WO2003065993A2 - Primary rat hepatocyte toxicity modeling - Google Patents

Primary rat hepatocyte toxicity modeling Download PDF

Info

Publication number
WO2003065993A2
WO2003065993A2 PCT/US2003/003482 US0303482W WO03065993A2 WO 2003065993 A2 WO2003065993 A2 WO 2003065993A2 US 0303482 W US0303482 W US 0303482W WO 03065993 A2 WO03065993 A2 WO 03065993A2
Authority
WO
WIPO (PCT)
Prior art keywords
genes
expression
tables
gene
probes
Prior art date
Application number
PCT/US2003/003482
Other languages
French (fr)
Other versions
WO2003065993A8 (en
Inventor
Donna Mendrick
Mark Porter
Kory Johnson
Brandon Higgs
Arthur Castle
Michael Orr
Michael Elashoff
Original Assignee
Gene Logic, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gene Logic, Inc. filed Critical Gene Logic, Inc.
Priority to JP2003565419A priority Critical patent/JP2006502693A/en
Priority to CA002471631A priority patent/CA2471631A1/en
Priority to EP03715981A priority patent/EP1578393A4/en
Priority to AU2003219713A priority patent/AU2003219713A1/en
Publication of WO2003065993A2 publication Critical patent/WO2003065993A2/en
Publication of WO2003065993A8 publication Critical patent/WO2003065993A8/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
    • 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
    • C12Q2600/142Toxicological screening, e.g. expression profiles which identify toxicity
    • 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
    • C12Q2600/158Expression markers

Definitions

  • the present invention is based on the elucidation of the global changes in gene expression in primary hepatocytes exposed to known toxins, in particular hepatotoxins, as compared to unexposed cells as well as the identification of individual genes that are differentially expressed upon toxin exposure.
  • the invention includes methods of predicting at least one toxic effect of a compound, predicting the progression of a toxic effect of a compound, and predicting the hepatoxicity of a compound.
  • the invention also includes methods of identifying agents that modulate the onset or progression of a toxic response. Also provided are methods of predicting the general pathology classes and cellular pathways that a compound modulates in a cell.
  • the invention includes methods of identifying agents that modulate protein activities.
  • the invention provides probes comprising sequences that specifically hybridize to genes in Tables 1-5XX. Also provided are solid supports comprising at least two of the previously mentioned probes.
  • the invention also includes a computer system that has a database containing information identifying the expression level in a tissue or cell sample exposed to a hepatotoxin of a set of genes comprising at least two genes in Tables 1-5XX. DETAILED DESCRIPTION
  • Changes in gene expression are also associated with the effects of various chemicals, drags, toxins, pharmaceutical agents and pollutants on an organism or cells.
  • the lack of sufficient expression of functional tumor suppressor genes and/or the over expression of oncogene/protooncogenes after exposure to an agent could lead to tumorgenesis or hype ⁇ lastic growth of cells (Marshall, Cell, 64: 313-326 (1991); Weinberg, Science, 254:1138-1146 (1991)).
  • changes in the expression levels of particular genes e.g. oncogenes or tumor suppressors
  • Monitoring changes in gene expression may also provide certain advantages during drag screening and development. Often drugs are screened for the ability to interact with a major target without regard to other effects the drugs have on cells. These cellular effects may cause toxicity in the whole animal, which prevents the development and clinical use of the potential drug.
  • the present inventors have examined primary rat hepatocytes exposed to the known hepatotoxins which induce detrimental liver effects, to identify global and individual changes in gene expression induced by these compounds. These global changes in gene expression, which can be detected by the production of expression profiles, as well as the individual genes, provide useful toxicity markers that can be used to monitor toxicity and/or toxicity progression by a test compound. Expression profiles, as well as the individual markers, may also be used to monitor or detect various disease or physiological states, disease progression, drug efficacy and drug metabolism.
  • Amiodarone (Cordarone®) is an anti-anhythmic agent whose chemical stracture contains a benzofuran ring (ring A) coupled to a p-OH-benzene structure substituted with 2 iodines and a diethyl-ethanolamine side chain (ring B).
  • This drug is known to cause damage to the liver and has been shown to adversely effect the mitochondria by uncoupling oxidative phosphorylation and inhibiting beta-oxidation and respiration. Inhibition of respiration decreases ATP and increases production of reactive oxygen species, which in turn cause lipid peroxidation.
  • Aromatic and aliphatic isothiocyanates are commonly used soil fumigants and pesticides (Shaaya et al, Pesticide Science 44(3):249-253 (1995); Cairns et al, J Assoc Official Analytical Chemists 71(3):547-550 (1988)).
  • ANIT ⁇ -naphthylisothiocyanate
  • ANIT fails to produce extensive necrosis, but was found to produce inflammation and edema in the portal tract of the liver (Maziasa et al, Toxicol Appl Pharmacol 110:365-373 (1991)).
  • ANIT-induced hepatotoxicity may also characterized by cholangiolitic hepatitis and bile duct damage.
  • Acute hepatotoxicity caused by ANIT in rats is manifested as neutrophil-dependent necrosis of bile duct epithelial cells (BDECs) and hepatic parenchymal cells. These changes minor the cholangiolitic hepatitis found in humans (Hill, Toxicol Sci 47:118-125 (1999)).
  • Histological changes include an infiltration of polymo ⁇ honuclear neutrophils and elevated number of apoptotic hepatocytes (Calvo et al, J Cell Biochem 80(4):461-470 (2001)).
  • Other known hepatotoxic effects of exposure to ANIT include a damaged antioxidant defense system, decreased activities of superoxide dismutase and catalase (Ohta et al, Toxicology 139(3):265-275 (1999)), and the release of proteases from the infiltrated neutrophils, alanine aminotransferase, cathepsin G, elastase, which mediate hepatocyte killing (Hill et ai, Toxicol Appl Pharmacol 148(1):169-175 (1998)).
  • Acetominophen is a widely used analgesic and antipyretic agent that is an effective substitute for aspirin. Although acetaminophen does not have anti-inflammatory properties, it is preferably given to patients with ulcers or patients in whom prolonged clotting times would not be desirable. It also preferably taken by people who do not tolerate aspirin well.
  • Acetominophen is metabolized to N-acetyl- -benzoquinoneimine (NAPQI) by N- hydroxylation in a cytochrome P450-mediated process.
  • NAPQI N-acetyl- -benzoquinoneimine
  • This highly reactive intermediate which reacts with sulfhydryl groups in glutathione, and in other liver proteins following the depletion of glutathione, can cause centrilobular hepatic necrosis (particularly in zone 3), renal tubular necrosis, and hepatic and renal failure (Goodman and Gilman' s The Pharmacological Basis of Therapeutics. Ninth Ed.. Hardman et al, eds., pp.
  • AY-25329 is a phenothiazine that has been shown to be toxic in liver and in kidney tissue, where it can cause nephrosis.
  • Phenothiazines are a class of psychoactive drugs that are used to treat schizophrenia, paranoia, mania, hyperactivity in children, some forms of senility, and anxiety (http://www.encyclopedia.com/articlesnew/ 36591.html). Side effects associated with prolonged use of these drugs are reduced blood pressure, Parkinsonism, reduction of motor activity, and visual impairment.
  • the present inventors have noted indications of liver and renal effects of AY-25329 by changes in serum chemistry. As early as 6 hours after the first dose, statistically significant increases in serum levels of creatinine, BUN, ALT, triglycerides and cholesterol were observed. Most of these markers of renal and liver dysfunction remained altered throughout the 14 day study period. Light microscopic analysis revealed effects in the liver as early as 6 and 24 hours, as evidenced by an increased number of hepatocytic mitotic figures and decreased glycogen content. Following 14 days of repeated dosing, nephrosis and alterations in the peripheral lobes of the liver and in the cytoplasm of hepatocytes were evident in rats dosed with 250 mg/kg/day of AY-25329.
  • Carbamazepine (Tegretol®) is an anti-epieleptic agent. In rats, it has been shown to induce a number of cytochrome P450 enzymes, in particular CYP2B, and the drug may also cause steatohepatitis in humans (Tateishi et al., Chem Biol Interact 117:257-268 (1999); Grieco et al, Eur J Gastioenterol 13(8):973-975 (2001)).
  • CCl 4 -induced hepatotoxicity is dependent on CC1 4 bioactivation to trichloromethyl free radicals by cytochrome P450 enzymes (CYP2E1), localized primarily in centrizonal hepatocytes. Formation of the free radicals leads to membrane lipid peroxidation and protein denaturation resulting in hepatocellular damage or death.
  • CYP2E1 cytochrome P450 enzymes
  • the onset of hepatic injury is rapid following acute administration of CC1 4 to male rats.
  • Mo ⁇ hologic studies have shown cytoplasmic accumulation of lipids in hepatocytes within 1 to 3 hours of dosing, and by 5 to 6 hours, focal necrosis and hydropic swelling of hepatocytes are evident.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • CC1 4 - mediated increases in c-jun and c-fos gene expression Pre-treatment with anti-TNF- ⁇ antibodies has been shown to prevent CC1 4 - mediated increases in c-jun and c-fos gene expression, whereas administration of TNF- ⁇ induced rapid expression of these genes (Braccoleri et al, Hepatol 25:133-141 (1997)).
  • TGF- ⁇ transforming growth factor- ⁇
  • TBRI-III transforming growth factor receptors
  • Chlo ⁇ romazine is a central nervous system depressant that is used as a sedative and also as an anti-nausea or anti-itching medication. The mechanism of action is not known.
  • the drug induces canalicular cholestasis, a condition characterized by a decrease in the volume of bile formed and impaired secretion of solutes into bile, resulting in elevated serum levels of bile salts and bilirubin.
  • Chlo ⁇ romazine has also been shown to inhibit bile acid uptake and canalicular contractility.
  • Bile plugs can form in the bile ducts and canaliculi.
  • Drag-induced cholestasis is also associated with cell swelling, inflammation and cell death (Casarett and Doull's Toxicology: The Basic Science of Poisons, 6th Ed.. Klaassen et al. eds., pp. 476-486, McGraw-Hill Medical Pub.
  • CI-1000 (4H-pynolo:3,2-d:pyrimidin-4-one, 2-amino-3,5-dihydro-7-(3- thienylmethyl)-monohydrochloride monohydrate) is a compound with anti-inflammatory properties. After treatment with CI-1000, increased serum ALT levels, a standard marker of liver toxicity, were observed in dogs.
  • Clofibrate a halogenated phenoxypropanoic acid derivative (ethyl ester of clofibric acid), is an antilipemic agent.
  • LDL low-density lipoprotein
  • HDL high-density lipoprotein
  • the drug has several antilipidemic actions, including activating lipoprotein lipase, which enhances the clearance of triglycerides and very-low-density lipoprotein (VLDL) cholesterol, inhibition of cholesterol and triglyceride biosynthesis, mobilization of cholesterol from tissues, increasing fecal excretion of neutral steroids, decreasing hepatic lipoprotein synthesis and secretion, and decreasing free fatty acid release.
  • activating lipoprotein lipase which enhances the clearance of triglycerides and very-low-density lipoprotein (VLDL) cholesterol
  • VLDL very-low-density lipoprotein
  • Clofibrate has a number of effects on the rat liver, including hepatocellular hypertrophy, cellular proliferation, hepatomegaly, induction of CYP450 isozymes, and induction of palmitoyl Co A oxidation. Long term administration of clofibrate causes increased incidence of hepatocellular carcinoma, benign testicular Leydig cell tumors, and pancreatic acinar adenomas in rats.
  • Clofibrate induces proliferation of peroxisomes in rodents and this effect, rather than genotoxic damage, is believed to be the causative event in rodent carcinogenesis (AHFS Drug Information Handbook 2001, McEvoy, ed., pp.1735- 1738; Electronic Physicians' Desk Reference- Atromid-S (clofibrate) at www.pdr.net; Brown and Goldstein, "Drugs used in the treatment of hyperliproteinemias," in Goodman and Gilman's The Pharmacological Basis of Therapeutics. Eighth ed during Goodman et al, eds., pp. 874-896, Pergamon Press, New York, 1990).
  • Clofibrate also increases hepatic lipid content and alters its normal composition by significantly increasing levels of phosphatidylcholine and phosphatidyl-ethanolamine (Adinehzadeh et al, Chem Res Toxicol 11(5):428-440 (1998)).
  • a rat study of liver hype ⁇ lasia and liver tumors induced by peroxisome proliferators revealed that administration of clofibrate increased levels of copper and altered copper-related gene expression in the neoplastic liver tissues.
  • Cyproterone acetate is a potent androgen antagonist and has been used to treat acne, male pattern baldness, precocious puberty, and prostatic hype ⁇ lasia and carcinoma (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9 ed., p. 1453, J.G. Hardman et al, Eds., McGraw Hill, New York, 1996). Additionally, CPA has been used clinically in hormone replacement therapy to protect the endometrium and decrease menopausal symptoms and the risk of osteoporotic fracture (Schneider, "The role of antiandrogens in hormone replacement therapy," Climacteric 3 (Suppl. 2): 21-27 (2000)).
  • CPA CPA was shown to induce unscheduled DNA synthesis in vitro. After a single oral dose, continuous DNA repair activity was observed after 16 hours. CPA also increased the occurrence of S phase cells, which conoborated the mitogenic potential of CPA in rat liver (Kasper et al, Carcinogenesis 17(10): 2271-2274 (1996)). CPA has also been shown to produce cinhosis in humans (Garty et al, Eur JPediatr 158(5): 367- 370 (1999)).
  • Diclofenac a non-steroidal anti-inflammatory drug
  • diclofenac is rapidly absorbed and then metabolized in the liver by cytochrome P450 isozyme of the CYC2C subfamily (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9 th ed., p. 637, J.G. Hardman et al, eds., McGraw Hill, New York, 1996).
  • diclofenac has been applied topically to treat pain due to corneal damage (Jayamanne et al, Eye ll(Pt. 1): 79-83 (1997); Domic et al, Am J Ophthalmol 125(5): 719-721 (1998)).
  • diclofenac has numerous clinical applications, adverse side-effects have been associated with the drug, such as corneal complications, including corneal melts, ulceration, and severe keratopathy (Guidera et al, Ophthalmology 108(5): 936-944 (2001)).
  • An additional report showed that a patient developed severe hepatitis five weeks after beginning diclofenac treatment for osteoarthritis (Bhogaraju et al, South MedJ92(7): 711-713 (1999)).
  • diclofenac-treated Wistar rats In one study on diclofenac-treated Wistar rats (Ebong et al, Afr JMed Sci 27(3-4): 243-246 (1998)), diclofenac treatment induced an increase in serum chemistry levels of alanine aminotransferase, aspartate aminotransferase, methaemoglobin, and total and conjugated bilirubin. Additionally, diclofenac enhanced the activity of alkaline phosphatase and 5'nucleotidase. A study on humans revealed elevated levels of hepatic transammases and serum creatine when compared to the control group (McKenna et al, Scand J Rheumatol 30(1): 11-18 (2001)).
  • NSAID non-steroidal anti-inflammatory drug
  • NSAID is a difluorophenyl derivative of salicylic acid (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9 th ed., p. 631, J.G. Hardman et al, Eds., McGraw Hill, New York, 1996). It is most frequently used in the treatment of osteoarthritis and musculoskeletal strains.
  • NSAIDs have analgesic, antipyretic and anti-inflammatory actions, however, hepatotoxicity is known to be an adverse side effect of NSAID treatment (Masubucbi et al, J Pharmacol Exp Ther 287:208-213 (1998)).
  • Diflunisal has been shown to be less toxic than other NSAIDs, but it can eventually have deleterious effects on platelet or kidney function (Bergamo et al, Am J Nephrol 9:460-463 (1989)). Other side effects that have been associated with diflunisal treatment are dianhea, dizziness, drowsiness, gas or heartburn, headache, nausea, vomiting, and insomnia (h1 ⁇ ://arthritisinsight.com/medical/ meds/dolobid.html). [0041] In a comparative hepatotoxicity study of 18 acidic NSAIDs, diflunisal was shown to increase LDH leakage in rat hepatocytes, a marker for cell injury, when compared to control samples.
  • DMN causes oxidative stress in liver cells, which may be the link between chronic liver damage and liver fibrosis.
  • Rats treated with DMN showed diffuse fibronectin deposition, elevated hydroxyproline levels (an indicator of fibrosis), increased levels of collagens, fibrous septa, and impaired oxidative balance.
  • Serum levels of ALT and malondialdehyde (MDA) were increased, while serum levels of SOD were decreased (Vendemiale et al, Toxicol Appl Pharmacol 175(2):130-139 (2001); Liu et al, Zhonghua Gan Zang Bing Za Zhi 9 Suppl: 18-20 (2001)).
  • 17 ⁇ -ethinylesfradiol a synthetic estrogen
  • 17 ⁇ -ethinylestradiol has been shown to cause a reversible intiahepatic cholestasis in male rats, mainly by reducing the bile-salt-independent fraction of bile flow (BSIF) (Koopen et al, Hepatology 27:537-545 (1998)). Plasma levels of bilirabin, bile salts, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in this study were not changed.
  • BSIF bile-salt-independent fraction of bile flow
  • the lipid-lowering drag gemfibrozil (Lopid®) is a know peroxisome proliferator in liver tissue, causing both hype ⁇ lasia and enlargement of liver cells.
  • gemfibrozil Upon exposure to gemfibrozil, hepatocarcinogenesis has been observed in rats and mice, and a decrease in alpha-tocopherol and an increase in DT-diaphorase activity have been observed in rats and hamsters (impaired antioxidant capability).
  • Peroxisome prohferators increase the activities of enzymes involved in peroxisomal beta-oxidation and omega-hydroxylation of fatty acids, which results in oxidative stress (O'Brien et al, Toxicol Sci 60(2):271-278 (2001); Carfhew et al, JAppl Toxicol 17(1):47-51 (1997)).
  • hydrazine sulphate used to treat cachexia in cancer patients, isoniazid, an anti-tuberculosis drug, and hydralazine, an anti-hypertensive.
  • These drags are metabolized in vivo to produce hydrazine, among other by-products. Consequently, exposure to hydrazine is by direct contact, e.g., among military and airline personnel, or the result of its production in the body, e.g., in patients with cancer or high blood pressure.
  • hepatotoxic changes characterized by GSH and ATP depletion and induction of fatty liver (increases in liver weight and triglycerides, with the appearance of fatty droplets, swelling of mitochondria and appearance of microbodies) were also found to be dose-dependent (Jenner et al., Arch Toxicol 68(6):349-357 (1994); Scales et al, J Toxicol Environ Health 10(6):941- 953 (1982)).
  • cytochrome P4502E1 cytochrome P4502E1
  • CYP2E1 cytochrome P4502E1
  • exposure to hydrazine and several hydrazine derivatives increased enzyme levels in kidney tissue, increased enzyme levels were not detected in liver tissue (Runge-Morris et al, Drug Metab Dispos 24(7):734-737 (1996)).
  • the mutagenic and hepatocarcinogenic effects of hydrazine were examined in hamster livers.
  • liver DNA showed the presence of methylated guanine, DNA adducts and the impairment of maintenance methylation (impaired methylation of deoxycytosine).
  • Hepatic adenomas and carcinomas also developed in a dose-dependent manner and could be conelated with decreased maintenance methylation (FitzGerald et al, Carcinogenesis 17(12):2703-2709 (1996)).
  • Imipramine a dibenzazepine derivative
  • Imipramine is a tricyclic anti-depressant agent commonly used for the treatment of major depression.
  • rats Experiments in rats have shown that the drug induces cytochrome P450-mediated formation of reactive metabolites, which cause acute cell injury. Decreased levels of glutathione and protein thiols, as well as lactate dehydrogenase leakage, all standard measures of liver toxicity, were also noted (Masubuchi et al., Arch Toxicol 73(3): 147-151 (1999).
  • Indomethacin is a non-steroidal antiinflammatory, antipyretic and analgesic drug commonly used to treat rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, gout and a type of severe, chronic cluster headache characterized by many daily occu ⁇ ences and jabbing pain.
  • This drug acts as a potent inhibitor of prostaglandin synthesis; it inhibits the cyclooxygenase enzyme necessary for the conversion of arachidonic acid to prostaglandins fPDR 47 th Ed.. Medical Economics Co., Inc., Montvale, NJ, 1993; Goodman & Gilman's The Pharmalogjcal Basis of Therapeutics 9 th Ed.. J.G. Hardman et al.
  • Renal papillary necrosis has been observed in rats, and interstitial nephritis with hematuria, proteinuria and nephrotic syndrome have been reported in humans. Patients suffering from renal dysfunction risk developing a reduction in renal blood flow, because renal prostaglandins play an important role in renal perfusion.
  • rats although indomethacin produces more adverse effects in the gastrointestinal tract than in the liver, it has been shown to induce changes in hepatocytic cytochrome P450. In one study, no widespread changes in the liver were observed, but a mild, focal, centrilobular response was noted. Serum levels of albumin and total protein were significantly reduced, while the serum level of urea was increased.
  • LPS lipopolysaccharide
  • LPS lipopolysaccharide
  • the membrane components of LPS are lipid- A, KDO (2-keto-3-deoxy-octulosonic acid), core polysaccharides and O-antigen polysaccharides, the polysaccharide units differing from one bacterium to another (Zjnsser Microbiology 20th Ed.. Joklik et al., eds., pp.
  • nitric oxide synthetase NOS
  • cellular hypertrophy vacuohzation
  • chromosomal emargination cytoplasmic DNA fragmentation and necrosis
  • LBP lipopolysaccharide-binding protein
  • the LPS-LBP complex interacts with the CD 14 receptor, inducing LPS sensitive genes.
  • LBP can be induced in hepatocytes isolated from rats that have not been primed with LPS, indicating that this key regulatory pathway is intact in primary rat hepatocytes (Wan et al, Infect Immun 63(7):2435-2442 (1995)).
  • Lovastatin (Mevacor®) is a cholesterol-lowering agent belonging to a class of compounds, the statins, that are potent inhibitors of HMG-CoA reductase.
  • HMG-CoA reductase inhibitors block the production of cholesterol in the liver leading to a reduction of LDL particles in the plasma.
  • Lovastatin has additional effects on lipid metabolism, including depletion of intracellular pools of sterols and increased synthesis of LDL receptors, leading to enhanced removal of LDL and LDL precursors from plasma.
  • plasma levels of VLDL, IDL and triglycerides Upon treatment with lovastatin, plasma levels of VLDL, IDL and triglycerides also decrease.
  • Oral doses of lovastatin are extensively absorbed by the liver, and the drug is excreted primarily via the liver; less than 13% of its metabolites are excreted in the urine (Goodman and Gilman's The Pharmacological Basis of Therapeutics. Ninth Ed.. Hardman et al, eds., pp. 884-888, McGraw-Hill, New York, 1996).
  • liver damage characterized by elevated levels of hepatic transammases (e.g., AST and ALT), creatinine phosphokinase and alkaline phosphatase, and myopathy, characterized by muscle pain and destruction of skeletal muscle cells.
  • hepatic transammases e.g., AST and ALT
  • creatinine phosphokinase and alkaline phosphatase e.g., AST and ALT
  • myopathy characterized by muscle pain and destruction of skeletal muscle cells.
  • Methotrexate is a widely used antineoplastic drag that is also frequently prescribed for the treatment of psoriasis (a disease characterized by abnormal proliferation of epidermal cells), juvenile lymphoblastic leukemia, rheumatoid arthritis, and a number of other cancerous diseases, such as leukemic meningitis and choriocarcinoma. Although generally not metabolized, at high dosages, metabolites such as 7-hydroxy-methotrexate, a nephrotoxin, do accumulate. Methotrexate polyglutamates are retained in the kidneys for weeks and in the liver for months ((Goodman and Gilman's The Pharmacological Basis of Therapeutics.
  • Methotrexate acts to prevent DNA synthesis and cell replication by inhibiting the rate-limiting enzyme in purine and thymidine synthesis, dihydrofolate reductase (DHFR) (Goodman and Gilman's, supra; Schwartz et al, Proc Nat Acad Sci USA 89(2):594-598 (1992)). It also acts as an suppressant of cell-mediated immune responses.
  • DHFR dihydrofolate reductase
  • biochemical toxicology of methotrexate has been well characterized in man, where long-term administration produces hepatic fibrosis or cinhosis, especially in heavy drinkers, which is linked to persistent, mild-to-moderate, increases in serum transammases, alkaline phosphatases and bilirubin (Reynolds et al, South Med J ' 1 '9(5):536-539 (1986); Tolman et al, JRheumatol 12 (Suppl 12):29-34 (1985)).
  • Methotrexate is a rather long-acting, rapidly reversible DHFR inhibitor, despite its high affinity for the target enzymes in many cell types, which may be due to the formation of methotrexate polyglutamates that reduce the ability of DHFR to pass through cell membranes.
  • the toxic effects of methotrexate may be due to the depletion of tetrahydrofolate cofactors that are required for purine and thymidylate synthesis (methylation reactions in hepatic 1 -carbon metabolism) and to the inliibition of folate- dependent enzymes involved in the metabolism of purines and thymidylate, the inhibition caused by the accumulation of methotrexate polyglutamates and dihydrofolate polyglutamates.
  • methotrexate-induced hepatotoxicity is not yet fully elucidated, partly because the pathological changes in humans are rather difficult to reproduce in animal models, although experiments in rats have shown that, in a dose-dependent fashion, methotrexate produces liver damage ranging from focal to confluent necrosis of zone 3 hepatocytes, with early stage fibrosis (Hall et al, Hepatology 14(5):906-10 (1991)). Other studies in rats have demonstrated that treatment with methotrexate produces intrahepatocytic fat deposits, along with fatty accumulations in hepatocytes that range from fine droplets to large vacuoles.
  • necrosis The areas of necrosis showed signs of the hypoxia associated with congestive failure, as well as anemic infarcts, fibrotic foci of the collapse type, atrophy of the hepatic cords, and hemosiderosis (Custer et al, JNatl Cancer Inst 58(4):1011-1015 (1977)). Hepatotoxicity probably involves interference with triglyceride and other lipid biosynthetic pathways in the liver. For example, studies on rats have shown that methotrexate inhibits the biosynthesis of lipotropic substances such as methionine and choline tlirough its interference with hepatic 1 -carbon metabolism.
  • the drug also inhibits the activity of vitamin B12, another lipotropic factor (Tuma et al, Biochem Pharmacol 24:1327-1331 (1975) and impairs RNA and protein synthesis, triglyceride secretion and total triglyceride esterification (Deboyser et al, Toxic in Vitro 6(2):129-132 (1992).
  • Methotrexate does not appear to be cytotoxic to cultured primary hepatocytes following short-term exposures (up to 3.5 hr), but significant effects on HepG2 growth curves have been observed at low concentrations during the course of 7-day exposures (Wu et al, Proc Natl Acad Sci USA 80(10):3078-3080 (1983)).
  • methotrexate increases hepatic glycogenolysis, oxygen consumption and calcium efflux and decreases glutathione levels (Yamamoto et al, Biochem Pharmacol 44(4):761-767, (1992); de Oliveira et al, Res Commun Chem Pathol Pharmacol 53(2):173-181 (1986); Lindenthal et al, Eur J Pharmacol 228(5-6):289-298 (1993)).
  • NAT2 hepatic N- acetyltransferase 2
  • Phenobarbital a barbiturate
  • Phenobarbital is used as an anti-epileptic, sedative or hypnotic drug and can also be used to treat neuroses with related tension states, such as hypertension, coronary artery disease, gastrointestinal disturbances and preoperative apprehension. Phenobarbital is also found in medications to treat insomnia and headaches (Remington: The Science and Practice of Pharmacy. 19th Ed.. A. R. Gennaro ed., pp. 1164-1165, Mack Publishing Co., Easton, Pennsylvania, 1995).
  • Phenobarbital induces a variety of drag metabolizing enzymes such as cytochrome P450 oxidoreductase, aldehyde dehydrogenases, UDP-glucuronyltransferase, GSTs, epoxide hydrolase, and an assortment of cytochrome P450 monooxygenases (Waxman et al, Biochem J 1281(Pt 3):577-592 (1992); Kaplowitz et al, Biochem J 146(2):351-356 (1975); Tank et al, Biochem Pharmacol 35(24):4563-4569 (1986).
  • cytochrome P450 oxidoreductase aldehyde dehydrogenases
  • UDP-glucuronyltransferase GSTs
  • epoxide hydrolase epoxide hydrolase
  • liver enzymes The induction of liver enzymes is usually accompanied by liver enlargement, proliferation of smooth endoplasmic reticulum, and tumor promotion (Waxman et al, supra; Rice et al, Carcinogenesis 15(2):395-402 (1994); Nims et al, Carcinogenesis 8(1):67-71, (1987). Incidences of cholestasis and hepatocellular injury have also been found (Selim et al, Hepatology 29(5):1347-1351 (1999); Gut et al, Environ Health Perspect 104(Suppl 6):1211-1218 (1999)).
  • Phenobarbital has been classified as nongenotoxic hepatocarcinogen as it induces liver tumors in rodents but lacks detectable signs of genotoxicity using short term in vivo or in vitro assays (Whysner et al, Pharmacol Ther 71(1-2): 153-191 (1996)).
  • phenobarbital The effects of phenobarbital on phospholipid metabolism in rat liver have been studied. In one study, phenobarbital, administered intraperitonially (i.p.), was found to cause an increase in the microsomal phosphatidylcholine content. Additionally, levels of glycerophosphate acyltransferase (GAT), phosphatidate cytidylyltransferase (PCT), phosphatidate phosphohydrolase (PPH) and choline phosphotransferase (CPT) were significantly increased (Hoshi et al, Chem Pharm Bull 38:3446-3448 (1990)).
  • GAT glycerophosphate acyltransferase
  • PCT phosphatidate cytidylyltransferase
  • PPH phosphatidate phosphohydrolase
  • CPT choline phosphotransferase
  • Tacrine l,2,3,4-tetiahydro-9-aminoacridine-hydrochloride
  • AChE acetylcholinesterase
  • Hepatotoxicty caused by tacrine is typically reversible, although cases of severe hepatotoxicity have been seen (Blackard et al, J Clin Gastroenterol 26:57-59 (1998)).
  • the toxicity is characterized by decreased levels of protein synthesis and the release of lactate dehydrogenase, as well as by increased transaminase levels and decreased levels of ATP, glycogen and glutathione. The decrease in protein synthesis may represent a signal leading to cell death (Lagadic-Gossmann et al, Cell Biol Toxicol 14(5):361-373 (1998)).
  • tacrine does not reveal classic signs of hepatotoxicity in rats, gene expression changes due to tacrine administration can be used to predict that the drug will be a liver toxin in humans. This suggests that toxicogenomics might be able to detect drugs that prove to be toxic in the clinic even when classical but more crude measures in preclinical screening fail to detect toxicity.
  • Tamoxifen is a nonsteroidal anti-estrogen used for breast cancer in males and females. Tamoxifen has been associated with changes in liver enzyme levels, disruption of mitochondrial metabolism and, occasionally, with a spectrum of more severe liver abnormalities including fatty liver, cholestasis, hepatic necrosis and NASH (nonalcoholic steatohepatitis) (Duthie et al, Xenobiotica 25(10):1151-1164 (1995); Cardoso et al, Toxicol Appl Pharmacol 176(3):145-152 (2001); Pinol et ai, Gastroenterol Hepatol 23(2):57 '-61 (2000); and Farrell, Semin Liver Dis 22(2):185-194 (2002)).
  • Tetracycline is a broad spectrum antibiotic whose main mechanism of action is the inhibition of bacterial protein synthesis. Hepatic toxicity, principally steatosis, has been observed in patients receiving high doses of tetracycline.
  • Valproate n-dipropylacetic acid, Depakene®
  • Depakene® n-dipropylacetic acid
  • valproate does not affect neuronal responses to GABA, it does increase the activity of the GABA synthetic enzyme, glutamic acid decarboxylase, and it inhibits enzymes that degrade GABA, GABA transaminase and succinic semialdehyde dehydrogenase (Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9th Ed.. Hardman et al, eds., pp. 462, 476 and 477, McGraw-Hill, New York, 1996).
  • hepatitis Fulminant hepatitis, microvesicular steatosis (fatty degeneration), hepatocyte necrosis and hepatic failure can also result. It is believed that hepatoxicity is caused by an accumulation of unsaturated metabolites of valproate, in particular 4-en- valproate, which is structurally similar to two known hepatotoxins, 4-en-pentanoate and methylenecyclopropylacetic acid (Eadie et al, Med Toxicol Adverse Drug Exp 3(2):85-106 (1988)).
  • valproate to rats has also been shown to cause enhanced excretion of dicarboxylic acids, a sign of impaired mitochondrial beta-oxidation.
  • Other metabolic effects include hypoglycemia, hyperammoneniia, decreased levels of beta-hydroxybutyrate and carnitine and decreased activities of acyl-CoA dehydrogenases, enzymes involved in fatty acid oxidation.
  • mRNA levels of genes involved in fatty acid oxidation were found to have increased (Kibayasbi et al, Pediatrlnt 41(l):52-60 (1999)).
  • Wy-14643 a tumor-inducing compound that acts in the liver, has been used to study the genetic profile of cells during the various stages of carcinogenic development, with a view toward developing strategies for detecting, diagnosing and treating cancers (Rockett et al, Toxicology 144(l-3):13-29 (2000)). In contrast to other carcinogens, Wy-14643 does not mutate DNA directly, instead, it acts on the peroxisome proliferator activated receptor-alpha (PPARalpha), as well as on other signaling pathways that regulate growth (Johnson et al, J Steroid Biochem Mol Biol 77(1): 59-71 (2001)).
  • PPARalpha peroxisome proliferator activated receptor-alpha
  • the genes and gene expression information, as well as the portfolios and subsets of the genes provided in Tables 1-5XX may be used to predict or identify at least one toxic effect, including the hepatotoxicity of a test or unknown compound.
  • at least one toxic effect includes, but is not limited to, a detrimental change in the physiological status of a cell or organism.
  • the response may be, but is not required to be, associated with a particular pathology, such as tissue necrosis. Accordingly, the toxic effect includes effects at the molecular and cellular level.
  • Hepatotoxicity is an effect as used herein and includes, but is not limited to, genotoxic and non-genotoxic carcinogenesis, cholestasis, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, and steatosis with hepatitis.
  • hepatoxicity includes the effect of direct- acting agents (such as metformin, rosiglitazone and dexamethasone), which are pharmaceuticals that act in the liver, but are not considered toxic to the liver. Exposure to these agents results in altered gene expression profiles.
  • a gene expression profile comprises any quantitative representation of the expression of at least one mRNA species in a cell sample or population and includes profiles made by various methods such as differential display, PCR, hybridization analysis, etc.
  • assays to predict the toxicity or hepatotoxicity of a test agent comprise the steps of exposing a cell population to the test compound, assaying or measuring the level of relative or absolute gene expression of one or more of the genes in Tables 5A-5XX and comparing the identified expression level(s) to the expression levels disclosed in the Tables and database(s) disclosed herein.
  • Assays may include the measurement of the expression levels of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 50, 75, 100, 200, 300, 400, 500, 1000 or more genes from Tables 5A-5XX to create multi-gene expression profiles.
  • all or substantially all of the genes of Tables 5A-5XX may be used to predict toxicity, etc.
  • the genes or subsets of the genes for each individual toxin model for instance, the genes of Table 5 A, may be used.
  • An "adequate amount" of the data of Tables 5A-5XX refers to an amount of information that allows toxicity identification or prediction (typically 2 or more genes).
  • “Substantially” or nearly all of the data in the tables refers to at least about 80% of the data for an individual model.
  • the gene expression level for a gene or genes induced by the test agent, compound or compositions may be comparable to the levels found in the Tables or databases disclosed herein if the expression level varies within a factor of about 2, about 1.5 or about 1.0 fold. In some cases, the expression levels are comparable if the agent induces a change in the expression of a gene in the same direction (e.g., up or down) as a reference toxin. "Comparing" may comprise determining the relationship of the database information to the sample gene expression profile with or without application of an algorithm to the results, differences or similarities between the two.
  • the cell population that is exposed to the test agent, compound or composition may be exposed in vitro or in vivo.
  • cultured or freshly isolated hepatocytes in particular rat hepatocytes, may be exposed to the agent under standard laboratory and cell culture conditions.
  • in vivo exposure may be accomplished by administration of the agent to a living animal, for instance a laboratory rat.
  • Procedures for designing and conducting toxicity tests in in vitro and in vivo systems are well known, and are described in many texts on the subject, such as Loomis et al, Loomis's Esstentials of Toxicology. 4th Ed.. Academic Press, New York, 1996; Echobichon, The Basics of Toxicity Testing. CRC Press, Boca Raton, 1992; Frazier, editor, In Vitro Toxicity Testing. Marcel Dekker, New York, 1992; and the like.
  • test organisms In in vitro toxicity testing, two groups of test organisms are usually employed: One group serves as a control and the other group receives the test compound in a single dose (for acute toxicity tests) or a regimen of doses (for prolonged or chronic toxicity tests). Because, in some cases, the extraction of tissue as called for in the methods of the invention requires sacrificing the test animal, both the control group and the group receiving compound must be large enough to permit removal of animals for sampling tissues, if it is desired to observe the dynamics of gene expression through the duration of an experiment. [0085] In setting up a toxicity study, extensive guidance is provided in the literature for selecting the appropriate test organism for the compound being tested, route of administration, dose ranges, and the like.
  • Water or physiological saline (0.9% NaCl in water) is the solute of choice for the test compound since these solvents permit administration by a variety of routes.
  • vegetable oils such as com oil or organic solvents such as propylene glycol may be used.
  • the volume required to administer a given dose is limited by the size of the animal that is used. It is desirable to keep the volume of each dose uniform within and between groups of animals. When rats or mice are used, the volume administered by the oral route generally should not exceed about 0.005 ml per gram of animal.
  • the routes of administration to the test animal should be the same as, or as similar as possible to, the route of administration of the compound to man for therapeutic pmposes.
  • a compound When a compound is to be administered by inhalation, special techniques for generating test atmospheres are necessary. The methods usually involve aerosolization or nebulization of fluids containing the compound. If the agent to be tested is a fluid that has an appreciable vapor pressure, it may be administered by passing air through the solution under controlled temperature conditions. Under these conditions, dose is estimated from the volume of air inhaled per unit time, the temperature of the solution, and the vapor pressure of the agent involved. Gases are metered from reservoirs. When particles of a solution are to be administered, unless the particle size is less than about 2 ⁇ m the particles will not reach the terminal alveolar sacs in the lungs.
  • the prefened method of administering an agent to animals is via the oral route, either by intubation or by inco ⁇ orating the agent in the feed.
  • the agent is exposed to cells in vitro or in cell culture
  • the cell population to be exposed to the agent may be divided into two or more subpopulations, for instance, by dividing the population into two or more identical aliquots.
  • the cells to be exposed to the agent are derived from liver tissue. For instance, cultured or freshly isolated rat hepatocytes may be used.
  • the methods of the invention may be used generally to predict at least one toxic response, and, as described in the Examples, may be used to predict the likelihood that a compound or test agent will induce various specific liver pathologies, such as genotoxic and non-genotoxic carcinogenesis, cholestasis, direct action toxicity, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, steatosis with hepatitis, or other pathologies associated with at least one of the toxins herein described.
  • the methods of the invention may also be used to determine the similarity of a toxic response to one or more individual compounds.
  • the methods of the invention may be used to predict or elucidate the potential cellular pathways influenced, induced or modulated by the compound or test agent due to the similarity of the expression profile compared to the profile induced by a known toxin (see Tables 5A-5G, 5J, 5K, 5M-5S, 5U-5Y, 5AA-5EE, 5HH-5JJ, 5MM, 5OO, 5PP and 5SS-5XX).
  • Tables 5A-5G, 5J, 5K, 5M-5S, 5U-5Y, 5AA-5EE, 5HH-5JJ, 5MM, 5OO, 5PP and 5SS-5XX the link between a specific liver pathology that is the result of exposure to a toxin and a specific gene expression profile allows for distinction of the genes in Tables 5A-5XX as markers of liver toxicity.
  • the genes and gene expression information or portfolios of the genes with their expression information as provided in Tables 5A-5XX may be used as diagnostic markers for the prediction or identification of the physiological state of tissue or cell sample that has been exposed to a compound or to identify or predict the toxic effects of a compound or agent.
  • a tissue sample such as a sample of peripheral blood cells or some other easily obtainable tissue sample may be assayed by any of the methods described above, and the expression levels from a gene or genes from Tables 5A-5XX may be compared to the expression levels found in tissues or cells exposed to the toxins described herein.
  • the levels of a gene(s) of Tables 5A-5XX, its encoded protein(s), or any metabolite produced by the encoded protein may be monitored or detected in a sample, such as a bodily tissue or fluid sample to identify or diagnose a physiological state of an organism.
  • samples may include any tissue or fluid sample, including urine, blood and easily obtainable cells such as peripheral lymphocytes.
  • the genes and gene expression information provided in Tables 5A-5XX may also be used as markers for the monitoring of toxicity progression, such as that found after initial exposure to a drug, drug candidate, toxin, pollutant, etc.
  • a tissue or cell sample may be assayed by any of the methods described above, and the expression levels from a gene or genes from Tables 5A-5XX may be compared to the expression levels found in tissue or cells exposed to the hepatotoxins described herein.
  • the comparison of the expression data, as well as available sequence or other information may be done by researcher or diagnostician or may be done with the aid of a computer and databases.
  • the genes identified in Tables 5A-5XX may be used as markers or drag targets to evaluate the effects of a candidate drug, chemical compound or other agent on a cell or tissue sample.
  • the genes may also be used as drag targets to screen for agents that modulate their expression and/or activity.
  • a candidate drug or agent can be screened for the ability to simulate the transcription or expression of a given marker or markers or to down-regulate or counteract the transcription or expression of a marker or markers.
  • an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down-regulating expression of the nucleic acid in a cell.
  • Assays to monitor the expression of a marker or markers as defined in Tables 5 A- 5XX may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention.
  • microanays containing probes to one, two or more genes from Tables 5 A-5XX may be used to directly monitor or detect changes in gene expression in the treated or exposed cell.
  • Cell lines, tissues or other samples are first exposed to a test agent and in some instances, a known toxin, and the detected expression levels of one or more, or preferably 2 or more of the genes of Tables 5A-5XX are compared to the expression levels of those same genes exposed to a known toxin alone.
  • Compounds that modulate the expression patterns of the known toxin(s) would be expected to modulate potential toxic physiological effects in vivo.
  • Tables 5 A-5XX are particularly appropriate marks in these assays as they are differentially expressed in cells upon exposure to a known hepatotoxin.
  • cell lines that contain reporter gene fusions between the open reading frame and/or the transcriptional regulatory regions of a gene in Tables 5A-5XX and any assayable fusion partner may be prepared. Numerous assayable fusion partners are known and readily available including the firefly luciferase gene and the gene encoding chloramphenicol acetyltransferase (Alam et al, Anal Biochem 188:245-254 (1990)). Cell lines containing the reporter gene fusions are then exposed to the agent to be tested under appropriate conditions and time. Differential expression of the reporter gene between samples exposed to the agent and control samples identifies agents which modulate the expression of the nucleic acid.
  • Additional assay formats may be used to monitor the ability of the agent to modulate the expression of a gene identified in Tables 5 A-5XX.
  • mRNA expression may be monitored directly by hybridization of probes to the nucleic acids of the invention.
  • Cell lines are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standard procedures such those disclosed in Sambrook et al (Molecular Cloning: A Laboratory Manual. 3d Ed.. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2001).
  • cells or cell lines are first identified which express the gene products of the invention physiologically.
  • Cell and/or cell lines so identified would be expected to comprise the necessary cellular machinery such that the fidelity of modulation of the transcriptional apparatus is maintained with regard to exogenous contact of agent with appropriate surface transduction mechanisms and/or the cytosolic cascades.
  • such cells or cell lines may be transduced or transfected with an expression vehicle (e.g., a plasmid or viral vector) construct comprising an operable non-translated 5 '-promoter containing end of the stractural gene encoding the gene products of Tables 5A-5XX fused to one or more antigenic fragments or other detectable markers, which are peculiar to the instant gene products, wherein said fragments are under the transcriptional control of said promoter and are expressed as polypeptides whose molecular weight can be distinguished from the naturally occurring polypeptides or may further comprise an immunologically distinct or other detectable tag.
  • an expression vehicle e.g., a plasmid or viral vector
  • Cells or cell lines transduced or transfected as outlined above are then contacted with agents under appropriate conditions; for example, the agent comprises a pharmaceutically acceptable excipient and is contacted with cells comprised in an aqueous physiological buffer such as phosphate buffered saline (PBS) at physiological pH, Eagles balanced salt solution (BSS) at physiological pH, PBS or BSS comprising serum or conditioned media comprising PBS or BSS and/or serum incubated at 37°C.
  • PBS phosphate buffered saline
  • BSS Eagles balanced salt solution
  • Said conditions may be modulated as deemed necessary by one of skill in the art.
  • a polypeptide fraction is pooled and contacted with an antibody to be further processed by immunological assay (e.g., ELISA, immunoprecipitation or Western blot).
  • immunological assay e.g., ELISA, immunoprecipitation or Western blot.
  • the pool of proteins isolated from the agent-contacted sample is then compared with the control samples (no exposure and exposure to a known toxin) where only the excipient is contacted with the cells and an increase or decrease in the immunologically generated signal from the agent-contacted sample compared to the contiol is used to distinguish the effectiveness and/or toxic effects of the agent.
  • Another embodiment of the present invention provides methods for identifying agents that modulate at least one activity of a protein(s) encoded by the genes in Tables 5 A- 5XX. Such methods or assays may utilize any means of monitoring or detecting the desired activity.
  • the relative amounts of a protein (Tables 5A-5XX) between a cell population that has been exposed to the agent to be tested compared to an un-exposed control cell population and a cell population exposed to a known toxin may be assayed.
  • probes such as specific antibodies are used to monitor the differential expression of the protein in the different cell populations.
  • Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time.
  • Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe, such as a specific antibody.
  • Agents that are assayed in the above methods can be randomly selected or rationally selected or designed.
  • an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of the a protein of the invention alone or with its associated substiates, binding partners, etc.
  • An example of randomly selected agents is the use a chemical library or a peptide combinatorial library, or a growth broth of an organism.
  • an agent is said to be rationally selected or designed when the agent is chosen on a nonrandom basis which takes into account the sequence of the target site and/or its conformation in connection with the agent's action.
  • Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites.
  • a rationally selected peptide agent can be a peptide whose amino acid sequence is identical to or a derivative of any functional consensus site.
  • the agents of the present invention can be, as examples, peptides, small molecules, vitamin derivatives, as well as carbohydrates. Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be introduced into cells to affect function. "Mimic” used herein refers to the modification of a region or several regions of a peptide molecule to provide a structure chemically different from the parent peptide but topographically and functionally similar to the parent peptide (see G . Grant in: Molecular Biology and Biotechnology. Meyers, ed., pp. 659-664, VCH Publishers, New York, 1995). A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • genes identified as being differentially expressed upon exposure to a known hepatotoxin may be used in a variety of nucleic acid detection assays to detect or quantititate the expression level of a gene or multiple genes in a given sample.
  • the genes described in Tables 5 A-5XX may also be used in combination with one or more additional genes whose differential expression is associate with toxicity in a cell or tissue.
  • the genes in Tables 5A-5XX may be combined with one or more of the genes described in prior and related applications 60/353,171; 60/363,534; 60/371,135; 60/371,134; 60/370,248; 60/371,150; 60/371,413; 60/373,601; 60/374,139; 60/394,253; 60/378,652; 60/373,602; 60/378,653; 60/378,665; 60/378,370; 60/394,230; 60/407,688;
  • Any assay format to detect gene expression may be used. For example, traditional Northern blotting, dot or slot blot, nuclease protection, primer directed amplification, RT- PCR, semi- or quantitative PCR, branched-chain DNA and differential display methods may be used for detecting gene expression levels. Those methods are useful for some embodiments of the invention. In cases where smaller numbers of genes are detected, amplification based assays may be most efficient. Methods and assays of the invention, however, may be most efficiently designed with hybridization-based methods for detecting the expression of a large number of genes.
  • Any hybridization assay format may be used, including solution-based and solid support-based assay formats.
  • Solid supports containing oligonucleotide probes for differentially expressed genes of the invention can be filters, polyvinyl chloride dishes, particles, beads, microparticles or silicon or glass based chips, etc. Such chips, wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755).
  • a prefened solid support is a high density anay or DNA chip. These contain a particular oligonucleotide probe in a predetermined location on the anay. Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence. Such predetermined locations are termed features. There may be, for example, from 2, 10, 100, 1000 to 10,000, 100,000 or 400,000 or more of such features on a single solid support. The solid support, or the area within which the probes are attached may be on the order of about a square centimeter. Probes conesponding to the genes of Tables 5 A-5XX or from the related applications described above may be attached to single or multiple solid support structures, e.g., the probes may be attached to a single chip or to multiple chips to comprise a chip set.
  • Oligonucleotide probe anays for expression monitoring can be made and used according to any techniques known in the art (see for example, Lockhart et al., Nat Biotechnol 14:1675-1680 (1996); McGall et al, Proc Nat Acad Sci USA 93:13555-13460 (1996)).
  • Such probe arrays may contain at least two or more oligonucleotides that are complementary to or hybridize to two or more of the genes described in Tables 5A-5XX.
  • such anays may contain oligonucleotides that are complementary or hybridize to at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 70, 100 or more the genes described herein.
  • Prefened anays contain all or nearly all of the genes listed in Tables 5A-5XX, 'or individually, the gene sets of Tables 5A-5XX.
  • anays are constructed that contain oligonucleotides to detect all or nearly all of the genes in any one of or all of Tables 5A-5XX on a single solid support substrate, such as a chip.
  • the sequences of the expression marker genes of Tables 5A-5XX are in the public databases. Table 1 provides the GenBank Accession Number, SEQ ID NO: and GLGC ID No.
  • Table 2 provides identification information for the human homologues of the genes of Tables 1 and 5A-5XX.
  • Table 3 identifies the metabolic pathways in which the genes of Tables 1 and 5A-5XX are believed to function.
  • Table 4 defines the model codes used in Tables 1, 2, 3 and 5A-5XX.
  • sequences of the genes in GenBank are expressly herein inco ⁇ orated by reference in their entirety as of the filing date of this application, as are related sequences, for instance, sequences from the same gene of different lengths, variant sequences, polymo ⁇ hic sequences, genomic sequences of the genes and related sequences from different species, including the human counte ⁇ arts, where appropriate. These sequences may be used in the methods of the invention or may be used to produce the probes and anays of the invention. In some embodiments, the genes in Tables 5 A-5XX that conespond to the genes or fragments previously associated with a toxic response may be excluded from the Tables. [00111] As described above, in addition to the sequences of the GenBank Accession Nos.
  • sequences such as naturally occurring variant or polymo ⁇ hic sequences may be used in the methods and compositions of the invention.
  • expression levels of various allelic or homologous forms of a gene disclosed in the Tables 5A-5XX may be assayed.
  • Any and all nucleotide variations that do not alter the functional activity of a gene listed in the Tables 5 A-5XX including all naturally occurring allelic variants of the genes herein disclosed, may be used in the methods and to make the compositions (e.g., anays) of the invention.
  • Probes based on the sequences of the genes described above may be prepared by any commonly available method. Oligonucleotide probes for screening or assaying a tissue or cell sample are preferably of sufficient length to specifically hybridize only to appropriate, complementary genes or transcripts. Typically the oligonucleotide probes will be at least about 10, 12, 14, 16, 18, 20 or 25 nucleotides in length. In some cases, longer probes of at least 30, 40, or 50 nucleotides will be desirable.
  • oligonucleotide sequences that are complementary to one or more of the genes described in Tables 5A-5XX refer to oligonucleotides that are capable of hybridizing under stringent conditions to at least part of the nucleotide sequences of said genes. Such hybridizable oligonucleotides will typically exhibit at least about 75% sequence identity at the nucleotide level to said genes, preferably about 80% or 85% sequence identity or more preferably about 90% or 95% or more sequence identity to said genes.
  • Bind(s) substantially refers to complementary hybridization between a probe nucleic acid and a target nucleic acid and embraces minor mismatches that can be accommodated by reducing the stringency of the hybridization media to achieve the desired detection of the target polynucleotide sequence.
  • background or “background signal intensity” refer to hybridization signals resulting from non-specific binding, or other interactions, between the labeled target nucleic acids and components of the oligonucleotide anay (e.g., the oligonucleotide probes, control probes, the anay substrate, etc.). Background signals may also be produced by intrinsic fluorescence of the anay components themselves. A single background signal can be calculated for the entire anay, or a different background signal may be calculated for each target nucleic acid.
  • background is calculated as the average hybridization signal intensity for the lowest 5% to 10%) of the probes in the anay, or, where a different background signal is calculated for each target gene, for the lowest 5% to 10% of the probes for each gene.
  • background may be calculated as the average hybridization signal intensity produced by hybridization to probes that are not complementary to any sequence found in the sample (e.g. probes directed to nucleic acids of the opposite sense or to genes not found in the sample such as bacterial genes where the sample is mammalian nucleic acids). Background can also be calculated as the average signal intensity produced by regions of the anay that lack any probes at all.
  • hybridizing specifically to refers to the binding, duplexing, or hybridizing of a molecule substantially to or only to a particular nucleotide sequence or sequences under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.
  • Assays and methods of the invention may utilize available formats to simultaneously screen at least about 100, preferably about 1000, more preferably about 10,000 and most preferably about 1,000,000 different nucleic acid hybridizations.
  • a "probe” is defined as a nucleic acid, capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, usually through complementary base pairing, usually through hydrogen bond formation.
  • a probe may include natural (t.e., A, G, U, C, or T) or modified bases (7- deazaguanosine, inosine, etc.).
  • probes may be joined by a linkage other than a phosphodiester bond, so long as it does not interfere with hybridization.
  • probes may be peptide nucleic acids in which the constituent bases are joined by peptide bonds rather than phosphodiester linkages.
  • the term "perfect match probe” refers to a probe that has a sequence that is perfectly complementary to a particular target sequence.
  • the test probe is typically perfectly complementary to a portion (subsequence) of the target sequence.
  • the perfect match (PM) probe can be a "test probe”, a "normalization, control” probe, an expression level control probe and the like.
  • a perfect match control or perfect match probe is, however, distinguished from a “mismatch control” or “mismatch probe.”
  • mismatch control or “mismatch probe” refer to a probe whose sequence is deliberately selected not to be perfectly complementary to a particular target sequence.
  • MM mismatch
  • PM conesponding perfect match
  • the mismatch may comprise one or more bases.
  • mismatch(s) may be located anywhere in the mismatch probe, terminal mismatches are less desirable as a terminal mismatch is less likely to prevent hybridization of the target sequence.
  • the mismatch is located at or near the center of the probe such that the mismatch is most likely to destabilize the duplex with the target sequence under the test hybridization conditions.
  • stringent conditions refers to conditions under which a probe will hybridize to its target subsequence, but with only insubstantial hybridization to other sequences or to other sequences such that the difference may be identified. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. Generally, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • stringent conditions will be those in which the salt concentration is at least about 0.01 to 1.0 M Na + ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes (e.g., 10 to 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • the "percentage of sequence identity” or “sequence identity” is determined by comparing two optimally aligned sequences or subsequences over a comparison window or span, wherein the portion of the polynucleotide sequence in the comparison window may optionally comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical submit (e.g. nucleic acid base or amino acid residue) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • Percentage sequence identity when calculated using the programs GAP or BESTFIT (see below) is calculated using default gap weights.
  • the high density anay will typically include a number of test probes that specifically hybridize to the sequences of interest. Probes may be produced from any region of the genes identified in the Tables and the attached representative sequence listing. In instances where the gene reference in the Tables is an EST, probes may be designed from that sequence or from other regions of the conesponding full-length transcript that may be available in any of the sequence databases, such as those herein described. See WO 99/32660 for methods of producing probes for a given gene or genes.
  • any available software may be used to produce specific probe sequences, including, for instance, software available from Molecular Biology Insights, Olympus Optical Co. and Biosoft International.
  • the anay will also include one or more control probes.
  • Test probes may be oligonucleotides that range from about 5 to about 500, or about 7 to about 50 nucleotides, more preferably from about 10 to about 40 nucleotides and most preferably from about 15 to about 35 nucleotides in length. In other particularly prefened embodiments, the probes are 20 or 25 nucleotides in length. In another prefened embodiment, test probes are double or single strand DNA sequences. DNA sequences are isolated or cloned from natural sources or amplified from natural sources using native nucleic acid as templates. These probes have sequences complementary to particular subsequences of the genes whose expression they are designed to detect. Thus, the test probes are capable of specifically hybridizing to the target nucleic acid they are to detect.
  • control probes In addition to test probes that bind the target nucleic acid(s) of interest, the high density anay can contain a number of control probes.
  • the control probes may fall into three categories refened to herein as 1) normalization controls; 2) expression level controls; and 3) mismatch controls.
  • Nonnalization controls are oligonucleotide or other nucleic acid probes that are complementary to labeled reference oligonucleotides or other nucleic acid sequences that are added to the nucleic acid sample to be screened.
  • the signals obtained from the normalization controls after hybridization provide a contiol for variations in hybridization conditions, label intensity, "reading" efficiency and other factors that may cause the signal of a perfect hybridization to vary between anays.
  • signals e.g., fluorescence intensity
  • read from all other probes in the anay are divided by the signal (e.g., fluorescence intensity) from the control probes thereby normalizing the measurements.
  • any probe may serve as a normalization control.
  • Prefened normalization probes are selected to reflect the average length of the other probes present in the anay, however, they can be selected to cover a range of lengths.
  • the normalization control(s) can also be selected to reflect the (average) base composition of the other probes in the anay, however in a prefened embodiment, only one or a few probes are used and they are selected such that they hybridize well (i.e., no secondary structure) and do not match any target-specific probes.
  • Expression level controls are probes that hybridize specifically with constitutively expressed genes in the biological sample. Virtually any constitutively expressed gene provides a suitable target for expression level controls. Typically expression level control probes have sequences complementary to subsequences of constitutively expressed "housekeeping genes" including, but not limited to the ⁇ -actin gene, the glyceraldehyde-3- phosphate dehydrogenase (GADPH) gene, the transferrin receptor gene and the like. [00131] Mismatch controls may also be provided for the probes to the target genes, for expression level controls or for normalization controls.
  • Mismatch controls are oligonucleotide probes or other nucleic acid probes identical to their conesponding test or control probes except for the presence of one or more mismatched bases.
  • a mismatched base is a base selected so that it is not complementary to the conesponding base in the target sequence to which the probe would otherwise specifically hybridize.
  • One or more mismatches are selected such that under appropriate hybridization conditions (e.g., stringent conditions) the test or control probe would be expected to hybridize with its target sequence, but the mismatch probe would not hybridize (or would hybridize to a significantly lesser extent) Prefened mismatch probes contain a central mismatch.
  • a conesponding mismatch probe will have the identical sequence except for a single base mismatch (e.g., substituting a G, a C or a T for an A) at any of positions 6 through 14 (the central mismatch).
  • Mismatch probes thus provide a control for non-specific binding or cross hybridization to a nucleic acid in the sample other than the target to which the probe is directed. For example, if the target is present the perfect match probes should be consistently brighter than the mismatch probes. In addition, if all central mismatches are present, the mismatch probes can be used to detect a mutation, for instance, a mutation of a gene in the accompanying Tables 5A-5XX. The difference in intensity between the perfect match and the mismatch probe provides a good measure of the concentration of the hybridized material.
  • Cell or tissue samples may be exposed to the test agent in vitro or in vivo.
  • appropriate mammalian liver extracts may also be added with the test agent to evaluate agents that may require biotransformation to exhibit toxicity.
  • primary isolates of animal or human hepatocytes which already express the appropriate complement of drag-metabolizing enzymes may be exposed to the test agent without the addition of mammalian liver extracts.
  • the genes which are assayed according to the present invention are typically in the form of mRNA or reverse transcribed mRNA.
  • the genes may be cloned or not.
  • the genes may be amplified or not. The cloning and/or amplification do not appear to bias the representation of genes within a population. In some assays, it may be preferable, however, to use polyA+ RNA as a source, as it can be used with less processing steps.
  • nucleic acid samples used in the methods and assays of the invention may be prepared by any available method or process. Methods of isolating total mRNA are well known to those of skill in the art.
  • RNA samples include RNA samples, but also include cDNA synthesized from a mRNA sample isolated from a cell or tissue of interest. Such samples also include DNA amplified from the cDNA, and RNA transcribed from the amplified DNA.
  • Biological samples may be of any biological tissue or fluid or cells from any organism as well as cells raised in vitro, such as cell lines and tissue culture cells. Frequently the sample will be a tissue or cell sample that has been exposed to a compound, agent, drag, pharmaceutical composition, potential environmental pollutant or other composition. In some formats, the sample will be a "clinical sample” which is a sample derived from a patient. Typical clinical samples include, but are not limited to, sputum, blood, blood-cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and pleural fluid, or cells therefrom.
  • Bio samples may also include sections of tissues, such as frozen sections or formalin fixed sections taken for histological pmposes.
  • oligonucleotide analogue anay can be synthesized on a single or on multiple solid substiates by a variety of methods, including, but not limited to, light-directed chemical coupling, and mechanically directed coupling (see Pirrung, U.S. Patent No. 5,143,854).
  • a glass surface is derivatized with a silane reagent containing a functional group, e.g., a hydroxyl or amine group blocked by a photolabile protecting group.
  • a functional group e.g., a hydroxyl or amine group blocked by a photolabile protecting group.
  • Photolysis through a photolithogaphic mask is used selectively to expose functional groups which are then ready to react with incoming 5' photoprotected nucleoside phosphoramidites.
  • the phosphoramidites react only with those sites which are illuminated (and thus exposed by removal of the photolabile blocking group).
  • the phosphoramidites only add to those areas selectively exposed from the preceding step. These steps are repeated until the desired anay of sequences have been synthesized on the solid surface. Combinatorial synthesis of different oligonucleotide analogues at different locations on the anay is determined by the pattern of illumination during synthesis and the order of addition of coupling reagents.
  • High density nucleic acid anays can also be fabricated by depositing pre-made or natural nucleic acids in predetermined positions. Synthesized or natural nucleic acids are deposited on specific locations of a substrate by light directed targeting and oligonucleotide directed targeting. Another embodiment uses a dispenser that moves from region to region to deposit nucleic acids in specific spots.
  • Nucleic acid hybridization simply involves contacting a probe and target nucleic acid under conditions where the probe and its complementary target can form stable hybrid duplexes through complementary base pairing. See WO 99/32660. The nucleic acids that do not form hybrid duplexes are then washed away leaving the hybridized nucleic acids to be detected, typically through detection of an attached detectable label. It is generally recognized that nucleic acids are denatured by increasing the temperature or decreasing the salt concentration of the buffer containing the nucleic acids. Under low stringency conditions (e.g., low temperature and/or high salt) hybrid duplexes (e.g., DNA:DNA, RNA:RNA, or RNA:DNA) will form even where the annealed sequences are not perfectly complementary.
  • low stringency conditions e.g., low temperature and/or high salt
  • hybridization conditions may be selected to provide any degree of stringency.
  • hybridization is performed at low stringency, in this case in 6X SSPET at 37°C (0.005% Triton X-100), to ensure hybridization and then subsequent washes are performed at higher stringency (e.g., I X SSPET at 37°C) to eliminate mismatched hybrid duplexes. Successive washes may be performed at increasingly higher stringency (e.g., down to as low as 0.25 X SSPET at 37°C to 50°C) until a desired level of hybridization specificity is obtained. Stringency can also be increased by addition of agents such as formamide.
  • Hybridization specificity may be evaluated by comparison of hybridization to the test probes with hybridization to the various controls that can be present (e.g., expression level control, normalization control, mismatch controls, etc.).
  • hybridization specificity stringency
  • signal intensity there is a tradeoff between hybridization specificity (stringency) and signal intensity.
  • the wash is performed at the highest stringency that produces consistent results and that provides a signal intensity greater than approximately 10% of the background intensity.
  • the hybridized array may be washed at successively higher stringency solutions and read between each wash. Analysis of the data sets thus produced will reveal a wash stringency above which the hybridization pattern is not appreciably altered and which provides adequate signal for the particular oligonucleotide probes of interest.
  • the hybridized nucleic acids are typically detected by detecting one or more labels attached to the sample nucleic acids.
  • the labels may be inco ⁇ orated by any of a number of means well known to those of skill in the art. See WO 99/32660.
  • the present invention includes relational databases containing sequence information, for instance, for the genes of Tables 5A-5XX, as well as gene expression information from tissue or cells exposed to various standard toxins, such as those herein described (see Tables 5A-5XX).
  • Databases may also contain information associated with a given sequence or tissue sample such as descriptive information about the gene associated with the sequence information (see Tables 1, 2 and 3), or descriptive information concerning the clinical status of the tissue sample, or the animal from which the sample was derived.
  • the database may be designed to include different parts, for instance a sequence database and a gene expression database. Methods for the configuration and construction of such databases and computer-readable media to which such databases are saved are widely available, for instance, see U.S. Patent No. 5,953,727, which is herein inco ⁇ orated by reference in its entirety.
  • GenBank www.ncbi.nlm.nih.gov/entrez.index.html
  • KEGG www.genome.ad.jp/kegg
  • SPAD www.grt.kyushu-u.ac.jp/spad/index.html
  • HUGO www.gene.uclac.uk/hugo
  • Swiss- Prot www.expasy.ch.sprot
  • Prosite www.expasy.ch/tools/scnpsitl.html
  • OMDM www.ncbi.nlm.nih.gov/omim
  • LocusLink www.ncbi.nlm.nih.gov/LocusLink/
  • RefSeq www.ncbi.nlm.nih.gov/LocusLink/refseq.html
  • GDB www.gdb.org
  • the external database is GenBank and the associated databases maintained by the National Center for Biotechnology Information (NCBI) (wivw. ncbi. nlm. nih.gov) .
  • NCBI National Center for Biotechnology Information
  • Any appropriate computer platform, user interface, etc. may be used to perform the necessary comparisons between sequence information, gene expression information and any other information in the database or information provided as an input.
  • a large number of computer workstations are available from a variety of manufacturers, such has those available from Silicon Graphics.
  • Client/server environments, database servers and networks are also widely available and appropriate platforms for the databases of the invention.
  • the databases of the invention may be used to produce, among other things, electronic Northerns that allow the user to determine the cell type or tissue in which a given gene is expressed and to allow determination of the abundance or expression level of a given gene in a particular tissue or cell.
  • the databases of the invention may also be used to present information identifying the expression level in a tissue or cell of a set of genes comprising one or more of the genes in Tables 5A-5XX, comprising the step of comparing the expression level of at least one gene in Tables 5A-5XX in a cell or tissue exposed to a test agent to the level of expression of the gene in the database.
  • Such methods may be used to predict the toxic potential of a given compound by comparing the level of expression of a gene or genes in Tables 5A-5XX from a tissue or cell sample exposed to the test agent to the expression levels found in a control tissue or cell samples exposed to a standard toxin or hepatotoxin such as those herein described.
  • Such methods may also be used in the drug or agent screening assays as described herein.
  • the invention further includes kits combining, in different combinations, high- density oligonucleotide arrays, reagents for use with the anays, protein reagents encoded by the genes of the Tables, signal detection and anay-processing instruments, gene expression databases and analysis and database management software described above.
  • the kits may be used, for example, to predict or model the toxic response of a test compound, to monitor the progression of hepatic disease states, to identify genes that show promise as new drug targets and to screen known and newly designed drags as discussed above.
  • the databases packaged with the kits are a compilation of expression patterns from human or laboratory animal genes and gene fragments (conesponding to the genes of Tables 5A-5XX).
  • the database software and packaged information that may contain the databases saved to a computer-readable medium include the expression results of Tables 5A-5XX that can be used to predict toxicity of a test agent by comparing the expression levels of the genes of Tables 5A-5XX induced by the test agent to the expression levels presented in Tables 5A-5XX.
  • database and software information may be provided in a remote electronic format, such as a website, the address of which may be packaged in the kit.
  • kits may be used in the pharmaceutical industry, where the need for early drug testing is strong due to the high costs associated with drug development, but where bioinformatics, in particular gene expression informatics, is still lacking. These kits will reduce the costs, time and risks associated with traditional new drag screening using cell cultures and laboratory animals. The results of large-scale drug screening of pre-grouped patient populations, pharmacogenomics testing, can also be applied to select drugs with greater efficacy and fewer side-effects. The kits may also be used by smaller biotechnology companies and research institutes who do not have the facilities for performing such large- scale testing themselves.
  • the hepatotoxins alpha-naphthylisothiocyante (ANIT), acetaminophen (APAP), AY-25329, carbon tetrachloride, clofibrate, diclofenac, 17 ⁇ - ethinylestradiol, hydrazine, indomethacin, lipopolysaccharide, lovastatin, methotrexate, tacrine, valproate and control compositions were administered to cultures of primary rat hepatocytes from male Sprague-Dawley rats at various time points using administration diluents, protocols and dosing regimes as previously described in the art and in the prior applications discussed above, as well as in Table 6.
  • administration diluents, protocols and dosing regimes as previously described in the art and in the prior applications discussed above, as well as in Table 6.
  • the source of the primary rat hepatocytes was Sprague Dawley Outbred CD® Rats (CRL:CD®[SD] IGS BR, Charles River Laboratories). Hepatocyte cultures were obtained in 24-well matrigel coated plates for the AlamarBlue® assay (175,000 cells/cm 2 ) or in T-75cm 2 matrigel coated flasks for RNA isolation for microanay analysis (187,000 cells/cm 2 ) . Primary rat hepatocytes were received the day after the cells were removed from the animals. After arrival, the cells, the cells were incubated overnight ( ⁇ 15hrs) before the toxin was added to the cultures.
  • the vehicle used in the toxicity experiments was HIM culture medium (Hepatocyte Incubation Medium, In Vitro Technologies Cat. No. Z90009) containing 0.2% DMSO (Sigma Cat. No. D-5879). Toxin or vehicle was administered to hepatocyte cultures as follows. For each treatment, i.e., vehicle alone, vehicle + toxin at low dose, or vehicle + toxin at high dose, cells were harvested after 3, 6 and 24-hour incubations with the toxin solution or with the vehicle.
  • HIM culture medium Hepatocyte Incubation Medium, In Vitro Technologies Cat. No. Z90009
  • DMSO Sigma Cat. No. D-5879
  • AlamarBlue® assay was performed as follows, using only the 24-hour time point samples.
  • HIM culture medium
  • the cells in all wells were incubated for 24 hours at 37°C, 5% CO 2 .
  • HDM medium was removed, and a solution containing 500 ⁇ l of fresh HIM medium + 50 ⁇ l AlamarBlue® (BioSource International, Inc., Cat. No. DAL1100) was added to each well.
  • the cells were incubated at 37°C, 5% CO 2 for 2 hours.
  • the data were evaluated to determine whether or not the toxin reduced cell viability. If so, the dose of the toxin that reduced cell viability by ⁇ 10-20%) was determined.
  • the medium from the flasks was discarded, and the cells were washed once with 20 ml of warm (37°C) RPMI-1640 + lOmM HEPES medium (Life Technologies, Cat. No. 22400-089).
  • 12 ml of Trizol (Life Technologies, Cat. No. 15596-018) was placed immediately into each T-75 flask. Each flask contained -10-20 million cells.
  • the contents of each flask were mixed vigorously for one minute with a vortex mixer and then aspirated up and down 5 times with a pipette.
  • the contents of each flask ( ⁇ 12 ml each) was collected into a 50 ml conical polypropylene tissue culture tube (Falcon), snap frozen in liquid nitrogen and stored at ⁇ -86° C.
  • Falcon conical polypropylene tissue culture tube
  • RNA sample preparation was conducted with minor modifications, following the protocols set forth in the Affymetrix GeneChip® Expression Analysis Manual. Frozen cells were ground to a powder using a Spex Certiprep 6800 Freezer Mill. Total RNA was extracted with Trizol (GibcoBRL) utilizing the manufacturer's protocol. The total RNA yield for each sample was 200-500 ⁇ g per 300 mg cells. mRNA was isolated using the Oligotex mRNA Midi kit (Qiagen) followed by ethanol precipitation. Double stranded cDNA was generated from mRNA using the Superscript Choice system (GibcoBRL). First strand cDNA synthesis was primed with a T7-(dT24) oligonucleotide.
  • cDNA was phenol- chloroform extracted and ethanol precipitated to a final concentration of 1 ⁇ g/ml.
  • cRNA was synthesized using Ambion' s T7 MegaScript in vitro Transcription Kit. [00161] To biotin label the cRNA, nucleotides Bio-11-CTP and Bio-16-UTP (Enzo Diagnostics) were added to the reaction. Following a 37°C incubation for six hours, impurities were removed from the labeled cRNA following the RNeasy Mini kit protocol (Qiagen).
  • cRNA was fragmented (fragmentation buffer consisting of 200 mM Tris-acetate, pH 8.1, 500 mM KOAc, 150 mM MgOAc) for thirty-five minutes at 94°C. Following the Affymetrix protocol, 55 ⁇ g of fragmented cRNA was hybridized on the Affymetrix rat anay set for twenty- four hours at 60 ⁇ m in a 45 °C hybridization oven. The chips were washed and stained with Streptavidin Phycoerythrin (SAPE) (Molecular Probes) in Affymetrix fluidics stations.
  • SAPE Streptavidin Phycoerythrin
  • SAPE solution was added twice with an anti-streptavidin biotinylated antibody (Vector Laboratories) staining step in between.
  • Hybridization to the probe arrays was detected by fluorometric scanning (Hewlett Packard Gene Anay Scanner). Data was analyzed using Affymetrix GeneChip® version 3.0 and Expression Data Mining Tool (EDMT) software (version 1.0), S-Plus, and the GeneExpress® software system.
  • EDMT Expression Data Mining Tool
  • Table 1 discloses those genes that are differentially expressed upon exposure to the named toxins with their conesponding SEQ ID NOS:, GenBank Accession or RefSeq ID Nos., GLGC ID Nos. (internal Gene Logic identification nos.), gene names and Unigene Sequence Cluster titles.
  • the metabolic pathways in which the genes of Table 1 function are indicated in Table 3, and the conesponding human homologues are given in Table 2.
  • the model codes, identified in Table 4 represent the various toxicity or liver pathology states associated with differential expression of each gene, as well as the individual toxin types associated with differential expression of each gene.
  • Tables 5A-5XX disclose the summary statistics for each of the comparisons performed. Each of these tables contains a set of predictive genes and creates a model for predicting the hepatoxicity of an unknown, i.e., untested compound. Each gene is identified by its Gene Logic identification number and can be cross-referenced to a gene name and representative SEQ LD NO. in Table 1.
  • the group mean for Tox samples is the mean signal intensity, as normalized for the various chip parameters that are being assayed.
  • the Non-tox mean represents the mean signal intensity, as normalized for the various chip parameters that are being assayed, in samples other than those tieated with the high dose of the specific toxin.
  • Tox samples were obtained from treated cells processed at the timepoint(s) indicated in the tables, while Non-tox samples were obtained from control cells processed at all time points in the experiments.
  • an increase in the Tox group mean compared to the Non-tox group mean indicates up-regulation upon exposure to a toxin.
  • a decrease in the Tox group mean compared to the Non-tox group mean indicates down-regulation.
  • the mean values are derived from Average Difference (AveDiff) values for a particular gene, averaged across the conesponding samples. Each individual Average Difference value is calculated by integrating the intensity information from multiple probe pairs that are tiled for a particular fragment.
  • the normalization multiplies each expression intensity for a given experiment (chip) by a global scaling factor. The intent of this normalization is to make comparisons of individual genes between chips possible.
  • the scaling factor is calculated as follows:
  • the value of 100 used here is the standard target value used.
  • Some AveDiff values may be negative due to the general noise involved in nucleic acid hybridization experiments. Although many conclusions can be made conesponding to a negative value on the GeneChip platform, it is difficult to assess the meaning behind the negative value for individual fragments. Our observations show that, although negative values are observed at times within the predictive gene set, these values reflect a real biological phenomenon that is highly reproducible across all the samples from which the measurement was taken. For this reason, those genes that exhibit a negative value are included in the predictive set. It should be noted that other platforms of gene expression measurement may be able to resolve the negative numbers for the conesponding genes. The predictive ability of each of those genes should extend across platforms, however. Each mean value is accompanied by the standard deviation for the mean.
  • the linear discriminant analysis score (discriminant score), as disclosed in the tables, measures the ability of each gene to predict whether or not a sample is toxic.
  • the discriminant score is calculated by the following steps:
  • the number of conect predictions is then the number of Yi's such that f(Yi)>.5 plus the number of X;'s such that f(Xj) ⁇ .5.
  • the discriminant score is then P/(n+t).
  • Linear discriminant analysis uses both the individual measurements of each gene and the calculated measurements of all combinations of genes to classify samples. For each gene, a weight is derived from the mean and standard deviation of the Tox and Non-tox sample groups. Every gene is multiplied by a weight and the sum of these values results in a collective discriminate score. This discriminant score is then compared against collective centioids of the Tox and Non-tox groups. These centroids are the average of all tox and nontox samples respectively. Therefore, each gene contributes to the overall prediction. This contribution is dependent on weights that are large positive or negative numbers if the relative distances between the Tox and Non-tox samples for that gene are large and small numbers if the relative distances are small. The discriminant score for each unknown sample and centroid values can be used to calculate a probability between zero and one as to the group in which the unknown sample belongs.
  • Samples were selected for grouping into Tox and Non-tox groups by examining each study individually with Principal Components Analysis (PCA) to determine which treatments had an observable response. Only sample groups where confidence of the tox- responding or non-tox-responding status (expression level affected by exposure to a specific toxin or expression level not affected by exposure to a specific toxin, respectively) was established were included in building a general toxicity prediction model.
  • Linear discriminant models were generated to describe Tox and Non-tox samples. The top discriminant genes and/or EST's were used to determine toxicity by calculating each gene's contribution with homo and heteroscedastic treatment of variance and inclusion or exclusion of mutual information between genes.
  • the above modeling methods provide broad approaches of combining the expression of genes to predict sample toxicity.
  • the spread of the group distribution and discriminate score alone provide enough information to enable a skilled person to generate all of the above types of models with accuracy that can exceed the discriminate ability of individual genes.
  • Some examples of methods that could be used individually or in combination after transformation of data types include but are not limited to: Discriminant Analysis, Multiple Discriminant Analysis, logistic regression, multiple regression analysis, linear regression analysis, conjoint analysis, canonical conelation, hierarchical cluster analysis, k-means cluster analysis, self-organizing maps, multidimensional scaling, structural equation modeling, support vector machine determined boundaries, factor analysis, neural networks, bayesian classifications, and resampling methods.
  • Example 4 Grouping of Individual compound and Pathology Classes [00180] Samples were grouped into individual pathology classes based on known toxicological responses and observed clinical chemical and pathology measurements or into observable toxicity produced by a compound (Tables 5A-5XX). The top 10, 25, 50, 100 genes based on individual discriminate scores were used in a model to ensure that a combination of genes provided a better prediction than individual genes. As described above, all combinations of two or more genes from this list could potentially provide better prediction than individual genes when selected in any order or by ordered, agglomerate, divisive, or random approaches. In addition, combining these genes with other genes could provide better predictive ability, but most of this predictive ability would come from the genes listed herein.
  • a sample may be considered a Tox sample if it scores positive in any pathological or individual compound class represented here, or in any modeling method mentioned under general toxicology models, based on a combination of the sample's time point and dosage group in a study using an individual compound (with known or potentially toxic properties) by comparisons obtainable from the data.
  • the pathological groupings and early and late phase models are prefened examples of all obtainable combinations of sample time and dose points. Most logical groupings with one or more genes and one or more sample dose and time points should produce better predictions of general toxicity, pathological specific toxicity, or similarity to a known toxin than individual genes.
  • Lipopolysaccharide lOug/ml lOOug/ml lOug/ml lOOug/ml lOug/ml lOOug/ml lOug/ml lOOug/ml
  • Tacrine 25uM 250uM 25uM 250uM 25uM 250uM 25uM 250uM
  • HTM cell culture medium In Vitro Technologies
  • DMSO Sigma cat. no. D-5879
  • AlamarBlue assay was performed only at the 24-hr time point following exposure to the toxin of interest.
  • a corresponding vehicle control (0.2% DMSO) sample was also isolated at 3, 6, and 24-hr time points for each toxin.

Abstract

The present invention is based on the elucidation of the global changes in gene expression and the identification of toxicity markers in tissues or cells exposed to a known toxin. The genes may be used as toxicity markers in drug screening and toxicity assays. The invention includes a database of genes characterized by toxin-induced differential expression that is designed for use with microarrays and other solid-phase probes.

Description

PRIMARY RAT HEPATOCYTE TOXICITY MODELING
INVENTORS: Donna MENDRICK, Mark PORTER, Kory JOHNSON, Brandon HIGGS, Arthur CASTLE, Michael ORR and Michael ELASHOFF
RELATED APPLICATIONS
[0001] This application claims priority under 35 U.S.C. §119(e) to U.S. Provisional
Applications 60/353,171, filed February 4, 2002; 60/363,534, filed March 13, 2002; 60/371,135, filed April 10, 2002; 60/371,134, filed April 10, 2002; 60/370,248, filed April 8, 2002; 60/371,150, filed April 10, 2002; 60/371,413, filed April 11, 2002; 60/373,601, filed April 19, 2002; 60/374,139, filed April 22, 2002; 60/394,253, filed July 9, 2002; 60/378,652, filed May 9, 2002; 60/373,602, filed April 19, 2002; 60/378,653, filed May 9, 2002; 60/378,665, filed May 9, 2002; 60/378,370, filed May 8, 2002; 60/394,230, filed My 9, 2002; and 60/407,688, filed September 4, 2002, all of which are herein incoφorated by reference in their entirety. [0002] This application is also related to pending U.S. Applications 09/917,800, filed July
31, 2001, 10/060,087, filed January 31, 2002, and PCT/US03/ , entitled "Molecular
Hepatotoxicology Modeling," filed January 31, 2003, as well as to PCT Application PCT/US01/23872, filed July 31, 2001, all of which are herein incoφorated by reference in their entirety.
SEQUENCE LISTING SUBMISSION ON COMPACT DISC
[0003] The Sequence Listing submitted concunently herewith on compact disc under 37
C.F.R. §§1.821(c) and 1.821(e) is herein incoφorated by reference in its entirety. Four copies of the Sequence Listing, one on each of four compact discs are provided. Copy 1, Copy 2 and Copy 3 are identical. Copies 1, 2 and 3 are also identical to the CRF. Each electronic copy of the Sequence Listing was created on February 3, 2003 with a file size of 6321 KB. The file names are as follows: Copy 1- gl5113wo.txt; Copy 2- gl5113wo.txt; Copy 3- gl5113wo.txt; CRF- gl5113wo.txt.
BACKGROUND OF THE INVENTION
[0004] The need for methods of assessing the toxic impact of a compound, pharmaceutical agent or environmental pollutant on a cell or living organism has led to the development of procedures which utilize living organisms as biological monitors. The simplest and most convenient of these systems utilize unicellular microorganisms such as yeast and bacteria, since they are most easily maintained and manipulated. Unicellular screening systems also often use easily detectable changes in phenotype to monitor the effect of test compounds on the cell. Unicellular organisms, however, are inadequate models for estimating the potential effects of many compounds on complex multicellular animals, as they do not have the ability to carry out biotransformations to the extent or at levels found in higher organisms. [0005] The biotransformation of chemical compounds by multicellular organisms is a significant factor in determining the overall toxicity of agents to which they are exposed. Accordingly, multicellular screening systems or screening systems using isolated eukaryotic cells may be prefened or required to detect the toxic effects of compounds. The use of multicellular organisms as toxicology screening tools has been significantly hampered, however, by the lack of convenient screening mechanisms or endpoints, such as those available in yeast or bacterial systems. In addition, previous attempts to produce toxicology prediction systems have failed to provide the necessary modeling data and statistical information to accurately predict toxic responses (e.g., WO 00/12760, WO 00/47761, WO 00/63435, WO 01/32928, WO 01/38579).
SUMMARY OF THE INVENTION
[0006] The present invention is based on the elucidation of the global changes in gene expression in primary hepatocytes exposed to known toxins, in particular hepatotoxins, as compared to unexposed cells as well as the identification of individual genes that are differentially expressed upon toxin exposure.
[0007] In various aspects, the invention includes methods of predicting at least one toxic effect of a compound, predicting the progression of a toxic effect of a compound, and predicting the hepatoxicity of a compound. The invention also includes methods of identifying agents that modulate the onset or progression of a toxic response. Also provided are methods of predicting the general pathology classes and cellular pathways that a compound modulates in a cell. The invention includes methods of identifying agents that modulate protein activities.
[0008] In a further aspect, the invention provides probes comprising sequences that specifically hybridize to genes in Tables 1-5XX. Also provided are solid supports comprising at least two of the previously mentioned probes. The invention also includes a computer system that has a database containing information identifying the expression level in a tissue or cell sample exposed to a hepatotoxin of a set of genes comprising at least two genes in Tables 1-5XX. DETAILED DESCRIPTION
[0009] Many biological functions are accomplished by altering the expression of various genes through transcriptional (e.g. through control of initiation, provision of RNA precursors, RNA processing, etc.) and/or translational control. For example, fundamental biological processes such as cell cycle, cell differentiation and cell death are often characterized by the variations in the expression levels of groups of genes.
[0010] Changes in gene expression are also associated with the effects of various chemicals, drags, toxins, pharmaceutical agents and pollutants on an organism or cells. For example, the lack of sufficient expression of functional tumor suppressor genes and/or the over expression of oncogene/protooncogenes after exposure to an agent could lead to tumorgenesis or hypeφlastic growth of cells (Marshall, Cell, 64: 313-326 (1991); Weinberg, Science, 254:1138-1146 (1991)). Thus, changes in the expression levels of particular genes (e.g. oncogenes or tumor suppressors) may serve as signposts for the presence and progression of toxicity or other cellular responses to exposure to a particular compound. [0011] Monitoring changes in gene expression may also provide certain advantages during drag screening and development. Often drugs are screened for the ability to interact with a major target without regard to other effects the drugs have on cells. These cellular effects may cause toxicity in the whole animal, which prevents the development and clinical use of the potential drug.
[0012] The present inventors have examined primary rat hepatocytes exposed to the known hepatotoxins which induce detrimental liver effects, to identify global and individual changes in gene expression induced by these compounds. These global changes in gene expression, which can be detected by the production of expression profiles, as well as the individual genes, provide useful toxicity markers that can be used to monitor toxicity and/or toxicity progression by a test compound. Expression profiles, as well as the individual markers, may also be used to monitor or detect various disease or physiological states, disease progression, drug efficacy and drug metabolism.
Identification of Toxicity Markers
[0013] To evaluate and identify gene expression changes that are predictive of toxicity, studies using selected compounds with well characterized toxicity have been conducted by the present inventors to catalogue altered gene expression during exposure in vivo and in vitro. In the present study, amiodarone, alpha-naphthylisothiocyante (ANIT), acetaminophen (APAP), AY-25329, carbamazepine, carbon tetrachloride, chloφromazine, CI-1000, clofibrate, CPA, diclofenac, diflunisal, dimethylmtiosamine (DMN), 17α-ethinylestradiol, gemfibrozil (Lopid®), hydrazine, imipramine (Janimine), indomethacin, lipopolysaccharide, lovastatin (Mevacor®), methotrexate, phenobarbital, tacrine, tamoxifen, tetracycline, valproate and Wy-14643 were selected as a known hepatotoxins.
[0014] Amiodarone (Cordarone®) is an anti-anhythmic agent whose chemical stracture contains a benzofuran ring (ring A) coupled to a p-OH-benzene structure substituted with 2 iodines and a diethyl-ethanolamine side chain (ring B). This drug is known to cause damage to the liver and has been shown to adversely effect the mitochondria by uncoupling oxidative phosphorylation and inhibiting beta-oxidation and respiration. Inhibition of respiration decreases ATP and increases production of reactive oxygen species, which in turn cause lipid peroxidation. The steatosis and hepatitis observed following treatment with amiodarone are believed to be due, at least in part, to lipid peroxidation products (Spaniol et al, JHepatol 35(5):628-636 (2001); Berson et /., Gastroenterology 114:764-774, (1998)). [0015] Aromatic and aliphatic isothiocyanates are commonly used soil fumigants and pesticides (Shaaya et al, Pesticide Science 44(3):249-253 (1995); Cairns et al, J Assoc Official Analytical Chemists 71(3):547-550 (1988)). These compounds are also environmental hazards, because they remain as toxic residues in plants (Cerny et al, J Agricultural and Food Chemistry 44(12):3835-3839 (1996)) and because they are released from the soil into the surrounding air (Gan et al, JAgricutural and Food Chemistry 46(3):986-990 (1998)).
[0016] Exposure to α-naphthylisothiocyanate (ANIT) has been shown to increase serum levels of total bilirubin, alkaline phosphatase, serum glutamic oxaloacetic transaminase and serum glutamic pyruvic transaminase, while total bile flow was reduced, all of which are indications of severe biliary dysfunction. ANIT also induces jaundice and cholestatis (the condition caused by failure to secrete bile, resulting in plasma accumulation of bile substances, liver cell necrosis and bile duct obstruction) (Tanaka et al, Clinical and Experimental Pharmacology and Physiology 20:543-547 (1993)). ANIT fails to produce extensive necrosis, but was found to produce inflammation and edema in the portal tract of the liver (Maziasa et al, Toxicol Appl Pharmacol 110:365-373 (1991)). ANIT-induced hepatotoxicity may also characterized by cholangiolitic hepatitis and bile duct damage. Acute hepatotoxicity caused by ANIT in rats is manifested as neutrophil-dependent necrosis of bile duct epithelial cells (BDECs) and hepatic parenchymal cells. These changes minor the cholangiolitic hepatitis found in humans (Hill, Toxicol Sci 47:118-125 (1999)). [0017] Histological changes include an infiltration of polymoφhonuclear neutrophils and elevated number of apoptotic hepatocytes (Calvo et al, J Cell Biochem 80(4):461-470 (2001)). Other known hepatotoxic effects of exposure to ANIT include a damaged antioxidant defense system, decreased activities of superoxide dismutase and catalase (Ohta et al, Toxicology 139(3):265-275 (1999)), and the release of proteases from the infiltrated neutrophils, alanine aminotransferase, cathepsin G, elastase, which mediate hepatocyte killing (Hill et ai, Toxicol Appl Pharmacol 148(1):169-175 (1998)). [0018] Acetominophen (APAP) is a widely used analgesic and antipyretic agent that is an effective substitute for aspirin. Although acetaminophen does not have anti-inflammatory properties, it is preferably given to patients with ulcers or patients in whom prolonged clotting times would not be desirable. It also preferably taken by people who do not tolerate aspirin well.
[0019] Acetominophen is metabolized to N-acetyl- -benzoquinoneimine (NAPQI) by N- hydroxylation in a cytochrome P450-mediated process. This highly reactive intermediate, which reacts with sulfhydryl groups in glutathione, and in other liver proteins following the depletion of glutathione, can cause centrilobular hepatic necrosis (particularly in zone 3), renal tubular necrosis, and hepatic and renal failure (Goodman and Gilman' s The Pharmacological Basis of Therapeutics. Ninth Ed.. Hardman et al, eds., pp. 631-633, McGraw-Hill, New York, 1996; Chanda et al, Hepatology 21(2):477-486 (1995)). Less serious side effects include skin rashes (erythemas and urticarias) and allergic reactions. [0020] Upon treatment of rats with acetaminophen, hepatotoxicity can be observed 24 hours after dosing, as determined by statistically significant elevations of ALT and AST in the serum and by hepatocellular necrosis visualized at the light microscopic level (Hessel et al, BrazJ Med Biol Res 29(6):793-796 (1996); Brack et al, Dig Dis Sci 44(6): 1228-1235 (1999)). High, but non-lethal, doses of acetaminophen given to rats also produced elevated levels of genes involved in hepatic acute phase response and liver cell maintenance and repair: arginase, beta-fibrinogen, alpha 1-acid glycoprotein, alpha-tubulin, histone 3, TGF beta and cyclin d. Expression levels of genes regulated by the cell cycle were decreased (Tygstrup et al, JHepatol 25(2):183-190 (1996); Tygsfrup et al, JHepatol 27(1):156-162 (1997)). In mice, expression levels of genes that encode growth anest and cell cycle regulatory proteins were increased, along with expression levels of stress-induced genes, transcription factor LRG-21, SOCS-2 (cytokine signaling repressor) and PAI-1 (plasminogen activator inhibitor-1) (Reilly et al, Biochem Biophys Res Comm 282(l):321-328 (2001)). [0021] AY-25329 is a phenothiazine that has been shown to be toxic in liver and in kidney tissue, where it can cause nephrosis. Phenothiazines are a class of psychoactive drugs that are used to treat schizophrenia, paranoia, mania, hyperactivity in children, some forms of senility, and anxiety (http://www.encyclopedia.com/articlesnew/ 36591.html). Side effects associated with prolonged use of these drugs are reduced blood pressure, Parkinsonism, reduction of motor activity, and visual impairment.
[0022] The present inventors have noted indications of liver and renal effects of AY-25329 by changes in serum chemistry. As early as 6 hours after the first dose, statistically significant increases in serum levels of creatinine, BUN, ALT, triglycerides and cholesterol were observed. Most of these markers of renal and liver dysfunction remained altered throughout the 14 day study period. Light microscopic analysis revealed effects in the liver as early as 6 and 24 hours, as evidenced by an increased number of hepatocytic mitotic figures and decreased glycogen content. Following 14 days of repeated dosing, nephrosis and alterations in the peripheral lobes of the liver and in the cytoplasm of hepatocytes were evident in rats dosed with 250 mg/kg/day of AY-25329.
[0023] Carbamazepine (Tegretol®) is an anti-epieleptic agent. In rats, it has been shown to induce a number of cytochrome P450 enzymes, in particular CYP2B, and the drug may also cause steatohepatitis in humans (Tateishi et al., Chem Biol Interact 117:257-268 (1999); Grieco et al, Eur J Gastioenterol 13(8):973-975 (2001)). [0024] The pathogenesis of acute carbon tetrachloride (CCl4)-induced hepatotoxicity follows a well-characterized course in humans and experimental animals resulting in centrilobular necrosis and steatosis, followed by hepatic regeneration and tissue repair. Severity of the hepatocellular injury is also dose-dependent and may be affected by species, age, gender and diet. [0025] Differences in susceptibility to CC14 hepatotoxicity are primarily related to the ability of the animal model to metabolize CC14 to reactive intennediates. CCl4-induced hepatotoxicity is dependent on CC14 bioactivation to trichloromethyl free radicals by cytochrome P450 enzymes (CYP2E1), localized primarily in centrizonal hepatocytes. Formation of the free radicals leads to membrane lipid peroxidation and protein denaturation resulting in hepatocellular damage or death. [0026] The onset of hepatic injury is rapid following acute administration of CC14 to male rats. Moφhologic studies have shown cytoplasmic accumulation of lipids in hepatocytes within 1 to 3 hours of dosing, and by 5 to 6 hours, focal necrosis and hydropic swelling of hepatocytes are evident. Centrilobular necrosis and inflammatory infiltration peak by 24 to 48 hours post dose. The onset of recovery is also evident within this time frame by increased DNA synthesis and the appearance of mitotic figures. Removal of necrotic debris begins by 48 hours and is usually completed by one week, with full restoration of the liver by 14 days. [0027] increases in serum transaminase levels also parallel CCl4-induced hepatic histopathology. In male Sprague Dawley (SD) rats, alanine aminotrasferase (ALT) and aspartate aminotransferase (AST) levels increase within 3 hours of CC14 administration (0.1,
1,2, 3, 4 mL/kg, ip; 2.5 lnL/kg, po) and reach peak levels (approximately 5-10 fold increases) within 48 hours post dose. Significant increases in seram -glutathione s-transferase (-GST) levels have also been detected as early as 2 hours after CC14 administration (25 L/kg, po) to male SD rats.
[0028] At the molecular level, induction of the growth-related proto-oncogenes, c-fos and c-jun, is reportedly the earliest event detected in an acute model of CCl4-induced hepatotoxicity (Schiaffonato et al, Liver 17:183-191 (1997)). Expression of these early- immediate response genes has been detected within 30 minutes of a single dose of CCL to mice (0.05 -1.5 mL/kg, ip) and by 1 to 2 hours post dose in rats (2 mL/kg, po; 5 mL/kg, po) (Schiaffonato et ai, supra, and Hong et ai, Yonsei Medical J 8:167 -177 (1997)). Similarly, hepatic c-myc gene expression is increased by 1 hour following an acute dose of CC14 to male SD rats (5 mL/kg, po) (Hong et al, supra). Expression of these genes following exposure to CC14 is rapid and transient. Peak hepatic mRNA levels for c-fos, c-jun, and c- myc, after acute administration of CC14 have been reported at 1 to 2 hours, 3 hours, and 1 hour post dose, respectively.
[0029] The expression of tumor necrosis factor-α (TNF-α) is also increased in the livers of rodents exposed to CC14, and TNF-α has been implicated in initiation of the hepatic repair process. Pre-treatment with anti-TNF-α antibodies has been shown to prevent CC14 - mediated increases in c-jun and c-fos gene expression, whereas administration of TNF-α induced rapid expression of these genes (Braccoleri et al, Hepatol 25:133-141 (1997)). Upregulation of transforming growth factor- β (TGF-β) and transforming growth factor receptors (TBRI-III) later in the repair process (24 and 48 hours after CC14 administration) suggests that TGF-β may play a role in limiting the regenerative response by induction of apoptosis (Grasl-Kraupp et al, Hepatol 28:717-7126 (1998)). [0030] Chloφromazine (Thorazine®) is a central nervous system depressant that is used as a sedative and also as an anti-nausea or anti-itching medication. The mechanism of action is not known. The drug induces canalicular cholestasis, a condition characterized by a decrease in the volume of bile formed and impaired secretion of solutes into bile, resulting in elevated serum levels of bile salts and bilirubin. Chloφromazine has also been shown to inhibit bile acid uptake and canalicular contractility. Bile plugs can form in the bile ducts and canaliculi. Drag-induced cholestasis is also associated with cell swelling, inflammation and cell death (Casarett and Doull's Toxicology: The Basic Science of Poisons, 6th Ed.. Klaassen et al. eds., pp. 476-486, McGraw-Hill Medical Pub. Div., New York, 2001). [0031] CI-1000 (4H-pynolo:3,2-d:pyrimidin-4-one, 2-amino-3,5-dihydro-7-(3- thienylmethyl)-monohydrochloride monohydrate) is a compound with anti-inflammatory properties. After treatment with CI-1000, increased serum ALT levels, a standard marker of liver toxicity, were observed in dogs.
[0032] Clofibrate, a halogenated phenoxypropanoic acid derivative (ethyl ester of clofibric acid), is an antilipemic agent. The exact mechanism by which clofibrate lowers serum concentrations of triglycerides and low-density lipoprotein (LDL) cholesterol, as well as raising high-density lipoprotein (HDL) cholesterol is uncertain. The drug has several antilipidemic actions, including activating lipoprotein lipase, which enhances the clearance of triglycerides and very-low-density lipoprotein (VLDL) cholesterol, inhibition of cholesterol and triglyceride biosynthesis, mobilization of cholesterol from tissues, increasing fecal excretion of neutral steroids, decreasing hepatic lipoprotein synthesis and secretion, and decreasing free fatty acid release.
[0033] Clofibrate has a number of effects on the rat liver, including hepatocellular hypertrophy, cellular proliferation, hepatomegaly, induction of CYP450 isozymes, and induction of palmitoyl Co A oxidation. Long term administration of clofibrate causes increased incidence of hepatocellular carcinoma, benign testicular Leydig cell tumors, and pancreatic acinar adenomas in rats. Clofibrate induces proliferation of peroxisomes in rodents and this effect, rather than genotoxic damage, is believed to be the causative event in rodent carcinogenesis (AHFS Drug Information Handbook 2001, McEvoy, ed., pp.1735- 1738; Electronic Physicians' Desk Reference- Atromid-S (clofibrate) at www.pdr.net; Brown and Goldstein, "Drugs used in the treatment of hyperliproteinemias," in Goodman and Gilman's The Pharmacological Basis of Therapeutics. Eighth ed„ Goodman et al, eds., pp. 874-896, Pergamon Press, New York, 1990). [0034] Clofibrate also increases hepatic lipid content and alters its normal composition by significantly increasing levels of phosphatidylcholine and phosphatidyl-ethanolamine (Adinehzadeh et al, Chem Res Toxicol 11(5):428-440 (1998)). A rat study of liver hypeφlasia and liver tumors induced by peroxisome proliferators revealed that administration of clofibrate increased levels of copper and altered copper-related gene expression in the neoplastic liver tissues. Down-regulation of the ceruloplasmin gene and of the Wilson's Disease gene (which encodes P-type ATPase), along with up-regulation of the metallothionein gene, were noted in these tissues (Eagon et al, Carcinogenesis 20(6):1091- 1096 (1999)). Clofibrate-induced peroxisome proliferation and carcinogenicity are believed to be rodent-specific, and have not been demonstiated in humans. [0035] Cyproterone acetate (CPA) is a potent androgen antagonist and has been used to treat acne, male pattern baldness, precocious puberty, and prostatic hypeφlasia and carcinoma (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9 ed., p. 1453, J.G. Hardman et al, Eds., McGraw Hill, New York, 1996). Additionally, CPA has been used clinically in hormone replacement therapy to protect the endometrium and decrease menopausal symptoms and the risk of osteoporotic fracture (Schneider, "The role of antiandrogens in hormone replacement therapy," Climacteric 3 (Suppl. 2): 21-27 (2000)). [0036] In experiments with rats, CPA was shown to induce unscheduled DNA synthesis in vitro. After a single oral dose, continuous DNA repair activity was observed after 16 hours. CPA also increased the occurrence of S phase cells, which conoborated the mitogenic potential of CPA in rat liver (Kasper et al, Carcinogenesis 17(10): 2271-2274 (1996)). CPA has also been shown to produce cinhosis in humans (Garty et al, Eur JPediatr 158(5): 367- 370 (1999)).
[0037] Diclofenac, a non-steroidal anti-inflammatory drug, has been frequently admimstered to patients suffering from rheumatoid arthritis, osteoarthritis, and ankylosing spondylitis. Following oral administration, diclofenac is rapidly absorbed and then metabolized in the liver by cytochrome P450 isozyme of the CYC2C subfamily (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9th ed., p. 637, J.G. Hardman et al, eds., McGraw Hill, New York, 1996). In addition, diclofenac has been applied topically to treat pain due to corneal damage (Jayamanne et al, Eye ll(Pt. 1): 79-83 (1997); Domic et al, Am J Ophthalmol 125(5): 719-721 (1998)).
[0038] Although diclofenac has numerous clinical applications, adverse side-effects have been associated with the drug, such as corneal complications, including corneal melts, ulceration, and severe keratopathy (Guidera et al, Ophthalmology 108(5): 936-944 (2001)). Another study investigated 180 cases of patients who had reported adverse reactions to diclofenac to the Food and Drug Administration (Banks et al, Hepatology 22(3): 820-827 (1995)). Of the 180 reported cases, the most common symptom was jaundice (75% of the symptomatic patients). Liver sections were taken and analyzed, and hepatic injury was apparent one month after drug tieatment. An additional report showed that a patient developed severe hepatitis five weeks after beginning diclofenac treatment for osteoarthritis (Bhogaraju et al, South MedJ92(7): 711-713 (1999)).
[0039] In one study on diclofenac-treated Wistar rats (Ebong et al, Afr JMed Sci 27(3-4): 243-246 (1998)), diclofenac treatment induced an increase in serum chemistry levels of alanine aminotransferase, aspartate aminotransferase, methaemoglobin, and total and conjugated bilirubin. Additionally, diclofenac enhanced the activity of alkaline phosphatase and 5'nucleotidase. A study on humans revealed elevated levels of hepatic transammases and serum creatine when compared to the control group (McKenna et al, Scand J Rheumatol 30(1): 11-18 (2001)).
[0040] Diflunisal, a non-steroidal anti-inflammatory drug (NSAID), is a difluorophenyl derivative of salicylic acid (Goodman & Gilman's The Pharmacological Basis of Therapeutics 9th ed., p. 631, J.G. Hardman et al, Eds., McGraw Hill, New York, 1996). It is most frequently used in the treatment of osteoarthritis and musculoskeletal strains. NSAIDs have analgesic, antipyretic and anti-inflammatory actions, however, hepatotoxicity is known to be an adverse side effect of NSAID treatment (Masubucbi et al, J Pharmacol Exp Ther 287:208-213 (1998)). Diflunisal has been shown to be less toxic than other NSAIDs, but it can eventually have deleterious effects on platelet or kidney function (Bergamo et al, Am J Nephrol 9:460-463 (1989)). Other side effects that have been associated with diflunisal treatment are dianhea, dizziness, drowsiness, gas or heartburn, headache, nausea, vomiting, and insomnia (h1φ://arthritisinsight.com/medical/ meds/dolobid.html). [0041] In a comparative hepatotoxicity study of 18 acidic NSAIDs, diflunisal was shown to increase LDH leakage in rat hepatocytes, a marker for cell injury, when compared to control samples. Additionally, tieatment with diflumsal led to decreased intiacellular ATP concentrations. In a study comparing the effects of diflunisal and ibuprofen, both drags appeared to cause abdominal cramping, even during short-term usage. Because the toxic dosages were selected to be below the level at which gastric ulceration occurs, more severe gastrointestinal effects were not detected. But, increased serum levels of creatinine, a sign of renal injury, were also observed (Muncie et al, Clin Ther 11:539-544 (1989)). [0042] Another model compound, dimethylnitiosamine (DMN), is a known carcinogen and inducer of liver fibrosis and lipid peroxidation. DMN causes oxidative stress in liver cells, which may be the link between chronic liver damage and liver fibrosis. Rats treated with DMN showed diffuse fibronectin deposition, elevated hydroxyproline levels (an indicator of fibrosis), increased levels of collagens, fibrous septa, and impaired oxidative balance. Serum levels of ALT and malondialdehyde (MDA) were increased, while serum levels of SOD were decreased (Vendemiale et al, Toxicol Appl Pharmacol 175(2):130-139 (2001); Liu et al, Zhonghua Gan Zang Bing Za Zhi 9 Suppl: 18-20 (2001)). Other studies in rats have noted severe centrilobular congestion and haemonhagic necrosis several days after a three-day period of DMN administration. Following additional periods of DMN treatment, the rats developed centrilobular necrosis and intense neutrophilic infiltration, which progressed to severe centrilobular necrosis, fiber deposition, focal fatty deposits, bile duct proliferation, bridging necrosis and fibrosis around the central veins (cinhosis-like symptoms). A decrease in total protein and increase in DNA were also observed (George et al. (2001) Toxicology 156(2-3):129-138).
[0043] 17α-ethinylesfradiol, a synthetic estrogen, is a component of oral contraceptives, often combined with the progestational compound norethindrone. It is also used in post- menopausal estrogen replacement therapy (PDR 47th Ed., pp. 2415-2420, Medical Economics Co., Inc., Montvale, NJ, 1993; Goodman & Gilman's The Pharmalogical Basis of Therapeutics 9th Ed., pp. 1419-1422, J.G. Hardman et al. Eds., McGraw Hill, New York, 1996).
[0044] The most frequent adverse effects of 17 -ethinylestiadiol usage are increased risks of cardiovascular disease: myocardial infarction, thromboembolism, vascular disease and high blood pressure, and of changes in carbohydrate metabolism, in particular, glucose intolerance and impaired insulin secretion. There is also an increased risk of developing benign hepatic neoplasia. Because this drug decreases the rate of liver metabolism, it is cleared slowly from the liver, and carcinogenic effects, such as tumor growth, may result. [0045] 17α-ethinylestradiol has been shown to cause a reversible intiahepatic cholestasis in male rats, mainly by reducing the bile-salt-independent fraction of bile flow (BSIF) (Koopen et al, Hepatology 27:537-545 (1998)). Plasma levels of bilirabin, bile salts, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in this study were not changed. This study also showed that 17α-ethinylestiadiol produced a decrease in plasma cholesterol and plasma triglyceride levels, but an increase in the weight of the liver after 3 days of drug administration, along with a decrease in bile flow. Further results from this study are as follows. The activities of the liver enzymes leucine aminopeptidase and alkaline phosphatase initially showed significant increases, but enzyme levels decreased after 3 days. Bilirubin output increased, although glutathione (GSH) output decreased. The increased secretion of bilirubin into the bile without affecting the plasma level suggests that the increased bilirubin production must be related to an increased degradation of heme from heme-containing proteins. Similar results were obtained in another experiment (Bouchard et al, Liver 13:193- 202 (1993)) in which the livers were also examined by light and election microscopy. Daily doses of 17α-ethinylestradiol have been shown to cause cholestasis as well, although, following drug tieatment, bile flow rates gradually returned to normal (Hamada et al, Hepatology 21 : 1455-1464 (1995)). Liver hypeφlasia, possibly in response to the effects of tumor promoters, has also been observed (Mayol, Carcinogenesis 13:2381-2388 (1992)). [0046] The lipid-lowering drag gemfibrozil (Lopid®) is a know peroxisome proliferator in liver tissue, causing both hypeφlasia and enlargement of liver cells. Upon exposure to gemfibrozil, hepatocarcinogenesis has been observed in rats and mice, and a decrease in alpha-tocopherol and an increase in DT-diaphorase activity have been observed in rats and hamsters (impaired antioxidant capability). Peroxisome prohferators increase the activities of enzymes involved in peroxisomal beta-oxidation and omega-hydroxylation of fatty acids, which results in oxidative stress (O'Brien et al, Toxicol Sci 60(2):271-278 (2001); Carfhew et al, JAppl Toxicol 17(1):47-51 (1997)). [0047] Hydrazine (NH2=NH2), is a component of many industrial chemicals, such as aerospace and aiφlane fuels, conosion inhibitors, dyes and photographic chemicals. Its derivatives are used in pharmaceuticals such as hydrazine sulphate, used to treat cachexia in cancer patients, isoniazid, an anti-tuberculosis drug, and hydralazine, an anti-hypertensive. These drags are metabolized in vivo to produce hydrazine, among other by-products. Consequently, exposure to hydrazine is by direct contact, e.g., among military and airline personnel, or the result of its production in the body, e.g., in patients with cancer or high blood pressure.
[0048] Studies on rat hepatocytes have shown that hydrazine causes a dose-dependent loss of viability, leakage of LDH, depletion of GSH and ATP and a decreased rate of protein synthesis (Delaney et al, Xenobiotica 25(12):1399-1410 (1995)). When administered to rats, hepatotoxic changes, characterized by GSH and ATP depletion and induction of fatty liver (increases in liver weight and triglycerides, with the appearance of fatty droplets, swelling of mitochondria and appearance of microbodies) were also found to be dose-dependent (Jenner et al., Arch Toxicol 68(6):349-357 (1994); Scales et al, J Toxicol Environ Health 10(6):941- 953 (1982)). The hepatoxicity, as well as renal toxicity, associated with hydrazine exposure has been linked to free radical damage resulting from oxidative metabolism by cytochrome P4502E1 (CYP2E1), which catalyzes the conjugation of free radicals with reduced glutathione. Although exposure to hydrazine and several hydrazine derivatives increased enzyme levels in kidney tissue, increased enzyme levels were not detected in liver tissue (Runge-Morris et al, Drug Metab Dispos 24(7):734-737 (1996)). [0049] The mutagenic and hepatocarcinogenic effects of hydrazine were examined in hamster livers. In vivo, hydrazine reacts with formaldehyde to form formaldehyde hydrazone (CH2=N-NH2), an alkylating intermediate that methylates guanine in DNA. Upon treatment with hydrazine, liver DNA showed the presence of methylated guanine, DNA adducts and the impairment of maintenance methylation (impaired methylation of deoxycytosine). Hepatic adenomas and carcinomas also developed in a dose-dependent manner and could be conelated with decreased maintenance methylation (FitzGerald et al, Carcinogenesis 17(12):2703-2709 (1996)).
[0050] Imipramine, a dibenzazepine derivative, is a tricyclic anti-depressant agent commonly used for the treatment of major depression. Experiments in rats have shown that the drug induces cytochrome P450-mediated formation of reactive metabolites, which cause acute cell injury. Decreased levels of glutathione and protein thiols, as well as lactate dehydrogenase leakage, all standard measures of liver toxicity, were also noted (Masubuchi et al., Arch Toxicol 73(3): 147-151 (1999). On rare occasions, imipramine has induced cholestasis and hepatitis in humans (Moskovitz et al, J Clin Psychiatry 43(4):165-066 (1982); Horst et al, Gastioenterology 79(3):550-544 (1980)).
[0051] Indomethacin is a non-steroidal antiinflammatory, antipyretic and analgesic drug commonly used to treat rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, gout and a type of severe, chronic cluster headache characterized by many daily occuπences and jabbing pain. This drug acts as a potent inhibitor of prostaglandin synthesis; it inhibits the cyclooxygenase enzyme necessary for the conversion of arachidonic acid to prostaglandins fPDR 47th Ed.. Medical Economics Co., Inc., Montvale, NJ, 1993; Goodman & Gilman's The Pharmalogjcal Basis of Therapeutics 9th Ed.. J.G. Hardman et al. eds., pp. 1074-1075, 1089- 1095, McGraw Hill, New York, 1996; Cecil Textbook of Medicine, 20th Ed., part XII, pp. 772-773, 805-808, J. C. Bennett andF. Plum Eds., W. B. Saunders Co., Philadelphia, 1996). [0052] The most frequent adverse effects of indomethacin treatment are gastrointestinal disturbances, usually mild dyspepsia, although more severe conditions, such as bleeding, ulcers and perforations can occur. Hepatic involvement is uncommon, although some fatal cases of hepatitis and jaundice have been reported. Renal toxicity can also result, particularly after long-term administration. Renal papillary necrosis has been observed in rats, and interstitial nephritis with hematuria, proteinuria and nephrotic syndrome have been reported in humans. Patients suffering from renal dysfunction risk developing a reduction in renal blood flow, because renal prostaglandins play an important role in renal perfusion. [0053] In rats, although indomethacin produces more adverse effects in the gastrointestinal tract than in the liver, it has been shown to induce changes in hepatocytic cytochrome P450. In one study, no widespread changes in the liver were observed, but a mild, focal, centrilobular response was noted. Serum levels of albumin and total protein were significantly reduced, while the serum level of urea was increased. No changes in creatinine or aspartate aminotransferase (AST) levels were observed (Falzon et al, Br J exp Path 66:527-534 (1985)). In another rat study, a single dose of indomethacin was shown to reduce liver and renal microsornal enzymes, including CYP450, and cause lesions in the GI tract (Fracasso et al, Agents Actions 31:313-316 (1990)).
[0054] LPS (lipopolysaccharide) is an endotoxin released by gram-negative bacteria upon breakage or rupture of the cells that induces an acute inflammatory response in mammals and that can cause septic shock. LPS is also a research tool used to initiate liver injury in rats through an inflammatory mechanism. Typically, the membrane components of LPS are lipid- A, KDO (2-keto-3-deoxy-octulosonic acid), core polysaccharides and O-antigen polysaccharides, the polysaccharide units differing from one bacterium to another (Zjnsser Microbiology 20th Ed.. Joklik et al., eds., pp. 82-87, Appleton & Lange, Norwalk, CT, 1992). [0055] Primary rat hepatocytes derived from liver parenchymal cells and sinusoidal cells of rats that have been exposed to LPS in vivo can directly respond to LPS in cell culture. Numerous effects of LPS-induced endotoxemia can be detected, including elevated levels of nitric oxide synthetase (NOS) with increased nitric oxide and nitrite production, cellular hypertrophy, vacuohzation, chromosomal emargination, cytoplasmic DNA fragmentation and necrosis (Pittner et al, Biochem Biophys Res Commun 185(l):430-435 (1992); Laskin et al, Hepatology 22(l):223-234 (1995); Wang et al, Am J Physiol 269(2 Pt l):G297-304 (1995)). Other studies have indicated that the presence of Kupffer cells with primary rat hepatocytes is essential for the induction of hepatocyte apoptosis by LPS (Hamada et al., JHepatol 30(5):807-818 (1999)).
[0056] Exposure of rats or primary hepatocytes to LPS induces the expression of a number of acute-phase proteins in the liver. Recent evidence has indicated that rat hepatocytes express soluble CD14 protein, and LPS is capable of markedly increasing levels of CD14 at both the gene expression and protein expression levels (Liu et al, Infect Immun 66(11):5089- 5098 (1998)). Soluble CD14 is believed to be a critical LPS recognition protein required for the activation of a variety of cells to toxic levels of LPS, even in endothelial and epithelial cells (Pugin et ai, Proc Natl Acad Sci USA 90(7):2744-2748 (1993)). Another key component of the LPS recognition system is lipopolysaccharide-binding protein (LBP), which binds to LPS. The LPS-LBP complex interacts with the CD 14 receptor, inducing LPS sensitive genes. LBP can be induced in hepatocytes isolated from rats that have not been primed with LPS, indicating that this key regulatory pathway is intact in primary rat hepatocytes (Wan et al, Infect Immun 63(7):2435-2442 (1995)). [0057] Lovastatin (Mevacor®) is a cholesterol-lowering agent belonging to a class of compounds, the statins, that are potent inhibitors of HMG-CoA reductase. This enzyme catalyzes the conversion of HMG-CoA to mevalonate, the rate-controlling enzyme in cholesterol biosynthesis. HMG-CoA reductase inhibitors block the production of cholesterol in the liver leading to a reduction of LDL particles in the plasma. Lovastatin has additional effects on lipid metabolism, including depletion of intracellular pools of sterols and increased synthesis of LDL receptors, leading to enhanced removal of LDL and LDL precursors from plasma. Upon treatment with lovastatin, plasma levels of VLDL, IDL and triglycerides also decrease. Oral doses of lovastatin are extensively absorbed by the liver, and the drug is excreted primarily via the liver; less than 13% of its metabolites are excreted in the urine (Goodman and Gilman's The Pharmacological Basis of Therapeutics. Ninth Ed.. Hardman et al, eds., pp. 884-888, McGraw-Hill, New York, 1996).
[0058] The most frequent side effects are liver damage, characterized by elevated levels of hepatic transammases (e.g., AST and ALT), creatinine phosphokinase and alkaline phosphatase, and myopathy, characterized by muscle pain and destruction of skeletal muscle cells. Cases of drug-induced hepatitis, accompanied by jaundice and elevated levels of liver enzymes, have also been reported, although the symptoms were reversible following withdrawal of the drug (Huchzermeyer et al, Deutsch Med Wochenschr 120(8):252-256 (1995); Heuer et al, Med Klin 95(ll):642-644 (2000)). Histologic examination of affected liver tissue showed centrilobular necrosis, centrilobular cholestasis, and infiltrates with mononuclear and polymoφhonuclear cells, including eosinophils (Grimbert et al, Dig Dis Sci 39(9):2032-2033 (1994)).
[0059] Experiments by the present inventors have found that when rats were dosed with lovastatin, at 9 or 90 mg/kg twice a day, no effects were noted in liver tissue after 6 or after 24 hours. After 14 days of treatment at the higher dosage, however, liver cells showed abnormal vacuolization of the cytoplasm. Hepatoxicity data from other studies of laboratory animals treated with lovastatin have not been widely reported. Therefore, in order to establish a more sensitive model for examining the changes in liver tissue caused by lovastatin, as well as to have a means of examining changes in expression level of individual genes as a result of exposure to lovastatin, experiments in cultured hepatocytes were undertaken.
[0060] Methotrexate is a widely used antineoplastic drag that is also frequently prescribed for the treatment of psoriasis (a disease characterized by abnormal proliferation of epidermal cells), juvenile lymphoblastic leukemia, rheumatoid arthritis, and a number of other cancerous diseases, such as leukemic meningitis and choriocarcinoma. Although generally not metabolized, at high dosages, metabolites such as 7-hydroxy-methotrexate, a nephrotoxin, do accumulate. Methotrexate polyglutamates are retained in the kidneys for weeks and in the liver for months ((Goodman and Gilman's The Pharmacological Basis of Therapeutics. Ninth Ed., Hardman et al, eds., pp. 1243-1247, McGraw-Hill, New York, 1996). [0061] Methotrexate acts to prevent DNA synthesis and cell replication by inhibiting the rate-limiting enzyme in purine and thymidine synthesis, dihydrofolate reductase (DHFR) (Goodman and Gilman's, supra; Schwartz et al, Proc Nat Acad Sci USA 89(2):594-598 (1992)). It also acts as an suppressant of cell-mediated immune responses. The biochemical toxicology of methotrexate has been well characterized in man, where long-term administration produces hepatic fibrosis or cinhosis, especially in heavy drinkers, which is linked to persistent, mild-to-moderate, increases in serum transammases, alkaline phosphatases and bilirubin (Reynolds et al, South Med J '1 '9(5):536-539 (1986); Tolman et al, JRheumatol 12 (Suppl 12):29-34 (1985)). Methotrexate is a rather long-acting, rapidly reversible DHFR inhibitor, despite its high affinity for the target enzymes in many cell types, which may be due to the formation of methotrexate polyglutamates that reduce the ability of DHFR to pass through cell membranes. The toxic effects of methotrexate may be due to the depletion of tetrahydrofolate cofactors that are required for purine and thymidylate synthesis (methylation reactions in hepatic 1 -carbon metabolism) and to the inliibition of folate- dependent enzymes involved in the metabolism of purines and thymidylate, the inhibition caused by the accumulation of methotrexate polyglutamates and dihydrofolate polyglutamates.
[0062] The mechanism of methotrexate-induced hepatotoxicity is not yet fully elucidated, partly because the pathological changes in humans are rather difficult to reproduce in animal models, although experiments in rats have shown that, in a dose-dependent fashion, methotrexate produces liver damage ranging from focal to confluent necrosis of zone 3 hepatocytes, with early stage fibrosis (Hall et al, Hepatology 14(5):906-10 (1991)). Other studies in rats have demonstrated that treatment with methotrexate produces intrahepatocytic fat deposits, along with fatty accumulations in hepatocytes that range from fine droplets to large vacuoles. The areas of necrosis showed signs of the hypoxia associated with congestive failure, as well as anemic infarcts, fibrotic foci of the collapse type, atrophy of the hepatic cords, and hemosiderosis (Custer et al, JNatl Cancer Inst 58(4):1011-1015 (1977)). Hepatotoxicity probably involves interference with triglyceride and other lipid biosynthetic pathways in the liver. For example, studies on rats have shown that methotrexate inhibits the biosynthesis of lipotropic substances such as methionine and choline tlirough its interference with hepatic 1 -carbon metabolism. The drug also inhibits the activity of vitamin B12, another lipotropic factor (Tuma et al, Biochem Pharmacol 24:1327-1331 (1975) and impairs RNA and protein synthesis, triglyceride secretion and total triglyceride esterification (Deboyser et al, Toxic in Vitro 6(2):129-132 (1992).
[0063] Methotrexate does not appear to be cytotoxic to cultured primary hepatocytes following short-term exposures (up to 3.5 hr), but significant effects on HepG2 growth curves have been observed at low concentrations during the course of 7-day exposures (Wu et al, Proc Natl Acad Sci USA 80(10):3078-3080 (1983)). Additionally, it has been demonstrated that methotrexate increases hepatic glycogenolysis, oxygen consumption and calcium efflux and decreases glutathione levels (Yamamoto et al, Biochem Pharmacol 44(4):761-767, (1992); de Oliveira et al, Res Commun Chem Pathol Pharmacol 53(2):173-181 (1986); Lindenthal et al, Eur J Pharmacol 228(5-6):289-298 (1993)). Experiments on cultured rat hepatocytes have shown that methotrexate also inhibits the activity of hepatic N- acetyltransferase 2 (NAT2), although the drug has only a slight inhibitory effect on rat NATI, enzymes that catalyze the acetylation of a variety of therapeutic arylamines (Zaher et al, Toxicol in Vitro 11:271-283 (1997)).
[0064] Phenobarbital, a barbiturate, is used as an anti-epileptic, sedative or hypnotic drug and can also be used to treat neuroses with related tension states, such as hypertension, coronary artery disease, gastrointestinal disturbances and preoperative apprehension. Phenobarbital is also found in medications to treat insomnia and headaches (Remington: The Science and Practice of Pharmacy. 19th Ed.. A. R. Gennaro ed., pp. 1164-1165, Mack Publishing Co., Easton, Pennsylvania, 1995).
[0065] Phenobarbital induces a variety of drag metabolizing enzymes such as cytochrome P450 oxidoreductase, aldehyde dehydrogenases, UDP-glucuronyltransferase, GSTs, epoxide hydrolase, and an assortment of cytochrome P450 monooxygenases (Waxman et al, Biochem J 1281(Pt 3):577-592 (1992); Kaplowitz et al, Biochem J 146(2):351-356 (1975); Tank et al, Biochem Pharmacol 35(24):4563-4569 (1986). The induction of liver enzymes is usually accompanied by liver enlargement, proliferation of smooth endoplasmic reticulum, and tumor promotion (Waxman et al, supra; Rice et al, Carcinogenesis 15(2):395-402 (1994); Nims et al, Carcinogenesis 8(1):67-71, (1987). Incidences of cholestasis and hepatocellular injury have also been found (Selim et al, Hepatology 29(5):1347-1351 (1999); Gut et al, Environ Health Perspect 104(Suppl 6):1211-1218 (1999)). Phenobarbital has been classified as nongenotoxic hepatocarcinogen as it induces liver tumors in rodents but lacks detectable signs of genotoxicity using short term in vivo or in vitro assays (Whysner et al, Pharmacol Ther 71(1-2): 153-191 (1996)).
[0066] The effects of phenobarbital on phospholipid metabolism in rat liver have been studied. In one study, phenobarbital, administered intraperitonially (i.p.), was found to cause an increase in the microsomal phosphatidylcholine content. Additionally, levels of glycerophosphate acyltransferase (GAT), phosphatidate cytidylyltransferase (PCT), phosphatidate phosphohydrolase (PPH) and choline phosphotransferase (CPT) were significantly increased (Hoshi et al, Chem Pharm Bull 38:3446-3448 (1990)). [0067] Tacrine (l,2,3,4-tetiahydro-9-aminoacridine-hydrochloride), a strong acetylcholinesterase (AChE) inhibitor, is used in the tieatment of mild to moderate cases Alzheimer's dimentias. Alzheimer's patients have synaptic loss, neuronal atrophy and degeneration of cholinergic nuclei in the forebrain, which are associated with reduced oxidative metabolism of glucose and decreased levels of ATP and acetyl Co A. Administration of AChE inhibitors, such as tacrine, is designed to increase cholinergic activity to combat this loss (Weinstock, Neurodegeneration 4(4):349-356 (1995)). The effect seen in the patients is a reversal of the cognitive and functional decline, but the drug does not appear to change the neurodegenerative process (Goodman & Gilman's The Pharmacological
Basis of Therapeutics 9m Ed.. Hardman et al. eds., p. 174, McGraw Hill, New York, 1996). [0068] Hepatotoxicty caused by tacrine is typically reversible, although cases of severe hepatotoxicity have been seen (Blackard et al, J Clin Gastroenterol 26:57-59 (1998)). The toxicity is characterized by decreased levels of protein synthesis and the release of lactate dehydrogenase, as well as by increased transaminase levels and decreased levels of ATP, glycogen and glutathione. The decrease in protein synthesis may represent a signal leading to cell death (Lagadic-Gossmann et al, Cell Biol Toxicol 14(5):361-373 (1998)). [0069] Preclinical studies have failed to detect adverse hepatic events, although tacrine displayed cytotoxicity to human hepatoma cell lines and primary rat hepatocytes (Viau et al, Drug Chem Toxicol 16:227-239 (1993)). While hepatotoxicity has been found in humans, in vivo rat studies have not shown a conelation between tacrine exposure and hepatotoxicity, and the mechanism of action is not completely understood. An in vitro study comparing the reaction of human and rat liver microsomal preparations to tacrine showed that the two species reacted differently to the drag, suggesting that the rat may not be the best model for monitoring tacrine-induced elevations in liver marker enzymes (Woolf et al, Drug Metab Dispos 21:874-882 (1993)).
[0070] While tacrine does not reveal classic signs of hepatotoxicity in rats, gene expression changes due to tacrine administration can be used to predict that the drug will be a liver toxin in humans. This suggests that toxicogenomics might be able to detect drugs that prove to be toxic in the clinic even when classical but more crude measures in preclinical screening fail to detect toxicity.
[0071] Tamoxifen is a nonsteroidal anti-estrogen used for breast cancer in males and females. Tamoxifen has been associated with changes in liver enzyme levels, disruption of mitochondrial metabolism and, occasionally, with a spectrum of more severe liver abnormalities including fatty liver, cholestasis, hepatic necrosis and NASH (nonalcoholic steatohepatitis) (Duthie et al, Xenobiotica 25(10):1151-1164 (1995); Cardoso et al, Toxicol Appl Pharmacol 176(3):145-152 (2001); Pinol et ai, Gastroenterol Hepatol 23(2):57 '-61 (2000); and Farrell, Semin Liver Dis 22(2):185-194 (2002)). A few of these serious cases included fatalities. A few cases of liver cancer have also been reported. Additionally, studies in mice and rats have shown that tamoxifen significantly alters the activities of liver enzymes and can induce hepatic carcinomas (Caballero et al, hit J Biochem Cell Biol 33(7):681-690 (2001); Guzelian, Semin Oncol 24(1 Suppl 1):S1-105-121 (1997)). [0072] Tetracycline is a broad spectrum antibiotic whose main mechanism of action is the inhibition of bacterial protein synthesis. Hepatic toxicity, principally steatosis, has been observed in patients receiving high doses of tetracycline. In rats and dogs, exposure to tetracycline has been shown to increase levels of total lipids and triglycerides in liver cells due to inhibition of mitochondrial lipid metabolism and beta-oxidation (Lewis et al., Am J Dig Dis 12:429-438, (1967); Amacher et al., Fundam Appl Toxicol 40(2):256-263 (1997). [0073] Valproate (n-dipropylacetic acid, Depakene®) is routinely used to tieat several types of epileptic seizures- absence seizures, myoclonic seizures and tonic-clonic seizures. Most other anti-epileptics are effective against only one type. Valproate acts on neurons to inhibit the sustained repetitive firing caused by depolarization of cortical or spinal cord neurons, and
+ a prolonged recovery of inactivated voltage-activated Na channels follows. The drag also acts by reducing the low-threshold Ca cunent and its multiple mechanisms contribute to its use in multiple types of seizures. Although valproate does not affect neuronal responses to GABA, it does increase the activity of the GABA synthetic enzyme, glutamic acid decarboxylase, and it inhibits enzymes that degrade GABA, GABA transaminase and succinic semialdehyde dehydrogenase (Goodman and Gilman's The Pharmacological Basis of Therapeutics. 9th Ed.. Hardman et al, eds., pp. 462, 476 and 477, McGraw-Hill, New York, 1996).
[0074] The most common side effects are gastrointestinal symptoms, including anorexia, nausea and vomiting. Effects on the CNS include sedation, ataxia and tremor. Rash, hair loss, increased appetite and teratogenic effects have also been observed (Briggs et al, A Reference Guide to Fetal and Neonatal Risk. Drugs in Pregnancy and Lactation. 4th ed.. p. 869, Williams & Wilkins, Baltimore, 1994). With respect to liver toxicity, valproate produces elevated levels of hepatic enzymes in about 40% of patients, which may be an asymptomatic condition, and elevated levels of hepatic lipids. Fulminant hepatitis, microvesicular steatosis (fatty degeneration), hepatocyte necrosis and hepatic failure can also result. It is believed that hepatoxicity is caused by an accumulation of unsaturated metabolites of valproate, in particular 4-en- valproate, which is structurally similar to two known hepatotoxins, 4-en-pentanoate and methylenecyclopropylacetic acid (Eadie et al, Med Toxicol Adverse Drug Exp 3(2):85-106 (1988)). [0075] In a study on rats, microvesicular steatosis caused by valproate was found to be accompanied by myeloid bodies, lipid vacuoles and mitochondrial abnormalities (Kesterson et al, Hepatology 4(6): 1143-1152 (1984)). Experiments on cultured rat hepatocytes have shown that valproate produces a dose-dependent leakage of lactic acid dehydrogenase and increased amounts of acyl-CoA esters, compounds that interfere with the beta-oxidation of fatty acids (Vance et al, Epilepsia 35(5):1016-1022 (1994)). Administration of valproate to rats has also been shown to cause enhanced excretion of dicarboxylic acids, a sign of impaired mitochondrial beta-oxidation. Other metabolic effects include hypoglycemia, hyperammoneniia, decreased levels of beta-hydroxybutyrate and carnitine and decreased activities of acyl-CoA dehydrogenases, enzymes involved in fatty acid oxidation. mRNA levels of genes involved in fatty acid oxidation, however, such as the short-, medium- and long-chain acyl-CoA dehydrogenases, were found to have increased (Kibayasbi et al, Pediatrlnt 41(l):52-60 (1999)).
[0076] Wy-14643, a tumor-inducing compound that acts in the liver, has been used to study the genetic profile of cells during the various stages of carcinogenic development, with a view toward developing strategies for detecting, diagnosing and treating cancers (Rockett et al, Toxicology 144(l-3):13-29 (2000)). In contrast to other carcinogens, Wy-14643 does not mutate DNA directly, instead, it acts on the peroxisome proliferator activated receptor-alpha (PPARalpha), as well as on other signaling pathways that regulate growth (Johnson et al, J Steroid Biochem Mol Biol 77(1): 59-71 (2001)). The effect is elevated and sustained cell replication, accompanied by a decrease in apoptosis (Rusyn et al, Carcinogenesis 21(12):2141-2145 (2000)). These authors (Rusyn et al.) noted an increase in the expression of enzymes that repair DNA by base excision, but no increased expression of enzymes that do not repair oxidative damage to DNA. In a study on rodents, Johnson et al noted that Wy- 14643 inhibited liver-X-receptor-mediated transcription in a dose-dependent manner, as well as de novo sterol synthesis.
[0077] In experiments with mouse liver cells (Peters et al. , Carcinogenesis 19(11): 1989- 1994 (1998)), exposure to Wy-14643 produced increased levels of acyl CoA oxidase and proteins involved in cell proliferation: CDK-1, 2 and 4, PCNA and c-myc. Elevated levels may be caused by accelerated transcription that is mediated directly or indirectly by PPARalpha. It is likely that the carcinogenic properties of peroxisome prohferators are due to the PPARalpha-dependent changes in levels of cell cycle regulatory proteins. [0078] Another study on rodents (Keller et al, Biochim Biophys Acta 1102(2):237-244 (1992)) showed that Wy-14643 was capable of uncoupling oxidative phosphorylation in rat liver mitochondria. Rates of urea synthesis from ammonia and bile flow, two energy- dependent processes, were reduced, indicating that the energy supply for these processes was disrupted as a result of cellular exposure to the toxin. Wy-14643 has also been shown to activate nuclear factor kappaB, NADPH oxidase and superoxide production in Kupffer cells (Rusyn et al, Cancer Res 60(17):4798-4803 (2000)). NADPH oxidase is known to induce mitogens, which cause proliferation of liver cells.
Toxicity Identification, Prediction and Modeling
[0079] The genes and gene expression information, as well as the portfolios and subsets of the genes provided in Tables 1-5XX may be used to predict or identify at least one toxic effect, including the hepatotoxicity of a test or unknown compound. As used, herein, at least one toxic effect includes, but is not limited to, a detrimental change in the physiological status of a cell or organism. The response may be, but is not required to be, associated with a particular pathology, such as tissue necrosis. Accordingly, the toxic effect includes effects at the molecular and cellular level. Hepatotoxicity is an effect as used herein and includes, but is not limited to, genotoxic and non-genotoxic carcinogenesis, cholestasis, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, and steatosis with hepatitis. In addition, hepatoxicity includes the effect of direct- acting agents (such as metformin, rosiglitazone and dexamethasone), which are pharmaceuticals that act in the liver, but are not considered toxic to the liver. Exposure to these agents results in altered gene expression profiles. As used herein, a gene expression profile comprises any quantitative representation of the expression of at least one mRNA species in a cell sample or population and includes profiles made by various methods such as differential display, PCR, hybridization analysis, etc.
[0080] In general, assays to predict the toxicity or hepatotoxicity of a test agent (or compound or multi-component composition) comprise the steps of exposing a cell population to the test compound, assaying or measuring the level of relative or absolute gene expression of one or more of the genes in Tables 5A-5XX and comparing the identified expression level(s) to the expression levels disclosed in the Tables and database(s) disclosed herein. Assays may include the measurement of the expression levels of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 50, 75, 100, 200, 300, 400, 500, 1000 or more genes from Tables 5A-5XX to create multi-gene expression profiles. In some embodiments, all or substantially all of the genes of Tables 5A-5XX may be used to predict toxicity, etc. In other embodiments, the genes or subsets of the genes for each individual toxin model, for instance, the genes of Table 5 A, may be used. An "adequate amount" of the data of Tables 5A-5XX refers to an amount of information that allows toxicity identification or prediction (typically 2 or more genes). "Substantially" or nearly all of the data in the tables refers to at least about 80% of the data for an individual model.
[0081] In the methods of the invention, the gene expression level for a gene or genes induced by the test agent, compound or compositions may be comparable to the levels found in the Tables or databases disclosed herein if the expression level varies within a factor of about 2, about 1.5 or about 1.0 fold. In some cases, the expression levels are comparable if the agent induces a change in the expression of a gene in the same direction (e.g., up or down) as a reference toxin. "Comparing" may comprise determining the relationship of the database information to the sample gene expression profile with or without application of an algorithm to the results, differences or similarities between the two.
[0082] The cell population that is exposed to the test agent, compound or composition may be exposed in vitro or in vivo. For instance, cultured or freshly isolated hepatocytes, in particular rat hepatocytes, may be exposed to the agent under standard laboratory and cell culture conditions. In another assay format, in vivo exposure may be accomplished by administration of the agent to a living animal, for instance a laboratory rat. [0083] Procedures for designing and conducting toxicity tests in in vitro and in vivo systems are well known, and are described in many texts on the subject, such as Loomis et al, Loomis's Esstentials of Toxicology. 4th Ed.. Academic Press, New York, 1996; Echobichon, The Basics of Toxicity Testing. CRC Press, Boca Raton, 1992; Frazier, editor, In Vitro Toxicity Testing. Marcel Dekker, New York, 1992; and the like.
[0084] In in vitro toxicity testing, two groups of test organisms are usually employed: One group serves as a control and the other group receives the test compound in a single dose (for acute toxicity tests) or a regimen of doses (for prolonged or chronic toxicity tests). Because, in some cases, the extraction of tissue as called for in the methods of the invention requires sacrificing the test animal, both the control group and the group receiving compound must be large enough to permit removal of animals for sampling tissues, if it is desired to observe the dynamics of gene expression through the duration of an experiment. [0085] In setting up a toxicity study, extensive guidance is provided in the literature for selecting the appropriate test organism for the compound being tested, route of administration, dose ranges, and the like. Water or physiological saline (0.9% NaCl in water) is the solute of choice for the test compound since these solvents permit administration by a variety of routes. When this is not possible because of solubility limitations, vegetable oils such as com oil or organic solvents such as propylene glycol may be used. [0086] Regardless of the route of administration, the volume required to administer a given dose is limited by the size of the animal that is used. It is desirable to keep the volume of each dose uniform within and between groups of animals. When rats or mice are used, the volume administered by the oral route generally should not exceed about 0.005 ml per gram of animal. Even when aqueous or physiological saline solutions are used for parenteral injection the volumes that are tolerated are limited, although such solutions are ordinarily thought of as being innocuous. The intravenous LD50 of distilled water in the mouse is approximately 0.044 ml per gram and that of isotonic saline is 0.068 ml per gram of mouse. In some instances, the route of administration to the test animal should be the same as, or as similar as possible to, the route of administration of the compound to man for therapeutic pmposes.
[0087] When a compound is to be administered by inhalation, special techniques for generating test atmospheres are necessary. The methods usually involve aerosolization or nebulization of fluids containing the compound. If the agent to be tested is a fluid that has an appreciable vapor pressure, it may be administered by passing air through the solution under controlled temperature conditions. Under these conditions, dose is estimated from the volume of air inhaled per unit time, the temperature of the solution, and the vapor pressure of the agent involved. Gases are metered from reservoirs. When particles of a solution are to be administered, unless the particle size is less than about 2 μm the particles will not reach the terminal alveolar sacs in the lungs. A variety of apparatuses and chambers are available to perform studies for detecting effects of irritant or other toxic endpoints when they are admimstered by inhalation. The prefened method of administering an agent to animals is via the oral route, either by intubation or by incoφorating the agent in the feed. [0088] When the agent is exposed to cells in vitro or in cell culture, the cell population to be exposed to the agent may be divided into two or more subpopulations, for instance, by dividing the population into two or more identical aliquots. In some prefened embodiments of the methods of the invention, the cells to be exposed to the agent are derived from liver tissue. For instance, cultured or freshly isolated rat hepatocytes may be used. [0089] The methods of the invention may be used generally to predict at least one toxic response, and, as described in the Examples, may be used to predict the likelihood that a compound or test agent will induce various specific liver pathologies, such as genotoxic and non-genotoxic carcinogenesis, cholestasis, direct action toxicity, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, steatosis with hepatitis, or other pathologies associated with at least one of the toxins herein described. The methods of the invention may also be used to determine the similarity of a toxic response to one or more individual compounds. In addition, the methods of the invention may be used to predict or elucidate the potential cellular pathways influenced, induced or modulated by the compound or test agent due to the similarity of the expression profile compared to the profile induced by a known toxin (see Tables 5A-5G, 5J, 5K, 5M-5S, 5U-5Y, 5AA-5EE, 5HH-5JJ, 5MM, 5OO, 5PP and 5SS-5XX). Further, the link between a specific liver pathology that is the result of exposure to a toxin and a specific gene expression profile allows for distinction of the genes in Tables 5A-5XX as markers of liver toxicity.
Diagnostic Uses for the Toxicity Markers
[0090] As described above, the genes and gene expression information or portfolios of the genes with their expression information as provided in Tables 5A-5XX may be used as diagnostic markers for the prediction or identification of the physiological state of tissue or cell sample that has been exposed to a compound or to identify or predict the toxic effects of a compound or agent. For instance, a tissue sample such as a sample of peripheral blood cells or some other easily obtainable tissue sample may be assayed by any of the methods described above, and the expression levels from a gene or genes from Tables 5A-5XX may be compared to the expression levels found in tissues or cells exposed to the toxins described herein. These methods may result in the diagnosis of a physiological state in the cell or may be used to identify the potential toxicity of a compound, for instance a new or unknown compound or agent. The comparison of expression data, as well as available sequence or other information may be done by researcher or diagnostician or may be done with the aid of a computer and databases as described below.
[0091] hi another format, the levels of a gene(s) of Tables 5A-5XX, its encoded protein(s), or any metabolite produced by the encoded protein may be monitored or detected in a sample, such as a bodily tissue or fluid sample to identify or diagnose a physiological state of an organism. Such samples may include any tissue or fluid sample, including urine, blood and easily obtainable cells such as peripheral lymphocytes.
Use of the Markers for Monitoring Toxicity Progression
[0092] As described above, the genes and gene expression information provided in Tables 5A-5XX may also be used as markers for the monitoring of toxicity progression, such as that found after initial exposure to a drug, drug candidate, toxin, pollutant, etc. For instance, a tissue or cell sample may be assayed by any of the methods described above, and the expression levels from a gene or genes from Tables 5A-5XX may be compared to the expression levels found in tissue or cells exposed to the hepatotoxins described herein. The comparison of the expression data, as well as available sequence or other information may be done by researcher or diagnostician or may be done with the aid of a computer and databases.
Use of the Toxicity Markers for Drug Screening
[0093] According to the present invention, the genes identified in Tables 5A-5XX may be used as markers or drag targets to evaluate the effects of a candidate drug, chemical compound or other agent on a cell or tissue sample. The genes may also be used as drag targets to screen for agents that modulate their expression and/or activity. In various formats, a candidate drug or agent can be screened for the ability to simulate the transcription or expression of a given marker or markers or to down-regulate or counteract the transcription or expression of a marker or markers. According to the present invention, one can also compare the specificity of a drug's effects by looking at the number of markers which the drag induces and comparing them. More specific drags will have less transcriptional targets. Similar sets of markers identified for two drags may indicate a similarity of effects. As used herein, an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down-regulating expression of the nucleic acid in a cell. [0094] Assays to monitor the expression of a marker or markers as defined in Tables 5 A- 5XX may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention.
[0095] In one assay format, microanays containing probes to one, two or more genes from Tables 5 A-5XX may be used to directly monitor or detect changes in gene expression in the treated or exposed cell. Cell lines, tissues or other samples are first exposed to a test agent and in some instances, a known toxin, and the detected expression levels of one or more, or preferably 2 or more of the genes of Tables 5A-5XX are compared to the expression levels of those same genes exposed to a known toxin alone. Compounds that modulate the expression patterns of the known toxin(s) would be expected to modulate potential toxic physiological effects in vivo. The genes in Tables 5 A-5XX are particularly appropriate marks in these assays as they are differentially expressed in cells upon exposure to a known hepatotoxin. [0096] In another format, cell lines that contain reporter gene fusions between the open reading frame and/or the transcriptional regulatory regions of a gene in Tables 5A-5XX and any assayable fusion partner may be prepared. Numerous assayable fusion partners are known and readily available including the firefly luciferase gene and the gene encoding chloramphenicol acetyltransferase (Alam et al, Anal Biochem 188:245-254 (1990)). Cell lines containing the reporter gene fusions are then exposed to the agent to be tested under appropriate conditions and time. Differential expression of the reporter gene between samples exposed to the agent and control samples identifies agents which modulate the expression of the nucleic acid.
[0097] Additional assay formats may be used to monitor the ability of the agent to modulate the expression of a gene identified in Tables 5 A-5XX. For instance, as described above, mRNA expression may be monitored directly by hybridization of probes to the nucleic acids of the invention. Cell lines are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standard procedures such those disclosed in Sambrook et al (Molecular Cloning: A Laboratory Manual. 3d Ed.. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2001).
[0098] In another assay format, cells or cell lines are first identified which express the gene products of the invention physiologically. Cell and/or cell lines so identified would be expected to comprise the necessary cellular machinery such that the fidelity of modulation of the transcriptional apparatus is maintained with regard to exogenous contact of agent with appropriate surface transduction mechanisms and/or the cytosolic cascades. Further, such cells or cell lines may be transduced or transfected with an expression vehicle (e.g., a plasmid or viral vector) construct comprising an operable non-translated 5 '-promoter containing end of the stractural gene encoding the gene products of Tables 5A-5XX fused to one or more antigenic fragments or other detectable markers, which are peculiar to the instant gene products, wherein said fragments are under the transcriptional control of said promoter and are expressed as polypeptides whose molecular weight can be distinguished from the naturally occurring polypeptides or may further comprise an immunologically distinct or other detectable tag. Such a process is well known in the art (see Sambrook et al, supra). [0099] Cells or cell lines transduced or transfected as outlined above are then contacted with agents under appropriate conditions; for example, the agent comprises a pharmaceutically acceptable excipient and is contacted with cells comprised in an aqueous physiological buffer such as phosphate buffered saline (PBS) at physiological pH, Eagles balanced salt solution (BSS) at physiological pH, PBS or BSS comprising serum or conditioned media comprising PBS or BSS and/or serum incubated at 37°C. Said conditions may be modulated as deemed necessary by one of skill in the art. Subsequent to contacting the cells with the agent, said cells are disrupted and the polypeptides of the lysate are fractionated such that a polypeptide fraction is pooled and contacted with an antibody to be further processed by immunological assay (e.g., ELISA, immunoprecipitation or Western blot). The pool of proteins isolated from the agent-contacted sample is then compared with the control samples (no exposure and exposure to a known toxin) where only the excipient is contacted with the cells and an increase or decrease in the immunologically generated signal from the agent-contacted sample compared to the contiol is used to distinguish the effectiveness and/or toxic effects of the agent.
[00100] Another embodiment of the present invention provides methods for identifying agents that modulate at least one activity of a protein(s) encoded by the genes in Tables 5 A- 5XX. Such methods or assays may utilize any means of monitoring or detecting the desired activity.
[00101] In one format, the relative amounts of a protein (Tables 5A-5XX) between a cell population that has been exposed to the agent to be tested compared to an un-exposed control cell population and a cell population exposed to a known toxin may be assayed. In this format, probes such as specific antibodies are used to monitor the differential expression of the protein in the different cell populations. Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time. Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe, such as a specific antibody. [00102] Agents that are assayed in the above methods can be randomly selected or rationally selected or designed. As used herein, an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of the a protein of the invention alone or with its associated substiates, binding partners, etc. An example of randomly selected agents is the use a chemical library or a peptide combinatorial library, or a growth broth of an organism.
[00103] As used herein, an agent is said to be rationally selected or designed when the agent is chosen on a nonrandom basis which takes into account the sequence of the target site and/or its conformation in connection with the agent's action. Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites. For example, a rationally selected peptide agent can be a peptide whose amino acid sequence is identical to or a derivative of any functional consensus site.
[00104] The agents of the present invention can be, as examples, peptides, small molecules, vitamin derivatives, as well as carbohydrates. Dominant negative proteins, DNAs encoding these proteins, antibodies to these proteins, peptide fragments of these proteins or mimics of these proteins may be introduced into cells to affect function. "Mimic" used herein refers to the modification of a region or several regions of a peptide molecule to provide a structure chemically different from the parent peptide but topographically and functionally similar to the parent peptide (see G . Grant in: Molecular Biology and Biotechnology. Meyers, ed., pp. 659-664, VCH Publishers, New York, 1995). A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
Nucleic Acid Assay Formats
[00105] The genes identified as being differentially expressed upon exposure to a known hepatotoxin (Tables 5A-5XX) may be used in a variety of nucleic acid detection assays to detect or quantititate the expression level of a gene or multiple genes in a given sample. The genes described in Tables 5 A-5XX may also be used in combination with one or more additional genes whose differential expression is associate with toxicity in a cell or tissue. In prefened embodiments, the genes in Tables 5A-5XX may be combined with one or more of the genes described in prior and related applications 60/353,171; 60/363,534; 60/371,135; 60/371,134; 60/370,248; 60/371,150; 60/371,413; 60/373,601; 60/374,139; 60/394,253; 60/378,652; 60/373,602; 60/378,653; 60/378,665; 60/378,370; 60/394,230; 60/407,688;
09/917,800; 10/060,087; PCT/US03/ , entitled "Molecular Hepatotoxicology
Modeling," filed January 31, 2003; and PCT/USOl/23872, all of which are incoφorated by reference on page 1 of this application.
[00106] Any assay format to detect gene expression may be used. For example, traditional Northern blotting, dot or slot blot, nuclease protection, primer directed amplification, RT- PCR, semi- or quantitative PCR, branched-chain DNA and differential display methods may be used for detecting gene expression levels. Those methods are useful for some embodiments of the invention. In cases where smaller numbers of genes are detected, amplification based assays may be most efficient. Methods and assays of the invention, however, may be most efficiently designed with hybridization-based methods for detecting the expression of a large number of genes.
[00107] Any hybridization assay format may be used, including solution-based and solid support-based assay formats. Solid supports containing oligonucleotide probes for differentially expressed genes of the invention can be filters, polyvinyl chloride dishes, particles, beads, microparticles or silicon or glass based chips, etc. Such chips, wafers and hybridization methods are widely available, for example, those disclosed by Beattie (WO 95/11755).
[00108] Any solid surface to which oligonucleotides can be bound, either directly or indirectly, either covalently or non-covalently, can be used. A prefened solid support is a high density anay or DNA chip. These contain a particular oligonucleotide probe in a predetermined location on the anay. Each predetermined location may contain more than one molecule of the probe, but each molecule within the predetermined location has an identical sequence. Such predetermined locations are termed features. There may be, for example, from 2, 10, 100, 1000 to 10,000, 100,000 or 400,000 or more of such features on a single solid support. The solid support, or the area within which the probes are attached may be on the order of about a square centimeter. Probes conesponding to the genes of Tables 5 A-5XX or from the related applications described above may be attached to single or multiple solid support structures, e.g., the probes may be attached to a single chip or to multiple chips to comprise a chip set.
[00109] Oligonucleotide probe anays for expression monitoring can be made and used according to any techniques known in the art (see for example, Lockhart et al., Nat Biotechnol 14:1675-1680 (1996); McGall et al, Proc Nat Acad Sci USA 93:13555-13460 (1996)). Such probe arrays may contain at least two or more oligonucleotides that are complementary to or hybridize to two or more of the genes described in Tables 5A-5XX. For instance, such anays may contain oligonucleotides that are complementary or hybridize to at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 70, 100 or more the genes described herein. Prefened anays contain all or nearly all of the genes listed in Tables 5A-5XX, 'or individually, the gene sets of Tables 5A-5XX. In a prefened embodiment, anays are constructed that contain oligonucleotides to detect all or nearly all of the genes in any one of or all of Tables 5A-5XX on a single solid support substrate, such as a chip. [00110] The sequences of the expression marker genes of Tables 5A-5XX are in the public databases. Table 1 provides the GenBank Accession Number, SEQ ID NO: and GLGC ID No. (Gene Logic reference no.) for each of the sequences (see www.ncbi.nlm.nih.gov/), while Table 2 provides identification information for the human homologues of the genes of Tables 1 and 5A-5XX. Table 3 identifies the metabolic pathways in which the genes of Tables 1 and 5A-5XX are believed to function. Table 4 defines the model codes used in Tables 1, 2, 3 and 5A-5XX. The sequences of the genes in GenBank are expressly herein incoφorated by reference in their entirety as of the filing date of this application, as are related sequences, for instance, sequences from the same gene of different lengths, variant sequences, polymoφhic sequences, genomic sequences of the genes and related sequences from different species, including the human counteφarts, where appropriate. These sequences may be used in the methods of the invention or may be used to produce the probes and anays of the invention. In some embodiments, the genes in Tables 5 A-5XX that conespond to the genes or fragments previously associated with a toxic response may be excluded from the Tables. [00111] As described above, in addition to the sequences of the GenBank Accession Nos. and GLGC ID Nos. disclosed in the Tables 5A-5XX, sequences such as naturally occurring variant or polymoφhic sequences may be used in the methods and compositions of the invention. For instance, expression levels of various allelic or homologous forms of a gene disclosed in the Tables 5A-5XX may be assayed. Any and all nucleotide variations that do not alter the functional activity of a gene listed in the Tables 5 A-5XX , including all naturally occurring allelic variants of the genes herein disclosed, may be used in the methods and to make the compositions (e.g., anays) of the invention.
[00112] Probes based on the sequences of the genes described above may be prepared by any commonly available method. Oligonucleotide probes for screening or assaying a tissue or cell sample are preferably of sufficient length to specifically hybridize only to appropriate, complementary genes or transcripts. Typically the oligonucleotide probes will be at least about 10, 12, 14, 16, 18, 20 or 25 nucleotides in length. In some cases, longer probes of at least 30, 40, or 50 nucleotides will be desirable.
[00113] As used herein, oligonucleotide sequences that are complementary to one or more of the genes described in Tables 5A-5XX refer to oligonucleotides that are capable of hybridizing under stringent conditions to at least part of the nucleotide sequences of said genes. Such hybridizable oligonucleotides will typically exhibit at least about 75% sequence identity at the nucleotide level to said genes, preferably about 80% or 85% sequence identity or more preferably about 90% or 95% or more sequence identity to said genes. [00114] "Bind(s) substantially" refers to complementary hybridization between a probe nucleic acid and a target nucleic acid and embraces minor mismatches that can be accommodated by reducing the stringency of the hybridization media to achieve the desired detection of the target polynucleotide sequence.
[00115] The terms "background" or "background signal intensity" refer to hybridization signals resulting from non-specific binding, or other interactions, between the labeled target nucleic acids and components of the oligonucleotide anay (e.g., the oligonucleotide probes, control probes, the anay substrate, etc.). Background signals may also be produced by intrinsic fluorescence of the anay components themselves. A single background signal can be calculated for the entire anay, or a different background signal may be calculated for each target nucleic acid. In a prefened embodiment, background is calculated as the average hybridization signal intensity for the lowest 5% to 10%) of the probes in the anay, or, where a different background signal is calculated for each target gene, for the lowest 5% to 10% of the probes for each gene. Of course, one of skill in the art will appreciate that where the probes to a particular gene hybridize well and thus appear to be specifically binding to a target sequence, they should not be used in a background signal calculation. Alternatively, background may be calculated as the average hybridization signal intensity produced by hybridization to probes that are not complementary to any sequence found in the sample (e.g. probes directed to nucleic acids of the opposite sense or to genes not found in the sample such as bacterial genes where the sample is mammalian nucleic acids). Background can also be calculated as the average signal intensity produced by regions of the anay that lack any probes at all.
[00116] The phrase "hybridizing specifically to" refers to the binding, duplexing, or hybridizing of a molecule substantially to or only to a particular nucleotide sequence or sequences under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.
[00117] Assays and methods of the invention may utilize available formats to simultaneously screen at least about 100, preferably about 1000, more preferably about 10,000 and most preferably about 1,000,000 different nucleic acid hybridizations. [00118] As used herein a "probe" is defined as a nucleic acid, capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, usually through complementary base pairing, usually through hydrogen bond formation. As used herein, a probe may include natural (t.e., A, G, U, C, or T) or modified bases (7- deazaguanosine, inosine, etc.). In addition, the bases in probes may be joined by a linkage other than a phosphodiester bond, so long as it does not interfere with hybridization. Thus, probes may be peptide nucleic acids in which the constituent bases are joined by peptide bonds rather than phosphodiester linkages.
[00119] The term "perfect match probe" refers to a probe that has a sequence that is perfectly complementary to a particular target sequence. The test probe is typically perfectly complementary to a portion (subsequence) of the target sequence. The perfect match (PM) probe can be a "test probe", a "normalization, control" probe, an expression level control probe and the like. A perfect match control or perfect match probe is, however, distinguished from a "mismatch control" or "mismatch probe."
[00120] The terms "mismatch control" or "mismatch probe" refer to a probe whose sequence is deliberately selected not to be perfectly complementary to a particular target sequence. For each mismatch (MM) contiol in a high-density anay there typically exists a conesponding perfect match (PM) probe that is perfectly complementary to the same particular target sequence. The mismatch may comprise one or more bases.
[00121] While the mismatch(s) may be located anywhere in the mismatch probe, terminal mismatches are less desirable as a terminal mismatch is less likely to prevent hybridization of the target sequence. In a particularly prefened embodiment, the mismatch is located at or near the center of the probe such that the mismatch is most likely to destabilize the duplex with the target sequence under the test hybridization conditions. [00122] The term "stringent conditions" refers to conditions under which a probe will hybridize to its target subsequence, but with only insubstantial hybridization to other sequences or to other sequences such that the difference may be identified. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. Generally, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
[00123] Typically, stringent conditions will be those in which the salt concentration is at least about 0.01 to 1.0 M Na+ ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes (e.g., 10 to 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. [00124] The "percentage of sequence identity" or "sequence identity" is determined by comparing two optimally aligned sequences or subsequences over a comparison window or span, wherein the portion of the polynucleotide sequence in the comparison window may optionally comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical submit (e.g. nucleic acid base or amino acid residue) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. Percentage sequence identity when calculated using the programs GAP or BESTFIT (see below) is calculated using default gap weights.
Probe design
[00125] One of skill in the art will appreciate that an enormous number of anay designs are suitable for the practice of this invention. The high density anay will typically include a number of test probes that specifically hybridize to the sequences of interest. Probes may be produced from any region of the genes identified in the Tables and the attached representative sequence listing. In instances where the gene reference in the Tables is an EST, probes may be designed from that sequence or from other regions of the conesponding full-length transcript that may be available in any of the sequence databases, such as those herein described. See WO 99/32660 for methods of producing probes for a given gene or genes. In addition, any available software may be used to produce specific probe sequences, including, for instance, software available from Molecular Biology Insights, Olympus Optical Co. and Biosoft International. In a prefened embodiment, the anay will also include one or more control probes.
[00126] High density anay chips of the invention include "test probes." Test probes may be oligonucleotides that range from about 5 to about 500, or about 7 to about 50 nucleotides, more preferably from about 10 to about 40 nucleotides and most preferably from about 15 to about 35 nucleotides in length. In other particularly prefened embodiments, the probes are 20 or 25 nucleotides in length. In another prefened embodiment, test probes are double or single strand DNA sequences. DNA sequences are isolated or cloned from natural sources or amplified from natural sources using native nucleic acid as templates. These probes have sequences complementary to particular subsequences of the genes whose expression they are designed to detect. Thus, the test probes are capable of specifically hybridizing to the target nucleic acid they are to detect.
[00127] In addition to test probes that bind the target nucleic acid(s) of interest, the high density anay can contain a number of control probes. The control probes may fall into three categories refened to herein as 1) normalization controls; 2) expression level controls; and 3) mismatch controls.
[00128] Nonnalization controls are oligonucleotide or other nucleic acid probes that are complementary to labeled reference oligonucleotides or other nucleic acid sequences that are added to the nucleic acid sample to be screened. The signals obtained from the normalization controls after hybridization provide a contiol for variations in hybridization conditions, label intensity, "reading" efficiency and other factors that may cause the signal of a perfect hybridization to vary between anays. In a prefened embodiment, signals (e.g., fluorescence intensity) read from all other probes in the anay are divided by the signal (e.g., fluorescence intensity) from the control probes thereby normalizing the measurements. [00129] Virtually any probe may serve as a normalization control. However, it is recognized that hybridization efficiency varies with base composition and probe length. Prefened normalization probes are selected to reflect the average length of the other probes present in the anay, however, they can be selected to cover a range of lengths. The normalization control(s) can also be selected to reflect the (average) base composition of the other probes in the anay, however in a prefened embodiment, only one or a few probes are used and they are selected such that they hybridize well (i.e., no secondary structure) and do not match any target-specific probes.
[00130] Expression level controls are probes that hybridize specifically with constitutively expressed genes in the biological sample. Virtually any constitutively expressed gene provides a suitable target for expression level controls. Typically expression level control probes have sequences complementary to subsequences of constitutively expressed "housekeeping genes" including, but not limited to the β-actin gene, the glyceraldehyde-3- phosphate dehydrogenase (GADPH) gene, the transferrin receptor gene and the like. [00131] Mismatch controls may also be provided for the probes to the target genes, for expression level controls or for normalization controls. Mismatch controls are oligonucleotide probes or other nucleic acid probes identical to their conesponding test or control probes except for the presence of one or more mismatched bases. A mismatched base is a base selected so that it is not complementary to the conesponding base in the target sequence to which the probe would otherwise specifically hybridize. One or more mismatches are selected such that under appropriate hybridization conditions (e.g., stringent conditions) the test or control probe would be expected to hybridize with its target sequence, but the mismatch probe would not hybridize (or would hybridize to a significantly lesser extent) Prefened mismatch probes contain a central mismatch. Thus, for example, where a probe is a 20 mer, a conesponding mismatch probe will have the identical sequence except for a single base mismatch (e.g., substituting a G, a C or a T for an A) at any of positions 6 through 14 (the central mismatch).
[00132] Mismatch probes thus provide a control for non-specific binding or cross hybridization to a nucleic acid in the sample other than the target to which the probe is directed. For example, if the target is present the perfect match probes should be consistently brighter than the mismatch probes. In addition, if all central mismatches are present, the mismatch probes can be used to detect a mutation, for instance, a mutation of a gene in the accompanying Tables 5A-5XX. The difference in intensity between the perfect match and the mismatch probe provides a good measure of the concentration of the hybridized material.
Nucleic Acid Samples
[00133] Cell or tissue samples may be exposed to the test agent in vitro or in vivo. When cultured cells or tissues are used, appropriate mammalian liver extracts may also be added with the test agent to evaluate agents that may require biotransformation to exhibit toxicity. In a prefened format, primary isolates of animal or human hepatocytes which already express the appropriate complement of drag-metabolizing enzymes may be exposed to the test agent without the addition of mammalian liver extracts.
[00134] The genes which are assayed according to the present invention are typically in the form of mRNA or reverse transcribed mRNA. The genes may be cloned or not. The genes may be amplified or not. The cloning and/or amplification do not appear to bias the representation of genes within a population. In some assays, it may be preferable, however, to use polyA+ RNA as a source, as it can be used with less processing steps. [00135] As is apparent to one of ordinary skill in the art, nucleic acid samples used in the methods and assays of the invention may be prepared by any available method or process. Methods of isolating total mRNA are well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in detail in Chapter 3 of Laboratory Techniques in Biochemistry and Molecular Biology. Vol. 24, Hybridization With Nucleic Acid Probes: Theory and Nucleic Acid Probes, P. Tijssen, Ed., Elsevier Press, New York, 1993. Such samples include RNA samples, but also include cDNA synthesized from a mRNA sample isolated from a cell or tissue of interest. Such samples also include DNA amplified from the cDNA, and RNA transcribed from the amplified DNA. One of skill in the art would appreciate that it is desirable to inhibit or destroy RNase present in homogenates before homogenates are used.
[00136] Biological samples may be of any biological tissue or fluid or cells from any organism as well as cells raised in vitro, such as cell lines and tissue culture cells. Frequently the sample will be a tissue or cell sample that has been exposed to a compound, agent, drag, pharmaceutical composition, potential environmental pollutant or other composition. In some formats, the sample will be a "clinical sample" which is a sample derived from a patient. Typical clinical samples include, but are not limited to, sputum, blood, blood-cells (e.g., white cells), tissue or fine needle biopsy samples, urine, peritoneal fluid, and pleural fluid, or cells therefrom.
[00137] Biological samples may also include sections of tissues, such as frozen sections or formalin fixed sections taken for histological pmposes.
Forming High Density Arrays
[00138] Methods of forming high density anays of oligonucleotides with a minimal number of synthetic steps are known. The oligonucleotide analogue anay can be synthesized on a single or on multiple solid substiates by a variety of methods, including, but not limited to, light-directed chemical coupling, and mechanically directed coupling (see Pirrung, U.S. Patent No. 5,143,854).
[00139] In brief, the light-directed combinatorial synthesis of oligonucleotide anays on a glass surface proceeds using automated phosphoramidite chemistry and chip masking techniques. In one specific implementation, a glass surface is derivatized with a silane reagent containing a functional group, e.g., a hydroxyl or amine group blocked by a photolabile protecting group. Photolysis through a photolithogaphic mask is used selectively to expose functional groups which are then ready to react with incoming 5' photoprotected nucleoside phosphoramidites. The phosphoramidites react only with those sites which are illuminated (and thus exposed by removal of the photolabile blocking group). Thus, the phosphoramidites only add to those areas selectively exposed from the preceding step. These steps are repeated until the desired anay of sequences have been synthesized on the solid surface. Combinatorial synthesis of different oligonucleotide analogues at different locations on the anay is determined by the pattern of illumination during synthesis and the order of addition of coupling reagents.
[00140] In addition to the foregoing, additional methods which can be used to generate an anay of oligonucleotides on a single substiate are described in PCT Publication Nos. WO 93/09668 and WO 01/23614. High density nucleic acid anays can also be fabricated by depositing pre-made or natural nucleic acids in predetermined positions. Synthesized or natural nucleic acids are deposited on specific locations of a substrate by light directed targeting and oligonucleotide directed targeting. Another embodiment uses a dispenser that moves from region to region to deposit nucleic acids in specific spots.
Hybridization
[00141] Nucleic acid hybridization simply involves contacting a probe and target nucleic acid under conditions where the probe and its complementary target can form stable hybrid duplexes through complementary base pairing. See WO 99/32660. The nucleic acids that do not form hybrid duplexes are then washed away leaving the hybridized nucleic acids to be detected, typically through detection of an attached detectable label. It is generally recognized that nucleic acids are denatured by increasing the temperature or decreasing the salt concentration of the buffer containing the nucleic acids. Under low stringency conditions (e.g., low temperature and/or high salt) hybrid duplexes (e.g., DNA:DNA, RNA:RNA, or RNA:DNA) will form even where the annealed sequences are not perfectly complementary. Thus, specificity of hybridization is reduced at lower stringency. Conversely, at higher stringency (e.g., higher temperature or lower salt) successful hybridization tolerates fewer mismatches. One of skill in the art will appreciate that hybridization conditions may be selected to provide any degree of stringency.
[00142] In a prefened embodiment, hybridization is performed at low stringency, in this case in 6X SSPET at 37°C (0.005% Triton X-100), to ensure hybridization and then subsequent washes are performed at higher stringency (e.g., I X SSPET at 37°C) to eliminate mismatched hybrid duplexes. Successive washes may be performed at increasingly higher stringency (e.g., down to as low as 0.25 X SSPET at 37°C to 50°C) until a desired level of hybridization specificity is obtained. Stringency can also be increased by addition of agents such as formamide. Hybridization specificity may be evaluated by comparison of hybridization to the test probes with hybridization to the various controls that can be present (e.g., expression level control, normalization control, mismatch controls, etc.). [00143] In general, there is a tradeoff between hybridization specificity (stringency) and signal intensity. Thus, in a prefened embodiment, the wash is performed at the highest stringency that produces consistent results and that provides a signal intensity greater than approximately 10% of the background intensity. Thus, in a prefened embodiment, the hybridized array may be washed at successively higher stringency solutions and read between each wash. Analysis of the data sets thus produced will reveal a wash stringency above which the hybridization pattern is not appreciably altered and which provides adequate signal for the particular oligonucleotide probes of interest.
Signal Detection
[00144] The hybridized nucleic acids are typically detected by detecting one or more labels attached to the sample nucleic acids. The labels may be incoφorated by any of a number of means well known to those of skill in the art. See WO 99/32660.
Databases
[00145] The present invention includes relational databases containing sequence information, for instance, for the genes of Tables 5A-5XX, as well as gene expression information from tissue or cells exposed to various standard toxins, such as those herein described (see Tables 5A-5XX). Databases may also contain information associated with a given sequence or tissue sample such as descriptive information about the gene associated with the sequence information (see Tables 1, 2 and 3), or descriptive information concerning the clinical status of the tissue sample, or the animal from which the sample was derived. The database may be designed to include different parts, for instance a sequence database and a gene expression database. Methods for the configuration and construction of such databases and computer-readable media to which such databases are saved are widely available, for instance, see U.S. Patent No. 5,953,727, which is herein incoφorated by reference in its entirety.
[00146] The databases of the invention may be linked to an outside or external database such as GenBank (www.ncbi.nlm.nih.gov/entrez.index.html); KEGG (www.genome.ad.jp/kegg); SPAD (www.grt.kyushu-u.ac.jp/spad/index.html); HUGO (www.gene.uclac.uk/hugo); Swiss- Prot (www.expasy.ch.sprot); Prosite (www.expasy.ch/tools/scnpsitl.html); OMDM (www.ncbi.nlm.nih.gov/omim); LocusLink (www.ncbi.nlm.nih.gov/LocusLink/); RefSeq (www.ncbi.nlm.nih.gov/LocusLink/refseq.html) and GDB (www.gdb.org). In a prefened embodiment, as described in Tables 1-3, the external database is GenBank and the associated databases maintained by the National Center for Biotechnology Information (NCBI) (wivw. ncbi. nlm. nih.gov) .
[00147] Any appropriate computer platform, user interface, etc. may be used to perform the necessary comparisons between sequence information, gene expression information and any other information in the database or information provided as an input. For example, a large number of computer workstations are available from a variety of manufacturers, such has those available from Silicon Graphics. Client/server environments, database servers and networks are also widely available and appropriate platforms for the databases of the invention.
[00148] The databases of the invention may be used to produce, among other things, electronic Northerns that allow the user to determine the cell type or tissue in which a given gene is expressed and to allow determination of the abundance or expression level of a given gene in a particular tissue or cell.
[00149] The databases of the invention may also be used to present information identifying the expression level in a tissue or cell of a set of genes comprising one or more of the genes in Tables 5A-5XX, comprising the step of comparing the expression level of at least one gene in Tables 5A-5XX in a cell or tissue exposed to a test agent to the level of expression of the gene in the database. Such methods may be used to predict the toxic potential of a given compound by comparing the level of expression of a gene or genes in Tables 5A-5XX from a tissue or cell sample exposed to the test agent to the expression levels found in a control tissue or cell samples exposed to a standard toxin or hepatotoxin such as those herein described. Such methods may also be used in the drug or agent screening assays as described herein.
Kits
[00150] The invention further includes kits combining, in different combinations, high- density oligonucleotide arrays, reagents for use with the anays, protein reagents encoded by the genes of the Tables, signal detection and anay-processing instruments, gene expression databases and analysis and database management software described above. The kits may be used, for example, to predict or model the toxic response of a test compound, to monitor the progression of hepatic disease states, to identify genes that show promise as new drug targets and to screen known and newly designed drags as discussed above.
[00151] The databases packaged with the kits are a compilation of expression patterns from human or laboratory animal genes and gene fragments (conesponding to the genes of Tables 5A-5XX). In particular, the database software and packaged information that may contain the databases saved to a computer-readable medium include the expression results of Tables 5A-5XX that can be used to predict toxicity of a test agent by comparing the expression levels of the genes of Tables 5A-5XX induced by the test agent to the expression levels presented in Tables 5A-5XX. In another format, database and software information may be provided in a remote electronic format, such as a website, the address of which may be packaged in the kit.
[00152] The kits may used in the pharmaceutical industry, where the need for early drug testing is strong due to the high costs associated with drug development, but where bioinformatics, in particular gene expression informatics, is still lacking. These kits will reduce the costs, time and risks associated with traditional new drag screening using cell cultures and laboratory animals. The results of large-scale drug screening of pre-grouped patient populations, pharmacogenomics testing, can also be applied to select drugs with greater efficacy and fewer side-effects. The kits may also be used by smaller biotechnology companies and research institutes who do not have the facilities for performing such large- scale testing themselves.
[00153] Databases and software designed for use with use with microaπays is discussed in Balaban et al, U.S. Patent Nos. 6,229,911, a computer-implemented method for managing information, stored as indexed tables, collected from small or large numbers of microanays, and 6,185,561, a computer-based method with data mining capability for collecting gene expression level data, adding additional attributes and reformatting the data to produce answers to various queries. Chee et al, U.S. Patent No. 5,974,164, discloses a software- based method for identifying mutations in a nucleic acid sequence based on differences in probe fluorescence intensities between wild type and mutant sequences that hybridize to reference sequences.
[00154] Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples therefore, specifically point out the prefened embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure.
EXAMPLES Example 1: Identification of Toxicity Markers in Rat Hepatocytes
[00155] To evaluate their toxicity, the hepatotoxins alpha-naphthylisothiocyante (ANIT), acetaminophen (APAP), AY-25329, carbon tetrachloride, clofibrate, diclofenac, 17α- ethinylestradiol, hydrazine, indomethacin, lipopolysaccharide, lovastatin, methotrexate, tacrine, valproate and control compositions were administered to cultures of primary rat hepatocytes from male Sprague-Dawley rats at various time points using administration diluents, protocols and dosing regimes as previously described in the art and in the prior applications discussed above, as well as in Table 6. Laboratory protocols for the administration of the hepatotoxins amiodarone, carbamazepine, chloφromazine, CI-1000, CPA, diflunisal, DMN, gemfibrozil, imipramine, phenobarbital, tamoxifen, tetracycline and Wy-14643 also appear in Table 6. Identification of toxicity markers was perfonned by microanay analysis and by the AlamarBlue® assay, a classical measure of cytotoxicity. The AlamarBlue® assay was performed in triplicate.
[00156] The source of the primary rat hepatocytes was Sprague Dawley Outbred CD® Rats (CRL:CD®[SD] IGS BR, Charles River Laboratories). Hepatocyte cultures were obtained in 24-well matrigel coated plates for the AlamarBlue® assay (175,000 cells/cm2) or in T-75cm2 matrigel coated flasks for RNA isolation for microanay analysis (187,000 cells/cm2) . Primary rat hepatocytes were received the day after the cells were removed from the animals. After arrival, the cells, the cells were incubated overnight (~15hrs) before the toxin was added to the cultures. The vehicle used in the toxicity experiments was HIM culture medium (Hepatocyte Incubation Medium, In Vitro Technologies Cat. No. Z90009) containing 0.2% DMSO (Sigma Cat. No. D-5879). Toxin or vehicle was administered to hepatocyte cultures as follows. For each treatment, i.e., vehicle alone, vehicle + toxin at low dose, or vehicle + toxin at high dose, cells were harvested after 3, 6 and 24-hour incubations with the toxin solution or with the vehicle.
[00157] The AlamarBlue® assay was performed as follows, using only the 24-hour time point samples.
1. Primary rat hepatocyte cultures were prepared as described above in a matrigel-coated plates at 175,000 cells/cm2.
2. The culture medium (HIM) was removed from each well and replaced with 500 μl of fresh HIM following arrival of the cells, and the cells were incubated overnight (approximately 15hrs) at 37°C, 5% CO2. 3. The next day, the HIM was removed and 500 μl of the medium containing either vehicle or a dose of toxin was added.
4. Lysis solution was used as a negative control. 450 μl medium + 50 μl 9% Triton XI 00 were added to each of 3 wells containing cells, for a final Triton concentration of 1%.
5. The cells in all wells were incubated for 24 hours at 37°C, 5% CO2.
6. HDM medium was removed, and a solution containing 500 μl of fresh HIM medium + 50 μl AlamarBlue® (BioSource International, Inc., Cat. No. DAL1100) was added to each well.
7. The cells were incubated at 37°C, 5% CO2 for 2 hours.
8. 100 μl medium was removed from each well of the 24-well plate and added to a well of a 96-well plate. The fluorescence was measured using 544 nm as the excitation and 590 nm as the emission on a Molecular Devices, SpectraMax Gemini, Softmax pro 2.6.1. Alternatively, two absorbance readings can be measured for the oxidized (600nm) and the reduced (570nrn) form of AlamarBlue®. After obtaining absorbance readings, results were calculated according to the manufacturer's protocol provided in the product description.
9. The data were evaluated to determine whether or not the toxin reduced cell viability. If so, the dose of the toxin that reduced cell viability by ~ 10-20%) was determined.
Collection of RNA from Rat Hepatocytes
[00158] More than 10 cells are typically prepared for each sample. RNA was collected at 3, 6 and 24 hours following addition of the toxin according to the following procedure. [00159] The medium from the flasks was discarded, and the cells were washed once with 20 ml of warm (37°C) RPMI-1640 + lOmM HEPES medium (Life Technologies, Cat. No. 22400-089). 12 ml of Trizol (Life Technologies, Cat. No. 15596-018) was placed immediately into each T-75 flask. Each flask contained -10-20 million cells. The contents of each flask were mixed vigorously for one minute with a vortex mixer and then aspirated up and down 5 times with a pipette. The contents of each flask (~12 ml each) was collected into a 50 ml conical polypropylene tissue culture tube (Falcon), snap frozen in liquid nitrogen and stored at < -86° C.
[00160] Microanay sample preparation was conducted with minor modifications, following the protocols set forth in the Affymetrix GeneChip® Expression Analysis Manual. Frozen cells were ground to a powder using a Spex Certiprep 6800 Freezer Mill. Total RNA was extracted with Trizol (GibcoBRL) utilizing the manufacturer's protocol. The total RNA yield for each sample was 200-500 μg per 300 mg cells. mRNA was isolated using the Oligotex mRNA Midi kit (Qiagen) followed by ethanol precipitation. Double stranded cDNA was generated from mRNA using the Superscript Choice system (GibcoBRL). First strand cDNA synthesis was primed with a T7-(dT24) oligonucleotide. The cDNA was phenol- chloroform extracted and ethanol precipitated to a final concentration of 1 μg/ml. From 2 μg of cDNA, cRNA was synthesized using Ambion' s T7 MegaScript in vitro Transcription Kit. [00161] To biotin label the cRNA, nucleotides Bio-11-CTP and Bio-16-UTP (Enzo Diagnostics) were added to the reaction. Following a 37°C incubation for six hours, impurities were removed from the labeled cRNA following the RNeasy Mini kit protocol (Qiagen). cRNA was fragmented (fragmentation buffer consisting of 200 mM Tris-acetate, pH 8.1, 500 mM KOAc, 150 mM MgOAc) for thirty-five minutes at 94°C. Following the Affymetrix protocol, 55 μg of fragmented cRNA was hybridized on the Affymetrix rat anay set for twenty- four hours at 60 φm in a 45 °C hybridization oven. The chips were washed and stained with Streptavidin Phycoerythrin (SAPE) (Molecular Probes) in Affymetrix fluidics stations. To amplify staining, SAPE solution was added twice with an anti-streptavidin biotinylated antibody (Vector Laboratories) staining step in between. Hybridization to the probe arrays was detected by fluorometric scanning (Hewlett Packard Gene Anay Scanner). Data was analyzed using Affymetrix GeneChip® version 3.0 and Expression Data Mining Tool (EDMT) software (version 1.0), S-Plus, and the GeneExpress® software system.
[00162] Differential expression of genes between the toxin-exposed and control samples conesponding to patterns indicative of toxicity was determined using the following criteria. [00163] Table 1 discloses those genes that are differentially expressed upon exposure to the named toxins with their conesponding SEQ ID NOS:, GenBank Accession or RefSeq ID Nos., GLGC ID Nos. (internal Gene Logic identification nos.), gene names and Unigene Sequence Cluster titles. The metabolic pathways in which the genes of Table 1 function are indicated in Table 3, and the conesponding human homologues are given in Table 2. The model codes, identified in Table 4, represent the various toxicity or liver pathology states associated with differential expression of each gene, as well as the individual toxin types associated with differential expression of each gene.
[00164] Tables 5A-5XX disclose the summary statistics for each of the comparisons performed. Each of these tables contains a set of predictive genes and creates a model for predicting the hepatoxicity of an unknown, i.e., untested compound. Each gene is identified by its Gene Logic identification number and can be cross-referenced to a gene name and representative SEQ LD NO. in Table 1. For each comparison of gene expression levels between samples in the toxicity group ("Tox" samples, i.e., samples affected by exposure to a specific toxin) and samples in the non-toxicity group ("Non-tox" samples, i.e., samples not affected by exposure to that same specific toxin), the group mean for Tox samples is the mean signal intensity, as normalized for the various chip parameters that are being assayed. The Non-tox mean represents the mean signal intensity, as normalized for the various chip parameters that are being assayed, in samples other than those tieated with the high dose of the specific toxin. These samples were treated with a low dose of the specific toxin, or with vehicle alone, or with a different toxin. Tox samples were obtained from treated cells processed at the timepoint(s) indicated in the tables, while Non-tox samples were obtained from control cells processed at all time points in the experiments. For individual genes, an increase in the Tox group mean compared to the Non-tox group mean indicates up-regulation upon exposure to a toxin. Conversely, a decrease in the Tox group mean compared to the Non-tox group mean indicates down-regulation.
[00165] The mean values are derived from Average Difference (AveDiff) values for a particular gene, averaged across the conesponding samples. Each individual Average Difference value is calculated by integrating the intensity information from multiple probe pairs that are tiled for a particular fragment. The normalization multiplies each expression intensity for a given experiment (chip) by a global scaling factor. The intent of this normalization is to make comparisons of individual genes between chips possible. The scaling factor is calculated as follows:
1. From all the unnormalized expression values in the experiment, delete the largest 2% and smallest 2% of the values. That is, if the experiment yields 10,000 expression values, order the values and delete the smallest 200 and the largest 200.
2. Compute the trimmed mean, which is equal to the mean of the remaining values.
3. Compute the scale factor SF = 100/(trimmed mean)
[00166] The value of 100 used here is the standard target value used. Some AveDiff values may be negative due to the general noise involved in nucleic acid hybridization experiments. Although many conclusions can be made conesponding to a negative value on the GeneChip platform, it is difficult to assess the meaning behind the negative value for individual fragments. Our observations show that, although negative values are observed at times within the predictive gene set, these values reflect a real biological phenomenon that is highly reproducible across all the samples from which the measurement was taken. For this reason, those genes that exhibit a negative value are included in the predictive set. It should be noted that other platforms of gene expression measurement may be able to resolve the negative numbers for the conesponding genes. The predictive ability of each of those genes should extend across platforms, however. Each mean value is accompanied by the standard deviation for the mean.
[00167] The linear discriminant analysis score (discriminant score), as disclosed in the tables, measures the ability of each gene to predict whether or not a sample is toxic. The discriminant score is calculated by the following steps:
Calculation of a discriminant score
[00168] Let Xj represent the AveDiff values for a given gene across the Group 1 samples, i=l...n.
[00169] Let Yj represent the AveDiff values for a given gene across the Group 2 samples, i=l...t.
[00170] The calculations proceed as follows:
[00171] Calculate mean and standard deviation for Xj's and Yj's, and denote these by niχ, mγ, sχ,sγ.
[00172] For all Xi's and Yi's, evaluate the function f(z) = ((l/sγ)*exp( -.5*( (z-mγ)/sγ)2)) /
(((l/sY)*exp( -.5*( (z-mY)/sY)2)) +((l/sx)*exp( -.5*( (z-mx)/sx)2))).
[00173] The number of conect predictions, say P, is then the number of Yi's such that f(Yi)>.5 plus the number of X;'s such that f(Xj)<.5.
[00174] The discriminant score is then P/(n+t).
[00175] Linear discriminant analysis uses both the individual measurements of each gene and the calculated measurements of all combinations of genes to classify samples. For each gene, a weight is derived from the mean and standard deviation of the Tox and Non-tox sample groups. Every gene is multiplied by a weight and the sum of these values results in a collective discriminate score. This discriminant score is then compared against collective centioids of the Tox and Non-tox groups. These centroids are the average of all tox and nontox samples respectively. Therefore, each gene contributes to the overall prediction. This contribution is dependent on weights that are large positive or negative numbers if the relative distances between the Tox and Non-tox samples for that gene are large and small numbers if the relative distances are small. The discriminant score for each unknown sample and centroid values can be used to calculate a probability between zero and one as to the group in which the unknown sample belongs.
Example 2: General Toxicity Modeling
[00176] Samples were selected for grouping into Tox and Non-tox groups by examining each study individually with Principal Components Analysis (PCA) to determine which treatments had an observable response. Only sample groups where confidence of the tox- responding or non-tox-responding status (expression level affected by exposure to a specific toxin or expression level not affected by exposure to a specific toxin, respectively) was established were included in building a general toxicity prediction model. [00177] Linear discriminant models were generated to describe Tox and Non-tox samples. The top discriminant genes and/or EST's were used to determine toxicity by calculating each gene's contribution with homo and heteroscedastic treatment of variance and inclusion or exclusion of mutual information between genes. Prediction of samples within the database exceeded 80% true positives with a false positive rate of less than 5%>. It was determined that combinations of genes and/or EST's generally provided a better prediction than individual genes and that the more genes and/or EST used, the better the prediction. Although the prefened embodiment includes fifty or more genes, many pairings or larger combinations of genes and/or EST can work better than individual genes. All combinations of two or more genes from the selected list could be used to predict toxicity. These combinations could be selected by pairing in an agglomerate, divisive, or random approach. Further, as yet undetermined genes and/or EST's could be combined with individual or a set of genes and/or EST's described here to increase predictive ability. However, the genes and/or EST's described here would contribute most of the predictive ability to any such undetermined combinations.
[00178] Other variations on the above method can provide adequate predictive ability. These include selective inclusion of components via agglomerate, divisive, or random approaches or extraction of loading and combining them in agglomerate, divisive, or random approaches. Also the use of composite variables in logistic regression to determine classification of samples can also be accomplished with linear discriminate analysis, neural or Bayesian networks, or other forms of regression and classification based on categorical or continual dependent and independent variables. Example 3: Modeling Methods
[00179] The above modeling methods provide broad approaches of combining the expression of genes to predict sample toxicity. One could also provide no weight in a simple voting method or determine weights in a supervised or unsupervised method using agglomerate, divisive, or random approaches. All or selected combinations of genes may be combined in ordered, agglomerate, or divisive, supervised or unsupervised clustering algorithms with unknown samples for classification. Any form of conelation matrix may also be used to classify unknown samples. The spread of the group distribution and discriminate score alone provide enough information to enable a skilled person to generate all of the above types of models with accuracy that can exceed the discriminate ability of individual genes. Some examples of methods that could be used individually or in combination after transformation of data types include but are not limited to: Discriminant Analysis, Multiple Discriminant Analysis, logistic regression, multiple regression analysis, linear regression analysis, conjoint analysis, canonical conelation, hierarchical cluster analysis, k-means cluster analysis, self-organizing maps, multidimensional scaling, structural equation modeling, support vector machine determined boundaries, factor analysis, neural networks, bayesian classifications, and resampling methods.
Example 4: Grouping of Individual compound and Pathology Classes [00180] Samples were grouped into individual pathology classes based on known toxicological responses and observed clinical chemical and pathology measurements or into observable toxicity produced by a compound (Tables 5A-5XX). The top 10, 25, 50, 100 genes based on individual discriminate scores were used in a model to ensure that a combination of genes provided a better prediction than individual genes. As described above, all combinations of two or more genes from this list could potentially provide better prediction than individual genes when selected in any order or by ordered, agglomerate, divisive, or random approaches. In addition, combining these genes with other genes could provide better predictive ability, but most of this predictive ability would come from the genes listed herein.
[00181] A sample may be considered a Tox sample if it scores positive in any pathological or individual compound class represented here, or in any modeling method mentioned under general toxicology models, based on a combination of the sample's time point and dosage group in a study using an individual compound (with known or potentially toxic properties) by comparisons obtainable from the data. The pathological groupings and early and late phase models are prefened examples of all obtainable combinations of sample time and dose points. Most logical groupings with one or more genes and one or more sample dose and time points should produce better predictions of general toxicity, pathological specific toxicity, or similarity to a known toxin than individual genes.
[00182] Although the present invention has been described in detail with reference to examples above, it is understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims. All cited patents, patent applications and publications refened to in this application are herein incoφorated by reference in their entirety.
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
Figure imgf000330_0001
Figure imgf000331_0001
Figure imgf000332_0001
Figure imgf000333_0001
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000348_0001
Figure imgf000349_0001
Figure imgf000350_0001
Figure imgf000351_0001
Figure imgf000352_0001
Figure imgf000353_0001
Figure imgf000354_0001
Figure imgf000355_0001
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Figure imgf000361_0001
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000370_0001
Figure imgf000371_0001
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
Figure imgf000375_0001
Figure imgf000376_0001
Figure imgf000377_0001
Figure imgf000378_0001
Figure imgf000379_0001
Figure imgf000380_0001
Figure imgf000381_0001
Figure imgf000382_0001
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0001
Figure imgf000386_0001
Figure imgf000387_0001
Figure imgf000388_0001
Figure imgf000389_0001
Figure imgf000390_0001
Figure imgf000391_0001
Figure imgf000392_0001
Figure imgf000393_0001
Figure imgf000394_0001
Figure imgf000395_0001
Figure imgf000396_0001
Figure imgf000397_0001
Figure imgf000398_0001
Figure imgf000399_0001
Figure imgf000400_0001
Figure imgf000401_0001
Figure imgf000402_0001
Figure imgf000403_0001
Figure imgf000404_0001
Figure imgf000405_0001
Figure imgf000406_0001
Figure imgf000407_0001
Figure imgf000408_0001
Figure imgf000409_0001
Figure imgf000410_0001
Figure imgf000411_0001
Figure imgf000412_0001
Figure imgf000413_0001
Figure imgf000414_0001
Figure imgf000415_0001
Figure imgf000416_0001
Figure imgf000417_0001
Figure imgf000418_0001
Figure imgf000419_0001
Figure imgf000420_0001
Figure imgf000421_0001
Figure imgf000422_0001
Figure imgf000423_0001
Figure imgf000424_0001
Figure imgf000425_0001
Figure imgf000426_0001
Figure imgf000427_0001
Figure imgf000428_0001
Figure imgf000429_0001
Figure imgf000430_0001
Figure imgf000431_0001
Figure imgf000432_0001
Figure imgf000433_0001
Figure imgf000434_0001
Figure imgf000435_0001
Figure imgf000436_0001
Figure imgf000437_0001
Figure imgf000438_0001
Figure imgf000439_0001
Figure imgf000440_0001
Figure imgf000441_0001
Figure imgf000442_0001
Figure imgf000443_0001
Figure imgf000444_0001
Figure imgf000445_0001
Figure imgf000446_0001
Figure imgf000447_0001
Figure imgf000448_0001
Figure imgf000449_0001
Figure imgf000450_0001
Figure imgf000451_0001
Figure imgf000452_0001
Figure imgf000453_0001
Figure imgf000454_0001
Figure imgf000455_0001
Figure imgf000456_0001
Figure imgf000457_0001
Figure imgf000458_0001
Figure imgf000459_0001
Figure imgf000460_0001
Figure imgf000461_0001
Figure imgf000462_0001
Figure imgf000463_0001
Figure imgf000464_0001
Figure imgf000465_0001
Figure imgf000466_0001
Figure imgf000467_0001
Figure imgf000468_0001
Figure imgf000469_0001
Figure imgf000470_0001
Figure imgf000471_0001
Figure imgf000472_0001
Figure imgf000473_0001
Figure imgf000474_0001
Figure imgf000475_0001
Figure imgf000476_0001
Figure imgf000477_0001
Figure imgf000478_0001
Figure imgf000479_0001
Figure imgf000480_0001
Figure imgf000481_0001
Figure imgf000482_0001
Figure imgf000483_0001
Figure imgf000484_0001
Figure imgf000485_0001
Figure imgf000486_0001
Figure imgf000487_0001
Figure imgf000488_0001
Figure imgf000489_0001
Figure imgf000490_0001
Figure imgf000491_0001
Figure imgf000492_0001
Figure imgf000493_0001
Figure imgf000494_0001
Figure imgf000495_0001
Figure imgf000496_0001
Figure imgf000497_0001
Figure imgf000498_0001
Figure imgf000499_0001
Figure imgf000500_0001
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
Figure imgf000505_0001
Figure imgf000506_0001
Figure imgf000507_0001
Figure imgf000508_0001
Figure imgf000509_0001
Figure imgf000510_0001
Figure imgf000511_0001
Figure imgf000512_0001
Figure imgf000513_0001
Figure imgf000515_0001
Figure imgf000516_0001
Figure imgf000517_0001
Figure imgf000518_0001
Figure imgf000519_0001
Figure imgf000520_0001
Figure imgf000521_0001
Figure imgf000522_0001
Figure imgf000523_0001
Figure imgf000524_0001
Figure imgf000525_0001
Figure imgf000526_0001
Figure imgf000527_0001
Figure imgf000528_0001
Figure imgf000529_0001
Figure imgf000530_0001
Figure imgf000531_0001
Figure imgf000532_0001
Figure imgf000533_0001
Figure imgf000534_0001
Figure imgf000535_0001
Figure imgf000536_0001
Figure imgf000537_0001
Figure imgf000538_0001
Figure imgf000539_0001
Figure imgf000540_0001
Figure imgf000541_0001
Figure imgf000542_0001
Figure imgf000543_0001
Figure imgf000544_0001
Figure imgf000545_0001
Figure imgf000546_0001
Figure imgf000547_0001
Figure imgf000548_0001
Figure imgf000549_0001
Figure imgf000550_0001
Figure imgf000551_0001
Figure imgf000552_0001
Figure imgf000553_0001
Figure imgf000554_0001
Figure imgf000555_0001
Figure imgf000556_0001
Figure imgf000557_0001
Figure imgf000558_0001
Figure imgf000559_0001
Figure imgf000560_0001
Figure imgf000561_0001
Figure imgf000562_0001
Figure imgf000563_0001
Figure imgf000564_0001
Figure imgf000565_0001
Figure imgf000566_0001
Figure imgf000567_0001
Figure imgf000568_0001
Figure imgf000569_0001
Figure imgf000570_0001
Figure imgf000571_0001
Figure imgf000572_0001
Figure imgf000573_0001
Figure imgf000574_0001
Figure imgf000575_0001
Figure imgf000576_0001
Figure imgf000577_0001
Figure imgf000578_0001
Figure imgf000579_0001
Figure imgf000580_0001
Figure imgf000581_0001
Figure imgf000582_0001
Figure imgf000583_0001
Figure imgf000584_0001
Figure imgf000585_0001
Figure imgf000586_0001
Figure imgf000587_0001
Figure imgf000588_0001
Figure imgf000589_0001
Figure imgf000590_0001
Figure imgf000591_0001
Figure imgf000592_0001
Figure imgf000593_0001
Figure imgf000594_0001
Figure imgf000595_0001
Figure imgf000596_0001
Figure imgf000597_0001
Figure imgf000598_0001
Figure imgf000599_0001
Figure imgf000600_0001
Figure imgf000601_0001
Figure imgf000602_0001
Figure imgf000603_0001
Figure imgf000604_0001
Figure imgf000605_0001
Figure imgf000606_0001
Figure imgf000607_0001
Figure imgf000608_0001
Figure imgf000609_0001
Figure imgf000610_0001
Figure imgf000611_0001
Figure imgf000612_0001
Figure imgf000613_0001
Figure imgf000614_0001
Figure imgf000615_0001
Figure imgf000616_0001
Figure imgf000617_0001
Figure imgf000618_0001
Figure imgf000619_0001
Figure imgf000620_0001
Figure imgf000621_0001
Figure imgf000622_0001
Figure imgf000623_0001
Figure imgf000624_0001
Figure imgf000625_0001
Figure imgf000626_0001
Figure imgf000627_0001
Figure imgf000628_0001
Figure imgf000629_0001
Figure imgf000630_0001
Figure imgf000631_0001
Figure imgf000632_0001
Figure imgf000633_0001
Figure imgf000634_0001
Figure imgf000635_0001
Figure imgf000636_0001
Figure imgf000637_0001
Figure imgf000637_0002
Figure imgf000638_0001
Figure imgf000639_0001
Figure imgf000640_0001
Figure imgf000641_0001
Figure imgf000642_0001
Figure imgf000643_0001
Figure imgf000644_0001
Figure imgf000645_0001
Figure imgf000646_0001
Figure imgf000647_0001
Figure imgf000648_0001
Figure imgf000649_0001
Figure imgf000650_0001
Figure imgf000651_0001
Figure imgf000652_0001
Figure imgf000653_0001
Figure imgf000654_0001
Figure imgf000655_0001
Figure imgf000656_0001
Figure imgf000657_0001
Figure imgf000658_0001
Figure imgf000659_0001
Figure imgf000660_0001
Figure imgf000661_0001
Figure imgf000662_0001
Figure imgf000663_0001
Figure imgf000664_0001
Figure imgf000664_0002
Figure imgf000665_0001
Figure imgf000666_0001
Figure imgf000667_0001
Figure imgf000668_0001
Figure imgf000669_0001
Figure imgf000670_0001
Figure imgf000671_0001
Figure imgf000672_0001
Figure imgf000673_0001
Figure imgf000674_0001
Figure imgf000675_0001
Figure imgf000676_0001
Figure imgf000677_0001
Figure imgf000678_0001
Figure imgf000679_0001
Figure imgf000680_0001
Figure imgf000681_0001
Figure imgf000682_0001
Figure imgf000683_0001
Figure imgf000684_0001
Figure imgf000685_0001
Figure imgf000686_0001
Figure imgf000687_0001
Figure imgf000688_0001
Figure imgf000689_0001
Figure imgf000690_0001
Figure imgf000691_0001
Figure imgf000692_0001
Figure imgf000693_0001
Figure imgf000694_0001
Figure imgf000695_0001
Figure imgf000696_0001
Figure imgf000697_0001
Figure imgf000698_0001
Figure imgf000699_0001
Figure imgf000700_0001
Figure imgf000701_0001
Figure imgf000702_0001
Figure imgf000703_0001
Figure imgf000704_0001
Figure imgf000705_0001
Figure imgf000706_0001
Figure imgf000707_0001
Figure imgf000708_0001
Figure imgf000709_0001
Figure imgf000710_0001
Figure imgf000711_0001
Figure imgf000712_0001
Figure imgf000713_0001
Figure imgf000714_0001
Figure imgf000715_0001
Figure imgf000716_0001
Figure imgf000717_0001
Figure imgf000718_0001
Figure imgf000719_0001
Figure imgf000720_0001
Figure imgf000721_0001
Figure imgf000722_0001
Figure imgf000723_0001
Figure imgf000724_0001
Figure imgf000725_0001
Figure imgf000726_0001
Figure imgf000727_0001
Figure imgf000728_0001
Figure imgf000729_0001
Figure imgf000730_0001
Figure imgf000731_0001
Figure imgf000733_0001
Figure imgf000734_0001
Figure imgf000735_0001
Figure imgf000736_0001
Figure imgf000737_0001
Figure imgf000738_0001
Figure imgf000739_0001
Figure imgf000740_0001
Figure imgf000741_0001
Figure imgf000742_0001
Figure imgf000743_0001
Figure imgf000744_0001
Figure imgf000745_0001
Figure imgf000746_0001
Figure imgf000747_0001
Figure imgf000748_0001
Figure imgf000749_0001
Figure imgf000750_0001
Figure imgf000751_0001
Figure imgf000752_0001
Figure imgf000753_0001
Figure imgf000754_0001
Figure imgf000755_0001
Figure imgf000756_0001
Figure imgf000757_0001
Figure imgf000758_0001
Figure imgf000759_0001
Figure imgf000760_0001
Figure imgf000761_0001
Figure imgf000762_0001
Figure imgf000763_0001
Figure imgf000764_0001
Figure imgf000765_0001
Figure imgf000766_0001
Figure imgf000767_0001
Figure imgf000768_0001
Figure imgf000769_0001
Figure imgf000770_0001
Figure imgf000771_0001
Figure imgf000772_0001
Figure imgf000773_0001
Figure imgf000774_0001
Figure imgf000775_0001
Figure imgf000776_0001
Figure imgf000777_0001
Figure imgf000778_0001
Figure imgf000779_0001
Figure imgf000781_0001
Figure imgf000782_0001
Figure imgf000783_0001
Figure imgf000784_0001
Figure imgf000785_0001
Figure imgf000786_0001
Figure imgf000787_0001
Figure imgf000788_0001
Figure imgf000789_0001
Figure imgf000790_0001
Figure imgf000791_0001
Figure imgf000792_0001
Figure imgf000793_0001
Figure imgf000794_0001
Figure imgf000795_0001
Figure imgf000796_0001
Figure imgf000797_0001
Figure imgf000798_0001
Figure imgf000799_0001
Figure imgf000800_0001
Figure imgf000801_0001
Figure imgf000802_0001
Figure imgf000803_0001
Figure imgf000804_0001
Figure imgf000805_0001
Figure imgf000806_0001
Figure imgf000807_0001
Figure imgf000808_0001
Figure imgf000809_0001
Figure imgf000810_0001
Figure imgf000811_0001
Figure imgf000812_0001
Figure imgf000813_0001
Figure imgf000814_0001
Figure imgf000815_0001
Figure imgf000816_0001
Figure imgf000817_0001
Figure imgf000818_0001
Figure imgf000819_0001
Figure imgf000820_0001
Figure imgf000821_0001
Figure imgf000822_0001
Figure imgf000823_0001
Figure imgf000824_0001
Figure imgf000825_0001
Figure imgf000826_0001
Figure imgf000827_0001
Figure imgf000828_0001
Figure imgf000829_0001
Figure imgf000830_0001
Figure imgf000831_0001
Figure imgf000832_0001
Figure imgf000833_0001
Figure imgf000834_0001
Figure imgf000835_0001
Figure imgf000837_0001
Figure imgf000838_0001
Figure imgf000839_0001
Figure imgf000840_0001
Figure imgf000841_0001
Figure imgf000842_0001
Figure imgf000843_0001
Figure imgf000844_0001
Figure imgf000845_0001
Figure imgf000846_0001
Figure imgf000847_0001
Figure imgf000848_0001
Figure imgf000849_0001
Figure imgf000850_0001
Figure imgf000851_0001
Figure imgf000852_0001
Figure imgf000853_0001
Figure imgf000854_0001
Figure imgf000855_0001
Figure imgf000856_0001
Figure imgf000857_0001
Figure imgf000858_0001
Figure imgf000859_0001
Figure imgf000860_0001
Figure imgf000861_0001
Figure imgf000862_0001
Figure imgf000863_0001
Figure imgf000864_0001
Table 6- Laboratory protocols for administration of toxins to hepatocyte cultures and Results of AlamarBlue® cell viability assays
Toxin 3 hours 6 hours 24 hours low dose high dose low dose high dose low dose high dose
Amiodarone, ICN cat. 6uM 60uM 6uM 60uM 6uM 60uM no. 15353583 % viability by -100 -80-85 AlamarBlue test
Carbamazepine, O.OlmM ImM O.OlmM ImM O.OlmM ImM
Sigma cat. no. C-8981 % viability by -100 -90 AlamarBlue test
Chlorpromazine, 3uM 30uM 3uM 30uM 3uM 30uM
Sigma cat. no. C-8138 % viability by -100 -75 AlamarBlue test
CI-1000 25uM 250uM 25uM 250uM 25uM 250uM
% viability by -90 -75 AlamarBlue test
Cyproterone acetate 40uM 400uM 40uM 400uM 40uM 400uM
Sigma cat. no. C-3412 % viability by -100 -65-70 AlamarBlue test
Diflunisal, 30uM 300uM 30uM 300uM 30uM 300uM
Sigma cat. no. D-3281 % viability by -100 -85-90 AlamarBlue test
DMN, ImM lOOmM ImM lOOmM ImM lOOmM
Sigma cat. no. N-7756 % viability by -100 -80-85 AlamarBlue test
Gemfibrozil, 0.3mM 3mM 0.3mM 3mM 0.3mM 3mM
Sigma cat. no. G-9518 % viability by -100 -50 AlamarBlue test
Imipramine, 5uM 50uM 5uM 50uM 5uM 50uM
Sigma cat. no. 1-7379 % viability by -100 -85-90 AlamarBlue test Toxin 3 hours 6 hours 24 hours low dose high dose low dose high dose low dose high dose
Phenobarbital, 0.8mM 8mM 0.8mM 8mM 0.8mM 8mM
Sigma cat. no. P-5178 % viability by 400 >95 AlamarBlue test
Tamoxifen, 4uM 40uM 4uM 40uM 4uM 40uM
Sigma cat. no. T-9262 % viability by -100 -45 AlamarBlue test
Tetracycline, O.lmM ImM O.lmM ImM O.lmM , ImM
Sigma cat. no. T-4062 % viability by -100 -85-90 AlamarBlue test
Wy-14643 lOuM lOOuM lOuM lOOuM lOuM lOOuM
Cayman Chem cat. no. 70730 % viability by 400 -90 AlamarBlue test
ANIT, 25uM 250uM 25uM 250uM 25uM 250uM
Sigma cat. no. N-9883 % viability by -100 -60 AlamarBlue test
Acetominophen, ImM lOmM ImM lOmM ImM lOmM
Sigma cat. no. A-7085 % viability by -100 -90 AlamarBlue test
AY-25329 5uM 50uM 5uM 50uM 5uM 50uM
% viability by -100 -90 AlamarBlue test
CC , Aldrich O.lmM lOmM O.lmM lOmM O.lmM lOmM cat. no. 31996-1 % viability by -100 -80-85 AlamarBlue test
Clofibrate, 0.5mM 5mM 0.5mM 5mM 0.5mM 5mM
Sigma cat . no. C-6643 % viability by -100 -80-85 AlamarBlue test
Diclofenac, 55uM 550uM 55uM 550uM 55uM 550uM
Sigma cat. no. D-6899 % viability by -100 -70 AlamarBlue test Toxin 3 hours 6 hours 24 hours low dose high dose low dose high dose low dose high dose
17α-ethinylestradioI, lOuM lOOuM lOuM lOOuM lOuM lOOuM
Sigma cat. no. E-4876 % viability by -100 -80 AlamarBlue test
Hydrazine, O.lmM ImM O.lmM ImM O.lmM ImM
Sigma cat. no. H-0883 % viability by -90-95 -80-85 AlamarBlue test
Indomethacin, O.lmM ImM O.lmM ImM O.lmM ImM
Sigma cat. no. 1-8280 % viability by -100 -85-90 AlamarBlue test
Lipopolysaccharide, lOug/ml lOOug/ml lOug/ml lOOug/ml lOug/ml lOOug/ml
Sigma cat. no. L-8274 % viability by -100 -100 AlamarBlue test
Lovastatin, O.lmM ImM O.lmM ImM O.lmM ImM
Merck, 40 mg tablets % viability by -100 -100 AlamarBlue test
Methotrexate, ImM lOmM ImM lOmM ImM lOmM
Sigma cat. no. M-9929 % viability by -100 -90 AlamarBlue test
Tacrine, 25uM 250uM 25uM 250uM 25uM 250uM
Sigma cat. no. A-3773 % viability by -100 -75-80 AlamarBlue test
Valproate, 0.4mg/ml 4mg/ml 0.4mg/ml 4mg/ml 0.4mg/ml 4mg/ml
Sigma cat. no. P-4543 % viability by -100 -95 AlamarBlue test
Notes:
1. Each compound was dissolved in HTM cell culture medium (In Vitro Technologies) containing 0.2% DMSO (Sigma cat. no. D-5879).
2. The AlamarBlue assay was performed only at the 24-hr time point following exposure to the toxin of interest. A corresponding vehicle control (0.2% DMSO) sample was also isolated at 3, 6, and 24-hr time points for each toxin.

Claims

WE CLAIM:
1. A method of determining whether a compound induces at least one toxic effect on a tissue or cell, comprising:
(a) preparing a gene expression profile of a tissue or cell sample exposed to said compound; and
(b) comparing the gene expression profile to a database comprising an adequate amount of the data or information of Tables 5A-5XX to determine whether the compound induces at least one toxic effect on the tissue or cell.
2. A method of claim 1 , wherein the gene expression profile prepared from the tissue or cell sample comprises the level of expression for at least one gene.
3. A method of claim 2, wherein the level of expression is compared to a Tox Mean and/or Non-tox Mean value in Tables 5A-5XX.
4. A method of claim 3, wherein the level of expression is normalized prior to comparison.
5. A method of claim 1, wherein the database comprises substantially all of the data or information in Tables 5 A-5XX.
6. A method of predicting at least one toxic effect of a compound, comprising:
(a) detecting the level of expression in a tissue or cell sample exposed to the compound of two or more genes from Tables 5A-5XX; wherein differential expression of the genes in Tables 5 A-5XX is indicative of at least one toxic effect.
7. A method of predicting the progression of a toxic effect of a compound, comprising: (a) detecting the level of expression in a tissue or cell sample exposed to the compound of two or more genes from Tables 5A-5XX; wherein differential expression of the genes in Tables 5A-5XX is indicative of toxicity progression.
8. A method of predicting the hepatotoxicity of a compound, comprising: (a) detecting the level of expression in a tissue or cell sample exposed to the compound of two or more genes from Tables 5A-5XX; wherein differential expression of the genes in Tables 5A-5XX is indicative of hepatotoxicity.
9. A method of identifying an agent that modulates the onset or progression of a toxic response, comprising:
(a) exposing a cell to the agent and a known toxin; and
(b) detecting the expression level of two or more genes from Tables 5A-5XX; wherein differential expression of the genes in Tables 5A-5XX is indicative of toxicity.
10. A method of predicting the cellular pathways that a compound modulates in a cell, comprising:
(a) detecting the level of expression in a tissue or cell sample exposed to the compound of two or more genes from Tables 5A-5XX; wherein differential expression of the genes in Tables 5 A-5XX is associated the modulation of at least one cellular pathway.
11. The method of any one of claims 6-10, wherein the expression levels of at least 3 genes are detected.
12. The method of any one of claims 6-10, wherein the expression levels of at least 5 genes are detected.
13. The method of any one of claims 6- 10, wherein the expression levels of at least 10 genes are detected.
14. The method of any one of claims 6-10, wherein the expression levels of at least 50 genes are detected.
15. The method of any one of claims 6-10, wherein the expression levels of at least 100 genes are detected.
16. The method of any one of claims 6-10, wherein the expression levels of at least 500 genes are detected.
17. A method of any one of claims 6-10, wherein substantially all of the genes in Tables 5A-5XX are detected.
18. A method of claim 51, wherein all of the genes in at least one of Tables 5A-5XX are detected.
19. A method of any one of claims 6-10, wherein the compound exposure is in vitro.
20. A method of claim 19, wherein the cell sample comprises rat hepatocytes.
21. A method of any one of claims 6- 10, wherein the level of expression is detected by an amplification or hybridization assay.
22. A method of claim 21, wherein the amplification assay is quantitative or semi- quantitative PCR.
23. A method of claim 21, wherein the hybridization assay is selected from the group consisting of Northern blot, dot or slot blot, nuclease protection and microarray assays.
24. The method of any one of claims 6-10, wherein the detected level of expression is compared to that found in cells exposed to a known toxin.
25. The method of claim 24, wherein the toxin is selected from the group consisting of amiodarone, alpha-naphthylisothiocyante (ANIT), acetaminophen (APAP), AY-25329, carbamazepine, carbon tetrachloride, chlorpromazine, CI-1000, clofibrate, cyproterone acetate (CPA), diclofenac, diflunisal, dimethylnitrosamine (DMN), 17α-ethinylestradiol, gemfibrozil, hydrazine, imipramine, indomethacin, lipopolysaccharide, lovastatin, methotrexate, phenobarbital, tacrine, tamoxifen, tetracycline, valproate and Wy-14643.
26. The method of claim 25, wherein the level of expression is compared to that found in Tables 5A-5XX.
27. The method of claim 26, wherein the cells are primary hepatocytes.
28. The method of claim 27, wherein the cells are rat primary hepatocytes.
29. A method of claim 6 or 7, wherein the effect is selected from the group consisting of genotoxic and non-genotoxic carcinogenesis, cholestasis, direct-acting toxicity, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, and steatosis with hepatitis.
30. A method of claim 8, wherein the hepatotoxicity is associated with at least one liver disease pathology selected from the group consisting of genotoxic and non-genotoxic carcinogenesis, cholestasis, direct-acting toxicity, hepatitis, liver enlargement, inflammation, necrosis, necrosis with steatosis, peroxisome proliferation, steatosis, and steatosis with hepatitis.
31. A method of claim 10, wherein the cellular pathway is modulated by a toxin selected from the group consisting of amiodarone, alpha-naphthylisothiocyante (ANIT), acetaminophen (APAP), AY-25329, carbamazepine, carbon tetrachloride, chlorpromazine, CI-1000, clofibrate, cyproterone acetate (CPA), diclofenac, diflunisal, dimethylnitrosamine (DMN), 17α-ethinylestradiol, gemfibrozil, hydrazine, imipramine, indomethacin, lipopolysaccharide, lovastatin, methotrexate, phenobarbital, tacrine, tamoxifen, tetracycline, valproate and Wy-14643.
32. A set of at least two probes, wherein each of the probes comprises a sequence that specifically hybridizes to a gene in Tables 5A-5XX.
33. A set of probes according to claim 32, wherem the set comprises probes that hybridize to at least 3 genes.
34. A set of probes according to claim 32, wherein the set comprises probes that hybridize to at least 10 genes.
35. A set of probes according to claim 32, wherein the set comprises probes that hybridize to at least 100 genes.
36. A set of probes according to claim 32, wherein the set comprises probes that hybridize to at least 500 genes.
37. A set of probes according to any one of claims 32-36, wherein the probes are attached to a solid support.
38. A set of probes according to claim 37, wherein the solid support is selected from the group consisting of a membrane, a glass support and a silicon support.
39. A solid support comprising at least two probes, wherein each of the probes comprises a sequence that specifically hybridizes to a gene in Tables 5A-5XX.
40. A solid support of claim 39, wherein the solid support is an array comprising at least 10 different oligonucleotides in discrete locations per square centimeter.
41. A solid support of claim 39, wherein the array comprises at least about 100 different oligonucleotides in discrete locations per square centimeter.
42. A solid support of claim 39, wherein the array comprises at least about 1000 different oligonucleotides in discrete locations per square centimeter.
43. A solid support of claim 39, wherein the array comprises at least about 10,000 different oligonucleotides in discrete locations per square centimeter.
44. A computer system comprising:
(a) a database containing information identifying the expression level in a tissue or cell sample exposed to a hepatotoxin of a set of genes comprising at least two genes in Tables 1-5XX; and
(b) a user interface to view the information.
45. The computer system of claim 44, wherein the cell samples are rat primary hepatocytes.
46. A computer system of claim 44, wherein the database further comprises sequence information for the genes.
47. A computer system of claim 44, wherein the database further comprises information identifying the expression level for the set of genes in the tissue or cell sample before exposure to a hepatotoxin.
48. A computer system of claim 44, wherein the database further comprises information identifying the expression level of the set of genes in a tissue or cell sample exposed to at least a second hepatotoxin.
49. A computer system of any of claims 44-48, further comprising records including descriptive information from an external database, which information correlates said genes to records in the external database.
50. A computer system of claim 49, wherein the external database is GenBank.
51. A method of using a computer system of any one of claims 44-48 to present information identifying the expression level in a tissue or cell of at least one gene in Tables 5A-5XX, comprising:
(a) comparing the expression level of at least one gene in Tables 5 A-5XX in a tissue or cell exposed to a test agent to the level of expression of the gene in the database.
52. A method of claim 51 , wherein the expression levels of at least 2 genes are compared.
53. A method of claim 51 , wherein the expression levels of at least 10 genes are compared.
54. A method of claim 51 , wherein the expression levels of at least 100 genes are compared.
55. A method of claim 51 , further comprising the step of displaying the level of expression of at least one gene in the tissue or cell sample compared to the expression level when exposed to a toxin.
56. A kit comprising at least one solid support of any one of claims 39-43 packaged with gene expression information for said genes.
57. A kit of claim 56, wherein the gene expression information comprises gene expression levels in a tissue or cell sample exposed to a hepatotoxin.
58. A kit of claim 57, wherein the gene expression information is in an electronic format.
59. A method of identifying an agent that modulates at least one activity of a protein encoded by a gene in Tables 5A-5XX comprising:
(a) exposing the protein to the agent; and
(b) assaying at least one activity of said protein.
60. A method of claim 59, wherein the agent is exposed to a cell expressing the protein.
61. A method of claim 60, wherein the cell is exposed to a known toxin.
62. A method of claim 61, wherein the toxin modulates the expression of the protein.
PCT/US2003/003482 2002-02-04 2003-02-04 Primary rat hepatocyte toxicity modeling WO2003065993A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2003565419A JP2006502693A (en) 2002-02-04 2003-02-04 Primary rat hepatotoxicity modeling
CA002471631A CA2471631A1 (en) 2002-02-04 2003-02-04 Primary rat hepatocyte toxicity modeling
EP03715981A EP1578393A4 (en) 2002-02-04 2003-02-04 Primary rat hepatocyte toxicity modeling
AU2003219713A AU2003219713A1 (en) 2002-02-04 2003-02-04 Primary rat hepatocyte toxicity modeling

Applications Claiming Priority (36)

Application Number Priority Date Filing Date Title
US35317102P 2002-02-04 2002-02-04
US60/353,171 2002-02-04
US36353402P 2002-03-13 2002-03-13
US60/363,534 2002-03-13
US37024802P 2002-04-08 2002-04-08
US60/370,248 2002-04-08
US37113402P 2002-04-10 2002-04-10
US37113502P 2002-04-10 2002-04-10
US37115002P 2002-04-10 2002-04-10
US60/371,150 2002-04-10
US60/371,135 2002-04-10
US60/371,134 2002-04-10
US37141302P 2002-04-11 2002-04-11
US60/371,413 2002-04-11
US37360102P 2002-04-19 2002-04-19
US37360202P 2002-04-19 2002-04-19
US60/373,601 2002-04-19
US60/373,602 2002-04-19
US37413902P 2002-04-22 2002-04-22
US60/374,139 2002-04-22
US37837002P 2002-05-08 2002-05-08
US60/378,370 2002-05-08
US37866502P 2002-05-09 2002-05-09
US37865202P 2002-05-09 2002-05-09
US37865302P 2002-05-09 2002-05-09
US60/378,652 2002-05-09
US60/378,665 2002-05-09
US60/378,653 2002-05-09
US39425302P 2002-07-09 2002-07-09
US39423002P 2002-07-09 2002-07-09
US60/394,230 2002-07-09
US60/394,253 2002-07-09
US40768802P 2002-09-04 2002-09-04
US60/407,688 2002-09-04
US44290003P 2003-01-28 2003-01-28
US60/442,900 2003-01-28

Publications (2)

Publication Number Publication Date
WO2003065993A2 true WO2003065993A2 (en) 2003-08-14
WO2003065993A8 WO2003065993A8 (en) 2006-04-27

Family

ID=27739584

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/003482 WO2003065993A2 (en) 2002-02-04 2003-02-04 Primary rat hepatocyte toxicity modeling

Country Status (5)

Country Link
EP (1) EP1578393A4 (en)
JP (1) JP2006502693A (en)
AU (1) AU2003219713A1 (en)
CA (1) CA2471631A1 (en)
WO (1) WO2003065993A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1361433A2 (en) * 2002-04-09 2003-11-12 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Method for estimating therapeutic efficacy of tumor necrosis factor (TNF)
WO2005039588A2 (en) * 2003-10-22 2005-05-06 Novartis Ag Methods for determining the risk of developing liver and lung toxicity
WO2006024144A1 (en) * 2004-08-30 2006-03-09 Centre Hospitalier Universitaire De Quebec New method for determination of anabolic activity
EP1693452A1 (en) * 2003-11-27 2006-08-23 Takeda Pharmaceutical Company Limited Method of estimating toxicity of drug
EP1849864A1 (en) * 2005-02-17 2007-10-31 Takeda Pharmaceutical Company Limited Method for determining phospholipidosis
US8481507B2 (en) 2007-07-31 2013-07-09 The Board Of Regents, The University Of Texas System Micro-RNAs that control myosin expression and myofiber identity
US8629119B2 (en) 2009-02-04 2014-01-14 The Board Of Regents, The University Of Texas System Dual targeting of MIR-208 and MIR-499 in the treatment of cardiac disorders
WO2016201349A1 (en) * 2015-06-11 2016-12-15 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE112009001245T5 (en) * 2008-05-28 2012-03-15 Basf Se Means and methods for the evaluation of liver enzyme induction
JP5804629B2 (en) * 2011-07-29 2015-11-04 株式会社メディクローム Method for evaluating toxicity of chemical substances by gene expression fluctuation analysis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6372431B1 (en) * 1999-11-19 2002-04-16 Incyte Genomics, Inc. Mammalian toxicological response markers
US20020110808A1 (en) * 2000-01-21 2002-08-15 Reidhaar-Olson John F. Toxicant-induced differential gene expression
AU2001268449A1 (en) * 2000-06-14 2001-12-24 Vistagen, Inc. Toxicity typing using liver stem cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1578393A4 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1361433A3 (en) * 2002-04-09 2005-02-23 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Method for estimating therapeutic efficacy of tumor necrosis factor (TNF)
EP1361433A2 (en) * 2002-04-09 2003-11-12 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Method for estimating therapeutic efficacy of tumor necrosis factor (TNF)
WO2005039588A2 (en) * 2003-10-22 2005-05-06 Novartis Ag Methods for determining the risk of developing liver and lung toxicity
WO2005039588A3 (en) * 2003-10-22 2005-10-06 Novartis Ag Methods for determining the risk of developing liver and lung toxicity
EP1693452A4 (en) * 2003-11-27 2008-09-10 Takeda Pharmaceutical Method of estimating toxicity of drug
EP1693452A1 (en) * 2003-11-27 2006-08-23 Takeda Pharmaceutical Company Limited Method of estimating toxicity of drug
JPWO2005052154A1 (en) * 2003-11-27 2007-06-21 武田薬品工業株式会社 Drug toxicity prediction method
JP4616178B2 (en) * 2003-11-27 2011-01-19 武田薬品工業株式会社 Drug toxicity prediction method
WO2006024144A1 (en) * 2004-08-30 2006-03-09 Centre Hospitalier Universitaire De Quebec New method for determination of anabolic activity
EP1849864A4 (en) * 2005-02-17 2008-08-27 Takeda Pharmaceutical Method for determining phospholipidosis
EP1849864A1 (en) * 2005-02-17 2007-10-31 Takeda Pharmaceutical Company Limited Method for determining phospholipidosis
JP4819791B2 (en) * 2005-02-17 2011-11-24 武田薬品工業株式会社 Method for determining phospholipidosis
US8481507B2 (en) 2007-07-31 2013-07-09 The Board Of Regents, The University Of Texas System Micro-RNAs that control myosin expression and myofiber identity
US8962588B2 (en) 2007-07-31 2015-02-24 The Board Of Regents, The University Of Texas System Micro-RNAS that control myosin expression and myofiber identity
US8629119B2 (en) 2009-02-04 2014-01-14 The Board Of Regents, The University Of Texas System Dual targeting of MIR-208 and MIR-499 in the treatment of cardiac disorders
WO2016201349A1 (en) * 2015-06-11 2016-12-15 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US11506672B2 (en) 2015-06-11 2022-11-22 Astute Medical, Inc. Follistatin-related protein 3 for diagnosis and prognosis of renal injury and renal failure

Also Published As

Publication number Publication date
AU2003219713A8 (en) 2003-09-02
EP1578393A2 (en) 2005-09-28
AU2003219713A1 (en) 2003-09-02
WO2003065993A8 (en) 2006-04-27
JP2006502693A (en) 2006-01-26
CA2471631A1 (en) 2003-08-14
EP1578393A4 (en) 2008-03-19

Similar Documents

Publication Publication Date Title
US20090197258A1 (en) Primary rat hepatocyte toxicity modeling
US20080215250A1 (en) Molecular toxicology modeling
US20090220970A1 (en) Molecular toxicology modeling
US20070015146A1 (en) Molecular nephrotoxicology modeling
Zidek et al. Acute hepatotoxicity: a predictive model based on focused illumina microarrays
EP1392871A2 (en) Molecular toxicology modeling
US20070027634A1 (en) Molecular hepatotoxicology modeling
US20060078921A1 (en) Biomarkers and expression profiles for toxicology
WO2003065993A2 (en) Primary rat hepatocyte toxicity modeling
US20070055448A1 (en) Primary rat hepatocyte toxicity modeling
US20110071767A1 (en) Hepatotoxicity Molecular Models
WO2007022419A2 (en) Molecular toxicity models from isolated hepatocytes
EP1412537A2 (en) Cardiotoxin molecular toxicology modeling
US20060240418A1 (en) Canine gene microarrays
US20070054269A1 (en) Molecular cardiotoxicology modeling
WO2006037025A2 (en) Molecular toxicity models from isolated hepatocytes
EP1344834A2 (en) Methods for the toxicity prediction of a compound

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 BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE 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 NO NZ OM PH PL PT RO RU SC SD SE SG SK SL 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): GH GM KE LS MW MZ 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 IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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

Ref document number: 2471631

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003565419

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003715981

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003715981

Country of ref document: EP

D17 Declaration under article 17(2)a