AU2001231143A1 - Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof - Google Patents

Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof

Info

Publication number
AU2001231143A1
AU2001231143A1 AU2001231143A AU3114301A AU2001231143A1 AU 2001231143 A1 AU2001231143 A1 AU 2001231143A1 AU 2001231143 A AU2001231143 A AU 2001231143A AU 3114301 A AU3114301 A AU 3114301A AU 2001231143 A1 AU2001231143 A1 AU 2001231143A1
Authority
AU
Australia
Prior art keywords
pyridinecarboxamide
nitrophenyl
chloro
optionally substituted
ethoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2001231143A
Inventor
Sui Xiong Cai
John A. Drewe
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cytovia Therapeutics LLC
Original Assignee
Cytovia Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cytovia Inc filed Critical Cytovia Inc
Publication of AU2001231143A1 publication Critical patent/AU2001231143A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Description

Substituted Nicotinamides and Analogs as Activators of Caspases and Inducers of Apoptosis and the Use Thereof
Field of the Invention
This invention is in the field of medicinal chemistry. In particular, the invention relates to substituted nicotinamides and analogs, and the discovery that these compounds are activators of caspases and inducers of apoptosis. The invention also relates to the use of these compounds as therapeutically effective anti-cancer agents.
Description of Background Art
Organisms eliminate unwanted cells by a process variously known as regulated cell death, programmed cell death or apoptosis. Such cell death occurs as a normal aspect of animal development as well as in tissue homeostasis and aging (Glucksmann, A., Biol. Rev. Cambridge Philos. Soc. 26:59-86 (1951); Glucksmann, A., Archives de Biologie
76:419-437 (1965); Ellis, et al, Dev. 112:591-603 (1991); Vaux, et al, Cell 76:777-179 (1994)). Apoptosis regulates cell number, facilitates morphogenesis, removes harmful or otherwise abnormal cells and eliminates cells that have already performed their function. Additionally, apoptosis occurs in response to various physiological stresses, such as hypoxia or ischemia (PCT published application WO96/20721).
There are a number of morphological changes shared by cells experiencing regulated cell death, including plasma and nuclear membrane blebbing, cell shrinkage (condensation of nucleoplasm and cytoplasm), organelle relocalization and compaction, chromatin condensation and production of apoptotic bodies (membrane enclosed particles containing intracellular material) (Orrenius, S., J. Internal Medicine 237:529-536 (1995)).
Apoptosis is achieved through an endogenous mechanism of cellular suicide (Wyllie, A.H., in Cell Death in Biology and Pathology, Bowen and Lockshin, eds., Chapman and Hall (1981), pp. 9-34). A cell activates its internally encoded suicide program as a result of either internal or external signals. The suicide program is executed through the activation of a carefully regulated genetic program (Wyllie, et al, Int. Rev. Cyt. 68:251 (1980); Ellis, et al, Ann. Rev. Cell Bio. 7:663 (1991)). Apoptotic cells and bodies are usually recognized and cleared by neighboring cells or macrophages before lysis. Because of this clearance mechanism, inflammation is not induced despite the clearance of great numbers of cells (Orrenius, S., J. Internal Medicine 237:529-536 (1995)).
It has been found that a group of proteases are a key element in apoptosis (see, e.g., Thornberry, Chemistry arid Biology 5:R97-R103 (1998); Thornbeπy, British Med. Bull 55:478-490 (1996)). Genetic studies in the nematode Caenorhabditis elegans revealed that apoptotic cell death involves at least 14 genes, two of which are the pro-apoptotic (death-promoting) ced (for cell death abnormal) genes, ced-3 and ced-4. CED-3 is homologous to interleukin 1 beta-converting enzyme, a cysteine protease, which is now called caspase-1. When these data were ultimately applied to mammals, and upon further extensive investigation, it was found that the mammalian apoptosis system appears to involve a cascade of caspases, or a system that behaves like a cascade of caspases. At present, the caspase family of cysteine proteases comprises 14 different members, and more may be discovered in the future. All known caspases are synthesized as zymogens that require cleavage at an aspartyl residue prior to forming the active enzyme. Thus, caspases are capable of activating other caspases, in the manner of an amplifying cascade.
Apoptosis and caspases are thought to be crucial in the development of cancer
(Apoptosis and Cancer Chemotherapy, Hickman and Dive, eds., Humana Press (1999)).
There is mounting evidence that cancer cells, while containing caspases, lack parts of the molecular machinery that activates the caspase cascade. This makes the cancer cells lose their capacity to undergo cellular suicide and the cells become cancerous. In the case of the apoptosis process, control points are known to exist that represent points for intervention leading to activation. These control points include the CED-9-BCL-like and CED-3-ICE-like gene family products, which are intrinsic proteins regulating the decision of a cell to survive or die and executing part of the cell death process itself, respectively (see, Schmitt, et al, Biochem. Cell. Biol. 75:301-314 (1997)). BCL-like proteins include BCL-xL and BAX-alpha, which appear to function upstream of caspase activation.
BCL-xL appears to prevent activation of the apoptotic protease cascade, whereas BAX- alpha accelerates activation of the apoptotic protease cascade.
It has been shown that chemotherapeutic (anti-cancer) drugs can trigger cancer cells to undergo suicide by activating the dormant caspase cascade. This may be a crucial aspect of the mode of action of most, if not all, known anticancer drugs (Los, et al, Blood
90:3118-3129 (1997); Friesen, et al, Nat. Med. 2:574 (1996)). The mechanism of action of current antineoplastic drugs frequently involves an attack at specific phases of the cell cycle. In brief, the cell cycle refers to the stages through which cells normally progress during their lifetimes. Normally, cells exist in a resting phase termed G0. During multiplication, cells progress to a stage in which DNA synthesis occurs, termed S. Later, cell division, or mitosis occurs, in a phase called M. Antineoplastic drugs such as cytosine arabinoside, hydroxyurea, 6-mercaptopurine, and methotrexate are S phase specific, whereas antineoplastic drugs such as vincristine, vinblastine, and paclitaxel are M phase specific. Many slow growing tumors, for example colon cancers, exist primarily in the G0 phase, whereas rapidly proliferating normal tissues, for example bone marrow, exist primarily in the S or M phase. Thus, a drug like 6-mercaptopurine can cause bone marrow toxicity while remaining ineffective for a slow growing tumor. Further aspects of the chemotherapy of neoplastic diseases are known to those skilled in the art (See, e.g., Hardman, et al., eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, Ninth Edition, McGraw-Hill, New York (1996), pp. 1225-1287). Thus, it is clear that the possibility exists for the activation of the caspase cascade, although the exact mechanisms for doing so are not clear at this point. It is equally clear that insufficient activity of the caspase cascade and consequent apoptotic events are implicated in various types of cancer. The development of caspase cascade activators and inducers of apoptosis is a highly desirable goal in the development of therapeutically effective antineoplastic agents. Moreover, since autoimmune disease and certain degenerative diseases also involve the proliferation of abnormal cells, therapeutic treatment for these diseases could also involve the enhancement of the apoptotic process through the administration of appropriate caspase cascade activators and inducers of apoptosis.
PCT published patent application WO95/25723 discloses anilide derivatives as fungicides:
wherein, X is O or S;
A is a 6 membered heteroaryl group comprising at least one nitrogen atom, which is optionally substituted by one or more of the group R ;
R1 is alkyl, cycloalkyl, cycloalkenyl, alkenyl, alkynyl, or amino, (each of which is optionally substituted), Y^X-, halogen, cyano, nitro, acyl, acyloxy, optionally substituted heterocyclyl or optionally substituted phenyl; or two adjacent groups together with the carbon atoms to which they are attached can form an optionally substituted benzo ring;
R2 is the same meaning as R1 or two adjacent groups together with the carbon atoms to which they are attached can form an optionally substituted heterocyclic ring;
Y is alkyl, cycloalkyl, cycloalkenyl, alkenyl or alkynyl, each of which is optionally substituted, hydrogen or acyl;
Y1 has the same meaning as Y or is optionally substituted phenyl or optionally substituted heterocyclyl;
Z is C(=X1)-X2-R3, cyano, nitro, amino, acyl, optionally substituted heterocyclyl,
-C(R5)=N-OR6 or-C(R5)=N-NR6R7; R is alkyl, cycloalkyl, cycloalkenyl, alkenyl, alkynyl, phenyl or heterocyclyl, each of which is optionally substituted, hydrogen or an inorganic or organic cationic group; X1 and X2, which may be the same or different, are O or S;
R5, R6 and R7, which may be the same or different, are alkyl, cycloalkyl, cycloalkenyl, alkenyl, alkynyl, phenyl or heterocyclyl, each of which is optionally substituted or hydrogen or R6 and R7 together with the atom(s) to which they are attached can form a ring; and n is 0 to 4, together with complexes with metal salts, as well as salts with bases of compounds which are acids and salts with acidsof compounds which are bases, with the proviso that when Y is hydrogen and i) when Z is carboxy, methoxycarbonyl or ethoxycarbonyl ring A is not unsubstututed pyridyl or pyrazinyl; and ii) when Z is carboxy and n is 0, A is not 2-chloro-3-pyridyl, 6-(2-diethylaminoethoxy)-3- pyridyl or a 2-pyridyl group.
PCT WO9936391 discloses benzenesulfonamide, benzamide, diarylsulfone and benzophenone compounds as pharmacological agents in the treatment of cancer, psoriasis, vascular restenosis, infections, atherosclerosis and hypercholesterolemia:
wherein, A represents N or C-R1, B represents N or C-R5; and
R1 and R5 independently represent hydrogen, halogen, (Cι-C8)alkyl, (Cl-C8)heteroalkyl, -OR6, -NR6R7, -S(O)mR6, -CN, -NO2, -S(O)nNR6R7, or -N3; wherein R6 and R7 are independently selected from hydrogen, (C1-C8)alkyl, ( -C^heteroalkyl; m is an integer of from 0 to 3; n is an integer of from 1 to 2;
R2 and R3 are independently -OR8, -SR8, and -NR8R9, wherein R8 and R9 are independently hydrogen, (C1-C6)alkyl, (C1-C6)heteroalkyl; R4 is hydrogen, (C1-C8)alkyl, (C1-C8)heteroalkyl, -OR10, -SR10, or -NR10Rπ; wherein R10 and R11 are independently hydrogen, (C1-C6)alkyl, (C1-C6)heteroalkyl; optionally, R1 is linked to R2 to form a fused ring, R2 is linked to R3 to form a fused ring, or R2 is linked to both R1 and R3 to form two additional fused rings; X represents -S(O)p or -C(O)-, wherein p is 1 or 2;
Y represents a single bond, -CH2- or -N(R12)-, wherein R12 is selected from hydrogen, (Ci- C6)alkyl, and arylalkyl; and Z represents an aryl group or an arylalkyl group.
Summary of the Invention
The present invention is related to the discovery that substituted nicotinamides and nicotinamide analogs as represented in Formula V are activators of the caspase cascade and inducers of apoptosis:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
Ar' and Ar are independently optionally substituted aryl or optionally substituted heteroaryl, provided that the ring structure of said optionally substituted heteroaryl comprises not more than two nitrogen atoms; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
Thus, an aspect of the present invention is directed to the use of compounds of Formula V as inducers of apoptosis.
A second aspect of the present invention is to provide a method for treating, preventing or ameliorating neoplasia and cancer by administering a compound of Formula V to a mammal in need of such treatment. Many of the compounds within the scope of the present invention are novel compounds. Therefore, a third aspect of the present invention is to provide novel compounds of Formula N, and to also provide for the use of these novel compounds for treating, preventing or ameliorating neoplasia and cancer. A fourth aspect of the present invention is to provide a pharmaceutical composition useful for treating disorders responsive to the induction of apoptosis, containing an effective amount of a compound of Formula N in admixture with one or more pharmaceutically acceptable carriers or diluents.
A fifth aspect of the present invention is directed to methods for the preparation of novel compounds of Formula N.
Brief Description of the Drawings
Figs. 1 A-B depict fluorescent micrographs of Jurkat cells as controls and as treated with drug and stained with a fluorescent DΝA probe, Sytolό. Fig. 1A depicts control cells. Fig. IB depicts cells treated with 5 μM of Ν-(4-methoxy-2-nitrophenyl)-3- pyridinecarboxamide for 24 h, showing shrunken and fragmented nuclei.
Fig. 2 is a graph showing mitotic arrest in Jurkat cells treated for 6 h with different concentrations of N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide. Fig. 2 shows increasing percent of mitotic arrest with increasing drug concentration up to a concentration of 5 μM.
Figs. 3A-B are graphs showing drug induced cell cycle arrest and apoptosis in
T47D cells. Fig. 3A: control cells showing most of the cells in G1(M2). Fig. 3B: cells treated with 10 μM of N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide for 48 h showing a reduction in the G1(M2), an increase in the G2/M (M4) and sub-diploid DNA content of cells (Ml).
Figs. 4 A-B are graphs showing drug induced cell cycle arrest and apoptosis in
T47D cells. Fig. 4A: control cells showing most of the cells in G1(M2). Fig. 4B: cells treated with 1 μM of 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide for 48 h showing a reduction in the G1(M2), an increase in the G2 M (M4) and sub-diploid DNA content of cells (Ml). Fig. 5 is a graph showing inhibition of proliferation of HeLa cells treated for 48 h with different concentrations of 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3- pyridinecarboxamide. Fig. 5 shows that 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3- pyridinecarboxa ide inhibit colony formation of HeLa cells with an IC50 of about 100 nM.
Detailed Description of the Invention
The present invention arises out of the discovery that substituted nicotinamides and nicotinamide analogs, as represented in Formula V, are potent and highly efficacious activators of the caspase cascade and inducers of apoptosis:
or a pharmaceutically acceptable salt or prodrug thereof, wherein: Ar' and Ar are independently optionally substituted aryl or optionally substituted heteroaryl, provided that the ring structure of said optionally substituted heteroaryl comprises not more than two nitrogen atoms; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted. Therefore compounds of Formula V are useful for treating disorders responsive to induction of apoptosis.
Specifically, compounds useful in this aspect of the present invention are represented by Formula I:
or pharmaceutically acceptable salts or prodrugs thereof, wherein:
A is N or C-R8; B is N or C-R9, D is N or C-R10, E is N or C-R6, F is N or C-R7, provided that not more than two of A, B, D, E, and F are N at the same time; Ar is optionally substituted and is aryl or heteroaryl; R6-Rιo are independently hydrogen, halo, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, haloalkoxy, carboxy, carbonylamido or alkylthiol; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
Preferred compounds of Formula I include compounds wherein Ar is optionally substituted phenyl, naphthyl, pyridyl, quinolyl, isoquinolyl, thienyl, furyl, or pyrrolyl. Preferred compounds of Formula I also include compounds wherein A is N, B is C-R9, D is C-R10, E is C-R6 and F is C-R7. Preferred compounds of Formula I also include compounds wherein A and D are N, B is C-R9, E is C-R6 and F is C-R7. Preferred compounds of Formula I also include compounds wherein A and E are N, B is C-R9, D is C-R10 and F is C-R7. Preferred compounds of Formula I also include compounds wherein Rπ is hydrogen.
Preferred structures of Formula I are substituted nicotinamides and analogs represented by Formulae II-TV. In particular, a preferred embodiment is represented by Formula II:
or pharmaceutically acceptable salts or prodrugs thereof, wherein R7, R , Rπ, D, E and Ar are as defined previously with respect to Formula I. Preferred compounds falling within the scope of Formula JJ include compounds wherein Ar is optionally substituted phenyl or pyridyl. Preferred compounds of Formula II also include compounds wherein D is C-R10 and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein D are N and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein E are N and D is C-Rto- Preferred compounds of Formula JJ also include compounds wherein R6-R and R^ are independently hydrogen or fluoro.
Another preferred embodiment is represented by Formula JJJ:
or pharmaceutically acceptable salts or prodrugs thereof, wherein R7, R9, Rπ, D, and E are as defined previously with respect to Formula I;
Ri-R5 are independently hydrogen, halo, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, haloalkoxy, carboxy, carbonylamido or alkylthiol; or
Ri and R2, or R2 and R3, or R3 and R4, or Rφ and R5 may be taken together to form a carbocycle or heterocycle, including -OCH2O-, -(CH2)3-, -(CH2)4-, -OCH2CH2O-, -CH2N(R)CH2-, -CH2CH2N(R)CH2- -CH2N(R)CH2CH2-, -CH=CH-CH=CH-, -N(R)-CH=CH~ -CH=CH-N(R)-, -O-CH=CH-, -CH=CH-O-, -S-CH=CH-, -CH=CH-S- -N=CH-CH=CH-, -CH=N-CH=CH- -CH=CH-N=CH-,
-CH=CH-CH=N- and -N=CH-CH=N-, wherein the carbocycle or heterocycle is optionally substituted, and R is hydrogen, CMO alkyl, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl or aminoalkyl.
Preferred compounds falling within the scope of Formula JJ include compounds wherein D is C-R10 and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein D are N and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein E are N and D is C-R10. Preferred compounds of Formula JJ also include compounds wherein R6-R7 and R10 are hydrogen or fluoro. Preferred compounds of Formula HI also include compounds wherein R and R3 is not hydrogen. Preferred compounds of Formula JJJ also include compounds wherein R2 and R4 are hydrogen or fluoro. Another group of preferred compounds of Formula JJJ include compounds wherein Rπ is hydrogen.
Another preferred embodiment is represented by Formula TV:
or pharmaceutically acceptable salts or prodrugs thereof, wherein Ri, R3, R5, R9, D arid E are as defined previously with respect to Formulae I and JJJ.
Preferred compounds falling within the scope of Formula TV include compounds wherein D is C-R10 and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein D are N and E is C-R6. Preferred compounds of Formula JJ also include compounds wherein E are N and D is C-R10. Preferred compounds of Formula IV also include compounds wherein Ri and R3 is not hydrogen. Especially preferred compounds of Formula TV include compounds wherein R\ is nitro, cyano, trifluoromethyl and methyl. Especially preferred compounds of Formula TV also include compounds wherein R3 is halo, haloalkyl, alkyl, amino, cyano, acyloxy, azido, alkoxy, aryloxy, arylalkoxy, haloalkoxy, or alkylthiol. Another preferred embodiment is represented by Formula VT:
or a pharmaceutically acceptable salt or prodrug thereof, wherein
R Rs, R7 and R9-Rio are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
Another preferred embodiment is represented by Formula VTT:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
RιrR3, RS-RΪO are are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted. Exemplary preferred compounds that may be employed in the method of the invention include, without limitation:
N-(4-Methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Chloro-N-(4-methyl-2-nitrophenyl)-3-pyridinecarboxamide;
6-Cr oro-N-(4-methoxy-2-nitrophenyl)-l-N-oxide-3-pyridinecarboxamide;
6-Chloro-iV-(4-chloro-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4,5-difluoro-2-nitrophenyl)-3-pyridinecarboxamide;
6-Fluoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Chloro-N-(4-fluoro-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(2-nitro-4-trifluoromethylphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(3-bromo-4-methoxy-6-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(2-nitro-4-trifluoromethoxyphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-benzyloxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Methyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
5,6-Dichloro-iV-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(2-methyl-4-methoxyphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-cyano-2-nitrophenyl)-3-pyridinecarboxamide;
4-Chloro-N-(4-ethoxy-2-nitrophenyl)-benzoylamide; 6-Cl oro-N-(4-ethoxy-2-ni1xophenyl)-N-methyl-3-pyridinecarboxamide;
6-Chloro-N-(2-cyano-4,5-dimethoxyphenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
N-(4-Methyl-2-nitrophenyl)-3-pyridinecarboxamide;
6-(2,2,2-Trifluoroethoxy)-N-(4-ethoxy-2-nitrophenyl)-3-p ridinecarboxamide; N-(4-Ethoxy-2-nitrophenyl)-2-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-5-pyrimidinecarboxamide; 6-Dimethylamino-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-t-butyl-2-nitoophenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-l-N-oxide-2-pyridinecarboxamide;
6-Trifluoromethyl-Λ^-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Chloro-iV-(4-chloro-2-trifluoromethylphenyl)-3-pyridinecarboxarnide;
6-Methyl-N-(4-emoxy-2-nitxophenyl)-3-pyrazinecarboxamide;
6-Chloro-N-(4-chloro-2-cyanophenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide;
6-Chloro-N-(2,4-dimethyl-6-nitrophenyl)-3-pyridinecarboxamide; 4-Chloromethyl-N-(4-ethoxy-2-mtrophenyl)-benzoylamide;
6-Clιloro-N-(3,4-dimethoxy-6-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(2-pyrazinyl)-3-pyridinecarboxamide;
6-Chloro-2V-( 1 -N-oxide-3-cyano-5-chloromethyl-2-pyrazinyl)-3- pyridinecarboxamide; 6-Chloro-N-(2-cyano-4-methylphenyl)-3-pyridinecarboxamide;
6-Cldoro-N-(4-chloro-2-methyl-6-ni1xophenyl)-3-pyridinecarboxamide;
4-Trifluoromethyl-Λ^-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
4-Chloro-2-nitro-N-(6-chloro-3-pyridyl)-benzoylamide; 4-Chloro-2-nitro-N-(6-methoxy-3-pyridyl)-benzoylamide;
4-Bromomethyl-3-nitro-N-(6-cUoro-3-pyridyl)-benzoylarnide;
N-(4-Ethoxy-2-nitrophenyl)-3-furancarboxamide; and
N-(4-Ethoxy-2-nitrophenyl)-3-pyrrolecarboxamide.
The present invention is also directed to novel compounds within the scope of Formulae I-NJJ. In one preferred embodiment, the novel compounds of the present invention are compounds of Formula TIT: or pharmaceutically acceptable salts or prodrugs thereof, wherein:
R1 and R5 are independently selected from the group consisting of hydrogen, hydroxy, alkyl, alkoxy, halogen, NO2, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl, provided that at least one of Ri and R5 is selected from the group consisting of NO2, cyano, alkyl and haloalkyl;
R2 and R4 are independently selected from the group consisting of hydrogen, hydroxy, halogen, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl;
R3 is alkyl, Cl, F, haloalkyl, alkoxy, arylalkoxy, cyano, haloalkyloxy, amino or aminoalkyl;
R6 is hydrogen, hydroxy, alkyl, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl;
R7 is hydrogen, hydroxy, alkyl, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl;
R9 is hydroxy, alkyl, halogen, NO2, haloalkyl, alkoxy, cyano, haloalkyloxy, amino or aminoalkyl;
R10 is hydrogen, hydroxy, alkyl, Cl, F, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl; and
Rπ is hydrogen, alkyl or haloalkyl.
In a further preferred embodiment, the compounds of the present invention are compounds of Formula TV:
or pharmaceutically acceptable salts or prodrugs thereof, wherein:
R3 is alkyl, Cl, F, haloalkyl, alkoxy, arylalkoxy, cyano, haloalkyloxy, amino or aminoalkyl; and
R9 is hydroxy, alkyl, halogen, NO2, haloalkyl, alkoxy, cyano, haloalkyloxy, amino or aminoalkyl.
In another preferred embodiment, the compounds of the present invention are compounds of Formula (NT):
or pharmaceutically acceptable salts or prodrugs thereof, wherein
Ri-R5, R7 and R9-Rio are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
Rii is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
In another preferred embodiment, the novel compounds of the present invention are compounds of Formula (Nil):
or pharmaceutically acceptable salts or prodrugs thereof, wherein:
R R3, R5-Rio are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
More preferably at least one of R6 and R7 is selected from the group consisting of NO2, cyano, alkyl and haloalkyl.
Exemplary preferred compounds of the present invention include, without limitation:
6-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-iV-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-iV-(4-methyl-2-nitrophenyl)-3-pyridinecarboxamide;
6-CWoro-N-(4-methoxy-2-ni1xophenyl)-l-N-oxide-3-pyridinecarboxamide;
6-Chloro-N-(4-chloro-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4,5-difluoro-2-nitrophenyl)-3-pyridinecarboxarride;
6-Fluoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-fluoro-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-1xifluoromethyl-2-nitrophenyl)-3-pyridinecarboxamide;
6-CUoro-N-(3-bromo-4-methoxy-6-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-N-(2-nitro-4-1xifluoromethoxyphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-benzyloxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Methyl-iV-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
5,6-Dichloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-CUoro-N-(2-methyl-4-methoxyphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-cyano-2-nitrophenyl)-3-pyridinecarboxamide; 4-Chloro-N-(4-ethoxy-2-nitrophenyl)-benzoylamide;
6-CWoro-N-(4-ethoxy-2-nitrophenyl)-N-methyl-3-pyridinecarboxarnide;
6-Chloro-N-(2-cyano-4,5-dimethoxyphenyl)-3-pyridinecarboxarnide;
6-(2,2,2-Trifluoroethoxy)-iV-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-2-pyridinecarboxamide; N-(4-Ethoxy-2-nitrophenyl)-5-pyrimidinecarboxamide;
6-Dimethylamino-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-iV-(4-t-butyl-2-nitrophenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)- 1 -N-oxide-2-pyridinecarboxamide;
6-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxarnide; 6-Chloro-N-(4-chloro-2-trifluoromethylphenyl)-3-pyridinecarboxamide;
6-Methyl-N-(4-ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide;
6-Chloro-N-(4-chloro-2-cyanophenyl)-3-pyridinecarboxamide;
N-(4-Ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide;
6-Chloro-N-(2,4-dimethyl-6-nitrophenyl)-3-pyridinecarboxamide; 4-Chloromethyl-iV-(4-ethoxy-2-nitrophenyl)-benzoylamide;
6-Chloro-N-(3,4-dimethoxy-6-nitrophenyl)-3-pyridinecarboxamide;
6-Chloro-iV-(2-pyrazinyl)-3-pyridinecarboxamide;
6-Chloro-N-( 1 -N-oxide-3-cyano-5-chloromethyl-2-pyrazinyl)-3- pyridinecarboxamide; 6-Chloro-iV-(2-cyano-4-methylphenyl)-3-pyridinecarboxamide;
6-Chloro-N-(4-chloro-2-methyl-6-nitrophenyl)-3-pyridinecarboxamide;
4-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
6-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
4-Chloro-2-nitro-N-(6-chloro-3-pyridyl)-benzoylarnide; 4-Chloro-2-mtro-N-(6-methoxy-3-pyridyl)-benzoylamide; and
4-Bromomethyl-3-nitro-N-(6-chloro-3-pyridyl)-benzoylamide. Useful alkyl groups include straight-chained and branched CMO alkyl groups, more preferably C1-6 alkyl groups. Typical C1-10 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, 3-pentyl, hexyl and octyl groups, which may be optionally substituted. Useful alkoxy groups include oxygen substituted by one of the Cno alkyl groups mentioned above, which may be optionally substituted.
Useful alkylthio groups include sulphur substituted by one of the .io alkyl groups mentioned above, which may be optionally substituted. Also included are the sulfoxides and sulfones of such alkylthio groups. Useful amino groups include -NH2, -NHRts and -NRi5R16, wherein R15 and R16 are C1-10 alkyl or cycloalkyl groups, or R15 and R16 are combined with the N to form a ring structure, such as a piperidine, or R15 and R16 are combined with the N and other group to form a ring, such as a piperazine. The alkyl group may be optionally substituted.
Optional substituents on the alkyl groups include one or more halo, hydroxy, carboxyl, amino, nitro, cyano, Ci-C6 acylamino, Ct-Cβ acyloxy, Ci-C6 alkoxy, aryloxy, alkylthio, C6-Cio aryl, C -C7 cycloalkyl, C -C6 alkenyl, C2-C6 alkynyl, C6-Cio aryl(C2-C6)alkenyl, C6-Cio aryl(C2-C6)alkynyl, saturated and unsaturated heterocyclic or heteroaryl.
Optional substituents on the aryl, aralkyl and heteroaryl groups include one or more halo, Ci-C6 haloalkyl, C6-C10 aryl, C4-C7 cycloalkyl, -Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C6-C10 aryl(C1-C6)alkyl, C6-C10 aryl(C2-C6)alkenyl, C6-C10 aryl(C2-C6)alkynyl,
Ci-C6 hydroxyalkyl, nitro, amino, ureido, cyano, Ci-C6 acylamino, hydroxy, thiol, - acyloxy, azido, Ci-C6 alkoxy or carboxy.
The term "aryl" as employed herein by itself or as part of another group refers to monocyclic, bicyclic or tricyclic aromatic groups containing from 6 to 14 carbons in the ring portion.
Useful aryl groups include C6-14 aryl, preferably C6-1o aryl. Typical C6-14 aryl groups include phenyl, naphthyl, phenanthrenyl, anthracenyl, indenyl, azulenyl, biphenyl, biphenylenyl and fluorenyl groups. Useful cycloalkyl groups are C3-8 cycloalkyl. Typical cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. Useful saturated or partially saturated carbocyclic groups are cycloalkyl groups as described above, as well as cycloalkenyl groups, such as cyclopentenyl, cycloheptenyl and cyclooctenyl.
Useful halo or halogen groups include fluorine, chlorine, bromine and iodine. Useful arylalkyl groups include any of the above-mentioned CMO alkyl groups substituted by any of the above-mentioned C6-14 aryl groups. Preferably the arylakyl group is benzyl, phenethyl or naphthylmethyl.
Useful haloalkyl groups include C1-10 alkyl groups substituted by one or more fluorine, chlorine, bromine or iodine atoms, e.g., fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl groups.
Useful acylamino (acylamido) groups are any C1-6 acyl (alkanoyl) attached to an amino nitrogen, e.g., acetamido, chloroacetamido, propionamido, butanoylamido, pentanoylamido and hexanoylamido, as well as aryl-substituted Ci-6 acylamino groups, e.g., benzoylamido, and pentafluorobenzoylamido.
Useful acyloxy groups are any C1-6 acyl (alkanoyl) attached to an oxy (-O-) group, e.g., formyloxy, acetoxy, propionoyloxy, butanoyloxy, pentanoyloxy and hexanoyloxy.
The term heterocycle is used herein to mean a saturated or partially saturated 3-7 membered monocyclic, or 7-10 membered bicyclic ring system, which consists of carbon atoms and from one to four heteroatoms independently selected from the group consisting of O, N, and S, wherein the nitrogen and sulfur heteroatoms can be optionally oxidized, the nitrogen can be optionally quatemized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring, and wherein the heterocyclic ring can be substituted on carbon or on a nitrogen atom if the resulting compound is stable.
Useful saturated or partially saturated heterocyclic groups include tetrahydrofuranyl, pyranyl, piperidinyl, piperazinyl, pyrrolidinyl, imidazolidinyl, imidazolinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, isochromanyl, chromanyl, pyrazolidinyl pyrazolinyl, tetronoyl and tetramoyl groups. The term "heteroaryl" as employed herein refers to groups having 5 to 14 ring atoms; 6, 10 or 14 π electrons shared in a cyclic array; and containing carbon atoms and 1, 2 or 3 oxygen, nitrogen or sulfur heteroactoms.
Useful heteroaryl groups include thienyl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxanthiinyl, 2H- pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, 4H-quinolizinyl, isoquinolyl, quinolyl, phthalzinyl, naphthyridinyl, quinozalinyl, cinnolinyl, pteridinyl, carbazolyl, β-carbolinyl, phenanthridinyl, acrindinyl, perimidinyl, phenanthrolinyl, phenazinyl, isothiazolyl, phenothiazinyl, isoxazolyl, furazanyl, phenoxazinyl, 1,4- dihydroquinoxaline-2,3-dione, 7-aminoisocoumarin, pyrido[l,2-a]pyrimidin-4-one, 1,2- benzoisoxazol-3-yl, benzimidazolyl, 2-oxindolyl and 2-oxobenzimidazolyl. Where the heteroaryl group contains a nitrogen atom in a ring, such nitrogen atom may be in the form of an N-oxide, e.g., a pyridyl N-oxide, pyrazinyl N-oxide and pyrimidinyl N-oxide. Some of the compounds of the present invention may exist as stereoisomers including optical isomers. The invention includes all stereoisomers and both the racemic mixtures of such stereoisomers as well as the individual enantiomers that may be separated according to methods that are well known to those of ordinary skill in the art.
Examples of pharmaceutically acceptable addition salts include inorganic and organic acid addition salts such as hydrochloride, hydrobromide, phosphate, sulphate, citrate, lactate, tartrate, maleate, fumarate, mandelate and oxalate; and inorganic and organic base addition salts with bases such as sodium hydroxy, Tris(hydroxymethyl)aminomethane (TRIS, tromethane) and N-methyl-glucamine.
Examples of prodrugs of the compounds of the invention include the simple esters of carboxylic acid containing compounds (e.g., those obtained by condensation with a C1-4 alcohol according to methods known in the art); esters of hydroxy containing compounds (e.g., those obtained by condensation with a C1-4 carboxylic acid, C3-6 dioic acid or anhydride thereof such as succinic and fumaric anhydrides according to methods known in the art); imines of amino containing compounds (e.g., those obtained by condensation with a Cι-4 aldehyde or ketone according to methods known in the art); carbamate of amino containing compounds such as those described by Leu, et. al, (J. Med. Chem. 42:3623- 3628 (1999)) and Greenwald, et. al, (J. Med. Chem. 42:3651-3661 (1999)); and acetals and ketals of alcohol containing compounds (e.g., those obtained by condensation with chloromethyl methyl ether or chloromethyl ethyl ether according to methods known in the art).
The compounds of this invention may be prepared using methods known to those skilled in the art, or the novel methods of this invention. Specifically, the compounds of this invention with Formulae I-Nπ may be prepared as illustrated by the exemplary reaction in Scheme 1. Reaction of an acyl chloride with an amine in the presence a base such as triethylamine or N,N-diisopropylethylamine produces the amide.
Scheme 1
Alternatively, reaction of an acid with an amine in the presence of a coupling reagent such as EDC, or cyanuric chloride, also produces the amide as shown by the exemplary reaction in Scheme 2.
Scheme 2
An important aspect of the present invention is the discovery that compounds having Formula I-Nπ are activators of caspases and inducers of apoptosis. Therefore, these compounds are useful in a variety of clinical conditions in which there is uncontrolled cell growth and spread of abnormal cells, such as in the case of cancer.
Another important aspect of the present invention is the discovery that compounds having Formula I-Nπ are potent and highly efficacious activators of caspases and inducers of apoptosis in drug resistant cancer cells, such as breast and prostate cancer cells, which enables these compounds to kill these drug resistant cancer cells. In comparison, most standard anti-cancer drugs are not effective in killing drug resistant cancer cells under the same conditions. Therefore, compounds of this invention are useful for the treatment of drag resistant cancer in animals. The present invention includes a therapeutic method useful to modulate in vivo apoptosis or in vivo neoplastic disease, comprising administering to a subject in need of such treatment an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis. The present invention also includes a therapeutic method comprising administering to an animal an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-VH, wherein said therapeutic method is useful to treat cancer, which is a group of diseases characterized by the uncontrolled growth and spread of abnormal cells. Such diseases include, but are not limited to, Hodgkin's disease, non-Hodgkin's lymphomas, acute lymphocytic leukemia, chronic lymphocytic leukemia, multiple myeloma, neuroblastoma, breast carcinomas, ovarian carcinomas, lung carcinomas, Wilms' tumor, cervical carcinomas, testicular carcinomas, soft-tissue sarcomas, primary macroglobulinemia, bladder carcinomas, chronic granulocytic leukemia, primary brain carcinomas, malignant melanoma, small-cell lung carcinomas, stomach carcinomas, colon carcinomas, malignant pancreatic insulinoma, malignant carcinoid carcinomas, malignant melanomas, choriocarcinomas, mycosis fungoides, head or neck carcinomas, osteogenic sarcoma, pancreatic carcinomas, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, genitourinary carcinomas, thyroid carcinomas, esophageal carcinomas, malignant hypercalcemia, cervical hyperplasia, renal cell carcinomas, endometrial carcinomas, polycythemia vera, essential thrombocytosis, adrenal cortex carcinomas, skin cancer, and prostatic carcinomas.
In practicing the therapeutic methods, effective amounts of compositions containing therapeutically effective concentrations of the compounds formulated for oral, intravenous, local and topical application, for the treatment of neoplastic diseases and other diseases in which caspase cascade mediated physiological responses are implicated, are administered to an individual exhibiting the symptoms of one or more of these disorders. The amounts are effective to ameliorate or eliminate one or more symptoms of the disorders. An effective amount of a compound for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce, the symptoms associated with the disease. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective. The amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease. Typically, repeated administration is required to achieve the desired amelioration of symptoms
Jn another embodiment, a pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt of said compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis in combination with a pharmaceutically acceptable vehicle is provided. Another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis, in combination with at least one known cancer chemotherapeutic agent, or a pharmaceutically acceptable salt of said agent. Examples of known cancer chemotherapeutic agents which may be used for combination therapy include, but not are limited to alkylating agents such as busulfan, cis-platin, mitomycin C, and carboplatin; antimitotic agents such as colchicine, vinblastine, paclitaxel, and docetaxel; topo I inhibitors such as camptothecin and topotecan; topo π inhibitors such as doxorubicin and etoposide; RΝA/DΝA antimetabolites such as 5-azacytidine, 5- fluorouracil and methotrexate; DΝA antimetabolites such as 5-fluoro-2'-deoxy-uridine, ara-C, hydroxyurea and thioguanine; antibodies such as Herceptin® and Rituxan®. Other known cancer chemotherapeutic agents which may be used for combination therapy include melphalan, chlorambucil, cyclophosamide, ifosfamide, vincristine, mitoguazone, epirubicin, aclarubicin, bleomycin, mitoxantrone, elliptinium, fludarabine, octreotide, retinoic acid, tamoxifen and alanosine. In practicing the methods of the present invention, the compound of the invention may be administered together with at least one known chemotherapeutic agent as part of a unitary pharmaceutical composition. Alternatively, the compound of the invention may be administered apart from at least one known cancer chemotherapeutic agent. In one embodiment, the compound of the invention and at least one known cancer chemotherapeutic agent are administered substantially simultaneously, i.e. the compounds are administered at the same time or one after the other, so long as the compounds reach therapeutic levels in the blood at the same time. On another embodiment, the compound of the invention and at least one known cancer chemotherapeutic agent are administered according to their individual dose schedule, so long as the compounds reach therapeutic levels in the blood.
Another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a bioconjugates of said compound of Formulae I-Vπ, which functions as a caspase cascade activator and inducer of apoptosis, in bioconjugation with at least one known therapeutically useful antibody, such as Herceptin® or Rituxan®, growth factors such as DGF, NGF, cytokines such as JL-2, JL-4, or any molecule that binds to the cell surface. The antibodies and other molecules will deliver the compound of Formulae I-VTT to its targets and make it an effective anticancer agent. The bioconjugates could also enhance the anticancer effect of therapeutically useful antibodies, such as Herceptin® or Rituxan®. Similarly, another embodiment of the present invention is directed to a composition effective in inhibiting neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis, in combination with radiation therapy. In this embodiment, the compound of the invention may be administered at the same time as the radiation therapy is administered or at a different time. Yet another embodiment of the present invention is directed to a composition effective for post-surgical treatment of cancer, comprising a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-VJJ, which functions as a caspase cascade activator and inducer of apoptosis. The invention also relates to a method of treating cancer by surgically removing the cancer and then treating the animal with one of the pharmaceutical compositions described herein.
A wide range of immune mechanisms operate rapidly following exposure to an infectious agent. Depending on the type of infection, rapid clonal expansion of the T and B lymphocytes occurs to combat the infection. The elimination of the effector cells following an infection is one of the major mechanisms for maintaining immune homeostasis. The elimination of the effector cells has been shown to be regulated by apoptosis. Autoimmune diseases have lately been determined to occur as a consequence of deregulated cell death. In certain autoimmune diseases, the immune system directs its powerful cytotoxic effector mechanisms against specialized cells such as oligodendrocytes in multiple sclerosis, the beta cells of the pancreas in diabetes mellitus, and thyrocytes in
Hashimoto's thyroiditis (Ohsako, S. & Elkon, K.B., Cell Death Differ. 6:13-21 (1999)). Mutations of the gene encoding the lymphocyte apoptosis receptor Fas/APO-l/CD95 are reported to be associated with defective lymphocyte apoptosis and autoimmune lymphoproliferative syndrome (ALPS), which is characterized by chronic, histologically benign splenomegaly, generalized lymphadenopathy, hypergammaglobulinemia, and autoantibody formation. (Infante, A.J., et al, J. Pediatr. 133:629-633 (1998) and Vaishnaw, A.K., et al, J. Clin. Invest. 703:355-363 (1999)). It was reported that overexpression of Bcl-2, which is a member of the bcl-2 gene family of programmed cell death regulators with anti-apoptotic activity, in developing B cells of transgenic mice, in the presence of T cell dependent costimulatory signals, results in the generation of a modified B cell repertoire and in the production of pathogenic autoantibodies (Lopez- Hoyos, M., et al, Int. J. Mol Med. 7:475-483 (1998)). It is therefore evident that many types of autoimmune disease are caused by defects of the apoptotic process. One treatment strategy for such diseases is to turn on apoptosis in the lymphocytes that are causing the autoimmune disease (O'Reilly, L.A. & Strasser, A., Inflamm. Res. 48:5-21
(1999)). Fas-Fas ligand (FasL) interaction is known to be required for the maintenance of immune homeostasis. Experimental autoimmune thyroiditis (EAT), characterized by autoreactive T and B cell responses and a marked lymphocytic infiltration of the thyroid, is a good model to study the therapeutic effects of FasL. Batteux, F., et al, (J. Immunol 752:603-608 (1999)) reported that by direct injection of DNA expression vectors encoding
FasL into the inflammed thyroid, the development of lymphocytic infiltration of the thyroid was inhibited and induction of infiltrating T cells death was observed. These results show that FasL expression on thyrocytes may have a curative effect on ongoing EAT by inducing death of pathogenic autoreactive infiltrating T lymphocytes. Bisindolylmaleimide NTH is known to potentiate Fas-mediated apoptosis in human astrocytoma 1321Ν1 cells and in Molt-4T cells; both of which were resistant to apoptosis induced by anti-Fas antibody in the absence of bisindolylmaleimide VTA. Potentiation of Fas-mediated apoptosis by bisindolylmaleimide VIH was reported to be selective for activated, rather than non-activated, T cells, and was Fas-dependent. Zhou T., et al, (Nat. Med. 5:42-48 (1999)) reported that administration of bisindolylmaleimide VTH to rats during autoantigen stimulation prevented the development of symptoms of T cell-mediated autoimmune diseases in two models, the Lewis rat model of experimental allergic encephalitis and the Lewis adjuvant arthritis model. Therefore, the application of a Fas- dependent apoptosis enhancer such as bisindolylmaleimide VHI may be therapeutically useful for the more effective elimination of detrimental cells and inhibition of T cell- mediated autoimmune diseases. Therefore an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis, should be an effective treatment for autoimmune diseases. Psoriasis is a chronic skin disease that is characterized by scaly red patches.
Psoralen plus ultraviolet A (PUVA) is a widely used and effective treatment for psoriasis vulgaris and Coven, et al, Photodermatol. Photoimmunol Photomed. 75:22-27 (1999), reported that lymphocytes treated with psoralen 8-MOP or TMP and UVA, displayed DΝA degradation patterns typical of apoptotic cell death. Ozawa, et al, J. Exp. Med. 189:111-718 (1999) reported that induction of T cell apoptosis could be the main mechanism by which 312-nm UNB resolves psoriasis skin lesions. Low doses of methotrexate may be used to treat psoriasis to restore a clinically normal skin. Heenen, et al, Arch. Dermatol. Res. 290:240-245 (1998), reported that low doses of methotrexate may induce apoptosis and that this mode of action could explain the reduction in epidermal hyperplasia during treatment of psoriasis with methotrexate. Therefore, an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis, should be an effective treatment for psoriasis.
Synovial cell hyperplasia is a characteristic of patients with rheumatoid arthritis (RA). It is believed that excessive proliferation of RA synovial cells, as well as defects in synovial cell death, may be responsible for synovial cell hyperplasia. Wakisaka, et al,
Clin. Exp. Immunol. 774:119-128 (1998), found that although RA synovial cells could die via apoptosis through a Fas FasL pathway, apoptosis of synovial cells was inhibited by proinflammatory cytokines present within the syriovium. Wakisaka, et al, also suggested that inhibition of apoptosis by the proinflammatory cytokines may contribute to the outgrowth of synovial cells, and lead to pannus formation and the destruction of joints in patients with RA. Therefore, an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Nπ, which functions as a caspase cascade activator and inducer of apoptosis, should be an effective treatment for rheumatoid arthritis. There has been an accumulation of convincing evidence that apoptosis plays a major role in promoting resolution of the acute inflammatory response. Νeutrophils are constitutively programmed to undergo apoptosis, thus limiting their pro-inflammatory potential and leading to rapid, specific, and non-phlogistic recognition by macrophages and semi-professional phagocytes (Savill, J., J. Leukoc. Biol. 57:375-380 (1997)). Boirivant, et al, Gastroenterology 775:557-565 (1999), reported that lamina propria T cells, isolated from areas of inflammation in Crohn's disease, ulcerative colitis, and other inflammatory states, manifest decreased CD2 pathway-induced apoptosis. In addition, studies of cells from inflamed Crohn's disease tissue indicate that this defect is accompanied by elevated Bcl-2 levels. Therefore, an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-VTT, which functions as a caspase cascade activator and inducer of apoptosis, should be an effective treatment for inflammation.
Pharmaceutical compositions within the scope of this invention include all compositions wherein the compounds of the present invention are contained in an amount that is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typically, the compounds may be administered to mammals, e.g., humans, orally at a dose of 0.0025 to 50 mg/kg of body weight, per day, or an equivalent amount of the pharmaceutically acceptable salt thereof, to a mammal being treated for apoptosis- mediated disorders. Preferably, about 0.01 to about 10 mg/kg of body weight is orally administered to treat or prevent such disorders. For intramuscular injection, the dose is generally about one-half of the oral dose. For example, a suitable intramuscular dose would be about 0.0025 to about 25 mg/kg of body weight, and most preferably, from about 0.01 to about 5 mg/kg of body weight. If a known cancer chemotherapeutic agent is also administered, it is administered in an amount that is effective to achieve its intended purpose. The amounts of such known cancer chemotherapeutic agents effective for cancer are well known to those of skill in the art.
The unit oral dose may comprise from about 0.01 to about 50 mg, preferably about 0.1 to about 10 mg of the compound of the invention. The unit dose may be administered one or more times daily as one or more tablets each containing from about 0.1 to about 10, conveniently about 0.25 to 50 mg of the compound or its solvates.
In a topical formulation, the compound may be present at a concentration of about 0.01 to 100 mg per gram of carrier.
In addition to administering the compound as a raw chemical, the compounds of the invention may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compounds into preparations which may be used pharmaceutically. Preferably, the preparations, particularly those preparations which may be administered orally and which may be used for the preferred type of administration, such as tablets, dragees, and capsules, and also preparations which may be administered rectally, such as suppositories, as well as suitable solutions for admimstration by injection or orally, contain from about 0.01 to 99 percent, preferably from about 0.25 to 75 percent of active compound(s), together with the excipient.
Also included within the scope of the present invention are the non-toxic pharmaceutically acceptable salts of the compounds of the present invention. Acid addition salts are formed by mixing a solution of the particular apoptosis inducers of the present invention with a solution of a pharmaceutically acceptable non-toxic acid such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid, oxalic acid, and the like. Basic salts are formed by mixing a solution of the particular apoptosis inducers of the present invention with a solution of a pharmaceutically acceptable non-toxic base such as sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, Tris, N-methyl-glucamine and the like.
The pharmaceutical compositions of the invention may be administered to any animal which may experience the beneficial effects of the compounds of the invention. Foremost among such animals are mammals, e.g., humans and veterinary animals, although the invention is not intended to be so limited.
The pharmaceutical compositions of the present invention may be administered by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
The pharmaceutical preparations of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, pharmaceutical preparations for oral use may be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above-mentioned starches and also carboxymefhyl-starch, cross- linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropymethyl- cellulose phthalate, are used. Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
Other pharmaceutical preparations which may be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules may contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added. Possible pharmaceutical preparations which may be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons. Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400 (the compounds are soluble in PEG-400) or cremophor, or cyclodextrins. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.
L accordance with one aspect of the present invention, compounds of the invention are employed in topical and parenteral formulations and are used for the treatment of skin cancer.
The topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments, gels and the like by choice of appropriate carriers. Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C12). The preferred carriers are those in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers may be employed in these topical formulations. Examples of such enhancers are found in U.S. Patent Nos. 3,989,816 and 4,444,762.
Creams are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil such as almond oil, is admixed. A typical example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil.
Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool. A typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight. The following examples are illustrative, but not limiting, of the method and compositions of the present invention. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.
Example 1 6"Chloro-N-(2-nitro-4-trifluoromethylphenyl)-3-pyridinecarboxamide
To a mixture of 6-chloronicotinoyl chloride (100 mg, 0.6 mmol) and 2-nitro-4- trifluoromethylaniline (117 mg, 0.6 mmol) in THF (10 ml) was added triethylamine (0.16 ml, 1.1 mmol). The mixture was stirred at room temperature overnight and then diluted with ethyl acetate. Water was added to dissolve the precipitate and the organic phase was separated and washed with water, then dried over anhydrous sodium sulfate. The product was purified by column chromatography to give the title compound as a pale yellow solid. 1H NMR (DMSO-d6): 11.27 (s, 1H), 8.97 (d, J = 2.7, 1H), 8.38-8.35 (m, 2H), 8.19 (d, J =
8.4, 1H), 7.96 (d, J = 8.7, 1H), 7.78 (d, J = 8.1, 1H).
Example 2 6-Chloro-N-(4-chloro-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-chloro-2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (DMSO-d6): 11.31 (s, 1H), 9.02 (d, J = 2.4, 1H), 8.96 (d, J = 9.3, 1H), 8.31 (d, J = 2.4, 1H), 8.24-8.20 (m, 1H), 7.73-7.69 (m, 1H), 7.54 (d, J = 8.4, 1H).
Example 3 6-Chloro-N-(4-fluoro-2-nitrophenyl)-3-pyrMinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-fluoro-2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (DMSO-d6): 10.97 (s, 1H), 8.94 (d, J = 2.7, 1H), 8.36-8.32 (m, 1H), 8.02-7.98 (m, 1H), 7.78-7.75 (m, 1H), 7.73-7.72 (m, 1H), 7.70 (d, J = 1.5, 1H).
Example 4 6-Chloro-N-(3-bromo-4-methoxy-6-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 3-bromo-4- methoxy-6-nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (DMSO-de): 10.84 (s, 1H), 8.93 (d, J = 2.4, 1H), 8.34-8.30 (m, 1H), 7.98 (s, 1H), 7.76 (d, J = 8.4, 1H), 7.70 (s, 1H), 3.97 (s, 3H).
Example 5 6-Chloro-N-(2,4-dimethoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2,4- dimethoxyaniline as described in Example 1. The reaction mixture was filtered to remove the precipitate and the filtrate was evaporated. The resulting solid was purified by CH2Cl2/hexane recrystallization to give a light purple solid in a yield of 59%. 1H NMR (DMSO-dβ): 9.77 (s, 1H), 8.93 (s, 1H), 8.35-8.31 (m, 1H), 7.68 (d, J = 7.8, 1H), 7.46 (d, J = 9.0, 1H), 6.67 (d, J = 2.4, 1H), 6.57-6.53 (m, 1H), 3.80 (s, 3H), 3.78 (s, 3H).
Example 6 2-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 2-chloronicotinoyl chloride and 4-methoxy-
2-nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDC13): 10.76 (s, 1H), 8.78 (d, J = 9.3, 1H), 8.58-8.55 (m, 1H), 8.13-8.09 (m, 1H), 7.73 (d, J = 3.0, 1H), 7.45-7.40 (m, 1H), 7.34-7.28 (m, 1H0, 3.90 (s, 3H). Example 7 6-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-methoxy- 2-nitroaniline and was obtained as a yellow solid as described in Example 1. XH NMR
(CDCI3): 11.13 (s, IH), 9.01 (d, J = 2.7, IH), 8.83 (d, J = 9.3, IH), 8.24-8.20 (m, IH), 7.76 (d, J = 2.7, IH), 7.52 (d, J = 8.7, IH), 7.35-7.31 (m, IH), 3.90 (s, 3H).
Example 8 6'Chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-ethoxy-2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR
(CDCI3): 11.13 (s, IH), 9.01 (d, J = 2.4, IH), 8.82 (d, J = 9.3, IH), 8.24-8.20 (m, IH), 7.74 (d, J = 3.0, IH), 7.51 (d, J = 7.8, IH), 7.34-7.30 (m, IH), 4.11 (q, J = 6.9, 2H), 1.47 (t, J =
6.9, 3H).
Example 9 6-Chloro-N-(4-methyl-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-methyl-2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDC13): 11.29 (s, IH), 9.02 (d, J = 2.7, IH), 8.81 (d, J = 8.7, IH), 8.24-8.21 (m, IH), 8.11 (s, IH), 7.58-7.50 (m, 2H), 2.44 (s, 3H).
Example 10 6-Chloro-N-(2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDCI3): 10.99 (s, IH), 8.95-8.94 (m, IH), 8.36-8.33 (m, IH), 8.02-7.99 (m, IH), 7.80- 7.69 (m, 3H), 7.48-7.42 (m, IH).
Example 11 6-Chloro-N-(4-methoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and ?-anisidine and was obtained as a white solid as described in Example 1. 1H NMR (CDC13): 8.85 (d, J = 2.1, IH), 8.19-8.15 (m, IH), 7.65 (s, IH), 7.54-7.46 (m, 3H), 6.96-6.91 (m, 2H), 3.83 (s, 3H).
Example 12 2,6-Dichloro-N-(4-methoxy'2-nitrophenyl)-4-pyridinecarboxamide
The title compound was prepared from 2,6-dichloropyridine-4-carbonyl chloride and 4-methoxy-2-nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDCI3): 11.12 (s, IH), 8.78 (d, J = 9.3, IH), 7.77 (d, J = 3.0, IH), 7.75 (s, 2H), 7.35-7.31 (m, IH), 3.91 (s, 3H).
Example 13 2-Chloro-4-t fluoromethyl-N-(4-methoxy-2-nitrophenyl)-5 yrimidinecarboxamide
The title compound was prepared from 2-chloro-4-(trifluoromethyl)pyrimidine-5- carbonyl chloride and 4-methoxy-2-nitroaniline and was obtained as a light yellow solid as described in Example 1. 1H NMR (CDCI3): 10.52 (s, IH), 9.04 (s, IH), 8.68 (d, J = 9.3, IH), 7.76 (d, J = 3.0, IH), 7.35-7.31 (m, IH), 3.91 (s, 3H). Example 14 N-(4-methoxy-2-nitrophenyl)-4-pyridinecarboxamide
The title compound was prepared from isonicotinoyl chloride and 4-methoxy-2- nitroaniline and was obtained as an orange yellow solid as described in Example 1. 1H
NMR (CDC13): 10.79 (s, IH), 8.83-8.81 (m, 2H), 7.85-7.83 (m, 2H), 7.61-7.55 (m, 2H), 7.40-7.36 (m, IH), 3.87 (s, 3H).
Example 15 2,6-Dichloro-N-(4-methoxy-2-nitrophenyl)'3-pyridinecarboxamide
To a solution of 2,6-dichloronicotinic acid (100 mg, 0.52 mmol) in CH2C12 (10 mL) was added cyanuric chloride (96 mg, 0.52 mmol). The mixture was stirred for half an hour, then 4-methoxy-2-nitroaniline (88 mg, 0.52 mmol) and triethylamine (0.1 mL, 0.72 mmol) were added. The resulting mixture was stirred at room temperature overnight and then diluted with ethyl acetate, washed with water. The organic phase was separated, dried over anhydrous Na2SO , concentrated and purified by chromatography to give the title compound (40 mg, 22%) as a yellow solid. 1H NMR (DMSO-cfc): 10.84 (bs, IH), 8.09 (d, J = 7.8, IH), 7.75 (d, J = 7.8, IH), 7.57-7.52 (m, 2H), 7.37-7.33 (m, IH), 3.86 (s, 3H).
Example 16 5,6-Diehloro-N-(4-methoxy-2-nitrophenyl)-3 yridinecarboxamide
The title compound was prepared from 5,6-dichloronicotinic acid and 4-methoxy- 2-nitroaniline and was obtained as a yellow solid as described in Example 15. 1H NMR
(DMSO-cfc): 10.85 (bs, IH), 8.88 (d, J = 2.4, IH), 8.59 (d, J = 2.4, IH), 7.58-7.54 (m, 2H), 7.39-7.35 (m, IH), 3.87 (s, 3H). Example 17 6-Chloro-N-(2,4-dinitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2,4- dinitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR
(CDC13): 11.69 (bs, IH), 9.28-9.23 (m, 2H), 9.06-9.05 (m, IH), 8.61-8.57 (m, IH), 8.28- 8.24 (m, IH), 7.59-7.56 (m, IH).
Example 18 4-Chloro-N-(4-ethoxy-2-nitrophenyl)-benzoylamide
The title compound was prepared from 4-chlorobenzoyl chloride and 4-ethoxy-2- nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDC13): 11.10 (bs, IH), 8.86 (d, J = 9.3, IH), 7.93 (d, J = 8.4, 2H), 7.73 (d, J = 3.0, IH), 7.51 (d, J = 8.4, 2H), 7.32-7.27 (m, IH), 4.11 (q, J = 6.9, 2H), 1.46 (t, J = 6.9, 3H).
Example 19 6-Chloro-N-(4,5-difluoro-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4,5- difluoro-2-nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDCI3): 11.49 (bs, IH), 9.04-8.97 (m, 2H), 8.25-8.19 (m, 2H), 7.56-7.53 (m, IH).
Example 20 6-Chloro-N-(4-methoxy-2-methylphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-methoxy- 2-methylaniline and was obtained as a white solid as described in Example 1. 1H NMR (CDCI3): 8.88 (bs, IH), 8.19 (d, J = 7.2, IH), 7.61-7.47 (m, 3H), 6.81-6.79 (m, 2H), 3.81 (s, 3H), 2.30 (s, 3H). Example 21 6-Chloro-N-(4-cyano~2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-amino-3- nitrobenzonitrile and was obtained as a light yellow solid as described in Example 1. 1H NMR (CDC13): 11.58 (s, IH), 9.18 (d, J = 8.7, IH), 9.04 (d, J = 2.4, IH), 8.65 (d, J = 2.1, IH), 8.26-8.22 (m, IH), 8.00-7.96 (m, IH), 7.56 (d, J = 9.0, IH).
Example 22
6-Fluoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
To the solution of KMnO4 (284 mg, 1.8 mmol) in H2O (50 mL) was added 2- fluoro-5-methylpyridine (100 mg, 0.9 mmol) at room temperature. The reaction mixture was then heated to 100°C for 4 h. The resulting precipitate was filtered and discarded.
The aqueous solution was washed with 3:2, hexane: ethyl acetate (2 x 20 mL), acidified with 2N HC1, then extracted with ethyl acetate (3 x 20 mL). The organic extracts were combined and washed with H2O and brine, dried over anhydrous Na2SO . The solvent was evaporated in vacuo to give 6-fluoronicotinic acid (30 mg) as a white solid. The title compound was prepared from 6-fluoronicotinic acid and 4-ethoxy-2-nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDC13): 11.09 (bs, IH), 8.88 (d, J = 2.4, IH), 8.81 (d, J = 9.3, IH), 8.42-8.35 (m, IH), 7.74 (d, J = 3.3, IH), 7.33-7.29 (m, IH), 7.13-7.10 (m, IH), 4.11 (q, J = 7.2, 2H), 1.47 (t, J = 7.2, 3H).
Example 23
6-Chloro-N-(2-nitro-4-trifluoromethoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-nitro-4- (trifluoromethoxy)aniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDCI3): 11.33 (bs, IH), 9.06 (d, J = 9.3, IH), 9.02 (d, J = 2.7, IH), 8.25-8.21 (m, IH), 8.19 (d, J = 2.7, IH), 7.65-7.61 (m, IH), 7.54 (d, J = 8.4, IH).
Example 24 6-Chloro-N-(4-benzoyl-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4-amino-3- nitrobenzophenone and was obtained as a yellow solid as described in Example 1. 1H
NMR (CDCI3): 11.61 (bs, IH), 9.12 (d, J = 8.7, IH), 9.06 (d, J = 2.4, IH), 8.79 (d, J = 1.8, IH), 8.28-8.25 (m, IH), 8.23-8.19 (m, IH), 7.82-7.81 (m, IH), 7.80-7.79 (m, IH), 7.67-
7.64 (m, IH), 7.58-7.53 (m, 3H).
Example 25 6-Chloro-N-(4-benzyloxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4- benzyloxy-2-nitroaniline and was obtained as a yellow solid as described in Example 1. 1H NMR (CDCI3): 11.13 (bs, IH), 9.00 (d, J = 3.0, IH), 8.83 (d, J = 9.3, IH), 8.23-8.20 (m, IH), 7.85 (d, J = 3.0, IH), 7.53-7.46 (m, IH), 7.44-7.37 (m, 6H), 5.15 (s, 2H).
Example 26 6-Methyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-methylnicotinic acid and 4-ethoxy-2- nitroaniline and was obtained as a yellow solid as described in Example 15. 1H NMR
(CDCI3): 11.09 (s, IH), 9.12 (d, J = 2.1, IH), 8.85 (d, J = 9.3, IH), 8.16-8.13 (m, IH), 7.73
(d, J = 3.0, IH), 7.34-7.29 (m, 2H), 4.11 (q, J = 6.9, 2H), 2.67 (s, 3H), 1.46 (t, J = 6.9, 3H). Example 27 6-Chloro-N-(4,5-dimethyl-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4,5- dimethyl-2-nitroaniline and was obtained as an orange solid as described in Example 1. 1H
NMR (CDC13): 11.38 (s, IH), 9.03 (d, J = 2.1, IH), 8.72 (s, IH), 8.24-8.21 (m, IH), 8.07 (s, IH), 7.52 (d, J = 8.7, IH), 2.41 (s, 3H), 2.34 (s, 3H).
Example 28 6-Chloro-N-(4-methoxy-2-nitrophenyl)-l-N-oxide-3-pyridinecarboxamide
To a stirred solution of 30% H2O2 (2 mL) was added trifluoroacetic anhydride (1 mL) dropwise at 0°C. The resulting solution was stirred at 0°C for 30 min, then 6-chloro- N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide (30 mg, 0.0975 mmol) was added. The mixture was stirred at 90°C for 30 min. The resulting yellow solution was cooled in an ice bath for 1 h. The precipitate was filtered, washed with H2O and MeOH, and dried to give 15 mg (0.0463 mmol, 47%) of the title compound as a yellow powder. 1H NMR (CDCI3): 11.11 (s, IH), 8.95-8.76 (m, IH), 8.78 (d, J = 9.3. IH), 7.76 (d, J = 3.0, IH), 7.66-7.65 (m, 2H), 7.35-7.31 (m, IH), 3.91 (s, 3H).
Example 29 N-(4-Methoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from nicotinoyl chloride and 4-methoxy-2- nitroaniline and was obtained as an orange solid as described in Example 1. 1H NMR
(DMSO-d6): 10.74 (s, IH), 9.10 (d, J = 2.4, IH), 8.80-8.78 (m, IH), 7.62-7.54 (m, 3H), 7.39-7.35 (m, IH), 3.87 (s, 3H). Example 30 6-Chloro-N-(4-ethoxy-2-nitrophenyl)-N-methyl-3-pyridinecarboxamide
To a solution of 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide (50 mg, 0.155 mmol) in DMF (2 ml) was added sodium methoxide (12 mg, 0.222 mmol) in
DMF (1 ml) and the reaction mixture was stirred for 10 min. To the mixture was added iodomethane (22 mg, 0.155 mmol) slowly and the mixture was stirred for at room temperature for 3 h. It was diluted with ethyl acetate and washed with water and brine, dried over anhydrous sodium sulfate, and evaporated. The residue was purified by column chromatography to yield the titled compound as a light brown liquid. 1H NMR (CDC13):
8.17 (d, J = 2.1, IH), 7.68-7.65 (m, IH), 7.35 (d, J = 2.7, IH), 7.21 (t, J = 7.8, 2H), 7.10- 7.06 (m, IH), 4.08-4.01 (m, 2H), 3.40 (s, 3H), 1.44 (t, J = 6.9, 3H).
Example 31 2,5-Dimethyl-N-(4-ethoxy-2-nitrophenyl)-3-furancarboxamide
The title compound was prepared from 2,5-dimethyl-3-furoic acid and 4-ethoxy-2- nitroaniline as an orange solid as described in Example 15. 1H NMR (CDCI3): 10.59 (s, IH), 8.83 (d, J = 9.0, IH), 7.70 (d, J = 3.0, IH), 7.27-7.25 (m, IH), 6.24 (s, IH), 4.08 (q, J = 6.9, 2H), 2.62 (s, 3H), 2.30 (s, 3H), 1.45 (t, J = 6.9, 3H).
Example 32 6-Chloro-N-(5-bromo-3-nitro-2-pyridyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino-5- bromo-3-nitropyridine as a light yellow solid as described in Example 1. 1H NMR (CDCI3): 10.56 (s, IH), 9.00 (d, J = 2.7, IH), 8.82 (d, J = 2.1, IH), 8.69 (d, J = 2.1, IH), 8.27-8.23 (m, IH), 7.53 (d, J = 8.1, IH). Example 33 N-(4-Ethoxy-2-nitrophenyl)-2-pyrazinecarboxamide
The title compound was prepared from 2-pyrazinecarboxylic acid and 4-ethoxy-2- nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDCI3): 12.34 (s,
IH), 9.50 (d, J = 1.2, IH), 8.91 (d, J = 9.0, IH), 8.84 (d, J = 2.4, IH), 8.72-8.70 (m, IH), 7.75 (d, J = 3.0, IH), 7.33-7.28 (m, IH), 4.12 (q, J = 6.9, 2H), 1.47 (t, J = 6.9, 3H).
Example 34 5-Methyl-N-(4-ethoxy-2-nitrophenyl)"2-pyrazinecarboxamuie
The title compound was prepared from 2-methylpyrazine-5-carboxylic acid and 4- ethoxy-2-nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDC13): 12.82 (s, IH), 9.36 (s, IH), 8.90 (d, J = 9.6, IH), 8.56 (d, J = 1.0. IH), 7.74 (d, J = 2.7, IH), 7.32-7.29 (m, IH), 4.11 (q, J = 6.9, 2H), 2.71 (s, 3H), 1.46 (t, J = 6.9, 3H).
Example 35 6-Chloro-N-(4-chloro-2-cyanophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino-5- chlorobenzonitrile as a white solid as described in Example 1. 1H NMR (CDCI3): 8.96 (d, J = 2.7, IH), 8.53-8.50 (M, IH), 8.24 (s, IH), 8.19-8.15 (m, IH), 7.67-7.63 (m, 2H), 7.54- 7.51 (m, IH).
Example 36
6-Chloro-N-(4-chloro-2-amidophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino-5- chlorobenzamide as a white solid as described in Example 1..1H NMR (DMSO-dg): 8.91 (d, J = 2.7, IH), 8.62 (d, J = 9.3, IH), 8.55 (s, IH), 8.30-8.27 (m, IH), 8.04 (s, IH), 8.01 (d, J = 2.7, IH), 7.78 (d, J = 8.4, IH), 7.69-7.66 (m, IH).
Example 37 6-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyrMinecarboxamide
The title compound was prepared from 6-trifluoromethylnicotinic acid and 4- ethoxy-2-nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDC13):
11.23 (s, IH), 9.32 (d, J = 3.6, IH), 8.83 (d, J = 9.3, IH), 8.47-8.43 (m, IH), 7.88 (d, J = 8.1, IH), 7.76 (d, J = 3.0, IH), 7.35-7.31 (m, IH), 4.12 (q, J = 6.9, 2H), 1.47 (t, J = 6.9,
3H).
Example 38 6-Chloro-N-(4-chloro-2-methoxycarbϋnylphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and methyl 2- amino-5-chlorobenzoate as a white solid as described in Example 1. 1H NMR (CDCI3): 12.13 (s, IH), 9.06 (d, J = 3.0, IH), 8.86 (d, J = 9.3, IH), 8.29-8.25 (m, IH), 8.08 (d, J = 2.4, IH), 7.60-7.57 (m, IH), 7.50 (d, J = 8.4, IH), 3.99 (s, 3H).
Example 39 5-Bromo-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 5-bromonicotinic acid and 4-ethoxy-2- nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDCI3): 11.11 (s,
IH), 9.11 (d, J = 2.1, IH), 8.88 (d, J = 1.8, IH), 8.80 (d, J = 9.3, IH), 8.42 (t, J = 2.1, IH), 7.75 (d, J = 2.7, IH), 7.34-7.30 (m, IH), 4.12 (q, J = 6.9, 2H), 1.47 (t, J = 6.9, 3H). Example 40 N-(4-Ethoxy-2-nitrophenyl)-2-pyridinecarboxamide N-oxide
The title compound was prepared from picolinic acid N-oxide and 4-ethoxy-2- nitroaniline as a yellow solid as described in Example 15. 1H NMR (DMSO-ck): 8.57-8.54
(m, IH), 8.40 (d, J = 9.0, IH), 8.37-8.33 (m, IH), 7.76-7.75 (m, 2H), 7.61 (d, J = 3.0, IH), 7.43-7.39 (m, IH), 4.14 (q, J = 6.9, 2H), 1.36 (t, J = 6.9, 3H).
Example 41 4-Amino-3-nitrophenyl 6-Chloro-3-pyridinecarboxylate (A) and 6-Chloro-N-(4- hydroxy-2-nitrophenyl)-3-pyridinecarboxamide (B)
A mixture of 4-amino-3-nitrophenol (200 mg, 1.30 mmol), 6-chloronicotinoyl chloride (229 mg, 1.30 mmol) and N,N-diisopropylethylamine (200 ul) in THF (5 ml) was stirred overnight at room temperature. The resulting solid was collected by filtration, wash with hexane:ethyl acetate (1:1) and dried under vacuo to give compound A as an orange solid (14 mg). 1H NMR (DMSO-d6): 9.08 (dd, J = 8.0, 2.6 Hz, IH), 8.47 (dd, J = 2.6, 8.3 Hz, IH), 7.97 (d, J = 2.7 Hz, IH), 7.78 (dd, J = 0.8, 8.3 Hz, IH), 7.55 (s, 2H), 7.45 (dd, J = 2.7, 9 Hz, IH), 7.10 (d, J = 9.0 Hz, IH). The filtrate was diluted with ethyl acetate (50 ml), washed with water (25 ml), aqueous saturated NaCl (25 ml) and dried over anhydrous sodium sulfate. The solution was concentrated in vacuo and the product was purified by column chromatogarphy using hexane:ethyl acetate (3:1) to give the product B as a dark yellow solid (9 mg). 1H NMR (DMSO-d6): 9.13 (d, J = 2.4 Hz, IH), 8.96 (d, J = 2.4 Hz, IH), 8.52 (dd, J = 1.8, 8.7 Hz, IH), 8.36 (dd, J = 2.4, 8.4 Hz, IH), 8.07 (s, IH), 7.85-7.80 (m, 2H), 7.73 (d, J = 8.4 Hz, IH). Example 42 N-(4-Ethoxy-2-nitrophenyl)-3-pyrrolecarboxamide
The title compound was prepared from pyrrole-3-carboxylic acid and 4-ethoxy-2- nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDCI3): 10.82 (s,
IH), 8.88 (d, J = 9.3, IH), 8.65 (s, IH), 7.70 (d, J = 3.0, IH), 7.54-7.52 (m, IH), 7.28-7.24 (m, IH), 6.87-6.85 (m, IH), 6.71-6.69 (m, IH), 4.08 (q, J = 6.9, 2H), 1.45 (t, J = 6.9, 3H).
Example 43 6-Chloro-N-(2-nitro-5-imidazolyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinonyl chloride and 5-amino- 4-nitroimidazole as a brown solid as described in Example 1. 1H NMR (DMSO-ck): 8.97- 8.96 (m, IH), 8.39-8.36 (m, IH), 7.79-7.76 (m, IH), 7.68 (s, IH), 7.37 (s, IH).
Example 44 6-Chloro-N-(4-t-butyl-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinic chloride and 4-(t-butyl)- 2-nitroaniline as a yellow solid as described in Example 1. 1H NMR (CDC13): 11.30 (s,
IH), 9.03 (d, J = 2.1, IH), 8.83 (d, J = 9.0, IH), 8.29 (d, J = 2.7, IH), 8.25-8.21 (m, IH), 7.80-7.76 (m, IH), 7.52 (d, J = 8.4, IH), 1.38 (s, 9H).
Example 45 N-(4-Ethoxy-2-nitrophenyl)-5-pyrimidinecarboxamide
The title compound was prepared from pyrimidine-5-carboxylic acid and 4-ethoxy- 2-nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDC13): 11.16 (s, IH), 9.43 (s, IH), 9.32 (s, 2H), 8.81 (d, J = 9.3, IH), 7.76 (d, J = 3.0, IH), 7.34-7.27 (m, IH), 4.12 (q, J = 6.9, 2H), 1.47 (t, J = 6.9, 3H). Example 46 N-(4-Ethoxy-2-nitrophenyl)-2-pyridinecarboxamide
The title compound was prepared from picolinic acid and 4-ethoxy-2-nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDC13): 12.54 (s, IH), 8.92 (d, J = 9.3, IH), 8.73 (d, J = 3.9, IH), 8.28 (d, J = 6.9, IH), 7.95-7.90 (m, IH), 7.74 (d, J = 3.0, IH), 7.54-7.50 (m, IH), 7.31-7.27 (m, IH), 4.11 (q, J = 6.9, 2H), 1.46 (t, J = 6.9, 3H).
Example 47
6-Chloro-N-(2-cyano-4,5-dimethoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino- 4,5-dimethoxybenzonitrile as a white solid as described in Example 1. XH NMR (CDCI3): 8.97 (d, J = 2.7, IH), 8.18-8.15 (m, 3H), 7.52 (d, J = 8.4, IH), 7.02 (s, IH), 4.01 (s, 3H),
3.92 (s, 3H).
Example 48
6-Chloro-N-(2-methoxycarbonyl-4,5-dimethoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino- 4,5-dimethoxybenzonic acid methyl ester as a light yellow solid as described in Example 1. 1H NMR (CDCI3): 12.34 (s, IH), 9.09 (d, J = 2.4, IH), 8.62 (s, IH), 8.29-8.26 (m, IH), 7.52 (s, IH), 7.50 (d, J = 8.4, IH), 4.03 (s, 3H), 3.96 (s, 3H), 3.93 (s, 3H).
Example 49 6-Chloro-N-(2-methylcarbonyl-4,5-methylenedioxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-amino- 4,5-methylenedioxyacetophenone as a light grey-yellow solid as described in Example 1. 1H NMR (CDCI3): 9.09-9.08 (m, IH), 8.55 (s, IH), 8.30-8.27 (m, IH), 7.50-7.47 (m, IH), 7.35 (s, IH), 6.10 (s, 2H), 2.64 (s, 3H).
Example 50 6-(2,2,2-Triflouroethoxy)-N-(4-ethoxy-2-nitrophenyl)-3-pyridinec rboxamide
The title compound was prepared from 6-(2,2,2-trifluoroethoxy)pyridine-3- carboxylic acid and 4-ethoxy-2-nitroaniline as a yellow solid as described in Example 15.
1H NMR (CDCI3): 11.05 (s, IH), 8.84-8.80 (m, 2H), 8.25-8.21 (m, IH), 7.73 (d, J = 3.0, IH), 7.32-7.28 (m, IH), 7.01 (d, J = 8.7, IH), 4.86 (q, J = 8.4, 2H), 4.11 (q, J = 6.9, 2H),
1.46 (t, J = 6.9, 3H).
Example 51 6-Chloro-N-(2,3-dinitro-4-methoxyphenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2,3-dinitro- 4-methoxyaniline as a white solid as described in Example 1. 1H NMR (CDC13): 9.99 (s, IH), 8.96 (d, J = 2.4, IH), 8.77 (d, J = 9.3, IH), 8.18-8.14 (m, IH), 7.53 (d, J = 9.0, IH), 7.45 (d, J = 9.3, IH), 4.02 (s, 3H).
Example 52 6-Chloro-N-(2-nitro-4,6-dimethylphenyl)-3φyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 2-nitro-4,6- dimethylaniline as a light yellow solid as described in Example 1. 1H NMR (CDC13): 9.09
(s, IH), 8.97 (d, J = 2.4, IH), 8.22-8.18 (m, IH), 7.60 (d, J = 1.2, IH), 7.50 (d, J = 8.1, IH), 7.42 (s, IH), 2.43 (s, 3H), 2.35 (s, 3H). Example 53 4-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-benzoylamide
The title compound was prepared from 4-chloromethylbenzoyl chloride and 4- ethoxy-2-nitroaniline as a yellow solid as described in Example 1. 1H NMR (CDCI3):
11.11 (s, IH), 8.87 (d, J = 9.3, IH), 7.98 (d, J = 8.4, 2H), 7.73 (d, J = 3.0, IH), 7.56 (d, J = 8.4, 2H), 7.32-7.28 (m, IH), 4.65 (s, 2H), 4.11 (q, J = 6.9, 2H), 1.46 (t, J = 6.9, 3H).
Example 54 6-Chloro-N-(4,5-dimethoxy-2-nitrophenyl)~3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 4,5- dimethoxy-2-nitroaniline as a yellow solid as described in Example 1. 1H NMR (CDCI3): 11.84 (s, IH), 9.04 (d, J = 1.8, IH), 8.66 (s, IH), 8.25-8.21 (m, IH), 7.77 (s, IH), 7.53 (d, J = 8.7, IH), 4.07 (s, 3H), 3.97 (s, 3H).
Example 55 6-Chloro-N-(2-methoxy-5-pyridyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 5-amino-2- methoxypyridine as a pink solid as described in Example 1. 1H NMR (CDCI3): 8.87 (d, J = 2.1, IH), 8.28 (d, J = 3.0, IH), 8.20-8.17 (m, IH), 7.99-7.96 (m, IH), 7.67 (s, IH), 7.49 (d, J = 8.7, IH), 6.81 (d, J = 8.7, IH), 3.95 (s, 3H).
Example 56
6-Chloro-N-(2-chloro-5-pyridyl)-3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 5-amino-2- chloropyridine as a white solid as described in Example 1. 1H NMR (CDCI3): 8.89-8.84 (m, IH), 8.52 (d, J = 2.1, IH), 8.27-8.23 (m, IH), 8.21-8.17 (m, IH), 7.83 (s, IH), 7.53- 7.50 (m, IH), 7.39 (d, J = 8.7, IH).
Example 57 6-Chloro-N-(4,6-dichloro-5-pyrimidyl)'3-pyridinecarboxamide
The title compound was prepared from 6-chloronicotinoyl chloride and 5-amino- 4,6-dichloropyrimidine as a white solid as described in Example 1. 1H NMR (CDCI3): 8.95 (d, J = 2.4, IH), 8.78 (s, IH), 8.25-8.22 (m, IH), 7.57 (s, IH), 7.54 (d, J = 8.4, IH).
Example 58 6-Cyano-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 6-cyanonicotinic acid and 4-ethoxy-2- nitroaniline as a yellow solid as described in Example 15. 1H NMR (CDCI3): 11.22 (s,
IH), 9.31-9.30 (m, IH), 8.81 (d, J = 9.3, IH), 8.42-8.38 (m, IH), 7.90-7.87 (m, IH), 7.76 (d, J = 3.0, IH), 7.34-7.30 (m, IH), 4.12 (q, J = 6.9, 2H), 1.47 (t, J = 6.9, 3H).
Example 59 2-Methyl-N-(4-ethoxy-2-nitrophenyl)-3-furancarboxamide
The title compound was prepared from 2-methyl-3-furoic acid and 4-ethoxy-2- nitroaniline as an orange yellow solid as described in Example 15. 1H NMR (CDC13): 10.66 (s, IH), 8.82 (d, J = 9.3, IH), 7.70 (d, J = 3.0, IH), 7.34 (d, J = 1.8, IH), 7.28-7.24 (m, IH), 6.68 (d, J = 1.8, IH), 4.09 (q, J = 6.9, 2H), 2.67 (s, 3H), 1.45 (t, J = 6.9, 3H). Example 60 6-Chloro-N-(4-chloro-2-trifluoromethylphenyl)-3-pyridinecarboxamide ,
The title compound was prepared from 6-chloronicotinoyl chloride and 4-chloro-2- trifluoromethylaniline as a white solid as described in Example 1. 1H NMR (CDCI3): 8.87
(d, J = 2.7, IH), 8.32 (d, J = 8.7, IH), 8.16-8.12 (m, IH), 8.06 (bs, IH), 7.82 (d, J = 2.4, IH), 7.53-7.50 (m, IH), 7.12 (d, J = 8.7, IH).
Example 61 4-Chloro-2-nitro-N-(6-chloro-3-pyridyl)-benzoylamide
The title compound was prepared from 4-chloro-2-nitrobenzoic acid and 5-amino- 2-chloropyridine as a white solid as described in Example 15.
Example 62
6-Chloro-N-(2-pyrazinyl)-3-pyridinecarboxamide
To a mixture of 6-chloronicotinoyl chloride (160 mg, 0.91 mmol), aminopyrazine (87 mg, 0.91 mmol) and THF (10 ml) was added triethylamine (0.25 ml, 1.82 mmol) and the mixture was stirred overnight at room temperature. The mixture was diluted with ethyl acetate (100 ml), wash with water (50 ml), saturated aqueous NaCl (50 ml), dried over anhydrous sodium sulfate and evaporated. The crude product was purified by column chromatography using hexane:ethyl acetate (3:2) to give 8 mg (4% yield) of the title compound as yellow solid. 1H NMR (CDC13): 9.68 (d, J = 1.2 Hz, IH), 8.95 (d, J = 2.7 Hz, IH), 8.45 (d, J = 2.7 Hz, IH), 8.4 (bs, IH), 8.33- 8.32 (m, IH), 8.23 (dd, J = 2.4, 8.1
Hz, IH), 7.53 (dd, J = 0.6, 8.4 Hz, IH). Example 63 4-Chloro-2-nitro-N-(6-methoxy-3-pyridyl)-benzoylamide
A mixture of cyanuric chloride (183 mg, 0.99 mmol) and 4-chloro-2-nitrobenzoic acid (200 mg, 0.99 mmol) in THE (5 ml) was stirred at room temperature for 30 minutes.
To the mixture was added a solution of 5-amino-2-methoxy-pyridine (123 mg, 0.99 mmol) in THF (5 ml), followed by triethylamine (0.28 ml, 1.98 mmol). The mixture was stirred overnight at room temperature. It was diluted with 100 ml of hexane:ethyl acetate (1:1), wash with 2N NaOH (50 ml), water (50 ml), followed by saturated aqueous NaCl (50 ml), dried over anhydrous sodium sulfate and evaporated. The crude product was purified by column chromatography using hexane:ethyl acetate (3:1) to give 16 mg (5% yield ) of the title compound as a pink solid. 1H NMR (Acetone-d6): 9.94 (s, IH), 8.16 (d, J = 1.8 Hz, IH), 8.07-7.86 (m, 4H), 6.81 (d, J= 9.3 Hz, IH), 3.89 (s, 3H).
Example 64
6-Chloro-N-(2-cyano-6-chloromethyl-4-oxide-3-pyrazinyl)-3-pyridinecarboxamide
The title compound was prepared from 3-amino-6-(chloromethyl)-2-pyrazine carbonitrile 4-oxide (203 mg, 1.1 mmol) and 6-chloronicotinoyl chloride (200 mg, 1.1 mmol) and was obtained as an yellow solid as described in Example 62. 1H NMR
(Acetone-d6): 9.00 (s, IH), 8.66 (s, IH), 8.42 (dd, J = 8.1, 2.1 Hz, IH), 7.65 (d, J = 8.1 Hz,
IH), 7.52 (s, IH), 5.38 (s, 2H).
Example 65 6-Chlorθ'N-(2-cyano-4-methylphenyl)-3-pyridinecarboxamide
A mixture of 2-amino-5-methyl-benzonitrile (80 mg, 0.61 mmol) and 6- chloronicotinoyl chloride (162 mg, 0.92 mmol) in anhydrous pyridine (5 ml) was refluxed for 4 hours. The mixture was diluted with 50 ml of hexane:ethyl acetate (1:1), wash with 2N HCl (25 ml), water (25 ml), followed by saturated aqueous NaCl (25 ml), dried over anhydrous sodium sulfate, and evaporated. The crude product was purified by column chromatography using hexane:ethyl acetate (2:1) to give 30 mg (18% yield) of the title compound as pale pink solid. 1H NMR (DMSO-d6): 10.82 (s, IH), 8.99 (d, J = 2.4 Hz, IH), 8.38 (dd, J = 2.4, 8.1 Hz, IH), 7.78-7.73 (m, 2H), 7.61-7.48 (m, 2H), 2.38 (s, 3H).
Example 66
6-Chloro-N-(4-chloro-6-methyl-2-nitrophenyl)-3-pyridinecarboxamide
The title compound was prepared from 4-chloro-2-methyl-6-nitroaniline (204 mg,
1.09 mmol) and 6-chloronicotinoyl chloride (192 mg, 1.09 mmol) and obtained as a light yellow solid as described in Example 62. 1H NMR (CDC13): 9.13 (s, IH), 8.96 (dd, J =
0.6, 2.4 Hz, IH), 8.20 (dd, J = 2.7, 8.4 Hz, IH), 7.93 (d, J = 2.4 Hz, IH), 7.59 (d, J = 2.4
Hz, IH), 7.51 (dd, 7= 0.6, 8.1 Hz, IH), 2.38 (s, 3H).
Example 67 4-Trifluoromethyl-N'(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
A mixture of 4-(trifluoromethyl)pyridine-3-carboxylic acid (100 mg, 0.52 mmol), 4-ethoxy-2-nitroaniline (94.7 mg, 0.52 mmol), and l-(3-dimethylamino-propyl)-3- ethylcarbodiimide hydrochloride (199.3 mg, 1.04 mmol) in THF (5 ml) was refluxed overnight. The mixture was cooled to room temperature, diluted with 100 ml of hexane:ethyl acetate (1:1), wash with 2N NaOH (50 ml), water (50 ml), saturated aqueous NaCl (50 ml), dried over anhydrous sodium sulfate and evaporated. The crude product was purified by column chromatography using hexane:ethyl acetate (2:1) to give 17.3 mg (9.4% yield) of the title compound as yellow solid. 1H NMR (CDC13): 10.48 (s, IH), 9.01- 8.96 (m, 2H), 8.74 (d, J = 9.3 Hz, IH), 7.74-7.69 (m, 2H), 7.32 (dd, J = 3.0, 9.3 Hz, IH),
4.13 (q, J = 6.9 Hz, 2H), 1.48 (t, J = 6.9 Hz, 3H). Example 68 4-Bromomethyl-3-nitro-N-(6-chloro-3-pyridyl)-benzoylamide
A mixture of 4-bromomethyl-3-nitrobenzoic acid (169 mg, 0.65 mmol), 5-amino- 2-chloropyridine (83 mg, 0.65 mmol) and l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (210 mg) in THF (10 ml) was refluxed for 24 h. The mixture was cooled to room temperature and diluted with hexane:ethyl acetate (1:1, 100 ml), washed with water (50 ml), 2N HCl (50 ml), brine (50 ml) and dried over anhydrous sodium sulfate. The solution was concentrated in vacuo to give a yellow solid which was washed with dichloromethane and dried to give 68 mg (28%) of the title compound. 1H NMR (DMSO- d6): 10.91 (s, IH), 8.77 (d, J = 2.1 Hz, IH), 8.63 (s, IH), 8.31 (d, J = 8.1 Hz, IH), 8.24 (dd, J = 2.4, 8.7 Hz, IH ), 7.96 (d, J = 8.1 Hz, IH), 7.56 (d, J = 8.7 Hz, IH), 5.11 (s, 2H).
Example 69 6-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide
A) Methyl 6-chloromethylnicotinate: A mixture of methyl 6-methylnicotinate (2.3 g, 15.2 mmol) and N-chlorosuccinimide (4.6 g, 34 mmol) in chloroform was refluxed for 24 h to give 1.3 g of the title compound as a white solid. B) 6-Chloromeihylnicotinic acid: A mixture of methyl 6- chloromethylnicotinate (1.3 g, 6.5 mmol) in 2N HCl (40 ml) was refluxed for 5 h. The mixture was cooled to room temperature to give 1.1 g of the title compound as a tan solid.
C) 6-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide: A mixture of 6-chloromethylnicotinic acid (155 mg, 0.69 mmol), 4-ethoxy-2-nitroaniline (146 mg), l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (238 mg) and pyridine in THF (10 ml) was refluxed for 17 h. The mixture was cooled to room temperature, diluted with hexane:ethyl acetate (1:1, 100 ml), washed with water (50 ml), 2N HCl (50 ml), brine (50 ml) and dried over anhydrous sodium sulfate. The solution was concentrated in vacuo and the product was purified by column chromatography using hexane:ethyl acetate (3:1) to give the title compound as a yellow solid (30 mg). 1H NMR
(CDC13): 11.14 (s, IH), 9.18 (m, IH), 8.84 (d, J = 9.3 Hz, IH), 8.30 (dd, J = 2.4, 8.1 Hz, 1H), 7.75 (d, J = 3.0 Hz, IH), 7.68 (d, J = 8.4 Hz, IH), 7.31 (dd, J = 3.0, 9.3 Hz, IH), 4.76 (s, 2H), 4.12 (m, 2H), 1.47 (t, J = 7.1 Hz, 3H).
Example 70 N-(4-Ethoxy-2-nitrophenyl)-3-furancarboxamide
The title compound was prepared from 3-furoic acid and 4-ethoxy-2-nitroaniline as a yellow solid as describedln Example 15. 1H NMR (DMSO-cfe): 10.25 (s, IH), 8.36 (d, J=2.4, IH), 7.83-7.81 (m, IH), 7.53-7.47 (m, 2H), 7.35-7.31 (m, IH), 6.94 (d, J=2.4, IH), 4.12 (q, J=6.9, 2H), 1.35 (t, J=6.9, 3H).
Example 71
Identification of N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide and Analogs as Caspase Cascade Activators and Inducers of Apoptosis in Solid Tumor Cells
Human breast cancer cell lines T47D and ZR75-1 were grown according to media component mixtures designated by American Type Culture Collection + 10% FCS (Jnvitrogen Corporation, Life Technologies Division), in a 5% CO2 -95% humidity incubator at 37 °C. T47D and ZR75-1 cells were maintained at a cell density between 30 and 80% confluency at a cell density of 0.1 to 0.6 x 106 cells/ml. Cells were harvested at
600xg and resuspended at 0.65 x 106 cells/ml into appropriate media + 10% FCS. An aliquot of 45 μl of cells was added to a well of a 96-well microtiter plate containing 5 μl of a 10% DMSO in RPMI-1640 media solution containing 1.6 to 100 μM of N-(4-methoxy- 2-nitrophenyl)-3-pyridinecarboxamide or other test compound (0.16 to 10 μM final). An aliquot of 45 μl of cells was added to a well of a 96-well microtiter plate containing 5 μl of a 10% DMSO in RPMI-1640 media solution without test compound as the control sample. The samples were mixed by agitation and then incubated at 37 C for 24 h in a 5% CO2- 95% humidity incubator. After incubation, the samples were removed from the incubator and 50 μl of a solution containing 20 μM of N-(Ac-DEND)-N'-ethoxycarbonyl-R110 (SEQ JD NO: 1) fluorogenic substrate (Cytovia, Inc.; WO99/18856), 20% sucrose (Sigma),
20 mM DTT (Sigma), 200 mM NaCl (Sigma), 40 mM Na PIPES buffer pH 7.2 (Sigma), and 500 μg/ml lysolecithin (Calbiochem) was added. The samples were mixed by agitation and incubated at room temperature. Using a fluorescent plate reader (Model 1420 Wallac Instruments), an initial reading (T = 0) was made approximately 1- 2 min after addition of the substrate solution, employing excitation at 485 nm and emission at 530 nm, to determine the background fluorescence of the control sample. After the 3 h incubation, the samples were read for fluorescence as above (T = 3 h).
Calculation:
The Relative Fluorescence Unit values (RFU) were used to calculate the sample readings as follows: RFU (τ=3h) - Control RFU (τ=0) = Net RFU(T=3h) .
The activity of caspase cascade activation was determined by the ratio of the net RFU value for N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide or other test compound to that of control samples. The EC50 (nM) was determined by a sigmoidal dose- response calculation (Prism 2.0, GraphPad Software Inc.). The caspase activity (Ratio) and potency (EC50) are summarized in Table I:
Table I. Caspase Activity and Potency
Thus, N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide (Example 29) and analogs are identified as potent caspase cascade activators and inducer of apoptosis in solid tumor cells.
Example 72
N-(4-Methoxy-2-nitrophenyl)-3-pyridinecarboxamide Induces Nuclear Fragmentation in Jurkat Cells
The ability of N-(4-memoxy-2-nitrophenyl)-3-pyridinecarboxamide to induce nuclear fragmentation was tested by treatment of Jurkat cells with the test compound followed by staining of the nucleus with Sytol6, a fluorescent DNA dye (Molecular Probes, Eugene, OR). The nuclei of Jurkat cells treated with vehicle control (DMSO) are seen to be round with dispersed chromatin that is moderately stained with Sytol6 (Fig. 1A). In contrast, Jurkat cells treated with 5 μM of N-(4-methoxy-2-nitrophenyl)-3- pyridinecarboxamide for 24 h have shrunken and fragmented nuclei (Fig. IB), which is a hallmark of caspase-mediated apoptosis. These results corroborate the caspase induction assays by showing that N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide can induce a key cellular marker of apoptosis. Example 73
N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide Induces Mitotic Arrest in Jurkat
Cells
Jurkat cells were incubated with a range of concentrations of N-(4-methoxy-2- nitrophenyl)-3-pyridinecarboxamide (0.02 μM to 5 μM) for 6 h under normal growth conditions; control cultures were treated with DMSO vehicle. The cells were then treated for 20 min. with 800 nM Syto 16 (Molecular Probes, Eugene, OR). Cytospin preparations were then prepared and the samples were viewed by fluorescent microscopy using a fluorescein filter set. For each concentration of test compound, the number of mitotic figures were counted and expressed as a percentage of the total number of cells. Three fields from each condition were evaluated and the mean and SEM were calculated and plotted as a function of drug concentration (Fig. 2). The results show that N-(4-methoxy- 2-nitrophenyl)-3-pyridinecarboxamide is an effective inducer of mitotic arrest in Jurkat cells up to a concentration of 5 μM.
Example 74
Treatment with N-(4'Methoxy-2-nitrophenyl)-3-pyridinecarboxamide or 6-Chloro-N-(4- ethoxy-2-nitrophenyl)-3-pyridinecarboxamide Leads to Cell Cycle Arrest and Apoptosis in Solid Tumor Cell Line
T47D, a breast cancer cell line, was maintained and harvested as described in Example 71. lxlO6 cells were treated with 10 μM of N-(4-methoxy-2-nitrophenyl)-3- pyridinecarboxamide or 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide for 48 h at 37 °C. As a control, cells were also incubated with equivalent amount of solvent as control (DMSO). Cells were harvested at 1,200 rpm and washed twice with 5 mM EDT A/PBS. Cells were then resuspended "in 300 μl EDT A/PBS and 700 μl of 100% ethanol, vortexed and incubated at room temperature for 1 h. Samples were spun down at 1,200 rpm for 5 min and the supernatant was removed. A solution containing 100 μg/ml of propidium iodide and 1 mg/ml of RNAse A (fresh) was added to the samples and incubated for 1 h at room temperature. Samples were then transferred to 12x75 mm polystyrene tubes and analyzed on a flow cytometer. All flow cytometry analyses were performed on FACScalibur (Becton Dickinson) using Cell Quest analysis software. On the x-axis is plotted the fluorescence intensity and on the y-axis is plotted the number of cells with that fluorescence intensity. The T47D control cell population profile is seen in Fig. 3A and the increase in G2/M DNA content (M4) cells that is seen when treated with N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide at 10 μM is seen in Fig. 3B. An increase in the sub-diploid DNA content of cells (marker Ml region, Fig 3) is also seen to increase from 2% to 25% with compound treatment. The sub-diploid amount of DNA (Ml) is indicative of apoptotic cells which have undergone DNA degradation or fragmentation. In cells treated with 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3- pyridinecarbox-amide, a more potent analog, at 1 μM (Fig. 4B), the accumulation of T47D cells in the G2 M phase (M4) is similarly seen. In addition, the sub-Gl population of cells with reduced DNA content (Ml) indicative of apoptosis is seen to increase substantially over control cells (Fig. 4A), indicating cell cycle arrest and induction of apoptosis by treatment with N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide and its more potent analog 6-chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide.
Example 75
6-Chloro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide Inhibits the Proliferation of Solid Tumor HeLa Cell Line
HeLa cells were grown according to media component mixtures designated by American Type Culture Collection + 10% FCS (Invitrogen Corporation, Life Technologies Division), in a 5% CO2 -95% humidity incubator at 37 °C. In a well of a 96 well plate, 30,000 cells were seeded and treated with compound at the indicated concentrations for 48 hr in a 5% CO2- 95% humidity incubator at 37°C. Control wells were treated with the same amount of solvent (DMSO) as the compound samples. After the indicated treatment time, the supernatant was removed to a sterile culture tube and the wells washed with phosphate buffered saline, and the adherent cells trypsinized for 5 min. The trypsinzed cells were added to the culture supernatant, cells were collected (1,200 rpm, 10 min), washed with phosphate buffered saline, and resuspended in fresh media.
The cells were counted for trypan blue negative cells, and the cells were diluted with fresh media to 1,000 cells/ml. To a well of a 24- well plate, 0.1 ml of the cell suspension was added along with 1 ml of fresh media (cell suspensions were passed through a 22G needle several times just before plating to form single cell suspensions). Plates are incubated in a 5% CO2- 95% humidity incubator at 37°C for 5-7 days. Colonies are counted when the sizes reached greater than 50 cells per colony. Cells are washed with phosphate buffered saline, fixed with 100% methanol for 15 min, and then stained with 0.5% gentian violet for 15 min. Colonies are rinsed with water and the colonies counted and the fraction surviving expressed as the percentage of the number of control colonies.
The results showed that after a 48 hr treatment, 6-chloro-N-(4-ethoxy-2- nitrophenyl)-3-pyridinecarboxamide inhibits the ability of HeLa cells to continue to proliferate and inhibited their colony forming ability with an IC50 of about 100 nM (Fig. 5)
Having now fully described this invention, it will be understood by those of ordinary skill in the art that the same may be performed within a wide and equivalent range of conditions, formulations and other parameters without affecting the scope of the invention or any embodiment thereof. All patents, patent applications and publications cited herein are fully incorporated by reference herein in their entirety.

Claims (1)

  1. What Is Claimed Is:
    1. A method of treating a disorder responsive to the induction of apoptosis in an animal suffering therefrom, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula N:
    or a pharmaceutically acceptable salt or prodrug thereof, wherein: Ar' and Ar are independently optionally substituted aryl or optionally substituted heteroaryl, provided that the ring structure of said optionally substituted heteroaryl comprises not more than two nitrogen atoms; and
    Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    The method of claim 1, wherein said compound is of Formula I:
    or pharmaceutically acceptable salts or prodrugs thereof, wherein:
    A is Ν or C-R8, B is Ν or C-R9, D is Ν or C-R10, E is Ν or C-R6 and F is Ν or C-R7, provided that not more than two of A, B, D, E and F are Ν in the same time; Ar is optionally substituted and is an aryl or heteroaryl;
    R6-Rio are independently hydrogen, halo, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, cyano, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, carboxy, carbonylamido or alkylthiol; and
    Rπ is hydrogen or optionally substituted alkyl, cycloalkyl, aryl, or heteroaryl.
    3. The method of claim 2, wherein A is N, B is C-R9 and F is C-R7.
    4. The method of claim 1, wherein Ar' is optionally substituted and is furyl, pyrrolyl, pyrazolyl, imidazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or phenyl.
    5. The method of claim 4, wherein Ar' is optionally substituted furyl.
    6. The method of claim 5, wherein Ar is optionally substituted phenyl.
    7. The method of claim 6, wherein Ar' is optionally substituted 3-furyl.
    8. The method of claim 7, wherein Ar' is unsubstituted 3-furyl.
    9. The method of claim 8, wherein said compound is JV-(4-ethoxy-2- nitrophenyl)-3-furancarboxamide.
    10. The method of claim 4, wherein Ar' is optionally substituted pyrrolyl.
    11. The method of claim 10, wherein Ar is optionally substituted phenyl.
    12. The method of claim 11, wherein Ar' is optionally substituted 3 -pyrrolyl.
    13. The method of claim 12, wherein Ar' is unsubstituted 3-pyrrolyl.
    14. The method of claim 13, wherein said compound is N-(4-ethoxy'-2- nitrophenyl)-3-pyrrolecarboxamide.
    15. The method of claim 4, wherein Ar' is optionally substituted phenyl.
    16. The method of claim 15, wherein Ar is optionally substituted phenyl.
    17. The method of claim 16, wherein said compound is selected from the group consisting of:
    4-Chloro-7V-(4-ethoxy-2-nitrophenyl)-benzoylamide; and 4-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-benzoylamide.
    18. The method of claim 15, wherein Ar is optionally substituted pyridyl.
    19. The method of claim 18, wherein said compound is selected from the group consisting of:
    4-Chloro-2-nitro-7V-(6-chloro-3-pyridyl)-benzoylamide; 4-Chloro-2-nitro-7V-(6-methyl-3-pyridyl)-benzoylamide; and
    4-Bromomethyl-3-nitro-7V-(6-chloro-3-pyridyl)-benzoylamide.
    20. The method of claim 4, wherein Ar' is optionally substituted pyrazinyl.
    21. The method of claim 20, wherein Ar is optionally substituted phenyl.
    22. The method of claim 21 , wherein Ar' is 3-pyrazinyl.
    23. The method of claim 22, wherein said compound is selected from the group consisting of:
    6-Methyl-7V-(4-ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide; and 7V-(4-Ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide.
    24. The method of claim 4, wherein Ar' is optionally substituted pyrimidinyl.
    25. The method of claim 24, wherein Ar is optionally substituted phenyl.
    26. The method of claim 25, wherein Ar' is optionally substituted 5-pyrimidinyl.
    27. The method of claim 26, wherein said compound is 7V-(4-ethoxy-2- nitrophenyl)-5-pyrimidinecarboxamide.
    28. The method of claim 4, wherein Ar' is optionally substituted pyridyl.
    29. The method of claim 28, wherein Ar is optionally substituted phenyl.
    30. The method of claim 29, wherein Ar' is optionally substituted 2-pyridyl.
    31. The method of claim 30, wherein said compound is selected from the group consisting of:
    N-(4-Ethoxy-2-nitrophenyl)-2-pyridinecarboxamide; and N-(4-Ethoxy-2-nitrophenyl)- 1 -7V-oxide-2-pyridinecarboxamide.
    32. The method of claim 29, wherein Ar' is optionally substituted 3-pyridyl.
    33. The method of claim 32, wherein said compound is of Formula JJJ:
    or a pharmaceutically acceptable salt or prodrug thereof, wherein Ri-R7 and R -Rio are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    34. The method of claim 33, wherein R\ and R2, or R2 and R3, or R3 and R4, or R4 and R5 are taken together to form an optionally substituted carbocycle or an optionally substituted heterocycle.
    35. The method of claim 34, wherein said Ri and R , or R2 and R3, or R3 and R4, or Rφ and R5 are taken together to form -OCH2O- -(CH2)3- -(CH2)4- -OCH2CH2O- -CH2N(R)CH2-, -CH2CH2N(R)CH2~, -CH2N(R)CH2CH2-, -CH=CH-CH=CH-, -N(R)-CH=CH-, -CH=CH-N(R)-, -O-CH=CH-, -CH=CH-O-
    -S-CH=CH-, -CH=CH-S-, -N=CH-CH=CH- -CH=N-CH=CH-, -CH=CH-N=CH- -CH=CH-CH=N- or -N=CH-CH=N-, wherein the carbocycle or heterocycle is optionally substituted, and R is hydrogen, alkyl, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl or aminoalkyl.
    36. The method of claim 33, wherein R6, R7 and Rio are independently hydrogen or fluoro.
    37. The method of claim 33, wherein Ri is nitro.
    38. The method of claim 33, wherein R2, R , and R5 are independently hydrogen or fluoro.
    39. The method of claim 33, wherein said compound is selected from the group consisting of:
    N-(4-Methyl-2-nitrophenyl)-3-pyridinecarboxamide;
    N-(4-Ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; N-(4-Methoxy-2-nitrophenyl)-3 -pyridinecarboxamide;
    6-Chloro-N-(4,5-difluoro-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-7Vr-(3-bromo-4-methoxy-6-nitrophenyl)-3-pyridinecarboxamide;
    5,6-Dichloro-7V-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(2-methyl-4-methoxyphenyl)-3-pyridinecarboxamide; 6-Chloro-7V-(4-ethoxy-2-nitrophenyl)-N-methyl-3-pyridinecarboxamide;
    6-Chloro-7V-(2-cyano-4,5-dimethoxyphenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-chloro-2-trifluoromethylphenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-chloro-2-cyanophenyl)-3-pyridinecarboxamide;
    6-CWoro-N-(2,4-dimethyl-6-nitrophenyl)-3-pyridinecarboxamide; 6-CUoro-N-(3,4-dimethoxy-6-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(2-cyano-4-methylphenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-chloro-2-methyl-6-nitrophenyl)-3-pyridinecarboxamide; and
    4-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide.
    40. The method of claim 33, wherein said compound is of Formula TV:
    or a pharmaceutically acceptable salt or prodrug thereof.
    41. The method of claim 40, wherein said compound is selected from the group consisting of:
    6-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Chloro-iV-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-methyl-2-nitrophenyl)-3-pyridinecarboxamide;
    6-CUoro-N-(4-methoxy-2-nitrophenyl)-l-7V-oxide-3-pyridinecarboxamide;
    6-Chloro-7y-(4-chloro-2-nitrophenyl)-3-pyridinecarboxamide; 6-Fluoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-fluoro-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-trifluoromethyl-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(2-nitro-4-trifluoromethoxylphenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-benzyloxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Methyl-7V-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-cyano-2-nitrophenyl)-3-pyridinecarboxamide;
    6-(2,2,2-Trifluoroethoxy)-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Dimethylamino-N-(4-ethoxy-2-nifrophenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-t-butyl-2-nitrophenyl)-3-pyridinecarboxamide; 6-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; and
    6-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide.
    42. A method for treating or preventing cancer, comprising administering to an animal in need of such treatment an effective amount of a compound of Formula N:
    or a pharmaceutically acceptable salt or prodrug thereof, wherein:
    Ar' and Ar are independently optionally substituted aryl or optionally substituted heteroaryl, provided that the ring structure of said optionally substituted heteroaryl comprises not more than two nitrogen atoms; and
    Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    43. The method of claim 42, wherein said compound is of Formula I:
    or pharmaceutically acceptable salts or prodrugs thereof, wherein:
    A is N or C-R8, B is N or C-R9, D is N or C-R10, E is N or C-R6 and F is N or C-R7, provided that not more than two of A, B, D, E and F are N in the same time; Ar is optionally substituted and is an aryl or heteroaryl;
    R6-R10 are independently hydrogen, halo, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, cyano, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, carboxy, carbonylamido or alkylthiol; and
    Rn is hydrogen or optionally substituted alkyl, cycloalkyl, aryl, or heteroaryl.
    44. The method of claim 42, wherein Ar' is optionally substituted and is furyl, pyrrolyl, pyrazolyl, imidazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or phenyl.
    45. The method of claim 42, wherein said cancer is selected from the group consisting of Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic leukemia, chronic lymphocytic leukemia, multiple myeloma, neuroblastoma, breast carcinoma, ovarian carcinoma, lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, soft-tissue sarcoma, primary macroglobulinemia, bladder carcinoma, chronic granulocytic leukemia, primary brain carcinoma, malignant melanoma, small-cell lung carcinoma, stomach carcinoma, colon carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, malignant melanoma, choriocarcinoma, mycosis fungoides, head or neck carcinoma, osteogenic sarcoma, pancreatic carcinoma, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, malignant hypercalcemia, cervical hyperplasia, renal cell carcinoma, endometrial carcinoma, polycythemia vera, essential thrombocytosis, adrenal cortex carcinoma, skin cancer and prostatic carcinoma.
    46. A method for the treatment of drug resistant cancer, comprising administering to an animal in need of such treatment an effective amount of a compound of the Formula N:
    or a pharmaceutically acceptable salt or prodrug thereof, wherein:
    Ar' and Ar are independently optionally substituted aryl or optionally substituted heteroaryl, provided that the ring structure of said optionally substituted heteroaryl comprises not more than two nitrogen atoms; and
    Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    47. The method of claim 46, wherein said compound is of Formula I:
    or pharmaceutically acceptable salts or prodrugs thereof, wherein:
    A is Ν or C-R8, B is Ν or C-R9, D is Ν or C-Rio, E is Ν or C-R6 and F is Ν or C-R7, provided that not more than two of A, B, D, E and F are Ν in the same time;
    Ar is optionally substituted and is an aryl or heteroaryl; R6-Rio are independently hydrogen, halo, haloalkyl, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, cyano, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, carboxy, carbonylamido or alkylthiol; and
    Rπ is hydrogen or optionally substituted alkyl, cycloalkyl, aryl, or heteroaryl.
    48. The method of claim 46, wherein Ar' is optionally substituted and is furyl, pyrrolyl, pyrazolyl, imidazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or phenyl.
    49. The method of claim 42 or 46, additionally comprising administering at least one known cancer chemotherapeutic agent, or a pharmaceutically acceptable salt of said agent.
    50. The method of claim 49, wherein said known cancer therapeutic agent is selected from the group consisting of busulfan, cis-platin, mitomycin C, carboplatin, colchicine, vinblastine, paclitaxel, docetaxel, camptothecin, topotecan, doxorubicin, etoposide, 5-azacytidine, 5-fluorouracil, methotrexate, 5-fluoro-2'-deoxy-uridine, ara-C, hydroxyurea, thioguanine, melphalan, chlorambucil, cyclophosamide, ifosfamide, vincristine, mitoguazone, epirubicin, aclarubicin, bleomycin, mitoxantrone, elliptinium, fludarabine, octreotide, retinoic acid, tamoxifen, Herceptin®, Rituxan® and alanosine.
    51. The method of claim 42 or 46, additionally comprising treating said animal with radiation-therapy.
    52. The method of claim 42 or 46, wherein said compound is administered after the surgical treatment of said animal for cancer.
    53. The method of claim 1, wherein said disorder is an autoimmune disease.
    54. The method of claim 1 , wherein said disorder is rheumatoid arthritis.
    55. The method of claim 1 , wherein said disorder is inflamatory bowel disease.
    56. The method of claim 1 , wherein said disorder is a skin disease.
    57. The method of claim 56, wherein said disorder is psoriasis.
    58. A compound of Formula JJJ:
    or a pharmaceutically acceptable salt or prodrug thereof, wherein:
    Ri and R5 are independently selected from the group consisting of hydrogen, hydroxy, alkyl, alkoxy, halogen, NO2, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl, provided that at least one of Ri and R5 is selected from the group consisting of NO2, cyano, alkyl and haloalkyl;
    R2 and J^. are independently selected from the group consisting of hydrogen, hydroxy, halogen, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl;
    R3 is alkyl, Cl, F, haloalkyl, alkoxy, arylalkoxy, cyano, haloalkyloxy, amino or aminoalkyl; R6 is hydrogen, hydroxy, alkyl, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl;
    R7 is hydrogen, hydroxy, alkyl, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl;
    R9 is hydroxy, alkyl, halogen, NO2, haloalkyl, alkoxy, cyano, haloalkyloxy, amino or aminoalkyl; R10 is hydrogen, hydroxy, alkyl, Cl, F, NO2, cyano, haloalkyl, haloalkyloxy, amino or aminoalkyl; and
    Rπ is hydrogen, alkyl or haloalkyl.
    59. The compound of claim 58, wherein said compound is selected from the group consisting of:
    6-ChlororN-(4,5-difluoro-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-7 -(3-bromo-4-methoxy-6-nitrophenyl)-3-pyridinecarboxamide;
    5,6-Dichloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide; 6-Chloro-N-(2-methyl-4-methoxyphenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-ethoxy-2-nifrophenyl)-N-methyl-3-pyridinecarboxamide;
    6-Chloro-N-(2-cyano-4,5-dimethoxyphenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-chloro-2-trifluoromethylphenyl)-3-pyridinecarboxamide;
    6-Chloro-7V-(4-chloro-2-cyanophenyl)-3-pyridinecarboxamide; 6-Chloro-N-(2,4-dimethyl-6-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(3,4-dimethoxy-6-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(2-cyano-4-methylphenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-chloro-2-methyl-6-nitrophenyl)-3-pyridinecarboxamide; and
    4-Trifluoromethyl-iV-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide.
    60. The compound of claim 58, wherein said compound is of Formula JN:
    or a pharmaceutically acceptable salt or prodrug thereof.
    61. The compound of claim 60, wherein said compound is selected from the group consisting of: 6-Chloro-N-(4-methoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-CUoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-methyl-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Cl loro-N-(4-methoxy-2-nitrophenyl)-l-7\'r-oxide-3-pyridinecarboxamide;
    6-Chloro-N-(4-chloro-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Fluoro-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Cbloro-N-(4-fluoro-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-trifluoromethyl-2-nitrophenyl)-3-pyridinecarboxamide;
    6-CUoro-N-(2-nitro-4-trifluoromethoxylphenyl)-3-pyridinecarboxamide;
    6-CMoro-N-(4-benzyloxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Methyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-cyano-2-nitrophenyl)-3-pyridinecarboxamide;
    6-(2,2,2-Trifluoroethoxy)-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Dimethylamino-N-(4-ethoxy-2-nifrophenyl)-3-pyridinecarboxamide;
    6-Chloro-N-(4-t-butyl-2-nitrophenyl)-3-pyridinecarboxamide;
    6-Trifluoromethyl-N-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide; and
    6-Chloromethyl-7Vr-(4-ethoxy-2-nitrophenyl)-3-pyridinecarboxamide.
    62. A compound of Formula (NT) :
    or a pharmaceutically acceptable salt or prodrug thereof, wherein
    Ri-Rs, R7 and R -Rio are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, . acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
    Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    63. The compound of claim 62, or a pharmaceutically acceptable salt thereof, with the prioviso that at least one of Ri and R5 is selected from the group consisting of NO2, cyano, alkyl and haloalkyl.
    64. The compound of claim 62 or a pharmaceutically acceptable salt or prodrug thereof, wherein:
    Ri and R5 are independently selected from the group consisting of hydrogen, hydroxy, alkyl, halogen, NO2, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl; R2 and R4 are independently selected from the group consisting of hydrogen, hydroxy, halogen, haloalkyl, haloalkoxy, amino and aminoalkyl;
    R3 is alkyl, Cl, F, haloalkyl, alkoxy, arylalkoxy, cyano, haloalkoxy, amino or aminoalkyl;
    R7, R9 and R10 are independently selected from the group consisting of hydrogen, hydroxy, alkyl, halogen, NO2, cyano, haloalkyl, alkoxy, haloalkoxy, amino and aminoalkyl; and
    Rπ is hydrogen, alkyl or haloalkyl.
    65. The compound of claim 64, wherein said compound is selected from the group consisting of:
    6-Methyl-7V-(4-ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide; and N-(4-Ethoxy-2-nitrophenyl)-3-pyrazinecarboxamide.
    66. A compound of Formula (NJJ):
    or a pharmaceutically acceptable salt or prodrug thereof, wherein: R1-R3, R5-R10 are independently hydrogen, halo, haloalkyl, haloalkoxy, aryl, fused aryl, carbocyclic, a heterocyclic group, a heteroaryl group, alkyl, alkenyl, alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, carbocycloalkyl, heterocycloalkyl, hydroxyalkyl, nitro, amino, aminoalkyl, cyano, cyanoalkyl, acyl, acylamido, hydroxy, thiol, acyloxy, azido, alkoxy, alkoxycarbonyl, aryloxy, arylalkoxy, carboxy, carbonylamido or alkylthiol; and
    Rπ is hydrogen; or alkyl, cycloalkyl, aryl or heteroaryl, each of which is optionally substituted.
    67. The compound of claim 66, or a pharmaceutically acceptable salt thereof, with the prioviso that at least one of R6 and R is selected from the group consisting of
    NO2, cyano, alkyl and haloalkyl.
    68. The compound of claim 67 or a pharmaceutically acceptable salt or prodrug thereof, wherein: Ri, R , R3 and R5 are independently selected from the group consisting of hydrogen, hydroxy, alkyl, halogen, NO2, cyano, haloalkyl, alkoxy, haloalkoxy, amino and aminoalkyl;
    R6 and R are independently selected from the group consisting of hydrogen, hydroxy, alkyl, halogen, NO2, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl; R8 and Rio are independently selected from the group consisting of hydrogen, hydroxy, alkyl, halogen, NO2, cyano, haloalkyl, haloalkoxy, amino and aminoalkyl; R is hydroxy, alkyl, halogen, NO , cyano, haloalkyl, alkoxy, haloalkoxy, amino or aminoalkyl; and
    Rπ is hydroxy, alkyl or haloalky.
    69. The compound of claim 66, wherein said compound is selected from the group consisting of:
    4-Chloro-2-nitro-7V-(6-chloro-3-pyridyl)-benzoylamide; 4-Chloro-2-nitro-7V-(6-methyl-3-pyridyl)-benzoylamide; and 4-Bromomethyl-3-nitro-N-(6-chloro-3-pyridyl)-benzoylamide.
    70. A compound selected from the group consisting of: N-(4-Ethoxy-2-nitrophenyl)-3-pyrrolylcarboxamide; 4-Chloro-N-(4-ethoxy-2-nifrophenyl)-benzoylarnide; 4-Chloromethyl-N-(4-ethoxy-2-nitrophenyl)-benzoylamide; N-(4-Ethoxy-2-nitrophenyl)-5-pyrimidinecarboxamide;
    N-(4-Ethoxy-2-nitrophenyl-2-pyridinecarboxamide; and N-(4-Ethoxy-2-nitrophenyl)- 1 -/V-oxide-2-pyridinecarboxamide.
    71. A pharmaceutical composition, comprising the compound of any one of claims 58-70, and a pharmaceutically acceptable carrier.
    72. The pharmaceutical composition of claim 71, further comprising at least one known cancer chemotherapeutic agent, or a pharmaceutically acceptable salt of said agent.
    73. The pharmaceutical composition of claim 72, wherein said known cancer chemotherapeutic agent is selected from the group consisting of busulfan, cis-platin, mitomycin C, carboplatin, colchicine, vinblastine, paclitaxel, docetaxel, camptothecin, topotecan, doxorubicin, etoposide, 5-azacytidine, 5-fluorouracil, methotrexate, 5-fluoro-2'- deoxy-uridine, ara-C, hydroxyurea, thioguanine, melphalan, chlorambucil, cyclophosamide, ifosfamide, vincristine, mitoguazone, epirabicin, aclarubicin, bleomycin, mitoxantrone, elliptinium, fludarabine, octreotide, retinoic acid, tamoxifen, Herceptin®, Rituxan® and alanosine.
AU2001231143A 2000-01-27 2001-01-26 Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof Abandoned AU2001231143A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US17764800P 2000-01-27 2000-01-27
US60177648 2000-01-27
PCT/US2001/002478 WO2001055115A1 (en) 2000-01-27 2001-01-26 Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof

Publications (1)

Publication Number Publication Date
AU2001231143A1 true AU2001231143A1 (en) 2001-08-07

Family

ID=22649398

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2001231143A Abandoned AU2001231143A1 (en) 2000-01-27 2001-01-26 Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof

Country Status (6)

Country Link
US (2) US6794397B2 (en)
EP (1) EP1257536A1 (en)
JP (1) JP2003520854A (en)
AU (1) AU2001231143A1 (en)
CA (1) CA2397493A1 (en)
WO (1) WO2001055115A1 (en)

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6495550B2 (en) * 1999-08-04 2002-12-17 Icagen, Inc. Pyridine-substituted benzanilides as potassium ion channel openers
AU779034B2 (en) 1999-08-04 2005-01-06 Icagen, Inc. Benzanilides as potassium channel openers
GB0001930D0 (en) 2000-01-27 2000-03-22 Novartis Ag Organic compounds
US20070208087A1 (en) 2001-11-02 2007-09-06 Sanders Virginia J Compounds, compositions and methods for the treatment of inflammatory diseases
WO2002053544A1 (en) * 2000-12-29 2002-07-11 Darwin Discovery Ltd. Pharmaceutical uses and synthesis of nicotinanilide-n-oxides
US6995162B2 (en) 2001-01-12 2006-02-07 Amgen Inc. Substituted alkylamine derivatives and methods of use
US7102009B2 (en) 2001-01-12 2006-09-05 Amgen Inc. Substituted amine derivatives and methods of use
US6878714B2 (en) 2001-01-12 2005-04-12 Amgen Inc. Substituted alkylamine derivatives and methods of use
US20030134836A1 (en) * 2001-01-12 2003-07-17 Amgen Inc. Substituted arylamine derivatives and methods of use
US7105682B2 (en) 2001-01-12 2006-09-12 Amgen Inc. Substituted amine derivatives and methods of use
US7398549B2 (en) 2001-05-18 2008-07-08 Imprivata, Inc. Biometric authentication with security against eavesdropping
DE10133665A1 (en) * 2001-07-11 2003-01-30 Boehringer Ingelheim Pharma Carboxylic acid derivatives, medicaments containing these compounds, their use and preparation
CA2480497A1 (en) 2002-04-26 2003-11-06 Pharmacia & Upjohn Company Substituted pyrazine derivatives
DK1511710T3 (en) 2002-05-31 2014-02-24 Proteotech Inc RELATIONSHIPS, PREPARATIONS AND METHODS FOR TREATING AMYLOID DISEASES AND SYNUCLEINOPATHES, SUCH AS ALZHEIMER'S DISEASE, TYPE 2-DIABETES AND PARKINSON'S DISEASE
AU2003249713A1 (en) * 2002-07-03 2004-01-23 Axys Pharmaceuticals, Inc. 3,4-dihydroisoquinolin-1-one derivatives as inducers of apoptosis
US7307088B2 (en) * 2002-07-09 2007-12-11 Amgen Inc. Substituted anthranilic amide derivatives and methods of use
US7432281B2 (en) 2003-10-07 2008-10-07 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
US7592466B2 (en) 2003-10-09 2009-09-22 Abbott Laboratories Ureas having antiangiogenic activity
US7507748B2 (en) 2004-07-22 2009-03-24 Amgen Inc. Substituted aryl-amine derivatives and methods of use
US7576099B2 (en) 2005-02-28 2009-08-18 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
JP2008533154A (en) * 2005-03-14 2008-08-21 レノビス, インコーポレイテッド Amide derivatives and pharmaceutical compositions as ion channel ligands and methods of using them
US8247556B2 (en) 2005-10-21 2012-08-21 Amgen Inc. Method for preparing 6-substituted-7-aza-indoles
AU2012202993B2 (en) * 2007-03-01 2015-06-25 Novartis Ag Pim kinase inhibitors and methods of their use
MX2009009304A (en) * 2007-03-01 2009-11-18 Novartis Ag Pim kinase inhibitors and methods of their use.
EP1987717A1 (en) 2007-04-30 2008-11-05 Bayer CropScience AG Pyridon carboxamides, agents containing these but not impacting useful plants and method for their manufacture and application
CA2693967A1 (en) * 2007-07-19 2009-01-29 Schering Corporation Heterocyclic amide compounds as protein kinase inhibitors
US20090239285A1 (en) 2008-03-19 2009-09-24 Jose Alberto Fernandez-Pol Tandem reapeat dna constructs producing proteins that attack plant pathogenic viruses, fungi, and bacteria by disrupting transcription factors essential for replication thereof in plants
EA020136B1 (en) 2008-09-02 2014-08-29 Новартис Аг Picolinamide derivatives as kinase inhibitors
JP6486002B2 (en) * 2010-08-23 2019-03-20 シントリックス・バイオシステムズ・インコーポレイテッドSyntrix Biosystems, Inc. Aminopyridine carboxamide and aminopyrimidine carboxamide as CXCR2 modulators
EP2861576B1 (en) 2012-05-15 2018-01-10 Novartis AG Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1
AP3613A (en) 2012-05-15 2016-02-29 Novartis Ag Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1 abl1abl2 bcr-abl1
CA2871332A1 (en) 2012-05-15 2013-11-21 Novartis Ag Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1
PL2900637T3 (en) 2012-05-15 2018-01-31 Novartis Ag Thiazole or imidazole substituted pyrimidine, pyridine and pyrazine amide derivatives and related compounds as abl1, abl2 and bcr-abl1 inhibitors for the treatment of cancer, specific viral infections and specific cns disorders
MX2014014253A (en) * 2012-05-21 2015-02-17 Novartis Ag Novel ring-substituted n-pyridinyl amides as kinase inhibitors.
CN103265479B (en) * 2013-06-14 2017-12-08 南开大学 A kind of synthetic method of the 6 chloromethyl nicotinic acid tert-butyl ester
MX2016000141A (en) 2013-07-09 2016-03-01 Bayer Cropscience Ag Use of selected pyridone carboxamides or salts thereof as active substances against abiotic plant stress.
EP3020183B1 (en) * 2013-07-11 2019-12-11 Harman International Industries, Inc. System and method for digital audio conference workflow management
WO2016016728A2 (en) * 2014-07-31 2016-02-04 University College Cardiff Consultants Limited Bcl-3 inhibitors
JO3589B1 (en) 2014-08-06 2020-07-05 Novartis Ag Protein kinase c inhibitors and methods of their use
US9716653B2 (en) * 2014-11-18 2017-07-25 Hauwei Technologies Co., Ltd. System and method for flow-based addressing in a mobile environment
AR103024A1 (en) 2014-12-18 2017-04-12 Bayer Cropscience Ag SELECTED PYRIDONCARBOXAMIDS OR ITS SALTS AS ACTIVE SUBSTANCES AGAINST ABIOTIC PLANTS STRESS
US20210120817A1 (en) * 2017-03-17 2021-04-29 Meiji Seika Pharma Co., Ltd. Plant disease control agent
CN111559977B (en) * 2020-06-04 2021-06-22 广州市朝利良生物科技有限公司 Micromolecular compound and application thereof in preparation of anti-tumor metastasis medicines
CN112358442B (en) * 2020-11-20 2022-03-04 内蒙古佳瑞米精细化工有限公司 Preparation method of 2-fluoro-5-formyl chloropyridine

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH438343A (en) 1962-11-08 1967-06-30 Thomae Gmbh Dr K Process for the preparation of 5,6-dihydro-6-oxo-11H-pyrido (2,3-b) (1,4) -benzodiazepines
NL7714092A (en) * 1976-12-22 1978-06-26 Monsanto Co PROCESS FOR PREPARING NEW PHTALAMIN AND NICOTINIC ACIDS.
US4261730A (en) * 1978-08-09 1981-04-14 Monsanto Company Substituted pyridyl phthalamic acids and their use as plant growth regulants
FR2636329B2 (en) * 1987-03-13 1990-11-02 Roussel Uclaf NOVEL PYRIDINE DERIVATIVES, THEIR PREPARATION PROCESS, THEIR USE AS MEDICAMENTS AND THE COMPOSITIONS CONTAINING THEM
FR2612189B1 (en) * 1987-03-13 1989-06-23 Roussel Uclaf NOVEL PYRIDINE DERIVATIVES, THEIR PREPARATION PROCESS AND THE NEW INTERMEDIATES OBTAINED, THEIR APPLICATION AS MEDICAMENTS AND THE COMPOSITIONS CONTAINING THEM
US4978385A (en) * 1987-05-29 1990-12-18 Daicel Chemical Industries Ltd. 4-halopyridine-3-carboxamide compounds and herbicidal compositions thereof
DE3804346A1 (en) * 1988-02-12 1989-08-24 Boehringer Mannheim Gmbh tert-Butylphenylpyridyl amides, process for their preparation, and medicaments containing these compounds
US5703075A (en) * 1988-12-21 1997-12-30 Pharmacia & Upjohn Company Antiatherosclerotic and antithrombotic 1-benzopyran-4-ones and 2-amino-1,3-benzoxazine-4-ones
IE920921A1 (en) * 1991-04-04 1992-10-07 Eisai Co Ltd Benzothiazole derivative
DE4127404A1 (en) * 1991-08-19 1993-02-25 Thomae Gmbh Dr K CYCLIC IMINODERIVATES, MEDICAMENTS CONTAINING SUCH COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
JPH06263702A (en) * 1991-12-12 1994-09-20 Koichi Shudo Carboxylic acid derivative
GB9212693D0 (en) * 1992-06-15 1992-07-29 Celltech Ltd Chemical compounds
US6087160A (en) 1993-06-24 2000-07-11 The General Hospital Corporation Programmed cell death genes and proteins
GB9405347D0 (en) 1994-03-18 1994-05-04 Agrevo Uk Ltd Fungicides
US6028111A (en) * 1996-03-08 2000-02-22 Oxigene, Inc. Compositions and use of benzamides and nicotinamides as anti-inflammatory agents
PT807633E (en) 1996-05-15 2003-02-28 Pfizer NEW PYRIMIDIN-4-ONES REPLACED IN 2.3, AND FUSED WITH HETEROARYL (5.6)
AUPO622597A0 (en) * 1997-04-15 1997-05-08 Fujisawa Pharmaceutical Co., Ltd. New heterocyclic compounds
AU751139B2 (en) * 1997-10-13 2002-08-08 Astellas Pharma Inc. Amide derivative
US6022884A (en) 1997-11-07 2000-02-08 Amgen Inc. Substituted pyridine compounds and methods of use
US6191170B1 (en) * 1998-01-13 2001-02-20 Tularik Inc. Benzenesulfonamides and benzamides as therapeutic agents
AU3053199A (en) * 1998-04-01 1999-10-25 Ono Pharmaceutical Co. Ltd. Fused thiophene derivatives and drugs containing the same as the active ingredient
WO2000004901A1 (en) 1998-07-21 2000-02-03 Thomas Jefferson University Small molecule inhibitors of bcl-2 proteins
EP1101759A4 (en) * 1998-07-31 2001-12-12 Nippon Soda Co Phenylazole compounds, process for producing the same and drugs for hyperlipemia
WO2000044216A2 (en) 1999-02-01 2000-08-03 Cytovia Inc Gambogic acid, analogs and derivatives as activators of caspases and inducers of apoptosis
JP2000256358A (en) 1999-03-10 2000-09-19 Yamanouchi Pharmaceut Co Ltd Pyrazole derivative
JP2002539183A (en) 1999-03-16 2002-11-19 サイトビア インコーポレイテッド Substituted 2-aminobenzamide caspase inhibitors and uses thereof
CN101481359A (en) * 1999-04-15 2009-07-15 布里斯托尔-迈尔斯斯奎布公司 Cyclic protein tyrosine kinase inhibitors
AR030154A1 (en) 1999-07-05 2003-08-13 Nihon Nohyaku Co Ltd DERIVED FROM FTALAMIDE, DERIVED FROM HETEROCICLIC AMINE USEFUL AS INTERMEDIARY FOR THE PRODUCTION OF THE SAME, AGROHORTICALLY INSECTICIDE AND METHOD TO USE SUCH INSECTICIDE
WO2001021598A1 (en) * 1999-09-23 2001-03-29 Astrazeneca Ab Therapeutic quinazoline compounds
WO2002053544A1 (en) * 2000-12-29 2002-07-11 Darwin Discovery Ltd. Pharmaceutical uses and synthesis of nicotinanilide-n-oxides

Also Published As

Publication number Publication date
CA2397493A1 (en) 2001-08-02
WO2001055115A1 (en) 2001-08-02
US20020010185A1 (en) 2002-01-24
US20040235846A1 (en) 2004-11-25
EP1257536A1 (en) 2002-11-20
WO2001055115A8 (en) 2001-11-29
JP2003520854A (en) 2003-07-08
US6794397B2 (en) 2004-09-21

Similar Documents

Publication Publication Date Title
US6794397B2 (en) Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof
US6716851B2 (en) Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators or caspases and inducers of apoptosis and the use thereof
US7041685B2 (en) Substituted 3-aryl-5-aryl-[1,2,4]-oxadiazoles and analogs as activators of caspases and inducers of apoptosis and the use thereof
EP1351691A1 (en) Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators of caspases and inducers of apoptosis and the use thereof
AU2002313633A1 (en) Substituted 3-aryl-5-aryl-[1,2,4]-oxadiazoles and analogs
EP1230232B1 (en) Substituted 4h-chromene and analogs as activators of caspases and inducers of apoptosis and the use thereof
US7015328B2 (en) Substituted coumarins and quinolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20070099941A1 (en) N-arylalkyl-thienopyrimidin-4-amines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US6716859B2 (en) Substituted N′-(Arylcarbonyl)-benzhydrazides, N′(Arylcarbonyl)-benzylidene-hydrazides and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20030065018A1 (en) Substituted 4H-chromenes and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20080096848A1 (en) Substituted N-Aryl-9-Oxo-9H-Fluorene-1-Carboxamides and Analogs as Activators of Caspases and Inducers of Apoptosis
WO2002102301A2 (en) Substituted indole-2-carboxylic acid benzylidene-hydrazides and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20050165053A1 (en) Substituted4-aryl-3-(3-aryl-1-oxo-2-propenyl)-2(1h)-quinolinones and analogs as activators of caspases and inducers of apoptosis and the use thereof
US6794400B2 (en) 4-substituted-1-(arylmethylidene)thiosemicarbazide, 4-substituted-1-(arylcarbonyl)thiosemicarbazide and analogs as activators of caspases and inducers of apoptosis and the use thereof

Legal Events

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