HK1117165A - Parp modulators and treatment of cancer - Google Patents
Parp modulators and treatment of cancer Download PDFInfo
- Publication number
- HK1117165A HK1117165A HK08112184.3A HK08112184A HK1117165A HK 1117165 A HK1117165 A HK 1117165A HK 08112184 A HK08112184 A HK 08112184A HK 1117165 A HK1117165 A HK 1117165A
- Authority
- HK
- Hong Kong
- Prior art keywords
- optionally substituted
- parp
- compound
- formula
- aromatic compound
- Prior art date
Links
Abstract
The invention relates to a method of modulating poly(ADP-ribose)polymerase-l (PARP-I) activity in a mammal comprising administering to a mammal an effective amount of an organic aromatic compound having from 4 to about 35 carbon atoms, wherein said organic aromatic compound is capable of binding the arginine-34 moiety located in Zinc finger- 1 of the PARP-I enzyme and wherein said organic aromatic compound has electron donating capabilities such that it's tr-electron system will interact with the positively charged (cationic) guanidinium moiety of the specific arginine-34 residue of the Zinc-1 finger of PARP-I and does not contain benzamide or lactam substituents. In particular, substituted benzopyrones and substituted indoles and their pharmaceutical compositions containing such compounds that modulate the activity of PARP-I, are described.; The invention is also directed to the composition of matter, kits and methods for their therapeutic and/or prophylactic use in treating diseases and disorders described herein, by administering effective amounts of such compounds. Preferably, the compositions and methods provided herein inhibit PARP activity.
Description
Cross-referencing
Priority of united states provisional application number 60/689,178 filed on 10.6.2005, this application is hereby incorporated by reference in its entirety.
Statement regarding federally sponsored research
The invention was made in part with the support of the U.S. government under the NIH funds HL 59693 and HL 35561.
Background
PARP (poly-ADP ribose polymerase) is involved in a variety of DNA-related functions including gene amplification, cell division, differentiation, apoptosis, DNA base excision repair, and also has an effect on telomere length and chromosome stability (d' Adda di Fagagna et al, 1999, Nature Gen.23 (1): 76-80). Oxidative stress-induced overactivation of PARP consumes NAD + and thus ATP, ultimately leading to cell dysfunction or necrosis. This cellular suicide mechanism involves the pathological mechanisms of stroke, myocardial ischemia, diabetes-related cardiovascular dysfunction, shock, traumatic central nervous system injury, arthritis, colitis, allergic encephalomyelitis and various forms of inflammation. PARP has also been shown to be associated with and regulate several transcription factors. The diverse functions of PARP make it the target of a variety of serious conditions, including various types of cancer and neurodegenerative diseases.
PARP-inhibition therapy is an effective method of treating a variety of diseases. In cancer patients, PARP inhibition may increase the therapeutic effect of radiation and chemotherapy. Targeting PARP can prevent tumor cells from repairing the DNA itself and from developing resistance, which can make them more susceptible to cancer treatment. PARP inhibitors have been demonstrated to have the ability to increase the effect of various chemotherapeutic agents (e.g., methylating agents, DNA topoisomerase inhibitors, cisplatin, etc.) as well as radiation therapy on a broad spectrum of tumors (e.g., glioma, melanoma, lymphoma, colorectal cancer, head and neck tumors).
The incidence of breast cancer in women has risen from 100.5 per 100000 in 1991 to 117.2 per 100000 in 2001; the average annual increase is 1.4%. Women with defects in BRCA1 and the 2 gene have up to 85% of the likelihood of developing breast cancer by age 70. PARP inhibitors can effectively kill tumor cells in humans that have been deficient in BRCA1 and BRCA 2. PARP inhibitors have the potential to help a particular subset of patients with these gene mutations. These mutations predispose patients to early onset of cancer and have been found in breast, ovarian, prostate, and pancreatic cancers.
PARP inhibitors can be combined with other chemotherapeutic agents, such as irinotecan or temozolomide, respectively, to improve the treatment of many cancers, such as colorectal and gastric cancers, as well as melanoma and glioma. PARP inhibitors may be combined with irinotecan to treat advanced colorectal cancer. Approximately 146000 new cases of colorectal cancer are expected in the united states in 2004, with 60-70% expected to be in advanced stages.
PARP inhibitors have been designed as analogs of benzamide that competitively bind with the natural substrate NAD at the catalytic site of PARP. This includes a number of cyclic benzamide analogs (i.e., lactams) that are potent inhibitors at the NAD site. However, the method using benzamide analogues is limited in vivo efficacy. These benzamides and lactams can bind to other ubiquitous NAD-utilizing enzymes and produce side effects and affect cell viability, metabolism and DNA synthesis. See Milan et al, (1984) "Inhibitors of Poly (Adenosine phosphate Ribose) Synthesis: effect on Other Metabolic Processes ", Science 223: 589-91. Thus, there remains a need for compounds that inhibit PARP activity with less side effects and produce effective and reliable results in inhibiting PARP activity and treating related diseases and conditions.
Accordingly, the present invention provides compositions and methods for modulating PARP activity in a mammal suffering from a PARP mediated disease.
Summary of The Invention
The present invention relates to a pharmaceutical composition comprising: (i) an effective amount of an organic aromatic compound containing 4 to about 35 carbon atoms that modulates PARP-1 activity in a mammal, wherein said organic aromatic compound (a) is capable of binding to an arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, and (b) wherein said organic aromatic compound has an electron donating ability such that its pi-electron system interacts with a positively charged (cationic) guanidinium (guanidinium) moiety of the specific arginine-34 residue referred to as zinc-1 of PARP-1, (c) wherein said aromatic compound comprises a heterocyclic ring containing a nitrogen atom, (d) said ring does not contain a carbonyl moiety; and (ii) a pharmaceutically acceptable carrier, excipient and/or diluent. Preferably, the compositions of the present invention inhibit PARP activity.
The present invention also relates to a method of modulating PARP-1 activity in a mammal comprising administering to the mammal an effective amount of an organic aromatic compound having from 4 to about 35 carbon atoms, wherein the organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, and wherein the organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of the specific arginine-34 residue referred to as zinc-1 of PARP-1, wherein the aromatic compound comprises a heterocyclic ring containing a nitrogen atom, which ring does not contain a carbonyl moiety and does not contain a lactam structure, and is not a benzamide analog, nor an analog of NAD. The compounds of the invention act through an ATP binding site and may or may not interact with an NAD site. Preferably, the methods of the invention inhibit PARP activity.
The present invention relates, inter alia, to a method of modulating PARP-1 activity in a mammal comprising administering to the mammal an effective amount of an organic aromatic compound of formula I or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof,
formula I
Wherein the organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein the organic aromatic compound has an electron donating ability such that its pi-electron system will interact with the positively charged (cationic) guanidinium moiety of the specific arginine-34 residue referred to as zinc-1 of PARP-1; wherein R is1、R2、R3And R4Independently selected from H, halogen, optionally substituted hydroxy, substituted amino, optionally substituted lower alkyl, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group. Preferably, the methods of the invention inhibit PARP activity.
In a preferred embodiment, the present invention relates to a compound of formula I or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof,
wherein R is1、R2Is H.
Another aspect of the present invention relates to a method of modulating PARP-1 activity in a mammal comprising administering to the mammal an effective amount of an organic aromatic compound of formula II or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof, wherein the organic aromatic compound is capable of binding to an arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein the organic aromatic compound has an electron donating ability such that its pi-electron system interacts with a positively charged (cationic) guanidinium moiety of a specific arginine-34 residue in the zinc finger-1 of PARP-1,
formula II
Wherein R is1、R2、R3、R4And R5Independently selected from H, halogen, nitro, nitroso, optionally substituted hydroxy, optionally substituted lower alkyl, optionally substituted amino, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group; x is H, N-oxide or optionally substituted alkyl. Preferably, the methods of the invention inhibit PARP activity.
In a preferred embodiment, the invention relates to a subgroup of compounds of formula II, as shown in formula IIa, or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof,
formula IIa
Wherein R is1And X is H, R2、R3、R4And R5Independently selected from halogen (preferably iodine), hydroxy, nitro, nitroso and optionally substituted amino (e.g. aminoalkyl).
A particularly preferred group of compounds of the formula IIa is that in which R2Is alkylamino, preferably propylamino.
Another preferred class of compounds of the formula IIa is that in which R3、R4Or R5Is halogen, preferably iodine.
Another preferred class of compounds of the formula IIa is that in which R3、R4Or R5Is a hydroxyl group.
One aspect of the present invention is a method of treating a PARP mediated disease comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound having 4 to about 35 carbon atoms, wherein the organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein the organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of a particular arginine-34 residue in the zinc-1 finger of PARP-1, wherein when the aromatic compound comprises a heterocyclic ring containing a nitrogen atom, the ring does not contain a carbonyl moiety.
Another aspect of the present invention is a method of treating PARP mediated diseases comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound of formula I, wherein the organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof, wherein the organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of the particular arginine-34 residue referred to as zinc-1 of PARP-1,
formula I
Wherein R is1、R2、R3And R4Independently selected from H, halogen, optionally substituted hydroxy, substituted amino, optionally substituted lower alkyl, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group.
Another aspect of the present invention is a method of treating PARP mediated diseases comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound of formula II, or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof, wherein said organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein said organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of the specific arginine-34 residue referred to as zinc-1 of PARP-1,
formula II
Wherein R is1、R2、R3、R4And R5Independently selected from H, halogen, nitro, nitroso, optionally substituted hydroxy, optionally substituted lower alkyl, optionally substituted amino, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substitutedC of (A)3-C8A cycloalkyl group; x is H, N-oxide or optionally substituted alkyl.
Particularly preferred examples of compounds of the present invention include, but are not limited to, the following compounds:
is incorporated by reference
All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
Brief Description of Drawings
The novel features of the invention are set forth with particularity in the appended claims. The features and advantages of the present invention will be better understood by reference to the following detailed description of illustrative embodiments in which the principles of the invention are utilized and the accompanying drawings, of which:
FIG. 1 is a graph illustrating the enzymatic activity of wild-type arginine-34 and arginine-138 mutated PARP-1.
FIG. 2 is a graph illustrating the effect of ATP on PARP-1 activity in Jurkat nuclei.
FIG. 3 is a graph illustrating the effect of BCNU on ATP sensitivity of PARP-1 activity of Jurkat nuclei.
FIG. 4 is a graph illustrating the effect of ATP on the glycohydrolase activity of Jurkat cell nuclear extracts.
Figure 5 is a graph illustrating the effect of chain length of PAR polymers on ATP sensitivity of purified PARG.
FIG. 6 is a graph illustrating the effect of ATP on PARG activity as a function of substrate (PAR) concentration.
FIG. 7 is a graph depicting one embodiment of the interaction between an aromatic π -system and the cationic guanidinium moiety of PARP-1, where X ═ OH or NH2。
Detailed Description
The term "alkyl" as used herein refers to straight and branched chain alkyl groups having from 1 to 8 carbon atoms. Examples of alkyl groups include methyl (Me), ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and the like. Substituted alkyl groups include aminoalkyl, hydroxyalkyl, alkoxyalkyl, and the like. Substituted alkyl also represents, for example, substituted or unsubstituted C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl, phenyl or C4-C10Alkyl substituted by heteroaryl.
The term "aminoalkyl" refers to-CH2-R-NH2Wherein R is an alkyl group as defined above.
The term "cycloalkyl" refers to a saturated carbocyclic ring of 3 to 8 carbon atoms, including bicyclic and tricyclic cycloalkyl structures. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
The term "halogen" refers to chlorine, fluorine, bromine or iodine. The term "halo" refers to chloro, fluoro, bromo, or iodo. The most preferred embodiment of the present invention includes iodine as the halogen group.
The term "heteroaryl" refers to the unsaturated or aromatic ring structures of mono-and poly-heterocyclic rings. Examples of the ring structure of the heterocycle include furyl, thienyl, pyrrolyl, nitrophenyl, pyridyl, pyrazolyl, imidazolyl, pyrazinyl, pyridazinyl, 1, 2, 3-triazinyl, 1, 2, 4-oxadiazolyl (oxadiazolyl), 1, 3, 4-oxadiazolyl, 1-H-tetrazol-5-yl, indolyl, quinolyl (quinolinyl), benzofuryl, phenylAnd thiophenyl (thioindenyl), and the like. These moieties may be optionally substituted with one or more suitable substituents, for example, selected from halogen (F, Cl, Br or I); a lower alkyl group; OH; NO2;CN;CO2H; o-lower alkyl; a phenyl group; phenyl-lower alkyl; CO 22CH3;CONH2;OCH2CONH2;NH2;SO2NH2;OCHF2;CF3(ii) a And OCF3And the like. These moieties may also optionally be fused ring structures or bridges such as OCH2-O.
The term "inhibit" or grammatical variations thereof, e.g., "inhibitory," is not intended to require a complete reduction in PARP activity. Such a reduction is preferably a reduction in molecular activity of at least about 50%, at least about 75%, at least about 90%, more preferably at least about 95% in the absence of inhibitory effects, e.g., in the absence of an inhibitor, such as compound I, II of the present invention and/or their preferred embodiments. Most preferably, the term refers to an observable or measurable reduction in activity. In a treatment regimen, it is preferred that the inhibitory effect is sufficient to produce a therapeutic and/or prophylactic benefit to the treated condition. The term "does not inhibit" or grammatical variations thereof does not require a complete lack of effect on activity. For example, it may refer to the case: in the absence of inhibitors, such as compound I, II of the present invention and/or their preferred embodiments, the reduction in PARP activity is less than about 20%, less than about 10%, preferably less than about 5%.
The term "pharmaceutically acceptable salts" refers to those salts that retain the biological effectiveness and properties of the compounds used in the present invention, and which are not biologically or otherwise undesirable. For example, pharmaceutically acceptable salts do not interfere with the beneficial effects of the compounds of the present invention in the treatment of cancer.
The term "pharmaceutically acceptable prodrug" refers to a compound that is converted to a specific compound or to a pharmaceutically acceptable salt of such a compound under physiological conditions or by solvolysis before exhibiting its pharmacological effect. The prodrugs are typically formulated for the following purposes: improved chemical stability, improved patient acceptability and compliance, improved bioavailability, extended duration of action, improved organ selectivity, improved formulation (e.g., increased water solubility), and/or reduced side effects (e.g., toxicity).
The term "pharmaceutically active metabolite" refers to a pharmacologically active product produced by the metabolism of a particular compound or salt thereof in the body. After entry into the body, most drugs are substrates for chemical reactions that can alter their physical properties and biological actions. However, in some cases, metabolism of the drug is required for therapeutic effect.
The term "therapeutically effective amount" refers to an amount effective to achieve a therapeutic or prophylactic benefit. Therapeutic benefit refers to the eradication or amelioration of the disease being treated. Eradication or amelioration of one or more of the physiological symptoms associated with the disease also achieves a therapeutic benefit, whereby an improvement in the patient's condition is observed, although the patient may still suffer from the disease. For prophylactic benefit, the composition can be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more physiological symptoms of a disease, even if the disease has not yet been diagnosed. The actual effective amount for a particular application will depend upon the patient (e.g., age, weight, etc.), the condition being treated, and the route of administration. Determination of an effective amount is well within the ability of those skilled in the art. An effective amount for a human may be determined from an animal model. For example, a dose for human use may be formulated to achieve circulating and/or gastrointestinal concentrations that have been found to be effective in animals.
Compositions and methods for PARP inhibitors
The present invention relates to a method for the preparation of a protein by binding an organic molecule to Zn localized to the PARP-1 enzyme2+Refers to arginine-34 in 1, to inhibit PARP-1. It is known that arginine residues in proteins can be involved in ATP sensing (Sensing) (Ogura et al (2004) J.struct.biol.146: 106-Two Zn2+Arginine residues were identified in all fingers without assigning a specific catalytic function (Molinet et al (1993) EMBO J.12: 2109-22117; Ikeyama et al (1990) J.biol.chem.265: 21907-21913). Analysis of the point mutation of arginine-34 showed Zn in PARP-12+In finger 1, the substitution of arginine-34 with other amino acids such as glycine did not affect the overall enzymatic activity of PARP-1, but the ATP inhibition was eliminated. Zn in PARP-12+In finger 2, the effect of the mutation of substituting arginine-138 with isoleucine on the inhibition of ATP was very slight, confirming Zn2+The idea that arginine-34 of finger 1 is the site of interaction of ATP with PARP-1.
The guanidino moiety of arginine is known to exhibit critical importance as a cation in cation-pi interactions (Zacharias et al (2002) Trends in pharmacological Sciences 23: 281-287; Woods et al (2004) J. proteome Res.3: 478-484). One aspect of the present invention relates to the inhibition of PARP-1 by cationic-pi interaction between the guanidino moiety of arginine and the pi-system of candidate molecules (as shown in figure 7, using 5-iodo-6-hydroxychromanone or 5-iodo-6-aminochromanone as examples). Replacement of the aromatic ring with electron donating substituents increases the electron density in the ring and thus the cationic-pi interaction with the guanidino moiety of arginine, thus increasing the inhibition of PARP-1 by using such organic aromatic molecules. Inhibiting the activity of PARP molecules includes decreasing the activity of these molecules.
The inhibition site at arginine-34 in zinc finger 1 of PARP-1 eliminates the need to inhibit PARP-1 at the NAD catalytic site, thus eliminating the need to use a benzamide or similar lactam, which competes with NAD in vivo and thus has disadvantages. The novel aromatic electron inhibitors at the arginine-34 position are a new class characterized in that they are designed to be free of benzamide or lactam groups. The compounds of the present invention are substituted 1, 2-benzopyranones, indoles or benzimidazoles which do not include a fusion with a third ring (i.e., are not tricyclic) and do not contain a lactam group; and are also not benzamide analogs, i.e., do not have a benzamide core.
Aromatic molecules that can act as pi-electron donors that interact with arginine-34 cations can be divided into two classes: (1) interaction inhibitors (preferably candidates for anti-cancer drugs), and (2) physiologically present molecules with aromatic groups that temporarily modulate PARP in response to cellular metabolic requirements. The choice of aromatic compound can be determined by the reactivity with the arginine-34 site, and the modification of the aromatic system can be determined by this reactivity. In general, kinetic evidence for reactivity with arginine-34 consists of additive inhibition of ATP (T.C.Chou and P.Talalay, adv.enzyme Regul.22: 27 (1984)).
In some preferred embodiments of the invention, the aromatic pi-system that interacts with the arginine-34 cation comprises a1, 2-benzopyranone (coumarin), such as formula I, an indole optionally substituted with iodine (formula II), or a benzimidazole optionally substituted with iodine (formula III).
Formula III
As noted, each moiety or functional group used for a variable in the formula may be optionally substituted with one or more suitable substituents. Examples of substituents include halogen (F, Cl, Br or I), lower alkyl, -OH, -NO2、-CN、-CO2H. -O-lower alkyl, -phenyl-lower alkyl, -CO2CH3、-CONH2、-OCH2 CONH2、-NH2、-SO2 NH2Haloalkyl (e.g., -CF)3、-CH2 CF3) -O-haloalkyl (e.g., -OCF)3、-OCHF2) And the like. The halogen is preferably an iodine group.
The present invention relates to a method of modulating, preferably inhibiting, PARP-1 activity in a mammal using an organic aromatic compound having from 4 to about 35 carbon atoms, including formula I, preferred embodiments thereof, formula II and/or preferred embodiments thereof, wherein said organic aromatic compound is capable of binding to an arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein said organic aromatic compound has an electron donating ability such that its pi-electron system will interact with a positively charged (cationic) guanidinium moiety of a particular arginine-34 residue of the zinc finger-1 of PARP-1, wherein when said aromatic compound comprises a heterocyclic ring containing a nitrogen atom, said ring does not contain a carbonyl moiety. The invention also relates to the therapeutic or prophylactic use of such compounds, and to methods of treating diseases and disorders associated with the activation of PARP.
Another aspect of the present invention is a method of treating PARP mediated diseases comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound of formula I, or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof, wherein the organic aromatic compound is capable of binding to an arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein the organic aromatic compound has an electron donating ability such that its pi-electron system interacts with a positively charged (cationic) guanidinium moiety of a particular arginine-34 residue of the zinc finger-1 of PARP-1,
formula I
Wherein R is1、R2、R3And R4Independently selected from H, halogen, optionally substituted hydroxy, substituted amino, optionally substituted nitro, optionally substituted lower alkyl, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group; and do not contain lactam groups nor carry lactam or benzamide substituents.
A preferred embodiment of formula I or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof:
wherein R is1、R2Is H.
Another aspect of the present invention is a method for treating PARP mediated diseases comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound of formula II or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof, wherein said organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein said organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of a specific arginine-34 residue of the zinc finger-1 of PARP-1
Formula II
Wherein R is1、R2、R3、R4And R5Independently selected from H, halogen, nitro, nitroso, optionally substituted hydroxy, optionally substituted lower alkyl, optionally substituted amino, optionally substituted nitro, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group; x is H, N-oxide or optionally substituted alkyl.
In a preferred embodiment, a subgroup of the compounds of formula II is represented by formula IIa, or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof,
formula IIa
Wherein R is1And X is H, R2、R3、R4And R5Independently selected from iodine, hydroxyl, nitro, nitroso and optionally substituted amino (e.g. aminoalkyl).
A particularly preferred group of compounds of the formula IIa is that in which R2Is an alkylamine, preferably propylamine.
In another preferred class of compounds of the formula IIa, R3、R4Or R5Is halogen, preferably iodine.
In another preferred class of compounds of the formula IIa, R3、R4Or R5Is a hydroxyl group.
Particularly preferred examples of compounds of the present invention include, but are not limited to, the following compounds:
the compounds of the invention may exhibit tautomerism. Although formulas I, II and IIa do not clearly describe all possible tautomeric forms, it should be understood that formulas I, II and IIa are intended to represent any tautomeric form of the compound described and are not limited solely to the specific compound form depicted by the structural formula diagram. Some of the compounds of the present invention may exist as individual stereoisomers (i.e., substantially free of other stereoisomers), racemates, and/or mixtures of enantiomers and/or diastereomers. All such individual stereoisomers, racemates and mixtures thereof are within the scope of the present invention. Preferably, the compounds of the invention having optical activity are used in optically pure form.
Furthermore, the formulae are intended to include solvated as well as unsolvated forms of the indicated structures. For example, formula I includes both hydrated and non-hydrated forms of the compounds of the structure. Other examples of solvates include structures in combination with isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine. In addition to compounds of formulas I, II and IIa, the present invention also includes pharmaceutically acceptable prodrugs, pharmaceutically active metabolites, and pharmaceutically acceptable salts of these compounds and metabolites.
The compounds described herein can be synthesized using techniques known in the art. Various substituents may be introduced into the aromatic nucleus of 1, 2-benzopyranones, indoles and benzimidazoles. In general, amino substituents can be introduced via standard nitration techniques followed by reduction of such nitro groups to amino groups. The amino group on the aromatic nucleus can be diazotized by Sandmeyer-type process and converted into a variety of other groups such as halogens and hydroxyl groups. In addition, halogenation can be performed directly on hydroxy-substituted and amino-substituted aromatic rings, giving di-substituted examples. Furthermore, a halogen group may be used as a leaving group to be substituted by a reagent (e.g., a metallo-organic) to introduce an alkyl group.
Poly (ADP-ribose) polymerase (PARP)
Poly (ADP-ribose) polymerase (PARP) is also known as poly (ADP-ribose) synthase and poly ADP ribosyltransferase. PARP is an enzyme located in the nucleus of various organs including muscle, heart and brain cells. PARP catalyzes the formation of poly (ADP-ribose) polymers that can attach to nuclear proteins (as well as to themselves) and thereby modify the activity of those proteins. The enzyme functions to enhance DNA repair, and may also function to regulate chromatin in the nucleus (see: D.D' ampoules et al "Poly (ADP-ribosylation interactions in the regulation of nuclear functions," biochem. J.342: 249-268 (1999)).
PARP-1 comprises an N-terminal DNA Binding Domain (DBD), a self-modifying domain and a C-terminal catalytic domain, and many cellular proteins interact with PARP-1. The N-terminal DNA binding domain contains two zinc finger motifs. Transcription enhancer factor-1 (TEF-1), retinoid X receptor alpha, DNA polymerase alpha, X-ray repair cross-complementing factor-1 (XRCC1), and PARP-1 itself interact with PARP-1 in this domain. The self-modifying domain includes the BRCT motif, which is a protein-protein interaction module. This motif was originally found at the C-terminus of BRCA1 (breast cancer sensitive protein 1) and is present in a variety of proteins involved in DNA repair, recombination, and cell cycle checkpoint control. Octameric transcription factor-1 (Oct-1), Yin Yang protein (YY)1 and ubiquitin conjugating enzyme 9(ubc9) containing the POU-homeodomain can interact with the BRCT motif in PARP-1.
More than 15 members of the PARP family of genes are present in the genome of mammals. PARP family proteins and poly (ADP-ribose) glycohydrolase (PARG) which degrades poly (ADP-ribose) to ADP-ribose may be involved in a variety of cellular regulatory functions, including DNA damage response and transcriptional regulation, and may be involved in carcinogenesis and many aspects of cancer biology.
Several PARP family proteins have been identified. Tankyrase has been found to be an interacting protein of telomere regulatory factor 1(TRF-1) and is involved in telomere regulation. Vault parp (vparp) is a component of the vault complex, which functions as the nuclear cytoplasmic transporter. PARP-2, PARP-3 and 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin inducible PARP (tiparp) have also been identified. Thus, poly (ADP-ribose) metabolism may be involved in a variety of cellular regulatory functions.
The most studied member of this gene family is PARP 1. The PARP1 gene product is expressed at high levels in the nucleus of cells, and its activation is dependent on DNA damage. Without being bound by any theory, it is believed that PARP1 binds to DNA single or double strand breaks through the amino-terminal DNA binding domain. The binding activates the catalytic domain at the carbonyl terminus and results in the formation of a polymer of ADP-ribose on the target molecule. Due to the centrally located self-modifying domain, PARP1 is itself a target for poly ADP-ribosylation. Nuclear glycosylation of PARP1 results in the dissociation of the PARP1 molecule from the DNA. The overall process of binding, ribosylation and isolation occurs very rapidly. It has been proposed that transient binding of PARP1 to a DNA damage site results in restoration of the DNA repair mechanism (recovery), or may act to inhibit recombination, and is long enough to restore the repair mechanism.
Bauer et al (int.J.Oncol.8, 239, 1996) demonstrated that poly ADP-ribosylation in cancer cells inhibits Ca2+-Mg2+Dependent dnase, and therefore can replicate cancer uncontrollably. The deregulation of dnase (by PARP-1 inhibition) can trigger cancer cell-specific DNA fragmentation and induce only cancer cell apoptosis. Physiologically present dsDNA is an excellent coenzyme for damaged DNA for PARP-1, thereby making PARP-1 function as a physiologically effective chromatin modulator in intact cells (Kun et al, j.biol.chem.277, 39066, 2002), which plays a distinct role in the cancer phenotype.
In human PARP, the N-terminal DBD in PARP-1 extends from the initiating methionine to threonine-373. This domain has a molecular weight of approximately 42kDa and contains two zinc fingers and two helix-turn-helix motifs. The DBD of PARP also contains a high proportion of basic residues that may be involved in the interaction of enzymes and DNA. PARP is a metalloenzyme that specifically binds to zinc molecules. The zinc binding site binds to a 29kDa PARP fragment produced by limited proteolysis of a protein with trypsin. Binding of PARP to zinc indicates that the enzyme has a zinc finger, which is subsequently confirmed by sequence analysis of the cloned cDNA. Zinc finger 1(F1) starts at cysteine-21 and ends at cysteine-56, while zinc finger 2(F2) is found between cysteine-125 and cysteine-162. See D' amuors et al, op.cit. (1999).
Without intending to be limited to one mechanism of action, one aspect of the invention relates to the bimodal effect of ATP on the poly ADP-ribose cycle, i.e., its degradation site and a specific poly ADP-ribose synthesis site. This involves the action of ATP on isolated nuclei, involves inhibition of poly ADP-ribosylation, and the action of ATP on specific glycohydrolase enzymes that regulate the degradation of protein-bound poly ADP-ribosyl chains. Isolated nuclei also respond to ATP-inhibiting poly (ADP-ribose) polymerase and ATP-activating poly (ADP-ribose) glycohydrolase, demonstrating that enzymatic results can be extrapolated to cellular systems.
Physiological concentrations of ATP (or its non-hydrolyzable analogs) have been demonstrated in the Zn of PARP-12+Inhibition of PARP-1 on finger 1 (Kun et al, Biochemistry 43, 210, 2004); this is not at the NAD catalytic site. By applying two Zn layers2+Amino acid mutations of the arginine residue in the finger further identified this inhibition site to pinpoint the ATP site (Bauer et al, int.j.mol.med.2005, accepted, now in print) which was found to be Zn2+Refers to arginine-34 in 1. Because arginine residues (Zacharias et al Trends in Pharmacol. Sci.23, 281, 2002) are able to react with "aggressive" phosphates (e.g. ATP) and fragrances (electron donors) (Woods, J.Proteomic Res.3, 478, 2004), the ATP "site" can aid in the identification of fragrances by virtue of the presence of Zn in the fragrance2+Refers to the site of reactive inhibition of PARP-1 by arginine-34 in 1. Thus, PARP inhibitors of the invention may be identified by their interaction with arginine-34 and may be identified kinetically by the additivity of ATP inhibition. Due to the high PARP-1 activity of cancer, which is a characteristic biochemical phenotype of cancer, the arginine-34 selective PARP-1 inhibitors of the present invention may act directly on tumor cells.
PARP mediated diseases
One aspect of the present invention is a method of treating a PARP mediated disease comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound having 4 to about 35 carbon atoms, including formula I above, preferred embodiments thereof, formula II and/or preferred embodiments thereof, wherein said organic aromatic compound is capable of binding to an arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein said organic aromatic compound has an electron donating capability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of a particular arginine-34 residue of the zinc finger-1 of PARP-1, wherein when said aromatic compound comprises a heterocyclic ring containing a nitrogen atom, said ring does not contain a carbonyl moiety.
Various PARP mediated diseases include, but are not limited to: cancers, including adrenocortical carcinoma, rectal cancer, aplastic anemia, biliary tract cancer, bladder cancer, bone metastasis, adult CNS brain tumor, pediatric CNS brain tumor, breast cancer, Castleman's disease, cervical cancer, pediatric non-Hodgkin's lymphoma, colorectal cancer, endometrial cancer, esophageal cancer, Ewing's tumor family, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, Hodgkin's disease, Kaposi's sarcoma, renal cancer, laryngeal carcinoma, acute lymphocytic leukemia, acute myelogenous leukemia, childhood leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, liver cancer, lung carcinoid tumor, non-Hodgkin's lymphoma, male breast cancer, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, cancer of the nasal cavity and sinuses, nasopharyngeal cancer, neuroblastoma, malignant melanoma, malignant lymphoma, cervical cancer, bone metastasis, colorectal cancer, Oral and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumor, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma (adult soft tissue cancer), melanoma skin cancer, non-melanoma skin cancer, gastric cancer, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, vaginal cancer, vulval cancer, and waler's isostan macroglobulinemia.
PARP mediated diseases include angiogenesis, inflammation, degenerative diseases, CNS diseases, autoimmune diseases and viral diseases (including HIV) in cancer. The compounds described herein are also useful for modulating cellular responses to pathogens. The present invention also provides methods of treating other PARP mediated diseases, such as viral diseases. Some viral diseases include, but are not limited to, Human Immunodeficiency Virus (HIV), herpes simplex virus types 1 and 2, and Cytomegalovirus (CMV), a dangerous HIV co-infection.
Other PARP mediated diseases include, but are not limited to: inflammatory bowel disease, arthritis, hyperglycemia, diabetes, endotoxic or septic shock, peripheral nerve injury, skin aging, epilepsy, stroke, parkinson's disease, amyotrophic lateral sclerosis, huntington's disease, schizophrenia, chronic pain, ischemia, post-hypoxic neuronal loss, alzheimer's disease, atherosclerosis, osteoarthritis, osteoporosis, muscular dystrophy, degenerative diseases of skeletal muscle involving replicative senescence of cells, age-related macular degeneration, immunosenescence, and other immunosenescence disorders. In some embodiments, the compounds and methods described herein are used to modulate, preferably inhibit, angiogenesis or inflammation.
Some examples of PARP mediated diseases are described herein, but are not limiting to the scope of the invention, and may be other PARP mediated diseases known in the art and are within the scope of the invention.
Examples of cancer
Examples of cancer include, but are not limited to: lymphomas, carcinomas, and hormone-dependent tumors (e.g., breast, prostate, or ovarian cancer). Abnormal cell proliferation disorders or cancers in adults or children that may be treated include solid/malignant tumors, locally advanced tumors, human soft tissue sarcomas, metastatic cancers (including lymphatic metastases), hematological malignancies (including multiple myeloma, acute and chronic leukemias, and lymphomas), head and neck cancers (including oral, laryngeal, and thyroid cancers), lung cancers (including small cell and non-small cell cancers), breast cancers (including small cell and ductal cancers), gastrointestinal cancers (including esophageal, gastric, colon, colorectal, and polyps associated with colorectal neoplasias), pancreatic, liver, urinary tract cancers (including bladder and prostate cancers), female reproductive tract malignancies (including ovarian, uterine (including endometrial) and solid tumors in the ovarian follicles), kidney cancers (including renal cell cancers), brain cancer (including endogenous brain tumors, neuroblastoma, astrocytic brain tumors, glioma, metastatic tumor cell invasion in the central nervous system), bone cancer (including osteoma), skin cancer (including malignant melanoma, human skin keratinocyte tumor progression, squamous cell carcinoma, basal cell carcinoma, hemangiopericyte tumor, and kaposi's sarcoma).
In some preferred embodiments of the invention, the cancer comprises colon adenocarcinoma, esophageal adenocarcinoma, liver hepatocellular carcinoma, squamous cell carcinoma, pancreatic adenocarcinoma, islet cell tumor, rectal adenocarcinoma, gastrointestinal stromal tumor, gastric adenocarcinoma, adrenal cortical carcinoma, follicular carcinoma, papillary carcinoma, breast cancer, ductal carcinoma, lobular carcinoma, intraductal carcinoma, mucinous carcinoma, phyllodes tumor, ovarian adenocarcinoma, endometrial adenocarcinoma, granular cell tumor, mucinous cystadenocarcinoma, cervical adenocarcinoma, vulva squamous cell carcinoma, basal cell carcinoma, prostate adenocarcinoma, giant cell tumor of bone, osteosarcoma, laryngeal carcinoma, lung adenocarcinoma, kidney carcinoma, urinary bladder carcinoma, and Wilm tumor.
In a more preferred embodiment of the invention, the cancer comprises mixed endometrial and mixed ductal and lobular infiltrates, Wilm's tumor, mixed ovarian muller's tumor, serous cystadenocarcinoma, ovarian adenocarcinoma (papillary serous), ovarian adenocarcinoma (endometrioid), metastatic infiltrative lobular breast cancer, testicular seminoma, benign nodular hyperplasia of the prostate, squamous lung carcinoma, large lung cell carcinoma, lung adenocarcinoma, endometrial adenocarcinoma (endometrioid), infiltrative ductal carcinoma, basal skin carcinoma, infiltrative lobular breast cancer, fibrocystic disease, fibroids, glioma (gleoma), chronic myelogenous leukemia, hepatocellular carcinoma of the liver, mucinous carcinoma, Schwannoma, transitional cell carcinoma of the kidney, hashimoto's thyroiditis, metastatic infiltrative ductal breast carcinoma, esophageal adenocarcinoma, thymoma, phyllodes tumor, melanoma, colon tumor, colon, Rectal adenocarcinoma, osteosarcoma, colon adenocarcinoma, papillary thyroid carcinoma, leiomyoma, and gastric adenocarcinoma.
Examples of inflammation
Examples of inflammation include, but are not limited to: systemic inflammation and disorders locally associated with the migration and attraction of monocytes, leukocytes and/or neutrophils. Inflammation may result from infection with pathogenic organisms (including gram positive bacteria, gram negative bacteria, viruses, fungi, and parasites, such as protozoa and helminths), transplant rejection (including rejection of solid organs such as kidney, liver, heart, lung, or cornea, and bone marrow transplantation, including Graft Versus Host Disease (GVHD)), or local chronic or acute autoimmunity or allergies. Autoimmune diseases include acute glomerulonephritis; rheumatoid or reactive arthritis; chronic glomerulonephritis; inflammatory bowel diseases, such as crohn's disease, ulcerative colitis, and necrotizing enterocolitis; granulocyte transfusion-related syndrome; inflammatory dermatoses such as contact dermatitis, atopic dermatitis, psoriasis; systemic Lupus Erythematosus (SLE), autoimmune thyroiditis, multiple sclerosis, and some forms of diabetes, or any other autoimmune situation in which attack of the subject's own immune system results in pathological tissue destruction. Allergies include allergic asthma, chronic bronchitis, acute and delayed hypersensitivity reactions. Systemic inflammatory disease conditions include inflammation associated with trauma, burns, reperfusion following ischemic events (e.g., thrombotic events in the heart, brain, bowel or peripheral vasculature including myocardial infarction and stroke), sepsis, ARDS or multiple organ dysfunction syndrome. Inflammatory cell recruitment also occurs in atherosclerotic plaques.
In some preferred embodiments, the inflammation comprises non-hodgkin's lymphoma, wegener's granulomatosis, hashimoto's thyroiditis, hepatocellular carcinoma, thymus atrophy, chronic pancreatitis, rheumatoid arthritis, reactive lymphoid tissue hyperplasia, osteoarthritis, ulcerative colitis, papillary carcinoma, crohn's disease, ulcerative colitis, acute cholecystitis, chronic cholecystitis, cirrhosis, chronic sialadenitis, peritonitis, acute pancreatitis, chronic gastritis, adenomyosis (adenomysis), endometriosis, acute cervicitis, chronic cervicitis, lymphoid tissue hyperplasia, multiple sclerosis, hypertrophy secondary to idiopathic thrombocytopenic purpura, primary IgA nephropathy, systemic lupus erythematosus, psoriasis, emphysema, chronic pyelonephritis, and chronic cystitis.
Examples of endocrine and neuroendocrine lesions
Examples of endocrine lesions include lesions of the adrenal gland, breast, gonads, pancreas, parathyroid gland, pituitary gland, dwarfism (dwarfism), and the like. Adrenal disorders include, but are not limited to, addison's disease, hirsutism (hirsutism), cancer, multiple endocrine tumors, congenital adrenal hyperplasia, and pheochromocytoma. Breast lesions include, but are not limited to, breast cancer, fibrocystic breast disease, and gynecomastia. Gonadal disorders include, but are not limited to, congenital adrenal hyperplasia, polycystic ovary syndrome, and turner's syndrome. Pancreatic pathologies include, but are not limited to, diabetes (type I and type II), hypoglycemia, and insulin resistance. Parathyroid disorders include, but are not limited to, hyperparathyroidism and hypoparathyroidism. Pituitary pathologies include, but are not limited to, acromegaly, cushing's syndrome, diabetes insipidus, empty sphenoid saddle syndrome, hypopituitarism and prolactinoma. Thyroid disorders include, but are not limited to, cancer, goiter, hyperthyroidism, hypothyroidism, nodules, thyroiditis, and Wilson's syndrome. Examples of neuroendocrine disorders include, but are not limited to, depression and anxiety disorders associated with hormonal imbalances, menstrual epilepsy, menopause, menstrual migraine, regenerative endocrine disorders, gastrointestinal disorders (such as visceral endocrine tumors, including carcinoid tumors, gastrinomas, and somatostatin tumors), achalasia, and Hirschsprung's disease. In some embodiments, the endocrine and neuroendocrine lesions include nodular hyperplasia, hashimoto's thyroiditis, islet cell tumors, and papillary carcinoma.
Endocrine and neuroendocrine disorders in children include growth-disordered endocrine disorders and diabetes insipidus. Growth retardation can be observed by congenital ectopy or congenital atrophy/hypoplasia of the pituitary gland, such as in anaplasia of the forebrain, hypoplasia of the septum, and cranial base bulge (basal encephalocel). Acquired disorders such as craniopharyngioma, optic/hypothalamic glioma may be present with clinical short stature (short status) and diencephalon syndrome. Precocious puberty and overgrowth can be seen in the following conditions: arachnoid cyst, hydrocephalus, hypothalamic hamartoma, and embryonal histiocytoma. Hypersecretion of growth hormone and corticotropin from pituitary adenomas can lead to pathologic high stature and trunk obesity in children. Diabetes insipidus can be secondary to an invasive process such as Langerhans cells for histiocytosis, tuberculosis, blastomas, post-traumatic/surgical injury of the pituitary stalk and hypoxic ischemic encephalopathy.
Examples of nutritional and metabolic pathologies
Examples of nutritional and metabolic pathologies include, but are not limited to: aspartylglucamine urine disease, biotinidase deficiency, carbohydrate-deficient glycoprotein syndrome (CDGS), Crigler-Najal syndrome, cystinosis, diabetes insipidus, Fabry's disease, fatty acid metabolism disorders, galactosemia, gaucher's disease, glucose-6-phosphate dehydrogenase (G6PD), glutaruria, Huller's disease, Huller-Sha's disease, Hunter's disease, hypophosphatemia, I-cell, Krabe's disease, lactic acidosis, Long-chain 3-hydroxy acyl CoA dehydrogenase deficiency (LCHAD), lysosomal storage disorders, mannosidosis, maple syrup urine, Maloto-Lamei disease, metachromatic leukodystrophy, mitochondria, Moryol disease, mucopolysaccharidoses, neuro-metabolism disorders, Niemann-pick's disease, acidosis, purines, Phenylketonuria (PKU), pompe, pseudo-Huller's disease, pyruvate dehydrogenase deficiency, sandhoff, san Fragile's disease, Share's disease, Spisis, Thai-Sacha, trimethylaminouria (Fish odor syndrome), Urea cycle disorders, vitamin D deficiency rickets, muscle metabolism disorders, inherited metabolic disorders, acid-base imbalance, acidosis, alkalosis, homogentisate uremia, alpha-mannosidosis, amyloidosis, anemia, iron deficiency, ascorbic acid deficiency, vitamin deficiency, beriberi, biotinases deficiency, glycoprotein deficiency syndrome, carnitine disorders, cystinosic, cystinuria, Fabry's disease, fatty acid oxidation disorders, fucosidosis, galactosemia, gaucher's disease, Gilbefibrate, glucose phosphate dehydrogenase deficiency, glutaremia, glycogen storage disease, Harter's sodium disease, hemochromatosis, hemosiderosis, hepatolenticular degeneration, histidinemia, homocystinuria, hyperbilirubinemia, hypercalcemia, hyperinsulinemia, hyperkalemia, hyperlipidemia, hyperoxaluria, vitamin a hyperactivity, hypocalcemia, hypoglycemia, hypokalemia, hyponatremia, hypophosphatasia, insulin resistance, iodine deficiency, iron overload, jaundice, chronic idiopathic xanthomatosis, leiomyelitis, lesch-nier syndrome, leucine metabolism disorders, lysosomal storage diseases, magnesium deficiency, maple syrup urine disease, MELAS syndrome, mengkins curl syndrome, metabolism syndrome X, mucolipidosis, mucopolysaccharidosis (mucopolysaccharosis), niemann-pick disease, obesity, ornithine aminoacyl-methyltransferase deficiency, osteomalacia, pellagra, peroxisome porphyria, erythropoiesis, porphyria (porphyies), premature aging, pseudo-gaucher disease, refsum disease, reyh syndrome, rickets, sandhoff disease, dangill disease, tay-saxas, tetrahydrobiopterin deficiency, trimethylaminouria (fish odor syndrome), tyrosinemia, urea cycle disorders, water-electrolyte imbalance, weirnike encephalopathy, vitamin a deficiency, vitamin B12 deficiency, vitamin B deficiency, wolmann disease, and zerwerg syndrome.
In some preferred embodiments, the metabolic disease includes diabetes and obesity.
Examples of diseases of the haemolymph system
Hematologic lymphatic diseases include hematologic and lymphatic diseases. "hematological disorders" include diseases, disorders or conditions that affect hematopoietic cells or tissues. Hematological disorders include diseases, disorders or conditions associated with abnormal hematological content or function. Examples of hematological lesions include lesions resulting from bone marrow irradiation or chemotherapy treatment of the following cancers, lesions: such as malignant anemia, hemorrhagic anemia, hemolytic anemia, aplastic anemia, sickle cell anemia, sideroblasts anemia, anemia associated with chronic infections such as malaria, trypanosomiasis, HIV, hepatitis virus or other viruses, myelopathic anemia resulting from bone marrow deficiency, renal failure resulting from anemia, polycythemia, Infectious Mononucleosis (IM), acute nonlymphocytic leukemia (ANLL), Acute Myelogenous Leukemia (AML), Acute Promyelocytic Leukemia (APL), acute myelomonocytic leukemia (AMMoL), polycythemia vera, lymphoma, Acute Lymphocytic Leukemia (ALL), chronic lymphocytic leukemia, Wilm's tumor, Ewing's sarcoma, retinoblastoma, hemophilia, pathologies associated with increased risk of thrombosis, Herpes, thalassemia, antibody-mediated pathologies such as transfusion reactions and erythroblastosis, mechanical damage to red blood cells such as micro-angiohemolytic anemia, thrombotic thrombocytopenic purpura and disseminated intravascular coagulation, infections caused by parasites such as plasmodium, chemical damage such as lead poisoning, and splenic hyperactivity.
Lymphoid disorders include, but are not limited to: lymphadenitis, lymphangitis, lymphedema, cystic lymphangioma, lymphoproliferative disorders, mucocutaneous lymph node syndrome, reticuloendotheliosis, spleen disease, thymic hyperplasia, thymic tumor, tuberculosis, lymphadenopathy, pseudolymphoma, and lymphoid abnormalities.
In some preferred embodiments, the lesions of the blood lymphatic system include non-hodgkin's lymphoma, chronic lymphocytic leukemia, and reactive lymphoid hyperplasia.
Examples of CNS disorders
Examples of CNS diseases include, but are not limited to: neurodegenerative diseases, drug abuse such as cocaine abuse, multiple sclerosis, schizophrenia, acute disseminated encephalomyelitis, transverse myelitis, demyelinating genetic diseases, spinal cord injury, virus-induced demyelination, progressive multifocal leukoencephalopathy, human lymphotrophic T-cell virus i (htlvi) -associated myelopathy, and nutritional metabolic pathologies.
In some preferred embodiments, the CNS disorders comprise parkinson's disease, alzheimer's disease, cocaine abuse, and schizophrenia.
Examples of neurodegenerative diseases
Neurodegenerative diseases in the methods of the invention include, but are not limited to: alzheimer's disease, pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Stel-Richcson syndrome), multiple system degeneration (Chari-Delerger syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxia, corticobasal degeneration, Guam ALS-Parkinson-dementia complex (ALS-Parkinson's-dementialia complex of guam), subacute panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatal substantia nigra degeneration, Marsdo-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degeneration, Gilledlla Tourette disease, bulbar palsy and pseudobulbar palsy, spinal and bulbar muscular atrophy (Kennedy's disease), primary lateral sclerosis, familial spastic paraparesis, Wer-Hover disease, Ku-Wer disease, Thai-Sacha disease, sandhoff disease, familial spasticity, Ware-Ku-Wer disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt-Jakob disease, Gerstmann-Straussler-Scheinker disease, Kuru disease, and fatal familial insomnia), Alexander disease, Aleper disease, amyotrophic lateral sclerosis, ataxia telangiectasia, Barbat disease, Kaempanan disease, Kjen's syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, Huntington disease, Kennedy disease, Krabbe disease, Lewy-body dementia, Masando-Joseph disease, spinocerebellar ataxia type 3, multiple sclerosis, multiple system atrophy, Parkinson's disease, familial intermediate-leaf sclerosis, refsum disease, schild disease, staffs-sertoli-barbyten disease, sertoli-richardson-olschuski disease, and tabes dorsalis.
Examples of urinary tract disorders
Urinary tract disorders in the methods of the invention include, but are not limited to: lesions of the kidney, ureter, bladder, and urethra. For example, urethritis, cystitis, pyelonephritis, renal hypoplasia, hydronephrosis, polycystic kidney disease, polycystic kidney, lower urinary tract obstruction, bladder eversion and cleavage of the urethra, bacterial urine, prostatitis, intrarenal and peripheral abscesses, benign prostatic hypertrophy, renal cell carcinoma, transitional cell carcinoma, Wilm's tumor, uremia, and glomerulonephritis.
Examples of respiratory diseases
Respiratory diseases and conditions include, but are not limited to: asthma, Chronic Obstructive Pulmonary Disease (COPD), adenocarcinoma, adenosquamous carcinoma, squamous cell carcinoma, large cell carcinoma, Cystic Fibrosis (CF), dyspnea, emphysema, wheezing, pulmonary hypertension, pulmonary fibrosis, hyper-responsive airways, increased adenosine or adenosine receptor levels, pulmonary bronchoconstriction, pneumonia and allergy, and surfactant depletion, chronic bronchitis, bronchoconstriction, dyspnea, obstructive and obstructive airways, adenosine testing of cardiac function, pulmonary vasoconstriction, obstructive breathing, Acute Respiratory Distress Syndrome (ARDS), administration of certain drugs, for example adenosine and drugs which elevate adenosine levels, and other drugs, for example for the treatment of supraventricular tachycardia (SVT), and also for the performance of adenosine stress tests, infant Respiratory Distress Syndrome (RDS), pain, allergic rhinitis, reduction of pulmonary surfactants, reduced levels of coenzyme Q, or chronic bronchitis, among others.
Examples of female reproductive system lesions
Pathologies of the female reproductive system include diseases of the vulva, vagina, cervix, uterus, fallopian tubes and ovaries. Some examples include, adnexal diseases. For example, fallopian tube disease, ovarian disease, leiomyoma, mucinous cystadenocarcinoma, serous cystadenocarcinoma, ovarian crown cyst, and pelvic inflammatory disease; endometriosis; genital tumors, e.g., fallopian tube, uterine, vaginal, vulvar, and ovarian tumors; vaginal atresia; genital herpes; infertility; sexual dysfunction, e.g., dyspareunia, and impotence; tuberculosis; uterine diseases, such as cervical diseases, endometrial hyperplasia, endometritis, hematuria, uterine bleeding, uterine tumors, uterine prolapse, uterine rupture, and uterine varus; vaginal diseases, such as dyspareunia, colporrhagia, vaginal fistulas, vaginal tumors, vaginitis, vaginal discharge, and candidiasis or vulvovaginal disease; vulvar diseases, e.g., vulvar trunk, pruritus, vulvar tumors, vulvitis, and candidiasis; and genitourinary disorders such as genitourinary abnormalities and genitourinary tumors.
Examples of disorders of the male reproductive system
Examples of lesions of the male reproductive system include, but are not limited to: epididymitis; genital tumors, e.g., penile tumors, prostate tumors, and testicular tumors; hematocele; genital herpes; scrotal cyst; infertility; penile diseases, such as balanitis, hypospadias, pelonemia, penile tumors, phimosis, and priapism; prostate diseases, such as, for example, prostatic hyperplasia, prostate tumors, and prostatitis; sexual dysfunction of the apparatus, e.g., dyspareunia, and impotence; twisting a spermatic cord; a cyst of sperm; testicular diseases, such as cryptorchidism, orchitis, and testicular tumors; tuberculosis; varicocele; urogenital diseases, e.g., urogenital abnormalities and urogenital tumors; and funier gangrene.
Examples of cardiovascular pathologies (CVS)
Cardiovascular pathologies include those that result in ischemia or those that result from cardiac reperfusion. Examples include, but are not limited to: atherosclerosis, coronary artery disease, granulomatous myocarditis, chronic myocarditis (non-granuloma), primary hypertrophic cardiomyopathy, Peripheral Artery Disease (PAD), stroke, angina pectoris, myocardial infarction, cardiovascular tissue damage due to cardiac arrest, cardiovascular tissue damage due to cardiac bypass, cardiogenic shock, and related conditions known to those skilled in the art or involving dysfunction of the heart or vasculature or tissue damage (particularly, but not limited to, tissue damage associated with PARP activation). In some preferred embodiments of the invention, CVS diseases include atherosclerosis, granulomatous myocarditis, myocardial infarction, myocardial fibrosis secondary to valvular heart disease, myocardial fibrosis without infarction, primary hypertrophic cardiomyopathy, and chronic myocarditis (non-granulomas).
Method of treatment
The methods provided herein may comprise administering a compound of formula I, II and/or preferred embodiments thereof. The compounds may also be administered in combination with other therapies. The choice of therapy that can be administered with the compositions of the invention will depend in part on the condition being treated. For example, for the treatment of acute myeloid leukemia, the compounds of some embodiments of the present invention may be used in combination with radiation therapy, monoclonal antibody therapy, chemotherapy, bone marrow transplantation, or a combination thereof.
A therapeutically effective amount of a PARP inhibitor is administered to a patient, preferably a mammal, more preferably a human, to affect the pharmacological activity associated with the inhibition of the PARP enzyme. Thus, the PARP inhibitors of the present invention may be used to treat or prevent a variety of diseases and disorders, including nerve tissue damage, cerebral ischemia and reperfusion injury or neurodegenerative diseases caused by cell damage or death due to necrosis or apoptosis in animals. In addition, the compounds of the present invention may be used to treat cardiovascular disease in an animal by administering to the animal an effective amount of a PARP inhibitor. Still further, the compounds of the present invention can be used for the treatment of cancer or for radiosensitizing (radiosensitize) or chemosensitizing (chemoradiosensitize) tumor cells.
In some embodiments of the invention, PARP inhibitors may be used to stimulate damaged neurons, promote neuronal regeneration, prevent neurodegeneration, and/or treat neurological disorders. PARP inhibitors inhibit PARP activity and are therefore useful for treating neural tissue damage in animals, particularly damage caused by cancer, cardiovascular disease, cerebral ischemia and reperfusion injury, or neurodegenerative disease. The PARP inhibitors of the present invention may be used to treat cardiac tissue damage in a patient, particularly damage caused by cardiac ischemia or resulting from reperfusion injury. The compounds of the invention are particularly useful for treating cardiovascular diseases selected from the group consisting of: coronary artery disease, such as atherosclerosis; angina pectoris; myocardial infarction; myocardial ischemia and cardiac arrest; cardiac bypass; and cardiogenic shock.
In another aspect, the PARP inhibitors of the invention may be used for the treatment of cancer, and for radiosensitizing and/or chemosensitizing tumor cells. The PARP inhibitors of the invention may be "anti-cancer agents", which term also includes "anti-tumor cell growth agents" and "anti-tumor agents". For example, PARP inhibitors of the invention may be used to treat cancer, and radiosensitize and/or chemosensitize tumor cells in cancer.
Radiosensitizers are known to increase the sensitivity of cancer cells to the toxic effects of electromagnetic radiation. Many cancer treatment protocols now use radiosensitizers activated by electromagnetic radiation of x-rays. Examples of x-ray activated radiosensitizers include, but are not limited to: metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU1069, SR4233, EO9, RB6145, niacinamide, 5-bromodeoxyuridine (BUdR), 5-iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives thereof.
Photodynamic therapy (PDT) of cancer uses visible light as a radiation activator of a sensitizer. Examples of photodynamic radiosensitizers include, but are not limited to: hematoporphyrin derivatives, photoporphyrins, benzoporphyrin derivatives, NPe6, stannorphyrin SnET2, pheoborbide- α, bacteriochlorophyll- α, naphthalocyanine, phthalocyanine, zinc phthalocyanine, and therapeutically effective analogs and derivatives thereof.
Radiosensitizers can be administered in combination with a therapeutically effective amount of one or more PARP inhibitors, including but not limited to: PARP inhibitors that promote the incorporation of radiosensitizers into target cells; PARP inhibitors that control the flow of therapeutic agents, nutrients and/or oxygen to a target. Similarly, chemosensitizers are also known to increase the sensitivity of cancer cells to the toxic effects of chemotherapeutic compounds. Exemplary chemotherapeutic agents that may be used in combination with PARP inhibitors include, but are not limited to, doxorubicin, camptothecin, dacarbazine, carboplatin, cisplatin, daunorubicin, docetaxel, adriamycin, interferons (α, β, γ), interleukin 2, irinotecan, paclitaxel, streptozotocin, temozolomide, topotecan, and therapeutically effective analogs and derivatives thereof. In addition, other therapeutic agents that may be used in combination with the PARP inhibitor include, but are not limited to, 5-fluorouracil, leucovorin, 5 '-amino-5' -deoxythymidine, oxygen, carbon potential (carbogen), red blood cell infusion, perfluorocarbons (e.g., Fluosol-DA), 2, 3-DPG, BW12C, calcium channel blockers, pentoxifylline, anti-angiogenic compounds, hydralazine, and L-BSO.
Formulation, route of administration and effective dose
Another aspect of the invention relates to the formulation and route of administration of pharmaceutical compositions comprising compounds of formula I, preferred embodiments thereof, II and/or IIa. Such pharmaceutical compositions may be used to treat cancer in the methods described in detail above.
The compounds of formula I, its preferred embodiments, II and/or IIa may be provided as prodrugs and/or may be converted into its form in vivo after administration. That is, the compounds or their pharmaceutically acceptable salts can be used to develop the formulations used in the present invention. Furthermore, in some embodiments, the compounds may be used in combination with or in one or more other forms. The two forms may be formulated together in the same dosage unit, for example, an emulsion, suppository, tablet, capsule or powder packet to be dissolved in a beverage; or each form may be formulated in separate units, e.g., two emulsions, two suppositories, two tablets, two capsules, one tablet and one liquid for dissolving the tablet, one powder packet and one liquid for dissolving the powder, etc.
Other active agents may be effective in compositions comprising a combination of compounds of formula I, preferred embodiments thereof, II and/or IIa. The two compounds and/or the two forms of the compounds may be formulated in the same dosage unit, for example as an emulsion, suppository, tablet, capsule or sachet to be dissolved in a beverage; or each form may be formulated in separate units, e.g., two emulsions, a suppository, a tablet, two capsules, a tablet and a liquid for dissolving the tablet, a powder packet and a liquid for dissolving the powder, and the like.
Typical salts are inorganic ionic salts such as sodium, potassium, calcium and magnesium ions. Such salts include those formed with inorganic or organic acids such as hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, methanesulfonic, p-toluenesulfonic, acetic, fumaric, succinic, lactic, mandelic, malic, citric, tartaric, or maleic acid. Furthermore, if a compound used in the present invention contains a carboxyl group or other acidic group, it can be converted into a pharmaceutically acceptable addition salt with an inorganic or organic base. Examples of suitable bases include sodium hydroxide, potassium hydroxide, aqueous ammonia, cyclohexylamine, dicyclohexylamine, ethanolamine, diethanolamine, and triethanolamine.
For oral administration, the compounds can be readily formulated by combining the active compound with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets (including chewable tablets), pills, dragees, capsules, lozenges, hard candies, liquids, gels, syrups, slurries, powders, suspensions, elixirs, and wafers, and the like, for oral ingestion by a patient to be treated. Such formulations may contain a pharmaceutically acceptable carrier, which includes solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents. In general, the compounds of the present invention are included at a concentration level of about 0.5%, about 5%, about 10%, about 20%, or about 30% to about 50%, about 60%, about 70%, about 80%, or about 90% by weight of the total composition of an oral dosage form in an amount sufficient to provide the desired dosage unit.
Aqueous suspensions may contain a compound of formula I, embodiments thereof, II and/or IIa, and pharmaceutically acceptable excipients, such as suspending agents (e.g. methylcellulose), wetting agents (e.g. lecithin, lysolecithin and/or long chain fatty alcohols), as well as coloring, preserving and flavoring agents and the like.
In some embodiments, an oil or non-aqueous solvent may be required to bring the compound into solution, for example due to the presence of a large lipophilic moiety. Alternatively, an emulsion, suspension or other preparation, such as a liposome preparation, may be used. With regard to the liposomal preparation, any known method of preparing liposomes for treating disorders can be used. See, for example, Bangham et al, j.mol.biol, 23: 238-: 4194-4198(1978), incorporated herein by reference. Ligands may also be attached to the liposomes to direct these compositions to specific sites of action. The compounds of the invention may also be incorporated into food products such as cream cheese, butter, salad dressings or ice cream to facilitate solubilization, administration and/or compliance in certain patient populations.
Pharmaceutical preparations for oral use can also be obtained as solid excipients, and after adding suitable auxiliaries, if desired, the resulting mixtures are optionally ground and the mixture of particles is processed to give tablets or dragee cores. In particular, suitable excipients are fillers, for example, sugars, including lactose, sucrose, mannitol, or sorbitol; flavouring ingredients, for example cellulose preparations, such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof (e.g., sodium alginate). The compounds may also be formulated as sustained release agents.
Dragee cores can be provided with suitable films. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyes or pigments may be added to the tablets or dragee films to distinguish or indicate different combinations of active compound doses.
Pharmaceutical preparations which can be taken 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. Push-fit capsules can contain the active ingredient in admixture with fillers, such as lactose, binders (e.g., starch), and/or lubricants, such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, for example fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for administration.
For injection, the inhibitors of the invention may be formulated as aqueous solutions, preferably physiologically compatible buffers, such as Hank's solution, Ringer's solution or physiological saline buffer. Such compositions may also comprise one or more excipients, such as preservatives, solubilizers, fillers, lubricants, stabilizers, albumin, and the like. Formulation methods are known in the art, for example, as disclosed in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing co. The compounds may also be formulated for transmucosal, buccal, by inhalation, parenteral, transdermal, and rectal administration.
In addition to the formulations described above, the compounds may also be formulated as depot formulations. Such long acting formulations may be administered by implantation or transdermal delivery (e.g. subcutaneous or intramuscular), intramuscular injection or using a transdermal patch. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (e.g. as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, e.g. as a sparingly soluble salt.
Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredient is present in an effective amount, i.e., an amount effective to obtain a therapeutic and/or prophylactic benefit in at least one of the cancers described herein. The actual effective amount for a particular application will depend on the condition being treated, the condition of the subject, the formulation and route of administration, and other factors known to those skilled in the art. Determining an effective amount of a compound of formula I, embodiments thereof, II and/or IIa is within the ability of those skilled in the art, based on the disclosure herein, and can be determined using routine optimization techniques.
The following formulations and examples are provided to illustrate the invention. The following examples are not intended to limit or narrow the scope of the present invention. Furthermore, those skilled in the art will appreciate that many changes and modifications may be made thereto without departing from the spirit and scope of the appended claims, and it is intended that such changes and modifications be within the scope of the present invention.
Examples
As reported earlier, PARP-1 was purified from calf thymus (Molinet et al (1993) EMBOJ.12: 2109-2117). Or recombinant PARP-1 was isolated from Spodoptera frugiperda (Sf9) cells infected with recombinant baculovirus expressing the human PARP-1 gene constructed according to the Pharmingen protocol. The cDNA for the amino acid exchange mutations R34G and R138il of PARP-1 was generated by the large primer method (Kannann et al (1989) Nucl Acids Res 17: 5404). The mutated gene was cloned into the transfer vector pV 1392 and recombinant virus was generated by the Baculogel technique from Pharmin. As reported, the mutated protein was expressed in Sf9 cells, purified and tested (Huang et al (2004) Biochemistry 43: 217-223; Kirsten et al (2004) Method in Molecular Biology 287, epigenetics 137-149).
Poly (ADP-ribose) glycohydrolase (PARG) is purchased from Biomol or Alexis Co, the enzymes of which are equivalent in performance. Jurkat cells were cultured as reported (Buday et al (1996) J Biol Chem 271: 6159-6163) and nuclei were prepared by published methods (Smirnova et al (2000) J Biol Chem 275: 9377-9384). An enzyme assay was performed on PARP-1 as published (DeMurcia (2000) From DNA-damage and Stress Signalling cell Death: Poly ADP-ribosylation Reactions). PARG activity was assayed using poly ADP-ribosylated PARP-1 as a substrate containing a long chain synthesized with spermine as a cofactor (Kun et al (2004) Biochemistry 43: 210-216) or a short chain whose cofactor was histone H1. The poly ADP-ribose is labeled with 3H or by biotinylated-NAD. PARG activity was measured quantitatively either by TLC determination of released 3H-ADP-ribose (Kirsten et al (1991) Exp Cell Res 194: 1-8) (chromatography on PEI-cellulose plates with 0.9M acetic acid and 0.3M LiCl as solvents) or by immunoassay of retained biotinylated (ADP-ribose) (Bakondi et al (2004) Exp Dermatol 13: 170-178). All other reagents were the highest analytical purity.
Example 1
Enzymatic Activity of wild-type arginine-34 and arginine-138 mutant PARP-1
The assay was performed three times (200. mu.M 3H-labeled NAD +, 28 dpm/pmol, 0.5pmol PARP-1, 3mM spermine, pH7.3 t ═ 7.5min) as described (Kun et al (2004) Biochemistry, 43: 210-. The same results were obtained when ATP was replaced by its non-hydrolyzable analogue (Kun et al (2004) Biochemistry, 43: 210-.
Zn in PARP-12+The effect of replacing arginine-34 with glycine in finger 1 is shown in FIG. 1. The total enzyme activity of PARP-1 is not affected by this mutation, whereas the inhibitory effect of ATP (or its non-hydrolysable analogue) is disrupted. These results indicate that only PARP-1 is sensitive to the modulation of ATP. Zn2+The negligible effect of the mutation of arginine-138 to isoleucine on the inhibition of ATP in finger 2 confirms our idea that Zn2+Arginine-34 of finger 1 is the interaction site of ATP with PARP-1.
Example 2
Effect of ATP on PARP-1 Activity of Jurkat nuclei
Equivalent to 2X 105Nuclei of Jurkat cells were preincubated in the presence of various concentrations of ATP. PARP activity was then determined by mixing with biotinylated-NAD (5 μ M final concentration) and incubating for 10 min. After separation of the proteins on an 8% SDS-PAGE gel, nitrocellulose blotted, labeled proteins were detected by incubation with streptavidin-HPO complex (1. mu.g/ml) and by fluorescence photography. Results were expressed in optical density units for three replicates.
The effect of externally added ATP (or non-hydrolyzable analogue thereof) on PARP-1 activity of isolated Jurkat nuclei is shown in FIG. 2. The dramatic inhibition of PARP-1 activity is evident, and inhibition in the nucleus can even be greater than that reported for isolated enzymes, since the Ki of ATP is between 2 and 2.5mM for pure enzyme (3), whereas in the nucleus 1mM ATP already inhibits 80% of PARP-1. This distinction may be due to a higher sensitivity of structure-related PARP-1 in the nucleus or to some loss of diffusible double stranded DNA-s that may occur during nuclear separations (Kun et al (2002) J Biol Chem 277: 39066-.
Example 3
Effect of BCNU on ATP sensitivity to PARR-1 Activity of Jurkat nuclei
The experiment was repeated three times as described in figure 2, except that the preincubation was performed with 400nM BCNU for 30 minutes. The first bar represents PARP activity of BCNU untreated nuclei.
The consequences of BCNU-induced DNA damage on PARP-1 activation and ATP inhibition of this pathophysiologically important process are illustrated in FIG. 3. The response to BCNU-induced DNA damage as determined by PARP-1 activity was completely abolished by externally added ATP, demonstrating that the effect of BCNU is dependent on the bioenergetic capacity of the target cancer cells.
Example 4
Effect of ATP on the Glycohydrolase Activity of Jurkat Nuclear extracts
Poly (ADP-ribosylation-PARP-1 (2. mu.g protein incubated with 50. mu.M biotinylated-NAD, as described above) was adsorbed onto the walls of 96-well plates (Kannann et al (1989) Nucl. acids Res.17: 5404.) Jurkat nuclear extract (50. mu.g protein) was incubated in the presence (. DELTA. -Delta.) or absence (. DELTA. -DEG.) of 10mM ATP for various times as indicated by the abscissa, the amount of PAR remaining adsorbed in the wells was determined in triplicate with the Trevigen assay.
When Jurkat cell extracts were incubated with poly ADP-ribosylated PARP-1 containing long polymers (50 ADP-ribose units), the breakdown of the polymers was evident as a result of PARG activity present in the cell extracts (fig. 4). The addition of 8mM ATP significantly promoted PARG activity.
Example 5
Effect of chain length of PAR polymers on ATP sensitivity of purified PARG
Short chain (. DELTA.. DELTA.) or long chain (-. ANG.. cndot.) PARP molecules are prepared as described in the method section and adsorbed onto the surface of the assay well. Purified PARG (15 mU/assay) was added to wells in the presence of various concentrations of ATP and incubated for 45 minutes. The amount of polymer adsorbed was determined by three replicates (zero minutes value of 0.5 OD for short chain polymers and 1.8 OD for long chain polymers).
The difference in sensitivity to PARG between long and short ADP-ribose oligomers is shown in figure 5, from which it is clear that the degradation of short oligomers is not accelerated by ATP, only the decomposition of longer oligomers (average chain length 50 ADPRs) is accelerated.
Example 6
Effect of ATP on PARG Activity as a function of substrate (PAR) concentration
PARG (15 mU/assay) was incubated with various concentrations of 32P-PAR (long chain polymer) in the presence or absence of 6mM ATP for 45 minutes and the amount of 32P-ADP-ribose released was determined by TLC and liquid scintillation of the cut-off portion of the plate positioned by autoradiography. The experiment was performed in triplicate.
After PARG catalyzed the reaction, the released 3H-ADP-ribose was also measured, as shown in fig. 6, confirming the activation of exonuclease activity of PARG when long chain polymers are substrates.
Example 7
Use of compounds for treating solid tumors colon cancer
Treating a subject afflicted with a solid tumor, colon cancer, with a therapeutically effective amount of a compound of the formula
Wherein the compound is administered orally or parenterally. After several days, the cancer symptoms were significantly reduced.
Example 8
Use of compounds for treating solid tumors colon cancer
The method of example 7 is repeated except that a compound of the formula is administered to a patient suffering from cancer
Similar results were obtained.
Example 9
Use of compounds for treating solid tumors colon cancer
The method of example 7 is repeated except that a compound of the formula is administered to a patient suffering from cancer
Similar results were obtained.
Example 10
Use of compounds for treating solid tumors colon cancer
The method of example 7 is repeated except that a compound of the formula is administered to a patient suffering from cancer
Similar results were obtained.
Example 11
Use of compounds for treating solid tumors colon cancer
The method of example 7 is repeated except that a compound of the formula is administered to a patient suffering from cancer
Similar results were obtained.
Example 12
Use of compounds for treating solid tumors colon cancer
The method of example 7 is repeated except that a compound of the formula is administered to a patient suffering from cancer
Similar results were obtained.
Example 13
Use of compounds in the treatment of inflammation
Treating a subject suffering from inflammation with a therapeutically effective amount of a compound of the formula
Wherein the compound is administered orally or parenterally. After several days, the inflammatory symptoms were significantly reduced.
Example 14
Use of compounds in the treatment of inflammation
The method of example 13 is repeated except that a compound of the formula is administered to a patient suffering from inflammation
Similar results were obtained.
Example 15
Use of compounds in the treatment of inflammation
The method of example 13 is repeated except that a compound of the formula is administered to a patient suffering from inflammation
Similar results were obtained.
Example 16
Use of compounds in the treatment of inflammation
The method of example 13 is repeated except that a compound of the formula is administered to a patient suffering from inflammation
Similar results were obtained.
Example 17
Use of compounds in the treatment of inflammation
The method of example 13 is repeated except that a compound of the formula is administered to a patient suffering from inflammation
Similar results were obtained.
Example 18
Use of compounds in the treatment of inflammation
The method of example 13 is repeated except that a compound of the formula is administered to a patient suffering from inflammation
Similar results were obtained.
Example 19
Use of compounds in the treatment of CNS disorders
Treating a subject suffering from a CNS disease with a therapeutically effective amount of a compound of the formula
Wherein the compound is administered orally or parenterally. After several days, symptoms of CNS diseases are markedly reduced.
Example 20
Use of compounds in the treatment of CNS disorders
The method of example 19 is repeated except that a compound of the formula is administered to a patient suffering from a CNS disease
Similar results were obtained.
Example 21
Use of compounds in the treatment of CNS disorders
The method of example 19 is repeated except that a compound of the formula is administered to a patient suffering from a CNS disease
Similar results were obtained.
Example 22
Use of compounds in the treatment of CNS disorders
The method of example 19 is repeated except that a compound of the formula is administered to a patient suffering from a CNS disease
Similar results were obtained.
Example 23
Use of compounds in the treatment of CNS disorders
The method of example 19 is repeated except that a compound of the formula is administered to a patient suffering from a CNS disease
Similar results were obtained.
Example 24
Use of compounds in the treatment of CNS disorders
The method of example 19 is repeated except that a compound of the formula is administered to a patient suffering from a CNS disease
Similar results were obtained.
Claims (14)
1. A method of modulating PARP-1 activity in a mammal comprising administering to the mammal an effective amount of an organic aromatic compound having from 4 to about 35 carbon atoms, wherein said organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, and wherein said organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of the particular arginine-34 residue referred to as zinc-1 of PARP-1, wherein when said aromatic compound comprises a heterocyclic ring containing a nitrogen atom, said ring does not contain a carbonyl moiety and does not contain a lactam structure, and said substituent does not contain a benzamide or lactam structure.
2. The method of claim 1, wherein the organic aromatic compound is selected from formulas I and II:
formula I
Wherein R is1、R2、R3And R4Independently selected from H, halogen, optionally substituted hydroxy, substituted amino, optionally substituted lower alkyl, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8Cycloalkyl, or a salt, solvate, isomer, tautomer, metabolite, or prodrug of a compound of formula I;
formula II
Wherein R is1、R2、R3、R4And R5Independently selected from H, halogen, nitro, nitroso, optionally substituted hydroxy, optionally substituted lower alkyl, optionally substituted amino, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group; x is H, N-oxide or optionally substituted alkyl, or a salt, solvate, isomer, tautomer, metabolite or prodrug of a compound of formula II.
3. The method of claim 1, wherein said modulation is inhibition.
4. The method of claim 1, wherein the inhibition is irreversible.
5. A compound of the formula or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof
6. A compound of formula IIa or a salt, solvate, isomer, tautomer, metabolite or prodrug thereof
Formula IIa
Wherein R is1、R2、R3、R4And R5Independently selected from iodine, hydroxyl, nitro, nitroso and optionally substituted amino.
7. The compound of claim 6, wherein R1、R2And R5Is hydrogen, R3Is hydroxy, and R4Is iodine.
8. The compound of claim 6, wherein R1、R2And R5Is hydrogen, R4Is hydroxy, and R3Is iodine.
9. The compound of claim 6, wherein R1、R2And R3Is hydrogen, R5Is iodine, and R4Is a hydroxyl group.
10. The compound of claim 6, wherein R2Is aminopropyl, R3Is iodine, and R4Is hydroxy, R5Is hydrogen.
11. The compound of claim 6, wherein R is2Is aminopropyl, R3Is hydrogen, and R4Is hydroxy, R5Is iodine.
12. A pharmaceutical composition comprising an effective amount of at least one compound of claim 5 or 6 and a pharmaceutically acceptable carrier, excipient and/or diluent.
13. A method of treating a PARP mediated disease comprising administering to a subject in need of such treatment a therapeutically effective amount of an organic aromatic compound having 4 to about 35 carbon atoms, wherein said organic aromatic compound is capable of binding to the arginine-34 moiety located in the zinc finger-1 of the PARP-1 enzyme, wherein said organic aromatic compound has an electron donating ability such that its pi-electron system interacts with the positively charged (cationic) guanidinium moiety of the specific arginine-34 residue referred to as zinc-1 of PARP-1, wherein when said aromatic compound comprises a heterocyclic ring containing a nitrogen atom, said ring does not contain a carbonyl moiety and does not contain a lactam structure, and said substituent does not contain a benzamide or lactam structure.
14. The method of claim 13, wherein the organic aromatic compound is selected from formulas I and II:
formula I
Wherein R is1、R2、R3And R4Independently selected from H, halogen, optionally substituted hydroxy, substituted amino, optionally substituted lower alkyl, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8Cycloalkyl, or salts, solvents of compounds of formula IA compound, isomer, tautomer, metabolite, or prodrug;
formula II
Wherein R is1、R2、R3、R4And R5Independently selected from H, halogen, nitro, nitroso, optionally substituted hydroxy, optionally substituted lower alkyl, optionally substituted amino, optionally substituted phenyl, optionally substituted C4-C10Heteroaryl and optionally substituted C3-C8A cycloalkyl group; x is H, N-oxide or optionally substituted alkyl, or a salt, solvate, isomer, tautomer, metabolite or prodrug of a compound of formula II.
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US60/689,178 | 2005-06-10 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| HK1117165A true HK1117165A (en) | 2009-01-09 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN101233121A (en) | PARP Modulators and Cancer Therapy | |
| CN113164495B (en) | Covalent targeting of E3 ligases | |
| JP5042888B2 (en) | Azaindoles useful as inhibitors of JAK and other protein kinases | |
| EP3151920B1 (en) | Use of inhibitor of apoptosis protein (iap) antagonists in hiv therapy | |
| KR20160029076A (en) | Nuclear transport modulators and uses thereof | |
| CN106488910A (en) | Inhibitors of KRAS G12C | |
| CN107674073A (en) | 3,5 2 as protein kinase modulators substitute 3H imidazoles (or [1,2,3] triazole) simultaneously [4,5 B] pyridine compounds | |
| CN107082779A (en) | It is used as the noval chemical compound of C MET protein kinase modulators | |
| TW200412966A (en) | Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors | |
| Miao et al. | A novel harmine derivative, N-(4-(hydroxycarbamoyl) benzyl)-1-(4-methoxyphenyl)-9H-pyrido [3, 4-b] indole-3-carboxamide (HBC), as histone deacetylase inhibitor: in vitro antiproliferation, apoptosis induction, cell cycle arrest, and antimetastatic effects | |
| KR20190062485A (en) | Methods of treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors | |
| CN119365198A (en) | Use of ALC1 inhibitors and synergy with PARPi | |
| US20150197522A1 (en) | Inauhzin analogues that induce p53, inhibit cell growth, and have antitumor activity | |
| CA2969786A1 (en) | Compositions and methods for enhancing oncolytic virus efficacy | |
| CA3252179A1 (en) | Cgas inhibitors and uses thereof | |
| CA3226555A1 (en) | 14-3-3 protein modulators as antitumor agents | |
| EP3440070B1 (en) | Mdr-reversing 8-hydroxy-quinoline derivatives | |
| HK1117165A (en) | Parp modulators and treatment of cancer | |
| Ao et al. | Design, synthesis, and biological evaluation of AV6 derivatives as novel dual reactivators of latent HIV-1 | |
| CA3224123A1 (en) | Small molecule inhibition of deubiquitinating enzyme josephin domain containing 1 (josd1) as a targeted therapy for leukemias with mutant janus kinase 2 (jak2) | |
| JP7461894B2 (en) | Method for preparing purified atomic quantum clusters | |
| WO2005034943A1 (en) | Indole antiviral compositions and methods | |
| Guo et al. | Novel β‐carboline‐based indole‐4, 7‐quinone derivatives as NAD (P) H: Quinone‐oxidoreductase‐1 inhibitor with potent antitumor activities by inducing reactive oxygen species, apoptosis, and DNA damage | |
| CN108698984B (en) | Bicyclic compounds for use as medicaments, especially for the treatment of Parkinson's disease | |
| CN101282728A (en) | combination of organic compounds |