WO2023146979A1 - Plateforme de cytométrie de masse à cellule unique pour cartographier les effets d'agents candidats sur le cartilage - Google Patents

Plateforme de cytométrie de masse à cellule unique pour cartographier les effets d'agents candidats sur le cartilage Download PDF

Info

Publication number
WO2023146979A1
WO2023146979A1 PCT/US2023/011645 US2023011645W WO2023146979A1 WO 2023146979 A1 WO2023146979 A1 WO 2023146979A1 US 2023011645 W US2023011645 W US 2023011645W WO 2023146979 A1 WO2023146979 A1 WO 2023146979A1
Authority
WO
WIPO (PCT)
Prior art keywords
cartilage
clusters
cells
chondrocytes
agent
Prior art date
Application number
PCT/US2023/011645
Other languages
English (en)
Inventor
Nidhi Bhutani
Fiorella C. GRANDI
Neety SAHU
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
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 The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2023146979A1 publication Critical patent/WO2023146979A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6848Methods of protein analysis involving mass spectrometry
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/10Signal processing, e.g. from mass spectrometry [MS] or from PCR
    • 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
    • G01N2800/105Osteoarthritis, e.g. cartilage alteration, hypertrophy of bone
    • 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
    • G01N2800/60Complex ways of combining multiple protein biomarkers for diagnosis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/30Unsupervised data analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients

Definitions

  • Cartilage repair remains an unmet medical need with no disease-modifying drug available for degenerative diseases like osteoarthritis (OA), for which clinical care is limited to pain management or total joint replacement.
  • OA osteoarthritis
  • multiple pathways and targets have been considered for OA treatment, the rate of drug failure in clinical trials is high.
  • OA is a complex disease with a known interplay of metabolic, epigenetic, genetic, and cellular factors that influence its etiology. Drug discovery efforts are further confounded by the late detection of the disease and ambiguity about the causal and early events in OA pathogenesis.
  • Another critical knowledge gap is the lack of understanding of the heterogeneity between patients and its molecular underpinnings.
  • the present disclosure provides a single cell platform directed to osteoarthritis drug discovery.
  • compositions and methods are provided for determining the effect of an agent on cartilage, using single cell flow cytometry for analysis of chondrocytes following treatment with an agent of interest.
  • mass cytometry analysis is performed on a population of chondrocytes cultured in vitro with an agent of interest, e.g. analyzed by time-of- flight (cyTOF).
  • cyTOF time-of- flight
  • the chondrocytes are human cells.
  • a population of chondrocytes obtained from a human cartilage sample may be analyzed for a plurality of markers, e.g. at least 5, at least 10, at least 15, at least 20, at least 25 markers, and may comprise 30 markers, or more.
  • a marker is one or more of SOX9; CD24; Idu; Ki67; Hif2a; PNF ⁇ B(S29); iNOS; CD126; CD121 B; CD121A; CD120B; P-SAPK/JNK (T183/Y185); p-SMAD1/5(S463/465); pStat3(pY705); TET1 ; SOD2; Runxl ; Runx2; CD106; CD73; Notch-1 ; Stro-1 ; CD171 ; CD105; CD33; CD49E; CD146; TLR4; TLR2; and p16ink4a.
  • the cells are clustered into discrete subpopulations based on the presence and level of expression of the plurality of markers, including the clusters as shown in Table 2, where the number of discrete clusters may be at least 5, at least 10, at least 15, at least 20, at least 25 or more.
  • Analysis may include, without limitation, changes in a cell sub-population size, changes in expression of markers of interest, changes in phosphorylation of markers, and the like.
  • cells expressing low levels of SOX9 are included in the clustering analysis.
  • expression of p16ink4a is included in the clustering analysis.
  • Sub-populations (clusters) of interest may comprise cartilage-progenitor cells (CPC) subtypes, including a CD105 + Notch-1 + Stro1 + , high p16 ink4a chondroprogenitor population (CPC III).
  • CPC cartilage-progenitor cells
  • An inflammation amplifying (Inf-A) population was characterized by the co-expression of two cytokine receptors, IL1 R1 (CD121A) and TNFRII (CD120B) and an inflammation dampening (Inf- D) subpopulation characterized by the expression of CD24.
  • the Inf-A, Inf-D, and CPC III populations respectively, all belonging to the SOX9 medium group.
  • clusters 6, 16 and 17 belong to the SOX9 medium group while clusters 11 and 24 belong to the SOX9 high group.
  • No p16 ink4a expressing senescent subpopulations were detected in SOX9 low or SOX9 low-medium groups.
  • an agent is a candidate agent where the effect of the agent on chondrocyte populations is determined, where the effect is determined on the change in the distribution of cells into clustered chondrocyte subpopulations.
  • a principal component analysis may be used to plot the distribution of cells from an individual based on cluster abundance and marker expression profiles in each sample with or without drug treatment. The change resulting from drug treatment may be compared to controls, e.g. a positive control and a negative control to determine if the change is indicative of responsiveness.
  • the cells are obtained from an individual with osteoarthritis.
  • analysis is patient-specific, where the effect of a candidate agent on cells obtained from an individual patient is assessed.
  • a patient sample is analyzed for responsiveness to a known drug.
  • analysis is patient-specific, where the effect of the known drug is determined on cells obtained from the individual patient.
  • the patient may have osteoarthritis.
  • the response of chondrocytes to a drug is determined by staining with a plurality of markers, as disclosed herein, followed by mass cytometry analysis. The presence and level of expression of the plurality of markers is used to cluster the cells. The clustering is used to compare the patient sample response to a reference response, and the individual patient is determined to be a responder or non-responder to the agent.
  • an analysis of the results is provided to the individual.
  • an individual patient is treated with the known drug after being determined to be a responder.
  • FIGS. 1A-1 G Stratification of OA chondrocytes based on SOX9 expression.
  • FIGS. 2A-2D Identification of inflammatory and senescent populations in the OA chondrocyte landscape.
  • B Heatmap shows median expression of markers used to precisely identify CPCIII (CD105 + Notch-1 + Stro1 + ), Inf A (Hif2a + CD121 A + CD120B + pNF- ⁇ B + pJNK + pSMAD1/5 + ) and Inf-D (CD24 + ) clusters.
  • C UMAP projection showing p16 ink4a expressing senescent clusters (SnC).
  • FIGS. 3A-3G Effect of NF- ⁇ B inhibition on the OA chondrocyte landscape.
  • FIGS. 4A-4D NF- ⁇ B pathway inhibition targets p16 ink4a expressing senescent cells.
  • (C) UMAP depicting the differential abundance of p16 ink4a expressing clusters in the combined control and BMS- treated samples (n 6). Dashed line encircles clusters significantly depleted upon BMS treatment (paired t-test, p ⁇ 0.05).
  • (D) UMAP of the median expression of p16 ink4a in the combined control and BMS-treated samples (n 6).
  • FIGS. 5A-5F Effect of Kartogenin treatment on the OA chondrocyte landscape.
  • B Percent abundance of significantly different clusters in paired samples with control or Kartogenin treatment. Statistical significance was calculated by paired t-test at 95% confidence level and represented by exact p values.
  • C UMAP of p16 ink4a expressing clusters in control and Kartogenin samples. Dashed line encircling SnC I cluster represents significant depletion following Kartogenin treatment.
  • FIGS. 6A-6D Delineating ‘responders’ and ‘non-responders’ to OA drugs using CyTOF analyses
  • A Heatmap with hierarchical clustering of OA chondrocytes treated with control, BMS, or Kartogenin based on the expression profile of panel markers in each sample.
  • B Heatmap with hierarchical clustering of OA chondrocytes treated with control, BMS or Kartogenin based on the percent abundance of clusters in each sample.
  • C Principal Component Analysis (PCA) plot the distribution of patients based on cluster abundance and marker expression profiles in each patient sample with or without drug treatment.
  • D Schematic outlining a platform for CyTOF based profiling and analyses to identify responders and non-responders to a drug candidate in patient cohorts.
  • FIG. 7 Median expression profile of p16 ink4a clusters. Heatmap of the median expression of markers in the four p16 ink4a clusters, namely SnC I, SnC II, SnC III, SnC CPC III, and Snc IV in combined DMSO-treated samples. The median intensities were z-scored row-wise (per marker) for standardization.
  • FIGS. 8A-8E Effect of NF- ⁇ B pathway inhibition on iNOS and Hif2a expression.
  • Venn diagram representing clusters where the expression of (C) NF- ⁇ B and iNOS, (D) NF- ⁇ B and Hif2a, and (E) NF- ⁇ B, iNOS and Hif2a were commonly downregulated in BMS-treated samples when compared with DMSO-treated controls.
  • FIGS. 9A-9D Effect of drugs on the OA secretory output.
  • Spent medium was used for 80-plex autoantibody assay by Luminex.
  • a and C Fold change in the raw mean fluorescence intensity (MFI) of all analytes in samples treated with BMS-345541 (A) or kartogenin (C) normalized to respective DMSO-treated samples.
  • MFI raw mean fluorescence intensity
  • FIGS. 10A-10B FlowSOM identified the OA chondrocyte landscape.
  • B Proportional representation of all clusters as a percent of total cells in each sample across treatment groups.
  • FIGS. 11A-11C Drug effects on OA chondrocyte clusters stratified by SOX9 expression.
  • B Frequency of SOX9low, SOX9low-medium, SOX9medium and SOX9high clusters in drug-treated groups. Bar graphs represent mean ⁇ standard deviation. Statistical significance was calculated by repeated measures one-way ANOVA with Bonferroni correction and significant differences were represented by adjusted p values.
  • C Clustered dot plot represents the frequency as a percent of total cells in each cluster (dot size) and the median SOX9 expression in each cluster (dot color) in individual samples.
  • FIG. 12 Drug treatment effects on the secretory profile of OA chondrocytes.
  • Heatmap represents the raw MFI of 80 analytes (scaled row-wise) measured by multiplex autoantibody assay by Luminex in all DMSO, BMS-345541 and Kartogenin treated samples. Hierarchical clustering of rows as a measure of Euclidean distance was performed to visualize treatment effects on analytes. Analytes that are dampened in BMS-345541 treatment group compared to DMSO or Kartogenin treatment groups cluster together and are indicated by rectangle box in red.
  • Cartilage is a hyperhydrated structure with water comprising 70% to 80% of its weight. The remaining 20% to 30% comprises type II collagen and proteoglycan. Collagen usually accounts for 70% of the dry weight of cartilage.
  • Proteoglycans are composed of a central protein core from which long chains of polysaccharides extend. These polysaccharides, called glycosaminoglycans, include: chondroitin-4-sulfate, chondroitin-6-sulfate, and keratan sulfate.
  • Cartilage has a characteristic structural organization consisting of chondrocytes dispersed within an endogenously produced and secreted extracellular matrix. The cavities in the matrix which contain the chondrocytes are called cartilage lacunae. Unlike bone, cartilage is neither innervated nor penetrated by either the vascular or lymphatic systems.
  • Hyaline cartilage consists of a gristly mass having a firm, elastic consistency, is translucent and is pearly blue in color. Hyaline cartilage is predominantly found on the articulating surfaces of articulating joints. It is found also in epiphyseal plates, costal cartilage, tracheal cartilage, bronchial cartilage and nasal cartilage.
  • Fibrocartilage is essentially the same as hyaline cartilage except that it contains fibrils of type I collagen that add tensile strength to the cartilage. The collagenous fibers are arranged in bundles, with the cartilage cells located between the bundles.
  • Fibrocartilage is found commonly in the annulus fibrosis of the invertebral disc, tendinous and ligamentous insertions, menisci, the symphysis pubis, and insertions of joint capsules.
  • Elastic cartilage also is similar to hyaline cartilage except that it contains fibers of elastin. It is more opaque than hyaline cartilage and is more flexible and pliant. These characteristics are defined in part by the elastic fibers embedded in the cartilage matrix.
  • elastic cartilage is present in the pinna of the ears, the epiglottis, and the larynx.
  • articular cartilage prevents direct contact of the opposing bone surfaces and permits the near frictionless movement of the articulating bones relative to one another.
  • Two types of articular cartilage defects are commonly observed in mammals and include full-thickness and partial-thickness defects. The two-types of defects differ not only in the extent of physical damage but also in the nature of repair response each type of lesion elicits.
  • Full-thickness articular cartilage defects include damage to the articular cartilage, the underlying subchondral bone tissue, and the calcified layer of cartilage located between the articular cartilage and the subchondral bone.
  • Full-thickness defects typically arise during severe trauma of the joint or during the late stages of degenerative joint diseases, for example, during osteoarthritis. Since the subchondral bone tissue is both innervated and vascularized, damage to this tissue is often painful. The repair reaction induced by damage to the subchondral bone usually results in the formation of fibrocartilage at the site of the full-thickness defect. Fibrocartilage, however, lacks the biomechanical properties of articular cartilage and fails to persist in the joint on a long term basis.
  • Partial-thickness articular cartilage defects are restricted to the cartilage tissue itself. These defects usually include fissures or clefts in the articulating surface of the cartilage. Partialthickness defects are caused by mechanical arrangements of the joint which in turn induce wearing of the cartilage tissue within the joint. In the absence of innervation and vasculature, partial-thickness defects do not elicit repair responses and therefore tend not to heal. Although painless, partial-thickness defects often degenerate into full-thickness defects.
  • OA Osteoarthritis
  • NSAIDs non-steroidal antiinflammatory drugs
  • It is a chronic arthropathy characterized by disruption and potential loss of joint cartilage along with other joint changes, including bone remodeling such as bone hypertrophy (osteophyte formation), subchondral sclerosis, and formation of subchondral cysts.
  • OA results in the degradation of joints, including degradation of articular cartilage and subchondral bone, resulting in mechanical abnormalities and joint dysfunction. Symptoms may include joint pain, tenderness, stiffness, sometimes an effusion, and impaired joint function. A variety of causes can initiate processes leading to loss of cartilage in OA.
  • OA may begin with joint damage caused by trauma to the joint; mechanical injury to the meniscus, articular cartilage, a joint ligament, or other joint structure; defects in cartilage matrix components; and the like.
  • Mechanical stress on joints may underlie the development of OA in many individuals, with the sources of such mechanical stress being many and varied, including misalignment of bones as a result of congenital or pathogenic causes; mechanical injury; overweight; loss of strength in muscles supporting joints; and impairment of peripheral nerves, leading to sudden or dyscoordinated movements that overstress joints.
  • synovial joints there are at least two movable bony surfaces that are surrounded by the synovial membrane, which secretes synovial fluid, a transparent alkaline viscid fluid that fills the joint cavity, and articular cartilage, which is interposed between the articulating bony surfaces.
  • the earliest gross pathologic finding in OA is softening of the articular cartilage in habitually loaded areas of the joint surface. This softening or swelling of the articular cartilage is frequently accompanied by loss of proteoglycans from the cartilage matrix.
  • fibrillation As OA progresses, the integrity of the cartilage surface is lost and the articular cartilage thins, with vertical clefts extending into the depth of the cartilage in a process called fibrillation.
  • Joint motion may cause fibrillated cartilage to shed segments and thereby expose the bone underneath (subchondral bone).
  • the subchondral bone is remodeled, featuring subchondral sclerosis, subchondral cycts, and ectopic bone comprising osteophytes.
  • the osteophytes (bone spurs) form at the joint margins, and the subchondral cysts may be filled with synovial fluid.
  • the remodeling of subchondral bone increases the mechanical strain and stresses on both the overlying articular cartilage and the subchondral bone, leading to further damage of both the cartilage and subchondral bone.
  • the tissue damage stimulates chondrocytes to attempt repair by increasing their production of proteoglycans and collagen.
  • efforts at repair also stimulate the enzymes that degrade cartilage, as well as inflammatory cytokines, which are normally present in only small amounts.
  • Inflammatory mediators trigger an inflammatory cycle that further stimulates the chondrocytes and synovial lining cells, eventually breaking down the cartilage.
  • Chondrocytes undergo programmed cell death (apoptosis) in OA joints.
  • OA is characterized by low-grade infiltration of inflammatory cells, primarily macrophages, but also B cells and T cells. These cells, again primarily macrophages, are capable of producing inflammatory cytokines and matrix metalloproteases (MMPs) in the OA joint.
  • MMPs matrix metalloproteases
  • tissue-resident cells within the joint including synovial fibroblasts and chondrocytes, can produce additional inflammatory cytokines, including IL-6, as well as multiple MMPs.
  • OA should be suspected in patients with gradual onset of joint symptoms and signs, particularly in older adults, usually beginning with one or a few joints. Pain can be the earliest symptom, sometimes described as a deep ache. Pain is usually worsened by weight bearing and relieved by rest but can eventually become constant. Joint stiffness in OA is associated with awakening or inactivity. If OA is suspected, plain x-rays should be taken of the most symptomatic joints. X-rays generally reveal marginal osteophytes, narrowing of the joint space, increased density of the subchondral bone, subchondral cyst formation, bony remodeling, and joint effusions. Standing x-rays of knees are more sensitive in detecting joint-space narrowing. Magnetic resonance imaging (MRI) can be used to detect cartilage degeneration, and several MRI-based based scoring systems exist for characterizing the severity of OA (Hunter et al, PM R. 2012 May;4(5 Suppl):S68-74).
  • MRI Magnetic resonance imaging
  • OA commonly affects the hands, feet, spine, and the large weight-bearing joints, such as the hips and knees, although in theory any joint in the body can be affected. As OA progresses, the affected joints appear larger, are stiff and painful, and usually feel better with gentle use but worse with excessive or prolonged use. Treatment generally involves a combination of exercise, lifestyle modification, and analgesics. If pain becomes debilitating, joint-replacement surgery may be used to improve quality of life.
  • OA Osteoarthritis
  • dogs including dogs, cats, horses, and other animals in which it can causes significant joint pain and dysfunction.
  • Osteoarthritis (OA) is the most common form of arthritis in dogs, affecting approximately a quarter of the population. It is a chronic joint disease characterized by loss of joint cartilage, thickening of the joint capsule and new bone formation around the joint (osteophytosis) and ultimately leading to pain and limb dysfunction.
  • the majority of OA in dogs occur secondarily to developmental orthopedic disease, such as cranial cruciate ligament disease, hip dysplasia, elbow dysplasia, OCD, patella (knee cap) dislocation.
  • OA occurs with no obvious primary causes and can be related to genetic and age.
  • Other contributing factors to OA in dogs include body weight, obesity, gender, exercise, and diet.
  • Other evidence of joint failure can be determined by direct or arthroscopic visualization of extensive, complete, or near-complete loss of joint space or cartilage, by biomechanical assessment of inability to maintain functional joint integrity, or by clinical assessment of joint failure, as evidenced by inability to perform full range of motion or to maintain normal joint function.
  • Patients with advanced OA frequently experience joint pain. On physical examination, patients with advanced OA can have bony enlargement, small effusions, crepitus, and malalignment of the synovial joints.
  • Examples of semiquantitative MRI scoring systems that can be used to classify the severity of OA include: WORMS (Whole-Organ Magnetic Resonance Imaging Score; Peterfy CG, et al. Osteoarthritis Cartilage 2004;12:177-190); KOSS (Knee Osteoarthritis Scoring System; Kornaat PR, et al. Skeletal Radiol 2005;34:95-102); BLOKS (Boston Leeds Osteoarthritis Knee Score; Hunter DJ, et al. Ann Rheum Dis 2008;67:206-211 ); MOAKS (MRI Osteoarthritis Knee Score; Hunter DJ, et al. Osteoarthritis Cartilage.
  • WORMS Whole-Organ Magnetic Resonance Imaging Score
  • Peterfy CG et al. Osteoarthritis Cartilage 2004;12:177-190
  • KOSS Knee Osteoarthritis Scoring System
  • Kornaat PR et al.
  • Mass cytometry Elemental mass spectrometry-based flow cytometry (mass cytometry) is a method to characterize single cells or particles with elemental metal isotope-labeled binding reagents. Because there are many stable metal isotopes available, and little overlap between measurement channels, dozens of molecules (parameters) can be readily measured.
  • An example of a mass cytometer used to read the metal tags is an inductively-coupled plasma mass spectrometer (ICP-MS).
  • ICP-MS inductively-coupled plasma mass spectrometer
  • cells are first incubated with antibodies/affinity binders conjugated to pure isotopes and subsequently the cell suspension is injected as a single cell stream into the mass cytometer.
  • Single cell droplets are generated via nebulization and are carried by an argon gas stream into a .about.7500 degrees Kelvin plasma where each single cell is completely atomized and ionized. Thereby generated metal ions are then directed into a time-of-flight (TOF) mass spectrometer and the mass over charge ratio and number of metal ions is measured per cell and thereby the abundance of the target epitope/molecules.
  • TOF time-of-flight
  • CCP Capacitively coupled plasma
  • Mass spectrometer means an instrument for producing ions in a gas and analyzing them according to their mass/charge ratio.
  • MIP Microwave induced plasma
  • GD low discharge
  • Graphite furnace means a spectrometer system that includes a vaporization and atomization source comprised of a heated graphite tube. Spectroscopic detection of elements within the furnace may be performed by optical absorption or emission, or the sample may be transported from the furnace to a plasma source (e.g. inductively coupled plasma) for excitation and determination by optical or mass spectrometry.
  • a plasma source e.g. inductively coupled plasma
  • the methods utilize ICP-MS.
  • the ICP-MS is performed with solution analysis, for example ELAN DRC II, Perkin-Elmer.
  • the analysis is performed with a mass cytometer (e.g. CyTOF, DVS Sciences), which uses a nebulizer to administer a suspension of cells, beads, or other particles in a single-particle stream to an ICP-MS chamber, thereby yielding single particle/cell data similar to a flow cytometer.
  • the analysis is performed by an elemental analysis-driven imaging system (e.g. laser ablation ICP-MS). Devices for such analytic methods are known in the art.
  • flow cytometry refers to a method and a process whereby cells within a sample can be detected and identified when transversing past a detector within an apparatus containing a detecting source and a flowing apparatus.
  • An active agent can be administered by any suitable means, including topical, oral, parenteral, intrapulmonary, and intranasal.
  • Parenteral infusions include intramuscular, intravenous (bolus or slow drip), intraarterial, intraperitoneal, intrathecal or subcutaneous administration.
  • An agent can be administered in any manner which is medically acceptable. This may include injections, by parenteral routes such as intravenous, intravascular, intraarterial, subcutaneous, intramuscular, intratumor, intraperitoneal, intraventricular, intraepidural, or others as well as oral, nasal, ophthalmic, rectal, or topical. Sustained release administration is also specifically included in the disclosure, by such means as depot injections or erodible implants.
  • an agent can be formulated with an a pharmaceutically acceptable carrier (one or more organic or inorganic ingredients, natural or synthetic, with which a subject agent is combined to facilitate its application).
  • a suitable carrier includes sterile saline although other aqueous and non-aqueous isotonic sterile solutions and sterile suspensions known to be pharmaceutically acceptable are known to those of ordinary skill in the art.
  • An "effective amount” refers to that amount which is capable of ameliorating or delaying progression of the diseased, degenerative or damaged condition. An effective amount can be determined on an individual basis and will be based, in part, on consideration of the symptoms to be treated and results sought. An effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • compositions comprising a pharmaceutically acceptable excipient.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • the diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
  • the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.
  • compounds which are "commercially available” may be obtained from commercial sources including but not limited to Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc.
  • the active agents of the invention and/or the compounds administered therewith are incorporated into a variety of formulations for therapeutic administration.
  • the agents are formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and are formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • administration of the active agents and/or other compounds can be achieved in various ways, usually by oral administration.
  • the active agents and/or other compounds may be systemic after administration or may be localized by virtue of the formulation, or by the use of an implant that acts to retain the active dose at the site of implantation.
  • the active agents and/or other compounds may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination with other pharmaceutically active compounds.
  • the agents may be combined, as previously described, to provide a cocktail of activities.
  • the following methods and excipients are exemplary and are not to be construed as limiting the invention.
  • the agents can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • Formulations are typically provided in a unit dosage form, where the term "unit dosage form,” refers to physically discrete units suitable as unitary dosages for human subjects, each unit containing a predetermined quantity of active agent in an amount calculated sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the unit dosage forms of the present invention depend on the particular complex employed and the effect to be achieved, and the pharmacodynamics associated with each complex in the host.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are commercially available.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are commercially available.
  • Any compound useful in the methods and compositions of the invention can be provided as a pharmaceutically acceptable base addition salt.
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid.
  • Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • the active agent may be administered in dosages of 0.01 mg to 500 mg /kg body weight per day, e.g. about 20 mg/day for an average person. Dosages will be appropriately adjusted for pediatric formulation.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above. Langer, Science 249: 1527, 1990 and Hanes, Advanced Drug Delivery Reviews 28: 97- 119, 1997.
  • the agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.
  • the pharmaceutical compositions are generally formulated as sterile, substantially isotonic and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration.
  • GMP Good Manufacturing Practice
  • Toxicity of the active agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index.
  • the data obtained from these cell culture assays and animal studies can be used in further optimizing and/or defining a therapeutic dosage range and/or a sub-therapeutic dosage range (e.g., for use in humans). The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.
  • polypeptide peptide
  • protein protein
  • amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • sequence identity refers to the subunit sequence identity between two molecules. When a subunit position in both of the molecules is occupied by the same monomeric subunit (e.g., the same amino acid residue or nucleotide), then the molecules are identical at that position. The similarity between two amino acid or two nucleotide sequences is a direct function of the number of identical positions. In general, the sequences are aligned so that the highest order match is obtained. If necessary, identity can be calculated using published techniques and widely available computer programs, such as the GCS program package (Devereux et al., Nucleic Acids Res. 12:387, 1984), BLASTP, BLASTN, FASTA (Atschul et al., J. Molecular Biol. 215:403, 1990).
  • protein variant or “variant protein” or “variant polypeptide” herein is meant a protein that differs from a wild-type protein by virtue of at least one amino acid modification.
  • the parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide.
  • Variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it.
  • the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • parent polypeptide By “parent polypeptide”, “parent protein”, “precursor polypeptide”, or “precursor protein” as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant.
  • a parent polypeptide may be a wild-type (or native) polypeptide, or a variant or engineered version of a wild-type polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, gamma- carboxyglutamate, and O-phosphoserine.
  • amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a-carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Amino acid modifications disclosed herein may include amino acid substitutions, deletions and insertions, particularly amino acid substitutions.
  • Variant proteins may also include conservative modifications and substitutions at other positions of the cytokine and/or receptor (e.g., positions other than those involved in the affinity engineering). Such conservative substitutions include those described by Dayhoff in The Atlas of Protein Sequence and Structure 5 (1978), and by Argos in EMBO J., 8:779-785 (1989).
  • amino acids belonging to one of the following groups represent conservative changes: Group I: Ala, Pro, Gly, Gin, Asn, Ser, Thr; Group II: Cys, Ser, Tyr, Thr; Group III: Vai, lie, Leu, Met, Ala, Phe; Group IV: Lys, Arg, His; Group V: Phe, Tyr, Trp, His; and Group VI: Asp, Glu. Further, amino acid substitutions with a designated amino acid may be replaced with a conservative change.
  • isolated refers to a molecule that is substantially free of its natural environment.
  • an isolated protein is substantially free of cellular material or other proteins from the cell or tissue source from which it is derived.
  • the term refers to preparations where the isolated protein is sufficiently pure to be administered as a therapeutic composition, or at least 70% to 80% (w/w) pure, more preferably, at least 80%-90% (w/w) pure, even more preferably, 90-95% pure; and, most preferably, at least 95%, 96%, 97%, 98%, 99%, or 100% (w/w) pure.
  • a “separated” compound refers to a compound that is removed from at least 90% of at least one component of a sample from which the compound was obtained. Any compound described herein can be provided as an isolated or separated compound.
  • subject is used interchangeably herein to refer to a mammal being assessed for treatment and/or being treated.
  • the mammal is a human.
  • subject encompass, without limitation, individuals having a disease.
  • Subjects may be human, but also include other mammals, particularly those mammals useful as laboratory models for human disease, e.g., mice, rats, etc.
  • sample with reference to a patient encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof.
  • the term also encompasses samples that have been manipulated in any way after their procurement, such as by treatment with reagents; washed; or enrichment for certain cell populations, such as diseased cells.
  • the definition also includes samples that have been enriched for particular types of molecules, e.g., nucleic acids, polypeptides, etc.
  • biological sample encompasses a clinical sample, and also includes tissue obtained by surgical resection, tissue obtained by biopsy, cells in culture, cell supernatants, cell lysates, tissue samples, organs, bone marrow, blood, plasma, serum, and the like.
  • a “biological sample” includes a sample obtained from a patient’s diseased cell, e.g., a sample comprising polynucleotides and/or polypeptides that is obtained from a patient’s diseased cell (e.g., a cell lysate or other cell extract comprising polynucleotides and/or polypeptides); and a sample comprising diseased cells from a patient.
  • a biological sample comprising a diseased cell from a patient can also include non-diseased cells.
  • Cells for use in the methods as described above may be collected from a cartilage sample from a subject or a donor, and may optionally may be separated from a mixture of cells by techniques that enrich for desired cells, or may be engineered and cultured without separation.
  • An appropriate solution may be used for dispersion or suspension.
  • Such solution will generally be a balanced salt solution, e.g. normal saline, PBS, Hank’s balanced salt solution, etc., conveniently supplemented with fetal calf serum or other naturally occurring factors, in conjunction with an acceptable buffer at low concentration, generally from 5-25 mM.
  • Convenient buffers include HEPES, phosphate buffers, lactate buffers, etc.
  • the collected and optionally enriched cell population may be used immediately or may be frozen at liquid nitrogen temperatures and stored, being thawed and capable of being reused.
  • the cells will usually be stored in 10% DMSO, 50% FCS, 40% RPMI 1640 medium.
  • Cells can be obtained from cartilage tissue by digestion to release individual chondrocytes, which can be expanded and manipulated in culture, e.g. by contacting with a candidate or known agent for treating osteoarthritis. These cells are then collected and fixed in preparation for cyTOF, followed by standard staining and analysis protocols, see, for example, Grandi et al. (2021 ) Bio Protoc. 11 (14): e4086, herein specifically incorporated by reference. Cartilage is shaved off the bone using a scalpel, and 1 -2 mm cartilage pieces placed in standard tissue culture treated plates in complete media overnight. The sample is digested overnight, e.g. with collagenase. The cells are then collected and washed. Cells can be maintained in culture for several months, preferably at high densities.
  • diagnosis is used herein to refer to the identification of a molecular or pathological state, disease or condition in a subject, individual, or patient.
  • prognosis is used herein to refer to the prediction of the likelihood of death or disease progression, including recurrence, spread, and drug resistance, in a subject, individual, or patient.
  • prediction is used herein to refer to the act of foretelling or estimating, based on observation, experience, or scientific reasoning, the likelihood of a subject, individual, or patient experiencing a particular event or clinical outcome. In one example, a physician may attempt to predict the likelihood that a patient will survive.
  • Prognosis can also refer to identifying whether an individual is a responder or non-responder to a candidate agent.
  • treatment refers to administering an agent, or carrying out a procedure, for the purposes of obtaining an effect on or in a subject, individual, or patient.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of effecting a partial or complete cure for a disease and/or symptoms of the disease.
  • Treatment may include treatment of OA in a mammal, particularly in a human, and includes: (a) inhibiting the disease, i.e., arresting its development; and (b) relieving the disease or its symptoms, i.e., causing regression of the disease or its symptoms.
  • Treating may refer to any indicia of success in the treatment or amelioration or prevention of a disease, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the disease condition more tolerable to the patient; slowing in the rate of degeneration or decline; or making the final point of degeneration less debilitating.
  • the treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of an examination by a physician.
  • treating includes the administration of an agent to prevent or delay, to alleviate, or to arrest or inhibit development of the symptoms or conditions associated with disease or other diseases.
  • therapeutic effect refers to the reduction, elimination, or prevention of the disease, symptoms of the disease, or side effects of the disease in the subject.
  • a "therapeutically effective amount” refers to that amount of the therapeutic agent sufficient to treat or manage a disease or disorder.
  • a therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the onset of disease, e.g., to delay or minimize the growth and spread of oesteoarthritis.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of a disease.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means the amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of a disease.
  • the term “dosing regimen” refers to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time.
  • a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses.
  • a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses.
  • all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts.
  • a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen).
  • the methods of the invention are to provide an analysis of the response of chondrocytes to an agent of interest, which may be a candidate drug for treatment of a disease of cartilage, a known drug, where the cells are tested for responsiveness, and the like.
  • the cells are profiled for the presence and level of analytes, e.g. by mass cytometry.
  • analytes refers to quantifiable components of cells or biological material, particularly components that can be accurately measured.
  • An analyte can be any cell component or cell product including cell surface determinant, receptor, protein or conformational or posttranslational modification thereof, lipid, carbohydrate, organic or inorganic molecule, nucleic acid, e.g. mRNA, DNA, etc. or a portion derived from such a cell component or combinations thereof.
  • Proteins are of particular interest. Some variability may be expected and a range of values may be obtained using standard statistical methods with a common statistical method used to provide single values.
  • Individuals may be monitored for treatment and/or selected for therapy by determining the phenotype of the chondrocytes, e.g. the expression levels of proteins selected from at least 5, at least 10, at least 15, at least 20, at least 25 markers, and may comprise 30 markers, or more selected from SOX9; CD24; Idu; Ki67; Hif2a; PNF ⁇ B(S29) ; iNOS; CD126; CD121 B; CD121A; CD120B; P-SAPK/JNK (T183/Y185); p-SMAD1/5(S463/465); pStat3(pY705); TET1 ; SOD2; Runxl ; Runx2; CD106; CD73; Notch-1 ; Stro-1 ; CD171 ; CD105; CD33; CD49E; CD146; TLR4; TLR2; and p16ink4a.
  • the phenotype of the chondrocytes e.g. the expression levels of proteins selected from at least
  • the cells are clustered into discrete subpopulations based on the presence and level of expression of the plurality of markers, including the clusters as shown in Table 2, where the number of discrete clusters may be at least 5, at least 10, at least 15, at least 20, at least 25 or more.
  • Analysis may include, without limitation, changes in a cell sub-population size, changes in expression of markers of interest, changes in phosphorylation of markers, and the like.
  • cells expressing low levels of SOX9 are included in the clustering analysis.
  • expression of p16ink4a is included in the clustering analysis.
  • a specific change in a chondrocyte population e.g. percentage of cells in the population, growth, viability, protein expression, protein phosphorylation, etc. is correlated with response to an agent.
  • Assessment responsiveness allows improved care by indicating where therapeutic action may be required, at an early stage of disease. Where an individual is analyzed and found to be responsive to a therapy, treatment may be continued, or the patient allowed to recover following a suitable response. Where an individual is treated and found to not be responsive, an alternative therapy is administered.
  • an individual is assessed for responsiveness to a therapeutic agent by culturing a population of chondrocytes with the agent. Following a period of time sufficient for response, e.g. at 12 hours, at least 18 hours, at least 24 hours, up to 3 weeks or more, up to 2 weeks, up to 10 days, up to 7 days, up to 5 days, up to 3 days, up to 48 hours, up to 24 hours, the cell population can be analyzed, or frozen for future analysis.
  • Multiple samples may be obtained and analyzed from an individual over time. Multiple samples may also be obtained and analyzed over a patient cohort group, for example in the context of clinical trials.
  • Analytes of interest include cytoplasmic, cell surface or secreted biomolecules, frequently biopolymers, e.g. polypeptides, polysaccharides, polynucleotides, lipids, etc.
  • analytes include specific epitopes. Epitopes are frequently identified using specific monoclonal antibodies or receptor probes.
  • the molecular entities comprising the epitope are from two or more substances and comprise a defined structure; examples include combinatorially determined epitopes associated with heterodimeric proteins.
  • An analyte may be detection of a specifically modified protein or oligosaccharide, e.g.
  • a phosphorylated protein such as a STAT transcriptional protein; or sulfated oligosaccharide, or such as the carbohydrate structure Sialyl Lewis x, a selectin ligand.
  • the presence of the active conformation of a receptor may comprise one analyte while an inactive conformation of a receptor may comprise another, e.g. the active and inactive forms of heterodimeric integrin.
  • Analytes of interest include polypeptides, and the epitope that is being quantitated by a primary amino acid epitope, an epitope formed by protein secondary or tertiary structure, an epitope formed by two or more interacting polypeptides, or an epitope that results from posttranslational modification of a polypeptide.
  • specific binding member refers to a member of a specific binding pair, i.e. two molecules, usually two different molecules, where one of the molecules through chemical or physical means specifically binds to the other molecule.
  • one of the molecules is an analyte as defined above, and generally the specific binding member is labeled for detection of fluorescence or elemental analysis, as known in the art.
  • the complementary members of a specific binding pair are sometimes referred to as a ligand and receptor; or receptor and counter-receptor.
  • Specific binding indicates that the agent can distinguish a target antigen, or epitope within it, from other non-target antigens. It is specific in the sense that it can be used to detect a target antigen above background noise ("non-specific binding").
  • a specific binding partner can detect a specific sequence or a topological conformation.
  • a specific sequence can be a defined order of amino acids or a defined chemical moiety (e.g., where an antibody recognizes a phosphotyrosine or a particular carbohydrate configuration, etc.) which occurs in the target antigen.
  • the term "antigen" is issued broadly, to indicate any agent which elicits an immune response in the body.
  • An antigen can have one or more epitopes.
  • Binding pairs of interest include antigen and antibody specific binding pairs, complementary nucleic acids, peptide-MHC-antigen complexes and T cell receptor pairs, biotin and avidin or streptavidin; carbohydrates and lectins; complementary nucleotide sequences; peptide ligands and receptor; effector and receptor molecules; hormones and hormone binding protein; enzyme cofactors and enzymes; enzyme inhibitors and enzymes; and the like.
  • the specific binding pairs may include analogs, derivatives and fragments of the original specific binding member.
  • an antibody directed to a protein antigen may also recognize peptide fragments, chemically synthesized peptidomimetics, labeled protein, derivatized protein, etc. so long as an epitope is present.
  • Immunological specific binding pairs include antigens and antigen specific antibodies; and T cell antigen receptors, and their cognate MHC-peptide conjugates.
  • Suitable antigens may be haptens, proteins, peptides, carbohydrates, etc.
  • Recombinant DNA methods or peptide synthesis may be used to produce chimeric, truncated, or single chain analogs of either member of the binding pair, where chimeric proteins may provide mixture(s) or fragment(s) thereof, or a mixture of an antibody and other specific binding members.
  • Antibodies and T cell receptors may be monoclonal or polyclonal, and may be produced by transgenic animals, immunized animals, immortalized human or animal B-cells, cells transfected with DNA vectors encoding the antibody or T cell receptor, etc.
  • the details of the preparation of antibodies and their suitability for use as specific binding members are well-known to those skilled in the art.
  • a nucleic acid based binding partner such as an oligonucleotide can be used to recognize and bind DNA or RNA based analytes.
  • the term "polynucleotide” as used herein may refer to peptide nucleic acids, locked nucleic acids, modified nucleic acids, and the like as known in the art.
  • the polynucleotide can be DNA, RNA, LNA or PNA, although it is not so limited. It can also be a combination of one or more of these elements and/or can comprise other nucleic acid mimics.
  • Binding partners can be primary or secondary. Primary binding partners are those bound to the analyte of interest. Secondary binding partners are those that bind to the primary binding partner.
  • analysis is performed on a mass cytometer, in which cells are introduced into a fluidic system and introduced into the mass cytometer one cell at a time.
  • cells are carried in a liquid suspension and sprayed into a plasma source by means of a nebulizer.
  • the cells may be hydrodynamically focused one cell at a time through a flow cell using a sheath fluid.
  • the cells may be compartmentalized in the flow cell by introduction of an immiscible barrier, e.g., using a gas (e.g., air or nitrogen) or oil, such that the cell is physically separated from other cells that are passing through the flow cell.
  • the cells may be compartmentalized prior to or during introduction of the cell into the flow cell by introducing an immiscible material (e.g., air or oil) into the flow path.
  • an immiscible material e.g., air or oil
  • results of such analysis may be compared to results obtained from reference compounds and cells, positive controls, concentration curves, controls, etc.
  • the comparison of results is accomplished by the use of suitable deduction protocols, Al systems, statistical comparisons, etc.
  • the method described above may be employed in a multiplex assay in which a heterogeneous population of cells is labeled with a plurality of distinguishably labeled binding agents (e.g., a number of different antibodies). After the population of cells is labeled, the cells are introduced into the flow cell, and individually analyzed using the method described above, where the viable cells are distinguished from non-viable cells by the presence of platinum derived from the viability reagent.
  • a heterogeneous population of cells is labeled with a plurality of distinguishably labeled binding agents (e.g., a number of different antibodies).
  • the cells are introduced into the flow cell, and individually analyzed using the method described above, where the viable cells are distinguished from non-viable cells by the presence of platinum derived from the viability reagent.
  • the analyte distribution pattern may be generated from a cell sample using any convenient protocol.
  • the readout may be a mean, average, median or the variance or other statistically or mathematically-derived value associated with the measurement.
  • the readout information may be further refined by direct comparison with the corresponding reference or control pattern.
  • a pattern may be evaluated on a number of points: to determine if there is a statistically significant change at any point in the data matrix; whether the change is an increase or decrease in prevalence of an isoform; and the like.
  • the absolute values will display a variability that is inherent in live biological systems.
  • the cells that are analyzed by mass cytometry are clustered into discrete subpopulations based on the presence and level of expression of the plurality of markers. Clustering allows for assignment of cells into different groups, and provides a means of determining how the population of cells changes in response to the presence of a candidate agent.
  • the clustering step utilizes an algorithm for hierarchical clustering.
  • Clustering algorithms group similar objects such that the objects in the same group are more similar to each other than the objects in the other groups.
  • the group of similar objects is called a cluster.
  • the analysis is hierarchical cluster analysis or HCA, which is an unsupervised clustering algorithm that involves creating clusters that have predominant ordering from top to bottom.
  • HCA hierarchical cluster analysis
  • the algorithm groups similar objects, where the endpoint is a set of clusters or groups, where each cluster is distinct from each other cluster, and the objects within each cluster are broadly similar to each other.
  • This clustering technique is divided into two types: Agglomerative Hierarchical Clustering; and Divisive Hierarchical Clustering.
  • the Agglomerative Hierarchical Clustering is the most common type of hierarchical clustering used to group objects in clusters based on their similarity. It's a “bottom-up” approach: each observation starts in its own cluster, and pairs of clusters are merged as one moves up the hierarchy. There are several ways to measure the distance between clusters in order to decide the rules for clustering, and they are often called Linkage Methods. Some of the common linkage methods are: Complete- linkage: the distance between two clusters is defined as the longest distance between two points in each cluster; and Single-linkage: the distance between two clusters is defined as the shortest distance between two points in each cluster. This linkage may be used to detect high values in your dataset which may be outliers as they will be merged at the end. Average-linkage: the distance between two clusters is defined as the average distance between each point in one cluster to every point in the other cluster. Centroid-linkage: finds the centroid of cluster 1 and centroid of cluster 2, and then calculates the distance between the two before merging.
  • Clusters can be sorted into a dendrogram, which is a type of tree diagram showing hierarchical relationships between different sets of data.
  • a dendrogram contains the memory of hierarchical clustering algorithm.
  • Divisive or DIANA is a top-down clustering method where all of the observations are assigned to a single cluster and then partitioned to two least similar clusters, proceeding recursively on each cluster until there is one cluster for each observation.
  • FlowSOM is an algorithm that speeds time to analysis and quality of clustering with Self-Organizing Maps (SOMs) that can reveal how all markers are behaving on all cells, and can detect subsets that might otherwise be missed. It clusters cells (or other observations) based on chosen clustering channels (or markers/features), generates a SOM of clusters, produces a Minimum Spanning Tree (MST) of the clusters, and assigns each cluster to a metacluster, effectively grouping them into a population.
  • SOMs Self-Organizing Maps
  • the FlowSOM algorithm outputs SOMs and MSTs showing population abundances and marker expression in various formats including pie charts, star plots, and channel-colored plots. See, for example, Van Gassen et al. (2015), FlowSOM: Using self-organizing maps for visualization and interpretation of cytometry data.
  • UMAP is an algorithm for dimension reduction based on manifold learning techniques and ideas from topological data analysis.
  • UMAP uses simplicial complexes as a means to construct topological spaces out of simple combinatorial components. This allows one to reduce the complexities of dealing with the continuous geometry of topological spaces to the task of relatively simple combinatorics and counting.
  • the algorithm constructs a simplicial complex by constructing a fuzzy topological representation, then optimizing the low dimensional representation to have as close a fuzzy topological representation as possible as measured by cross entropy.
  • the obtained clustering pattern is compared to a single reference/control profile to obtain information regarding the phenotype of the cells being assayed.
  • the obtained protein distribution pattern is compared to two or more different reference/control profiles to obtain more in depth information regarding the phenotype of the cells.
  • the obtained protein distribution pattern may be compared to a positive and negative reference profile to obtain confirmed information regarding whether a cell has the phenotype of interest.
  • An algorithm that will discriminate robustly between groups of cells in different classifications with respect to responsiveness to therapy, and controls for confounding variables and evaluating potential interactions may be used for identifying candidate drugs.
  • the protein expression pattern is determined by the methods described above.
  • the quantitative data thus obtained is can be subjected to an analytic classification process.
  • the raw data is manipulated according to an algorithm, where the algorithm has been pre-defined by a training set of data, for example as described in the examples provided herein.
  • An algorithm may utilize the training set of data provided herein, or may utilize the guidelines provided herein to generate an algorithm with a different set of data.
  • Classification can be made according to predictive modeling methods that set a threshold for determining the probability that a sample belongs to a given class, i.e. responsive, non- responsive, etc.
  • the probability preferably is at least 50%, or at least 60% or at least 70% or at least 80% or higher.
  • Classifications also may be made by determining whether a comparison between an obtained protein distribution pattern and a reference protein distribution pattern yields a statistically significant difference. If such a comparison is not statistically significantly different from the reference protein distribution pattern, then the sample from which the protein distribution pattern was obtained is classified as belonging to the reference protein distribution pattern class.
  • a desired quality threshold is a predictive model that will classify a sample with an accuracy of at least about 0.7, at least about 0.75, at least about 0.8, at least about 0.85, at least about 0.9, at least about 0.95, or higher.
  • a desired quality threshold may refer to a predictive model that will classify a sample with an AUG (area under the curve) of at least about 0.7, at least about 0.75, at least about 0.8, at least about 0.85, at least about 0.9, or higher.
  • the relative sensitivity and specificity of a predictive model can be “tuned” to favor either the selectivity metric or the sensitivity metric, where the two metrics have an inverse relationship.
  • the limits in a model as described above can be adjusted to provide a selected sensitivity or specificity level, depending on the particular requirements of the test being performed.
  • One or both of sensitivity and specificity may be at least about at least about 0.7, at least about 0.75, at least about 0.8, at least about 0.85, at least about 0.9, or higher.
  • the raw data may be initially analyzed by measuring the values for each marker, usually in triplicate or in multiple triplicates.
  • the data may be manipulated, for example, raw data may be transformed using standard curves, and the average of triplicate measurements used to calculate the average and standard deviation for each patient. These values may be transformed before being used in the models, e.g. log-transformed, Box-Cox transformed (see Box and Cox (1964) J. Royal Stat. Soc., Series B, 26:211 —246), etc.
  • the data are then input into a predictive model, which will classify the sample according to the state.
  • the resulting information may be transmitted to a patient or health professional.
  • Candidate agents of interest are biologically active agents that encompass numerous chemical classes, primarily organic molecules, which may include organometallic molecules, inorganic molecules, genetic sequences, etc.
  • An important aspect of the invention is to evaluate candidate drugs, select therapeutic antibodies and protein-based therapeutics, with preferred biological response functions.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, frequently at least two of the functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules, including peptides, polynucleotides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. [00112] Included are pharmacologically active drugs, genetically active molecules, etc. Compounds of interest include chemotherapeutic agents, anti-inflammatory agents, hormones or hormone antagonists, ion channel modifiers, and neuroactive agents.
  • Exemplary of pharmaceutical agents suitable for this invention are those described in, "The Pharmacological Basis of Therapeutics," Goodman and Gilman, McGraw-Hill, New York, N.Y., (1996), Ninth edition, under the sections: Drugs Acting at Synaptic and Neuroeffector Junctional Sites; Drugs Acting on the Central Nervous System; Autacoids: Drug Therapy of Inflammation; Water, Salts and Ions; Drugs Affecting Renal Function and Electrolyte Metabolism; Cardiovascular Drugs; Drugs Affecting Gastrointestinal Function; Drugs Affecting Uterine Motility; Chemotherapy of Parasitic Infections; Chemotherapy of Microbial Diseases; Chemotherapy of Neoplastic Diseases; Drugs Used for Immunosuppression; Drugs Acting on Blood-Forming organs; Hormones and Hormone Antagonists; Vitamins, Dermatology; and Toxicology, all incorporated herein by reference. Also included are toxins, and biological and chemical warfare agents
  • Candidate agents include all of the classes of molecules described above, and may further comprise samples of unknown content. Of interest are complex mixtures of naturally occurring compounds derived from natural sources such as plants. While many samples will comprise compounds in solution, solid samples that can be dissolved in a suitable solvent may also be assayed. Samples of interest include environmental samples, e.g. ground water, sea water, mining waste, etc.; biological samples, e.g. lysates prepared from crops, tissue samples, etc.; manufacturing samples, e.g. time course during preparation of pharmaceuticals; as well as libraries of compounds prepared for analysis; and the like. Samples of interest include compounds being assessed for potential therapeutic value, i.e. drug candidates.
  • samples also includes the fluids described above to which additional components have been added, for example components that affect the ionic strength, pH, total protein concentration, etc.
  • the samples may be treated to achieve at least partial fractionation or concentration.
  • Biological samples may be stored if care is taken to reduce degradation of the compound, e.g. under nitrogen, frozen, or a combination thereof.
  • the volume of sample used is sufficient to allow for measurable detection, usually from about 0.1 .mu.I to 1 ml of a biological sample is sufficient.
  • Compounds, including candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds, including biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs.
  • Agents are screened for biological activity by adding the agent to the chondrocyte cultures disclosed herein.
  • the change in parameter readout in response to the agent is measured, desirably normalized, and clustered.
  • the agents are conveniently added in solution, or readily soluble form, to the medium of cells in culture.
  • the agents may be added in a flow-through system, as a stream, intermittent or continuous, or alternatively, adding a bolus of the compound, singly or incrementally, to an otherwise static solution.
  • a flow-through system two fluids are used, where one is a physiologically neutral solution, and the other is the same solution with the test compound added. The first fluid is passed over the cells, followed by the second.
  • a bolus of the test compound is added to the volume of medium surrounding the cells. The overall concentrations of the components of the culture medium should not change significantly with the addition of the bolus, or between the two solutions in a flow through method.
  • Preferred agent formulations do not include additional components, such as preservatives, that may have a significant effect on the overall formulation.
  • preferred formulations consist essentially of a biologically active compound and a physiologically acceptable carrier, e.g. water, ethanol, DMSO, etc.
  • a physiologically acceptable carrier e.g. water, ethanol, DMSO, etc.
  • the formulation may consist essentially of the compound itself.
  • a plurality of assays may be run in parallel with different agent concentrations to obtain a differential response to the various concentrations.
  • determining the effective concentration of an agent typically uses a range of concentrations resulting from 1 :10, or other log scale, dilutions.
  • the concentrations may be further refined with a second series of dilutions, if necessary.
  • one of these concentrations serves as a negative control, i.e. at zero concentration or below the level of detection of the agent or at or below the concentration of agent that does not give a detectable change in the phenotype.
  • Mass cytometry may be used to quantitate parameters such as the presence of cell surface proteins or conformational or posttranslational modification thereof; intracellular or secreted protein, where permeabilization allows antibody (or probe) access, and the like.
  • the readouts of selected parameters are capable of being read simultaneously, or in sequence during a single analysis, as for example through the use of itotope labeled antibodies to proteins.
  • Databases can typically comprise distribution pattern information from various conditions, such as responses of cells to a variety of treatments.
  • the results and databases thereof may be provided in a variety of media to facilitate their use.
  • Media can refer to a manufacture that contains the distribution pattern information; and methods of analysis as described above.
  • the databases and comparative algorithms can be recorded on computer readable media, e.g. any medium that can be read and accessed directly by a computer.
  • Such media include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage medium, and magnetic tape; optical storage media such as CD- ROM; electrical storage media such as RAM and ROM; and hybrids of these categories such as magnetic/optical storage media.
  • magnetic storage media such as floppy discs, hard disc storage medium, and magnetic tape
  • optical storage media such as CD- ROM
  • electrical storage media such as RAM and ROM
  • hybrids of these categories such as magnetic/optical storage media.
  • Recorded refers to a process for storing information on computer readable medium, using any such methods as known in the art. Any convenient data storage structure may be chosen, based on the means used to access the stored information. A variety of data processor programs and formats can be used for storage, e.g. word processing text file, database format, etc.
  • a computer-based system refers to the hardware means, software means, and data storage means used to analyze the information provided herein.
  • the minimum hardware of the computer-based systems of the present invention comprises a central processing unit (CPU), input means, output means, and data storage means.
  • CPU central processing unit
  • input means input means
  • output means output means
  • data storage means may comprise any manufacture comprising a recording of the present information as described above, or a memory access means that can access such a manufacture.
  • a variety of structural formats for the input and output means can be used to input and output the information in the computer-based systems of the present invention. Such presentation provides a skilled artisan with a ranking of similarities and identifies the degree of similarity contained in the test expression repertoire.
  • the data analysis may be implemented in hardware or software, or a combination of both.
  • a machine-readable storage medium comprising a data storage material encoded with machine readable data which, when using a machine programmed with instructions for using said data, is capable of displaying a any of the datasets and data comparisons of this invention.
  • Such data may be used for a variety of purposes, such as drug discovery, analysis of interactions between cellular components, and the like.
  • the analysis is implemented in computer programs executing on programmable computers, comprising a processor, a data storage system (including volatile and non-volatile memory and/or storage elements), at least one input device, and at least one output device.
  • Program code is applied to input data to perform the functions described above and generate output information.
  • the output information is applied to one or more output devices, in known fashion.
  • the computer may be, for example, a personal computer, microcomputer, or workstation of conventional design.
  • Each program can be implemented in a high level procedural or object oriented programming language to communicate with a computer system.
  • the programs can be implemented in assembly or machine language, if desired. In any case, the language may be a compiled or interpreted language.
  • Each such computer program can be stored on a storage media or device (e.g., ROM or magnetic diskette) readable by a general or special purpose programmable computer, for configuring and operating the computer when the storage media or device is read by the computer to perform the procedures described herein.
  • the system may also be considered to be implemented as a computer-readable storage medium, configured with a computer program, where the storage medium so configured causes a computer to operate in a specific and predefined manner to perform the functions described herein.
  • a variety of structural formats for the input and output means can be used to input and output the information in the computer-based systems.
  • One format for an output tests datasets possessing varying degrees of similarity to a trusted repertoire. Such presentation provides a skilled artisan with a ranking of similarities and identifies the degree of similarity contained in the test repertoire.
  • a method of storing and/or transmitting, via computer, data collected by the methods disclosed herein Any computer or computer accessory including, but not limited to software and storage devices, can be utilized to practice the present invention.
  • Data can be input into a computer by a user either directly or indirectly.
  • any of the devices which can be used to perform or analyze NIA can be linked to a computer, such that the data is transferred to a computer and/or computer-compatible storage device.
  • Data can be stored on a computer or suitable storage device (e.g., CD).
  • Data can also be sent from a computer to another computer or data collection point via methods well known in the art (e.g., the internet, ground mail, air mail).
  • data collected by the methods described herein can be collected at any point or geographical location and sent to any other geographical location.
  • reagents and kits thereof for practicing one or more of the above- described methods.
  • the subject reagents and kits thereof may vary greatly.
  • Reagents of interest include reagents specifically designed for use in production of the above described protein distribution patterns associated with chondrocytes and their responsiveness to therapy.
  • cyTOF conducted with 30 metal isotope-labeled antibodies parsed the effects of the drugs on inflammatory, senescent, and chondroprogenitor cell populations.
  • the NF- ⁇ B pathway inhibition decreased the expression of p-NF- ⁇ B, HIF2A, and inducible NOS in multiple chondrocyte clusters and significantly depleted 4 p16 ink4a -expressing senescent populations, including NOTCH1 + STRO1 + chondroprogenitor cells. While kartogenin also affected select p16 ink4a - expressing senescent clusters, there was a less discernible effect on chondroprogenitor cell populations.
  • Osteoarthritic chondrocytes were harvested from patients undergoing total knee arthroplasty and treated with two preclinically successful drugs for OA, an NF- ⁇ B pathway inhibitor, BMS-345541 and a stem cell modulator, Kartogenin. The cells were stained with 30 metal isotope- labeled antibodies and cyTOF acquired single-cell data was analyzed by unsupervised hierarchical clustering by FlowSOM to identify inflammatory, senescent and chondroprogenitor populations.
  • the single cell analyses parsed out the effects of the drugs on chondrocyte subpopulations and their redistribution.
  • the NF- ⁇ B pathway inhibitor significantly decreased NF- ⁇ B signaling in most but not all chondrocyte clusters along with Hif2a and iNOS expression.
  • NF- ⁇ B inhibition significantly depleted four p16 ink4a expressing senescent populations including Notch1 + Stro-1 + chondroprogenitor cells and reshaped the OA landscape. While both drugs led to depletion or expansion of multiple cell populations, Kartogenin had a modest effect in comparison to BMS-345541.
  • Single cell cyTOF-based screening platform can provide insightful predictive analyses of the uniformity of drug response as well as to stratify putative responders and non-responders. Such approaches can accelerate precision medicine-based drug discovery efforts for OA.
  • CPC chondroprogenitor cell
  • Inf-A an inflammation-amplifying population termed Inf-A, defined by elevated levels of IL1 R1 (CD121A) and TNFRII (CD120B) receptors. TNFRII has been shown to have antiinflammatory effects as opposed to TNFRI in some cell types. Chondrocytes coexpressing IL1 R1 (CD121A) and TNFRII (CD120B), however, showed active signaling through SMAD1/5 and JNK pathways, and inhibition of either of these pathways dampened secretion of inflammatory cytokines.
  • Another functional, inflammation- dampening population, Inf-D was characterized by the expression of CD24, a molecule we had previously identified as a negative regulator of NF- ⁇ B-mediated inflammation. Activation of Inf-D in combination with Inf-A inhibition affected multiple cytokines for an immunomodulatory effect on OA cartilage secretome.
  • DMSO control
  • Drug doses were determined based on prior literature and validation: 25 ⁇ M NF- ⁇ B inhibitor BMS- 345541 (Sigma-Aldrich B9935) and 25 ⁇ M kartogenin (Sigma-Aldrich SML0370).
  • Table 1 List of antibody markers categorized into chondrocyte, cell cycle, inflammation, signaling, chondroprogenitor (CPC) or TLRs and senescence related marker types with corresponding metal isotope conjugates identified by unique mass numbers.
  • markers for clustering is approximate to the methodology in previous study where surface markers with SOX9 and Hif2a were used so that previously defined subpopulations, particularly Inf-A, Inf-D and CPC clusters are adequately distinguishable.
  • 25 clusters and 10 metaclusters with 10 iterations were used as pre-determined input parameters as calibrated and utilized in previous study to identify subpopulations of interest with sufficient resolution.
  • Downsampling was avoided and therefore, all cells per sample were included in creating self-organizing maps (SOMs) for cluster generation. Abundances of clusters were represented as a percentage of total cells analyzed per sample.
  • UMAP projection was performed using Cytobank’s online platform to visualize FlowSOM generated clusters using 8043 cells per sample, pheatmap package was used to generate heatmaps in RStudio software.
  • Principal Component Analysis was performed using Factoextra package in Rstudio based on all clusters and median expression of all markers in individual patient samples.
  • the optimal cluster and metacluster numbers were selected after calibration and deemed sufficient in resolving the clusters (each cluster should have non-zero and greater than 1000 events across all samples) and identifying distinct subpopulations. Using these parameters, we were able to easily define the chondrocyte sub-populations identified in our previous study i.e inflammation-amplifying (Inf-A), inflammation-dampening (Inf-D) and chondroprogenitor population (CPCs) in the present dataset.
  • Inf-A inflammation-amplifying
  • Inf-D inflammation-dampening
  • CPCs chondroprogenitor population
  • SOX9 low sOX9 low medium
  • SOX9 medium SOX9 high
  • the SOX9 low group consisted of four clusters (4, 5, 9 and 10) and comprised only 2.6% of the total live cells (FIG. 1 D).
  • the SOX9 mid low group consisted of five clusters (1 , 3, 8, 13 and 22) and comprised 10.7% of the total live cells. Together, these cells that were excluded from analyses in our previous study comprised 13.3% of cells, hence were overall a small fraction of the OA chondrocytes.
  • clusters in the SOX9 low group were distinct from all other groups in having quite low median expression levels for the markers for chondroprogenitor cells (CPC) or inflammatory populations, suggesting that these are either OA chondrocytes with low inflammation or chondrocytes undergoing dedifferentiation (FIG. 1 G).
  • clusters in the SOX9 mid- low group were like the previously profiled SOX9 medium and SOX9 high groups in consisting of clusters that had either CPC-like phenotype (like CD105 + cluster 13) or had high inflammation (like cluster 22 with the expression of IL1 ⁇ receptors, CD121A and CD121 B).
  • the S0X9 medium group was the largest, comprising 71.5% of total live cells while the SOX9 high group comprised the rest 15%.
  • SOX9 high clusters (11 ,20,21 ,24) appeared highly inflamed with a higher overall expression of Hif2, NF- ⁇ B, iNOS, the IL6 receptor CD126 and IL1 ⁇ receptor CD121 B.
  • the SOX9 medium group on the other hand is mixed, consisting of clusters with relatively lower levels of inflammation (clusters 2, 7 and 15) along with clusters showing higher levels of NF- ⁇ B, iNOS and IL6 receptor CD126 (clusters 6, 12 and 16).
  • p16 ink4a expressing senescent cell populations in OA chondrocytes are distinct from Inf-A cells.
  • Our previously reported sub-populations in OA cartilage were easily identified in this study.
  • an inflammation-amplifying population was termed Inf-A that was defined by elevated levels of ILi ⁇ (CD121A) and TNFaRII (CD120B) receptors.
  • TNF RII has been shown to have pro-inflammatory effects as opposed to TNFRI in some cell types (Bluml, et al, Arthritis Rheum, 2010).
  • p16 ink4a was previously identified to mark a functional senescent population in OA cartilage that upon ablation or inhibition showed a decrease in senescence-associated secretory phenotype (SASP) and delayed OA in a mouse model (Jeon et al. Nat Med, 2017).
  • SASP senescence-associated secretory phenotype
  • FlowSOM identified five different clusters that expressed high levels of p16 ink4a -clusters 6, 11 , 16, 17, and 24 (FIG. 2C,D). Out of these, clusters 6, 16 and 17 belong to the SOX9 medium group while clusters 11 and 24 belong to the SOX9 high group.
  • chondroprogenitor cells can undergo senescence in OA cartilage.
  • SnC CPC III cells also expressed CD33 and TLR2 (FIG. 7), markers that are also coexpressed with p16 ink4a in clusters 6 and 11 (labeled SnC I and SnC II).
  • the other two clusters, 17 and 24 (labeled SnC III and SnC IV) showed higher expression of CD105 and p16 ink4a but not of CD33, TLR2, Notch-1 or Strol (FIG. 6).
  • NF- ⁇ B inhibition greatly alters the OA landscape depleting p16 ink4a expressing senescent clusters.
  • the NF- ⁇ B pathway is known to play a significant role in the function of inflammatory cytokines including IL1 ⁇ and TNFa in OA pathogenesis and drugs targeting this pathway have been extensively investigated. Since activation of inhibitor of ⁇ B (I ⁇ B) kinase is essential for NF- ⁇ B activation, we used a known selective inhibitor of IkB kinase (BMS 345541 ) to test the precise effect of NF- ⁇ B inhibition. This inhibitor BMS 345541 has already been shown to be effective in inhibiting OA pathogenesis in a preclinical mouse model of OA.
  • Cluster 3 of the SOX9 lo/mid group and clusters 7 and 14 of the SOX9 mid group were significantly depleted clusters.
  • Cluster 5 of the SOX9 lo group consisting of low-inflammation chondrocytes, was the only cluster that showed significant expansion.
  • RUNX2 expression was significantly downregulated in SnC I (cluster 6) among others (clusters 12, 20, 21 , and 25) (FIG. 5E).
  • kartogenin was shown to regulate the nuclear shuttling of core-binding factor ⁇ (CBF
  • 3 core-binding factor ⁇
  • NF- ⁇ B pathway inhibition by BMS-345541 significantly depleted the frequencies of the SOX9 mid and SOX9 hi groups, whereas treatment of OA chondrocytes with kartogenin had no significant effect in comparison with respective DMSO-treated controls.
  • both BMS-345541 and kartogenin increased the frequency of the SOX9 lo group.
  • the median expression of SOX9 in individual clusters belonging to all groups was maintained in kartogenin-treated samples to levels comparable to those in DMSO-treated controls.
  • NF- ⁇ B inhibition modulates the secretome of OA chondrocytes to a greater degree than kartogenin.
  • NF- ⁇ B pathway inhibition and kartogenin treatment had distinct effects on the OA landscape.
  • BMS-345541 and kartogenin were used to compare the effect of NF- ⁇ B pathway inhibition by BMS-345541 and kartogenin on the inflammatory secretome of OA chondrocytes.
  • An 80-plex autoantibody assay comprising cytokines, chemokines, and growth factors was chosen to assess the effect of the drug treatments on the OA chondrocyte secretome.
  • Multiplex autoantibody assay by Luminex on the culture supernatants of the treatment groups revealed substantial dissimilarity in the secretory profile of BMS-345541-treated OA chondrocytes compared with kartogenin treatment and DMSO-treated controls.
  • the levels of a plurality of secreted factors were decreased upon NF- ⁇ B pathway inhibition by BMS-345541 (FIG. 6A).
  • FMS-related tyrosine kinase 3 ligand FMS-related tyrosine kinase 3 ligand
  • PDGF- AA platelet-derived growth factor-AA
  • sFAS soluble FAS
  • PAl1 plasminogen activator inhibitor-1
  • FLT3L, PDGF-AA, and sFAS are known to be accumulated in the synovial fluid in OA patients.
  • PAI1 has pleiotropic roles dependent on tissue context such that it inhibits MMP activity under normal conditions whereas it increases ECM protein turnover, leading to fibrosis, under pathological conditions.
  • IL-10 is an antiinflammatory cytokine that regulates TNF- ⁇ -mediated effects in OA cartilage.
  • local fluctuations in IL-10 are transient but TNF- ⁇ expression levels can persist in OA; hence the downward trend in the levels of TNF- ⁇ and TNF- ⁇ in most patient samples implies positive anabolic regulation by BMS-345541 treatment (FIG. 6B).
  • BMS-345541 BMS-345541 treatment
  • the levels of macrophage migration inhibitory factor (MIF) were substantially higher compared with the levels observed in IL-20, -33, and -3.
  • cytokines such as IL-1 ⁇ as well as enhanced cell proliferation and antiapoptotic pathways.
  • SASP senescence- associated secretory phenotype
  • the secretory profile of kartogenin-treated samples did not deviate much from that of the respective DMSO- treated controls (FIG. 6C). Notable exceptions were the significantly elevated levels of epidermal growth factor (EGF) and a modest increase in the death signaling receptor FasL. As with BMS- 34551 , the levels of select SASP-associated cytokines were variable between patient samples in kartogenin-treated samples (FIG. 6D).
  • EGF epidermal growth factor
  • a high-resolution cellular atlas of OA articular cartilage is useful for understanding disease pathology and for devising new therapeutic strategies.
  • CPC cartilage progenitor cells
  • Inf-A and Inf-D two rare chondrocyte subpopulations
  • p16 ink4a was previously identified to mark a functional senescent population in OA cartilage.
  • Ablation or inhibition of p16 ink4a expressing cells showed a decrease in senescence-associated secretory phenotype (SASP) and delayed OA in a mouse model (Jeon et al. Nat Med, 2017).
  • p16 ink4a is just one of the senescent markers and a comprehensive analyses of other markers including p21 , p53, uPAR.
  • three of the SnC clusters (besides SnC III and IV) showed a co-expression of TLR2 and CD33 (Siglec 3) with p16 ink4a .
  • TLR2 has been previously observed to regulate oncogene-induced senescence and p16 ink4a function.
  • TLRs in addition to the other players like p21 , p53 and uPAR in cartilage- specific senescent cells and the multiple contributions to the senescence-associated secretory phenotype (SASP) will be interesting to explore in future studies.
  • Taylor SEB Smeriglio P, Dhulipala L, Rath M, Bhutani N.
  • a highly selective inhibitor of l?B kinase, BMS-345541 blocks both joint inflammation and destruction in collagen-induced arthritis in mice. Arthritis & Rheumatism 2003;48:2652-2659.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Medical Informatics (AREA)
  • Databases & Information Systems (AREA)
  • Theoretical Computer Science (AREA)
  • Computer Vision & Pattern Recognition (AREA)
  • Data Mining & Analysis (AREA)
  • Artificial Intelligence (AREA)
  • Epidemiology (AREA)
  • Evolutionary Computation (AREA)
  • Signal Processing (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Software Systems (AREA)
  • Evolutionary Biology (AREA)
  • Bioethics (AREA)

Abstract

L'invention concerne des compositions et des méthodes destinées à déterminer l'effet d'un agent sur le cartilage, à l'aide d'une cytométrie en flux à cellule unique pour l'analyse de chondrocytes après traitement avec un agent d'intérêt.
PCT/US2023/011645 2022-01-27 2023-01-26 Plateforme de cytométrie de masse à cellule unique pour cartographier les effets d'agents candidats sur le cartilage WO2023146979A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263303679P 2022-01-27 2022-01-27
US63/303,679 2022-01-27

Publications (1)

Publication Number Publication Date
WO2023146979A1 true WO2023146979A1 (fr) 2023-08-03

Family

ID=87472566

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/011645 WO2023146979A1 (fr) 2022-01-27 2023-01-26 Plateforme de cytométrie de masse à cellule unique pour cartographier les effets d'agents candidats sur le cartilage

Country Status (1)

Country Link
WO (1) WO2023146979A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200030323A1 (en) * 2014-01-28 2020-01-30 Buck Institute For Research On Aging Treating cognitive decline and other neurodegenerative conditions by selectively removing senescent cells from neurological tissue
WO2021067749A2 (fr) * 2019-10-02 2021-04-08 The Board Of Trustees Of The Leland Stanford Junior University Méthodes et compositions pour le traitement de l'arthrose
WO2021081374A1 (fr) * 2019-10-25 2021-04-29 Massachusetts Institute Of Technology Méthodes et compositions pour le criblage comprimé à haut rendement d'agents thérapeutiques

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200030323A1 (en) * 2014-01-28 2020-01-30 Buck Institute For Research On Aging Treating cognitive decline and other neurodegenerative conditions by selectively removing senescent cells from neurological tissue
WO2021067749A2 (fr) * 2019-10-02 2021-04-08 The Board Of Trustees Of The Leland Stanford Junior University Méthodes et compositions pour le traitement de l'arthrose
WO2021081374A1 (fr) * 2019-10-25 2021-04-29 Massachusetts Institute Of Technology Méthodes et compositions pour le criblage comprimé à haut rendement d'agents thérapeutiques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SAHU: "A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis", JCI INSIGHT, vol. 7, no. 20, 4 October 2022 (2022-10-04), pages e160702, XP093083447, DOI: 10.1172/jci.insight.160702 *

Similar Documents

Publication Publication Date Title
Mobasheri Osteoarthritis year 2012 in review: biomarkers
Hsueh et al. Synergistic roles of macrophages and neutrophils in osteoarthritis progression
Xu et al. The role of MCP-1-CCR2 ligand-receptor axis in chondrocyte degradation and disease progress in knee osteoarthritis
Kraus et al. Application of biomarkers in the development of drugs intended for the treatment of osteoarthritis
Chen et al. Divergent urinary metabolic phenotypes between major depressive disorder and bipolar disorder identified by a combined GC–MS and NMR spectroscopic metabonomic approach
Adamyan et al. Direct mass spectrometry differentiation of ectopic and eutopic endometrium in patients with endometriosis
Izbicka et al. Plasma biomarkers distinguish non-small cell lung cancer from asthma and differ in men and women
CN101971026A (zh) 检测tiabs的组合物和方法
Pesapane et al. Discovery of new small molecules inhibiting 67 kDa laminin receptor interaction with laminin and cancer cell invasion
WO2010045180A1 (fr) Biomarqueurs pour la maladie intestinale inflammatoire et leurs procédés d’utilisation
Speziali et al. Chondropenia: current concept review
Sahu et al. A single-cell mass cytometry platform to map the effects of preclinical drugs on cartilage homeostasis
CN110662966A (zh) 用于检测结直肠癌和晚期腺瘤的蛋白质生物标志物小组
Yang et al. Serum connective tissue growth factor is a highly discriminatory biomarker for the diagnosis of rheumatoid arthritis
JP2012073274A (ja) 変形性関節症のタンパク質プロフィール
CN102348982B (zh) 肌变性疾病的检测方法以及治疗效果判定方法
Rocha et al. Integrative metabolic pathway analysis reveals novel therapeutic targets in osteoarthritis
CN110702930B (zh) 24-羟基胆固醇在制备抑郁症诊断治疗相关产品的用途
Haartmans et al. Mass spectrometry-based biomarkers for knee osteoarthritis: a systematic review
Lyu et al. Metabolomics analysis reveals four biomarkers associated with the gouty arthritis progression in patients with sequential stages
Yan et al. A serum lipidomics study for the identification of specific biomarkers for endometrial polyps to distinguish them from endometrial cancer or hyperplasia
Zhang et al. High systemic immune‐inflammation index is relevant to osteoporosis among middle‐aged and older people: A cross‐sectional study
Torres-Torrillas et al. Treating full depth cartilage defects with intraosseous infiltration of plasma rich in growth factors: an experimental study in rabbits
WO2023146979A1 (fr) Plateforme de cytométrie de masse à cellule unique pour cartographier les effets d'agents candidats sur le cartilage
Cao et al. The association between methylmalonic acid, a biomarker of mitochondria dysfunction, and phenotypic age acceleration: A population-based study

Legal Events

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

Ref document number: 23747609

Country of ref document: EP

Kind code of ref document: A1