![]() Use of anti-cd26 antibody levels as biomarkers of autoimmune and/or inflammatory diseases. (Machine-
专利摘要:
The present invention relates to the use of anti-cd26 antibodies as markers for the screening (screening), diagnosis or monitoring of subjects suspected of having an autoimmune and/or inflammatory disease, such as rheumatoid arthritis, particularly those in an early stage of the disease. More specifically, it relates to an in vitro method for the screening of a subject that is suspected of having an autoimmune and/or inflammatory disease and comprises the following steps: A) the determination of anti-cd26 igg antibody levels in a sample of the patient containing the antibodies. B) compare the levels in that sample containing antibodies with a reference value, so that an increase in anti-cd26 igg antibody levels in the subject sample with respect to said reference value is indicative of the disease and in the that said reference value corresponds to the average levels of anti-cd26 igg igg antibodies in healthy subjects. (Machine-translation by Google Translate, not legally binding) 公开号:ES2642723A1 申请号:ES201630620 申请日:2016-05-12 公开日:2017-11-17 发明作者:José María PEGO REIGOSA;Óscar CORDERO SANTAMARÍA;Rubén VARELA CALVIÑO 申请人:Universidade de Santiago de Compostela;Servizo Galego de Saude SERGAS; IPC主号:
专利说明:
USE OF ANTI-CD26 ANTIBODY LEVELS AS BIOMARCATORS OF AUTOIMMUNE AND / OR INFLAMMATORY DISEASES FIELD OF THE INVENTION The present invention can be included in the field of personalized medicine, in which specific biomarkers are used for screening, diagnosis, prognosis, prediction of treatment results, and / or monitoring of certain disease or disorder. Specifically, anti-CD26 antibodies are used in the present invention as markers for screening, diagnosis or monitoring of human subjects who have, or are suspected of having, an autoimmune disease and / or inflammatory disease, such such as rheumatoid arthritis, and in particular those at an early stage of the disease. BACKGROUND OF THE INVENTION Rheumatoid Arthritis (RA) is considered an autoimmune and inflammatory disease. It is the most common form of chronic inflammatory disease of the joints and affects 1-2% of the general population. The disease is characterized by inflammation of the joints, joint pain, and destruction of synovial joints, leading to severe disability and premature mortality. The optimal use of Disease Modifying Antirheumatic Drugs (DMARDs), such as methotrexate, and the incorporation into the therapeutic scheme of RA of new biological agents in the last ten years has dramatically increased the success in the management of RA. In this regard, there is a growing number of evidences that indicate that the destruction of the joints and the functional decline (disability) in RA are improved by early and aggressive therapeutic intervention. The treatment of patients at a stage in which the evolution of joint destruction can still be prevented would be ideal, therefore the need for an unequivocal early diagnosis has become critical (Aletaha D et al, Arthritis Rheum 2010, 62 : 2569-2581). The diagnosis of RA is still based on specific clinical parameters, such as the number of affected joints or duration of symptoms. However, the early clinical presentation of RA is initially indistinguishable from other forms of arthritis (Rooy DP et al, Rheumatology 2011, 50: 93-100). Analytes or biomarkers such as erythrocyte sedimentation rate (ESR), C-Reactive Protein (CRP) concentration levels), Rheumatoid Factor (EP0175270) and antibodies anti-MBL (Mannose Binding Lectin) (Gupta B et al, J Autoimmun 2006,, 27: 125-133, Afzal N et al, Clin Lab 2011, 57: 895-899, Takizawa Y et al, Ann Rheum Dis 2006, 65: 1013-1020, Vossenaar ER et al, Arthritis Res Ther 2004, 6: R142-R150) have been used with limited diagnostic value. Current tests for monitoring disease progression and response to RA treatment also have sensitivity limitations (González-Quintela A et al, Clin Exp Immunol 2007, 151: 42-50). It has been seen that antigens such as citrullinated vimentin, type II collagen, fibrinogen and alpha enolase produce high levels of autoantibodies (ACPA) that appear in the serum of patients with established disease (Ohmura K et al, Rheumatology 2010, 49: 22982304, Vossenaar ER et al, Arthritis Res Ther 2004, 6: 107-111). These antibodies against citrullinated peptides / proteins (ACPA) are not only highly specific for RA, but also appear early in the disease process when diagnosis is more difficult and intervention more effective. However, there is a subset of the negative population for this biomarker, about 1/4 (Aletaha D et al, Arthritis Rheum 2010, 62: 2569-2581). Specifically, the sensitivity of these antibodies is 80% in patients with established RA, 55% in early RA and 40% in very early RA (Nicaise Roland P et al, Arthritis Res Ther 2008; 10 (6): R142 , Ohmura K et al, Rheumatology 2010, 49: 2298-2304). Recent research on ACPAs is revealing information about the specificity of antigens that initiate or perpetuate autoimmune inflammatory reactions before the onset of the disease. For example, post-translational modifications of the own proteins that participate in breaking the tolerance of T and B cells have been described (Reynisdóttir G et al, Arthritis Rheum 2014, 66: 31-9). It has also been shown that ACPA immunity it begins outside the joints, with the lungs, lymph nodes and gingival tissue being the identified tissues in which the pathogenesis of RA begins (Catrina AI et al, Nat Rev Rheumatol 2014, 10: 645-53; Demoruelle MK et al, Curr Opin Rheumatol 2014, 26: 64-71). It has been described that ACPA levels are higher in synovial fluid compared to serum, which already demonstrates a dominant synovial manifestation (Aletaha D et al Arthritis Rheum 2010, 62: 2569-2581, Snir O et al, Arthritis Rheum 2010, 62: 44-52). In this context, researchers are now looking for autoantigens with high titers of autoantibodies preferably in the serum in an attempt to get closer to the pathogenesis of RA and obtain tools for early diagnosis (Reynisdóttir G et al, Arthritis Rheum, 2014 66: 31-9, Catrina AI et al, Nat Rev Rheumatol 2014, 10: 645-53, Snir O et al, 2010 Arthritis Rheum 62, 44-52, Trouw LA et al, Autoimmun Rev 2012, 12 : 318-22. Human dipeptidyl peptidase IV (DPP-IV / CD26), a type II membrane glycoprotein with intrinsic DPP-IV activity, is a key molecule in immune regulation and autoimmune diseases (Hosono et al, Modern Rheumatology 2003, 13, 199-204). It has been described that DPP IV purified from human serum lacks the transmembrane domain of the membrane-bound enzyme (Iwaki-Egawa S et al, Jap J Biochem 1998; 124: 428-33), however it is not affected its enzymatic activity or its ability to bind adenosine deaminase (ADA). DPP-IV activity is a contributing factor to the pathogenesis of rheumatoid arthritis (Busso N et al, Am J Pathol 2005, 166: 433-42, Sedo A et al, Arthritis Res Ther 2005, 7: 25369). As a result of its X-Pro N-terminal dipeptide cleavage activity, it can regulate chemotactic responses to various inflammatory chemokines, including SDF-1, to several biologically active peptides, such as NPY and VIP, recently implicated in RA, in addition of being a neutrophil chemo repellent (Herlihy SE et al, Arthritis Rheumatol 2015, 67: 2634-8, Cordero OJ et al, PLoS One 2015, 10: e0131992). Several authors, including the inventors, (Cordero OJ et al, PLoS One 2015, 10: e0131992, Cordero OJ et al, Rheumatol Int 2001, 21: 69-74, Muscat C et al, Clin Exp Immunol1994, 98: 252- 6, Ellingsen T et al, Scand J Immunol 2007, 66: 451-7) have observed reduced levels of sCD26 and DPP-IV activity in patients with RA and have found significant correlations between CD26 expression in some subtypes of T cells with disease activity (in the form of the DAS28 indicator) and serum levels of sCD26 (Cordero et al, PLoS One 2015, 10: e0131992). Barr et al predicted the existence of autoantibodies against human serum DPP-IV (sCD26) (Barr VA et al 1995. J Biol Chem. 270: 27834-44), which were first described by Cuchacovich et al (Clin Exp Rheumatol 2001 , 19: 673-80). Cuchacovich et al determined the mean serum levels of DPP-IV, the specific activity of DPP-IV and the levels of anti-CD26 antibodies of the IgG, IgM and IgA isotypes in patients suffering from autoimmune and inflammatory diseases, specifically in RA , Systemic Lupus Erythematosus (SLE) and Primary Sjögren's Syndrome (SS). They found that serum DPP IV levels of patients with RA were similar to those of normal controls; however, they were reduced in the serum of patients with SLE and SS when compared to normal controls. Interestingly, while the concentration of DPP IV in the serum of patients with RA It seemed normal, its specific activity was significantly reduced. In addition, it was investigated whether anti-DPP IV antibodies can modify the activity of the enzyme. The results showed that serum levels of anti-DPP IV autoantibodies of the IgA class were significantly higher in the sera of RA, SLE and patients with SS when compared with those of normal controls. The serum levels of anti-DPP IV autoantibodies of the IgG and IgM classes in the three groups of patients were found not to be significantly different from those of the normal controls, whereby Cuchacovich et al concluded that only the anti-autoantibodies DPP IV of the IgA class may have some relevance in the immune response against DPP IV. US2006 / 0177886 refers to a method for the diagnosis of RA which comprises measuring DPP-IV activity in a biological sample obtained from a mammal. In particular, a statistically significant (about 5 times higher) increase in DPP-IV activity was reported in patients with RA compared to patients with osteoarthritis (OA). No correlation was observed between DPP-IV activity in RA and the severity of the disease. The detection and early diagnosis of RA has been associated with a more positive result of therapy and is vital to establish a correct evidence-based treatment, according to the state of disease progression. From the doctor's perspective, early detection, accurate diagnosis and classification of the disease based on its degree of development would have potential benefits with respect to patient orientation, assessment of disease prognosis, monitoring and progression of disease relapses, and in particular with the choice of the most appropriate treatment. Despite the significant progress that has been made in recent years in the discovery of molecular and serological markers related to RA, there is a continuing need for improved methods for early detection, accurate diagnosis, classification, study of progression and / or prognosis of RA and autoimmune and related inflammatory diseases. SUMMARY OF THE INVENTION The levels of IgA, IgM and anti-CD26 IgG were measured in a cohort of patients with RA who were subjected to different biological and non-biological therapies (Example 3), and for which the disease activity was previously assessed by the inventors. as published in Cordero OJ et al, 2015. In the new data generated, higher levels of anti-CD26 (Ab) autoantibody, about twice for each isotype, were found in that cohort with respect to healthy donors, while that the quotients with the respective titles of Total Ig were different for each isotype. More specifically, it was found that levels of anti-CD26 IgA, IgM and IgG and anti-CD26 / total IgG isotype antibody ratios were statistically significantly different compared to healthy donors (Example 3.1). Based on this finding, the discriminatory value of anti-CD26 antibodies was determined between patients with RA (undergoing different therapies) and healthy donors throughout the cohort, where IgG titres showed a sensitivity of 82% and a specificity of 96 % with the cut points selected (see Example 4.1). In addition, the detection / diagnosis value was tested in the group undergoing DMARD but without biological therapy (noBT), which would be more like a group of patients diagnosed and / or patients closer to pre-AR, in the sense that These therapies do not change the expression of CD26. A sensitivity of 77% was found for the anti-CD26 IgG isotype and a sensitivity of 73% for the anti-CD26 IgM (Example 4.2). Only 2 of the 22 patients were negative for all three isotypes. These results point to the usefulness of these markers in the detection or diagnosis of RA. In addition, correlations of the levels of anti-CD26 were found with disease activity parameters (Example 6), in particular the anti-CD26 antibodies of the three isotypes are negatively correlated with the Inflamed Joints Count (TJC), which suggests a specific relationship of these autoantibodies with the joints. Thus, according to these particular findings, the present invention provides in a first aspect an in vitro method for screening a subject who has, or is suspected of having an autoimmune and / or inflammatory disease comprising the following steps: a) the determination of the levels of anti-CD26 IgG and / or anti-CD26 IgM antibodies in a isolated sample of said subject containing the antibody; b) compare the levels in said sample containing antibodies with a value of reference. In a related aspect, the invention relates to an in vitro method for obtaining useful data for the diagnosis of an autoimmune and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect. In a more extensive related aspect, the invention relates to an in vitro method for the diagnosis of an autoimmune and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect. In a further related aspect, the invention relates to an in vitro method for classifying subjects as i) healthy subjects or as ii) subjects who have, or are suspected of having an autoimmune and / or inflammatory disease, said method comprising stages a) and b) of the first aspect. In another aspect, the invention relates to an in vitro method of controlling the disease and / or monitoring the ability to respond to a treatment in a subject who has or is suspected of having an autoimmune and / or inflammatory disease, said said comprising. method steps a) and b) of the first aspect of the invention. In a related aspect, the invention relates to an in vitro method for classifying a subject as a respondent to a treatment for an autoimmune and / or inflammatory disease, said method comprising steps a) and b) of the first aspect of the invention. In a further aspect, the present invention relates to a method of treating a subject having an autoimmune and / or inflammatory disease, said method comprising identifying the subject to be treated by a method comprising steps a) and b) of the first aspect of the invention and c) the treatment of said subject who has or is suspected of having an autoimmune disease and / or inflammatory disease. In a further aspect, the invention relates to an in vitro method for determining the stage of progression of an autoimmune and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect of the invention. In a related aspect, the invention relates to an in vitro method of determining a treatment for a subject having an autoimmune and / or inflammatory disease in accordance with its progression stage comprising steps a) and b) of the first aspect of the invention. In a related aspect, the invention relates to the use of in vitro levels of anti-CD26 IgG and / or anti-CD26 IgM and / or anti-CD26 levels of antibody IgG / total IgG levels, optionally in combination with the levels of IgA anti-CD26 antibodies determined in vitro, in a sample, isolated from a subject, containing antibody for the detection, diagnosis and / or monitoring of an autoimmune and / or inflammatory disease; and / or to control the efficacy of a treatment in an autoimmune and / or inflammatory disease. In a further aspect, the invention relates to a kit comprising reagents for the determination of the levels of anti-CD26-antibodies of the IgG isotype in a sample containing antibody, isolated from a subject, in which said kit comprises: a) a reagent to determine the levels of anti-CD26 antibodies of the IgG isotype; b) optionally, instructions for the use of said reagent in determining the levels of anti-CD26 antibodies of the IgG isotype in a sample, isolated from said subject, containing antibody. In another aspect, the invention relates to a kit comprising reagents for the determination of the levels of anti-CD26 antibodies of the IgM isotype in a sample containing antibody isolated from a subject, wherein said kit comprises: a) a reagent to determine the levels of anti-CD26 antibodies of the IgM isotype; b) optionally, instructions for the use of said reagent in determining the anti-CD26 antibody levels of the IgM isotype in a sample, containing antibody, isolated from said subject. Still in a further aspect, the invention relates to the use of a kit of the preceding aspect for the detection, diagnosis and / or monitoring of an autoimmune and / or inflammatory disease; and / or for monitoring the efficacy of a treatment in a sample, containing antibody, isolated from a subject. In a further aspect, the invention relates to an in vitro method for the detection, obtaining useful data for the diagnosis, diagnosis or monitoring of a subject who suffers, or is suspected of suffering from an autoimmune and / or inflammatory disease comprising the next steps: a) the determination of the total antibody levels of the IgA isotype (total levels of IgA antibodies) in a sample, containing antibodies, isolated from said subject; b) compare the levels in said sample containing antibodies with a reference value. In a further aspect, the present invention relates to a method of treating a subject having an autoimmune and / or inflammatory disease, said method comprising identifying the subject to be treated by a method comprising the steps of: a) the determination of total antibody levels of the IgA isotype (total levels of IgA antibodies) in a sample, containing antibodies, isolated from said subject; b) compare the levels in said sample containing the antibody with a value of reference; and c) the treatment of said subject who has or is suspected of having an autoimmune disease and / or inflammatory disease. In a further aspect, the invention relates to a kit comprising reagents for the determination of total levels of IgA isotype antibodies in a sample, containing antibodies, isolated from a subject, in which said kit comprises: a) a reagent to determine the total antibody levels of the IgA isotype; b) optionally, instructions for the use of said reagent in determining the Total levels of IgA antibodies in a sample, containing antibodies, isolated from said subject. Still in a further aspect, the invention relates to the use of a kit for the detection, diagnosis and / or monitoring of an autoimmune and / or inflammatory disease; and / or for monitoring the efficacy of a treatment in a sample, containing antibody, isolated from a subject, in which said kit comprises reagents for the determination of total IgA antibody levels according to the kits described in the previous aspect. . BRIEF DESCRIPTION OF THE FIGURES Fig. 1. Cut-off values of anti-CD26 / DPP-IV antibody levels and frequency of anomalous values in patients with RA of the noBT group (patients under DMARD) and healthy donors (DS ) or control donors. The levels of IgM (A), IgG (B) and IgA are shown (C) in both groups. Cut-off values (20, 2.85 and 1.8 mLg mL-1, respectively). Fig. 2. Correlations between the parameters of RA disease activity and the levels of anti-CD26 IgA / DPP-IV autoantibodies in the anti-TNF group. In (A) correlations of the IgA isotype with the TJC (inflamed joint counts); in (B) correlations of the IgA isotype with the DAS28 score. Fig. 3. Correlations between the parameters of AR disease activity and the anti-CD26 IgG / DPP-IV autoantibody titers in the anti-CD20 group. In (A) correlations of the IgG isotype with the TJC; in (B) with the SJC (damaged joints); in (C) with the DAS28 correlations of the IgA isotype with the DAS28 score; and in (D) with the PGA (subjective component of DAS28). Fig. 4. The anti-CD26 / DPP-IV autoantibodies are not ACPA. In (A) the supernatants recovered from the ACPA test after the incubation of serum from 3 healthy donors (DS) and patients were then used for the anti-CD26 test. The same absorbance values were observed when serum samples were used directly in this anti-CD26 test (not shown). In (B), the dark gray bars show the absorbance values of the ACPA test performed on the serum of the same individuals (a control was also positive) and the light gray bars show the absorbance values of the ACPA tests taken to carried out on the supernatants recovered from the anti-CD26 test of the same individuals; Differences were observed in a patient with RA (3rd) and in a healthy donor (1st). Background noise values have already been extracted from the absorbance data shown. Fig. 5. The correlations between the levels of anti-CD26 / DPP-IV autoantibodies and the total Ig levels of the three isotypes (A, G or M), or their ratios (autoantibodies / total Ig) with the serum concentration of sCD26 and the enzymatic activity of DPP-IV in the cohort of patients with RA in different therapies. (A) Patients under DMARD (without BT therapy), (B), (C) and (D) patients under biological therapy (BT): anti-TNF, anti-CD20 and anti-IL6R / Ig-CTLA4, respectively. sCD26 refers to the concentration of DPP-IV. The numbers represent the Pearson coefficient, r, the asterisks are for statistical significance, * p <0.05, ** p <0.005). Only in the anti-TNF group there were some coincidences. Fig. 6. The effects of treatments on antibody levels are observed in the ratios between the Ig A, G or M isotypes. Columns 1-4 refer to samples from patients who have received DMARD; anti-TNF-; anti-CD20; and anti-IL6R / Ig-CTLA4 respectively. In many cases, the effects on the ratios of the autoantibody isotypes (lower row) were the opposite of that observed in the total antibody isotype coefficients (upper row), demonstrating the specificity of anti-CD26 anti-autoantibodies. Fig. 7. ROC curve (Receiver Operating Characteristic) for anti-CD26 IgM throughout the cohort. Area under the curve (AUC) = 0.748, the X axis represents 1 -specificity and the Y axis represents the sensitivity. Fig. 8. ROC curve for anti-CD26 IgG throughout the cohort. Area under the curve (AUC) = 0.881, the X axis represents 1 -specificity and the Y axis represents the sensitivity. Fig. 9. ROC curve for IgA anti-CD26 throughout the cohort. Area under the curve (AUC) = 0.661, the X axis represents 1 - specificity and the Y axis represents the sensitivity. Fig. 10. ROC curve for anti-CD26 IgG / total IgG throughout the cohort. Area under the curve (AUC) = 0.741, the X axis represents 1 - specificity and the Y axis represents the sensitivity. Fig. 11. ROC curve for anti-CD26 IgM in patients undergoing DMARD but without biological therapy. Area under the curve (AUC) = 0.751, the X axis represents 1 -specificity and the Y axis represents the sensitivity. Fig. 12. ROC curve for IgG anti-CD26 in patients undergoing DMARD but without biological therapy. Area under the curve (AUC) = 0.846, the X axis represents 1 - specificity and the Y axis represents the sensitivity. Fig. 13. ROC curve for IgA anti-CD26 in patients undergoing DMARD but without biological therapy. Area under the curve (AUC) = 0.672, X axis 1 -specificity, Y axis sensitivity. Fig. 14. ROC curve for anti-CD26 IgG / total IgG in patients undergoing DMARD but without biological therapy. Area under the curve (AUC) = 0.804, X axis 1 -specificity, Y axis sensitivity. Fig. 15. ROC curve for total IgA throughout the cohort. Area under the curve (AUC) = 0.979, X axis 1 -specificity, Y axis sensitivity. Fig. 16. ROC curve for total IgA in patients undergoing DMARD but not biological therapy. Area under the curve (AUC) = 0.94, the X axis represents 1 - specificity and the Y axis represents the sensitivity. DETAILED DESCRIPTION OF THE INVENTION Definitions The term "screening" (in English "screening") is understood here as the examination or analysis of a group of asymptomatic individuals belonging to the general population, or a group of individuals suspected of suffering from a disease, with the aim of discriminating healthy individuals from those who have or are suspected of having a disease. The term "subject who has a disease", as used herein, refers to those subjects diagnosed for a given disease. The term "diagnosis" as used herein refers to determining the presence or absence of a disease in a subject suspected of having the disease. A "diagnostic test" could be used to suggest or rule out the disease. The term "subject suspected of having a disease", as used herein, refers to a subject who has one or more signs or symptoms indicative of said disease. A subject suspected of having a disease may also have one or more risk factors (that is, a subject that may develop or is at risk of developing a disease). It also includes an individual who has received a preliminary diagnosis, but for whom a confirmation test has not been made. In addition, those individuals in remission are included. The term "subject" as used herein refers to a mammalian subject. Preferably, it is selected from a human being, companion animals, non-domestic farm animals or zoo animals. For example, the selected subject may be a human being, dog, cat, cow, pig, sheep, horse, bear, and so on. In a preferred embodiment, said mammalian subject is a human subject. The term "inflammatory rheumatic disease" as used herein refers to a disease of the musculoskeletal system and / or connective tissues in which inflammatory mechanisms play a relevant role. A rheumatic disease can affect the joints, bones, cartilage, tendons, ligaments and muscles. The term "progression phase" of an inflammatory rheumatic disease may refer to a stage of disease progression in which there is no irreversible damage (for example, there is no irreversible damage to the joints). The term "early rheumatoid arthritis", "early RA" or "eRA", as used herein may refer to a subject diagnosed with Rheumatoid Arthritis, typically with duration of symptoms of ≤ 6 months who has not received any treatment Frontline for the treatment of RA (i.e. corticosteroid or therapy FAME) and whose conventional radiographs of the hands and feet showed no structural damage. The term "very early rheumatoid arthritis" or "veRA" as used herein may refer to a subject diagnosed with early RA with symptom duration of ≤ 3 months. More details on the classification of patients with RA are provided in the ACR / EULAR classification criteria (Ann Rheum Dis 2010; 69: 1580-1588). The term "control" (also referred to as "monitoring" or "monitoring") of the disease, "as used herein, refers to the determination of disease progression, for example determining whether there is a disease remission, is that is, a reduction of the signs and / or symptoms that characterize the clinical disease; or on the contrary if there is disease progression or relapse. The term "response to a treatment" as used herein, refers to the degree to which a treatment achieves the desired or expected results, for example, the ability of a drug to achieve the desired prophylactic and / or therapeutic effect. The term "treatment" encompasses both a prophylactic or therapeutic treatment. The term "therapeutic treatment" or "therapy" as used herein refers to bringing a body from a pathological state or disease to its normal and healthy state. The term "prophylactic treatment" as used herein refers to the prevention of a pathological state. The term "sample containing antibody" or "biological sample containing antibody" as used herein includes biological fluids, such as whole blood, serum, plasma, synovial fluid, cerebrospinal fluid, bronchial lavage, ascites fluid, bone marrow aspirate , pleural effusion, urine, as well as any tissue or any other body constituent that could contain antibodies or cells expressing them. Cells that express and secrete antibodies (B cells) are present in, and where appropriate, can be isolated from different tissues, organs and biological fluids of individuals. B cells can be found in the central or primary lymphoid organs that generate lymphocytes of immature progenitor cells, such as the thymus and bone marrow. Alternatively, B cells can be found in the secondary lymphoid tissue that provides the environment for the antigen to interact with lymphocytes, such as lymph nodes, lymphoid follicles in the tonsils, Peyer's plaques, the spleen, the adenoids, the skin, etc. Which are associated with the lymphoid tissue associated with the mucosa. It is observed that the mononuclear cells in the Spleen contain a higher percentage of IgG secretory cells. B cells can be found in biological fluids such as blood, cerebrospinal, synovial or pleural fluids. Alternatively, lymphocytes can also be found at sites of chronic infection and / or inflammation. The term "reference value" as used herein refers to a certain value in one or more samples, in general, the average values of, preferably, a statistically significant number of samples, obtained from a reference population or control. The determination of the "reference value" is carried out under similar or identical conditions as the biological sample of the subject under test. Typically, said reference samples are of the same type of tissue or cell and have undergone the same procedures for obtaining and preserving them. Alternatively, said "reference values" are preset values, in general, the average values, in the control or reference population. The term "kit" or "test kit" refers to combinations of reagents and additives necessary for an analysis. Although a test kit is usually recorded in most cases of several units, the one-piece analysis elements are also valid and should also be considered as test kits. The term "partially identical" as used herein refers to a sequence that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to a reference sequence. In some embodiments, the partially identical sequence may be substantially identical to a reference sequence, which is at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to said reference sequence. In one embodiment, the partially identical sequence is 100% identical to a reference sequence. The term "identity" as used herein refers to an exact nucleotide to nucleotide or amino acid to amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more sequences (polynucleotide or amino acids) can be compared by determining their "percent identity." The "percent identity" of two sequences, whether nucleic acid or amino acid sequences, is the number of matches exact between two aligned sequences divided by the length of the shortest sequence and multiplied by 100. Suitable programs for calculating the percentage of identity or similarity between sequences are well known in the art, such as the NCBI BLAST program, which is used for example, with the default parameters (http: // www. ncbi. NLM. gov / cgi-bin / BLAST). The term "Receiver Characteristic Curves (ROC)" as used herein refers to a graphic representation that illustrates the operation of a binary classifier system in which the mobility of the threshold value to be considered can be varied. The curve is created by plotting the true positive rate versus the false positive rate in various threshold settings. The true positive rate is also known as sensitivity. The false positive rate is calculated as 1 -specificity. The ROC curve is therefore a way to graphically show the true positive rate versus the false positive rate (sensitivity to (1-specificity)) through a range of cut-off values and to select the optimum cut-off point for clinical use The precision that is expressed as the area under the ROC curve (AUC) provides a useful parameter to compare the performance of the assay. If AUC approaches 1 it indicates that the test is very sensitive as well as highly specific, while an AUC that approaches 0.5 indicates that the test is neither sensitive nor specific. In general, a test is considered to be an adequate discriminator if the AUC is 0.6 to 0.75, to have a high discrimination capacity if the AUC is 0.75 to 0.9 and to be an excellent discriminator if the AUC is 0.9 to 1. To more details, see for example, Zweig MH and Campbell G, Clin Chem 1993; 39: 561-577, or Greiner et al, Prev Vet Medicine 2000; 45: 23-41. DETAILED DESCRIPTION An in vitro method for screening a subject who has, or is suspected of having an autoimmune and / or inflammatory disease and related methods In a first aspect, the present invention relates to an in vitro method for the screening of a subject who has, or is suspected of having an autoimmune and / or inflammatory disease comprising the following steps: a) the determination of the levels of anti-CD26 IgG and / or anti-CD26 IgM antibodies in a isolated sample of said subject containing antibodies. b) compare the levels in said sample containing antibodies with a value of reference. In a related aspect, the invention relates to an in vitro method for obtaining useful data for the diagnosis of an autoimmune and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect. In a further related aspect, the invention relates to an in vitro method for the diagnosis of an autoimmune and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect. In a further related aspect, the invention relates to an in vitro method for classifying subjects as i) healthy subjects or as ii) subjects who have, or are suspected of having an autoimmune and / or inflammatory disease, said method comprising stages a) and b) of the first aspect. The first aspect and the related aspects described above herein can be collectively referred to as the first aspect of the invention. The term "inflammatory disease" means any disease in which there is an excessive or altered inflammatory response that leads to inflammatory symptoms. Such inflammatory diseases include, without limitation, Addison's disease, acne vulgaris, alopecia areata, amyloidosis, ulcerations, aphthous stomatitis, arteriosclerosis, arthritis, osteoarthritis, rheumatoid arthritis, idiopathic arthritis, psoriatic arthritis, ankylosing spondylitis, bronchial asthma, Bechet's disease , Boeck's disease, Inflammatory Bowel Disease, Crohn's disease, choroiditis, ulcerative colitis, celiac disease, cryoglobulinemia, macular degeneration, dermatitis, herpetiform dermatitis, dermatomyositis, insulin-dependent diabetes, juvenile diabetes, inflammatory demyelinating disease, Dupuytren's contracture, encephalomyelitis , allergic encephalomyelitis, endophthalmia, allergic enteritis, autoimmune enteropathy syndrome, leprosy erythema, idiopathic facial paralysis, chronic fatigue syndrome, rheumatic fever, cystic fibrosis, gingivitis, glomerulonephritis, Goodpasture syndrome, syndrome of Graves, Hashimoto's disease, chronic hepatitis, histiocytosis, regional ileitis, iritis, disseminated lupus erythematosus, systemic lupus erythematosus, cutaneous lupus erythematosus, lymphogranuloma, infectious mononucleosis, myasthenia gravis, transverse myelitis, idiopathic myxedema, primary obesity nephritis, primary obesity, nephtrosis, primary obesity , granulomatous orchitis, pancreatitis, panniculitis, pemphigus vulgaris, periodontitis, polyarteritis nodosa, chronic polyarthritis, polymyositis, acute polyradiculitis, psoriasis, chronic obstructive pulmonary disease, purple, gangrenous pyoderma, Reiter's syndrome, diabetic retinopathy, rosacea, rosacea, rosacea , progressive systemic sclerosis, scleritis, scleroderma, multiple sclerosis, sclerosis disseminated, acute anterior uveitis, vitiligo, Whipple's disease, and AIDS-associated diseases, severe combined immunodeficiency and / Epstein Barr virus, such as Sjogren's syndrome, osteoarticular tuberculosis, and parasitic diseases such as leishmaniasis. The term "autoimmune disease" as used herein refers to a non-malignant disease or disorder that arises from and directed against an individual's own tissues. Autoimmune diseases in this document specifically exclude diseases. or malignant or cancerous conditions, especially excluding B-cell lymphoma, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (LLC), hair cell leukemia and chronic myeloblastic leukemia. Examples of autoimmune diseases or disorders include, but are not limited to, inflammatory responses such as inflammatory skin diseases including psoriasis and dermatitis (eg, atopic dermatitis); scleroderma and systemic sclerosis; responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome; ARDS); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions such as eczema and asthma and other conditions that involve T-cell infiltration and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus (for example, type I diabetes mellitus or insulin dependent diabetes mellitus mellitus); multiple sclerosis; Reynaud's syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjorgen syndrome; juvenile onset diabetes; and the immune responses associated with acute delayed hypersensitivity mediated by cytokines and T lymphocytes are typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison's disease); diseases involving leukocyte diapédesis; inflammatory disorder of the central nervous system (CNS); multiple organ injury syndrome; hemolytic anemia (including, but not limited to cryoglobinemia or Coombs positive anemia); myasthenia gravis; diseases mediated by antigen-antibody complexes; anti-glomerular basement membrane disease; antiphospholipid syndrome; allergic neuritis; Graves disease; Myasthenic Lambert-Eaton syndrome; bullous pemphigoid; pemphigus; autoimmune polyocrinopathies; Reiter's disease; rigid man syndrome; Behcet's disease; giant cell arteritis; immune complex nephritis; IgA nephropathy; IgM polyneuropathies; immune thrombocytopenic purpura (ITP) or autoimmune thrombocytopenia, etc. In a particular embodiment, said autoimmune and / or inflammatory disease is an inflammatory rheumatic disease. Preferably, said inflammatory rheumatic disease It is selected from the group consisting of arthritis, rheumatoid arthritis, systemic lupus erythematosus, Sjögren's syndrome, juvenile rheumatoid arthritis, psoriatic arthritis and spondyloarthropathy. Elevated levels of TNF are found in the synovial fluid of patients with rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis and ankylosing spondylitis and play an important role in both the pathological inflammation and the destruction of the joints that are characteristic of these diseases. Accordingly, in a preferred embodiment, said inflammatory rheumatic disease is selected from rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. In another embodiment, said autoimmune and / or inflammatory disease is selected from rheumatoid arthritis, systemic lupus erythematosus and Sjögren's syndrome. In a preferred embodiment, said autoimmune and / or inflammatory disease is rheumatoid arthritis. The correlations of anti-CD26 levels with disease activity parameters, in particular with joint sensitivity, have been observed by the inventors (Example 6). It should be noted that, according to Example 6.1, the anti-CD26 levels of the three isotypes appear to be negatively correlated with the painful joint count (TJC) when considering the entire cohort. In addition, the CD26 antigen used in the ELISA is non-citrullinated which suggests that the autoantibodies detected would recognize a non-citrullinated sCD26 form. Taken together, these data may suggest that anti-CD26 antibodies appear at an early stage of the pathogenesis of RA. Accordingly, in a preferred embodiment, said autoimmune and / or inflammatory disease is in a state of early progression, preferably very early. Step (a) of the method according to the first aspect of the invention requires the determination of anti-CD26 antibody levels in a sample, isolated from a subject, containing antibody. The term "CD26" as used herein refers to human CD26 (UniProtKB -P27487), also known as dipeptidyl peptidase IV (DPP-IV). Its enzymatic activity is characterized by the release of an N-terminal dipeptide, Xaa-Yaa- | -Zaa-, from a polypeptide, preferably when Yaa is Pro, provided Zaa is neither Pro nor hydroxyproline. The term "CD26" encompasses 240 kDa of homodimeric type II membrane glycoprotein composed of two 120 kDa subunits. (Mentlein R, Int Citology Journal, 2004, 235: 165-213). Each of the homodimers of the membrane bound form are characterized by a sequence consisting of positions 1 to 766 of SEQ ID NO: 1 and the soluble form or "sCD26" is characterized by a sequence consisting of positions 39 to 766 of SEQ ID NO: 1, as shown in Table 1 below. SEQ ID NO positions: 1 LengthDescriptionUNIPROT identifierSEQ NOID 1 - 766 766Dipeptidyl peptidase 4 membrane formPRO_0000027213SEQ NO: 1ID 39 - 766 728Dipeptidyl peptidase 4 soluble formPRO_0000027214SEQ NO: 2ID SEQ ID NO: 1 (PRO_0000027213) has the following sequence: MKTPWKVLLG LLGAAALVTI ITVPVVLLNK GTDDATADSR KTYTLTDYLK NTYRLKLYSL 60 RWISDHEYLY KQENNILVFN AEYGNSSVFL ENSTFDEFGH SINDYSISPD GQFILLEYNY 120 VKQWRHSYTA SYDIYDLNKR QLITEERIPN NTQWVTWSPV GHKLAYVWNN DIYVKIEPNL 180 PSYRITWTGK EDIIYNGITD WVYEEEVFSA YSALWWSPNG TFLAYAQFND TEVPLIEYSF 240 YSDESLQYPK TVRVPYPKAG AVNPTVKFFV VNTDSLSSVT NATSIQITAP ASMLIGDHYL 300 CDVTWATQER ISLQWLRRIQ NYSVMDICDY DESSGRWNCL VARQHIEMST TGWVGRFRPS 360 EPHFTLDGNS FYKIISNEEG YRHICYFQID KKDCTFITKG TWEVIGIEAL TSDYLYYISN 420 EYKGMPGGRN LYKIQLSDYT KVTCLSCELN PERCQYYSVS FSKEAKYYQL RCSGPGLPLY 480 TLHSSVNDKG LRVLEDNSAL DKMLQNVQMP SKKLDFIILN ETKFWYQMIL PPHFDKSKKY 540 PLLLDVYAGP CSQKADTVFR LNWATYLAST ENIIVASFDG RGSGYQGDKI MHAINRRLGT 600 FEVEDQIEAA RQFSKMGFVD NKRIAIWGWS YGGYVTSMVL GSGSGVFKCG IAVAPVSRWE 660 YYDSVYTERY MGLPTPEDNL DHYRNSTVMS RAENFKQVEY LLIHGTADDN VHFQQSAQIS 720 KALVDVGVDF QAMWYTDEDH GIASSTAHQH IYTHMSHFIK QCFSLP 766 The term "CD26" may also encompass partially identical sequences, preferably substantially identical to SEQ ID NO: 1 or SEQ ID NO: 2. These are collectively known as "CD26 variants" and can be obtained by modifying SEQ ID NO: one or SEQ ID NO: 2 by substitution, deletion and / or addition. Suppression variants of CD26 and antigenic fragments of CD26 are characterized by containing at least one antigenic region or epitope. Illustrative, non-limiting examples of CD26 epitopes are those defined by the binding of known anti-CD26 mAbs as described above, such as 4G8, 11H9, 1F7, 10F8A, 12E3B, 14D10, Tal, 5F8, 2F9, TA5. 9, 18H3A, 16D4B, 9C11 (see anti-CD26 mAb and its respective binding to CD26 in Figure 1 of Dong RP, et al, Mol Immunol 1998, 35: 13-21) and anti-FLAG, 22C3, DS2-7 , TA5.9, 1F7, 202-36, L272, Та.1, BA5, CB.1, A3, A10, B10, F11 and C3 (see anti-CD26 mAb and its respective binding to CD26 variants in Table 1 of Hühn J et al, Cell Immunol 1999; 192: 33-40, or Torimoto Y et al, Mol Immunol 1992; 29: 183-92, or Granados et al, Nat Commun 2014; 5: 3600) have recently described positions of amino acids 145-154 of human CD26 as an associated MHC-I peptide (MIP) recognized by CD8 T cells. In one embodiment, a variant of CD26 may have one or more amino acid residues. Said additional amino acid residue (s) may be any amino acid, which may be either an L-and / or D-amino acid, of natural origin and otherwise. Preferably, the amino acid is any naturally occurring amino acid such as alanine, cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan or tyrosine. However, the amino acid residue (s) may also be (a) modified (s) or unusual amino acid (s). Examples are 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, 2-aminobutyric acid, 4-aminobutyric acid, 6-aminocaproic acid, 2aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2-aminopimelic acid, 2,4-diaminobutyric acid, desmosin, 2,2'-diaminopimelic acid, 2,3-diaminopropionic acid, Netylglycine, N-ethylasparagine, hydroxylysine, alo-hydroxylysine, 3-hydroxyproloine, 4hydroxyproloine, isodesmosine, alo-isoleucine, N-methylglycine , N-methylisoleucine, 6-Nmetillisine, N-methylvaline, norvaline, norleucine or ornithine. In addition, the amino acid (s) may be subject to modifications such as post-translational modifications. Examples of modifications include acetylation, amidation, blocking, formylation, hydroxylation of γ-carboxyglutamic acid, glycosylation, methylation, phosphorylation, sulfation and citrullination. Preferably, CD26 is not citrullinated. If more than one additional amino acid residue is present in the polypeptide, the amino acid residues may be the same or different from each other. The CD26 polypeptide may be flanked by the C-terminal, N-terminal, or C-and N-terminal amino acid (s). In a particular embodiment of the invention, said CD26 variant is a variant having at least 70%, at least 80%, at least 90%, at least 95%, at least 99% identity with SEQ ID NO: 1 or SEQ ID NO: 2 that is derived by conservative substitutions. Conservative substitutions are those that take place within a family of amino acids that are related in their side chains and chemical properties and are well known to a person skilled in the art. Examples of such families are amino acids with basic side chains, with acidic side chains, with non-polar aliphatic side chains, with non-polar aromatic side chains, with unloaded polar side chains, with small side chains, with large side chains etc. In one embodiment, a conservative substitution is included in the polypeptide. In another embodiment, two conservative substitutions or less are included in the polypeptide. In a further embodiment, three conservative substitutions or less are included in the polypeptide. CD26 can be obtained by recombinant protein production techniques or by purification from human serum. Standard recombinant protein production techniques use prokaryotic or eukaryotic host cells to produce CD26. Suitable host cells include bacteria (for example E. coli), yeasts, insects, or mammalian cells. A number of eukaryotic expression systems are available for the expression of secreted proteins and membrane bound proteins, including those that contain unique post-translational modifications. The most common eukaryotic expression platforms today include yeasts (e.g., Pichia pastoris and Saccharomyces cerevisiae), insect cells (Spodoptera frugiperda or Trichoplusia ni), and mammalian cell systems (including a variety of transformed cell lines and / or genetically modified). Mammalian cell systems include, but are not limited to ape, human, dog and rodent cells. Examples of human cells are cells (WO01 / 38362), MRC-5 (ATCC CCL-171), WI-38, HEK-293 cells (ATCC CRL-1573), HeLa cells (ATCC CCL-75) (ATCC PER. C6 CCL2), and rhesus fetal lung cells (ATCC Cl-160). Examples of non-human primate cells are Vero cells (ATCC CCL81), COS-1 cells (ATCC CRL-1650) or COS-7 cells (ATCC CRL1651). Examples of dog cells are MDCK cells (ATCC CCL-34). Examples of rodent cells are hamster cells, such as BHK21-F, HKCC cells, or CHO cells. Any compatible protein expression system can be used to produce CD26. Suitable expression systems include transgenic animals described in Gene Expression Systems, Academic Press, eds. by Fernández et al., 1999. In addition, any expression vector known in the art can be used. Enzymes can be used to generate proteins of less than full length, by enzymatic proteolysis from the full length protein or from partial length proteins. Chemical synthesis methods, such as solid phase peptide synthesis, can also be used to synthesize CD26. In another particular embodiment, CD26 has been synthesized by any technique known in the art of peptide synthesis. CD26 can be purified by any technique known in the protein purification technique. Examples of these techniques include ion exchange chromatography, hydrophobic interaction chromatography and immunoaffinity methods. In a particular embodiment, CD26 is the soluble form of CD26 (sCD26), including variants thereof. In a preferred embodiment, sCD26 has been obtained by recombinant means and purified from a suitable host cell as described above. In a more preferred embodiment, said recombinant sCD26 has been purified from the NS0 mouse myeloma cell line, such as the commercially available sCD26 from RnD Systems, USA. (1180-SE-010). In another more preferred embodiment, said recombinant sCD26 has been purified from Sf9 cells infected with baculovirus, such as the commercially available sCD26 from Sigma-Aldrich, USA. (D4943). The methods of the present invention can be applied to samples of individuals of both sexes, that is, men or women, and at any age. In some embodiments, said subject has not previously received or is not subjected to any biological treatment against said autoimmune and / or inflammatory disease, preferably has not previously received or is not subjected to any treatment against said autoimmune and / or inflammatory disease. In other embodiments, said subject has previously received or is subjected to a treatment against said autoimmune and / or inflammatory disease, preferably, wherein said treatment is selected from the group consisting of: i. FAME; ii. anti-TNF alpha agents; iii. anti-CD20 agents; Y iv. anti-IL6R agents and / or CTLA4-Fc fusion proteins. The term "sample containing antibody" has been defined above. These types of samples are commonly used in clinical practice and a person skilled in the art will know how to identify the most appropriate means for obtaining and preserving them. Once a sample has been obtained, it can be used fresh, or it can be frozen or stored using the appropriate means (for example, as a sample of paraffin-fixed tissue included in formalin). When quantification is carried out by analysis of immunohistochemistry (IHC), thin sections of the biological sample immobilized on coated slides are typically used. These sections, when derived from a tissue sample included in paraffin, are deparaffinized and preferably treated in a suitable manner to recover the protein to be detected. Detection can be carried out in individual samples or in tissue microarrays. Preferably, this sample containing antibody is whole blood, serum or plasma, more preferably it is serum. The quantification of the antibody levels in step a) of the first aspect of the invention can be expressed in terms of "antibody concentration" and / or "antibody levels". The term "antibody levels" and "antibody concentration" may be used interchangeably herein. The term "antibody concentration" as used herein refers to the total amount of antibody (protein) in solution regardless of function. The antibody concentration can be estimated using a general protein assay or a specific immunoglobulin method. The term "antibody level" as used herein refers to the functional dilution (or working concentration) of an antibody sample that is necessary to achieve a specific minimum level of detection in a given test method. The exact minimum acceptable value is set by the researcher, but is usually defined by reference to a statistically significant relationship of a value above the background noise (or white, or background). Antibody levels are generally considered a specific measure for each assay. Preferably, the determination of antibody levels in step a) of the first aspect of the invention is carried out by a specific immunoglobulin method. Various types of immunoassays are known to a person skilled in the art for the quantification of proteins of interest. These methods are based on the use of affinity reagents, which can be any antibody or ligand that specifically binds to the target protein, and that is preferably labeled for detection. For example, transfer or immunoblotting techniques allow the comparison of the relative abundance of proteins separated by an electrophoretic gel (for example, native proteins by the 3-D structure or proteins denatured by the length of the polypeptide). Immunoblotting techniques use antibodies (or other specific ligands in related techniques) to identify target proteins among a number of unrelated protein species. They involve the identification of the target protein through specific antigen-antibody (or protein-ligand) reactions. Proteins are typically separated by electrophoresis and transferred to a sheet of polymeric material (usually nitrocellulose, nylon, or polyvinylidene difluoride). Dot and slot blot tests are simplified procedures in which protein samples are not separated by electrophoresis but immobilized directly on a membrane. In some embodiments of the invention, the immunoglobulin quantification can be performed by quantifying the cells expressing immunoglobulins (for example B cells that produce anti-CD26 antibodies of a given isotype). This may be possible, for example, by immunohistochemical assays, where proteins are detected directly in cells of a tissue. The role of B cells and autoantibodies in tissue destructive events in autoimmune diseases is being highlighted, and thus increasing interest in identifying the presence and location of autoreactive B cells and autoantibody-secreting plasma cells. For the visualization and analysis of self-reactive B cells, the antigen of interest is selected, produced and purified from native or recombinant sources. Appropriate labeling (eg biotinylation) of the purified antigen and its subsequent use in immunohistochemistry in sections of the tissue under analysis allow the detection of self-reactive B cells and plasma cells. Double staining with specific cell markers or by the presence of intracellular Ig allows the characterization of the cell or the determination of the Ig isotype of the autoantibody produced by individual self-reactive B cells. With this technique, the identification, as well as the quantification of self-reactive cells within the tissues can be performed; It is also possible to analyze the spatial relationship with respect to residual cells or other infiltrating cells of the target organ. For illustrative purposes, a non-limiting example of how this can be accomplished is described in Wharen-Herlenius and Salomonsson (Methods Mol Med 2001, 36: 19-24). Quantification of cells expressing immunoglobulin may also be possible in any type of biological fluids containing antibodies using techniques well known in the art such as flow cytometry or fluorescence activated cell sorter (FACS). A selection and quantification of cells that secrete specific antibody can be performed using one of the many methods described in the literature, usually based on the expression of specific antibody isotypes and / or cell surface markers on their surface. In particular, several technologies for Purification of antibody secreting cells from human samples make use of different means and conditions for positive or negative selection thereof. These cells are more easily and efficiently selected by physical separation from those that express specific cell surface markers for cells that express and secrete antibodies (eg, B cells). Specific protocols can be found in the literature (see Callard R and Kotowicz К "Human B-cell responses to cytokines" in Cytokine Cell Biology: A practical Approach. Balkwill F. (ed.) Oxford University Press, 2000, p.17 -31). Traditionally, the quantification of proteins in solution has been carried out by immunoassays on a solid support. Said immunoassay can be, for example, an enzyme-linked immunosorbent assay (ELISA), a fluorescence-linked immunosorbent assay (FLISA), chemiluminescence immunoassay (CIA), or radioimmunoassay (RIA), multiplied enzyme immunoassay, solid phase radioimmunoassay (SPROA), fluorescence polarization assay (FP), fluorescence resonance energy transfer assay (FRET), time resolved fluorescence resonance energy transfer assay (TR-FRET), plasmon resonance assay Superficial (SPR) They also specifically cover Multiplex and any next-generation version of any of the above, such as sphere-based flow cytometry immunoassays (for example, based on Luminex xMAP technology). In a particular embodiment, said immunoassay is an ELISA assay, an ELISA cell or any multiplex version thereof. Other methods that can be used for protein quantification are techniques based on mass spectrometry (MS) such as liquid chromatography coupled to mass spectrometry (LC / MS), which is described for example in US2010 / 0173786, or in Tandem LC-MS / MS (WO2012 / 155019, US2011 / 0039287, Rauh M, J Chromatogr B Analyt Technol Biomed Life Sci 2012; 59-67) and the use of peptide matrices, proteins or antibodies and multiplex versions of the prior art , as well as the next generation of such techniques and combinations thereof. In a particular embodiment, the levels of anti-sCD26 antibodies present in a suspension sample containing antibodies are determined by an immunoassay in which sCD26 has been coated on a solid phase support. In another embodiment, the levels of anti-CD26 antibodies present in a suspension sample containing antibodies are determined by an immunoassay in which a cell expressing CD26 is on a solid phase support (eg cell-ELISA). A general method for the determination of anti-CD26 antibodies of a selected isotype is described below. sCD26, or any antigenic fragment thereof, is bound to the solid support. It can be adsorbed or chemically coupled to a solid phase support. Any means known in the art for the immobilization of a protein or peptide to a solid support can be used. sCD26 can be either covalently or attached to the solid phase support by techniques such as covalent bonding through an amide or ester bond, or by non-covalent adsorption. It can also be linked by binding pairs such as biotin and avidin or antibody and antigen. After sCD26 is fixed to the solid phase, the solid phase support can be incubated with a blocking solution (containing a blocking protein such as bovine serum albumin) to reduce non-specific adsorption of antibodies in a sample of support surface test. Various solid phase supports can be used, including, but not limited to, glass, polystyrene, polypropylene, nitrocellulose, dextran or other materials. Suitable forms of solid phase supports include beads, microparticles, tubes, tissues or plates formed from, or coated with these materials. In a preferred claim, the solid support comprises microtiter wells, such as a 96-well microtiter plate. A detection antibody is used to evaluate the anti-sCD26 antibody bound to the solid phase support. The detection antibody used for this purpose may be isotype specific, for example, IgA, IgD, IgE, IgG, or IgM. The isotype specific detection antibody can be labeled and detected. Sub-isotypes of the antibody can also be determined. The detection antibody may be labeled. The marker can be any detectable composition. Any means of analysis known in the art can be used to determine or detect the detection antibody. These means include the use of spectroscopy, chemistry, photochemistry, biochemistry, immunochemistry, or optics. The label can be, for example, an enzyme (for example, horseradish peroxidase (HRP), alkaline phosphatase, beta-galactosidase, and others commonly used in an ELISA), a radiolabel (for example, 3H, 125I, 35S, 14C , or 32P), a chemiluminescent compound (e.g., luciferin, and 2,3-dihydroftalazinediones, luminol, etc.), a fluorescent dye (e.g., fluorescein isothiocyanate, Texas red, rhodamine, etc.), or any other dye known in the art. The label can be directly or indirectly coupled (eg, through binding pairs such as biotin and avidin) to the detection of antibodies according to methods well known in the art. As indicated above, a wide variety of labels can be used. The choice of the label may depend on the sensitivity required, the ease of conjugation with the compound, stability requirements, the available instrumentation, or disposal arrangements. Detection antibodies can be detected or quantified by any suitable method known in the art. A marker in an antibody can be detected by a gamma counter if the label is a radioactive gamma emitter, or by a fluorimeter, if the label is a fluorescent material. In a preferred claim, the detection antibody is a species specific immunoglobulin conjugated to HRP. Any known HRP substrate can be used. Non-limiting examples are ABTS (2,2'azinobis [3-ethylbenzothiazoline-6-sulfonic acid] -diammonium salt), OPD (o-phenylenediamine dihydrochloride) and TMB (3,3 ', 5,5'-tetramethylbenzidine). With each of these substrates a reaction product is obtained which can be detected optically with a maximum absorbance at 410 nm, 650 nm, 492 nm or 450 nm wavelength, for the cases mentioned. The test results can be qualitative or quantitative. The amount of marker associated with the support can be compared with the positive and negative controls in order to determine the presence of anti-sCD26 antibodies. The controls are usually executed concomitantly with the sample to be tested. A positive control can be a serum that contains antibodies that are immunoreactive with sCD26. A negative control may be a serum that does not contain antibodies that are immunoreactive with sCD26. For quantification, a standard curve is used using known amounts of antibody of the same isotype, which may, but does not have to be, anti-sCD26. Antibodies for use as positive controls can be produced using fragments of, or the complete sequence, of sCD26 amino acids. "Antibody" as used herein includes intact immunoglobulin molecules, as well as fragments thereof, such as Fab, F (ab ') 2, and Fv, which are capable of binding to an epitope of the target protein (by example, sCD26). Any type of antibody known in the art can be generated to specifically bind an epitope of sCD26. Monoclonal or polyclonal antibodies can be made as is well known in the art. Any available technique can be used for the purification of the antibodies. For example, antibodies can be purified by known methods, such as affinity separation using protein A, high pressure liquid chromatography on reverse phase alkylated silica gel, or gel filtration. Antibodies can also be passed through a solid phase to which sCD26 binds. Anti-sCD26 antibodies will bind to sCD26 bound to the solid support and contaminants can be washed. Bound antibodies can be eluted, for example, with a buffer having a high salt concentration, or low pH. The particular parameters used in the assay for the determination of anti-CD26 antibody levels can vary widely depending on various factors such as the concentration of antibody in the sample, the nature of the sample, the type of immunoassay employed and the like. Optimum conditions can be easily established by those skilled in the art. Typical test conditions include a temperature range of about 4 ° C to about 45 ° C and a range of pH values of about 5 to 9. Incubation times can also vary widely depending on the nature of the assay, and generally vary from about 0.1 minutes to about 24 hours. A wide variety of buffers, for example TRIS buffered saline, or phosphates, can be used, and other reagents such as salt to improve ionic strength, proteins such as serum albumin, stabilizers and non-ionic detergents can also be included. . Exemplary conditions for total immunoglobulins and anti-CD26 autoantibodies of the IgG, IgM and IgA isotypes are given in Example 1. The relationship between levels of IgG anti-CD26 antibodies and total levels of IgG antibodies Alternatively, or in addition to determining the levels of anti-CD26 IgG antibodies, the methods of the invention may include determining the relationship between anti-CD26 levels of antibody IgG and total levels of IgG antibodies (hereinafter referred to as as "anti-CD26 IgG / total IgG ratio"). Total antibody levels of a particular isotype can be determined by an immunoassay on a solid support in which, instead of sCD26 as described above, an anti-human antibody of the corresponding isotype (eg, IgM, IgG or IgA) in general from another animal species is attached to the solid support, for example, an anti-human IgM, anti-IgG or anti-IgA polyclonal rabbit. Consequently, the relationship between IgG anti-CD26 antibody levels and levelsof total IgG antibodies (ie, the "anti-CD26 IgG ratio") is obtained herein.document dividing the anti-CD26 levels of IgG antibodies by the total levels ofIgG antibodies. Accordingly, in another particular embodiment, the methods of the invention comprise theNext steps:a) determine the levels of anti-CD26 IgG antibodies and / or the ratio of anti-CD26 IgG inan isolated sample of said subject containing antibody;b) compare the level and / or the ratio in said sample containing antibody with a valuereference. In a further particular embodiment, the methods of the invention comprise theNext steps:a) determine the levels of anti-CD26 IgM antibodies and / or the ratio of anti-CD26 IgG ina sample containing antibody isolated from said subject;b) compare the level and / or the ratio in said sample containing antibody with a valuereference. In a preferred embodiment, the method according to the first aspect of the invention, andAny related aspect as described above, comprises the followingstages:a) the determination of IgG anti-CD26 antibody levels in a sample thatcontains antibody isolated from said subject;b) compare the levels in said sample containing antibody with a reference value. In step b), the levels and / or ratio determined in said biological sample are comparedWith reference values. A reference value represents a threshold value of the relevant biomarker (for example,anti-CD26 IgG antibody levels) to which levels can be comparedMeasured of a subject. The reference level can be selected based on the purposeplanned of the test or trial to be performed. For example, a reference levelcan be determined for the purposes of identifying subjects affected by aautoimmune and / or inflammatory disease, while the same or different level of Reference can be used to monitor the progression or severity of the disease in a subject or the ability to respond to a treatment. The selection of a suitable reference level for a given test or trial is within the level of experience in the art. The choice of the reference value is not absolute. For example, a relatively low value can be used advantageously to reduce the incidence of false negatives, but it can also increase the likelihood of false positives. Consequently, as for early detection, diagnosis or monitoring techniques, the reference value may be based on a number of factors, including but not limited to, costs, the benefit of early diagnosis and treatment, the invasiveness of diagnostic and follow-up methods for people who have false positive results, the intended purpose of the test or test, the limits of precise detection in the particular type of sample, or the prevalence and mean of the biomarker given in the relevant population, the Desired level of statistical significance of the results, the level of quantification desired in terms of disease risk, presence, progression, or severity, and other factors that are routinely considered in the design of screening trials. The threshold value or cut level can be determined by use in the methods of the invention. The term "reference value" as used herein also encompasses a threshold or cut-off point. A variety of statistical and mathematical methods for setting the threshold or cutoff level for a particular biomarker are known in the prior art and can be selected, for example, based on ROC curve data. One skilled in the art will appreciate that this threshold or cut-off point of expression levels can be varied, for example, by moving along the ROC chart for a biomarker, or particular combinations thereof, to obtain different values for sensitivity. or specificity without affecting the overall test performance. The best cut-off point refers to the value obtained from the ROC line for a particular biomarker that produces the best sensitivity and specificity. Sensitivity and specificity values are calculated over the threshold range (cut-off). Thus, the threshold cut-off values or can be selected such that the sensitivity and / or specificity are at least about 70%, and can be, for example, at least 75%, at least 80%, at least 85% at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or at least 100% in at least 60% of the population of patients tested, or at least minus 65%, 70%, 75%, 80%, 85%, 90%, 95% or, preferably, 100% of the population tested. In some embodiments of the methods in the first and related aspects of the invention, said reference value corresponds to the respective levels of anti-CD26 antibodies, or the value of the mentioned ratio, in healthy subjects. The term "healthy subjects" as used herein refers to an appropriate unaffected population. In other embodiments of the methods in the first and related aspects of the invention, said disease is an inflammatory rheumatic disease and said reference value corresponds to the value of the respective levels of anti-CD26 antibodies, or the ratio, of a rheumatic disease that It is not an inflammatory rheumatic disease, such as osteoarthritis. When comparing the subject's levels with said reference or control value (for example, the mean or median of the levels or a cut-off value), the degree of variation of the levels in the samples examined with respect to a value of reference may be, for example, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least one 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, or more. Alternatively, or in addition, subjects having more than about 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 8, 10 or 20 times higher biomarker concentration than the reference value (for example, the mean or the median levels or a cut-off value) for an appropriate unaffected population as measured under similar or identical conditions can be identified as having, or suspected to have, an autoimmune disease and / or inflammatory disease. Preferably, this variation is statistically significant. The term "significant" or "statistically significant" when referring to the differences between the test sample values and the control or reference value, refers to the condition, when the appropriate statistical analysis is used, of the probability of that it is less than 5%, eg p <0.05. In other words, the probability of obtaining the same results on a completely random basis is less than 5 out of 100 attempts. One skilled in the art will know how to choose the appropriate statistical analysis. In general, the appropriate statistical analysis is determined based on whether the variable under study has a normal distribution, for example, by using the Kolmogorov-Smirnov test and whether there is homocedasticity, which is determined, for example, by the Levene test. Preferably, in cases where there is a normal distribution and homocedasticity, a parametric model such as t-test or the ANOVA test is used; and where at least one of these two requirements is not carried out then a non-parametric model such as the Mann-Whitney test or the Kruskal-Wallis test is generally used to compare continuous (uncategorized) variables. For the categorized variables, the Chi-square test or Fisher's exact test are normally carried out. Dunnett's t-test is normally used for multiple comparisons in a single cohort. In preferred embodiments of the methods of the invention, an increase in the levels of relevant anti-CD26 antibodies as defined above (eg levels of IgG anti-CD26 antibody and / or the ratio of anti-CD26 IgG / total IgG and / or levels of anti-CD26 IgM antibodies, and similar ratio) in the subject sample, in relation to the reference value, is indicative of disease. Preferably, wherein said reference value corresponds to the respective reference value in healthy subjects. The methods under the first aspect and related aspects as defined above may further comprise the determination of the levels of anti-CD26 antibodies of the IgA isotype in said antibody-containing sample. Accordingly, the levels of anti-CD26 antibodies, in some embodiments of the invention are selected from the group consisting of: i. anti-CD26 IgG antibody levels and / or the anti-CD26 / total IgG ratio; ii. anti-CD26 IgM antibody levels; iii. the levels of IgG and / or the quotient cited in i) and the levels of anti-CD26 IgM antibodies; iv. the levels of IgG and / or the quotient cited in i) and the levels of anti-CD26 antibodies of IgA; Y v. the levels of IgG and / or the quotient cited in i) and the anti-CD26 IgM, and the levels of anti-CD26 antibodies of IgA. wherein, for each of the defined groups, an increase of any and preferably of all antibody levels or the quotient in the subject's sample, with respect to said reference value, is indicative of disease and in which said reference value corresponds to the respective reference value in healthy subjects. In a preferred embodiment, said method comprises: a) the determination of the levels of anti-CD26 isotype IgG antibodies in a sample ofcontaining the antibody;b) comparison of levels of anti-CD26 isotype IgG antibodies in a samplecontaining the antibody with a reference value; Y It also includes: c) the determination of the levels of anti-CD26 antibodies IgM and / or IgA isotypes in asample containing these antibodies;d) comparison of the levels of anti-CD26 antibodies IgM and / or IgA isotypes in a samplecontaining these antibodies with a reference value; wherein an increase in the value of the levels of anti-CD26 isotype IgG and / or IgM antibodies and / or the levels of anti-CD26 isotype IgA antibodies in a sample of the subject in relation to the reference value is indicative of the disease, and in which said reference value corresponds to the corresponding reference value (eg, average levels) in healthy subjects. In a preferred embodiment of any of the above, the increase in the levels of said anti-CD26 antibody with respect to said reference value is at least 1.2x, preferably at least 1.3x, more preferably at least 1 , 4x, even more preferably at least 1.5x, and most preferably at least 2x. In a more preferred embodiment, the increase in IgG is at least 1.3x, the increase in IgM is at least 1.4x, and the increase in IgA is at least 1.3x. The method according to the first aspect and related aspects may further comprise the determination of total IgA antibody levels, which have demonstrated a high discrimination value between healthy donors and the entire cohort (see Figure 15, AUC: 0.979) and among healthy donors and patients undergoing DMARDs but no biological therapy (Figure 16, AUC: 0.947). The method according to the first aspect and related aspects may further comprise the determination of other biomarkers associated with said autoimmune and / or inflammatory diseases, preferably, said biomarkers are selected from the group consisting of anti-citrullinated protein antibodies (ACPA), rheumatoid factor antibodies, anti-mannose-binding lectin antibodies (MBL), erythrocyte sedimentation rate (VSG), C-reactive protein (CRP), platelet count, hemoglobin and hematocrit levels. In a particular embodiment, these are selected from a list comprising ESR, the levels of PCR concentration, anti-citrullinated protein antibodies (ACPA), rheumatoid factor antibodies and anti-mannin-binding lectin antibodies (MBL) that have been previously used as diagnostic markers. More details are provided in Gupta B et al, J Autoimmun 2006; 27: 125-133, Afzal N et al, Clin Lab 2011, 57: 895-899, Takizawa Y et al, Ann Rheum Dis 2006; 65: 1013-1020, Vossenaar ER et al, Arthritis Res Ther 2004; 6: R142-R150 and EP0175270 which are incorporated herein by reference. In a particular embodiment, the anti-citrullinated protein antibodies (ACPA) are determined to be the negative subject for these anti-citrullinated protein antibodies (ACPA). In another particular embodiment, optionally in combination with any of the above, the methods under the first aspect and related aspects may further comprise the determination of clinical parameters. Signs and / or symptoms whose presence / absence can be determined are: morning stiffness, arthritis of three or more joints, involvement of the hand joint, symmetric arthritis, rheumatoid nodules, and radiographic changes (see Table 1 from Rindfleisch and Muller (American Family Physician 2005, 72 (6), 1037-1047) and the ACR / EULAR classification criteria (Ann Rheum Dis 2010; 69: 1580-1588). A "confirmed diagnosis of rheumatoid arthritis" or "definitive rheumatoid arthritis" may be based on the confirmed presence of synovitis in at least one joint, the absence of an alternative diagnosis that better explains synovitis, and obtaining a total score of 6 or greater (from a maximum possible of 10) from the individual scores in four domains: number and location of affected joints (range 0-5), serological alterations (range 0-3), high acute phase response (range 0 -1) and duration of symptoms (two levels, range 0-1). The meaning of these parameters are defined in the ACR / EULAR classification criteria (Ann Rheum Dis 2010; 69: 1580-1588). The term "joint involvement", as used herein refers to any inflamed or tender joint in the examination, with confirmed synovitis. by image, Distal interphalangeal joints, first carpometacarpal joints and first metatarsophalangeal joints are typically excluded from the evaluation. Joint distribution categories are generally classified according to the location and number of joints affected, with placement in the highest possible category, based on the pattern of joint involvement. The term "large joints" as used herein refers to shoulders, elbows, hips, knees and ankles. The term "small joints" as used herein refers to metacarpophalangeal joints, proximal interphalangeal joints, second to fifth metatarsophalangeal metatarsophalangeal joints, interphalangeal joints of the thumb and wrists. Preferably, the method of the invention further comprises storing the results of the method in a data carrier. The results of the method of the invention include the UNR values of gene expression, the classification of a good or bad prognosis of the subject, the classification of the subject at the time of having short or long term results after surgery, and any other determination according to any of the methods of the invention, as well as any mathematical or statistical treatment thereof. Said data carrier may be a sheet of paper or, preferably, a computer readable medium. As used herein, "a computer readable medium" may be any apparatus that may include, store, communicate, propagate or transport the results of the determination of the method of the invention. The medium can be an electronic, magnetic, optical, electromagnetic, infrared, or semiconductor system (or apparatus or device) or a propagation medium. The methods of the present invention can be implemented by a computer. Therefore, a further aspect of the invention relates to a computer-implemented method, in which the method is any of the methods described herein or any combination thereof. It is noted that any computer program capable of implementing any of the methods of the present invention or the program used to implement any of these methods or any combination thereof, is also part of the present invention. Also included is any device or apparatus comprising, or carrying, a computer program for the application of any of the methods of the present invention or any combination that, therefore, is included as part of the present specification. An in vitro method of disease control and / or monitoring the ability to respond to a treatment in a subject who has, or is suspected of having, an autoimmune disease and / or inflammatory disease and related methods In another aspect, the invention relates to an in vitro method of disease control and / or monitoring the ability to respond to a treatment in a subject who has, or is suspected of having, an autoimmune disease and / or inflammatory disease. , said method comprising steps a) and b) of the first aspect of the invention. In a particular embodiment, an increase in levels of anti-CD26 IgG antibodies and / or levels of anti-CD26 IgM antibodies and / or the ratio between anti-CD26 antibody levels / total IgG antibody levels in the subject sample in relation to the reference value is indicative of the disease. In a related aspect, the invention relates to an in vitro method for classifying a subject as a respondent to a treatment for an autoimmune and / or inflammatory disease, said method comprising steps a) and b) of the first aspect of the invention. In a particular embodiment, an increase in levels of anti-CD26 IgG antibodies and / or levels of anti-CD26 IgM antibodies and / or the ratio between levels of anti-CD26 IgG antibodies / total / total IgG antibody levels in the sample of the subject in relation to a reference value is indicative of lack of response. For any of the aforementioned aspects, said reference value may correspond to a reference value (for example, the average levels of anti-CD26 antibodies or their quotients) in healthy subjects. It may also correspond to the levels of anti-CD26 antibodies determined in an appropriate sample isolated from a patient at an earlier time in time, such as might be close to diagnosis and / or before treatment. Preferred features and embodiments are as defined above for other aspects of the invention. A method of treating a subject and secondary medical uses of the invention In a further aspect, the present invention relates to a method of treating a subject having an autoimmune disease and / or an inflammatory disease, said method comprising the identification of the subject to be treated by a method thatit comprises steps a) and b) of the first aspect of the inventionYc) treatment of said subject who suffers or is suspected of suffering from a diseaseAutoimmune and / or inflammatory disease.In a particular embodiment, an increase in any of the levels of anti-antibodyCD26 IgG and / or levels of anti-CD26 IgM antibodies and / or the ratio between the levels ofIgG anti-CD26 antibodies / total IgG antibody levels in a subject's sampleWith respect to a reference value it is indicative of disease. Said reference value may correspond to a reference value (for example, the levelrelevant anti-CD26 antibody medium or its ratio) in healthy subjects.Such therapies may include one or more of the antirheumatic drugs modifying thedisease (DMARD); non-steroidal anti-inflammatory drugs (NSAIDs); biological therapies, whichcan be divided as biological anti-TNF alpha (for example, adalimumab, pegolcetolizumab, etanercept, golimumab or infliximab) and biological non-TNF alpha (for example,anti-CD20 antibodies, anti-IL6R antibodies or CTLA4-Fc fusion proteins);corticosteroids; and JAK kinase inhibitors (such as tofacitinib). NSAIDs including, but not limited to Cox-2 inhibitors, diclofenac,ibuprofen, naproxen, celecoxib, mefenamic acid, etoricoxib, indomethacin and aspirin. TheCorticosteroids include, but are not limited to triamcinolone, cortisone, prednisone andmethylprednisolone The disease modifying antirheumatic drugs (DMARDs),include, but are not limited to auranofin, chloroquine, cyclosporine, cyclophosphamide,gold preparations, hydroxychloroquine sulfate, leflunomide, methotrexate, penicillamine,sodium aurothiomalate and sulfasalazine. Illustrative, non-limiting examples of antibodiesTherapeutics used for autoimmune diseases and / or inflammatory diseases areprovided in Table 1 of Chan and Carter, in Nature Rev Immunol, 2010; 10: 301-315. Singh et al., Arthritis & Rheumatol 2016; 68: 1-16 provides a guideline for theRA treatment of the American College of Rheumatology. Table 1 presents aSummary of current therapeutic treatments for RA. In particular, theTreatment recommendations for patients with early symptomatic RA areprovided in the table on page 8. In a particular embodiment, said method of treatment comprises the use of one or moreagents selected from the group consisting of: i. DMARDs, preferably methotrexate, and / or leflunomide; ii. anti-TNF alpha agents; iii. anti-CD20 agents; iv. anti-IL6R agents; Y v. CTLA4-Fc fusion proteins, such as abatacept. In a related aspect, the invention relates to a drug selected from the list consisting of: i. FARMEs; ii. anti-TNF alpha agents; iii. anti-CD20 agents; Y iv. anti-IL6R agents and / or CTLA4-Fc fusion proteins, for use in a method of treating a subject who suffers or is suspected of suffering from an autoimmune disease and / or an inflammatory disease, which has been selected using a method according to any of the methods described herein. Preferred features and embodiments are as defined above for other aspects of the invention. An in vitro method to determine the stage of the progression of an autoimmune and / or inflammatory disease in a subject and related methods In a further aspect, the invention relates to an in vitro method for determining the stage of progression of an autoimmune disease and / or inflammatory disease in a subject, said method comprising steps a) and b) of the first aspect of the invention. In a related aspect, the invention relates to an in vitro method of determining a treatment for a subject having an autoimmune and / or inflammatory disease in accordance with its progression stage comprising steps a) and b) of the first aspect of the invention . In a particular embodiment of any of the above aspects, an increase in any of the levels of anti-CD26 IgG antibodies and / or levels of anti-CD26 IgM antibodies and / or the ratio between levels of anti-CD26 IgG antibodies / Total IgG antibody levels in the subject sample with respect to a reference value is associated with an early stage of progression. In addition, for any of the aforementioned aspects, said reference value may correspond to a reference value (for example, the average levels of relevant anti-CD26 antibodies or the quotient) in healthy subjects. It may also correspond to a reference value (for example, the average levels of relevant anti-CD26 antibodies or the quotient) in patients who have or are suspected of having an anti-inflammatory and / or autoimmune disease, preferably an inflammatory rheumatic disease, more preferably the same inflammatory and / or autoimmune disease. Preferred features and embodiments are as defined above for other aspects of the invention. In a preferred embodiment, optionally in combination with any of the embodiments described herein, it refers to an in vitro method for determining the stage of progression of an autoimmune disease and / or inflammatory disease in a subject, said method comprising the steps a) and b) of the first aspect, where an increase in the levels of anti-CD26 IgG antibodies in the subject's sample with respect to a reference value is associated with the early stage of progression; where said reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. In another preferred embodiment, optionally in combination with any of the embodiments described herein, it refers to an in vitro method of determining a treatment for a subject having an autoimmune and / or inflammatory disease according to its stage of progression. comprising steps a) and b) of the first aspect, where an increase in the levels of anti-CD26 IgG antibodies in the subject's sample with respect to a reference value is indicative of the early stage of progression; where the reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. The use of in vitro levels of anti-CD26 antibodies as biomarkers of an autoimmune disease and / or inflammatory disease The use of in vitro levels of anti-CD26 IgG antibodies and / or levels of anti-CD26 IgM antibodies and / or the ratio between anti-CD26 IgG antibody levels / total IgG antibody levels, optionally in combination with the levels determined in vitro of anti-CD26 IgA antibodies, in a sample of a subject for the detection, diagnosis and / or monitoring of an autoimmune disease and / or an inflammatory disease; and / or to control the efficacy of a treatment in an autoimmune disease and / or an inflammatory disease. Preferred features and embodiments are as defined above for other aspects of the invention. Kit and uses thereof In a further aspect, the invention relates to a kit comprising the reagents for the determination of anti-CD26 antibody levels according to step a) of the first aspect of the invention. In some embodiments, the kit includes a purified or synthesized sCD26 polypeptide or an antigenic fragment thereof (which can be labeled or not) and one or more auxiliary reagents. In other embodiments, the kit includes a suitable host cell (eg, a mammalian cell) that expresses CD26 and one or more auxiliary reagents. CD26, including sCD26 and fragments thereof have been described in the first aspect of the invention. In these embodiments, when the levels of anti-CD26 antibodies are quantified, sCD26 or a cell expressing CD26 is typically employed for the purpose of capture and a secondary antibody for the purpose of detection. In other additional embodiments, the levels of anti-CD26 immunoglobulin produced by cells expressing it are quantified, for example, by immunohistochemistry (IHC) or cytometry, the kit typically includes labeled sCD26 for the purpose of detection and one or more auxiliary reagents. Kit anti-CD26 isotype IgG In a particular embodiment, said kit comprises reagents for the determination of the levels of anti-CD26 antibodies of the IgG isotype in a sample of a subject containing the antibody, wherein said kit comprises: a) a reagent to determine the levels of anti-CD26 antibodies of the IgG isotype; b) optionally, instructions for the use of said reagent in the determination of the levels of anti-CD26 antibodies of the IgG isotype in a sample of a subject. Preferably, a) corresponds to: i. CD26, an antigenic fragment thereof or a cell that expresses any of them; ii. a reagent to determine the levels of captured IgG isotype anti-CD26 antibodies (eg, a labeled anti-IgG antibody) In a preferred embodiment, said kit further comprises:c) a reagent to determine the levels of anti-CD26 antibodies of the IgA isotype and / or theIgM isotype;d) optionally, instructions for the use of said reagent in determining theanti-CD26 antibody levels of the IgA isotype and / or the IgM isotype in said samplecontaining the antibody. Preferably, c) corresponds to: iii. a reagent to determine the levels of anti-CD26 antibodies of the IgA isotype and / or of the captured IgM isotype (for example, a labeled anti-IgA antibody and / or a labeled anti-IgM antibody). IgM isotype anti-CD26 kit In another particular embodiment, said kit comprises reagents for the determination of the levels of anti-CD26 antibodies of the IgM isotype in a sample of a subject containing the antibody, wherein said kit comprises: a) a reagent to determine the levels of anti-CD26 antibodies of the IgM isotype; b) optionally, instructions for the use of said reagent in the determination of the levels of anti-CD26 antibody levels of the IgM isotype in a sample of a subject. Preferably, a) corresponds to: i. CD26, an antigenic fragment thereof or a cell that expresses any of them; ii. a reagent for the determination of captured levels of anti-CD26 antibody of the IgM isotype (eg, a labeled anti-IgM antibody); In a preferred embodiment, said kit further comprises:c) a reagent to determine the levels of anti-CD26 antibodies of the IgA isotype and / or of theisotype;d) optionally, instructions for the use of said reagent in determining thelevels of anti-CD26 antibodies of the IgA isotype and / or of the IgG isotype in said samplecontaining the antibody. Preferably, c) corresponds to: iii. a reagent for determining the levels of anti-CD26 antibodies of the IgA isotype and / or of the IgG isotype (for example, a labeled anti-IgA antibody and / or a labeled anti-IgG antibody). Preferably, said kit comprises a solid support or a surface coated with i). The solid support may include any support known in the state of the art in which a protein of the present disclosure can be immobilized. In some embodiments, said solid supports are microtiter plates, slides (for example, glass slides), chips (for example, protein chips, biosensor chips, such as Biacore chips), microfluid cartridges, cuvettes, beads (for example, magnetic beads, xMAP® beads) or resins. Said reagent for the determination of antibody levels of the IgM, IgG and / or isotypes or IgA can be a secondary antibody. Secondary antibodies in the present invention may include, for example, an anti-human IgA antibody, an anti-human IgG antibody, or an anti-human IgM antibody. The secondary antibodies may be monoclonal or polyclonal antibodies. Secondary antibodies can be obtained from any mammal, including mouse, rat, hamster, goat, camel, chicken, rabbit, and others. Secondary antibodies may be conjugated to any suitable detection means, such as enzymes (for example, horseradish peroxidase (HRP), alkaline phosphatase (AP), luciferase, and the like) or dyes (eg, colorimetric dyes, fluorescent dyes, dyes by fluorescence resonance energy transfer (FRET), FRET dyes with temporal resolution (TR), and the like). In some embodiments, the secondary antibody is a goat anti-human IgG polyclonal antibody (or anti-human IgM or anti-human IgA depending, respectively, on the isotype to be determined), conjugated with HRP. Any of the above embodiments may further comprise:e) a reagent to determine the total antibody levels of the IgM, IgG and / or isotypesIgA; f) optionally, instructions for the use of said reagent in the determination of the total antibody levels of the IgM, IgG and / or IgA isotypes in a sample containing said antibodies. Preferably, e) corresponds to: iv. a reagent for the capture of antibodies of the IgM, IgG and / or IgA isotypes (for example, an anti-IgM, anti-IgG and / or IgA antibody) v. a reagent to determine the levels of antibodies captured from the IgM, IgG and / or IgA isotypes (for example, an antibody labeled anti-IgM, anti-IgG and / or anti-IgA); Typically, the reagent in iv) and v) are polyclonal antibodies against the corresponding isotype from different animal species (for example, a rabbit anti-human IgG polyclonal antibody and a goat human anti-IgG polyclonal antibody, respectively). These can also be monoclonal antibodies, with the proviso that they bind to different epitopes and there is no steric interference. Preferably, said kit comprises a solid surface or support, which is coated with iv). This surface may or may not be the same as it is coated with i). Preferably, the surface coated with i) and the surface coated with iv) are separate surfaces. Auxiliary reagents typically used in an immunoassay, such as a solid support immunoassay, may include, for example, an immobilization buffer, an immobilization reagent, a dilution buffer, a secondary antibody, a detection reagent, a buffer. blocking, a wash buffer, a detection buffer, a detection reagent, a stop solution, a system wash buffer, and a system cleaning solution that are well known to a person skilled in the art. Preferred features and embodiments are as defined above for other aspects of the invention. In particular, an example of an immunoassay for determining levels of anti-CD26 antibodies is provided within the framework of the first aspect of the invention. In a further aspect, the invention relates to the use of a kit of the above aspect for the detection, diagnosis and / or monitoring of an autoimmune disease and / or an inflammatory disease; and / or for monitoring the efficacy of a treatment in a sample of a subject containing the antibody. Preferred features and embodiments are as defined above for other aspects of the invention. Total levels of IgA as a biomarker for the detection, diagnosis, monitoring of an inflammatory and / or autoimmune disease and related aspects. As stated in Example 5, total IgA antibody levels have demonstrated a high discrimination value between healthy donors and the entire cohort of patients treated with different therapies (see Figure 15, AUC of 0.979) and between healthy donors and patients undergoing DMARDs but no biological therapy (Figure 16, AUC: 0.947). Consequently, in a further aspect, the invention relates to an in vitro method for screening, for obtaining useful data for diagnosis, for diagnosis or for monitoring a subject who has, or is suspected of having, an autoimmune disease and / or an inflammatory disease comprising the following steps:a) determination of total antibody levels of the IgA isotype (total levels ofIgA antibodies) in a sample of a subject containing the antibody;b) comparison of the levels in said sample containing the antibody with a value ofreference. In a particular embodiment, an increase in total antibody levels of the IgA isotype in the subject's sample with respect to said reference value is indicative of disease. Said reference value may correspond to a reference value for total antibody levels of the IgA isotype (for example, the mean or median of these levels or a calculated cut-off value) in healthy subjects. It may also correspond to said levels of total IgA antibodies determined in an appropriate sample of a patient at an earlier time in time, such as close to diagnosis and / or before treatment. Total levels of IgA antibodies (for example, total levels of human IgA antibodies) can be determined by any method well known in the art. These are typically quantified by the use of an immunoassay including, in particular, an immunoassay on a solid support as described in detail in the first aspect of the invention, wherein the solid support is coated with an anti-IgA antibody (byexample, an anti-human IgA antibody). In a further aspect, the present invention relates to a method of treating asubject who has an autoimmune disease and / or an inflammatory disease,said method comprising the identification of the subject to be treated by a method thatIt comprises the stages of: a) determination of total antibody levels of the IgA isotype (total levels ofIgA antibodies) in a sample of the subject containing said antibodies;b) comparison of antibody levels in said sample with a reference value;Yc) treatment of said subject who has or is suspected of having an autoimmune diseaseand / or an inflammatory disease. In a particular embodiment, an increase in total antibody levels of the IgA isotypein the sample of the subject with respect to said reference value is indicative ofdisease. Said reference value may correspond to the reference value of the total level ofIgA antibodies (for example, the mean or median levels or a cut-off value) inhealthy subjects In a further aspect, the invention relates to a kit comprising reagents for thedetermination of total antibody levels of the IgA isotype in a sample of asubject containing said antibody:a) a reagent for the determination of total antibody levels of the IgA isotype;b) optionally, instructions for the use of said reagent in determining thetotal levels of IgA isotype antibodies in a sample of a subject containing saidantibody. In a particular embodiment, said kit further comprises:c) a reagent for the determination of the levels of an anti-CD26 antibody of the IgG isotype;d) optionally, instructions for the use of said reagent in determining thelevels of an IgG isotype anti-CD26 antibody in a sample of a subject that containsthe antibody In another particular embodiment, optionally in combination with the previous embodiment, saidkit also includes:e) a reagent to determine the levels of anti-CD26 antibody of the IgA isotype and / or of theIgM isotype;f) optionally, instructions for the use of said reagent in determining thelevels of anti-CD26 antibodies of the IgA isotype and / or of the IgM isotype. In a further aspect, the invention relates to the use of a kit for screening, diagnosis and / or monitoring of an autoimmune disease and / or an inflammatory disease; and / or for monitoring the efficacy of a treatment in a sample of a subject containing antibody, in which said kit is as described in the previous aspect. Preferred embodiments and features of any of these aspects are as described above for the other aspects of the invention. It is contemplated that any embodiment described herein with respect to one aspect of the invention also applies to other aspects of the invention, and vice versa, more specifically can be implemented in relation to any method, kit, and use of the invention herein. described. It will be understood that the particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The main features of this invention can be used in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or will be able to determine using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered within the scope of this invention and are covered by the claims. All publications and patent applications mentioned in the specifications are indicative of the level of experience of those skilled in the art to which this invention pertains. All publications and patent applications are incorporated herein by reference to the same extent as if each individual publication or patent application is specifically and individually indicated to be incorporated by reference. The use of the word "a" or "a" may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more of one." The use of the term "other" may also refer to one or more. The use of the term "or" in the claims is used to mean "and / or" unless expressly stated to refer to alternatives only or the alternatives are mutually exclusive. As used herein and claim (s), the words "comprising" (and any form of "comprising", such as "comprise" and "comprising"), "having" (and any form of having, such as "have" and "have"), "that includes" (and any way of including, such as "includes" and "include") or "that contains" (and any form of containing, such as "contains" and "contain ") are inclusive and do not exclude additional elements not cited, or stages of the procedure. The term "comprises" also expressly encompasses and discloses the terms "consists of" and "consists essentially of". As used herein, the phrase "consisting essentially of" limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel feature (s) of the claimed invention. As used herein, the phrase "consisting of" excludes any element, stage, or ingredient not specified in the application with the exception of, for example, impurities normally associated with the element or limitation. The term "or combinations thereof," as used herein, refers to all permutations and combinations of the preceding enumerated elements of the term. For example, "A, B, C, or combinations thereof" is intended to include at least one of: A, B, C, AB, AC, BC or ABC, and if the order is important in a particular context, also BA, CA, CB, ACB, ACB, ACB, BAC, or CAB. Following this example, combinations containing repetitions of one or more elements or terms, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so on, are expressly included. The person skilled in the art will understand that normally there is no limit on the number of elements or terms in any combination, except others evident from the context. A person skilled in the art generally understands that a value includes the standard deviation of the error for the device or method used to determine the value. This can be emphasized in this document by the use of approximation words such as, without limitation, "about", "about", "approximately", refers to a condition thus modified is understood to be not necessarily absolute or perfect, but it would be considered close enough by those skilled in the art to ensure that it designates that said condition is present. The extent to which the description may vary will depend on how large a change can be established and yet those skilled in the art recognize that the modified characteristic still has the required characteristics and capabilities of the unmodified function. In general, but subject to the previous discussion, a numerical value in this document that is modified by a word of Approximation as "approximately" may vary with respect to the declared value of ± 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15%. Preferably, the term "approximately" means exactly the indicated value (± 0%). The following examples serve to illustrate the present invention and should not be construed as limiting the scope thereof. EXAMPLES Example 1.-Material and Methods Study design 110 patients from the Rheumatology Service (Hospital Meixoeiro-CHUVI) were recruited in a cross-sectional case-control study. The patients met the criteria of the American College of Rheumatology (ACR) / EULAR of 2010, Aletaha D et al, 2010; Arthritis Rheum 62: 2569-2581) and treated with different therapies, including biological therapies (BT). The only exclusion criterion was the patient's decision not to be included in the study. A group of 25 healthy donors was also recruited. All the procedures described were performed in accordance with the clinical ethical practices of the Spanish and European administrations and approved by the local ethics committee. Written informed consent was obtained from all participants. The disease activity was previously evaluated in this group of patients using the DAS-28 index, which takes into account the number of painful joints (TJC), inflamed joints (SJC), erythrocyte sedimentation rate (ESR) and the overall evaluation of the patient (PGA) of disease activity, noted by a numerical classification scale (NRS 0-100). The erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), hemoglobin levels and platelet concentration were also recorded as markers of RA activity. The DAS-28 index and how to determine it is well known to one skilled in the art, see for example J. S. Smolen et al. Arthritis Rheum 1995, 38: 38-43, whose content is included here for reference. All patients also completed the HAQ (Stanford Health Assessment Questionnaire: Fries J, Spitz PW, Young DY. The dimensions of health outcomes: the health assessment questionnaire, disability and pain scales, J Rheumathol 1982, 9: 789- 93), which is an evaluation of functional status (disability). The results in the parameters evaluated of disease activity have been published previously by the inventors (Lamb OJ et al., PLoS One 2015, 10: E0131992) and the content of this document is included as a reference. Definition of different groups of patients depending on the therapy. The patients studied were classified in different groups according to the therapies they were receiving, which may have different effects on antibody values. The cohort was divided into four groups according to the mechanism of action of the different therapies: a) disease-modifying anti-rheumatic drugs (DMARDs, methotrexate and / or leflunomide); b) anti-TNF-alpha agents (adalimumab, etanercept, infliximab, golimumab), c) anti-CD20 monoclonal antibodies (mAb) (rituximab), and d) anti-IL6R (tocilizumab) or Ig-CTLA4 (abatacept) mAb. In this last subgroup both treatments showed a similar immune response, so the results of the patients were grouped. Smoking History The smoking history of the patients was evaluated at the time of inclusion. Current smokers were those who reported active smoking. The ex-smokers were all those patients who had quit smoking before the first exam at the time of inclusion. Non-smokers were those without a history of smoking at any time. Biological samples For serum collection, peripheral venous blood was taken in BD SST ™ II Advance tubes, allowed to coagulate at room temperature and centrifuged at 2000 x g for 15 min. The serum was stored at -80 ° C until use. Blood cells were collected using TransFix® Vacuum Blood Collection tubes (Cytomark, Buckingham, United Kingdom) and stored at 4 ° C until use. Determination of DPP-IV enzyme activity and soluble DC26 protein (sCD26) Both techniques have been described previously (Lamb OJ et al., Plos One 2015; 10 (7): e0131992, Cuchacovich M et al., Clin Exp Rheumatol 2001; 19: 673-80, Ellingsen T et al., Scand J Immunol 2007; 66 : 451-7] Since previous studies [Lamb OJ et al., Immunobiology 1997; 197: 522-33, De Chiara L et al., Dis Markers 2009; 27: 311-6. Doi: 10.3233 / DMA-2009-0679 , Lambeir AM et al., Crit Rev Clin Lab Sci 2003; 40: 209-94) have shown small non-statistically significant differences in the levels of sCD26 and DPPIV enzyme activity depending on sex or age, control values and patients with RA were not matched according to these two parameters. Determination of total immunoglobulins and anti-CD26 autoantibodies by enzyme linked immunosorbent assays. The concentration of total IgA, IgG and IgM as well as anti-CD26 / DPPIV autoantibodies in the patients' serum was determined by ELISA using 96-well culture plates coated with rDPP-IV (0.5 g / mL) (Sigma -Aldrich Spain or R&D Systems, USA) or with rabbit polyclonal antibody anti-IgA, anti-IgG and anti-human IgM (2 g / mL) (DakoCytomation, Denmark) prepared in PBS pH 7.4 and they were blocked overnight with 0.5% BSA PBS. The plates were incubated with different serum dilutions for 1 h at 37 ° C and then washed four times with PBS 0.05% Tween20. Goat anti-IgM antibodies (specific for--chain), anti-IgG (specific for Fab) and anti-IgA (specific for--chain) (all from Sigma-Aldrich) were used to detect the antibodies captured together with the standard OPD substrate (Sigma-Aldrich) following the manufacturer's instructions. Absorbance at 450 nm was recorded using a BioRad plate reader (BioRad, Madrid, Spain). Concentrations were calculated from calibration curves constructed with purified human IgA, IgG or IgM. ACPA test The Elia ™ CCP test (Thermo Scientific / Phadia, Sweden) was used to analyze whether anti-CD26 autoantibodies in RA were antibodies against citrullinated CD26. Briefly, the assay was used to bind the anti-CCP antibodies of the patients' serum to the plaque containing CPP antigens, according to the manufacturer's instructions. The remaining serum is recovered as a negative fraction for ACPA. After washing the Elia-CCP plate to avoid nonspecific binding, the ACPAs were eluted from the plate with elution buffer (0.1 M Gly-HCl pH 2.7) and this was neutralized with 1 M Tris-HCl pH 9 to get the positive fraction for ACPA. Both fractions were now evaluated by the anti-CD26 ELISA described above. In the same way, the latter ELISA was used to obtain a negative fraction for anti-CD26 antibodies (recovery of serum from patients after antibody capture to CD26 coated plates) and a positive fraction for anti-CD26 antibodies (obtained after washing and eluting the plate with the captured antibodies); the two fractions were analyzed by the Elia ™ CCP test. Sera from three patients with RA who show high levels of anti-CD26 antibodies of the IgG isotype and those from three healthy donors were chosen. In all ELISA assays only reagents were used for the detection of antibodies of the IgG isotype (described above). Statistic analysis All analyzes were parametric. The ANOVA test was performed to compare the variables between the four groups of patients with or without biological therapies. The Scheffé post-hoc test was used for variables with homogeneous variances and Dunnett's posthoc C test for variables without homogeneous variances. Dunnett's t-test was performed to compare each group with a control group, either the group without biological treatment or the group of healthy donors. The Student's t-test was also used to compare variables between two groups. Statistical analyzes were performed using SPSS software version 20 (SPSS, Chicago, IL, USA). Example 2.-Levels of anti-CD26 / DPP-IV autoantibodies and their correlation with serum DPP-IV activity and sCD26 concentrations in the cohort of healthy donors Autoantibodies against CD26 have been described in healthy donors and their participation in the clearance of this antigen has been proposed through their secretion in the bile (Cuchacovich M et al. Clin Exp Rheumatol 2001. 19: 673-80). With our ELISA, the titres measured in serum (n = 25) for the total immunoglobulin concentrations for the three isotypes under study were consistent with the expected values, 2.37 ± 1.24 mg / ml for IgM, 2.28 ± 0.27 mg / ml for IgA and 12.28 ± 2.79 mg / ml for IgG (González-Quintela A et al. Clin Exp Immunol 2007; 151: 42-50). The mean serum titers of anti-CD26 antibodies are also quite similar to those published previously (Snir O et al. Arthritis Rheum 2010; 62: 44-52; DOI: 10.1002 / art.25036), 13.51 ± 5, 62 g / mL for IgM, 1.28 ± 0.27 g / mL for IgA and 2.14 ± 0.64 g / mL for IgG, with the current values being slightly lower for the IgM and IgA isotypes in Comparison with previous results published by Cuchacovich et al (Cuchacovich M et al Clin Exp Rheumatol 2001; 19: 67380) probably due to the fact that recombinant human antigen was used instead of purified CD26 from human serum. However, comparing the ratios between anti-CD26 autoantibodies / total immunoglobulin isotype published by Cuchacovich et al. With our data (7.12 for IgM, 0.57 for IgA and 0.19 for IgG), our results are more consistent with the expected values for total immunoglobulin titers. Importantly, none of the serum titres shown, including the reduced levels of anti-CD26 autoantibodies of the IgA isotype, or their ratios, correlate with DPP-IV enzyme activity or serum sCD26 concentration in this cohort of healthy donors, which suggests that if its function is related to antigen clearance, its effect on sCD26 levels is undetectable. Example 3.-Total levels of antibodies and anti-CD26 / DPP-IV autoantibodies in patients with RA following different treatments 3.1 Total levels of anti-CD26 / DPP-IV antibodies and autoantibodies in the complete cohort When analyzing the complete cohort of patients with RA (n = 106) we have found higher levels of both total immunoglobulins and anti-CD26 autoantibodies. Total immunoglobulin concentrations for the three isotypes are 4.33 ± 3.79 mg / mL for IgM, 5.61 ± 1.77 mg / mL for IgA and 21.81 ± 15.35 mg / mL for IgG (Lambeir AM et al Crit Rev Clin Lab Sci 2003; 40: 209-94) .The concentrations of anti-CD26 autoantibodies are 27.97 ± 19.09 /g / mL for IgM, 2.01 ± 1.34 /g / mL for IgA and 5.87 ± 3.13 /g / mL for IgG, and the auto-antibody / total isotype ratios are 17.10 for IgM, 0.43 for IgA and 0.75 for IgG. Thus, although all total immunoglobulin values increased twice in general, it is interesting to note that the ratios for IgM isotypes and in particular IgA are lower compared to healthy donors, while this was not the case for the IgG isotype. , which is slightly higher and significantly different compared to healthy donors (p = 0.03; ANOVA test), in addition to all average values for total immunoglobulin titers and anti-CD26 autoantibodies of the three isotypes (p < 0.001 for most of them with the Student t test). These results point to the usefulness of anti-CD26 antibody titers as a biological marker of RA. 3.2 Total antibody levels and anti-CD26 / DPP-IV depending on therapy The group under anti-CD20 therapy can serve as a control. Therapeutic monoclonal antibodies directed against the CD20 antigen in B cells are used successfully in the clinic for the reduction of these B cells in the treatment of forms of cancer and autoimmune diseases, since by their mechanism of action they reduce immunoglobulin levels in general. Anti-CD20 therapy has been shown to reduce levels of total IgG and in particular IgM, as expected, but had no effect on total IgA (Table 2) compared to the group without biological treatment. The decrease observed, however, does not reach the levels of antibodies observed in healthy donors. On the contrary, it is shown that the other biological therapies raise the total levels of the IgG and IgM isotypes, with the group receiving anti-IL6R / Ig-CTLA4 being the only one that shows a slightly lower level of IgA (Table 2). However, the effect of the different treatments on the levels of anti-CD26 autoantibodies was different. The IgG isotype was not affected by any of the therapies; he Anti-CD20 treatment strongly reduces only the IgM isotype and finally the IgA isotype is affected by both anti-TNF-alpha treatment and anti-IL6R / Ig-CTLA4 therapies but in opposite directions (Table 2). As a consequence, the quotients for each of the three isotypes, in particular for IgM and IgA are quite different in comparison with those of healthy subjects, underlining the specificity of our data. Most of these differences were statistically significant (see Table 2), in particular for the IgM isotype in patients treated with anti-TNF and anti-IL6R / Ig-CTLA4, for the IgA isotype in patients treated with anti-CD20 e IL6R / Ig-CTLA4 BT, and for the IgG isotype for patients in treatment with non-biological agents and in treatment with IL6R / Ig10 CTLA4. Similar conclusions can be drawn when examining the quotients for the different isotypes (A / M, G / A, G / M) for both the total immunoglobulin and autoantibody titers: there are statistically significant differences for any of these ratios. between patients and healthy donors (Table 3) and important differences between treatments (Fig. 6, check the different behavior of the G / A ratio for titres 15 total and autoantibodies in patients with anti-CD20 therapy). Table 2. Levels of total Ig and anti-CD26 isotypes in RA patients according to therapy. 54 Ig, immunoglobulin, RA, rheumatoid arthritis, n = number of samples; BT = biological therapy; CI = confidence interval; DS = standard deviation. * Significantly different values between groups (unspecified) at p <0.05 with the Student t test + Values significantly different from those of the group without biological therapy (No BT) at p <0.05 with Dunnett's t-test Table 3. Levels of total and anti-CD26 Ig isotypes throughout the cohort of RA patients (treated with different therapies) Controls (n = 25) AR patients (n = 106) Mean ± SD Mean ± SD Total IQ ratio A / M 1.3 ± 1.0 3.7 ± 6.7 ** Total IQ ratio G / A 5.4 ± 1.3 4.8 ± 7.3 Total IQ ratio G / M 6.7 ± 5.2 11.3 ± 18.6 * Auto IQ ratio A / M 0.11 ± 0.04 0.13 ± 0.21 Auto Ig ratio G / A 1.7 ± 0.6 7.5 ± 27.1 Auto Ig ratio G / M 0.17 ± 0.06 0.32 ± 0.35 ** Ig, immunoglobulin, RA, rheumatoid arthritis, n = number of samples; SD = standard deviation. * Significantly different values between groups (unspecified) at p <0.05 with the Student t test ** Significantly different values between groups (unspecified) at p <0.001 with the Student t test Example 4.-Diagnostic value of anti-CD26 / DPP-IV antibody levels Since our cohort of patients with RA showed elevated titers of anti-CD26 autoantibodies, which indicates its usefulness in following patients' capacity to respond to therapy at the individual level, we have assessed whether any of the isotype titres could have value diagnosis. 4.1 Anti-CD26 / DPP-IV levels throughout the cohort Cohort patients under study have already been diagnosed and treated. Thus,this group of subjects is not ideal for the evaluation of the diagnostic value of the titles ofanti-CD26 antibodies. However, IgG isotype titres have shown thatthey remain unchanged with the therapies employed and therefore can be a goodindicator. In addition, to test the performance of biomarkers in a screening test /diagnosis, ROC curves representing sensitivity values have been preparedobtained against the 1-specificity values for the anti-CD26 autoantibodies of theIgG, IgM and IgA isotypes, finding the following values of AUC, specificity (E) andSensitivity (S) in the calculation of the optimum cut-off point: -anti-CD26 IgM (Fig. 7): AUC (95% CI): 0.748 (0.666-0.830); cut-off point: 19 g / mL, 62%S, 88% E-anti-CD26 IgG (Fig. 8): AUC (95% CI): 0.881 (0.825-0.938); cut-off point; 2.85 /g / mL,82% S, 96% E-anti-CD26 IgA (Fig. 9): AUC (95% CI): 0.661 (, 573-, 749); cut-off point: 1.5 g / mL, 60% S,80% E-anti-CD26 IgG / total IgG (Figure 10): AUC (95% CI): 0.741 (0.653 to 0.829); cut point:0.24 /g / mg, 60% S, 84% E These cut-off points are, respectively, 1.41, 1.33, 1.17 and 1.26 times the values ofreference (mean concentrations of healthy donor biomarkers). 4.2 Levels of anti-CD26 / DPP-IV autoantibodies in patients undergoing DMARD but without any biological therapy Next, we evaluated the group undergoing DMARDs but without any biological therapy (noBT), which would look better to undiagnosed patients and / or patients close to a pre-RA in the sense that these therapies do not change the expression of CD26 at least in leukocyte populations (Ellingsen T et al., Scand J Immunol 2007; 66: 4517). A sensitivity of 77% (17/22) is found for the anti-CD26 IgG isotype (with a cut-off point of 2.85 /g / mL). A similar result of 73% (16/22) sensitivity was found for anti-CD26 IgM (with a cut-off value of 20 g / mL) and IgA 64% (with a cut-off value of 1.7 g / mL) (Figure 1). Notably, only 3 of 22 patients were negative for both IgG and IgM isotypes and 2/22 for all three. In addition, to test the performance of biomarkers in a screening / diagnostic test, ROC curves have been prepared representing the sensitivity values obtained versus the 1-specificity values for the anti-CD26 autoantibodies of the IgG, IgM and IgA isotypes, finding the following AUC values, specificity (e) and sensitivity (S) in the calculation of the optimum cut-off point: -anti-CD26 IgM (Fig. 11): AUC (95% CI): 0.751 (0.583 to 0.919); 20 g / mL cut-off point, 73% S, 92% E-anti-CD26 IgG (Fig. 12): AUC (95% CI): 0.846 (0.718-0.975); 2.85 g / mL cut-off point, 77% S, 96% E-anti-CD26 IgA (Fig. 13): AUC (95% CI): 0.672 (0.495-0.848); cut-off point of 1.7 /g / mL, 64% S, 80% E-anti-CD26 IgG / total IgG (Fig. 14): AUC (95% CI): 0.804 (0.670-0.938); cut-off point: 0.24 /g / mg (same as in the complete cohort), 68% S, 84% E; and with a cut-off point of: 0.25 g / mL, 68% S, 88% E These cut-off points are, respectively, 1.48, 1.33, 1.33 and 1.26 times than the reference values (mean concentrations of healthy donor biomarkers). It is observed that an important difference between the results of the inventors and those previously published by Cuchacovich et al (Cuchacovich M et al, Clin Exp Rheumatol 2001; 19: 673-80) is that higher titers of autoantibodies were found in the previous study anti-CD26 in RA and other autoimmune diseases, in particular, of the IgA isotype. However, the inventors have found high levels of the three classes evaluated but in particular for the IgM and IgG isotypes, which in fact showed better diagnostic values compared to the IgA isotype. It should be noted that Cuchacovich et al. Only the ratios between the level of autoantibodies / total Ig for each isotype are shown, while the results for each of these are presented in Example 3 above. individual determinations and for their quotient, and in fact show that the levels of anti-CD26 antibodies of each isotype were independent of the changes in total Ig levels for each isotype. The differences between the titles of anti-CD26 autoantibodies between both studies may also be related in part to the fact that we have used a recombinant sCD26 protein as an antigen for the ELISA instead of an isolated sCD26 antigen from patient sera, which has been described as a more complex molecule (Cuchacovich M et al., Clin Exp Rheumatol 2001; 19: 673-80, or Lambeir AM et al., Crit Rev Clin Lab Sci 2003; 40: 209-94, or Lamb OJ et al, Cancer Immunol Immunother 2009; 58: 1723-47). Example 5.-Diagnostic value of total IgA antibody levels Interestingly, during the elaboration of the ROC curves for the anti-CD26 autoantibodies, total antibodies and the ratios thereof for the IgA, IgM and IgG isotypes, the inventors have observed that the total antibodies of the IgA isotype have proven to be a very good discriminator between healthy donors (HD) and the population of RA patients, both for the complete cohort (see Figure 15, AUC (95% CI) of 0.979 (0.953-1,000); cut-off point: 0.24 g / mL , 60% S, 84% e) and the group of patients with non-biological therapy, which is assimilable to undiagnosed or pre-RA patients (see Figure 16, AUC (95% CI) of 0.947 (0.859 -1,000) , cut-off point: 2.5 mg / mL, 96% S, 76% e, and cut-off point: 3 mg / mL, 91% S, 100% E). Example 6.-Correlations of anti-CD26 / DPP-IV antibody levels with disease activity parameters of patients with RA The inventors have previously shown correlations between the DAS28 score, DAS28 components (clinical: TJC, SJC, PGA), and laboratory variables: ESR and CRP, or other markers of RA activity (HAQ, hemoglobin, platelets and hematocrit) ) with subsets of T cells defined by CD26 cell surface expression (Lamb OJ et al., PLoS One 2015, 10; e0131992). 6.1 Levels of anti-CD26 / DPP-IV autoantibodies in the entire cohort. In this case, correlations of disease activity were first analyzed against the total levels of each isotype, including rheumatoid factor (RF) and ACPA, in the entire cohort. The total titles of the three classes of Ig do not correlate with DAS28, however there are some statistically significant correlations between the total titles for the three isotypes and ESR (r = 0.272, p = 0.005 for IgM; r = 0.213, p = 0.028 for IgG; r = 0.280, p = 0.004 for IgA). On the other hand, the IgG isotype correlates negatively with TJC and total IgM with HAQ. The levels of anti-CD26 also do not correlate with the DAS28 score in the complete cohort. However, the anti-CD26 autoantibody titres of the three isotypes correlate negatively with TJC (r = -0.272, p = 0.036 for IgM; r = -0.210, p = 0.032 for IgG; r = -0.273, p = 0.005 for IgA); in addition, levels of anti-CD26 IgG autoantibodies correlate negatively with hematocrit and levels of anti-CD26 IgA autoantibodies with ESR (r = -0.360, p <0.001). Finally, the quotients for the IgG and IgA isotypes correlate with the CRP. 6.2 Levels of anti-CD26 / DPP-IV antibodies depending on therapy. To verify the effect that any of the different therapies can have on the correlations between antibody levels and clinical scores, the same analysis (together with internal controls) was performed for each group of patients undergoing different treatments. In the group without BT, while strong correlations are observed among all total Ig titers (and that show correlations with platelet count), only the anti-CD26 autoantibody titers of the IgA and IgM isotypes show a correlation between them. and only the quotient for the IgA isotype show a very strong correlation with the TJC (r = 0.704, p <0.001). In the anti-TNF group, total IgA titres do not correlate with the levels of the other two isotypes, showing only a positive correlation with ESR, positively (data not shown). Total IgM titers also correlate positively with Hb and hematocrit and negatively with CRP, while IgG titers correlate negatively with ESR and platelet count (data not shown). Interestingly, the titles of anti-CD26 IgA autoantibodies correlate strongly with the TJC and DAS28 (Figure 2A and 2B) and the levels of anti-CD26 IgM autoantibodies positively correlate with the TJC (r = 0.295, p = 0.024). No correlations were found for the IgG isotype. However, in the anti-CD20 group, these titles of anti-CD26 IgG autoantibodies correlate very strongly with the TJC, SJC, DAS28 and PGA (Figure 3 A-D). No correlations were found for the other two isotypes, although the titles of anti-CD26 autoantibodies IgG and IgA are correlated with each other. In this group, the total titles of each isotype do not correlate with each other and the only correlations detected were between the ratio of the IgM isotype with both HAQ and PGA and the ratio between the isotype IgG with platelets (data not shown). Finally, in the anti-IL6R / Ig-CTLA4 group, while total IgA levels do not correlate with the titles of the other isotypes (as in the anti-TNF group) and the anti-CD26 IgA autoantibody titres and IgM do not correlate with each other (contrary to what happens in the noBT group), the autoantibody titers for the other combinations (IgA with IgG and IgM with IgG) are. In this group, only the quotient for the IgG isotype correlates with TJC and the total IgG with the hematocrit. Together, current data suggest: a) a specific relationship between anti-CD26 autoantibodies with joint activity and disease, b) the levels of these autoantibodies are modified differently by each therapy, and c) that provide information different compared to most of the disease activity parameters frequently used today (VSG, PCR, platelet count, Hb or hematocrit levels). Example 7.-Detection of higher anti-CD26 autoantibody titers in smokers. Tobacco is an environmental factor that triggers autoimmunity in patients with RA positive for ACPA (Reynisdóttir G et al., Arthritis Rheum 2014; 66: 31-9, doi: 10.1002 / art.38201, or Catrina AI et al., Nat Rev Rheumatol 2014; 10: 645-53, doi: 10.1038 / nrrheum. 2014,115, or Vossenaar ER et al., Arthritis Res Ther 2004; 6: 107-111). We decided to compare the mean titles of anti-CD26 autoantibodies for the three isotypes by dividing the complete cohort of RA patients into three groups based on their tobacco use, current smokers (n = 18), ex-smokers (n = 28) or not smokers (n = 59). While the IgA titres were similar between the three groups, the IgM and IgG isotypes showed higher levels in ex-smokers in particular (26.0 mg / mL in non-smokers compared to 31.8 mg / ml in ex-smokers for IgM, and 5.5 mg / mL to 6.7 mg / mL for IgG), although these differences did not reach statistical significance. Next, we analyze the complete cohort of RA patients taking into account the therapy (Table 4). Interestingly, in the non-BT group, the titles of anti-CD26 IgM autoantibodies are significantly higher in ex-smokers compared to non-smokers. Although there are few current smoking patients in this group, they also showed higher autoantibody titres. The anti-CD26 IgG autoantibodies also showed a tendency to a higher degree in ex-smokers compared to other groups, although this trend did not reach statistical significance. Note that while the levels of anti-CD26 IgG or IgA autoantibodies do not change between the different therapies, the levels of anti-CD26 IgM autoantibodies were attenuated in the ex-smokers in the anti-TNF group and in all patients in the group of anti-CD20 treatment; however, in the group treated with anti-IL6R / Ig-CTLA4, non-smokers showed elevated levels of anit-CD26 autoantibodies of the IgM isotype (Table 4). Table 4. Levels of anti-CD26 Ig isotypes in RA patients with different therapies according to their smoking status. 62 Example 8.-ACPA Test The results obtained, combining the well-established second-generation Elia ™ CCP tool in clinical practice together with our homemade ELISA for anti-CD26 autoantibodies (only the IgG isotype was tested) lead us to the conclusion that CD26 anti-auto antibodies They are not ACPA. When serum supernatants from both healthy donors and patients with RA were recovered from the ACPA test and tested for the presence of anti-CD26 autoantibodies, an absorbance similar to that obtained with the same serum sample tested was detected only using our anti-CD26 ELISA test (not shown) (Figure 4A, patients with RA showing higher absorbance data). The ACPA test showed the expected results, with the highest serum levels of the three patients analyzed compared to three control samples from healthy donors, except for one of them, which was also positive (a candidate for the development of arthritis) (Figure 4B, dark gray). To verify these results, the reverse analysis was also performed, where supernatants recovered from the anti-CD26 ELISA were subsequently used in the ACPA tests. Although the absorbances were similar to those obtained after direct incubation of the same serum in the ACPA trial, there were minor differences in four of the samples and more significant in two of them, a healthy control and a patient with RA (Figure 4B, in light gray). In this case, when the proteins captured in the assay were eluted from the plate (see Methods), a lower positive signal was observed (data not shown), which points to the possibility that a smaller part of ACPA autoantibodies can also bind to CD26. Example 9.-Serum anti-CD26 autoantibodies and their correlations with DPP-IV activity and sCD26 concentrations in the cohort of patients with RA treated with different therapies 9.1 Serum anti-CD26 autoantibodies and their correlations with DPP-IV activity and sCD26 concentrations throughout the cohort Determined for the healthy control group (see example 2 above), we analyzed the correlations of Ig isotypes and their relationships with serum DPP-IV activity and sCD26 concentrations in the entire patient cohort. We found positive correlations close to the statistical significance between total IgG and anti-CD26 IgG with the concentration of sCD26 but not with the enzymatic activity for the total cohort. 9.2. Anti-CD26 serum autoantibodies and their correlations with DPP-IV activity and sCD26 concentrations in relation to the therapy used. When the same analysis was performed for each of the patient groups according to the therapy received, many interesting correlations were found. In the group without TB (noTB), the total levels of IgA and IgM correlate positively with the activity of DPP-IV, but not with the concentration of sCD26 (Figure 5A), while in the group of anti-IL6R / Ig-CTLA4, the total titles of IgM and IgG have a negative correlation with DPP-IV activity (Figure 5D). In the anti-TNFa group that show increased levels of DPP-IV activity (Cuchacovich M et al., Clin Exp Rheumatol 2001; 19: 673-80), in addition to the total IgA and IgM titers, the IgG titres anti-CD26 also positively correlated with DPP-IV activity (Figure 5B). Only in this group (Figure 5B), total IgM and IgG titers and anti-CD26 IgG and IgM titers positively correlate with the concentration of sCD26, while in the antiCD20 group, which shows decreased levels of IgM autoantibodies , both the anti-CD26 IgM titres and the ratio show a strong negative correlation with the concentration of sCD26 (Figure 5C). These results point to the idea that: a) the levels of auto-Antibodies are not related to the reduction of the concentrations of their antigens (sCD26) in patients with RA, since the only correlation seen was observed in the anti group -TNF, with a positive correlation; b) DPP-IV activity and sCD26 concentration, reduced in patients with RA and affected by therapies (Cordero OJ et al., PLoS ONE 2015; 10: e0131992), are related to the same route that increases autoantibody titres anti-CD26; c) together with the fact that different levels of correlation between the activity of DPP-IV and the concentration of sCD26 were found in each group of patients (which do not correlate in the non-TB group and in the anti-treatment group -IL6R / Ig-CTLA4 and correlate more strongly in the anti-CD20 group, data not shown), we also conclude that DPP-IV activity and sCD26 concentration are involved in the pathogenesis of RA in different ways; and d) finally, different therapies have a profound impact on these pathways, even in opposite ways. Interestingly, Cuchacovich et al. Showed a negative correlation between the circulating levels of DPP-IV and the titles of anti-CD26 IgA autoantibodies in RA (Cuchacovich M et al., Clin Exp Rheumatol 2001; 19: 673-80, Cuchacovich M and cols, Clin Immunol Diagn Lab 2002; 9: 1253-9), a correlation that was not found in the present study. On the contrary, when correlations were found, these were positive and especially for the IgG and IgM isotypes of anti-CD26. We found a negative correlation in the patient group treated with anti-CD20, but in this group, the therapy did not seem to decrease autoantibody levels in general. It can be concluded, therefore, that the deterioration of the activity of DPP-IV and the concentration of sCD26 in patients with RA is related to the same route that increases the titers of anti-CD26 autoantibodies that, therefore, do not It appears to be involved in the serum elimination of sCD26.
权利要求:
Claims (28) [1] one. In vitro method for the screening of a subject having an inflammatory rheumatic disease comprising the following steps: a) the determination of the levels of anti-CD26 IgG antibodies in an isolated sample of said subject containing antibodies; b) comparing the levels in said sample containing antibodies with a reference value, wherein an increase in the levels of anti-CD26 IgG antibodies in the subject's sample with respect to said reference value is indicative of disease and in which said reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. [2] 2. In vitro method for obtaining useful data for the diagnosis of an inflammatory rheumatic disease in a subject, said method comprising steps a) and b) of claim 1, wherein an increase in the levels of anti-CD26 IgG antibodies in the Sample of the subject with respect to said reference value is indicative of the disease and in which said reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. [3] 3. In vitro method for the diagnosis of an inflammatory rheumatic disease in a subject, said method comprising steps a) and b) of claim 1, wherein an increase in the levels of anti-CD26 IgG antibodies in the subject's sample with respect to said reference value is indicative of disease and in which said reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. [4] Four. In vitro method for classifying subjects as i) healthy subjects or as ii) subjects having an inflammatory rheumatic disease, said method comprising steps a) and b) of claim 1, wherein an increase in the levels of anti-CD26 IgG antibodies in the subject's sample with respect to said reference value it is indicative of disease and in which said reference value corresponds to the average levels of anti-CD26 IgG antibodies in healthy subjects. [5] 5. In vitro method of disease monitoring and / or response monitoring toa treatment in a subject who has an inflammatory rheumatic disease,said method comprising steps a) and b) of claim 1,where an increase in the levels of anti-CD26 IgG antibodies in the subject's sample withwith respect to said reference value it is indicative of disease andwherein said reference value corresponds to the average levels of anti-antibodyCD26 IgG in healthy subjects. [6] 6. In vitro method for classifying a subject as a respondent to a treatment foran inflammatory rheumatic disease,said method comprises steps a) and b) of claim 1,where an increase in the levels of anti-CD26 IgG antibodies in the subject's sample withwith respect to said reference value it is indicative of the lack of response andwherein said reference value corresponds to the average levels of anti-antibodyCD26 IgG in healthy subjects. [7] 7. The method according to any one of claims 1 to 6, wherein therheumatic inflammatory disease is selected from the group consisting of arthritisrheumatoid, systemic lupus erythematosus, Sjögren's syndrome, juvenile rheumatoid arthritis,psoriatic arthritis and spondyloarthropathy. [8] 8. The method according to any one of claims 1 to 7, wherein saidInflammatory rheumatic disease is rheumatoid arthritis. [9] 9. The method according to any of claims 1 to 8, which methodIt comprises steps a) and b) of claim 1 and further comprising:a) the determination of the levels of anti-CD26 antibodies of IgM and / or IgA isotype in saidsample containing antibodies;b) compare the levels of anti-CD26 antibodies of IgM and / or IgA isotype in said samplecontaining antibodies with a reference value;where an increase in the value of anti-CD26 IgG antibody levels and levelsof the IgM and / or IgA isotypes in the subject's sample with respect to the reference valuecorresponding is indicative of disease;in which the reference value corresponds to the average levels in healthy subjects. [10] 10. The method according to any one of claims 1 to 9, wherein the levelsof certain anti-CD26 antibodies are selected from the group consisting of: i. anti-CD26 IgG antibody levels and anti-CD26 IgM antibody levels; ii. anti-CD26 IgG antibody levels and anti-CD26 IgA antibody levels and iii. anti-CD26 IgG antibody levels, anti-CD26 IgM antibody levels and anti-CD26 IgA antibody levels. [11] eleven. The method according to any one of claims 1 to 10, wherein the increase in said levels of anti-CD26 antibodies with respect to said reference value is at least 1.3 times, preferably at least 1.4 times and, more preferably, at least 1.5 times. [12] 12. The method according to any one of claims 1 to 11, wherein said subject is a human subject. [13] 13. The method according to any of claims 1 to 12, wherein said antibody-containing sample is whole blood, serum or plasma, preferably serum. [14] 14. The method according to any of claims 1 to 13, wherein the antibody levels are determined using an ELISA assay, an ELISA cell assay or any multiplex version thereof. [15] fifteen. The method according to any one of claims 1 to 14, wherein said anti-CD26 antibodies are specific antibodies against recombinant soluble CD26. [16] 16. The method according to any of claims 1 to 15, wherein said subject has not previously received or is not receiving treatment for said inflammatory rheumatic disease. [17] 17. The method according to any of claims 1 to 16, wherein said subject has previously received or is being treated for said inflammatory rheumatic disease, preferably, wherein said treatment is selected from the following group of drugs: to. Disease modifying anti-rheumatic drugs (FAME); b. anti-TNF alpha agents; C. anti-CD20 agents and d. anti-IL6R agents and / or Fc-CTLA4 fusion proteins. [18] 18. The method according to any one of claims 1 to 17, wherein said method further comprises the determination of other biomarkers associated with said inflammatory rheumatic disease, preferably, the biomarkers are selected from the group consisting of citrullinated anti-peptide antibodies (ACPA ), rheumatoid factor antibodies, anti-mannin-linked lectin antibodies (MBL), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), platelet count, hemoglobin and hematocrit levels. [19] 19. The method according to any of claims 1 to 18, wherein said method further comprises the determination of clinical parameters, wherein said clinical parameters are selected from the group consisting of morning stiffness, arthritis in three or more joints, involvement of the hand joint, symmetric arthritis, rheumatoid nodules, and radiographic changes. [20] twenty. The method according to any of claims 1 to 19, wherein said method further comprises storing the results of the method in a data carrier, preferably wherein said data carrier is a computer readable medium. [21] twenty-one. The method according to any of claims 1 to 20, characterized by being implemented in a computer. [22] 22 Use of a drug selected from the following list: to. Disease modifying anti-rheumatic drugs (FAME); b. Anti-TNF alpha agents; C. Anti-CD20 agents; Y d. Anti-IL6R agents and / or Fc-CTLA4 fusion proteins; for the preparation of a medicament for the treatment of a subject having an inflammatory rheumatic disease, wherein said subject has been selected using a method according to any one of claims 1 to 21. [23] 23. Kit comprising reagents for the determination of anti-CD26 antibody levels in an isolated sample of a subject containing antibodies, in which said kit comprises a. The following reagents for the determination of anti-CD26 antibody levels of the IgG and IgM isotypes: i. Purified or synthesized soluble CD26 (sCD26), an antigenic fragment thereof or a cell expressing CD26 or an antigenic fragment thereof; ii. a labeled anti-IgG antibody; Y iii. a labeled anti-IgM antibody; Y b. optionally, instructions for the use of said reagents in determining the levels of the IgG and IgM isotypes of the anti-CD26 antibodies in a sample containing antibodies isolated from said subject. [24] 24. The kit according to claim 23, wherein said kit further comprises: to. a reagent for the determination of the levels of anti-CD26 antibody of the IgA isotype, wherein said reagent consists of a labeled anti-IgA antibody; Y b. optionally, instructions for the use of said reagent in the determination of the levels of anti-CD26 antibodies of the IgA isotype in said sample containing antibodies. [25] 25. The kit according to any of claims 23 or 24, wherein said kit further comprises: to. a reagent to determine the total antibody levels of the IgM, IgG and / or IgA isotypes, wherein said reagent consists of: i. an anti-IgM, anti-IgG and / or anti-IgA antibody; Y ii. an anti-IgM, anti-IgG and / or labeled anti-IgA antibody; Y b. optionally, instructions for the use of said reagent in the determination of the total antibody levels of isotype IgM, IgG and / or IgA in said sample containing antibodies. [26] 26. Use of a kit for screening, diagnosis and / or monitoring of an inflammatory rheumatic disease and / or for monitoring the response to a treatment in a sample containing antibodies isolated from a subject, in which said kit comprises: to. The following reagents for the determination of the levels of anti-CD26 antibodies of the IgG isotype: i. Purified or synthesized soluble CD26 (sCD26), an antigenic fragment thereof or a cell expressing CD26 or an antigenic fragment thereof; Y ii. a labeled anti-IgG antibody; Y b. optionally, instructions for the use of said reagent in the determination of the levels of IgG isotype anti-CD26 antibodies in a sample containing antibodies isolated from said subject. [27] 27. The use of a kit according to claim 26, wherein said kit further comprises: 5 a. a labeled anti-IgM antibody; Y b. optionally, instructions for the use of said reagent in the determination of the levels of anti-CD26 antibodies of IgM isotype in said sample containing antibodies. [28] 28. The use of a kit according to any of claims 26 or 27, wherein said kit 10 further comprises: to. a labeled anti-IgA antibody; Y b. optionally, instructions for the use of said reagent in the determination of the levels of anti-CD26 antibodies of isotype IgA in said sample containing antibodies. The use of a kit according to any one of claims 26 to 28, wherein said kit further comprises: to. a reagent to determine the total antibody levels of isotype IgM, IgG and / or IgA, wherein said reagent consists of: i. an anti-IgM, anti-IgG and / or anti-IgA antibody; and 20 ii. an anti-IgM, anti-IgG and / or labeled anti-IgA antibody; Y b. optionally, instructions for the use of said reagent in the determination of the total antibody levels of isotype IgM, IgG and / or IgA in said sample containing antibodies. Fig. 1 Fig 2 Fig. 3 Fig. 4 Fig. 5 Fig. 6 Fig. 7 Fig. 8 Fig. 9 Fig. 10 Fig. 11 Fig. 12 Fig. 13 Fig. 14 Fig. 15 Fig. 16 <110> Servizo Galego de Saúde (SERGAS) University of Santiago de Compostela (USC) <120> USE OF ANTI-CD26 ANTIBODY LEVELS AS AUTOIMMUNE AND / OR INFLAMMATORY DISEASE BIOMARKERS <130> 901 050 <160> 2 <170> PatentIn version 3.5 <210> one <211> 766 <212> PRT <213> Homo sapiens <220> <221> Dipeptidyl peptidase 4 <222> (1) .. (766) <223> membrane form <400> one Met Lys Thr Pro Trp Lys Val Leu Leu Gly Leu Leu Gly Wing Wing Wing1 5 10 15 Leu Val Thr Ile Ile Thr Val Pro Val Val Leu Leu Asn Lys Gly Thr20 25 30 Asp Asp Wing Thr Wing Asp Ser Arg Lys Thr Tyr Thr Leu Thr Asp Tyr35 40 45 Leu Lys Asn Thr Tyr Arg Leu Lys Leu Tyr Ser Leu Arg Trp Ile Ser50 55 60 Asp His Glu Tyr Leu Tyr Lys Gln Glu Asn Asn Ile Leu Val Phe Asn65 70 75 80 Wing Glu Tyr Gly Asn Ser Ser Val Phe Leu Glu Asn Ser Thr Phe Asp85 90 95 Glu Phe Gly His Ser Ile Asn Asp Tyr Ser Ile Ser Pro Asp Gly Gln100 105 110 Phe Ile Leu Leu Glu Tyr Asn Tyr Val Lys Gln Trp Arg His Ser Tyr115 120 125 Thr Ala Ser Tyr Asp Ile Tyr Asp Leu Asn Lys Arg Gln Leu Ile Thr130 135 140 Glu Glu Arg Ile Pro Asn Asn Thr Gln Trp Val Thr Trp Ser Pro Val145 150 155 160 Gly His Lys Leu Wing Tyr Val Trp Asn Asn Asp Ile Tyr Val Lys Ile165 170 175one Glu Pro Asn Leu Pro Ser Tyr Arg Ile Thr Trp Thr Gly Lys Glu Asp180 185 190 Ile Ile Tyr Asn Gly Ile Thr Asp Trp Val Tyr Glu Glu Glu Val Phe195 200 205 Be Wing Tyr Be Wing Leu Trp Trp Be Pro Asn Gly Thr Phe Leu Wing210 215 220 Tyr Ala Gln Phe Asn Asp Thr Glu Val Pro Leu Ile Glu Tyr Ser Phe225 230 235 240 Tyr Ser Asp Glu Ser Leu Gln Tyr Pro Lys Thr Val Arg Val Pro Tyr245 250 255 Pro Lys Ala Gly Ala Val Asn Pro Thr Val Lys Phe Phe Val Val Asn260 265 270 Thr Asp Ser Leu Ser Ser Val Thr Asn Ala Thr Ser Ile Gln Ile Thr275 280 285 Pro Wing Wing Be Met Leu Ile Gly Asp His Tyr Leu Cys Asp Val Thr290 295 300 Trp Wing Thr Gln Glu Arg Ile Ser Leu Gln Trp Leu Arg Arg Ile Gln305 310 315 320 Asn Tyr Ser Val Met Asp Ile Cys Asp Tyr Asp Glu Ser Ser Gly Arg325 330 335 Trp Asn Cys Leu Val Ala Arg Gln His Ile Glu Met Ser Thr Thr Gly340 345 350 Trp Val Gly Arg Phe Arg Pro Ser Glu Pro His Phe Thr Leu Asp Gly355 360 365 Asn Ser Phe Tyr Lys Ile Ile Ser Asn Glu Glu Gly Tyr Arg His Ile370 375 380 Cys Tyr Phe Gln Ile Asp Lys Lys Asp Cys Thr Phe Ile Thr Lys Gly385 390 395 400 Thr Trp Glu Val Ile Gly Ile Glu Ala Leu Thr Ser Asp Tyr Leu Tyr405 410 415 Tyr Ile Ser Asn Glu Tyr Lys Gly Met Pro Gly Gly Arg Asn Leu Tyr420 425 430 Lys Ile Gln Leu Ser Asp Tyr Thr Lys Val Thr Cys Leu Ser Cys Glu435 440 445 2 Leu Asn Pro Glu Arg Cys Gln Tyr Tyr Ser Val Ser Phe Ser Lys Glu450 455 460 Wing Lys Tyr Tyr Gln Leu Arg Cys Ser Gly Pro Gly Leu Pro Leu Tyr465 470 475 480 Thr Leu His Ser Ser Val Asn Asp Lys Gly Leu Arg Val Leu Glu Asp485 490 495 Asn Ser Ala Leu Asp Lys Met Leu Gln Asn Val Gln Met Pro Ser Lys500 505 510 Lys Leu Asp Phe Ile Ile Leu Asn Glu Thr Lys Phe Trp Tyr Gln Met515 520 525 Ile Leu Pro Pro His Phe Asp Lys Ser Lys Lys Tyr Pro Leu Leu Leu530 535 540 Asp Val Tyr Wing Gly Pro Cys Ser Gln Lys Wing Asp Thr Val Phe Arg545 550 555 560 Leu Asn Trp Ala Thr Tyr Leu Ala Ser Thr Glu Asn Ile Ile Val Ala565 570 575 Ser Phe Asp Gly Arg Gly Ser Gly Tyr Gln Gly Asp Lys Ile Met His580 585 590 Wing Ile Asn Arg Arg Leu Gly Thr Phe Glu Val Glu Asp Gln Ile Glu595 600 605 Wing Wing Arg Gln Phe Ser Lys Met Gly Phe Val Asp Asn Lys Arg Ile610 615 620 Wing Ile Trp Gly Trp Ser Tyr Gly Gly Tyr Val Thr Ser Met Val Leu625 630 635 640 Gly Ser Gly Ser Gly Val Phe Lys Cys Gly Ile Ala Val Ala Pro Val Wing645 650 655 Ser Arg Trp Glu Tyr Tyr Asp Ser Val Tyr Thr Glu Arg Tyr Met Gly660 665 670 Leu Pro Thr Pro Glu Asp Asn Leu Asp His Tyr Arg Asn Ser Thr Val675 680 685 Met Ser Arg Ala Glu Asn Phe Lys Gln Val Glu Tyr Leu Leu Ile His690 695 700 Gly Thr Wing Asp Asp Asn Val His Phe Gln Gln Ser Wing Gln Ile Ser705 710 715 720 3 Lys Wing Leu Val Asp Val Gly Val Asp Phe Gln Wing Met Trp Tyr Thr725 730 735 Asp Glu Asp His Gly Ile Ala Ser Ser Thr Ala His Gln His Ile Tyr740 745 750 Thr His Met Ser His Phe Ile Lys Gln Cys Phe Ser Leu Pro755 760 765 <210> 2 <211> 727 <212> PRT <213> Homo sapiens <220> <221> Dipeptidyl peptidase 4 <222> (1) .. (727) <223> soluble form <400> 2 Arg Lys Thr Tyr Thr Leu Thr Asp Tyr Leu Lys Asn Thr Tyr Arg Leu1 5 10 15 Lys Leu Tyr Ser Leu Arg Trp Ile Ser Asp His Glu Tyr Leu Tyr Lys20 25 30 Gln Glu Asn Asn Ile Leu Val Phe Asn Wing Glu Tyr Gly Asn Ser Ser35 40 45 Val Phe Leu Glu Asn Ser Thr Phe Asp Glu Phe Gly His Ser Ile Asn50 55 60 Asp Tyr Ser Ile Ser Pro Asp Gly Gln Phe Ile Leu Leu Glu Tyr Asn65 70 75 80 Tyr Val Lys Gln Trp Arg His Ser Tyr Thr Ala Ser Tyr Asp Ile Tyr85 90 95 Asp Leu Asn Lys Arg Gln Leu Ile Thr Glu Glu Arg Ile Pro Asn Asn100 105 110 Thr Gln Trp Val Thr Trp Ser Pro Val Gly His Lys Leu Ala Tyr Val115 120 125 Trp Asn Asn Asp Ile Tyr Val Lys Ile Glu Pro Asn Leu Pro Ser Tyr130 135 140 Arg Ile Thr Trp Thr Gly Lys Glu Asp Ile Ile Tyr Asn Gly Ile Thr145 150 155 160 Asp Trp Val Tyr Glu Glu Glu Val Phe Ser Ala Tyr Ser Ala Leu Trp165 170 175 4 Trp Ser Pro Asn Gly Thr Phe Leu Wing Tyr Wing Gln Phe Asn Asp Thr180 185 190 Glu Val Pro Leu Ile Glu Tyr Ser Phe Tyr Ser Asp Glu Ser Leu Gln195 200 205 Tyr Pro Lys Thr Val Arg Val Pro Tyr Pro Lys Wing Gly Wing Val Asn210 215 220 Pro Thr Val Lys Phe Phe Val Val Asn Thr Asp Ser Leu Ser Ser Val225 230 235 240 Thr Asn Ala Thr Ser Ile Gln Ile Thr Ala Pro Ala Ser Met Leu Ile245 250 255 Gly Asp His Tyr Leu Cys Asp Val Thr Trp Ala Thr Gln Glu Arg Ile260 265 270 Ser Leu Gln Trp Leu Arg Arg Ile Gln Asn Tyr Ser Val Met Asp Ile275 280 285 Cys Asp Tyr Asp Glu Ser Ser Gly Arg Trp Asn Cys Leu Val Ala Arg290 295 300 Gln His Ile Glu Met Ser Thr Thr Gly Trp Val Gly Arg Phe Arg Pro305 310 315 320 Ser Glu Pro His Phe Thr Leu Asp Gly Asn Ser Phe Tyr Lys Ile Ile325 330 335 Ser Asn Glu Glu Gly Tyr Arg His Ile Cys Tyr Phe Gln Ile Asp Lys340 345 350 Lys Asp Cys Thr Phe Ile Thr Lys Gly Thr Trp Glu Val Ile Gly Ile355 360 365 Glu Ala Leu Thr Ser Asp Tyr Leu Tyr Tyr Ile Ser Asn Glu Tyr Lys370 375 380 Gly Met Pro Gly Gly Arg Asn Leu Tyr Lys Ile Gln Leu Ser Asp Tyr385 390 395 400 Thr Lys Val Thr Cys Leu Ser Cys Glu Leu Asn Pro Glu Arg Cys Gln405 410 415 Tyr Tyr Ser Val Ser Phe Ser Lys Glu Ala Lys Tyr Tyr Gln Leu Arg420 425 430 Cys Ser Gly Pro Gly Leu Pro Leu Tyr Thr Leu His Ser Ser Val Asn435 440 445 Asp Lys Gly Leu Arg Val Leu Glu Asp Asn Ser Ala Leu Asp Lys Met5 450 455 460 Leu Gln Asn Val Gln Met Pro Ser Lys Lys Leu Asp Phe Ile Ile Leu465 470 475 480 Asn Glu Thr Lys Phe Trp Tyr Gln Met Ile Leu Pro Pro His Phe Asp485 490 495 Lys Ser Lys Lys Tyr Pro Leu Leu Leu Asp Val Tyr Ala Gly Pro Cys500 505 510 Ser Gln Lys Wing Asp Thr Val Phe Arg Leu Asn Trp Wing Thr Tyr Leu515 520 525 Ala Ser Thr Glu Asn Ile Ile Val Ala Ser Phe Asp Gly Arg Gly Ser530 535 540 Gly Tyr Gln Gly Asp Lys Ile Met His Ala Ile Asn Arg Arg Leu Gly545 550 555 560 Thr Phe Glu Val Glu Asp Gln Ile Glu Ala Ala Arg Gln Phe Ser Lys565 570 575 Met Gly Phe Val Asp Asn Lys Arg Ile Ile Wing Trp Gly Trp Ser Tyr580 585 590 Gly Gly Tyr Val Thr Ser Met Val Leu Gly Ser Gly Ser Gly Val Phe595 600 605 Lys Cys Gly Ile Wing Val Wing Pro Val Ser Arg Trp Glu Tyr Tyr Asp610 615 620 Ser Val Tyr Thr Glu Arg Tyr Met Gly Leu Pro Thr Pro Glu Asp Asn625 630 635 640 Leu Asp His Tyr Arg Asn Ser Thr Val Met Ser Arg Ala Glu Asn Phe645 650 655 Lys Gln Val Glu Tyr Leu Leu Ile His Gly Thr Wing Asp Asp Asn Val660 665 670 His Phe Gln Gln Ser Ala Gln Ile Ser Lys Ala Leu Val Asp Val Gly675 680 685 Val Asp Phe Gln Wing Met Trp Tyr Thr Asp Glu Asp His Gly Ile Ala690 695 700 Ser Ser Thr Ala His Gln His Ile Tyr Thr His Met Ser His Phe Ile705 710 715 720 Lys Gln Cys Phe Ser Leu Pro7256
类似技术:
公开号 | 公开日 | 专利标题 Dolcino et al.2014|Crossreactive autoantibodies directed against cutaneous and joint antigens are present in psoriatic arthritis Sieghart et al.2018|Determination of autoantibody isotypes increases the sensitivity of serodiagnostics in rheumatoid arthritis WO2012039161A1|2012-03-29|Novel testing method and testing reagent for angiitis Vanhecke et al.2012|Identification of a major linear C1q epitope allows detection of systemic lupus erythematosus anti‐C1q antibodies by a specific peptide‐based enzyme‐linked immunosorbent assay EP2564202A1|2013-03-06|Methods for detecting antibodies Sato et al.2014|Serum level of soluble triggering receptor expressed on myeloid cells-1 as a biomarker of disease activity in relapsing polychondritis JP5914536B2|2016-05-11|Biomarkers for predicting responsiveness to anti-tumor necrosis factor alpha | treatment JP2018524585A|2018-08-30|Prognosis and monitoring of membranous nephropathy based on analysis of PLA2R1 epitope profile and PLA2R1 epitope spreading Kaga et al.2019|Comparison of measurements of anti-PLA2R antibodies in Japanese patients with membranous nephropathy using in-house and commercial ELISA Cha et al.2004|Autoantibodies to glucose‐6‐phosphate isomerase are elevated in the synovial fluid of rheumatoid arthritis patients JP2019508694A|2019-03-28|Methods and compositions for detecting and diagnosing kidney disease and periodontal disease ES2642723B1|2018-10-22|Use of anti-CD26 antibody levels as biomarkers of autoimmune and / or inflammatory diseases. JP2014506994A|2014-03-20|Diagnosis method of urinary tract infection US20110287458A1|2011-11-24|Method and composition for the diagnosis and monitoring of inflammatory diseases DK2406633T3|2019-04-01|COMPOSITIONS AND PROCEDURES FOR CHARACTERIZING ARTHRITIC CONDITIONS US11242408B2|2022-02-08|Monoclonal antibody specific for gamma-glutamyl-l-epsilon-lysine for the monitoring of apoptosis WO2014185145A1|2014-11-20|Biomarker for psychiatric and neurological disorders AU2010241416B1|2012-05-24|Cancer biomarkers KR102128251B1|2020-06-30|Biomarker Composition for Diagnosing Colorectal Cancer Specifically Binding to Arginine-methylated Dopamine Receptor D2 KR102131860B1|2020-07-08|Biomarker Composition for Diagnosing Colorectal Cancer Specifically Binding to Arginine-methylated Gamma-glutamyl Transferase 1 JP2021167805A|2021-10-21|Methods and Reagents for Diagnosing SARS-CoV-2 Infection JPWO2019098328A1|2020-12-24|IgA nephropathy diagnostic kit WO2009055880A2|2009-05-07|New method for diagnosing sjogren's syndrome JP6312054B2|2018-04-18|Biomarker for interstitial pneumonia CN114174827A|2022-03-11|Method for diagnosing colorectal cancer
同族专利:
公开号 | 公开日 WO2017194779A1|2017-11-16| EP3454842A1|2019-03-20| EP3454842B1|2020-07-08| US20200393461A1|2020-12-17| ES2642723B1|2018-10-22|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 WO1996038550A1|1995-06-01|1996-12-05|Dana-Farber Cancer Institute, Inc.|A novel form of dipeptidylpeptidase iv found in human serum, antibodies thereto, and uses therefor| US20060177886A1|2005-02-05|2006-08-10|Tadahiko Hazato|Method for diagnosis of rheumatoid arthritis| CA1261257A|1984-09-19|1989-09-26|Helgi Valdimarsson|Prognostic value of rheumatoid factor isotypes andtheir association with disease activity| EP1103610A1|1999-11-26|2001-05-30|Introgene B.V.|Production of vaccines from immortalised mammalian cell lines| JP4913587B2|2006-12-28|2012-04-11|雪印メグミルク株式会社|Peptide and protein quantification| WO2008146100A1|2007-06-01|2008-12-04|INSERM |Method for absolute quantification of polypeptides| EP2700945B1|2011-04-22|2016-11-02|Kyoto University|Use of myelin basic protein as a novel genetic factor for rheumatoid arthritis| ES2567276T3|2011-05-12|2016-04-21|Genentech, Inc.|LC-MS / MS method of monitoring multiple reactions to detect therapeutic antibodies in animal samples using frame-changing peptides|CN108226534B|2018-01-12|2020-06-30|广州市康润生物科技有限公司|Application of biomarker in judging phospholipid-resistant syndrome and kit thereof|
法律状态:
2018-10-22| FG2A| Definitive protection|Ref document number: 2642723 Country of ref document: ES Kind code of ref document: B1 Effective date: 20181022 |
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 ES201630620A|ES2642723B1|2016-05-12|2016-05-12|Use of anti-CD26 antibody levels as biomarkers of autoimmune and / or inflammatory diseases.|ES201630620A| ES2642723B1|2016-05-12|2016-05-12|Use of anti-CD26 antibody levels as biomarkers of autoimmune and / or inflammatory diseases.| PCT/EP2017/061537| WO2017194779A1|2016-05-12|2017-05-12|Use of anti-cd26 antibody levels as autoimmune and/or inflammatory disease biomarkers| EP17726564.2A| EP3454842B1|2016-05-12|2017-05-12|Use of anti-cd26 antibody levels as autoimmune and/or inflammatory disease biomarkers| US16/300,494| US20200393461A1|2016-05-12|2017-05-12|Use of anti-cd26 antibody levels as autoimmune and/or inflammatory disease biomarkers| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|