WO2005067650A2 - Gene expression profiles and methods of use - Google Patents

Gene expression profiles and methods of use Download PDF

Info

Publication number
WO2005067650A2
WO2005067650A2 PCT/US2005/000518 US2005000518W WO2005067650A2 WO 2005067650 A2 WO2005067650 A2 WO 2005067650A2 US 2005000518 W US2005000518 W US 2005000518W WO 2005067650 A2 WO2005067650 A2 WO 2005067650A2
Authority
WO
WIPO (PCT)
Prior art keywords
expression
genes
level
gene products
cell
Prior art date
Application number
PCT/US2005/000518
Other languages
French (fr)
Other versions
WO2005067650A3 (en
Inventor
Nicole Pauloski
Mark J. Lynch
Liya Tang
Original Assignee
Bayer Pharmaceuticals Corporation
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 Bayer Pharmaceuticals Corporation filed Critical Bayer Pharmaceuticals Corporation
Publication of WO2005067650A2 publication Critical patent/WO2005067650A2/en
Publication of WO2005067650A3 publication Critical patent/WO2005067650A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds

Definitions

  • the present invention relates to gene expression profiles, microa ⁇ ays comprising nucleic acid sequences representing gene expression profiles, and methods of using gene expression profiles and microarrays.
  • one mechanism of activating unregulated growth is to increase the number of genes coding for oncogene proteins or to increase the level of expression of these oncogenes (e.g., in response to cellular or environmental changes), and another mechanism is to lose genetic material or to decrease the level of expression of genes that code for tumor suppressors.
  • This model is supported by the losses and gains of genetic material associated with glioma progression (Mikkelson, et al., J. Cellular Biochem. 46:3-8, 1991).
  • changes in the expression (transcription) levels of particular genes serve as signposts for the presence and progression of various cancers.
  • Compounds which are used as therapeutics to treat these various diseases presumably reverse some, or all, of these gene expression changes.
  • the expression change of at least some of these genes may therefore, be used as a method to monitor, or even predict, the efficacy of such therapeutics.
  • the analysis of these expression changes may be performed in the target tissue of interest (e.g., tumor) or in some surrogate cell population (e.g., peripheral blood leukocytes). In the latter case, co ⁇ elation of the gene expression changes with efficacy (e.g., tumor shrinkage or non-growth) must be especially strong for the expression change pattern to be used as a marker for efficacy.
  • VEGF vascular endothelial growth factor
  • PDGFR- ⁇ platelet- derived growth factor receptor-beta
  • VEGF and PDGFR- ⁇ work together to promote the formation of neovasculature and to recruit stromal cells to the maturing blood vessels.
  • VEGF is the most commonly up-regulated pro-angiogenic factor found in tumors. It is strongly induced by hypoxia and hypoglycemia, two conditions commly found in the center of poorly vascularized tumors (Takagi, et al. Invest. Ophthalmol. Vis. Sci. 37: 1311-1321, 1996). VEGF expression induces the formation of nascent blood vessels and is a survival factor for endothelial cells (Fe ⁇ ara, et al, Curr. Opin. Nephrol. Hypertens. 5:35-44, 1996).
  • PDGFR- ⁇ is expressed by endothelial cells and is up-regulated in tumors of the breast, colon, lung, and prostate. (George, et al, Semin. Oncol. 28:27-33, 2001). It promotes the recruitment of pericytes and smooth muscle cells to the neovasculature, which form the structural support for the mature vessels (Guo, et al. Am. J. Pathol. 162:1083-1093, 2003).
  • an anti-angiogenic agent must target both the nascent, VEGF-dependent and mature stromal supported, and PDGFR- ⁇ dependent vessels. It has been demonstrated experimentally that a selective VEGF inhibitor is ineffective against large tumors with well- established vasculature while a PDGFR- ⁇ selective inhibitor has poor anti-tumor activity (Bergers, et al, J. Clin. Invest. 111:1287-1295, 2003). However, VEGF and PDGFR- ⁇ inhibitors work synergistically to inhibit even large tumors.
  • PDGFR- ⁇ inhibition cause pericytes and smooth muscles cells to detach from endothelial cells, making them susceptible to apoptosis when VEGF signaling is inhibited. This combination of events leads to the collapse of the tumor vasculature and to tumor cell death. Therefore, the data suggests that small molecule inhibitors of PDGFR- ⁇ activity will be an important therapeutic mechanism in the treatment of cancer.
  • the present invention is directed to gene expression profiles, microa ⁇ ays comprising nucleic acid sequences representing said gene expression profiles, and methods of using said gene expression profiles and microa ⁇ ays.
  • the gene expression profile is an expression profile comprising one or more genes (e.g, SEQ ID NOs: 1-20) that demonstrate altered expression following exposure to a drug.
  • the expression profile comprises one or more genes that demonstrate altered expression following exposure to a PDGFR- ⁇ inhibitor.
  • the expression profile is an expression profile comprising one or more polypeptides (e.g, SEQ ID NOs: 21-40) that demonstrate altered expression following exposure to a drug.
  • the expression profile comprises one or more polypeptides that demonstrate altered expression following exposure to an PDGFR- ⁇ inhibitor.
  • the present invention is also directed to the discovery of the gene expression profile of cancer cells.
  • the gene expression profiles of the present invention represents the profiles of cancer cells following treatment with an PDGFR- ⁇ inhibitor.
  • PDGFR- ⁇ inhibitor As described in the Examples and in Table 1, compound-treated cancer cells have genes which are expressed at higher levels (i.e, which are up-regulated) and genes which are expressed at lower levels (i.e, which are down-regulated) relative to cells of the same type in untreated subjects.
  • PDGFR- ⁇ inhibitor As described in Table 1, it has been shown that following treatment with an PDGFR- ⁇ inhibitor, several genes are up-regulated or down- regulated in the cancer cells relative to the co ⁇ esponding untreated cancer cells. Genes which are up-regulated or down-regulated are refe ⁇ ed to herein as "genes characteristic of small molecule efficacy.”
  • microa ⁇ ays comprising one or more genes that demonstrate altered expression following exposure to an PDGFR- ⁇ inhibitor
  • the microa ⁇ ay may be a microa ⁇ ay comprising one or more genes selected from the group consisting of the genes listed in Table 1 (SEQ ID NO: 1-20).
  • the microa ⁇ ay may be a microa ⁇ ay comprising one or more biomarkers isolated from the group comprising the genes listed in Table 1 (SEQ ID NO: 1-20).
  • This invention also relates to methods for using said microarrays which include, but are not limited to, screening the effects of a drug or treatment (e.g, PDGFR- ⁇ inhibitor) on tissue or cell samples, screening toxicity effects on tissue or cell samples, identifying a disease state in a tissue or cell sample, providing a patient diagnosis, predicting a patient's response to treatment, distinguishing between control and drug-treated samples, distinguishing between normal and tumor samples, discovering novel drugs, and determining the level of gene expression in a tissue or cell sample.
  • a drug or treatment e.g, PDGFR- ⁇ inhibitor
  • Another embodiment of the present invention is a method for screening the effects of a drug (e.g, PDGFR- ⁇ inhibitor) on a tissue or cell sample comprising the step of analyzing the level of expression of one or more genes (e.g, SEQ ED NOs: 1-20) and/or gene products (e.g, SEQ ID NOs: 21-40), wherein the gene expression and/or gene product levels in the tissue or cell sample are analyzed before and after exposure to the drug, and a variation in the expression level of the gene and/or gene product is indicative of a drug effect or provides a patient diagnosis or predicts a patient's response to the treatment.
  • a drug e.g, PDGFR- ⁇ inhibitor
  • Another aspect of the present invention is a method for discovering novel drugs comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of the cells are analyzed before and after exposure to the drug, and a variation in the expression level of the gene and/or gene product is indicative of drug efficacy.
  • the invention further provides a method for identifying a compound useful for the treatment of cancer comprising administering to a subject with cancer a test compound, and measuring the activity of the polypeptide (e.g, the polypeptides encoded by SEQ ID NO: 21-40), wherein a change in the activity of the polypeptide is indicative of the test compound being useful for the treatment of cancer.
  • the polypeptide e.g, the polypeptides encoded by SEQ ID NO: 21-40
  • the invention provides methods which may be used to identify compounds which may act, for example, as regulators or modulators such as agonists and antagonists, partial agonists, inverse agonists, activators, co-activators, and inhibitors. Accordingly, the invention provides reagents and methods for regulating the expression of a polynucleotide or a polypeptide associated with cancer.
  • Reagents that modulate the expression, stability, or amount of a polynucleotide or the activity of the polypeptide may be a protein, a peptide, a peptidomimetic, a nucleic acid, a nucleic acid analogue (e.g, peptide nucleic acid, locked nucleic acid), or a small molecule.
  • the present invention also provides a method for providing a patient diagnosis comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of normal and patient samples are analyzed, and a variation in the expression level of the gene and/or gene product in the patient sample is diagnostic of a disease.
  • the patient samples include, but are not limited to, blood, amniotic fluid, plasma, semen, bone ma ⁇ ow, and tissue biopsy.
  • the present invention still further provides a method of diagnosing cancer in a subject comprising measuring the activity of the polypeptide in a subject suspected of having cancer, wherein if there is a difference in the activity of the polypeptide, relative to the activity of the polypeptide in a subject not suspected of having cancer, then the subject is diagnosed has having cancer.
  • the invention provides a method for detecting cancer in a patient sample in which an antibody to a protein is used to react with proteins in the patient sample.
  • Another aspect of the present invention is a method for distinguishing between normal and disease states comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of normal and disease tissues are analyzed, and a variation in the expression level of the gene and/or gene product is indicative of a disease state.
  • the invention pertains to a method of determining the phenotype of cells comprising detecting the differential expression, relative to normal cells, of at least one gene, wherein the gene is differentially expressed as compared to normal cells.
  • the invention pertains to a method of determining the phenotype of cells, comprising detecting the differential expression, relative to normal cells, of at least one polypeptide, wherein the protein is differentially expressed as compared to normal cells.
  • the invention pertains to a method for determining the phenotype of cells from a patient by providing a nucleic acid probe comprising a nucleotide sequence having at least about 10, at least about 15, at least about 25, or at least about 40 consecutive nucleotides, obtaining a sample of cells from a patient, optionally providing a second sample of cells substantially all of which are non-cancerous, contacting the nucleic acid probe under stringent conditions with mRNA of each of said first and second cell samples, and comparing (a) the amount of hybridization of the probe with mRNA of the first cell sample, with (b) the amount of hybridization of the probe with mRNA of the second cell sample, wherem a difference in the amount of hybridization with the mRNA of the first cell sample as compared to the amount of hybridization with the mRNA of the second cell sample is indicative of the phenotype of cells in the first cell sample.
  • the invention provides a test kit for identifying the presence of cancerous cells or tissues, comprising a probe/primer, for measuring a level of a nucleic acid in a sample of cells isolated from a patient.
  • the kit may further include instructions for using the kit, solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a nucleic acid susceptible to hybridization, solutions for lysing cells, or solutions for the purification of nucleic acids.
  • the invention provides a test kit for identifying the presence of cancer cells or tissues, comprising an antibody specific for a protein.
  • the kit further includes instructions for using the kit.
  • the kit may further include solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a polypeptide susceptible to the binding of an antibody, solutions for lysing cells, or solutions for the purification of polypeptides.
  • the invention provides a test kit for monitoring the efficacy of a compound or therapeutic in cancerous cells or tissues, comprising a probe/primer, for measuring a level of a nucleic acid in a sample of cells isolated from a patient.
  • the kit may further include instructions for using the kit, solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a nucleic acid susceptible to hybridization, solutions for lysing cells, or solutions for the purification of nucleic acids.
  • the invention provides a test kit for monitoring the efficacy of a compound or therapeutic in cancer cells or tissues, comprising an antibody specific for a protein.
  • the kit further includes instructions for using the kit.
  • the kit may further include solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a polypeptide susceptible to the binding of an antibody, solutions for lysing cells, or solutions for the purification of polypeptides.
  • This invention is also related to methods of identifying biomarkers comprising the steps of selecting a set of biomarker genes from a gene expression profile representing a disease or drug treatment.
  • a co ⁇ esponding normal cell of or "normal cell co ⁇ esponding to” or "normal counte ⁇ art cell of a diseased cell refers to a normal cell of the same type as that of the diseased cell.
  • An "address" on an a ⁇ ay refers to a location at which a element, for example, an oligonucleotide, is attached to the solid surface of the a ⁇ ay.
  • agonist is meant to refer to an agent that mimics or up- regulates (e.g, potentiates or supplements) the bioactivity of a protein.
  • An agonist may be a wild- type protein or derivative thereof having at least one bioactivity of the wild-type protein.
  • An agonist may also be a compound that up-regulates expression of a gene or which increases at least one bioactivity of a protein.
  • An agonist can also be a compound which increases the interaction of a polypeptide with another molecule, for example, a target peptide or nucleic acid.
  • amplification relates to the production of additional copies of a nucleic acid sequence.
  • amplification may be carried out using polymerase chain reaction (PCR) technologies which are well known in the art. (see, e.g., Dieffenbach, C. W. and G. S. Dveksler (1995) PCR Primer, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y.)
  • PCR polymerase chain reaction
  • Antagonist is meant to refer to an agent that down-regulates (e.g, suppresses or inhibits) at least one bioactivity of a protein.
  • An antagonist may be a compound which inhibits or decreases the interaction between a protein and another molecule, for example, a target peptide or enzyme substrate.
  • An antagonist may also be a compound that down-regulates expression of a gene or which reduces the amount of expressed protein present.
  • an PDGFR- ⁇ inhibitor is an example of such an antagonist.
  • antibody is intended to include whole antibodies, for example, of any isotype (IgG, IgA, IgM, IgE, etc.), and includes fragments thereof which are also specifically reactive with a vertebrate (e.g, mammalian) protein.
  • Antibodies may be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies.
  • the term includes segments of proteolytically-cleaved or recombinantly-prepared portions of an antibody molecule that are capable of selectively reacting with a certain protein.
  • Non-limiting examples of such proteolytic and/or recombinant fragments include Fab, F(ab')2, Fab', Fv, and single chain antibodies (scFv) containing a V[L] and/or V[H] domain joined by a peptide linker.
  • the scFv's may be covalently or non-covalently linked to form antibodies having two or more binding sites.
  • the subject invention includes polyclonal, monoclonal, or other purified preparations of antibodies and recombinant antibodies.
  • a ⁇ ay or “matrix” refer to an a ⁇ angement of addressable locations or "addresses" on a device.
  • the locations can be a ⁇ anged in two-dimensional a ⁇ ays, three- dimensional a ⁇ ays, or other matrix formats.
  • the number of locations may range from several to at least- hundreds of thousands. Most importantly, each location represents a totally independent reaction site.
  • a "nucleic acid a ⁇ ay” refers to an a ⁇ ay containing nucleic acid probes, such as oligonucleotides or larger portions of genes.
  • the nucleic acid on the a ⁇ ay may be single-stranded.
  • a ⁇ ays wherein the probes are oligonucleotides are refe ⁇ ed to as “oligonucleotide a ⁇ ays” or “oligonucleotide chips.”
  • a “rnicroa ⁇ ay,” also refe ⁇ ed to herein as a “biochip” or “biological chip,” is an a ⁇ ay of regions having a density of discrete regions of at least about 100/cm 2 , or at least about 1000/cm 2 .
  • the regions in a microa ⁇ ay have typical dimensions, for example, diameters, in the range of between about 10-250 ⁇ m, and are separated from other regions in the a ⁇ ay by about the same distance.
  • Bioactivity means an effector or antigenic function that is directly or indirectly performed by a polypeptide (whether in its native or denatured conformation), or by any subsequence thereof.
  • Biological activities include binding to polypeptides, binding to other proteins or molecules, activity as a DNA binding protein, as a transcription regulator, ability to bind damaged DNA, etc.
  • a bioactivity can be modulated by directly affecting the subject polypeptide.
  • a bioactivity can be altered by modulating the level of the polypeptide, such as by modulating expression of the co ⁇ esponding gene.
  • biological sample refers to a sample obtained from an organism or from components (e.g, cells) of an organism.
  • the sample may be of any biological tissue or fluid.
  • the sample may be a "clinical sample” which is a sample derived from a patient.
  • 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 taken for histological pu ⁇ oses.
  • biomarker encompasses a broad range of intra- and extra-cellular events as well as whole-organism physiological changes.
  • Biomarkers may be represent essentially any aspect of cell function, for example, but not limited to, levels or rate of production of signaling molecules, transcription factors, metabolites, gene transcripts as well as post-translational modifications of proteins.
  • Biomarkers may include whole genome analysis of transcript levels or whole proteome analysis of protein levels and/or modifications.
  • a biomarker may also refer to a gene or gene product which is up- or down-regulated in a compound-treated, diseased cell of a subject having the disease compared to an untreated diseased cell.
  • a biomarker is a gene or gene product that is characteristic of efficacy of a compound in a diseased cell or the response of that diseased cell to treatment by the compound.
  • a nucleotide sequence is "complementary" to another nucleotide sequence if each of the bases of the two sequences match, that is, are capable of forming Watson-Crick base pairs.
  • complementary strand is used herein interchangeably with the term “complement.”
  • the complement of a nucleic acid strand may be the complement of a coding strand or the complement of a non-coding strand.
  • Detection agents of genes refers to agents that can be used to specifically detect the gene or other biological molecules relating to it, for example, RNA transcribed from the gene or polypeptides encoded by the gene.
  • exemplary detection agents are nucleic acid probes, which hybridize to nucleic acids co ⁇ esponding to the gene, and antibodies.
  • Differential gene expression pattern between, for example, a normal cell and a disease cell refers to a pattern reflecting the differences in gene expression between a normal cell and a disease cell.
  • a differential gene expression pattern may also be obtained between a cell at one time point and a cell at another time point, or between a cell incubated or contacted with a compound and a cell that has not been incubated with or contacted with the compound.
  • cancer includes, but is not limited to, solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases.
  • solid tumors such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases.
  • lymphomas, sarcomas, and leukemias include lymphomas, sarcomas, and leukemias.
  • breast cancer examples include, but are not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
  • cancers of the respiratory tract include, but are not limited to, small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastema.
  • brain cancers include, but are not limited to, brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
  • Tumors of the male reproductive organs include, but are not limited to, prostate and testicular cancer.
  • Tumors of the female reproductive organs include, but are not limited to, endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumors of the digestive tract include, but are not limited to, anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumors of the urinary tract include, but are not limited to, bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
  • Eye cancers include, but are not limited to, intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to, hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to, squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to, laryngeal / hypopharyngeal / nasopharyngeal / oropharyngeal cancer, and lip and oral cavity cancer.
  • Lymphomas include, but are not limited to, AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to, sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
  • a diseased cell of cancer refers to a cell present in subjects having cancer. That is, a cell which is a modified form of a normal cell and is not present in a subject not having cancer, or a cell which is present in significantly higher or lower numbers in subjects having cancer relative to subjects not having cancer.
  • Equivalent is understood to include nucleotide sequences encoding functionally equivalent polypeptides. Equivalent nucleotide sequences may include sequences that differ by one or more nucleotide substitutions, additions, or deletions, such as allelic variants; and may, therefore, include sequences that differ from the nucleotide sequence of the nucleic acids refe ⁇ ed to in Table 1 due to the degeneracy of the genetic code.
  • expression profile which is used interchangeably herein with “gene expression profile” and “finge ⁇ rint” of a cell refers to a set of values representing mRNA levels of one or more genes in a cell.
  • An expression profile may comprise, for example, values representing expression levels of at least about 2 genes, at least about 5 genes, at least about 10 genes, or at least about 50, 100, 200 or more genes. Expression profiles may also comprise an mRNA level of a gene which is expressed at similar levels in multiple cells and conditions (e.g, a housekeeping gene such as GAPDH).
  • the term "gene” refers to a nucleic acid sequence that comprises control and coding sequences necessary for the production of a polypeptide or precursor.
  • the polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence.
  • the gene may be derived in whole or in part from any source known to the art, including a plant, a fungus, an animal, a bacterial genome or episome, eukaryotic, nuclear or plasmid DNA, cDNA, viral DNA, or chemically synthesized DNA.
  • a gene may contain one or more modifications in either the coding or the untranslated regions which could affect the biological activity or the chemical structure of the expression product, the rate of expression, or the manner of expression control.
  • Such modifications include, but are not limited to, mutations, insertions, deletions, and substitutions of one or more nucleotides.
  • the gene may constitute an uninterrupted coding sequence or it may include one or more introns, bound by the appropriate splice junctions.
  • Hybridization refers to any process by which a strand of nucleic acid binds with a complementary strand through base pairing.
  • two single-stranded nucleic acids "hybridize” when they form a double-stranded duplex.
  • the region of double-strandedness may include the full-length of one or both of the single-stranded nucleic acids, or all of one single- stranded nucleic acid and a subsequence of the other single-stranded nucleic acid, or the region of double-strandedness may include a subsequence of each nucleic acid.
  • Hybridization also includes the formation of duplexes which contain certain mismatches, provided that the two strands are still forming a double-stranded helix.
  • “Stringent hybridization conditions” refers to hybridization conditions resulting in essentially specific hybridization.
  • isolated refers to molecules separated from other DNAs or RNAs, respectively, that are present in the natural source of the macromolecule.
  • isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • an "isolated nucleic acid” may include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • isolated is also used herein to refer to polypeptides is meant to encompass both purified and recombinant polypeptides.
  • label and “detectable label” refer to a molecule capable of detection, including, but not limited to, radioactive isotopes, fluorophores, chemiluminescent moieties, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, dyes, metal ions, ligands (e.g, biotin or haptens), and the like.
  • fluorescer refers to a substance or a portion thereof which is capable of exhibiting fluorescence in the detectable range.
  • level of expression refers to the level of mRNA, as well as pre-mRNA nascent transcript(s), transcript processing intermediates, mature n RNA(s), and degradation products, encoded by a gene in the cell.
  • level of expression also refers to the level of protein or polypeptide in a cell.
  • nucleic acid refers to polynucleotides such as deoxyribonucleic acid (DNA) and, where appropriate, ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • Chromosomes, cDNAs, mRNAs, rRNAs, and ESTs are representative examples of molecules that may be refe ⁇ ed to as nucleic acids.
  • nucleic acid co ⁇ esponding to a gene refers to a nucleic acid that can be used for detecting the gene, for example, a nucleic acid which is capable of hybridizing specifically to the gene.
  • nucleic acid sample derived from RNA refers to one or more nucleic acid molecules (e.g, RNA or DNA) that may be synthesized from the RNA, and includes DNA produced from methods using PCR (e.g, RT-PCR).
  • oligonucleotide refers to a nucleic acid molecule comprising, for example, from about 10 to about 1000 nucleotides. Oligonucleotides for use in the present invention may be from about 15 to about 150 nucleotides, or from about 150 to about 1000 in length.
  • the oligonucleotide may be a naturally occurring oligonucleotide or a synthetic oligonucleotide. Oligonucleotides may be prepared by the phosphoramidite method (Beaucage and Carruthers, Tetrahedron Lett. 22:1859-62, 1981), or by the triester method (Matteucci, et al, J. Am. Chem. Soc. 103:3185, 1981), or by other chemical methods known in the art.
  • patient or "subject” as used herein includes mammals (e.g, humans and animals).
  • percent identical refers to sequence identity between two amino acid sequences or between two nucleotide sequences. For example, identity between two sequences may be determined by comparing a particular position in each sequence which may be aligned for potposes of comparison. When an equivalent position in the compared sequences is occupied by the same base or amino acid, then the molecules are identical at that position. When the equivalent site is occupied by the same or a similar amino acid residue (e.g, similar in steric and/or electronic nature), then the molecules may be refe ⁇ ed to as homologous (similar) at that position. Expression as a percentage of homology, similarity, or identity refers to a function of the number of identical or similar amino acids at positions shared by the compared sequences.
  • FASTA FASTA
  • BLAST BLAST
  • ENTREZ is available through the National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD.
  • percent identity of two sequences may be determined by the GCG program with a gap weight of 1 (e.g, each amino acid gap is weighted as if it were a single amino acid or nucleotide mismatch between the two sequences).
  • gap weight e.g, each amino acid gap is weighted as if it were a single amino acid or nucleotide mismatch between the two sequences.
  • Nucleic acid-encoded amino acid sequences may be used to search both protein and DNA databases. Databases with individual sequences are described in Methods in Enzymology, ed. Doolittle, supra. Databases include, for example, Genbank, EMBL, and DNA Database of Japan (DDBJ).
  • a nucleic acid or other molecule attached to an a ⁇ ay is refe ⁇ ed to as a "probe” or “capture probe.”
  • probe or capture probe
  • a gene-probe set may consist of, for example, about 2 to about 20 probes, from about 2 to about 10 probes, or about 5 probes.
  • the "profile" of a cell's biological state refers to the levels of various constituents of a cell that are known to change in response to drug treatments and other perturbations of the biological state of the cell.
  • Constituents of a cell include, for example, levels of RNA, levels of protein abundances, or protein activity levels.
  • protein protein
  • polypeptide peptide
  • An. expression profile in one cell is "similar" to an expression profile in another cell when the level of expression of the genes in the two profiles are sufficiently similar that the similarity is indicative of a common characteristic, for example, the same type of cell.
  • Small molecule refers to a composition with a molecular weight of less than about 5 kD or less than about 4 kD. Small molecules can be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids, or other organic or inorganic molecules. Many pharmaceutical companies have extensive libraries of chemical and/or biological mixtures, often fungal, bacterial, or algal extracts, which can be screened with any of the assays of the invention to identify compounds that modulate a bioactivity.
  • hybridization of a probe to a target site of a template nucleic acid refers to hybridization of the probe predominantly to the target, such that the hybridization signal can be clearly inte ⁇ reted.
  • such conditions resulting in specific hybridization vary depending on the length of the region of homology, the GC content of the region, and the melting temperature ("Tm") of the hybrid.
  • Tm melting temperature
  • hybridization conditions may vary in salt content, acidity, and temperature of the hybridization solution and the washes.
  • a "variant" of polypeptide refers to a polypeptide having an amino acid sequence in which one or more amino acid residues is altered.
  • the variant may have "conservative” changes, wherein a substituted amino acid has similar structural or chemical properties (e.g, replacement of leucine with isoleucine).
  • a variant may also have "nonconservative” changes (e.g, replacement of glycine with tryptophan).
  • Analogous minor variations may include amino acid deletions or insertions, or both.
  • Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological or immunological activity may be identified using computer programs well known in the art, for example, LASERGENE software (DNASTAR).
  • variants when used in the context of a polynucleotide sequence, may encompass a polynucleotide sequence related to that of a particular gene or the coding sequence thereof. This definition may also include, for example, "allelic,” “splice,” “species,” or “polymo ⁇ hic” variants.
  • a splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternate splicing of exons during mRNA processing.
  • the co ⁇ esponding polypeptide may possess additional functional domains or an absence of domains.
  • Species variants are polynucleotide sequences that vary from one species to another.
  • a polymo ⁇ hic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species.
  • Polymo ⁇ hic variants also may encompass "single nucleotide polymo ⁇ hisms" (SNPs) in which the polynucleotide sequence varies by one base. The presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state.
  • determining expression profiles with microa ⁇ ays involves the following steps: (a) obtaining an mRNA sample from a subject and preparing labeled nucleic acids therefrom (the "target nucleic acids” or “targets”); (b) contacting the target nucleic acids with an a ⁇ ay under conditions sufficient for the target nucleic acids to bind to the co ⁇ esponding probes on the a ⁇ ay, for example, by hybridization or specific binding; (c) optional removal of unbound targets from the a ⁇ ay; (d) detecting the bound targets, and (e) analyzing the results, for example, using computer based analysis methods.
  • nucleic acid probes or “probes” are nucleic acids attached to the a ⁇ ay
  • target nucleic acids are nucleic acids that are hybridized to the a ⁇ ay.
  • Nucleic acid specimens may be obtained from an individual to be tested using either “invasive” or “non-invasive” sampling means.
  • a sampling means is said to be “invasive” if it involves the collection of nucleic acids from within the skin or organs of an animal (including murine, human, ovine, equine, bovine, porcine, canine, or feline animal).
  • invasive methods include blood collection, semen collection, needle biopsy, pleural aspiration, umbilical cord biopsy, etc. Examples of such methods are discussed by Kim, et al, (J. Virol. 66:3879-3882, 1992); Biswas, et al, (Ann. NY Acad. Sci. 590:582-583, 1990); and Biswas, et al, (J. Clin. Microbiol. 29:2228-2233, 1991).
  • a “non-invasive” sampling means is one in which the nucleic acid molecules are recovered from an internal or external surface of the animal.
  • Examples of such "non-invasive" sampling means include, for example, "swabbing," collection of tears, saliva, urine, fecal material, sweat or perspiration, hair, etc.
  • PBLs peripheral blood leukocytes
  • cells may be isolated from other cells using a variety of techniques, such as isolation with an antibody binding to an epitope on the cell surface of the desired cell type.
  • desired cells are in a solid tissue
  • particular cells may be dissected, for example, by microdissection or by laser capture microdissection (LCM) (see, e.g., Bonner, et al. Science 278:1481, 1997; Emmert-Buck, et al. Science 274:998, 1996; Fend, et al. Am. J. Path. 154:61, 1999; and Murakami, et al. Kidney hit. 58:1346, 2000).
  • LCM laser capture microdissection
  • RNA may be extracted from tissue or cell samples by a variety of methods, for example, guanidium thiocyanate lysis followed by CsCl centrifugation (Chirgwin, et al. Biochemistry 18:5294-5299, 1979).
  • RNA from single cells may be obtained as described in methods for preparing cDNA libraries from single cells (see, e.g., Dulac, Cu ⁇ . Top. Dev. Biol. 36:245, 1998; Jena, et al, J. Immunol. Methods 190:199, 1996).
  • RNA sample can be further enriched for a particular species, h one embodiment, for example, poly(A)+ RNA may be isolated from an RNA sample, h particular, poly-T oligonucleotides may be immobilized on a solid support to serve as affinity ligands for mRNA. Kits for this pu ⁇ ose are commercially available, for example, the MessageMaker kit (Life Technologies, Grand Island, NY).
  • the RNA population may be enriched for sequences of interest, such as the genes described in Table 1 (e.g, SEQ ID NOs: 1-20). Enrichment may be accomplished, for example, by primer-specific cDNA synthesis, or multiple rounds of linear amplification based on cDNA synthesis and template-directed in vitro transcription (see, e.g., Wang, et al, Proc. Natl. Acad. Sci. USA 86:9717, 1989; Dulac, et al, supra; Jena, et al, supra).
  • RNA enriched or not in particular species or sequences
  • the population of RNA, enriched or not in particular species or sequences, may be further amplified. Such amplification is particularly important when using RNA from a single cell or a few cells.
  • a variety of amplification methods are suitable for use in the methods of the present invention, including, for example, PCR; ligase chain reaction (LCR) (see, e.g., Wu and Wallace, Genomics 4:560, 1989; Landegren, et al. Science 241: 1077, 1988); self-sustained sequence replication (SSR) (see, e.g., Guatelli, et al, Proc. Natl. Acad. Sci.
  • LCR ligase chain reaction
  • SSR self-sustained sequence replication
  • PCR technology Principles and Applications for DNA Amplification (ed. H. A. Erlich, Freeman Press, N.Y, N.Y, 1992); PCR Protocols: A Guide to Methods and Applications (eds. Innis, et al. Academic Press, San Diego, Calif, 1990); Mattila, et al. Nucleic Acids Res.
  • RNA amplification and cDNA synthesis may also be conducted in cells in situ (see, e.g., Eberwine, et al. Proc. Natl. Acad. Sci. USA 89:3010, 1992).
  • the target molecules will be labeled to permit detection of hybridization of the target molecules to a microa ⁇ ay.
  • the probe may comprise a member of a signal producing system and thus, is detectable, either directly or through combined action with one or more additional members of a signal producing system.
  • directly detectable labels include isotopic and fluorescent moieties inco ⁇ orated, usually by a covalent bond, into a moiety of the probe, such as a nucleotide monomeric unit (e.g, dNMP of the primer), or a photoactive or chemically active derivative of a detectable label which can be bound to a functional moiety of the probe molecule.
  • Nucleic acids may be labeled during or after enrichment and/or amplification of RNAs.
  • reverse transcription may be carried out in the presence of a dNTP conjugated to a detectable label, for example, a ftuorescently labeled dNTP.
  • the cDNA or RNA probe may be synthesized in the absence of detectable label and may be labeled subsequently, for example, by inco ⁇ orating biotinylated dNTPs or rNTP, or some similar means (e.g, photo-cross-linking a psoralen derivative of biotin to RNAs), followed by addition of labeled streptavidin (e.g, phycoerythrin-conjugated streptavidin) or the equivalent.
  • labeled streptavidin e.g, phycoerythrin-conjugated streptavidin
  • Fluorescent moieties or labels of interest include coumarin and its derivatives (e.g, 7- amino-4-methylcoumarin, aminocoumarin); bodipy dyes such as Bodipy FL and cascade blue; fluorescein and its derivatives (e.g, fluorescein isothiocyanate, Oregon green); rhodamine dyes (e.g, Texas red, tetramethylrhodamine); eosins and erythrosins; cyanine dyes (e.g, Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7); FluorX, macrocyclic chelates of lanthanide ions (e.g, quantum dyeTM); fluorescent energy transfer dyes such as thiazole orange-ethidium heterodimer, TOTAB, dansyl, etc.
  • bodipy dyes such as Bodipy FL and cascade blue
  • fluorescein and its derivatives e.g, fluorescein isothiocyanate, Oregon green
  • Chemiluminescent labels include luciferin and 2,3-dihydrophthalazinediones, for example, luminol.
  • Labels may also be members of a signal producing system that act in concert with one or more additional members of the same system to provide a detectable signal.
  • Illustrative of such labels are members of a specific binding pair, such as ligands, for example, biotin, fluorescein, digoxigenin, antigen, polyvalent cations, chelator groups and the like.
  • Members may specifically bind to additional members of the signal producing system, and the additional members may provide a detectable signal either directly or indirectly, for example, an antibody conjugated to a fluorescent moiety or an enzymatic moiety capable of converting a substrate to a chromogenic product (e.g, alkaline phosphatase conjugate antibody and the like).
  • Additional labels of interest include those that provide a signal only when the probe with which it is associated is specifically bound to a target molecule.
  • Such labels include "molecular beacons" as described in Tyagi and Kramer (Nature Biotech. 14:303, 1996) and EP 0 070 685 Bl.
  • Other labels of interest include those described in U.S. Patent No. 5,563,037; WO 97/17471; and WO 97/17076.
  • the target nucleic acid may not be labeled.
  • hybridization may be determined, for example, by plasmon resonance (see, e.g., Thiel, et al. Anal. Chem. 69:4948, 1997).
  • a plurality (e.g, 2, 3, 4, 5, or more) of sets of target nucleic acids are labeled and used in one hybridization reaction ("multiplex" * analysis).
  • one set of nucleic acids may co ⁇ espond to RNA from one cell and another set of nucleic acids may co ⁇ espond to RNA from another cell.
  • the plurality of sets of nucleic acids may be labeled with different labels, for example, different fluorescent labels (e.g, fluorescein and rhodamine) which have distinct emission spectra so that they can be distinguished.
  • the sets may then be mixed and hybridized simultaneously to one microa ⁇ ay (see, e.g., Shena, et al. Science 270:467-470, 1995).
  • Examples of distinguishable labels for use when hybridizing a plurality of target nucleic acids to one a ⁇ ay are well known in the art and include: two or more different emission wavelength fluorescent dyes such as Cy3 and Cy5; combination of fluorescent proteins and dyes such as phicoerythrin and Cy5; two or more isotopes with different energy of emission such as 32 P and 33 P; gold or silver particles with different scattering spectra; labels which generate signals under different treatment conditions such as temperature, pH, treatment with additional chemical agents, etc.; or generate signals at different time points after treatment.
  • the quality of labeled nucleic acids may be evaluated prior to hybridization to an a ⁇ ay.
  • the GeneChip ® Test3 A ⁇ ay from Affymetrix (Santa Clara, CA) may be used for that pu ⁇ ose.
  • This a ⁇ ay contains probes representing a subset of characterized genes from several organisms including mammals.
  • the quality of a labeled nucleic acid sample can be determined by hybridization of a fraction of the sample to an a ⁇ ay.
  • Microa ⁇ ays for use according to the invention include one or more probes of genes characteristic of small molecule efficacy.
  • the microa ⁇ ay comprises probes co ⁇ esponding to one or more of genes selected from the group consisting of genes which are up- regulated in cancer and genes which are down-regulated in cancer.
  • the microa ⁇ ay may comprise, for example, probes co ⁇ esponding to at least 2, at least 5, at least 10, at least 100 or more characteristic of small molecule efficacy.
  • the microa ⁇ ay may comprise probes co ⁇ esponding to each gene or gene product listed in Table 1.
  • a microa ⁇ ay may contain from 2 to 20 probes co ⁇ esponding to one gene or about 5 to 10.
  • the probes may co ⁇ espond to the full-length RNA sequence or complement thereof of genes characteristic of small molecule efficacy, or the probe may co ⁇ espond to a portion thereof, which portion is of sufficient length to permit specific hybridization.
  • Such probes may comprise from about 50 nucleotides to about 100, 200, 500, or 1000 nucleotides or more than 1000 nucleotides.
  • microa ⁇ ays may contain oligonucleotide probes, consisting of about 10 to 50 nucleotides, about 15 to 30 nucleotides, or about 20-25 nucleotides.
  • the probes are may be single-stranded and will have sufficient complementarity to its target to provide for the desired level of sequence specific hybridization.
  • the a ⁇ ays used in the present invention will have a site density of greater than 100 different probes per cm 2 .
  • the a ⁇ ays may have a site density of, for example, greater than 500/cm 2 , greater than about 1000/cm 2 , or greater than about 10,000/cm 2 .
  • the a ⁇ ays may have, for example, more than 100 different probes on a single substrate, greater than about 1000 different probes, greater than about 10,000 different probes, or greater than 100,000 different probes on a single substrate.
  • a ⁇ ays may also include control and reference nucleic acids.
  • Control nucleic acids include, for example, prokaryotic genes such as bioB, bioC and bioD, ere from PI bacteriophage or polyA controls, such as dap, lys, phe, thr, and t ⁇ .
  • Reference nucleic acids allow the normalization of results from one experiment to another and the comparison of multiple experiments on a quantitative level.
  • Exemplary reference nucleic acids include housekeeping genes of known expression levels, for example, GAPDH, hexokinase, and actin.
  • an a ⁇ ay of oligonucleotides may be synthesized on a solid support.
  • exemplary solid supports include glass, plastics, polymers, metals, metalloids, ceramics, organics, etc. Using chip masking technologies and photoprotective chemistry, it is possible to generate ordered a ⁇ ays of nucleic acid probes.
  • a ⁇ ays which are known, for example, as “DNA chips” or very large scale immobilized polymer a ⁇ ays (“VLSIPSTM” a ⁇ ays), may include millions of defined probe regions on a substrate having an area of about 1 cm 2 to several cm 2 , thereby inco ⁇ orating from a few to millions of probes (see, e.g., U.S. Patent No. 5,631,734).
  • a nucleic acid probe may be at least, for example, about 10, 15, 20, 25, 30, 50, 100 or more nucleotides, and may comprise the full-length gene.
  • probes may be those that hybridize specifically to the genes listed in Table 1.
  • Nucleic acid probes may be obtained, for example, by PCR amplificartion of gene segments from genomic, cDNA (e.g, RT-PCR), or cloned sequences.
  • cDNA probes may be prepared according to methods known in the art and further described herein, for example, by reverse-transcription PCR (RT-PCR) of RNA using sequence specific primers. Sequences of genes or cDNA from which probes are generated may be obtained, for example, from GenBank, other public databases, or publications.
  • Oligonucleotide probes may also be synthesized by standard methods known in the art, for example, by automated DNA synthesizer or any other chemical method.
  • phosphorothioate oligonucleotides may be synthesized by the method of Stein, et al, (Nucl. Acids Res. 16:3209, 1988), and methylphosphonate oligonucleotides may be prepared by controlled pore glass polymer supports (see, e.g., Sarin, et al, Proc. Natl. Acad. Sci. U.S.A. 85:7448-7451, 1988).
  • the oligonucleotide may be a 2'-)-methylribonucleotide (Inoue, et al, Nucl. Acids Res. 15:6131-6148, 1987), or a chimeric RNA-DNA analog (Inoue, et al, FEBS Lett. 215:327-330, 1987).
  • Nucleic acid probes may be natural nucleic acids or chemically modified nucleic acids (e.g, composed of nucleotide analogs); however, the probes should possess activated hydroxyl groups compatible with the linking chemistry.
  • the protective groups may be photolabile, or the protective groups may be labile under certain chemical conditions (e.g, acid).
  • the surface of the solid support may contain a composition that generates acids upon exposure to light. Thus, exposure of a region of the substrate to light generates acids in that region that remove the protective groups in the exposed region.
  • the synthesis method may use 3'- protected 5'-0- phosphoramidite-activated deoxynucleoside. In this case, the oligonucleotide is synthesized in the 5' to 3' direction, which results in a free 5' end.
  • oligonucleotides of an a ⁇ ay may be synthesized using a 96-well automated multiplex oligonucleotide synthesizer (A.M.O.S.) that is capable of producing thousands of oligonucleotides (see, e.g., Lashkari, et al, Proc. Natl. Acad. Sci. USA 93: 7912, 1995).
  • A.M.O.S. automated multiplex oligonucleotide synthesizer
  • labeled nucleic acids may be contacted with the a ⁇ ay under conditions sufficient for binding between the target nucleic acid and the probe on the a ⁇ ay.
  • the hybridization conditions may be selected to provide for the desired level of hybridization specificity; that is, conditions sufficient for hybridization to occur between the labeled nucleic acids and probes on the microa ⁇ ay.
  • Hybridization may be carried out in conditions permitting essentially specific hybridization.
  • the length and GC content of the nucleic acid will determine the thermal melting point and thus, the hybridization conditions necessary for obtaining specific hybridization of the probe to the target nucleic acid. These factors are well known to a person of skill in the art, and may also be tested in assays. An extensive guide to nucleic acid hybridization may be found in
  • stringent conditions may be selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • Highly stringent conditions may be selected to be equal to the Tm point for a particular probe.
  • dissociation temperature Td is used to define the temperature at which at least half of the probe dissociates from a perfectly matched target nucleic acid.
  • Tm or Td a variety of techniques for estimating the Tm or Td are available, and generally are described in Tijssen, supra.
  • G-C base pairs in a duplex are estimated to contribute about 3°C to the Tm
  • A-T base pairs are estimated to contribute about 2°C, up to a theoretical maximum of about 80-100°C.
  • more sophisticated models of Tm and Td are available in which G-C stacking interactions, solvent effects, the desired assay temperature, and the like are taken into account.
  • non-specific binding or background signal may be reduced by the use of a detergent (e.g, C-TAB) or a blocking reagent (e.g, sperm DNA, cot-1 DNA, etc.) during the hybridization, hi one embodiment, the hybridization may be performed in the presence of about 0.5 mg/ml DNA (e.g, herring sperm DNA).
  • a detergent e.g, C-TAB
  • a blocking reagent e.g, sperm DNA, cot-1 DNA, etc.
  • the hybridization may be performed in the presence of about 0.5 mg/ml DNA (e.g, herring sperm DNA).
  • the use of blocking agents in hybridization is well known to those of skill in the art (see, e.g., Tijssen, supra).
  • a ⁇ ays may be employed for each physiological source or the same a ⁇ ay may be screened multiple times.
  • the above methods may be varied to provide for multiplex analysis by employing different and distinguishable labels for the different target populations (e.g, different physiological sources). According to this multiplex method, the same a ⁇ ay may be used at the same time for each of the different target populations.
  • the methods described above result in the production of hybridization patterns of labeled target nucleic acids on the a ⁇ ay surface.
  • the resultant hybridization patterns of labeled nucleic acids may be visualized or detected in a variety of ways, with the particular mamier of detection selected based on the particular label of the target nucleic acid.
  • Representative detection means include scintillation counting, autoradiography, fluorescence measurement, colorimetric measurement, light emission measurement, light scattering, and the like.
  • One such method of detection utilizes an a ⁇ ay scanner that is commercially available (Affymetrix, Santa Clara, CA), for example, the 417TM A ⁇ ayer, the 418TM A ⁇ ay Scanner, or the Agilent GeneA ⁇ ayTM Scanner.
  • This scanner is controlled from a system computer with an interface and easy-to-use software tools. The output may be directly imported into or directly read by a variety of software applications. Scanning devices are described in, for example, U.S. Patent Nos. 5,143,854 and 5,424,186.
  • the fluorescence emissions at each site of a transcript a ⁇ ay may be detected by scanning confocal laser microscopy.
  • a laser may be used that allows simultaneous specimen illumination at wavelengths specific to the two fluorophores and emissions from the two fluorophores may be analyzed simultaneously (see, e.g., Shalon, et al. Genome Res. 6:639-645, 1996).
  • the a ⁇ ays may be scanned with a laser fluorescent scanner with a computer controlled X-Y stage and a microscope objective. Fluorescence laser scanning devices are described in Shalon, et al, supra.
  • the data will typically be reported to a data analysis operation.
  • the data obtained by the reader from the device may be analyzed using a digital computer.
  • the computer will be appropriately programmed for receipt and storage of the data from the device, as well as for analysis and reporting of the data gathered, for example, subtraction of the background, deconvolution of multi-color images, flagging or removing artifacts, verifying that controls have performed properly, normalizing the signals, inte ⁇ reting fluorescence data to determine the amount of hybridized target, normalization of background and single base mismatch hybridizations, and the like.
  • a system comprises a search function that allows one to search for specific patterns, for example, patterns relating to differential gene expression, for example, between the expression profile of a cancer cell and the expression profile of a counte ⁇ art normal cell in a subject.
  • a system allows one to search for patterns of gene expression between more than two samples.
  • Various algorithms are available for analyzing gene expression profile data, for example, the type of comparisons to perform.
  • One embodiment for identifying such groups of genes involves clustering algorithms (for reviews of clustering algorithms, see, e.g., Fukunaga, 1990, Statistical Pattern Recognition, 2nd Ed, Academic Press, San Diego; Everitt, 1974, Cluster Analysis, London: Heinemann Educ. Books; Hartigan, 1975, Clustering Algorithms, New York: Wiley; Sneath and Sokal, 1973, Numerical Taxonomy, Freeman; Anderberg, 1973, Cluster Analysis for Applications, Academic Press: New York).
  • Clustering may be based on other characteristics of the genes, for example, their level of expression (see, e.g., U.S. Patent No. 6,203,987), or permit clustering of time curves (see, e.g. U.S. Patent No. 6,263,287).
  • Examples of clustering algorithms include K-means clustering and hierarchical clustering. Clustering may also be achieved by visual inspection of gene expression data using a graphical representation of the data (e.g. a "heat map").
  • An example of software which contains clustering algorithms and a means to graphically represent gene expression data is Spotfire DecisionSite (Spotfire, Inc, Somerville, Massachusetts and Goteborg, Sweden).
  • Comparison of the expression levels of one or more genes characteristic of small molecule efficacy with reference expression levels may be conducted using computer systems.
  • expression levels may be obtained from two cells and these two sets of expression levels may be introduced into a computer system for comparison.
  • one set of expression levels is entered into a computer system for comparison with values that are already present in the computer system, or in computer-readable form that is then entered into the computer system.
  • the computer system may also contain a database comprising values representing levels of expression of one or more genes characteristic of small molecule efficacy.
  • the database may contain one or more expression profiles of genes characteristic of small molecule efficacy in different cells.
  • the invention provides a computer-readable form of the gene expression profile data, or of values co ⁇ esponding to the level of expression of at least one gene characteristic of cancer in a diseased cell.
  • the values may be mRNA expression levels obtained from experiments, for example, microa ⁇ ay analysis.
  • the values may also be mRNA levels normalized relative to a reference gene whose expression is constant in numerous cells under numerous conditions (e.g, GAPDH).
  • the values in the computer may be ratios of, or differences between, normalized or non-normalized mRNA levels in different samples.
  • the expression profiles expression profiles from cancer cells of one or more subjects, which cells are treated in vivo or in vitro with a drug, for example, an PDGFR- ⁇ inhibitor.
  • Expression data of a cell of a subject treated in vitro or in vivo with the drug is entered into a computer and the computer is instructed to compare the data entered to the data in the computer, and to provide results indicating whether the expression data input into the computer are more similar to those of a cell of a subject that is responsive to the drug or more similar to those of a cell of a subject that is not responsive to the drug.
  • the results indicate whether the subject is likely to respond to the treatment with the drug or unlikely to respond to it.
  • the invention also provides a machine-readable or computer-readable medium including program instructions for performing the following steps: (i) comparing a plurality of values co ⁇ esponding to expression levels of one or more genes characteristic of small molecule efficacy in a query cell with a database including records comprising reference expression or expression profile data of one or more reference cells and an annotation of the type of cell; and (ii) indicating to which cell the query cell is most similar based on similarities of expression profiles.
  • the reference cells may be cells from subjects at different stages of cancer.
  • the reference cells may also be cells from subjects responding or not responding to a particular drug treatment and optionally incubated in vitro or in vivo with the drug.
  • the reference cells may also be cells from subjects responding or not responding to several different treatments, and the computer system indicates a prefe ⁇ ed treatment for the subject.
  • the invention provides a method for selecting a therapy for a patient having cancer, the method comprising: (i) providing the level of expression of one or more genes characteristic of small molecule efficacy in a diseased cell of the patient; (ii) providing a plurality of reference profiles, each associated with a therapy, wherein the subject expression profile and each reference profile has a plurality of values, each value representing the level of expression of a gene characteristic of cancer; and (iii) selecting the reference profile most similar to the subject expression profile, to thereby select a therapy for said patient, hi one embodiment, step (iii) may be performed by a computer.
  • the most similar reference profile may be selected by weighing a comparison value of the plurality using a weight value associated with the co ⁇ esponding expression data.
  • the relative abundance of an mRNA in two biological samples may be scored as a perturbation and its magnitude determined (i.e, the abundance is different in the two sources of mRNA tested), or as not perturbed (i.e, the relative abundance is the same). In various embodiments, a difference between the two sources of RNA is scored as a perturbation. Perturbations r ⁇ ay be used by a computer for calculating and expression comparisons.
  • the invention also provides methods for designing and optimizing drugs for cancer, for example, those which have been identified as described herein, hi one embodiment, compounds may be screened by comparing the expression level of one or more genes characteristic of small molecule efficacy following incubation of a diseased cell of cancer or similar cell with the test compound.
  • the expression level of the genes may be determined using microa ⁇ ays, and comparing the gene expression profile of a cell in response to the test compound with the gene expression profile of a normal cell co ⁇ esponding to a diseased cell of cancer (a "reference profile").
  • the expression profile may also be compared to that of a diseased cell of cancer.
  • the comparisons may be done by introducing the gene expression profile data of the cell treated with drug into a computer system comprising reference gene expression profiles, which are stored in a computer readable form, using appropriate algorithms.
  • Test compounds may be screened for those that alter the level of expression of genes characteristic of small molecule efficacy. Such compounds, that is, compounds which are capable of normalizing the expression of essentially all genes characteristic of small molecule efficacy, are candidate therapeutics.
  • the efficacy of the compounds may then be tested in additional in vitro and in vivo assays, and in animal models (e.g, xenograft model).
  • the test compound may be administered to the test animal, and one or more symptoms of the disease may be monitored for improvement of the condition of the animal.
  • Expression of one or more genes characteristic of small molecule efficacy may also be measured before and after administration of the test compound to the animal. A normalization of the expression of one or more of these genes is indicative of the efficiency of the compound for treating cancer in the animal.
  • a test compound may be administered to the test animal (either normal or cancer-containing) at the same doses that have been observed to be efficacious in treating cancer in that animal model. Blood may be drawn from the animal at various time points (e.g, 1, 4, 7, and 24 hours following the first, mid-point, and last day of a regimen of multiple day dosing).
  • RNA may be isolated from PBLs, and can be used to generate probes for hybridization to microa ⁇ ays.
  • the hybridization results may then be analyzed using computer programs and databases, as described above.
  • the resulting expression profile may be compared directly to the analogous profile from the treated cancer tissue for similarities or simply co ⁇ elated with efficacy (e.g, in terms of doses and time points) in the animal model.
  • human blood may be treated ex vivo with a therapeutic compound at a dose consistent with the therapeutic dose in the animal model, or at a dose that is consistent with known plasma levels of the therapeutic dose in the animal model.
  • the blood may be treated (e.g, rocking at 37°C) with the therapeutic compound immediately, or after some period of incubation time (e.g, 24 hours) to allow for gene expression to re-equilibrate after the blood draw.
  • the blood may also be treated with the therapeutic compound for various timepoints (e.g, 4 and 24 hours), and then PBL RNA isolated and used to create a probe for hybridization to a microa ⁇ ay.
  • a compound solubilization agent e.g, DMSO
  • the resulting expression profile may be compared directly to the analogous profile from the treated cancer tissue for similarities or simply co ⁇ elated with efficacy (e.g, in terms of doses and time points) in the animal model.
  • the toxicity of the candidate therapeutic compound may be evaluated, for example, by determining whether the compound induces the expression of genes known to be associated with a toxic response. Expression of such toxicity related genes may be determined in different cell types, for example, those that are known to express the genes, hi fact, alterations in gene expression may serve as a more sensitive marker of human toxicity than routine preclinical safety studies. Microa ⁇ ays may be used for detecting changes in the expression of genes known to be associated with a toxic response. It may be possible to perform proof of concept studies demonstrating that changes in gene expression levels may predict toxic events that were not identified by routine preclinical safety testing (see, e.g., Huang, et al, Toxicol. Sci. 63: 196-207, 2001; Waring, et al, Toxicol. Appl. Pharamacol. 175:28-42, 2001).
  • Drug screening may be performed by adding a test compound to a sample of cells, and monitoring the effect. A parallel sample which does not receive the test compound may also be monitored as a control.
  • the treated and untreated cells are then compared by any suitable phenotypic criteria, including but not limited to microscopic analysis, viability testing, ability to replicate, histological examination, the level of a particular RNA or polypeptide associated with the cells, the level of enzymatic activity expressed by the cells or cell lysates, and the ability of the cells to interact with other cells or compounds. Differences between treated and untreated cells indicates effects attributable to the test compound.
  • Desirable effects of a test compound include an effect on any phenotype that was confe ⁇ ed by the cancer-associated marker nucleic acid sequence. Examples include a test compound that limits the overabundance of mRNA, limits production of the encoded protein, or limits the functional effect of the protein. The effect of the test compound would be apparent when comparing results between treated and untreated cells.
  • the present invention provides nucleic acid sequences which are differentially regulated in cancer, and a method for identifying such sequences.
  • the present invention provides a method for identifying a nucleotide sequence which is differentially regulated in a subject with cancer, comprising: hybridizing a nucleic acid sample co ⁇ esponding to RNA obtained from the subject to a nucleic acid sample comprising one or more nucleic acid molecules of known identity; and measuring the hybridization of the nucleic acid sample to the one or more nucleic acid molecules of known identity, wherein a difference in the hybridization of the nucleic acid sample to the one or more nucleic acid molecules of known identity relative to a nucleic acid sample obtained from a subject without cancer is indicative of the differential expression of the nucleotide sequence in a subject with cancer.
  • the present invention provides a method for identifying nucleic acid sequences which are differentially regulated in a subject with cancer comprising isolating messenger RNA from a subject, generating cRNA from the mRNA sample, hybridizing the cRNA to a microa ⁇ ay comprising a plurality of nucleic acid molecules stably associated with discrete locations on the a ⁇ ay, and identifying patterns of hybridization of the cRNA to the a ⁇ ay.
  • a nucleic acid molecule which hybridizes to a given location on the a ⁇ ay is said to be differentially regulated if the hybridization signal is, for example, higher or lower than the hybridization signal at the same location on an identical a ⁇ ay hybridized with a nucleic acid sample obtained from a subject that does not have cancer.
  • Expression patterns may be used to derive a panel of biomarkers that can be used to predict the efficacy of drug treatment in the patients.
  • the biomarkers may consist of gene expression levels from microa ⁇ ay experiments on RNA isolated from biological samples, RNA isolated from frozen samples of tumor biopsies, or mass spectrometry-derived protein masses in the serum.
  • plasma may be applied to a hydrophobic SELDI- target, washed extensively in water, and analyzed by the SELDI-Tof mass spectrometer. This may be repeated on 100 or more patient samples. The protein profiles resulting from the intensities of some 16,000 m/z values in each sample would be statistically analyzed in order to identify sets of specific m/z values that are predictive of drug efficacy. Identical experiments using other SELDI- targets, such as ion-exchange or IMAC surfaces, could also be conducted. These will capture different subsets of the proteins present in plasma. Furthermore, the plasma may be denatured and prefractionated prior to application onto the SELDI target.
  • the present invention provides methods for determining whether a subject is at risk for developing a disease or condition characterized by unwanted cell proliferation by detecting biomarkers, that is, nucleic acids and/or polypeptide markers for cancer.
  • human tissue samples may be screened for the presence and/or absence of biomarkers identified herein.
  • samples could consist of needle biopsy cores, surgical resection samples, lymph node tissue, or serum.
  • these methods include obtaining a biopsy, which is optionally fractionated by cryostat sectioning to enrich tumor cells to about 80% of the total cell population.
  • nucleic acids extracted from these samples may be amplified using techniques well known in the art. The levels of selected markers detected would be compared with statistically valid groups of metastatic, non-metastatic malignant, benign, or normal tissue samples.
  • the diagnostic method comprises determining whether a subject has an abnormal mRNA and/or protein level of the biomarkers, such as by Northern blot analysis, reverse transcription-polymerase chain reaction (RT-PCR), in situ hybridization, immunoprecipitation, Western blot hybridization, or immunohistochemistry.
  • cells may be obtained from a subject and the levels of the biomarkers, protein, or mRNA level, are determined and compared to the level of these markers in a healthy subject.
  • An abnormal level of the biomarker polypeptide or mRNA levels is likely to be indicative of cancer.
  • the method comprises using a nucleic acid probe to determine the presence of cancerous cells in a tissue from a patient. Specifically, the method comprises:
  • nucleic acid probe comprising a nucleotide sequence, for example, at least 10, 15, 25 or 40 nucleotides, and up to all or nearly all of the coding sequence which is complementary to a portion of the coding sequence of a nucleic acid sequence and is differentially expressed in tumors cells;
  • RNA of each of said first and second tissue samples e.g, in a Northern blot or in situ hybridization assay
  • the method comprises in situ hybridization with a probe derived from a given marker nucleic acid sequence.
  • the method comprises contacting the labeled hybridization probe with a sample of a given type of tissue potentially containing cancerous or pre-cancerous cells as well as normal cells, and determining whether the probe labels some cells of the given tissue type to a degree significantly different than the degree to which it labels other cells of the same tissue type.
  • Also within the invention is a method of determining the phenotype of a test cell from a given human tissue, for example, whether the cell is (a) normal, or (b) cancerous or precancerous, by contacting the mRNA of a test cell with a nucleic acid probe, for example, at least about 10, 15, 25, or 40 nucleotides, and up to all or nearly all of a sequence which is complementary to a portion of the coding sequence of a nucleic acid sequence, and which is differentially expressed in tumor cells as compared to normal cells of the given tissue type; and determining the approximate amount of hybridization of the probe to the mRNA, an amount of hybridization either more or less than that seen with the mRNA of a normal cell of that tissue type being indicative that the test cell is cancerous or pre-cancerous.
  • a nucleic acid probe for example, at least about 10, 15, 25, or 40 nucleotides, and up to all or nearly all of a sequence which is complementary to a portion of the coding sequence of
  • the above diagnostic assays may be carried out using antibodies to detect the protein product encoded by the marker nucleic acid sequence.
  • the assay would include contacting the proteins of the test cell with an antibody specific for the gene product of a nucleic acid, the marker nucleic acid being one which is expressed at a given control level in normal cells of the same tissue type as the test cell, and determining the approximate amount of immunocomplex formation by the antibody and the proteins of the test cell, wherein a statistically significant difference in the amount of the immunocomplex formed with the proteins of a test cell as compared to a normal cell of the same tissue type is an indication that the test cell is cancerous or pre-cancerous.
  • Another such method includes the steps of: providing an antibody specific for the gene product of a marker nucleic acid sequence, the gene product being present in cancerous tissue of a given tissue type at a level more or less than the level of the gene product in non-cancerous tissue of the same tissue type; obtaining from a patient a first sample of tissue of the given tissue type, which sample potentially includes cancerous cells; providing a second sample of tissue of the same tissue type (which may be from the same patient or from a normal control, e.g.
  • this second sample containing normal cells and essentially no cancerous cells; contacting the antibody with protein (which may be partially purified, in lysed but unfractionated cells, or in situ) of the first and second samples under conditions permitting immunocomplex formation between the antibody and the marker nucleic acid sequence product present in the samples; and comparing (a) the amount of immunocomplex formation in the first sample, with (b) the amount of immunocomplex formation in the second sample, wherein a statistically significant difference in the amount of immunocomplex formation in the first sample less as compared to the amount of immunocomplex formation in the second sample is indicative of the presence of cancerous cells in the first sample of tissue.
  • protein which may be partially purified, in lysed but unfractionated cells, or in situ
  • the subject invention further provides a method of determining whether a cell sample obtained from a subject possesses an abnormal amount of marker polypeptide which comprises (a) obtaining a cell sample from the subject, (b) quantitatively determining the amount of the marker polypeptide in the sample so obtained, and (c) comparing the amount of the marker polypeptide so determined with a known standard, so as to thereby determine whether the cell sample obtained from the subject possesses an abnormal amount of the marker polypeptide.
  • marker polypeptides may be detected by immunohistochemical assays, dot-blot assays, ELISA, and the like.
  • Immunoassays are commonly used to quantitate the levels of proteins in cell samples, and many other immunoassay techniques are known in the art.
  • the invention is not limited to a particular assay procedure, and therefore, is intended to include both homogeneous and heterogeneous procedures.
  • exemplary immunoassays which may be conducted according to the invention include fluorescence polarization immunoassay (FPIA), fluorescence immunoassay (FIA), enzyme immunoassay (EIA), nephelometric inhibition immunoassay (NIA), enzyme-linked immunosorbent assay (ELISA), and radioimmunoassay (RIA).
  • FPIA fluorescence polarization immunoassay
  • FIA fluorescence immunoassay
  • EIA enzyme immunoassay
  • NIA nephelometric inhibition immunoassay
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • An indicator moiety, or label group may be attached to the subject antibodies and is selected so as to meet the needs of various uses of the method which are often dictated by the availability of assay equipment and compatible immunoas
  • the level of the encoded product, or alternatively the level of the polypeptide, in a biological fluid (e.g, blood or urine) of a patient may be determined as a way of monitoring the level of expression of the marker nucleic acid sequence in cells of that patient.
  • a biological fluid e.g, blood or urine
  • Such a method would include the steps of obtaining a sample of a biological fluid from the patient, contacting the sample (or proteins from the sample) with an antibody specific for an encoded marker polypeptide, and determining the amount of immune complex formation by the antibody, with the amount of immune complex formation being indicative of the level of the marker encoded product in the sample. This determination is particularly instructive when compared to the amount of immune complex formation by the same antibody in a control sample taken from a normal individual or in one or more samples previously or subsequently obtained from the same person.
  • the method may be used to determine the amount of marker polypeptide present in a cell, which in turn may be co ⁇ elated with progression of a hype ⁇ roliferative disorder.
  • the level of the marker polypeptide may be used predictively to evaluate whether a sample of cells contains cells which are, or are predisposed towards becoming, transformed cells.
  • the subject method may be used to assess the phenotype of cells which are known to be transformed, the phenotyping results being useful in planning a particular therapeutic regimen. For example, very high levels ofthe marker polypeptide in sample cells is a powerful diagnostic and prognostic marker for a cancer. The observation of marker polypeptide levels may be utilized in decisions regarding, for example, the use of more aggressive therapies.
  • one aspect ofthe present invention relates to diagnostic assays for determining, in the context of cells isolated from a patient, if the level of a marker polypeptide is significantly reduced in the sample cells.
  • the term "significantly reduced” refers to a cell phenotype wherein the cell possesses a reduced cellular amount ofthe marker polypeptide relative to a normal cell of similar tissue origin.
  • the assay evaluates the level of marker polypeptide in the test cells, and may compare the measured level with marker polypeptide detected in at least one control cell, for example, a normal cell and/or a transformed cell of known phenotype.
  • Another aspect ofthe subject invention is the ability to quantitate the level of marker polypeptide as determined by the number of cells associated with a normal or abnormal marker polypeptide level.
  • the number of cells with a particular marker polypeptide phenotype may then be co ⁇ elated with patient prognosis, hi one embodiment ofthe invention, the marker polypeptide phenotype of a lesion is determined as a percentage of cells in a biopsy which are found to have abnormally high/low levels ofthe marker polypeptide.
  • Such expression may be detected by immunohistochemical assays, dot-blot assays, ELISA, and the like.
  • immunohistochemical staining may be used to determine the number of cells having the marker polypeptide phenotype.
  • a multiblock of tissue may be taken from the biopsy or other tissue sample and subjected to proteolytic hydrolysis, employing such agents as protease K or pepsin.
  • proteolytic hydrolysis employing such agents as protease K or pepsin.
  • tissue samples are fixed by treatment with a reagent such as formalin, glutaraldehyde, methanol, or the like.
  • a reagent such as formalin, glutaraldehyde, methanol, or the like.
  • the samples are then incubated with an antibody (e.g, a monoclonal antibody) with binding specificity for the marker polypeptides.
  • This antibody may be conjugated to a label for subsequent detection of binding.
  • Samples are incubated for a time sufficient for formation ofthe immunocomplexes. Binding ofthe antibody is then detected by virtue of a label conjugated to this antibody.
  • a second labeled antibody may be employed, for example, which is specific for the isotype ofthe anti-marker polypeptide antibody.
  • labels which may be employed include radionuclides, fluorescers, chemiluminescers, enzymes, and the like.
  • the substrate for the enzyme may be added to the samples to provide a colored or fluorescent product.
  • suitable enzymes for use in conjugates include horseradish peroxidase, alkaline phosphatase, malate dehydrogenase, and the like. Where not commercially available, such antibody-enzyme conjugates are readily produced by techniques known to those skilled in the art.
  • the assay is performed as a dot blot assay. The dot blot assay finds particular application where tissue samples are employed as it allows determination ofthe average amount ofthe marker polypeptide associated with a single cell by co ⁇ elating the amount of marker polypeptide in a cell-free extract produced from a predetermined number of cells.
  • tumor cells ofthe same type e.g, lung and/or colon tumor cells
  • the invention provides for a battery of tests utilizing a number of probes ofthe invention, i order to improve the reliability and/or accuracy ofthe diagnostic test.
  • the present invention also provides a method wherein nucleic acid probes are immobilized on a DNA chip in an organized a ⁇ ay.
  • Oligonucleotides may be bound to a solid support by a variety of processes, including lithography.
  • a chip may hold up to 250,000 oligonucleotides.
  • These nucleic acid probes comprise a nucleotide sequence, for example, at least about 12, 15, 25, or 40 nucleotides in length, and up to all or nearly all of a sequence which is complementary to a portion ofthe coding sequence of a marker nucleic acid sequence and is differentially expressed in tumor cells.
  • the present invention provides significant advantages over the available tests for various cancers, because it increases the reliability ofthe test by providing an a ⁇ ay of nucleic acid markers on a single chip.
  • the method includes obtaining a biopsy, which is optionally fractionated by cryostat sectioning to enrich tumor cells.
  • the DNA or RNA is then extracted, amplified, and analyzed with a DNA chip to determine the presence of absence ofthe marker nucleic acid sequences.
  • the nucleic acid probes are spotted onto a substrate in a two- dimensional matrix or a ⁇ ay.
  • Samples of nucleic acids may be labeled and then hybridized to the probes.
  • Double-stranded nucleic acids, comprising the labeled sample nucleic acids bound to probe nucleic acids, may be detected once the unbound portion ofthe sample is washed away.
  • the probe nucleic acids may be spotted on substrates including glass, nitrocellulose, etc.
  • the probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions.
  • the sample nucleic acids can be labeled using radioactive labels, fluorophores, chromophores, etc.
  • the invention contemplates using a panel of antibodies which are generated against the marker polypeptides of this invention.
  • a panel of antibodies may be used as a reliable diagnostic probe for cancer.
  • the assay ofthe present invention comprises contacting a biopsy sample containing cells, for example, lung cells, with a panel of antibodies to one or more ofthe encoded products to determine the presence or absence ofthe marker polypeptides.
  • the diagnostic methods ofthe subject invention may also be employed as follow-up to treatment, for example, quantitation ofthe level of marker polypeptides may be indicative ofthe effectiveness of cu ⁇ ent or previously employed cancer therapies as well as the effect of these therapies upon patient prognosis.
  • marker nucleic acids or marker polypeptides may be utilized as part of a diagnostic panel for initial detection, follow-up screening, detection of reoccu ⁇ ence, and post- treatment monitoring for chemotherapy or surgical treatment.
  • the present invention makes available diagnostic assays and reagents for detecting gain and/or loss of marker polypeptides from a cell in order to aid in the diagnosis and phenotyping of proliferative disorders arising from, for example, tumorigenic transfo ⁇ nation of cells.
  • the diagnostic assays described above may be adapted to be used as prognostic assays, as well. Such an application takes advantage ofthe sensitivity ofthe assays ofthe invention to events which take place at characteristic stages in the progression of a tumor. For example, a given marker gene may be up- or down-regulated at a very early stage, perhaps before the cell is i ⁇ eversibly committed to developing into a malignancy, while another marker gene may be characteristically up- or down-regulated only at a much later stage.
  • Such a method could involve the steps of contacting the mRNA of a test cell with a nucleic acid probe derived from a given marker nucleic acid which is expressed at different characteristic levels in cancerous or precancerous cells at different stages of tumor progression, and determining the approximate amount of hybridization ofthe probe to the mRNA ofthe cell, such amount being an indication of the level of expression ofthe gene in the cell, and thus an indication ofthe stage of tumor progression ofthe cell; alternatively, the assay may be carried out with an antibody specific for the gene product ofthe given marker nucleic acid, contacted with the proteins ofthe test cell.
  • a battery of such tests will disclose not only the existence and location of a tumor, but also will allow the clinician to select the mode of treatment most appropriate for the tumor, and to predict the likelihood of success of that treatment.
  • the methods ofthe invention may also be used to follow the clinical course of a tumor.
  • the assay ofthe invention may be applied to a tissue sample from a patient; following treatment ofthe patient for the cancer, another tissue sample is taken and the test repeated. Successful treatment will result in either removal of all cells which demonstrate differential expression characteristic ofthe cancerous or precancerous cells, or a substantial increase in expression ofthe gene in those cells, perhaps approaching or even su ⁇ assing normal levels.
  • the invention provides methods for determining whether a subject is at risk for developing a disease, such as a predisposition to develop cancer, associated with abe ⁇ ant activity of a polypeptide, wherein the abe ⁇ ant activity ofthe polypeptide is characterized by detecting the presence or absence of a genetic lesion characterized by at least one of (a) an alteration affecting the integrity of a gene encoding a marker polypeptides, or (b) the mis- expression ofthe encoding nucleic acid.
  • such genetic lesions may be detected by ascertaining the existence of at least one of (i) a deletion of one or more nucleotides from the nucleic acid sequence, (ii) an addition of one or more nucleotides to the nucleic acid sequence, (iii) a substitution of one or more nucleotides ofthe nucleic acid sequence, (iv) a gross chromosomal rearrangement ofthe nucleic acid sequence, (v) a gross alteration in the level of a messenger RNA transcript ofthe nucleic acid sequence, (vi) abe ⁇ ant modification ofthe nucleic acid sequence, such as ofthe methylation pattern ofthe genomic DNA, (vii) the presence of a non-wild type splicing pattern of a messenger RNA transcript ofthe gene, (viii) a non-wild type level ofthe marker polypeptide, (ix) allelic loss ofthe gene, and/or (x) inappropriate post-translational modification ofthe marker polypeptide.
  • the present invention provides assay techniques for detecting lesions in the encoding nucleic acid sequence. These methods include, but are not limited to, methods involving sequence analysis, Southern blot hybridization, restriction enzyme site mapping, and methods involving detection of absence of nucleotide pairing between the nucleic acid to be analyzed and a probe.
  • Specific diseases or disorders are associated with specific allelic variants of polymo ⁇ hic regions of certain genes, which do not necessarily encode a mutated protein.
  • the presence of a specific allelic variant of a polymo ⁇ hic region of a gene in a subject may render the subject susceptible to developing a specific disease or disorder.
  • Polymo ⁇ hic regions in genes may be identified, by determining the nucleotide sequence of genes in populations of individuals. If a polymo ⁇ hic region is identified, then the link with a specific disease may be determined by studying specific populations of individuals, for example, individuals which developed a specific disease, such as cancer.
  • a polymo ⁇ hic region may be located in any region of a gene, for example, exons, in coding or non-coding regions of exons, introns, and promoter region.
  • a nucleic acid composition comprising a nucleic acid probe including a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence of a gene or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with the subject genes or naturally occu ⁇ ing mutants thereof.
  • the nucleic acid of a cell is rendered accessible for hybridization, the probe is contacted with the nucleic acid ofthe sample, and the hybridization ofthe probe to the sample nucleic acid is detected.
  • Such techniques may be used to detect lesions or allelic variants at either the genomic or mRNA level, including deletions, substitutions, etc, as well as to determine mRNA transcript levels.
  • An example of a detection method is allele specific hybridization using probes overlapping the mutation or polymo ⁇ hic site and having about 5, 10, 20, 25, or 30 nucleotides around the mutation or polymo ⁇ hic region.
  • several probes capable of hybridizing specifically to allelic variants are attached to a solid phase support, for example, a "chip.” Mutation detection analysis using these chips comprising oligonucleotides, also termed "DNA probe a ⁇ ays" is described, for example, by Cronin, et al, (Human Mutation 7:244, 1996).
  • a chip may comprise all the allelic variants of at least one polymo ⁇ hic region of a gene. The solid phase support is then contacted with a test nucleic acid and hybridization to the specific probes is detected. Accordingly, the identity of numerous allelic variants of one or more genes may be identified in a simple hybridization experiment.
  • detection ofthe lesion comprises utilizing the probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligase chain reaction (LCR) (see, e.g., Landegran, et al. Science 241: 1077-1080, 1988; Nakazaw, et al, Proc. Natl. Acad. Sci. USA 91:360-364, 1994), the latter of which can be particularly useful for detecting point mutations in the gene (see, e.g., Abravaya, et al, Nuc.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • the method includes the steps of (i) collecting a sample of cells from a patient, (ii) isolating nucleic acid (e.g, genomic, mRNA, or both) from the cells ofthe sample, (iii) contacting the nucleic acid sample with one or more primers which specifically hybridize to a nucleic acid sequence under conditions such that hybridization and amplification ofthe nucleic acid (if present) occurs, and (iv) detecting the presence or absence of an amplification product, or detecting the size ofthe amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any ofthe techniques used for detecting mutations described herein.
  • nucleic acid e.g, genomic, mRNA, or both
  • the invention thus, also encompasses methods of screening for agents which inhibit or enhance the expression ofthe nucleic acid markers in vitro, comprising exposing a cell or tissue in which the marker nucleic acid mRNA is detectable in cultured cells to an agent in order to determine whether the agent is capable of inhibiting or enhancing production ofthe mRNA; and determining the level of mRNA in the exposed cells or tissue, wherein a decrease in the level of the mRNA after exposure ofthe cell line to the agent is indicative of inhibition ofthe marker nucleic acid mRNA production and an increase in mRNA levels is indicative of enhancement of maker mRNA production.
  • the screening method may include in vitro screening of a cell or tissue in which marker protein is detectable in cultured cells to an agent suspected of inhibiting or enhancing production ofthe marker protein; and determining the level ofthe marker protein in the cells or tissue, wherein a decrease in the level of marker protein after exposure ofthe cells or tissue to the agent is indicative of inhibition of marker protein production and an increase on the level of marker protein is indicative of enhancement of marker protein production.
  • the invention also encompasses in vivo methods of screening for agents which inhibit or enhance expression ofthe marker nucleic acids, comprising exposing a subject having tumor cells in which marker mRNA or protein is detectable to an agent suspected of inhibiting or enhancing production of marker mRNA or protein; and determining the level of marker mRNA or protein in tumor cells ofthe exposed mammal.
  • a decrease in the level of marker mRNA or protein after exposure ofthe subject to the agent is indicative of inhibition of marker nucleic acid expression and an increase in the level of marker mRNA or protein is indicative of enhancement of marker nucleic acid expression.
  • the invention provides a method comprising incubating a cell expressing the marker nucleic acids with a test compound and measuring the mRNA or protein level.
  • the invention further provides a method for quantitatively determining the level of expression ofthe marker nucleic acids in a cell population, and a method for determining whether an agent is capable of increasing or decreasing the level of expression ofthe marker nucleic acids in a cell population.
  • the method for determining whether an agent is capable of increasing or decreasing the level of expression ofthe marker nucleic acids in a cell population comprises the steps of (a) preparing cell extracts from control and agent-treated cell populations, (b) isolating the marker polypeptides from the cell extracts, and (c) quantifying (e.g, in parallel) the amount of an immunocomplex formed between the marker polypeptide and an antibody specific to said polypeptide.
  • the marker polypeptides of this invention may also be quantified by assaying for its bioactivity.
  • Agents that induce an increase in the marker nucleic acid expression may be identified by their ability to increase the amount of immunocomplex formed in the treated cell as compared with the amount ofthe immunocomplex formed in the control cell.
  • agents that decrease expression ofthe marker nucleic acid may be identified by their ability to decrease the amount ofthe immunocomplex formed in the treated cell extract as compared to the control cell.
  • a biomarker associated with the anti-cancer agent may be analyzed in a biological sample (e.g, tumor sample, plasma) before, during, and following treatment.
  • a biological sample e.g, tumor sample, plasma
  • Another approach to monitor treatment is an evaluation of serum proteomic spectra.
  • plasma samples may be subjected to mass spectroscopy (e.g, surface-enhanced laser deso ⁇ tion and ionization) and a proteomic spectra may be generated for each patient.
  • mass spectroscopy e.g, surface-enhanced laser deso ⁇ tion and ionization
  • a set of spectra, derived from analysis of plasma from patients before and during treatment may be analyzed by an iterative searching algorithm, which can identify a proteomic pattern that completely discriminates the treated samples from the untreated samples. The resulting pattern may then be used to predict the clinical benefit following treatment.
  • RNA isolated from cells derived from whole blood from patients before and during treatment may be used to generate blood cell gene expression profiles utilizing Affymetrix GeneChip technology and algorithms. These gene expression profiles may then predict the clinical benefit from treatment with a particular anti-cancer agent.
  • Drug-, disease-, and genetic-stimuli have been shown to produce metabolic-specific changes in baseline urine profiles that are indicative ofthe timeline and magnitude ofthe metabolic response to the stimuli. These analyses are multi- variant and therefore use pattern recognition techniques to improve data inte ⁇ retation. Urinary metabolic profiles may be co ⁇ elated with clinical endpoints to determine the clinical benefit.
  • kits for determining the expression level of genes characteristic of small molecule efficacy may be useful for identifying subjects that are predisposed to developing cancer or who have cancer, as well as for identifying and validating therapeutics for cancer.
  • the kit comprises a computer readable medium on which is stored one or more gene expression profile of diseased cells of cancer, or at least values representing levels of expression of one or more genes characteristic of small molecule efficacy in a diseased cell.
  • the computer readable medium can also comprise gene expression profiles of counte ⁇ art normal cells, diseased cells treated with a drug, and any other gene expression profile described herein.
  • the kit can comprise expression profile analysis software capable of being loaded into the memory of a computer system.
  • a kit can comprise a microarray comprising probes of genes characteristic of small molecule efficacy.
  • a kit can comprise one or more probes or primers for detecting the expression level of one or more genes characteristic of small molecule efficacy and/or a solid support on which probes attached and which can be used for detecting expression of one or more genes characteristic of small molecule efficacy in a sample.
  • a kit may further comprise nucleic acid controls, buffers, and instructions for use.
  • kits provide compositions for treating cancer.
  • a kit can also comprise one or more nucleic acids co ⁇ esponding to one or more genes characteristic of small molecule efficacy (e.g, for use in treating a patient having cancer).
  • the nucleic acids can be included in a plasmid or a vector (e.g, a viral vector).
  • kits comprise a polypeptide encoded by a gene characteristic of cancer or an antibody to a polypeptide.
  • kits comprise compounds identified herein as agonists or antagonists of genes characteristic of small molecule efficacy.
  • the compositions may be pharmaceutical compositions comprising a pharmaceutically acceptable excipient.
  • mice Female nude mice ranging between 11-19 weeks of age, and with an average weight of approximately 18-25 grams were used in these studies.
  • human Breast (MDA-MB-231) cancer cell lines were grown in tissue culture to approximately 70% confluency. Cells were harvested on the day of implant (5 x 10 6 cells/mouse), and were suspended in Hanks Balanced Salt Solution from the time of harvest to the time of implant at which time each mouse received a 0.2 ml injection ofthe cell suspension ofthe appropriate cell innoculum. The cells were injected subcutaneously in the right flank of each mouse and tumors were monitored for growth. Time of staging (dosing) was deteratined when tumors reached a size of 200-300 mm 3 .
  • the vehicle used for the PDGFR- ⁇ inhibitor was 40% PEG, 10% ethanol, and 50% 50 mM methanesulfonic acid. Dosing was oral at 25 mg/kg and 75 mg/kg b.i.d. for 4 days. On day 4, at 4 hours after the dose, mice were euthanized and three tumor xenografts for each drug level and three vehicle-treated tumors were harvested and snap frozen. Table 1 describes the genes that were up- and down- regulated greater than or equal to two-fold in the PDGFR- ⁇ inhibitor-treated xenografts relative to vehicle-treated xenografts.
  • a ⁇ ays were stained with Phycoerythrm-conjugated Streptavidin, placed in an Agilent GeneA ⁇ ay Scanner and then exposed to a 488 n laser, causmg excitation of the phycoerythrin.
  • the Microa ⁇ ay Suite 5.0 software digitally converts the intensity of light given off by the a ⁇ ay into a numeric value indicative of levels of gene expression. Because each a ⁇ ay represented a single animal sample, treated animals were compared to the vehicle animals and relative fold changes of genes were obtained. Those genes increased or decreased by at least 2- fold change were considered significant and chosen for further analysis and are listed on Table 1.

Abstract

The present invention relates to gene expression profiles, microarrays comprising nucleic acid sequences representing gene expression profiles, and methods of using expression profiles and microarrays. The invention also provides methods and compositions for diagnostic assays for detecting cancer and therapeutic methods and compositions for treating cancer. The invention also provides methods for designing, identifying, and optimizing therapeutics for cancer.

Description

GENE EXPRESSION PROFILES AND METHODS OF USE
[001] This application claims benefit of U.S. Provisional Application Serial No. 60/534,825, filed January 6, 2004, the contents of which are incoφorated herein by reference in their entirety.
FIELD OF THE INVENTION
[002] The present invention relates to gene expression profiles, microaπays comprising nucleic acid sequences representing gene expression profiles, and methods of using gene expression profiles and microarrays.
BACKGROUND OF THE INVENTION
[003] Many disease states are characterized by differences in the expression levels of various genes either through changes in the copy number of the genetic DNA or through changes in levels of transcription of particular genes (e.g., through control of initiation, provision of RNA precursors, RNA processing, etc.). For example, losses and gains of genetic material play an important role in malignant transformation and progression. These gains and losses are thought to be "driven" by at least two kinds of genes, oncogenes and tumor suppressor genes. Oncogenes are positive regulators of tumorgenesis, while tumor suppressor genes are negative regulators of tumorgenesis (Marshall, Cell 64:313-326, 1991; Weinberg, Science 254: 1138-1146, 1991). Therefore, one mechanism of activating unregulated growth is to increase the number of genes coding for oncogene proteins or to increase the level of expression of these oncogenes (e.g., in response to cellular or environmental changes), and another mechanism is to lose genetic material or to decrease the level of expression of genes that code for tumor suppressors. This model is supported by the losses and gains of genetic material associated with glioma progression (Mikkelson, et al., J. Cellular Biochem. 46:3-8, 1991). Thus, changes in the expression (transcription) levels of particular genes (e.g., oncogenes or tumor suppressors) serve as signposts for the presence and progression of various cancers.
[004] Compounds which are used as therapeutics to treat these various diseases (e.g., cancer) presumably reverse some, or all, of these gene expression changes. The expression change of at least some of these genes may therefore, be used as a method to monitor, or even predict, the efficacy of such therapeutics. The analysis of these expression changes may be performed in the target tissue of interest (e.g., tumor) or in some surrogate cell population (e.g., peripheral blood leukocytes). In the latter case, coπelation of the gene expression changes with efficacy (e.g., tumor shrinkage or non-growth) must be especially strong for the expression change pattern to be used as a marker for efficacy. [005] Even for well-established tumors, blocking angiogenesis can lead to tumor regression if the right combination of growth factor signaling pathways are inhibited. Two of the most important angiogenic growth factors are vascular endothelial growth factor (VEGF) and platelet- derived growth factor receptor-beta (PDGFR-β) (Guo, et al., Am. J. Pathol. 162:1083-1093, 2003; Feπara, Am. J. Physiol. Cell Physiol. 280:C1358-1366, 2001; Nakanishi, et al. Mod. Pathol. 10:341-347, 1997; Feπara, et al, Cuπ. Opin. Nephrol. Hypertens. 5:35-44, 1996). VEGF and PDGFR-β work together to promote the formation of neovasculature and to recruit stromal cells to the maturing blood vessels. VEGF is the most commonly up-regulated pro-angiogenic factor found in tumors. It is strongly induced by hypoxia and hypoglycemia, two conditions commly found in the center of poorly vascularized tumors (Takagi, et al. Invest. Ophthalmol. Vis. Sci. 37: 1311-1321, 1996). VEGF expression induces the formation of nascent blood vessels and is a survival factor for endothelial cells (Feπara, et al, Curr. Opin. Nephrol. Hypertens. 5:35-44, 1996). PDGFR-β is expressed by endothelial cells and is up-regulated in tumors of the breast, colon, lung, and prostate. (George, et al, Semin. Oncol. 28:27-33, 2001). It promotes the recruitment of pericytes and smooth muscle cells to the neovasculature, which form the structural support for the mature vessels (Guo, et al. Am. J. Pathol. 162:1083-1093, 2003).
[006] To be effective, an anti-angiogenic agent must target both the nascent, VEGF-dependent and mature stromal supported, and PDGFR-β dependent vessels. It has been demonstrated experimentally that a selective VEGF inhibitor is ineffective against large tumors with well- established vasculature while a PDGFR-β selective inhibitor has poor anti-tumor activity (Bergers, et al, J. Clin. Invest. 111:1287-1295, 2003). However, VEGF and PDGFR-β inhibitors work synergistically to inhibit even large tumors. The inhibition of PDGFR-β cause pericytes and smooth muscles cells to detach from endothelial cells, making them susceptible to apoptosis when VEGF signaling is inhibited. This combination of events leads to the collapse of the tumor vasculature and to tumor cell death. Therefore, the data suggests that small molecule inhibitors of PDGFR-β activity will be an important therapeutic mechanism in the treatment of cancer.
SUMMARY OF THE INVENTION
[007] The present invention is directed to gene expression profiles, microaπays comprising nucleic acid sequences representing said gene expression profiles, and methods of using said gene expression profiles and microaπays.
[008] In one embodiment of the present invention, the gene expression profile is an expression profile comprising one or more genes (e.g, SEQ ID NOs: 1-20) that demonstrate altered expression following exposure to a drug. For example, the expression profile comprises one or more genes that demonstrate altered expression following exposure to a PDGFR-β inhibitor. [009] In another embodiment, the expression profile is an expression profile comprising one or more polypeptides (e.g, SEQ ID NOs: 21-40) that demonstrate altered expression following exposure to a drug. For example, the expression profile comprises one or more polypeptides that demonstrate altered expression following exposure to an PDGFR-β inhibitor.
[010] The present invention is also directed to the discovery of the gene expression profile of cancer cells. The gene expression profiles of the present invention represents the profiles of cancer cells following treatment with an PDGFR-β inhibitor. As described in the Examples and in Table 1, compound-treated cancer cells have genes which are expressed at higher levels (i.e, which are up-regulated) and genes which are expressed at lower levels (i.e, which are down-regulated) relative to cells of the same type in untreated subjects. As described in Table 1, it has been shown that following treatment with an PDGFR-β inhibitor, several genes are up-regulated or down- regulated in the cancer cells relative to the coπesponding untreated cancer cells. Genes which are up-regulated or down-regulated are refeπed to herein as "genes characteristic of small molecule efficacy."
[011] Also within the scope of the present invention are microaπays comprising one or more genes that demonstrate altered expression following exposure to an PDGFR-β inhibitor, hi another embodiment of the present invention, the microaπay may be a microaπay comprising one or more genes selected from the group consisting of the genes listed in Table 1 (SEQ ID NO: 1-20). In a further embodiment, the microaπay may be a microaπay comprising one or more biomarkers isolated from the group comprising the genes listed in Table 1 (SEQ ID NO: 1-20).
[012] In addition, it is another aspect of the invention to provide methods and reagents for the prediction, diagnosis, prognosis, and therapy of cancer.
[013] This invention also relates to methods for using said microarrays which include, but are not limited to, screening the effects of a drug or treatment (e.g, PDGFR-β inhibitor) on tissue or cell samples, screening toxicity effects on tissue or cell samples, identifying a disease state in a tissue or cell sample, providing a patient diagnosis, predicting a patient's response to treatment, distinguishing between control and drug-treated samples, distinguishing between normal and tumor samples, discovering novel drugs, and determining the level of gene expression in a tissue or cell sample.
[014] Another embodiment of the present invention is a method for screening the effects of a drug (e.g, PDGFR-β inhibitor) on a tissue or cell sample comprising the step of analyzing the level of expression of one or more genes (e.g, SEQ ED NOs: 1-20) and/or gene products (e.g, SEQ ID NOs: 21-40), wherein the gene expression and/or gene product levels in the tissue or cell sample are analyzed before and after exposure to the drug, and a variation in the expression level of the gene and/or gene product is indicative of a drug effect or provides a patient diagnosis or predicts a patient's response to the treatment.
[015] Another aspect of the present invention is a method for discovering novel drugs comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of the cells are analyzed before and after exposure to the drug, and a variation in the expression level of the gene and/or gene product is indicative of drug efficacy.
[016] The invention further provides a method for identifying a compound useful for the treatment of cancer comprising administering to a subject with cancer a test compound, and measuring the activity of the polypeptide (e.g, the polypeptides encoded by SEQ ID NO: 21-40), wherein a change in the activity of the polypeptide is indicative of the test compound being useful for the treatment of cancer.
[017] The invention, thus, provides methods which may be used to identify compounds which may act, for example, as regulators or modulators such as agonists and antagonists, partial agonists, inverse agonists, activators, co-activators, and inhibitors. Accordingly, the invention provides reagents and methods for regulating the expression of a polynucleotide or a polypeptide associated with cancer. Reagents that modulate the expression, stability, or amount of a polynucleotide or the activity of the polypeptide may be a protein, a peptide, a peptidomimetic, a nucleic acid, a nucleic acid analogue (e.g, peptide nucleic acid, locked nucleic acid), or a small molecule.
[018] The present invention also provides a method for providing a patient diagnosis comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of normal and patient samples are analyzed, and a variation in the expression level of the gene and/or gene product in the patient sample is diagnostic of a disease. The patient samples include, but are not limited to, blood, amniotic fluid, plasma, semen, bone maπow, and tissue biopsy.
[019] The present invention still further provides a method of diagnosing cancer in a subject comprising measuring the activity of the polypeptide in a subject suspected of having cancer, wherein if there is a difference in the activity of the polypeptide, relative to the activity of the polypeptide in a subject not suspected of having cancer, then the subject is diagnosed has having cancer.
[020] In another embodiment, the invention provides a method for detecting cancer in a patient sample in which an antibody to a protein is used to react with proteins in the patient sample. [021] Another aspect of the present invention is a method for distinguishing between normal and disease states comprising the step of analyzing the level of expression of one or more genes and/or gene products, wherein the gene expression and/or gene product levels of normal and disease tissues are analyzed, and a variation in the expression level of the gene and/or gene product is indicative of a disease state.
[022] hi another embodiment, the invention pertains to a method of determining the phenotype of cells comprising detecting the differential expression, relative to normal cells, of at least one gene, wherein the gene is differentially expressed as compared to normal cells.
[023] In yet another embodiment, the invention pertains to a method of determining the phenotype of cells, comprising detecting the differential expression, relative to normal cells, of at least one polypeptide, wherein the protein is differentially expressed as compared to normal cells.
[024] In another embodiment, the invention pertains to a method for determining the phenotype of cells from a patient by providing a nucleic acid probe comprising a nucleotide sequence having at least about 10, at least about 15, at least about 25, or at least about 40 consecutive nucleotides, obtaining a sample of cells from a patient, optionally providing a second sample of cells substantially all of which are non-cancerous, contacting the nucleic acid probe under stringent conditions with mRNA of each of said first and second cell samples, and comparing (a) the amount of hybridization of the probe with mRNA of the first cell sample, with (b) the amount of hybridization of the probe with mRNA of the second cell sample, wherem a difference in the amount of hybridization with the mRNA of the first cell sample as compared to the amount of hybridization with the mRNA of the second cell sample is indicative of the phenotype of cells in the first cell sample.
[025] In another embodiment, the invention provides a test kit for identifying the presence of cancerous cells or tissues, comprising a probe/primer, for measuring a level of a nucleic acid in a sample of cells isolated from a patient. In certain embodiments, the kit may further include instructions for using the kit, solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a nucleic acid susceptible to hybridization, solutions for lysing cells, or solutions for the purification of nucleic acids.
[026] In one embodiment, the invention provides a test kit for identifying the presence of cancer cells or tissues, comprising an antibody specific for a protein. In certain embodiments, the kit further includes instructions for using the kit. In certain embodiments, the kit may further include solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a polypeptide susceptible to the binding of an antibody, solutions for lysing cells, or solutions for the purification of polypeptides. [027] In another embodiment, the invention provides a test kit for monitoring the efficacy of a compound or therapeutic in cancerous cells or tissues, comprising a probe/primer, for measuring a level of a nucleic acid in a sample of cells isolated from a patient. In certain embodiments, the kit may further include instructions for using the kit, solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a nucleic acid susceptible to hybridization, solutions for lysing cells, or solutions for the purification of nucleic acids.
[028] In one embodiment, the invention provides a test kit for monitoring the efficacy of a compound or therapeutic in cancer cells or tissues, comprising an antibody specific for a protein. In certain embodiments, the kit further includes instructions for using the kit. In certain embodiments, the kit may further include solutions for suspending or fixing the cells, detectable tags or labels, solutions for rendering a polypeptide susceptible to the binding of an antibody, solutions for lysing cells, or solutions for the purification of polypeptides.
[029] This invention is also related to methods of identifying biomarkers comprising the steps of selecting a set of biomarker genes from a gene expression profile representing a disease or drug treatment. DETAILED DESCRIPTION OF THE INVENTION
[030] It is to be understood that this invention is not limited to the particular methodology, protocols, cell lines, animal species or genera, constructs, and reagents described and as such may vary. It is also to be understood that the teπninology used herein is for the pmpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
[031] It must be noted that as used herein and in the appended claims, the singular forms "a," "and," and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "a gene" is a reference to one or more genes and includes equivalents thereof known to those skilled in the art, and so forth.
[032] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the prefeπed methods, devices and materials are now described.
[033] All publications and patents mentioned herein are hereby incoφorated herein by reference for the puφose of describing and disclosing, for example, the constructs and methodologies that are described in the publications which might be used in connection with the presently described invention. The publications discussed above and throughout the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention.
Definitions
[034] For convenience, the meaning of certain terms and phrases employed in the specification, examples, and appended claims are provided below.
[035] The phrase "a coπesponding normal cell of or "normal cell coπesponding to" or "normal counteφart cell of a diseased cell refers to a normal cell of the same type as that of the diseased cell.
[036] An "address" on an aπay (e.g, a microaπay) refers to a location at which a element, for example, an oligonucleotide, is attached to the solid surface of the aπay.
[037] The term "agonist," as used herein, is meant to refer to an agent that mimics or up- regulates (e.g, potentiates or supplements) the bioactivity of a protein. An agonist may be a wild- type protein or derivative thereof having at least one bioactivity of the wild-type protein. An agonist may also be a compound that up-regulates expression of a gene or which increases at least one bioactivity of a protein. An agonist can also be a compound which increases the interaction of a polypeptide with another molecule, for example, a target peptide or nucleic acid.
[038] "Amplification," as used herein, relates to the production of additional copies of a nucleic acid sequence. For example, amplification may be carried out using polymerase chain reaction (PCR) technologies which are well known in the art. (see, e.g., Dieffenbach, C. W. and G. S. Dveksler (1995) PCR Primer, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y.)
[039] "Antagonist," as used herein, is meant to refer to an agent that down-regulates (e.g, suppresses or inhibits) at least one bioactivity of a protein. An antagonist may be a compound which inhibits or decreases the interaction between a protein and another molecule, for example, a target peptide or enzyme substrate. An antagonist may also be a compound that down-regulates expression of a gene or which reduces the amount of expressed protein present. For example, an PDGFR-β inhibitor is an example of such an antagonist.
[040] The term "antibody," as used herein, is intended to include whole antibodies, for example, of any isotype (IgG, IgA, IgM, IgE, etc.), and includes fragments thereof which are also specifically reactive with a vertebrate (e.g, mammalian) protein. Antibodies may be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies. Thus, the term includes segments of proteolytically-cleaved or recombinantly-prepared portions of an antibody molecule that are capable of selectively reacting with a certain protein. Non-limiting examples of such proteolytic and/or recombinant fragments include Fab, F(ab')2, Fab', Fv, and single chain antibodies (scFv) containing a V[L] and/or V[H] domain joined by a peptide linker. The scFv's may be covalently or non-covalently linked to form antibodies having two or more binding sites. The subject invention includes polyclonal, monoclonal, or other purified preparations of antibodies and recombinant antibodies.
[041] The terms "aπay" or "matrix" refer to an aπangement of addressable locations or "addresses" on a device. The locations can be aπanged in two-dimensional aπays, three- dimensional aπays, or other matrix formats. The number of locations may range from several to at least- hundreds of thousands. Most importantly, each location represents a totally independent reaction site. A "nucleic acid aπay" refers to an aπay containing nucleic acid probes, such as oligonucleotides or larger portions of genes. The nucleic acid on the aπay may be single-stranded. Aπays wherein the probes are oligonucleotides are refeπed to as "oligonucleotide aπays" or "oligonucleotide chips." A "rnicroaπay," also refeπed to herein as a "biochip" or "biological chip," is an aπay of regions having a density of discrete regions of at least about 100/cm2, or at least about 1000/cm2. The regions in a microaπay have typical dimensions, for example, diameters, in the range of between about 10-250 μm, and are separated from other regions in the aπay by about the same distance.
[042] "Biological activity," "bioactivity," "activity," or "biological function," which are used interchangeably, herein mean an effector or antigenic function that is directly or indirectly performed by a polypeptide (whether in its native or denatured conformation), or by any subsequence thereof. Biological activities include binding to polypeptides, binding to other proteins or molecules, activity as a DNA binding protein, as a transcription regulator, ability to bind damaged DNA, etc. A bioactivity can be modulated by directly affecting the subject polypeptide. Alternatively, a bioactivity can be altered by modulating the level of the polypeptide, such as by modulating expression of the coπesponding gene.
[043] The term "biological sample," as used herein, refers to a sample obtained from an organism or from components (e.g, cells) of an organism. The sample may be of any biological tissue or fluid. The sample may be a "clinical sample" which is a sample derived from a patient. Such 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 taken for histological puφoses.
[044] The term "biomarker" or "marker" encompasses a broad range of intra- and extra-cellular events as well as whole-organism physiological changes. Biomarkers may be represent essentially any aspect of cell function, for example, but not limited to, levels or rate of production of signaling molecules, transcription factors, metabolites, gene transcripts as well as post-translational modifications of proteins. Biomarkers may include whole genome analysis of transcript levels or whole proteome analysis of protein levels and/or modifications. [045] A biomarker may also refer to a gene or gene product which is up- or down-regulated in a compound-treated, diseased cell of a subject having the disease compared to an untreated diseased cell. That is, the gene or gene product is sufficiently specific to the treated cell that it may be used, optionally with other genes or gene products, to identify, predict, or detect efficacy of a small molecule. Thus, a biomarker is a gene or gene product that is characteristic of efficacy of a compound in a diseased cell or the response of that diseased cell to treatment by the compound. [046] A nucleotide sequence is "complementary" to another nucleotide sequence if each of the bases of the two sequences match, that is, are capable of forming Watson-Crick base pairs. The term "complementary strand" is used herein interchangeably with the term "complement." The complement of a nucleic acid strand may be the complement of a coding strand or the complement of a non-coding strand.
[047] "Detection agents of genes" refers to agents that can be used to specifically detect the gene or other biological molecules relating to it, for example, RNA transcribed from the gene or polypeptides encoded by the gene. Exemplary detection agents are nucleic acid probes, which hybridize to nucleic acids coπesponding to the gene, and antibodies.
[048] "Differential gene expression pattern" between, for example, a normal cell and a disease cell refers to a pattern reflecting the differences in gene expression between a normal cell and a disease cell. A differential gene expression pattern may also be obtained between a cell at one time point and a cell at another time point, or between a cell incubated or contacted with a compound and a cell that has not been incubated with or contacted with the compound.
[049] The term "cancer" includes, but is not limited to, solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases. The term also includes lymphomas, sarcomas, and leukemias.
[050] Examples of breast cancer include, but are not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
[051] Examples of cancers of the respiratory tract include, but are not limited to, small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastema.
[052] Examples of brain cancers include, but are not limited to, brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor. [053] Tumors of the male reproductive organs include, but are not limited to, prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to, endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
[054] Tumors of the digestive tract include, but are not limited to, anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
[055] Tumors of the urinary tract include, but are not limited to, bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
[056] Eye cancers include, but are not limited to, intraocular melanoma and retinoblastoma.
[057] Examples of liver cancers include, but are not limited to, hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
[058] Skin cancers include, but are not limited to, squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
[059] Head-and-neck cancers include, but are not limited to, laryngeal / hypopharyngeal / nasopharyngeal / oropharyngeal cancer, and lip and oral cavity cancer.
[060] Lymphomas include, but are not limited to, AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
[061] Sarcomas include, but are not limited to, sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
[062] Leukemias include, but are not limited to, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
[063] "A diseased cell of cancer" refers to a cell present in subjects having cancer. That is, a cell which is a modified form of a normal cell and is not present in a subject not having cancer, or a cell which is present in significantly higher or lower numbers in subjects having cancer relative to subjects not having cancer.
[064] The term "equivalent" is understood to include nucleotide sequences encoding functionally equivalent polypeptides. Equivalent nucleotide sequences may include sequences that differ by one or more nucleotide substitutions, additions, or deletions, such as allelic variants; and may, therefore, include sequences that differ from the nucleotide sequence of the nucleic acids refeπed to in Table 1 due to the degeneracy of the genetic code. [065] The term "expression profile," which is used interchangeably herein with "gene expression profile" and "fingeφrint" of a cell refers to a set of values representing mRNA levels of one or more genes in a cell. An expression profile may comprise, for example, values representing expression levels of at least about 2 genes, at least about 5 genes, at least about 10 genes, or at least about 50, 100, 200 or more genes. Expression profiles may also comprise an mRNA level of a gene which is expressed at similar levels in multiple cells and conditions (e.g, a housekeeping gene such as GAPDH).
[066] The term "gene" refers to a nucleic acid sequence that comprises control and coding sequences necessary for the production of a polypeptide or precursor. The polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence. The gene may be derived in whole or in part from any source known to the art, including a plant, a fungus, an animal, a bacterial genome or episome, eukaryotic, nuclear or plasmid DNA, cDNA, viral DNA, or chemically synthesized DNA. A gene may contain one or more modifications in either the coding or the untranslated regions which could affect the biological activity or the chemical structure of the expression product, the rate of expression, or the manner of expression control. Such modifications include, but are not limited to, mutations, insertions, deletions, and substitutions of one or more nucleotides. The gene may constitute an uninterrupted coding sequence or it may include one or more introns, bound by the appropriate splice junctions.
[067] "Hybridization" refers to any process by which a strand of nucleic acid binds with a complementary strand through base pairing. For example, two single-stranded nucleic acids "hybridize" when they form a double-stranded duplex. The region of double-strandedness may include the full-length of one or both of the single-stranded nucleic acids, or all of one single- stranded nucleic acid and a subsequence of the other single-stranded nucleic acid, or the region of double-strandedness may include a subsequence of each nucleic acid. Hybridization also includes the formation of duplexes which contain certain mismatches, provided that the two strands are still forming a double-stranded helix. "Stringent hybridization conditions" refers to hybridization conditions resulting in essentially specific hybridization.
[068] The term "isolated," as used herein, with respect to nucleic acids, such as DNA or RNA, refers to molecules separated from other DNAs or RNAs, respectively, that are present in the natural source of the macromolecule. The term "isolated" as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Moreover, an "isolated nucleic acid" may include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state. The term "isolated" is also used herein to refer to polypeptides is meant to encompass both purified and recombinant polypeptides.
[069] As used herein, the terms "label" and "detectable label" refer to a molecule capable of detection, including, but not limited to, radioactive isotopes, fluorophores, chemiluminescent moieties, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, dyes, metal ions, ligands (e.g, biotin or haptens), and the like. The term "fluorescer" refers to a substance or a portion thereof which is capable of exhibiting fluorescence in the detectable range. Particular examples of labels which may be used in the present invention include fluorescein, rhodamine, dansyl, umbelliferone, Texas red, luminol, NADPH, alpha - beta -galactosidase, and horseradish peroxidase.
[070] The phrase "level of expression" refers to the level of mRNA, as well as pre-mRNA nascent transcript(s), transcript processing intermediates, mature n RNA(s), and degradation products, encoded by a gene in the cell. The phrase "level of expression" also refers to the level of protein or polypeptide in a cell.
[071] As used herein, the term "nucleic acid" refers to polynucleotides such as deoxyribonucleic acid (DNA) and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs and, as applicable to the embodiment being described, single-stranded (sense or antisense) and double- stranded polynucleotides. Chromosomes, cDNAs, mRNAs, rRNAs, and ESTs are representative examples of molecules that may be refeπed to as nucleic acids.
[072] The phrase "nucleic acid coπesponding to a gene" refers to a nucleic acid that can be used for detecting the gene, for example, a nucleic acid which is capable of hybridizing specifically to the gene.
[073] The phrase "nucleic acid sample derived from RNA" refers to one or more nucleic acid molecules (e.g, RNA or DNA) that may be synthesized from the RNA, and includes DNA produced from methods using PCR (e.g, RT-PCR).
[074] The term "oligonucleotide" as used herein refers to a nucleic acid molecule comprising, for example, from about 10 to about 1000 nucleotides. Oligonucleotides for use in the present invention may be from about 15 to about 150 nucleotides, or from about 150 to about 1000 in length. The oligonucleotide may be a naturally occurring oligonucleotide or a synthetic oligonucleotide. Oligonucleotides may be prepared by the phosphoramidite method (Beaucage and Carruthers, Tetrahedron Lett. 22:1859-62, 1981), or by the triester method (Matteucci, et al, J. Am. Chem. Soc. 103:3185, 1981), or by other chemical methods known in the art. [075] The term "patient" or "subject" as used herein includes mammals (e.g, humans and animals).
[076] The term "percent identical" refers to sequence identity between two amino acid sequences or between two nucleotide sequences. For example, identity between two sequences may be determined by comparing a particular position in each sequence which may be aligned for puiposes of comparison. When an equivalent position in the compared sequences is occupied by the same base or amino acid, then the molecules are identical at that position. When the equivalent site is occupied by the same or a similar amino acid residue (e.g, similar in steric and/or electronic nature), then the molecules may be refeπed to as homologous (similar) at that position. Expression as a percentage of homology, similarity, or identity refers to a function of the number of identical or similar amino acids at positions shared by the compared sequences. Various alignment algorithms and/or programs may be used including, for example, FASTA, BLAST, or ENTREZ. FASTA and BLAST are available as a part of the GCG sequence analysis package (University of Wisconsin, Madison, Wis.), and may be used with, for example, default settings. ENTREZ is available through the National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD. In one embodiment, the percent identity of two sequences may be determined by the GCG program with a gap weight of 1 (e.g, each amino acid gap is weighted as if it were a single amino acid or nucleotide mismatch between the two sequences). Other techniques for alignment are described in Methods in Enzymology (vol. 266: Computer Methods for Macromolecular Sequence Analysis (1996), ed. Doolittle, Academic Press, Inc., a division of Harcourt Brace & Co, San Diego, California, USA). An alignment program that permits gaps in the sequence may be utilized to align the sequences. For example, the Smith-Waterman is one type of algorithm that permits gaps in sequence alignments (see, e.g., Meth. Mol. Biol. 70:173-187, 1997). Also, the GAP program using the Needleman and Wunsch alignment method may be utilized to align sequences. An alternative search strategy uses MPSRCH software, which runs on a MASPAR computer. MPSRCH uses a Smith-Waterman algorithm to score sequences on a massively parallel computer. This approach improves the ability to detect distantly related matches, and is especially tolerant of small gaps and nucleotide sequence eπors. Nucleic acid-encoded amino acid sequences may be used to search both protein and DNA databases. Databases with individual sequences are described in Methods in Enzymology, ed. Doolittle, supra. Databases include, for example, Genbank, EMBL, and DNA Database of Japan (DDBJ).
[077] As used herein, a nucleic acid or other molecule attached to an aπay is refeπed to as a "probe" or "capture probe." When an aπay contains several probes coπesponding to one gene, these probes are refeπed to as a "gene-probe set." A gene-probe set may consist of, for example, about 2 to about 20 probes, from about 2 to about 10 probes, or about 5 probes.
[078] The "profile" of a cell's biological state refers to the levels of various constituents of a cell that are known to change in response to drug treatments and other perturbations of the biological state of the cell. Constituents of a cell include, for example, levels of RNA, levels of protein abundances, or protein activity levels.
[079] The term "protein," "polypeptide," and "peptide" are used interchangeably herein when referring to a gene product.
[080] An. expression profile in one cell is "similar" to an expression profile in another cell when the level of expression of the genes in the two profiles are sufficiently similar that the similarity is indicative of a common characteristic, for example, the same type of cell.
[081] "Small molecule," as used herein, refers to a composition with a molecular weight of less than about 5 kD or less than about 4 kD. Small molecules can be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids, or other organic or inorganic molecules. Many pharmaceutical companies have extensive libraries of chemical and/or biological mixtures, often fungal, bacterial, or algal extracts, which can be screened with any of the assays of the invention to identify compounds that modulate a bioactivity.
[082] The term "specific hybridization" of a probe to a target site of a template nucleic acid refers to hybridization of the probe predominantly to the target, such that the hybridization signal can be clearly inteφreted. As further described herein, such conditions resulting in specific hybridization vary depending on the length of the region of homology, the GC content of the region, and the melting temperature ("Tm") of the hybrid. Thus, hybridization conditions may vary in salt content, acidity, and temperature of the hybridization solution and the washes.
[083] A "variant" of polypeptide refers to a polypeptide having an amino acid sequence in which one or more amino acid residues is altered. The variant may have "conservative" changes, wherein a substituted amino acid has similar structural or chemical properties (e.g, replacement of leucine with isoleucine). A variant may also have "nonconservative" changes (e.g, replacement of glycine with tryptophan). Analogous minor variations may include amino acid deletions or insertions, or both. Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological or immunological activity may be identified using computer programs well known in the art, for example, LASERGENE software (DNASTAR).
[084] The term "variant," when used in the context of a polynucleotide sequence, may encompass a polynucleotide sequence related to that of a particular gene or the coding sequence thereof. This definition may also include, for example, "allelic," "splice," "species," or "polymoφhic" variants. A splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternate splicing of exons during mRNA processing. The coπesponding polypeptide may possess additional functional domains or an absence of domains. Species variants are polynucleotide sequences that vary from one species to another. The resulting polypeptides generally will have significant amino acid identity relative to each other. A polymoφhic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species. Polymoφhic variants also may encompass "single nucleotide polymoφhisms" (SNPs) in which the polynucleotide sequence varies by one base. The presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state.
Microarrays for Determining the Level of Expression of Genes
[085] Generally, determining expression profiles with microaπays involves the following steps: (a) obtaining an mRNA sample from a subject and preparing labeled nucleic acids therefrom (the "target nucleic acids" or "targets"); (b) contacting the target nucleic acids with an aπay under conditions sufficient for the target nucleic acids to bind to the coπesponding probes on the aπay, for example, by hybridization or specific binding; (c) optional removal of unbound targets from the aπay; (d) detecting the bound targets, and (e) analyzing the results, for example, using computer based analysis methods. As used herein, "nucleic acid probes" or "probes" are nucleic acids attached to the aπay, whereas "target nucleic acids" are nucleic acids that are hybridized to the aπay. Each of these steps is described in more detail below.
[086] Nucleic acid specimens may be obtained from an individual to be tested using either "invasive" or "non-invasive" sampling means. A sampling means is said to be "invasive" if it involves the collection of nucleic acids from within the skin or organs of an animal (including murine, human, ovine, equine, bovine, porcine, canine, or feline animal). Examples of invasive methods include blood collection, semen collection, needle biopsy, pleural aspiration, umbilical cord biopsy, etc. Examples of such methods are discussed by Kim, et al, (J. Virol. 66:3879-3882, 1992); Biswas, et al, (Ann. NY Acad. Sci. 590:582-583, 1990); and Biswas, et al, (J. Clin. Microbiol. 29:2228-2233, 1991).
[087] In contrast, a "non-invasive" sampling means is one in which the nucleic acid molecules are recovered from an internal or external surface of the animal. Examples of such "non-invasive" sampling means include, for example, "swabbing," collection of tears, saliva, urine, fecal material, sweat or perspiration, hair, etc. [088] In one embodiment of the present invention, one or more cells from the subject to be tested are obtained and RNA is isolated from the cells. In one embodiment, a sample of peripheral blood leukocytes (PBLs) cells is obtained from the subject. It is also possible to obtain a cell sample from a subject, and then to enrich the sample for a desired cell type. For example, cells may be isolated from other cells using a variety of techniques, such as isolation with an antibody binding to an epitope on the cell surface of the desired cell type. Where the desired cells are in a solid tissue, particular cells may be dissected, for example, by microdissection or by laser capture microdissection (LCM) (see, e.g., Bonner, et al. Science 278:1481, 1997; Emmert-Buck, et al. Science 274:998, 1996; Fend, et al. Am. J. Path. 154:61, 1999; and Murakami, et al. Kidney hit. 58:1346, 2000).
[089] RNA may be extracted from tissue or cell samples by a variety of methods, for example, guanidium thiocyanate lysis followed by CsCl centrifugation (Chirgwin, et al. Biochemistry 18:5294-5299, 1979). RNA from single cells may be obtained as described in methods for preparing cDNA libraries from single cells (see, e.g., Dulac, Cuπ. Top. Dev. Biol. 36:245, 1998; Jena, et al, J. Immunol. Methods 190:199, 1996).
[090] The RNA sample can be further enriched for a particular species, h one embodiment, for example, poly(A)+ RNA may be isolated from an RNA sample, h particular, poly-T oligonucleotides may be immobilized on a solid support to serve as affinity ligands for mRNA. Kits for this puφose are commercially available, for example, the MessageMaker kit (Life Technologies, Grand Island, NY).
[091] In one embodiment, the RNA population may be enriched for sequences of interest, such as the genes described in Table 1 (e.g, SEQ ID NOs: 1-20). Enrichment may be accomplished, for example, by primer-specific cDNA synthesis, or multiple rounds of linear amplification based on cDNA synthesis and template-directed in vitro transcription (see, e.g., Wang, et al, Proc. Natl. Acad. Sci. USA 86:9717, 1989; Dulac, et al, supra; Jena, et al, supra).
[092] The population of RNA, enriched or not in particular species or sequences, may be further amplified. Such amplification is particularly important when using RNA from a single cell or a few cells. A variety of amplification methods are suitable for use in the methods of the present invention, including, for example, PCR; ligase chain reaction (LCR) (see, e.g., Wu and Wallace, Genomics 4:560, 1989; Landegren, et al. Science 241: 1077, 1988); self-sustained sequence replication (SSR) (see, e.g., Guatelli, et al, Proc. Natl. Acad. Sci. USA 87:1874, 1990); nucleic acid based sequence amplification (NASBA) and transcription amplification (see, e.g., Kwoh, et al, Proc. Natl. Acad. Sci. USA 86: 1173, 1989). Methods for PCR technology are well known in the art (see, e.g., PCR Technology: Principles and Applications for DNA Amplification (ed. H. A. Erlich, Freeman Press, N.Y, N.Y, 1992); PCR Protocols: A Guide to Methods and Applications (eds. Innis, et al. Academic Press, San Diego, Calif, 1990); Mattila, et al. Nucleic Acids Res. 19:4967, 1991; Eckert, et al, PCR Methods and Applications 1:17, 1991; PCR (eds. McPherson, et al, IRL Press, Oxford); and U.S. Pat. No. 4,683,202). Methods of amplification are described, for example, by Ohyama, et al, (BioTechniques 29:530, 2000); Luo, et al, (Nat. Med. 5:117, 1999); Hegde, et al, (BioTechniques 29:548, 2000); Kacharmina, et al, (Meth. Enzymol. 303:3, 1999); Livesey, et al, Cuπ. Biol. 10:301, 2000); Spirin, et al, (Invest. Ophtalmol. Vis. Sci. 40:3108, 1999); and Sakai, et al, (Anal. Biochem. 287:32, 2000). RNA amplification and cDNA synthesis may also be conducted in cells in situ (see, e.g., Eberwine, et al. Proc. Natl. Acad. Sci. USA 89:3010, 1992).
[093] Generally, the target molecules will be labeled to permit detection of hybridization of the target molecules to a microaπay. That is, the probe may comprise a member of a signal producing system and thus, is detectable, either directly or through combined action with one or more additional members of a signal producing system. Examples of directly detectable labels include isotopic and fluorescent moieties incoφorated, usually by a covalent bond, into a moiety of the probe, such as a nucleotide monomeric unit (e.g, dNMP of the primer), or a photoactive or chemically active derivative of a detectable label which can be bound to a functional moiety of the probe molecule.
[094] Nucleic acids may be labeled during or after enrichment and/or amplification of RNAs. For example, reverse transcription may be carried out in the presence of a dNTP conjugated to a detectable label, for example, a ftuorescently labeled dNTP. In another embodiment, the cDNA or RNA probe may be synthesized in the absence of detectable label and may be labeled subsequently, for example, by incoφorating biotinylated dNTPs or rNTP, or some similar means (e.g, photo-cross-linking a psoralen derivative of biotin to RNAs), followed by addition of labeled streptavidin (e.g, phycoerythrin-conjugated streptavidin) or the equivalent.
[095] Fluorescent moieties or labels of interest include coumarin and its derivatives (e.g, 7- amino-4-methylcoumarin, aminocoumarin); bodipy dyes such as Bodipy FL and cascade blue; fluorescein and its derivatives (e.g, fluorescein isothiocyanate, Oregon green); rhodamine dyes (e.g, Texas red, tetramethylrhodamine); eosins and erythrosins; cyanine dyes (e.g, Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7); FluorX, macrocyclic chelates of lanthanide ions (e.g, quantum dye™); fluorescent energy transfer dyes such as thiazole orange-ethidium heterodimer, TOTAB, dansyl, etc. Individual fluorescent compounds which have functionalities for linking to an element desirably detected in an apparatus or assay of the invention, or which may be modified to incoφorate such functionalities may also be utilized (see, e.g., Kricka, 1992, Nonisotopic DNA Probe Techniques, Academic Press San Diego, Calif). [096] Chemiluminescent labels include luciferin and 2,3-dihydrophthalazinediones, for example, luminol. [097] Labels may also be members of a signal producing system that act in concert with one or more additional members of the same system to provide a detectable signal. Illustrative of such labels are members of a specific binding pair, such as ligands, for example, biotin, fluorescein, digoxigenin, antigen, polyvalent cations, chelator groups and the like. Members may specifically bind to additional members of the signal producing system, and the additional members may provide a detectable signal either directly or indirectly, for example, an antibody conjugated to a fluorescent moiety or an enzymatic moiety capable of converting a substrate to a chromogenic product (e.g, alkaline phosphatase conjugate antibody and the like).
[098] Additional labels of interest include those that provide a signal only when the probe with which it is associated is specifically bound to a target molecule. Such labels include "molecular beacons" as described in Tyagi and Kramer (Nature Biotech. 14:303, 1996) and EP 0 070 685 Bl. Other labels of interest include those described in U.S. Patent No. 5,563,037; WO 97/17471; and WO 97/17076.
[099] In other embodiments, the target nucleic acid may not be labeled. In this case, hybridization may be determined, for example, by plasmon resonance (see, e.g., Thiel, et al. Anal. Chem. 69:4948, 1997).
[100] In one embodiment, a plurality (e.g, 2, 3, 4, 5, or more) of sets of target nucleic acids are labeled and used in one hybridization reaction ("multiplex"*analysis). For example, one set of nucleic acids may coπespond to RNA from one cell and another set of nucleic acids may coπespond to RNA from another cell. The plurality of sets of nucleic acids may be labeled with different labels, for example, different fluorescent labels (e.g, fluorescein and rhodamine) which have distinct emission spectra so that they can be distinguished. The sets may then be mixed and hybridized simultaneously to one microaπay (see, e.g., Shena, et al. Science 270:467-470, 1995).
[101] Examples of distinguishable labels for use when hybridizing a plurality of target nucleic acids to one aπay are well known in the art and include: two or more different emission wavelength fluorescent dyes such as Cy3 and Cy5; combination of fluorescent proteins and dyes such as phicoerythrin and Cy5; two or more isotopes with different energy of emission such as 32P and 33P; gold or silver particles with different scattering spectra; labels which generate signals under different treatment conditions such as temperature, pH, treatment with additional chemical agents, etc.; or generate signals at different time points after treatment. Using one or more enzymes for signal generation allows for the use of an even greater variety of distinguishable labels, based on different substrate specificity of enzymes (e.g, alkaline phosphatase/peroxidase). [102] The quality of labeled nucleic acids may be evaluated prior to hybridization to an aπay. In one embodiment, the GeneChip® Test3 Aπay from Affymetrix (Santa Clara, CA) may be used for that puφose. This aπay contains probes representing a subset of characterized genes from several organisms including mammals. Thus, the quality of a labeled nucleic acid sample can be determined by hybridization of a fraction of the sample to an aπay.
[103] Microaπays for use according to the invention include one or more probes of genes characteristic of small molecule efficacy. In one embodiment, the microaπay comprises probes coπesponding to one or more of genes selected from the group consisting of genes which are up- regulated in cancer and genes which are down-regulated in cancer. The microaπay may comprise, for example, probes coπesponding to at least 2, at least 5, at least 10, at least 100 or more characteristic of small molecule efficacy. The microaπay may comprise probes coπesponding to each gene or gene product listed in Table 1.
[104] There may be one or more than one probe coπesponding to each gene on a microaπay. For example, a microaπay may contain from 2 to 20 probes coπesponding to one gene or about 5 to 10. The probes may coπespond to the full-length RNA sequence or complement thereof of genes characteristic of small molecule efficacy, or the probe may coπespond to a portion thereof, which portion is of sufficient length to permit specific hybridization. Such probes may comprise from about 50 nucleotides to about 100, 200, 500, or 1000 nucleotides or more than 1000 nucleotides. As further described herein, microaπays may contain oligonucleotide probes, consisting of about 10 to 50 nucleotides, about 15 to 30 nucleotides, or about 20-25 nucleotides. The probes are may be single-stranded and will have sufficient complementarity to its target to provide for the desired level of sequence specific hybridization.
[105] Typically, the aπays used in the present invention will have a site density of greater than 100 different probes per cm2. The aπays may have a site density of, for example, greater than 500/cm2, greater than about 1000/cm2, or greater than about 10,000/cm2. The aπays may have, for example, more than 100 different probes on a single substrate, greater than about 1000 different probes, greater than about 10,000 different probes, or greater than 100,000 different probes on a single substrate.
[106] A number of different microaπay configurations and methods for their production are known to those of skill in the art and are disclosed in U.S. Patent Nos: 5,242,974; 5,384,261; 5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,436,327; 5,445,934; 5,556,752; 5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,436,327; 5,472,672; 5,527,681; 5,529,756; 5,545,531; 5,554,501; 5,561,071; 5,571,639; 5,593,839; 5,624,711; 5,700,637; 5,744,305; 5,770,456; 5,770,722; 5,837,832; 5,856,101; 5,874,219; 5,885,837; 5,919,523; 6,022,963; 6,077,674; and 6,156,501; Shena, et al, Tibtech 16:301, 1998; Duggan, et al, Nat. Genet. 21:10, 1999; Bowtell, et al, Nat. Genet. 21:25, 1999; Lipshutz, et al, 21 Nature Genet. 20-24, 1999; Blanchard, et al, 11 Biosensors and Bioelectronics, 687-90, 1996; Maskos, et al, 21 Nucleic Acids Res. 4663-69, 1993; Hughes, et al, Nat. Biotechol. 19:342, 2001; the disclosures of which are herein incoφorated by reference. Patents describing methods of using aπays in various applications include: U.S. Pat. Nos. 5,143,854; 5,288,644; 5,324,633; 5,432,049; 5,470,710; 5,492,806; 5,503,980; 5,510,270; 5,525,464; 5,547,839; 5,580,732; 5,661,028; 5,848,659; and 5,874,219; the disclosures of which are herein incoφorated by reference.
[107] Aπays may also include control and reference nucleic acids. Control nucleic acids include, for example, prokaryotic genes such as bioB, bioC and bioD, ere from PI bacteriophage or polyA controls, such as dap, lys, phe, thr, and tφ. Reference nucleic acids allow the normalization of results from one experiment to another and the comparison of multiple experiments on a quantitative level. Exemplary reference nucleic acids include housekeeping genes of known expression levels, for example, GAPDH, hexokinase, and actin.
[108] In one embodiment, an aπay of oligonucleotides may be synthesized on a solid support. Exemplary solid supports include glass, plastics, polymers, metals, metalloids, ceramics, organics, etc. Using chip masking technologies and photoprotective chemistry, it is possible to generate ordered aπays of nucleic acid probes. These aπays, which are known, for example, as "DNA chips" or very large scale immobilized polymer aπays ("VLSIPS™" aπays), may include millions of defined probe regions on a substrate having an area of about 1 cm2 to several cm2, thereby incoφorating from a few to millions of probes (see, e.g., U.S. Patent No. 5,631,734).
[109] A nucleic acid probe may be at least, for example, about 10, 15, 20, 25, 30, 50, 100 or more nucleotides, and may comprise the full-length gene. For example, probes may be those that hybridize specifically to the genes listed in Table 1.
[110] Nucleic acid probes may be obtained, for example, by PCR amplificartion of gene segments from genomic, cDNA (e.g, RT-PCR), or cloned sequences. cDNA probes may be prepared according to methods known in the art and further described herein, for example, by reverse-transcription PCR (RT-PCR) of RNA using sequence specific primers. Sequences of genes or cDNA from which probes are generated may be obtained, for example, from GenBank, other public databases, or publications.
[Ill] Oligonucleotide probes may also be synthesized by standard methods known in the art, for example, by automated DNA synthesizer or any other chemical method. As an example, phosphorothioate oligonucleotides may be synthesized by the method of Stein, et al, (Nucl. Acids Res. 16:3209, 1988), and methylphosphonate oligonucleotides may be prepared by controlled pore glass polymer supports (see, e.g., Sarin, et al, Proc. Natl. Acad. Sci. U.S.A. 85:7448-7451, 1988). In another embodiment, the oligonucleotide may be a 2'-)-methylribonucleotide (Inoue, et al, Nucl. Acids Res. 15:6131-6148, 1987), or a chimeric RNA-DNA analog (Inoue, et al, FEBS Lett. 215:327-330, 1987).
[112] Nucleic acid probes may be natural nucleic acids or chemically modified nucleic acids (e.g, composed of nucleotide analogs); however, the probes should possess activated hydroxyl groups compatible with the linking chemistry. The protective groups may be photolabile, or the protective groups may be labile under certain chemical conditions (e.g, acid). The surface of the solid support may contain a composition that generates acids upon exposure to light. Thus, exposure of a region of the substrate to light generates acids in that region that remove the protective groups in the exposed region. Also, the synthesis method may use 3'- protected 5'-0- phosphoramidite-activated deoxynucleoside. In this case, the oligonucleotide is synthesized in the 5' to 3' direction, which results in a free 5' end.
[113] hi one embodiment of the present invention, oligonucleotides of an aπay may be synthesized using a 96-well automated multiplex oligonucleotide synthesizer (A.M.O.S.) that is capable of producing thousands of oligonucleotides (see, e.g., Lashkari, et al, Proc. Natl. Acad. Sci. USA 93: 7912, 1995).
[114] To compare expression levels, labeled nucleic acids may be contacted with the aπay under conditions sufficient for binding between the target nucleic acid and the probe on the aπay. hi one embodiment, the hybridization conditions may be selected to provide for the desired level of hybridization specificity; that is, conditions sufficient for hybridization to occur between the labeled nucleic acids and probes on the microaπay.
[115] Hybridization may be carried out in conditions permitting essentially specific hybridization. The length and GC content of the nucleic acid will determine the thermal melting point and thus, the hybridization conditions necessary for obtaining specific hybridization of the probe to the target nucleic acid. These factors are well known to a person of skill in the art, and may also be tested in assays. An extensive guide to nucleic acid hybridization may be found in
Tijssen, et al. (Laboratory Techniques in Biochemistry and Molecular Biology, Vol. 24:
Hybridization With Nucleic Acid Probes, P. Tijssen, ed. Elsevier, N.Y, (1993)). Generally, stringent conditions may be selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Highly stringent conditions may be selected to be equal to the Tm point for a particular probe. Sometimes the term "dissociation temperature" (Td) is used to define the temperature at which at least half of the probe dissociates from a perfectly matched target nucleic acid. In any case, a variety of techniques for estimating the Tm or Td are available, and generally are described in Tijssen, supra. Typically, G-C base pairs in a duplex are estimated to contribute about 3°C to the Tm, while A-T base pairs are estimated to contribute about 2°C, up to a theoretical maximum of about 80-100°C. However, more sophisticated models of Tm and Td are available in which G-C stacking interactions, solvent effects, the desired assay temperature, and the like are taken into account.
[116] . In one embodiment, non-specific binding or background signal may be reduced by the use of a detergent (e.g, C-TAB) or a blocking reagent (e.g, sperm DNA, cot-1 DNA, etc.) during the hybridization, hi one embodiment, the hybridization may be performed in the presence of about 0.5 mg/ml DNA (e.g, herring sperm DNA). The use of blocking agents in hybridization is well known to those of skill in the art (see, e.g., Tijssen, supra).
[117] If the target sequences are detected using the same label, different aπays may be employed for each physiological source or the same aπay may be screened multiple times. The above methods may be varied to provide for multiplex analysis by employing different and distinguishable labels for the different target populations (e.g, different physiological sources). According to this multiplex method, the same aπay may be used at the same time for each of the different target populations.
[118] The methods described above result in the production of hybridization patterns of labeled target nucleic acids on the aπay surface. The resultant hybridization patterns of labeled nucleic acids may be visualized or detected in a variety of ways, with the particular mamier of detection selected based on the particular label of the target nucleic acid. Representative detection means include scintillation counting, autoradiography, fluorescence measurement, colorimetric measurement, light emission measurement, light scattering, and the like.
[119] One such method of detection utilizes an aπay scanner that is commercially available (Affymetrix, Santa Clara, CA), for example, the 417™ Aπayer, the 418™ Aπay Scanner, or the Agilent GeneAπay™ Scanner. This scanner is controlled from a system computer with an interface and easy-to-use software tools. The output may be directly imported into or directly read by a variety of software applications. Scanning devices are described in, for example, U.S. Patent Nos. 5,143,854 and 5,424,186.
[120] For fluorescent labeled probes, the fluorescence emissions at each site of a transcript aπay may be detected by scanning confocal laser microscopy. Alternatively, a laser may be used that allows simultaneous specimen illumination at wavelengths specific to the two fluorophores and emissions from the two fluorophores may be analyzed simultaneously (see, e.g., Shalon, et al. Genome Res. 6:639-645, 1996). For example, the aπays may be scanned with a laser fluorescent scanner with a computer controlled X-Y stage and a microscope objective. Fluorescence laser scanning devices are described in Shalon, et al, supra.
[121] Following the data gathering operation, the data will typically be reported to a data analysis operation. To facilitate the sample analysis operation, the data obtained by the reader from the device may be analyzed using a digital computer. Typically, the computer will be appropriately programmed for receipt and storage of the data from the device, as well as for analysis and reporting of the data gathered, for example, subtraction of the background, deconvolution of multi-color images, flagging or removing artifacts, verifying that controls have performed properly, normalizing the signals, inteφreting fluorescence data to determine the amount of hybridized target, normalization of background and single base mismatch hybridizations, and the like.
[122] In one embodiment, a system comprises a search function that allows one to search for specific patterns, for example, patterns relating to differential gene expression, for example, between the expression profile of a cancer cell and the expression profile of a counteφart normal cell in a subject. For example, a system allows one to search for patterns of gene expression between more than two samples.
[123] Various algorithms are available for analyzing gene expression profile data, for example, the type of comparisons to perform. In certain embodiments, it is desirable to group genes that are co-regulated. This allows for the comparison of large numbers of profiles. One embodiment for identifying such groups of genes involves clustering algorithms (for reviews of clustering algorithms, see, e.g., Fukunaga, 1990, Statistical Pattern Recognition, 2nd Ed, Academic Press, San Diego; Everitt, 1974, Cluster Analysis, London: Heinemann Educ. Books; Hartigan, 1975, Clustering Algorithms, New York: Wiley; Sneath and Sokal, 1973, Numerical Taxonomy, Freeman; Anderberg, 1973, Cluster Analysis for Applications, Academic Press: New York).
[124] Clustering may be based on other characteristics of the genes, for example, their level of expression (see, e.g., U.S. Patent No. 6,203,987), or permit clustering of time curves (see, e.g. U.S. Patent No. 6,263,287). Examples of clustering algorithms include K-means clustering and hierarchical clustering. Clustering may also be achieved by visual inspection of gene expression data using a graphical representation of the data (e.g. a "heat map"). An example of software which contains clustering algorithms and a means to graphically represent gene expression data is Spotfire DecisionSite (Spotfire, Inc, Somerville, Massachusetts and Goteborg, Sweden).
[125] Comparison of the expression levels of one or more genes characteristic of small molecule efficacy with reference expression levels, for example, expression levels in diseased cells of cancer or in normal counteφart cells, may be conducted using computer systems. In one embodiment, expression levels may be obtained from two cells and these two sets of expression levels may be introduced into a computer system for comparison. For example, one set of expression levels is entered into a computer system for comparison with values that are already present in the computer system, or in computer-readable form that is then entered into the computer system.
[126] hi one embodiment, the computer system may also contain a database comprising values representing levels of expression of one or more genes characteristic of small molecule efficacy. The database may contain one or more expression profiles of genes characteristic of small molecule efficacy in different cells.
[127] In another embodiment, the invention provides a computer-readable form of the gene expression profile data, or of values coπesponding to the level of expression of at least one gene characteristic of cancer in a diseased cell. The values may be mRNA expression levels obtained from experiments, for example, microaπay analysis. The values may also be mRNA levels normalized relative to a reference gene whose expression is constant in numerous cells under numerous conditions (e.g, GAPDH). In other embodiments, the values in the computer may be ratios of, or differences between, normalized or non-normalized mRNA levels in different samples.
[128] In one embodiment, the expression profiles expression profiles from cancer cells of one or more subjects, which cells are treated in vivo or in vitro with a drug, for example, an PDGFR-β inhibitor. Expression data of a cell of a subject treated in vitro or in vivo with the drug is entered into a computer and the computer is instructed to compare the data entered to the data in the computer, and to provide results indicating whether the expression data input into the computer are more similar to those of a cell of a subject that is responsive to the drug or more similar to those of a cell of a subject that is not responsive to the drug. Thus, the results indicate whether the subject is likely to respond to the treatment with the drug or unlikely to respond to it.
[129] The invention also provides a machine-readable or computer-readable medium including program instructions for performing the following steps: (i) comparing a plurality of values coπesponding to expression levels of one or more genes characteristic of small molecule efficacy in a query cell with a database including records comprising reference expression or expression profile data of one or more reference cells and an annotation of the type of cell; and (ii) indicating to which cell the query cell is most similar based on similarities of expression profiles. The reference cells may be cells from subjects at different stages of cancer. The reference cells may also be cells from subjects responding or not responding to a particular drug treatment and optionally incubated in vitro or in vivo with the drug. [130] The reference cells may also be cells from subjects responding or not responding to several different treatments, and the computer system indicates a prefeπed treatment for the subject. Accordingly, the invention provides a method for selecting a therapy for a patient having cancer, the method comprising: (i) providing the level of expression of one or more genes characteristic of small molecule efficacy in a diseased cell of the patient; (ii) providing a plurality of reference profiles, each associated with a therapy, wherein the subject expression profile and each reference profile has a plurality of values, each value representing the level of expression of a gene characteristic of cancer; and (iii) selecting the reference profile most similar to the subject expression profile, to thereby select a therapy for said patient, hi one embodiment, step (iii) may be performed by a computer. The most similar reference profile may be selected by weighing a comparison value of the plurality using a weight value associated with the coπesponding expression data.
[131] The relative abundance of an mRNA in two biological samples may be scored as a perturbation and its magnitude determined (i.e, the abundance is different in the two sources of mRNA tested), or as not perturbed (i.e, the relative abundance is the same). In various embodiments, a difference between the two sources of RNA is scored as a perturbation. Perturbations rαay be used by a computer for calculating and expression comparisons.
Drug Design Using Microarrays
[132] The invention also provides methods for designing and optimizing drugs for cancer, for example, those which have been identified as described herein, hi one embodiment, compounds may be screened by comparing the expression level of one or more genes characteristic of small molecule efficacy following incubation of a diseased cell of cancer or similar cell with the test compound. In another embodiment, the expression level of the genes may be determined using microaπays, and comparing the gene expression profile of a cell in response to the test compound with the gene expression profile of a normal cell coπesponding to a diseased cell of cancer (a "reference profile"). In a further embodiment, the expression profile may also be compared to that of a diseased cell of cancer. The comparisons may be done by introducing the gene expression profile data of the cell treated with drug into a computer system comprising reference gene expression profiles, which are stored in a computer readable form, using appropriate algorithms. Test compounds may be screened for those that alter the level of expression of genes characteristic of small molecule efficacy. Such compounds, that is, compounds which are capable of normalizing the expression of essentially all genes characteristic of small molecule efficacy, are candidate therapeutics.
[133] The efficacy of the compounds may then be tested in additional in vitro and in vivo assays, and in animal models (e.g, xenograft model). The test compound may be administered to the test animal, and one or more symptoms of the disease may be monitored for improvement of the condition of the animal. Expression of one or more genes characteristic of small molecule efficacy may also be measured before and after administration of the test compound to the animal. A normalization of the expression of one or more of these genes is indicative of the efficiency of the compound for treating cancer in the animal.
[134] In the clinical setting, obtaining human-derived samples of tissue exhibiting cancer may be difficult, if not prohibitive. Therefore, identification of gene expression changes indicative of efficacy of a therapeutic compound may be determined in a more easily accessible, suπogate cell population, for example, peripheral blood leukocytes (PBLs). This method may be performed either in a human or animal model system. In one embodiment, a test compound may be administered to the test animal (either normal or cancer-containing) at the same doses that have been observed to be efficacious in treating cancer in that animal model. Blood may be drawn from the animal at various time points (e.g, 1, 4, 7, and 24 hours following the first, mid-point, and last day of a regimen of multiple day dosing). Animals dosed with vehicle may be used as controls. RNA may be isolated from PBLs, and can be used to generate probes for hybridization to microaπays. The hybridization results may then be analyzed using computer programs and databases, as described above. The resulting expression profile may be compared directly to the analogous profile from the treated cancer tissue for similarities or simply coπelated with efficacy (e.g, in terms of doses and time points) in the animal model.
[135] In another embodiment, human blood may be treated ex vivo with a therapeutic compound at a dose consistent with the therapeutic dose in the animal model, or at a dose that is consistent with known plasma levels of the therapeutic dose in the animal model. The blood may be treated (e.g, rocking at 37°C) with the therapeutic compound immediately, or after some period of incubation time (e.g, 24 hours) to allow for gene expression to re-equilibrate after the blood draw. The blood may also be treated with the therapeutic compound for various timepoints (e.g, 4 and 24 hours), and then PBL RNA isolated and used to create a probe for hybridization to a microaπay. A compound solubilization agent (e.g, DMSO) may be used as a control. The resulting expression profile may be compared directly to the analogous profile from the treated cancer tissue for similarities or simply coπelated with efficacy (e.g, in terms of doses and time points) in the animal model.
[136] The toxicity of the candidate therapeutic compound may be evaluated, for example, by determining whether the compound induces the expression of genes known to be associated with a toxic response. Expression of such toxicity related genes may be determined in different cell types, for example, those that are known to express the genes, hi fact, alterations in gene expression may serve as a more sensitive marker of human toxicity than routine preclinical safety studies. Microaπays may be used for detecting changes in the expression of genes known to be associated with a toxic response. It may be possible to perform proof of concept studies demonstrating that changes in gene expression levels may predict toxic events that were not identified by routine preclinical safety testing (see, e.g., Huang, et al, Toxicol. Sci. 63: 196-207, 2001; Waring, et al, Toxicol. Appl. Pharamacol. 175:28-42, 2001).
[137] Drug screening may be performed by adding a test compound to a sample of cells, and monitoring the effect. A parallel sample which does not receive the test compound may also be monitored as a control. The treated and untreated cells are then compared by any suitable phenotypic criteria, including but not limited to microscopic analysis, viability testing, ability to replicate, histological examination, the level of a particular RNA or polypeptide associated with the cells, the level of enzymatic activity expressed by the cells or cell lysates, and the ability of the cells to interact with other cells or compounds. Differences between treated and untreated cells indicates effects attributable to the test compound.
[138] Desirable effects of a test compound include an effect on any phenotype that was confeπed by the cancer-associated marker nucleic acid sequence. Examples include a test compound that limits the overabundance of mRNA, limits production of the encoded protein, or limits the functional effect of the protein. The effect of the test compound would be apparent when comparing results between treated and untreated cells.
Diagnostic and Prognostic Assays
[139] The present invention provides nucleic acid sequences which are differentially regulated in cancer, and a method for identifying such sequences. The present invention provides a method for identifying a nucleotide sequence which is differentially regulated in a subject with cancer, comprising: hybridizing a nucleic acid sample coπesponding to RNA obtained from the subject to a nucleic acid sample comprising one or more nucleic acid molecules of known identity; and measuring the hybridization of the nucleic acid sample to the one or more nucleic acid molecules of known identity, wherein a difference in the hybridization of the nucleic acid sample to the one or more nucleic acid molecules of known identity relative to a nucleic acid sample obtained from a subject without cancer is indicative of the differential expression of the nucleotide sequence in a subject with cancer.
[140] Generally, the present invention provides a method for identifying nucleic acid sequences which are differentially regulated in a subject with cancer comprising isolating messenger RNA from a subject, generating cRNA from the mRNA sample, hybridizing the cRNA to a microaπay comprising a plurality of nucleic acid molecules stably associated with discrete locations on the aπay, and identifying patterns of hybridization of the cRNA to the aπay. According to the present invention, a nucleic acid molecule which hybridizes to a given location on the aπay is said to be differentially regulated if the hybridization signal is, for example, higher or lower than the hybridization signal at the same location on an identical aπay hybridized with a nucleic acid sample obtained from a subject that does not have cancer.
[141] Expression patterns may be used to derive a panel of biomarkers that can be used to predict the efficacy of drug treatment in the patients. The biomarkers may consist of gene expression levels from microaπay experiments on RNA isolated from biological samples, RNA isolated from frozen samples of tumor biopsies, or mass spectrometry-derived protein masses in the serum.
[142] Although the precise mechanism for data analysis will depend upon the exact nature of the data, a typical procedure for developing a panel of biomarkers is as follows. The data (gene expression levels or mass spectra) are collected for each patient prior to treatment. As the study progresses, the patients are classified according to their response to the drug treatment; either as efficacious or non-efficacious. Multiple levels of efficacy can be accommodated in a data model, but a binary comparison is considered optimal, particularly if the patient population is less than several hundred. Assuming adequate numbers of patients in each class, the protein and/or gene expression data may be analyzed by a number of techniques known in the art. Many of the techniques are derived from traditional statistics as well from the field of machine learning. These techniques serve two puφoses: 1. Reduce the dimensionality of data - In the case of mass spectra or gene expression microaπays, data is reduced from many thousands of individual data points to, for example, about three to ten. The reduction is based upon the predictive power of the data points when taken as a set. 2. Training - These three to ten data points are then used to train multiple machine learning algorithms which then "learn" to recognize, in this case, patterns of protein masses or gene expression which distinguish efficacious drug treatment from non-efficacious. All patient samples can be used to train the algorithms.
[143] The resulting trained algorithms are then tested in order to measure their predictive power. Typically, when less than many hundreds of training examples are available, some form of cross- validation is performed. To illustrate, consider a ten-fold cross validation. In this case, patient samples are randomly assigned to one often bins. In the first round of validation the samples in nine of the bins are used for training and the remaining samples in the tenth bin are used to test the algorithm. This is repeated an additional nine times, each time leaving out the samples in a different bin for testing. The results (coπect predictions and eπors) from all ten rounds are combined and the predictive power is then assessed. Different algorithms, as well as different panels, may be compared in this way for this study. The "best" algorithm/panel combination will then be selected. This "smart" algorithm may then be used in future studies to select the patients that are most likely to respond to treatment.
[144] Many algorithms benefit from additional information taken for the patients. For example, gender or age could be used to improve predictive power. Also, data transformations such as normalization and smoothing may be used to reduce noise. Because of this, a large number of algorithms may be trained using many different parameters in order to optimize the outcome. If predictive patterns exist in the data, it is likely that an optimal, or near-optimal, "smart" algorithm can be developed. If more patient samples become available, the algorithm can be retrained to take advantage of the new data.
[145] As an example using mass spectrometry, plasma may be applied to a hydrophobic SELDI- target, washed extensively in water, and analyzed by the SELDI-Tof mass spectrometer. This may be repeated on 100 or more patient samples. The protein profiles resulting from the intensities of some 16,000 m/z values in each sample would be statistically analyzed in order to identify sets of specific m/z values that are predictive of drug efficacy. Identical experiments using other SELDI- targets, such as ion-exchange or IMAC surfaces, could also be conducted. These will capture different subsets of the proteins present in plasma. Furthermore, the plasma may be denatured and prefractionated prior to application onto the SELDI target.
[146] The present invention provides methods for determining whether a subject is at risk for developing a disease or condition characterized by unwanted cell proliferation by detecting biomarkers, that is, nucleic acids and/or polypeptide markers for cancer.
[147] In clinical applications, human tissue samples may be screened for the presence and/or absence of biomarkers identified herein. Such samples could consist of needle biopsy cores, surgical resection samples, lymph node tissue, or serum. For example, these methods include obtaining a biopsy, which is optionally fractionated by cryostat sectioning to enrich tumor cells to about 80% of the total cell population. In certain embodiments, nucleic acids extracted from these samples may be amplified using techniques well known in the art. The levels of selected markers detected would be compared with statistically valid groups of metastatic, non-metastatic malignant, benign, or normal tissue samples.
[148] hi one embodiment, the diagnostic method comprises determining whether a subject has an abnormal mRNA and/or protein level of the biomarkers, such as by Northern blot analysis, reverse transcription-polymerase chain reaction (RT-PCR), in situ hybridization, immunoprecipitation, Western blot hybridization, or immunohistochemistry. According to the method, cells may be obtained from a subject and the levels of the biomarkers, protein, or mRNA level, are determined and compared to the level of these markers in a healthy subject. An abnormal level of the biomarker polypeptide or mRNA levels is likely to be indicative of cancer.
[149] In one embodiment, the method comprises using a nucleic acid probe to determine the presence of cancerous cells in a tissue from a patient. Specifically, the method comprises:
1. providing a nucleic acid probe comprising a nucleotide sequence, for example, at least 10, 15, 25 or 40 nucleotides, and up to all or nearly all of the coding sequence which is complementary to a portion of the coding sequence of a nucleic acid sequence and is differentially expressed in tumors cells;
2. obtaining a tissue sample from a patient potentially comprising cancerous cells;
3. providing a second tissue sample containing cells substantially all of which are non- cancerous;
4. contacting the nucleic acid probe under stringent conditions with RNA of each of said first and second tissue samples (e.g, in a Northern blot or in situ hybridization assay); and
5. comparing (a) the amount of hybridization of the probe with RNA of the first tissue sample, with (b) the amount of hybridization of the probe with RNA of the second tissue sample; wherein a statistically significant difference in the amount of hybridization with the RNA of the first tissue sample as compared to the amount of hybridization with the RNA of the second tissue sample is indicative of the presence of cancerous cells in the first tissue sample.
[150] In one aspect, the method comprises in situ hybridization with a probe derived from a given marker nucleic acid sequence. The method comprises contacting the labeled hybridization probe with a sample of a given type of tissue potentially containing cancerous or pre-cancerous cells as well as normal cells, and determining whether the probe labels some cells of the given tissue type to a degree significantly different than the degree to which it labels other cells of the same tissue type.
[151] Also within the invention is a method of determining the phenotype of a test cell from a given human tissue, for example, whether the cell is (a) normal, or (b) cancerous or precancerous, by contacting the mRNA of a test cell with a nucleic acid probe, for example, at least about 10, 15, 25, or 40 nucleotides, and up to all or nearly all of a sequence which is complementary to a portion of the coding sequence of a nucleic acid sequence, and which is differentially expressed in tumor cells as compared to normal cells of the given tissue type; and determining the approximate amount of hybridization of the probe to the mRNA, an amount of hybridization either more or less than that seen with the mRNA of a normal cell of that tissue type being indicative that the test cell is cancerous or pre-cancerous.
[152] Alternatively, the above diagnostic assays may be carried out using antibodies to detect the protein product encoded by the marker nucleic acid sequence. Accordingly, in one embodiment, the assay would include contacting the proteins of the test cell with an antibody specific for the gene product of a nucleic acid, the marker nucleic acid being one which is expressed at a given control level in normal cells of the same tissue type as the test cell, and determining the approximate amount of immunocomplex formation by the antibody and the proteins of the test cell, wherein a statistically significant difference in the amount of the immunocomplex formed with the proteins of a test cell as compared to a normal cell of the same tissue type is an indication that the test cell is cancerous or pre-cancerous.
[153] The method for producing polyclonal and/or monoclonal antibodies which specifically bind to polypeptides useful in the present invention is known to those of skill in the art and may be found in, for example, Dymecki, et al, (J. Biol. Chem. 267:4815, 1992); Boersma & Van Leeuwen, (J. Neurosci. Methods 51:317, 1994); Green, et al, (Cell 28:477, 1982); and Arnheiter, et al, (Nature 294:278, 1981).
[154] Another such method includes the steps of: providing an antibody specific for the gene product of a marker nucleic acid sequence, the gene product being present in cancerous tissue of a given tissue type at a level more or less than the level of the gene product in non-cancerous tissue of the same tissue type; obtaining from a patient a first sample of tissue of the given tissue type, which sample potentially includes cancerous cells; providing a second sample of tissue of the same tissue type (which may be from the same patient or from a normal control, e.g. another individual or cultured cells), this second sample containing normal cells and essentially no cancerous cells; contacting the antibody with protein (which may be partially purified, in lysed but unfractionated cells, or in situ) of the first and second samples under conditions permitting immunocomplex formation between the antibody and the marker nucleic acid sequence product present in the samples; and comparing (a) the amount of immunocomplex formation in the first sample, with (b) the amount of immunocomplex formation in the second sample, wherein a statistically significant difference in the amount of immunocomplex formation in the first sample less as compared to the amount of immunocomplex formation in the second sample is indicative of the presence of cancerous cells in the first sample of tissue.
[155] The subject invention further provides a method of determining whether a cell sample obtained from a subject possesses an abnormal amount of marker polypeptide which comprises (a) obtaining a cell sample from the subject, (b) quantitatively determining the amount of the marker polypeptide in the sample so obtained, and (c) comparing the amount of the marker polypeptide so determined with a known standard, so as to thereby determine whether the cell sample obtained from the subject possesses an abnormal amount of the marker polypeptide. Such marker polypeptides may be detected by immunohistochemical assays, dot-blot assays, ELISA, and the like. [156] Immunoassays are commonly used to quantitate the levels of proteins in cell samples, and many other immunoassay techniques are known in the art. The invention is not limited to a particular assay procedure, and therefore, is intended to include both homogeneous and heterogeneous procedures. Exemplary immunoassays which may be conducted according to the invention include fluorescence polarization immunoassay (FPIA), fluorescence immunoassay (FIA), enzyme immunoassay (EIA), nephelometric inhibition immunoassay (NIA), enzyme-linked immunosorbent assay (ELISA), and radioimmunoassay (RIA). An indicator moiety, or label group, may be attached to the subject antibodies and is selected so as to meet the needs of various uses of the method which are often dictated by the availability of assay equipment and compatible immunoassay procedures. General techniques to be used in perforating the various immunoassays noted above are known to those of ordinary skill in the art.
[157] i another embodiment, the level of the encoded product, or alternatively the level of the polypeptide, in a biological fluid (e.g, blood or urine) of a patient may be determined as a way of monitoring the level of expression of the marker nucleic acid sequence in cells of that patient. Such a method would include the steps of obtaining a sample of a biological fluid from the patient, contacting the sample (or proteins from the sample) with an antibody specific for an encoded marker polypeptide, and determining the amount of immune complex formation by the antibody, with the amount of immune complex formation being indicative of the level of the marker encoded product in the sample. This determination is particularly instructive when compared to the amount of immune complex formation by the same antibody in a control sample taken from a normal individual or in one or more samples previously or subsequently obtained from the same person.
[158] In another embodiment, the method may be used to determine the amount of marker polypeptide present in a cell, which in turn may be coπelated with progression of a hypeφroliferative disorder. The level of the marker polypeptide may be used predictively to evaluate whether a sample of cells contains cells which are, or are predisposed towards becoming, transformed cells. Moreover, the subject method may be used to assess the phenotype of cells which are known to be transformed, the phenotyping results being useful in planning a particular therapeutic regimen. For example, very high levels ofthe marker polypeptide in sample cells is a powerful diagnostic and prognostic marker for a cancer. The observation of marker polypeptide levels may be utilized in decisions regarding, for example, the use of more aggressive therapies. [159] As set out above, one aspect ofthe present invention relates to diagnostic assays for determining, in the context of cells isolated from a patient, if the level of a marker polypeptide is significantly reduced in the sample cells. The term "significantly reduced" refers to a cell phenotype wherein the cell possesses a reduced cellular amount ofthe marker polypeptide relative to a normal cell of similar tissue origin. The assay evaluates the level of marker polypeptide in the test cells, and may compare the measured level with marker polypeptide detected in at least one control cell, for example, a normal cell and/or a transformed cell of known phenotype. [160] Another aspect ofthe subject invention is the ability to quantitate the level of marker polypeptide as determined by the number of cells associated with a normal or abnormal marker polypeptide level. The number of cells with a particular marker polypeptide phenotype may then be coπelated with patient prognosis, hi one embodiment ofthe invention, the marker polypeptide phenotype of a lesion is determined as a percentage of cells in a biopsy which are found to have abnormally high/low levels ofthe marker polypeptide. Such expression may be detected by immunohistochemical assays, dot-blot assays, ELISA, and the like.
[161] Where tissue samples are employed, immunohistochemical staining may be used to determine the number of cells having the marker polypeptide phenotype. For such staining, a multiblock of tissue may be taken from the biopsy or other tissue sample and subjected to proteolytic hydrolysis, employing such agents as protease K or pepsin. In certain embodiments, it may be desirable to isolate a nuclear fraction from the sample cells and detect the level ofthe marker polypeptide in the nuclear fraction.
[162] The tissue samples are fixed by treatment with a reagent such as formalin, glutaraldehyde, methanol, or the like. The samples are then incubated with an antibody (e.g, a monoclonal antibody) with binding specificity for the marker polypeptides. This antibody may be conjugated to a label for subsequent detection of binding. Samples are incubated for a time sufficient for formation ofthe immunocomplexes. Binding ofthe antibody is then detected by virtue of a label conjugated to this antibody. Where the antibody is unlabeled, a second labeled antibody may be employed, for example, which is specific for the isotype ofthe anti-marker polypeptide antibody. Examples of labels which may be employed include radionuclides, fluorescers, chemiluminescers, enzymes, and the like.
[163] Where enzymes are employed, the substrate for the enzyme may be added to the samples to provide a colored or fluorescent product. Examples of suitable enzymes for use in conjugates include horseradish peroxidase, alkaline phosphatase, malate dehydrogenase, and the like. Where not commercially available, such antibody-enzyme conjugates are readily produced by techniques known to those skilled in the art. [164] In one embodiment, the assay is performed as a dot blot assay. The dot blot assay finds particular application where tissue samples are employed as it allows determination ofthe average amount ofthe marker polypeptide associated with a single cell by coπelating the amount of marker polypeptide in a cell-free extract produced from a predetermined number of cells. [165] It is well established in the cancer literature that tumor cells ofthe same type (e.g, lung and/or colon tumor cells) may not show uniformly increased expression of mdividual oncogenes or uniformly decreased expression of mdividual tumor suppressor genes. There may also be varying levels of expression of a given marker gene even between cells of a given type of cancer, further emphasizing the need for reliance on a battery of tests rather than a single test. Accordingly, in one aspect, the invention provides for a battery of tests utilizing a number of probes ofthe invention, i order to improve the reliability and/or accuracy ofthe diagnostic test.
[166] In one embodiment, the present invention also provides a method wherein nucleic acid probes are immobilized on a DNA chip in an organized aπay. Oligonucleotides may be bound to a solid support by a variety of processes, including lithography. For example, a chip may hold up to 250,000 oligonucleotides. These nucleic acid probes comprise a nucleotide sequence, for example, at least about 12, 15, 25, or 40 nucleotides in length, and up to all or nearly all of a sequence which is complementary to a portion ofthe coding sequence of a marker nucleic acid sequence and is differentially expressed in tumor cells. The present invention provides significant advantages over the available tests for various cancers, because it increases the reliability ofthe test by providing an aπay of nucleic acid markers on a single chip.
[167] The method includes obtaining a biopsy, which is optionally fractionated by cryostat sectioning to enrich tumor cells. The DNA or RNA is then extracted, amplified, and analyzed with a DNA chip to determine the presence of absence ofthe marker nucleic acid sequences.
[168] In one embodiment, the nucleic acid probes are spotted onto a substrate in a two- dimensional matrix or aπay. Samples of nucleic acids may be labeled and then hybridized to the probes. Double-stranded nucleic acids, comprising the labeled sample nucleic acids bound to probe nucleic acids, may be detected once the unbound portion ofthe sample is washed away.
[169] The probe nucleic acids may be spotted on substrates including glass, nitrocellulose, etc. The probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions. The sample nucleic acids can be labeled using radioactive labels, fluorophores, chromophores, etc.
[170] In yet another embodiment, the invention contemplates using a panel of antibodies which are generated against the marker polypeptides of this invention. Such a panel of antibodies may be used as a reliable diagnostic probe for cancer. The assay ofthe present invention comprises contacting a biopsy sample containing cells, for example, lung cells, with a panel of antibodies to one or more ofthe encoded products to determine the presence or absence ofthe marker polypeptides.
[171] The diagnostic methods ofthe subject invention may also be employed as follow-up to treatment, for example, quantitation ofthe level of marker polypeptides may be indicative ofthe effectiveness of cuπent or previously employed cancer therapies as well as the effect of these therapies upon patient prognosis.
[172] In addition, the marker nucleic acids or marker polypeptides may be utilized as part of a diagnostic panel for initial detection, follow-up screening, detection of reoccuπence, and post- treatment monitoring for chemotherapy or surgical treatment.
[173] Accordingly, the present invention makes available diagnostic assays and reagents for detecting gain and/or loss of marker polypeptides from a cell in order to aid in the diagnosis and phenotyping of proliferative disorders arising from, for example, tumorigenic transfoπnation of cells.
[174] The diagnostic assays described above may be adapted to be used as prognostic assays, as well. Such an application takes advantage ofthe sensitivity ofthe assays ofthe invention to events which take place at characteristic stages in the progression of a tumor. For example, a given marker gene may be up- or down-regulated at a very early stage, perhaps before the cell is iπeversibly committed to developing into a malignancy, while another marker gene may be characteristically up- or down-regulated only at a much later stage. Such a method could involve the steps of contacting the mRNA of a test cell with a nucleic acid probe derived from a given marker nucleic acid which is expressed at different characteristic levels in cancerous or precancerous cells at different stages of tumor progression, and determining the approximate amount of hybridization ofthe probe to the mRNA ofthe cell, such amount being an indication of the level of expression ofthe gene in the cell, and thus an indication ofthe stage of tumor progression ofthe cell; alternatively, the assay may be carried out with an antibody specific for the gene product ofthe given marker nucleic acid, contacted with the proteins ofthe test cell. A battery of such tests will disclose not only the existence and location of a tumor, but also will allow the clinician to select the mode of treatment most appropriate for the tumor, and to predict the likelihood of success of that treatment.
[175] The methods ofthe invention may also be used to follow the clinical course of a tumor. For example, the assay ofthe invention may be applied to a tissue sample from a patient; following treatment ofthe patient for the cancer, another tissue sample is taken and the test repeated. Successful treatment will result in either removal of all cells which demonstrate differential expression characteristic ofthe cancerous or precancerous cells, or a substantial increase in expression ofthe gene in those cells, perhaps approaching or even suφassing normal levels.
[176] In yet another embodiment, the invention provides methods for determining whether a subject is at risk for developing a disease, such as a predisposition to develop cancer, associated with abeπant activity of a polypeptide, wherein the abeπant activity ofthe polypeptide is characterized by detecting the presence or absence of a genetic lesion characterized by at least one of (a) an alteration affecting the integrity of a gene encoding a marker polypeptides, or (b) the mis- expression ofthe encoding nucleic acid. To illustrate, such genetic lesions may be detected by ascertaining the existence of at least one of (i) a deletion of one or more nucleotides from the nucleic acid sequence, (ii) an addition of one or more nucleotides to the nucleic acid sequence, (iii) a substitution of one or more nucleotides ofthe nucleic acid sequence, (iv) a gross chromosomal rearrangement ofthe nucleic acid sequence, (v) a gross alteration in the level of a messenger RNA transcript ofthe nucleic acid sequence, (vi) abeπant modification ofthe nucleic acid sequence, such as ofthe methylation pattern ofthe genomic DNA, (vii) the presence of a non-wild type splicing pattern of a messenger RNA transcript ofthe gene, (viii) a non-wild type level ofthe marker polypeptide, (ix) allelic loss ofthe gene, and/or (x) inappropriate post-translational modification ofthe marker polypeptide.
[177] The present invention provides assay techniques for detecting lesions in the encoding nucleic acid sequence. These methods include, but are not limited to, methods involving sequence analysis, Southern blot hybridization, restriction enzyme site mapping, and methods involving detection of absence of nucleotide pairing between the nucleic acid to be analyzed and a probe.
[178] Specific diseases or disorders, for example, genetic diseases or disorders, are associated with specific allelic variants of polymoφhic regions of certain genes, which do not necessarily encode a mutated protein. Thus, the presence of a specific allelic variant of a polymoφhic region of a gene in a subject may render the subject susceptible to developing a specific disease or disorder. Polymoφhic regions in genes, may be identified, by determining the nucleotide sequence of genes in populations of individuals. If a polymoφhic region is identified, then the link with a specific disease may be determined by studying specific populations of individuals, for example, individuals which developed a specific disease, such as cancer. A polymoφhic region may be located in any region of a gene, for example, exons, in coding or non-coding regions of exons, introns, and promoter region.
[179] In an exemplary embodiment, there is provided a nucleic acid composition comprising a nucleic acid probe including a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence of a gene or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with the subject genes or naturally occuπing mutants thereof. The nucleic acid of a cell is rendered accessible for hybridization, the probe is contacted with the nucleic acid ofthe sample, and the hybridization ofthe probe to the sample nucleic acid is detected. Such techniques may be used to detect lesions or allelic variants at either the genomic or mRNA level, including deletions, substitutions, etc, as well as to determine mRNA transcript levels.
[180] An example of a detection method is allele specific hybridization using probes overlapping the mutation or polymoφhic site and having about 5, 10, 20, 25, or 30 nucleotides around the mutation or polymoφhic region. In one embodiment ofthe invention, several probes capable of hybridizing specifically to allelic variants are attached to a solid phase support, for example, a "chip." Mutation detection analysis using these chips comprising oligonucleotides, also termed "DNA probe aπays" is described, for example, by Cronin, et al, (Human Mutation 7:244, 1996). In one embodiment, a chip may comprise all the allelic variants of at least one polymoφhic region of a gene. The solid phase support is then contacted with a test nucleic acid and hybridization to the specific probes is detected. Accordingly, the identity of numerous allelic variants of one or more genes may be identified in a simple hybridization experiment.
[181] In certain embodiments, detection ofthe lesion comprises utilizing the probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligase chain reaction (LCR) (see, e.g., Landegran, et al. Science 241: 1077-1080, 1988; Nakazaw, et al, Proc. Natl. Acad. Sci. USA 91:360-364, 1994), the latter of which can be particularly useful for detecting point mutations in the gene (see, e.g., Abravaya, et al, Nuc. Acid Res. 23:675-682, 1995). In an illustrative embodiment, the method includes the steps of (i) collecting a sample of cells from a patient, (ii) isolating nucleic acid (e.g, genomic, mRNA, or both) from the cells ofthe sample, (iii) contacting the nucleic acid sample with one or more primers which specifically hybridize to a nucleic acid sequence under conditions such that hybridization and amplification ofthe nucleic acid (if present) occurs, and (iv) detecting the presence or absence of an amplification product, or detecting the size ofthe amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any ofthe techniques used for detecting mutations described herein.
[182] The invention thus, also encompasses methods of screening for agents which inhibit or enhance the expression ofthe nucleic acid markers in vitro, comprising exposing a cell or tissue in which the marker nucleic acid mRNA is detectable in cultured cells to an agent in order to determine whether the agent is capable of inhibiting or enhancing production ofthe mRNA; and determining the level of mRNA in the exposed cells or tissue, wherein a decrease in the level of the mRNA after exposure ofthe cell line to the agent is indicative of inhibition ofthe marker nucleic acid mRNA production and an increase in mRNA levels is indicative of enhancement of maker mRNA production.
[183] Alternatively, the screening method may include in vitro screening of a cell or tissue in which marker protein is detectable in cultured cells to an agent suspected of inhibiting or enhancing production ofthe marker protein; and determining the level ofthe marker protein in the cells or tissue, wherein a decrease in the level of marker protein after exposure ofthe cells or tissue to the agent is indicative of inhibition of marker protein production and an increase on the level of marker protein is indicative of enhancement of marker protein production.
[184] The invention also encompasses in vivo methods of screening for agents which inhibit or enhance expression ofthe marker nucleic acids, comprising exposing a subject having tumor cells in which marker mRNA or protein is detectable to an agent suspected of inhibiting or enhancing production of marker mRNA or protein; and determining the level of marker mRNA or protein in tumor cells ofthe exposed mammal. A decrease in the level of marker mRNA or protein after exposure ofthe subject to the agent is indicative of inhibition of marker nucleic acid expression and an increase in the level of marker mRNA or protein is indicative of enhancement of marker nucleic acid expression.
[185] Accordingly, the invention provides a method comprising incubating a cell expressing the marker nucleic acids with a test compound and measuring the mRNA or protein level. The invention further provides a method for quantitatively determining the level of expression ofthe marker nucleic acids in a cell population, and a method for determining whether an agent is capable of increasing or decreasing the level of expression ofthe marker nucleic acids in a cell population. The method for determining whether an agent is capable of increasing or decreasing the level of expression ofthe marker nucleic acids in a cell population comprises the steps of (a) preparing cell extracts from control and agent-treated cell populations, (b) isolating the marker polypeptides from the cell extracts, and (c) quantifying (e.g, in parallel) the amount of an immunocomplex formed between the marker polypeptide and an antibody specific to said polypeptide. The marker polypeptides of this invention may also be quantified by assaying for its bioactivity. Agents that induce an increase in the marker nucleic acid expression may be identified by their ability to increase the amount of immunocomplex formed in the treated cell as compared with the amount ofthe immunocomplex formed in the control cell. In a similar manner, agents that decrease expression ofthe marker nucleic acid may be identified by their ability to decrease the amount ofthe immunocomplex formed in the treated cell extract as compared to the control cell. Predictive Assays
[186] Laboratory-based assays, which can predict clinical benefit from a given anti-cancer agent, will greatly enhance the clinical management of patients with cancer. In order to assess this effect, a biomarker associated with the anti-cancer agent may be analyzed in a biological sample (e.g, tumor sample, plasma) before, during, and following treatment.
[187] Another approach to monitor treatment is an evaluation of serum proteomic spectra. Specifically, plasma samples may be subjected to mass spectroscopy (e.g, surface-enhanced laser desoφtion and ionization) and a proteomic spectra may be generated for each patient. A set of spectra, derived from analysis of plasma from patients before and during treatment, may be analyzed by an iterative searching algorithm, which can identify a proteomic pattern that completely discriminates the treated samples from the untreated samples. The resulting pattern may then be used to predict the clinical benefit following treatment.
[188] Global gene expression profiling of biological samples (e.g, tumor biopsy samples, blood samples) and bioinformatics-driven pattern identification may be utilized to predict clinical benefit and sensitivity, as well as development of resistance to an anti-cancer agent. For example, RNA isolated from cells derived from whole blood from patients before and during treatment may be used to generate blood cell gene expression profiles utilizing Affymetrix GeneChip technology and algorithms. These gene expression profiles may then predict the clinical benefit from treatment with a particular anti-cancer agent.
[189] Analysis ofthe biochemical composition of urine by ID -NMR (Nuclear Magnetic Resonance) may also be utilized as a predictive assay. Pattern recognition techniques may be used to evaluate the metabolic response to treatment with an anti-cancer agent and to coπelate this response with clinical endpoints. The biochemical or endogenous metabolites excreted i urine have been well-characterized by proton NMR for normal subjects (Zuppi, et al, Clin Chim Acta 265:85-97, 1997). These metabolites (approximately 30-40) represent the by-products ofthe major metabolic pathways, such as the citric acid and urea cycles. Drug-, disease-, and genetic-stimuli have been shown to produce metabolic-specific changes in baseline urine profiles that are indicative ofthe timeline and magnitude ofthe metabolic response to the stimuli. These analyses are multi- variant and therefore use pattern recognition techniques to improve data inteφretation. Urinary metabolic profiles may be coπelated with clinical endpoints to determine the clinical benefit.
Kits
[190] The invention further provides kits for determining the expression level of genes characteristic of small molecule efficacy. The kits may be useful for identifying subjects that are predisposed to developing cancer or who have cancer, as well as for identifying and validating therapeutics for cancer. In one embodiment, the kit comprises a computer readable medium on which is stored one or more gene expression profile of diseased cells of cancer, or at least values representing levels of expression of one or more genes characteristic of small molecule efficacy in a diseased cell. The computer readable medium can also comprise gene expression profiles of counteφart normal cells, diseased cells treated with a drug, and any other gene expression profile described herein. The kit can comprise expression profile analysis software capable of being loaded into the memory of a computer system.
[191] A kit can comprise a microarray comprising probes of genes characteristic of small molecule efficacy. A kit can comprise one or more probes or primers for detecting the expression level of one or more genes characteristic of small molecule efficacy and/or a solid support on which probes attached and which can be used for detecting expression of one or more genes characteristic of small molecule efficacy in a sample. A kit may further comprise nucleic acid controls, buffers, and instructions for use.
[192] Other kits provide compositions for treating cancer. For example, a kit can also comprise one or more nucleic acids coπesponding to one or more genes characteristic of small molecule efficacy (e.g, for use in treating a patient having cancer). The nucleic acids can be included in a plasmid or a vector (e.g, a viral vector). Other kits comprise a polypeptide encoded by a gene characteristic of cancer or an antibody to a polypeptide. Yet other kits comprise compounds identified herein as agonists or antagonists of genes characteristic of small molecule efficacy. The compositions may be pharmaceutical compositions comprising a pharmaceutically acceptable excipient.
EXAMPLES [193] It will be apparent to those skilled in the art that the examples and embodiments described herein are by way of illustration and not of limitation, and that other examples may be used without departing from the spirit and scope ofthe present invention, as set forth in the claims.
Example 1. Tumor Xenograft Gene Expression Profiling Protocol
A. Tumor implantation and excision
[194] Female nude mice ranging between 11-19 weeks of age, and with an average weight of approximately 18-25 grams were used in these studies. Separately, human Breast (MDA-MB-231) cancer cell lines were grown in tissue culture to approximately 70% confluency. Cells were harvested on the day of implant (5 x 106 cells/mouse), and were suspended in Hanks Balanced Salt Solution from the time of harvest to the time of implant at which time each mouse received a 0.2 ml injection ofthe cell suspension ofthe appropriate cell innoculum. The cells were injected subcutaneously in the right flank of each mouse and tumors were monitored for growth. Time of staging (dosing) was deteratined when tumors reached a size of 200-300 mm3. The vehicle used for the PDGFR-β inhibitor was 40% PEG, 10% ethanol, and 50% 50 mM methanesulfonic acid. Dosing was oral at 25 mg/kg and 75 mg/kg b.i.d. for 4 days. On day 4, at 4 hours after the dose, mice were euthanized and three tumor xenografts for each drug level and three vehicle-treated tumors were harvested and snap frozen. Table 1 describes the genes that were up- and down- regulated greater than or equal to two-fold in the PDGFR-β inhibitor-treated xenografts relative to vehicle-treated xenografts.
B. RNA extraction and cRNA preparation
[195] Total RNA was extracted from tumor explants or cell lines using TRIzol reagent (Life Technologies, Rockville, MD) according to a modified vendor protocol which utilizes the RNeasy protocol (Qiagen, Valencia, CA). After homogenization with a Brinkmann Polytron PT10/35 (Brinkmann, Westbury, NY) and phase separation with chloroform, samples were applied to RNeasy columns. RNA samples were treated with DNase I using RNase-free DNase Set (Qiagen, Valencia, CA).
[196] After elution and quantitation with UV spectrophotometry, each sample was reverse transcribed into double-stranded cDNA using the Gibco Superscript II Choice System for RT- PCR according to vendor protocol (Invitrogen, Carlsbad, CA).
[197] Samples were organically extracted and ethanol precipitated. Approximately 1 μg cDNA was then used in an in vitro transcription reaction incoφorating biotinylated nucleotides using an RNA labeling kit (Enzo Diagnostics, NY). The resulting cRNA was put through RNeasy clean-up protocol and then quantified using UV spectrophotometry. The cRNA (15 μg) was fragmented in the presence of MgOAc and KOAc at 94°C. Fragmented RNA (10 μg) was loaded onto each aπay, one cRNA sample per aπay. Aπays were hybridized for 16 hours at 45°C rotating at 60 φm in an Affymetrix GeneChip Hybridization Oven 640.
C. Microarray Suite 5.0 analysis
[198] Following hybridization, aπays were stained with Phycoerythrm-conjugated Streptavidin, placed in an Agilent GeneAπay Scanner and then exposed to a 488 n laser, causmg excitation of the phycoerythrin. The Microaπay Suite 5.0 software digitally converts the intensity of light given off by the aπay into a numeric value indicative of levels of gene expression. Because each aπay represented a single animal sample, treated animals were compared to the vehicle animals and relative fold changes of genes were obtained. Those genes increased or decreased by at least 2- fold change were considered significant and chosen for further analysis and are listed on Table 1.
TABLE 1
Figure imgf000044_0001
TABLE 1 (cont'd)
Figure imgf000045_0001
*(aa): amino acid SEQ ID NO.

Claims

We claim:
1. A method to monitor the response of a patient being treated for cancer by administering an anti-cancer agent, comprising the steps of: (a) determining the level of expression of one or more genes or gene products in a first biological sample taken from the patient prior to treatment with the anti-cancer agent; (b) determining the level of expression of one or more genes or gene products in at least a second biological sample taken from the patient subsequent to the treatment with the anti-cancer agent; and (c) comparing the level of expression of one or more one genes(s) or gene products in the second biological sample with the level of expression of one or more one genes(s) or v gene products in the first biological sample; wherein a change in the level of expression of one or more genes or gene products in the second biological sample compared to the level of expression of one or more genes or gene products in the first biological sample indicates the efficacy ofthe treatment with the anti-cancer agent.
2. The method of claim 1, wherein the anti-cancer agent is an inhibitor of PDGFR-β.
3. The method of claim 1 or 2, wherein one or more genes are selected from the group consisting of SEQ ID NOs: 1-20.
4. The method of claim 1 or 2, wherein one or more gene products are polypeptides selected from the group consisting of SEQ ID NOs: 21-40.
5. A method to predict the response of a patient to an anti-cancer agent, comprising the steps of: (a) determining the level of expression of one or more genes or gene products in a first biological sample taken from the patient prior to treatment with the anti-cancer agent; (b) determining the level of expression of one or more genes or gene products in at least a second biological sample taken from the patient, wherein the second biological sample is exposed to the anti-cancer agent; and (c) comparing the level of expression of one or more one genes(s) or gene products in the second biological sample with the level of expression of one or more one genes(s) or gene products in the first biological sample; wherein a change in the level of expression of one or more genes or gene products in the second biological sample compared to the level of expression of one or more genes or gene products in the first biological sample predicts the response ofthe patient to an anti- cancer agent.
6. The method of claim 5, wherein the anti-cancer agent is an inhibitor of PDGFR-β.
7. The method of claim 5 or 6, wherein one or more genes are selected from the group consisting of SEQ ID NOs: 1-20.
8. The method of claim 5 or 6, wherein one or more gene products are polypeptides selected from the group consisting of SEQ ID NOs: 21-40.
9. A method for identifying a compound useful for the treatment of cancer comprising the steps of: (a) analyzing the level of expression of one or more genes and/or gene products in a cell or tissue sample prior to treatment with the compound; (b) analyzing the level of expression of one or more genes and/or gene products in a cell or tissue sample subsequent to treatment with the compound; wherein a variation in the expression level ofthe gene and/or gene product is indicative of drug efficacy.
10. The method of claim 9, wherein the compound is an inhibitor of PDGFR-β.
11. The method of claim 9 or 10, wherein one or more genes are selected from the group consisting of SEQ ID NOs: 1-20.
12. The method of claim 9 or 10, wherein one or more gene products are polypeptides selected from the group consisting of SEQ ID NOs: 21-40.
13. A method for providing a patient diagnosis for cancer, comprising the steps of: (a) determining the level of expression of one or more genes or gene products in a first biological sample taken from the patient; (b) determining the level of expression of one or more genes or gene products in at least a second biological sample taken from a normal patient sample; and (c) comparing the level of expression of one or more genes or gene products in the first biological sample with the level of expression of one or more genes or gene products in the second biological sample; wherein a change in the level of expression of one or more genes or gene products in the first biological sample compared to the level of expression of one or more genes or gene products in the second biological sample is a diagnostic ofthe disease.
14. The method of claim 13, wherein one or more genes are selected from the group consisting of SEQ ID NOs: 1-20.
15. The method of claim 13, wherein one or more gene products are polypeptides selected from the group consistmg of SEQ ID NOs: 21-40.
16. An aπay comprising two or more probes coπesponding to two or more genes selected from the group consisting of SEQ ID NOs: 1-20.
17. An aπay comprising two or more polypeptides selected from the group consisting of SEQ ID NOs: 21-40.
18. A test kit comprising a primer or probe for measuring the expression level of a nucleic acid selected from SEQ ID NOs: 1-20.
19. The test kit of claim 18, further comprising one or more components selected from solutions for suspending or fixing tissue or cell samples, solutions for lysing cells, hybridization solutions, solutions for the isolation of nucleic acids, control samples, and instructions for using the kit.
20. A test comprising an antibody specific for a polypeptide selected from SEQ ID NOs: 21- 40.
21. The test kit of claim 20, wherein the antibody is labeled.
22. The test kit of claim 20 or 21, further comprising one or more components selected from solutions for suspending or fixing tissue or cell samples, solutions for lysing cells, substrates, buffer reagents, blocker reagents, blotting reagents, control samples, and instructions for using the kit.
PCT/US2005/000518 2004-01-06 2005-01-05 Gene expression profiles and methods of use WO2005067650A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US53482504P 2004-01-06 2004-01-06
US60/534,825 2004-01-06

Publications (2)

Publication Number Publication Date
WO2005067650A2 true WO2005067650A2 (en) 2005-07-28
WO2005067650A3 WO2005067650A3 (en) 2006-08-17

Family

ID=34794323

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/000518 WO2005067650A2 (en) 2004-01-06 2005-01-05 Gene expression profiles and methods of use

Country Status (1)

Country Link
WO (1) WO2005067650A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030152980A1 (en) * 2001-12-21 2003-08-14 Whitehead Institute For Biomedical Research Prostate cancer diagnosis and outcome prediction by expression analysis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030152980A1 (en) * 2001-12-21 2003-08-14 Whitehead Institute For Biomedical Research Prostate cancer diagnosis and outcome prediction by expression analysis

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE MEDLINE [Online] ABRAMS ET AL.: 'SU11248 inhibits KIT, and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer' Retrieved from STN Database accession no. (2003226905) & MOLECULAR CANCER THERAPEUTICS vol. 2, no. 5, May 2003, pages 471 - 478 *
KHAN ET AL.: 'Expression profiling in cancer using cDNA microarrays' ELECTROPHORESIS vol. 20, 1999, pages 223 - 229 *

Also Published As

Publication number Publication date
WO2005067650A3 (en) 2006-08-17

Similar Documents

Publication Publication Date Title
US20040146921A1 (en) Expression profiles for colon cancer and methods of use
US8329408B2 (en) Methods for prognosis and monitoring cancer therapy
EP1945819B1 (en) Gene expression profiles and methods of use
US20070065827A1 (en) Gene expression profiles and methods of use
US20070178494A1 (en) Methods for prediction and prognosis of cancer, and monitoring cancer therapy
WO2005032495A2 (en) Gene expression profiles and methods of use
US20040191819A1 (en) Expression profiles for breast cancer and methods of use
US20070134670A1 (en) Methods for prediction and prognosis of cancer, and monitoring cancer therapy
US20040121375A1 (en) Methods for prediction and prognosis of cancer, and monitoring cancer therapy
WO2006032009A2 (en) Methods for prediction and prognosis of cancer, and monitoring cancer therapy
WO2005067650A2 (en) Gene expression profiles and methods of use

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 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

NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
122 Ep: pct application non-entry in european phase