AU726522B2 - Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions - Google Patents

Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions Download PDF

Info

Publication number
AU726522B2
AU726522B2 AU71631/96A AU7163196A AU726522B2 AU 726522 B2 AU726522 B2 AU 726522B2 AU 71631/96 A AU71631/96 A AU 71631/96A AU 7163196 A AU7163196 A AU 7163196A AU 726522 B2 AU726522 B2 AU 726522B2
Authority
AU
Australia
Prior art keywords
carboxamide
mmol
pyrimidine
compound
compounds
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.)
Ceased
Application number
AU71631/96A
Other versions
AU7163196A (en
Inventor
Leah M. Gayo
Mark E. Goldman
Moorthy S. S. Palanki
Lynn J. Ransone-Fong
Robert W. Sullivan
Mark J. Suto
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.)
Signal Pharmaceuticals LLC
Original Assignee
Signal Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/581,473 external-priority patent/US5811428A/en
Application filed by Signal Pharmaceuticals LLC filed Critical Signal Pharmaceuticals LLC
Publication of AU7163196A publication Critical patent/AU7163196A/en
Application granted granted Critical
Publication of AU726522B2 publication Critical patent/AU726522B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • 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
    • 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]
    • 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
    • C07D239/30Halogen atoms or nitro radicals
    • 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
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen atom
    • 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
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Description

WO 97/09315 PCT/US96/15108 1 Description PYRIMIDINE CARBOXAMIDES AND RELATED COMPOUNDS AND METHODS FOR TREATING INFLAMMATORY CONDITIONS Technical Field The present invention relates generally to compounds that block intracellular signal transduction and activation of transcription factors, and to methods for preventing or treating immunoinflammatory and autoimmune diseases.
Background of the Invention Signals necessary for cell growth, differentiation, response to bioregulatory molecules, infectious agents and physiological stress involve changes in the rates of gene expression. The ability to respond appropriately to such signaling events challenge the survival of the cell and ultimately the organism. Perturbations in the normal regulation of these specific genetic responses can result in pathogenic events which lead to acute and chronic disease.
In certain autoimmune diseases or chronic inflammatory states, continuous activation of T-cells eventually leads to a self-perpetuating destruction of normal tissues or organs. This is caused by the induction of adhesion molecules, chemotaxis of leukocytes, activation of leukocytes and the production of mediators of inflammation. All of these events are regulated at the level of transcription for the production of new proteins, including cytokines. The production of cytokines, as well as a number of other cellular regulators, is controlled by a family of proteins known as transcription factors (TFs). These transcription factors, when activated, bind to specific regions on the DNA and act as molecular switches or messengers to induce or upregulate gene expression. The activation of these TFs is caused by a variety of external signals including physiological stress, infectious agents and other bioregulatory molecules. Once the plasma membrane receptors are activated, a cascade of protein kinases and second messengers are induced which, in turn, result in the production of RNA transcripts. The end result is the production of proinflammatory proteins via translation and processing of the RNA transcripts.
This activation system can, at times, be very robust. For example, a specific set of external signals could result in a single transcription factor to induce many proteins responsible for a given disease. Therefore, regulating this process by disrupting WO 97/09315 PCT/US96/15108 2 the production of activated TF(s) has the potential to attenuate the production of the associated pathological proteins, thereby halting or reversing the course of the disease.
Two transcription factors, NFcB and AP-1, have been shown to regulate the production of many proinflammatory cytokines and related proteins that are elevated in immunoinflammatory diseases. These TFs regulate interleukin-1 interleukin-2 tumor necrosis factor-a (TNFa), interleukin-6 (IL-6) and interleukin-8 (IL-8) levels in a variety of cell types. For example, NFiB and other related complexes are involved in the rapid induction of genes whose products function in protective and proliferative responses upon exposure of cells to external stimuli. Similarly, AP-1 has a significant role in the regulation of interleukin-2 (IL-2) and tumor necrosis factor-a (TNF-a) transcription during T-cell activation. In addition, TNF-oa and IL-1 are strong activators of collagenase, gelatinase and stromelysin gene expression, which require a single AP-1 binding site in the promoter region of these genes. Therefore, an inhibitor of NFB and/or AP-1 activation would coordinately repress the activities of a series of proteinases. In addition, cell adhesion molecules are also controlled by these TFs. All of these proteins have been shown to play a role in diseases, including osteoarthritis, transplant rejection, ischemia, reperfusion injury, trauma, certain cancers and viral disorders, and autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, psoriasis, inflammatory bowel disease, glomerulonephritis, lupus and juvenile diabetes.
In summary, the role of these TFs is to act as a transducer for certain stimuli that lead to immune, inflammatory, and acute phase responses.
Since many diseases are caused by the inappropriate production of proteins, conventional therapeutic approaches have focused on inhibiting function or activity of individual effector proteins. These treatments have not always proved to be effective and, at times, are associated with many undesirable side effects. Therefore, there is a need for new therapies for the prevention and/or treatment of immunoinflammatory and autoimmune diseases. More specifically, there is a need for compounds that prevent, preferably by inhibiting transcription at an early stage, the production of proteins associated with immunoinflammatory and autoimmune diseases.
Furthermore, these compounds should inhibit the kinase(s) that regulate the activation of TFs such as NFiB and AP-1. The present invention fulfills these needs and provides further related advantages.
Summary of the Invention In brief, this invention is directed to compounds that block the activation of transcription factors (TFs), particularly NFKB and AP-1, and are believed to function P'\OPER\POD M63 I-96..isd-i4/9/GO -3through inhibition of a family of specific kinases. This results in a decrease in a number of proinflammatory proteins, including IL-1, IL-2, IL-8 and/or TNFa, which are responsible for tissue and organ damage associated with diseases such as rheumatoid arthritis, osteoarthritis, related autoimmune disorders and tissue rejection. Accordingly, compounds of the present invention are useful in, for example, the prevention of organ and tissue rejection associated with transplantation. Furthermore, the compounds of this invention also have utility in the prevention and/or treatment of immunoinflammatory and autoimmune diseases, as well as having general activity as anti-inflammatory agents.
In a first aspect, the invention provides a method for treating an inflammatory condition in a warm blooded animal in need thereof comprising administering to said animal an effective amount of a compound having the structure:
R
R4aRRs
**Y
including pharmaceutically acceptable salts thereof, wherein 15 Rs is selected from the following chemical moieties: R RS SN S N R RT r R R7 N O N
S
R
7 is selected from hydrogen, -CH 3 and -CH2C 6 Hs;
R
8 is selected from hydrogen and an unsubstituted or substituted Cvsalkyl, as hereinbefore defined, C 6 .12aryl, C 7 1 2 aralkyl, C 3 -1 2 heterocycle and a C4.20heterocyclealkyl; R2a is selected from halogen, an unsubstituted or substituted Cl.salkyl as hereinbefore defined, C 6 -1 2 aryl, C 7 1 2 aralkyl, C3-12heterocycle or C4_2oheterocyclealkyl, CN, -OR, -NRR, H, -NHNH 2
NHC
6 Hs, and -NRCOR; PAOPER\PDB\716J1-9 6 claiMSdoc-1409/ -3A- R4a is selected from hydrogen, halogen, an unsubstituted or substituted Ci-salkyl as hereinbefore defined, C6-12aryl, C 7 12 aralkyl, C3-12heterocycle or C 4 -20heterocyclealkyl, CN, -OR, -NRR and -NRCOR; and R6 is selected from hydrogen, halogen and an unsubstituted or substituted Ci-salkyl; and wherein each occurrence of R is independently selected from an unsubstituted or substituted Ci.salkyl as hereinbefore defined, C6-1 2 arl, C 7 -1 2 aralkyl, C3-12heterocycle or
C
4 2 oheterocyclealkyl; with the provisos that: when R5 is -CONR 7 R, R 7 and Rs are not both hydrogen, (ii) R2a is not selected from -N(CH 3 2
-N(CH
2
CH
3 2 -OR, and an unsubstituted, straight chain or branched, non-cyclic, saturated C 1 3 alkyl, -N(CH 3 2
-N(CH
2
CH
3 2 and OR, (iii) when R 2 a is -Cl and R6 is R4a is not selected from -CF 3 -Cl, -CH 3 and
C(CH
3 3 (iv) when R2a is -C1, and both R4a and R6 are Rs is not -CH(CN)C 6
H
5 and when R2a is -Cl and R 4 a is -Cl, R6 is not selected from -Cl and -CH 2 Cl; and when 1 R 5 is -N(R 7 R2a is not selected from -CH 3
-OCH
3 and -N(CH 3 2 and (ii) Rs is not selected from -H and -CH 3 In another aspect, the invention provides a method for treating an inflammatory condition 9 00.6 *0 *in a warm blooded animal in need thereof comprising administering to said animal an 9 0 0 0effective amount of a compound having the structure:
RS
R4b N N R R2b including pharmaceutically acceptable salts thereof, wherein
R
5 is selected from the following chemical moieties: I N O 300 N 7 S\ N
R
7 R7\$
S
YI
I
P.'OPER\PDB\71631-96.claims.doc-14/09/00 -3B-
R
7 is selected from hydrogen, -CH 3 and -CH 2
C
6
H
5
R
8 is selected from hydrogen and an unsubstituted or substituted Cl- 8 alkyl, as hereinbefore defined, C6-12aryl, C7-12aralkyl, C3.12heterocycle and a C4- R2b is halogen; R4a is selected from hydrogen, halogen, -CN, and an unsubstituted or substituted Cig 8 alkyl as hereinbefore defined, C6-12aryl, C7-12aralkyl, C 3 12heterocycle or C4-20heterocyclealkyl; and R is selected from hydrogen,
-CH
3
-CF
3 and -CH 2
CH
3 S 10 with the proviso that when R 5 is -CONR 7
R
8 and R2b is -Cl, R4b and RI are not both hydrogen.
The invention also provides for the use of a compound as defined herein in the manufacture of a medicament for treating an inflammatory condition.
*n These and other aspects of this invention will become evident upon reference to the attached figures and the following detailed description.
o Brief Description of the Drawings Figure 1 illustrates a reaction scheme for the synthesis of representative pyrimidine-containing compounds of this invention.
Figure 2 illustrates a reaction scheme for the synthesis of representative pyrazinecontaining compounds of this invention.
WO 97/09315 PCT/US96/15108 Figure 3 illustrates the ability of a representative compound of this invention to inhibit the activation of NFicB and AP-1.
Figure 4 illustrates the ability of a representative compound of this invention to inhibit IL-2 and IL-8.
Figure 5 illustrates the ability of a representative compound of this invention to cause a dose-dependent suppression of alloantigen-induced
PLN
proliferation.
Detailed Description of the Invention As mentioned above, the compounds of this invention block activation of transcription factors (TFs), and thus have utility as anti-inflammatory agents in general, and in the prevention and/or treatment of a variety of conditions, including (but not limited to) immunoinflammatory and autoimmune diseases. The compounds are believed to function by inhibiting, at an early stage, transcription of deleterious proteins associated with such conditions or diseases. It is believed that this is achieved by inhibiting the kinase(s) that regulate the activation of TFs, such as NFicB and/or AP-1.
By disrupting the production of these activated TFs, synthesis of pathological proteins, including proinflammatory cytokines, associated with a series of immunoinflammatory and autoimmune diseases are effectively blocked at a transcriptional level. Accordingly, the compounds of this invention have activity in both the prevention and treatment of immunoinflammatory diseases such as rheumatoid arthritis, osteoarthritis and transplant rejection (tissue and organ), as well as autoimmune diseases such as multiple sclerosis.
The compounds of this invention are generally represented by the following general structure R4 N. B R2
(I)
wherein A is C-Re when B is N, and A is N when B is C-RI, and wherein R 1 R2, R4, Rs and R 6 are as defined below. Thus, when A is C-R 6 and B is N, structure is a pyrimidine-containing compound having structure and when A is N and B is C-RI, structure is a pyrazine-containing compound having structure (III): WO 97/09315 PCT/US96/15108 R4a-" R6 R 4
-O-N
N N N Rk NYN Ri R2a R2b (II)
(III)
In structures (II) and (III) above, Rs is selected from the following chemical moieties through (iv): OyN% SyN, R7\ N R7\ N R N N R 7 R S I I I (ii) (iii) (iv) wherein R7 is selected from hydrogen, -CH 3 and -CH 2
C
6 Hs; and
R
8 is selected from hydrogen and an unsubstituted or substituted
C
1 alkyl, C6- 1 2 aryl, C- 1 2 aralkyl, C 3 .1 2 heterocycle and a C4-16heterocyclealkyl.
The compounds of this invention further include pharmaceutically and prophylactically acceptable salts of compounds of structure Compounds of structure may contain proton donating groups a carboxylic acid group) and/or proton accepting groups a group with a nitrogen atom having a free lone pair of electrons, such as an amine group), and the salts of compounds of structure may be formed and utilized in the practice of the invention. Thus, compounds of the invention may be in the form of a base addition salt a salt of a proton donating group) or in the form of an acid addition salt a salt of a proton accepting group), as well as the free acid or free base forms thereof.
Acid addition salts of a free base amino compound of the invention may be prepared by methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include acetic, ascorbic, benzenesulfonic, benzoic, fumaric, maleic, methanesulfonic, and succinic acids. Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric and nitric acids. Base addition salts of a free acid carboxylic acid compound of the invention may also be prepared by methods well known in the art, and may be formed from organic and inorganic bases.
Thus, the compounds of this invention also include those salts derived from inorganic bases such as the hydroxide or other salt of sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like, and organic bases such as substituted ammonium salts.
As used herein, the above terms have the following meaning: A "C-.salkyl" is a straight chain or branched, cyclic or non-cyclic, saturated or unsaturated carbon chain containing from 1 to 8 carbon atoms. In one embodiment, the C-.salkyl is a fully saturated, straight chain alkyl selected from methyl, ethyl, n-propyl, n-butyl, n-pentyl and n-hexyl. In another embodiment, the Ci.salkyl is a fully saturated cyclic alkyl selected from (but not limited to) cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methylenecyclopropyl and methylenecyclohexyl. In still a further embodiment, the Cl.salkyl is a fully saturated, branched alkyl selected from (but ,oo not limited to) isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl and isohexyl. In yet a further embodiment, the Cl.salkyl is an unsaturated straight chain alkyl selected from :(but not limited to) ethylenyl, propylenyl, 1-butenyl, 1-pentenyl and 1-hexenyl.
A "C6 1 2 aryl" is an aromatic moiety containing from 6 to 12 carbon atoms. In one embodiment, the C6 12 aryl is selected from (but not limited to) phenyl, tetralinyl, and napthalenyl. In a preferred embodiment, the C.
12 aryl is phenyl.
A "C 7 1 2 aralkyl" is an arene containing from 7 to 12 carbon atoms, and has both aliphatic and aromatic units. In one embodiment, the C 7 1 2 aralkyl is selected 20 from (but not limited to) benzyl, ethylbenzyl, propylbenzyl and isobutylbenzyl.
A "C 3 1 2 heterocycle" is a compound that contains a ring made up of more than one kind of atom, and which contains 3 to 12 carbon atoms. In one embodiment, the C 3 1 2 heterocycle is selected from (but not limited to) pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyrrolidinyl, pyridinyl, pyrimidinyl and purinyl.
In a further embodiment, the C 3 1 2 heterocycle includes the following structures: o0 -N -N O
O
A "C4-2oheterocyclealkyl" is a compound that contains a C3- 1 2 heterocycle linked to L CI-_alkyl. In one embodiment, the C 4 20 heterocyclealkyl is a methylene furan having the ollowing structure: P \OPER\PDB\71631-96.cji,,ido.-IS/ 9 /0 -7- A "substituted" Cixsalkyl, C6-12arl,
C
7 -12aralkyl, C3-12heterocycle or C4-2oheterocyclealkyl is a C 18 alkyl, C6-12arl, C7-12aralkyl, C3-12heterocycle or C4-20heterocyclealkyl having one or more hydrogens replaced with a substituent selected from halogen (including -Cl, -Br and -OH, -OR, -COOH, -COOR,
-COR,
-CONH
2
-NH
2 -NHR, -NRR, -SH, -SR, -SOOR, -SO 3 R and -SOR, where each occurrence of R is independently selected from an unsubstituted or substituted CIsalkyl,
C
6 12 aryl, C 7 -12aralkyl, C3-12heterocycle or C420heterocyclealkyl as defined above. In one embodiment, the substituted C1salkyl is a CI.shaloalkyl including (but not limited to) -CF 3 and -C 2
F
5 In structure (II) above, R 2 a is selected from halogen, an unsubstituted or substituted
CI
8 alkyl, C6-12arl, C7-12aralkyl, C3-1 2 heterocycle or C4-20heterocyclealkyl, -CN, -OR, -NHR, H, -NHNH 2
-NHC
6
H
5 NRR and -NRCOR, wherein each occurrence of R is independently selected from an unsubstituted or substituted
C
1 8 alkyl, C6-12aryl,
C
7 -1 2 aralkyl, C 3 1 2heterocycle or C4-2heterocyclealkyl as defined above. In one embodiment,
R
2 a is selected from -C1, -CN and -CF 3 In structure (III) above, R2b is halogen, such as -Cl or -F.
In structure (II) above, R 4 a is selected from hydrogen, halogen, and unsubstituted or substituted C1-salkyl, C6-12ary1, C7-12aralkyl, C3-12heterocycle or C4-20heterocyclealkyl,
-CN,
-OR, -NHR, -NRR and -NRNCOR, wherein each occurrence of R is independently selected from an unsubstituted or substituted Ci.
8 alkyl, C6-12aryl, C7-12aralkyl, C3-12heterocycle or C4-2heterocyclealkyl as defined above. In one embodiment, R4a is selected from hydrogen,
-CH
3
-CF
3
-C
2
F
5
-C
6
H
5 and -CH 2
C
6
H
5 In structure (III) above, R4b is selected from hydrogen, halogen, -CN, and an unsubstituted or substituted CIsalkyl, C6-12arl, C7- 12 aralkyl, C3-12heterocycle or C4-20heterocyclealkyl.
In structures and (II) above, R 6 is selected from hydrogen, halogen, and an unsubstituted or substituted
C
18 alkyl, including (but not limited to) a C 1 8 haloalkyl (such as -CF 3 and -C 2
F
5 In one embodiment,
R
6 is selected from hydrogen -Cl, -CH 3 and
CF
3 In structures and (III) above, R 1 is selected from hydrogen, -CH 3
-CF
3 and
-C
2
H
5 SkALL, In one embodiment, the compounds of this invention have structure (II) above, wherein R 1 is the chemical moiety In this embodiment, the compounds disclosed herein have the following structure
(IV):
8 14 0 N, R7 R4 R6
NN
R2a
(IV)
where R2, R 6 R7 and R are as defined above. In a preferred ,embodiment, representative compounds of structure (IV) contain R2., R 4
R
6
R
7 and Rs moieties as identified in Table 1 below.
Table 1 Compounds of Structure (IV) R2. R4. R6 R7 Rs -Cl -CF3 -H
-H
-OCH
3 -CI
-CF
3
-CH
3 z 0 -H -F -CH 3 -N(CHa)2
-CH
3 -Cl
-CF
3
-H
-CN
-C
2
F
-NHNH2 -NHPh wherein X, Y and Z are the same or different, and independently selected from hydrogen, -OH, -OR, -COOH, -COOR, -COR, -CONH 2
NH
2
-NM
-NRR, -SH, -SR, -SOOR, -SO 3 R and -SOR, where each occurrence of R is independently selected from an unsubstituted or substituted
C
4 .alkyl,
C&
12 aryl, C7.- 2 aralkyl, C3-12heterocycle or C4-2heterocyclealkyl.
4 WO 97/09315 WO 9709315PCT/US96/15108 9 In a preferred embodiment of the compounds disclosed in Table I above, X, Y and Z are the same or different, and independently selected from -Cl, -CF 3 -OH, -CH 3 and -OCH 3 In a further preferred embodiment, R8 is a bis(trifluoromethyl)phenyl moiety or a 3-trifluoromethyl-5-halo-phenyl moiety.
As mentioned above, in one embodiment of this invention the compounds have structure Within one aspect of this embodiment, R 4 is -CF 3 and R 2 is -CI.
Such compounds include (but are not limited to): 2-chloro-4-trifluoromethyl-5-N-(3',5'bistrifluoromethylphenyl)pyrimidine carboxamide; 2-chloro-4-trifluoromethyl-5-N-(3 dichlorophenyl)pyrimidine carboxamide; 2-chloro-4-trifluoromethyl-5-N-(4'trifluoromethylphenyl)pyrimidine carboxamide; 2-chloro-4-trifluoromethyl-5-N- (phenyl)pyrimidine carboxamide; 2 carboxamide; 2-chloro-4-trifluoromethyl-5-N-(3',4,5'-trichlorophenyl)pyrimidine carboxamide; 2-chioro-4-trifluoromethyl-5-N-(benzyl)pyrimidine carboxamide; 2-chloro- 4-trifluoromethyl-5-N-(4'-(2', 1',3'-benzothiadiazole))pyrimidine carboxamide; 2-chloro- 4-trifluoromethyl-5-N-(3',5'-dichloro-6'-hydroxyphenyl)pyrimidine carboxamide; 2chloro-4-trifluoromethyl-5-N-(5'-(3'-methylisoxazole))pyrimidine carboxamide; 2chloro-4-trifluoromethyl-5-N-(3'-N-acyl-4'-fluoroaniline)pynimidine carboxaunide; 2chloro-4-trifluoromethyl-5-N-(3-trifluoromethyl-5'-ethoxycarbonylphenyl)pyrimidine carboxaniide; 2-chloro-4-trifluoromethyl-5-N-(3'-trifluoromethyl-5- (carboxaniide)phenyl) pyrimidine carboxamide; 2-chloro-4-trifluoromethyl-5-N-(3 dichlorophenyl)-N-(methyl)pyrimidine carboxamide; and 2-chloro-4-trifluoromethyl-5carboxamide.
Within another aspect of this embodiment, R 4 a is -CF 3 and R2. is a moiety other than -Cl. Such compounds include (but are not limited to): 2-fluoro-4tfifluoromethyl-5-N-(3', 5'-bistfifluoromethyl)pyrimidine carboxamide, 5-(3 bis(trifluoromethyl)phenacyl)-2-methoxy-4-trifluoromethylpyrimidine; 4-trifluoromethyl- 5-N-(3',5'-dichlorophenyl)pyrimidine carboxamide; 2-dimethylamino-4-tfifluoromethyl- 5'-dichlorophenyl)pyrimidine carboxamide; 2-triethylammonium chloride-4trifluoromethyl-5-N-(3', 5'-dichlorophenyl)pyrimidine carboxamide; 2-cyano-4trifluoromethyl-5-N-[3', 5'-(bistrifiuoromethyl)phenyl]pyrimidine carboxamide; 2hydrazino-4-trifluoromethyl-5-[N-(3 5'-dichlorophenyl)pyrimidine-5-carboxamide; 2-[N- (1 -Aminocitraconamide)]-4-trifluoromethyl-5-[N-(3', 5'-dichlorophenyl) carboxaniide; and 2-aniinophenyl-4-trifluoromethyl-N-(3,5'-dichlorophenyl)pyrimidine- WO 97/09315 PCTIUS96/15108 Within yet a further aspect of this embodiment, R2, is -CI and RP 4 is a moiety other than -CF 3 Such compounds include (but are not limited to): bis(trifluoromethyl)phenyl)-2,4-dichloro-6-methyl-pyrimidine carboxamide; 2-chloro-4methyl-5-N-(3',5'-(bistrifluoromethyl)phenyl)pyrimidine carboxamide; 2,4-dichloro-5-N- (3',5'-bis(trifluoromethyl)benzyl)pyrimidine-5-carboxamide; and 2 carboxamide.
In another embodiment, the compounds of this invention have structure (III) above. Within one aspect of this embodiment, R 1 is selected from hydrogen, -CH 3 and -CF 3 Such compounds include (but are not limited to) pyrazine-containing compounds which correspond to the pyrimidine-containing compounds disclosed above.
In one embodiment of structure (III), R2b is -Cl, R4b is -CF 3 and R5 is a moiety of structure above.
A small number of compounds which fall within structure above have been previously disclosed and/or are commercially available. However, such compounds have not been associated with the utilities of the present invention, or possess no recognized utility. Accordingly, compounds that fall within the scope of structure and which have recognized utility, are specifically excluded from the novel compounds of structure However, to the extent such compounds have not been disclosed for the utilities of the present invention, they are included in the various methods of this invention.
To this end, the novel compounds of this invention do not include compounds of structure (IV) above where R7 and Rs are both hydrogen, and where R2.
is selected from an unsubstituted, straight chain or branched, non-cyclic, saturated Ci.3 alkyl -CH 3
-CH
2
CH
3
-(CH
2 2
CH
3 and -CH(CH 3 2
-N(CH
3 2
-N(CH
2
CH
3 2 and -OR, where R is as defined above. Similarly, the novel compounds of structure (IV) are subject to the following provisos: when R 2 is -Cl and R 6 is R4, is not -CF 3 -Cl,
-CH
3 or -C(CH 3 3 (b)when R2Z is -Cl and both R4, and RP are RP is not
-CH(CN)CH
5 or -(CH 2 5
CH
3 and when R2, is -Cl and R 4 is -CI, R 6 is not -Cl or
-CH
2
C.
The novel compounds of this invention also do not include compounds of structure (II) when Rs is moiety (iii) and R2 is -CH 3
-OCH
3 or -N(CH 3 2 or Rs is -H or -CH 3 Furthermore, the novel compounds of structure (III) when Rs is moiety are subject to the following proviso: when R2b is -CI, R4b and R, are not both hydrogen.
WO 97/09315 PCTIUS96/15108 11 The compounds of this invention may be made by one skilled in organic synthesis by known techniques, as well as by the synthetic routes disclosed herein. For purpose of convenience, the compounds have been separated into pyrimidine-containing (structure and pyrazine-containing (structure (III)) compounds as set forth below.
The pyrimidine-containing compounds of this invention may be prepared as illustrated by the reaction scheme of Figure 1. In general, commercially available f-keto esters 1 are heated at elevated temperatures (75-110 0 C) with a mixture of urea and triethylorthoformate (or a substituted orthoformate) to provide ureido derivatives 2.
Treatment of these intermediates with sodium alkoxides, such as sodium ethoxide in an alcoholic solvent at 35-100 0 C, gives 2-hydroxypyrimidine esters 3 which, upon treatment with a chlorinating agent such as phosphorous oxychloride at elevated temperatures (75-120 0 yields 2-chloropyrimidine esters 4. The 2-hydroxypyrimidine esters 3 may also be treated with a mild base, such as lithium hydroxide, sodium hydroxide or potassium carbonate to provide the corresponding acid 3A, which may then be converted with a chlorinating agent, such as phosphorous oxychloride or thionyl chloride in an inert solvent or neat at 25-75 0 C, to the acid chloride 5. Compounds of structure 6 may be prepared using standard conditions known in the art by reacting the acid chloride 5 with an amine in the presence of a base, such as potassium carbonate or dimethylaminopyridine (DMAP), in a non-protic solvent, such as methylene chloride or EtOAc at 25-40 0 C, followed by standard workup.
Alternatively, pyrimidine-containing compounds of this invention may also be made by the following combinatorial procedure. Commercially available and/or readily synthesized amines, anilines and related compounds may be reacted with the acid chloride 5 in EtOAc in the presence of basic Amberlyst 21 resin. The reactions are quenched with 50 uL of water and the final products are obtained in the organic layer and concentrated. This procedure may be done in a 96 well (1 mL deep well) plate and the final products isolated as dry powders. TLC analysis is performed on each compound and indicates the purity, and GC/MS and HPLC analysis demonstrates that the desired products are synthesized (mass spectral analysis, molecular weight) and are greater than 80% pure. By this method, eighty distinct pyrimidine-containing compounds may be routinely synthesized at the same time in one 96 well plate.
In addition, compound 4 may be reacted with various nucleophiles in an aprotic solvent and at ambient temperature to provide derivatives 7. These compounds can be hydrolyzed with base to yield compounds having structure 8. Compounds of structure 8 can be converted to the acid chloride as described above, and reacted with various amines to give compounds having structure 9 using known conditions, including WO 97/09315 PCT/US96/15108 12 the combinatorial approach described above. Alternatively, compounds of structure 7 can also be prepared by reacting the P-keto ester 1 in a sequential fashion with triethylorthoformate and acetic anhydride or N,N-dimethylformamide dimethyl acetal in DMF to give intermediate 10. Reacting intermediate 10 with a variety of amidines in alcoholic solvents provides intermediate 11 which, upon addition of base, provides compounds of structure 7.
Pyrazine-containing compounds of structure (III) may be prepared as illustrated by the reaction scheme of Figure 2. The synthesis of these compounds may begin with readily available pyruvic acid derivatives 12. These compounds are condensed with commercially available 2-cyano-l,2-diamino-2-substituted ethenes 13 in an alcoholic solvent (such as MeOH) in the presence of an acid (such as HCI) at ambient temperatures (25-60 0 C) to provide the cyano pyrazines of structure 14. The pyrazines may then be converted to the corresponding carboxylic acids 15 using a strong base such as sodium hydroxide in water, or a strong acid such as HCI, at elevated temperatures (70-110°C). These carboxylic acids may then be converted to 5-chloro-2-carbonyl acid chloride derivatives 16 using a chlorinating agent such as POCI 3 or SOC1 2 Treatment of 16 with various amines or anilines at ambient temperatures in an inert solvent such as EtOAc or CH 2 C1 2 provides compounds of structure 17.
The carboxylic acids of structure 15 can also be converted to the hydroxy ester 18 by treatment with SOC1 2 and MeOH at a temperature of 25-60 0 C Treatment of 18 with a chlorinating agent such as SOC12 or POCI 3 in the presence of DMF gives the chloro ester 19. Compound 19 can also be converted to the acid chloride 16 using a mild base such as potassium carbonate in an a protic solvent such as MeOH, followed by treatment with a chlorinating agent such as oxalyl chloride in an inert solvent such as methylene chloride at ambient temperatures.
The pyrazine-containing compounds of this invention may also be synthesized by appropriate combinatorial techniques as described. In short, commercially available and/or readily synthesized amines, anilines and related compounds may be reacted with the acid chloride 16 in EtOAc in the presence of basic Amberlyst 21 resin. The reactions are quenched with 50 jiL of water and the final products are obtained in the organic layer and concentrated. This procedure may be done in a 96 well (1 mL deep well) plate and the final products isolated as dry powders.
TLC analysis is performed on each compound and indicates the purity, and GC and HPLC analysis demonstrates that the desired products are synthesized (mass spectral analysis, molecular weight) and are greater than 80% pure. By this method, eighty pyrazine-containing compounds may be routinely synthesized in one 96 well plate.
WO 97/09315 PCT/US96/15108 13 Once synthesized, the compounds of this invention may be formulated for administration to a warm-blooded animal by a variety of techniques known to those skilled in the art. In one embodiment, the compound is in the form of a pharmaceutical composition for prophylactic or therapeutic use, and which contains at least one compound of this invention in combination with a pharmaceutically acceptable carrier or diluent. The compound is present in the composition in an amount which, upon administration to the animal, is effective in preventing or treating the condition of interest. Preferably, the composition includes a compound of this invention in an amount ranging from 0.01 mg to 250 mg per dosage, depending upon the route of administration, and more preferably from 1 mg to 60 mg. Appropriate concentrations, dosages and modes of administration may be readily determined by one skilled in the art.
Suitable carriers or diluents are familiar to those skilled in the formulation field. For compositions formulated as liquid solutions, acceptable carrier or diluents include saline and sterile water, and may optionally include antioxidants, buffers, bacteriostats and other common additives. The compositions of this invention may also be formulated as pills, capsules, granules or tablets which contain, in addition to the compound of this invention, diluents, dispersing and surface active agents, binders and lubricants. One skilled in the art may further formulate the compounds of this invention in any appropriate manner, and in accordance with accepted practices, such as those disclosed in Remington's Pharmaceutical Sciences, Gennaro, Ed., Mack Publishing Co., Easton, PA, 1990 (incorporated herein by reference).
In another embodiment, the present invention provides methods for preventing or treating a variety of conditions. Such methods include administering a compound of this invention to a warm-blooded animal in need thereof in an amount sufficient to prevent or treat the condition. Such methods include systemic administration of a compound of this invention, preferably in the form of a composition as disclosed above. As used herein, systemic administration includes oral and parental methods of administration. For oral administration, suitable pharmaceutical compositions include powders, granules, pills, tablets and capsules, as well as liquids, syrups, suspensions and emulsions. These compositions may also include flavorants, preservatives, suspending, thickening and emulsifying agents, and other pharmaceutically acceptable additives. For parental administration, the compounds of the present invention may be prepared in aqueous injectable solutions which may contain, in addition to the compound of this invention, buffers, antioxidants, bacteriostats and other additives commonly employed in such solutions.
WO 97/09315 PCT/US96/15108 14 As mentioned above, compounds of the present invention can be used to prevent or treat a wide variety of disorders, diseases and/or illnesses. In particular, the compounds may be administered to a warm-blooded animal for prevention or treatment of rheumatoid arthritis, osteoarthritis, tissue and/or organ transplant rejection, sepsis, ARDS, asthma, trauma, oxidative stress, cell death, irradiation damage, ischemia, reperfusion, cancer, viral infection, and autoimmune diseases such as psoriasis, inflammatory bowel disease, glomerulonephritis, lupus, uveitis and chronic hepatitis.
Compounds of this invention may be screened by known and accepted techniques for their ability to function as prophylactically and/or therapeutically active agents. For example, the compounds may be evaluated in in vitro and/or in vivo assays indicative of the compound's antinflammatory and immunosuppressive properties. To this end, such compounds may first be evaluated in a number of cell-based assays which determine the ability of a compound to prevent activation of NFcB and AP-l(see Example 56). Next, the compound's ability to attenuate cytokine levels (such as IL-2 and IL-8), which are known to be elevated in certain disease states, may be determined (see Example 57). The compounds may then be evaluated in an appropriate animal model, including rodent models of inflammation and immunosuppression (see Example 58).
It should be recognized that, for example, in the case of immunosuppressive drugs and other agents which have utility for the treatment of rheumatoid arthritis numerous studies have been performed directed to the activity of such drugs. To this end, cyclosporin A has been used in clinical trials since the late 1970's as a second-line drug and is recommended to be used only in patients with active RA. Thus, Experiment 58 was performed utilizing cyclosporin A as a positive control.
A recent review of such immunosuppressive drugs, including relevant assays for the same, is presented by R.P. Carlson in Exp. Opin. Invest. Drugs 4(9):853-859, 1995 (incorporated herein by reference in its entirety, including cited references).
The following examples are presented for purpose of illustration, not limitation.
EXAMPLES
To summarize the examples that follow, Examples 1-54 disclose the synthesis of representative compounds of this invention, as well as intermediates thereof; Example 55 discloses the synthesis of representative compounds by combinational chemistry techniques; Examples 56-57 disclose the ability of representative compounds of this invention to inhibit NFKB, AP-1 and cytokines; and Example 58 discloses the WO 97/09315PCJS6150 PCT/US96/15108 activity of a representative compound of this invention in both graft versus host disease and contact sensitivity models.
Example 1 2-CHLORO-4-TRIFLUORoMETHYL-5-N-.
5'-BIS(TRIFLUOROMETHYL)PHENYL]PYRIM1NE
CARBOXAMIDE
To a mixture of 3,5-bistrifluoromethylaniline (0.20 g, 0.92 mmol), Amberlyst A-21 ion exchange resin (0.02 g) in EtOAc (5 rnL) was added a solution of 2chloride (0.27 g, 1. 13 mmol) in EtOAc (5 mL). The mixture was stirred for 0.5 h, then quenched with water (0.20 niL). The organic layer was separated, dried over MgSO 4 filtered and the solvent removed under reduced pressure. The resulting oil was recrystallized from EtOHIH 2 O to provide the title compound (0.21 g, 53% yield) as a white solid; m.p. 162-163 0
C.
Example 2 2-CHmoRo-4-TRmLuoRo~NErHYL-5-N- (4'-TRIFLUOROMETHYLPHE-NYL)PYRMID1N
CARBOXAMIDE
The title compound was prepared as described in Example 1, but employing 4-trifluoromethylaniline 1 g, 0.41 mmol) in place of bistrifluoromethylaniline and the acid chloride 10 g, 0. 41 mmol), resulting in a 24% yield; m.p. 172-173'C.
Example 3 2-CHLoRo-4-RILUORONETYL- 5-N-(PHENYL)PYRimiDINE cARBoxAmiDE The title compound was prepared as described in Example 1, but employing aniline (0.04 g, 0.39 mmol) and the acid chloride (0.22 g, 0.90 mmol), resulting in a 62% yield; m.p. 108-181'C.
Example 4 2-CHLoRo4-TLUORONTHnr-YL-
CARBOXAMIDE
The title compound was prepared as described in Example 1, but employing cyclohexylamine (0.02 g, 0.18 mmol) and the acid chloride (0.05 g, 3 5 0.22 mmol), resulting in a 3 3% yield; m.p. 150-15 1 0
C.
WO 97/09315 WO 9709315PCT/US96/1 5108 16 Example 2-CHLoRo-4-TRIFLUOROMTHYL-
CARBOXAMIDE
The title compound was prepared as described above in Example 1, but employing benzylamnine (0.09 g, 0.92 mmol) and the acid chloride (0.25 g, 1.0 mmol), resulting in a 78% yield; m.p. 152-153*C.
Example 6 2-CBLoRo-4-TRiFLuoRoM ETHYL-5-N-(3',4',5'- TRICHLOROPH-ENYL)PYRIMIDINE
CARBOXAMIDE
The title compound was prepared as described in Example 1, but employing 3,4,5-trichioroaniline 15 g, 0.61 mmol) and the acid chloride 15 g, 0. 61 mmol), resulting in a 5 5% yield; m. p. 200-201 0
C.
Example 7 2-CHLoRo-4-TRIFLuoRo~vITHYL-5-N-(4-(2', I ,3' -BENZOTHIADIAZOLE))PYRIMIDINE
CARBOXAMIDE
The title compound was prepared as described above in Example 1, but employing 4-amino-2,1,3-benzothiadiazole (0.01 g, 0.07 mmol) and the acid chloride (0.025 g, 0. 10 mmol), resulting in a 60% yield; m.p. 179-180'C.
Example 8 2-CHiLoRo-4-TRIFLuoRoMETJ-L-5-N-(3 6'-HYDRoxYPHENYL)PYRimiMDmN cARBoxAmID The title compound was prepared as described in Example 1, but employing 3,5-dichloro-6-hydroxyaniline (0.02 g, 0. 11 mmol) and the acid chloride (0.04 g, 0. 16 mmol), and purified by chromatography (SiO 2 1:1 hexanes/EtOAc) to provide the compound in a 10% yield; m. p. 21 1-213'C.
Example 9 2-CHLoRo-4-TRIFLUOROMTHYL-5-N-[5'-(3'-NcTHyIL ISOXAZOLE)]PYRIMDINE CARBOXAMIDE The title compound was prepared as described in Example 1, but employing 5-amino-3-methylisoxazole (0.02 g, 0. 17 mmol) and the acid chloride (0.03 g, 3 5 0. 10 mmol), resulting in a 75% yield; m. p. 170-171IT.
WO 97/09315 PCT/US96/15108 17 Example 2-CHLoRO-4-TRIFLUOROMETHYL-5-N-(3'-N-ACYL- 4'-FLuoRo-ANILNE)PYRIMIiNE
CARBOXAMIDE
A solution of 2-fluoro-5-nitroaniline (1.97 g, 12.60 mmol) and a 1:1 mixture of Ac 2 0/pyridine (20 mL) was stirred for 18 h. The resulting precipitate was filtered and washed with MeOH to provide The N-acyl-2-fluoro-5-nitroaniline (0.99 g, 5.00 mmol) was dissolved in EtOH (25 mL), and then 10% Pd/C (0.12 g) was added and the solution stirred under H 2 for 5 h. The suspension was filtered through celite and the filtrate evaporated to dryness. The resulting oil was chromatographed (SiO 2 1:3 hexanes/EtOAc) to provide 3-N-acyl-4-fluoro-aniline as a yellow oil. The aniline derivative was then coupled to 2chloride as described in Example 1 to provide the title compound in a 47% yield; m.p. 126-127 0
C.
Example 11 2 -CHLORO-4-TRFLUOROETHYL-5-N-(3'-TR
FLUOROETHYL-
PYRIMIDINE CARBOXAMIDE To a solution of 3 -nitro-5-trifluoromethylbenzoic acid (1.00 g, 4.25 mmol) in CH 2
CI
2 (50 mL) was added oxalyl chloride (1.45 g, 13.8 mmol) followed by DMF (3 drops). An immediate evolution of gas occurred and the reaction was stirred for 18 h. The solvent was removed under reduced pressure, the resulting oil was dissolved in THF (80 mL) and cooled to 0 0 C. To the cold solution, NH 4 0H (22 mL) in THF (15 mL) was added dropwise and the mixture was stirred 18 h at room temperature. The mixture was concentrated to remove the THF and the resulting precipitate was filtered and dried. The solid was dissolved in EtOH (25 mL) and Pd/C (0.12 g) was added, and the suspension was stirred 15 h under a blanket of H 2 The reaction was filtered through celite, and the filtrate evaporated to dryness to provide 3 -carboxamide-5-trifluoromethylaniline as a yellow oil. This compound was then coupled to 2-chloro-4-trifluoromethylpyrimidine-5-carbonyl chloride as described in Example 1 to provide the title compound in a 55% yield; m.p. 218-219 0
C.
Example 12 2-CHLoRo-4-TRFLuoRoMETHYL-5-N-(3'-TRIFLUOROMETHYL- PYRIMIDINE CARBOXAMIDE To a solution of 3-nitro-5-trifluoromethylbenzoic acid (0.36 g, 1.53 mmol) in CH 2
C
2 (20 mL) was added oxalyl chloride (0.58 g, 4.60 mmol) followed by WO 97/09315 PCT/US96/15108 18 DMF (3 drops). An immediate evolution of gas occurred and the reaction was stirred for 18 h. The solvent was removed under reduced pressure, the resulting oil was dissolved in THF (80 mL) and cooled to 0°C. To the cooled solution was added EtOH mL) in THF (15 mL) and the mixture was stirred for 18 h at room temperature. The mixture was concentrated to remove the THF and the resulting precipitate was filtered and dried. The solid was dissolved in EtOH (25 mL) and 10% Pd/C (0.12 g) was added and the suspension was stirred for 15 h under a blanket of H 2 The reaction was filtered through celite and the filtrate evaporated to dryness to provide trifluoromethylaniline as a yellow oil. This compound was then coupled to 2-chloro-4trifluoromethyl pyrimidine-5-carbonyl chloride as described above to provide the title compound in a 12% yield; m.p. 67-71°C.
Example 13 2-CHLORO-4-TRIFLUOROMETHYL-5-N-(3',5'-DICHLOROPHENYL)- 5-N-(METHYL)PYRIMIDINE
CARBOXAMIDE
To a solution of 2 -chloro-4-trifluoromethyl-5-N-(3,5-dichlorophenyl)pyrimidine carboxamide (0.086 g, 0.23 mmol) in DMF (20 mL) was added NaH (0.02 g, 0.53 mmol). The mixture was stirred for 0.3 h at room temperature and then Mel (0.100 mL, 1.61 mmol) was added and stirring continued for 2 h. The solution was acidified with 2N HCI and then extracted with EtOAc The combined organic layers were dried over MgSO 4 filtered and the solvent removed under reduced pressure. The resulting oil was chromatographed (SiO 2 7:1 hexanes/EtOAc) to provide the title compound yield) as a white solid; m.p. 124-125 0
C.
Example 14 2-CHLORO-4-TRIFLUOROMETHYL-5-N-(3',
CARBOXAMIDE
A mixture of benzaldehyde (1.04 g, 9.40 mmol), 3,5-dichloroaniline (1.71 g, 10.60 mmol), and HOAc (0.20 mL) in MeOH (35 mL) was cooled to 0°C. Then a solution of NaBH 3 CN (28.0 mL, 28.0 mmol, 1.0 M solution in THF) was added dropwise via a syringe pump over 0.25 h. The solution was allowed to stir an additional 0.3 h at 0°C, and then room temperature for 18 h. The excess NaBH 3 CN was quenched with HCI and the solvent was removed under reduced pressure. The resulting oil was dissolved in EtOAc/H 2 0, basified with NaOH, and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO 4 filtered and the solvent removed under reduced pressure. The resulting oil was purified by WO 97/09315 PCT/US96/1 5108 19 chromatography (SiO 2 15:1 hexanes/EtOAc) to provide N-benzyl-3,5-dichloroaniline as a white solid. This compound was coupled to 2 carbonyl chloride as described and purified by chromatography (SiO 2 9:1 hexanes/EtOAc) to provide the title compound (15% yield) as a white foam; m.p. 102- 104 0
C.
Example 2 4 -DicHLoRo-6-ETHyPYjamff cARBoxAmIID 5-Carbethoxy-6-methyluracil was prepared as reported in the literature (Lamon, J1 Het. Chem., 261, 1969); m.p. 180-182'C. The ethyl ester was then hydrolyzed as described for 2 -hydroxy4-methylpyrimidine5carboxyic acid to provide 2 4 -dihydroxy-6-methylpytimidine-5-carboxylic acid in a 95% yield; m.p. >230'C.
The 2,4-dihydroxy-6-methyl pyrimidine-5-carboxylic acid was heated at reflux with POC1 3 The reaction mixture was concentrated and 2,4-dichloro-6chloride was obtained by distillation 70-80'C, mm/Hg). The 2 4 -dichloro-6-methylpyrimidine5carbonyI chloride 15 g, 0.67 mmol) was immediately reacted with 3 ,5-bis(trifluoromethyl)aniline (0.15 g, 0.67 mmol) in a similar manner to that described in Example I to provide the title compound (0.06 g, 24% based upon starting 2 4 -dihydroxy-6-methylpyrimidine-5-carboxyiic acid); m.p.
174-1 76 0
C.
Example 16 2 4 -DicHoRopY~imim5.CARONYL
CHLORIDE
The title compound was prepared as described in the literature (Smith and Christensen, J Org. Chem. 20:829, 1955) starting from 2 4 carboxylic acid. The compound was obtained by distillation; b.p. 90-100TC mm/Hg) in a yield of 46%; 'HNMR (CDCI 3 8 9.29.
Example 17 ETHYL UREIDOMETHYLENE
ACETOACETATE
A mixture of ethyl acetoacetate (200 g, 1.54 mol), urea (105 g, 1.54 mole) and triethyl orthoformate (228 g, 1.54 mol) was heated at 140'C under N 2 for 22 h. The reaction mixture was cooled and filtered to provide the title compound in a 51% yield (156 m.p. 173-174*C.
WO 97/09315 PCT/US96/15108 Example 18 ETHYL UREIDOMETHYLENE BENZOYLACETATE The title compound was prepared as described in Example 17, but employing ethyl benzoylacetate (30 g, 156 mmol), resulting in a yield of 21% (12 g); m.p. 124-126 0
C.
Example 19 ETHYL 2-HYDROXY-4-METHYLPYRIMIDINE-5-CARBOXYLATE A solution of ethyl ureidomethylene acetoacetate (50 g, 250 mmol) NaOEt (22.1 g, 325 mmol) in EtOH (500 mL) was stirred at room temperature under N 2 for 3 days. The resulting solid was filtered and dried to yield the title compound as a sodium salt in a yield of 88% (45 m.p. >220 0 C (dec.).
Example ETHYL 2-HYDROXY-4-PHENYLPYRMIDINE-5-CARBOXYLATE The title compound was prepared as described in Example 19, but employing ethyl ureidomethylene benzoyl acetate (12 g, 45 mmol), resulting in a yield of (6 m.p. >260 0 C, (dec.).
Example 21 ETHYL 2-CHLORO-4-METHYLPYRIMIDINE-5-CARBOXYLATE A solution of ethyl 2-hydroxy-4-methylpyrimidine-5-carboxylate (5 g, 27.5 mmol) and POCI 3 (84 g, 550 mmol) was heated at reflux under N 2 for 1 h. The reaction was cooled and concentrated. The residue was partitioned between CHCI 3 and
H
2 0 and the organic layer was dried (Na 2
SO
4 filtered, and concentrated to yield the title compound in a yield of 27% (1.5 'HNMR (CDCI 3 5 9.04 1H), 4.42 2H), 2.85 3H), 1.43 3H).
Example 22 ETHYL 2-CHLORO-4-PHENYLPYRIMIDINE-5-CARBOXYLATE The title compound was prepared as described in Example 21, but employing 2-hydroxy-4-phenylpyrimidine-5-carboxylate (6 g, 25 mmol) to give the title compound (5.5 g, m.p. 45-47 0
C.
WO 97/09315 PCT/US96/15108 21 Example 23 2-CLR-4-METHYLPYRIMMINE-5-CARBOXYLIC ACID A solution of ethyl 2-chloro-4-methylpyrimidine-5-carboxylate (1.0 g, mmol), NaOH (0.24 g, 6 mmol) in H20 (30 mL) was stirred at room temperature for 3 h. The solution was acidified with 6N HCI and the resulting solid was filtered and dried to give the title compound (0.67 g 'HNMR (DMSO-d) 8 9.01 1H), 2.75 (s, 3H).
Example 24 2-CHLORO-4-PHENYLPYRIMIDINE-5-CARBOXYLIC ACID The title compound was prepared as described in Example 23, but employing 2-chloro-4-phenylpyrimidine-5-carboxylate (4.5 g, 17 mmol), resulting in a yield of 87% (3.9 m.p. 105-110 0
C.
Example 2-CHLORO-4-METHYLPYRIMIDNE-5-ARBONYL CHLORIDE A solution of 2-chloro-4-methylpyrimidine-5-carboxylic acid (0.81 g, 4.70 mmol), oxalyl chloride (0.89 g, 7.05 mmol), DMF (2 drops) in CH 2
I
2 (23 mL) was stirred at room temperature under N 2 for 4 h. The solution was concentrated and distilled to give the title compound (0.55 b.p. 90-100 0 C, 1.3 mm/Hg; 'HNJMR (CDCl 3 5 d 9.02 1H), 2.74 3H).
Example 26 2-CHLoRo-4-PHENYLPYRDINE-5-CARBONYL CHLORIDE The compound was prepared as described above in Example 25, but employing 2-chloro-4-phenylpyrimidine-5-carboxylic acid (3.8 g, 14 mmol), resulting in a yield of 53 m.p. 42 0
C.
Example 27 The compound was prepared as described in the literature (see, Arukwe, J. Undheim, K. Acta Chemica Scand. B40:764, 1986).
WO 97/09315 PCT/US96/I 5108 22 Example 28 ETHYL ETHOXYMETHYLENE-4,4,4-TRIFLUOROACETOACETATE A solution of 4 ,4,4-trifluoroacetoacetate (46 g, 0.25 mol) triethyl orthoformate (74 g, 0.50 mol) and AC 2 0 (77 g, 0.75 mol) was heated at 120-140 0 C for 7 h. The mixture was concentrated and distilled to give the title compound in a 98% yield (58.6 b.p. 80-90'C, 1.5 mm/Hg.
Example 29 2 4 -Bis(RLuoRo myL)P ysimiEcA~oNYL CiHLORIDE A solution of ethyl etoyehln444trfuraeoctt (15 g, 62.5 mmol) and trifluoroacetamidine (12.6 g, 112.5 mmol) in EtOH (50 mL) was heated at reflux for 24 h under N 2 The reaction mixture was cooled and concentrated.
Chromatography (SiO 2 20% EtOAc/hexane) afforded ethyl-2,4-bis as an oil (7.0 g, 39 'HNMR (CDCI 3 9.37 IH), 3.70 2H), 1.27 3H).
A solution of ethyl- 2 4 -bis(trifluoromethyl)pyrimidine-5-carboxylate g, 17 mmol) and NaGH (0.72 g, 18 mmol) in EtOH (20 mL) and H20 (50 mL) was stirred at room temperature for I h. The solution was acidified (HCI) and the resulting solid was filtered and dried to give 2,4-bis (trifluoromethyl)-pyrimidine5carboxylic acid (1.5 g, m.p. 59 0 C, 'HINMR (DMSO-d 6 8 9.62 IH).
The desired acid chloride was obtained from 2,4-bis(trifluoromethyl)acid in a manner similar to that described in Example 25 in a yield of 44%; b.p. 105 0 C (1.5 mm/Hg); 'HNMR (CDCl 3 8 9.12 IFH).
Example 2-Hoo4TILOOEHLYiim5CROYI
ACID
A solution of 2 -chloro- 4 -trifluoromethylpyrimidine-5-.carbonyI chloride (2.1 g, 8.6 mmol) in H 2 0 (10 mL) was stirred at 0 0 C under N 2 for 0.5 h. The resulting solid was filtered and dried to give the title compound (1.91 g, 98% yield); m.p. 232- 234'C (dec.).
Example 31 2-Y~--RFURNTYP~~N--A~~~Hom To a solution of 2 -chloro- 4 -trifluoromethylpyrimidines...carboxyic acid (2.80 g, 12.4 mmol) in TIHE (50 mL) at 0 0 C. was added Me 3 ;N (bubbled for 5 minutes).
The reaction was kept at 0 0 C for 0.25 h and the resulting solid was filtered to provide 2- WO 97/09315 PCTIUS96/15108 23 trimethylammonium chloride-4-trifluoromethylpy iridine-5-carboxylic acid (3.40 g, 97% yield); m.p. 120-1216C (dec.).
A solution of 2-trimethylammonium chloride-4acid (3.62 g, 12.7 mmol) and KCN (0.99 g, 15.2 mmol) in DMF (36.5 mL) and H 2 0 (18.3 m.L) was stirred at room temperature under N 2 for 0.25 h. The reaction mixture was concentrated and dissolved in EtOAc (400 mL).
The EtOAc layer was washed with 1120 (4 X 100 brine (100 mL) and dried (Na 2 SO4. The EtOAc layer was filtered and concentrated to yield 2-cyano-4acid (2.03 g, 74% yield); m.p. 148-149'C (dec.).
A solution of 2 -cyano- 4 -trifiuoromethylpyrimidine5carb 0 ,xjj 0 acid g, 9.2 mmol), oxalyl chloride (1.4 g, 11I mmol) and DMff (4 drops) in CH 2
CI
2 (46 mL) was stirred at room temperature under N 2 for 0.75 h. The reaction was concentrated and distilled 100 0 C, 1.5 mm/Hg) to give the title compound (1.8 g, 82% yield); 'HNMvR (CDCI 3 8 9.49 1H).
Example 32 2 -PHENYLPYRim1DINE,-5-cARBoNYL cHLoRiDE A solution of ethyl 3 -N,N-dimethylamino-2-formylacrylate (4.0 g, 23 mniol) (Arnold, Coll. Czech. Chem. Commun. 26:3051, 1961), benzamnidine hydrochloride (4.0 g, 26 mmol) and sodium (0.65 g, 28 mmol) in EtOH (40 mL) was heated at reflux for 1 h. The solution was filtered and concentrated and the residue partitioned between EtOAc and dilute HCI The organic layer was dried (Na 2 SO4, and concentrated to give ethyl 2 -phenylpyrimidine-5-carboxylate (4.0 g, yield); m.p. >220 0 C (dec.).
The corresponding 2 -phenylpyrimidine-5-carboxylic acid was prepared in a yield of 80% (0.35 g) starting from ethyl 2 -phenylpyrimidine-5-carboxylate in a similar manner to that described in Example 23; m.p. 220'C (dec.).
The title compound was prepared in a quantitative yield from 2acid in a similar manner to that described in Example 3 0 m. p. 135 0
C.
Example 33 ETHYL 2 -TRIFLUOROiN A solution of diethyl ethoxymethylenemnalonate (35.0 g, 162 mmol), 3 5 trifluoroacetamidine (18 g, 162 mmol) and NaOEt (11. -0 g, 162 mmol) in EtOH (200 m.L) was heated at reflux for 6 h. The reaction mixture was concentrated and H120 (48 WO 97/09315 PCT/US96/15108 24 mL) was added. The resulting solid was filtered, washed with Et20 (300 mL) and (200 mL), and dried to give the title compound (21 g, 50% yield); m.p. >220 0 C (dec.); 'HNMR (DMSO-d 6 8 8.38,4.16 2H), 1.25 3H).
Example 34 2-TRFLUOROMETHYL-4-CHLOROPYRIDINE-5-CARBONYL
CHLORIDE
A solution of ethyl 2 (5.00 g, 19.4 mmol) and NaOH (0.93 g, 23.3 mmol) in H20 (20 mL) was stirred at 60 0
C
for 15 h. The reaction was acidified (conc. HCI) and concentrated until a solid began to form. The solid was filtered and dried to give 2 carboxylic acid (2.1 g, 53% yield); 'HNMR (DMSO-d 6 8 8.83 1H).
A solution of 2 -trifluoromethyl-4-hydroxypyrimidine-5-carboxylic acid g, 10.4 mmol), POCb (32 g, 212 mmol) and SOC1 2 (25 g, 212 mmol) was heated at reflux for 4 days. The reaction was concentrated and distilled 90-95 0 C, mm/Hg) to provide the title compound (2.1 g, 81% yield), 'HNMR (CDCI 3 6 9.45 (s, 1H).
Example 2 -CHLORO-4-PENTAFLUOROETHYLPYRIMIDINE-5-CARBONYL
CHLORIDE
A solution of ethyl 2-hydroxy-4-pentafluoroethylpyrimidine-5carboxylate (4.0 g, 13 mmol) and NaOH (1.60 g, 39 mmol) in EtOH (20 mL) and mL) was heated at reflux for I h. The solution was cooled and acidified (conc.
HCI). The resulting solid was filtered and dried to provide 2-hydroxy-4acid (3.3 g, 98% yield); 'H NMR (DMSO-d 6 6 9.90 (bs, 1H), 8.43 1H).
A solution of 2 -hydroxy- 4 -pentafluoroethylpyrimidine-5-carboxylic acid (3.33 g, 12.9 mmol) in SOC 2 (27.7 g, 233 mmol) was heated at reflux for 0.5 h. Then
POC
3 (35.6 g, 233 mmol) was added to the reaction mixture and heating continued for 36 h. The reaction mixture was concentrated and distilled 80-85 0 C, I mm/Hg) to give the title compound (1.2 g, 35% yield). 'H NMR (DMSO-d 6 6 9.18 1H).
Example 36
CARBOXAMIDE
A solution of 2-chloro-4-trifluoromethyl-5-N-(3,5dichlorophenyl)pyrimidine carboxamide (0.10 g, 0.27 mmol), Mg 2 0O (0.024 g, 0.59 WO 97/09315PCUS/1 0 PCT/US96/15108 mmol) and 5% Pd/C (0.01 g) in EtOH (1.8 mL) and water (0.9 ml,) was stirred at room temperature under a blanket of H 2 for 2.5 h. The reaction mixture was concentrated and chromatographed (SiOz 9% EtOAc/hexane) to yield the title compound (0.05 g, 53% yield); m.p. 189-190'C.
Example 37 2-DMETHYLAmiNo-4-TRiFLuoRo~MTHYL.
5-N-(3',5'-DioHLoRP-mNYL) PYRnvm flIEcARBoxAmTD)E A solution of 2-chloro-4-trifluoromethyl.5..N-(3,5.
dichlorophenyl)pyrimidine carboxamide 13 g, 0. 36 mmol) and dimethyl amine 10 g, 2.20 mmol) in MeOH was stirred at room temperature for 3 h. The reaction mixture was concentrated and chromatographed (SiO 2 5% EtOAc/hexane) to afford the title compound (0.022 g, 16% yield); m.p. 163-164'C.
Example 38 2-TRiETHYLAmmvoNmuN cHLoRiDE-4-TRFLuoRoMmTHYL 5-N-(3 5'-DICI-LOROPHENYL) PYRIMIDINE CARBOXAMIDE A solution of 2-chloro-4-trifluoromethyl-5-N-(3',5'dichlorophenyl)pyrimidine carboxamide 10 g, 0.27 mmol) and triethylamine (0.027 g, 0.27 mmol) in dry THE was stirred for 24 h. The solid was filtered, washed with Et 2
O,
and dried to afford the title compound (0.031 g, 24% yield); m.p. 158-1 59'C.
Example 39 2-CHiLoRo-4-METHYL-5-N..{3 METHYL)PHENYL]PYRIMDINE
CARBOXAMIDE
A solution of 2 -chloro-4-methylpyrimidine-5-carbonyl chloride (0.10 g, 0.53 mmol), 3,5-bis(trifluoromethyl)aniline (0.12 g, 0.53 mmol) and Amberlyst A-21 resin 1lOg) in EtOAc (5.3 mL) was stirred at room temperature for I h. The solution was filtered, concentrated and chromatographed (SiO 2 10% EtOAc/hexane) to afford the title compound 17 g, 84% yield); m.p. 156-157'C.
Example 2,4-Dic-LmoRo-5-N-[3', The title compound was prepared as described in Example 1, but employing 2,4-dichloropyrimidine-5-carbonylchlofide (0.10 g, 0.40 mmol) and WO 97/09315 PCTIUS96/15108 26 bistrifluoromethylbenzylamine (0.10 g, 0.45 mmol) to give the compound in a 61% yield (0.12 m.p. 144-145 0
C.
Example 41 2 4 -DICLoRo-5-N-[3',5'1BIS(TRpnLUORO.
The title compound was prepared as described in Example 1, but employing 2 4 -dichloropyrimidine-5-carbony chloride to give the compound in a 97% yield (0.28 m.p. 104-105C.
Example 42 2 -CYANo-4-TRFLoRoME THYL5-N-3 ETHYL)PHENYLPYRIMIDINE
CARBOXAMIDE
The title compound was prepared as described in Example 1, but employing 2 -cyano- 4 -trifiuoromethylpyrimidine-5-carbony chloride (0.11 g, 0.46 mnol) to give the compound in a 96% yield (0.19 m.p. 146-147C.
Example 43 2-CHLORO-4-PIENYL-5-N-[3 mETHYL)PHENYL]PYRIMIDINE
CARBOXAMIDE
A solution of 2 -chloro-4-phenylpyrimidine-5-carbony chloride (0.10 g, 0.40 mmol), 3,5-bis(trifiuoromethyl) aniline (0.08 g, 0.40 mmol) and Et 3 N (0.04 g, 0.40 mmol) in EtOAc was stirred at room temperature for 2 h. The solution was concentrated and chromatographed (SiO 2 5% EtOHICHCI 3 to afford the title compound (0.08 g, 45% yield); m.p. 154*C.
Example 44 2 (3 A solution of 2 -chloro4-trifiuoromethyl-5-N(3',5'dichlorophenyl)pyrimidine carboxamide (0.10 g, 0.27 mmol) and hydrazine (0.009 g, 0.54 mmol) in TIF was stirred under N 2 at room temperature for 14 h. The solution was filtered, concentrated and chromatographed (SiO 2 20% EtOAc/hexane) to afford the title compound (0.08 g, 79% yield), 'HNMR (acetone-d 6 6 10.08 (bs, 1H), 9.64 (bs, IH), 8.89 IH), 7.80 2H), 7.24 1H), 2.79 (bs, 2H).
WO 97/09315PCUS61w PCT/US96/15108 27 Example I-AMINOCITRACoNAm[DE)]-4-TRiFLuoRoMETHyL- 3 A solution of 2-hydrazino-4-trifluoromethyl-5-rN-(3', 59dichlorophenyl)pyrimidine carboxamide (0.08 g, 0.21 mmol) and citraconic anhydride (0.024 g, 0.21 mmol) in CHC1 3 (2.1 mL) was heated at reflux under N 2 for 24 h. The solution was concentrated and chromatographed (SiO 2 33% EtOAc/hexane) to afford the title compound (0.06 g, 62% yield); m.p. 182-183'C.
Example 46 2-PHENYLAmiNo-4-TRIFLUOROMETHYL- 5-N-(3 A solution of 2-chloro-4-trifluoromethyl-5-N-(3',5'dichlorophenyl)pyrimidine carboxamide (0.10 g, 0.27 mmol) and aniline (0.06 g, 0.59 mmol) in dry TIHE (2.7 mL) was stirred at room temperature under N 2 for 18 h. The reaction mixture was filtered, concentrated and chromatographed (SiO 2 CHCl 3 /hexane) to afford the title compound 10 g, 91 yield); m.p. 228-229'C.
Example 47 METHYL 5-cHwoRo-6-M THYL-2-PYRAzINE cARBoxyLATE.
To a solution of methyl 4 ,5-dihydro-6-methyl-5-oxo-2-pyrazine carboxylate Mano, T. Seo, K. Imai, Chem. Pharm. Bull 10:3057-3063, 1980) in DWvI (20 mL) was added POC1 3 (20 mL). The reaction was refluxed for 0.5 h and then poured into ice. The aqueous layer was extracted with CHC1 3 dried (MgSO 4 and concentrated. The residue was chromatographed (SiO 2
CHCI
3 to provide the title compound (2.34 g, 52% yield); m.p. 49-50"C.
Example 48 5-CHLoRo-6-METHYL-2-PYRAzINcARBOXYLIC
ACID
A mixture of methyl 5-chloro-6-methyl-2-pyrazine carboxylate 16 g, 0.86 mmol), K 2 C0 3 (0.31 g, 2.18 mmol) and H 2 0 was stirred for 2 h at room temperature. The reaction was filtered and acidified (20% HQI, and the resulting solid collected to provide the title compound (0.057 g, 39% yield); m.p. 116-1 17 0
C.
WO 97/09315 WO 9709315PCTIUS96/1 5108 28 Example 49 ANmiLNE) PYRAZINE CARBOXAMIDE The title compound was prepared in a yield of 51% (0.08 g) using the same procedure as outlined in Example 1, except substituting carbonyl chloride 1 g, 0. 57 mmol.) in place of the pyrimidine carbonyl chloride; m. p.
101-102 0
C.
Example 2-TRIMETHYLAMMONIUM C~HoRLiDE-4-TRiFLuoRoMETHYLcARBoxyuic AmI A solution of 2-chloro-4-trifiuoromethylpyrimidine-5-carboxylic acid g, 27 mmol) and excess trimethyl amine in THF (60 mL) was stirred for 5 min. The solid was filtered and dried to yield 97% (7.1 g) of the title compound; 'H NMvR (DMSO-d6) 859.19 IH), 2.72 9H).
Example 51 2-FLuoRo-4-TRiFLuoRomETHYL- CARBoxyLic AmI A mixture of 2-trimethylammonium pyrimidine carboxylic acid (4.3 g, 15 mmol), KF (1.8 g, 30 mmol), DMF (40 mL) and
H
2 0 (20 mL) was stirred for 0.5h. The mixture was concentrated, acidified and extracted with Et 2 O. The Et 2 O layer was concentrated to yield 47% (1.6 g) of the title compound; 'H NMR (DMSO-d6) 5 9.41 1H).
Example 52 2.-FLuoRo-4-TRIFLUORONETHYL- CARBoNYL CHLORIDE The title compound was prepared as described in Example 25, but employing a solution of 2-fiuoro-4-tfifiuoromethylpyrimidine-5-carboxylic acid (1.5 g, 7.1 mmol) and oxalyl chloride (1.0 g, 8 mmol), DMF (2 drops) in CH 2
CI
2 (30 mL) resulted in a 75% yield (1.2 'H NMR (CDC1 3 5 9.42 IH).
WO 97/09315 PCT/US96/15108 29 Example 53 2-FLUORO-4-TRIFLUOROMETHYL-5-N-[3',5'-BIS(TRIFLUOROMETHYL)PHENYL] PYRIMIDINE CARBOXAMIDE The title compound was prepared as described in Example 1, but employing a solution of 2 -fluoro-4-trifluoromethylpyrimidine-5-carbonyl chloride (0.05 g, 0.22 mmol) and 3,5-bis(trifluoromethyl)aniline (45 mg, 0.2 mmol) in EtOAc (2 mL) resulted in a 22% yield (0.02 m.p. 133-135 0
C.
Example 54 2 -CHLORO-4-TRIFLUOROMETHYL- CARBONYL CHLORIDE The title compound was prepared as described in Example 25, but employing a solution of 2 -chloro-4-trifluoromethylpyrimidine-5-carboxylic acid (1.5 g, 7.1 mmol) and oxalyl chloride (1.0 g, 8 mmol) in CH 2 C12 (30 mL) resulted in a yield 'HNMR(CDC 3 8 9.31 1H).
Example SYNTHESIS OF REPRESENTATIVE COMPOUNDS BY COMBINATORIAL CHEMISTRY TECHNIQUES This example illustrates the synthesis of a representative class of compounds of this invention by combinatorial chemistry. It should be understood that, while a specific class of compounds are illustrated in this example, the following procedure may be employed to synthesize other compounds of this invention.
Into wells 2-11 of a 96 well 1 mL plate (rows 1 and 12 left open as controls) was added 5 mg of Amberlyst 21 resin, 0.2 mL of EtOAc and 22.4 pmol of different amine derivatives. Then to each well was added 25.0 pmol of the appropriate chloride (for example 2 -chloro- 4 chloride). The 96 well plate was sonicated for 0.3 h and 50 pL of H 2 0 was added to each well. The plate was sonicated for an additional 0.25 h, and the EtOAc layer from each well was removed and concentrated to provide 80 individual compounds. Thinlayer chromatography, HPLC and GC/MS analysis indicated that the desired compounds had been produced at >90% purity. This approach can be used to generate large numbers of derivatives for each substituted pyrimidine prepared, and can be used to routinely prepare >160 derivatives for each of the different 5-carbonyl pyrimidines.
WO 97/09315 PCT/US96/15108 Example 56 INHIBITION OF THE ACTIVATION OF NFKB AND AP- A. NFKB ASSAY Stable human Jurkat T-cells containing an NFxB binding site (from the MHC promoter) fused to a minimal SV-40 promoter driving luciferase expression were used in this experiment. Cells were split to 3 x 105 cells/mL every 2-3 days (cell concentration should not exceed 1 x 106 cells/mL to keep the cells proliferating in log phase). These cells were counted, resuspended in fresh medium containing 10% Serum- Plus at a density of 1 x 106 cells/mL and plated in 96 well round bottom plates (200 L per well) 18 hours prior to starting the experiment.
Compounds of this invention, dissolved in dimethyl sulfoxide 0.33 and 0.03 tg/mL), were then added to the 96 well plates containing the cells and the plates are incubated for 0.5 h at 370C. Then 50 ng/mL of phorbol 12-myristate-13acetate (PMA) and 1 pg/mL of phytohemagglutinin (PHA) was added to each well and the cells were incubated for an additional 5 h at 370C. The plates were centrifuged at 2200 RPM for 3 minutes at room temperature and then the medium was removed. To each well was added 60 pL of cell lysis buffer and the plates were left at room temperature for 0.25 h. Then 40 pL of each cell extract was transferred to a black 96 well plate and 50 uL of luciferase substrate buffer was added. Luminescence was immediately measured using a Packard TopCount.
B. AP-1 ASSAY For AP-1, the assay was run as described above for NFKB except stable Jurkat T-cells were used that contained a collagenase promoter driving luciferase expression. In addition, the concentration of PMA used was 5 ng/mL.
C. RESULTS The results of the above assays for a representative compound of this invention, 2-chloro-4-trifluoromethyl-5-N-(3',5'-bistrifluoromethylphenyl)pyrimidine carboxamide, as percent inhibition versus control are presented in Figure 3. This figure also indicates activity of P-actin which was employed in these assays as a control cell line indicating effects on transcription. The lack of P-actin activity evidences selectivity of the test compounds for the transcription factors AP-1 and NFKB.
Expressed as ICso's, the results of these assays on additional test compounds are summarized in Table 2 below.
WO 97/09315 PCT/US96/15108 31 Table 2 Test Compound NFrB/AP-1 (Example 1 0.03 2 0.75 6 0.8 8 11 12 0.4 13 0.8 39 0.075 41 0.6 42 43 Based on the results of this experiment, representative compounds of this invention were found to be effective at inhibiting the activation of transcription factors NFKB and AP-1) involved in gene transcription, and therefore have utility as, for example, immunosuppressive agents.
Example 57 INHIBITION OF CYTOKINES To determine the effects of compounds on PMA/PHA-induced cytokine production, supernatants from either the NFxB (for IL-8) and AP-1 (for IL-2) reporter gene assays of Example 56 were collected and saved. Cytokine levels in the supernatants (25-50 pL aliquots) were determined by ELISA. The results of this experiment for a representative compound of this invention, 2-chloro-4-trifluoromethyl- 5-N-(3',5'-bistrifluoromethylphenyl)pyrimidine carboxamide, is presented in Figure 4 (expressed as percent inhibition versus control).
Example 58 IN Vivo ACTIVITY OF REPRESENTATIVE COMPOUND The murine popliteal lymph node (PLN) assay is a graft vs. host model that predicts activity of compounds in blocking human transplant rejection. The delayedtype hypersensitivity response to oxazolone is a standard contact sensitivity model. Both WO 97/09315 PCT/US96/15108 32 of these models are used routinely to evaluate compounds that are used clinically. For example, cyclosporin and cyclophosphamide are active in these models and are used clinically (Morris et al., Transplantation Proceedings 22(Suppl. 110-112, 1990).
A. POPLITEAL LYMPH NODE MODEL Spleens were removed from donor BALB/c mice and splenocytes were isolated then irradiated (3,000 rads) to prevent donor cell proliferation. After washing and adjusting cell density, 2.5x10 6 cells were injected subcutaneously into the left hind footpad ofC3H mice. On day 4, the mice were sacrificed and left popliteal lymph nodes (PLNs) were weighed.
The compound of Example 1, 2 -chloro-4-trifluoromethyl-5-N-(3',5'bistrifluoromethylphenyl)pyrimidine carboxamide, was administered once daily by intraperitoneal injection beginning one day before footpad injection (day 0) through day 4. The compound was suspended, immediately prior to use, at a concentration of mg/mL in 0.25% methyl cellulose (Sigma) using a glass-teflon homogenizer. For doses of 10, 20 and 30 mg/kg, appropriate dilutions of the stock solution were made so that 0.1 mL/10 g body weight was administered by intraperitoneal injection.
The results of this experiment, presented in Figure 5, demonstrate that a representative compound of this invention caused a dose-dependent suppression of alloantigen-induced PLN proliferation. The lowest dose of this compound, 10 mg/kg, caused a 52% inhibition of proliferation whereas cyclosporin A, at 12 mg/kg, caused a inhibition.
B. DELAYED TYPE HYPERSENSITIVITY
STUDY
On day 0, oxazolone (100 pL of a 3% solution) was applied to the shaved abdomen of mice. On day 7, a challenge application of oxazolone was applied pL) around the right ear. The compound of Example 1, 2 -chloro-4-trifluoromethyl- 3 ,5-bistrifluoromethylphenyl)pyrimidine carboxamide, was administered from days -2 to 7 by intraperitoneal injection. It was prepared immediately prior to use by suspending it in 0.25% methyl cellulose (Sigma) using a glass-teflon homogenizer. For each dose, 0.1 mL/10 g body weight of the suspension was administered. The compound was prepared at the highest concentration for that study and appropriate dilutions of the stock solution were made so that 0.1 mL/10 g body weight was administered. Twenty four hours later, the difference in right vs. left ear thickness was measured. The results of this experiment are presented in Table 3 below.
Table 3 Effect on the DTH Response to Oxazolone Dose Right-Left Ear (mg/kg) (mean SEM) Compound P Value (vs. vehicle) Vehicle only Test Cpd.
Test Cpd.
Cyclophosphamide *One animal died during study 10 30 50 0.30 0.02 0.27 0.01 0.13 0.02 0.08 0.01 0.163 <0.001* <0.001
S
S
S
S
S
S
S
*5 Sr 5 The test compound (30 mg/kg and cyclophosphamide (50 mg/kg significantly attenuated the delayed-type response to oxazolone by 56% and 73%, respectively.
It will be appreciated that, although specific embodiments of this invention have been described herein for purpose of illustration, various modifications may be made without departing from the spirit and scope of the invention.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

Claims (12)

  1. 34- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: 1. A method for treating an inflammatory condition in a warm blooded animal in need thereof comprising administering to said animal an effective amount of a compound having the structure: R R 4 .a yR 999*N N N *2a including pharmaceutically acceptable salts thereof, wherein 5 is selected from the following chemical moieties: S 10 Rg R RgR O IN S N R R 7 N "k 07 R7 N R7isseete fomhdrge,-C3,anN'SC65 R8 is 1eetdfo yrgnada nustttdo usiue I8 ,a *eenbfr deieC-2rl 71ar9*,C-2eeoyl ndaC-oeeoylak ~R4, is selected from hydrogen, Chlg, an -CH 2 C 6 H 5 o ubtiuedC-8lla hereinbefore. defined, C 6 .i 2 aryl, C7l 2 arllcyl, C 3 12 heterocycle andr C4- 2 ohleterocyclealkyl; CN, -OR, -NRR, Hd -NHNII 2 nd1 5 an. NCR is selected from hydrogen, halogenan an unsubstituted or substituted C i -alkyl; and wherein each occurrence of R is independently selected from an unsubstituted or substituted C 1 8 salkyl as hereinbefore defined, C 6 12 aryl, C7- 12 aralkyl, C 3 12 hterocycle or C4-2oheterocyclealkyl; ),EJwi th the provisos that: when R 5 is -CONR 7 R 8 R7, and R8 are not both P \OPER PDBR\7! 631 -96,I,,.sdo-14/09/00 straight chain or branched, non-cyclic, saturated C 1 3 alkyl, -N(CH 3 2 -N(CH 2 CH 3 2 and OR, (iii) when R 2 a is -Cl and R,5 is R 4 a is not selected from -CF 3 -Cl, -CH 3 and C(CH 3 3 (iv) When R 2 a is -Cl, and both R 4 a and R 6 are R 8 is not -CH(CN)C 6 H 5 and when R 2 a is -Cl and R~a is -Cl, R 6 is not selected from -Cl and -CH 2 CL; and when R 5 is -N(R 7 )C(0)R 8 R 2 a is not selected from -CH 3 -OCH 3 and -N(CH 3 2 and (ii) R 8 is not selected from -H and -CH 3 2. The method of claim 1 wherein said compound has the structure: o N, R N N 3. The method of claim 2 wherein R. 4 a is selected from -CF 3 -Cl, -CH 3 and -H. be.. C. S.. 0 4 0 C S* 0* *0 C C* 0 9 C C U C 4. CF 3 The method of claim 2 wherein R 2 a is selected from -Cl, -OCH 3 -N(CH 3 2 -CN, -NHNI{ 2 and -NHC 6 H 5 The method of claim 2 wherein R 6 is selected from -CF 3 -CH 3 and -Cl. 6. The method of claim 2 wherein R 7 is selected from -H and -CH 3 7. The method of claim 2 wherein R 8 is z P',OPERPDB\71631-96 111i.I.A.- 14,09/00 -36- wherein X, Y, and Z are the same or different, and independently selected from hydrogen, -OH, -OR, -COOH, -COOR, -COR, -CONH2, -NH2, -NER, -NRR, -SH, SR, -SOOR, -S03R and -SOR. 8. The method of claim 2 wherein R4a is -CF3 and R2a is -Cl- 9. The method of claim 2 wherein R4a is -CF3- The method of claim 9 wherein the compound is selected from 2-fluoro-4- trifluoromethyl-5-N-(3',5'-bistrifluoromethyl)pyrimidine carboxamide, bis(tn'fluoromethyl)phenacyl)-2-methoxy-4-tn'fluoromethylpyrimidine carboxamide; 2-dimethylamino-4-trifluoromethyl-5-N- V. 660 carboxamide; 2-triethylammonium chloride-4- trifluoromethyl-5-N-(3',5'-dichlorophenyl)pyTimidine carboxamide; 2-cyano-4- trifluoromethyl-5-N-[3',5'-(bistrifluoromethyl)phenyl]pyrimidine carboxamide; 2- hydrazino-4-trifluromethyl-5-[N-(3',5'-dichlorophenyl)pyrimidine-5-carboxamide; 2-[N- (I -aminocitraconamide)]-4-trifluromethyl-5-[N-(3',5'-dichlorophenyl) 06 carboxamide; and 2-aminophenyl-4-trifluromethyl-N-(3',5'-dichlorophenyl)pyrimidine-5- 0, carboxamide. 11. The method of claim 2 wherein R2a is -Cl. 12. The method of claim I I wherein the compound is selected from bis(tn'fluoromethyl)phenyl]-2,4-dichloro-6-methyl-pyrimidine carboxamide; 2-chloro-4- methyl-5-N-[3',5'-(bistn'fluoromethyl)phenyl]pyrimidine carboxamide; 2,4-dichloro-5-N- [3',5'-bis(tn'fluromethyl)benzyl]pyrimidine-5-carboxarnide; and 2-chloro-4-phenyl-5-N- carboxamide. 13. The method of claim 2 wherein R8 is a 3,5-disubstitued phenyl moiety, wherein both substituents are electron withdrawing groups. P\OPERPDB\71631-96 claims.doc-1409/00 -37- 14. The method of claim 13 wherein both substituents are -CF 3 The method of claim 13 wherein at least one of the substituents is -CF 3 16. The method of claim 2 wherein R 4 a is selected from -CH 3 -CF 3 -CF 2 CF 3 C 6 H 5 and -CH 2 C 6 H 5 17. The method of claim 2 wherein R 2 a is selected from -Cl, -CN and -CF 3 10 18. The method of claim 2 wherein R 2 ais selected from -Cl and -F. 19. The method of claim 2 wherein R 6 is -H. A method for treating an inflammatory condition in a warm blooded animal in need 15 thereof comprising administering to said animal an effective amount of a compound having the structure: a. ."5 S: 5R4b N' RI R2b including pharmaceutically acceptable salts thereof, wherein R 5 is selected from the following chemical moieties: Rg R Rg Rg 0 N S N 7 R 7 NO R 7 Nk y R 7 R7 N O N S R7 is selected from hydrogen, -CH 3 and -CH 2 C 6 H 5 Rg is selected from hydrogen and an unsubstituted or substituted Cl-salkyl, PAOPER\PDH\II63I96chi,-doc 3/09/00 38 as hereinbefore defined, C 6 -1 2 aryl, C 7 12 aralkyl, C3l1 2 heterocycle and a C 4 2oheterocyclealkyl; R2b is halogen; R4a is selected from hydrogen, halogen, -CN, and an unsubstituted or substituted CI-8alkyl as hereinbefore defined, C 6 -1 2 aryl, C 7 -1 2 aralkyl, C 3 i 2 heterocycle or C 4 2 heterocyclealkyl; and R, is selected from hydrogen, -CH 3 -CF 3 and -CH 2 CH 3 with the proviso that when R 5 is -CONR 7 R 8 and R2b is -Cl, R4b and R, are not both hydrogen. 0* 9 *21. The method of claim 20 wherein ROb is an unsubstituted C 1 salkyl as hereinbefore defined. 22. The method of claim 20 wherein R2b is selected from -Gl and -F. 23. The method of claim 20 wherein R, is selected from -H and -CH 3 24 Th.ehdo li 0weenR sslce rm- n C3 24. The method of claim 20 wherein Rg is slce rm- n C 3 pq z wherein X, Y and Z are the same or different, and independently selected from hydrogen, -OH, -OR, -COOH, -COOR, -COR, -CON-H 2 -NH 2 -NHR, -NRR, -SH, SR, -SOOR, -SO 3 R and -SOR. 26. The method of claim 20 wherein R4b is -CF 3 and R2b is -Cl. P OPERPDB.71631-96 claimns doc-13 09/00 9S** a -39- 27. The method of claim 20 wherein R4b is -CF 3 28. The method of claim 20 wherein R2b is -Cl. 29. The method of claim 20 wherein R 8 is a 3,5-disubstituted phenyl moiety, wherein both substituents are electron withdrawing groups. 30. The method of claim 29 wherein both substituents are -CF 3 31. The method of claim 29 wherein at least one of the substituents is -CF 3 32. The method of claim 20 wherein R4b is selected from -CH 3 -CF 3 -CF 2 CF 3 C 6 H 5 and -CH 2 C 6 H 5 15 33. The method of claim 20 wherein Ri is -H. 34. The method of claim 1 or 20 wherein the inflammatory condition is an immunoinflammatory condition.
  2. 35. The method of claim 1 or 20 wherein the immunoinflammatory condition is selected from rheumatoid arthritis, osteoarthritis, transplant rejection, sepsis, ARDS and asthma.
  3. 36. The method of claim 1 or 20 wherein the immunoinflammatory condition is rheumatoid arthritis.
  4. 37. The method of claim 1 or 20 wherein the inflammatory condition is an autoimmune disease.
  5. 38. The method of claim 1 or 20 wherein the autoimmune disease is selected from multiple sclerosis, psoriasis, inflammatory bowel disease, glomerulonephritis, lupus, 19- 3-00; 3:31PM;DAVIES COLLISON CAVE ;613 92542771] 4/ 7 P:PE*RIi71if63I-96. Ei /6D4) uveitis and chronic hepatitis. @9 @9 9 9 9 9 o°999@ *O *0* 9 99 @9 0990 9* *990 9. @9 o **ooo*
  6. 39. The method of claim I or 20 wherein the inflammatory condition is selected from trauma, oxidative stress, cell death, irradiation damage, ischaemia, reperfusion, cancer and viral infection. The method of claim I or 20 wherein the inflammatory condition is transplant rejection. 10 41. Use of a compound as defined in any one of claims 1-33 for the manufacture of a medicament for treating an inflammatory condition.
  7. 42. The use of claim 41 wherein the inflammatory condition is an immunoinflammatory condition.
  8. 43. The use of claim 42 wherein the immrnunoinflammatory condition is selected from rheumatoid arthritis, osteoarthritis, transplant rejection, sepsis, ARDS and asthma.
  9. 44. The use of claim 42 wherein the immunoinflammatory condition is rheumatoid arthritis. The use of claim 41 wherein the inflammatory condition is an autoimmune disease.
  10. 46. The use of claim 45 wherein the autoimmune disease is selected from multiple sclerosis, psoriasis, inflammatory bowel disease, glomemrulonephritis, lupus, uveitis and chronic hepatitis.
  11. 47. The use of claim 41 wherein the inflammatory condition is selected from trauma, oxidative stress, cell death, irradiation damage, ischaemia, reperfusion, cancer and viral infection. 19/09 '00 TUE 15:30 [TX/RX NO 5947] 19-9-0 a; 3!31PM;DAVIES COLLISON CAVE ;613 92542770 5/ 7 -41-
  12. 48. The use of claim 41 wherein the inflarmmatory condition is transplan rejection. 49, A method according to claim I or 20 substantially as hereinibefore described or exemplified. A use according to claim 41 substantially as hereinbefore described or exemplified. Dated this 13th day of September 2000 Signal Pharmaceuticals, Inc. 10 By Its Patent Attorneys DAVIES COLLISON CAVE 19/09 '00 TUE 15:30 [TX/RX NO 5947]
AU71631/96A 1995-09-01 1996-08-30 Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions Ceased AU726522B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US310995P 1995-09-01 1995-09-01
US60/003109 1995-09-01
US08/581,473 US5811428A (en) 1995-12-18 1995-12-18 Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions
US08/581473 1995-12-18
PCT/US1996/015108 WO1997009315A1 (en) 1995-09-01 1996-08-30 Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions

Publications (2)

Publication Number Publication Date
AU7163196A AU7163196A (en) 1997-03-27
AU726522B2 true AU726522B2 (en) 2000-11-09

Family

ID=26671336

Family Applications (1)

Application Number Title Priority Date Filing Date
AU71631/96A Ceased AU726522B2 (en) 1995-09-01 1996-08-30 Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions

Country Status (5)

Country Link
EP (1) EP0850228A1 (en)
JP (1) JPH11512399A (en)
AU (1) AU726522B2 (en)
CA (1) CA2230894A1 (en)
WO (1) WO1997009315A1 (en)

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5939421A (en) * 1997-07-01 1999-08-17 Signal Pharmaceuticals, Inc. Quinazoline analogs and related compounds and methods for treating inflammatory conditions
US6022884A (en) 1997-11-07 2000-02-08 Amgen Inc. Substituted pyridine compounds and methods of use
US6462019B1 (en) 1998-07-10 2002-10-08 Osteoscreen, Inc. Inhibitors of proteasomal activity and production for stimulating bone growth
US6902721B1 (en) 1998-07-10 2005-06-07 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone growth
US6838436B1 (en) 1998-07-10 2005-01-04 Osteoscreen Inc. Inhibitors of proteasomal activity for stimulating bone growth
ATE266662T1 (en) 1998-07-22 2004-05-15 Daiichi Suntory Pharma Co Ltd NF-KAPPA B INHIBITORS CONTAINING INDANDERIVATES AS THE ACTIVE INGREDIENTS
ID30460A (en) 1999-04-15 2001-12-06 Bristol Myers Squibb Co INHIBITORS, INHIBITORS, THYROCIN CINASE PROTEIN
US7125875B2 (en) 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
JO2308B1 (en) * 1999-05-31 2005-09-12 اف. هوفمان- لاروش أيه جي‏ 5-phenyl purinidine derivatives
TW550258B (en) 1999-05-31 2003-09-01 Hoffmann La Roche 4-phenyl-pyrimidine derivatives
CN1212861C (en) * 1999-09-17 2005-08-03 第一三得利制药株式会社 Preventives or remedies for myocarditis, dilated cardiomyopathy and cardiac insufficiency containing NF-K B inhibitors as the active ingredient
WO2002047690A1 (en) * 2000-12-12 2002-06-20 Cytovia, Inc. Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US6716851B2 (en) 2000-12-12 2004-04-06 Cytovia, Inc. Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators or caspases and inducers of apoptosis and the use thereof
AR036375A1 (en) * 2001-08-30 2004-09-01 Novartis Ag PIRROLO [2,3-D] PIRIMIDINE -2- CARBONITRILE COMPOUNDS, A PROCESS FOR THEIR PREPARATION, A PHARMACEUTICAL COMPOSITION AND THE USE OF SUCH COMPOUNDS FOR THE PREPARATION OF MEDICINES
US7205307B2 (en) 2002-02-14 2007-04-17 Icagen, Inc. Pyrimidines as novel openers of potassium ion channels
EP1487796A4 (en) * 2002-03-28 2005-11-16 Neurogen Corp Substituted biaryl amides as c5a receptor modulators
US6858637B2 (en) 2002-03-28 2005-02-22 Neurogen Corporation Substituted biaryl amides as C5a receptor modulators
WO2003103647A1 (en) * 2002-06-05 2003-12-18 株式会社医薬分子設計研究所 Inhibitors against the activation of ap-1 and nfat
JP4743382B2 (en) 2002-06-06 2011-08-10 株式会社医薬分子設計研究所 O-substituted hydroxyaryl derivatives
SE0201976D0 (en) 2002-06-24 2002-06-24 Astrazeneca Ab Novel compounds
ES2273077T3 (en) * 2002-08-21 2007-05-01 Glaxo Group Limited DERIVATIVES OF 2-PHENYLAMINE-4-TRIFLUOROMETIL-5 (BENCIL- OR PIRIDIN-4-IL-METIL) CARBAMOILPIRIMIDINE AS SELECTIVE MODULATORS OF THE CANNABINOID CB2 RECEIVER.
UY27939A1 (en) * 2002-08-21 2004-03-31 Glaxo Group Ltd COMPOUNDS
GB0220187D0 (en) 2002-08-30 2002-10-09 Novartis Ag Organic compounds
GB0402355D0 (en) * 2004-02-03 2004-03-10 Glaxo Group Ltd Novel compounds
GB0402356D0 (en) * 2004-02-03 2004-03-10 Glaxo Group Ltd Novel compounds
EP1718620A1 (en) * 2004-02-23 2006-11-08 Glaxo Group Limited Pyrimidine derivatives as cannabinoid receptor modulators
GB0404105D0 (en) * 2004-02-24 2004-03-31 Glaxo Group Ltd Novel compounds
CA2571710A1 (en) 2004-06-24 2006-11-02 Nicholas Valiante Small molecule immunopotentiators and assays for their detection
US7671058B2 (en) 2006-06-21 2010-03-02 Institute Of Medicinal Molecular Design, Inc. N-(3,4-disubstituted phenyl) salicylamide derivatives
CA2679198C (en) 2007-02-26 2011-07-12 Pfizer Products Inc. Nicotinamide derivatives as inhibitors of h-pgds and their use for treating prostaglandin d2 mediated diseases
US8063058B2 (en) 2008-04-16 2011-11-22 Portola Pharmaceuticals, Inc. Inhibitors of syk and JAK protein kinases
US8138339B2 (en) 2008-04-16 2012-03-20 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
EP2415765A4 (en) 2009-03-30 2012-08-15 Astellas Pharma Inc Pyrimidine compound
US8580805B2 (en) * 2010-08-31 2013-11-12 Hubert Maehr Pyrimidine carboxamide derivatives
AU2010363329A1 (en) 2010-11-07 2013-05-09 Targegen, Inc. Compositions and methods for treating myelofibrosis
KR102019530B1 (en) 2011-11-23 2019-09-06 포톨라 파마슈티컬스, 인코포레이티드 Pyrazine kinase inhibitors
WO2017122209A2 (en) * 2016-01-12 2017-07-20 Yeda Research And Development Co. Ltd. NF-kappaB INHIBITORS
EP3651747A4 (en) 2017-11-17 2020-07-22 Cellix Bio Private Limited Compositions and methods for the treatment of eye disorders

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1898195A (en) * 1994-03-18 1995-10-09 Agrevo Uk Limited Anilide derivatives as fungicides

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE489416A (en) *
US3517010A (en) * 1968-04-05 1970-06-23 American Home Prod 5-acetamido-4-pyrimidinecarboxylic acids and related compounds
US3517009A (en) * 1968-04-05 1970-06-23 American Home Prod 4-hydroxy(lower)alkylamino-n,2-disubstituted-5-pyrimidinecarboxamides
CH549339A (en) * 1971-05-12 1974-05-31 Ciba Geigy Ag HERBICIDAL AGENT.
GR66581B (en) * 1978-02-21 1981-03-27 Delalande Sa
DE3205638A1 (en) * 1982-02-17 1983-08-25 Hoechst Ag, 6230 Frankfurt Trisubstituted pyrimidine-5-carboxylic acids and their derivatives, processes for their preparation, and their use as pesticides
JPS63107966A (en) * 1986-05-22 1988-05-12 Fujisawa Pharmaceut Co Ltd Pyrimidine derivative
JPH075568B2 (en) * 1987-02-13 1995-01-25 ダイセル化学工業株式会社 Pyrimidine-5-carboxamide derivative and plant growth inhibitor
JPH01180805A (en) * 1988-01-05 1989-07-18 Daicel Chem Ind Ltd Herbicide composition
JPH01180804A (en) * 1988-01-05 1989-07-18 Daicel Chem Ind Ltd Herbicide composition
HUT63941A (en) * 1992-05-15 1993-11-29 Hoechst Ag Process for producing 4-alkyl-substituted pyrimidine-5-carboxanilide derivatives, and fungicidal compositions comprising same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1898195A (en) * 1994-03-18 1995-10-09 Agrevo Uk Limited Anilide derivatives as fungicides

Also Published As

Publication number Publication date
CA2230894A1 (en) 1997-03-13
EP0850228A1 (en) 1998-07-01
JPH11512399A (en) 1999-10-26
WO1997009315A1 (en) 1997-03-13
AU7163196A (en) 1997-03-27

Similar Documents

Publication Publication Date Title
AU726522B2 (en) Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions
US5811428A (en) Pyrimidine carboxamides and related compounds and methods for treating inflammatory conditions
AU726058B2 (en) Pyrimidine carboxylates and related compounds and methods for treating inflammatory conditions
US5935966A (en) Pyrimidine carboxylates and related compounds and methods for treating inflammatory conditions
RU2485113C2 (en) Cytokine inhibitors
US4861891A (en) Antidepressant N-substituted nicotinamide compounds
KR100565439B1 (en) Novel pyridazine derivatives and drugs containing the same as the active ingredient
AU2006335967B2 (en) Novel heterocycles
US20070082912A1 (en) Pyrrole compounds for the treatment of prostaglandine mediated diseases
US5852028A (en) Pyrimidine carboxylates and related compounds and methods for treating inflammatory conditions
JP2005535570A (en) New pyrimidone derivatives
PL170373B1 (en) Method of obtaining novel, substituted 5-aryl pyrimidines
WO1989003818A1 (en) Novel fenamic acid hydroxamate derivatives having cyclooxygenase and 5-lipoxygenase inhibition
CZ220499A3 (en) Derivative of [pyrrolo 3,4-d]pyrimidinone and application thereof as a medicament
US7317014B2 (en) Bio-active pyrimidine molecules
IE852118L (en) Ethylenediamine monoamide derivatives.
Goto et al. Synthesis and biological evaluation of 5-carbamoyl-2-phenylpyrimidine derivatives as novel and potent PDE4 inhibitors
AU2009208712B2 (en) Novel heterocycles
US20090163521A1 (en) Novel Pyrazolopyrimidinone Derivatives
AU2005223423A1 (en) Derivatives of heteroaryl-alkylcarbamates, preparation method thereof and use of same as FAAH enzyme inhibitors
US4769380A (en) Cardiotonic 5-benzoyl-1,2-dihydro-2-oxo-3-pyridinecarboxylates
KR20040107499A (en) Novel pyrimidone derivatives
EP0801062A1 (en) Pyrimidine derivatives
AU604084B2 (en) (2-cyano-2-(phenyl or naphthyl)-2-substituted ethyl) pyrazines, pyrimidines and pyridazines
US4992452A (en) Certain 5-acyl-2-(1H)-pyridinones useful in treating cardiac failure

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired