Title of Invention

THIOPHENE COMPOUNDS

Abstract The present invention provides compounds of formula (I): (I) pharmaceutical compositions containing the same, processes for preparing the same and their use as pharmaceutical agents, including their use for the treatment of cancer.
Full Text THIOPHENE COMPOUNDS
BACKGROUND OF THE INVENTION
The present invention relates to novel compounds, pharmaceutical formulations
comprising these compounds, and the use of these compounds in therapy. More
particularly, the present invention relates to novel compounds and methods for
treating conditions mediated by Polo-like Kinase, susceptible neoplasms, and other
conditions.
Polo-like kinases ("PLK") are evolutionary conserved serine/threonine kinases that
play critical roles in regulating processes in the cell cycle. PLK plays a role in the entry
into and the exit from mitosis in diverse organisms from yeast to mammalian cells.
PLK includes PLK1, PLK2, and PLK3.
Polo-like kinases are known to be essential for mitosis in yeast, Drosophila, and
Xenopus. For example, mutants of the homologous PLK genes in these organisms
result in disordered mitotic spindles, and in Drosophila mutations can be embryonic
lethal. RNA interference experiments on Drosophila polo have shown that ablation of
polo in S2 cells results in G2/M arrest and apoptosis. PLK1 is the human homolog of
Drosophila polo. It is believed to be involved in the entry into mitosis through the
activation of cdk1 by phosphorylating and activating the phosphatase cdc25C, which
in turn removes inhibitory phosphates from cdk1. This sets up an activation loop for
cdk1 that leads to mitotic entry. PLK1 also phosphorylates cyclin B1, the cyclin
partner of cdk1, resulting in nuclear localization. During mitosis, PLK1 has been
shown to play roles in centrosome maturation and microtubule dynamics involved in
formation of the mitotic spindle. PLK1 is also involved in the exit of cells from mitosis
by phosphorylating and activating subunits of the anaphase-promoting complex
(cdc16 and cdc27). PLK1 also phosphorylates cohesin proteins that hold sister
chromatids together, exposing separase cleavage sites, and allowing separation of
sister chromatids during anaphase. PLK1 may also play a role in cytokinesis through
mosphorylation of the kinesin-like motor protein MKLP1. Inhibition of PLK1 thus has
the potential to interfere with several stages of mitosis. Expression and activity of PLK
protein increases during the cell cycle, reaching its peak during mitosis when it is also
maximally phosphorylated. PLK1 mRNA is highly expressed in cells with a high mitotic
index. PLK2 (serum-inducible kinase, SNK) and" PLK3 (Proliferation-related kinase PRK
Fibroblast Growth Factor-inducible Wnase, FNK) were originally identified as
immediate-early genes. PLK2 is not very well characterized, but PLK3 appears to be
involved In regulation of cell cycle progression through M phase but functions
differently from PLK1. Recent published work suggests that PLK3 plays an important
role in the regulation of microtubule dynamics and function of the centrosome during
mitosis.
Overexpression of PLK1 appears to be strongly associated with neoplastic cells
(including cancers). A published study has shown high levels of PLK1 RNA expression
in >80% of lung and breast tumors, with little to no expression in adjacent normal
tissue. Several studies have shown correlations between PLK expression, histological
grade, and prognosis in several types of cancer. Significant correlations were found
between percentages of PLK-positive cells and histological grade of ovarian and
endometrial cancer (P Invading endometrial carcinoma cells and that this could reflect the degree of
malignancy and proliferation in endometrial carcinoma. Using RT-PCR analysis, PLK
overexpression was detected in 97% of esophageal carcinomas and 73% of gastric
carcinomas as compared to the corresponding normal tissues. Further, patients with
high levels of PLK overexpression in esophageal carcinoma represented a significantly
poorer prognosis group than those with low levels of PLK overexpression. In head and
neck cancers, elevated mRNA expression of PLK1 was observed in most tumors; a
Kaplan-Meier analysis showed that those patients with moderate levels of PLK1
expression survived longer than those with high levels of PLK1 expression. Analysis of
patients with non-small cell lung carcinoma showed similar outcomes related to PLK1
expression.
bisruption of mitosis with anti-microtubule drugs has been a successful approach in
cancer chemotherapy. The taxanes and vinca alkaloids have been effectively used in
the clinic, but they have undesirable side effects. In addition, many tumors appear to
have weakened G2/M cell cycle checkpoints; in response to mitotic disruption these
tumors attempt to bypass mitosis, leading to mitotic catastrophe and cell death.
Several studies suggest that the disruption of mitosis by targeting PLK may be a
feasible approach to selective tumor cell destruction. There remains a need in the art
for new approaches to the treatment of neoplasms.
BRIEF SUMMARY OF THE INVENTION
According to a first aspect of the invention there is provided a compound of formula
wherein:
R1 is selected from the group consisting of H, alkyl, alkenyl, alkynyl, -C(O)R7, -CO2R7,
-C(O)NR7R8. -C(O)N(R7)OR8. -C(O)N(R7)-R2-OR8, -C(O)N(R7)-Ph,
-C(O)N(R7)-R2-Ph, -C(O)N(R7)C(O)R8, -C(O)N(R7)CO2R8, -C(O)N(R7)C(O)NR7R8,
-C(O)N(R7)S(O)2R8, -R2-OR7, -R2-O-C(O)R7, -C(S)R7, -C(S)NR7R8, -C(S)N(R7)-Ph,
-C(O)N(R7)-R2-Ph, -R2-SR7, -C(=NR7)NR7R8. -C(=NR7)N(R8)-Ph,
-C(-NR7)N(R8)-R2-Ph, -R2-NR7R8, -CN, -OR7, -S(O)tR7. -S(O)2NR7R8,
-S(O)2N(R7)-Ph, -S(O)2N(R7)-R2-Ph, -NR7R8, N(R7)-Ph, -N(R7)-R2-Ph, -N(R7)-SO2R8
and Het;
Ph is phenyl optionally substituted from 1 to 3 times with a substituent selected from
the group consisting of halo, alkyl, -OH, -R2-OH, -O-alkyl, -R2-O-alkyl, -NH2,
-N(H)alkyl. -N(alkyl)2 -CN and -N3;
Het is a 5-7 membered heterocycle having 1,2,3 or 4 heteroatoms selected from N, O
and S, or a 5-6 membered heteroaryl having 1, 2,3 or 4 heteroatoms selected
from N, O and S, each optionally substituted from 1 to 2 times with a
substituent selected from the group consisting of halo, alkyl, oxo, -OH, -R2-OH,
-O-alkyl. -R2-O-alkyl, -NH2, -N(H)alkyl, -N(alkyl)2 -CN and -N3;
Q1 is a group of formula: -(R2)(Y1)(R2)c-R3
a, b and c are the same or different and are ea"ch independently 0 or 1 and at least
one of a or b is 1 ;
n is 0, 1,2,3 or 4;
Q2 is a group of formula: -(R2)aa-(Y2)bb-(R2)cc-R4
or two adjacent Q2 groups are selected from the group consisting of alkyl,
alkenyl, -OR7, -S(O)fR7 and -NR7R8 and together with the carbon atoms to
which they are bound, they form a C5-6cycloalkyl, C5-6cycloalkenyl, phenyl, 5-7
membered heterocycle having 1 or 2 heteroatoms selected from N, 0 and S, or
5-6 membered heteroaryl having 1 or 2 heteroatoms selected from N, 0 and S;
aa, bb and cc are the same or different and are each independently 0 or 1 ,•
each Y1 and Y2 is the same or different and is independently selected from the group
consisting of-O-, -S(O)f, -NfR7)-, -C(O)-, -OC(O)-, -CO2-, -C(O)N(R7)-,
-C(O)N(R7)S(O)2-, -OC(O)N(R7)-, -OS(O)2-, -S(O)2N(R7)-. -S(O)2N(R7)C(O)-,
-N(R7)S(O)2-, -N(R7)C(O)-, -N(R7)CO2- and -N(R7)C(O)N(R7)-;
each R2 is the same or different and is independently selected from the group
consisting of alkylene, alkenylene and alkynylene;
each R3 and R4 is the same or different and is each independently selected from the
group consisting of H, halo, alkyl, alkenyl, alkynyl, -C(O)R7, -C(0)NR7R8, -CO2R7,
-C(S)R7, -C(S)NR7R8. -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)fR7,
-S(O)2NR7R8, -NR7R8. -N(R7)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN, -N3and a group of
C5-10cycyIoaIkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from N, 0 and S and 5-10 membered heteroaryl
having 1,2 or 3 heteroatoms selected from N, 0 and S
each d is 0 or 1 ;
e is 0,1,2,3 or 4;
each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, Ph,
Het, -CH(OH)-R2-OH, -C(O)R7, -C02R7, -CO2-R2-Ph, -CO2-R2-Het,
-C(O)NR7R8, -C(O)N(R7)C(O)R7, -C(O)N(R7)CO2R7, -C(O)N(R7)C(O)NR7R8,
-C(O)N(R7)S(O)2R7, -C(S)R7. -C(S)NR7R8, -C(=NR7R8, -C(=NR7)NR7R8,
-CR7=N-OR8, =0, -OR7, -OC(O)R7, -OC(O)Ph, -OC(O)Het, -OC(O)NR7R8,
-O-R2-S(O)2R7, -S(O)fR7, -S(O)2NR7R8, -S(O)2Ph, -S(0)2Het. -NR7R8,
-N(R7)C(O)R8, -N(R7)CO2R8, -N(R7)-R2-CO2R8, -N(R7)C(O)NR7R8,
-N(R7)-R2-C(O)NR7R8, -N(R7)C(O)Ph, -N(R7)C(O)Het, -N(R7)Ph, -N(R7)Het,
-N(R7)C(O)NR7-R2-NR7R8, -N(R7)C(O)N(R7)Ph,-N(R7)C(O)N(R7)Het,
N(R7)C(O)N(R7)-R7-Het, -N(R7)S(O)2R8, -N(R7)-R2-S(O)2R8, -N02, -CN and
-N3;
wherein when Q1 is defined where b is 1 and c is 0, R3 is not halo, -C(0)R7, -C(O)NR7R8,
-CO2R7, -C(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7,
-S(O)rR7, -S(O)2NR7R8, -NR"R8, -N(R7)G(O)R8, N(R7)S(O)2R8, -N02, -CN or -N3;
wherein when Q2 is defined where bb is 1 and cc is O, R4 is not halo, -C(O)R7,
-C(O)NR7R8, -CO2R7, -C(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8,
-CR7=N-0R7, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8, -N(R7)C(O)R8, -N(R7)S(O)2R8,
-NO2,-CN or-N3;
R6 is selected from the group consisting of H, halo, alkyl. cycloalkyl, 0R7, -S(O)fR7,
-NR7R8, -NHC(0)R7, -NHC(O)NR7R8 and -NHS(O)2R7;
f is 0,1 or 2; and
each R7 and each R8 are the same or different and are each independently selected
from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl and
cycloalkenyl;
wherein when R1 is -CO2CH3 and n is 0, Q1 is not -OH;
it a pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof.
In another aspect of the invention there is provided a pharmaceutical composition
comprising a compound of formula (I). In one embodiment, the pharmaceutical
composition further comprises a pharmaceutically acceptable carrier, diluent or
excipient
In a third aspect of the invention, there is provided a method for treating a condition
mediated by PLK in an animal. The method comprises administering to the animal a
therapeuticaliy effective amount of a compound of formula (1) or a pharmaceuticaily
acceptable salt, solvate or physiologically functional derivative thereof.
In a fourth aspect-of the invention, there is provided a method for treating a
susceptible neoplasm in an animal. The method comprises administering to the animal
a therapeuticaliy effective amount of a compound of formula (I) or a pharmaceutically
acceptable salt solvate or physiologically functional derivative thereof. The
susceptible neoplasm may be selected from the group consisting of breast cancer,
colon cancer, lung cancer, prostate cancer, lymphoma, leukemia, endometrial cancer,
melanoma, pancreatic cancer, ovarian cancer, squamous carcinoma, carcinoma of the
head and neck, and esophageal carcinoma.
In a fifth aspect of the invention, there is provided a method for treating a condition
characterized by inappropriate cellular proliferation. The method comprises
contacting the cell with a therapeuticaliy effective amount of a compound of formula
(I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof.
In a sixth aspect, the present invention provides a method for inhibiting proliferation
of a cell. The method comprises contacting the cell with an amount of a compound of
farmula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof sufficient to inhibit proliferation of the cell.
In another aspect, the present invention provides a method for inhibiting mitosis in a
cell. The method comprises administering to the cell an amount of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof sufficient to inhibit mitosis in the cell.
In another aspect, there is provided a process for preparing a compound of formula (I)
comprising reacting a compound of formula (III):
wherein R10 is selected from the group consisting of alky), alkenyl, alkynyl,
cyctoalkyl, cycloalkenyl and suitable carboxylic acid protecting groups.
In another aspect, the present invention provides a radiolabeled compound of formula
(I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof. In one embodiment, the present invention provides a tritiated
compound of formula (I) or a pharmaceutically acceptable salt, solvate or
physiologically functional derivative thereof. In another aspect, the present invention
provides a biotinylated compound of formula (I) or a pharmaceutically acceptable salt,
solvate or physiologically functional derivative thereof.
In another aspect, the present invention provides a compound of formula (I) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in therapy.
in yet another aspect, the present invention provides a compound of formula (I) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in the treatment of a condition mediated by PLK in an animal.
In yet another aspect, the present invention provides a compound of formula (I) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in the treatment of a susceptible neoplasm in an animal.
In another aspect, the present invention provides a compound of formula (I) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in the treatment of a condition characterized by inappropriate cellular
proliferation.
In yet another aspect, the present invention provides a compound of formula (1) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in inhibiting proliferation of a cell.
In yet another aspect, the present invention provides a compound of formula (I) or a
pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof for use in inhibiting mitosis in a cell.
In yet another aspect, the present invention provides the use of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof for the preparation of a medicament for the treatment of condition
mediated by PLK in an animal.
In yet another aspect, the present invention provides the use of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof for the preparation of a medicament for the treatment of a
susceptible neoplasm in an animal.
In yet another aspect, the present invention provides the use of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof for the preparation of a medicament for the treatment of a
condition characterized by inappropriate cellular proliferation in an animal.
In yet another aspect, the present invention provides the use of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof for the preparation of a medicament for inhibiting proliferation of a
cell.
In yet another aspect, the present invention provides the use of a compound of
formula (I) or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof for the preparation of a medicament for inhibiting mitosis in a cell.
In yet another aspect, the present"invention provides a pharmaceutical composition
comprising a compound of formula (i) for use in the treatment of a susceptible
neoplasm in an animal.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, "a compound of the invention" or "a compound of formula (I)" means
a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or
physiologically functional derivative thereof. Similarly, with respect to isolatable
intermediates such as for example, compounds of formula (III) and (VIII) the phrase "a
compound of formula (number)" means a compound having that formula and
pharmaceutically acceptable salts, solvates and physiologically functional derivatives
thereof.
As used herein, the terms "alkyl" (and "alkylene") refer to straight or branched
hydrocarbon chains containing from 1 to 8 carbon atoms. Examples of "alkyl" as used
herein include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl,
isobutyl, isopropyl, and tert-butyl. Examples of "alkylene" as used herein include, but
atrnot limited to, methylene, ethylene, propylene, butylene, and isobutylene. "Alkyl"
also Includes substituted alkyl. The alkyl groups may be optionally substituted one or
more times with a halogen. Thus, the term "alkyl" includes trifluoromethyl and
trifluoroethyl, among other halogenated alkyls.
As used herein, the term "alkenyl" refers to straight or branched hydrocarbon chains
containing from 2 to 8 carbon atoms (unless a different number of atoms is specified)
and at least one and up to three carbon-carbon double bonds. Examples of "alkenyl"
as used herein include, but are not limited to ethenyl and propenyl. "Alkenyl" also
includes substituted alkenyl: The alkenyl groups may optionally be substituted one or
more times with a halogen.
As used herein, the term "alkynyl" refers to straight or branched hydrocarbon chains
containing from 2 to 8 carbon atoms (unless a different number of atoms is specified)
and at least one and up to three carbon-carbon triple bonds. Examples of "alkynyl" as
used herein include, but are not limited to ethynyl and propynyl. "Alkynyl" also
includes substituted alkynyl. The alkynyl groups may optionally be substituted one or
more times with a halogen.
As used herein, the term "cycloalkyl" refers to a non-aromatic monocyclic carbocyclic
ring having from 3 to 8 carbon atoms (unless a different number of atoms is specified)
and no carbon-carbon double bonds. "Cycloalkyl" includes by way of example
cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
"Cycloalkyl" also includes substituted cycloalkyl. The cycloalkyl may optionally be
substituted on any available carbon with one or more substituents selected from the
group consisting of halo, C1-3alkyl (including haloalkyl, e.g., perfluoroalkyl), -OH,
-O-C1-3aikyl, -NHa, -NH(C1-3alkyl) -N(C1-3alkyl)2 -CN and -N3. Preferred cycloalkyl
groups include C3-6cycloalkyl and substituted C3-6cycloalkyl.
As used herein, the term "cycloalkenyl" refers to a non-aromatic monocyclic
carbocyclic ring having from 3 to 8 carbon atoms (unless a different number of atoms
(specified) and up to 3 carbon-carbon double bonds. "Cycloalkenyl" includes by way
of example cyclobutenyl, cyclopentenyl and cyclohexenyl. "Cycloalkenyl" also includes
substituted cycloalkenyl. The cycloalkenyl may optionally be substituted on any
available carbon with one or more substituents selected from the group consisting of
halo, C1-3alkyl (including haloalkyl, e.g, perfluoroalkyl), -OH, -O-C1-3alkyl, -NH2,
-NH(C1-3alkyl) -N(C1-3alkyl)2, -CN and -N3
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine.
The term "oxo" as used herein refers to the group =0 attached directly to a carbon
atom of a hydrocarbon ring (i.e., cycloalkenyl, aryl, heterocycle or heteroaryl ring) as
well as -N-oxides, sulfones and sulfoxides wherein the N or S are atoms of a
heterocyclic or heteroaryl ring.
The term "aryl" refers to monocyclic carbocyclic groups and fused bicyclic carbocyclic
groups having from 6 to 13 carbon atoms (unless a different number of atoms is
specified) and having at least one aromatic ring. Examples of particular aryl groups
include but are not limited to phenyl and naphthyl. One particular aryl group
according to the invention is phenyl.
The terms "heterocycle" and "heterocyclic" refer to monocyclic saturated or
unsaturated non-aromatic groups and fused bicyclic saturated or unsaturated non-
aromatic groups, having the specified number of members and containing 1,2,3 or 4
heteroatoms selected from N, 0 and S (unless a different number of heteroatoms is
specified). Examples of particular heterocyclic groups include but are not limited to
tetrahydrofuran, dihydropyran, tetrahydropyran, pyran, tetrahydropyran, thietane, 1,4-
dioxane, 1,3-dioxane, 1,3-dioxalane, piperidine, piperazine, tetrahydropyrimidine,
pyrrolidine, morpholine, thiomorpholine, thiazolidine, oxazolidine,
tetrahydrothiopyran, tetrahydrothiophene, and the like.
The term "heteroaryl" refers to aromatic monocyclic groups and fused bicyclic groups
wherein at least one ring is aromatic, having the specified number of members and
containing 1,2,3, or 4 beteroatoms selected from N, O and S (unless a different
number of heteroatoms is specified). Examples of particular heteroaryl groups include
but are not limited to furan, thiophene, pyrrole, imidazole, pyrazole, triazole, tetrazole,
thiazole, oxazole, isoxazole, oxadiazole, thiadiazole, isothtazole, pyridine, pyridazine,
pyrazine, pyrimidine, quinoline, isoquinoline, benzofuran, benzothiophene, indole, and
indazole.
The term "members" (and variants thereof e.g., "membered") in the context of
heterocyclic and heteroaryl groups refers to the total atoms, carbon and heteroatoms
N, 0 and/or S, which form the ring. Thus, an example of a 6-membered heterocyclic
ring is piperidine and an example of a 6-membered heteroaryl ring is pyridine.
As used herein, the term "optionally" means that the subsequently described event(s)
may or may not occur, and includes both event(s) that occur and events that do not
occur.
wherein:
R1 is selected from the group consisting of H, alkyl, alkenyl, alkynyl, -C(O)R7, -CO2R7,
-C(O)NR7R8, -C(O)N(R7)OR8. -C(O)NR7)-R2-OR8, -C(O)N(R7)-Ph,
-C(O)NR7R8-Ph, -C(O)N(R7)C(O)R8, -C(O)N(R7)CO2R8, -C(O)N(R7)C(O)NR7R8,
_ -C(Q)N(R7)S(O)aRB1 -R2-OR7, -R2-O-C(O)R7,-C(S)R7,-C(S)NR7R",-C(S)N(R7)-Ph,
-C(S)N(R7)-R2-Ph, -R2-SR7, -C(=NR7)NR7R8, -C(=NR7)N(R8)-Ph,
-C(-NR7)N(R8)-R2-Ph, -R2-NR7R8, -CN, -OR7, -S(O)fR7, -S(O)2NR7R8,
-S(O)2N(R7)-Ph,-S(O)2N(R7)-R2-Ph, -NR7R8, N(R7)-Ph, -N(R7)-R2-Ph, -N(R7)-SO2R8
and Het;
Ph is phenyl optionally substituted from 1 to 3 times with a substituent selected from
the group consisting of halo, alkyl, -OH, -R2-OH, -O-alkyl. -R2-O-alkyl, -NH2,
-N(H)alkyl, -N(alkyl)2 -CN and -N3;
Het is a 5-7 membered heterocycle having 1,2,3 or 4 heteroatoms selected from N, 0
and S, or a 5-6 membered heteroaryl having 1,2,3 or 4 heteroatoms selected
from N, 0 and S, each optionally substituted from 1 to 2 times with a
substituent selected from the group consisting of halo, alkyl, oxo, -OH, -R2-0H,
-O-alkyl. -R2-O-alkyl. -NH2, -N(H)alkyl. -N(alkyl)2 -CN and -N3;
Q1 is a group of formula: -(R2)a(Y1)b(R2)c-R3
a, b and c are the same or different and are each independently 0 or 1 and at least
one of a or b is 1;
n is 0,1,2,3 or 4;
Q2 is a group of formula: -(R2)aa(Y2)bb(R2)cc-R4
or two adjacent Q2 groups are selected from the group consisting of alkyl,
alkenyl, -OR7, -S(O)fR7 and -NR7R8 and together with the carbon atoms to
which they are bound, they form a C5-6cycloalkyl, C5-6cycloalkenyl, phenyl, 5-7
membered heterocycle having 1 or 2 heteroatoms selected from N, 0 and S, or
5-6 membered heteroaryl having 1 or 2 heteroatoms selected from N, 0 and S;
aa, bb and cc are the same or different and are each independently 0 or 1 ;
each Y1 and Y2 is the same or different and is independently selected from the group
consisting of-O-, -S(O) -N(R7)-, -C(O)-, -0C(0)-. -C02-, -C(O)N(R7)-,
-C(O)N(R7)S(O)2-, -OC(O)N(R7)-, -OS(O)2-. -S(O)2N(R7)-, -S(O)2N(R7)C(O)-,
-N(R7)S(O)2-, -N(R7)C(O)-, -N(R7)CO2- and -N(R7)C(O)N(R7)-;
each R2 is the same or different and is independently selected from the group
consisting of alkylene, alkenylene and alkynylene;
each R3 and R4 is the same or different and is each independently selected from the
group consisting of H, halo, alkyl, alkenyl, alkynyl, -C(0)R7, -C(0)NR7R8, -CO2R7,
-C(S)R7, -G(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)rR7,
-S(O)2NR7R8, -NR7R8, -N(R7)C(O)R8. -N(R7)S(0)2R8, -N02. -CN1 -N3 and a group of
wherein:
Ring A is selected from the group consisting of C5-10cycloalkyl,
C5-10ocycyloalkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from N, 0 and S and 5-10 membered heteroaryl
having 1,2 or 3 heteroatoms selected from N, 0 and S
each d is 0 or 1;
e is O, 1,2,3 or 4;
each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl,Ph,
Het, -CH(OH)-R2)H, -C(O)R7, -CO2R7, -C02-R2-Ph. -CO2-R2-Het,
-C(O)NR7R8, -G(O)N(R7)C(O)R7, -C(O)N(R7)CO2R7. -C(O)N(R7)C(O)NR7R8,
-C(O)N(R7)S(0)2R7, -C(S)R7. -C(S)NR7R8, -C(=NR7)R8, -C(-NR7)NR7R8,
-CR7=N-OR8, =0, -OR7, -OC(O)R7, -OC(O)Ph. -OC(O)Het, -0C(0)NR7R8,
-0-R2-S(O)2R7. -S(O)fR7, -S(O)2NR7R8, -S(O)2Ph. -S(O)2Het, -NR7R8,
-N(R7)C(O)R8, -N(R7)CO2R8, -N(R7)-CO2R8, -N(R7)C(O)NR7R8,
-N(R7)-R2-C(O)NR7R8, -N(R7)C(O)Ph, -N(R7)C(O)Het, -N(R7)Ph, -N(R7)Het,
-N(R7)C(O)NR7-R2-NR2R8, -N(R7)C(O)N(R7)Ph, -N(R7)C(O)N(R7)Het,
-N(R7)C(O)N(R7)-R8-Het, -N(R7)S(0)2R8. -N(R7)-R2-S(0)2R8, -NO2, -CN and
-N3;
R5 is selected from the group consisting of H, halo, alkyl, cycloalkyl. OR7, -S(O)fR7,
-NR7R8, -NHC(O)R7, -NHC(O)NR7R8 and -NHS(O)2R7;
f is 0,1 or 2; and
each R7 and each RB are the same or different and are each independently selected
from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl and
cycloalkenyl;
and pharmaceutically acceptable salts, solvates and physiologically functional
derivatives thereof.
In the embodiment, the compounds of formula (I) are defined wherein R1 is selected
from the group consisting of alkyl. alkenyl, alkynyl, -C(O)R7, -CO2R7, -C(O)NR7R8.
-C(O)N(R7)-OR8, -R2-OR7, -C(S)NR7R8. -C(=NR7)NR7R8, -CN, -S(O)fR7, -S(O)2NR7R8,
and Het, or any subset thereof. In one embodiment, the compounds of formula (I) are
defined wherein R1 is selected from the group consisting of -C(O)R7, -CO2R7,
-C(S)NR7R8. Het, and -C(O)NR7R8, or any subset thereof. In one embodiment, the
compounds of formula (I) are defined wherein R1 is selected from the group consisting
of -C(O)R7, -CO2R7 and -C(O)NR7R8, or any subset thereof. In one particular
embodiment, R1 is selected from the group consisting of -CO2R7 and -C(O)NR7R8, or
any subset thereof. In one embodiment R1 is -CO2R7. In one embodiment, R1 is
-C(O)NR7R8.
Specific examples of groups defining R1 include but are not limited to -COH, -COCH3,
-COOH, -COOCH3, -C(O)NH2, -CONH(alkyl). -CON(alkyl)(alkyl). -CONH(Et-OH),
-CONH(benzyl), -CONH(phenyl), -S(O)2NH2 and -S(O)2N(H)CH3, -CH2OH. -C(S)NH2, -CN,
and -tetrazole, or any subset thereof. In one particular embodiment, R1 is selected
from the group consisting of -CO2H and -C(O)NH2.
Q1 is defined as a group of formula: -(R2)a-(Y1)b(R2)c-R3.
In the foregoing formula, a, b and c are the same or different and are each
independently 0 or 1.
In one embodiment, Q1 is defined wherein a is 0. In the embodiment wheren a is 1
and thus the (R2)a group is present, R2 is typically alkylene or alkenylene, more
particularly alkylene. In one particular embodiment, Q1 is defined where a is 1 and
(R2)a is C1-3alkylene.
In one embodiment, Q1 in the compounds of formula (I) is defined where b is 1; thus
Y1 is present In one such embodiment, Y1 is selected from -O-, -S(O)f-, -N(R7)-,
-C(O)-, -OC(O)-, -CO2-, -C(O)N(R7)-, -C(O)N(R7)S(O)2-, -OC(O)N(R7)-, -OS(O)2-,
-S(O)2N(R7)-, -S(O)2N(R7)C(O)-. -N(R7)S(O)2-, -N(R7)C(O)-, -N(R7)CO2- and
(R7)C(O)N(R7)-. In one particular embodiment, Y1 is selected from -0-, -N(R7)-,
-C(O)-. -OC(O)-, -C(O)N(R7)-, -0S(0)2-, -S(O)2N(R7)-, N(R7)S(O)2-, and -N(R7)C(O)-, or
any subset thereof. In another particular embodiment, Yl is selected from -0-,
-N(R7)-, -C(0)-, -OS(O)2- N(R7)S(O)2-, and -N(R7)C(O)-, or any subset therof. In one
particular embodiment, b is 1 and Y" is -0-, -ISKR7)-, -C(O)- or -OS(O)2-, or any subset
thereof. In one particular embodiment, b is 1 and y1 is -O-. In another paticular
embodiment, b is 1 and Y1 is-N(R7)- and R7 is H or alkyl, more particularly H. In
another particular embodiment, b is 1 and Y1 is -C(0)-. In another particular
embodiment, b is 1 and Y1 is -OS(O)2-.
The variable c in the formula Q1 can be 0 or 1. In one embodiment, c is 1. In one such
embodiment (R2)c is alkylene or alkenylene, more particularly alkylene. In one
particular embodiment, Q1 is defined where c is 1 and (R2)c is C1-3alkylene.
In one embodiment of the invention, the compounds of formula (I) are defined to
include a substitution at the position indicated by Q1; thus, when a, b and c are all 0,
then R3 is not H. In one particular embodiment the compounds of the present
invention are defined wherein, at least one of a or b is 1. In one particular
embodiment, Q1 is defined wherein both b and c are 1. In one particular embodiment,
Q1 is defined wherein a is 0 and both b and c are 1.
Consistent with the definition of b, Y1 and c, the group R3 may be selected from the
group consisting of H, halo, alkyl, alkenyl, alkynyl, -C(O)R7, -C(O)NR7R8, -CO2R7, -C(S)R7,
-C(S)NR7R8. -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)rR7. -S(O)2NR7R8,
-NR7R8. -N(R7)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN, -N3 and a group of formula (ii):
In one embodiment, R3 in the definition of Q1 is selected from the group consisting of
H, alkyl, alkenyl, alkynyl, and a group of formula (ii), or any subset thereof. In one
Particular embodiment, R3 is selected from the group consisting of H, alkyl, alkenyl
and alkynyl, or any subset thereof. In one embodiment, when R3 is alkyl, R3 is C2-6alkyl.
In one particular embodiment, R3 is a group of formula (ii).
( A ) in formula (ii) is referred to herein as "Ring A." Ring A is selected from
Cs-iocycloalkyl, C5-10cycloalkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from N, 0 and S and 5-10 membered heteroaryl having 1,2 or 3
heteroatoms selected from N, 0 and S. In Q1 Ring A may be bonded to R2, Yl (when c
is 0) or the thiophene ring (when a, b and c are 0) through any suitable carbon or
heteroatom. In one embodiment, Q1 is defined wherein R3 is a group of formula (ii)
and Ring A is selected from C5-10cycloalkyl, C5-10 cycloalkenyl, aryl, 5-10 membered
heterocycle having 1,2 or 3 heteroatoms selected from N, 0 and S and 5-10
membered heteroaryl having 1,2 or 3 heteroatoms selected from N, 0 and S. In one
embodiment, Q1 is defined wherein R3 is a group of formula (ii) and Ring A is selected
from aryl, 5-10 membered heterocycle having 1,2 or 3 heteroatoms selected from N,
0 and S and 5-10 membered heteroaryl having 1,2 or 3 heteroatoms selected from N,
0 and S. In one particular embodiment, Q1 is defined wherein R3 is a group of formula
(ii) and Ring A is selected from aryl and 5-10 membered heteroaryl having 1,2 or 3
heteroatoms selected from N. 0 and S.
In one embodiment, Q1 is defined wherein R3 is a group of formula (ii) and Ring A is
selected from the group consisting of cycloalkyl, tetrahydropyran, tetrahydrofuran,
morpholine, piperidine, phenyl, naphthyl, thiophene, furan, pyrrole, pyrrolidine,
pyrrolidinone, imidazole, benzofuran, benzimidazole, pyridyl,
or any subset thereof. In one particular embodiment, Ring A is phenyl. In one
particular embodiment Ring A is pyridyl.
Particular, more specific, examples of groups defining Q1 in the compounds of formula
(I) are selected from the group consisting of:
In one embodiment the compounds of formula (I) are defined wherein R3 is a group of
formula (ii) and d is 0 or 1. In a particular embodiment, wherein R3 is a group of
formula (ii) and d is 1, R2 is C1-3alkylene. In one embodiment, d is 0.
In one embodiment, wherein the compounds of formula (1) are defined wherein R3 is a
group of formula (ii), e is 0,1,2 or 3. In one particular embodiment, e is 0 or 1. In
one embodiment, e is 1. In one embodiment, e is 2.
In one embodiment, wherein the compounds of formula (I) are defined wherein R3 is a
group of formula (ii), each R6 is the same or different and is independently selected
from the group consisting of H, halo, alkyl, alkenyl, alkynyl, cycloalkyl, Ph, Het,
-CH(OH)-R2-OH, -C(O)R7, -C(O)NR7R8, =0, -OR7, -S(O)fR7, -S(0)2NR7R8, -SO2Ph, -NR7RB.
-N(R7)C(O)R8, -N(R7)C02R8, -N(R7)S(O)2R8, -NO2. -CN and -N3, or any subset thereof. In
one particular embodiment, R3 is a group of formula (ii) and each R6 is the same or
different and is independently selected from the group consisting of H, halo, alkyl,
alkenyl, alkynyl, cycloalkyl, -OR7. -S(0)rR7, -S(0)2NR7R8, -NR7R8, -N(R7)S(O)2R8, -NO2 and
-CN or any subset thereof. In one particular embodiment, R3 is a group of formula (ii)
and each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyl, -OR7, -S(0)fR7, -S(O)2NR7R8 and -NO2, or any subset
thereof.
More specifically, in one embodiment wherein R3 is a group of formula (ii), each R6 is
the same or different and is independently selected from the group consisting of H, F,
Cl, Br, I, methyl, trifluoromethyl, ethyl, propyl, isopropyl, cycloprbpyl, iso-butyl, t-butyl,
ethenyl, propenyl, acetylene, 0-methyl, O-difluoromethyl, Q-trifluoromethyl, O-ethyl,
O-propyl. O-isopropyl, O-cyclopropyl, -S02-methyl, -SO2NH2, -NH2, -NH(alkyl),
-N(alkyl)alkyl, -NH(cyclopropyl), -NHS02-methyl, -NO2, and -CN, or any subset
thereof.
In one embodiment, Q1 is defined such that when b is 1 and c is 0, R3 is not halo,
-C(O)R7, -C(O)NR7R8, -CO2R7, -C(S)R7, -C(S)NR7R8, -C(=NR7)R8. -C(=NR7)NR7R8,
-CR7=N-OR7, -OR7, -S(O)fR7, -S(0)2NR7R8, -NR7R8, -N(R7)C(O)R8, -N(R7)S(O)iR8, -NO2, -CN
or -N3.
In one embodiment, wherein when R1 is -CO2CH3 and n is 0, Q1 is not -OH. In one
embodiment, Q1 is not -OH.
In one embodiment, n is 0,1 or 2, or any subset thereof- In one particular
embodiment, n is 0, and thus the benzimidazcHe ring is unsubstituted at positions C-4,
C-5, C-6 and C-7. In one embodiment, n is 2 and Q2 is at C-5 and C-6. In another
particular embodiment, n is 1. In one particular embodiment n is 2.
Q2 is a group of formula -(R2)aa-(y2)bb-(R2)a-R4 Q2 may be located at any of C-4, C-5,
C-6 and/or C-7 of the benzimidazole ring. In one embodiment, n is 1 and Q2 is at C-5.
In one embodiment, n is 1 and Q2 is at C-6.
In the foregoing formula, aa, bb and cc are the same or different and are each
independently 0 or 1.
In one embodiment, aa is 0; thus the group (R2)aa is not present. In the embodiment
wherein aa is 1, (R2)aa is typically alkylene or alkenylene, more particularly alkylene. In
one particular embodiment, Q2 is defined where aa is 1 and (R2)aa. is C1-3alkylene.
In one embodiment, the compounds of formula (I) are defined wherein bb is 0. In
another embodiment, Q2 in the compounds of formula (I) is defined where bb is 1;
thus Y2 is present. In one such embodiment, Y2 is selected from -O-, -S(O)i-, -N(R7)-,
-C(O)-, -0C(0) -CO*-, -C(O)N(R7)-, -C(O)N(R7)S(O)2-, -OC(O)N(R7)-, -OS(O)2-,
-S(O)2(R7)-, -S(O)2N(R7)C(O)-, -N(R7)S(O)2-. -N(R7)C(O)-, -N(R7)CO2- and
-N(R7)C(O)N(R7)-. In one particular embodiment, bb is 1 and Y2 is selected from -O-,
-S(0) -N(R7)-. -C(O)-, -OC(O)-, -CO2-, -C(O)N(R7)-, -OS(O)2-, -N(R7)S(O)2-,
-N(R7)C(O)-, -N(R7)CO2- and -N(R7)C(O)N(R7)-, or any subset thereof. In another
particular embodiment, bb is 1 and Y2 is selected from -O-, -S(O)f-, -N(R7)-, -CO2-,
-C(O)N(R7)-, -N(R7)S(O)2-, and -N(R7)C(O)-N(|P)CO2- N(R7)C(O)N(R7)-, or any subset
thereof. In one particular embodiment, Q2 is defined wherein bb is 1 and Y2 is selected
from -O-, -S(O)f-, -N(R7)-. -CO2- and -C(O)N(R7)-, or any subset thereof. In one
particular embodiment, Q2 is defined wherein bb is 1 and Y2 is -O-. In one particular
embodiment, Q2 is defined wherein bb is 1 and Y2 is -S(0)f, wherein f is 2. In another
particular embodiment, bb is 1 and Y2 is -N(R7)- and R7 is H or alkyl, more particularly
H. In another particular embodiment, bb is 1 and Y2 is -CO2-. In another particular
embodiment, bb is 1 and Y2 is -C(O)N(R7)-.
The variable cc in the formula Q2 can be 0 or 1. In one embodiment, cc is 1. In one
such embodiment (R2)cc is alkylene or alkenylene, more particularly alkylene. In one
particular embodiment, Q2 is defined where cc is 1 and (R2)cc is C1-3alkylene.
Consistent with the definition of bb, Y2 and cc, the group R4 may be selected from the
group consisting of H, halo, alkyl. alkenyl, alkynyl, -C(O)R7, -C(O)NR7R8, -CO2R7, -C(S)R7,
-C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)fR7. -S(O)2NR7R8,
-NR7R8, -N(R7)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN, -N3 and a group of formula (ii):
In one embodiment R4 in the definition of Q2 is selected from the group consisting H,
halo, alkyl, alkenyl, alkynyl, -C(O)NR7R8, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8,
N(R7)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN, -N3and a group of formula (ii), or any subset
thereof. In one particular embodiment, R4 is selected from the group consisting of H,
halo, alkyl, -OR7, -S(O)rR7, -S(O)2NR7R8, -NR7R8, and a group of formula (ii), or any
subset thereof. In one embodiment, R4 is selected from H, halo, alkyl, -OR7, -NR7R8,
and a group of formula (ii), or any subset thereof.
In one particular embodiment, R4 is a group of formula (ii). In the embodiment,
wherein R4 is a group of formula (ii), Ring A is selected from C5-10cycloalkyl,
C5-10cycloalkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3 heteroatoms
selected from N, 0 and S and 5-10 membered heteroaryl having 1,2 or 3 heteroatoms
selected from N, 0 and S. In one embodiment wherein R4 is a group of formula (ii),
Ring A is selected from C5-6cycloalkyl, C5-6cycloalkenyl, aryl, 5-10 membered
heterocycle having 1,2 or 3 heteroatoms selected from N, 0 and S and 5-10
membered heteroaryl having 1,2 or 3 heteroatoms selected from N, 0 and S. In Q2,
Ring A may be bonded to the R2, Y2 (when cc is 0) or the benzimidazole (when aa, bb
and cc are 0) through any suitable carbon or heteroatom. In one embodiment, Q2 is
defined wherein R4 is a group of formula (ii) and Ring A is selected from aryl, 5-10
membered heterocycle having 1.2 or 3 heteroatoms selected from N, 0 and S and 5-
10 membered heteroaryl having 1,2 or 3 heteroatoms selected from N, 0 and S. In
one particular embodiment, Q2 is defined wherein R4 is a group of formula (ii) and
Ring A is selected from aryl and 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from N, 0 and S.
In one embodiment, Q2 is defined wherein R4 is a group of formula (ii) and Ring A.is
selected from the group consisting of cycloalkyl, oxetane, oxazole, thiazole,
morpholine, piperidine, piperazine, phenyl, naphthyl, thiophene, furan, pyrrolidine,
pyrrolidinone, imidazole, triazole, imidazolidinone, benzofuran, benzodioxolane,
benzimidazole and pyridyl, or any subset thereof. In one particular embodiment, Ring
A is selected from morpholine, piperidine, piperazine, phenyl, pyrrolidinone,
imidazoiidinone and pyrrolidine, or any subset thereof.
More specifically, in one embodiment each R4 is the same or different and is
independently selected from the group consisting of H, F, Cl, Br, I, methyl,
trifluoromethyl, ethyl, propyl, isopropyl, cyclopropyl, iso-butyl, t-butyl, ethenyl,
propenyl, acetylene, O-methyl, O-trifluoromethyl, O-ethyl, O-propyl, O-isopropyl, O-
cyclopropyl, -SO2-methyl, -SO2NH2, -NH2, -NH(alkyl), -N(alkyl)alkyl,
-NH(cyclopropyl), -NHC(O)-methyl, -NHC(0)NH2, -NHSO2-methyl, morpholino and
piperizinyl, or any subset thereof.
particular, more specific, examples of groups defining Q2 in the compounds of formula
(I) are selected from the group consisting of:
In one embodiment, Q2 is -O-alkyl. In one particular embodiment, Q2 is halo.
In one embodiment the compounds of formula (I) are defined wherein R4 is a group of
formula (ii) and d is 0 or 1. In a particular embodiment, wherein R4 is a group of
formula (ii) and d is 1, R2 is Ci-3alkylene. In one embodiment, d is 0.
In one embodiment wherein the compounds of formula (I) are defined wherein R4 is a
group of formula (ii), e is 0,1,2 or 3. In one particular embodiment, e is 0 or 1. In
one embodiment, e is 0. In one embodiment, e is 1. In one embodiment, e is 2.
In one embodiment wherein the compounds of formula (I) are defined wherein R4 is a
group of formula (ii), each R6 is the same or different and is independently selected
from the group consisting of H, halo, alkyl, alkenyl, alkynyl, Het, -C(O)R7, -CChR7,
-C(O)NR7R8, =0, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R" and -N(R7)S(O)2R8, or any subset
thereof. In one particular embodiment, each R6 is the same or different and is
independently selected from the group consisting of H, halo, alkyl. =O. -OR7, -S(O)fR7,
(O)2NR7R8 and -NR7R8, or any subset thereof.
More specifically, in one embodiment, each R6 is the same or different and is
independently selected from the group consisting of H, methyl, ethyl, propyl,
isopropyl, iso-butyl, t-butyl, ethenyl, propenyl/cyclopropyl, pyrimidyl, -C(O)-alkyl,
-CO2-alkyl, -C(O)NH2, acetylene, oxo, O-methyl, O-ethyl, O-propyl, O-isopropyl, O-
cyclopropyl, -SO2-methyl, -SO2NH2, -NH2, -NH(alkyl),
-N(alkyl)alkyl, -NH(cyclopropyl) and -NHS02-methyl, or any subset thereof.
In another embodiment of the present invention, two adjacent Qz groups are selected
from the group consisting of alkyl, alkenyl, -OR7, -S(0)fR7 and -NR7R8 and together
with the carbon atoms to which they are bound, they form a C5-6cycloalkyl,
C5-6cycloalkenyl,phenyl,5-7 membered heterocycle having 1 or 2 heteroatoms
selected"from N, 0 and S, or 5-p membered heteroaryl having 1 or 2 heteroatoms
selected from N, 0 and S. By "two adjacent Q2 groups" is meant that two Q2 groups
are bonded to adjacent carbon atoms (e.g., C-4 and C-5). For example, in one
embodiment two adjacent Q2 groups are -OR7 and together with the atoms to which
they are bonded, they form a heterocyclic group such as:
In another embodiment, two adjacent Q2 groups are alkyl and together with the atoms
to which they are bonded, they form a cycloalkyl group such as:
In another embodiment two adjacent Q2 groups are defined as -OR7 and -NR7R8
respectively and together with the atoms to which they are bonded, they form a
heterocyclic group such as:
from these examples, additional embodiments can be readily ascertained by those
skilled in the art. Preferably the compounds of formula (I) are defined wherein when
n is 2, two adjacent Q2 groups together with the atoms to which they are bonded do
not form a C5-6cycloalkyl, C5-6cycloalkenyl, phenyl, 5-7 membered heterocycle having 1
or 2 heteroatoms selected from N, 0 and S, or 5-6 membered heteroaryl having 1 or 2
heteroatoms selected from N, 0 and S.
In one embodiment, Q2 is defined such that when bb is 1 and cc is O, R4 is not halo,
-C(O)R7, -C(O)NR7Ra, -CO2R7, -C(S)R7, -C(S)NR7R8. -C(-NR7)RB. -C(=NR7)NR8,
-CR7=N-OR7, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8, -N(R7)C(O)R8. -N(R7)S(O)2R8,
-NO2,-CN or-N3;
In one embodiment, R5 is selected from the group consisting of H, halo, alkyl, -NR7R8
and -S(O)fR7, or any subset thereof. In another embodiment, R5 is selected from the
group consisting of H, halo, alkyl and -NR7RS, or any subset thereof, in one particular
embodiment, R5 is H. In one particular embodiment, R5 is -NH2.
More specifically, in one embodiment, R5 is selected from the group consisting of H, F,
Cl, Br, I, methyl, trifluoromethyl, ethyl, propyl, isopropyl, -S-methyl, -SO2-methyl and
-NH2, or any subset thereof.
wherein all variables are as defined above, and pharmaceutically acceptable salts,
solvates and physiologically functional derivatives thereof.
The present invention also provides compounds of formula (1b):
wherein each R9 is the same or different and is selected from H, halo and alkyl; and all
other variables are as defined above, and pharmaceuticaily acceptable salts, solvates
and physiologically functional derivatives thereof.
It is to be understood that the present invention includes all combinations and subsets
of the particular groups defined hereinabove.
Specific compounds of formula (I) include but are not limited to those compounds
described in the Example section that follows. Some particular compounds of formula
(I) include but are not limited to:
5-(5,6-Dimethoxy-1H-ben2imidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzylloxy}thiophene-2-carboxamide;
5-(5-(Methyloxy)-6-{[2-(4-methyl-1-piperazinyl)ethyl]oxy}-1H-benzimidazol-1-yl)-
3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxamide;
3-[1 -(2-Chlorophenyl)ethoxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1 -yl)thiophene-2-
carboxamide;
5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-[1-(2-methylphenyl)ethoxy] thiophene-2-
carboxamide;
5-(5-Amino-1 H-benzimidazol-1 -y))-3-[1 -(2-chlorophenyl)ethoxy]thiophene-2~
carboxamide;
5-{6-[(4-Piperidinylmethyl)oxy]-1 H-benzimidazol-1-yl}-3-({[2-
(trifluoromethyl)phenyll-methyl}oxy)-2-thiophenecarboxamide;
5-(6-(Methyloxy)-5-{[3-(2-oxo-1-pyrrolidinyl)propyl]oxy}-1H-benzimidazol-1-yl)-3-
({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxamide;
5-6-{[3-(Dimethylamino)propyl]oxy}-5-(methyloxy)-H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxamide;
5-(5-(Methyloxy)-6-{[2-(4-morpholinyl)ethyl]oxy}-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxamide;
5-[6-(2-Morpholin-4-yiethoxy)-1 H-benzimidazol-1 -yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide;
5-[6-(2-Pyrrolidin-1-ylethoxy)-1H-benzimidazoh1-yl]-3-{[2-
(trlfluoromethyi)benzyl]oxy}thiophene-2-carboxamider
5-[5-Fluoro-6-(2-morpholin-4-ylethoxy)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide;
5-[6-(Methylsulfonyl)-1H-benzimidazol-1-yl]-3-{[2-(trifIuoromethyl)benzyl]oxy}-
thiophene-2-carboxamide;
3-[(3-Bromopyridin-4-yl)methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
5-(5l6-Dimethoxy-H-benzimidazol-1-yl)-3-{[2-(trifluoromethoxy)benzyl] oxy}
thiophene-2-carboxamide;
3-{[2-(Difluoromethoxy)benzyl]oxy}-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
3-[(2-Chloropyridin-3-yl)methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
.5-(5,6-Dimethoxy-1 H-benzimidazol-1 -yl)-3-[(2-f luoropyridin-3-
yI)methoxy]thiophene-2-carboxamide;
3-[(2-Aminopyridin-4-yl)methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
3-[(6-Chloro-1,3-benzodioxol-5-yl}methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
5-(5,6-Dimethoxy-1H benzimidazol-1-yl)-3-[(2-nitrobenzyl)oxy]thiophene-2-
carboxamide;
3-[(3-Aminobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
5i-(6-Broino-1H-benzimJdazol-1-yl)-3-{[2-(trifluoromethyI)benzyl]-oxy}thiophene-2-
carboxamide;
3-[(2,6-Dichlorobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
3-[(2-Bromobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
5-(5,6-Dimethoxy-1H-benzimidazol-l-yl)-3-[(2-formylbenzyl)oxy]thiophene-2-
carboxamide;
5-(1H-Benzimidazol-1-yl)-3-{(2-(trifluoromethyl)benzyl]oxy}thiophene-2-
carboxamide;
5-(1H-BenzimidazoI-1-yl)-3-[(2-nitrobenzyl)oxy]thiophene-2-carboxamide;
5-{6-Methoxy-1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)benzyl]oxy}thiophenc-
2-carboxamide;
2-(Aminocarbonyl)-5-(5,6-dimethoxy-1 H-benzimidazol-1 -yl)thien-3-yl 2-
methylbenzenesulfonate
and pharmaceutically acceptable salts, solvates and physiologically functional
derivatives thereof.
It will be appreciated by those skilled in the art that the compounds of the present
invention may also be utilized in the form of a pharmaceutically acceptable salt or
solvate or physiologically functional derivative thereof. The pharmaceutically
acceptable salts of the compounds of formula (I) include conventional salts formed
from pharmaceutically acceptable inorganic or organic acids or bases as well as
quaternary ammonium salts. More specific examples of suitable acid salts include
hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, perchloric, fumaric, acetic,
propionic, succinic, glycolic, formic, lactic, maleic, tartaric, citric, palmoic, malonic,
hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, fumaric, toluenesulfonic,
methanesulfonic (mesylate), naphthalene-2-sulfonic, benzenesulfonic
hydroxynaphthoic, hydroiodic, malic, steroic, tannic and the like.
Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may
be useful in the preparation of salts useful as intermediates in obtaining the
compounds of the invention and their pharmaceutically acceptable salts. More
specific examples of suitable basic salts include sodium, lithium, potassium,
magnesium, aluminium, calcium, zinc, N.N"-dibe"nzylethylenediamine, chloroprocaine,
choline, diethanolamine. ethylenediamine, N-methylglucamine and procaine salts.
The term "solvate" as used herein refers to a complex of variable stoichiometry formed
by a solute (a compound of formula (I)) and a solvent. Solvents, by way of example,
include water, methanol, ethanol, or acetic acid.
The term "physiologically functional derivative" as used herein refers to any
pharmaceutically acceptable derivative of a compound of the present invention, for
example, an ester or an amide of a compound of formula (I), which upon
administration to an animal, particularly a mammal, such as a human, is capable of
providing (directly or indirectly) a compound of the present invention or an active
metabolite thereof See, for example, Burger"s Medicinal Chemistry And Drug
Discovery, 5th Edition, Vol 1: Principles And Practice.
Processes for preparing pharmaceutically acceptable salts, soivates and physiologically
functional derivatives of the compounds of formula (I) are conventional in the art.
See, e.g., Burger"s Medicinal Chemistry And Drug Discovery 5th Edition, Vol 1:
Principles And Practice.
As will be apparent to those skilled in the art, in the processes described below for the
preparation of compounds of formula (I), certain intermediates, may be in the form of
pharmaceutically acceptable salts, soivates or physiologically functional derivatives of
the compound. Those terms as applied to any intermediate employed in the process of
preparing compounds of formula (I) have the same meanings as noted above with
respect to compounds of formula (I). Processes for preparing pharmaceutically
acceptable salts, soivates and physiologically functional derivatives of such
tenediates are known in the art and are analogous to the process for preparing
pharmaceutically acceptable salts, solvates and physiologically functional derivatives
of the compounds of formula (I).
Certain compounds of formula (I) may exist in stereoisomeric forms (e.g. they may
contain one or more asymmetric carbon atoms or may exhibit cis-trans isomerism).
The individual stereoisomers (enantiomers and diastereomers) and mixtures of these
are included within the scope of the present invention. The present invention also
covers the individual isomers of the compounds represented by formula (I) as mixtures
with isomers thereof in which one or more chiral centres are inverted. Certain
compounds of formula (l}may be prepared as a mixture of regioisomers. The present
invention covers both the mixture of regioisomers as well as the individual
compounds. Likewise, it is understood that compounds of formula (I) may exist in.
tautomeric forms other than that shown in the formula and these are also included
within the scope of the present invention. In one particular embodiment of the
present invention, the chiral compounds are present in the R conformation (i.e., the R-
isomer of the compound).
The compounds of the present invention are typically inhibitors of PLK. By PLK
inhibitor is meant a compound which exhibits pIC50 greater than 4 in the PLK
Inhibition assay described below in the examples or an IC50 less than 100 µM in the
Methylene Blue Growth Inhibition assay described below in the examples; more
particularly a PLK inhibitor is a compound which exhibits a pICso greater than 5 or an
IC50 less than 10 µM using the methods described in the examples below.
The present invention further provides compounds of formula (I) for use in medical
therapy in an animal, e.g. a mammal such as a human. In particular, the present
invention provides compounds of formula (I) for use in the treatment of a condition
mediated by PLK. The present invention also provides compounds of formula (I) for
use in the treatment of a susceptible neoplasm. The present invention provides
compounds of formula (I) for use in treating a condition characterized by
Imppropriate cellular proliferation. The present invention also provides compounds of
formula (I) for use in inhibiting proliferation of a cell. The present invention also
provides compounds of formula (I) for use in inhibiting mitosis in a cell.
The present invention provides methods for the treatment of several conditions or
diseases, all of which comprise the step of administering a therapeutically effective
amount of a compound of formula (I). As used herein, the term "treatment" refers to
alleviating the specified condition, eliminating or reducing the symptoms of the
condition, slowing or eliminating the progression of the condition and preventing or
delaying the reoccurrance of the condition in a previously afflicted subject
As used herein, the term "therapeufrcally effective amount" means an amount of a
compound of formula (I) which is sufficient, in the subject to which it is administered,
to elicit the biological or medical response of a cell culture, tissue, system, animal
(including human) that is being sought, for instance, by a researcher or clinician. For
example, a therapeutically effective amount of a compound of formula (I) for the
treatment of a condition mediated by PLK is an amount sufficient to treat the PLK
mediated condition in the subject Similarly, a therapeutically effective amount of a
compound of formula (I) for the treatment of a susceptible neoplasm is an amount
sufficient to treat the susceptible neoplasm in the subject. In one embodiment of the
present invention, the therapeutically effective amount of a compound of formula 0)
is an amount sufficient to inhibit cell mitosis. In one embodiment of the present
invention, a therapeutically effective amount of a compound of formula (I) is an
amount sufficient to regulate, modulate, bind or inhibit PLK.
The precise therapeutically effective amount of the compounds of formula (I) will
depend on a number of factors including, but not limited to, the age and weight of
the subject being treated, the precise disorder requiring treatment and its severity, the
nature of the formulation, and the route of administration, and will ultimately be at
the discretion of the attendant physcian or veternarian. Typically, the compound of
formula (I) will be given for treatment in the range of 0.1 to 200 mg/kg body weight
of recipient (animal) per day and more usually in the range of 1 to 100 mg/kg body
weight per day. Acceptable daily dosages, may be from about 0.1 to about 2000
mg/day, and preferably from about 0.1 to about 100 mg/day.
As one aspect, the present invention provides methods of regulating, modulating,
binding, or inhibiting PLK for the treatment of conditions mediated by PLK.
"Regulating, modulating, binding or inhibiting PLK" refers to regulating, modulating,
binding or inhibiting PLK activity, as well as regulating, modulating, binding or
inhibiting overexpression of PLK. Such conditions include certain neoplasms
(including cancers and tumors) which have been associated with PLK and conditions
characterized by inappropriate cellular proliferation.
The present invention provides a method for treating a condition mediated by PLK in
an animal such as a mammal (e.g., a human), which method comprises administering
to the animal a therapeuticaily effective amount of the compound of formula (I).
Conditions which are mediated by PLK are known in the art and include but are not
limited to neoplasms and conditions characterized by inappropriate cellular
proliferation.
The present invention also provides a method for treating a susceptible neoplasm
(cancer or tumor) in an animal such as a mammal (e.g., a human), which method
comprises administering to the animal a therapeuticaily effective amount of the
compound of formula (I). "Susceptible neoplasm" as used herein refers to neoplasms
which are susceptible to treatment with a PLK inhibitor. Neoplasms which have been
associated with PLK and are therefor susceptible to treatment with a PLK inhibitor are
known in the art, and include both primary and metastatic tumors and cancers. For
example, susceptible neoplasms within the scope of the present invention include but
are not limited to breast cancer, colon cancer, lung cancer (including small cell lung
cancer and non-small cell lung cancer), prostate cancer, lymphoma, leukemia,
endometrial cancer, melanoma, ovarian cancer, pancreatic cancer, squamous
carcinoma, carcinoma of the head and neck, and esophageal carcinoma. The
compounas of formula (I) can be used alone in the treatment of such susceptible
neoplasms or can be used to provide additive or synergistic effects with certain
existing chemotherapies, and/or be used to restore effectiveness of certain existing
chemotherapies and radiation.
The present invention also provides a method for treating a condition characterized by
inappropriate cellular proliferation. By "inapproriate cellular proliferation" is meant
cellular proliferation resulting from inappropriate cell growth, cellular proliferation
resulting from excessive cell division, cellular proliferation resulting from cell division
at an accelerated rate, cellular proliferation resulting from inappropriate cell survival,
and/or cellular proliferation in a normal cell occurring at a normal rate, which is
neverthless undesired. Conditions characterized by inappropriate cellular proliferation
include but are riot limited to neoplasms, blood vessel proliferative disorders, fibrotic
disorders, mesangial cell proliferative disorders and metabolic diseases. Blood vessel
proliferative disorders include arthritis and restenosis. Fibrotic disorders include
hepatic cirrhosis and atherosclerosis. Mesangial cell proliferative disorders include
glomerulonephritis, malignant nephrosclerosis, thrombotic microangiopathy
syndromes, organ transplant rejection and glomeruiopathies. Metabolic disorders
include psoriasis, chronic wound healing, inflammation and neurodegenerative
diseases. Osteoarthritis and other osteoclast proliferation dependent diseases of
excess bone resorbtion are examples of conditions characterized by inapproprate
cellular proliferation in which the cellular proliferation occurs in normal cells at a
normal rate, but is nevertheless undesired.
The present invention also provides a method for inhibiting proliferation of a cell,
which method comprises contacting the cell with an amount of a compound of
formula (I) sufficient to inhibit proliferation of the cell. In one particular embodiment,
the cell is a neoplastic cell. In one particular embodiment, the cell is an
inappropriately proliferative cell. The term "inappropriately proliferative cell" as used
herein refers to cells that grow inappropriately (abnormally), cells that divide
excessively or at an accelerated rate, ceils that inappropriately (abnormally) survive
and/or normal cells that proliferate at a normal rate but for which proliferation is
undesired. Neoplastic ceils (including cancer cells) are an example of inappropriately
proliferative cells but are not the only inappropriately proliferative cells.
PLK is essential for cellular mitosis and accordingly, the compounds of formula (I) are
effective for inhibiting mitosis. "Inhibiting mitosis" refers to inhibiting the entry into
the M phase of the cell cycle, inhibiting the normal progression of the M phase of the
cell cycle once M phase has been entered and inhibiting the normal exit from the M
phase of the cell cycle. Thus, the compounds of the present invention may inhibit
mitosis by inhibiting the cell"s entry into mitosis, by inhibiting the cell"s progression
through mitosis or by inhibiting the cell"s exit from mitosis. As one aspect, the present
invention provides a method for inhibiting mitosis in a. cell, which method comprises
administering to the cell an amount of a compound of formula (I) sufficient to inhibit
mitosis. In one particular embodiment, the cell is a neoplastic cell. In one particular
embodiment, the cell is an inappropriately proliferative cell.
The present invention also provides the use of a compound of formula (I) for the
preparation of a medicament for the treatment of condition mediated by PLK in an
animal, such as a mammal (e.g., a human). The present invention further provides the
use of a compound of formula (I) for the preparation of a medicament for the
treatment of a susceptible neoplasm in an animal. The present invention further
provides the use of a compound of formula (I) for the preparation of a medicament
for the treatment of a condition characterized by inappropriate cellular proliferation.
The present invention further provides the use of a compound of formula (I) for the
preparation of a medicament for inhibiting proliferation of a cell. The present
invention further provides the use of a compound of formula (I) for the preparation of
a medicament for inhibiting mitosis in a cell.
While it is possible that, for use in therapy, a therapeutically effective amount of a
compound of formula (I) may be administered as the raw chemical, it is typically
presented as the active ingredient of a pharmaceutical composition or formulation.
Accordingly, the invention further provides a pharmaceutical composition comprising
a compound of the formula (I). The pharmaceutical composition may further comprise
one or more pharmaceutically acceptable carriers, diluents, and/or excipients. The
carriers), diluent(s) and/or excipient(s) must be acceptable in the sense of being
compatible with the other ingredients of the formulation and not deleterious to the
recipient thereof. In accordance with another aspect of the invention there is also
provided a process for the preparation of a pharmaceutical formulation including
admixing a compound of the formula (I) with one or more pharmaceutically
acceptable carriers, diluents and/or excipients.
Pharmaceutical formulations may be presented in unit dose form containing a
predetermined amount of active ingredient per unit dose. Such a unit may contain a
therapeutically effective dose of the compound of formula (I) or a fraction of a
therapeutically effective dose such that multiple unit dosage forms might be
administered at a given time to achieve the desired therapeutically effective dose.
Preferred unit dosage formulations are those containing a daily dose or sub-dose, as
herein above recited, or an appropriate fraction thereof, of an active ingredient
Furthermore, such pharmaceutical formulations may be prepared by any of the
methods well known in the pharmacy art.
Pharmaceutical formulations may be adapted for administration by any appropriate
route, for example by the oral (including buccal or sublingual), rectal, nasal, topical
(including buccal, sublingual or transdermal), vaginal or parenteral (including
subcutaneous, intramuscular, intravenous or intradermal) route. Such formulations
may be prepared by any method known in the art of pharmacy, for example by
bringing into association the active ingredient with the carriers) or excipient(s).
Pharmaceutical formulations adapted for oral administration may be presented as
discrete units such as capsules or tablets; powders or granules; solutions or
suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water
liquid emulsions or water-in-oil liquid emulsions.
For instance, for oral administration in the form of a tablet or capsule, the active drug
component can be combined with an oral, non-toxic pharmaceutically acceptable
inert carrier such as ethanol, glycerol, water and the like. Powders are prepared by
comminuting the compound to a suitable fine size and mixing with a similarly
comminuted pharmaceutical carrier such as an"edible carbohydrate, as, for example,
starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be
present
Capsules are made by preparing a powder mixture as described above, and filling
formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc,
magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the
powder mixture before the filling operation. A disintegrating or solubilizing agent
such as agar-agar, calcium carbonate or sodium carbonate can also be added to
improve the availability of the medicament when the capsule is ingested.
Moreover, when desired or necessary, suitable binders, lubricants, disintegrating
agents and coloring agents can also be incorporated into the mixture. Suitable
binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn
sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate,
carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in
these dosage forms include sodium oleate, sodium stearate, magnesium stearate,
sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators
include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and
the like. Tablets are formulated, for example, by preparing a powder mixture,
granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
A powder mixture is prepared by mixing the compound, suitably comminuted, with a
diluent or base as described above, and optionally, with a binder such as
carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone. a solution
retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an
absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder
mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia
pactlage or solutions of cellulosic or polymeric materials and forcing through a
screen. As an alternative to granulating, the powder mixture can be run through the
tablet machine and the result is imperfectly formed slugs broken into granules. The
granules can be lubricated to prevent sticking to the tablet forming dies by means of
the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture
is then compressed into tablets. The compounds of the present invention can also be
combined with a free flowing inert carrier and compressed into tablets directly
without going through the granulating or slugging steps. A clear or opaque
protective coating consisting of a sealing coat of shellac, a coating of sugar or
polymeric material and a polish coating of wax can be provided. Dyestuffs can be
added to these coatings to distinguish different unit dosages.
oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so
that a given quantity contains a predetermined amount of active ingredient Syrups
can be prepared by dissolving the compound in a suitably flavored aqueous solution,
while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy
ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or
natural sweeteners or saccharin or other artificial sweeteners, and the like can also be
added.
Where appropriate, dosage unit formulations for oral administration can be
microencapsulated. The formulation can also be prepared to prolong or sustain the
release as for example by coating or embedding particulate material in polymers, wax
or the like.
The compounds of formula (I) can also be administered in the form of liposome
delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and
multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such
as cholesterol, stearylamine or phosphatidylcholines.
The compounds of formula (I) may also be delivered by the use of monoclonal
antibodies as individual carriers to which the compound molecules are coupled. The
compounds may also be coupled with soluble polymers as targetable drug carriers.
Such polymers can include peptides, polyvinylpyrrolidone, pyran copolymer,
polyhydroxypropylmethacrylamide -phenol, polyhydroxyethylaspartamidephenol, or
polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the
compounds may be coupled to a class of biodegradable polymers useful in achieving
controlled release of a drug, for example, polylactic acid, polepsilon caprolactone,
polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans,
polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
Pharmaceutical formulations adapted for transdermal administration may be
presented as discrete patches intended to remain in intimate contact with the
epidermis of the recipient for a prolonged period of time. For example, the active
ingredient may be delivered from the patch by iontophoresis as generally described in
Pharmaceutical Research, 3(6):318 (1986).
Pharmaceutical formulations adapted for topical administration may be formulated as
ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays,
aerosols or oils.
For treatments of the eye or other external tissues, for example mouth and skin, the
formulations are preferably applied as a topical ointment or cream. When formulated
in an ointment, the active ingredient may be employed with either a paraffinic or a
water-miscible ointment base. Alternatively, the active ingredient may be formulated
in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical formulations adapted for topical administrations to the eye include
eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier,
especially an aqueous solvent
rmaceutical formulations adapted for topical administration in the mouth include
lozenges, pastilles and mouth washes.
Pharmaceutical formulations adapted for rectal administration may be presented as
suppositories or as enemas.
Pharmaceutical formulations adapted for nasal administration wherein the carrier is a
solid include a coarse powder having a particle size for example in the range 20 to 500
microns which is administered in the manner in which snuff is taken, i.e. by rapid
inhalation through the nasal passage from a container of the powder held close up to
the nose. Suitable formulations wherein the carrier is a liquid, for administration as a
nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
Pharmaceutical formulations adapted for administration by inhalation include fine
particle dusts or mists, which may be generated by means of various types of metered,
dose pressurised aerosols, nebulizers or insufflators.
Pharmaceutical formulations adapted for vaginal administration may be presented as
pessaries, tampons, creams, gels, pastes, foams or spray formulations.
Pharmaceutical formulations adapted for parenteral administration include aqueous
and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers,
bacteriostats and solutes which render the formulation isotonic with the blood of the
intended recipient; and aqueous and non-aqueous sterile suspensions which may
include suspending agents and thickening agents. The formulations may be presented
in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may
be stored in a freeze-dried (lyophilized) condition requiring only the addition of the
sterile liquid carrier, for example water for injections, immediately prior to use.
Extemporaneous injection solutions and suspensions may be prepared from sterile
powders, granules and tablets.
it should be understood that in addition to the ingredients particularly mentioned
above, the formulations may include other agents conventional in the art having
regard to the type of formulation in question, for example those suitable for oral
administration may include flavouring agents.
In the above-described methods of treatment and uses, a compound of formula (I)
may be employed alone, in combination with one or more other compounds of
formula (I) or in combination with other therapeutic agents. In particular, in methods
of treating conditions mediated by PLK and methods of treating susceptible
neoplasms, combination with other chemotherapeutic, hormonal and/or antibody
agents is envisaged as well as combination with surgical therapy and radiotherapy.
The term "chemotherapeutic" as used herein refers to any chemical agent having a
therapeutic effect on the subject to which it is administered. "Chemotherapeutic"
agents include but are not limited to anti-neoplastic agents, analgesics and anti-
emetics. As used herein, "anti-neoplastic agents" include both cytostatic and
cytotoxic agents. Combination therapies according to the present invention thus
comprise the administration of at least one compound of formula (I) and the use of at
least one other cance treatment method. In one embodiment, combination therapies
according to the present invention comprise the administration of at least one
compound of formula (I) and at least one other chemotherapeutic agent. In one
particular embodiment, the present invention comprises the administration of at least
one compound of formula (I) and at least one anti-neoplastic agent As an additional
aspect, the present invention provides the methods of treatment and uses as described
above, which comprise administering a compound of formula (I) together with at least
one chemotherapeutic agent In one particular embodiment, the chemotherapeutic
agent is an anti-neoplastic agent. In another embodiment, the present invention
provides a pharmaceutical composition as described above further comprising at least
one other chemotherapeutic agent, more particularly, the chemotheraDeutic agent is
an anti-neoplastic agent
iccally, any chemotherapeutic agent that has activity versus a susceptible neoplasm
being treated may be utilized in combination with the compounds of formula (I),
provided that the particular agent is clinically compatible with therapy employing a
compound of formula (I). Typical anti-neoplastic agents useful in the present
invention include, but are not limited to, anft-microtubule agents such as diterpenoids
and vinca alkaloids; platinum coordination complexes; alkylating agents such as
nitrogen mustards, oxazaphosphor-ines, alkylsulfonates, nitrosoureas, and triazenes;
antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase 11
inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine
analogues and anti-folate compounds; topoisomerase I inhibitors such as
camptothecins; hormones and hormonal analogues; signal transduction pathway
inhibitors; non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic
agents; proapoptotic agents; and cell cycle signaling inhibitors.
Anti-microtubule or anti-mitotic agents are phase specific agents active against the
microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples
of anti-microtubule agents include, but are not limited to, diterpenoids and vinca
alkaloids. Examples of diterpenoids include, but are not limited to, paclitaxel and its
analog docetaxel. Examples of vinca alkaloids include, but are not limited to,
vinblastine, vincristine, and vinorelbine.
Platinum coordination complexes are non-phase specific anti-neoplastic agents, which
are interactive with DNA. The platinum complexes enter tumor cells, undergo,
aquation and form intra- and interstrand crosslinks with DNA causing adverse
biological effects to the tumor. Examples of platinum coordination complexes
include, but are not limited to, cisplatin and carboplatin.
Alkylating agents are non-phase anti-neoplastic specific agents and strong
electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to
DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino,
and hydroxyl groups. Such alkylation disrupts nucleic acid function leading to cell
path Examples of alkylating agents include, but are not limited to, nitrogen
mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates
such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
Antibiotic chemotherapeutic agents are non-phase specific agents, which bind or
intercalate with DNA. Typically, such action results in stable DNA complexes or strand
breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
Examples of antibiotic anti-neoplastic agents include, but are not limited to,
actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and
doxorubicin; and bleomycins.
Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the
mandrake plant Epipodophyllotoxins typically affect cells in the S and G2 phases of
the cell cycle by forming a ternary complex with topoisomerase II and DNA causing
DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of
epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at
S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting
purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently,
S phase does not proceed and cell death follows. Examples of antimetabolite anti-
neoplastic agents include, but are not limited to, fluorouracil, methotrexate,
cytarabine, mecaptopurine and thioguanine.
Camptothecins, including, camptothecin and camptothecin derivatives are available or
under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is
believed to be related to its Topoisomerase I inhibitory activity. Examples of
camptothecins include, but are not limited to irinotecan, topotecan, and the various
optical forms of 7-(4-methy!piperazino-methylene)-10,11 -ethylenedioxy-20-
camptothecin.
Narones and hormonal analogues are useful compounds for treating cancers in
which there is a relationship between the hormone(s) and growth and/or lack of
growth of the cancer. Examples of hormones and hormonal analogues believed to be
useful in the treatment of neoplasms include, but are not limited to, adrenocorti-
costeroids such as prednisone and prednisolone which are useful in the treatment of
malignant lymphoma and acute leukemia in children; aminoglutethimide and other
aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in
the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma
containing estrogen receptors; progestrins such as megestrol acetate useful in the
treatment of hormone dependent breast cancer and endometrial carcinoma;
estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide,
cyproterone acetate and 5 the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-
estrogens such as tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene
useful in the treatment of hormone dependent breast carcinoma; and gonadotropin-
releasing hormone (GnRH) and analogues thereof which stimulate the release of
leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment
prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin
acetate and luprolide.
Signal transduction pathway inhibitors are those inhibitors which block or inhibit a
chemical process which evokes an intracellular change. As used herein this change is
cell proliferation or differentiation. Signal tranduction inhibitors useful in the present
invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine
kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphotidyl inositol-3
kinases, myo-inositol signaling, and Ras oncogenes.
Several protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl
residues in various proteins involved in the regulation of cell growth. Such protein
tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
Recptor tyrosine kinases are transmembrane proteins having an extracellular ligand
binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor
tyrosine kinases are involved in the regulation of cell growth and are sometimes
termed growth factor receptors. Inappropriate or uncontrolled activation of many of
these kinases, i.e. aberrant kinase growth factor receptor activity, for example by over-
expression or mutation, has been shown to result in uncontrolled cell growth.
Accordingly, the aberrant activity of such kinases has been linked to malignant tissue
growth. Consequently, inhibitors of such kinases could provide cancer treatment
methods. Growth factor receptors include, for example, epidermal growth factor
receptor (EGFr, ErbB2 and ErbB4,), platelet derived growth factor receptor (PDGFr),
vascular endotbelial growth factor receptor (VEGFR), tyrosine kinase with
immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin
growth factor-l receptor (IGF-I), macrophage colony stimulating factor (cfms), BTK,
ckit, cmet, Fibrobtast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and
TrkC), ephrin (eph) receptors, and the RET protooncogene. Several inhibitors of growth
factor receptors are under development and include ligand antagonists, antibodies,
tyrosine kinase inhibitors, anti-sense oligonucleotides and aptamers. Growth factor
receptors and agents that inhibit growth factor receptor function are described, for
instance, in Kath, John C, Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al
DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth Factor Receptors as
Targets", New Molecular Targets for Cancer Chemotherapy, Ed. Workman, Paul and
Kerr, David, CRC Press 1994, London.
Tyrosine kinases, which are not growth factor receptor kinases are termed non
receptor tyrosine kinases. Non-receptor tyrosine kinases useful in the present
invention, which are targets or potential targets of anti-neoplastic drugs, include cSrc,
Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-
Abl. Such non-receptor kinases and agents which inhibit non-receptor tyrosine kinase
function are described in Sinh, S. and Corey, SJ., (1999) Journal of Hematotherapy and
Stem Cell Research 8 (5): 465 - 80; and Bolen, J.B., Brugge, J.S., (1997) Annual Review
of Immunology. 15:371-404.
SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a
variety of enzymes or adaptor, proteins including, P13-K p85 subunit, Src family
kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. SH2/SH3 domains as
targets for anti-cancer drugs are discussed in Smithgall, T.E (1995), Journal of
Pharmacological and Toxicological Methods. 34(3) 125-32.
Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which
include blockers of Raf kinases (Rafk), Mitogen or Extracellular Regulated Kinase
(MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family
member blockers including blockers of subtypes of PKCs (alpha, beta, gamma, epsilon,
rriu, lambda, iota, zeta), IkB kinase family (IKKa, IKKb), PKB family kinases, Akt kinase
family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and
inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal
of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000),
Biochemical Pharmacology, 60.1101-1107; Massague, J., Weis-Garcia, F. (1996)
Cancer Surveys. 27:41-64; Philip, PA, and Harris, A.L (1995), Cancer Treatment and
Research. 78:3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10),
2000,223-226; and Martinez-lacati, L, et al, Int J. Cancer (2000), 88(1), 44-52.
Inhibitors of Phosphotidyl lnositol-3 Kinase family members including blockers of
PI3-kinase, ATM, DNA-PK, and Ku are also useful in combination with the present
invention. Such kinases are discussed in Abraham, R.T (1996), Current Opinion in
Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-
3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29
(7):935-8; and Zhong, H. et al, Cancer Res, (2000) 60(6), 1541-1545.
Also useful in combination with the present invention are Myo-inositol signaling
inhibitors such as phospholipase C blockers and Myoinositol analogues. Such signal
inhibitors are described in Powis. G., and Kozikowski A., (1994) New Molecular Targets
for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC Press 1994, London.
other group of signal transduction pathway inhibitors useful in combination with
the present invention are inhibitors of Ras Oncogene. Such inhibitors include
inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as
well as anti-sense oiigonucleotides, ribozymes and immunotherapy. Such inhibitors
have been shown to block Ras activation in cells containing wild type mutant Ras,
thereby acting as antiproliferation agents. Ras oncogene inhibition is discussed in
Scharovsky, O.G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical
Science. 7(4) 292-8; Ashby. M.N. (1998). Current Opinion in Lipidology. 9(2)99-102;
and BioChim. Biophys. Acta, (1989) 1423(3):19-30.
As mentioned above, antibodies to receptor kinase ligand binding may also serve as
signal transduction inhibitors. This group of signal transduction pathway inhibitors
includes the use of humanized antibodies to the extracellular ligand binding domain
of receptor tyrosine kinases. For example, Imclone C225 EGFR specific antibody (see
Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat Rev.,
(2000), 26(4), 269-286); Herceptin® ErbB2 antibody (see Tyrosine Kinase Signaling in
BreasfrCancer:ErbB Family Receptor Tyrosine Kinases, Breast Cancer Res., 2000,2(3),
176-183); and 2CB VE6FR2 specific antibody (see Brekken, RA et al, Selective
Inhibition of VEGFR2 Activity by a Monoclonal Anti-VEGF Antibody Blocks Tumor
Growth in Mice, Cancer Res. (2000) 60.5117-5124).
Receptor kinase angiogenesis inhibitors may also find use in the present invention.
Inhibitors of angiogenesis related VEGFR and TIE2 are discussed above in regard to
signal transduction inhibitors (both receptors are receptor tyrosine kinases). Other
inhibitors may be used in combination with the compounds of the present invention.
For example, anti-VEGF antibodies, which do not recognize VEGFR (the receptor
tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alphav
betas) that will inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also
prove useful in combination with PLK inhibitors.
Agents used in immunotherapeutic regimens may also be useful in combination with
the compounds of formula (I).
Agents used in proapoptotic regimens (e.g., bcl-2 antisense oligonucleotides) may also
be used in the combination of the present invention. Members of the Bcl-2 family of
proteins block apoptosis. Upregulation of bcl-2 has therefore been linked to
chemoresistance. Studies have shown that the epidermal growth factor (EGF)
stimulates anti-apoptotic members of the bcl-2 family (i.e., md-1). Therefore,
strategies designed to downregulate the expression of bcl-2 in tumors have
demonstrated clinical benefit and are now in Phase ll/lll trials, namely Genta"s G3139
bcl-2 antisense oligonucleotide. Such proapoptotic strategies using the antisense
oligonucleotide strategy for bcl-2 are discussed in Water JS et al., J. Clin. Oncol.
18:1812-1823 (2000);and Kitada S et al., Antisense Res. Dev. 4:71-79 (1994).
Cell cycle signaling inhibitors inhibit molecules involved in the control of the cell
cycle. Cyclin dependent kinases (CDKs) and their interaction cyclins control
progression through the eukaryotic cell cycle. The coordinated activation and
inactivatibn of different cydin/CDK complexes is necessary for normal progression
through the cell cycle. Several inhibitors of cell cycle signaling are under
development For instance, examples of cyclin dependent kinases, including CDK2,
CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania, et
al., Exp. Opin. Ther. Patents 10(2):215-230 (2000).
in one embodiment, the methods of the present invention comprise administering to
the animal a compound of formula (I) in combination with a signal transduction
pathway inhibitor, particularly gefitinib (IRESSA®).
The methods and uses employing these combinations may comprise the administration
of the compound of formula (I) and the other chemotherapeutic/anti-neoplastic agent
either sequentially in any order or simultaneously in separate or combined
pharmaceutical compositions. When combined in the same formulation it will be
appreciated that the two compounds must be stable and compatible with each other
and the other components of the formulation and may be formulated for
administration. When formulated separately they may be provided in any convenient
formulation, in such a manner as are known for such compounds in the art.
When a compound of formula (I) is used in combination with a chemotherapeutic
agent, the dose of each compound may differ from that when the compound is used
alone. Appropriate doses will be readily appreciated by those skilled in the art The
appropriate dose of the compound(s) of formula (I) and the other therapeutically
active agent(s) and the relative timings of administration will be selected in order to
achieve the desired combined therapeutic effect, and are within the expertise and
discretion of the attendent clinician.
Compounds of formula (I) may be conveniently prepared by the methods outlined in
Scheme 1 below.
wherein:
R1 is selected from the group consisting of H, alkyl, alkenyl. alkynyl, -C(O)R7, -CO2R7,
-C(O)NR7R8. -C(O)N(R7)OR8 -C(O)N(R7)-R2-OR8. -C(O)N(R7)-Ph.
-C(O)N(R7)-R2-Ph, -C(O)N(R7)C(O)R8, -C(O)N(R7)CO2R8, -C(O)N(R7)C(O)NR7R8,
-C(O)N(R7)S(O)2R8, -R2-OR7, -R2-O-C(O)R7, -C(S)R7. -C(S)NR7R8. -C(S)N(R7)-Ph,
-C(S)N(R7)R2-Ph, -R2-SR7. -C(=NR7)NR7R8. -C(=NR7)N(R8)-Ph,
-C(=NR7)N(R8)-Ph,, -R2-NR7R8, -CN, -OR7, -S(O)fR7, -S(O)2NR7R8,
-S(O)2N(R7)-Ph. -S(O)2N(R7)-R2-Ph. -NR7R8, N(R7)rPh. -N(R7)-R2-Ph, -N(R7)-SO2R8
and Het;
Fa is phenyl optionally substituted from 1 to 3 times with a substituent selected from
the group consisting of halo, alkyl, -OH, -R2-OH, -O-alkyl, -R2-O-alkyl, -NH2,
-N(H)alkyl, -N(alkyl)2. -CN and -N3;
Het is a 5-7 membered heterocycle having 1,2,3 or 4 heteroatoms selected from N, O
and S, or a 5-6 membered heteroaryl having 1,2,3 or 4 heteroatoms selected
from N, 0 and S, each optionally substituted from 1 to 2 times with a
substituent selected from the group consisting of halo, alkyl, oxo, -OH, -R2-OH,
-O-alkyl, -R2-O-alkyl. -NH2, -N(H)alkyl, -N(alkyl)2 -CN and -N3;
Q1 is a group of formula: -(R2)a-(Y1)(R2)c-R3
a, b and c are the same or different and are each independently 0 or 1 and at least
one of a or b is 1;
n is 0,1,2,3 or 4;
Q2 is a group of formula: -(R2)aa(Y2)bb(R2)ccR4
or two adjacent Q2 groups are selected from the group consisting of alkyl,
alkenyl, -OR7, -S(O)fR7 and -NR7R8 and together with the carbon atoms to
which they are bound, they form a C5-6cycloalkyl, C5-6cycloalkenyl, phenyl, 5-7
membered heterocycle having 1 or 2 heteroatoms selected from N, 0 and S, or
5-6 membered heteroaryl having 1 or 2 heteroatoms selected from N, 0 arid S;
aa, bb and cc are the same or different and are each independently 0 or 1 ;
each Y1 and Y2 is the same or different and is independently selected from the group
consisting of -O-, -S(O)t-, -N(R7)-, -C(O)-, -0C(O}-. -CO2-, -C(O)N(R7)-,
-C(O)N(R7)S(O)2-, -OC(O)N(R7)-, -OS(O)2-, -S(O)2N(R7)-. -S(O)2N(R7)C(O)-,
-N(R7)S(O)2 -N(R7)C(O)-, -N(R7)CO2- and -N(R7)C(O)N(R7)-;
each R2 Is the same or different and is independently selected from the group
consisting of alkylene, alkenylene and alkynylene;
each R3 and R4 is the same or different and is each independently selected from the
group consisting of H, halo, alkyl, alkenyl, alkynyl, -C(O)R7, -C(0)NR7R8, -CO2R7,
-C(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)fR7,
-S(O)2NR7R8, -NR7R8. -N(R7)C(O)R8, -N(R7)S(O)2R8.-NO2, -CN, -N3and a group of
Ring A is selected from the group consisting of C5-10cycloalkyl,
C5-10cycloalkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from N, 0 and S and 5-10 membered heteroaryl
having 1,2 or 3 heteroatoms selected from N, 0 and S
eachd is 0 or 1;
e is 0,1,2,3 or 4;
each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, Ph,
Het, -CH(OH)-R2-OH, -C(O)R7, -C02R7, -CO2-R2-Ph, -CO2-R2-Het.
-C(O)NR7R8, -C(O)N(R7)C(O)R7, -C(O)N(R7)CO2R7, -C(O)N(R7)C(O)NR7R8.
-C(O)N(R7)S(O)2R7, -C(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)R8,
-CR7=N-OR8, =0, -OR7, -OC(O)R7, -OC(O)Ph, -OC(O)Het, -OC(O)NR7R8,
-O-R2-S(O)2R7, -S(0)fR7, -S(0)2NR7R8, -S(0)2Ph, -S(O)2Het, -NR7R8,
-N(R7)C(O)R8, -N(R7)CO2R8,.-N(R7)-R2-CO2R8, -N(R7)C(O)NR7R8,
-N(R7)-C(O)NR7R8. -N(R7)C(O)Ph, -N(R7)C(O)Het, -N(R7)Ph, -N(R7)Het.
-N(R7)C(O)NR7,-R2-NR7R8 -N(R7)C(O)NR7Ph. -N(R7)C(O)N(R7)Het,
-N(R7)C(O)N(R7)-R2-Het -N(R7)S(O)2R8, -N(R7)-S(O)2R8. -NO2 -CN and
-N3.
wherein when Q1 is defined where b is 1 and c is 0, R3 is not halo, -C(0)R7, -C(0)NR7R8,
-CO2R7. -C(S)R7, -C(S)NR7R8, -C(-NR7)R8, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7,
-S(O)fR7, -S(0)2NR7Ra, -NR7R8. -N(R7)C(O)R8, -N(R7)S(O)2R8, -NOz, -CN or -N3;
wherein when Q2 is defined where bb is 1 and cc is 0, R4 is not halo, -C(0)R7,
-C(0)NR7R8, -C&R7, -e(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8,
-CR7-N-0R7, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8, -N(R7)C(O)R8, -N(R7)S(O)2R8,
-N02, -CN or -N3;
Rs is selected from the group consisting of H, halo, alkyl. cycloalkyl, OR7, -S(O)fR7,
-NR7R8, -NHC(0)R7, -NHC(0)NR7R8 and -NHS(O)2R7;
0, 1 or 2; and
each R7 and each R8 are the same or different and are each independently selected
from the group consisting of H, aikyl, alkenyl, alkynyl, cycloalkyl and
cycloalkenyl; and
R10 is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkenyl and suitable carboxylic acid protecting groups.
Generally, the process for preparing the compounds of formula (I) (all formulas and all
variables having been defined above in connection with Scheme 1) comprises the
steps of:
a) reacting a compound of formula (HI) with a compound of formula (IV) to
prepare a compound of formula (I);
b) optionally converting the compound of formula (I) to a pharmaceutically •
acceptable salt, solvate or physiologically functional derivative thereof; and
c) optionally converting the compound of formula (I) or a pharmaceutically
acceptable salt, solvate or physiologically functional derivative thereof to a different
compound of formula (I) or a pharmaceutically acceptable salt, solvate or
physiologically functional derivative thereof.
More specifically, compounds of formula (I) can be prepared by reacting a compound
of formula (IV) with a compound of formula (III) to prepare a compound of formula (l-
A compound of formula (I-A) may be converted into a pharmaceutically acceptable
salt, solvate or physiologically functional derivative thereof or may be converted to a
different compound of formula (I) or a pharmaceutically acceptable salt, solvate or
physiologically functional derivative thereof using techniques described hereinbelow
and those conventional in the art.
The reaction of a compound of formula (III) with a compound of formula (IV) is
typically carried-out in an inert solvent at room temperature. Typically two molar
equivalents of a compound of formula (III) are combined with one molar equivalent of
a compound of formula (IV). Examples of suitable inert solvents for this reaction
include but are not limited to, chloroform, dichloromethane, tetrahydrofuran, dioxane,
and toluene.
wherein all variables are as defined in connection with Scheme 1.
Compounds of formula (V) are commercially available or can be prepared using
conventional knowledge in the art Typically, reaction of a compound of formula (V)
with sulfuryl chloride at room temperature provides a compound of formula (IV).
Excess sulfuryl chloride may be used if desired. Examples of suitable solvents include
but are not limited to chloroform, dichloromethane, and toluene. See, Corral, C;
Lissavetzky, J. Synthesis 847-850 (1984).
A compound of formula (III) can be prepared by several methods. According to one
method, a compound of formula (III) is prepared according to Scheme 2 below.
Denerally, this process for preparing a compound of formula (Ill) (all formulas and all
variables having been defined above in connection with Scheme 1) comprises the
steps of:
a) reducing the compound of formula (VII) to prepare a compound of formula
(VIII); and
b) reacting the compound of formula (VIII) with a ring forming reagent to
prepare a compound of formula (III).
The order of the foregoing steps is not critical to the practice of the invention and the
process may be practiced by performing the steps in any suitable order based on the
knowledge of those skilled in the art
More specifically, a compound of formula (III) can be.prepared by reacting a
compound of formula (VIII) with a ring forming reagent There are several ring
forming reagents which may be employed in this process step. In one embodiment,
the compound formula (III-A.) (i.e., a compound of formula (III) wherein Ft5 is H or
alkyl) is prepared by reacting a compound of formula (VIII) with a ring forming
This reaction may be carried out using conventional techniques. See, White, A., et al.,
J. Med. Chem. 43:4084-4097 (2000); Jiang, J.-L, et al., Synthetic Comm; 28:4137-
4142 (1998); Tanaka, A., et al., Chem. Pharm. Bull. 42:560-569 (1994); Tian, W., et al.,
Synthesis 12:1283-1286 (1992); Buckle, D. R., et al., J. Med. Chem. 30:2216-2221
(1987); and Raban, M., et al., J. Org. Chem. 50:2205-2210 (1985). This reaction may be
carried out neat or in a suitable solvent The reaction may optionally be heated to a
temperature of from about 50 to about 230 °C The reaction is typically carried out
with an excess of the compound of formula (IX). An additional acid may be used.
Exemples of suitable acids include but are not limited to, hydrochloric acid,
hydrobromic acid, perchloric acid, sulfuric acid, p-toluenesulfonic acid,
methanesulfonic acid, and trifluoromethanesulfonic acid. Examples of suitable
solvents for this reaction include but are not limited to water, methanol, ethanol,
isopropanol, tetrahydrofuran, dichloromethane,toluene, N,N-dimethylformamide,
dimethylsulfoxide, and acetonitrile. The compounds of formula (IX) are commercially
available.
A compound of formula (VIII) may be prepared by reducing a compound of formula
The reduction can be carried out using conventional techniques and reducing agents.
See, Rangarajan, M., et al., Bioorg. Med. Chem. 8:2591-2600 (2000); White, A.W., et al.,
J. Med. Chcm. 43:4084-4097 (2000); Silvestri, R., et al., Bioorg. Med. Chem. 8:2305-
2309 (2000); Nagaraja, D., et al., Tetrahedron Lett 40:7855-7856 (1999); Jung, F., et
al., 1 Med. Chem. 34:1110-1116 (1991); Srivastava, R.P., et al., Pharmazie 45:34-37
(1990); Hankovszky, H.O., et al., Can. J. Chem. 67:1392-1400 (1989); Ladd, D.L, et al.. J.
Org. Chem. 53:417-420 (1988); Mertens, A., et al., J. Med. Chem. 30:1279-1287
(1987); and Sharma, K.S., et al., Synthes/s 4:316-318 (1981). Examples of suitable
reducing agents for this reaction include but are not limited to, palladium with
hydrogen, palladium with ammonium formate, platinum oxide with hydrogen, nickel
with hydrogen, tin(ll) chloride, iron with acetic acid, aluminum with ammonium
chloride, borane, sodium dithionite, and hydrazine. The reaction may optionally be
heated to between about 50 and about 120 °C. Suitable solvents for this reaction vary
and include but are not limited to, water, methanol, ethanol, ethyl acetate,
tetrahydrofuran, and dioxane.
A compound of formula (VII) may be prepared by several methods. In one
embodiment, the compound of formula (VII) is prepared by reacting a compound of
formula (VI) with ammonia.
wherein all variables are as defined in connection with Scheme 1.
This reaction may be carried out using conventional techniques. See, Silvestri, R., et
al., Bioorg. Med. Chem. 8:2305-2309 (2000); Hankovszky, H.O., et al., Can. J. Chem.
67:1392-1400 (1989); Nasielski-Hinkens, R.;etal., Weterocye/es 26:2433-2442 (1987);
Chu, K.Y., et al., J. Chem. Soc, Pcrkin Trans. 1 10:1194-1198 (1978). This reaction is
typically carried out with an excess of ammonia and may be optionally heated to a
temperature of from about 50 to about 100 °C, Examples of suitable solvents for this
reaction include but are not limited to, water, methanol, ethanol, isopropanol,
tetrahydrofuran, dioxane, and 1,2-dimethoxyethane.
The compounds of formula (VI) are commercially available or may be prepared using
conventional techniques and reagents.
In another embodiment, the compound of formula (VII) can be prepared by reacting a
protected compound of formula (X) under nitration conditions to prepare a protected
compound of formula (VII) (i.e.,VH-A) and then removing the protecting group from
the compound of formula (Vll-A).
protection of anilines is a common transformation well known to one skilled in
the art See, Kocienski, PJ. Protecting Groups, Georg Thieme Verlag, Stuttgart, 1994;
and Greene, T.W., Wuts, P. G. M. Protecting Groups in Organic Synthesis (2nd Edition),
J. Wiley and Sons, 1991. Suitable protecting groups for this application include but
are not limited to acetyl; trifluoroacetyl, benzyloxycarbonyl, allyloxycarbonyl, 2-
(rimethylsilyl)ethoxycarbonyl, phenylsulfonyl, and p-toluenesulfonyl. Reagents and
conditions vary according to the nature of the particular protecting group. Some
typical reagents include but are not limited to acetic anhydride, trifluoroacetic
anhydride, benzyl chloroformate, allyl chloroformate, 4-nitrophenyI 2-
(trimethylsilyl)ethyl carbonate, phenylsulfonyl chloride, and p-tolueniulfonyl chloride.
In certain cases the addition of some base is required. Examples of suitable bases
include but are not limited to potassium carbonate, sodium carbonate, trialkylamines,
pyridine, and potassium t-butoxide. Suitable solvents for these conversions include
but are not limited to dichloromethane, chloroform, tetrahydrofuran, acetic acid,
methanol, ethanol, water, toluene, and diethyl ether.
The nitration of anilines is also well documented in the literature and the foregoing
reaction may be carried out using these conventional techniques. See, Wissner, A., et.
al., J. Med.Chem. 46:49-63 (2003); Duggan, S. A., et a!., J. Org. Chem. 66:4419-4426
(2001); Clews, J et al.. Tetrahedron 56:8735-8746 (2000); and Kagechika, H., J. Med.
Chem. 31:2182-2192 (1988). The nitration may be carried out with a variety of
nitrating reagents including but not limited to 70% aqueous nitric acid, red fuming
nitric acid, ammonium nitrate with trifluoroacetic anhydride, and potassium nitrate
with trifluoromethanesulfonic acid. The reaction is typically conducted at room
temperature, but may be optionally heated to a temperature of from about 40 to
about 100 °C in certain cases. Suitable solvents include but are not limited to acetic
acid, sulfuric add, acetic anhydride, dichloromethane, and chloroform.
The nitration results in a compound of formula (Vll-A), (i.e., a protected compound of
formula (VII)). The cleavage of the aniline protecting group, to result in a compound of
formula (VII) can be accomplished through many different conventional methods.
See, Kocienski, PJ. Protecting Groups, Georg Thieme Verlag, Stuttgart, 1994; and
Greene, T.W., Wuts, P. G. M. Protecting Groups in Organic Synthesis (2nd Edition), J.
Wiley and Sons, 1991.
The compounds of formula (X) may be prepared by installing a protecting group on
the corresponding aniline. Such Anilines are commercially available or may be
prepared using conventional techniques.
A compound of formula (Ill-A) may optionally be converted to a compound of formula
(Ill-B). This conversion may be effected by halogenating the compound of formula
(Ill-A) to prepare a compound of formula (Ill-B).
. wherein X1 is halo (particularly Cl, Br or I) and all other variables are as defined
in connection with Scheme 1.
This type of transformation is well established in the literature. See, Taylor, E. C, et
al., J. Org. Chem. 56:6937-6939 (1991); Mistry, A. G., et al., Tetrahedron Lett 27:1051-
1054 (1986); and Apen, P. G., et al., Heterocycles 29:1325-1329 (1989). Suitable
halogenating agents include but are not limited to, A/-chlorosuccinimide, N-
bromosuccinimide, AModosuccinimide, chlorine, bromine, and iodine. Examples of
suitable solvents include but are not limited to, dichloromethane, chloroform, diethyl
ether, tetrahydrofuran, and acetone.
A compound of formula (Ill-B) may also be prepared directly from a compound of
formula (VIII). The process comprises the steps of i) reacting a compound of formula
(VIII) with a phosgene or phosgene equivalent compound to prepare a compound of
formula (Xil) and ii) reacting the compound of formula (XII) with phosphorous oxy
halide to prepare a compound of formula (Ill-B).
each R12 is the same or different and is independently selected from the group
consisting of Cl, methoxy, ethoxy, trichloromethoxy, amlno and N-
imidazolyl;
X1 is halo (particularly Cl, Br or I; more particularly Cl or Br); and
all other variables are as defined in connection with Scheme 1.
The phosgene or phosgene equivalent compound is the ring forming reagent and is
typically a compound of formula (XI) as shown above. Phosgene and phosgene
equivalent compounds of formula. (XI) are commercially available. Examples of
suitable compounds of formula (XI) include but are not limited to phosgene, dimethyl
carbonate, diethyl carbonate, 1,1"-carbonyldiimidazole, urea, and triphosgene. The
reaction of a compound of formula (VIII) with the phosgene or phosgene equivalent
compound can be carried out using conventional techniques. See, Silvestri, R., et al.,
Bioorg. Meet Chem. 8:2305-2309 (2000); Wright, J. L, et al., J. Med. Chan. 43:3408-
3419 (2000); Penieres, G. C, et al., Synthetic Comm. 30:2191-2195 (2000); and Von
der Saal, W., et al.. J. Med. Chem. 32:1481-1491 (1989). The reaction is typically run in
an inert solvent or neat The reaction may be optionally heated to a temperature of
from about 50 to about 250 °C. The optional addition of a suitable base to the
reaction may be desirable. Examples of such bases include but are not limited to,
trialkylamines, pyridine, 2,6-lutidine, potassium carbonate, sodium carbonate, and
sodium bicarbonate. Examples of suitable solvents for this reaction include but are
not limited to dichloromethane, chloroform, N,N-dimethylformamide,
tetrahydrofuran, toluene, and acetone.
The reaction of the compound of formula (XII) with the phosphorous oxy halide to
prepare a compound of formula (lll-B) can be carried out using conventional
techniques. See, Blythin, D. 1, et al., J. Med. Chem. 29:1099-1113 (1986); and Crank,
G., Aust J. Chem. 35:775-784 (1982). Examples of suitable reagents include but are
not limited to phosphorous oxychloride and phosphorous oxybromide. Suitable
solvents include but are not limited to, dichloromethane, chloroform, dichloroethane,
and toluene. Optional heat ranging from about 50 to about 150 °C may be used.
A compound of formula (Ill-B), prepared by any method, may optionally be converted
to a compound of formula (Ill-G) by reacting with an amine of formula HNR7R8.
The reaction of a halo-substituted benzimidazole of formula (Ill-B) with an amine to
prepare a compound of formula (Ill-C) can be carried out using conventional
techniques. See, Alcalde, E., et al., J. Org. Chem. 56:4233-4238 (1991); Katsushima, T.,
et al., J. Med. Chem. 33:1906-1910 (1990); Young, R. C, et al., J. Med. Chem. 33:2073-
2080 (1990); lemura, R., et al., J. Med. Chem. 29:1178-1183 (1986); and Benassi, R., et
al., J. Chem. Soc., Perkin Trans. 210:1513-1521 (1985). An acid catalyst may be
employed if desired. Examples of suitable acid catalysts include but are not limited to,
hydrochloric acid and p-toluenesulfonic acid. The reaction can optionally be heated
to a temperature of from about 50 to about 220 °C. Suitable solvents for this reaction
include but are not limited to, water, ethanol, isopropanol, i-methyl-2-pyrrolidinone,
N,N-dimethylformamide, dimethylsulfoxide, toluene, xylenes and tetrahydrofuran.
In another embodiment, a compound of formula (Ill-D) (i.e., a compound of formula
((III) wherein R5 is H or alkyl) is prepared according to the process outlined in Scheme
3 below.
wherein R13 is H or alkyl and all other variables are as defined in connection
with Scheme 1.
Generally, this process for preparing the a compound of formula (III—D) (all formulas
and all variables having been defined above in connection with Scheme 1) comprises
the steps of:
a) reacting a compound of formula (XIII) with a suitable acylating agent to
prepare a compound of the formula (XIV);
b) reacting a compound of formula (XIV) under nitration conditions to prepare a
compound of the formula (XV);
c) reducing a compound of formula (XV) to prepare a compound of formula (XVI);
and
d) cyclizing a compound of formula (XVI) to prepare a compound of formula (III-
D).
The order of the foregoing steps is not critical to the practice of the invention and the
process may be practiced by performing the steps in any suitable order based on the
knowledge of those skilled in the art.
WE are specifically, a compound of formula (Ill-D) can be prepared bycyclizing a
compound of formula (XVI).
wherein all variables are as defined in .connection with Schemes 1-3.
This type of cyclization reaction is well documented in the literature. See, Brafia, M. F.,
et al., J. Med. Chem. 45:5813-5816 (2002); Fonseca, T., et. al., Tetrahedron 57:1793-
1799 (200,1); White, A. W., et al., JL Med. Chem. 43:4084-4097 (2000); and Tamura, S.
Y., et al., Biorg.Med. Chem. Lett 7:1359-1364 (1997). This reaction may be carrfed
out neat or in a suitable solvent The reaction may optionally be heated to a
temperature of from about 50 to about 200 °C. Typically an excess of a suitable acid
is used. Examples of suitable acids include but are not limited to acetic acid,
trifluoroacetic acid, hydrochloric acid, hydrobromic acid, sulfuric acid,
methanesulfonic acid, p-toluenesulfonic acid, and pyridinium p-toluenesulfonate. A
dehydrating reagent may optionally be used as well. Examples of suitable dehydrating
reagents include but are not limited to magnesium sulfate, sodium sulfate,
phosphorous pentoxide, and molecular sieves. Examples of suitable solvents include
but are not limited to dichloromethane, chloroform, toluene, xylenes, methanol,
ethanol, and water.
A compound of formula (XVI) may be prepared by reducing a compound of formula
The deduction can be carried out using conventional techniques and reducing agents.
See, Rangarajan, M., et al., Bioorg. Med. Chem. 8:2591-2600 (2000); White. A.W., et al.,
J. Med. Chem. 43:4084-4097 (2000); Silvestri, R., et al., Bioorg. Med. Chem. 8:2305-
2309 (2000); Nagaraja, D., et al., Tetrahedron Lett 40:7855-7856 (1999); Jung, F., et
al., J. Med. Chem. 34:1110-1116 (1991); Srivastava, R.P., et al., Pharmazie 45:34-37
(1990); Hankovszky, H.O., et al., Can. J. Chem. 67:1392-1400 (1989); Ladd, D.L, et al., J.
Org. Chem. 53:417-420 (1988); Mertens, A., et al.. J. Med. Chem. 30:1279-1287
(1987); and Sharma. K.S.. et al.. Synthesis 4:316-318 (1981). Examples of suitable
reducing agents for this reaction include but are not limited to, palladium with
hydrogen, palladium with ammonium formate, platinum oxide with hydrogen, nickel
with hydrogen, tin(ll) chloride, iron with acetic acid, aluminum with ammonium
chloride, borane, sodium dithionite, and hydrazine. The reaction may optionally be
heated to between about 50 and about 120 CC. Suitable solvents for this reaction vary
and include but are not limited to, water, methanol, ethanol, ethyl acetate,
tetrahydrofuran, and dioxane.
A compound of formula (XV) may be prepared by reacting a compound of formula
(XIV) under nitration conditions.
wherein all variables are as defined in connection with Schemes 1-3.
The reaction of the compound of formula (XIV) under nitration conditions may be
carried out in the same manner as described above for the nitration of a compound of
formula (X).
A compound of formula (XIV) may be prepared by acylating a compound of formula
wherein all variables are as defined in connection with Schemes 1-3.
Acylation of anilines is a common transformation well known to one skilled in the art
and such conventional acylation techniques may be employed for carrying out the
foregoing reaction. See, Larock, R. C. Comprehensive Organic Transformations, VGH
Publishers, Inc., New York, pp. 972-976, 979, 981 (1989). The acylation reaction is
typically carried out using an acylating agent such as an acid halide, acid anhydride, or
carboxylic acich in the presence of a coupling reagent(s). Examples of suitable
coupling reagents include but are not limited to N,N-dicyclohexylcarbodiimide, 1-(3-
dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, 0-(7-azabenzotriazol-1 -yl)-
N,N,N,N-tetramethyluronium hexafluorophosphate, and- N,N"-carbonyldiimidazole.
Suitable solvents include but are not limited to N,N-dimethylformamide,
tetrahydrofuran, dioxane, toluene, benzene, 1,2-dimethoxyethane, and 1-methyl-2-
pyrrolidinone. Anilines of formula (XIII) are commercially available or readily prepared
from commercially available material using conventional techniques.
As will be apparent to those skilled in the art, a compound of formula (I) may be
converted to another compound of formula (I) using techniques well known in the art
For example, a compound of formula (I-A) may optionally be converted to a
compound of formula (I-B) or (I-C) according to the process outlined in Scheme 4.
wherein
Q3 is a group of formula: -(R2)a-(y3)i-(R2)c-R3
j is 0 or 1 ;
Y3 is selected from the group consisting of -S(O)f, -N(R7)-, -C(O)-, -OC(O)-, -
CO2-.
-C(O)N(R7)-, -C(O)N(R7)S(O)2-1 -OC(O)N(R7)-, -OS(O)2-, -S(O)2N(R7)-,
-S(O)2N(R7)C(O)-, -N(R7)(O)2-. -N(R7)C(O)-, -N(R7)CO2- and -
N(R7)C(O)N(R7)-;
LG is a suitable leaving group; and
all other variables are as defined in connection with Scheme 1 above.
In general the process for preparing a compound of formula (I-B) comprises the steps
of:
a) reacting the compound of formula (I-A) with a base and a compound of
formula (XVIII) to prepare a compound of the formula (I-B); or
b) reacting the compound of formula (I-A) with a compound of formula (IXX)
under Mitsunobu conditions to prepare a compound of formula (I-B).
More specifically, a compound of formula (I-B) can be prepared by reacting a
compound of formula (I-A) with a compound of formula (XVIII). The compounds of
formula (XVIII) are commercially available or can be prepared using conventional
knowledge in the art. The reaction may be carried out in an inert solvent, conveniently
at room temperature, in the presence of a suitable base. The compound of formula
(I-A) and the compound of formula (XVIII) may be present in equimolar amounts;
however, a slight excess of the compound of formula (XVIII) may be employed if
desired. Examples of suitable bases for this reaction include but are not limited to,
potassium carbonate, sodium carbonate, cesium carbonate, sodium hydride, and
potassium hydride. Examples of suitable inert solvents for this reaction include but
are not limited to, N,N-dimethylformamide, tetrahydrofuran, dioxane, and 1,2-
dimethoxyethane.
In another embodiment, a compound of formula (I-B) can be prepared by reacting a
compound of formula (I-A) with a compound of formula (IXX). The compounds of
formula (IXX) are commercially available or can be prepared using conventional
knowledge in the art The reaction is carried out in an inert solvent under standard
Mitsunobu conditions. See, Hughes, D.L, Org. React. 42:335-656 (1992); and
Mitsunobu, 0., Synthesis 1-28 (1981). Typically the compound of formula (I-A), the
compound of formula (IXX), a triarylphosphine, and a dialkyl azodicarboxylate are
reacted together at room temperature. Examples of suitable triarylphosphines include
but are not limited to, triphenylphosphine, tri-p-tolylphosphine, and
trimesitylphosphine. Examples of suitable dialkyl azodicarboxylates include but are
not limited to, diethyl azodicarboxylate, diisopropyl azodicarboxylate, and di-tert-
butyl azodicarboxylate. Examples of suitable inert solvents for this reaction include
but are not limited to, tetrahydrofuran, dioxane, 1,2-dimethoxyethane,
dichloromethane, and toluene.
A compound of formula (I-A) may also be converted to a compound of formula (I-C)
according to the following Scheme 5.
wherein M is -B(OH)2. -B(OR14)2, -Sn(R14)2, Zn-halo, Zn-R14, Mg-halo, Cu-halo.
Cu-Ru where R14 is alkyl or cycloalkyl, and all other variables are as defined in
connection with Schemes 1-4 above.
Generally, the process for preparing a compound of formula (I-C) comprises the steps
of:
a) reacting a compound of formula (I-A) with a suitable triflating reagent to
prepare a compound of formula (XX); and
b) coupling the compound of formula (XX).with a compound selected from the
group consisting of a compound of formula (XXI), (XXII), and (XXIII) using a palladium
(0) catalyst to prepare a compound of the formula (I-C).
More specifically, a compound of formula (I-C) can be prepared by reacting a
compound of formula (XX) with a compound selected from the group consisting of a
compound of formula (XXI), (XXII), and (XXIII) using a palladium (0) catalyst. This
reaction may be carried out in an inert solvent, in the presence of palladium (0). The
reaction may optionally be heated to a temperature of from about SO to about 150 °C.
Typically, the reaction is carried out by reacting an equimolar amount of a compound
of formula (XX) with an equtmolar amount of the compound selected from the group
consisting of compounds of formula (XXI), (XXII) and (XXIII). The palladium (0) catalyst
is typically present in 1-10 mole percent compared to the compound of formula (XX).
Examples of suitable palladium catalysts include but are not limited to,
tetrakis(triphenylphosphine)palladium (0) and tris(dibenzylideneacetone)dipalladium
(0). It is also possible to generate the palladium (0) catalyst in situ using palladium (II)
sources. Examples of suitable palladium (II) sources include but are not limited to,
palladium (II) acetate, palladium (II) chloride, palladium (II) trifluoroacetate,
dichiorobis(triphenyl-phosphine)palladium (II), and bis(diphenylphosphinoferrocene)-
palladium (II) dichloride. Suitable solvents for this reaction include but are not limited
to N,N-dimethylformamide, tetrahydrofuran, dioxane, toluene, benzene, 1,2-
dimethoxyethane, and 1-methyl-2-pyrroltdinone. Bases and phosphines may be
included as additives in the reaction if desired. Examples of suitable bases include but
are not limited to cesium carbonate, sodium carbonate, and trialkylamines. Examples
of suitable phosphine additives include but are not limited to triphenylphosphine,
tributylphosphine, diphenylphosphinoethane, and 2,2"-bis(diphenylphosphino)-1,11-
binaphthyl. Compounds of the formula (XXI), (XXII) and (XX111) may be obtained from
commercial sources or prepared either as discreet compounds or generated in situ
using conventional knowledge in the art See, Luker.TJ., et al., Tetrahedron Lett
41 -.7731-773S (2000); Yin, J., et al., Org. Lett 2:1101 -1104 (2000); Wolfe, J.P., et al..
Corn. J. Chem. 78:957-962 (2000); Littke, A.F., et al., J. Am. Chem. Soc 122:4020-4028
(2000); Hundertmark. T., et al.. Org. Lett 2:1729-1731 {2000); Buchwald, S,L, Ace.
Chem. Res. 31:805-818 (1998); Suzuki. A., J. Organomet Chem. 576:147-168 (1999);
Negishi, E., J. Organomet Chem. 576:179-194 (1999); Stanforth, S.P., Tetrahedron
54:263-303 (1998); Littke, A.F., Angew. Chem., Int. Ed. 37:3387-3388 (1999); and
Thorand, S., et al., J. Org. Chem. 63:8551-8553 (1998).
A compound of formula (XX) can be prepared from a compound of formula (I-A) using
a suitable inflating reagent. This reaction is typically carried out in an inert solvent
using a base and a reagent designed for conversion of alcohols into triflates (i.e., a
triflating reagent). Examples of suitable bases include but are not limited to sodium
carbonate, trialkylamines, pyridine, sodium hydride, and lithium bis(trimethylsilyl)
amide. The reaction is preferably run at a temperature of from about 0 to about 25
°C. Suitable triflating reagents for this reaction include but are not limited to,
trifluoromethanesulfonic anhydride, trifluoromethanesulfonyl chloride, and N-
phenyltrifluoromethanesulfonimide. Suitable Inert solvents for this reaction include
but are not limited to tetrahydrofuran, dichioromethane, toluene, chloroform, diethyl
ether, and dioxane.
As a further example of methods for converting a compound of formula (I) to another
compound of formula (I), a compound of formula (I-A), (I-B), or (1-C) (collectively
referred to as a compound of formula "(I-D)" may be converted to a different
R1 is other than-CO2R10;
and all other variables are as defined in connection with Schemes 1-5.
Several methods, using conventional techniques can be employed to convert a
compound of formula (I-D) to a different compound of formula (I), depending upon
the particular compound of formula (I) that is desired. For example, according to one
method, a compound of formula (I-D) can be converted to a compound of formula (I-
E) by removal of the carboxylic acid protecting group.
There are several options for carrying out this conversion. Examples of suitable
conditions include but are not limited to, basic hydrolysis where R" is -CO2MC
deprotection with protic acid where R1 is -CO2t-Bu, deprotection under palladium (0)
catalysis where R1 is CO2CH2CH=CH2, deprotection with tetrabutylammonium fluoride
where R1 is CO2CH2CH2Si(CH3)3 and hydrogenolysis where R1 is CO2CH2Ph. Other
suitable conditions for compounds with various R10 definitions will be apparent to
those skilled in the art The choice of protecting group and deprotection conditions
will be apparent to one skilled in the art and, detailed information on this subject is
available in the literature. See, Kocienski, PJ. Protecting Groups, Georg Thieme Verlag,
Stuttgart, 1994; and Greene, T.W., Wuts, P. G. M. Protecting Groups in Organic
Synthesis (2nd Edition), J. Wiley and Sons, 1991.
A compound of formula (I-E) may be further converted to a compound of formula (I-
F) by heating.
This reaction may be performed in an inert solvent Typically, the reaction is heated
to a temperature of from about 80 to about 120 °C. Examples of suitable solvents for
this reaction include but are not limited to acetic acid, propionic acid, N,N-
dimethylformamide, dimethylsulfoxide, ethanol, dioxane and toluene.
A compound of formula (I-E) may be further converted to a compound of formula (I-
G) using conventional amide bond coupling reactions with an amine of formula
HNR7R8.
This reaction can be carried out in an inert solvent using a variety of commercially
available coupling reagents. Suitable coupling reagents include but are not limited to
N,N-dicyclohexylcarbodiimide, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
hydrochloride, 1,1-carbonyldiimidazole, and benzotriazol-1-yloxytris(dimethyl-
amino)phosphonium hexafluorophosphate. Other suitable coupling reagents will be
readily apparent to those skilled in the art The carboxylic acid"optionally may be.
converted into the corresponding acid chloride and subsequently treated with the
amine of formula HNR7R8. Suitable reagents for the reaction of such acid chlorides
include but are not limited to oxalyl chloride, thionyl chloride, and 1-chloro-N,N,2-
trimethyl-1-propenylamine. Base may be optionally added to the coupling reaction.
The reaction may optionally require heating to a temperature of from about 40 to
about 100 °C. Suitable bases include but are not limited to trialkyiammes, pyridine,
and 4-(dimethylamino)pyridine. Examples of suitable solvents for this reaction include
but are not limited to dichloromethane, chloroform, benzene, toluene, N,N-
dimethylformamide and dichloroethane.
In an alternative embodiment, a compound of formula (1-G") is prepared directly from
a compound of formula (I-D).
This reaction is typically performed in a sealed vessel with an excess of ammonia. The
reaction is typically heated to a temperature of from about 50 to about 120 °C.
Suitable solvents for this reaction include but are not limited to methanol, ethanol,
isopropanol, tetrahydrofuran, and dioxane.
Dehydration of the compound of formula (I-G") may be used to prepare a compound
of formula (I-H).
wherein all variables are as defined in connection with Schemes 1-5.
The dehydration reaction can be carried out using a variety of reagents. Suitable
dehydration reagents include but are not limited to thionyl chloride, trifluoroacetic
anhydride, phosphorous oxychloride, phosphorous pentoxide, and N,N-
dicyclohexylcarbodiimide. The reaction may be optionally heated to from about 50 to
about 150 °C. Suitable solvents for this reaction include but are not limited to
dichloromethane, chloroform, benzene, toluene, N,N-dimethylformamide, and
dichloroethane.
A compound of formula (I-J) may be prepared through a two step conversion process,
comprising a) converting a compound of formula (I-E) to a compound of formula (l-l)
by coupling with N,O-dimethylhydroxylamine, and b) reacting the compound of
formula (l-l) with a nucleophile of formula M1-R7.
wherein
M1 is Li, Mg-halo, Cu-halo or Ce-halo; and
all variables are as defined in connection with Schemes 1-5.
The coupling reaction with N,O-dimethylhydroxylamine may be carried out in the
same manner as described above for the conversion of a compound of formula (I-E) to
a compound of formula (I-G). The addition of the nucleophile to the Weinreb amide
(l-l) is typically carried out at a temperature ranging from about -30 to about 5 °C.
Suitable solvents for this reaction include but are not limited to, tetrahydrofuran,
dioxane, diethyl ether, toluene, 1,2-dimethoxyethane, and hexanes. See, Weinreb,
S.M., et al.. Tetrahedron Lett 22:3815-3818 (1981). Nucleophiles of formula M1-R7 are
commercially available or can be prepared using con ventional knowledge in the art
A compound of formula (I-K) may be prepared from a compound of formula (I-D)
through a hydride reduction.
This reaction may be carried out in an inert solvent at a temperature ranging from
about -78 to about 25 °C. Suitable reducing agents include but are not limited to
diisobutylaluminum hydride, lithium aluminum hydride, and lithium borohydride.
Suitable solvents vary considerably depending on the chosen reducing agent.
Appropriate selection of a solvent for this reaction will be apparent to those skilled in
the art based upon the choice of reducing agent Examples of suitable solvents
include but are not limited to tetrahydrofuran, diethyl ether, 1,2-dimethoxyethane,
dioxane, dichloromethane, toluene, and hexanes.
A compound pf formula (I-K) may be oxidized to prepare a compound of formula (I-L).
This reaction-can be carried out using a wide variety of conventional oxidizing agents.
Suitable oxidizing agents include but are not limited to, manganese dioxide, dimethyl
sulfoxide / oxalyl chloride / triethylamine, pyridinium chlorochromate, pyridinium
dichromate, and tetrapropylammonium perruthenate / 4-methylmorpholine /V-oxide.
Examples of suitable solvents for the oxidation reaction include but are not limited to,
dichloromethane, chloroform, diethyl ether, toluene, and tetrahydrofuran.
A compound of formula (I-L) may be further converted to a compound of formula (I-
M) by reacting with a nucleophile of formula M1-R7.
wherein M1 is Li, Mg-halo, Cu-halo or Ce-halo,
R16 is H, alkyl, alkenyl or alkynyl; and
all other variables are as defined in connection with Schemes 1-5.
The addition of the nucleophile M1-R16 to the aldehyde of formula (I-L) is typically
carried out at a temperature ranging from about -78 to about 5 °C. Suitable solvents
for this reaction include but are not limited to, tetrahydrofuran, dioxane, diethyl
ether, toluene, 1,2-dimethoxyethane, and hexanes.
As an alternative to the previously described method, a compound of the formula (I-J)
may also be prepared by conversion from a compound of formula (I-M). More
specifically, a compound of formula (I-J) may be prepared by oxidation of a compound
of formula (I-M).
wherein R16 is H, alkyl, alkenyl or alkynyl; and
all other variables are as defined in connection with Schemes 1-5.
This reaction can be carried out using a wide variety of conventional oxidizing agents.
Examples of suitable oxidizing agents include but are not limited to, manganese
dioxide, dimethyl suIf oxide / oxalyl chloride / triethyl amine, pyridinium
chlorochromate, pyridinium dichromate, and tetrapropylammonium perruthenate / 4-
methylmorpholine N-oxide. Suitable solvents for this reaction include but are not
limited to, dichloromethane, chloroform, diethyl ether, toluene and tetrahydrofuran.
Further, a compound of formula (I-J) may be converted to a compound of formula (I-
M") by reacting with a nucleophile of formula M"-R16.
wherein M1 is Li, Mg-halo, Cu-halo or Ce-halo;
R16 is H, alkyl, alkenyl or alkynyl; and
all other variables are as defined above in connection with Schemes 1-5.
Nucleophiles of formula M1-R16 are commercially available or can be prepared using
conventional knowledge in the art
The addition of the nucleophile to the aldehyde of formula (I-J) is typically carried out
at a temperature ranging from about -78 to about 5 °C. Suitable solvents for this
reaction include but are not limited to, tetrahydrofuran, dioxane, diethyl ether,
toluene, 1,2-dimethoxyethane,and hexanes.
A compound of formula (I-M) may be further converted to a compound of formula (I-
N) by halogenating the compound of formula (I-M).
wherein X2 is halo;
R16 is H, alkyl, alkenyl or alkynyl; and
all other variables are as defined in connection with Schemes 1-5.
This reaction may be carried out using any conventional halogenating reagent
Examples of suitable halogenating reagents include but are not limited to
triphenylphosphine / iodine / imidazole, triphenylphosphine / carbon tetrabromide,
phosphorous pentachloride, thionyl chloride, phosphorous tribromide, hydrofluoric
acid / potassium fluoride, and dimethyl sulfide / A/-bromosuccinimide. Suitable
solvents for this reaction include but are not limited to tetrahydrofuran, dioxane,
diethyl ether, dichloromethane, chloroform, acetonitrile, toluene, 1,2-
dimethoxyethane, and hexanes.
A compound of formula (I-N) may be further converted to a compound of formula (I-
0) using a reduction.
wherein X2 is halo;
R16 is H, alkyl. aikenyl or alkynyl; and
all other variables are as defined above in connection with Scheme 2.
This reaction may be carried out in an inert solvent using a variety of conditions.
Examples of suitable reducing agents for this reaction include but are not limited to,
lithium / ammonia, zinc / acetic acid, lithium triethylborohydride, tributyltin hydride,
lithium aluminum hydride, and samarium (II) iodide. Suitable solvents for this reaction
vary considerably depending upon the chosen reducing agent. Examples of suitable
solvents include but are not limited to, tetrahydrofuran, diethyl ether, 1,2-
dimethoxyethane, dioxane, toluene, and hexanes.
A compound of formula (I-L) may be further converted to a compound of formula (I-
P) by reacting with a compound of the formula (XXV).
wherein all variables are as defined above in connection with Schemes 1-5.
This reaction is carried out in an inert solvent, conveniently at room temperature. The
synthesis and use of the compound of formula (XXV) is analogous to that described in
Mueller, S., et al., Synlett 6:521-522(1996). Typically, the reaction is carried out using
methanol as the solvent and a base such as potassium carbonate.
In another embodiment, a compound of formula (I-Q) may be converted to a
compound of formula (I-R), which may in turn be converted to a compound of
formula (I-S), or a compound of formula (I-Q) may be converted directed to a
compound of formula (I-S).
wherein
n"is 0,1,2 or 3;
each L6 is the same or different suitable leaving group; and
all other variables are as defined above in connection with Schemes 1-5.
Compounds of formula (I-Q) may be prepared according to any of the methods
described herein above. The compound of formula (I-Q) may then be converted to a
compound of formula (I-R) or a compound of formula (I-S).
The compound of formula (I-R) may be prepared by either of two methods. According
to one method, a compound of formula (I-R) is prepared by reacting a compound of
formula (I-Q) with a compound of formula: LG-(R2)cc-LG (XXVII), wherein all variables
are as defined above. Specific examples of suitable leaving groups include but are not
limited to -Cl, -Br, -I, -OSO2CH3 and -OSO2-Phenyl. Suitable compounds of formula
(XXVII) are commercially available or may be prepared using conventional techniques.
The reaction may be carried out in an inert solvent, conveniently at room temperature,
in the presence of a suitable base. Examples of suitable bases for this reaction include
but are not limited to, potassium carbonate, sodium carbonate, cesium carbonate,
sodium hydride, and potassium hydride. Examples of suitable inert solvents for this
reaction include but are not limited to, N,N-dimethylformamide, tetrahydrofuran,
dioxane.and 1,2-dimethoxyethane.
According to a second method, a compound of formula (I-R) is prepared by reacting a
compound of formula (I-Q) with a compound of formula: HO-(R2)cc-LG (XXVIII),
wherein all variables are as defined above. Specific examples of suitable leaving •
groups include those described above. Compounds of formula (XXVIII) are
commercially available or can be prepared using conventional techniques. The
reaction is carried out in an inert solvent under standard Mitsunobu conditions. See,
Hughes, D.L, Org. React. 42335-656 (1992); and Mitsunobu, 0., Synthesis 1-28
(1981). Typically the compound of formula (I-Q) and the compound of formula
(XXVIII) are reacted together with a triarylphosphine, and a dialkyl azodicarboxylate at
room temperature. Examples of suitable triarylphosphines include but are not limited
to, triphenylphosphine, tri-p-tolylphosphine, and trimesitylphosphine. Examples of
suitable dialkyl azodicarboxylates include but are not limited to, diethyl
azodicarboxylate, diisopropyl azodicarboxylate, and di-te/t-butyl azodicarboxylate.
Examples of suitable inert solvents for this reaction include but are not limited to,
tetrahydrofuran, dioxane, 1,2-dimethoxyethane, dichloromethane, and toluene.
The compound of formula (I-R) may be converted to a compound of formula (I-S) by
reaction with a suitable nucleophile for installing the group R4 Examples of suitable
nucleophiles include but are not limited to ammonia, primary and secondary amines,
metal alkoxides, metal thioalkoxides. potassium cyanide, sodium azide, organolithium
reagents, organocuprates, and Grignard reagents. The specific conditions for these
displacements vary, but the use of these types of nucleophiles for the installation of a
group as defined by R4 are conventional in the art Displacement of the leaving group
with such a nucleophile would either install the R4 functionality or provide an
intermediate from which, the R4 functional group could be readily installed according
to conventional methods by one skilled in the art.
Alternatively, a compound of formula (I-S) may be prepared directly from a compound
of formula (I-Q) using procedures analogous to those described above for the
conversion of a compound of formula (1-Q) to a compound of formula (I-R). More
specifically, a compound of formula (I-S) may be prepared by reacting a compound of
formula (I-Q) with a compound of formula: LG-(R2)cc-R4 (XXIX),using conditions
analogous to those described above for the reaction of a compound of formula (I-Q)
with a compound of formula (XXVII). Compounds of formula (XXIX) are commercially
available or can be prepared using conventional techniques.
In another embodiment, a compound of formula (I-Q) is converted to a compound of
formula (I-S) by reacting with a compound of formula: HO-(R2)cc-R4 (XXX) under the
conditions described above for the reaction of a compound of formula (I-Q) with a
compound of formula (XXVIII). Compounds of formula (XXX) are commercially
available or can be prepared using conventional techniques.
As a further example, a compound of formula (l-T) may be converted to a compound
of formula (I-U), which may optionally be further converted to a compound of
formula (I-V).
wherein:
R15 is alkyl or phenyl; and
all other variables are as defined in connection with Schemes 1-5 above.
A compound of formula (I-T) may be converted to a compound of formula (I-U) by
reacting with a suitable acid, such as trifluoroacetic acid (TFA). This reaction may be
carried out neat or in an inert solvent at ambient temperature. Suitable solvents for
this reaction include but are not limited to, dichloromethane and chloroform.
The compound of formula (I-U) may be further converted to a compound of formula
(I-V) by reacting with sulfonyl chlorides of formula (XXXI). The reaction may be
carried out in an inert solvent at ambient temperature using a variety of bases.
Examples of suitable bases include but are not limited to, triethylamine, N,N-
diisopropylethylamine, and pyridine. Suitable solvents for this reaction include but are
not limited to, dichloromethane, chloroform, tetrahydrofuran, 1,2-dimethoxyethane,
dioxane, and N,N-dimethyiformamide.
In another embodiment, a compound of formula (I-W) may be converted to a
compound of formula (l-X). A compound of formula (I-X) may be further converted
to a compound of formula (I-Y).
wherein R5a is selected from the group consisting of -OR7 and -NR7R8; and
all other variables are as defined in connection with Schemes 1-5 above.
A compound of formula (I-W) may be oxidized to a compound of formula (I-X) using a
conventional oxidizing agent such as for example, 3-chlorpperoxybenzoic acid.
Reaction of the compound of formula (I-X) with a suitable nucleophile of formula R5a
will convert a compound of formula (I-X) to a compound of formula (I-Y). Specific
examples of suitable nucleophiles for this reaction include but are not limited to
sodium hydroxide, sodium acetate, ammonia, and mono and di-substituted amines.
The reaction with the nucleophile is typically carried out using equimolar or a slight
excess of the nucleophile in an inert solvent, such as THF, at ambient or elevated
temperatures. In another embodiment, a compound of formula (I-X) may be
converted to a compound of formula (I-Y) in a sealed tube at elevated temperatures
between 80°C and 120°G, using excess ammonia in an appropriate solvent such as
methanol, ethanol, isopropanol, tetrahydrofuran and dioxane.
Similarly, a compound of formula (I-AA) may also be converted to a compound of
formula (I-BB) by oxidation, and the compound of formula (I-BB) may be converted to
a compound of formula (I-CC) by reaction with ammonia.
wherein all variables are as defined in connection Schemes 1-5 above.
The step of converting a compound of formula (I-AA) to a compound of formula (I-
BB) may be carried out by reacting a compound of formula (I-AA) with a suitable
oxidizing agent, such as for example 3-chloroperoxybenzoic acid. The compound of
formula (I-BB) may be converted to a compound of formula (I-CC) by reaction with
excess ammonia in a sealed tube at elevated temperature between about 80 and
about 120 °C in a suitable solvent Suitable solvents for this reaction include but are
not limited to methanol, ethanol, isopropanol, tetrahydrofuran and dioxane.
A further example of a process for converting a compound of formula (I) to a different
compound of formula (I) includes the reaction of a compound of formula (I-DD) with
a thionating reagent to prepare a compound of formula (I-EE).
The reaction may be carried out in an inert solvent and optionally heated to a
temperature of from about 65 to above about 100°C. Examples of suitable thionating
reagents include but are not limited to phosphorus pentasulfide, 2,4-bis(4-
methoxyphenyl)-1l3-dithia-2,4-diphosphetane-2,4-disulfide and the like. Suitable
solvents include but are not limited to xylene; dioxane and toluene.
Further, a compound of formula (I-FF) may be converted to a compound of formula (I-
6G) by reaction with an azide source in an inert solvent
wherein all variables are as defined in connection with Schemes 1-5 above.
Examples of suitable azide sources include but are not limited to hydrazoic aicd,
sodium azide with ammonium chloride, sodium azide with aluminum chloride, and
sodium azide with zinc(ll) bromide. By way of example some preferred solvents include
but are not limited to dimehtylformamide, dimethylsulfoxide, N-methylpyrrolidinone,
toluene and the like. The reaction may be optionally heated to a temperature of from
about 23 to about 150°C.
In another embodiment, a compound of formula (I-HH) may be converted to a
compound of formula (l-ll) using a coupling protocol.
wherein all variables are as defined in any of Schemes 1-5.
The conversion reaction can be carried out by reacting a compound of formula (I-HH)
with a suitable coupling reagent in an inert solvent followed by the addition of a
hydroxylarnine source, and optionally a base. Suitable coupling reagents include but
are not limited to 1,1-carbonyldiimidazole,oxalyl chloride, dicyclohexylcarbodiimide
and 1-(N,N-diphenylcarbamoyl)pyridinium chloride. Preferably the.hydroxylamine is
hydroxylamine hydrochlorlde. Suitable bases include but are not limited to
triethylamine, sodium methoxide and diisoproylethylamine. The reaction may be
optionally heated to a temperature of from about O°C to about 80°C. Examples of
suitable solvents for this reaction include but are not limited to dimethylformamide,
dichloromethane and tetrahydrofuran.
In yet another example of a conversion using a coupling protocol a compound of
formula (I-KK) is prepared from a compound of formula (I-JJ) as follows.
wherein n" is 0,1,2 or 3;
PG is a protecting group and
all other variables are as defined in any of Schemes 1-5 above.
The protecting group is typically carboxylic acid protecting group which when
removed yields the acid. The cleavage of the carboxylic acid protecting group can be
accomplished through many different methods conventional in the art See,
Kocienski, P.J. Protecting Groups, Georg Thieme Verlag, Stuttgart, 1994; and Greene,
T.W., Wuts, P. G. M. Protecting Groups in Organic Synthesis (2nd Edition), J. Wiley and
Sons, 1991.
Following the removal of the protecting group, the resulting carboxylic acid is reacted
using a coupling protocol to yield the compound of formula (I-KK). The reaction can
be carried out by reacting the deprotected compound of formula (I-JJ) with a suitable
coupling reagent in an inert solvent, followed by the addition of a primary or
secondary amine, and optionally a base. Suitable coupling reagents include but are
not limited to 1,1-carbonyldiimidazole, oxalyl chloride, dicyclohexylcarbodiimide and
0-(7-azabenzotriazol-1-yl)-1,1,3l3-tetramethyluronium hexafluorophosphate.
Suitable bases include but are not limited to triethylamine, diisoproylethylamine and
the like. The reaction may be optionally heated to a temperature of from about 0°C to
about 80°C. Examples of suitable solvents include but are not limited to
dimethylformamide, dichloromethane and tetrahydrofuran.
in yet another example of a conversion using a coupling protocol a compound of
formula (I-MM) is prepared from a compound of formula (I-LL) as follows.
wherein n" is 0,1,2 or 3;
PG is a protecting group and
all other variables are as defined in any of Schemes 1-5 above.
The protecting group is amino protecting group which when removed yields the
amine. The cleavage of the amino protecting group can be accomplished through
many different methods conventional in the art. See, Kocienski, PJ. Protecting
Groups, Georg Thieme Verlag, Stuttgart, 1994; and Greene, T.W., Wuts, P. G. M.
Protecting Groups in Organic Synthesis (2nd Edition), J. Wiley and Sons, 1991.
Following the removal of the protecting group, the resulting amine is reacted using a
coupling protocol to yield the compound of formula (I-MM). The reaction can be
carried out by reacting the deprotected compound of formula (I-LL) with a carboxylic
acid in the presence of a suitable coupling reagent in an inert solvent, and optionally a
base. Suitable coupling reagents include but are not limited to 1,1-
carbonyldiimidazole, oxalyl chloride, dicyclohexylcarbodiimide and 0-(7-
azabenzotriazol-1-yl)-1,1,3l3-tetramethyluroniumhexatluorophosphate. Suitable
bases include but are not limited to triethylamine, diisoproylethylamine and the like.
The reaction may be optionally heated to a temperature of from about 0°C to about
80°C. Examples of suitable solvents include but are not limited to dimethylformamide,
dichloromethane and tetrahydrofuran.
Based upon this disclosure and the examples contained herein one skilled in the art
can readily convert a compound of formula (I) or a pharmaceutically acceptable salt,
solvate or physiologically functional derivative thereof into another compound of
formula (I), or a pharmaceutically acceptable salt, solvate or physiologically functional
derivative thereof.
The present invention also provides radiolabeled compounds of formula (1) and
biotinylated compounds of formula (I) and solid-support-bound versions thereof.
Radiolabeled compounds of formula (!) and biotinylated compounds of formula (I) can
be prepared using conventional techniques. For example, radiolabeled compounds of
formula (f) can be prepared by reacting the compound of formula (I) with tritium gas
in the presence of an appropriate catalyst to produce radiolabeled compounds of
formula (I).
In one embodiment the compounds of formula (I) are tritiated.
The radiolabeled compounds of formula (I) and biotinylated compounds of formula (i)
are useful in assays for the identification of compounds which inhibit PLK, for the
identification of compounds for the treatment of a condition mediated by PLK, for the
treatment of susceptible neoplasms, for the treatment of conditions characterized by
inappropriate proliferation, for the inhibition of proliferation of a cell and for the
inhitibion of mitosis in a cell. Accordingly, the present invention provides an assay
method for identifying such compounds, which method comprises the step of
specifically binding the radiolabeled compound of formula (I) or the biotinyiated
compound of formula (I) to the target protein or cellular homogenates. More
specifically, suitable assay methods will include competition binding assays. The
radiolabeled compounds of formula (I) and biotinyiated compounds of formula (I) and
solid-support-bound verstions thereof, can be employed in assays according to the
methods conventional in the art.
The following examples are intended for illustration only and are not intended to limit
the scope of the invention in any way, the invention being defined by the claims
which follow.
Reagents are commercially available or are prepared according to procedures in the
literature. In the following structures, "Me" refers to the group -CHs.
Example 1: Methyl 2-chloro-3-oxo-2,3-dihydro-2-thiophenecarboxylate
To a solution of methyl 3-hydroxy-2-thiophenecarboxylate (5.00 g, 31.6 mmol) in
chloroform (10 mL) was added 1M sulfuryl chloride in dichloromethane (34.8 mL, 34.8
mmol) dropwise over 2 minutes under a N2 atmosphere. The mixture was stirred for 4
hours at room temperature and the volatiles removed under reduced presssure. The
solids were recrystallized from hexane to give methyl 2-chloro-3-oxo-2,3-dihydro-2-
thiophenecarboxylate (4.60 g, 76%) as white needles. 1H NMR (CDCl3): 8 8.38 (d, 1 H),
6.23 (d, 1 H), 3.84 (s. 3 H); MS m/z 193 (M+1).
To a solution of methyl 2-chloro-3-oxo-2,3-dihydro-2-thiophenecarboxylate (0.050 g,
0.26 mmol) in chloroform (1.0 mL) (and in a separate reaction acetic acid (1.0 mL)) was
added benzimidazole (0.061 g, 0.52 mmol) to each reaction. The chloroform reaction
was stirred for 22 hours at room temperature and then diluted with chloroform (2.0
mL). The organic phase was washed with water (1.0 ml) and the phases were
separated. The organic phase was analyzed by LC-MS and then concentrated under
reduced pressure to give a solid residue. The residue was triturated with water (2 mL),
filtered and dried. The acetic acid reaction was stirred at room temperature for 66
hours, and analyzed by LC-MS. The reaction was diluted with water (5 mL), then
cooled on ice for 30 minutes and the solids collected by filtration and dried at 50°C
under vacuum. The solids from both the chlorofofm and acetic acid reactions were
analyzed by 1H-nmr When both reactions were of sufficient purity they were
combined to give methyl 5-(1H-benzimidazol-1-yl)-3-hydroxy-2-
thiophertecarboxylate (0.058 g, 41%) as an orange-brown solid. "H NMR (DMSO-de):
610.87 (br s. 1H), 8.69 (s, 1H), 7.80 (m, 2H), 7.39 (m, 2H), 7.14 (s, 1H). 3.79 (s, 3H). MS
m/z 275 (M+1).
Example 2B: Methyl 5-(1H benzimidazol-1-vl)-3-[(2-methvlbenzvl)oxv]-2-
thiophenecarboxylate and 5-(1 H-Benzimidazol-1-vl)-3-[(2-methvlbenzyl)oxv]-2-
thiophenecarboxamide.
To a mixture of methyl 5-(1H-benzirnidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate
(0.058 g, 0.21 mmol) and potassium carbonate (0.032 g, 0.23 mmol) in
dimethylformamide (0.50 mL) was added a-bromo-o-xylene (31 (µL, 0.23 mmol). The
mixture was stirred for 6 hours at room temperature and then diluted with water (1.0
mL). The mixture was extracted with ether (2x3 mL) and the combined ether extract
was concentrated to dryness under reduced pressure. The residue was treated with
2M ammonia in methanol (3 mL) in a Pyrex test tube sealed with a Teflon-lined screw
cap, and the reaction heated to 80°C with magnetic stirring for 3 days. The reaction
was cooled and fresh 2M ammonia in methanol (2 mL) was added and the test tube
re-sealed and heated at 80°C for an additional 2 days. The reaction was cooled and
silica gel (0.5 g) was added to the reaction mixture, followed by evaporation of the
volatiles under reduced pressure. The pre-adsorbed solids were loaded into a solid
loading cartridge and subjected to a gradient elution using ethyl acetaterhexane
(25:75) to ethyl acetate (100%) using a RediSep silica gel cartridge (4.2 g; ISCO). The
methyl ester (higher Rf) was readily separated from the carboxamide product and the
appropriate fractions were combined and concentrated under reduced pressure to give
methyl 5-(1 H-benzimidazol-1 -yt)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate
(0.0092 g) as ah off-White solid. "H NMR (DMSO-de): 8 8.72 (s, 1H), 7.86 (d, 1H), 7.81
(d, 1H), 7.76 (s, 1H), 7.55 (d, 1H). 7.42 (m, 1H), 7.38 (dd, 1H). 7.26 (m, 3H), 5.38 (s, 2H),
3.77 (s, 3H), 2.39 (s, 3H). MS m/z 379 (M+1); and 5-(1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxamide (0.0136 g) as a tan solid. "H NMR (DMSO-
de): 8 8.65 (s, 1H), 7.80 (d, 1H), 7.68 (s+br s. 2H), 7.49 (d, 1H), 7.40 (m, 3H), 7.28 (m,
3H), 6.85 (br s, 1H), 5.43 (s. 2H), 2.39 (s, 3H). MS m/z 364 (M+1).
Example 3: Methyl 5-(1H-benzimidazol-1-yl)-3-[(2-methvlbenzvl)oxy]r2-
thiophenecarboxylate
To a mixture of methyl 5-(1H-benzimidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate
(0.500 g, 1.82 mmol) and potassium carbonate (0.277 g, 2.01 mmol) in
dimethylformamide (5.0 mL) was added a-bromo-o-xylene (0.27 mL, 2.01 mmol). The
mixture was stirred for 18 hours at room temperature and then diluted with water (20
mL) and extracted with ether (2 x 50 mL). The organic layer was washed with water
(10 mL), saturated brine (10 mL) and dried (MgSCU). Concentration of the organic
phase under reduced pressure gave 0.395 g of crude methyl 5-(1 H-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate as a yellow solid. 1H NMR
(DMSO-d6): d 8.71 (s. 1H). 7.84 (d, 1H), 7.79 (d, 1H), 7.75 (s, 1H), 7.53 (d, 1H). 7.42 (dd,
1H), 7.38 (dd, 1H), 7.24 (m, 3H), 5.36 (s, 2H). 3.75 (s, 3H), 2.37 (s, 3H). MS m/z 379
(M+1).
Example 4: 5-(1H-Benzimidazol-1 -vl)-3-[(2-methvlbenzyl)oxy]-2-
thiophenecarboxamide
A mixture of methyl 5-(1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxylate (0.114 g, 0.302 mmol) and 2M methanolic ammonia (5 mL) was
heated at 80°C for 48 h in a Pyrex test tube fitted with a Teflon-lined screw cap. The
reaction was cooled and charged with fresh 2M methanolic ammonia (2 mL) and
heated at 80°C for 72 h. The reaction was again cooled and recharged with fresh 2M
methanolic ammonia (2 mL) and heated at 80°C for 48 h. The reaction mixture was
concentrated under reduced pressure and the solid residue was dissolved in
methanohethyl acetate (1:1). Silica gel (0.5 g) was added to the solution and the
volatiles were removed under reduced pressure. The pre-adsorbed material was
packed into a solid loading cartridge and eluted onto a RediSep silica gel cartridge (4.2
g; ISCO) using ethyl acetate; collected 18 mL fractions. The appropriate fractions were
combined and concentrated to dryness to give a solid residue. The solids were
triturated with methanol-.ether (1:2) and collected by filtration, rinsed with ether (2
mL) and dried to give 0.021 g of 5-(1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-
2-thiophenecarboxamide as a light yellow solid. 1H NMR (DMSO-de): 8 8.65 (s, 1H),
7.80 (d, 1H), 7.69 (s, 1H), 7.77 ft 6.85 (2xbr s, 2H), 7.48 (d, 1H), 7.40 (m, 3H), 7.28 (m,
3H), 5.43 (s, 2H), 2.39 (s, 3H). MS m/z 364 (M+1).
Example 5: 5-(1H-Benzimidazol-1-vl)-3-[(2-methvlbenzyl)oxyl-2-
thiophenecarboxylic acid
To a solution of methyl 5-(1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxylate (0.393 g, 1.04 mmol) in dioxane (4.0 mL) was added aqueous 1M
lithium hydroxide (4.0 mL). The mixture was stirred for 18 hours at room temperature.
The reaction mixture was acidified to pH 1-2 with 1N hydrochloric acid (4 ml) and the
solids were collected by filtration and dried to give 0.334 g of 5-(1H-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylic acid as a yellow solid. "H NMR
(DMS0-d6): d 12.8 (br S, 1H), 8.69 (s, 1H), 7.80 (2xd, 2H), 7.70 (s. 1H), 7.52 (d, 1H), 7.40
(m, 2H), 7.24 (m, 3H), 5.32 (s, 2H), 2.37 (s, 3H). MS m/z 365 (M+1).
Example 6: 5-(1 H-Benzimidazol-1 -yl)-N-methy|-3-[(2-methvlbenzyl)oxy]-2-
To a mixture of 5-(1 H-benzimidazol-1-Yl)-3-[(2-methYlbenzyl)oxy]-2-
thiophenecarboxylic acid (0.050 g, 0.14 mmol) in dichloromethane (2 mL) was added
1-chloro-2,N,N-trimethylpropenylamine (0.027 mL, 0.20 mmol) and the reaction
mixture was stirred for 1 hour at room temperature. Methylamine (8M) in ethanol
(52 µl, 0.42 mmol) was added to the reaction mixture, followed by addition of
diisopropylethylamine (49 µL, 0.28 mmol). The reaction was complete after two hours.
After stirring for 66 hours the reaction was partitioned between dichloromethane (3
mL) and water (1 mL). The biphasic mixture was separated and the organic phase
dried over MgSCk The organic phase was concentrated under reduced pressure and
the residue was triturated with ether. The solids were collected by filtration and dried
to give 0.037 g of 5-(1 H-benzimidazol-1-yl)-N-methyl-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxamide as a yellow solid. 1H NMR (DMSO-d6): 6 8.63 (s, 1H), 7.80 (d,
1H), 7.74 (d, 1H), 7.63 (s, 1H), 7.42 (m, 4H), 7.27 (m, 3H), 5.44 (s, 2H), 2.81 (d, 3H), 2.39
(s,3H). MS m/z 378(M+1).
Example 7: 5-(1 H-Benzimidazol-1 -yl)-N,N-dimethvl-3-[(2-methylbenzyl)oxy]-2-
In a similar manner as described for Example 6,5-(1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxylic acid (0.050 g, 0.14 mmol) in
dichloromethane (2 mL), 1-chloro-2,NIN-trimethylpropenylamine (0.027 mL, 0.20
mmol), dimethylamine (2M) in tetrahydrofuran (210 U.L, 0.42 mmol) and
diisopropylethylamine (49 µL 0.28 mmol) gave 5-(1H-enzimidazol-1-yl)-N,N-
dimethyl-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxamide (0.032 g, 60%) as a tan
solid. "H NMR (DMSO-de): d 8.63 (s, 1H), 7.79 (2xd. 2H), 7.64 (s, 1H). 7.40 (m, 3H), 7.26
(m, 3H), 5.30 (s, 2H), 2.98 (s, 6H), 2.34 (s, 3H). MSm/z 392 (M+1).
Example 8: 5-(1 H-Benzimidazol-t-yl)-N-isopropvl-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxamide
In a similar manner as described for Example 6,5-(1 H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxylic acid (0.050 g, 0.14 mmol) in
dichloromethane (2 mL), 1-chloro-2,N,N-trimethylpropenylamine (0.027 mL, 0.20
mmol), isopropylamine (36 \xl, 0.42 mmol) and diisopropylethylamine (49 nL, 0.28
mmol) gave 5-(1 H-benzimidazol-1-yl)-/V-isopropyl-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxamide (0.033 g, 59%) as a yellow solid. 1H NMR (DMSO-de): 5 8.66 (s,
1H), 7.81 (2xd. 2H), 7.73 (s, 1H), 7.52 (d, 1H), 7.44 (m, 1H), 7.38 (m, 1H), 7.30 (m, 3H),
7.14 (d, 1H), 5.44 (s, 2H), 3.99 (m, 1H), 2.41 (s, 3H), 1.06 (d, 6H). MS m/z 406 (M+1).
Example 9: 5-(1 H-Benzimidazol-1-yl)-N-(2-hydroxvethvl)-3-[(2-methylbenzyl)oxy]-
2-thiophenecarboxamide
In a similar manner as described for Example 6,5-(1H-benzimidazol-1-yl)-3-(2-
methylbenzyloxy)-2-thiophenecarboxylic acid (0.050 g, 0.14 mmol) in
dichloromethane (2 mL), 1-chloro-2,N,N-trimethylpropenylamine (0.027 mL, 0.20
mmol), ethanolamine (25 µL, 0.42 mmol) and diisopropylethylamine (49 µL, 0.28
mmol) gave 5-(1 H-benzimidazoI-1-yl)-N-(2-hydroxyethyl)-3-[(2-methylbenzyl)oxy]-
2-thiophenecarboxamide (0.036 g, 64%) as a yellow solid. 1H NMR (DMSO-de): 8 8.65
(s, 1H), 7.80 (2xd, 2H), 7.71 (s. 1H), 7.54 (m, 2H), 7.44 (m, 1H), 7.37 (m, 1H), 7.27 (m,
3H), 5.45 (s, 2H). 4.80 (t, 1H), 3.46 (m, 2H), 3.36 (m, 2H), 2.40 (s, 3H). MS m/z 408
(M+1).
Example 10: 5-(1 H-Benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]-N-phenyl-2-
thiophenecarboxamide
In a similar manner as described for Example 6,5-(1 H-benzimidazol-1 -yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxylic acid (0.050 g, 0.14 mmol) in
dichloromethane (2 mL), 1-chloro-2,N,N-trimethylpropenylamine (0.027 mL, 0.20
mmol), aniline (38 µL, 0.42 mmol) and diisopropylethylamine (49 jaL, 0.28 mmol) gave
5-(1H-benzimidazol-1-yI)-3-[(2-methylbenzyl)oxy]-N-phenyl-2-
thiophenecarboxamide (0.044 g, 73%) as a yellow solid. 1H NMR (DMSO-d6): 89.80 (s,
1H), 8.72 (s, 1H), 7.85 (m, 2H). 7.81 (s, 1H), 7.61 (d, 1H), 7.41 (m, 4H). 7.32 (m. 5H), 7.09
(m, 1H), 5.56 (s, 2H), 2.44 (s, 3H). MS m/z 440 (M+1).
Example 11: 5-(1 H-Benzimidazol-1-yl)-N-benzyl-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxamide
In a similar manner as described for Example 6,5-(1 W-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxylic acid (0.050 g, 0.14 mmol) in
dichloromethane (2 mL), 1-chloro-2,N,N-trimethylpropenylamine (0.027 mL, 0.20
mmol), benzylamine (46 µL, 0.42 mmol) and diisopropylethylamine (49 nl, 0.28 mmol)
gave 5-(i W-benzimidazol-1-yl)-A/-benzyl-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxamide (0.038 g, 61%) as a yellow solid. 1H NMR (DMSO-ds): 5 8.65 (s,
1H), 7.81 (m, 3H), 7.69 (s, 1H), 7.42 (m, 3H), 7.27 (m, 8H), 5.43 (s, 2H), 4.49 (d, 2H), 2.29
(s,3H). MS m/z 454 (M+1).
Example 12: 5-(1H-Benzimidazol-1-yl)-3-benzvloxv-2-thiophenecarboxamide
In a similar manner as described for Example 4, methyl 5-(1H-benzimidazol-1-yl)-3-
[(2-methylbenzyl)oxy]-2-thiophenecarboxylate (0.109 g, 0.299 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(1H-benzimidazol-1--yl)-3-benzyloxy-2-
thiophenecarboxamide (0.031 g, 30%) as a white solid. 1H NMR (DMSO-ds): 8 8.63 (s,
1H). 7.76 (dd, 2H), 7.70 ft 7.01 (2xbr s, 2H), 7.64 (s, 1H), 7.55 (d. 2H), 7.44 (m, 5H), 5.42
(s,2H). MS m/z 350 (M+1).
Example 13: 5-(1 H-Benzimidazol-1 -vl)-3-[(3-methylbenzyl)oxy]-2-
thiophenecarboxamide
In a similar manner as described for Example 4, methyl 5~(1 W-benzimidazol-1-yl)-3-
[(3-methylbenzyl)oxy]-2-thiophenecarboxylate (0.114 g, 0.301 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(1H-benzimidazol-1-yl)-3-l(3-methylbenzyl)oxy]-
2-thiophenecarboxamide (0.019 g, 17%) as a white solid. "H NMR (DMSO-ds): 5 8.63
(s, 1H), 7.77 (dd, 2H), 7.70 ft 7.00 (2xbr s, 2H), 7.63 (s, 1H), 7.36 (m, 5H), 7.19 (d, 1H),
5.37 (s, 2H), 2.33 (s, 3H). MS m/z 364 (M+1).
Example 14: 5-(1 H-Benzimidazoi-1-vO-3-[(3-methoxybenzyl)oxv]-2-
thiophenecarboxamide
In a similar manner as described for Example 4, methyl 5-(1 H-benzimidazol-1-yl)-3-
[{3-methoxybenzyl)oxy]-2-thiophenecarboxylate (0.118 g, 0.299 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(1H-benzimidazol-1-yl)-3-[(3-
methoxybenzyl)oxy]-2-thiophenecarboxamide (0.034 g, 30%) as an off-white solid.
1H NMR (DMSO-d6): 8 8.63 (s. 1H), 7.77 (dd, 2H), 7.66 ft 7.05 (2xbr s, 2H), 7.63 (s, 1H),
7.38 (m, 3H), 7.12 (m, 2H), 6.94 (d, 1H), 5.38 (s, 2H), 3.76 (s, 3H). MS m/z 380 (M+1).
Example 15: 5-(1H-Benzimidazol-1-yl)-3-[(3-chlorobenzyl)oxy]-2-
thiophenecarboxamide
In a similar manner as described for Example 4, methyl 5-(1 H-benzimidazol-1-yl)-3-
[(3-chlorobenzyl)oxy]-2-thiophenecarboxylate (0.120 g, 0.301 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(1H-benzimidazoi-1-yl)-3-[(3-chlorobenzyl)oxy]-
2-thiophenecarboxamide (0.031 g. 27%) as a white solid. ,"H NMR (DMSO-de): 8 8.62
(s, 1H), 7.80 (d, 1H). 7.70 (m, 4H), 7.63 (s, 1H), 7.54 ft 7.09 (2xbr s, 2H), 7.42 (m, 3H),
5.41 (s, 2H). MS m/z 384 (M+1).
Example 16: 5-(1H-Benzimidazol-1-yl)-3-[(4-methylbenzyl)oxy]-2-
thiophenecarboxamide
In a similar manner as described for Example 4, methyl 5-(1 H-benzimidazol-1-yl)-3-
[(4-methylbenzyl)oxy]-2-thiophenecarboxylate (0.114 g, 0.301 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(iW-benzimidazol-1-yl)-3-[(4-methylbenzyl)oxy]-
2-thiophenecarboxamide (0.0069 g, 6%) as an off-white solid. "H NMR (DMSO-de): 8
8.63 (s, 1H). 7.78 (dd, 2H)t 7.69 ft 6.98 (2xbrs, 2H), 7.64 (s, 1H), 7.40 (m, 4H), 7.24 (d.
2H), 5.36 (s, 2H), 2.31 (s. 3H). MS m/z 364 (M+i).
Example 17: 5-(1H-Benzimidazol-1-yl)-3-[(4--chlorobenzyl)oxyl-2-
thiophenecarboxamide.
In a similar manner as described for Example 4 methyl 5-(1 H-benzimidazol-1-yl)-3-
[(4-chlorobenzyl)oxyJ-2-thiophenecarboxylate (0.120 g, 0.301 mmol) and 2M
methanolic ammonia (5 mL) gave 5-(1H-benzimidazol-1-Yl)-3-[(4-chlorobenzy!)oxy]-
2-thiophenecarboxamide (0.015 g, 13%) as an off-white solid. 1H NMR (DMSO-de): 8
8.62 (s, 1H), 7.78 (dd, 2H). 7.70 ft 7.03 (2xbr s, 2H), 7.62 (s, 1H). 7.54 (AB q, 4H)r7.40
(m, 2H), 5.41 (s, 2H). MS m/z 384 (M+1).
Example 18A: Methyl 3-hydroxv-5-(5-methyl-1H-benzimidazol-1-yl)-2-
thiophenecarboxylate and Methyl 3-hydroxy-5-(6-methyl-1H-benzimidazol-1-yl)-2-
thiophenecarboxylate
In a similar manner as described for Example 2A, methyl 2-chloro-3-oxo-2,3-dihydro-
2-thiophenecarboxylate (0.050 g, 0.26 mmol) and 5-methyl-1 H-benzimidazole (0.069
g, 0.52 mmol) in chloroform (1.0 mL), and in a separate reaction acetic acid (1.0 mL),
gave a 1:1 isomer mixture of methyl 3-hydroxy-5-(5-methyI-1H-benzimidazol-1-yl)-
2-thiophenecarboxylate and methyl 3-hydroxy-5-(6-methyl-1 H-benzimidazol-1-yl)-
2-thiophenecarboxylate (0.063 g, 42%) as a light yellow solid 1H NMR (DMSO-ds): 5
10.84 (br s, 2H), 8.63,8.59 (2xs, 2H), 7.65 (m, 4H), 7.22 (m, 2H), 7.12 (d, 2H), 3.79,3.78
(2xs, 6H), 2.47, 2.44 (2xs, 6H). MS m/z 289 (M+1).
Example 18B: Methyl 5-(5-methyl-1H-benzimidazol-1-yl)-3-[(2-methvlbenzyl)oxy]-
2-thiophenecarboxylate / Methyl 5-(6-methyl-1H-benzimidazol-1-yl)-3-[(2-
methylbenzvl)oxy]-2-thiophenecarboxylate and 5-(5-Methyl-1 H-benzimidazol-1-yl)-
3-[(2-methylbenzyl)oxy]-2-thiophenecarboxamide/5-(6-Methyl-1//-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxvl-2-thiophenecarboxamide
In a similar manner as described for Example 2B, a 1:1 isomer mixture of methyl 3-
hydroxy-5-(5-methyl-1H-benzimidazol-1-yl)-2-thiophenecarboxylate and methyl 3-
hydroxy-5-(6-methyl-1 H-benzimidazol-1-yI)-2-thtophenecarboxylate (0.055 g, 0.19
mmol). potassium carbonate (0.029 g, 0.21 mmol), a-bromo-o-xylene (28 µL 0.21
mmol) and dimethylformamide (0.50 mL), followed by 2M methanolic ammonia (3
mL), gave a 1:1 isomer mixture of methyl 5-(5-methyl-1 H-benzimidazol-1-yl)-3-l(2-
methyl-benzyl)oxy]-2-thiophenecarboxylate and methyl 5-(6-methyl-1H-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate (0.017 g, 23%)
as an amber oil. 1H NMR (DMSO-dB): 6 8.67 (s, 1H), 8.62 (s, 1H), 7.74 (d, 1H), 7.73 (s.
2H). 7.67 (d, 1H), 7.60 (s, 2H), 7.54 (d, 2H), 7.26 (m, 8H), 5.37 (s, 4H), 4.09 (q, 2H), 3.77,
3.76 (2xs, 6H), 3.16 (d, 4H), 2.45,2.39 (2xs, 6H). MS m/z 393 (M+1); and a 1:1 isomer
mixture of 5-(5-methyl-1 «-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-
thiophene-carboxamide and 5-(6-methyl-1 H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-2-thiophenecarboxamide (0.057 g, 79%) as a tan solid. 1H NMR
(DMSO-d6): 8 8.59,8.55 (2xs, 2H), 7.67 (m, 4H), 8.64 (s, 2H), 8.59,8.53 (2xs, 2H), 7.50 ft
6.87 (2 br s, 4H), 7.28 (m, 8H), 5.42 (s, 4H), 3.32,3.31 (2xs. 6H), 2.45,2.39 (2xs, 6H). MS
m/z365(M+1).
Example 19A: Methyl 3-hydroxy-5-(5,6-dimethyl-1H-benzimidazol-1-yl)-2-
thiophenecarboxylate.
In a similar manner as described for Example 2A, methyl 2-chloro-3-oxo-2,3-dihydro-
2-thiophenecarboxylate (0.050 g, 0.26 mmol) and 5,6-dimethyl-1 H-benzimidazote
(0.076 g, 0.52 mmol) in chloroform (1.0 ml), and in a separate reaction acetic acid (1.0
mL), gave, methyl 3-hydroxy-5-(5,6-dimethyl-1 H-benzimidazoI-1 -yl)-2-
thiophenecarboxylate (0.079 g, 50%) as a light yellow solid. 1H NMR (DMSO-d6): d
10.81 (brs, 1H), 8.54 ts, 1H), 7.59 (s, 1H), 7.56 (s, 1H). 7.11 (s, 1H), 3.79 (s, 3H), 2.37 (s,
3H), 2.33 (s, 3H). MS m/z 303 (M+1).
Example 19B: Methyl 5-(5.6-dimethyl-1H-benzimidazol-1-yl)-3-[(2-
methylbenzvl)oxv]-2-thiophenecarboxylate and 5-(5,6-Dimethyl-1 H-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxamide.
In a similar manner as described for Example 2B, methyl 3-hydroxy-5-(5,6-dimethyl-
1 H-benzimidazol-1-yl)-2-thiophenecarboxylate (0.074 g, 0.24 mmol), potassium
carbonate (0.037 g, 0.27 mmol), a-bromo-o-xylene (36 µl, 0.27 mmol) and
dimethylformamide (0.50 mL), followed by 2M methanolic ammonia (3 mL), gave
methyl 5-(5,6-dimethyl-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-
thiophenecarboxylate (0.011 g, 11%) as a pale yellow solid. 1H NMR (DMSO-d6): d 8.58
(s, 1H), 7.70 (s, 1H), 7.58 (m, 3H), 7.26 (m, 3H), 5.37 (s, 2H), 3.76 (s, 3H), 2.39 (s, 6H),
2.34 (s, 3H). MS m/z 407 (M+1); and 5-(5,6-dimethyl-1 H-benzimidazol-1-yl)-3-[(2-
methylben.zyl)oxy]-2-thiophenecarboxamide (0.0066 g, 7%) as an off-white solid. 1H
NMR (DMSO-d6): 8 8.50 (s, 1H), 7.68,6.85 (2xbr s, 2H), 7.62 (s, 1H), 7.54 (d, 2H), 7.50
(d, 1H), 7.28 (m, 3H), 5.42 (s, 2H), 2.39 (s, 3H), 2.37 (s, 3H), 2.34 (s, 3H). MS m/z 392
(M+1).
Example 20A: Methyl 5-(5-chloro-1H-benzimidazol-1-yl)-3-hydroxy-2-
thiophenecarboxylate and Methyl 5-(6-chloro-1H-benzimidazol-1-yl)-3-hvdroxv-2-
thiophenecarboxylate
In a similar manner as described for Example 2A, methyl 2-chloro-3-oxo-2,3-dihydro-
2-thiophenecarboxylate (0.050 g, 0.26 mmol) and 5-chloro-1 H-benzimidazole (0.079
g, 0.52 mmol) in chloroform (1.0 mL), and in a separate reaction acetic acid (1.0 ml),
gave a 1:1 isomer mixture of methyl 5-(5-chloro-1H-benzimidazol-1-yl)-3-hydroxy-2-
thiophenecarboxylate and methyl 5-(6-chloro-1 H-benzimidazol-1-yl)-3-hydroxy-2-
thiophenecarboxylate (0.103 g, 64%) as a light yellow solid. ]H NMR (DMSO-de): 8
10.91,10.89 (2xbr s, 2H), 8.76,8.71 (2xs, 2H), 7.89 (s, 1H), 7.82 (d, 1H), 7.81 (s, 2H).
7.42 (m, 2H), 7.17,7.15 (2xs, 2H), 3.79 (2xs, 6H). MS m/z 309 (M+1).
Example 20B: Methyl 5-(5-chloro-1 H-benzimidazol-1-vl)-3-[(2-methylbenzyl)oxv]-2-
thiophenecarboxylate / Methyl S-(6-chloro-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxv3-2-thiophenecarboxylate and 5-(5-Chloro-1H-benzimidazol-1 -yl)-
3-[(2-methylbenzyl)oxy]-2-thiophenecarboxamide / 5-(6-Chloro-l A/-benzirnidazol-1-
yl)-3-[(2-methylbenzyl)bxy]-2-thiophenecarboxamide.
In a similar manner as described for Example 2B, a 1:1 isomer mixture of methyl 5-(5-
chloro-1H-benzimidazol-l-yl)-3-hydroxy-2-thiophenecarboxylate and methyl 5-(6-
chloro-1H-benzimidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate (0.095 g, 0.31
mmol), potassium carbonate (0.047 g, 0.34 mmol), a-bromo-o-xylene (46 U.L, 0.34
mmol) and dimethylformamide (0.50 mL), followed by workup, gave a solid mixture.
Treatment of the residual solids with 2M methanolic ammonia (3 mL) at elevated
temperature, followed by chromatography, gave a mixture of methyl 5-(5-chloro-iH-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate and methyl 5-
(6-chloro-1 H-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate
(0.016 g. 6%) as a pale yellow solid. 1H NMR (DMSO-d6): 5 8:79 (s, 1H), 7.90 (d. 1H),
7.86 (d, 1H), 7.78 (s, 1H), 7.50 (m, 2H), 7.26 (m, 3H), 5.37 (s, 2H), 3.77 (s, 3H), 2.38 (s,
3H). MS m/z 413 (M+D; and a mixture of 5-(5-chloro-1 H-benzimidazol-1-yl)-3-[(2-
methyibenzyl)oxy]-2-thiophenecarboxamide and 5-(6-chloro-1 W-benzimidazol-1 -yl)-
3-[(2~methylbenzyl)oxy]-2-thiophenecarboxamide (0.021 g, 8.5%) as a pale yellow
solid. "H NMR (DMSO-d6): 8 8.72. 8.67 (2 x s, 2H). 7.80 (m. 4H), 7.72 ft 6.88 (2 x br s,
4H), 7.70 (s, 2H). 7.44 (m, 4H), 7.28 (m, 6H), 5.43, 5.42 (2 x s, 4H), 2.39 (2 x s, 6H). MS
m/z 398 (M+1).
Example 21: Methyl 5-(1H-benzimidazol-1-yl)-3-isopropoxy-2-thiophenecarboxylate
To a mixture of methyl 5-(1 H-benztmidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate
(0.150 g, 0.55 mmol) and potassium carbonate (0.083 g, 0.60 mmol) in dimethyl-
formamide (5.0 mL) was added 2-iodopropane (60 pi, 0.60 mmol). The mixture was
heated at 65°C for 3 hours and then additional 2-iodopropane (164 ul) was added to
the reaction. The mixture was heated at 80°C for 64 hours and then diluted with
water (2.0 mL) and extracted with ether (2 x 5.0 mL). The organic layer was washed
with saturated brine (2.0 mL) and dried (MgSO*). The organic layer was filtered and
concentrated under reduced pressure to give a residue which was dissolved in EtOAc
and pre-adsorbed to silica gel (1.5 g) Elution of the silica-adsorbed material on a
RediSep column (4.2 g; ISCO) using a gradient elution EtOAc:hexanes (25:75) to EtOAc
(100) gave 0.082 g of methyl 5-(1H-benzimidazol-1-yl)-3-isopropoxy-2-
thiophenecarboxylate as a yellow solid. MS m/z 317 (M+1).
Example 22: 5-(1H-benzimidazol-1-yl)-3-isopropoxy-2-thiophenecarboxamide
in a similar manner as described for Example 4, methyl 5-(1H-benzimidazol-1-yl)-3-
isopropoxy-2-thiophenecarboxylate (0.080 g, 0.25 mmol) and 7M methanolic
ammonia (3.0 mL) gave 5-(1H-benzimidazol-1-yl)-3-isopropoxy-2-
thiophenecarboxamide (0.045 g, 60%) as an off-white solid. 1H NMR (DMSO-d6): d
8.64 (s, 1H), 7.78 (2xd, 2H), 7.68 ft 6.93 (2xbr s, 2H), 7.55 (s, 1H), 7.37 (2xt, 2H), 4.80
(m, 1H). 1.36 (d, 6H). MS m/z 302 (M+1).
Example 23: 5-(1 H-Benzimidazol-1 -yl)-3-[(2-methvlbenzyl)oxy]thiophene-2-
carbonitrile
5-(1 H-Benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxamide (0.0285
g, 0.0784 mmol) was dissolved in 2 mL of pyridine and cooled to 0 °C. Trifluoroacetic
anhydride (0.017 mL, 0.120 mmol) was added dropwise via syringe. The mixture was
stirred for 15 minutes and warmed to room temperature. After 1 hour, 2 mL of
dichloromethane followed by five drops of trifluoroacetic anhydride were added to
dissolve the insoluble components of the mixture. After 14 hours, the reaction was
poured into dichloromethane and brine. The layers were separated and the aqueous
layer was washed twice with dichloromethane. The combined organic layers were
dried over MgSO4, filtered, and concentrated in vacuo. Purification by flash
chromatography provided 0.0075 g (28%) of 5-(1 W-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carbonitrile as a pale yellow solid. 1H NMR (300 MHz,
DMSO-d6) d 8.71 (s, 1H), 7.83 (s+m, 3H), 7.49-7.25 (m, 6H), 5.44 (s, 2H), 2.40 (s. 3H).
MS (m/z) 346 (m+1).
Example 24: (5-(1 H-Benzimidazol-1-yl)-3-[(2-methylbenzyl)oxv]thien-2-vl)rnethariol
Methyl 5-(1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylate
(0.276 g, 0.729 mmol) was dissolved in 7 mL of dichloromethane and cooled to -78 °C.
Diisobutylaluminum hydride (1.5 M in toluene, 2.0 mL, 3.0 mmol) was added dropwise
via syringe. After 1 hour, an additional quantity of diisobutylaluminum hydride (1.5 M
in toluene, 1.0 mL, 1.5 mmol) was added dropwise via syringe. The reaction was
allowed to stir for an additional 10 minutes. Methanol (1-2 mL) was added dropwise
by pipet, and the mixture was warmed to room temperature. Dilute aqueous
hydrochloric acid {5 percent HCI w/v) was added carefully by pipet The mixture was
poured into ethyl acetate and water, and the layers were separated. The organics were
washed with brine, and the combined aqueous layers were extracted with ethyl
acetate. The combined organic layers were dried over MgSO4, filtered, and
concentrated in vacuo. Purification by flash chromatography provided afforded 0.175
g (68%) of {5-(1H-benzirnidazol-1-yl)-3-[(2-methylbenzyl)oxy]thien-2-yl}methanol
as a tan solid. 1H NMR (300 MHz. DMSO-d6) 8 8.52 (s, 1H), 7.78 (d, J - 7.4 Hz, 1H).
7.64 (d, J= 7.4 Hz, 1H), 7,48 (s, 1H). 7.45-7.19 (m, 6H), 5.42 (br s, 1H), 5.16 (s, 2H), 2.37
(s,3H). MS(m/z)351 (m+1).
Example 25: 5-(1 H-Benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-2-
carbaldehyde
{5-(1 H-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thien-2-yl}methanol (0.0535 g.
0.153 mmol) was dissolved in 5 mL of dichloromethane with stirring. Manganese
dioxide (0.133 g, 1.53 mmol) was added in single portion. The mixture was allowed to
stir for 1 hour and then filtered through a celite pad, washing well with
dichloromethane. The solvent was removed in vacuo, and the solid dried under high
vacuum conditions to yield 0.0508 g (95%) of 5-(1 W-benzimidazol-1 -yl)-3-[(2-
methylbenzyI)oxy]thiophene-2-carbaldehyde as a tan solid. 1H NMR (300 MHz,
DMSO-d6) 8 9.96 (s, 1H), 8.79 (s, 1H), 7.93 (d, J = 7.9 Hz, 1H), 7.83 (s, 1H), 7.83 (d, J -
7.6 Hz, 1H), 7.77-7.35 (m, 3H), 7.31-7.22 (m, 3H), 5.47 (s, 2H), 2.40 (s, 3H).
Example 26: (+/-)-1 -{5-(1 H-Benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thien-2-
ylkthanol
Methyl magnesium bromide (0.35 mL, 3.0 M in diethyl ether, 1.05 mmol) was added to
3 mL of diethyl ether with stirring. The solution was cooled to 0 °C, and 5-(1 H-
benzimidazol-1 -yl)-3~[(2-methylbenzyl)oxy]thiophene-2-carbaldehyde (0.0943 g,
0.271 mmol) in 3 mL of dichloromethane was added dropwise via syringe. The
reaction was stirred for 30 minutes and quenched by the addition of 5 mL of water.
The mixture was warmed to room temperature and enough 5% HCI solution was
added to dissolve the magnesium salts. The mixture was poured into ethyl acetate,
and the layers were separated. The organic layer was washed with brine, and the
combined aqueous layers were extracted with ethyl acetate. The combined organic
layers were dried over MgSO4, filtered, and concentrated in vacuo to afford 0.0965 g
(98%) of (+/-)-1-{5-(1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thien-2-
yl}ethanol as a brown solid. 1H NMR (300 MHz, DMSO-d6) 5 8.51 (s, 1H), 7.77 (d, J =
7.3 Hz, 1H), 7.64 (d, J= 7.5 Hz, 1H), 7.48-7.22 (m, 7H), 5.61 (m, 1H), 5.15 (s. 2H), 5.08
(m, 1H), 2.38 (s, 3H), 1.39,1.36 (2xs, 3H).
Example 27: 1-{5-(1H-Benzimidazol-1-vl)-3-[(2-methvlbenzvl)oxy]thien-2-
yljethanone
Using a procedure as described in Example 25 afforded 1-{5-(1 H-benzimidazol-1-yl)-
3-[(2-methylbenzyl)oxy]thien-2-yl}ethanone. "H NMR (300 MHz, DMSO-d6) 5 8.76 (s,
1H), 7.90 (d, J m 7.9 Hz, 1H), 7.82 (d, J - 7.6 Hz, 1H), 7.78 (s, 1H), 7.55-7.24 (m, 6H),
5.44 (s, 2H), 2.46 (s, 3H), 2.41 (s, 3H).
Example 28: 1-{4-[(2-Methylbenzvl)oxy]thien-2-yl)-1H-benzimidazole
5-(1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-2-thiophenecarboxylic acid. (0.105
g, 0.288 mmol) was dissolved in 4 mL of acetic acid in a flask fitted with a reflux
condenser. The flask was placed in an oil bath set to 80 °C. After 65 hours, the
reaction was cooled to room temperature and poured into ethyl acetate. The solution
was washed with saturated NaHCO3 (3X) and brine. The organic layer was dried over
MgSO4. filtered, and concentrated in vacuo. The crude product was filtered through a
short column of silica gel washing with 1:1 ethyl acetate/hexanes. The filtrate was
concentrated in vacuo to afford 0.0850 g (92%) of 1-{4-[(2-methylbenzyl)oxy]thien-
2-yl}-1 H-benzimidazole as a dark orange oil, which later solidified upon standing. 1H
NMR (300 MHz, DMSO-d6) 8 8.54 (s, 1H), 7.77 (d, J = 7.3 Hz, 1H), 7.69 (d, J = 7.5 Hz,
1H), 7.46-7.20 (m, 7H), 6.80 (d, J - 1.9 Hz, 1H), 5.11 (s, 2H), 2.36 (s, 3H).
Example 29: (5-(1 H-Benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy3thien-2-yl I methyl
acetate
{5-(1 H-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thien-2-yl}methanol (0.0278 g,
0.0793 mmol) was dissolved in 4 mL of dichloromethane with stirring. 4-
Dimethylamino-pyridine (0.0194 g, 0.159 mmol) was added in a single portion. Acetic
anhydride (0.075 mL, 0.795 mmol) was added via syringe. After two hours, the
reaction was poured into ethyl acetate. The organic layer was washed with 5% HCI,
saturated NaHCO3, and Brine. The organic layer was dried over MgSO4, filtered, and
concentrated in vacuo. The residue was filtered through a short column of silica gel
washing with 1:1 ethyl acetate/hexanes. The filtrate was concentrated in vacuo to
provide 0.0276 g (89%) of {5-(1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thien-
2-yl}methyl acetate as a dark oil, which later solidified upon standing. 1H NMR (300
MHz, DMSO-d6) 8 8.56 (s, 1H), 7.79 (d, J - 7.4 Hz, 1H), 7.68 (d, J = 7.5 Hz, 1H), 7.59 (s,
1H), 7.46-7.19 (m, 6H), 5.23 (s, 2H), 5.14 (s, 2H), 2.36 (s, 3H), 2.03 (s, 3H).
Example 30: Methyl 5-(1H-benz»midazo)-1-yl)-3-{[(trifluoromethyl)su1fonvl]oxy}-
thtophene-2-carboxylate
Methyl 5-(1H-benzimidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate (0.275 g, 1.00
mmol) was dissolved in 7 mLof dichloromethane with stirring. N,N-Diisopropylethyl-
amine (0.230 mL, 1.32 mmol) was added via syringe. N-Phenyltrifluoromethane-
sulfonamide (0.429 g, 1.20 mmol) was added in a single portion. After 18 hours, the
reaction was poured into dichloromethane and brine. The layers were separated, and
the aqueous washed with dichloromethane. The combined organic layers were dried
over MgSO4, filtered, and concentrated in vacuo. Purification by flash
chromatography afforded 0.406 g (100%) of methyl 5-(1H-benzimidazol-1-yl)-3-
{[(trifluoromethyl)-sulfonyl]oxy}-thiophene-2-carboxylate as a white solid. 1H NMR
(300 MHz, DMSO-d6) 8 8.77 (s, 1H), 7.88 (s, 1H), 7.85 (d, J - 8.4 Hz, 1H), 7.83 (d, J = 8.5
Hz, 1H), 7.49-7.38 (m, 2H), 3.91 (s, 3H).
Example 31: Methyl 3-anilino-5-(1H-benzimidazol-1-Yl)thiophene-2-carboxylate
Methyl 5-(1H-benzimidazol-1 -yl)-3-{t(trifluoromethyl)sulfonyl]oxy}-thlophene-2-
carbbxylate (0.200 g, 0.492 mmol), cesium carbonate (0.224 g, 0.687 mmol), rao-2,21-
bis(diphenylphosphino)-1,1"-binaphthyl (0.0306 g, 0.0490 mmol). and
tris(dibenzylidene-acetone)dipalladium(O) (0.0225 g, 0.0250 mmol) were combined in
flask equipped with a reflux condenser. 5 mL of toluene was added followed by
aniline (0.0540 mL, 0.593 mmol). The mixture was heated to 110 °C and maintained at
that temperature for 18 hours. The mixture was cooled to room temperature and
adsorbed onto silica gel. Purification by flash chromatography afforded 0.138 g (80%)
of methyl 3-anilino-5-(1H-benzimidazol-1-yl)thiophene-2-carboxylate as an off-
white solid. 1H NMR (300 MHz. DMSO-d6) 8 9.01 (s, 1H), 8.77 (s, 1H), 7.84 (d,J= 7.7
Hz, 1H), 7.79 (d, J m 7.6 Hz, 1H), 7.51 (s, 1H), 7.45-7.33 (m, 6H), 7.08 (m, 1H), 3.84 (s,
3H).
Example 32: Methyl 5-(1//-benzimidazol-1-yl)-3-(benzovlamino)thiophene-2-
carboxylate
Methyl 5-(1H-benzimidazol-1-yI)-3-{[(trifluoromethyl)sulfonyl]oxy}-thiophene-2-
carboxylate (0.350 g, 0.861 mmol), cesium carbonate (0.393 g, 1.21 mmol), rac-2,21-
bis(diphenylphosphino)-1,1"-binaphthyl (0.0536 g, 0.0860 mmol), and
tris(dibenzylidene-acetone)dipalladium(0) (0.0394 g, 6.0430 mmol) were combined in
flask equipped with a reflux condenser. 12 mL of toluene was added followed by
benzamide (0.125 g, 1.03 mmol). The mixture was heated to 100 °C and maintained at
that temperature for 40 hours. The mixture was cooled to room temperature and
adsorbed onto silica gel. Purification by flash chromatography afforded 0.282 g (87%)
of methyl 5-(1H-benzimidazol-1-yl)-3-(benzoylamino)thiophene-2-carboxylate as a
white solid. 1H NMR (300 MHz, DMSO-d6) 5 11.11 (s, 1H), 8.81 (s, 1H), 8.40 (s, 1H),
8.00 (m, 2H), 7.83 (m, 2H), 7.72-7.60 (m, 3H), 7.50-7.38 (m. 2H), 3.93 (s, 3H). MS (m/z)
378 (m+1).
Example 33: 5-(1 H-Benzimidazol-1-yl)-3-(benzoylamino)thiophene-2-carboxylic acid
Methyl 5-(1H-benzimidazol-1-yl)-3-(benzoylamino)thiophene-2-carboxylate (0.275 g,
0.729 mmol) was dissolved in 15 mL of dioxane with stirring. 15 mL of 1M LiOH
solution was added, and the mixture was stirred for 16 hours at room temperature. 15
mL of 2M HCI solution was added slowly via pipet, resulting in the formation of a
solid. The mixture was filtered and the solid was washed with diethyl ether. The solid
was collected and dried under high vacuum to yield 0.0963 g (36%) of 5-(1H-
benzimidazol-1-yl)-3-(benzoylamino)thiophene-2-carboxylic acid as a tan solid. "H
NMR (300 MHz, DMSO-d6) d 11.31 (s, 1H), 8.79 (s, 1H), 8.39 (s, 1H), 7.97 (m, 2H). 7:83
(m, 2H), 7.73-7.60 (mr3H), 7.50-7.36 (m, 2H).
Example 34: 5-{5-Chloro-1 H-benzimidazol-1-yl)l-3-[((2-methylbenzyl)oxvlthiophene-
2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5-chloro-
1 H-benzimidazoi-1-yl)-3-l(2-methylbenzyl)oxy]thiophene-2-carboxylate (0.323 g,
0.782 mmol) provided 0.253 g (81%) of 5-(5-chloro-1 H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylic acid as a pale yellow solid. 1H NMR (300
MHz, DMSO-d6) 5 12.81 (br s, 1H), 8.77 (s, 1H), 7.90 (d, J= 1.9 Hz, 1H), 7.85 (d, J- 8.7
Hz, 1H), 7.72 (s, 1H), 7.54-7.44 (m, 2), 7.28-7.20 (m, 3H), 5.33 (s. 2H), 2.38 (s, 3H).
Example 35: 5-(6-Chloro-1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-
2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(6-chloro-
1 W-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxylate (0.176 g,
0.426 mmol) provided 0.150 g (88%) of 5-(6-chloro-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylic acid as a pale yellow solid. "H NMR (300
MHz, DMSO-d6) 812.81 (s, 1H), 8.71 (s, 1H), 7.82 (m, 2H), 7.72 (s, 1H), 7.54 (m, 1H),
7.40 (dd, J = 8.7,1.8 Hz, 1H), 7.29-7.21 (m, 3H), 5.35 (s, 2H), 2.39 (s, 3H).
Example 36: 5-(5-Chloro-1 H-benzimidazol-1-yl)-N-methoxy-N-methyl-3-[(2-
methvlbenzvl)oxv]thiophene-2-carboxamide
5-(5-Chloro-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxylic
acid (0.100 g, 0.251 mmol). N,O-dimethylhydroxylamine hydrochloride (0.0490 g,
0.502 mmol), and 4-dimethylaminopyridlne (0.0062 g, 0.051 mmol) were dissolved in 5
mL of dichloromethane. Triethylamine (0.077 mL, 0.550 mmol) was added via syringe,
followed by the addition of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
hydrochloride (0.0870 g, 0.454 mmol) in a single portion. The reaction was stirred for
65 hours and poured into ethyl acetate and water. The layers were separated, and the
organic layer was washed with brine. The combined aqueous layers were extracted
with ethyl acetate, and the combined organic layers were dried over MgSO4
Filtration, concentration in vacuo, and purification by flash chromatography afforded
0.0772 g (70%) of 5-(5-chloro-1H-benzimidazol-1-yl)-N-methoxy-N-methyl-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxamide as an oil which solidified upon standing.
1H NMR (300 MHz, DMSO-d6) d 8.76 (s, 1H), 7.90 (d, J = 2.0 Hz, 1H), 7.84 (d, J- 8.8
Hz, 1H), 7.71 (s, 1H), 7.55 (d, J= 7.4 Hz, 1H), 7.46 (dd, J = 8.8 Hz, 2.0 Hz, 1H), 7.29-7.20
(m, 3H), 5.30 (s, 2H), 3.69 (s, 3H), 3.21 (s, 3H), 2.37 (s, 3H).
Example 37: 5-(6-Chloro-1H-benzimidazol-1-yl)-N-methoxv-/V-methyl-3-[(2-
methylbenzvl)oxy]thiophene-2-carboxamide
An analogous procedure to that described in Example 36 with 5-(6-chloro-1H-
benzimidazol-1-yl)-3-[(2-methYlbenzyl)oxy]thiophene-2-carboxyiic acid (0.0430"g,
0.108 mmol) afforded 0.0423 g (89%) of 5-(6-chloro-1H-benzimidazol-1-yl)-N-
methoxy-N-methyl-3-[(2-methylbenzyl)oxy]thiophene-2-carboxamide as an oil which
solidified upon standing. "H NMR (300 MHz, DMSO-d6) 8 8.70 (s, 1H), 7.82 (d, J= 8.8
Hz, 1H), 7.80 (s, 1H), 7.71 (s, 1H), 7.56 (d, J - 7.3 Hz, 1H), 7.41 (dd, J - 8.5,2.1 Hz, 1H),
7.29-7.20 (m, 3H), 5.32 (s. 2H), 3.68 (s, 3H), 3.32 (s, 3H), 2.38 (s, 3H).
Example 38: t-{5-(5-Chloro-1 H-benzimidazol-1-yl)-3-(2-methylbenzyl)oxy3thten-2-
yllethanorie
5-(5-Chloro-1H-benzimidazol-1-yl)-N-methoxy-N-methyl-3-[(2-methylbenzyl)-
oxy]thiophene-2-carboxamide (0.0750 g, 0.170 mmol) was dissolved in 5 mLof
tetrahydrofuran and cooled to -78 °C. Methyl magnesium bromide (0.170 mL, 3.0 M
in diethyl ether, 0.510 mmol) was added dropwise via syringe. After 5 minutes, the
reaction was warmed to 0 °C, where it was maintained for an additional 30 minutes.
The reaction was quenched by the dropwise addition of 2 mL of 5% HCI. The mixture
was poured into ethyl acetate and brine, and the layers were separated. The aqueous
layer was extracted with ethyl acetate, and the combined organic layers were dried
over MgS(k Filtration, concentration in vacuo, and purification by flash
chromatography provided 0.0658 g (98%) of 1-{5-(5-chloro-1H-benzimidazol-1-yl)-
3-[(2-methy!benzy!)oxy]thien-2-yl}ethanone as a bright yellow solid. 1H NMR (300
MHz, DMSO-d6) 8 8.83 (s, 1H), 7.91 (m, 2H), 7.79 (s, 1H), 7.53 (m, 1H), 7.49 (dd, J= 8.8,
2.1 Hz, 1H), 7.29 (s, 1H), 7.28 (m. 2H), 5.43 (s, 2H), 2.46 (s, 3H), 2.41 (s, 3H).
Example 39: 1-{5-(6-Chloro-1 tf-benzimidazol-1-yl)-3-[(2-methvlbenzyl)oxv]thien-2-
yljethanone
An analogous procedure to that described in Example 38 with 5-(6-Chloro-1 H-
benzimidazol-1-yl)-N-methoxy-N-methyl-3-[(2-methylbenzyl)-oxy]thiophene-2-
carboxamide (0.0400 g, 0.0905 mmol) provided 0.0320 g (89%) of 1-{5-(6-chloro-1 H-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thien-2-yl}ethanone as a yellow solid. 1H
NMR (300 MHz, DMSO-d6) 5 8.77 (s, 1H), 7.89 (d, J - 1.7 Hz, 1H),7.83 (d.J=8.6 Hz,
1H), 7.78 (s, 1H), 7.55 (d, J - 6.6 Hz, 1H), 7.43 (dd, J = 8.6 Hz, 1.9 Hz. 1H), 7.33-7.25 (m,
3H), 5.45 (s, 2H), 2.46 (s, 3H), 2.41 (s, 3H).
Example 40: Methyl 5-(5-fluoro-1H-benzimidazol-1-yl)-3-hvdroxythiophene-2-
carboxylate and Methyl 5-(6-fluoro-1/7-benzimidazol-1-yl)-3-hvdroxythiophene-2-
carboxylate
Methyl 2-chloro-3-oxo-2,3-dihydro-2-thiophenecarboxylate (0.250 g, 1.30 mmol) was
dissolved in 15 mL of chloroform with stirring. 5-fluorobenzimidazole (0.389 g, 2.86
mmol) was added, and the mixture was allowed to stir for 65 hours. The reaction was
poured into half-saturated NaCI and dichloromethane. The layers were separated, and
the aqueous layer was extracted twice with dichloromethane. The combined organic
layers were dried over MgSO4, filtered, and concentrated in vacuo. Purification by
flash chromatography afforded 0.267 g (70%) of a 1:1 regioisomeric mixture of
methyl 5-(5-fluoro-1 H-benzimidazol-1-yl)-3-1iydroxythiophene-2-carboxylate and
methyl 5-(6-fluoro-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate as a
tan solid. 1H NMR (300 MHz, DMSO-d6) 8 10.90,10.87 (2xs, 1H), 8.75,8.68 (2xs, 1H),
7.84-7.79 (m, 1H), 7.66-7.59 (m, 1H), 7.32-7.20 (m, 1H), 7.15 (s, 1H), 3.79 (s, 3H).
Example 41: Methyl a-hydroxv-5-(5-methoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxylate and Methyl 3-hvdroxy-5-(6-methoxv-1H-benzimidazol-1-yl)thiophene-2-
carboxylate
An analogous procedure to that described in Example 40 with 5-
methoxybenzimidazole (0.424 g, 2.86 mmol) provided 0.260 g (66%) of a 1:1
regioisomeric mixture of methyl 5-(5-methoxy-1H-benzimidazol-1-yl)-3-
hydroxythiophene-2-carboxylate and methyl 5-(6-methoxy-1 H-benzimidazol-1-yl)-3-
hydroxythiophene-2-carboxylate as a tan solid. "H NMR (300 MHz, DMSO-d6) d10.85
(s, 1H). 8.63, &52 (2xs, 1H). 7.70, 7.67 (2xd, J - 8.0 Hz, 1 H), 7.33,7.23 (2xd, J = 2.4 Hz.
1H), 7.14,7.11 (2xs, 1H), 7.03, 6.97 (2xdd, J = 9.0,2.4 Hz, 1H), 3.84.3.82,3.79,3.78 (4 x
s, 12H).
Example 42: Methyl 5-(5-bromo-1H-benzimidazol-1-vl)-3-hydroxythiophene-2-
carboxylate and Methyl 5-(6-brorno-1f/-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate
An analogous procedure to that described in Example 40 with 5-bromobenzimidazole
(2,20 g, 11.2 mmol) provided 1.03 g (53%) of a 1:1 regioisomeric mixture of methyl 5-
(5-bromo-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate and methyl 5-
(6-bromo-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate as a tan solid.
1H NMR (300 MHz, DMSO-d6) 6 10.90 (s, 1H), 8.74, 8.70 (2xs, 1H), 8.02.7.93 (2xd, J =
1.8 Hz, 1H), 7.77 (m, 1H), 7.54 (m, 1H), 7.17, 7.15 (2xs, 1H), 3.79 (s, 3H).
Example 43: Methyl 5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-hydroxvthiophene-2-
carboxylate
An analogous procedure to that described in Example 40 with 5,6-
dichlorobenzimidazole (2.15 g, 11.5 mmol) provided 0.359 g (18%) of methyl 5-(5,6-
dichloro-1//-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate as a tan solid. 1H
NMR (300 MHz, DMSO-d6) 8 10.90 (s, 1H), 8.78 (s, 1H), 8.12 (s, 1H), 8.02 (s, 1H), 7.18 (s,
1H),3.79(s,3H).
Example 44: Methyl 5-(5.6-dimethoxv-1H-benzimidazol-1-yl)-3-hydroxythiophene-
2-carboxylate
An analogous procedure to that described in Example 40 with 5,6-dimethoxy-
benzimidazole (2.00 g, 11.22 mmol) provided 0.632 g (34%) of methyl 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate as a tan solid.
1H NMR (300 MHz, DMSO-d6) 6 10.81 (s, 1H), 8.46 (s, 1H), 7.34 (s, 1H), 7.24 (s, 1H), 7.13
(s, 1H), 3.85 (s, 3H). 3.82 (s, 3H), 3.79 (s, 3H).
Example 45: Methyl 5-(5.6-dichloro-1H-benzimidazol-1-yl)-3-[(2-
methvlbenzvl)oxv]thiophene-2-carboxylate
Methyl 5-(5,6-dichloro-1 H-benzimidazol-1 -yI)-3-hydroxythiophene-2-carboxylate
(0.0900 g, 0.262 mmol) was dissolved in 5 mL of N,N-dimethylformamide with stirring.
Solid potassium carbonate (0.0430 g, 0.311 mmol) was added in a single portion. 2-
Methylbenzyl bromide (0.042 mL, 0.31 mmol) was added via syringe. The reaction was
stirred for 65 hours and poured into ethyl acetate and water. The layers were
separated, and the organic layer was washed with brine. The combined aqueous layers
were extracted with ethyl acetate, and the combined organic layers were dried over
MgSO4 The solution was filtered, concentrated in vacuo, and purified by flash
chromatography to afford 0.107 g (91%) of methyl 5-(5,6-dichloro-1/7-benzimidazol-
1-yl)-3-[(2-methyibenzyl)oxy]thiophene-2-carboxylate as an off-white solid. 1H NMR
(300 M Hz, DMSO-d6) 8 8.80 (s, 1H), 8.14 (s, 1H), 8.05 (s, 1 H)f 7.79 (s, 1H), 7.55 (d, J -
7.5 Hz, 1H), 7.28-7.24 (m, 3H), 5.38 (s, 2H), 3.77 (s, 3H). 2.39 (s, 3H).
Example 46: Methyl 5-(5-fluoro-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)-
oxy]thiophene-2-carboxvlate and Methyl 5-(6-fluoro-1H-benzimidazol-1-yl)-3-[(2-
methvlbenzvl)oxy]thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with a 1:1 regioisomeric
mixture of methyl 5-(5-fluoro-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate and methyl 5-(6-fluoro-1 H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate (0.262 g , 0.896 mmol) provided 0,291 g (82%) of a 1:1 regioisomeric
mixture of methyl 5-(5-fluoro-1 H-benzimidazol-l-yl)-3-[(2-methylbenzyl)-
oxy]thiophene-2-carboxylate and methyl 5-(6-fluoro-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylate as an off-white solid. 1H NMR (300 MHz,
DMSO-d6) 8 8.78, 8.71 (2xs, 1H), 7.95-7.50 (m, 5H), 7.35-7.22 (m, 3H). 5.39, 5.37 (2xs,
2H), 3.77 (s, 3H), 2.39 (s, 3H).
Example 47: Methyl 5-(5-methoxv-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-
thiophene-2-carboxylate and Methyl 5-(6-methoxy-1H-benzimidazolr-1-yl)-3-((2-
methvlbenzyl)oxv]thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with a 1:1 regioisomeric
mixture of methyl 5-(5-methoxy-1 W-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate and methyl 5-(6-methoxy-1 W-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate (0.255 g, 0.838 mmol) gave 0.249 g (73%) of a 1:1 regioisomeric mixture
of methyl 5-(5-methoxy-1 W-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]-thiophene-
2-carboxylate and methyl 5-(6-methoxy-1 W-benzimidazol-1 -yI)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylate as an off-white solid. 1H NMR (300 MHz,
DMSO-ds) 8 8.67. 8.55 (2xs, 1H), 7.95,7.76-7.67, 7.56-7.53 (m, 3H), 7.34, 7.30-7.21,
7.07-6.97 (m, 5H), 5.38, 5.37 (2xs, 2H), 3.84,3.83, 3.77,3.76 (4 x s, 12H), 2.39 (s, 3H).
Example 48: Methyl 5-(5-bromo-1H-benzimidazol-1-vl)-3-[(2-methylbenzyl)oxy]-
thiophene-2-carboxylate and Methyl 5-(6-bromo-1H-benzimidazol-1-yl)-3-[(2-
methvlbenzyl)oxyl-thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with a 1:1 regioisomeric
mixture of methyl 5-(5-bromo-1 H-benzimidazol-l~yl)-3-hydroxythiophene-2-
earboxylate and methyl 5-(6-bromo-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate (0.750 g . 2.12 mmol) provided 0.681 g (70%) of a 1:1 regioisomeric
mixture of methyl 5-(5-bromo-1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)-
oxy]thiophene-2-carboxylate and methyl 5-(6-bromo-1H-benzimidazoi-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylate as an off-white solid. 1H NMR (300 MHz,
DMSO-d6) 6 8.77. 8.71 (2xs, 1H), 8.04,7.95 (2xd, J - 1.8 Hz. 1H). 7.83-7.75. 7.60-7.52,
7.27-7.11(m, 7H), 5.38, 5.37 (2xs, 2H), 3.77 (s, 3H). 2.40, 2.39 (s, 3H).
Example 49: Methyl 5-(6-chloro-1H-benzimidazol-1 -yl)-3-[(2-chloro-4-
fluorobenzvl)oxy]thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(6-chloro-
1 W-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.100 g, 0.324 mmol) and
2-chloro-4-fluorobenzyl bromide (0.0869 g, 0.389 mmol) provided 0.131 g (90%) of
methyl 5-(6-chloro-1Hbenzimidazol-1-yl)-3-[(2-chloro-4-fluorobenzyl)oxy]-
thiophene-2-carboxylate as an off-white solid. 1H NMR (300 MHz, DMSO-d6) 5 8.75
(s, 1H), 7.89 (d, J = 1.9 Hz, 1H). 7.84-7.78 (m, 2H), 7.78 (s, 1H), 7.56 (dd, J = 8.8,2.7 Hz,
1H), 7.42 (dd, J = 8.6,1.9 Hz. 1H), 7.35 (ddd, J - 8.7.8.7. 2.7 Hz, 1H), 5.42 (s. 2H). 3.78
(s,3H).
Example 50: Methyl 5-(6-chloro-1H-benzimidazol-1-yl)-3-[(2.4-difluorobenzvl)oxy]-
thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(6-chloro-
1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.100 g, 0.324 mmol) and
2,4-difluorobenzy! bromide (0.054 mL, 0.39 mmol} provided 0.122 g (87%) of methyl
5-(6-chloro-1W-benzimidazol-1-yl)-3-[(2,4-difluorobenzyl)oxy]thiopherie-2-
carboxylate as an off-white solid. 1H NMR (300 MHz, DMSO-d6) 8 8.74 (s, 1H), 7.89 (d,
J = 1.9 Hz, 1H). 7.83 (d, J = 8.6 Hz, 1H), 7.77-7.69 (m, 1H), 7.76 (s, 1H), 7.42 (dd, J = 8.6,
1.9 Hz, 1H), 7.35 (m, 1H), 7.19 (m, 1H), 5.41 (s, 2H), 3.77 (s, 3H).
Example 51: Methyl 5-(6-chloro-1H-benzimidazo1-1-yl)-3-(pyridin-3-
ylmethoxv)thiophene-2-carboxylate
An analogous, procedure to that described in Example 45 with methyl 5-(6-chloro-
1 H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.100 g, 0.324), 3-
(bromomethyl)pyridine hydrobromide (0.0980g, 0.387 mmol), and potassium
carbonate (0.107 g, 0.774 mmol) yielded 0.0393 g (30%) of methyl 5-(6-chloro-1 H-
benzimidazol-1-yl)-3-(pyridin-3-ylmethoxy)thiophene-2-carboxy)ate as a tan solid.
1H NMR (300 MHz, DMSO-de) 8 8.72 (s, 1H), 8.72 (m, 1H), 8.58 (dd. J - 4.8,1.5 Hz, 1H),
7.93 (m, 1H), 7.86 (d,J = 1.9 Hz, 1H), 7.83 (d, J= 8.7 Hz, 1H), 7.73 (s, 1H), 7.48 (m, 1H),
7.42 (dd, J m 8.7,1.9 Hz, 1H), 5.45 (s, 2H), 3.79 (s, 3H).
Example 52: Methyl 5-(1H-benzimidazol-1-yl)-3-(prop-2-vnvloxy)thiophene-2-
carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(1 H-
benzimidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate (0.250 g, 0.911 mmol) and
propargyl bromide (0.12 mL, 80% in toluene, 1.08 mmol) afforded 0.211 g (74%) of
methyl 5-(1H-benzimidazol-1-yl)-3-(prop-2-ynyloxy)thiophene-2-carboxyIate as a
tan solid. 1H NMR (300 MHz, DMSO-d6) 5 8.70 (s, 1H), 7.88 (d, J - 7.7 Hz, 1H), 7.81 (d, J
- 7.5 Hz, 1H), 7.61 (s, 1H), 7.49-7.36 (m, 2H), 5.07 (d, J = 2.3 Hz, 2H), 3.78 (s, 3H), 3.73
(t, J = 2.3 Hz, 1H). MS (m/z) 313 (m+1).
Example 53: Methyl 5-(5-bromo-1 H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzyl]oxv)thiophene-2-carboxylate and Methyl 5-(6-bromo-1 H-benzimidazol-1 -yl)-
3-{[2-(trifluoromethyl)benzyl3oxy}thiophene-2-carboxvlate
An analogous procedure to that described in Example 45 with a 1:1 regioisomeric
mixture of methyl 5-(5-bromo-1 H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate and methyl 5-(6-bromo-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxylate (0.200 g, 0.566 mmol) and 2-trifluoromethylbenzyl bromide (0.163 g,
0.682 mmol) provided a 1:1 regioisomeric mixture of products. The mixture was
separated by flash chromatography to afford 0.0952 g (33%) of methyl 5-(5-bromo-
1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-benzyl]oxy}thiophene-2-carboxy]ate
as an off-white solid: 1H NMR (300 MHz, DMSO-ds) 8 8.79 (s, 1H), 8.04 (d, J = 1.8 Hz,
1H), 7.97 (d, J - 7.6 Hz, 1H). 7.85-7.77 (m, 2H). 7.75 (s, 1H), 7.62 (m, 1H), 7.60 (d, J -
1.9 Hz, 1H). 7.58 (d, J= 1.8 Hz, 1H), 5.50 (s, 2H), 3.78 (s, 3H), and 0.0970 g (34%) of
methyl 5-(6-bromo-1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzyl]oxy}thiophene-2-carboxylate as an off-white solid: "H NMR (300 MHz, DMSO-
ds) 8 8.73 (s, 1H), 7.99-7.94 (m, 2H), 7.85-7.71 (m, 4H). 7.62 (m, 1H), 7.53 (dd, J - 8.6,
1.5 Hz, 1H), 5.52 (s, 2H), 3.78 (s, 3H).
Example 54: Methyl 5-(5.6-dimethoxv-1H-benzimidazol-1-yl)-3-[(2-trifluoromethyl-
benzyl)oxy]thiophene-2-earboxylate
An analogous procedure to that described in Example 45 with methyl 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.108 g, 0.323
mmol} and 2-trifluoromethylbenzyl bromide (0.232 g, 0.971 mmol) afforded 0.109 g
(69%) of methyl 5-(5,6-dimethoxy-1 H-benzimidazol-1 -yl)-3-[(2-
trifluoromethylbenzyl)-oxy]thiophene-2-carboxylate as an off-white solid. "H NMR
(300 MHz, DMSO-d6) 8 8.50 (s, 1H), 7.96 (d, J - 7.5 Hz, 1H), 7.84-7.76 (m, 2H), 7.66 (s,
1H), 7.61 (dd, J - 7.7,7.7 Hz, 1H), 7.35 (s, 1H), 7.26 (s, 1H). 5.53 (s, 2H), 3.84 (s, 3H),
3.83 (s. 3H), 3.78 (s, 3H).
Example 55: Methyl 3-[(2,6-dichlorobenzyl)oxvI-5-(5,6-dimethoxy-1H-benzimidazol-
1-yl)thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.100 g, 0.299
mmol) and 2,6-dichlorobenzyl bromide (0.0869 g, 0.362 mmol) provided 0.117 g (79%)
of methyl 3-[(2,6-dichlorobenzyl)oxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1-
yl)thiophene-2-carboxylate as an off-white solid. 1H NMR (300 MHz, DMSO-ds) 8 8.52
(s, 1H), 7.78 (s, 1H), 7.62 (d, J - 1.5 Hz, 1H), 7.59 (s. 1H), 7.51 (dd, J - 9.3, 6.8 Hz, 1H),
7.35 (s, 1H), 7.31 (s, 1H), 5.52 (s, 2H), 3.87 (s, 3H), 3.83 (s, 3H), 3.71 (s, 3H).
Example 56: Methyl3-[(2-bromobertzyl)oxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1-
yl)thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(5,6-
dimethoxy-1 H-benzimidazpl-1-yl)-3-hydroxythiophene-2-carboxylate (0.100 g, 0.299
mmol) and 2-bromobenzyl bromide (0.0905 g, 0.362 mmol) provided 0.114 g (76%) of
methyl 3-[(2-bromobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-
2-carboxylate as an off-white solid. 1H NMR (300 MHz, DMS0-d6) 8 8.51 (s, 1H), 7.73
(ddd, J= 7.6, 7.6,1.0 Hz, 1H), 7.68 (m. 1H), 7.68 (s, 1H), 7.49 (ddd, .7 = 7.6,7.6,1.2 Hz,
1H), 7.35 (s, 1H), 7.34 (ddd, J - 7.6,7.6,1.6 Hz, 1H), 7.26 (s, 1H), 5.40 (s, 2H), 3.84 (s,
3H), 3.83 (s, 3H), 3.79 (s, 3H).
Example 57: Methyl 5-(5,6-dichloro-1tf-benzimidazol-1-yl)-3-(3-furylmethoxy)-
thiophene-2-carboxylate
Methyl 5-(5,6-dichloro-1 H-benzimidazol-1 -yl)-3-hydroxythiophene-2-carboxylate
(0.0900 g, 0.262 mmol) and triphenylphosphine (0.0890 g, 0.339 mmol) were dissolved
in 4 mL of tetrahydrofuran with stirring. The reaction was cooled to 0 °C, and 3-
furanmethanol (0.030 mL, 0.35 mmol) was added via syringe. Diethyl azodicarboxylate
(0.053 mL, 0.34 mmol) was added dropwise via syringe. The reaction was warmed to
room temperature and stirred for 3 hours. The mixture was adsorbed onto silica gel
and purification by flash chromatography afforded 0.0725 g (65%) of methyl 5-(5,6-
dichloro-1H-benzimidazol-1-yl)-3-(3-furylmethoxy)-thiophene-2-carboxylate as an
inseparable mixture with diethyl hydrazine-1,2-dicarboxylate, which could be easily
removed in the workup of the following reaction. 1H NMR (300 MHz, DMSO-d6) 8 8.79
(s. 1H),8.14(s, 1H),8.07 (s, 1H),7.85 (dd,J= 1.6,0.9 Hz, 1H), 7.72 (s, 1H),7.70(dd, J =
1.6,1.6 Hz. 1H), 6.61 (dd, J - 1.9,0.8 Hz, 1H), 5.25 (s, 2H), 3.77 (s, 3H).
Example 58: Methyl 5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-(2-furylmethoxy)-
thiophene-2-carboxylate
An analogous procedure to that described in Example 57 with methyl 5-(5,6-
dichloro-1 H-benzimfdazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.0900 g, 0.262
mmol) and furfuryl alcohol (0.029 mL, 0.34 mmol) provided 0.0525 g (47%) of methyl
5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-(2-furylmethoxy)-thiophene-2-carboxylate
as an inseparable mixture with diethyl hydrazine-1,2-dicarboxylate, which could be
easily removed in the workup of the following reaction. 1H NMR (300 MHz, DMSO-d6)
d8.79 (s, 1H),8.14 (s, 1H), 8.09 (s, 1H), 7.76(s, 1H), 7.75 (dd, J = 1.9.0.8 Hz, 1H), 6.71
(dd, J = 3.2,0.8 Hz, 1H), 6.51 (dd. J - 3.2,1.9 Hz, 1H), 5.36 (s. 2H). 3.75 (s. 3H).
Example 59: Methyl 5-(5.6-dichloro-1H-benzimidazol-1-yl)-3-(thien-3-ylmethoxy)-
thiophene-2-carboxylate
An analogous procedure to that described in Example 57 with methyl 5-(5,6-
dichloro-1 H-benzimidazol-1 -yl)-3-hydroxythiophene-2-carboxylate (0.0900 g, 0.262
mmol) and 3-thiophenemethanol (0.032 mL.0.34 mmol) provided 0.0745 g (65%) of
methyl 5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-(thien-3-ylmethoxy)-thiophene-2-
carboxylate as an inseparable mixture with diethyl hydrazine-1,2-dicarboxylate, which
could be easily removed in the workup of the following reaction. "H NMR (300 MHz,
DMSO-d6) 8 8.78 (s, 1H), 8.14 (s, 1H), 8.04 (s, 1H), 7.71 (s, 1H), 7.66 (m, 1H), 7.60 (dd, J
=5.0,2.9 Hz, 1H), 7.22 (dd, J= 5.0,1.2 Hz, 1H), 5.38 (s, 2H), 3.78 (s, 3H).
Example 60: Methyl 5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-(thien-2-vlmethoxy)-
thiophene-2-carboxylate
An analogous procedure to that described in Example 57 with methyl 5-(5,6-
dichloro-1H-benzimidazol-1-yl)-3-hydroxythiophene-2-carboxylate (0.0775 g, 0.226
mmol) and 2-thiophenemethanol (0.028 mL, 0.30 mmol) provided 0.0599 g (60%) of
methyl 5-(5,6-dichloro-1H-benzimidazol-1-yl)-3-(thien-2-ylmethoxy)-thiophene-2-
carboxylate as an inseparable mixture with diethyl hydrazine-1,2-dicarboxylate, which
could be easily removed in the workup of the following reaction. 1H NMR (300 MHz,
DMSO-de) 8 8.78 (s, 1H), 8.14 (s, 1H), 8.05 (s, 1H). 7.75 (s, 1H), 7.61 (dd, J - 5.0,1.2 Hz,
1H), 7.30 (dd, J - 3.5,1.2 Hz, 1H), 7.07 (dd, J = 5.0.3;5 Hz, 1H), 5.57 (s, 2H), 3.77 (s,
3H).
Example 61: 5-(6-Chloro-1H-benzimidazol-1-yl)-3-[(2-methvlbenzyl)oxy]thiophene-
2-carboxamide
Methyl 5-(6-chloro-1 H-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thiophene-2-
carboxylate (0.172 g, 0.417 mmol) was placed in sealed tube. Ammonia in methanol
(15.0 mL. 2.0 M in MeOH, 30 mmol) was added, and the vessel was sealed. The tube
was placed in an oil bath preheated to 80 °C, and stirred at that temperature for 24
hours. The reaction was cooled to room temperature, and an additional 15.0 mL of
the ammonia in methanol solution was added. The vessel was resealed and heating
continued for an additional 44 hours. The reaction was cooled to room temperature
and adsorbed onto silica gel. Purification by flash chromatography provided 0.0417 g
(24%) of recovered starting material and 0.0820 g (49%) of 5-(6-chloro-1 H-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxamide as a yellow
solid. 1H NMR (300 MHz, DMSO-d6) 6 8.68 (s, 1H), 7.82 (d, J = 8.8 Hz, 1H), 7.78 (d, J - .
2.1 Hz, 1H), 7.72 (br s, 1H), 7.70 (s, 1H), 7.51 (d, J = 7.0 Hz, 1H), 7.40 (dd, J = 8.6, 2.1
Hz, 1H), 7.34-7.21 (m, 3H), 6.88 (br s, 1H), 5.44 (s, 2H), 2.40 (s, 3H).
Example 62: 5-(6-Bromo-1H-benzimidazol-1-vl)-3-{[2-(trifluoromethyl)benzyl]-
oxy}thiophene-2-carboxamide
An analogous procedure to that described in Example 61 with methyl 5-(6-bromo-
1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyI)benzyl]oxy}thiophene-2-carboxylate
,(0,0950 g, 0.186 mmol) afforded 0.0557 g (60%) of 5-(6-bromo-1H-benzimidazol-1-
yl)-3-{[2-(trifluoromethyl)benzyl]-oxy}thiophene-2-carboxamide as an off-white
solid. 1H NMR (300 MHz, DMSO-d6) 5 8.67 (s, 1H), 7.91 (d, J = 1.6 Hz, 1H), 7.89-7.71
(m, 5H), 7.68 (s, 1H), 7.67 (m, 1H), 7.52 (dd, J - 8.6,1.8 Hz, 1H), 6.81 (br s, 1H), 5.56 (s,
2H).
Example 63: 5-(5.6-Dimethoxv-1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzyl3oxv}thiophene-2-carboxamide
An analogous procedure to that described in Example 61 with methyl 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-[(2-trifiuoromethylbenzyl)oxy]thiophene-2-
carboxylate provided 0.0351 g (34%) of 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-
{[2-(trifluoromethyl)-benzyl]oxy}thiophene-2-carboxamide as a light tan solid. 1H
NMR (300 MHz, DMSO-d6) d 8.43 (s, 1H), 7.90-7.58 (m, 5H), 7.60 (s, 1H), 7.34 (s, 1H),
7.21 (s, 1H), 6.82 (br s, 1H), 5.56 (s, 2H).
Example 64: 3-[(2,6-Dichlorobenzvl)oxy]-5-(5.6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide
An analogous procedure to that described in Example 61 with methyl 3-[(2,6-
dichlorobenzyl)oxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1 -yl)thiophene-2-
carboxylate (0.115 g. 0.233 mmol) afforded 0.0392 g (35%) of 3-[(2,6-
dichlorobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide as an off-white solid. 1H NMR (300 MHz, DMSO-ds) 8 8.46 (s, 1H), 7.79
(s, 1H), 7.68 (br s, 1H), 7.63 (d, J = 1.5 Hz, 1H), 7.60 (s, 1H), 7.52 (dd. J = 9.1, 6.9 Hz,
1H), 7.35 (s, 1H), 7.30 (s, 1H). R63 (br s, 1H), 5.58 (s. 2H), 3.87 (s, 3H), 3.83 (s, 3H).
Example 65: 3-[(2-Bromobenzyl)oxv]-5-(5.6-dimethoxv-1 H-benzimidazol-1-
yl)thiophene-2-carboxamide
An analogous procedure to that described in Example 61 with methyl 3-[(2-
bromobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-carboxylate
(0.112 g, 0.222 mmol) afforded 0.0296 g (27%) of 3-[(2-bromobenzyl)oxy]-5-(5,6-
dimethoxy-1 H-benzimidazol-1-yl)thiophene-2-carboxamide as a yellow solid. 1H
NMR (300 MHz, DMSO-ds) 8 8.43 (s, 1H), 7.78-7.64 (m, 3H), 7.66 (s, 1H), 7.47 (m, 1H).
7.86 (m. 1H), 7.34 (s, 1H), 7.21 (s, 1H), 6.91 (br s, 1H). 5.46 (s, 2H).
Example 66: 5-(5,6-Dichloro-1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]-
thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dichloro-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxylate
(0.105 g, 0.235 mmol) yielded 0.0695 g (68%) of 5-(5,6-dichloro-1 H-benzimidazol-1-
yl)~3-[(2-methylbenzyl)oxy]thiophene-2-carboxylic acid as a light tan solid. 1H NMR
(300 MHz, DMS0-d6) 8 12.84 (s, 1H), 8.78 (s, 1H), 8.14 (s, 1H), 8.04 (s, 1H), 7.73 (s, 1H),
7.53 (m, 1H), 7.29-7.22 (m, 3H), 5.35 (s, 2H), 2.39 (s, 3H).
Example 67: 5-(5-Fluoro-1 H-benzimidazol-1 -yl)-3-[(2-methvlbenzyl)oxy]thiophene-
2-carboxylic acid and 5-(6-Ruoro-1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxv]-
thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with a 1:1 regioisomeric
mixture of methyl 5-(5-fluoro-1 H-benzimidazol-1-yl)-3-[(2-methylbenzyl)-
oxy]thiophene-2-carboxylate and methyl 5-(6-fluoro-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)-oxy]thiophene-2-carboxylate (0.285 g, 0.719 mmol) provided 0.215 g
(78%) of a 1:1 regioisomeric mixture of 5-(5-fluoro-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-thiophene-2-carboxylic acid and 5-(6-fluoro-1H-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxy]-thiophene-2-carboxylic acid as a yellow solid. lH NMR
(300 MHz, DMSO-d6) 6 12.81 (br s, 1H), 8.76, 8.69 (2xs, 1H), 7.84 (m, 1H), 7.72,7.70
(2xs, 1H), 7.66 (m, 1H), 7.53 (d, J - 6.3 Hz, 1H), 7.36-7.19 (m, 4H), 5.35, 5.34 (2xs, 2H),
2.38 (s, 3H).
Example 68: 5-(5-Methoxv-1H-benzimidazol-1-yl)-3-[(2-
methvibenzyl)oxvltrtiophene-2-carboxylic acid and 5-(6-Methoxy-1 H-benzimidazol-
1 -yl)-3-[(2-methylbenzyl)-oxy]thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with a 1 ;1 regioisomeric
mixture of niethyl 5-(5-methoxy-1H-benzimidazol-1-yl)-3-[(2-methylbenzyI)oxy]-
thiophene-2-carboxylate and methyl 5-(6-methoxy-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylate (0.243 g, 0.595 mtnol) provided 0.217 g
(92%) of a 1:1 regioisomeric mixture of 5-(5-methoxy-1H-benzimidazol-1-yl)-3-[{2-
methylbenzyl)oxy]thiophene-2-carboxylic acid and 5-(6-methoxy-1H-benzimidazol-
1-yl)-3-[(2-methy1benzyl)oxy]thiophene-2-carboxylic acid as a pale yellow solid. 1H
NMR (300 MHz, DMSO-d6) 8 8.93,8.79 (2xs, 1H), 7.80-7.68 (m, 2H), 7.53 (d, J - 6.6 Hz,
1H), 7.35, 7.31-7.17 (m, 4H),7.-K), 7.04 (2xdd, J= 9.0, 2.4 Hz, J= 8.9, 2.3 Hz, 1H), 5.34
(s, 2H), 2.38 (s, 3H).
Example 69: 5-(5-Bromo-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thiophene-
2-carboxylic acid and 5-(6-Bromo-1H-benzimidazol-1-yl)-3-[(2-
methvlbenzvl)oxylthiophene-2-carboxyricacid
An analogous procedure to that described in Example 33 with a 1:1 regioisomeric
mixture of methyl 5-(5-bromo-1H-benzimidazoI-1-yl)-3-[(2-methYlbenzyl)-
oxy]thiophene-2-carboxylate and methyl 5-(6-bromo-1Hbenzimidazol-1-yl)-3-[(2-
methylbenzyl)-oxy]thiophene-2-carboxylate (0.100 g, 0.219 mmol) provided 0.0599 g
(62%) of a 1:1 regioisomeric mixture of 5-(5-bromo-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]-thiophene-2-carboxylic acid and 5-(6-bromo-1 W-benzimidazol-1-
yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxylic acid as a yellow solid. 1H NMR
(300 MHz, DMSO-d6) 5 12.81 (brs, 1H), 8.75,8.70 (s, 1H), 8.04, 7.93 (2xd,J= 1.8 Hz, J
= 1.8 Hz, 1H), 7.81, 7.77 (2xd, J - 8.8 Hz, J = 8.7 Hz, 1H), 7.73, 7.72 (2xs, 1H). 7.61 -7.50
(m, 2H), 7.31-7.20 (m, 3H), 5.35, 5.33 (2xs, 2H), 2.39, 2.38 (s, 3H).
Example 70: 5-(6-Chloro-1 H-benzimidazol-1-yl)-3-[(2-Chloro-4-fluorobenzyl)oxy3-
thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(6-chloro-
1H-benzimidazol-1-yl)-3-[(2-chloro-4-fluorobenzyl)oxy]thiophene-2-carboxylate
(0.128 g, 0.284 mmol) yielded 0.0805 g (65%) of 5-(6-chloro-1H-benzimidazol-1-yl)-
3-[(2-chloro-4-fluorobenzyl)oxy]-thiophene-2-carboxylic acid as a white solid. 1H
NMR (300 MHz, DMSO-d6) d 12.88 (br s, 1H), 8.73 (s. 1H), 7.93-7.74 (m, 3H), 7.71 (s,
1H), 7.55 (dd, J = 8.8,2.5 Hz, 1H), 7.41 (dd, J =8.6,1.9 Hz, 1H), 7.34 (ddd, J = 9.7, 8.5,
2.5 Hz, 1H),5.39(s,2H).
Example 71: 5-(6-Chloro-1 H-benzimidazol-1-yl)-3-[(2,4-difluorobenzyl)oxy]-
thiophene-2-carboxyric acid
An analogous procedure to that described in Example 33 with methyl 5-(6-chloro-
1H-benzimidazol-1-yl)-3-[(2,4-difluorobenzyl)oxy]thiophene-2-carboxylate (0.119 g,
0.274 mmol) yielded 0.0860 g (75%) of 5-(6-chloro-1H-benzimidazol-1-yl)-3-[(2,4-
difluorobenzyl)oxy]-thiophene-2-carboxylic acid as an off-white solid. 1H NMR (300
MHz, DMSO-d6) 8 8.72 (s, 1H), 7.87 (d, J = 1.8 Hz, 1H), 7.82 (d, J= 8.6 Hz, 1H), 7.72 (m,
1H), 7.71 (s, 1H), 7.41 (dd, J= 8.6,2.0 Hz, 1H), 7.34 (m, 1H), 7.18 (m, 1H), 5.38 (s, 2H).
Example 72: 5-(6-Chloro-1 H-benzimidazol-1 -yl)-3-(pyridin-3-vlmethoxy)thiophene-
2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(6-chloro-
1 H-benzimidazol-1-yl)-3-(pyridin-3-ylmethoxy)thiophene-2-carboxylate (0.0380 g,
0.0950 mmol) afforded 0.010 g (27%) of 5-(6-chloro-1H-benzimidazol-1-yl)-3-
(pyridin-3-ylmethoxy)thiophene-2-carboxylic acid as a tan solid. "H NMR (300 MHz,
DMSO-d6) 8 8.68 (s, 1H), 8.77 (m, 1H), 8.72 (s, 1H), 8.32 (d, J= 7.9 Hz, 1H), 7.87-7.79
(m, 3H), 7.69 (s, 1H), 7.42 (dd. J=- 8.6, 2.0 Hz, 1H), 5.52 (s, 2H).
Example 73: 5-(1 H-Benzimidazol-1 -yl)-3-(prop-2-vnvloxy)thiophene-2-carboxylic
acid
An analogous procedure to that described in Example 33 with methyl 5-(1H-
benzimidazol-1-yl)-3-(prop-2-ynyloxy)thiophene-2-carboxylate (0.183 g, 0.586 mmol)
gave 0.175 g (100%) of 5-(1 H-benzimidazol-1~yl)-3-(prop-2-ynyloxy)thiophene-2-
carboxylic acid as a tan solid. 1H NMR (300 MHz, CD3OD) 8 9.83 (s, 1H), 7.98 (m, 2H),
7.77 (m, 2H), 7.71 (s, 1H). 5.02 (d, J = 2.3 Hz, 2H), 3.17 (t. J - 2.3 Hz. 1H). MS (m/z) 299
(m+1).
Example 74: 5-(6-Bromo-1 H-benzimidazol-1-yl)-3-{[2-(trifluoromethvl)-
benzyl]oxy)thiophene-2-carboxylicacid
An analogous procedure to that described in Example 33 with methyl 5-(6-bromo-
1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-benzyl]oxy}thiophene-2-carboxylate
(0.0155 g, 0.0303 mmol) gave 0.0080 g (53%) of 5-(6-bromo-1H-benzimidazol-1-yl)-
3-{[2-(trifluoromethyl)-benzyl]oxy}thiophene-2-carboxylic acid as a yellow solid. 1H
NMR (300 MHz, DMSO-ds) 5 8.71 (s, 1H). 7.98-7.93 (m, 2H), 7.84-7.74 (m, 3H), 7.68 (s,
1H), 7.62 (m, 1H), 7.53 (dd, J= 8.6,1.9 Hz, 1H), 5.50 (s, 2H).
Example 75: 5-(5,6-Dimethoxy-1 tf-benzimidazol-1-vl)-3-{[2-(trifluoromethvl)-
benzyl]-oxy}thiophene-2-carboxylicacid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-[(2-trifluoromethylbenzyl)-oxy]thiophene-2-
carboxylate (0.0691 g, 0.140 mmol) yielded 0.0558 g (83%) of 5-(5,6-dimethoxy-1H-
benzimidazol-1-yl)-3-{[2-(trifluoromethyl)benzyl]-oxy}thiophene-2-carboxylicacid
as a yellow solid. 1H NMR (300 MHz, DMSO-d6) d 8.49 (s, 1H), 7.95 (d, J - 7.6 Hz, 1H),
7.84-7.74 (m, 2H), 7.65-7.56 (m, 2H), 7.34 (s, 1H), 7.25 (s, 1H), 5.50 (s, 2H), 3.84 (s, 3H),
3.83 (s, 3H).
Example 76: 3-[(2-Bromobenzvl)oxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1-
Vl)thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 3-[(2-
bromobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-carboxylate
(0.0719 g, 0.143 rrimol) afforded 0.0597 g (85%) of 3-[(2-bromobenzyl)oxy]-5-(5.6-
dimethoxy-1H-benzimidazol-1-yl)thiophene-2-carboxylic acid as a yellow solid. 1H
NMR (300 MHz, DMSO-d6) 8 8.54 (s, 1H). 7.77-7.66 (m, 2H), 7.63 (s, 1H), 7.47 (m, 1H),
7.38-7.29 (m, 2H), 7.26 (s, 1H), 5.37 (s, 2H), 3.84 (s, 3H), 3.83 (s, 3H).
Example 77: 5-(5,6-Dichloro-1 /V-benzimidazol-1-vl)-3-(3-furylmethoxv)thiophene-2-
carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dichloro-1 W-benzimidazol-1 -yl)-3-(3-furylmethoxy)-thiophene-2-carboxylate (0.0715
g, 0.169 mmol) provided 0.0476 g (69%) of 5-(5,6-dichloro-1 W-benzimidazol-1 -yl)-3-
(3-furylmethoxy)thiophene-2-carboxylic acid as a tan-orange solid. 1H NMR (300
MHz, DMSO-ds) 8 12.82 (br s, 1H), 8.78 (s, 1H), 8.13 (s, 1H), 8.06 (s, 1H), 7.85 (s, 1H),
7.69 (m, 1H), 7.68 (s, 1H). 6.61 (m. 1H), 5.21 (s, 2H).
Example 78: 5-(5,6-Dichloro-1 H-benzimidazol-1-yl)-3-(2-furylrnethoxv)thiophene-2-
carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dichloro-1 H-benzimidazol-1 -Yl)-3-(2-furylmethoxy)-thiophene-2-carboxyiate (0.0525
g, 0.124 mmol) gave 0.0289 g (57%) of 5-(5,6-dichloro-1 W-benzimidazol-1 -yl)-3-(2-
furylmethoxy)thiophene-2-carboxylic acid as a yellow solid. 1H NMR (300 MHz,
DMSO-de) 8 12.85 (br s, 1H), 8.78 (s, 1H). 8.14 (s, 1H), 8.08 (s. 1H), 7.74 (dd, J= 1.9.0.7
Hz, 1H), 7.71 (s, 1H), 6.70 (d. J = 3.2 Hz, 1H), 6.51 (dd, J - 3.2,1.9 Hz, 1H), 5.32 (s, 2H).
Example 79: 5-(5,6-Dichloro-1 H-benzimidazol-1 -yl)-3-(thien-3-ylmethoxy)-
thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dichloro-1 W-benzirnidazol-1 -yI)-3-(thien-3-ylmethoxy)-thiophene-2-carboxylate
(0.0730 g, 0.166 mmol) afforded 0:0476 g (67%) of 5-(5,6-dich!oro-1H-benzirmdazol-
1-yl)-3-(thien-3-ylmethoxy)thiophene-2-carboxyljc acid as a yellow solid. 1H NMR
(300 MHz, DMSO-d6) d 12.84 (br s, 1H), 8.77 (s. 1H), 8.13 (s. 1H), 8.02 (s, 1H), 7.67 (s,
1H). 7.66 (m, 1H), 7.59 (dd. J= 5.0.3.0 Hz, 1H). 7.22 (dd, J = 5.0.1.2 Hz, 1H). 5.35 (s.
2H).
Example 80: 5-(5.6-Dichloro-1 H-benzimidazol-1 -yl)-3-(thien-2-ylmethoxy)-
thiophene-2-carboxylic acid
An analogous procedure to that described in Example 33 with methyl 5-(5,6-
dichloro-1H-benzimidazol-1-yl)-3-(thien-2-ylmethoxy)-thiophene-2-carboxylate
(0.0580 g, 0.132 mmol) afforded 0.0341 g (61%) of 5-(5,6-dichloro-1H-benzimidazol-
1-yl)-3-(thien-2-ylmethoxy)thiophene-2-carboxylic acid as a pale yellow solid. "H
NMR (300 MHz, DMSO-d6) 6 8.77 (s, 1H), 8.13 (s, 1H), 8.03 (s, 1H), 7.70 (s, 1H), 7.61 (dd,
J = 5.0,1.1 Hz, 1H), 7.30 (dd, J = 3.5,1.1 Hz, 1H), 7.07 (dd, J = 5.0,3.5 Hz, 1H), 5.54 (s,
2H).
Example 81: Methyl 3-[(2-chloro-4-fluorobertzy))oxy]-5-(5,6-dimethoxy-1H-
benzimidazol-1-yl)thiophene-2-carboxylate
An analogous procedure to that described in Example 45 with methyl 5-(5,6-
dimethoxy-1 H-benzimidazol-1-yl)-3-hydroxythipphene-2-carboxylate (0.100 g, 0.299
mmol) and 2-chloro-4-fIurobenzyl bromide (0.0809 g, 0.362 mmol) provided 0.0963 g
(68%) of methyl 3-[(2-chloro-4-fluorobenzyl)oxy]-5-(5,6-dimethoxy-1 H-
benzimidazol-1-yl)thiophene-2-carboxylate as a yellow solid. 1H NMR (300 MHz,
DMSO-ds) 8 8.50 (s, 1H), 7.80 (dd, J - 8.6, 6.2 Hz, 1H), 7.70 (s, 1H), 7.55 (dd, J - 8.8, 2.6
Hz, 1H), 7.39-7.31 (m, 1H), 7.35 (s, 1H), 7.27 (s, 1H), 5.41 (s, 2H), 3.85 (s, 3H), 3.83 (s,
3H). 3.78 (s, 3H).
Example 82: A/-({5-(5-methoxv-1H-benzimidazol-1-yl)-3-[(2-methylbenzyl)-
oxy]thieri-2-yl}carborivl)methanesulfonamide and N-({5-(6-methoxy-1 H-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxy]thien-2-yl}carbonyl)methanesulfonamide.
A 1:1 regioisomeric mixture of 5-(5-methoxy-1H-benzimidazol-1-yl)-3-[(2-
methylbenzyl)oxy]thiophene-2-carboxylic acid and 5-(6-methoxy-1H-benzimidazol-
1-yl)-3-[(2-methylbenzyl)oxy]thiophene-2-carboxylic acid (0.100 g, 0.254 mmol), 4-
dimethylaminopyridine (0.0403 g, 0.330 mmol), and methanesulfonamide (0.0313 g,
0.329 mmol) were dissolved in 4 mL of dichloromethane with stirring. Triethylamine
(0.046 mL, 0.33 mmol) was added via syringe followed by the addition 1-(3-
dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (0.0633 g, 0.330 mmol) in a
single portion. The mixture was stirred for 12 hours and subsequently poured into 5°/o
aqueous HCI solution and ethyl acetate. The layers were separated, and the organic
layer was washed with brine. The combined aqueous layers were extracted with ethyl
acetate, and the combined organic layers were dried over MgSO4. Filtration,
concentration in vacuo, and purification by flash chromatography afforded 0.0826 g
(69%) of a 1:1 regioisomeric mixture of N-({5-(5-methoxy-1H-benzimidazol-1-yl)-3-
[(2-methylbenzyl)oxy]thien-2-yl}carbonyl)-methanesulfonamide and N-({5-(6-
methoxy-1 H-benzimidazol-1 -yl)-3-[(2-methylbenzyl)oxy]thien-2-
yl}carbonyl)methanesulfonamide as a pale green solid. 1H NMR (300 MHz, DMSO-d6)
8 9.97 (br s, 1H), 8.70, 8.58 (2xs, 1H), 7.83-7.68 (m, 2H), 7.55 (m, 1H), 7.37-7.21 (m,
4H), 7.07, 7.01 (2xdd, J = 8.8, 2;3 Hz, 1H), 5.51 (s, 2H), 3.85,3.83 (2xs, 3H), 3.37,3.36
(2xs, 3H), 2.41 (s, 3H). MS (m/z) 472 (m+1).
Examples 83-158.
Unless otherwise noted, the following compounds were prepared similarly according
to general procedures outlined for Examples 2A, 33,40,45,57 (or Intermediate
Example 21), and 61 (where 7M NH3 in MeOH was used instead of 2M NH3 in MeOH).
Example 83: 5-(5-Chloro-2-methyl-1H-benzim8dazol-1-yl)-3-[(2-
methvlbenzvl)oxy]thiophene-2-carboxvlic acid and 5-(6-Chloro-2-methyl-1 H-
benzimidazol-1-yl)-3-[(2-methylbenzyl)oxv]thiophene-2-carboxylicacid
1H NMR (400 MHz, CD3OD) 8 7.61-7.56 (m, 1H); 7.46 (d,J = 7.2 Hz, 1H); 7.28-7.21 (m,
6H); 5.34 (s, 2H); 2.52 (s, 3H); 2.43 (s, 3H). MS (ES-, m/z) 411 (m-1).
Example 84: 3-(Benzyloxy)-5-(5-chloro-1 H-benzimidazol-1-yl)thiophene-2-
carboxamide
1H NMR (400 MHz, DMSO-d6) 5 8.70 (s, 1H), 7.88 (d, J = 2.01 Hz, 1H), 7.78-7.70 (m,
2H), 7.65 (s, 1H). 7.56-7.52 (m, 2H), 7.46-7.35 (m, 4H), 7.01 (s, 1H), 5.40 (s, 2H). MS
(ES+, m/z) 383 (m+1).
Example 85: 5-(5-Chloro-1H-benzimidazol-1-yl)-3-({2-
[(phenylsulfonyl)methyl]benzyl}oxy)thiophene-2-carboxvlic acid and 5-(6-Chloro-1H-
benzimidazol-1-vl)-3-({2-[(phenylsulfonyl)methyl]benzyl}oxv)thiophene-2-carboxylic
acid
1H NMR (400 MHz, DMSO-d6) 5 8.75 (s, 1H), 8.70 (s, 1H), 7.91 (d, J - 1.96 Hz, 1H), 7.84
- 7.58 (m, 17H), 7.47 (dd, J = 1.96 Hz, 8.74 Hz, 1H), 7.43 - 7.26 (m, 5H), 7.12 (t, J =
7.76 Hz, 2H), 5.38 (s, 4H), 4.93 (s, 4H). MS (ES+, m/z) 540 (m+1).
Example 86: 5-(5-Chloro-1H-benzimidazol-1-yl)-3-{1-[3-
(trifluoromethvl)phenyl1ethoxy)thiophene-2-carboxylic acid and 5-(6-Chloro-1 H-
benzimidazol-1 -yl)-3-( 1 -[3-(trifluoromethyl)phenvl]ethoxy}thiophene-2-carboxylic
acid
1H NMR (400 MHz, DMSO-d6) d 12.94 (br s, 2H), 8.70 (s, 1H), 8.65 (s, 1H), 7.94 - 7.63
(m, 13H), 7.58 (s,2H), 7.41 (t, J = 8.03,2H), 5.88 (dd,J = 6.06 Hz, 11.06 Hz,2H), 1.64 (t
J » 6.24 Hz, 6H). MS (ES+, m/z) 467 (m+1).
Example 87: 5-[6-(2,2,2-Trifluoroethoxy)-1H-bt:nzimidazol-1-yll-3-U2-
(trifluoromethvt)benzylloxy}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.55 (s. 1H), 7.86 - 7.63 (m, 7H), 7.38 (d, J - 2.38 Hz,
1H), 7.10 (dd, J= 2.29 Hz, 8.88 Hz, 1H), 6.82(br s, 1H), 5.56 (s, 2H), 4.86(q.J- 8.85 Hz,
2H). MS (ES+, m/z) 516 (m+1).
Example 88: 5-(2,2-Difluoro-5H-[1 ,3]dioxolol4.5-flbenzirnidazot-5-y1)-3-{[2-
(trifluoromethyl)benzYlloxy>thiophene-2-carboxamide
"H NMR (400 MHz, DMSO-d6) 8 3.66 (s, 1H), 7.92 (s, 1H)f 7.88 (s, 1H), 7.87-7.64 (m,
6H). 6.79 (br s, 1H), 5.56 (s. 2H). MS (ES+, m/z) 498 (m+1).
Example 89: 5-(7,8-Dihvdro-1H,6H-[1.4]dioxepino[2.3-flbenzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzYl]oxv}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.56 (s, 1H). 7.85 (s, TH). 7.83 (s, 1H), 7.77 (t.J- 7.60
Hz, 1H), 7.69 (br s, 1H), 7.64 (t, J = 7.60 Hz, TH), 7.60 (s. 1H), 7.36 (s, 2H), 6.76 (br s,
1H), 5.54 (s, 2H),4.15-4.06 (m, 4H), 2.11 (t,J = 4.94 Hz, 2H). MS (ES+, m/z) 490 (m+1).
Example 90: 5-(5,6-Dimethoxv-1H-benzimidazol-1-yl)-3-{[3-
(dimethytamino)benzyl]oxy}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.40 (s, 1H), 7.73 (br s, 1H), 7.60 (s, 1H), 7.33 (s, 1H),
7.20 (t, J - 7.87 Hz, 1H), 7.15 (s, 1H), 7.07 (br s, 1H), 6.88 (s, H), 6.79 (d, J - 7.51 Hz,
1H), 6.70 (dd, J= 2.29 Hz, 8.33 Hz, 1H), 5.34 (s, 2H), 3.82 (s, 6H), 2.89 (s, 6H).
Example 91: 3-[(6-Chloro-1,3-benzodioxol-5-yl)methoxy]-5-(5,6-dimethoxy-1H-
benzimidazol-1-yl)thiophene-2-carboxamide
"H NMR (400 MHz, DMSO-d6) d 8.43 (s, 1H),7.73 (br s, 1H), 7.67 (s, 1H), 7.35 (s, 1H),
7.33 (s, 1H), 6.90 (br s, 1H), 6.11 (s, 2H), 5.36 (s, 2H), 3.86 (s, 3H), 3.83 (s, 3H). MS (ES+,
m/z) 488 (m+1).
Example 92: 5-(5,6-Dimethoxy-1 H-benzimidazol-1 -yl)-3-[(2-
nitrobenzvl)oxvlthiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 6 8.38 (s, 1H), 8.19 (d, J= 8.1 Hz, 1H), 7.84 (t, J= 7.6 Hz,
1H), 7.78-7.76 (m, 2H), 7.65 (m, 1H), 7.57 (s. 1H), 7.32 (s, 1H). 7;09 (br s, 1H). 7.07 (s,
1H) 5.79 (s, 2H), 3.81 (s, 3H), 3.76 (s, 3H). MS (ES+, m/z) 455 (m+1).
Example 93: 3-(1,1"-Biphenvl-2-ylmethoxy)-5-(5,6-dimethoxv-1 H-benzimidazol-1-
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.36 (s, 1H), 7.72 (m. 1H), 7.61 (br s, 1H), 7.52-7.48 (m,
2H), 7.46-7.33 (m, 8H), 7.15 (s, 1H), 6.62 (br s, 1H) 5.34 (s, 2H), 3.83 (s, 3H). 3.82 (s, 3H).
MS (ES+. m/z) 486 (m+1).
Example 94: 5-(5,6-Dimethbxy-1 H-benzimidazol-1-yl)-3-[(3-iodobenzyl)oxy]-
thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 5 8.40 (s, 1H), 7.96 (m, 1H), 7.73 (d, J = 7.3 Hz, 1H), 7.59-
7.57 (m, 3H), 7.34 (s, 1H), 7.15 (s, 1H), 7.10 (br s, 1H). 5.38 (s, 2H), 3.84 (s, 3H), 3.83 (s,
3H). MS (ES+. m/z) 536 (m+1).
Example 95: 3-[(2-Cvanobenzyl)oxy]-5-(5,6-dirriethoxy-1 H-benzimidazoi-1 -
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.47 (s, 1H), 7.98 (d, J"- 7.3 Hz, 1H), 7.85-7.77 (m, 3H),
7.70 (s, 1H), 7.62 (m, 1H). 7.35 (s, 1H), 7.22 (s, 1H), 6.92 (br s, 1H), 5.60 (s, 2H), 3.85 (s,
3H). 3.83 (s, 3H). MS (ES+. m/z) 435 (m+1).
Example 96: 3-[(3-Aminobenzyl)oxy]-5-(5,6-dimethoxv-1 H-benzimidazol-1 -
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.41 (s, 1H). 7.73 (br s, 1H), 7.53 (s, 1H), 7.34 (s, 1H),
7.16 (s, 1H), 7.04 (t, J - 7.7 Hz, 1H), 7.00 (br s, 1H). 6.67-6.63 (m, 2H), 6.64 (d, J = 7.8
Hz, 1H), 5.27 (s, 2H), 5.18 (d, J= 7.8 Hz, 2H), 3.83 (m, 6H). MS (B+, m/z) 425 (m+1).
Example 97: 5-(5,6-Dimethoxy-1 H-benzimidazol-1-yl)-3-{[2-(methylthio)benzyl]-
oxy}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.42 (s, 1H), 7.70 (br s, 1H). 7.66 (s, 1H), 7.55 (d, J= 7.5
Hz, 1H), 7.41 (m, 2H), 7.33 (s, 1H), 7.21 (s, 2H), 6.87 (br s, 1H), 5.40 (s, 2H). 3.84 (s. 3H),
3.81 (s, 3H), 2.50 (s, 3H). MS (ES+, m/z) 456 (m+1).
Example 98: 5-(5,6-Dimethoxy-1H benzimidazol-1 -yl)-3-{[2-
(methylsulfinyl)benzyl]oxy}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.42 (s, 1H). 7.99 (d, J= 7.7 Hz, 1H), 7.73-7.68 (m, 3H),
7.65-7.62 (m. 2H), 7.34 (s, 1H), 7.21 (s, 2H), 6.92 (br s, 1H), 5.50 (in, 2H), 3.84 (s, 3H),
3.83 (s, 3H), 2.77 (s, 3H). MS (ES+, m/z) 472 (m+1).
Example 99: 5-(5,6-Dimethoxv-1H-benzimidazol-1-yl)-3-{[2-
(methylsulfonyl)benzyl]oxy)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.42 (s, 1H), 8.03 (d, J- 7.8 Hz, 1H), 7.86-7.79 (m, 2H),
7.70-7.67 (m, 2H), 7.59 (s, 1H), 7.33 (s, 1H), 7.19 (s, 1H), 7.11 (br s, 1H), 5.79 (s, 2H),
3.82 (m, 6H), 3.34 (s, 3H). MS (ES+, m/z) 488 (m+1).
Example 100: 3-[(2-Aminopvrid"m-4-yl)methoxy]-5-(5,6-dimethoxy-1 H-
benzimidazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.39 (s. 1H), 7.91 (d,J=5.1 Hz, 1H), 7.76 (br s, 1H),
7.46 (s, 1H), 7.33 (s, 1H), 7.12 (s, 1H), 7.07 (br s, 1H), 6.56 (d, J - 5.2 Hz, 1H), 6.49 (s, 1H)
6.03 (s, 2H), 5.31 (s, 2H), 3.82 (s, 3H), 3.81 (s, 3H). MS (ES+, m/z) 426 (m+1).
Example 101: 3-[(2-Chloropyridin-3-vl)methoxv]-5-(5,6-dimethoxy-1 H-
benzimidazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) S 8.45 (dd, J= 4.8,1.9 Hz, 1H), 8.43 (s, 1H), 8.11 (dd, J =
7.7,1.8 Hz, 1H),7.75(s, iH),7.66(s, 1H),7.53 (dd, J = 7.4,4.8 Hz, 1H),7.34(s, 1H),7.21
(s, 1H), 7.00 (br s, 1H), 5.49 (s, 2H), 3.84 (s, 3H), 3.83 (s, 3H). MS (ES+, m/z) 445 (m+1).
Example 102: 5-(5.6-Dimethoxy-1 H-benzimidazol-1-yl)-3-[(2-fluoropyridin-3-
yl)methoxy]thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.43 (s, 1H), 8.28 (d, J= 4.5 Hz, 1H), 8.19 (m, 1H), 7.87
(m, 1H), 7.67 (s, 1H), 7.45 (m, 1H), 7.34 (s, 1H), 7.21 (s, 1H), 6.97 (br s, 1H), 5.49 (s, 2H),
3.85 (s, 3H), 3.83 (s. 3H). MS (ES+, m/z) 429 (rh+1).
Example 103: 5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-[(2-
vinylbenzvl)oxy]thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.46 (s, 1H), 7.73-7.70 (m, 2H), 7.61 (m, 1H), 7.48-7.36
(m, 2H), 7.26 (s, 1H), 7:24-7.14 (m, 3H), 6.82 (br s, 1H), 5.87 (d, J = 16.6 Hz, 1H), 5.54 (d,
J =11.8 Hz, 1H), 5.54 (s, 2H), 3.88 (s, 3H), 3.86 (s, 3H). MS (ES+, m/z) 436 (m+1).
Example 104: 3-{[4-(Ammocarbonyl)benzyl]oxy}-5-(5,6-dimethoxv-1H
benzimidazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz. DMSO-d6) 8 8.43 (s, 1H), 8.01 (br s, 1H), 7.93 (d, J = 8.2Hz, 2H), 7.76
(brs, 1H), 7.65 (d, J - 8.2 Hz, 2H), 7.61 (s, 1H), 7.43 (br s, 1H), 7.36 (s, 1H), 7.16 (s, 1H),
7.12 (br s. 1H). 5.51 (s, 2H), 3.85 (m. 6H). MS (ES+, m/z) 453 (m+1).
Example 105: 3-[(2-Acetylbenzyl)oxy]-5-(5,6-dimethoxy-1 H-benzimidazol-1-
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-ds) S 8.43 (s, 1H), 8.05 (d, J=7.8 Hz, 1H), 7.72-7.64 (m, 2H),
7.59-7.55 (m, 2H),7.35 (s, 1H), 7.17 (s, 1H), 7.17 (m, 2H), 5.66 (s, 2H), 3.85 (s, 3H), 3.84
(s. 3H), 2.65 (s, 3H). MS (ES+. m/z) 452 (m+1).
Example 106: 5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-[(2-
ethvriylbenzvl)oxy]thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.43 (s, 1H), 7.72 (br s, 1H), 7.66-7.51 (m, 3H). 7.49-
7.41 (m, 2H), 7.34 (s, 1H), 7.20 (s, 1H), 6.94 (br s, 1H), 5.50 (s, 2H), 4.54 (s, 1H), 3.85 (s,
3H), 3.83 (s, 3H). MS (ES+, m/z) 434 (m+1).
Example 107: 5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethoxy)benzyl] oxy} thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.43 (s, 1H), 7.76 (m, 2H), 7.65 (s, 1H), 7.56 (m, 1H),
7.50-7.46 (m, 2H), 7.34 (s, 1H), 7.22 (s, 1H), 6.86 (br s, 1H), 5.48 (s, 2H), 3.84 (s, 3H),
3.83 (s, 3H). MS (ES+, m/z) 494 (m+1).
Example 108: 3-{[2-(Difluoromethoxy)benzyl]oxy}-5-(5.6-dimethoxy-1 H-
benzimtdazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.42 (s, 1H), 7.72 (br s, 1H), 7.67 (d,7= 7.7 Hz, 1H),
7.64 (s, 1H), 7.51-7.47 (m, 2H), 7.34 (s, 1H), 7.32-7.28 (m. 2H), 7.21 (s, 1H), 6.91 (br s,
1H), 5.43 (s, 2H), 3.84 (s, 3H), 3.83 (s, 3H). MS (ES+, m/z) 476 (m+1).
Example 109: 3-{ [2-(1,2-Dihvdroxvethvl)benzvl]oxy}-5-(5.6-dimethoxy-1H-
benzimidazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 8 8.40 (is, 1H), 7.64 (br s, 1H), 7.59 (s, 1H), 7.52 (d, J = 7.5
Hz, 1H), 7.46 (d. J= 7.3 Hz, 1H), 7.37 (m, 1H). 7.32-7.28 (m, 2H), 7.17 (s, 1H), 6.92 (br s,
1H), 5.49 (m. 2H), 5.35 (d, J = 4.0 Hz, 1H), 4.87 (m, 1H), 4.81 (t, J - 5.8 Hz, 1H), 3.98 (m,
1H), 3.80 (s, 6H), 3.53 (m, 1H). MS (ES+, m/z) 470 (m+1).
Example 110: 5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-[(2-
formvlbenzyl)oxy]thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 10.28 (s, 1H), 8.41 (s, 1H), 8.02 (m, 1H), 7.73-7.64 (m,
4H), 7.58 (s. 1H), 7.34 (s, 1H), 7.15 (s, 1H), 7.02 (m. 1H), 5.81 (s, 2H), 3.82 (s, 3H), 3.81 (s,
3H), 2.65 (s, 3H). MS (ES+, m/z) 438 (m+1).
Example 111: 3-(Cyclohexylmethoxy)-5-(5,6-dimethoxy-1 H-benzimidazol-1-
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) 5 8.43 (s, 1H), 7.73 (br s, 1H), 7.55 (s, 1H), 7.34 (s, 1H),
7.25 (s, 1H), 6.89 (s, 1H), 4.13 (d. J = 6.3 Hz, 2H), 3.85 (s, 3H), 3.83 (s, 3H), 1.88-1.64 (m
6H), 1.31-1.01 (m, 5H). MS (ES+, m/z) 416 (m+1).
Example 112: 5-(5,6-Dimethoxv-1H-benzimidazol-1-yl)-3-(tetrahydro-2H-Pyran-2-
ylmethoxy) thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.42 (s, 1H), 7.72 (br s, 1H), 7.53 (s, 1H), 7.33 (s, 1H),
7.25 (s, 1H), 7.09 (br s, 1H), 4.30 (dd, J = 10.8,3.2 Hz, 1H), 4.20 (dd, J = 10.6, 6.9 Hz,
1H), 3.92 (d. J= 11.1 Hz, 1H),3.86 (s, 3H),3.83 (s, 3H). 3.72 (m, 1H), 1.83 (m, 1H), 1.64
(d, J= 13.1HZ, 1H), 1.54-1.47 (m.4H), 1.38 (m, 1H). MS (ES+, m/z) 418 (m+1).
Example 113: 5-(5.6-Djmethoxy-1 H-benzimidazol-1 -yl)-3-(2-morpholin-4-
ylethoxy)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.41 (s, 1H), 7.75 (br s, 1H), 7.56 (s, 1H), 7.52 (br s. 1H),
7.34 (s, 1H), 7.25 (s, 1H). 4.38 (t J= 4.6 Hz, 2H), 3.85 (s, 3H), 3.83 (s, 3H), 3.59 (m, 4H),
2.72 (t, J m 6.7 Hz, 2H), 2.47 (m, 4H). MS (ES+, m/z) 433 (m+1).
Example 114: 5-(5,6-Dimethoxv-1H-benzimidazol-1-yl)-3-(2-phenvlethoxy)-
thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.42 {s, 1H), 7.64 (br s, 1H), 7.56 (s, 1H), 7.37-7.31 (m,
5H), 7.26-7.23 (m, 2H), 6.75 (br s, 1H), 4.53 (t, J - 6.8 Hz, 2H), 3.85 (s, 3H), 3.82 (s, 3H),
3.15 (t, J = 6.7 Hz. 2H). MS (ES+, m/z) 424 (m+1).
Example 415: 5-(5.6-Dimetrtoxy-1H-benzirnidazol-1-yl)-3-(3-
phenvlpropoxy)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.42 (s, 1H), 7.70 (br s, 1H), 7.52 (s, 1H), 7.34 (s, 1H),
7.31-7.24 (m, 5H), 7.19 (m, 1H), 6.97 (br s, 1H), 4.31 (t, J= 6.8 Hz, 2H), 3.83 (s, 3H),
3.83 (s, 3H). 2.76 (t, J = 6.7 Hz, 2H), 2.13 (m, 2H). MS (ES+, m/z) 438 (m+i).
Example 116:5-(1H-Benzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzvl]oxy)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.67 (s, 1H). 7.87-7.85 (m, 2H), 7.82-7.77 (m, 3H), 7.72-
7.64 (m, 3H), 7.45-7.36 (m, 2H), 6.79 (br s, 1H), 5.56 (s, 2H). MS (ES+. m/z) 418 (m+1).
Example 117: 5-(1H-Benzimidazol-1 -yl)-3-[(2-nitrobenzyl)oxy]thiophene-2-
carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.61 (s, 1H), 8.19 (d, J = 7.6 Hz, 1H), 7.84-7.62 (m, 7H),
7.41-7.35 (m, 2H), 7.05 (br s, 1H), 5.78 (s, 2H). MS (ES+, m/z) 395 (m+1).
Example 118: 5-(6-Methoxy-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide
1H NMR (300 MHz, DMSO-d6) d 8.49 (s, 1H), 7.87-7.61 (m, 7H), 7.21 (d, J - 2.4 Hz, 1H),
6.98 (dd, J = 8:9,2.4 Hz, 1H), 6.81 (br s, 1H), 5.56 (s. 2H), 3.83 (s, 3H).
Example 119: 3-[(2-Bromobenzyl)oxy]-5-[6-(trifluoromethyl)-1H-benzimidazol-1-
yl]thiophene-2-carboxamide
1H NMR (300 MHz, DMSO-d6) d 8.86 (s. 1H), 8.05-7.99 (m. 2H), 7.83-7.67 (m, 5H), 7.47
(ddd, J m 8.8,7.5,1.3 Hz, 1H), 7.37 (ddd, J - 9.4,7.6,1.8 Hz, 1H), 6.95 (br s, 1H), 5.46 (s,
2H).
Example 120: 3-[(3-Bromopyridin-4-yl)methoxy]-5-(5,6-dimethoxv-1 H-
benzimidazol-1-yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.81 (s, 1H), 8.63 (d, J= 5.0 Hz, 1H), 8.42 (s, 1H), 7.77
(br s. 1H), 7.62 (d, J = 5.0 Hz, 1H), 7.59 (s, 1H), 7.33 (s, 1H), 7.19 (s, 1H), 7.08 (br s, 1H),
5.49 (s, 2H), 3.824 (s, 3H), 3.818 (s, 3H). MS (ES+, m/z) 489, 491 (m+1).
Example 121: 5-[6-(Methylsulfonyl)-1H-benzimidazol-1-yl]-3-f[2-
(trifluoromethyl)benzyl]oxy}-thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.93 (s, 1H), 8.24 (d, J= 1.7 Hz. 1H), 8.05 (d, J = 8.60
Hz, 1H), 7.92 (dd, J= 8.4,1.7 Hz, 1H), 7.89-7.77 (m, 5H), 7.65 (m, 1H), 6.84 (brs, 1H),
5.54 (s, 2H), 3.29 (s, 3H). MS (ES+. m/z) 496 (m+1).
Example 122: 5-{6-[(Methvlsulfonvi)amino]-1H-benzimidazol-1-yl}-3-{[2-
(trifluoromethyl)benzyll-oxy)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 9.85 (s, 1H), 8.64 (s, 1H). 7.86 (d, J - 7.9 Hz. 1H), 7.82-
7.71 (m, 6H). 7.66 (m, 1H). 7.25 (dd, J - 8.8, 2.0 Hz, 1H), 6.79 (br s, 1H), 5.52 (s, 2H),
2.98 (s, 3H).
Example 123; 5-(6.7-Dihydro-1 H-[1,4]dioxino[2,3-flbenzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzvl]-oxy}thiophene-2-carboxamide
1H NMR (300 MHz, DMSO-d6) d 8.48 (s, 1H), 7.87-7.74 (m, 3H), 7.70-7.61 (m, 2H), 7.60
(s, 1H), 7.25 (s, 1H), 7.24 (s, 1H), 6.76 (br is, 1H), 5.55 (s, 2H), 4.29 (m, 4H). MS (ES+,
m/z) 476 (m+1).
Example 124: 5-(6.7-Dihydro-1 H-[1,4]dioxino[2.3-flbenzimidazol-1-yl)-3-{[1-
(methylsulfonyl)-piperidin-4-yl]methoxy}thiophene-2-carboxamide
1H NMR (300 MHz, DMSO-d6) d 8.48 (s, 1H), 7.71 (br s, 1H), 7.51 (s, 1H), 7.27 (s, 1H),
7.23 (s. 1H), 6.87 (br s, 1H), 4.29 (br s, 4H), 421 (m, 2H), 3.60 (m, 2H), 2.85 (s, 3H), 2.74
(m, 2H), 2.08-1.81(m, 3H), 1.36 (m, 2H). MS (ES+, m/z) 493 (m+1).
Example 125: 1-[5-(Aminocarbonyl)-4-({[2-(trifluorometriyl)phenvl]methyl}oxy)-2-
thienyl]-1 H-benzimidazole-5-earboxamide
1H NMR (400 MHz, DMSO-d6) d 8.75 (s, 1H). 8.36 (d, J = 0.9 Hz, 1H), 8.10 (br s, 1H),
7.99 (dd, J m 8.6,1.4 Hz, 1H), 7.88-7.62 (m, 7H), 7.41 (br s, 1H), 6.80 (br s, 1H), 5.56 (s,
2H). MS (ES+, m/z) 461 (m+1).
Example 126: 3-[1-(2-Chlorophenyl)ethoxy]-5-(5,6-dimethoxy-1 fl-benzimidazol-1-
yl)thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.35 (s, 1H), 7.83 (br s, 1H), 7.68 (dd, J - 7.7,2.0 Hz,
1H), 7.48 (dd, J= 7.8, 1.2 Hz, 1H), 7.43 (ddd, J= 7.5,7.4, 1.2 Hz, 1H), 7.35 (ddd, J= 7.8,
7.6,1.8 Hz, 1H), 7.32 (s, 1H), 7.14 (br s, 1H), 7.13 (s, 1H), 7.02 (s, 1H), 6.01 (q, J - 6.4 Hz,
1H), 3J81 (s, 3H), 3.79 (s, 3H), 1.72 (d, J = 6.4 Hz, 3H). MS (ES+, m/z) 458 (m+1).
Example 127: 5-(5.6-Dimethoxy-1H-benzimidazol-1-yl)-3-[1-(2-
methylphenyl)ethoxyl thiophene-2-carboxamide
"H NMR (400 MHz, DMSO-d6) d 8.31 (s, 1H), 7.96-7.92 (m, 1H), 7.84 (br s, 1H), 7.81-
7.73 (m, 2H), 7.58-7.52 (m, 1H), 7.31 (s, 1H), 7.15 (br s, 1H), 7.05 (s, 1H), 7.01 (s, 1H),
6.01-5.96 (m, 1H), 3.81 (s, 3H), 3.78 (s, 3H), 1.75 (d, J= 6.0 Hz, 3H). MS (ES+, m/z) 492
(m+1).
Example 128: 5-(5,6-0imethoxy-1H-benzimidazol-1-yl)-3-[(4-
methoxvbenzyl)oxv]thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-ds) d 8.39 (s, 1H), 7.68 (br s, 1H), 7.60 (s, 1H), 7.48 (d, J = 8.8
Hz, 2H), 7.32 (s, 1H), 7.15, (s. 1H), 6.99 (br s. 1H), 6.95 (d, J = 8.8 Hz, 2H), 5.32 (s, 2H),
3.83 (s, 3H), 3.81 (s, 3H), 3.74 (s, 3H). MS (ES+, m/z) 440 (m+l).
Intermediate Example 1: Methyl 5-(1H-benzimidazol-1-vl)-3-(phenylethynyl)-2-
thiophenecarboxylate
Methyl 5-(1 H-benzimidazol-1 -yl)-3-{[(trifluoromethyl)sulfonyl]oxy}-2-
thiophenecarboxylate (0.300 g, 0.738 mmol) was dissolved in 7 mL of N,N-
dimethylformamide with stirring. Triethylamine (0.21 mL, 1.5 mmol) was added via
syringe. Copper (I) iodide (0.0141 g, 0.0740 mmol) was added followed by trans-
dichlorobis(triphenylphosphine) palladium (II) (0.0258 g, 0.0368 mmol).
Phenylacetylene (0.12 mL, 1.1 mmol) was added via syringe, and the mixture was
heated to 80 °C for 16 hours. The mixture was cooled to room temperature and
poured into ethyl acetate and water. The layers were separated, and the organic layer
was washed with brine. The combined aqueous layers were extracted with ethyl
acetate. The combined organic layers were dried over MgSO4, filtered, and
concentrated in vacuo. Purification by flash chromatography afforded 0.212 g (80%)
of methyl 5-(1 H-benzimidazol-1-yl)-3-(phenylethynyl)-2-thiophenecarboxylate. 1H
NMR (300 MHz, DMSO-ds) d 8.76 (s, 1H), 7.85 (m, 2H), 7.80 (s, 1H), 7.64-7.58 (m, 2H),
7.52-7.35 (m, 5H), 3.92 (s, 3H). MS (ES+, m/z) 359 (m+1)..
Example 129: 5-(1 H-Benzimidazol-1 -yl)-3-(phenylethynyflthiophene-2-carboxamide
5-(1 H-benzimidazol-1-yl)-3-(phenylethynyl)thiophene-2-carboxamide was prepared
from methyl 5-(1 H-benzimidazol-1-yl)-3-(phenylethynyl)-2-thiqphenecarboxylate
using procedure similarly described in Example 61 except 7M NH3 in MeOH was used
instead of 2M NH3 in MeOH. 1H NMR (400 MHz, DMSO-d6) d 8.71 (s, 1H), 8.09 (br s,
1H), 7.85-7.80 (m, 2H), 7.72 (s, 1H), 7.67-7.63 (m, 2H), 7.53-7.36 (m, 6H). MS (ES+.
m/z) 344 (m+1).
Intermediate Example 2: Methyl 5-(1H-benzimidazol-1-yl)-3-(2-phenylethyl)-2-
thiophenecarboxylate
Methyl 5-(1H-benzimidazol-1-yI)-3-(phenylethynyl)-2-thiophenecarboxylate (0.110 g,
0.307 mmol) was dissolved in 10 mL of ethyl acetate with stirring. 10% Palladium on
carbon (0.0327 g, 0.0307 mmol) was added, and the reaction placed under 1
atmosphere of hydrogen for 16 hours. The mixture was filtered through celite,
washing with ethyl acetate. The filtrate was concentrated to afford 0.109 g (98%) of
methyl 5-(1H-benzimidazol-1-yl)-3-(2-phenylethyl)-2-thiophenecarboxylate. "H NMR
(300 MHz, DMSO-ds) d 8.67 (s, 1H). 7.80 (d, J - 7.5 Hz, 1H), 7.72 (d, J - 7.2 Hz, 1H),
7.49 (s, 1H), 7.46-7.17 (m, 7H), 3.84 (s, 3H), 3.32 (m, 2H), 2.95 (m, 2H). MS (ES+, m/z)
363 (m+1).
Example 130: 5-(1tf-Benzimidazol-1-yl)-3-(2-phenylethvl)thiophene-2-carboxamide
5-(1 H-benzimidazol-1-yl)-3-(2-phenylethyl)thiophene-2-carboxamide was prepared
from methyl 5-(1 H-benzimidazol-1-yl)-3-(2-phenylethyl)-2-thjophenecarboxylate
using procedure similarly described in Example 61 except 7M NH3 in MeOH was used
instead of 2M NH3 in MeOH. 1H NMR (300 MHz, DMSO-d6) d 8.58 (s, 1H), 7.79 (d, J =
7.3 Hz, 1H), 7.65 (d, J = 8.1 Hz, 1H), 7.56 (br S; 2H), 7.44-7.17 (m, 8H), 3.22 (m, 2H),
2.95 (m, 2H). MS (ES+, m/z) 348 (m+1).
Example 131:.. 5-(1H-Benzimidazol-1-yl)-3-[methyl(phenyl)amino]thiophene-2-
carboxamide
Compound was prepared using procedures similarly described for Example 31 and 61.
1H NMR (300 MHz, DMSO-d6) d 8.65 (s, 1H), 7.83-7.67 (m, 3H), 7.46-7.23 (m. 6H), 6.91-
6.84 (m, 3H), 3.28 (s, 3H). MS (ES+, m/z) 349 (m+1).
Example 132: 5-(1H-Benzimidazol-1 -yl)-3-[(phenylsulfonyl)amirto]thiophene-2-
carboxamide
Compound was prepared using procedures similarly described for Example 32 except
sulfonamide was used instead of benzamide and Example 61. 1H NMR (300 MHz,
DMSO-d6) d 11.40 (s, 1H), 8.75 (s. 1H), 7.95-7.90 (m, 3H), 7.88 (br s, 1H), 7.82 (m, 1H),
7.71 (m, 1H), 7.65-7.58 (m, 3H), 7.51 (s, 1H), 7.45 (m, 1H), 7.40 (m, 1H). MS (ES+, m/z)
399 (m+1).
Intermediate Example 3: Methyl 5-(1H-benzimidazol-1-yl)-3-
({[(phenylmethvl)oxy]carbonyl}amino)-2-thiophenecarboxylate
Methyl 5-(1H-benzimidazol-1-yI)-3-{[(trifluoromethyl)sulfonyl]oxy}-2-
thiophenecarboxylate (1.11 g, 2.73 mmol), cesium carbonate (1.25 g, 3.84 mmol), 2,2"-
bis(diphenylphosphino)-1,1"-binaphthyl (0.0850 g, 0.137 mmol), and
tris(dibenzylideneacetone dipalladium (0) (0.0625 g, 0.0683 mmol) were combined in a
reaction flask with 30 mL of toluene with stirring. Benzyl carbamate (0.495 g, 3.27
mmol) was added, and the reaction was heated to 100 °C for 40 hours. The reaction
was cooled to room temperature, adsorbed directly onto silica gel, and purified by
flash chromatography to afford 0.604 g (54%) of methyl 5-(1H-benzimidazol-1-yl)-3-
({[(phehylmethyl)oxy]carbonyl}amino)-2-thiophenecarboxyIate. 1H NMR (400 MHz,
DMSO-d6) d 9.67 (s, 1H), 8.76 (s, 1H), 8.03 (s, 1H), 7.84-7.77 (m, 2H), 7.49-7.28 (m, 7H),
5.26 (s, 2H), 3.86 (s. 3H). MS (ES+, m/z) 408 (m+1).
Intermediate Example 4: Methyl 5-(1 H-benzimidazol-1-yl)-3-
(([(phenvlmethyl)oxy]carbonyl}{[2-(trjfluQromethyl)phenyl]methvl}amino)-2-
thiophenecarboxylate
Methyl 5-(1H-benzimidazol-1-yl)-3-({[(phenylmethyl)oxy]carbonyl}amino)-2-
thiophenecarboxylate (0.400 g, 0.982 mmol) and cesium carbonate (1.02 g, 3.13 mmol)
were placed in a flask with 12 mL of N,N-dimethylformamide with stirring. 2-
(Trifluoromethyl)benzyl bromide (0.704 g, 2.95 mmol) was added, and the reaction was
stirred for 16 hours. The mixture was poured into water and ethyl acetate, and the
layers were separated. The organic layer was washed with brine, and the combined
aqueous layers were extracted with ethyl acetate. The combined organic layers were
dried over MgSO4, filtered, and concentrated in vacuo. Flash chromatography provided
somewhat impure methyl 5-(1H-benzimidazol-1-yl)-3-({[(phenylmethyl)pxy]-
carbonyl}{[2-(trifluoromethyl)phenyl]methyl}amino)-2-thiophenecarboxylatethat
was carried directly into the next step. MS (ES+, m/z) 567 (m+1).
Intermediate Example 5:5-(1H-benzimidazol-1-yl)-3-({[(phenylmethyl)oxy]carbonyl}-
{[2-(trifluoromethyl)phenyl]methyl|amino)-2-thiophenecarboxylicacid
Methyl 5-(iH-benzimidazol-1-yl)-3-({[(phenylmethyl)oxy]carbonyl}{[2-
(trifluoromethyl)phenyl]methyl}amino)-2-thiophenecarboxylate (0.555 g, 0.982 ramol)
was dissolved in 10 mL of tetrahydrofuran with stirring. 10 mL of 1N LiOH solution
was added, and the mixture was allowed to stir for 16 hours. The mixture was poured
into diethyl ether and water, and the layers were separated. The organic layer was
washed with water, and the diethyl ether layer was subsequently discarded. The
combined aqueous layers were acidified to pH ~ 2 with concentrated HCI and
extracted with ethyl acetate three times. The combined organic layers were dried over
MgSO4. filtered, and concentrated in vacuo to afford 0.528 g (97%) of 5-(1 H-
benzimidazol-1 -yl)-3-({[(phenylmethyl)oxy]carbonyl} {[2-
(trifluoromethyl)phenyl]methyl}amino)-2-thiophenecarboxyiic acid as an off-white
solid. 1H NMR (400 MHz. DMSO-d6) d 13.60 (br s, 1H), 8.58 (s, 1H), 7.85 (d. J - 7.7 Hz,
1H), 7.76 (d, J - 7.0 Hz, 1H), 7.70 (d, J = 7.7 Hz, 1H), 7.65 (dd, J - 7.7. 7.7 Hz, 1H), 7.54-
7.43 (m, 4H), 7.41-7.24 (m, 6H), 5.14 (s, 2H), 5.14 (s, 2H). MS (ES+, m/z) 552 (m+1).
Intermediate Example 6: Benzyl 2-(aminocarbonvl)-5-(1H-benzimidazol-1-yl)thienr-3-
yl[2-(trifluoromethyl)-benzyl]carbamate
5-(1H-Benzimidazol-1-yl)-3-({[(phenylmethyl)oxy]carbonyl}{[2-
(trifluordmethyl)phenyl]methyl}amino)-2-thiophenecarboxylic acid (0.200 g, 0.363
mmol) and ammonium chloride (0.0388 g, 0.725 mmol) were added to a flask with 5
mL of N,N-dimethylformamtde with stirring. N-Methylmorpholine (0.080 mL, 0.73
mmol) was added via syringe. 1-Hydroxybenzotriazole (0.0981 g. 0.726 mmol) was
added followed by 1-(3-dimethylaminopropy!)-3-ethylcarbodiimide hydrochloride
(0.0974 g, 0.508 mmol). The mixture was stirred for 16 hours and poured into ethyl
acetate and 1N HCI. The layers were separated; and the organic layer was washed
with brine. The combined aqueous layers were extracted with ethyl acetate. The
combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo.
Purification by flash chromatography afforded 0.171 g (86%) of benzyl 2-
(aminocarbonyl)-5-(1H-benzimidazol-1-yl)thien-3-yl[2-(trifluoromethyl)-
benzyl)carbamate as an off-white solid. AH NMR (400 MHz. DMSO-d6) 5 8.53 (s, 1H),
7.85-7.75 (m, 2H), 7.75-7.69 (m, 3H), 7.66 (dd, J = 7.4, 7.4 Hz, 1H), 7.50 (dd, J = 7.5,7.5
Hz, 1H), 7.47-7.27 (m, 9H), 5.16 (s, 2H), 5.11 (br s, 2H). MS (ES+. m/z) 551 (m+1).
Example 133:5-(1 H-Benzimidazol-l-yl)-3-{[2-(trtfluoromethyl)benzyl]amtno}
thiophene-2-carboxamide
Phenylmethyl [2-(aminocarbonyl)-5-(1 H-benzimidazol-1 -yl)-3-thienyl] {[2-
(trifluoromethyl)phenyl]methyl}carbamate (0.157 g, 0.285 mmol) was dissolved in 10
mL of ethyl acetate with stirring. 10% Palladium on carbon (0.0606 g, 0.0570 mmol)
was added, and the solution was placed under 1 atmosphere of hydrogen. The
reaction was stirred for 48 hours and was judged incomplete. The reaction mixture
was filtered through a celite pad and washed with ethyl acetate. The filtrate was
concentrated in vacuo and purified by flash chromatography to afford 0.0257 g (22%)
of pure product and 0.112 mg of a mixture of starting material and product. 1H NMR
(400 MHz, DMSO-d6) d 8.61 (s, 1H). 8.16 (dd. J= 6.6. 6.6 Hz, 1H), 7.81-7.74 (m, 2H),
7.73-7.67 (m. 2H), 7.64 (d, J= 7.7 Hz, 1H), 7.50 (dd, J= 7.5, 7.5 Hz, 1H), 7.43-7.33 (m,
2H), 7.19 (s, 1H), 7.14 (br s, 2H), 4.71 (d, J - 6.2 Hz, 2H). MS (ES+, m/z) 417 (m+1).
Intermediate Example 7: 2-(Methyloxv)-5-nitrophenyl 2,2-dimethytpropanoate
Commercially available 2-Methoxy-5-nitrophenol (10.0 g, 59.1 mmol) was dissolved in
150 mL of dichloromethane with 4-dimethylaminopyridine (0.722 g, 5.91 mmol),
Triethylamine (9.88 mL, 70.9 mmol) was added via syringe. Pivaloyl chloride (8.01 mL,
65.0 mmol) was added slowly via syringe. The reaction was stirred for ten minutes and
poured into 1N HCI. The layers were separated, and the aqueous layer was washed
with dichloromethane. The combined organic layers were washed with saturated
NaHC03 and brine. The organic layers were dried over MgS04, filtered, and
concentrated in vacuo. The isolated solid was triturated with hexanes, filtered, and
washed with hexanes and 2-methylbutane. The solid was air dried and collected to
afford 13.0 g (87%) of 2-(methyloxy)-5-nitrophenyl 2,2-dimethylpropanoate. 1H NMR
(400 MHz. CDCI3) 5 8.16 (dd, J - 9.2,2.8 Hz, 1H), 7.95 (d, J - 2.8 Hz, 1H). 7.01 (d, J -
9.2 Hz, 1H). 3.92 (s, 3H), 1.38 (s, 9H).
Intermediate Example 8: 5-Amino-2-(methyloxy)phenvl 2.2-dimethylpropanoate
(Methyloxy)-5-nitrophenyl 2,2-dimethylpropanoate (13.0 g, 51.4 mmol) was
dissolved in 150 mL of ethyl acetate with stirring. 10% Palladium on carbon (1.64 g,
1.54 mmol) was added and the solution was stirred under 1 atmosphere of hydrogen
for 16 hours. The reaction was filtered through celite and washed well with ethyl
acetate. The filtrate was concentrated in vacuo to afford 11.3 g (98%) of 5-amino-2-
(methyloxy)phenyl 2,2-dimethylpropanoate as a pink solid. "H NMR (400 MHz, CDCI3)
5 6.80 (d, J - 8.6 Hz, 1H), 6.55 (dd, J = 8.6, 2.8 Hz, 1H), 6.45 (d. J = 2.8 Hz, 1H), 3.73 (s,
3H),1.35(s,9H).
Intermediate Example 9: 2-(Methvloxv)-4-nitro-5-[(trifluoroacetyl)amino]phenyl 2,2-
dimethylpropanoate
5-Amino-2-(methyloxy)phenyl 2,2-dimethylpropanoate (10.53 g, 47.1 mmol) was
dissolved in 200 mL of chloroform with stirring. Ammonium nitrate (6.79 g, 84.8
mmol) was added in a single portion. The mixture was cooled to 0 °C, and
trifiuoroacetic anhydride (36 mL, 260 mmol) was added dropwise via addition funnel
over 1 hour. The reaction was warmed to room temperature and stirred for an
additional six hours. The reaction was quenched by the careful addition of 100 mL of
saturated NaHCO3 and stirred for 15 minutes. The mixture was poured into a
separatory funnel, and the layers were separated. The aqueous layer was washed with
dichloromethane. The combined organic layers were dried over MgSO4, filtered, and
concentrated in vacuo to provide 16.5 g (96%) of 2-(methyloxy)-4-nitro-5-
[(trifluoroacetyl)amino]phenyl 2,2-dimethylpropanoate as a yellow solid. 1H NMR (400
MHz, CDCI3) d 11.36 (br s, 1H), 8.49 (s, 1H), 7.84 (s, 1H), 3.91 (s, 3H), 1.38 (s, 9H).
termediate Example 10: 5-Amino-2-(methyloxy)-4-nitrophenol
2-(Methyloxy)-4-nitro-5-[(trifluoroacetyl)amino]phenyl 2,2-dimethylpropanoate (16.5
g, 45.2 mmol) was dissolved in 200 mL of methanol-and 200 mL of water with stirring.
Potassium carbonate (31.2 g, 226 mmol) was added and the solution was stirred for
sixteen hours at room temperature. At this point the reaction was judged incomplete
and heated to reflux for two hours. The mixture was cooled to room temperature, and
the majorityof the methanol was removed in vacuo. Ethyl acetate was added, and the
pH of the solution was adjusted to approximately 7 using concentrated HCI. The
layers were separated, and the organic layer was washed with brine. The combined
aqueous layers were saturated with NaCI and further extracted with ethyl acetate (2X)
and 20% isopropanol in ethyl acetate (4X). The combined organic extracts were dried
over MgSO4, filtered, and concentrated in vacuo. The isolated solid was triturated with
diethyl ether, filtered, and washed with diethyl ether and 2-methylbutane. The orange
solid was air dried and collected to yield 7.15 g (86%) of 5-amino-2-(methyloxy)-4-
nitrophenol. 1H NMR (400 MHz. CDCl3) 5 10.66 (br s, 1H), 7.36 (br s, 2H), 7.32 (s, 1H),
6.36 (s, 1H), 3.73 (s, 3H). MS (ES+, m/z) 185 (m+1).
Intermediate Example 11: 5-{[(1,1-Dimethylethvl)(diphenyl)silyl]oxy}-4-(methyloxy)-
2-nitroaniline
5-Amino-2-(methyloxy)-4-nitropheno! (6.90 g, 37.5 mmol) was dissolved in 100 mL of
acetonitrile with stirring. Triethylamine (6.30 mL, 45.2 mmol) was added via syringe.
t-Butylchlorodiphenylsilane (9.75 mL, 37.5 mmol) was added slowly via syringe. The
reaction was stirred for 2 hours and judged incomplete. Additional triethylamine (1.57
mL, 11.3 mmol) and t-butylchlorodiphenylsilane (2.94 mL, 11.3 mmol) were added via
syringe. The reaction was stirred an additional 15 minutes and poured into ethyl
acetate and 1N NaOH. The layers were separated, and the organic layer was washed
with brine. The combined aqueous layers were extracted with ethyl acetate. The
combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo.
The isolated material was passed through a plug of silica gel, and the fractions
containing product concentrated. The isolated viscous oil was a mixture of
unidentified silyl byproducts and 5-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-4-
(methyloxy)-2-nitroaniline. The yield was not determined, and the impure material
carried forward to the next step. MS (ES+, m/z) 423 (m+1).
Intermediate Example 12: [5-U(1,1-Pimethytethyl)(diphenyl)silyl]oxy}-4-(methyloxy)-
2-nitrophenyl]formamide
Acetic anhydride (17.7 mL, 188 mL) was slowly added to formic acid (14.1 mL, 374
mmot) with stirring. The mixture was placed in a 50 °C oil bath for one hour. After
cooling to room temperature, the impure material containing 5-{[(1,1-
dimethylethyl)(diphenyl)-silyl]oxy}-4-(methyloxy)-2-nitroaniline was dissolved in 100
mL of diehloromethane and added to the reaction. The reaction was allowed to stir
for 14 hours and quenched by the careful addition of 100 mL of water. The reaction
was slowly poured into saturated NaHC03 and diehloromethane. The layers were
separated, and the aqueous layer washed with diehloromethane. The combined
organic layers were dried over MgS04, filtered; and concentrated to provide impure
material containing [5-{[(1,1-dimethylethyl)(diphenyl)-silyl]oxy}-4-(methyloxy)-2-
nitrophenyl]formamide. The yield was not determined, and the impure material
carried forward to the next step. MS (ES+, m/z) 451 (m+1).
Intermediate Example 13: [2-Amino-5-{[(1,1-dimethylethyl)(diphenyl)silvl]oxy}-4-
(methyloxy)phenyl]-formamide
The impure material containing [5-{[(1,1-dimethylethyl)(diphenyl)-silyl]oxy}-4-
(methyloxy)-2-nitrophenyl]formamide was dissolved in 200 mL of ethyl acetate with
stirring. 10% Palladium on carbon (1.20 g, 1.13 mmol) was added, and the mixture
was placed under 1 atmosphere of hydrogen for 24 hours. The reaction was filtered
through celite washing with ethyl acetate and chloroform. The filtrate was
concentrated Invacuo to afford impure material containing [2-amino-5-{[(1,1-
dimethylethyl)(diphenyl)-silyl]oxy}-4-(methyloxy)phenyl]-formamide. The yield was
not determined, and the impure material carried forward to the next step. MS (ES+,
m/z) 421 (m+1).
Intermediate Example 14: 5-{[(1.1-Dimethvlethvl)(diphenvl)silyl]oxv}-6-(methvloxv)-
1 H-benzimidazole
The impure material containing [2-amino-5-{[(1,1-dimethylethyl)(diphenyl)-
silyl]oxy}-4-(methyloxy)phenyll-formamide was dissolved in 200 mL of chloroform
with stirring. Magnesium sulfate (13.54 g, 112 mmol) was added in a single portion.
pyridinium p-toluenesulfonate (11.3 g, 45.0 mmol) was added, and the reaction was
allowed to stir for 16 hours. The reaction was judged incomplete, therefore the
mixture was heated to between 40 and 50 °C for 8 additional hours. The reaction was
cooled to room temperature and solid NaHC03 (10g) was added. The mixture was
stirred for 30 minutes and filtered to remove all solid particles. The filtrate was
concentrated to approximately 100-200 mL total volume at which point significant
solid formation had occurred. 200 mL of diethyl ether and hexanes (1:1) was added,
and the mixture was filtered. The solid was washed with hexanes and 2-
methylbutane. The solid was air dried, collected, and determined to be the tosyl salt
of the desired product The solid was placed in a separatory funnel with 1N NaOH and
extracted twice with isopropanol in dichloromethane (1:4). The combined organic
layers were dried over MgSO4, filtered, and concentrated in vacuo to afford 7.80 g
(52% over 4 steps) of 5-{[(1,1-dimethylethylKdiphenyl)silyl]oxy}-6-(methyIoxy)-1 H-
benzimidazole. 1H NMR (400 MHz, DMSO-d6) d 12.00 (br s, 1H), 7.93 (s, 1H), 7.72-7.67
(m, 4H). 7.49-7.39 (m. 6H), 7.07 (s, 1H), 6.78 (s. 1H), 3.65 (s, 3H), 1.07 (s. 9H). MS (ES+,
m/z) 403 (m+1).
Intermediate Example 15: Methyl 5-[6-{[(1.1-dimethylethyl)(diphenyl)silyl]oxy}-5-
(methyloxy)-1 H-benzimidazol-1-yl]-3-hydroxv-2-thiophenecarboxylate and Methyl 5-
[5- {[(1,1 -dimethyiethyl)(diphenyl)silyl]oxy}-6-(methvloxy)-1 H-benzimidazol-1 -yl]-3-
hvdroxv-2-thiophenecarboxylate
5-{[(1,l-Dimethylethyl)(diphenyl)silyl]oxy}-6-(methyloxy)-1 H-benzimidazole (4.12 g.
10.2 mmol) was dissolved in 50 mL of chloroform with stirring. Methyl 2-chloro-3-
oxo-2,3-dihydro-2-thiophenecarboxylate (0.982 g, 5.10 mmol) was added in a single
portion. The reaction was allowed to stir for 5 days. 50 ml of water was added, and
the pH was adjusted to approximately 6-7 using saturated NaHCO3 The layers were,
separated, and the aqueous layer was extracted with dichloromethane (1X) and ethyl
acetate (1X). The combined organic layers were dried over MgSO)4, filtered, and
concentrated in vacuo. The residue was purified by flash chromatography to afford
2.40 g (84%) of a 1.2-1.4:1 regioisomeric mixture of methyl 5-[6-{[(1,1-
dimethylethyl)(diphenyl)silyl]oxy}-5-(methyloxy)-1H-benzimidazol-1-yl]-3-hydroxy-
2-thiophenecarboxylate and methyl 5-[5-{[(1,1-dimethYlethyl)(diphenYl)silylloxy}-6-
(methyloxy)-1H-benzimidazol-1-yl]-3-hydroxy-2-thiophenecarboxylate. 1H NMR (300
MHz, DMSO-d6) d 10.84,10.74 (br s, 1H), 8.50, 8.42 (s, 1H), 7.76-7.69 (m, 4H), 7.54-
41 (m, 6H). 7.36,7.23 (s, 1H), 7.13, 7.00 (s. 1H), 6.93, 6.91 (s, 1H), 3.86.3.82 (s, 3H),
3.744,3.737 (s, 3H), 1.12,1.11 (s, 9H). MS (ES+, m/z) 403 (m+1).
Intermediate Example 16: Methyl 57.[6-{[(1,1-dimethvlethyl)(diphenyl)silyl]oxyl-5-
(methyloxy)-1 H-benzimidazol-1 -yl]-3-({ [2-(trif luoromethy0phenyl]methyl}oxy)-2-
thiophenecarboxylate and Methyl 5-[5-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-6-
(methyloxv)-1H-benzimidazol-1-vl]-3-({[2-(trifluoromethvl)phenyl]methyl}oxy)-2-
thiophenecarboxylate
A regioisomeric mixture of methyl 5-[6-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-5-
(methyloxy)-1H-benzimidazol-1 -yl]-3-hydroxy-2-thiophenecarboxylate and methyl 5-
[5-{[(1,1 -dimethylethyl)(diphenyl)silyl]oxy}-6-(methyloxy)-1H-benzimidazol-1 -yl]-3-
hydroxy-2-thiophenecarboxylate (3.97 g, 7.11 mmol) was dissolved in 40 mL of N,N-
dimethylformamide with stirring. Potassium carbonate (1.18 g, 8.54 mmol) was added
in a single portion. 2-(Trifluoromethyl)benzyl bromide (2.04 g, 8.53 mmol) was added
in a single portion. The reaction was allowed to stir for 16 hours and poured into
water and ethyl acetate. The layers were separated, and the organic layer was washed
with brine. The combined aqueous layers were extracted with ethyl acetate. The
combined organic layers were dried over MgS04, filtered, and concentrated in vacuo.
Purification by flash chromatography afforded 2.65 g (52%) of methyl 5-[5-{[(1,1-
dimethylethyl)(diphenyl)-silyl]oxy}-6-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy)r2-thiophenecarboxylate and 2.13 g (42%) of
methyl 5-[6-{[(1,1 -dimethylethyl)(diphenyl)silyl]oxy}-5-(methyloxy)-1 H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate. Data for (5-OTBDPS, 6-OMe): 1H NMR (300 MHz, DMSO-d6) d
8.47 (s, 1H), 7.98 (d, J= 7.4 Hz, 1H), 7.88-7.60 (m, 8H), 7.55-7.42 (m, 6H), 7.25 (s, 1H),
6.94 (s, 1H), 5.54 (s, 2H). 3.81 (s, 3H), 3.73 (s, 3H), 1.11 (s, 9H). MS (ES+, m/z) 717
(m+1). Data for (5-OMe, 6-OTBDPS): 1H NMR (300 MHz, DMSO-d6) d 8.57 (s, 1H), 7.98
(d, J m 7.6 Hz, 1H), 7.88-7.79 (m, 2H), 7.76-7.61 (m. 5H), 7.56 (s, 1H), 7.51-7.41 (m, 6H),
7.37 (s, 1H), 7.05 (s, 1H). 5.43 (s, 2H), 3.84 (s, 3H), 3.74 (s, 3H), 1.12 (s, 9H). MS (ES+,
m/z) 717 (m+1).
Intermediate Example 17: Methyl 5-[5-hydroxv-6-(methyloxy)-1H-benzimidazol-1-
yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiopheneearboxylate
Methyl 5-[5-{[(1,1-dimethylethyl)(diphenyl)-silyl]oxy}-6-(methyloxy)-1H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenylI-methyl}oxy)-2-
thiophenecarboxylate (1.54 g, 2.15 mmol) was dissolved in 20 mL of tetrahydrofuran
with stirring. The solution was cooled to 0 °C, and tetrabutylammonium fluoride (3.20
mL, 1.0M in THF, 3.20 mmol) was added slowly via syringe. The reaction was stirred for
ten minutes and quenched by the addition of 50 mL of 0.5N HCI. The mixture was
poured into ethyl acetate, and the layers were separated. The organic layer was
washed with brine, and the combined aqueous layers were extracted with ethyl
acetate. The combined organic layers were dried over MgSO4, filtered, and
concentrated in vacuo. Purification by flash chromatography provided 0.761 g (74%)
of methyl 5-[5-hydroxy-6-(methyloxy)-1 H-benzimidazol-1-yl]-3-({[2-
(tritluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate as an off-white solid;
1H NMR (400 MHz, DMSO-d6) d 9.07 (s, 1H), 8.47 (s, 1H), 7.96 (d, J= 7.9 Hz, 1H), 7.84-
7.76 (m. 2H), 7.65 (s, 1H), 7.62 (dd,J = 7.9. 7.7 Hz, 1H), 7.24 (s, 1H), 7.13 (s, 1H), 5.53 (s,
2H), 3.86 (s,3H), 3.78 (s,3H).
Intermediate Example 18: Methyl 5-[6-hydroxy-5-(methyloxy)-1/7-benzimidazol-1-
yl]-3-({[2-(trifluoromethyl)phenvl]methvl)oxy)-2-thiophenecarboxvlate
This compound was prepared in a similar manner to that previously described for the
synthesis of methyl 5-[5-hydroxy-6-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy)-2-thiophenecarboxylate. Reaction of methyl 5-
[6-{[(1,1-dimethylethyl)(diphenyl)silylloxy}-5-(methyloxy)-1H-benzimidazol-1-yl]-3-
({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate (3.22 gf 4.49 mmol)
with tetrabutylammonium fluoride (6.74 mL, 1.0M in THF, 6.74 mmol) afforded 1.76 g
(82%) of methyl 5-[6-hydroxy-5-(methYloxy)-1H-benzimidazol-1-yll-3-({[2-
(trifluoromethyl)-phenyl]methyl}oxy)-2-thiophenecarboxylate as a pale yellow solid.
1H NMR (400 MHz. DMSO-d6) d 8.72 (s, 1H). 7.98 (d, J= 7.7 Hz, 1H), 7.85-7.77 (m, 2H),
7.72 (s, 1H), 7.62 (dd, J= 7.9,7.7 Hz, 1H), 7.32 (s, 1H), 7.30 (s, 1H), 5.50 (s, 2H), 3.86 (s,
3H),3.78(s,3H).
Intermediate Example 19:5-([(1,1-Dimethylethyl)(diphenvl)silvl1oxy)-1H-
benzimidazole
This compound was prepared in four steps from commercially available 4-amino-3-
nitrophenol using a procedure similar to that outlined for the synthesis of 5-{[(1,1-
dimethylethyl)(diphenyl)silyl]oxy}-6-(methyloxy)-1H-benzimidazole. 1H NMR (400
MHz, DMSO-d6) d 12.15 (br s, 1H), 8.03 (s, 1H), 7.74-7.67 (m, 4H). 7.51-7.39 (m. 6H),
7.37 (d, J = 8.6 Hz. 1H). 6.81 (d, J = 2.2 Hz. 1H), 6.75 (dd, J - 8.6. 2,2 Hz. 1H), 1.05 (s,
9H). MS(ES+,m/z)373(m+1).
Intermediate Example 20: Methyl S-(6-{[(1,1-dimethvlethyl)(diphenyl)silyl]oxy}--1H-
benzimidazol-1 -yl)-3-hydroxy-2-thiopheneearboxylate-and Methyl 5-(5-{[(1.1 -
dimethylethyl)-(diphenvl)silyl]oxy}-1H-benzimidazol-1-yl)-3-hydroxy-2-
thiophenecarboxylate
5-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-6-(methyloxy)-1H-benzimidazole (9.43 g,
25.3 mmol) was dissolved in 125 mL of chloroform with stirring. Methyl 2-chloro-3-
oxo-2,3-dihydro-2-thiophenecarboxylate (2.44 g, 12.7 mmol) was added in a single
portion. The reaction was allowed to stir for 10 days. 100 mL of water was added*,
and the pH was adjusted to approximately 6-7 using saturated NaHCO3. The layers
were separated, and the aqueous layer was extracted with dichloromethane (1X) and
ethyl acetate (1X). The combined organic layers were dried over MgSO4, filtered, and
concentrated in vacuo. The residue was purified by flash chromatography to afford
5.48 g (82%)of a 1.0-1.1:1 regioisomeric mixture of methyl 5-(6-{[(1,1-
dimethylethyi)(diphenyl)sHyl]oxy}-1 H-benzimtdazol-1-y!)-3-hydroxy-2-
thiophenecarboxylate and methyl 5-(5-{[(i,1-dimethylethyl)-(diphenyl)silyl]oxy}-1 H-
benzimidazol-1-yl)-3-hydroxyr2-thiophenecarboxylate. 1H NMR (300 MHz, DMSO-d6)
d 10.85,10.78 (br s, 1H), 8.59,8.54 (s, 1H), 7.78-7.70 (m, 4H), 7.64,7.60 (dd, J - 8.8,0.6
Hz and d, J - 8.8 Hz, 1H), 7.56-7.43 (m, 6H), 7.10,6.96 (s, 1H), 7.05-6.88 (m, 2H), 3.85,
3.81 (s, 3H), 1.11,1.09 (s, 9H). MS (ES+, m/z) 529 (m+1).
termediate Example 21: Methyl 5-(6-{[(1,1-dimethylethyl)(diphenyl)silyl]oxyl-1H-
benzimidazol-1-yl)-3-({[2-(trtfluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate and Methyl 5-(5-{[(1,1-dimethvlethvl)(diphenyl)silyl]oxy}-1H-
ben2imidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate
Polystyrene triphenylphoshine (9.84 g, 1.58 mmol/gram, 15.5 mmol) was stirred in 100
mL of dichloromethane for ten minutes. The regioisomeric mixture of methyl 5-(6-
{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-1H-benzimidazol-1-yl)-3-hydroxy-2-
thiophenecarboxylate and methyl 5-(5-{[(1,1-dimethylethyl)-(diphenyl)silyl]oxy}-1 H-
benzimidazol-1-yl)-3-hydroxy-2-thiophenecarboxylate (5.48 g, 10.4 mmol) was added
in a single portion. 2-(TrifIuromethyl)benzyl alcohol (1.68 mL, 12.6 mmol) was added
via syringe, and the solution was cooled to 0°C. Di-Tert-butyl azodicarboxylate (3.58
g, 15.5 mmol) was dissolved in 20 ml of dichloromethane and added dropwise via
addition funnel. The reaction was warmed to room temperature and stirred for 1.5
hours. The mixture was filtered through filter paper, and the solid was washed with
dichloromethane and methanol. The filtrate was concentrated and purified by flash
ehromatography to afford 2.89 g (41%) of methyl 5-(5-{[(1,1-
dimethylethyl)(diphenyl)silyl]oxy}-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate and 2.69 g (38%) of
methyl 5-(6-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-1 H-benzimidazoi-1 -yl)-3-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate. Data for 5-OTBDPS
regioisomer: 1H NMR (300 MHz, DMSO-d6) d 8.66 (s. 1H), 7.98 (d, J= 7.6 Hz, 1H). 7.86-
7.60 (m. 9H), 7.56-7.44 (m. 6H), 7.01 (s, 1H). 6.99 (ddrJ= 6.7, 2.4 Hz, 1H), 5.51 (s, 2H),
3.79 (s, 3H), 1.10 (s, 9H). MS (ES+, m/z) 687 (m+1). Data for 6-OTBDPS regioisomer:
1H NMR (300 MHz, DMSO-d6) d 8.60 (s. 1H), 7.99 (d, J= 7.6 Hz, 1H), 7.87-7.57 (m, 9H),
54-7.42 (m, 6H), 7.07 (d, J - 2.0 Hz, 1H), 6.92 (dd, J = 8.8, 2.3 Hz, 1H), 5.46 (s, 2H),
3.84 (s, 3H), 1.11 (s, 9H). MS (ES+. m/z) 687 (m+1).
Intermediate Example 22: Methyl 5-(6-hydroxy-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethyl)-phenvl]methyl)oxy)-2-thiophenecarboxylate
This compound was prepared in a similar manner to that previously described for the
synthesis of methyl 5-[5-hydroxy-6-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenYl]-methyl}oxy)-2-thiophenecarboxylate. Reaction of methyl 5-
(6-{[(1,1-dimethylethyl)(diphenyl)silyl]oxy}-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethyl)-phenyl]methyl}oxy)-2-thiophenecarboxylate (2.69 g, 3.92 mmol)
with tetrabutylammonium fluoride (5.9 mL, 1.0M in THF, 5.9 mmol) afforded 1.42 g
(81%) of methyl 5-(6-hydroxy-1H-benzimidazol-1-yl)-3-({[2-(trifluoromethyl)-
phenyl]methyl}-oxy)-2-thiophenecarboxylate as an off-white solid. 1H NMR (300
MHz, DMSO-d6) d 9.72 (s, 1H), 8.60 (s, 1H), 8.01 (d, J - 7.7 Hz, 1H), 7.89-7.79 (m, 2H),
7.75 (s. 1H), 7.67 (d, J = 7.7 Hz, 1H), 7.62 (d, J - 8.7 Hz, 1H), 7.27 (d, J = 1.8 Hz, 1H),
6.87 (dd, 7= 8.7, 2.2 Hz, 1H), 5.53 (s, 2H), 3.81 (s, 3H).
Intermediate Example 23: Methyl 5-(5-hydroxy-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethvl)-phenyl]methyl}oxy)-2-thiophenecarboxylate
This compound was prepared in a similar manner to that previously described for the
synthesis of methyl 5-[5-hydroxy-6-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy)-2-thiophenecarboxylate. Reaction of methyl 5-
(5-{[(1,1-dimethylethyl)(diphenyl)sllyl]oxy}-1H-benzimidazol-1-yl)-3-({[2-
(trifluoromethyl)-phenyl]methyl}oxy)-2-thiophenecarboxylate (2.89 g, 4.21 mmol)
with tetrabutylammonium fluoride (6.3 mL, 1.0M in THF, 6.3 mmol) afforded 1.56 g
(83%) of methyl 5-(5-hydroxy-1H-benzimidazol-1-yl)-3-({[2-(trifluoromethyl)-
phenyl]methyl}-oxy)-2-thiophenecarboxylate as an off-white solid. 1H NMR (400
MHz, DMS0-d6) d 9.46 (s, 1H), 8.64 (s, 1H), 7.97 (d, J= 7.0 Hz, 1H), 7.86-7.76 (m, 2H),
7.72-7.59 (m, 3H), 7.09 (s, 1H), 6.92 (d, J = 8.1 Hz, 1H), 5.51 (s, 2H), 3.77 (s, 3H).
Intermediate Example 24: Methyl 5T(6-(methyloxy)-5-{[3-(2-oxo-1-
pvrrolidinyl)propvlloxvl-1H-behzimidazol-1-yl)-3-({[2-(trifluoromethyl)-
phenyl]methvl}oxv)-2-thiophenecarboxylate
Polystyrene-triphenylphosphine (0.397 g, 1.58 mmol/gram, 0.627 mmol) was placed in
a flask with 6 mL of dichloromethane and stirred for 5 minutes. 5-[5-Hydroxy-6-
(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate (0.150 g, 0.314 mmol) was added in a single portion. 1-(3-
Hydroxypropyl)pyrrolidinone (0.059 mL, 0.412 mmol) was added via syringe, and the
mixture was cooled to 0 °C. Di-tert-butyl azodicarboxylate (0.144 g, 0.625 mmol) was
dissolved in 1 mL dichloromethane and added dropwise via syringe. The reaction was
warmed to room temperature and stirred for 1.5 hours. The reaction was filtered
through filter paper and washed with dichloromethane and methanol. The filtrate
was concentrated in vacuo and purified by flash chromatography to afford 0.152 g
(80%) of methyl 5-(6-(methyloxy)-5-{[3-(2-oxo-1-pyrrolidinyl)propyl]oxy}-1H-
benzimidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate. 1H NMR (400 MHz, DMSO-d6) d 8.52 (s, 1H), 7.97 (d, J= 7.9 Hz,
1H), 7.84-7.77 (m, 2H), 7.68 (s, 1H), 7.62 (dd, J - 7.3,7.3 Hz, 1H), 7.34 (s, 1H), 7.28 (s.
1H), 5.54 (s, 2H), 4.02 (t, J = 6.3 Hz, 2H), 3.86 (s, 3H), 3.79 (s, 3H), 3.41 -3.29 (m, 4H),
2.21 (t. J = 8.1 Hz, 2H), 1.99-1.88 (m, 4H). MS (ES+, m/z) 604 (m+1).
example 134: 5-(6-(Methyloxy)-5-{[3-(2-oxo-1 -pyrrolidinvl)propvl]oxy}-1 H-
benzimidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxy-2-
thiophenecarboxamide
5-(6-(methyloxy)-5-{[3-(2-oxo-1-pyrrolidinyl)propy1]oxy}-1H-benzimidazol-1-yl)-3-
({[2-(trifluoromethyl)phenyllmethyl}oxY)-2-thiophenecarboxamide was prepared
from methyl 5-(6-(methyloxy)-5-{[3-(2-oxo-t-pyrrolidinyl)propyl]oxy}-1H-
benzimidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxylate using procedure similarly described in Example 61 except 7M
NH3 in MeOH was used instead of 2M NH3 in MeOH. 1H NMR (400 MHz, DMSO-d6) d
8.42 (s, 1H), 7.86-7.60 (m, 5H), 7.59 (s, 1H), 7.30 (s, 1H), 7.20 (s, 1H), 6.80 (br s, 1H),
5.54 (s, 2H), 3.99 (t, J - 6.2 Hz, 2H), 3.82 (s, 3H). 3.39-3.28 (m, 4H), 2.18 (t. J - 8.1 Hz,
2H), 1.97-1.85 (m, 4H). MS (ES+, m/z) 549 (m+1).
Intermediate Example 25: Methyl 5-[6-{[3-(dimethylamino)propyl]oxy}-5-
(methyloxv)-1H-benzimidazol-1-yll-3-({[2-(triftuoromethvl)phenyl]methyl}oxy)-2-
thiophenecarboxylate
Methyl 5-[6-hydroxy-5-(methyloxy)-1 H-benzimidazol-1-yl]-3-({[2-(trifluoromethyl)-
phenyl]methyl}oxy)-2-thiophenecarboxylate (0.150 g, 0.313 mmol) and
triphenylphosphine (0.361 g, 1.38 mmol) were stirred in 6 mL of dichloromethane. 3-
Dimethylamino-1-propanol (0.13 mL, 1.1 mmol) was added via syringe, and the
solution was cooled to 0 gC. Diethyl azodicarboxylate (0.12 mL, 0.76 mmol) was added
dropwise via syringe, and the solution was warmed to room temperature. After 3
hours the reaction was quenched by the addition of 2-3 mL of methanol. The reaction
nature was absorbed directly onto silica gel, and purification by flash
chromatography afforded 0.112 g (63%) of methyl 5-[6-{[3-
(dimethylamino)propyl]oxy}-5-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate. 1H NMR (400 MHz,
DMSO-d6) d 8.51 (s, 1H), 7.97 (d, J = 7.7 Hz, 1H), 7.84-7.77 (m, 2H), 7.67 (s, 1H), 7.62
(dd, J = 7.5, 7.5 Hz, 1H), 7.35 (s, 1H), 7.29 (s, 1H). 5.53 (s, 2H), 4.06 (t, J - 6.4 Hz, 2H),
3.84 (s, 3H), 3.79 (s, 3H), 2.47 (t, J - 7.0 Hz, 2H), 2.21 (s, 6H), 1.91 (m, 2H). MS (ES+,
m/z) 564 (m+1).
Example 135: 5-[6-{[3-(Dimethvlamino)propyl]oxy)-5-(methyloxy)-1 H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxamide
5-[6-[3-(dimethylamlno)propyl]oxy}-5-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyI)phenyl]methyl}oxy)-2-thiophenecarboxamide was prepared from
methyl 5-[6-{[3-(dimethylamino)propylloxy}-5-(methyloxy}-1H-benzimidazol-1-yl]-
3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylate using procedure
similarly described in Example 61 except 7M NH3 in MeOH was used instead of 2M
NH3 in MeOH.
1H NMR (400 MHz, DMSO-d6) d 8.42 (s, 1H), 7.88-7.60 (m, 5H), 7.59 (s, 1H), 7.32 (s, 1H),
7.20 (s, 1H), 6.8 (br s, 1H), 5.53 (s, 2H), 4.02 (t, J - 6.3 Hz, 2H), 3.81 (s, 3H), 2.35 (t. J =
7.0 Hz, 2H), 2.11 (s, 6H), 1.86 (m, 2H). MS (ES+, m/z) 549 (m+1).
intermediate Example 26: Methyl 5-[6-[(2-chloroethyl)oxv3-5-(methyloxy)-1H-
benzimidazol-l-yl]-3-({[2-(trifluoromethvl)phenyllmethyl}oxy)-2-
thiophenecarboxylate
This compound was prepared in a similar manner to that previously described for the
synthesis of methyl 5-[6-{[3-(dimethylamino)propyl]oxy}-5-(methyloxy)-1 H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]methyI}oxy)-2-
thiophenecarboxylate Reaction of methyl 5-[6-hydroxy-5-(methy|oxy)-1H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)-phenyl]methyl}oxy)-2-
thiophenecarboxylate (0.150 g,0.313 mmpl), triphenylphosphine (0.740 g, 2.82 mmol),
2-chloroethanol (0.13 mL, 1.9 mmol), and diethyl azodicarboxylate (0.25 mL, 1.6
mmol) provided 0.117 g (69%) of methyl 5-[6-[(2-chloroethyl)oxy]-5-(methyloxy)-1H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]-methyl}oxy)-2-
thiophenecarboxylate. 1H NMR (400 MHz, DMSO-d6) d 8.52 (s, 1H), 7.95 (d,7 = 7.7 Hz,
1H), 7.83-7.75 (m, 2H), 7.67 (s, 1H), 7.60 (dd, J= 7.9, 7.7 Hz, 1H), 7.37 (s. 1H), 7.31 (s.
1H), 5.51 (s, 2H), 4.30 (t J - 5.1 Hz, 2H), 3.97 (t, J = 5.1 Hz, 2H), 3.84 (s, 3H), 3.77 (s,
3H). MS (ES+, m/z) 541 (m+1).
Intermediate Example 27: 5-[6-[(2-Chloroethyl)oxy3-5-(methyloxy)-1 H-benzimidazol-
1-yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-thiophenecarboxylic acid
Methyl 5-[6-[(2-chloroethyl)oxy]-5-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy)-2-thiophenecarboxylate (0.115 g, 0.213 mmol)
was dissolved in 10 mL of methanol with stirring. A 1.0M lithium hydroxide solution
(10 mL, 10 mmol) was added and the mixture was stirred for 24 hours. The reaction
was judged incomplete so it was heated to 40 °C for an additional 24 hours. The
reaction was cooled to room temperature and poured into 0.5N NaOH and diethyl
ether. The layers were separated, and the aqueous layer was washed with diethyl
ether. The diethyl ether layers were discarded, and the aqueous layer acidified with
concentrated HCI. The aqueous layer was extracted with ethylacetate (2X) and
dichloromethane. The combined organic layers were dried over MgSO4, filtered, and
concentrated to yield 0.0800 g (71%) of 5-[6-[(2-chloroethyl)oxy]-5-(methyloxy)-1H-
benzimidazol-1-yl]-3-({[2-(trifluoromethyl)-phenyl]methyi}oxy)-2-
thiophenecarboxylic acid as a white solid. 1H NMR (400 MHz, DMSO-d6) d 12.85 (brs,
1H), 8.52 (s, 1H), 7.96 (d, J= 7.5 Hz, 1H), 7.84-7.75 (m, 2H), 7.64-7.58 (m, 2H), 7.38 (s,
1H), 7.31 (s, 1H), 5.50 (s, 2H), 4.31 (t, J = 5.1 Hz, 2H). 3.98 (t, J = 5.1 Hz, 2H), 3.85 (s,
3H). MS (ES+, m/z) 527 (m+1).
Intermediate Example 28: 5-[6-[(2-Chloroethyl)oxy1-5-(methyloxy)-1H-benzimidazol-
1 -yl]-3-({ [2-(trifluoromethyl)phenyl] methyl}oxy)-2-thiophenecarboxamide
5-[6-[(2-Chloroethyl)oxy]-5-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-(trifluoro-
methyI)phenyl]methyl}oxy)-2-thiophenecarboxylic acid (0.0790 g, 0.150 mmol) and
ammonium chloride (0.0160 g, 0.299 mmol} were placed in a flask. 5 mL of N,N-
dimethylformamide was added, and the mixture was stirred. N-Methylmorpholine
(0.032 mL, 0.29 mmol) was added via syringe. 1-Hydroxybenzotriazole (0.0405 g,
0.300 mmol) was added in a single portion. 1-(3-Dimethytaminopropyl)-3-
ethylcarbodiimide hydrochloride (0.0403 g, 0.210 mmol) was added, and the mixture
was stirred for 64 hours. The reaction was poured into ethyl acetate and 1N HCI, and
the layers were separated. The organic layer was washed with brine, and the
combined aqueous layers were extracted with ethyl acetate. The combined organic
layers were dried over MgSO4, filtered, and concentrated. Purification by flash
chromatography provided 0.0760 g (96%) of 5-[6-[(2-chloroethyl)oxy]-5-(methyloxy)-
1H-benzimidazol-1-yl]-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxamide as an off-white solid. 1H NMR (300 MHz, DMSO-d6) d 8.49 (s,
1H), 7.91-7.64 (m, 5H), 7.65 (s, 1H), 7.41 (s, 1H), 7,31 (s, 1H), 6.84 (brs, 1H), 5.59 (s,
2H), 4.34 (t, J = 5.0 Hz, 2H), 4.02 (t, J - 5.0 Hz, 2H), 3.88 (s, 3H).
Example 136: 5-(5-(Methvloxv)-6-{[2-(4-methyl-1-piperazinyl)ethvl]oxv}-1 H-
benzimidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxy)-2-
thiophenecarboxamide
5-[6-[(2-Chloroethyl)oxyl-5-(methyloxy)-1H-benzimidazol-1-yl]-3-({[2-
(triflupromethyl)-phenyl]methyl}oxy)-2-thiophenecarboxamide (0.0750 g, 0.143
mmol) was dissolved In 3 mL of 1-methylpiperazine and heated to 90 °C with an oil
bath. After 3 hours cool to room temperature and adsorb onto a mixture of NaHCO3
and silica gel (1:5). The sample was purified by flash chromatography and
concentrated in vacuo. The residue was dissolved in approximately 5 mL of methanol
and 1 mL of 1N HCI in diethyl ether was added with swirling. Excess diethyl ether was
added to induce precipitation of a white solid. The mixture was filtered, and the solid
washed with diethyl ether. The solid was air dried and collected to provide 0.0496 g
(52%) of 5-(5-(methyloxy)-6-{[2-(4-methy}-1-piperazinyl)ethyl3oxy}-1 H-
benzimidazol-1-yl)-3-({[2-(trifluoromethyl)phenyl]methyl}-oxy)-2-
thiophenecarboxamide as its di-HCI salt For NMR analysis solid Na2CO3 was added to
the NMR tube to free base the sample in situ. 1H NMR (400 MHz, DMSO-d6) d 8.42 (s,
1H), 7.88-7.58 (m, 5H), 7.59 (s, 1H). 7.32 (s, 1H), 7,24 (s, 1H), 6.80 (br s, 1H), 5.54 (s.
2H), 4.10 (t. J - 5.7 Hz. 2H), 3.81 (s. 3H), 2.69 (t, J - 5.8 Hz, 2H), 2.48-2.15 (m, 8H), 2.10
(s. 3H). MS (ES+, m/z) 590 (m+1).
Unless otherwise noted, the following compounds were prepared according to general
procedures outlined for Examples 134,135 and 136 with appropriate intermediates.
Example 137: 5-(5-(Methvloxy)-6-([2-(4-morpholinyl)ethyl]oxy}-1 H-benzimidazol-
1-vl)-3-({[2-(trifluoromethyl)phenyl]methyl}oxv)-2-thiophenecarboxamicle
1H NMR (400 MHz, DMSO-d6) d 8.44 (s. 1H), 7.87-7.63 (m, 5H), 7.62 (s, 1H), 7.34 (s, 1H),
7.27 (s, 1H), 6.82 (br s, 1H), 5.56 (s, 2H), 4.13 (t, J= 5.9 Hz, 2H), 3.83 (s, 3H), 3.59-3.54
(m, 4H), 2.73 (t, J= 5.9 Hz, 2H). MS (ES+, m/z) 577 (m+1).
Example 138: 5-[6-(2-Morpholin-4-ylethoxy)-1 H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thibphene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.50 (s, 1H). 7.86 (d, J = 8.06 Hz, 2H), 7.79 (t, J - 7.6
Hz, 1H), 7.73 (br s, 1H), 7.68 (s, 1H), 7.65 (d, J - 6.41 Hz, 2H), 7.23 (d, J - 1.65 Hz, 1H),
6.99 (dd, J = 2.01 Hz, J m 8.79 Hz, 1H), 6.82 (br s, 1H), 5.56 (s, 1H), 4.15 (t, J - 5.58 Hz,
2H), 3.58 (t, J - 4.39 Hz, 4H), 2.73 (t, J - 5.58 Hz, 2H). MS (ES+, m/z) 547 (m+1).
Example 139: 5-[6-(2-Pvrrolidin-1-ylethoxy)-1 H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxylthiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.50 (s, 1H), 7.86 (s, 1H). 7.84 (s, 1H), 7.79 (t, 7= 7.60
Hz, 1H), 7.73 (br s, 4H).7.68-7.64 (m, 3H). 7.23 (d, J = 1.83 Hz, 1H), 6.99 (dd, J- 2.11
Hz, 8.70 Hzt 1H), 6.82 (br s, 1H), 5.56 (s, 2H), 4.14 (t, J = 5.58 Hz, 2H), 2.85 (br s, 2H),
2.57 (br s, 4H), 1.70 (br s, 4H). MS (ES+, m/z) 531 (m+1).
Example 140: 5-[5-Fluoro-6-(2-morpho)in-4-vlethoxy)-1 H-benzimidazol-1-yl]-3-
{[2-(trifluoromethyl))benzvl]oxy}thiophene-2-carboxamide
1H NMR (400 MHz, DMSO-d6) d 8.54(s,1H),7.85 - 7.63 (m, 7H); 7.43 (d, J= 7.48 Hr,
1H), 6.83 (br s, 1H). 5.55 (s, 2H), 4.23 (t, J = 5.64 Hz, 2H), 3.57 (t, J - 4.43 Hz, 4H), 2.75
(t, J = 5.64 Hz, 2H). MS (ES+. m/z) 565 (M+1).
Example 141: 5-(5-Hydroxy-1 H-benzimidazol-1-vl)-3-[(2-methylbenzyl)oxy]-
thiophene-2-carboxylic acid
1H NMR (400 MHz, CD3OD) d 9.64 (s, 1H); 7.69-7.63 (m, 2H); 7.49 (d, J= 7.4 Hz, 1H);
7.24-7.18 (m, 5H); 5.37 (s, 2H); 2.43 (s, 3H). MS (ES+, m/z) 380 (M+).
Example 142: 5-[5-(2-Methoxvethoxy)-1H-benzirnidazol-1-y)]-3-[(2-
methvlbenzyl)oxylthiophene-2-carboxamide
1HNMR (400 MHz, CD3OD) d 8.45 (s, 1H); 7.83-7.78 (m, 2H); 7.73 (t, .J = 7.1 Hz, 1H);
7.65-7.59 (m, 2H); 7.43 (s, 1H); 7.29 (d, J = 2.2 Hz, 1H); 7.10 (dd, J = 2.2,4.7 Hz, 1H);
5.58 (s, 2H); 4.20-4.18 (m, 2H); 3.80-3.78 (m, 2H); 3.44 (s, 3H). MS (ES+, m/z) 491 (M+).
Intermediate Example 29: 1,1-Pimethylethyl 4-[({1-[5-[(methyloxy)carbonyl]-4-({[2-
(trifluoromethyl)phenyl3methyl}oxy)-2-thienylI-1H-benzirnidazol-6-yl}oxy)methyl]-
1-piperidinecarboxylate
Methyl 5-(6-hydroxy-1H-benzimidazol-1-yl)-3-({[2-(trifluoromethyl)-
phenyl]methyl}-oxy)-2-thiophenecarboxylate (0.150 g, 0.335 mmol) and 1,1-
d!methylethYl4-({[{4-methylphenyl)sulfonyl]oxy}methyl)-1-piperidinecarboxyiate
(0.161 g, 0.436 mmol) were dissolved in 5 mL of N,N-dimethylformamide with stirring.
Cesium carbonate (0.164 g, 0.503 mmol) was added in a single portion, and the
reaction was heated to 60 °C with an oil bath. The reaction was stirred at this
temperature for seven hours and cooled to room temperature. The mixture was
poured into water and ethyl acetate, and the layers were separated. The organic layer
was washed with brine, and the combined aqueous layers were extracted with ethyl
acetate. The combined organic layers were dried over MgSO4. filtered, and
concentrated in vacuo. The residue was purified by flash chromatography to provide
0.186 g (86%) of 1,1-dimethylethyl 4-[({1-[5-[(methyloxy)carbonyl]-4-({[2-
(trifluoromethyl)phenyl]methyl}oxy)-2-thienyl]-1H-benzimidazol-6-yl}oxy)methyl}-
1-piperidinecarboxylate. 1H NMR (400 MHz, DMSO-d6) d 8.59 (s, 1H), 8-00 (d, J= 8.0
Hz, 1H), 7.87-7.79 (m. 2H), 7.74-7.60 (m, 3H), 7.28 (d, J = 2.1 Hz, 1H), 7.03 (dd. 7- 8.8,
2.2 Hz, 1H). 5.56 (s, 2H), 4.02 (m, 2H), 3.95 (d, J - 6.5 Hz, 1H), 3.82 (s, 3H), 2.78 (br s.
1H), 2.00 (br s, 1H), 1.87-1.76 (m, 2H), 1.43 (s, 9H), 1.84-1.12 (m, 2H). MS (ES+, m/z)
646 (m+1).
Example 143: 5-{6-[(4-Piperidmylmethyl)oxy]-1H-benzimidazol-1-yl}-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy)-2-thiophenecarboxamide
1,1-Dimethylethyl 4-[({1-[5-(aminocarbonyl)-4-({[2-(trifluoromethyl)phenyl]-
methyl}-oxy)-2-thienyl]-1H-benzimidazol-6-yl}oxy)methyl]-t-piperidinecarboxylate
was dissolved in 7 mL of methanol with stirring. 4 mL of concentrated HCI was added
and the solution was heated to 45 °C for 1 hour. The solution was cooled to room
temperature and concentrated in vacuo to afford 0.0866 g (87%) of 5-{6-[(4-
piperidinylmethyl)oxy]-1H-benzimidazol-1 -yl}-3-({[2-(trifluoromethyl)phenyl]-.
methyl}oxy)-2-thiophenecarboxamide as its HCI salt For 1H NMR analysis solid
Na2CO3 was added to the NMR tube to free base the sample in situ. 1H NMR (400
MHz, DMSO-d6) d 8.47 (s, 1H), 7.85-7.81 (m, 2H), 7.80-7.71 (m, 2H), 7.67-7.60 (m, 3H),
7.16 (d,J = 2.2 Hz, 1H), 6.96 (dd. J = 8.8, 2.2 Hz, 1H), 6.81 (br s, 1H), 5.54 (s, 2H), 4.09
(m, 2H),3.89-3.81 (m, 2H), 2.93 (d,J= 10.6 Hz, 1H), 1.83 (brs, 1H), 1.73-1.62 (m, 2H),
1.27-1.05 (m, 2H). MS (ES+, m/z) 531 (m+1).
Example 144: 5-(1 H-Benzimidazol-1-yl)-3-(benzyloxy)-N-hydroxythiophene-2-
carboxamide
To a cooled (0°C) solution of 5-(1H-benzimidazol-1-yl)-3-(benzyloxy)thiophene-2-
carboxylic acid (100 mg, 0.28 mmol) in dichloromethane (2.0 mL) was added
dimethylformamide (22 microL, 0.28 mmol) followed by a 2.0M solution of oxalyl
chloride in dichloromethane (310 microL, 0.62 mmol). The reaction was stirred at 0°C
for 40 minutes then added to a solution of hydroxylamine hydrochloride (78 mg, 1.12
mmol) and triethylamine (233 microL, 1.67 mmol) in 85:15 tetrahydrofuran/HzO (1
mL). The reaction was stirred at room temperature for 45 minutes then poured into
1M aqueous HCI and extracted with dichloromethane. The organic extracts were
washed with brine and dried over Na2SO4. Filtration and concentration followed by
reverse-phase PREP HPLG (30-to-70°/o acetonitrile/H2O with 0.1% formic acid) gave 5-
(1 H-benzimidazol-1-yl)-3-(benzyloxy)-N-hydroxythiophene-2-carboxamide (10 mg,
10%) as an off-white solid. 1H NMR (400 MHz, CDCI3) 8 9.51 (s, 1H), 8.08 (s, 1H), 8.89-
8.84 (m, 1H), 7.60-7.56 (m, 1H), 7.47-7.37 (m, 8H), 6.95 (s, 1H), 5.30 (s, 2H). MS (ES+.
m/z) 365 (m+1).
Example 145: 3-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzyl]oxy}thiophene-2-carbothioamide
To a solution of 5-(5,6-dimethoxy-1H-benzimidazol~1-yl)-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide (50 mg, 0.10 mmol) in 1,4-
dioxane (1.5 mL) was added Lawesson"s Reagent (32 mg, 0.08 mmol). The reaction was
heated to 80°C for 3 hrs, cooled to room temperature and additional Lawesson"s
Reagent was added (32 mg, 0.08 mmol). The reaction was heated to 80°C for 2hrs then
cooled to room temperature. The reaction was poured into aqueous 1M HCI and
extracted with dicholormethane. The organic extracts were dried over Na2S0«.
Filtration and concentration followed by reverse-phase PREP HPLC (30-to-70%
acetonitrile/H2O with 0.1% formic acid) gave 5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carbothioamide (25 mg, 48%) as a
light yellow solid. 1H NMR (400 MHz, DMSO-d6) d 9.63 (s, 1H), 8.35 (m, 2H), 7.76-7.65
(m, 3H), 7.56-7.51 (m, 1H), 7.47 (s, 1H), 7.23 (s, 1H), 7.11 (s, 1H), 5.49 (s, 2H), 3.72 (s,
3H), 3.71 (s, 3H). MS (ES+, m/z) 493 (m+1).
Example 146: 5-(5.6-Dimethoxv-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carbonitrile
To a solution of 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide (150 mg, 0.31 mmol) in
dichloromethane (2 mL) was added 2-chloro-1,3-dimethylimidazolinium chloride (120
mg, 0.71 mmol) and trifluoroacetic acid (50 microL, 0.65 mmol). To this solution was
added triethylamine (200 microL, 1.44 mmol). The mixture was stirred 18 hrs, then
additional 2-chloro-1,37dimethylimidazolinium chloride (120 mg, 0.71 mmol) and
trifluoroacetic acid (50 microL, 0.65 mmol) were added, followed by triethylamine (
200 microL, 1.44 mmol). The mixture was stirred for 4 hrs then poured into H2O and
extracted with dichloromethane. The organic extracts were washed with aqueous 5%
HCI, aqueous (saturated) NaHCO)3, brine and dried over Na2SO4. Filtration and
concentration followed by silica gel chromatography (eluting with a 40-to-95%
EtOAc/hexane gradient) gave 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carbonitrile (66 mg, 46%) as a yellow solid.
1H NMR (400 MHz, CDCI3) 8 7.90 (s, 1H), 7.80-7.72 (m, 2H), 7.66 (t, J - 7.60 Hz, 1H),
7.51 (t, J - 7.51 Hz, 1H), 7.31 (s, 1H), 6.99 (s. 1H), 6.82 (s, 1H), 5.55 (s, 2H), 3.96 (s, 3H),
3.92 (s. 3H). MS (ES+, m/z) 459 (m+1).
Example 147: 5.6-Dimethoxv-1-(5-(1H-tetraazol-5-yl)-4-{[2-(trifluoromethvl)-
benzyl]oxy}thien-2-vl)-1H-benzimidazole
To a Smithcreator Microwave reaction vessal was added 5-(5,6-dimethoxy-1H-
benzimidazol-1-yl)-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carbonitrile(53
mg, 0.11 mmol), sodium azide (20 mg, 0.31 mmol), ammonium chloride (16 mg, 0.31
mmol) and dimethylformamide (2.0 mL). The reaction vessal was sealed and heated at
120°C for 20 minutes in the Smithcreator Microwave. The reaction vessal was cooled
to room temperature, opened, and additional sodium azide (20 mg, 0.31 mmol) and
ammonium chloride (16 mg, 0.31 mmol) was added. The vessal was sealed and heated
at 120°C for 10 minutes on the microwave, then cooled to room temperature and
opened. The mixture was poured into aqueous (saturated) NaHCO3 and washed with
diethyl ether. The aqueous layer was then acidified to pH 1.0 by addition of
concentrated HCI, then extracted with ethyl acetate. The organic extract was washed
with brine and dried over Na2SO4 Filtration and concentration followed by reverse-
phase PREP HPLC (30-to-70% acetonitrile/H2O with 0.1% formic acid) gave 5,6-
dimethoxy-1-(5-(1H-tetraazol-5-yl)-4-{[2-(trifluoromethyl)benzyl]oxy}thien-2-yl)-
1H-benzimidazole (25 mg, 43%) as a white solid. 1HNMR (400 MHz, CDCI3) 8 7.97 (s,
1H), 7.81 (d, J - 7.87 Hz, 1H), 7.69-7.56 (m. 3H), 7.32 (s, 1H), 7.08 (s, 1H), 6.99 (s, 1H),
5.57 (s, 2H), 3.95 (s, 3H), 3.93 (s, 3H). MS (ES+, m/z) 502 (m+1).
Intermediate Example 30: Methyl 3-hydroxy-5-[2-(methylthio)-1 H-benzimidazol-1-
vflthiophene-2-carboxylate
A mixture of 2-(methylthio)-1 H-benzimidazole (5.0 g, 25.9 mmol) and methyl 2-
ehloro-3-oxo-2,3-dihydrothiophene-2-carboxylate (8.53 g, 51.9 mmol) were dissolved
in chloroform (100ml.) and glacial acetic acid (12 mL). Stirred at room temperature
for 72 hrs. Poured reaction mixture into separatory funnel containing
dichloromethane (150 mL), washed with distilled water (2x100 mL). Extracted
combined aqueous layers with dichloromethane (2x50 mL)- Washed combined organic
layers with distilled water (3x100 mL). Dried organic layer (MgSO4), filtered and
concentrated under reduced pressure. Dissolved residue in dichloromethane and
methanol, and added silica gel (35 g). Following evaporation of the volatiles under
reduced pressure, the pre-adsorbed solids were loaded into a solid loading cartridge
and subjected to an isocratic elution with dichloromethane (100%) using a RediSep
silica gel cartridge (330 g; ISCO). The appropriate fractions were combined and
concentrated under reduced pressure to give methyl 3-hydroxy-5-[2-(methyithio)-1H-
benzimidazol-1-yl]thiophene-2-carboxylate (4.75 g) as an off-white solid. 1H NMR
(400 MHz, CDCI3): 8 9.77 (s. 1H), 7.71-7.68 (m.1IH), 7.36-7.34 (m, 1H), 7.30-7.26 (m,
1H), 7.24-7.20 (m, 1H), 6.87 (s, 1H), 3.93 (s, 3H). 2.78 (s, 3H). MS (ES+. m/z) 321 (M+1).
Intermediate Example 31: Methyl 5-[2-(methvlthio)-1tf-benzimidazoM-yl]-3-{[2-
(trifluoromethyl)benzvl]oxy)thiophene-2-carboxvlate
In a similar manner as described for Example 54,3-hydroxy-5-[2-(methylthio)-1H-
benzimidazol-1-yl]thiophene-2-carboxylate (4.5 g, 14.0 mmol) and 1-(bromomethyl)-
2-(trifluoromethyl)benzene (3.36 g, 14.0 mmol) gave methyl 5-[2-(methylthio)-1H-
benzimidazol-1-yl]-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carboxylate
(5.99 g) as a tan solid. 1H NMR (400 MHz, CDCl3): 6 7.93 (d, J= 7.7 Hz, 1H), 7.8-7.76
(m, 2H), 7.65-7.58 (m, 3H), 7.37-7.34 (m, 1H), 7.29-7.21 (m, 2H), 5.46 (s, 2H), 3.77 (s,
3H), 2.71 (s. 3H). MS (ES+, m/z) 479 (M+1).
Intermediate Example 32: Methyl 5-[2-(methvlsulfonyl)-1H-benzimidazol-1-yl]-3-
{[2-(trifluoromethvl)benzvlloxy|thiophene-2-carboxylate
To a solution of methyl 5-[2-(methylthio)-1H-benzirnidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxylate (150 mg, 0.31 mmol) in
dichloromethane (5 mL) under nitrogen atmosphere was added 3-chloroperoxybenzoic
acid (77%) (178 mg, 0.79 mmol) and stirred at room temperature for 24 hours.
Concentrated under reduced pressure to give an off-white solid. Dissolved in
chloroform (100 mL) and poured reaction mixture into separatory funnel. Washed
with saturated NaHCO3 aqueous solution (2x50 mL), and brine (2x50 mL). Dried
organic layer (MgS04), filtered and concentrated under reduced pressure to give a gold
oil. Dissolved in dichloromethane (25 mL) and added silica gel (500 mg), followed by
evaporation of the volatiles under reduced pressure. The pre-adsorbed solids were
loaded into a solid loading cartridge and subjected to a gradient elution using ethyl
acetate:hexanes (20:80) to ethyl acetate:hexanes (50:50) using a RediSep silica gel
cartridge (12 g; ISCO). The appropriate fractions were combined and concentrated
under reduced pressure to give methyl 5-[2-(methylsu!fonyl)-1H-benzimidazol-1-yl]-
3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carboxylate (130 mg) as a white solid.
1H NMR (400 MHz, CDCl3): 8 7.97 (d, J - 7.8 Hz, 1H), 7.89-7.86 (m, 1H), 7.69-7.62 (m,
2H), 7.49-7.39 (m, 4H), 7.16 (s, 1H), 5.46 (s, 2H), 3.91 (3, 3H), 3.50 (s, 3H). MS (ES+.
m/z)511 (M+1).
entermediate Example 33: 5-[2-(Methvlthio)-1H-benzimidazol-1-yl1-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide
In a similar manner as described for Example 61, methyl 5-[2-(methylthio)-1H-
benzimidazol-1-yl]-3-{[2-(tritluoromethyl)benzyl]oxy}thiophene-2-carboxylate(160
mg, 0.343 mmol) and 7N NH3 in methanol (10 mL, 70.0 mmol) gave 5-[2-(methylthio)-
1H-benzimidazol-1-yl]-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide
(136 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6): d 7.84-7.75 (m, 4H), 7.65-
7.62 (m, 2H), 7.56 (s, 1H), 7.32-7.30 (m, 1H), 7.28-7.20 (m, 2H), 6.87 (bs, 1H), 5.50 (s,
2H), 2.70 (s, 3H). MS (ES+, m/z) 464 (M+1).
Intermediate Exmaple 34: 5-[2-(Methylsulfonyl)-1H-benzimidazol-1-yl]-3-{[2-
(trifluorotnethyl)benzyl]oxy}thiophene-2-carboxamide
To a solution of 5-[2-(methylthio)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide (1.25 g, 2.69 mmol) in
dichloromethane (50 mL) under nitrogen atmosphere was added 3-
chloroperoxybenzoic acid (77%) (1.86 g, 8.29 mmol) and stirred at room temperature
for 24 hours. Concentrated under reduced pressure to give an off-white solid.
Dissolved in dichloromethane and methanol, added silica gel (10.0 g), followed by
evaporation of the volatiles under reduced pressure. The pre-adsorbed solids were
loaded into a solid loading cartridge and subjected to a gradient elution using ethyl
acetatethexanes (15:85) to ethyl acetate:hexanes (60:40) using a RediSep silica gel
cartridge (40 g; ISCO). The appropriate fractions were combined and concentrated
under reduced pressure to give 5-[2-(methylsulfonyl)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide (869 mg) as an off-white solid.
1H NMR (400 MHz, CDCl3): 5 7.89-7.87 (m, 1H), 7.76-7.74 (m, 1H), 7.66-7.61 (m, 2H),
7.54-7.44 (m, 4H), 7.25 (s, 1H), 7.02 (bs, 1H), 5.69 (bs, 1H), 5.44 (s, 2H), 3.51 (s, 3H). MS
(ES+, m/z) 496 (M+1).
Example 148: 5~(2-Amino-1H-benzimidazol-1-yl)-3-{[2-
(trifluoromethvl)benzyl]oxy}thiophene-2-carboxamide
Method A: In a sealed tube, a mixture of 5-[2-(methylsulfonyl)-1 H-benzimidazol-1-
yl]-3-{[2-(trifluoromethyl)benzyl]oxy}th[ophene-2-carboxamide (410 mg, 0.827
mmol) in 7N NH3 in methanol (20 mL, 140 mmol) was heated to 80°C for 24 hours.
Cooled reaction mixture to room temperature, and filtered precipitate over glass-
fritted funnel. The filtrate was concentrated under reduced pressure to give a solid
residue (180 mg), which was dissolved in methanol and dichloromethane. Added silica
gel (250 mg), followed by evaporation of the volatiles under reduced pressure. The
pre-adsorbed solids were loaded into a solid loading cartridge and subjected to a
gradient elution using dichloromethane:methanol (100:0) to
dichloromethane:methanol (85:15) using a RediSep silica gel cartridge (4g; ISCO). The
appropriate fractions were combined and concentrated under reduced pressure to give
5-(2-amino-1H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-
carboxamide (25 mg) as a tan solid. 1H NMR (400 MHz, DMS0-d6): d 8.83 (s, 2H), 7.90-
7.75 (m, 4H), 7.65-7.62 (m, 2H), 7.42 (d, J - 7.9 Hz, 1H). 7.34-7.23 (m, 2H), 7.17 (d, J =
8.6 Hz, 1H). 6.93 (bs, 1H), 5.47 (s, 2H). MS (ES+, m/z) 433 (M+1).
Method B: In a sealed tube, a mixture of methyl 5-[2-(methylsulfonyl)-1H-
benzimidazol-1-yl]-3-{[2-(trifluoromethy!)benzyl]oxy}thiophene-2-carboxylate in 7N
NH3 in methanol were reacted together to give the 5-(2-amino-1H-benzimidazol-1-
yl)-3-{[2-(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide.
Example 149: Methyl 5-(5.6-dimethoxy-1H-benzimidazol-1-yl)-3-{[(2-
nitrophenyl)sulfonyljoxy} thiophene-2-carboxylate
To a solution of 5-(5,6-dimethoxy-1 H-benzimidazol-1-yl)-3-hydroxythiophene-2-
carboxamide (170 mg, 0.50 mmol) and N,N-diisopropylethylamine (0.12 mL, 0.70
mmol) in dichloromethane (5 mL) was added 2-nitrobenzenesulfonyl chloride (130
mg, 0.60 mmol). The solution was stirred 1h, at which time silica gel (5g) was added.
The volatiles were evaporated under reduced pressure, and the pre-adsorbed solids
were loaded into a solid loading cartridge and subjected to a gradient elution using
hexanes:ethyl acetate (80:20) to hexanesrethyl acetate (0:100) using a RediSep silica
gel cartridge (4 g; ISCO). The appropriate fractions were combined and concentrated
under reduced pressure to give methyl 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-
{[(2-nitrophenyl)sulfonyl]oxy} thiophene-2-carboxylate (240 mg) as a white solid. 1H
NMR (400 MHz, CDCl3) 8 8.31 (dd,J= 8.0,1.5 Hz, 1H). 7.96 (s, 1H), 7.91-7.81 (m, 3H).
7.32 (s, 1H), 7.19 (s, 1H), 7.15 (s, 1H), 3.98 (s, 3H), 3.97 (s, 3H), 3.76 (s, 3H). MS (ES+,
m/z) 520 (m+1).
Example 150: Methyl 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-
{[(trifluoromethyl)sulfonvl]oxy) thiophene-2-carboxylate
Compound was prepared according to general procedure outlined for Example 30. 1H
NMR (400 MHz, DMSO-ds) d 8.52 (s, 1H), 7.84 (s, 1H), 7.35 (s, 1H), 7.26 (m, 3H), 3.89 (s,
3H), 3.84 (s, 3H), 3.81 (s, 3H). MS (ES+, m/z) 467 (m+1).
Example 151: 5T(5.6-Dimethoxy-1H-benzimidazol-1-yl)-3-{[(2-
methylphenyl)sulfonyl]oxy)thiophene-2-carboxylicacid
To a solution of methyl 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-{[(2-
methylphenyI)sulfonyl]oxy}thiophene-2-carboxylate (100 mg,0.20 mmol) in
tetrahydrofuran (2 mL) was added 0.1 N NaOH (2 mL, 0.20 mmol). The solution was
stirred 1 h, at which time the solution was neutralized by the addition of 0.1 N HCI (2
mL, 0.20 mmol), and a white solid precipitated. Vacuum filtration provided 5-(5,6-
dimethoxy-1H-benzimidazol-1-yl)-3-{[(2-methylphenyl)sulfonyl]oxy}thiophene-2-
carboxylic acid (7 mg) as a white solid. 1H NMR (400 MHz, CDCl3) 8 9.50 (s, 1H), 7.88
(d, J - 8.0 Hz, 1H), 7.54 (dd, J - 8.3,7.1 Hz, 1H), 7.49 (s, 1H), 7.42 (d, J - 9.3 Hz, 1H),
7.32-7.27 (m 1H), 7.19 (s, 1H), 7.15 (s, 1H), 4.03 (s, 3H), 4.02 (s, 3H), 2.81 (s, 3H). MS
(ES+, m/z) 475 (m+1).
Example 152: 5-(5,6-Dimethoxy-1 H-benzimidazol-1 -yl)-3-hydroxythiophene-2-
carboxamide trifluoroacetate
To solid 5-(5,6-dimethoxy-1H-benzimidazol-1-yl)-3-[(4-
methoxybenzyl)oxy]thiophene-2-carboxamide (400 mg, 0.91 mmol) was added
trifluoroacetic add (2 ml). The bright red solution was stirred 10 minutes, at which
time.ether (20 mL) was added, and a pink solid precipitated. Vacuum filtration
provided 5-(5,6-dimethoxy-1H-benzimidazol-1 -yl)-3-hydroxythiophene-2-
carboxamide trifluoroacetate (300 mg) as a pink solid. 1H NMR (400 MHz, DMSO-d6) d
8.70 (s, 1H), 7.33 (s, 1H), 7.23 (s, 1H), 7.10 (s, 1H), 7.05 (br s, 1H), 3.83 (s, 3H), 3.82 (s.
3H). MS(ES+,m/z)320(m+1).
Example 153: 2-(Aminocarbonvl}-5-(5,6-dimethoxy-1 H-benzimidazol-1-yl)thien-3-yl
2-nitrobenzenesulfonate
To a solution 5-(5,6-dimethoxy-1 H-benzimidazol-1 -yl)-3-hydroxythiophene-2-
carboxamide trifluoroacetate (44 mg, 0.10 mmol) and N,N-diisopropylethylamine
(0.058 mL, 0.33 mmol) in dichloromethane (2 mL) was added 2-nitrobenzenesulfonyl
chloride (24 mg, 0.11 mmol). The solution was stirred 3h, at which time silica gel (2 g)
was added. The volatiles were evaporated under reduced pressure, and the pre-
adsorbed solids were loaded into a solid loading cartridge and subjected to a gradient
elution using ethyl acetate (100%) to ethyl acetate:methanol (80:20) using a RediSep
silica gel cartridge (4 g; ISCO). The appropriate fractions were combined and
concentrated under reduced pressure to give methyl 2-(aminocarbonyl)-5-(5,6-
dimethoxy-1H-benzimidazol-1 -yl)thien-3-yl 2-nitrobenzenesulfonate (37 mg) as a
white solid. 1H NMR (400 MHz, DMSO-d6) d 8.38 (s, 1H), 8.22-7.93 (m, 4H), 7.80 (brs,
1H), 7.40 (s, 1H), 7.34 (br s, 1H), 7.33 (s, 1H), 7.15 (s, 1 H)t 3.82 (s, 3H), 3.81 (s, 3H). MS
(ES+, m/z) 505 (m+1).
Example 154: 2-(Aminocarbonyl)-5-(5,6-dimethoxy-1H-benzimidazol-1 -yl)thien-3-yl
2-methylbenzenesulfonate
2-(Aminocarbonyl)-5-(5,6-dimethoxy-1H-benzimidazol-1 -yI)thien-3-yl 2-
methylbenzenesulfonate was prepared using similar procedure described above for the
preparation of 2-(amihocarbonyl)-5-(5,6-dimethoxy-1H-benzimidazoI-1 -yl)thien-3-yl
2-nitrobenzenesulfonat except 2-methylsulfonyl chloride was used instead of 2-
nitrobenzenesulfonyl chloride. 1H NMR (400 MHz, DMSO-d6) d 8.35 (s, 1H). 7.91 (dd, J
= 8.0,1.2 Hz, 1H), 7.79, (brs, 1H), 7.72 (ddd,J= 7.7, 7.4,1.3 Hz. 1H), 7.56 (d, J= 7.4 Hz,
1H), 7.45 (dd, J= 7:7, 7.7 Hz, 1H). 7.34 (br s, 1H), 7.32 (s. 1H), 7.15 (s, 1H), 7.05 (s, 1H),
3.80 (s, 3H), 3.80 (s. 3H). 2.68 (s, 3H). MS (ES+, m/z) 474 (m+1).
Intermediate Example 35: 1-(5-(Methoxycarbonyl)-4-{[2-
(trifluoromethvl)benzyl]oxy}thien-2-yl)-1H-benzimidazole-5-carboxvlic acid
To a solution of vinyl 1-(5-(methoxycarbonyl)-4-{[2-
(trifluoromethyl)benzyl]oxy}thien-2-yl)-1H-benzimidazole-5-carboxylate (500 mg,
0.97 mmol) in tetrahydrofuran (3.0 mL) was added morpholine (178 microL, 2.04
mmoi) followed by tetrakis(tripheny+lphosphine)-palladium (0) (56 mg, 0.05 mmol).
The reaction was stirred at room temperature for 1 hour then poured into 0.5M
aqueous HCI and ethyl acetate. The organic layer was washed with water, brine, and
dried over Na2SO4 Filtration and concentration gave 1-(5-(methoxycarbonyl)-4-{[2-
(trifluoromethyl)benzyl]oxy}thien-2-yl)-1H-benzimidazole-5-carboxylic acid (455 mg,
98%) as a tan solid. 1H NMR (400 MHz, DMSO-d6) d 13.02 (b, 1H), 8.87 (s, 1H), 8.33 (s,
1H), 8.03 (dd, J - 8.60 and 1.46 Hz, 1H), 7.92-7.98 (m, 2H), 7.77-7.83 (m, 3H), 7.59-
7.64 (m, 1H), 5.51 (s, 2H), 3.78 (s, 3H). MS (ES+, m/z) 476 (m+1).
Example 155: 1-(5-(Aminoearbonvl)-4-{[2-(trifluoromethvl)benzvl1oxy}thien-2-vl)-
A/-[2-(methylsulfonyl)ethvl]-1H-benzimidazole-5-carboxamide
To a solution of l-(5-(methoxycarbonyl)-4-{[2-(trifluoromethyl)benzyl]oxy}thien-2-
yl)-1H-benzimidazole-5-carboxylic acid (35 mg, 0.073 mmol), 2-
(methylsulfonyl)ethanamine (14 mg, 0.11 mmol) and diisopropylethylamine (35
microL, 0.20 mmol) in dimethylformamide (1.0 mL) was added [0-(7-azabenzotriazol-
1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate] (35 mg, 0.092 mmol). The
reaction was stirred for 12 hours then poured into aqueous saturated NaHC03 and
extracted with ethyl acetate. The combined organics were washed with water, brine,
and dried over Na2SO4. Filtration and concentration gave crude methyl 5-[5-({[2-
(methylsulfonyl)ethyl]amino}carbonyl)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxylate (40 mg, 95%) as a light brown
oil. The oil was stirred as a solution in 7 M ammonia in methanol (10 mL, 70 mmol), at
80°C in a sealed, thick-walled glass pressure tube for 16 hours. The reaction was
cooled to room temperature-concentrated and purified by reverse-phase PREP HPLC
(10-90% gradient of acetonitrile/H20 with 0.1% formic acid) to give 1-(5-
(aminocarbonyl)-4-{[2-(trifluoromethyl)benzyl]oxy}thien-2-yl)-N-[2-
(methylsulfonyl)ethyl]-1H-benzimidazole-5-carboxamide (23 mg, 55%) as a white
solid. 1H NMR (400 MHz, DMSO-d6) d 8.84 (t, J - 5.58 Hz, 1H), 8.76 (s, 1H), 8.30 (s, 1H),
7.94 (dd, J = 8.60 and 1.28 Hz, 1H), 7.70-7.89 (m, 6H), 7.65 (t, J = 7.60 Hz. 1H), 6.79 (b.
1H), 5.55 (s, 2H), 3.71 (q, J = 6.41 Hz 2H). 3.41 (t, J - 6.87 Hz, 2H), 3.04 (s, 3H). MS
(ES+, m/z) 566 (m+1).
Example 156: 1-(5-(Aminocarbonvl)-4-{[2-(trifluoromethyl)benzyl]oxy)thien-2-vl)-
Al[2-(2-oxoimidazolidin-1-yl)ethyl]-1H-benzimidazole-5-carboxamide
To a solution of 1-(5-(methoxycarbonyl)-4-{[2-(trifluoromethyl)benzyl]oxy}thien-2-
yl)-1H-benzimidazole-5-carboxylic acid (112 mg,0.23 mmol), 1-(2-
aminoethyl)imidazolidin-2-one (85 mg, 0.35 mmol) and diisopropylethylamine (110
microi, 0.62 mmol) in dimethylformamide (2.0 mL) was added [0-(7-azabenzotriazol-
1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate] (115 mg, 0.30 mmol). The
reaction was stirred for 2 hours then poured into ethyl acetate and washed with
aqueous 5% HCI, aqueous saturated NaHCOa, water, brine, and dried over Na2SO4
nitration and concentration gave crude methyl 5-[5-({[2-(2-oxoimidazolidin-1-
yl)ethyl]amino}carbonyl)-1H-benzimidazol-1-yl]-3-{[2-(trifluoromethyl)-
benzyl]oxy}thiophene-2-carboxylate (128 mg, 95%) as tan solid. The solid was stirred
as a solution in 7 M ammonia in methanol (10 mL, 70 mmol), at 80°C in a sealed,
thick-walled glass pressure tube for 16 hours. The reaction was cooled to -10°C and
cold diethyl ether was added. The resulting slurry was filtered, washing the solids with
cold diethyl ether. The solids were then dried under vacuum to give 1-(5-
(aminocarbonyl)-4-{[2-(trifluoromethyl)benzyl]oxy}thien-2-yl)-N-[2-(2-
oxoimidazolidin-1-yl)ethyl]-1H-benzimidazole-5-carboxamide (53 mg, 44%) as a
white solid. 1H NMR (400 MHz, DMSO-d6) d 8.75 (s, 1H), 8.64 (t, J - 5.49 Hz, 1H), 8.28
(s, 1H), 7.70-7.94 (m. 7H), 7.65 (t, J - 7.60 Hz, 1H), 6.79 (b, 1H), 6.28 (s, 1H), 5.55 (s,
2H), 3.36-3.44 (m, 4H), 3.18-3.27 (m, 4H). MS (ES+, m/z) 572 (m+1).
Intermediate Example 36: Methyl 5-{6-[(tert-butoxycarbonyl)amino]-1H-
benzimidazol-l-yl}-3-hydroxythiophene-2-carboxylate and methyl 5-{5-[(tert-
butoxycarbonyl)amino]-1H-benzimidazol-1-yl}-3-hvdroxythiophene-2-carboxylate
Compounds were prepared using procedure similarly described in Example 2A. M
(ES-, m/z) 388 (m-1).
Intermediate Example 37: Methyl 5-{6-[(tert-butoxvcarbonyl)amino]-1 H-
benzimidazo1-1-yl}-3-[1-(2-chlbrophenyl)ethoxy]thiophene-2-carboxylateand
Methyl 5-{5-[(tert-butoxycarbonyl)amino1-1H-benzimidazol-1-yl}-3-[1-(2-
chlorophenyl)ethoxy]thiophene-2-carboxylate
Compounds were prepared using procedure similarly described in Example 57 or
Intermediate Example 21. MS (ES+, m/z) 428 (m+1).
Intermediate Example 38: Methyl 5-(6-amino-1H-benzimidazol-1-yl)-3-[1-(2-
chlorophenyl)ethoxy3thiophene-2-carboxvlate and Methyl 5-(5-amino-1 H-
benzimidazol-1-yl)-3-[1-(2-chlorophenyl)ethoxy]thiophene-2-carboxylate
A regioisomeric mixture of methyl 5-{6-[(tert-butoxycarbonyl)amino]-1H-
benzimidazol-1-yl}-3-[1-(2-chlorophenyl)ethoxy]thiophene-2-carboxylateand
methyl 5-{5-[(fert-butoxycarbonyl)amino]-1H-benzimidazol-1-yl}-3-[1 -(2-
chlorophenyl)ethoxy]thiophene-2-carboxylate (0.610 g, 1.57 mmol) was dissolved in
20 mL of dichloromethane with stirring. Trifluoroacetic acid (6 mL) was added via
syringe. The reaction was allowed to stir for 2 hours at room temperature and the
reaction was then diluted with ethyl acetate and neutralised with bicarbonate. The
layers were separated, and the organic layer was washed with brine. The combined
aqueous layers were extracted with ethyl acetate. The combined organic layers were
dried over MgSO4, filtered, and concentrated in vacuo. Purification by flash
chromatography afforded 0.1915 g (39%) of methyl 5-(6-amino-1H-benzimidazol-1-
yl)-3-[l-(2-chlorophenyl)ethoxy]thiophene-2-carboxylate and 0.1182 g (24%) of
methyl 5-(5-amino-1 H-benzimidazol-1 -yl)-3-[1 -(2-chlorophenyl)ethoxy]thiophene-2-
carboxylate. Data for (6-NH2): 1H NMR (400 MHz. DMSO-d6) d 8.32 (s, 1H), 7.75 (dd, J
- 7.8,1.6 Hz, 1H), 7.50-7.30 (m, 6H),6.92 {d,J = 1.8 Hz, 1H), 6.62 (dd,J = 8.6,2.0 Hz,
1H). 5.93 (q, J - 6.2 Hz, 1H). 5.30 (bs, 2H). 3.80 (s, 3H), 1.61 (d, J = 6.2 Hz, 3H). MS (ES+,
m/z) 428 (m+1). Data for (5-NH2): 1H NMR (400 MHz. DMS0-d6) d 8.44 (s. 1H), 7.72
(dd, J= 7.7,1.7 Hz, 1H), 7.49-7.39 (m, 2H), 7,38-7.31 (m. 2H), 7.30 (s, 1H). 6.84 (d, J =
2.2 Hz, 1H). 6.69 (dd, J = 8.7, 2.1 Hz, 1H), 5.96 (q, J - 6.4 Hz, 1H), 5.05 (bs, 2H), 3.80 (s,
3H). 1.61 (d. J - 6.4 Hz. 3H). MS (ES+, m/z) 428 (m+1).
Example 157: 5-(5-Amino-1H-benzimidazol-1-yl)-3-[1-(2-
chlorophenyl)ethoxv]thiophene-2-carboxamide
5-(5-Amino-1H-benzimidazol-1-yl)-3-[1-(2-chlorophenyl)ethoxy]thiophene-2-
carboxamide was prepared from methyl 5-(5-amino-1H-benzimidazol-1-yl)-3-[1-(2-
chlorophenyl)ethoxy]thiophene-2-carboxylate using procedure similarly described irr
Example 61 except 7M NH3 in MeOH was used instead of 2M NHa in MeOH. 1H NMR
(400 MHz, DMSO-d6) d 8.33 (s,1H), 7.77 (bs, 1H), 7.67 (dd,J= 7.7,1.7 Hz, 1H), 7.50
(ddr J = 8.0,1.4 Hz, 1H), 7.48-7.33 (m, 2H), 7.23 (d, J = 8.8 Hz. 1H), 7.09 (bs, 1H), 7.07
(s, 1H), 6.85 (d, J = 1.8 Hz, 1H), 6.68 (dd, J - 8.6, 2.0 Hz, 1H), 5.98 (q, J = 6.4 Hz, 1H).
5.06 (bs, 2H), 1.72 (d, J = 6.4 Hz, 3H). MS (ES+, m/z) 413 (m+1).
Intermediate Example 39: Methyl 3-[1-(2-chlorophenyl)ethoxv]-5-(6-{[(1-
methylpiperidin-3-yl)carbonyl]amino)-1H-benzimidazol-1-yl)thiophene-2-
carboxylate
A soultion of 1-methylpiperidine-3-carboxylic acid hydrochloride (63 mg, 0.35 mmol),
HATU (133 mgr 0.35 mmol) and diisopropylethylamine (0.12 mL, 0.70 mmol) in DMF (3
ml) was added to a stirring solution of methyl 5-(6-amino-1H-benzimidazol-1-yl)-3-
[1-(2-chlorophenyl)ethoxy]thiophene-2-carboxylate (149 mg, 0.35 mmol) in DMF
(3mL). The resultant solution was allowed to stir at room temperature for 2h. The
reaction mixture was then diluted with EtOAc and washed several times with water.
The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo
The residue was purified by flash column chromatography to yield methyl 3-[1-(2-
chlorophenyl)ethoxy]-5-(6-{[(1-methylpiperidin-3-yl)carbonyl]amino}-1H-
enzirnidazol-1-yl)thiophene-2-carboxylate (123 mg, 64%). Data: 1H NMR (400 MHz,
CDCl3) 5 8.39 (bs, 1H), 7.91 (s, 1H), 7.73-7.66 (m, 2H), 7.35-7.27 (m, 2H). 7.25-7.19 (m,
1H), 7.15 (bs, 1H), 6.72 (s, 1H), 5.83 (q, J= 6.4 Hz, 1H), 3.90 (s, 3H), 3.03 (bs, 2H), 2.86
(bs, 2H), 2.52 (bs, 3H), 1.90 (bs, 4H). 1.73 (d, J= 6.4 Hz, 3H). MS (ES+, m/z) 553 (m+1).
Intermediate Example 40: Methyl 3-[1-(2-chlorophenvl)ethoxy]-5-(5-{[(1-
methvlpiperidin-3-yl)carbonvl]amino}-1H-benztmidazol-1-yl)thiophene-2-
carboxylate
Compound was prepared using procedure similarly described in Intermediate Example
39. 1H NMR (400 MHz, CDCl3) 8 7.97 (bs, 2H), 7.69-7.62 (m, 2H), 7.41 -7.29 (m, 3H).
7.27-7.22 (m, 1H), 6.69 (s, 1H), 5.82 (q,J= 6.3 Hz. 1H), 3.91 (s, 3H), 3.04 (bs, 2H), 2.85
(bs, 2H), 2.48 (bs, 3H). 1.99 (bs, 2H), 1.86 (bs, 2H), 1.74 (d. J = 6.3 Hz, 3H). MS (ES-, m/z)
551 (m-1).
Example 158: 3-[1-(2-Chlorophenvl)ethoxv1-5-(6-([f1-methylpiperidin-3-
yl)carbonyl]amino)-1tf-benzimidazol-1-yl)thiophene-2-carboxarnide
Compound was prepared using procedure similarly described in Intermediate Example
61 except 7M NH3 in MeOH was used instead of 2M NH3 in MeOH. 1H NMR (400 MHz,
DMSOrds) d 10.19 (s, TH), 8;50 (s, 1H), 8.38 (s, 1H), 7.84 (bs, 1H), 7.73-7.66 (m, 2H),
7.51-7.32 (m, 4H), 7.30 (s,4H), 7.11 (bs, 1H), 5.94 (q, J= 6.4 Hz, 1H), 2.90-2.86 (m, 1H).
2.75-2.71 (m, 1H), 2.63-2.57 (m, 1H), 2.20 (s, 3H). 2.10-2.01 (m, 1H), 1.93-1.79 (m, 2H),
1.74 (d, J= 6.4 Hz, 3H), 1.72-1.67 (m. 1H), 1.53-1.38 (m, 2H). MS (ES+, m/z) 538
(m+1).
Example 159: Biological Examples
I. Assay for inhibition of PLK1
A. Preparation of 6x N-terminal His-tagged PLK kinase domain
6x N-terminal His-tagged PLK kinase domain (amino acids 21-346 preceded by
MKKGHHHHHHD) SEQ ID: No. 1. was prepared from baculovirus infected T. ni cells
under polyhedrin promoter control. All procedures were performed at 4oC. Cells were
lysed in 50 mM HEPES, 200 mM NaCI, 50 mM imidazole, 5% glycerol; pH 7.5. The
homogenate was centrifuged at 14K rpm in a SLA-1500 rotor for 1 hr and the
supernatant filtered through a 1.2 micron filter. The supernatant was loaded onto a
Nickel chelating Sepharose (Amersham Pharmacia) column and washed with lysis •
buffer. Protein was eluted using 20%, 30% and 100% buffer B steps where buffer B is
50 mM HEPES, 200 mM NaCI, 300 mM imidazole, 5% glycerol; pH 7.5. Fractions
containing PLK were determined by SDS-PAGE. Fractions containing PLK were diluted
five-fold with 50 mM HEPES, 1 mM DTT, 5% glycerol; pH 7.5. then loaded on an SP
Sepharose (Amersham Pharmacia) column. After washing the column with 50 mM
HEPES, 1 mM DTT, 5% glycerol; pH 7.5, PLK was step eluted with 50 mM HEPES, 1 mM
DTT, 500 mM NaCI; 5% glycerol; pH 7.5. PLK was concentrated using a 10 kDa
molecular weight cutoff membrane and then loaded onto a Superdex 200 gel
filtration (Amersham Pharmacia) column equilibrated in 25 mM HEPES, 1 mM DTT, 500
mM NaCI, 5% glycerol; pH 7.5. Fractions containing PLK were determined by SDS-
PAGE PLK was pooled, aliquoted and stored at -80"C. Samples were quality
controlled using mass spectrometry, N-terminal sequencing and amino acid analysis..
B. Enzyme activity +/- inhibitors was determined as follows:
Compounds were added to the plate (1µl in 100% DMSO). DMSO (2% final) and EDTA
(55.5mM final) were used as controls. Reaction Mix A is prepared as follows at 4°C:
Reaction Mix A (substrate Mix):
25mM HEPES. pH 7.2
15mM MgCl2
2µM ATP
0.1µCi/well33P-y ATP (10Ci/mMol)
2µM substrate peptide (Biotin-Ahx-SFNDTLDFD) SEQ ID:No. 2.
Reaction Mix B is prepared as follows at 4°C:
Reaction Mix B (Enzyme Mix)
25mM HEPES, pH 7.2
15mM MgCl2
0.15mg/mlBSA
2mMDTT
2-10 nM PLK1 kinase domain
Reaction Mix A (20µl) is added per well. Reaction Mix B (20µl) is added per well.
Incubate 1.5hrs. at RT. The enzymatic reaction is stopped with 175µl of SPA/EDTA
bead mix (29mM EDTA, 2.5 mg/ml Streptavidin-coated SPA in Standard Dulbecco"s
PBS (without Mg2+ and Ca2+), 60µM ATP). Plates are sealed spun (after a 1 hr
incubation at RT) at 1,000 x g for 7 min or settled overnight, then plates counted in
Packard TopCount for 30 seconds/well.
C. Results
The data obtained is reported in Table 1 below. In Table 1, + - plC50 plC50 5-7; +++ - plC50 >7.
II. Methylene Blue Growth Inhibition Assay
Normal Human foreskin fibroblasts (HFF) and human colon (HCT116, RKO), lung
(H460), prostate (PC3), and breast tumor (MCF7) cell lines were cultured in high
glucose DMEM (Life Technologies) containing 10% fetal bovine serum (FBS) at 37°C in
a humidified 10% CO2, 90% air incubator. Cells were harvested using trypsin/EDTA,
counted using a haemocytometer, and plated in 100 µl of the appropriate media, at
the following densities, in a 96-well tissue culture plate (Falcon 3075): HFF 5,000
Cells/well, HCT116 3,000 cells/well, RKO 2,500 cells/well, H460 2,000 cells/well, PC3
8,000 cells/well, MCF7 4,000 cells/well. The next day, compounds were diluted in
DMEM containing 100 u.g/ml gentamicin, at twice the final required concentration,
from 10 mM stock solutions in DMSO. 100 µl/well of these dilutions were added to
the 100 µl of media currently on the cell plate"s. Medium containing 0.6% DMSO was
added to control wells. Compounds diluted in DMEM were added to all cell lines. The
final concentration of DMSO in all wells was 0.3%. Cells were incubated at 37°C, 10%
CO2 for 3 days. Medium was removed by aspiration. Cell biomass was estimated by
staining cells with 90 µl per well methylene blue (Sigma M9140,0.5% in 50:50
ethanokwater), and incubation at room temperature for at least 30 minutes. Stain
was removed, and the plates rinsed under a gentle stream of water, and air-dried. To
release stain from the cells 100 µl of solubilization solution was added (1% N-lauroyl
sarcosine, Sodium salt, Sigma L5125, in PBS), and plates were shaken gently for about
30 minutes. Optical density at 620 nM was measured on a microplate reader. Percent
inhibition of cell growth was calculated relative to vehicle treated control wells.
Concentration of compound that inhibits 50% of cell growth (IC50) was interpolated
using nonlinear regression (LevenbergrMarquardt) and the equation, y = Vnax*(1-
(x/(K+x))) + Y2, where "K" was equal to the ICso. The data obtained reported in Table 1
below* In Table 1, + * 10->30uM;++ = 1 -10 uM: +++ = SEQUENCE LISTING
SmithKline Beecham Corporation
THIOPHENE COMPOUNDS
PU4870
to be assigned

60/402,008
2002-08-08
2
FastSEQ.for Windows Version 4.0
1
11
PRT
baculovirus infected T.ni cells
1
Met Lys Lys Gly His His His His His His Asp
1 5 10
2
9
PRT
Artificial Sequence

optimized PLK peptide substrate
2
Ser Phe Asn Asp Thr Leu Asp Phe Asp
1 5
CLAIMS
1. A compound of formula (I):
wherein:
R1 is selected from the group consisting of H, alkyl, alkenyl, alkynyl, -C(0)R7, -CO2R7,
-C(O)NR7R8, -C(O)NtR7)OR8, -C(O)N(R7)-RZ-OR8, -C(O)N(R7)-Ph,
-C(O)N(R7)-R2-Ph, -C(O)N(R7)C(O)R8, -C(O)N(R7)CO2R8, -C(O)N(R7)C(O)NR7R8,
-C(O)N(R7)S(O)2R8, -R2-OR7, -R2-O-C(O)R7, -C(S)R7, -C(S)NR7R8, -C(S)N(R7)-ph,
-C(S)N(R7)-R2-Ph. -R2-SR7, -C(=NR7)NR7R8, -C(=NR7)N(R8)-Ph,
-C(=NR7)N(R8)-R2-Ph, -R2-NR7R8. -CNr -OR7, -S(O)fR7, -S(O)2NR7R8,
-S(0)2N(R7)-Ph, -S(O)2N(R7)-R2-Ph, -NR7R8, N(R7)-Ph, -N(R7)-R2-Ph, -N(R7)-SO2R8
and Het;
Ph is phenyl optionally substituted from 1 to 3 times with a substituent selected from
the group consisting of halo, alkyl, -OH, -R2-OH, -O-alkyl. -R2-O-alkyl, -NH2,
-N(H)alkyl, -N(alkyl)2, -CN and -N3;
Het is a 5-7 membered heterocycle having 1,2,3 or 4 heteroatoms selected from N, O
and S, or a 5-6 membered heteroaryl having 1,2,3 or 4 heteroatoms selected
from N", 0 and S, each optionally substituted from 1 to 2 times with a
substituent selected from the group consisting of halo, alkyl, oxo, -OH, -R2-OH,
-O-alkyl, -R2-O-alkyl, -NHz, -N(H)alkyl, -N(alkyl)2, -CN and -N3;
Q1 is a group of formula: -(R2)a-(Y1)b(R2)c-R3
a, b and c are the same or different and are each independently 0 or 1 and at least
one of a or b is 1;
n is 0,1,2,3 or 4;
Q2 is a group of formula: -(R2)aa-(Y2)bb-(R2)cc-R4
or two adjacent Q2 groups are selected from the group consisting of alkyl,
alkenyl, -OR7, -S(O)fR7 and -NR7R8 and together with the carbon atoms to
which they are bound, they form a C5-6cycloalkyl, C5-6cycloalkenyl, phenyl, 5-7
membered heterocycle having 1 or 2 heteroatoms selected from N, 0 and S, or
5-6 membered heteroaryl having 1 or 2 heteroatoms selected from N, 0 and S;
aa, bb and cc are the same or different and are each independently 0 or 1;
each Y" and Y2 is the same or different and"is independently selected from the group
consisting of -O-, -S(O)f-, -N(R7)-, -C(0)-, -OC(O)-, -COz-, -C(O)N(R7)-,
-C(O)N(R7)S(O)2, -OC(O)N(R7)-, -OS(O)2, -S(O)2N(R7)-, -S(O)2N(R7]C(O)-,
-N(R7)S(O)2-, -N(R7)C(O)-, -N(R7)CO2- and -N(R7)C(O)N(R7)-;
each R2 is the same or different and is independently selected from the group
consisting of alkylene/alkenylene and alkynylene;
each R3 and R4 is the same or different and is each independently.selected from the
group consisting of H, halo, alkyl. alkenyl, alkynyl, -C(O)R7r -C(O)NR7R8, -CO2R7,
-C(S)R7, -C(S)NR7RB, -C(=NR7)RB, -C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)fR7,
-S(O)2NR7Rfl,-NR7R8,-N(R7)C(O)R8,-N(R7)S(O)2R8,-NO2,-CN,-N3and a group of
formula (ii):
wherein:
Ring A is selected from the group consisting of C5-10cycloallcyl,
C5-10cycloalkenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
•heteroatoms selected from N, 0 and S and 5-10 membered heteroaryl
having 1,2 or 3 heteroatoms selected from N, O and S
each d is 0 or 1;
e is 0, 1,2,3 or 4;
each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyl, alkenyi, alkynyl, cycloalkyi, cycloalkenyl, Ph,
Het, -CH(OH)-R2-OH, -C(O)R7, -CO2R7, -CO2-R2-Ph, -CO2-R2-Het,
-C(O)NR7R8, -C(O)N(R7)C(O)R7, -C(O)N(R7)CO2R7, -C(O)N(R7)C(O)NR7RB,
-C(0)N(R7)S(0)2R7, -C(S)R7r -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8,
-CR7=N-OR8, -O, -OR7, -0C(0)R7T -OC(O)Ph, -OC(O)Het, -OC(O)NR7R8,
-O-R2-S(0)2R7, -S(0)fR7, -S(0)2NR7R8, -S(O)iPh, -S(O)zHet, -NR7R8,
. -N(R7)C(O)R8, ~N(R7)CO2R8, -N(R7)-R2-CO2R3, -N(R7)C(O)NR7R8,
-N(R7)-R2-C(O)NR7R8, -N(R7)C(O)Ph, -N(R7)C(O)Het, -N(R7)Ph, -N(R7)Het,
-N(R7)C(O)NR7-R2-NR7RB, -N(R7)C(O)N(RJ)Ph, -N(R7)C(O)N(R7)Het,
-M(R7)C(0)N(R7)-R2-Het, -N(R7)S(O)2R8, -N(R7)-R2-S(O)2R8, -NO2, -CN and
-N3;
wherein when Q" is defined where b is 1 and c is O, R3 is not halo, -C(O)R7, -C(O)NR7Rs,
-C0zR7, -C(S)R7, -C(S)NR7R8r -C(=NR7)R8t -C(=NR7)NR7R8, -CR7=N-OR7, -OR7,
-S(0)fR7, -S(O)2NR7R8, -MR7R8, -N(R7)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN or -N3;
wherein when Q2 is defined where bb is 1 and cc is Or R4 is not halo, -C(O)R7,
-C(O)NR7R8, -CO2R7, -G(S)R7; -C(S)NR7R8, -C(=NR7)R3, -C(=NR7)NR7R8,
-CR7-N-OR7, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8, -N(R7)C(O)R8, -N(R7)S(O)2R8,
-NO2, -CN or -N3;
R5 is selected from the group consisting of H, halo, alkyl, cycloalkyl, OR7, -S(O)fR7,
-NR7R8, -NHC(O)R7, -NHC(O)NR7R8 and -NHS(O)2R7;
f is 0,1 or 2; and
each R7 and each R8 are-the same or different and are each independently selected
from the group consisting of H, alkyl. alkenyl, alkynyl, cycloalkyl and
cycloaikenyl;
wherein when R1 is -CO2CH3 and n is 0, Q1 is not -OH;
or a pharmaceuticallyacceptable salt, solvate or physiologically functional derivative
thereof.
2. A compound as claimed in claim 1 selected from the group consisting of:
5-(5,6-Dimethoxy-1 H-benzimidazol-1-yl)-3-{[2-(trifluoromethyl)-
benzylloxy}thiophene-2-carboxamide;
5-(5-(Methyloxy)-.6-{[2-(4-methyl,-1-pipera2inyl)ethyl]oxy}-1H-benzimidazol-1-yl)-
3r({[2-(triflupromethyl)phenyl]methyl}oxy)-2-thiophenecarboxamide;
3-[1-(2-Chlorophenyl)ethoxy]-5-(S,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
. 5-(5,G-Dim"ethoxy-1H-bcnzimtdazo-1-yl)-3-[1 -(2-methylphenyl)ethoxy] thiophene-2-
carboxamide;
5-(5-Amino-1H-benzimidaz6l-1-yl)-3-[1-(2-chlbrophenyl)ethoxy]thiophene-2-
carboxamide;
5-{6-[(4-Piperidinylmethyl)oxy]-1H-benzimidaza[-1-yl}-3-({[2-
(trifluoromethyl)phenyl]-methyl}oxy}-2-thiophenecarboxamide;
5-[6r(Methyloxy)-.5-{[3-(2-oxo-1-pyrroIidinyl)propyl]oxy}-1H-benzimidazol-1-yl)-3-
({[2-(trifiuoromethyl)phenyl]methyl}dxy)-2-thiophenecarboxarrtide;
5-[6-{[3-(Dimethylamino)propyl]oxy}-5-(methyIoxy)-1H-benzimidazol-1-yl]-3-({[2-
(trif[uoromethyl]phenyl]methyl}oxy}-2-thiophenecarboxamide;
5-(5-(Methyloxy)-6-{[2-(4-morphoIinyl)ethyl]oxy}-1H-benzimidazol-1-yl)-3-({[2-
{trifluoromethynphenyl]methyl}oxy}-2-thiophenecarboxamide;
5-[6-(2-Marp.holin-4-ylethoxy)-1 H-benzimidazol-1 -yl]-3-{[2-
(trifluoromethyl)benzyfloxy}thiophene-2-carboxamide;
5-[6-(2-Pyrrolidin-1-ylethoxy)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethy!)benzyl]oxy}thiophene-2-carboxamide;
5-[5-Fluoro-6-(2-morphoIin-4-ylethoxy)-1H-benzimidazol-1-yl]-3-{[2-
(trifluoromethyl)benzyl]oxy}thiophene-2-carboxamide;
5-[6-(MethylsuIfonyl)-1H-benzimidazol-1-yl)-3-{[2-(trifIuoromethyl)benzyl]oxy}-
thiophene-2-carboxamide;
3-[(3-Bromopyridin-4-yl)methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
S-(5,6-Diraethoxy-1H-benzlmidazol-1-yl)-3-{[2-(trifluoromethoxy)benzyl]uxyi
thiophene-2-carboxamide;
3-{[2-(DifIuoromethoxy)benzyl]oxy}-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
3-[(2-Chloropyridm-3-yl)methoxyl-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
5-(5,6-Dimcthoxy-1 H-benzimtdazol-1-yl)-3-[(2-fIuoropyridin-3-
yl)methoxy]thiophene-2-carboxamide;
3-[(2-Aminopyridin-4-yl)methoxyl-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
3-[(6-Chloro-1,3-benzodioxol-5-yl)methoxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-
yl)thiophene-2-carboxamide;
5-(5,6-Dimcthoxy-1H-benzimidazol-1-yl)-3-[(2-nitrobenzyl)oxy]thiophene-2-
carboxamide;
. 3-[(3rAminobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
5-(6-Bromo-1H-benzimidazoI-1-yl)-3-{[2-(trifluoromethyl)benzyl]-oxy}thiophene-2-
carboxamide;
3-[(2,6-Dichlorobenzyl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thiophene-2-
carboxamide;
3-[(2-BromobenzYl)oxy]-5-(5,6-dimethoxy-1H-benzimidazol-1-yl)thlophene-2-
carboxamide;
5-(5,6-Dimethoxy-1H-benzimidazol-1-yl)-3-[(2-formylben2yl)oxy]thiophene-2-
carboxamide;
5-(1H-Benzimidazol-1-yl)-3-{[2-(trlfluoromethyl)benzyl3oxy}thiophene-2-
carboxamide;
5-(lH-Benzimidazol-1-yl)-3-[(2-nttrobenzyl)oxy]thiophene-2-carboxamide;
5-[6-Methoxy-1H-benzimidazol-1-y!)-3-{[2-(trfflupromethyl)benzyl]oxy}thiophene-
2-carboxamide;
2-(Aminocarbonyl)-5-(5,6-dlmethoxy-1 H-benzimidazol-1-yl)thien-3-yl 2-
methylbenzenesulfonate
and pharmaceutically acceptable salts, solvates and physiologically functional
derivatives thereof.
3. A pharmaceutical composition comprising a compound as claimed in any of
claims 1-2.
4. The pharmaceutical composition as claimed in claim 3, optionally comprising a
pharmaceutically acceptable carrier, diluent or excipient.
5. The pharmaceutical composition as claimed in claim 3, optionally comprising a
chemotherapeutic agent.
6. A pharmaceutical composition as claimed in claim 3, for treating a condition
mediated by PLK in an animal.
7. A pharmaceutical composition as claimed in claim 3, for treating a susceptible
neoplasm in an animal.
8. A pharmaceutical composition as claimed in claim 7, wherein said susceptible neoplasm is
selected from the group consisting-of breast cancer, colon cancer, lung cancer,
prostate cancer, lymphorrta, leukemia, endometriai cancer, melanoma, ovarian cancer,
pancreatic cancer, squamous carcinoma, carcinoma of the head and neck, and
esophageal carcinoma.
wherein;
R1 Is selected from the group consisting of H, alkyl, alkenyi, alkynyl, -C(O)R7,
-CO2R7, -C(O)NR7R8, -C(O)N(R7)OR8, -C(O)N(R7)-R2-OR8,
-C(O)N(R7)-Ph, -C(O)N(R7)-R2-Ph, -C(O)N(R7)C(O)R8,
-C(O)N(R7)CO2R8, -C(O)N(R7)C(O)NR7R8, -C(O)N(R7)S(O)2R8,
-R2-OR7, -R2-O-C(O)R7, -C(S)R7, -C(S)NR7R8, -C(S)N(R7)-Ph,
-C(S)N(R^)-R2-Ph, -R2-SR7, -C(=NR7)NR7R8,-C(=NR7)N(R8)-Ph,
-C(=NR7)N(R8)-R2-Ph, -R2-NR7R8, -CN, -OR7, -S(O)fR7, -S(O)2NR7R8,
-S(O)2N(R7)-Ph, -S(O)2N(R7)-R2-Ph, -NR7R8, N(R7)-Ph, -N(R7)-R2-Ph,
-N(R7)-SO2R8 and Het;
Ph is phenyl optionally substituted from 1 to 3 times with a substituent selected from the
group consisting of halo, alkyl, -OH, -R2-OH, -O-aikyi, -R2-O-alkyl, -NH2, -N(H)alkyi, -
N(alkyl)2,-CN and-N3;
Het is a 5-7 membered heterocycle having 1, 2,3 or 4 heteroatoms selected from N, O and
S, or a 5-6 membered heteroaryl having 1,2,3 or-4 heteroatoms selected from N, Q
and S, each optionally substituted from .1 to 2 times with a substituent selected from
the group consisting of halo, alkyl, oxo, -OH, -R2-OH, -O-alkyl, -R2-O-alkyl, -NH2, -
N(H)alkyl,
-N(alkyl)2, -CN and -N3;
n is 0,-1, 2,3 or 4;
Q2 is a group of formula: -(R2)aa-(Y2)bb-(R2)cc-R4
or two adjacent Q2 groups are selected-from the group consisting of alkyl, alkenyl, -
OR7, -S(6)(R7 and -NR7R8 and together with the carbon atoms to which they are
bound, they form a C5-6cycloalkyi, C5-6cydoalkenyl, phenyl, 5-7 membered
heterocycle having i or 2 heteroatoms selected from N, O and S, or-5-6 membered
heteroaryl having t. or 2 heteroatoms selected from N, O and S;
aa, bb and cc are the same or different and are each independently 0 or 1;
each Y2 is the same or different and is independently selected from the group consisting of -
O-, -S(O)f-, -N{R7)-, -C(O)-, -OC(O)-, -C02-, -C(O)N(R7)-, -C(O)N(R7)S(O)2-, -
OC(O)W(R7)-,-OS(O)2-1-S{O)2N(R7)-;
-S(O)2N(R7)C(O)-, -N(R7)S(O)2-, -N(R7)G(O)-, -N(R7)CO2- and
-N(R7)C(O)N(R7)-;
each R2 is the same .or different and is-independently selected from the group consisting of
-alkylene, alkenylene and aikynylene;
each R4 is the same ondifferent and is each independently selected from the group
consisting of H; halo, alkyi, alkenyl, aikynyl, -C(O)R7,
-C(O)NR7R8, -CO2R7, -C(S)R7, -C(S)NR7R8, -C(=NR7)R8r
-C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(6)fR7, -S(O)2NR7R8, -NR7R&,
-N(R7)C(O)R8-, -N(R7}S(O)2R8, -NO2, -CN, -N3and a group of formula (ii):
wherein;
Ring A is selected from the group consisting of C5-10cycloalkyl,
C5-10cycloaikenyl, aryl, 5-10 membered heterocycle having 1,2 or 3
heteroatoms selected from H, 0 and S and 5-10 membered heteroaryi having
1, 2 or 3 heteroatoms selected from N, O and S
each d is 0 or 1;
e is 0, 1,2, 3 or 4;
each R6 is the same or different and is independently selected from the group
consisting of H, halo, alkyi, aikenyl, alkynyi, cycloaikyl, cycioalkenyi, Ph, Het,
CH(OH)-R2-OH,-C(O)R7,-CO2R7,
-CO2-R2-Ph,CO2-R2-Het, -C(O)NR7R8, -C{O)N(R7)C(O)R7,
-C(O)N(R7)CO2R7, -C(O)N(R7)C(O)MR7R8, -C(O)N(R7)S(O)2R7,
-C(S)R7, -C(S)NR7R8, -C(=NR7)R8, -C(=NR7)NR7R8,
-CR7=N-OR8, =O, -OR7, -OC(O)R7, -OC(O)Ph, -OC(O)Het,
-OC(O)NR7R8, -O-R2-S(O)2R7, -S(O)fR7, -S(O)2NR7R8, -S(O)2Ph, -S(O)2Het,
NR7R8, -N(R7)C(O)R8, -N(R7)CO2R8,
-N(R7)-R2-CO2R8, -N(R7)C(O)NR7R8, -N(R7)-R2-C(O)NR7R8,
-N(R7)C(O)Ph, -N(R7)C(O)Het, -N(R7)Pfi,"-N(R7)Het,
-N(R7)C(O)NR7-R2-NR7R8, -N(R7)C(O)N(R7)Ph,
-N(R7)C(O)N(R7)Het, -N(R7)C(O)N(R7)-R2-Het, -N(R7)S(O)2R8,
-N(R7)-R2-S(O)2R8, -NO2, -CN and -N3;
wherein when Q2 is defined where bb is 1 and cc is O, R4 is not halo, -C(O)R7,
-C(O)NR7R8, -CO2R7, -C(S)R7, -C(S)NR7R8,-C(=NR7)R8r
-C(=NR7)NR7R8, -CR7=N-OR7, -OR7, -S(O)fR7, -S(O)2NR7R8, -NR7R8,
-N(R7.)C(O)R8, -N(R7)S(O)2R8, -NO2, -CN or -N3; -
R5 is selected from the group consisting of H, halo, alkyl, cycioalkyl, OR7,
-S(O)fR7, -NR7R8, -NHC(O)R7, -NHC(O)NR7R8 and -NHS(O)2R7;
f is 0, 1 or 2; and
each R7 and each R8 are the same or different and are each independently selected from
the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl; and
each R9 is the same or different and is selected from H, halo and alky!;
or a pharmaceutically acceptable salt, solvate or physiologically functional derivative
thereof.
10. An R-isomer of a compound as claimed in claim 9.
11. An R-isomer of a compound selected from
5-(5-Amino-1H-benzimidazol-1-yl)-3-[1-(2-chlorophenyl)ethoxy]thiophene-2-carboxamide;
3-[1-(2-Chlorophenyl)ethoxy]-5-(5,5-dimethoxy-1H-benzimidazol-1-yi)th!ophene-2-
carboxamide;
5-(5,6-Dimethoxy-1H-benzimidazol-1 -yl)-3-[1 -(2-methylphenyi)ethoxy] thiophene-2-
carboxamide;
and pharmaceutically acceptable salts, solvates and physiologically functional derivatives
thereof.
12. A compound of formula (Ib):
wherein:
R1 is -C(O)NH2;
each R9 is the same or different and is selected from H, halo and alkyl;
Ring A is phenyl;
d is 0;
e is 1;
R6 is trifluoromethyl;
n is 1 and Q2 is at C-6;
R5 is H;
f is 0, 1 or 2; and
each R7 and each R8 are the same or different and are each independently selected from the group
consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl; and
Q2 is a group of formula: -(R2)aa-(Y2)bb-(R2)cc-R4, wherein:
aa is 0;
bb is 0;
cc is 1 and (R2)cc is C1-3alkylene; and
R4 is a group of formula (ii):
wnerein:
Ring A is selected from the group consisting of morpholine, piperidine, piperazine, phenyl,
pyrrolidinone, imidazolidinone and pyrrolidine
d is 0;
e is 1; and
R6 is selected from the group consisting of H, halo, alkyl, =0, -OR7, -S(O)fR7,
-S(O)2NR7R5 and -NR7R8;
or a pharmaceutically acceptable salt thereof.
The present invention provides benzimidazole thiophene compounds of formula (I):
pharmaceutical compositions containing the same, processes for preparing the same and their use as
pharmaceutical agents, including their use for the treatment of cancer.

Documents:

321-KOLNP-2005-CORRESPONDENCE 1.1.pdf

321-kolnp-2005-granted-abstract.pdf

321-kolnp-2005-granted-assignment.pdf

321-kolnp-2005-granted-claims.pdf

321-kolnp-2005-granted-correspondence.pdf

321-kolnp-2005-granted-description (complete).pdf

321-kolnp-2005-granted-examination report.pdf

321-kolnp-2005-granted-form 1.pdf

321-kolnp-2005-granted-form 13.pdf

321-kolnp-2005-granted-form 18.pdf

321-kolnp-2005-granted-form 3.pdf

321-kolnp-2005-granted-form 5.pdf

321-kolnp-2005-granted-gpa.pdf

321-kolnp-2005-granted-letter patent.pdf

321-kolnp-2005-granted-reply to examination report.pdf

321-kolnp-2005-granted-specification.pdf

321-KOLNP-2005-PA.pdf


Patent Number 215541
Indian Patent Application Number 321/KOLNP/2005
PG Journal Number 09/2008
Publication Date 29-Feb-2008
Grant Date 27-Feb-2008
Date of Filing 02-Mar-2005
Name of Patentee SMITHKLINE BEECHAM CORPORATION
Applicant Address ONE FRANKLIN PLAZA, PHILADELPHIA, PA 19101
Inventors:
# Inventor's Name Inventor's Address
1 ANDREWS, III, CLARENCE, W CLAXOSMITHKLINE, FIVE MOORE DRIVE, PO BOX 13398, RESEARCH TRIANGLE PARK, NC 27709
2 VEAL, JAMES, MARVIN 8916 WEAVER CROSSING ROAD, APEX, NC27502
3 CHEUNG, MUI -DO-
4 DAVIS-WARD, RONDA, G -DO-
5 DREWRY, DAVID, HAROLD -DO-
6 EMMITTE, KYLE, ALLEN -DO-
7 HUBBARD, ROBERT, DALE -DO-
8 KUNTZ, KEVIN, W -DO-
9 LINN, JAMES, ANDREW -DO-
10 MOOK, ROBERT, ANTHONY -DO-
11 SMITH, GARY, KEITH -DO-
PCT International Classification Number C 07 D 409/04
PCT International Application Number PCT/US2003/024272
PCT International Filing date 2003-08-04
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/402,008 2002-08-08 U.S.A.