WO2017151755A1 - Biomarkers and methods for predicting response to discontinuation of inflammatory disease therapy - Google Patents

Biomarkers and methods for predicting response to discontinuation of inflammatory disease therapy Download PDF

Info

Publication number
WO2017151755A1
WO2017151755A1 PCT/US2017/020181 US2017020181W WO2017151755A1 WO 2017151755 A1 WO2017151755 A1 WO 2017151755A1 US 2017020181 W US2017020181 W US 2017020181W WO 2017151755 A1 WO2017151755 A1 WO 2017151755A1
Authority
WO
WIPO (PCT)
Prior art keywords
score
remission
subject
biomarkers
therapy
Prior art date
Application number
PCT/US2017/020181
Other languages
French (fr)
Inventor
Nadine Defranoux
Ching-chang HWANG
Wanying Li
Yoshiya Tanaka
Original Assignee
Crescendo Bioscience
University Of Occupational And Environmental Health, Japan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Crescendo Bioscience, University Of Occupational And Environmental Health, Japan filed Critical Crescendo Bioscience
Publication of WO2017151755A1 publication Critical patent/WO2017151755A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/101Diffuse connective tissue disease, e.g. Sjögren, Wegener's granulomatosis
    • G01N2800/102Arthritis; Rheumatoid arthritis, i.e. inflammation of peripheral joints
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/54Determining the risk of relapse

Definitions

  • RA Rheumatoid arthritis
  • RA Rheumatoid arthritis
  • the immune system of the RA subject targets the subject's joints as well as other organs including the lung, blood vessels and pericardium, leading to inflammation of the joints (arthritis), widespread endothelial inflammation, and even destruction of joint tissue. Erosions and joint space narrowing are largely irreversible and result in cumulative disability.
  • methotrexate (MTX), yet did not demonstrate any clinical improvement, as measured by DAS (Disease Activity Score) and CRP (C -reactive protein).
  • DAS Disease Activity Score
  • CRP C -reactive protein
  • Biological agents have provided unprecedented efficacy and improvement in long-term outcomes for patients RA.
  • these treatments can be associated with increased risk of serious infection and substantial costs, and in some patients lifelong treatment may not be desirable.
  • premature or improper withdrawal from therapy can result in disease flare, which runs the risk of irreversible joint damage. Therefore, there is interest in reducing or stopping biologic treatment in RA patients who achieve sustained remission.
  • the European League against Rheumatism (EULAR) and the American College of Rheumatology both recommend that tapering both non-biologic DMARDs and biological agents be considered for patients who are in persistent remission (Singh JA, et al, Arthritis Care Res (Hoboken).
  • the present teachings relate to biomarkers associated with inflammatory disease, and with autoimmune disease, including RA, and methods of using the biomarkers to measure disease activity in a subject, and in particular, in response to therapy.
  • a method for recommending a therapeutic regimen in a subject having an autoimmune disorder comprises a) placing the subject on a therapy regimen; b) determining whether the subject responds to the therapy regimen; c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatin
  • the at least four biomarkers comprise IL6, EGF, SAAl, and CRP. In an embodiment, the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAAl, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1.
  • performance of the at least one immunoassay comprises: obtaining the sample, wherein the sample comprises the protein markers; contacting the sample with a plurality of distinct reagents; generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data.
  • the immunoassays comprise a multiplex assay.
  • the autoimmune disorder is an arthritic disorder.
  • the arthritic disorder is rheumatoid arthritis.
  • the rheumatoid arthritis is early rheumatoid arthritis (eRA).
  • the therapy regimen comprises one or more of methotrexate (MTX) and a biologic.
  • the biologic comprises a TNF inhibitor.
  • the TNF inhibitor is adalimumab.
  • the score is predictive of a clinical assessment.
  • the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28- ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC).
  • the score is low if on a scale of 1-100, the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is greater than about 30. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of sustained clinical remission (SC-REM) and the score is less than or equal to about 23.
  • SC-REM sustained clinical remission
  • the score is high if on a scale of 1- 100, the score is predictive of SC-REM and the score is greater than about 23. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of functional remission and the score is less than or equal to about 26. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of functional remission and the score is greater than about 26.
  • the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission. In an embodiment, the remission is sustained if DAS28-ESR is ⁇ 2.6 for >24 weeks. In an embodiment, the remission is SC-REM. In an embodiment, the remission is functional remission. In an embodiment, the reduction of the therapy regimen is discontinuation of the therapy regimen.
  • a method for predicting flare or remission in a subject having an autoimmune disorder comprises: a) placing the subject on a therapy regimen; b) determining whether the subject responds to the therapy regimen; c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, pro
  • the at least four biomarkers comprise IL6, EGF, SAA1, and CRP. In an embodiment, the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAA1, VCAMl, CRP, MMP1, MMP3, T FRSF1A, RETN, and CHI3L1.
  • performance of the at least one immunoassay comprises: obtaining the sample, wherein the sample comprises the protein markers; contacting the sample with a plurality of distinct reagents; generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data.
  • the immunoassays comprise a multiplex assay.
  • the autoimmune disorder is an arthritic disorder.
  • the arthritic disorder is rheumatoid arthritis.
  • the rheumatoid arthritis is early rheumatoid arthritis (eRA).
  • the therapy regimen comprises one or more of methotrexate (MTX) and a biologic.
  • the biologic comprises a TNF inhibitor.
  • the TNF inhibitor is adalimumab.
  • the score is predictive of a clinical assessment.
  • the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28-ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC).
  • the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30.
  • the score is low if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is sustained clinical remission (SC-REM) and the score is less than or equal to about 23.
  • the score is high if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is SC-REM and the score is greater than about 23.
  • the score is low if on a scale of 1-100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is less than or equal to about 26.
  • the score is high if on a scale of 1- 100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is greater than about 26.
  • the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission.
  • the remission is sustained if DAS28-ESR is ⁇ 2.6 for >24 weeks.
  • the flare is DAS28-ESR>3.2.
  • the remission is sustained clinical remission (SC-REM).
  • the remission is functional remission. In an
  • the SC-REM is DAS28-ESR ⁇ 2.6.
  • the therapy regimen is not discontinued if flare or no remission is predicted.
  • Figure 1 depicts correlations of the MBDA algorithm predictions and CRP with clinical assessments of disease activity, as described in Example 1.
  • Figure 2 depicts the MBDA scores for subjects at baseline and six-month visits, according to the description in Example 1. MBDA scores are shown by treatment arm and time point. Only subjects with MBDA scores available at both baseline and six months are shown.
  • Figure 3A depicts the distribution of MBDA scores at ADA discontinuation for 3
  • Figure 4 depicts p-values by univariate logistic regression 6 months after ADA discontinuation compared with and without flare, clinical remission, and functional remission.
  • Figure 5 depicts p-values by univariate logistic regression 1 year after ADA discontinuation compared with and without flare, clinical remission, and functional remission.
  • Figure 6 depicts MBDA scores at ADA discontinuation: flare vs. no flare.
  • Figure 7 depicts flare rate by 3 MBDA categories. P-values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 7A depicts flare rate at 6 months post ADA discontinuation.
  • Figure 7B depicts flare rate at 1-year post ADA discontinuation.
  • Figure 8 depicts flare rate by 4 MBDA categories. P-values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 9 depicts MBDA scores at ADA discontinuation: SC-REM vs. no SC-
  • Figure 10 depicts sustained clinical remission rate by 3 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 11 depicts sustained clinical remission rate by 4 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 11 A depicts rates of SC-REM at 6 months.
  • Figure 1 IB depicts rates of SC-REM at 1 year.
  • Figure 13 depicts sustained functional remission rate by 3 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 14 depicts sustained functional remission rate by 4 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
  • Figure 15 depicts Kaplan-Meier estimates examining the time to flare show a lower probability of flare among subjects with low ( ⁇ 30) or moderate (30-44) MBDA scores at baseline, compared to those with high MBDA scores (>44).
  • Figure 16 depicts receiver operating characteristic (ROC) curves of clinical remission, flare, and functional remission at 6 months and 1 year after discontinuation of ADA.
  • ROC receiver operating characteristic
  • FIG 17 illustrates a high-level block diagram of a computer (1600). Illustrated are at least one processor (1602) coupled to a chipset (1604). Also coupled to the chipset (1604) are a memory (1606), a storage device (1608), a keyboard (1610), a graphics adapter (1612), a pointing device (1614), and a network adapter (1616). A display (1618) is coupled to the graphics adapter (1612). In one embodiment, the functionality of the chipset (1604) is provided by a memory controller hub 1620) and an I/O controller hub (1622). In another embodiment, the memory (1606) is coupled directly to the processor (1602) instead of the chipset (1604).
  • the storage device 1608 is any device capable of holding data, like a hard drive, compact disk readonly memory (CD-ROM), DVD, or a solid-state memory device.
  • the memory (1606) holds instructions and data used by the processor (1602).
  • the pointing device (1614) may be a mouse, track ball, or other type of pointing device, and is used in combination with the keyboard (1610) to input data into the computer system (1600).
  • the graphics adapter (1612) displays images and other information on the display (1618).
  • the network adapter (1616) couples the computer system (1600) to a local or wide area network.
  • the present teachings relate generally to the identification of biomarkers associated with subjects having inflammatory and/or autoimmune diseases, for example RA, and that are useful in determining or assessing disease activity, and in particular, in response to inflammatory disease therapy for recommending optimal therapy.
  • “Accuracy” refers to the degree that a measured or calculated value conforms to its actual value. “Accuracy” in clinical testing relates to the proportion of actual outcomes (true positives or true negatives, wherein a subject is correctly classified as having disease or as healthy/normal, respectively) versus incorrectly classified outcomes (false positives or false negatives, wherein a subject is incorrectly classified as having disease or as healthy/normal, respectively). Other and/or equivalent terms for “accuracy” can include, for example,
  • sensitivity in the context of the present teachings, refers to the repeatability and predictability of the measurement process. Analytical accuracy can be summarized in such measurements as, e.g., coefficients of variation (CV), and tests of concordance and calibration of the same samples or controls at different times or with different assessors, users, equipment, and/or reagents. See, e.g., R. Vasan, Circulation 2006, 113(19):2335-2362 for a summary of considerations in evaluating new biomarkers.
  • CV coefficients of variation
  • administering refers to the placement of a composition into a subject by a method or route that results in at least partial localization of the composition at a desired site such that a desired effect is produced.
  • Routes of administration include both local and systemic administration. Generally, local administration results in more of the composition being delivered to a specific location as compared to the entire body of the subject, whereas, systemic administration results in delivery to essentially the entire body of the subject.
  • algorithm encompasses any formula, model, mathematical equation, algorithmic, analytical or programmed process, or statistical technique or classification analysis that takes one or more inputs or parameters, whether continuous or categorical, and calculates an output value, index, index value or score.
  • algorithms include but are not limited to ratios, sums, regression operators such as exponents or coefficients, biomarker value
  • transformations and normalizations including, without limitation, normalization schemes that are based on clinical parameters such as age, gender, ethnicity, etc.
  • rules and guidelines including, without limitation, rules and guidelines, statistical classification models, and neural networks trained on populations.
  • statistical classification models including, without limitation, statistical classification models, and neural networks trained on populations.
  • linear and non-linear equations and statistical classification analyses to determine the relationship between (a) levels of biomarkers detected in a subject sample and (b) the level of the respective subject's disease activity.
  • analyte in the context of the present teachings can mean any substance to be measured, and can encompass biomarkers, markers, nucleic acids, electrolytes, metabolites, proteins, sugars, carbohydrates, fats, lipids, cytokines, chemokines, growth factors, proteins, peptides, nucleic acids, oligonucleotides, metabolites, mutations, variants, polymorphisms, modifications, fragments, subunits, degradation products and other elements.
  • APOAl as used herein can refer to the gene APOAl and also the protein ApoAI.
  • To “analyze” includes determining a value or set of values associated with a sample by measurement of analyte levels in the sample. “Analyze” may further comprise and comparing the levels against constituent levels in a sample or set of samples from the same subject or other subject(s).
  • the biomarkers of the present teachings can be analyzed by any of various conventional methods known in the art. Some such methods include but are not limited to: measuring serum protein or sugar or metabolite or other analyte level, measuring enzymatic activity, and measuring gene expression.
  • the term “antibody” refers to any immunoglobulin-like molecule that reversibly binds to another with the required selectivity.
  • the term includes any such molecule that is capable of selectively binding to a biomarker of the present teachings.
  • the term includes an immunoglobulin molecule capable of binding an epitope present on an antigen.
  • the term is intended to encompass not only intact immunoglobulin molecules, such as monoclonal and polyclonal antibodies, but also antibody isotypes, recombinant antibodies, bi-specific antibodies, humanized antibodies, chimeric antibodies, anti-idiopathic (anti-ID) antibodies, single-chain antibodies, Fab fragments, F(ab') fragments, fusion protein antibody fragments, immunoglobulin fragments, F v fragments, single chain F v fragments, and chimeras comprising an immunoglobulin sequence and any modifications of the foregoing that comprise an antigen recognition site of the required selectivity.
  • Autoimmune disease encompasses any disease, as defined herein, resulting from an immune response against substances and tissues normally present in the body.
  • autoimmune diseases include rheumatoid arthritis, early rheumatoid arthritis, axial spondylarthritis, juvenile idiopathic arthritis, seronegative spondyloarthropathies, ankylosing spondylitis, psoriatic arthritis, antiphospholipid antibody syndrome, autoimmune hepatitis, Behcet's disease, bullous pemphigoid, coeliac disease, Crohn's disease, dermatomyositis, Goodpasture's syndrome, Graves' disease, Hashimoto's disease, idiopathic thrombocytopenic purpura, IgA nephropathy, Kawasaki disease, systemic lupus erythematosus, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, polymyositis, primary biliary cirrhosis, psoriasis, scleroderma, Sjogren's syndrome, ulcerative co
  • a “biologic” or “biotherapy” or “biopharmaceutical” is a pharmaceutical therapy product manufactured or extracted from a biological substance.
  • a biologic can include vaccines, blood or blood components, allergenics, somatic cells, gene therapies, tissues, recombinant proteins, and living cells; and can be composed of sugars, proteins, nucleic acids, living cells or tissues, or combinations thereof.
  • biologic drugs can include but are not limited to biological agents that target the tumor necrosis factor (T F)-alpha molecules and the TNF inhibitors, such as infliximab, adalimumab, etanercept and golimumab.
  • Other classes of biologic drugs include IL1 inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab.
  • Biomarker in the context of the present teachings encompasses, without limitation, cytokines, chemokines, growth factors, proteins, peptides, nucleic acids, oligonucleotides, and metabolites, together with their related metabolites, mutations, isoforms, variants, polymorphisms, modifications, fragments, subunits, degradation products, elements, and other analytes or sample-derived measures.
  • Biomarkers can also include mutated proteins, mutated nucleic acids, variations in copy numbers and/or transcript variants.
  • Biomarkers also encompass non-blood borne factors and non-analyte physiological markers of health status, and/or other factors or markers not measured from samples (e.g., biological samples such as bodily fluids), such as clinical parameters and traditional factors for clinical assessments. Biomarkers can also include any indices that are calculated and/or created mathematically. Biomarkers can also include combinations of any one or more of the foregoing measurements, including temporal trends and differences.
  • Biomarkers can include, but are not limited to, apolipoprotein A-I (APOA1); apolipoprotein C-III (APOC3); calprotectin; chemokine (C-C motif) ligand 22 (CCL22); chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1, or YKL- 40); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); intercellular adhesion molecule 1 (ICAMl); ICTP; interleukin 18 (interferon-gamma- inducing factor) (IL18); interleukin 1, beta (IL1B); interleukin 6 receptor (IL6R); interleukin 8 (IL8); keratan sulfate, or KS; leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallo
  • a "clinical assessment,” or “clinical datapoint” or “clinical endpoint,” in the context of the present teachings can refer to a measure of disease activity or severity.
  • a clinical assessment can include a score, a value, or a set of values that can be obtained from evaluation of a sample (or population of samples) from a subject or subjects under determined conditions.
  • a clinical assessment can also be a questionnaire completed by a subject.
  • a clinical assessment can also be predicted by biomarkers and/or other parameters.
  • the clinical assessment for RA can comprise, without limitation, one or more of the following: DAS (defined herein), DAS28, DAS28-ESR, DAS28-CRP, health assessment questionnaire (HAQ), modified HAQ (mHAQ), multi-dimensional HAQ (MDHAQ), visual analog scale (VAS), physician global assessment VAS, patient global assessment VAS, pain VAS, fatigue VAS, overall VAS, sleep VAS, simplified disease activity index (SDAI), clinical disease activity index (CDAI), routine assessment of patient index data (RAPID), RAPID3, RAPID4, RAPID 5, American College of Rheumatology (ACR), ACR20, ACR50, ACR70, SF-36 (a well-validated measure of general health status), RA MRI score (RAMRIS; or RA MRI scoring system), total Sharp score (TSS), van der Heijde-modified TSS, van der Heijde- modified Sharp score (or Sharp-van der Heijde score (
  • a clinical parameter can be used to derive a clinical assessment of the subject's disease activity.
  • Clinical parameters can include, without limitation: therapeutic regimen (including but not limited to DMARDs, whether conventional or biologies, steroids, etc.), TJC, SJC, morning stiffness, arthritis of three or more joint areas, arthritis of hand joints, symmetric arthritis, rheumatoid nodules, radiographic changes and other imaging, gender/sex, age, race/ethnicity, disease duration, diastolic and systolic blood pressure, resting heart rate, height, weight, body-mass index, family history, CCP status ⁇ i.e., whether subject is positive or negative for anti-CCP antibody), CCP titer, RF status, RF titer, ESR, CRP titer, menopausal status, and whether a smoker/non-smoker.
  • therapeutic regimen including but not limited to DMARDs, whether conventional or biologies, steroids, etc.
  • CRP concentration can be used as a clinical assessment of disease activity; or, it can be used as a measure of the health status of a subject, and thus serve as a clinical parameter.
  • FIG. 15 is a high-level block diagram of a computer (1600).
  • a "computer” can have different and/or other components than those shown in FIG. 15.
  • the computer 1600 can lack certain illustrated components.
  • the storage device (1608) can be local and/or remote from the computer (1600) (such as embodied within a storage area network (SAN)).
  • the computer (1600) is adapted to execute computer program modules for providing functionality described herein.
  • the term "module” refers to computer program logic utilized to provide the specified functionality.
  • a module can be implemented in hardware, firmware, and/or software.
  • program modules are stored on the storage device (1608), loaded into the memory (1606), and executed by the processor (1602).
  • Embodiments of the entities described herein can include other and/or different modules than the ones described here.
  • the processor 1602
  • modules can be performed by other or different modules in other embodiments. Moreover, this description occasionally omits the term "module" for purposes of clarity and convenience.
  • cytokine in the present teachings refers to any substance secreted by specific cells that can be of the immune system that carries signals between cells and thus has an effect on other cells.
  • cytokines encompasses "growth factors.”
  • Cyokines are also cytokines. They are a subset of cytokines that are able to induce chemotaxis in cells; thus, they are also known as “chemotactic cytokines.”
  • DAS Disease Activity Score
  • DAS28 involves the evaluation of 28 specific joints. It is a current standard well- recognized in research and clinical practice. Because the DAS28 is a well-recognized standard, it may be referred to as "DAS.”
  • DAS may refer to calculations based on 66/68 or 44 joint counts, unless otherwise specified, “DAS” herein will encompass the DAS28.
  • DAS28 can refer to a DAS28-ESR or DAS28-CRP, as obtained by any of the four formulas described above; or, DAS28 can refer to another reliable DAS28 formula as may be known in the art.
  • a DAS28 can be calculated for an RA subject according to the standard as outlined at the das-score. nl website, maintained by the Department of Rheumatology of the University Medical Centre in Nijmegen, the Netherlands. The number of swollen joints, or swollen joint count out of a total of 28 (SJC28), and tender joints, or tender joint count out of a total of 28 (TJC28) in each subject is assessed.
  • GH general health
  • VAS 100mm Visual Analogue Scale
  • DAS28-CRP is a DAS28 assessment calculated using CRP in place of ESR (see below).
  • CRP is produced in the liver. Normally there is little or no CRP circulating in an individual's blood serum - CRP is generally present in the body during episodes of acute inflammation or infection, so that a high or increasing amount of CRP in blood serum can be associated with acute infection or inflammation.
  • a blood serum level of CRP greater than 1 mg/dL is usually considered high. Most inflammation and infections result in CRP levels greater than 10 mg/dL.
  • the amount of CRP in subject sera can be quantified using, for example, the DSL- 10-42100 ACTIVE ® US C-Reactive Protein Enzyme-Linked Immunosorbent Assay (ELISA), developed by Diagnostics Systems Laboratories, Inc. (Webster, TX).
  • ELISA C-Reactive Protein Enzyme-Linked Immunosorbent Assay
  • CRP production is associated with radiological progression in RA. See M. Van Leeuwen et al, Br. J. Rheum. 1993, 32(suppl.):9-13).
  • CRP is thus considered an appropriate alternative to ESR in measuring RA disease activity. See R. Mallya et al, J. Rheum. 1982, 9(2):224-228, and F. Wolfe, J.
  • the DAS28-CRP can be calculated according to either of the formulas below, with or without the GH factor, where "CRP” represents the amount of this protein present in a subject's blood serum in mg/L, “sqrt” represents the square root, and “In” represents the natural logarithm:
  • the "DAS28-ESR” is a DAS28 assessment wherein the ESR for each subject is also measured (in mm/hour).
  • the DAS28-ESR can be calculated according to the formula:
  • a “dataset” is a set of numerical values resulting from evaluation of a sample (or population of samples) under a desired condition.
  • the values of the dataset can be obtained, for example, by experimentally obtaining measures from a sample and constructing a dataset from these measurements; or alternatively, by obtaining a dataset from a service provider such as a laboratory, or from a database or a server on which the dataset has been stored.
  • a “difference” as used herein refers to an increase or decrease in the measurable expression of a biomarker or panel of biomarkers as compared to the measurable expression of the same biomarker or panel of biomarkers in a second samples.
  • disease in the context of the present teachings encompasses any disorder, condition, sickness, ailment, etc. that manifests in, e.g., a disordered or incorrectly functioning organ, part, structure, or system of the body, and results from, e.g., genetic or developmental errors, infection, poisons, nutritional deficiency or imbalance, toxicity, or unfavorable environmental factors.
  • a DMARD can be conventional or biologic.
  • DMARDs that are generally considered conventional include, but are not limited to, MTX, azathioprine (AZA), bucillamine (BUC), chloroquine (CQ), ciclosporin (CSA, or cyclosporine, or cyclosporin), doxycycline (DOXY), hydroxychloroquine (HCQ), intramuscular gold (IM gold), leflunomide (LEF), levofloxacin (LEV), and sulfasalazine (SSZ).
  • MTX azathioprine
  • BUC bucillamine
  • CQ chloroquine
  • CQ chloroquine
  • CSA ciclosporin
  • DOXY hydroxychloroquine
  • HCQ hydroxychloroquine
  • IM gold intramuscular gold
  • LEF leflunomide
  • LEV levofloxacin
  • SSZ sulfasalazine
  • biologic DMARDs include but are not limited to biological agents that target the tumor necrosis factor (TNF)-alpha molecules such as infliximab, adalimumab, etanercept and golimumab.
  • TNF tumor necrosis factor
  • Other classes of biologic DMARDs include IL1 inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab.
  • the term "flare” as used herein is a sudden and severe increase in the onset of symptoms and clinical manifestations including, but not limited to, an increase in SJC, increase in TJC, increase in serologic markers of inflammation (e.g., CRP and ESR), decrease in subject function (e.g., ability to perform basic daily activities), increase in morning stiffness, and increases in pain that commonly lead to therapeutic intervention and potentially to treatment intensification.
  • an increase in SJC increase in TJC
  • increase in serologic markers of inflammation e.g., CRP and ESR
  • subject function e.g., ability to perform basic daily activities
  • increase in morning stiffness e.g., ability to perform basic daily activities
  • increases in pain that commonly lead to therapeutic intervention and potentially to treatment intensification.
  • an "immunoassay” as used herein refers to a biochemical assay that uses one or more antibodies to measure the presence or concentration of an analyte or biomarker in a biological sample.
  • Inflammatory disease in the context of the present teachings encompasses, without limitation, any disease, as defined herein, resulting from the biological response of vascular tissues to harmful stimuli, including but not limited to such stimuli as pathogens, damaged cells, irritants, antigens and, in the case of autoimmune disease, substances and tissues normally present in the body.
  • Non-limiting examples of inflammatory disease include rheumatoid arthritis (RA), eRA, ankylosing spondylitis, psoriatic arthritis, atherosclerosis, asthma, autoimmune diseases, chronic inflammation, chronic prostatitis, glomerulonephritis, hypersensitivities, inflammatory bowel diseases, pelvic inflammatory disease, reperfusion injury, transplant rejection, and vasculitis.
  • RA rheumatoid arthritis
  • eRA ankylosing spondylitis
  • psoriatic arthritis atherosclerosis
  • asthma autoimmune diseases, chronic inflammation, chronic prostatitis, glomerulonephritis, hypersensitivities, inflammatory bowel diseases, pelvic inflammatory disease, reperfusion injury, transplant rejection, and vasculitis.
  • Interpretation function means the transformation of a set of observed data into a meaningful determination of particular interest; e.g., an interpretation function may be a predictive model that is created by utilizing one or more statistical algorithms to transform a dataset of observed biomarker data into a meaningful determination of disease activity or the disease state of a subject.
  • Measurement in the context of the present teachings refers to determining the presence, absence, quantity, amount, or effective amount of a substance in a clinical or subject-derived sample, including the concentration levels of such substances, or evaluating the values or categorization of a subject's clinical parameters.
  • a “multiplex assay” as used herein refers to an assay that simultaneously measure multiple analytes, e.g., protein analytes, in a single run or cycle of the assay.
  • Performance in the context of the present teachings relates to the quality and overall usefulness of, e.g., a model, algorithm, or diagnostic or prognostic test.
  • Factors to be considered in model or test performance include, but are not limited to, the clinical and analytical accuracy of the test, use characteristics such as stability of reagents and various components, ease of use of the model or test, health or economic value, and relative costs of various reagents and components of the test. Performing can mean the act of carrying out a function.
  • a "population" is any grouping of subjects of like specified characteristics.
  • the grouping could be according to, for example but without limitation, clinical parameters, clinical assessments, therapeutic regimen, disease status (e.g. with disease or healthy), level of disease activity, etc.
  • an aggregate value can be determined based on the observed MBDA scores of the subjects of a population; e.g., at particular timepoints in a longitudinal study.
  • the aggregate value can be based on, e.g., any mathematical or statistical formula useful and known in the art for arriving at a meaningful aggregate value from a collection of individual datapoints; e.g., mean, median, median of the mean, etc.
  • multivariate model or simply a “model,” is a mathematical construct developed using a statistical algorithm or algorithms for classifying sets of data.
  • the term “predicting” refers to generating a value for a datapoint without actually performing the clinical diagnostic procedures normally or otherwise required to produce that datapoint; "predicting” as used in this modeling context should not be understood solely to refer to the power of a model to predict a particular outcome.
  • Predictive models can provide an interpretation function; e.g., a predictive model can be created by utilizing one or more statistical algorithms or methods to transform a dataset of observed data into a meaningful determination of disease activity or the disease state of a subject. See Calculation of the MBDA score for some examples of statistical tools useful in model development.
  • a "prognosis” is a prediction as to the likely outcome of a disease. Prognostic estimates are useful in, e.g., determining an appropriate therapeutic regimen for a subject.
  • a “quantitative dataset” or “quantitative data” as used in the present teachings refers to the data derived from, e.g., detection and composite measurements of expression of a plurality of biomarkers ⁇ i.e., two or more) in a subject sample.
  • the quantitative dataset can be used to generate a score for the identification, monitoring and treatment of disease states, and in characterizing the biological condition of a subject. It is possible that different biomarkers will be detected depending on the disease state or physiological condition of interest.
  • Recommending refers to making a recommendation for a therapeutic regimen or excluding ⁇ i.e., not recommending) a certain therapeutic regimen for a subject. Such a recommendation shall serve optionally together with other information as a basis for a clinician to apply a certain therapeutic regimen for an individual subject.
  • remission refers to the state of absence of disease activity in patients known to have a chronic illness that usually cannot be cured.
  • sustained clinical remission or “SC-REM” as used herein refers to a state of clinical remission sustained as evaluated based on clinical assessments, for example, DAS28 for at least six months.
  • functional remission refers to a state of remission as evaluated using functional assessment measures such as but not limited to HAQ. Sustained remission can be used interchangeably with maintained remission.
  • sample in the context of the present teachings refers to any biological sample that is isolated from a subject.
  • a sample can include, without limitation, a single cell or multiple cells, fragments of cells, an aliquot of body fluid, whole blood, platelets, serum, plasma, red blood cells, white blood cells or leucocytes, endothelial cells, tissue biopsies, synovial fluid, lymphatic fluid, ascites fluid, and interstitial or extracellular fluid.
  • sample also encompasses the fluid in spaces between cells, including synovial fluid, gingival crevicular fluid, bone marrow, cerebrospinal fluid (CSF), saliva, mucous, sputum, semen, sweat, urine, or any other bodily fluids.
  • Bood sample can refer to whole blood or any fraction thereof, including blood cells, red blood cells, white blood cells or leucocytes, platelets, serum and plasma.
  • Samples can be obtained from a subject by means including but not limited to venipuncture, excretion, ejaculation, massage, biopsy, needle aspirate, lavage, scraping, surgical incision, or intervention or other means known in the art.
  • a “score” is a value or set of values selected so as to provide a quantitative measure of a variable or characteristic of a subject's condition, and/or to discriminate, differentiate or otherwise characterize a subject's condition.
  • the value(s) comprising the score can be based on, for example, quantitative data resulting in a measured amount of one or more sample constituents obtained from the subject, or from clinical parameters, or from clinical assessments, or any combination thereof.
  • the score can be derived from a single constituent, parameter or assessment, while in other embodiments the score is derived from multiple constituents, parameters and/or assessments.
  • the score can be based upon or derived from an interpretation function; e.g., an interpretation function derived from a particular predictive model using any of various statistical algorithms known in the art.
  • a "change in score” can refer to the absolute change in score, e.g. from one time point to the next, or the percent change in score, or the change in the score per unit time (e.g., the rate of score change).
  • a “score” as used herein can be used interchangeably with MBDA score as defined below.
  • a "multi-biomarker disease activity score,” "MBDA score,” or simply “MBDA,” in the context of the present teachings, is a score that uses quantitative data to provide a quantitative measure of inflammatory disease activity or the state of inflammatory disease in a subject.
  • a set of data from particularly selected biomarkers, such as from the disclosed set of biomarkers, is input into an interpretation function according to the present teachings to derive the MBDA score.
  • the interpretation function in some embodiments, can be created from predictive or multivariate modeling based on statistical algorithms. Input to the interpretation function can comprise the results of testing two or more of the disclosed set of biomarkers, alone or in combination with clinical parameters and/or clinical assessments, also described herein.
  • the MBDA score is a quantitative measure of autoimmune disease activity. In some embodiments, the MBDA score is a quantitative measure of RA disease activity. MBDA as used herein can also refer to a VDA or VECTRA ® DA score.
  • a “multiplex assay” as used herein refers to an assay that simultaneously measures multiple analytes, e.g., protein analytes, in a single run or cycle of the assay.
  • Statistically significant in the context of the present teachings means an observed alteration is greater than what would be expected to occur by chance alone (e.g., a "false positive”).
  • Statistical significance can be determined by any of various methods well- known in the art.
  • An example of a commonly used measure of statistical significance is the p- value.
  • the p-value represents the probability of obtaining a given result equivalent to a particular datapoint, where the datapoint is the result of random chance alone.
  • a result is often considered highly significant (not random chance) at a p-value less than or equal to 0.05.
  • a "subject” in the context of the present teachings is generally a mammal.
  • the subject can be a patient.
  • the term "mammal” as used herein includes but is not limited to a human, non-human primate, dog, cat, mouse, rat, cow, horse, and pig. Mammals other than humans can be advantageously used as subjects that represent animal models of inflammation.
  • a subject can be male or female.
  • a subject can be one who has been previously diagnosed or identified as having an inflammatory disease.
  • a subject can be one who has already undergone, or is undergoing, a therapeutic intervention for an inflammatory disease.
  • a subject can also be one who has not been previously diagnosed as having an inflammatory disease; e.g., a subject can be one who exhibits one or more symptoms or risk factors for an inflammatory condition, or a subject who does not exhibit symptoms or risk factors for an inflammatory condition, or a subject who is asymptomatic for inflammatory disease.
  • a "therapeutic regimen,” “therapy” or “treatment(s),” as described herein, includes all clinical management of a subject and interventions, whether biological, chemical, physical, or a combination thereof, intended to sustain, ameliorate, improve, or otherwise alter the condition of a subject. These terms may be used synonymously herein.
  • Treatments include but are not limited to administration of prophylactics or therapeutic compounds (including conventional DMARDs, biologic DMARDs, non-steroidal anti -inflammatory drugs (NSAID's) such as COX-2 selective inhibitors, and corticosteroids), exercise regimens, physical therapy, dietary modification and/or supplementation, bariatric surgical intervention, administration of pharmaceuticals and/or anti-inflammatories (prescription or over-the-counter), and any other treatments known in the art as efficacious in preventing, delaying the onset of, or ameliorating disease.
  • a "response to treatment” includes a subject's response to any of the above-described treatments, whether biological, chemical, physical, or a combination of the foregoing.
  • treatment course relates to the dosage, duration, extent, etc. of a particular treatment or therapeutic regimen.
  • An initial therapeutic regimen as used herein is the first line of treatment.
  • a "time point” as used herein refers to a manner of describing a time, which can be substantially described with a single point.
  • a time point may also be described as a time range of a minimal unit which can be detected.
  • a time point can refer to a state of the aspect of a time or a manner of description of a certain period of time. Such a time point or range can include, for example, an order of seconds, minutes to hours, or days.
  • biomarkers can be used in the derivation of a MBDA score, as described herein, which MBDA score can be used to provide diagnosis, prognosis and monitoring of disease state and/or disease activity in inflammatory disease and in autoimmune disease.
  • the MBDA score can be used to provide diagnosis, prognosis and monitoring of disease state and/or disease activity of RA or early RA in response to therapy.
  • the MBDA score can be used to recommend discontinuation of a therapeutic regimen, or the MBDA score can be used to recommend no change in a therapeutic regimen.
  • the MBDA score can be used to predict disease activity, such as flare or remission, in a subject having an autoimmune disorder.
  • the optimal cutoff for flare, sustained clinical remission, and functional remission 6 months after discontinuation is an MBDA score of 30, 23, and 26, respectively.
  • the optimal cutoff for flare, sustained clinical remission, and functional remission 12 months after discontinuation is an MBDA score of 26, 25, and 27, respectively.
  • Identifying the state of inflammatory disease in a subject allows for a prognosis of the disease, and thus for the informed selection of, initiation of, adjustment of or increasing or decreasing various therapeutic regimens in order to delay, reduce or prevent that subject's progression to a more advanced disease state.
  • subjects can be identified as having a particular level of inflammatory disease activity and/or as being at a particular state of disease, or flare, based on the determination of their MBDA scores, and so can be selected to begin or accelerate treatment, as treatment is defined herein, to prevent or delay the further progression of inflammatory disease.
  • subjects that are identified via their MBDA scores as having a particular level of inflammatory disease activity, and/or as being at a particular state of inflammatory disease can be selected to have their treatment decreased or discontinued, where improvement or remission in the subject is seen.
  • subjects that are identified via their MBDA scores as having a particular level of inflammatory disease activity, and/or as being at a particular state of inflammatory disease can have therapy selected based on disease activity levels.
  • RA treatment provide a means for more profound disease management and optimal treatment of RA within the first months of symptom onset, which in turn result in significantly improved outcomes. See F. Wolfe, Arth. Rheum. 2000, 43(12):2751-2761; M. Matucci-Cerinic, Clin. Exp. Rheum. 2002, 20(4):443-444; and, V. Nell et. al, Lancet 2005, 365(9455): 199-200.
  • multi- biomarker algorithms can be derived from the disclosed set of biomarkers.
  • the MBDA score derived as described herein, can be used to rate inflammatory disease activity; e.g., as high, medium or low.
  • the score can be varied based on a set of values chosen by the practitioner. For example, a score can be set such that a value is given a range from 0-100, and a difference between two scores would be a value of at least one point.
  • the practitioner can then assign disease activity based on the values. For example, in some embodiments a score of about 1 to 29 represents a low level of disease activity, a score of about 30 to 44 represents a moderate level of disease activity, and a score of about 45 to 100 represents a high level of disease activity.
  • a score of ⁇ 38 can represent a low or lower score, and a score of >38 can represent a high or higher score.
  • a score of ⁇ 30 can represent a low or lower score, and a score of >30 can represent a high or higher score.
  • an MBDA scores of about ⁇ 25 is remission, about 26-29 is low, about 30-44 is moderate, and about >44 is high.
  • the cutoffs can vary.
  • a low score can be a score of ⁇ 30, although for other utilities, a low score can be a score of ⁇ 29 or ⁇ 31.
  • the disease activity score can also change based on the range of the score.
  • a score of 1 to 58 can represent a low level of disease activity when a range of 0-200 is utilized. Differences can be determined based on the range of score possibilities. For example, if using a score range of 0-100, a small difference in scores can be a difference of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 points; a moderate difference in scores can be a difference of about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 points; and large differences can be a change in about 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, or 50 points.
  • a practitioner can define a small difference in scores as about ⁇ 6 points, a moderate difference in scores as about 7-20 points, and a large difference in scores as about >20 points.
  • the difference can be expressed by any unit, for example, percentage points.
  • a practitioner can define a small difference as about ⁇ 6 percentage points, moderate difference as about 7-20 percentage points, and a large difference as about >20 percentage points.
  • autoimmune disease activity can be so rated.
  • RA disease activity can be so rated.
  • the MBDA score correlates well with traditional clinical assessments of inflammatory disease activity, e.g. in RA
  • bone damage itself in a subject or population, and thus disease progression can be tracked via the use and application of the MBDA score.
  • the MBDA score can be used for several purposes. On a subject-specific basis, it provides a context for understanding the relative level of disease activity.
  • the MBDA rating of disease activity can be used, e.g., to guide the clinician in determining treatment, in setting a treatment course, and/or to inform the clinician that the subject is in remission. Moreover, it provides a means to more accurately assess and document the qualitative level of disease activity in a subject. It is also useful from the perspective of assessing clinical differences among populations of subjects within a practice. For example, this tool can be used to assess the relative efficacy of different treatment modalities. Moreover, it is also useful from the perspective of assessing clinical differences among different practices. This would allow physicians to determine what global level of disease control is achieved by their colleagues, and/or for healthcare management groups to compare their results among different practices for both cost and comparative effectiveness. Because the MBDA score demonstrates strong association with established disease activity assessments, such as the DAS28, the MBDA score can provide a quantitative measure for monitoring the extent of subject disease activity, and response to treatment.
  • Certain embodiments of the present teachings can also be used to screen subject populations in any number of settings.
  • a health maintenance organization, public health entity or school health program can screen a group of subjects to identify those requiring interventions, as described above.
  • Other embodiments of these teachings can be used to collect disease activity data on one or more populations of subjects, to identify subject disease status in the aggregate, in order to, e.g., determine the effectiveness of the clinical management of a population, or determine gaps in clinical management.
  • Insurance companies e.g., health, life, or disability
  • Such data arrays or collections can be stored in machine-readable media and used in any number of health-related data management systems to provide improved healthcare services, cost-effective healthcare, and improved insurance operation, among other things. See, e.g., U.S. Patent Application No. 2002/0038227; U.S. Patent Application No. 2004/0122296; U.S. Patent Application No. 2004/0122297; and U.S. Pat. No. 5,018,067.
  • Such systems can access the data directly from internal data storage or remotely from one or more data storage sites as further detailed herein.
  • various embodiments of the present teachings provide an improvement comprising the use of a data array encompassing the biomarker measurements as defined herein, and/or the resulting evaluation of disease status and activity from those biomarker
  • inflammatory disease activity in a subject is measured by: determining the levels in inflammatory disease subject serum of two or more biomarkers, then applying an interpretation function to transform the biomarker levels into a single MBDA score, which provides a quantitative measure of inflammatory disease activity in the subject, correlating well with traditional clinical assessments of inflammatory disease activity ⁇ e.g., a DAS28 or CDAI score in RA), as is demonstrated in the Examples below.
  • the disease activity so measured relates to an autoimmune disease.
  • the disease activity so measured relates to RA.
  • the biomarkers can include apolipoprotein A-I (APOAl); apolipoprotein C-III (APOC3); calprotectin; chemokine (C-C motif) ligand 22 (CCL22); chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1, or YKL-40); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); intercellular adhesion molecule 1 (ICAMl); ICTP; interleukin 18 (interferon-gamma-inducing factor) (IL18); interleukin 1, beta (IL1B); interleukin 6 receptor (IL6R); interleukin 8 (IL8); keratan sulfate, or KS; leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase)
  • APOAl apolipoprotein A-I
  • MMP1 matrix metallopeptidase 3 (stromelysin 1, progelatinase)
  • MMP3 matrix metallopeptidase 3 (stromelysin 1, progelatinase)
  • PYD pyridinoline (crosslinks formed in collagen, derived from three lysine residues), which may be referred to herein as PYD; resistin (RETN); serum amyloid Al (SAA1); tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA).
  • FOGFA vascular endothelial growth factor A
  • ICTP is the carboxyterminal telopeptide region of type I collagen, and is liberated during the degradation of mature type I collagen.
  • Type I collagen is present as fibers in tissue; in bone, the type I collagen molecules are crosslinked.
  • the ICTP peptide is immunochemically intact in blood. (For the type I collagen gene, see official symbol COL1 Al, HUGO Gene Nomenclature Committee; also known as 014; alpha 1 type I collagen; collagen alpha 1 chain type I; collagen of skin, tendon and bone, alpha- 1 chain; and, pro-alpha-1 collagen type 1).
  • Keratan sulfate is not the product of a discrete gene, but refers to any of several sulfated glycosaminoglycans. They are synthesized in the central nervous system, and are found especially in cartilage and bone. Keratan sulfates are large, highly hydrated molecules, which in joints can act as a cushion to absorb mechanical shock.
  • the biomarkers can comprise two, or three, or four, or five, or six, or seven, or eight, or nine, or ten, or eleven, or twelve of IL6, EGF, VEGFA, LEP, SAA1, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1.
  • the biomarkers can comprise at least four biomarkers that comprise IL6, EGF, SAA1, and CRP.
  • the interpretation function is based on a predictive model.
  • ANOVA Bayesian networks; boosting and Ada-boosting; bootstrap aggregating (or bagging) algorithms; decision trees classification techniques, such as Classification and Regression Trees (CART), boosted CART, Random Forest (RF), Recursive Partitioning Trees (RPART), and others; Curds and Whey (CW); Curds and Whey -Lasso; dimension reduction methods, such as principal component analysis (PCA) and factor rotation or factor analysis; discriminant analysis, including Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELD A), and quadratic discriminant analysis; Discriminant Function Analysis (DFA); factor rotation or factor analysis; genetic algorithms; Hidden Markov Models; kernel based machine algorithms such as kernel density estimation, kernel partial least squares algorithms, kernel matching pursuit algorithms, kernel Fisher's discriminate analysis algorithms, and kernel principal components analysis algorithms; linear regression and generalized linear models, including or utilizing Forward Linear Stepwise Regression, Lasso (or LASSO) shrinkage and selection method, and Elastic Net regularization
  • KNN Kth-nearest neighbor
  • NNN Kth-nearest neighbor
  • SC shrunken centroids
  • StepAIC Standard for the Exchange of Product model data, Application Interpreted Constructs
  • SPC super principal component
  • SVM Support Vector Machines
  • RSVM Recursive Support Vector Machines
  • clustering algorithms as are known in the art can be useful in determining subject sub-groups.
  • Logistic Regression is the traditional predictive modeling method of choice for dichotomous response variables; e.g., treatment 1 versus treatment 2. It can be used to model both linear and non-linear aspects of the data variables and provides easily interpretable odds ratios.
  • DFA Discriminant Function Analysis
  • a forward step-wise DFA can be used to select a set of analytes that maximally discriminate among the groups studied. Specifically, at each step all variables can be reviewed to determine which will maximally discriminate among groups. This information is then included in a discriminative function, denoted a root, which is an equation consisting of linear combinations of analyte concentrations for the prediction of group membership. The discriminatory potential of the final equation can be observed as a line plot of the root values obtained for each group.
  • This approach identifies groups of analytes whose changes in concentration levels can be used to delineate profiles, diagnose and assess therapeutic efficacy.
  • the DFA model can also create an arbitrary score by which new subjects can be classified as either "healthy” or “diseased.” To facilitate the use of this score for the medical community the score can be rescaled so a value of 0 indicates a healthy individual and scores greater than 0 indicate increasing disease activity.
  • Classification and regression trees CART perform logical splits (if/then) of data to create a decision tree. All observations that fall in a given node are classified according to the most common outcome in that node. CART results are easily interpretable - one follows a series of if/then tree branches until a classification results.
  • Support vector machines classify objects into two or more classes.
  • classes include sets of treatment alternatives, sets of diagnostic alternatives, or sets of prognostic alternatives.
  • Each object is assigned to a class based on its similarity to (or distance from) objects in the training data set in which the correct class assignment of each object is known.
  • the measure of similarity of a new object to the known objects is determined using support vectors, which define a region in a potentially high dimensional space (>R6).
  • a logistical regression model if a logistical regression is bagged 1000 times, there will be 1000 logistical models, and each will provide the probability of a sample belonging to class 1 or 2.
  • Curds and Whey (CW) using ordinary least squares (OLS) is another predictive modeling method. See L. Breiman and JH Friedman, J. Royal. Stat. Soc. B 1997, 59(l):3-54. This method takes advantage of the correlations between response variables to improve predictive accuracy, compared with the usual procedure of performing an individual regression of each response variable on the common set of predictor variables X.
  • Another method is Curds and Whey and Lasso in combination (CW-Lasso). Instead of using OLS to obtain B, as in CW, here Lasso is used, and parameters are adjusted accordingly for the Lasso approach.
  • biomarker selection techniques such as, for example, forward selection, backwards selection, or stepwise selection
  • biomarker selection methodologies in their own techniques.
  • These techniques can be coupled with information criteria, such as Akaike's Information Criterion (AIC), Bayes Information Criterion (BIC), or cross-validation, to quantify the tradeoff between the inclusion of additional biomarkers and model improvement, and to minimize overfit.
  • AIC Akaike's Information Criterion
  • BIC Bayes Information Criterion
  • cross-validation to quantify the tradeoff between the inclusion of additional biomarkers and model improvement, and to minimize overfit.
  • the resulting predictive models can be validated in other studies, or cross-validated in the study they were originally trained in, using such techniques as, for example, Leave-One-Out (LOO) and 10- Fold cross-validation (10-Fold CV).
  • MBDA b 0 + b 1 * DAIMRK X - b 2 * DAIMRK 2 x - b 3 * DAIMRK 3 x * DAIMRK leisure x ; where MBDA is the MBDA score, b 0 - n are constants, and DAIMRKi. n x are the serum
  • MBDA scores thus obtained for RA subjects with known clinical assessments ⁇ e.g., DAS28 scores) can then be compared to those known assessments to determine the level of correlation between the two assessments, and hence determine the accuracy of the MBDA score and its underlying predictive model. See Examples below for specific formulas and constants.
  • MBDA ⁇ F(DAIMRK X , DAIMRK 2 X , DAIMRK n x ) where MBDA is the MBDA score, F is the function, and DAIMRK].
  • n x are the serum concentrations to the X th power of n different biomarkers selected from the biomarkers disclosed herein. The function is described in the following paragraph.
  • An interpretation function for providing a MBDA score can also be derived based on models built to predict components of a disease activity assessment, such as DAS28-CRP, rather than predicting disease activity entirely. See Example 1. An example of such a function is given by the following, wherein biomarkers are used to provide improved predicted components of the DAS score:
  • MBDA score ((0.56 * sqrt(IPTJC)) + (0.28 * sqrt(IPSJC)) + (O. J 4 * PPGA) + (0.36 * ln(CRP/10 6 + l))+0.96) * 10.53 + 1;
  • MBDA score ((0.56 * sqrt(IPTJC)) + (0.28 * sqrt(IPSJC)) + (0.14 * (PPGA)) + (0.36 * ln(CRP + 1)) + 0.96) * 10.53 + 1.
  • the MBDA score can be further rounded and capped, in order to provide a whole number between 1 and 100, the scaled MBDA score.
  • scaled MBDA score round(max(min((0.56 * sqrt(IPTJC) + (0.28 * sqrt(IPSJC)) +(0 .14 * (PPGA)) + (0.36 * ln(CRP + 1) + 0.96) * 10.53 + 1, 100), 1)).
  • Biomarker gene names provided in the above formulas represent the concentrations of those markers, and will depend on the types of assays used.
  • the MBDA score be compared to any pre-determined "reference,” "normal,” “control,” “standard,”
  • the amount of the biomarker(s) can be measured in a sample and used to derive a MBDA score, which MBDA score is then compared to a "normal" or “control” level or value, utilizing techniques such as, e.g., reference or discrimination limits or risk defining thresholds, in order to define cut-off points and/or abnormal values for inflammatory disease.
  • the normal level then is the level of one or more biomarkers or combined biomarker indices typically found in a subject who is not suffering from the inflammatory disease under evaluation.
  • normal or control are, e.g., "reference,” “index,” “baseline,” “standard,” “healthy,” “pre-disease,” etc. Such normal levels can vary, based on whether a biomarker is used alone or in a formula combined with other biomarkers to output a score. Alternatively, the normal level can be a database of biomarker patterns from previously tested subjects who did not convert to the inflammatory disease under evaluation over a clinically relevant time period. Reference (normal, control) values can also be derived from, e.g., a control subject or population whose inflammatory disease activity level or state is known.
  • the reference value can be derived from one or more subjects who have been exposed to treatment for inflammatory disease, or from one or more subjects who are at low risk of developing inflammatory disease, or from subjects who have shown improvements in inflammatory disease activity factors (such as, e.g., clinical parameters as defined herein) as a result of exposure to treatment.
  • the reference value can be derived from one or more subjects who have not been exposed to treatment; for example, samples can be collected from (a) subjects who have received initial treatment for inflammatory disease, and (b) subjects who have received subsequent treatment for inflammatory disease, to monitor the progress of the treatment.
  • a reference value can also be derived from disease activity algorithms or computed indices from population studies.
  • the quantity of one or more biomarkers of the present teachings can be indicated as a value.
  • the value can be one or more numerical values resulting from the evaluation of a sample, and can be derived, e.g., by measuring level(s) of the biomarker(s) in a sample by an assay performed in a laboratory, or from dataset obtained from a provider such as a laboratory, or from a dataset stored on a server.
  • Biomarker levels can be measured using any of several techniques known in the art.
  • the present teachings encompass such techniques, and further include all subject fasting and/or temporal-based sampling procedures for measuring biomarkers.
  • the actual measurement of levels of the biomarkers can be determined at the protein or nucleic acid level using any method known in the art.
  • Protein detection comprises detection of full-length proteins, mature proteins, pre-proteins, polypeptides, isoforms, mutations, variants, post-translationally modified proteins and variants thereof, and can be detected in any suitable manner.
  • Levels of biomarkers can be determined at the protein level, e.g., by measuring the serum levels of peptides encoded by the gene products described herein, or by measuring the enzymatic activities of these protein biomarkers.
  • Such methods are well- known in the art and include, e.g., immunoassays based on antibodies to proteins encoded by the genes, aptamers or molecular imprints. Any biological material can be used for the
  • a suitable method can be selected to determine the activity of proteins encoded by the biomarker genes according to the activity of each protein analyzed.
  • the activities can be determined in vitro using enzyme assays known in the art.
  • assays include, without limitation, protease assays, kinase assays, phosphatase assays, reductase assays, among many others.
  • Modulation of the kinetics of enzyme activities can be determined by measuring the rate constant KM using known algorithms, such as the Hill plot, Michaelis-Menten equation, linear regression plots such as Lineweaver-Burk analysis, and Scatchard plot.
  • sequence information provided by the public database entries for the biomarker
  • expression of the biomarker can be detected and measured using techniques well- known to those of skill in the art.
  • nucleic acid sequences in the sequence databases that correspond to nucleic acids of biomarkers can be used to construct primers and probes for detecting and/or measuring biomarker nucleic acids.
  • probes can be used in, e.g., Northern or Southern blot hybridization analyses, ribonuclease protection assays, and/or methods that quantitatively amplify specific nucleic acid sequences.
  • sequences from sequence databases can be used to construct primers for specifically amplifying biomarker sequences in, e.g., amplification-based detection and quantitation methods such as reverse- transcription based polymerase chain reaction (RT-PCR) and PCR.
  • RT-PCR reverse- transcription based polymerase chain reaction
  • sequence comparisons in test and reference populations can be made by comparing relative amounts of the examined DNA sequences in the test and reference populations.
  • expression can be measured using RT-PCR; e.g., polynucleotide primers specific for the differentially expressed biomarker mRNA sequences reverse-transcribe the mRNA into DNA, which is then amplified in PCR and can be visualized and quantified.
  • Biomarker RNA can also be quantified using, for example, other target amplification methods, such as TMA, SDA, and NASBA, or signal amplification methods (e.g., bDNA), and the like.
  • Ribonuclease protection assays can also be used, using probes that specifically recognize one or more biomarker mRNA sequences, to determine gene expression.
  • biomarker protein and nucleic acid metabolites can be measured.
  • the term "metabolite” includes any chemical or biochemical product of a metabolic process, such as any compound produced by the processing, cleavage or consumption of a biological molecule (e.g., a protein, nucleic acid, carbohydrate, or lipid).
  • Metabolites can be detected in a variety of ways known to one of skill in the art, including the refractive index spectroscopy (RI), ultra-violet spectroscopy (UV), fluorescence analysis, radiochemical analysis, near-infrared spectroscopy (near-IR), nuclear magnetic resonance spectroscopy (NMR), light scattering analysis (LS), mass spectrometry, pyrolysis mass spectrometry, nephelometry, dispersive Raman spectroscopy, gas chromatography combined with mass spectrometry, liquid chromatography combined with mass spectrometry, matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) combined with mass spectrometry, ion spray spectroscopy combined with mass spectrometry, capillary electrophoresis, MR and IR detection.
  • RI refractive index spectroscopy
  • UV ultra-violet spectroscopy
  • fluorescence analysis radiochemical analysis
  • radiochemical analysis near-inf
  • biomarker analytes can be measured using the above-mentioned detection methods, or other methods known to the skilled artisan.
  • circulating calcium ions Ca 2+
  • fluorescent dyes such as the Fluo series, Fura-2A, Rhod-2, among others.
  • Other biomarker metabolites can be similarly detected using reagents that are specifically designed or tailored to detect such metabolites.
  • a biomarker is detected by contacting a subject sample with reagents, generating complexes of reagent and analyte, and detecting the complexes.
  • kits include but are not limited to nucleic acid primers and antibodies.
  • an antibody binding assay is used to detect a biomarker; e.g., a sample from the subject is contacted with an antibody reagent that binds the biomarker analyte, a reaction product (or complex) comprising the antibody reagent and analyte is generated, and the presence (or absence) or amount of the complex is determined.
  • the antibody reagent useful in detecting biomarker analytes can be monoclonal, polyclonal, chimeric, recombinant, or a fragment of the foregoing, as discussed in detail above, and the step of detecting the reaction product can be carried out with any suitable immunoassay.
  • the sample from the subject is typically a biological fluid as described above, and can be the same sample of biological fluid as is used to conduct the method described above.
  • Immunoassays carried out in accordance with the present teachings can be homogeneous assays or heterogeneous assays. Immunoassays carried out in accordance with the present teachings can be multiplexed. In a homogeneous assay the immunological reaction can involve the specific antibody ⁇ e.g., anti -biomarker protein antibody), a labeled analyte, and the sample of interest. The label produces a signal, and the signal arising from the label becomes modified, directly or indirectly, upon binding of the labeled analyte to the antibody. Both the immunological reaction of binding, and detection of the extent of binding, can be carried out in a homogeneous solution. Immunochemical labels which can be employed include but are not limited to free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, and coenzymes. Immunoassays include competition assays.
  • the reagents can be the sample of interest, an antibody, and a reagent for producing a detectable signal.
  • Samples as described above can be used.
  • the antibody can be immobilized on a support, such as a bead (such as protein A and protein G agarose beads), plate or slide, and contacted with the sample suspected of containing the biomarker in liquid phase.
  • the support is separated from the liquid phase, and either the support phase or the liquid phase is examined using methods known in the art for detecting signal.
  • the signal is related to the presence of the analyte in the sample.
  • Methods for producing a detectable signal include but are not limited to the use of radioactive labels, fluorescent labels, or enzyme labels.
  • an antibody which binds to that site can be conjugated to a detectable (signal-generating) group and added to the liquid phase reaction solution before the separation step.
  • detectable group on the solid support indicates the presence of the biomarker in the test sample.
  • suitable immunoassays include but are not limited to oligonucleotides,
  • immunoblotting immunoprecipitation, immunofluorescence methods, chemiluminescence methods, electrochemiluminescence (ECL), and/or enzyme-linked immunoassays (ELISA).
  • Antibodies can be conjugated to a solid support suitable for a diagnostic assay (e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding.
  • a diagnostic assay e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene
  • Antibodies as described herein can likewise be conjugated to detectable labels or groups such as radiolabels (e.g., 35S, 1251, 1311), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein, Alexa, green fluorescent protein, rhodamine) in accordance with known techniques.
  • radiolabels e.g., 35S, 1251, 1311
  • enzyme labels e.g., horseradish peroxidase, alkaline phosphatase
  • fluorescent labels e.g., fluorescein, Alexa, green fluorescent protein, rhodamine
  • Antibodies may also be useful for detecting post-translational modifications of biomarkers.
  • post-translational modifications include, but are not limited to tyrosine phosphorylation, threonine phosphorylation, serine phosphorylation, citrullination and glycosylation (e.g., O-GlcNAc).
  • Such antibodies specifically detect the phosphorylated amino acids in a protein or proteins of interest, and can be used in the immunoblotting,
  • the present invention provides methods of recommending therapeutic regimens, including withdrawal from therapeutic regiments, following the determination of differences in expression of the biomarkers disclosed herein. Measuring scores derived from expression levels of the biomarkers disclosed herein over a period time can provide a clinician with a dynamic picture of a subject's biological state. These embodiments of the present teachings thus will provide subject-specific biological information, which will be informative for therapy decision and will facilitate therapy response monitoring, and should result in more rapid and more optimized treatment, better control of disease activity, and an increase in the proportion of subjects achieving remission.
  • autoimmune disorders such as RA
  • RA autoimmune disorders
  • a biomarker-derived algorithm can be used to quantify therapy response in RA subjects.
  • methotrexate is sometimes recommended as a first-line treatment and in non-responders both the addition of conventional non-biological disease modifying anti -rheumatic drug therapy (triple DMARD therapy) and of biological (anti- T F) therapy are supported by data. Identification of patients with a higher likelihood of responding to one or the other of these options would lead to more personalized medicine and increased effectiveness of therapy, which is a primary objective of this invention.
  • the levels of one or more analyte biomarkers or the levels of a specific panel of analyte biomarkers in a sample are compared to a reference standard
  • reference standard in order to direct treatment decisions. Expression levels of the one or more biomarkers can be combined into a score, which can represent disease activity.
  • the reference standard used for any embodiment disclosed herein may comprise average, mean, or median levels of the one or more analyte biomarkers or the levels of the specific panel of analyte biomarkers in a control population.
  • the reference standard may further include an earlier time point for the same subject. For example, a reference standard may include a first time point, and the levels of the one or more analyte biomarkers can be examined again at second, third, fourth, fifth, sixth time points, etc. Any time point earlier than any particular time point can be considered a reference standard.
  • the reference standard may additionally comprise cutoff values or any other statistical attribute of the control population, or earlier time points of the same subject, such as a standard deviation from the mean levels of the one or more analyte biomarkers or the levels of the specific panel of analyte biomarkers.
  • the control population may comprise healthy individuals or the same subject prior to the administration of any therapy.
  • a score may be obtained from the reference time point, and a different score may be obtained from a later time point.
  • a first time point can be when an initial therapeutic regimen is begun.
  • a first time point can also be when a first immunoassay is performed.
  • a time point can be hours, days, months, years, etc. In some embodiments, a time point is one month.
  • a time point is two months. In some embodiments, a time point is three months. In some embodiments, a time point is four months. In some embodiments, a time point is five months. In some embodiments, a time point is six months. In some embodiments, a time point is seven months. In some embodiments, a time point is eight months. In some embodiments, a time point is nine months. In some embodiments, a time point is ten months. In some embodiments, a time point is eleven months. In some embodiments, a time point is twelve months. In some embodiments, a time point is two years. In some embodiments, a time point is three years. In some embodiments, a time point is four years. In some embodiments, a time point is five years. In some embodiments, a time point is ten years.
  • a difference in the score can be interpreted as a decrease in disease activity.
  • lower score can indicate a lower level of disease activity, or remission.
  • a second score having a lower score than the reference score, or first score means that the subject's disease activity has been lowered (improved) between the first and second time periods, or is in remission.
  • a higher score can indicate a lower level of disease activity, or remission.
  • a second score having a higher score than the reference score, or first score also means that the subject's disease activity has improved between the first and second time periods, or is in remission.
  • a difference in the score can also be interpreted as an increase in disease activity. For example, lower score can indicate a higher level of disease activity, or flare. In these
  • a second score having a lower score than the reference score, or first score means that the subject's disease activity has been increased (worsened) between the first and second time periods.
  • a higher score can indicate a higher level of disease activity, or flare.
  • a second score having a higher score than the reference score, or first score also means that the subject's disease activity has worsened between the first and second time periods, or is flaring.
  • the optimal cutoff for flare, sustained clinical remission, and functional remission 6 months after discontinuation is an MBDA score of 30, 23, and 26, respectively.
  • the optimal cutoff for flare, sustained clinical remission, and functional remission 12 months after discontinuation is an MBDA score of 26, 25, and 27, respectively.
  • the differences can be variable. For example, when a difference in the score is interpreted as a decrease in disease activity, a large difference can mean a greater decrease in disease activity than a lower or moderate difference. Alternatively, when a difference in the score is interpreted as an increase in disease activity, a large difference can mean a greater increase in disease activity than a lower or moderate difference.
  • a patient is treated more or less aggressively than a reference therapy based on the difference of scores.
  • a reference therapy is any therapy that is the standard of care for the autoimmune disorder.
  • the standard of care can vary temporally and
  • a more aggressive therapy than the standard therapy comprises beginning treatment earlier than in the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises administering additional treatments than in the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises treating on an accelerated schedule compared to the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises administering additional treatments not called for in the standard therapy.
  • a less aggressive therapy than the standard therapy comprises delaying treatment relative to the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering less treatment than in the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering treatment on a decelerated schedule compared to the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering no treatment. Therapy withdrawal
  • prediction of autoimmune disease patients in particular RA patients, who can successfully withdrawal from or discontinue therapy, can be based on an MBDA score.
  • the therapy to be considered for withdrawal can be any therapy as described herein.
  • a high MBDA score as described herein at baseline can be an independent predictor of disease progression within a certain period of time following discontinuation of therapy.
  • a moderate MBDA score as described herein at baseline can be an independent predictor of disease progression within a certain period of time following discontinuation of therapy.
  • a low MBDA score as described herein at baseline can be an independent predictor of disease progression, or remission, within a certain period of time following discontinuation of therapy.
  • the disease progression for therapy withdrawal is flare. In some embodiments, the disease progression for therapy withdrawal is radiographic progression. In other embodiments, the disease progression for therapy withdrawal is any other consequence of autoimmune diseases, in particular RA, known in the art.
  • the period of time in which an MBDA score can predict disease progression can be one, two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve months.
  • the period of time in which an MBDA score can predict disease progression can also be one, two, three, four, five, ten, fifteen, twenty, twenty -five, or more years.
  • a low MBDA score as described herein can result in the recommendation that therapy is withdrawn or discontinued.
  • a low or moderate MBDA score as described herein can result in the recommendation that therapy is withdrawn or discontinued.
  • a moderate or high MBDA score as described herein can result in the recommendation that therapy is not withdrawn or discontinued.
  • a high MBDA score as described herein can result in the recommendation that therapy is not withdrawn or discontinued.
  • the therapy can be partially withdrawn.
  • the therapy can be withdrawn partially in relation to the MBDA score.
  • a low MBDA score as described herein can result in the
  • dose can be adjusted based on the MBDA score.
  • the practitioner discontinues a therapy regimen if a score is low. In one embodiment, the practitioner does not change the therapy regimen if the score is high. In one embodiment, the practitioner adjusts the therapy based on a comparison between difference scores, or based on an initial predictive score. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different combination of drugs. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage. In one embodiment, the practitioner adjusts the therapy by adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting drug dosage.
  • the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage and dose schedule. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting dose schedule and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, and adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, and adjusting length of therapy.
  • the practitioner adjusts the therapy by selecting and administering a different drug, adjusting dose schedule, and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage, adjusting dose schedule, and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, adjusting dose schedule, and adjusting length of therapy.
  • a less aggressive therapy comprises no change in the therapy regimen. In one embodiment a less aggressive therapy comprises delaying treatment. In one embodiment a less aggressive therapy comprises selecting and administering less potent drugs. In one embodiment a less aggressive therapy comprises decreasing the frequency treatment. In one embodiment a less aggressive therapy comprises shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and decreasing drug dosage. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and decelerating dose schedule. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and shortening length of therapy. In one embodiment, less aggressive therapy comprises decreasing drug dosage and decelerating dose schedule. In one embodiment, less aggressive therapy comprises decreasing drug dosage and shortening length of therapy.
  • less aggressive therapy comprises decelerating dose schedule and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, and decelerating dose schedule. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decelerating dose schedule, and shortening length of therapy. In one embodiment, less aggressive therapy comprises decreasing drug dosage, decelerating dose schedule, and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, decelerating dose schedule, and shortening length of therapy. In some embodiments, a less aggressive therapy comprises administering only non-drug-based therapies.
  • treatment comprises a more aggressive therapy than a reference therapy.
  • a more aggressive therapy comprises increased length of therapy.
  • a more aggressive therapy comprises increased frequency of the dose schedule.
  • more aggressive therapy comprises selecting and administering more potent drugs and increasing drug dosage.
  • more aggressive therapy comprises selecting and administering more potent drugs and accelerating dose schedule.
  • more aggressive therapy comprises selecting and administering more potent drugs and increasing length of therapy.
  • more aggressive therapy comprises increasing drug dosage and accelerating dose schedule.
  • more aggressive therapy comprises increasing drug dosage and increasing length of therapy.
  • more aggressive therapy comprises accelerating dose schedule and increasing length of therapy.
  • more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, and accelerating dose schedule. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, and increasing length of therapy. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, accelerating dose schedule, and increasing length of therapy. In one embodiment, more aggressive therapy comprises increasing drug dosage, accelerating dose schedule, and increasing length of therapy. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, accelerating dose schedule, and increasing length of therapy. In some embodiments, a more aggressive therapy comprises administering a combination of drug- based and non-drug-based therapies.
  • Therapies can be conventional or biologic.
  • therapies such as disease modifying anti -rheumatic drugs (DMARD) that are generally considered conventional include, but are not limited to, MTX, azathioprine (AZA), bucillamine (BUC), chloroquine (CQ), ciclosporin (CSA, or cyclosporine, or cyclosporin), doxycycline (DOXY), hydroxychloroquine (HCQ), intramuscular gold (EVI gold), leflunomide (LEF), levofloxacin (LEV), and sulfasalazine (SSZ).
  • MTX azathioprine
  • BUC bucillamine
  • CQ chloroquine
  • CSA ciclosporin
  • DOXY hydroxychloroquine
  • HCQ hydroxychloroquine
  • EVI gold intramuscular gold
  • LEF leflunomide
  • LEV levofloxacin
  • SSZ sulfasalazine
  • biologic drugs can include but are not limited to biological agents that target the tumor necrosis factor (T F)-alpha molecules and the TNF inhibitors, such as infliximab, adalimumab, etanercept and golimumab.
  • T F tumor necrosis factor
  • Other classes of biologic drugs include ILl inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab.
  • a test sample from the subject can also be exposed to a therapeutic agent or a drug, and the level of one or more biomarkers can be determined.
  • the level of one or more biomarkers can be compared to sample derived from the subject before and after treatment or exposure to a therapeutic agent or a drug, or can be compared to samples derived from one or more subjects who have shown improvements in inflammatory disease state or activity (e.g., clinical parameters or traditional laboratory risk factors) as a result of such treatment or exposure.
  • MBDA scores are tailored to the population, endpoints or clinical assessment, and/or use that is intended.
  • a MBDA score can be used to assess subjects for primary prevention and diagnosis, and for secondary prevention and management.
  • the MBDA score can be used for prediction and risk stratification for future conditions or disease sequelae, for the diagnosis of inflammatory disease, for the prognosis of disease activity and rate of change, and for indications for future diagnosis and therapeutic regimens.
  • the MBDA score can be used for prognosis and risk stratification.
  • the MBDA score can be used for clinical decision support, such as determining whether to defer intervention or treatment, to recommend preventive check-ups for at-risk patients, to recommend increased visit frequency, to recommend increased testing, and to recommend intervention.
  • the MBDA score can also be useful for therapeutic selection, determining response to treatment, adjustment and dosing of treatment, monitoring ongoing therapeutic efficiency, and indication for change in therapeutic regimen.
  • the MBDA score can be used to aid in the diagnosis of inflammatory disease, and in the determination of the severity of inflammatory disease.
  • the MBDA score can also be used for determining the future status of intervention such as, for example in RA, determining the prognosis of future joint erosion with or without treatment.
  • Certain embodiments of the present teachings can be tailored to a specific treatment or a combination of treatments. X-ray is currently considered the gold standard for assessment of disease progression, but it has limited capabilities since subjects may have long periods of active symptomatic disease while radiographs remain normal or show only nonspecific changes. Conversely, subjects who seem to have quiescent disease (subclinical disease) may slowly progress over time, undetected clinically until significant radiographic progression has occurred.
  • Tests for measuring disease activity can be implemented on a variety of systems typically used for obtaining test results, such as results from immunological or nucleic acid detection assays.
  • Such systems may comprise modules that automate sample preparation, that automate testing such as measuring biomarker levels, that facilitate testing of multiple samples, and/or are programmed to assay the same test or different tests on each sample.
  • the testing system comprises one or more of a sample preparation module, a clinical chemistry module, and an immunoassay module on one platform.
  • Testing systems are typically designed such that they also comprise modules to collect, store, and track results, such as by connecting to and utilizing a database residing on hardware.
  • Test systems also generally comprise a module for reporting and/or visualizing results.
  • reporting modules include a visible display or graphical user interface, links to a database, a printer, etc. See section Machine-readable storage medium, below.
  • One embodiment of the present invention comprises a system for determining the inflammatory disease activity of a subject.
  • the system employs a module for applying a formula to an input comprising the measured levels of biomarkers in a panel, as described herein, and outputting a score.
  • the measured biomarker levels are test results, which serve as inputs to a computer that is programmed to apply the formula.
  • the system may comprise other inputs in addition to or in combination with biomarker results in order to derive an output score; e.g., one or more clinical parameters such as therapeutic regimen, TJC, SJC, morning stiffness, arthritis of three or more joint areas, arthritis of hand joints, symmetric arthritis, rheumatoid nodules, radiographic changes and other imaging, gender/sex, age, race/ethnicity, disease duration, height, weight, body-mass index, family history, CCP status, RF status, ESR, smoker/non-smoker, etc.
  • the system can apply a formula to biomarker level inputs, and then output a disease activity score that can then be analyzed in conjunction with other inputs such as other clinical parameters.
  • the system is designed to apply a formula to the biomarker and non-biomarker inputs (such as clinical parameters) together, and then report a composite output disease activity index.
  • ARCHITECT is a registered trademark of Abbott Laboratories, Abbott Park, 111. 60064.
  • C. Wilson et al. "Clinical Chemistry Analyzer Sub-System Level Performance,” American Association for Clinical Chemistry Annual Meeting, Chicago, 111., Jul. 23-27, 2006; and, HJ Kisner, "Product development: the making of the Abbott ARCHITECT,” Clin. Lab. Manage. Rev. 1997 Nov.-Dec, 11(6):419-21;
  • A. Ognibene et al. "A new modular
  • VITROS is a registered trademark of Johnson & Johnson Corp., New
  • DIMENSION system (DIMENSION is a registered trademark of Dade Behring Inc., Deerfield 111.) - a system for the analysis of body fluids, comprising computer software and hardware for operating the analyzers, and analyzing the data generated by the analyzers. [00139] The testing required for various embodiments of the present teachings, e.g.
  • measuring biomarker levels can be performed by laboratories such as those certified under the Clinical Laboratory Improvement Amendments (42 U.S.C. Section 263(a)), or by laboratories certified under any other federal or state law, or the law of any other country, state or province that governs the operation of laboratories that analyze samples for clinical purposes.
  • laboratories include, for example, Laboratory Corporation of America, 358 South Main Street, Burlington, NC 27215 (corporate headquarters); Quest Diagnostics, 3 Giralda Farms, Madison, NJ 07940 (corporate headquarters); and other reference and clinical chemistry laboratories.
  • kits comprise oligonucleotides that specifically identify one or more biomarker nucleic acids based on homology and/or complementarity with biomarker nucleic acids.
  • the oligonucleotide sequences may correspond to fragments of the biomarker nucleic acids.
  • the oligonucleotides can be more than 200, 200, 150, 100, 50, 25, 10, or fewer than 10 nucleotides in length.
  • the kits comprise antibodies to proteins encoded by the biomarker nucleic acids.
  • kits of the present teachings can also comprise aptamers.
  • the kit can contain in separate containers a nucleic acid or antibody (the antibody either bound to a solid matrix, or packaged separately with reagents for binding to a matrix), control formulations (positive and/or negative), and/or a detectable label, such as but not limited to fluorescein, green fluorescent protein, rhodamine, cyanine dyes, Alexa dyes, luciferase, and radiolabels, among others.
  • Instructions for carrying out the assay including, optionally, instructions for generating a MBDA score, can be included in the kit; e.g., written, tape, VCR, or CD-ROM.
  • the assay can for example be in the form of a Northern hybridization or a sandwich ELISA as known in the art.
  • biomarker detection reagents can be immobilized on a solid matrix, such as a porous strip, to form at least one biomarker detection site.
  • the measurement or detection region of the porous strip can include a plurality of sites containing a nucleic acid.
  • the test strip can also contain sites for negative and/or positive controls. Alternatively, control sites can be located on a separate strip from the test strip.
  • the different detection sites can contain different amounts of immobilized nucleic acids, e.g., a higher amount in the first detection site and lesser amounts in subsequent sites.
  • the number of sites displaying a detectable signal provides a quantitative indication of the amount of biomarker present in the sample.
  • the detection sites can be configured in any suitably detectable shape and can be, e.g., in the shape of a bar or dot spanning the width of a test strip.
  • the kit can contain a nucleic acid substrate array comprising one or more nucleic acid sequences.
  • the nucleic acids on the array specifically identify one or more nucleic acid sequences represented by the MBDA markers.
  • the expression of one or more of the sequences represented by the MBDA markers can be identified by virtue of binding to the array.
  • the substrate array can be on a solid substrate, such as what is known as a "chip.” See, e.g., U.S. Pat. No. 5,744,305.
  • the substrate array can be a solution array; e.g., xMAP (Luminex, Austin, TX), Cyvera (Illumina, San Diego, CA), RayBio Antibody Arrays (RayBiotech, Inc., Norcross, GA), CellCard (Vitra Bioscience, Mountain View, CA) and Quantum Dots' Mosaic (Invitrogen, Carlsbad, CA).
  • xMAP Luminex, Austin, TX
  • Cyvera Illumina, San Diego, CA
  • RayBio Antibody Arrays RayBiotech, Inc., Norcross, GA
  • CellCard Vitra Bioscience, Mountain View, CA
  • Quantum Dots' Mosaic Invitrogen, Carlsbad, CA.
  • a machine-readable storage medium can comprise, for example, a data storage material that is encoded with machine-readable data or data arrays.
  • the data and machine- readable storage medium are capable of being used for a variety of purposes, when using a machine programmed with instructions for using said data. Such purposes include, without limitation, storing, accessing and manipulating information relating to the inflammatory disease activity of a subject or population over time, or disease activity in response to inflammatory disease treatment, or for drug discovery for inflammatory disease, etc.
  • Data comprising measurements of the biomarkers of the present teachings, and/or the evaluation of disease activity or disease state from these biomarkers, can be implemented in computer programs that are executing on programmable computers, which comprise a processor, a data storage system, one or more input devices, one or more output devices, etc.
  • Program code can be applied to the input data to perform the functions described herein, and to generate output information. This output information can then be applied to one or more output devices, according to methods well-known in the art.
  • the computer can be, for example, a personal computer, a
  • microcomputer or a workstation of conventional design.
  • the computer programs can be implemented in a high-level procedural or object- oriented programming language, to communicate with a computer system such as for example, the computer system illustrated in FIG. 15.
  • the programs can also be implemented in machine or assembly language.
  • the programming language can also be a compiled or interpreted language.
  • Each computer program can be stored on storage media or a device such as ROM, magnetic diskette, etc., and can be readable by a programmable computer for configuring and operating the computer when the storage media or device is read by the computer to perform the described procedures.
  • Any health-related data management systems of the present teachings can be considered to be implemented as a computer-readable storage medium, configured with a computer program, where the storage medium causes a computer to operate in a specific manner to perform various functions, as described herein.
  • the biomarkers disclosed herein can be used to generate a "subject biomarker profile" taken from subjects who have inflammatory disease.
  • the subject biomarker profiles can then be compared to a reference biomarker profile, in order to diagnose or identify subjects with inflammatory disease, to monitor the progression or rate of progression of inflammatory disease, or to monitor the effectiveness of treatment for inflammatory disease.
  • the biomarker profiles, reference and subject, of embodiments of the present teachings can be contained in a machine- readable medium, such as analog tapes like those readable by a CD-ROM or USB flash media, among others.
  • Such machine-readable media can also contain additional test results, such as measurements of clinical parameters and clinical assessments.
  • the machine-readable media can also comprise subject information; e.g., the subject's medical or family history.
  • the machine- readable media can also contain information relating to other disease activity algorithms and computed scores or indices, such as those described herein.
  • Example 1 Deriving an MBDA score
  • This example demonstrates a method of deriving a Multi-Biomarker Disease Activity (MBDA) score, based on a dataset of quantitative data for biomarkers.
  • MBDA Multi-Biomarker Disease Activity
  • a MBDA score is determined from the biomarker data using an interpretation function that is based on a set of predictive models, where each predictive model is predictive of a component of the DAS28-CRP, in this example TJC, SJC and patient global health assessment (GHA).
  • GHA patient global health assessment
  • a MBDA algorithm was trained using clinical and biomarker data for patients in the InFoRM and BRASS studies.
  • the InFoRM study (Index For Rheumatoid Arthritis
  • Measurement is a multi-center observational study of the North American RA population.
  • the patients used in algorithm training were recruited between April and September 2009 from 25 sites in the U.S. and Canada.
  • Inclusion criteria were: age >18 years with a diagnosis of RA made by a board-certified rheumatologist.
  • Patients concurrently enrolled in therapeutic drug trials involving a biologic agent and a placebo arm were excluded.
  • the study includes three visits for each patient, each with clinical data and biological sample collection, at approximately three- month intervals.
  • BRASS is an observational study of approximately 1,000 RA patients receiving care at the RB Brigham Arthritis and Musculoskeletal Diseases Clinical Research Center, at the Brigham and Women's Hospital, Boston, Mass. Inclusion criteria were: age >18 years with a diagnosis of RA made by a board-certified rheumatologist. The study includes annual visits with clinical data and biological sample collection, and patient questionnaires between visits.
  • the first data set used in training consisted of visit 1 data for 512 InFoRM patients.
  • the 512 patient visits were chosen to have clinical characteristics representative of the entire study population at the time of selection, and also to have been evaluated by a limited number of joint assessors.
  • the number of joint assessors was limited to 12 so that assessor- specific biases could be evaluated and taken into account in algorithm development.
  • the average age of these patients was 58.9 years (range 20-91), and 76% were female.
  • the mean SJC and TJC were 4.28 and 5.49, respectively.
  • biomarkers for 25 candidate biomarkers were run on serum from the 512 InFoRM visits. Those biomarkers were SAA1, IL6, TNFRSF1A, VEGFA, PYD, MMP1, KAMI, calprotectin, CHI3L1 (YKL40), MMP3, EGF, IL1RA, VCAM1, LEP, RETN, CRP, IL8, APOA1, APOC3, CCL22, IL1B, IL6R, IL18, keratan sulfate and ICTP. All the biomarker assays were run on the Meso Scale Discovery (MSD ® ) platform. See Example 1 of US
  • biomarkers were prioritized based on (1) univariate association with disease activity, (2) contribution to multivariate models for disease activity, and (3) assay performance.
  • the assays for 20 candidate biomarkers were run in a second set of patient samples, comprising 167 samples from BRASS and 29 from InFoRM. These 20 candidate biomarkers were SAA1, IL6, T FRSF1A, VEGFA, PYD, MMP1, ICAM1, calprotectin, YKL40, MMP3, EGF, ILIRA, VCAM1, leptin, resistin, CRP, IL8, CCL22, IL1B and IL6R. The samples were selected to enrich the overall training data for extremes of disease activity, while also having good representation of patients with moderate disease activity.
  • Enriching for extreme phenotypes can result in improved algorithm training, as long as the resulting training population still fully represents the types of patients on which the algorithm will used in independent validation and intended use populations.
  • the 167 BRASS samples were intended to represent similar numbers of patients with low, moderate and high disease activity.
  • the 29 InFoRM samples were selected to represent patients with high disease activity, since low and moderate activity patients were already well represented by the first 512 training samples.
  • Lasso is used to prioritize biomarkers (based on R 2 values) and to obtain a Lasso model.
  • the "lasso" in this model minimizes the residual sum of squares, subject to the sum of the absolute value of the coefficients being less than a constant. This method produces interpretable models and exhibits the stability of ridge regression. See R. Tibshirani, J. Royal Stat. Soc. B 1996, 58(l):267-288.
  • the Elastic Net mixtures of Lasso and ridge penalties are applied. It encourages a grouping effect, where strongly correlated predictors segregate together, either tending to be in or out of the model together. See T. Zou, J. Royal Stat. Soc. B 2005, 67(2):301-320. For each of the above three methods, the marker selected at each step is recorded.
  • the fourth method (4) is Multivariate Response with Curds and Whey (CW) using ordinary least squares (OLS). See L. Breiman and JH Friedman, J. Royal. Stat. Soc. B 1997, 59(l):3-54.
  • This method takes advantage of the correlations between the response variables ⁇ e.g., components of DAS) to improve predictive accuracy, compared with the usual procedure of performing an individual regression of each response variable on the common set of predictor variables X.
  • B is obtained using OLS
  • S is the shrinkage matrix computed from the canonical co-ordinate system.
  • the fifth method (5) is Curds and Whey and Lasso in combination (CW-Lasso).
  • the 20 candidate biomarkers examined in all training samples were prioritized according to a number of criteria, including: strength of association with disease activity and contribution to multivariate models; consistency of correlation with disease activity across feasibility and training data sets; CRP was excluded from any sub-models for TJC, SJC, and PGA both because it is included in the DAS28-CRP4 and because it did not increase sub-model prediction accuracy in independent test samples (CRP is used, however, in the final MBDA score calculation as part of the MBDA formula); robust assay performance (ILIB was excluded from final modeling because its concentrations too frequently fall below the limits of detection of immunoassays); known drug effects (IL6R was excluded from final modeling because it is known to be strongly affected by tocilizumab, independently of the effects of the drug on disease activity); and, stability (TL8 was excluded from final modeling because its measurable levels are known to rise dramatically when serum samples are not kept cold). These criteria led to 15 candidate biomarkers being considered for inclusion in the final algorithm. See Table 1.
  • IPTJC Improved Prediction of TJC
  • IPSJC Improved Prediction of SJC
  • PPGHA Predicted PGHA
  • PDAS Predicted DAS28-CRP.
  • Table 4 demonstrates the correlation of the values predicted by the PDAS algorithm with actual values for TJC, SJC, PGHA and DAS28-CRP, in the two cohorts studied, CAMERA and InFoRM.
  • PSJC -25.444 + 4.051 * (SAA1) 1/10 + 16.154 * (IL6) o - 11.847 * (EGF) 1/10 +3.091 * (CHI3L1) 1/10 + 0.353 * (TNFRSF1A) 1/10 ;
  • PPGHA -13.489 + 5.474 * (IL6) 1/10 + 0.486 * (SAA1) 1/10 + 2.246 * (MMP1) 1/10 + 1.684 * (leptin) 1/10 + 4.14 * (TNFRSF1A) 1/10 + 2.292 * (VEGFA) 1/10 - 1.898 * (EGF) 1/10 + 0.028 * (MMP3) 1/10 - 2.892 * (VCAMl) 1/w - .506 * (REIN) o ;
  • IPTJC max(0.1739 * PTJC + 0.7865 * PSJC,0);
  • IPSJC max(0.1734 * PTJC + 0.7839 * PSJC, 0);
  • MBDA score round(max(min((56 * sqrt(IPTJC) + .28 * sqrt(IPSJC) + .14 * PPG A + .36 * ln(CRP/10 6 + 1)) * 10.53 + 1, 100), 1)).
  • Rheumatology (ACR) criteria for rheumatoid arthritis were asked to participate in this two-year randomized, open-label prospective multicentre strategy trial. As a result, 299 patients were studied. Patients visited the outpatient clinic of one of the six rheumatology departments in the region of Utrecht, the Netherlands, collaborating in the Utrecht Rheumatoid Arthritis Cohort study group. Inclusion criteria were that patients must have exhibited symptoms for less than one year, with age greater than 16 years. Exclusion criteria were the previous use of
  • glucocorticoids or any DMARD use of cytotoxic or immunosuppressive drugs within a period of three months before inclusion, alcohol abuse, defined as more than two units per day, and psychological problems, which would make adherence to the study protocol impossible.
  • alcohol abuse defined as more than two units per day
  • psychological problems which would make adherence to the study protocol impossible.
  • This screening included a chest X-ray, liver enzymes, albumin, hepatitis serology, serum creatinine and complete blood count.
  • An independent person performed randomization in blocks of nine per hospital. The medical ethics committees of all participating hospitals approved this study, and all patients gave written informed consent before entering the study.
  • the cohort for this study had the following characteristics: 69% female, 68% CCP positive, 74% RF positive, 100% on MTX, 100% on non-biologic DMARDs, and 0% on biologic DMARDs. Additionally, the mean age of the cohort was 52 years (standard deviation (SD) +/- 14.7), with a minimum age of 17 and a maximum age of 78. The mean DAS28-CRP for this cohort was 5.0 (SD +/- 1.9), with a minimum of 0.9 and a maximum of 8.4.
  • the first algorithm or "prototype algorithm,” using a linear combination of protein biomarkers, was trained on subject samples to estimate the DAS28 directly and was provided by the formula described elsewhere herein according to:
  • MBDA b 0 + b 1 * DAIMRK X - b 2 * DAIMRK 2 x - b 3 * DAIMRK 3 x . . . - b n * DAIMRK till x ; where MBDA is the MBDA score, b 0 . detox are constants, and DAIMRK].
  • n x are the serum
  • concentrations, transformed to the x th power, of n different biomarkers selected from the panel of biomarkers are concentrations, transformed to the x th power, of n different biomarkers selected from the panel of biomarkers.
  • MBDA (-16.1564) - (0.0606 * Calprotectin 1/w ) + (0.2194 * CHI3L1 1/W ) + (1.1886 *
  • ICAM1 1/W (2.7738 * IL6 1/W ) + (0.7254 * MMP1 1/W ) - (0.8348 * MMP3 1/W ) + (1.0296 * PYD 1/W ) + (1.1792 * SAA1 1/W ) + (2.4422 * TNFRSF1A 1/W ) + (0.3272 * VEGFA 1/W ).
  • the second algorithm was derived using serum biomarker concentrations to separately estimate the three clinical assessments of TJC28, SJC28 and PGHA. Note that all of these are components of the formula used in calculating DAS28-CRP:
  • DAS28-CRP 0.56 * sqrt(TJC28) + 0.28 * sqrt(SJC28) + 0.36 * ln(CRP+l) + (0.014 * PGHA) + 0.96.
  • Biomarkers were then selected to predict and estimate clinical assessments of disease activity, specifically PGHA, TJC28 and SJC28. The resulting estimates were combined with a serum CRP concentration measurement to calculate an overall MBDA score. See FIG. 2, which indicates the three panels of biomarkers predictive of clinical disease activity
  • the CW-Lasso method was used to predict the individual components of the DAS28; i.e., TJC28, SJC28 and PGHA.
  • biomarker terms are included in the CW-Lasso if they help to improve cross-validated model performance, and this criterion does not imply that each term is statistically significant by univariate analysis. A biomarker could make a significant contribution to a multivariate model even if it does not have a significant univariate correlation, and could not make a significant contribution to a
  • This examples includes data from the following set of biomarkers: SAA1, IL6, CHI3L1, EGF, TNFRSF1A, LEP, VEGFA and VCAMl for PTJC; SAA1, IL6, EGF, CHI3L1 and TNFRSFIA for PSJC; SAA1, MMP1, LEP, TNFRSFIA, VEGFA, EGF, MMP3, VCAMl and RETN for PPGHA; plus CRP.
  • data from the following set of 12 markers was used to derive a MBDA score: CHI3L1, CRP, EGF, IL6, LEP, MMP1, MMP3, RETN, SAA1, TNFRSFIA, VCAMl and VEGFA.
  • the predicted clinical assessments of disease activity were developed according to the following formulas:
  • PTJC -38.564 + (3.997 * SAA1 1/W ) + (17.331 * IL6 1/1U ) + (4.665 * CHI3Lr w ) - (15.236 * EGF o ) + (2.651 * TNFRSFIA 1 " 10 ) + (2.641 * LEP 1/W ) + (4.026 * VEGFA 1110 ) - (1.47 * VCAMl 1110 );
  • PSJC -25.444 + (4.051 * SAA1 1/10 ) + (16.154 * IL6 1/W ) - (11.847 * EGF 1/W ) + (3.091 * CHI3L1 1/W ) + (0.353 * TNFRSFIA 1 " 10 );
  • this Example demonstrates that serum protein biomarkers representing a variety of biological pathways were consistently associated with RA disease activity.
  • a pre-specified MBDA algorithm combining information from several of these biomarkers performed well in predicting RA disease activity when evaluated in an independent test set.
  • the algorithm's estimates of TJC, SJC and PGHA correlated to actual clinical measures of disease activity.
  • subsequent MBDA scores of the subjects analyzed decreased compared to initial MBDA scores following and in response to treatment.
  • Example 2 Predictive value of the multi-biomarker disease activity (MBDA) score for flare and sustained remission
  • the objective of this example is to determine the predictive value of the multi- biomarker disease activity (MBDA) score as described in Example 1 for flare and sustained remission after discontinuation of adalimumab (ADA) in patients with rheumatoid arthritis (RA).
  • MBDA multi- biomarker disease activity
  • ADA adalimumab
  • RA rheumatoid arthritis
  • DAS28 (ESR) adalimumab
  • Clinical disease activity, functional status, and joint damage were recorded at ADA discontinuation (baseline), and after 24 and 52 weeks. Twenty-four weeks following ADA discontinuation, remission was observed in 58% of patients. At 1 year, 48% of patients were in remission, 40% had flared (DAS28 (ESR) >3.2) and little change was observed in physical function or radiographic damage (Hirata S. et. al.
  • DAS28 was only independent baseline predictor of remission at one year, observed in 68% of patients with baseline DAS28 (ESR) ⁇ 1.98, compared with 25% with DAS28 (ESR) >1.98.
  • HONOR demonstrated that it was possible to maintain disease activity score (DAS28, ESR ⁇ 2.6) clinical remission following discontinuation of ADA in approximately half of patients and that a very low DAS28 (ESR), termed “deep remission,” was predictive of a favorable clinical outcome without functional and radiographic progression and that very low DAS28 (ESR) at the time of discontinuation was associated with successful ADA- free DS28 (ESR) ⁇ 2.6 (Hirata et al. 2013; Tanaka et al. 201).
  • the baseline time point represents the time of ADA discontinuation in the HONOR study.
  • Serum samples were available for 42 of the 51 subjects in the ADA discontinuation arm and were included in the present analysis.
  • Clinical disease activity, functional status, and joint damage were assessed at the time of ADA discontinuation (baseline for the present study), and at 6 and 12 months post-discontinuation.
  • DAS disease activity score
  • SDAI simple disease activity index
  • CDAI clinical disease activity index
  • Flare was defined as DAS28>3.2.
  • Sustained clinical remission (SC- REM) was defined as absence of flare and DAS28 ⁇ 2.6 at all visits.
  • Physical function was assessed using the Health Assessment Questionnaire (HAQ-DI).
  • Baseline MBDA score and individual biomarkers were tested by univariate logistic regression models as the sole independent predictors of sustained low disease activity, i.e., DAS28-ESR ⁇ 3.2; time patient remained free of ADA; functional good outcome (also referred as to as remission), i.e., HAQ ⁇ 0.5; structural remission over 1 year, i.e., annual rate of AmTSS ⁇ 0.5; and structural progression over 1 year, i.e., annual rate of AmTSS >0.5, >3, and >5.
  • Multivariate logistic regression models were used to evaluate baseline MBDA score and individual biomarkers as predictors of each outcome when adjusted for one or more of the baseline patient and disease-related factors. Baseline variables with p-values ⁇ 0.2 in univariate analyses were included in the multivariate model.
  • Receiver operating characteristic (ROC) curves of the ability of MBDA score to predict clinical remission, flare, and functional remission at 6 months and 1 year after discontinuation of ADA were generated and analyzed to identify the optimal cut-off value of baseline MBDA scores. P-values were based on 2-sided tests; any p-value ⁇ 0.05 was considered statistically significant and ⁇ 0.1 marginally significant.
  • MBDA scores (remission, ⁇ 25; low, 26-29; moderate, 30-44; high, >44) were determined from serum samples collected at baseline. The ability of predicting flare (DAS28- ESR >3.2) or sustained clinical remission (SC-REM) (DAS28-ESR ⁇ 2.6) by MBDA score and patient characteristics were determined. Patient characteristics at ADA discontinuation are shown in Table 7.
  • the MBDA score at discontinuation can be used as a predictor of sustained functional remission. Percentage of sustained functional remission at year 1 by MBDA category (low/moderate/high) was 67.9%/55.6%/40% ( Figure 13). Percentage of sustained functional remission at year 1 by MBDA category (remission/low/moderate/high) was
  • the MBDA score at discontinuation can be used as a predictor of sustained clinical remission (SC-REM) (DAS28 ⁇ 2.6, without flare). Rates of SC-REM were higher among individuals with lower baseline MBDA score categories at both 6 months post- discontinuation and at 1-year post discontinuation ( Figure 1 1). Rates of SC-REM at 6 months and 1 year increased with increasing baseline MBDA score utilizing three MBDA categories ( Figure 10) and four MBDA categories with a lower MBDA cut off value of ⁇ 25 (MBDA remission) ( Figure 1 1 A and 1 IB). Similarly, baseline MBDA scores tended to be lower among patients with SC-REM than those without SC-REM at both 6 months and 1 year post- discontinuation (Figure 9).
  • Multivariate logistic is a predictor of SC-REM.
  • Table 8 shows a multivariable logistic regression analysis of the contribution of baseline MBDA score and SC-REM controlling for potential confounders.
  • Multivariate model includes MBDA score and disease duration.
  • b Multivariate model includes BMDA score, disease duration, and their interaction
  • Table 9 demonstrates odds rations for flare/remission using an MBDA cut off of
  • Odds ratio and 95% CI were from Fisher's Exact Test for count data. *Odds ratio is significantly different from 1 because the 95% CI excludes 1.
  • Figure 16 displays ROC curves of clinical remission, flare, and functional remission at 6 months and 1 year after ADA discontinuation.

Abstract

Provided herein are methods for assessing response to inflammatory disease therapy. The methods include performing immunoassays to generate scores based on quantitative data for expression of biomarkers relating to inflammatory biomarkers to predict flare and sustained remission. The methods further include recommending discontinuation of a therapeutic regimen, or no change in a therapeutic regimen, based on the predictions generated by the quantitative data.

Description

BIOMARKERS AND METHODS FOR PREDICTING RESPONSE TO DISCONTINUATION OF INFLAMMATORY DISEASE THERAPY
BACKGROUND
[0001] This application claims priority to U.S. provisional application number
62/302,487, filed March 2, 2016, the entire contents of which are hereby incorporated by reference.
[0002] This application is directed to the fields of bioinformatics and inflammatory and autoimmune diseases, with methods of assessing response to inflammatory disease therapy. Rheumatoid arthritis ("RA") is an example of an inflammatory disease, and is a chronic, systemic autoimmune disorder. It is one of the most common systemic autoimmune diseases worldwide. The immune system of the RA subject targets the subject's joints as well as other organs including the lung, blood vessels and pericardium, leading to inflammation of the joints (arthritis), widespread endothelial inflammation, and even destruction of joint tissue. Erosions and joint space narrowing are largely irreversible and result in cumulative disability.
[0003] The precise etiology of RA has not been established, but underlying disease pathogenesis is multifactorial and includes inflammation and immune dysregulation. The precise mechanisms involved are different in individual subjects, and can change in those subjects over time. Variables such as race, sex, genetics, hormones, and environmental factors can impact the development and severity of RA disease. Emerging data are also beginning to reveal the characteristics of new RA subject subgroups and complex overlapping relationships with other autoimmune disorders. Disease duration and level of inflammatory activity is also associated with other comorbidities such as risk of lymphoma, extra-articular manifestations, and cardiovascular disease. See, e.g., S. Banerjee et a/., Am. J. Cardiol. 2008, 101(8): 1201-1205; E. Baecklund et a/., Arth. Rheum. 2006, 54(3):692-701; and, N. Goodson et a/., Ann. Rheum. Dis. 2005, 64(11): 1595-1601.
[0004] Traditional models for treating RA are based on the expectation that controlling disease activity (i.e., inflammation) in an RA subject should slow down or prevent disease progression, in terms of radiographic progression, tissue destruction, cartilage loss and joint erosion. There is evidence, however, that disease activity and disease progression can be uncoupled, and may not always function completely in tandem. Indeed, different cell signaling pathways and mediators are involved in these two processes. See W. van den Berg et al., Arth. Rheum. 2005, 52:995-999. The uncoupling of disease progression and disease activity is described in a number of RA clinical trials and animal studies. See, e.g., PE Lipsky et al, N. Engl. J. Med. 2003, 343 : 1594-602.; AK Brown et al, Arth. Rheum. 2006, 54:3761-3773; and, AR Pettit et al, Am. J. Pathol. 2001, 159: 1689-99. Studies of RA subjects indicate limited association between clinical and radiographic responses. See E. Zatarain and V. Strand, Nat. Clin. Pract. Rheum. 2006, 2(11):611-618 (Review). RA subjects have been described who demonstrated radiographic benefits from combination treatment with infliximab and
methotrexate (MTX), yet did not demonstrate any clinical improvement, as measured by DAS (Disease Activity Score) and CRP (C -reactive protein). See JS Smolen et al, Arth. Rheum. 2005, 52(4): 1020-30. To track the uncoupling of disease activity and remission, and to analyze the relationship between disease activity, treatment, and progression, RA subjects should be assessed frequently for both disease activity and progression during therapy. Recent advances in assessing inflammatory disease activity and progression are described in US 2011/0137851, which is hereby incorporated by reference in its entirety.
[0005] There is a need to classify subjects by disease activity in order to ensure that each receives treatment that is appropriate and optimized for that patient. For patients with early RA ("eRA"), MTX is recommended as first-line treatment and in MTX non-responders both the addition of conventional non-biological disease modifying anti -rheumatic ("DMARD") therapy and/or biological {e.g., anti-T F) therapy are known in the art.
[0006] Biological agents have provided unprecedented efficacy and improvement in long-term outcomes for patients RA. However, these treatments can be associated with increased risk of serious infection and substantial costs, and in some patients lifelong treatment may not be desirable. However, premature or improper withdrawal from therapy can result in disease flare, which runs the risk of irreversible joint damage. Therefore, there is interest in reducing or stopping biologic treatment in RA patients who achieve sustained remission. The European League Against Rheumatism (EULAR) and the American College of Rheumatology both recommend that tapering both non-biologic DMARDs and biological agents be considered for patients who are in persistent remission (Singh JA, et al, Arthritis Care Res (Hoboken). 2016 Jan; 68(1): 1-25 (2016); Smolen JS, et al, Ann Rheum Dis. Mar;73(3):492-509 (2014)). Clearly there exists a need to accurately predict which subjects receiving biologic drug therapy for autoimmune diseases can be removed from the therapy in order to establish an optimal treatment regimen.
SUMMARY
[0007] The present teachings relate to biomarkers associated with inflammatory disease, and with autoimmune disease, including RA, and methods of using the biomarkers to measure disease activity in a subject, and in particular, in response to therapy.
[0008] In one embodiment, a method for recommending a therapeutic regimen in a subject having an autoimmune disorder is provided. The method comprises a) placing the subject on a therapy regimen; b) determining whether the subject responds to the therapy regimen; c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); resistin (RETN); serum amyloid Al (SAAl); tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA) if the subject responds to the therapy regiment response as determined in step b); and d)
recommending i) reduction of the therapy regimen if the score is low; or ii) no change in the therapy regimen if the score is high. In an embodiment, the at least four biomarkers comprise IL6, EGF, SAAl, and CRP. In an embodiment, the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAAl, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1. In an embodiment, performance of the at least one immunoassay comprises: obtaining the sample, wherein the sample comprises the protein markers; contacting the sample with a plurality of distinct reagents; generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data. In an embodiment, the immunoassays comprise a multiplex assay. In an embodiment, the autoimmune disorder is an arthritic disorder. In an embodiment, the arthritic disorder is rheumatoid arthritis. In an embodiment, the rheumatoid arthritis is early rheumatoid arthritis (eRA). In an embodiment, the therapy regimen comprises one or more of methotrexate (MTX) and a biologic. In an embodiment, the biologic comprises a TNF inhibitor. In an embodiment, the TNF inhibitor is adalimumab. In an embodiment, the score is predictive of a clinical assessment. In an embodiment, the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28- ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC). In an
embodiment, the score is low if on a scale of 1-100, the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is greater than about 30. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of sustained clinical remission (SC-REM) and the score is less than or equal to about 23. In an embodiment, the score is high if on a scale of 1- 100, the score is predictive of SC-REM and the score is greater than about 23. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of functional remission and the score is less than or equal to about 26. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of functional remission and the score is greater than about 26. In an embodiment, the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission. In an embodiment, the remission is sustained if DAS28-ESR is <2.6 for >24 weeks. In an embodiment, the remission is SC-REM. In an embodiment, the remission is functional remission. In an embodiment, the reduction of the therapy regimen is discontinuation of the therapy regimen.
[0009] In another embodiment, a method for predicting flare or remission in a subject having an autoimmune disorder is provided. The method comprises: a) placing the subject on a therapy regimen; b) determining whether the subject responds to the therapy regimen; c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); resistin (RETN); serum amyloid Al (SAA1); tumor necrosis factor receptor superfamily, member 1 A (TNFRSF1 A); vascular cell adhesion molecule 1 (VCAMl); and, vascular endothelial growth factor A (VEGFA) if the subject responds to the therapy regiment response as determined in step b); and d) predicting i) no flare or sustained remission if the score is low; or ii) flare or no sustained remission if the score is high. In an embodiment, the at least four biomarkers comprise IL6, EGF, SAA1, and CRP. In an embodiment, the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAA1, VCAMl, CRP, MMP1, MMP3, T FRSF1A, RETN, and CHI3L1. In an embodiment, performance of the at least one immunoassay comprises: obtaining the sample, wherein the sample comprises the protein markers; contacting the sample with a plurality of distinct reagents; generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data. In an embodiment, the immunoassays comprise a multiplex assay. In an embodiment, the autoimmune disorder is an arthritic disorder. In an embodiment, the arthritic disorder is rheumatoid arthritis. In an embodiment, the rheumatoid arthritis is early rheumatoid arthritis (eRA). In an embodiment, the therapy regimen comprises one or more of methotrexate (MTX) and a biologic. In an embodiment, the biologic comprises a TNF inhibitor. In an embodiment, the TNF inhibitor is adalimumab. In an embodiment, the score is predictive of a clinical assessment. In an embodiment, the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28-ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC). In an embodiment, the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is sustained clinical remission (SC-REM) and the score is less than or equal to about 23. In an embodiment, the score is high if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is SC-REM and the score is greater than about 23. In an embodiment, the score is low if on a scale of 1-100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is less than or equal to about 26. In an embodiment, the score is high if on a scale of 1- 100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is greater than about 26. In an embodiment, the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission. In an embodiment, the remission is sustained if DAS28-ESR is <2.6 for >24 weeks. In an embodiment, the flare is DAS28-ESR>3.2. In an embodiment, the remission is sustained clinical remission (SC-REM). In an embodiment, the remission is functional remission. In an
embodiment, the SC-REM is DAS28-ESR<2.6. In an embodiment, the therapy regimen is not discontinued if flare or no remission is predicted.
BRIEF DESCRIPTION OF THE DRAWINGS
[0010] The skilled artisan will understand that the drawings, described below, are for illustration purposes only. The drawings are not intended to limit the scope of the present teachings in any way.
[0011] Figure 1 depicts correlations of the MBDA algorithm predictions and CRP with clinical assessments of disease activity, as described in Example 1.
[0012] Figure 2 depicts the MBDA scores for subjects at baseline and six-month visits, according to the description in Example 1. MBDA scores are shown by treatment arm and time point. Only subjects with MBDA scores available at both baseline and six months are shown.
[0013] Figure 3A depicts the distribution of MBDA scores at ADA discontinuation for 3
MBDA categories which combine remission and low disease activity (N=42). Figure 3B depicts the distribution of MBDA scores at the time of ADA discontinuation for 4 MBDA categories (N=42).
[0014] Figure 4 depicts p-values by univariate logistic regression 6 months after ADA discontinuation compared with and without flare, clinical remission, and functional remission.
[0015] Figure 5 depicts p-values by univariate logistic regression 1 year after ADA discontinuation compared with and without flare, clinical remission, and functional remission. [0016] Figure 6 depicts MBDA scores at ADA discontinuation: flare vs. no flare.
P=0.293 for 6 months after discontinuation and p=0.058 for 1 year after discontinuation by two- sided Wilcoxon rank sum test.
[0017] Figure 7 depicts flare rate by 3 MBDA categories. P-values were determined by a two-sided Cochran- Armitage trend test. Figure 7A depicts flare rate at 6 months post ADA discontinuation. Figure 7B depicts flare rate at 1-year post ADA discontinuation.
[0018] Figure 8 depicts flare rate by 4 MBDA categories. P-values were determined by a two-sided Cochran- Armitage trend test.
[0019] Figure 9 depicts MBDA scores at ADA discontinuation: SC-REM vs. no SC-
REM at 6 months (Figure 9A) and 1 year (Figure 9B). P=0.06 for 6 months after discontinuation and p=0.039 for 1 year after discontinuation (difference in median MBDA: 9) by two-sided Wilcoxon rank sum test.
[0020] Figure 10 depicts sustained clinical remission rate by 3 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
[0021] Figure 11 depicts sustained clinical remission rate by 4 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test. Figure 11 A depicts rates of SC-REM at 6 months. Figure 1 IB depicts rates of SC-REM at 1 year.
[0022] Figure 12 depicts MBDA scores at ADA discontinuation: sustained functional remission vs. no sustained functional remission. P=0.051 for 6 months after discontinuation and p=0.097 for 1 year after discontinuation by two-sided Wilcoxon rank sum test.
[0023] Figure 13 depicts sustained functional remission rate by 3 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test.
[0024] Figure 14 depicts sustained functional remission rate by 4 MBDA categories. P- values were determined by a two-sided Cochran- Armitage trend test. [0025] Figure 15 depicts Kaplan-Meier estimates examining the time to flare show a lower probability of flare among subjects with low (<30) or moderate (30-44) MBDA scores at baseline, compared to those with high MBDA scores (>44).
[0026] Figure 16 depicts receiver operating characteristic (ROC) curves of clinical remission, flare, and functional remission at 6 months and 1 year after discontinuation of ADA.
[0027] Figure 17 illustrates a high-level block diagram of a computer (1600). Illustrated are at least one processor (1602) coupled to a chipset (1604). Also coupled to the chipset (1604) are a memory (1606), a storage device (1608), a keyboard (1610), a graphics adapter (1612), a pointing device (1614), and a network adapter (1616). A display (1618) is coupled to the graphics adapter (1612). In one embodiment, the functionality of the chipset (1604) is provided by a memory controller hub 1620) and an I/O controller hub (1622). In another embodiment, the memory (1606) is coupled directly to the processor (1602) instead of the chipset (1604). The storage device 1608 is any device capable of holding data, like a hard drive, compact disk readonly memory (CD-ROM), DVD, or a solid-state memory device. The memory (1606) holds instructions and data used by the processor (1602). The pointing device (1614) may be a mouse, track ball, or other type of pointing device, and is used in combination with the keyboard (1610) to input data into the computer system (1600). The graphics adapter (1612) displays images and other information on the display (1618). The network adapter (1616) couples the computer system (1600) to a local or wide area network.
DESCRIPTION OF VARIOUS EMBODIMENTS
[0028] These and other features of the present teachings will become more apparent from the description herein. While the present teachings are described in conjunction with various embodiments, it is not intended that the present teachings be limited to such embodiments. On the contrary, the present teachings encompass various alternatives, modifications, and equivalents, as will be appreciated by those of skill in the art.
[0029] The present teachings relate generally to the identification of biomarkers associated with subjects having inflammatory and/or autoimmune diseases, for example RA, and that are useful in determining or assessing disease activity, and in particular, in response to inflammatory disease therapy for recommending optimal therapy.
[0030] Most of the words used in this specification have the meaning that would be attributed to those words by one skilled in the art. Words specifically defined in the specification have the meaning provided in the context of the present teachings as a whole, and as are typically understood by those skilled in the art. In the event that a conflict arises between an art- understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification, the specification shall control. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise.
Definitions
[0031] "Accuracy" refers to the degree that a measured or calculated value conforms to its actual value. "Accuracy" in clinical testing relates to the proportion of actual outcomes (true positives or true negatives, wherein a subject is correctly classified as having disease or as healthy/normal, respectively) versus incorrectly classified outcomes (false positives or false negatives, wherein a subject is incorrectly classified as having disease or as healthy/normal, respectively). Other and/or equivalent terms for "accuracy" can include, for example,
"sensitivity," "specificity," "positive predictive value (PPV)," "the AUC," "negative predictive value (NPV)," "likelihood," and "odds ratio." "Analytical accuracy," in the context of the present teachings, refers to the repeatability and predictability of the measurement process. Analytical accuracy can be summarized in such measurements as, e.g., coefficients of variation (CV), and tests of concordance and calibration of the same samples or controls at different times or with different assessors, users, equipment, and/or reagents. See, e.g., R. Vasan, Circulation 2006, 113(19):2335-2362 for a summary of considerations in evaluating new biomarkers.
[0032] The term "administering" as used herein refers to the placement of a composition into a subject by a method or route that results in at least partial localization of the composition at a desired site such that a desired effect is produced. Routes of administration include both local and systemic administration. Generally, local administration results in more of the composition being delivered to a specific location as compared to the entire body of the subject, whereas, systemic administration results in delivery to essentially the entire body of the subject.
[0033] The term "algorithm" encompasses any formula, model, mathematical equation, algorithmic, analytical or programmed process, or statistical technique or classification analysis that takes one or more inputs or parameters, whether continuous or categorical, and calculates an output value, index, index value or score. Examples of algorithms include but are not limited to ratios, sums, regression operators such as exponents or coefficients, biomarker value
transformations and normalizations (including, without limitation, normalization schemes that are based on clinical parameters such as age, gender, ethnicity, etc.), rules and guidelines, statistical classification models, and neural networks trained on populations. Also of use in the context of biomarkers are linear and non-linear equations and statistical classification analyses to determine the relationship between (a) levels of biomarkers detected in a subject sample and (b) the level of the respective subject's disease activity.
[0034] The term "analyte" in the context of the present teachings can mean any substance to be measured, and can encompass biomarkers, markers, nucleic acids, electrolytes, metabolites, proteins, sugars, carbohydrates, fats, lipids, cytokines, chemokines, growth factors, proteins, peptides, nucleic acids, oligonucleotides, metabolites, mutations, variants, polymorphisms, modifications, fragments, subunits, degradation products and other elements. For simplicity, standard gene symbols may be used throughout to refer not only to genes but also gene products/proteins, rather than using the standard protein symbol; e.g., APOAl as used herein can refer to the gene APOAl and also the protein ApoAI. In general, hyphens are dropped from analyte names and symbols herein (IL-6 = IL6).
[0035] To "analyze" includes determining a value or set of values associated with a sample by measurement of analyte levels in the sample. "Analyze" may further comprise and comparing the levels against constituent levels in a sample or set of samples from the same subject or other subject(s). The biomarkers of the present teachings can be analyzed by any of various conventional methods known in the art. Some such methods include but are not limited to: measuring serum protein or sugar or metabolite or other analyte level, measuring enzymatic activity, and measuring gene expression. [0036] The term "antibody" refers to any immunoglobulin-like molecule that reversibly binds to another with the required selectivity. Thus, the term includes any such molecule that is capable of selectively binding to a biomarker of the present teachings. The term includes an immunoglobulin molecule capable of binding an epitope present on an antigen. The term is intended to encompass not only intact immunoglobulin molecules, such as monoclonal and polyclonal antibodies, but also antibody isotypes, recombinant antibodies, bi-specific antibodies, humanized antibodies, chimeric antibodies, anti-idiopathic (anti-ID) antibodies, single-chain antibodies, Fab fragments, F(ab') fragments, fusion protein antibody fragments, immunoglobulin fragments, Fv fragments, single chain Fv fragments, and chimeras comprising an immunoglobulin sequence and any modifications of the foregoing that comprise an antigen recognition site of the required selectivity.
[0037] "Autoimmune disease" encompasses any disease, as defined herein, resulting from an immune response against substances and tissues normally present in the body.
Examples of suspected or known autoimmune diseases include rheumatoid arthritis, early rheumatoid arthritis, axial spondylarthritis, juvenile idiopathic arthritis, seronegative spondyloarthropathies, ankylosing spondylitis, psoriatic arthritis, antiphospholipid antibody syndrome, autoimmune hepatitis, Behcet's disease, bullous pemphigoid, coeliac disease, Crohn's disease, dermatomyositis, Goodpasture's syndrome, Graves' disease, Hashimoto's disease, idiopathic thrombocytopenic purpura, IgA nephropathy, Kawasaki disease, systemic lupus erythematosus, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, polymyositis, primary biliary cirrhosis, psoriasis, scleroderma, Sjogren's syndrome, ulcerative colitis, vasculitis, Wegener's granulomatosis, temporal arteritis, Takayasu's arteritis, Henoch- Schonlein purpura, leucocytoclastic vasculitis, polyarteritis nodosa, Churg-Strauss Syndrome, and mixed cryoglobulinemic vasculitis.
[0038] A "biologic" or "biotherapy" or "biopharmaceutical" is a pharmaceutical therapy product manufactured or extracted from a biological substance. A biologic can include vaccines, blood or blood components, allergenics, somatic cells, gene therapies, tissues, recombinant proteins, and living cells; and can be composed of sugars, proteins, nucleic acids, living cells or tissues, or combinations thereof. Examples of biologic drugs can include but are not limited to biological agents that target the tumor necrosis factor (T F)-alpha molecules and the TNF inhibitors, such as infliximab, adalimumab, etanercept and golimumab. Other classes of biologic drugs include IL1 inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab.
[0039] "Biomarker," "biomarkers," "marker" or "markers" in the context of the present teachings encompasses, without limitation, cytokines, chemokines, growth factors, proteins, peptides, nucleic acids, oligonucleotides, and metabolites, together with their related metabolites, mutations, isoforms, variants, polymorphisms, modifications, fragments, subunits, degradation products, elements, and other analytes or sample-derived measures. Biomarkers can also include mutated proteins, mutated nucleic acids, variations in copy numbers and/or transcript variants. Biomarkers also encompass non-blood borne factors and non-analyte physiological markers of health status, and/or other factors or markers not measured from samples (e.g., biological samples such as bodily fluids), such as clinical parameters and traditional factors for clinical assessments. Biomarkers can also include any indices that are calculated and/or created mathematically. Biomarkers can also include combinations of any one or more of the foregoing measurements, including temporal trends and differences. Biomarkers can include, but are not limited to, apolipoprotein A-I (APOA1); apolipoprotein C-III (APOC3); calprotectin; chemokine (C-C motif) ligand 22 (CCL22); chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1, or YKL- 40); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); intercellular adhesion molecule 1 (ICAMl); ICTP; interleukin 18 (interferon-gamma- inducing factor) (IL18); interleukin 1, beta (IL1B); interleukin 6 receptor (IL6R); interleukin 8 (IL8); keratan sulfate, or KS; leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); pyridinoline (crosslinks formed in collagen, derived from three lysine residues), which may be referred to herein as PYD; resistin (RETN); serum amyloid Al (SAA1); tumor necrosis factor receptor superfamily, member 1A (TNFRSF 1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA).
[0040] A "clinical assessment," or "clinical datapoint" or "clinical endpoint," in the context of the present teachings can refer to a measure of disease activity or severity. A clinical assessment can include a score, a value, or a set of values that can be obtained from evaluation of a sample (or population of samples) from a subject or subjects under determined conditions. A clinical assessment can also be a questionnaire completed by a subject. A clinical assessment can also be predicted by biomarkers and/or other parameters. One of skill in the art will recognize that the clinical assessment for RA, as an example, can comprise, without limitation, one or more of the following: DAS (defined herein), DAS28, DAS28-ESR, DAS28-CRP, health assessment questionnaire (HAQ), modified HAQ (mHAQ), multi-dimensional HAQ (MDHAQ), visual analog scale (VAS), physician global assessment VAS, patient global assessment VAS, pain VAS, fatigue VAS, overall VAS, sleep VAS, simplified disease activity index (SDAI), clinical disease activity index (CDAI), routine assessment of patient index data (RAPID), RAPID3, RAPID4, RAPID 5, American College of Rheumatology (ACR), ACR20, ACR50, ACR70, SF-36 (a well-validated measure of general health status), RA MRI score (RAMRIS; or RA MRI scoring system), total Sharp score (TSS), van der Heijde-modified TSS, van der Heijde- modified Sharp score (or Sharp-van der Heijde score (SHS)), Larsen score, TJC, swollen joint count (SJC), CRP titer (or level), and erythrocyte sedimentation rate (ESR).
[0041] The term "clinical parameters" in the context of the present teachings
encompasses all measures of the health status of a subject. A clinical parameter can be used to derive a clinical assessment of the subject's disease activity. Clinical parameters can include, without limitation: therapeutic regimen (including but not limited to DMARDs, whether conventional or biologies, steroids, etc.), TJC, SJC, morning stiffness, arthritis of three or more joint areas, arthritis of hand joints, symmetric arthritis, rheumatoid nodules, radiographic changes and other imaging, gender/sex, age, race/ethnicity, disease duration, diastolic and systolic blood pressure, resting heart rate, height, weight, body-mass index, family history, CCP status {i.e., whether subject is positive or negative for anti-CCP antibody), CCP titer, RF status, RF titer, ESR, CRP titer, menopausal status, and whether a smoker/non-smoker.
[0042] "Clinical assessment" and "clinical parameter" are not mutually exclusive terms.
There may be overlap in members of the two categories. For example, CRP concentration can be used as a clinical assessment of disease activity; or, it can be used as a measure of the health status of a subject, and thus serve as a clinical parameter.
[0043] The term "computer" carries the meaning that is generally known in the art; that is, a machine for manipulating data according to a set of instructions. For illustration purposes only, FIG. 15 is a high-level block diagram of a computer (1600). As is known in the art, a "computer" can have different and/or other components than those shown in FIG. 15. In addition, the computer 1600 can lack certain illustrated components. Moreover, the storage device (1608) can be local and/or remote from the computer (1600) (such as embodied within a storage area network (SAN)). As is known in the art, the computer (1600) is adapted to execute computer program modules for providing functionality described herein. As used herein, the term "module" refers to computer program logic utilized to provide the specified functionality. Thus, a module can be implemented in hardware, firmware, and/or software. In one
embodiment, program modules are stored on the storage device (1608), loaded into the memory (1606), and executed by the processor (1602). Embodiments of the entities described herein can include other and/or different modules than the ones described here. In addition, the
functionality attributed to the modules can be performed by other or different modules in other embodiments. Moreover, this description occasionally omits the term "module" for purposes of clarity and convenience.
[0044] The term "cytokine" in the present teachings refers to any substance secreted by specific cells that can be of the immune system that carries signals between cells and thus has an effect on other cells. The term "cytokines" encompasses "growth factors." "Chemokines" are also cytokines. They are a subset of cytokines that are able to induce chemotaxis in cells; thus, they are also known as "chemotactic cytokines."
[0045] "DAS" refers to the Disease Activity Score, a measure of the activity of RA in a subject, well-known to those of skill in the art. See D. van der Heijde et al., Ann. Rheum. Dis. 1990, 49(11):916-920. "DAS" as used herein refers to this particular Disease Activity Score. The "DAS28" involves the evaluation of 28 specific joints. It is a current standard well- recognized in research and clinical practice. Because the DAS28 is a well-recognized standard, it may be referred to as "DAS." Although "DAS" may refer to calculations based on 66/68 or 44 joint counts, unless otherwise specified, "DAS" herein will encompass the DAS28. Unless otherwise specified herein, the term "DAS28," as used in the present teachings, can refer to a DAS28-ESR or DAS28-CRP, as obtained by any of the four formulas described above; or, DAS28 can refer to another reliable DAS28 formula as may be known in the art. [0046] A DAS28 can be calculated for an RA subject according to the standard as outlined at the das-score. nl website, maintained by the Department of Rheumatology of the University Medical Centre in Nijmegen, the Netherlands. The number of swollen joints, or swollen joint count out of a total of 28 (SJC28), and tender joints, or tender joint count out of a total of 28 (TJC28) in each subject is assessed. In some DAS28 calculations the subject's general health (GH) is also a factor, and can be measured on a 100mm Visual Analogue Scale (VAS). GH may also be referred to herein as PG or PGA, for "patient global health assessment" (or merely "patient global assessment"). A "patient global health assessment VAS," then, is GH measured on a Visual Analogue Scale.
[0047] "DAS28-CRP" (or "DAS28CRP") is a DAS28 assessment calculated using CRP in place of ESR (see below). CRP is produced in the liver. Normally there is little or no CRP circulating in an individual's blood serum - CRP is generally present in the body during episodes of acute inflammation or infection, so that a high or increasing amount of CRP in blood serum can be associated with acute infection or inflammation. A blood serum level of CRP greater than 1 mg/dL is usually considered high. Most inflammation and infections result in CRP levels greater than 10 mg/dL. The amount of CRP in subject sera can be quantified using, for example, the DSL- 10-42100 ACTIVE® US C-Reactive Protein Enzyme-Linked Immunosorbent Assay (ELISA), developed by Diagnostics Systems Laboratories, Inc. (Webster, TX). CRP production is associated with radiological progression in RA. See M. Van Leeuwen et al, Br. J. Rheum. 1993, 32(suppl.):9-13). CRP is thus considered an appropriate alternative to ESR in measuring RA disease activity. See R. Mallya et al, J. Rheum. 1982, 9(2):224-228, and F. Wolfe, J.
Rheum. 1997, 24: 1477-1485.
[0048] The DAS28-CRP can be calculated according to either of the formulas below, with or without the GH factor, where "CRP" represents the amount of this protein present in a subject's blood serum in mg/L, "sqrt" represents the square root, and "In" represents the natural logarithm:
(a) DAS28-CRP with GH (or DAS28-CRP4) = (0.56*sqrt(TJC28) + 0.28*sqrt(SJC28) +
0.36*ln(CRP+l)) + (0.014 * GH) + 0.96; or, (b) DAS28-CRP without GH (or DAS28-CRP3) = (0.56*sqrt(TJC28) + 0.28*sqrt(SJC28) + 0.36*ln(CRP+l)) * 1.10 + 1.15.
[0049] The "DAS28-ESR" is a DAS28 assessment wherein the ESR for each subject is also measured (in mm/hour). The DAS28-ESR can be calculated according to the formula:
(a) DAS28-ESR with GH (or DAS28-ESR4) = 0.56 * sqrt(TJC28) + 0.28 * sqrt(SJC28) + 0.70 * In(ESR) + 0.014 * GH; or,
(b) DAS28-ESR without GH = 0.56 * sqrt(TJC28) + 0.28 * sqrt(SJC28) + 0. 70 * In(ESR) * 1.08 + 0.16.
[0050] A "dataset" is a set of numerical values resulting from evaluation of a sample (or population of samples) under a desired condition. The values of the dataset can be obtained, for example, by experimentally obtaining measures from a sample and constructing a dataset from these measurements; or alternatively, by obtaining a dataset from a service provider such as a laboratory, or from a database or a server on which the dataset has been stored.
[0051] A "difference" as used herein refers to an increase or decrease in the measurable expression of a biomarker or panel of biomarkers as compared to the measurable expression of the same biomarker or panel of biomarkers in a second samples.
[0052] The term "disease" in the context of the present teachings encompasses any disorder, condition, sickness, ailment, etc. that manifests in, e.g., a disordered or incorrectly functioning organ, part, structure, or system of the body, and results from, e.g., genetic or developmental errors, infection, poisons, nutritional deficiency or imbalance, toxicity, or unfavorable environmental factors.
[0053] A DMARD can be conventional or biologic. Examples of DMARDs that are generally considered conventional include, but are not limited to, MTX, azathioprine (AZA), bucillamine (BUC), chloroquine (CQ), ciclosporin (CSA, or cyclosporine, or cyclosporin), doxycycline (DOXY), hydroxychloroquine (HCQ), intramuscular gold (IM gold), leflunomide (LEF), levofloxacin (LEV), and sulfasalazine (SSZ). Examples of other conventional DMARDs include, but are not limited to, folinic acid, D-pencillamine, gold auranofin, gold aurothioglucose, gold thiomalate, cyclophosphamide, and chlorambucil. Examples of biologic DMARDs (or biologic drugs) include but are not limited to biological agents that target the tumor necrosis factor (TNF)-alpha molecules such as infliximab, adalimumab, etanercept and golimumab. Other classes of biologic DMARDs include IL1 inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab.
[0054] The term "flare" as used herein is a sudden and severe increase in the onset of symptoms and clinical manifestations including, but not limited to, an increase in SJC, increase in TJC, increase in serologic markers of inflammation (e.g., CRP and ESR), decrease in subject function (e.g., ability to perform basic daily activities), increase in morning stiffness, and increases in pain that commonly lead to therapeutic intervention and potentially to treatment intensification.
[0055] An "immunoassay" as used herein refers to a biochemical assay that uses one or more antibodies to measure the presence or concentration of an analyte or biomarker in a biological sample.
[0056] "Inflammatory disease" in the context of the present teachings encompasses, without limitation, any disease, as defined herein, resulting from the biological response of vascular tissues to harmful stimuli, including but not limited to such stimuli as pathogens, damaged cells, irritants, antigens and, in the case of autoimmune disease, substances and tissues normally present in the body. Non-limiting examples of inflammatory disease include rheumatoid arthritis (RA), eRA, ankylosing spondylitis, psoriatic arthritis, atherosclerosis, asthma, autoimmune diseases, chronic inflammation, chronic prostatitis, glomerulonephritis, hypersensitivities, inflammatory bowel diseases, pelvic inflammatory disease, reperfusion injury, transplant rejection, and vasculitis.
[0057] "Interpretation function," as used herein, means the transformation of a set of observed data into a meaningful determination of particular interest; e.g., an interpretation function may be a predictive model that is created by utilizing one or more statistical algorithms to transform a dataset of observed biomarker data into a meaningful determination of disease activity or the disease state of a subject. [0058] "Measuring" or "measurement" in the context of the present teachings refers to determining the presence, absence, quantity, amount, or effective amount of a substance in a clinical or subject-derived sample, including the concentration levels of such substances, or evaluating the values or categorization of a subject's clinical parameters.
[0059] A "multiplex assay" as used herein refers to an assay that simultaneously measure multiple analytes, e.g., protein analytes, in a single run or cycle of the assay.
[0060] "Performance" in the context of the present teachings relates to the quality and overall usefulness of, e.g., a model, algorithm, or diagnostic or prognostic test. Factors to be considered in model or test performance include, but are not limited to, the clinical and analytical accuracy of the test, use characteristics such as stability of reagents and various components, ease of use of the model or test, health or economic value, and relative costs of various reagents and components of the test. Performing can mean the act of carrying out a function.
[0061] A "population" is any grouping of subjects of like specified characteristics. The grouping could be according to, for example but without limitation, clinical parameters, clinical assessments, therapeutic regimen, disease status (e.g. with disease or healthy), level of disease activity, etc. In the context of using the MBDA score in comparing disease activity between populations, an aggregate value can be determined based on the observed MBDA scores of the subjects of a population; e.g., at particular timepoints in a longitudinal study. The aggregate value can be based on, e.g., any mathematical or statistical formula useful and known in the art for arriving at a meaningful aggregate value from a collection of individual datapoints; e.g., mean, median, median of the mean, etc.
[0062] A "predictive model," which term may be used synonymously herein with
"multivariate model" or simply a "model," is a mathematical construct developed using a statistical algorithm or algorithms for classifying sets of data. The term "predicting" refers to generating a value for a datapoint without actually performing the clinical diagnostic procedures normally or otherwise required to produce that datapoint; "predicting" as used in this modeling context should not be understood solely to refer to the power of a model to predict a particular outcome. Predictive models can provide an interpretation function; e.g., a predictive model can be created by utilizing one or more statistical algorithms or methods to transform a dataset of observed data into a meaningful determination of disease activity or the disease state of a subject. See Calculation of the MBDA score for some examples of statistical tools useful in model development.
[0063] A "prognosis" is a prediction as to the likely outcome of a disease. Prognostic estimates are useful in, e.g., determining an appropriate therapeutic regimen for a subject.
[0064] A "quantitative dataset" or "quantitative data" as used in the present teachings, refers to the data derived from, e.g., detection and composite measurements of expression of a plurality of biomarkers {i.e., two or more) in a subject sample. The quantitative dataset can be used to generate a score for the identification, monitoring and treatment of disease states, and in characterizing the biological condition of a subject. It is possible that different biomarkers will be detected depending on the disease state or physiological condition of interest.
[0065] "Recommending" as used herein refers to making a recommendation for a therapeutic regimen or excluding {i.e., not recommending) a certain therapeutic regimen for a subject. Such a recommendation shall serve optionally together with other information as a basis for a clinician to apply a certain therapeutic regimen for an individual subject.
[0066] The term "remission" refers to the state of absence of disease activity in patients known to have a chronic illness that usually cannot be cured. The term "sustained clinical remission" or "SC-REM" as used herein refers to a state of clinical remission sustained as evaluated based on clinical assessments, for example, DAS28 for at least six months. The term "functional remission" as used herein refers to a state of remission as evaluated using functional assessment measures such as but not limited to HAQ. Sustained remission can be used interchangeably with maintained remission.
[0067] A "sample" in the context of the present teachings refers to any biological sample that is isolated from a subject. A sample can include, without limitation, a single cell or multiple cells, fragments of cells, an aliquot of body fluid, whole blood, platelets, serum, plasma, red blood cells, white blood cells or leucocytes, endothelial cells, tissue biopsies, synovial fluid, lymphatic fluid, ascites fluid, and interstitial or extracellular fluid. The term "sample" also encompasses the fluid in spaces between cells, including synovial fluid, gingival crevicular fluid, bone marrow, cerebrospinal fluid (CSF), saliva, mucous, sputum, semen, sweat, urine, or any other bodily fluids. "Blood sample" can refer to whole blood or any fraction thereof, including blood cells, red blood cells, white blood cells or leucocytes, platelets, serum and plasma.
Samples can be obtained from a subject by means including but not limited to venipuncture, excretion, ejaculation, massage, biopsy, needle aspirate, lavage, scraping, surgical incision, or intervention or other means known in the art.
[0068] A "score" is a value or set of values selected so as to provide a quantitative measure of a variable or characteristic of a subject's condition, and/or to discriminate, differentiate or otherwise characterize a subject's condition. The value(s) comprising the score can be based on, for example, quantitative data resulting in a measured amount of one or more sample constituents obtained from the subject, or from clinical parameters, or from clinical assessments, or any combination thereof. In certain embodiments the score can be derived from a single constituent, parameter or assessment, while in other embodiments the score is derived from multiple constituents, parameters and/or assessments. The score can be based upon or derived from an interpretation function; e.g., an interpretation function derived from a particular predictive model using any of various statistical algorithms known in the art. A "change in score" can refer to the absolute change in score, e.g. from one time point to the next, or the percent change in score, or the change in the score per unit time (e.g., the rate of score change). A "score" as used herein can be used interchangeably with MBDA score as defined below.
[0069] A "multi-biomarker disease activity score," "MBDA score," or simply "MBDA," in the context of the present teachings, is a score that uses quantitative data to provide a quantitative measure of inflammatory disease activity or the state of inflammatory disease in a subject. A set of data from particularly selected biomarkers, such as from the disclosed set of biomarkers, is input into an interpretation function according to the present teachings to derive the MBDA score. The interpretation function, in some embodiments, can be created from predictive or multivariate modeling based on statistical algorithms. Input to the interpretation function can comprise the results of testing two or more of the disclosed set of biomarkers, alone or in combination with clinical parameters and/or clinical assessments, also described herein. In some embodiments of the present teachings, the MBDA score is a quantitative measure of autoimmune disease activity. In some embodiments, the MBDA score is a quantitative measure of RA disease activity. MBDA as used herein can also refer to a VDA or VECTRA® DA score.
[0070] A "multiplex assay" as used herein refers to an assay that simultaneously measures multiple analytes, e.g., protein analytes, in a single run or cycle of the assay.
[0071] "Statistically significant" in the context of the present teachings means an observed alteration is greater than what would be expected to occur by chance alone (e.g., a "false positive"). Statistical significance can be determined by any of various methods well- known in the art. An example of a commonly used measure of statistical significance is the p- value. The p-value represents the probability of obtaining a given result equivalent to a particular datapoint, where the datapoint is the result of random chance alone. A result is often considered highly significant (not random chance) at a p-value less than or equal to 0.05.
[0072] A "subject" in the context of the present teachings is generally a mammal. The subject can be a patient. The term "mammal" as used herein includes but is not limited to a human, non-human primate, dog, cat, mouse, rat, cow, horse, and pig. Mammals other than humans can be advantageously used as subjects that represent animal models of inflammation. A subject can be male or female. A subject can be one who has been previously diagnosed or identified as having an inflammatory disease. A subject can be one who has already undergone, or is undergoing, a therapeutic intervention for an inflammatory disease. A subject can also be one who has not been previously diagnosed as having an inflammatory disease; e.g., a subject can be one who exhibits one or more symptoms or risk factors for an inflammatory condition, or a subject who does not exhibit symptoms or risk factors for an inflammatory condition, or a subject who is asymptomatic for inflammatory disease.
[0073] A "therapeutic regimen," "therapy" or "treatment(s)," as described herein, includes all clinical management of a subject and interventions, whether biological, chemical, physical, or a combination thereof, intended to sustain, ameliorate, improve, or otherwise alter the condition of a subject. These terms may be used synonymously herein. Treatments include but are not limited to administration of prophylactics or therapeutic compounds (including conventional DMARDs, biologic DMARDs, non-steroidal anti -inflammatory drugs (NSAID's) such as COX-2 selective inhibitors, and corticosteroids), exercise regimens, physical therapy, dietary modification and/or supplementation, bariatric surgical intervention, administration of pharmaceuticals and/or anti-inflammatories (prescription or over-the-counter), and any other treatments known in the art as efficacious in preventing, delaying the onset of, or ameliorating disease. A "response to treatment" includes a subject's response to any of the above-described treatments, whether biological, chemical, physical, or a combination of the foregoing. A
"treatment course" relates to the dosage, duration, extent, etc. of a particular treatment or therapeutic regimen. An initial therapeutic regimen as used herein is the first line of treatment.
[0074] A "time point" as used herein refers to a manner of describing a time, which can be substantially described with a single point. A time point may also be described as a time range of a minimal unit which can be detected. A time point can refer to a state of the aspect of a time or a manner of description of a certain period of time. Such a time point or range can include, for example, an order of seconds, minutes to hours, or days.
Use of the present teachings in the diagnosis, prognosis, and assessment of disease
[0075] In some embodiments of the present teachings, biomarkers can be used in the derivation of a MBDA score, as described herein, which MBDA score can be used to provide diagnosis, prognosis and monitoring of disease state and/or disease activity in inflammatory disease and in autoimmune disease. In certain embodiments, the MBDA score can be used to provide diagnosis, prognosis and monitoring of disease state and/or disease activity of RA or early RA in response to therapy. In certain embodiments, the MBDA score can be used to recommend discontinuation of a therapeutic regimen, or the MBDA score can be used to recommend no change in a therapeutic regimen. In certain embodiments, the MBDA score can be used to predict disease activity, such as flare or remission, in a subject having an autoimmune disorder. In some embodiments, the optimal cutoff for flare, sustained clinical remission, and functional remission 6 months after discontinuation is an MBDA score of 30, 23, and 26, respectively. In other embodiments, the optimal cutoff for flare, sustained clinical remission, and functional remission 12 months after discontinuation is an MBDA score of 26, 25, and 27, respectively.
[0076] Identifying the state of inflammatory disease in a subject allows for a prognosis of the disease, and thus for the informed selection of, initiation of, adjustment of or increasing or decreasing various therapeutic regimens in order to delay, reduce or prevent that subject's progression to a more advanced disease state. In some embodiments, therefore, subjects can be identified as having a particular level of inflammatory disease activity and/or as being at a particular state of disease, or flare, based on the determination of their MBDA scores, and so can be selected to begin or accelerate treatment, as treatment is defined herein, to prevent or delay the further progression of inflammatory disease. In other embodiments, subjects that are identified via their MBDA scores as having a particular level of inflammatory disease activity, and/or as being at a particular state of inflammatory disease, can be selected to have their treatment decreased or discontinued, where improvement or remission in the subject is seen. In other embodiments, subjects that are identified via their MBDA scores as having a particular level of inflammatory disease activity, and/or as being at a particular state of inflammatory disease, can have therapy selected based on disease activity levels.
[0077] In regards to the need for early and accurate diagnosis of RA, recent advances in
RA treatment provide a means for more profound disease management and optimal treatment of RA within the first months of symptom onset, which in turn result in significantly improved outcomes. See F. Wolfe, Arth. Rheum. 2000, 43(12):2751-2761; M. Matucci-Cerinic, Clin. Exp. Rheum. 2002, 20(4):443-444; and, V. Nell et. al, Lancet 2005, 365(9455): 199-200.
Unfortunately, most subjects do not receive optimal treatment within this narrow window of opportunity, resulting in poorer outcomes and irreversible joint damage, in part because of the limits of current diagnostic laboratory tests. Numerous difficulties exist in diagnosing RA subject. This is in part because at their early stages, symptoms may not be fully differentiated. It is also because diagnostic tests for RA were developed based on phenomenological findings, not the biological basis of disease. In various embodiments of the present teachings, multi- biomarker algorithms can be derived from the disclosed set of biomarkers.
Rating disease activity
[0078] In some embodiments of the present teachings, the MBDA score, derived as described herein, can be used to rate inflammatory disease activity; e.g., as high, medium or low. The score can be varied based on a set of values chosen by the practitioner. For example, a score can be set such that a value is given a range from 0-100, and a difference between two scores would be a value of at least one point. The practitioner can then assign disease activity based on the values. For example, in some embodiments a score of about 1 to 29 represents a low level of disease activity, a score of about 30 to 44 represents a moderate level of disease activity, and a score of about 45 to 100 represents a high level of disease activity. In some embodiments on a scale of 1-100 a score of < 38 can represent a low or lower score, and a score of >38 can represent a high or higher score. In some embodiments on a scale of 1-100 a score of < 30 can represent a low or lower score, and a score of >30 can represent a high or higher score. In some embodiments, an MBDA scores of about <25 is remission, about 26-29 is low, about 30-44 is moderate, and about >44 is high. The cutoffs can vary. For example, in some embodiments a low score can be a score of <30, although for other utilities, a low score can be a score of <29 or <31.
[0079] The disease activity score can also change based on the range of the score. For example a score of 1 to 58 can represent a low level of disease activity when a range of 0-200 is utilized. Differences can be determined based on the range of score possibilities. For example, if using a score range of 0-100, a small difference in scores can be a difference of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 points; a moderate difference in scores can be a difference of about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 points; and large differences can be a change in about 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, or 50 points. Thus, by way of example, a practitioner can define a small difference in scores as about <6 points, a moderate difference in scores as about 7-20 points, and a large difference in scores as about >20 points. The difference can be expressed by any unit, for example, percentage points. For example, a practitioner can define a small difference as about <6 percentage points, moderate difference as about 7-20 percentage points, and a large difference as about >20 percentage points.
[0080] In some embodiments of the present teachings, autoimmune disease activity can be so rated. In other embodiments, RA disease activity can be so rated. Because the MBDA score correlates well with traditional clinical assessments of inflammatory disease activity, e.g. in RA, in other embodiments of the present teachings bone damage itself in a subject or population, and thus disease progression, can be tracked via the use and application of the MBDA score. [0081] The MBDA score can be used for several purposes. On a subject-specific basis, it provides a context for understanding the relative level of disease activity. The MBDA rating of disease activity can be used, e.g., to guide the clinician in determining treatment, in setting a treatment course, and/or to inform the clinician that the subject is in remission. Moreover, it provides a means to more accurately assess and document the qualitative level of disease activity in a subject. It is also useful from the perspective of assessing clinical differences among populations of subjects within a practice. For example, this tool can be used to assess the relative efficacy of different treatment modalities. Moreover, it is also useful from the perspective of assessing clinical differences among different practices. This would allow physicians to determine what global level of disease control is achieved by their colleagues, and/or for healthcare management groups to compare their results among different practices for both cost and comparative effectiveness. Because the MBDA score demonstrates strong association with established disease activity assessments, such as the DAS28, the MBDA score can provide a quantitative measure for monitoring the extent of subject disease activity, and response to treatment.
Subject screening
[0082] Certain embodiments of the present teachings can also be used to screen subject populations in any number of settings. For example, a health maintenance organization, public health entity or school health program can screen a group of subjects to identify those requiring interventions, as described above. Other embodiments of these teachings can be used to collect disease activity data on one or more populations of subjects, to identify subject disease status in the aggregate, in order to, e.g., determine the effectiveness of the clinical management of a population, or determine gaps in clinical management. Insurance companies (e.g., health, life, or disability) may request the screening of applicants in the process of determining coverage for possible intervention. Data collected in such population screens, particularly when tied to any clinical progression to conditions such as inflammatory disease and RA, will be of value in the operations of, for example, health maintenance organizations, public health programs and insurance companies. [0083] Such data arrays or collections can be stored in machine-readable media and used in any number of health-related data management systems to provide improved healthcare services, cost-effective healthcare, and improved insurance operation, among other things. See, e.g., U.S. Patent Application No. 2002/0038227; U.S. Patent Application No. 2004/0122296; U.S. Patent Application No. 2004/0122297; and U.S. Pat. No. 5,018,067. Such systems can access the data directly from internal data storage or remotely from one or more data storage sites as further detailed herein. Thus, in a health-related data management system, wherein it is important to manage inflammatory disease progression for a population in order to reduce disease-related employment productivity loss, disability and surgery, and thus reduce healthcare costs in the aggregate, various embodiments of the present teachings provide an improvement comprising the use of a data array encompassing the biomarker measurements as defined herein, and/or the resulting evaluation of disease status and activity from those biomarker
measurements.
Calculation of scores
[0084] In some embodiments of the present teachings, inflammatory disease activity in a subject is measured by: determining the levels in inflammatory disease subject serum of two or more biomarkers, then applying an interpretation function to transform the biomarker levels into a single MBDA score, which provides a quantitative measure of inflammatory disease activity in the subject, correlating well with traditional clinical assessments of inflammatory disease activity {e.g., a DAS28 or CDAI score in RA), as is demonstrated in the Examples below. In some embodiments, the disease activity so measured relates to an autoimmune disease. In some embodiments, the disease activity so measured relates to RA. The biomarkers can include apolipoprotein A-I (APOAl); apolipoprotein C-III (APOC3); calprotectin; chemokine (C-C motif) ligand 22 (CCL22); chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1, or YKL-40); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); intercellular adhesion molecule 1 (ICAMl); ICTP; interleukin 18 (interferon-gamma-inducing factor) (IL18); interleukin 1, beta (IL1B); interleukin 6 receptor (IL6R); interleukin 8 (IL8); keratan sulfate, or KS; leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase)
(MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); pyridinoline (crosslinks formed in collagen, derived from three lysine residues), which may be referred to herein as PYD; resistin (RETN); serum amyloid Al (SAA1); tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA). Selection of the biomarkers of the present invention is described in detail in US 2011/0137851. Calprotectin is a heteropolymer, comprising two protein subunits of gene symbols S100A8 and S100A9. ICTP is the carboxyterminal telopeptide region of type I collagen, and is liberated during the degradation of mature type I collagen. Type I collagen is present as fibers in tissue; in bone, the type I collagen molecules are crosslinked. The ICTP peptide is immunochemically intact in blood. (For the type I collagen gene, see official symbol COL1 Al, HUGO Gene Nomenclature Committee; also known as 014; alpha 1 type I collagen; collagen alpha 1 chain type I; collagen of skin, tendon and bone, alpha- 1 chain; and, pro-alpha-1 collagen type 1). Keratan sulfate (KS, or keratosulfate) is not the product of a discrete gene, but refers to any of several sulfated glycosaminoglycans. They are synthesized in the central nervous system, and are found especially in cartilage and bone. Keratan sulfates are large, highly hydrated molecules, which in joints can act as a cushion to absorb mechanical shock. In some embodiments, the biomarkers can comprise two, or three, or four, or five, or six, or seven, or eight, or nine, or ten, or eleven, or twelve of IL6, EGF, VEGFA, LEP, SAA1, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1. In some embodiments, the biomarkers can comprise at least four biomarkers that comprise IL6, EGF, SAA1, and CRP.
[0085] In some embodiments, the interpretation function is based on a predictive model.
Established statistical algorithms and methods well-known in the art, useful as models or useful in designing predictive models, can include but are not limited to: analysis of variants
(ANOVA); Bayesian networks; boosting and Ada-boosting; bootstrap aggregating (or bagging) algorithms; decision trees classification techniques, such as Classification and Regression Trees (CART), boosted CART, Random Forest (RF), Recursive Partitioning Trees (RPART), and others; Curds and Whey (CW); Curds and Whey -Lasso; dimension reduction methods, such as principal component analysis (PCA) and factor rotation or factor analysis; discriminant analysis, including Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELD A), and quadratic discriminant analysis; Discriminant Function Analysis (DFA); factor rotation or factor analysis; genetic algorithms; Hidden Markov Models; kernel based machine algorithms such as kernel density estimation, kernel partial least squares algorithms, kernel matching pursuit algorithms, kernel Fisher's discriminate analysis algorithms, and kernel principal components analysis algorithms; linear regression and generalized linear models, including or utilizing Forward Linear Stepwise Regression, Lasso (or LASSO) shrinkage and selection method, and Elastic Net regularization and selection method; glmnet (Lasso and Elastic Net-regularized generalized linear model); Logistic Regression (LogReg); meta-learner algorithms; nearest neighbor methods for classification or regression, e.g. Kth-nearest neighbor (KNN); non-linear regression or classification algorithms; neural networks; partial least square; rules based classifiers; shrunken centroids (SC); sliced inverse regression; Standard for the Exchange of Product model data, Application Interpreted Constructs (StepAIC); super principal component (SPC) regression; and, Support Vector Machines (SVM) and Recursive Support Vector Machines (RSVM), among others. Additionally, clustering algorithms as are known in the art can be useful in determining subject sub-groups.
[0086] Logistic Regression is the traditional predictive modeling method of choice for dichotomous response variables; e.g., treatment 1 versus treatment 2. It can be used to model both linear and non-linear aspects of the data variables and provides easily interpretable odds ratios.
[0087] Discriminant Function Analysis (DFA) uses a set of analytes as variables (roots) to discriminate between two or more naturally occurring groups. DFA is used to test analytes that are significantly different between groups. A forward step-wise DFA can be used to select a set of analytes that maximally discriminate among the groups studied. Specifically, at each step all variables can be reviewed to determine which will maximally discriminate among groups. This information is then included in a discriminative function, denoted a root, which is an equation consisting of linear combinations of analyte concentrations for the prediction of group membership. The discriminatory potential of the final equation can be observed as a line plot of the root values obtained for each group. This approach identifies groups of analytes whose changes in concentration levels can be used to delineate profiles, diagnose and assess therapeutic efficacy. The DFA model can also create an arbitrary score by which new subjects can be classified as either "healthy" or "diseased." To facilitate the use of this score for the medical community the score can be rescaled so a value of 0 indicates a healthy individual and scores greater than 0 indicate increasing disease activity. [0088] Classification and regression trees (CART) perform logical splits (if/then) of data to create a decision tree. All observations that fall in a given node are classified according to the most common outcome in that node. CART results are easily interpretable - one follows a series of if/then tree branches until a classification results.
[0089] Support vector machines (SVM) classify objects into two or more classes.
Examples of classes include sets of treatment alternatives, sets of diagnostic alternatives, or sets of prognostic alternatives. Each object is assigned to a class based on its similarity to (or distance from) objects in the training data set in which the correct class assignment of each object is known. The measure of similarity of a new object to the known objects is determined using support vectors, which define a region in a potentially high dimensional space (>R6).
[0090] The process of bootstrap aggregating, or "bagging," is computationally simple. In the first step, a given dataset is randomly resampled a specified number of times (e.g., thousands), effectively providing that number of new datasets, which are referred to as
"bootstrapped resamples" of data, each of which can then be used to build a model. Then, in the example of classification models, the class of every new observation is predicted by the number of classification models created in the first step. The final class decision is based upon a "majority vote" of the classification models; i.e., a final classification call is determined by counting the number of times a new observation is classified into a given group, and taking the majority classification (33%+ for a three-class system). In the example of logistical regression models, if a logistical regression is bagged 1000 times, there will be 1000 logistical models, and each will provide the probability of a sample belonging to class 1 or 2.
[0091] Curds and Whey (CW) using ordinary least squares (OLS) is another predictive modeling method. See L. Breiman and JH Friedman, J. Royal. Stat. Soc. B 1997, 59(l):3-54. This method takes advantage of the correlations between response variables to improve predictive accuracy, compared with the usual procedure of performing an individual regression of each response variable on the common set of predictor variables X. In CW, Y = XB * S, where Y = (ykj ) with k for the kth patient and j for jth response (j =1 for TJC, j = 2 for SJC, etc.), B is obtained using OLS, and S is the shrinkage matrix computed from the canonical coordinate system. Another method is Curds and Whey and Lasso in combination (CW-Lasso). Instead of using OLS to obtain B, as in CW, here Lasso is used, and parameters are adjusted accordingly for the Lasso approach.
[0092] Many of these techniques are useful either combined with a biomarker selection technique (such as, for example, forward selection, backwards selection, or stepwise selection), or for complete enumeration of all potential panels of a given size, or genetic algorithms, or they can themselves include biomarker selection methodologies in their own techniques. These techniques can be coupled with information criteria, such as Akaike's Information Criterion (AIC), Bayes Information Criterion (BIC), or cross-validation, to quantify the tradeoff between the inclusion of additional biomarkers and model improvement, and to minimize overfit. The resulting predictive models can be validated in other studies, or cross-validated in the study they were originally trained in, using such techniques as, for example, Leave-One-Out (LOO) and 10- Fold cross-validation (10-Fold CV).
[0093] One example of an interpretation function that provides a MBDA score, derived from a statistical modeling method as described above, is given by the following function:
MBDA = b0 + b1 * DAIMRK X - b2 * DAIMRK2 x - b3 * DAIMRK3 x * DAIMRK„ x; where MBDA is the MBDA score, b0-n are constants, and DAIMRKi.n x are the serum
concentrations to the xth power of n different biomarkers selected from the biomarkers disclosed herein. MBDA scores thus obtained for RA subjects with known clinical assessments {e.g., DAS28 scores) can then be compared to those known assessments to determine the level of correlation between the two assessments, and hence determine the accuracy of the MBDA score and its underlying predictive model. See Examples below for specific formulas and constants.
[0094] More generally, the function can be described as:
MBDA = ~F(DAIMRK X , DAIMRK2 X, DAIMRKn x) where MBDA is the MBDA score, F is the function, and DAIMRK].n x are the serum concentrations to the Xth power of n different biomarkers selected from the biomarkers disclosed herein. The function is described in the following paragraph. [0095] An interpretation function for providing a MBDA score can also be derived based on models built to predict components of a disease activity assessment, such as DAS28-CRP, rather than predicting disease activity entirely. See Example 1. An example of such a function is given by the following, wherein biomarkers are used to provide improved predicted components of the DAS score:
MBDA score = ((0.56 * sqrt(IPTJC)) + (0.28 * sqrt(IPSJC)) + (O. J 4 * PPGA) + (0.36 * ln(CRP/106 + l))+0.96) * 10.53 + 1;
IPTJC = Improved PTJC = max(0.1739 * PTJC + 0. 7865 * PSJC, 0); IPSJC = Improved PSJC = max(0.1734 * PTJC + 0.7839 * PSJC, 0);
PTJC = Prediction of Tender Joint Count = -38.564 + 3.997 * (SAA1) 1/10 + 17.331 * (IL6) 1/10 + 4.665 * (CHI3L1) 1/10 - 15.236 * (EOF) 1/10 + 2.651 * (TNFRSF1A) 1/10 + 2.641 * (LEP) 1/10 + 4.026 * (VEGFA) 1/10 - 1.47 * (VCAMl) 1/w;
PSJC = Prediction of Swollen Joint Count = -25.444 + 4.051 * (SAA1) 1/10 + 16.154 * (IL6) 1/10 - 11.847 * (EGF) 1/10 +3.091 * (CHI3L1) 1/10 + 0.353 * (TNFRSF1A) M0;
PPGA = Prediction of Patient Global Assessment = -13.489 + 5.474 * (IL6) 1/10 + 0.486 * (SAA1) 1/10 + 2.246 * (MMP1) 1/10 + 1.684 * (leptin) 1/10 + 4.14 * (TNFRSF1A) 1/10 + 2.292 * (VEGFA) 1/10 - 1.898 * (EGF) 1/10 + 0.028 * (MMP3) 1/10 - 2.892 * (VCAMl) 1/10-.506*(RETN) 1/10 in which serum levels x for all biomarkers but CRP are transformed as xul °, units for all biomarkers are in pg/mL, and In is natural log, or loge.
[0096] Where CRP units are obtained in mg/L and other markers are pg/mL, MBDA score = ((0.56 * sqrt(IPTJC)) + (0.28 * sqrt(IPSJC)) + (0.14 * (PPGA)) + (0.36 * ln(CRP + 1)) + 0.96) * 10.53 + 1.
[0097] The MBDA score can be further rounded and capped, in order to provide a whole number between 1 and 100, the scaled MBDA score. To accomplish this, the immediately preceding function can be re-written: scaled MBDA score = round(max(min((0.56 * sqrt(IPTJC) + (0.28 * sqrt(IPSJC)) +(0 .14 * (PPGA)) + (0.36 * ln(CRP + 1) + 0.96) * 10.53 + 1, 100), 1)). Biomarker gene names provided in the above formulas represent the concentrations of those markers, and will depend on the types of assays used.
[0098] In some embodiments of the present teachings, it is not required that the MBDA score be compared to any pre-determined "reference," "normal," "control," "standard,"
"healthy," "pre-disease" or other like index, in order for the MBDA score to provide a quantitative measure of inflammatory disease activity in the subject.
[0099] In other embodiments of the present teachings, the amount of the biomarker(s) can be measured in a sample and used to derive a MBDA score, which MBDA score is then compared to a "normal" or "control" level or value, utilizing techniques such as, e.g., reference or discrimination limits or risk defining thresholds, in order to define cut-off points and/or abnormal values for inflammatory disease. The normal level then is the level of one or more biomarkers or combined biomarker indices typically found in a subject who is not suffering from the inflammatory disease under evaluation. Other terms for "normal" or "control" are, e.g., "reference," "index," "baseline," "standard," "healthy," "pre-disease," etc. Such normal levels can vary, based on whether a biomarker is used alone or in a formula combined with other biomarkers to output a score. Alternatively, the normal level can be a database of biomarker patterns from previously tested subjects who did not convert to the inflammatory disease under evaluation over a clinically relevant time period. Reference (normal, control) values can also be derived from, e.g., a control subject or population whose inflammatory disease activity level or state is known. In some embodiments of the present teachings, the reference value can be derived from one or more subjects who have been exposed to treatment for inflammatory disease, or from one or more subjects who are at low risk of developing inflammatory disease, or from subjects who have shown improvements in inflammatory disease activity factors (such as, e.g., clinical parameters as defined herein) as a result of exposure to treatment. In some embodiments the reference value can be derived from one or more subjects who have not been exposed to treatment; for example, samples can be collected from (a) subjects who have received initial treatment for inflammatory disease, and (b) subjects who have received subsequent treatment for inflammatory disease, to monitor the progress of the treatment. A reference value can also be derived from disease activity algorithms or computed indices from population studies.
Measurement of biomarkers
[00100] The quantity of one or more biomarkers of the present teachings can be indicated as a value. The value can be one or more numerical values resulting from the evaluation of a sample, and can be derived, e.g., by measuring level(s) of the biomarker(s) in a sample by an assay performed in a laboratory, or from dataset obtained from a provider such as a laboratory, or from a dataset stored on a server. Biomarker levels can be measured using any of several techniques known in the art. The present teachings encompass such techniques, and further include all subject fasting and/or temporal-based sampling procedures for measuring biomarkers.
[00101] The actual measurement of levels of the biomarkers can be determined at the protein or nucleic acid level using any method known in the art. "Protein" detection comprises detection of full-length proteins, mature proteins, pre-proteins, polypeptides, isoforms, mutations, variants, post-translationally modified proteins and variants thereof, and can be detected in any suitable manner. Levels of biomarkers can be determined at the protein level, e.g., by measuring the serum levels of peptides encoded by the gene products described herein, or by measuring the enzymatic activities of these protein biomarkers. Such methods are well- known in the art and include, e.g., immunoassays based on antibodies to proteins encoded by the genes, aptamers or molecular imprints. Any biological material can be used for the
detection/quantification of the protein or its activity. Alternatively, a suitable method can be selected to determine the activity of proteins encoded by the biomarker genes according to the activity of each protein analyzed. For biomarker proteins, polypeptides, isoforms, mutations, and variants thereof known to have enzymatic activity, the activities can be determined in vitro using enzyme assays known in the art. Such assays include, without limitation, protease assays, kinase assays, phosphatase assays, reductase assays, among many others. Modulation of the kinetics of enzyme activities can be determined by measuring the rate constant KM using known algorithms, such as the Hill plot, Michaelis-Menten equation, linear regression plots such as Lineweaver-Burk analysis, and Scatchard plot. [00102] Using sequence information provided by the public database entries for the biomarker, expression of the biomarker can be detected and measured using techniques well- known to those of skill in the art. For example, nucleic acid sequences in the sequence databases that correspond to nucleic acids of biomarkers can be used to construct primers and probes for detecting and/or measuring biomarker nucleic acids. These probes can be used in, e.g., Northern or Southern blot hybridization analyses, ribonuclease protection assays, and/or methods that quantitatively amplify specific nucleic acid sequences. As another example, sequences from sequence databases can be used to construct primers for specifically amplifying biomarker sequences in, e.g., amplification-based detection and quantitation methods such as reverse- transcription based polymerase chain reaction (RT-PCR) and PCR. When alterations in gene expression are associated with gene amplification, nucleotide deletions, polymorphisms, post- translational modifications and/or mutations, sequence comparisons in test and reference populations can be made by comparing relative amounts of the examined DNA sequences in the test and reference populations.
[00103] As an example, Northern hybridization analysis using probes which specifically recognize one or more of these sequences can be used to determine gene expression.
Alternatively, expression can be measured using RT-PCR; e.g., polynucleotide primers specific for the differentially expressed biomarker mRNA sequences reverse-transcribe the mRNA into DNA, which is then amplified in PCR and can be visualized and quantified. Biomarker RNA can also be quantified using, for example, other target amplification methods, such as TMA, SDA, and NASBA, or signal amplification methods (e.g., bDNA), and the like. Ribonuclease protection assays can also be used, using probes that specifically recognize one or more biomarker mRNA sequences, to determine gene expression.
[00104] Alternatively, biomarker protein and nucleic acid metabolites can be measured. The term "metabolite" includes any chemical or biochemical product of a metabolic process, such as any compound produced by the processing, cleavage or consumption of a biological molecule (e.g., a protein, nucleic acid, carbohydrate, or lipid). Metabolites can be detected in a variety of ways known to one of skill in the art, including the refractive index spectroscopy (RI), ultra-violet spectroscopy (UV), fluorescence analysis, radiochemical analysis, near-infrared spectroscopy (near-IR), nuclear magnetic resonance spectroscopy (NMR), light scattering analysis (LS), mass spectrometry, pyrolysis mass spectrometry, nephelometry, dispersive Raman spectroscopy, gas chromatography combined with mass spectrometry, liquid chromatography combined with mass spectrometry, matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) combined with mass spectrometry, ion spray spectroscopy combined with mass spectrometry, capillary electrophoresis, MR and IR detection. See WO 04/056456 and WO 04/088309, each of which is hereby incorporated by reference in its entirety. In this regard, other biomarker analytes can be measured using the above-mentioned detection methods, or other methods known to the skilled artisan. For example, circulating calcium ions (Ca2+) can be detected in a sample using fluorescent dyes such as the Fluo series, Fura-2A, Rhod-2, among others. Other biomarker metabolites can be similarly detected using reagents that are specifically designed or tailored to detect such metabolites.
[00105] In some embodiments, a biomarker is detected by contacting a subject sample with reagents, generating complexes of reagent and analyte, and detecting the complexes.
Examples of "reagents" include but are not limited to nucleic acid primers and antibodies.
[00106] In some embodiments of the present teachings an antibody binding assay is used to detect a biomarker; e.g., a sample from the subject is contacted with an antibody reagent that binds the biomarker analyte, a reaction product (or complex) comprising the antibody reagent and analyte is generated, and the presence (or absence) or amount of the complex is determined. The antibody reagent useful in detecting biomarker analytes can be monoclonal, polyclonal, chimeric, recombinant, or a fragment of the foregoing, as discussed in detail above, and the step of detecting the reaction product can be carried out with any suitable immunoassay. The sample from the subject is typically a biological fluid as described above, and can be the same sample of biological fluid as is used to conduct the method described above.
[00107] Immunoassays carried out in accordance with the present teachings can be homogeneous assays or heterogeneous assays. Immunoassays carried out in accordance with the present teachings can be multiplexed. In a homogeneous assay the immunological reaction can involve the specific antibody {e.g., anti -biomarker protein antibody), a labeled analyte, and the sample of interest. The label produces a signal, and the signal arising from the label becomes modified, directly or indirectly, upon binding of the labeled analyte to the antibody. Both the immunological reaction of binding, and detection of the extent of binding, can be carried out in a homogeneous solution. Immunochemical labels which can be employed include but are not limited to free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, and coenzymes. Immunoassays include competition assays.
[00108] In a heterogeneous assay approach, the reagents can be the sample of interest, an antibody, and a reagent for producing a detectable signal. Samples as described above can be used. The antibody can be immobilized on a support, such as a bead (such as protein A and protein G agarose beads), plate or slide, and contacted with the sample suspected of containing the biomarker in liquid phase. The support is separated from the liquid phase, and either the support phase or the liquid phase is examined using methods known in the art for detecting signal. The signal is related to the presence of the analyte in the sample. Methods for producing a detectable signal include but are not limited to the use of radioactive labels, fluorescent labels, or enzyme labels. For example, if the antigen to be detected contains a second binding site, an antibody which binds to that site can be conjugated to a detectable (signal-generating) group and added to the liquid phase reaction solution before the separation step. The presence of the detectable group on the solid support indicates the presence of the biomarker in the test sample. Examples of suitable immunoassays include but are not limited to oligonucleotides,
immunoblotting, immunoprecipitation, immunofluorescence methods, chemiluminescence methods, electrochemiluminescence (ECL), and/or enzyme-linked immunoassays (ELISA).
[00109] Those skilled in the art will be familiar with numerous specific immunoassay formats and variations thereof which can be useful for carrying out the method disclosed herein. See, e.g., E. Maggio, Enzyme -Immunoassay (1980), CRC Press, Inc., Boca Raton, FL. See also U.S. Pat. No. 4,727,022 to C. Skold et al, titled "Novel Methods for Modulating Ligand- Receptor Interactions and their Application"; U.S. Pat. No. 4,659,678 to GC Forrest et a/., titled "Immunoassay of Antigens"; U.S. Pat. No. 4,376,110 to GS David et a/., titled "Immunometric Assays Using Monoclonal Antibodies"; U.S. Pat. No. 4,275,149 to D. Litman et al, titled "Macromolecular Environment Control in Specific Receptor Assays"; U.S. Pat. No. 4,233,402 to E. Maggio et al, titled "Reagents and Method Employing Channeling"; and, U.S. Pat. No. 4,230,797 to R. Boguslaski et a/., titled "Heterogenous Specific Binding Assay Employing a Coenzyme as Label." [00110] Antibodies can be conjugated to a solid support suitable for a diagnostic assay (e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding. Antibodies as described herein can likewise be conjugated to detectable labels or groups such as radiolabels (e.g., 35S, 1251, 1311), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein, Alexa, green fluorescent protein, rhodamine) in accordance with known techniques.
[00111] Antibodies may also be useful for detecting post-translational modifications of biomarkers. Examples of post-translational modifications include, but are not limited to tyrosine phosphorylation, threonine phosphorylation, serine phosphorylation, citrullination and glycosylation (e.g., O-GlcNAc). Such antibodies specifically detect the phosphorylated amino acids in a protein or proteins of interest, and can be used in the immunoblotting,
immunofluorescence, and ELISA assays described herein. These antibodies are well-known to those skilled in the art, and commercially available. Post-translational modifications can also be determined using metastable ions in reflector matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF). See U. Wirth et al, Proteomics 2002, 2(10): 1445- 1451.
Therapeutic regimens
[00112] The present invention provides methods of recommending therapeutic regimens, including withdrawal from therapeutic regiments, following the determination of differences in expression of the biomarkers disclosed herein. Measuring scores derived from expression levels of the biomarkers disclosed herein over a period time can provide a clinician with a dynamic picture of a subject's biological state. These embodiments of the present teachings thus will provide subject-specific biological information, which will be informative for therapy decision and will facilitate therapy response monitoring, and should result in more rapid and more optimized treatment, better control of disease activity, and an increase in the proportion of subjects achieving remission.
[00113] Treatment strategies for autoimmune disorders are confounded by the fact that some autoimmune disorders, such as RA, is a classification given to a group of subjects with a diverse array of related symptoms that can flare or go into remission. This suggests that certain subtypes of RA are driven by specific cell type or cytokine. As a likely consequence, no single therapy has proven optimal for treatment. Given the increasing numbers of therapeutic options available for RA, the need for an individually tailored treatment directed by immunological prognostic factors of treatment outcome is imperative. In various embodiments of the present teachings, a biomarker-derived algorithm can be used to quantify therapy response in RA subjects. For patients with early RA (eRA), methotrexate (MTX) is sometimes recommended as a first-line treatment and in non-responders both the addition of conventional non-biological disease modifying anti -rheumatic drug therapy (triple DMARD therapy) and of biological (anti- T F) therapy are supported by data. Identification of patients with a higher likelihood of responding to one or the other of these options would lead to more personalized medicine and increased effectiveness of therapy, which is a primary objective of this invention.
Reference Standards for Treatment
[00114] In many embodiments, the levels of one or more analyte biomarkers or the levels of a specific panel of analyte biomarkers in a sample are compared to a reference standard
("reference standard" or "reference level") in order to direct treatment decisions. Expression levels of the one or more biomarkers can be combined into a score, which can represent disease activity. The reference standard used for any embodiment disclosed herein may comprise average, mean, or median levels of the one or more analyte biomarkers or the levels of the specific panel of analyte biomarkers in a control population. The reference standard may further include an earlier time point for the same subject. For example, a reference standard may include a first time point, and the levels of the one or more analyte biomarkers can be examined again at second, third, fourth, fifth, sixth time points, etc. Any time point earlier than any particular time point can be considered a reference standard. The reference standard may additionally comprise cutoff values or any other statistical attribute of the control population, or earlier time points of the same subject, such as a standard deviation from the mean levels of the one or more analyte biomarkers or the levels of the specific panel of analyte biomarkers. In some embodiments, the control population may comprise healthy individuals or the same subject prior to the administration of any therapy. [00115] In some embodiments, a score may be obtained from the reference time point, and a different score may be obtained from a later time point. A first time point can be when an initial therapeutic regimen is begun. A first time point can also be when a first immunoassay is performed. A time point can be hours, days, months, years, etc. In some embodiments, a time point is one month. In some embodiments, a time point is two months. In some embodiments, a time point is three months. In some embodiments, a time point is four months. In some embodiments, a time point is five months. In some embodiments, a time point is six months. In some embodiments, a time point is seven months. In some embodiments, a time point is eight months. In some embodiments, a time point is nine months. In some embodiments, a time point is ten months. In some embodiments, a time point is eleven months. In some embodiments, a time point is twelve months. In some embodiments, a time point is two years. In some embodiments, a time point is three years. In some embodiments, a time point is four years. In some embodiments, a time point is five years. In some embodiments, a time point is ten years.
[00116] A difference in the score can be interpreted as a decrease in disease activity. For example, lower score can indicate a lower level of disease activity, or remission. In these circumstances a second score having a lower score than the reference score, or first score, means that the subject's disease activity has been lowered (improved) between the first and second time periods, or is in remission. Alternatively, a higher score can indicate a lower level of disease activity, or remission. In these circumstances, a second score having a higher score than the reference score, or first score, also means that the subject's disease activity has improved between the first and second time periods, or is in remission.
[00117] A difference in the score can also be interpreted as an increase in disease activity. For example, lower score can indicate a higher level of disease activity, or flare. In these
circumstances a second score having a lower score than the reference score, or first score, means that the subject's disease activity has been increased (worsened) between the first and second time periods. Alternatively, a higher score can indicate a higher level of disease activity, or flare. In these circumstances, a second score having a higher score than the reference score, or first score, also means that the subject's disease activity has worsened between the first and second time periods, or is flaring. In some embodiments, the optimal cutoff for flare, sustained clinical remission, and functional remission 6 months after discontinuation is an MBDA score of 30, 23, and 26, respectively. In other embodiments, the optimal cutoff for flare, sustained clinical remission, and functional remission 12 months after discontinuation is an MBDA score of 26, 25, and 27, respectively.
[00118] The differences can be variable. For example, when a difference in the score is interpreted as a decrease in disease activity, a large difference can mean a greater decrease in disease activity than a lower or moderate difference. Alternatively, when a difference in the score is interpreted as an increase in disease activity, a large difference can mean a greater increase in disease activity than a lower or moderate difference.
Reference Therapy for Treatment
[00119] In some embodiments, a patient is treated more or less aggressively than a reference therapy based on the difference of scores. A reference therapy is any therapy that is the standard of care for the autoimmune disorder. The standard of care can vary temporally and
geographically, and a skilled person can easily determine the appropriate standard of care by consulting the relevant medical literature.
[00120] In some embodiments, a more aggressive therapy than the standard therapy comprises beginning treatment earlier than in the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises administering additional treatments than in the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises treating on an accelerated schedule compared to the standard therapy. In some embodiments, a more aggressive therapy than the standard therapy comprises administering additional treatments not called for in the standard therapy.
[00121] In some embodiments, a less aggressive therapy than the standard therapy comprises delaying treatment relative to the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering less treatment than in the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering treatment on a decelerated schedule compared to the standard therapy. In some embodiments, a less aggressive therapy than the standard therapy comprises administering no treatment. Therapy withdrawal
[00122] In some embodiments, prediction of autoimmune disease patients, in particular RA patients, who can successfully withdrawal from or discontinue therapy, can be based on an MBDA score. The therapy to be considered for withdrawal can be any therapy as described herein.
[00123] In some embodiments, a high MBDA score as described herein at baseline can be an independent predictor of disease progression within a certain period of time following discontinuation of therapy. In some embodiments, a moderate MBDA score as described herein at baseline can be an independent predictor of disease progression within a certain period of time following discontinuation of therapy. In some embodiments, a low MBDA score as described herein at baseline can be an independent predictor of disease progression, or remission, within a certain period of time following discontinuation of therapy.
[00124] In some embodiments, the disease progression for therapy withdrawal is flare. In some embodiments, the disease progression for therapy withdrawal is radiographic progression. In other embodiments, the disease progression for therapy withdrawal is any other consequence of autoimmune diseases, in particular RA, known in the art.
[00125] The period of time in which an MBDA score can predict disease progression can be one, two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve months. The period of time in which an MBDA score can predict disease progression can also be one, two, three, four, five, ten, fifteen, twenty, twenty -five, or more years.
[00126] In some embodiments, a low MBDA score as described herein can result in the recommendation that therapy is withdrawn or discontinued. In some embodiments, a low or moderate MBDA score as described herein can result in the recommendation that therapy is withdrawn or discontinued. In some embodiments, a moderate or high MBDA score as described herein can result in the recommendation that therapy is not withdrawn or discontinued. In some embodiments, a high MBDA score as described herein can result in the recommendation that therapy is not withdrawn or discontinued. [00127] In some embodiments, the therapy can be partially withdrawn. In some embodiments, the therapy can be withdrawn partially in relation to the MBDA score. For purposes of illustration, a low MBDA score as described herein can result in the
recommendation that therapy is completely withdrawn; whereas a moderate MBDA score as described herein can result in the recommendation that therapy is reduced, for example, a lower dose can be recommended. Thus, in some embodiments, dose can be adjusted based on the MBDA score.
Treatment of autoimmune disorders
[00128] In one embodiment, the practitioner discontinues a therapy regimen if a score is low. In one embodiment, the practitioner does not change the therapy regimen if the score is high. In one embodiment, the practitioner adjusts the therapy based on a comparison between difference scores, or based on an initial predictive score. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different combination of drugs. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage. In one embodiment, the practitioner adjusts the therapy by adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting drug dosage. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug combination and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage and dose schedule. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting dose schedule and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, and adjusting dose schedule. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting dose schedule, and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by adjusting drug dosage, adjusting dose schedule, and adjusting length of therapy. In one embodiment, the practitioner adjusts the therapy by selecting and administering a different drug, adjusting drug dosage, adjusting dose schedule, and adjusting length of therapy.
[00129] In one embodiment a less aggressive therapy comprises no change in the therapy regimen. In one embodiment a less aggressive therapy comprises delaying treatment. In one embodiment a less aggressive therapy comprises selecting and administering less potent drugs. In one embodiment a less aggressive therapy comprises decreasing the frequency treatment. In one embodiment a less aggressive therapy comprises shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and decreasing drug dosage. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and decelerating dose schedule. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs and shortening length of therapy. In one embodiment, less aggressive therapy comprises decreasing drug dosage and decelerating dose schedule. In one embodiment, less aggressive therapy comprises decreasing drug dosage and shortening length of therapy. In one embodiment, less aggressive therapy comprises decelerating dose schedule and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, and decelerating dose schedule. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decelerating dose schedule, and shortening length of therapy. In one embodiment, less aggressive therapy comprises decreasing drug dosage, decelerating dose schedule, and shortening length of therapy. In one embodiment, less aggressive therapy comprises selecting and administering less potent drugs, decreasing drug dosage, decelerating dose schedule, and shortening length of therapy. In some embodiments, a less aggressive therapy comprises administering only non-drug-based therapies.
[00130] In another aspect of the present application, treatment comprises a more aggressive therapy than a reference therapy. In one embodiment a more aggressive therapy comprises increased length of therapy. In one embodiment a more aggressive therapy comprises increased frequency of the dose schedule. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs and increasing drug dosage. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs and accelerating dose schedule. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs and increasing length of therapy. In one embodiment, more aggressive therapy comprises increasing drug dosage and accelerating dose schedule. In one embodiment, more aggressive therapy comprises increasing drug dosage and increasing length of therapy. In one embodiment, more aggressive therapy comprises accelerating dose schedule and increasing length of therapy. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, and accelerating dose schedule. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, and increasing length of therapy. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, accelerating dose schedule, and increasing length of therapy. In one embodiment, more aggressive therapy comprises increasing drug dosage, accelerating dose schedule, and increasing length of therapy. In one embodiment, more aggressive therapy comprises selecting and administering more potent drugs, increasing drug dosage, accelerating dose schedule, and increasing length of therapy. In some embodiments, a more aggressive therapy comprises administering a combination of drug- based and non-drug-based therapies.
[00131] Therapies can be conventional or biologic. Examples of therapies, such as disease modifying anti -rheumatic drugs (DMARD) that are generally considered conventional include, but are not limited to, MTX, azathioprine (AZA), bucillamine (BUC), chloroquine (CQ), ciclosporin (CSA, or cyclosporine, or cyclosporin), doxycycline (DOXY), hydroxychloroquine (HCQ), intramuscular gold (EVI gold), leflunomide (LEF), levofloxacin (LEV), and sulfasalazine (SSZ). Examples of other conventional therapies include, but are not limited to, folinic acid, D- pencillamine, gold auranofin, gold aurothioglucose, gold thiomalate, cyclophosphamide, and chlorambucil. Examples of biologic drugs can include but are not limited to biological agents that target the tumor necrosis factor (T F)-alpha molecules and the TNF inhibitors, such as infliximab, adalimumab, etanercept and golimumab. Other classes of biologic drugs include ILl inhibitors such as anakinra, T-cell modulators such as abatacept, B-cell modulators such as rituximab, and IL6 inhibitors such as tocilizumab. [00132] To identify additional therapeutics or drugs that are appropriate for a specific subject, a test sample from the subject can also be exposed to a therapeutic agent or a drug, and the level of one or more biomarkers can be determined. The level of one or more biomarkers can be compared to sample derived from the subject before and after treatment or exposure to a therapeutic agent or a drug, or can be compared to samples derived from one or more subjects who have shown improvements in inflammatory disease state or activity (e.g., clinical parameters or traditional laboratory risk factors) as a result of such treatment or exposure.
Clinical assessments of the present teachings
[00133] In some embodiments of the present teachings, MBDA scores are tailored to the population, endpoints or clinical assessment, and/or use that is intended. For example, a MBDA score can be used to assess subjects for primary prevention and diagnosis, and for secondary prevention and management. For the primary assessment, the MBDA score can be used for prediction and risk stratification for future conditions or disease sequelae, for the diagnosis of inflammatory disease, for the prognosis of disease activity and rate of change, and for indications for future diagnosis and therapeutic regimens. For secondary prevention and clinical
management, the MBDA score can be used for prognosis and risk stratification. The MBDA score can be used for clinical decision support, such as determining whether to defer intervention or treatment, to recommend preventive check-ups for at-risk patients, to recommend increased visit frequency, to recommend increased testing, and to recommend intervention. The MBDA score can also be useful for therapeutic selection, determining response to treatment, adjustment and dosing of treatment, monitoring ongoing therapeutic efficiency, and indication for change in therapeutic regimen.
[00134] In some embodiments of the present teachings, the MBDA score can be used to aid in the diagnosis of inflammatory disease, and in the determination of the severity of inflammatory disease. The MBDA score can also be used for determining the future status of intervention such as, for example in RA, determining the prognosis of future joint erosion with or without treatment. Certain embodiments of the present teachings can be tailored to a specific treatment or a combination of treatments. X-ray is currently considered the gold standard for assessment of disease progression, but it has limited capabilities since subjects may have long periods of active symptomatic disease while radiographs remain normal or show only nonspecific changes. Conversely, subjects who seem to have quiescent disease (subclinical disease) may slowly progress over time, undetected clinically until significant radiographic progression has occurred. If subjects with a high likelihood of disease progression could be identified in advance, the opportunity for early aggressive treatment could result in much more effective disease outcomes. See, e.g., M. Weinblatt et al, N. Engl. J. Med. 1999, 340:253-259.
Systems for implementing disease activity tests
[00135] Tests for measuring disease activity according to various embodiments of the present teachings can be implemented on a variety of systems typically used for obtaining test results, such as results from immunological or nucleic acid detection assays. Such systems may comprise modules that automate sample preparation, that automate testing such as measuring biomarker levels, that facilitate testing of multiple samples, and/or are programmed to assay the same test or different tests on each sample. In some embodiments, the testing system comprises one or more of a sample preparation module, a clinical chemistry module, and an immunoassay module on one platform. Testing systems are typically designed such that they also comprise modules to collect, store, and track results, such as by connecting to and utilizing a database residing on hardware. Examples of these modules include physical and electronic data storage devices as are well-known in the art, such as a hard drive, flash memory, and magnetic tape. Test systems also generally comprise a module for reporting and/or visualizing results. Some examples of reporting modules include a visible display or graphical user interface, links to a database, a printer, etc. See section Machine-readable storage medium, below.
[00136] One embodiment of the present invention comprises a system for determining the inflammatory disease activity of a subject. In some embodiments, the system employs a module for applying a formula to an input comprising the measured levels of biomarkers in a panel, as described herein, and outputting a score. In some embodiments, the measured biomarker levels are test results, which serve as inputs to a computer that is programmed to apply the formula. The system may comprise other inputs in addition to or in combination with biomarker results in order to derive an output score; e.g., one or more clinical parameters such as therapeutic regimen, TJC, SJC, morning stiffness, arthritis of three or more joint areas, arthritis of hand joints, symmetric arthritis, rheumatoid nodules, radiographic changes and other imaging, gender/sex, age, race/ethnicity, disease duration, height, weight, body-mass index, family history, CCP status, RF status, ESR, smoker/non-smoker, etc. In some embodiments the system can apply a formula to biomarker level inputs, and then output a disease activity score that can then be analyzed in conjunction with other inputs such as other clinical parameters. In other embodiments, the system is designed to apply a formula to the biomarker and non-biomarker inputs (such as clinical parameters) together, and then report a composite output disease activity index.
[00137] A number of testing systems are presently available that could be used to implement various embodiments of the present teachings. See, for example, the ARCHITECT series of integrated immunochemistry systems - high-throughput, automated, clinical chemistry analyzers (ARCHITECT is a registered trademark of Abbott Laboratories, Abbott Park, 111. 60064). See C. Wilson et al., "Clinical Chemistry Analyzer Sub-System Level Performance," American Association for Clinical Chemistry Annual Meeting, Chicago, 111., Jul. 23-27, 2006; and, HJ Kisner, "Product development: the making of the Abbott ARCHITECT," Clin. Lab. Manage. Rev. 1997 Nov.-Dec, 11(6):419-21; A. Ognibene et al., "A new modular
chemiluminescence immunoassay analyzer evaluated," Clin. Chem. Lab. Med. 2000 March, 38(3):251-60; JW Park et al., "Three-year experience in using total laboratory automation system," Southeast Asian J. Trop. Med. Public Health 2002, 33 Suppl 2:68-73; D. Pauli et al., "The Abbott Architect c8000: analytical performance and productivity characteristics of a new analyzer applied to general chemistry testing," Clin. Lab. 2005, 5 l(l-2):31-41.
[00138] Another testing system useful for embodiments of the present teachings is the
VITROS system (VITROS is a registered trademark of Johnson & Johnson Corp., New
Brunswick, NJ) - an apparatus for chemistry analysis that is used to generate test results from blood and other body fluids for laboratories and clinics. Another testing system is the
DIMENSION system (DIMENSION is a registered trademark of Dade Behring Inc., Deerfield 111.) - a system for the analysis of body fluids, comprising computer software and hardware for operating the analyzers, and analyzing the data generated by the analyzers. [00139] The testing required for various embodiments of the present teachings, e.g.
measuring biomarker levels, can be performed by laboratories such as those certified under the Clinical Laboratory Improvement Amendments (42 U.S.C. Section 263(a)), or by laboratories certified under any other federal or state law, or the law of any other country, state or province that governs the operation of laboratories that analyze samples for clinical purposes. Such laboratories include, for example, Laboratory Corporation of America, 358 South Main Street, Burlington, NC 27215 (corporate headquarters); Quest Diagnostics, 3 Giralda Farms, Madison, NJ 07940 (corporate headquarters); and other reference and clinical chemistry laboratories.
Kits
[00140] Other embodiments of the present teachings comprise biomarker detection reagents packaged together in the form of a kit for conducting any of the assays of the present teachings. In certain embodiments, the kits comprise oligonucleotides that specifically identify one or more biomarker nucleic acids based on homology and/or complementarity with biomarker nucleic acids. The oligonucleotide sequences may correspond to fragments of the biomarker nucleic acids. For example, the oligonucleotides can be more than 200, 200, 150, 100, 50, 25, 10, or fewer than 10 nucleotides in length. In other embodiments, the kits comprise antibodies to proteins encoded by the biomarker nucleic acids. The kits of the present teachings can also comprise aptamers. The kit can contain in separate containers a nucleic acid or antibody (the antibody either bound to a solid matrix, or packaged separately with reagents for binding to a matrix), control formulations (positive and/or negative), and/or a detectable label, such as but not limited to fluorescein, green fluorescent protein, rhodamine, cyanine dyes, Alexa dyes, luciferase, and radiolabels, among others. Instructions for carrying out the assay, including, optionally, instructions for generating a MBDA score, can be included in the kit; e.g., written, tape, VCR, or CD-ROM. The assay can for example be in the form of a Northern hybridization or a sandwich ELISA as known in the art.
[00141] In some embodiments of the present teachings, biomarker detection reagents can be immobilized on a solid matrix, such as a porous strip, to form at least one biomarker detection site. In some embodiments, the measurement or detection region of the porous strip can include a plurality of sites containing a nucleic acid. In some embodiments, the test strip can also contain sites for negative and/or positive controls. Alternatively, control sites can be located on a separate strip from the test strip. Optionally, the different detection sites can contain different amounts of immobilized nucleic acids, e.g., a higher amount in the first detection site and lesser amounts in subsequent sites. Upon the addition of test sample, the number of sites displaying a detectable signal provides a quantitative indication of the amount of biomarker present in the sample. The detection sites can be configured in any suitably detectable shape and can be, e.g., in the shape of a bar or dot spanning the width of a test strip.
In other embodiments of the present teachings, the kit can contain a nucleic acid substrate array comprising one or more nucleic acid sequences. The nucleic acids on the array specifically identify one or more nucleic acid sequences represented by the MBDA markers. In various embodiments, the expression of one or more of the sequences represented by the MBDA markers can be identified by virtue of binding to the array. In some embodiments the substrate array can be on a solid substrate, such as what is known as a "chip." See, e.g., U.S. Pat. No. 5,744,305. In some embodiments the substrate array can be a solution array; e.g., xMAP (Luminex, Austin, TX), Cyvera (Illumina, San Diego, CA), RayBio Antibody Arrays (RayBiotech, Inc., Norcross, GA), CellCard (Vitra Bioscience, Mountain View, CA) and Quantum Dots' Mosaic (Invitrogen, Carlsbad, CA).
Machine-readable storage medium
[00142] A machine-readable storage medium can comprise, for example, a data storage material that is encoded with machine-readable data or data arrays. The data and machine- readable storage medium are capable of being used for a variety of purposes, when using a machine programmed with instructions for using said data. Such purposes include, without limitation, storing, accessing and manipulating information relating to the inflammatory disease activity of a subject or population over time, or disease activity in response to inflammatory disease treatment, or for drug discovery for inflammatory disease, etc. Data comprising measurements of the biomarkers of the present teachings, and/or the evaluation of disease activity or disease state from these biomarkers, can be implemented in computer programs that are executing on programmable computers, which comprise a processor, a data storage system, one or more input devices, one or more output devices, etc. Program code can be applied to the input data to perform the functions described herein, and to generate output information. This output information can then be applied to one or more output devices, according to methods well-known in the art. The computer can be, for example, a personal computer, a
microcomputer, or a workstation of conventional design.
[00143] The computer programs can be implemented in a high-level procedural or object- oriented programming language, to communicate with a computer system such as for example, the computer system illustrated in FIG. 15. The programs can also be implemented in machine or assembly language. The programming language can also be a compiled or interpreted language. Each computer program can be stored on storage media or a device such as ROM, magnetic diskette, etc., and can be readable by a programmable computer for configuring and operating the computer when the storage media or device is read by the computer to perform the described procedures. Any health-related data management systems of the present teachings can be considered to be implemented as a computer-readable storage medium, configured with a computer program, where the storage medium causes a computer to operate in a specific manner to perform various functions, as described herein.
[00144] The biomarkers disclosed herein can be used to generate a "subject biomarker profile" taken from subjects who have inflammatory disease. The subject biomarker profiles can then be compared to a reference biomarker profile, in order to diagnose or identify subjects with inflammatory disease, to monitor the progression or rate of progression of inflammatory disease, or to monitor the effectiveness of treatment for inflammatory disease. The biomarker profiles, reference and subject, of embodiments of the present teachings can be contained in a machine- readable medium, such as analog tapes like those readable by a CD-ROM or USB flash media, among others. Such machine-readable media can also contain additional test results, such as measurements of clinical parameters and clinical assessments. The machine-readable media can also comprise subject information; e.g., the subject's medical or family history. The machine- readable media can also contain information relating to other disease activity algorithms and computed scores or indices, such as those described herein. EXAMPLES
[00145] Aspects of the present teachings can be further understood in light of the following examples, which should not be construed as limiting the scope of the present teachings in any way.
[00146] The practice of the present teachings employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., T. Creighton, Proteins: Structures and Molecular Properties, 1993, W. Freeman and Co.; A. Lehninger, Biochemistry, Worth Publishers, Inc. (current addition); J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition, 1989; Methods In Enzymology, S.
Colowick and N. Kaplan, eds., Academic Press, Inc.; Remington's Pharmaceutical Sciences, 18th Edition, 1990, Mack Publishing Company, Easton, PA; Carey and Sundberg, Advanced Organic Chemistry, Vols. A and B, 3rd Edition, 1992, Plenum Press.
[00147] The practice of the present teachings also employ, unless otherwise indicated, conventional methods of statistical analysis, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., J. Little and D. Rubin, Statistical Analysis with Missing Data, 2nd Edition 2002, John Wiley and Sons, Inc., NJ; M. Pepe, The Statistical Evaluation of Medical Tests for Classification and Prediction (Oxford Statistical Science Series) 2003, Oxford University Press, Oxford, UK; X. Zhoue et al, Statistical Methods in Diagnostic Medicine 2002, John Wiley and Sons, Inc., NJ; T. Hastie et. al, The Elements of Statistical Learning: Data Mining, Inference, and Prediction, Second Edition 2009, Springer, NY; W. Cooley and P. Lohnes, Multivariate procedures for the behavioral science 1962, John Wiley and Sons, Inc. NY; E. Jackson, A User 's Guide to Principal Components 2003, John Wiley and Sons, Inc., NY.
Example 1: Deriving an MBDA score
[00148] This example demonstrates a method of deriving a Multi-Biomarker Disease Activity (MBDA) score, based on a dataset of quantitative data for biomarkers. In this example, a MBDA score is determined from the biomarker data using an interpretation function that is based on a set of predictive models, where each predictive model is predictive of a component of the DAS28-CRP, in this example TJC, SJC and patient global health assessment (GHA).
Deriving an MBDA score as described in this example is described in detail in USSN
12/905,984, herein incorporated by reference in its entirety.
MBDA algorithm development and evaluation
Training data
[00149] A MBDA algorithm was trained using clinical and biomarker data for patients in the InFoRM and BRASS studies. The InFoRM study (Index For Rheumatoid Arthritis
Measurement) is a multi-center observational study of the North American RA population. The patients used in algorithm training were recruited between April and September 2009 from 25 sites in the U.S. and Canada. Inclusion criteria were: age >18 years with a diagnosis of RA made by a board-certified rheumatologist. Patients concurrently enrolled in therapeutic drug trials involving a biologic agent and a placebo arm were excluded. The study includes three visits for each patient, each with clinical data and biological sample collection, at approximately three- month intervals.
[00150] BRASS is an observational study of approximately 1,000 RA patients receiving care at the RB Brigham Arthritis and Musculoskeletal Diseases Clinical Research Center, at the Brigham and Women's Hospital, Boston, Mass. Inclusion criteria were: age >18 years with a diagnosis of RA made by a board-certified rheumatologist. The study includes annual visits with clinical data and biological sample collection, and patient questionnaires between visits.
[00151] The first data set used in training consisted of visit 1 data for 512 InFoRM patients. The 512 patient visits were chosen to have clinical characteristics representative of the entire study population at the time of selection, and also to have been evaluated by a limited number of joint assessors. The number of joint assessors was limited to 12 so that assessor- specific biases could be evaluated and taken into account in algorithm development. The average age of these patients was 58.9 years (range 20-91), and 76% were female. The mean SJC and TJC were 4.28 and 5.49, respectively.
[00152] Assays for 25 candidate biomarkers were run on serum from the 512 InFoRM visits. Those biomarkers were SAA1, IL6, TNFRSF1A, VEGFA, PYD, MMP1, KAMI, calprotectin, CHI3L1 (YKL40), MMP3, EGF, IL1RA, VCAM1, LEP, RETN, CRP, IL8, APOA1, APOC3, CCL22, IL1B, IL6R, IL18, keratan sulfate and ICTP. All the biomarker assays were run on the Meso Scale Discovery (MSD®) platform. See Example 1 of US
2011/0137851 for specifics of biomarker assay development and evaluation.
[00153] The biomarkers were prioritized based on (1) univariate association with disease activity, (2) contribution to multivariate models for disease activity, and (3) assay performance.
[00154] The assays for 20 candidate biomarkers were run in a second set of patient samples, comprising 167 samples from BRASS and 29 from InFoRM. These 20 candidate biomarkers were SAA1, IL6, T FRSF1A, VEGFA, PYD, MMP1, ICAM1, calprotectin, YKL40, MMP3, EGF, ILIRA, VCAM1, leptin, resistin, CRP, IL8, CCL22, IL1B and IL6R. The samples were selected to enrich the overall training data for extremes of disease activity, while also having good representation of patients with moderate disease activity. Enriching for extreme phenotypes can result in improved algorithm training, as long as the resulting training population still fully represents the types of patients on which the algorithm will used in independent validation and intended use populations. The 167 BRASS samples were intended to represent similar numbers of patients with low, moderate and high disease activity. The 29 InFoRM samples were selected to represent patients with high disease activity, since low and moderate activity patients were already well represented by the first 512 training samples.
Data analysis
[00155] Prior to statistical analyses, all assay data were reviewed for pass/fail criteria on parameters including inter-assay CV, intra-assay CV, percent of samples within the measureable range of the calibration curve, and four serum process controls within the range of the calibration curve. The biomarker values that were not in the measureable range of the calibration curves were marked as missing data, and imputed with the lowest/highest detected value across all the samples within a given biomarker assay during the data export process. If the intra-assay CV of the biomarker concentration, computed from two replicates, was greater than 30%, it was also considered missing and excluded from univariate analyses. For multivariate analysis, individual biomarkers were excluded entirely if more than 20% of their data values were missing, and other missing data were imputed by the KNN algorithm (with k = 5 nearest neighbors). In the data used for algorithm training, no biomarkers were excluded from multivariate analysis because they all had less than 20% missing values. Concentration values were transformed as xO.1 prior to further analysis in order to make the distribution of values for each biomarker more normal. This transformation has a similar effect to log transformation but avoids the generation of negative values. The transformed, imputed biomarker dataset is denoted as X_(nxm), where X is the protein data from n markers and m samples.
[00156] In univariate analysis, the Pearson correlations between the levels of each biomarker and disease activity measures including DAS28-CRP4, DAS28-ESR4, SJC, TJC, GHA, SDAI and CDAI were calculated.
[00157] In multivariate analysis, statistical models were developed by five different regression methods. In the first regression method (1), forward stepwise ordinary least square regression, the equation Y = Χβ + ε applies, where Y is the column vector with observed values, β is the vector of coefficients, and ε is the residuals. The forward selection begins with no variables in the model. Then, given a collection of predictors X, the one having the largest absolute correlation with the response Y is selected and a simple linear regression of Y on XI is performed. The residual vector is now orthogonal to XI, and is taken to be the new response variable. The other predictors are then projected orthogonally to XI and the forward selection process is repeated.
[00158] In the second method (2), Lasso is used to prioritize biomarkers (based on R2 values) and to obtain a Lasso model. The "lasso" in this model minimizes the residual sum of squares, subject to the sum of the absolute value of the coefficients being less than a constant. This method produces interpretable models and exhibits the stability of ridge regression. See R. Tibshirani, J. Royal Stat. Soc. B 1996, 58(l):267-288.
[00159] In the third method (3), the Elastic Net, mixtures of Lasso and ridge penalties are applied. It encourages a grouping effect, where strongly correlated predictors segregate together, either tending to be in or out of the model together. See T. Zou, J. Royal Stat. Soc. B 2005, 67(2):301-320. For each of the above three methods, the marker selected at each step is recorded. [00160] The fourth method (4) is Multivariate Response with Curds and Whey (CW) using ordinary least squares (OLS). See L. Breiman and JH Friedman, J. Royal. Stat. Soc. B 1997, 59(l):3-54. This method takes advantage of the correlations between the response variables {e.g., components of DAS) to improve predictive accuracy, compared with the usual procedure of performing an individual regression of each response variable on the common set of predictor variables X. In CW, Y = XB * S, where Y = (ν¾ ) with k for the kth patient and j for j"1 response (j =1 for TJC, j = 2 for SJC, etc.), B is obtained using OLS, and S is the shrinkage matrix computed from the canonical co-ordinate system. Hence, this approach will yield submodels corresponding to each component of DAS.
[00161] The fifth method (5) is Curds and Whey and Lasso in combination (CW-Lasso).
Instead of using OLS to obtain B as in CW, Lasso was used, and the parameters were adjusted accordingly for the Lasso approach.
[00162] The performance of the five regression methods was compared in 70/30 cross validation (repeatedly training in a randomly selected 70% of the data and testing in the remaining 30%). The number of markers in each regression model was chosen by using nested 10-fold cross-validation once the number of markers was selected for a given analysis method the best-fitting model of that size was used to represent the method. In the CW approaches (methods 4 and 5), nested 10 fold cross validation was used for each sub-model corresponding to each component of DAS. The models developed using the CW-Lasso method performed best overall. The following sections consist of results mainly using CW-Lasso approach.
[00163] The 20 candidate biomarkers examined in all training samples were prioritized according to a number of criteria, including: strength of association with disease activity and contribution to multivariate models; consistency of correlation with disease activity across feasibility and training data sets; CRP was excluded from any sub-models for TJC, SJC, and PGA both because it is included in the DAS28-CRP4 and because it did not increase sub-model prediction accuracy in independent test samples (CRP is used, however, in the final MBDA score calculation as part of the MBDA formula); robust assay performance (ILIB was excluded from final modeling because its concentrations too frequently fall below the limits of detection of immunoassays); known drug effects (IL6R was excluded from final modeling because it is known to be strongly affected by tocilizumab, independently of the effects of the drug on disease activity); and, stability (TL8 was excluded from final modeling because its measurable levels are known to rise dramatically when serum samples are not kept cold). These criteria led to 15 candidate biomarkers being considered for inclusion in the final algorithm. See Table 1.
Table 1
Figure imgf000057_0001
Training the algorithm
[00164] While all data was used in prioritizing biomarkers, a subset was used for training the final algorithm. This subset was selected to have a broad range of disease activity levels, so that patients at all levels of disease activity were well represented. A comparison was made of the performance of models trained using: only BRASS samples (167 total); BRASS samples plus InFoRM samples (167+-100) selected to have a uniform disease activity distribution; or, BRASS samples plus InFoRM samples (167+-100) with a disease activity distribution like that of the BRASS samples.
[00165] The model performance was evaluated in an independent set of BRASS and InFoRM samples (70 total) set aside for this purpose. The DAS28-CRP distribution of this independent test set was similar to that of past studies (approximately normal). As shown below, correlation (r) to the DAS28-CRP and area under the ROC curve (AUROC) for predicting high and low DAS using median cut off were higher when training used BRASS samples plus "BRASS-like" InFoRM samples, although the differences were not statistically significant. The following Table 2 uses the Lasso regression method.
Table 2
Figure imgf000058_0001
[00166] For final training, the combination of BRASS plus "BRASS-like" InFoRM samples was selected. The CW-Lasso regression method was chosen for development of the final algorithm because of its superior performance in cross validation within the training set and in testing using InFoRM 512 patients and CAMERA patients (see below, MBDA algorithm performance, for a description of algorithm testing in another cohort of samples). In the application of this method, the shrinkage matrix was applied to the predictions of TJC and SJC. Ten-fold cross-validation indicated that the following 13 markers were optimal for performance. See Table 3.
Table 3
Figure imgf000058_0002
PYD X X
RETN X
SAA1 X X X
TNFRSF 1A X X
VCAM1 X X
VEGF 1 X X
[00167] From this set, PYD and calprotectin were excluded due to elevated assay failure rates. The remaining 1 1 biomarkers gave very similar algorithm performance to the full set of 13. An algorithm was chosen for validation that was developed by CW-Lasso regression using this 1 1- marker to estimate the DAS28-CRP in data from the BRASS + BRASS-like InFoRM samples. The estimates of TJC, SJC and PGHA were combined with a CRP test result in a formula similar to that used to calculate the DAS28-CRP.
DAS28CRP = + lj + 0.96
PDAS = 0.56J
Figure imgf000059_0001
+ l) + 0.96
[00168] Here IPTJC = Improved Prediction of TJC, IPSJC = Improved Prediction of SJC, PPGHA = Predicted PGHA, and PDAS is Predicted DAS28-CRP. (Details are defined below; see Selected algorithm). The MBDA score is the result from this formula.
[00169] Table 4 demonstrates the correlation of the values predicted by the PDAS algorithm with actual values for TJC, SJC, PGHA and DAS28-CRP, in the two cohorts studied, CAMERA and InFoRM.
Table 4
Figure imgf000059_0002
Selected algorithm
[00170] The 11 -marker + CRP Lasso model selected from the training process is as follows:
PTJC = -38.564 + 3.997 * (SAA1) 1/w + 17.331 * (IL6) 4.665 * (CHI3L1) 1/w - 15.236 *
(EGF) 1/10 + 2.651 * (TNFRSF1A) 1/10 + 2.641 * (LEP) 1/w + 4.026 * (VEGFA) 1/w- 1.47 *
(VCAM1) 1/10.
PSJC = -25.444 + 4.051 * (SAA1) 1/10 + 16.154 * (IL6) o- 11.847 * (EGF) 1/10 +3.091 * (CHI3L1) 1/10 + 0.353 * (TNFRSF1A) 1/10 ;
PPGHA = -13.489 + 5.474 * (IL6) 1/10 + 0.486 * (SAA1) 1/10 + 2.246 * (MMP1) 1/10 + 1.684 * (leptin) 1/10 + 4.14 * (TNFRSF1A) 1/10 + 2.292 * (VEGFA) 1/10 - 1.898 * (EGF) 1/10 + 0.028 * (MMP3) 1/10 - 2.892 * (VCAMl) 1/w- .506 * (REIN) o;
IPTJC = max(0.1739 * PTJC + 0.7865 * PSJC,0);
IPSJC = max(0.1734 * PTJC + 0.7839 * PSJC, 0);
MBDA score = round(max(min((56 * sqrt(IPTJC) + .28 * sqrt(IPSJC) + .14 * PPG A + .36 * ln(CRP/106 + 1)) * 10.53 + 1, 100), 1)).
[00171] For the final DA algorithm, the results from the 11 -marker + CRP CW-Lasso model were scaled and rounded to be integers on a scale of 1-100 such that a MBDA score of 1 would be equivalent to a DAS28-CRP value of 0, and a MBDA score of 100 would be equivalent to a DAS28-CRP value of 9.4.
[00172] Gene names in the above formulas correspond to serum protein concentrations, as obtained by the MSD® platform. Biomarker concentrations were obtained in the ranges shown in Table 5 (95% interval).
Table 5
Figure imgf000060_0001
pg/ml
Biomarker Lower Limit Upper Limit
SAA1 636,889 99,758,140
VCAM1 354,026 1,054,681
CRP 245,332 76,399,801
MM P I 3,047 39,373
MMP3 9,203 134,262
T FRSF1A 1, 139 4,532
RETN 3,635 19,308
CHI3L1 25,874 442, 177
MBDA algorithm performance
[00173] In order to independently test the performance of the algorithm developed above in this Example, a total of 120 serum samples were analyzed, obtained from the CAMERA study. Samples were obtained from the Computer Assisted Management in Early Rheumatoid Arthritis Study (CAMERA). From 1999-2003, all early rheumatoid arthritis patients (i.e., disease duration of one year or less) who fulfilled the 1987 revised American College of
Rheumatology (ACR) criteria for rheumatoid arthritis were asked to participate in this two-year randomized, open-label prospective multicentre strategy trial. As a result, 299 patients were studied. Patients visited the outpatient clinic of one of the six rheumatology departments in the region of Utrecht, the Netherlands, collaborating in the Utrecht Rheumatoid Arthritis Cohort study group. Inclusion criteria were that patients must have exhibited symptoms for less than one year, with age greater than 16 years. Exclusion criteria were the previous use of
glucocorticoids or any DMARD, use of cytotoxic or immunosuppressive drugs within a period of three months before inclusion, alcohol abuse, defined as more than two units per day, and psychological problems, which would make adherence to the study protocol impossible. At baseline all patients were monitored for medical conditions that would interfere with MTX usage. This screening included a chest X-ray, liver enzymes, albumin, hepatitis serology, serum creatinine and complete blood count. An independent person performed randomization in blocks of nine per hospital. The medical ethics committees of all participating hospitals approved this study, and all patients gave written informed consent before entering the study.
[00174] The cohort for this study had the following characteristics: 69% female, 68% CCP positive, 74% RF positive, 100% on MTX, 100% on non-biologic DMARDs, and 0% on biologic DMARDs. Additionally, the mean age of the cohort was 52 years (standard deviation (SD) +/- 14.7), with a minimum age of 17 and a maximum age of 78. The mean DAS28-CRP for this cohort was 5.0 (SD +/- 1.9), with a minimum of 0.9 and a maximum of 8.4.
[00175] A subpopulation of 72 subjects was selected from the CAMERA cohort for this
Example. All 72 patients were represented by baseline (time 0) visits and samples, and 48 were also represented by six-month visits and samples. Within the visits selected, a wide distribution of DAS28-CRP scores were represented, ranging from a minimum of 0.96 to a maximum of 8.4.
[00176] Of these, 72 samples were taken from subject baseline visits, and 48 were from visits six months subsequent to baseline. The concentrations of 23 serum protein biomarkers were measured in each sample: APOAl, APOC3, calprotectin, CCL22, CHI3L1 (YKL40), CRP, EGF, ICAMl, IL18, ILIB, ILIRA, IL6, IL6R, IL8, LEP, MMPl, MMP3, PYD, RETN, SAAl, TNFRSF1 A, VCAMl, and VEGFA. The concentrations of the markers were determined by customized immunoassays using either the Meso Scale Discovery SECTOR® Imager 6000 or individual ELISAs.
[00177] The associations between individual biomarkers and the clinical assessment measurements of DAS28-CRP, SJC28 and TJC28 were assessed by Pearson correlation (r) for log-transformed concentrations. The correlation p-values were adjusted for multiple hypothesis testing by estimating false discovery rates (FDR) using the method of Benjamini and Hochberg. See J. Royal Stat. Soc. B 1995 57(l):289-300.
[00178] Of the 23 proteins examined, fourteen were statistically significantly correlated with DAS28-CRP, eleven with SJC28 and nine with TJC28 (FDR < 0.05). See Table 6, which shows the Pearson correlations (r) between individual biomarkers and each clinical disease activity measure. The q-values reflect the FDRs, and were calculated by adjusting the p-values for multiple hypothesis testing. Statistically significant associations (q < 0.05) are in bold. As Table 6 shows, the individual biomarkers associated with disease activity represented a range of pathways associated with RA disease pathophysiology (Functional Category).
Table 6
Figure imgf000063_0001
[00179] Two pre-specified algorithms, a prototype and a final algorithm, using subsets of these 23 biomarkers were applied to calculate a total MBDA score for each subject at each visit (baseline and six-month). These algorithms were trained in prior studies using independent samples from other clinical cohorts. Algorithm performance was evaluated by Pearson correlation (r) and area under the ROC curve (AUROC) for identifying high and low disease activity at the baseline and six-month visits. The reference classification for ROC analysis was based on a DAS28-CRP of 2.67, the threshold separating remission/low disease activity from moderate and high disease activity.
Prototype algorithm for multivariate model
[00180] The first algorithm, or "prototype algorithm," using a linear combination of protein biomarkers, was trained on subject samples to estimate the DAS28 directly and was provided by the formula described elsewhere herein according to:
MBDA = b0 + b1 * DAIMRK X - b2 * DAIMRK2 x - b3 * DAIMRK3 x . . . - bn * DAIMRK„ x; where MBDA is the MBDA score, b0.„ are constants, and DAIMRK].n x are the serum
concentrations, transformed to the xth power, of n different biomarkers selected from the panel of biomarkers.
[00181] The prototype algorithm used in this Example was:
MBDA = (-16.1564) - (0.0606 * Calprotectin1/w) + (0.2194 * CHI3L11/W) + (1.1886 *
ICAM11/W) + (2.7738 * IL61/W) + (0.7254 * MMP11/W) - (0.8348 * MMP31/W) + (1.0296 * PYD1/W) + (1.1792 * SAA11/W) + (2.4422 * TNFRSF1A1/W) + (0.3272 * VEGFA1/W).
[00182] The prototype algorithm achieved a Pearson correlation (r) of 0.65 and an AUROC of 0.84 relative to the DAS28-CRP.
Biomarker selection for final algorithm
[00183] The second algorithm was derived using serum biomarker concentrations to separately estimate the three clinical assessments of TJC28, SJC28 and PGHA. Note that all of these are components of the formula used in calculating DAS28-CRP:
DAS28-CRP = 0.56 * sqrt(TJC28) + 0.28 * sqrt(SJC28) + 0.36 * ln(CRP+l) + (0.014 * PGHA) + 0.96. [00184] Biomarkers were then selected to predict and estimate clinical assessments of disease activity, specifically PGHA, TJC28 and SJC28. The resulting estimates were combined with a serum CRP concentration measurement to calculate an overall MBDA score. See FIG. 2, which indicates the three panels of biomarkers predictive of clinical disease activity
measurements, the union thereof, and CRP. The CW-Lasso method was used to predict the individual components of the DAS28; i.e., TJC28, SJC28 and PGHA. Note that biomarker terms are included in the CW-Lasso if they help to improve cross-validated model performance, and this criterion does not imply that each term is statistically significant by univariate analysis. A biomarker could make a significant contribution to a multivariate model even if it does not have a significant univariate correlation, and could not make a significant contribution to a
multivariate model even though it has a significant univariate correlation. Indeed, a comparison of each algorithm predictive for a clinical assessment, (a) - (c) above, with the biomarkers of Table 3 shows that not all biomarkers in each algorithm were individually statistically correlated with that clinical assessment. For example, values for serum concentrations of EGF, LEP, VEGFA and VCAMl are all included in the algorithm for predicting TJC28, yet each of these markers individually demonstrated a q-value for correlation with TJC of > 0.28. Including these markers, however, improves multivariate model performance in independent cross-validation test sets.
[00185] The overall MBDA score derived according to the methods of the present Example was given as a whole number between 1 and 100. The formula used to derive this score was provided by:
MBDA Score = ((0.56 * sqrt(PTJC) + 0.28*sqrt(PSJC) + 0.36 * log(CRP/106 + 1) + (0.14 * PPGHA) + 0.96) * 10.53) + 1, where PTJC = predicted TJC28, PSJC = predicted SJC28, and PPGHA = predicted PGA. This examples includes data from the following set of biomarkers: SAA1, IL6, CHI3L1, EGF, TNFRSF1A, LEP, VEGFA and VCAMl for PTJC; SAA1, IL6, EGF, CHI3L1 and TNFRSFIA for PSJC; SAA1, MMP1, LEP, TNFRSFIA, VEGFA, EGF, MMP3, VCAMl and RETN for PPGHA; plus CRP. In total, therefore, data from the following set of 12 markers was used to derive a MBDA score: CHI3L1, CRP, EGF, IL6, LEP, MMP1, MMP3, RETN, SAA1, TNFRSFIA, VCAMl and VEGFA. The predicted clinical assessments of disease activity were developed according to the following formulas:
(a) PTJC = -38.564 + (3.997 * SAA11/W) + (17.331 * IL61/1U) + (4.665 * CHI3Lrw) - (15.236 * EGF o) + (2.651 * TNFRSFIA1"10) + (2.641 * LEP1/W) + (4.026 * VEGFA1110) - (1.47 * VCAMl1110);
(b) PSJC = -25.444 + (4.051 * SAA11/10) + (16.154 * IL61/W) - (11.847 * EGF1/W) + (3.091 * CHI3L11/W) + (0.353 * TNFRSFIA1"10); and,
(c) PPGHA = -13.489 + (5.474 * IL *
Figure imgf000066_0001
LEP1/W) + (4.14 * TNFRSF1A1/W) + (2.292 * VEGFA1'10) - (1.898 * EGF1'10) + (0.028 * MMP31'10) - (2.892 * VCAMl1'10) - (0.506 * RETN1'10).
[00186] The performance of the above algorithm in deriving a MBDA score was evaluated by Pearson correlation (r) and area under the ROC curve (AUROC) for identifying high and low disease activity at the baseline and six-month visits. The Pearson correlation was 0.73, and the AUROC was 0.87, with the reference classification for ROC analysis based on a threshold DAS28-CRP of 2.67, the threshold separating remission/low disease activity from moderate and high disease activity. The changes in biomarker-based MBDA scores between the baseline and six-month visits were assessed by the paired Wilcoxon rank sum test.
[00187] To ensure that performance of the second algorithm was not overestimated due to the inclusion of two samples for some patients, subsets of samples were also analyzed that included only one randomly selected visit for each subject. The algorithm performed equally well in these subsets. Possible bias in the AUROC due to an imbalance in numbers between low and high disease activity groups was also analyzed using a DAS28-CRP cutoff of 2.67. When the cutoff was set at the median DAS28-CRP of 4.6, the AUROC was 0.83.
[00188] When the predictions of the individual components of the DAS28 generated by the MBDA algorithm were correlated to the actual TJC28, SJC28 and PGHA, the correlation coefficients were seen to trend higher (and thus provide better correlation with clinical disease activity measurements) than the coefficients for CRP, a marker commonly used alone as an indicator of RA disease activity. See FIG. 1. [00189] An analysis was then done to determine whether the MBDA score changed in response to the treatment protocols used in the CAMERA study. For all subjects for whom MBDA Scores were available for both visits (baseline and six-month), the median score dropped from 52 to 37 (p = 2.2E-6; n = 46). See FIG. 2. The intensive and conventional treatment arms were considered separately. There was also a significant decrease in median MBDA Score in the intensive treatment arm, from 52 to 36 (p = 2.5E-5; n = 31). In the conventional treatment arm, the median MBDA Score decreased from 59 to 45 (p = 0.06; n = 15).
[00190] In conclusion, this Example demonstrates that serum protein biomarkers representing a variety of biological pathways were consistently associated with RA disease activity. A pre-specified MBDA algorithm combining information from several of these biomarkers performed well in predicting RA disease activity when evaluated in an independent test set. The algorithm's estimates of TJC, SJC and PGHA correlated to actual clinical measures of disease activity. Furthermore, subsequent MBDA scores of the subjects analyzed decreased compared to initial MBDA scores following and in response to treatment.
Example 2: Predictive value of the multi-biomarker disease activity (MBDA) score for flare and sustained remission
[00191] The objective of this example is to determine the predictive value of the multi- biomarker disease activity (MBDA) score as described in Example 1 for flare and sustained remission after discontinuation of adalimumab (ADA) in patients with rheumatoid arthritis (RA). This example and corresponding figures use "MBDA" and "Vectra DA" and "VDA"
interchangeably.
Methods
[00192] The HONOR (Humira discontinuation withOut functional and radiographic damage progressioN following sustained Remission) study evaluated withdrawal of adalimumab (ADA) treatment, with continued use of methotrexate, from RA patients who were in stable remission, defined as DAS28 (ESR) <2.6 for at least the prior 24 weeks. Clinical disease activity, functional status, and joint damage were recorded at ADA discontinuation (baseline), and after 24 and 52 weeks. Twenty-four weeks following ADA discontinuation, remission was observed in 58% of patients. At 1 year, 48% of patients were in remission, 40% had flared (DAS28 (ESR) >3.2) and little change was observed in physical function or radiographic damage (Hirata S. et. al. Arthritis Res Ther. 25; 15(5):R135 (2013); Tanaka Y et al, Ann Rheum Dis . 74(2):389-95 (2015)). DAS28 (ESR) was only independent baseline predictor of remission at one year, observed in 68% of patients with baseline DAS28 (ESR) <1.98, compared with 25% with DAS28 (ESR) >1.98. Thus, HONOR demonstrated that it was possible to maintain disease activity score (DAS28, ESR < 2.6) clinical remission following discontinuation of ADA in approximately half of patients and that a very low DAS28 (ESR), termed "deep remission," was predictive of a favorable clinical outcome without functional and radiographic progression and that very low DAS28 (ESR) at the time of discontinuation was associated with successful ADA- free DS28 (ESR) < 2.6 (Hirata et al. 2013; Tanaka et al. 201).
[00193] The present study focused on the subset of patients who experienced ADA discontinuation (n=51), (Hirata et al. 2013) and excluded those who did not discontinue ADA. For subjects included in the present analysis, the baseline time point represents the time of ADA discontinuation in the HONOR study. Serum samples were available for 42 of the 51 subjects in the ADA discontinuation arm and were included in the present analysis. Clinical disease activity, functional status, and joint damage were assessed at the time of ADA discontinuation (baseline for the present study), and at 6 and 12 months post-discontinuation. Disease activity measures included the disease activity score (DAS) using 28 joints (DAS28) based on erythrocyte sedimentation rate (DAS28), (simplified disease activity index (SDAI), and clinical disease activity index (CDAI). Flare was defined as DAS28>3.2. Sustained clinical remission (SC- REM) was defined as absence of flare and DAS28 <2.6 at all visits. Physical function was assessed using the Health Assessment Questionnaire (HAQ-DI).
[00194] Baseline MBDA score and individual biomarkers were tested by univariate logistic regression models as the sole independent predictors of sustained low disease activity, i.e., DAS28-ESR<3.2; time patient remained free of ADA; functional good outcome (also referred as to as remission), i.e., HAQ <0.5; structural remission over 1 year, i.e., annual rate of AmTSS<0.5; and structural progression over 1 year, i.e., annual rate of AmTSS >0.5, >3, and >5. Multivariate logistic regression models were used to evaluate baseline MBDA score and individual biomarkers as predictors of each outcome when adjusted for one or more of the baseline patient and disease-related factors. Baseline variables with p-values <0.2 in univariate analyses were included in the multivariate model.
[00195] Receiver operating characteristic (ROC) curves of the ability of MBDA score to predict clinical remission, flare, and functional remission at 6 months and 1 year after discontinuation of ADA were generated and analyzed to identify the optimal cut-off value of baseline MBDA scores. P-values were based on 2-sided tests; any p-value <0.05 was considered statistically significant and <0.1 marginally significant.
[00196] MBDA scores (remission, <25; low, 26-29; moderate, 30-44; high, >44) were determined from serum samples collected at baseline. The ability of predicting flare (DAS28- ESR >3.2) or sustained clinical remission (SC-REM) (DAS28-ESR <2.6) by MBDA score and patient characteristics were determined. Patient characteristics at ADA discontinuation are shown in Table 7.
Table 7
Figure imgf000069_0001
* Anti-CCP status was missing for 5 patients
Results
[00197] At ADA discontinuation, all patients had DAS28-ESR <2.6 81% female, 69%
RF+, 81%) ACPA+ and 30 months mean disease duration. The median MBDA score was 24.5 [quartile; 14.3, 30.8] with 22 (52.4%) patients in remission, 6 (14.3%) low, 9 (21.4%) moderate and 5 (1 1.9%) high (Figure 3).
[00198] The MBDA score at discontinuation can be used as a predictor of flare. At 52 weeks, flare was observed in 12/42 (28.6%) (Figure 6). At 6 months post-discontinuation, there was a stepwise trend for increasing flare rate by increasing MBDA category (3.6% for low (<30), 1 1.1% for moderate (30-44), and 20.0% for high (>44) MBDA categories, respectively; p=0.158) (Figure 7A). A similar trend was observed at 1-year post ADA discontinuation (21.4% for low (<30) MBDA, 33.3% among moderate (30-44) MBDA, and 60.0% among those with high (>44) MBDA, although the trend did not reach statistical significance (p=0.08) (Figure 7B). Kaplan- Meier estimates examining the time to flare show a lower probability of flare among subjects with low (<30) or moderate (30-44) MBDA scores at baseline, compared to those with high MBDA scores (>44) (Figure 15).
[00199] The MBDA score at discontinuation can be used as a predictor of sustained functional remission. Percentage of sustained functional remission at year 1 by MBDA category (low/moderate/high) was 67.9%/55.6%/40% (Figure 13). Percentage of sustained functional remission at year 1 by MBDA category (remission/low/moderate/high) was
77.3%/33.3%/55.6%/40.0% (p=0.076) (Figure 14).
[00200] The MBDA score at discontinuation can be used as a predictor of sustained clinical remission (SC-REM) (DAS28<2.6, without flare). Rates of SC-REM were higher among individuals with lower baseline MBDA score categories at both 6 months post- discontinuation and at 1-year post discontinuation (Figure 1 1). Rates of SC-REM at 6 months and 1 year increased with increasing baseline MBDA score utilizing three MBDA categories (Figure 10) and four MBDA categories with a lower MBDA cut off value of <25 (MBDA remission) (Figure 1 1 A and 1 IB). Similarly, baseline MBDA scores tended to be lower among patients with SC-REM than those without SC-REM at both 6 months and 1 year post- discontinuation (Figure 9).
[00201] Univariate regression analyses identified MBDA score and disease duration as predictors of flare at 52 weeks (p<0.05). P-values for univariate logistic regression are illustrated in Figure 4 (6 months after ADA discontinuation) and in Figure 5 (1 year after ADA discontinuation).
[00202] Multivariate logistic is a predictor of SC-REM. Table 8 shows a multivariable logistic regression analysis of the contribution of baseline MBDA score and SC-REM controlling for potential confounders.
Table 8
Figure imgf000071_0001
Multivariate model includes MBDA score and disease duration.
b Multivariate model includes BMDA score, disease duration, and their interaction
[00203] Table 9 demonstrates odds rations for flare/remission using an MBDA cut off of
30).
Table 9
Figure imgf000071_0002
Odds ratio and 95% CI were from Fisher's Exact Test for count data. *Odds ratio is significantly different from 1 because the 95% CI excludes 1.
[00204] The optimal cutoffs of MBDA for flare and remission, based on the maximum of the sum of sensitivity and specificity, are shown in Table 10.
Table 10
Figure imgf000072_0001
[00205] Figure 16 displays ROC curves of clinical remission, flare, and functional remission at 6 months and 1 year after ADA discontinuation.
Conclusion
[00206] These findings suggest that the MBDA score could predict flare and biologic-free SC-REM in RA patients who were in stable clinical remission undergoing ADA withdrawal while maintaining MTX treatment in a clinical setting. These results support the potential utility of the MBDA score for guiding treatment decisions in patients receiving biologies.
[00207] All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
[00208] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the invention as defined in the appended claims.

Claims

Listing of Claims: What is claimed is:
1. A method for recommending a therapeutic regimen in a subject having an autoimmune disorder, the method comprising:
a) placing the subject on a therapy regimen;
b) determining whether the subject responds to the therapy regimen;
c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); resistin (RETN); serum amyloid Al (SAAl); tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA) if the subject responds to the therapy regiment response as determined in step b); and
d) recommending
i) reduction of the therapy regimen if the score is low; or
ii) no change in the therapy regimen if the score is high.
2. The method of claim 1, wherein the at least four biomarkers comprise IL6, EGF, SAAl, and CRP.
3. The method of claim 1, wherein the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAAl, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1.
4. The method of claim 1, wherein performance of the at least one immunoassay comprises:
obtaining the sample, wherein the sample comprises the protein markers;
contacting the sample with a plurality of distinct reagents;
generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data.
5. The method of claim 1, wherein the immunoassays comprise a multiplex assay.
6. The method of claim 1, wherein the autoimmune disorder is an arthritic disorder.
7. The method of claim 6, wherein the arthritic disorder is rheumatoid arthritis.
8. The method of claim 7, wherein the rheumatoid arthritis is early rheumatoid arthritis (eRA).
9. The method of claim 1, wherein the therapy regimen comprises one or more of methotrexate (MTX) and a biologic.
10. The method of claim 9, wherein the biologic comprises a TNF inhibitor.
11. The method of claim 10, wherein the TNF inhibitor is adalimumab.
12. The method of claim 1, wherein the score is predictive of a clinical assessment.
13. The method of claim 12, wherein the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28-ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC).
14. The method of claim 1, wherein the score is low if on a scale of 1-100, the score is less than or equal to about 30.
15. The method of claim 1, wherein the score is high if on a scale of 1-100, the score is greater than about 30.
16. The method of claim 1, wherein the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30.
17. The method of claim 1, wherein the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30.
18. The method of claim 1, wherein the score is low if on a scale of 1-100, the score is predictive of sustained clinical remission (SC-REM) and the score is less than or equal to about 23.
19. The method of claim 1, wherein the score is high if on a scale of 1-100, the score is predictive of SC-REM and the score is greater than about 23.
20. The method of claim 1, wherein the score is low if on a scale of 1-100, the score is predictive of functional remission and the score is less than or equal to about 26.
21. The method of claim 1, wherein the score is high if on a scale of 1-100, the score is predictive of functional remission and the score is greater than about 26.
22. The method of claim 1, wherein the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission.
23. The method of claim 22, wherein the remission is sustained if DAS28-ESR is <2.6 for >24 weeks.
24. The method of claim 1, wherein the remission is SC-REM.
25. The method of claim 1, wherein the remission is functional remission.
26. The method of claim 1, where the reduction of the therapy regimen is discontinuation of the therapy regimen.
27. A method for predicting flare or remission in a subject having an autoimmune disorder, the method comprising:
a) placing the subject on a therapy regimen;
b) determining whether the subject responds to the therapy regimen;
c) performing an immunoassay on a sample from the subject to generate a score based on a set of quantitative data, wherein the set of quantitative data comprises expression data for at least four biomarkers, wherein the at least four biomarkers are selected from a group comprising chitinase 3 -like 1 (cartilage glycoprotein-39) (CHI3L1); C-reactive protein, pentraxin-related (CRP); epidermal growth factor (beta-urogastrone) (EGF); interleukin 6 (interferon, beta 2) (IL6); leptin (LEP); matrix metallopeptidase 1 (interstitial collagenase) (MMP1); matrix metallopeptidase 3 (stromelysin 1, progelatinase) (MMP3); resistin (RETN); serum amyloid Al (SAA1); tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A); vascular cell adhesion molecule 1 (VCAM1); and, vascular endothelial growth factor A (VEGFA) if the subject responds to the therapy regiment response as determined in step b); and
d) predicting
i) no flare or sustained remission if the score is low; or
ii) flare or no sustained remission if the score is high.
28. The method of claim 27, wherein the at least four biomarkers comprise IL6, EGF, SAAl, and CRP.
29. The method of claim 27, wherein the at least four biomarkers comprise IL6, EGF, VEGFA, LEP, SAAl, VCAM1, CRP, MMP1, MMP3, TNFRSFIA, RETN, and CHI3L1.
30. The method of claim 27, wherein performance of the at least one immunoassay comprises:
obtaining the sample, wherein the sample comprises the protein markers;
contacting the sample with a plurality of distinct reagents;
generating a plurality of distinct complexes between the reagents and markers; and detecting the complexes to generate the data.
31. The method of claim 27, wherein the immunoassays comprise a multiplex assay.
32. The method of claim 27, wherein the autoimmune disorder is an arthritic disorder.
33. The method of claim 32, wherein the arthritic disorder is rheumatoid arthritis.
34. The method of claim 33, wherein the rheumatoid arthritis is early rheumatoid arthritis (eRA).
35. The method of claim 27, wherein the therapy regimen comprises one or more of
methotrexate (MTX) and a biologic.
36. The method of claim 35, wherein the biologic comprises a TNF inhibitor.
37. The method of claim 36, wherein the TNF inhibitor is adalimumab.
38. The method of claim 27, wherein the score is predictive of a clinical assessment.
39. The method of claim 38, wherein the clinical assessment is selected from the group consisting of: a DAS, a DAS28, a DAS28-CRP, a DAS28-ESR, a Sharp score, a tender joint count (TJC), and a swollen joint count (SJC).
40. The method of claim 27, wherein the score is low if on a scale of 1-100, the score is predictive of flare and the score is less than or equal to about 30.
41. The method of claim 27, wherein the score is high if on a scale of 1-100, the score is predictive of flare and the score is greater than about 30.
42. The method of claim 27, wherein the score is low if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is sustained clinical remission (SC-REM) and the score is less than or equal to about 23.
43. The method of claim 27, wherein the score is high if on a scale of 1-100, the score is predictive of sustained remission wherein the sustained remission is SC-REM and the score is greater than about 23.
44. The method of claim 27, wherein the score is low if on a scale of 1-100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is less than or equal to about 26.
45. The method of claim 27, wherein the score is high if on a scale of 1-100, the score is predictive of sustained remission and the sustained remission is functional remission and the score is greater than about 26.
46. The method of claim 27, wherein the subject is determined to have responded to the therapy regimen if the autoimmune disorder is in sustained remission.
47. The method of claim 46, wherein the remission is sustained if DAS28-ESR is <2.6 for >24 weeks.
48. The method of claim 27, wherein the flare is DAS28-ESR>3.2.
49. The method of claim 27, wherein the remission is sustained clinical remission (SC-REM).
50. The method of claim 27, wherein the remission is functional remission.
51. The method of claim 27, wherein the SC-REM is DAS28-ESR<2.6.
52. The method of claim 27, wherein the therapy regimen is not discontinued if flare or no remission is predicted.
PCT/US2017/020181 2016-03-02 2017-03-01 Biomarkers and methods for predicting response to discontinuation of inflammatory disease therapy WO2017151755A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662302487P 2016-03-02 2016-03-02
US62/302,487 2016-03-02

Publications (1)

Publication Number Publication Date
WO2017151755A1 true WO2017151755A1 (en) 2017-09-08

Family

ID=59744397

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/020181 WO2017151755A1 (en) 2016-03-02 2017-03-01 Biomarkers and methods for predicting response to discontinuation of inflammatory disease therapy

Country Status (1)

Country Link
WO (1) WO2017151755A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112466477A (en) * 2019-09-06 2021-03-09 广州市妇女儿童医疗中心 Pre-dose MTX treatment JIA efficacy prediction system and establishment method thereof
CN112466478A (en) * 2019-09-06 2021-03-09 广州市妇女儿童医疗中心 Efficacy prediction system for MTX treatment JIA after drug administration and establishment method thereof
US20230419489A1 (en) * 2017-07-24 2023-12-28 Ventana Medical Systems, Inc. Methods and systems for evaluation of immune cell infiltrate in tumor samples

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090017472A1 (en) * 2007-05-31 2009-01-15 Bruno Stuhlmuller BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNFalpha INHIBITORS IN AUTOIMMUNE DISORDERS
US20090142792A1 (en) * 2007-06-21 2009-06-04 Robinson William H Biomarkers for the diagnosis of autoimmune disease
US20110137851A1 (en) * 2009-10-15 2011-06-09 Crescendo Bioscience Biomarkers and methods for measuring and monitoring inflammatory disease activity
US20140142861A1 (en) * 2010-11-06 2014-05-22 Oklahoma Medical Research Foundation Biomarkers For Predicting Progressive Joint Damage
WO2015191423A1 (en) * 2014-06-09 2015-12-17 Crescendo Bioscience Biomarkers and methods for assessing response to inflammatory disease therapy

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090017472A1 (en) * 2007-05-31 2009-01-15 Bruno Stuhlmuller BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNFalpha INHIBITORS IN AUTOIMMUNE DISORDERS
US20090142792A1 (en) * 2007-06-21 2009-06-04 Robinson William H Biomarkers for the diagnosis of autoimmune disease
US20110137851A1 (en) * 2009-10-15 2011-06-09 Crescendo Bioscience Biomarkers and methods for measuring and monitoring inflammatory disease activity
US20140142861A1 (en) * 2010-11-06 2014-05-22 Oklahoma Medical Research Foundation Biomarkers For Predicting Progressive Joint Damage
WO2015191423A1 (en) * 2014-06-09 2015-12-17 Crescendo Bioscience Biomarkers and methods for assessing response to inflammatory disease therapy

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230419489A1 (en) * 2017-07-24 2023-12-28 Ventana Medical Systems, Inc. Methods and systems for evaluation of immune cell infiltrate in tumor samples
CN112466477A (en) * 2019-09-06 2021-03-09 广州市妇女儿童医疗中心 Pre-dose MTX treatment JIA efficacy prediction system and establishment method thereof
CN112466478A (en) * 2019-09-06 2021-03-09 广州市妇女儿童医疗中心 Efficacy prediction system for MTX treatment JIA after drug administration and establishment method thereof

Similar Documents

Publication Publication Date Title
US20220214341A1 (en) Biomarkers and methods for assessing psoriatic arthritis disease activity
CA2777800C (en) Biomarkers and methods for measuring and monitoring inflammatory disease activity
US20220057394A1 (en) Biomarkers and methods for measuring and monitoring axial spondyloarthritis activity
US20210208139A1 (en) Biomarkers and methods for assessing response to inflammatory disease therapy withdrawal
WO2015191423A1 (en) Biomarkers and methods for assessing response to inflammatory disease therapy
EP2635966A1 (en) Biomarkers for predicting progressive joint damage
JP2018531398A6 (en) Biomarkers and methods for assessing disease activity in psoriatic arthritis
EP3682240B1 (en) Assessing myocardial infarction and serious infection risk in rheumatoid arthritis patients
US20200249243A1 (en) Adjusted multi-biomarker disease activity score for inflammatory disease assessment
US20220057395A1 (en) Biomarkers and methods for assessing response to inflammatory disease therapy
WO2017151755A1 (en) Biomarkers and methods for predicting response to discontinuation of inflammatory disease therapy
EASTMAN et al. Patent 3021343 Summary

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17760710

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17760710

Country of ref document: EP

Kind code of ref document: A1