Treatment of cancer with TG02

Information

  • Patent Grant
  • 11865116
  • Patent Number
    11,865,116
  • Date Filed
    Friday, March 24, 2017
    7 years ago
  • Date Issued
    Tuesday, January 9, 2024
    3 months ago
  • Inventors
  • Original Assignees
    • COTHERA BIOSCIENCE, INC.
  • Examiners
    • Canella; Karen A.
    Agents
    • Sterne, Kessler, Goldstein & Fox P.L.L.C.
Abstract
The present disclosure provides therapeutic methods of treating a cancer patient with TG02 and a second therapeutic agent, e.g., TG02 and an immune checkpoint inhibitor, TG02 and a COX-2 inhibitor, or TG02 and an immune checkpoint inhibitor and a COX-2 inhibitor.
Description
BACKGROUND OF THE INVENTION
Field of the Invention

The present disclosure provides therapeutic methods of treating a cancer patient with TG02 and a second therapeutic agent, e.g., TG02 and an immune checkpoint inhibitor, TG02 and a COX-2 inhibitor, and TG02 and an immune checkpoint inhibitor and a COX-2 inhibitor.


Background

TG02 is a pyrimidine-based multi-kinase inhibitor that inhibits CDKs 1, 2, 5, 7 and 9 together with JAK2 and FLT3. It dose-dependently inhibits signaling pathways downstream of CDKs, JAK2 and FLT3 in cancer cells with the main targets being CDKs. TG02 is anti-proliferative in a broad range of tumor cell lines, inducing G1 cell cycle arrest and apoptosis. Primary cultures of progenitor cells derived from acute myeloid leukemia (AML) and polycythemia vera patients are very sensitive to TG02. Comparison with reference inhibitors that block only one of the main targets of TG02 demonstrate the benefit of combined CDK and JAK2/FLT3 inhibition in cell lines as well as primary cells. See Goh et al., Leukemia 26:236-43 (2012). TG02 is also known as SB1317 and by its chemical name: (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene. TG02 is disclosed as Compound 1 in U.S. Pat. No. 8,143,255. U.S. Pat. No. 9,120,815 discloses various salt, e.g., TG02 citrate, and crystalline forms of TG02. The chemical structure of TG02 is:




embedded image


BRIEF SUMMARY OF THE INVENTION

In one aspect, the present disclosure provides therapeutic methods of treating a cancer patient, the methods comprising administering to the patient a therapeutically effective amount of TG02. In another aspect, the patient's cancer is characterized as overexpressing of MYC, MCL1, or both.


In another aspect, the present disclosure provides therapeutic methods of treating a cancer patient, the methods comprising administering to the patient therapeutically effective amounts of TG02 and an immune checkpoint inhibitor, e.g., a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, a LAG3 inhibitor, a TIM3 inhibitor, or a cd47 inhibitor.


In another aspect, the present disclosure provides therapeutic methods of treating a cancer patient, the methods comprising administering to the patient therapeutically effective amounts of TG02 and a COX inhibitor, e.g., apricoxib or 6-bromo-8-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid.


In another aspect, the present disclosure provides therapeutic methods of treating a cancer patient, the methods comprising administering to the patient therapeutically effective amounts of TG02, an immune checkpoint inhibitor, and a COX-2 inhibitor.


In another aspect, present disclosure provides therapeutic methods of treating a cancer patient who has tumors that overexpress MYC, MCL1, or both.


In another aspect, the present disclosure provides kits comprising TG02, TG02 and an immune checkpoint inhibitor, TG02 and a COX-2 inhibitor, and TG02 and an immune checkpoint inhibitor and a COX-2 inhibitor.


In another aspect, the present disclosure provides a pharmaceutical composition comprising TG02, a COX-2 inhibitor, e.g., apricoxib or 6-bromo-8-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid and a pharmaceutically acceptable excipient.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 is a bar graph showing the in vitro activity of TG02, TMZ (temozolomide), and TG02+TMZ in GSC923 cells.



FIG. 2 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in LN18 cells.



FIG. 3 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in T98G cells.



FIG. 4 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in U251 cells.



FIG. 5 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in U87 cells.



FIG. 6 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in LN299 cells.



FIG. 7 is a bar graph showing the in vitro activity of TG02, TMZ, and TG02+TMZ in GSC827 cells



FIG. 8 is a bar graph showing the in vitro cytotoxicity of TG02, TMZ, and TG02+TMZ (T+T) in GSC923 cells.



FIG. 9 is a bar graph showing the in vitro cytotoxicity of TG02, TMZ, and TG02+TMZ (T+T) in U251 cells.



FIG. 10 is a bar graph showing a lack of in vitro activity of TG02, TMZ, and TG02+TMZ (T+T) in human pulmonary arterial endothelial cells.



FIG. 11 is a bar graph showing a lack of in vitro activity of TG02, TMZ, and TG02+TMZ (T+T) in human astrocytes.



FIG. 12 is a dose response curve showing the in vitro activity of TG02 and TG02+TMZ in GSC923 cells.



FIG. 13 is a dose response curve showing the in vitro activity of TMZ and TG02+TMZ in GSC923 cells.



FIG. 14 is a dose response curve showing the in vitro activity of TG02 and TG02+TMZ in U251 cells.



FIG. 15 is a dose response curve showing the in vitro activity of TMZ and TG02+TMZ in U251 cells.



FIG. 16 is a schematic illustration of TG02 and TMZ administration in a mouse glioma GL261 cell allograft model.



FIG. 17 is a line graph showing percent survival following TG02, TMZ, and TG02+TMZ administration in a mouse glioma GL261 cell allograft model.



FIG. 18 is a line graph showing the tumor burden following TG02, TMZ, and TG02+TMZ administration in a mouse glioma GL261 cell allograft model.



FIG. 19 is an illustration showing the effect of TG02 on MYC protein levels in hepatocellular carcinoma (HCC) cells.



FIG. 20 is a dose response curve showing the effect of TG02 on MYC protein levels in HCC cells.



FIG. 21 is is an illustration showing showing the effect of TG02 on MYC protein levels in HCC tumor cells.



FIG. 22 is a line graph showing the in vivo activity of TG02 and TG02+sorafenib in an orthotopic model of HepG2 HCC xenografts.



FIG. 23 is a bar graph showing PD-L1 expression following treatment with TG02 in a transgenic mouse model of MYC-induced T cell acute lymphoblastic leukemia.



FIG. 24 is a bar graph showing CD47 expression following treatment with TG02 in a transgenic mouse model of MYC-induced T cell acute lymphoblastic leukemia.



FIG. 25 is a bar graph showing BCL-xL expression following treatment with TG02 in a transgenic mouse model of MYC-induced T cell acute lymphoblastic leukemia.



FIG. 26 is a bar graph showing MYC expression following treatment with TG02 in a transgenic mouse model of MYC-induced T cell acute lymphoblastic leukemia.



FIG. 27 is a line graph showing the efficacy of TG02 in combination with anti-PD-1 in a mouse syngeneic GL261 orthotopic glioblastoma model.



FIG. 28 is an illustration showing that BT245 tumor cells exposed to TG02 show inhibition of MYC and MCL-1 expression.



FIG. 29 is a bar graph showing the area under the curve (AUC) for TG02 induced inhibition in glioblastoma (GBM) cells.



FIG. 30 is a scatter graph showing that high MYC expression correlates with low AUC in GBM cells.



FIG. 31 is a series of six line graphs showing the activity of TG02 in combination with radiation in glioblastoma cell lines.



FIG. 32 is a bar graph showing the activity of TG02 on 26 patient-derived GBM stem cell lines.



FIG. 33 is an illustration showing the expression level of CDK9, MYC, and Mcl-1 in patient-derived GBM stem cell lines following treatment with TG02.





DETAILED DESCRIPTION OF THE INVENTION

In one embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient a therapeutically effective amount of TG02, wherein one or more of the genes listed in Table 1, see below, is differentially present in a biological sample taken from the patient as compared with a biological sample taken from a subject of another phenotypic status. In another embodiment, MYC overexpression is differentially present in a sample taken from the patient. In another embodiment, MCL1 overexpression is differentially present in a sample taken from the patient.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient a therapeutically effective amounts of TG02 and an immune checkpoint inhibitor, wherein one or more of the genes listed in Table 1, see below, is differentially present in a biological sample taken from the patient as compared with a biological sample taken from a subject of another phenotypic status. In another embodiment, MYC overexpression is differentially present in a sample taken from the patient. In another embodiment, MCL1 overexpression is differentially present in a sample taken from the patient. In another embodiment, TG02 is administered to the patient before the immune checkpoint inhibitor. In another embodiment, TG02 is administered to the patient after the immune checkpoint inhibitor. In another embodiment, TG02 is administered to the patient at the same time as an immune checkpoint inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient a therapeutically effective amounts of TG02, an immune checkpoint inhibitor, and a COX-2 inhibitor wherein one or more of the genes listed in Table 1, see below, is differentially present in a biological sample taken from the patient as compared with a biological sample taken from a subject of another phenotypic status. In another embodiment, MYC overexpression is differentially present in a sample taken from the patient. In another embodiment, MCL1 overexpression is differentially present in a sample taken from the patient. In another embodiment, TG02 is administered to the patient before the COX-2 inhibitor. In another embodiment, TG02 is administered to the patient after the COX-2 inhibitor. In another embodiment, TG02 is administered to the patient at the same time as the COX-2 inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient therapeutically effective amounts of TG02 and an immune checkpoint inhibitor. In another embodiment, TG02 is administered to the patient before the immune checkpoint inhibitor. In another embodiment, TG02 is administered to the patient after the immune checkpoint inhibitor. In another embodiment, TG02 is administered to the patient at the same time as an immune checkpoint inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient therapeutically effective amounts of TG02, an immune checkpoint inhibitor, and a COX-2 inhibitor. In another embodiment, TG02 is administered to the patient before the COX-2 inhibitor. In another embodiment, TG02 is administered to the patient after the COX-2 inhibitor. In another embodiment, TG02 is administered to the patient at the same time as the COX-2 inhibitor.


In another embodiment, the present disclosure provides kits comprising TG02 and an immune checkpoint inhibitor, and instructions for administering TG02 and the immune checkpoint inhibitor to a patient having cancer. In another embodiment, the kit further comprises a COX-2 inhibitor.


In another embodiment, the present disclosure provides kits comprising TG02 and a COX-2 inhibitor, and instructions for administering TG02 and the COX-2 inhibitor to a patient having cancer.


In another embodiment, the kit is packaged in a manner that facilitates its use to practice methods of the present disclosure.


In another embodiment, the kit includes TG02 (or a composition comprising TG02) packaged in a container, such as a sealed bottle or vessel, with a label affixed to the container or included in the kit that describes use of TG02 or composition to practice the method of the disclosure. In one embodiment, TG02 is packaged in a unit dosage form. The kit further can include a device suitable for administering the composition according to the intended route of administration.


The disclosure provides various therapeutic methods, kits, and compositions relating to the treatment of cancer. In one embodiment, the cancer is a solid tumor. In another embodiment, the cancer is a hematological malignancy. In another embodiment, the cancer selected from the group consisting of adrenal cancer, acinic cell carcinoma, acoustic neuroma, acral lentigious melanoma, acrospiroma, acute eosinophilic leukemia, acute erythroid leukemia, acute lymphoblastic leukemia, acute megakaryoblastic leukemia, acute monocytic leukemia, acute promyelocytic leukemia, adenocarcinoma, adenoid cystic carcinoma, adenoma, adenomatoid odontogenic tumor, adenosquamous carcinoma, adipose tissue neoplasm, adrenocortical carcinoma, adult T-cell leukemia/lymphoma, aggressive NK-cell leukemia, AIDS-related lymphoma, alveolar rhabdomyosarcoma, alveolar soft part sarcoma, ameloblastic fibroma, anaplastic large cell lymphoma, anaplastic thyroid cancer, angioimmunoblastic T-cell lymphoma, angiomyolipoma, angiosarcoma, astrocytoma, atypical teratoid rhabdoid tumor, B-cell chronic lymphocytic leukemia, B-cell prolymphocytic leukemia, B-cell lymphoma, basal cell carcinoma, biliary tract cancer, bladder cancer, blastoma, bone cancer, Brenner tumor, Brown tumor, Burkitt's lymphoma, breast cancer, brain cancer, carcinoma, carcinoma in situ, carcinosarcoma, cartilage tumor, cementoma, myeloid sarcoma, chondroma, chordoma, choriocarcinoma, choroid plexus papilloma, clear-cell sarcoma of the kidney, craniopharyngioma, cutaneous T-cell lymphoma, cervical cancer, colorectal cancer, Degos disease, desmoplastic small round cell tumor, diffuse large B-cell lymphoma, dysembryoplastic neuroepithelial tumor, dysgerminoma, embryonal carcinoma, endocrine gland neoplasm, endodermal sinus tumor, enteropathy-associated T-cell lymphoma, esophageal cancer, fetus in fetu, fibroma, fibrosarcoma, follicular lymphoma, follicular thyroid cancer, ganglioneuroma, gastrointestinal cancer, germ cell tumor, gestational choriocarcinoma, giant cell fibroblastoma, giant cell tumor of the bone, glial tumor, glioblastoma, glioma, gliomatosis cerebri, glucagonoma, gonadoblastoma, granulosa cell tumor, gynandroblastoma, gallbladder cancer, gastric cancer, hairy cell leukemia, hemangioblastoma, head and neck cancer, hemangiopericytoma, hematological malignancy, hepatoblastoma, hepatocellular carcinoma, hepatosplenic T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, invasive lobular carcinoma, intestinal cancer, kidney cancer, laryngeal cancer, lentigo maligna, lethal midline carcinoma, leukemia, leydig cell tumor, liposarcoma, lung cancer, lymphangioma, lymphangiosarcoma, lymphoepithelioma, lymphoma, acute lymphocytic leukemia, acute myelogeous leukemia, chronic lymphocytic leukemia, liver cancer, small cell lung cancer, non-small cell lung cancer, MALT lymphoma, malignant fibrous histiocytoma, malignant peripheral nerve sheath tumor, malignant triton tumor, mantle cell lymphoma, marginal zone B-cell lymphoma, mast cell leukemia, mediastinal germ cell tumor, medullary carcinoma of the breast, medullary thyroid cancer, medulloblastoma, melanoma, meningioma, merkel cell cancer, mesothelioma, metastatic urothelial carcinoma, mixed Mullerian tumor, mucinous tumor, multiple myeloma, muscle tissue neoplasm, mycosis fungoides, myxoid liposarcoma, myxoma, myxosarcoma, nasopharyngeal carcinoma, neurinoma, neuroblastoma, neurofibroma, neuroma, nodular melanoma, ocular cancer, oligoastrocytoma, oligodendroglioma, oncocytoma, optic nerve sheath meningioma, optic nerve tumor, oral cancer, osteosarcoma, ovarian cancer, Pancoast tumor, papillary thyroid cancer, paraganglioma, pinealoblastoma, pineocytoma, pituicytoma, pituitary adenoma, pituitary tumor, plasmacytoma, polyembryoma, precursor T-lymphoblastic lymphoma, primary central nervous system lymphoma, primary effusion lymphoma, preimary peritoneal cancer, prostate cancer, pancreatic cancer, pharyngeal cancer, pseudomyxoma periotonei, renal cell carcinoma, renal medullary carcinoma, retinoblastoma, rhabdomyoma, rhabdomyosarcoma, Richter's transformation, rectal cancer, sarcoma, Schwannomatosis, seminoma, Sertoli cell tumor, sex cord-gonadal stromal tumor, signet ring cell carcinoma, skin cancer, small blue round cell tumors, small cell carcinoma, soft tissue sarcoma, somatostatinoma, soot wart, spinal tumor, splenic marginal zone lymphoma, squamous cell carcinoma, synovial sarcoma, Sezary's disease, small intestine cancer, squamous carcinoma, stomach cancer, T-cell lymphoma, testicular cancer, thecoma, thyroid cancer, transitional cell carcinoma, throat cancer, urachal cancer, urogenital cancer, urothelial carcinoma, uveal melanoma, uterine cancer, verrucous carcinoma, visual pathway glioma, vulvar cancer, vaginal cancer, Waldenstrom's macroglobulinemia, Warthin's tumor, and Wilms' tumor.


In another embodiment, the cancer is selected from the group consisting of squamous cell carcinoma of the head and neck, adenocarcinoma squamous cell carcinoma of the esophagus, adenocarcinoma of the stomach, adenocarcinoma of the colon, hepatocellular carcinoma, cholangiocarcinoma of the biliary system, adenocarcinoma of gall bladder, adenocarcinoma of the pancreas, ductal carcinoma in situ of the breast, adenocarcinoma of the breast, adenocarcinoma of the lungs, squamous cell carcinoma of the lungs, transitional cell carcinoma of the bladder, squamous cell carcinoma of the bladder, squamous cell carcinoma of the cervix, adenocarcinoma of the cervix, endometrial carcinoma, penile squamous cell carcinoma, and squamous cell carcinoma of the skin.


In another embodiment, a precancerous tumor is selected from the group consisting of leukoplakia of the head and neck, Barrett's esophagus, metaplasia of the stomach, adenoma of the colon, chronic hepatitis, bile duct hyperplasia, pancreatic intraepithelial neoplasia, atypical adenomatous hyperplasia of the lungs, dysplasia of the bladder, cervical initraepithelial neoplasia, penile intraepithelial neoplasia, and actinic keratosis of the skin.


In another embodiment, the patient has tumors that overexpress MYC, MCL1, or both. The tumors may be determined to overexpress MYC, MCL1, or both, by methods known in the art.


In another embodiment, the cancer is selected from the group consisting of hepatocellular carcinoma, glioblastoma, lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, and colorectal cancer.


In another embodiment, the cancer is selected from the group consisting of glioblastoma, hepatocellular carcinoma, non-small cell and small-cell lung cancer, head and neck cancer, colorectal carcinoma, and triple-negative breast cancer.


In another embodiment, the cancer has become resistant to conventional cancer treatments. The term “conventional cancer treatments” as used herein refers to any cancer drugs or biologics, or combination of cancer drugs and/or biologics that have been tested and/or approved for therapeutic use in humans by the U.S. Food and Drug Administration, European Medicines Agency, or similar regulatory agency.


In another embodiment, the patient has been treated previously with an immune checkpoint inhibitor without TG02. For example, the previous immune checkpoint therapy may be an anti-PD-1 therapy.


In another embodiment, the patient has been treated previously with a COX-2 inhibitor without TG02.


In another embodiment, the present disclosure provides a pharmaceutical composition comprising TG02, a COX-2 inhibitor, and a pharmaceutically acceptable excipient.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient a therapeutically effective amount of TG02, wherein the phenotypic status of the patient is overexpression of MYC, overexpression of MCL1, or overexpression of MYC and MCL1. In another embodiment, the cancer is selected from the group consisting of hepatocellular carcinoma, glioblastoma, lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, and colorectal cancer.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, the method comprising administering to the patient therapeutically effective amounts of TG02 and a second therapeutic agent, wherein the second therapeutic agent is neither an immune checkpoint inhibitor nor a COX-2 inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, comprising administering to the patient therapeutically effective amounts of TG02, an immune checkpoint inhibitor, and a third therapeutic agent, wherein the third therapeutic agent is not a COX-2 inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, comprising administering to the patient therapeutically effective amounts of TG02, a COX-2 inhibitor, and a third therapeutic agent, wherein the third therapeutic agent is not an immune checkpoint inhibitor.


In another embodiment, the present disclosure provides therapeutic methods of treating a patient having cancer, comprising administering to the patient therapeutically effective amounts of TG02, an immune checkpoint inhibitor, a COX-2 inhibitor, and a fourth therapeutic agent, wherein the fourth therapeutic agent is neither an immune checkpoint inhibitor nor a COX-2 inhibitor.


In another embodiment, the present disclosure provides personalized medicine for cancer patients, and encompasses the selection of treatment options with the highest likelihood of successful outcome for individual cancer patients. In another aspect, the disclosure relates to the use of an assay(s) to predict the treatment outcome, e.g., the likelihood of favorable responses or treatment success, in patients having cancer.


In another embodiment, the present disclosure provides methods of selecting a patient, e.g., a human subject for treatment of cancer with TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, comprising obtaining a biological sample, e.g., blood cells, from the patient, testing a biological sample from the patient for the presence of a biomarker, e.g., overexpression of MYC, overexpression of MCL1, or both, and selecting the patient for treatment if the biological sample contains that biomarker. In another embodiment, the methods further comprise administering a therapeutically effective amount of TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, to the patient if the biological sample contains the biomarker. Examples of cancer biomarkers are provided in Table 1. In another embodiment, the cancer is a solid tumor. In another embodiment, the cancer is a hematological malignancy. In another embodiment, the cancer is selected from the group consisting of hepatocellular carcinoma, glioblastoma, lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, and colorectal cancer.


In another embodiment, the present disclosure provides methods of predicting treatment outcomes in a patient having cancer, comprising obtaining a biological sample, from the patient, testing the biological sample from the patient for the presence of a biomarker, e.g., overexpression of MYC, overexpression of MCL1, or both, wherein the detection of the biomarker indicates the patient will respond favorably to administration of a therapeutically effective amount of TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor. Favorable responses include, but are not limited to, a decrease in tumor size and an increase in progression-free or overall survival.


In another embodiment, the present disclosure provides methods of treating cancer, comprising administering a therapeutically effective amount of TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, to a patient, e.g., a human subject, with cancer in whom the patient's cells contain a biomarker. In another embodiment, the patient is selected for treatment with TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, after the patient's cells have been determined to contain an overexpression of MYC. In another embodiment, the patient is selected for treatment with TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, after the patient's cells have been determined to contain an overexpression of MCL1. In another embodiment, the patient is selected for treatment with TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, after the patient's cells have been determined to contain an overexpression of MYC and an overexpression of MCL1.


In another embodiment, the method of treating a patient having cancer comprises obtaining a biological sample from the patient, determining whether the biological sample contains a biomarker, e.g., overexpression of MYC, overexpression of MCL1, or both, and administering to the patient a therapeutically effective amount of TG02 and, optionally, an immune checkpoint inhibitor and/or a COX-2 inhibitor, if the biological sample contains the biomarker. In another embodiment, the methods provided herein comprise determining whether the patient's cells contain an overexpression of MYC. In another embodiment, the methods provided herein comprise determining whether the patient's cells contain an overexpression of MCL1. In another embodiment, the methods provided herein comprise determining whether the patient's cells contain an overexpression of MYC and MCL1.


In another embodiment, the disclosure provides a method of treating a subject having cancer, the method comprising obtaining a biological sample from the subject, determining the expression level of MYC, MCL1, or both in the biological sample; and administering a therapeutically effective amount of TG02 and a second therapeutic agent, e.g., temozolomide, carfilzomib, sorafenib, bortezomib, doxorubicin, cisplatin, lenalidomide, dexamethasone, or Ara-C, to the subject if the biological sample shows overexpression of MYC, MCL1, or both.


In another embodiment, the patient has been treated previously with immune checkpoint inhibitor alone. For example, the previous immune checkpoint therapy may be an anti-PD-1 therapy.


In another embodiment, the patient has been treated previously with COX-2 inhibitor alone.


I. Immune checkpoint inhibitors

Immune checkpoint inhibitors are therapies that blockade immune system inhibitor checkpoints. Immune checkpoints can be stimulatory or inhibitory. Blockade of inhibitory immune checkpoint activates immune system function and can be used for cancer immunotherapy. Pardoll, Nature Reviews. Cancer 12:252-64 (2012). Tumor cells turn off activated T cells when they attach to specific T-cell receptors. Immune checkpoint inhibitors prevent tumor cells from attaching to T cells, which results in T cells remaining activated. In effect, the coordinated action by cellular and soluble components combats pathogens and injuries by cancers. The modulation of immune system pathways may involve changing the expression or the functional activity of at least one component of the pathway to then modulate the response by the immune system. U.S. 2015/0250853. Examples of immune checkpoint inhibitors include PD-1 inhibitors, PD-L1 inhibitors, CTLA-4 inhibitors, LAG3 inhibitors, TIM3 inhibitors, cd47 inhibitors, and B7-H1 inhibitors. Thus, in one embodiment, the immune checkpoint inhibitor is selected from the group consisting of a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, a LAG3 inhibitor, a TIM3 inhibitor, and a cd47 inhibitor.


In another embodiment, the immune checkpoint inhibitor is a programmed cell death (PD-1) inhibitor. PD-1 is a T-cell coinhibitory receptor that plays a pivotal role in the ability of tumor cells to evade the host's immune system. Blockage of interactions between PD-1 and PD-L1, a ligand of PD-1, enhances immune function and mediates antitumor activity. Examples of PD-1 inhibitors include antibodies that specifically bind to PD-1. Particular anti-PD-1 antibodies include, but are not limited to nivolumab, pembrolizumab, STI-1014, and pidilzumab. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies of anti-PD-1 antibodies, see U.S. 2013/0309250, U.S. Pat. Nos. 6,808,710, 7,595,048, 8,008,449, 8,728,474, 8,779,105, 8,952,136, 8,900,587, 9,073,994, 9,084,776, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a PD-L1 (also known as B7-H1 or CD274) inhibitor. Examples of PD-L1 inhibitors include antibodies that specifically bind to PD-L1. Particular anti-PD-L1 antibodies include, but are not limited to, avelumab, atezolizumab, durvalumab, and BMS-936559. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies, see U.S. Pat. No. 8,217,149, U.S. 2014/0341917, U.S. 2013/0071403, WO 2015036499, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a CTLA-4 inhibitor. CTLA-4, also known as cytotoxic T-lymphocyte antigen 4, is a protein receptor that downregulates the immune system. CTLA-4 is characterized as a “brake” that binds costimulatory molecules on antigen-presenting cells, which prevents interaction with CD28 on T cells and also generates an overtly inhibitory signal that constrains T cell activation. Examples of CTLA-4 inhibitors include antibodies that specifically bind to CTLA-4. Particular anti-CTLA-4 antibodies include, but are not limited to, ipilimumab and tremelimumab. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies, see U.S. Pat. Nos. 6,984,720, 6,207,156, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a LAG3 inhibitor. LAG3, Lymphocyte Activation Gene 3, is a negative co-simulatory receptor that modulates T cell homeostatis, proliferation, and activation. In addition, LAG3 has been reported to participate in regulatory T cells (Tregs) suppressive function. A large proportion of LAG3 molecules are retained in the cell close to the microtubule-organizing center, and only induced following antigen specific T cell activation. U.S. 2014/0286935. Examples of LAG3 inhibitors include antibodies that specifically bind to LAG3. Particular anti-LAG3 antibodies include, but are not limited to, GSK2831781. For a general discussion of the availability, methods of production, mechanism of action, and studies, see, U.S. 2011/0150892, U.S. 2014/0093511, U.S. 20150259420, and Huang et al., Immunity 21:503-13 (2004).


In another embodiment, the immune checkpoint inhibitor is a TIM3 inhibitor. TIM3, T-cell immunoglobulin and mucin domain 3, is an immune checkpoint receptor that functions to limit the duration and magnitude of TH1 and TC1 T-cell responses. The TIM3 pathway is considered a target for anticancer immunotherapy due to its expression on dysfunctional CD8+ T cells and Tregs, which are two reported immune cell populations that constitute immunosuppression in tumor tissue. Anderson, Cancer Immunology Research 2:393-98 (2014). Examples of TIM3 inhibitors include antibodies that specifically bind to TIM3. For a general discussion of the availability, methods of production, mechanism of action, and studies of TIM3 inhibitors, see U.S. 20150225457, U.S. 20130022623, U.S. Pat. No. 8,522,156, Ngiow et al., Cancer Res 71: 6567-71 (2011), Ngiow, et al., Cancer Res 71:3540-51 (2011), and Anderson, Cancer Immunology Res 2:393-98 (2014).


In another embodiment, the immune checkpoint inhibitor is a cd47 inhibitor. See Unanue, E. R., PNAS 110:10886-87 (2013).


The term “antibody” is meant to include intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity. In another embodiment, “antibody” is meant to include soluble receptors that do not possess the Fc portion of the antibody. In one embodiment, the antibodies are humanized monoclonal antibodies and fragments thereof made by means of recombinant genetic engineering.


Another class of immune checkpoint inhibitors include polypeptides that bind to and block PD-1 receptors on T-cells without triggering inhibitor signal transduction. Such peptides include B7-DC polypeptides, B7-H1 polypeptides, B7-1 polypeptides and B7-2 polypeptides, and soluble fragments thereof, as disclosed in U.S. Pat. No. 8,114,845.


Another class of immune checkpoint inhibitors include compounds with peptide moieties that inhibit PD-1 signaling. Examples of such compounds are disclosed in U.S. Pat. No. 8,907,053 and have the structure:




embedded image



or a pharmaceutically acceptable salt thereof, wherein the compound comprises at least 5 amino acids useful as therapeutic agents capable of inhibiting the PD-1 signaling pathway.


Another class of immune checkpoint inhibitors include inhibitors of certain metabolic enzymes, such as indoleamine 2,3 dioxygenase (IDO), which is expressed by infiltrating myeloid cells and tumor cells. The IDO enzyme inhibits immune responses by depleting amino acids that are necessary for anabolic functions in T cells or through the synthesis of particular natural ligands for cytosolic receptors that are able to alter lymphocyte functions. Pardoll, Nature Reviews. Cancer 12:252-64 (2012); Löb, Cancer Immunol Immunother 58:153-57 (2009). Particular IDO blocking agents include, but are not limited to levo-1-methyl typtophan (L-1MT) and 1-methyl-tryptophan (1MT). Qian et al., Cancer Res 69:5498-504 (2009); and Löb et al., Cancer Immunol Immunother 58:153-7 (2009).


In one embodiment, the immune checkpoint inhibitor is nivolumab, pembrolizumab, pidilizumab, STI-1110, avelumab, atezolizumab, durvalumab, STI-1014, ipilimumab, tremelimumab, GSK2831781, BMS-936559 or MED14736.


II. COX-2 Inhibitors

Cyclooxygenase-2 (COX-2) is an enzyme that promotes inflammation and plays a role in tumor progression. COX-2 inhibitors include non-selective inhibitors such as aspirin, ibuprofen, sulindac sulphone, sulindac sulphide, diclofenac, nabumetone, naproxen, indomethacine, and piroxicam, selective inhibitors such as celecoxib, rofecoxib, valdecoxib, ANS-398, Cay10404, SC-236, and DUP697, and preferential inhibitors such as meloxicam and nimesulide. Other COX-2 inhibitors include apricoxib, tilmacoxib, and cimicoxib. Any COX-2 inhibitor is contemplated for use in the therapeutic methods of this disclosure. See Sobolewski et al., “The Role of Cyclooxygenase-2 in Cell Proliferation and Cell Death in Human Malignancies,” International Journal of Cell Biology, vol. 2010, Article ID 215158, 21 pages, 2010. doi:10.1155/2010/215158.


In another embodiment, the COX-2 inhibitor is apricoxib. See Kirane et al., Clin. Cancer Res. 18:5031-5042 (2012).


In another embodiment, the COX-2 inhibitor is selected from the group consisting of:

  • 8-(ethyl-D5)-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid;
  • 6-chloro-8-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carb oxylic acid;
  • 6-bromo-8-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carb oxylic acid;
  • 8-chloro-6-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carb oxylic acid;
  • 6,8-dibromo-5,7-(dimethyl-D6)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid;
  • 8-(1-methylhexyl-D15)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid;
  • 6-chloro-8-(1-methylhexyl-D15)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid;
  • 8-(hexyl-D13)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid;
  • 7,8-(dimethyl-D6)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid; and
  • 6-chloro-8-(hexyl-D13)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid.


    See US 2015/0133538.


In another embodiment, the COX-2 inhibitor is 6-bromo-8-(methyl-D3)-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid.


III. Optional Therapeutic Agents

In certain therapeutic methods of the disclosure, a second therapeutic agent is administered to a cancer patient in combination with TG02, a third therapeutic agent is administered to a cancer patient in combination with TG02 and an immune checkpoint inhibitor or in combination with TG02 and a COX-2 inhibitor, or a fourth therapeutic agent is administered to a cancer patient in combination with TG02, an immune checkpoint inhibitor, and a COX-2 inhibitor. The second, third and fourth therapeutic agents used in the therapeutic methods of the present disclosure are referred to as “optional therapeutic agents.” Such optional therapeutic agents useful in the treatment of cancer patients are known in the art. In one embodiment, the optional therapeutic agent combined with TG02 is an anticancer agent that is neither an immune checkpoint inhibitor nor a COX-2 inhibitor.


Optional therapeutic agents are administered in an amount to provide their desired therapeutic effect. The effective dosage range for each optional therapeutic agent is known in the art, and the optional therapeutic agent is administered to an individual in need thereof within such established ranges.


TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent can be administered together as a single-unit dose or separately as multi-unit doses, and in any order, e.g., wherein TG02 is administered before the immune checkpoint inhibitor, COX-2 inhibitor, and/or the optional therapeutic agent, or vice versa. One or more doses of TG02, the immune checkpoint inhibitor, the COX-2 inhibitor and/or the optional therapeutic agent can be administered to the patient.


In one embodiment, the optional therapeutic agent is an epigenetic drug. As used herein, the term “epigenetic drug” refers to a therapeutic agent that targets an epigenetic regulator. Examples of epigenetic regulators include the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases. Histone deacetylase inhibitors include, but are not limited to, vorinostat.


In another embodiment, the optional therapeutic agent is a chemotherapeutic agent or other anti-proliferative agent that can be administered in combination with TG02, or a pharmaceutically acceptable salt thereof, to treat cancer. Examples of therapies and anticancer agents that can be used in combination with TG02, or a pharmaceutically acceptable salt thereof, include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, a biologic response modifier (e.g., an interferon, an interleukin, tumor necrosis factor (TNF), hyperthermia and cryotherapy, an agent to attenuate any adverse effect (e.g., an antiemetic), and any other approved chemotherapeutic drug.


Nonlimiting exemplary antiproliferative compounds include an aromatase inhibitor; an anti-estrogen; an anti-androgen; a gonadorelin agonist; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active agent; an alkylating agent, e.g., temozolomide; a retinoid, a carontenoid, or a tocopherol; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an antimetabolite; a platin compound; a methionine aminopeptidase inhibitor; a bisphosphonate; an antiproliferative antibody; a heparanase inhibitor; an inhibitor of Ras oncogenic isoforms; a telomerase inhibitor; a proteasome inhibitor; a compound used in the treatment of hematologic malignancies; a Flt-3 inhibitor; an Hsp90 inhibitor; a kinesin spindle protein inhibitor; a MEK inhibitor; an antitumor antibiotic; a nitrosourea; a compound targeting/decreasing protein or lipid kinase activity, a compound targeting/decreasing protein or lipid phosphatase activity, or any further anti-angiogenic compound.


Nonlimiting exemplary aromatase inhibitors include steroids, such as atamestane, exemestane, and formestane, and non-steroids, such as aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole, and letrozole.


Nonlimiting anti-estrogens include tamoxifen, fulvestrant, raloxifene, and raloxifene hydrochloride. Anti-androgens include, but are not limited to, bicalutamide. Gonadorelin agonists include, but are not limited to, abarelix, goserelin, and goserelin acetate.


Nonlimiting exemplary topoisomerase I inhibitors include topotecan, gimatecan, irinotecan, camptothecin and its analogues, 9-nitrocamptothecin, and the macromolecular camptothecin conjugate PNU-166148. Topoisomerase II inhibitors include, but are not limited to, anthracyclines, such as doxorubicin, daunorubicin, epirubicin, idarubicin, and nemorubicin; anthraquinones, such as mitoxantrone and losoxantrone; and podophillotoxines, such as etoposide and teniposide.


Microtubule active agents include microtubule stabilizing, microtubule destabilizing compounds, and microtubulin polymerization inhibitors including, but not limited to, taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine, vinblastine sulfate, vincristine, and vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof.


Nonlimiting exemplary alkylating agents include cyclophosphamide, ifosfamide, melphalan, and nitrosoureas, such as carmustine and lomustine.


Nonlimiting exemplary matrix metalloproteinase inhibitors (“MMP inhibitors”) include collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, batimastat, marimastat, prinomastat, metastat, BMS-279251, BAY 12-9566, TAA211, MMI270B, and AAJ996.


Nonlimiting exemplary mTOR inhibitors include compounds that inhibit the mammalian target of rapamycin (mTOR) and possess antiproliferative activity such as sirolimus, everolimus, CCI-779, and ABT578.


Nonlimiting exemplary antimetabolites include 5-fluorouracil (5-FU), capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists, such as pemetrexed.


Nonlimiting exemplary platin compounds include carboplatin, cis-platin, cisplatinum, and oxaliplatin.


Nonlimiting exemplary methionine aminopeptidase inhibitors include bengamide or a derivative thereof and PPI-2458.


Nonlimiting exemplary bisphosphonates include etridonic acid, clodronic acid, tiludronic acid, pamidronic acid, alendronic acid, ibandronic acid, risedronic acid, and zoledronic acid.


Nonlimiting exemplary heparanase inhibitors include compounds that target, decrease, or inhibit heparin sulfate degradation, such as PI-88 and OGT2115.


Nonlimiting exemplary compounds which target, decrease, or inhibit the oncogenic activity of Ras include farnesyl transferase inhibitors, such as L-744832, DK8G557, tipifarnib, and lonafarnib.


Nonlimiting exemplary telomerase inhibitors include compounds that target, decrease, or inhibit the activity of telomerase, such as compounds that inhibit the telomerase receptor, such as telomestatin.


Nonlimiting exemplary proteasome inhibitors include compounds that target, decrease, or inhibit the activity of the proteasome including, but not limited to, bortezomib. In some embodiments, the proteasome inhibitor is carfilzomib.


Nonlimiting exemplary FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R) include interferon, I-β-D-arabinofuransylcytosine (ara-c), and bisulfan; and ALK inhibitors, which are compounds which target, decrease, or inhibit anaplastic lymphoma kinase.


Nonlimiting exemplary Flt-3 inhibitors include PKC412, midostaurin, a staurosporine derivative, SU11248, and MLN518.


Nonlimiting exemplary HSP90 inhibitors include compounds targeting, decreasing, or inhibiting the intrinsic ATPase activity of HSP90; or degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins, or antibodies that inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.


Nonlimiting exemplary protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, include a) a compound targeting, decreasing, or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as a compound that targets, decreases, or inhibits the activity of PDGFR, such as an N-phenyl-2-pyrimidine-amine derivatives, such as imatinib, SU101, SU6668, and GFB-111; b) a compound targeting, decreasing, or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); c) a compound targeting, decreasing, or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as a compound that targets, decreases, or inhibits the activity of IGF-IR; d) a compound targeting, decreasing, or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) a compound targeting, decreasing, or inhibiting the activity of the Axl receptor tyrosine kinase family; f) a compound targeting, decreasing, or inhibiting the activity of the Ret receptor tyrosine kinase; g) a compound targeting, decreasing, or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) a compound targeting, decreasing, or inhibiting the activity of the c-Kit receptor tyrosine kinases, such as imatinib; i) a compound targeting, decreasing, or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. Bcr-Abl kinase) and mutants, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib; PD180970; AG957; NSC 680410; PD173955; or dasatinib; j) a compound targeting, decreasing, or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK1, PKB/Akt, and Ras/MAPK family members, and/or members of the cyclin-dependent kinase family (CDK), such as a staurosporine derivative disclosed in U.S. Pat. No. 5,093,330, such as midostaurin; examples of further compounds include UCN-01, safingol, BAY 43-9006, bryostatin 1, perifosine; ilmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; LY333531/LY379196; a isochinoline compound; a farnesyl transferase inhibitor; PD184352 or QAN697, or AT7519; k) a compound targeting, decreasing or inhibiting the activity of a protein-tyrosine kinase, such as imatinib mesylate or a tyrphostin, such as Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); 1) a compound targeting, decreasing, or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as CP 358774, ZD 1839, ZM 105180; trastuzumab, cetuximab, gefitinib, erlotinib, OSI-774, C1-1033, EKB-569, GW-2016, antibodies E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 and E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives; and m) a compound targeting, decreasing, or inhibiting the activity of the c-Met receptor.


Nonlimiting exemplary compounds that target, decrease, or inhibit the activity of a protein or lipid phosphatase include inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.


Further anti-angiogenic compounds include compounds having another mechanism for their activity unrelated to protein or lipid kinase inhibition, e.g., thalidomide and TNP-470.


Additional, nonlimiting, exemplary chemotherapeutic compounds, one or more of which may be used in combination with TG02, or a pharmaceutically acceptable salt thereof, include: avastin, daunorubicin, adriamycin, Ara-C, VP-16, teniposide, mitoxantrone, idarubicin, carboplatinum, PKC412, 6-mercaptopurine (6-MP), fludarabine phosphate, octreotide, SOM230, FTY720, 6-thioguanine, cladribine, 6-mercaptopurine, pentostatin, hydroxyurea, 2-hydroxy-1H-isoindole-1,3-dione derivatives, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate, angiostatin, endostatin, anthranilic acid amides, ZD4190, ZD6474, SU5416, SU6668, bevacizumab, rhuMAb, rhuFab, macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, RPI 4610, bevacizumab, porfimer sodium, anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortex olone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone, dexamethasone, fluocinolone, a plant alkaloid, a hormonal compound and/or antagonist, a biological response modifier, such as a lymphokine or interferon, an antisense oligonucleotide or oligonucleotide derivative, shRNA, and siRNA.


A number of suitable optional therapeutic, e.g., anticancer, agents are contemplated for use in the therapeutic methods provided herein. Indeed, the methods provided herein can include, but are not limited to, administration of numerous optional therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siRNA); polypeptides (e.g., enzymes and antibodies); biological mimetics (e.g., gossypol or BH3 mimetics); agents that bind (e.g., oligomerize or complex) with a Bcl-2 family protein such as Bax; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides; biological response modifiers (e.g., interferons (e.g., IFN-α) and interleukins (e.g., IL-2)); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid); gene therapy reagents (e.g., antisense therapy reagents and nucleotides); tumor vaccines; angiogenesis inhibitors; proteosome inhibitors: NF-κB modulators; anti-CDK compounds; HDAC inhibitors; and the like. Numerous other examples of optional therapeutic agents such as chemotherapeutic compounds and anticancer therapies suitable for co-administration with the disclosed compounds are known to those skilled in the art.


In certain embodiments, anticancer agents comprise agents that induce or stimulate apoptosis. Agents that induce or stimulate apoptosis include, for example, agents that interact with or modify DNA, such as by intercalating, cross-linking, alkylating, or otherwise damaging or chemically modifying DNA. Agents that induce apoptosis include, but are not limited to, radiation (e.g., X-rays, gamma rays, UV); tumor necrosis factor (TNF)-related factors (e.g., TNF family receptor proteins, TNF family ligands, TRAIL, antibodies to TRAIL-R1 or TRAIL-R2); kinase inhibitors (e.g., epidermal growth factor receptor (EGFR) kinase inhibitor. Additional anticancer agents include: vascular growth factor receptor (VGFR) kinase inhibitor, fibroblast growth factor receptor (FGFR) kinase inhibitor, platelet-derived growth factor receptor (PDGFR) kinase inhibitor, and Bcr-Abl kinase inhibitors (such as GLEEVEC)); antisense molecules; antibodies (e.g., HERCEPTIN, RITUXAN, ZEVALIN, and AVASTIN); anti-estrogens (e.g., raloxifene and tamoxifen); anti-androgens (e.g., flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids); cyclooxygenase 2 (COX-2) inhibitors (e.g., celecoxib, meloxicam, NS-398, and non-steroidal anti-inflammatory drugs (NSAIDs)); anti-inflammatory drugs (e.g., butazolidin, DECADRON, DELTASONE, dexamethasone, dexamethasone intensol, DEXONE, HEXADROL, hydroxychloroquine, METICORTEN, ORADEXON, ORASONE, oxyphenbutazone, PEDIAPRED, phenylbutazone, PLAQUENIL, prednisolone, prednisone, PRELONE, and TANDEARIL); and cancer chemotherapeutic drugs (e.g., irinotecan (CAMPTOSAR), CPT-11, fludarabine (FLUDARA), dacarbazine (DTIC), dexamethasone, mitoxantrone, MYLOTARG, VP-16, cisplatin, carboplatin, oxaliplatin, 5-FU, doxorubicin, gemcitabine, bortezomib, gefitinib, bevacizumab, TAXOTERE or TAXOL); cellular signaling molecules; ceramides and cytokines; staurosporine, and the like.


In still other embodiments, the therapeutic methods provided herein include administering to a cancer patient a therapeutically effective amount of TG02 and at least one additional anti-hyperproliferative or antineoplastic agent selected from alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).


Alkylating agents suitable for use in the present methods include, but are not limited to: 1) nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin); and chlorambucil); 2) ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa); 3) alkyl sulfonates (e.g., busulfan); 4) nitrosoureas (e.g., carmustine (BCNU); lomustine (CCNU); semustine (methyl-CCNU); and streptozocin (streptozotocin)); and 5) triazenes (e.g., dacarbazine (DTIC; dimethyltriazenoimid-azolecarboxamide).


In some embodiments, antimetabolites suitable for use in the present methods include, but are not limited to: 1) folic acid analogs (e.g., methotrexate (amethopterin)); 2) pyrimidine analogs (e.g., fluorouracil (5-fluorouracil; 5-FU), floxuridine (fluorode-oxyuridine; FudR), and cytarabine (cytosine arabinoside)); and 3) purine analogs (e.g., mercaptopurine (6-mercaptopurine; 6-MP), thioguanine (6-thioguanine; TG), and pentostatin (2′-deoxycoformycin)).


In still further embodiments, chemotherapeutic agents suitable for use in the methods of the present disclosure include, but are not limited to: 1) vinca alkaloids (e.g., vinblastine (VLB), vincristine); 2) epipodophyllotoxins (e.g., etoposide and teniposide); 3) antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin (daunomycin; rubidomycin), doxorubicin, bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin C)); 4) enzymes (e.g., L-asparaginase); 5) biological response modifiers (e.g., interferon-alfa); 6) platinum coordinating complexes (e.g., cisplatin (cis-DDP) and carboplatin); 7) anthracenediones (e.g., mitoxantrone); 8) substituted ureas (e.g., hydroxyurea); 9) methylhydrazine derivatives (e.g., procarbazine (N-methylhydrazine; MIH)); 10) adrenocortical suppressants (e.g., mitotane (o,p′-DDD) and aminoglutethimide); 11) adrenocorticosteroids (e.g., prednisone); 12) progestins (e.g., hydroxyprogesterone caproate, medroxyprogesterone acetate, and megestrol acetate); 13) estrogens (e.g., diethylstilbestrol and ethinyl estradiol); 14) antiestrogens (e.g., tamoxifen); 15) androgens (e.g., testosterone propionate and fluoxymesterone); 16) antiandrogens (e.g., flutamide): and 17) gonadotropin-releasing hormone analogs (e.g., leuprolide).


Any oncolytic agent that is routinely used in a cancer therapy context finds use in the therapeutic methods of the present disclosure. For example, the U.S. Food and Drug Administration (FDA) maintains a formulary of oncolytic agents approved for use in the United States. International counterpart agencies to the FDA maintain similar formularies. Those skilled in the art will appreciate that the “product labels” required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents.


Anticancer agents further include compounds which have been identified to have anticancer activity. Examples include, but are not limited to, 3-AP, 12-O-tetradecanoylphorbol-13-acetate, 17AAG, 852A, ABI-007, ABR-217620, ABT-751, ADI-PEG 20, AE-941, AG-013736, AGRO100, alanosine, AMG 706, antibody G250, antineoplastons, AP23573, apaziquone, APC8015, atiprimod, ATN-161, atrasenten, azacitidine, BB-10901, BCX-1777, bevacizumab, BG00001, bicalutamide, BMS 247550, bortezomib, bryostatin-1, buserelin, calcitriol, CCI-779, CDB-2914, cefixime, cetuximab, CG0070, cilengitide, clofarabine, combretastatin A4 phosphate, CP-675,206, CP-724,714, CpG 7909, curcumin, decitabine, DENSPM, doxercalciferol, E7070, E7389, ecteinascidin 743, efaproxiral, eflornithine, EKB-569, enzastaurin, erlotinib, exisulind, fenretinide, flavopiridol, fludarabine, flutamide, fotemustine, FR901228, G17DT, galiximab, gefitinib, genistein, glufosfamide, GTI-2040, histrelin, HKI-272, homoharringtonine, HSPPC-96, hu14.18-interleukin-2 fusion protein, HuMax-CD4, iloprost, imiquimod, infliximab, interleukin-12, IPI-504, irofulven, ixabepilone, lapatinib, lenalidomide, lestaurtinib, leuprolide, LMB-9 immunotoxin, lonafarnib, luniliximab, mafosfamide, MB07133, MDX-010, MLN2704, monoclonal antibody 3F8, monoclonal antibody J591, motexafin, MS-275, MVA-MUC1-IL2, nilutamide, nitrocamptothecin, nolatrexed dihydrochloride, nolvadex, NS-9, O6-benzylguanine, oblimersen sodium, ONYX-015, oregovomab, OSI-774, panitumumab, paraplatin, PD-0325901, pemetrexed, PHY906, pioglitazone, pirfenidone, pixantrone, PS-341, PSC 833, PXD101, pyrazoloacridine, R115777, RAD001, ranpirnase, rebeccamycin analogue, rhuAngiostatin protein, rhuMab 2C4, rosiglitazone, rubitecan, S-1, S-8184, satraplatin, SB-, 15992, SGN-0010, SGN-40, sorafenib, SR31747A, ST1571, SU011248, suberoylanilide hydroxamic acid, suramin, talabostat, talampanel, tariquidar, temsirolimus, TGFa-PE38 immunotoxin, thalidomide, thymalfasin, tipifarnib, tirapazamine, TLK286, trabectedin, trimetrexate glucuronate, TroVax, UCN-1, valproic acid, vinflunine, VNP40101M, volociximab, vorinostat, VX-680, ZD1839, ZD6474, zileuton, and zosuquidar trihydrochloride.


For a more detailed description of anticancer agents and other optional therapeutic agents, those skilled in the art are referred to any number of instructive manuals including, but not limited to, the Physician's Desk Reference and to Goodman and Gilman's “Pharmaceutical Basis of Therapeutics” tenth edition, Eds. Hardman et al., 2002.


In some embodiments, methods provided herein comprise administering TG02 to a cancer patient in combination with radiation therapy. The methods provided herein are not limited by the types, amounts, or delivery and administration systems used to deliver the therapeutic dose of radiation to a patient. For example, the patient may receive photon radiotherapy, particle beam radiation therapy, other types of radiotherapies, and combinations thereof. In some embodiments, the radiation is delivered to the patient using a linear accelerator. In still other embodiments, the radiation is delivered using a gamma knife.


The source of radiation can be external or internal to the patient. External radiation therapy is most common and involves directing a beam of high-energy radiation to a tumor site through the skin using, for instance, a linear accelerator. While the beam of radiation is localized to the tumor site, it is nearly impossible to avoid exposure of normal, healthy tissue. However, external radiation is usually well tolerated by patients. Internal radiation therapy involves implanting a radiation-emitting source, such as beads, wires, pellets, capsules, particles, and the like, inside the body at or near the tumor site including the use of delivery systems that specifically target cancer cells (e.g., using particles attached to cancer cell binding ligands). Such implants can be removed following treatment, or left in the body inactive. Types of internal radiation therapy include, but are not limited to, brachytherapy, interstitial irradiation, intracavity irradiation, radioimmunotherapy, and the like.


The patient may optionally receive radiosensitizers (e.g., metronidazole, misonidazole, intra-arterial Budr, intravenous iododeoxyuridine (IudR), nitroimidazole, 5-substituted-4-nitroimidazoles, 2H-isoindolediones, [[(2-bromoethyl)-amino]methyl]-nitro-1H-imidazole-1-ethanol, nitroaniline derivatives, DNA-affinic hypoxia selective cytotoxins, halogenated DNA ligand, 1,2,4 benzotriazine oxides, 2-nitroimidazole derivatives, fluorine-containing nitroazole derivatives, benzamide, nicotinamide, acridine-intercalator, 5-thiotretrazole derivative, 3-nitro-1,2,4-triazole, 4,5-dinitroimidazole derivative, hydroxylated texaphrins, cisplatin, mitomycin, tiripazamine, nitrosourea, mercaptopurine, methotrexate, fluorouracil, bleomycin, vincristine, carboplatin, epirubicin, doxorubicin, cyclophosphamide, vindesine, etoposide, paclitaxel, heat (hyperthermia), and the like), radioprotectors (e.g., cysteamine, aminoalkyl dihydrogen phosphorothioates, amifostine (WR 2721), IL-1, IL-6, and the like). Radiosensitizers enhance the killing of tumor cells. Radioprotectors protect healthy tissue from the harmful effects of radiation.


Any type of radiation can be administered to an patient, so long as the dose of radiation is tolerated by the patient without unacceptable negative side-effects. Suitable types of radiotherapy include, for example, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) or particle beam radiation therapy (e.g., high linear energy radiation). Ionizing radiation is defined as radiation comprising particles or photons that have sufficient energy to produce ionization, i.e., gain or loss of electrons (as described in, for example, U.S. Pat. No. 5,770,581 incorporated herein by reference in its entirety). The effects of radiation can be at least partially controlled by the clinician. In one embodiment, the dose of radiation is fractionated for maximal target cell exposure and reduced toxicity.


In one embodiment, the total dose of radiation administered to a patient is about 0.01 Gray (Gy) to about 100 Gy. In another embodiment, about 10 Gy to about 65 Gy (e.g., about 15 Gy, 20 Gy, 25 Gy, 30 Gy, 35 Gy, 40 Gy, 45 Gy, 50 Gy, 55 Gy, or 60 Gy) are administered over the course of treatment. While in some embodiments a complete dose of radiation can be administered over the course of one day, the total dose is ideally fractionated and administered over several days. Desirably, radiotherapy is administered over the course of at least about 3 days, e.g., at least 5, 7, 10, 14, 17, 21, 25, 28, 32, 35, 38, 42, 46, 52, or 56 days (about 1-8 weeks). Accordingly, a daily dose of radiation will comprise approximately 1-5 Gy (e.g., about 1 Gy, 1.5 Gy, 1.8 Gy, 2 Gy, 2.5 Gy, 2.8 Gy, 3 Gy, 3.2 Gy, 3.5 Gy, 3.8 Gy, 4 Gy, 4.2 Gy, or 4.5 Gy), or 1-2 Gy (e.g., 1.5-2 Gy). The daily dose of radiation should be sufficient to induce destruction of the targeted cells. If stretched over a period, in one embodiment, radiation is not administered every day, thereby allowing the animal to rest and the effects of the therapy to be realized. For example, radiation desirably is administered on 5 consecutive days, and not administered on 2 days, for each week of treatment, thereby allowing 2 days of rest per week. However, radiation can be administered 1 day/week, 2 days/week, 3 days/week, 4 days/week, 5 days/week, 6 days/week, or all 7 days/week, depending on the animal's responsiveness and any potential side effects. Radiation therapy can be initiated at any time in the therapeutic period. In one embodiment, radiation is initiated in week 1 or week 2, and is administered for the remaining duration of the therapeutic period. For example, radiation is administered in weeks 1-6 or in weeks 2-6 of a therapeutic period comprising 6 weeks for treating, for instance, a solid tumor. Alternatively, radiation is administered in weeks 1-5 or weeks 2-5 of a therapeutic period comprising 5 weeks. These exemplary radiotherapy administration schedules are not intended, however, to limit the methods provided herein.


IV. Therapeutic Methods

In the therapeutic methods provided herein, TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic, e.g., anticancer, agent may be administered to a cancer patient under one or more of the following conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc.


In some embodiments, TG02 is administered prior to the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks prior to the administration of the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent.


In some embodiments, TG02 is administered after the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks after the administration of the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent.


In some embodiments, TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent are administered concurrently but on different schedules, e.g., TG02 is administered daily while the immune checkpoint inhibitor is administered once a week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, TG02 is administered once a day while the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent is administered once a week, once every two weeks, once every three weeks, or once every four weeks.


The therapeutic methods provided herein comprise administering TG02 to a cancer patient in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typically, TG02 may be administered in an amount from about 1 mg/kg to about 500 mg/kg, about 1 mg/kg to about 100 mg/kg, or about 1 mg/kg to about 50 mg/kg. The dosage of a composition can be at any dosage including, but not limited to, 30-600 mg/day. Particular doses include 50, 100, 200, 250, 300, 400, 500, and 600 mg/day. In one embodiment, TG02 is administered once a day on 3-7 consecutive days prior to the administration of the immune checkpoint inhibitor. In another embodiment, 250 mg/day of TG02 is administered. In another embodiment, 250 mg/day of TG02 is administered twice weekly. In another embodiment, TG02 administration continues on the day of the immune checkpoint inhibitor and continues for additional days until disease progression or until TG02 administration is no longer beneficial. These dosages are exemplary of the average case, but there can be individual instances in which higher or lower dosages are merited, and such are within the scope of this disclosure. In practice, the physician determines the actual dosing regimen that is most suitable for an individual patient, which can vary with the age, weight, and response of the particular patient.


The unit oral dose of TG02 may comprise from about 0.01 to about 1000 mg, e.g., about 10 to about 500 mg of TG02. In one embodiment, the unit oral dose of TG02 is 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, 250 mg, 260 mg, 270 mg, 280 mg, 290 mg, or 300 mg. The unit dose may be administered one or more times daily, e.g., as one or more tablets or capsules.


In addition to administering TG02 as a raw chemical, it may be administered as part of a pharmaceutical preparation or composition. In some embodiments, the pharmaceutical preparation or composition can include one or more pharmaceutically acceptable carriers, excipients, and/or auxiliaries. In some embodiments, the one or more carriers, excipients, and auxiliaries facilitate processing of TG02 into a preparation or composition which can be used pharmaceutically. The preparations, particularly those preparations which can be administered orally or topically and which can be used for one type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by intravenous infusion, injection, topically or orally, contain from about 0.01 to 99 percent, in one embodiment from about 0.25 to 75 percent of active compound(s), together with the one or more carriers, excipients, and/or auxiliaries.


The pharmaceutical compositions of provided herein may be administered to any patient which may experience the beneficial effects of TG02. Foremost among such patients are mammals, e.g., humans, although the methods and compositions provided herein are not intended to be so limited. Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).


The pharmaceutical preparations provided herein are manufactured by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.


Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries can be suitable flow-regulating agents and lubricants. Suitable auxiliaries include, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, are used. Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.


Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are in one embodiment dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added.


Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.


Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.


The present disclosure encompasses the use of solvates of TG02. Solvates typically do not significantly alter the physiological activity or toxicity of a compound, and as such may function as pharmacological equivalents. The term “solvate” as used herein is a combination, physical association and/or solvation of TG02 with a solvent molecule such as, e.g. a disolvate, monosolvate or hemisolvate, where the ratio of solvent molecule to TG02 is about 2:1, about 1:1 or about 1:2, respectively. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate can be isolated, such as when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. Thus, “solvate” encompasses both solution-phase and isolatable solvates. TG02 can be present as solvated forms with a pharmaceutically acceptable solvent, such as water, methanol, ethanol, and the like, and it is intended that the disclosure includes both solvated and unsolvated forms of TG02. One type of solvate is a hydrate. A “hydrate” relates to a particular subgroup of solvates where the solvent molecule is water. Solvates typically can function as pharmacological equivalents. Preparation of solvates is known in the art. See, for example, M. Caira et al, J. Pharmaceut. Sci., 93(3):601-611 (2004), which describes the preparation of solvates of fluconazole with ethyl acetate and with water. Similar preparation of solvates, hemisolvates, hydrates, and the like are described by E. C. van Tonder et al., AAPS Pharm. Sci. Tech., 5(1):Article 12 (2004), and A. L. Bingham et al., Chem. Commun. 603-604 (2001). A typical, non-limiting, process of preparing a solvate involves dissolving TG02 in a desired solvent (organic, water, or a mixture thereof) at temperatures above 20° C. to about 25° C., then cooling the solution at a rate sufficient to form crystals, and isolating the crystals by known methods, e.g., filtration. Analytical techniques such as infrared spectroscopy can be used to confirm the presence of the solvent in a crystal of the solvate.


Therapeutically effective amounts of TG02 and/or the immune checkpoint inhibitor, and/or the COX-2 inhibitor, and/or the optional therapeutic agent formulated in accordance with standard pharmaceutical practices, are administered to a human patient in need thereof. Whether such a treatment is indicated depends on the individual case and is subject to medical assessment (diagnosis) that takes into consideration signs, symptoms, and/or malfunctions that are present, the risks of developing particular signs, symptoms and/or malfunctions, and other factors.


TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent can be administered by any suitable route, for example by oral, buccal, inhalation, sublingual, rectal, vaginal, intracisternal or intrathecal through lumbar puncture, transurethral, nasal, percutaneous, i.e., transdermal, or parenteral (including intravenous, intramuscular, subcutaneous, intracoronary, intradermal, intramammary, intraperitoneal, intraarticular, intrathecal, retrobulbar, intrapulmonary injection and/or surgical implantation at a particular site) administration. Parenteral administration can be accomplished using a needle and syringe or using a high pressure technique.


Pharmaceutical compositions include those wherein TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent are administered in an effective amount to achieve its intended purpose. The exact formulation, route of administration, and dosage is determined by an individual physician in view of the diagnosed condition or disease. Dosage amount and interval can be adjusted individually to provide levels of TG02, immune checkpoint inhibitor, COX-2 inhibitor, and/or optional therapeutic agent that is sufficient to maintain therapeutic effects.


Toxicity and therapeutic efficacy of TG02, the immune checkpoint inhibitor, the COX-2 inhibitor, and/or the optional therapeutic agent can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the maximum tolerated dose (MTD) of a compound, which defines as the highest dose that causes no toxicity in a patient. The dose ratio between the maximum tolerated dose and therapeutic effects (e.g. inhibiting of tumor growth) is the therapeutic index. The dosage can vary within this range depending upon the dosage form employed, and the route of administration utilized. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.


A therapeutically effective amount of TG02, immune checkpoint inhibitor, COX-2 inhibitor, and/or optional therapeutic agent required for use in therapy varies with the nature of the condition being treated, the length of time that activity is desired, and the age and the condition of the patient, and ultimately is determined by the attendant physician. For example, dosage amounts and intervals can be adjusted individually to provide plasma levels of TG02 and immune checkpoint inhibitor that are sufficient to maintain the desired therapeutic effects. The desired dose conveniently can be administered in a single dose, or as multiple doses administered at appropriate intervals, for example as one, two, three, four or more subdoses per day. Multiple doses often are desired, or required. For example, TG02 and immune checkpoint inhibitor can be administered at a frequency of: one dose per day; four doses delivered as one dose per day at four-day intervals (q4d×4); four doses delivered as one dose per day at three-day intervals (q3d×4); one dose delivered per day at five-day intervals (qd×5); one dose per week for three weeks (qwk3); five daily doses, with two days rest, and another five daily doses (5/2/5); or, any dose regimen determined to be appropriate for the circumstance.


The immune checkpoint inhibitor is administered in therapeutically effective amounts. When the immune checkpoint inhibitor is a monoclonal antibody, 1-20 mg/kg is administered as an intravenous infusion every 2-4 weeks. For example, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1000 mg, 1100 mg, 1200 mg, 1300 mg, 1400 mg, 1500 mg, 1600 mg, 1700 mg, 1800 mg, 1900 mg and 2000 mg of the antibody may be administered.


For example, when the immune checkpoint inhibitor is the anti-PD-1 antibody nivolumab, 3 mg/kg may be administered by intravenous infusion over 60 minutes every two weeks. When the immune checkpoint inhibitor is the anti-PD-1 antibody pembrolizumab, 2 mg/kg may be administered by intravenous infusion over 30 minutes every two or three weeks. When the immune checkpoint inhibitor is the anti-PD-L1 antibody avelumab, 10 mg/kg may be administered by intravenous infusion as frequently as every 2 weeks. Disis et al., J. Clin Oncol. 33 (2015) (suppl; abstr 5509). When the immune checkpoint inhibitor is the anti-PD-L1 antibody MPDL3280A, 20 mg/kg may be administered by intravenous infusion every 3 weeks. Herbst et al., Nature 515:563-80 (2014). When the immune checkpoint inhibitor is the anti-CTLA-4 antibody ipilumumab, 3 mg/kg may be administered by intravenous infusion over 90 minutes every 3 weeks. When the immune checkpoint inhibitor is the anti-CTLA-4 antibody tremelimumab, 15 mg/kg may be administered by intravenous infusion every 12 weeks. Naido et al., British Journal of Cancer 111:2214-19 (2014); Drugs R D, 10:123-32 (2010). When the immune checkpoint inhibitor is the anti-LAG3 antibody GSK2831781, 1.5 to 5 mg/kg may be administered by intravenous infusion over 120 minutes every 2-4 weeks. When the immune checkpoint inhibitor is an anti-TIM3 antibody, 1-5 mg/kg may be administered by intravenous infusion over 30-90 minutes every 2-4 weeks. When an inhibitor of indoleamine 2,3-dioxygenase (IDO) pathway is inhibitor indoximod in combination with temozolomide, 18.5 mg/kg/dose BID with an escalation to 27.7 mg/kg/dose BID of indoximod with 200 mg/m2 every 5 days of temozolomide.


The COX-2 inhibitor is also administered in therapeutically effective amounts, e.g., from about 1 mg/kg to about 500 mg/kg, about 1 mg/kg to about 100 mg/kg, or about 1 mg/kg to about 50 mg/kg. The unit oral dose of the COX-2 inhibitor may comprise from about 0.01 to about 1000 mg, e.g., about 1 to about 250 mg of the COX-2 inhibitor. In one embodiment, the unit oral dose of the COX-2 inhibitor is 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, or 250 mg. The unit dose may be administered one or more times daily, e.g., as one or more tablets or capsules.


In one embodiment, the immune checkpoint inhibitor is an antibody and 1-20 mg/kg is administered by intravenous infusion every 2-4 weeks. In another embodiment, 50-2000 mg of the antibody is administered by intravenous infusion every 2-4 weeks. In another embodiment, TG02 is administered prior to administration of the antibody. In another embodiment, TG02 is administered 3-7 days prior to the day of administration of the antibody. In another embodiment, TG02 is also administered the day the antibody is administered and on consecutive days thereafter until disease progression or until TG02 administration is no longer beneficial.


In one embodiment, the cancer patient has tumors with a biomarker, e.g., overexpression of MYC and/or MCL1, and receives 2 mg/kg pembrolizumab administered by intravenous infusion every three weeks and 30-600 mg of TG02 administered for 3-7 days prior to pembrolizumab administration, on the day of pembrolizumab administration, and thereafter until disease progression or until there is no therapeutic benefit.


In another embodiment, the cancer patient has tumors with a biomarker, e.g., overexpression of MYC and/or MCL1, and receives 3 mg/kg nivolumab administered by intravenous infusion every 2 weeks and 30-600 mg TG02 administered orally for 3-7 days prior to nivolumab administration, on the day of nivolumab administration, and thereafter until disease progression or until there is no therapeutic benefit.


In another embodiment, the cancer patient has tumors with a biomarker, e.g., overexpression of MYC and/or MCL1, and receives 3 mg/kg nivolumab administered by intravenous infusion every 2 weeks and 30-600 mg TG02 administered orally twice weekly prior to nivolumab administration, on the day of nivolumab administration, and thereafter until disease progression or until there is no therapeutic benefit.


In another embodiment, the treatment of the cancer patient with an immune checkpoint inhibitor and TG02 induces anti-proliferative response faster than when the immune checkpoint inhibitor is administered alone.


In another embodiment, the treatment of the cancer patient with a COX-2 inhibitor and TG02 induces anti-proliferative response faster than when the COX-2 inhibitor is administered alone.


V. Biomarkers

The term “biomarker” as used herein refers to any biological compound, such as a gene, a protein, a fragment of a protein, a peptide, a polypeptide, a nucleic acid, etc., that can be detected and/or quantified in a cancer patient in vivo or in a biological sample obtained from a cancer patient. A biomarker can be the entire intact molecule, or it can be a portion or fragment thereof. In one embodiment, the expression level of the biomarker is measured. The expression level of the biomarker can be measured, for example, by detecting the protein or RNA, e.g., mRNA, level of the biomarker. In some embodiments, portions or fragments of biomarkers can be detected or measured, for example, by an antibody or other specific binding agent. In some embodiments, a measurable aspect of the biomarker is associated with a given state of the patient, such as a particular stage of cancer. For biomarkers that are detected at the protein or RNA level, such measurable aspects may include, for example, the presence, absence, or concentration, i.e., expression level, of the biomarker in a cancer patient, or biological sample obtained from the cancer patient. For biomarkers that are detected at the nucleic acid level, such measurable aspects may include, for example, allelic versions of the biomarker or type, rate, and/or degree of mutation of the biomarker, also referred to herein as mutation status.


For biomarkers that are detected based on expression level of protein or RNA, expression level measured between different phenotypic statuses can be considered different, for example, if the mean or median expression level of the biomarker in the different groups is calculated to be statistically significant. Common tests for statistical significance include, among others, t-test, ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney, Significance Analysis of Microarrays, odds ratio, etc. Biomarkers, alone or in combination, provide measures of relative likelihood that a subject belongs to one phenotypic status or another. Therefore, they are useful, inter alia, as markers for disease and as indicators that particular therapeutic treatment regimens will likely result in beneficial patient outcomes.


Biomarkers include, but are not limited, the genes listed in Table 1. In one embodiment, the measurable aspect of the biomarker is its expression status. In one embodiment, the measurable aspect of the biomarker is its mutation status.











TABLE 1





Gene
Gene synonym
Gene description







A2M
CPAMD5, FWP007, S863-7
Alpha-2-macroglobulin


ABCB1
ABC20, CD243, CLCS,
ATP-binding cassette, sub-family B



GP170, MDR1, P-gp, PGY1
(MDR/TAP), member 1


ABCC1
GS-X, MRP, MRP1
ATP-binding cassette, sub-family C




(CFTR/MRP), member 1


ABCC2
CMOAT, cMRP, DJS,
ATP-binding cassette, sub-family C



MRP2
(CFTR/MRP), member 2


ABCC3
cMOAT2, EST90757,
ATP-binding cassette, sub-family C



MLP2, MOAT-D, MRP3
(CFTR/MRP), member 3


ABCC5
EST277145, MOAT-C,
ATP-binding cassette, sub-family C



MRP5, SMRP
(CFTR/MRP), member 5


ABCC6
ARA, EST349056, MLP1,
ATP-binding cassette, sub-family C



MRP6, PXE, URG7
(CFTR/MRP), member 6


ABCG2
ABCP, BCRP, CD338,
ATP-binding cassette, sub-family G



EST157481, MXR
(WHITE), member 2 (Junior blood




group)


ABL1
ABL, c-ABL, JTK7, p150
ABL proto-oncogene 1, non-receptor




tyrosine kinase


ABL2
ABLL, ARG
ABL proto-oncogene 2, non-receptor




tyrosine kinase


ACAP1
CENTB1, KIAA0050
ArfGAP with coiled-coil, ankyrin




repeat and PH domains 1


ACLY
ACL, ATPCL, CLATP
ATP citrate lyase


ACPP
ACP-3, ACP3
Acid phosphatase, prostate


ACVR1B
ActRIB, ACVRLK4, ALK4,
Activin A receptor, type IB



SKR2


ACVR2A
ACTRII, ACVR2
Activin A receptor, type IIA


ACVR2B
ActR-IIB
Activin A receptor, type IIB


ADAM9
CORD9, KIAA0021,
ADAM metallopeptidase domain 9



MCMP, MDC9, Mltng


ADAMTS1
C3-C5, KIAA1346, METH1
ADAM metallopeptidase with




thrombospondin type 1 motif, 1


ADAMTS14

ADAM metallopeptidase with




thrombospondin type 1 motif, 14


ADAMTS18
ADAMTS21
ADAM metallopeptidase with




thrombospondin type 1 motif, 18


ADAMTS20
GON-1
ADAM metallopeptidase with




thrombospondin type 1 motif, 20


ADAMTS3
ADAMTS-4, KIAA0366
ADAM metallopeptidase with




thrombospondin type 1 motif, 3


ADAMTS4
ADAMTS-2, ADMP-1,
ADAM metallopeptidase with



KIAA0688
thrombospondin type 1 motif, 4


ADAMTS5
ADAMTS11, ADMP-2
ADAM metallopeptidase with




thrombospondin type 1 motif, 5


ADAMTS6
ADAM-TS6
ADAM metallopeptidase with




thrombospondin type 1 motif, 6


ADAMTS8
ADAM-TS8, FLJ41712,
ADAM metallopeptidase with



METH2
thrombospondin type 1 motif, 8


ADAMTS9
KIAA1312
ADAM metallopeptidase with




thrombospondin type 1 motif, 9


ADM
AM
Adrenomedullin


ADRA1B

Adrenoceptor alpha 1B


AFP
FETA, HPAFP
Alpha-fetoprotein


AGER
RAGE
Advanced glycosylation end product-




specific receptor


AHR
bHLHe76
Aryl hydrocarbon receptor


AHSG
A2HS, FETUA, HSGA
Alpha-2-HS-glycoprotein


AKAP12
AKAP250, SSeCKS
A kinase (PRKA) anchor protein 12


AKR1B1
ALDR1, AR
Aldo-keto reductase family 1, member




B1 (aldose reductase)


AKT1
AKT, PKB, PRKBA, RAC
V-akt murine thymoma viral oncogene




homolog 1


AKT2

V-akt murine thymoma viral oncogene




homolog 2


AKT3
PKBG, PRKBG, RAC-
V-akt murine thymoma viral oncogene



gamma
homolog 3


ALB

Albumin


ALCAM
CD166, MEMD
Activated leukocyte cell adhesion




molecule


ALDOA

Aldolase A, fructose-bisphosphate


ALDOB

Aldolase B, fructose-bisphosphate


ALDOC

Aldolase C, fructose-bisphosphate


ALPL
HOPS, TNSALP
Alkaline phosphatase,




liver/bone/kidney


ALPP

Alkaline phosphatase, placental


ANG
RNASE5
Angiogenin, ribonuclease, RNase A




family, 5


ANGPT1
Ang1, KIAA0003
Angiopoietin 1


ANGPT2
Ang2
Angiopoietin 2


ANXA1
ANX1, LPC1
Annexin A1


ANXA11
ANX11
Annexin A11


ANXA2
ANX2, ANX2L4, CAL1H,
Annexin A2



LIP2, LPC2D


ANXA4
ANX4
Annexin A4


ANXA7
ANX7
Annexin A7


AOC3
HPAO, VAP-1, VAP1
Amine oxidase, copper containing 3


AP2B1
ADTB2, CLAPB1
Adaptor-related protein complex 2,




beta 1 subunit


APAF1
APAF-1, CED4
Apoptotic peptidase activating factor 1


APEX1
APE, APE-1, APEN, APEX,
APEX nuclease (multifunctional DNA



APX, HAP1, REF-1, REF1
repair enzyme) 1


APOA1

Apolipoprotein A-I


APOA2

Apolipoprotein A-II


APOC1

Apolipoprotein C-I


APOC3

Apolipoprotein C-III


APOD

Apolipoprotein D


APOE
AD2
Apolipoprotein E


APPBP2
Hs.84084, KIAA0228,
Amyloid beta precursor protein



PAT1
(cytoplasmic tail) binding protein 2


AR
AIS, DHTR, HUMARA,
Androgen receptor



NR3C4, SBMA, SMAX1


AREG
AREGB, SDGF
Amphiregulin


ARG2

Arginase 2


ARNT
bHLHe2, HIF-1beta
Aryl hydrocarbon receptor nuclear




translocator


ASPH
BAH, CASQ2BP1, HAAH,
Aspartate beta-hydroxylase



JCTN


ATM
ATA, ATC, ATD, ATDC,
ATM serine/threonine kinase



TEL1, TELO1


ATOH1
bHLHa14, HATH1, MATH-
Atonal homolog 1 (Drosophila)



1, Math1


ATP7B
WND
ATPase, Cu++ transporting, beta




polypeptide


AURKA
AIK, ARK1, AurA, BTAK,
Aurora kinase A



PPP1R47, STK15, STK6,



STK7


AURKB
Aik2, AIM-1, ARK2, AurB,
Aurora kinase B



IPL1, PPP1R48, STK12,



STK5


AZGP1
ZA2G, ZAG
Alpha-2-glycoprotein 1, zinc-binding


B2M

Beta-2-microglobulin


BAD
BBC2, BCL2L8
BCL2-associated agonist of cell death


BAG1

BCL2-associated athanogene


BAI1

Brain-specific angiogenesis inhibitor 1


BAX
BCL2L4
BCL2-associated X protein


BCL11A
BCL11A-L, BCL11A-S,
B-cell CLL/lymphoma 11A (zinc



BCL11A-XL, CTIP1, EVI9,
finger protein)



HBFQTL5, ZNF856


BCL2
Bcl-2, PPP1R50
B-cell CLL/lymphoma 2


BCL2A1
ACC-1, ACC-2, BCL2L5,
BCL2-related protein A1



BFL1, GRS, HBPA1


BCL2L1
Bcl-X, bcl-xL, bcl-xS,
BCL2-like 1



BCL2L, BCLX, PPP1R52


BCL2L2
BCL-W, KIAA0271,
BCL2-like 2



PPP1R51


BCL2L2-

BCL2L2-PABPN1 readthrough


PABPN1


BCL3
BCL4, D19S37
B-cell CLL/lymphoma 3


BCL6
BCL5, BCL6A, LAZ3,
B-cell CLL/lymphoma 6



ZBTB27, ZNF51


BDNF

Brain-derived neurotrophic factor


BIRC2
API1, c-IAP1, cIAP1, hiap-
Baculoviral IAP repeat containing 2



2, MIHB, RNF48


BIRC3
API2, c-IAP2, cIAP2, hiap-
Baculoviral IAP repeat containing 3



1, MALT2, MIHC, RNF49


BIRC5
API4, EPR-1, survivin
Baculoviral IAP repeat containing 5


BIRC6
BRUCE
Baculoviral IAP repeat containing 6


BLK
MGC10442
BLK proto-oncogene, Src family




tyrosine kinase


BLMH
BH
Bleomycin hydrolase


BMI1
PCGF4, RNF51
BMI1 proto-oncogene, polycomb ring




finger


BMP2
BMP2A
Bone morphogenetic protein 2


BMP4
BMP2B
Bone morphogenetic protein 4


BNIP3
Nip3
BCL2/adenovirus E1B 19 kDa




interacting protein 3


BNIP3L
BNIP3a, Nix
BCL2/adenovirus E1B 19 kDa




interacting protein 3-like


BRCA1
BRCC1, PPP1R53, RNF53
Breast cancer 1, early onset


BRCA2
BRCC2, FACD, FAD,
Breast cancer 2, early onset



FAD1, FANCD, FANCD1


BRMS1
DKFZP564A063
Breast cancer metastasis suppressor 1


BTG2
MGC126063, MGC126064,
BTG family, member 2



PC3, TIS21


C18orf8
HsT2591, MIC-1, MIC1
Chromosome 18 open reading frame 8


C1QBP
gC1Q-R, gC1qR, HABP1,
Complement component 1, q



p32, SF2p32
subcomponent binding protein


C6

Complement component 6


C7

Complement component 7


CA8
CALS, CARP
Carbonic anhydrase VIII


CALCA
CALC1
Calcitonin-related polypeptide alpha


CALM1
CALML2, CAMI, DD132,
Calmodulin 1 (phosphorylase kinase,



PHKD
delta)


CALM2
CAMII, PHKD
Calmodulin 2 (phosphorylase kinase,




delta)


CALM3
PHKD
Calmodulin 3 (phosphorylase kinase,




delta)


CALR
cC1qR, CRT, FLJ26680,
Calreticulin



RO, SSA


CANX
CNX, IP90, P90
Calnexin


CAPN6
CalpM, CANPX, CAPNX
Calpain 6


CASC3
BTZ, MLN51
Cancer susceptibility candidate 3


CASP1
ICE, IL1BC
Caspase 1, apoptosis-related cysteine




peptidase


CASP10
MCH4
Caspase 10, apoptosis-related cysteine




peptidase


CASP2
ICH1, MGC2181, NEDD2,
Caspase 2, apoptosis-related cysteine



PPP1R57
peptidase


CASP3
apopain, CPP32, CPP32B,
Caspase 3, apoptosis-related cysteine



Yama
peptidase


CASP4
ICE(rel)II, ICH-2, TX
Caspase 4, apoptosis-related cysteine




peptidase


CASP5
ICE(rel)III
Caspase 5, apoptosis-related cysteine




peptidase


CASP6
MCH2
Caspase 6, apoptosis-related cysteine




peptidase


CASP7
CMH-1, ICE-LAP3, MCH3
Caspase 7, apoptosis-related cysteine




peptidase


CASP8
Casp-8, FLICE, MACH,
Caspase 8, apoptosis-related cysteine



MCH5
peptidase


CASP9
APAF-3, ICE-LAP6,
Caspase 9, apoptosis-related cysteine



MCH6, PPP1R56
peptidase


CAT

Catalase


CAV1
CAV
Caveolin 1, caveolae protein, 22 kDa


CBL
c-Cbl, CBL2, RNF55
Cbl proto-oncogene, E3 ubiquitin




protein ligase


CCKBR

Cholecystokinin B receptor


CCL11
eotaxin, MGC22554,
Chemokine (C-C motif) ligand 11



SCYA11


CCL13
CKb10, MCP-4,
Chemokine (C-C motif) ligand 13



MGC17134, NCC-1,



SCYA13, SCYL1


CCL14
CKb1, HCC-1, HCC-3,
Chemokine (C-C motif) ligand 14



MCIF, NCC-2, SCYA14,



SCYL2


CCL16
CKb12, HCC-4, LCC-1,
Chemokine (C-C motif) ligand 16



LEC, LMC, Mtn-1, NCC-4,



SCYA16, SCYL4


CCL18
AMAC-1, CKb7, DC-CK1,
Chemokine (C-C motif) ligand 18



DCCK1, MIP-4, PARC,
(pulmonary and activation-regulated)



SCYA18


CCL19
CKb11, ELC, exodus-3,
Chemokine (C-C motif) ligand 19



MIP-3b, SCYA19


CCL2
GDCF-2, HC11, MCAF,
Chemokine (C-C motif) ligand 2



MCP-1, MCP1, MGC9434,



SCYA2, SMC-CF


CCL21
6Ckine, CKb9, ECL,
Chemokine (C-C motif) ligand 21



exodus-2, SCYA21, SLC,



TCA4


CCL23
Ckb-8, CKb8, MIP-3,
Chemokine (C-C motif) ligand 23



MPIF-1, SCYA23


CCL3
G0S19-1, LD78ALPHA,
Chemokine (C-C motif) ligand 3



MIP-1-alpha, SCYA3


CCL4
Act-2, AT744.1, LAG1,
Chemokine (C-C motif) ligand 4



MIP-1-beta, SCYA4


CCL5
D17S136E, MGC17164,
Chemokine (C-C motif) ligand 5



RANTES, SCYA5, SISd,



TCP228


CCL7
FIC, MARC, MCP-3,
Chemokine (C-C motif) ligand 7



MCP3, NC28, SCYA6,



SCYA7


CCL8
HC14, MCP-2, SCYA8
Chemokine (C-C motif) ligand 8


CCNA1
CT146
Cyclin A1


CCNA2
CCN1, CCNA
Cyclin A2


CCNB1
CCNB
Cyclin B1


CCNB2
HsT17299
Cyclin B2


CCND1
BCL1, D11S287E, PRAD1,
Cyclin D1



U21B31


CCND2

Cyclin D2


CCNE1
CCNE
Cyclin E1


CCNE2
CYCE2
Cyclin E2


CCNG1
CCNG
Cyclin G1


CCNG2

Cyclin G2


CCNH
CycH, p34, p37
Cyclin H


CCR10
GPR2
Chemokine (C-C motif) receptor 10


CCR7
BLR2, CD197, CDw197,
Chemokine (C-C motif) receptor 7



CMKBR7, EBI1


CD14

CD14 molecule


CD27
S152, TNFRSF7, Tp55
CD27 molecule


CD36
FAT, GP3B, GP4, GPIV,
CD36 molecule (thrombospondin



SCARB3
receptor)


CD38

CD38 molecule


CD40
Bp50, p50, TNFRSF5
CD40 molecule, TNF receptor




superfamily member 5


CD40LG
CD154, CD40L, gp39,
CD40 ligand



hCD40L, HIGM1, IMD3,



TNFSF5, TRAP


CD44
CD44R, CSPG8, HCELL,
CD44 molecule (Indian blood group)



IN, MC56, MDU2, MDU3,



MIC4, Pgp1


CD46
MCP, MGC26544, MIC10,
CD46 molecule, complement



TLX, TRA2.10
regulatory protein


CD52
CDW52
CD52 molecule


CD59
16.3A5, EJ16, EJ30, EL32,
CD59 molecule, complement



G344, MIC11, MIN1,
regulatory protein



MIN2, MIN3, MSK21, p18-



20


CD70
CD27L, CD27LG, TNFSF7
CD70 molecule


CD74
DHLAG
CD74 molecule, major




histocompatibility complex,




class II invariant chain


CD82
IA4, KAI1, R2, ST6,
CD82 molecule



TSPAN27


CD9
BA2, MIC3, MRP-1, P24,
CD9 molecule



TSPAN29


CDC16
ANAPC6, APC6, CUT9
Cell division cycle 16


CDC20
CDC20A, p55CDC
Cell division cycle 20


CDC25A

Cell division cycle 25A


CDC25B

Cell division cycle 25B


CDC25C
CDC25, PPP1R60
Cell division cycle 25C


CDC34
E2-CDC34, UBC3,
Cell division cycle 34



UBE2R1


CDC37
P50CDC37
Cell division cycle 37


CDC6
CDC18L
Cell division cycle 6


CDH1
CD324, UVO, uvomorulin
Cadherin 1, type 1, E-cadherin




(epithelial)


CDH17
cadherin, HPT-1
Cadherin 17, LI cadherin (liver-intestine)


CDH5
7B4, CD144
Cadherin 5, type 2 (vascular




endothelium)


CDK1
CDC2, CDC28A
Cyclin-dependent kinase 1


CDK2

Cyclin-dependent kinase 2


CDK4
PSK-J3
Cyclin-dependent kinase 4


CDK6
PLSTIRE
Cyclin-dependent kinase 6


CDK7
CAR, CAK1, CDKN7,
Cyclin-dependent kinase 7



MO15, STK1


CDKN1A
CAP20, CDKN1, CIP1,
Cyclin-dependent kinase inhibitor 1A



P21, p21CIP1,
(p21, Cip1)



p21Cip1/Waf1, SDI1,



WAF1


CDKN1C
BWCR, BWS, KIP2, P57
Cyclin-dependent kinase inhibitor 1C




(p57, Kip2)


CDKN2A
ARF, CDK4I, CDKN2,
Cyclin-dependent kinase inhibitor 2A



CMM2, INK4, INK4a,



MLM, MTS1, p14,



p14ARF, p16, p16INK4a,



p19, p19Arf


CEACAM5
CD66e, CEA
Carcinoembryonic antigen-related cell




adhesion molecule 5


CEACAM6
CD66c, NCA
Carcinoembryonic antigen-related cell




adhesion molecule 6 (non-specific




cross reacting antigen)


CENPF
hcp-1
Centromere protein F, 350/400 kDa


CFHR1
CFHL, CFHL1, CFHL1P,
Complement factor H-related 1



CFHR1P, FHR1, H36-1,



H36-2, HFL1, HFL2


CFLAR
c-FLIP, CASH, CASP8AP1,
CASP8 and FADD-like apoptosis



Casper, CLARP, FLAME,
regulator



FLIP, I-FLICE, MRIT


CFTR
ABC35, ABCC7, CF,
Cystic fibrosis transmembrane



CFTR/MRP, dJ760C5.1,
conductance regulator (ATP-binding



MRP7, TNR-CFTR
cassette sub-family C, member 7)


CGA
FSHA, GPHa, GPHA1,
Glycoprotein hormones, alpha



HCG, LHA, TSHA
polypeptide


CGB
CGB3
Chorionic gonadotropin, beta




polypeptide


CGB5
HCG
Chorionic gonadotropin, beta




polypeptide 5


CGB7
CG-beta-a
Chorionic gonadotropin, beta




polypeptide 7


CGB8

Chorionic gonadotropin, beta




polypeptide 8


CHD7
CRG, FLJ20357, FLJ20361,
Chromodomain helicase DNA binding



KIAA1416
protein 7


CHEK1
CHK1
Checkpoint kinase 1


CHEK2
bA444G7, CDS1, CHK2,
Checkpoint kinase 2



HuCds1, PP1425, RAD53


CHFR
FLJ10796, RNF196
Checkpoint with forkhead and ring




finger domains, E3 ubiquitin protein




ligase


CHGA

Chromogranin A (parathyroid secretory




protein 1)


CHI3L1
GP39, YKL40
Chitinase 3-like 1 (cartilage




glycoprotein-39)


CHP2

Calcineurin-like EF-hand protein 2


CIB2
DFNB48, KIP2, USH1J
Calcium and integrin binding family




member 2


CKB
CKBB
Creatine kinase, brain


CKS1B
CKS1, ckshs1
CDC28 protein kinase regulatory




subunit 1B


CKS2

CDC28 protein kinase regulatory




subunit 2


CLDN3
C7orf1, CPE-R2, CPETR2,
Claudin 3



HRVP1, RVP1


CLDN4
CPE-R, CPETR, CPETR1,
Claudin 4



hCPE-R, WBSCR8


CLDN7
CEPTRL2, CPETRL2,
Claudin 7



Hs.84359


CLEC3B
TN, TNA
C-type lectin domain family 3, member




B


CLIC1
NCC27, p64CLCP
Chloride intracellular channel 1


CLIP1
CLIP, CLIP-170, CLIP170,
CAP-GLY domain containing linker



CYLN1, RSN
protein 1


CLSTN1
CDHR12, CSTN1,
Calsyntenin 1



KIAA0911


CLU
APOJ, CLI, CLU1, CLU2,
Clusterin



KUB1, SGP-2, SP-40,



TRPM-2


CNN1
Sm-Calp, SMCC
Calponin 1, basic, smooth muscle


CNTF
HCNTF
Ciliary neurotrophic factor


COL11A1
CO11A1, COLL6, STL2
Collagen, type XI, alpha 1


COL17A1
BP180, BPAG2
Collagen, type XVII, alpha 1


COL18A1
KNO, KNO1, KS
Collagen, type XVIII, alpha 1


COL1A1
OI4
Collagen, type I, alpha 1


COL1A2
OI4
Collagen, type I, alpha 2


COL4A2
DKFZp686I14213,
Collagen, type IV, alpha 2



FLJ22259


COL4A3

Collagen, type IV, alpha 3




(Goodpasture antigen)


COL4A4
CA44
Collagen, type IV, alpha 4


COL4A5
ASLN, ATS
Collagen, type IV, alpha 5


COL6A1

Collagen, type VI, alpha 1


COX17

COX17 cytochrome c oxidase copper




chaperone


CP

Ceruloplasmin (ferroxidase)


CRABP1
CRABP, CRABP-I,
Cellular retinoic acid binding protein 1



CRABPI, RBP5


CRADD
RAIDD
CASP2 and RIPK1 domain containing




adaptor with death domain


CREBBP
CBP, KAT3A, RSTS, RTS
CREB binding protein


CRP
PTX1
C-reactive protein, pentraxin-related


CRYAB
CRYA2, HSPB5
Crystallin, alpha B


CSE1L
CAS, CSE1, XPO2
CSE1 chromosome segregation 1-like




(yeast)


CSF1
M-CSF, MCSF, MGC31930
Colony stimulating factor 1




(macrophage)


CSF1R
C-FMS, CD115, CSFR,
Colony stimulating factor 1 receptor



FMS


CSF2
GM-CSF, GMCSF
Colony stimulating factor 2




(granulocyte-macrophage)


CSF2RA
CD116, CSF2R
Colony stimulating factor 2 receptor,




alpha, low-affinity (granulocyte-




macrophage)


CSF3
C17orf33, G-CSF, GCSF,
Colony stimulating factor 3



MGC45931
(granulocyte)


CSN1S1
CASA, CSN1
Casein alpha s1


CSNK1E
CKIE, CKIepsilon, HCKIE
Casein kinase 1, epsilon


CSNK2A1

Casein kinase 2, alpha 1 polypeptide


CSNK2A2
CSNK2A1
Casein kinase 2, alpha prime




polypeptide


CSNK2B

Casein kinase 2, beta polypeptide


CST3

Cystatin C


CST6

Cystatin E/M


CSTA
STF1, STFA
Cystatin A (stefin A)


CSTB
CST6, EPM1, PME, STFB
Cystatin B (stefin B)


CTAG1A
ESO1, LAGE2A
Cancer/testis antigen 1A


CTAG1B
CT6.1, CTAG, CTAG1,
Cancer/testis antigen 1B



ESO1, LAGE2A, LAGE2B,



NY-ESO-1


CTAG2
CAMEL, CT6.2a, CT6.2b,
Cancer/testis antigen 2



ESO2, LAGE-1, LAGE-1a,



LAGE-1b, LAGE1,



MGC138724, MGC3803


CTGF
CCN2, IGFBP8
Connective tissue growth factor


CTNNB1
armadillo, beta-catenin,
Catenin (cadherin-associated protein),



CTNNB
beta 1, 88 kDa


CTNNBL1
C20orf33, FLJ21108, NAP,
Catenin, beta like 1



NYD-SP19, P14, P14L


CTSB

Cathepsin B


CTSD
CLN10, CPSD
Cathepsin D


CTSH
ACC-4, ACC-5, CPSB
Cathepsin H


CTSL
CTSL1, FLJ31037
Cathepsin L


CUL2

Cullin 2


CUL5
VACM-1
Cullin 5


CXCL1
FSP, GRO1, GROa, MGSA,
Chemokine (C-X-C motif) ligand 1



MGSA-a, NAP-3, SCYB1
(melanoma growth stimulating activity,




alpha)


CXCL10
C7, crg-2, gIP-10, IFI10,
Chemokine (C-X-C motif) ligand 10



INP10, IP-10, mob-1,



SCYB10


CXCL13
ANGIE, ANGIE2, BCA-1,
Chemokine (C-X-C motif) ligand 13



BLC, BLR1L, SCYB13


CXCL2
CINC-2a, GRO2, GROb,
Chemokine (C-X-C motif) ligand 2



MGSA-b, MIP-2a, SCYB2


CXCL5
ENA-78, SCYB5
Chemokine (C-X-C motif) ligand 5


CXCL8
3-10C, AMCF-I, b-ENAP,
Chemokine (C-X-C motif) ligand 8



GCP-1, GCP1, IL-8, IL8,



K60, LECT, LUCT,



LYNAP, MDNCF,



MONAP, NAF, NAP-1,



NAP1, SCYB8, TSG-1


CXCL9
CMK, crg-10, Humig, MIG,
Chemokine (C-X-C motif) ligand 9



SCYB9


CXCR1
CD181, CDw128a, CKR-1,
Chemokine (C-X-C motif) receptor 1



CMKAR1, IL8RA


CXCR2
CD182, CMKAR2, IL8RB
Chemokine (C-X-C motif) receptor 2


CXCR4
CD184, D2S201E, fusin,
Chemokine (C-X-C motif) receptor 4



HM89, HSY3RR, LESTR,



NPY3R, NPYR, NPYY3R


CYB5R3
DIA1
Cytochrome b5 reductase 3


CYP19A1
ARO, ARO1, aromatase,
Cytochrome P450, family 19,



CPV1, CYAR, CYP19, P-
subfamily A, polypeptide 1



450AROM


CYP1A2
CP12, P3-450
Cytochrome P450, family 1, subfamily




A, polypeptide 2


CYP2C19
CPCJ, CYP2C, P450IIC19
Cytochrome P450, family 2, subfamily




C, polypeptide 19


CYP2E1
CYP2E
Cytochrome P450, family 2, subfamily




E, polypeptide 1


CYP3A4
CYP3A3
Cytochrome P450, family 3, subfamily




A, polypeptide 4


CYP3A5
CP35, P450PCN3, PCN3
Cytochrome P450, family 3, subfamily




A, polypeptide 5


DAD1
OST2
Defender against cell death 1


DAPK1
DAPK
Death-associated protein kinase 1


DAXX
DAP6
Death-domain associated protein


DBI
ACBD1, ACBP
Diazepam binding inhibitor (GABA




receptor modulator, acyl-CoA binding




protein)


DCC
IGDCC1, NTN1R1
DCC netrin 1 receptor


DCDC1

Doublecortin domain containing 1


DCN
DSPG2, SLRR1B
Decorin


DDB2
DDBB, FLJ34321, UV-
Damage-specific DNA binding protein



DDB2
2, 48 kDa


DDIT3
CHOP, CHOP10,
DNA-damage-inducible transcript 3



GADD153


DEFA1
DEF1, DEFA2, HNP-1,
Defensin, alpha 1



MRS


DEFA1B

Defensin, alpha 1B


DEFA3
DEF3, HNP-3
Defensin, alpha 3, neutrophil-specific


DEK
D6S231E
DEK proto-oncogene


DES
CMD1I, CSM1, CSM2
Desmin


DHFR

Dihydrofolate reductase


DIAPH3
AN, AUNA1, DRF3,
Diaphanous-related formin 3



FLJ34705, NSDAN


DLC1
ARHGAP7, DLC-1, HP,
DLC1 Rho GTPase activating protein



p122-RhoGAP, STARD12


DNAJC2
MPHOSPH11, MPP11,
DnaJ (Hsp40) homolog, subfamily C,



ZRF1, ZUO1, zuotin
member 2


DST
BP240, BPA, BPAG1,
Dystonin



CATX-15, FLJ13425,



FLJ21489, FLJ30627,



FLJ32235, KIAA0728,



MACF2


DUSP1
CL100, HVH1, MKP-1,
Dual specificity phosphatase 1



PTPN10


DUSP14
MKP-L, MKP6
Dual specificity phosphatase 14


DUSP4
HVH2, MKP-2, TYP
Dual specificity phosphatase 4


DVL3
KIAA0208
Dishevelled segment polarity protein 3


DYNLL1
DLC1, DLC8, DNCL1,
Dynein, light chain, LC8-type 1



hdlc1, LC8, PIN


DYRK2

Dual-specificity tyrosine-(Y)-




phosphorylation regulated kinase 2


E2F1
RBBP3, RBP3
E2F transcription factor 1


E2F3

E2F transcription factor 3


E2F5

E2F transcription factor 5, p130-




binding


EBAG9
EB9, RCAS1
Estrogen receptor binding site




associated, antigen, 9


EDN1
ET1
Endothelin 1


EEF2
EEF-2, EF2
Eukaryotic translation elongation factor




2


EFNA1
ECKLG, EPLG1, LERK1,
Ephrin-A1



TNFAIP4


EFNA2
ELF-1, EPLG6, LERK6
Ephrin-A2


EFNA5
AF1, EPLG7, LERK7
Ephrin-A5


EFNB1
CFNS, Elk-L, EPLG2,
Ephrin-B1



LERK2


EFNB2
EPLG5, Htk-L, HTKL,
Ephrin-B2



LERK5, MGC126226,



MGC126227, MGC126228


EFNB3
EPLG8, LERK-8
Ephrin-B3


EGF

Epidermal growth factor


EGFR
ERBB, ERBB1
Epidermal growth factor receptor


EGR1
AT225, G0S30, KROX-24,
Early growth response 1



NGFI-A, TIS8, ZIF-268,



ZNF225


EI24
EPG4, PIG8, TP53I8
Etoposide induced 2.4


EIF3H
eIF3-gamma, eIF3-p40,
Eukaryotic translation initiation factor



eIF3h, EIF3S3
3, subunit H


EIF4E
EIF4E1, EIF4EL1, EIF4F
Eukaryotic translation initiation factor




4E


EIF4EBP1
4E-BP1, PHAS-I
Eukaryotic translation initiation factor




4E binding protein 1


EIF4G1
EIF4F, EIF4G, p220,
Eukaryotic translation initiation factor



PARK18
4 gamma, 1


EIF4H
KIAA0038, WBSCR1,
Eukaryotic translation initiation factor



WSCR1
4H


EIF5A
EIF-5A, EIF5A1,
Eukaryotic translation initiation factor



MGC104255, MGC99547
5A


ELANE
ELA2, HLE, HNE, NE
Elastase, neutrophil expressed


ELK3
ERP, NET, SAP2
ELK3, ETS-domain protein (SRF




accessory protein 2)


ENC1
ENC-1, KLHL37, NRPB,
Ectodermal-neural cortex 1 (with BTB



PIG10, TP53I10
domain)


ENG
CD105, END, HHT1, ORW,
Endoglin



ORW1


ENO1
ENO1L1, MBP-1, MPB1,
Enolase 1, (alpha)



PPH


ENO2

Enolase 2 (gamma, neuronal)


ENPP2
ATX, PD-IALPHA, PDNP2
Ectonucleotide




pyrophosphatase/phosphodiesterase 2


EPAS1
bHLHe73, HIF2A, HLF,
Endothelial PAS domain protein 1



MOP2, PASD2


EPCAM
17-1A, 323/A3, CD326,
Epithelial cell adhesion molecule



CO-17A, EGP-2, EGP34,



EGP40, Ep-CAM, ESA,



GA733-2, HEA125, KS1/4,



KSA, Ly74, M4S1, MH99,



MIC18, MK-1, MOC31,



TACST-1, TACSTD1,



TROP1


EPHA1
EPH, EPHT, EPHT1
EPH receptor A1


EPHA2
ECK
EPH receptor A2


EPHA3
ETK, ETK1, HEK, HEK4,
EPH receptor A3



TYRO4


EPHA4
Hek8, TYRO1
EPH receptor A4


EPHA7
Hek11
EPH receptor A7


EPHA8
EEK, Hek3
EPH receptor A8


EPHB2
DRT, EPHT3, ERK, Hek5,
EPH receptor B2



Tyro5


EPHB3
ETK2, Hek2, Tyro6
EPH receptor B3


EPHB4
HTK, Tyro11
EPH receptor B4


EPHX1
EPHX
Epoxide hydrolase 1, microsomal




(xenobiotic)


EPO
EP
Erythropoietin


EPOR

Erythropoietin receptor


ERBB2
CD340, HER-2, HER2,
V-erb-b2 avian erythroblastic leukemia



NEU, NGL
viral oncogene homolog 2


ERBB3
HER3, LCCS2
V-erb-b2 avian erythroblastic leukemia




viral oncogene homolog 3


ERBB4
ALS19
V-erb-b2 avian erythroblastic leukemia




viral oncogene homolog 4


ERCC1
RAD10
Excision repair cross-complementation




group 1


ERCC2
EM9, MAG, MGC102762,
Excision repair cross-complementation



MGC126218, MGC126219,
group 2



TFIIH, XPD


ERCC3
BTF2, GTF2H, RAD25,
Excision repair cross-complementation



TFIIH, XPB
group 3


ERCC4
FANCQ, RAD1, XPF
Excision repair cross-complementation




group 4


ERCC5
ERCM2, XPGC
Excision repair cross-complementation




group 5


ERCC6
ARMD5, CKN2, CSB,
Excision repair cross-complementation



RAD26
group 6


ESR1
Era, ESR, NR3A1
Estrogen receptor 1


ESR2
Erb, NR3A2
Estrogen receptor 2 (ER beta)


ETHE1
HSCO, YF13H12
Ethylmalonic encephalopathy 1


ETV4
E1A-F, E1AF, PEA3
Ets variant 4


ETV5
ERM
Ets variant 5


EXT1
LGCR, LGS, ttv
Exostosin glycosyltransferase 1


EZH2
ENX-1, EZH1, KMT6,
Enhancer of zeste 2 polycomb



KMT6A
repressive complex 2 subunit


EZR
VIL2
Ezrin


F13A1
F13A
Coagulation factor XIII, A1




polypeptide


F13B
FXIIIB
Coagulation factor XIII, B polypeptide


F2

Coagulation factor II (thrombin)


F3
CD142
Coagulation factor III (thromboplastin,




tissue factor)


FABP1
L-FABP
Fatty acid binding protein 1, liver


FABP2
I-FABP
Fatty acid binding protein 2, intestinal


FABP4
A-FABP, aP2
Fatty acid binding protein 4, adipocyte


FABP5
E-FABP, KFABP, PA-
Fatty acid binding protein 5 (psoriasis-



FABP
associated)


FADD
GIG3, MORT1
Fas (TNFRSF6)-associated via death




domain


FAF1
CGI-03, hFAF1, HFAF1s,
Fas (TNFRSF6) associated factor 1



UBXD12, UBXN3A


FAM129A
C1orf24, GIG39, NIBAN
Family with sequence similarity 129,




member A


FAP
DPPIV
Fibroblast activation protein, alpha


FAS
APO-1, APT1, CD95,
Fas cell surface death receptor



FAS1, TNFRSF6


FASLG
APT1LG1, CD178, FasL,
Fas ligand (TNF superfamily, member



TNFSF6
6)


FASN
FAS, SDR27X1
Fatty acid synthase


FBXO6
FBG2, FBS2, FBX6, Fbx6b
F-box protein 6


FCER2
CD23, CD23A, CLEC4J,
Fc fragment of IgE, low affinity II,



FCE2
receptor for (CD23)


FEN1
FEN-1, MF1, RAD2
Flap structure-specific endonuclease 1


FES
FPS
FES proto-oncogene, tyrosine kinase


FGA

Fibrinogen alpha chain


FGB

Fibrinogen beta chain


FGF1
AFGF, ECGF, ECGF-beta,
Fibroblast growth factor 1 (acidic)



ECGFA, ECGFB, FGF-



alpha, FGFA, GLIO703,



HBGF1


FGF17
FGF-13
Fibroblast growth factor 17


FGF18
FGF-18, ZFGF5
Fibroblast growth factor 18


FGF19

Fibroblast growth factor 19


FGF2
FGFB
Fibroblast growth factor 2 (basic)


FGF23

Fibroblast growth factor 23


FGF3
HBGF-3, INT2
Fibroblast growth factor 3


FGF4
HBGF-4, HST, HST-1,
Fibroblast growth factor 4



HSTF1, K-FGF, KFGF


FGF6

Fibroblast growth factor 6


FGF7
KGF
Fibroblast growth factor 7


FGF8
AIGF
Fibroblast growth factor 8 (androgen-




induced)


FGF9

Fibroblast growth factor 9


FGFR1
BFGFR, CD331, CEK,
Fibroblast growth factor receptor 1



FLG, FLT2, H2, H3, H4,



H5, KAL2, N-SAM


FGFR2
BEK, CD332, CEK3, CFD1,
Fibroblast growth factor receptor 2



ECT1, JWS, K-SAM,



KGFR, TK14, TK25


FGFR3
ACH, CD333, CEK2, JTK4
Fibroblast growth factor receptor 3


FGFR4
CD334, JTK2
Fibroblast growth factor receptor 4


FGG

Fibrinogen gamma chain


FHIT
AP3Aase, FRA3B
Fragile histidine triad


FIGF
VEGF-D, VEGFD
C-fos induced growth factor (vascular




endothelial growth factor D)


FKBP5
FKBP51, FKBP54, P54,
FK506 binding protein 5



PPIase, Ptg-10


FKBP8
FKBP38, FKBPr38
FK506 binding protein 8, 38 kDa


FLT1
FLT, VEGFR1
Fms-related tyrosine kinase 1


FLT4
PCL, VEGFR3
Fms-related tyrosine kinase 4


FMO5

Flavin containing monooxygenase 5


FN1
CIG, FINC, GFND2, LETS,
Fibronectin 1



MSF


FOLH1
FOLH, GCP2, GCPII,
Folate hydrolase (prostate-specific



NAALAD1, NAALAdase,
membrane antigen) 1



PSM, PSMA


FOS
AP-1, c-fos
FBJ murine osteosarcoma viral




oncogene homolog


FOSL1
fra-1
FOS-like antigen 1


FOXJ1
FKHL13, HFH-4, HFH4
Forkhead box J1


FOXM1
FKHL16, HFH-11, HNF-3,
Forkhead box M1



INS-1, MPHOSPH2, MPP2,



TGT3, trident


FOXO1
FKH1, FKHR, FOXO1A
Forkhead box O1


FOXO3
AF6q21, FKHRL1, FOXO2,
Forkhead box O3



FOXO3A


FOXQ1
HFH1
Forkhead box Q1


FSCN1
FLJ38511, p55, SNL
Fascin actin-bundling protein 1


FSHB

Follicle stimulating hormone, beta




polypeptide


FST
FS
Follistatin


FTH1
FHC, FTH, FTHL6, PIG15,
Ferritin, heavy polypeptide 1



PLIF


FTL
MGC71996, NBIA3
Ferritin, light polypeptide


FZD1
DKFZp564G072
Frizzled class receptor 1


FZD2

Frizzled class receptor 2


G6PD
G6PD1
Glucose-6-phosphate dehydrogenase


GADD45A
DDIT1, GADD45
Growth arrest and DNA-damage-




inducible, alpha


GADD45G
CR6, DDIT2,
Growth arrest and DNA-damage-



GADD45gamma, GRP17
inducible, gamma


GAS1

Growth arrest-specific 1


GAST
GAS
Gastrin


GATA3
HDR
GATA binding protein 3


GCLM
GLCLR
Glutamate-cysteine ligase, modifier




subunit


GDF15
MIC-1, MIC1, NAG-1,
Growth differentiation factor 15



PDF, PLAB, PTGFB


GDNF
ATF1, ATF2, HFB1-GDNF
Glial cell derived neurotrophic factor


GH1
GH, GH-N, GHN, hGH-N
Growth hormone 1


GH2
GH-V, GH2, GHL, GHV,
Growth hormone 2



hGH-V


GJA1
CX43, GJAL, ODD,
Gap junction protein, alpha 1, 43 kDa



ODDD, ODOD, SDTY3


GJB5
CX31.1
Gap junction protein, beta 5, 31.1 kDa


GLO1
GLOD1
Glyoxalase I


GMNN
Gem
Geminin, DNA replication inhibitor


GNAS
GNAS1, GNASXL, GPSA,
GNAS complex locus



NESP, NESP55, SCG6


GPA33
A33
Glycoprotein A33 (transmembrane)


GPC3
DGSX, OCI-5, SDYS, SGB,
Glypican 3



SGBS, SGBS1


GPI
AMF, NLK
Glucose-6-phosphate isomerase


GPX1

Glutathione peroxidase 1


GPX2
GSHPX-GI
Glutathione peroxidase 2




(gastrointestinal)


GRB10

Growth factor receptor-bound protein




10


GRB2
NCKAP2
Growth factor receptor-bound protein 2


GRB7

Growth factor receptor-bound protein 7


GSK3A

Glycogen synthase kinase 3 alpha


GSN
DKFZp313L0718
Gelsolin


GSR

Glutathione reductase


GSTM1
GST1, H-B, MU
Glutathione S-transferase mu 1


GSTM3
GST5
Glutathione S-transferase mu 3 (brain)


GSTP1
FAEES3, GST3, GSTP
Glutathione S-transferase pi 1


HDAC10
DKFZP761B039
Histone deacetylase 10


HDAC2
RPD3, YAF1
Histone deacetylase 2


HDAC5
FLJ90614, KIAA0600, NY-
Histone deacetylase 5



CO-9


HGF
DFNB39, F-TCF, HGFB,
Hepatocyte growth factor (hepapoietin



HPTA, SF
A; scatter factor)


HGFAC
HGFA, HGFAP
HGF activator


HIF1A
bHLHe78, HIF-1alpha,
Hypoxia inducible factor 1, alpha



HIF1, MOP1, PASD8
subunit (basic helix-loop-helix




transcription factor)


HIP1R
FLJ14000, HIP12, HIP3,
Huntingtin interacting protein 1 related



ILWEQ, KIAA0655


HIST1H2AC
H2AFL
Histone cluster 1, H2ac


HK1

Hexokinase 1


HK2

Hexokinase 2


HLA-G

Major histocompatibility complex,




class I, G


HMGA1
HMGIY
High mobility group AT-hook 1


HMGA2
BABL, HMGIC, LIPO
High mobility group AT-hook 2


HMOX1
bK286B10, HO-1
Heme oxygenase (decycling) 1


HOXA5
HOX1, HOX1C
Homeobox A5


HOXA9
HOX1, HOX1G
Homeobox A9


HP

Haptoglobin


HPGD
SDR36C1
Hydroxyprostaglandin dehydrogenase




15-(NAD)


HPN
TMPRSS1
Hepsin


HRAS
HRAS1
Harvey rat sarcoma viral oncogene




homolog


HSF1
HSTF1
Heat shock transcription factor 1


HSP90AA1
FLJ31884, Hsp89, Hsp90,
Heat shock protein 90 kDa alpha



HSP90N, HSPC1, HSPCA
(cytosolic), class A member 1


HSP90AB1
HSPC2, HSPCB
Heat shock protein 90 kDa alpha




(cytosolic), class B member 1


HSP90B1
GP96, GRP94, TRA1
Heat shock protein 90 kDa beta




(Grp94), member 1


HSPA1A
HSP70-1, HSPA1
Heat shock 70 kDa protein 1A


HSPA1B
HSP70-2
Heat shock 70 kDa protein 1B


HSPA1L
HSP70-HOM, hum70t
Heat shock 70 kDa protein 1-like


HSPA2

Heat shock 70 kDa protein 2


HSPA4
HS24/P52, HSPH2
Heat shock 70 kDa protein 4


HSPA8
HSC70, HSC71, HSP73,
Heat shock 70 kDa protein 8



HSPA10


HSPB1
Hs.76067, Hsp25, HSP27,
Heat shock 27 kDa protein 1



HSP28


HSPD1
GROEL, HSP60, SPG13
Heat shock 60 kDa protein 1




(chaperonin)


HSPE1
CPN10, GROES
Heat shock 10 kDa protein 1


HSPH1
HSP105A, HSP105B,
Heat shock 105 kDa/110 kDa protein 1



KIAA0201, NY-CO-25


IBSP
BSP, BSP-II, SP-II
Integrin-binding sialoprotein


ICAM1
BB2, CD54
Intercellular adhesion molecule 1


ID1
bHLHb24, dJ857M17.1.2
Inhibitor of DNA binding 1, dominant




negative helix-loop-helix protein


ID2
bHLHb26, GIG8
Inhibitor of DNA binding 2, dominant




negative helix-loop-helix protein


ID3
bHLHb25, HEIR-1
Inhibitor of DNA binding 3, dominant




negative helix-loop-helix protein


IDO1
IDO, INDO
Indoleamine 2, 3-dioxygenase 1


IFNA1
IFL, IFN, IFN-ALPHA,
Interferon, alpha 1



IFN-alphaD, IFNA13,



IFNA@


IFNA13

Interferon, alpha 13


IFNAR1
IFNAR, IFRC
Interferon (alpha, beta and omega)




receptor 1


IFNAR2
IFNABR
Interferon (alpha, beta and omega)




receptor 2


IFNB1
IFB, IFF, IFNB
Interferon, beta 1, fibroblast


IFNG

Interferon, gamma


IGF1
IGF-I, IGF1A, IGFI
Insulin-like growth factor 1




(somatomedin C)


IGF1R
CD221, IGFIR, IGFR,
Insulin-like growth factor 1 receptor



JTK13, MGC18216


IGF2
C11orf43, FLJ44734, IGF-II
Insulin-like growth factor 2


IGF2R
CD222, CIMPR, M6P-R,
Insulin-like growth factor 2 receptor



MPR1, MPRI


IGFBP2
IBP2
Insulin-like growth factor binding




protein 2, 36 kDa


IGFBP3
BP-53, IBP3
Insulin-like growth factor binding




protein 3


IL10
CSIF, IL-10, IL10A, TGIF
Interleukin 10


IL11
AGIF, IL-11
Interleukin 11


IL12A
CLMF, IL-12A, NFSK,
Interleukin 12A



NKSF1, p35


IL13
ALRH, BHR1, IL-13,
Interleukin 13



MGC116786, MGC116788,



MGC116789, P600


IL13RA2
CD213a2, CT19, IL-13R,
Interleukin 13 receptor, alpha 2



IL13BP


IL15
IL-15, MGC9721
Interleukin 15


IL16
FLJ16806, FLJ42735,
Interleukin 16



HsT19289, IL-16, LCF,



prIL-16


IL17A
CTLA8, IL-17, IL-17A,
Interleukin 17A



IL17


IL17B
IL-17B, IL-20,
Interleukin 17B



MGC138900, MGC138901,



NIRF, ZCYTO7


IL18
IGIF, IL-18, IL-1g, IL1F4
Interleukin 18


IL1A
IL-1A, IL1, IL1-ALPHA,
Interleukin 1, alpha



IL1F1


IL1B
IL-1B, IL1-BETA, IL1F2
Interleukin 1, beta


IL1R1
CD121A, D2S1473, IL1R,
Interleukin 1 receptor, type I



IL1RA


IL1R2
CD121b, IL1RB
Interleukin 1 receptor, type II


IL1RN
ICIL-1RA, IL-1RN, IL1F3,
Interleukin 1 receptor antagonist



IL1RA, IRAP, MGC10430


IL2
IL-2, TCGF
Interleukin 2


IL24
C49A, FISP, IL-24, IL10B,
Interleukin 24



mda-7, Mob-5, ST16


IL2RA
CD25, IDDM10, IL2R
Interleukin 2 receptor, alpha


IL2RB
CD122, IL15RB
Interleukin 2 receptor, beta


IL2RG
CD132, CIDX, IMD4,
Interleukin 2 receptor, gamma



SCIDX1


IL4
BCGF-1, BCGF1, BSF1, IL-4,
Interleukin 4



MGC79402


IL4R
CD124
Interleukin 4 receptor


IL5
EDF, IL-5, TRF
Interleukin 5


IL6
BSF2, HGF, HSF, IFNB2,
Interleukin 6



IL-6


IL6R
CD126
Interleukin 6 receptor


IL6ST
CD130, GP130
Interleukin 6 signal transducer


IL7
IL-7
Interleukin 7


IL9
HP40, IL-9, P40
Interleukin 9


ILF3
DRBP76, MPHOSPH4,
Interleukin enhancer binding factor 3,



MPP4, NF90, NFAR-1
90 kDa


ILK

Integrin-linked kinase


INHBA

Inhibin, beta A


INHBB

Inhibin, beta B


INS
IDDM1, IDDM2
Insulin


IRF1
MAR
Interferon regulatory factor 1


IRF4
LSIRF, MUM1
Interferon regulatory factor 4


ITGA1
CD49a, VLA1
Integrin, alpha 1


ITGA2
CD49B
Integrin, alpha 2 (CD49B, alpha 2




subunit of VLA-2 receptor)


ITGA2B
CD41, CD41B, GP2B,
Integrin, alpha 2b (platelet glycoprotein



PPP1R93
IIb of IIb/IIIa complex, antigen CD41)


ITGA3
CD49c, GAP-B3, MSK18,
Integrin, alpha 3 (antigen CD49C,



VCA-2, VLA3a
alpha 3 subunit of VLA-3 receptor)


ITGA4
CD49D
Integrin, alpha 4 (antigen CD49D,




alpha 4 subunit of VLA-4 receptor)


ITGA5
CD49e, FNRA
Integrin, alpha 5 (fibronectin receptor,




alpha polypeptide)


ITGA6
CD49f
Integrin, alpha 6


ITGAM
CD11B, CR3A, MAC-1
Integrin, alpha M (complement




component 3 receptor 3 subunit)


ITGAV
CD51, MSK8, VNRA,
Integrin, alpha V



VTNR


ITGB1
CD29, FNRB, GPIIA,
Integrin, beta 1 (fibronectin receptor,



MDF2, MSK12
beta polypeptide, antigen CD29




includes MDF2, MSK12)


ITGB3
CD61, GP3A, GPIIIa
Integrin, beta 3 (platelet glycoprotein




IIIa, antigen CD61)


ITGB4
CD104
Integrin, beta 4


ITGB5

Integrin, beta 5


ITGB6

Integrin, beta 6


ITGB8

Integrin, beta 8


ITIH4
H4P, IHRP, ITIHL1
Inter-alpha-trypsin inhibitor heavy




chain family, member 4


JKAMP
C14orf100, CDA06,
JNK1/MAPK8-associated membrane



HSPC213, HSPC327, JAMP
protein


JTB
hJT
Jumping translocation breakpoint


JUN
AP-1, c-Jun
Jun proto-oncogene


JUND
AP-1
Jun D proto-oncogene


JUP
CTNNG, DP3, DPIII,
Junction plakoglobin



PDGB, PKGB


KAT2B
GCN5, GCN5L, P/CAF,
K(lysine) acetyltransferase 2B



PCAF


KDR
CD309, FLK1, VEGFR,
Kinase insert domain receptor (a type



VEGFR2
III receptor tyrosine kinase)


KIF2A
HK2, KIF2
Kinesin heavy chain member 2A


KIF2C
CT139, KNSL6, MCAK
Kinesin family member 2C


KISS1

KiSS-1 metastasis-suppressor


KIT
C-Kit, CD117, PBT, SCFR
V-kit Hardy-Zuckerman 4 feline




sarcoma viral oncogene homolog


KITLG
FPH2, Kitl, KL-1, MGF,
KIT ligand



SCF, SF


KLF4
EZF, GKLF
Kruppel-like factor 4 (gut)


KLF5
BTEB2, CKLF, IKLF
Kruppel-like factor 5 (intestinal)


KLK10
NES1, PRSSL1
Kallikrein-related peptidase 10


KLK11
PRSS20, TLSP
Kallikrein-related peptidase 11


KLK13
KLK-L4
Kallikrein-related peptidase 13


KLK14
KLK-L6
Kallikrein-related peptidase 14


KLK15
ACO, HSRNASPH,
Kallikrein-related peptidase 15



prostinogen


KLK2

Kallikrein-related peptidase 2


KLK3
APS, PSA
Kallikrein-related peptidase 3


KLK4
EMSP, EMSP1, KLK-L1,
Kallikrein-related peptidase 4



PRSS17, PSTS


KLK5
KLK-L2, SCTE
Kallikrein-related peptidase 5


KLK6
Bssp, Klk7, neurosin,
Kallikrein-related peptidase 6



PRSS18, PRSS9


KLK7
PRSS6, SCCE
Kallikrein-related peptidase 7


KLK8
HNP, neuropsin, ovasin,
Kallikrein-related peptidase 8



PRSS19, TADG14


KLRK1
CD314, D12S2489E, KLR,
Killer cell lectin-like receptor



NKG2-D, NKG2D
subfamily K, member 1


KRAS
KRAS1, KRAS2
Kirsten rat sarcoma viral oncogene




homolog


KRT13
CK13, K13, MGC161462,
Keratin 13



MGC3781


KRT14
EBS3, EBS4
Keratin 14


KRT15
CK15, K15, K1CO
Keratin 15


KRT17
PCHC1
Keratin 17


KRT18

Keratin 18


KRT19
CK19, K19, K1CS,
Keratin 19



MGC15366


KRT4
CK4, CYK4, K4
Keratin 4


KRT8
CARD2, CK8, CYK8,
Keratin 8



K2C8, K8, KO


LALBA
LYZL7
Lactalbumin, alpha-


LAMB1
CLM
Laminin, beta 1


LAMC1
LAMB2
Laminin, gamma 1 (formerly LAMB2)


LCN1
MGC71975, PMFA, TLC,
Lipocalin 1



TP, VEGP


LDHA

Lactate dehydrogenase A


LEP
OB, OBS
Leptin


LGALS3
GALIG, LGALS2, MAC-2
Lectin, galactoside-binding, soluble, 3


LGALS3BP
90K, BTBD17B, CyCAP,
Lectin, galactoside-binding, soluble, 3



gp90, M2BP, MAC-2-BP,
binding protein



TANGO10B


LGALS4
GAL4
Lectin, galactoside-binding, soluble, 4


LGI1
EPITEMPIN, EPT, ETL1,
Leucine-rich, glioma inactivated 1



IB1099


LGMN
LGMN1, PRSC1
Legumain


LHB
CGB4, hLHB, LSH-B
Luteinizing hormone beta polypeptide


LHX1
LIM-1, LIM1
LIM homeobox 1


LIF
CDF, DIA, HILDA
Leukemia inhibitory factor


LIG4

Ligase IV, DNA, ATP-dependent


LIMK1
LIMK
LIM domain kinase 1


LMNA
CMD1A, HGPS, LGMD1B,
Lamin A/C



LMN1, LMNL1, PRO1


LRP1B
LRP-DIT, LRPDIT
Low density lipoprotein receptor-




related protein 1B


LRP6
ADCAD2
Low density lipoprotein receptor-




related protein 6


LTA
LT, TNFB, TNFSF1
Lymphotoxin alpha


LTA4H

Leukotriene A4 hydrolase


LTB
p33, TNFC, TNFSF3
Lymphotoxin beta (TNF superfamily,




member 3)


LTBR
D12S370, TNF-R-III,
Lymphotoxin beta receptor (TNFR



TNFCR, TNFR-RP,
superfamily, member 3)



TNFR2-RP, TNFRSF3


LTF
HLF2
Lactotransferrin


MAD2L1
HSMAD2, MAD2
MAD2 mitotic arrest deficient-like 1




(yeast)


MAD2L2
MAD2B, POLZ2, REV7
MAD2 mitotic arrest deficient-like 2




(yeast)


MAGEA3
CT1.3, HIP8, HYPD,
Melanoma antigen family A, 3



MAGE3, MGC14613


MAGEA4
CT1.4, MAGE-41, MAGE-
Melanoma antigen family A, 4



X2, MAGE4, MAGE4A,



MAGE4B, MGC21336


MAGEA6
CT1.6, MAGE6
Melanoma antigen family A, 6


MAGEB5
CT3.3, MAGE-B5
Melanoma antigen family B, 5


MAGEB6
CT3.4, FLJ40242, MAGE-
Melanoma antigen family B, 6



B6, MAGEB6A


MAGEC1
CT7, CT7.1, MAGE-C1,
Melanoma antigen family C, 1



MGC39366


MAGEC2
CT10, MAGE-C2,
Melanoma antigen family C, 2



MAGEE1


MAGEC3
CT7.2, HCA2, MAGE-C3
Melanoma antigen family C, 3


MAGED1
DLXIN-1, NRAGE
Melanoma antigen family D, 1


MAGED2
11B6, BCG1, HCA10, JCL-
Melanoma antigen family D, 2



1, MAGE-D2, MAGED,



MGC8386


MAGI1
AIP3, BAIAP1, BAP1,
Membrane associated guanylate kinase,



MAGI-1, TNRC19, WWP3
WW and PDZ domain containing 1


MAP2K1
MAPKK1, MEK1,
Mitogen-activated protein kinase



PRKMK1
kinase 1


MAP2K2
MEK2, PRKMK2
Mitogen-activated protein kinase




kinase 2


MAP2K4
JNKK1, MEK4, MKK4,
Mitogen-activated protein kinase



PRKMK4, SERK1
kinase 4


MAPK1
ERK, ERK2, MAPK2,
Mitogen-activated protein kinase 1



p41mapk, PRKM1, PRKM2


MAPK14
CSBP1, CSBP2, CSPB1,
Mitogen-activated protein kinase 14



Mxi2, p38, PRKM14,



PRKM15


MAPK3
ERK1, p44erk1, p44mapk,
Mitogen-activated protein kinase 3



PRKM3


MAPK7
BMK1, ERK5, PRKM7
Mitogen-activated protein kinase 7


MAPK8
JNK, JNK1, PRKM8,
Mitogen-activated protein kinase 8



SAPK1


MAPKAPK2

Mitogen-activated protein kinase-




activated protein kinase 2


MBD1
CXXC3, PCM1
Methyl-CpG binding domain protein 1


MBD2

Methyl-CpG binding domain protein 2


MBD4
MED1
Methyl-CpG binding domain protein 4


MCL1
BCL2L3, Mcl-1
Myeloid cell leukemia 1


MCM2
BM28, CCNL1, cdc19,
Minichromosome maintenance



CDCL1, D3S3194,
complex component 2



KIAA0030


MCM3

Minichromosome maintenance




complex component 3


MCM5
CDC46
Minichromosome maintenance




complex component 5


MCM7
CDC47, MCM2, PPP1R104
Minichromosome maintenance




complex component 7


MDH1

Malate dehydrogenase 1, NAD




(soluble)


MDK
FLJ27379, MK, NEGF2
Midkine (neurite growth-promoting




factor 2)


MDM2
HDM2, MGC5370
MDM2 proto-oncogene, E3 ubiquitin




protein ligase


MECP2
MRX16, MRX79, RTT
Methyl CpG binding protein 2


MED1
CRSP1, CRSP200,
Mediator complex subunit 1



DRIP230, PBP, PPARBP,



PPARGBP, RB18A,



TRAP220, TRIP2


MET
HGFR, RCCP2
MET proto-oncogene, receptor tyrosine




kinase


MFGE8
BA46, EDIL1, hP47,
Milk fat globule-EGF factor 8 protein



HsT19888, MFG-E8,



OAcGD3S, SED1, SPAG10


MGMT

O-6-methylguanine-DNA




methyltransferase


MIA
CD-RAP
Melanoma inhibitory activity


MIF
GIF, GLIF
Macrophage migration inhibitory factor




(glycosylation-inhibiting factor)


MKI67
MIB-, PPP1R105
Marker of proliferation Ki-67


MLH1
COCA2, FCC2, HNPCC,
MutL homolog 1



HNPCC2


MLLT11
AF1Q
Myeloid/lymphoid or mixed-lineage




leukemia (trithorax homolog,





Drosophila); translocated to, 11



MME
CALLA, CD10, NEP
Membrane metallo-endopeptidase


MMP1
CLG
Matrix metallopeptidase 1 (interstitial




collagenase)


MMP10
STMY2
Matrix metallopeptidase 10




(stromelysin 2)


MMP11
STMY3
Matrix metallopeptidase 11




(stromelysin 3)


MMP12
HME
Matrix metallopeptidase 12




(macrophage elastase)


MMP13
CLG3
Matrix metallopeptidase 13




(collagenase 3)


MMP14
MT1-MMP
Matrix metallopeptidase 14




(membrane-inserted)


MMP15
MT2-MMP, MTMMP2,
Matrix metallopeptidase 15



SMCP-2
(membrane-inserted)


MMP16
C8orf57, DKFZp761D112,
Matrix metallopeptidase 16



MT3-MMP
(membrane-inserted)


MMP2
CLG4, CLG4A, TBE-1
Matrix metallopeptidase 2 (gelatinase




A, 72 kDa gelatinase, 72 kDa type IV




collagenase)


MMP3
STMY, STMY1
Matrix metallopeptidase 3 (stromelysin




1, progelatinase)


MMP7
MPSL1, PUMP-1
Matrix metallopeptidase 7 (matrilysin,




uterine)


MMP8
CLG1
Matrix metallopeptidase 8 (neutrophil




collagenase)


MMP9
CLG4B
Matrix metallopeptidase 9 (gelatinase




B, 92 kDa gelatinase, 92 kDa type IV




collagenase)


MPO

Myeloperoxidase


MRE11A
ATLD, MRE11
MRE11 meiotic recombination 11




homolog A (S. cerevisiae)


MSH6
GTBP
MutS homolog 6


MSLN
CAK1, MPF
Mesothelin


MSMB
IGBF, MSP, MSPB, PN44,
Microseminoprotein, beta-



PRPS, PSP, PSP-94, PSP57,



PSP94


MSR1
CD204, SCARA1
Macrophage scavenger receptor 1


MT1A
MT1, MT1S
Metallothionein 1A


MT1G
MT1, MT1K
Metallothionein 1G


MTA1

Metastasis associated 1


MUC1
ADMCKD, ADMCKD1,
Mucin 1, cell surface associated



CD227, MCD, MCKD,



MCKD1, PEM, PUM


MUTYH
MYH
MutY homolog


MVP
LRP, VAULT1
Major vault protein


MXI1
bHLHc11, MAD2, MXD2,
MAX interactor 1, dimerization protein



MXI


MYBL2
B-MYB, BMYB
V-myb avian myeloblastosis viral




oncogene homolog-like 2


MYC
bHLHe39, c-Myc, MYCC
V-myc avian myelocytomatosis viral




oncogene homolog


MYOCD
MYCD
Myocardin


MYOD1
bHLHc1, MYF3, MYOD,
Myogenic differentiation 1



PUM


MYOG
bHLHc3, MYF4
Myogenin (myogenic factor 4)


NAGA
D22S674
N-acetylgalactosaminidase, alpha-


NAIP
BIRC1, NLRB1
NLR family, apoptosis inhibitory




protein


NAMPT
PBEF, PBEF1
Nicotinamide




phosphoribosyltransferase


NAT2
AAC2
N-acetyltransferase 2 (arylamine N-




acetyltransferase)


NCAM1
CD56, NCAM
Neural cell adhesion molecule 1


NCOA3
ACTR, AIB1, bHLHe42,
Nuclear receptor coactivator 3



CAGH16, KAT13B, p/CIP,



RAC3, SRC-3, SRC3,



TNRC16, TRAM-1


NDRG1
CAP43, DRG1, NDR1,
N-myc downstream regulated 1



RTP, TDD5


NEDD8
Nedd-8
Neural precursor cell expressed,




developmentally down-regulated 8


NEO1
HsT17534, IGDCC2, NGN,
Neogenin 1



NTN1R2


NFKB1
KBF1, NF-kappaB, NF-
Nuclear factor of kappa light



kB1, NFkappaB, NFKB-
polypeptide gene enhancer in B-cells 1



p50, p105, p50


NFKB2
LYT-10, NF-kB2, p105, p52
Nuclear factor of kappa light




polypeptide gene enhancer in B-cells 2




(p49/p100)


NFKBIA
IkappaBalpha, IKBA,
Nuclear factor of kappa light



MAD-3, NFKBI
polypeptide gene enhancer in B-cells




inhibitor, alpha


NFKBIE
IKBE
Nuclear factor of kappa light




polypeptide gene enhancer in B-cells




inhibitor, epsilon


NGF
NGFB
Nerve growth factor (beta polypeptide)


NGFR
CD271, p75NTR,
Nerve growth factor receptor



TNFRSF16


NKX3-1
BAPX2, NKX3.1, NKX3A
NK3 homeobox 1


NME1
NDPKA, NM23, NM23-H1
NME/NM23 nucleoside diphosphate




kinase 1


NME2
NDPKB, NM23-H2
NME/NM23 nucleoside diphosphate




kinase 2


NOS1
nNOS, NOS
Nitric oxide synthase 1 (neuronal)


NOS2
HEP-NOS, iNOS, NOS,
Nitric oxide synthase 2, inducible



NOS2A


NOS3
ECNOS, eNOS
Nitric oxide synthase 3 (endothelial




cell)


NOTCH1
TAN1
Notch 1


NOTCH2

Notch 2


NOTCH3
CADASIL, CASIL
Notch 3


NQO1
DHQU, DIA4, DTD,
NAD(P)H dehydrogenase, quinone 1



NMOR1, QR1


NR0B1
AHC, AHCH, DAX1, DSS,
Nuclear receptor subfamily 0, group B,



NR0B1
member 1


NRG1
GGF, HGL, HRG, NDF,
Neuregulin 1



NRG1-IT2


NRG2
Don-1, HRG2, NTAK
Neuregulin 2


NRG3

Neuregulin 3


NRP1
CD304, NRP, VEGF165R
Neuropilin 1


NRP2
VEGF165R2
Neuropilin 2


NTF3
NGF2
Neurotrophin 3


NTF4
GLC1O, NT-4/5, NTF5
Neurotrophin 4


NTHL1
NTH1, OCTS3
Nth endonuclease III-like 1 (E. coli)


NTN1
NTN1L
Netrin 1


NTRK1
MTC, TRK, TRKA
Neurotrophic tyrosine kinase, receptor,




type 1


NTRK2
TRKB
Neurotrophic tyrosine kinase, receptor,




type 2


NTRK3
TRKC
Neurotrophic tyrosine kinase, receptor,




type 3


NUDT1
MTH1
Nudix (nucleoside diphosphate linked




moiety X)-type motif 1


NUMB
C14orf41
Numb homolog (Drosophila)


OGG1
HMMH, HOGG1, MUTM,
8-oxoguanine DNA glycosylase



OGH1


OR51E2
PSGR
Olfactory receptor, family 51,




subfamily E, member 2


ORM1

Orosomucoid 1


OSM
MGC20461
Oncostatin M


PAGE4
CT16.7, GAGEC1, PAGE-4
P antigen family, member 4 (prostate




associated)


PAPPA
ASBABP2, DIPLA1,
Pregnancy-associated plasma protein



IGFBP-4ase, PAPA, PAPP-
A, pappalysin 1



A, PAPPA1


PARP1
ADPRT, PARP, PPOL
Poly (ADP-ribose) polymerase 1


PARVB
CGI-56
Parvin, beta


PAX5
BSAP
Paired box 5


PAX8

Paired box 8


PCNA

Proliferating cell nuclear antigen


PDGFA
PDGF-A, PDGF1
Platelet-derived growth factor alpha




polypeptide


PDGFB
SIS, SSV
Platelet-derived growth factor beta




polypeptide


PDGFRA
CD140a, PDGFR2
Platelet-derived growth factor




receptor, alpha polypeptide


PDGFRB
CD140b, JTK12, PDGFR,
Platelet-derived growth factor receptor,



PDGFR1
beta polypeptide


PDZD4
FLJ34125, KIAA1444,
PDZ domain containing 4



LU1, PDZK4, PDZRN4L


PF4
CXCL4, SCYB4
Platelet factor 4


PGC

Progastricsin (pepsinogen C)


PGF
D12S1900, PGFL, PLGF,
Placental growth factor



PIGF-2, SHGC-10760


PGR
NR3C3, PR
Progesterone receptor


PHF20
C20orf104, dJ1121G12.1,
PHD finger protein 20



TDRD20A


PIGR

Polymeric immunoglobulin receptor


PIK3CA
PI3K
Phosphatidylinositol-4,5-bisphosphate




3-kinase, catalytic subunit alpha


PIK3R1
GRB1, p85, p85-ALPHA
Phosphoinositide-3-kinase, regulatory




subunit 1 (alpha)


PIK3R2
p85, P85B
Phosphoinositide-3-kinase, regulatory




subunit 2 (beta)


PIK3R3
p55
Phosphoinositide-3-kinase, regulatory




subunit 3 (gamma)


PIM1
PIM
Pim-1 proto-oncogene, serine/threonine




kinase


PIM2

Pim-2 proto-oncogene, serine/threonine




kinase


PIM3

Pim-3 proto-oncogene, serine/threonine




kinase


PIN1
dod
Peptidylprolyl cis/trans isomerase,




NIMA-interacting 1


PIP4K2B
PIP5K2B, PIP5KIIB,
Phosphatidylinositol-5-phosphate 4-



PIP5KIIbeta
kinase, type II, beta


PKM
OIP3, PK3, PKM2, THBP1
Pyruvate kinase, muscle


PLAT

Plasminogen activator, tissue


PLAU
UPA, URK
Plasminogen activator, urokinase


PLAUR
CD87, UPAR, URKR
Plasminogen activator, urokinase




receptor


PLG

Plasminogen


PLK1
PLK
Polo-like kinase 1


PLP1
GPM6C, PLP, SPG2
Proteolipid protein 1


PMEPA1
STAG1, TMEPAI
Prostate transmembrane protein,




androgen induced 1


PML
MYL, RNF71, TRIM19
Promyelocytic leukemia


PMP22
GAS-3, HNPP, Sp110
Peripheral myelin protein 22


PNMT
PENT
Phenylethanolamine N-




methyltransferase


POMC
ACTH, CLIP, LPH, MSH,
Proopiomelanocortin



NPP, POC


PON1
ESA, PON
Paraoxonase 1


POSTN
OSF-2, periostin, PN
Periostin, osteoblast specific factor


POU2F2
OCT2, OTF2
POU class 2 homeobox 2


PPA2
FLJ20459
Pyrophosphatase (inorganic) 2


PPARG
NR1C3, PPARG1,
Peroxisome proliferator-activated



PPARG2, PPARgamma
receptor gamma


PPARGC1A
PGC1, PGC1A, PPARGC1
Peroxisome proliferator-activated




receptor gamma, coactivator 1 alpha


PPM1D
PP2C-DELTA, Wip1
Protein phosphatase, Mg2+/Mn2+




dependent, 1D


PPP1R15A
GADD34
Protein phosphatase 1, regulatory




subunit 15A


PPY
PNP
Pancreatic polypeptide


PRDM13

PR domain containing 13


PRDM16
KIAA1675, MEL1,
PR domain containing 16



MGC166915, PFM13


PRDX2
MGC4104, NKEFB, PRP,
Peroxiredoxin 2



PRX2, PRXII, TDPX1, TSA


PRDX4
AOE37-2
Peroxiredoxin 4


PRKCA
PKCA
Protein kinase C, alpha


PRKCB
PKCB, PRKCB1, PRKCB2
Protein kinase C, beta


PRKCE

Protein kinase C, epsilon


PRKCH
PKC-L, PKCL, PRKCL
Protein kinase C, eta


PRKCI
DXS1179E, PKCI
Protein kinase C, iota


PRKCQ

Protein kinase C, theta


PRKDC
DNA-PKcs, DNAPK,
Protein kinase, DNA-activated,



DNPK1, HYRC, HYRC1,
catalytic polypeptide



p350, XRCC7


PRL

Prolactin


PROC

Protein C (inactivator of coagulation




factors Va and VIIIa)


PRSS1
TRY1
Protease, serine, 1 (trypsin 1)


PSCA

Prostate stem cell antigen


PSMD4
AF, AF-1, Rpn10, S5A
Proteasome (prosome, macropain) 26S




subunit, non-ATPase, 4


PTCH1
BCNS, NBCCS, PTCH
Patched 1


PTCH2

Patched 2


PTGS1
COX1, PGHS-1, PTGHS
Prostaglandin-endoperoxide synthase 1




(prostaglandin G/H synthase and




cyclooxygenase)


PTGS2
COX2
Prostaglandin-endoperoxide synthase 2




(prostaglandin G/H synthase and




cyclooxygenase)


PTH
PTH1
Parathyroid hormone


PTHLH
HHM, PLP, PTHR, PTHRP
Parathyroid hormone-like hormone


PTK2
FADK, FAK, FAK1,
Protein tyrosine kinase 2



PPP1R71


PTN
HBGF8, HBNF, NEGF1
Pleiotrophin


PTPRO
GLEPP1, NPHS6, PTP-oc,
Protein tyrosine phosphatase, receptor



PTP-U2, PTPU2
type, O


PTTG1
EAP1, HPTTG, PTTG,
Pituitary tumor-transforming 1



securin, TUTR1


PURA
PUR-ALPHA, PUR1,
Purine-rich element binding protein A



PURALPHA


PZP
CPAMD6
Pregnancy-zone protein


RAB11FIP3
eferin, KIAA0665, Rab11-
RAB11 family interacting protein 3



FIP3
(class II)


RAB18

RAB18, member RAS oncogene




family


RAB25
CATX-8
RAB25, member RAS oncogene




family


RAC1
p21-Rac1, Rac-1, TC-25
Ras-related C3 botulinum toxin




substrate 1 (rho family, small GTP




binding protein Rac1)


RAD23A
HHR23A, MGC111083
RAD23 homolog A (S. cerevisiae)


RAD23B
HHR23B, HR23B, P58
RAD23 homolog B (S. cerevisiae)


RAD51
BRCC5, HsRad51,
RAD51 recombinase



HsT16930, RAD51A,



RECA


RAD51D
HsTRAD, R51H3,
RAD51 paralog D



RAD51L3, Trad


RAD52

RAD52 homolog (S. cerevisiae)


RAD54B
RDH54
RAD54 homolog B (S. cerevisiae)


RAF1
c-Raf, CRAF, Raf-1
Raf-1 proto-oncogene, serine/threonine




kinase


RARA
NR1B1, RAR
Retinoic acid receptor, alpha


RARB
HAP, NR1B2, RRB2
Retinoic acid receptor, beta


RARG
NR1B3, RARC
Retinoic acid receptor, gamma


RASA1
CM-AVM, GAP, p120GAP,
RAS p21 protein activator (GTPase



p120RASGAP, RASA
activating protein) 1


RB1
OSRC, PPP1R130, RB
Retinoblastoma 1


RBBP4
lin-53, NURF55, RbAp48
Retinoblastoma binding protein 4


RBL1
cp107, p107, PRB1
Retinoblastoma-like 1


RBL2
p130, Rb2
Retinoblastoma-like 2


RBM6
3G2, DEF-3, DEF3, g16,
RNA binding motif protein 6



NY-LU-12


RBP4

Retinol binding protein 4, plasma


REL
c-Rel, I-Rel
V-rel avian reticuloendotheliosis viral




oncogene homolog


RELA
NFKB3, p65
V-rel avian reticuloendotheliosis viral




oncogene homolog A


RELB
REL-B
V-rel avian reticuloendotheliosis viral




oncogene homolog B


RET
CDHF12, CDHR16,
Ret proto-oncogene



HSCR1, MEN2A, MEN2B,



MTC1, PTC, RET51


RHOA
ARH12, ARHA, Rho12,
Ras homolog family member A



RhoA, RHOH12


RHOB
ARH6, ARHB, MST081,
Ras homolog family member B



RhoB, RHOH6


RHOC
ARH9, ARHC, RhoC
Ras homolog family member C


RPA2

Replication protein A2, 32 kDa


RPL27
L27
Ribosomal protein L27


RPS3
FLJ26283, FLJ27450,
Ribosomal protein S3



MGC87870, S3


RPS6KA1
HU-1, RSK, RSK1
Ribosomal protein S6 kinase, 90 kDa,




polypeptide 1


RPS6KA3
CLS, HU-3, MRX19, RSK,
Ribosomal protein S6 kinase, 90 kDa,



RSK2
polypeptide 3


RXRA
NR2B1
Retinoid X receptor, alpha


RXRB
H-2RIIBP, NR2B2, RCoR-1
Retinoid X receptor, beta


RXRG
NR2B3
Retinoid X receptor, gamma


S100A1
S100-alpha, S100A
S100 calcium binding protein A1


S100A2
CAN19, S100L
S100 calcium binding protein A2


S100A4
18A2, 42A, CAPL, FSP1,
S100 calcium binding protein A4



MTS1, P9KA, PEL98


S100A6
2A9, CABP, CACY, PRA
S100 calcium binding protein A6


S100A7
PSOR1, S100A7c
S100 calcium binding protein A7


S100A8
60B8AG, CAGA, CFAG,
S100 calcium binding protein A8



CGLA, MRP8, P8


S100A9
60B8AG, CAGB, CFAG,
S100 calcium binding protein A9



CGLB, LIAG, MAC387,



MIF, MRP14, NIF, P14


S100B
S100beta
S100 calcium binding protein B


S1PR1
CD363, D1S3362, edg-1,
Sphingosine-1-phosphate receptor 1



EDG1


SAA1
PIG4, SAA, TP53I4
Serum amyloid A1


SAA2

Serum amyloid A2


SART1
Ara1, SNRNP110, Snu66
Squamous cell carcinoma antigen




recognized by T cells


SCGB1A1
CC10, CC16, CCSP, UGB
Secretoglobin, family 1A, member 1




(uteroglobin)


SCGB1D2
LIPB, LPHB
Secretoglobin, family 1D, member 2


SCGB2A1
LPHC, MGB2, MGC71973,
Secretoglobin, family 2A, member 1



UGB3


SCGB2A2
MGB1, MGC71974, UGB2
Secretoglobin, family 2A, member 2


SDC1
CD138, SDC, SYND1,
Syndecan 1



syndecan


SELE
CD62E, ELAM, ELAM1,
Selectin E



ESEL


SELL
CD62L, hLHRc, LAM-1,
Selectin L



LAM1, Leu-8, LNHR,



LSEL, Lyam-1, LYAM1,



PLNHR


SELP
CD62, CD62P, GMP140,
Selectin P (granule membrane protein



GRMP, PADGEM, PSEL
140 kDa, antigen CD62)


SEMA3B
LUCA-1, SemA, sema5,
Sema domain, immunoglobulin domain



SEMAA, semaV
(Ig), short basic domain, secreted,




(semaphorin) 3B


2-Sep
DIFF6, hNedd5, KIAA0158,
Septin 2



NEDD5, Pnutl3


SERPINA1
A1A, A1AT, AAT, alpha-1-
Serpin peptidase inhibitor, clade A



antitrypsin, alpha1AT, PI,
(alpha-1 antiproteinase, antitrypsin),



PI1
member 1


SERPINA3
AACT, ACT, alpha-1-
Serpin peptidase inhibitor, clade A



antichymotrypsin
(alpha-1 antiproteinase, antitrypsin),




member 3


SERPINA5
PAI3, PCI, PLANH3,
Serpin peptidase inhibitor, clade A



PROCI
(alpha-1 antiproteinase, antitrypsin),




member 5


SERPINB2
HsT1201, PAI2, PLANH2
Serpin peptidase inhibitor, clade B




(ovalbumin), member 2


SERPINB3
HsT1196, SCC, SCCA1,
Serpin peptidase inhibitor, clade B



T4-A
(ovalbumin), member 3


SERPINB4
LEUPIN, PI11, SCCA-2,
Serpin peptidase inhibitor, clade B



SCCA1, SCCA2
(ovalbumin), member 4


SERPINE1
PAI, PAI1, PLANH1
Serpin peptidase inhibitor, clade E




(nexin, plasminogen activator inhibitor




type 1), member 1


SERPINF1
EPC-1, PEDF, PIG35
Serpin peptidase inhibitor, clade F




(alpha-2 antiplasmin, pigment




epithelium derived factor), member 1


SFN
YWHAS
Stratifin


SHBG
ABP, MGC126834,
Sex hormone-binding globulin



MGC138391, TEBG


SIRT2
SIR2L
Sirtuin 2


SKP2
FBL1, FBXL1, p45
S-phase kinase-associated protein 2, E3




ubiquitin protein ligase


SLC19A1
FOLT
Solute carrier family 19 (folate




transporter), member 1


SLC2A1
DYT18, GLUT, GLUT1,
Solute carrier family 2 (facilitated



HTLVR
glucose transporter), member 1


SLC3A2
4F2, 4F2HC, 4T2HC,
Solute carrier family 3 (amino acid



CD98, CD98HC, MDU1,
transporter heavy chain), member 2



NACAE


SLPI
ALK1, ALP, BLPI, HUSI,
Secretory leukocyte peptidase inhibitor



HUSI-I, WAP4, WFDC4


SMAD1
JV4-1, MADH1, MADR1
SMAD family member 1


SMAD2
JV18-1, MADH2, MADR2
SMAD family member 2


SMAD3
HsT17436, JV15-2,
SMAD family member 3



MADH3


SMAD4
DPC4, MADH4
SMAD family member 4


SMYD3
KMT3E, ZMYND1,
SET and MYND domain containing 3



ZNFN3A1


SOD1
ALS, ALS1, IPOA
Superoxide dismutase 1, soluble


SOD2

Superoxide dismutase 2, mitochondrial


SOX1

SRY (sex determining region Y)-box 1


SOX9
CMD1, CMPD1, SRA1
SRY (sex determining region Y)-box 9


SP1

Sp1 transcription factor


SPARC
ON
Secreted protein, acidic, cysteine-rich




(osteonectin)


SPARCL1
MAST9
SPARC-like 1 (hevin)


SPINK1
PCTT, PSTI, Spink3, TATI
Serine peptidase inhibitor, Kazal type 1


SPINT1
HAI, MANSC2
Serine peptidase inhibitor, Kunitz type




1


SPINT2
HAI-2, Kop
Serine peptidase inhibitor, Kunitz type,




2


SPP1
BNSP, BSPI, ETA-1, OPN
Secreted phosphoprotein 1


SPRR1B
GADD33, SPRR1
Small proline-rich protein 1B


SPRR3

Small proline-rich protein 3


SPRY1
hSPRY1
Sprouty homolog 1, antagonist of FGF




signaling (Drosophila)


SRC
ASV, c-src, SRC1
SRC proto-oncogene, non-receptor




tyrosine kinase


SRD5A1

Steroid-5-alpha-reductase, alpha




polypeptide 1 (3-oxo-5 alpha-steroid




delta 4-dehydrogenase alpha 1)


SRD5A2

Steroid-5-alpha-reductase, alpha




polypeptide 2 (3-oxo-5 alpha-steroid




delta 4-dehydrogenase alpha 2)


SST
SMST
Somatostatin


SSX2
CT5.2a, HD21, HOM-MEL-
Synovial sarcoma, X breakpoint 2



40, MGC119055,



MGC15364, MGC3884,



SSX


SSX2B
CT5.2b
Synovial sarcoma, X breakpoint 2B


ST14
HAI, MT-SP1, PRSS14,
Suppression of tumorigenicity 14



SNC19, TMPRSS14
(colon carcinoma)


STARD3
es64, MLN64
StAR-related lipid transfer (START)




domain containing 3


STAT4

Signal transducer and activator of




transcription 4


STAT5A
MGF, STAT5
Signal transducer and activator of




transcription 5A


STEAP1
PRSS24, STEAP
Six transmembrane epithelial antigen




of the prostate 1


STMN1
C1orf215, FLJ32206, Lag,
Stathmin 1



LAP18, OP18, PP17, PP19,



PR22, SMN


STRAP
MAWD, pt-wd, UNRIP
Serine/threonine kinase receptor




associated protein


STT3A
ITM1, MGC9042, STT3-A,
STT3A, subunit of the



TMC
oligosaccharyltransferase complex




(catalytic)


SULT1E1
EST, STE
Sulfotransferase family 1E, estrogen-




preferring, member 1


TAGLN
DKFZp686P11128, SM22,
Transgelin



SMCC, TAGLN1, WS3-10


TDRD6
bA446F17.4, CT41.2, NY-
Tudor domain containing 6



CO-45, SPATA36


TEK
CD202b, TIE-2, TIE2,
TEK tyrosine kinase, endothelial



VMCM, VMCM1


TERT
EST2, hEST2, TCS1, TP2,
Telomerase reverse transcriptase



TRT


TF
PRO1557, PRO2086
Transferrin


TFAP2B
AP2-B
Transcription factor AP-2 beta




(activating enhancer binding protein 2




beta)


TFDP1
Dp-1, DP1, DRTF1
Transcription factor Dp-1


TFDP2
Dp-2
Transcription factor Dp-2 (E2F




dimerization partner 2)


TFF1
BCEI, D21S21, HP1.A,
Trefoil factor 1



HPS2, pNR-2, pS2


TFF2
SML1
Trefoil factor 2


TFF3
HITF, ITF
Trefoil factor 3 (intestinal)


TFRC
CD71, p90, TFR1
Transferrin receptor


TG
AITD3, TGN
Thyroglobulin


TGFA

Transforming growth factor, alpha


TGFB1
CED, DPD1, TGFB,
Transforming growth factor, beta 1



TGFbeta


TGFB2

Transforming growth factor, beta 2


TGFB3
ARVD, ARVD1
Transforming growth factor, beta 3


TGFBR3
betaglycan, BGCAN
Transforming growth factor, beta




receptor III


TGM4
TGP
Transglutaminase 4


TGM7
TGMZ
Transglutaminase 7


THBS1
THBS, THBS-1, TSP, TSP-
Thrombospondin 1



1, TSP1


THBS2
TSP2
Thrombospondin 2


THBS4

Thrombospondin 4


THPO
MGDF, MPLLG, TPO
Thrombopoietin


THRA
AR7, EAR-7.1/EAR-7.2,
Thyroid hormone receptor, alpha



ERBA, ERBA1, NR1A1,



THRA1, THRA2, THRA3


THRB
ERBA-BETA, ERBA2,
Thyroid hormone receptor, beta



GRTH, NR1A2, PRTH,



THR1, THRB1, THRB2


TIE1
JTK14, TIE
Tyrosine kinase with immunoglobulin-




like and EGF-like domains 1


TIMP1
CLGI, EPO, TIMP
TIMP metallopeptidase inhibitor 1


TIMP2
CSC-21K
TIMP metallopeptidase inhibitor 2


TIMP3
SFD
TIMP metallopeptidase inhibitor 3


TK1

Thymidine kinase 1, soluble


TMF1
ARA160, TMF
TATA element modulatory factor 1


TMPRSS2
PRSS10
Transmembrane protease, serine 2


TMPRSS3
DFNB10, DFNB8
Transmembrane protease, serine 3


TNC
DFNA56, HXB,
Tenascin C



MGC167029, TN


TNF
DIF, TNF-alpha, TNFA,
Tumor necrosis factor



TNFSF2


TNFAIP2
B94, EXOC3L3
Tumor necrosis factor, alpha-induced




protein 2


TNFAIP3
A20, OTUD7C
Tumor necrosis factor, alpha-induced




protein 3


TNFRSF10A
Apo2, CD261, DR4,
Tumor necrosis factor receptor



TRAILR-1
superfamily, member 10a


TNFRSF10B
CD262, DR5, KILLER,
Tumor necrosis factor receptor



TRAIL-R2, TRICK2A,
superfamily, member 10b



TRICKB


TNFRSF10C
CD263, DcR1, LIT,
Tumor necrosis factor receptor



TRAILR3, TRID
superfamily, member 10c, decoy




without an intracellular domain


TNFRSF10D
CD264, DcR2, TRAILR4,
Tumor necrosis factor receptor



TRUNDD
superfamily, member 10d, decoy with




truncated death domain


TNFRSF11B
OCIF, OPG, TR1
Tumor necrosis factor receptor




superfamily, member 11b


TNFRSF12A
CD266, FN14, TweakR
Tumor necrosis factor receptor




superfamily, member 12A


TNFRSF14
ATAR, CD270, HVEA,
Tumor necrosis factor receptor



HVEM, LIGHTR, TR2
superfamily, member 14


TNFRSF1A
CD120a, TNF-R, TNF-R-I,
Tumor necrosis factor receptor



TNF-R55, TNFAR, TNFR1,
superfamily, member 1A



TNFR60


TNFRSF1B
CD120b, p75, TNF-R-II,
Tumor necrosis factor receptor



TNF-R75, TNFBR, TNFR2,
superfamily, member 1B



TNFR80


TNFRSF4
ACT35, CD134, OX40,
Tumor necrosis factor receptor



TXGP1L
superfamily, member 4


TNFRSF8
CD30, D1S166E, KI-1
Tumor necrosis factor receptor




superfamily, member 8


TNFRSF9
4-1BB, CD137, ILA
Tumor necrosis factor receptor




superfamily, member 9


TNFSF10
Apo-2L, CD253, TL2,
Tumor necrosis factor (ligand)



TRAIL
superfamily, member 10


TNFSF11
CD254, ODF, OPGL,
Tumor necrosis factor (ligand)



RANKL, TRANCE
superfamily, member 11


TNFSF13
APRIL, CD256
Tumor necrosis factor (ligand)




superfamily, member 13


TNFSF13B
BAFF, BLYS, CD257,
Tumor necrosis factor (ligand)



TALL-1, TALL1, THANK,
superfamily, member 13b



TNFSF20


TNFSF4
CD252, gp34, OX-40L,
Tumor necrosis factor (ligand)



TXGP1
superfamily, member 4


TNFSF8
CD153, CD30LG
Tumor necrosis factor (ligand)




superfamily, member 8


TNK2
ACK, ACK1, p21cdc42Hs
Tyrosine kinase, non-receptor, 2


TOP2A
TOP2
Topoisomerase (DNA) II alpha 170 kDa


TP53
LFS1, p53
Tumor protein p53


TP53BP2
53BP2, ASPP2, PPP1R13A
Tumor protein p53 binding protein 2


TPD52
D52, hD52, N8L
Tumor protein D52


TPI1

Triosephosphate isomerase 1


TPM1
C15orf13, CMH3
Tropomyosin 1 (alpha)


TPM2
AMCD1, DA1, NEM4
Tropomyosin 2 (beta)


TPX2
C20orf1, C20orf2, DIL-2,
TPX2, microtubule-associated



p100


TRAF1
EBI6
TNF receptor-associated factor 1


TRAF2
TRAP3
TNF receptor-associated factor 2


TRAF4
CART1, MLN62, RNF83
TNF receptor-associated factor 4


TRIM25
EFP, RNF147, ZNF147
Tripartite motif containing 25


TRIP4
HsT17391, ZC2HC5
Thyroid hormone receptor interactor 4


TRO
KIAA1114, MAGE-D3,
Trophinin



MAGED3


TSG101
TSG10, VPS23
Tumor susceptibility 101


TSPAN8
CO-029, TM4SF3
Tetraspanin 8


TSPO
BZRP, DBI, IBP, MBR,
Translocator protein (18 kDa)



mDRC, PBR, pk18, PKBS


TTR
CTS, CTS1, HsT2651,
Transthyretin



PALB


TUSC2
C3orf11, FUS1, PAP,
Tumor suppressor candidate 2



PDAP2


TWIST1
ACS3, bHLHa38, BPES2,
Twist family bHLH transcription factor



BPES3, CRS, CRS1, H-
1



twist, SCS, TWIST


TXLNA
DKFZp451J0118
Taxilin alpha


TYMP
ECGF1, MNGIE
Thymidine phosphorylase


TYMS
HsT422, TMS, TS, Tsase
Thymidylate synthetase


TYRO3
Brt, Dtk, RSE, Sky, Tif
TYRO3 protein tyrosine kinase


UBA1
A1S9T, CFAP124, GXP1,
Ubiquitin-like modifier activating



POC20, UBE1, UBE1X
enzyme 1


UBE2C
UBCH10
Ubiquitin-conjugating enzyme E2C


UBE2I
UBC9
Ubiquitin-conjugating enzyme E2I


UBE2N
MGC8489, UBC13, UbcH-
Ubiquitin-conjugating enzyme E2N



ben


UGT1A10
UGT1J
UDP glucuronosyltransferase 1 family,




polypeptide A10


UGT1A3
UGT1C
UDP glucuronosyltransferase 1 family,




polypeptide A3


UGT1A4
HUG-BR2, UGT1D
UDP glucuronosyltransferase 1 family,




polypeptide A4


UGT1A8
UGT1H
UDP glucuronosyltransferase 1 family,




polypeptide A8


UGT1A9
HLUGP4, LUGP4, UGT1AI
UDP glucuronosyltransferase 1 family,




polypeptide A9


USH1C
AIE-75, DFNB18,
Usher syndrome 1C (autosomal



harmonin, NY-CO-37, NY-
recessive, severe)



CO-38, PDZ-73, PDZ73,



PDZD7C


VAMP3
CEB
Vesicle-associated membrane protein 3


VCAM1
CD106
Vascular cell adhesion molecule 1


VEGFA
VEGF, VEGF-A, VPF
Vascular endothelial growth factor A


VEGFB
VEGFL, VRF
Vascular endothelial growth factor B


VEGFC
VRP
Vascular endothelial growth factor C


VHL
VHL1
Von Hippel-Lindau tumor suppressor,




E3 ubiquitin protein ligase


VIL1
D2S1471, VIL
Villin 1


VIP

Vasoactive intestinal peptide


VTN
VN
Vitronectin


VWF
F8VWF
Von Willebrand factor


WEE1

WEE1 G2 checkpoint kinase


WFDC2
dJ461P17.6, EDDM4, HE4,
WAP four-disulfide core domain 2



WAP5


WISP1
CCN4
WNT1 inducible signaling pathway




protein 1


WNT1
INTI
Wingless-type MMTV integration site




family, member 1


WNT2
INT1L1, IRP
Wingless-type MMTV integration site




family member 2


WRN
RECQ3, RECQL2
Werner syndrome, RecQ helicase-like


WT1
AWT1, GUD, WAGR,
Wilms tumor 1



WIT-2


XBP1
XBP2
X-box binding protein 1


XIAP
API3, BIRC4, hILP
X-linked inhibitor of apoptosis


XPA
XP1, XPAC
Xeroderma pigmentosum,




complementation group A


XPC
RAD4, XPCC
Xeroderma pigmentosum,




complementation group C


XRCC2

X-ray repair complementing defective




repair in Chinese hamster cells 2


XRCC3

X-ray repair complementing defective




repair in Chinese hamster cells 3


XRCC4

X-ray repair complementing defective




repair in Chinese hamster cells 4


XRCC5
KARP-1, KU80, Ku86,
X-ray repair complementing defective



KUB2
repair in Chinese hamster cells 5




(double-strand-break rejoining)


XRCC6
D22S671, D22S731, G22P1,
X-ray repair complementing defective



KU70, ML8
repair in Chinese hamster cells 6


YBX1
BP-8, CSDA2, CSDB,
Y box binding protein 1



DBPB, MDR-NF1, NSEP-1,



NSEP1, YB-1, YB1


YWHAB
YWHAA
Tyrosine 3-monooxygenase/tryptophan




5-monooxygenase activation protein,




beta


YWHAE
FLJ45465
Tyrosine 3-monooxygenase/tryptophan




5-monooxygenase activation protein,




epsilon


YWHAH
YWHA1
Tyrosine 3-monooxygenase/tryptophan




5-monooxygenase activation protein,




eta


ZBTB16
PLZF, ZNF145
Zinc finger and BTB domain




containing 16


ZMAT3
FLJ12296, MGC10613,
Zinc finger, matrin-type 3



PAG608, WIG-1, WIG1









In one embodiment, the biomarker is MYC. In one embodiment, the measurable aspect of MYC is its expression status. In one embodiment, the biomarker is overexpression of MYC.


Thus, in certain aspects of the disclosure, the biomarker is MYC which is differentially present in a subject of one phenotypic status, e.g., a patient having cancer, e.g., hepatocellular carcinoma (HCC), glioblastomas (GBM), lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, or colorectal cancer, as compared with another phenotypic status, e.g., a normal undiseased subject or a patient having cancer without overexpression MYC.


Biomarker standards can be predetermined, determined concurrently, or determined after a biological sample is obtained from the subject. Biomarker standards for use with the methods described herein can, for example, include data from samples from subjects without cancer; data from samples from subjects with cancer, e.g., GBM, that is not a progressive, recurrent, and/or metastatic cancer; and data from samples from subjects with cancer, e.g., GBM, that is a progressive, recurrent, and/or metastatic cancer. Comparisons can be made to establish predetermined threshold biomarker standards for differenct classes of subjects, e.g., diseased vs. non-diseased subjects. The standards can be run in the same assay or can be known standards from a previous assay.


In one embodiment, the biomarker is MCL1. In one embodiment, the measurable aspect of MCL1 is its expression status. In one embodiment, the biomarker is overexpression of MCL1.


A biomarker is differentially present between different phenotypic status groups if the mean or median expression or mutation levels of the biomarker is calculated to be different, i.e., higher or lower, between the groups. Thus, biomarkers provide an indication that a subject, e.g., a cancer patient, belongs to one phenotypic status or another.


Thus, in certain aspects of the disclosure, the biomarker is MCL1 which is differentially present, i.e., overexpressed, in a subject of one phenotypic status, e.g., a patient having cancer, e.g., hepatocellular carcinoma (HCC), glioblastomas (GBM), lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, colorectal cancer, medulloblastoma, or general brain tumors, as compared with another phenotypic status, e.g., an undiseased patient or a cancer patient without overexpression MCL1.


In addition to individual biological compounds, e.g., MYC or MCL1, the term “biomarker” as used herein is meant to include groups, sets, or arrays of multiple biological compounds. For example, the combination of MYC and MCL1 may comprise a biomarker. The term “biomarker” may comprise one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, twenty five, thirty, or more, biological compounds.


The determination of the expression level or mutation status of a biomarker in a patient can be performed using any of the many methods known in the art. Any method known in the art for quantitating specific proteins and/or detecting MYC and/or MCL1 expression, or the expression or mutation levels of any other biomarker in a patient or a biological sample may be used in the methods of the disclosure. Examples include, but are not limited to, PCR (polymerase chain reaction), or RT-PCR, Northern blot, Western blot, ELISA (enzyme linked immunosorbent assay), RIA (radioimmunoassay), gene chip analysis of RNA expression, immunohistochemistry or immunofluorescence. See, e.g., Slagle et al. Cancer 83:1401 (1998). Certain embodiments of the disclosure include methods wherein biomarker RNA expression (transcription) is determined. Other embodiments of the disclosure include methods wherein protein expression in the biological sample is determined. See, for example, Harlow et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., (1988) and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York 3rd Edition, (1995). For northern blot or RT-PCR analysis, RNA is isolated from the tumor tissue sample using RNAse free techniques. Such techniques are commonly known in the art.


In one embodiment of the disclosure, a biological sample is obtained from the patient and cells in the biopsy are assayed for determination of biomarker expression or mutation status.


In one embodiment of the disclosure, PET imaging is used to determine biomarker expression.


In another embodiment of the disclosure, Northern blot analysis of biomarker transcription in a tumor cell sample is performed. Northern analysis is a standard method for detection and/or quantitation of mRNA levels in a sample. Initially, RNA is isolated from a sample to be assayed using Northern blot analysis. In the analysis, the RNA samples are first separated by size via electrophoresis in an agarose gel under denaturing conditions. The RNA is then transferred to a membrane, crosslinked and hybridized with a labeled probe. Typically, Northern hybridization involves polymerizing radiolabeled or nonisotopically labeled DNA, in vitro, or generation of oligonucleotides as hybridization probes. Typically, the membrane holding the RNA sample is prehybridized or blocked prior to probe hybridization to prevent the probe from coating the membrane and, thus, to reduce non-specific background signal. After hybridization, typically, unhybridized probe is removed by washing in several changes of buffer. Stringency of the wash and hybridization conditions can be designed, selected and implemented by any practitioner of ordinary skill in the art. Detection is accomplished using detectably labeled probes and a suitable detection method. Radiolabeled and non-radiolabled probes and their use are well known in the art. The presence and or relative levels of expression of the biomarker being assayed can be quantified using, for example, densitometry.


In another embodiment of the disclosure, biomarker expression and/or mutation status is determined using RT-PCR. RT-PCR allows detection of the progress of a PCR amplification of a target gene in real time. Design of the primers and probes required to detect expression and/or mutation status of a biomarker of the disclosure is within the skill of a practitioner of ordinary skill in the art. RT-PCR can be used to determine the level of RNA encoding a biomarker of the disclosure in a tumor tissue sample. In an embodiment of the disclosure, RNA from the biological sample is isolated, under RNAse free conditions, than converted to DNA by treatment with reverse transcriptase. Methods for reverse transcriptase conversion of RNA to DNA are well known in the art. A description of PCR is provided in the following references: Mullis et al., Cold Spring Harbor Symp. Quant. Biol. 51:263 (1986); EP 50,424; EP 84,796; EP 258,017; EP 237,362; EP 201,184; U.S. Pat. Nos. 4,683,202; 4,582,788; 4,683,194.


RT-PCR probes depend on the 5′-3′ nuclease activity of the DNA polymerase used for PCR to hydrolyze an oligonucleotide that is hybridized to the target amplicon (biomarker gene). RT-PCR probes are oligonucleotides that have a fluorescent reporter dye attached to the 5, end and a quencher moiety coupled to the 3′ end (or vice versa). These probes are designed to hybridize to an internal region of a PCR product. In the unhybridized state, the proximity of the fluor and the quench molecules prevents the detection of fluorescent signal from the probe. During PCR amplification, when the polymerase replicates a template on which an RT-PCR probe is bound, the 5′-3′ nuclease activity of the polymerase cleaves the probe. This decouples the fluorescent and quenching dyes and FRET no longer occurs. Thus, fluorescence increases in each cycle, in a manner proportional to the amount of probe cleavage. Fluorescence signal emitted from the reaction can be measured or followed over time using equipment which is commercially available using routine and conventional techniques.


In another embodiment of the disclosure, expression of proteins encoded by biomarkers are detected by western blot analysis. A western blot (also known as an immunoblot) is a method for protein detection in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate denatured proteins by mass. The proteins are then transferred out of the gel and onto a membrane (e.g., nitrocellulose or polyvinylidene fluoride (PVDF)), where they are detected using a primary antibodythat specifically bind to the protein. The bound antibody can then detected by a secondary antibody that is conjugated with a detectable label (e.g., biotin, horseradish peroxidase or alkaline phosphatase). Detection of the secondary label signal indicates the presence of the protein.


In another embodiment of the disclosure, the expression of a protein encoded by a biomarker is detected by enzyme-linked immunosorbent assay (ELISA). In one embodiment of the disclosure, “sandwich ELISA” comprises coating a plate with a capture antibody; adding sample wherein any antigen present binds to the capture antibody; adding a detecting antibody which also binds the antigen; adding an enzyme-linked secondary antibody which binds to detecting antibody; and adding substrate which is converted by an enzyme on the secondary antibody to a detectable form. Detection of the signal from the secondary antibody indicates presence of the biomarker antigen protein.


In another embodiment of the disclosure, the expression of a biomarker is evaluated by use of a gene chip or microarray. Such techniques are within ordinary skill held in the art.


VI. Definitions

The disclosure provides various therapeutic methods, kits, and pharmaceutical compositions comprising TG02. The term “TG02” as used herein refers to (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene in any crystalline or amorphous form as a free base or as a pharmaceutically acceptable salt or solvate. In one embodiment, TG02 refers to the free base of (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene. In another embodiment, TG02 refers to a pharmaceutically acceptable salt of (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene. A pharmaceutically acceptable salt of TG02 can be prepared during the final isolation and purification of TG02 or separately by reacting TG02 with pharmaceutically acceptable acids. Examples of acids which can be employed to form pharmaceutically acceptable salts include inorganic acids such as nitric, boric, hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Nonlimiting examples of salts of TG02 include, but are not limited to, the hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, 2-hydroxyethansulfonate, phosphate, hydrogen phosphate, acetate, adipate, alginate, aspartate, benzoate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerolphsphate, hemisulfate, heptanoate, hexanoate, formate, succinate, fumarate, maleate, ascorbate, isethionate, salicylate, methanesulfonate, mesitylenesulfonate, naphthylenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproprionate, picrate, pivalate, propionate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, paratoluenesulfonate, undecanoate, lactate, citrate, tartrate, gluconate, methanesulfonate, ethanedisulfonate, benzene sulfonate, and p-toluenesulfonate salts.


In another embodiment, TG02 refers to the citrate salt of (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene. This is referred to as TG02 citrate or (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6). 1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene—citric acid.


The term “biological sample” as used herein refers any tissue or fluid from a patient that is suitable for detecting a biomarker, such as MYC and/or MCL1 expression status. Examples of useful biological samples include, but are not limited to, biopsied tissues and/or cells, e.g., solid tumor, lymph gland, inflamed tissue, tissue and/or cells involved in a condition or disease, blood, plasma, serous fluid, cerebrospinal fluid, saliva, urine, lymph, cerebral spinal fluid, and the like. Other suitable biological samples will be familiar to those of ordinary skill in the relevant arts. A biological sample can be analyzed for biomarker expression and/or mutation using any technique known in the art and can be obtained using techniques that are well within the scope of ordinary knowledge of a clinical practioner. In one embodiment of the disclosure, the biological sample comprises blood cells.


The terms “a”, “an”, “the”, and similar referents in the context of describing the disclosure (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated. Recitation of ranges of values herein merely are intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The use of any and all examples, or exemplary language, e.g., “such as,” provided herein, is intended to better illustrate the disclosure and is not a limitation on the scope of the disclosure unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.


The term “about,” as used herein, includes the recited number ±10%. Thus, “about 10” means 9 to 11.


As used herein, the terms “treat,” “treating,” “treatment,” and the like refer to eliminating, reducing, or ameliorating a disease or condition, and/or symptoms associated therewith. Although not precluded, treating a disease or condition does not require that the disease, condition, or symptoms associated therewith be completely eliminated. However, in one embodiment, administration of TG02 and/or an immune checkpoint inhibitor and/or COX-2 inhibitor leads to complete remission of the cancer.


The term “therapeutically effective amount,” as used herein, refers to that amount of the therapeutic agent sufficient to result in amelioration of one or more symptoms of a disorder, or prevent advancement of a disorder, or cause regression of the disorder. For example, with respect to the treatment of cancer, in one embodiment, a therapeutically effective amount will refer to the amount of a therapeutic agent that causes a therapeutic response, e.g., normalization of blood counts, decrease in the rate of tumor growth, decrease in tumor mass, decrease in the number of metastases, increase in time to tumor progression, and/or increase patient survival time by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, or more.


The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable vehicle” encompasses any of the standard pharmaceutical carriers, solvents, surfactants, or vehicles. Suitable pharmaceutically acceptable vehicles include aqueous vehicles and nonaqueous vehicles. Standard pharmaceutical carriers and their formulations are described in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 19th ed. 1995.


The term “container” means any receptacle and closure therefore suitable for storing, shipping, dispensing, and/or handling a pharmaceutical product.


The term “insert” means information accompanying a pharmaceutical product that provides a description of how to administer the product, along with the safety and efficacy data required to allow the physician, pharmacist, and patient to make an informed decision regarding use of the product. The package insert generally is regarded as the “label” for a pharmaceutical product.


“Concurrent administration,” “administered in combination,” “simultaneous administration,” and similar phrases mean that two or more agents are administered concurrently to the subject being treated. By “concurrently,” it is meant that each agent is administered either simultaneously or sequentially in any order at different points in time. However, if not administered simultaneously, it is meant that they are administered to an individual in a sequence and sufficiently close in time so as to provide the desired therapeutic effect and can act in concert. For example, TG02 can be administered at the same time or sequentially in any order at different points in time as the immune checkpoint inhibitor and/or the COX-2 inhibitor and/or the optional therapeutic agent. TG02 and the immune checkpoint inhibitor and/or the COX-2 inhibitor and/or the optional therapeutic agent can be administered separately, in any appropriate form and by any suitable route. When TG02 and the immune checkpoint inhibitor and/or the COX-2 inhibitor and/or the optional therapeutic agent are not administered concurrently, it is understood that they can be administered in any order to a patient in need thereof. For example, TG02 can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the immune checkpoint inhibitor and/or COX-2 inhibitor, to an individual in need thereof. In various embodiments, TG02 and the immune checkpoint inhibitor and/or COX-2 inhibitor are administered 1 minute apart, 10 minutes apart, 30 minutes apart, less than 1 hour apart, 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart or no more than 48 hours apart. In one embodiment, the components of the combination therapies are administered at about 1 minute to about 24 hours apart. In one embodiument, TG02 is administered 3-7 days prior to the day the immune checkpoint inhibitor is administered. In another embodiment, TG02 is also administered on the day the immune checkpoint inhibitor is administered and continues to be administered until disease progression or TG02 therapy is no longer beneficial.


EXAMPLES
Example 1

This study is being done to compare progression-free or overall survival using pembrolizumab (p) or nivolumab (n) to p or n in combination with TG02 for participants with cancer who are untreated or have progressed after prior therapy and who have been selected for overexpressed MYC and/or MCL1 status. Participants will be randomized to receive either standard anti-PD-1 therapy plus placebo or standard anti-PD-1 therapy plus TG02.


Primary Outcome Measures: Progression-free-survival (PFS) and/or Overall survival (OS)


Secondary Outcome Measures: Overall response rate (ORR) and/or Response Duration


Eligibility






    • Ages Eligible for Study: Generally—18 Years and older
      • For medulloblastoma patients—6 months or older

    • Genders Eligible for Study: Both


      Inclusion Criteria:





Histologically or cytologically confirmed diagnosis of cancer not amenable to local therapy


Must consent to allow correlative studies; must provide a newly obtained tissue/biopsy specimen (or specimen obtained within 60 days of consenting)


Radiographically measurable disease


Eastern Cooperative Oncology Group Performance Status of 0 or 1


Patient has disease with overexpressed MYC and/or MCL1


Exclusion Criteria:


Chemotherapy, radiation therapy, or biological therapy within four weeks prior to the first dose of study drug, or not recovered from the AEs due to cancer therapies administered more than four weeks earlier


Participating or has participated in a study of an investigational agent or using an investigational device within 30 days of the first dose of study drug


Expected to require any other form of systemic or localized antineoplastic therapy while on study


Chronic systemic steroid therapy within two weeks before the planned date for first dose randomized treatment or on any other form of immunosuppressive medication


Known history of any other than the current malignancy excepting adequately treated basal or squamous cell carcinoma of the skin, superficial bladder cancer, in situ cervical cancer, breast cancer, or other in situ cancers


Known active central nervous system (CNS) metastases and/or carcinomatous meningitis


Active autoimmune disease or a documented history of autoimmune disease or syndrome that requires systemic steroids or immunosuppressive agents


Prior treatment with any other anti-programmed cell death (PD) agent


Active infection requiring systemic therapy


Known history of Human Immunodeficiency Virus (HIV)


Active Hepatitis B or Hepatitis C


Regular user (including recreational use of) illicit drugs or had a recent history (within the last year) of substance abuse (including alcohol)


Pregnant or breastfeeding, or expecting to conceive or father children within the projected duration of the study.


Protocols:


A first group of patients receive 2-10 mg/kg pembrolizumab (or flat dose equivalent) administered by intravenous infusion every three weeks and TG02 administered orally at 100, 200 or 300 mg once daily until disease progression or is no longer beneficial. TG02 administration is started 3-7 days prior to initiating pembrolizumab therapy, continues on the day of pembrolizumab administration, and continues until disease progression or until TG02 therapy is no longer beneficial. The control patients receive 2-10 mg/kg pembrolizumab (or flat dose equivalent) administered by intravenous infusion every three weeks.


A second group of patients receive 3 mg/kg nivolumab administered over 60 minutes by intravenous infusion every 2 weeks and TG02 administered orally at 100, 200, or 300 mg once daily. TG02 administration is started 3-7 days prior to initiating nivolumab therapy, continues on the day of nivolumab administration, and continues until disease progression or until TG02 therapy is no longer beneficial. The control patients receive 3 mg/kg nivolumab administered over 60 minutes by intravenous infusion every 2 weeks.


Results:


TG02 in combination with pembrolizumab or nivolumab results in better antitumor clinical activity than the immune checkpoint inhibitors alone in patients whose tumors overexpress MYC and/or MCL1. Unexpected objective responses are obtained associated with lack of tumor progression and extension of long term survival compared to historical controls using (the antibody) alone. In one embodiment, patients receiving TG02 and the immune checkpoint inhibitor achieve an extension of time to progression (or progression-free survival) of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months. In another embodiment, at least some of the patients receiving TG02 and the immune checkpoint inhibitor achieve an extension of duration of response of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months.


Example 2

Open label Phase 2 study assessing the combination of checkpoint blockade immunotherapy and TG02 in patients relapsing from or refractory to standard anti-PD-1 therapy (p) or (n).

    • Primary endpoint: ORR
    • Secondary endpoints: PFS, OS, Duration of Response, Safety


      Inclusion Criteria:


Histologically confirmed diagnosis of cancer not amenable to local therapy


Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1


At least one measurable lesion


Adequate organ function


Prior therapy with an anti-PD-1 or anti-PD-L1 antibody


Patient has disease with overexpressed MYC and/or MCL1


Exclusion Criteria:


Chemotherapy, targeted small molecule therapy, radiotherapy, or biological cancer therapy (including monoclonal antibodies) within 4 weeks prior to the first dose of trial treatment, or not recovered (<=Grade 1 or baseline) from adverse events due to a previously administered agent.


Expected to require any other form of systemic or localized antineoplastic therapy while in study.


Known active central nervous system (CNS) metastases and/or carcinomatous meningitis.


Documented history of clinically severe autoimmune disease, or a syndrome that requires systemic steroids or immunosuppressive agents.


Receiving systemic steroid therapy or any other form of immunosuppressive therapy within 1 week prior to the first dose of study treatment.


Received a live vaccine within 4 weeks prior to the first dose of trial treatment.


History or evidence of active pneumonitis.


Human immunodeficiency virus (HIV)-positive.


Active Hepatitis B or C.


Pregnant, breastfeeding, or expecting to conceive or father children within the projected duration of the trial treatment through 120 days after the last dose of study medication.


Dosing Protocol:









TABLE 1







TG02 + Checkpoint Inhibitor Combination Dosing & Schedules











Every
Every
Every



2 weeks
3 weeks
4 weeks
















Pembrolizumab 2 mg/kg
X
X




Pembrolizumab 10 mg/kg
X
X



Pembrolizumab 200 mg
X
X



Pembrolizumab 300 mg
X
X



Nivolumab 3 mg/kg
X
X
X



Nivolumab 1 mg/kg
X
X
X



Pidilizumab 3 mg/kg
X
X
X



Pidilizumab 1.5 mg/kg
X
X
X



STI-110 2 mg/kg
X
X
X



STI-110 2 mg/kg
X
X
X



Durvalumab 10 mg/kg
X
X



Durvalumab 2 mg/kg
X
X



Durvalumab 15, g/kg

X
X



Avelumab 1200 mg
X
X
X



Avelumab 10 mg/kg
X
X
X



Avelumab 5 mg/kg
X
X
X



Atezolizumab 1200 mg

X



STI-1014 10 mg/kg
X
X
X



STI-1014 15 mg/kg
X
X
X







*TG02 is dosed weekly (50-400 mg) starting at least 5 days prior to initiating checkpoint inhibitor therapy and continuing until disease progression or investigator decision







Results


Combining TG02 with at least one checkpoint inhibitor in patients with overexpressed MYC and/or MCL1 tumors reverses immune evasion and induces clinically relevant responses in patients previously nonresponding to or failing checkpoint inhibitor therapy or de novo cancer patients. Unexpected objective responses are obtained associated with lack of tumor progression and extension of long term survival compared to historical controls using (the antibody) alone. In one embodiment, patients receiving TG02 and at least one immune checkpoint inhibitor achieve an extension of time to progression (or progression-free survival) of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months. In another embodiment, at least some of the patients receiving TG02 and at least one immune checkpoint inhibitor achieve an extension of duration of response of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months.


Example 3
Placebo-Controlled, Randomized Phase 2 Study of Pembrolizumab+TG02 vs. Pembrolizumab+Placebo in Participants with Previously-Treated Locally Advanced Unresectable or Metastatic Colorectal Cancer Exhibiting Overexpressed MYC and/or MCL1 Status





    • Primary Endpoint: PFS

    • Secondary Endpoint: ORR, Duration of Response


      Inclusion Criteria:





Histologically-proven locally advanced unresectable or metastatic high colorectal carcinoma


Previously treated with at least two lines of approved standard therapies, which must include fluoropyrimidine, oxaliplatin, irinotecan, bevacizumab, and cetuximab or panitumumab


Eastern Cooperative Oncology Group performance status of 0 or 1


Patient has disease with overexpressed MYC and/or MCL1


Life expectancy of greater than 3 months


At least one measureable lesion


Female participants of childbearing potential should be willing to use 2 methods of birth control or be surgically sterile, or abstain from heterosexual activity for the course of the study through 120 days after the last dose of study medication


Male participants should agree to use an adequate method of contraception starting with the first dose of study therapy through 120 days after the last dose of study medication


Adequate organ function


Exclusion Criteria:


Currently participating in another study and receiving trial treatment, participated in a study of an investigational agent and received trial treatment within 4 weeks of the first dose of medication in this study, or used an investigational device within 4 weeks of the first dose of medication in this study


Active autoimmune disease that has required systemic treatment in past 2 years


Diagnosis of immunodeficiency or receiving systemic steroid therapy or any other form of immunosuppressive therapy within 7 days prior to the first dose of study medication


Known active central nervous system (CNS) metastases and/or carcinomatous meningitis


Prior monoclonal antibody (mAb), chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks prior to study Day 1 or not recovered (i.e., Grade 1 or at baseline) from adverse events due to a previously administered agent


Prior therapy with an anti-programmed cell death (PD)-1, anti-PD-L1, or anti-PD-L2 agent, or participant has previously participated in Merck pembrolizumab (MK-3475) clinical trial


Known additional malignancy that is progressing or requires active treatment with the exception of basal cell carcinoma of the skin or squamous cell carcinoma of the skin that has undergone potentially curative therapy, or in situ cervical cancer


Received a live vaccine within 30 days of planned start of study medication


Known history of human immunodeficiency virus (HIV)


Known active Hepatitis B or C


Known history or any evidence of interstitial lung disease or active, non-infectious pneumonitis


Active infection requiring systemic therapy


Known psychiatric or substance abuse disorders that would interfere with cooperation with the requirements of the trial


Pregnant or breastfeeding, or expecting to conceive or father children within the projected duration of the trial, starting with the screening visit through 120 days after the last dose of trial medication


Dosing Protocol:


Patients receive 2-10 mg/kg pembrolizumab administered by intravenous infusion every three weeks and TG02 administered orally at 1, 2 or 3 mg/kg 3-7 days prior to pembrolizumab administration, on the day of pembrolizumab administration, and continuously thereafter until disease progression or until it is no longer beneficial. The control patients receive 2 mg/kg pembrolizumab administered by intravenous infusion every three weeks.


Results:


When used in patients with tumors overexpressing MYC and/or MCL1, TG02 combined with pembrolizumab provides superior clinical activity than pembrolizumab alone in the same patients. Unexpected objective responses are obtained in patients associated with lack of tumor progression and extension of long term survival compared to historical controls using (the antibody) alone. In one embodiment, patients receiving TG02 and pembrolizumab achieve an extension of time to progression (or progression-free survival) of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months. In another embodiment, at least some of the patients receiving TG02 and pembrolizumab achieve an extension of duration of response of at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months or at least 12 months.


Example 4
TG02 in Combination with Carfilzomib in Carfilzomib (CFZ) Refractory Multiple Myeloma (MM) Patients

Methods


An open-label Phase 1b study enrolled MM patients that previously received ≥2 lines of therapy. The primary objective was to determine the maximum tolerated dose (MTD) of TG02 in combination with carfilzomib (TG02/CFZ). Secondary objectives included anti-tumor activity and safety. TG02 was administered once daily on days 1, 4, 8, 11, 15, 18 of a 28-day schedule (BIW). The TG02 starting dose was 150 mg. TG02 dosing was escalated in 50 mg increments up to 300 mg. CFZ was dosed according to the Prescribing Information. Responses were assessed using standard criteria.


Results


Fourteen patients were enrolled for dose escalation and 10 patients were enrolled for the MTD cohort expansion. Patients were heavily pretreated: median 6 previous treatments [min 3; max 15] and 92% patients received CFZ in a prior regimen. The best response to previous therapy was progressive disease in 46% patients. The MTD was 250 mg TG02 combined with CFZ. Two dose-limiting toxicities were observed (including Grade (Gr) 4 sepsis and Gr 4 neutropenia), both on the 300 mg cohort. The most common drug-related adverse events (AEs) were diarrhea (Gr 1-2: 71% Gr 3: 17%), nausea (Gr 1-2: 79%), vomiting (Gr 1-2: 50%), fatigue (Gr 1-2: 38%, Gr 3: 4%), anorexia (Gr 1: 21%), anemia (Gr 1-2: 4%, Gr 3: 17%) and thrombocytopenia (Gr 3: 8%, Gr 4: 13%). Six patients (25%) discontinued treatment due to an AE. Serious AEs occurred in 50% patients; only acute renal failure and febrile neutropenia occurred in >1 pt (8% each). The severity of AEs was similar to single agent TG02. The incidence of diarrhea was increased in the TG02/CFZ administration (88% vs 67%) but the incidence of other AEs was similar to single agent TG02. Fourteen patients administered TG02 at the MTD were evaluable for response. The overall response rate (≥PR) was 27%; the clinical benefit rate (≥MR) was 45% (1 very good partial response, 2 partial response and 2 minimal response). All responders (MR or better) were CFZ-refractory in a previous treatment regimen. Durable stable disease was observed in 27% patients.


Conclusion


The safety profile of TG02 BIW/CFZ was similar to that of TG02 alone. The most common drug-related AEs were diarrhea, nausea, and vomiting; grade 4 AEs were infrequent. Objective responses were observed in CFZ-refractory patients.


Example 5
TG02 Activity in Glioma Cells and Allograft Model

Several standard glioblastoma multiforme (GBM) cell lines and one stem cell line expressing O6-methylguanine DNA methyltransferase (MGMT) were treated with TG02, temozolomiode (TMZ), or the combination of TG02 and TMZ in a 72-hour cell proliferation assay. See FIGS. 1-3. TG02, TMZ, and the TG02+TMZ combination was also tested in cell lines without MGMT expression. See FIGS. 4-7. The cells were seeded onto 12-well plates and treated with 50 nM of TG02, 100 μM of TMZ or TG02+TMZ for 72 hr. Cell viability was determined by cell counting.


The cytotoxic effect TG02, TMZ, and the combination of TG02 and TMZ was examined via the colony formation assay in GSC923 and U251 cells. See FIGS. 8 and 9.


Pulmonary arterial endothelial cells and human astrocytes were tested with 50 nM of TG02, 100 μM of TMZ, or TG02+TMZ for 72 hr. The cells were then changed to normal medium and cultured for another 7 days. Cell viability was determined by cell counting. See FIGS. 10 and 11.


GSC923 (FIGS. 12 and 13) and U251 (FIGS. 14 and 15) cells were exposed to various concentrations of TG02, TMZ, and TG02+TMZ for 72 hr, and cell viability was examined via cell counting. The synergistic effect of TG02+TMZ was determined by Combination Index (CI). CI values were calculated by COMPUSYN software and shown Table 2 for GSC923 cells and Table 3 for U251 cells. CI<1 is a synergistic, CI=1 is an additive, and CI>1 is an antagonistic effect of the two compounds combined.












TABLE 2







Fraction Affected (Fa)
CI value



















0.25
0.077



0.5
0.136



0.75
0.242



0.90
0.430




















TABLE 3







Fraction Affected (Fa)
CI value



















0.25
0.029



0.5
0.104



0.75
0.376



0.90
1.333











FIG. 16 shows a schematic illustration of drug administration in an mouse glioma GL261 cell allograft model. Mouse glioma GL261 cells were injected stereotactically into the striatum of female C57BL/6 albino mice (n=5-7 per cohort) followed by vehicle, TG02, TMZ, and TG02+TMZ combination treatment. A median overall survival of 24, 24.5, 27.5, and 32 days, respectively was observed. See FIG. 17. The result was analyzed using Logrank test for trend in GraphPad Prism software (Chi square=9.063, df=1, P value=0.0026**). The turmor burden was determined by bioluminescence imaging (BLI) taken using the PerkinElmer IVIS® Spectrum. The intensity of BLI was calculated and normalized to the initial intensity at day 5. See FIG. 18.


Example 6
TG02 Activity in Hepatocellular Carcinoma (HCC) Cells and Xenograft Model

The effect of TG02 on MYC expression in HCC cell lines was investigated. Five HCC cell lines with varying levels of MYC expression were treated with 0.5 TG02 for 24 hours and MYC expression was evaluated by western blot. See FIG. 19. MYC expression levels were decreased by TG02 treatment in HepG2, SNU398 and HUH-1 cell lines but not in the JHH-5 line. Hep3B cells did not express MYC.


Eight HCC cell lines with either high or low MYC expression were then treated with TG02 in vitro. TG02 treatment resulted in inhibition of cell proliferation in all HCC cell lines tested. TG02 is selectively more potent in the cells lines with high levels of MYC expression as compared to those cells with low levels of MYC expression, with mean IC50 values of 84 nM and 524 nM, respectively. See FIG. 20.


Inhibition of MYC expression was also measured in vivo. HepG2 hepatocellular carcinoma xenografts was grown orthotopically in Balb/c nude mice. TG02 or vehicle was given orally at 50 mg/kg to 5 mice each; tumors were collected 8 hours post-treatment and MYC protein expression levels were measured by western blot. MYC expression was observed in each of the control tumors. In the TG02 treatment group, MYC expression levels were decreased in 4 of 5 animals, with substantial MYC depletion in two animals and partial reduction in two other animals. See FIG. 21.


The therapeutic efficacy of TG02 as a single agent or in combination with sorafenib in the treatment of orthotopic HepG2 human liver cancer xenograft model was evaluated in BALB/c nude mice. On Day 19 post-inoculation, mice were randomized into treatment groups based on baseline serum AFP levels which trace tumor volume in the liver. TG02 was given orally twice weekly at 50 mg/kg and reduced to 40 mg/kg. Sorafenib was given orally daily at 15 mg/kg. TG02 as a single agent had a modest effect on tumor volume. TG02 combined with sorafenib led to significant anti-tumor activity. See FIG. 22.


Example 7
TG02-Mediated CDK9 Inhibition

A Tet-off transgenic mouse model of MYC-induced T cell acute lymphoblastic leukemia (MYC T-ALL) which over-expresses and is dependent on MYC, was treated with TG02 at 100 or 500 μM.


MYC is a transcription factor that regulates the expression of a multitude of gene products involved in cell proliferation, growth, differentiation and apoptosis. The MYC gene is genetically activated and over-expressed in many human cancers and this over-expression has been causally linked to tumorigenesis, driving malignant growth and immune evasion. See, e.g., Alvarez-Fernandez et al., Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 19:2677-2687 (2013); Carter et al., Blood 105:4043-4050 (2005); Casey et al., Science 352:227-231 (2016); Hannah, A. L., Curr. Mol. Med. 5:625-642 (2005); and Parcells et al., Stem Cells Day. Ohio 24:1174-1184 (2006).


As shown in FIGS. 23-26, when MYC was turned “on,” PD-L1 was expressed as detected by RT-PCR (t=0 hr). But when MYC was turned “off”, both CD47 and PD-L1 expression was significantly reduced in a time dependent manner. TG02 led to a time- and dose-dependent down-regulation of both PD-L1 and CD47 mRNA expression in MYC T-ALL cells (FIGS. 23 and 24, respectively) and both BCL-xL and MYC expression were down-regulated (FIGS. 25 and 26, respectively). Reduced expression of both CD47 and PD-L1 on tumor cells may result in reduced immune evasion and increased tumor cell death.


Example 8
TG02 in Combination with Anti-PD-1

The combination of TG02 and PD-1 mAb (anti-PD-1) was tested in an orthotopic GL261 glioma model. GL261 allografts were established for 3 days and the mice were then randomized into 6 treatment cohorts (n=8) based on the bioluminescent tumor volumes.


Mice were treated with vehicle, TG02 alone (20 or 40 mg/kg), PD-1 mAb alone (500 ug), and the combination of TG02 and anti-PD-1. Median survival times of the mice were 27.5, 26.5, 33, 32, 78 and >95 days, respectively (FIG. 27).


There was a significant survival benefit in TG02 alone at 40 mg/kg (0.009), PD-1 mAb alone (0.003), TG02 20 mg/kg+PD-1 mAb (0.0001), and TG02 40 mg/kg+PD-1 mAb (0.0001) treated-mice compared with the vehicle group.


Example 9
TG02 Induces Cell Death and Synergizes with Radiation in MYC-Driven Glioblastoma

The relationship between TG02 antitumor activity and MYC expression was tested in a panel of patient-derived GBM cell lines (PDCLs). TG02 inhibited six out of twelve PDCLs at an IC50 of less than 0.2 μM. See Table 4.












TABLE 4







Patient-derived GBM lines
TG02 IC50 (μM)



















BT245
0.066



BT360
0.073



BT145
0.052



BT224
0.109



BT187
0.176



BT164
0.178



BT228
0.267



BT286
0.371



BT182
0.416



BT359
0.624



BT139
57.88



BT181
~4828










Downregulation of MYC and Mcl-1 was observed in MYC-amplified BT245 cell line as early as 6 hours, while complete downregulation was seen at 24 hours, which coincided with a significant increase in apoptosis (FIG. 28). The AUC of TG02 induced inhibition of cell viability was calculated in this cell line panel to correlate with MYC expression levels (FIG. 29). TG02 was a more potent inhibitor of PDCLs exhibiting high MYC expression (FIG. 30). In vitro sensitivity (AUC) negatively correlated with MYC expression in GBM cells (P value=0.02)


Example 10

Radiation is an effective treatment for glioblastoma. But tumor resistance and recurrence develops in all patients.


A panel of GBM PDCLs, see Example 9, were chosen for evaluation of the combination of TG02 and radiation therapy for the treatment of glioblastoma (FIG. 31). Cells were treated first with TG02 at increasing concentrations. Within 30 minutes, cells were treated with increasing doses of radiation and cell proliferation was measured 72 hours post-treatment. TG02 alone had anti-proliferative activity in these cell lines. The addition of TG02 augmented the effects radiation in a synergistic manner. The combination of TG02 and radiation exceeds the Bliss predicted model (greater than a 10% change from the Bliss predicted model), demonstrating synergy between TG02 and radiation in multiple PDCLs.


Example 11
TG02 Activity Correlates with MYC Expression in Glioblastoma Cell Lines

In a panel of 26 patient-derived GBM stem cell lines, the activity of TG02 on GBM stem cell proliferation was evaluated (FIG. 32). TG02 was potent in this panel with sixteen cell lines achieving an IC50 of less than 250 nM.


The expression level of CDK9 and downstream markers, including MYC and Mcl-1, were measured to explore whether a correlation exists between protein expression and IC50 values in this GBM panel. High MYC expression was found to correlate with greater sensitivity to TG02 treatment (FIG. 33).


Having now fully described the methods, compounds, and compositions herein, it will be understood by those of skill in the art that the same can be performed within a wide and equivalent range of conditions, formulations, and other parameters without affecting the scope of the methods, compounds, and compositions provided herein or any embodiment thereof. All patents, patent applications and publications cited herein are fully incorporated by reference herein in their entirety.

Claims
  • 1. A method of treating a patient having cancer, the method comprising administering to the patient a therapeutically effective amount of (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene and radiotherapy, wherein MYC overexpression, MCL1 overexpression, or MYC and MCL1 overexpression is differentially present in a biological sample taken from the patient as compared with a biological sample taken from a subject of another phenotypic status.
  • 2. The method of claim 1, wherein (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene is administered to the patient before radiotherapy.
  • 3. The method of claim 1, wherein (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene is administered to the patient after radiotherapy.
  • 4. The method of claim 1, wherein a therapeutically effective amount of (16E)-14-methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene is administered to the patient at the same time as radiotherapy.
  • 5. The method of claim 1, wherein the cancer is selected from the group consisting of acoustic neuroma, acute lymphoblastic leukemia, acute monocytic leukemia, acute promyelocytic leukemia, adenocarcinoma, adult T-cell leukemia/lymphoma, alveolar rhabdomyosarcoma, angiosarcoma, astrocytoma, B-cell chronic lymphocytic leukemia, B-cell prolymphocytic leukemia, B-cell lymphoma, basal cell carcinoma, bladder cancer, blastoma, Burkitt's lymphoma, breast cancer, brain cancer, carcinoma, carcinoma in situ, carcinosarcoma, chondroma, chordoma, choriocarcinoma, craniopharyngioma, cervical cancer, colorectal cancer, diffuse large B-cell lymphoma, embryonal carcinoma, esophageal cancer, fibrosarcoma, follicular lymphoma, follicular thyroid cancer, ganglioneuroma, germ cell tumor, gestational choriocarcinoma, glioblastoma, glioma, hemangioblastoma, head and neck cancer, hematological malignancy, hepatoblastoma, hepatocellular carcinoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, invasive lobular carcinoma, intestinal cancer, kidney cancer, laryngeal cancer, lentigo maligna, lethal midline carcinoma, leukemia, liposarcoma, lung cancer, lymphangiosarcoma, acute lymphocytic leukemia, acute myelogeous leukemia, chronic lymphocytic leukemia, liver cancer, small cell lung cancer, non- small cell lung cancer, medullary carcinoma of the breast, medulloblastoma, melanoma, meningioma, multiple myeloma, myxosarcoma, neurinoma, neuroblastoma, neuroma, nodular melanoma, oligodendroglioma, oral cancer, osteosarcoma, ovarian cancer, Pancoast tumor, papillary thyroid cancer, prostate cancer, pancreatic cancer, pharyngeal cancer, pseudomyxoma periotonei, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, Richter's transformation, rectal cancer, sarcoma, Schwannomatosis, seminoma, skin cancer, small cell carcinoma, somatostatinoma, squamous cell carcinoma, synovial sarcoma, squamous carcinoma, stomach cancer, T-cell lymphoma, testicular cancer, thyroid cancer, uterine cancer, verrucous carcinoma, Waldenstrom's macroglobulinemia, Warthin's tumor, and Wilms' tumor.
  • 6. The method of claim 1, wherein the cancer is selected from the group consisting of astrocytoma, hepatocellular carcinoma, lung cancer, breast cancer, head and neck cancer, prostate cancer, melanoma, multiple myeloma, glioma, glioblastoma, and colorectal cancer.
  • 7. The method of claim 1, wherein the cancer is glioma.
  • 8. The method of claim 1, wherein the cancer is glioblastoma.
PCT Information
Filing Document Filing Date Country Kind
PCT/US2017/023965 3/24/2017 WO
Publishing Document Publishing Date Country Kind
WO2017/165732 9/28/2017 WO A
US Referenced Citations (4)
Number Name Date Kind
8143255 Blanchard et al. Mar 2012 B2
9120815 Mansfield et al. Sep 2015 B2
10544162 Mansfield et al. Jan 2020 B2
20200262843 Mansfield et al. Aug 2020 A1
Foreign Referenced Citations (6)
Number Date Country
WO 2015004534 Jan 2015 WO
WO 2015026634 Feb 2015 WO
WO-2015095840 Jun 2015 WO
WO 2015153870 Oct 2015 WO
WO 2015157093 Oct 2015 WO
WO 2016040892 Mar 2016 WO
Non-Patent Literature Citations (27)
Entry
The abstract of Burrows et al (Clinical Lymphoma, Myeloma &Leukemia, 2014, Sep. 2014, p. S152, abstract No. 607) (Year: 2014).
Abstract of Hofmeister et al (Blood, Dec. 3, 2015, vol. 126, No. 23) (Year: 2015).
“Prescribing Information” for Kyprolis (revised Jul. 2012) (Year: 2012).
The abstract of Barnhardt et al (Blood, Nov. 15, 2013, vol. 122, No. 21, p. 3171) (Year: 2013).
NCT01204164 (Apr. 8, 2015 version). (Year: 2015).
Williams et al (Journal of Medicinal Chemistry, 2012, vol. 55, pp. 169-196) (Year: 2012).
Abstract of Park et al (Blood, Dec. 2015, vol. 126, No. 23) (Year: 2015).
Sanchez-Martinez, Bioorganic and Medicinal Chemistry Letters, 2015, vol. 25, pp. 3420-3435 (Year: 2015).
Flakus (IAEA Bulletin, 1982, vol. 23, pp. 31-36 (Year: 1982).
Qaim (Radiochimica Acta, 2001, vol. 89, pp. 297-302) (Year: 2001).
The abstract of Stepanek et al (Acta Oncologica, 1996, vol. 35, pp. 863-868) (Year: 1996).
Hdeib and Slaon (Expert Opinion on Biological Therapy, 2011, vol. 11, pp. 799-806). (Year: 2011).
Álvarez-Fernández, S., et al., “Potent antimyeloma activity of a novel ERK5/CDK inhibitor,” Clinical Cancer Research 19(10): 2677-2687, American Association for Cancer Research, United States (2013).
Burrows, F., et al., “TG02: A novel, multi-kinase inhibitor with potent activity against solid tumors,” Journal of Clinical Oncology 28(15_suppl):e13549, American Society of Clinical Oncology, United States (2010).
Goh, K. C., et al., “TG02, a novel oral multi-kinase inhibitor of CDKs, JAK2 and FLT3 with potent anti-leukemic properties,” Leukemia 26(2): 236, Nature Publishing Group, United Kingdom (2012).
Matsumoto, F., et al., “Abstract 2487: A multi-kinase inhibitor, XL228, enhanced human cancer cell radiosensitivity and suppressed cell invasion and migration,” Cancer Research 71(8_suppl):2487, American Association for Cancer Research, United States (2011).
Munshi, A., et al., “Radiosensitization of human melanoma cells by sorafenib (BAY 43- 9006), a multi-kinase inhibitor,” Cancer Research 66(8_Suppl):4671, American Association for Cancer Research, United States (2006).
Pallis, M., et al., “The multi-kinase inhibitor TG 02 overcomes signalling activation by survival factors to deplete MCL 1 and XIAP and induce cell death in primary acute myeloid leukaemia cells,” British Journal of Haematology 159(2): 191-203, Wiley-Blackwell Publishing Ltd., United Kingdom (2012).
Pelton, K., et al., “EXTH-63. Preclinical Efficacy of a CDK Inhibitor (TG02) in Glioblastoma.” Neuro-Oncology18(suppl_6):vi73, Oxford University Press, United Kingdom (2016).
PR Web, “Tragara and Lee's Pharma Enter into Exclusive Licensing Agreement to Develop and Commercialize TG02 in Greater China Market and South East Asia,” press release dated Nov. 16, 2015, 3 pages.
Pasha. M.K et al., “Preclinical Metabolism And Pharmacokinetics of SB1317 (TG02), a Potent CDK/JAK2/FLT3 Inhibitor,” Drug Metabolism Letters 6(1):33-42, Bentham Science Publishers (2012).
Parrott, T. et al. “P08.32 TG02, an oral CDK inhibitor, demonstrates activity in glioma models: EORTC Brain Tumor Group Conducts Phase 1b study (STEAM / EORTC 1608)” Neuro-Oncology 18(Suppl 4): iv48, Oxford University Press (2016).
Wadhwa, E. et al. “PDTM-41. TG02, a Novel Multikinase Inhibitor, is Effective in Pediatric Brain Tumors, With Selective Potency in Those With MYC Expression” Neuro-Oncology 19(Suppl 6): vi198-vi199, Oxford University Press (2017).
Sonawane, Y. A. et al. “Cyclin Dependent Kinase 9 Inhibitors for Cancer Therapy,” J. Med. Chem. 59:8667-8684, American Chemical Society (2016).
Inuzuka, H. et al., “SCFFbw7 Regulates Cellular Apoptosis by Targeting Mcl-1 for Ubiquitination and Destruction,” Nature 471:104-9, Springer Nature Publishing AG (2011).
Sehgal, A., et al. “PD-1 Checkpoint Blockade in Acute Myeloid Leukemia,” Expert Opin Biol Ther. 15(8):1191-1203 (2015).
Brahmer, J.R. et al., “Nivolumab: targeting PD-1 to bolster antitumor immunity,” Future Oncology 11(9):1307-1326, Future Science Group, London, UK (2015).
Related Publications (1)
Number Date Country
20200323862 A1 Oct 2020 US
Provisional Applications (2)
Number Date Country
62423468 Nov 2016 US
62312712 Mar 2016 US