WO2008070100A1 - Compositions comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique ou leurs sels et leurs procédés d'utilisation - Google Patents

Compositions comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique ou leurs sels et leurs procédés d'utilisation Download PDF

Info

Publication number
WO2008070100A1
WO2008070100A1 PCT/US2007/024873 US2007024873W WO2008070100A1 WO 2008070100 A1 WO2008070100 A1 WO 2008070100A1 US 2007024873 W US2007024873 W US 2007024873W WO 2008070100 A1 WO2008070100 A1 WO 2008070100A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
akt
administered
erlotinib
compounds
Prior art date
Application number
PCT/US2007/024873
Other languages
English (en)
Other versions
WO2008070100A8 (fr
Inventor
Jin Q. Cheng
Said M. Sebti
Original Assignee
University Of South Florida
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 University Of South Florida filed Critical University Of South Florida
Publication of WO2008070100A1 publication Critical patent/WO2008070100A1/fr
Publication of WO2008070100A8 publication Critical patent/WO2008070100A8/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This application relates to combination therapies including triciribine compounds and epidermal growth factor receptor inhibitor compounds, particularly erlotinib-like compounds and compositions with reduced toxicity for the treatment and prevention of tumors, cancer, and other disorders associated with abnormal cell proliferation.
  • Cancer is an abnormal growth of cells. Cancer cells rapidly reproduce despite restriction of space, nutrients shared by other cells, or signals sent from the body to stop reproduction. Cancer cells are often shaped differently from healthy cells, do not function properly, and can spread into many areas of the body. Abnormal growths of tissue, called tumors, are clusters of cells that are capable of growing and dividing uncontrollably. Tumors can be benign (noncancerous) or malignant (cancerous). Benign tumors tend to grow slowly and do not spread. Malignant tumors can grow rapidly, invade and destroy nearby normal tissues, and spread throughout the body. [004] Cancers are classified according to the kind of fluid or tissue from which they originate, or according to the location in the body where they first developed.
  • cancers are of mixed types. Cancers can be grouped into five broad categories, carcinomas, sarcomas, lymphomas, leukemias, and myelomas, which indicate the tissue and blood classifications of the cancer.
  • Carcinomas are cancers found in body tissue known as epithelial tissue that covers or lines surfaces of organs, glands, or body structures. For example, a cancer of the lining of the stomach is called a carcinoma. Many carcinomas affect organs or glands that are involved with secretion, such as breasts that produce milk. Carcinomas account for approximately eighty to ninety percent of all cancer cases.
  • Sarcomas are malignant tumors growing from connective tissues, such as cartilage, fat, muscle, tendons, and bones.
  • sarcoma a tumor on the bone, usually occurs in young adults.
  • sarcoma include osteosarcoma (bone) and chondrosarcoma (cartilage).
  • Lymphoma refers to a cancer that originates in the nodes or glands of the lymphatic system, whose job it is to produce white blood cells and clean body fluids, or in organs such as the brain and breast. Lymphomas are classified into two categories: Hodgkin's lymphoma and non-Hodgkin's lymphoma.
  • Leukemia also known as blood cancer, is a cancer of the bone marrow that keeps the marrow from producing normal red and white blood cells and platelets. White blood cells are needed to resist infection.
  • Red blood cells are needed to prevent anemia. Platelets keep the body from easily bruising and bleeding.
  • leukemia include acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia.
  • myelogenous and lymphocytic indicate the type of cells that are involved.
  • myeloma cells collect in many bones, forming many bone tumors. This is called multiple myeloma.
  • Tumor induction and progression are often the result of accumulated changes in the tumor-cell genome.
  • Such changes can include inactivation of cell growth inhibiting genes, or tumor suppressor genes, as well as activation of cell growth promoting genes, or oncogenes.
  • Hundreds of activated cellular oncogenes have been identified to date in animal models, however, only a small minority of these genes have proven to be relevant to human cancers (Weinberg et al., Oncogenes and the Molecular Origins of Cancer. 1989. Cold Spring Harbor, NY; Stanbridge J. et al., Cell, 1990. 63: p 867-874; Godwin et al., in Gynecological oncology: principles and practice.
  • oncogenes in human cancers can result from factors such as increased gene copy number or structural changes. These factors can cause numerous cellular effects, for example, they can result in overexpression of a gene product.
  • oncogenes involved in human cancer can be activated through gene overexpression.
  • Receptor tyrosine kinases such as EGFR, ErbB2, VEGFR and insulin-like growth factor I receptor (IGF-IR) are intimately involved in the development of many human cancers including colorectal pancreatic, breast and ovarian cancers (Khaleghpour K. et al., Carcinogenesis, 2004. 25(2): p. 241-8.; Sekharam M. et al., Cancer Res, 2003. 63(22): p. 7708-16).
  • Binding of ligands such as EGF, VEGF and IGF-I to their receptors promotes stimulation of the intrinsic tyrosine kinase activity, autophosphorylation of specific tyrosines in the cytoplasmic domain of the receptors and recruitment of signaling proteins that trigger a variety of complex signal transduction pathways (Olayioye M.A. et al., Embo J, 2000. 19(13): p. 3159-67; Porter A.C. and Vaillancourt R.R., Oncogene, 1998. 17(11 Reviews): p. 1343-52).
  • Akt is a serine/threonine protein kinase (also known as PKB), which has 3 family members Aktl, Akt2 and Akt3. Stimulation of cells with growth or survival factors results in recruitment to the receptors of the lipid kinase phosphoinositide-3-OH-
  • PI3K phosphoinositol-4,5-biphosphate
  • Akt phosphatase PTEN dephophorylates PIP3 to PIP2 and hence prevents the activation of Akt.
  • the majority of human cancers contain hyperactivated Akt (Datta S.R.et al., Genes Dev, 1999. 13(22): p. 2905-27; Bellacosa A. et al., Int J Cancer, 1995. 64(4): p. 280-5; Sun M. et al., Am J Pathol, 2001. 159(2): p. 431-7).
  • Akt is overexpressed and/or hyperactivated in 57%, 32%, 27% and 36% of human colorectal, pancreatic, breast and ovarian cancers, respectivel (Roy H.K. et al., Carcinogenesis, 2002. 23(1): p. 201-5; Altomare D.A. et al., J Cell Biochem, 2003. 88(1): p. 470-6; Sun M. et al., Cancer Res, 2001. 61(16): p. 5985-91; Stal O. et al., Breast Cancer Res, 2003. 5(2): p. R37-44; Cheng J.Q. et al., Proc Natl Acad Sci U S A, 1992.
  • Akt Hyperactivation of Akt is due to amplification and/or overexpression of Akt itself as well as genetic alterations upstream of Akt including overexpression of receptor tyrosine kinases and/or their ligands and deletion of the phosphatase PTEN (Khaleghpour K. et al., Carcinogenesis, 2004. 25(2): p. 241-8; Sekharam M. et al., Cancer Res, 2003. 63(22): p. 7708-16; Cohen B.D. et al., Biochem Soc Symp, 1998.
  • a combination therapy including a triciribine compound and erlotinib-like compound hold promise as a potential therapy for treating tumors, cancer, and abnormal cell proliferation while synergistically reducing toxicity or adverse side effects caused by currently administered cancer compounds.
  • the present invention provides novel therapeutic regimens of triciribine, triciribine phosphate and related compounds in combination with one or more erlotinib- like compounds to treat tumors or cancer in a subject while limiting systemic toxicity.
  • the invention is based on the discovery that tumors or cancers, which overexpress Akt kinase are particularly sensitive to the cytotoxic effects of TCN and related compounds and a synergistic affect would arise with a combination of erlotinib-like compounds.
  • the invention encompasses a composition including:
  • each R2', R3' and R5' are independently hydrogen, optionally substituted phosphate or phosphonate (including mono-, di-, or triphosphate or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); amide, sulfonate ester including alkyl or arylalkyl; sulfonyl, including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as for example as described in the definition of an aryl given herein; optionally substituted arylsulfonyl; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; or cholesterol; or other pharmaceutically acceptable leaving group that, in vivo, provides a compound wherein R2', R3' or R5' is independently H or mono-, di- or tri-phosphate;
  • Rx and Ry are independently hydrogen, optionally substituted phosphate; acyl (including lower acyl); amide, alkyl (including lower alkyl); aromatic, polyoxyalkylene such as polyethyleneglycol, optionally substituted arylsulfonyl; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; or cholesterol; or
  • the compound is administered as a 5'-phosphoether lipid or a 5 '-ether lipid.
  • Rl and R2 each are independently H, optionally substituted straight chained, branched or cyclic alkyl (including lower alkyl), alkenyl, or alkynyl, CO-alkyl, CO- alkenyl, CO-alkynyl, CO-aryl or heteroaryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, sulfonyl, alkylsulfonyl, arylsulfonyl, aralkylsulfonyl; [0018] (ii) erlotinib-like compounds as in formula V:
  • each Rl and R2 is independently hydrogen, independently optionally substituted alkoxy, optionally substituted amine, aromatic amine, heteroaromatic amine, optionallysubstituted straight chained, branched or cyclic alkyl;
  • each R3 and R4 is independently hydrogen, independently optionally substituted aromatic amine, heteroaromatic amine, or cyclic amine;
  • the invention encompasses a method of treating a tumor or cancer in a mammal including administering to the mammal an effective amount of a composition including: [0024] (i) a compound of formula I-IV:
  • each R2', R3' and R5' are independently hydrogen, optionally substituted phosphate or phosphonate (including mono-, di-, or triphosphate or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower
  • Rx and Ry are independently hydrogen, optionally substituted phosphate; acyl (including lower acyl); amide, alkyl (including lower alkyl); aromatic, polyoxyalkylene such as polyethyleneglycol, optionally substituted arylsulfonyl; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; or cholesterol; or other pharmaceutically acceptable leaving group.
  • the compound is administered as a 5'-phosphoether lipid or a 5 '-ether lipid.
  • Rl and R2 each are independently H, optionally substituted straight chained, branched or cyclic alkyl (including lower alkyl), alkenyl, or alkynyl, CO-alkyl, CO- alkenyl, CO-alkynyl, CO-aryl or heteroaryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, sulfonyl, alkylsulfonyl, arylsulfonyl, aralkylsulfonyl; and [0028] (ii) erlotinib-like compounds as in formula V:
  • each Rl and R2 is independently hydrogen, independently optionally substituted alkoxy, optionally substituted amine, aromatic amine, heteroaromatic amine, optionally substituted straight chained, branched or cyclic alkyl;
  • each R3 and R4 is independently hydrogen, independently optionally substituted aromatic amine, heteroaromatic amine, or cyclic amine.
  • Methods are useful to treat tumors and cancers that are particulary susceptible to the toxic effects of TCN, TCN-P and/or related compounds.
  • methods are provided for treating a tumor in a mammal, particularly a human that includes (i) obtaining a biological sample from the tumor; (ii) determining whether the tumor overexpresses an Akt kinase, and (iii) treating the tumor that overexpresses Akt kinase with triciribine, triciribine phosphate or a related compound in combination with one or more erlotinib-like compounds, as described herein.
  • the level of Akt kinase expression can be determined by assaying the tumor or cancer for the presence of a phosphorylated Akt kinase, for exmple, by using an antibody that can detect the phosphorylated form.
  • the level of Akt expression can be determined by assaying a tumor or cancer cells obtained from a subject and comparing the levels to a control tissue.
  • the Akt can be overexpressed at least 2, 2.5, 3 or 5 fold in the cancer sample compared to the control.
  • the overexpressed Akt kinase can be a hyperactivated and phosphorylated Akt kinase.
  • dosing regimens are provided that limit the toxic side effects of TCN and related compounds.
  • such dosing regimens minimize or eliminate toxic side effects, including, but not limited to, hepatoxicity, thrombocytopenia, hyperglycemia, vomiting, hypocalcemia, anemia,
  • TCN, TCN-P or related compounds provides at least a partial, such as at least 15, 20 or 30%, or complete response in vivo in at least 15, 20, or 25 % of the subjects.
  • a method is provided to treat a subject who has been diagnosed with a tumor by administering to the subject an effective amount of TCN, TCN-P or a related compound and one or more erlotinib-like compounds according to a dosing schedule that includes administering the the triciribine compound and erlotinib- like compounds approximately one time per week for approximately three weeks followed by a one week period wherein the triciribine compound and erlotinib-like compounds are not administered.
  • methods are provided to treat tumor or cancer in a subject by administering to the subject a dosing regimen of 10 mg/m2 or less of TCN, TCN-P or a related compound and one or more erlotinib-like compounds each one time per week.
  • the triciribine compound and one or more erlotinib-like compounds can be administered as a single bolus dose over a short period of time, for example, about 5, 10 or 15 minutes.
  • dosing schedules are provided in which the triciribine compound and one or more erlotinib-like compounds are administered via continuous infusion for at least 24, 48, 72, 96, or 120 hours.
  • the continuous administration can be repeated at least once a week, once every two weeks and/ or once a month.
  • the triciribine compound and one or more erlotinib-like compounds can be administered at least once every three weeks.
  • the compounds can be administered at least once a day for at least 2, 3, 4 or 5 days.
  • the triciribine compound and one or more erlotinib-like compounds as disclosed herein can be administered to patients in an amount that is effective in causing tumor regression.
  • the administration the triciribine compound and one or more erlotinib-like compounds can provide at least a partial, such as at least 15, 20 or 30%, or complete response in vivo in at least 15-20% of the subjects.
  • at least 2, 5, 10, 15, 20, 30 or 50 mg/m2 of the triciribine compound and at least about 10, 25, 50, 75, 100, 150, 200, 250 or 500 mgone or more erlotinib-like compounds disclosed herein can be administered to a subject.
  • the administration of the triciribine compound and one or more erlotinib-like compounds can be conducted according to any of the therapeutic regimens disclosed herein.
  • the dosing regimen can include administering less than 20 mg/m2 of the triciribine compound and one or more erlotinib-like compounds. In one embodiment, less than 10 mg/m2 of the triciribine compound and less than about 200 mg of one or more erlotinib-like compounds can be administered once a week.
  • dosages of or less than 2 mg/m2, 5 mg/m2, 10 mg/m2, and/or 15 mg/m2 of the triciribine compound and 10 mg, 20 mg, 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, or 500 mg of one or more erlotinib-like compounds can be administered to a subject.
  • less than 10 mg/m2 of the triciribine compound and less than 150 mg of the erlotinib compound can be administered to a subject via continuous infusion for at least five days.
  • the triciribine compound and one or more erlotinib-like compounds as disclosed herein can be used for the treatment of pancreatic, prostate, colo-rectal and/or ovarian cancer.
  • the triciribine compound and one or more erlotinib-like compounds and/or therapeutic regimens of the present invention can be used to prevent
  • the triciribine compound and one or more erlotinib-like compounds can be used to treat solid tumors.
  • the triciribine compound and one or more erlotinib-like compounds and compositions disclosed herein can be used for the treatment of a tumor or cancer, such as, but not limited to cancer of the following organs or tissues: breast, prostate, bone, lung, colon, including, but not limited to colorectal, urinary, bladder, non-Hodgkin lymphoma, melanoma, kidney, renal, pancreas, pharnx, thyroid, stomach, brain, and/or ovaries.
  • the triciribine compound and one or more erlotinib-like compounds can be used for the treatment of pancreatic, breast, colorectal and/or ovarian cancer.
  • the triciribine compound and one or more erlotinib-like compounds disclosed herein can be used in the treatment of angiogenesis-related diseases.
  • methods are provided to treat leukemia via continuous infusion of , the triciribine compound and one or more erlotinib-like compounds via continuous infusion for at least 24, 48, 72 or 96 hours.
  • the continuous infusion can be repeated, for example, at least once every two, three or four weeks.
  • a method for the treatment of tumors, cancer, and others disorders associated with an abnormal cell proliferation in a host includes administering to the host an effect amount of the triciribine compound and one or more erlotinib-like compounds optionally in combination with a pharmaceutically acceptable carrier.
  • the triciribine compound and one or more erlotinib-like compounds and compositions can be administered in combination and can form part of
  • the triciribine compound and one or more Erlotinib-like compounds as disclosed herein can be used to treat tumors or cancers resistant to one or more known anti-cancer drugs, including the embodiments of tumors or cancers and compounds disclosed herein.
  • the triciribine compound and one or more erlotinib-like compounds as disclosed herein is administered in an effective amount for the treatment of a patient with a drug resistant tumor or cancer, for example, multidrug resistant tumors or cancer including, but not limited to, those resistant to taxol alone, rapamycin, tamoxifen, cisplatin, and/ or gefitinib (iressa).
  • a method including administering to a host in need thereof an effective amount of a triciribine compound and one or more erlotinib- like compounds disclosed herein, or pharmaceutical composition including a triciribine compound and one or more erlotinib-like compounds, in an effective amount for the treatment of the treatment of tumors, cancer, and others disorders associated with an abnormal cell proliferation in a host.
  • a method for the treatment of a tumor or cancer including an effective amount of a compound disclosed herein, or a salt, isomer, prodrug or ester thereof, to an individual in need thereof, wherein the cancer is for example, carcinoma, sarcoma, lymphoma, leukemia, or myeloma.
  • the compound, or salt, isomer, prodrug or ester thereof is optionally provided in a pharmaceutically acceptable composition including the appropriate carriers, such as water, which is formulated for the desired route of administration to an individual in need thereof.
  • the compound is administered in combination or alternation with at least one additional therapeutic agent for the treatment of tumors or cancer.
  • a compound disclosed herein or a salt, prodrug or ester thereof in the treatment of a tumor or cancer, optionally in a pharmaceutically acceptable carrier; and the use of a triciribine compound and one or more erlotinib-like compounds disclosed herein or a salt, prodrug or ester thereof in the manufacture of a medicament for the treatment of cancer or tumor, optionally in a pharmaceutically acceptable carrier.
  • Figure 1 demonstrates the identification of API-2 (triciribine) as a candidate of Akt inhibitor from the NCI Diversity Set.
  • Figure IA illustrates the chemical structure of API-2 (triciribine).
  • Figure IB demonstrates that API-2 inhibits phosphorylation levels of AKT2 in AKT2-transformed NIH3T3 cells.
  • Wile type AKT2-transformed NIH3T3 cells were treated with API-2 (1 ⁇ M) for indicated times and subjected to immunoblotting analysis with anti-phospho-Akt-T308 and -S473 antibodies (top and middle panels). The bottom panel shows expression of total AKT2.
  • Figure 1C it is shown that API-2 inhibits three isoforms of Akt.
  • HEK293 cells were transfected with HA-Aktl , -AKT2 and -AKT3 and treated with API-2 (1 ⁇ M) or wortmannin (15 ⁇ M) prior to EGF stimulation, the cells were lysed and immunoprecipitated with anti-HA antibody. The immunoprecipitates were subjected to in vitro kinase assay (top) and immunoblotting analysis with anti-phospho-Akt-T308 (bottom) antibody. Middle panel shows expression of transfected Akt 1, AKT2 and AKT3. Figure ID illustrates that API- 2 did not inhibit Akt in vitro. In vitro kinase assay of constitutively active AKT2 recombinant protein in a kinase buffer containing 1 ⁇ M API-2 (lane 3).
  • Figure 2 demonstrates that API-2 does not inhibit PI3K, PDKl and the closely related members of AGC kinase family.
  • Figure 2A demonstrates an in vitro PI3K kinase assay.
  • HEK293 cells were serum-starved and treated with API-2 (1 ⁇ M) or Wortmannin (15 ⁇ M) fro 30 minutes prior to EGF stimulation. Cells were lysed and immunoprecipitated with anti-pllO ⁇ antibody. The immunoprecipitates were subjected to in vitro kinase assay using PI-4-P as substrate.
  • Bottom panels are immunoblotting analysis of HEK293 cells that were transfected with Myc-PDKl and treated with wortmannin or API-2 prior to EGF stimulation. The immunoblots were detected with indicated antibodies.
  • Figure 2C illustrates an immunoblot analysis of phosphorylation levels of PKC D with anti-phospho-PKC ⁇ T638 (top) and total PKC ⁇ (bottom) antibodies following treatment with API-2 or a nonselective PKC inhibitor Ro31-8220.
  • FIG. 2D shows an in vitro SGK kinase assay.
  • HEK293 cells were transfected with HA-SGK and treated with API-2 or wortmannin prior to EGF stimulation.
  • In vitro kinase was performed with HA-SGK immunoprecipitates using MBP as substrate (top).
  • Bottom panel shows the expression of transfected HA-SGK.
  • Figure 2E illustrates the results of a PKA kinase assay. Immuno-purified PKA was incubated in ADB buffer (Upstate Biotechnology Inc) containing indicated inhibitors (API-2 or PKAI) and substrate Kemptide. The kinase activity was quantified.
  • ADB buffer Upstate Biotechnology Inc
  • PKAI indicated inhibitors
  • FIG. 2F a western blot is shown.
  • OVCAR3 cells were treated with API-2 for indicated times. Cell lysates were immunoblotted with indicated anti-phospho-antibodies (panels 1-4) and anti-
  • Figure 3 demonstrates that API-2 inhibits Akt activity and cell growth and induces apoptosis in human cancer cells with elevated Akt.
  • Figure 3A is a western blot, following treatment with API-2, phosphorylation levels of Akt were detected with anti- phospho-Akt-T308 antibody in indicated human cancer cell lines. The blots were reprobed with anti-total Akt antibody (bottom panels).
  • Figure 3B a cell proliferation assay is shown. Cell lines as indicated in the figure were treated with different doses of API-2 for 24 h and 48 h and then analyzed with CellTiter 96 Cell Proliferation Assay kit (Promega).
  • Figure 3C provides an apoptosis analysis.
  • FIG. 4 shows that API-2 inhibits downstream targets of Akt and exhibits antitumor activity in cancer cell lines with elevated Akt in mouse xenograft.
  • Figure 4A it is demonstrated that API-2 inhibits Akt phosphorylation of tuberin, Bad, AFX and GSK-3 ⁇ .
  • OVAR3 cells were lysed and immunoblotted with indicated antibodies.
  • Figure 4B shows that API-2 inhibits tumor growth. Tumor cells were subcutaneously injected into nude mice with low level of Akt cells on left side and elevated level of Akt cells on right side.
  • FIG. 4C illustrates a representation of the mice with OVCAR3 (right) and OVCAR5 (left) xenograft treated with API-2 or vehicle (control).
  • Figure 4D shows examples of tumor size (bottom) and weight (top) at the end of experiment.
  • Figure 4E immunoblot analysis of tumor lysates was performed with anti-phospho-Akt-S473 (top) and anti-AKT2 (bottom) antibodies in O VC AR-3 -derived tumors that were treated (T3 and T4) and untreated (Tl and T2) with API-2.
  • Figure 5 shows that API-2 (triciribine) inhibits Akt kinase activity in vitro.
  • API-2 triciribine
  • In vitro kinase assay was performed with recombinant of PDKl and Akt in a kinase buffer containing phosphatidylinositol-3,4,5-P3 (PIP3), API-2 and histone H2B as substrate.
  • PIP3 phosphatidylinositol-3,4,5-P3
  • Figure 6 provides the mRNA and amino acid sequence of human Aktl, restriction enzyme sites are also noted.
  • Figure 7 provides the mRNA and amino acid sequence of human Akt2 restriction enzyme sites are also noted.
  • Figure 8 provides the mRNA and amino acid sequence of human Akt3 restriction enzyme sites are also noted.
  • Figure 9 shows growth inhibition by the combination of trastuzumab and Akt/mTOR pathway inhibitors. PTEN anti-sense or non-specific oligonucleotide transfected BT474.ml cells were treated with inhibitors of the Akt/mTOR pathway alone or in combination with trastuzumab and the relative cell growth was assessed.
  • Figure 9A shows a panel of Akt/mTOR inhibitors. Growth inhibition was assessed in PTEN AS transfected BT474.ml cells.
  • TCN Triciribine
  • RADOOl 0.2 nM
  • QLT0267 10 ⁇ M
  • KP 372-1 0.05 ⁇ M
  • 4ADPIB 4ADPIB 5 ⁇ M
  • Edelfosine 7.5 ⁇ M 7.5 ⁇ M
  • trastuzumab Ttzm 2 ⁇ g/ml.
  • SD standard deviation
  • Results shown are the combined data from 2-3 experiments with triplicates of each treatment within each experiment.
  • Figure 9B shows TCN inhibits cell growth in combination with trastuzumab.
  • BT474.ml cells were transfected with PTEN AS oligonucleotide or non-specific (NS) oligonucleotide, treated with trastuzumab and TCN, alone and in combination, at multiple doses of TCN and
  • Figure 7C shows RADOOl inhibits cell growth in combination with trastuzumab.
  • BT474.ml cells were transfected with PTEN AS oligonucleotide or non-specific (NS) oligonucleotide, treated with trastuzumab and RADOOl, alone and in combination, at multiple doses of RADOOl and assayed for growth inhibition.
  • Figures 9B and 9C * indicates a significant difference in growth inhibition following combination treatment as compared to either trastuzumab or TCN/TADOOl alone. P ⁇ 0.05 was considered significant. Error bars depict the SEM.
  • FIG. 10 shows the synergistic effects on apoptosis. Twenty-four hours after plating, PTEN AS and NS transfected BT474.ml cells were treated as indicated with trastuzumab (Ttzm), TCN and/or RADOOl at the following concentrations: trastuzumab 2 ⁇ g/ml; triciribine 2.5 ⁇ M; RADOOl 0.4 nM. Apo-BrdU Tunel assays were performed to assess apoptosis. The experiment was performed 3 times and the data shown is the mean apoptosis.Error bars depitc the standard deviation. Trastuzumab + triciribine treatment significantly induced apoptosis (p ⁇ 0.01) as compared with all other treatments.
  • FIG 11 shows inhibition of Akt and p70S6K activity.
  • PTEN AS and NS oligonucleotides were transfected int BT474.ml cells. Two days later, the cells were treated for 2 hours with trastuzumab and triciribine (TCN) ( Figure 1 IA) or trastuzumab and RADOOl ( Figure 1 IB). Total cell lysates were collected, separated by SDS-PAGE and immunoblotted as indicated. The concentration of trastuzumab was 2 ⁇ g/ml, triciribine was 2.5 ⁇ M and RADOOl was 0.4 nM. The experiments were repeated at least twice to insure that results were reproducible.
  • Figure 12 shoes combination treatments inhibited tumor growth in a SCID mice xenograft model.
  • SCID mice received BT474.ml breast cancer cell xenografts in mammary fat pad. The xenografts grew for 3 weeks to generate tumors with an average size of 100-150 mm 3 .
  • PTEN antisense oligonucleotides, trastuzumab, triciribine ( Figure 12A) and RADOOl ( Figure 12B) were administered. The tumors were measured twice weekly with- calipers and tumor size was averaged for each treatment group. Error bars denote the standard error of the mean. * indicates a significant difference in growth inhibition following combination treatment as compared to either trastuzumab (Ttzm), TCN or DMSO alone. P ⁇ 0.05 was considered significant.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth, i.e., proliferative disorders.
  • proliferative disorders include cancers such as carcinoma, lymphoma, blastoma, sarcoma, and leukemia, as well as other cancers disclosed herein.
  • cancers include breast cancer, prostate cancer, colon cancer, squamous cell cancer, small-cell lung cancer, non- small cell lung cancer, gastrointestinal cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, e.g., hepatic carcinoma, bladder cancer, colorectal cancer, endometrial carcinoma, kidney cancer, and thyroid cancer.
  • cancers are basal cell carcinoma, biliary tract cancer;; bone cancer; brain and CNS cancer; choriocarcinoma; connective tissue cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; intraepithelial neoplasm; larynx cancer; lymphoma including Hodgkin's and Non-Hodgkin's lymphoma; melanoma; myeloma; neuroblastoma; oral cavity cancer (e.g., lip, tongue, mouth, and pharynx); pancreatic cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; sarcoma; skin cancer; stomach cancer; testicular cancer; uterine cancer; cancer of the urinary system, as well as other carcinomas and sarcomas.
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • a particular cancer may be characterized by a solid mass tumor.
  • the solid tumor mass if present, may be a primary tumor mass.
  • a primary tumor mass refers to a growth of cancer cells in a tissue resulting from the transformation of a normal cell of that tissue. In most cases, the primary tumor mass is
  • alkyl includes a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of for example Ci to C 24 , and specifically includes methyl, trifluoromethyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, f-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3- dimethylbutyl.
  • the alkyl is optionally substituted, e.g., with one or more substituents such as halo (F, Cl, Br or I), (e.g. CF 3 , 2-Br-ethyl, CH 2 F, CH 2 Cl, CH 2 CF 3 or CF 2 CF 3 ), hydroxyl (e.g. CH 2 OH), amino (e.g. CH 2 NH 2 , CH 2 NHCH 3 or CH 2 N(CH 3 ) 2 ), alkylamino, arylamino, alkoxy, aryloxy, nitro, azido (e.g. CH 2 N 3 ), cyano (e.g.
  • alkyl refers to a Ci to C 4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms.
  • alkylamino or arylamino includes an amino group that has one or two
  • amino acid includes naturally occurring and synthetic ⁇ , ⁇ , ⁇ or ⁇ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine.
  • the amino acid is in the L- configuration.
  • the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, ⁇ -alanyl, ⁇ -valinyl, ⁇ -leucinyl, ⁇ -isoleuccinyl, ⁇ -prolinyl, ⁇ -phenylalaninyl, ⁇ -tryptophanyl, ⁇ -methioninyl, ⁇ -glycinyl, ⁇ -serinyl, ⁇ -threoninyl, ⁇ - cysteinyl,
  • amino acid When the term amino acid is used, it is considered to be a specific and independent disclosure of each of the esters of a natural or synthetic amino acid, including but not limited to ⁇ , ⁇ , ⁇ or ⁇ glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine in the D and L-configurations.
  • ⁇ , ⁇ , ⁇ or ⁇ glycine alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and his
  • the term "protected” as used herein and unless otherwise defined includes a group that is added to an oxygen, nitrogen, sulfur or phosphorus atom to prevent its further reaction or for other purposes.
  • oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis (see Greene and Wuts, Protective Groups in Organic Synthesis. 1999, 3d Ed., John Wiley & Sons, Inc., NY).
  • aryl as used herein, and unless otherwise specified, includes phenyl,
  • the aryl group is optionally substituted with one or more moieties such as halo, hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene et al., Protective Groups in Organic Synthesis. 1999, 3 rd Ed., John Wiley and Sons, NY.
  • alkaryl or alkylaryl includes an alkyl group with an aryl substituent.
  • aralkyl or arylalkyl includes an aryl group with an alkyl substituent.
  • halo as used herein, includes chloro, bromo, iodo, and fluoro.
  • acyl includes a carboxylic acid ester in which the non-carbonyl moiety of the ester group is selected from straight, branched, or cyclic alkyl or lower alkyl, alkoxyalkyl including methoxymethyl, aralkyl including benzyl, aryloxyalkyl such as phenoxymethyl, aryl including phenyl optionally substituted with halogen, Ci to C 4 alkyl or Ci to C 4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxytrityl, substituted benzyl, trialkylsilyl (e.g.
  • the term “isolated” refers to a compound composition that includes at least 85% or 90% by weight, preferably 95% to 98 % by weight, and even more preferably 99% to 100% by weight, of the compound, the remainder comprising other chemical species or enantiomers.
  • the term “independently” is used herein to indicate that the variable, which is independently applied, varies independently from application to application. Thus, in a compound such as R"XYR", wherein R" is “independently carbon or nitrogen,” both R" can be carbon, both R" can be nitrogen, or one R" can be carbon and the other R” nitrogen.
  • pharmaceutically acceptable salt or prodrug is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, phosphate ester, salt of an ester or a related group) of a compound, which, upon administration to a patient, provides the compound.
  • Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium and magnesium, among numerous other acids well known in the pharmaceutical art.
  • Pharmaceutically acceptable prodrugs refer to a compound that is metabolized, for example hydrolyzed or oxidized, in the host to form the compound of the present invention.
  • prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound.
  • Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, dephosphorylated to produce the active compound.
  • esters as used herein, unless otherwise specified, includes those esters of one or more compounds, which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of hosts without • undue toxicity, irritation, allergic response and the like, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • subject refers to an animal, preferably a mammal, most preferably a human. Mammals can include non-human mammals, including, but not limited to, pigs, sheep, goats, cows (bovine), deer, mules, horses, monkeys and other non-human primates, dogs, cats, rats, mice, rabbits or any other known or disclosed herein.
  • non-human mammals including, but not limited to, pigs, sheep, goats, cows (bovine), deer, mules, horses, monkeys and other non-human primates, dogs, cats, rats, mice, rabbits or any other known or disclosed herein.
  • the present invention provides for the use of TCN, TCN-P and related compounds in combination with an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof for use in particular therapeutic regimens for the treatment of proliferative disorders.
  • an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof for use in particular therapeutic regimens for the treatment of proliferative disorders.
  • triciribine compounds and “triciribine and related compounds” refers to compounds having the following structures:
  • each R2', R3' and R5' are independently hydrogen, optionally substituted phosphate or phosphonate (including mono-, di-, or triphosphate or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); amide, sulfonate ester including alkyl or arylalkyl; sulfonyl, including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as for example as described in the definition of an aryl given herein; optionally substituted arylsulfonyl; a lipid, including a phospholipid; an amino
  • R x and R y are independently hydrogen, optionally substituted phosphate; acyl (including lower acyl); amide, alkyl (including lower alkyl); aromatic, polyoxyalkylene such as polyethyleneglycol, optionally substituted arylsulfonyl; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; or cholesterol; or other pharmaceutically acceptable leaving group.
  • the compound is administered as a 5'-phosphoether lipid or a 5 '-ether lipid.
  • Ri and R 2 each are independently H, optionally substituted straight chained, branched or cyclic alkyl (including lower alkyl), alkenyl, or alkynyl, CO-alkyl, CO- alkenyl, CO-alkynyl, CO-aryl or heteroaryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, sulfonyl, alkylsulfonyl, arylsulfonyl, aralkylsulfonyl.
  • R2' and R3' are hydrogen.
  • R2' and R5' are hydrogen.
  • R2', R3' and R5' are hydrogen. In yet another embodiment, R2', R3' , R5', Rl and R2 are hydrogen. [0081] In another embodiment, the triciribine compound has the following structure:
  • R 3 is H, optionally substituted straight chained, branched or cyclic alkyl (including lower alkyl), alkenyl, or alkynyl, NH 2 , NHR 4 , N(R 4 ) 2 , aryl, alkoxyalkyl, aryloxyalkyl, or substituted aryl; and [0083] each R 4 independently is H, acyl including lower acyl, alkyl including lower alkyl such as but not limited to methyl, ethyl, propyl and cyclopropyl, alkenyl, alkynyl, cycloalkyl, alkoxy, alkoxyalkyl, hydroxyalkyl, or aryl.
  • R 3 is a straight chained Cl-I l alkyl, iso-propyl, t-butyl, or phenyl.
  • the triciribine compounds provided herein have the following structure:
  • the triciribine compounds provided herein have the following structure:
  • the triciribine compounds provided herein have the following structure:
  • each R 4 independently is H, acyl including lower acyl, alkyl including lower alkyl such as but not limited to methyl, ethyl, propyl and cyclopropyl, alkenyl, alkynyl, cycloalkyl, alkoxy, alkoxyalkyl, hydroxyalkyl, or aryl.
  • R 6 is ethyl, CH 2 CH 2 OH, or CH 2 -phenyl.
  • the triciribine compounds provided herein have the following structure:
  • the triciribine compounds provided herein have the following structure:
  • the triciribine compounds provided herein have the following structure:
  • the term "epidermal growth factor receptor inhibitor” refers to compounds that target the epidermal growth factor receptor (EGFR) tyrosine kinase, which is highly expressed and occasionally mutated in various forms of cancer.
  • the compounds bind in a reversible fashion to the adenosine triphosphate (ATP) binding site of the receptor.
  • ATP adenosine triphosphate
  • two members of the EGFR family come together to form a homodimer.
  • epidermal growth factor receptor inhibitor compounds include erlotinib-like compounds such as for example, gefitinib and erlotinib.
  • each Rl and R2 is independently hydrogen, independently optionally substituted alkoxy, optionally substituted amine, aromatic amine, heteroaromatic amine, optionally substituted straight chained, branched or cyclic alkyl;
  • each R3 and R4 is independently hydrogen, independently optionally substituted aromatic amine, heteroaromatic amine, or cyclic amine.
  • the erlotinib-like compounds include, but are limited to, the following
  • the compounds disclosed herein may contain chiral centers. Such chiral centers may be of either the (R) or (S) configuration, or may be a mixture thereof.
  • the compounds provided herein may be enantiomerically pure, or be stereoisomeric or diastereomeric mixtures.
  • the disclosure of a compound herein encompasses any racemic, optically active, polymorphic, or steroisomeric form, or mixtures therof, which preferably possesses the useful properties described herein, it being well known in the art how to prepare optically active forms and how to determine activity using the standard tests described herein, or using other similar tests which are will known in the art.
  • Examples of methods that can be used to obtain optical isomers of the compounds include the following: [00102] (i) physical separation of crystals- a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct;
  • enzymatic asymmetric synthesis a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
  • chemical asymmetric synthesis a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce assymetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries;
  • diastereomer separations a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers.
  • the resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;
  • (x) chiral liquid chromatography a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase.
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
  • triciribine, triciribine phosphate (TCN-P), triciribine 5'-phosphate (TCN-P), or the DMF adduct of triciribine (TCN-DMF) are provided.
  • TCN can be synthesized by any technique known to one skilled in the art, for exmple, as described in Tetrahedron Letters, 1971. 49: p. 4757-4760.
  • TCN-P can be prepared by any technique known to one skilled in the art, for example, as described in U.S. Pat. No. 4,123,524.
  • TCN-DMF The synthesis of TCN-DMF is described, for example, in INSERM, 1978. 81 : p. 37-82.
  • Other compounds related to TCN as described herein can be synthesized, for example, according to the methods disclosed in Gudmundsson K.S. et al., Nucleosides Nucleotides Nucleic Acids, 2001. 20(10-11): p. 1823-1830; Porcari A.R. et al., J Med Chem, 2000. 43(12): p. 2457-2463; Porcari A.R. et al., Nucleosides Nucleotides, 1999. 18(11-12): p. 2475-2497; Porcari A.R.
  • compositions include those derived from pharmaceutically acceptable inorganic or organic bases and acids. Suitable salts include those derived from alkali metals such as potassium and sodium,
  • alkaline earth metals such as calcium and magnesium
  • examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ot-ketoglutarate, and a- glycerophosphate.
  • Suitable inorganic salts may also be formed, including, sulfate, . nitrate, bicarbonate, and carbonate salts.
  • nucleotide prodrug Any of the nucleotides described herein can be administered as a nucleotide prodrug to increase the activity, bioavailability, stability or otherwise alter the properties of the nucleoside. A number of nucleotide prodrug ligands are known.
  • alkylation, acylation or other lipophilic modification of the mono, di or triphosphate of the nucleoside will increase the stability of the nucleotide.
  • substituent groups that can replace one or more hydrogens on the phosphate moiety are alkyl, aryl, steroids, carbohydrates, including sugars, 1,2-diacylglycerol and alcohols. Many are described in R. Jones and N. Bischofberger, Antiviral Research, 1995. 27: p. 1-17. Any of these can be used in combination with the disclosed nucleosides to achieve a desired effect.
  • the triciribine or a related compound is provided as
  • suitable lipophilic substituents that can be covalently incorporated into the nucleoside, preferably at the 5'-OH position of the nucleoside or lipophilic preparations, include U.S. Patent Nos. 5,149,794, 5,194,654, 5,223,263, 5,256,641, 5,411,947, 5,463,092, 5,543,389, 5,543,390, 5,543,391, and 5,554,728, all of which are incorporated herein by reference.
  • derivatives of triciribine or related compounds are those that contain substituents as described in the following publications. These derivatized triciribine or related compounds can be used for the indications described in the text or otherwise as antiviral agents, including as anti-HTV or anti-HBV agents.
  • Alkyl hydrogen phophonate derivatives of the anti-HIV agent AZT may be less toxic than the parent nucleoside analogue.
  • One preferred phosphate prodrug group is the S-acyl-2-thioethyl group, also referred to as "SATE.”
  • prodrugs that can be used are those described in the following patents and patent applications: U.S. Patent Nos. 5,614,548, 5,512,671, 5,770,584, 5,962,437, 5,223,263, 5,817,638, 6,252,060, 6,448,392, 5,411,947, 5,744,592, 5,484,809, 5,827,831, 5,696,277, 6,022,029, 5,780,617, 5,194,654, • 5,463,092, 5,744,461, 4,444,766, 4,562,179, 4,599,205, 4,493,832, 4,221,732,
  • dosing regimens are provided that limit the toxic side effects of TCN and related compounds.
  • such dosing regimens minimize the following toxic side effects, including, but not limited to, hepatoxicity, thrombocytopenia, hyperglycemia, vomiting, hypocalcemia, anemia, hypoalbunemia, myelosuppression, hypertriglyceridemia, hyperamylasemia,
  • the administration of TCN, TCN-P or related compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof provides at least a partial or complete response in vivo in at least 15-20% of the subjects.
  • a partial reponse can be at least 15, 20, 25, 30, 35, 40, 50, 55, 60, 65, 70, 75, 80 or 85% regression of the tumor.
  • this response can be evident in at least 15, 15, 20, 25, 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85 or 90% of the subjects treated with the therapy.
  • such response rates can be obtained by any therapeutic regimen disclosed herein.
  • methods are provided to treat a subject that has been diagnosed with cancer by administering to the subject an effective amount of TCN, TCN-P or a related compound and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, according to a dosing schedule that includes administering the the triciribine compound and/ an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, one time per week for three weeks followed by a one week period wherein the drug is not administered (i.e., via a 28 day cycle).
  • such 28 day cycles can be repeated at least 2, 3, 4, or 5 times or until regression of the tumor is evident.
  • a 42 day cycle in which the compounds disclosed herein can be administered once a week for four weeks followed by a two week period in which the the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, is not administered.
  • such 42 day cycles can be repeated at least 2, 3, 4, or 5 times or until regression of the tumor is evident. In a particular embodiment, less than 12, less
  • 1 -W A/2866539.1 than 11 or less than 10 mg/m 2 of TCN, TCN-P or a related compound can be administered according to a 42 day cycle.
  • 2, 3, 4, 5, 6, 7, 8, 9, 10 or 15 mg/m 2 of TCN, TCN-P or a related compound can be administered according to a 42 day cycle.
  • about 1 to about 50 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof is administered.
  • 1, 5, 10, 15, 20, 25, 30, 35, or 40 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be administered according to a 42 day cycle.
  • methods are provided to treat cancer in a subject by administering to the subject a dosing regimen of 10 mg/m 2 or less of TCN, TCN-P or a related compound and less than about 30 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, one time per week.
  • a dosing regimen 10 mg/m 2 or less of TCN, TCN-P or a related compound and less than about 30 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, one time per week.
  • 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 mg/m 2 of TCN, TCN-P or a related compound as dislosed herein can be administered one time per week
  • 1, 5, 10, 15, 20, 25, 30, 35, or 40 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be administered one time per week.
  • the compounds disclosed herein can be administered simultaneously as a single bolus dose over a short period of time, for example, about 5, 10, 15, 20, 30 or 60 minutes.
  • dosing schedules are provided in which the compounds are administered simultaneously via continuous infusion for at least 24, 48, 72, 96, or 120 hours.
  • the administration of the the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, via continuous or bolus injections can be administered simultaneously as a single bolus dose over a short period of time, for example, about 5, 10, 15, 20, 30 or 60 minutes.
  • dosing schedules are provided in which the compounds are administered simultaneously via continuous infusion for at least 24, 48, 72, 96, or 120 hours.
  • the administration of the the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, via continuous or bolus injections can
  • 1 -W A/2866539.1 be repeated at a certain frequency at least: once a week, once every two weeks, once every three weeks, once a month, once every five weeks, once every six weeks, once every eight weeks, once every ten weeks and/or once every twelve weeks.
  • the type and frequency of administrations can be combined in any manner disclosed herein to create a dosing cycle.
  • the the triciribine compound and/or an erlotinib-like compound, for example, gef ⁇ tinib, erlotinib or a salt thereof, can be repeatedly administered via a certain dosing cycles, for example as a bolus injection once every two weeks for three months.
  • the dosing cycles can be administered for at least: one, two three, four five, six, seven, eight, nine, ten, eleven, twelve, eighteen or twenty four months. Alternatively, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15 or 20 dosing cycles can be administered to a patient.
  • the triciribine compound and/or an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof, can be administered according to any combination disclosed herein, for example, the the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, can be administered once a week every three weeks for 3 cycles.
  • the compounds can be administered separately at least once a day for at least 2, 3, 4, 5, 6, 7, 8, 9 or 10 days. Such administration can be followed by corresponding periods in which the the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, are not administered
  • TCN, TCN-P and related compounds and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof, as disclosed herein can be administered to patients in an amount that is effective in causing tumor regression.
  • the administration of TCN, TCN-P or related compounds and an erlotinib-like compound can be administered to patients in an amount that is effective in causing tumor regression.
  • gefitinib, erlotinib or a salt thereof can provide at least a partial, such as at least 15, 20 or 30%, or complete response in vivo in at least 15-20% of the subjects.
  • at least 2, 5, 10, 15, 20, 30 or 50 mg/m 2 of a triciribine compound disclosed herein can be administered to a subject.
  • At least about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 12, 15, 17, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 165, 175, 200, 250, 300, or 350 mg/m 2 of TCN, TCN-P or a related compound disclosed herein can be administered to a subject.
  • 1, 5, 10, 15, 20, 25, 30, 35, or 40 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be administered to a subject.
  • the administration of the compound can be conducted according to any of the therapeutic regimens disclosed herein.
  • the dosing regimen includes administering less than about 20 mg/m 2 of TCN and related compounds and less than about 30 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, either concurrently, sequentially, or conducted over a period of time.
  • less than 20 mg/m 2 of TCN or related compounds can be administered once a week concurrently with less than about 30 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof.
  • less than 20 mg/m 2 of TCN or related compounds can be administered once a week and less than about 30 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be administered the following week.
  • 2 mg/m 2 , 5 mg/m 2 , 10 mg/m 2 , and/or 15 mg/m 2 of TCN or a related compound and less than about 300, 250, 200, 150, or 100 mg an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be
  • I -W A/2866539.1 administered to a subject.
  • less than 10 mg/m 2 of a triciribine compound and less than about 300 mg of an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be administered to a subject via continuous infusion for at least five days.
  • the present invention provides for any combination of dosing type, frequency, number of cycles and dosage amount disclosed herein.
  • methods are provided to identify cancers or tumors that are susceptible to the toxic effects of triciribine (TCN) and related compounds.
  • methods are provided to treat a cancer or tumor in a mammal by (i) obtaining a biological sample from the tumor; (ii) determining whether the cancer or tumor overexpresses Akt kinase or hyperactivated and phosphorylated Akt kinase, and (iii) treating the cancer or tumor with triciribine or a related compound as described herein.
  • the biological sample can be a biopsy.
  • the biological sample can be fluid, cells and/or aspirates obtained from the tumor or cancer.
  • the biological sample can be obtained according to any technique known to one skilled in the art.
  • a biopsy can be conducted to obtain the biological sample.
  • a biopsy is a procedure performed to remove tissue or cells from the body for examination.
  • Some biopsies can be performed in a physician's office, while others need to be done in a hospital setting.
  • some biopsies require use of an anesthetic to numb the area, while others do not require any sedation.
  • an endoscopic biopsy can be performed. This type of biopsy is performed through a fiberoptic endoscope (a long, thin tube that has a close-focusing telescope on the end for viewing) through a natural body orifice (i.e., rectum) or a small
  • the endoscope is used to view the organ in question for abnormal or suspicious areas, in order to obtain a small amount of tissue for study. Endoscopic procedures are named for the organ or body area to be visualized and/or treated.
  • the physician can insert the endoscope into the gastrointestinal tract (alimentary tract endoscopy), bladder (cystoscopy), abdominal cavity (laparoscopy), joint cavity (arthroscopy), mid-portion of the chest (mediastinoscopy), or trachea and bronchial system (laryngoscopy and bronchoscopy).
  • a bone marrow biopsy can be performed. This type of biopsy can be performed either from the sternum (breastbone) or the iliac crest hipbone (the bone area on either side of the pelvis on the lower back area). The skin is cleansed and a local anesthetic is given to numb the area. A long, rigid needle is inserted into the marrow, and cells are aspirated for study; this step is occasionally uncomfortable. A core biopsy (removing a small bone 'chip' from the marrow) may follow the aspiration. [00137] In a further embodiment, an excisional or incisional biopsy can be performed on the mammal. This type of biopsy is often used when a wider or deeper portion of the skin is needed.
  • a scalpel surgical knife
  • a full thickness of skin is removed for further examination, and the wound is sutured (sewed shut with surgical thread).
  • an excisional biopsy technique When the entire tumor is removed, it is referred to as an excisional biopsy technique. If only a portion of the tumor is removed, it is referred to as an incisional biopsy technique.
  • Excisional biopsy is often the method usually preferred, for example, when melanoma (a type of skin cancer) is suspected.
  • a fine needle aspiration (FNA) biopsy can be used. This type of biopsy involves using a thin needle to remove very small pieces
  • FNA is not, for example, used for diagnosis of a suspicious mole, but may be used, for example, to biopsy large lymph nodes near a melanoma to see if the melanoma has metastasized (spread).
  • a computed tomography scan can be used to guide a needle into a tumor in an internal organ such as the lung or liver.
  • punch shave and/ or skin biopsies can be conducted.
  • Punch biopsies involve taking a deeper sample of skin with a biopsy instrument that removes a short cylinder, or "apple core," of tissue. After a local anesthetic is administered, the instrument is rotated on the surface of the skin until it cuts through all the layers, including the dermis, epidermis, and the most superficial parts of the subcutis (fat).
  • a shave biopsy involves removing the top layers of skin by shaving it off.
  • Shave biopsies are also performed with a local anesthetic.
  • Skin biopsies involve removing a sample of skin for examination under the microscope to determine if, for example, melanoma is present.
  • Akt kinase overexpression can refer to the phosphorylation state of the kinase. Hyperphosphorylation of Akt can be detected according to the methods described herein.
  • a tumor biopsy can be compared to a control tissue.
  • the control tissue can be a normal tissue from the mammal in which the biopsy was obtained or a normal tissue from a healthy mammal.
  • Akt kinase overexpression or hyperphosphorylation can be determined if the tumor biopsy contains greater amounts of Akt kinase and/ or Akt kinase phosphorylation than the control tissue, such as, for example, at least approximately 1.5, 2, 2.25, 2.5, 2.75, 3,
  • the present invention provides a method to detect aberrant Akt kinase expression in a subject or in a biological sample from the subject by contacting cells, cell extracts, serum or other sample from the subjects or said biological sample with an immunointeractive molecule specific for an Akt kinase or antigenic portion thereof and screening for the level of immunointeractive molecule-Akt kinase complex formation, wherein an elevated presence of the complex relative to a normal cell is indicative of an aberrant cell that expresses or overexpresses Akt.
  • cells or cell extracts can be screened immunologically for the presence of elevated levels of Akt kinase.
  • the aberrant expression of Akt in a cell is detected at the genetic level by screening for the level of expression of a gene encoding an Akt kinase wherein an elevated level of a transcriptional expression product (i.e., mRNA) compared to a normal cell is indicative of an aberrant cell.
  • mRNA a transcriptional expression product
  • real-time PCR as well as other PCR procedures can be used to determine transcriptional activity.
  • mRNA can be obtained from cells of a subject or from a biological sample from a subject and cDNA optionally generated.
  • the mRNA or cDNA can then be contacted with a genetic probe capable of hybridizing to and/or amplifying all or part of a nucleotide sequence encoding Akt kinase or its complementary nucleotide sequence and then the level of the mRNA or cDNA can be detected wherein the presence of elevated levels of the mRNA or cDNA compared to normal controls can be assessed.
  • a genetic probe capable of hybridizing to and/or amplifying all or part of a nucleotide sequence encoding Akt kinase or its complementary nucleotide sequence
  • an antibody, monoclonal or polyclonal, to Akt kinase in a quantitative or semiquantitative diagnostic kit to determine relative levels of Akt kinase in suspected cancer cells from a patient which can include all the reagents necessary to perform the assay.
  • a kit utilizing reagents and materials necessary to perform an ELISA assay is provided.
  • Reagents can include, for example, washing buffer, antibody dilution buffer, blocking buffer, cell staining solution, developing solution, stop solution, anti-phospho-protein specific antibodies, anti-Pan protein specific antibodies, secondary antibodies, and distilled water.
  • the kit can also include instructions for use and can optionally be automated or semi-automated or in a form which is compatible with automated machine or software.
  • a phosphor-ser-473 Akt antibody that detects the activated form of AKT (Akt phosphorylated at serine 474) can be utilized as the antibody in a diagnostic kit. See, for example, Yuan et al., Oncogene, 2000. 19: p. 2324-2330.
  • Akt also named PKB 3 , represents a subfamily of the serine/threonine kinase. Three members, AKTl , AKT2, and AKT3, have been identified in this subfamily. Akt is activated by extracellular stimuli in a PI3K-dependent manner (Datta S.R. et al., Genes Dev, 1999. 13: p. 2905-2927). Full activation of Akt requires phosphorylation of Thr 308 in the activation loop and Ser 473 in the C-terminal activation domain. Akt is negatively regulated by PTEN tumor suppressor.
  • Akt kinase can be any known Akt family kinase, or kinase related thereto, including, but not limited to Akt 1 , Akt 2, Akt 3.
  • Akt 1 , Akt 2, Akt 3 The mRNA and amino acid sequences of human Aktl, Akt2, and Akt 3 are illustrated in Figures 6a-c, 7a-d, and 8a-c, respectively.
  • a method for detecting the aberrant expression of an Akt kinase in a cell in a mammal or in a biological sample from the mammal, by contacting cells, cell extracts or serum or other sample from the mammal or biological sample with an immunointeractive molecule specific for an Akt kinase or
  • the immunointeractive molecule can be a molecule having specificity and binding affinity for an Akt kinase or its antigenic parts or its homologs or derivatives thereof.
  • the immunointeractive molecule can be an immunglobulin molecule.
  • the immunointeractive molecules can be an antibody fragments, single chain antibodies, and/or deimmunized molecules including humanized antibodies and T-cell associated antigen-binding molecules (TABMs).
  • the antibody can be a monoclonal antibody. In another particular embodiment, the antibody can be a polyclonal antibody.
  • the immunointeractive molecule can exhibit specificity for an Akt kinase or more particularly an antigenic determinant or epitope on an Akt kinase.
  • An antigenic determinant or epitope on an Akt kinase includes that part of the molecule to which an immune response is directed.
  • the antigenic determinant or epitope can be a B-cell epitope or where appropriate a T-cell epitope.
  • the antibody is a phosphor-ser 473 Akt antibody.
  • One embodiment of the present invention provides a method for diagnosing the presence of cancer or cancer-like growth in a mammal, in which aberrant Akt activity is present, by contacting cells or cell extracts from the mammal or a biological sample from the subject with an Akt kinase-binding effective amount of an antibody having specificity for the Akt kinase or an antigenic determinant or epitope thereon and then quantitatively or qualitatively determining the level of an Akt kinase- antibody complex wherein the presence of elevated levels of said complex compared to
  • Antibodies can be prepared by any of a number of means known to one skilled in the art.
  • antibodies can be generally but not necessarily derived from non-human animals such as primates, livestock animals (e.g. sheep, cows, pigs, goats, horses), laboratory test animals (e.g. mice, rats, guinea pigs, rabbits) and/or companion animals (e.g. dogs, cats).
  • Antibodies may also be recombinantly produced in prokaryotic or eukaryotic host cells.
  • antibody based assays can be conducted in vitro on cell or tissue biopsies.
  • an antibody is suitably deimmunized or, in the case of human use, humanized, then the antibody can be labeled with, for example, a nuclear tag, administered to a patient and the site of nuclear label accumulation determined by radiological techniques.
  • the Akt kinase antibody can be a cancer targeting agent.
  • another embodiment of the present invention provides deimmunized forms of the antibodies for use in cancer imaging in human and non-human patients.
  • the enzyme is required to be extracted from a biological sample whether this be from animal including human tissue or from cell culture if produced by recombinant means.
  • the Akt kinase can be separated from the biological sample by any suitable means.
  • the separation may take advantage of any one or more of the Akt kinase's surface charge properties, size, density, biological activity and its affinity for another entity (e.g. another protein or chemical compound to which it binds or otherwise associates).
  • separation of the Akt kinase from the biological fluid can be achieved by any one or more of ultra-centrifugation, ion-exchange chromatography (e.g. anion exchange chromatography, cation exchange chromatography), electrophoresis (e.g.
  • polyacrylamide gel electrophoresis isoelectric focussing
  • size separation e.g., gel filtration, ultra-filtration
  • affinity-mediated separation e.g. immunoaffinity separation including, but not limited to, magnetic bead separation such as Dynabead (trademark) separation, immunochromatography, immuno-precipitation.
  • the separation of Akt kinase from the biological fluid can preserve conformational epitopes present on the kinase and, thus, suitably avoids techniques that cause denaturation of the enzyme.
  • the kinase can be separated from the biological fluid using any one or more of affinity separation, gel filtration and/or ultra-filtration.
  • Immunization and subsequent production of monoclonal antibodies can be carried out using standard protocols known in the art, such as, for example, described by Kohler and Milstein (Kohler and Milstein, Nature, 1975. 256: p. 495-499; Kohler and Milstein, Eur J Immunol, 1976. 6(7): p. 511-519; Coligan et al., (Current Protocols in Immunology, 1991-1997, John Wiley & Sons, Inc.) or Toyama et al. (Monoclonal Antibody, Experiment Manual, 1987, Kodansha Scientific).
  • an animal is immunized with an Akt kinase-containing biological fluid or fraction thereof or a recombinant form of Akt kinase by standard methods to produce antibody-producing cells, particularly antibody-producing somatic cells (e.g. B lymphocytes). These cells can then be removed from the immunized animal for immortalization.
  • a fragment of an Akt kinase can be used to the generate antibodies.
  • the fragment can be associated with a carrier.
  • the carrier can be any substance of typically high molecular weight to which a non- or poorly immunogenic substance (e.g. a hapten) is naturally or artificially linked to enhance its immunogenicity.
  • Immortalization of antibody-producing cells can be carried out using methods which are well-known in the art.
  • the immortalization may be
  • antibody-producing cells are immortalized using the cell fusion method (described in Coligan et al., 1991-1997, supra), which is widely employed for the production of monoclonal antibodies.
  • somatic antibody-producing cells with the potential to produce antibodies, particularly B cells, are fused with a myeloma cell line.
  • somatic cells may be derived from the lymph nodes, spleens and peripheral blood of primed animals, preferably rodent animals such as mice and rats.
  • mice spleen cells can be used.
  • rat, rabbit, sheep or goat cells can also be used.
  • Specialized myeloma cell lines have been developed from lymphocytic tumours for use in hybridoma-producing fusion procedures (Kohler and Milstein, 1976, supra; Shulman et al., Nature, 1978. 276: p. 269-270; VoIk et al., J Virol, 1982. 42(1): p. 220-227).
  • Many myeloma cell lines can also be used for the production of fused cell hybrids, including, e.g.
  • P3.times.63-Ag8 P3.times.63-AG8.653, P3/NS1- Ag4-1 (NS-I), Sp2/0-Agl4 and Sl 94/5.XXO-Bu.1.
  • the P3.times.63-Ag8 and NS-I cell lines have been described by Kohler and Milstein (1976, supra).
  • Shulman et al. (1978, supra) developed the Sp2/0-Agl4 myeloma line.
  • the S194/5.XXO.Bu.l line was reported by Trowbridge (J Exp Med, 1978. 148(1): p. 313-323).
  • Methods for generating hybrids of antibody-producing spleen or lymph node cells and myeloma cells usually involve mixing somatic cells with myeloma cells in a 10: 1 proportion (although the proportion may vary from about 20: 1 to about 1:1), respectively, in the presence of an agent or agents (chemical, viral or electrical) that promotes the fusion of cell membranes. Fusion methods have been described (Kohler and Milstein, 1975, supra; Kohler and Milstein, 1976, supra; Gefter et al., Somatic Cell Genet, 1977. 3: p. 231-
  • fusion-promoting agents used by those investigators were Sendai virus and polyethylene glycol (PEG).
  • means to select the fused cell hybrids from the remaining unfused cells, particularly the unfused myeloma cells are provided.
  • the selection of fused cell hybrids can be accomplished by culturing the cells in media that support the growth of hybridomas but prevent the growth of the unfused myeloma cells, which normally would go on dividing indefinitely.
  • the somatic cells used in the fusion do not maintain long-term viability in in vitro culture and hence do not pose a problem.
  • each cell line may be propagated in either of two standard ways.
  • a suspension of the hybridoma cells can be injected into a histocompatible animal. The injected animal will then develop tumours that secrete the specific monoclonal antibody produced by the fused cell hybrid.
  • the body fluids of the animal such as serum or ascites fluid, can be tapped to provide monoclonal antibodies in high concentration.
  • the individual cell lines may be propagated in vitro in laboratory culture vessels.
  • the culture medium containing high concentrations of a single specific monoclonal antibody can be harvested by decantation, filtration or centrifugation, and subsequently purified.
  • the cell lines can then be tested for their specificity to detect the Akt kinase of interest by any suitable immunodetection means.
  • cell lines can be aliquoted into a number of wells and incubated and the supernatant from each well is analyzed by enzyme-linked immunosorbent assay (ELISA), indirect fluorescent antibody technique, or the like.
  • ELISA enzyme-linked immunosorbent assay
  • the cell line(s) producing a monoclonal antibody capable of recognizing the target LIM kinase but which does not recognize non-target epitopes are identified and then directly cultured in vitro or injected into a histocompatible animal to form tumours and to produce, collect and purify the required antibodies.
  • the present invention provides, therefore, a method of detecting in a sample an Akt kinase or fragment, variant or derivative thereof comprising contacting the sample with an antibody or fragment or derivative thereof and detecting the level of a complex containing the antibody and Akt kinase or fragment, variant or derivative thereof compared to normal controls wherein elevated levels of Akt kinase is determined.
  • Any suitable technique for determining formation of the complex may be used.
  • an antibody according to the invention, having a reporter molecule associated therewith, may be utilized in immunoassays.
  • Immunoassays include but are not limited to radioimmunoassays (RIAs), enzyme-linked immunosorbent assays (ELISAs) immunochromatographic techniques (ICTs), and Western blotting which are well known to those of skill in the art. Immunoassays can also include competitive RIAs, enzyme-linked immunosorbent assays (ELISAs) immunochromatographic techniques (ICTs), and Western blotting which are well known to those of skill in the art. Immunoassays can also include competitive methods (RIAs), enzyme-linked immunosorbent assays (ELISAs) immunochromatographic techniques (ICTs), and Western blotting which are well known to those of skill in the art. Immunoassays can also include competitive methods (RIAs), enzyme-linked immunosorbent assays (ELISAs) immunochromatographic techniques (ICTs), and Western blotting which are well known to those of skill in the art. Immunoassays can also include competitive blotting and Western blotting which are well known to those of
  • 1 -W A/2866539.1 assays The present invention encompasses qualitative and quantitative immunoassays. [00158] Suitable immunoassay techniques are described, for example, in U.S. Pat.
  • the invention further provides methods for quantifying Akt protein expression and activation levels in cells or tissue samples obtained from an animal, such as a human cancer patient or an individual suspected of having cancer.
  • the invention provides methods for quantifying Akt protein expression or activation levels using an imaging system quantitatively.
  • the imaging system can be used to receive, enhance, and process images of cells or tissue samples, that have been stained with AKT protein-specific stains, in order to determine the amount or activation level of AKT protein expressed in the cells or tissue samples from such an animal.
  • a calibration curve of AKTl and AKT2 protein expression can be generated for at least two cell lines expressing differing amounts of AKT protein.
  • AKT protein expression in a cell or tissue sample can be quantified using an enzyme-linked immunoabsorbent assay (ELISA) to determine the amount of AKT protein in a sample.
  • ELISA enzyme-linked immunoabsorbent assay
  • Akt kinase In such assays, an enzyme is conjugated to the second antibody, generally by means of glutaraldehyde or periodate.
  • the substrates to be used with the specific enzymes are generally chosen for the production of, upon hydrolysis by the corresponding enzyme, a detectable colour change. It is also possible to employ fluorogenic substrates, which yield a fluorescent product rather than the chromogenic substrates.
  • the enzyme-labeled antibody can be added to the first antibody-antigen complex, allowed to bind, and then the excess reagent washed away. A solution containing the appropriate substrate can then be added to the complex of antibody- antigen-antibody.
  • the substrate can react with the enzyme linked to the second antibody, giving a qualitative visual signal, which may be further quantitated, usually spectrophotometrically, to give an indication of the amount of antigen which was present in the sample.
  • fluorescent compounds such as fluorescein, rhodamine and the lanthanide, europium (EU)
  • EU europium
  • the fluorochrome-labeled antibody When activated by illumination with light of a particular wavelength, the fluorochrome-labeled antibody adsorbs the light energy, inducing a state to excitability in the molecule, followed by emission of the light at a characteristic colour visually detectable with a light microscope.
  • the fluorescent- labeled antibody is allowed to bind to the first antibody-antigen complex.
  • IFMA Immunofluorometric assays
  • ELISA-mediated detection of Akt kinase especially in serum or other circulatory fluid This can be accomplished by immobilizing anti-Akt kinase antibodies to a solid support and contacting these with a biological extract such as serum, blood, lymph or other bodily fluid, cell extract or cell biopsy. Labeled anti-Akt kinase antibodies can then be used to detect immobilized Akt kinase.
  • This assay can be varied in any number of ways and all variations are encompassed by the present invention and known to one skilled in the art. This approach can enable rapid detection and quantitation of Akt kinase levels using, for example, a serum-based assay.
  • an Akt ELISA assay kit may be used in the present invention.
  • a Cellular Activation of Signaling ELISA kit for Akt S473 from SuperArray Bioscience can be utilized in the present invention.
  • the antibody can be an anti-pan antibody that recognizes Akt S473.
  • ELISA assay kit containing an anti-Akt antibody and additional reagents including, but not limited to, washing buffer, antibody dilution buffer, blocking buffer, cell staining solution, developing solution, stop solution, secondary antibodies, and distilled water.
  • a method to detect Akt kinases is provided by detecting the level of expression in a cell of a polynucleotide encoding an Akt kinase. Expression of the polynucleotide can be determined using any suitable technique known to one skilled in the art. In one embodiment, a labeled polynucleotide encoding an Akt kinase can be utilized as a probe in a Northern blot of an RNA extract obtained from the cell. In other embodiments, a nucleic acid extract from an animal can be utilized in
  • oligonucleotide primers corresponding to sense and antisense sequences of a polynucleotide encoding the kinase, or flanking sequences thereof, in a nucleic acid amplification reaction such as RT PCR.
  • a variety of automated solid-phase detection techniques are also available to one skilled in the art, for example, as described by Fodor et al. (Science, 1991. 251 : p. 767-777) and Kazal et al. (Nature Medicine, 1996. 2: p. 753-759).
  • RNA can be isolated from a cellular sample suspected of containing Akt kinase RNA, e.g. total RNA isolated from human cancer tissue.
  • RNA can be isolated by methods known in the art, e.g. using TRIZOL reagent (GIBCO- BRL/Life Technologies, Gaithersburg, Md.).
  • Oligo-dT, or random-sequence oligonucleotides, as well as sequence-specific oligonucleotides can be employed as a primer in a reverse transcriptase reaction to prepare first-strand cDNAs from the isolated RNA.
  • Resultant first-strand cDNAs can then amplified with sequence-specific oligonucleotides in PCR reactions to yield an amplified product.
  • PCR Polymerase chain reaction
  • RNA and/or DNA are amplified as described, for example, in U.S. Pat. No. 4,683,195.
  • sequence information from the ends of the region of interest or beyond is employed to design oligonucleotide primers. These primers will be identical or similar in sequence to opposite strands of the template to be amplified.
  • PCR can be used to amplify specific RNA sequences and cDNA transcribed from total cellular RNA. See generally Mullis et al. (Quant Biol, 1987. 51: p. 263; Erlich, eds., PCR Technology, 1989, Stockton Press, NY).
  • amplification of specific nucleic acid sequences by PCR relies upon
  • oligonucleotides or "primers” having conserved nucleotide sequences wherein the conserved sequences are deduced from alignments of related gene or protein sequences, e.g. a sequence comparison of mammalian Akt kinase genes.
  • one primer is prepared which is predicted to anneal to the antisense strand and another primer prepared which is predicted to anneal to the sense strand of a cDNA molecule which encodes a Akt kinase.
  • the reaction mixture is typically subjected to agarose gel electrophoresis or other convenient separation technique and the relative presence of the Akt kinase specific amplified DNA detected.
  • Akt kinase amplified DNA may be detected using Southern hybridization with a specific oligonucleotide probe or comparing its electrophoretic mobility with DNA standards of known molecular weight. Isolation, purification and characterization of the amplified Akt kinase DNA can be accomplished by excising or eluting the fragment from the gel (for example, see references Lawn et al., Nucleic Acids Res, 1981. 2: p. 6103; Goeddel et al., Nucleic Acids Res, 1980. 8: p.
  • telomere sequence can then be determined.
  • real-time PCR can be used to determine transcriptional levels of Akt nucleotides. Determination of transcriptional activity also includes a measure of potential translational activity based on available mRNA transcripts.
  • Real-time PCR as well as other PCR procedures use a number of chemistries for detection of PCR product including the binding of DNA binding fluorophores, the 5' endonuclease, adjacent liner and hairpin oligoprobes and the self- fluorescing amplicons. These chemistries and real-time PCR in general are discussed,
  • the aberrant expression of Akt can be identified by contacting a nucleotide sequences isolated from a biological sample with an oligonucleotide probe having a sequence complementary to an Akt sequences selected from the nucleotide sequences of figures 6a-c, 7a-d, or 8a-c, or fragment thereof, and then detecting the sequence by hybridizing the probe to the sequence, and comparing the results to a normal sample.
  • the hybridization of the probe to the biological sample can be detected by labeling the probe using any detectable agent.
  • the probe can be labeled for example, with a radioisotope, or with biotin, fluorescent dye, electron-dense reagent, enzyme, hapten or protein for which antibodies are available.
  • the detectable label can be assayed by any desired means, including spectroscopic, photochemical, biochemical, immunochemical, radioisotopic, or chemical means.
  • the probe can also be detected using techniques such as an oligomer restriction technique, a dot blot assay, a reverse dot blot assay, a line probe assay, and a 5' nuclease assay.
  • the probe can be detected using any of the generally applicable DNA array technologies, including macroarray, microarray and DNA microchip technologies.
  • the oligonucleotide probe typically includes approximately at least 14, 15, 16, 18, 20, 25 or 28 nucleotides that hybridize to the nucleotides selected from figures 6a-c, 7a-d, and 8a-c, or a fragment thereof. It is generally not preferred to use a probe that is greater than approximately 25 or 28 nucleotides in length.
  • the oligonucleotide probe is designed to identify an Akt nucleotide sequence. [00170] Kinase Assays
  • the activity of the Akt kinases can be measured using any suitable kinase assay known in the art.
  • any suitable kinase assay known in the art.
  • the methods described in Hogg et al (Oncogene, 1994. 9: p. 98-96), Mills et al (J Biol Chem, 1992. 267: p. 16000-006) and Tomizawa et al. (FEBS Lett, 2001. 492: p. 221-7), Schmandt et al. (J Immunol, 1994. 152: p. 96-105) can be used.
  • serine, threonine and tyrosine kinase assays are described in Ausubel et al. (Short Protocols in Molecular Biology, 1999, unit 17.6).
  • Akt kinase assays can generally use an Akt polypeptide, a labeled donor substrate, and a receptor substrate that is either specific or non-specific for Akt.
  • Akt transfers a labeled moiety from the donor substrate to the receptor substrate, and kinase activity is measured by the amount of labeled moiety transferred from the donor substrate to the receptor substrate.
  • Akt polypeptide can be produced using various expression systems, can be purified from cells, can be in the form of a cleaved or uncleaved recombinant fusion protein and/or can have non-Akt polypeptide sequences, for example a His tag or .beta.-galactosidase at its N- or C-terminus.
  • Akt activity can be assayed in cancerous cells lines if the cancerous cell lines are used as a source of the Akt to be assayed.
  • Suitable donor substrates for Akt assays include any molecule that is susceptible to dephosphorylation by Akt., such as, for example include .gamma.-labeled ATP and ATP analogs, wherein the label is 33 P, 32 P, . 35 S or any other radioactive isotope or a suitable fluorescent marker.
  • Suitable recipient substrates for Akt assays include any polypeptide or other molecule that is susceptible to phosphorylation by Akt.
  • Recipient substrates can be derived from fragments of in vivo targets of Akt. Recipient substrates fragments can be 8 to 50 amino acids in length , usually 10 to 30 amino acids and particularly of about 10, 12, 15, 18, 20 and 25 amino
  • Recipient substrates for TTK can be capable of being purified from other components of the reaction once the reaction has been performed. This purification is usually done through a molecular interaction, where the recipient substrates is biotinylated and purified through its interaction with streptavidin, or a specific antibody is available that can specifically recognize the recipient substrates.
  • the reaction can be performed in a variety of conditions, such as on a solid support, in a gel, in solution or in living cells.
  • the choice of detection methods depends on type of label used for the donor molecule and may include, for example, measurement of incorporated radiation or fluorescence by autoradiography, scintillation, scanning or fluorography. 6.
  • the compounds and pharmaceutical compositions provided herein can be used in the treatment of a condition including tumors, cancer, and other disorders associated with abnormal cell proliferation.
  • the compounds of the present invention can be used to treat a carcinoma, sarcoma, lymphoma, leukemia, and/or myeloma.
  • the compounds disclosed herein can be used to treat solid tumors.
  • the compounds of the present invention invention can be used for the treatment of cancer, such as, but not limited to cancer of the following organs or tissues: breast, prostate, lung, bronchus, colon, urinary, bladder, non-Hodgkin lymphoma, melanoma, kidney, renal, pancreas, pharnx, thyroid, stomach, brain, multiple myeloma, esophagus, liver, intrahepatic bile duct, cervix, larynx, acute myeloid leukemia, chronic lymphatic leukemia, soft tissue, such as heart, Hodgkin lymphoma, testis, small
  • intestine chronic myeloid leukemia, acute lymphatic leukemia, anus, anal canal, anorectal, thyroid, vulva, gallbladder, pleura, eye, nose nasal cavity, middle ear, nasopharnx, ureter, peritoneum, omentum, mesentery, and gastrointestineal, high grade glioma, glioblastoma, colon, rectal, pancreatic, gastric cancers, hepatocellular carcinoma; head and neck cancers, carcinomas; renal cell carcinoma; adenocarcinoma; sarcomas; hemangioendothelioma; lymphomas; leukemias, mycosis fungoides.
  • the compounds of the present invention can be used to treat skin diseases including, but not limited to, the malignant diseases angiosarcoma, hemangioendothelioma, basal cell carcinoma, squamous cell carcinoma, malignant melanoma and Kaposi's sarcoma, and the non-malignant diseases or conditions such as psoriasis, lymphangiogenesis, hemangioma of childhood, Sturge-Weber syndrome, verruca vulgaris, neurofibromatosis, tuberous sclerosis, pyogenic granulomas, recessive dystrophic epidermolysis bullosa, venous ulcers, acne, rosacea, eczema, molluscum contagious, seborrheic keratosis, and actinic keratosis.
  • skin diseases including, but not limited to, the malignant diseases angiosarcoma, hemangioendothelioma, basal cell carcinoma, squa
  • compositions including the compounds of the invention can be used to treat these cancers and other cancers at any stage from the discovery of the cancer to advanced stages.
  • compositions including compounds of the invention can be used in the treatment of the primary cancer and metastases thereof.
  • the compounds described herein can be used for the treatment of cancer, including, but not limited to the cancers listed in Table 2 below.
  • Antiangiogenic small molecules include thalidomide, which acts in part by inhibiting NFkB, 2-methoxyestradiol, which influences microtubule activation and hypoxia inducing factor (HIFIa) activation, cyclo-oxygenase 2 (COX2) inhibitors, and low doses of conventional chemotherapeutic agents, including cyclophosphamide, taxanes, and vinca alkaloids (vincristine, vinblastine) (D'Amato R.J. et al., Proc. Natl. Acad. Sci. U. S. A., 1994.
  • thalidomide which acts in part by inhibiting NFkB
  • 2-methoxyestradiol which influences microtubule activation and hypoxia inducing factor (HIFIa) activation
  • COX2 cyclo-oxygenase 2
  • conventional chemotherapeutic agents including cyclophosphamide, taxanes, and vinca alkaloids (vincristine,
  • tyrosine kinase inhibitors indirectly decrease angiogenesis by decreasing production of VEGF and other proangiogenic factors by tumor and stromal cells.
  • these drugs include Herceptin, imatinib (Glivec), and Iressa (Bergers G et al., J Clin Invest, 2003. 111 : p. 1287-1295; Ciardiello F. et al., Clin Cancer Res, 2001. 7: p. 1459-1465; Plum S.M. et al., Clin Cancer Res, 2003. 9: p. 4619-4626).
  • angiogenesis inhibitors have moved from animal models to
  • Angiogenesis inhibitors represent a promising treatment for a variety of cancers.
  • Avastin a high affinity antibody against vascular endothelial growth factor (VEGF)
  • VEGF vascular endothelial growth factor
  • Angiogenesis-related diseases include, but are not limited to, inflammatory, autoimmune, and infectous diseases; angiogenesis-dependent cancer, including, for example, solid tumors, blood bom tumors such as leukemias, and tumor metastases; benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; rheumatoid arthritis; psoriasis; eczema; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis; Osier-Webber Syndrome; myocardial angiogenesis; plaque neovascularization; telangiectasia; hemophiliac joints; angiofibroma; and wound granulation.
  • angiogenesis-dependent cancer including, for
  • compositions of this invention can be used to treat diseases such as, but not limited to, intestinal adhesions, atherosclerosis, scleroderma, warts, and hypertrophic scars (i.e., keloids).
  • Compositions of this invention can also be used in the treatment of diseases that have angiogenesis as a pathologic consequence such as cat scratch disease (Rochele minalia quintosa), ulcers (Helobacter pylori), tuberculosis, and leprosy.
  • the invention provides compounds that can be used to treat drug resistant cancer, including the embodiments of cancers and the triciribine compound
  • Multidrug resistance occurs in human cancers and can be a significant obstacle to the success of chemotherapy. Multidrug resistance is a phenomenon whereby tumor cells in vitro that have been exposed to one cytotoxic agent develop cross-resistance to a range of structurally and functionally unrelated compounds. In addition, MDR can occur intrinsically in some cancers without previous exposure to chemotherapy agents.
  • the present invention provides methods for the treatment of a patient with a drug resistant cancer, for example, multidrug resistant cancer, by administration of TCN, TCN-P or a related compound and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof.
  • TCN, TCN-P and related compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof can be used to treat cancers that are resistant to taxol alone, rapamycin, tamoxifen, cisplatin, and/ or gefitinib (iressa).
  • TCN, TCN-P or a related compound and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof can be used for the treatment of drug resistent cancers of the colon, bone, kidney, adrenal, pancreas, liver and/or any other cancer known in the art or described herein.
  • the triciribine compounds and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof can be administered together with other cytotoxic agents.
  • the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof and compositions thereof, when used in the treatment of solid tumors can be administered the use of radiation.
  • the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof and compositions disclosed herein can be combined with at least one additional chemotherapeutic agent.
  • the additional agents can be administered in combination or alternation with the compounds disclosed herein.
  • the drugs can form part of the same composition, or be provided as a separate composition for administration at the same time or a different time.
  • the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof disclosed herein can be combined with antiangiogenic agents to enhance their effectiveness, or combined with other antiangiogenic agents and administered together with other cytotoxic agents.
  • the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof and compositions, when used in the treatment of solid tumors can be administered with the agents selected from, but not limited to IL- 12, retinoids, interferons, angiostatin, endostatin, thalidomide, thrombospondin-1, thrombospondin-2, captopryl, anti-neoplastic agents such as alpha interferon, COMP (cyclophosphamide, vincristine, methotrexate and prednisone), etoposide, mBACOD (methortrexate, bleomycin, doxorubicin, cyclophosphamide, vincristine and dexamethasone), PRO-MACE/MOPP (prednisone, methotrexate (w/leucovin rescue), doxorubicin, cyclophosphamide
  • drugs with antimitotic effects such as those which target cytoskeletal elements, including podophylotoxins or vinca alkaloids (vincristine, vinblastine); antimetabolite drugs (such as 5-fluorouracil, cytarabine, gemcitabine, purine analogues such as pentostatin, methotrexate); alkylating agents or nitrogen mustards (such as nitrosoureas, cyclophosphamide or ifosphamide); drugs which target DNA such as the antracycline drugs adriamycin, doxorubicin, pharmorubicin or epirubicin; drugs which target topoisomerases such as etoposide; hormones and hormone agonists or antagonists such as estrogens, antiestrogens (tamoxifen and related compounds) and androgens, flutamide, leuprorelin, goserelin, cyprotrone or o
  • interferons can be used in combinations with the compounds of the present invention.
  • Suitable intereferons include: interferon alpha-2a, interferon alpha-2b, pegylated interferon alpha, including interferon alpha-2a and interferon alpha 2b, interferon beta, interferon gamma, interferon tau, interferon omega, INFERGEN (interferon alphacon-1) by InterMune, OMNIFERON (natural interferon) by Viragen, ALBUFERON by Human Genome Sciences, REBIF (interferon beta- Ia) by Ares-Serono, Omega Interferon by BioMedicine, Oral Interferon Alpha by
  • TCN TCN-P or a related compound and an erlotinib-
  • 1 -W A/2866539.1 like compound for example, gefitinib, erlotinib or a salt thereof as disclosed herein can be used in combination or alternation with additional chemotherapeutic agents, such as those described herein or in Table 3, for the treatment of drug resistant cancer, for example multiple drug resistant cancer.
  • Drug resistent cancers can include cancers of the colon, bone, kidney, adrenal, pancreas, liver and/or any other cancer known in the art or described herein.
  • the additional chemotherapeutic agent can be a P-glycoprotein inhibitor.
  • the P-glycoprotein inhibitor can be selected from the following drugs: verapamil, cyclosporin (such as cyclosporin A), tamoxifen, calmodulin antagonists, dexverapamil, dexniguldipine, valspodar (PSC 833), biricodar (VX-710), tariquidar (XR9576), zosuquidar (LY335979), laniquidar (R101933), and/or ONT-093. 7.
  • cyclosporin such as cyclosporin A
  • tamoxifen such as cyclosporin A
  • calmodulin antagonists such as cyclosporin A
  • dexverapamil such as cyclosporin A
  • dexniguldipine valspodar
  • PSC 833 biricodar
  • tariquidar XR9576
  • zosuquidar LY335979
  • laniquidar R101933
  • compositions including triciribine compounds and an erlotinib-like compound can optionally be administered with a pharmaceutical carrier or excipient.
  • Pharmaceutical carriers suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • the triciribine compounds and in combination with an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof may be formulated as the sole pharmaceutically active ingredients in the composition or may be combined.
  • compositions including the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof may be suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal, or parenteral (including subcutaneous, intramuscular, subcutaneous, intravenous, intradermal, intraocular,
  • compositions are administered intravenously.
  • compositions may conveniently be presented in unit dosage form and may be prepared by conventional pharmaceutical techniques. Such techniques include the step of bringing into association one or more compositions of the present invention and one or more pharmaceutical carriers or excipients.
  • the triciribine compounds and and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof and compositions thereof can be formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers.
  • the triciribine compounds described above are formulated into pharmaceutical compositions using techniques and procedures well known in the art ⁇ see, e.g., Ansel, Introduction to Pharmaceutical Dosage Forms, Fourth Ed., 1985, p.
  • compositions effective concentrations of one or more compounds or pharmaceutically acceptable derivatives thereof may be mixed with one or more suitable pharmaceutical carriers.
  • the compounds of the invention may be derivatized as the corresponding salts, esters, enol ethers or esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, hydrates or prodrugs prior to formulation.
  • concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration, that treats, prevents, or ameliorates one or more of the symptoms of the target disease or disorder.
  • the concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration, that treats, prevents, or ameliorates one or more of the symptoms of the target disease or disorder.
  • compositions are formulated for single dosage administration.
  • the weight fraction of compound is dissolved, suspended, dispersed or otherwise mixed in a selected carrier at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
  • compositions suitable for oral administration may be presented as discrete units such as, but not limited to, tablets, caplets, pills or dragees capsules, or cachets, each containing a predetermined amount of one or more of the compositions; as a powder or granules; as a solution or a suspension in an aqueous liquid or a nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil emulsion or as a bolus, etc.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing a triciribine compound and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents, preservatives, flavoring agents, and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents, preservatives, flavoring agents, and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • compositions of the present invention suitable for topical administration in the mouth include for example, lozenges, having the ingredients in a flavored basis,
  • sucrose and acacia or tragacanth usually sucrose and acacia or tragacanth; pastilles, having one or more triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof of the present invention in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes, having one or more of the compositions of the present invention administered in a suitable liquid carrie
  • the tablets, pills, capsules, troches and the like can contain one or more of the following ingredients, or compounds of a similar nature: a binder; a lubricant; a diluent; a glidant; a disintegrating agent; a coloring agent; a sweetening agent; a flavoring agent; a wetting agent; an emetic coating; and a film coating.
  • binders include microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, molasses, polvinylpyrrolidine, povidone, crospovidones, sucrose and starch paste.
  • Lubricants include talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • Diluents include, for example, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • Glidants include, but are not limited to, colloidal silicon dioxide.
  • Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate.
  • Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors.
  • Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and
  • compositions suitable for topical administration to the skin may be presented as ointments, creams, gels, and pastes, having one or more of the compositions administered in a pharmaceutical acceptable carrier.
  • compositions for rectal administration may be presented as a suppository with a suitable base including, for example, cocoa butter or a salicylate.
  • compositions suitable for nasal administration when the carrier is a solid, include a coarse powder having a particle size, for example, in the range of 20 to 500 microns which is administered in the manner in which snuff is taken, (i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose).
  • the carrier is a liquid (for example, a nasal spray or as nasal drops)
  • one or more of the compositions can be admixed in an aqueous or oily solution, and inhaled or sprayed into the nasal passage.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing one or more of the compositions and appropriate carriers.
  • Compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented in unit-
  • 87 l-WA/2866539.1 dose or multi-dose containers for example, sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water for injections, immediately prior to use.
  • sterile liquid carrier for example, water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets of the kind previously described above.
  • compositions suitable for enteral or parenteral administration can be used to fabricate the compositions.
  • compositions including triciribine compounds and an erlotinib-like compound for example, gef ⁇ tinib, erlotinib or a salt thereof may be used in combination with one or more pharmaceutically acceptable carrier mediums and/or excipients.
  • pharmaceutically acceptable carrier medium includes any and all carriers, solvents, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants, adjuvants, vehicles, delivery systems, disintegrants, absorbents, preservatives, surfactants, colorants, flavorants, or sweeteners and the like, as suited to the particular dosage form desired.
  • compositions including triciribine compounds and an erlotinib-like compound may be combined with pharmaceutically acceptable excipients, and, optionally, sustained- release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • pharmaceutically acceptable excipient includes a non-toxic solid, semi-solid or liquid
  • the specific therapeutically effective dose level for any particular host will depend upon a variety of factors, including for example, the disorder being treated and the severity of the disorder; activity of the specific composition employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration; route of administration; rate of excretion of the specific compound employed; the duration of the treatment; the triciribine compound and/or the an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof used in combination or coincidential with the specific composition employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the composition at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • compositions including triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of the composition appropriate for the host to be treated. Each dosage should contain the quantity of composition calculated to produce the desired therapeutic affect either as such, or in association with the selected pharmaceutical carrier medium.
  • Preferred unit dosage formulations are those containing a daily dose or unit, daily sub-dose, or an appropriate fraction thereof, of the administered ingredient.
  • 89 l-WA/2866539.1 For example, approximately 1-5 mg per day of a compound disclosed herein can reduce the volume of a solid tumor in mice.
  • the dosage will depend on host factors such as weight, age, surface area, metabolism, tissue distribution, absorption rate and excretion rate. In one embodiment, approximately 0.5 to 7 grams per day of a triciribine compound disclosed herein may be administered to humans. Optionally, approximately 1 to 4 grams per day of the compound can be administered to humans. In certain embodiments 0.001-5 mg/day is administered to a human.
  • the therapeutically effective dose level will depend on many factors as noted above. In addition, it is well within the skill of the art to start doses of the composition at relatively low levels, and increase the dosage until the desired effect is achieved.
  • compositions including triciribine compounds and an erlotinib-like compound for example, gef ⁇ tinib, erlotinib or a salt thereof may be used with a sustained-release matrix, which can be made of materials, usually polymers, which are degradable by enzymatic or acid-based hydrolysis or by dissolution. Once inserted into the body, the matrix is acted upon by enzymes and body fluids.
  • a sustained-release matrix which can be made of materials, usually polymers, which are degradable by enzymatic or acid-based hydrolysis or by dissolution.
  • a sustained-release matrix for example is chosen from biocompatible materials such as liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), polylactide co- glycolide (copolymers of lactic acid and glycolic acid), polyanhydrides, poly(ortho)esters, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxcylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • a preferred biodegradable matrix is a matrix of one of either polylactide, polyglycolide, or polylactide co-glycolide
  • the triciribine compounds and an erlotinib-like compound may also be administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically- acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the liposome can contain, in addition to one or more compositions of the present invention, stabilizers, preservatives, excipients, and the like. Examples of lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art.
  • the triciribine compounds and an erlotinib-like compound may be formulated as aerosols for application, such as by inhalation.
  • These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation will, in one embodiment, have diameters of less than 50 microns, in one embodiment less than 10 microns.
  • compositions including the triciribine compounds and an erlotinib-like compound, for example, gef ⁇ tinib, erlotinib or a salt thereof may be used in combination with other compositions and/or procedures for the treatment of the conditions described above.
  • a tumor may be treated conventionally with surgery, radiation, or chemotherapy combined with one or more compositions of the present invention and then one or more compositions of the present invention may be subsequently
  • compositions including triciribine compounds and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof can be formulated according to known methods for preparing pharmaceutically useful compositions.
  • Formulations are described in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. describes formulations which can be used in connection with the subject invention.
  • Formulations suitable for administration include, for example, aqueous sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powder, granules, tablets, etc. It should be understood that in addition to the ingredients particularly mentioned above, the formulations of the subject invention can include other agents conventional in the art having regard to the type of formulation in question.
  • the methods of the present invention for example, for inhibiting the growth of a cancerous cell, can be advantageously combined with at least one additional
  • 1 -W A/2866539.1 therapeutic method including but not limited to chemotherapy, radiation therapy, therapy that selectively inhibits Ras oncogenic signaling, or any other therapy known to those of skill in the art of the treatment and management of cancer, such as administration of an anti-cancer agent.
  • Administration of API-2 (triciribine) as a salt may be carried out.
  • Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, alpha-ketoglutarate, and alpha-glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • the triciribine compounds and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof can be formulated as pharmaceutical compositions and administered to a subject, such as a human or veterinary patient, in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the triciribine compounds and an erlotinib-like compound for example, gefitinib, erlotinib or a salt thereof of the present invention may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle (i.e., carrier) such as an inert diluent or an assimilable edible carrier.
  • a pharmaceutically acceptable vehicle i.e., carrier
  • an inert diluent such as an inert diluent or an assimilable edible carrier.
  • compositions and preparations should contain at least 0.1% of active agent.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of the active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • binders such as gum tragacanth, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, fructose, lactose or aspartame or
  • the unit dosage form When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like.
  • a syrup or elixir may contain the compounds of the invention, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially
  • the triciribine compounds and an erlotinib-like compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active agents or their salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders including the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium including, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the
  • injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating compounds of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the triciribine compounds and an erlotinib- like compound for example, gefitinib, erlotinib or a salt thereof may be applied in pure- form, i.e., when they are liquids.
  • a dermatologically acceptable carrier which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds of the invention can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be
  • the concentration of the active agent in a liquid composition can be from about 0.1-25 wt-%, or from about 0.5-10 wt.-%.
  • the concentration in a semi-solid or solid composition such as a gel or a powder can be about 0.1-5 wt.-%, preferably about 0.5- 2.5 wt.-%.
  • single dosages for injection, infusion or ingestion will generally vary between 5-1500 mg, and may be administered, i.e., 1-3 times daily, to yield levels of about 0.1-50 mg/kg, for adults.
  • a non-limiting dosage of the present invention can be between 7.5 to 45 mg per clay, administered orally, with appropriate adjustment for the body weight of an individual.
  • the present invention includes a pharmaceutical composition including triciribine compounds and an erlotinib-like compound, for example, gef ⁇ tinib, erlotinib or a salt thereof in combination with a pharmaceutically acceptable carrier.
  • 97 l-WA/2866539 1 compound for example, gefitinib, erlotinib or a salt thereof, constitute a preferred embodiment of the invention.
  • the dose administered to a subject, particularly a human, in the context of the present invention should be sufficient to affect a therapeutic response in the patient over a reasonable time frame.
  • dosage will depend upon a variety of factors including the condition of the animal, the body weight of the animal, as well as the severity and stage of the cancer.
  • a suitable dose is that which will result in a concentration of the triciribine compounds and an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof in tumor tissue which is known to affect the desired response.
  • the preferred dosage is the amount which results in maximum inhibition of cancer cell growth, without unmanageable side effects.
  • Administration of API-2 (or a pharmaceutically acceptable salt thereof) can be continuous or at distinct intervals, as can be determined by a person of ordinary skill in the art.
  • Mammalian species which benefit from the disclosed methods for the inhibition of cancer cell growth include, but are not limited to, primates, such as apes, chimpanzees, orangutans, humans, monkeys; domesticated animals (e.g., pets) such as dogs, cats, guinea pigs, hamsters, Vietnamese pot-bellied pigs, rabbits, and ferrets; domesticated farm animals such as cows, buffalo, bison, horses, donkey, swine, sheep, and goats; exotic animals typically found in zoos, such as bear, lions, tigers, panthers, elephants, hippopotamus, rhinoceros, giraffes, antelopes, sloth, gazelles, zebras, wildebeests, prairie dogs, koala bears, kangaroo, opossums, raccoons, pandas, hyena, seals, sea lions, elephant seals
  • patient and “subject” are used herein interchangeably and are intended to include such human and non-human mammalian species.
  • Patients in need of treatment using the methods of the present invention can be identified using standard techniques known to those in the medical profession.
  • Example 1 In Vitro Screening
  • the NCI Structural Diversity Set is a library of
  • Screening for Inhibition of Akt-transformed Cell Growth ⁇ AKT2 transformed NIH3T3 cells or LXSN vector-transfected NIH3T3 control cells (Cheng J.Q. et al., Oncogene, 1997. 14: p. 2793-2801) are plated into 96-well tissue culture plate. Following treatment with 5 ⁇ M of NCI Diversity Set compound, cell growth can be detected with CellTier 96 One Solution Cell Proliferation kit (Promega). Compounds that inhibit growth in AKT2-transformed but not LXSN-transfected NIH3T3 cells are considered as candidates of Akt inhibitor and subjected to further analysis.
  • Akt is considered as an attractive molecular target for development of novel cancer therapeutics.
  • a chemical library of 1,992-compounds from the NCI is evaluated for agents capable of inhibition of growth in AKT2- transformed but not empty vector LXSN-transfected NIH3T3 cells. Repeated experiments showed that 32 compounds inhibited growth only in AKT2-transformaed cells.
  • API-2 (NCI identifier: NSC 154020), can suppress cell growth at a concentration of 50 nM.
  • Figure IA shows the chemical structure of API-2, which is also known as triciribine (Schweinsberg P.D. et al., Biochem Pharmacol, 1981. 30: p. 2521-2526).
  • API-2 inhibits selectively AKT-2 transformed cells over untransformed parental cells prompted us to determine whether API-2 is an inhibitor of AKT2 kinase.
  • AKT2 is immunoprecipitated with anti-AKT2 antibody from AKT-2 transformed NIH3T3 cells following treatment with API-2.
  • AKT2 immunoprecipitates were immunoblotted with anti-phospho-Akt antibodies.
  • API-2 significantly inhibited
  • HEK293 cells are transfected with HA-Aktl, -AKT2 and -AKT3, serum-starved overnight and treated with API-2 for 60 min prior to EGF (50 ng/ml) stimulation.
  • API-2 Does Not Inhibit Known Upstream Activators of Akt. It has been well documented that Akt is activated by extracellular stimuli and intracellular signal molecules, such as active Ras and Src, through a PI3K-dependent manner. Therefore, API-2 inhibition of Akt could result from targeting upstream molecule(s) of Akt. As PI3K and PDKl are direct upstream regulators of Akt (Datta S.R. et al., Genes Dev, 1999. 13: p. 2905-2927), whether API-2 inhibits PI3K and/or PDKl is examined.
  • HEK293 cells are serum-starved and then can be treated with API-2 or PI3K inhibitor, wortmannin, for 30 min prior to EGF stimulation.
  • PI3K is immunoprecipitated with anti-pllOQantibody.
  • the immunoprecipitates are subjected to in vitro PI3K kinase assay using PI-4-P as a substrate.
  • the EGF-induced PI3K activity is inhibited by wortmannin but not by API-2.
  • API-2 Is Highly Selective for the Akt over PKC, PKA, SGK, STAT,
  • Akt belongs to AGC (PKA/PKG/PKC) kinase family, which also include PKA, PKC, serum- and glucocorticoid-inducible kinase (SGK), p90 ribosomal S6 kinase, p70 S6K , mitogen- and stress-activated protein kinase and PKC-related kinase.
  • PKA PKA/PKG/PKC
  • SGK serum- and glucocorticoid-inducible kinase
  • SGK serum- and glucocorticoid-inducible kinase
  • SGK serum- and glucocorticoid-inducible kinase
  • SGK serum- and glucocorticoid-inducible kinase
  • SGK serum- and glucocorticoid-inducible kinase
  • HEK293 cells are transfected with HA-tagged PKA, PKC ⁇ % or SGK.
  • In vitro kinase assay and immunoblotting analysis show that the kinase activities of PKA and PKC D are inhibited by PKAI and Ro 31-8220, a PKC inhibitor, respectively, whereas API-2 exhibits no effect on their activities ( Figure 2C and 2E). Further, serum-induced SGK kinase activity is attenuated by wortmannin but not by API-2 ( Figure 2D). In addition, it is determined whether API-2 has effect on other oncogenic survival pathways.
  • Western blotting analyses with commercially available anti-phospho-antibodies reveals that phosphorylation levels of Stat3, JNK, p38 and Erkl/2 were not affected by API-2
  • API-2 Suppresses Cell Growth and Induces Apoptosis in Akt- overexpressing/activating Human Cancer Cell Lines.
  • the ability of API-2 to selectively inhibit the Akt pathway suggests that it should inhibit proliferation and/or induces apoptosis preferentially in those tumor cells with aberrant expression/activation of Akt.
  • API-2 is used to treat the cells that express constitutively active Akt, caused by overexpression of AKT2 (OVCAR3, OVCAR8, PANC 1 and AKT2-transformed NIH3T3) or mutations of the PTEN gene (PC-3, LNCaP, MDA-MB-468), and cells that do not (OVCAR5, DU-145, T47D, COLO357 and LXSN-NIH3T3) as well as melanoma cells that are activated by IGF-I to activate Akt or do not respond to growth stimulation by IGF-I (Satyamoorthy K. et al., Cancer Res, 2001. 61: p.
  • API-2 treatment inhibited cell proliferation by approximately 50-60% in Akt-overexpressing/activating cell lines, LNCaP, PC-3, OVCAR3, OVCA8, PANCl, MDA-MB-468, and WM35, whereas only by about 10- 20% in DU145, OVCAR5, COLO357, T47D and WM852 cells, which exhibit low levels of Akt or do not respond to growth stimulation by IGF-I.
  • API-2 induces apoptosis by 8-fold (OVCAR3), 6-fold (OVCAR8), 6-fold (PANCl), and 3-fold (AKT2-NIH3T3). No significant difference of apoptosis is observed between API-2
  • API-2 inhibits cell growth and induces apoptosis preferentially in cells that express aberrant Akt.
  • API-2 Inhibits Downstream Targets of Akt It has been shown that Akt exerts its cellular effects through phosphorylation of a number of proteins (Datta
  • Akt substrates More than 20 proteins have been identified as Akt substrates, including the members of Forkhead protein family (FKHR, AFX and FKHRLl), tuberlin/TSC2, p70 S6K , GSK-3D, p21 WAF1/cipI , p27 kipl , MDM2, Bad, ASKl and IKKD etc. It is next examined whether API-2 inhibits downstream targets of Akt. As anti-phospho-tuberlin, -Bad, -AFX, and -GSK-3 ⁇ antibodies are commercially available, therefore, the effect of API-2 on their phosphorylation induced by Akt was determined.
  • FIG. 4A shows that API-2 considerably inhibited the phosphorylation levels of tuberlin leading to stabilization and upregulation of tuberin (Dan, H.C., et al., J Biol Chem, 2002. 277: p. 35364-35370).
  • the phosphorylation levels of Bad, GSK-3 ⁇ , and AFX are partially attenuated by API-2.
  • API-2 inhibition of Akt downstream targets at different degrees could be due to the fact that phosphorylation sites of these targets are also regulated by other kinase(s), for instance, Bad serine- 136 is phosphorylated by PAKl in addition to Akt (Schurmann, A., et al. MoI Cell Biol, 2000. 20: p. 453-461).
  • Example 2 Antitumor Activity in the Nude Mouse Tumor Xenograft
  • Tumor cells can be harvested, suspended in PBS, and can be injected s.c. into the right and left flanks (2 x 10 6 cells/flank) of 8-week-old female nude mice as reported previously ( Sun, J. et al., Cancer Res, 1999. 59: p. 4919-4926, 1999).
  • animals are randomized and dosed i.p. with 0.2 ml vehicle of the triciribine compound and/or an erlotinib-like compound, for example, gefitinib, erlotinib or a salt thereof, daily.
  • Control animals receive DMSO (20%) vehicle, whereas treated animals can be injected with API-2 (1 mg/kg/day) in 20% DMSO.
  • API-2 Inhibits the Growth of Tumors in Nude Mice that Overexpress
  • Akt Frequent overexpression/activation and/or amplification of AKTl and AKT2 in human ovarian and pancreatic cancer was shown (Cheng J.Q. and Nicosia S.V., in Encyclopedic Reference of Cancer. 2001, Schwab D, Ed. Springer, Berlin Heidelberg and New York, p. 35-7). Inhibition of Akt pathway by inhibitors of PI3K, HSP70, Src and farnesyltransferase resulted in cell growth arrest and induction of apoptosis (Solit D.B. et al., Cancer Res, 2003. 63: p. 2139-2144; Xu, W., et al. Cancer Res, 2003.63: p. 7777-7784).
  • mice implanted into the right flank, and those cell lines that express low levels of Akt (OVCAR5 and COLO357) into the left flank of mice.
  • the tumors reach an average size of about 100-150 mm 3 , the animals are randomized and treated i.p. with
  • Example 3 TCN directly inhibits wild type Akt kinase activity
  • API-2 can directly inhibit wild type Akt kinase activity induced by PDKl in vitro ( Figure 1). This result supports that API-2 is a direct Akt inhibitor and that the underlying mechanism may be API-2 binding to PH domain and/or threonine- 308 of Akt.
  • An in vitro kinase assay is performed with recombinant of PDKl and Akt in a kinase buffer containing phosphatidylinositol-3,4,5-P3 (PIP3), API-2 and histone H2B as substrate. After incubation of 30 min, the reactions were separated by SDS-PAGE and exposed in a film.
  • TCN is effective in cancer resistant cells
  • TCN potentiates growth inhibition by trastuzumab and
  • Akt inhibitor 4ADPIB (4-amino-2(3,4-dichloro-phenyl) -N- (1 H-indazol-5-yl) -butyramide) (US patent 6,919340), was synthesized.
  • PTEN antibodies were from Santa Cruz Biotechnology (Santa Cruz, CA).
  • 3-actin antibodies were from Sigma (St. Louis, MO). All other antibodies were purchased from Cell Signaling Technology (Danvers, MA).
  • Treated cells were compared to control DMSO treated BT474.ml cells to calculate percentage of growth inhibition.
  • BT474.ml cells were plated in 6-well plates (4-6xlO 5 cells/ well). Twenty-four hours after plating, the cells were treated as indicated for 72 hours with trastuzumab, TCN and/or RADOOl. The floating and adherent cells were collected, labeled and stained using the APO-BRDUTM TUNEL assay kit (Phoenix Flow Systems, San Diego, CA) according to the manufacturer's protocol. Data was collected and analyzed using a
  • BT474.ml cells are a tumorigenic subline of the BT474 breast cancer cell line and express high levels of ErbB2. When PTEN levels are decreased by transfection with
  • PTEN AS PTEN antisense oligonucleotides
  • BT474 BT474.ml breast cancer cells become more resistant to the anti-proliferative effects of trastuzumab than cells with normal levels of PTEN and provide a good experimental model for breast cancers in which trastuzumab resistance is caused by PTEN loss (Nagata Y. et al., Cancer Cell, 2004. 6(2): p. 117-27 and Figures 9B and 9C).
  • Nonspecific oligonucleotides (NS) were transfected as controls. Treatment with PTEN AS oligonucleotides effectively lowered PTEN levels ( Figure 1 IA and HB). Neither PTEN AS nor NS control oligonucleotides altered ErbB2 levels in the cells.
  • PTEN AS and NS-transfected BT474.ml cells were treated with each of the 6 compounds or trastuzumab alone and in combination for 5 days and evaluated cell proliferation as compared to DMSO-treated control.
  • growth inhibition as a biological endpoint, we compared the ability of each drug to exhibit cooperative effects with trastuzumab using doses of drug that resulted in -20-40% growth inhibition when administered alone ( Figure 9A).
  • Figure 9A Almost all of the compounds displayed growth inhibitory effects, particularly at high concentrations and in cells with intact PTEN ( Figure 9A).
  • Example 6 TCN inhibits activation of Akt and mTOR Inhibition of downstream signaling molecules.
  • Immunoblot analysis verified that triciribine and RADOOl blocked activation of Akt and mTOR, important signaling molecules activated by ErbB2 and the targets of triciribine and RADOOl respectively.
  • Example 7 TCN and trastuzumab inhibit tumor growth in PTEN- deficient tumors
  • mice 111 l-WA/2866539.1 severe combined immunodeficiency mice were from Taconic Farms (Hudson, NY). Tumor xenografts were performed as described in Nagata Y. et al., Cancer Cell, 2004. 6(2): p. 117-27. When the xenograft tumors reached the average size of 100-150 mm 3 , the mice were divided into 6 groups, each with 7 mice and an even distribution of tumor sizes, and treated as follows. PTEN antisense (30 pg) oligonucleotides were administered to each mouse weekly via intratumor injection. One week after PTEN AS oligonucleotide administration was initiated, drug treatment began.

Abstract

Cette invention concerne des traitements combinés comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique. L'invention concerne en particulier des composés du type erlotinib et des compositions à toxicité réduite utilisés dans le traitement et la prévention des tumeurs, du cancer et d'autres affections associées avec la prolifération anormale de cellules.
PCT/US2007/024873 2006-12-05 2007-12-05 Compositions comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique ou leurs sels et leurs procédés d'utilisation WO2008070100A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US87277806P 2006-12-05 2006-12-05
US60/872,778 2006-12-05

Publications (2)

Publication Number Publication Date
WO2008070100A1 true WO2008070100A1 (fr) 2008-06-12
WO2008070100A8 WO2008070100A8 (fr) 2008-08-14

Family

ID=39148288

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/024873 WO2008070100A1 (fr) 2006-12-05 2007-12-05 Compositions comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique ou leurs sels et leurs procédés d'utilisation

Country Status (2)

Country Link
TW (1) TW200835507A (fr)
WO (1) WO2008070100A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008057503A2 (fr) * 2006-11-06 2008-05-15 Vioquest Pharmaceuticals, Inc. Compositions contenant de la triciribine et des taxanes et méthodes d'utilisation associées
WO2009139766A1 (fr) * 2008-05-12 2009-11-19 University Of South Florida Thérapie de combinaison anticancéreuse comprenant de la triciribine
US8796455B2 (en) 2005-11-17 2014-08-05 OSI Pharmaceuticals, LLC Fused bicyclic mTOR inhibitors
US8901086B2 (en) 2004-03-29 2014-12-02 University Of South Florida Compositions including triciribine and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005094322A2 (fr) * 2004-03-29 2005-10-13 University Of South Florida Traitement efficace des tumeurs et du cancer a l'aide de la triciribine et de composes associes
WO2005110477A2 (fr) * 2004-04-09 2005-11-24 University Of South Florida Polytherapies pour le cancer et des angiopathies proliferantes
WO2006125539A2 (fr) * 2005-05-27 2006-11-30 Bayer Healthcare Ag Therapie de combinaisons comprenant des urees diaryle destinee a traiter des maladies

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005094322A2 (fr) * 2004-03-29 2005-10-13 University Of South Florida Traitement efficace des tumeurs et du cancer a l'aide de la triciribine et de composes associes
WO2005110477A2 (fr) * 2004-04-09 2005-11-24 University Of South Florida Polytherapies pour le cancer et des angiopathies proliferantes
WO2006125539A2 (fr) * 2005-05-27 2006-11-30 Bayer Healthcare Ag Therapie de combinaisons comprenant des urees diaryle destinee a traiter des maladies

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANNALS NEW YORK ACADEMY OF SCIENCE, 2005, XP009097340 *
KONECNY GOTTFRIED E ET AL: "Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells", CANCER RESEARCH, vol. 66, no. 3, February 2006 (2006-02-01), pages 1630 - 1639, XP002472825, ISSN: 0008-5472 *
LI Q: "Recent progress in the discovery of Akt inhibitors as anticancer agents", EXPERT OPINION ON THERAPEUTIC PATENTS 2007 GB, vol. 17, no. 9, 2007, pages 1077 - 1130, XP002472827, ISSN: 1354-3776 *
LU CHIEN-HSING ET AL: "Preclinical testing of clinically applicable strategies for overcoming trastuzumab resistance caused by PTEN deficiency.", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH 1 OCT 2007, vol. 13, no. 19, 1 October 2007 (2007-10-01), pages 5883 - 5888, XP002472826, ISSN: 1078-0432 *
NORMANNO N ET AL: "Is the gefitinib plus trastuzumab combination feasible in breast cancer patients?", ANNALS OF ONCOLOGY : OFFICIAL JOURNAL OF THE EUROPEAN SOCIETY FOR MEDICAL ONCOLOGY / ESMO OCT 2005, vol. 16, no. 10, October 2005 (2005-10-01), pages 1709, XP002472824, ISSN: 0923-7534 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8901086B2 (en) 2004-03-29 2014-12-02 University Of South Florida Compositions including triciribine and methods of use thereof
US9511084B2 (en) 2004-03-29 2016-12-06 University Of Florida Compositions including triciribine and methods of use thereof
US8796455B2 (en) 2005-11-17 2014-08-05 OSI Pharmaceuticals, LLC Fused bicyclic mTOR inhibitors
WO2008057503A2 (fr) * 2006-11-06 2008-05-15 Vioquest Pharmaceuticals, Inc. Compositions contenant de la triciribine et des taxanes et méthodes d'utilisation associées
WO2008057503A3 (fr) * 2006-11-06 2008-09-04 Vioquest Pharmaceuticals Inc Compositions contenant de la triciribine et des taxanes et méthodes d'utilisation associées
WO2009139766A1 (fr) * 2008-05-12 2009-11-19 University Of South Florida Thérapie de combinaison anticancéreuse comprenant de la triciribine

Also Published As

Publication number Publication date
TW200835507A (en) 2008-09-01
WO2008070100A8 (fr) 2008-08-14

Similar Documents

Publication Publication Date Title
CA2561513C (fr) Traitement efficace des tumeurs et du cancer a l'aide de la triciribine et de composes associes
WO2008086005A1 (fr) Compositions contenant de la triciribine et du bortézomib et des dérivés de ces derniers, procédés d'utilisation correspondants
US9486492B2 (en) Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
US9486522B2 (en) Compositions including triciribine and trastuzumab and methods of use thereof
WO2008070100A1 (fr) Compositions comprenant des composés à base de triciribine et des composés inhibiteurs des récepteurs du facteur de croissance épidermique ou leurs sels et leurs procédés d'utilisation
WO2008121261A2 (fr) Traitement efficace de tumeurs et de cancer au moyen de promédicaments de la triciribine
WO2008060581A2 (fr) Compositions renfermant de la triciribine et du trastuzumab, et leurs méthodes d'utilisation
US10780104B2 (en) Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
CA2724246C (fr) Therapie de combinaison anticancereuse comprenant de la triciribine
WO2008070136A1 (fr) Compositions comprenant de la triciribine et un ou plusieurs composés de platine, et leurs procédés d'utilisation
EP2182939A2 (fr) Compositions contenant de la triciribine et des taxanes et méthodes d'utilisation associées
US9486470B2 (en) Compositions including triciribine and one or more platinum compounds and methods of use thereof
US20110223154A1 (en) Compositions including triciribine and methods of use thereof
WO2008069922A9 (fr) Compositions comprenant des analogues de la triciribine et de l'anthracycline et procédés d'utilisation de celles-ci

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07862515

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07862515

Country of ref document: EP

Kind code of ref document: A1