AU2014200040A1 - Method of radio-sensitizing tumors using a radio-sensitizing agent - Google Patents

Method of radio-sensitizing tumors using a radio-sensitizing agent Download PDF

Info

Publication number
AU2014200040A1
AU2014200040A1 AU2014200040A AU2014200040A AU2014200040A1 AU 2014200040 A1 AU2014200040 A1 AU 2014200040A1 AU 2014200040 A AU2014200040 A AU 2014200040A AU 2014200040 A AU2014200040 A AU 2014200040A AU 2014200040 A1 AU2014200040 A1 AU 2014200040A1
Authority
AU
Australia
Prior art keywords
carcinoma
alkyl
cancer
radiation
leukemia
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2014200040A
Inventor
James L. Diebold
Robert L. Hudkins
Sheila J. Miknyoczki
Bruce Ruggeri
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cephalon LLC
Original Assignee
Cephalon LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2007321987A external-priority patent/AU2007321987B2/en
Application filed by Cephalon LLC filed Critical Cephalon LLC
Priority to AU2014200040A priority Critical patent/AU2014200040A1/en
Publication of AU2014200040A1 publication Critical patent/AU2014200040A1/en
Abandoned legal-status Critical Current

Links

Abstract

The present invention relates to a method of treating cancer using PARP inhibitors as radio-sensitization agents of tumors. Specifically the present 5 invention relates to a method of radio-sensitization of tumors using a compound of Formula (1) or a pharmaceutically acceptable salt form thereof. The present invention also relates to pharmaceutical compositions of PARP inhibitors for radiosensitizing tumors.

Description

Method of Radio-Sensitizing Tomiors Using a Radio-Sensitizing Agent Field of the Invention The present invention relates to a method of treating cancer using PARP inhibitors 5 as radio-sensitization agents of tumors. Specifically the present invention relates to a method of radiosensitization of tumors using a compound of Formula (1) H-C, 0 / .20 N H (I) 10 or a pharmaceutically acceptable salt form thereof. The present invention also relates to a pharmaceutical compositions of PARP inhibitors for radiosensitizing tumors. Background of the Invention Radiation is a cytotOxic treatment modality that induces cellular damage by 15 creating DNA strand breaks. Poly (ADP-ribose) polymerase 1 (PARP-1) a nuclear zinc finger DNA binding protein which is activated by awd iuplicated in DNA radiation induced-damage and repair, PARP binds to DNA strand breaks which may serve to protect them from nuclease attack or recombination, Since PARP acts to aid in DNA repair, inhibitors have the potential to enhance the chemno- and radio-sensitization of 20 cytotoxic agents (Curtin, 2005), The most significant cause for treatment failure and cancer mortality is radiolchemo-resistance, Agents to overcome cancer cell resistance to cytotoxic agents may be a key factor in successful cancer therapy. The potential application of PARP inhibitors therapeutically as chemo- and radio-sensitizers has, until relatively recently, 25 been limited by the potency, selectivity, and pharmaceutic properties of these agents (Griffin et aL, 1998; Bowman, et al, 1998; Bowman et at, 2001, Chen & Pan, 1998; Delany et at., 2000; Griffin et aL, 1995; Lui, et aL, 1999). Recently, more potent and selective PARP inhibitors (benzimidazole-4-carboxamides and quinazoin-4-[3H-ones) have been developed that have demonstrated the ability to potentiate the effects of 30 radiation and of chemotherapeutic agents such as camptothecin (CPT), topotecan, irinotecan, cisplatin, eroposide, bleomycin, BCNU, and temozolomide (TMZ) in vitro and in vvo using both human and murine tumor models of leukemia, lymophma metastases to the central nervous system, colon, lung and breast carcinomas agents (Griffin et aL, 1998; Bowman, et at, 1998; Bowman et at, 2001, Chen & Pan, 1998; Delany et aL, 2000; 5 Griffin et at, 195; Lui, et at, 1999, Tentori, et at, 2002). A PARP inhibitor that is able to sensitize tumor cells to the actions of different classes of chemotherapeutic agents and/or radiation could increase the success rate of established cancer therapies. PARP-1 is a 11 6kD nuclear zinc finger DNA binding protein that uses NAD+ as a substrate to transfer ADP -riose onto acceptor proteins such as histones polyrierases, 10 ligases, and PARP itself (automodification) (Griffin et al, 1998; Tentori, et al 2002; Blaldwin et aL, 2002), PARP belongs to a family of proteins that currently includes 18 members, of these PAR?- I and PARP-2 are the only enzymes activated by DNA damage (Curtin, 2005; Tentori, et al, 2002), Activation of PARP-2 may also induce pro inflammatory activity (Jagtap and Szabo, 2005), indicating that inhibition of PARP-2 in 15 tumor cells may be of additional therapeutic benefit. Although the pathophysiological and physiological process modulated by the various PARP isofonms are the subject of extensive study (Ame et at, 2004), the best characterized member of this family, and the major focus of targeted drug discovery efforts therapeutically in oncology, is PARP-1. PARP is active in the regulation of many different biological processes, including 20 protein expression at the transcriptional level, replication and differentiation, telornerase activity, and cytoskeletal organization. However, it is the role PARP plays in DNA repair and maintenance of genomic integrity that is of interest for the use of PAR? inhibitors as chemo/radio-sensitizing agents (Smith, 2001). This role is illustrated via the use of PARP I deficient cells which demonstrate delayed base excision repair and a high frequency of 25 sister chromatid exchange upon exposure to ionizing radiation or treatment with alkylating agents. In addition, high levels of ionizing radiation and alkylating agents elicit higher lethality in PARP-1 deficient mice as compared to wild type mice (Smith, 2001; Virag & Szabo, 2002), Among the members of the PARP family, PARP- I (and PARP-2) is specifically 30 activated by, and implicated in, the repair of DNA strand breaks caused directly by ionizing radiation, or indirectly following enzymatic repair of DNA lesions due to nethylating agents, topoisomerase I inhibitors, and other chemotherpeutic agents such as cisplatin and bleoinycin (Griffin et at, 1998; Delany at al., 2000; Tentori et at, 2002; de Murcia et at, 1997). There is a substantial body of biochemical and genetic evidence demonstrating that PARP-I plays a role in cell survival and repair following sub-lethal massive DNA damage. Funhermore, as exemplified by PARP-1 knockout mice, PARP- I function in the absence of DNA damage is not critical for cell survival has made inhibition of PARP-1 a potentially viable therapeutic strategy for use with chemo- and/or radio 5 therpy (Delany et aL, 2000; Birkle et aL, 1993) Early generations of PARP-1 inhibitors such as 3-aminobenzarnide, nicotinamide and related derivatives, potentiated both the in vitro and in vivo cytotoxic activities of radiation, bleomycin, CT, cisplatin and TMZ in human and muine tumor models in vitro and in vivo. The inherent limitations in the potency, selectivity, and deliverability of these 10 compounds precluded assigning unequivocally the potentiation of anti-tumor efficacy observed in vitro and in vivo to the inhibition of PARP- specifically versus non-specific activities of these molecules (Griffin et al, 1998; Grif'n et at. 1995; Masuntani et a,, 2000; Kato et at, 1988), These issues were influential in the development of more potent and selective structural classes of PRAP-1 inhibitors including various benzimidazole-4 15 carboxamides and quinazolin4[3H-one derivatives. In vitro and In vivo analyses revealed that these compounds were able to potentiate the efficacy of chemotherapeutic agents using both human and marine tumor models (Griffin et at., 1998; Bowman, et at, 1998; Bowman et at, 2001; Chen & Pan, 1998; Delany et at, 2000; Griffin et at, 1995; Liu, et at, 1999), 20 PCT publication W02001085686, published Nov, 15, 2001, discloses carbazole compounds with PARP inhibitory activity. There is a need to discover and develop PARP inhibitors as radio-sensitization agents for the treatment of cancer which have high selectivity for PARP, high potency, improved deliverability, and improved tolerability profiles. 25 Summary of the Invention The present invention provides a method of using a 4-methoxy-carbazole to cause radio-sensitization in tumors by the in vivo inhibition of PARP-l The method comprises a 4-methoxy-carbazole of Formula 1a): 300N (1a) and prodrugs thereof, preferably a Mannich base prodrug thereof, to provide solubility and stability, and to aid in the in vivo delivery of the active drug, 7-methoxy-1,2,3,11 tetrahydro-S, 11 -diaza-benzo[altrindene-4,6-dione. 5 The present invention further provides for a method of treating cancer by administering a radiosensitizing agent of Formula (I), X HC,. 0 / 0 ~WN N H \/ (I) 10 or a pharmaceutically acceptable salt form thereof, wherein, X is H or a prodrug moiety, as defined herein; to a mammal suffering from cancer and applying ionizing radiation to said mammal tissue, Another object of the present invention is to provide pharnaceutical compositions comprising the compounds of the present invention wherein the compositions comprise 15 one or more pharmaceutially acceptable recipients and a therapeutically effective amount of at least one of the compounds of the present invention, or a pharmaceutically acceptable salt or ester form thereof Another object of the present invention is to provide a compound of Formula (1): 0.-N / O0 20 or a pharmaceutically acceptable salt fomni thereof. In another embodiment, the present invention provides use of a compound of Formula (1) for the manufacture ofa a medicament for the tre~atment of cancer. -4 4- These and other objects, features and advantages of the invention will be disclosed in the -following detailed description of the patent disclosure, 5 Brief Description of the Diagrams Fig.1: Shows the effect of the Mannich base prodrug in combination with Radiation using radio-resistant U87MG glioblastoma xenografis on the growth delay of the tumors. Fig,2: Magnitude of effect with combination therapy stronger than that achieved with a comparable regimen of radio-therapy or the prodrug only 10 Figa3: Radio-sensitizing Effect of Example 7 in U87MG Human Glioblastoma Xenografts in Nude Mice (Non-optimized Schedule). Fig. 4 shows a synthetic schematic including a compound within the scope of the present invention and precursors thereto. Fig,5: Radio-sensitizing Effect of Example 7 administered orally in U87MG Human 15 Glioblastoma Xenografts in Nude Mice, Detailed Description of the Invention in a first embodiment, the present invention provides a method of treating cancer 20 by administering a radiosensitizing agent of Formnla (1): H. C 0 H (I) or a pharmaceutically acceptable salt fonn thereof, wherein, X is H or a prodrug moiety; 25 to a manual suffering from cancer and applying ionizing radiation to said mammal tissue. In a preferred embodinent the radiosensitizing agent is present within or proximate to said tissue increases the efficiency of conversion of said applied ionizing radiation into localized therapeutic effects. ~-5.- In a preferred embodiment the radiosensitizing hgent is present in an amount effective to radiosensitize cancer cells, in a preferred embodiment the ionizing radiation of said tissue is performed with a dose of radiation effective to destroy said cells. 5 In a preferred embodiment the ionizing radiation is of clinically acceptable or recommended radiotheraputic protocols for a given cancer type. In a preferred embodiment the cancer is malignant. In a preferred embodiment the cancer is benign. In a preferred embodiment the the prodrug moiety is selected from the group 10 consisting of-CH rNRR2, -CH2OC(=0)R CH 2 OP(=0)(OR) 2 , and C(=0)R 4 ; wherein; R1 is H or C1 alkyl;
R
2 is H or Cu alkyl; alternatively, R' and R1, together with the nitrogen atom to which they are attached, form 1$ a heterocyclyl group selected from pyrrolyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl, wherein said heterocyclyl group is optionally substituted with C! alkyl;
R
5 is selected from the group consisting of -CIA alky[NRRK -C14 alkyl-OR, pyridinyl, phenyl(CH2NR 1
R
2 ) and -CH(Rt)NH 2 ; 20 R 4 is selected from the group consisting of -O(C 1 4 aikyl)NR 1
R
2 -- (C 4 aikyl)-ORt and -CH(R 6 )Nlt; R is H or C1A alkyl; and R" is the side chain of a naturally occurring amino acid, In a preferred embodiment the prodrug moiety is -CH 2
NRR
2 , R' is H or Cu 25 alkyl; R2 is H or Cu alkyl; and alternatively, R 1 and R2, together with the nitrogen atom to which they are attached, form a heterocyclyl group selected from pyrrolyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl, wherein said heterocyclyl group is optionally substituted with C..
1 alkyL In a preferred embodiment the prodrug moiety is a Mannich base, 30 In a preferred embodiment the Mannich base is selected form 4-methyl-piperazln 1i-ylmethyl-, morpholin-4-ylmethyl-, and 5-diethylaminomethy In a preferred embodiment the Mannich base is 4-methyl-piperazin-1 -ylmethyl. In a preferred embodiment the route of administration is intravenous, subcutaneous. oral or intraperitoneally. -6- In a preferred embodiment the route of administration is intravenous, In a preferred embodiment the cancer is selected from head and neck squamous cell carcinoma (eye, lip, oral , pharynx, Larynx, nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (epidermis), giablastoma, 5 astrocytoma, oligodendroglioma, oligoastrocytoma, meinngioma, neuroblastoma, rhabdomyosarcoma, soft-tissue sarcomas, osteosarcoma, hematolog icmalignancy at the ens site, breast carcinoma (ductal and carcinoma in situ), thyroid carcinoma (papillary and follicular), lung carcinoma (bronchioloalweolar carcinoma, small cell lung carcinoma mixed small cell/large cell carcinoma, combined small cell carcinoma, non-small cell lung 10 carcinoma, squamous cell carcinoma, large cell carcinoma, and adenocarcinoma of the lung), hepatocellular carcinoma, colo-recial carcinoma, cervical carcinoma, ovarian carcinoma, prostatic carcinoma, testicular carcinoma, gastric carcinoma, pancreatic carcinoma, cholangiosarcoma, lymphoma (Hodgkins and non-Hodgkins types of T-and B cell origin), leukemia (acute and chronic leukemia of myeloid and lymphoid origins), and 15 bladder carcinoma. In a preferred embodiment the cancer is selected froin head and neck squamous cell carcinoma (eye, lip, oral, pharynx, larynx, nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (epidermis), glioblastoma, neuroblastoma, rhabdomyosarcoma, lung carcinoma, (bronchioloalveolar carcinoma, 20 small cell lung carcinoma, mixed small cell/large cell carcinoma, combined small cell carcinoma, non-small cell lung carcinoma, squamous cell carcinoma large cell carcinoma-, and adenocarcinoma of the lung), lymphoma (Hodgkins and non-Hodgkins types of T-and fl-cell origin), and leukemia (acute and chronic leukemia of myeloid and lymphoid origins). 25 In a preferred embodiment, the present invention provides a method of treating cancer by administering a radiosensitizing agent of formula 7-mrethoxy- 1. ,2.3,11 tetrahydro-5)1iI -diaza-benzofaltrindene-4,6-dione. In a preferred embodiment, the present invention provides a method of treating cancer by administering a radiosensitizing agent of formula 7-methoxy-1,2,3,11 30 tetrahydro-5,1 -diaza-benzo altrindene-4,6-dione. In a second embodiment, the present invention provides a pharmaceutical composition for radiosensitizing cancer cells comprising a radiosensitizing amount of a compound of Formula (1): -7-- H C 0) or a pharmaceutically acceptable salt form thereof, wherein X is H or a prodrug moiety; and a pharmaceutically acceptable carrier. 5 In a preferred embodiment, the prodrug moiety is selected from the group consisting of-CH 2
NR
2 R -CH 2 OC(=O)R -CHOP(=O)(OH)2, and -C(=O)R 4 ; Ri is H or C>4 alkyl;
R
2 is H or C4 alkyl; alternatively R 1 and R2, together with the nitrogen atom to which they are attached, form 10 a heterocyclyl group selected front pyrrolyt, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl, wherein said heterocyclyl group is optionally substituted with Cj alkyl; Re is selected from the group consisting of M- alkyl-NRW', ~C alkyl-OR, pyridinyi, ~ phenyi(CH2NR R2), and -CH(R%)NH 2 ; 15 R 4 is selected from the group consisting of -0-(C alkyl)-NR R O 2 , -0(C alkyl)-ORt and -CH(R)Nht; R is H or C4 alkyl; and R is the side chain of a naturally occurring amino acid, In a preterred embodiment, the prodnig moiety is -CH7NR'R 2 . 20 R1 is H or C3 alkyl; R2 is H or C4 alkyl; and alternatively, R and n together with the nitrogen atom to which they are attached, form a heterocyclyl group selected from pyrrolyl, pyrrolidinyl, piperidinyl, morpholiny, thiomorpholinyl, and piperazinyl, wherein said heterocyclyl group is optionally substituted 25 with C4 alkyL In a preferred embodiment, the compound is -8- HC N A or a pharIaceutically acceptable salt form thereof In a preferred embodiment, the compound is "'00 0 AN /,-N 'NA N A" N O N H \ 50 or a phannaceutically acceptable salt form thereof In a third embodiment, the present invention provides .fcr a compound of Formula (II): -N -N / F0orma ha reutianfactabre sfat fon n t thereof, rateto wxw ln -a preferred embodiment, the pressent invention provides uise of a comnpound of Formula (11) for the mamufacture of a medicament for the treatment of cancer. 15 It is appreciated that certain featureosof the invenion, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment, Conversely, various features of the invention which are, for brevity , - 9 described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. The following tenns and expressions contained herein are defined as follows: As used herein, the term "about" refers to a range of values from i 10% of a 5 specified value, For example, the phrase "about 50 mag" includes ± 10% of 50, or from 45 to 55 mg. As used herein, the term "alkyl" refers to a. straight-chain, or branched, alkyl group having I to 4 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec butyl, and tert-butyL A designation such as "CrCK 4 alkyl" refers to an alkyl radical 10 containing from I to 4 carbon atoms. As used herein, the term "amino acid" means a molecule containing both an amino group and a carboxyl group. It includes an "c-amino acid" which is well known to one skilled in the art as a carboxylic acid that bears an amino fnctionality on the carbon adjacent to the carboxyl group, Amino acids can be naturally occurring or non-naturally 15 occurring, "Naturally occurring amino acids" include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isolucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine. As used herein, the tern "heterocyclyl" refers to a 5 or 6 membered cyclic group 20 containing carbon atoms and at least heteroatom selected forn 0, N, or S, wherein said heterocyclyl group may be saturated or unsaturated and wherein said heterocyclyl group may be substituted or imsubstituted. The nitrogen and sulfur heteroatoms may be optionally oxidized. Examples of heterocyclyl groups include pyrrolyl, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and methylpiperazinyl, 2$ As used herein, the term mammalt" refers to a wann blooded animal such as a mouse, rat, cat, dog, monkey or human, preferably a human, or a human child, which is afflicted with, or has the potential to be afflicted with, one or more diseases and conditions described herein. As used herein, a "pharmaceutically acceptable" component is one that is suitable 30 for use with humans and/or animals without undue adverse side effects (such as toxicity, irritation and allergic response) commensurate with a reasonable benefitlrisk ratio, As used herein, the term "safe and effective amount" refers to the quantity of a component which is sufficient to yield a desired therapeutic response without undue -10 adverse side effects (such as toxicity, irritation, or allergic response) commensurate with a reasonable benefit/risk. ratio when used in the manner of this invention. By "therapeutically effective amount" is meant an amount of a compound of the present invention effective to yield the desired therapeutic response, For example, an amount 5 effective to delay the growth of or to cause a cancer, either a sarcoma or lymphoma, or to shrink the cancer or prevent metastasis. The specific safe and effective amount or therapeutically effective amount will vary with sueh factors as the particular condition being treated, the physical condition of the patient, the type of mammal or animal being treated, the duration of the treatment, the nature of concurrent therapy (if any), and the 10 specific formulations employed and the structure of the compounds or its derivatives. In the present invention, the term "ionizing radiation" means radiation comprising particles or photons that have sufficient energy or can produce sufficient energy via nuclear interactions to produce ionization (gain or loss of electrons), An exemplary and preferred ionizing radiation is an x-radiation, Means for delivering x-radiation to a target 15 tissue or cell are well known in the art. The amount of ionizing radiation needed in a given cell generally depends on the nature of that cell. Means for determining an effective amount of radiation are well known in the art, Used herein, the term "an effective dose" of ionizing radiation means a dose of ionizing radiation that produces an increase in cell damage or death when given in conjunction with the compounds of the invention. 20 Dosage ranges for x-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 weeks), to single doses of 2000 to 6000 roentgens, Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells. Any suitable means for delivering radiation to a tissue may be employed in the 25 present invention. Common means of delivering radiation to a tissue is by an ionizing radiation source external to the body being treated. Alternative methods for delivering radiation to a tissue include, for example, first delivering in vivo a radiolabeled antibody that immnunoreacts with an antigen of the tumor, followed by delivering in vivo an effective amount of the radiolabeed antibody to the tumor. In addition, radioisotopes may 30 be used to deliver ionizing radiation to a tissue or cell. Additionally, the radiation may be delivered by means of a radiomnimetic agent. A.s used herein a "radiominetic agent" is a chemnotherapeutic agent, for example melphalan, that causes the same type of cellular damage as radiation therapy, but without the application of radiation.
As used herein the term. "prodrug moiety' means; the prodrug can be converted under physiological conditions to the biologically active drug by a number of chemical and biological mechanisms. In one embodiment, conversion of the prodrug to the biologically active drug can be accomplished by hydrolysis of the prodrug moiety 5 provided the prodrug moiety is chemically or enzymatically hydrolyzable with water. The reaction with water typically results in removal of the prodrug moiety and liberation of the biologically active drug. Yet another aspect of the invention provides conversion of the prodrug to the biologically active drug by reduction of the prodrug moiety, Typically in this embodiment, the prodrug moiety is reducible under physiological conditions in the 10 presence of a reducing enzymatic process, The reduction preferably results in removal of the prodrug moiety and liberation of the biologically active drug, In another embodiment., conversion of the prodrug to the biologically active drug can also be accomplished by oxidation of the prodrug moiety, Typically in this embodiment, the prodrug moiety is oxidizable under physiological conditions in the presence of an oxidative enzymatic 15 process. The oxidation preferably results in removal of the prodrug moiety and liberation of the biologically active drug. A further aspect of the invention encompasses conversion of the prodrug to the biologically active drug by elimination of the prodnig moiety. Generally speaking, in this embodiment the prodrug moiety is removed under physiological conditions with a chemical or biological reaction, The elimination results in 20 removal of the prodrug moiety and liberation of the biologically active drug. Of course, any prodrug compound of the present invention may undergo any combination of the above detailed mechanisms to convert the prodrug to the biologically active compound. For exnple, a particular compound may undergo hydrolysis, oxidation, elimination, and reduction to convert the prodrug to the biologically active compound. Equally, a particular 25 compound may undergo only one of these mechanisms to convert the prodrug to the biologically active compound. As used herein, "cancer" refers to all types of cancer or neoplasm or malignant or benign tumors found in mammals, including carcinomas and sarcomnas. Examples of cancers are cancer of the brain, breast, pancreas, cervix, colon, head & neck, kidney, lung 30 non-small cell lung, melanoma, mesothelioma, ovary, sarcoma, stomach, uterus and Medulioblastoma. The term "leukemia' refers broadly to progressive, malignant diseases of the blood-forming organs and is generally characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow, Leukemia is generally clinically classified on the basis of (1) the duration and character of the disease-acute or chronic; (2) the type of cell involved; myeloid (myelogenous), lymphoid (lymphogenous), or monocytic; and (3) the increase or non-increase in the number abnormal cells in the blood-leukemic or aleukemic (subleukemic). The P388 leukemia 5 model is widely accepted as being predictive of in vivo anti-leukemic activity, It is believed that compounds that tests positive in the P388 assay will generally exhibit some level of antideukemic activity in vivo regardless of the type of leukemia being treated, Accordingly, the present invention includes a method of treating leukemia, and, preferably, a method of treating acute nonlymphocytic leukemia chronic lymphocytic 10 leukemia acute granulocytic leukemia, chronic granulocytic leukemia, acute promyelocyic leukemia, adult T-cell leukemia., aleukemic leukemia, a leukocythemic leukemia, basophylic leukemia, blast cell leukemia, bovine leukemia, chronic myelocytic leukemia, leukemia cutis, embryonal leukemia, eosinophilic leukemia, Gross' leukemia, hairy-cell leukemia, hemoblastic leukemia, hemocytoblastic leukemia, histiocytic 15 leukemia, stem cell leukemia, acute monocytic leukemia, leukopenic leukemia, lymphatic leukemia, lymphoblastic leukemia, lymphocytic leukemia, lymphogenous leukemia, lymphoid leukemia, lymphosarcoma cell leukemia, mast cell leukemia, megakaryocytic leukemia, micromyeloblastic leukemia, monocytic leukemia, myeloblastic leukemia, myelocytic leukemia, myeloid granulocytic leukemia, myelomonocytic leukemia, Naegeli 20 leukemia, plasma cell leukemia, plasmacytic leukemia, promyelocytic leukemia, Rieder cell leukemia, Schilling's leukemia, stem cell leukemia, subleukemic leukemia, and undifferentiated cell leukemia. The term "sarcoma" generally refers to a tumor which is made up of a substance like the embryonic connective tissue and is generally composed of closely packed cells 25 embedded in a fibrillar or homogeneous substance. Sarcomas which can be treated with 4 methoxy-carbazole and radiotherapy include a chondrosarcoma, eholangiosarcoma, frosarcoma, lymnphosarcoma. melanosarcoma, myxosarcoma, osteosarcoma, Abemethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' 30 tumor sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, granulocyticsarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented hemorrhagic sarcoma, immunoblastic sarcoma of B cells, lymphoma, immunoblastic sarcoma of T-cells, Jensen's sarcoma, Kaposi's sarcoma, Kupffer cell sarcoma, angiosarcoma leukosarcorna, malignant mesenchymoma sarcoma - 13 ~ parosteal sarcoma, reticulocytic sarcoma, Ros sarcoma serocystic sarcoma, softtissue sarcoma, synovial sarcoma, and telangiecialtic sarcoma. The termnmelanoma" is taken to mean a tumor arising from the mclanoctric system of the skin and other organs. Melanomas which can be treated with 4-methoxy 5 carbazole and radiotherapy include, for example, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma, Harding Passey melanoma, juvenie melanoma, lentigo maligna melanoma, malignant melanoma, nodular melanoma, subungal melanoma, and superficial spreading melanoma. The term "carcinoma" refers to a malignant new growth made up of epithelial cells 10 tending to infiltrate the surounding tissues and give rise to metastases. Exemplary carcinomas which can be treated with 4-methoxy-carbazole and radiotherapy include, for example, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, breast Carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma 15 basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bladder carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriforn carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colloid carcinoma, colo rectual carcinoma, cervical carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical 20 cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiennoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex ulcere, carcinoma fibrosum, gastric carcinoma, gelatinifonn carcinoma, gelatinous carcinoma, giant cell carcinoma, carcinoma gigantocellulare, glandular carcinoma, granulosa cell carcinoma, hair-matrix carcinoma, hematoid carcinoma, 25 hepatocellular carcinoma, Hurthie cell carcinoma, hyaline carcinoma, hypemephroid carcinoma, infantile embryonal carcinoma, carcinoma in situ, intruepidermal carcinoma, intraepithelial carcinoma, Krompecher's carcinoma, Kulchitzky-cell carcinoma, large-cell carcinoma, lenticular carcinoma, carcinoma lenticulare, lipomatous carcinoma, lung carcinoma, lymphoepithelial carcinoma, carcinoma medullare, medullary carcinoma, 30 melanotic carcinoma, carcinoma molle, mucirmus carcinoma, carcinoma muciparum, carcinoma mucocellulare, mucoepidermoid carcinoma, carcinoma mucosum, mucous carcinoma, carcinoma myxomatodes, nasopharyngeal carcinoma, oat cell carcinoma, carcinoma ossificans, ostecid carcinoma, ovarian carcinoma, pancreatic carcinoma, prostatic carcinoma, papillary carcinoma, periportal carcinoma, preinvasive carcinoma, ~ 14prickle cell carcinoma, pultaceous carcinoma, renal cell carcinoma of kidney, reserve cell carcinoma, carcinoma sarcomatodes, schneiderian carcinoma, scirrhous carcinoma, carcinoma scrod signet-ring cell carcinoma, carcinoma simplex. small-cell carcinoma, solanoid carcinoma, spheroidal cell carcinoma, spindle cell carcinoma, carcinoma 5 spongiosum, squamous carcinoma, squamous cell carcinoma, string carcinoma, carcinoma telangiectaticum, carcinoma telangiectodes, testicular carcincoma, transitional cell carcinoma, thyroid carcinoma, carcinoma tuberosun, tuberous carcinoma, verrucous carcinoma, and carcinoma villosum. Preferred cancers which can be treated with compounds according to the invention 10 include, head and neck squamous cell carcinoma (eye, lip, oral, pharynx, larynx, nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (epidermiis), glioblastoma, astrocytoma, oligodendrogiioma, oligoasurocytoma, meningioma, neuroblastona, thabdomyosarcoma, soft-tissue sarcomas, osteosarcoma, hematologic malignancy at the ens site, breast carcinoma (ductal and carcinoma in situ), 15 thyroid carcinoma (papillary and follicular), lung carcinoma (bronchioloalveolar carcinoma, small cell lung carcinoma, mixed small cell/iarge cell carcinoma, combined small cell carcinoma, non-small cell lung carcinoma, squamous cell carcinoma, large cell carcinoma, and adenocarcinoma of the lung), hepatocellular carcinoma, colo-rectal carcinoma, cervical carcinoma, ovarian carcinoma, prostatic carcinoma, testicular 20 carcinoma, gastric carcinoma, pancreatic carcinoma, cholangiosarcoma, lymphoma (Hodgkins and non-Hodgkins types of T-and B-cell origin), leukemia (acute and chronic leukemias of myeloid and lymphoid origins), and bladder carcinoma. More preferred cancers which can be treated with compounds according to the invention include, head and neck squamous cell carcinoma (eye, lip, oral, pharynx, larynx, 25 nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (Cpidermis) glioblastoma, neuroblastoma, rhabdomyosarcoma, lung carcinoma, (bronchioloalveolar carcinoma, small cell lung carcinoma, mixed small cell/large cell carcinoma, combined small cell carcinoma, non-small cell lung carcinoma, squamous cell carcinoma, large cell carcinoma, and adenocarcinoma of the lung), lymphoma (Hodgkins 30 and non-Hodgkins types of T-and B-cell origin), and leukemia (acute and chronic leukeniias of myeloid and lymphoid origins). As used herein, the term "4~methoxy-carbazole" is used to mean those chemicals having the formula: ~-15 ~ 0. / "0 N H / or a pharmaceutically acceptable salt form thereof, wherein, X is El or a prodrug moiety. The compound of the present invention may contain a prodrug moiety. Examples of a prodrug moiety contemplated by the invention can be selected from phosphate esters, 5 amino acid esters, amino acid amides, aminoalkyl carbamates, alkoxyakyl carbamates, hydroxyalkyl carbamates, alkoxyaikyl esters, hydroxyalkyl esters, benzoic acid esters, niotinic esters. piperazine acetates, morpholine acetates, and Mannich bases, Examples of a prodrug moiety contemplated by the invention can be selected from: 0 0 ....- 0 0H 0N~"N H s/y0 -O -O)(OH) O NH rNH2 10R6 R6 0) 0 0 O N(R) 2 OR >H 0 {0R NtON JOH 15 O 0 O N(R) N(R) -l6- N0 N 0 f O NN >N NN--' 0oN -N / N I N N; ;and A preferred prodnig moiety is a Mannich base, Prefrred Mannich bases include, but are not limited to, 4-methylpiperazin ylmethyl~, morpholin~4-ylmethyl-, and diethylaminonethyl-. Compounds of the present invention also may take the form of a 10 pharmacologically acceptable salt, hydrate, solvate, or rmetabolite, Pharmacologically acceptable salts include basic salts of inorganic and organic acids, including but not limited to hydrochloric acid, hydrobronmic acid, sulphuric acid, phosphoric acid nethanesulphonic acid, ethanesulfonic acid, malic acid, acetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, futmaric acid, succinic acid, maleic acid, salicylic acid, benzoic 15 acid, phenylacetic acid, mandelic acid, ascorbic acid, gluconic acid and the like. When compounds of the invention include an acidic function, such as a carboxy group, then suitable pharmaceutically acceptable cation pairs for the carboxy group are well known to those skilled in the art and include alkaline, alkaline earth, ammoniuim, quaternary arnmonium cations and -the like, It is contemplated by the invention that when compounds 20 of the present invention take the form of a pharmacologically acceptable salt, said salt form may be generated in situ or as an isolated solid. The compounds of the present invention, particularly in the form of the salts just described, can be combined with various excipient vehicles and/or adjuvants well known in this art which serve as phannaceutically acceptable carriers to permit drug 25 administration in the form of, e'g., injections, suspensions, emulsions, tablets, capsules, and ointments, These pharmaceutical compositions, containing a radiosensitizing amount of the described compounds, may be administered by any acceptable means which results in the radiosensitization of hypoxic tumor cells, For warm-blooded animals, and in -17particular, for humans undergoing radiotherapy treatment, administration can be oral, subcutaneous, intraperitoneally or intravenous, To destroy hypoxic tumor cells, the pharmaceutical composition containing the radiosensitizing agent is administered in an amount effective to radiosensitize the hypoxic tumor cells, The specific dosage $ administered will be dependent upon such factors as the general health and physical condition of the patient as well as his age and weight, the stage of the patient's disease condition, and the existence of any concurrent treatments. The method of administering an effective amount also varies depending on the disorder or disease being treated. It is believed that treatment by intravenous application of 10 the 4~methoxy-carbazole, fonnulated with an appropriate carrier, additional cancer inhibiting compound or compounds or diluent to facilitate application will be the preferred method of administering the compounds to warm blooded animals, Compounds described herein may be administered in pure form, combined with other active ingredients, or combined with pharnaceutically acceptable nontoxic 15 excipients or carriers, Oral compositions will generally include an inert dilient carrier or an edible carrier. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. Tablets, pills, capsules, troches and the like can contain any of the Iliowing ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch 20 or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil, In addition dosage unit 25 forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents. Further, a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes, colorings, and flavorings. The amount of compound administered to the patient is sufficient to radiosensitize 30 the malignant neoplasm to be treated but below that which may elicit toxic effects. This amount will depend upon the type of tumor, the species of the patient being treated, the indication dosage intended and the weight or body surface of the patient. The radiation may be administered to humans in a variety of different fractionation regimes, i.e., the total radiation dose is given in portions over a period of several days to several weeks. -18- These are most likely to vary from daily (i.e, five times per week) doses for up to six weeks, to once weekly doses for four to six weeks. The acmnt of a s ompound administered to the patient may be given prior to radiation treatment, during radiation treatmem, or after radiation treatment. 5 Hlowever, it is preferred that the compounds of the invention be administered prior to radiation treatment. After administration of the radiosensitizing composition to the hypoxic tumor cells and the passage of a time interval sufficient to enhance radiosensitization of the hypoxic tumor cells, the hypoxic tumor cells are irradiated with a dose of radiation effective to 10 destroy the hypoxic tumor cells. Generally, the patient will receive a radiation dosage of about 2 Gy per day for five days. Generally, the patient will receive a total radiation dosage of about 70 to about 80 Gy over seven to eight weeks, each individual radiation dose to be given within approximately I to 4 hrs after administration of the radiosensitizer. Such sequences of radiosensitization treatments and irradiation are repeated as needed to 15 abate and, optimally, reduce or eliminate, the spread of the malignancy. However, it is understood by one skilled in the art that daily radiation dosage and total radiation dosage will vary depending on a patient's tumor type, treatment protocol, and physical condition, For example, the daily dose of the present compounds is not specifically limited but can vary with a patient's age, cancer, body weight, and current treatment protocol and/or 20 medications. Additionally, the present compounds are useful as radiosensitizer and can be administered in one or more doses, i.e. one to several doses, prior to the exposure to radiation. initial Radio-sensitzing Studies Using UMG Radio-,reslstne Xnograis in Nude Mice 25 (Non-optimized Dosing schedule) Irradiation of cells induces check point arrest, which allows cells to repair DNA damage, with activated PARP facilitating the repair of DNA damage, Hypothetically, administration of a PARP inhibitor in combination with single dose or fractionated 30 radiation will reduce the ability of irradiated cells to repair DNA damage and increase cell kill. Therefore, a PARP inhibitor should work synergistically with fractionated radiation to increase tumor growth delay. The initial test of this hypothesis with Example 7/Example 6 was conducted in radio-resistant U87MG human glioblastoma xenografns in nude mice, As shon in Figure 3, administration of Example 7 alone, radiation alone, 3$ and Example 7 in combination with radiation (100 mg/kg dose equivalents of Example 6, - 19sc, qd two days prior to radiation and in combination with 7.5 Gy radiation for 3 days), was done in mice bearing established tumors, Example 7 administered as a single agent had no effect on tumor growth, Tumors treated with vehicle or Example 7 reached a tumor volume of 2000 mm3 in 10.0 days or 9,6 days ( p = 0,798, vs. control), respectively 5 Administration of radiation alone increased the time to reach 2000 aim3 to 16.1 days, an increase in tumor growth delay (TGD) of 6.1 days (p=-0.033, vs. control). In contrast, administration of Example 7 with radiation therapy increased the time for tumors to reach 2000 mm3 to 24.8 days, corresponding to a 14.8 day TOD, The magnitude of effect with the combination therapy was stronger than that seen by a comparable regimen of Example 10 7 only (p=0,001), or radiation only (p=0.00 6 ) indicating that Example 7 exhibits the profile of a true radio-sensitizer. Plasma levels of Example 6 at Cmax associated with efficacy (at 100 mg/kg Example 7) were 23 pM, comparable to those achieved at this dose in chemo-sensitization studies. 15 Radia-sensitzing Studies with Example 7 and a Clinically Relevant .Fractionated Radiotherapy Dosing Schedule Using US7MG Radioresistant Xenograps in Nude AMice A subsequent radio-sensitization study was evaluating Example 7 (30 and 100 mg/kg, s.c,) in combination with a clinically relevant fractionated radiotherapy schedule (2 20 Gy X dayss, Example 7 was administered 0,5 hr after radiation for 5 days, and dosing of Example 7 continued for 16 days after the radiation regimen was completed The rationale for this dosing schedule was based on the fact that DNA repair from radiation damage occurs 1012 days post-radiation, therefore, continual dosing of Example 7 and modulation of PARP activity covers cell cycle arrest and DNA repair time which should 25 act synergistically with fractionated radiation to increase radio-sensitivity and tumor growth delay. As shown in Figures 1 and 2. administration of radiation alone (2 Gy X days) resulted in a TGD of 2.5 days as compared to vehicle treated tumors. Administration of Example 7 (CEP 30; 30 mg&g s,c.) increased the TGD to 15 days, a 4 fold increase compared to radiation alone (ps0,05); and 26 days, a 6-fold increase 30 compared to Example 7 alone (ps 0.01). Plasma levels of Example 6 at Cmax associated with radio-sensitization efficacy were 5.5 pM, Administration of Example 7 (100 mg/kg, szc,) with fractionated radiotherapy resulted in significant anti-tumor efficacy, but 80% mortality by day 11, Plasma levels at Cmax at the 100 mg/k., s~c. dose were 21 pM, in ~20 agreement with exposure levels achieved at this dose in chemo-sensitization studies and the initial radio-sensitization studies described above, These data demonstrate that a greater increase in TGD was observed at a lower concentration of Example 7 (CEP 30; 30 mg/kg dose equivalents of Example 6 sc, qd X2 1 5 days ) using a clinically relevant fractionated dosing schedule, In addition, Example 7 (CEP 30; 30 mg/kg dose equivalents of Example 6 stc, qd X21 days) alone had no effect on tumor growth inhibition demonstrating that Example 7 acts as a "tne" radio-sensitizer. To evaluate therapeutic gain, Example 7(30 and 100 mg/kg dose equivalents of Example 6 so) plus 2 Gy radiation X 5 days was evaluated in bone marrow and jejunal 10 crypt assays to determine if Example 7 potentiated radiation-induced normal tissue (NT) toxicity. Evaluation of bone marrow and intestinal mucosa revealed that Example 7 (30 and 100 mg/kg dose equivalents of Example 6 se) did not potentate radiation toxicity in these tissues studies indicate that CEP-9722 exerts radio-sensitizing effects when administered orally. These combined data indicate that Example 7 acts as a 15 radiosensitizer by increasing the effectiveness of fractionated radiotherapy in a radio resistant glioma model in a greater than additive manner and does not potentiate radiation induced NT toxicity. Examples 20 The compounds of the present invention may be prepared in a number of methods well known to those skilled in the art, including, but not limited to those described below, or through modifications of these methods by applying standard techniques known to those skilled in the art of organic synthesis. All processes disclosed in association with the present invention are contemplated to be practiced on any scale, including milligram, 25 gram, multigram, kilogram, muhtikilograrm or commercial industrial scale. The present invention features methods for preparing the muhicyclic compounds described herein which are usefil as inhibitors of PARP, The method consists of a multistep synthesis starting with 4-methoxyindole. Specifically, 4-methoxyindole A, is 30 treated serially, for example, with butyllithium, carbon dioxide, t-butyllithium and a ketone B to provide a 2~substituted 4-methoxyindole tertiary alcohol C. This tertiary alcohol is eliminated, for example, under acidic conditions using hydrochloric acid or touenesulfonic acid, to afford a substituted 2~-vinylindole, D, Diels->Alder cycloaddition of D with a dienophile such as, but not limited to, maleimide (E) afTbrds the cycloaddition -21intermediate F Aromatization of the cycloaddition intermediate, for example, with oxygen in the presence of a catalyst such as palladium or platinum or with an oxidant such as DDQ or tetrachloroquinone, produces carbazoie G, Further treatment of G with an alkylating or acylating reagent gives indole-N-substituted 5 carbazole derivatives of the present invention. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Prodrugs; Sloane, K, B,, Ed,; Marcel Dekker: New York, 1992, incorporated by reference herein in its entirety, The compounds of the present invention are PARP inhibitors. The potency of the 10 inhibitor can be tested by measuring PARP activity in vitro or in vivo, A preferred assay monitors transfer of radiolabeled ADP-ribose units from [ 32 P]NADY to a protein acceptor such as histone or PARP itself Routine assays for PAR? are disclosed in Ptrnell and Whish, Biochemn J 1980, 185, 775, incorporated herein by reference. 15 Example I Cell Line U87MG human glioblastoma cells were cultured in commercially available Minimum Essential Medium (MEM) with 1.5 gIL sodium bicarbonate, 0.1nM non essential amino acids, ,OnM sodium pyruvate with 10% Fetal Bovine Serum (FBS). 20 Example 2 Tumor Cell Implantation and Growth Exponentially growing cells were harvested and injected ((2xl0t) cells/mouse) into the right flank of commercially available athymic NCR NUM nude mice. Animals 25 bearing tumors of 200-400mm 3 were randomized according to size into the appropriate treatment groups (n=10), Tumors were measured every 3-4 days using a vernier caliper, Tumor volumes were calculated using the following formula: V(nun 2 ) 0-5236 x length (nun) x width (mm) [length (mm) + width (mm)/2]. 30 Example 3 Methods: U87MG human glioblastoma cells were injected subcutaneously (s c.) into the right hind limb of athymic NCR NUM mice and allowed to grow to a mean tumor volume of 200mm 3 . Mice that received radiotherapy were anesthetized prior to irradiation with 100 mg/kg Ketamine + 1.0 mgkg xylezine or 37.5 mg/kg Ketamine + 0.2 mg/kg ~22 acepromazine, s.c. to provide 25-30 min of sedation. Anesthetized mice were positioned in malleable lead shielding which conforms to the animal's body size and shape without undue pressure. The body was shielded by lead. The tumor bearing leg or exposed tumor was irradiated with the appropriate dose, After tumors were irradiated, the mice are 5 returned to cages on heating pads until recovered from the anesthetics, Example 7 was given as soon possible (within 30 min) after radiation (RD Figure 3: Mice were randomized into the following treatment groups (n=l 0): 1) vehicle, 2) radiation only (7.5 Gy for 3 days), 3) Example 7 only (100 mg/kg dose equivalents of Example 6, s.c., QD for 5 days), and 4) Example 7 plus radiation, Either the Example 7 or vehicle was 10 administered s.c. on days 1-5 and 30 minutes after radiation on day 2, 3, and 4. Analysis of data was performed using mixed effects regression to model the base-I 0 logarithm of tumor volume as a function of time and treatment. Analyses were performed in SAS 8.3 (SAS Institute Inc,, Cary, NC). Figures 1&2: Mice were randomized into the following treatment groups and administered: 1) vehicle , 2) RT (5 X 2 Gy), 3) RT plus Example 7 15 (30 or 100 ngkg s.c. dose equivalents of Example 6, qd X2ld ) or 4) Example 7 (30 or 100 mg/kg dose equivalents of Example 6 s.c., qd,X2ld) only, Example 7 was given on days 1 -21 and RT was given on days 1-5, All of the animals were measured on the same day, Individual tumor volume measurements were modeled in a log transformed linear model and the best fit time for tumors to reach approximately 2000 mm 3 was determined. 20 One-way ANOVA and post hoc analysis was used to determine significance. A P value <0,05 was considered significant. Results: All groups started treatment with similar-sized tumors of 200mm.
3 (P=0,83 comparing groups at day 0), As shown in Figure 3, administration of Example 7 alone, radiation alone, and Example 7 in combination with radiation (100 mg/kg dose equivalents 25 of Example 6, sc, qd two days prior to radiation and in combination with 7.5 Gy radiation for 3 days), was done in mice bearing established tumors. Example 7 administered as a single agent had no effect on tumor growth. Tmnors treated with vehicle or Example 7 only reached a tumor volume of 2000 mm 3 in 10.0 days or 9,6 days ( P:= 0.798, vs, control), respectively, Administration of radiation alone increased the time to reach 2000 30 mm3 to 16.1 days, an increase in tumor growth delay (TGD) of 61 days (P=0.033, vs. control). The combination therapy of Example 7 with radiation therapy increased the time for tumors to reach 2000 mm 3 to 24,8 days, corresponding to a 14.8 day TGD. The magnitude of effect with the combination therapy was stronger than that seen by a comparable regimen of Example 7 only (P=0.001), or radiation only (P=0.006) indicating that Example 7 exhibits the profile of a true radio-sensitizer, As shown in Figures 1&2, administration of Example 7(CEP 30; 30 mg/kg dose equivalents of Example 6 s.c.) in combination with RT increased the TGD to 15 days, a 4 fold increase compared to radiation alone (PS0,05); and 26 days, a 6-fold increase compared to Example 7 alone (PS 5 0,001), Admintistration of Example 7 (100 mg/kg, sc.) with fractionated radiotherapy resulted in significant anti-tumor efficacy, but 80% mortality by day Ii These data demonstrate that a greater increase in TOD was observed at a lower concentration of Example 7 (CE P 30; 30 ng/kg) using a clinically relevant fractionated dosing schedule. In addition, administration of Example 7 alone had no effect on tumor growth inhibition 10 demonstrating that Example 7 acts as a "true" radio-sensitizer, Example 4 Evaluation of DNA Damage Antibodies: Primary antibodies can be used against phospho histone H2AX (Cell 15 Signaling, #2577, 1:1000) and CAPDH (Abcam, #9484, 1:5000). Secondarv antibodies can be Goat anti-mouse IRDye800 (Rockland, #610-132- 121) and Goat anti-rabbit Alexa fluor 700 (Molecular Probes, #A21038), U87MG cells can be irradiated with 3Gy or 5Gy radiation, followed by treatment 20 with Example 6 (300 n.M and 1 pM) 0.5 hs post-radiation. Samples can then be collected at 0,5, 1, and 4 hours after the addition of Example 6, The cells can then be lysed on ice in RIPA buffer (150 mM NaCl, 1% NP-40, 0,5% Sodium deoxycholate, 0. 1% SDS, 50 mM Tris pH 8,0) plus inhibitor cocktail (Protease Inhibitor Cocktail Set 111, Calbiochem), and 1 mM NaV& 1 can then be quantitated using the BCA protein assay kit (Pierce 25 #23225). Samples can be resolved by electrophoresis (15 pig protein) using a 4-12% bis tries gel(Novex #NP0336) with MES SDS buffer (Novex, #NP0002) at 140 volts, and then transferred to a nitrocellulose membrane (Biorad, #162-0145) by semi-dry transfer (18 volts for 35 minutes) using 2X transfer buffer (Novex, #NP0006), Membranes can then be blocked for I hour at room temperature in Odyssey Blocking Buffer (Licor # 927-40000) 30 diluted I 1 with IX TBS and then incubated overnight at 4*C with both primary antibodies in Odyssey Blocking Buffer diluted 1:1 with IX TBS-T 0.05%, The next day, membranes can be washed four limes with IX TBS-T 0.2% for 10 minutes each wash, and then incubated with both secondary antibodies at 1:10,000 (in Odyssey Blocking Buffer diluted 1:1 with 1 X TBS-T 0.05%) for 1.5 hours at room temperature protected -24 from light, Blots can be washed four times with 1X TBS-T 0,2% for 10 minutes each wash (protected from light) and then read on the Odyssey Infrared Imager. GAPDH can be visualized using the 800nm signal and the phospho-H2AX then detected with 700nm. Size expected for phospho histone 1H2AX is 15kDa and GAPDH is 36kDa. 5 Example 5 Cell Cycle Analysis U387MG cells can be irradiated at 30y or 5Gy radiation and then treated with Example 6 (300 nM and I pM) 0.5 hours post-radiation. Samples can then be collected 8, 10 24, and 48 hours (or any times determined by one skilled in the art) afier the addition of Example 6, Cells can be fixed in 100% ethanol overnight at 4*(. The next day cells can be incubated with cell cycle reagent (Guava Technologies #4500-0220) for I hour at room temperature protected from light. Stained nuclei are analyzable by flow cytometry (Guava EasyCyte; using settings known to one skilled in the art, for example 427 X8; acquisition 15 data 5,000 events/sample). The percentage of cells in each phase of the cell cycle can be determined using Cell Cycle analysis software (Guava Technologies). Example 6 7-Methoxy-12,3,1 1-tetrahydro-5,1I-diaza-benzojaitrinden-4,6-dione 20 ?tO~ 0 "N Step 1: To a cooled (-78 0 C) solution of 4-methoxyindole (210 g 13.1 mmol) in dry THF (20 mL.) was slowly added nBuLi in hexanes (2.5 M, 52 maL, 13,1 mmol), The mixture 25 was stirred at -78 0(7 for another 30 min, and CO 2 gas was then bubbled into the reaction mixture for 15 min, followed by additional stirring of 15 min. Excess CO 2 and half the THY volume was removed at reduced pressure, Additional dry THF (10 mL) was added to the reaction mixture that was cooled back to -78 'C. 1.7 M t-BuLi (7,7 mL, 13.1 mtmol) was slowly added to the reaction mixture over 30 min, Stirring was continued for 2 h at 30 78 0C, followed by slow addition of a solution of cyclopentanone (17 g, 20,4 nmol) in dry THF (5 mL). After an additional stirring of lh at -78 C the reaction mixture was quenched by dropwise addition of water (5 mL) followed by saturated NH4CI solution (20 mL), Ethyl ether (50 mL) was added and the mixture was stirred for 10 min at room temperature. The organic layer was separated, dried (MgSO4) and concentrated to give a 5 mixture of alcohol (1~(4-methoxy-1Hindol-2~y)cyclopentanol) and diene (2-cyclopent 1-enyl-4-methoxy~1H-indole). To the mixture in acetone (15 mL) was added 2 N HCI (5 mL), The mixture was stirred for another 10 min, water (50 mL) was added and the diene product 2-cyclopent-1-enyl-4-methoxy-iH-indole collected and dried under vacuum. The product was purified by silica gel chromatography (EtOAC/hexanes 9;1) 'H NMR 10 (DMSO-d6) 6 1,9-2.1 (m, 3 H) 26-2,75 (m, 3Hb, 3 (s, 3H) 6.1 (s, 1), 6.3 (s, 1H), 6.4 (m, IH), 6.9-7.0 (m, 2H), 11.1 (s, IH). This product was used directly in the next step. Step 2: A mixture of 2-cyclopent- 1 -enyl-4-methoxy- I 4-indole (0.1 g, 0.47 mmol) and maleimide (0.0.9 g, 0.91 rmmol) in acetic acid (5 mL) were stirred for 1 hour at room temperature. Water was added and the product extracted with EtOAc, which was washed 15 with 2 N NaCO solution, water, and saturated NaCI solution and dried (MgSO4), The drying agent was removed by filtration and the solvent concentrated to give 0.13 g MS: m/z 309 (M ~ H). Stcp 3: The product from step 2 (0.123 g, &0A mmol) in a toluene (2 mnL) and acetic acid (3 rL was added 2,3-dichioro-5,6-dicyano-1,4-benzoquinone (DDQ, 185 mag, 0.8 mmol). 20 After stirring 30 rin at 0 C the mixture was concentrated and treated with EtOAC and ascorbic acid. After 30 min the mixture was made basic with 2 N Na2C0 3 The EtOAc layer was washed with water. saturated NaCl solution, dried (MgSO 4 ) and concentrated to give the product 0,095 mg; MS: nz 305 (M -H) 'H1 NMR (DMSO-d6) 6 2.26-2.31 (m, 2H), 3,1-3.2 (m, 2H), 3.3~3.4 (m, 2H), 3,9 (s, 3H), 6,7 (M, 1H), 7.1 (m I H), 6.4 (in, I H), 25 7.4 (m, I H), 10,6 (s, IH14), 11,9 (s, 114), Example 7 7-Methoxy-5-(4-mnethyl-piperazin-4I-ybmethyl)4,2,3,11l-tetrahydro-5,t 1-diaza~ benzolaltrindene-4,6-dione - 26 cN' To a slurry of Example 6 (10.0 g 30 mmol) and N-methylpiperazine (12.4 g, 124 mml) in ethanol (950 mL.) was added paraformaldehyde (5.60 g, 62-4 mmol) in 0.5 hr 5 and stirred 24 hr. The slurry was evaporated to dryness. To the residue was added hexane (500 mL), sonicated 15 min, stirred 1.5 hr. and cooled at 0"C for 15 Pm. A yellow solid was collected and washed with cold hexane. This product was dissolved in warm tetrahydrofuran (THF) (250 mL) and filtered, The filtrate was added dropwise into hexane (3 L), stirred 15 rain., and Example 7 collected the precipitate and washed with hexane 10 (120 g, 96% yield), H NMR (DMSO-d4) 2 12 (s,3H), 2.35 (m,8H) 2,53 (m,4H), 3.18 (m;'2H), 4,44 (s,3H), 630 (d, H), 7.10 (diH), 7.40 (t,1H), 11.96 (s,1H1). MS miz 419 (M Example 8 15 7~Methoxy~5-(diethylaminomethyl)-1,2,3,1 -tetrahydro-5,1 I -diaza-benzo[altrindene. 4,6-dione (Ex. Sa) 7-Methexy-$,11(bis-diethy amitneehyl)41,2,3,11-tetrahydro~5,11-diaza benzofaltrindene-4,6-dione (Ex 81) /CHC, 27- To a slurry of Example 6 (50 mg, 0,16 mmol) in DMF (5 mL) was added paraformaldehyde (73 mg, 0.81 mmol), diethylamine (84 pL 0;81 mmol) and stirred at room temperature for I day. The reaction was evaporated and the residue triturated with hexane and evaporated to give two products as an oil, (ratio 6-1, 1 b: 16c) 'H-NMR 5 (DMSO4d) 0,98 (t,3H4),1 11 (t,3), 2.27 (m,21), 2,53 (m,8H), 257(m15H), 3,17 (t,2H) 3.50 (mIH) 3,97 (s,3H), 4.14 (d,2H), 4.71 (d,211, 6,82 (t,2H), 6.75 (d,2H), 713 (d,21), 7.33 (mH), 7.46 (t,3H)7.52 (m,1H), 11.95 (s,1H) 16b: MS inz 392, 16c MS r/z 476. Example 9 10 7-.Methoxy-5,11-(bis-morpholin-4-yhnethyl)-,2,3,11-tetrahydro-5,11-diaza~ benzo ja]trindene-4,6-d lone i5 To a sturry of Example 6 (15 mg, 0.049n-mmol1) in DMF (I. mL) was added paraformaldehyde (42 mg, 0.05 pL[), morpholine (160 mng, 1.9 mmol) andi heated at 70 "C ,for 18 hr. Thbe mixture was evaporated. The residue was triturated with hexane, then dissolved in C12C12, fibered and evaporated, The residue was triturated with Et2OQ and Example 9 collected as a yellow solid (5 mg, 20%)j HNMR (DMSO-d-152(1H,73 20 (d, I H).6 82 (d, I H), 5.0 (s, 2H4), 4.46 (s. 2H), 3.98 (s, 3H), 3 , 6 (s, 6H-), 3,49 (s, 4H), 2,50 (,61-), 2-49 (s, 4H1), 2.45 (m, 2H); MS miz 505 (M + H1), Example 10 25 benzolaltrindene-4,6-dione -28~ To a slurry of Example 6 (50 mgO 1 6 mmol) in ethanol (10 mIL) was added parafo rmaldehyd e (72 mg, 0, m mot), morph.-line ( 100 g, I1.1 mol1) and heated at IS0*T for 5 5.hr, The reaction was evaporated, water added (1-5 mLf.) and a yellow solid collecd (59 mg). 'H NMR (DMSO-ds,) 11,98 (s, 1H), '7,45 (t, 11-), 7. 13 (d, 1H-), 6,75 (d, I H), 4,44 (s, 2H)97 (s, 3 H), 3.5 6 (s, 4 h),3 18 (t, 2 h) 229 (t, 2 h). MS mn/7, 4 06 (M + H.). Example 11I 10 Measu rement of PARP Enzy matic Activ ity. PARP activity,4was monitored by transfer of radiolabeled ADP-ribose imits from [EPNA* t aproei acepor uc ashiton o PAP tsef.The assay mixtures contained 100 mM Tris (p1 mM DTT; 10 mMN1 MgOl,,20 ugiml DNA (nicked by sonication), '20 mg/mil histone H-1, 5 ng recombiant human PARPI, and inhibhor or is DMISO (< 2.5% (v/v)) in a final volume of 100 til. The reactions were initialed by the addition~~~~~~~~ of10p A*splmne ih2u [PNAD' ml and maintained at room temperature for 12 minutes. Assays we.vre terminated by the addition of 100 g-M of 50% T'CA and the radiolabeled precipitate was collected oni a 96-well filter plate (Millipore, MADP NOB 50), washed with 25% TCA, The amournof acid-ins'oluble 20 radioactivity, corresponding to polyADP-ribosylated protein, vwas quantitated in a Wallac MicroBeta scintillation counter, Determination of ICRI for Inhibitors& Single-point inhibitionidata were calculated by comparing PARP, VEGFR2, fir 25 MILK3 activity in the presence of Inhibitor to activity in the presence of DMSO only, Inhibition cuirves for compounds were generated by plotting percent inhibition versus logof of the concentration of compound. IC-, values were calculated by nonlinear regression using the sigmoidal dose-resiponse (variable slope) equation in GraphPad Prism as follows: Y::= bottom + (top - botm/1+ 10 ^ 4") ~ 9
CI~
where y is the % activity at a given concentration of compound, x is the logarithm of the concentration of compound, bottom is the % inhibition at the lowest compound concentration tested, and top is the % inhibition at the highest compound concentration examined. The values for bottom and top were fixed at 0 and 100, respectively, iCjo 5 values are reported as the average of at least three separate determinations. Using the assays disclosed herein the following Table 2 demonstrates the utility of compounds of the invention for PARP inhibition. Compounds of the present invention are considered active if their 1(59 values are less than 50 tM., In the following Table, for the inhibition of PARP, compounds of the present invention with a "+" are less than 10000 10 nM; compounds of the present invention with a "++" are less than 1000 nM; and compounds of the present invention with a "+++" are less than 100 nM in ICu for PARP inhibition. Where no ICS value is represented, data has yet to be determined, Table 2 Example No. PARP 1I29 (UM) 6 6 .. 7 7++ 8 8a/8b + 9 9) +++ . ................................... +... . . . . . . 10 10 15 Example 12 A preliminary study was conducted to determine the radio-sensitizing ability of orally administered Example 7. 20 Tumnor Cell inplanation and Growth Exponentially growing cells were harvested and injected (2x10 ellsmouse) into the right flank of commercially available athymic NCR. nu/nu nude mice. Animals bearing tumors of 200-400mn 1 were randomized according to size into the appropriate 25 treatment groups (n=4). Tumors were measured every 3-4 days using a vernier caliper. Tumor volumes were calculated using the following formula: V=ab/2, where a and b are the short and long dimensions, respectively. -30- Methods: U87MG human glioblastoma cells were injected subcutaneously (suc,) into the right hind limb of athymic NCR nuhin nude mice and allowed to grow to a mean tumor volume of 200mmnr Mice that received radiotherapy were anesthetized prior to 5 irradiation with 100 mg/kg Ketamine A 10 mg/kg xylezine or 37.5 mg/kg Ketamine + 0,2 mg/kg acepromazine. s.c. to provide 25-30 min of sedation. Anesthetized mice were positioned in malleable lead shielding which conforms to the animals body size and shape without undue pressure. The body was shielded by lead. The tumor bearing leg or exposed tumor was irradiated with the appropriate dose. After tumors were irradiated, the mice are 10 returned to cages on heating pads until recovered from the anesthetics. Example 7 was given as soon possible (within 30 miin) after radiation (R), Mice were randomized into the following treatment groups and administered: 1) vehicle , 2) RT (2 Gy X$d), 3) RTr plus Example 7 (200 or 300 mg/kg p.o, dose equivalents of Example 6, qd X21d) or 4) Example 7 (200 or 300 mg/kg dose equivalents of Example 6p.o, qd X2ld ) only, 15 Example 7 was given on days 1-21 and RT was given on days 1-5. All of the animals were measured on the same day. Individual tumor volume measurements were modeled in a log transformed linear model and the best fit time for tumors to reach approximately 2000 mm 3 was determined, 20 Results: As shown in Figure 5, administration o Example 7 (Cep 300; 300 mg/kg dose equivalents of Example 6 p.o, qdX21 d) plus R T (2 Gy X5d) resulted tumor growth stasis starting on day 8 and continuing throughout the study (day 31), while adminiastration of Example 7 (Cep 200; 200 mg/kg dose equivalents of Example 6 po. qdX21 d) phs RT (2 Gy X5d) and Example 7 alone (200 and 300 mg/kg dose equivalents 25 of Example 6 p.o. qdX2 I d) had no effect on tumor growth as compared to RT alone. The obtained indicating that Example 7 administration only had no ef'eet on tumor growth confines data obtained from s.c. dosing, Those skilled in the art will appreciate that numerous changes and modifications 30 can be made to the preferred embodiments of the invention and that such changes and modifications can he made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention. -31- All references cited herein are hereby incorporated herein in their entireties by reference. REFERENCES 5 Curtin, NJ. (2005) PAR? inhibitors for cancer therapy Expert Rev Mlec Med 4: 1-20. Griffin, R et at, (1998) Resistance-modifying agents. 5, Synthesis and biological properties of quinazolnone inhibitors of the DNA repair enzyme poly(ADP rihose)polymerase (PARP),. Med Chem, 42: 5247-5256 10 Bowman, KJ, et at (1998) Potentiation of anti-cancer agent cytotoxicity by the potent poly(ADP-ribose)polymerase inhibitosr NU 1025 and NIU 1064 Br, J. Cancer 78: 1269 1277. 15 Bowmain, KJ, et at (2001) Differential effects of the poly(ADP-ribose)polymerase inhibitor NU, 1025 on topoisomerase I and 11 inhibitor cytotoxicity in L1210 cells in vitro Br. 1 Cancer 84: 106-112. Chen & Pan (1988) Potentiation of antitumor activity of cisplatin in mice by 3 20 amiinobenzamide and nictotinamide Cancer Chemoth Pharmacol. 22: 303-307. Delaney, CA et at, (2000) Potentiation of temozolomide and topotecan growth inhibition and cytotoxicity by novel poly(adenosine diphosphoribose)poymerase inhibitors in a panel of human tumor lines CHUn Cancer Res. 6: 2860-2867. Griffin R et al, (1995) The role of inhibitors of poly(ADP-ribose)polymerase as resistance-modifying agents in cancer therapy Biochimie 77: 408-422. Liu, L, et at, (1999) Pharmacologic disruption, of base excision repair sensitizes mismatch 30 repair-deficient and-proficient colon cancer cells to methylating agents Clin. Cancer Res. 5: 2908-2917, - 2 Tentori, L et at (2002) Combined treatment with temozolomide and poly(ADPribose) polymerase inhibitor enhances survival of mice hearing hematologic malignancy at the central nervous system site Blood 15: 2241-2244, 5 Baldwin J. (2002) FDA evaluating oxaliplatin fbr advance colorectal cancer treatment I Vat!. Cancer Inst. 94: 1191-1193, 2002, Jagtap P and Szabo C (2005). Poly (ADP-Rihose) polymerase and the therapeutic effects of its inhibitors. Nature Rev Drug Disc 4: 421-440. 10 Ame JC, et al (2004). The PARP superfamily. Bioessays 26: 882-893. Smith., S. (2001) The World According to PARP Trends Biochm.Sc? 26: 174-179, 15 Virag, L and Sabo, C. (2002) The therapeutic potential of poly(ADP-ribose) polymerase inhibitors Pharmaocoi. Rev. 54: 375-429, de Murcia, JM, et at, (1997) Requirement of poly(ADP-ribose)polymerase in recovery from DNA damage in mice and cells Proc Nod AcadSci USA 94: 7303-7307, 20 Masuntani , et al. (2000) The response of PARP knockout mice against DNA damaging agents Mural, Res. 462: 159-166. Kato, T et at (1988) Enhancement of bleomycin activity by 3-aminobenzamide, a 25 poly(ADP-ribose) synthesis inhibitor, in vitro and in vivo Anticancer Res. 8: 239-244. Smith, L et al (2005) The novel poly(ADP-Ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks CI Cancer Res. 11: 8449-8457, 30 -33 -

Claims (24)

1. A method of treating cancer by administering a radiosensitizing agent of Fonnula (I): H3CsO O N/ 0 N O N H 5 (I) or a pharmaceutically acceptable salt form thereof, wherein, X is -1 or a prodrug moiety; to a mammal suffering from cancer and applying ionizing radiation to said mammal tissue. 10
2. The method of Claim 1 wherein said radiosensitizing agent is present within or proximate to said tissue increases the efficiency of conversion of said applied ionizing radiation into localized therapeutic effects.
3. The method of Claim 2 wherein said radiosensitizing agent is present in an amount 15 effective to radiosensitize cancer cells.
4. The method of Claim 3 wherein ionizing radiation of said tissue is performed with a dose of radiation effective to destroy said cells. 20
5. The method of Claim 4 wherein said ionizing radiation is of clinically acceptable or recommended radiotheraputic protocols for a given cancer type.
6. The method of Claim 4 wherein said cancer is malignant. 25
7. The method of Claim 4 wherein said cancer is benign.
8. The method of Claim 1 wherein the prodrug moiety is selected from the group consisting of -CH2NR'R 2 , -CH 2 0C(=O)R 3 , -CI-12OP(=O)(OH) 2 , and -C(=O)R 4 ; wherein; - 34 - R' is B. or C alkyl; R 2 is H or C alkyl; alternatively, R and R, together with the nitrogen atom to which they are attached, form a heterocyclyl group selected from pyrrolyl, pyrrolidinyl, piperidinyl, 5 morpholinyl, thiomorpholinyl, and piperazinvl, wherein said heterocyclyl group is optionally substituted with C4 alkyl; R is selected from the group consisting of -C4 alkyl-NR 1 R -C 1 4 alkyl-ORK pyridinyl, -phenyl(CH 2 NR 1 R 2 ), and -CH(R ')NH 2 ; R4 is selected from the group consisting of O-(C, alkyl)~NR'R 2 , -O-(Cz., alkyl) 10 OR', and -CH(R)NHt; Re is H or C1 alkyl; and R5 is the side chain of a naturally occurring amino acid,
9, The method of Claim I wherein the prodrug moiety is -CH2NR I 15 R1 is H or CM alkyl; R is H or Ct alkyl; and alternatively, R and R , together with the nitrogen atom to which they are attached, form a heterocyclyl group selected from pyrroly, pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholiny, and piperazinyl, wherein said heterocyclyl group is 20 optionally substituted with Ct alkyl,
10, The method of Claim I wherein the prodrug moiety is a Marnich base,
11. The method of Claim 8 wherein the Mannich base is selected form 4-methyl 25 piperazin -1-ylmethylk morpholin-4-yimethylb and 5-diethylaminomethyl,
12, The method of Claim & wherein the Mannich base is 4-nethyl-piperazin- 1 -yimethyl,
13. A method of ClRim 1 wherein the route of administration is intravenous, subcutaneous, 30 oral or intraperitoneally,
14, A method of Claim I wherein the route of administration is intravenous, ~-35 -
15. A method according to Claim I wherein said cancer is selected from head and neck squamous cell carcinoma (eye, lip, oral , pharynx, larynx, nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (epidermis), glioblastoma, astrocytoma, oligodendroglioma, oligoastrocytoma, meningioma, 5 neuroblastoma, rhabdomyosarcoma, soft-tissue sarcomas, osteosarcoma, hematologic malignancy at the cns site, breast carcinoma (ductal and carcinoma in situ), thyroid carcinoma (papillary and follicular), lung carcinoma (bronchioloalveolar carcinoma, small cell lung carcinoma, mixed small cell/large cell carcinoma, combined small cell carcinoma, non-small cell lung carcinoma, squamous cell carcinoma, large cell carcinoma, 10 and adenocarcinoma of the lung), hepatoceilular carcinoma, co-lorectal carcinoma, cervical carcinoma, ovarian carcinoma, prostatic carcinoma, testicular carcinoma, gastric carcinoma, pancreatic carcinoma, cholangiosarcoma, lymuphoma (Hodgkins and non Hodgkins types of T-and B-cell origin), leukemia (acute and chronic leukemias of myeloid and lymphoid origins), and bladder carcinoma, 15
16. A method according to Clain 1 wherein said cancer is selected from head and neck squamous cell carcinoma (eye, lip, oral, pharynx, larynx, nasal, carcinoma of the tongue, and esophogeal carcinoma), melanoma, squamous cell carcinoma (epiderms, glioblastoma, neuroblastoma, rhabdomyosarcoma, lung carcinoma, (bronchioloalveolar 20 carcinoma, small cell lung carcinoma mixed small ceIl/large celi carcinoma, combined small cell carcinoma, non-small cell lung carcinoma, squamous cell carcinoma, large cell carcinoma, and adenocarcinoma of the lung), lymphoma (Hodgkins and non-Hodgkins types of T-and B-cell origin), and leukemia (acute and chronic leukemias of nyeloid and lymphoid origins). 25
17, A method of treating cancer by administering a radiosensitizing agent of formula 7 imethoxy-1,2, 3,11 -tetrahydro-5,1 -diaza-benzo ajtrindene-4,6-dione, or a pharmaceuically acceptable salt frmi thereof, to a mammal suffering from cancer, and applying radiation to said mammal 30
18 A method of treating cancer by administering a radiosensitizing agent of formula 7 nmethoxy-5~(4-miethyl-piperazin- I-ylmethyfl)~1,2,3,11-tretrahydro-5,1 1-diaza benzo[altrindene-4,6-dione, or a pharmaceutically acceptable salt form thereof, to a mammal suffering from cancer, and applying radiation to said mammal, - 36
- 19, A phannacetical composition for radiosensitizing cancer cells comprising a radiosensitizing amount of a compound of Formula (1); H3C , 0 ox a pharmaceutically acceptable salt form thereof, wherein X is H1 or a prodrug moiety and a pharmaceutically acceptable carrier, 10
20. The phracu ica position of C13aim1 19 Wherein the prodnag moiety is selected from the group consisting of -CJ.4,NR 1R2, CH2OC(=)R -CH2O =0 and C(=O)R R' is H or C _ alkyl; R2is 1-1 or C Ialkyl; 15 alternatively, R" an~d Re, together withi the nitrogen atom to which they are attached, form a heterocyclyl. group selected from pyrrolyl1, pyrrOlidinyL, piperidinyl, morphoinyl, yhiomorpholinyl, an tierof, wherein saod pro ygrog p is opionallysubstituted with Cap a alkyl; R d is selected from the group consisting of -CHNakyRNR R"C ' alkyl-OR' 20 pyridinyl, -phenyl(CHlNRoR), and -CH(R)NR; R4 is selectd from the group consisting of-O-(C alkyl)-NR'Rz -0-(C4 alkyl) OR , and -CH(R6)N142 R' is H or C, alkyl; and ais the side chain ofa naturally occurring amino acid,
21 The pharmaceutical composition of Caim 19 we cghrein the prodrug moiety is -CHNR in R' is H r C the ginlkya; R is H or C% alkyl; and - 37 - alternatively, R and R, together with the nitrogen atom to which they are attached, form a heterocyclyl group selected from pyrrolyl, pyrroldinyl, piperidinyl, morpholinyl, thiornorpholinyl, and piperazinyl, wherein said heterocyclyl group is optionally substituted with CM alkyl, 5
22. A pharmaceutical composition as set forth in Claim 19 wherein the compound is 1-bC.. 0 I N' H 10 or a phannaceutically acceptable salt form thereof
23. A pharmaceutical composition as set forth in Claim 19 wherein the compound is lt- N -N N H C) 0 H 15 or a pharmaceutically acceptable salt form thcreof
24, A compound of Formula (11): 38 - N H 3 C-,0 0 N N N H (II) or a pharmaceutically acceptable salt form thereof. CEPHALON, INC. WATERMARK, PATENT AND TRADE MARKS ATTORNEYS P31872AU01 -39-
AU2014200040A 2006-11-20 2014-01-06 Method of radio-sensitizing tumors using a radio-sensitizing agent Abandoned AU2014200040A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2014200040A AU2014200040A1 (en) 2006-11-20 2014-01-06 Method of radio-sensitizing tumors using a radio-sensitizing agent

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/860,036 2006-11-20
AU2007321987A AU2007321987B2 (en) 2006-11-20 2007-11-20 Method of radio-sensitizing tumors using a radio-sensitizing agent
AU2014200040A AU2014200040A1 (en) 2006-11-20 2014-01-06 Method of radio-sensitizing tumors using a radio-sensitizing agent

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2007321987A Division AU2007321987B2 (en) 2006-11-20 2007-11-20 Method of radio-sensitizing tumors using a radio-sensitizing agent

Publications (1)

Publication Number Publication Date
AU2014200040A1 true AU2014200040A1 (en) 2014-01-23

Family

ID=49956575

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2014200040A Abandoned AU2014200040A1 (en) 2006-11-20 2014-01-06 Method of radio-sensitizing tumors using a radio-sensitizing agent

Country Status (1)

Country Link
AU (1) AU2014200040A1 (en)

Similar Documents

Publication Publication Date Title
AU2007321987B2 (en) Method of radio-sensitizing tumors using a radio-sensitizing agent
US20210100813A1 (en) Combination therapy for cancer using bromodomain and extra-terminal (bet) protein inhibitors
CA2802644C (en) Antitumor agent using compounds having kinase inhibitory effect in combination
RU2580609C2 (en) Anticancer therapeutic agent
CA3222841A1 (en) Use of atr inhibitors in combination with parp inhibitors for treating cancer
JP2018052974A (en) Combination products with tyrosine kinase inhibitors and their use
KR20190130621A (en) Combination of CHK1 Inhibitors and WEE1 Inhibitors
CN105658809B (en) Tumor selective combination therapy
CA3167746A1 (en) Compounds and uses thereof
JP6526789B2 (en) Combination therapy
AU2011323899A1 (en) Methods of treatment and/or prevention of scleroderma, UV injury or sunburn, formation of scars or keloids by using haloaryl substituted Aminopurines
IL293810A (en) Use of atr inhibitors in combination with parp inhibitors
WO2017041040A1 (en) Androgen receptor antagonists
AU2014200040A1 (en) Method of radio-sensitizing tumors using a radio-sensitizing agent
US20050176696A1 (en) Synergistic anit-cancer compounds
JP2024511466A (en) ALK-5 inhibitors and their uses
EP3538529B1 (en) Selective inhibitors of genotoxic stress-induced ikk/nf-kb pathways
EP3753563A1 (en) Anti-herpesvirus guanosine analogue for the inhibition of basal autophagy
CN116546986A (en) ALK-5 inhibitors and uses thereof
WO2023193114A1 (en) Methods of using atr inhibitors
CN116322680A (en) Combination regimens for PARP inhibitors
NZ786604A (en) Combination therapy with notch and cdk4/6 inhibitors for the treatment of
NZ786609A (en) Combination therapy with notch and pi3k/mtor inhibitors for use in treating cancer

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application