WO2005098448A2 - Method and biomarkers for detecting tumor endothelial cell proliferation - Google Patents

Method and biomarkers for detecting tumor endothelial cell proliferation Download PDF

Info

Publication number
WO2005098448A2
WO2005098448A2 PCT/US2005/009874 US2005009874W WO2005098448A2 WO 2005098448 A2 WO2005098448 A2 WO 2005098448A2 US 2005009874 W US2005009874 W US 2005009874W WO 2005098448 A2 WO2005098448 A2 WO 2005098448A2
Authority
WO
WIPO (PCT)
Prior art keywords
endogro
bfgf
vegf
genes
tumor
Prior art date
Application number
PCT/US2005/009874
Other languages
French (fr)
Other versions
WO2005098448A3 (en
Inventor
James Hardwick
Hongyue Dai
John R. Lamb
Laura Sepp-Lorenzino
Michael E. Severino
Chunsheng Zhang
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CA002561338A priority Critical patent/CA2561338A1/en
Priority to JP2007505185A priority patent/JP2007530047A/en
Priority to EP05731529A priority patent/EP1733051A4/en
Publication of WO2005098448A2 publication Critical patent/WO2005098448A2/en
Publication of WO2005098448A3 publication Critical patent/WO2005098448A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the field of this invention relates to methods, biomarkers, and expression signatures for assessing the proliferative rate of vascular endothelial cells within tumors. More specifically, the invention provides a set of genes which can be used as biomarkers for evaluating the pharmacodynamic effects of cancer therapies designed to regulate the proliferation of endothelial cells in tumor vasculature. In one aspect the invention provides a method of evaluating the efficacy of a compounds designed to inhibit kinase receptor activity, such as a mammalian KDR receptor activity.
  • Vascular endothelial cells form a luminal non-thrombogenic monolayer throughout the vascular system. Solid tumors require a vascular system to expand beyond small nodules limited by the diffusion of nutrients and metabolic by products. Although tumor cells can initially colonize existing host capillaries, their growth leads to the collapse of these preexisting normal vessels resulting in hypoxia. Therefore, angiogenesis is critical to the progression of numerous cancers.
  • tumor angiogenesis neovascularization that is achieved by the ingrowth of new host blood vessels.
  • Tumors induce proliferation, migration and differentiation resulting in neovascularization by secreting growth factors for vascular endothelial cells.
  • Angiogenesis is critical to the progression of numerous cancers. Tumors induce endothelial cell migration, proliferation and differentiation resulting in neovascularization arising from existing blood vessels. Tumor cells induce angiogenesis primarily through the production and secretion of vascular endothelial growth factor (VEGF), a secreted protein that is a potent endothelial cell mitogen and ligand for the kinase insert domain receptor (KDR, FLK-1, or VEGF receptor).
  • VEGF vascular endothelial growth factor
  • Tyrosine kinases are a class of enzymes that catalyze the transfer of the terminal phosphate of adenosine triphosphate to tyrosine residues in protein substrates. Tyrosine kinases are believed, by way of substrate phosphorylation, to play critical roles in signal transduction for a number of cell functions and have been shown to be important contributing factors in cell proliferation, carcinogenesis and cell differentiation. Tyrosine kinases can be categorized as receptor type or non receptor type.
  • Receptor type tyrosine kinases typically have an extracellular, a transmembrane, and an intracellular portion, while non- receptor type tyrosine kinases typically are wholly intracellular, while examples exist of membrane receptors that upon ligand binding recruit intracellular kinases to bind to the intracellular portion of the receptor which, by itself, does not have kinase activity. Both receptor-type and non-receptor type tyrosine kinases are implicated in cellular signaling pathways leading to numerous pathogenic conditions, including cancer, psoriasis and hyperimmune responses.
  • the receptor-type tyrosine kinases are comprised of a large number of transmembrane receptors with diverse biological activity.
  • KDR kinase insert domain receptor
  • Tumor cells induce angiogenesis primarily through the production and secretion of vascular endothelial growth factor (VEGF) a potent endothelial cell mitogen and ligand for the kinase insert domain receptor (KDR, FLK-1, or VEGF receptor 2) (3-7).
  • VEGF vascular endothelial growth factor
  • KDR kinase insert domain receptor
  • FLK-1 FLK-1
  • VEGF receptor 2 kinase insert domain receptor 2
  • VEGF binds with high affinity to two transmembrane tyrosine kinase-linked receptors, Flt-1 (VEGFR-1) and KDR (Flk-l/VEGFR-2), that are expressed by vascular endothelial cells.
  • KDR initiates a signal transduction cascade, is internalized and ultimately degraded. Inhibition of the VEGF/KDR system has been shown to inhibit VEGF-dependent tumor angiogenesis and growth in several animal models. Because VEGF produced and secreted by tumor cells activates KDR and induces endothelial cell proliferation, it is acknowledged that inhibition of KDR kinase activity should lead to decreases in the proliferation of tumor endothelial cells. Accordingly, numerous proposed cancer therapeutics target vascular endothelial cell growth factor (VEGF) or the kinase insert domain receptor (KDR/VEGFR-2/FLK-l), the primary VEGF receptor on endothelial cells.
  • VEGF vascular endothelial cell growth factor
  • KDR/VEGFR-2/FLK-l kinase insert domain receptor
  • KDR is a particularly attractive target to antagonize VEGF-dependent tumor angiogenesis and growth. Inhibition of KDR catalytic activity blocks tumor neoangiogenesis, reduces vascular permeability, and in animal moldes, inhibits tumor growth and metastasis.
  • KDR protein is not expressed at high levels in readily accessible biological material, such as peripheral blood or bone marrow aspirates, clinical assessment of the in vivo pharmacodynamic efficacy of KDR kinase inhibitors is challenging.
  • One described method for the in vivo assessment of EC proliferation involves dual immunohistochemical (MC) staining of tumor sections for the endothelial cell marker CD-31 and a nuclear marker of cellular proliferation, Ki-67(11). While an immunohistochemical method such as this can determine the fraction of ECs that are proliferating, the experimental protocol is technically complex and difficult and the analysis required for each stained tumor section is extremely time-consuming. Each of these factors makes clinical use of an MC-based assay unlikely.
  • the methods disclosed and claimed herein are based on the discovery and characterization of biomarkers and gene expression signatures that are specific for proliferating endothelial cells.
  • Gene expression profiling data from cultured primary endothelial cells, cultured tumor cells, and tissue from animal tumor models treated with KDR inhbitors was used to identify a set of genes that are selectively overexpressed in tumor endothelial cells relative to tumor cells, and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation. It is contemplated that the biomarkers and endothelial cell-specific expression signatures which are disclosed and claimed herein will find utility in the context of providing a pharmacodynamic readout for any cancer therapy that aims to inhibit proliferation of endothelial cells in tumor vasculature.
  • the expression levels of these genes serve as the basis of a simple pharmacodynamic assay for the activity of small molecule inhibitors of the KDR receptor tyrosine kinase.
  • the methods disclosed and claimed herein can be used as the basis for a pharmacodynamic assay capable of supporting the clinical development of small molecule inhibitors of the KDR receptor tyrosine kinase.
  • the invention provides a method for assessing the in vivo effects of a KDR kinase inhibitor on the proliferative rate of vascular endothelial cells within tumors. In one aspect the invention provides a method for determining the proliferative status (or rate) of endothelial cells.
  • the disclosed method can be used to evaluate the proliferative status of endothelial cells in either an in vitro or in vivo format.
  • the disclosed gene expression-based pharmacodynamic assays which can be established based on the disclosure provided herein can be used to support screening assays established to evaluate the efficacy of therapeutic agents intended to regulate the proliferative status of endothelial cells.
  • the invention provides a method for evaluating the proliferative rate of vascular endothelial cells within tumors.
  • the invention provides a gene expression-based pharmacodynamic assay that is suitable for use to support clinical development of cancer therapies designed to regulate the proliferation of endothelial cells in tumor vasculature.
  • the disclosed methods can be used to establish pharmacodynamic assays that can distinguish tumors containing proliferating endothelial cells from tumors containing mostly quiescent endothelial cells.
  • the method may include detecting the expression level of one or more genes selected from a group consisting of Angpt-2, Clu (ApoJ), Cyr ⁇ l (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA).
  • the invention provides a method for evaluating the activity of anti-angiogenesis therapeutics intended to regulate the proliferative rate of vascular endothelial cells within tumors.
  • the invention provides a method for evaluating the efficacy of small molecule inhibitors of receptor-type kinase inhibitors, such as KDR.
  • this aspect of the invention provides a method which is suitable for use to support clinical development of KDR kinase inhibitors, such as Compound A. Using the information provided herein, particularly the Compound A-and B- induced endothelial cell-specific expression signatures provided in Tables 5.
  • Table 6 provides the summary information describing the changed (suppressed) expression of endothelial cell specific biomarkers (collectively referred to as the proliferation sequence) observed in response to the in vivo administration of KDR Kinase inhibitors (Compounds A and B), provided in Table 6. It is well within the abilities of a skilled artisan to design and validate a gene expression-based assay that is suitable for evaluating the efficacy of anti-angiogenesis agents. It is contemplated that using the disclosure provided herein a skilled artisan can utilize the information provided in the tables summarizing the proliferation signatures disclosed herein to identify compound-specific expression signatures that will facilitate evaluating the efficacy of alternative therapeutic agents intended to regulate endothelial cell proliferation.
  • this aspect of the invention provides a method which is suitable for use to support clinical development of KDR kinase inhibitors, such as Compound A.
  • KDR kinase inhibitors such as Compound A.
  • the efficacy of Compound A could be evaluated in vivo by establishing an assay which detects changes in the expression of a gene signature comprising the Angpt-2, Clu (ApoJ), Cyr ⁇ l (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA) genes.
  • the invention provides a composition of genes or biomarkers which are selectively overexpressed in tumor endothelial cells relative to tumor cells, and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation.
  • compositions comprising at least two oligonucleotides, wherein each of the oligonucleotides comprises a sequence that specifically hybridizes to a gene disclosed in Tables 3 or 4 as well as solid supports comprising at least two probes, wherein each of the probes comprises a sequence that specifically hybridizes to a gene in Tables 3, 4, 5 or 6.
  • the composition will comprise oligonucleotides and/or probes which hybridize with the following genes: Angpt-2, Clu (ApoJ), Cyr ⁇ l (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA), in combination with other oligonucleotides or probes specific for other genes identified in Tables 3-6.
  • the invention provides gene expression signatures, which can be used to establish expression-based pharmacodynamic assays for evaluating the efficacy of therapeutic agents designed to regulate the proliferation of endothelial cells.
  • the gene expression signature disclosed and claimed herein can be used distinguish tumors containing mostly proliferating ECs from tumors containing mostly quiescent ECs. It is contemplated that the disclosed assay will have the ability to detect inhibition of angiogenesis relatively quickly after initiating therapy, eliminating the longer period of time required to visualize morphological changes in tumor microvasculature. In addition, it is envisioned that the disclosed methods will be particularly useful in circumstances where immunohistochemistry is inappropriate or impractical, such as with small tissue samples from biopsies (i.e. fine needle aspirates) or from tissue samples with poor morphology.
  • the disclosed assay In a real time quantitative reverse transcription- polymerase chain reaction (PCR) format, the disclosed assay is predicted to represent an extremely sensitive assay that is readily compatible with existing clinical laboratory instrumentation. Expression profiling-based monitoring of the pharmacodynamic effects of cancer therapy potentially has many benefits. Used in the clinical setting, this technology provides for rapid, quantitative, reproducible, and inexpensive assays that are compatible with current clinical laboratory instrumentation. Carefully designed, gene expression-based asssays, such as the assays disclosed herein, have the potential to make dosing of anti-neoplastic agents more efficient, to identify patient populations most likely to benefit from specific therapies, and to reduce clinical development time of novel therapeutics. Each of these aspects will lead to increased tumor response rates and improved human health.
  • PCR reverse transcription- polymerase chain reaction
  • VEC Vascular Endothelial Cell
  • HDMVECs or RHMVECs were trypsinization following the third passage in culture and seeded in fibronectin-coated, six-well tissue culture plates at a density of 10,000 cells/well. Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 ⁇ g/ml ENDOGRO. Wells with unstimulated cells and wells containing un-arrested cells were included as controls.
  • FIG. 2A and 2B Identification of a gene expression profile in proliferating vascular endothelial cells in vitro.
  • HDMVECs and RHMVECs were grown in culture and mitogen deprived for 24 hr as described in Figure 1 and Methods.
  • culture media was aspirated quickly and the cells lysed in an RNA stabilizing buffer. Matched control plates that received no supplemental stimulatory growth factor were present for each stimulation condition and RNAs isolated from them served as the reference to which the RNAs from the stimulated cells was compared.
  • FIG. 3A-3D Bars corresponding to genes which are regulated (e.g., upregulated or downregulated) are indicated by various shades of gray. Color intensity represents the degree of regulation, not mRNA copy number.
  • Figures 3A-3D Specific suppression of VEGF-induced gene expression in cultured vascular endothelial cells. EC monolayers were maintained in complete MCDB-131 media until reaching -75% confluence, then induced into a quiescent state by mitogen starvation for 24 hr. Cells were then stimulated to proliferate with 100 ng/ml VEGF for 24 hr in the presence or absence of Compound B.
  • RNA populations isolated from cells exposed to VEGF or VEGF + Compound B were compared to matched control RNAs isolated from quiescent cells exposed to neither VEGF nor Compound B.
  • Each point in the plots represents a gene sequence present on the DNA oligonucleotide microarray and is plotted according to the ratio of the two mRNA levels (experimental sample intensity: control sample intensity, vertical-axis) and the total mRNA quantity (experimental sample intensity + control sample intensity, horizontal-axis) for that gene. Dark-colored points indicate upregulated genes. Light Gray colored points indicate downregulated genes.
  • Figures 4A and 4B Growth kinetics of established rat tumors following exposure to a KDR kinase inhibitor. Tumor studies were performed as described in Materials and Methods.
  • FIGS 4A and 4B illustrate tumor volumes from animals in the C6 profiling study (Fig. 4A) and the Mattlll profiling study (Fig. 4B) as determined by caliper measurements. Tumors were calipered in two dimensions (length and width) and tumor volume was calculated according to the formula (length) x (width) x (V2 width).
  • Figures 5 A-5C Identification of gene expression changes induced in rat tumors by KDR kinase inhibitors in vivo.Each row represents a distinct tumor from an individual animal. Each column represents a gene. Gray colored points/bars indicate genes that are regulated (e.g., upregulated or downregulated) by KDR kinase.
  • Figure 5A illustrates changes in the expression of genes from rat C6 flank tumors that are regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound B.
  • Figure 5B illustrates changes in the expression of genes from rat C6 flank tumors regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound A.
  • Figure 5C illustrates changes in the expression of genes from rat MatBHI mammary tumors regulated following 100 hrs of systemic exposure to the KDR kinase inhibitor Compound A.
  • Figures 6A-6B illustrates changes in the expression of genes from rat C6 flank tumors that are regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound B.
  • Figure 5B illustrates changes in the expression of genes from rat C6 flank tumors regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound A.
  • Figure 5C
  • TheVenn diagram illustrated in figure 6A illustrates the degree of overlap between the tumor gene expression responses to KDR kinase inhibitors in C6 flank tumors and MatBIJI mammary tumors.
  • the Venn diagram provided in Panel B illustrates the degree of overlap between the sets of endothelial cell-specific genes regulated both in vitro by mitogens and in tumor tissue by KDR kinase inhibitors. All of the genes (biomakers) depicted in figure 6B are regulated in vivo by KDR kinase inhibitors in a manner opposite that observed in vitro following exposure to mitogens.
  • Figures 7A-7B Confirmation of microarray data by real time quantitative real time PCR.
  • FIG. 7A illustrates fold changes in gene expression in tumors from KDR kinase-treated animals relative tumors from vehicle-treated animals were calculated using the ⁇ CT method (see Materials and Methods).
  • Figure 7B illustrates mRNA levels for each gene in the rat tumors relative the calibrator RNA pool.
  • Figure 8. Biomarker protein expression in rat mammary tumors is localized to vasculature.
  • the present invention provides compositions and methods to detect the level of expression of genes that may be differentially expressed dependent upon the state of the cell, i.e., proliferating versus quiescent cells.
  • detecting the level expression includes methods that quantify expression levels as well as methods that determine whether a gene of interest is expressed at all.
  • an assay which provides a yes or no result without necessarily providing quantification of an amount of expression is an assay that requires "detecting the level of expression" as that phrase is used herein.
  • genes identified as being differentially expressed in proliferating endothelial cells may be used in a variety of nucleic acid detection assays to detect or quantify the expression level of a gene or multiple genes in a given sample. For example, traditional Northern blotting, nuclease protection, RT- PCR and differential display methods may be used for detecting gene expression levels.
  • oligonucleotide sequences that are complementary to one or more of the genes described herein refers to oligonucleotides that are capable of hybridizing under stringent conditions to at least part of the nucleotide sequence of said genes.
  • hybridizable oligonucleotides will typically exhibit at least about 75% sequence identity at the nucleotide level to said genes, preferably about 80% or 85% sequence identity or more preferably about 90% or 95% or more sequence identity to said genes.
  • “Bind(s) substantially” refers to complementary hybridization between a probe nucleic acid and a target nucleic acid and embraces minor mismatches that can be accommodated by reducing the stringency of the hybridization media to achieve the desired detection of the target polynucleotide sequence.
  • hybridizing specifically to refers to the binding, duplexing or hybridizing of a molecule substantially to or only to a particular nucleotide sequence or sequences under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.
  • Assays and methods of the invention may utilize available formats to simultaneously screen at least about 100, preferably about 1000, more preferably about 10,000 and most preferably about 1,000,000 or more different nucleic acid hybridizations. Directly assessing the pharmacodynamics of anti-angiogenesis therapeutics targeted to the VEGF signaling pathway is difficult.
  • Inhibition of the KDR tyrosine protein kinase suppresses endothelial cell proliferation, but it is difficult to assess the rate of proliferation of these cells in vivo.
  • One method that has been used is double immunohistochemical staining of tumor sections for CD31 and Ki67 in order to quantitate proliferating endothelial cells.
  • a second method in use is to assess changes in vascular permeability by magnetic resonance imaging (MRI). Both methods have disadvantages.
  • Immunohistochemistry (MC) is limited to studies where relatively large, intact tumor samples are available. Even then, it is rare to have paired tumor samples taken obtained before and after treatment with a drug candidate for comparison.
  • Fine needle aspiration (FNA) biopsy samples from a clinical setting are not analyzable by this method.
  • Changes in the expression profile from a baseline profile can be used as an indication of such effects.
  • Those skilled in the art can use any of a variety of known techniques to evaluate the expression of one or more of the genes and/or ESTs identified in the instant application in order to observe changes in the expression profile. Beginning with a large set of genes shown to be regulated in vitro by mitogen-induced proliferation of primary endothelial cells, we identified a subset that was relatively specific to endothelial cells. We then identified the subset of genes from the in vitro proliferation signature that were endothelial cell-specific.
  • Two distinct syngeneic tumor models are used to demonstrate that in vivo exposure to KDR kinase inhibitors mediated robust gene expression changes in a manner consistent with suppression of the proliferative rate of vascular endothelial cells within tumors.
  • Gene expression changes consistent with inhibition of VEGF-signaling and inhibition of endothelial cell proliferation were detected in tumors from each animal model.
  • the endothelial cell specificity of the putative biomarkers was confirmed by immunofluorescence microscopy.
  • the biomakers were further validated by correlating their in vivo gene expression changes to an independent, immunohistochemical measure of endothelial cell proliferation.
  • Genes regulated by systemic exposure to KDR kinase inhibitors in at least two of the three tumor models were selected as endothelial cell proliferation biomarkers. Gene expression changes of these biomarkers (as determined by microarray hybridization) were confirmed by quantitative real time PCR, both in the tumors that were profiled as well as in tumors from an additional, independent animal tumor study. The disclosed set of biomarkers was validated by correlating the compound-induced gene expression changes to compound-induced differences in proliferating tumor endothelial cell number as determined by immunohistochemical staining (again in the same rat tumors that were profiled).
  • the endothelial cell specificity (in the context of our rat tumor models) of the biomarker expression (gene signature) disclosed and claimed herein expression is established by showing that the protein products of the identified genes are restricted to CD31 -expressing cells. Based on the disclosure provided herein it is contemplated that it may be possible to identify a gene expression signature that reflects the proliferation rate of vascular endothelial cells within tumors, thereby allowing a clinician to predict tumor responsiveness to therapy.
  • the instant invention provides a set of genes or biomarkers, collectively referred to herein as a gene signature, that are regulated both in vitro during mitogen-induced proliferation of primary microvascular endothelial cells and in vivo in response to systemic exposure to KDR kinase inhibitors. Changes in expression levels of these biomarkers in response to inhibition of KDR are indicators of change in tumor endothelial cell proliferation rate. It is contemplated that identification of the gene expression signature (or biomarkers) disclosed and claimed herein, the regulation of which indicative of changes in the proliferation rate of tumor vascular endothelial cells provides a non-invasive and inexpensive assay following exposure to anti-angiogenesis therapeutics.
  • the angiopoietin-2 protein (ANGPT2/ANG2) is a well characterized ligand for the Tie-2 receptor tyrosine kinase that functions in concert with VEGF and angiopoietin-1 to regulate vascular remodeling (25).
  • Angiopoietin-2 gene expression has been previously reported to be directly upregulated by VEGF, both in vivo and in vitro, consistent with our results (26).
  • the type B endothelin receptor (EDNRB/ET(B)) is a seven transmembrane G-protein coupled receptor that is mutated in Waardenburg-Hirschsprung disease, a congenital malformation of neuronal ganglia in the hindgut (27).
  • EDNRB is an alphal, 3-fucosyltransferase involved in the synthesis of myeloglycan, the major physiological binder of E-selectin (31).
  • Clusterin is a secreted glycoprotein that appears to be overexpressed in apoptotic cells (33-35) but whose function is still largely unknown (33). Clusterin expression has been shown to be anti- proliferative (36) and down-regulated in advanced prostate cancer (37-39). Reduction in serum clusterin levels also correlates with esophageal squamous cell carcinoma tumorigenesis (40). Cysteine rich protein 61 (CYR61) is an extracellular matrix-associated heparin-binding protein with pro-angiogenic properties (41).
  • the urokinase type-plasminogen activator (PLAU or uPA) is a proteolytic enzyme that plays a critical role in angiogenesis, tumor invasion, and metastasis by contributing to remodeling of the extracellular matrix (49, 50). It has been characterized as a pro-tumor invasion and pro-metastatic factor.
  • the effect of PLAU activity is the conversion of plasminogen to plasmin.
  • Cyr ⁇ l it is unclear why we observe an increase in Plau gene expression in tumors exposed to KDR kinase inhibitors rather than the decrease we would have expected to accompany a decrease in neovascularization.
  • Plau expression is a compensatory mechanism elicited by inhibition of the VEGF signaling pathway, but clearly, more investigation is required to determine the mechanism underlying our observations.
  • Ifit3 interferon-induced protein with tetratricopeptide repeats 3, also known as Garg-49 (glucocorticoid-attenuated response genes) and ERG2 (interferon responsive gene 2) is a gene that as yet has no known function. Cloned from the mouse as part of studies to identify glucocorticoid attenuated response genes induced by lipopolysaccharide or interferon, the highly conserved tetratricopeptide repeat domains of IFIT3 are believed to mediate protein-protein interactions (51-54).
  • Ift4 is 60% identical and 78% similar by protein sequence (BLASTP, (55))
  • BLASTP protein sequence
  • RNA for real time PCR can be isolated from low milligram quantities of sample tissue. Quantitative thermal cyclers may now be used with microfluidics cards preloaded with reagents making routine clinical use of multigene expression-based assays a realistic goal.
  • alternative assay formats may be used to monitor the ability of putative cancer therapeutic agent to modulate the expression of a gene identified in Tables 3-6.
  • mRNA expression may be monitored directly by hybridization of probes to the nucleic acids of the invention.
  • methods and assays of the invention are most efficiently designed with array or chip hybridization-based methods for detecting the expression of a large number of genes.
  • Any hybridization assay format may be used, including solution-based and solid support-based assay formats.
  • a preferred solid support is a high density array also known as a DNA chip or a gene chip.
  • gene chips containing probes to at least two genes from Tables 5-6 may be used to directly monitor or detect changes in gene expression in biological samples containing endothelial cells prepared from subjects exposed to putative cancer therapeutics designed to regulate the proliferation of endothelial cells in tumor vasculature.
  • Solid supports containing oligonucleotide probes for differentially expressed genes can be any solid or semisolid support material known to those skilled in the art. Suitable examples include, but are not limited to, membranes, filters, tissue culture dishes, polyvinyl chloride dishes, beads, test strips, silicon or glass based chips and the like. Suitable glass wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755).
  • a preferred solid support is a high density array or DNA chip. These contain a particular oligonucleotide probe in a predetermined location on the array. Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence. Such predetermined locations are termed features. There may be, for example, from 2, 10, 100, 1000 to 10,000, 100,000 or 400,000 of such features on a single solid support.
  • Oligonucleotide probe arrays for expression monitoring can be made and used according to any techniques known in the art (see for example, Lockhart et al., Nat. Biotechnol. (1996) 14, 1675-1680; McGall et al., Proc. Nat. Acad. Sci. USA (1996) 93, 13555-13460).
  • Such probe arrays may contain at least two or more oligonucleotides that are complementary to or hybridize to two or more of the genes described herein.
  • Such arrays my also contain oligonucleotides that are complementary or hybridize to at least 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 70 or more the genes described herein.
  • Any hybridization assay format may be used, including solution-based and solid support-based assay formats.
  • Solid supports containing oligonucleotide probes for differentially expressed genes of the invention can be filters, polyvinyl chloride dishes, silicon or glass based chips, etc. Such wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755). Any solid surface to which oligonucleotides can be bound, either directly or indirectly, either covalently or non-covalently, can be used.
  • a preferred solid support is a high density array or DNA chip.
  • Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence.
  • Such predetermined locations are termed features. There may be, for example, about 2, 10, 100, 1000 to 10,000; 100,000 or 400,000 of such features on a single solid support. The solid support, or the area within which the probes are attached may be on the order of a square centimeter.
  • RT- PCR which can be used to compare mRNA levels in different sample populations, in normal and tumor tissues, with or without drug treatment, to characterize patterns of gene expression, to discriminate between closely related mRNAs, and to analyze RNA structure.
  • the first step is the isolation of mRNA from a target sample.
  • the starting material is typically total RNA isolated from human tumors or tumor cell lines, and corresponding normal tissues or cell lines, respectively.
  • RNA can be isolated from a variety of primary tumors, including breast, lung, colon, prostate, brain, liver, kidney, pancreas, spleen, thymus, testis, ovary, uterus, etc., tumor, or tumor cell lines, with pooled DNA from healthy donors.
  • mRNA can be extracted, for example, from frozen or archived paraffin-embedded and fixed (e.g. formalin-fixed) tissue samples.
  • the first step in gene expression profiling by RT- PCR is the reverse transcription of the RNA template into cDNA, followed by its exponential amplification in a PCR reaction.
  • the two most commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT).
  • AMV-RT avilo myeloblastosis virus reverse transcriptase
  • MMLV-RT Moloney murine leukemia virus reverse transcriptase
  • the reverse transcription step is typically primed using specific primers, random hexamers, or oligo-dT primers, depending on the circumstances and the goal of expression profiling.
  • extracted RNA can be reverse-transcribed using a GeneAmp RNA PCR kit
  • RNAs most frequently used to normalize patterns of gene expression are mRNAs for the housekeeping gene glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), as used herein, or the ⁇ -actin gene.
  • GPDH housekeeping gene
  • the PCR step can use a variety of thermostable DNA-dependent DNA polymerases, it typically employs the Taq DNA polymerase, which has a 5'-3' nuclease activity but lacks a 3'-5' proofreading endonuclease activity.
  • TaqMan.RTM PCR typically utilizes the 5'-nuclease activity of Taq or Tth polymerase to hydrolyze a hybridization probe bound to its target amplicon, but any enzyme with equivalent 5' nuclease activity can be used.
  • Two oligonucleotide primers are used to generate an amplicon typical of a PCR reaction.
  • a third oligonucleotide, or probe is designed to detect nucleotide sequence located between the two PCR primers.
  • the probe is non-extendible by Taq DNA polymerase enzyme, and is labeled with a reporter fluorescent dye and a quencher fluorescent dye. Any laser-induced emission from the reporter dye is quenched by the quenching dye when the two dyes are located close together as they are on the probe.
  • the Taq DNA polymerase enzyme cleaves the probe in a template-dependent manner. The resultant probe fragments disassociate in solution, and signal from the released reporter dye is free from the quenching effect of the second fluorophore.
  • RT-PCR can be performed using commercially available equipment, such as, for example, ABI PRISM 7700.TM. Sequence Detection System. TM. (Perkin-Elmer-Applied Biosystems, Foster City, Calif, USA), or Lightcycler (Roche Molecular Biochemicals, Mannheim, Germany).
  • ABI PRISM 7700.TM. Sequence Detection System. TM. Perkin-Elmer-Applied Biosystems, Foster City, Calif, USA
  • Lightcycler Roche Molecular Biochemicals, Mannheim, Germany.
  • the 5' nuclease procedure is run on a real-time quantitative PCR device such as the ABI PRISM 7700.TM. Sequence Detection System.TM..
  • the system consists of a thermocycler, laser, charge-coupled device (CCD), camera and computer.
  • the system amplifies samples in a 96-well format on a thermocycler.
  • laser-induced fluorescent signal is collected in real-time through fiber optics cables for all 96 wells, and detected at the CCD.
  • the system includes software for running the instrument and for analyzing the data.
  • a more recent variation of the RT-PCR technique is the real time quantitative PCR, which measures PCR product accumulation through a dual-labeled fluorigenic probe (i.e., TaqMan.RTM. probe).
  • Real time PCR is compatible both with quantitative competitive PCR, where internal competitor for each target sequence is used for normalization, and with quantitative comparative PCR using a normalization gene contained within the sample, or a housekeeping gene for RT-PCR.
  • the genes which are assayed according to the present invention are typically in the form of mRNA or reverse transcribed mRNA.
  • the genes may be cloned or not and the genes may be amplified or not. The cloning itself does not appear to bias the representation of genes within a population. However, it may be preferable to use polyA+RNA as a source, as it can be used with less processing steps.
  • RNA isolation can be performed using purification kit, buffer set and protease from commercial manufacturers, such as Qiagen, according to the manufacturer's instructions.
  • Other commercially available RNA isolation kits include MasterPure.TM.
  • RNA from tissue samples can be isolated using RNA Stat-60 (Tel-Test).
  • RNA prepared from tumor can be isolated, for example, by cesium chloride density gradient centrifugation.
  • nucleic acid samples used in the methods and assays of the invention may be prepared by any available method or process. Methods of isolating total mRNA are also well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in detail in Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I Theory and Nucleic Acid
  • RNA samples include RNA samples, but also include cDNA synthesized from a mRNA sample isolated from a cell or tissue of interest. Such samples also include DNA amplified from the cDNA, and an RNA transcribed from the amplified DNA.
  • RNA samples may be of any biological tissue or fluid or cells from any organism as well as cells raised in vitro, such as cell lines and tissue culture cells. Frequently the sample will be a "clinical sample” which is a sample derived from a patient.
  • Typical clinical samples include, but are not limited to, sputum, blood, blood-cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and pleural fluid, or cells therefrom.
  • Biological samples may also include sections of tissues, such as frozen sections or formalin fixed sections taken for histological purposes. The following non-limiting examples are presented to better illustrate the invention. Methods and Materials The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, and biochemistry, which are within the skill of the art.
  • HDMVEC human dermal microvascular endothelial cells
  • RVEC rat heart microvascular endothelial cells
  • VEC Technologies Renneslaer, NY
  • endothelial cell monolayers were maintained at 37°C in a 5% C02 humidified atmosphere in tissue culture flasks coated with human fibronectin (Sigma, St. Louis, MO) using complete MCDB-131 media (MCDB-131 supplemented with 10% fetal bovine serum (FBS, Invitrogen, Carlsbad, CA) and the growth factor cocktail ENDOGRO, VEC Technologies).
  • FBS fetal bovine serum
  • ENDOGRO fetal bovine serum
  • cells were harvested by trypsinization between passage 3-6 following initiation of culture from frozen stocks, counted, and seeded in fibronectin-coated tissue culture plates at 75% confluence (1.5 x 106 cells/per plate, 100 mm diameter plates).
  • Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 ⁇ g ml ENDOGRO.
  • the culture media was changed to pre-warmed DMEM supplemented with 10% FBS.
  • For stimulation of cell growth the growth arrest media was replaced with MDCB-131 supplemented with 10% FBS and the appropriate growth factor.
  • Matched control plates that received no supplemental stimulatory growth factor were made for each stimulation condition.
  • the culture media was removed quickly by aspiration, and the cells were lysed in 1.2 ml RLT buffer (guanidine thiocyanate lysis buffer for RNA stabilization and purification, QIAGEN, Valencia, CA).
  • Cell lysates were homogenized in QIAshredders and total RNA was isolated with RNeasy MINI affinity columns (QIAGEN, Valencia, CA).
  • Gene expression profiles from a total of 8 independent VEGF-stimulated cultures, 7 ENDOGRO- stimulated cultures, and 4 bFGF-stimulated cultures were determined for HDMVECs. Profiles from 4 independent VEGF-stimulated cultures, 4 ENDOGRO-stimulated cultures, and 4 bFGF-stimulated cultures were determined For RHMVECs.
  • a rat glial cell line (C6, ATCC CCL-107) and a rat mammary adenocarcinoma (MatBni, ATCC CRL-1666) were used for our animal tumor models.
  • C6 cells were maintained in culture at 37°C in a 5% C02 humidified atmosphere in Ham's F-12 medium supplemented with 2 mM L- glutamine, 1 mg/ml sodium bicarbonate, 15% horse serum, 2.5% fetal bovine serum, 10 U/ml penicillin, and 10 ⁇ g/ml streptomycin (all media components from Invitrogen).
  • MatBffl cells were grown in McCoy's 5a medium supplemented with 1.5 mM glutamine, 10% FBS 10 U/ml penicillin, and 10 ⁇ g/ml streptomycin.
  • McCoy's 5a medium supplemented with 1.5 mM glutamine, 10% FBS 10 U/ml penicillin, and 10 ⁇ g/ml streptomycin.
  • RNA isolation from C6 or MatBIII cells 2x106 cells growing in a 100 mm diameter tissue culture plate were lysed directly in 1.2 ml RLT buffer. Following lysate homogenization with a QIAshredder, total RNA was isolated with RNeasy MINI affinity columns.
  • C6 glial cells and MatBHI adenocarcinoma cells were chosen for animal models because they were obtained from Fischer 344 (F344) rats and therefore could be used to create syngeneic tumors in immunocompetent F344 animals.
  • C6 glioma flank tumor model C6 cells were injected subcutaneously into the right flank of male F344 rats (150-175 g, 107 cells per animal). Following cell injection, animals were randomized according to body weight to receive either vehicle (0.5% methylcellulose) or drug (10 mpk/dose Compound B or 40 mpk/dose Compound A in 0.5% methylcellulose) (12-15).
  • MatBi ⁇ Breast Cancer Metastasis Model MatBIII cells between passage 20-30 were injected on the mammary fat pad around the 4th left nipple (106 cells/animal) of female F344 rats (150-175 g). Prior to dosing, animals were randomized into groups according to tumor size and body weight. Once daily oral dosing of Compound A (40 mpk dose formulated in 0.5% methylcellulose) or vehicle (0.5% methylcellulose) began on day 7 post tumor cell implantation and continued for 4 additional days. Six vehicle-treated and six compound-treated animals were sacrificed on day 11, four hrs after final dosing. At the time of necropsy, tumors were weighed immediately upon removal.
  • RNA samples from the compound-treated rats were combined to form a control RNA pool.
  • RNAs isolated from each of the tumor samples from the compound-treated rats was compared to the control pool of RNA during microarray hybridization.
  • RNAs from individual vehicle treated animals was compared to the vehicle treated pool in order to assess inter-animal variability.
  • RNA isolated from cultured cells or tumor tissue samples was used to make fluorescently-labeled complementary RNA (cRNA) that was hybridized to DNA microarrays as previously described (16, 17). Briefly, 4 ⁇ g total RNA from an individual tumor sample or in vitro endothelial cell culture was used to synthesize double-stranded DNA through reverse transcription. cRNA was produced by in vitro transcription and labeled post-synthetically with Cy3 or Cy5. Two populations of labeled cRNA, a reference population and experimental population, were compared to each other by competitive hybridization to oligonucleotide arrays synthesized in situ with inkjet technology.
  • cRNA fluorescently-labeled complementary RNA
  • Quantitative real time PCR Quantitative real time polymerase chain reaction was performed with gene-specific PCR primer pairs and amplicon-specific fluorescent probes (TaqMan, Applied Biosystems Inc. (ABI), Foster City, CA) according to published protocols (ABI Assays-on-DemandTM Gene Expression Protocol, Rev A, http://docs.appliedbiosystems.com/pebiodocs/04333458.pdf).
  • One-step quantitative reverse transcription PCR reactions were performed using ABI's TaqMan® One-Step RT-PCR Master Mix Reagents (ABI Product# 4309169) and 25 ng total RNA template on an ABI Prism 7900HT Sequence Detection System.
  • Two-step reverse transcription PCR experiments were initiated by cDNA synthesis from 25 ng total RNA as template using ABI's High Capacity cDNA Archive Kit (ABI Product# 4322171). Second step quantitative real time PCR was performed with standard reagents (TaqMan® Universal PCR Master Mix, ABI Product# 4324018) on the ABI Prism 7900HT Sequence Detection System. Real time PCR reactions were performed in duplicate in a 25 ⁇ l reaction volume in 384-well plates. Primer and probe sequences used for each gene are listed below. For every RNA sample, transcript abundance of GAPDH was determined. In addition, transcript abundance of genes of interest and GAPDH were determined for calibrator RNA samples, either total human lung RNA or total rat lung RNA.
  • the endothelial cell specific signatures defined in Tables 5 and 6 also provide the GenBank Accession Number and GeneSymbol for the biomarkers comprising the expression signature.
  • the following list provides the name, Accession Number and primer and probes used to detect a subset of the biomarkers: Hs refers to Homo sapiens.
  • Rn refers to Rattus norvegicus.
  • Tumor sam pies were fixed immei by submersion in a Zn-Tris fixative solution for immunohistochemistry (MC Zinc Fixative, BD Biosciences-Pharmingen, San Diego, CA) for 24 hr at room temperature (RT, 22°C) followed by submersion in 70% ethanol at RT for an additional 24 hrs. All subsequent steps were performed at RT.
  • Tumor samples were embedded in paraffin (Tissue-Tek VIP Processing/Embedding Medium, Sakura Finetek, Torrance, CA) and cut into 3 ⁇ m sections on a Sakura Accu-Cut SRM microtome (Sakura Finetek).
  • Tissue sections were de-waxed in xylene and re-hydrated through graded ethanol washes. Following washes in deionized H20 (dH20) and tris-buffered saline (TBS), a hydrophobic barrier was placed around the tissue section with a hydrophobic pen (Super Pap Pen, EMS #71310).
  • dH20 deionized H20
  • TBS tris-buffered saline
  • CD31 staining CD31 is a validated endothelial cell-specific protein (18-20). Sections were blocked with Protein Block (Biogenex, San Ramon, CA) for 30 min and incubated with anti-CD31 antibodies (mouse anti-rat, Serotec, Raleigh, NC) diluted 1: 1000 in DAKO Antibody Diluent with Blockers
  • Ki67 Staining Ki67 is a validated nuclear protein expressed only in proliferating cells (21, 22). To facilitate antibody recognition of Ki67, we used a high temperature antigen retrieval strategy. Sections were submerged in Target Retrieval Solution (lx DakoCytomation S1699 diluted with dH20) in a Decloaking Chamber (Biocare Medical, DC2002) and heated to 195°C for 1 min.
  • Sections were cooled with running dH20 into the retrieval solution and then rinsed in TBS. Residual peroxidase activity was blocked by incubating the sections with 3.0% H202 in TBS for 20 min. Sections were washed several times in TBS, then incubated with anti-Ki67 antibodies (rabbit anti-human, Novacastra, Newcastle upon Tyne, UK) diluted 1:2000 in antibody diluent for 2 hrs. Sections were washed with TBST, and then incubated with undiluted biotinylated anti-rabbit IgG (DakoCytomation, Link K-0609) for 10 min.
  • Sections were washed in TBST, and then incubated with streptavidin coupled to horseradish peroxidase (DakoCytomation, K0609) for 10 min. Sections were washed again in TBST, and antibodies bound to Ki67 were visualized by incubation with diaminobenzidine plus substrate (DakoCytomation, DAB+) for 5 min (color development monitored microscopically). Sections were washed in dH20, incubated with DAB Enhance for 20 min RT, and washed again with dH20.
  • Tumor sections were counterstained with filtered Mayer's Hematoxylin (Lillie's Formulation, DakoCytomation) for two min, and then washed with tap H20 until no color remained in the wash water. Sections were then rinsed in dH20, dehydrated with 100% ethanol, cleared with xylenes and mounted with Permount (Fisher Scientific, Hampton, NH).
  • Endothelial cell proliferation percentages represent the combined analysis results from at least 100 images with CD31 staining per tumor section.
  • Immunofluorescence Microscopy Tumor samples were fixed, embedded, sectioned, de-waxed, and re- hydrated as described for immunohistochemistry above. All subsequent steps were performed at room temperature. After a brief rinse in TBS, tissue sections were blocked by incubation with Sniper Blocking Reagent (Biocare Medical) for 5-10 min, rinsed in TBS and incubated with primary antibodies diluted 1: 1000 in DAKO Antibody Diluent for 2 hrs (Antibodies against ANGPT2, CLU, CYR61, and PLAU were from Santa Cruz Biotechnology, Santa Cruz, CA and were raised in goat or rabbit; antibodies against EDNRB were from Calbiochem, San Diego, CA and raised in sheep, antibodies against CD31 were from Serotec and raised in mouse).
  • Sections were then washed with Tris-buffered saline containing 0.2% Tween-20 (TBST, Sigma) and incubated with appropriate secondary antibodies diluted 1:200 (lOug/ml) in DAKO Antibody Diluent with blocking serum for 45 min (Alexa Fluor 488 donkey anti- goat IgG, Alexa Fluor 488 goat anti-rabbit IgG, Alexa Fluor 488 donkey anti-sheep IgG, Molecular Probes, Eugene, OR; Normal donkey and normal goal blocking serum, Sigma). Following additional washes with TBST, sections were counterstained with DAPI (Molecular Probes, 1:2000 dilution of 1 mg/ml stock in MQH20) for 30 min.
  • DAPI Molecular Probes, 1:2000 dilution of 1 mg/ml stock in MQH20
  • Sections were then washed in TBST, dehydrated in 100% EtOH, cleared in xylene, and mounted under coverslips with Permount. Images were captured with a Zeiss Axiocam mHR CCD camera connected to a Zeiss Axiovert 135 inverted fluorescence microscope equipped with a 40x objective. For each fluorophore, all images were captured using equal camera integration times.
  • HDMVECs Primary human dermal microvascular endothelial cells
  • RHMVECs rat heart microvascular endothelial cells
  • HDMVECs or RHMVECs were trypsinization following the third passage in culture and seeded in fibronectin-coated, six-well tissue culture plates at a density of 10,000 cells well.
  • Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 ⁇ g/ml ENDOGRO.
  • VEGF vascular endothelial growth factor
  • bFGF a bFGF-rich bovine brain extract, VEC Technologies
  • ENDOGRO a bFGF-rich bovine brain extract, VEC Technologies
  • the data provided in Figure 1 demonstrates that exposure of HDMVEC (Figure 1A) and RHMVEC (Figure IB) to Compound A selectively inhibits in vitro microvascular endothelial cell proliferation induced by VEGF as determined by viable cell counting with a hemocytometer.
  • the gene expression profile illustrated in Figure 2 graphically illustrates a gene expression signature that is characteristic of proliferating HDMVEC (panel A) and RHMVEC (panel B) cultures. Color intensity represents the degree of regulation, not mRNA copy number.
  • Table 3 provides a list of genes comprising the HDMVEC Proliferative Signature identified in the expression profiles illustrated in Figure 2.
  • Table 4 provides a list of genes comprising the RHMVEC Proliferative Signature identified in the expression profiles illustrated in Figure 2.
  • VEGF binds to and activates the fms-like tyrosine kinase (FLT1) and KDR (23, 24). Both FLTl and KDR are inhibited by Compound B (Table 1). bFGF binds to FGFR1 and FGFR2, but not FLTl or KDR. Both FGFR1 and FGFR2 are relatively insensitive to Compound B (see Table 1). Briefly, EC monolayers were maintained in complete MCDB-131 media until reaching -75% confluence, then induced into a quiescent state by mitogen starvation for 24 hr. Cells were then stimulated to proliferate with 100 ng/ml VEGF for 24 hr in the presence or absence of Compound B.
  • RNA populations isolated from cells exposed to VEGF or VEGF + Compound B were compared to matched control RNAs isolated from quiescent cells exposed to neither VEGF nor Compound B.
  • Parallel experiments were performed with RHMVECs (data not shown). Each point in the plots represents a gene sequence present on the DNA oligonucleotide microarray and is plotted according to the ratio of the two mRNA levels (experimental sample intensity ontrol sample intensity, vertical-axis) and the total mRNA quantity (experimental sample intensity + control sample intensity, horizontal-axis) for that gene.
  • EXAMPLE 3 IDENTIFICATION OF AN ENDOTHELIAL CELL-SPECIFIC PROLIFERATION SIGNATURE
  • the experimental data provided above identifies gene expression profiles, or expression signatures specific for proliferating endothelial cells. However, the majority of genes regulated during endothelial cell proliferation will also be expressed in other types of proliferating cells (genes that regulate cell cycle and metabolic processes, for example). Tumors contain a complex mixture of cell types, where approximately 1 in 2000 cells (0.05%) are proliferating endothelial cells (Joanne Antanavage, Rosemary McFall, and Ken Thomas, personal communications).
  • Candidate endothelial cell-specific genes were defined as genes characterized by regulated expression during an in vitro proliferative response to mitogens, but expressed at relatively low levels in non-endothelial cells.
  • microarray intensity data which corresponds to the number of labeled cRNAs bound to each array feature and is proportional to mRNA copy number, from previous expression profiling studies and compared it with the microarray intensity data from our HDMVEC proliferation experiments.
  • the tumor models use C6 glioma and MatBIII mammary carcinoma cell lines, both derived from Fischer 344 rats. These cell lines each secrete VEGF and form highly vascularized tumors that are sensitive to KDR kinase inhibitors.
  • Glioma Flank Tumor Model C6 cells were injected subcutaneously into the right flank of rats and allowed to form tumors for seven days. At that time, once-daily oral dosing with Compound A,
  • Figure 4 illustrates the growth kinetics of established rat tumors following exposure to a KDR kinase inhibitor. Tumor volumes were determined by caliper measurements. Tumors were calipered in two dimensions (length and width) and tumor volume was calculated according to the formula (length) x (width) x ( l ⁇ width). Genome-wide gene expression in tumors isolated from compound-treated animals was compared to gene expression from tumors isolated from vehicle-treated animals. In the data provided in Figure 5 each row represents a distinct tumor from an individual animal. Each column represents a gene.
  • Points corresponding to genes which are regulated (upregulated or downregulated) are indicated by various shades of gray.
  • the data presented in Panel A of Figure 5 identifies genes from rat C6 flank tumors that are regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound B.
  • the data presented in Panel B of Figure 5 identifies genes from rat C6 flank tumors regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound A.
  • MatBIII breast Cancer Metastasis Model MatBIII mammary adenocarcinoma cells were injected into a mammary fat pad of female rats. After allowing tumors to establish for seven days once-daily oral dosing of Compound A began and continued for a total of 5 days (Figure 4B).
  • the data presented in Panel C of Figure 5 identifies genes from rat MatBIII mammary tumors regulated following 100 hrs of systemic exposure to the KDR kinase inhibitor Compound A.
  • Figure 5 Panel C, p-value ⁇ 0.05 for individual sequences. While there was overlap in the gene expression changes induced by the KDR kinase inhibition between the three studies, the majority of gene expression changes were study-specific (data not shown).
  • FIG. 6 provides Venn diagrams which summarize the degree of overlap between the set of genes identified in the various assays formats disclosed herein. More specifically, Figure 6A summarizes the degree of overlap between the tumor gene expression responses to KDR kinase inhibitors in C6 flank tumors and MatBIII mammary tumors.
  • Figure 6B indicates the degree of overlap between the sets of endothelial cell-specific genes determined to be regulated both in vitro by mitogens and in tumor tissue by KDR kinase inhibitors. All genes/sequences represented in Panel B were observed to be regulated in vivo by KDR kinase inhibitors in a manner opposite that observed in vitro following exposure to mitogens. Most interestingly, we found in each study that some of those genes were regulated in a manner consistent with suppression of endothelial cell proliferation. In effect, these genes were oppositely regulated in our in vitro proliferation experiments as compared to our in vivo tumor studies.
  • Tables 5 and 6 provide a list of genes which were observed to be regulated by Compound A.
  • Table 5 utilizes the data obtained in the C6 Flank Tumor and MatBill Breast Cancer Metatesis Models to define a Compound induced endothelial cell-specific expression signatures.
  • the table provides the GeneBank Accession number, the gene symbol and summarizes the compound-induced fold change in gene expression that was observed.
  • Table 5 provides a summary of the changes (i.e., suppressed expression) observed for individual biomarkers comprising the EC- specific proliferation signatures disclosed herein in response to in vivo KDR inhibitor administration.
  • NM_002421 MMP1 18.54 2.29 VEGF
  • NM_003812 ADAM23 18.65 -1.34 VEGF
  • NM_005460 SNCAIP 5.44 2.20 VEGF
  • NM_014029 HSPC022 32.99 1.25 ENDOGRO NM_014059 RGC32 28.92 -1.65 ENDOGRO NM_014074 PRO0529 4.39 -1.42 VEGF NM_014143 B7-H1 9.24 -1.30 VEGF NM_014331 SLC7A11 16.15 -1.61 ENDOGRO NM_014344 FJX1 7.64 1.39 ENDOGRO NM_014349 APOL3 11.47 -1.43 bFGF NM_014363 SACS 13.77 -1.44 VEGFE NM_014391 CARP 46.40 -1.80 ENDOGRO NM_014397 NEK6 3.95 -1.28 ENDOGRO NM_014398 LAMP3 15.90 -1.35 ENDOGRO NM_014465 ST1 B2 39.94 -1.84 ENDOGRO NM_014476 ALP 15.82 -1.19 ENDOGRO NM_01

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

Methods, biomarkers and expression signatures are disclosed for assessing the proliferative rate of vascular endothelial cells. More specifically, the invention provides a set of genes which can be used as biomarkers and gene signatures for evaluating the pharmacodynamic effects of cancer therapies designed to regulate the proliferation of endothelial cells in tumor vasculature. In one aspect the invention provides a method of evaluating the efficacy of a compounds designed to inhibit kinase receptor activity, such as a mammalian KDR receptor activity.

Description

TITLE OF THE INVENTION
METHOD AND BIOMARKERS FOR DETECTING TUMOR ENDOTHELIAL CELL
PROLIFERATION
CROSS-REFERENCE TO RELATED APPLICATIONS The present application claims the benefit U.S. Provisional Application No. 60/556,645, filed March 26, 2004, hereby incorporated by reference herein.
FIELD OF THE INVENTION The field of this invention relates to methods, biomarkers, and expression signatures for assessing the proliferative rate of vascular endothelial cells within tumors. More specifically, the invention provides a set of genes which can be used as biomarkers for evaluating the pharmacodynamic effects of cancer therapies designed to regulate the proliferation of endothelial cells in tumor vasculature. In one aspect the invention provides a method of evaluating the efficacy of a compounds designed to inhibit kinase receptor activity, such as a mammalian KDR receptor activity.
BACKGROUND OF THE INVENTION In the description that follows, the teachings of various scientific references are relied on to support particular findings and statements. The numerical citations included at the end of particular sentences refer to the numbered list of references included at the end of the specification. Vascular endothelial cells form a luminal non-thrombogenic monolayer throughout the vascular system. Solid tumors require a vascular system to expand beyond small nodules limited by the diffusion of nutrients and metabolic by products. Although tumor cells can initially colonize existing host capillaries, their growth leads to the collapse of these preexisting normal vessels resulting in hypoxia. Therefore, angiogenesis is critical to the progression of numerous cancers. Subsequent tumor growth requires neovascularization that is achieved by the ingrowth of new host blood vessels, denoted tumor angiogenesis. Tumors induce proliferation, migration and differentiation resulting in neovascularization by secreting growth factors for vascular endothelial cells. Angiogenesis is critical to the progression of numerous cancers. Tumors induce endothelial cell migration, proliferation and differentiation resulting in neovascularization arising from existing blood vessels. Tumor cells induce angiogenesis primarily through the production and secretion of vascular endothelial growth factor (VEGF), a secreted protein that is a potent endothelial cell mitogen and ligand for the kinase insert domain receptor (KDR, FLK-1, or VEGF receptor). Tyrosine kinases are a class of enzymes that catalyze the transfer of the terminal phosphate of adenosine triphosphate to tyrosine residues in protein substrates. Tyrosine kinases are believed, by way of substrate phosphorylation, to play critical roles in signal transduction for a number of cell functions and have been shown to be important contributing factors in cell proliferation, carcinogenesis and cell differentiation. Tyrosine kinases can be categorized as receptor type or non receptor type. Receptor type tyrosine kinases typically have an extracellular, a transmembrane, and an intracellular portion, while non- receptor type tyrosine kinases typically are wholly intracellular, while examples exist of membrane receptors that upon ligand binding recruit intracellular kinases to bind to the intracellular portion of the receptor which, by itself, does not have kinase activity. Both receptor-type and non-receptor type tyrosine kinases are implicated in cellular signaling pathways leading to numerous pathogenic conditions, including cancer, psoriasis and hyperimmune responses. The receptor-type tyrosine kinases are comprised of a large number of transmembrane receptors with diverse biological activity. In fact, about twenty different subfamilies of receptor-type tyrosine kinases have been identified. The kinase insert domain receptor (KDR) belongs to the FLK subfamily of receptor-type tyrosine kinases. KDR is a transmembrane receptor tyrosine kinase expressed primarily in vascular endothelial cells that transduces the majority of physiological functions attributed to VEGF (3, 8-10). Inhibition of KDR catalytic activity blocks tumor neo-angiogenesis, reduces vascular permeability, and, in animal models, inhibits tumor growth and metastasis. Tumor cells induce angiogenesis primarily through the production and secretion of vascular endothelial growth factor (VEGF) a potent endothelial cell mitogen and ligand for the kinase insert domain receptor (KDR, FLK-1, or VEGF receptor 2) (3-7). VEGF binds with high affinity to two transmembrane tyrosine kinase-linked receptors, Flt-1 (VEGFR-1) and KDR (Flk-l/VEGFR-2), that are expressed by vascular endothelial cells. The binding of dimeric VEGF to the extracellular region of KDR promotes receptor dimerization that brings the intracellular tyrosine kinase domains together and promotes phosphorylation of several receptor tyrosine residues, at least some of which are critical for mitogenic signal transduction. Extensive efforts are underway to identify anti-angiogenic therapies for the treatment of human cancers. Since VEGF produced and secreted by tumor cells activates KDR and induces endothelial cell proliferation, inhibition of KDR by a small molecule should lead to a decrease in the proliferation rate of tumor endothelial cells. Currently, several small molecule inhibitors of KDR activity are being evaluated as anti-cancer agents in clinical trials. Once activated, KDR initiates a signal transduction cascade, is internalized and ultimately degraded. Inhibition of the VEGF/KDR system has been shown to inhibit VEGF-dependent tumor angiogenesis and growth in several animal models. Because VEGF produced and secreted by tumor cells activates KDR and induces endothelial cell proliferation, it is acknowledged that inhibition of KDR kinase activity should lead to decreases in the proliferation of tumor endothelial cells. Accordingly, numerous proposed cancer therapeutics target vascular endothelial cell growth factor (VEGF) or the kinase insert domain receptor (KDR/VEGFR-2/FLK-l), the primary VEGF receptor on endothelial cells. Clinically, it is difficult to assess directly the pharmacodynamic effects of KDR inhibitors because KDR protein is not easily detectable in readily available clinical samples. Measurement of changes in vascular permeability within a tumor by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) is currently the most common pharmacodynamic assay, but provides an indirect readout and is a complex and expensive procedure. Thus, there is a need in the clinical setting for a rapid, quantitative, reproducible, and inexpensive assay that is compatible with current clinical laboratory instrumentation and which is capable of assessing the efficacy of anti-angiogenic agents.
SUMMARY OF THE INVENTION As the mitogenically and angiogenically competent VEGF receptor, KDR is a particularly attractive target to antagonize VEGF-dependent tumor angiogenesis and growth. Inhibition of KDR catalytic activity blocks tumor neoangiogenesis, reduces vascular permeability, and in animal moldes, inhibits tumor growth and metastasis. However, because KDR protein is not expressed at high levels in readily accessible biological material, such as peripheral blood or bone marrow aspirates, clinical assessment of the in vivo pharmacodynamic efficacy of KDR kinase inhibitors is challenging. Accordingly, current pharmacodynamic assays for KDR inhibition generally rely on surrogate protein kinase markers whose activity is also sensitive to the compound being evaluated (i.e. Fms-related tyrosine kinase-3 (Flt-3) tyrosine phosphorylation in the case of many KDR kinase inhibitors) or on imaging techniques such as DCE-MRI that can assess changes in vascular permeability. These methods have the disadvantage of being indirect measures of KDR function and endothelial cell proliferation. An alternate approach is to assess the pharmacodynamic effects of putative KDR inhibitors on the proliferation rate of tumor endothelial cells. One described method for the in vivo assessment of EC proliferation involves dual immunohistochemical (MC) staining of tumor sections for the endothelial cell marker CD-31 and a nuclear marker of cellular proliferation, Ki-67(11). While an immunohistochemical method such as this can determine the fraction of ECs that are proliferating, the experimental protocol is technically complex and difficult and the analysis required for each stained tumor section is extremely time-consuming. Each of these factors makes clinical use of an MC-based assay unlikely. The methods disclosed and claimed herein are based on the discovery and characterization of biomarkers and gene expression signatures that are specific for proliferating endothelial cells. Gene expression profiling data from cultured primary endothelial cells, cultured tumor cells, and tissue from animal tumor models treated with KDR inhbitors was used to identify a set of genes that are selectively overexpressed in tumor endothelial cells relative to tumor cells, and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation. It is contemplated that the biomarkers and endothelial cell-specific expression signatures which are disclosed and claimed herein will find utility in the context of providing a pharmacodynamic readout for any cancer therapy that aims to inhibit proliferation of endothelial cells in tumor vasculature. As shown herein, the expression levels of these genes serve as the basis of a simple pharmacodynamic assay for the activity of small molecule inhibitors of the KDR receptor tyrosine kinase. The methods disclosed and claimed herein can be used as the basis for a pharmacodynamic assay capable of supporting the clinical development of small molecule inhibitors of the KDR receptor tyrosine kinase. More specifically, the invention provides a method for assessing the in vivo effects of a KDR kinase inhibitor on the proliferative rate of vascular endothelial cells within tumors. In one aspect the invention provides a method for determining the proliferative status (or rate) of endothelial cells. As shown herein, the disclosed method can be used to evaluate the proliferative status of endothelial cells in either an in vitro or in vivo format. One of skill in the art will acknowledge that the disclosed gene expression-based pharmacodynamic assays which can be established based on the disclosure provided herein can be used to support screening assays established to evaluate the efficacy of therapeutic agents intended to regulate the proliferative status of endothelial cells. In a second aspect the invention provides a method for evaluating the proliferative rate of vascular endothelial cells within tumors. In a particular embodiment, the invention provides a gene expression-based pharmacodynamic assay that is suitable for use to support clinical development of cancer therapies designed to regulate the proliferation of endothelial cells in tumor vasculature. For example, it is contemplated that the disclosed methods can be used to establish pharmacodynamic assays that can distinguish tumors containing proliferating endothelial cells from tumors containing mostly quiescent endothelial cells. In a particular embodiment the method may include detecting the expression level of one or more genes selected from a group consisting of Angpt-2, Clu (ApoJ), Cyrόl (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA). In a third aspect the invention provides a method for evaluating the activity of anti-angiogenesis therapeutics intended to regulate the proliferative rate of vascular endothelial cells within tumors. In a particular embodiment, the invention provides a method for evaluating the efficacy of small molecule inhibitors of receptor-type kinase inhibitors, such as KDR. For example, this aspect of the invention provides a method which is suitable for use to support clinical development of KDR kinase inhibitors, such as Compound A. Using the information provided herein, particularly the Compound A-and B- induced endothelial cell-specific expression signatures provided in Tables 5. Table 6 provides the summary information describing the changed (suppressed) expression of endothelial cell specific biomarkers (collectively referred to as the proliferation sequence) observed in response to the in vivo administration of KDR Kinase inhibitors (Compounds A and B), provided in Table 6. It is well within the abilities of a skilled artisan to design and validate a gene expression-based assay that is suitable for evaluating the efficacy of anti-angiogenesis agents. It is contemplated that using the disclosure provided herein a skilled artisan can utilize the information provided in the tables summarizing the proliferation signatures disclosed herein to identify compound-specific expression signatures that will facilitate evaluating the efficacy of alternative therapeutic agents intended to regulate endothelial cell proliferation. For example, this aspect of the invention provides a method which is suitable for use to support clinical development of KDR kinase inhibitors, such as Compound A. For example, it is contemplated that the efficacy of Compound A could be evaluated in vivo by establishing an assay which detects changes in the expression of a gene signature comprising the Angpt-2, Clu (ApoJ), Cyrόl (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA) genes. In a fourth aspect, the invention provides a composition of genes or biomarkers which are selectively overexpressed in tumor endothelial cells relative to tumor cells, and whose pattern of expression correlates with the rate of tumor endothelial cell proliferation. One embodiment of this aspect of the invention provides compositions comprising at least two oligonucleotides, wherein each of the oligonucleotides comprises a sequence that specifically hybridizes to a gene disclosed in Tables 3 or 4 as well as solid supports comprising at least two probes, wherein each of the probes comprises a sequence that specifically hybridizes to a gene in Tables 3, 4, 5 or 6. In a particular embodiment, the composition will comprise oligonucleotides and/or probes which hybridize with the following genes: Angpt-2, Clu (ApoJ), Cyrόl (CCN1), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA), in combination with other oligonucleotides or probes specific for other genes identified in Tables 3-6. In another aspect the invention provides gene expression signatures, which can be used to establish expression-based pharmacodynamic assays for evaluating the efficacy of therapeutic agents designed to regulate the proliferation of endothelial cells. It is contemplated that one of skill in the art will be able to utilize the information provided in this disclosure, in particular the information contained in the Proliferation Signature Tables, referred to herein as Table 3 (HDMVEC Proliferation Signature), Table 4 (RHMVEC Prolilferation Signature), Table 5 (Compound A-and B-induced Endothelial Cell Specific Sequences) and Table 6 (Changes in EC-specific proliferation signature by KDR Kinase Inhibitor Asministration), to elucidate endothelial cell proliferation signatures that are suitable for monintoring the in efficacy of other anti-angiogenic compounds. As shown herein, the gene expression signature disclosed and claimed herein can be used distinguish tumors containing mostly proliferating ECs from tumors containing mostly quiescent ECs. It is contemplated that the disclosed assay will have the ability to detect inhibition of angiogenesis relatively quickly after initiating therapy, eliminating the longer period of time required to visualize morphological changes in tumor microvasculature. In addition, it is envisioned that the disclosed methods will be particularly useful in circumstances where immunohistochemistry is inappropriate or impractical, such as with small tissue samples from biopsies (i.e. fine needle aspirates) or from tissue samples with poor morphology. In a real time quantitative reverse transcription- polymerase chain reaction (PCR) format, the disclosed assay is predicted to represent an extremely sensitive assay that is readily compatible with existing clinical laboratory instrumentation. Expression profiling-based monitoring of the pharmacodynamic effects of cancer therapy potentially has many benefits. Used in the clinical setting, this technology provides for rapid, quantitative, reproducible, and inexpensive assays that are compatible with current clinical laboratory instrumentation. Carefully designed, gene expression-based asssays, such as the assays disclosed herein, have the potential to make dosing of anti-neoplastic agents more efficient, to identify patient populations most likely to benefit from specific therapies, and to reduce clinical development time of novel therapeutics. Each of these aspects will lead to increased tumor response rates and improved human health.
BRIEF DESCRIPTION OF THE DRAWINGS Figures 1A and IB. Inhibition of Vascular Endothelial Cell (VEC) proliferation in vitro by KDR kinase inhibition. HDMVECs or RHMVECs were trypsinization following the third passage in culture and seeded in fibronectin-coated, six-well tissue culture plates at a density of 10,000 cells/well. Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 μg/ml ENDOGRO. Wells with unstimulated cells and wells containing un-arrested cells were included as controls. At 72 hr following growth factor stimulation, cells were removed from the culture plates by trypsinization and counted on a hemocytometer under bright-field microscopy. Figures 2A and 2B. Identification of a gene expression profile in proliferating vascular endothelial cells in vitro. HDMVECs and RHMVECs were grown in culture and mitogen deprived for 24 hr as described in Figure 1 and Methods. Following a 24 hr stimulation with growth factor, culture media was aspirated quickly and the cells lysed in an RNA stabilizing buffer. Matched control plates that received no supplemental stimulatory growth factor were present for each stimulation condition and RNAs isolated from them served as the reference to which the RNAs from the stimulated cells was compared. Bars corresponding to genes which are regulated (e.g., upregulated or downregulated) are indicated by various shades of gray. Color intensity represents the degree of regulation, not mRNA copy number. Figures 3A-3D. Specific suppression of VEGF-induced gene expression in cultured vascular endothelial cells. EC monolayers were maintained in complete MCDB-131 media until reaching -75% confluence, then induced into a quiescent state by mitogen starvation for 24 hr. Cells were then stimulated to proliferate with 100 ng/ml VEGF for 24 hr in the presence or absence of Compound B. RNA populations isolated from cells exposed to VEGF or VEGF + Compound B were compared to matched control RNAs isolated from quiescent cells exposed to neither VEGF nor Compound B. Each point in the plots represents a gene sequence present on the DNA oligonucleotide microarray and is plotted according to the ratio of the two mRNA levels (experimental sample intensity: control sample intensity, vertical-axis) and the total mRNA quantity (experimental sample intensity + control sample intensity, horizontal-axis) for that gene. Dark-colored points indicate upregulated genes. Light Gray colored points indicate downregulated genes. Figures 4A and 4B. Growth kinetics of established rat tumors following exposure to a KDR kinase inhibitor. Tumor studies were performed as described in Materials and Methods. Figures 4A and 4B illustrate tumor volumes from animals in the C6 profiling study (Fig. 4A) and the Mattlll profiling study (Fig. 4B) as determined by caliper measurements. Tumors were calipered in two dimensions (length and width) and tumor volume was calculated according to the formula (length) x (width) x (V2 width). Figures 5 A-5C. Identification of gene expression changes induced in rat tumors by KDR kinase inhibitors in vivo.Each row represents a distinct tumor from an individual animal. Each column represents a gene. Gray colored points/bars indicate genes that are regulated (e.g., upregulated or downregulated) by KDR kinase. Figure 5A illustrates changes in the expression of genes from rat C6 flank tumors that are regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound B. Figure 5B illustrates changes in the expression of genes from rat C6 flank tumors regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound A. Figure 5C illustrates changes in the expression of genes from rat MatBHI mammary tumors regulated following 100 hrs of systemic exposure to the KDR kinase inhibitor Compound A. Figures 6A-6B. Distinct tumor gene expression responses elicited by KDR inhibitors.TheVenn diagram illustrated in figure 6A illustrates the degree of overlap between the tumor gene expression responses to KDR kinase inhibitors in C6 flank tumors and MatBIJI mammary tumors. The Venn diagram provided in Panel B illustrates the degree of overlap between the sets of endothelial cell-specific genes regulated both in vitro by mitogens and in tumor tissue by KDR kinase inhibitors. All of the genes (biomakers) depicted in figure 6B are regulated in vivo by KDR kinase inhibitors in a manner opposite that observed in vitro following exposure to mitogens. Figures 7A-7B. Confirmation of microarray data by real time quantitative real time PCR. Quantitative real time PCR was performed with gene-specific PCR primer pairs and amplicon-specific fluorescent probes (TaqMan). For each RNA sample tested, transcript abundance of GAPDH was determined. In addition, transcript abundance of genes of interest and GAPDH were determined for a calibrator RNA sample (total rat lung RNA). Figure 7A illustrates fold changes in gene expression in tumors from KDR kinase-treated animals relative tumors from vehicle-treated animals were calculated using the ΔΔCT method (see Materials and Methods). Figure 7B illustrates mRNA levels for each gene in the rat tumors relative the calibrator RNA pool. Figure 8. Biomarker protein expression in rat mammary tumors is localized to vasculature. De- waxed, re-hydrated MatBiπ tumor sections were incubated with antibodies against CD31 and one of the following biomarker proteins: CLU, ANGPT2, CYR61, ENDRB, or PLAU. Primary antibodies bound to the biomarker proteins and CD31 were visualized with Alexa488-labeled and Alexa546-labeled secondary antibodies, respectively as described in Materials and Methods. After mounting under coverslips, images were captured with a Zeiss Axiocam through a Zeiss Axiovert 135 fluorescence microscope equipped with a 40x objective and an Axiocam mHR CCD camera.
DETAILED DESCRIPTION OF THE INVENTION In the description that follows, numerous terms and phrases known to those skilled in the art are used. In the interest of clarity and consistency of interpretation, the definitions of certain terms and phrases are provided. The present invention provides compositions and methods to detect the level of expression of genes that may be differentially expressed dependent upon the state of the cell, i.e., proliferating versus quiescent cells. As used herein, the phrase "detecting the level expression" includes methods that quantify expression levels as well as methods that determine whether a gene of interest is expressed at all. Thus, an assay which provides a yes or no result without necessarily providing quantification of an amount of expression is an assay that requires "detecting the level of expression" as that phrase is used herein. The genes identified as being differentially expressed in proliferating endothelial cells may be used in a variety of nucleic acid detection assays to detect or quantify the expression level of a gene or multiple genes in a given sample. For example, traditional Northern blotting, nuclease protection, RT- PCR and differential display methods may be used for detecting gene expression levels. As used herein, oligonucleotide sequences that are complementary to one or more of the genes described herein, refers to oligonucleotides that are capable of hybridizing under stringent conditions to at least part of the nucleotide sequence of said genes. Such hybridizable oligonucleotides will typically exhibit at least about 75% sequence identity at the nucleotide level to said genes, preferably about 80% or 85% sequence identity or more preferably about 90% or 95% or more sequence identity to said genes. "Bind(s) substantially" refers to complementary hybridization between a probe nucleic acid and a target nucleic acid and embraces minor mismatches that can be accommodated by reducing the stringency of the hybridization media to achieve the desired detection of the target polynucleotide sequence. The phrase "hybridizing specifically to" refers to the binding, duplexing or hybridizing of a molecule substantially to or only to a particular nucleotide sequence or sequences under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA. Assays and methods of the invention may utilize available formats to simultaneously screen at least about 100, preferably about 1000, more preferably about 10,000 and most preferably about 1,000,000 or more different nucleic acid hybridizations. Directly assessing the pharmacodynamics of anti-angiogenesis therapeutics targeted to the VEGF signaling pathway is difficult. Inhibition of the KDR tyrosine protein kinase suppresses endothelial cell proliferation, but it is difficult to assess the rate of proliferation of these cells in vivo. One method that has been used is double immunohistochemical staining of tumor sections for CD31 and Ki67 in order to quantitate proliferating endothelial cells. A second method in use is to assess changes in vascular permeability by magnetic resonance imaging (MRI). Both methods have disadvantages. Immunohistochemistry (MC) is limited to studies where relatively large, intact tumor samples are available. Even then, it is rare to have paired tumor samples taken obtained before and after treatment with a drug candidate for comparison. Fine needle aspiration (FNA) biopsy samples from a clinical setting are not analyzable by this method. Furthermore, to accurately assess microvascular density or the percentage of proliferating endothelial cells throughout the tumor is labor intensive even with semi-automated or automated microscopy equipment. Analyzing MRI images to assess changes in vascular permeability is also labor intensive, requiring highly trained personnel both to operate the imager and to interpret the images. Our strategy in designing a gene expression-based pharmacodynamic assay for endothelial cell proliferation was to employ genome-wide gene expression profiling to first identify a general mitogen- induced proliferation signature in cultured primary microvascular endothelial cells. Expression profiles of genes in particular, tissues, disease states or disease progression stages provide molecular tools for evaluating toxicity, drug efficacy, drug metabolism, development, and disease monitoring. Changes in the expression profile from a baseline profile can be used as an indication of such effects. Those skilled in the art can use any of a variety of known techniques to evaluate the expression of one or more of the genes and/or ESTs identified in the instant application in order to observe changes in the expression profile. Beginning with a large set of genes shown to be regulated in vitro by mitogen-induced proliferation of primary endothelial cells, we identified a subset that was relatively specific to endothelial cells. We then identified the subset of genes from the in vitro proliferation signature that were endothelial cell-specific. Two distinct syngeneic tumor models are used to demonstrate that in vivo exposure to KDR kinase inhibitors mediated robust gene expression changes in a manner consistent with suppression of the proliferative rate of vascular endothelial cells within tumors. Gene expression changes consistent with inhibition of VEGF-signaling and inhibition of endothelial cell proliferation were detected in tumors from each animal model. The endothelial cell specificity of the putative biomarkers was confirmed by immunofluorescence microscopy. The biomakers were further validated by correlating their in vivo gene expression changes to an independent, immunohistochemical measure of endothelial cell proliferation. Genes regulated by systemic exposure to KDR kinase inhibitors in at least two of the three tumor models were selected as endothelial cell proliferation biomarkers. Gene expression changes of these biomarkers (as determined by microarray hybridization) were confirmed by quantitative real time PCR, both in the tumors that were profiled as well as in tumors from an additional, independent animal tumor study. The disclosed set of biomarkers was validated by correlating the compound-induced gene expression changes to compound-induced differences in proliferating tumor endothelial cell number as determined by immunohistochemical staining (again in the same rat tumors that were profiled). The endothelial cell specificity (in the context of our rat tumor models) of the biomarker expression (gene signature) disclosed and claimed herein expression is established by showing that the protein products of the identified genes are restricted to CD31 -expressing cells. Based on the disclosure provided herein it is contemplated that it may be possible to identify a gene expression signature that reflects the proliferation rate of vascular endothelial cells within tumors, thereby allowing a clinician to predict tumor responsiveness to therapy. Based on the data described herein, the instant invention provides a set of genes or biomarkers, collectively referred to herein as a gene signature, that are regulated both in vitro during mitogen-induced proliferation of primary microvascular endothelial cells and in vivo in response to systemic exposure to KDR kinase inhibitors. Changes in expression levels of these biomarkers in response to inhibition of KDR are indicators of change in tumor endothelial cell proliferation rate. It is contemplated that identification of the gene expression signature (or biomarkers) disclosed and claimed herein, the regulation of which indicative of changes in the proliferation rate of tumor vascular endothelial cells provides a non-invasive and inexpensive assay following exposure to anti-angiogenesis therapeutics. While the expectation by random chance of identifying a gene that met all our selection criteria was low, we identified a set of seven potential biomarker genes (Angpt-2, Endrb (Etb), Fut-4, Clu (ApoJ), Cyrόl (CCNl), Plau (uPA), and Ifit-3 (Garg49)). Significantly, each of the seven identified biomarker is known or implicated to be involved in endothelial cell biology. We biased our biomarker selection towards endothelial cell specific genes, but there was no guarantee that genes meeting our multiple criteria would have any known function in endothelial cells. Surprisingly, nearly all the genes identified have been implicated or shown to be directly involved in the regulation of endothelial cell function. The angiopoietin-2 protein (ANGPT2/ANG2) is a well characterized ligand for the Tie-2 receptor tyrosine kinase that functions in concert with VEGF and angiopoietin-1 to regulate vascular remodeling (25). Angiopoietin-2 gene expression has been previously reported to be directly upregulated by VEGF, both in vivo and in vitro, consistent with our results (26). The type B endothelin receptor (EDNRB/ET(B)) is a seven transmembrane G-protein coupled receptor that is mutated in Waardenburg-Hirschsprung disease, a congenital malformation of neuronal ganglia in the hindgut (27). Most published studies of EDNRB describe its role in the neuronal system during neural crest development. However, it does control vasoconstriction and vascular cell proliferation induced by the endothelins and EDNRB has been shown to be overexpressed in primary melanomas(28). EDNRB antagonists have been reported to inhibit vascular cell proliferation and human melanoma cell growth in vitro and in vivo (29, 30). Fucosyltransferase 4 (FUT4) is an alphal, 3-fucosyltransferase involved in the synthesis of myeloglycan, the major physiological binder of E-selectin (31). It is also involved in the synthesis of many other glycosylated proteins, but it is reported to be highly expressed in some tumors with inverse correlation to prognosis (32). Clusterin is a secreted glycoprotein that appears to be overexpressed in apoptotic cells (33-35) but whose function is still largely unknown (33). Clusterin expression has been shown to be anti- proliferative (36) and down-regulated in advanced prostate cancer (37-39). Reduction in serum clusterin levels also correlates with esophageal squamous cell carcinoma tumorigenesis (40). Cysteine rich protein 61 (CYR61) is an extracellular matrix-associated heparin-binding protein with pro-angiogenic properties (41). In vitro CYR61, promotes cell adhesion to extracellular matrix and chemotaxis (42, 43). It stimulates cell motility through interaction with integrin αVβ5, αόβl, αMβ2 and stimulates endothelial cell proliferation through interaction with aVb3 (44-48). Our observations that the Cyrόl gene was upregulated in rat tumor tissue following exposure to KDR kinase inhibitors did not appear to be in agreement with an inhibition of endothelial cell proliferation. However it may be that the upregulation of the Cyrόl gene is a response mechanism the endothelial cell attempting to compensate for the lack of functional KDR. The urokinase type-plasminogen activator (PLAU or uPA) is a proteolytic enzyme that plays a critical role in angiogenesis, tumor invasion, and metastasis by contributing to remodeling of the extracellular matrix (49, 50). It has been characterized as a pro-tumor invasion and pro-metastatic factor. The effect of PLAU activity is the conversion of plasminogen to plasmin. As with Cyrόl, it is unclear why we observe an increase in Plau gene expression in tumors exposed to KDR kinase inhibitors rather than the decrease we would have expected to accompany a decrease in neovascularization. We can surmise that increased Plau expression is a compensatory mechanism elicited by inhibition of the VEGF signaling pathway, but clearly, more investigation is required to determine the mechanism underlying our observations. Ifit3 (interferon-induced protein with tetratricopeptide repeats 3, also known as Garg-49 (glucocorticoid-attenuated response genes) and ERG2 (interferon responsive gene 2) is a gene that as yet has no known function. Cloned from the mouse as part of studies to identify glucocorticoid attenuated response genes induced by lipopolysaccharide or interferon, the highly conserved tetratricopeptide repeat domains of IFIT3 are believed to mediate protein-protein interactions (51-54). No human ortholog of Ifit3 has been identified in human cells, but a homologous gene designated Ift4 is 60% identical and 78% similar by protein sequence (BLASTP, (55)) In practice a gene expression-based pharmacodynamic assay based on a small number of genes can be performed with relatively little effort using existing quantitative real time PCR technology familiar to clinical laboratories. Sufficient RNA for real time PCR can be isolated from low milligram quantities of sample tissue. Quantitative thermal cyclers may now be used with microfluidics cards preloaded with reagents making routine clinical use of multigene expression-based assays a realistic goal. It is to be understood that alternative assay formats, other than the methodologies exemplified herein, may be used to monitor the ability of putative cancer therapeutic agent to modulate the expression of a gene identified in Tables 3-6. For instance, as described above, mRNA expression may be monitored directly by hybridization of probes to the nucleic acids of the invention. However, methods and assays of the invention are most efficiently designed with array or chip hybridization-based methods for detecting the expression of a large number of genes. Any hybridization assay format may be used, including solution-based and solid support-based assay formats. A preferred solid support is a high density array also known as a DNA chip or a gene chip. In one assay format, gene chips containing probes to at least two genes from Tables 5-6 may be used to directly monitor or detect changes in gene expression in biological samples containing endothelial cells prepared from subjects exposed to putative cancer therapeutics designed to regulate the proliferation of endothelial cells in tumor vasculature. Solid supports containing oligonucleotide probes for differentially expressed genes can be any solid or semisolid support material known to those skilled in the art. Suitable examples include, but are not limited to, membranes, filters, tissue culture dishes, polyvinyl chloride dishes, beads, test strips, silicon or glass based chips and the like. Suitable glass wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755). Any solid surface to which oligonucleotides can be bound, either directly or indirectly, either covalently or non-covalently, can be used. In some embodiments, it may be desirable to attach some oligonucleotides covalently and others non-covalently to the same solid support. A preferred solid support is a high density array or DNA chip. These contain a particular oligonucleotide probe in a predetermined location on the array. Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence. Such predetermined locations are termed features. There may be, for example, from 2, 10, 100, 1000 to 10,000, 100,000 or 400,000 of such features on a single solid support. The solid support, or the area within which the probes are attached may be on the order of a square centimeter. Oligonucleotide probe arrays for expression monitoring can be made and used according to any techniques known in the art (see for example, Lockhart et al., Nat. Biotechnol. (1996) 14, 1675-1680; McGall et al., Proc. Nat. Acad. Sci. USA (1996) 93, 13555-13460). Such probe arrays may contain at least two or more oligonucleotides that are complementary to or hybridize to two or more of the genes described herein. Such arrays my also contain oligonucleotides that are complementary or hybridize to at least 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 70 or more the genes described herein. Any hybridization assay format may be used, including solution-based and solid support-based assay formats. Solid supports containing oligonucleotide probes for differentially expressed genes of the invention can be filters, polyvinyl chloride dishes, silicon or glass based chips, etc. Such wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755). Any solid surface to which oligonucleotides can be bound, either directly or indirectly, either covalently or non-covalently, can be used. A preferred solid support is a high density array or DNA chip. These contain a particular oligonucleotide probe in a predetermined location on the array. Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence. Such predetermined locations are termed features. There may be, for example, about 2, 10, 100, 1000 to 10,000; 100,000 or 400,000 of such features on a single solid support. The solid support, or the area within which the probes are attached may be on the order of a square centimeter. Of the techniques listed above, the most sensitive and most flexible, quantitative method is RT- PCR, which can be used to compare mRNA levels in different sample populations, in normal and tumor tissues, with or without drug treatment, to characterize patterns of gene expression, to discriminate between closely related mRNAs, and to analyze RNA structure. The first step is the isolation of mRNA from a target sample. The starting material is typically total RNA isolated from human tumors or tumor cell lines, and corresponding normal tissues or cell lines, respectively. Thus RNA can be isolated from a variety of primary tumors, including breast, lung, colon, prostate, brain, liver, kidney, pancreas, spleen, thymus, testis, ovary, uterus, etc., tumor, or tumor cell lines, with pooled DNA from healthy donors. If the source of mRNA is a primary tumor, mRNA can be extracted, for example, from frozen or archived paraffin-embedded and fixed (e.g. formalin-fixed) tissue samples. As RNA cannot serve as a template for PCR, the first step in gene expression profiling by RT- PCR is the reverse transcription of the RNA template into cDNA, followed by its exponential amplification in a PCR reaction. The two most commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT). The reverse transcription step is typically primed using specific primers, random hexamers, or oligo-dT primers, depending on the circumstances and the goal of expression profiling. For example, extracted RNA can be reverse-transcribed using a GeneAmp RNA PCR kit
(Perkin Elmer, California, USA), following the manufacturer's instructions. The derived cDNA can then be used as a template in the subsequent PCR reaction. To minimize errors and the effect of sample-to- sample variation, RT-PCR is usually performed using an internal standard. The ideal internal standard is expressed at a constant level among different tissues, and is unaffected by the experimental treatment. RNAs most frequently used to normalize patterns of gene expression are mRNAs for the housekeeping gene glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), as used herein, or the β-actin gene. Although the PCR step can use a variety of thermostable DNA-dependent DNA polymerases, it typically employs the Taq DNA polymerase, which has a 5'-3' nuclease activity but lacks a 3'-5' proofreading endonuclease activity. Thus, TaqMan.RTM. PCR typically utilizes the 5'-nuclease activity of Taq or Tth polymerase to hydrolyze a hybridization probe bound to its target amplicon, but any enzyme with equivalent 5' nuclease activity can be used. Two oligonucleotide primers are used to generate an amplicon typical of a PCR reaction. A third oligonucleotide, or probe, is designed to detect nucleotide sequence located between the two PCR primers. The probe is non-extendible by Taq DNA polymerase enzyme, and is labeled with a reporter fluorescent dye and a quencher fluorescent dye. Any laser-induced emission from the reporter dye is quenched by the quenching dye when the two dyes are located close together as they are on the probe. During the amplification reaction, the Taq DNA polymerase enzyme cleaves the probe in a template-dependent manner. The resultant probe fragments disassociate in solution, and signal from the released reporter dye is free from the quenching effect of the second fluorophore. One molecule of reporter dye is liberated for each new molecule synthesized, and detection of the unquenched reporter dye provides the basis for quantitative interpretation of the data. TaqMan.RTM. RT-PCR can be performed using commercially available equipment, such as, for example, ABI PRISM 7700.TM. Sequence Detection System. TM. (Perkin-Elmer-Applied Biosystems, Foster City, Calif, USA), or Lightcycler (Roche Molecular Biochemicals, Mannheim, Germany). In a preferred embodiment, the 5' nuclease procedure is run on a real-time quantitative PCR device such as the ABI PRISM 7700.TM. Sequence Detection System.TM.. The system consists of a thermocycler, laser, charge-coupled device (CCD), camera and computer. The system amplifies samples in a 96-well format on a thermocycler. During amplification, laser-induced fluorescent signal is collected in real-time through fiber optics cables for all 96 wells, and detected at the CCD. The system includes software for running the instrument and for analyzing the data. A more recent variation of the RT-PCR technique is the real time quantitative PCR, which measures PCR product accumulation through a dual-labeled fluorigenic probe (i.e., TaqMan.RTM. probe). Real time PCR is compatible both with quantitative competitive PCR, where internal competitor for each target sequence is used for normalization, and with quantitative comparative PCR using a normalization gene contained within the sample, or a housekeeping gene for RT-PCR. For further details see, e.g. Held et al., Genome Research 6:986-994 (1996). The genes which are assayed according to the present invention are typically in the form of mRNA or reverse transcribed mRNA. The genes may be cloned or not and the genes may be amplified or not. The cloning itself does not appear to bias the representation of genes within a population. However, it may be preferable to use polyA+RNA as a source, as it can be used with less processing steps. General methods for mRNA extraction are well known in the art and are disclosed in standard textbooks of molecular biology, including Ausubel et al., Current Protocols of Molecular Biology, John Wiley and Sons (1997). Methods for RNA extraction from paraffin embedded tissues are disclosed, for example, in Rupp and Locker, Lab Invest. 56:A67 (1987), and De Andrs et al., BioTechniques 18:42044 (1995). In particular, RNA isolation can be performed using purification kit, buffer set and protease from commercial manufacturers, such as Qiagen, according to the manufacturer's instructions. Other commercially available RNA isolation kits include MasterPure.TM. Complete DNA and RNA Purification Kit (EPICENTRE.RTM., Madison, Wis.), and Paraffin Block RNA Isolation Kit (Ambion, Inc.). Total RNA from tissue samples can be isolated using RNA Stat-60 (Tel-Test). RNA prepared from tumor can be isolated, for example, by cesium chloride density gradient centrifugation. As is apparent to one of ordinary skill in the art, nucleic acid samples used in the methods and assays of the invention may be prepared by any available method or process. Methods of isolating total mRNA are also well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in detail in Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I Theory and Nucleic Acid
Preparation, Tijssen, (1993) (editor) Elsevier Press. Such samples include RNA samples, but also include cDNA synthesized from a mRNA sample isolated from a cell or tissue of interest. Such samples also include DNA amplified from the cDNA, and an RNA transcribed from the amplified DNA. One of skill in the art would appreciate that it may be desirable to inhibit or destroy RNase present in homogenates before homogenates can be used. Biological samples may be of any biological tissue or fluid or cells from any organism as well as cells raised in vitro, such as cell lines and tissue culture cells. Frequently the sample will be a "clinical sample" which is a sample derived from a patient. Typical clinical samples include, but are not limited to, sputum, blood, blood-cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and pleural fluid, or cells therefrom. Biological samples may also include sections of tissues, such as frozen sections or formalin fixed sections taken for histological purposes. The following non-limiting examples are presented to better illustrate the invention. Methods and Materials The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, and biochemistry, which are within the skill of the art. Such techniques are explained fully in the literature, such as, "Molecular Cloning: A Laboratory Manual", 2.sup.nd edition (Sambrook et al., 1989); "Oligonucleotide Synthesis" (M. J. Gait, ed., 1984); "Animal Cell Culture" (R. I. Freshney, ed., 1987); "Methods in Enzymology" (Academic Press, Inc.); "Handbook of Experimental Immunology", 4.sup.th edition (D. M. Weir & C. C. Blackwell, eds., Blackwell Science Inc., 1987); "Gene Transfer Vectors for Mammalian Cells" (J. M. Miller & M. P. Calos, eds., 1987); "Current Protocols in Molecular Biology" (F. M. Ausubel et al., eds., 1987); and "PCR: The Polymerase Chain Reaction", (Mullis et al., eds., 1994).
Compounds: The structure of the small molecule KDR kinase inhibitors, Compound A and Compound B, used in the exemplification of the present invention are as follows:
Compound A
Figure imgf000018_0001
Compound B
Figure imgf000018_0002
Cell Culture: Primary human dermal microvascular endothelial cells (HDMVEC) and rat heart microvascular endothelial cells (RHMVEC) were purchased from VEC Technologies (Renneslaer, NY) and grown in culture according to the supplier's directions, endothelial cell monolayers were maintained at 37°C in a 5% C02 humidified atmosphere in tissue culture flasks coated with human fibronectin (Sigma, St. Louis, MO) using complete MCDB-131 media (MCDB-131 supplemented with 10% fetal bovine serum (FBS, Invitrogen, Carlsbad, CA) and the growth factor cocktail ENDOGRO, VEC Technologies). For in vitro MVEC proliferation experiments, cells were harvested by trypsinization between passage 3-6 following initiation of culture from frozen stocks, counted, and seeded in fibronectin-coated tissue culture plates at 75% confluence (1.5 x 106 cells/per plate, 100 mm diameter plates). Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 μg ml ENDOGRO. For growth arrest the culture media was changed to pre-warmed DMEM supplemented with 10% FBS. For stimulation of cell growth the growth arrest media was replaced with MDCB-131 supplemented with 10% FBS and the appropriate growth factor. Matched control plates that received no supplemental stimulatory growth factor were made for each stimulation condition. At the desired time following growth factor stimulation, the culture media was removed quickly by aspiration, and the cells were lysed in 1.2 ml RLT buffer (guanidine thiocyanate lysis buffer for RNA stabilization and purification, QIAGEN, Valencia, CA). Cell lysates were homogenized in QIAshredders and total RNA was isolated with RNeasy MINI affinity columns (QIAGEN, Valencia, CA). Gene expression profiles from a total of 8 independent VEGF-stimulated cultures, 7 ENDOGRO- stimulated cultures, and 4 bFGF-stimulated cultures were determined for HDMVECs. Profiles from 4 independent VEGF-stimulated cultures, 4 ENDOGRO-stimulated cultures, and 4 bFGF-stimulated cultures were determined For RHMVECs.
Animal Tumor Models: A rat glial cell line (C6, ATCC CCL-107) and a rat mammary adenocarcinoma (MatBni, ATCC CRL-1666) were used for our animal tumor models. C6 cells were maintained in culture at 37°C in a 5% C02 humidified atmosphere in Ham's F-12 medium supplemented with 2 mM L- glutamine, 1 mg/ml sodium bicarbonate, 15% horse serum, 2.5% fetal bovine serum, 10 U/ml penicillin, and 10 μg/ml streptomycin (all media components from Invitrogen). MatBffl cells were grown in McCoy's 5a medium supplemented with 1.5 mM glutamine, 10% FBS 10 U/ml penicillin, and 10 μg/ml streptomycin. For RNA isolation from C6 or MatBIII cells, 2x106 cells growing in a 100 mm diameter tissue culture plate were lysed directly in 1.2 ml RLT buffer. Following lysate homogenization with a QIAshredder, total RNA was isolated with RNeasy MINI affinity columns. C6 glial cells and MatBHI adenocarcinoma cells were chosen for animal models because they were obtained from Fischer 344 (F344) rats and therefore could be used to create syngeneic tumors in immunocompetent F344 animals. Prior to implantation, cells were collected, washed in phosphate- buffered saline and resuspended in Hanks Balanced Saline Solution (Invitrogen) at a density of 2x107 (C6) or 2x106 (MatBDI) cells/ml. C6 glioma flank tumor model: C6 cells were injected subcutaneously into the right flank of male F344 rats (150-175 g, 107 cells per animal). Following cell injection, animals were randomized according to body weight to receive either vehicle (0.5% methylcellulose) or drug (10 mpk/dose Compound B or 40 mpk/dose Compound A in 0.5% methylcellulose) (12-15). Once-daily oral dosing began 7 days post tumor cell implantation and continued for 1, 2, or 3 days at which point the animals were sacrificed. The tumors were bisected with half preserved for RNA extraction by snap-freezing in liquid nitrogen and half fixed for histology or immunofluorescence microscopy. Five vehicle-treated and five compound-treated animals were sacrificed at each timepoint. RNA was extracted from tumor samples with RNeasy Mini columns according to standard protocols (QIAGEN). Briefly, frozen tumor samples were weighed, placed in sample tubes containing RLT buffer (600 μl RLT per 30 mg tissue), and immediately homogenized for 10-20 seconds using a rotor/stator homogenizer. Total RNA was isolated from homogenized tissue lysate with RNeasy affinity columns, resuspended in DEPC-treated water and frozen at -80°C. RNAs from the five tumors in each vehicle-treated cohort were combined to form three reference RNA pools. RNAs isolated from each of the tumor samples from the five compound-treated rats in each cohort were compared to the appropriate time-matched reference pool of RNA during microarray hybridization. In addition, RNA from individual vehicle treated rats was compared to time matched vehicle treated pool in order to assess inter-animal variability.
MatBiπ Breast Cancer Metastasis Model: MatBIII cells between passage 20-30 were injected on the mammary fat pad around the 4th left nipple (106 cells/animal) of female F344 rats (150-175 g). Prior to dosing, animals were randomized into groups according to tumor size and body weight. Once daily oral dosing of Compound A (40 mpk dose formulated in 0.5% methylcellulose) or vehicle (0.5% methylcellulose) began on day 7 post tumor cell implantation and continued for 4 additional days. Six vehicle-treated and six compound-treated animals were sacrificed on day 11, four hrs after final dosing. At the time of necropsy, tumors were weighed immediately upon removal. Half of each tumor was fixed in Zn-Tris for histology or immunofluorescence microscopy and the other half immediately snap frozen in liquid nitrogen for RNA extraction. Total RNA was isolated in the same manner as with the C6 tumor studies. RNAs from the vehicle-treated cohort were combined to form a control RNA pool. RNAs isolated from each of the tumor samples from the compound-treated rats was compared to the control pool of RNA during microarray hybridization. RNAs from individual vehicle treated animals was compared to the vehicle treated pool in order to assess inter-animal variability.
Gene Expression Profiling: Total RNA isolated from cultured cells or tumor tissue samples was used to make fluorescently-labeled complementary RNA (cRNA) that was hybridized to DNA microarrays as previously described (16, 17). Briefly, 4 μg total RNA from an individual tumor sample or in vitro endothelial cell culture was used to synthesize double-stranded DNA through reverse transcription. cRNA was produced by in vitro transcription and labeled post-synthetically with Cy3 or Cy5. Two populations of labeled cRNA, a reference population and experimental population, were compared to each other by competitive hybridization to oligonucleotide arrays synthesized in situ with inkjet technology. Two hybridizations were performed with each cRNA sample pair using a fluorescent dye reversal strategy. For animal tumor studies, reference cRNA pools were made by pooling equal amounts of cRNA from each tumor in the appropriate vehicle-dosed group. After hybridization, arrays were scanned and fluorescence intensities for each feature were recorded. Ratios of transcript abundance (experimental to control) were obtained following normalization and correction of the array intensity data. Gene expression data analysis was performed with the Rosetta Resolver Client software (v3.2, Rosetta Biosciences, Kirkland, WA). A one-way ANOVA test was used to determine statistically significant changes in gene expression.
Quantitative real time PCR: Quantitative real time polymerase chain reaction was performed with gene- specific PCR primer pairs and amplicon-specific fluorescent probes (TaqMan, Applied Biosystems Inc. (ABI), Foster City, CA) according to published protocols (ABI Assays-on-Demand™ Gene Expression Protocol, Rev A, http://docs.appliedbiosystems.com/pebiodocs/04333458.pdf). One-step quantitative reverse transcription PCR reactions were performed using ABI's TaqMan® One-Step RT-PCR Master Mix Reagents (ABI Product# 4309169) and 25 ng total RNA template on an ABI Prism 7900HT Sequence Detection System. Two-step reverse transcription PCR experiments were initiated by cDNA synthesis from 25 ng total RNA as template using ABI's High Capacity cDNA Archive Kit (ABI Product# 4322171). Second step quantitative real time PCR was performed with standard reagents (TaqMan® Universal PCR Master Mix, ABI Product# 4324018) on the ABI Prism 7900HT Sequence Detection System. Real time PCR reactions were performed in duplicate in a 25 μl reaction volume in 384-well plates. Primer and probe sequences used for each gene are listed below. For every RNA sample, transcript abundance of GAPDH was determined. In addition, transcript abundance of genes of interest and GAPDH were determined for calibrator RNA samples, either total human lung RNA or total rat lung RNA. Fold changes in gene expression were calculated using the ΔΔCT method (ABI User Bulletin #2, Rev B: Relative Quantitation Of Gene Expression, http://docs.appliedbiosystems.com/pebiodocs/04303859.pdf) The teachings of which are incorporated herein by reference. The sequences of the individual biomarker genes which comprise the proliferation and expression signatures disclosed in tables 3-6 are readily available in public databases. The signatures defined in Tables 3 and 4 (HDMVEC and RHMVEC proliferation signatures, respectively) provide the GenBank Accession Number and gene symbol for the included biomarkers, the sequences of which are hereby incorporated by reference. The endothelial cell specific signatures defined in Tables 5 and 6 also provide the GenBank Accession Number and GeneSymbol for the biomarkers comprising the expression signature. The following list provides the name, Accession Number and primer and probes used to detect a subset of the biomarkers: Hs refers to Homo sapiens. Rn refers to Rattus norvegicus. Gene RefSeq Acc# Primer Sequence (or ABI Assay-on-Demand™ ID)
Angpt-2 XM_225004 (Rn)
SEQ ID NO: 1 Forward Primer 5' - GAC AGA GTC CGA ATG CAT GCT - 3' SEQ ID NO: 2 Reverse Primer 5' - TGC GGG TCT GGA GAA ATA CC - 3' SEQ ID NO: 3 TaqMan Probe 5' - CCC TGT GAT TCT AAC CAT GGC CTT CTC A - 3'
NM_001147 (Hs) Hs00169867 ml
Ifit-3 XM_220059 (Rn)
SEQ ID NO: 4 Forward Primer 5' - CGG TTG TTA TCA GGC TCA TAGGAT - 3' SEQ ID NO: 5 Reverse Primer 5' - TGT GGGAGG CAA CAC GAT TT - 3' SEQ ID NO: 6 TaqMan Probe 5' - TCA GGA ATA GGCTGC CTG CAC CCC - 3'
Fut-4 NM_022219 (Rn)
SEQ ID NO: 7 Forward Primer 5' - GAC CGA AAC GTG GCT GTC TAT C - 3' SEQ ID NO: 8 Reverse Primer 5' - GTG ATGTGC ACC GCA TAG CT - 3' SEQ ID NO: 9 TaqMan Probe 5' - CCG CTA CTT CCA CTGGCG TCG G- 3'
NM_002033 (Hs)
SEQ H NO: 10 Forward Primer 5' - AAT TGG GCT CCT GCA CAC -3' SEQ ID NO: 11 Reverse Primer 5' - CCA GGT GCT GCG AGT TCT C - 3' SEQ ID NO: 12 TaqMan Probe 5' - TGG CCC GCT ACA AGT TCT ACC TGG CTT - 3'
Plau NM_013085 (Rn) Rn00565261_ml NM_002658 (Hs) Hs00170182_ml Clu NM_012679 (Rn) Rn00562081_ml NM_001831 (Hs) Hs00156548_ml
Etb NM_017333 (Rn) Rn00569139_ml NM_000115 (Hs) Hs00240752_ml
Cyrόl NM_031327 (Rn) Rn00580055_ml NM_001554 (Hs) Hs00155479_ml
GAPDH NM_017008 (Rn) 4308313
GAPDH NM_002046 (Hs) 402869
Immunohistochemistry: Tumor sam pies were fixed immei by submersion in a Zn-Tris fixative solution for immunohistochemistry (MC Zinc Fixative, BD Biosciences-Pharmingen, San Diego, CA) for 24 hr at room temperature (RT, 22°C) followed by submersion in 70% ethanol at RT for an additional 24 hrs. All subsequent steps were performed at RT. Tumor samples were embedded in paraffin (Tissue-Tek VIP Processing/Embedding Medium, Sakura Finetek, Torrance, CA) and cut into 3 μm sections on a Sakura Accu-Cut SRM microtome (Sakura Finetek). Tissue sections were de-waxed in xylene and re-hydrated through graded ethanol washes. Following washes in deionized H20 (dH20) and tris-buffered saline (TBS), a hydrophobic barrier was placed around the tissue section with a hydrophobic pen (Super Pap Pen, EMS #71310).
CD31 staining: CD31 is a validated endothelial cell-specific protein (18-20). Sections were blocked with Protein Block (Biogenex, San Ramon, CA) for 30 min and incubated with anti-CD31 antibodies (mouse anti-rat, Serotec, Raleigh, NC) diluted 1: 1000 in DAKO Antibody Diluent with Blockers
(DakoCytomation, Carpinteria, CA) for two hours. After several brief washes in TBST (TBS + 0.1% Tween-20), sections were incubated with biotinylated anti-Mouse IgG secondary antibody (DakoCytomation Alkaline Phosphatase Kit Link K-0610) for 10-30 min, washed several times with TBST, and incubated with streptavidin coupled to alkaline phosphatase (DAKO Alkaline Phosphatase Kit K-0610) for 10-30 min. Sections were then washed again with several changes of TBST and CD31 bound antibodies were visualized by incubation with Vulcan Fast Red Substrate (Biocare Medical, Walnut Creek, CA) for lOmin (color development monitored microscopically). Sections were then washed in dH20 stored overnight in TBS. Ki67 Staining: Ki67 is a validated nuclear protein expressed only in proliferating cells (21, 22). To facilitate antibody recognition of Ki67, we used a high temperature antigen retrieval strategy. Sections were submerged in Target Retrieval Solution (lx DakoCytomation S1699 diluted with dH20) in a Decloaking Chamber (Biocare Medical, DC2002) and heated to 195°C for 1 min. Sections were cooled with running dH20 into the retrieval solution and then rinsed in TBS. Residual peroxidase activity was blocked by incubating the sections with 3.0% H202 in TBS for 20 min. Sections were washed several times in TBS, then incubated with anti-Ki67 antibodies (rabbit anti-human, Novacastra, Newcastle upon Tyne, UK) diluted 1:2000 in antibody diluent for 2 hrs. Sections were washed with TBST, and then incubated with undiluted biotinylated anti-rabbit IgG (DakoCytomation, Link K-0609) for 10 min. Sections were washed in TBST, and then incubated with streptavidin coupled to horseradish peroxidase (DakoCytomation, K0609) for 10 min. Sections were washed again in TBST, and antibodies bound to Ki67 were visualized by incubation with diaminobenzidine plus substrate (DakoCytomation, DAB+) for 5 min (color development monitored microscopically). Sections were washed in dH20, incubated with DAB Enhance for 20 min RT, and washed again with dH20. Tumor sections were counterstained with filtered Mayer's Hematoxylin (Lillie's Formulation, DakoCytomation) for two min, and then washed with tap H20 until no color remained in the wash water. Sections were then rinsed in dH20, dehydrated with 100% ethanol, cleared with xylenes and mounted with Permount (Fisher Scientific, Hampton, NH).
Immunohistochemical Analysis of Endothelial Cell Proliferation: Sequential brightfield images of CD31/Ki67 double-labeled tumor sections were obtained with a 3-CCD color video camera (Optronics) attached to an Olympus BX-50 microscope equipped with an automated stage (Prior H128, Watertown, MA) and a 40x objective. The number of images per section varied between 1000 and 4000 depending on total tissue area. CD31 staining and Ki67 staining were quantitated for each image using the ImageProPlus software package (Media Cybernetics, Carlsbad, CA). Proliferating endothelial cells were identified as those cells with cytoplasmic CD31 staining and nuclear Ki67 staining. Cells staining positive for CD31 but without nuclear staining for Ki67 were scored as non-proliferating endothelial cells. The percentage of proliferating endothelial cells was calculated by dividing the Ki67 -positive nuclear area associated with endothelial cells by the total nuclear area associated with endothelial cells (both Ki67+ and Ki67-). Endothelial cell proliferation percentages represent the combined analysis results from at least 100 images with CD31 staining per tumor section.
Immunofluorescence Microscopy: Tumor samples were fixed, embedded, sectioned, de-waxed, and re- hydrated as described for immunohistochemistry above. All subsequent steps were performed at room temperature. After a brief rinse in TBS, tissue sections were blocked by incubation with Sniper Blocking Reagent (Biocare Medical) for 5-10 min, rinsed in TBS and incubated with primary antibodies diluted 1: 1000 in DAKO Antibody Diluent for 2 hrs (Antibodies against ANGPT2, CLU, CYR61, and PLAU were from Santa Cruz Biotechnology, Santa Cruz, CA and were raised in goat or rabbit; antibodies against EDNRB were from Calbiochem, San Diego, CA and raised in sheep, antibodies against CD31 were from Serotec and raised in mouse). Sections were then washed with Tris-buffered saline containing 0.2% Tween-20 (TBST, Sigma) and incubated with appropriate secondary antibodies diluted 1:200 (lOug/ml) in DAKO Antibody Diluent with blocking serum for 45 min (Alexa Fluor 488 donkey anti- goat IgG, Alexa Fluor 488 goat anti-rabbit IgG, Alexa Fluor 488 donkey anti-sheep IgG, Molecular Probes, Eugene, OR; Normal donkey and normal goal blocking serum, Sigma). Following additional washes with TBST, sections were counterstained with DAPI (Molecular Probes, 1:2000 dilution of 1 mg/ml stock in MQH20) for 30 min. Sections were then washed in TBST, dehydrated in 100% EtOH, cleared in xylene, and mounted under coverslips with Permount. Images were captured with a Zeiss Axiocam mHR CCD camera connected to a Zeiss Axiovert 135 inverted fluorescence microscope equipped with a 40x objective. For each fluorophore, all images were captured using equal camera integration times.
EXAMPLE 1 IDENTIFICATION OF GENE EXPRESSION CHANGES IN PROLIFERATING MICROVASCULAR ENDOTHELIAL CELLS In order to identify genes that are regulated in proliferating endothelial cells relative to quiescent endothelial cells, we employed an in vitro angiogenesis model in which primary cultured microvascular endothelial cells were driven to proliferate from a quiescent state by incubation with growth factors. Primary human dermal microvascular endothelial cells (HDMVECs) or rat heart microvascular endothelial cells (RHMVECs) between passage 4 and 7 were grown in monolayers in tissue culture dishes, mitogen-starved for 24 hr, then induced to proliferate by exposure to VEGF, bFGF, or ENDOGRO. HDMVECs or RHMVECs were trypsinization following the third passage in culture and seeded in fibronectin-coated, six-well tissue culture plates at a density of 10,000 cells well. Cell growth was arrested for 24h by mitogen withdrawal and then stimulated by the addition of 100 ng/ml VEGF, 100 ng/ml bFGF or 200 μg/ml ENDOGRO. Wells with unstimulated cells and wells containing un-arrested cells were included as controls. At 72 hr following growth factor stimulation, cells were removed from the culture plates by trypsinization and counted on a hemocytometer under bright-field microscopy. To confirm that VEGF, bFGF, and ENDOGRO (a bFGF-rich bovine brain extract, VEC Technologies) were signaling through different growth factor receptors, we exposed cells to VEGF, bFGF, or ENDOGRO in the presence of Compound A, which is small molecule KDR kinase inhibitor that is 100-fold less active against FGFR1 and FGFR2 (Table 1) (12-15).
Figure imgf000026_0001
A comparison of the gene expression pattern of mitogen-starved, quiescent HDMVECs and RHMVECs to the expression pattern of actively dividing endothelial cells, indicates significant
(p-value <0.01)gene expression changes, that are characteristic of proliferating vascular endothelial cells. Briefly, endothelial cell cultures were grown in culture and mitogen deprived for 24 hr as described above. Following a 24 hr stimulation with growth factor, culture media was aspirated quickly and the cells lysed in an RNA stabilizing buffer. Matched control plates that received no supplemental stimulatory growth factor were present for each stimulation condition and RNAs isolated from them served as the reference to which the RNAs from the stimulated cells was compared. Results: Although growth media supplemented with 10% FBS was not sufficient to drive endothelial cell proliferation, 10% FBS plus additional growth factor induced rapid endothelial cell proliferation. The data provided in Figure 1 demonstrates that exposure of HDMVEC (Figure 1A) and RHMVEC (Figure IB) to Compound A selectively inhibits in vitro microvascular endothelial cell proliferation induced by VEGF as determined by viable cell counting with a hemocytometer. The gene expression profile illustrated in Figure 2 graphically illustrates a gene expression signature that is characteristic of proliferating HDMVEC (panel A) and RHMVEC (panel B) cultures. Color intensity represents the degree of regulation, not mRNA copy number. Table 3 provides a list of genes comprising the HDMVEC Proliferative Signature identified in the expression profiles illustrated in Figure 2. Table 4 provides a list of genes comprising the RHMVEC Proliferative Signature identified in the expression profiles illustrated in Figure 2. Using the information provided in Tables 3 and 4 it is well within the abilities of a skilled artisan to design and validate a gene expression-based assay that is suitable for evaluating the proliferation of vascular endothelial cells in either in vitro or in vivo screening assay format. EXAMPLE 2 SUPPRESSION OF VEGF-INDUCED GENE EXPRESSION SIGNATURES IN PRIMARY ENDOTHELIAL CELLS BY A KDR KINASE INHIBITOR To determine if the growth factor-induced proliferation signatures identified in Example lare sensitive to KDR kinase inhibitors, we stimulated HDMVECs or RHMVECs with VEGF or bFGF for 24 hrs in the presence of 100 nM Compound B. VEGF binds to and activates the fms-like tyrosine kinase (FLT1) and KDR (23, 24). Both FLTl and KDR are inhibited by Compound B (Table 1). bFGF binds to FGFR1 and FGFR2, but not FLTl or KDR. Both FGFR1 and FGFR2 are relatively insensitive to Compound B (see Table 1). Briefly, EC monolayers were maintained in complete MCDB-131 media until reaching -75% confluence, then induced into a quiescent state by mitogen starvation for 24 hr. Cells were then stimulated to proliferate with 100 ng/ml VEGF for 24 hr in the presence or absence of Compound B. RNA populations isolated from cells exposed to VEGF or VEGF + Compound B were compared to matched control RNAs isolated from quiescent cells exposed to neither VEGF nor Compound B. Results: The data provided in Figure 3 demonstrate that the HDMVEC VEGF-induced gene expression signature was effectively suppressed by Compound B while the bFGF-induced signature was unaffected. Parallel experiments were performed with RHMVECs (data not shown). Each point in the plots represents a gene sequence present on the DNA oligonucleotide microarray and is plotted according to the ratio of the two mRNA levels (experimental sample intensity ontrol sample intensity, vertical-axis) and the total mRNA quantity (experimental sample intensity + control sample intensity, horizontal-axis) for that gene.
EXAMPLE 3 IDENTIFICATION OF AN ENDOTHELIAL CELL-SPECIFIC PROLIFERATION SIGNATURE The experimental data provided above identifies gene expression profiles, or expression signatures specific for proliferating endothelial cells. However, the majority of genes regulated during endothelial cell proliferation will also be expressed in other types of proliferating cells (genes that regulate cell cycle and metabolic processes, for example). Tumors contain a complex mixture of cell types, where approximately 1 in 2000 cells (0.05%) are proliferating endothelial cells (Joanne Antanavage, Rosemary McFall, and Ken Thomas, personal communications). Therefore, in attempting to develop a pharmacodynamic assay that specifically measure tumor endothelial cell proliferation, it was acknowledged that there was a need to identify the endothelial cell-specific portion of the HDMVEC and RHMVEC proliferation signatures. Candidate endothelial cell-specific genes were defined as genes characterized by regulated expression during an in vitro proliferative response to mitogens, but expressed at relatively low levels in non-endothelial cells. We used microarray intensity data, which corresponds to the number of labeled cRNAs bound to each array feature and is proportional to mRNA copy number, from previous expression profiling studies and compared it with the microarray intensity data from our HDMVEC proliferation experiments. Existing intensity data from a panel of actively growing tumor-derived cell lines (MOLT-4, HL-60, Raji, SW480, Daudi, G361, A549, K562, MCF7) was used to remove from consideration those genes with EC:tumor microarray intensity ratios less than 3: 1. 702 HDMVEC sequences were selected as endothelial cell-specific in this manner (see, Table 3). We identified many known endothelial cell-specific genes by this method (i.e. ESM-1KDR, FLT-1) as well as numerous novel sequences. In parallel, we obtained a measure of endothelial cell specificity for genes regulated in proliferating RHMVECs by comparing microarray intensity data from the RHMVEC experiments to data from gene expression profiling experiments with rat C6 glioma cells actively growing in culture. We identified 493 genes with RHMVEC:C6 intensity ratios greater than 3:1 (see, Table 4).
EXAMPLE 4 ORALLY-DOSED KDR KINASE INHIBΠΌRS INDUCE SIGNIFICANT GENE EXPRESSION CHANGES IN SYNGENEIC ANIMAL TUMORS Two syngeneic rat tumor models (i.e., C6 glioma flank tumor model and MatBIII Breast Cancer Metastasis Model), were used to assess the effect of the small molecule KDR kinase inhibitors, Compound A and Compound B, on thE genes identified as endothelial cell-specific which were regulated during in vitro endothelial cell proliferation. Tumor studies were performed as described above in
Materials and Methods. The tumor models use C6 glioma and MatBIII mammary carcinoma cell lines, both derived from Fischer 344 rats. These cell lines each secrete VEGF and form highly vascularized tumors that are sensitive to KDR kinase inhibitors. Glioma Flank Tumor Model: C6 cells were injected subcutaneously into the right flank of rats and allowed to form tumors for seven days. At that time, once-daily oral dosing with Compound A,
Compound B, or vehicle commenced and continued for a total of 1, 2 or 3 days (Figure 4, Panel A "C6 Profiling Study). Figure 4 illustrates the growth kinetics of established rat tumors following exposure to a KDR kinase inhibitor. Tumor volumes were determined by caliper measurements. Tumors were calipered in two dimensions (length and width) and tumor volume was calculated according to the formula (length) x (width) x (lΛ width). Genome-wide gene expression in tumors isolated from compound-treated animals was compared to gene expression from tumors isolated from vehicle-treated animals. In the data provided in Figure 5 each row represents a distinct tumor from an individual animal. Each column represents a gene. Points corresponding to genes which are regulated (upregulated or downregulated) are indicated by various shades of gray. The data presented in Panel A of Figure 5 identifies genes from rat C6 flank tumors that are regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound B. The data presented in Panel B of Figure 5 identifies genes from rat C6 flank tumors regulated following 24, 48, or 72 hrs of systemic exposure to the KDR kinase inhibitor Compound A. We observed that both Compound A (Figure 5, Panel B) and Compound B (Figure 5, Panel A) induced robust gene expression changes in C6 tumor gene expression, particularly after 48 hrs or more of compound exposure (p-value < 0.05 for individual sequences).
MatBIII Breast Cancer Metastasis Model: MatBIII mammary adenocarcinoma cells were injected into a mammary fat pad of female rats. After allowing tumors to establish for seven days once-daily oral dosing of Compound A began and continued for a total of 5 days (Figure 4B). The data presented in Panel C of Figure 5 identifies genes from rat MatBIII mammary tumors regulated following 100 hrs of systemic exposure to the KDR kinase inhibitor Compound A. When the pattern of tumor gene expression from compound-treated rats was compared to vehicle-treated controls, we again found significant differences (Figure 5, Panel C, p-value < 0.05 for individual sequences). While there was overlap in the gene expression changes induced by the KDR kinase inhibition between the three studies, the majority of gene expression changes were study-specific (data not shown).
EXAMPLE 5 IDENTIFICATION OF GENE EXPRESSION BIOMARKERS OF ENDOTHELIAL CELL PROLIFERATION In each of the three animal studies we performed, we found that we could detect KDR kinase inhibitor-induced changes in expression for a fraction of those genes we had identified as specific to proliferating RHMVECs in culture. Figure 6 provides Venn diagrams which summarize the degree of overlap between the set of genes identified in the various assays formats disclosed herein. More specifically, Figure 6A summarizes the degree of overlap between the tumor gene expression responses to KDR kinase inhibitors in C6 flank tumors and MatBIII mammary tumors. Figure 6B indicates the degree of overlap between the sets of endothelial cell-specific genes determined to be regulated both in vitro by mitogens and in tumor tissue by KDR kinase inhibitors. All genes/sequences represented in Panel B were observed to be regulated in vivo by KDR kinase inhibitors in a manner opposite that observed in vitro following exposure to mitogens. Most interestingly, we found in each study that some of those genes were regulated in a manner consistent with suppression of endothelial cell proliferation. In effect, these genes were oppositely regulated in our in vitro proliferation experiments as compared to our in vivo tumor studies. In both cases the genes were highly expressed when endothelial cells were proliferating and expressed at low levels under non-proliferating conditions. Thus, we identified endothelial cell-specific genes that were "oppositely regulated" in each of the three animal tumor studies and identified genes that were regulated as such in multiple studies (Figure 6B, and Tables 5 and 6). Tables 5 and 6 provide a list of genes which were observed to be regulated by Compound A. Table 5 utilizes the data obtained in the C6 Flank Tumor and MatBill Breast Cancer Metatesis Models to define a Compound induced endothelial cell-specific expression signatures. The table provides the GeneBank Accession number, the gene symbol and summarizes the compound-induced fold change in gene expression that was observed. It is contemplated that the expression signatures provided in Table 5 will find utility in evaluating the in vivo efficacy of anti-angiogenic agents in general and KDR kinase inhibitors in particular. Table 6 provides a summary of the changes (i.e., suppressed expression) observed for individual biomarkers comprising the EC- specific proliferation signatures disclosed herein in response to in vivo KDR inhibitor administration. By imposing a requirement that genes to be considered as biomarkers should have compound- induced in vivo expression changes of at least 1.6 fold, we identified seven genes that were "oppositely regulated" in both Compound A and Compound B studies and two genes that were "oppositely regulated" in all three studies. Based on the data provided in the instant disclosure, the seven genes (Angpt2, Ednrb, Plau, Clu, Fut4, Ifit3, and Cyrόl) identified by both Compound A animal studies were identified as potential biomarkers for tumor endothelial cell proliferation. These genes are identified and described in Table 2.
Table 2
Figure imgf000031_0001
Significantly, each of these genes has been reported to be involved or implicated in endothelial cell function. Table. 3
HDMVEC Proliferation Signature GenBank Accession EC:MCF7 Intensity Growth Number Gene Symbol (Expression) Ratio Fold Change Factor AA013218 FLJ 14079 6.81 1.33 ENDOGRO AA029441 8.22 -1.17 VEGF AA046478 NT5 4.48 1.35 ENDOGRO AA053711 RBM8B 86.86 -1.38 ENDOGRO AA053806 LOC64148 35.28 -1.45 ENDOGRO AA102600 12.51 -1.51 bFGF AA148511 FLJ22724 71.39 -1.18 bFGF AA156672 28.11 1.51 ENDOGRO AA166703 10.11 1.37 VEGF AA173992 4.15 1.42 ENDOGRO AA224245 14.16 -1.22 ENDOGRO AA404374 FLJ21935 21.27 1.40 VEGF AA449120 28.44 2.02 VEGF AA464846 33.20 -1.34 VEGF AA489383 BMP2 5.09 1.66 VEGF AA522536 9.14 -1.55 bFGF AA534774 3.09 -1.38 ENDOGRO AA541787 12.99 -2.77 bFGF AA584310 28.05 -1.36 ENDOGRO AA617813 4.03 -1.35 VEGF AA621714 8.79 -1.38 VEGF AA628517 10.91 1.58 ENDOGRO
AA632012 12.16 -1.37 VEGF
AA707332 6.09 1.27 bFGF
AA740709 8.73 -1.72 ENDOGRO
AA758545 MMP2 93.44 -1.41 ENDOGRO
AA811265 10.22 -1.30 bFGF
AA815048 FLJ 12649 4.37 -1.79 bFGF
AA858297 152.47 1.61 bFGF
AA868377 FLJ22233 3.66 1.45 ENDOGRO
AA868615 3.33 -2.01 bFGF
AA873008 8.82 -2.04 bFGF
AA897516 8.35 1.62 ENDOGRO
AA903334 29.77 1.57 ENDOGRO
AA923461 12.34 -1.30 ENDOGRO
AA932206 11.75 1.94 VEGF
AA946945 14.82 2.11 ENDOGRO
AB007954 KIAA0485 3.69 -1.44 VEGFD
AB011099 KIAA0527 104.01 -1.30 ENDOGRO
AB014538 KIAA0638 6.54 1.31 VEGF
AB014567 KIAA0667 3.53 -1.29 ENDOGRO
AB014604 KIAA0704 23.34 1.33 ENDOGRO
AB018301 KIAA0758 243.54 1.90 VEGF
AB018333 KIAA0790 3.12 1.57 ENDOGRO
AB018339 SYNE-1 B 9.29 -1.44 bFGF
AB028019 LATS2 5.74 -1.43 ENDOGRO
AB028976 KIAA1053 11.78 -1.69 bFGF
AB028981 KIAA1058 6.73 1.24 VEGF
AB032971 KIAA1145 13.30 2.04 bFGF
AB033006 NDRG4 23.14 -1.79 VEGF
AB033035 KIAA1209 12.08 1.68 VEGF
AB033093 DKFZP727C091 4.05 -1.59 VEGFE
AB033100 KIAA1274 32.44 1.41 ENDOGRO
AB033101 KIAA1275 5.36 -1.86 bFGF
AB037722 KIAA1301 4.32 1.80 VEGF
AB037726 KIAA1305 5.96 -1.48 ENDOGRO
AB037751 KIAA1330 8.51 -1.42 ENDOGRO
AB037784 KIAA1363 3.50 1.65 ENDOGRO
AB037820 KIAA1399 289.61 -1.44 ENDOGRO
AB037821 PCDH10 8.07 -1.72 ENDOGRO
AB037857 KIAA1436 3.61 -1.16 ENDOGRO
AF007150 18.82 1.66 ENDOGRO
AF035121 KDR 70.01 1.39 bFGF
AF035306 12.18 -1.34 bFGF
AF035318 47.64 -1.68 bFGF
AF041037 SPRY1 10.05 1.90 VEGF
AF052169 83.10 -1.37 VEGF
AF061034 FIP2 8.51 -1.62 ENDOGRO AF062341 CTNND1 5.61 -1.12 ENDOGRO
AF070569 4.62 -1.41 ENDOGRO
AF070641 8.76 4.19 ENDOGRO
AF091434 PDGFC 67.66 -1.37 ENDOGRO
AF095719 CPA4 19.14 -1.56 ENDOGRO
AF101051 CLDN1 3.73 -2.39 ENDOGRO
AF114264 unknown 35.87 -2.74 ENDOGRO
AF119663 LOC55970 6.47 -1.26 ENDOGRO
AF131762 4.22 1.34 ENDOGRO
AF131817 13.30 -1.49 VEGF
AF134404 FADS3 3.00 -1.58 ENDOGRO
AF181265 EHD4 5.55 1.09 ENDOGRO
AF 186780 KIAA0959 14.14 1.53 VEGF
AF218942 FMN2 6.56 -1.75 bFGF
AF234532 MYO10 6.56 1.26 ENDOGRO
AF238083 SPHK1 4.48 1.37 ENDOGRO
AI005420 4.74 1.48 VEGFE
AI031794 7.72 -1.99 bFGF
AI039171 3.98 -1.33 ENDOGRO
AI051390 3.00 -1.32 ENDOGRO
AI05251 1 12.16 1.52 VEGF
AI073464 PLG 79.09 -1.25 ENDOGRO
AI073669 56.60 1.82 ENDOGRO
AI079944 5.90 1.44 VEGF
AI085787 29.37 3.06 VEGF
AI088104 8.74 -1.40 VEGF
AH 25204 16.83 -2.15 bFGF
AH 25425 7.25 1.68 ENDOGRO
AM 39987 FLJ23056 32.76 -1.69 bFGF
AH 41554 2709.99 -1.59 VEGF
AI141700 LOC63875 6.99 1.30 VEGFE
AM 68436 7.67 -1.40 bFGF
AI188161 109.32 -2.23 bFGF
AI188513 3.42 1.27 ENDOGRO
AI200874 23.78 -1.45 ENDOGRO
AI203531 ART4 3.56 -2.05 ENDOGRO
AI206317 34.47 -1.39 ENDOGRO
AI208788 5.00 -2.02 bFGF
AI218538 20.83 -1.81 ENDOGRO
AI223799 3.29 -1.15 ENDOGRO
AI224533 DKFZp762L0311 3.19 1.70 ENDOGRO
AI275691 13.51 1.18 ENDOGRO
AI277316 4.21 -1.98 bFGF
AI291779 45.84 -1.44 VEGF
AI301312 6.62 -1.97 ENDOGRO
AI338631 11.26 -1.33 VEGFE
AI343000 8.60 -1.36 ENDOGRO AI351898 14.00 1.13 ENDOGRO
AI357650 AD026 4.02 1.87 VEGF
AI375677 8.94 1.47 VEGF
AI376749 SDC2 5.18 -1.89 ENDOGRO
AI378647 6.03 -1.84 ENDOGRO
AI392987 HOXB6 4.40 1.44 ENDOGRO
AI418293 54.98 -1.34 VEGFE
AI418530 4.00 1.37 PIGF
AI418596 10.33 -1.62 bFGF
AI420933 54.00 -2.63 VEGF
AI433789 OS4 3.70 -1.44 ENDOGRO
AI433914 4.47 1.32 VEGF
AI439093 8.40 1.27 ENDOGRO
AI453557 24.20 -1.50 ENDOGRO
AI478770 MYH9 4.94 -1.39 ENDOGRO
AI479854 FLJ20980 46.10 -1.24 VEGF
AI498132 3.84 1.33 ENDOGRO
AI523391 3.67 -1.41 bFGF
AI539275 3.09 -1.22 VEGF
AI569689 3.53 -2.60 ENDOGRO
AI608902 17.83 -1.64 ENDOGRO
AI610727 4.23 -1.21 ENDOGRO
AI633826 5.30 3.89 VEGF
AI633890 48.69 -1.40 VEGF
AI635050 FLJ22252 327.52 -1.79 ENDOGRO
AI652289 31.67 1.32 ENDOGRO
AI652898 30.60 2.98 bFGF
AI652991 13.27 -1.76 ENDOGRO
AI654230 21.37 1.31 ENDOGRO
AI655345 9.29 1.47 ENDOGRO
AI659533 ARGBP2 108.45 -2.15 ENDOGRO
AI659800 4.82 -1.45 ENDOGRO
AI672407 HOXB8 3.72 2.26 bFGF
AI674404 7.93 1.21 bFGF
AI681538 FLJ23403 40.51 -1.48 VEGF
AI681805 15.66 -1.42 ENDOGRO
AI682468 3.07 -1.43 bFGF
AI683621 8.18 -1.77 bFGF
AI684489 CSF2RB 7.89 -1.59 ENDOGRO
AI684705 9.25 -1.29 VEGFE
AI688546 ARHGEF1 35.74 1.25 VEGFE
AI693178 22.73 -1.48 ENDOGRO
AI733194 5.60 -1.26 ENDOGRO
AI733570 FLJ20898 566.31 2.21 VEGF
AI739507 DAB2 1074.51 -1.73 ENDOGRO
AI741128 53.39 2.07 VEGF
AI741880 131.82 1.49 VEGF AI742043 5.43 1.37 ENDOGRO
AI742210 4.99 1.28 ENDOGRO
AI742878 6.05 -1.77 VEGF
AI742936 8.15 1.23 ENDOGRO
AI743880 7.62 -1.36 ENDOGRO
AI743942 6.90 1.84 ENDOGRO
AI744591 31.91 1.34 ENDOGRO
AI745230 7.26 -1.52 VEGF
AI745614 27.20 -1.44 ENDOGRO
AI754423 83.30 -1.58 ENDOGRO
AI760613 6.16 -1.89 VEGF
AI765437 15.10 -1.55 ENDOGRO
AI767993 5.35 1.46 VEGF
AI769801 ALB 4.74 1.87 VEGF
AI803656 5.73 -1.27 ENDOGRO
AI806221 81.13 1.75 VEGF
AI806313 FLJ23091 34.79 -1.15 ENDOGRO
AI807266 6.54 1.46 ENDOGRO
AI810042 FLJ21841 16.31 1.44 ENDOGRO
AI822137 5583.85 -1.44 ENDOGRO
AI823801 SE57-1 5.65 1.35 ENDOGRO
AI825936 KIAA1350 12.43 1.42 ENDOGRO
AI827455 45.22 1.46 VEGF
AI828007 9.30 1.37 ENDOGRO
AI857683 3.74 1.27 VEGF
AI861824 SLC1A1 60.49 1.41 ENDOGRO
AI862120 6.44 -1.33 bFGF
AI887362 3.58 1.48 ENDOGRO
AI889160 5.58 -1.51 ENDOGRO
AI912975 71.62 -1.52 ENDOGRO
AI913402 75.58 -1.34 VEGF
AI924550 4.97 1.54 ENDOGRO
AI926697 6.91 1.67 ENDOGRO
AI927454 8.47 -1.14 ENDOGRO
AI927919 30.58 -1.27 ENDOGRO
AI928427 24.79 1.50 bFGF
AI936034 20.65 -1.58 ENDOGRO
AI949647 18.22 3.10 VEGF
AI949827 10.53 -1.63 bFGF
AI950109 FLJ 12604 37.36 1.35 ENDOGRO
AI968085 WNT5A 3.31 -1.63 ENDOGRO
AI972337 6.36 1.65 ENDOGRO
AI979166 3.20 2.29 ENDOGRO
AI982765 10.21 -1.29 bFGF
AI992251 7.09 -1.36 bFGF
AK000401 4.30 -1.30 ENDOGRO
AK000711 TAX1 BP1 12.69 -1.40 ENDOGRO AK000776 110.22 -1.47 ENDOGRO
AK000884 LRRFIP1 17.73 1.26 VEGF
AK000959 FLJ 10097 3.63 -1.19 ENDOGRO
AK001020 22.23 -1.37 bFGF
AK001362 3.02 -1.45 bFGF
AK001438 FBXL2 8.83 -1.33 VEGF
AK001560 LOC57863 16.74 -1.45 ENDOGRO
AK001630 126.78 1.38 VEGF
AK001872 PDL2 104.85 -1.24 bFGF
AK001903 11.23 -2.25 bFGF
AK001942 25.29 1.33 ENDOGRO
AK002195 6.46 1.31 VEGF
AL035306 STX12 4.35 -1.36 ENDOGRO
AL040051 6.17 -1.76 ENDOGRO
AL043980 PELI1 4.30 -1.28 ENDOGRO
AL049257 69.89 -1.59 ENDOGRO
AL049279 38.68 1.35 VEGF
AL049367 16.98 -1.38 VEGFE
AL049370 4.60 -1.99 ENDOGRO
AL049969 7.15 -1.70 ENDOGRO
AL049998 3.45 -1.17 ENDOGRO
AL050090 DKFZP586F1018 27.49 -1.25 ENDOGRO
AL050166 4.34 -1.37 ENDOGRO
AL110152 10.87 -1.20 ENDOGRO
AL110164 4.96 -1.52 ENDOGRO
AL110171 8.99 -1.30 VEGF
AL110202 4.31 -1.21 VEGF
AL110207 82.49 1.32 ENDOGRO
AL110255 4.30 -1.42 ENDOGRO
AL110280 7.38 -2.19 ENDOGRO
AL117427 4.11 -1.51 VEGF
AL117468 DKFZP586N1922 9.05 -1.51 ENDOGRO
AL117523 KIAA1053 15.24 -1.62 bFGF
AL117525 AKT3 32.46 -1.38 bFGF
AL117604 DLC1 15.23 -1.60 ENDOGRO
AL117615 DKFZP564D0764 257.68 -1.31 ENDOGRO
AL117617 4.04 1.32 bFGF
AL117664 DKFZP586L2024 117.73 -1.61 bFGF
AL122098 FLJ11937 3.98 -1.28 ENDOGRO
AL133118 30.13 1.63 VEGF
AL133596 12.02 -1.47 ENDOGRO
AL133605 PELI2 11.52 -1.71 bFGF
AL133640 DKFZp586C1021 4.67 -1.70 bFGF
AL133706 25.69 -1.39 VEGF
AL137540 NTN4 16.43 -1.86 ENDOGRO
AL137663 4.31 -1.46 bFGF
AL157431 DKFZp762A227 7.70 1.53 ENDOGRO AL157475 DKFZp761 G151 5.36 1.64 ENDOGRO AL157482 FLJ23399 7.50 -1.57 ENDOGRO AL157488 18.63 -1.45 VEGFE AL157502 MSTP032 5.99 1.36 VEGF AL359062 2726.41 -1.58 ENDOGRO AL572015 4.16 -1.81 bFGF AW015537 3.74 1.14 ENDOGRO AW015898 11.49 -1.28 VEGF AW023373 5.60 2.35 VEGF AW024527 3.55 1.44 ENDOGRO AW043571 FLJ20505 46.96 -1.63 ENDOGRO AW069166 CALD1 275.85 -1.68 ENDOGRO AW081929 KIAA1571 3.68 1.20 PIGF AW102613 3.22 1.66 VEGF AW 117242 32.57 1.42 VEGF AW 119059 4.57 -1.60 bFGF AW131552 8.49 2.99 VEGF AW 138207 FLJ22969 6.39 4.22 bFGF AW 139097 95.87 -1.21 ENDOGRO AW 139393 FTH1 37.66 1.63 ENDOGRO AW 139567 32.44 -1.70 ENDOGRO AW 139834 7.44 -1.43 VEGF AW151025 ARHE 7.93 -1.92 ENDOGRO AW 173150 4.05 1.55 ENDOGRO AW183161 42.71 -1.27 ENDOGRO AW 189467 3.79 -1.40 ENDOGRO
AW 190823 LOC58514 4.54 1.57 ENDOGRO
AW 195720 20.83 1.41 VEGF
AW237511 181.16 -2.84 VEGF
AW242009 4.21 1.42 ENDOGRO
AW243046 14.10 1.37 ENDOGRO
AW269515 FLJ20481 13.01 -1.61 ENDOGRO
AW269818 FLJ23144 7.49 -1.40 VEGF
AW271825 45.27 1.33 VEGF
AW274396 3.22 -1.11 ENDOGRO
AW274929 12.68 2.37 ENDOGRO
AW276078 24.62 1.61 bFGF
AW291331 16.15 -1.68 bFGF
AW291988 556.06 1.26 VEGF
AW292303 3.41 1.40 ENDOGRO
AW292755 8.16 1.28 VEGF
AW293366 3.66 -1.32 ENDOGRO
AW293770 1515.01 -1.50 ENDOGRO
AW294011 13.79 -1.41 ENDOGRO
AW294653 MOX2 20.80 1.68 VEGF
AW995919 PRG2 5.74 -1.26 ENDOGRO
BC005133 45.68 1.25 bFGF BM263824 7.39 1.48 ENDOGRO
D38522 KIAA0080 31.39 -1.47 ENDOGRO
D43636 KIAA0096 9.12 1.77 bFGF
D50406 RECK 60.03 -1.30 ENDOGRO
G26403 LOC64148 15.13 -1.30 ENDOGRO
H05089 FLJ 14033 1 1.27 -1.25 ENDOGRO
H09749 8.90 -1.78 bFGF
H11724 8.28 -1.21 ENDOGRO
H 16409 28.49 -1.22 ENDOGRO
H56091 5.97 -1.81 bFGF
H80726 5.53 1.57 VEGF
M 12758 HLA-A 4.93 -1.34 bFGF
M26383 IL8 91.08 1.30 VEGFE
M60721 HLX1 49.44 5.23 VEGFE
M68874 PLA2G4A 176.88 -1.18 ENDOGRO
M80783 TNFAIP1 3.66 -1.41 ENDOGRO
M90657 TM4SF1 17.14 1.26 ENDOGRO
M93718 N0S3 34.51 1.59 ENDOGRO
N36156 31.71 1.63 VEGF
N92500 4.02 1.16 bFGF
N92541 FLJ23462 7.63 -1.46 ENDOGRO
N95435 16.21 -1.73 bFGF
N99256 FLJ 11808 4.58 -1.29 VEGF
NM_000019 ACAT1 3.04 -1.25 PIGF
NM_000024 ADRB2 27.96 1.94 ENDOGRO
NM_000089 C0L1A2 16.78 -2.59 bFGF
NM_000093 C0L5A1 4.58 -1.39 ENDOGRO
NM_000109 DMD 3.44 -1.73 VEGF
NM_000115 EDNRB 5.52 1.43 ENDOGRO
NM_000124 ERCC6 5.51 -1.40 VEGF
NM_000138 FBN1 38.80 -1.48 ENDOGRO
NM_000143 FH 3.13 -1.53 ENDOGRO
NM_000165 GJA1 636.59 -1.50 ENDOGRO
NM_000170 GLDC 3.21 1.60 ENDOGRO
NM_000201 ICAM1 5.70 -2.41 bFGF
NM_000204 IF 3.96 1.42 ENDOGRO
NM_000210 ITGA6 15.87 2.14 VEGF
NM_000237 LPL 9.12 -1.95 bFGF
NM_000240 MAOA 5.87 -2.02 ENDOGRO
NM_000304 PMP22 13.23 1.22 ENDOGRO
NM_000361 THBD 5.73 1.56 bFGF
NM_000393 COL5A2 20.35 -1.51 ENDOGRO
NM_000416 IFNGR1 4.03 -1.30 ENDOGRO
NM_000428 LTBP2 90.66 -1.42 VEGF
NM_000436 OXCT 3.56 -1.46 ENDOGRO
NM_000439 PCSK1 76.58 1.35 ENDOGRO
NM_000441 SLC26A4 5.08 1.89 VEGF NM_000450 SELE 12.46 -3.51 bFGF
NM_000552 VWF 359.59 -1.26 bFGF
NM_000584 IL8 465.02 1.30 VEGFE
NM_000596 IGFBP1 17.07 1.50 bFGF
NM_000627 LTBP1 6.90 -1.14 ENDOGRO
NM_000689 ALDH1A1 133.54 -1.34 PIGF
NM_000784 CYP27A1 5.82 -1.35 ENDOGRO
NM_000793 DI02 5.13 -1.87 VEGF
NM_000820 GAS6 22.28 -1.37 bFGF
NM_000824 GLRB 27.89 -1.41 ENDOGRO
NM_000885 ITGA4 20.27 -2.03 bFGF
NM_000902 MME 7.99 1.44 bFGF
NM_000919 PAM 3.37 -1.29 ENDOGRO
NM_000929 PLA2G5 3.66 -1.78 bFGF
NM_000930 PLAT 15.88 1.87 bFGF
NM_000931 PLAT 17.20 1.90 bFGF
NM_000950 PRRG1 9.96 -1.48 ENDOGRO
NM_000958 PTGER4 4.64 1.54 ENDOGRO
NM_000963 PTGS2 233.39 -2.13 ENDOGRO
NM_001066 TNFRSF1 B 23.17 -1.38 VEGF
NM_001078 VCAM1 61.45 -7.79 ENDOGRO
NM_001122 ADFP 17.09 -1.32 VEGF
NM_001147 ANGPT2 9.05 3.98 VEGF
NM_001159 A0X1 5.24 1.68 ENDOGRO
NM_001165 BIRC3 3.66 -2.59 bFGF
NM_001166 BIRC2 4.58 -1.30 ENDOGRO
NM_001202 BMP4 14.66 -1.92 ENDOGRO
NMJD01223 CASP1 23.83 -1.37 bFGF
NM_001290 LDB2 63.23 1.33 VEGF
NM_001336 CTSZ 41.31 1.63 VEGFE
NM_001343 DAB2 381.87 -1.77 ENDOGRO
NM_001399 ED1 3.60 1.40 ENDOGRO
NM_001430 EPAS1 5.28 -1.25 ENDOGRO
NM_001442 FABP4 7.69 2.05 VEGF
NM_001444 FABP5 9.91 1.50 ENDOGRO
NM_001450 FHL2 4.24 -1.41 VEGF
NM_001457 FLNB 3.39 -1.46 bFGF
NM_001613 ACTA2 11.65 -1.39 VEGF
NM_001627 ALCAM 7.03 -1.98 ENDOGRO
NM_001674 ATF3 4.18 -3.56 ENDOGRO
N _001709 BDNF 357.01 -2.53 bFGF
NM_001711 BGN 3.05 -1.31 ENDOGRO
NM_001718 BMP6 66.96 -1.33 ENDOGRO
NM_001721 BMX 144.68 1.37 ENDOGRO
NM_001724 BPGM 13.36 -1.40 PIGF
NM_001773 CD34 310.68 -1.28 ENDOGRO
NM_001797 CDH11 17.51 1.32 bFGF NM_001839 CNN3 243.45 -1.39 ENDOGRO
NM_001845 COL4A1 20.78 -1.32 ENDOGRO
NM_001850 COL8A1 74.23 -1.60 ENDOGRO
NM_001885 CRYAB 5.53 -1.53 ENDOGRO
NM_001924 GADD45A 3.89 -1.46 bFGF
NM_001945 DTR 38.30 -1.94 bFGF
NM_001946 DUSP6 32.18 2.13 ENDOGRO
NM_001955 EDN1 52.88 -1.74 ENDOGRO
NM_001992 F2R 83.85 -1.62 ENDOGRO
NM_002006 FGF2 34.80 -1.55 ENDOGRO
NM_002019 FLT1 88.72 2.54 VEGF
NM_002053 GBP1 6.07 -1.94 ENDOGRO
NM_002131 HMGIY 5.99 1.70 ENDOGRO
NM_002133 HMOX1 3.75 1.38 ENDOGRO
NM_002153 HSD17B2 32.56 2.49 bFGF
NM_002165 ID1 6.91 1.60 bFGF
NM_002185 IL7R 38.68 -1.31 ENDOGRO
NM_002189 IL15RA 8.70 1.53 ENDOGRO
NM_002192 INHBA 8.93 -2.11 bFGF
NM_002210 ITGAV 4.70 -2.03 ENDOGRO
NM_002223 ITPR2 5.74 -1.46 ENDOGRO
NM_002313 ABLIM 5.35 -1.46 ENDOGRO
NM_002341 LTB 5.48 -1.17 bFGF
NM_002350 LYN 6.10 1.37 VEGF
NM_002397 MEF2C 3.31 2.13 VEGF
NM_002402 MEST 3.13 -1.61 VEGF
NM_002421 MMP1 18.54 2.29 VEGF
NM_002425 MMP10 12.05 1.63 VEGF
NM_002438 MRC1 34.54 -1.35 ENDOGRO
NM_002451 MTAP 40.27 1.48 bFGF
NM_002475 MYL1 3.19 -1.13 ENDOGRO
NM_002526 NT5 5.84 1.73 VEGF
NM_002575 SERPINB2 47.87 3.04 ENDOGRO
NM_002595 PCTK2 4.39 1.28 ENDOGRO
NM_002599 PDE2A 10.28 1.61 ENDOGRO
NM_002600 PDE4B 12.97 1.60 ENDOGRO
NM_002608 PDGFB 11.22 -1.47 bFGF
NM_002659 PLAUR 6.03 2.16 bFGF
NM_002662 PLD1 11.32 -1.42 ENDOGRO
NM_002763 PR0X1 6.17 1.69 ENDOGRO
NM_002837 PTPRB 102.95 -1.47 ENDOGRO
NM_002845 PTPRM 36.50 -1.27 bFGF
NM_002923 RGS2 12.21 2.02 ENDOGRO
NM_002933 RNASE1 25.36 -1.22 ENDOGRO
NM_002937 RNASE4 28.30 -1.49 ENDOGRO
NM_002982 SCYA2 10.53 -3.48 ENDOGRO
NM_002993 SCYB6 5.70 -2.66 bFGF NM_002996 SCYD1 14.41 -5.15 ENDOGRO
NM_003003 SEC14L1 5.78 -1.13 ENDOGRO
NM_003082 SNAPC1 4.43 -1.60 VEGF
NM_ 003113 SP100 4.98 -1.58 ENDOGRO
NM_003115 UAP1 9.18 -1.37 VEGF
NM_003186 TAGLN 36.78 -1.67 VEGF
NM_003199 TCF4 181.64 1.22 VEGFE
NM_003238 TGFB2 9.39 -2.64 bFGF
NM_003246 THBS1 78.99 -1.77 bFGF
NM_003266 TLR4 18.63 -1.44 ENDOGRO
NM_003289 TPM2 7.89 -1.32 ENDOGRO
NM_003330 TXNRD1 5.26 -1.74 bFGF
NMJD03459 SLC30A3 3.97 -1.73 bFGF
NM_003483 HMGIC 55.41 3.05 bFGF
NMJD03494 DYSF 84.07 1.48 VEGF
NM_003603 ARGBP2 11.04 -1.90 ENDOGRO
NM_003607 PK428 3.85 -1.24 ENDOGRO
NM_003633 ENC1 3.48 -1.89 VEGF
NM_003662 PIR 3.05 -1.25 VEGF
NM_003676 DEGS 5.23 1.07 ENDOGRO
NM_003693 SREC 232.16 1.34 ENDOGRO
NM_003706 PLA2G4C 13.06 -2.90 bFGF
NM_003798 CTNNAL1 4.07 -1.23 ENDOGRO
NM_003810 TNFSF10 3.21 -2.50 VEGFE
NM_003812 ADAM23 18.65 -1.34 VEGF
NM_003816 ADAM9 6.16 -1.36 ENDOGRO
NM_003914 CCNA1 10.33 1.31 ENDOGRO
NM_003919 SGCE 8.43 -1.17 ENDOGRO
NM_003947 HAPIP 18.68 1.65 bFGF
NM_003956 CH25H 7.43 -3.03 bFGF
NM_003965 CCRL2 8.41 2.00 bFGF
NM_003975 SH2D2A 3.60 1.33 bFGF
NM_003991 EDNRB 5.28 1.39 ENDOGRO
NM_004010 DMD 5.12 -1.90 bFGF
NM_004024 ATF3 4.83 -4.15 bFGF
NM_004105 EFEMP1 23.60 -1.39 ENDOGRO
NM_004126 GNG11 3428.47 1.47 ENDOGRO
NM_004155 SERPINB9 5.22 1.16 ENDOGRO
NM_004156 PPP2CB 3.27 -1.20 ENDOGRO
NM_004159 PSMB8 10.81 -1.19 bFGF
NM_004184 WARS 4.49 -1.89 bFGF
NM_004267 CHST2 86.99 2.83 bFGF
NM_004289 NFE2L3 6.65 -1.49 VEGF
NM_004334 BST1 165.62 -1.96 bFGF
NM_004342 CALD1 120.74 -1.64 ENDOGRO
NM_004385 CSPG2 14.18 -1.75 VEGF
NM_004414 DSCR1 7.74 2.13 VEGF NM_004417 DUSP1 5.71 1.53 bFGF
NM_004454 ETV5 16.90 2.73 ENDOGRO
NM_004490 GRB14 3.77 1.66 ENDOGRO
NM_004556 NFKBIE 4.86 -1.41 bFGF
NM_004609 TCF15 30.48 1.72 bFGF
NM_004675 ARHI 31.17 -1.47 ENDOGRO
NM_004791 ITGBL1 7.10 -1.40 ENDOGRO
NM_004796 NRXN3 5.25 -1.24 PIGF
NM_004808 NMT2 13.1 1 -1.73 bFGF
NM_004811 LPXN 3.02 1.36 ENDOGRO
NM_004877 GMFG 127.79 1.49 VEGF
NM_004881 PIG3 7.60 -1.15 ENDOGRO
NM_004895 C1 orf7 3.19 1.64 ENDOGRO
NM_005012 R0R1 62.08 -1.64 VEGF
NM_005045 RELN 10.98 -1.42 ENDOGRO
NM_005072 SLC12A4 10.32 -1.48 ENDOGRO
NM_005100 AKAP12 152.34 1.62 VEGF
NM_005118 TNFSF15 3.83 -1.64 VEGF
NM_005127 CLECSF2 16.36 1.73 bFGF
NM_005168 ARHE 6.11 -1.69 ENDOGRO
NM_005203 C0L13A1 3.85 1.31 ENDOGRO
NM_005238 ETS1 3.48 1.49 VEGF
NM_005282 GPR4 8.75 2.25 ENDOGRO
NM_005308 GPRK5 12.51 1.53 bFGF
NM_005360 MAF 5.11 -2.52 bFGF
NM_005420 STE 64.47 -2.17 bFGF
NM_005429 VEGFC 11.68 1.91 VEGF
NM_005438 F0SL1 3.17 1.71 ENDOGRO
NM_005460 SNCAIP 5.44 2.20 VEGF
NM_005541 INPP5D 40.84 1.40 ENDOGRO
NM_005556 KRT7 3.46 -1.17 ENDOGRO
NMJ305574 LM02 14.62 1.35 VEGF
NM_005585 MADH6 3.60 -1.38 VEGF
NM_005627 SGK 27.82 -1.17 ENDOGRO
NM_005630 SLC21A2 102.05 -1.39 ENDOGRO
NM_005711 EDIL3 10.36 -1.35 ENDOGRO
NM_005755 EBI3 3.54 -1.65 ENDOGRO
NM_005795 CALCRL 12.74 1.96 VEGF
NM_005953 MT2A 12.90 1.21 ENDOGRO
NM_005965 MYLK 3.93 -1.37 ENDOGRO
NM_006006 ZNF145 14.58 -2.00 bFGF
NM_006074 STAF50 74.18 -1.43 bFGF
NM_006094 DLC1 9.76 -1.69 ENDOGRO
NM_006100 ST3GALVI 26.24 1.42 VEGF
NM_006102 PGCP 8.16 -1.43 ENDOGRO
NM_006169 NNMT 530.16 -1.79 ENDOGRO
NM_006226 PLCE 13.52 1.64 ENDOGRO NM_006227 PLTP 4.50 -1.33 bFGF
NM_006255 PRKCH 4.77 -1.18 ENDOGRO
NM_006320 PMBP 3.38 -1.33 ENDOGRO
NM_006398 UBD 33.18 -5.18 bFGF
NM_006404 PROCR 52.74 1.20 bFGF
NM_006407 JWA 3.65 -1.19 ENDOGRO
NM_006454 MAD4 3.47 -1.39 ENDOGRO
NM_006457 LIM 6.95 -1.73 ENDOGRO
NM_006474 T1A-2 9.84 1.45 bFGF
NM_006475 OSF-2 870.46 -1.34 bFGF
NM_006509 RELB 4.29 -2.33 bFGF
NM_006528 TFPI2 11.72 3.00 bFGF
NM_006691 XLKD1 5.82 2.91 bFGF
NM_006719 ABLIM 6.08 -1.43 ENDOGRO
NM_006729 DIAPH2 5.11 -1.34 ENDOGRO
NM_006779 CEP2 4.35 1.32 ENDOGRO
NM_006834 RAB32 3.33 -1.38 VEGF
NM_006855 KDELR3 3.47 -1.40 bFGF
NM_006905 PSG1 3.03 -1.49 VEGF
NM_006988 ADAMTS1 24.94 2.05 ENDOGRO
NM_007005 BCE-1 3.54 -1.32 VEGF
NM_007021 DEPP 3.11 1.64 VEGF
NM_007034 DNAJB4 53.46 -1.28 VEGF
NM_007066 PKIG 4.23 1.33 ENDOGRO
NM_007283 HU-K5 14.74 -1.39 bFGF
NM_007288 MME 7.05 1.44 bFGF
NM_007289 MME 5.59 1.41 bFGF
NM_007308 SNCA 120.69 -1.16 ENDOGRO
NM_007361 NID2 22.18 3.23 VEGF
NM_009588 LTB 5.38 -1.19 bFGF
NM_012082 FOG2 10.62 -1.25 ENDOGRO
NM_012250 TC21 9.43 -1.29 VEGF
NM_012269 HYAL4 5.99 -2.44 bFGF
NM_012323 MAFF 3.56 1.25 VEGF
NM_012449 STEAP 301.60 1.45 ENDOGRO
NM_013231 FLRT2 1584.59 -1.71 ENDOGRO
NM_013250 ZNF215 4.12 2.11 ENDOGRO
NM_013352 SART-2 7.17 -1.93 bFGF
NM_013372 CKTSF1 B1 19.71 1.44 bFGF
NM_013423 ARHGAP6 8.68 2.49 VEGF
NM_013956 NRG1 29.07 -1.81 VEGF
NM_013957 NRG1 8.43 -1.74 VEGF
NM_013958 NRG1 13.34 -1.44 VEGF
NM_013961 NRG1 3.70 -1.37 VEGF
NM_013962 NRG1 20.39 -1.38 VEGF
NM_013989 DI02 7.84 -2.07 VEGF
NM_014029 HSPC022 32.99 1.25 ENDOGRO NM_014059 RGC32 28.92 -1.65 ENDOGRO NM_014074 PRO0529 4.39 -1.42 VEGF NM_014143 B7-H1 9.24 -1.30 VEGF NM_014331 SLC7A11 16.15 -1.61 ENDOGRO NM_014344 FJX1 7.64 1.39 ENDOGRO NM_014349 APOL3 11.47 -1.43 bFGF NM_014363 SACS 13.77 -1.44 VEGFE NM_014391 CARP 46.40 -1.80 ENDOGRO NM_014397 NEK6 3.95 -1.28 ENDOGRO NM_014398 LAMP3 15.90 -1.35 ENDOGRO NM_014465 ST1 B2 39.94 -1.84 ENDOGRO NM_014476 ALP 15.82 -1.19 ENDOGRO NM_014521 SH3BP4 3.55 -1.52 ENDOGRO NM_014570 ARFGAP1 3.13 -1.17 ENDOGRO NM_014585 SLC11A3 46.27 -1.41 VEGFE NM_014634 KIAA0015 7.30 1.52 bFGF NM_014686 KIAA0355 3.24 -1.27 ENDOGRO NMJD14705 KIAA0716 87.07 1.36 ENDOGRO NM_014721 KIAA0680 3.70 1.43 VEGF NM_014731 KIAA0552 3.25 -1.79 ENDOGRO NM_014737 RASSF2 23.65 1.40 ENDOGRO NM_014782 KIAA0512 22.06 -1.38 ENDOGRO NM_014795 ZFHX1 B 158.26 -1.36 VEGF NM_014822 SEC24D 3.59 -1.32 ENDOGRO NM_014832 KIAA0603 3.75 1.56 VEGF NM_014840 KIAA0537 7.79 -1.77 ENDOGRO NM_014890 D0C1 12.99 -2.25 ENDOGRO NM_014909 KIAA1036 5.45 1.42 VEGF NM_014933 KIAA0905 3.78 -1.20 ENDOGRO NM_014945 KIAA0843 4.21 1.20 ENDOGRO NM_014959 KIAA0955 7.96 1.84 ENDOGRO NM_014965 KIAA1042 4.04 -1.44 bFGF NM_015376 KIAA0846 9.01 2.98 VEGF NM_015675 GADD45B 3.51 -1.94 ENDOGRO NM_015881 DKK3 286.01 -1.24 ENDOGRO NM_016061 LOC51646 3.01 -1.64 ENDOGRO NM_016109 PGAR 31.81 -1.99 VEGF NM_016134 LOC51670 4.61 -1.36 ENDOGRO NM_016203 PRKAG2 5.09 -1.22 VEGF NM_016232 IL1 RL1 15.56 -1.40 VEGF NM_016235 GPRC5B 5.37 -1.53 bFGF NM_016270 KLF2 5.53 2.17 bFGF NM_016274 LOC51177 5.28 -1.48 ENDOGRO NM_016352 CPA4 54.01 -1.55 ENDOGRO NM_016357 EPLIN 4.98 -1.30 VEGF NM_016385 HSPC057 4.00 -1.40 ENDOGRO NM 016602 GPR2 4.94 -1.29 ENDOGRO NM_016619 LOC51316 5.50 -1.38 ENDOGRO
NM_016848 SHC3 41.19 -1.31 VEGF
NM_016931 NOX4 80.07 1.17 ENDOGRO
NM_017415 KLHL3 40.42 1.72 ENDOGRO
NM_017577 DKFZp434C0328 3.90 -1.35 ENDOGRO
NM_017585 SLC2A6 8.27 -1.47 ENDOGRO
NM_017596 KIAA0449 3.45 1.41 ENDOGRO
NM_017718 FLJ20220 5.02 1.32 ENDOGRO
NM_017734 FLJ20271 124.25 -1.32 ENDOGRO
NM_017752 FLJ20298 6.73 -1.46 ENDOGRO
NM_017805 FLJ20401 41.04 1.27 ENDOGRO
NM_017905 FLJ20623 3.11 -1.30 ENDOGRO
NM_017980 FLJ 10044 125.20 -1.42 ENDOGRO
NM_018004 FLJ10134 17.08 -1.46 bFGF
NM_018012 FLJ 10157 3.96 -1.59 VEGF
NM_018057 FLJ10316 8.37 -1.52 ENDOGRO
NM_018071 FLJ 10357 3.71 -1.59 bFGF
NM_018159 FLJ 10628 6.29 1.17 ENDOGRO
NM_018192 FLJ10718 154.28 -2.02 bFGF
NM_018295 FLJ 11000 4.10 -1.43 ENDOGRO
NM_018324 FLJ11106 5.31 -1.97 bFGF
NM_018326 FLJ11110 64.14 1.66 bFGF
NM_018357 FLJ11196 4.28 -1.34 ENDOGRO
NM_018370 FLJ 11259 6.87 -1.49 ENDOGRO
NM_018384 FLJ 11296 11.26 2.13 bFGF
NM_018401 HSA250839 3.61 1.19 bFGF
NM_018413 C4ST 3.30 1.51 ENDOGRO
NM_018476 HBEX2 106.48 -1.62 VEGF
NM_018482 DDEF1 3.45 -1.19 ENDOGRO
NM_018567 TNS 5.72 1.43 ENDOGRO
NM_018841 LOC55970 12.04 -1.33 ENDOGRO
NM_019858 A 14.27 1.21 ENDOGRO
NM_020130 C8orf4 22.69 -3.60 ENDOGRO
NM_020152 C21orf7 4.33 1.87 VEGF
NM_020163 LOC56920 30.25 -5.28 VEGFE
NM_020186 DC11 25.99 1.58 ENDOGRO
NM_020190 HNOEL-iso 3.76 -1.55 bFGF
NM_020353 LOC57088 426.58 -1.52 VEGF
NM_020651 PELI1 3.43 -1.38 bFGF
NM_021069 ARGBP2 192.99 -2.09 ENDOGRO
NM_021106 RGS3 9.55 -2.34 ENDOGRO
NM_021154 PSA 5.38 1.61 ENDOGRO
NM_021226 LOC58504 6.03 1.38 ENDOGRO
NM_021255 PELI2 6.20 -1.77 bFGF
R49042 16.17 -1.54 bFGF
R92031 6.32 1.33 bFGF
T57773 4.79 -2.27 VEGF T81424 K-ALPHA-1 12.52 1.45 ENDOGRO
T87544 4.04 -1.40 ENDOGRO
T89094 RGS4 1156.70 1.71 ENDOGRO
U10991 G2 205.85 1.46 ENDOGRO
U 17077 BENE 12.42 2.43 VEGF
U27655 RGS3 11.59 -2.23 bFGF
U27768 RGS4 102.15 1.68 ENDOGRO
U50534 13CDNA73 14.49 1.54 bFGF
U61166 ITSN1 4.45 -1.56 ENDOGRO
U79271 SDCCAG8 20.19 -1.33 ENDOGRO
U90908 LOC58504 7.01 1.30 ENDOGRO
W02693 5.01 -1.87 ENDOGRO
W44435 FLJ 12649 6.30 -1.65 ENDOGRO
W46280 5.82 -1.65 ENDOGRO
W46364 630.21 -1.92 ENDOGRO
W60844 6.45 -1.30 ENDOGRO
W69778 32.69 -1.64 bFGF
W87772 472.27 1.26 VEGFE
X04706 H0XD4 38.81 1.55 ENDOGRO
X05610 COL4A2 83.40 -1.35 ENDOGRO
X66945 FGFR1 4.04 1.20 ENDOGRO
X68742 ITGA1 4.63 1.73 VEGF
X93921 DUSP7 6.85 1.34 ENDOGRO
Table. 4
RHMVEC Proliferation Signature
GenBank
Accession EC:MCF7 Intensity Growth
Number Gene Symbol (Expression) Ratio Fold Change Factor
600507553R1 600507553R1 6.40 -1.56 VEGF
600511339R1 600511339R1 10.95 -1.65 ENDOGRO
600513062R1 600513062R1 11.30 -1.84 VEGF
600516127R1 600516127R1 20.18 -1.97 VEGF
600516223R1 600516223R1 3.14 -1.25 bFGF
600518689R1 600518689R1 19.16 -1.92 VEGF
600523987R1 600523987R1 3.36 1.54 VEGF
600524312R1 600524312R1 9.60 -1.78 bFGF
700039220H1 700039220H1 3.64 1.31 bFGF
700067654H1 700067654H1 5.16 1.33 VEGF
700588837H1 700588837H1 3.28 -1.58 ENDOGRO
700690490H1 700690490H1 5.36 1.29 VEGF
701347738H1 701347738H1 4.31 -1.55 ENDOGRO
701349191 H1 701349191 H1 5.30 -1.50 bFGF
701350288H1 701350288H1 8.43 1.20 ENDOGRO
701353618H1 701353618H1 5.33 1.28 VEGF
701354577H1 701354577H1 5.05 1.49 VEGF
701354657H1 701354657H1 3.71 1.27 VEGF
701417958H1 701417958H1 3.13 1.20 bFGF
701419627H1 701419627H1 15.42 -1.76 ENDOGRO
AA799503 g2862458 4.90 -1.23 ENDOGRO
AA799750 Erg 482.85 -1.31 bFGF
AA800192 g2863147 4.60 -1.49 bFGF
AA800293 g2863248 104.39 -1.37 bFGF
AA800550 g2863505 39.16 1.20 VEGF
AA801220 g2864175 222.44 1.25 ENDOGRO
AA848714 g2936254 9.08 -1.26 bFGF
AA850055 g2937595 4.18 -1.26 bFGF
AA850311 g2937851 3.44 1.22 VEGF
AA851637 Lu 42.24 -1.51 ENDOGRO
AA859260 g2948611 3.32 -1.34 VEGF
AA859278 g2948629 4.10 1.36 bFGF
AA859444 g2947975 10.74 -1.73 bFGF
AA874964 g2979912 5.37 -1.33 VEGF
AA875261 g2980209 8.23 1.41 bFGF
AA891911 g3018790 9.75 1.35 bFGF
AA899923 g3035277 3.48 -1.44 bFGF
AA925057 g3072193 25.13 -1.36 VEGF AA926129 74.21 -1.32 bFGF
AA944413 g3104329 3.96 1.21 VEGF
AA944542 g4132423 3.97 -1.23 bFGF
AA944936 g3104852 6.51 1.72 bFGF
AA945463 g3105379 3.61 -1.45 VEGF
AA945677 g3105593 29.07 -1.25 ENDOGRO
AA945788 g3105704 5.63 -1.20 VEGF
AA946190 g3106106 14.83 -1.28 VEGF
AA946201 g3106117 102.57 -1.25 ENDOGRO
AA946350 g3106266 80.11 1.45 bFGF
AA955134 g3513034 3.01 1.30 ENDOGRO
AA956085 g3119780 3.61 -1.23 bFGF
AA957335 g3121030 158.89 -1.55 bFGF
AA957776 g2936580 38.11 -1.18 bFGF
AA963106 g3136598 7.68 1.47 bFGF
AA964004 Pter 4.69 1.35 ENDOGRO
AA964264 g3137756 14.05 -1.20 bFGF
AA996897 g3187452 23.71 -1.24 ENDOGRO
AA997073 g3187934 5.63 -1.21 VEGF
AA998510 g3189161 26.21 -1.78 ENDOGRO
AA998516 g3189167 5.07 1.19 VEGF
AA998618 g3189269 3.48 -1.48 VEGF
AA999079 g3189670 3.67 -1.18 VEGF
AB000199 cca2 3.23 -1.31 VEGF
AB005540 PCTAIRE2 3.66 1.20 bFGF
AB010467 Abcc3 4.40 -1.28 VEGF
AB015308 Gna15 19.47 1.21 bFGF
AB015746 Il4r 22.40 -1.17 bFGF
AB019120 AB019120 67.88 -1.55 bFGF
AB020978 GADD45gamma 3.19 1.27 bFGF
AB032085 RM3 4.60 -1.28 bFGF
AB032087 g3730102 3.17 -1.18 ENDOGRO
AB060092 Scyb2 7.67 -1.76 ENDOGRO
AF003835 Idi1 3.42 1.32 ENDOGRO
AF021350 NKG2A 3.04 1.64 bFGF
AF029241 RT1.S3 5.62 -1.32 bFGF
AF047707 Ugcg 3.82 -1.65 VEGF
AF072816 mrp3 5.04 -1.30 bFGF
AF102262 beta1 -4GT 3.91 -1.25 VEGF
AF154245 chemotactic protein-3 5.09 -1.32 bFGF
AF164039 AF164039 18.79 -1.31 bFGF
AF189709 collagen XVIII 14.86 -1.25 ENDOGRO interferon-gamma
AF201901 receptor 4.50 -1.50 bFGF AF205717 LRTM4 8.60 1.20 VEGF AF244366 FLIP short form 16.14 1.32 bFGF AF254801 AF254801 69.37 1.29 bFGF AF259504 Bak 4.27 -1.27 VEGF
AF259898 E3karp 10.92 -1.69 VEGF
AF271786 Fgf13 11.13 1.31 VEGF
AF314657 clusterin 10.00 -1.43 ENDOGRO
AF324255 Erol l 3.40 -1.66 bFGF
AF368269 Cyp2t1 19.99 -1.38 ENDOGRO
AI008035 700068780H1 9.35 1.32 bFGF
AI008526 g3222358 16.48 2.48 bFGF
AI009368 g3223200 8.25 -1.34 VEGF
AI009736 g3223568 6.35 1.32 bFGF
AI009780 g3223612 3.02 1.23 bFGF
AI009783 g3223615 3.33 -1.32 ENDOGRO
AI010312 g4133226 31.15 -1.34 bFGF
AI011501 g3225333 17.92 -1.20 bFGF
AI012580 g3226412 10.77 -1.22 bFGF
AI012597 g3226429 13.23 1.19 bFGF
AI013470 g4133876 3.54 -1.48 ENDOGRO
AI013562 g3227618 7.03 -3.34 VEGF
AI029460 g3247286 5.84 -1.40 bFGF
AI031004 g3248830 80.92 -1.39 bFGF
AI043630 g3290365 13.96 1.23 bFGF
AI043724 g3290459 4.08 1.22 ENDOGRO
AI043851 g3290586 210.09 -1.35 bFGF
AI043958 g3290693 4.08 -1.20 VEGF
AI044026 g3290761 6.81 -1.34 bFGF
AI044530 g3291391 8.51 -2.18 bFGF
AI044674 g3291535 5.62 -1.49 VEGF
AI044760 g3291621 26.81 1.43 VEGF
AI044802 g3290865 4.52 -2.03 ENDOGRO
AI044912 g3291731 33.22 1.40 ENDOGRO
AI044948 g3291767 10.36 1.36 bFGF
AI045186 g3292005 21.40 -1.52 ENDOGRO
AI045920 g3292739 5.07 1.24 ENDOGRO
AI058759 g3332536 25.51 1.23 bFGF
AI059060 g3332837 3.24 -1.62 VEGF
AI059103 g3332880 14.45 -1.36 VEGF
AI059204 g3332981 55.91 -1.44 VEGF
AI059363 g3333140 124.38 1.40 bFGF
AI059449 g3333226 7.39 -1.34 bFGF
AI059450 g3333227 8.45 -1.69 VEGF
AI060115 g3333892 66.25 -1.25 VEGF
AI070068 g3396319 6.13 -2.29 bFGF
AI070370 g3396621 3.50 -1.23 ENDOGRO
AI072357 g3398551 7.13 1.22 ENDOGRO
AH 01062 g3706050 4.44 -1.91 VEGF
AH 01250 g4133997 4.27 1.45 bFGF
AH 01270 g4134000 87.91 -1.28 bFGF AH 01402 g3706309 3.52 1.18 bFGF AI101757 g3706619 6.05 -1.20 bFGF AI101945 g3706786 6.82 -1.26 VEGF AH 02248 g4134070 13.24 -1.22 VEGF AH 02320 g3707114 3.90 -1.25 bFGF AH 03007 g3704802 3.47 -1.29 bFGF AH03106 g3704827 3.90 1.47 bFGF AH03618 g3708145 3.01 -1.36 VEGF AH03939 g3704876 4.41 1.22 ENDOGRO AH 04128 g3708534 3.38 -1.36 VEGF AH05417 g3709501 45.14 -1.30 VEGF AH05452 g3709529 3.95 -1.44 VEGF AH 12636 g3512585 5.56 -1.25 bFGF AH 37629 g3638406 18.18 1.41 bFGF AH 37826 g3638603 3.28 -1.33 ENDOGRO AH37944 g3638721 222.74 -1.76 bFGF AH45002 g3666801 23.08 1.23 VEGF AH45832 g3667631 6.35 -1.23 bFGF AH68952 g3705260 5.36 1.21 VEGF AH69422 g3705730 10.92 -1.37 VEGF AH69635 g3709675 15.87 -1.21 ENDOGRO AI171908 g3711948 3.49 -1.23 ENDOGRO AH72056 g4134696 16.18 1.34 VEGF AH 72117 g4134703 10.06 -1.32 ENDOGRO AH 75466 g3726104 6.20 -1.45 bFGF AH 76486 g3727124 146.17 1.31 bFGF AH76965 g4133497 82.43 -1.88 bFGF AH76983 g3727621 3.19 1.26 bFGF AH77055 g3727693 19.15 1.29 VEGF AI177120 g3829605 4.30 -1.39 bFGF AI177198 g3727836 4.37 1.16 VEGF AH77396 g3728034 3.05 -1.62 bFGF AH77621 g3728259 264.10 -1.28 bFGF AH77939 g4135031 36.48 -1.32 VEGF AH78222 g3728860 7.21 1.23 ENDOGRO AH 78718 g4135092 114.85 1.41 bFGF AH78978 g3729616 7.39 -1.25 ENDOGRO AH79786 g3730424 6.81 -1.33 VEGF AH80386 g3731024 21.30 -1.50 VEGF AI230758 g3814645 89.23 -1.24 ENDOGRO AI230762 g3814649 3.23 -1.25 VEGF AI230918 g3814805 3.00 -1.24 bFGF AI231805 g3815685 43.95 -1.18 ENDOGRO AI231999 g3815879 22.28 1.31 ENDOGRO AI233099 g3816979 3.19 1.31 VEGF AI233773 Mawbp 68.74 -1.27 bFGF AI235721 g3829227 3.22 -1.32 VEGF AI235960 g3829466 229.73 -1.23 VEGF
AI236381 g3829887 4.52 -2.09 bFGF
AI236799 g4136246 25.50 1.32 bFGF
AI236912 Nab1 7.23 1.24 bFGF
AI237544 g3831050 4.46 -1.28 bFGF
AI407547 g3727827 3.84 -1.17 bFGF
AI409186 g2938420 31.41 1.29 ENDOGRO
AI409841 g3706639 3.36 -1.26 VEGF
AI411054 g3812200 3.37 1.46 bFGF
AI711100 g3224246 385.26 -1.29 VEGF
AJ000696 KIF1D 4.52 -2.61 bFGF
AJ011116 nos3 58.27 -1.31 VEGF
AW140657 600510887R1 6.53 -1.44 ENDOGRO
AW142011 g3333577 3.20 -1.35 bFGF
AW142194 g2864225 41.07 1.23 VEGF
AW142519 g3071124 3.12 -1.39 bFGF
AW523549 g3224204 17.12 -1.22 bFGF
AW914004 g3224058 306.81 1.35 VEGF
AW916926 Slc22a7 13.17 -1.37 ENDOGRO
AW917188 Dpyd 9.05 1.63 VEGF
AW920825 701222534H1 32.82 1.57 VEGF
AY024364 GATA-3 4.64 -1.24 bFGF
BE098266 g3071239 323.96 -1.21 bFGF
BE108269 g3246659 6.26 1.34 ENDOGRO
BE121287 700032038H1 4.23 -1.16 ENDOGRO
BF283084 600520366R1 5.93 1.27 VEGF
BF398271 g3292264 7.25 -1.36 bFGF
BF416417 g3707631 4.83 -1.27 bFGF
BF548232 g3828538 3.62 -1.58 VEGF
BF551997 BF551997 21.20 1.33 bFGF
BG381698 g2672938 4.40 1.20 bFGF
BG664717 701222952H1 12.84 1.54 VEGF
BI275290 g3020546 3.50 -1.26 bFGF
BI277635 g3728852 9.73 -1.39 VEGF
BI283830 g3817681 9.65 -1.28 ENDOGRO
BI284263 g3137012 511.46 -1.25 bFGF
BI285246 g3830698 61.06 1.22 ENDOGRO
BI287221 g3705125 32.89 -1.24 VEGF
BI294910 g3705813 43.33 -1.51 ENDOGRO
BI296015 g3711886 5.26 -1.37 VEGF
BM384585 g2862699 6.07 -1.31 ENDOGRO
BM384701 g3224341 20.20 -1.42 VEGF
BM388598 g3187570 6.05 -1.56 bFGF
BM388852 g3106350 28.05 -1.19 bFGF
BM391182 600518660R1 4.10 -1.28 VEGF
BQ190671 g4132974 3.42 1.21 VEGF
BQ192029 g4133216 376.26 -1.31 bFGF BQ198730 g3224083 39.70 1.24 VEGF
BQ199612 g3727571 29.44 -1.43 bFGF
BQ199678 g3818042 3.96 -1.35 bFGF
BQ200399 g3727330 128.03 1.21 bFGF
BQ203036 g2937343 10.46 1.20 VEGF
BQ203060 g3221834 19.92 -1.27 bFGF
BQ203246 g3102732 188.91 1.28 bFGF
BQ204980 g3222960 4.72 1.37 VEGF
BQ205927 g3103480 7.39 -1.25 bFGF
BQ779673 700052535F1 3.36 -1.43 bFGF
BQ780778 g2937314 29.44 -1.23 bFGF
BQ781420 g2936107 3.31 1.36 bFGF
BU671151 3.04 1.28 VEGF
BU671574 701216766H1 3.51 1.44 VEGF
CA333942 600524307R1 6.48 -1.57 VEGF
CA503512 g4134955 3.09 -1.30 VEGF
CA504040 g3709254 3.01 1.29 VEGF
CA506715 g2938431 102.56 1.24 bFGF
CA507008 g976837 3.86 -1.36 ENDOGRO
CA508330 g3813525 3.00 -1.25 VEGF
CA509105 600513095R1 3.10 -1.21 bFGF
CA509955 g3730145 37.61 1.42 bFGF
CA513003 701353736H1 3.92 1.23 bFGF
D00636 b5R 3.13 -1.26 bFGF
D11444 gro 3.41 -2.00 bFGF
D16339 g6981681 10.14 1.33 ENDOGRO
D16339 Ttpa 15.43 1.53 bFGF
D28860 D28860 3.78 -3.13 VEGF
D31838 weel 4.16 1.34 VEGF
D42148 600520458R1 94.76 -1.47. bFGF
D86086 Abcc2 5.88 1.21 bFGF
G2887744 9.40 -1.24 VEGF
G2887885 3.21 -1.58 ENDOGRO
G2888124 16.17 -1.70 ENDOGRO
G2937254 3.04 -1.36 bFGF
G2937336 3.11 -1.48 ENDOGRO
G2938797 6.83 1.24 bFGF
G2938805 3.65 -1.21 bFGF
G2938831 3.04 -1.33 bFGF
G2939578 6.47 -1.68 ENDOGRO
G3019428 39.84 1.20 VEGF
G3020059 7.27 -1.29 bFGF
G3021176 6.67 -1.33 bFGF
G3035644 5.53 1.32 bFGF
G3071714 3.04 -1.46 ENDOGRO
G3071902 167.52 -1.28 bFGF
G3072603 8.46 -1.26 bFGF G3072712 56.89 1.22 ENDOGRO
G3073258 7.51 1.54 bFGF
G3102686 9.14 -1.38 bFGF
G3103279 34.67 1.38 ENDOGRO
G3137338 5.03 -1.53 VEGF
G3137782 22.69 1.33 VEGF
G3137957 273.68 -1.34 bFGF
G3137994 8.42 -1.34 bFGF
G3138006 6.05 -1.25 bFGF
G3189628 3.21 1.35 bFGF
G3224642 3.55 -1.69 VEGF
G3225906 14.76 1.21 bFGF
G3226018 5.47 -1.21 bFGF
G3226140 98.14 -1.31 bFGF
G3227787 13.27 -1.25 VEGF
G3246847 3.40 -1.48 bFGF
G3246942 135.33 -1.52 ENDOGRO
G3247784 14.84 -1.31 VEGF
G3247854 199.84 -1.30 bFGF
G3248097 26.74 1.35 bFGF
G3248367 21.02 -1.88 bFGF
G3291935 5.77 -1.46 ENDOGRO
G3291949 3.22 -1.16 VEGF
G3292531 18.79 -1.34 ENDOGRO
G3292629 3.80 -1.27 VEGF
G3333900 8.95 -1.34 ENDOGRO
G3333935 5.10 -1.21 bFGF
G3396358 3.87 -1.40 bFGF
G3396493 3.05 -1.32 VEGF
G3396557 26.97 -1.36 bFGF
G3396633 8.07 -1.45 bFGF
G3397437 304.78 -1.22 VEGF
G3397680 3.38 1.24 ENDOGRO
G3397969 9.69 -1.63 VEGF
G3398076 61.74 -1.60 bFGF
G3398171 4.81 -1.20 ENDOGRO
G3399145 22.81 -1.58 bFGF
G3399177 4.24 1.34 VEGF
G3399284 11.67 1.21 VEGF
G3399406 13.82 1.54 bFGF
G3511710 5.60 -1.27 VEGF
G3512937 31.87 -1.40 VEGF
G3513230 4.55 -1.21 bFGF
G3636910 5.68 1.20 VEGF
G3637432 14.94 -1.28 VEGF
G3637746 6.69 1.20 bFGF
G3638675 36.35 -1.27 VEGF G3666728 3.09 -1.26 bFGF G3666899 64.34 -1.48 bFGF G3667173 8.07 -1.28 bFGF
G3707946 3.06 -1.27 bFGF
G3708389 17.55 -2.00 bFGF
G3708633 4.16 -1.35 VEGF
G3708986 95.05 -1.44 bFGF
G3709440 3.09 1.18 bFGF
G3710527 6.48 1.22 bFGF
G3710770 5.18 -1.19 bFGF
G3711240 8.06 -2.85 ENDOGRO
G3711421 22.68 -1.76 bFGF
G3711520 26.51 1.39 VEGF
G3711533 5.04 -1.39 bFGF
G3711566 31.85 1.34 ENDOGRO
G3712171 4.18 1.37 VEGF
G3725993 3.63 -1.22 ENDOGRO
G3726061 3.64 -1.16 ENDOGRO
G3727230 14.55 -1.37 VEGF
G3729898 1566.10 1.38 bFGF
G3730814 6.28 -2.55 bFGF
G3811611 6.82 -1.27 bFGF
G3812445 3.50 -1.28 bFGF
G3813207 398.22 -1.31 VEGF
G3813483 8.65 -1.50 ENDOGRO
G3829163 3.61 -1.32 bFGF
G4131670 3.34 1.24 VEGF
G4131679 5.44 -1.24 bFGF
G4131762 4.42 -1.46 ENDOGRO
G4132317 47.79 1.22 VEGF
G4132471 3.75 -1.22 bFGF
G915019 3.97 -1.30 VEGF
G976993 11.77 1.50 VEGF
G977252 3.56 -1.74 bFGF
G977371 9.43 -1.28 ENDOGRO
G977384 3.01 1.66 VEGF
G977468 3.82 -1.41 bFGF
G977854 5.07 -1.42 bFGF
G977919 5.12 -1.31 bFGF
G979053 10.84 -1.41 bFGF
H32317 g977734 3.08 1.32 VEGF
H32799 g978216 3.31 -1.65 VEGF
H34328 g979745 17.72 1.76 bFGF
H34385 g979802 7.34 -1.19 bFGF
H34603 g980020 4.26 1.43 VEGF
H34681 g980098 5.73 1.38 VEGF
J03637 AD mRNA 4.24 -2.80 bFGF J03819 erb62 7.20 1.86 VEGF
L11995 cyclin B 3.64 1.22 VEGF
L20468 cerebroglycan 31.80 1.30 ENDOGRO
L22294 PDH 4.81 -1.47 ENDOGRO
L23128 L23128 3.23 1.20 bFGF
L27651 Slc22a7 7.71 -1.34 ENDOGRO
L34049 megalin 4.23 1.27 bFGF
M17412 LOC60380 4.02 -1.73 ENDOGRO
M26125 XEH mRNA 223.02 -1.53 ENDOGRO
M58040 transferrin receptor 3.87 1.28 bFGF
M80367 Gbp2 9.84 -1.62 bFGF
M81855 Abcbl 9.60 -1.79 bFGF
M83143 M83143 3.03 1.31 bFGF
M91235 M91235 11.54 -1.94 ENDOGRO
NM_012502 Ar 8.93 2.18 bFGF
NM_012528 Chrnbl 25.35 -1.31 bFGF
NM_012548 Edn1 392.42 -2.23 bFGF
NM_012566 Gfi1 14.50 1.39 bFGF
NM_012620 Serpinel 54.90 -2.51 ENDOGRO
NM_012679 Clu 3.34 -1.30 ENDOGRO
NM_012715 Adm 9.14 -1.97 ENDOGRO
NM_012762 Caspl 5.76 -1.26 bFGF
NM_012827 Bmp4 80.49 -1.44 bFGF
NM_012912 Atf3 4.89 -1.43 bFGF
NM_013000 Pam 7.26 1.40 bFGF
NM_013062 Kdr 405.26 1.34 bFGF
NM_013085 Plau 9.54 2.20 bFGF
NM_013130 Madhl 6.04 -1.20 ENDOGRO
NM_013145 Gnail 14.79 1.48 bFGF
NM_013151 Plat 5.26 -1.38 ENDOGRO
NM_016987 Acly 3.21 -1.28 VEGF
NM_017028 Mx1 14.33 -1.79 bFGF
NM_017076 Tage4 10.54 1.78 bFGF
NM_017079 Cd1 d 26.25 -1.30 bFGF
NM_017105 Bmp3 19.82 -2.20 ENDOGRO
NM_017112 Hpn 3.02 -1.91 VEGF
NM_017225 Pctp 5.87 1.30 bFGF
NM_017259 Btg2 26.14 -1.27 ENDOGRO
NM_017317 ram 4.89 -1.51 bFGF
NM_017350 Plaur 7.45 1.31 bFGF
NM_019144 Acp5 223.88 -1.54 ENDOGRO
NM_019147 Jag1 65.16 -1.46 VEGF
NM_019234 Dncid 32.61 -1.51 bFGF
NM_019249 Ptprf 22.72 -1.20 ENDOGRO
NM_019261 Klrc2 11.07 1.78 bFGF
NM_019285 Adcy4 15.84 -1.18 bFGF
NM_019370 LOC54410 34.00 1.25 VEGF NM_019371 SM-20 7.13 -1.61 ENDOGRO
NM_020072 Ppal 3.37 1.51 VEGF
NM_020082 Rnase4 25.93 1.41 VEGF
NM_021655 Chga 3.64 1.28 VEGF
NM_021679 Nxph3 6.16 1.28 ENDOGRO
NM_021751 LOC60357 140.84 -1.40 bFGF
NM_021846 Mcl1 8.92 1.36 VEGF
NM_022005 Fxyd6 812.43 -1.31 bFGF
NM_022177 Sdf1 9.97 -1.68 VEGF
NM_022224 Rpr1 3.40 1.38 bFGF
NM_022297 Ddahl 4.46 -1.28 VEGF
NM_022441 AcvrH 61.21 -1.34 bFGF
NM_022528 Hif3a 3.50 -1.47 bFGF
NM_022705 Mch 12.79 -1.27 bFGF
NM_022715 Mvp 3.80 1.29 VEGF
NM_022856 Nab1 4.04 1.23 bFGF
NM_023103 Mug1 3.24 1.26 ENDOGRO
NM_023960 Kcnmb4 5.42 1.43 VEGF
NM_024160 Cyba 44.10 -1.34 bFGF
NM_030834 Mct3 27.50 -1.81 ENDOGRO
NM_030868 Nov 193.60 1.20 ENDOGRO
NM_030985 Agtrla 33.27 2.02 VEGF
NM_031059 Msx1 4.48 -1.40 bFGF
NM_031100 RpHO 4.31 -1.32 VEGF
NM_031242 Cds1 10.30 -1.37 bFGF
NM_031321 Slit3 5.51 -1.58 ENDOGRO
NM_031327 Cyr61 24.10 -1.65 ENDOGRO
NM_031544 Ampd3 24.58 -1.42 bFGF
NM_031550 Cdkn2a 7.80 1.40 bFGF
NM_031645 Rampl 4.16 -1.47 ENDOGRO
NM_031646 Ramp2 14.25 -1.50 VEGF
NM_031771 Thbd 14.40 1.38 bFGF
NM_031807 Tpbg 66.41 -1.25 bFGF
NM_031970 Hspbl 52.18 -1.36 bFGF
NM_033237 Gal 546.73 -4.46 ENDOGRO
S68135 GLUT1 5.83 -1.52 ENDOGRO
S70011 g3137721 3.01 1.41 VEGF
S70011 tricarboxylate carrier 4.01 1.23 ENDOGRO
U03388 cyclooxygenase 1 22.47 -1.42 bFGF
U05989 Pawr 45.60 1.29 VEGF
U 16858 testin 4.35 -1.47 ENDOGRO
U 17604 rS-Rex-b 4.23 1.61 bFGF
U 18060 PGHS-1 11.82 1.59 bFGF
U22830 P2Y purinoceptor 3.89 1.30 bFGF
U24174 WAF1 3.08 -1.32 ENDOGRO
U38376 Pla2g4a 10.63 1.23 bFGF
U39208 CYP4F6 8.18 -1.40 bFGF U44948 SmLIM 33.23 1.19 VEGF chemokine receptor U54791 LCR1 20.18 1.41 ENDOGRO
U60085 CYP3A9 18.44 1.40 bFGF
U65656 gelatinase A 21.42 -1.33 bFGF U72353 lamin B1 4.02 1.20 VEGF
U94330 OPG 3.13 -1.89 bFGF
U94709 EP4 prostanoid receptor 3.47 -2.55 VEGF X00469 Cyp1 a1 15.35 -2.04 bFGF X00722 g3727098 3.04 -1.37 bFGF X02601 Mmp3 8.70 1.55 bFGF X13016 OX-45 mRNA 3.55 1.19 VEGF
X14264 CaMII 3.33 1.36 VEGF
X14977 Aldh2 3.11 -1.20 bFGF
X54862 Mgmt 23.74 -1.28 bFGF X58830 vgr 22.26 -1.34 ENDOGRO
X63515 phosphorylase 3.93 -1.91 ENDOGRO X63744 Slc1 a3 7.36 -1.38 VEGF X66539 TNF-alpha 3.97 -1.48 ENDOGRO X68101 trg 4.28 1.19 VEGF
X70706 T-plastin 4.40 1.50 bFGF X84004 CI100 20.03 -1.20 ENDOGRO
Y17328 CDK108 7.15 -1.45 bFGF
Z18877 Oas1 9.02 -3.11 bFGF
Table. 5
C6 Flank Tumor Model, Compound A-induced Signature, EC-specific Sequences Compound-induced Fold Changes in Gene Expression
GenBank
Accession 1 2 3
Number Gene Symbol Dose Doses Doses
M 10934 Rbp4 2.54 1.59 1.45
600519878R1 600519878R1 1.02 -1.94 -1.46
AA925717 1.09 1.34 1.62
AB028461 AB028461 1.36 2.08 1.51
AF056034 AF056034 -1.91 1.34 -2.03
AF058786 JE/MCP-1 1.47 1.58 1.38
AF058786 JE/MCP-1 1.44 1.62 1.36 chemotactic
AF154245 protein-3 1.26 1.63 1.22
AF158385 ATP1 B4 -2.03 1.40 -1.71
AF276998 Jam 1.07 -1.42 -1.84
AF295535 Ata3 -1.79 -1.30 -2.23
AJ299016 Ret 1.05 -1.65 -1.71
BF564460 BF564460 -1.08 -1.49 -1.78
D13871 glut 5 coding -1.54 -1.65 -1.19
AA800146 g2863101 1.08 1.85 1.60
AA818658 g2888244 1.23 2.07 1.57
AA818845 g2888431 -2.40 1.13 -1.63
BG378083 g2888538 -1.80 1.42 -1.84
AA819832 g2889019 -1.26 -1.74 -1.75
AA848809 g2936349 1.28 1.85 1.59
G2937470 g2937470 -1.76 1.09 -2.73
BI282277 g2939494 -1.73 1.30 -1.66
BM390487 g2948168 -1.13 -1.81 -2.19
G3019978 g3019978 1.06 -1.39 -1.68
G3020570 g3020570 -1.01 -1.50 -1.65
AA900587 g3035941 -2.36 1.58 -2.63
AC091752 g3071324 1.22 1.79 1.46
G3073040 g3073040 1.02 -1.31 -1.69
G3103294 g3103294 -1.11 -1.70 -1.46
AA943790 g3103706 -2.49 1.33 -4.44
AA944827 g3104743 1.20 1.91 1.56
AA946094 Mb -2.04 1.18 -2.31
AA946201 g3106117 1.47 2.25 1.31
AW251703 g3120965 1.20 1.73 1.51
G3136659 g3136659 -1.57 1.33 -1.70
G3136765 g3136765 -1.67 -1.54 -1.19
AA963444 g3137002 -1.68 -1.58 -1.27
AA996414 g3186969 1.01 -1.50 -1.66
AA996581 g3187136 -1.85 -1.23 -1.49
AI008203 g3222035 -1.72 -1.52 -1.32
CA503524 g3222465 1.24 1.95 1.60
AI009946 g3223778 -1.82 -1.59 -1.25
AI044213 g3291116 1.05 -1.44 -1.65
AI044263 g3291166 1.23 1.78 1.58 AI044643 g3291504 -1.01 -1.52 -1.66
AI059122 g3332899 1.14 1.70 1.26
BF558524 g3333166 -2.26 1.24 -2.52
AI059446 g3333223 -1.19 -1.77 -1.87
AI059511 g3333288 -1.66 -1.65 -1.30
G3398050 g3398050 -1.15 -2.01 -1.68
AI072733 g3398927 -1.47 1.16 -1.61
AH 13026 g3512975 -1.19 -1.29 -1.71
G3637152 g3637152 -1.02 -1.36 -1.63
G3637289 g3637289 -1.02 -1.66 -1.96
AH 37249 g3638026 -1.56 1.38 -1.78
AH 37425 g3638202 -1.06 -1.91 -1.28
M31591 g3704673 1.44 2.24 1.90
G3708698 g3708698 -2.63 1.39 -2.88
AH 04620 g3708949 -1.96 1.40 -1.71
AH 05417 g3709501 -1.15 -1.85 -1.91
AH 70840 g3710880 -2.23 1.43 -2.16
BQ211970 g3711814 -2.26 1.52 -2.37
G3725683 g3725683 -2.15 1.08 -3.22
CA503625 g3726615 -1.15 -1.66 -1.98
G3727000 g3727000 -1.19 -1.25 -1.64
AA924506 g3728041 1.17 -1.60 -1.35
AH78585 g3729223 1.03 -1.53 -1.77
G3811945 g3811945 1.15 1.89 1.53
G3813551 g3813551 1.10 -1.78 -1.47
AI231826 g3815706 -1.12 -1.72 -2.24
AI232784 g3816664 1.06 -1.36 -1.62
CA509598 g3817191 1.22 2.30 1.38
AI233962 g3817842 -1.81 1.19 -1.90
G977684 g977684 -1.61 -1.42 -1.20
K02111 MYHC mRNA -1.70 -1.36 -1.50
L19998 Sult1a1 1.10 -1.80 -1.48
M11596 Caleb 1.03 1.41 1.59
M26125 XEH mRNA 1.06 -1.76 -1.48
M26744 IL6 1.37 1.71 1.02 alpha(B)-
M55534 crystallin 1.04 1.65 1.41
M81785 syndecan 1.22 1.59 1.35
NM 012561 Fst 1.41 2.01 1.92
NM 012945 Dtr 1.27 1.95 1.28
NM 012949 Eno3 -2.30 1.36 -2.24
NM 013012 Prkg2 1.06 1.62 1.51
NM 013153 Has2 1.27 1.59 1.42
NM 017099 Kcnjδ 1.06 -1.33 -1.63
NM 017117 Capn3 -2.03 1.43 -2.17
NM 017123 Areg 1.22 1.61 1.54
NM 017178 Bmp2 1.46 1.71 1.40
NM 017210 Dio3 1.21 3.37 1.69
NM 019156 Vtn 1.01 -1.53 -1.71
NM 019341 Rgs5 -1.11 -1.62 -2.01
NM 021588 Mb -1.85 1.44 -1.99
NM 021666 Trdn -1.55 1.43 -1.72
NM 022235 Kcne3 -1.58 -2.05 -2.07
NM 022604 Pg25 -1.66 -2.21 -2.73
NM 024483 Adrald 1.25 1.61 1.29
NM 031327 Cyr61 1.13 1.65 1.29 NM 031345 Gilz -1.29 -1.59 -1.60
NM 031970 Hspbl -1.42 1.10 -1.77
U05341 p55CDC -1.59 -1.42 -1.29
U23407 CRABP II 1.29 1.74 1.25
U53855 ratpgis 1.89 1.71 1.31
U94330 OPG 1.28 1.65 1.24
U94330 OPG 1.30 1.65 1.22
U94330 OPG 1.32 1.68 1.30
X15679 X15679 -1.37 -1.67 1.05
X52883 SulH al 1.11 -1.76 -1.54
Y13275 D6.1A protein -1.24 -1.60 -2.50
C6 Flank Tumor Model, Compound B- induced Signature, EC-specific Sequences Compound-induced Fold Changes in Gene Expression
GenBank Accession 1 2 3
Number Gene Symbol Dose Doses Doses
600507547R1 600507547R1 1.33 2.22 1.53
AW 140657 600510887R1 1.23 2.48 1.54
600522244R1 600522244R1 1.17 1.62 1.38
AI454872 700031879H1 -1.05 2.01 1.64
BQ206769 700064878H1 1.34 1.68 1.44
700510178H1 700510178H1 1.10 1.85 1.30
701216526H1 701216526H1 -1.04 -1.81 -1.43
AW920825 701222534H1 1.10 1.90 1.48
701347825H1 701347825H1 -1.70 -1.84 -1.99
701348620H1 701348620H1 -1.12 -1.62 -1.27
701350232H1 701350232H1 -1.10 -1.63 -1.23 fatty acid
AB005743 transporter 1.35 1.87 1.45 PPAR- gamma
AB011365 protein 1.16 1.83 1.36 PPAR- gamma
AB011365 protein 1.17 1.95 1.45 AB032828 AB032828 -1.08 1.72 1.40 AB036792 ficolin-B -1.77 -2.44 -2.34 AB060092 Scyb2 1.43 2.31 1.92 AF016387 RXRgamma -1.09 -1.76 -1.41 AF058786 JE/MCP-1 1.18 1.78 1.61 AF058786 JE/MCP-1 1.22 1.78 1.56 AF081582 Kpl1 1.01 -1.81 -1.34 AF084934 RT1.D(u) -1.39 -1.81 -1.41 AF087946 Gpr37 -1.07 -1.71 -1.36 AF131294 Abcd2 1.09 -1.65 -1.26 AF131294 Abcd2 1.06 -1.62 -1.25 AF146518 Enpep 1.15 2.35 1.75 chemotactic
AF154245 protein-3 -1.05 1.78 1.45
AF314657 clusterin -1.41 -1.61 -1.42
AW914760 AW914760 -1.23 -1.63 -1.42
AW914760 AW914760 -1.24 -1.60 -1.45
AW914760 AW914760 -1.23 -1.59 -1.36
AW920598 AW920598 1.21 1.61 1.50
D14839 rat FGF-9 1.10 1.74 1.66
D28560 NPH-type III -1.20 -2.11 -1.37
D85509 MT3-MMP -1.07 -2.11 -1.40
D89730 g2429082 1.21 4.18 1.82
BG381698 g2672938 1.01 1.66 1.42
AA799278 g2862233 -1.03 -1.89 -1.31
AA799657 g2862612 1.18 1.60 1.35
AA800145 g2863100 -1.03 1.68 1.30 AA800146 g2863101 1.20 1.75 1.49
BE115875 g2864040 1.12 1.67 1.40
G2888594 g2888594 1.07 2.38 1.65
G2888948 g2888948 -1.16 -1.62 -1.50
AA818607 g2889346 1.25 1.65 1.54
AA848639 g2936179 -1.20 2.23 1.52
AA848993 g2936533 1.39 1.70 1.46
AA849479 g2937019 -1.15 -1.68 -1.26
G2938081 g2938081 -1.06 1.67 1.29
G2939209 g2939209 1.17 1.82 1.41
AA858479 g2948819 1.12 -1.70 -1.28
AA875261 g2980209 1.08 1.67 1.37
BU758985 g3019743 1.02 1.76 1.26
G3019865 g3019865 -1.06 2.11 1.32
AA893022 g3019901 1.08 2.19 1.57
AA899521 g3034875 -1.26 -1.98 -1.41
AA925019 g3072155 -1.01 1.98 1.48
G3073258 g3073258 1.07 1.62 1.35
G3103279 g3103279 -1.15 2.14 1.61
AA943907 g3103823 -1.02 -1.62 -1.29
BQ204813 g3104100 1.16 2.06 1.45
BG375318 g3104739 -1.01 -1.79 -1.24
AA944827 g3104743 1.24 1.69 1.52
AA945643 g3105559 -1.92 -1.34 -1.71
G3105912 g3105912 1.10 2.31 1.38
AA946201 g3106117 1.11 2.01 1.41
AA946355 g3106271 -1.18 -1.61 -1.15
BQ201957 g3106396 -1.34 -1.99 -1.57
G3137782 g3137782 -1.06 1.77 1.33
G3137957 g3137957 -1.02 1.82 1.41
AA996727 g3187282 -1.41 -1.72 -1.63
BM391207 g3188195 -1.10 2.11 1.65
AA998510 g3189161 1.63 3.15 1.81
BQ202244 g3189311 -1.06 -1.83 -1.36
AA998953 g3189544 1.01 2.07 1.58
BQ203060 g3221834 -1.01 1.70 1.39
AI008526 g3222358 -1.04 1.70 1.33
CA503524 g3222465 1.18 2.02 1.38
AI009946 g3223778 1.03 -2.08 -1.39
BQ198730 g3224083 -1.00 1.76 1.37
AI010304 g3224136 1.23 1.72 1.40
AI502256 g3226632 -1.55 -2.23 -1.54
BI290624 g3227931 -1.06 -1.73 -1.35
AI029379 g3247205 1.17 1.64 1.31
G3247660 g3247660 -1.00 1.65 1.43
BQ196623 g3248862 1.43 2.17 1.43
AI411352 g3290939 -1.10 1.61 1.46
AI044052 g3290955 -1.32 -1.95 -1.44
AI044556 g3291417 1.04 2.21 1.51
AI044912 g3291731 1.06 1.89 1.53
AI044948 g3291767 1.29 1.80 1.29
AI045191 g3292010 1.01 3.41 1.60
G3292531 g3292531 1.12 2.03 1.65
AI058759 g3332536 1.35 1.80 1.54
AH03652 g3332682 1.22 1.91 1.36
AI059735 g3333512 -1.27 -1.47 -1.59 AF327511 Smhsl .06 1.66 1.35
BG377159 g3397026 .03 1.71 1.22
BE099056 g3397110 .02 1.61 1.31
G3397860 g3397860 .13 1.64 1.42
AI072959 Mgll .00 2.03 1.37
G3512385 g3512385 .05 -1.85 -1.34
G3513248 g3513248 .05 1.98 1.38
AH36855 g3637632 .20 1.79 1.36
G3637836 g3637836 .06 1.70 1.29
BI289488 g3638183 .06 1.70 1.34
G3638417 g3638417 .30 -2.15 -1.25
G3638675 g3638675 .27 2.08 1.53
G3638748 g3638748 .18 1.67 1.36
G3666553 g3666553 .05 1.71 1.48
G3666899 g3666899 .09 1.72 1.31
G3667679 g3667679 .11 -2.68 -1.59
G3667962 g3667962 .12 -2.02 -1.45
G3668045 g3668045 .24 1.63 1.34
M31591 g3704673 .13 1.97 1.67
BI294910 g3705813 .04 1.83 1.36
AH 01330 g3706248 .13 -1.66 -1.28
AH 02081 g3706915 .08 -1.76 -1.35
G3708538 g3708538 .29 1.87 1.71
G3708830 g3708830 .26 2.40 1.54
G3710429 g3710429 .04 2.10 1.48
G3711566 g3711566 .12 1.62 1.32
AH72274 g3712314 .09 1.92 1.42
G3727230 g3727230 .09 1.63 1.35
G3727318 g3727318 .14 1.79 1.38
AH76957 g3727595 .05 2.12 1.51
AH78367 g3729005 .12 -1.61 -1.49
AH79184 g3729822 .23 -1.96 -1.40
G3729898 g3729898 .36 2.21 1.47
BE117878 g3730830 .07 2.04 1.41
BF282318 g3730930 .90 -1.47 -2.02
G3811572 g3811572 .07 2.42 1.58
BQ206905 g3812156 .02 1.62 1.24
G3812233 g3812233 .13 -1.68 -1.23
G3813551 g3813551 .50 1.87 1.28
BI283128 g3815073 .16 1.93 1.45
NM_175582 g3816235 .45 -2.46 -1.96
AI232356 g3816236 .16 1.77 1.34
AI232402 g3816282 .07 1.70 1.25
CA509598 g3817191 .10 1.68 1.25
AI407821 g3817623 .01 1.93 1.29
G3817759 g3817759 .08 -1.88 -1.35
G3828430 g3828430 .01 1.99 1.40
G3828465 g3828465 .16 2.40 1.71
BI278601 g3829380 .03 1.64 1.32
AI236212 g3829718 .11 1.72 1.33
AI317824 g4033091 .37 -2.14 -1.88
AA800480 g4131501 .35 2.36 1.41
G4132436 g4132436 .00 2.43 1.94
BQ192029 g4133216 .06 1.64 1.27
AH 01250 g4133997 .02 1.73 1.36
AH72271 g4134732 .17 2.01 1.48 AH 77939 g4135031 1.06 1.74 1.30 G807364 g807364 -1.15 1.63 1.28 H32476 g977893 -1.01 1.92 1.34 J03637 AD mRNA -1.14 3.17 1.42 platelet factor
M 15254 4 1.67 2.18 1.39 M 18847 M 18847 1.05 -1.68 -1.30 M23601 Maobf3 -1.06 1.74 1.32 potassium
M26161 channel 1.03 -1.96 -1.34
M26744 IL6 1.14 1.92 1.50
M81785 syndecan 1.06 1.72 1.32
NM_012561 Fst 1.10 1.84 1.65
NMJ312733 Rbp1 -1.00 1.83 1.54
NM_012771 Lyz -1.49 -1.53 -1.69
NM_012908 Tnfsfδ 1.04 -1.64 -1.39
NM_012908 Tnfsfδ 1.02 -1.61 -1.39
NM_013012 Prkg2 1.31 2.03 1.48
NM_013057 F3 1.06 1.65 1.46
NM_013080 Ptprzl -1.11 -1.61 -1.30
NM_013191 S100b 1.04 -1.86 -1.37
NM_016994 C3 -1.74 -1.94 -2.10
NM_017061 Lox -1.22 2.17 1.63
NM_017083 Myo5b 1.82 3.00 1.62
NM_017161 Adora2b 1.07 2.13 1.45
NM_017173 Serpinhl 1.03 1.83 1.32
NM_017210 Dio3 1.97 1.69 2.45
NM_017354 RNU16845 1.05 -2.04 -1.33
NM_019343 Rgs7 1.09 1.87 1.43
NM_019355 CPG2 -1.22 -2.12 -1.24
NM_019358 Gp38 1.05 1.94 1.25
NM_019370 LOC54410 1.02 2.31 1.70
NM_020082 Rnase4 -1.03 1.86 1.43
NM_020542 LOC57301 1.22 1.64 1.39
NM_021698 F13a 1.21 1.64 1.31
NM_022182 Fgf7 1.00 1.79 1.41
NM_022226 Prsd 1.03 1.65 1.24
NM_022230 Stc2 1.07 1.59 1.45
NM_022236 Pde10a -1.02 1.59 1.17
NMJ522849 Crpd -1.07 1.70 1.39
NM_022925 Ptprq 1.29 1.71 1.13
NM_024142 LOC79110 -1.06 -1.60 -1.49
NM_024159 Dab2 1.03 1.82 1.29
NM_030656 Agxt 1.18 1.61 1.24
NM_030832 Fabp7 1.17 2.32 1.70
NM_030990 Plp1 -1.05 -2.33 -1.53
NM_031012 An pep -1.14 1.67 1.35
NM_031022 Cspg4 -1.14 -1.70 -1.23
NM_031050 Lum -1.17 1.97 1.52
NM_031334 Cdh1 -1.32 -2.12 -1.39
NM_031645 Rampl 1.07 1.63 1.21
NM_031712 Pdzkl -1.43 -1.86 -1.62
NM_031713 Pirb -1.68 -1.37 -1.59
NM_031716 Wispl -1.03 1.88 1.43
NM_031736 Slc27a2 -1.07 -1.72 -1.71
NM 031761 Figf -1.03 1.84 1.39 NM_031807 Tpbg 1.04 1.77 1.44 NM_032060 C3ar1 1.20 1.68 1.29 glutathione S- transferase
S72505 Yd subunit 1.72 2.68 1.39 glutathione S- transferase
S72505 Yd subunit 2.01 2.79 1.50 glutathione S- transferase
S72505 Yd subunit 2.02 2.92 1.65 S82820 GSTA5 1.91 2.60 1.46 U04808 Rbs11 -1.18 -1.61 -1.33 cytochrome
U36992 P450 Cyp7b1 -1.22 1.78 1.49 cytochrome
U36992 P450 Cyp7b1 -1.20 1.82 1.57 cytochrome
U36992 P450 Cyp7b1 -1.22 1.84 1.63 U39943 CYP2J3 -1.12 -1.72 -1.40 U53855 ratpgis 1.33 2.53 1.59 U57062 RNKP-4 -2.10 -1.70 -2.07 U65217 U65217 -1.50 -2.21 -1.55 U65656 gelatinase A -1.01 2.05 1.43 U66470 U66470 1.16 2.05 1.41 U75581 A-FABP 1.55 2.22 1.69 U75581 A-FABP 1.60 2.46 1.73 U75581 A-FABP 1.60 2.48 1.79 U81037 NrCAM 1.24 2.20 1.43 U85512 Gchfr 1.41 1.74 1.64 U94330 OPG 1.01 1.66 1.34 X15679 X15679 1.07 -2.00 -1.33 X59859 DCN -1.02 2.43 1.60 X68312 igM -1.19 -2.16 -1.50 X78997 cadherin -1.21 -1.79 -1.35 Z30663 Z30663 1.06 -1.74 -1.36
MatBIII Tumor Model, Compound A-induced Signature, EC-specific Sequences Compound-induced Fold Change in Gene
GenBank Accession Number Gene Symbol Expression
600511261 R1 600511261 R1 1.61
600512030R1 600512030R1 -2.01
600516277R1 600516277R1 1.70
600521787R1 600521787R1 2.45
600521928R1 600521928R1 -1.72
600522339R1 600522339R1 1.72
700031150H1 Hbb -2.93
700031292H1 Hbb -2.95
700033761 H1 700033761 H1 -1.69
700035711H1 700035711H1 -1.77
700036014H1 Hbb -3.21
700037874H1 700037874H1 -2.14
700040546H1 700040546H1 -2.36
700041049H1 700041049H1 -2.47
700509540H1 700509540H1 1.59
701217088H1 701217088H1 1.80
701347811H1 701347811H1 -1.74
AA012768 g1473830 1.72
AA799471 g2862426 3.28
AA799964 g2862919 1.62
AA800145 g2863100 -1.75
AA800690 700036377H1 -3.01
AA800790 g2863745 -2.53
AA801013 g2863968 2.88
AA818163 g2888043 1.78
AA818207 700035764H1 -3.04
AA818804 g2888390 1.81
AA818845 g2888431 2.74
AA819500 -1.68
AA848265 g2935805 1.88
AA848809 g2936349 1.60
AA858479 g2948819 1.65
AA859373 g2948724 2.23
AA874889 g2979837 -2.20
AA874924 g2979872 -1.69
AA892298 g3019177 -1.61
AA900587 g3035941 1.99
AA924082 g3071218 1.59
AA943742 g3103658 -1.76
AA943743 g3103659 -2.53
AA944410 g3104326 -1.74
AA945100 g3105016 -1.60 AA9451 19 Hbb -3.64
AA945677 g3105593 1 .95
AA945951 g4132652 5.93
AA946094 Mb 2.83
AA946457 g4132801 -2.19
AA956834 g3120529 -2.23
AA963068 g3136560 -2.08
AA963366 g3136858 2.43
AA996698 g3187253 -1.67
AA997375 g3187690 1.71
AB011533 MEGF7 -1.70
AC091752 g3071324 1.78
AF009329 SHARP-1 1 .83
AF015304 Slc29a1 1 .69
AF037272 ps20 1 .75
AF055286 3-Oct 1 .80
AF056034 AF056034 2.30
AF058786 JE/MCP-1 -1 .80
AF058786 JE/MCP-1 -1 .76
AF058786 JE/MCP-1 -1 .69
AF063102 CIRL-2 -1 .80
AF109393 podocalyxin -2.34
AF109674 Lgl1 1 .61
AF134409 Rhes protein 2.29
AF146518 Enpep -2.03
AF150082 DDP1 -1.66
AF157005 MYHC 6.94
AF158385 ATP1 B4 2.57
AF159103 Tnfip6 -1.67
AF160978 C1qRp -2.02
AF173834 calpain isoform Rt88' 1.96
AF269251 mob-5 1.97
AF271786 Fgf13 2.06
AF314657 clusterin 2.13
AF323174 Clicδ 2.57
AF364071 Smpx 2.64
AF372834 g4135229 1.63
AF404762 g3704880 1.67
AF450248 Actn3 3.96
AI008526 g3222358 -2.88
AI009020 600518256R1 1.61
AI009669 g3223501 -1.61
AI01 1757 g3225589 -1.62
AI013912 g3227968 -1.72
AI029383 g3247209 3.23
AI030556 g3248382 1.77
AI043880 g3290615 1.76 AI044257 g3291160 1.99 AI044658 g3291519 2.38 AI044948 g3291767 -1.94 AI045276 g3292095 -2.89 AI058243 700034477H1 -1.69 AI059662 g3333439 1.98 AI070419 g3396670 -1.65 AI071230 g3397445 1.80 AI071570 600520320R1 2.07 AI072669 g3398863 1.78 AI072687 g3398881 1.64 AI072733 g3398927 1.68 AI072751 g3398945 -1.70 AH02560 g3707304 -2.36 AH04898 g3708043 2.57 AH04955 g3709178 2.22 AH 13026 g3512975 -2.36 AH 37425 g3638202 -2.48 AH37674 g3638451 1.79 AH44943 g3666742 1.87 AH 69239 g3705547 -3.08 AH 69311 g3705619 1.78 AH 70840 g3710880 2.19 AH 70948 g3710988 -1.77 AH71466 g3711506 1.87 AH72271 g4134732 -2.04 AH75988 g3726626 3.28 AH76957 g3727595 -2.20 AH77013 g3727651 1.65 AH77057 g4134951 -3.44 AH77392 g3728030 -1.67 AH77951 g3728589 -1.82 AH78376 g4135065 1.60 AH78585 g3729223 -2.30 AH 78996 g3729634 1.59 AI231053 g3814933 -1.91 AI231438 g3815318 1.61 AI233773 Mawbp 1.77 AI235210 g4136063 -4.17 AI235960 g3829466 -1.66 AI236229 g4136175 3.67 AI409186 g2938420 -2.62 AI411737 Hbb -3.37 AI412460 g3704629 -1.65 AI603145 g3707784 2.62 AJ242926 irl685 2.05 AJ426426 600524449R1 1.68 AW253240 700039226H1 -3.10
AW915763 600523193R1 1.65
AW916287 g2980074 -1.71
AY027527 NADPH oxidase 4 -1.66
AY1 15564 g3712131 2.68
BE109616 g2862726 2.56
BE117010 g2949577 -2.03
BF287821 700065626H1 -1.63
BF393825 g3814029 -1.59
BF395396 g3187656 2.88
BF401710 g2863152 1.63
BF419896 600508357R1 -1.67
BF558524 g3333166 1.94
BG373503 g3138384 -1.65
BG374556 700510594H1 1.61
BG378083 g2888538 2.54
BG670348 g3332207 -1.62
BI274062 Trela 2.08
BI277462 g3729596 -1.96
BI279373 g3814946 1.70
BI282277 g2939494 2.23
BI283128 g3815073 -2.27
BI285246 g3830698 -1.81
BI291451 g2889589 -1.78
BI294910 g3705813 -1.88
BI296277 g3705123 -1.67
BM386121 g2862284 -2.23
BM390441 g3707901 -1.67
BQ190196 g3831151 -1.78
BQ191387 g3512957 1.72
BQ194973 g3118966 -1.93
BQ 196248 g2938644 -1.65
BQ 199466 g3712138 -1.67
BQ 199747 g3707262 -1.65
BQ204163 g3291004 4.70
BQ206937 g3816206 1.59
BQ208795 LOC85383 1.75
BQ211970 g3711814 1.84
BQ779790 g3709350 2.08
BU758944 g3729204 -1.62
CA503430 g3818031 -2.01
CA503625 g3726615 -1.97
CA505509 g3020114 2.75
CA507161 g3226571 -1.77
CA508003 g3707933 -1.89
CA509083 g3397522 1.59
CA509145 g3727217 1.93 CA509211 g3704741 -1.60
CA509598 g3817191 2.15
CA509955 g3730145 -1.74
D12520 nitric oxide synthase 1.63
D14051 nitric oxide synthase 1.62
D28561 glucose transporter 2.60
D63164 cyclin E -1.66
D86800 D86800 -1.92
D86800 D86800 -1.85
G2863860 3.31
G2889725 2.55
G2939329 -1.67
G3018718 -1.64
G3018906 -2.03
G3020570 -1.63
G3036209 -1.61
G3072525 3.27
G3072678 2.28
G3073004 -1.77
G3105912 -1.63
G3136659 2.50
G3138335 -2.07
G3187018 -1.69
G3225225 -1.92
G3226067 -2.21
G3226140 5.03
G3226701 2.48
G3227787 -2.28
G3291802 -1.77
G3292543 2.04
G3396388 1.91
G3396690 -1.99
G3396723 1.77
G3397083 1.59
G3397437 -1.72
G3397917 -1.62
G3397969 -2.39
G3398441 -2.39
G3399406 -2.47
G3513002 1.96
G3513248 -2.52
G3636896 -1.94
G3637290 1.97
G3637649 1.60
G3637836 1.69
G3638063 -1.64
G3638106 -1.60 G3638248 -1 .86
G 3666834 -1 .75
G3667558 -1 .64
G3667798 -1 .59
G3708538 -1 .59
G3708698 3.29
G3708830 1 .72
G3708934 1 .61
G3709332 1 .78
G3709581 -1.76
G3710109 -1.87
G3710353 -1.89
G3710419 -1.60
G3710427 -1 .67
G3710782 1.72
G3712041 -1.70
G3712094 3.52
G3712205 1.79
G3712229 1 .68
G3725978 3.71
G3726013 2.02
G3726093 1 .91
G3726475 -3.16
G3727000 1 .91
G3727318 -1 .61
G3811492 -2.18
G3812333 -1.88
G3813207 -1.60
G3817985 -1.66
G4131487 1.95
G4131762 1.84
G4135671 2.78
G977371 1.70
G978154 -1.86
H35065 g980482 3.04
J02582 apoE 1.64
J02585 s-CoA d mRNA 2.04
K00781 g3247405 1.94
L00381 L00381 2.50
L16764 HSP70 1.60
L16764 HSP70 1.70
L16764 HSP70 1.79
L20681 Ets-1 -1.59
M11670 cat mRNA 1.60
M11851 MLC2 mRNA 2.69
M26744 IL6 -2.45
M26744 IL6 -2.38 M26744 IL6 -2.35
M29853 P-450 mRNA 2.02
M29853 P-450 mRNA 2.03
M34097 -1.64
M55149 pap 2.08
M58040 transferrin receptor -1.81
M60616 UMRCase 1.81
NM_012491 Add2 -1.78
NM_012497 Aldoc 1.60
NM_012505 Atp1 a2 3.08
NM_012530 Ckm 2.64
NM_012588 Igfbp3 -3.62
NM_012604 Myh3 2.35
NM_012605 Myl2 2.36
NM_012771 Lyz -1.74
NM_012786 Coxδh 3.99
NM_012812 Cox6a2 2.55
NM_012864 Mmp7 2.08
NM_012949 Eno3 3.06
NM_012966 Hspel -1 .71
NM_013037 111 rl1 -1 .79
NM_013044 Tmod 2.36
NM_013062 Kdr -3.79
NM_013153 Has2 -1 .82
NM_013186 Kcnbl 2.96
NM_013197 Alas2 -2.48
NM_017049 Slc4a3 1 .67
NM_017066 Ptn -1.97
NM_017104 Csf3 1.85
NMJ317115 Myog 2.53
NM_017117 Capn3 2.47
NM_017131 Casq2 1.66
NM_017184 Tnnil 1 .96
NM_017185 Tnni2 3.43
NM_017328 Pgam2 3.97
NM_017333 Etb -2.74
NM_019131 Tpm1 1.97
NM_019212 Actal 3.55
NM_019278 Resp18 1.69
NM_019282 Cktsf1b1 -1.82
NM_019292 Ca3 2.82
NM_019334 Pitx2 2.84
NM_019341 Rgs5 -1.83
NM_021588 Mb 2.44
NM_021593 Kmo -1.62
NM_021666 Trdn 1.64
NM 021693 LOC59329 1.61 NM_022235 Kcne3 -1 1.33
NM_022396 Gng11 -1.80
NM_022604 Pg25 -6.48
NM_022631 Wntδa -1.74
NM_022674 H2afz -1.74
NM_023991 Prkaa2 3.17
NM_024141 Thox2 2.32
NM_030856 Lrrn3 -1.87
NM_031007 Adcy2 1.94
NM_031022 Cspg4 -1 .77
NM_031039 Gpt 1 .70
NM_031511 Igf2 3.83
NM_031531 Spin2c 2.18
NM_031612 Apel -2.36
NM_031715 Pfkm 1 .68
NM_031813 Mybph 1 .84
NM_032063 DII1 -1 .76
NMJD32072 Appbpl -1.71
U22520 IP-10 -2.40
U25281 CR16 1 .60
U25684 U25684 -1 .60
U31935 CAP2 2.53
U94330 OPG -1.68 glutathione
X67654 transferase 1 .69 X81449 g587519 1 .68 X82152 fibromodulin, unnamed 1 .92 X92069 P2X5 1 .86 X98517 Mmp12 1.66
Table. 6
C6 Flank Tumor Model, Compound A
GenBank Accession Number Gene Symbol
600520 86R1
701217994H1
AA945677 g3105593
AA957449 g3121144
AA964264 g3137756
AB015308 Gna15 chemotactic
AF154245 protein-3
AF244366 FLIP short form
AF314657 clusterin
AI010322 g3224154
AI012597 g3226429
AI059363 g3333140
AH 36847 g3637624
AI236799 g4136246
AI454865 g3103424
AW142194 g2864225
BI285246 g3830698
BQ207019 g3730060
G2938456
G2938797
G3021176
G3137780
G3138247
G3225906
G3396115
H32810 g978227
H34187 g979604
L20468 cerebroglycan
NM_012922 Casp3
NM_013085 Plau
NM_017105 Bmp3
NM_031327 Cyr61
NM_080783 600519254R1
U 17604 rS-Rex-b
U 18060 PGHS-1
Y15685 g2648067 C6 Flank Tumor Model, Compound B
GenBank Accession Number Gene Symbol
600520186R1
700510178H1
701217994H1
701419627H1
AA964264 g3137756
AB015308 Gna15
AF075704 Ata1
AF140232 S100A6
AF154245 chemotactic protein-3
AI008035 700068780H1
AI009736 g3223568
AI010322 g3224154
AI012085 g4133563
AI012597 g3226429
AI059363 g3333140
AH 36847 g3637624
AH77939 g4135031
AI228076 g4135309
AI454865 g3103424
AW140657 600510887R1
BE115875 g2864040
BI278601 g3829380
BI285246 g3830698
BI294910 g3705813
BQ192029 g4133216
BQ196623 g3248862
BQ203060 g3221834
D89730 g2429082
G2938081
G2946933
G3072603
G3137780
G3137957
G3138247
G3292531
G3396115
G3397675
G3512937
G3638675
G3666553
G3666899
G3710770
G3726475
G3813551 G3814512
G977854
M81855 Abcbl
NM_012566 Gfi1
NM_019261 Klrc2
NM_022925 Ptprq
NM_031518 Mox2
N M_080783 600519254R 1
U36992 cytochrome P450 Cyp7b1
U60085 CYP3A9
U60085 CYP3A9
U65656 gelatinase A
Y15685 g2648067
MatBIII Tumor Model, Compound A
GenBank Accession Number Gene Symbol
600520186R1
701219674H1
AA800293 g2863248
AA848602 g2936142
AA859467 g2948987
AA945677 g3105593
AA957449 g3121144
AA963106 g3136598
AA996897 g3187452
AF022774 Rph3al
AF248543 iGb3 synthase
AF314657 clusterin
AF368269 Cyp2t1
AI008526 g3222358
AI010322 g3224154
AI012085 g4133563
AI012597 g3226429
AI044404 g3291307
AI044948 g3291767
AI045276 g3292095
AH 69311 g3705619
AH70948 g3710988
AH 76957 g3727595
AI228076 g4135309
AI233773 Mawbp
AI409186 g2938420
AI454865 g3103424
AW142194 g2864225
BI278601 g3829380
BI285246 g3830698
BM388852 g3106350
BM390441 g3707901
BQ196557 g3729556
D16339 g6981681
D16339 Ttpa
D88666 PS-PLA1
G2938797
G3034536
G3226140
G3396115
G3398076
G3399406
G3513248 G3638802
G3666553
G3666899
G3667173
G371 1240
G3725764
G3730626
G3813079
G977371
H32810 g978227
H35065 g980482
NM_012715 Adm
NM_013062 Kdr
NM_021676 Shank3
NM_021751 LOC60357
NM_022242 Niban
NM_022959 P-cip1
NM_031327 Cyr61
NM_031544 Ampd3
NM_031658 Msln
U03388 cyclooxygenase 1
U18771 Rab26
U38419 LOC64305
X63515 phosphorylase
EXAMPLE 6 BIOMARKER VALIDATION In order to confirm that the seven genes we identified as potential biomarkers of tumor endothelial cell proliferation were specifically expressed in tumor vasculature and whose expression levels reflected endothelial cell proliferation rates, we performed several validation experiments. First, we independently assessed gene expression levels in the animal tumor RNA samples by quantitative realtime PCR to confirm the microarray hybridization results. The biomarker data obtained from the microarray experiments described above was validated by real time quantitative real time PCR. Results: Quantitative real time PCR was performed with gene-specific PCR primer pairs and amplicon- specific fluorescent probes (TaqMan). For each RNA sample tested, transcript abundance of GAPDH was determined. In addition, transcript abundance of genes of interest and GAPDH were determined for a calibrator RNA sample (total rat lung RNA). (A) Fold changes in gene expression in tumors from KDR kinase-treated animals relative tumors from vehicle-treated animals were calculated using the ΔΔCT method (see Materials and Methods). mRNA levels for each gene in the rat tumors are also shown relative the calibrator RNA pool (B). As shown in Figure 7, the results obtained from the real time PCR studies closely matched those from the DNA microarrays (Figure 7). We also measured the expression levels of the biomarker genes in an additional set of rat MatBIII tumors from a fourth animal study for which no gene expression profiling was performed. In this independent study, animals with established MatBIII breast tumors (seven days post cell implantation) were dosed orally once per day with Compound A or vehicle for a total of eight days. Half of each tumor from our animal tumor studies was fixed and preserved for sectioning and immunohistochemistry as described in Materials and Methods. In order to determine if expression of the biomarker genes was specific to the endothelial cells within tumors, we visualized their protein products in rat tumor tissue sections by immunofluorescence microscopy. Antibodies are available commercially for five of the seven biomarker protein products and we were able determine optimal conditions for immunofluorescence staining. Individual tumor sections approximately 3-5 um thick were de-waxed, rehydrated and incubated with antibodies against one of the biomarker proteins and also with antibodies against the endothelial cell surface protein CD31 (to label the tumor vasculature). De-waxed, re-hydrated MatBIII tumor sections were incubated with antibodies against CD31 and one of the following biomarker proteins: CLU, ANGPT2, CYR61, ENDRB, or PLAU. Primary antibodies bound to the biomarker proteins and CD31 were visualized with Alexa488-labeled and Alexa546-labeled secondary antibodies, respectively as described in Materials and Methods. After mounting under coverslips, images were captured with a Zeiss Axiocam through a Zeiss Axiovert 135 fluorescence microscope equipped with a 40x objective. Results: We found that the five proteins we examined (ANGPT2, CLU, CYR61, EDNRB, and PLAU) were each localized specifically to the tumor vasculature in MatBIII tumors (Figure 8). Indicating that biomarker protein expression in rat mammary tumors is localized to vasculature. The expression levels of all genes with the exception of Cyrόl changed as expected in response to exposure to the KDR kinase inhibitor. Finally, we correlated the confirmed expression changes of the biomarker genes in Compound A- treated tumors with an independent measure of tumor endothelial cell proliferation. Using a modification of the method described by Mundhenke, et al. (Mundhenke, 2001), we measured endothelial cell proliferation rates by double immunohistochemical staining of tumor sections for the endothelial cell marker CD31 and the nuclear proliferation marker Ki67. We analyzed C6 flank tumors from five vehicle- treated animals and five Compound A-treated animals (3 doses vehicle or compound over 72 hrs). Results: We determined the endothelial cell proliferation rate in tumors from vehicle treated animals to be 34% +/- 5%. In contrast, we determined that the endothelial cell proliferation rate was only 19 % +/- 5% in tumors from animals treated with Compound A. While the present invention has been described with reference to what are considered to be the specific embodiments, it is to be understood that the invention is not limited to such embodiments. To the contrary, the invention is intended to cover various modifications and equivalents included within the spirit and scope of the appended claims. For example, while the disclosure focuses on using the disclosed biomarkers for detecting the efficacy of a KDR kinase inhibitor, the use of similar methods to evaluate the ability of other cancer therapeutics to regulate the proliferative state of vascular endothelial cells within tumors are specifically within the scope herein. All references cited throughout the disclosure are hereby expressly incorporated by reference.
References Cited in the Specification
1. N. Ferrara, H. Heinsohn, C. E. Walder, S. Bunting, G. R. Thomas, Ann N Y Acad Sci 752, 246- 56 (Mar 27, 1995).
2. N. Ferrara, H. P. Gerber, J. LeCouter, Nat Med 9, 669-76 (Jun, 2003). 3. D. W. Leung, G. Cachianes, W. J. Kuang, D. V. Goeddel, N. Ferrara, Science 246, 1306-9 (Dec 8, 1989).
4. N. Ferrara, Nat Rev Cancer 2, 795-803 (Oct, 2002).
5. K. J. Kim et al., Nature 362, 841-4 (Apr 29, 1993).
6. T. P. Quinn, K. G. Peters, C. De Vries, N. Ferrara, L. T. Williams, Proc Natl Acad Sci U S A 90, 7533-7 (Aug 15, 1993).
7. R. L. Kendall et al., J Biol Chem 274, 6453-60 (Mar 5, 1999).
8. H. P. Gerber, N. Ferrara, J Mol Med 81, 20-31 (Jan, 2003).
9. N. Ferrara, Semin Oncol 29, 10-4 (Dec, 2002).
10. H. Gille et al., J Biol Chem 276, 3222-30 (Feb 2, 2001). 11. C. Mundhenke et al., Clin Cancer Res 7, 3366-74 (Nov, 2001).
12. M. E. Fraley et al., Bioorg Med Chem Lett 12, 3537-41 (Dec 16, 2002).
13. P. J. Manley et al., Bioorg Med Chem Lett 13, 1673-7 (May 19, 2003).
14. M. T. Bilodeau et al., Bioorg Med Chem Lett 13, 2485-8 (Aug 4, 2003).
15. M. E. Fraley et al., Bioorg Med Chem Lett 13, 2973-6 (Sep 15, 2003). 16. T. R. Hughes et al., Nat Biotechnol 19, 342-7 (Apr, 2001).
17. M. J. Marton et al., Nat Med 4, 1293-301 (Nov, 1998).
18. H. M. DeLisser, P. J. Newman, S. M. Albelda, Curr Top Microbiol Immunol 184, 37-45 (1993).
19. B. R. DeYoung et al., J Cutan Pathol 22, 215-22 (Jun, 1995).
20. H. M. DeLisser, P. J. Newman, S. M. Albelda, Immunol Today 15, 490-5 (Oct, 1994). 21. D. C. Brown, K. C. Gatter, Histopathology 40, 2-11 (Jan, 2002).
22. D. C. Brown, K. C. Gatter, Histopathology 17, 489-503 (Dec, 1990).
23. B. A. Keyt et al., J Biol Chem 271, 5638-46 (Mar 8, 1996).
24. C. de Vries et al., Science 255, 989-91 (Feb 21, 1992).
25. J. Lauren, Y. Gunji, K. Alitalo, Am J Pathol 153, 1333-9 (Nov, 1998). 26. L. Zhang et al., Cancer Res 63, 3403-12 (Jun 15, 2003).
27. P. J. Svensson, M. Anvret, M. L. Molander, A. Nordenskjold, Hum Genet 103, 145-8 (Aug, 1998).
28. K. Kikuchi et al., Biochem Biophys Res Commun 219, 734-9 (Feb 27, 1996).
29. M. Okada, M. Nishikibe, Cardiovasc Drug Rev 20, 53-66 (Winter, 2002). 30. R. Lahav, G. Heffner, P. H. Patterson, Proc Natl Acad Sci U S A 96, 11496-500 (Sep 28, 1999). 31. D. A. Withers, S. I. Hakomori, J Biol Chem 275, 40588-93 (Dec 22, 2000).
32. A. Taniguchi, R. Suga, K. Matsumoto, Biochem Biophys Res Commun 273, 370-6 (Jun 24, 2000).
33. I. P. Trougakos, E. S. Gonos, Int J Biochem Cell Biol 34, 1430-48 (Nov, 2002). 34. S. E. Jones, C. Jomary, Int J Biochem Cell Biol 34, 427-31 (May, 2002).
35. C. Koch-Brandt, C. Morgans, Prog Mol Subcell Biol 16, 130-49 (1996).
36. W. Zhou, L. Janulis, Park, fl, C. Lee, Life Sci 72, 11-21 (Nov 22, 2002).
37. M. Scaltriti et al., Int J Cancer 108, 23-30 (Jan 1, 2004).
38. S. Bettuzzi et al., Oncogene 21, 4328-34 (Jun 20, 2002). 39. S. Bettuzzi, Acta Biomed Ateneo Parmense 74, 101-4 (Aug, 2003).
40. L. Y. Zhang et al., World J Gastroenterol 9, 650-4 (Apr, 2003).
41. J. A. Menendez, I. Mehmi, D. W. Griggs, R. Lupu, Endocr Relat Cancer 10, 141-52 (Jun, 2003).
42. M. L. Kireeva et al., Exp Cell Res 233, 63-77 (May 25, 1997).
43. M. L. Kireeva, F. E. Mo, G. P. Yang, L. F. Lau, Mol Cell Biol 16, 1326-34 (Apr, 1996). 44. T. M. Grzeszkiewicz, D. J. Kirschling, N. Chen, L. F. Lau, J Biol Chem 276, 21943-50 (Jun 15, 2001).
45. S. J. Leu, S. C. Lam, L. F. Lau, J Biol Chem 277, 46248-55 (Nov 29, 2002).
46. S. J. Leu et al., J Biol Chem 278, 33801-8 (Sep 5, 2003).
47. J. M. Schober, L. F. Lau, T. P. Ugarova, S. C. Lam, J Biol Chem 278, 25808-15 (Jul 11, 2003). 48. T. M. Grzeszkiewicz, V. Lindner, N. Chen, S. C. Lam, L. F. Lau, Endocrinology 143, 1441-50
(Apr, 2002).
49. P. F. Choong, A. P. Nadesapillai, Clin Orthop, S46-58 (Oct, 2003).
50. A. P. Mazar, J. Henkin, R. H. Goldfarb, Angiogenesis 3, 15-32 (1999).
51. J. B. Smith, H. R. Herschman, Arch Biochem Biophys 330, 290-300 (Jun 15, 1996). 52. J. B. Smith, H. R. Herschman, J Biol Chem 270, 16756-65 (Jul 14, 1995).
53. C. G. Lee, J. Demarquoy, M. J. Jackson, W. E. O'Brien, J Immunol 152, 5758-67 (Jun 15, 1994).
54. M. J. de Veer, H. Sim, J. C. Whisstock, R. J. Devenish, S. J. Ralph, Genomics 54, 267-77 (Dec 1, 1998).
55. S. F. Altschul et al., Nucleic Acids Res 25, 3389-402 (Sep 1, 1997).

Claims

WHAT IS CLAIMED IS:
1. A method for determining the proliferative status of a population of endothelial cells comprising: a) providing an array comprising a substrate having a plurality of addresses, wherein each address has disposed thereon a capture probe or oligonucleotide that can specifically bind an endothelial cell proliferative biomarker; b) preparing a nucleic acid test sample from a population of endothelial cells; c) contacting the nucleic acid sample with the array; and d) determining an expression profile by detecting binding of the nucleic acids in the test sample to each address of the plurarity of addresses present on the array, thereby determining the proliferative rate of the endothelial cells.
2. The method of claim 1 wherein the array comprises oligonucleotides that specifically binds one or more genes selected from the gene sequences set forth in Table 3 or Table 4.
3. The method of claim 2 wherein the array comprises oligonucleotides that specifically binds to Angpt-2, Clu (ApoJ), Cyrόl (CCNl), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA) genes in combination with other biomarkers selected from the genes identified in Tables 3 through 6.
4. The method of claim 1 wherein the nucleic acid sample is prepared from a population of vascular endothelial cells obtained from a tumor biopsy.
5. The method of claim 4 wherein the sample is prepared from a cancer patient receiving treatmentwith a therapeutic agent intended to inhibit the proliferation of endothelial cells in tumor vasculature.
6. The method of claim 5 wherein the patient is being treated with a KDR kinase inhibitor.
7. The method of claim 6 wherein the patient is being treated with Compound A.
8. A method for screening a plurality of therapeutic agents for anti-angiogenic activity comprising: contacting a compound with a population of cells containing a polynucleotide comprising a nucleic acid sequence selected from the group consisting of the biomarkers identified in Tables 4 and Table 5 under conditions wherein said polynucleotide is being expressed, and determining a change in expression of an endothelial cell proliferation signature, wherein a change in expression is indicative of anti-angiogenic activity.
9. The method of claim 8 wherein the population of cells is isolated from a tumor biopsy of a cancer patient who is being treated with a therapeutic agent intended to inhibit the proliferation of endothelial cells in tumor vasculature.
10. The method according to claim 10 wherein the therapeutic agent is a receptor-type kinase inhibitor.
11. The method of claim 8 wherein the change in of the endothelial cell expression signature is determined by hybridization to a microarray comprising oligonucleotide that can specifically bind an endothelial cell proliferative biomarker or by RT-PCR.
12. The method of claim 11 wherein the endothelial cell proliferation signature comprises capture probes or oligonucleotides that specifically binds to Angpt-2, Clu (ApoJ), Cyrόl (CCNl), Endrb (Etb), Ifit-3 (Garg49), Fut-4, Plau (uPA) genes in combination with other biomarkers selected from the genes identified in Table 5 or Table 6.
13. An array comprising a substrate having a plurality of addresses, wherein each address has disposed thereon a capture probe or oligonucleotide that can specifically bind a polynucleotide sequence of a biomarker gene seleceted from the group consisting of the genes comprising a proliferation signature defined in Table 3 and 4.
14. A composition comprising the biomarker genes comprising the proliferation signatures set forth in Table 3 or Table 4.
15. A composition comprising the biomarker genes comprising the expression signatures set forth in Table 5 or Table 6.
16. An endothelial cell proliferation signature comprising Angpt-2, Clu (ApoJ), Cyrόl (CCNl), Endrb (Etb), Ifit-3 (Garg49), Fut-4, and Plau (uPA) genes.
PCT/US2005/009874 2004-03-26 2005-03-24 Method and biomarkers for detecting tumor endothelial cell proliferation WO2005098448A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA002561338A CA2561338A1 (en) 2004-03-26 2005-03-24 Method and biomarkers for detecting tumor endothelial cell proliferation
JP2007505185A JP2007530047A (en) 2004-03-26 2005-03-24 Methods and biomarkers for detecting tumor endothelial cell proliferation
EP05731529A EP1733051A4 (en) 2004-03-26 2005-03-24 Method and biomarkers for detecting tumor endothelial cell proliferation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US55664504P 2004-03-26 2004-03-26
US60/556,645 2004-03-26

Publications (2)

Publication Number Publication Date
WO2005098448A2 true WO2005098448A2 (en) 2005-10-20
WO2005098448A3 WO2005098448A3 (en) 2005-12-29

Family

ID=35125712

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/009874 WO2005098448A2 (en) 2004-03-26 2005-03-24 Method and biomarkers for detecting tumor endothelial cell proliferation

Country Status (4)

Country Link
EP (1) EP1733051A4 (en)
JP (1) JP2007530047A (en)
CA (1) CA2561338A1 (en)
WO (1) WO2005098448A2 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013200287A (en) * 2012-03-26 2013-10-03 Kawasaki Gakuen POSITIVE RATE MEASUREMENT METHOD USING DOUBLE DYEING METHOD FOR Ki-67 INDEX CALCULATION
EP3364190A1 (en) * 2017-02-20 2018-08-22 Panka Cancer Research AG Method of detecting cancer or cancer cells

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000011164A1 (en) * 1998-08-20 2000-03-02 Incyte Pharmaceuticals, Inc. Human angiopoietin-3
DE60208792T2 (en) * 2001-06-22 2006-08-31 Merck & Co., Inc. TYROSINE KINASE INHIBITORS
US20040033495A1 (en) * 2001-08-03 2004-02-19 Eos Biotechnology, Inc. Methods of diagnosis of angiogenesis, compositions and methods of screening for angiogenesis modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1733051A4 *

Also Published As

Publication number Publication date
EP1733051A4 (en) 2008-03-26
WO2005098448A3 (en) 2005-12-29
CA2561338A1 (en) 2005-10-20
EP1733051A2 (en) 2006-12-20
JP2007530047A (en) 2007-11-01

Similar Documents

Publication Publication Date Title
JP6378273B2 (en) ECM composition, tumor microenvironment platform and method thereof
Klein et al. Wnt2 acts as a cell type–specific, autocrine growth factor in rat hepatic sinusoidal endothelial cells cross‐stimulating the VEGF pathway
Drake et al. Effects of intermittent parathyroid hormone treatment on osteoprogenitor cells in postmenopausal women
EP3263717A1 (en) Novel tissue protective erythropoietin receptor (nepor) and methods of use
US20180231554A1 (en) Programmed cell death (pd-1) inhibitor therapy for patients with pd-1-expressing cancers
US20100068717A1 (en) Cancer therapy prognosis
Fujita et al. High EGFR mRNA expression is a prognostic factor for reduced survival in pancreatic cancer after gemcitabine-based adjuvant chemotherapy
US20130143753A1 (en) Methods for predicting outcome of breast cancer, and/or risk of relapse, response or survival of a patient suffering therefrom
US20120046197A1 (en) Biomarkers of therapeutic responsiveness
Terasaki et al. Effects of prolactin on TSC2-null Eker rat cells and in pulmonary lymphangioleiomyomatosis
EP1601795A2 (en) Method for determining susceptibility of tumor to treatment with anti-neoplastic agent
WO2009014761A9 (en) Method for predicting response to tamoxifen
Hardwick et al. Identification of biomarkers for tumor endothelial cell proliferation through gene expression profiling
Caspar et al. In vitro cultivation of primary prostate cancer cells alters the molecular biomarker pattern
WO2005098448A2 (en) Method and biomarkers for detecting tumor endothelial cell proliferation
Massa et al. The phosphotyrosine phosphatase η mediates somatostatin inhibition of glioma proliferation via the dephosphorylation of ERK1/2
Jia et al. Wnt7a predicts poor prognosis, and contributes to growth and metastasis in tongue squamous cell carcinoma
Li et al. CXCR4 expression in bladder transitional cell carcinoma and its relationship with clinicopathological features
KR101141190B1 (en) Biomarker indicative of prostate cancer and diagnosis using the same
KR20110096292A (en) Use of socs6 as a hepatocellular carcinomar diagnostic marker
Hodges et al. Prostate cancer biomarkers: Current status
KR20190129765A (en) Method for diagnosing rheumatoid arthritis by using metabolomics and biomarker composition
Wolczyk et al. Stress granules assembly affects detection of mRNA in living cells by the NanoFlares; an important aspect of the technology
US11982675B2 (en) Method of assessing ABC transporter activity using fluorescent dye accumulation assay
Kajdaniuk et al. Transcriptional activity of TGFβ1 and its receptors genes in thyroid gland

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007505185

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2005731529

Country of ref document: EP

Ref document number: 2561338

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWP Wipo information: published in national office

Ref document number: 2005731529

Country of ref document: EP