WO2004087941A1 - Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs - Google Patents

Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs Download PDF

Info

Publication number
WO2004087941A1
WO2004087941A1 PCT/US2003/004786 US0304786W WO2004087941A1 WO 2004087941 A1 WO2004087941 A1 WO 2004087941A1 US 0304786 W US0304786 W US 0304786W WO 2004087941 A1 WO2004087941 A1 WO 2004087941A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
tumor
antigen
cancer cells
cell
Prior art date
Application number
PCT/US2003/004786
Other languages
English (en)
Inventor
James T. Kurnick
Paul J. Durda
Original Assignee
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The General Hospital Corporation filed Critical The General Hospital Corporation
Priority to AU2003222221A priority Critical patent/AU2003222221A1/en
Priority to EP03717900A priority patent/EP1509614A4/fr
Priority to CA 2475206 priority patent/CA2475206A1/fr
Publication of WO2004087941A1 publication Critical patent/WO2004087941A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • 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

  • This invention relates to methods and compositions for the treatment of tumors.
  • the invention relates to methods and agents for the treatment of tumors expressing a tumor-antigen [also known as a tumor-associated antigen (TAA), lineage specific antigen, differentiation antigen, or self-antigen].
  • TAA tumor-associated antigen
  • the invention also relates to methods for identifying agents that regulate expression of tumor-antigens.
  • TIL tumor-infiltrating lymphocytes
  • TAA tumor-associated antigens
  • TILs immunodominant epitopes recognized by TILs have also been described for widely-expressed lineage-specific antigens, for example, the HLA-A2- restricted Melan-A/MART-1 in melanomas (Sensi M, et al., Proc N ⁇ tl Ac ⁇ d Sci USA, 1995, 92:5674-8; Kawakami Y, et al., JExp Med, 1994, 180:347-52).
  • Melanomas are aggressive, frequently metastatic tumors derived from either melanocytes or melanocyte related nevus cells (Cellular and Molecular Immunology, 1991, (eds) Abbas A. K., Lechtman, A. H., Pober, J.
  • the invention provides methods for identifying agents that modulate expression of tumor-associated antigens in tumor cells.
  • the invention also provides agents and pharmaceutical compositions containing such agents that modulate expression of tumor- associated antigens in tumor cells.
  • the invention therefore, is particularly useful, inter alia, for treating subjects with autologous, solid tumors having cells that express, or that can be induced to express, tumor-associated antigens.
  • One category of materials according to the invention is tumor-antigen expression up-regulating agents. These agents include modulators of the MAPK/ERK signaling cascade, and more specifically modulators of MEK1/2 and ERK1/2 activity.
  • a method of upregulating tumor-antigen expression in a tumor cell invlves contacting a tumor cell with a MEK1/2 inhibitor in an amount effective to increase tumor-antigen expression in the tumor cell.
  • the contacting may occur in vivo and/or in vitro.
  • the origin of the tumor cells may be of: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical carcinoma; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T-cell leukemia lymphoma; intraepithelial neoplasms, including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; pancreas cancer; prostate cancer; rectal cancer; sarcomas, including
  • the tumor-antigen can be Melan-A/MART-1, melanoma GP75, PGP 9.5, Annexin I and 11, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC)-C017-1A/GA733, Ab2 BR3E4, CI17-1A/GA733, HSPPC-96, Hsp96, gp96-associated cellular peptides, G250, Herpes simplex thymidine kinase (HSVtk), Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, amll, Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), Ad5-PSA, Parathyroid-
  • the MEK1/2 inhibitor is PD98059, U0126, PP1 PP2A, a b-Raf inhibitor, or a c-Raf inhibitor.
  • the b-Raf inhibitor is a PK-A inhibitor.
  • a method of upregulating tumor- antigen expression in a tumor cell involves contacting a tumor cell with a ERK1/2 inhibitor in an amount effective to increase tumor-antigen expression in the tumor cell.
  • the contacting may occur in vivo and/or in vitro.
  • the origin of the tumor cells may be of: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical carcinoma; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T-cell leukemia lymphoma; intraepithelial neoplasms, including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; pancreas cancer; prostate cancer; rectal cancer; sarcomas, including
  • the tumor-antigen can be Melan-A/MART-1, melanoma GP75, PGP 9.5, Annexin I and II, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC) ⁇ C017-1A/GA733, Ab2 BR3E4, CI17-1A GA733, HSPPC-96, Hsp96, gp96-associated cellular peptides, G250, Herpes simplex thymidine kinase (HSVtk), Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, amll, Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), Ad5-PSA, Parathyroid-
  • Rab GTPase activating protein members of Rab-GAP (Rab GTPase-activating proteins) e.g., Rab GTPase-activating proteins
  • PARIS-1 T-cell receptor/CD3-zeta chain, cTAGE-1 and SCP-1, Glycolipid antigens- GM2, GD2, GD3, FucosylGMl, Glycoprotein (mucin) antigens-Tn, sialyl Tn., TF and MUC1, Hspl05, MAGE-family of tumor antigens, GAGE-1,2, BAGE, RAGE, GnT-V, MUM-1, CDK4, tyrosinase, p53, MUC family, HER2/neu, ErbB-2/neu, p21ras, RCAS1, -fetoprotein, E-cadherin, ⁇ -catenin, ⁇ -catenin and ⁇ -catenin, pl20ctn, gpl00 PmelU7 , PRAME, GA733/EoCam, thyroglobulin, NY-ESO-1, cdc27, adenomatous polyposis coli protein (AP
  • preferred tumor cells are melanoma cells and a preferred tumor-antigen is Melan-A/MART- 1.
  • a method of enhancing a tumor-specific immune response in a subject with cancer involves administering to a subject in need of such treatment a MEK-1/2 inhibitor or an ERK-1/2 inhibitor, in an amount effective to enhance a tumor-specific immune response in the subject such as that induced by tumor vaccine or adoptive transfer of tumor-reactive cells.
  • a tumor-specific immune response in the subject such as that induced by tumor vaccine or adoptive transfer of tumor-reactive cells.
  • Preferred cancers/tumors are as described elsewhere herein.
  • a method of enhancing a melanoma-specific immune response in a subject with melanoma involves administering to a subject in need of such treatment a MEK-1/2 inhibitor or an ERK-1/2 inhibitor, in an amount effective to increase Melan-A/MART-1 expression in malignant melanoma cells and enhance a melanoma-specific immune response in the subject such as that induced by tumor vaccine or adoptive transfer of tumor-reactive cells.
  • Preferred MEK-1/2 inhibitors and ERK-1/2 inhibitors are as described elsewhere herein. According to a further aspect, methods for preparing medicaments useful in enhancing a tumor-specific immune response in a subject with cancer and/or a melanoma- specific immune response in a subject with melanoma, are also provided. According to a further aspect of the invention, a method of identifying genes that modulate tumor-antigen expression in a tumor cell.
  • the method provides (a) contacting a tumor cell known to express a tumor-antigen with a modulator of the MAPK/ERK signaling cascade, (b) measuring tumor-antigen expression on the tumor cell, (c) determining whether tumor-antigen expression on the tumor cell is modulated compared to a control, and (d) identifying genes that modulate expression of tumor-antigen expression in the tumor cell.
  • the genes are identified using differential expression technology and are then isolated and characterized using conventional methodology.
  • the tumor cell is a cell selected from the group consisting of of acute lymphoblastic leukemia cells, glioma cells, bladder cancer cells, billiary cancer cells, breast cancer cells, cervical carcinoma cells, colon carcinoma cells, colorectal cancer cells, choriocarcinoma cells, epithelial cancer cells, gastric cancer cells, hepatocellular cancer cells, Hodgkins lymphoma cells, lung cancer cells, lymphoid cell-derived leukemia cells, myeloma cells, non-small cell lung carcinoma cells, nasopharyngeal cancer cells, ovarian cancer cancer cells, prostate cancer cells, pancreatic cancer cells, renal cancer cells, testicular cancer cells, T cell leukemia cells, and melanoma cells, and the tumor-antigen is selected from the group consisting of Melan-A/MART-1, melanoma GP75, PGP 9.5, Annexin I and II, Dipeptidyl peptidase IV (DPPIV), adeno
  • the contacting occurs in the presence or absence of Oncostatin M.
  • Preferred MAPK/ERK signaling cascade modulators are as described elsewhere herein.
  • the cells amy be those of a cell line selected from the group consisiting of 136.2, 453A, MM96L, MU, MU-X, EW, IGR-39D, and A375.
  • a solid-phase nucleic acid molecule array is provided.
  • the array consists essentially of a set of nucleic acid molecules, expression products thereof, or fragments thereof, fixed to a solid substrate.
  • the set of nucleic acid molecules comprises nucleic acid molecules of a tumor cell known to express a tumor-antigen, said tumor cell previously contacted with a modulator of the MAPK/ERK signaling cascade.
  • the solid-phase nucleic acid molecule array further comprises at least one control set of nucleic acid molecules.
  • a preferred control set of nucleic acid molecules consists essentially of nucleic acid molecules, expression products thereof, or fragments thereof, fixed to a solid substrate, wherein the set of nucleic acid molecules comprises nucleic acid molecules of a tumor cell known to express a tumor-antigen, said tumor cell not previously contacted with a modulator of the MAPK ERK signaling cascade.
  • Figure 1 shows graphs depicting the down-modulation of Antigen Expression in Melanoma Cell Line MU by OSM.
  • MU tumor cells were cultured for 3 days in control medium or in 20ng/ml of OSM (first and last panels), or in supernatants from EW (contains OSM) (2 nd panel) or A375 tumor cells (does not contain OSM) (3 rd panel).
  • Cells were stained for cytoplasmic expression of MA/MI protein (first 3 panels) or gplOO (last panel) and assayed by flow cytometry. Mean channel of fluorescence is shown within each panel.
  • Figure 2 is a graph depicting the inhibition of OSM activity on MU tumor cells by
  • MU tumor cells were cultured for three days in the presence of 1 Ong/ml of recombinant hOSM (OSM curve) as compared to control medium
  • MU tumor cells were treated with lOmM of PD98059 before adding 1 Ong/ml of recombinant hOSM (PD98059+OSM). Cultured cells were stained with monoclonal anti-Melan-A/MART-1 antibody (A103) and FITC-anti mouse antibody to detect cytoplasmic antigen. Histograms of the three cultures are overlaid to demonstrate the reversal of OSM effect by the PD98059 treatment.
  • Figure 3 shows graphs depicting up-regulation of Melan-A/MART-1 expression in melanoma cells by MEKl/2 inhibitors U0126 and PD98059.
  • Both Melan-A/MART-1 positive tumor cells, such as 453 A, and Melan- A/ MART- 1 -deficient cell lines, including EW, IGR-39D and A375 demonstrate up-regulation of this melanocyte-lineage antigen after 3 to 6 Days in the presence of 40-50 ⁇ M U0126 or PD98059.
  • Mean channel fluorescence for the Control and MEK-inhibitor-treated cells is shown for each cell line treated. In each case, the treated cells have readily-demonstrable increases in MA Ml staining, with the curve on the right representing the treated cells in each panel. (Note: Although MA/Ml deficient cells show up-regulation, they do not express as much antigen as the typical Melan-A/MART- 1 -positive melanoma tumor cells) .
  • Figure 4 shows an RT-PCR product depicting up-regulation of Melan-A/MART-1 mRNA expression in MU tumor cells by PD98059 and thus reversal of OSM-down- modulation.
  • Figure 5 shows RT-PCR product depicting the effect of U0126 (MEKl/2 inhibitor) on MA/Ml and tyrosinase mRNA levels and on OSM responses.
  • Figure 6 shows RT-PCR product depicting the up-regulation of Melan-A/MART-1 mRNA expression in MU-X tumor cells treated with MEKl/2 Inhibitor U0126.
  • Melan- A/MART-1 -deficient tumor cells, MU-X were treated for 3 days with 20 ⁇ M MEKl/2 inhibitor, U0126 (Lane 1), to demonstrate elevated mRNA levels (after PCR amplification of cDNAs) for Melan-A/MART-1. In contrast no mRNA can be detected in "control"
  • Figure 7 is a graph showing the induction of Cytotoxic T Cell Recognition by an MEKl/2 inhibitor. Cloned MA Ml specific cytotoxic T lymphocytes (CTL) were tested against MA/Ml-expressing MU tumor cells vs. low MA Ml -expressing A375 tumor cells.
  • CTL cytotoxic T lymphocytes
  • the A375 tumor cells were incubated for 6 days in the presence of MEK 1/2 inhibitor
  • agents and pharmaceutical compositions containing such agents that modulate expression of tumor-associated antigens in tumor cells can be used, inter alia, in vivo or in vitro, for the purpose of inhibiting growth of a tumor having cells expressing tumor-associated antigens, and in a variety of screening assays in order to identify additional agents that modulate expression of tumor-associated antigens in tumor cells.
  • tumor cells which normally express or present TAAs (intracellularly or on their surface) "lose” (or down-modulate) such TAA expression/ presentation when cultured at high density (Ramirez-Montagut T, et al., Clin Exp Immunol, 2000, 119(l).T l-8; U.S. Provisional App.
  • TAA down-modulation is mediated through a mitogen-activated protein kinase (MAPK) pathway. More specifically, we have discovered that TAA down-modulation is mediated through mitogen-activated protein kinase kinase family members MEK-1 and MEK-2 (or MEKl/2 for both), and their substrates ERK-1 and ERK-2 (or ERK1/2 for both, members of the mitogen-activated protein kinase family).
  • MEK-1 and MEK-2 or MEKl/2 for both
  • ERK-1 and ERK-2 or ERK1/2 for both, members of the mitogen-activated protein kinase family.
  • an immunoreactive cell sample is used in conjunction with the methods of the invention described herein.
  • immunoreactive cell is meant a cell which can mature into an immune cell (such as a B cell, a helper T cell, or a cytolytic T cell) upon appropriate stimulation.
  • immunoreactive cells include CD34 + hematopoietic stem cells, immature T cells and immature B cells.
  • the immunoreactive cell When it is desired to produce cytolytic T cells which recognize a tumor antigen or a tumor-associated antigen, the immunoreactive cell is contacted with a cell which expresses a tumor antigen or a tumor-associated antigen under conditions favoring production, differentiation and/or selection of cytolytic T cells; the differentiation of the T cell precursor into a cytolytic T cell upon exposure to antigen is similar to clonal selection of the immune system.
  • Some therapeutic approaches are premised on a response by a subject's immune system, leading to lysis of antigen presenting cells, such as cancer cells which present one or more tumor antigens or tumor-associated antigens.
  • antigen presenting cells such as cancer cells which present one or more tumor antigens or tumor-associated antigens.
  • One such approach is the administration of autologous CTLs specific to a cancer-associated antigen/MHC complex to a subject with abnormal cells of the phenotype at issue. It is within the ability of one of ordinary skill in the art to develop such CTLs in vitro.
  • An example of a method for T cell differentiation is presented in International Application number PCT/US96/05607.
  • a sample of cells taken from a subject such as blood cells, are contacted with a cell presenting the complex and capable of provoking CTLs to proliferate.
  • the target cell can be a transfectant, such as a COS cell. These transfectants present the desired complex of their surface and, when combined with a CTL of interest, stimulate its proliferation. COS cells are widely available, as are other suitable host cells. Specific production of CTL clones is well known in the art. The clonally expanded autologous CTLs then are administered to the subject. Another method for selecting antigen-specific CTL clones has recently been described (Altaian et al., Science 274:94-96, 1996; Dunbar et al., Curr. Biol. 8:413-416, 1998). The reactive CTLs isolated by the method can then be expanded in vitro for use as described herein.
  • MAPKs e.g., ERK1/2
  • MAPKs comprise a family of related protein kinases that are themselves activated by MAPK-activating enzymes (MAPK/ERK kinases or MEKs) by phosphorylation on their threonine and tyrosine residues. Once activated they can regulate targets in the cytosol and can also translocate to the nucleus where they can phosphorylate a variety of transcription factors regulating gene expression of important cell-cycle and differentiation-specific proteins.
  • MAPK-activating enzymes are unusual in their ability to catalyze phosphorylation on both threonine and tyrosine residues.
  • MEKs are themselves activated by phosphorylation of their serine residues by upstream kinases (e.g., c-Raf).
  • modulators of MEKl/2 and ERK1/2 activity can up- regulate TAA expression/presentation, thus enhancing recognition/lysis of tumors expressing tumor-associated antigens by tumor-infiltrating lymphocytes (TILs) .
  • TILs tumor-infiltrating lymphocytes
  • down-modulation and up-modulation are used interchangeably with the terms “down-regulation” and “up-regulation,” respectively, throughout this application.
  • Down-modulating (down-regulating), refers to inhibition of tumor-antigen (or TAA) expression.
  • Inhibition of tumor-antigen expression refers to inhibiting (i.e., reducing to a detectable extent) expression/presentation of the specific antigen intracellularly and/or at the surface of a tumor cell.
  • Such inhibition of tumor- antigen expression can be directly determined by detecting a decrease in the level of mRNA for the gene encoding the antigen, or the level of peptide expression of the tumor- antigen, using any suitable means known to the art, such as nucleic acid hybridization or, preferably, antibody detection methods, respectively (see Examples).
  • Inhibition of tumor- antigen expression can also be determined indirectly, for example, by detecting a change in tumor-cell lysis ability by TILs that specifically recognize the tumor-antigen.
  • up-modulating (up-regulating), refers to an increase in tumor-antigen (or TAA) expression.
  • Upregulation of tumor-antigen expression refers to increasing (i.e., increasing to a detectable extent) expression/presentation of the specific antigen intracellularly and/or at the surface of a tumor cell.
  • the present invention relates in one aspect to down-modulators (or inhibitors) of MEKl/2 and/or ERK1/2 activity.
  • MEKl/2 activity inhibitors are known to those of ordinary skill in the art and include agents that can inhibit the phosphorylation of both threonine and tyrosine residues on MEKl/2 substrates such as ERK1/2 or ATP.
  • the MEKl/2 inhibitor is selected from the group consisting of small molecule organic compounds, inhibitory antibodies, synthetic kinase substrate peptides, dominant negative MEK1 proteins, antisense nucleic acids, and ribozymes which reduce the expression of translatable MEKl/2 transcripts.
  • the MEKl/2 inhibitor is a small molecule organic compound, particularly a tricyclic flavone or a (phenylthio)butadiene.
  • the MEK1 inhibitor is 2-(2-amino-3-methoxyphenyl)-4-oxo-4H- [l]benzopyran (PD98059), l,4-diamino-2,3-dicyano-l,4-bis-(phenylthio)butadiene (U0125) or l,4-diamino-2,3-dicyano- l,4-bis-(2-aminophenylthio)butadiene (U0126).
  • MEKl/2 inhibitors include a compound selected from the group consisting of PP1 PP2A, a b-Raf inhibitor, or a c-Raf inhibitor.
  • the b-Raf inhibitor is a PK-A inhibitor.
  • Variants of the foregoing (phenylthio)butadiene compounds which retain the MEKl/2 inhibitory activity of the (phenylthio)butadiene compounds also can be used in accordance with the invention. Substitutions preferably are made for the phenyl rings, or for one or both of the NH group on the phenyl rings of U0126, above.
  • Chemical groups which can be added to one or both ends of the molecule include: hydrido, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, acyl, amino, acyloxy, acylamino, carboalkoxy, carboxyamido, carboxyamido, halo and thio groups. Substitutions also can be made for the terminal groups on the butadiene portion of the molecule (i.e., CN, NH 2 ) > Molecular terms, when used in this application, have their common meaning unless otherwise specified.
  • the term "hydrido" denotes a single hydrogen atom (H).
  • H hydrogen atom
  • acyl is defined as a carbonyl radical attached to an alkyl, alkenyl, alkynyl, cycloalkyl, heterocycyl, aryl or heteroaryl group, examples of such radicals being acetyl and benzoyl.
  • amino denotes a nitrogen radical containing two substituents independently selected from the group consisting of hydrido, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
  • acyloxy denotes an oxygen radical adjacent to an acyl group.
  • acylamino denotes a nitrogen radical adjacent to an acyl group.
  • carbboalkoxy is defined as a carbonyl radical adjacent to an alkoxy or aryloxy group.
  • carboxyamido denotes a carbonyl radical adjacent to an amino group.
  • Carboxy embraces a carbonyl radical adjacent to an alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl group.
  • halo is defined as a bromo, chloro, fluoro or iodo radical.
  • thio denotes a radical containing a substituent group independently selected from hydrido, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, s attached to a divalent sulfur atom, such as, methylthio and phenylthio.
  • alkyl is defined as a linear or branched, saturated radical having one to about ten carbon atoms unless otherwise specified. Preferred alkyl radicals are "lower alkyl” radicals having one to about five carbon atoms.
  • One or more hydrogen atoms can also be replaced by a substitutent group selected from acyl, amino, acylamino, acyloxy, o carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • alkyl groups include methyl, tert-butyl, isopropyl, and methoxymethyl.
  • alkenyl embraces linear or branched radicals having two to about twenty carbon atoms, preferably three to about ten carbon atoms, and containing at least one carbon- 5 carbon double bond.
  • One or more hydrogen atoms can also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • alkenyl groups include ethylenyl or phenyl ethylenyl.
  • alkynyl denotes linear or o branched radicals having from two to about ten carbon atoms, and containing at least one carbon-carbon triple bond.
  • One or more hydrogen atoms can also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • alkynyl groups include propynyl.
  • aryl denotes aromatic radicals in a single or fused carbocyclic ring system, having from five to twelve ring members.
  • One or more hydrogen atoms may also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy
  • Heteroaryl embraces aromatic radicals which contain one to four hetero atoms selected from oxygen, nitrogen and sulfur in a single or fused heterocyclic ring system, having from five to fifteen ring members.
  • One or more hydrogen atoms may also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • heteroaryl groups include, pyridinyl, thiazolyl, thiadiazoyl, isoquinolinyl, pyrazolyl, oxazolyl, oxadiazoyl, triazolyl, and pyrrolyl groups.
  • cycloalkyl is defined as a saturated or partially unsaturated carbocyclic ring in a single or fused carbocyclic ring system having from three to twelve ring members.
  • One or more hydrogen atoms may also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • cycloalkyl group examples include cyclopropyl, cyclobutyl, cyclohexyl, and cycloheptyl.
  • heterocyclyl embraces a saturated or partially unsaturated ring containing zero to four hetero atoms selected from oxygen, nitrogen and sulfur in a single or fused heterocyclic ring system having from three to twelve ring members.
  • One or more hydrogen atoms may also be replaced by a substituent group selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxy, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfoxy, and formyl.
  • a heterocyclyl group include morpholinyl, piperidinyl, and pyrrolidinyl.
  • alkoxy denotes oxy-containing radicals substituted with an alkyl, cycloalkyl or heterocyclyl group. Examples include methoxy, tert-butoxy, benzyloxy and cyclohexyloxy.
  • aryloxy denotes oxy-containing radicals substituted with an aryl or heteroaryl group.
  • sulfoxy is defined as a hexavalent sulfur radical bound to two or three substituents selected from the group consisting of oxo, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein at least one of said substituents is OXO.
  • the invention embraces the use of antisense oligonucleotides that selectively bind to a nucleic acid molecule encoding a MEKl/2 polypeptide, to decrease MEKl/2 transcription or translation.
  • Antisense molecules in this manner, can be used to decrease or prevent the effects mediated by MEKl/2.
  • antisense oligonucleotide or “antisense” describes an oligonucleotide that is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide, or modified oligodeoxyribonucleotide which hybridizes under physiological conditions to DNA comprising a particular gene or to an mRNA transcript of that gene and, thereby, inhibits the transcription of that gene and/or the translation of that mRNA.
  • the antisense molecules are designed so as to interfere with transcription or translation of a target gene upon hybridization with the target gene or transcript.
  • the exact length of the antisense oligonucleotide and its degree of complementarity with its target will depend upon the specific target selected, including the sequence of the target and the particular bases which comprise that sequence. It is preferred that the antisense oligonucleotide be constructed and arranged so as to bind selectively with the target under physiological conditions, i.e., to hybridize substantially more to the target sequence than to any other sequence in the target cell under physiological conditions.
  • antisense oligonucleotides Based upon the MEK1 cDNA sequence , for example (GenBank accession numbers L02526 (mouse) and LI 1284 (human)), or upon allelic or homologous genomic and/or cDNA sequences, one of skill in the art can easily choose and synthesize any of a number of appropriate antisense molecules for use in accordance with the present invention.
  • such antisense oligonucleotides should comprise at least 10 and, more preferably, at least 15 consecutive bases which are complementary to the target, although in certain cases modified oligonucleotides as short as 7 bases in length have been used successfully as antisense oligonucleotides (Wagner et al., Nature Biotechnol. 14:840-844, 1996). Most preferably, the antisense oligonucleotides comprise a complementary sequence of 20-30 bases.
  • oligonucleotides may be chosen which are antisense to any region of the gene or mRNA transcripts, in preferred embodiments the antisense oligonucleotides j correspond to N-terminal or 5' upstream sites such as translation initiation, transcription initiation or promoter sites. In addition, 3 '-untranslated regions may be targeted. Targeting to mRNA splicing sites has also been used in the art but may be less preferred if alternative mRNA splicing occurs. In addition, the antisense is targeted, preferably, to sites in which mRNA secondary structure is not expected (see, e.g., Sainio et al., Cell Mol.
  • RNA interference RNA interference
  • hairpin RNA interference
  • duplex RNA interference
  • siRNAs 20 mRNA degradation in the process of RNA interference (RNAi).
  • siRNAs are 18-23 nucleotide duplexes. Most frequently, siRNAs are 21-23 nucleotide duplexes.
  • RNA interference is the process of sequence-specific, post-transcriptional gene silencing in animals and plants, initiated by double-stranded RNA (dsRNA) that is homologous in sequence to the silenced gene.
  • dsRNA double-stranded RNA
  • 25 messenger RNA degradation are preferably 21- and 22-nucleotide small interfering RNAs (siRNAs) generated by ribonuclease III cleavage from longer dsRNAs (Elbashir SM et al. Nature 2001, May 24, 411, 428-429)
  • siRNA duplexes with overhanging 3' ends mediate efficient target RNA cleavage in the lysate, and the cleavage site is located near the center
  • the antisense oligonucleotides of the invention may be composed of "natural" deoxyribonucleotides, ribonucleotides, or any combination thereof. That is, the 5' end of one native nucleotide and the 3' end of another native nucleotide may be covalently linked, as in natural systems, via a phosphodiester internucleotide linkage.
  • oligonucleotides may be prepared by art recognized methods which may be carried out manually or by an automated synthesizer. They also may be produced recombinantly by vectors. In preferred embodiments, however, the antisense oligonucleotides of the invention also may include "modified" oligonucleotides. That is, the oligonucleotides may be modified in a number of ways which do not prevent them from hybridizing to their target but which enhance their stability or targeting or which otherwise enhance their therapeutic effectiveness.
  • modified oligonucleotide as used herein describes an oligonucleotide in which (1) at least two of its nucleotides are covalently linked via a synthetic internucleoside linkage (i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide) and/or (2) a chemical group not normally associated with nucleic acids has been covalently attached to the oligonucleotide.
  • a synthetic internucleoside linkage i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide
  • Preferred synthetic internucleoside linkages are phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidates, carboxymethyl esters and peptides.
  • modified oligonucleotide also encompasses oligonucleotides with a covalently modified base and/or sugar.
  • modified oligonucleotides include oligonucleotides having backbone sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3' position and other than a phosphate group at the 5' position.
  • modified oligonucleotides may include a 2'-0- alkylated ribose group.
  • modified oligonucleotides may include sugars such as arabinose instead of ribose.
  • Modified oligonucleotides also can include base analogs such as C-5 propyne modified bases (Wagner et al., Nature Biotechnology 14:840-844, 1996).
  • the present invention contemplates the use of pharmaceutical preparations containing modified antisense molecules that are complementary to and hybridizable with, under physiological conditions, nucleic acids encoding MEKl/2 polypeptides, together with pharmaceutically acceptable carriers.
  • the invention also provides, in certain embodiments, the use of "dominant negative" MEKl/2 polypeptides.
  • a dominant negative polypeptide is an inactive variant of a protein, which, by interacting with the cellular machinery, displaces an active protein from its interaction with the cellular machinery or competes with the active protein, thereby reducing the effect of the active protein.
  • dominant negative MEK 1 proteins include MEKl/2 proteins having a catalytically-inactive kinase domain which interacts normally with target proteins but does not phosphorylate the target proteins, or which does not interact with normally with target proteins, or both.
  • Dominant negative MEKl/2 proteins include variants in which a portion of the kinase domain has been mutated or deleted to reduce or eliminate substrate binding or kinase activity.
  • the end result of the expression of a dominant negative MEKl/2 polypeptide in a cell is a reduction in function of active proteins.
  • One of ordinary skill in the art can assess the potential for a dominant negative variant of a protein, and using standard mutagenesis techniques to create one or more dominant negative variant polypeptides. For example, given the nucleotide sequence of MEKl/2, one of ordinary skill in the art can modify the sequence of the MEKl/2 polypeptide by site-specific mutagenesis, scanning mutagenesis, partial gene deletion or truncation, and the like. See, e.g., U.S. Pat. No. 5,580,723 and Sambrook et al., Molecular Cloning.
  • the invention also embraces MEKl/2 binding agents which can be antibodies or fragments of antibodies having the ability to selectively bind to MEKl/2 polypeptides. Such agents can be used to inhibit the native activity of the MEKl/2 polypeptides by binding to such polypeptides.
  • Antibodies include polyclonal and monoclonal antibodies, prepared according to conventional methodology.
  • the pFc' and Fc regions are effectors of the complement cascade but are not involved in antigen binding.
  • an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region, designated an Fab fragment, retains one of the antigen binding sites of an intact antibody molecule.
  • Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd.
  • the Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
  • CDRs complementarity determining regions
  • FRs framework regions
  • CDR1 through CDR3 complementarity determining regions
  • non-CDR regions of a mammalian antibody may be replaced with similar regions of conspecific or heterospecific antibodies while retaining the epitopic specificity of the original antibody.
  • This is most clearly manifested in the development and use of "humanized" antibodies in which non-human CDRs are covalently joined to human FR and/or Fc/pFc' regions to produce a functional antibody. See, e.g., U.S. Pat. Nos. 4,816,567, 5,225,539, 5,585,089, 5,693,762 and 5,859,205.
  • PCT International Publication Number WO 92/04381 teaches the production and use of humanized murine RSV antibodies in which at least a portion of the murine FR regions have been replaced by FR regions of human origin.
  • Such antibodies including fragments of intact antibodies with antigen-binding ability, are often referred to as "chimeric" antibodies.
  • the present invention also provides for F(ab') 2 , Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDRl and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab') fragment antibodies in which the FR and/or CDRl and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDRl and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDRl and/or CDR2 regions have been replaced by homologous human or non-human sequences.
  • the present invention also includes so-called single chain antibodies.
  • polypeptides of numerous size and type that bind specifically to MEKl/2 polypeptides, and complexes of both MEKl/2 polypeptides and their binding partners.
  • polypeptide binding agents can be provided by degenerate peptide libraries which can be readily prepared in solution, in immobilized form or as phage display libraries.
  • Combinatorial libraries also can be synthesized of peptides containing one or more amino acids. Libraries further can be synthesized of peptoids and non-peptide synthetic moieties.
  • Phage display can be particularly effective in identifying binding peptides useful according to the invention, including human antibodies. Briefly, one prepares a phage library (using e.g. ml3, fd, or lambda phage), displaying inserts from 4 to about 80 amino acid residues using conventional procedures. The inserts may represent, for example, a completely degenerate or biased array. One then can select phage-bearing inserts which bind to the MEKl/2 polypeptide. This process can be repeated through several cycles of reselection of phage that bind to the MEKl/2 polypeptide. Repeated rounds lead to enrichment of phage bearing particular sequences. DNA sequence analysis can be conducted to identify the sequences of the expressed polypeptides.
  • the minimal linear portion of the sequence that binds to the MEKl/2 polypeptide can be determined.
  • Yeast two-hybrid screening methods also may be used to identify polypeptides that bind to the MEKl/2 polypeptides.
  • MEKl/2 polypeptides, or fragments thereof can be used to screen peptide libraries, including phage display libraries, to identify and select peptide binding partners of MEKl/2. Additional methodologies are described by Zhang et al., Nature Biotechnol. 18:71-74, 2000. Such molecules can be used, as described, for interfering directly with the functioning of
  • MEKl/2 activity For example U.S. Pat. No. 5,525,625, the disclosure of which is incorporated herein by reference, describes several assays which are useful for determining the MEKl/2 inhibitory potential of a test compound.
  • the assays include in vitro kinase assays, whole cell kinase assays, and cell growth assays including assays of monolayer growth and growth in soft agar.
  • the Examples below provide an in vivo assay of focal cerebral ischemia and in vitro cell-based assays for testing the activity of MEKl/2 inhibitors. Additional assays are described by Favata et al. (J. Biol. Chem.
  • test compound is able to inhibit the MEK 1 activity, then it is a compound which is useful in the treatment of ischemia, particularly stroke, and other conditions including hypoxia and glutamate toxicity.
  • the test compound can be determined readily to be a specific inhibitor of MEKl/2 activity.
  • ERK1/2 are serine/threonine kinases that are activated in the cytosol in response to specific extracellular signals and can be translocated to the nucleus. They are known to phosphorylate many different proteins including transcription factors that regulate expression of important cell-cycle and differentiation-specific proteins.
  • ERK1/2 activity inhibitors are known to those of ordinary skill in the art and include agents that can inhibit the phospho ylation of a number of proteins, for example, the actin-associated protein caldesmon (CaD) (D'Angelo G, et al., Am J Physiol Heart Circ Physiol, 2002, Feb;282:H602-10), or inhibit cell functions such as the attenuation or enhancement of FSH-induced progesterone or estradiol production in granulosa cells (Moore RK, et al., Biochem Biophys Res Commun., 2001, Dec 14; 289:796-800), or the FGF-induced lens cell proliferation and fibre differentiation (Lovicu FJ, et al.,
  • the ERKl/2 inhibitor is MKP-1, MKP-3, or a MEKl/2 inhibitor.
  • MEKl/2 and ERKl/2 activities are well established in the art, and as a result, techniques for measuring such activities are also well established and known to those of ordinary skill in the art (see, e.g., Favata MF, et al, JBiol Chem., 1998, Jul 17;273:18623-
  • a tumor cell may be of a cancer or tumor type thought to escape immune recognition.
  • Such cancers or tumors may be of the folowing origin: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical carcinoma; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T- cell leukemia lymphoma; intraepithelial neoplasms, including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from epithelial cells, stromal cells, germ cells and mesenchy
  • the tumor-antigen can be Melan-A MART-1, melanoma GP75, PGP 9.5, Annexin I and II, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC)-C017-1A/GA733, Ab2 BR3E4, CI17-1A/GA733, HSPPC-96, Hsp96, gp96-associated cellular peptides, G250, Herpes simplex thymidine kinase (HSVtk), Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, amll, Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), Ad5-PSA, Parathyroid-
  • MAGE-family of tumor antigens GAGE- 1,2, BAGE, RAGE, GnT-V, MUM-1, CDK4, tyrosinase, p53, MUC family, HER2/neu, ErbB-2/neu, p21ras, RCAS1, ⁇ -fetoprotein, E- cadherin, ⁇ -catenin, ⁇ -catenin and ⁇ -catenin, pl20ctn, gpl00 Pmel117 , PRAME,
  • the fraction from the plurality of fractions can be undiluted or concentrated.
  • cancers or tumors escaping immune recognition and tumor-antigens associated with such tumors include acute lymphoblastic leukemia (etv6; amll; cyclophilin b), glioma (E-cadherin; ⁇ -catenin; ⁇ - catenin; ⁇ -catenin; pl20ctn; PHF3), bladder cancer (p21ras), billiary cancer (p21ras), breast cancer (MUC family; HER2/neu; ErbB-2/neu; c-erbB-2, PDEF; Cytochrome oxidase 1; splOO; Ran GTPase activating protein; NY-ESO-1; LAGE-1; SCP-1; SSX-1; SSX-2; SSX-4), cervical carcinoma (p53; p21ras), colon carcinoma (p21ras; FJER2/neu; c- erbB-2; MUC family; NY-ESO-1; LAGE
  • T cell leukemia HTLV-1 epitopes, cTAGE-1, SCP-1
  • melanoma Melan- A/MART-1; cdc27; MAGE-3; p21ras; gpl00 Pmel117 .
  • a subject is a human, non-human primate, cow, horse, pig, sheep, goat, dog, cat or rodent. In all embodiments, human subjects are preferred.
  • the down-modulation inhibitors of the invention are administered in therapeutically effective amounts.
  • a therapeutically effective amount means that amount necessary to delay the onset of, inhibit the progression of, or halt altogether the particular condition being treated.
  • a therapeutically effective amount will vary with the subject's age, condition, and sex, as well as the nature and extent of the disease in the subject, all of which can be determined by one of ordinary skill in the art.
  • the dosage may be adjusted by the individual physician or veterinarian, particularly in the event of any complication.
  • a therapeutically effective amount typically varies from 0.01 mg/kg to about 1000 mg/kg, preferably from about 0.1 mg/kg to about 200 mg/kg, and most preferably from about 0.2 mg//kg to about 20 mg/kg, in one or more dose administrations daily, for one or more days.
  • the therapeutically effective amount of the tumor-antigen expression down- modulation inhibitors of the invention is that amount effective to inhibit tumor-antigen expression down-modulation, and can be determined using, for example, standard tests known in the art.
  • a direct way to measure tumor-antigen (e.g., Melan- A/MART-1) expression the tumor cell (e.g., melanoma) is to use antibodies specific for the tumor-antigen and a number of immunocyto- and immunohisto- chemical protocols well known in the art.
  • Antibodies specific for the Melan-A/MART-1 antigen, for example, are fully described in U.S.
  • Patent 5,674,749 to Chen et al entitled: "Monoclonal antibodies which bind to tumor rejection antigen precursor Melan-A, and uses thereof.”
  • the tumor-antigen expression down-modulation inhibitors of the invention i.e., modulators of the MAPK/ERK signaling cascade, and more specifically down-modulators of MEKl/2 and ERKl/2 activity
  • an anti- cancer agent other than an agent of the invention e.g., other than a MEKl/2 and/or an ERK1/2 inhibitor
  • an agent of the invention e.g., other than a MEKl/2 and/or an ERK1/2 inhibitor
  • co-administered means administered substantially simultaneously with another agent (e.g., in different or same compositions/formulations).
  • a MEKl/2 and/or an ERKl/2 inhibitor of the invention is administered to the subject close enough in time with the administration of the other agent (e.g., an anti-cancer agent) whereby the two agents may exert an additive or even synergistic effect to upregulate tumor-antigen expression and inhibit growth and/or proliferation of the cancer.
  • Anti-cancer agents other than agents of the invention include, but are not limited to: Acivicin; Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycin
  • Losoxantrone Hydrochloride Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin;
  • Mitocromin Mitogillin; Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone
  • Paclitaxel Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Phcamycin; Plomestane; Podofilox;
  • Porfimer Sodium Porfiromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin;
  • Spirogermanium Hydrochloride Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Sulofenur; Talisomycin; Taxotere; Tecogalan Sodium; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfm; Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglyc
  • the above-described drug therapies are well known to those of ordinary skill in the art and are administered by modes known to those of skill in the art.
  • the drug therapies are administered in amounts which are effective to achieve the physiological goals in combination with the MEKl/2 and/or an ERKl/2 inhibitors of the invention.
  • a MEKl/2 and/or an ERKl/2 inhibitor may be administered alone or in combination with the above-described drug therapies as part of a pharmaceutical composition.
  • a pharmaceutical composition may include the MEKl/2 and/or an ERKl/2 inhibitor in combination with any standard physiologically and/or pharmaceutically acceptable carriers which are known in the art.
  • the compositions can be sterile and contain a therapeutically effective amount of the MEKl/2 and/or an ERKl/2 inhibitor in a unit of weight or volume suitable for administration to a patient.
  • pharmaceutically-acceptable carrier as used herein means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration into a human or other animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • Pharmaceutically acceptable further means a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism.
  • the characteristics of the carrier will depend on the route of administration. Physiologically and pharmaceutically acceptable carriers include diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials which are well known in the art. A variety of administration routes are available.
  • the particular mode selected will depend, of course, upon the particular drug selected, the severity of the condition being treated, and the dosage required for therapeutic efficacy.
  • the methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects.
  • modes of administt-ation include oral, rectal, topical, nasal, intradermal, or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are not particularly suitable for long-term therapy and prophylaxis. They could, however, be preferred in emergency situations. Oral administration will be preferred for prophylactic treatment because of the convenience to the patient as well as the dosing schedule.
  • compositions suitable for parenteral administration conveniently comprise a sterile aqueous preparation of the MEKl/2 and/or an ERKl/2 inhibitor, which is preferably isotonic with the blood of the recipient.
  • This aqueous preparation may be formulated according to l ⁇ iown methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation also may be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di- glycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectables.
  • Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found in Remington's Pharmaceutical Sciences,
  • compositions may conveniently be presented in unit dosage
  • J form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the MEKl/2 and/or an ERKl/2 inhibitor into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the MEKl/2 and/or an ERKl/2 inhibitor into association with a liquid carrier, a finely divided solid carrier, or 0 both, and then, if necessary, shaping the product.
  • compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the MEKl/2 and/or an ERKl/2 inhibitor.
  • Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the MEKl/2 and/or ERKl/2 inhibitors described above, increasing convenience to the subject and the physician. Many types of release delivery systems are available and l ⁇ iown to those of ordinary skill in the art.
  • polymeric systems include the above-described polymeric systems, as well as o polymer base systems such as poly(lactide-gtycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • o polymer base systems such as poly(lactide-gtycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.
  • Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Patent 5,075,109.
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such 5 as mono- di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such 5 as mono- di- and tri-glycerides
  • hydrogel release systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such 5 as mono- di- and tri-glycerides
  • sylastic systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such 5 as mono- di- and tri-glycerides
  • peptide based systems such as mono- di- and tri-glycerides
  • wax coatings such as those described in U.S. Patent Nos.
  • Long-term sustained release means that the implant is constructed and arranged to delivery therapeutic levels of the active ingredient for at least 30 days, and preferably 60 days.
  • Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • the invention also embraces methods for identifying genes that modulate tumor- antigen expression in a tumor cell.
  • a method according to this aspect of the invention typically involves: (a) contacting a tumor cell known to express a tumor-antigen with a modulator of the MAPK/ERK signaling cascade, (b) measuring tumor-antigen expression on the tumor cell, (c) determining whether tumor-antigen expression on the tumor cell is modulated compared to a control, and (d) identifying genes that modulate expression of tumor-antigen expression in the tumor cell.
  • the genes are identified using differential expression technology and are then isolated and characterized using conventional methodology.
  • a tumor cell l ⁇ iown to express a tumor-antigen includes: acute lymphoblastic leukemia (etv6; amll; cyclophilin b), glioma (E-cadherin; ⁇ -catenin; ⁇ -catenin; ⁇ -catenin; pl20ctn; PHF3), bladder cancer (p21ras), billiary cancer (p21ras), breast cancer (MUC family; HER2/neu; ErbB-2/neu; c-erbB-2, PDEF; Cytochrome oxidase 1; splOO; Ran GTPase activating protein; NY-ESO-1; LAGE-1; SCP-1; SSX-1; SSX-2; SSX-4), cervical carcinoma (p53; p21ras), colon carcinoma (p21ras; HER2/neu; c-erbB-2; MUC family; NY-ESO-1; LAGE-1; SCP-1; SSX-1; SSX
  • GTPase-activating proteins e.g., PARIS-1, Ad5-PSA, PTH-rP, PAP
  • pancreatic cancer e.g., PARIS-1, Ad5-PSA, PTH-rP, PAP
  • HER2/neu c-erbB-2; OFA-iLR; G250
  • testicular cancer NY-ESO-1
  • T cell leukemia HTLV-1 epitopes, cTAGE-1, SCP-1
  • melanoma Melan-A/MART-1 ; cdc27;
  • Typical controls include identically isolated and cultured cells, with the exception, for example, that the supernatant medium in the control cultures is removed at regular intervals during the culture period (e.g. every 2-6 hours), being replaced with fresh culture medium. This media change effectively eliminates any down-regulatory effects a control-tumor cell isolate may exert on the control-tumor cell tumor-antigen expression.
  • Another control would include identically isolated and cultured cells, with the exception, for example, of the addition of tumor-antigen expression modulators such as OSM and/or MEKl/2, ERKl/2 inhibitors.
  • differential display is one of the techniques designed to identify genes that are differentially regulated by cells under various physiological or experimental conditions (for example, differentiation, carcinogenesis, pharmacologic treatment).
  • This technique was introduced by Liang and Pardee and described in U.S. Pat. No. 5,262,311. Prior to Liang and Pardee's introduction of this technique, those interested in identifying differentially expressed genes were compelled to resort either to differential hybridization screening (Zimmerman, G. R., et al., Cell, Vol. 21, pp.
  • Liang et al. describes a protocol which involves the reverse transcription of a messenger ribonucleic acid ('mRNA') population, in independent reactions, with each of twelve anchor primers (T ⁇ 2 MN), where M can be G (guanine), A (adenine) or C (cystosine) and N can be G, A, C or T (thymidine).
  • T ⁇ 2 MN twelve anchor primers
  • M can be G (guanine)
  • N can be G, A, C or T (thymidine).
  • the resulting single- stranded cDNAs are then amplified by the polymerase chain reaction (hereinafter, 'PCR') using the same anchor primer used for reverse transcription together with an upstream or 5' decamer of arbitrary sequence.
  • PCR products which are labeled by incorporation of tracer amounts of a radioactive nucleotide, are resolved for analysis by denaturating polyacrylamide gel electrophoresis (PAGE).
  • PAGE denaturating polyacrylamide gel electrophoresis
  • the invention also embraces solid-phase nucleic acid molecule arrays.
  • the array consists essentially of a set of nucleic acid molecules, expression products thereof, or fragments (of either the nucleic acid or the polypeptide molecule) thereof.
  • the set of nucleic acid molecules comprises nucleic acid molecules of a tumor cell known to express a tumor-antigen, said tumor cell previously contacted with a modulator of the MAPK/ERK signaling cascade.
  • the solid-phase nucleic acid molecule array further comprises at least one control set of nucleic acid molecules.
  • a preferred control set of nucleic acid molecules consists essentially of nucleic acid molecules, expression products thereof, or fragments thereof, fixed to a solid substrate, wherein the set of nucleic acid molecules comprises nucleic acid molecules of a tumor cell l ⁇ iown to express a tumor-antigen, said tumor cell not previously contacted with a modulator of the
  • microarray technology which is also known by other names including: DNA chip technology, gene chip technology, and solid-phase nucleic acid array technology, is well known to those of ordinary skill in the art and is based on, but not limited to, obtaining an array of identified nucleic acid probes (e.g., sets of antigen- expressing tumor cell-line specific cDNA molecules) on a fixed substrate, labeling target molecules with reporter molecules (e.g., radioactive, chemiluminescent, or fluorescent tags such as fluorescein, Cye3-dUTP, or Cye5-dUTP), hybridizing target nucleic acids to the probes, and evaluating target-probe hybridization.
  • reporter molecules e.g., radioactive, chemiluminescent, or fluorescent tags such as fluorescein, Cye3-dUTP, or Cye5-dUTP
  • a probe with a nucleic acid sequence that perfectly matches the target sequence will, in general, result in detection of a stronger reporter-molecule signal than will probes with less perfect matches.
  • Many components and techniques utilized in nucleic acid microarray technology are presented in The Chipping Forecast, Nature Genetics, Vol.21, Jan 1999, the entire contents of which is incorporated by reference herein.
  • microarray substrates may include but are not limited to glass, silica, aluminosilicates, borosilicates, metal oxides such as alumina and nickel oxide, various clays, nitrocellulose, or nylon. In all embodiments a glass substrate is preferred.
  • probes are selected from the group of nucleic acids including, but not limited to: DNA, genomic DNA, cDNA, and oligonucleotides; and may be natural or synthetic. Oligonucleotide probes preferably are 20 to 25-mer oligonucleotides and DNA/cDNA probes preferably are 500 to 5000 bases in length, although other lengths may be used.
  • probe length may be determined by one of ordinary skill in the art by following art-known procedures.
  • preferred probes are a set of nucleic acid molecules (e.g., cDNAs, RNAs, etc.) that comprise nucleic acid molecules of a tumor cell known to express a tumor-antigen, said tumor cell previously contacted with a modulator of the MAPK/ERK signaling cascade.
  • modulators of the MAPK ERK signaling cascade include modulators of MEKl/2 and ERKl/2 activity (e.g., MEKl/2 and ERKl/2 inhibitors). Probes may be purified to remove contaminants using standard methods known to those of ordinary skill in the art such as gel filtration or precipitation.
  • the microarray substrate may be coated with a compound to enhance synthesis of the probe on the substrate.
  • a compound to enhance synthesis of the probe on the substrate include, but are not limited to, oligoethylene glycols.
  • coupling agents or groups on the substrate can be used to covalently link the first nucleotide or olignucleotide to the substrate. These agents or groups may include, but are not limited to: amino, hydroxy, bromo, and carboxy groups. These reactive groups are preferably attached to the substrate through a hydrocarbyl radical such as an alkylene or phenylene divalent radical, one valence position occupied by the chain bonding and the remaining attached to the reactive groups.
  • hydrocarbyl groups may contain up to about ten carbon atoms, preferably up to about six carbon atoms.
  • Alkylene radicals are usually preferred containing two to four carbon atoms in the principal chain.
  • probes are synthesized directly on the substrate in a predetermined grid pattern using methods such as light-directed chemical synthesis, photochemical deprotection, or delivery of nucleotide precursors to the substrate and subsequent probe production.
  • the substrate may be coated with a compound to enhance binding of the probe to the substrate.
  • Such compounds include, but are not limited to: polylysine, amino silanes, amino-reactive silanes (Chipping Forecast, 1999) or chromium (Gwynne and Page, 2000).
  • presynthesized probes are applied to the substrate in a precise, predetermined volume and grid pattern, utilizing a computer- controlled robot to apply probe to the substrate in a contact-printing manner or in a non- contact manner such as ink jet or piezo-electric delivery.
  • Probes may be covalently linked to the substrate with methods that include, but are not limited to, UV-irradiation.
  • probes are linked to the substrate with heat.
  • Targets are nucleic acids selected from the group, including but not limited to: DNA, genomic DNA, cDNA, RNA, mRNA and may be natural or synthetic. In all embodiments, nucleic acid molecules from subjects suspected of developing or having a cardiovascular condition, are preferred. In certain embodiments of the invention, one or more control nucleic acid molecules are attached to the substrate. Preferably, control nucleic acid molecules allow determination of factors including but not limited to: nucleic acid quality and binding characteristics; reagent quality and effectiveness; hybridization success; and analysis thresholds and success. Control nucleic acids may include, but are not limited to, expression products of genes such as housekeeping genes or fragments thereof.
  • the expression data generated by, for example, microarray analysis of gene expression is preferably analyzed to determine which nucleic acids in differentially treated (e.g., with or without a MAPK/ERK signaling cascade modulator) tumor-antigen expressing cells, are significantly differentially expressed.
  • the significance of gene expression can be determined using Permax computer software, although any standard statistical package that can discriminate significant differences is expression may be used. Permax performs permutation 2-sample t-tests on large arrays of data. For high dimensional vectors of observations, the Permax software computes t-statistics for each attribute, and assesses significance using the permutation distribution of the maximum and minimum overall attributes.
  • the main use is to determine the attributes (genes) that are the most different between two groups (e.g., control untreated cell line and a MAPK/ERK signaling cascade modulator treated cell line, in the presence or absence of another agent, e.g., OSM), measuring "most different” using the value of the t-statistics, and their significance levels.
  • Expression of nucleic acid molecules can also be determined using protein measurement methods to determine expression of the set of nucleic acids by determining the expression of polypeptides encoded by the set of nucleic acids.
  • Preferred methods of specifically and quantitatively measuring proteins include, but are not limited to: mass spectroscopy-based methods such as surface enhanced laser desorption ionization (SELDI; e.g., Ciphergen ProteinChip System), non-mass spectroscopy-based methods, and immunohistochemistry-based methods such as 2-dimensional gel electrophoresis.
  • mass spectroscopy-based methods such as surface enhanced laser desorption ionization (SELDI; e.g., Ciphergen ProteinChip System), non-mass spectroscopy-based methods, and immunohistochemistry-based methods such as 2-dimensional gel electrophoresis.
  • SELDI methodology may, through procedures l ⁇ iown to those of ordinaiy skill in the art, be used to vaporize microscopic amounts of tumor protein and to create a "fingerprint" of individual proteins, thereby allowing simultaneous measurement of the abundance of many proteins in a single sample.
  • SELDI-based assays may be utilized to characterize cardiovascular conditions as well as stages of such conditions. Such assays preferably include, but are not limited to the following examples. Gene products discovered by RNA microarrays may be selectively measured by specific
  • SELDI protein disc e.g., selective SELDI
  • Gene products discovered by protein screening e.g., with 2-D gels, may be resolved by "total protein SELDI" optimized to visualize those particular nucleic acids of interest from among the set of nucleic acids.
  • TIL Tumor and Tumor Infiltrating Lymphocytes
  • TIL tumor infiltrating lymphocytes
  • TIL were cloned by limiting dilution using irradiated mononuclear feeder cells together with PHA as a polyclonal stimulus as previously described. Limiting dilution was performed on TIL which had been in culture for two weeks prior to cloning. A ⁇ minimum of 5 x 10 cells were utilized for functional assays and PCR analyses.
  • Tumor cell lines MU, MO, MA and EW were obtained from cutaneous metastatic melanoma deposits and some (MU, MO and MA), were previously described (Pandolfi F, et al., Clin. Exp. Immunol, 1994, 95:141-7; Pandolfi F, et al., Cancer Res, 1991, 51:3164-
  • Melan-A/MART-1 negative variant MU-X were obtained by culture of MU tumor cells at high density (>5xl0 cells/ml) for several days prior to immunoselection with anti- Melan- A/MART- 1 specific TIL. After 1 week of co-culture of tumor and lymphocytes in the presence of recombinant human IL-2, the tumor cells which propagate were collected and maintained in culture in the absence of T cells. The melanoma origin of the lines was confirmed using antibodies to melanoma-associated antigen S-100 and HMB-45 (Ordonez N, et al., Am J Clin Pathol, 1988, 90:385-90).
  • MU and MA tumor s expressed HLA- A2; MU tumor cells were also HLA-A1 positive.
  • MO tumor was derived from a patient whose leukocytes expressed HLA-A2, while MO tumor cells did not express this class I MHC antigen. Tumor cells were cultured either in DMEM supplemented with 10% human serum (MU), or in RPMI 1640 supplemented with 5% human serum (MA and MO). Expression of Melan-A/MART-1 by tumor cells
  • TIL were assayed for the ability to lyse melanoma target cells in 4 hour Cr- release assays as previously described (Hishii M, et al., Proc. Natl. AcadSci USA, 1997, 94: 1378-1383).
  • the melanoma target cells with high constitutive expression of Melan- A/MART-1 were generated by low density culture (1-2 x 10 /ml) were compared with respect to their susceptibility to cytolysis with the same cells cultured for 3 to 6 days in the presence of conditioned medium from the Melan-A/MART-1 negative variant, MU-X, to derive target cells with low Melan-A/MART-1 expression.
  • EBV-transformed B lymphocyte targets EBV-3 (HLA-A1, B8, DR3), EBV-19
  • Pulsing included the following melanocyte lineage-derived peptides: Tyrosinase (Rivoltini, L., et al., J. Immunol, 1995, 154:2257-2265): MLLAVLYCL (SEQ ID NO:2) or YMNGTMSQV(SEQ ID NO:3), MAGE-3 (Gaugler B, et al., JExp Med, 1994, 179:921-30): EVDPIGHLY
  • Clones were screened for cytotoxic activity at effector to target ratios of 50:1 and below.
  • MA/Ml Tumor-Conditioned Medium on Me ⁇ an-A/Mart-1
  • Oncostatin-M (OSM), and at least one additional soluble factor, which we designate
  • Oncostatin M is the subject of United States patent 5,618,715 to Shoyab et al., which is expressly incorporated herein by reference.
  • Anti- Oncostatin-M antibodies are l ⁇ iown in the art, and include Mouse and Goat anti-human Oncostatin M from Research Diagnostics, Flanders, NJ.
  • the loss of MA/Ml as shown in flow cytometry is associated with a marked diminution in the ability of T cells to lyse tumor cells which have been treated with MASA-containing supernatants or recombinant OSM.
  • the loss of T cell-mediated lysis can be overcome by the addition of the MA/Ml -derived peptide, AAGIGILTV, which restores the targets to cytolytic susceptibility.
  • the loss of MA/Ml is generally accompanied by diminished gplOO and tyrosinase, as well as other melanocyte lineage proteins, indicating that there is a 'sea- change' in the tumor cells.
  • the down-modulation of antigen expression appears to be somewhat selective as the HLA Class I antigen needed for presentation of the melanoma peptide is not down-modulated.
  • the MASA-containing conditioned medium was removed from the MA/Ml expressing tumor cells, there was renewed expression of this antigen.
  • These antigen positive cells are again lysed by MA/Ml -specific cytotoxic T cells.
  • tumors with low or absent MA/Ml are also relatively deficient in tyrosinase and gplOO as well, and 3 of 4 low-MA/Ml melanomas have low Mitf-M, including the MU-X line derived from MA/M1+ MU cells.
  • the soxlO regulator of Mitf- M expression is deficient in 2 of 4 of the low-MA/Ml melanomas, while another melanocyte-lineage transcription factor, tbx2, was deficient at the mRNA level only in the MA/Ml -low EW cell line (see Table 1 below).
  • OSM induces down-modulation of a variety of melanocyte-related genes, including
  • these agents can upregulate Melan-A/MART-1 expression in Melan- A/MART-1 expressing and deficient melanoma cells.
  • antigen-A/MART-1 As shown in Figure 3, antigen-A/MART-1
  • 5 positive cell lines, such as 453 A show dramatic up-regulation of Melan-A/MART-1, while the relatively deficient tumor cells, such as EW, IGR-39D and A375, show parallel, but more modest levels of Melan-A/MART-1 expression after MEKl/2 inhibitor treatment.
  • the up-regulation of Melan-A/MART-1 by MEK inhibitors is dose-dependent, and cellular toxicity limits the dose at which these inhibitors can be tested.
  • the up-regulation of Melan-A/MART-1 expression can also be demonstrated at the mRNA level, where both U0126 and PD98059 show a dose-dependent up-regulation of Melan-A/MART-1 mRNA transcription in both antigen-positive cells, such as MU ( Figure 4), and Melan-A MART-1 deficient tumor cells, MU-X ( Figure 5).
  • the up-regulation of Melan-A/MART-1 in deficient cells demonstrates that these antigen-negative cells have
  • the U0126-stimulated upregulation of mRNA and cytoplasmic protein expression in MA/Ml -deficient cell lines correlates with an increase in cytotoxic T cell recognition of tumor cells.
  • the MA/Ml -specific T cell clone is highly 30 effective, so that even at low effector:target ratios (2:1), it still shows almost maximal lysis of MU target cells.
  • the A375 are lysed only slightly (10%) at all ratios, while the addition of 40mM U0126 for 6 days enhanced the lysis of A375 cells to approximately 30%. Still, the lysis of treated A375 cells still did not approach the 70% lysis achieved against MA/Ml high expressing MU target cells.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pathology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Hospice & Palliative Care (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention a trait à des procédés pour l'identification d'agents modulateurs de l'expression des antigènes spécifiques de tumeurs dans des cellules tumorales. L'invention a également trait à des agents et des compositions pharmaceutiques contenant de tels agents modulateurs de l'expression des antigènes spécifiques de tumeurs dans des cellules tumorales. Par conséquent l'invention est particulièrement utile, entre autres, pour le traitement de sujets atteints de tumeurs solides autologues présentant des cellules exprimant, ou pouvant être induites à exprimer, des antigènes spécifiques de tumeurs. Une catégorie de matières selon l'invention est constituée par d'agents de régulation positive d'antigènes spécifiques de tumeurs. Ces agents comprennent des modulateurs de la cascade de signalisation de MAPK/ERKL, et plus spéficiquement des modulateurs de l'activité de MEK1/2 et de ERK1/2.
PCT/US2003/004786 2002-02-15 2003-02-18 Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs WO2004087941A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2003222221A AU2003222221A1 (en) 2002-02-15 2003-02-18 Map-kinase inhibitors as regulators of tumour-associated antigen expression
EP03717900A EP1509614A4 (fr) 2002-02-15 2003-02-18 Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs
CA 2475206 CA2475206A1 (fr) 2002-02-15 2003-02-18 Inhibiteurs de map-kinase comme regulateurs d'expression antigenique associe aux tumeurs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US35758602P 2002-02-15 2002-02-15
US60/357,586 2002-02-15

Publications (1)

Publication Number Publication Date
WO2004087941A1 true WO2004087941A1 (fr) 2004-10-14

Family

ID=33130319

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/004786 WO2004087941A1 (fr) 2002-02-15 2003-02-18 Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs

Country Status (4)

Country Link
EP (1) EP1509614A4 (fr)
AU (1) AU2003222221A1 (fr)
CA (1) CA2475206A1 (fr)
WO (1) WO2004087941A1 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006010628A1 (fr) * 2004-07-29 2006-02-02 Creabilis Therapeutics S.P.A. Emploi de k-252a et d'inhibiteurs de kinase pour la prevention ou le traitement de pathologies associees a hmgb1
WO2008082730A2 (fr) * 2006-09-19 2008-07-10 Novartis Ag Biomarqueurs de modulation cible, efficacité, diagnostic et/ou pronostic pour les inhibiteurs de la raf
WO2009069668A1 (fr) * 2007-11-28 2009-06-04 National University Corporation Nagoya University Agent pour augmenter l'expression d'un antigène du mélanome malin, et son utilisation
US7994185B2 (en) 2008-05-06 2011-08-09 Glaxo Smith Kline LLC Benzene sulfonamide thiazole and oxazole compounds
CN102274513A (zh) * 2011-07-06 2011-12-14 南开大学 ERK1/2抑制剂通过刺激巨噬细胞干扰素γ表达抑制肿瘤生长
WO2013001372A3 (fr) * 2011-06-30 2013-04-25 University Of Oslo Procédés et compositions pour inhiber l'activation des lymphocytes t régulateurs
CN103097546A (zh) * 2010-06-18 2013-05-08 许执恒 Mtea在诊断和治疗恶性肿瘤中的应用
EP2665835A4 (fr) * 2011-01-18 2016-07-20 Everist Genomics Inc Signature pronostique de la récurrence d'un cancer colorectal
WO2016168264A1 (fr) * 2015-04-13 2016-10-20 Kiromic, Llc Méthodes et compositions pour traiter un cancer avec des cellules dendritiques
US10000546B2 (en) 2013-03-13 2018-06-19 Health Research, Inc. Compositions and method for use of recombinant T cell receptors for direct recognition of tumor antigen

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5525625A (en) * 1995-01-24 1996-06-11 Warner-Lambert Company 2-(2-Amino-3-methoxyphenyl)-4-oxo-4H-[1]benzopyran for treating proliferative disorders

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU8856201A (en) * 2000-09-01 2002-03-13 Andel Inst Van Inhibition of mitogen-activated protein kinase (mapk) pathway: a selective therapeutic strategy against melanoma

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5525625A (en) * 1995-01-24 1996-06-11 Warner-Lambert Company 2-(2-Amino-3-methoxyphenyl)-4-oxo-4H-[1]benzopyran for treating proliferative disorders

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ENGLARO ET AL.: "Inhibition of the mitogen-activated protein kinase pathway triggers B16 melanoma cell differentiation", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 273, no. 16, 17 April 1998 (1998-04-17), pages 9966 - 9970, XP002225752 *
See also references of EP1509614A4 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006010628A1 (fr) * 2004-07-29 2006-02-02 Creabilis Therapeutics S.P.A. Emploi de k-252a et d'inhibiteurs de kinase pour la prevention ou le traitement de pathologies associees a hmgb1
WO2008082730A2 (fr) * 2006-09-19 2008-07-10 Novartis Ag Biomarqueurs de modulation cible, efficacité, diagnostic et/ou pronostic pour les inhibiteurs de la raf
WO2008082730A3 (fr) * 2006-09-19 2009-01-08 Novartis Ag Biomarqueurs de modulation cible, efficacité, diagnostic et/ou pronostic pour les inhibiteurs de la raf
JP2010504530A (ja) * 2006-09-19 2010-02-12 ノバルティス アーゲー Raf阻害剤の標的調節、効果、診断、および/または予後診断のためのバイオマーカー
WO2009069668A1 (fr) * 2007-11-28 2009-06-04 National University Corporation Nagoya University Agent pour augmenter l'expression d'un antigène du mélanome malin, et son utilisation
US8642759B2 (en) 2008-05-06 2014-02-04 Glaxosmithkline Llc Benzene sulfonamide thiazole and oxazole compounds
US7994185B2 (en) 2008-05-06 2011-08-09 Glaxo Smith Kline LLC Benzene sulfonamide thiazole and oxazole compounds
US8415345B2 (en) 2008-05-06 2013-04-09 Glaxo SmithKline LLC Benzene sulfonamide thiazole and oxazole compounds
US9233956B2 (en) 2008-05-06 2016-01-12 Novartis Ag Benzene sulfonamide thiazole and oxazole compounds
CN103097546A (zh) * 2010-06-18 2013-05-08 许执恒 Mtea在诊断和治疗恶性肿瘤中的应用
EP2665835A4 (fr) * 2011-01-18 2016-07-20 Everist Genomics Inc Signature pronostique de la récurrence d'un cancer colorectal
WO2013001372A3 (fr) * 2011-06-30 2013-04-25 University Of Oslo Procédés et compositions pour inhiber l'activation des lymphocytes t régulateurs
CN102274513A (zh) * 2011-07-06 2011-12-14 南开大学 ERK1/2抑制剂通过刺激巨噬细胞干扰素γ表达抑制肿瘤生长
US10000546B2 (en) 2013-03-13 2018-06-19 Health Research, Inc. Compositions and method for use of recombinant T cell receptors for direct recognition of tumor antigen
US11155595B2 (en) 2013-03-13 2021-10-26 Health Research, Inc. Compositions and methods for use of recombinant T cell receptors for direct recognition of tumor antigen
WO2016168264A1 (fr) * 2015-04-13 2016-10-20 Kiromic, Llc Méthodes et compositions pour traiter un cancer avec des cellules dendritiques

Also Published As

Publication number Publication date
EP1509614A1 (fr) 2005-03-02
EP1509614A4 (fr) 2007-09-12
AU2003222221A1 (en) 2004-10-25
CA2475206A1 (fr) 2003-08-15

Similar Documents

Publication Publication Date Title
Schwitalle et al. Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers
Carrascal et al. Sialyl Tn-expressing bladder cancer cells induce a tolerogenic phenotype in innate and adaptive immune cells
EP3182994B1 (fr) Récepteur d'antigène chimérique spécifique de l'antigène ssea4
US20210363215A1 (en) T cell receptors with mage-b2 specificity and uses thereof
AU2014298504B2 (en) Tumor antigens for determining cancer therapy
US8153421B2 (en) Prostate stem cell
AU2010215691B2 (en) Methods and compositions for diagnosis and treatment of cancer
EP3590530A1 (fr) Récepteurs de lymphocytes t spécifiques aux antigènes et épitopes de lymphocytes t
AU2014275166A1 (en) Compositions and methods for identification, assessment prevention, and treatment of cancer using PD-L1 isoforms
EP2441468A2 (fr) Immunothérapie par l'utilisation de cellules capable de co-exprimer l'antigène cible et le CD1d et impulsées par le ligand CD1d
US20230330145A1 (en) Natural killer cells
WO2004087941A1 (fr) Inhibiteurs de la proteine kinase associee aux membranes en tant que regulateurs de l'expression de l'antigene specifique de tumeurs
EP3027203B1 (fr) Antigenes de tumeur pour determiner la therapie du cancer
KR20220095112A (ko) Mdsc 활성 저해 약물의 선별 방법
Wang et al. Expression of the human Sda β-1, 4-N-acetylgalactosaminyltransferase II gene is dependent on the promoter methylation status
WO2001036472A2 (fr) Compositions et methodes permettant de reguler l'expression des antigenes specifiques de tumeurs
CN108697762A (zh) 治疗癌症的组合物和方法
US20210169913A1 (en) Fcrl6 and its uses related to cancer
Voigt et al. CD28-mediated costimulation impacts on the differentiation of DC vaccination-induced T cell responses
WO2018175673A1 (fr) Méthodes et compositions de détection et de traitement de cancer
EP2249159A1 (fr) Identification des marqueurs associés aux tumeurs pour diagnostic et thérapie
CN104640568B (zh) 用于治疗mds的cd95信号传导途径的抑制剂
中山和典 NFκB and TGFβ contribute to the expression of PTPN3 in activated human lymphocytes
Schröder Tumor-specific CD8 T cells in a primary human breast carcinoma: a detailed analysis
KR20230004458A (ko) Apc-결핍 암의 치료를 위한 방법 및 조성물

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2475206

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003222221

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003717900

Country of ref document: EP

Ref document number: 535004

Country of ref document: NZ

AK Designated states

Kind code of ref document: A1

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 SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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
WWP Wipo information: published in national office

Ref document number: 2003717900

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Ref document number: JP