individuals at risk of developing RA [45]. Other antibodies of growing interest in RA target PAD enzymes. These enzymes catalyse the conversion of peptidyl-arginine into peptidyl-citrulline, a posttranslational modification known as citrullination. In particular, anti-PAD3 and PAD4 antibodies have emerged as significant participants in the pathogenesis of RA [46]. Ren J et al. reported a meta-analysis showing the high value of anti-PAD4 in the diagnosis of RA and the high specificity but relatively low sensitivity of anti-PAD4 [39]. Moreover, anti-PAD4 antibodies have been associated with severe erosive joint disease in RA [40]. Recently, anti-PAD2 has been described for the first time in RA [47]. Anti-PAD2 seems to be characteristic of a clinically distinct subtype of RA causing less severe baseline joint inflammation and slower joint disease progression [47]. Anti-mutated citrullinated vimentin (anti-MCV) antibodies have demonstrated comparable diagnostic value to RF and ACPA and might be an effective diagnostic marker for RA [41]. An association between anti-MCV levels and extra-articular manifestations of RA has been detected [48]. Moreover, anti-MCV seems to also be a predictive marker of the response to rituximab treatment [49]. Several antibody types have been identified only in certain subsets of RA and present a complex pattern with partial overlaps between the different antibodies. Antibodies to stress-related proteins [50] and modified type II collagens [51] and their role in RA have also been explored. Alterations in the glycosylation status of antibodies are associated with autoimmunity [52]. Interestingly, glycosylation of the Immunoglobulin G (IgG) ACPA V domain and its value as a predictive marker of RA development in ACPA-positive individuals have been reported [53].
Serum 14-3-3η protein has been considered a promising joint-derived biomarker for RA [42,54]. Combined with other serological measurements, detection of the 14-3-3η protein enhances the diagnosis of RA, especially in patients seronegative for RF and ACPA [42]. A positive 14-3-3η status has also been significantly associated with radiographic progression in early RA at years 1, 3 and 5, indicating the prognostic utility [55]. A recent meta-analysis demonstrated that serum and synovial calprotectin are correlated with disease activity in RA [43]. Furthermore, calprotectin plays a potential role as an independent predictor of radiological progression [56].
Proteomics research focuses on the discovery of novel biomarkers, which would not only provide an early diagnosis and allow us to understand the different pathological mechanisms underlying the heterogeneity of RA but also would allow to stratify patients, which is critical to enable effective treatments [57]. Patients show a huge variability in response to different treatments, and some of them are exposed to adverse effects. For example, although anti-tumour necrosis factor (TNF) α therapy is effective in the majority of cases, 30 to 40% of treated patients show an inadequate response [58]. Several approaches have shown the advantage of using proteomics to identify predictors of treatment response in RA [59], mainly with the TNF blockers etanercept and infliximab [60,61,62,63,64]. Biomarkers for prognosis of RA would help identifying the patients with the highest progression rates which will allow focusing on them with more aggressive treatments. It has been a long time, since Hueber et al. last described in 2005 an autoantibody signature with the potential to stratify patients by disease severity [65]. They analysed serum samples with antigen microarrays, showing potential to stratify RA patients with less disease severity. However, the search for predictors of prognosis and/or treatment response in RA patients remains an area of development. More efforts need to be made in order to discover new biomarkers, validate the findings and translate them into the clinical practice.

4. Multiplex Immunoassays: A Promising Path to Personalized Medicine

Proteomics dynamically study the set of proteins expressed by an organism at a given time and under certain specific conditions of time and environment [66,67]. This type of studies can be approached with different techniques, such as gel-based and mass spectrometry-based techniques, single-molecule sequencing and multiplexed immunoassays. All of them are complementary and valid techniques to study the proteome, with their particular advantages and disadvantages that condition their applicability. Despite all of them being equally interesting, in this review, we have focused on multiplexed immunoassays. We trust that they have the greatest potential for their application in clinical practice and will be the most interesting for readers.

4.1. Limitations of Current Proteomic Technologies

Proteomic research reveals hundreds of candidate proteins every day around the world. However, despite this large number of proteins, the number of candidate protein biomarkers that eventually reach clinical use remains very low. In some cases, proteins have either unknown or incompletely characterized clinical significance, while other proteins do not exhibit expression that is consistent or sufficient enough to warrant their use in clinical practice [68,69,70].
Any proposed biomarker should be of clinical interest. This implies that its role in the disease must be well established; it must be measurable in biological fluids; it must result in improved sensitivity and specificity compared to existing biomarkers, and its detection must be cost-effective. Plasma and urine are the most commonly used biological samples for laboratory tests. Only in special cases are extravascular fluids, such as cerebrospinal fluid or synovial fluid, used. Blood is the gold standard because it travels through almost every part of the human body, and therefore, serum/plasma biomarkers are particularly valuable for monitoring health status [71]. Apart from blood, urine is a very interesting sample type for studying secreted proteins, and it can be obtained by non-invasive techniques, which makes it especially attractive in some cases. Research projects that identify candidate proteins using other types of human biological samples (homogenized tissue, cell supernatants, etc.) must confirm two things: that the proteins are measurable in plasma or urine and that the same alterations previously observed between healthy controls and patients are corroborated in one of those two types of specimens.
The direct discovery of biomarkers in serum or plasma using proteomic techniques is not a simple task but a great challenge due to the complexity and high dynamic range of protein expression, which extends through eleven orders of magnitude, from albumin to cytokines [72]. Moreover, most candidate protein biomarkers are often present at very low concentrations and are often bound to other carrier proteins. In biological fluids, such as plasma and urine, high-abundance proteins are predominant and repress the signals of lower-abundance proteins, which then become undetectable by common proteomic techniques, such as two-dimensional gel electrophoresis and chromatography. Different commercial approaches have been developed predominantly to eliminate high-abundance serum proteins [73,74,75]. However, this required step involves altering the natural composition of the plasma and introduces a source of variability that can influence the validity of subsequent results. In addition, this step does not occur in clinical practice, so before considering the clinical application of candidate proteins, it is necessary to ensure that they can also be quantified directly in plasma using clinical analysers. The problem that may arise is that predominant, high-abundance serum proteins can significantly interfere with and influence the performance of the test as well as the quality of the result, resulting in the so-called serum matrix effect [76].
There is another problem derived from the limitations of proteomic techniques. Most of these techniques are essentially manual and time-consuming, unlike those used in clinical practice, where the entire analysis process is fast and automated. Many intermediate steps are necessary to study the low-abundance proteome (removing high-abundance proteins, using different buffers, etc.) and the higher the number of intermediate steps is, the larger the difference between the final sample and plasma is. In addition, since proteomic techniques are mainly manual, the final results depend greatly on the experience of the researcher, affecting the reproducibility of the results. This is unlike what occurs in clinical practice, where the analytical process is fully automated, and this guarantees very high reproducibility.
Overall, the limitations of proteomic techniques mean that the results must be carefully evaluated. The differences observed in the candidate proteins must be confirmed under clinical conditions. A complex analytical validation process to determine the sensitivity, specificity, linearity range, detection limit, etc., as well as the dependence of these parameters on the nature of the biological sample used and its integrity over time is also necessary [77]. Only if all of the drawbacks previously mentioned are successfully overcome will candidate protein biomarkers be able to be considered for clinical use.

4.2. The Challenge of Multiplex Immunoassays

Conventional multiplex immunoassays are conducted by incubating microbeads or a microarray with a biological sample, followed by the addition of a mixture of detection antibodies with the expectation that each detection antibody will bind to the target proteins bound to the corresponding capture antibody. While thermal agitation ensures that each reagent encounters its target, three major challenges affect the design and limit the number of proteins that can be used: antibody cross-reactivity, non-specific antibody binding and antibody interference [78] (Figure 1).
Cross-reactivity toward nontarget proteins is pervasive and widespread for antibodies [79,80]. It often occurs when proteins in the sample are structurally similar to the protein of interest. The antibody pair interacts with a protein other than the targeted protein. Cross-reactivity is arguably the biggest obstacle in establishing high-performance and large-scale multiplexed assays and requires that manufacturers carefully select and exhaustively test in-house-developed antibody pairs to ensure analyte specificity [81]. Nonspecific binding occurs when the antibody pair interacts with the sample container or other assay surfaces and contributes to the background. This interference can be reduced by blocking. Finally, antibody interference occurs when endogenous antibodies within samples cross-link with assay antibodies or substances within the sample and prevent proper binding of the target protein to both the capture and detection antibodies. In biological samples, such as serum and plasma, common antibody interfering substances include human anti-mouse antibodies (HAMA) and RF as well as any substance present at an exceptionally high concentration [82,83].
Interfering factors can be found in complex sample matrices and may also be introduced to biological samples via reagents. Interference can result in either a reduced or elevated signal relative to the actual concentration of the target protein, yielding untrustworthy data. Therefore, interfering factors may compromise assay performance and design. Many protein combinations of interest may not be possible because nonspecific binding produces a large background signal, reducing the sensitivity. Although the main advantage of multiplex immunoassays is the possibility of quantifying several proteins simultaneously in a single test, a proper balance must be achieved to provide acceptable sensitivity and an appropriate dynamic range for each of the multiplexed proteins [84]. Consequently, the challenge of scaling up assays and enhancing their sensitivity requires that cross-reactivity is adequately addressed and suppressed or at least mitigated.
The appropriate composition of diluents and assay buffers represents another key factor in multiplex immunoassays. The diluent refers to the solution that is used for serial dilution of the protein standard to establish a calibration curve as well as the dilution of samples at concentrations above the upper limit of quantification. Thus, the composition of the diluent is an important factor that can affect the reliability and accuracy of immunoassays [85]. Ideally, the diluent must be optimized to maximize specific binding and detection, minimize nonspecific binding and interference, and mimic the sample matrix for the dilution of calibrators and samples [86]. Assay buffers should interact effectively with all reagents and proteins included under normal assay conditions. However, differences in hydrophobicity, electrical charge and post-translational modifications lead to the requirement for specific reaction conditions for each protein. Small changes in buffer pH or ionic strength can irreversibly change the structure, which can significantly affect the signal intensity and sensitivity of the assay [85].

4.3. Multiplex Immunoassay Platforms

Multiplex immunoassays use either antibodies or proteins/peptides as binding molecules to capture circulating proteins or autoantibodies, respectively, during incubation with biological specimens. Unbound proteins are removed by washing, and captured proteins are usually detected by using various labelled reporter ligands. After quantification of the detection label, the signal intensities can be converted into mass units using calibration curves or evaluated qualitatively [87,88].
Two different formats may be used: planar-based assays and microbead-based suspension assays (Figure 2). In the planar assay, the capture antibodies are immobilized on a two-dimensional support, and the fluorescent or chemiluminescent signals are identified. Planar arrays can be produced in two formats, which are either slide-based or microtiter-based. The slide-based format supports various layouts where repeated or individual assays composed of specific sets of antibodies are printed robotically upon the activated slide surface. Microtiter-based immunoassays involve antibodies within the wells of a standard protein-binding plate, similar to conventional enzyme-linked immunosorbent assay ELISA [88,89]. In the suspension assay, the capture antibodies are immobilized on colour- or size-coded microspheres that can be distinguished by their fluorescence intensity in a flow cytometer [88,89]. Each microsphere accommodates a “sandwich” consisting of the captured antibody, the target analyte and the detection antibody, which is labelled with a single chemiluminescent/fluorescent reporter. After the washing stages, lasers excite the reporters, and the emitted light is collected by a series of detectors for quantitative analysis. The use of different coloured microspheres enables the simultaneous detection of many analytes in the same sample [8].
Thousands of proteins can be quantified in parallel through the use of multiplexed immunoassays. The application of these assays has had a profound impact on a wide range of research and diagnostic areas. A number of multiplex assays have been developed for the diagnostic market. Currently, Luminex’s xMAP® Technology (Luminex Corp., Austin, TX, USA) (Luminex Corp., Austin, TX, USA) is the most commonly used platform in commercial assays. It combines advanced fluidics, optics and digital signal processing with proprietary microsphere technology to provide multiplexed assay capabilities. Moreover, its open-architecture design can be configured to perform a wide variety of protein or nucleic acid assays quickly, cost-effectively and accurately [90].
Suspension microsphere-based multiplexed immunoassays have been used to analyse the expression of cytokines, chemokines and growth factors in diverse samples (serum, plasma and tissue culture) and therefore serve as a very straightforward approach for biomarker discovery [91,92,93,94,95,96]. Cytokines, chemokines and growth factors are cell signalling proteins that mediate a wide range of physiological responses, including immunity, inflammation and haematopoiesis. Changes in the levels of these biomarkers are associated with a spectrum of diseases, including autoimmune diseases such as RA [97]. Some studies have shown that the levels of signalling molecules and markers of bone metabolism circulating in the blood of RA patients could be additional markers used in assessing the degree of activity in RA [98,99]. Recently, O’Neil et al. showed that the serum proteome provides a source of proteins that serves both to identify the molecular pathways involved in the development of RA and to classify at-risk individuals [100]. The multiple biomarker approach could utilize a variety of new potential biomarkers and could be used for the early diagnosis of seronegative RA patients, the measurement of disease activity and as a prognostic tool during treatment therapy.

5. Conclusions

The preclinical period in RA development is characterized by immune dysregulation and inflammation without typical manifestations of the disease. During this period, classical laboratory tests (RF and ACPA) can be negative. Multiple genetic studies have revealed the genetic contribution to RA to be between 30% and 60%. The high genetic contribution indicates that genetic studies are essential to identify individuals at risk for RA and to initiate prevention measures, including pharmacological targeting and dietary and lifestyle interventions. For cases without a genetic contribution, novel and more sensitive protein biomarkers are particularly necessary.
The advancement of proteomic techniques in the last two decades has allowed the identification of a large number of candidate proteins that can act as potential biomarkers for several diseases [101,102,103,104,105,106]. This development in particular shows promise for the treatment of highly prevalent disorders, such as cardiovascular disease or cancer, in which early diagnosis is currently impeded by the absence of adequate diagnostic biomarkers. In many other diseases, such as RA, proteomic techniques represent a golden opportunity to improve the current diagnostic strategy. Multiplex immunoassays are a novel tool that allows the application of the knowledge obtained via proteomics research for the diagnosis, monitoring and treatment of diseases [84]. Classical diagnostic approaches based on single parameters have shown enormous limitations in most diseases. Only the specific characteristics of some disorders, such as diabetes, allow such approaches to remain valid. Most diseases are complex and multifactorial processes, for which a more global and ambitious approach based on the analysis of multiple protein biomarkers is required.
In contrast to the single biomarker approach, which focuses only on a particular aspect of the pathologic process, multiplexed immunoassays allow the simultaneous measurement of multiple proteins in biological samples, which has great potential for use in both basic research and clinical diagnostics. From a clinical perspective, the combination of a large number of proteins in multiplex panels would allow for the monitoring of disease states in a much more complete way, with most pathophysiological processes being evaluated at the same time. Additionally, multiplexing not only offers advantages from the point of view of the diagnosis and management of diseases but is also cost-effective. Multiplex assays could provide a more complete picture of the disease and the pathways involved, which would reduce the number of imaging tests required, prevent the repetition of unnecessary laboratory tests and reduce hospital stays, among other benefits [107,108].
Despite all these advantages, multiplex immunoassays also present distinctive challenges not encountered with single-plex assays. Companies such as Millipore, Bio-Rad, Invitrogen, Qiagen and PerkinElmer currently offer multi-protein panels for research purposes in several species, including humans. The widespread adoption of multiplexing for in vitro diagnosis will depend on the availability of robust, affordable analytical platforms and validated multiplex-optimized antibody panels as well as a high level of automation. In RA in particular, simultaneous analysis of multiple biomarkers will yield biomarker signatures of RA subtypes that will allow patients to benefit from personalized medicine.

Author Contributions

Conceptualization, R.D.-P. and C.M.L.; methodology, R.D.-P. and P.C.-S.; writing—original draft preparation, R.D.-P., P.C.-S. and C.M.L.; writing—review and editing, R.D.-P., P.C.-S. and C.M.L.; investigation, R.D.-P., P.C.-S. and C.M.L.; visualization, R.D.-P. and C.M.L.; supervision, R.D.-P. and C.M.L.; project administration, R.D.-P.; funding acquisition, R.D.-P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Fondecyt, grant n° 11130198.

Conflicts of Interest

Carlos M. Laborde is currently employee of Dreamgenics, Inc. The authors declare no conflict of interest.

References

  1. Tobón, G.J.; Youinou, P.; Saraux, A. The environment, geo-epidemiology, and autoimmune disease: Rheumatoid arthritis. J. Autoimmun. 2010, 35, 10–14. [Google Scholar] [CrossRef] [PubMed]
  2. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O.; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 Rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010, 62, 2569–2581. [Google Scholar] [CrossRef] [PubMed]
  3. Conaghan, P.G.; Green, M.J.; Emery, P. Established rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 1999, 13, 561–575. [Google Scholar] [CrossRef] [PubMed]
  4. Castro-Santos, P.; Laborde, C.M.; Díaz-Peña, R. Genomics, proteomics and metabolomics: Their emerging roles in the discovery and validation of rheumatoid arthritis biomarkers. Clin. Exp. Rheumatol. 2015, 33, 279–286. [Google Scholar] [PubMed]
  5. Vander Cruyssen, B.; Hoffman, I.E.A.; Peene, I.; Union, A.; Mielants, H.; Meheus, L.; De Keyser, F. Prediction models for rheumatoid arthritis during diagnostic investigation: Evaluation of combinations of rheumatoid factor, anti-citrullinated protein/peptide antibodies and the human leucocyte antigen-shared epitope. Ann. Rheum. Dis. 2007, 66, 364–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Van Beers-Tas, M.H.; Turk, S.A.; Van Schaardenburg, D. How does established rheumatoid arthritis develop, and are there possibilities for prevention? Best Pract. Res. Clin. Rheumatol. 2015, 29, 527–542. [Google Scholar] [CrossRef] [PubMed]
  7. Kyburz, D.; Gabay, C.; Michel, B.A.; Finckh, A. The long-term impact of early treatment of rheumatoid arthritis on radiographic progression: A population-based cohort study. Rheumatology (Oxf.) 2011, 50, 1106–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Lukas, C.; Combe, B.; Ravaud, P.; Sibilia, J.; Landewé, R.; Van Der Heijde, D. Favorable effect of very early disease-modifying antirheumatic drug treatment on radiographic progression in early inflammatory arthritis: Data from the ôtude et Suivi des polyarthrites indifférenciées récentes (study and followup of early undifferentiate. Arthritis Rheum. 2011, 63, 1804–1811. [Google Scholar] [CrossRef] [PubMed]
  9. Gerlag, D.M.; Raza, K.; Van Baarsen, L.G.M.; Brouwer, E.; Buckley, C.D.; Burmester, G.R.; Gabay, C.; Catrina, A.I.; Cope, A.P.; Cornelis, F.; et al. EULAR recommendations for terminology and research in individuals at risk of rheumatoid arthritis: Report from the Study Group for Risk Factors for Rheumatoid Arthritis. Ann. Rheum. Dis. 2012, 71, 638–641. [Google Scholar] [CrossRef]
  10. Sparks, J.A.; Costenbader, K.H. Genetics, environment, and gene-environment interactions in the development of systemic rheumatic diseases. Rheum. Dis. Clin. N. Am. 2014, 40, 637–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. MacGregor, A.J.; Snieder, H.; Rigby, A.S.; Koskenvuo, M.; Kaprio, J.; Aho, K.; Silman, A.J. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000, 43, 30–37. [Google Scholar] [CrossRef]
  12. Gregersen, P.K.; Silver, J.; Winchester, R.J. The shared epitope hypothesis. an approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum. 1987, 30, 1205–1213. [Google Scholar] [CrossRef]
  13. Okada, Y.; Wu, D.; Trynka, G.; Raj, T.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Yoshida, S.; et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2013, 506, 376–381. [Google Scholar] [CrossRef] [PubMed]
  14. Arend, W.P.; Firestein, G.S. Pre-rheumatoid arthritis: Predisposition and transition to clinical synovitis. Nat. Rev. Rheumatol. 2012, 8, 573–586. [Google Scholar] [CrossRef] [PubMed]
  15. Bossini-Castillo, L.; de Kovel, C.; Kallberg, H.; van ‘t Slot, R.; Italiaander, A.; Coenen, M.; Tak, P.P.; Posthumus, M.D.; Wijmenga, C.; Huizinga, T.; et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann. Rheum. Dis. 2015, 74, e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sugiyama, D.; Nishimura, K.; Tamaki, K.; Tsuji, G.; Nakazawa, T.; Morinobu, A.; Kumagai, S. Impact of smoking as a risk factor for developing rheumatoid arthritis: A meta-analysis of observational studies. Ann. Rheum. Dis. 2010, 69, 70–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Catrina, A.I.; Ytterberg, A.J.; Reynisdottir, G.; Malmström, V.; Klareskog, L. Lungs, joints and immunity against citrullinated proteins in rheumatoid arthritis. Nat. Rev. Rheumatol. 2014, 10, 645–653. [Google Scholar] [CrossRef]
  18. Klareskog, L.; Rönnelid, J.; Saevarsdottir, S.; Padyukov, L.; Alfredsson, L. The importance of differences; On environment and its interactions with genes and immunity in the causation of rheumatoid arthritis. J. Intern. Med. 2020, 287, 514–533. [Google Scholar] [CrossRef]
  19. De Roos, A.J.; Koehoorn, M.; Tamburic, L.; Davies, H.W.; Brauer, M. Proximity to Traffic, Ambient Air Pollution, and Community Noise in Relation to Incident Rheumatoid Arthritis. Environ. Health Perspect. 2014, 122, 1075–1080. [Google Scholar] [CrossRef] [Green Version]
  20. Heliovaara, M. Coffee consumption, rheumatoid factor, and the risk of rheumatoid arthritis. Ann. Rheum. Dis. 2000, 59, 631–635. [Google Scholar] [CrossRef] [Green Version]
  21. Yahya, A.; Bengtsson, C.; Larsson, P.; Too, C.L.; Mustafa, A.N.; Abdullah, N.A.; Muhamad, N.A.; Klareskog, L.; Murad, S.; Alfredsson, L. Silica exposure is associated with an increased risk of developing ACPA-positive rheumatoid arthritis in an Asian population: Evidence from the Malaysian MyEIRA case-control study. Mod. Rheumatol. 2013, 24, 271–274. [Google Scholar] [CrossRef] [PubMed]
  22. Martins, P.; Fonseca, J.E. How to investigate: Pre-clinical rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101438. [Google Scholar] [CrossRef] [PubMed]
  23. Hazes, J.M.W.; Coulie, P.G.; Geenen, V.; Vermeire, S.; Carbonnel, F.; Louis, E.; Masson, P.; De Keyser, F. Rheumatoid arthritis and pregnancy: Evolution of disease activity and pathophysiological considerations for drug use. Rheumatology (Oxf.) 2011, 50, 1955–1968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Qin, B.; Yang, M.; Fu, H.; Ma, N.; Wei, T.; Tang, Q.; Hu, Z.; Liang, Y.; Yang, Z.; Zhong, R. Body mass index and the risk of rheumatoid arthritis: A systematic review and dose-response meta-analysis. Arthritis Res. Ther. 2015, 17, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Scher, J.U.; Littman, D.R.; Abramson, S.B. Microbiome in Inflammatory Arthritis and Human Rheumatic Diseases. Arthritis Rheumatol. 2016, 68, 35–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Scher, J.U.; Joshua, V.; Artacho, A.; Abdollahi-Roodsaz, S.; Öckinger, J.; Kullberg, S.; Sköld, M.; Eklund, A.; Grunewald, J.; Clemente, J.C.; et al. The lung microbiota in early rheumatoid arthritis and autoimmunity. Microbiome 2016, 4, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Potempa, J.; Mydel, P.; Koziel, J. The case for periodontitis in the pathogenesis of rheumatoid arthritis. Nat. Rev. Rheumatol. 2017, 13, 606–620. [Google Scholar] [CrossRef] [PubMed]
  28. Bergström, U.; Jacobsson, L.T.H.; Nilsson, J.Å.; Berglund, G.; Turesson, C. Pulmonary dysfunction, smoking, socioeconomic status and the risk of developing rheumatoid arthritis. Rheumatology (Oxf.) 2011, 50, 2005–2013. [Google Scholar] [CrossRef] [Green Version]
  29. Eriksson, J.K.; Neovius, M.; Ernestam, S.; Lindblad, S.; Simard, J.F.; Askling, J. Incidence of Rheumatoid Arthritis in Sweden: A Nationwide Population-Based Assessment of Incidence, Its Determinants, and Treatment Penetration. Arthritis Care Res. (Hoboken) 2013, 65, 870–878. [Google Scholar] [CrossRef]
  30. Spear, B.B.; Heath-Chiozzi, M.; Huff, J. Clinical application of pharmacogenetics. Trends Mol. Med. 2001, 7, 201–204. [Google Scholar] [CrossRef]
  31. Mahler, M.; Martinez-Prat, L.; Sparks, J.A.; Deane, K.D. Precision medicine in the care of rheumatoid arthritis: Focus on prediction and prevention of future clinically-apparent disease. Autoimmun. Rev. 2020, 19, 102506. [Google Scholar] [CrossRef] [PubMed]
  32. Trouw, L.A.; Rispens, T.; Toes, R.E.M. Beyond citrullination: Other post-translational protein modifications in rheumatoid arthritis. Nat. Rev. Rheumatol. 2017, 13, 331–339. [Google Scholar] [CrossRef] [PubMed]
  33. Sutton, B.; Corper, A.; Bonagura, V.; Taussig, M. The structure and origin of rheumatoid factors. Immunol. Today 2000, 21, 177–183. [Google Scholar] [CrossRef]
  34. Conigliaro, P.; Chimenti, M.S.; Triggianese, P.; Sunzini, F.; Novelli, L.; Perricone, C.; Perricone, R. Autoantibodies in inflammatory arthritis. Autoimmun. Rev. 2016, 15, 673–683. [Google Scholar] [CrossRef] [Green Version]
  35. Daha, N.A.; Toes, R.E.M. Rheumatoid arthritis: Are ACPA-positive and ACPA-negative RA the same disease? Nat. Rev. Rheumatol. 2011, 7, 202–203. [Google Scholar] [CrossRef] [PubMed]
  36. Wysocki, T.; Olesińska, M.; Paradowska-Gorycka, A. Current Understanding of an Emerging Role of HLA-DRB1 Gene in Rheumatoid Arthritis-From Research to Clinical Practice. Cells 2020, 9, 1127. [Google Scholar] [CrossRef] [PubMed]
  37. Robinson, W.H.; Lindstrom, T.M.; Cheung, R.K.; Sokolove, J. Mechanistic biomarkers for clinical decision making in rheumatic diseases. Nat. Rev. Rheumatol. 2013, 9, 267–276. [Google Scholar] [CrossRef] [Green Version]
  38. Shi, J.; Knevel, R.; Suwannalai, P.; Van Der Linden, M.P.; Janssen, G.M.C.; Van Veelen, P.A.; Levarht, N.E.W.; Van Der Helm-van Mil, A.H.M.; Cerami, A.; Huizinga, T.W.J.; et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc. Natl. Acad. Sci. USA 2011, 108, 17372–17377. [Google Scholar] [CrossRef] [Green Version]
  39. Ren, J.; Sun, L.; Zhao, J. Meta-analysis: Diagnostic accuracy of antibody against peptidylarginine deiminase 4 by ELISA for rheumatoid arthritis. Clin. Rheumatol. 2017, 36, 2431–2438. [Google Scholar] [CrossRef]
  40. Halvorsen, E.H.; Pollmann, S.; Gilboe, I.-M.; van der Heijde, D.; Landewe, R.; Odegard, S.; Kvien, T.K.; Molberg, O. Serum IgG antibodies to peptidylarginine deiminase 4 in rheumatoid arthritis and associations with disease severity. Ann. Rheum. Dis. 2007, 67, 414–417. [Google Scholar] [CrossRef]
  41. Zhu, J.-N.; Nie, L.-Y.; Lu, X.-Y.; Wu, H.-X. Meta-analysis: Compared with anti-CCP and rheumatoid factor, could anti-MCV be the next biomarker in the rheumatoid arthritis classification criteria? Clin. Chem. Lab. Med. 2019, 57, 1668–1679. [Google Scholar] [CrossRef] [PubMed]
  42. Zeng, T.; Tan, L. 14-3-3η protein: A promising biomarker for rheumatoid arthritis. Biomark. Med. 2018, 12, 917–925. [Google Scholar] [CrossRef] [PubMed]
  43. Bae, S.-C.; Lee, Y.H. Calprotectin levels in rheumatoid arthritis and their correlation with disease activity: A meta-analysis. Postgrad. Med. 2017, 129, 531–537. [Google Scholar] [CrossRef] [PubMed]
  44. Shi, J.; van Steenbergen, H.W.; van Nies, J.A.B.; Levarht, E.W.N.; Huizinga, T.W.J.; van der Helm-van Mil, A.H.M.; Toes, R.E.M.; Trouw, L.A. The specificity of anti-carbamylated protein antibodies for rheumatoid arthritis in a setting of early arthritis. Arthritis Res. Ther. 2015, 17, 339. [Google Scholar] [CrossRef] [Green Version]
  45. Verheul, M.K.; Böhringer, S.; van Delft, M.A.M.; Jones, J.D.; Rigby, W.F.C.; Gan, R.W.; Holers, V.M.; Edison, J.D.; Deane, K.D.; Janssen, K.M.J.; et al. Triple Positivity for Anti-Citrullinated Protein Autoantibodies, Rheumatoid Factor, and Anti-Carbamylated Protein Antibodies Conferring High Specificity for Rheumatoid Arthritis. Arthritis Rheumatol. 2018, 70, 1721–1731. [Google Scholar] [CrossRef] [Green Version]
  46. Foulquier, C.; Sebbag, M.; Clavel, C.; Chapuy-Regaud, S.; Al Badine, R.; Méchin, M.-C.; Vincent, C.; Nachat, R.; Yamada, M.; Takahara, H.; et al. Peptidyl arginine deiminase type 2 (PAD-2) and PAD-4 but not PAD-1, PAD-3, and PAD-6 are expressed in rheumatoid arthritis synovium in close association with tissue inflammation. Arthritis Rheum. 2007, 56, 3541–3553. [Google Scholar] [CrossRef] [PubMed]
  47. Darrah, E.; Giles, J.T.; Davis, R.L.; Naik, P.; Wang, H.; Konig, M.F.; Cappelli, L.C.; Bingham, C.O.; Danoff, S.K.; Andrade, F. Autoantibodies to Peptidylarginine Deiminase 2 Are Associated With Less Severe Disease in Rheumatoid Arthritis. Front. Immunol. 2018, 9, 2696. [Google Scholar] [CrossRef] [PubMed]
  48. Turesson, C.; Mathsson, L.; Jacobsson, L.T.H.; Sturfelt, G.; Rönnelid, J. Antibodies to modified citrullinated vimentin are associated with severe extra-articular manifestations in rheumatoid arthritis. Ann. Rheum. Dis. 2013, 72, 2047–2048. [Google Scholar] [CrossRef]
  49. Lindenberg, L.; Spengler, L.; Bang, H.; Dorner, T.; Maslyanskiy, A.L.; Lapin, S.V.; Ilivanova, E.I.; Martinez-Gamboa, L.; Bastian, H.; Wittenborn, E.; et al. Restrictive IgG antibody response against mutated citrullinated vimentin predicts response to rituximab in patients with rheumatoid arthritis. Arthritis Res. Ther. 2015, 17, 206. [Google Scholar] [CrossRef] [Green Version]
  50. Bläß, S.; Union, A.; Raymackers, J.; Schumann, F.; Ungethum, U.; Muller-Steinbach, S.; De Keyser, F.; Engel, J.M.; Burmester, G.R. The stress protein BiP is overexpressed and is a major B and T cell target in rheumatoid arthritis. Arthritis Rheum. 2001, 44, 761–771. [Google Scholar] [CrossRef]
  51. Strollo, R.; Ponchel, F.; Malmström, V.; Rizzo, P.; Bombardieri, M.; Wenham, C.Y.; Landy, R.; Perret, D.; Watt, F.; Corrigall, V.M.; et al. Autoantibodies to Posttranslationally Modified Type II Collagen as Potential Biomarkers for Rheumatoid Arthritis. Arthritis Rheum. 2013, 65, 1702–1712. [Google Scholar] [CrossRef] [PubMed]
  52. Seeling, M.; Brückner, C.; Nimmerjahn, F. Differential antibody glycosylation in autoimmunity: Sweet biomarker or modulator of disease activity? Nat. Rev. Rheumatol. 2017, 13, 621–630. [Google Scholar] [CrossRef] [PubMed]
  53. Hafkenscheid, L.; Moel, E.; Smolik, I.; Tanner, S.; Meng, X.; Jansen, B.C.; Bondt, A.; Wuhrer, M.; Huizinga, T.W.J.; Toes, R.E.M.; et al. N-Linked Glycans in the Variable Domain of IgG Anti–Citrullinated Protein Antibodies Predict the Development of Rheumatoid Arthritis. Arthritis Rheumatol. 2019, 71, 1626–1633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Maksymowych, W.P.; Naides, S.J.; Bykerk, V.; Siminovitch, K.A.; van Schaardenburg, D.; Boers, M.; Landewé, R.; van der Heijde, D.; Tak, P.-P.; Genovese, M.C.; et al. Serum 14-3-3η is a Novel Marker that Complements Current Serological Measurements to Enhance Detection of Patients with Rheumatoid Arthritis. J. Rheumatol. 2014, 41, 2104–2113. [Google Scholar] [CrossRef] [Green Version]
  55. Maksymowych, W.P.; Marotta, A. 14-3-3η: A novel biomarker platform for rheumatoid arthritis. Clin. Exp. Rheumatol. 2014, 32, S35–S39. [Google Scholar]
  56. Jonsson, M.K.; Sundlisæter, N.P.; Nordal, H.H.; Hammer, H.B.; Aga, A.-B.; Olsen, I.C.; Brokstad, K.A.; van der Heijde, D.; Kvien, T.K.; Fevang, B.-T.S.; et al. Calprotectin as a marker of inflammation in patients with early rheumatoid arthritis. Ann. Rheum. Dis. 2017, 76, 2031–2037. [Google Scholar] [CrossRef]
  57. Lourido, L.; Blanco, F.J.; Ruiz-Romero, C. Defining the proteomic landscape of rheumatoid arthritis: Progress and prospective clinical applications. Expert Rev. Proteom. 2017, 14, 431–444. [Google Scholar] [CrossRef]
  58. Oliver, J.; Plant, D.; Webster, A.P.; Barton, A. Genetic and genomic markers of anti-TNF treatment response in rheumatoid arthritis. Biomark. Med. 2015, 9, 499–512. [Google Scholar] [CrossRef]
  59. Lequerré, T.; Rottenberg, P.; Derambure, C.; Cosette, P.; Vittecoq, O. Predictors of treatment response in rheumatoid arthritis. Jt. Bone Spine 2019, 86, 151–158. [Google Scholar] [CrossRef]
  60. Fabre, S.; Dupuy, A.M.; Dossat, N.; Guisset, C.; Cohen, J.D.; Cristol, J.P.; Daures, J.P.; Jorgensen, C. Protein biochip array technology for cytokine profiling predicts etanercept responsiveness in rheumatoid arthritis. Clin. Exp. Immunol. 2008, 153, 188–195. [Google Scholar] [CrossRef]
  61. Obry, A.; Lequerré, T.; Hardouin, J.; Boyer, O.; Fardellone, P.; Philippe, P.; Le Loët, X.; Cosette, P.; Vittecoq, O. Identification of S100A9 as Biomarker of Responsiveness to the Methotrexate/Etanercept Combination in Rheumatoid Arthritis Using a Proteomic Approach. PLoS ONE 2014, 9, e115800. [Google Scholar] [CrossRef] [PubMed]
  62. Obry, A.; Hardouin, J.; Lequerré, T.; Jarnier, F.; Boyer, O.; Fardellone, P.; Philippe, P.; Marcelli, C.; Loët, X.L.; Vittecoq, O.; et al. Identification of 7 Proteins in Sera of RA Patients with Potential to Predict ETA/MTX Treatment Response. Theranostics 2015, 5, 1214–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Trocme, C.; Marotte, H.; Baillet, A.; Pallot-Prades, B.; Garin, J.; Grange, L.; Miossec, P.; Tebib, J.; Berger, F.; Nissen, M.J.; et al. Apolipoprotein A-I and platelet factor 4 are biomarkers for infliximab response in rheumatoid arthritis. Ann. Rheum. Dis. 2009, 68, 1328–1333. [Google Scholar] [CrossRef] [PubMed]
  64. Ortea, I.; Roschitzki, B.; Ovalles, J.G.; Longo, J.L.; de la Torre, I.; González, I.; Gómez-Reino, J.J.; González, A. Discovery of serum proteomic biomarkers for prediction of response to infliximab (a monoclonal anti-TNF antibody) treatment in rheumatoid arthritis: An exploratory analysis. J. Proteom. 2012, 77, 372–382. [Google Scholar] [CrossRef] [Green Version]
  65. Hueber, W.; Kidd, B.A.; Tomooka, B.H.; Lee, B.J.; Bruce, B.; Fries, J.F.; Sønderstrup, G.; Monach, P.; Drijfhout, J.W.; van Venrooij, W.J.; et al. Antigen microarray profiling of autoantibodies in rheumatoid arthritis. Arthritis Rheum. 2005, 52, 2645–2655. [Google Scholar] [CrossRef]
  66. Pandey, A.; Mann, M. Proteomics to study genes and genomes. Nature 2000, 405, 837–846. [Google Scholar] [CrossRef] [PubMed]
  67. Blackstock, W.P.; Weir, M.P. Proteomics: Quantitative and physical mapping of cellular proteins. Trends Biotechnol. 1999, 17, 121–127. [Google Scholar] [CrossRef]
  68. Trojanowski, J.Q.; Vandeerstichele, H.; Korecka, M.; Clark, C.M.; Aisen, P.S.; Petersen, R.C.; Blennow, K.; Soares, H.; Simon, A.; Lewczuk, P.; et al. Update on the biomarker core of the Alzheimer’s Disease Neuroimaging Initiative subjects. Alzheimer’s Dement. 2010, 6, 230–238. [Google Scholar] [CrossRef] [Green Version]
  69. Brooks, J.D. Translational genomics: The challenge of developing cancer biomarkers. Genome Res. 2012, 22, 183–187. [Google Scholar] [CrossRef] [Green Version]
  70. Vanmassenhove, J.; Vanholder, R.; Nagler, E.; Van Biesen, W. Urinary and serum biomarkers for the diagnosis of acute kidney injury: An in-depth review of the literature. Nephrol. Dial. Transplant. 2013, 28, 254–273. [Google Scholar] [CrossRef] [Green Version]
  71. Chen, R.; Snyder, M. Systems biology: Personalized medicine for the future? Curr. Opin. Pharmacol. 2012, 12, 623–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Anderson, N.L.; Anderson, N.G. The Human Plasma Proteome. Mol. Cell. Proteom. 2002, 1, 845–867. [Google Scholar] [CrossRef] [Green Version]
  73. Fountoulakis, M.; Juranville, J.-F.; Jiang, L.; Avila, D.; Jakob, P.; Berndt, P.; Evers, S.; Langen, H. Depletion of the high-abundance plasma proteins. Amino Acids 2004, 27, 249–259. [Google Scholar] [CrossRef] [PubMed]
  74. Boschetti, E.; Righetti, P.G. The ProteoMiner in the proteomic arena: A non-depleting tool for discovering low-abundance species. J. Proteom. 2008, 71, 255–264. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, C.; Duan, J.; Liu, T.; Smith, R.D.; Qian, W.-J. Contributions of immunoaffinity chromatography to deep proteome profiling of human biofluids. J. Chromatogr. B 2016, 1021, 57–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wild, D.; Sheehan, C. Standardization and Calibration. In The Immunoassay Handbook; Elsevier Ltd.: Amsterdam, The Netherlands, 2013; pp. 167–176. [Google Scholar]
  77. Chau, C.H.; Rixe, O.; McLeod, H.; Figg, W.D. Validation of Analytic Methods for Biomarkers Used in Drug Development. Clin. Cancer Res. 2008, 14, 5967–5976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ward, G.; Simpson, A.; Boscato, L.; Hickman, P.E. The investigation of interferences in immunoassay. Clin. Biochem. 2017, 50, 1306–1311. [Google Scholar] [CrossRef] [Green Version]
  79. Holm, A.; Wu, W.; Lund-Johansen, F. Antibody array analysis of labelled proteomes: How should we control specificity? New Biotechnol. 2012, 29, 578–585. [Google Scholar] [CrossRef]
  80. Landegren, U.; Vänelid, J.; Hammond, M.; Nong, R.Y.; Wu, D.; Ullerås, E.; Kamali-Moghaddam, M. Opportunities for Sensitive Plasma Proteome Analysis. Anal. Chem. 2012, 84, 1824–1830. [Google Scholar] [CrossRef]
  81. Juncker, D.; Bergeron, S.; Laforte, V.; Li, H. Cross-reactivity in antibody microarrays and multiplexed sandwich assays: Shedding light on the dark side of multiplexing. Curr. Opin. Chem. Biol. 2014, 18, 29–37. [Google Scholar] [CrossRef] [PubMed]
  82. Tate, J.; Ward, G. Interferences in immunoassay. Clin. Biochem. Rev. 2004, 25, 105–120. [Google Scholar] [PubMed]
  83. Koshida, S.; Asanuma, K.; Kuribayashi, K.; Goto, M.; Tsuji, N.; Kobayashi, D.; Tanaka, M.; Watanabe, N. Prevalence of human anti-mouse antibodies (HAMAs) in routine examinations. Clin. Chim. Acta 2010, 411, 391–394. [Google Scholar] [CrossRef] [PubMed]
  84. Kingsmore, S.F. Multiplexed protein measurement: Technologies and applications of protein and antibody arrays. Nat. Rev. Drug Discov. 2006, 5, 310–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Pfleger, C.; Schloot, N.; Veld, F. ter Effect of serum content and diluent selection on assay sensitivity and signal intensity in multiplex bead-based immunoassays. J. Immunol. Methods 2008, 329, 214–218. [Google Scholar] [CrossRef]
  86. Fu, Q.; Schoenhoff, F.S.; Savage, W.J.; Zhang, P.; Van Eyk, J.E. Multiplex assays for biomarker research and clinical application: Translational science coming of age. Proteom. Clin. Appl. 2010, 4, 271–284. [Google Scholar] [CrossRef]
  87. Ling, M.M.; Ricks, C.; Lea, P. Multiplexing molecular diagnostics and immunoassays using emerging microarray technologies. Expert Rev. Mol. Diagn. 2007, 7, 87–98. [Google Scholar] [CrossRef] [PubMed]
  88. Ellington, A.A.; Kullo, I.J.; Bailey, K.R.; Klee, G.G. Antibody-Based Protein Multiplex Platforms: Technical and Operational Challenges. Clin. Chem. 2010, 56, 186–193. [Google Scholar] [CrossRef]
  89. Fu, Q.; Zhu, J.; Van Eyk, J.E. Comparison of Multiplex Immunoassay Platforms. Clin. Chem. 2010, 56, 314–318. [Google Scholar] [CrossRef]
  90. Zheng, W.; He, L. Multiplexed Immunoassays. In Advances in Immunoassay Technology; InTech: Rijeka, Croatia, 2012; pp. 143–164. [Google Scholar]
  91. Carson, R.T.; Vignali, D.A. Simultaneous quantitation of 15 cytokines using a multiplexed flow cytometric assay. J. Immunol. Methods 1999, 227, 41–52. [Google Scholar] [CrossRef]
  92. de Jager, W.; te Velthuis, H.; Prakken, B.J.; Kuis, W.; Rijkers, G.T. Simultaneous Detection of 15 Human Cytokines in a Single Sample of Stimulated Peripheral Blood Mononuclear Cells. Clin. Diagnostic Lab. Immunol. 2003, 10, 133–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Prabhakar, U.; Eirikis, E.; Davis, H.M. Simultaneous quantification of proinflammatory cytokines in human plasma using the LabMAPTM assay. J. Immunol. Methods 2002, 260, 207–218. [Google Scholar] [CrossRef]
  94. Olsson, A.; Vanderstichele, H.; Andreasen, N.; De Meyer, G.; Wallin, A.; Holmberg, B.; Rosengren, L.; Vanmechelen, E.; Blennow, K. Simultaneous Measurement of β-Amyloid(1–42), Total Tau, and Phosphorylated Tau (Thr181) in Cerebrospinal Fluid by the xMAP Technology. Clin. Chem. 2005, 51, 336–345. [Google Scholar] [CrossRef] [PubMed]
  95. DEJAGER, W.; RIJKERS, G. Solid-phase and bead-based cytokine immunoassay: A comparison. Methods 2006, 38, 294–303. [Google Scholar] [CrossRef] [PubMed]
  96. Maier, R.; Weger, M.; Haller-Schober, E.M.; El-Shabrawi, Y.; Theisl, A.; Barth, A.; Aigner, R.; Hass, A. Application of multiplex cytometric bead array technology for the measurement of angiogenic factor in the vitreous. Mol. Vis. 2006, 12, 1143–1147. [Google Scholar]
  97. Eastman, P.S.; Manning, W.C.; Qureshi, F.; Haney, D.; Cavet, G.; Alexander, C.; Hesterberg, L.K. Characterization of a multiplex, 12-biomarker test for rheumatoid arthritis. J. Pharm. Biomed. Anal. 2012, 70, 415–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Milman, N.; Karsh, J.; Booth, R.A. Correlation of a multi-cytokine panel with clinical disease activity in patients with rheumatoid arthritis. Clin. Biochem. 2010, 43, 1309–1314. [Google Scholar] [CrossRef] [PubMed]
  99. Meyer, P.W.A.; Hodkinson, B.; Ally, M.; Musenge, E.; Wadee, A.A.; Fickl, H.; Tikly, M.; Anderson, R. Circulating cytokine profiles and their relationships with autoantibodies, acute phase reactants, and disease activity in patients with rheumatoid arthritis. Mediat. Inflamm. 2010, 2010. [Google Scholar] [CrossRef] [Green Version]
  100. O’Neil, L.J.; Spicer, V.; Smolik, I.; Meng, X.; Goel, R.R.; Anaparti, V.; Wilkins, J.; El-Gabalawy, H.S. A Serum Protein Signature is associated with Rheumatoid Arthritis development. Arthritis Rheumatol. 2020. [Google Scholar] [CrossRef]
  101. Laborde, C.M.; Alonso-Orgaz, S.; Mourino-Alvarez, L.; Moreu, J.; Vivanco, F.; Padial, L.R.; Barderas, M.G. The plasma proteomic signature as a strategic tool for early diagnosis of acute coronary syndrome. Proteome Sci. 2014, 12, 43. [Google Scholar] [CrossRef] [Green Version]
  102. Whelan, C.D.; Mattsson, N.; Nagle, M.W.; Vijayaraghavan, S.; Hyde, C.; Janelidze, S.; Stomrud, E.; Lee, J.; Fitz, L.; Samad, T.A.; et al. Multiplex proteomics identifies novel CSF and plasma biomarkers of early Alzheimer’s disease. Acta Neuropathol. Commun. 2019, 7, 169. [Google Scholar] [CrossRef]
  103. Rader, J.S.; Pan, A.; Corbin, B.; Iden, M.; Lu, Y.; Vellano, C.P.; Akbani, R.; Mills, G.B.; Simpson, P. Identification and validation of a prognostic proteomic signature for cervical cancer. Gynecol. Oncol. 2019, 155, 324–330. [Google Scholar] [CrossRef] [PubMed]
  104. Baldan-Martin, M.; Martin-Rojas, T.; Corbacho-Alonso, N.; Lopez, J.A.; Sastre-Oliva, T.; Gil-Dones, F.; Vazquez, J.; Arevalo, J.M.; Mourino-Alvarez, L.; Barderas, M.G. Comprehensive Proteomic Profiling of Pressure Ulcers in Patients with Spinal Cord Injury Identifies a Specific Protein Pattern of Pathology. Adv. Wound Care 2020, 9, 277–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Corbacho-Alonso, N.; Baldán-Martín, M.; López, J.A.; Rodríguez-Sánchez, E.; Martínez, P.J.; Mourino-Alvarez, L.; Martin-Rojas, T.; Sastre-Oliva, T.; Madruga, F.; Vázquez, J.; et al. Novel molecular plasma signatures on cardiovascular disease can stratify patients throughout life. J. Proteom. 2020, 222, 103816. [Google Scholar] [CrossRef] [PubMed]
  106. Martinez, P.J.; Agudiez, M.; Molero, D.; Martin-Lorenzo, M.; Baldan-Martin, M.; Santiago-Hernandez, A.; García-Segura, J.M.; Madruga, F.; Cabrera, M.; Calvo, E.; et al. Urinary metabolic signatures reflect cardiovascular risk in the young, middle-aged, and elderly populations. J. Mol. Med. 2020, 1–11. [Google Scholar] [CrossRef] [PubMed]
  107. Dincer, C.; Bruch, R.; Kling, A.; Dittrich, P.S.; Urban, G.A. Multiplexed Point-of-Care Testing—xPOCT. Trends Biotechnol. 2017, 35, 728–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ahsan, H. The biomolecules of beauty: Biochemical pharmacology and immunotoxicology of cosmeceuticals. J. Immunoass. Immunochem. 2019, 40, 91–108. [Google Scholar] [CrossRef]
Figure 1. Potential sources of interference in multiplex immunoassays; (A) antibody cross-reactivity, (B) non-specific antibody binding and (C) antibody interference.
Figure 1. Potential sources of interference in multiplex immunoassays; (A) antibody cross-reactivity, (B) non-specific antibody binding and (C) antibody interference.
Jpm 10 00202 g001
Figure 2. Multiplexed immunoassay systems: (A) planar-based assays and (B) microbead-based suspension assays.
Figure 2. Multiplexed immunoassay systems: (A) planar-based assays and (B) microbead-based suspension assays.
Jpm 10 00202 g002
Table 1. Environmental factors associated with the development of rheumatoid arthritis (RA).
Table 1. Environmental factors associated with the development of rheumatoid arthritis (RA).
Predisposing FactorsACPA
SmokingPositive
PollutionPositive
PeriodontitisPositive
PregnancyNegative
ObesityNegative
DietNegative
ExerciseNegative
MicrobiomeNegative
ACPA: Anti-cyclic citrullinated peptide antibodies.
Table 2. Overview of biomarkers with potential utility in rheumatoid arthritis.
Table 2. Overview of biomarkers with potential utility in rheumatoid arthritis.
BiomarkerCommentsReference
ACPADiagnostic and prognostic value. Included in the 2010 ACR RA classification criteria.[34]
RFDiagnostic and prognostic value. Included in the 2010 ACR RA classification criteria. Moderate specificity[34]
Anti-CarP antibodiesLower sensitivity than ACPAs and RF but showed promising results in combination with ACPA/RF. Associated with erosive disease.[38]
Anti-PAD antibodiesAnti-PAD4 antibodies have been associated with radiographic progression.[39,40]
Anti-MCV antibodiesAssociated with bone erosion[41]
14-3-3η proteinDiagnostic utility for RA. Associated with radiographic progression in early RA[42]
CalprotectinAssociated with disease activity.[43]
Anti-CarP: Anti-carbamylated protein; ACPA: Anti-citrullinated protein autoantibodies; ACR: American College of Rheumatology; anti-MCV: Anti-mutated citrullinated vimentin; anti-PAD: Anti-peptidylarginine deiminase; RF: Rheumatoid factor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Laborde, C.M.; Castro-Santos, P.; Díaz-Peña, R. Contribution of Multiplex Immunoassays to Rheumatoid Arthritis Management: From Biomarker Discovery to Personalized Medicine. J. Pers. Med. 2020, 10, 202. https://doi.org/10.3390/jpm10040202

AMA Style

Laborde CM, Castro-Santos P, Díaz-Peña R. Contribution of Multiplex Immunoassays to Rheumatoid Arthritis Management: From Biomarker Discovery to Personalized Medicine. Journal of Personalized Medicine. 2020; 10(4):202. https://doi.org/10.3390/jpm10040202

Chicago/Turabian Style

Laborde, Carlos M., Patricia Castro-Santos, and Roberto Díaz-Peña. 2020. "Contribution of Multiplex Immunoassays to Rheumatoid Arthritis Management: From Biomarker Discovery to Personalized Medicine" Journal of Personalized Medicine 10, no. 4: 202. https://doi.org/10.3390/jpm10040202

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop