Gene Expression Inflammatory Age and Its Uses

Information

  • Patent Application
  • 20220291237
  • Publication Number
    20220291237
  • Date Filed
    February 25, 2022
    2 years ago
  • Date Published
    September 15, 2022
    a year ago
Abstract
The compounds and methods described herein can improve iAge (Inflammatory Age) of patients with a specific immunotype. For example, a patients iAge can be moved into a responders cohort from a non-responders cohort for an immunotherapy. The compounds and methods described herein can also improve cardiovascular patient outcomes using cAge to stratify CVD patients into risk cohorts for therapy and monitoring. Higher risk CVD patients can be converted to lower risk patients by treating the patients with molecules that reduce their cAge.
Description
BACKGROUND

Over the past five years cancer immunotherapy treatments have witnessed a great deal of clinical success in multiple cancer types often with extended disease-free survival periods of >10 years. Examples of successful immunotherapies are immune checkpoint inhibitors, which have demonstrated unprecedented rates of durable responses in many difficult-to-treat cancers. However, regardless of the organ affected and cancer type, only a limited percentage of patients (˜20%) benefit from these approaches. Thus, there is a growing need to identify biomarkers that will improve the selection of patients who will respond to therapy.


Biomarkers are needed both before and during treatment to enable identification of patients likely to respond to immunotherapy treatment in order to reduce inappropriate drug use. Objective clinical responses are defined as a reduction in tumor size during the course of treatment. Multiple baseline factors associated with disease prognosis have been linked to response rates. For example, patients with small-sized tumors or low baseline levels of serum lactate dehydrogenase (LDH) are more likely to respond to anti-PD-1 treatment. Circulating tumor DNA (ctDNA) that can be released by dead tumor cells and detected in the serum of some patients correlate strongly with tumor progression.


Response to anti-PD-1 treatment can partially be predicted by the expression of the ligand PD-L1 within the tumor microenvironment. Although PD-L1 expression is correlated with treatment efficacy in melanoma patients, it is not in patients with other cancers such as squamous cell carcinoma, non-small cell lung cancer and Merkel cell carcinoma.


The presence of neoantigens on tumor cells promotes immunogenicity against tumors and improves treatment efficacy. Thus, high genetic variation between tumor cells and host cells is one indicator of checkpoint inhibitor treatment efficacy. This is particularly true for anti-CTLA-4 treatment in melanoma patients and anti-PD-1 treatment in patients with colorectal cancer or non-small cell lung cancer with high mutation rates.


Other immunological factors associated with improved treatment responses prior to immunotherapy treatment include elevated eosinophil and lymphocyte counts; high numbers of CD8+ T cells infiltrating the tumor, and increased TGF-β levels in the serum from melanoma patients treated with anti-PD-1.


A number of post-treatment immune biomarkers have also been suggested to be associated with improved responses to cancer immunotherapy. For instance, patients who were more likely to respond to anti-CTLA-4 treatment had increased counts of inducible co-stimulatory molecule (ICOS)(+) T cells and lower neutrophil/lymphocyte ratios.


SUMMARY

The disclosure describes a method for treating subjects based on their gene expression inflammatory age (GE iAge) levels. Subjects can receive individualized interventions to reduce their gene expression inflammatory age. Improved GE iAge can improve a subject's clinical and immune responses to immunotherapy treatments, improve responses to vaccines and antimicrobial treatment, and improve overall health and well-being.


A gene expression inflammatory age scoring system (GE iAge) can be used to classify patients into those who will mount an objective clinical response to immunotherapy versus those who will not. The gene expression inflammatory age scoring system can be used to guide initial therapy targeting inflammation to enable optimal objective responses in those patients who were classified as non-responders.


The disclosure also describes a method for treating patients with cardiovascular disease or patients at risk of cardiovascular disease whereby subjects can be stratified based on risk for cardiovascular disease based on their inflammatory factor level; and can receive individualized interventions to treat and/or reduce the inflammatory factors and improve their risk profile, cardiovascular health, and response to cardiovascular treatments.


The disclosure also describes a method for treating patients with cardiovascular disease or patients at risk of cardiovascular disease whereby subjects can be stratified based on risk for cardiovascular disease based on their GE iAge.


The disclosure also describes methods of improving health, well being and longevity of patients by reducing the iAge score of certain markers so that the patients iAge score is lowered. Described herein are immunotypes that can be used to stratify patients into groups with similar GE iAge characteristics. Described herein are ten (10) different immunotypes. Four of the immunotypes subclassify those patients who have a significant decrease in iAge from their chronological age. The other six (6) immunotypes classify the remainder of the patients. Subjects in each immunotype can receive similar interventions to improve iAge of any subject with that immunotype. Thus, patients of the same immunotype can be provided with a treatment based upon their immunotype. Each immunotype treatment reduces the iAge score for patients in that immunotype which improves patient health, well-being and longevity.


The disclosure also describes methods of improving health, well being and longevity of patients by reducing the GE iAge score of a subject by changing the levels of certain markers so that the patients GE iAge score is lowered.


An inflammatory age scoring system (iAge) can be used to classify patients into those who have higher risk for cardiovascular disease versus those who have a low risk. The inflammatory age scoring system can be used to guide initial therapy targeting inflammation to improve outcomes of patients receiving treatment for cardiovascular disease, and to reduce risk of cardiovascular disease in asymptomatic patients (e.g., prophylactic treatment). MIG, EOTAXIN, Mip-1α, LEPTIN, IL-1(3, IL-5, IFN-α and IL-4 (positive contributors) and TRAIL, IFN-γ, CXCL1, IL-2, TGF-α, PAI-1 and LIF (negative contributors) are related to iAge and can be used to make up the iAge score. MIG, LIF and Sirtuin-3 are strongly related to cardiac aging and risk for cardiovascular disease and can be used alone or combination with other factors to define the risk level of a patient.


The disclosure describes a method for treating subjects with immunotherapy (e.g., cancer patients), vaccines (e.g., subjects who will benefit from vaccination), and antipathogen therapeutics (e.g., antibiotics, antivirals, antifungals, etc.) whereby subjects can be stratified based on their inflammatory age levels; and can receive individualized interventions to reduce inflammatory age and improve clinical and immune responses to the therapeutic treatment (e.g., cancer immunotherapy, vaccination, anti-pathogen therapeutic).


A gene expression inflammatory age scoring system (iAge) can be used to classify subjects (e.g., cancer patients, vaccination subjects, subjects with an infectious disease) into those who have an immune system that can mount an effective response (e.g., mount an objective clinical response to immunotherapy, produce a protective response to a vaccine, or mount an immune response against a pathogen) versus those who will not. The gene expression inflammatory age scoring system can be used to guide initial therapy targeting inflammation to enable optimal objective responses in those patients who were classified as non-responders. The GE iAge can also be used to stratify subjects for different courses of vaccines or antipathogen therapy.


Based on a subject's GE iAge, iAge, CRS, and/or Jak-STAT responses the subject can be classified as a responder or a nonresponder for the immunotherapy. Patients who are classified as nonresponders can be treated to lower their GE iAge, iAge, increase their CRS, and/or increase their Jak-STAT response so that the subject moves into a responder category. Classifications are made by comparing the subjects GE iAge, iAge, CRS, and/or Jak-STAT response to those of patients of similar chronological age. When a subject's GE iAge, iAge, CRS, and/or Jak-STAT response places them at a younger iAge for their age cohort, or a more responsive CRS and/or Jak-STAT score the subject can be a responder for immunotherapy. Subjects with older GE iAge and/or iAge for their age cohort, and/or lower scores for CRS and/or Jak-STAT can be treated to lower their GE iAge and/or iAge and/or increase their CRS and/or Jak-STAT score so that they move into a responder group for immunotherapy.


Based on a subject's GE iAge, iAge, CRS, Jak-STAT responses, cAge, and/or levels of MIG, LIF and/or SIRT3 the subject can be classified as high risk or low risk for cardiovascular disease. Patients who are classified as high risk can be treated to lower their GE iAge, iAge, increase their CRS, increase their Jak-STAT response, lower cAge, lower MIG, raise LIF and/or raise SIRT3 so that the subject moves into the low risk category. Classifications are made by comparing the subject's GE iAge, iAge, CRS, Jak-STAT responses, cAge, and/or levels of MIG, LIF and/or SIRT3 to those of patients of similar chronological age. When a subject's GE iAge, iAge, CRS, Jak-STAT responses, cAge, and/or levels of MIG, LIF and/or SIRT3 places them at a younger GE iAge for their age cohort, a younger iAge for their age cohort, or a more responsive CRS and/or Jak-STAT score, a lower cAge, a lower MIG, a higher LIF, and/or a higher SIRT3 the subject is less at risk for cardiovascular disease. Subjects with older GE iAge for their age cohort, iAge for their age cohort, lower scores for CRS and/or Jak-STAT, older cAge, a higher MIG, a lower LIF, and/or a lower SIRT3 can be treated to lower their iAge, increase their CRS and/or Jak-STAT score, lower cAge, lower MIG, increase LIF, and/or increase SIRT3 so that they move into the lower risk cohort of patients.


A subject's MIG, LIF, and Sirtuin-3 levels can also be used to classify risk for cardiovascular disease. Patients can be classified by their levels of MIG, Sirtuin-3, LIF, and optionally other factors. For example, the patients can be assigned a cardiac age based on these factors with or without other factors. When a patient's levels of MIG, SIRT3, LIF, and/or cardiac age (cAge) places them in a younger quartile, quintile, decile (or other quantile) for their age cohort the subject is less at risk for cardiovascular disease. Subjects with older levels of MIG, SIRT3, LIF, and/or cAge for their age cohort can be treated to lower their levels of MIG, SIRT3, LIF, and/or cAge so that they move into the lower risk cohort of patients.


In an aspect, the disclosure describes diagnosing cardiovascular disease, monitoring cardiovascular disease progression, monitoring the treatment of cardiovascular disease, prognosing cardiovascular disease, treating cardiovascular disease, alleviating symptoms of cardiovascular disease, inhibiting progression of cardiovascular disease, and preventing cardiovascular disease, in a mammal using the markers, combinations of markers, treatments, prophylactic treatments, and/or agents provided herein.


In an aspect, the disclosure describes compounds and methods for modifying GE iAge, iAge (or cAge) of a subject. The GE iAge, iAge (or cAge) modification can reclassify the cohort of a subject undergoing cancer treatment, immunotherapy, or cardiovascular disease treatment. The compounds and methods can modify one or more markers involved in the GE iAge or iAge determination.


In an aspect, the disclosure describes compositions which can be used to improve the GE iAge or iAge of individuals within certain immunotypes. Treatments can include, for example, combinations of components that can alter the level of a GE iAge or an iAge marker to healthier levels (lowers GE iAge or iAge) for one or more of the GE iAge markers (GBP5, MMP9, SIGLEC5, S100P, OLFM1, CISH, MT1A, IGLL1, RPLP0, SLC16A10, FCER1A, DDX3Y, or MAN1A1) or iAge markers (TRAIL, GROA, IFNg, MIG, or EOTAXIN). Such combinations can include a combination of one or more of the following: iron, biotin, caffeine, manganese chloride, niacin, carrageenan, beta-carotene, leutin, zinc-sulfate, vitamin D2, guar gum, kawain, indole-3-carbinol, and/or picetannol.


The disclosure also describes methods for identifying drugs, food compounds and other molecules that modify iAge (or cAge). These methods identify drugs, food compounds, and other molecules that interact with and modify the levels of certain markers involved with the iAge (or cAge) determination. These drugs, food compounds, and other molecules can be used with subjects to modify their iAge (or cAge) and so change the cohort in which the subject stratifies and so alter the response of the subject to treatment and/or risk of disease.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A, 1B and 1C show graphs of iAge, naïve CD8(+) T-cells, and Jak STAT signaling responses.



FIG. 2 shows the stratification of cancer patients by iAge and CRS into responders and nonresponders.



FIG. 3 shows the stratification of cancer patients using iAge.



FIG. 4 depicts the fold change in MIG mRNA and SIRT3 mRNA after passage of cells following differentiation into endothelial cells from hiPSCs.



FIG. 5 is a bar graph showing the relative expression levels of CXCR3 in different cell types.



FIG. 6 shows the four immunotypes of the super-healthy group of patients.



FIG. 7 shows the six immunotypes of the normal group of patients.



FIG. 8 shows the levels of EOTAXIN, TRAIL, GROA, IFNg, and MIG (after normalization of the data as discussed below) for each the super-healthy immunotypes (SH1-4).



FIG. 9 shows the levels of EOTAXIN, TRAIL, GROA, IFNg, and MIG (after normalization of the data as discussed below) for each the normal immunotypes (N1-6).





DETAILED DESCRIPTION

Before the various embodiments are described, it is to be understood that the teachings of this disclosure are not limited to the particular embodiments described, and as such can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present teachings will be limited only by the appended claims.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present teachings, some exemplary methods and materials are now described.


It must be noted that as used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims can be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation. Numerical limitations given with respect to concentrations or levels of a substance are intended to be approximate, unless the context clearly dictates otherwise. Thus, where a concentration is indicated to be (for example) 10 μg, it is intended that the concentration be understood to be at least approximately or about 10 μg.


As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which can be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present teachings. Any recited method can be carried out in the order of events recited or in any other order which is logically possible.


Definitions

In reference to the present disclosure, the technical and scientific terms used in the descriptions herein will have the meanings commonly understood by one of ordinary skill in the art, unless specifically defined otherwise. Accordingly, the following terms are intended to have the following meanings.


As used herein, “activation” is defined to be a physiological condition upon exposure to a substance, allergen, drug, protein, chemical, or other stimulus, or upon removal of a substance, allergen, drug, protein, chemical or other stimulus.


As used herein, an “antibody” is defined to be a protein functionally defined as a ligand-binding protein and structurally defined as comprising an amino acid sequence that is recognized by one of skill as being derived from the variable region of an immunoglobulin. An antibody can consist of one or more polypeptides substantially encoded by immunoglobulin genes, fragments of immunoglobulin genes, hybrid immunoglobulin genes (made by combining the genetic information from different animals), or synthetic immunoglobulin genes. The recognized, native, immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes and multiple D-segments and J-segments. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. Antibodies exist as intact immunoglobulins, as a number of well characterized fragments produced by digestion with various peptidases, or as a variety of fragments made by recombinant DNA technology. Antibodies can derive from many different species (e.g., rabbit, sheep, camel, human, or rodent, such as mouse or rat), or can be synthetic. Antibodies can be chimeric, humanized, or humaneered. Antibodies can be monoclonal or polyclonal, multiple or single chained, fragments or intact immunoglobulins.


As used herein, an “antibody fragment” is defined to be at least one portion of an intact antibody, or recombinant variants thereof, and refers to the antigen binding domain, e.g., an antigenic determining variable region of an intact antibody, that is sufficient to confer recognition and specific binding of the antibody fragment to a target, such as an antigen. Examples of antibody fragments include, but are not limited to, Fab, Fab′, F(ab′)2, and Fv fragments, scFv antibody fragments, linear antibodies, single domain antibodies such as sdAb (either VL or VH), camelid VHH domains, and multi-specific antibodies formed from antibody fragments. The term “scFv” is defined to be a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chain variable regions are contiguously linked via a short flexible polypeptide linker, and capable of being expressed as a single chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived. Unless specified, as used herein an scFv may have the VL and VH variable regions in either order, e.g., with respect to the N-terminal and C-terminal ends of the polypeptide, the scFv may comprise VL-linker-VH or may comprise VH-linker-VL.


As used herein, an “antigen” is defined to be a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both. The skilled artisan will understand that any macromolecule, including, but not limited to, virtually all proteins or peptides, including glycosylated polypeptides, phosphorylated polypeptides, and other post-translation modified polypeptides including polypeptides modified with lipids, can serve as an antigen. Furthermore, antigens can be derived from recombinant or genomic DNA. A skilled artisan will understand that any DNA, which comprises a nucleotide sequences or a partial nucleotide sequence encoding a protein that elicits an immune response therefore encodes an “antigen” as that term is used herein. Furthermore, one skilled in the art will understand that an antigen need not be encoded solely by a full length nucleotide sequence of a gene. It is readily apparent that the present invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene and that these nucleotide sequences are arranged in various combinations to encode polypeptides that elicit the desired immune response. Moreover, a skilled artisan will understand that an antigen need not be encoded by a “gene” at all. It is readily apparent that an antigen can be synthesized or can be derived from a biological sample, or can be a macromolecule besides a polypeptide. Such a biological sample can include, but is not limited to a tissue sample, a tumor sample, a cell or a fluid with other biological components.


As used herein, the terms “Chimeric Antigen Receptor” and the term “CAR” are used interchangeably. As used herein, a “CAR” is defined to be a fusion protein comprising antigen recognition moieties and cell-activation elements.


As used herein, a “CAR T-cell” or “CAR T-lymphocyte” are used interchangeably, and are defined to be a T-cell containing the capability of producing CAR polypeptide, regardless of actual expression level. For example a cell that is capable of expressing a CAR is a T-cell containing nucleic acid sequences for the expression of the CAR in the cell.


As used herein, an “effective amount” or “therapeutically effective amount” are used interchangeably, and defined to be an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result.


As used herein, an “epitope” is defined to be the portion of an antigen capable of eliciting an immune response, or the portion of an antigen that binds to an antibody. Epitopes can be a protein sequence or subsequence that is recognized by an antibody.


As used herein, an “expression vector” and an “expression construct” are used interchangeably, and are both defined to be a plasmid, virus, or other nucleic acid designed for protein expression in a cell. The vector or construct is used to introduce a gene into a host cell whereby the vector will interact with polymerases in the cell to express the protein encoded in the vector/construct. The expression vector and/or expression construct may exist in the cell extrachromosomally or integrated into the chromosome. When integrated into the chromosome the nucleic acids comprising the expression vector or expression construct will be an expression vector or expression construct.


As used herein, “heart failure” often called congestive heart failure (CHF) or congestive cardiac failure (CCF), means a condition that occurs when the heart is unable to provide sufficient pump action to maintain blood flow to meet the needs of the body. Heart failure can cause a number of symptoms including shortness of breath, leg swelling, and exercise intolerance. The condition is typically diagnosed by patient physical examination and confirmed with echocardiography. Common causes of heart failure include myocardial infarction and other forms of ischemic heart disease, hypertension, valvular heart disease, and cardiomyopathy. The term heart failure is sometimes incorrectly used for other cardiac-related illnesses, such as myocardial infarction (heart attack) or cardiac arrest, which can cause heart failure but are not equivalent to heart failure.


As used herein, “heterologous” is defined to mean the nucleic acid and/or polypeptide is not homologous to the host cell. Alternatively, “heterologous” means that portions of a nucleic acid or polypeptide that are joined together to make a combination where the portions are from different species, and the combination is not found in nature.


As used herein, the term “impaired immune function” is defined to be any reduction in immune function in an individual, as compared to a fully healthy individual. Individuals with an impaired immune function are readily identifiable by substantially increased abundance of CD8+ CD28− cells or more broadly by reduced cytokine responses, increased baseline phosphoprotein levels and other co-occurring measure.


As used herein, the term “inflammasome” is defined as cytosolic multiprotein complexes that are composed of an inflammasome-initiating sensor, apoptosis-associated speck-like protein containing a CARD (Caspase Activation and Recruitment Domain) acts as an adaptor protein and the protease-caspase-1. Inflammasome-initiating sensors include members of the NLRs the pyrin and HIN domain-containing (also known as PYHIN, Aim 2-like receptors, or ALRs; e.g., Aim2), or the TRIM (e.g., pyrin) family. Complex assembly leads to caspase-1-dependent cleavage of cytokines pro-interleukin 1β (pro-IL-1β) and pro-IL-18 into secreted mature forms. In addition, inflammasomes initiate pyroptotic cell death.


As used herein, a “single chain antibody” (scFv) is defined as an immunoglobulin molecule with function in antigen-binding activities. An antibody in scFv (single chain fragment variable) format consists of variable regions of heavy (VH) and light (VL) chains, which are joined together by a flexible peptide linker.


Immunological Age

The Jak/STAT signaling pathway is critical for meeting the multiple challenges encountered by the immune system, from fighting infections to maintaining immune tolerance. Clearly STATs are also involved in the development and function of the immune system in humans and play a key role in maintaining immune surveillance of cancer (Nature. 2007; 450(7171):903-7; Nat Rev Cancer (2009) 9:798-809).


The Jak-STAT pathway can be profoundly altered with aging and this is one major cause of immune dysfunction in older adults. A cytokine response score (CRS) can be used to predict immune decline and reduction in immune surveillance of cancer.


An inflammatory age scoring system (iAge) can also be used to predict age-associated multimorbidity and mortality. iAge can be extremely sensitive as a biomarker of cardiovascular health since elevated levels predict left ventricular remodeling and arterial stiffness even in very healthy older subject with no clinical or laboratory cardiovascular risk factors. iAge can also identify subclinical immunodeficient young patients (10% of subjects 16-35 years old) who cannot mount responses to any strain of the influenza vaccine in any of the years studied (up to 6 years follow-up). These subjects are characterized by having an older-like immunological phenotype with regards to their immune cell composition, ex vivo responses to multiple acute stimuli, and expression of gene modules associated with advanced age.


Since the cytokine response score CRS and iAge are independent measures of inflammation, diminished Jak-STAT signaling pathway in T cells, and low naïve CD8(+) T cell counts (FIG. 1A-C) these measures can be used to stratify cancer patients with respect to their clinical responses to immunotherapy. The methods described herein use blood inflammatory markers CRS and iAge to stratify cancer patients into responder and nonresponders groups for immunotherapy. The nonresponders can be treated to reduce their iAge and/or increase their CRS (and/or Jak-STAT score) so that the nonresponders obtain iAge and/or CRS (and/or Jak-STAT score) that places them into a responder group.


The procedure involves the extraction of peripheral blood samples by venipuncture, or by any appropriate method, from candidate cancer patients prior to infusion with immunotherapy treatment (FIG. 2). Immunotherapy treatment may comprise the use of certain molecules including antibodies, small molecules, etc. against inhibitory immune receptors. Blood serum is separated from blood cells by centrifugation of clogged blood, or by any other appropriate method (FIG. 2).


Construction of iAge: For serum protein determination, the resulting sera can be mixed with antibody-linked magnetic beads on 96-well filter-bottom plates and can be incubated at room temperature for 2 h followed by overnight incubation at 4° C. Room temperature incubation steps can be performed on an orbital shaker at 500-600 rpm. Plates can be vacuum filtered and washed twice with wash buffer, then incubated with biotinylated detection antibody for 2 h at room temperature. Samples can be then filtered and washed twice as above and re-suspended in streptavidin-PE. After incubation for 40 minutes at room temperature, two additional vacuum washes can be performed, and the samples can be re-suspended in Reading Buffer. Each sample can be measured in duplicate or triplicate. Plates can be read using a Luminex 200 instrument with a lower bound of 100 beads per sample per cytokine and mean fluorescence intensity (MFI) is recorded.


To derive inflammatory age (iAge) (FIG. 2), the mean fluorescence intensity can be normalized and used for multiple regression analysis, which is computed using the following regression coefficients: MIG: 0.6357, TRAIL: −0.3760, IFNG: −0.3235, EOTAXIN: 0.2912, GROA: −0.2723, IL2: −0.2063, TGFA: −0.1978, PAI1: −0.1587, LIF: −0.1587, LEPTIN: 0.1549, MIP1A: 0.1547, IL1B: 0.1471. The MFI can be multiplied by the regression coefficient for the protein, and these numbers can be all added together to give the iAge of the subject. Table 1 below lists the ranges of iAge within chronological age decades.









TABLE 1







iAge Ranges










Chronological Age (years)
iAge Range







10-19
18.1-58.3



20-29
18.5-78.9



30-39
16.9-70.2



40-49
21.5-74.1



50-59
23.1-74.4



60-69
28.1-76.6



70-79
35.6-77.6



80-96
37.9-78.5










Those markers with positive regression coefficients increased in serum concentration with age (MIG, EOTAXIN, LEPTIN, MIP1A, and IL1B) and those with negative regression coefficients decreased in serum concentration with age (TRAIL, IFNG, GROA, IL2, TGFA, PAIL and LIF).


MIG (monokine induced by gamma interferon) is a small cytokine belonging to the CXC chemokine family. MIG is one of the chemokines which plays a role to induce chemotaxis, promote differentiation and multiplication of leukocytes, and cause tissue extravasation. MIG regulates immune cell migration, differentiation, and activation. Tumor-infiltrating lymphocytes are a key for clinical outcomes and prediction of the response to checkpoint inhibitors. In vivo studies suggest the axis plays a tumorigenic role by increasing tumor proliferation and metastasis. MIG predominantly mediates lymphocytic infiltration to the focal sites and suppresses tumor growth. MIG binds to C—X—C motif chemokine 3 of the CXCR3 receptor.


TRAIL (TNF-related apoptosis-inducing ligand) is a cytokine that is produced and secreted by most normal tissue cells. It is thought to cause apoptosis primarily in tumor cells by binding to certain death receptors. TRAIL has also been designated CD253 (cluster of differentiation 253) and TNFSFlO (tumor necrosis factor (ligand) superfamily, member 10). TRAIL is described in Wiley et al Immunity 1005 3: 673-82 as well as Pitti J. Biol. Chem. 1996 271: 12687-90.


INFG (otherwise known as interferon gamma, IFNy or type II interferon) is a dimerized soluble cytokine that is the only member of the type II class of interferons. IFNG is critical for innate and adaptive immunity against viral, some bacterial and protozoan infections. INFG is an important activator of macrophages and inducer of Class II major histocompatibility complex (MHC) molecule expression. INFG is described In Schoenborn et al Adv. Immunol. 2007 96: 41-101 as well as Gray Nature. 1982 298: 859-63.


Eotaxin (also known as C—C motif chemokine I I or eosinophil chemotactic protein) is a small cytokine belonging to the CC chemokine family. Eotaxin selectively recruits eosinophils by inducing their chemotaxis, and therefore, is implicated in allergic responses. The effects of eotaxin is mediated by its binding to a G-protein-linked receptor known as a chemokine receptor. Chemokine receptors for which CCLI I is a ligand include CCR2, CCR3 and CCR5. Eotaxin is described in Kitaura et al The Journal of Biological Chemistry I 996 271: 7725-30 and Jose et al The Journal of Experimental Medicine 1994 I 79: 881-7.


GROA (also known as CXCLI, the GROI oncogene, GROa, KC, neutrophilactivating protein 3 (NAP-3) and melanoma growth stimulating activity, alpha (MSGA-a)) is secreted by human melanoma cells, has mitogenic properties and is implicated in melanoma pathogenesis. GROA is expressed by macrophages, neutrophils and epithelial cells, and has neutrophil chemoattractant activity. This chemokine elicits its effects by signaling through the chemokine receptor CXCR2. GROA is described in Haskill et al Proc. Natl. Acad. Sci. U.S.A. 190 87 (19): 7732-6.


IL-2 is one of the key cytokines with pleiotropic effects on the immune system. It is a 15.5-16 kDa protein that regulates the activities of white blood cells (leukocytes, often lymphocytes) that are responsible for immunity. The major sources of IL-2 are activated CD4+ T cells, activated CD8+ T cells, NK cells, dendritic cells and macrophages. IL-2 is an important factor for the maintenance of CD4+ regulatory T cells and plays a critical role in the differentiation of CD4+ T cells into a variety of subsets. It can promote CD8+ T-cell and NK cell cytotoxicity activity, and modulate T-cell differentiation programs in response to antigen, promoting naive CD4+ T-cell differentiation into T helper-1 (Th1) and T helper-2 (Th2) cells while inhibiting T helper-17 (Th17) differentiation.


TGFA (transforming growth factor alpha) is a polypeptide of 5.7 kDa that is partially homologous to EGF. TGFA is a growth factor that is a ligand for the epidermal growth factor receptor, which activates a signaling pathway for cell proliferation, differentiation and development. TGFA also is a potent stimulator of cell migration. TGFA can be produced in macrophages, brain cells, and keratinocytes. TGFA can induce epithelial development. TGFA can also upregulate TLR expression and function augmenting host cell defense mechanisms at epithelial surfaces. TGFA may act as either a transmembrane-bound ligand or a soluble ligand. TGFA has been associated with many types of cancers, and it may also be involved in some cases of cleft lip/palate. Alternatively spliced transcript variants encoding different isoforms have been found for this gene.


PAI1 (plasminogen activator inhibitor-1) is a member of the serine proteinase inhibitor (serpin) superfamily. PALL is the principal inhibitor of tissue plasminogen activator (tPA) and urokinase (uPA), and hence is an inhibitor of fibrinolysis. PALL is also a regulator of cell migration. PAI1 can play a role in a number of age-related, conditions including, for example, inflammation, atherosclerosis, insulin resistance, obesity, comorbidities, and Werner syndrome. PAI1 can play a host protective role during the acute phase of infection by regulating interferon gamma release. IFNG regulates PAI-1 expression, which suggests an intricate interplay between PAI-1 and IFNG. PAI1 can also activate macrophages through Toll-like receptor 4 (TLR4) and can promote migration of pro-cancer M2 macrophages into tumors.


LIF (leukemia inhibitory factor) is interleukin 6 class cytokine with pleiotropic effects impacting several different systems. When LIF levels drop, cells differentiate. LIF has the capacity to induce terminal differentiation in leukemic cells. Its activities include the induction of hematopoietic differentiation in normal and myeloid leukemia cells, the induction of neuronal cell differentiation, and the stimulation of acute-phase protein synthesis in hepatocytes. The protein encoded by this gene is a pleiotropic cytokine with roles in several different systems. It is involved in the induction of hematopoietic differentiation in normal and myeloid leukemia cells, induction of neuronal cell differentiation, regulator of mesenchymal to epithelial conversion during kidney development, and may also have a role in immune tolerance at the maternal-fetal interface. Alternatively spliced transcript variants encoding multiple isoforms have been observed for this gene. LIF functions through both autocrine and paracrine manners. LIF binds to its specific receptor LIFR, then recruits gp130 to form a high affinity receptor complex to induce the activation of the downstream signal pathways including JAK/STAT3, PI3K/AKT, ERK1/2 and mTOR signaling. Further studies have clearly proven that LIF is a multifunctional protein which has a broad biological functions in neuronal, hepatic, endocrine, inflammatory and immune systems. LIF regulates the embryonic stem cell self-renewal and is an indispensable factor to maintain mouse embryonic stem cell pluripotency. The expression of LIF is induced under inflammatory stress as an anti-inflammatory agent.


LEPTIN is secreted by white adipocytes into the circulation and plays a major role in the regulation of energy homeostasis. LEPTIN binds to the leptin receptor in the brain, which activates downstream signaling pathways that inhibit feeding and promote energy expenditure. LEPTIN also has several endocrine functions, and is involved in the regulation of immune and inflammatory responses, hematopoiesis, angiogenesis, reproduction, bone formation and wound healing. LEPTIN can directly link nutritional status and pro-inflammatory T helper 1 immune responses, and a decrease of LEPTIN plasma concentration during food deprivation can lead to an impaired immune function. LEPTIN is associated with the pathogenesis of chronic inflammation, and elevated circulating LEPTIN levels in obesity appear to contribute to low-grade inflammation which makes obese individuals more susceptible to increased risk of developing cardiovascular diseases, type II diabetes, and degenerative disease including autoimmunity and cancer. Reduced levels of LEPTIN such as those found in malnourished individuals have been linked to increased risk of infection and reduced cell-mediated immune responses. Mutations in this gene and its regulatory regions cause severe obesity and morbid obesity with hypogonadism in human patients. A mutation in this gene has also been linked to type 2 diabetes mellitus development.


MIP1A (macrophage inflammatory protein) is a member of the CC or beta chemokine subfamily. MIP1A regulates leukocyte activation and trafficking. MIP1A acts as a chemoattractant to a variety of cells including monocytes, T cells, B cells and eosinophils. MIP1A plays a role in inflammatory responses through binding to the receptors CCR1, CCR4 and CCR5.


IL-1B (Interleukin-1 beta) is a member of the interleukin 1 cytokine family. IL-1B is an important mediator of the inflammatory response, and is involved in a variety of cellular activities, including cell proliferation, differentiation, and apoptosis. LI-1B is produced by activated macrophages as a proprotein, which is proteolytically processed to its active form by caspase 1 (CASP1/ICE).


iAge predicts pulse-wave velocity (a measure of arterial stiffness, or the rate at which pressure waves move down the vessel) which correlates with cardiovascular health.


Construction of GE iAge


Gene expression measurements from were obtained for 397 subjects. For example, the Illumina Direct Hyb labeling method was used. This Illumina system performs 3′-based gene expression measurements through reverse transcription and in vitro transcription techniques that incorporate biotin-labeled nucleotides into the nascent products. The iAge was also calculated from serum protein levels for the 397 subjects. The iAge data was regressed into the gene expression data using a LASSO regression (glmnet R) of Friedman et al., J Stat Softw. 33:1-22 (2010), which is incorporated by reference in its entirety for all purposes. For the final selected genes, we filter for genes that were selected 100 out of 100 times from the regressions.


To derive gene expression inflammatory age (GE iAge), the mean gene expression signal can be normalized and used for multiple regression analysis, which is computed using the following regression coefficients: GBP5: 1.3452, MMP9: 0.6083, SIGLEC5: 0.5419, S100P: 0.4408, OLFM1: 0.4155, CISH: 0.2270, MT1A: 0.1978, CHURC1: −0.1308, IGLL1: −0.2250, RPLP0: −0.2861, SLC16A10: −0.4742, FCER1A: −0.6154, CD248: −0.6580, DDX3Y: −0.6937, MAN1A1: −0.7070. The gene expression signal can be multiplied by the regression coefficient for the gene, and these numbers can be all added together to give the GE iAge of the subject.


Table 2 below list regression coefficients for a number of other gene expression signals. One or more of these can be used for GE iAge:









TABLE 2







Regression Coefficients










Name
Coefficient














BCAT1
−1.3369



LRRC8D
−1.2064



FAM153B
−1.1522



MAN1A1
−0.9758



SLC16A10
−0.9609



DDX3Y
−0.8888



FTH1
−0.8109



ATP6V0A2
−0.7822



SRXN1
−0.7617



CYB5R1
−0.6936



FCER1A
−0.6855



CD248
−0.6440



CCR1
−0.6257



RPLP0
−0.5934



CEACAM4
−0.5823



IGLL1
−0.5557



HIST1H4C
−0.4970



CYP27A1
−0.4945



GSTT1
−0.4877



P2RX4
−0.4722



MPPE1
−0.4398



FBXL13
−0.4396



IL8
−0.4230



ERV3
−0.3961



MED9
−0.3939



CPA3
−0.3923



SLC25A29
−0.3557



HIST1H3D
−0.3533



HIST1H3H
−0.3359



LPAR5
−0.3278



D4S234E
−0.3195



ASF1A
−0.3084



AUTS2
−0.2770



FOXO3
−0.2517



SERPINE2
−0.2409



BNIP3L
−0.2386



SLC46A2
−0.2205



HP
−0.1997



CYP1B1
−0.1822



HRK
−0.1806



OASL
−0.1705



KRT23
−0.1638



KCNJ10
−0.1619



YIPF6
−0.1517



ISL2
−0.1423



CHURC1
−0.1324



PDE3B
−0.1305



LPCAT2
−0.1167



EIF2C2
−0.0968



HIST1H3F
−0.0932



STARD10
−0.0439



DAPK1
−0.0208



NUTF2
−0.0070



PNPLA2
3.9454E−05



IL17RA
0.0011



TAF8
0.00441



ZNF653
0.0049



NFKBIE
0.0192



CKAP4
0.0281



ST3GAL6
0.0315



BLMH
0.0409



LRRC25
0.0511



CISH
0.0776



SPOCD1
0.08208



OLIG1
0.08530



ACP5
0.1122



HAPLN3
0.1135



MYBPC3
0.1357



P2RY10
0.1406



GTSF1
0.1468



ITGB3
0.1535



ASPRV1
0.1562



CD79A
0.1863



PPP2R5B
0.1915



LYZ
0.2145



MMP9
0.2154



ALDH1A1
0.2308



CARD8
0.2356



ACTA2
0.2461



FOSB
0.2527



CHI3L1
0.2722



VNN 3.00
0.2885



GLT1D1
0.3497



PGM5
0.3821



MT1A
0.3828



ZP3
0.3845



ENTPD4
0.4186



CDKN1C
0.4231



BTN3A2
0.4519



PIK3C3
0.4631



EIF4G3
0.4736



S100P
0.4796



MPL
0.4890



HBZ
0.5202



LY75
0.5841



OLFM1
0.5884



ZNF696
0.6595



CSTA
0.6627



E2F1
0.6958



ORM1
0.7330



SLC39A11
0.7359



STX2
0.7364



ARF3
0.7470



SIGLEC5
0.8026



RAP1GAP
0.8326



CREBL2
0.8350



GBP5
1.4730



NCF4
1.5256



CCNG1
2.3670










Those markers with positive regression coefficients increased in gene expression with age (GBP5, MMP9, SIGLEC5, S100P, OLFM1, CISH, and MT1A) and those with negative regression coefficients decreased in gene expression with age (CHURC1, IGLL1, RPLP0, SLC16A10, FCER1A, CD248, DDX3Y, and MAN1A1).


GBP5. Guanylate Binding Protein 5. This gene belongs to the TRAFAC class dynamin-like GTPase superfamily. The encoded protein acts as an activator of NLRP3 inflammasome assembly and has a role in innate immunity and inflammation. Alternative splicing results in multiple transcript variants. Diseases associated with GBP5 include Chronic Active Epstein-Barr Virus Infection. Among its related pathways are Interferon gamma signaling and Innate Immune System. Gene Ontology (GO) annotations related to this gene include identical protein binding and GTPase activity. An important paralog of this gene is GBP3. As an activator of NLRP3 inflammasome assembly, plays a role in innate immunity and inflammation. Promotes selective NLRP3 inflammasome assembly in response to microbial and soluble, but not crystalline, agents.


MMP9. Matrix Metallopeptidase 9. The enzyme encoded by this gene degrades type IV and V collagens. Studies in rhesus monkeys suggest that the enzyme is involved in IL-8-induced mobilization of hematopoietic progenitor cells from bone marrow, and murine studies suggest a role in tumor-associated tissue remodeling. Diseases associated with MMP9 include Metaphyseal Anadysplasia 2 and Metaphyseal Anadysplasia. Among its related pathways are Regulation of Wnt-mediated beta catenin signaling and target gene transcription and Transcriptional misregulation in cancer. May play an essential role in local proteolysis of the extracellular matrix and in leukocyte migration. Could play a role in bone osteoclastic resorption. Cleaves KiSS1 at a Gly-|-Leu bond. Cleaves type IV and type V collagen into large C-terminal three quarter fragments and shorter N-terminal one quarter fragments. Degrades fibronectin but not laminin or Pz-peptide.


SIGLEC5. Sialic Acid Binding Ig Like Lectin 5. This gene encodes a member of the sialic acid-binding immunoglobulin-like lectin (Siglec) family. These cell surface lectins are characterized by structural motifs in the immunoglobulin (Ig)-like domains and sialic acid recognition sites in the first Ig V set domain. The encoded protein is a member of the CD33-related subset of Siglecs and inhibits the activation of several cell types including monocytes, macrophages and neutrophils. Binding of group B Streptococcus (GBS) to the encoded protein plays a role in GBS immune evasion. Diseases associated with SIGLEC5 include Ovarian Cystic Teratoma and Herpetic Whitlow. Among its related pathways are Innate Immune System and Class I MHC mediated antigen processing and presentation. Gene Ontology (GO) annotations related to this gene include carbohydrate binding. SIGLEC 5 can be a putative adhesion molecule that mediates sialic-acid dependent binding to cells. Binds equally to alpha-2,3-linked and alpha-2,6-linked sialic acid. The sialic acid recognition site may be masked by cis interactions with sialic acids on the same cell surface.


S100P. S100 Calcium Binding Protein P. May function as a calcium sensor and contribute to cellular calcium signaling. In a calcium-dependent manner, functions by interacting with other proteins, such as EZR and PPP5C, and indirectly plays a role in physiological processes like the formation of microvilli in epithelial cells. May stimulate cell proliferation in an autocrine manner via activation of the receptor for activated glycation end products (RAGE). Diseases associated with S100P include Geographic Tongue and Atrophic Glossitis. Among its related pathways are Innate Immune System and Ca, cAMP and Lipid Signaling. Gene Ontology (GO) annotations related to this gene include calcium ion binding and calcium-dependent protein binding. An important paralog of this gene is S100A1. This protein, in addition to binding Ca2+, also binds Zn2+ and Mg2+.


OLFM1. Olfactomedin 1. Contributes to the regulation of axonal growth in the embryonic and adult central nervous system by inhibiting interactions between RTN4R and LINGO1. Inhibits RTN4R-mediated axon growth cone collapse (By similarity). May play an important role in regulating the production of neural crest cells by the neural tube (By similarity). May be required for normal responses to olfactory stimuli (By similarity). Diseases associated with OLFM1 include Ectopic Pregnancy and Neuroblastoma. Gene Ontology (GO) annotations related to this gene include amyloid-beta binding. An important paralog of this gene is OLFM3.


CISH. Cytokine Inducible SH2 Containing Protein. The protein encoded by this gene contains a SH2 domain and a SOCS box domain. The protein thus belongs to the cytokine-induced STAT inhibitor (CIS), also known as suppressor of cytokine signaling (SOCS) or STAT-induced STAT inhibitor (SSI), protein family. CIS family members are known to be cytokine-inducible negative regulators of cytokine signaling. The expression of this gene can be induced by IL2, IL3, GM-CSF and EPO in hematopoietic cells.


MT1A. Metallothionein 1A. Proteins encoded by this gene family are low in molecular weight, are cysteine-rich, lack aromatic residues, and bind divalent heavy metal ions. The conserved cysteine residues co-ordinate metal ions using mercaptide linkages. These proteins act as anti-oxidants, protect against hydroxyl free radicals, are important in homeostatic control of metal in the cell, and play a role in detoxification of heavy metals. These proteins are transcriptionally regulated by both heavy metals and glucocorticoids. Diseases associated with MT1A include Pthirus Pubis Infestation and Lice Infestation. Among its related pathways are Platinum Pathway and Metal ion SLC transporters.


CHURC1. Churchill Domain Containing 1. Transcriptional activator that may mediate FGF signaling during neural development. Can play a role in the regulation of cell movement. Diseases associated with CHURC1 include Hypercholesterolemia, Familial, 4 and Spindle Cell Hemangioma.


IGLL1. Immunoglobulin Lambda Like Polypeptide 1. This gene encodes one of the surrogate light chain subunits that interacts with Ig mu heavy chain in the B-cell receptor. IGLL1 is a member of the immunoglobulin gene superfamily. This gene does not undergo rearrangement. Mutations in this gene can result in B cell deficiency and agammaglobulinemia, an autosomal recessive disease in which few or no gamma globulins or antibodies are made.


RPLP0. Ribosomal Protein Lateral Stalk Subunit P0. This gene encodes a ribosomal protein that is a component of the 60S subunit. The protein, which is the functional equivalent of the E. coli L10 ribosomal protein, belongs to the L10P family of ribosomal proteins. It is a neutral phosphoprotein with a C-terminal end that is nearly identical to the C-terminal ends of the acidic ribosomal phosphoproteins P1 and P2. The P0 protein can interact with P1 and P2 to form a pentameric complex consisting of P1 and P2 dimers, and a P0 monomer. SLC16A10. Solute Carrier Family 16 Member 10. SLC16A10 mediates the Na(+)-independent transport of aromatic amino acids across the plasma membrane.


FCER1A. Fc Fragment of IgE Receptor Ia. Binds to the Fc region of immunoglobulins epsilon. High affinity receptor. Responsible for initiating the allergic response. Binding of allergen to receptor bound IgE leads to cell activation and the release of mediators (such as histamine) responsible for the manifestations of allergy. The same receptor also induces the secretion of important lymphokines.


CD248. Endosialin. CD248 is a transmembrane glycoprotein that is dynamically expressed on pericytes and fibroblasts during tissue development, tumor neovascularization and inflammation. In tissue remodeling, CD248 is associated with increased stromal cell proliferation and migration. CD248 is a C-type lectin transmembrane receptor which can play a role not only in cell—cell adhesion processes but also in host defense.


DDX3Y. DEAD-Box Helicase 3 Y-Linked. DDX3Y can be an ATP-dependent RNA helicase. During immune response, may enhance IFNB1 expression via the IRF3/IRF7 pathway.


MAN1A1. Mannosidase Alpha Class 1A Member 1. MAN1A1 is a class I mammalian Golgi 1,2-mannosidase which is a type II transmembrane protein. This protein catalyzes the hydrolysis of three terminal mannose residues from peptide-bound Man(9)-GlcNAc(2) oligosaccharides. MAN1A1 is involved in the maturation of Asn-linked oligosaccharides. Progressively trims alpha-1,2-linked mannose residues from Man(9)G1cNAc(2) to produce Man(5)G1cNAc(2).


To further validate the clinical implication of the iAge score, we leveraged the data from the Framingham Heart Study, Mahmood et al. The Framingham Heart Study and the epidemiology of cardiovascular disease: a historical perspective. Lancet. 2014; 383:999-1008, a longitudinal cohort tracking thousands of individuals for decades. The gene-expression iAge (GE iAge) was significantly associated with all-causes of mortality following adjustment to multiple covariates associated with mortality, including age, gender, smoking, cholesterol levels, blood pressure, diabetes, and existence of a cardiovascular disease (p=0.02, cox proportional hazard model, N=2,290 individuals). Causes of mortality included, for example, coronary heart disease, cardiovascular disease, stroke, and cancer.


Construction of CRS: Separation of immune cells may comprise the use of differential centrifugation of blood by density gradient (FIG. 2). The resulting cell pellet can be suspended in warm media, wash twice and resuspended at 0.5×10{circumflex over ( )}6 viable cells/mL. 200 uL of cell sample can be plated per well in 96-well deep-well plates. After resting for 1 hour at 37° C., cells can be stimulated by adding 50 ul of cytokine (IFNa, IFNg, IL-6, IL-7, IL-10, IL-2, or IL-21) (FIG. 2) and incubated at 37° C. for 15 minutes. The cells can be fixed with paraformaldehyde, permeabilized with methanol, and kept at −80C overnight. Each well can then be bar-coded using a combination of Pacific Orange and Alexa-750 dyes (Invitrogen, Carlsbad, Calif.) and pooled in tubes. The cells can be washed with FACS buffer (PBS supplemented with 2% FBS and 0.1% soium azide), and stained with the following antibodies (all from BD Biosciences, San Jose, Calif.): CD3 Pacific Blue, CD4 PerCP-Cy5.5, CD20 PerCp-Cy5.5, CD33 PE-Cy7, CD45RA Qdot 605, pSTAT-1 AlexaFluor488, pSTAT-3 AlexaFluor647, pSTAT-5 PE. The samples can be washed and resuspended in FACS buffer. 100,000 cells per stimulation condition are collected using DIVA 6.0 software on an LSRII flow cytometer (BD Biosciences). Data analysis can be performed using FlowJo v9.3 by gating on live cells based on forward versus side scatter profiles, then on singlets using forward scatter area versus height, followed by cell subset-specific gating.


Fold-change difference due to stimulation can be computed as the ratio of the cell, cytokine stimulation, phosphoprotein measure to the raw, un-normalized, cell-phosphoprotein matching baseline that was measured on the same plate. The data can be normalized by scaling individual's by the average of the assay on the day in which they were measured.


To construct the Cytokine Response Score (CRS) (FIG. 2) 15 reproducible age-associated normalized cytokine responses can be expressed as fold increases over baseline (unstimulated) and the fold increases for the following can be summed: CD8+ cells, stimulate with IFNa and measure pSTAT1, 3 and 5; CD8+ cells, stimulate with IL6 and measure pSTAT1, 3 and 5, CD8+ cells, stimulate with IFNg and measure pSTAT1, CD8+ cells, stimulate with IL21 and measure pSTAT1; CD4+ cells, stimulate with IFNa and measure pSTAT5, CD4+ cells, stimulate with IL6 and measure pSTAT5, CD20+ cells, stimulate with IFNa and measure pSTAT1, Monocytes stimulate with IL10 and measure pSTAT3, Monocytes stimulate with IFNg and measure pSTAT3, Monocytes stimulate with IFNa and measure pSTAT3, and Monocytes stimulate with IL6 and measure pSTAT3.


IFNA (Interferon alpha) is a member of the type I interferon class. And has thirteen (13) variants in humans. IFNA is secreted by hematopoietic cells, predominately plasmacytoid dendritic cells. IFNA can have either protective or deleterious roles. IFNA can be induced by ssRNA, dsRNA, and cytosolic DNA from viruses or bacteria. IFNA can induce caspase-11 expression, which contributes to activation of non-canonical inflammasome. Use of recombinant IFNA has been shown to be effective in reducing the symptoms and duration of the common cold.


INFG (Interferon gamma) is a member of the type II interferon class. The encoded protein is secreted by cells of both the innate and adaptive immune systems. The active protein is a homodimer that binds to the interferon gamma receptor which triggers a cellular response to viral and microbial infections. Mutations in this gene are associated with an increased susceptibility to viral, bacterial and parasitic infections and to several autoimmune diseases.


IL6 is a cytokine with pleiotropic effects on inflammation, immune response, and hematopoiesis. IL6 is promptly and transiently produced in response to infections and tissue injuries, contributes to host defense through the stimulation of acute phase responses, hematopoiesis, and immune reactions. IL6 functions in inflammation and the maturation of B cells. In addition, IL6 has been shown to be an endogenous pyrogen capable of inducing fever in people with autoimmune diseases or infections. IL6 is primarily produced at sites of acute and chronic inflammation, where it is secreted into the serum and induces a transcriptional inflammatory response through interleukin 6 receptor, alpha. IL6 is implicated in a wide variety of inflammation-associated disease states, including susceptibility to diabetes mellitus and systemic juvenile rheumatoid arthritis. Dysregulated, continual synthesis of IL-6 plays a pathological effect on chronic inflammation and autoimmunity. Alternative splicing results in multiple transcript variants.


IL10 is a cytokine with pleiotropic effects in immunoregulation and inflammation. IL-10 is an anti-inflammatory cytokine and during infection it inhibits the activity of Th1 cells, NK cells, and macrophages, all of which are required for optimal pathogen clearance but also contribute to tissue damage. IL10 can directly regulate innate and adaptive Th1 and Th2 responses by limiting T cell activation and differentiation in the lymph nodes as well as suppressing proinflammatory responses in tissues. It also enhances B cell survival, proliferation, and antibody production. This cytokine can block NF-kappa B activity, and is involved in the regulation of the JAK-STAT signaling pathway. Knockout studies in mice suggested the function of this cytokine as an essential immunoregulator in the intestinal tract.


IL21 is a member of the common-gamma chain family of cytokines with immunoregulatory activity. IL21 plays a role in both the innate and adaptive immune responses by inducing the differentiation, proliferation and activity of multiple target cells including macrophages, natural killer cells, B cells, cytotoxic T cells, and epithelial cells. IL21 is important to anti-tumor and antiviral responses and also exerts major effects on inflammatory responses that promote the development of autoimmune diseases and inflammatory disorders.


pSTAT1 (phosphorylated signal transducer and activator of transcription 1) mediates cellular responses to interferons (IFNs), cytokine KITLG/SCF and other cytokines and other growth factors. Following type I IFN (IFN-alpha and IFN-beta) binding to cell surface receptors, signaling via protein kinases leads to activation of Jak kinases (TYK2 and JAK1) and to tyrosine phosphorylation of STAT1 and STAT2. The phosphorylated STATs dimerize and associate with ISGF3G/IRF-9 to form a complex termed ISGF3 transcription factor, that enters the nucleus (PubMed:28753426). ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of IFN-stimulated genes (ISG), which drive the cell in an antiviral state. In response to type II IFN (IFN-gamma), STAT1 is tyrosine- and serine-phosphorylated (PubMed:26479788). It then forms a homodimer termed IFN-gamma-activated factor (GAF), migrates into the nucleus and binds to the IFN gamma activated sequence (GAS) to drive the expression of the target genes, inducing a cellular antiviral state.


pSTAT 3 (phosphorylated signal transducer and activator of transcription 3) mediates cellular responses to interleukins, KITLG/SCF, LEP and other growth factors. Once activated, recruits coactivators, such as NCOA1 or MED1, to the promoter region of the target gene. Binds to the interleukin-6 (IL-6)-responsive elements identified in the promoters of various acute-phase protein genes. Activated by IL31 through IL31RA. Acts as a regulator of inflammatory response by regulating differentiation of naive CD4+ T-cells into T-helper Th17 or regulatory T-cells (Treg): deacetylation and oxidation of lysine residues by LOXL3, disrupts STAT3 dimerization and inhibits its transcription activity.


pSTAT 5 (phosphorylated signal transducer and activator of transcription 5) is activated by Janus-activated kinases (JAK) downstream of cytokine receptors. STAT5 proteins are activated by a wide variety of hematopoietic and nonhematopoietic cytokines and growth factors, all of which use the JAK-STAT signaling pathway as their main mode of signal transduction. STAT5 proteins critically regulate vital cellular functions such as proliferation, differentiation, and survival. STAT5 plays an important role in the maintenance of normal immune function and homeostasis, both of which are regulated by specific members of IL-2 family of cytokines, which share a common gamma chain (γ(c)) in their receptor complex. STAT5 critically mediates the biological actions of members of the γ(c) family of cytokines in the immune system. Essentially, STAT5 plays a critical role in the function and development of Tregs, and consistently activated STAT5 is associated with a suppression in antitumor immunity and an increase in proliferation, invasion, and survival of tumor cells.


Immunotypes

Five markers which are significant contributors to iAge were used in deriving immunotypes. The five markers are Eotaxin, GroA, INFg, MIG, and TRAIL. Using information from patients, the levels of these markers were subjected to a Principal Component Analysis. In a PCA, the levels of the five (5) markers can be standardized so that each contributes equally. This standardized data can then be subject to a covariance matrix computation to see if some of the variables are behaving in a correlated fashion. Eigenvectors and Eigenvalues can be calculated for the covariance matrix to identify the Principle Components. Principal components are new variables that are constructed as linear combinations or mixtures of the initial variables. Principal Components represent the directions of the data that explain a maximal amount of variance, that is to say, the lines that capture most information of the data. Components of lesser value can be discarded and more significant components can be kept (reducing the dimensionality of the data set). This data can be recast along the Principal Component axes.


The PCA of the patient data produced two groups: a super-healthy group and a normal health group. The super healthy group was divided into four immunotypes 1-4 which are shown in FIG. 6. In FIG. 6, the level of each protein marker was standardized (50 percentile=0) and minus 4 was the level of marker in patients with an iAge 20 years younger than their chronological age. Immunotype SH1 had low iAge score levels for Eotaxin, GroA, INFg, and MIG, but high iAge score levels of TRAIL. Immunotype SH2 had low iAge scores for Eotaxin and TRAIL, high iAge score levels of GroA and INFg, and moderate iAge score levels for MIG. Immunotype SH3 had low iAge score levels for all five markers (Eotaxin, GroA, IFNg, MIG, TRAIL). Immunotype SH4 had low iAge scores for GroA, IFNg, and TRAIL, high iAge score levels of MIG, and moderate iAge score levels for Eotaxin.


The normal group was divided into six Immunotypes 1-6 which are shown in FIG. 7. In FIG. 7, the level of each protein marker was standardized (50 percentile=0) and minus 4 was the level of marker in patients with an iAge 20 years younger than their chronological age. In addition, two additional components were added for the PCA of the normal group: number of markers in the minus direction (low iAge), and the difference between iAge and chronological age. Immunotype N1 had negative iAge score levels for Eotaxin and TRAIL, and high iAge score levels for GroA, IFNg and MIG. Immunotype N2 had moderate iAge score levels for Eotaxin, GroA, IFNg, and MIG, and high iAge score levels for TRAIL. Immunotype N3 had moderate iAge score levels for MIG and TRAIL, and high iAge score levels for Eotaxin, GroA, and IFNg. Immunotype N4 had moderate iAge score levels for Eotaxin and MIG, and high iAge score levels for GroA, IFNg, and TRAIL. Immunotype N4 had moderate iAge score levels for MIG and TRAIL, and high iAge score levels for Eotaxin, GroA, and IFNg. Immunotype N5 had low iAge score levels for TRAIL, and high iAge score levels for GroA, IFNg, MIG, and TRAIL. Immunotype N6 had moderate iAge score levels for GroA and IFNg, and high iAge score levels for Eotaxin, MIG, and TRAIL.



FIGS. 8 and 9 show that each of the ten immunotypes has a different signature for the five markers (different levels of the five markers after the markers are normalized as discussed above, i.e., normalized so zero=50th percentile and 4=level in patients with an iAge 20 years younger than their chronological age). Thus, a common treatment of each immunotype can be provided to members of the immunotype to lower the iAge of any member of the immunotype which will lower iAge and improve the patients health, well-being and longevity.


Construction of Cardiac Age.


To derive Cardiac age (cAge), patient samples are obtained and processed similar to the description above for iAge. The mean fluorescence intensity can be normalized and used for multiple regression analysis. The levels of MIG, LIF and SIRT3 are diagnostic for risk of cardiovascular health. Other parameters that can be used to compute cardiac age include, for example, aortic pulse wave velocity, a measure of vascular stiffness; relative wall thickness (RWT), a measure of ventricular remodeling, and early diastolic mitral annular velocities (e′), a measure of ventricular relaxation. Still other parameters include, for example, sex, BMI, heart rate, systolic blood pressure, fasting glucose and total cholesterol to HDL ratio. The levels of MIG, LIF, SIRT3, and/or other measurements for a subject can be compared to those of other subjects of the same age and/or different ages to determine the quantile of the subject for each factor, or for the factors of subjects of different ages. Low quantile rank for MIG is diagnostic for low risk of cardiovascular disease, and high quantile rank for MIG is diagnostic for a higher risk of cardiovascular disease. High quantile rank for LIF and/or SIRT3 are diagnostic for low risk of cardiovascular disease, and high quantile rank for LIF and/or SIRT3 are diagnostic for a higher risk of cardiovascular disease. Other parameters (factors) can also be included in the analysis and, for example, high quantile rank for pulse wave velocity are diagnostic for a higher risk of cardiovascular disease, high quantile rank for abnormal RWT are diagnostic for a higher risk of cardiovascular disease, and lower quantile early diastolic mitral annular velocities are also diagnostic for higher risk of cardiovascular disease. Multiple parameters and/or factors can be combined to compute cardiac age, for example, MIG, LIF and SIRT3 can be used to derive cardiac age, or these factors can be combined with other parameters (e.g., aortic pulse wave velocity, RWT, and/or early diastolic mitral annular velocities) to derive cardiac age. When these factors and/or parameters are combined, high quantile rank will correlate with older cardiac age and a higher risk of cardiovascular disease, and a low quantile rank will correlate with younger cardiac age and a lower risk of cardiovascular disease. Quantile ranks can include, for example, quartiles, quintiles or deciles.


SIRT3 (Situin-3, a NAD-dependent deacetylase) is member of the mammalian sirtuin family of proteins, which are homologs to the yeast Sir2 protein. SIRT3 exhibits NAD+-dependent deacetylase activity. SIRT3 is a regulator of the mitochondrial adaptive response to stress, such as metabolic reprogramming and antioxidant defense mechanisms. SIRT3 mediates cellular resistance toward various forms of stress by maintaining genomic stability and mitochondrial integrity. SIRT3 is central to the maintenance of appropriate mitochondrial function by limiting oxidative stress, and reducing reactive oxygen species (ROS) production with a decrease in mitochondrial membrane potential. SIRT3 has cardio-protective properties involved in mitochondrial homeostasis, stem cell and tissue maintenance during aging, and linked to the beneficial effects of diet, caloric restriction and exercise in maintaining cardiovascular health and longevity.


MIG positively correlates with cardiovascular aging markers PWV (R=0.22), a measure of arterial stiffness, and RWT (R=0.3), a measure of cardiac remodeling; and a negative correlation between LIF and PWV (R=−0.27), and RWT (R=−0.22). Subclinical cardiac tissue remodeling and increased arterial stiffness can be found in otherwise healthy individuals with elevated levels of MIG and low levels of LIF.


Patients with subclinical cardiac tissue remodeling and increased arterial stiffness can be otherwise healthy individuals who have elevated levels of MIG and low levels of SIRT-3 and LIF. Cardiac tissue remodeling and increased arterial stiffness are risk factors associated with poorer outcomes in cardiovascular disease. The largest contributor to the inflammatory clock, MIG, was positively correlated with subclinical levels of arterial stiffness and cardiac remodeling even in healthy older adults with no clinical or laboratory evidence of cardiovascular disease. The inflammatory clock (iAge) can also be used as an early molecular marker for cardiovascular malfunctioning.


At least two sources of MIG-mediated inflammation can ensue with aging based on our findings; one that is age-intrinsic and observed in aging endothelia, and one independent of age (likely as a response to cumulative exposure to environmental insults). In contrast, there was no significant correlation between known disease risk factors (BMI, smoking, dyslipidemia) and the levels of MIG gene or protein expression. MIG overproduction can be caused by cellular aging per-se, which triggers metabolic dysfunction with production of damage-associated molecular patterns (DAMPs) such as adenine and N4-acetylcytidine. These DAMPs can then act through the inflammasome machinery, such as NLRC4, to regulate multiple inflammatory signals including IL-1β and MIG.


Endothelium has a critical role in the etiology of hypertension and arterial stiffness, and more advanced signs of cardiovascular aging such as tissue remodeling and cardiac hypertrophy are often preceded and may be initiated by malfunctioning of aged endothelia. Endothelial cells show a time-dependent increase in MIG transcript levels, which was concomitant with a drop in SIRT3 expression, and with a decrease in the number of vascular networks formed by the endothelial cells. Young endothelia is a target of MIG from other sources, and MIG can down-regulate SIRT3 expression in the endothelial cells. In addition, endothelia cells made from hiPSC (human, induced pluripotent stem cells) but not cardiomyocytes made from hiPSC, express CXCR3 he receptor for MIG. MIG can act both in a paracrine fashion, wherein increasing levels of this chemokine from immune sources affect endothelial cell function, and in an autocrine fashion on endothelial cells likely producing a positive feedback loop where increasing doses of MIG and expression of its receptor in these cells leads to cumulative deterioration of endothelial function in aging. Exposure of endothelial cells to MIG can also reduce the endothelial cell's capacity for forming tubular networks, and MIG can reduce vasorelaxation in the aorta.


Immunosenescence

The immune system undergoes marked shifts in composition and function with aging, a pattern of changes that are together termed “immunosenescence”.


Immunosenescence impacts both the innate and adaptive arms of the immune system and major features of immunosenescence include alteration in immune cell subset frequencies, defective antigen presentation, reduced cytotoxic function, and restricted T cell repertoire (Pawelec G, Larbi A. (2008), Immunity and ageing in man: Annual Review 2006/2007. Exper Gerontol 43:34-38; Weiskopf D, Weinberger B, GrubeckLoebenstein B. (2009), The aging of the immune system. Transpl Int. 22:1041-1050, both of which are incorporated by reference in their entirety for all purposes).


Other changes occurring during immunosenescence include reduction in cytokine signaling responses, increased baseline levels of phospho-STAT, and elevation in memory cell populations (Shen-Orr et al. (2016). Defective Signaling in the JAK-STAT Pathway Tracks with Chronic Inflammation and Cardiovascular Risk in Aging Humans, Cell Systems. 3(4):374-384.E4, which is incorporated by reference in its entirety for all purposes). Of notable clinical importance, immunosenescence also results in defects in antibody responses, with many older individuals failing to generate protective antibody titers following vaccination (Furman D et al. (2013). Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Molecular Systems Biology 9:659, which is incorporated by reference in its entirety for all purposes).


Immunosenescence impacts both the host's capacity to respond to infections and the development of long-term immune memory, especially by vaccination. Immunosenescence is associated with the accumulation of memory and effector cells as a result of repeated infections and by continuous exposure to antigens (inhalant allergens, food, etc.). This chronic inflammation characterizes immunosenescence and can have a significant impact on survival and fragility. Immunosenescence can also be associated with remodeling of the immune system caused by oxidative stress.


Immunosenescence can occur from an imbalance between inflammatory and antiinflammatory mechanisms producing chronic inflammation. This chronic inflammation can be due to chronic antigen stimulation occurring over the course of life and to the oxidative stress that involves the production of oxygen free radicals and toxic products. These factors are able to modify the potential of apoptotic lymphocytes, and this remodeling of the lymphocyte compartment and the chronic expression of proinflammatory cytokines are implicated in the processes of longevity and diseases related to immunosenescence.


Canonical acute inflammation proteins (C-reactive protein, Interleukin-6, etc.) have been associated with immunosenescence in previous studies, but the relationship with systemic chronic inflammation has not yet been established. Using a well-known marker for immunosenescence (the frequency of naïve CD8 (+) T cells) contribution of iAge to immunosenescence was estimated after controlling for Age, CMV, and sex by a multiple regression model. Age was the strongest contributor to changes in naïve CD8 (+) T cells followed by iAge, CMV (negative contributors) and sex (frequency of total CD8 (+) T cells in females was 24% vs. 30% in males). iAge was significantly correlated with the frequency of naïve CD8 (+) T cells to a similar extent to CMV positivity. Chronological age was the strongest contributor (P<10−15), followed by iAge (P<10−5), CMV (P<10−3) and gender (P=0.012) (A).


The effect of chronic inflammation on the immune response was also measured using a functional immune assay (phospho-flow) in which cells are stimulated ex vivo and the phosphorylation of various intracellular proteins is measured by using antibodies against phosphorylated forms of these proteins. In particular, the responses to four independent stimuli (Interferon-alpha, Interleukin-6, Interleukin-10 and Interleukin-21) were measured in a total of 818 individuals and the fold-increase in phospho-STAT1, -STAT3 and -STAT5 in B cells, total CD4 (+) T cells (and the CD45RA(+) and CD45RA(−) subsets), total CD8 (+) T cells (and the CD45RA(+) and CD45RA(−) subsets), and in monocytes were determined. Multiple regression analysis controlling age, CMV and sex, surprisingly showed there was a general decrease of the B cell and T cell responses to stimuli and an overall potentiation of the monocyte responses associated with increasing iAge (combined P<10−5).


These results demonstrate that iAge is an important immune predictor of immune function decline (immunosenescence) and can be used as a ‘metric’ for immunological health.


Immunosenescence is associated with lowered ability of the immune system to kill cancer cells, protect against infections from pathogenic organisms, and produce efficacious response to vaccines. Treating a subject to lower their iAge can reduce the immunosenescence in the subject and improve the ability of the subject's immune system to kill cancer cells, protect against infections from pathogenic organisms, and produce efficacious responses to vaccines. Agents and methods for lowering iAge and thereby reducing immunosenescence are described below.


Immunotherapies

In recent years, there has been a sharp rise in the development and implementation of cancer immunotherapies against cancer. The approval of anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and anti-programmed cell death protein 1 (PD-1) antibodies has resulted in significant improvements in disease outcomes for a variety of cancers. Unlike chemo- and radiotherapy, which aim to interfere with tumor cell growth and survival, immunotherapies indirectly target the tumor by boosting the anti-tumor immune responses of the patient. Despite the huge success of these therapies in many forms of cancer, the success rates are generally low and biomarkers to define objective clinical responses are still lacking.


The biological foundation of current cancer immunotherapies is the concept of cancer immune surveillance, which proposes that the immune system eliminates tumor cells because these possess new antigens and trigger an immune reaction with regression of the tumor and no clinical signs of its existence. In a seminal study to test this theory, it was shown that chemically-induced sarcomas grew faster and more aggressively in immune-incompetent mice than in wild type animals because the former lack lymphocytes (have a engineered mutation of the recombination-activating (RAG) gene) or their inability to respond to IFNγ, either because of the loss of the IFNγ receptor gene, or the STAT-1 gene. Immune surveillance was shown by using mice with a double mutation RAG2−/−/STAT1−/− which spontaneously developed tumors. These tumors resemble some of the major malignancies of humans, such as breast, lung, or colon. Cancer immunoediting was shown by transplanting tumors between mice. When a tumor was transplanted from an immune incompetent mouse to a immune competent mouse, 40% of the tumors were rejected. Whereas no rejection occured when transplants were performed using tumors from syngeneic immune-competent mice. This clearly demonstrated that immunoediting had occurred in immune-competent animals, even if they were incapable of rejecting their own tumor, enabling their escape from immune-surveillance. After decades of follow-up work a novel theory of tumor immunity was introduced. The theory proposed three steps: 1) elimination of tumors at early stages (immune surveillance hypothesis), 2) equilibrium which refers to the state in which the immune system controls the tumor, and 3) escape when tumor cells are immune-edited and grow without immune control. This three E's theory is still the theory accepted worldwide as the basis to understand the interaction of cancer cells with the immune system. The theory also paved the way for the exploding field of cancer immunotherapy.


Immunotherapy for cancer boosts the body's natural defenses to fight cancer. It uses substances made by the body or in a laboratory to improve or restore immune system function. Cancer immunotherapies include, for example, monoclonal antibodies, immune checkpoint inhibitors, cancer vaccines, immune cells modified with, for example, chimeric antigen receptors, and other nonspecific immunotherapies that boost the immune system function or action by, for example, specifically targeting cancer cells, overcoming inhibition of the immune system (e.g., by myeloid suppressor cells), etc.


Monoclonal antibodies for treating cancer include, for example, anti-CD20 antibody (e.g., Bexxar®, Zevalin®, Rituxan®, Gazyvaro®, Arzerra®), anti-Her2 antibody (e.g., Herceptin®, Kadcyla®, Perjeta®), anti-CD30 antibody (e.g., Adcetris®), anti-CD19 and anti-CD3 bispecific antibody (e.g., Blincyto®), anti-VegF antibody (e.g., Avastin®, Cyramza®), anti-EGFR antibody (e.g., Erbitux®, Portrazza®, Vectibix®), anti-PDGFR-α antibody (e.g., Lartruvo®), anti-CD38 antibody (e.g., Darzalex®), antiSLAMF7 antibody (e.g., Empliciti®), anti-GD2 antibody (e.g., Unituxin®), anti-CD19 antibody (e.g., Blincyto®), anti-RANKL antibody (e.g., Xgeva®, Prolia®), anti-EpCAM and anti-CD3 antibody (e.g., Removab®), anti-EpCAM antibody (e.g., Proxinium®), anti-CD52 antibody (e.g., Campath®), and anti-CD33 antibody (e.g., Mylotarg®).


Checkpoint inhibitors for treating cancer include, for example, Nivolumab (Opdivo), Pembrolizumab (Keytruda), Atezolizumab (Tecentriq), Ipilimumab (Yervoy), Durvalumab (Imfinzi®), Avelumab (Bavencio®), Lirilumab, and BMS-986016 (Relatlimab). Nivolumab, Atezolizumab, Pembrolizumab, Durvalumab, and Avelumab act at the checkpoint protein PD-1/PD-L1 and inhibit apoptosis of anti-tumor immune cells. Ipilimumab acts at CTLA4 and prevents CTLA4 from downregulating activated Tcells in the tumor. Lirilumab acts at KIR and facilitates activation of Natural Killer cells. BMS-986016 acts at LAG3 and activates antigen-specific T-lymphocytes and enhances cytotoxic T cell-mediated lysis of tumor cells.


Chimeric Antigen Receptors for treating cancer include, for example, an antiCD19 CAR in T-cells (e.g., Kymriah® and Yescarta®). CAR therapy can also be directed at a variety of tumor-associated antigens including, for example, 4-1BB, 5T4, adenocarcinoma antigen, alpha-fetoprotein, BAFF, B-lymphoma cell, C242 antigen, CA125, carbonic anhydrase 9 (CA-IX), C-MET, CCR4, CD152, CD19, CD20, CD21, CD22, CD23 (IgE receptor), CD28, CD30 (TNFRSF8), CD33, CD4, CD40, CD44 v6, CD51, CD52, CD56, CD74, CD80, CEA, CNT0888, CTLA-4, DR5, EGFR, EpCAM, CD3, FAP, fibronectin extra domain-B, folate receptor 1, GD2, GD3 ganglioside, glycoprotein 75, GPNMB, HER2/neu, HGF, human scatter factor receptor kinase, IGF-1 receptor, IGF-I, IgG1, L1-CAM, IL-13, IL-6, insulin-like growth factor I receptor, alpha 5β1integrin, integrin αvβ3, MORAb-009, MS4A1, MUC1, mucin CanAg, Nglycolylneuraminic acid, NPC-1C, PDGF-Rα, PDL192, phosphatidylserine, prostatic carcinoma cells, RANKL, RON, ROR1, SCH 900105, SDC1, SLAMF7, TAG-72, tenascin C, TGF β2, TGF-β, TRAIL-R1, TRAIL-R2, tumor antigen CTAA16.88, VEGFA, VEGFR-1, VEGFR2, 707-AP, ART-4, B7H4, BAGE, β-catenin/m, Bcr-abl, MN/C IX antibody, CAMEL, CAP-1, CASP-8, CD25, CDC27/m, CDK4/m, CT, Cyp-B, DAM, ErbB3, ELF2M, EMMPRIN, ETV6-AML1, G250, GAGE, GnT-V, Gp100, HAGE, HLA-A*0201-R170I, HPV-E7, HSP70-2M, HST-2, hTERT (or hTRT), iCE, IL-2R, IL5, KIAA0205, LAGE, LDLR/FUT, MAGE, MART-1/melan-A, MART-2/Ski, MC1R, myosin/m, MUM-1, MUM-2, MUM-3, NA88-A, PAP, proteinase-3, p190 minor bcr-abl, Pml/RARα, PRAIVIE, PSA, PSM, PSMA, RAGE, RU1 or RU2, SAGE, SART-1 or SART-3, survivin, TPI/m, TRP-1, TRP-2, TRP-2/INT2, WT1, NY-Eso-1 or NY-Eso-B or vimentin.


Cancer vaccines include, for example, human papilloma virus (HPV) vaccine, dendritic cell vaccines (e.g., Provenge® for prostate cancer), tumor cell vaccines, antigen vaccines, oncolytic virus vaccines (e.g., Imlygic™), Non-Hodgkin's lymphoma and mantle cell lymphoma vaccine (e.g., BioVaxID™), breast cancer vaccine (e.g., Neuvax™), brain cancer vaccine (e.g., DCVax™, CDX-110™), pancreatic cancer vaccine (e.g., GVAX Pancreas, HyperAcute™ Pancreas), colorectal cancer vaccine (e.g., Imprime PGG®), bladder cancer vaccine (e.g., BCG™), solid tumor vaccine (e.g., OK432™), lung cancer and gastrointestinal cancer vaccine (e.g., PSK™), cervical cancer vaccine (e.g., Schizophyllan™), and stomach cancer vaccine (e.g., Lentinan™).


Other immunotherapies for treating cancer include, for example, an IL-2 diphtheria toxin fusion protein (e.g., Ontak®),


Types of Cancer

Cancers that can be treated with the methods described herein, include, for example, the approved indications for the FDA approved immunotherapies, such as melanoma, non-small cell lung cancer, Head and Neck squamous cell cancer, classical Hodgkin's lymphoma, primary mediastinal large B-cell lymphoma, urothelial carcinoma, microsatellite instability-high cancer, gastric cancer, cervical cancer, hepatocellular carcinoma, Merkel Cell carcinoma, renal cell carcinoma (Keytruda®); advanced or metastatic urothelial carcinoma, unresectable, stage III non-small cell lung cancer (Imfinzi®); unresectable or metastatic melanoma, metastatic non-small cell lung cancer, advanced renal cell carcinoma, classical Hodgkin's lymphoma, recurrent or metastatic squamous cell carcinoma, advanced or metastatic urothelial carcinoma, microsatellite instability high, or mismatch repair deficient metastatic colorectal cancer, hepatocellular carcinoma (Opdivo®); urothelial carcinoma, non-small cell lung cancer, triple negative breast cancer, small cell lung cancer (Tecentriq®); metastatic Merkel cell carcinoma (Bavencio®); unresectable of metastatic melanoma, advanced renal cell carcinoma, microsatellite instability high, or mismatch repair deficient metastatic colorectal cancer (Yervoy®); refractory diffuse B-cell lymphoma, relapsed or refractory acute lymphoblastic leukemia (Kymriah®); or diffuse large B-cell lymphoma, primary mediastinal B-cell lymphoma, High grade B-cell lymphoma (Yescarta®).


Cancers that can be treated with the methods described herein, also include, for example the indications under development such as, acute myeloid leukemia, bladder cancer, squamous cell carcinoma of the head and neck, chronic lymphocytic leukemia, multiple myeloma, metastatic solid malignancies (Lirilumab™); or melanoma, advanced colorectal cancer, advanced Chordoma, metastatic melanoma, gastro/esophageal cancer, solid tumors, gastric cancer, advanced renal cell cancer, advanced non-small cell lung cancer (Relatlimab™).


Other cancers that can be treated with the methods herein include, for example, sarcoma, carcinoma, melanoma, chordoma, malignant histiocytoma, mesothelioma, glioblastoma, neuroblastoma, medulloblastoma, malignant meningioma, malignant schwannoma, leukemia, lymphoma, myeloma, myelodysplastic syndrome, myeloproliferative disease. In some embodiments, the cancer is a leukemia, lymphoma, myeloma, myelodysplastic syndrome, and/or myeloproliferative disease.


Vaccines

Vaccines can be substances used to stimulate a protective immune response in a subject (e.g., an antibody response or a cell mediated response) and provide immunity against one or several diseases. Vaccines protect against more than many debilitating or life-threatening diseases/infectious agents, including for example, adenovirus, anthrax, cervical cancer, chicken pox, cholera, dengue, diphtheria, Haemophilus influenza, hepatitis A, hepatitis B, hepatitis E, HPV, influenza, Japanese encephalitis, malaria, measles, meningitis, meningococcal (MenACWY), serogroup B meningococcal, mumps, pneumococcus, polio, rabies, rotavirus, rubella, shingles, small pox, tetanus, tuberculosis, typhoid, varicella, whooping cough, and yellow fever.


Vaccines can be prepared from the causative agent of a disease, its products, or a synthetic substitute, treated to act as an antigen without inducing the disease. Examples of vaccine types include, for example, live or attenuated vaccines (e.g., measles, mumps, rubella, varicella, influenza, coronavirus, rotavirus, zoster, yellow fever), inactivated or killed vaccines (e.g., polio, hepatitis A, rabies), toxoid (inactivated toxoid) vaccines (e.g., diphtheria, and tetanus), and subunit/conjugate vaccines (e.g., hepatitis B, influenza, coronavirus, Haemophilus influenza type b, pertussis, pneumococcal, meningococcal, HPV).


Attenuated vaccines can be made in several different ways. Some of the most common methods involve passing the disease-causing virus through a series of cell cultures or animal embryos (typically chick embryos). Using chick embryos as an example, the virus is grown in different embryos in a series. With each passage, the virus becomes better at replicating in chick cells, but loses its ability to replicate in human cells. A virus targeted for use in a vaccine may be grown through—“passaged” through—upwards of 200 different embryos or cell cultures. Eventually, the attenuated virus will be unable to replicate well (or at all) in human cells, and can be used in a vaccine. All of the methods that involve passing a virus through a non-human host produce a version of the virus that can still be recognized by the human immune system, but cannot replicate well in a human host. When the resulting vaccine virus is given to a human, it will be unable to replicate enough to cause illness, but will still provoke an immune response that can protect against future infection.


Killed or inactivated vaccines can be created by inactivating a pathogen, typically using heat or chemicals such as formaldehyde or formalin. This destroys the pathogen's ability to replicate, but keeps it “intact” so that the immune system can still recognize it. (“Inactivated” is generally used rather than “killed” to refer to viral vaccines of this type, as viruses are generally not considered to be alive.) Because killed or inactivated pathogens can't replicate at all, they can't revert to a more virulent form capable of causing disease (as discussed above with live, attenuated vaccines). However, these vaccines tend to provide a shorter length of protection than live vaccines, and are more likely to require boosters to create long-term immunity.


Immunizations created using inactivated toxins are called toxoids. Toxoids can actually be considered killed or inactivated vaccines, but are sometimes given their own category to highlight the fact that they contain an inactivated toxin, and not an inactivated form of bacteria.


Both subunit and conjugate vaccines contain only pieces of the pathogens they protect against. Subunit vaccines use only part of a target pathogen to provoke a response from the immune system. This may be done by isolating a specific protein from a pathogen and presenting it as an antigen on its own. The acellular pertussis vaccine and influenza vaccine (in shot form) are examples of subunit vaccines. Another type of subunit vaccine can be created via genetic engineering. Conjugate vaccines can be made using a combination of two different components. Conjugate vaccines, however, are made using pieces from the coats of bacteria. These coats are chemically linked to a carrier protein, and the combination is used as a vaccine.


Pathogens

Pathogenic organisms are capable of causing disease in a subject. A human pathogen is capable of causing illness in humans. Common examples of pathogenic organisms include specific strains of bacteria such as, for example, Actinomyces israelii, Bacillus anthracis, Bacteroides fragilis, Bordetella pertussis, Borrelia, Brucella, Campylobacter jejuni, Chlamydophila psittaci, Corynebacterium diphtheria, Ehrlichia, Enterococcus, Francisella tularensis, Haemophilus influenza, Helicobacter pylori, Klebsiella pneumoniae, Legionella pneumophila, Leptospira species, Listeria monocytogenes, Mycobacterium, Mycoplasma pneumoniae, Pseudomonas aeruginosa, Nocardia asteroids, Rickettsia rickettsia, Salmonella, Shigella, Treponema pallidum, Vibrio cholera, Yersinia pestis, Listeria E. coli, Staphylococcus, Streptococcus, Neisseria, Clostridia, Chlamydia, mycoplasmas.


Pathogenic organisms also include viruses such as, for example, adenoviruses, herpesviruses, influenza, coronavirus, hepatitis, poxviruses, papovaviruses, paramyxoviruses, coronaviruses, picornaviruses, Reoviruses, togaviruses, flaviviruses, arenaviruses, rhabdoviruses, retroviruses, hepadnaviruses, Cryptosporidium.


Pathogenic organisms include fungi such as, for example, Candida, Aspergillus, Cryptococcus, Histoplasma, Pneumocystis, and Stachybotrys.


Pathogens also include the above organisms which are the target of vaccines.


Anti-pathogen therapies can include, for example, antibiotics for bacterial pathogens, anti-viral therapies for viral pathogens, and anti-fungal therapies for fungal pathogens. Antibodies can also be administered for the treatment of certain infectious diseases caused by bacteria, viruses or fungi.


Cancer Treatments Using iAge


Subjects with cancer who are candidates for immunotherapy (as described above) have their blood drawn and an iAge and CRS are calculated as described above. If the subject's iAge places them in the youngest iAge quartile for their age group (see Table 1) they can be classified as responders and move forward with the immunotherapy. If the subject's iAge places them in the middle two quartiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be treated with the immunotherapy. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quartile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quartile.


Alternatively, if the subject's iAge places them in the youngest iAge quintile for their age group (see Table 1) they can be classified as responders and move forward with the immunotherapy. If the subject's iAge places them in the middle three quintiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be treated with the immunotherapy. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quintile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quintile.


Still alternatively, if the subject's iAge places them in the youngest iAge tertile for their age group (see Table 1) they can be classified as responders and move forward with the immunotherapy. If the subject's iAge places them in the middle tertile, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be treated with the immunotherapy. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest tertile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge tertile.


Subjects who are candidates for vaccinations (e.g., the elderly) can have their blood drawn and an iAge and CRS are calculated as described above. If the subject's iAge places them in the youngest iAge quartile for their age group (see Table 1) they can be classified as responders and move forward with the vaccination. If the subject's iAge places them in the middle two quartiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be vaccinated. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quartile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quartile. Subjects classified as nonresponders can also be treated with higher doses of vaccines and/or more aggressive vaccine formulations (e.g., cocktails of antigens, adjuvants, and/or immunostimulants) to account for the immunosenescence in the subject.


Alternatively, if the subject's iAge places them in the youngest iAge quintile for their age group (see Table 1) they can be classified as responders and move forward with the vaccination. If the subject's iAge places them in the middle three quintiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be vaccinated. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quintile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quintile. Subjects classified as nonresponders can also be treated with higher doses of vaccines and/or more aggressive vaccine formulations (e.g., cocktails of antigens, adjuvants, and/or immunostimulants) to account for the immunosenescence in the subject.


Still alternatively, if the subject's iAge places them in the youngest iAge tertile for their age group (see Table 1) they can be classified as responders and move forward with the vaccination. If the subject's iAge places them in the middle tertile, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can be vaccinated. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest tertile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge tertile. Subjects classified as nonresponders can also be treated with higher doses of vaccines and/or more aggressive vaccine formulations (e.g., cocktails of antigens, adjuvants, and/or immunostimulants) to account for the immunosenescence in the subject.


Subjects who have been exposed to a pathogenic organism, are infected with a pathogenic organism, and/or are susceptible to infection by a pathogenic organism can have their blood drawn and an iAge and CRS are calculated as described above. If the subject's iAge places them in the youngest iAge quartile for their age group (see Table 1) they can be classified as responders and move forward with standard treatment for the pathogenic organism. If the subject's iAge places them in the middle two quartiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can move forward with standard treatment for the pathogenic organism. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quartile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quartile. Subjects classified as nonresponders can also be treated with more aggressive therapies and/or higher doses of therapeutics to account for the immunosenescence in the subject.


Alternatively, if the subject's iAge places them in the youngest iAge quintile for their age group (see Table 1) they can be classified as responders and move forward with standard treatment for the pathogenic organism. If the subject's iAge places them in the middle three quintiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as responders and can move forward with standard treatment for the pathogenic organism. Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest quintile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge quintile. Subjects classified as nonresponders can also be treated with more aggressive therapies and/or higher doses of therapeutics to account for the immunosenescence in the subject.


Still alternatively, if the subject's iAge places them in the youngest iAge tertile for their age group (see Table 1) they can be classified as responders and move forward with standard treatment for the pathogenic organism. If the subject's iAge places them in the middle tertile, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose JakSTAT activity places them in the highest quartile can be classified as responders and can move forward with standard treatment for the pathogenic organism. Subjects whose JakSTAT activity places them in the lower three quartiles can be classified as nonresponders and are treated to lower iAge (and increase their Jak-STAT score) into a responder group. If the subject's iAge places them in the oldest tertile, they can be classified as nonresponders and are treated to lower their iAge (see below) into a responder group of a younger iAge tertile. Subjects classified as nonresponders can also be treated with more aggressive therapies and/or higher doses of therapeutics to account for the immunosenescence in the subject.


Cardiovascular Disease

Cardiovascular disease include a class of diseases that involve the heart, the blood vessels (arteries, capillaries, and veins) or both. Cardiovascular disease refers to any disease that affects the cardiovascular system, principally cardiac disease including cardiomyopathies, vascular diseases of the brain and kidney, and peripheral arterial disease. Cardiovascular disease can refer to a disease that primarily affects the heart, and can be referred to as cardiac disease. Cardiovascular disease can refer to a disease in which the pathology begins with cardiac damage, malfunction, or malformation, as opposed to disease in which cardiac damage, malfunction, or malformation is a result of a primary pathology present at a site remote from the heart (e.g., cardiovascular disease as a comorbidity to another disease or condition). For example, heart failure, cardiac dysrhythmias (abnormalities of heart rhythm including increased QT duration and atrial flutter and/or fibrillation), inflammatory heart disease including endocarditis (inflammation of the inner layer of the heart, the endocardium, most commonly the heart valves); inflammatory cardiomegaly (enlarged heart, cardiac hypertrophy); myocarditis (inflammation of the myocardium); valvular heart disease; congenital heart disease; and rheumatic heart disease (heart muscle and valve damage due to rheumatic fever caused by streptococcal bacteria infections) are examples of cardiac damage, malfunction, or malformation in which the primary pathology can be or is present in the heart, and subsequently can result in vascular or other systemic disease. Alternatively, coronary heart disease (also ischaemic heart disease or coronary artery disease); hypertensive heart disease (diseases of the heart secondary to high blood pressure); cor pulmonale (failure at the right side of the heart with respiratory system involvement); cerebrovascular disease (disease of blood vessels that supplies to the brain such as stroke); peripheral arterial disease (disease of blood vessels that supplies to the arms and legs); and artherosclerosis are a result of pathology present initially at a site remote from the heart. Cardiovascular disease initiated either at the heart or at a site remote from the heart can result in heart failure. Cardiovascular disease can include disease in which the initial pathology is at a site remote from the heart. Cardiovascular disease also includes conditions affecting the heart, heart valves, and vasculature (e.g., arteries and veins) of the body and encompasses diseases and conditions including, but not limited to arteriosclerosis, atherosclerosis, myocardial infarction, acute coronary syndrome, angina, congestive heart failure, aortic aneurysm, aortic dissection, iliac or femoral aneurysm, pulmonary embolism, primary hypertension, atrial fibrillation, stroke, transient ischemic attack, systolic dysfunction, diastolic dysfunction, myocarditis, atrial tachycardia, ventricular fibrillation, endocarditis, arteriopathy, vasculitis, atherosclerotic plaque, vulnerable plaque, acute coronary syndrome, acute ischemic attack, sudden cardiac death, peripheral vascular disease, coronary artery disease (CAD), peripheral artery disease (PAD), and cerebrovascular disease.


Cardiomyopathy includes one or more conditions (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more of) selected from the group consisting of increased QT duration, arrhythmias, myocardial ischemia, hypertension and thromboembolic complications, myocardial dysfunction, cardiomyopathy, heart failure, atrial fibrillation, cardiomyopathy and heart failure, heart failure and LV dysfunction, atrial flutter and fibrillation, and, heart valve damage and heart failure. In certain embodiments, cardiomyopathy does not include cardiomyopathy as a comorbidity to another disease or condition.


Heart failure often called congestive heart failure (CHF) or congestive cardiac failure (CCF), includes conditions that occur when the heart is unable to provide sufficient pump action to maintain blood flow to meet the needs of the body. Heart failure can cause a number of symptoms including shortness of breath, leg swelling, and exercise intolerance. The condition is typically diagnosed by patient physical examination and confirmed with echocardiography. Common causes of heart failure include myocardial infarction and other forms of ischemic heart disease, hypertension, valvular heart disease, and cardiomyopathy.


Cardiovascular disease includes atherosclerosis a chronic disease process characterized by lipid deposits and fibrosis of the intima, irregularly distributed in large and medium sized arteries. The disease is progressive and most often becomes clinically manifest in the middle-aged and elderly. When severe, the atherosclerotic plaque causes a reduction of the cross-sectional area of the arterial lumen, with and without thrombosis. Atherosclerotic plaques can occur in essentially any or all of the blood vessels of the body, resulting in cardiovascular diseases involving the heart (e.g., acute coronary syndrome, heart failure, and myocardial infarction), the brain (e.g., stroke, transient ischemic attack, and brain infarction), the kidney (e.g., acute and chronic kidney disease, hypertension), and the extremities (e.g., peripheral vascular disease, lower and/or upper extremity claudication, and lower and/or upper extremity ischemia). Resultant ischemic manifestations include: angina pectoris, rayocardial infarction, stroke, intermittent claudication, gangrene of the lower extremities, and renovascular hypertension. Atherosclerosis can be considered to be an inflammatory disease. For example, the lesions of atherosclerosis appear to represent a series of highly-specific cellular and molecular responses that can be described as an inflammatory disease. See, e.g., Ross, “Atherosclerosis—An inflammatory disease” N Engl J Med (1999), 340:115-126; the publications cited in Ross (1999); and subsequent publications that cite Ross (1999); each of which is incorporated herein in reference in its entirety.


A subject can be identified as having cardiovascular disease by the presence of any one of: documented coronary artery disease, documented cerebrovascular disease, documented carotid disease, documented peripheral arterial disease, or combinations thereof. A subject can also be identified as having cardiovaswcular disease if the subject is at least 45 years old and: (a) has one or more stenosis of greater than 50% in two major epicardial coronary arteries; (b) has had a documented prior MI; (c) has been hospitalized for high-risk NSTE ACS with objective evidence of ischemia (e.g., ST-segment deviation and/or biomarker positivity); (d) has a documented prior ischemic stroke; (e) has symptomatic artery disease with at least 50% carotid arterial stenosis; (0 has asymptomatic carotid artery disease with at least 70% carotid arterial stenosis per angiography or duplex ultrasound; (g) has an ankle-brachial index (“ABI”) of less than 0.9 with symptoms of intermittent claudication; and/or (h) has a history of aorto-iliac or peripheral arterial intervention (catheter-based or surgical).


Cardiovascular Treatments Using iAge and Cardiac Markers


Subjects with cardiovascular disease or at risk for cardiovascular disease have their blood drawn and an iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and cAge are calculated as described above. If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the youngest quartile for their age group they can be classified as low risk for cardiovascular disease and move forward with the standard therapy (CVD patients) or no therapy (patients at risk but no CVD at the time). If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the middle two quartiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) can be tested for Jak-STAT activity (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as low risk and move forward with standard therapy (CVD patients) or no therapy (patients at risk but no CVD at the time). Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as higher risk for cardiovascular disease and can be treated to lower iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge (and increase their Jak-STAT score) into a low risk group. If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the oldest quartile, they can be classified as higher risk patients and can be treated to lower their iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge (see above) into a low risk group.


Alternatively, if the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the youngest iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge quintile for their age group (see Table 1) they can be classified as low risk and move forward with the standard therapy (CVD patients) or no therapy (patients with no CVD at the time). If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the middle three quintiles, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as low risk and move forward with the standard therapy (CVD patients) or no therapy (patients at risk but no CVD at the time). Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as higher risk and can be treated to lower iAge, CRS, cardiac marker levels (lower MIG, raise LIF, raise SIRT3), and/or cAge (and increase their Jak-STAT score) into a low risk group. If the subject's iAge places them in the oldest quintile, they can be classified as higher risk and can be treated to lower their iAge, CRS, cardiac marker levels (lower MIG, increase LIF, increase SIRT3), and/or cAge (see above) into a low risk group of a younger iAge quintile.


Still alternatively, if the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the youngest iAge tertile for their age group (see Table 1) they can be classified as low risk and move forward with the standard therapy (CVD patients) or no therapy (patients at risk but no CVD at the time). If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the middle tertile, the subject's blood cells (e.g., CD4+ and CD8+ cells) are stimulated and Jak-STAT activity is measured (see, e.g., Example 1 below). Subject's whose Jak-STAT activity places them in the highest quartile can be classified as low risk and move forward with the standard therapy (CVD patients) or no therapy (patients at risk but no CVD at the time). Subjects whose Jak-STAT activity places them in the lower three quartiles can be classified as higher risk and can be treated to lower iAge, CRS, cardiac marker levels (lower MIG, increase LIF, increase SIRT3), and/or cAge (and increase their Jak-STAT score) into a low risk group. If the subject's iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge places them in the oldest tertile, they can be classified as higher risk and are treated to lower their iAge, CRS, cardiac marker levels (lower MIG, increase LIF, increase SIRT3), and/or cAge (see above) into a low risk group of a younger iAge, CRS, cardiac marker levels (MIG, LIF, SIRT3), and/or cAge tertile.


Agents for Improving iAge and Cardiac Markers


In addition to using iAge to classify patients (FIG. 3) or cAge to classify patients, these can be used to derive individual inflammatory profiles by comparing subject's individual protein levels with those of a population (e.g., of similar chronological age). The resulting signatures (or barcodes) are used for protein-compound association (PCI) analysis using the drugbank database (www.drugbank.ca) and a personalized initial therapy to reduce iAge can be generated (FIG. 3). Patients following personalized recommendations can be monitored weekly for changes in iAge and/or cAge until they reach optimal levels (below group average for a given age bracket) and they convert into a responder treatment phenotype (FIG. 3) and/or lower risk for cardiovascular disease. The patient is then classified as a responder and is suitable for immunotherapy treatment, or lower risk for CVD.


A subject may reduce their iAge with treatments that lower the levels of TRAIL, IFNG, GROA, IL2, TGFA, PAIL and/or LIF to their optimal levels for a person's chronological age. A subject may reduce their cAge with treatments that raise the levels of LIF or SIRT3 A subject may also reduce their iAge with treatments that raise the levels of MIG, EOTAXIN, LEPTIN, IL-1B, or MIP1A to their optimal levels for a person's age. A subject may also reduce their cAge with treatments that lower the levels of MIG.


A subject may also reduce their iAge and/or cAge by reducing any systemic chronic inflammation, using any of the following, whether alone or in combination: (1) pharmacological treatment, including without limitation anti-inflammatory drugs (NSAIDs such as, for example, aspirin, ibuprofen, naproxen, diclofenac, celecoxib, oxaprozin, piroxicam, indomethacin, meloxicam, fenoprofen, diflunisal, etodolac, ketorolac, meclofenamate, nabumetone) or corticosteroids (e.g., glucocorticoids, mineralocorticoids); (2) neutraceuticals or nutritional supplements, including without limitation fish oil, lipoic acid, and curcumin, or spices/herbs such as ginger, garlic, turmeric, hyssop, cannabis, Harpagophytum procumbens, and cayenne; (3) dietary change, including without limitation increasing the intake of foods that are high in antioxidants and polyphenols, such as olive oil, leafy greens (e.g., kale and spinach), broccoli, avocados, green tea, bell peppers, chili peppers, mushrooms, dark chocolate, cocoa, tomatoes, fatty fish (e.g., salmon, sardines, herring, anchovies, and mackerel), nuts (walnuts and almonds), and fruits (e.g., cherries, blackberries, blueberries, raspberries, strawberries, grapes, and oranges), and/or decreasing the intake of foods that can increase inflammation such as refined carbohydrates (e.g., white bread and pastries), high-fructose corn syrup, refined sugar, processed and packaged food, fried foods, red meat, excessive alcohol, and processed meat; and (4) lifestyle changes including without limitation eliminating or reducing smoking and alcohol intake, maintaining a healthy body weight, and reducing stress levels.


Cardiac markers can be improved by providing a subject with treatments that improve the level of the cardiac marker (lowering cAge). A subject's cardiac marker score (cAge) can be lowered by reducing the MIG in a patient, increasing Sirtuin-3 in the patient, increasing LIF in the patient, and/or reducing cell signaling from CXCR3 (the receptor for MIG). A variety of agents are known which can reduce MIG expression, increase Sirtuin-3 expression, increase LIF activity (or LIF like activity), and/or act as antagonists for CXCR3 (the receptor for MIG).


Agents that can lower MIG (and so improve the cardiac marker and iAge marker) include, for example arsenic trioxide, Roxarsone, Selenium, and/or a variety of antibodies. Antibodies include, for example, MIG-2F5.5 (anti-human CXCL9 antibody, BioLegend Cat. #740072), Anti-human CXCL9 antibody, NSJ Bioreagents, Cat #R30501, Mouse MAb clone 49106 (anti-human CXCL9, R&D Systems Cat #MAB392), Mouse monoclonal MAb for human CXCL9 (neutralizing, GeneTex, Cat #GTX52673), Mouse monoclonal anti-human CXCL9 antibody (OriGene, Cat #PM1209P), MIG Antibody (MM0220-7F11) (Novus Biologicals, NBP2-12236), MIG Antibody (1F5) (Novus Biologicals, H00004283-M06), Mouse MAb anti human CXCL9 (ThermoFisherCat #MA5-23746, Cat #MA5-30320, Cat #MA5-23628, Cat #MA5-23544).


Arsenic trioxide (As2O3), a component of traditional Chinese medicine, has been used successfully for the treatment of acute promyelocytic leukemia (APL), and As2O3 is of potential therapeutic value for the treatment of other promyelocytic malignancies and some solid tumors including breast cancer. As2O3 treatment changed the expression level of several genes that involved in cell cycle regulation, signal transduction, and apoptosis. Notably, As2O3 treatment increased the mRNA and protein levels of the cell cycle inhibitory proteins, p21 and p27. Interestingly, knocking down p21 or p27 individually did not alter As2O3-induced apoptosis and cell cycle arrest; however, the simultaneous down-regulation of both p21 and p27 resulted in attenuating of G1, G2/M arrest and reduction in apoptosis, thus indicating that p21 and p27 as the primary molecular targets of As2O3.


Roxarsone is an organoarsonic acid where the organyl group is 4-hydroxy-3-nitrophenyl. It has a role as a coccidiostat, an antibacterial drug, an agrochemical and an animal growth promotant. It is an organoarsonic acid and a member of 2-nitrophenols. Roxarsone was found to exhibit a higher angiogenic index than As' at lower concentrations. Increased endothelial nitric oxide synthase (eNOS) activity was observed for roxarsone but not for AsIII-induced angiogenesis. However, AsIII caused more rapid and pronounced phosphorylation of eNOS.


Selenium (Se) is a potential anticarcinogenic nutrient, and the essential role of Se in cell growth is well recognized but certain cancer cells appear to have acquired a survival advantage under conditions of Se-deficiency. Se can exert its effects through increasing the expression of a humoral defense gene (A2M) and tumor suppressor-related genes (IGFBP3, HHIP) while decreasing pro-inflammatory gene (MIG, HSPB2) expression.


Agents that raise Sirtuin-3 levels include, for example, Berberine and Resveratrol. Berberine (molecular formula C20H19NO5 and molecular weight of 353.36) is the main active component of an ancient Chinese herb Coptis chinensis French, which has been used to treat diabetes for thousands of years. Berberine is an Over-the-Counter (OTC) drug, which is used to treat gastrointestinal infections in China. Berberine has been shown to regulate glucose and lipid metabolism in vitro and in vivo. Berberine is also a potent oral hypoglycemic agent with beneficial effects on lipid metabolism.


Resveratrol (3,5,4′-trihydroxy-trans-stilbene) belongs to polyphenols' stilbenoids group, possessing two phenol rings linked to each other by an ethylene bridge. This natural polyphenol has been detected in more than 70 plant species, especially in grapes' skin and seeds, and was found in discrete amounts in red wines and various human foods. It is a phytoalexin that acts against pathogens, including bacteria and fungi. As a natural food ingredient, numerous studies have demonstrated that resveratrol possesses a very high antioxidant potential. Resveratrol also exhibit antitumor activity, and is considered a potential candidate for prevention and treatment of several types of cancer. Indeed, resveratrol anticancer properties have been confirmed by many in vitro and in vivo studies, which shows that resveratrol is able to inhibit all carcinogenesis stages (e.g., initiation, promotion and progression). Even more, other bioactive effects, namely as anti-inflammatory, anticarcinogenic, cardioprotective, vasorelaxant, phytoestrogenic and neuroprotective have also been reported.


Agents that raise LIF levels include Aminodarone, arsenic trioxide, Azathioprine, Estradiol, Chlorambucil, Clomiphene, Coumaphos, Cyclosporine, decitabine, Cisplatin, Vincristine, Formaldehyde, Glucose, Hydrogen Peroxide, letrozole, Lindane, Methotrexate, Quercetin, Oxyquinoline, resorcinol, resveratrol, Silicon Dioxide, Tretinoin, and troglitazone.


The expression of LIF is regulated by many cytokines. In normal human bone marrow stromal cells, IL-1α, IL-1β, TGF-β and tumor necrosis factor-α (TNF-α) can all increase the transcription of LIF mRNA. The induction of LIF by IL-1β and TNF-α was also observed in gingival fibroblasts and several cell types in human airways. In addition, the induction of LIF expression by other cytokines, including IL-6, IL-2 has been observed in different cell types, including airway smooth-muscles and MT-2 cells. The expression of LIF can also be inhibited by some factors, including 1α, 25-dihydroxyvitamin D3 and dexamethasone. The analysis of the LIF promoter revealed that transcription factor STAT5 can bind to the LIF promoter and induce its expression in myeloid cell lines. In addition, the LIF promoter region contains several ETS binding sites. The binding of ETS transcription factors to the LIF promoter is critical for the induction of LIF in response to T cell activators.


Amiodarone is a primarily a class III antiarrhythmic and is one of the most commonly used anti-arrhythmic drugs. While the United States FDA has labeled amiodarone for the treatment of life-threatening ventricular arrhythmias, the drug is commonly used off-label to treat supraventricular tachyarrhythmias such as atrial fibrillation as well as for the prevention of ventricular tachyarrhythmias (VTs) in high-risk patients. Like other antiarrhythmic drugs of this class, amiodarone works primarily by blocking potassium rectifier currents that are responsible for repolarization of the heart during phase 3 of the cardiac action potential. This potassium channel-blocking effect results in increased action potential duration and a prolonged effective refractory period in cardiac myocytes. Unlike other class III agents, amiodarone also interferes with beta-adrenergic receptors, calcium channels, and sodium channels.


Clomiphene is an ovulatory stimulant designated chemically as 2-[p-(2-chloro-1,2-diphenylvinyl)phenoxy]triethylamine citrate (1:1). It has the molecular formula of C26H28ClNO•C6H8O7 and a molecular weight of 598.09. Clomiphene is capable of interacting with estrogen-receptor-containing tissues, including the hypothalamus, pituitary, ovary, endometrium, vagina, and cervix. It may compete with estrogen for estrogen-receptor-binding sites and may delay replenishment of intracellular estrogen receptors. Clomiphene initiates a series of endocrine events culminating in a preovulatory gonadotropin surge and subsequent follicular rupture. The first endocrine event in response to a course of clomiphene therapy is an increase in the release of pituitary gonadotropins.


Coumaphos is an organothiophosphate insecticide, an organic thiophosphate and an organochlorine compound. It has a role as an agrochemical, an acaricide, an antinematodal drug, an avicide and an EC 3.1.1.8 (cholinesterase) inhibitor. Coumaphos is used for control of a wide variety of insects on cattle and parasitic mites (Varroa jacobson) on bees. It is also used in veterinary medicine for the treatment of screwworms, maggots, and ear ticks on livestock. In humans coumaphos causes muscarinic effects (parasympathetic), nicotinic effects (sympathetic and motor), and CNS effects associated with massive overstimulation of the chlorinergic system.


Lindane also known as gamma-hexachlorocyclohexane (γ-HCH), gammaxene, and Gammallin is an organochlorine chemical and an isomer of hexachlorocyclohexane that has been used both as an agricultural insecticide and as a pharmaceutical treatment for lice and scabies. Lindane is a neurotoxin that interferes with GABA neurotransmitter function by interacting with the GABAA receptor-chloride channel complex at the picrotoxin binding site. In humans, lindane affects the nervous system, liver, and kidneys, and may well be a carcinogen.


Oxyquinoline is a heterocyclic phenol and Oxyquinoline Sulfate is its salt, both of which are described as cosmetic biocides for use in cosmetic formulations. Oxyquinoline can be used as an antiseptic, disinfectant, and has pesticide properties. Oxyquinoline is also a chelating agent which has been used for the quantitative determination of metal ions.


Decitabine (5-aza-2′-deoxycytidine or 5-Aza-Cdr) is a cytosine analogue that was first synthesized in the early 1960s by Pliml and Sorm and is currently marketed as Dacogen® by Eisai (Tokyo, Japan). It differs from deoxycytidine by the substitution of nitrogen for carbon at the 5-position of the pyrimidine ring. It was noted to have an antileukemic effect in cell lines, with more potency in vitro than cytarabine. Initially, its cytotoxicity was attributed to its ability to impair DNA synthesis and cause DNA damage similar to other antimetabolites. At low doses, decitabine induces differentiation by reversing DNA methylation-induced gene silencing. Once inside a cell, decitabine is phosphorylated and activated by the enzyme deoxycytidine kinase to its triphosphate form aza-dCTP. It then competes with and replaces cytosine in the CpG (cytosine-guanosine dinucleotide) islands that occur in clusters in promoter regions. During subsequent cell divisions, aza-dCTP inhibits methylation of the promoter by forming a covalent bond with the enzyme DNA methyltransferase (DNMT), and thereby traps and contributes to degradation of the enzyme.


Chlorambucil and Cisplatin are alkylating agents used to treat cancer. Chlorambucil is in the class of nitrogen mustards, and Cisplatin is a platinum based-agent. Chlorambucil produces its anti-cancer effects by interfering with DNA replication and damaging the DNA in a cell. The DNA damage induces cell cycle arrest and cellular apoptosis via the accumulation of cytosolic p53 and subsequent activation of Bcl-2-associated X protein, an apoptosis promoter. Cisplatin crosslinks DNA in several different ways, interfering with cell division by mitosis. The damaged DNA elicits DNA repair mechanisms and activates apoptosis.


Vincristine is a chemotherapy drug that belongs to a group of drugs called vinca alkaloids. Vincristine works by stopping the cancer cells from separating into 2 new cells. Vincristine works partly by binding to the tubulin protein, stopping the tubulin dimers from polymerizing to form microtubules, causing the cell to be unable to separate its chromosomes during the metaphase. The cell then undergoes apoptosis.


Letrozole is an aromatase inhibitor which is used in the treatment of hormonally-responsive breast cancer after surgery. Letrozole is also for ovulation induction. Letrozole blocks the production of estrogens in this way by competitive, reversible binding to the heme of its cytochrome P450 unit. Letrozole has shown to reduce estrogen levels by 98 percent while raising testosterone levels.


Tretinoin is a derivative of vitamin A. It is used on the skin (topically) in the treatment of mild to moderate acne and on skin that has been damaged by excessive exposure to the sun. Tretinoin irritates the skin and causes the cells of the skin to grow (divide) and die more rapidly, increasing the turnover of cells. Tretinoin can also induce acute promyelocytic leukemia cells to differentiate and stops them from proliferating; in people there is evidence that it forces the primary cancerous promyelocytes to differentiate into their final form.


Estradiol is the main circulating oestrogen in women and reaches a plasma concentration of 30-400 pg/mL before menopause. Estradiol regulates growth and the development of the reproductive system, also, helps to maintain the osseous tissue, the central nervous system and the vasodilatation in the vascular tissue. The protective effect of Estradiol in the vasculature and against cardiovascular disease (CVD) has been demonstrated in several hormone replacement studies. Estradiol activates BK channels via a process that requires the presence of the β1 subunit. Valverde et al. were the first to propose that Estradiol affected BK channels by binding to β1, but it is still a matter of debate whether the agonistic action of Estradiol on BK channels is caused by its binding to the β1 subunit or to the α/β1 complex. Moreover, the molecular nature of the Estradiol binding site and the mode of action of the hormone are at present unknown. Acute application of Estradiol (100 nM) decreases smooth muscle excitability by activating BK channels. Notably, Estradiol or its membrane-impermeant form (E2-BSA) can induce a fast increase in BK channel activity in MCF-7 breast epithelial cancer cells with an EC50 of 80 pM reaching a maximal effect at 10 nM34. Rapid effects of Estradiol have also been reported in neurons of the area postrema where nanomolar concentrations of E2 can decrease the firing rate most probably by increasing BK current35. All these examples underscore the physiological importance of the regulation of BK channels by E2 and made worthwhile efforts in determining the molecular nature of the interaction between this hormone and the BK channel.


Cyclosporine has been a core component of immunosuppression in both immune dysregulatory disorders and organ transplantation. For immune disorders involving ophthalmologic, dermatologic, hematologic, gastroenterologic, neurologic, or musculoskeletal systems, cyclosporine has demonstrated marked efficacy in relieving clinical symptoms and reversing pathological developments. Additionally, after the drug's implementation in transplantation medicine, rates of acute rejection and one-year graft survival have improved dramatically.


Methotrexate is a chemotherapy agent and immune system suppressant. It is used to treat cancer, autoimmune diseases, ectopic pregnancy, and for medical abortions. Types of cancers it is used for include breast cancer, leukemia, lung cancer, lymphoma, and osteosarcoma. Types of autoimmune diseases it is used for include psoriasis, rheumatoid arthritis, and Crohn's disease. It can be given by mouth or by injection. Methotrexate is an antimetabolite of the antifolate type. It is thought to affect cancer and rheumatoid arthritis by two different pathways. For cancer, methotrexate competitively inhibits dihydrofolate reductase (DHFR), an enzyme that participates in the tetrahydrofolate synthesis. The affinity of methotrexate for DHFR is about 1000-fold that of folate. DHFR catalyses the conversion of dihydrofolate to the active tetrahydrofolate. Folic acid is needed for the de novo synthesis of the nucleoside thymidine, required for DNA synthesis. Also, folate is essential for purine and pyrimidine base biosynthesis, so synthesis will be inhibited. Methotrexate, therefore, inhibits the synthesis of DNA, RNA, thymidylates, and proteins. For the treatment of rheumatoid arthritis (immune suppression), multiple mechanisms appear to be involved, including the inhibition of enzymes involved in purine metabolism, leading to accumulation of adenosine; inhibition of T cell activation and suppression of intercellular adhesion molecule expression by T cells; selective down-regulation of B cells; increasing CD95 sensitivity of activated T cells; and inhibition of methyltransferase activity, leading to deactivation of enzyme activity relevant to immune system function. Another mechanism of MTX is the inhibition of the binding of interleukin 1-beta to its cell surface receptor.


Troglitazone is an antidiabetic and anti-inflammatory drug, and a member of the drug class of the thiazolidinediones. Troglitazone is an oral antihyperglycemic agent which acts primarily by decreasing insulin resistance. Troglitazone is used in the management of type II diabetes. Troglitazone binds to nuclear receptors (PPAR) that regulate the transcription of a number of insulin responsive genes critical for the control of glucose and lipid metabolism. Troglitazone decrease nuclear factor kappa-B (NF-κB) and increase its inhibitor (IκB).


Azathioprine is a purine analogue with cytotoxic and immunosuppressive activity. Azathioprine is a prodrug that is converted by hepatic xanthine oxidase to its active metabolite 6-mercaptopurine (6-MP). 6-MP is further metabolized by hypoxanthine-guanine phosphoribosyltransferase (HGPRT) into 6-thioguanosine-5′-phosphate (6-thio-GMP) and 6-thioinosine monophosphate (6-thio-IMP), both inhibit nucleotide conversions and de novo purine synthesis. This leads to inhibition of DNA, RNA, and protein synthesis. As a result, cell proliferation may be inhibited, particularly in lymphocytes and leukocytes. Azathioprine an immunosuppressive agent in organ transplantation to prevent rejection and in autoimmune diseases as a corticosteroid sparing agent.


Quercetin, a flavonoid found in fruits and vegetables, has unique biological properties that may improve mental/physical performance and reduce infection risk. These properties form the basis for potential benefits to overall health and disease resistance, including anti-carcinogenic, anti-inflammatory, antiviral, antioxidant, and psychostimulant activities, as well as the ability to inhibit lipid peroxidation, platelet aggregation and capillary permeability, and to stimulate mitochondrial biogenesis. Quercetin is a naturally occurring polar auxin transport inhibitor. Quercetin inhibits lipopolysaccharide (LPS)-induced tumor necrosis factor α (TNF-α) production in macrophages and LPS-induced IL-8 production in lung A549 cells. Moreover, in glial cells it was even shown that quercetin can inhibit LPS-induced mRNA levels of TNF-α and interleukin IL-1α, this effect of quercetin resulted in a diminished apoptotic neuronal cell death induced by microglial activation. Quercetin inhibits production of inflammation-producing enzymes (cyclooxygenase (COX) and lipoxygenase (LOX)). It limits LPS-induced inflammation via inhibition of Src- and Syk-mediated phosphatidylinositol-3-Kinase (PI3K)-(p85) tyrosine phosphorylation and subsequent Toll Like Receptor 4 (TLR4)/MyD88/PI3K complex formation that limits activation of downstream signaling pathways in RAW 264.7 cells. It can also inhibit FcεRI-mediated release of pro-inflammatory cytokines, tryptase and histamine from human umbilical cord blood-derived cultured mast cells (hCBMCs); this inhibition appears to involve in inhibition of calcium influx, as well as phospho-protein kinase C (PKC).


Resorcinol is an organic compound with the formula C6H4(OH)2. Resorcinol is used as an antiseptic and disinfectant in topical pharmaceutical products in the treatment of skin disorders and infections such as acne, seborrheic dermatitis, eczema, psoriasis, corns, calluses, and warts. It is also used to treat corns, calluses, and warts. It exerts a keratolytic activity.


Agents that reduce expression of CXCR3 (the receptor for MIG) include, for example, formaldehyde and taurine. Agents that are antagonists for CXCR3 include, for example, piperazinyl-piperidines (e.g., SCH546738), 8-azaquinazolinones (e.g., AMG 487), 3-phenyl-3H-quinazolin-4-ones, aryl piperazine, 4-aryl-5-piperazinylthiazoles, arylpiperazines, benzetimide derivatives, imidazolidines, imidazolium, lysergic acid derivative, diaminocyclobutenediones, zinc phthalocyanine, and NBI-74330. (See Andrews et al., J. Med. Chem. 59:2894-917 (2016), which is incorporated by reference in its entirety for all purposes). Chemical structures for specific antagonists of CXCR3 are found in Andrews 2016, and are hereby incorporated by reference in their entirety for all purposes. A few of the specific structures are shown below:




text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


Other small molecule antagonists are found, for example, in US20060036093, WO2009/105435, which all are incorporated by reference in their entirety for all purposes.


Any of the foregoing antibodies or fragments thereof (collectively antibodies) can be engineered for use in humans by methods such as, for example, chimerization, humanization, humaneering, etc, which are known in the art.


In addition, any of the foregoing antibodies or fragments thereof (collectively antibodies) can include a protracting moiety that extends a half-life (T1/2) or/and the duration of action of the antibody. The protracting moiety can extend the circulation T1/2, blood T1/2, plasma T1/2, serum T1/2, terminal T1/2, biological T1/2, elimination T1/2 or functional T1/2, or any combination thereof, of the antibody. One or more protracting moieties can be combined (covalently or noncovalently) with an antibody. Protracting moieties include, for example, hydrophilic polymers (e.g., PEG, dextran, etc.), a synthetic polymer, glycosylation, human serum albumin (HSA) or a portion thereof (e.g., domain III) that binds to the neonatal Fc receptor (FcRn), or a carboxy-terminal peptide (CTP).


Additional agents (drugs, foods and other molecules) that can alter iAge by affecting genes involved in systemic chronic inflammation comprising MIG, TNFSF10, IFNg, CCL11 or CXCL1 are listed below in Tables 7, 8, and 9. These molecules were obtained using methods described below with a combined confidence score >500 (q value of <0.05, nominal p value of <0.005) Table 7 shows drugs and other molecules that can change iAge by interacting with immune genes involved in the inflammatory response including MIG, TNFSF10, IFNg, CCL11 or CXCL1 and changing the levels of these proteins in the subject. Table 8 shows food compounds and other molecules that can change iAge by interacting with immune genes involved in the inflammatory response including MIG, TNFSF10, IFNg, CCL11 or CXCL1 and changing the levels of these proteins in the subject. Table 9 shows drugs that can upregulate or downregulate MIG, TNFSF10, IFNg, CCL11 or CXCL1, and whether the up- or down-regulation is beneficial (lowers) or detrimental (raises) to iAge. Other drugs and other molecules that interact with genes/proteins involved in inflammation and/or the inflammatory response can be used to reduce the iAge of the subject through indirect effects on the levels of the iAge markers which are described above. Other food compounds or other molecules that interact with genes/proteins involved in inflammation and/or the inflammatory response can be used to reduce the iAge of the subject through indirect effects on the levels of the iAge markers which are described above.


Methods for Determining the Effect of a Substance on iAge


Compounds that can modify iAge are identified from DrugBank and FooDB using a compound-gene interaction database, machine learning for drug repurposing and food compound mapping for anti-inflammatory activity, and medication usage studies from the 1KIP cohort.


The STITCH database v. 5.0 can be used as the compound-gene interaction database to find immune genes with which a drug or food compound interacts. Compound-protein interactions are extracted from the STITCH database v5.0 by matching the InChI keys of drugs/food compounds. STITCH collects information from multiple sources and individual scores from each source are combined into an overall confidence score.


An immune gene set (n=4275) is obtained from the HIM CHip panel. The immune gene set is then matched with the two compound-gene interaction datasets above to extract immune genes that interact with drugs or food compounds. The immune gene set is used in the STITCH database, and FDA-approved drugs (n=1692) are selected from the DrugBank database and food compounds (n=7962) are selected from the FoodDB database as previously described in Veselkov et 1., Sci Rep. 2019; 9: 9237, PMCID: PMC6610092, which is incorporated by reference in its entirety for all purposes.


All interactions with a combined confidence score of less than 500 are removed. The Ensemble peptide identifiers for protein are then converted to HGNC gene symbols using Biomart (version 2.42.0). As a result, there are 1617 immune genes interact with drugs and 1774 immune genes interact with food compounds. After pre-processing, two data sets are obtained: i) drug-gene interaction dataset containing 1670 drugs, 9642 genes with 118,342 interaction ii) food compound-gene interaction dataset containing 3447 compounds, 10,942 genes, and 166,431 interactions.


To investigate compound-immunity association, statistical significance for the enrichment of compound genes in the immune gene set is calculated using Fisher's exact test. The universal gene set contains all genes that interact with at least one compound. The compound with low p-value interacts with a higher proportion of the immune gene set than that expected by chance. The statistical analysis is performed using R.


FDA-approved drugs (n=1692) are selected from the DrugBank database as previously described in Veselkov et 1., Sci Rep. 2019; 9: 9237, PMCID: PMC6610092, which is incorporated by reference in its entirety for all purposes. The set of drugs is mapped to the DrugCentral database via InChI keys to identify drugs indicated for anti-inflammatory treatment (n=49), that are denoted as ‘positive class’. All drugs with no known association with anti-inflammatory activity are denoted as ‘negative class’. This set of drugs will be used as a training set to train machine learning models. Food compounds (n=7962) are selected from the FoodDB database as previously described. The set of food compounds will be used as a test data set.


Compound-protein interactions are extracted from the STITCH database v5.0 by matching the InChI keys of drugs/food compounds. STICH collects information from multiple sources and individual scores from each source are combined into an overall confidence score. The threshold for the significant score is not set at a fixed value and considered as an adjustable parameter for ML model optimization.


Gene-gene interactions are extracted from public sources including STRING, UniProt, COSMIC, BioPlex, and NCBI Gene as previously described in Veselkov et 1., Sci Rep. 2019; 9: 9237, PMCID: PMC6610092, which is incorporated by reference in its entirety for all purposes. This results in a gene-gene interactome dataset containing 20,256 genes with ˜11 million interactions.


The gene profile for each compound is represented as a sparse matrix, in which a ‘1’ indicates genes that directly interact with the compound and a ‘0’ for all other genes. The network propagation (Random Walk with Restart) algorithm is then applied to spread this gene profile on to the human interactome. As a result, a genome-wide profile of gene scores is obtained for each compound. The restart parameter ‘c’ is considered as an adjustable parameter for ML model optimization.


ML can use Linear SVM as a classifier for optimization. The interaction score threshold can be set at 600, the restart parameter ‘c’ for network propagation can be set at 0.1. Linear SVM can be used to identify anti-inflammatory drugs based on their genome-wide profile obtained from network propagation. The regularization parameter ′C; can be optimized during the model training using a nested cross-validation strategy. The F-score, that balances sensitivity and specificity, is used to evaluate the outcome of each model. The best model is defined as a model with the highest F-score.


The anti-inflammatory ‘likeness’ of drugs is calculated using the selected model. These values are used to identify potential drugs for anti-inflammatory repurposing. Similarly, the selected model is applied to the food compound dataset. The probability estimates for the anti-inflammatory activity of each food compound are calculated. Food compounds with high anti-inflammatory probability (i.e., >0.8) are selected for validation.


Medication prescribed for 1KIP cohorts are divided into 23 groups. Medication usage is represented as a sparse matrix, in which ‘1’ indicates the patient took at least one of the drugs in that group and ‘0’ indicates none of the drugs in that group was not taken.


Several analyses were performed as follows: build a model to predict iAge based on medication usage using lasso method implemented in ‘glmnet’ package; study correlation between medication usage and cytokine/chemokine levels using PLS method implemented in ‘pls’ package; and build a model to predict cytokine/chemokine levels based on medication usage using lasso in ‘glmnet’ and lasso with interaction in ‘glinternet’.


Additional ways to find agents that can lower iAge include a deep neural network approach implemented using DeepCOP (Woo et. al. 2019). Level 5 expression scores from the LINCS L1000 study can be used to label genes as up- or down-regulated if scores were more than a 50% perturbation above the top-threshold. Compounds in LINCS L1000 and compounds from FooDB (Wishart 2019) were represented using calculated Morgan Fingerprints from SMILES using RDkit (Landrum 2013). Genes were represented as commonly occurring gene ontologies (at least three times). The deep neural network was trained on LINCS L1000 compounds on CD34 and HUVEC cells, independently, using the target genes plus the imputed expression for the genes that encode the Inflammatory Age proteins. Predicted expression probabilities for the compounds in FooDB were used to score these interventions. Compounds that only upregulate anti-inflammatory markers and downregulate pro-inflammatory markers are considered as a possible interventions.


The network propagation algorithm can be implemented using the method in (Veselkov., et al, 2019). The assumption is that compounds with similar network profiles would have similar effect of regulation of a certain gene. Compound-gene interactions can be extracted from the STITCH database v5.0. STITCH collects information from multiple sources and individual scores from each source are combined into an overall confidence score. Gene-gene interactions can be extracted from public sources including STRING, UniProt, COSMIC, BioPlex, and NCBI Gene as previously described (Veselkov,. et al, 2019). This results in a gene-gene interactome dataset containing 20,256 genes with ˜11 million interactions.


The gene profile for each compound can be represented as a sparse matrix, in which a ‘1’ indicates genes that directly interact with the compound and a ‘0’ for all other genes. The network propagation (Random Walk with Restart) algorithm can then applied to spread this gene profile onto the gene-gene interactome. As a result, a genome-wide profile of gene scores can be obtained for each compound. The restart parameter ‘c’ can be considered as an adjustable parameter for ML model optimization.


The LINCS compounds that are already known the regulation direction for a certain gene can be used to train a linear SVM classification model using network propagation profile as features. The regularization parameter ′C; can be optimized during the model training using a nested cross-validation strategy. The F-score, that balances sensitivity and specificity, can be used to evaluate the outcome of each model. The best model can be defined as a model with the highest F-score. The selected model can then applied to the FooDB compound dataset. The probability estimates for the desired regulation direction of each food compound can be calculated and can be used as a score to determine the probability of each intervention.


Treating Immunotypes to Lower iAge


Table 3 below lists the protein markers that can have their levels changed to improve the iAge for patients in the different immunotypes.









TABLE 3







Protein Markers to Change for Each Immunotype










Protein Levels to change
ImmunoType







TRAIL
SH1



GroA/IFNG/MIG
SH2, N1



None
SH3



MIG
SH4



TRAIL/IFNG
N2




custom-charactercustom-character

N3



GroA/IFNG/TRAIL
N4



Eotaxin/GroA/MIG/IFNG
N5



Eotaxin/MIG/TRAIL
N6










Table 4 below shows GRAS (generally regarded as safe) compounds that can be used to lower each of the five protein markers in the Immunotypes.









TABLE 4







GRAS Compounds for Each Protein Marker










Protein Marker
GRAS Compounds







TRAIL
iron/biotin/caffeine



GroA
iron/manganese chloride/niacin/carrageenan



IFNg
manganese chloride/beta carotene/leutin/zinc




sulfate



MIG
vitamin d2/niacin/guar gum



Eotaxin
kawain/indole-3-carbinol/picetannol










The level of one or more of the five protein markers can be improved for iAge by providing a patient with one or more of the GRAS compounds listed in Table 4. Table 3 above lists the proteins markers to improve for each immunotype. Table 4 identifies GRAS compounds that can be used for each protein marker to improve iAge. For example, a patient in Immunotype SH1 can be administered one or more of iron, biotin and/or caffeine. Table 5 below shows the GRAS compounds that can be used to improve iAge for the ten immunotypes.









TABLE 5







GRAS Compounds for Improving iAge of Each Immunotype











Immuno-






type
intervention 1
intervention 2
intervention 3
intervention 4





1-1
iron/biotin/






caffeine


1-2, 2-1
iron/manganese
manganese
vitamin



chloride/niacin/
chloride/beta
d2/niacin/guar



carrageenan
carotene/leutin/
gum




zinc sulfate


1-3


1-4
vitamin



d2/niacin/guar



gum


2-2
iron/biotin/
manganese



caffeine
chloride/beta




carotene/leutin/




zinc sulfate


2-3
iron/manganese
manganese
kawain/indole-3-



chloride/niacin/
chloride/beta
carbinol/



carrageenan
carotene/leutin/
picetannol




zinc sulfate


2-4
iron/biotin/
iron/manganese
manganese



caffeine
chloride/niacin/
chloride/beta




carrageenan
carotene/leutin/





zinc sulfate


2-5
iron/manganese
manganese
vitamin
kawain/indole-3-



chloride/niacin/
chloride/beta
d2/niacin/guar
carbinol/



carrageenan
carotene/leutin/
gum
picetannol




zinc sulfate


2-6
iron/biotin/
vitamin
kawain/indole-3-



caffeine
d2/niacin/guar
carbinol/




gum
picetannol









Table 10 shows other agents that can be administered to patients to improve the levels of Eotaxin, GroaA, INFg, MIG, and/or TRAIL and lower the patient's iAge. The agents to be used can depend upon the patient's immunotype (see Table 3 and 4). Combinations of agents can be made (e.g., see Table 5) for individual immunotypes using agents from Table 4. One or more, or all of the GRAS compounds listed in each entry of the table can be administered to a subject.


Additional agents that can be used to improve the levels of Eotaxin, GroaA, INFg, MIG, and/or TRAIL and lower the patient's iAge are in Table 6. The agents in Table 6 can be used as described above to improve the levels of Eotaxin, GroaA, INFg, MIG, and/or TRAIL and lower the iAge of patients in certain immunotypes. In addition, combinations of one or more of these agents in Table 6 can be used to improve the levels of Eotaxin, GroaA, INFg, MIG, and/or TRAIL and lower the iAge of patients in certain immunotypes.


The inventions disclosed herein will be better understood from the experimental details which follow. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the inventions as described more fully in the claims which follow thereafter. Unless otherwise indicated, the disclosure is not limited to specific procedures, materials, or the like, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.


EXAMPLES
Example 1: iAge Correlates with Naïve CD8(+) T Cells and with the Ex Vivo Jak-STAT Signaling Responses to Stimulation

The frequency of circulating naïve CD8(+) T cells decline with high iAge (A) and iAge can predict poor ex vivo Jak-STAT signaling responses to stimulation (B and C). A total of 96 cytokine-cell-STAT combinations were analyzed with respect of a subject's iAge. These included eight cell types: B cells, CD4(+) T cells (and their CD45(+) and (−) subsets), CD8(+) T cells (and their CD45(+) and (−) subsets), and monocytes; four cytokines: Interleukin-6 (IL-6), IL-10, IL-21 and Interferon-alpha; and three STAT proteins (STAT1, 3 and 5). FIG. 1B: volcano plot, result of a multiple regression analysis with permutation tests to estimate false discovery rates (Benjamini-Hochberg FDR) (y-axis) as a function of the regression coefficients obtained for iAge after adjusting for Age, Gender and cytomegalovirus status. FIG. 1C: normalized ex vivo CD8(+) T cell phosphor-STAT-1 responses to Interleukin-6. The lower tertile for iAge shows significantly more robust responses than the higher tertile for iAge (C).


iAge is negatively correlated with naïve CD8(+) T cells and with the ex vivo Jak-STAT signaling responses to stimulation.


Example 2: Stratification of Cancer Patients Using iAge and CRS

A blood sample is obtained from patients prior to immunotherapy treatment. Serum and immune cells are separated by standard methods. Serum samples are used to measure protein concentration for inflammatory age (iAge) determination; and cells are cytokine-stimulated ex vivo to measure phosphorylation of intracellular signal transducer and activator of transcription (STAT) proteins to derive a cytokine response score (CRS). iAge and CRS can independently predict patient's response to immunotherapy treatment. FIG. 2 shows a flow diagram of this process.


iAge and CRS can be used to stratify cancer patients prior to treatment as responders versus non-responder for immunotherapy.


Example 3: Stratification of Cancer Patients Using iAge

iAge can be used to classify cancer patients into responder and non-responders to immunotherapy treatment (A), and to derive iAge individual inflammatory protein signature (barcode), which is fed to iAge personalized recommendation engine to create an individualized initial therapy aimed to reduce iAge, inform medical decision and hence, convert those non-responder patients into responder patients (suitable for immunotherapy) (B). FIG. 3 shows a flow diagram of this process.


iAge is used to stratify patients for cancer immunotherapy and help convert non-responders into responder for immunotherapy.


Example 4: Endothelial Cells Derived from hiPSCs Produce MIG

Human induced pluripotent stem cells (hiPSCs) were obtained from isolated fibroblasts (N=5, in duplicates) using the Yamanaka factors (Takahashi and Yamanaka, 2006) and differentiated them into endothelial cells (hiPSC-ECs) under well-defined conditions as previously described (Hu et al., 2016).


Expression levels of MIG and SIRT3 were measured by RT-PCR. A significant age-dependent increase in MIG mRNA expression levels is observed (P<0.01), which reaches a plateau after the sixth cell passage. (See FIG. 4) Concomitant with the increase in MIG, down-regulation in SIRT3 mRNA can be observed after the second cell passage (P<0.01). (See FIG. 4)


Example 5: Expression of CXCR3 in Endothelial Cells

Human induced pluripotent stem cells were made as described in Example 4. Expression of the MIG receptor, CXCR3, was measured in young cardiomyocytes derived from hiPSCs (hiPSC-CM) as well as in hiPSC-ECs (endothelial cells derived from hiPSC), HUVEC cells, freshly isolated fibroblasts and hiPSCs.


Elevated expression of CXCR3 is observed in hiPSC-ECs, HUVEC cells but not in other cell types (F) suggesting that the endothelium but not other cell subsets is a target of MIG and potentially other CXCR3 ligands as well. (See FIG. 5)


Example 6: MIG Impairs Endothelial Cell Function

Mouse thoracic aortas were carefully dissected, and vessels were cut into small rings and mounted on an isometric wire myograph chambers (Danish Myo Technology) and subjected to a normalization protocol. Following normalization, the vessels were incubated with either PBS or different concentrations of recombinant mouse MIG (R&D systems, catalog number 492-MM). A concentration-dependent contraction curve was created by accumulative application of the prostaglandin agonist U46619. Subsequently, concentration-dependent relaxation curves of Acetylcholine were conducted on these vessels and percent relaxation calculated for each dose.



FIG. 6 shows a line graph of percent relaxation of mouse thoracic aortic sections to Acetylcholine after exposure to different amounts of MIG. FIG. 6 shows impaired vascular reactivity with increasing concentrations of MIG. MIG causes a dose-dependent effect on vasorelaxation in treated aortas demonstrating that MIG impairs vascular function, and can contribute to arterial stiffness and premature aging of the cardiovascular system.


Example 7: Compositions for Treating Immunotypes

Tables 2 to 4 show agents that can be used to lower iAge by improving the levels of the markers Eotaxin, GroA, IFNg, MIG, and/or TRAIL. Table 4 shows how the treatment of certain markers correlates to immunotype, and Table 5 shows combinations of agents that can administer to patients in the different immunotypes to improve their iAge.


Table 6 below, shows dosages that can be used in formulating the agents into compositions that can be administered to patients of certain immunotypes.









TABLE 6







Dose of Agents for Improving Levels


of an iAge Marker in a Patient











Agent
Marker
Dosage per day
















iron
TRAIL/GroA
45
mg



manganese chloride
IFNG/GroA
9
mg



vitamin D2
MIG
.05
mg



niacin
MIG/GroA
250
mg



guar gum
MIG
2000
mg



kawain
Eotaxin
125
mg



indole-3-carbinol
Eotaxin
400
mg



L-methonine
Eotaxin
500
mg



Piceatannol
Eotaxin
500
mg



biotin
TRAIL
10
mg



caffeine
TRAIL
200
mg



beta-carotene
IFNG
15
mg



leutin
IFNG
20
mg



zinc sulfate
IFNG
220
mg



Carrageenan
GroA
450
mg










For example, treatment of patients from Immunotypes SH2 or N1 can improve the levels of GroA, IFNg, and MIG. A composition for improving GroA can have iron, manganese chloride, niacin, and carrageenan. The dose of each of these per day is 45 mg iron, 9 mg manganese chloride, 250 mg niacin, and 450 mg carrageenan. A composition for improving IFNg can have manganese chloride, beta carotene, leutin, and zinc sulfate. The dose of each of these is 15 mg beta-carotene, 20 mg leutin, 220 mg zinc sulfate, and the manganese chloride doses is the same as for GroA. A composition for improving MIG can have vitamin d2, niacin, and guar gum. The dose of each of these is 0.05 mg vitamin d2, 2000 mg guar gum, and dose of niacin is the same as for GroA. These compositions can be combined into one dosage form, placed in separate dosage forms, or the components of each can be mixed and matched into separate dosage forms for administering to a patient.


Using Tables 3, 4, and 5 combined with the dosing in Table 6 one can make dosage forms containing agents that can improve the iAge of patients in any of the immunotypes as was shown above for Immunotypes SH2 or N1.


Example 8: Gene Expression iAge (GE iAge)

The Framingham Heart Study gene-expression, phenotypic clinical data, and longitudinal survival data, were downloaded from dbGap and preprocessed as detailed in Alpert et al, A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat Med. 2019; 25:487-495. The enrichment of the gene-signature in the Framingham Heart Study samples was calculated using single-sample gene-set enrichment analysis, Foroutan M, Bhuva DD, Lyu R, Horan K, Cursons J and Davis M J. Single sample scoring of molecular phenotypes. BMC Bioinformatics. 2018; 19:404.


To evaluate iAge in the Framingham Heart Study, we used the gene expression signature of iAge as described above. For survival analysis, we calculated a multivariate Cox regression model regressing all-cause mortality against the clinical covariates: age, gender, smoking status, diabetes, total cholesterol, HDL cholesterol, blood pressure, a cardiovascular disease status assessed on the date of the 8th exam, and the iAge score.


We observed that gene expression iAge was significantly associated with all-cause mortality following adjustment to multiple covariates associated with mortality, including age, gender, smoking, cholesterol levels, blood pressure, diabetes, and existence of a cardiovascular disease (p=0.02, cox proportional hazard model, N=2,290 individuals).


All publications, patents and patent applications discussed and cited herein are incorporated herein by reference in their entireties. It is understood that the disclosed invention is not limited to the particular methodology, protocols and materials described as these can vary. It is also understood that the terminology used herein is for the purposes of describing particular embodiments only and is not intended to limit the scope of the present invention which will be limited only by the appended claims.


Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.









TABLE 7







Molecules that Change iAge











Compound
pval
q_value
hgnc_symbol
combined.score














Indomethacin
9.21E−14
1.28E−11
CXCL9
786


Dinoprostone
7.08E−12
7.88E−10
CXCL9
912


Celecoxib
4.46E−11
3.89E−09
CXCL9
786


Calcium
1.41E−09
9.41E−08
CXCL9
907


Histamine
2.08E−09
1.29E−07
CXCL9
909


Norepinephrine
2.99E−05
0.00075721 
CXCL9
900


Adenosine
4.93E−05
0.001158718
CXCL9
900


Vidarabine
4.93E−05
0.001158718
CXCL9
900


Acetylcholine
5.26E−05
0.001220383
CXCL9
923


Melatonin
0.000291384
0.005289249
CXCL9
900


Dopamine
0.000810821
0.012654873
CXCL9
900


Yohimbine
0.000914411
0.014009785
CXCL9
900


Caffeine
0.001416067
0.019872531
CXCL9
900


Epinephrine
0.001477318
0.020559348
CXCL9
900


Arbutin
0.001939689
0.026123226
CXCL9
900


Chloramphenicol
0.002229433
0.02863964 
CXCL9
900


Camphor
0.002436302
0.030591166
CXCL9
900


Papaverine
0.003039802
0.036521365
CXCL9
900


Bortezomib
1.45E−24
2.42E−21
TNFSF10
756


Simvastatin
1.54E−17
6.41E−15
TNFSF10
700


Acetylsalicylic_acid
4.48E−15
8.31E−13
TNFSF10
700


Fluorouracil
1.46E−11
1.52E−09
TNFSF10
760


Sorafenib
5.16E−11
4.11E−09
TNFSF10
727


Vorinostat
6.65E−10
4.63E−08
TNFSF10
751


Azacitidine
6.29E−07
2.50E−05
TNFSF10
700


Capsaicin
1.95E−06
6.66E−05
TNFSF10
700


Zucapsaicin
1.95E−06
6.66E−05
TNFSF10
700


Lovastatin
0.000266545
0.005001469
TNFSF10
700


Irinotecan
0.000398311
0.006928944
TNFSF10
767


Acetylcysteine
0.000575174
0.009510311
TNFSF10
724


Oxaliplatin
0.001395422
0.019748775
TNFSF10
773


Ribavirin
0.001491127
0.020580019
TNFSF10
700


Simvastatin
1.54E−17
6.41E−15
IFNG
721


Tretinoin
4.61E−16
1.28E−13
IFNG
959


Alitretinoin
4.61E−16
1.28E−13
IFNG
959


Calcitriol
4.08E−15
8.31E−13
IFNG
852


Nitric_Oxide
8.45E−14
1.28E−11
IFNG
885


Dinoprostone
7.08E−12
7.88E−10
IFNG
556


Fluorouracil
1.46E−11
1.52E−09
IFNG
730


Chromium_Cr-51
4.66E−11
3.89E−09
IFNG
774


Pentoxifylline
2.63E−09
1.57E−07
IFNG
914


Budesonide
3.59E−09
2.06E−07
IFNG
728


Dexamethasone
5.51E−09
2.88E−07
IFNG
700


Betamethasone
5.51E−09
2.88E−07
IFNG
700


Isoprenaline
1.94E−06
6.66E−05
IFNG
700


Benzylpenicillin
5.06E−06
0.000156574
IFNG
953


Decitabine
1.02E−05
0.000294988
IFNG
700


Titanium_dioxide
1.23E−05
0.000348701
IFNG
700


Fenofibrate
0.000204957
0.003979987
IFNG
800


Dronabinol
0.000285001
0.005230242
IFNG
751


Glucosamine
0.000561713
0.009380608
IFNG
800


Acetylcysteine
0.000575174
0.009510311
IFNG
819


Morphine
0.00359675 
0.041712305
IFNG
827


Calcium
1.41E−09
9.41E−08
CCL11
700


Budesonide
3.59E−09
2.06E−07
CCL11
931


Losartan
3.47E−06
0.00011148 
CCL11
629


Ethanol
0.000228915
0.004344177
CCL11
674


Montelukast
0.000276008
0.005121491
CCL11
924


Morphine
0.00359675 
0.041712305
CCL11
629


Nitric_Oxide
8.45E−14
1.28E−11
CXCL1
687


Dinoprostone
7.08E−12
7.88E−10
CXCL1
925


Calcium
1.41E−09
9.41E−08
CXCL1
900


Histamine
2.08E−09
1.29E−07
CXCL1
918


Estradiol
1.66E−06
6.02E−05
CXCL1
831


Titanium_dioxide
1.23E−05
0.000348701
CXCL1
739


Sodium_Chloride
1.89E−05
0.000518049
CXCL1
694


Norepinephrine
2.99E−05
0.00075721 
CXCL1
900


Adenosine
4.93E−05
0.001158718
CXCL1
907


Vidarabine
4.93E−05
0.001158718
CXCL1
907


Acetylcholine
5.26E−05
0.001220383
CXCL1
900


Mesalazine
0.000131842
0.002729337
CXCL1
700


Melatonin
0.000291384
0.005289249
CXCL1
900


Acetylcysteine
0.000575174
0.009510311
CXCL1
676


Dopamine
0.000810821
0.012654873
CXCL1
900


Yohimbine
0.000914411
0.014009785
CXCL1
900


Oxaliplatin
0.001395422
0.019748775
CXCL1
818


Caffeine
0.001416067
0.019872531
CXCL1
900


Epinephrine
0.001477318
0.020559348
CXCL1
900


Arbutin
0.001939689
0.026123226
CXCL1
900


Chloramphenicol
0.002229433
0.02863964 
CXCL1
900


Camphor
0.002436302
0.030591166
CXCL1
900


Papaverine
0.003039802
0.036521365
CXCL1
900
















TABLE 8







Food Compounds and Other Compounds that Change iAge














hgnc
combined


Molecule
pval
q_value
symbol
score














Prostaglandin_E2
5.83E−13
9.13E−11
CXCL9
912


8-isoprostaglandin_E2
5.83E−13
9.13E−11
CXCL9
912


ADP
9.74E−13
1.46E−10
CXCL9
900


Ca2+
8.84E−12
1.22E−09
CXCL9
907


Histamine
1.78E−10
1.81E−08
CXCL9
909


Lipoxin_A4
7.55E−07
4.34E−05
CXCL9
900


D-Glutamic_acid
8.08E−07
4.57E−05
CXCL9
900


Parthenolide
1.15E−06
6.20E−05
CXCL9
900


Prostaglandin_D2
2.99E−06
0.000151552
CXCL9
900


Leukotriene_D4
5.31E−06
0.00025799
CXCL9
919


xi-Norepinephrine
6.31E−06
0.000292475
CXCL9
900


LPA(18:1(9Z)/0:0)
3.64E−05
0.001548382
CXCL9
900


Leukotriene_E4
6.45E−05
0.002647045
CXCL9
900


leukotriene-C4
0.000124578
0.004473114
CXCL9
900


5-KETE
0.000192008
0.006618506
CXCL9
900


2-Arachidonylglycerol
0.000258189
0.008351519
CXCL9
900


1-Isothiocyanato-2-phenylethane
0.000277685
0.008781464
CXCL9
900


Serotonin
0.000308348
0.009575447
CXCL9
900


(−)-alpha-Narcotine
0.000336004
0.010249596
CXCL9
900


( )-alpha-Narcotine
0.000336004
0.010249596
CXCL9
900


Quassin
0.000473125
0.01244933
CXCL9
900


Cucurbitacin_B
0.000473125
0.01244933
CXCL9
900


Artemorin
0.000473125
0.01244933
CXCL9
900


Tatridin_B
0.000473125
0.01244933
CXCL9
900


2-Ethylpyrazine
0.000473125
0.01244933
CXCL9
900


(R)-3-Hydroxyoctanoic_acid
0.000473125
0.01244933
CXCL9
900


3-Hydroxyoctanoic_acid
0.000473125
0.01244933
CXCL9
900


Salicin
0.000473125
0.01244933
CXCL9
900


2-Propenyl_glucosinolate
0.000473125
0.01244933
CXCL9
900


Sinigrin
0.000473125
0.01244933
CXCL9
900


Isohumulone_B
0.000473125
0.01244933
CXCL9
900


Amygdalin
0.000473125
0.01244933
CXCL9
900


Grosheimin
0.000473125
0.01244933
CXCL9
900


Cascarillin
0.000473125
0.01244933
CXCL9
900


Linonin
0.000538779
0.013655661
CXCL9
900


Limonin
0.000538779
0.013655661
CXCL9
900


N-Arachidonoyl_glycine
0.000538779
0.013655661
CXCL9
900


Cucurbitacin_E
0.000694933
0.016869247
CXCL9
900


Aloin
0.000890075
0.020353866
CXCL9
900


Arbutin
0.000890075
0.020353866
CXCL9
900


Falcarindiol
0.000890075
0.020353866
CXCL9
900


boman-2-one
0.001132243
0.024153542
CXCL9
900


GDP
0.001134364
0.024153542
CXCL9
900


coumarin
0.001430726
0.030071413
CXCL9
900


(R)-3-Hydroxybutyric_acid
0.002240724
0.044389521
CXCL9
900


3-Hydroxybutanoic_acid
0.002240724
0.044389521
CXCL9
900


(S)-3-Hydroxybutyric_acid
0.002240724
0.044389521
CXCL9
900


trans-resveratrol
4.13E−19
9.48E−17
TNFSF10
943


Resveratrol
4.13E−19
9.48E−17
TNFSF10
943


(Z)-Resveratrol
4.13E−19
9.48E−17
TNFSF10
943


Simvastatin
2.12E−18
4.30E−16
TNFSF10
700


Genistein
5.93E−11
6.39E−09
TNFSF10
741


Quercetin
2.63E−08
2.11E−06
TNFSF10
742


Platinum
0.000706672
0.017034249
TNFSF10
773


Celastrol
0.001210457
0.025597816
TNFSF10
700


N-acetyl--D-glucosamine
6.35E−51
5.47E−48
IFNG
800


N-acetyl-_-D-glucosamine
6.35E−51
5.47E−48
IFNG
800


N-acetyl-_-D-galactosamine
6.35E−51
5.47E−48
IFNG
800


N-Acetylglucosamine
6.35E−51
5.47E−48
IFNG
800


L-Galactose
1.59E−30
4.56E−28
IFNG
829


alpha-D-Glucopyranose
1.59E−30
4.56E−28
IFNG
829


beta-D-Galactose
1.59E−30
4.56E−28
IFNG
829


alpha-D-Galactose
1.59E−30
4.56E−28
IFNG
829


L-Gulose
1.59E−30
4.56E−28
IFNG
829


D-Galactose
1.59E−30
4.56E−28
IFNG
829


_-L-galactose
1.59E−30
4.56E−28
IFNG
829


beta-D-(1−>3)-Glucan
1.59E−30
4.56E−28
IFNG
829


trans-resveratrol
4.13E−19
9.48E−17
IFNG
824


Resveratrol
4.13E−19
9.48E−17
IFNG
824


(Z)-Resveratrol
4.13E−19
9.48E−17
IFNG
824


Guar_gum
1.54E−18
3.31E−16
IFNG
918


Simvastatin
2.12E−18
4.30E−16
IFNG
721


9_13-cis-Retinoate
2.46E−17
4.72E−15
IFNG
959


Calcitriol
4.87E−16
8.83E−14
IFNG
852


Prostaglandin_E2
5.83E−13
9.13E−11
IFNG
556


8-isoprostaglandin_E2
5.83E−13
9.13E−11
IFNG
556


ADP
9.74E−13
1.46E−10
IFNG
900


15-Deoxy-delta-12_14-
7.94E−12
1.14E−09
IFNG
824


prostaglandin_J2


Chromium
1.65E−11
2.19E−09
IFNG
774


Genistein
5.93E−11
6.39E−09
IFNG
806


Dexamethasone
2.01E−09
1.82E−07
IFNG
700


n-butanoate
4.85E−07
2.88E−05
IFNG
700


S-Nitrosoglutathione
1.15E−06
6.20E−05
IFNG
746


Parthenolide
1.15E−06
6.20E−05
IFNG
827


Rotenone
2.88E−06
0.000148094
IFNG
700


Maltotetraose
3.18E−06
0.000158638
IFNG
786


Beryllium
1.77E−05
0.000783919
IFNG
836


ATP
7.25E−05
0.002907726
IFNG
914


Adenosine_triphosphate
7.25E−05
0.002907726
IFNG
914


2_2′-Bis(4-
0.000247643
0.008129781
IFNG
700


hydroxyphenyl)propane


Galactosamine
0.000390286
0.01149843
IFNG
800


Glucosamine
0.000390286
0.01149843
IFNG
800


beta-D-Glucosamine
0.000390286
0.01149843
IFNG
800


acrolein
0.000763576
0.018027727
IFNG
700


(−)-Morphine
0.001903742
0.0390607
IFNG
827


trans-resveratrol
4.13E−19
9.48E−17
CCL11
885


Resveratrol
4.13E−19
9.48E−17
CCL11
885


(Z)-Resveratrol
4.13E−19
9.48E−17
CCL11
885


Ca2+
8.84E−12
1.22E−09
CCL11
700


Genistein
5.93E−11
6.39E−09
CCL11
629


12-HETE
1.25E−09
1.19E−07
CCL11
629


leukotriene-C4
0.000124578
0.004473114
CCL11
875


GDP
0.001134364
0.024153542
CCL11
900


(−)-Morphine
0.001903742
0.0390607
CCL11
629


trans-resveratrol
4.13E−19
9.48E−17
CXCL1
700


Resveratrol
4.13E−19
9.48E−17
CXCL1
700


(Z)-Resveratrol
4.13E−19
9.48E−17
CXCL1
700


Prostaglandin_E2
5.83E−13
9.13E−11
CXCL1
925


8-isoprostaglandin_E2
5.83E−13
9.13E−11
CXCL1
925


ADP
9.74E−13
1.46E−10
CXCL1
900


Ca2+
8.84E−12
1.22E−09
CXCL1
900


Gallocatechin_3-gallate
2.62E−11
3.01E−09
CXCL1
629


ent-Gallocatechin_3-gallate
2.62E−11
3.01E−09
CXCL1
629


ent-Epigallocatechin_3-gallate
2.62E−11
3.01E−09
CXCL1
629


Epigallocatechin_3-gallate
2.62E−11
3.01E−09
CXCL1
629


Histamine
1.78E−10
1.81E−08
CXCL1
918


Epiestradiol
3.21E−07
2.05E−05
CXCL1
831


Silica
3.65E−07
2.29E−05
CXCL1
707


n-butanoate
4.85E−07
2.88E−05
CXCL1
629


Lipoxin_A4
7.55E−07
4.34E−05
CXCL1
900


D-Glutamic_acid
8.08E−07
4.57E−05
CXCL1
900


Parthenolide
1.15E−06
6.20E−05
CXCL1
900


Prostaglandin_D2
2.99E−06
0.000151552
CXCL1
900


Leukotriene_D4
5.31E−06
0.00025799
CXCL1
900


xi-Norepinephrine
6.31E−06
0.000292475
CXCL1
900


LPA(18:1(9Z)/0:0)
3.64E−05
0.001548382
CXCL1
900


Leukotriene_E4
6.45E−05
0.002647045
CXCL1
900


leukotriene-C4
0.000124578
0.004473114
CXCL1
900


5-KETE
0.000192008
0.006618506
CXCL1
900


2-Arachidonylglycerol
0.000258189
0.008351519
CXCL1
900


1-Isothiocyanato-2-phenylethane
0.000277685
0.008781464
CXCL1
900


Serotonin
0.000308348
0.009575447
CXCL1
900


(−)-alpha-Narcotine
0.000336004
0.010249596
CXCL1
900


( )-alpha-Narcotine
0.000336004
0.010249596
CXCL1
900


Quassin
0.000473125
0.01244933
CXCL1
900


Cucurbitacin_B
0.000473125
0.01244933
CXCL1
900


Artemorin
0.000473125
0.01244933
CXCL1
900


Tatridin_B
0.000473125
0.01244933
CXCL1
900


2-Ethylpyrazine
0.000473125
0.01244933
CXCL1
900


(R)-3-Hydroxyoctanoic_acid
0.000473125
0.01244933
CXCL1
900


3-Hydroxyoctanoic_acid
0.000473125
0.01244933
CXCL1
900


Salicin
0.000473125
0.01244933
CXCL1
900


2-Propenyl_glucosinolate
0.000473125
0.01244933
CXCL1
900


Sinigrin
0.000473125
0.01244933
CXCL1
900


Isohumulone_B
0.000473125
0.01244933
CXCL1
900


Amygdalin
0.000473125
0.01244933
CXCL1
900


Grosheimin
0.000473125
0.01244933
CXCL1
900


Cascarillin
0.000473125
0.01244933
CXCL1
900


Linonin
0.000538779
0.013655661
CXCL1
900


Limonin
0.000538779
0.013655661
CXCL1
900


N-Arachidonoyl_glycine
0.000538779
0.013655661
CXCL1
900


Cucurbitacin_E
0.000694933
0.016869247
CXCL1
900


Platinum
0.000706672
0.017034249
CXCL1
818


Aloin
0.000890075
0.020353866
CXCL1
900


Arbutin
0.000890075
0.020353866
CXCL1
900


Falcarindiol
0.000890075
0.020353866
CXCL1
900


boman-2-one
0.001132243
0.024153542
CXCL1
900


GDP
0.001134364
0.024153542
CXCL1
900


coumarin
0.001430726
0.030071413
CXCL1
900


(R)-3-Hydroxybutyric_acid
0.002240724
0.044389521
CXCL1
900


3-Hydroxybutanoic_acid
0.002240724
0.044389521
CXCL1
900


(S)-3-Hydroxybutyric_acid
0.002240724
0.044389521
CXCL1
900
















TABLE 9







Drugs that Can Change iAge










DRUG
GENE
CHANGE
EFFECT





Arsenic trioxide
CXCL9
downregulated
Beneficial


Selenium
CXCL9
downregulated
Beneficial


Beta carotene
CXCL9
upregulated
Detrimental


Formaldehyde
CXCL9
upregulated
Detrimental


Gadodiamide
CXCL9
upregulated
Detrimental


Progesterone
CXCL9
upregulated
Detrimental


Silicon dioxide
CXCL9
upregulated
Detrimental


Vincristine
CXCL9
upregulated
Detrimental


Acetaminophen
TNFSF10
downregulated
Detrimental


Acetylcysteine
TNFSF10
downregulated
Detrimental


Arsenic trioxide
TNFSF10
downregulated
Detrimental


Estradiol
TNFSF10
downregulated
Detrimental


Bortezomib
TNFSF10
downregulated
Detrimental


Chromium
TNFSF10
downregulated
Detrimental


Curcumin
TNFSF10
downregulated
Detrimental


Ciclosporin
TNFSF10
downregulated
Detrimental


Progesterone
TNFSF10
downregulated
Detrimental


Selenium
TNFSF10
downregulated
Detrimental


Silicon dioxide
TNFSF10
downregulated
Detrimental


Silver
TNFSF10
downregulated
Detrimental


Selenium
TNFSF10
downregulated
Detrimental


Vitamin E
TNFSF10
downregulated
Detrimental


Alitretinoin
TNFSF10
upregulated
Beneficial


Aluminum oxide
TNFSF10
upregulated
Beneficial


Arsenic trioxide
TNFSF10
upregulated
Beneficial


Beta carotene
TNFSF10
upregulated
Beneficial


Estradiol
TNFSF10
upregulated
Beneficial


Capsaicin
TNFSF10
upregulated
Beneficial


Ciclosporin
TNFSF10
upregulated
Beneficial


Cytarabine
TNFSF10
upregulated
Beneficial


Decitabine
TNFSF10
upregulated
Beneficial


Cisplatin
TNFSF10
upregulated
Beneficial


Doxorubicin
TNFSF10
upregulated
Beneficial


Fluorouracil
TNFSF10
upregulated
Beneficial


Formaldehyde
TNFSF10
upregulated
Beneficial


Iron
TNFSF10
upregulated
Beneficial


Methotrexate
TNFSF10
upregulated
Beneficial


Oxygen
TNFSF10
upregulated
Beneficial


Pentoxifylline
TNFSF10
upregulated
Beneficial


Oxyquinoline
TNFSF10
upregulated
Beneficial


Isotretinoin
TNFSF10
upregulated
Beneficial


Tretinoin
TNFSF10
upregulated
Beneficial


Vincristine
TNFSF10
upregulated
Beneficial


Vorinostat
TNFSF10
upregulated
Beneficial


Alitretinoin
IFNG
downregulated
Detrimental


Sodium stibogluconate
IFNG
downregulated
Detrimental


Clozapine
IFNG
downregulated
Detrimental


Dronabinol
IFNG
downregulated
Detrimental


Dexamethasone
IFNG
downregulated
Detrimental


Formaldehyde
IFNG
downregulated
Detrimental


Histamine
IFNG
downregulated
Detrimental


Methotrexate
IFNG
downregulated
Detrimental


Nitroprusside
IFNG
downregulated
Detrimental


Resveratrol
IFNG
downregulated
Detrimental


Ribavirin
IFNG
downregulated
Detrimental


Theophylline
IFNG
downregulated
Detrimental


Tretinoin
IFNG
downregulated
Detrimental


Cimetidine
IFNG
upregulated
Beneficial


Formaldehyde
IFNG
upregulated
Beneficial


Gadodiamide
IFNG
upregulated
Beneficial


Gadolinium
IFNG
upregulated
Beneficial


Oxygen
IFNG
upregulated
Beneficial


Simvastatin
IFNG
upregulated
Beneficial


Vincristine
IFNG
upregulated
Beneficial


Zinc
IFNG
upregulated
Beneficial


Zinc sulfate
IFNG
upregulated
Beneficial


Methotrexate
CCL11
downregulated
Beneficial


Resveratrol
CCL11
downregulated
Beneficial


Tacrolimus
CCL11
downregulated
Beneficial


Zinc
CCL11
upregulated
Detrimental


Alitretinoin
CXCL1
downregulated
Detrimental


Clozapine
CXCL1
downregulated
Detrimental


Curcumin
CXCL1
downregulated
Detrimental


Calcitriol
CXCL1
downregulated
Detrimental


Sodium aurothiomalate
CXCL1
downregulated
Detrimental


Methotrexate
CXCL1
downregulated
Detrimental


Methylprednisolone
CXCL1
downregulated
Detrimental


Mifepristone
CXCL1
downregulated
Detrimental


Piroxicam
CXCL1
downregulated
Detrimental


Prednisolone
CXCL1
downregulated
Detrimental


Progesterone
CXCL1
downregulated
Detrimental


Resveratrol
CXCL1
downregulated
Detrimental


Isotretinoin
CXCL1
downregulated
Detrimental


Tretinoin
CXCL1
downregulated
Detrimental


Tretinoin
CXCL1
downregulated
Detrimental


Estradiol
CXCL1
upregulated
Beneficial


Bortezomib
CXCL1
upregulated
Beneficial


Eugenol
CXCL1
upregulated
Beneficial


Formaldehyde
CXCL1
upregulated
Beneficial


Gemcitabine
CXCL1
upregulated
Beneficial


Methotrexate
CXCL1
upregulated
Beneficial


Selenium
CXCL1
upregulated
Beneficial


Silicon dioxide
CXCL1
upregulated
Beneficial


Silicon dioxide
CXCL1
upregulated
Beneficial


Ursodeoxycholic acid
CXCL1
upregulated
Beneficial
















TABLE 10







Agents for Lowering Inflammatory Age












Compound
CCL11
CXCL1
IFNG
CXCL9
TRAIL





BRD-K60297835
X
X

X
X


BRD-K94390040
X
X

X


nickel monoxide
X
X

X


BRD-A55554369
X
X


X


Copper
X
X


X


VU-0420363-1
X
X


BRD-K02701279
X
X


3,3′,4,5′-tetrahydroxystilbene
X

X


BRD-K75040614
X


X
X


alda-1
X


X


kawain
X


X


cycloserine
X



X


Virolongin B
X


Methionine sulfoxide
X


W-5
X


BRD-K64970062
X


Cacodylic Acid
X


indole-3-carbinol
X


TUL-XXI039

X
X
X


decabromobiphenyl ether

X
X
X


Cobalt

X
X
X


BRD-K88668992

X
X

X


Paraquat

X
X

X


gadolinium chloride

X
X

X


manganese chloride

X
X


Acrolein

X
X


Aldehydes

X
X


Colchicine

X
X


Dietary Sucrose

X
X


gallium arsenide

X
X


cinnamic aldehyde

X
X


4-phenylenediamine

X
X


zinc protoporphyrin

X
X


Dinitrofluorobenzene

X
X


Ursodeoxycholic Acid

X
X


22-hydroxycholesterol

X
X


mono-(2-ethylhexyl)phthalate

X
X


2-methyl-4-isothiazolin-3-one

X
X


tcdA protein, Clostridium difficile

X
X


trovafloxacin

X
X


C646 compound

X
X


fluoranthene

X
X


iodopravadoline

X
X


ginsan

X
X


1,2-Dihydroxybenzene-3,5-Disulfonic Acid

X
X


Disodium Salt


neocuproine

X
X


Polycyclic Aromatic Hydrocarbons

X
X


pyrrolidine dithiocarbamic acid

X
X


3-(2-hydroxy-4-(1,1-dimethylheptyl)phenyl)-4-

X
X


(3-hydroxypropyl)cyclohexanol


trimethyltin

X
X


Amino Acids, Peptides, and Proteins

X
X


3-(4-methylphenylsulfonyl)-2-propenenitrile

X
X


4-methyl-N1-(3-phenylpropyl)benzene-1,2-

X
X


diamine


benzyloxycarbonylleucyl-leucyl-leucine

X
X


aldehyde


Folic Acid

X
X


Acetylmuramyl-Alanyl-Isoglutamine

X
X


lipopolysaccharide, E coli O55-B5

X
X


curdlan

X
X


BRD-K37940862

X

X
X


ST-4066738

X

X
X


hydroquinine

X

X
X


nalbuphine

X

X
X


BRD-K78206606

X

X
X


BRD-K03857568

X

X
X


RS-16566

X

X
X


BRD-K80094086

X

X
X


cytarabine

X

X
X


BRD-K79554012

X

X
X


BRD-K88329126

X

X
X


balsalazide

X

X
X


BRD-K01229043

X

X
X


BRD-K72429558

X

X
X


tyrphostin-B44

X

X
X


droxinostat

X

X
X


BRD-K68863575

X

X
X


pyroxamide

X

X
X


GW-9662

X

X
X


metformin

X

X
X


BRD-K60067222

X

X
X


lopinavir

X

X
X


BRD-K03358980

X

X
X


karakoline

X

X
X


LY-225910

X

X
X


DMAB-anabaseine

X

X
X


nTZDpa

X

X
X


GW-9508

X

X
X


BRD-A09164687

X

X
X


L-694247

X

X
X


BRD-A25829966

X

X
X


chlorphensin

X

X
X


BRD-K75308990

X

X
X


BRD-K45256078

X

X
X


CAY-10415

X

X
X


BRD-K41775742

X

X
X


BRD-K32111336

X

X
X


BRD-K67090983

X

X
X


nateglinide

X

X
X


BRD-K83998179

X

X
X


BRD-A16581344

X

X
X


KU-C103656

X

X
X


BRD-K70751730

X

X
X


mianserin

X

X
X


PSH-008

X

X
X


BRD-K33297438

X

X
X


niacin

X

X


LY-341495

X

X


KU-C103867

X

X


nitrazepam

X

X


BRD-K11841836

X

X


BRD-K78167523

X

X


tolazoline

X

X


GW-311616

X

X


idebenone

X

X


BRD-K73082558

X

X


1-benzylimidazole

X

X


BRD-K13318664

X

X


phenprobamate

X

X


BRD-A65516924

X

X


trap-101

X

X


benidipine

X

X


cetraxate

X

X


YM-298198

X

X


BRD-K92458042

X

X


KU-C104218

X

X


triclabendazole

X

X


BRD-K55630727

X

X


BRD-K04238564

X

X


ARG-CSC-91

X

X


BRD-K57019930

X

X


SCH-442416

X

X


BRD-K66896231

X

X


BRD-K05672354

X

X


ICI-204448

X

X


mestinon

X

X


BRD-K16358524

X

X


BRD-K89412978

X

X


BRD-K99381377

X

X


NSC-119889

X

X


VU-0366968

X

X


MW-MITF22

X

X


EMF-sumol-6

X

X


Glafenine

X

X


Croton Oil

X

X


alpha-Chlorohydrin

X

X


JNJ-10191584

X


X


BRD-K28284219

X


X


bongkrek-acid

X


X


ICI-118551

X


X


BRD-K31851742

X


X


BRD-K90430314

X


X


tyrphostin-AG-126

X


X


levomepromazine

X


X


BRD-K01578344

X


X


FTase-inhibitor-B581

X


X


BRD-A38793261

X


X


BRD-K65983740

X


X


taxifolin

X


X


cytosporone-b

X


X


JAS07-009

X


X


BRD-K02223034

X


X


cefoxitin

X


X


BRD-A13367427

X


X


bemesetron

X


X


dicloxacillin

X


X


JFD02227

X


X


chloramphenicol

X


X


BRD-K22831754

X


X


BRD-K34711351

X


X


BRD-K43267173

X


X


apoptosis-activator-II

X


X


BW-723C86

X


X


dipropyl-5ct

X


X


phentermine

X


X


SQ-22536

X


X


BRD-K98439022

X


X


BRD-K09460252

X


X


bimatoprost

X


X


YL-61

X


X


AR-C133057XX

X


X


neurodazine

X


X


LE-300

X


X


xanthoxyline

X


X


BRD-K61679744

X


X


BRD-K37724988

X


X


BRD-K69422440

X


X


BRD-K50360938

X


X


BJM-HSC2-10

X


X


MW-RAS9

X


X


PSH-012

X


X


BRD-K44890999

X


X


BRD-K82206012

X


X


BRD-K72504866

X


X


BRD-K76425766

X


X


moexipril

X


X


MW-A1-18

X


X


6-hydroxytropinone

X


X


chromanol

X


X


BRD-K55186349

X


X


BRD-K71599932

X


X


BRD-K92754162

X


X


PSB-1115

X


X


Iron

X


X


Magnetite Nanoparticles

X


X


Succimer

X


X


Peonidin

X


Peonidin 3-[4-hydroxycinnamoyl-b-D-

X


glucopyranoside]


3,4′,5,6,8-Pentamethoxyflavone

X


5,7-Dimethoxyisoilavone

X


Ursodeoxycholic acid

X


Pentanal

X


2-mercaptobenzothiazole

X


BRD-K03176945

X


BRD-K25731886

X


BRD-K97522823

X


norethindrone

X


BRD-A05831822

X


BRD-K29345712

X


pipamperone

X


rhapontin

X


parecoxib

X


VX-222

X


tetramethyl-haematoxylone

X


BRD-A13964793

X


BRD-K89814851

X


BRD-K90610876

X


BRD-K88013557

X


BRD-K11181277

X


pseudoephedrine

X


BRD-A01973626

X


BRD-K63220166

X


fexaramine

X


BRD-K30781400

X


procyclidine

X


isamoltan

X


BRD-K61921568

X


BRD-K20401833

X


BRD-K97926541

X


buddleoflavonoloside

X


BRD-K74534237

X


BRD-K89306950

X


BRD-K21401822

X


BRD-A34421300

X


BRD-K53987746

X


KU-C103654

X


BRD-K84803513

X


BRD-K95606637

X


mepivacaine

X


BRD-K84301121

X


cefatrizine

X


BRD-K54070802

X


vicriviroc

X


methysergide

X


psoromic-acid

X


BRD-K08451418

X


BRD-K96456313

X


VU-0414833-2

X


BRD-K36490574

X


quinethazone

X


mesna

X


BRD-K41059137

X


BRD-K03339442

X


RS-45041-190

X


dihydroptaeroxylin

X


BRD-A73847809

X


nevirapine

X


BRD-K62902879

X


BRD-K59794046

X


BRD-K12707296

X


SD-6-035-B4

X


RO-19-4605

X


sulfinpyrazone

X


BRD-K08311783

X


methyl-angolensate

X


imiquimod

X


ganciclovir

X


physostigmine

X


BRD-K35568648

X


BRD-K84571636

X


rathyronine

X


BRD-A76732176

X


BAY-59-3074

X


BRD-A42969626

X


BRD-K12623437

X


VU-0366037-2

X


BRD-K41284916

X


Carrageenan

X


Diacetyl

X


Hydrochloric Acid

X


Taurocholic Acid

X


Soman

X


actein

X


CI 1044

X


erionite

X


pentanal

X


primycin

X


undecane

X


siponimod

X


Ceruletide

X


nonivamide

X


SCH 351591

X


corannulene

X


Floxacillin

X


naphthalene

X


tobacco tar

X


aflatoxin G1

X


arsenic acid

X


Naphthalenes

X


Polystyrenes

X


pseudocumene

X


zinc nitrate

X


1-nitropyrene

X


butyraldehyde

X


Dicloxacillin

X


Isoflurophate

X


n-tetradecane

X


zinc chromate

X


cobalt sulfate

X


Leukotriene C4

X


cadmium sulfate

X


propionaldehyde

X


vanadyl sulfate

X


2-dichlorobenzene

X


Asbestos, Amosite

X


cyanuric chloride

X


3 -nitrofluoranthene

X


beta-Naphthoflavone

X


Estrogens

X


Pregnenolone Carbonitrile

X


2-chloroethyl ethyl sulfide

X


2,4-dinitrobenzenesulfonic acid

X


macrophage stimulatory lipopeptide 2

X


lipopolysaccharide, Helicobacter pylori

X


Metyrapone

X


AM 251

X


HU 211

X


carvacrol

X


Gold

X


Polyethylene Glycols

X


WZ4003

X


amphotericin B, deoxycholate drug

X


combination


amlexanox

X


Antimycin A

X


Dimercaprol

X


AEOL 10150

X


Domperidone

X


SB 225002

X


JTE 607

X


1-methylanthracene

X


Oleanolic Acid

X


Lipids

X


Metoclopramide

X


BIRB 796

X


Shiga Toxin 2

X


spautin-1

X


Chlorogenic Acid

X


benazepril

X


3-methyladenine

X


Sitagliptin Phosphate

X


Blood Glucose

X


obeticholic acid

X


Peptides

X


Atropine

X


maslinic acid

X


angiotensin I (1-7)

X


corosolic acid

X


astressin-2B

X


Hexadimethrine Bromide

X


2″-galloylhyperin

X


trimethyltin chloride

X


O(2)-vinyl-1-(pyrrolidin-1-yl)diazen-1-ium-

X


1,2-diolate


muricholic acid

X


tauromuricholic acid

X


7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene

X


9,10-oxide


alisol A 24-acetate

X


3-(pyridin-3-ylsulfonyl)-5-(trifluoromethyl)-

X


2H-chromen-2-one


CAP18 lipopolysaccharide-binding protein

X


2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-

X


7H-purin-7-yl)-N-(4-


isopropylphenyl)acetamide


captax

X


Graphite

X


fullerene C60

X


Deoxycholic Acid

X


Chenodeoxycholic Acid

X


tetrabromobisphenol A

X


hexyl cinnamic aldehyde

X


resolvin D1

X


resolvin D2

X


Ketamine

X


17-hydroxy-4,7,10,13,15,19-docosahexaenoic

X


acid


tin protoporphyrin IX

X


Rimonabant

X


2-Deoxy-25-Methyldolichosterone


X
X


3-epi-2-Deoxy-25-methyldolichosterone


X
X


6-Deoxo-25-methyldolichosterone


X
X


ent-Epiafzelechin-(2alpha−>7,4alpha−>8)-


X
X


catechin


Substance P


X
X


Linalyl benzoate


X
X


4′-Methylcatechin


X
X


(−)-Epiafzelechin


X
X


Afzelechin


X
X


Calamenene


X
X


Arachin


X
X


1-S-cis-Calamenene


X
X


Lormetazepam


X
X


Xylene


X
X


Sandosapogenol


X
X


Isoeugenol benzyl ether


X
X


1,2-Dimethoxy-4-methylbenzene


X
X


Tolualdehydes (mixed o-, m-, p-)


X
X


3-Methyl-9H-carbazole-9-carboxaldehyde


X
X


3-O-trans-Coumaroylmaslinic acid


X
X


(3b,24R,25x)-26-Benzoyloxystigmast-5-ene-3-


X
X


ol


(3xi,16xi,21xi)-12-Ursene-3,16,21-triol


X
X


Epicatechin-(2beta−>7,4beta−>6)-catechin


X
X


Epicatechin-(2beta−>7,4beta−>6)-ent-


X
X


epicatechin


1-Methyl-3-(2-thiazolyl)-1H-indole


X
X


beta-Boswellic acid acetate


X
X


beta-Boswellic acid


X
X


Brein


X
X


Phyllanthusol B


X
X


Isopimaric acid


X
X


Proanthocyanidin A2


X
X


7-Hydroxy-5-methoxyflavan


X
X


Prunus inhibitor b


X
X


Epicatechin 3-(3-methylgallate)


X
X


alpha-Terpinyl cinnamate


X
X


Ornithokinin


X
X


Kitagine


X
X


Arachidoside


X
X


3-Phenyl-4-pentenal


X
X


Avenanthramide E


X
X


Carissol


X
X


cis-p-Coumaroylcorosolic acid


X
X


21b-Hydroxygypsogenin


X
X


(2a,3a)-Dihydroxy-12-ursen-24-oic acid


X
X


Flupoxam


X
X


3′-Methylcatechin


X
X


dCTP


X
X


Angiotensin II


X
X


Angiotensin IV


X
X


Bradykinin


X
X


Cobinamide


X
X


Bradykinin hydroxyproline


X
X


Kinetensin 4-8


X
X


Kinetensin


X
X


Neuromedin N (1-4)


X
X


Neuromedin N


X
X


1,5-Diferuloylquinic acid


X
X


3′-Methylepicatechin


X
X


4′-Methylepicatechin


X
X


ajmaline


X
X


ϱ-amyrin


X
X


3′-Methylepicatechin 4′-sulfate


X
X


Catechin 4′-sulfate


X
X


Ursolic acid (cis-3-O-hydroxycinnamoyl-)


X
X


Ursolic acid (trans-3-O-hydroxycinnamoyl-)


X
X


Phe-Phe-Pro-Arg


X
X


Urodilatin


X
X


Phenylalanylisoleucine


X
X


5-p-Coumaroylquinic acid


X


p-Coumaroylquinic acid


X


2-Methylbenzenethiol


X


3-Methylbenzaldehyde


X


1-(1-Methoxy-1-methylethyl)-4-


X


methylbenzene


Ginkgolide M


X


Epicatechin-(4beta−>6)-catechin 3-gallate


X


Arctiol


X


3,3′-Dihydroxy-4,5-dimethoxybibenzyl


X


3-O-Feruloylquinic acid


X


Notoginsenoside G


X


1,4-Dimethylpyrrolo[1,2-a]pyrazine


X


23-Hydroxyphysalolactone


X


alpha-Catechin


X


Germacrene D


X


2,4-Dimethylquinoline


X


Salicyaldehyde


X


Melonin


X


Cuparene


X


1-p-Menthen-3-one


X


1-Methyl-4-isobutylbenzene


X


24(28)-Dehydromakisterone


X


trans-[6]-Shogaol


X


2-Methyl-4-phenyl-2-butanol


X


Lansimide 2


X


alpha-Terpinyl formate


X


alpha-Terpinyl propanoate


X


1-Methoxy-2-methylbenzene


X


L-Menthone 1,2-glycerol ketal


X


2-Methyl-1,4-naphthalenediol bis(dihydrogen


X


phosphate)


Menadiol sodium diphosphate


X


Epicatechin-(6′−>8)-epicatechin


X


10-Methylacridone


X


4-Methylpyrrolo[1,2-a]pyrazine


X


Glycylserylprolylmethionylphenylalanylvalinamide


X


(2S,2″S,3S,3″R,4S)-3,4′,5,7-


X


Tetrahydroxyflavan(2−>7,4−>8)-3,4′,5,7-


tetrahydroxyflavan


Cabbage identification factor 1


X


Cabbage identification factor 2


X


4beta-Hydroxywithanolide E


X


6-Deoxodolichosterone


X


Physalolactone


X


Dolichosterone


X


Quillajasaponin


X


Ginsenoyne M


X


6beta-6,11-Dihydroxy-4-eudesmen-3-one


X


(S)-Phellandral


X


Dukunolide B


X


alpha-Terpineol


X


p-Menth-1-ene-8-thiol


X


(R)-alpha-Terpineol


X


Phellandral


X


Obacunone


X


Phyllanthusol A


X


Thymol methyl ether


X


3beta-Dihydroxymarasmene


X


1 alpha,15-Dihydroxymarasmene


X


Marasmen-3-one


X


(S)-alpha-Terpineol


X


alpha,alpha-Dimethylphenethyl formate


X


(S)-gamma-Calacorene


X


3,4-Dihydrocadalene


X


Yucalexin P21


X


Glechomafuran


X


Mustakone


X


Linalyl phenyl acetate


X


Procyanidin A1


X


Proanthocyanidin A1


X


Proanthocyanidin A5′


X


Prunus inhibitor a


X


Tragopogonsaponin B


X


ent-Epicatechin


X


Ginsenoside Rh5


X


3alpha-O-trans-Feruloyl-2alpha-hydroxy-12-


X


ursen-28-oic acid


alpha-Terpinyl anthranilate


X


Mintsulfide


X


Rishitinol


X


alpha-Bisabolol oxide B


X


Azaspiracid 4


X


Pediocin AcH


X


Gambiriin B3


X


Procyanidin B5


X


Brassicanal B


X


z-Clausenamide


X


Methoxybrassenin A


X


Pavetannin A2


X


(E)-1-Cinnamoylpyrrolidine


X


Epicatechin 3-p-hydroxybenzoate


X


Ginsenoside Rh7


X


Chamomillol


X


Mahuannin A


X


(2S,2′R,3S,3′R,4S)-3,4′,5,7-


X


Tetrahydroxyflavan(2−>7,4−>8)-3,3′,5,5′,7-


pentahydroxyflavan


Physapruin A


X


Zanthobisquinolone


X


(2S,2″R,3S,3″S,4S)-3,4′,5,7-


X


Tetrahydroxyflavan(2−>7,4−>8)-3,4′,5,7-


tetrahydroxyflavan


(3alpha,5alpha,7alpha)-14-Methylergosta-


X


9(11),24(28)-dien-3,7-diol


ent-Epicatechin-(4alpha−>6)-ent-epicatechin


X


18-Nor-4(19),8,11,13-abietatetraene


X


xi-11,12,13-Trinor-7-calamenone


X


p-Tolyl phenylacetate


X


5-Androstene-3b,16b,17a-triol


X


5-Androstene-3b,16a,17a-triol


X


5-Androstenetriol


X


dCMP


X


dCDP


X


Morphiceptin


X


Demethylated antipyrine


X


Pseudoecgonyl-CoA


X


3-Methylene-indolenine


X


Oxytocin 1-8


X


5-Androstene-3alpha,16beta,17beta-triol


X


benzylmethyl ether


X


1-methyl-5-methylene-8-(1-methylethyl)-1,6-


X


cyclodecadiene


1,7,7-trimethylbicyclo[2.2.1]heptan-2-ol


X


benzoate


3′-Methylcatechin 4′-glucuronide


X


3′-Methylepicatechin 4′-glucuronide


X


Quadrangularin A


X


Phe-Pro-Ile


X


Val-Phe-Val-Tyr


X


(23S)-23,25-Dihydroxy-24-oxovitamine D3


X
X


23-(beta-glucuronide)


25-Hydroxyvitamin D2 25-(beta-glucuronide)


X
X


25-Hydroxyvitamin D2-25-glucuronide


X
X


Ercalcitriol


X
X


BRD-K50634134


X
X


PNU-96415E


X
X


Metformin


X
X


ornidazole


X

X


MW-STK33-1B


X

X


tripterine


X

X


Plicamycin


X

X


3,5-bis(2-fluorobenzylidene)piperidin-4-one


X

X


Pentachlorophenol


X

X


Dichloroacetic Acid


X

X


ϲ-zeacarotene


X


beta-Zeacarotene


X


gamma-Carotene


X


1′-hydroxy-œ≥-carotene


X


(9Z)-beta,beta-Carotene


X


cis-b,b-Carotene


X


delta-Carotene


X


Licoricesaponin F3


X


alpha-Carotene


X


Isozeaxanthin


X


Dehydro-beta-carotene


X


Licoricesaponin E2


X


(3R,3′R,all-E)-Zeaxanthin


X


(3R,3′R,13Z)-Zeaxanthin


X


(3R,3′R,15Z)-Zeaxanthin


X


(3R,3′R,9Z)-Zeaxanthin


X


(3S,3′S,all-E)-Zeaxanthin


X


epsilon-Carotene


X


Vitamin D5


X


Apo-9-zeaxanthinone


X


Licoricesaponin J2


X


Vitamin A2 aldehyde


X


5,6-trans-Vitamin D3


X


Acetylsoyasaponin A6


X


Acetylsoyasaponin A4


X


Acetylsoyasaponin A5


X


Soyasaponin II


X


Dehydrosoyasaponin I


X


Soyasapogenol B 3-O-[a-L-rhamnosyl-(1−>2)-


X


[b-D-glucosyl-(1−>3)]-b-D-galactosyl-(1−>2)-


b-D-glucuronide] 22-O-[b-D-glucosyl-(1−>2)-


a-L-arabinoside]


Acetylsoyasaponin A3


X


Acetylsoyasaponin A1


X


Acetylsoyasaponin A2


X


Soyasaponin A-d


X


Soyasaponin IV


X


Soyasapogenol B 3-O-b-D-glucuronide


X


Soyasaponin III


X


Soyasaponin V


X


ϳ-carotene


X


ϱ-carotene


X


(13Z)-beta-Carotene


X


beta-Carotene


X


Carotene


X


ϴ-carotene


X


Lutein


X


Lutein ester


X


Zeaxanthin


X


Vitamin D3


X


all-trans-Carophyll yellow


X


Retinyl beta-glucuronide


X


Retinoyl beta-glucuronide


X


b-Cryptoxanthin palmitate


X


all-trans-retinyl palmitate


X


Retinol palmitate


X


4-Oxo-9-cis-retinoyl-beta-glucuronide


X


Hoduloside I


X


Retinol acetate


X


4-Hydroxyretinoic acid glucuronide


X


Piceid (cis-)


X


Zeaxanthin dipalmitate


X


7,7′,8,8′-Tetrahydro-beta,beta-carotene


X


7′,8′-Dihydro-8′-hydroxycitraniaxanthin


X


(8′R)-Neochrome


X


Sintaxanthin


X


8′-Apo-b-caroten-8′-ol


X


Cryptoxanthin-alpha


X


Cryptocapsin


X


(6b,7b,13R)-6,7-Diacetoxy-8,14-labdadiene-


X


13-ol


all-trans-18-Hydroxyretinoic acid


X


Notoginsenoside N


X


(S)-1′,2′-Epoxy-1′,2′-dihydro-b,y-carotene


X


Deepoxyneoxanthin


X


Anhydroamarouciaxanthin B


X


Citranaxanthin


X


Parasiloxanthin


X


Notoginsenoside R1


X


(3S,5R,6R)-3,5-dihydroxy-6,7-didehydro-5,6-


X


dihydro-12′-apo-œ≤-caroten-12′-al


1-(2,6,6-Trimethyl-1-cyclohexen-1-yl)-1-


X


penten-3-one


Idoxanthin


X


12′-Apo-b-carotene-3,12′-diol


X


beta-Citraurol


X


(3b,5a,22E,24S)-Stigmasta-7,22,25-trien-3-ol


X


3-glucoside


Theasaponin B1


X


œ≤-carotene 15,15′ epoxide


X


Koryoginsenoside R1


X


(3′RS,3′SR)-Astaxanthin


X


(3S,3′S)-Astaxanthin


X


3-Hydroxysintaxanthin


X


(13Z)-Retinol


X


11-cis-Retinol


X


9-cis-Retinol


X


all-trans-retinol


X


Tragopogonsaponin K


X


Janthitrem G


X


4-Oxoretinol


X


(24E)-3alpha,15alpha-Diacetoxy-23-oxo-


X


7,9(11),24-lanostatrien-26-oic acid


5,6-Diepikarpoxanthin


X


5,6-Dihydro-b,b-carotene-3,3′,5,6-tetrol


X


Rubixanthin


X


(all-E)-Rubixanthin


X


Cinncassiol A


X


beta-Carotene 5,6-epoxide


X


beta-Carotene-5,6-monoepoxide


X


Prenigroxanthin


X


Notoginsenoside H


X


(3beta,5alpha,6beta,9alpha,22E,24R)-23-


X


Methylergosta-7,22-diene-3,5,6,9-tetrol


Pseudoginsenoside RT3


X


Notoginsenoside R9


X


Ganoderiol C


X


4-Oxo-retinoic acid


X


Cryptocapsone


X


beta-Micropteroxanthin


X


2,6-Cyclolycopene-1,5-diol


X


Phomopsin B


X


10′-Apo-beta-caroten-10′-al


X


2′-Apo-beta-carotenal


X


9-cis-Retinoic acid


X


9,13-cis-Retinoate


X


Pancreatin


X


Retinoic acid


X


13-cis-Retinoic acid


X


Unknown 370


X


Cryptoxanthin epoxide


X


Semi-beta-carotenone


X


torulene


X


Mytiloxanthin


X


Pithecelloside


X


Persicaxanthin


X


Neoxanthin


X


9′-cis-Neoxanthin


X


cis-Neoxanthin


X


Anhydrocinnzeylanine


X


Hydratopyrrhoxanthinol


X


Delphinidin 3-[6-(4-(Z)-p-


X


coumarylrhamnosyl)glucoside] 5-glucoside


4-Oxo-13-cis-retinoate


X


Ginsenoside A2


X


7,8-Dehydroastaxanthianthin


X


Tragopogonsaponin N


X


5-Dehydroavenasterol


X


Valenciaxanthin


X


(9E)-Valenciaxanthin


X


(9Z)-Valenciaxanthin


X


Notoginsenoside R2


X


Ginsenoside A1


X


4-Hydroxyretinoic acid


X


Floratheasaponin C


X


delta-Methylionone


X


beta-Carotinal


X


Philosamixanthin


X


Latoxanthin


X


Hydroxysintaxanthin 5,6-epoxide


X


Canthaxanthin


X


(3beta,5alpha,9alpha,22E,24R)-3,5,9-


X


Trihydroxy-23-methylergosta-7,22-dien-6-one


Reticulataxanthin


X


(S)-Neomammein


X


Notoginsenoside M


X


Notoginsenoside R6


X


Ginsenoside Rh5


X


15,15′-dihydroxy-œ≤-carotene


X


22-Angeloylbarringtogenol C


X


gamma-Eudesmol rhamnoside


X


Neochrome


X


11-cis-Retinaldehyde


X


Retinal


X


13-cis-Retinal


X


9-cis-Retinal


X


all-trans-retinal


X


Vinaginsenoside R2


X


Cryptochrome


X


3,5,9-Trihydroxyergost-7-en-6-one


X


œμ,œμ-carotene-3-diol


X


Eleutheroside L


X


Ginsenoside Rh8


X


3,6-Epoxide-5-hydroxy-5,6-dihydrozeaxanthin


X


Cucurbitaxanthin A


X


cis-Cryptoxanthin


X


beta-Cryptoxanthin


X


Assamsaponin D


X


Theasaponin


X


Cartormin


X


Colupdox a


X


Cobalamin


X


(3beta,5alpha,6beta,22E,24R)-23-


X


Methylergosta-7,22-diene-3,5,6-triol


Hoduloside VII


X


Ginsenoside Rf


X


Tuberoside


X


(3b,16a,21b,22a)-12-Oleanene-3,16,21,23,28-


X


pentol-22-angeloyloxy-23-al


23-Methylergosterol


X


Hoduloside X


X


Ganodermic acid TQ


X


Ginsenoside Rs1


X


5-Hydroxycapsanthin


X


Goshonoside F3


X


Chondrillasterol 3-[glucosyl-(1−>2)-glucosyl-


X


(1−>2)-glucoside]


Aleuriaxanthin


X


2-Hydroxyestrone-4-S-glutathione


X


Persicachrome


X


(3S,5R,8R)-Persicachrome


X


(3S,5R,8S)-Persicachrome


X


alpha-Spinasterol 3-glucoside


X


Trigoneoside XIIb


X


Ginsenoside III


X


(3R,3′R,6′R,9-cis)-Carotene-3,3′-diol


X


Calthaxanthin


X


(3R,3′R,6′R,13-cis)-beta,epsilon-Carotene-3,3′-


X


diol


(3R,3′R,6′R,9′-cis)-beta,epsilon-Carotene-3,3′-


X


diol


Mutachrome


X


22-Angeloyltheasapogenol A


X


Lutein 5,6-epoxide


X


Apo-14′-zeaxanthinal


X


Cryptoxanthin diepoxide


X


Xanthohumol E


X


4-Oxoretinal


X


Blumealactone A


X


Campesteryl alpha-linolenate


X


alpha-Spinasterol gentiobioside


X


Ginsenoside F1


X


Theasaponin A1


X


Salmoxanthin


X


Lansioside B


X


Apo-13-zeaxanthinone


X


Vinaginsenoside R4


X


1,25-Dihydroxy vitamin D3-26,23-lactone


X


Pisumoside A


X


Tragopogonsaponin G


X


4-Hydroxyretinal


X


Vinaginsenoside R15


X


(3beta,22E,24R)-5,8-Epidioxy-23-


X


methylergosta-6,22-dien-3-ol


8-Epidiosbulbin E acetate


X


5,6-Epoxy-5,6-dihydro-10′-apo-b,y-carotene-


X


3,10′-diol


Sinensiaxanthin


X


Vinaginsenoside R14


X


Chakasaponin II


X


Floratheasaponin B


X


Ginsenoside Rh6


X


Strigoi


X


Hoduloside IX


X


Notoginsenoside B


X


Ginsenoside F3


X


beta-Selinene


X


Ginsenoside Rh1


X


3-Hydroxy-10′-apo-b,y-carotenal


X


Apo-10′-zeaxanthinal


X


Ginsenoside F5


X


3,6:5′,8′-Diepoxy-5,5′,6,8′-tetrahydro-b,b-


X


carotene-3′,5-diol


Cucurbitachrome 1


X


(20S,24E)-20,26-Dihydroxy-24-dammaren-3-


X


one


(3S,5R,8S)-5,8-Epoxy-5,8-dihydro-beta,beta-


X


caroten-3-ol


7-Methylhypoxanthine


X


25-Azacholesterol


X


Flavochrome


X


Goyaglycoside e


X


delta-Carotene-1,2-epoxide


X


Amaranthussaponin I


X


6′-Hydroxylutein


X


Ginsenoside Rs2


X


Quinquenoside R1


X


Tragopogonsaponin H


X


Isofucosterol glucoside


X


Erinacine C


X


Schidigerasaponin C1


X


Crispane


X


TR-Saponin C


X


Cucurbitaxanthin B


X


5′,6′-Epoxy-5′,6′-dihydro-beta,beta-caroten-3-ol


X


Triphasiaxanthin


X


Valenciachrome


X


28-Glucosyloleanolic acid 3-[rhamnosyl-(1−>2)-


X


galactosyl-(1−>3)-glucuronide]


2′-Dehydroplectaniaxanthin


X


Lansioside C


X


Phyllanthusol A


X


Kaempferol 3-(2″-(E)-feruloylgalactosyl-


X


(1−>4)-glucoside)


Lablaboside F


X


Capsanthin 5,6-epoxide


X


Capsanthone


X


Madlongiside C


X


Crispanone


X


13-cis-Capsanthin


X


9-cis-Capsanthin


X


Capsanthin


X


Heteroxanthin


X


(3S,3′R,4xi)-beta,beta-Carotene-3,3′,4-triol


X


Erinacine B


X


Aurochrome


X


Cholestane-3,7,12,25-tetrol-3-glucuronide


X


3′-(1″-(3-Methylbutanoyl))-angeloyl vaginidiol


X


Capsorubin


X


Ganoderiol B


X


Phaseoloside A


X


baruol


X


Corbisterol


X


(+/−)-Ethyl 3-hydroxy-2-methylbutyrate


X


Protobassic acid


X


Ginsenoside Ia


X


Momordicasaponin I


X


Nigroxanthin


X


Anhydrocinnzeylanol


X


CE(DiMe(11,3))


X


CE(DiMe(9,3))


X


(3R,3′R,13-cis)-beta,beta-Carotene-3,3′-diol


X


(3R,3′R,15-cis)-beta,beta-Carotene-3,3′-diol


X


(3R,3′R,9-cis)-beta,beta-Carotene-3,3′-diol


X


5-dehydroepisterol


X


5-Dehydroepisterol


X


Chinenoside VI


X


Amasterol


X


Cryptoflavin


X


Theasaponin G1


X


Apo-12′-zeaxanthinal


X


beta-Citraurin


X


(3beta,4alpha,5alpha,24Z)-4-Methylstigmasta-


X


8(14),24(28)-dien-3-ol


Camelliasaponin B1


X


Sitoindoside II


X


Sitosterol 3-O-(6′-O-oleyl-beta-D-glucoside)


X


Physapruin B


X


Retinyl ester


X


Quinquenoside L1


X


(3beta,5alpha,6beta,24R)-Stigmastane-3,5,6-


X


triol


(6S,6′S)-epsilon,epsilon-Carotene-3,3′-dione


X


epsilon,epsilon-Carotene-3,3′-dione


X


Ganoderic acid X


X


Lablaboside A


X


Retinoyl CoA


X


(3b,24R,25x)-26-Benzoyloxystigmast-5-ene-3-


X


ol


Melilotussaponin O1


X


Hovenolactone


X


Quinquenoside II


X


Momordicoside L


X


Pseudoginsenoside Rc1


X


Quinquenoside III


X


all-trans-5,6-Epoxyretinoic acid


X


Isoachifolidiene


X


28-Glucosylarjunolate 3-[rhamnosyl-(1−>3)-


X


glucuronide]


Euglobal III


X


28-Galloylglucosylpomolate 3-arabinoside


X


5,7-Megastigmadien-9-ol glucoside


X


Araliasaponin I


X


Sanguisorbin E


X


(S)-Abscisic acid


X


Abscisic acid


X


Apo-15-zeaxanthinal


X


5,8-Epoxy-5,8-dihydro-10′-apo-b,y-carotene-


X


3,10′-diol


Occidentoside


X


Ganoderic acid Me


X


Araliasaponin III


X


3-cis-Hydroxy-beta,epsilon-Caroten-3′-one


X


beta-Citraurinene


X


Ginsenoside Rh7


X


Ganoderic acid Mf


X


Physalolactone B 3-glucoside


X


1′-O-Acetylpaxilline


X


13-Hydroxy-5′-O-methylmelledonal


X


7,8-Dehydro-beta-micropteroxanthin


X


2,4,7,10,13,16,19-Docosaheptaenoyl-CoA


X


1-hydroxylycopene


X


Chrysanthemaxanthin


X


Flavoxanthin


X


Colupox a


X


Clerosterol 3-glucoside


X


CE(MonoMe(11,3))


X


Ganoderic acid delta


X


Ursololactone


X


(R)-3,4-Dihydro-2-methyl-2-(4,8,12-trimethyl-


X


3,7,11-tridecatrienyl)-2H-1-benzopyran-6-ol


Vitamin D2 3-glucuronide


X


beta-Ionone


X


Lansioside A


X


beta-Sitosterol 3-O-beta-D-galactopyranoside


X


Coriandrinol


X


Schidigerasaponin C2


X


7-O-Acetylhorminone


X


beta-Doradecin


X


Apo-10′-violaxanthal


X


Quinquenoside V


X


Eleutheroside M


X


Cyanidin 3-(6″-p-coumarylsambubioside)


X


2,3,23-Triacetylsericic acid


X


Pyrrhoxanthinol


X


24-Hydroxyglabrolide


X


Ginsenoside Ra1


X


Cinncassiol D1 glucoside


X


Sterebin G


X


Momordicoside I


X


(+)-cis-abscisic aldehyde


X


Isoalantolactone


X


Vomifoliol 9-[xylosyl-(1−>6)-glucoside]


X


CE(DiMe(11,5))


X


CE(DiMe(13,5))


X


CE(DiMe(9,5))


X


Quillaic acid 3-[xylosyl-(1−>3)-[galactosyl-


X


(1−>2)]-glucuronide]


Dulcin


X


5,6-Epoxy-5,6-dihydrolycopene


X


(13Z)-Violaxanthin


X


(all-E)-Violaxanthin


X


9-cis-violaxanthin


X


cis-Violaxanthin


X


Violaxanthin


X


Theasaponin F3


X


7-dehydrodesmosterol


X


7,8-Didehydrodesmosterol


X


(3b,12a,13b)-3-Hydroxy-12-oxo-28,13-


X


oleanolide 3-acetate


Cryptoxanthin 5,6:5′,8′-diepoxide


X


Quinquenoside I


X


Momordicoside K


X


alpha-Cryptoxanthin


X


zeinoxanthin


X


Momordicoside G


X


(3S,3′R,5R,6R)-7′,8′-Didehydro-3,6-epoxy-5,6-


X


dihydro-beta,beta-carotene-3′,5-diol


25-Cinnamoylvulgaroside


X


24-methylenecycloartanyl ferulate


X


Arctiol


X


Tarasaponin I


X


Ganodermic acid Jb


X


Lucyoside M


X


Mangiferadiol


X


Ganoderenic acid E


X


Majonoside R2


X


45-Hydroxyhomoyessotoxin


X


Edulisin IV


X


3′-Geranyl-2′,4,4′,6′-tetrahydroxychalcone


X


5,9-Epidioxy-3-hydroxyergost-7-en-6-one


X


Methyl phaeophorbide b


X


(5R,5′R,6S,8′R)-Luteochrome


X


Medicoside L


X


Blumealactone C


X


6-O-p-Coumaroyl-1,2-digalloylglucose


X


Zingiberol


X


(3beta,5alpha)-Ergosta-7,23-dien-3-ol


X


4-Hydroxyestrone-2-S-glutathione


X


Dehydroxypaxilline


X


Araliasaponin VIII


X


Epoxyganoderiol B


X


Vinaginsenoside R3


X


Silenoside A


X


beta-Sitosterol acetate


X


2-Hydroxyestrone-1-S-glutathione


X


4-(2,6,6-Trimethyl-1-cyclohexen-1-yl)-2-


X


butanone


Methuyl tanshinonate


X


Balanitesin


X


24-Methylene-cycloartanol palmitate


X


Vinaginsenoside R5


X


11-Hydroxyprogesterone 11-glucuronide


X


Tarasaponin II


X


PC-M5′


X


Protopanaxadiol 3-glucoside 20-[arabinosyl-


X


(1−>2)-glucoside]


Calendulaglycoside B


X


(1(10)E,4a,5E)-1(10),5-Germacradiene-12-


X


acetoxy-4,11-diol


Melilotussaponin O2


X


Melledonal C


X


29-Methylisofucosterol


X


Delphinidin 3-(6-p-coumaroylglucoside)


X


Melleolide K


X


Acutoside F


X


Ganolucidic acid D


X


3-trans,5-cis-Octadienoyl-CoA


X


Notoginsenoside G


X


Sasanquol


X


(5R,5′R,6S,6′S)-5,6:5′,6′-Diepoxy-5,5′,6,d′-


X


tetrahydro-beta,beta-carotene


Acutoside G


X


Austalide I


X


beta-Amyrin palmitate


X


7′,8′-Dihydro-8′-hydroxyreticulataxanthin


X


Vinaginsenoside R17


X


Celosianin


X


Notoginsenoside I


X


Ganoderic acid VI


X


(3S,5R,6S)-5,6-epoxy-3-hydroxy-5,6-dihydro-


X


12′-apo-œ≤-caroten-12′-al


(3S,5R,6S)-5,6-Epoxy-5,6-dihydro-3-hydroxy-


X


8′-apo-beta,psi-carotenal


Apo-12′-violaxanthal


X


beta-Citraurin epoxide


X


Tsugaric acid A 21-glucosyl ester


X


Campesterol glucoside


X


Melilotoside D


X


Vinaginsenoside R6


X


Vinaginsenoside R16


X


Vinaginsenoside R7


X


caffeoyl hexose


X


Campesteryl linoleate


X


Pelargonidin 3-p-coumarylglucoside


X


Elatoside I


X


Campesteryl stearate


X


methoxyhedyotisol A


X


Majonoside R1


X


(4E,9a)-9-(3-Methyl-2E-pentenoyloxy)-


X


4,10(14)-oplopadien-3-one


(4Z,9a)-9-(3-Methyl-2E-pentenoyloxy)-


X


4,10(14)-oplopadien-3-one


Quinquenoside IV


X


(3beta,4alpha,5alpha,23E)-4-Methylergosta-


X


7,23-dien-3-ol


trans-24-Methyl-23-dehydrolophenol


X


Alliosterol 1-rhamnoside 16-galactoside


X


CE(MonoMe(11,5))


X


CE(MonoMe(13,5))


X


CE(MonoMe(9,5))


X


Ganolucidic acid A


X


Lablaboside E


X


Calcitroic acid


X


Cyclocalopin F


X


Acutoside D


X


(3a,22x,24E)-3,22-Dihydroxycycloart-24-en-


X


26-oic acid


Saringosterol 3-glucoside


X


Ganoderic acid epsilon


X


Basilimoside


X


Withaperuvin D


X


Acutoside B


X


trans-2-all-cis-6,9,12,15,18,21-


X


Tetracosaheptaenoyl-CoA


8-Hydroxyhyperforin 8,1-hemiacetal


X


Medicoside H


X


(3alpha,5beta)-24-Oxo-24-[(2-


X


sulfoethyl)amino]cholan-3-yl-beta-D-


glucopyranosiduronic acid


all-trans-13,14-Dihydroretinol


X


(3beta,4alpha,5alpha,24(28)E)-4,14-


X


Dimethylstigmasta-8,24(28)-dien-3-ol


3-O-p-trans-Coumaroylalphitolic acid


X


(3beta,5alpha,24R)-Ergost-8(14)-en-3-ol


X


Tarasaponin IV


X


Phytolaccoside I


X


Tyromycic acid


X


Ganoderic acid DM


X


Kudzusaponin SA3


X


7,13-Eperudien-15-oic acid


X


(3beta,5alpha,24R)-Stigmast-8(14)-en-3-ol


X


Medicagenic acid 3-O-b-D-glucoside 28-O-[b-


X


D-xylosyl-(1−>4)-a-L-rhamnosyl-(1−>2)-a-L-


arabinosyl] ester


24-Ethyl-24(25)-dehydrolathosterol


X


7,24-Stigmastadien-3beta-ol


X


Peposterol


X


Armillyl everninate


X


Capsochrome


X


Delphinidin 3-(3″-p-coumaroylglucoside)


X


3-Hydroxystigmast-5-en-7-one


X


CE(18:3(9Z,12Z,15Z))


X


CE(20:4(8Z,11Z,14Z,17Z))


X


CE(22:5(7Z,10Z,13Z,16Z,19Z))


X


Ganolucidic acid C


X


3-Hexadecanoyloleanolic acid


X


6-Hydroxysandoricin


X


Theasaponin H1


X


Tulipinolide


X


Calenduloside H methyl ester


X


5-methyl-2,4-bis(3-methyl-2-butenyl)-6-(2-


X


methyl-1-oxopropyl)-5-(4-methyl-3-


pentenyl)cyclohexanone


26-Glucosyl-1,3,11,22-tetrahydroxyergosta-


X


5,24-dien-26-oate


Ginsenoside Ra3


X


Ampeloside Bf2


X


Medicagenic acid 3-O-[b-D-glucosyl-(1−>2)-b-


X


D-glucoside] 28-O-[b-D-xylosyl-(1−>4)-a-L-


rhamnosyl-(1−>2)-a-L-arabinosyl] ester


Ganolucidic acid B


X


Withanolide B


X


(3beta,4alpha,22beta)-22-[[2-O-(6-Deoxy-


X


alpha-L-mannopyranosyl)-alpha-L-


arabinopyranosyl]oxy]-23-hydroxyolean-12-


en-3-yl O-6-deoxy-alpha-L-mannopyranosyl-


(1−>2)-O-beta-D-galactopyranosyl-(1−>2)-beta-


D-glucopyranosiduronic acid


Ginsenoside Ra2


X


7-Dehydrocholesterol


X


Lumisterol 3


X


29-Norcycloartane-3,24-dione


X


23-trans-p-Coumaroyloxytormentic acid


X


Kaempferol 3-(2-feruloylsophoroside) 7-


X


glucoside


Ampeloside Bf1


X


Gibberellin A111


X


Soybean saponin BG


X


Taccagenin


X


Lepidiumterpenoid


X


2-trans,4-trans-Octadienoyl-CoA


X


Peltatol A


X


Ginsenoside Rb3


X


ent-kaur-16-en-19-oate


X


Cyclomammeisin


X


Ginkgolide J


X


28-Homobrassinolide


X


Furomammea D


X


ϔ24-25-sitosterol


X


Betavulgaroside II


X


Glucoconvallasaponin B


X


Quillaic acid 3-[galactosyl-(1−>2)-glucuronide]


X


Notoginsenoside Fc


X


Spinosin A


X


Avenestergenin A2


X


Spinasaponin A


X


Antheraxanthin


X


cis-Antheraxanthin


X


Amaranthussaponin III


X


Butyl 3-O-caffeoylquinate


X


Withangulatin A


X


Nuatigenin


X


Notoginsenoside Fa


X


Notoginsenoside L


X


Koryoginsenoside Rg2


X


beta-Carotenone


X


Chikusetsusaponin VI


X


Notoginsenoside R4


X


Arnamiol


X


Ginsenoside Rd


X


Glycyrrhizin


X


2-Deoxybrassinolide


X


7-oxateasterone


X


7-oxatyphasterol


X


Demissidine


X


Araloside A


X


Tuberoside A (Ullucus tuberosus)


X


beta-Rotunol


X


Oxysolavetivone


X


Glucosyl passiflorate


X


Cycloheterophyllin


X


Notoginsenoside D


X


Araliasaponin II


X


Ganoderal A


X


Cajanone


X


Schleicherastatin 5


X


Cafestol


X


Pitheduloside K


X


Cryptotanshinone


X


Physapubescin


X


3,4-Didehydroretinol


X


Ginsenoside Rg5


X


(3beta,5alpha,6beta,22E,24R)-Ergosta-


X


7,9(11),22-triene-3,5,6-triol


Majoroside F3


X


Tsugarioside B


X


Araloside G


X


Ganoderic acid Ma


X


Vitexin 7-glucoside 2″-p-coumarate


X


Ganolucidic acid E


X


(22±)-hydroxy-isofucosterol


X


Notoginsenoside E


X


Ganoderenic acid A


X


(3beta,24(241)Z)-24-Ethylidenelanost-9(11)-


X


en-3-ol


Ananasic acid


X


Glyciram


X


stigmasterol 3-O-ϲ-D-glucoside


X


Stigmasteryl glucoside


X


2-O-(4-Hydroxycinnamoyl)-1-O-galloyl-beta-


X


D-glucopyranoside


Ustiloxin A


X


3-Hydroxy-beta-ionol 3-[glucosyl-(1−>6)-


X


glucoside]


(3b,7b,22x)-Cucurbita-5,24-diene-3,7,23-triol


X


7-glucoside


2-O-(4-Hydroxycinnamoyl)-1,6-di-O-galloyl-


X


beta-D-glucopyranose


2-O-p-Coumaroyl-1,6-digalloyl-beta-D-


X


glucopyranoside


(2xi,3xi)-2,3-Dihydroxy-12,18-ursadien-28-oic


X


acid diacetate


Alliospiroside A


X


Auroxanthin


X


Epoxyganoderiol A


X


Tragopogonsaponin B


X


Araloside B


X


Calendulaglycoside A


X


Stipuleanoside R2


X


Pitheduloside E


X


(24R)-14alpha-Methyl-24-ethyl-5alpha-


X


cholest-9(11)-en-3beta-ol


Ganoderic acid beta


X


2-Cinnamoyl-1-galloyl-beta-D-glucopyranose


X


5,8-Epoxy-5,8-dihydro-3-hydroxy-8′-apo-b,y-


X


carotenal


2-Prenyl-6alpha-hydroxyphaseollidin


X


2,10-Di-(gamma,gamma-dimethylallyl)-


X


glycinol


Jurubine


X


28-Hydroxywithanolide E


X


4alpha-Methyl-24-ethyl-cholesta-7,24-dien-


X


3beta-ol


Gibberellin A97


X


Coffeasterene


X


Isocyperol


X


Mollicellin A


X


3-(2-Methylpropanoyloxy)-8-(3-


X


methylbutanoyloxy)-9,10-epoxy-p-mentha-


1,3,5-triene


17-alpha-Estradiol-3-glucuronide


X


17b-Estradiol 3-glucuronide


X


Estradiol-17alpha 3-D-glucuronoside


X


Pitheduloside A


X


2-Cinnamoyl-1,6-digalloyl-beta-D-


X


glucopyranose


Ganoderic acid W


X


(3beta,4alpha,5alpha)-4-Methylergosta-


X


8,14,24(28)-trien-3-ol


4ϱ-methyl-5ϱ-ergosta-8,14,24(28)-trien-


X


3ϲ-ol


Epoxyganoderiol C


X


8,5′-diferulic acid


X


Campesteryl brassidate


X


Campesteryl elaidate


X


Amaranthussaponin IV


X


Melilotoside C


X


Capsorubinidione


X


1-(2,6,6-Trimethyl-2-cyclohexen-1-yl)-1-


X


penten-3-one


Physagulin B


X


Monoglucuronylglycyrrhetinic acid


X


Calenduloside A


X


Lablaboside C


X


24-Methylenepollinasterol


X


(E)-3-oxo-beta-ionone


X


Perulactone


X


Zerumbodienone


X


Antibiotic CJ 14258


X


Isoscoparin 2″-(6-(E)-p-coumaroylglucoside)


X


2,3-Dehydrosalvipisone


X


cis-2-Methyl-5-isopropylhexa-2,5-dienoyl-


X


CoA


trans-2-Methyl-5-isopropylhexa-2,5-dienoyl-


X


CoA


Ginsenoside RAO


X


Ginsenoside Rb1


X


Ginsenoside Rc


X


Portensterol


X


4′-Dihydroabscisic acid


X


Capsicoside A1


X


6-oxocampestanol


X


Cholesterol glucuronide


X


Momordicoside D


X


Janthitrem C


X


(3beta,5alpha,6alpha,9alpha,22E,24R)-Ergosta-


X


7,22-diene-3,5,6,9-tetrol


Elatoside G


X


Spinosin C


X


Apo-11-zeaxanthinal


X


Psidinin C


X


Goyaglycoside c


X


precorrin-2


X


2,9-Bis(3-methyl-2E-pentenoyl)-2b,9a-


X


dihydroxy-4Z,10(14)-oplopadien-3-one


Umbellifolide


X


Kaempferol 3-sophoroside 7-(2-


X


feruloylglucoside)


Momordin IIa


X


2,6,6-Trimethyl-1-cyclohexen-1-acetaldehyde


X


TR-Saponin B


X


Elatoside D


X


4-Hydroxyestradiol


X


Trigoneoside XIb


X


Phytocassane D


X


Cyclolaudenyl palmitate


X


Triterpenoid


X


Calenduloside B


X


(4Z,9a)-9-Angeloyloxy-4,10(14)-oplopadien-3-


X


one


Glyceollidin II


X


(3b,5a,22E,24S)-Stigmasta-7,22,25-trien-3-ol


X


25(27)-Dehydrochondrillasterol


X


5alpha-Stigmasta-7,22,25-trien-3beta-ol


X


Bassic acid


X


28-[Rhamnosyl-(1−>2)-[b-D-glucosyl-(1−>3)]-


X


[2R-methylbutanoyl-(−>4)]-fucosyl]-30-


methyl-3b,23-dihydroxy-12-oleanene-28,30-


dioate 3-[galactosyl-(1−>2)-glucuronide]


Blumealactone B


X


Camellioside D


X


alpha-Selinene


X


Physagulin D


X


PRE


X


Cyclocommunol


X


Spinacetin 3-[E-feruloyl-(−>2)-xylosyl-(1−>2)-


X


[glucosyl-(1−>3)]-glucoside]


(24Z)-Ethylidenelathosterol


X


(3b,5a,24(28)E)-Stigmasta-7,24(28)-dien-3-ol


X


avenasterol


X


delta7-Avenasterol


X


Stigmasta-7,24(28)-dien-3-ol


X


Phyllanthusol B


X


D5-Avenasterol


X


delta-5-Avenasterol


X


Fucosterol


X


Isofucosterol


X


Alloalantolactone


X


2-alpha-Ethoxydihydrophytuberin


X


2-beta-Ethoxydihydrophytuberin


X


3b,5a,6b-Cholestanetriol


X


Cucurbitacin A


X


Physagulin A


X


Hoduloside VI


X


Ganoderic acid Y


X


Ganoderic acid C2


X


3,11,12-Trihydroxy-1(10)-spirovetiven-2-one


X


Cyclopassifloside VI


X


7-Oxostigmasterol


X


Sonchifolin


X


Estriol-16-glucuronide


X


(3alpha,22R,23R,24S)-3,22,23-


X


Trihydroxystigmastan-6-one


2-Deoxy-28-homocastasterone


X


Cucurbitacin I 2-glucoside


X


Galanolactone


X


CE(22:6(4Z,7Z,10Z,13Z,16Z,19Z))


X


Masticadienediol


X


Araloside C


X


Gynosaponin I


X


Ganoderal B


X


Clupanodonyl CoA


X


ϱ-linolenoyl-CoA


X


8-Epiisoivangustin


X


Schleicherastatin 6


X


isobutanoyl-CoA


X


(15a,20R)-Dihydroxypregn-4-en-3-one 20-


X


[glucosyl-(1−>4)-6-acetyl-glucoside]


Matesaponin 3


X


Cyclopassifloside IV


X


3-Methyl-4-(2,6,6-trimethyl-2-cyclohexen-1-


X


yl)-3-buten-2-one


Medicagenic acid 3-O-[a-D-glucosyl-(1−>4)-b-


X


D-glucoside]


Petasitin


X


Glycochenodeoxycholic acid 3-glucuronide


X


25-Ethoxy-24-methoxycylcoartanol


X


Elatoside E


X


trans-Caffeic acid [apiosyl-(1−>6)-glucosyl]


X


ester


Arctiopicrin


X


Azukisaponin I


X


(8R)-Mutatoxanthin


X


(8S)-Mutatoxanthin


X


Mutatoxanthin


X


Cinncassiol D2


X


(5alpha,6beta,14alpha,20R,22R)-5,6,14,20,27-


X


Pentahydroxy-1-oxowith-24-enolide


Macrophorin D


X


Gibberellin Ga115


X


(3beta,4alpha,5alpha,24Z)-4-Methylstigmasta-


X


8,24(28)-dien-3-ol


Junicedral


X


Oleanolic acid 3-[glucosyl-(1−>4)-xyloside]


X


Azuki saponin VI


X


10,11-Dihydro-6-oxoatlantone


X


22,23-Dihydroergosterol


X


3beta-3-Hydroxy-18-lupen-21-one


X


Goshonoside F2


X


22-Dehydroclerosterol


X


25(27)-Dehydroporiferasterol


X


(3b,21b)-12-Oleanene-3,21,28-triol 28-


X


[arabinosyl-(1−>3)-arabinosyl-(1−>3)-


arabinoside]


3beta-O-beta-D-Glucopyranosiduronic acid


X


(1_2)-beta-D-glucopyranosyloxy]-machaerinic


acid_-lactone


Ginsenoside F2


X


Cyclocalamin


X


trans-2-Enoyl-OPC8-CoA


X


Lablaboside D


X


Helinorbisabone


X


11-Ketoetiocholanolone


X


Isoscoparin 2″-(6-(E)-feruloylglucoside) 4′-


X


glucoside


(3b,5b,22a,25R)-Furostane-22-methoxy-3,26-


X


diol 3-[glucosyl-(1−>2)-glucoside] 26-


glucoside


Fasciculol F


X


Janthitrem B


X


Ponasteroside A


X


1-(2,6,6-Trimethyl-2-cyclohexen-1-yl)-1,6-


X


heptadien-3-one


Cinncassiol D1


X


Vinaginsenoside R8


X


Taraxasterol palmitate


X


thalianol


X


Methyl (7Z,9Z,9′Z)-6′-apo-y-caroten-6′-oate


X


Methyl 6′-apo-9′Z-lycopen-6′-oate


X


Methyl apo-6′-lycopenate


X


Salviol


X


Goyaglycoside g


X


Armillaripin


X


Isocyclocalamin


X


28-Glucosyloleanolic acid 3-[arabinosyl-


X


(1−>2)-6-methylglucuronide]


2,4-Methylene cholesterol


X


Campesteryl p-coumarate


X


Ganoderiol F


X


OPC6-CoA


X


OPC8-CoA


X


Cynarasaponin C


X


2a-Hydroxygypsogenin 3-O-b-D-glucoside


X


Gibberellin A38 glucosyl ester


X


Tiglyl-CoA


X


(3beta,4alpha,5alpha,24S)-4,14-


X


Dimethylstigmasta-8,25-dien-3-ol


24epsilon-Ethyl-31-norlanosta-8,25(27)-dien-


X


3beta-ol


(E)-2-methylcrotonoyl-CoA


X


Euglobal IVa


X


Rosmaridiphenol


X


Ovalicin


X


gibberellin A8-catabolite


X


Panaxoside B


X


Calenduloside H


X


Stigmastanediol


X


abieta-7,13-dien-18-ol


X


Kuguacin C


X


Acetylbalchanolide


X


Lucidal


X


(3alpha,5beta,7alpha,12alpha)-24-


X


[(Carboxymethyl)amino]-1,12-dihydroxy-24-


oxocholan-3-yl-beta-D-glucopyranosiduronic


acid


Eucalyptone


X


(24E)-3ϱ,7ϱ,12ϱ-trihydroxy-


X


5ϲ-cholest-24-enoyl-CoA


3,5-Dihydroxy-6,7-megastigmadien-9-one


X


4ϱ-methyl-5ϱ-cholesta-8,14,24-trien-3ϲ-ol


X


Punicacortein B


X


Glabrolide


X


Carthamoside A1


X


Isocolumbin


X


Retrocalamin


X


Sandosaponin B


X


Ginsenoside Rh3


X


beta-Ionyl acetate


X


Deterrol stearate


X


Epiisolubimin


X


Hericenone F


X


Hericenone G


X


Apiumoside


X


gibberellin A15 (open lactone form)


X


Notoginsenoside A


X


Bauerenyl acetate


X


alpha-Amyrin cerotate


X


alpha-Amyrin palmitate


X


alpha-Amyrin tetratriacontanoate


X


Ganoderiol D


X


Macrophorin A


X


Quercilicoside A


X


Rishitinone


X


5-Dehydro-avenasterol


X


Cycloeucalanone


X


Ganoderenic acid D


X


Lucyoside K


X


6beta-Hydroxy-3-oxo-12-oleanen-28-oic acid


X


3-O-{beta-D-Glucopyranosyl(1_2)-[alpha-L-


X


arabinopyranosyl(1_6)]-beta-D-


glucopyranosyl}-oleanolic acid


(3b,4b,5a,24Z)-4-Methylstigmasta-7,24(28)-


X


dien-3-ol


24-ethylidenelophenol


X


alpha 1-Sitosterol


X


Epicitrostadienol


X


Armillarinin


X


Tarasaponin VI


X


Cyanidin 3-(disinapoylsophoroside) 5-


X


glucoside


Pelargonidin 3-O-[b-D-Xylopyranosyl-(1−>2)-


X


a-D-glucopyranoside]


Shinflavanone


X


3-O-≤-D-glucosyl-brassicasterol


X


Basellasaponin D


X


Chakasaponin VI


X


28-[Glucosyl-(1−>6)-glucosyl]oleanolic acid 3-


X


[glucosyl-(1−>3)-[arabinosyl-(1−>4)]-


glucuronide]


4ϱ-carboxy-stigmasta-7,24(241)-dien-3ϲ-ol


X


Momordin Ia


X


(3beta,8x,9x,15x,24x)-8,9-Epoxyergosta-5,22-


X


diene-3,15-diol


Eriojaposide A


X


Gynosaponin S


X


Notoginsenoside Fe


X


Notoginsenoside K


X


Bisabolene oxide


X


beta-Sitosterol behenate


X


beta-Sitosterol palmitate


X


caffeoyl pentose


X


Ganodermanontriol


X


23-Dehydrocholesterol


X


Periandrin III


X


(20R)-Ginsenoside Rg3


X


Avenestergenin A1


X


Hemsloside Ma 2


X


Spinasaponin B


X


Erythrodiol 3-decanoate


X


Erythrodiol 3-palmitate


X


10-Epijunenol


X


Yucalexin P17


X


cholesteryl-ϲ-D-glucoside


X


3-Hydroxy-beta-ionone


X


Fasciculic acid C


X


Congmunoside X


X


TR-Saponin A


X


(24E)-3ϱ,7ϱ-dihydroxy-5ϲ-cholest-24-


X


enoyl-CoA


Ginsenoyne M


X


Fasciculic acid A


X


Cyclopassifloside X


X


Camellenodiol


X


20(29)-Lupene-2a,3b,28-triol


X


Lucidenic acid F


X


Furohyperforin


X


jasmonoyl-CoA


X


Melleolide B


X


Bn-NCC-1


X


Camelliagenin B


X


Alkaloid AQC2


X


(3b,20R,22R)-3,20,27-Trihydroxy-1-oxowitha-


X


5,24-dienolide 3-glucoside


FADH2


X


4-Deoxyhumulone


X


Isovitexin 2″-(6″′-feruloylglucoside)


X


Physalolactone B


X


Portuloside A


X


Trilobinol


X


campestanyl-16:0


X


Gibberellin A17


X


Kuguacin A


X


Pitheduloside F


X


3-trans-p-Coumaroylrotundic acid


X


Calotropenol acetate


X


Isoscoparin 2″-(6-(E)-feruloylglucoside)


X


Hoduloside VIII


X


Momordin Ie


X


3b,6a-Dihydroxy-alpha-ionol 9-[apiosyl-


X


(1−>6)-glucoside]


Hericenone H


X


Arvensoside D


X


cis-5,6-Dihydro-5,6-dihydroxy-carotene


X


Celereoside


X


Tricyclodehydroisohumulone


X


2alpha-Hydroxypyracrenic acid


X


Kuwanol D


X


4ϱ-carboxy-ergosta-7,24(241)-dien-3ϲ-ol


X


Spinosin B


X


Cymbopogonol


X


Araliasaponin V


X


Medicagenic acid 3-O-b-D-glucuronide 28-0-


X


[b-D-xylosyl-(1−>4)-a-L-rhamnosyl-(1−>2)-a-


L-arabinosyl] ester


Isovitexin 7-(6″′-sinapoylglucoside)


X


Ginsenoside M7cd


X


(3Z)-phytochromobilin


X


2beta-Hydroxytestosterone


X


Oxyglutinosone


X


(3b,6b,8b,12a)-8,12-Epoxy-7(11)-


X


eremophilene-6-angeloyloxy-8,12-dimethoxy-


3-ol


CE(20:5(5Z,8Z,11Z,14Z,17Z)


X


Torvonin A


X


Erythrabyssin II


X


Ginsenoside Ro


X


3-oxo-23,24-bisnorchol-4-en-22-oyl-CoA


X


Cyclopassifloside V


X


4-(2,6,6-Trimethyl-1-cyclohexenyl)-2-butanol


X


Pitheduloside B


X


3-oxo-2-(cis-2′-pentenyl)-cyclopentane-1-


X


octanoate


Cyclomorusin


X


LPS core


X


Dulcisisoflavone


X


Polyporusterone B


X


Estradiol acetate glucuronide


X


Betavulgaroside I


X


(S)-Nerolidol 3-O-[a-L-Rhamnopyranosyl-


X


(1−>4)-a-L-rhamnopyranosyl-(1−>2)-[4-(4-


hydroxy-3-methoxycinnamoyl)-(E)-a-L-


rhamnopyranosyl-(1−>6)]-b-D-


glucopyranoside]


Ergosterol


X


Obtusifoliol


X


Assafoetidin


X


Cynarasaponin F


X


Ganoderenic acid C


X


5,11-Selinadiene


X


Alectrol


X


Kuwanol E


X


(2E)-Dodecenoyl-CoA


X


(2E)-Hexadecenoyl-CoA


X


Camelliagenin A


X


3,5-Di-O-caffeoylquinic acid


X


3,5-Dicaffeoylquinic acid


X


Dicaffeoylquinic acid


X


3(S)-3-Hydroxy dodecen-(5Z)-oyl-CoA


X


3,4,4′-Trihydroxy-5,5′-diisopropyl-2,2′-


X


dimethylbiphenyl


23-Hydroxy-3-oxocycloart-24-en-26-oic acid


X


Nootkatol


X


5-Megastigmen-7-yne-3,9-diol 9-glucoside


X


Acutoside A


X


Heteroside


X


Lucyoside L


X


Cyclodehydroisolubimin


X


5,7-Dihydroxy-3′,4′-dimethoxy-8-(3-hydroxy-


X


3-methylbutyl)-isoflavone 7-glucoside


28-Glucuronyloleanolic acid 3-[galactosyl-


X


(1−>3)-glucoside]


24-Methylene-31-nor-5alpha-cycloartan-3-one


X


Cycloeucalenone


X


Sudachiin C


X


24-Methylene-24-dihydroparkeol


X


3beta-24-Methylenelanost-9(11)-en-3-ol


X


Alloxanthin


X


Asparagoside B


X


Isomasticadienonic acid


X


Piceid 4′-sulfate


X


Methylprotodioscin


X


Estriol 3-sulfate 16-glucuronide


X


Cloversaponin I


X


Myricadiol


X


Hebevinoside XIV


X


Cucurbitacin C


X


Cholesteryl acetate


X


campest-4-en-3ϲ-ol


X


Withaperuvin E


X


cathasterone


X


Mollicellin B


X


Lucyoside O


X


Glyuranolide


X


Sclareol


X


24-Methylcholesterol


X


Campesterol


X


Dihydrobrassicasterol


X


beta-Santalene


X


Garcilivin A


X


Quinoside D


X


(3beta,4alpha,5alpha,24(28)Z)-4,14-


X


Dimethylstigmasta-9(11),24(28)-dien-3-ol


Taraxerol


X


1alpha-Hydroxyarbusculin A


X


1-18:1-2-16:3-monogalactosyldiacylglycerol


X


1-18:2-2-16:3-monogalactosyldiacylglycerol


X


1-18:3-2-16:0-monogalactosyldiacylglycerol


X


1-18:3-2-16:1-monogalactosyldiacylglycerol


X


1-18:3-2-16:2-monogalactosyldiacylglycerol


X


1-18:3-2-18:2-monogalactosyldiacylglycerol


X


2′,7-Dihydroxy-4′-methoxy-8-prenylflavan


X


2′,7-diglucoside


Kuguacin E


X


1-(2,4-Dihydroxyphenyl)-3-[8-hydroxy-2-


X


methyl-2-(4-methyl-3-pentenyl)-2H-1-


benzopyran-5-yl]-1-propanone


25-Acetyl-6,7-didehydrofevicordin F 3-


X


glucoside


Peltatol B


X


YGM 5A


X


Fasciculic acid B


X


Phytolaccoside F


X


Petunidin 3-O-[6-O-(4-O-(4-O-(beta-D-


X


glucopyranosyl)-feruloyl)-alpha-L-


rhamnopyranosyl)-beta-D-glucopyranoside]-5-


O-[beta-D-glucopyranoside]


Methyl dihydrophaseate


X


1,10-Epoxy-3,8-dihydroxy-4,11(13)-


X


germacradien-12,6-olide


Artonol B


X


4ϱ-hydroxymethyl-stigmasta-7,24(241)-


X


dien-3ϲ-ol


(3beta,5alpha,9alpha,14alpha,22E,24R)-


X


3,5,9,14-Tetrahydroxyergosta-7,22-dien-6-one


Gibberellin A37 glucosyl ester


X


Hebevinoside V


X


(24S)-14alpha,24-Dimethyl-9beta,19-cyclo-


X


5alpha-cholest-25en-3beta-ol


(3beta,5alpha)-14-Methyl-9,19-cycloergost-25-


X


en-3-ol


(3beta,4alpha,5alpha)-4-Methyl-8(14)-


X


cholesten-3-ol


4alpha-Methyl-5alpha-cholest-8(14)-en-3beta-


X


ol


(3beta,5alpha,6a)-Cholesta-8,14-diene-3,6-diol


X


Ursolic acid lactone


X


Anhydrosafflor Yellow B


X


Gibberellin A21


X


psi-Taraxasteryl acetate


X


3b-Hydroxy-6b-angeloyloxy-7(11)-


X


eremophilen-12,8b-olide


3b-Hydroxy-6b-tigloyloxy-7(11)-eremophilen-


X


12,8b-olide


15beta,19-ent-Dihydroxy-7-trachylobanone


X


Isogomphrenin II


X


ϔ9-tetrahydrocannabinolate


X


dammarenediol II


X


Furost-5-ene-3beta,22,26-triol


X


6-O-Methylarmillaridin


X


Mollicellin H


X


5′-O-Methylmelledonal


X


28-Hydroxy-14-taraxeren-3-one


X


Basellasaponin B


X


14-Hydroxy-7-methylrosmanol


X


1-18:1-2-16:2-monogalactosyldiacylglycerol


X


1-18:2-2-16:0-monogalactosyldiacylglycerol


X


1-18:2-2-16:1-monogalactosyldiacylglycerol


X


1-18:2-2-16:2-monogalactosyldiacylglycerol


X


1-18:2-2-18:2-monogalactosyldiacylglycerol


X


Momordin I


X


Isopimaric acid


X


3beta-12,21-Baccharadien-3-ol


X


24-Methyl-lanost-9(11)-en-3-beta-ol


X


Ganoderiol H


X


Petunidin 3-O-[6-O-(4-O-(E)-caffeoyl-O-


X


alpha-rhamnopyranosyl)-beta-


glucopyranoside]-5-O-beta-glucopyranoside


Phytolaccoside E


X


(1S,2S,4R,8R)-p-Menthane-1,2,9-triol


X


(25S)-26-Hydroxy-24-methylenecycloartan-3-


X


one


Panaxatriol


X


alpha-Campholene acetate


X


Gibberellin A95


X


11-Hydroxyandrosterone


X


2-Hydroxyestrone sulfate


X


TG(16:1(9Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:1(11Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:1(9Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:2(9Z,12Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:3(6Z,9Z,12Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:3(9Z,12Z,15Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(18:4(6Z,9Z,12Z,15Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(20:1(11Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(20:2n6/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(20:3n6/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(20:4(8Z,11Z,14Z,17Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(22:1(13Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(22:2(13Z,16Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(22:4(7Z,10Z,13Z,16Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(22:5(4Z,7Z,10Z,13Z,16Z)/O-


X


18:0/18:3(9Z,12Z,15Z))


TG(22:5(4Z,7Z,10Z,13Z,16Z)/O-


X


18:0/18:4(6Z,9Z,12Z,15Z))


TG(22:5(4Z,7Z,10Z,13Z,16Z)/O-


X


18:0/20:4(8Z,11Z,14Z,17Z))


TG(22:5(4Z,7Z,10Z,13Z,16Z)/O-


X


18:0/22:5(7Z,10Z,13Z,16Z,19Z))


TG(22:5(7Z,10Z,13Z,16Z,19Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


TG(24:1(15Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


Tragopogonsaponin A


X


Majoroside F4


X


Kaurenoic acid thujanol ester


X


Phaseoluside A


X


24-Methyl-lathosterol


X


24-Methylcholest-7-en-3-beta-ol


X


delta-7-Campesterol


X


Medicagenic acid 28-O-[b-D-xylosyl-(1−>4)-a-


X


L-rhamnosyl-(1−>2)-a-L-arabinosyl] ester


CDP-


X


DG(18:3(9Z,12Z,15Z)/18:3(9Z,12Z,15Z))


Petanin


X


Ganodermatriol


X


PC(18:3(9Z,12Z,15Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z))


PC(20:4(8Z,11Z,14Z,17Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z))


PC(22:5(7Z,10Z,13Z,16Z,19Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z))


PC(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


18:3(9Z,12Z,15Z))


PC(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


20:4(8Z,11Z,14Z,17Z))


PC(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z))


Torvoside B


X


28-Glucosyl-19(29)-dehydroursolic acid 3-


X


arabinoside


Periandrin V


X


Zaluzanin D


X


Dolicholide


X


4ϱ-hydroxymethyl,4ϲ,14ϱ-dimethyl-


X


9ϲ,19-cyclo-5ϱ-ergost-24(241)-en-3ϲ-ol


Acetylvalerenolic acid


X


24-Epibrassinolide


X


Brassinolide


X


trans-2-Enoyl-OPC6-CoA


X


LysoPC(18:3(9Z,12Z,15Z))


X


LysoPC(20:4(8Z,11Z,14Z,17Z))


X


LysoPC(22:5(7Z,10Z,13Z,16Z,19Z))


X


12-Hydroxy-7-oxo-8,11,13-abietatrien-18-al


X


Oxyhumulinic acid


X


24-Hydroxyglycyrrhetic acid


X


Homodolicholide


X


TG(14:1(9Z)/O-


X


18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))


Ganoderiol E


X


Gibberellin A53


X


DG(18:3(9Z,12Z,15Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0)


DG(20:4(8Z,11Z,14Z,17Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0)


DG(22:5(7Z,10Z,13Z,16Z,19Z)/


X


22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0)


DG(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


18:3(9Z,12Z,15Z)/0:0)


DG(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


20:4(8Z,11Z,14Z,17Z)/0:0)


DG(22:6(4Z,7Z,10Z,13Z,16Z,19Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z)/0:0)


Notoginsenoside J


X


Vinaginsenoside R21


X


Vinaginsenoside R22


X


7,11-Dioxolanost-8-en-3-yl acetate


X


Momordicilin


X


Oleragenoside


X


Cyanidin 3-(6-coumaryl-2′-


X


sinapoylsophoroside) 5-glucoside


24-Ethylcholesterol


X


alpha-Sitosterol


X


beta-Sitosterol


X


gamma-Sitosterol


X


Spinacetin 3-O-glucosyl-(1−>6)-


X


[apiosyl(1−>2)]-glucoside


Fauronyl acetate


X


Ergosterol peroxide


X


Hericene A


X


Hericene C


X


(3xi,4alpha,5alpha,14alpha)-4,14-


X


Dimethylergosta-8,24-dien-3-ol


4alpha,14alpha,24-Trimethyl-cholesta-8(24)-


X


dien-3beta-ol


Ginsenoyne N


X


Goyaglycoside f


X


Gibberellin A44


X


24-Methylenecycloartan-3-ol


X


3beta-24-Methylenecycloartan-3-ol


X


Torososide B


X


Androsterone


X


Epiandrosterone


X


Epietiocholanolone


X


Etiocholanolone


X


Cyclotricuspidoside C


X


TG(14:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(15:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(16:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(16:1(9Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(18:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(18:1(11Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(18:1(9Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(18:2(9Z,12Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(18:3(6Z,9Z,12Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(20:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(20:1(11Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(20:2n6/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(20:3n6/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(22:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(22:1(13Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(22:2(13Z,16Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(22:4(7Z,10Z,13Z,16Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


TG(24:0/O-18:0/22:5(4Z,7Z,10Z,13Z,16Z))


X


TG(24:1(15Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


(3beta,6alpha,19alpha)-3,6,19-Trihydroxy-12-


X


ursen-28-oic acid


Clerosterol


X


Elasterol


X


Medicagenic acid 3-O-[b-D-glucosyl-(1−>6)-b-


X


D-glucosyl-(1−>3)-b-D-glucoside]


3b,8b-Dihydroxy-6b-(3-chloro-2-hydroxy-2-


X


methylbutanoyloxy)-7(11)-eremophilen-12,8-


olide


Ginsenoside Rh2


X


Solavetivone


X


campesteryl-18:2


X


Caraganoside A


X


2-cis,4-trans-xanthoxin


X


Momordicoside E


X


(13R,14R)-8-Labdene-13,14,15-triol


X


3-O-trans-Coumaroylmaslinic acid


X


(3alpha,5alpha,7alpha)-14-Methylergosta-


X


9(11),24(28)-dien-3,7-diol


Annoglabasin F


X


Kudzusaponin SA1


X


Quinoside A


X


(E)-5,8-Megastigmadien-4-one


X


25-Acetyl-6,7-didehydrofevicordin F 3-


X


[glucosyl-(1−>6)-glucoside]


Withaperuvin F


X


Lupinisoflavone A


X


3a,7a,12a-Trihydroxy-5b-cholest-24-enoyl-


X


CoA


TG(14:1(9Z)/O-


X


18:0/22:5(4Z,7Z,10Z,13Z,16Z))


1,3,5,8-Tetrahydroxy-6-methoxy-2-


X


methylanthraquinone 8-O-b-D-glucoside


Azukisaponin III


X


Goyaglycoside a


X


CE(10:0)


X


CE(12:0)


X


CE(14:0)


X


CE(16:0)


X


CE(18:0)


X


CE(19:0)


X


CE(20:0)


X


CE(22:0)


X


CE(24:0)


X


24-Ethyllathosterol


X


7beta-Stigmasten-3beta-ol


X


Schottenol


X


Stigmast-7-en-3-ol


X


Pubesenolide


X


Medicagenic acid 3-O-beta-D-glucoside


X


2-methylbutanoyl-CoA


X


Ambonic acid


X


3-Hydroxy-28,13-lupanolide


X


Indosterol


X


beta-Eudesmol


X


Cyclopassifloside VII


X


Ganoderic acid U


X


Tuberoside C (Ullucus tuberosus)


X


CE(18:2(9Z,12Z))


X


CE(20:2(6Z,9Z))


X


CE(20:3(8Z,11Z,14Z))


X


CE(22:2(13Z,16Z))


X


CE(22:4(7Z,10Z,13Z,16Z)


X


(22:4(7Z,10Z,13Z,16Z))


X


Vulgarolide


X


2-succinyl-5-enolpyruvyl-6-hydroxy-3-


X


cyclohexene-1-carboxylate


(3beta,5alpha,24S)-Stigmasta-7,25-dien-3-ol


X


5alpha-Stigmasta-7,25-dien-3beta-ol


X


Ganoderiol A


X


3-Keto-4-methylzymosterol


X


4a-Methyl-5a-cholesta-8,24-dien-3-one


X


14alpha-Hydroxypaxilline


X


Caffeic acid ethyl ester


X


Copalic acid


X


6-Epi-beta-bisabolol


X


beta-Bisabolol


X


Formebolone


X


Cyclopassifloside II


X


1-18:3-2-18:3-phosphatidylcholine


X


PC(18:3(9Z,12Z,15Z)/18:3(9Z,12Z,15Z))


X


PC(18:3(9Z,12Z,15Z)/18:4(6Z,9Z,12Z,15Z))


X


PC(18:3(9Z,12Z,15Z)/20:4(8Z,11Z,14Z,17Z))


X


PC(18:3(9Z,12Z,15Z)/22:5(7Z,10Z,13Z,16Z,19Z))


X


PC(18:4(6Z,9Z,12Z,15Z)/18:3(9Z,12Z,15Z))


X


PC(18:4(6Z,9Z,12Z,15Z)/20:4(8Z,11Z,14Z,17Z))


X


PC(18:4(6Z,9Z,12Z,15Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z))


PC(20:4(8Z,11Z,14Z,17Z)/18:3(9Z,12Z,15Z))


X


PC(20:4(8Z,11Z,14Z,17Z)/18:4(6Z,9Z,12Z,15Z))


X


PC(20:4(8Z,11Z,14Z,17Z)/20:4(8Z,11Z,14Z,17Z))


X


PC(20:4(8Z,11Z,14Z,17Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z))


PC(22:5(7Z,10Z,13Z,16Z,19Z)/18:3(9Z,12Z,15Z))


X


PC(22:5(7Z,10Z,13Z,16Z,19Z)/18:4(6Z,9Z,12Z,15Z))


X


PC(22:5(7Z,10Z,13Z,16Z,19Z)/20:4(8Z,11Z,14Z,17Z))


X


PC(22:5(7Z,10Z,13Z,16Z,19Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z))


Austalide H


X


Kudzusaponin SA4


X


7-Ketochol esterol


X


Cycloartan-29-ol


X


Camelliagenin C


X


Liquoric acid


X


24-Methylcycloartenol


X


Cyclobranol


X


(3beta,23xi)-323-epi-Dihydroxycycloart-24-en-


X


26-oic acid


Edulisin V


X


24-hydroxy-3-oxocholest-4-en-26-oyl-CoA


X


Adenosyl cobinamide phosphate


X


5alpha-Cholest-8(14)-en-3beta-ol


X


Doristerol


X


DG(18:3(9Z,12Z,15Z)/18:3(9Z,12Z,15Z)/0:0)


X


DG(18:3(9Z,12Z,15Z)/18:4(6Z,9Z,12Z,15Z)/0:0)


X


DG(18:3(9Z,12Z,15Z)/20:4(8Z,11Z,14Z,17Z)/0:0)


X


DG(18:3(9Z,12Z,15Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z)/0:0)


DG(18:4(6Z,9Z,12Z,15Z)/18:3(9Z,12Z,15Z)/0:0)


X


DG(18:4(6Z,9Z,12Z,15Z)/20:4(8Z,11Z,14Z,17Z)/0:0)


X


DG(18:4(6Z,9Z,12Z,15Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z)/0:0)


DG(20:4(8Z,11Z,14Z,17Z)/18:3(9Z,12Z,15Z)/0:0)


X


DG(20:4(8Z,11Z,14Z,17Z)/18:4(6Z,9Z,12Z,15Z)/0:0)


X


DG(20:4(8Z,11Z,14Z,17Z)/20:4(8Z,11Z,14Z,17Z)/0:0)


X


DG(20:4(8Z,11Z,14Z,17Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z)/0:0)


DG(22:5(7Z,10Z,13Z,16Z,19Z)/


X


18:3(9Z,12Z,15Z)/0:0)


DG(22:5(7Z,10Z,13Z,16Z,19Z)/


X


18:4(6Z,9Z,12Z,15Z)/0:0)


DG(22:5(7Z,10Z,13Z,16Z,19Z)/


X


20:4(8Z,11Z,14Z,17Z)/0:0)


DG(22:5(7Z,10Z,13Z,16Z,19Z)/


X


22:5(7Z,10Z,13Z,16Z,19Z)/0:0)


hederagenin


X


Oxepahyperforin


X


Zinc sulfate


X


Gadolinium


X


BRD-K60685007


X


W-12


X


liquiritigenin


X


Ammonium Sulfate


X


Linoleic Acid


X


Niacin


X


Niacinamide


X


Taurine


X


boric acid


X


Glucans


X


beta-Glucans


X


alpha-Linolenic Acid


X


Zinc Sulfate


X


rhein


X


Ziram


X


Thiram


X


AZD8848


X


SZU-101


X


Uranium


X


BCH 1393


X


Methanol


X


Mitogens


X


Perfadex


X


Titanium


X


Ditiocarb


X


Enflurane


X


Ribomunyl


X


Acrylamide


X


allergovit


X


Nevirapine


X


Permethrin


X


Turpentine


X


cyhalothrin


X


gardiquimod


X


nonylphenol


X


Oxidopamine


X


punarnavine


X


astaxanthine


X


caffeic acid


X


CpG ODN 2006


X


enilconazole


X


FEC protocol


X


Insecticides


X


Methoxychlor


X


Penicillin G


X


saikosaponin


X


doxifluridine


X


pendimethalin


X


Penicillamine


X


Tetanus Toxoid


X


Thioglycolates


X


cerous chloride


X


Eosine I Bluish


X


Epoxy Compounds


X


Gadolinium DTPA


X


furfuryl alcohol


X


Lithium Chloride


X


o-Phthalaldehyde


X


Carbamates


X


Hypochlorous Acid


X


indoleacetic acid


X


lipoteichoic acid


X


methyl isocyanate


X


monomethylpropion


X


emulphogene BC 720


X


Isocyanates


X


Piperonyl Butoxide


X


sodium perchlorate


X


sym-trinitrobenzene


X


ciji-hua'ai-baosheng


X


tolmetin glucuronide


X


zomepirac glucuronide


X


4-bromophenacyl bromide


X


1-naphthaleneacetic acid


X


Acetylglucosamine


X


bis(tri-n-butyltin)oxide


X


isopentenyl pyrophosphate


X


Polychlorinated Biphenyls


X


Water Pollutants, Chemical


X


enterotoxin A, Staphylococcal


X


enterotoxin F, Staphylococcal


X


Vaccines, Virus-Like Particle


X


1,6-hexamethylene diisocyanate


X


taurolithocholic acid 3-sulfate


X


Heroin


X


Maneb


X


GW 7647


X


SPL-334


X


GGTI 298


X


TO-901317


X


8-Hydroxy-2-(di-n-propylamino)tetralin


X


Herbicides


X


fucoidan


X


nymphayol


X


Hemin


X



Dermatophagoides pteronyssinus antigen p 1



X


Iodine


X


MK 473


X


NS3728


X


TBE 31


X


GW 1929


X


NS 1652


X


resiniferatoxin


X


Dimaprit


X


Peptidoglycan


X


Butyrates


X


taxifolin


X


trans-phytol


X


Vanadates


X


Berberine


X


Erianin


X


GW 506033X


X


herbimycin


X


malondialdehyde-low density lipoprotein,


X


human


manumycin


X


molibresib


X


protein-bound polysaccharide K,


X


Basidiomycetes


Ro 41-5253


X


sauchinone


X


2-amino-1-methyl-6-phenylimidazo(4,5-


X


b)pyridine


geraniol


X


centipedic acid


X


Coumarins


X


hiiranlactone D


X


tetrandrine


X


Clodronic Acid


X


Alcohols


X


alpha-galactosylceramide


X


bromodichloromethane


X


Ellagic Acid


X


Phenylbutyrates


X


Propanil


X


5-aminoisoquinolinone


X


caryophyllene oxide


X


Silymarin


X


4-methylhistamine


X


diethylamine


X


hesperetin


X


chlorophyllin


X


Cholera Toxin


X


(glycyl-glycyl)GLP-2


X


salvianolic acid A


X


notoginsenoside R1


X


5′-methylthioadenosine


X


cyanoginosin LR


X


Malondialdehyde


X


1-trifluoromethoxyphenyl-3-(1-propionylpiperidine-4-yl)urea


X


decursin


X


Rifaximin


X


sinomenine


X


ginsenoside Rg1


X


tetramethylpyrazine


X


Glyoxal


X


Manganese


X


dai-kenchu-to


X


dibenzothiazyl disulfide


X


Melatonin


X


Organoselenium Compounds


X


12-acetoxyhawtriwaic acid lactone


X


1-oleoyl-2-acetoyl-sn-glycerol


X


S-Nitroso-N-Acetylpenicillamine


X


Antigens


X


Cholesterol, Dietary


X


NG-Nitroarginine Methyl Ester


X


oleanolic acid 3-acetate


X


Fatty Acids, Unsaturated


X


resiquimod


X


1,1-dimethylbutyl-1-deoxy-Delta(9)-THC


X


BRL 44408


X


Dinoprost


X


2,6-diaminohexanoic acid (4-(5-


X


fluorobenzothiazol-2-yl)-2-


methylphenyl)amide


polydatin


X


((1-ethylpyrrolidin-2-yl)methyl)-4-hydroxy-7-


X


methoxy-quinoline-3-carboxamide


zingerone


X


nifuroxazide


X


platycodin D


X


5-nitro-2-(3-phenylpropylamino)benzoic acid


X


kuwanon G


X


butane-2,3-dione thiosemicarbazone oxime


X


licochalcone A


X


alpha-cyano-(3,4-dihydroxy)-N-


X


benzylcinnamide


coniferaldehyde


X


wogonin


X


catalpol


X


Camosine


X


Glyburide


X


heptanoyl-gamma-D-glutamyl-L-meso-


X


diaminopimelyl-D-alanine


pimagedine


X


Nitrofurazone


X


cholan-2,3,24-tryl-2,3,24-trisulfate


X


glutathione-C4


X


I-152 compound


X


N-(3-propylcarbamoyloxirane-2-carbonyl)-


X


isoleucyl-proline


1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl)


X


imidazole


4-cresol sulfate


X


7,14-dihydroxydocosa-4,8,10,12,16,19-


X


hexaenoic acid


Lithium Carbonate


X


2,4-diaminotoluene


X


lactacystin


X


adefovir dipivoxil


X


Sulfobromophthalein


X


Trichloroacetic Acid


X


Aminosalicylic Acids


X


omega-N-Methylarginine


X


2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic


X


acid ethyl amide



Mangifera indica extract



X


Organometallic Compounds


X


Heme


X


phenyl-N-tert-butylnitrone


X


Pam(3)CSK(4) peptide


X


seryl-leucyl-isoleucyl-glycyl--arginyl-


X


leucinamide


4,4′,4″-(4-propyl-((1)H)-pyrazole-1,3,5-triyl)


X


tris-phenol


2-(1′H-indole-3′-carbonyl)thiazole-4-


X


carboxylic acid methyl ester


orazipone


X


GW0072


X


SN50 peptide


X


latrunculin B


X


Thiourea


X


Cytochalasin D


X


jasplakinolide


X


cyanidin 3-O-glucoside


X


2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine


X


3,5-diethoxycarbonyl-1,4-dihydrocollidine


X


2-(2-amino-3-methoxyphenyl)-4H-1-


X


benzopyran-4-one


4,4′-Diisothiocyanostilbene-2,2′-Disulfonic


X


Acid


Biliverdine


X


Methylene Chloride


X



Dermatophagoides pteronyssinus antigen p 2



X


Chaicones


X


2-fluoro-N-methyl-4-(7-(quinolin-6-yl-


X


methyl)imidazo(1,2-b)(1,2,4)triazin-2-


yl)benzamide


toxB protein, Clostridium difficile


X


1-naphthylethyl-6,7-dihydroxy-1,2,3,4-


X


tetrahydroisoquinoline


2-hydroxy-4-(2,2,3,3,3-


X


pentafluoropropoxy)benzoic acid


3,4-dihydro-5-(4-(1-piperidinyl)butoxy)-1(2H)-


X


isoquinolinone


2-methyl-2H-pyrazole-3-carboxylic acid (2-


X


methyl-4-o-tolylazophenyl)amide


2-morpholin-4-yl-6-thianthren-1-yl-pyran-4-


X


one


2-(6-((3-(4-


X


(diphenylmethoxy)piperidino)propyl)amino)imidazo(1,2-


b)pyridazin-2-yl)-2-methylpropionic acid


1-(beta-naphthylmethyl)-6,7-dihydroxy-


X


1,2,3,4-tetra-hydroisoquinoline


platycodigenin 3-O-(glucopyranosyl-(1-3)-


X


glucopyranosyl)-28-O-(apiofuranosyl-(1-3)-


xylopyranosyl-(1-4)-rhamnopyranosyl-(1-2)-


arabinopyranoside)


3-O-methylsphingomyelin


X


N-(2-(4-(2-methoxyphenyl)-1-


X


piperazinyl)ethyl)-N-(2-


pyridinyl)cyclohexanecarboxamide


N-((4-dimethylaminophenyl)methyl)-N-(4-


X


isopropylphenyl)-7-methoxy-1,2,3,4-


tetrahydronaphthalen-1-carboxamide


1,1-bis(3′-indolyl)-1-(4-


X


trifluoromethylphenyl)methane


Imiquimod


X


Hemagglutinins


X


beryllium sulfate


X


Imatinib Mesylate


X


Active Hexose Correlated Compound


X


Dacarbazine


X


Mevalonic Acid


X


Sulfapyridine


X


geranylgeranyl pyrophosphate


X


Mesalamine


X


methyldithiocarbamate


X


Environmental Pollutants


X


Reactive Oxygen Species


X


BRD-K41143549



X
X


meloxicam



X
X


BRD-K69021489



X
X


BRD-A38425832



X
X


BRD-K55216615



X
X


oxymetazoline



X
X


BRD-K93578426



X
X


BRD-K52446745



X
X


saclofen



X
X


triprolidine



X
X


lavendustin-a



X
X


BRD-K58674774



X
X


BRD-K45205755



X
X


domperidone



X
X


BRD-K00442147



X
X


glycopyrrolate



X
X


acetyl-geranyl-cysteine



X
X


BRD-A27640568



X
X


zolmitriptan



X
X


ioxaglic-acid



X
X


oxiconazole



X
X


BRD-K76999748



X
X


benzthiazide



X
X


BRD-A01999384



X
X


BRD-K11362964



X
X


BRD-K37340241



X
X


SA-94315



X
X


BRD-A81416315



X
X


BRD-K84677391



X
X


glycodeoxycholic-acid



X
X


BRD-K36767861



X
X


oleanolic-acid



X
X


BRD-K29500995



X
X


BRD-K74711637



X
X


UGISS-451



X
X


RO-04-5595



X
X


propranolol



X
X


quercetagetin



X
X


L-745870



X
X


CAY-10577



X
X


BRD-K60228169



X
X


DH-97



X
X


ML-4054



X
X


tolmetin



X
X


BRD-K67619794



X
X


TUL-XVIII076



X
X


BRD-K70960308



X
X


ST-4075437



X
X


RO-16-6941



X
X


BRD-K00637033



X
X


RDR-01440SC



X
X


BRD-K40480443



X
X


BRD-A83706824



X
X


BRD-K76188144



X
X


BRD-K93127801



X
X


BRD-K35961280



X
X


BRD-K78481586



X
X


somatostatin



X
X


BRD-K12105018



X
X


BRD-K74767048



X
X


BRD-K40577058



X
X


lithocholic-acid



X
X


BRD-K74109131



X
X


BRD-K09902564



X
X


hyperoside



X
X


BRD-A84445767



X
X


BRD-K45232279



X
X


KUC104235N



X
X


flupirtine



X
X


BRD-A88029023



X
X


BRD-K73700643



X
X


BRD-K06206016



X
X


BRD-A45206415



X
X


didanosine



X
X


BRD-A83802330



X
X


2-(4-methoxybenzylthio)-6-methylpyrimidin-



X
X


4-ol


rondual-kinase-inhibitor



X
X


BRD-K62719675



X
X


ZD-7155



X
X


BRD-K74155249



X
X


Isoproterenol



X
X


5,6-trans-25-Hydroxyvitamin D3



X


25,26-Dihydroxyvitamin D3



X


1-alpha,24R,25-Trihydroxyvitamin D2



X


24,25-Dihydroxyvitamin D3



X


24R,25-Dihydroxyvitamin D3



X


25-Hydroxyvitamin D2



X


25-Hydroxycholecalciferol



X


25-Hydroxytachysterol3



X


24-Hydroxycalcitriol



X


2-[4-(3-Hydroxypropyl)-2-methoxyphenoxy]-



X


1,3-propanediol 1-xyloside


Momordicoside C



X


Roxarsone



X


BRD-A30083233



X


linoleamide



X


BRD-K63368502



X


BRD-K32321439



X


BRD-K44931544



X


raltegravir



X


BRD-A13461302



X


BRD-K02381115



X


BRD-K16346331



X


PD-169316



X


SDZ-WAG-994



X


BRD-K18970629



X


homoveratrylamine



X


BRD-K56983253



X


PSH-026



X


entacapone



X


BRD-K17773477



X


BRD-K15107389



X


BRD-K72864428



X


ramipril



X


BRD-K63157263



X


BRD-K31453413



X


BRD-K27881911



X


piperacetazine



X


KU-C103676



X


BRD-K30523950



X


VUF-5681



X


BRD-K46532897



X


cefotaxime



X


BRD-K65774694



X


ST-014147



X


coumarin



X


ichthynone



X


cromoglicic-acid



X


prima-1-met



X


BRD-K19027210



X


Capecitabine



X


BRD-K83589469



X


ropivacaine



X


BRD-K51126483



X


aminoindazole



X


BRD-K67742177



X


ST-012030



X


DM-163



X


prestwick-559



X


sulfafurazole



X


BRD-K95117060



X


BRD-K79251086



X


ungerine



X


KM-03439



X


etifenin



X


BRD-K37618507



X


strychnine



X


BRD-A13346522



X


JAS07-001



X


BRD-K17969842



X


BRD-K32949592



X


mepyramine



X


BRD-K99303177



X


BRD-K06652922



X


BRD-K32580836



X


I-OMe-AG-538



X


BRD-K70974948



X


diphemanil



X


BRD-K92489476



X


SD-6-035-A1



X


BRD-K71879957



X


BRD-K53445087



X


levetiracetam



X


BRD-K08777248



X


BRD-K49477212



X


troxipide



X


JAS07-008



X


icosapent



X


benzydamine



X


BRD-K04136539



X


ARG-A1-2



X


BRD-K83929831



X


L-693403



X


BRD-K34596778



X


quinapril



X


zaldaride



X


BRD-K14842454



X


SC-10



X


BRD-K71140693



X


KU-C103868



X


UBP-302



X


DY-44



X


BRD-K32132269



X


BRD-K56446541



X


metoclopramide



X


diprotin-a



X


BRD-K35100517



X


BRD-K29138830



X


CEP 33779



X


Roxarsone



X


Astemizole



X


perfosfamide



X


adrenocorticotropin zinc



X


endotoxin, Escherichia coli



X


1,2-dielaidoylphosphatidylethanolamine



X


N-Methyl-3,4-methylenedioxy amphetamine



X


N-(2-(1,1′-bicyclopropyl)-2-ylphenyl)-3-



X


(difluoromethyl)-1-methyl-1H-pyrazole-4-


carboxamide


ethylene dichloride



X


Pertussis Toxin



X


Malonyl CoA



X


Cinnamoyl-CoA



X


4-Coumaroyl-CoA



X


Hordatine B glucoside



X


2-Ethylpyridine



X


D-Perillaldehyde



X


4,6-Benzylidene-D-glucose



X


Oleoresin



X


alpha-Himachalene



X


Eugenyl benzoate



X


Guar gum



X


8-Acetoxy-4′-methoxypinoresinol



X


8-Acetoxy-4′-methoxypinoresinol 4-glucoside



X


8-Acetoxypinoresinol



X


4-Methoxycinnamoyloleanolic acid methyl



X


ester


N1,N10-Dicoumaroylspermidine



X


Ceftiofur



X


Vinaginsenoside R15



X


Xanthohumol B



X


Ursolic aldehyde



X


(R)-Perillaldehyde



X


Avenestergenin B2



X


(6b,7b,13R)-6,7-Diacetoxy-8,14-labdadiene-



X


13-ol


Galanal B



X


(S)-Perillaldehyde



X


Perillaldehyde



X


2-[(2-Furanylmethyl)thio]-6-methylpyrazine



X


Eugenyl formate



X


gamma-Himachalene



X


(1(10)E,4a,5E)-1(10),5-Germacradiene-12-



X


acetoxy-4,11-diol


Medicagenic acid 28-O-[b-D-xylosyl-(1−>4)-a-



X


L-rhamnosyl-(1−>2)-a-L-arabinosyl] ester


Curcumin II



X


Grossamide



X


3alpha-12-Ursene-3,24-diol



X


Enkephalin L



X


Endomorphin-2



X


Arginine vasopressin 1-8



X


Dynorphin B



X


(24R,25R)-3ϱ,7ϱ,12ϱ,24-tetrahydroxy-



X


5ϲ-cholestanoyl CoA


(24R,25R)-3ϱ,7ϱ,24-trihydroxy-5ϲ-



X


cholestanoyl CoA


(E)-cinnamoyl-CoA



X


(S)-3-hydroxybutanoyl-CoA



X


(S)-3-hydroxyhexanoyl-CoA



X


3-cis-dodecenoyl-CoA



X


4-(2′-carboxyphenyl)-4-oxobutyryl-CoA



X


monovinyl protochlorophyllide a



X


N-methylanthraniloyl-CoA



X


4′-Methylepicatechin 3′-sulfate



X


Catechin 3′-sulfate



X


Val-Tyr-Leu-Arg



X


biotin




X


caffeine




X


remoxipride




X


GR-113808




X


benzylpenicillin




X


larixinic-acid




X


BRD-K10846167




X


SCH-23390




X


BRD-A51929314




X


BRD-K36817836




X


BRD-A96272097




X


BRD-K40955748




X


BRD-U32049204




X


oxalomalic-acid




X


BRD-A03436432




X


parachlorophenol




X


BRD-K21237738




X


flucytosine




X


ropinirole




X


SD-6-035-B2




X


VU-0400193-3




X


BRD-K98676510




X


BRD-K53816294




X


dehydroisoandosterone




X


BRD-K65404805




X


L-655708




X


maackiain




X


SC-68376




X


BRD-K75784474




X


fenbufen




X


BRD-K14975149




X


3,6-dimethoxyflavone




X


SA-423131




X


acebutolol




X


BRD-K51454562




X


thioperamide




X


NNC-63-0532




X


azacyclonol




X


BRD-K25784575




X


ST-208960




X


BRD-K10609180




X


moxonidine




X


BRD-K33676730




X


BRD-K58223721




X


BRD-K98079119




X


BML-257




X


hydrocotarnine




X


ethisterone




X


BRD-K97951054




X


BRD-K90705745




X


medetomidine




X


benzatropine




X


BRD-K68783079




X


perospirone




X


BRD-K11883928




X


BRD-A48052484




X


BRD-K47695085




X


azaperone




X


BRD-K46171503




X


VU-0415556-1




X


apafant




X


BRD-K88735704




X


BRD-K69231968




X


KUC103903N




X


pemoline




X


BRD-K56855930




X


BRD-K74348865




X


metyrapone




X


BRD-K52640952




X


buspirone




X


PETCM




X


BRD-K57443339




X


BRD-K42006476




X


BRD-K91836367




X


BRD-A69421747




X


BRD-K37124285




X


BRD-K91818252




X


KU-C104226




X


BRD-K27170250




X


BRD-K33586157




X


SA-103123




X


BRD-K27290412




X


BRD-K26822808




X


BRD-K75329208




X


ketotifen




X


BRD-K76595177




X


BRD-K73888531




X


bethanechol




X


BRD-K22006123




X


BRD-K19875179




X


CO-102862




X


alfaxalone




X


telenzepine




X


RO-25-6981




X


BRD-K22828860




X


BRD-K45675241




X


BRD-K48199208




X


BRD-A98050686




X


BRD-K99135512




X


BRD-K53142391




X


leu-enkephalin




X


BRD-K00816165




X


BRD-K71292008




X


cefalexin




X


BRD-K00555438




X


BRD-A06131576




X


BRD-K10806285




X


benazepril




X


BRD-K36317223




X


BRD-K91671201




X


eudesmic-acid




X


BRD-K28680267




X


pirenzepine




X


BRD-K37413063




X


quinpirole




X


BRD-K26367064




X


BRD-K27305388




X


andarine




X


pirinixic-acid




X


KU-C104491




X


BRD-K69496360




X


BRD-K47147974




X


nafcillin




X


BRD-K70524666




X


EHNA




X


CS-1657




X


BRD-K19117583




X


BRD-K63784565




X


JAS07-007




X


tropicamide




X


KUC103420N




X


BRD-K51083659




X


BRD-K22384978




X


flurofamide




X


AICA-ribonucleotide




X


BRD-K28040935




X


WAY-161503




X


BRD-A82976242




X


BRD-K13842623




X


ormetoprim




X


BRD-K23644387




X


BRD-K59633395




X


BIX-01338




X


BRD-K90667125




X


relcovaptan




X


BRD-K17830172




X


BRD-K55071917




X


BRD-K16046246




X


ST-4029487




X


BRD-A74134989




X


pseudopelletierine




X


BRD-A29721261




X


BRD-K58784778




X


diphenidol




X


BRD-K88059273




X


amantadine




X


BRD-K17693482




X


K784-3391




X


ryanodine




X


busulfan




X


RS-67333




X


ponalrestat




X


cinalukast




X


BRD-K73418978




X


BRD-K76612204




X


KU-C104253




X


BRD-A74699940




X


BRD-K46445327




X


Ro-04-6790




X


BRD-K53327986




X


BRD-K06432680




X


bicuculline




X


BRD-K71746704




X


BRD-K98025142




X


BRD-K55972177




X


BRD-K76293260




X


BRD-K58033748




X


dosulepin




X


BRD-K63801873




X


BRD-K12182524




X


ARG-CSC2-23




X


BRD-K82230692




X


haematoxylin-pentaacetate




X


BRD-K23774426




X


BRD-K30802967




X


Iron




X


Cytarabine




X


Aluminum oxide




X


Oxyquinoline




X


N,N-dimethylformamide




X


Styrene




X


casticin




X


cinnabar




X


danthron




X


vicanicin




X


Cytarabine




X


Cantharidin




X


fusarenon-X




X


riddelliine




X


vinclozolin




X


Oxyquinoline




X


panduratin A




X


sanguinarine




X


tinuvin




X


Dimethylformamide




X


Aluminum Oxide




X


protolichesterinic acid




X


1,2,5,6-dibenzanthracene




X


Ro 31-8220




X


tanshinone




X


Egtazic Acid




X


10′(Z),13′(E),15′(E)-




X


heptadecatrienylhydroquinone


Thiophenes




X


Aluminum




X


Viper Venoms




X


staurosporine aglycone




X


hispanolone




X


Glatiramer Acetate




X


3,4-dihydroxybenzohydroxamic acid




X


N-(2-aminoethyl)-5-chloroisoquinoline-8-




X


sulfonamide


chrysene




X


2-(1H-indazol-4-yl)-6-(4-




X


methanesulfonylpiperazin-1-ylmethyl)-4-


morpholin-4-ylthieno(3,2-d)pyrimidine








Claims
  • 1. A method, comprising the steps of: obtaining a sample from a subject; and measuring the expression of three or more genes in a cell in the sample from the subject selected from a group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein only genes from the group are measured.
  • 2. The method of claim 1, wherein the sample is a blood sample.
  • 3. The method of claim 1, wherein the sample is a serum sample.
  • 4. The method of claim 1, wherein the measuring of the three or more genes is done in a multiplex assay.
  • 5. The method of claim 4 further comprising the step of measuring a cholesterol, a gender or a BMI for the subject.
  • 6. The method of claim 1, further comprising the step of measuring the expression of a fourth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the fourth gene is different from the three genes.
  • 7. The method of claim 6, further comprising the step of measuring the expression of a fifth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 8. The method of claim 7, further comprising the step of measuring the expression of a sixth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 9. The method of claim 8, further comprising the step of measuring the expression of a seventh gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 10. The method of claim 9, further comprising the step of measuring the expression of an eighth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 11. The method of claim 10, further comprising the step of measuring the expression of a tenth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 12. The method of claim 11, further comprising the step of measuring the expression of an eleventh gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 13. The method of claim 12, further comprising the step of measuring the expression of a twelfth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 14. The method of claim 13, further comprising the step of measuring the expression of a thirteenth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 15. The method of claim 14, further comprising the step of measuring the expression of a fourteenth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 16. The method of claim 15, further comprising the step of measuring the expression of a fifteenth gene in the sample from the subject selected from the group consisting of a GBP5, a MMP9, a SIGLEC5, a S100P, an OLFM1, a CISH, a MT1A, a CHURC1, an IGLL1, a RPLP0, a SLC16A10, a FCER1A, a CD248, a DDX3Y, and a MAN1A1, wherein the genes measured are different.
  • 17. The method of claim 1, wherein expression of the three or more genes are measured with a RNAseq.
  • 18. The method of claim 1, wherein expression of the three or more genes are measured with a PCR assay.
  • 19. The method of claim 16, wherein expression of the fifteen genes is measured with a RNAseq.
  • 20. The method of claim 16, wherein the expression of the fifteen genes are measured with a polymerase chain reaction.
Provisional Applications (1)
Number Date Country
63156868 Mar 2021 US