Note: Descriptions are shown in the official language in which they were submitted.
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
PRO-NEUROGENIC COMPOUNDS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Application No. 13/594,223 filed
August 24, 2012, which is a
continuation-in-part application of U.S. Application No. 13/177,981 filed July
7, 2011, which is a
continuation-in-part of U. S. Application No. 12/832,056 filed July 7, 2010,
which is a continuation-in-part of
U. S. Application No. 12/685,652 filed January 11, 2010, which claims the
benefit of and priority to U.S.
Provisional Application No. 61/143,755, filed January 9, 2009; each of these
prior applications is incorporated
herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
This invention was made with government support under Grant Numbers
5DPI0D00027605,
5R37MH05938809, and 1R01MH087986, which were awarded by the National Institute
of Health; the
Government has certain rights in the invention.
TECHNICAL FIELD
This invention relates generally to the discovery of pro-neurogenic compounds
capable of promoting
neurogenesis and/or reducing neuronal cell death.
BACKGROUND
It is now accepted that the adult vertebrate brain fosters the birth and
functional incorporation of newly
formed neurons (Goldman and Nottebohm, Proc Natl Acad Sci USA 1983, 80: 2390-
2394; Paton and
Nottebohm, Science 1984, 225, 1046-1048; Burd and Nottebohm, J Comp Neurol
1985, 240:143-152).
However, it was long thought that no new neurons could be added to the adult
mammalian brain. This dogma
was challenged in the 1960's when autoradiographic evidence of new neuron
formation in the hippocampal
dentate gyms, olfactory bulb, and cerebral cortex of the adult rat was
presented (Altman, J. Science 1962, 135,
1127-1128; Altman, J. J Comp Neurol 1966, 128:431-474; Altman, Anat Rec 1963,
145:573-591; Altman and
Das, J. Comp. Neurol. 1965, 124, 319-335; Altman and Das, J Comp Neurol 1966,
126:337-390). It is now
accepted that within all mammalian species, including humans (Eriksson et al.,
Nat. Med. 1998, 4(11), 1313-
1317), there are two major reservoirs of neuronal stem cells, one located in
the subgranular zone (SGZ) of the
hippocampal dentate gyms and another in the subventricular zone (SVZ) (Gross,
Natl. Rev. 2000, 1, 67-72).
Neural stem cells in the SVZ facilitate formation of new neurons that migrate
rostrally to populate the
olfactory bulb, while neural stem cells in the SGZ produce neurons that
integrate locally in the granular layer
of the dentate gyms, a region of the hippocampus that exhibits lifelong
structural and functional plasticity.
1
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The process of new neuron formation in the adult mouse brain can be influenced
by environmental,
chemical and genetic variables. As demonstrated by Gage and colleagues,
neurogenesis in the adult mouse
brain is enhanced when animals are exposed to an enriched environment
(Kempermann et al., Nature 1997,
386, 493-495) or able to exercise voluntarily (van Praag et al., Nat. Neuro-
sci. 1999, 2, 266-270). More
recently, anti-depressant drugs have been shown to enhance levels of adult
neurogenesis in animals, including
humans (Schmidt et al., Behav Pharmacol. 2007 Sep;18(5-6):391-418; Boldrini et
al.,
Neuropsychopharmacology 2009, 34, 2376-2389). Among many genes reported to
impact adult neurogenesis
is the gene encoding neuronal PAS domain protein 3 (NPAS3), a central nervous
system (CNS)-specific
transcription factor that has been associated with schizophrenia and bipolar
disorder (Kamnasaran et al., J.
Med. Genet. 2003, 40, 325-332; Pickard et al., Am. J. Med. Genet. B.
Neuropsychiatr. Genet. 2005, 136B, 26-
32; Pickard et al., Aim. Med. 2006, 38, 439-448; Pickard et al., Mol.
Psychiatry 2009, 14, 874-884; Lavedan
et al., Pharmacogenomics 2008, 9: 289-301). Animals missing both copies of the
NPAS3 gene suffer a
profound loss of adult hippocampal neurogenesis coupled with significant
behavioral deficits (Pieper et al.,
Proc. Natl. Acad. Sci. USA 2005, 102, 14052-14057). Knowing that impaired post-
natal neurogenesis elicits
unfavorable phenotypic deficits, it is predicted that pro-neurogenic chemical
compounds should exhibit
favorable therapeutic benefits.
SUMMARY
This invention relates generally to compounds that promote the generation or
the survival of existing
neurons in the mammalian brain. For the purpose of simplicity these compounds
are referred to as being pro-
neurogenic. In certain embodiments, the compounds promote the generation or
survival of neurons in the
post-natal mammalian brain. In certain embodiments, the compounds promote the
survival, growth,
development and/or function of neurons, particularly CNS, brain, cerebral, and
hippocampal neurons. In
certain embodiments, the compounds stimulate post-natal hippocampal
neurogenesis, which while not wishing
to be bound by theory, is believed to represent a therapeutic target for a
variety of neuropsychiatric and
neurodegenerative diseases, including (but not limited to) schizophrenia,major
depression, bipolar disorder,
normal aging, epilepsy, traumatic brain injury, post-traumatic stress
disorder, Parkinson's disease, Alzheimer's
disease, Down syndrome, spinocerebellar ataxia, amyotrophic lateral sclerosis,
Huntington's disease, stroke,
radiation therapy, chronic stress, abuse of neuro-active drugs (such as
alcohol, opiates, methamphetamine,
phencyclidine, and cocaine), retinal degeneration, spinal cord injury, and
peripheral nerve injury. In certain
embodiments, the compounds stimulate post-natal hypothalamic neurogenesis,
which can provide therapeutic
benefits in weight management, such as physiological weight loss associated
with various conditions,
including but not limited to, normal aging, chemotherapy, radiation therapy,
stress, drug abuse, anorexia, as
well as other diseases discussed herein.
2
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The presently disclosed embodiments also feature compositions (e.g.,
pharmaceutical compositions)
that include such compounds as well as methods of making, identifying, and
using such compounds. Other
features and advantages are described in, or will be apparent from, the
present specification and accompanying
drawings.
Accordingly, in one aspect, methods for promoting post-natal mammalian
neurogenesis and/or
reducing neuronal cell death in a subject in need thereof are described, the
method comprising administering
an effective amount of a compound having formula (I) or a pharmaceutically
acceptable salt thereof:
R4
R'
R3
R2 101 C3
/ 2
N%C
\
L ¨A
R1
(I)
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
Ci-C6 haloalkyl, C2-C6
alkynyl, cyclopropyl, -N3, cyano, -NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and nitro;
R and R' are defined according to (1), (2), (3), (4), or (5) below:
(1) R and R' together with C2 and C3, respectively, form a fused phenyl
ring having formula (II):
R6
R7
C2 R8
VINV"
(II)
wherein each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, Ci-C6
thiohaloalkoxy, Ci-C6 alkyl, Ci-C6
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; OR
3
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) each of R and R' is, independently, hydrogen, C1-C6 alkyl, or C1-C6
haloalkyl; OR
(3) R and R' together with C2 and C3, respectively, form a fused
heterocyclic ring containing from
5-6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C6 alkyl),
NC(0)(C1-C6 alkyl), 0, and S; and wherein said heterocyclic ring is optionally
substituted with from 1-3
independently selected Ra; OR
(4) R and R' together with C2 and C3, respectively, form a fused C5-C6
cycloalkyl ring that is
optionally substituted with from 1-4 independently selected Ra; OR
(5) R and R' together with C2 and C3, respectively, form a fused heteroaryl
ring containing from
5-6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C3 alkyl), 0,
and S; and wherein said heteroaryl ring is optionally substituted with from 1-
3 independently selected Rb;
Ll is:
(i) C1-C3 straight chain alkylene, which is optionally substituted with from 1-
2 independently
selected Re; or
(ii) a bond that directly connects N in the 5-membered ring of formula (I) to
A in formula (I);
L2 is:
(i) C1-C3 straight chain alkylene, which is optionally substituted with from 1-
2 independently
selected Re; or
(ii) a bond that directly connects A in formula (I) to Z in formula (I);
A is:
(i) cRAI A2,
K wherein each of RA1 and RA2 is independently selected
from hydrogen, halo, CI-
C3 alkyl, or OR9; or
(ii) C=0; or
(iii) C3-05 cycloalkylene that is (a) substituted with 1 oxo; and (b)
optionally further
substituted with from 1-4 independently selected le; or
(iv) heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of
the ring atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heterocycloalkylene is
(a) substituted with 1 oxo; and (b) is optionally further substituted with
from 1-4 independently
selected Ra;
Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2 or
4
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected Ra;
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
(viii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(ix) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(x) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(xi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R9 is hydrogen; or C1-C3 alkyl that is optionally substituted with hydroxyl or
C1-C3 alkoxy;
5
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (1) below:
(a) hydrogen;
(b) C6-Cio aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl);
(0 C2-C6 alkenyl or C2-C6 alkynyl;
(g) C8-C arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(h) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(i) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(j) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
6
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(k) C3-C8 cycloalkyl or C3-C8 cycloalkenyl, each of which is optionally
substituted with from
1-4 independently selected Ra; and
(1) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
R.12 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb; or
(iii) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
or
(iv) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(v) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vii) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
7
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R" is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(iii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(iv) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(v) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
8
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
Ra at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), Ci-
C6 alkyl, Ci-C6 haloalkyl, -
NH2, -NH(Ci -C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6 haloalkoxy; Ci-C6 thioalkoxy; Ci-C6 thiohaloalkoxy; -
0-(CH2)1-3-
[0(CH2)1-3]1_3-1-1; -Ci-C6 alkyl, Ci-C6 haloalkyl,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; Ci-C6 thiohaloalkoxy; Ci-C6 alkyl,
and C1-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, Ci-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, CI-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, Ci-C6 alkoxy;
Ci-C6 thioalkoxy; CI-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(Ci-C6 alkyl); N(Ci-C6 alky1)2; -
NHC(0)(Ci-C6 alkyl); cyano;
-C(0)H; -C(0)(Ci-C6 alkyl); -C(0)(Ci-C6 haloalkyl); C(0)0H; -C(0)0(C-C6
alkyl); -C(0)NH2; -
C(0)NH(Ci-C6 alkyl); C(0)N(Ci-C6 alky1)2; -S02(Ci-C6 alkyl); -SO2NH2; -
SO2NH(Ci-C6 alkyl); -SO2N(Ci-C6
9
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
alky1)2; and L3-(C1-C6 alkylene)-Cy, where in L3 is a -0-, -NH-, -NCH3-, -C(0)-
, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-, and Cy is a saturated, partially unsaturated or
aromatic carbocyclic or heterocyclic
ring system;
or a pharmaceutically acceptable salt thereof
In some embodiments, one or more of (A), (B), or (C) apply.
(A) Provided that when R and R' are defined according to definition
(3), then:
(i) each of L1 and L2 must be C1-C3 alkylene, which is optionally substituted
with from 1-2
independently selected Re when A is CH2; or
(ii) Z must be other than heteroaryl containing from 5-14 (e.g., 5-6 or 6)
ring atoms, wherein from 1-6
of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; and wherein said heteroaryl
is optionally substituted with from 1-4 independently selected Rb; e.g., other
than substituted pyridyl, e.g.,
other than pyridyl substituted with C1-C3 alkyl (e.g., CH3), e.g., other than
2 or 6-methylpyridyl.
(B) Each of R1 and R11 cannot be optionally substituted naphthyl
(e.g., each of R1 and R11 cannot
be unsubstituted naphthyl). In embodiments, each of R1 and R11 is other than
optionally substituted naphthyl
(e.g., unsubstituted naphthyl) when R and R' are defined according to
definitions (1), (2), and (4); and A is
CRA1RA2 (e.g., CHOR9, e.g., CHOH), and each of L1 and L2 is C1-C3 alkylene
(e.g., each of L1 and L2 is CH2).
(C) R12 and/or R13 cannot be substituted phenyl. In embodiments,
R12 and/or R13 cannot be
substituted phenyl when R and R' are defined according to definition (1); and
A is CRA1RA2 (e.g., CHOR9,
e.g., CHOH), and each of L1 and L2 is C1-C3 alkylene (e.g., each of L1 and L2
is CH2).
In some embodiments, (A), (B), or (C) applies. In other embodiments, (A) and
(B); or (A) and (C); or
(B) and (C) applies. In still other embodiments, (A), (B), and (C) apply.
In another aspect, methods for promoting post-natal mammalian neurogenesis in
a subject in need
thereof are featured. The method includes administering to the subject an
effective amount of a compound
having formula (I) or a pharmaceutically acceptable salt thereof
R4
R'
R3 /
R2 02 C3
/
N%C --R
\
R1 \
L2---,z
(I)
wherein:
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
Ci-C6 haloalkyl, cyano, -NH2,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
R and R' are defined according to (1), (2), (3), (4), or (5) below:
(1) R and R' together with C2 and C3, respectively, form a fused phenyl
ring having formula (II):
R6
R6.......____ R7
1
....-- C3 .....: .."%.........s.
..--µ ,........
C2 R8
I
µrtrtftr
I (II)
wherein each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6
halothioalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, -NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6
alkyl), and nitro; OR
(2) each of R and R' is, independently, hydrogen, CI-C6 alkyl, or CI-C6
haloalkyl; OR
(3) R and R' together with C2 and C3, respectively, form a fused
heterocyclic ring containing from
5-6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C6 alkyl),
NC(0)(C1-C6 alkyl), 0, and S; and wherein said heterocyclic ring is optionally
substituted with from 1-3
independently selected Ra; OR
(4) R and R' together with C2 and C3, respectively, form a fused C5-C6
cycloalkyl ring that is
optionally substituted with from 1-4 independently selected Ra; OR
(5) R and R' together with C2 and C3, respectively, form a fused
heteroaryl ring containing from
5-6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C3 alkyl), 0,
and S; and wherein said heteroaryl ring is optionally substituted with from 1-
3 independently selected Rb;
Ll is:
(i) C1-C3 straight chain alkylene, which is optionally substituted with from 1-
2 independently
selected Re; or
(ii) a bond that directly connects N in the 5-membered ring of formula (I) to
A in formula (I);
L2 is:
(i) C1-C3 straight chain alkylene, which is optionally substituted with from 1-
2 independently
selected Re; or
(ii) a bond that directly connects A in formula (I) to Z in formula (I);
A is:
11
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(i) cRAI -K A2,
wherein each of RA1 and RA2 is independently selected from hydrogen, halo, CI-
C3 alkyl, or OR9; or
(ii) C=0; or
(iii) C3-05 cycloalkylene that is (a) substituted with 1 oxo; and (b)
optionally further
substituted with from 1-4 independently selected le; or
(iv) heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of
the ring atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heterocycloalkylene is
(a) substituted with 1 oxo; and (b) is optionally further substituted with
from 1-4 independently
selected Ra;
In certain embodiments, A is not CH2. In some embodiments, when A is CRA1RA2,
one of RA1
and RA2 can be hydrogen, halo, C1-C3 alkyl, or OR9; and the other of RA1 and
RA2 can be halo, Ci-C3
alkyl, or OR9.
Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2 or
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected Ra;
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
(viii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(ix) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
12
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(x) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(xi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R9 is hydrogen; or C1-C3 alkyl that is optionally substituted with hydroxyl or
C1-C3 alkoxy;
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (1) below:
(a) hydrogen;
(b) C6-Cio aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl);
(0 C2-C6 alkenyl or C2-C6 alkynyl;
(g) C8-C arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(h) arylheterocyclyl containing from 8-14 ring atoms, wherein:
13
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(i) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(j) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(k) C3-C8 cycloalkyl or C3-C8 cycloalkenyl, each of which is optionally
substituted with from
1-4 independently selected Ra; and
(1) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
R.12 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb; or
(iii) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
or
(iv) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
14
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(v) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vii) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R" is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(iii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(iv) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(v) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
Ra at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, Ci-C6 thioalkoxy,
CI-C6 haloalkoxy, Ci-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), Ci-
C6 alkyl, Ci-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; Ci-C6haloalkoxy; Ci-C6thioalkoxy; Ci-C6thiohaloalkoxy; Ci-
C6 alkyl,
Ci-C6 haloalkyl, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl),
wherein the alkyl portion
of each is optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6
alkyl); -SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
16
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(c1d) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), C i-C6
alkoxy; C1-C6 haloalkoxy; CI-C6 thioalkoxy; CI-C6 thiohaloalkoxy; CI-C6 alkyl,
and C1-C6 haloalkyl;
Ie at each occurrence is, independently selected from halo, CI-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, C1-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; CI-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-Cy, where in L3 is a -0-, -NH-, -NCH3-, -C(0)-
,
-C(0)NH-, -C(0)NCH3-, -NHC(0)-, or -NCH3C(0)-, and Cy is a saturated,
partially unsaturated or aromatic
carbocyclic or heterocyclic ring system;
or a salt (e.g., pharmaceutically acceptable salt) thereof
In some embodiments, one or more of (A), (B), or (C) apply.
(A) Provided that when R and R' are defined according to definition
(3), then:
(i) each of L1 and L2 must be C1-C3 alkylene, which is optionally substituted
with from 1-2
independently selected Re when A is CH2; or
(ii) Z must be other than heteroaryl containing from 5-14 (e.g., 5-6 or 6)ring
atoms, wherein from 1-6
of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; and wherein said heteroaryl
is optionally substituted with from 1-4 independently selected Rb; e.g., other
than substituted pyridyl, e.g.,
other than pyridyl substituted with CI-C3 alkyl (e.g., CH3), e.g., other than
2 or 6-methylpyridyl.
(B) Each of RI and R11 cannot be optionally substituted naphthyl
(e.g., each of RI and R11 cannot
be unsubstituted naphthyl). In embodiments, each of RI and R11 is other than
optionally substituted naphthyl
(e.g., unsubstituted naphthyl) when R and R' are defined according to
definitions (1), (2), and (4); and A is
CRA1RA2 (e.g., CHOR9, e.g., CHOH), and each of L1 and L2 is C1-C3 alkylene
(e.g., each of L1 and L2 is CH2).
(C) R12 and/or R13 cannot be substituted phenyl. In embodiments,
R12 and/or R13 cannot be
substituted phenyl when R and R' are defined according to definition (1); and
A is CRA1RA2 (e.g., CHOR9,
e.g., CHOH), and each of L1 and L2 is C1-C3 alkylene (e.g., each of L1 and L2
is CH2).
17
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
In embodiments, (A), (B), or (C) applies. In other embodiments, (A) and (B);
or (A) and (C); or (B)
and (C) applies. In still other embodiments, (A), (B), and (C) apply.
In another aspect, methods for promoting post-natal mammalian neurogenesis in
a subject in need
thereof are featured. The methods include administering to the subject an
effective amount of a compound
having formula (I) or a pharmaceutically acceptable salt thereof, in which R
and R' together with C2 and C3,
respectively, form a fused phenyl ring having formula (II):
R6
R5....._... R7
1
..\....---.C3.......... ...:,"%..........
C2 R8
I
VINV"
i (H).
For purposes of clarification, it is understood that compounds in which R and
R' together with C2 and
C3, respectively, form a fused phenyl ring having formula (II) correspond to
compounds having the following
general formula:
R6
R5
R4
R3
111 R7
R2 0 N R8
\
W \L2----Z (III)
in which RI, R2, R3, R4, LI, L2, A, and Z can be as defined anywhere herein.
In embodiments, (A), (B), or (C) applies. In other embodiments, (A) and (B);
or (A) and (C); or (B)
and (C) applies. In still other embodiments, (A), (B), or (C) apply.
In another aspect, methods for promoting post-natal mammalian neurogenesis in
a subject in need
thereof are featured. The method includes administering to the subject an
effective amount of a compound
having formula (I) or a pharmaceutically acceptable salt thereof, in which:
each of LI and L2 is CH2;
A is CRAIRA2, wherein one of RAI and RA2 is OR9, and the other is hydrogen.;
Z is -NRI0R11; and
each of le and Ril is independently selected from
18
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(a) hydrogen;
(b) C6-Cio aryl that is optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
C2-C6 alkenyl or C2-C6 alkynyl.
In embodiments, (A), (B), or (C) applies. In other embodiments, (A) and (B);
or (A) and (C); or (B)
and (C) applies. In still other embodiments, (A), (B), and (C) apply.
In one aspect, compositions (e.g., a pharmaceutical composition) are featured,
which includes a
compound of formula (I) (and/or a compound of any of the other formulae
described herein) or a salt (e.g., a
pharmaceutically acceptable salt) thereof as defined anywhere herein and a
pharmaceutically acceptable
carrier. In some embodiments, the compositions can include an effective amount
of the compound or salt. In
some embodiments, the compositions can further include one or more additional
therapeutic agents. These
may include, but are not limited to, antidepressant medications (including
selective serotonin reuptake
inhibitors, tricyclic antidepressants, monoamine oxidase inhibitors, and other
antidepressant medications
including but not limited to venlafaxine, nefazadone, bupropion, mirtazapine,
lithium and trazodone) and
acetylcholinesterase inhibitors (including but not limited to Aricept,
Reminyl, and Exelon).
In another aspect, dosage forms are featured, which includes from about 0.05
milligrams to about
2,000 milligrams (e.g., from about 0.1 milligrams to about 1,000 milligrams,
from about 0.1 milligrams to
about 500 milligrams, from about 0.1 milligrams to about 250 milligrams, from
about 0.1 milligrams to about
100 milligrams, from about 0.1 milligrams to about 50 milligrams, or from
about 0.1 milligrams to about 25
milligrams) of a compound of formula (I) (and/or a compound of any of the
other formulae described herein)
or a salt (e.g., a pharmaceutically acceptable salt) thereof as defined
anywhere herein. The dosage forms can
further include a pharmaceutically acceptable carrier and/or an additional
therapeutic agent.
In one aspect, the compounds of formula (I) themselves (and/or a compound of
any of the other
formulae described herein) or a salt (e.g., a pharmaceutically acceptable
salt) thereof as defined anywhere
herein are featured. In another aspect, any of the formula (I) compounds
specifically described herein are
featured.
In one aspect, compounds having formula (I) are featured.
R4
R'
R3
C3
R2
\
L ¨A
W
L2--,z
(I)
19
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
C1-C6 haloalkyl, C2-C6
alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
R and R' are defined according to (1) or (2) below:
(1) R and R' together with C2 and C3, respectively, form a fused phenyl
ring having formula (II):
R6
R6....._... R7
1
..--µ....=====C3 .....: .7=.............
..........
C2 R8
I
µrtrtAf
1 (H)
wherein each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; OR
(2) R and R' together with C2 and C3, respectively, form a fused R and R'
together with C2 and
C3, respectively, form a fused heteroaryl ring containing 6 ring atoms,
wherein from 1-2 independently
selected ring atoms is N; and wherein said heteroaryl ring is optionally
substituted with from 1-2
independently selected Rb;
each of Ll and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is:
(i) CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen, halo, C1-
C3 alkyl, and OR9, wherein R9 is hydrogen or C1-C3 alkyl that is optionally
substituted with hydroxyl
or C1-C3 alkoxy; or
(ii) C=0; or
(iii) C3-05 cycloalkylene that is (a) substituted with 1 oxo; and (b)
optionally further
substituted with from 1-4 independently selected le; or
(iv) heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of
the ring atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heterocycloalkylene is
(a) substituted with 1 oxo; and (b) is optionally further substituted with
from 1-4 independently
selected Ra;
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2 or
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected Ra;
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
(viii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(ix) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(x) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(xi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
21
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (1) below:
(a) hydrogen;
(b) C6-Cio aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl);
(0 C2-C6 alkenyl or C2-C6 alkynyl;
(g) C8-C arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(h) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(i) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(j) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
22
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(k) C3-C8 cycloalkyl or C3-C8 cycloalkenyl, each of which is optionally
substituted with from
1-4 independently selected Ra; and
(1) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
provided that one of le and Ril must be selected from (b), (c), (g), (h), (i),
(j), and (k);
R.12 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb; or
(iii) C1-C6 alkyl or C1-C6 haloalkyl, each of which is substituted with from 1-
3 Rd;
(iv) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(v) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
23
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vii) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R" is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(iii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(iv) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(v) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
24
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
or
(vi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
Ra at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), Ci-
C6 alkyl, Ci-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; -
0-(CH2)1-3-
[0(CH2)1-3]1_3-1-1; -C1-C6 alkyl, C1-C6 haloalkyl,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl,
and Ci-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, C1-C6 alkoxy, C1-
C6 thioalkoxy, Ci-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, CI-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; C1-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-, -
C(0)-, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-; or a pharmaceutically acceptable salt thereof
In embodiments, 1, 2, 3, 4, 5, or 6 of the following can apply
= provided that R3 and R6 cannot both be hydrogen when A is CH2, and R and
R' are defined
according to definition (1);
= provided that R3 cannot be hydrogen when A is CH2, and R and R' are
defined according to
definition (2);
= provided that R3 and R6 cannot both be chloro when A is CH2, R and R' are
defined according
to definition (1), Z is -0R12, and R12 is unsubstituted phenyl;
= provided that R3 and R6 cannot both be bromo when A is CH2, R and R' are
defined according
to definition (1), Z is -0R12, and R12 is phenyl that is substituted with
pyridyl or alkyl that is
substituted with from 1-3 Re;
= provided that R3 and R6 cannot both be hydrogen when A is CH(CH3), R and R'
are defined
according to definition (1), Z is NRI0R11, RI is CH3, and R11 is
unsubstituted phenyl;
= provided that when A is CRA1RA2, and one of RAland RA2 is OH (i.e., R9 is
H), then the other
of RAland RA2 is C1-C3 alkyl.
In another aspect, pharmaceutical compositions are featured that include the
above-described
compounds (or salts thereof as described herein) and a pharmaceutically
acceptable carrier. In embodiments,
1, 2, 3, 4, 5, or 6 of the above described provisions can apply.
In one aspect, compounds having formula (I) are featured.
R4
R'
R3 /
0
/C2---R
i
R2 N
\
R1 \
L2--,z
(I)
26
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
C1-C6 haloalkyl, C2-C6
alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
R and R' are defined according to (1) or (2) below:
(1) R and R' together with C2 and C3, respectively, form a fused phenyl
ring having formula (II):
R6
R6....._... R7
1
..--µ....=====C3 .....: .7=.............
..........
C2 R8
I
urtrtrtf
1 (H)
wherein each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; OR
(2) R and R' together with C2 and C3, respectively, form a fused R and R'
together with C2 and
C3, respectively, form a fused heteroaryl ring containing 6 ring atoms,
wherein from 1-2 independently
selected ring atoms is N; and wherein said heteroaryl ring is optionally
substituted with from 1-2
independently selected Rb;
each of Ll and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is:
(i) CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen, halo, C1-
C3 alkyl, and OR9, wherein R9 is C1-C3 alkyl that is optionally substituted
with hydroxyl or C1-C3
alkoxy; or
(ii) C=0; or
(iii) C3-05 cycloalkylene that is (a) substituted with 1 oxo; and (b)
optionally further
substituted with from 1-4 independently selected le; or
(iv) heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of
the ring atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heterocycloalkylene is
(a) substituted with 1 oxo; and (b) is optionally further substituted with
from 1-4 independently
selected Ra;
27
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2 or
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected Ra;
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
(viii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(ix) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(x) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(xi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
28
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (1) below:
(a) hydrogen;
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl);
(0 C2-C6 alkenyl or C2-C6 alkynyl;
(g) C8-C arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(h) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(i) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(j) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
29
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(k) C3-C8 cycloalkyl or C3-C8 cycloalkenyl, each of which is optionally
substituted with from
1-4 independently selected Ra; and
(1) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
provided that one of le and Ril must be selected from (b), (c), (g), (h), (i),
(j), and (k);
R.12 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb; or
(iii) C1-C6 alkyl or C1-C6 haloalkyl, each of which is substituted with from 1-
3 Rd;
(iv) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(v) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vii) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
R" is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(iii) C8-C14 arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(iv) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(v) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
31
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
or
(vi) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
Ra at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), Ci-
C6 alkyl, Ci-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; -
0-(CH2)1-3-
[0(CH2)1-3]1_3-1-1; -C1-C6 alkyl, C1-C6 haloalkyl,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl,
and Ci-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, C1-C6 alkoxy, C1-
C6 thioalkoxy, Ci-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
32
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, CI-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; C1-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-, -
C(0)-, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-;
or a pharmaceutically acceptable salt thereof
In embodiments, 1, 2, 3, 4, or 5 of the following can apply
= provided that R3 and R6 cannot both be hydrogen when A is CH2, and R and
R' are defined
according to definition (1);
= provided that R3 cannot be hydrogen when A is CH2, and R and R' are
defined according to
definition (2);
= provided that R3 and R6 cannot both be chloro when A is CH2, R and R' are
defined according
to definition (1), Z is ¨0R12, and R12 is unsubstituted phenyl;
= provided that R3 and R6 cannot both be bromo when A is CH2, R and R' are
defined according
to definition (1), Z is ¨0R12, and R12 is phenyl that is substituted with
pyridyl or alkyl that is
substituted with from 1-3 Re; and
= provided that R3 and R6 cannot both be hydrogen when A is CH(CH3), R and
R' are defined
according to definition (1), Z is NRI0R11, RI is CH3, and R11 is
unsubstituted phenyl.
In another aspect, pharmaceutical compositions are featured that include the
above-described
compounds (or salts thereof as described herein) and a pharmaceutically
acceptable carrier. In embodiments,
1, 2, 3, 4, or 5 of the above described provisions can apply.
In another aspect, compounds having formula (I) are featured
R4
R'
R3 I
R20 C3
/ 2
\Ll¨A
W \
L2---,z
(I)
33
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
C1-C6 haloalkyl, C2-C6
alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
R and R' are defined according to (1) or (2) below:
(1) R and R' together with C2 and C3, respectively, form a fused phenyl
ring having formula (II):
R6
R6....._... R7
1
..--µ....=====C3 .....: .7=.............
..........
C2 R8
I
urtrtrtf
1 (H)
wherein each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; OR
(2) R and R' together with C2 and C3, respectively, form a fused R and R'
together with C2 and
C3, respectively, form a fused heteroaryl ring containing 6 ring atoms,
wherein from 1-2 independently
selected ring atoms is N; and wherein said heteroaryl ring is optionally
substituted with from 1-2
independently selected Rb;
each of Ll and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is CRA1RA2, wherein one of RA1 and RA2 is -OH, and the other of RA1 and RA2
is hydrogen or Ci-C3
alkyl;
Z is -0R12 or -S(0),A13, wherein n is 0, 1, or 2;
each of R12 and R13 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(iii) C1-C6 alkyl or C1-C6 haloalkyl (e.g., C1-C6 alkyl), each of which is
substituted with from
1-3 Rd; or
(iv) C8-C14 arylcycloalkyl, wherein:
34
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
or
(v) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vi) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
or
(vii) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
Ra at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), Ci-
C6 alkyl, Ci-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6haloalkoxy; C1-C6thioalkoxy; C1-C6thiohaloalkoxy; -0-
(CH2)1-3-
[0(CH2)1-3]1_3-H; -C1-C6 alkyl, C1-C6 haloalkyl,
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl,
and Ci-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, C1-C6 alkoxy, C1-
C6 thioalkoxy, Ci-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, CI-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, Ci-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; C1-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-, -
C(0)-, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-;
or a pharmaceutically acceptable salt thereof
In embodiments, 1, 2, 3, or 4 of the following can apply:
= provided that R3 and R6 cannot both be hydrogen when R and R' are defined
according to
definition (1);
= provided that R3 and R6 cannot both be chloro when R and R' are defined
according to
definition (1), Z is ¨0R12, and R12 is phenyl substituted with chloro, formyl,
or -NHC(0)CH3;
= provided that R3 and R6 cannot both be bromo when R and R' are defined
according to
definition (1), Z is ¨0R12, and R12 is phenyl substituted with -NHC(0)CH3; and
36
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
= provided that R3 and R6 cannot both be bromo when R and R' are defined
according to
definition (1), Z is ¨SR13, and R13 is phenyl substituted with ¨OH.
In another aspect, pharmaceutical compositions are featured that include the
above-described
compounds (or salts thereof as described herein) and a pharmaceutically
acceptable carrier. In embodiments,
1, 2, 3, 4, or 5 of the above described provisions can apply.
In another aspect, compounds having formula (I) are featured:
R4
R'
R3 /
R20 C3
2
\Ll¨A
R1 \
L2--........z
(I)
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
Ci-C6 haloalkyl, C2-C6
alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
R and R' together with C2 and C3, respectively, form a fused heterocyclic ring
containing from 5-6
ring atoms, wherein from 1-2 of the ring atoms is independently selected from
N, NH, N(C1-C6 alkyl),
NC(0)(C1-C6 alkyl), 0, and S; and wherein said heterocyclic ring is optionally
substituted with from 1-3
independently selected Ra;
each of Ll and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is:
(i) CRA1RA2, wherein one of RA1 and RA2 is independently selected from
hydrogen, halo, Ci-C3
alkyl, and OR9; and the other of RA1 and RA2 is independently selected from
halo, C1-C3 alkyl, and
OR9; wherein R9 is hydrogen or C1-C3 alkyl that is optionally substituted with
hydroxyl or C1-C3
alkoxy; or
(ii) C=0;
Z is:
(i) -NR10R11; or
37
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2 or
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (g) below:
(a) hydrogen;
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6haloalkyl), or -C(0)0(C1-C6 alkyl);
(f) C2-C6 alkenyl or C2-C6 alkynyl;
and
(g) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
provided that one of le and Ril must be selected from (b) and (c);
R.12 is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
R" is:
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
le at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6thioalkoxy,
C1-C6haloalkoxy, C1-C6thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), C1-C6
alkyl, C1-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
38
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6 haloalkoxy; Ci-C6 thioalkoxy; Ci-C6 thiohaloalkoxy; -
0-(CH2)1-3-
[0(CH2)1-3]1_3-H; -Ci-C6 alkyl, Ci-C6 haloalkyl,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl,
and Ci-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, Ci-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, C1-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, ¨
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; C1-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-, -
C(0)-, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-;
or a pharmaceutically acceptable salt thereof
In embodiments, provision (A) described herein can apply.
In another aspect, compounds having formula (I) are featured:
39
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
R4
R'
R3 I
R2 0 C3
/ 2
\
W \
L2---,z
(I)
wherein:
each of le, R2, R3, and R4 is independently selected from hydrogen, halo,
hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl,
C1-C6 haloalkyl, C2-C6
-- alkynyl, cyclopropyl, -N3, cyano,
-NH2, -NH(Ci-C 6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro;
each of R and R' is, independently, hydrogen, C1-C6 alkyl, or C1-C6 haloalkyl;
each of Ll and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is:
(i) CRA1RA2, wherein one of RA1 and RA2 is independently selected from
hydrogen, fluoro,
chloro, C1-C3 alkyl, and OR9; and the other of RA1 and RA2 is independently
selected from fluoro,
chloro, C1-C3 alkyl, and OR9; wherein R9 is hydrogen or C1-C3 alkyl that is
optionally substituted with
hydroxyl or C1-C3 alkoxy; or
(ii) C=0;
Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0),A13, wherein n is 0, 1, or 2 or
(vi) C6-C10 aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb; or
each of Rl and Ril is independently selected from the substituents delineated
collectively in (a)
through (g) below:
(a) hydrogen;
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6haloalkyl), or -C(0)0(C1-C6 alkyl);
(f) C2-C6 alkenyl or C2-C6 alkynyl;
and
(g) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
provided that one of R'b and Rn must be selected from (b) and (c);
each of R" and R" is::
(i) C6-C10 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
le at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6thioalkoxy,
C1-C6haloalkoxy, C1-C6thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), C1-C6
alkyl, C1-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano;
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6haloalkoxy; Ci-C6thioalkoxy; Ci-C6thiohaloalkoxy; -0-
(CH2)1-3-
[0(CH2)1-3]1_3-H; -Ci-C6 alkyl, Ci-C6haloalkyl,
-NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), wherein the alkyl
portion of each is
optionally substituted with from 1-3 independently selected Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6haloalkyl); C(0)0H;
-C(0)0(C1-C6 alkyl); -C(0)NH2; -C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -
S02(C1-C6 alkyl); -
SO2NH2; -SO2NH(C1-C6 alkyl); -SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
41
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), Ci-C6
alkoxy; C1-C6 haloalkoxy; Ci-C6 thioalkoxy; Ci-C6 thiohaloalkoxy; Ci-C6 alkyl,
and C1-C6 haloalkyl;
Ir at each occurrence is, independently selected from halo, Ci-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, C1-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; CI-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); N(C1-C6 alky1)2; -
NHC(0)(C1-C6 alkyl); cyano;
-C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-C6
alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-, -
C(0)-, -C(0)NH-, -C(0)NCH3-, -
NHC(0)-, or -NCH3C(0)-;
or a pharmaceutically acceptable salt thereof
In one aspect, compounds of formula (III) are featured in which:
A is CRAIRA2, in which each of RAI and RA2 is, independently, hydrogen, halo,
or C1-C3 alkyl; or
A is CRAIRA2, in which one of RAI and RA2 is halo (e.g., fluoro), and the
other of RAI and RA2 is,
independently, hydrogen, halo, or Ci-C3 alkyl (e.g., hydrogen); or
A is CRAIRA2, in which one of RAI and RA2 is halo (e.g., fluoro), and the
other of RAI and RA2 is
hydrogen; and
RI, R2, R3, R4, LI, L2, and Z can be as defined anywhere herein; or a salt
(e.g., pharmaceutically
acceptable salt) thereof
In embodiments, (B) and/or (C) applies.
In one aspect, compounds of formula (III) are featured in which:
one of RAI and RA2 can be OR9. In embodiments, the other of RAI and RA2 can be
as defined anywhere
herein; e.g., the other of RAI and RA2 can be hydrogen or C1-C3 alkyl. For
example, one of RAI and RA2 can be
OR9, and the other of RAI and RA2 is hydrogen or C1-C3 alkyl. In embodiments,
R9 can be hydrogen or C1-C3
alkyl; and
42
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
RI, R2, R3, R4, LI, L2, and Z can be as defined anywhere herein; or a salt
(e.g., pharmaceutically
acceptable salt) thereof
In embodiments, one or more of the following apply, e.g., when A is CHOH and Z
is NRI0R11:
= each of R3 and R6 is CH3; and/or each of R3 and R6 is bromo; and/or each
of R3 and R6 is
chloro; and/or one of R3 and R6 is CH3 (e.g., R6), and the other is bromo
(e.g., R3);
= each of RI and RII is other than hydrogen;
= each of RI and RII is hydrogen;
= one of RI and RII is heteroaryl as defined anywhere herein;
= LI and/or L2 is C2-C3 alkylene (optionally substituted);
= (B) and/or (C) applies.
In one aspect, compounds of formula (III) are featured in which Z is other
than NRI0R11; and RI, R2,
R3, R4, LI, L2, Z, and A can be as defined anywhere herein; or a salt (e.g.,
pharmaceutically acceptable salt)
thereof In embodiments, (B) and/or (C) applies.
In one aspect, compounds of formula (III) are featured in which Z is -0R12
and/or ¨S(0)õR13; and RI,
R2, R3, R4, LI, L2, and A can be as defined anywhere herein; or a salt (e.g.,
pharmaceutically acceptable salt)
thereof In embodiments, (B) and/or (C) applies.
In one aspect, compounds of formula (III) are featured in which A is (ii) C=0;
and/or (iv)
heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of the
ring atoms is independently
selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heterocycloalkylene is (a) substituted with 1
oxo; and (b) is optionally further substituted with from 1-4 independently
selected le; and RI, R2, R3, R4, LI,
L2, and Z can be as defined anywhere herein; or a salt (e.g., pharmaceutically
acceptable salt) thereof
In yet another aspect, compounds of formula (VI) are feutured:
R4
R3 R5
*
X
R2 Ni
1
R1 L1,,A
I
L2
Z
(VI)
wherein:
43
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
R1 ¨ R5 are each independently selected from hydrogen, halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, Ci-C6 alkyl, Ci-C6
haloalkyl, C2-C6 alkynyl,
cyclopropyl, -N3, cyano, -NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-
C6 alkyl), and nitro;
X is C6-C10 aryl that is optionally substituted with 1-4 Rb; or heteroaryl
containing 5-14 ring atoms,
wherein from 1-6 of the ring atoms is independently selected from N, NH, N(C1-
C3 alkyl), 0, and S, and
wherein said heteroaryl is optionally substituted with 1-4 Rb;
each of Ll and L2 is, independently, Ci-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re;
A is CRA1RA2, wherein one of RA1 and RA2 is independently selected from
hydrogen, fluoro, chloro,
C1-C3 alkyl, and OR9; and the other of RA1 and RA2 is independently selected
from fluoro, chloro, C1-C3 alkyl,
and OR9; wherein R9 is hydrogen or C1-C3 alkyl that is optionally substituted
with hydroxyl or C1-C3 alkoxy;
Z is -NR10R11 or -0R12;
each of le and Ril is independently selected from the substituents delineated
collectively in (a)
through (g) below:
(a) hydrogen;
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl);
(0 C2-C6 alkenyl or C2-C6 alkynyl;
and
(g) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
provided that one of R" and Rn must be selected from (b) and (c);
R12 is::
(i) C6-Ci0 aryl that is optionally substituted with from 1-4 Rb; or
(ii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
le at each occurrence is, independently selected from halo, hydroxyl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, oxo, thioxo, =NH, =N(C1-C6 alkyl), C1-
C6 alkyl, C1-C6 haloalkyl, -
NH2, -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and cyano;
44
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Rb at each occurrence is independently selected from the substituents
delineated in (aa) through (dd)
below:
(aa) C1-C6 alkoxy; C1-C6 haloalkoxy; Ci-C6 thioalkoxy; Ci-C6 thiohaloalkoxy; -
0(CH2)1-
3[0(CH2)1_3]1_3H; Ci-C6 alkyl, Ci-C6 haloalkyl, -NH(C1-C6 alkyl), -N(C1-C6
alky1)2, -NHC(0)(C1-C6
alkyl), wherein the alkyl portion of each is optionally substituted with from
1-3 independently selected
Re;
(bb) halo; hydroxyl; cyano; nitro; -NH2; azido; sulfhydryl; C2-C6 alkenyl; C2-
C6 alkynyl; -
C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); -C(0)0H; -C(0)0(C1-C6
alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -
SO2N(C1-C6 alky1)2;
(cc) C3-C6 cycloalkyl or heterocyclyl containing from 5-6 ring atoms, wherein
from 1-2 of the
ring atoms of the heterocyclyl is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein each of said phenyl and heterocyclyl is
optionally substituted with from
1-3 independently selected Ra; and
(dd) phenyl or heteroaryl containing from 5-6 ring atoms, wherein from 1-2 of
the ring atoms
of the heteroaryl is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; wherein each of said
phenyl and heteroaryl is optionally substituted with from 1-3 substituents
independently selected from
halo; hydroxyl; cyano; nitro; -NH2; -NH(C1-C6 alkyl), -N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), C1-C6
alkoxy; C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl,
and Ci-C6 haloalkyl;
Re at each occurrence is, independently selected from halo, Ci-C6 alkoxy, C1-
C6 thioalkoxy, C1-C6
haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -NH(C1-
C6 alkyl), -N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and cyano;
Rd at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy,
C1-C6 thioalkoxy, C1-
C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, -NH2, -
NH(C1-C6 alkyl), -N(C1-C6 alky1)2,
-NHC(0)(C1-C6 alkyl), and cyano; and
Re at each occurrence is, independently selected from hydroxyl, C1-C6 alkoxy;
C1-C6 thioalkoxy; C1-
C6 haloalkoxy; C1-C6 thiohaloalkoxy; -NH2; -NH(C1-C6 alkyl); -N(C1-C6 alky1)2;
-NHC(0)(C1-C6 alkyl);
cyano; -C(0)H; -C(0)(C1-C6 alkyl); -C(0)(C1-C6 haloalkyl); -C(0)0H; -C(0)0(C1-
C6 alkyl); -C(0)NH2; -
C(0)NH(C1-C6 alkyl); -C(0)N(C1-C6 alky1)2; -S02(C1-C6 alkyl); -SO2NH2; -
SO2NH(C1-C6 alkyl); -SO2N(C1-
C6 alky1)2; and L3-(C1-C6 alkylene)-biotin, where in L3 is a -0-, -NH-, -NCH3-
, -C(0)-, -C(0)NH-, -
C(0)NCH3-, -NHC(0)-, or -NCH3C(0)-;
or a pharmaceutically acceptable salt thereof
In certain embodiments, compound of formula (VI) can have a R3 that is
selected from halo, hydroxyl,
sulfhydryl, C1-C6 alkoxy, C1-C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl, C1-C6
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano, -NH2, -NH(C1-C6 alkyl), -
N(C1-C6 alky1)2, -NHC(0)(C1-C6
alkyl), and nitro. In some embodiments, R3 is halo such as bromo. In certain
embodiments, each of RI, R2, R4
and R5 is hydrogen.
In certain embodiments, compound of formula (VI) can have X that is C6-Cio
aryl substituted with one
or more halo such as bromo. For example, X can be 4-bromophenyl. X can also be
heteroaryl containing 5-14
ring atoms, wherein from 1-6 of the ring atoms is independently selected from
N, NH, N(C1-C3 alkyl), 0, and
S, and wherein said heteroaryl is optionally substituted with 1-4 Rb. For
example, X can be pyridine optionally
substituted with 1-4 Rb.
In certain embodiments, compound of formula (VI) can have A that is CRA1RA2,
wherein each of RA1
and RA2 is, independently, hydrogen, C1-C3 alkyl, or OR9. In some embodiments,
one of RA1 and RA2 is OR9;
and the other of RA1 and RA2 is hydrogen or C1-C3 alkyl. For example, one of
RA1 and RA2 can be OH; and the
other of RA1 and RA2 can be hydrogen.
In some embodiments, A is CRA1RA2 and wherein the carbon attached to RA1 and
RA2 is substituted
with four different substituents. The carbon attached to to RA1 and RA2 can be
(R) or (S) configured. In an
embodiment, the (R) configured formula (VI) compound can be substantially free
of a formula (VI) compound
that is S configured at the carbon atom attached to to RA1 and R. In some
embodiments, the (S) configured
formula (VI) compound can be substantially free of a formula (VI) compound
that is (R) configured at the
carbon atom attached to to RA1 and R.
The compound of formula (VI), in some embodiments, can be (+) or (-)
(dextrorotatory).
In some embodiments, the (+) (dextrorotatory) compound can be substantially
free of a formula (I) compound
that is (levororotatory). In some embodiments, the (-) (levororotatory)
compound can be substantially free of
a formula (I) compound that is (+) (dextrorotatory).
Any of the aforementioned compounds can be used in any of the methods or
compositions described
anywhere herein.
The presently disclosed embodiments relate generally to stimulating
neurogenesis (e.g., post-natal
neurogenesis, e.g., post-natal hippocampal and/or hypothalamic neurogenesis)
and protecting neurons from
death with a compound of formula (I) (and/or a compound of any of the other
formulae described herein) or a
salt (e.g., a pharmaceutically acceptable salt) thereof as defined anywhere
herein.
For example, methods of promoting the generation of neurons are featured. As
another example,
methods of promoting the survival, growth, development and/or function of
neurons, particularly CNS, brain,
cerebral, hippocampal and hypothalamic neurons are featured. As a further
example, methods of stimulating
post-natal hippocampal and/or hypothalamic neurogenesis are featured.
In some embodiments, such methods can include in vitro methods, e.g.,
contacting a sample (e.g., a
cell or tissue) with a compound of formula (I) (and/or a compound of any of
the other formulae described
46
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
herein) or a salt (e.g., a pharmaceutically acceptable salt) thereof as
defined anywhere herein. In other
embodiments, the methods can include administering a compound of formula (I)
(and/or a compound of any of
the other formulae described herein) or a salt (e.g., a pharmaceutically
acceptable salt) thereof as defined
anywhere herein to a subject (e.g., a mammal, such as a human).
Accordingly, in yet another aspect, the presently disclosed embodiments
include and feature methods
of screening for (thereby identifying) compounds that stimulate neurogenesis
(e.g., post-natal neurogenesis,
e.g., post-natal hippocampal and/or hypothalamic neurogenesis) or protect
newborn neurons from cell death.
E.g., such as those described in the Examples section.
In one aspect, methods for treating (e.g., controlling, relieving,
ameliorating, alleviating, or slowing
the progression of) or methods for preventing (e.g., delaying the onset of or
reducing the risk of developing)
one or more diseases, disorders, or conditions caused by, or associated with
insufficient (e.g., aberrant)
neurogenesis or unwanted neuronal cell death in a subject in need thereof are
featured. The methods include
administering to the subject an effective amount of a compound of formula (I)
(and/or a compound of any of
the other formulae described herein) or a salt (e.g., a pharmaceutically
acceptable salt) thereof as defined
anywhere herein to the subject.
In another aspect, the use of a compound of formula (I) (and/or a compound of
any of the other
formulae described herein) or a salt (e.g., a pharmaceutically acceptable
salt) thereof as defined anywhere
herein in the preparation of, or for use as, a medicament for the treatment
(e.g., controlling, relieving,
ameliorating, alleviating, or slowing the progression of) or prevention (e.g.,
delaying the onset of or reducing
the risk of developing) of one or more diseases, disorders, or conditions
caused by, or associated with,
insufficient (e.g., aberrant) neurogenesis or unwanted neuronal cell death is
featured.
In embodiments, the one or more diseases, disorders, or conditions can include
neuropathies, nerve
trauma, and neurodegenerative diseases. In embodiments, the one or more
diseases, disorders, or conditions
can be diseases, disorders, or conditions caused by, or associated with
insufficient neurogenesis (e.g., aberrant
hippocampal and/or hypothalamic neurogenesis) as is believed to occur in
neuropsychiatric diseases, or
aberrant neuronal cell death as is believed to occur in neurodegenerative
diseases. Examples of the one or
more diseases, disorders, or conditions include, but are not limited to,
schizophrenia, major depression,
bipolar disorder, normal aging, epilepsy, traumatic brain injury, post-
traumatic stress disorder, Parkinson's
disease, Alzheimer's disease, Down syndrome, spinocerebellar ataxia,
amyotrophic lateral sclerosis,
Huntington's disease, stroke, radiation therapy, chronic stress, and abuse of
neuro-active drugs (such as
alcohol, opiates, methamphetamine, phencyclidine, and cocaine), retinal
degeneration, spinal cord injury,
peripheral nerve injury, physiological weight loss associated with various
conditions, and cognitive decline
associated with normal aging, radiation therapy, and chemotherapy.
In some embodiments, the subject can be a subject in need thereof (e.g., a
subject identified as being in
need of such treatment, such as a subject having, or at risk of having, one or
more of the diseases or conditions
47
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
described herein). Identifying a subject in need of such treatment can be in
the judgment of a subject or a
health care professional and can be subjective (e.g. opinion) or objective
(e.g. measurable by a test or
diagnostic method). In some embodiments, the subject can be a mammal. In
certain embodiments, the subject
can be a human.
In another aspect, methods of making the compounds described herein are
featured. In embodiments,
the methods include taking any one of the intermediate compounds described
herein and reacting it with one or
more chemical reagents in one or more steps to produce a compound of formula
(I) (and/or a compound of any
of the other formulae described herein) or a salt (e.g., a pharmaceutically
acceptable salt) thereof as defined
anywhere herein.
In some embodiments, compounds in which A is CHOH, and each of LI and L2 is C1-
C3 alkylene (e.g.,
each of LI and L2 is CH2) can be converted to compounds in which A is C(0),
and each of LI and L2 is C1-C3
alkylene (e.g., each of LI and L2 is CH2) that is substituted with C1-C6
thioalkoxy (e.g., -SCH3). The methods
include contacting the starting material with an oxidizing agent sulfur
trioxide pyridine complex (see, e.g.,
Example 7a and 7b).
In one aspect, methods of making the pharmaceutical compositions described
herein are featured. In
embodiments, the methods include taking any one or more of the compounds of
formula (I) (and/or
compounds of any of the other formulae described herein) or a salt (e.g., a
pharmaceutically acceptable salt)
thereof as defined anywhere herein, and mixing said compound(s) with one or
more pharmaceutically
acceptable carriers.
In one aspect, kits for the treatment (e.g., controlling, relieving,
ameliorating, alleviating, or slowing
the progression of) or prevention (e.g., delaying the onset of or reducing the
risk of developing) of one or more
diseases, disorders, or conditions caused by, or associated with insufficient
(e.g., aberrant) neurogenesis or
unwanted neuronal cell death are featured. The kits include (i) a compound of
formula (I) (and/or compounds
of any of the other formulae described herein) or a salt (e.g., a
pharmaceutically acceptable salt) thereof as
defined anywhere herein; and (ii) instructions that include a direction to
administer said compound to a subject
(e.g., a patient).
Embodiments can include, for example, any one or more of the following
features.
R3 is selected from halo, hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6
thioalkoxy, Ci-C6 haloalkoxy, C1-
C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 alkynyl, cyclopropyl, -
N3, cyano, -NH2, -NH(C1-C6
alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro. In embodiments, R3
is halo (e.g., bromo). In
embodiments, each of RI, R2, and R4 is hydrogen.
R and R' together with C2 and C3, respectively, form a fused phenyl ring
having formula (II):
48
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
R6
R6---...-- R7
1
)2a.....--C3......... ..."Nõ...........
C2 R8
I
I (H).
R6 is selected from halo, hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy, Cl-
C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 alkynyl, cyclopropyl, -
N3, cyano, -NH2, -NH(C1-C6
alkyl), -N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro. In embodiments, R6
is halo (e.g., bromo) or C1-C6
alkyl (e.g., CH3). In embodiments, R6 is halo (e.g., bromo). In embodiments,
each of R5, R7, and R8 is
hydrogen.
In embodiments, each of R3 and R6 is an independently selected substituent
that is other than
hydrogen. In certain embodiments, each of R3 and R6 is independently selected
from halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, C2-C6 alkynyl, cyclopropyl, -N3, cyano, -NH2, -NH(C1-C6 alkyl), -
N(C1-C6 alky1)2, -NHC(0)(C1-C6
alkyl), and nitro. For example, R3 can be halo (e.g., bromo); and R6 can be
halo (e.g., bromo) or C1-C6 alkyl
(e.g., CH3); e.g., halo (e.g., bromo). In embodiments, each of RI, R2, and R4
is hydrogen; and each of R5, R7,
and R8 is hydrogen.
In embodiments, R and R' together with C2 and C3, respectively, form a fused
heteroaryl ring
containing from 5-6 ring atoms, wherein from 1-2 of the ring atoms is
independently selected from N, NH,
N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl ring is optionally
substituted with from 1-3
independently selected Rb.
For example, R and R' together with C2 and C3, respectively, form a fused
heteroaryl ring containing -
6 ring atoms, wherein from 1-2 independently selected ring atoms is N; and
wherein said heteroaryl ring is
optionally substituted with from 1-2 independently selected Rb.
In embodiments, R and R' together with C2 and C3, respectively, form a fused
heterocyclic ring
containing from 5-6 ring atoms, wherein from 1-2 of the ring atoms is
independently selected from N, NH,
N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said heterocyclic
ring is optionally substituted
with from 1-3 independently selected R.
For example, R and R' together with C2 and C3, respectively, form a fused
heterocyclic ring containing
6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C6 alkyl), and
NC(0)(C1-C6 alkyl); and wherein said heterocyclic ring is optionally
substituted with from 1-3 independently
selected R.
49
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In embodiments, R and R' is, independently, hydrogen, C1-C6 alkyl, or C1-C6
haloalkyl (e.g., C1-C6
alkyl, or C1-C6 haloalkyl; e.g., C1-C6 alkyl).
Each of L1 and L2 is, independently, C1-C3 straight chain alkylene, which is
optionally substituted with
from 1-2 independently selected Re. For example, each of L1 and L2 is CH2.
A is CRA1RA2, in which each of RA1 and RA2 is, independently, hydrogen, halo,
C1-C3 alkyl, or OR9.
In some embodiments, A is other than CH2.
In embodiments, one of RA1 and RA2 can be independently selected from
hydrogen, halo, C1-C3 alkyl,
and OR9; and the other of RA1 and RA2 can be independently selected from halo,
Ci-C3 alkyl, and OR9. For
example, one of RA1 and RA2 is halo, C1-C3 alkyl, or OR9 (e.g., halo or OR9);
and the other is hydrogen or Cl -
C3 alkyl.
In embodiments, one of RA1 and RA2 is halo, and the other of RA1 and RA2 is
hydrogen or halo. For
example, one of RA1 and RA2 is fluoro, and the other of RA1 and RA2 is
hydrogen or fluoro. In either
embodiments, one of RA1 and RA2 is OR9; and the other of RA1 and RA2 is C1-C3
alkyl. For example, one of RA1
and RA2 is OH; and the other of RA1 and RA2 is CH3.
In embodiments, the carbon attached to RA1 and RA2 is substituted with four
different substituents (for
purposes of clarification, these four substituents include RA1 and RA2) and is
therefore a stereogenic center.
In certain embodiments, the carbon attached to RA1 and RA2 is (R) configured,
meaning that the carbon
attached to RA1 and RA2 has the (R) configuration (Cahn Ingold Prelog sequence
rules notation). Such
compounds are sometimes referred to herein as an "(R)-configured compound"
(this term also includes
compounds that further contain one or more stereogenic centers in addition to
the (R)-CRA1RA2 stereogenic
center).
In other embodiments, the carbon attached to RA1 and RA2 is (S) configured,
meaning that the carbon
attached to RA1 and RA2 has the (S) configuration (Cahn Ingold Prelog sequence
rules notation). Such
compounds are sometimes referred to herein as an "(S)-configured compound"
(this term also includes
compounds that further contain one or more stereogenic centers in addition to
the (5)-CRA1RA2 stereogenic
center).
In embodiments, the (R) configured compound (or salt, e.g., a pharmaceutically
acceptable salt,
thereof) is substantially free of (e.g., contains less than about 5% of, less
than about 2% of, less than about 1%,
less than about 0.5% of) a formula (I) compound (or salt thereof as described
herein) that is (S) configured at
the carbon attached to RA1 and RA2 (i.e., a formula (I) compound in which the
carbon attached to RA1 and RA2
has the (S) configuration). For example, the (R) configured compound can be an
(R)-enantiomer that is
substantially free of its opposing (S) enantiomer. As another example, an (R)
configured compound can be
substantially free of a diastereomer in which the carbon attached to RA1 and
RA2 has the (S) configuration. In
certain embodiments, the (R) configured compound can be additionally in
substantially pure form (e.g.,
contains less than about 5% of, less than about 2% of, less than about 1%,
less than about 0.5% of other
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
substances, including, for example, one or more of other formula (I)
compounds, non-formula (I) compounds,
or biological media).
In embodiments, the (S) configured compound (or salt, e.g., a pharmaceutically
acceptable salt,
thereof) is substantially free of (e.g., contains less than about 5% of, less
than about 2% of, less than about 1%,
less than about 0.5% of) a formula (I) compound (or salt thereof as described
herein) that is (R) configured at
the carbon attached to RA1 and RA2 (i.e., a formula (I) compound in which the
carbon attached to RA1 and RA2
has the (R) configuration). For example, the (S) configured compound can be an
(S)-enantiomer that is
substantially free of its opposing (R) enantiomer. As another example, the (S)
configured compound can be
substantially free of a diastereomer in which the carbon attached to RA1 and
RA2 has the (R) configuration. In
certain embodiments, the (S) configured compound can be additionally in
substantially pure form (e.g.,
contains less than about 5% of, less than about 2% of, less than about 1%,
less than about 0.5% of other
substances, including, for example, one or more of other formula (I)
compounds, non-formula (I) compounds,
or biological media).
In certain embodiments, a formula (I) compound is (+)(dextrorotatory) when in
the presence of plane
polarized light.
In certain embodiments, a formula (I) compound is (-) (levororotatory) when in
the presence of plane
polarized light.
In embodiments, the (+)(dextrorotatory) compound is substantially free of
(e.g., contains less than
about 5% of, less than about 2% of, less than about 1%, less than about 0.5%)
a formula (I) compound (or salt
thereof as described herein) that is (-) (levororotatory). In certain
embodiments, the (+)(dextrorotatory)
compound can be additionally in substantially pure form (e.g., contains less
than about 5% of, less than about
2% of, less than about 1%, less than about 0.5% of other substances,
including, for example, one or more of
other formula (I) compounds, non-formula (I) compounds, or biological media).
In embodiments, the (-) (levororotatory) compound is substantially free of
(e.g., contains less than
about 5% of, less than about 2% of, less than about 1%, less than about 0.5%)
a formula (I) compound (or salt
thereof as described herein) that is (+)(dextrorotatory). In certain
embodiments, the (-) (levororotatory)
compound can be additionally in substantially pure form (e.g., contains less
than about 5% of, less than about
2% of, less than about 1%, less than about 0.5% of other substances,
including, for example, one or more of
other formula (I) compounds, non-formula (I) compounds, or biological media).
A is: (i) CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen, halo, Ci-C3
alkyl, and OR9, wherein R9 is C1-C3 alkyl that is optionally substituted with
hydroxyl or Ci-C3 alkoxy; or (ii)
C=0.
A is CRA1RA2, wherein each of RA1 and RA2 is, independently, hydrogen, halo,
C1-C3 alkyl, or OR9.
In embodiments, one of RA1 and RA2 is independently selected from hydrogen,
halo, Ci-C3 alkyl, and
OR9; and the other of RA1 and RA2 is independently selected from halo, C1-C3
alkyl, and OR9.
51
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, one of RAI and RA2 is halo, and the other of RAI and
RA2 is hydrogen, halo, or
C1-C3 alkyl. In embodiments, one of RAI and RA2 is halo, and the other of RAI
and RA2 is hydrogen. For
example, one of RAI and RA2 is fluoro, and the other of RAI and RA2 is
hydrogen.
In other embodiments, each of RAI and RA2 is, independently, halo; e.g., each
of RAI and RA2 is fluoro.
In embodiments, one of RAI and RA2 is -OH, and the other of RAI and RA2 is
hydrogen.
In embodiments, A is CRAIRA2, wherein one of RAI and RA2 is independently
selected from hydrogen,
halo, C1-C3 alkyl, and OR9; and the other of RAI and RA2 is independently
selected from halo, Ci-C3 alkyl, and
OR9; wherein R9 is hydrogen or Ci-C3 alkyl that is optionally substituted with
hydroxyl or C1-C3 alkoxy.
In certain embodiments, one of RAI and RA2 is OR9, and the other is hydrogen,
wherein R9 is
hydrogen.
In embodiments, one of RAI and RA2 is halo, and the other of RAI and RA2 is
hydrogen or halo. For
example, one of RAI and RA2 is fluoro, and the other of RAI and RA2 is
hydrogen or fluoro.
In other embodiments, one of RAI and RA2 is OR9; and the other of RAI and RA2
is C1-C3 alkyl. For
example, one of RAI and RA2 is OH; and the other of RAI and RA2 is CH3.
Z is: (i) -NR10R11; or (ii) -C(0)NRI0R11; or (iii) -0R12; or (iv) -S(0),A13,
wherein n is 0, 1, or 2.
Z is -NRI0R11. In embodiments, one of RI and RII is: (b) C6-C10 aryl that is
optionally substituted
with from 1-4 Rb; or (c) heteroaryl containing from 5-14 ring atoms, wherein
from 1-6 of the ring atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is optionally
substituted with from 1-4 Rb; and the other of RI and RII is hydrogen or C1-
C6 alkyl.
Z is -0R12 or ¨S(0),A13.
In embodiments, Z is -0R12. In certain embodiments, RI2 is C6-Cio aryl that is
optionally substituted
with from 1-4 Rb.
In embodiments, RI2 is C1-C6 alkyl or C1-C6 haloalkyl (e.g., Ci-C6 alkyl),
each of which is substituted
with from 1-3 Rd. In other embodiments, RI2 is other than Ci-C6 alkyl or Ci-C6
haloalkyl (e.g., Ci-C6 alkyl),
each of which is unsubstituted or substituted with from 1-3 Rd.
R3 can be selected from halo, hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy,
C1-C6 thiohaloalkoxy, C1-C6 alkyl, Ci-C6 haloalkyl, cyano, -NH2, -NH(C1-C6
alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and nitro. E.g., R3 can be halo (e.g., bromo). In
embodiments, each of RI, R2, and R4
can be hydrogen.
L1 can be C1-C3 straight chain alkylene, which is optionally substituted with
from 1-2 independently
selected Re. E.g., LI can be CH2.
L2 can be C1-C3 straight chain alkylene, which is optionally substituted with
from 1-2 independently
selected Re. E.g., L2 can be CH2.
Each of LI and L2 can be, independently, C1-C3 straight chain alkylene, which
is optionally substituted
with from 1-2 independently selected R. E.g., each of LI and L2 can be CH2.
52
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
A can be CRA1RA2, in which each of RA1 and RA2 is, independently, hydrogen,
halo, C1-C3 alkyl, or
OR9.
A can be CRA1RA2, in which each of RA1 and RA2 is, independently, hydrogen,
halo, or C1-C3 alkyl.
A can be CRA1RA2, in which one of RA1 and RA2 is halo (e.g., fluoro), and the
other of RA1 and RA2 is,
independently, hydrogen, halo, or Ci-C3 alkyl (e.g., hydrogen).
A can be CRA1RA2, in which one of RA1 and RA2 is halo (e.g., fluoro), and the
other of RA1 and RA2 is
hydrogen.
One of RA1 and RA2 can be halo or OR9, and the other is hydrogen.
One of RA1 and RA2 can be OR9. In embodiments, the other of RA1 and RA2 can be
as defined
anywhere herein; e.g., the other of RA1 and RA2 can be hydrogen or C1-C3
alkyl. For example, one of RA1 and
RA2 can be OR9, and the other of RA1 and RA2 is hydrogen. In embodiments, R9
can be hydrogen.
One of RA1 and RA2 can be halo. In embodiments, the other of RA1 and RA2 can
be as defined
anywhere herein; e.g., the other of RA1 and RA2 can be hydrogen, C1-C3 alkyl,
or halo. For example, one of
RA1 and RA2 can be halo (e.g., fluoro), and the other of RA1 and RA2 is
hydrogen.
The carbon attached to RA1 and RA2 can have the R configuration.
The carbon attached to RA1 and RA2 can have the S configuration.
Each of L1 and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re. E.g., each of L1 and L2 can be CH2.
Z can be -NR10R11.
One of R1 and R11 can be C6-C10 aryl that is optionally substituted with from
1-4 Rb.
One of R1 and R11 can be C6-Cio aryl that is optionally substituted with from
1-4 Rb, and the other is
hydrogen or C1-C6 alkyl.
One of R1 and R11 can be C6-Cio aryl that is optionally substituted with from
1-4 Rb, and the other is
hydrogen. For example, one of R1 and R11 can be unsubstituted phenyl, and the
other is hydrogen. As
another example, one of R1 and R11 can be phenyl that is substituted with 1
Rb, and the other is hydrogen. In
embodiments, Rb can be Ci-C6 alkoxy (e.g., OCH3). For example, one of R1 and
R11 can be 3-methoxyphenyl,
and the other is hydrogen.
Z can be -0R12. In embodiments, R12 can be Ci-C6 alkyl or Ci-C6 haloalkyl,
each of which is
optionally substituted with from 1-3 R. In other embodiments, R12 can be C6-
Cio aryl that is optionally
substituted with from 1-4 Rb. For example, R12 can be unsubstituted phenyl.
Z can be -S(0)õR13, in which n can be 0, 1, or 2. In other embodiments, R13
can be C6-C10 aryl that is
optionally substituted with from 1-4 Rb. For example, R13 can be unsubstituted
phenyl.
Z can be heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3
of the ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected R.
53
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
R and R' together with C2 and C3, respectively, form a fused phenyl ring
having formula (II):
R6
R6--...õ.s..........-- R7
1
....\,--C3,44, ....,74...............
C2 R8
I
1 (II).
R6 can be selected from halo, hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy,
Ci-C6 thiohaloalkoxy, Ci-C6 alkyl, Ci-C6 haloalkyl, cyano, -NH2, -NH(C1-C6
alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and nitro. E.g., R6 can be halo (e.g., bromo). In
embodiments, each of R5, R7, and R8
can be hydrogen. Any one or more of the RI, R2, R3, R4, LI, L2, A, and Z
embodiments described herein can
be combined with any one or more of the R5, R6, R7, and R8 embodiments
described herein.
Each of LI and L2 can be CH2.; A can be CRAIRA2, wherein one of RAI and RA2 is
OR9, and the other is
hydrogen.; Z is -NRI0R11; and each of le and Ril can be independently selected
from: (a) hydrogen; (b) C6-
C10 aryl that is optionally substituted with from 1-4 Rb; (d) C1-C6 alkyl or
C1-C6 haloalkyl, each of which is
optionally substituted with from 1-3 Rd; and (I) C2-C6 alkenyl or C2-C6
alkynyl.
Each of R3 and R6 can be halo (e.g., bromo); and each of RI, R2, R4, R5, R7,
and R8 can be hydrogen.
R9 can be hydrogen. One of RI and R11 can be C6-C10 aryl that is optionally
substituted with from 1-4 Rb, and
the other is hydrogen. One of RI and R11 can be unsubstituted phenyl, and the
other is hydrogen. One of RI
and R11 can be phenyl that is substituted with 1 Rb, and the other is
hydrogen. Rb can be C1-C6 alkoxy (e.g.,
OCH3). One of RI and R11 can be 3-methoxyphenyl, and the other is hydrogen.
Each of LI and L2 is CH2.; A is CRAIRA2, wherein one of RAI and RA2 is OR9,
and the other is
hydrogen.; Z is -NRI0R11; and each of le and Ril is independently selected
from: (a) hydrogen; (b) C6-C10
aryl that is optionally substituted with from 1-4 Rb; (d) C1-C6 alkyl or C1-C6
haloalkyl, each of which is
optionally substituted with from 1-3 Rd; and (1) C2-C6 alkenyl or C2-C6
alkynyl. Embodiment can include one
or more of the following features.
Each of R3 and R6 is halo (e.g., bromo); and each of RI, R2, R4, R5, R7, and
R8 is hydrogen. R9 can be
hydrogen. One of RI and R11 can be C6-C10 aryl that is optionally substituted
with from 1-4 Rb, and the other
is hydrogen. One of RI and R11 can be unsubstituted phenyl, and the other is
hydrogen. One of RI and RII
can be phenyl that is substituted with 1 Rb, and the other is hydrogen. Rb can
be Ci-C6 alkoxy (e.g., OCH3).
One of RI and R11 can be 3-methoxyphenyl, and the other is hydrogen.
In embodiments, (A), (B), or (C) applies. In other embodiments, (A) and (B);
or (A) and (C); or (B)
and (C) applies. In still other embodiments, (A), (B), or (C) apply.
54
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Each of R and R' can be, independently, hydrogen, C1-C6 alkyl, or C1-C6
haloalkyl. Each of R and R'
can be, independently, Ci-C6 alkyl (e.g., each of R and R' can be CH3). Each
of R and R' can be hydrogen.
The compound of the present invention can include any one or more compounds
selected from:
R-1 -(3,6-Dibromo-9H-carbazol-9-y1)-3 -(3-methoxyphenylamino)-propan-2-ol;
S-1 -(3 ,6-Dibromo -9H-c arb azol-9-y1)-3 -(3 -methoxyphenylamino)-propan-2-
ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-iminopyridin-1(2H)-yl)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylthio)propan-2-ol;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(3 -
methoxyphenyl)acetamide;
54(3,6-dibromo-9H-carbazol-9-yl)methyl)-3-(3-methoxypheny1)-oxazolidin-2-one;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-propan-2-one;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-methoxypropy1)-3-methoxyaniline;
1-(3,6-Dimethy1-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol;
1-(3-Bromo-6-methy1-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-propan-2-ol;
1-(3,6-Dichloro-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol;
1-(5-bromo-2,3-dimethy1-1H-indo1-1-y1)-3-(phenylamino)propan-2-ol;
1-(3,6-Dibromo-9H-pyrido[3,4-b]indo1-9-y1)-3-(phenylamino)propan-2-ol;
1-(3-Azidophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1,3-Bis(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(9H-Carbazol-9-y1)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxy-N-(3-methoxypheny1)-propanamide;
Ethyl 5-(2-Hydroxy-3-(3-methoxyphenylamino)propy1)-8-methy1-3,4-dihydro-1H-
pyrido[4,3-
b]indole-2(5H)-carboxylate;
4-(3,6-dibromo-9H-carbazol-9-y1)-1-(phenylamino)butan-2-ol;
N-(3 -(3,6-dibromo-9H-carbazol-9-yl)propyl)aniline;
1-(3,6-dibromo-9H-carbazol-9-y1)-4-(phenylamino)butan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-2-ylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-((3-methoxyphenyl)(methyl)-amino)propan-2-
ol;
3-(3,6-dibromo-9H-carbazol-9-y1)-1-(3-methoxyphenylamino)-1-(methylthio)propan-
2-one;
3-amino-1 -(3 -(3 ,6-dibromo -9 H-carb azol-9 -y1)-2-hydro
xypropyl)pyridinium;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyrimidin-2-ylamino)propan-2-ol;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxy-N-
methylaniline;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-methoxypropan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-4-phenylbutan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(1H-indo1-1-y1)propan-2-ol;
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
3-(1-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-1H-1,2,3-triazol-4-
y1)propan-1-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-ethoxyphenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,5-dimethy1-1H-pyrazol-1-y1)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylsulfinyl)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylsulfonyl)propan-2-ol;
1-(3-bromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol;
N-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropylamino)phenoxy)penty1)-
2-(7-
(dimethylamino)-2-oxo-2H-chromen-4-y1)acetamide;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-ol;
N-(2-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropoxy)ethyl)-acetamide;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-3-ylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-4-ylamino)propan-2-ol;
1-(2,8-dimethy1-3,4-dihydro-1H-pyrido[4,3-b]indol-5(2H)-y1)-3-
(phenylamino)propan-2-ol;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2,2-difluoropropy1)-3-methoxyanifine;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(o-tolylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(m-tolylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-methoxyphenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(naphthalen-1-ylamino)propan-2-ol;
1-(4-bromophenylamino)-3-(3,6-dichloro-9H-carbazol-9-yl)propan-2-ol;
1-(4-bromophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(4-ethoxyphenylamino)propan-2-ol;
1-(4-chlorophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenethylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-hydroxyethylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2,4-dimethoxyphenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2,3-dimethylphenylamino)propan-2-ol;
1-(2-chlorophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(tert-butylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(isopropylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(4-methoxyphenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(m-tolylamino)propan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,5-dimethylphenylamino)propan-2-ol;
56
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(3 ,4-dimethylphenylamino)propan-2-ol;
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(3 ,4-dimethylphenylamino)propan-2-ol;
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(2,5 -dimethylphenylamino)propan-2-ol;
1 -(4-bromophenylamino)-3 -(2,3 -dimethy1-1H-indo1-1 -yl)propan-2-ol;
1 -(2,3 -dimethy1-1H-indo1-1 -y1)-3 -(4-methoxyphenylamino)propan-2-ol;
1 -(2,3 -dimethy1-1H-indo1-1 -y1)-3 -(4-ethoxyphenylamino)propan-2-ol;
1 -(2,3 -dimethy1-1H-indo1-1 -y1)-3 -(p-tolylamino)prop an-2-ol;
1 -(2,3 -dimethy1-1H-indo1-1 -y1)-3 -(phenylamino)propan-2-ol oxalate;
1 -(1H-indo1-1 -y1)-3 -(4-methoxyphenylamino)propan-2-ol hydrochloride;
1 -(1H-indo1-1 -y1)-3 -(phenylamino)propan-2-ol oxalate;
1 -(3 ,4-dihydro-1H-carbazol-9(2H)-y1)-3 -(m-tolylamino)propan-2-ol;
1 -(9H-carbazol-9-y1)-3 -(phenylamino)propan-2-ol;
1 -(3 ,6-dichloro-9H-carbazol-9-y1)-3 -(phenylamino)propan-2-ol;
1 -(9H-carbazol-9-y1)-3 -(p-tolylamino)propan-2-ol;
1 -(3 ,6-dichloro-9H-carbazol-9-y1)-3 -(p-tolylamino)propan-2-ol;
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(p-tolylamino)propan-2-ol;
N-(4-(3-(9H-carbazol-9-y1)-2-hydroxypropoxy)phenyl)acetamide;
1 -(9H-carbazol-9-y1)-3 -phenoxypropan-2-ol;
1 -(9H-carbazol-9-y1)-3 -(4-methoxyphenylamino)propan-2-ol;
1 -(benzylamino)-3 -(9H-carbazol-9-yl)propan-2-ol;
methyl 4-(3-(9H-carbazol-9-y1)-2-hydroxypropoxy)benzoate;
1 -(9H-carbazol-9-y1)-3 -(4-methoxyphenoxy)propan-2-ol;
1 -amino-3 -(3 ,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
(S)-1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -phenoxypropan-2-ol;
(R)-1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -phenoxypropan-2-ol;
3 ,6-dibromo-9-(2-fluoro-3 -phenoxypropy1)-9H-carbazole;
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(3 -methoxyphenylamino)-2-methylprop an-
2-ol;
1 -(2,8 -dimethy1-3,4-dihydro-1H-pyrido [4,3 -b]indo1-5(2H)-y1)-3 -(3 -
methoxyphenylamino)propan-2-ol;
1 -(4-azidophenylamino)-3 -(3 ,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1 -(3 -azido-6-bromo-9H-carbazol-9-y1)-3 -(3 -methoxyphenylamino)propan-2-ol;
1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(4-methoxyphenoxy) propan-2-ol;
1 -(3 ,6-dichloro-9H-carbazol-9-y1)-3 -(phenylsulfonyl)propan-2-ol;
3 ,6-dibromo-9-(2-fluoro-3 -(phenylsulfonyl)propy1)-9H-carbazole;
S)-1 -(3 ,6-dibromo-9H-carb azol-9-y1)-3 -(phenylsulfonyl) propan-2-ol;
(R)-1 -(3 ,6-dibromo-9H-carbazol-9-y1)-3 -(phenylsulfonyl) propan-2-ol;
57
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1-(3,6-dicyclopropy1-9H-carbazol-9-y1)-3-(phenylamino) propan-2-ol;
1-(3,6-diiodo-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
1-(3,6-diethyny1-9H-carbazol-9-y1)-3 -(3 -methoxyphenylamino) propan-2-ol;
9-(2-hydroxy-3 -(3 -methoxyphenylamino)propy1)-9H-carbazole-3,6-
dicarbonitrile;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropyl)aniline;
3,6-dibromo-9-(2,2-difluoro-3-phenoxypropy1)-9H-carbazole;
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-4-methoxyaniline;
N-(2-bromo-3-(3,6-dibromo-9H-carbazol-9-yl)propy1)-N-(4-methoxypheny1)-4-
nitrobenzenesulfonamide;
Ethyl 2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)acetate; and
N-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-4-(2-(2-
methoxyethoxy)ethoxy)aniline;
N-(2-(2-(4-(3 -(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)acetamido)ethyl)-5 -(2-
oxohexahydro-1H-thieno [3,4-d]imidazol-4-yl)pentanamide;
2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropylamino)phenoxy)-N,N-
dimethylacetamide;
2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropylamino)phenoxy)-N-(2-
hydroxyethyl)acetamide;
1-(bis(4-bromophenyl)amino)-3-(phenylamino)propan-2-ol;
(E)-3,6-dibromo-9-(3-phenoxyally1)-9H-carbazole;
(E)-3,6-dibromo-9-(3 -phenoxyprop-l-en-1 -y1)-9H-carbazole;
1-(3,6-bis(trifluoromethyl)-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
1-(2,8 -Dibromo-10,11 -dihydro-5H-dibenzo[Mazepin-5-y1)-3 -(3 -
methoxyphenylamino)propan-2-ol;
1-(3,6-Dibromo-9H-carbazol-9-y1)-3 -(3 -methoxyphenylthio)propan-2-ol;
1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(4-methoxyphenylthio)propan-2-ol;
3,6-Dibromo-9-(2-fluoro-3 -(3 -methoxyphenylthio)propy1)-9H-carbazole;
3,6-Dibromo-9-(2-fluoro-3-(4-methoxyphenylthio)propy1)-9H-carbazole;
3,6-Dibromo-9-(2-fluoro-3 -(3 -methoxyphenylsulfonyl)propy1)-9H-carbazole;
1-(3,6-Dibromo-9H-carbazol-9-y1)-3 -(3 -methoxyphenylsulfonyl)propan-2-ol;
3,6-Dibromo-9-(2-fluoro-3-(4-methoxyphenylsulfonyl)propy1)-9H-carbazole;
1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(4-methoxyphenylsulfonyl)propan-2-ol;
3 -(3 -(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropylthio)phenol;
4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropylthio)phenol;
3 -(3 -(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropylsulfonyl)phenol;
4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropylsulfonyl)phenol;
1-(3-Aminophenylthio)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
1-(4-Aminophenylthio)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
58
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-amine;
N-Benzy1-2-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropylthio)-
phenoxy)acetamide;
N-Benzy1-2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropylthio)-
phenoxy)acetamide;
3-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-fluoropropylsulfonyl)phenol;N-Benzy1-2-
(3-(3-(3,6-dibromo-
9H-carbazol-9-y1)-2-hydroxypropylsulfony1)-phenoxy)acetamide;
4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-fluoropropylsulfonyl)phenol;
5-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylamino)phenoxy)pentylcarbamoy1)-2-(6-
hydroxy-3-oxo-3H-xanthen-9-yl)benzoic acid;
1-(8-bromo-3,4-dihydro-1H-pyrido[4,3-b]indo1-5(2H)-y1)-3-phenoxypropan-2-ol;
1-(8-bromo-2-cyclopropy1-3,4-dihydro-1H-pyrido[4,3-b]indo1-5(2H)-y1)-3-
phenoxypropan-2-ol;
8-bromo-5-(2-hydroxy-3-phenoxypropy1)-3,4-dihydro-1H-pyrido[4,3-b]indole-2(5H)-
carbonitrile;
8-bromo-5-(2-fluoro-3-phenoxypropy1)-2,3,4,5-tetrahydro-1H-pyrido[4,3-
b]indole;
1-(cyclohexylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
(9-(2-hydroxy-3-(phenylthio)propy1)-9H-carbazole-3,6-dicarbonitrile;
9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3,6-dicarbonitrile;
R-N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline
S-N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline
N-(2-(3,6-dibromo-9H-carbazol-9-yl)ethyl)aniline;
2-(6-Amino-3-imino-3H-xanthen-9-y1)-4-(6-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-
y1)-2-
hydroxypropylamino)phenoxy)pentylamino)-6-oxohexylcarbamoyl)benzoic acid AND 2-
(6-amino-3-imino-
3H-xanthen-9-y1)-5-(6-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylamino)phenoxy)pentylamino)-6-oxohexylcarbamoyl)benzoic acid;
1-(8-bromo-2-methy1-3,4-dihydro-1H-pyrido[4,3-b]indo1-5(2H)-y1)-3-
phenoxypropan-2-ol;
6-((4-bromophenyl)(2-hydroxy-3-phenoxypropyl)amino)nicotinonitrile;
1-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropyl)pyridin-2(111)-one;
9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carbonitrile;
tert-butyl (5-(4-((3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropyl)sulfonyl)
phenoxy)pentyl)carbamate;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carbonitrile;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carboxamide;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-2-yloxy)propan-2-ol;
methyl 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carboxylate;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carboxylic acid;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[2,3-b]indole-3-carbonitrile;
9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[2,3-b]indole-3-carbonitrile;
59
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
tert-butyl 3-(2-(2-(2-(3-((3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)amino)phenoxy)ethoxy)ethoxy)ethoxy)propanoate;
1-(3,6-dibromo-1,4-dimethoxy-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
1-(3,6-dibromo-1,8-dimethy1-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
2-(3,6-dibromo-9H-carbazol-9-yl)acetic acid;
1-(6-bromo-3-methoxy-1-methy1-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
1-(4,6-dibromo-3-methoxy-1-methy1-9H-carbazol-9-y1)-3-(phenylamino)propan-2-
ol;
1-(3,6-dibromo-4-methoxy-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol;
9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylic acid;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylic
acid;
ethyl 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-
carboxylate;
9-(2-fluoro-3-phenoxypropy1)-9H-carbazole-3,6-dicarbonitrile;
9-(2-hydroxy-2-methyl-3-phenoxypropy1)-9H-carbazole-3,6-dicarbonitrile;
1-(cyclohexyloxy)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol;
(E)-N-(3 -(3,6-dibromo-9H-carbazol-9-yl)prop-1 -en-1 -y1)-1,1,1 -trifluoro-N-
(3 -
methoxyphenyl)methanesulfonamide;
1-(3,6-dibromo-9H-pyrido[2,3-b]indo1-9-y1)-3-phenoxypropan-2-ol;
1-(3,6-dibromo-9H-carbazol-9-y1)-3-((6-methoxypyridin-2-yl)amino)propan-2-ol;
1-(8-bromo-5H-pyrido[4,3-b]indo1-5-y1)-3-phenoxypropan-2-ol;
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxamide;
8-bromo-5-(2-hydroxy-3-phenoxypropy1)-5H-pyrido[4,3-b]indole 2-oxide;
8-bromo-5-(2-hydroxy-3-phenoxypropy1)-5H-pyrido[3,2-b]indole 1-oxide;
(6-bromo-9H-pyrido[3,4-b]indo1-3-yl)methanol;
ethyl 6-bromo-9H-pyrido[3,4-b]indole-3-carboxylate;
tert-butyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropyl)carbamate;
2-(3,6-dibromo-9H-carbazol-9-y1)-N-methylacetamide;
3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropan-1 -amine hydrochloride;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropyl)acetamide;
2-(3,6-dibromo-9H-carbazol-9-yl)propanamide;
6-bromo-9H-pyrido[3,4-b]indole-3-carbonitrile;
6-bromo-3-methy1-9H-pyrido[3,4-b]indole;
methyl (2-(3,6-dibromo-9H-carbazol-9-yl)acetyl)carbamate;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-6-methoxypyridin-2-amine;
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide;
1-(3,6-dibromo-9H-carbazol-9-y1)-34(4-methoxybenzyl)(3-
methoxyphenyl)amino)propan-2-ol;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-2,2,2-trifluoroacetamide;
tert-butyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropyl)carbamate;
5-(2-hydroxy-3-phenoxypropy1)-5H-pyrimido[5,4-b]indole-2-carboxylic acid;
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropyl)acetamide;
ethyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropyl)carbamate;
6-bromo-9-(3-(4-bromophenoxy)-2-hydroxypropy1)-9H-carbazole-3-carbonitrile;
methyl 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylate;
N-(3-(3-bromo-6-methy1-9H-carbazol-9-y1)-2-fluoropropy1)-6-methoxypyridin-2-
amine;
or a salt (e.g., a pharmaceutically acceptable salt) thereof (or any one or a
subset thereof, e.g., as
delineated in the claims).
In certain embodiments, the compound having formula (I) can be 1-(3,6-dibromo-
9H-carbazol-9-y1)-
3-(phenylamino)propan-2-ol; or a salt (e.g., a pharmaceutically acceptable
salt) thereof
In certain embodiments, the compound having formula (I)can be R-1-(3,6-Dibromo-
9H-carbazol-9-
y1)-3-(3-methoxyphenylamino)-propan-2-ol; or a salt (e.g., a pharmaceutically
acceptable salt) thereof In
embodiments, R-1 -(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-
propan-2-ol or a salt (e.g., a
pharmaceutically acceptable salt) thereof can be substantially free of (e.g.,
contains less than about 5% of, less
than about 2% of, less than about 1%, less than about 0.5% of) S-1-(3,6-
Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol or a salt (e.g., a pharmaceutically acceptable
salt) thereof
In certain embodiments, the compound having formula (I) can be S-1-(3,6-
Dibromo-9H-carbazol-9-
y1)-3-(3-methoxyphenylamino)-propan-2-ol; or a salt (e.g., a pharmaceutically
acceptable salt) thereof In
embodiments, S-1 -(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-
propan-2-ol or a salt (e.g., a
pharmaceutically acceptable salt) thereof can be substantially free of (e.g.,
contains less than about 5% of, less
than about 2% of, less than about 1%, less than about 0.5% of) R-1-(3,6-
Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol or a salt (e.g., a pharmaceutically acceptable
salt) thereof
In certain embodiments, the compound having formula (I) can be the (+)
(dextrorotatory) enantiomer
of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-propan-2-ol as
described herein or a salt
(e.g., a pharmaceutically acceptable salt) thereof See, e.g., Example la and
lb. In embodiments, the (+)
(dextrorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol as
described herein or a salt (e.g., a pharmaceutically acceptable salt) thereof
can be substantially free of (e.g.,
contains less than about 5% of, less than about 2% of, less than about 1%,
less than about 0.5% of) the (-)
(levorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol as
described herein or a salt (e.g., a pharmaceutically acceptable salt) thereof
61
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, the compound having formula (I) can be the (-)
(levorotatory) enantiomer of
1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-propan-2-ol as
described herein or a salt (e.g.,
a pharmaceutically acceptable salt) thereof See, e.g., Example la and lb. In
embodiments, the (-)
(levorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol as
described herein or a salt (e.g., a pharmaceutically acceptable salt) thereof
can be substantially free of (e.g.,
contains less than about 5% of, less than about 2% of, less than about 1%,
less than about 0.5% of) the (+)
(dextrorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol as
described herein or a salt (e.g., a pharmaceutically acceptable salt) thereof
In certain embodiments, the compound can be (+) (dextrorotatory)-N-(3-(3,6-
dibromo-9H-carbazol-9-
y1)-2-fluoropropy1)-3-methoxyaniline as described herein or a salt (e.g., a
pharmaceutically acceptable salt)
thereof See, e.g., Example 144a and 144b. In embodiments, the (+)
(levorotatory) enantiomer of N-(3-(3,6-
dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline as described herein
or a salt (e.g., a
pharmaceutically acceptable salt) thereof can be substantially free of (e.g.,
contains less than about 5% of, less
than about 2% of, less than about 1%, less than about 0.5% of) the (-)
(dextrorotatory) enantiomer of N-(3-
(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline as described
herein or a salt (e.g., a
pharmaceutically acceptable salt) thereof
In certain embodiments, the compound can be (-) (dextrorotatory)-N-(3-(3,6-
dibromo-9H-carbazol-9-
y1)-2-fluoropropy1)-3-methoxyaniline as described herein or a salt (e.g., a
pharmaceutically acceptable salt)
thereof See, e.g., Example 144a and 144b. In embodiments, the (-)
(levorotatory) enantiomer of N-(3-(3,6-
dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline as described herein
or a salt (e.g., a
pharmaceutically acceptable salt) thereof can be substantially free of (e.g.,
contains less than about 5% of, less
than about 2% of, less than about 1%, less than about 0.5% of) the (+)
(dextrorotatory) enantiomer of N-(3-
(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline as described
herein or a salt (e.g., a
pharmaceutically acceptable salt) thereof
Compounds of formula (I), (II), (III), and (IV) are featured, including title
compounds of Examples la,
lb, 3a, 3b, 3d, 6a, 10, 13, 21, 22, 88b, 90, 92, 96, 97a, 97b, 102, 116, 117,
118, 119, 120, 121, 122, 132, 143,
and 144a; or a pharmaceutically acceptable salt thereof
In various embodiments, compounds of formula (I), (II), (III), and (IV) can be
used in a method for
the treatment of a disease, disorder, or condition caused by unwanted neuronal
cell death or associated with
insufficient neurogenesis in a subject in need thereof The method can include
administering to the subject an
effective amount of a compound having formula (I), (II), (III), or (VI), or a
pharmaceutically acceptable salt
thereof, as defined herein.
The methods can further include detecting a resultant neurotrophism (e.g.,
neurogenesis; and/or
determining that the patient has aberrant neurotrophism, particularly aberrant
neurogenesis, particularly
62
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
aberrant hippocampal and/or hypothalamic neurogenesis, or a disease or
disorder associated therewith,
particularly by detecting and/or diagnosing the same.
The methods can further include detecting a resultant neurotrophism.
The methods can further include detecting determining that the subject has
aberrant neurogenesis or
death of neurons or a disease or disorder associated therewith, by detecting
the same in said subject.
The methods can further include detecting a resultant hippocampal and/or
hypothalamic neurogenesis.
The disease, disorder, or condition can be a neuropsychiatric and
neurodegenerative disease, including
(but not limited to) schizophrenia, major depression, bipolar disorder, normal
aging, epilepsy, traumatic brain
injury, post-traumatic stress disorder, Parkinson's disease, Alzheimer's
disease, Down syndrome,
spinocerebellar ataxia, amyotrophic lateral sclerosis, Huntington's disease,
stroke, radiation therapy, chronic
stress, and abuse of neuro-active drugs (such as alcohol, opiates,
methamphetamine, phencyclidine, and
cocaine), retinal degeneration, spinal cord injury, peripheral nerve injury,
physiological weight loss associated
with various conditions, and cognitive decline associated with normal aging,
and chemotherapy.
In some embodiments, the compounds having formula (I) or a salt (e.g., a
pharmaceutically acceptable
salt) thereof provide at least about 27 (x10- 6) BrdU+ cells / mm3 dentate
gyms when evaluated in the assay
described in conjunction with Table 1 (i.e., evaluated for pro-neurogenic
efficacy / neuroprotection in our
standard in vivo assay at 10 .1\4 concentration in four 12 week old adult
male C57/B16 mice..
In some embodiments, the compounds having formula (I) or a salt (e.g., a
pharmaceutically acceptable
salt) thereof provide at least about 19 (x10- 6) BrdU+ cells / mm3 dentate
gyms when evaluated in the assay
described in conjunction with Table 1.
In some embodiments, the compounds having formula (I) or a salt (e.g., a
pharmaceutically acceptable
salt) thereof provide from about 18 to about 30 (e.g., 18-27, 19-26, 20-25, 27-
30, 27-29) (x10- 6) BrdU+ cells /
mm3 dentate gyms when evaluated in the assay described in conjunction with
Table 1.
In some embodiments, the compounds having formula (I) or a salt (e.g., a
pharmaceutically acceptable
salt) thereof provide from about 18 to about 26 (e.g., 19-26, 20-25) (x10- 6)
BrdU+ cells / mm3 dentate gyms
when evaluated in the assay described in conjunction with Table 1.
In some embodiments, the compounds having formula (I) or a salt (e.g., a
pharmaceutically acceptable
salt) thereof provide from about 27 to about 30 (e.g., 27-29) (x10- 6) BrdU+
cells / mm3 dentate gyms when
evaluated in the assay described in conjunction with Table 1.
In embodiments, a composition (e.g., a pharmaceutical composition) can include
an amount effective
to achieve the levels described above.
In embodiments, any compound, composition, or method described herein can also
include any one or
more of the other features delineated in the detailed description and/or in
the claims.
63
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Definitions
The term "mammal" includes organisms, which include mice, rats, cows, sheep,
pigs, rabbits, goats,
horses, monkeys, dogs, cats, and humans.
"An effective amount" refers to an amount of a compound that confers a
therapeutic effect (e.g., treats,
e.g., controls, relieves, ameliorates, alleviates, or slows the progression
of; or prevents, e.g., delays the onset of
or reduces the risk of developing, a disease, disorder, or condition or
symptoms thereof) on the treated subject.
The therapeutic effect may be objective (i.e., measurable by some test or
marker) or subjective (i.e., subject
gives an indication of or feels an effect). An effective amount of the
compound described above may range
from about 0.01 mg/kg to about 1000 mg/kg, (e.g., from about 0.1 mg/kg to
about 100 mg/kg, from about 1
mg/kg to about 100 mg/kg). Effective doses will also vary depending on route
of administration, as well as the
possibility of co-usage with other agents.
The term "halo" or "halogen" refers to any radical of fluorine, chlorine,
bromine or iodine.
In general, and unless otherwise indicated, substituent (radical) prefix names
are derived from the
parent hydride by either (i) replacing the "ane" in the parent hydride with
the suffixes "yl," "diyl," "triyl,"
"tetrayl," etc.; or (ii) replacing the "e" in the parent hydride with the
suffixes "yl," "diyl," "triyl," "tetrayl," etc.
(here the atom(s) with the free valence, when specified, is (are) given
numbers as low as is consistent with any
established numbering of the parent hydride). Accepted contracted names, e.g.,
adamantyl, naphthyl, anthryl,
phenanthryl, furyl, pyridyl, isoquinolyl, quinolyl, and piperidyl, and trivial
names, e.g., vinyl, allyl, phenyl,
and thienyl are also used herein throughout. Conventional numbering/lettering
systems are also adhered to for
substituent numbering and the nomenclature of fused, bicyclic, tricyclic,
polycyclic rings.
The following definitions are used, unless otherwise described. Specific and
general values listed
below for radicals, substituents, and ranges, are for illustration only; they
do not exclude other defined values
or other values within defined ranges for the radicals and substituents.
Unless otherwise indicated, alkyl,
alkoxy, alkenyl, and the like denote both straight and branched groups.
The term "alkyl" refers to a saturated hydrocarbon chain that may be a
straight chain or branched
chain, containing the indicated number of carbon atoms. For example, C1-C6
alkyl indicates that the group
may have from 1 to 6 (inclusive) carbon atoms in it. Any atom can be
optionally substituted, e.g., by one or
more subsituents. Examples of alkyl groups include without limitation methyl,
ethyl, n-propyl, isopropyl, and
tert-butyl.
As used herein, the term "straight chain Cii, alkylene," employed alone or in
combination with other
terms, refers to a non-branched divalent alkyl linking group having n to m
carbon atoms. Any atom can be
optionally substituted, e.g., by one or more subsituents. Examples include
methylene (i.e., -CH2-).
The term "haloalkyl" refers to an alkyl group, in which at least one hydrogen
atom is replaced by halo.
In some embodiments, more than one hydrogen atom (e.g., 2, 3, 4, 5, 6, 7, 8,
9, 10, 11, 12, 13, or 14) are
replaced by halo. In these embodiments, the hydrogen atoms can each be
replaced by the same halogen (e.g.,
64
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
fluoro) or the hydrogen atoms can be replaced by a combination of different
halogens (e.g., fluoro and chloro).
"Haloalkyl" also includes alkyl moieties in which all hydrogens have been
replaced by halo (sometimes
referred to herein as perhaloalkyl, e.g., perfluoroalkyl, such as
trifluoromethyl). Any atom can be optionally
substituted, e.g., by one or more substituents.
As referred to herein, the term "alkoxy" refers to a group of formula
-0(alkyl). Alkoxy can be, for example, methoxy (-0CH3), ethoxy, propoxy,
isopropoxy, butoxy, iso-butoxy,
sec-butoxy, pentoxy, 2-pentoxy, 3-pentoxy, or hexyloxy. Likewise, the term
"thioalkoxy" refers to a group of
formula -S(alkyl). Finally, the terms "haloalkoxy" and "thioalkoxy" refer to -
0(haloalkyl) and -S(haloalkyl),
respectively. The term "sulfhydryl" refers to -SH. As used herein, the term
"hydroxyl," employed alone or in
combination with other terms, refers to a group of formula -OH.
The term "aralkyl" refers to an alkyl moiety in which an alkyl hydrogen atom
is replaced by an aryl
group. One of the carbons of the alkyl moiety serves as the point of
attachment of the aralkyl group to another
moiety. Any ring or chain atom can be optionally substituted e.g., by one or
more substituents. Non-limiting
examples of "aralkyl" include benzyl, 2-phenylethyl, and 3-phenylpropyl
groups.
The term "alkenyl" refers to a straight or branched hydrocarbon chain
containing the indicated number
of carbon atoms and having one or more carbon-carbon double bonds. Any atom
can be optionally
substituted, e.g., by one or more substituents. Alkenyl groups can include,
e.g., vinyl, allyl, 1-butenyl, and 2-
hexenyl. One of the double bond carbons can optionally be the point of
attachment of the alkenyl substituent.
The term "alkynyl" refers to a straight or branched hydrocarbon chain
containing the indicated number
of carbon atoms and having one or more carbon-carbon triple bonds. Alkynyl
groups can be optionally
substituted, e.g., by one or more substituents. Alkynyl groups can include,
e.g., ethynyl, propargyl, and 3-
hexynyl. One of the triple bond carbons can optionally be the point of
attachment of the alkynyl substituent.
The term "heterocycly1" refers to a fully saturated monocyclic, bicyclic,
tricyclic or other polycyclic
ring system having one or more constituent heteroatom ring atoms independently
selected from 0, N (it is
understood that one or two additional groups may be present to complete the
nitrogen valence and/or form a
salt), or S. The heteroatom or ring carbon can be the point of attachment of
the heterocyclyl substituent to
another moiety. Any atom can be optionally substituted, e.g., by one or more
substituents. Heterocyclyl
groups can include, e.g., tetrahydrofuryl, tetrahydropyranyl, piperidyl
(piperidino), piperazinyl, morpholinyl
(morpholino), pyrrolinyl, and pyrrolidinyl. By way of example, the phrase
"heterocyclic ring containing from
5-6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C6 alkyl),
NC(0)(C1-C6 alkyl), 0, and S; and wherein said heterocyclic ring is optionally
substituted with from 1-3
independently selected Ra" would include (but not be limited to)
tetrahydrofuryl, tetrahydropyranyl, piperidyl
(piperidino), piperazinyl, morpholinyl (morpholino), pyrrolinyl, and
pyrrolidinyl.
The term "heterocycloalkenyl" refers to partially unsaturated monocyclic,
bicyclic, tricyclic, or other
polycyclic hydrocarbon groups having one or more (e.g., 1-4) heteroatom ring
atoms independently selected
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
from 0, N (it is understood that one or two additional groups may be present
to complete the nitrogen valence
and/or form a salt), or S. A ring carbon (e.g., saturated or unsaturated) or
heteroatom can be the point of
attachment of the heterocycloalkenyl substituent. Any atom can be optionally
substituted, e.g., by one or more
substituents. Heterocycloalkenyl groups can include, e.g., dihydropyridyl,
tetrahydropyridyl, dihydropyranyl,
4,5-dihydrooxazolyl, 4,5-dihydro-1H-imidazolyl, 1,2,5,6-tetrahydro-
pyrimidinyl, and 5,6-dihydro-2H-
[1,3]oxazinyl.
The term "cycloalkyl" refers to a fully saturated monocyclic, bicyclic,
tricyclic, or other polycyclic
hydrocarbon groups. Any atom can be optionally substituted, e.g., by one or
more substituents. A ring carbon
serves as the point of attachment of a cycloalkyl group to another moiety.
Cycloalkyl moieties can include,
e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl,
adamantyl, and norbornyl
(bicycle[2.2.1]hepty1).
The term "cycloalkenyl" refers to partially unsaturated monocyclic, bicyclic,
tricyclic, or other
polycyclic hydrocarbon groups. A ring carbon (e.g., saturated or unsaturated)
is the point of attachment of the
cycloalkenyl substituent. Any atom can be optionally substituted e.g., by one
or more substituents.
Cycloalkenyl moieties can include, e.g., cyclohexenyl, cyclohexadienyl, or
norbornenyl.
As used herein, the term "cycloalkylene" refers to a divalent monocyclic
cycloalkyl group having the
indicated number of ring atoms.
As used herein, the term "heterocycloalkylene" refers to a divalent monocyclic
heterocyclyl group
having the indicated number of ring atoms.
The term "aryl" refers to an aromatic monocyclic, bicyclic (2 fused rings), or
tricyclic (3 fused rings),
or polycyclic (>3 fused rings) hydrocarbon ring system. One or more ring atoms
can be optionally
substituted, e.g., by one or more substituents. Aryl moieties include, e.g.,
phenyl and naphthyl.
The term "heteroaryl" refers to an aromatic monocyclic, bicyclic (2 fused
rings), tricyclic (3 fused
rings), or polycyclic (> 3 fused rings) hydrocarbon groups having one or more
heteroatom ring atoms
independently selected from 0, N (it is understood that one or two additional
groups may be present to
complete the nitrogen valence and/or form a salt), or S. One or more ring
atoms can be optionally substituted,
e.g., by one or more substituents.
Examples of heteroaryl groups include, but are not limited to, 2H-pyrrolyl, 3H-
indolyl, 4H-
quinolizinyl, acridinyl, benzo[b]thienyl, benzothiazolyl, P-carbolinyl,
carbazolyl, coumarinyl, chromenyl,
cinnolinyl, dibenzo[b,d]furanyl, furazanyl, furyl, imidazolyl, imidizolyl,
indazolyl, indolyl, isobenzofuranyl,
isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxazolyl,
perimidinyl, phenanthridinyl,
phenanthrolinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxathiinyl,
phenoxazinyl, phthalazinyl,
pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl,
pyrimidinyl, pyrrolyl, quinazolinyl,
quinolyl, quinoxalinyl, thiadiazolyl, thianthrenyl, thiazolyl, thienyl,
triazolyl, and xanthenyl.
66
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The terms "arylcycloalkyl" and "arylheterocycly1" refer to bicyclic,
tricyclic, or other polycyclic ring
systems that include an aryl ring fused to a cycloalkyl and heterocyclyl,
respectively. Similarly, the terms
"heteroarylheterocyclyl," and "heteroarylcycloalkyl" refer to bicyclic,
tricyclic, or other polycyclic ring
systems that include a heteroaryl ring fused to a heterocyclyl and cycloalkyl,
respectively. Any atom can be
substituted, e.g., by one or more substituents. For example, arylcycloalkyl
can include indanyl;
arylheterocyclyl can include 2,3-dihydrobenzofuryl, 1,2,3,4-
tetrahydroisoquinolyl, and 2,2-
dimethylchromanyl.
The descriptors "C=0" or "C(0)" refers to a carbon atom that is doubly bonded
to an oxygen atom.
The term "oxo" refers to double bonded oxygen when a substituent on carbon.
When oxo is a
substituent on nitrogen or sulfur, it is understood that the resultant groups
has the structures N¨>0- and 5(0)
and SO2, respectively.
As used herein, the term "cyano," employed alone or in combination with other
terms, refers to a
group of formula -CN, wherein the carbon and nitrogen atoms are bound together
by a triple bond.
In general, when a definition for a particular variable includes both hydrogen
and non-hydrogen (halo,
alkyl, aryl, etc.) possibilities, the term "substituent(s) other than
hydrogen" refers collectively to the non-
hydrogen possibilities for that particular variable.
The term "substituent" refers to a group "substituted" on, e.g., an alkyl,
haloalkyl, cycloalkyl,
heterocyclyl, heterocycloalkenyl, cycloalkenyl, aryl, or heteroaryl group at
any atom of that group. In one
aspect, the substituent(s) on a group are independently any one single, or any
combination of two or more of
the permissible atoms or groups of atoms delineated for that substituent. In
another aspect, a substituent may
itself be substituted with any one of the above substituents.
Further, as used herein, the phrase "optionally substituted" means
unsubstituted (e.g., substituted with
a H) or substituted. As used herein, the term "substituted" means that a
hydrogen atom is removed and
replaced by a substituent. It is understood that substitution at a given atom
is limited by valency.
Descriptors such as "C6-C10 aryl that is optionally substituted with from 1-4
independently selected
Rb" (and the like) is intended to include both an unsubstituted C6-C10 aryl
group and a C6-Cio aryl group that is
substituted with from 1-4 independently selected Rb. The use of a substituent
(radical) prefix names such as
alkyl without the modifier "optionally substituted" or "substituted" is
understood to mean that the particular
substituent is unsubstituted. However, the use of "haloalkyl" without the
modifier "optionally substituted" or
"substituted" is still understood to mean an alkyl group, in which at least
one hydrogen atom is replaced by
halo.
In some embodiments, Rb can be as defined in any one, two, three, or all of
(aa) through (dd). For
example, Rb can be as defined in (aa) and (bb) or combinations thereof
67
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The phrase "Cy is a saturated, partially unsaturated or aromatic carbocyclic
or heterocyclic ring
system" in the definition of Re is understood to include each of the rings
systems defined above (e.g., Cy can
be coumarinyl or the ring component of biotin optionally substituted as
defined anywhere herein).
The details of one or more embodiments are set forth in the description below.
Other features and
advantages of the presently disclosed embodiments will be apparent from the
description and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: Pulse-chase analysis of BrdU-labeling identified magnitude and
timing of cell death following birth
of new neurons in the dentate gyms. 12 week old wild type male C57/B6 mice
were individually housed
without access to running wheels and injected on day 0 with BrdU (50 mg/kg,
i.p.). Neural precursor cell
proliferation in the dentate gyms (DG) subgranular zone (SGZ) and granular
layer (GL) was subsequently
monitored through immunohistochemistry for BrdU on days 1, 5, 10, 15, 20, and
25 days post-injection. Four
mice were evaluated at each time point, and 25-30 adjacent coronal sections
through the hippocampus
(progressing posteriorly from the point where the suprapyramidal and
infrapyramidal blades are joined at the
crest region and the dentate gyrus is oriented horizontally beneath the corpus
callosum) from each mouse were
examined. On days 1 and 5, almost 100% of BrdU-positive cells within the DG
were localized in the SGZ.
The total number of cells decreased approximately 40% between days 1 and 5, in
accordance with the
appearance of apoptotic cell bodies in the SGZ. By day 10, some BrdU positive
cells had migrated into the
GL, with no significant change in total number of BrdU-positive cells in the
DG. By day 15, BrdU-positive
cells in the SGZ declined as the number of BrdU-positive cells in the GL
stayed constant, suggesting that some
of the cells migrating out of the SGZ and into the GL between days 10 and 15
underwent apoptosis. This trend
continued through days 20-25. These results indicated that daily injection of
BrdU over a one week period of
continuous molecule infusion, a time period during which 40% of newborn cells
in the SGZ normally die,
would allow detection of compounds that enhance either proliferation or
survival of newborn cells in the
dentate gyms.
Figure 2: Surgical placement of cannula and pumps did not affect hippocampal
neurogenesis or survival of
newborn neurons on the contralateral side of the brain. Mice infused with
vehicle (artificial cerebrospinal
fluid) over seven days by means of surgically implanted Alzet osmotic
minipumps (Vehicle Infusion, n=5)
displayed no difference in hippocampal neural precursor cell proliferation, as
assessed by BrdU incorporation
normalized for dentate gyrus volume, from mice treated identically except not
having undergone surgery (No
Surgery, n=4). When Alzet osmotic minipumps were loaded with fibroblast growth
factor 2 (FGF-2; 10
mg/mL) (n=5), however, hippocampal neural precursor cell proliferation roughly
doubled with respect to both
of the other two groups(*, p<0.001, Student's t test).
68
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Figure 3: Ectopic incorporation of BrdU served to eliminate molecules from
further consideration.
Immunohistochemical staining of BrdU in the hippocampal field should normally
be restricted to the SGZ of
the dentate gyrus, as shown on the left. The in vivo neurogenic screen
employed was designed to detect small
molecules that selectively stimulated BrdU incorporation into replicating
cells of the SGZ. Infrequently, some
compounds exhibited non-specific BrdU incorporation in ectopic regions, such
as CA3, CA1, cortex, and
striatum, as shown on the right. Any molecules that demonstrated ectopic
incorporation of BrdU were
eliminated from the study.
Figure 4: Screening of 100 pools of 10 compounds identified 10 pools with pro-
neurogenic efficacy. The
total number of BrdU-labeled cells in the dentate gyms subgranular zone (SGZ)
approximately doubled
following seven day infusion with fibroblast growth factor 2 (FGF-2; 10 mg/mL)
(n=5) relative to mice
infused with vehicle (artificial cerebrospinal fluid (aCSF) (n=5). Each pool
of ten compounds was tested for
pro-neurogenic efficacy over a 7 day period in two independent mice at 10
.1\4 concentration for each
individual compound. Pools 7, 14, 18, 19, 41, 53, 54, 61, 69 and 70 displayed
comparable stimulation of
neural precursor cell proliferation as FGF-2 infusion. The majority of pools
displayed no effect on
hippocampal neural precursor cell proliferation.
Figure 5: Re-evaluation of positive pools verified statistical significance of
enhanced BrdU-incorporation.
Subsequent to their initial identification, pools 7, 14, 18, 19, 41, 53, 54,
61, 69, and 70 were re-evaluated in 2
additional mice each. Results shown are average with SEM of all 4 mice
evaluated for each compound. All
pools significantly (*, P<0.001, Student's t test) stimulated neural precursor
cell proliferation in the
hippocampal dentate gyms SGZ relative to vehicle control.
Figure 6: Pro-neurogenic pools were broken down to identify individual pro-
neurogenic compounds. Figure
6A- In vivo evaluation of the ten individual compounds that composed pool #7
revealed that compound #3
stimulated either the proliferation or survival of neural precursor cells in
the SGZ, whereas the remaining
individual components of pool #7 did not. In this document this molecule is
interchangeably referred to as
"P7C3" or "Example 45 Compound." Each compound was infused at two different
concentrations (100 .1\4
(A and B) and 10 ,M (C and D)) in two mice each. Example 45 Compound showed
either pro-neurogenic or
neuroprotective activity at both concentrations. Below the graphs are typical
results of BrdU incorporation in
the SGZ, which is notably greater in animals infused with either Pool #7 or
Example 45 Compound. Figure
6B- Molecular formulas and weights of individual pro-neurogenic compounds
identified through the in vivo
screen. Figure 6C- Re-supplied compounds were evaluated in three mice per
compound at 10[tM
concentration to verify that the pro-neurogenic or neuroprotective effect on
neural stem cells was not an
artifact of storage conditions in the UTSWMC chemical compound library. Re-
supplied compounds were
69
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
verified to be 99% pure by mass spectrometry and shown to retain either pro-
proliferative or neuroprotective
properties in vivo in neural stem cells. All compounds significantly (*,
P<0.001, Student's t test) stimulated
neural precursor cell proliferation in the hippocampal dentate gyrus SGZ
relative to vehicle control.
Figure 7: Neurogenic efficacy of orally administered Example 45 Compound was
dose-related. The graph on
the top shows that the concentration of Example 45 Compound in brain tissue of
mice that were administered
compound by daily oral gavage for 7 consecutive days correlated with the dose
of Example 45 Compound
administered. The graph on the bottom shows that pro-neurogenic or
neuroprotective efficacy of Example 45
Compound was roughly double that of vehicle control at doses ranging from 5 to
40 mg/kg. At decreasing
dosage of Example 45 Compound the amount of neurogenesis decreased
accordingly, until it reached levels no
greater than vehicle control at compound doses below 1.0 mg/kg. Results shown
are the average obtained
from analysis of 5 adult wild type male mice at each dose.
Figure 8: Analysis of molecules related structurally to Example 45 Compound
(P7C3) revealed a region of
the compound that could be chemically modified without loss of in vivo
activity. An in vivo SAR study was
conducted using 37 chemical analogs of Example 45 Compound, each evaluated in
4 or 5 adult C57/B6 male
mice. Some analogs revealed activity comparable to the parent compound,
whereas others showed
significantly diminished activity, or evidence of pro-neurogenic effect
intermediate between vehicle and FGF
controls. This exercise enabled identification of regions of the parent
compound that might be amenable to
chemical modification without loss of activity. As an example, Example 62
Compound retained robust
activity with the aniline ring of Example 45 Compound substituted by an
anisidine. This derivative compound
was exploited to yield a fluorescent derivative by attaching a coumarin moiety
to the N-phenyl ring.
Figure 9: Activity of Example 62 Compound is enantiomer-specific. Figure 9A-
(+) and (-) enantiomers of
Example 62 Compound were prepared. Figure 9B- Evaluation of Example 62
Compound enantiomers
showed that in vivo pro-neurogenic or neuroprotective efficacy was fully
retained by the (+) enantiomer in a
dose-dependent manner, while the (-) enantiomer showed diminished activity.
Each enantiomer was evaluated
at each dose in between 3 and 5 three month old adult wild type male C57/B6
mice.
Figure 10: Example 45 Compound enhances the survival of newborn neurons in the
dentate gyrus. Figure
10A- Immunohistochemical staining for doublecortin (DCX), an antigen
specifically and transiently expressed
in proliferating hippocampal neural precursor cells when they become
irreversibly committed to neuronal
differentiation, was substantially increased in newborn neurons in mice that
were administered Example 45
Compound (20 mg/kg) daily for 30 days by oral gavage, relative to that seen in
mice that received vehicle
only. These results are representative of 10 sections each from 5 mice in each
group, and demonstrate that
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 45 Compound specifically promoted hippocampal neurogenesis. (Figure
10B) Example 45
Compound enhances hippocampal neurogenesis by promoting survival of newborn
neurons. Three month old
wild type C57/B6 male mice were exposed to orally-delivered Example 45
Compound or vehicle for 30 days
(n=5 animals / group), administered a single pulse of BrdU via IP injection
(150 mg/kg), and then sacrificed 1
hour, 1 day, 5 days or 30 days later for immunohistochemical detection of BrdU
incorporation into cells
localized in the subgranular layer of the dentate gyms. No significant
differences were observed between
groups at the 1 hour or 1 day time points, though at one day there was a trend
towards increased BrdU+ cells
in the Example 45 Compound-treated group. At the 5 day time point, by which
time 40% of newborn neurons
normally die, animals that received Example 45 Compound showed a statistically
significant (*, P<0.001,
Student's t test) 25% increase in BrdU+ cells compared to the vehicle-only
control group. This difference
between groups progressed with time such that mice that received a daily oral
dose of Example 45 Compound
for 30 days, starting 24 hours after the pulse administration of BrdU,
exhibited a 5-fold increase in the
abundance of BrdU+ cells in the dentate gyms relative to vehicle-only
controls. In this longer-term trial,
BrdU+ cells were observed both in the SGZ and the granular layer of the
dentate gyms.
Figure 11: Quantification of short term (1 hour pulse) BrdU incorporation and
cleaved-caspase 3 (CCSP3)
formation in the dentate gyrus showed that NPAS3-deficient mice have the same
rate of proliferation of
newborn cells in the dentate as wild type littermates (BrdU), but roughly
twice the level of programmed cell
death (CCSP3) (*, P<0.01, Student's t test). Three 6 week old male mice (NPAS3-
deficient or wild type
littermates) in each group were evaluated.
Figure 12: Granule cell neurons in the dentate gyms of NPAS3-deficient mice
displayed morphological
deficits in dendritic branching and spine density. (Figure 12A) Golgi-Cox
staining of the dentate gyms
illustrates that dendritic arborization of dentate gyrus granule cell neurons
in npas3-/- mice is substantially less
developed than in wild type littermates. Results shown are representative of
15 sections from five 12-14 week
old adult male mice of each genotype. (Figure 12B) In addition to obviously
reduced dendritic length and
branching, granular neurons in the dentate gyrus of npas3-/- mice also
exhibited significantly reduced spine
density relative to wild type littermates (*, P < 0.00001, Student's t test).
These genotype-specific differences
were not exhibited by neurons in the CA1 region of the hippocampus.
Figure 13: In hippocampal slice preparation from npas3-/- mice, synaptic
transmission was increased both in
the outer molecular layer of the dentate gyrus (Figure 13A) and the CA1 region
of the hippocampus (Figure
13B) relative to hippocampal slices from wild type mice. Extended treatment
with Example 45 Compound
normalized synaptic responses in the dentate gyms but not the CA1 region of
npas3-/- mice. Extended
71
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
treatment with Example 45 Compound did not affect wild-type responses. Data
are presented as the mean
SEM. Each group consisted of 1 or 2 slice preparation from each of 5 mice.
Figure 14: Example 45 Compound has pro-neurogenic or neuroprotective efficacy
in the dentate gyms of
NPAS3-deficient animals. Six 12 week old npas3-/- mice were orally
administered vehicle or Example 45
Compound (20 mg/kg/d) for 12 days, and also injected daily with BrdU (50
mg/kg). At the end of day 12,
mice were sacrificed and tissue was stained for BrdU and doublecortin (DCX).
BrdU staining showed that
Example 45 Compound increased the magnitude of neurogenesis in npas3-/- mice
by roughly 4-fold, as
graphically represented above (*, P<0.001, Student's t test). DCX staining
shows that Example 45 Compound
also promoted more extensive process formation in differentiating neurons of
the adult dentate gyms in npas3-
/-
mice.
Figure 15: Golgi-Cox staining of neurons in the dentate gyrus shows that
extended daily treatment of npas3-/-
mice with Example 45 Compound (20 mg/kg/d) enhanced dendritic arborization. Hi-
power micrographs are
shown on top, and a lower power micrograph illustrating the entire dentate
gyms is shown below.
Figure 16: Measured thickness of hippocampal subfields in npas3-/- and wild
type littermate mice that were
treated with Example 45 Compound (20 mg/kg/d) or vehicle every day from
embryonic day 14 until 3 months
of age demonstrated that Example 45 Compound selectively increased the
thickness of the dentate gyrus
granular cell layer to a level approaching wild type thickness (*, P<0.01,
Student's t test), without affecting
thickness of the pyramidal cell layers of CA1 or CA3 regions.
Figure 17: Immunohistochemical detection of cleaved caspase 3 (CCSP3), a
marker of apoptosis, showed
elevated levels of programmed cell death in the dentate gyrus of NPAS3-
deficient animals. Apoptosis in
NPAS3-deficient animals was inhibited by treatment with Example 45 Compound
(20 mg/kg/d, p.o., for 12
days), whereas analogous treatment with vehicle alone had no effect. Images
shown are representative of 10-
12 sections evaluated per animal, with 3-5 eight-week-old male NPAS3-deficient
mice per group.
Figure 18: Example 45 Compound acts mechanistically in the mitochondria.
(Figure 18A) Example 45
Compound preserved mitochondrial membrane potential following exposure to the
calcium ionophore A23187
in a dose dependent manner as judged by fluorescent imaging of TMRM dye, a
cell-permeant, cationic red-
orange fluorescent dye that is readily sequestered by intact mitochondria.
(Figure 18B) The protective effect
of Example 62 Compound was enantiomeric specific, with the (+) enantiomer
retaining activity more so than
the (-) enantiomer.
72
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Figure 19: Example 45 Compound as compared to a known drug. (Figure 19A) Both
Example 45
Compound and the Dimebon anti-histamine enhanced hippocampal neurogenesis
(Figure 19B), and protected
mitochondria from dissolution following toxic exposure to the calcium
ionophore A23187 (Figure 19C). In
the in vivo assay of neurogenesis the Example 45 Compound exhibited a higher
ceiling of efficacy than the
Dimebon anti-histamine. In all three assays, the Example 45 Compound performed
with greater relative
potency than the Dimebon anti-histamine.
Figure 20: Effect of Example 45 Compound in aged rats. (Figure 20A) Example 45
Compound (20 mg/kg/d,
i.p.) and BrdU (50 mg / kg, i.p.) were administered daily for 7 days to 12-18
month old Fisher 344 rats (n = 4
in each group). P7C3 promoted neural precursor cell proliferation by roughly 5
fold compared to vehicle. (*p
<0.001, Students t test). DCX staining demonstrates that P7C3 specifically
promoted neuronal differentiation
and dendritic branching. These micrographs were taken at the same
magnification. Scale bar = 50 mm. Data
are expressed as mean +/- SEM. (Figure 20B) Latency to find the hidden
platform in the Morris water maze
task, as well as (Figure 20C) swim speed and locomotor activity (Figure 20D)
in aged rats treated with P7C3
or vehicle both before and after 2 months of treatment did not differ between
groups. Data are expressed as
mean +/- SEM. (Figure 20E) Quantification of food intake (upper panel) and
fasting blood glucose levels in
aged rats did not differ with respect to whether rats received P7C3 or
vehicle. Data are expressed as mean +/-
SEM.
Figure 21: Example 45 Compound Enhances Hippocampal Neurogenesis, Ameliorates
Cognitive Decline, and
Prevents Weight Loss in Terminally Aged Rats (Figure 21A) Prior to treatment,
both groups (n -- 23 for each
group) showed similar frequency of crossings through the goal platform. After
2 months of treatment,
however, Example 45 Compound-treated rats displayed a statistically
significant increase of crossings through
the goal platform area relative to vehicle treated rats. (Figure 21B) Example
45 Compound-treated rats
displayed significantly enhanced hippocampal neurogenesis, as assessed by BrdU
incorporation, relative to
vehicle treated rats. Many more of the BrdU-labeled cells were noted to have
migrated into the granular layer
in Example 45 Compound-treated rats in comparison to vehicle treated animals,
consistent with their
functional incorporation into the dentate gyms as properly wired neurons. The
scale bar represents 50 mM.
(Figure 21C) Relative to vehicle-treated animals, Example 45 Compound-treated
rats displayed significantly
lower number of cleaved caspase 3-positive cells in the dentate gyrus,
indicating that P7C3 was capable of
inhibiting apoptosis in the aged rat brain. The scale bar represents 50 mM.
(Figure 21D) Relative to vehicle-
treated animals, Example 45 Compound-treated rats were observed to maintain
stable body weight as a
function of terminal aging. In all graphs data are expressed as mean + SEM.
73
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Figure 22: Example 45 Compound Preserves Mitochondrial Membrane Potential in
Parallel to Proneurogenic
Activity U2OS cells were loaded with tetramethylrhodamine methyl ester (TMRM)
dye and then exposed to
the calcium ionophore A23187 either in the presence or absence of test
compounds. Example 45 Compound
(Figure 22A) preserved mitochondrial membrane potential following exposure to
the calcium ionophore
A23187 in a dose-dependent manner. The protective effect of P7C3 was
enantiomeric specific. The (R)-
enantiomer of another compound (Figure 22B) blocked dye release at levels as
low as 1 nM, whereas the (S)-
enantiomer (Figure 22C) failed to block dye release even at the highest drug
dose tested (100 nM). A
proneurogenic compound, P7C3A20 (Figure 22D) exhibited dye release protection
at all doses tested, yet
compounds having less proneurogenic activity (Figure 22E and Figure 22F) were
less effective in preserving
mitochondrial membrane potential at any test dose. Each compound was evaluated
in triplicate with similar
results.
Figure 23: Example 45 Compound Preserves Mitochondrial Membrane Potential in
Cultured Primary Cortical
Neurons. Cortical neurons cultures from rats on embryonic day 14 were loaded
with tetramethylrhodamine
methyl ester (TMRM) dye after 6 days of maturation. The top panels (no calcium
ionophore) show that the dye
alone did not affect the health of neurons. The remaining panels are from
cells that were exposed to the
calcium ionophore A23187 at time zero. With vehicle-alone, cortical neuron
mitochondrial membrane
potential was rapidly lost after exposure to the ionophore. Escalating doses
of Example 45 Compound (Figure
23A) preserved mitochondrial membrane potential following exposure to the
calcium ionophore A23187 in a
dose dependent manner, with full protection achieved at 1 mM. The less active
compound (Figure 23B) was
less effective in preserving mitochondrial membrane potential at any dose
tested. Results shown are
representative of 10 fields analyzed in each of 2 experimental runs for all
conditions.
Figure 24. Example 45 Compound (P7C3) Provides Therapeutic Benefit in Animal
Model of Amyotrophic
Lateral Sclerosis (ALS). Female G93A SOD1 mice (n=30 in each group, with all
mice sibling matched across
treatment groups) were treated with either vehicle or P7C3 (10 mg/kg i.p.
twice daily) starting at 40 days of
age. P7C3-treated mice showed a significant delay in disease progression, as
evidenced by the later age by
which they dropped to 10% below their maximum weight (Figure 24A). P7C3-
treated mice also attain a
neurological severity score of 2 at a later age than vehicle treated mice
(Figure 24B), again indicating that
P7C3-treatment slows disease progression. This score is determined as follows:
'0' = full extension of hind
legs away from lateral midline when the test mouse is suspended by its tail,
and can hold this for 2 seconds,
suspended 2-3 times; '1' = collapse or partial collapse of leg extension
towards lateral midline (weakness) or
trembling of hind legs during tail suspension; '2' = toes curl under at least
twice during walking of 12 inches,
or any part of foot drags along cage bottom / table; '3' = rigid paralysis or
minimal joint movement, foot not
being used for forward motion; and '4' = mouse cannot right itself within 30
seconds from either side. With
74
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
further disease progression, vehicle-treated mice show the expected decline in
retention time on the
accelerating rotarod, with retention time averaged across 4 trials (Figure
24C, open bars). P7C3-treated mice,
however, show a consistent trend towards improved performance on this task
after onset of disease (Figure
24C, filled bars), with statistically significant improvement on days 131, 138
and 145 (*, p<0.001, Student's t
Test). All graphical data shown above is mean +/- SEM, with statistical
analysis conducted using the Student's
t Test). As another means of disease progression, walking gait was evaluated.
Figure 24D shows footprint
data from two sisters (VEH and P7C3) on day 92 (before disease onset) and day
118 (after disease onset).
Front paws are dipped in red ink, and back paws are dipped in black ink. The
VEH-treated mouse shows the
expected decline in gait after disease onset on day 188, while her P7C3-
treated sister showed preservation of
normal gait on day 118. All analysis was conducted blind to treatment group.
Figure 25. Example 6a Compound (P7C3A20) Provides Therapeutic Benefit in
Animal Model of Parkinson's
Disease. Mice were treated with MPTP (30 mg/kg i.p.) or Vehicle only for 5
days and then
immunohistochemically analyzed for tyrosine hydroxylase staining (TH) 21 days
later (Figure 25A).
Treatment with MPTP and Vehicle (n=6) reduced the number of TH+ neurons in the
substantia nigra (Figure
25B) by approximately 50% (*, p=0.0002, Student's t test) relative to mice
that received Vehicle only (n=8).
MPTP-mediated cell death in the substantia nigra was significantly attenuated
(**, p=0.005) in mice that
additionally received P7C3A20 (10 mg/kg i.p. twice daily) (n=5). TH+ neurons
in the substantia nigra of
every mouse were counted blind to treatment group by two investigators using
Image J software, and results
were averaged.
Figure 26. Example 45 Compound (P7C3) Provides Therapeutic Benefit in Animal
Model of Huntington's
Disease. 40 female R6/2 mice were included in each of VEH (vehicle) and P7C3
(10 mg/kg P7C3 i.p. twice
daily) groups, and treatment was begun at 6 weeks of age. (Figure 26A)
Treatment with P7C3 statistically
significantly extends survival of R6/2 mice (p<0.001, Gehan-Breslow-Wilcoxon
test). (Figure 26B) At 14
weeks of age, P7C3-treated R6/2 mice also show statistically improved
objective measures of general
condition (lower score corresponds to better general better condition, *
p<0.0001, Student's t Test). All
measurements were conducted blind to genotype and treatment group.
Figure 27. Example 45 Compound (P7C3) Augments Hypothalamic Neurogenesis.
Administration of P7C3
for a one month period of time augments proliferation of hypothalamic neural
precursor cells (shown in red) in
the arcuate nucleus (ARC), dorsomedial hypothalamus (DMH) and ventralmedial
hypothalamus (VMH).
Micrographs shown are representative of staining from every third section
throughout the hypothalamus in 4-6
mice for each treatment group.
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Figure 28. Neuroprotective efficacy of P7C3, P7C3A20 and Dimebon for newborn
neurons in the adult
hippocampus. Test compounds were evaluated by dose response assay for their
ability to block normal
apoptotic cell death of newborn neural precursor cells in the adult dentate
gyrus. P7C3A20 exhibits the
greatest potency and ceiling of efficacy, and Dimebon the least. P7C3 is
intermediate in both measures. 6
animals were tested per group. Dosing is expressed as total mg / day, and
compounds were administered
intraperitoneally in divided doses twice daily. Data are expressed as mean
SEM. Values for P7C3 and
P7C3A20 were compared to those of vehicle (VEH), and values for Dimebon were
compared to those of saline
(SAL).
Figure 29. Neuroprotective efficacy of P7C3 and P7C3A20 from MPTP-toxicity to
SNc dopaminergic
neurons. Figure 29A- Test compounds were evaluated by dose response assay for
their ability to block
MPTP-neurotoxicity in the SNc. P7C3A20 showed greater potency and CoE than
P7C3, and Dimebon
showed no protective efficacy. 15 animals were tested per group. VEH group
contained 30 animals: 15
animals that received the P7C3A20/P7C3 vehicle, and 15 animals that received
Dimebon vehicle (saline).
These 2 control groups did not differ in number of surviving TH+ neurons, and
were thus combined.
Representative immunohistochemical pictures of TH-staining in the SNc are
shown below the graph. Dosing
is expressed as total mg / day, and was administered intraperitoneally in
divided doses twice daily. Data are
expressed as mean SEM. Figure 29B- Representative images of TH-staining from
the striata of individual
animals demonstrate that three weeks after a five day course of daily MPTP
administration both P7C3 and
P7C3A20 block the loss of dopaminergic axon terminals. P7C3A20 does so with
greater effect, and Dimebon
offers no protection. Striatal sections were obtained from the same mice used
in Figure 2, and compound
treatment groups are from mice that received a dose of 20 mg/kg/d.
Figure 30. Brain and blood levels of P7C3, P7C3A20 and Dimebon three weeks
after MPTP administration.
Relative neuroprotective activity within a test compound correlated with brain
levels of that compound, and
brain levels correlated with blood levels of the test compounds. Only about
one-tenth the amount of P7C3A20
accumulated in the brain compared to P7C3. Brain accumulation of Dimebon was
equivalent to P7C3. Data
are expressed as mean SEM. Three animals were tested per group.
Figure 31. Neuroprotective efficacy of P7C3 and P7C3A20 for MPP toxicity to
dopaminergic neurons in C.
elegans. Worms were treated with 5 mM MPP for 40 hrs, with pre-incubation for
30 minutes with different
concentrations of test compounds or vehicle. VEH animals not exposed to MPP
exhibited normal GFP
expression in dopaminergic neurons (filled arrow). By contrast, GFP expression
was lost after 40 hrs of MPP
exposure (unfilled arrow). Both P7C3A20 and P7C3 dose-dependently protected
dopaminergic neurons from
76
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
MPP toxicity, with P7C3A20 exhibiting greater potency and ceiling of
efficacy. 20 worms were analyzed per
group, and each group was performed in triplicate. Data are expressed as mean
SEM.
Figure 32. Protective efficacy of P7C3 and P7C3A20 for MPP -induced mobility
deficits in C. elegans. The
top panels show worms with the head identified by a green dot. The second row
of panels shows the path
taken by each worm in 10 seconds, determined by tracking the green dot.
Tracking is visualized as starting
with blue color and progressing to white by the completion of 10 seconds. The
green dot was used to
determine locomotion, defined as the distance traveled by the head of the worm
in 10 seconds divided by body
length. Scale bars represent 7011M. Quantitative analysis of locomotion showed
that untreated VEH controls
had a value of 16.2 0.49 (n=30). When worms were treated with MPP ,
locomotion was reduced more than
50% (7.2 0.68; n=31, p<0.0001). 10 [LM P7C3A20 protected mobility to almost
80% of normal (12.8 0.81;
n=34, *p<0.01), and 10 [LM P7C3 protected almost 60% (m.i. of 9.6 0.72;
n=28, *p<0.05). 10 [LM Dimebon
did not offer any protection. (7.7 1.0; n=30). Experiments were performed in
triplicate and data are
expressed as mean SEM.
Figure 33. Efficacy of new P7C3 analogs in the in vivo hippocampal
neurogenesis assay correlates with
activity in the in vivo MPTP-neuroprotection assay. Figure 33A- P7C3-57
differs from P7C3 by replacing the
aniline NH with sulfide linker. P7C3-58 differs from P7C3 by replacing the
aniline phenyl ring with a
pyrimidine. P7C3-525 differs from P7C3 by replacing the aniline moiety with a
dimethyl pyrazole. P7C3-540
and S41 differ from P7C3 by replacing the aniline NH with and oxygen linker,
and they are R and S single
enantiomers, respectively. P7C3-554 differs from P7C3 mainly by the addition
of a methyl group to the
central carbon of the propyl linker and an OMe group on the aniline. P7C3-S165
differs from P7C3 by
replacing the aniline and carbinol fragments with a carboxylic acid. P7C3-5184
differs from P7C3 by
replacing the bromines on the carbazole with chlorines and replacing the
aniline with a naphthyl amine.
Figure 33B- New analogs of P7C3 were subjected to in vivo testing in the
hippocampal neurogenesis (4 mice
each) and MPTP-protection (10 mice each) assays. Results show that activity in
these two assays correlates,
such that the in vivo neurogenesis screen is useful for predicting
neuroprotective efficacy of P7C3 analogs for
dopaminergic neurons in the substantia nigra. LC/MS/MS assay of blood and
brain levels of all compounds
administered a single time to C57BL/6 mice at 10 mg/kg (i.p.) indicates that
they cross the blood brain barrier
following intraperitoneal administration.
Figure 34. P7C3A20 and P7C3 block motor neuron cell death in the spinal cord
when administered at the
time of disease onset to G93A-SOD1 mutant mice. Treatment of G93A-SOD1 mutant
mice with 20 mg/kg/d
of P7C3A20, P7C3 or Dimebon, or the appropriate vehicle, was initiated on day
80. Five mice from each
group were sacrificed on days 90, 100, 110 and 120. The number of spinal cord
motor neurons per cubic
77
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
millimeter of lumbar spinal cord was determined through immunohistochemical
staining for ChAT and
quantification with NIH Image J software. All images were analyzed blind to
treatment group. Spinal cord
motor neurons in G93A-SOD1 mutant mice died over time as expected. Spinal cord
motor neuron cell death
was blocked by administration of P7C3A20. P7C3 was intermediately protective,
while Dimebon had no
neuroprotective efficacy. (Figure 34A) Representative staining of ChAT of one
ventral horn from each of the
5 mice in each treatment group at 110 days. (Figure 34B) Graph representation.
Figure 35. P7C3A20 preserves performance in the accelerating rotarod test when
administered at the time of
disease onset to G93A-SOD1 mutant mice. Treatment of G93A-SOD1 mutant mice
with 20 mg/kg/d of
P7C3A20, P7C3 or Dimebon, or the appropriate vehicle, was initiated on day 80,
with 20 mice per group. All
compounds were administered at 20 mg/kg/day i.p. in divided doses. Each
compound-treated mouse had a
sex-matched sibling that received vehicle. Only sibling pairs were analyzed at
each time point. By week 16,
there were 13 compound-vehicle pairs remaining in each group. All vehicle
treated mice showed the expected
decline in retention time on the accelerating rotarod over time, and P7C3 and
Dimebon groups showed no
difference in retention time compared to their vehicle groups. Mice treated
with P7C3A20 showed
significantly higher retention time on the rotarod at weeks 13, 14, 15 and 16.
All testing and analysis was
performed blind to treatment group.
Figure 36. P7C3A20 preserves walking gait when administered at the time of
disease onset to G93A-SOD1
mutant mice. Figure 36A- The schematic diagram shows parameters used to
measure gait. Front and back
stride were collected as a straight line from back paw print to the following
paw print. Back-front distance
was collected as a straight line from back paw print to the corresponding
front paw print. 20 measurements
(10 on each side) for each parameter were measured per mouse, and 20 mice per
group were evaluated at 90
and 118 day time points. All measurements were conducted blind to treatment
group, and the student's t test
was used for statistical comparison of a treatment group to its matched
vehicle group. Figure 36B- At 90
days, there were no differences between any groups in back stride, front
stride and back-front distance. By
day 118, all vehicle groups, and P7C3- and Dimebon-treated mice, showed a
significant difference in these
measures, reflecting disease progression. Back stride and front stride were
preserved to near-normal levels in
P7C3A20 treated mice on day 118. By day 132, P7C3 and Dimebon treated mice
were too sick to participate
in the task. At this time point, P7C3A20 treated mice continued to show
normalized back stride and front
stride levels.
Figure 37. Plasma, brain and spinal cord levels of P7C3A20, P7C3 and Dimebon.
Five mice for each
compound group were treated for 21 days with 20 mg/kg/day of the compound,
starting on day 85. Blood,
78
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
brain and spinal cord were harvested six hours after the last injection and
compound levels were measured by
LC/MS/MS. Concentrations are presented as mean SEM.
DETAILED DESCRIPTION
The presently disclosed embodiments relate generally to stimulating
neurogenesis (e.g., post-natal
neurogenesis, e.g., post-natal hippocampal and/or hypothalamic neurogenesis)
and/or promoting the survival
of existing neurons by reducing neuronal cell death.
COMPOUNDS
In one aspect, the presently disclosed embodiments feature compounds having
general formula (I):
R4
R'
R3
R2 101 C3
/ 2
N%C ¨R
\
L ¨A
R1
2
z
1 0
(I)
Here and throughout this specification, RI, R2, R3, R4, ¨
K R', LI, L2, A, and Z can be as defined
anywhere herein.
It is appreciated that certain features of the presently disclosed
embodiments, which are, for clarity,
described in the context of separate embodiments, can also be provided in
combination in a single
embodiment. Conversely, various features of the presently disclosed
embodiments which are, for brevity,
described in the context of a single embodiment, can also be provided
separately or in any suitable sub-
combination.
Thus, for ease of exposition, it is also understood that where in this
specification, a variable (e.g., RI)
is defined by "as defined anywhere herein" (or the like), the definitions for
that particular variable include the
first occurring and broadest generic definition as well as any sub-generic and
specific definitions delineated
anywhere in this specification.
Variables R1, R2, R3, R4
In some embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g., R3)
is selected from halo,
hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6 thioalkoxy, C1-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl,
79
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
C1-C6 haloalkyl, cyano, -NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6
alkyl), and nitro; and the
others are hydrogen.
In certain embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g.,
R3) is selected from halo,
C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, cyano, and
nitro; and the others are hydrogen.
In certain embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g.,
R3) is selected from halo,
C1-C6 alkyl, and C1-C6 haloalkyl; and the others are hydrogen.
In certain embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g.,
R3) is selected from halo
and C1-C6 alkyl; and the others are hydrogen.
In certain embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g.,
R3) is halo (e.g., bromo or
chloro) and C1-C6 alkyl; and the others are hydrogen.
In certain embodiments, one or two of RI, R2, R3, and R4 (e.g., one of, e.g.,
R3) is bromo; and the
others are hydrogen.
In some embodiments, R3 is selected from halo, hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, cyano, -
NH2, -NH(C1-C6 alkyl), N(C1-
C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; and each of RI, R2, and R4 can be
as defined anywhere herein.
In certain embodiments, R3 is selected from halo, hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, -NH2, -NH(C1-C6
alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; and each of RI, R2,
and R4 is hydrogen.
In some embodiments, R3 is selected from halo, C1-C6 alkoxy, C1-C6 haloalkoxy,
Ci-C6 alkyl, Ci-C6
haloalkyl, cyano, and nitro; and each of RI, R2, and R4 can be as defined
anywhere herein.
In certain embodiments, R3 is selected from halo, Ci-C6 alkoxy, C1-C6
haloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, and nitro; and each of RI, R2, and R4 is hydrogen.
In some embodiments, R3 is selected from halo, Ci-C6 alkyl, and Ci-C6
haloalkyl; and each of RI, R2,
and R4 can be as defined anywhere herein.
In certain embodiments, R3 is selected from halo, C1-C6 alkyl, and Ci-C6
haloalkyl; and each of RI, R2,
and R4 is hydrogen.
In some embodiments, R3 is selected from halo and Ci-C6 alkyl; and each of RI,
R2, and R4 can be as
defined anywhere herein.
In certain embodiments, R3 is selected from halo and C1-C6 alkyl; and each of
RI, R2, and R4 is
hydrogen.
In some embodiments, R3 is halo (e.g., bromo or chloro); and each of RI, R2,
and R4 can be as defined
anywhere herein..
In certain embodiments, R3 is halo (e.g., bromo or chloro); and each of RI,
R2, and R4 is hydrogen.
In some embodiments, R3 is bromo; and each of RI, R2, and R4 can be as defined
anywhere herein..
In certain embodiments, R3 is bromo; and each of RI, R2, and R4 is hydrogen.
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In some embodiments, each of RI, R2, R3, and R4 is independently selected from
hydrogen, halo, and
CI-C6 alkyl.
In certain embodiments, each of RI, R2, R3, and R4 is independently selected
from hydrogen and
halo(e.g., bromo or chloro).
In some embodiments, each of RI, R2, R3, and R4 is hydrogen.
In some embodiments, when any one or more of RI, R2, R3, and R4 can be a
substituent other than
hydrogen, said substituent, or each of said substituents, is other than Ci-C6
alkyl (e.g., other than C1-C3 alkyl,
e.g., other than CH3).
Variable Ll
In some embodiments, LI is C1-C3 (e.g., C1-C2) straight chain alkylene, which
is optionally substituted
with from 1-2 independently selected Re.
In certain embodiments, LI is methylene (i.e., -CH2-). In other embodiments,
LI is methylene that is
substituted with 1 or 2 (e.g., 1) independently selected R. In embodiments, Re
is C1-C6 alkyl (e.g., C1-C3
alkyl, e.g., CH3).
In certain embodiments, LI is ethylene (i.e., -CH2CH2-). In other embodiments,
LI is ethylene that is
substituted with 1 or 2 (e.g., 1) independently selected R. In embodiments, Re
is C1-C6 alkyl (e.g., C1-C3
alkyl, e.g., CH3).
Variable L2
In some embodiments, L2 is C1-C3 (e.g., C1-C2) straight chain alkylene, which
is optionally substituted
with from 1-2 independently selected R.
In certain embodiments, L2 is methylene (i.e., -CH2-). In other embodiments,
LI is methylene that is
substituted with 1 or 2 (e.g., 1) independently selected R. In embodiments, Re
is C1-C6 alkyl (e.g., C1-C3
alkyl, e.g., CH3). In embodiments, Re is C1-C6 alkoxy, C1-C6 thioalkoxy, C1-C6
haloalkoxy, or C1-C6
thiohaloalkoxy. For example, Re can be C1-C6 (e.g., C1-C3) thioalkoxy, such as
-SCH3.
In certain embodiments, L2 is ethylene (i.e., -CH2CH2-). In other embodiments,
L2 is ethylene that is
substituted with 1 or 2 (e.g., 1) independently selected R. For example, the
ethylene carbon more proximal to
Z in formula (I) can be substituted as described in the preceding paragraph.
In certain embodiments, L2 is a bond that directly connects A in formula (I)
to Z in formula (I).
Non-Limiting Combinations of Variables Ll and L2
In some embodiments, each of LI and L2 is, independently, C1-C3 alkylene,
which is optionally
substituted with from 1-2 independently selected R.
In certain embodiments, each of LI and L2 is CH2.
In certain embodiments, one of LI and L2 is CH2 (e.g., LI), and the other
(e.g., L2) is methylene that is
substituted with 1 or 2 (e.g., 1) independently selected Re, in which Re can
be as defined anywhere herein.
81
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, each of L1 and L2 is methylene that is substituted
with 1 or 2 (e.g., 1)
independently selected Re, in which Re can be as defined anywhere herein.
In some embodiments, L1 is C1-C3 (e.g., C1-C2) straight chain alkylene, which
is optionally substituted
with from 1-2 independently selected Re, and L2 is a bond that directly
connects A in formula (I) to Z in
formula (I). In embodiments, L1 can be, for example, methylene (i.e., -CH2-)
or methylene that is substituted
with 1 or 2 (e.g., 1) independently selected Re (e.g., Ci-C6 alkyl, e.g., Ci-
C3 alkyl, e.g., CH3).
Variable A
[I] In some embodiments, A is:
(i) CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen,
halo, C1-C3 alkyl, or OR9; or
(ii) C=0; or
(iv) heterocycloalkylene containing from 3-5 ring atoms, wherein from 1-2 of
the ring
atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and
wherein said
heterocycloalkylene is (a) substituted with 1 oxo; and (b) is optionally
further substituted with
from 1-4 independently selected R.
In some embodiments, A is CRA1RA2, in which each of RA1 and RA2 is,
independently, hydrogen, halo,
C1-C3 alkyl, or OR9 (e.g., hydrogen, halo, or OR9).
In certain embodiments, A can be CRA1RA2, in which each of RA1 and RA2 is,
independently, hydrogen,
halo, or C1-C3 alkyl.
In certain embodiments, A can be CRA1RA2, in which one of RA1 and RA2 is halo
(e.g., fluoro), and the
other of RA1 and RA2 is, independently, hydrogen, halo, or Ci-C3 alkyl (e.g.,
hydrogen).
In certain embodiments, one of RA1 and RA2 is hydrogen. In embodiments, one of
RA1 and RA2 is halo
or OR9, and the other is hydrogen.
In certain embodiments, one of RA1 and RA2 can be OR9. In embodiments, the
other of RA1 and RA2
can be as defined anywhere herein; e.g., the other of RA1 and RA2 can be
hydrogen or Ci-C3 alkyl. For
example, one of RA1 and RA2 can be OR9, and the other of RA1 and RA2 is
hydrogen. In embodiments, R9 can
be hydrogen or R9 can be Ci-C3 alkyl (e.g., CH3).
In certain embodiments, one of RA1 and RA2 can be halo. In embodiments, the
other of RA1 and RA2
can be as defined anywhere herein; e.g., the other of RA1 and RA2 can be
hydrogen, C1-C3 alkyl, or halo. For
example, one of RA1 and RA2 can be halo (e.g., fluoro), and the other of RA1
and RA2 is hydrogen.
In embodiments, one of RA1 and RA2 is halo or OR9, and the other is hydrogen.
For example, one of RA1 and RA2 can be OR9, and the other is hydrogen. In
embodiments, R9 can be
hydrogen. R9 can be C1-C3 alkyl (e.g., CH3).
As another example, one of RA1 and RA2 can be halo (e.g., fluoro), and the
other is hydrogen.
In other embodiments, each of RA1 and RA2 is a substituent other than
hydrogen.
82
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
For example, each of RA1 and RA2 can be halo (e.g., fluoro).
As another example, one of RA1 and RA2 can be OR9 (e.g., in which R9 is
hydrogen), and the other is
Ci-C3 alkyl (e.g., CH3).
As a further example, each of RA1 and RA2 can be Ci-C3 alkyl (e.g., CH3).
In still other embodiments, each of RA1 and RA2 is hydrogen.
Embodiments can further include any one or more of the following features.
When the carbon attached to RA1 and RA2 is substituted with four different
substituents, the carbon
attached to RA1 and RA2 can have the R configuration.
When the carbon attached to RA1 and RA2 is substituted with four different
substituents, the carbon
attached to RA1 and RA2 can have the S configuration.
III] In some embodiments, A is C=0.
RI] In some embodiments, A is heterocycloalkylene containing from 3-
5 ring atoms, in which
from 1-2 of the ring atoms is independently selected from N, NH, N(C1-C3
alkyl), 0, and S; and wherein said
heterocycloalkylene is (a) substituted with 1 oxo (e.g., 1 oxo on a ring
carbon); and (b) is optionally further
substituted with from 1-4 independently selected R.
In certain embodiments, A is heterocycloalkylene containing 5 ring atoms, in
which from 1-2 of the
ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and
wherein said
heterocycloalkylene is (a) substituted with 1 oxo; and (b) is optionally
further substituted with from 1-4
independently selected R. For example, A can be:
N--
0
Non-Limiting Combinations of Variables Ll, L2, and A
In some embodiments:
A is (i) CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen, halo, C1-C3
alkyl, or OR9; or (ii) C=0; and
each of L1 and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re.
In some embodiments:
A is CRA1RA2, wherein each of RA1 and RA2 is independently selected from
hydrogen, halo, Ci-C3
alkyl, or OR9; and
each of L1 and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re.
83
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Embodiments can include one or more of the following features
Each of RA1 and RA2 can be as defined anywhere herein.
Each of L1 and L2 is CH2.
One of L1 and L2 is CH2 (e.g., L1), and the other (e.g., L2) is methylene that
is substituted with 1 or 2
(e.g., 1) independently selected Re, in which Re can be as defined anywhere
herein. For example:
= Li can be CH2; and
= One of RA1 and RA2 is hydrogen; and
= L2 can be methylene that is substituted with 1 or 2 (e.g., 1)
independently selected Re (e.g., C1-
C6 (e.g., C1-C3) alkyl, such as CH3; or Ci-C6 (e.g., C1-C3) thioalkoxy, such
as -SCH3);
Each of L1 and L2 is methylene that is substituted with 1 or 2 (e.g., 1)
independently selected Re, in
which Re can be as defined anywhere herein. For example:
= each of RA1 and RA2 can be a substituent other than hydrogen (e.g., one
of which is CH3), and
= each of L1 and L2 is methylene that is substituted with C1-C3 alkyl, such
as CH3).
In some embodiments:
A is heterocycloalkylene containing from 3-5 (e.g., 5) ring atoms, in which
from 1-2 of the ring atoms
is independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein
said heterocycloalkylene is (a)
substituted with 1 oxo; and (b) is optionally further substituted with from 1-
4 independently selected le; and
L1 is C1-C3 (e.g., C1-C2) straight chain alkylene, which is optionally
substituted with from 1-2
independently selected Re, and
L2 is a bond that directly connects A in formula (I) to Z in formula (I).
Variable Z
II] In some embodiments, Z is:
(i) -NR10R11; or
(ii) -C(0)NR10R11; or
(iii) -0R12; or
(iv) -S(0)õR13, wherein n is 0, 1, or 2; or
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NHC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected le;
(vi) C6-Cio aryl that is optionally substituted with from 1-4 independently
selected Rb; or
(vii) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 independently selected Rb.
In certain embodiments, Z is as defined in (i), (iii), (iv), (v), (vi), or
(vii) in the preceding paragraph.
In certain embodiments, Z is as defined in (i), (iii), (iv), (v), or (vii) in
the preceding paragraph.
84
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, Z is as defined in (i), (iii), (v), or (vii) in the
preceding paragraph.
In certain embodiments, Z is as defined in (i), (iii), or (iv) in the
preceding paragraph.
In certain embodiments, Z is:
(i) -NR10R11; or
(iii) -0R12; or
(v) heterocycloalkenyl containing from 5-6 ring atoms, wherein from 1-3 of the
ring atoms is
independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected R.
In certain embodiments, Z is: (i) -NR10R11; or (iii) -0R12.
In certain embodiments, Z is: (i) -NR10R11; or (iv) -S(0)õR13, wherein n is 0,
1, or 2.
In certain embodiments, Z is: (iii) -0R12; or (iv) -S(0)õR13, wherein n is 0,
1, or 2.
In certain embodiments, Z does not include heterocyclyl (e.g., a nitrogenous
heterocyclyl, e.g.,
piperazinyl or piperidinyl) as part of its structure (e.g., as a fused ring or
attached to another ring by a bond).
In certain embodiments, Z is other than heterocycloalkenyl containing from 5-6
ring atoms, wherein
from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6
alkyl), NC(0)(C1-C6 alkyl), 0, and
S; and wherein said heterocycloalkenyl is optionally substituted with from 1-4
independently selected R.
In certain embodiments, Z is other than heteroaryl containing from 5-14 ring
atoms, wherein from 1-6
of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; and wherein said heteroaryl
is optionally substituted with from 1-4 independently selected Rb (e.g., other
than pyridyl).
[II] In some embodiments, Z is -NR10R11.
[A] In some embodiments, one of R1 and R11 is hydrogen, and the other of
R1 and R11 is a
substituent other than hydrogen.
In some embodiments, one of R1 and R11 is hydrogen or a substituent other
than hydrogen, and the
other of R1 and R11 is a substituent other than hydrogen.
In some embodiments, each of R1 and R11 is a substituent other than hydrogen.
In some embodiments, each of R1 and R11 is hydrogen.
[B] In some embodiments, one of R1 and R11 is independently selected from
the substituents
delineated collectively in (b), (c), (g) through (k), and (1) below:
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is
independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said
heteroaryl is
optionally substituted with from 1-4 Rb;
(g) C8-C arylcycloalkyl, wherein:
(1) the aryl portion is optionally substituted with from 1-4 independently
selected Rb,
and
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(h) arylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) the aryl portion from is optionally substituted with from 1-4
independently
selected Rb, and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(i) heteroarylheterocyclyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) from 1-2 of the ring atoms of the heterocyclyl portion is independently
selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclyl
portion is optionally substituted with from 1-3 independently selected Ra;
(j) heteroarylcycloalkyl containing from 8-14 ring atoms, wherein:
(1) from 1-2 of the ring atoms of the heteroaryl portion is independently
selected from
N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl portion is
optionally substituted
with from 1-3 independently selected Rb; and
(2) the cycloalkyl portion is optionally substituted with from 1-4
independently
selected Ra;
(k) C3-C8 cycloalkyl or C3-C8 cycloalkenyl, each of which is optionally
substituted with from
1-4 independently selected Ra; and
(1) C7-C12 aralkyl, wherein the aryl portion is optionally the aryl portion
from is optionally
substituted with from 1-4 independently selected Rb,
and the other of RI and R11 can be as defined anywhere herein.
In some embodiments, RI and R11 cannot be C3-C8 cycloalkyl or C3-C8
cycloalkenyl, each of which is
optionally substituted with from 1-4 independently selected R.
In some embodiments, one of RI and R11 is independently selected from the
substituents delineated
collectively in (b), (c), (g) through (j), and (1) above; and the other of RI
and R11 can be as defined anywhere
herein.
In some embodiments, one of RI and R11 is independently selected from the
substituents delineated
collectively in (b), (c), and (g) through (j); and the other of RI and R11
can be as defined anywhere herein.
In some embodiments, one of RI and R11 is independently selected from:
(b) C6-C10 aryl that is optionally substituted with from 1-4 Rb;
86
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(c) heteroaryl containing from 5-14 ring atoms, wherein from 1-6 of the ring
atoms is independently
selected from N, NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl is
optionally substituted with from
1-4 Rb;
and the other of RI and RII can be as defined anywhere herein.
In some embodiments, one of RI and RII is C6-C10 aryl (e.g., C6) that is
optionally substituted with
from 1-4 (e.g., 1-3, 1-2, or 1) Rb; and the other of RI and RII can be as
defined anywhere herein.
In certain embodiments, Rb at each occurrence is independently selected from
halo; or CI-C6 alkoxy;
C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; CI-C6 alkyl, CI-C6
haloalkyl, -NH(C1-C6 alkyl),
N(C1-C6 alky1)2, and -NHC(0)(C1-C6 alkyl), each of which is optionally
substituted with from 1-3
independently selected Re.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy; C1-C6
haloalkoxy; C1-C6 thioalkoxy; and C1-C6 thiohaloalkoxy, each of which is
optionally substituted with from 1-3
independently selected R. In embodiments, Rb can further include halo.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy and C1-C6
haloalkoxy, each of which is optionally substituted with from 1-3
independently selected R. In embodiments,
Rb can further include halo.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy, each of
which is optionally substituted with from 1-3 independently selected R. In
embodiments, Rb is C1-C6 alkoxy
(e.g., OCH3). In embodiments, Rb can further include halo.
In certain embodiments, one of RI and RII is unsubstituted phenyl, and the
other of RI and RII can be
as defined anywhere herein.
In certain embodiments, one of RI and RII is phenyl that is substituted with
1 Rb, and the other of RI
and RII can be as defined anywhere herein. Rb can be as defined anywhere
herein (e.g., Rb can be C1-C6
alkoxy, e.g., OCH3). For example, one of RI and RII can be 3-methoxyphenyl.
In embodiments, Rb can
further include halo.
[C] In some embodiments, when one of RI and RII is independently
selected from the
substituents delineated collectively in (b), (c), (g) through (k), and (1)
above, the other of RI and RII can be:
(a) hydrogen; or
(d) C1-C6 alkyl or C1-C6 haloalkyl (e.g., C1-C6 alkyl), each of which is
optionally substituted with from
1-3 Rd; or
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl); or
(0 C2-C6 alkenyl or C2-C6 alkynyl.
In certain embodiments, the other of RI and RII is:
(a) hydrogen; or
87
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(d) C1-C6 alkyl or C1-C6 haloalkyl (e.g., C1-C6 alkyl), each of which is
optionally substituted with from
1-3 Rd; or
(e) -C(0)(C1-C6 alkyl), -C(0)(C1-C6 haloalkyl), or -C(0)0(C1-C6 alkyl).
In certain embodiments, the other of RI and RII is:
(a) hydrogen; or
(d) C1-C6 alkyl or C1-C6 haloalkyl (e.g., C1-C6 alkyl), each of which is
optionally substituted with from
1-3 Rd; or
(e) -C(0)(C1-C6 alkyl), or-C(0)(Ci-C6 haloalkyl).
In certain embodiments, the other of RI and RII can be:
(a) hydrogen; or
(d) C1-C6 alkyl (e.g., C1-C3 alkyl, e.g., CH3), which is optionally
substituted with from 1-3 Rd; or
(e) -C(0)(C1-C6 alkyl), e.g., C1-C3 alkyl, e.g., CH3.
In certain embodiments, the other of RI and RII can be:
(a) hydrogen; or
(d) C1-C6 alkyl (e.g., C1-C3 alkyl, e.g., CH3), which is optionally
substituted with from 1-3 Rd.
In certain embodiments, the other of RI and RII can be hydrogen.
In certain embodiments, the other of RI and RII can be (d) or (e) or any
subset thereof
[E] In some embodiments, one of RI and RII is C6-C10 (e.g., C6) aryl that
is optionally substituted
with from 1-4 Rb, and the other is hydrogen or C1-C6 alkyl (e.g., C1-C3 alkyl,
e.g., CH3).
In some embodiments, one of RI and RII is C6-Cio (e.g., C6) aryl that is
optionally substituted with
from 1-4 Rb, and the other is hydrogen.
In certain embodiments, one of RI and RII is unsubstituted phenyl, and the
other is hydrogen.
In certain embodiments, one of RI and RII is phenyl that is substituted with
1 Rb, and the other is
hydrogen. In embodiments, Rb is C1-C6 alkoxy (e.g., C1-C3 alkoxy, e.g., OCH3).
For example, one of RI and
RII is 3-methoxyphenyl, and the other is hydrogen.
[F] In some embodiments, each of RI and RII cannot be optionally
substituted naphthyl (e.g.,
each of RI and RII cannot be unsubstituted naphthyl). In embodiments, each of
RI and RII is other than
optionally substituted naphthyl (e.g., unsubstituted naphthyl) when R and R'
are defined according to
definitions (1), (2), and (4); and A is CRAIRA2 (e.g., CHOR9, e.g., CHOH), and
each of LI and L2 is C1-C3
alkylene (e.g., each of LI and L2 is CH2).
[G] In some embodiments, one of RI and RII is hydrogen, and the other is
heteroaryl containing
from 5-14 ring atoms, wherein from 1-6 of the ring atoms is independently
selected from N, NH, N(C1-C3
alkyl), 0, and S; and wherein said heteroaryl is optionally substituted with
from 1-4 Rb.
88
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, one of RI and RII is hydrogen, and the other is
heteroaryl containing from 5-
6 ring atoms, wherein from 1-2 of the ring atoms is independently selected
from N, NH, N(C1-C3 alkyl), 0,
and S; and wherein said heteroaryl is optionally substituted with from 1-2 Rb.
RI] In some embodiments, Z is -0R12.
In some embodiments, RI2 is C1-C6 alkyl or C1-C6 haloalkyl, each of which is
optionally substituted
with from 1-3 Re.
In some embodiments, RI2 is C1-C6 alkyl, which is optionally substituted with
from 1-3 R.
In certain embodiments, RI2 is Ci-C6 alkyl (e.g., C1-C3 alkyl, e.g., CH3).
In certain embodiments, RI2 is C1-C6 alkyl (e.g., C1-C3 alkyl, e.g., CH3),
which is optionally
substituted with from 1-3 (e.g., 1 or 2, e.g., 1) R. In embodiments, each
occurrence of Re can be
independently selected from -NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, and -
NHC(0)(C1-C6 alkyl).
In some embodiments, RI2 is C6-C10 aryl that is optionally substituted with
from 1-4 (e.g., 1-3, 1-2, or
1) Rb.
In certain embodiments, Rb at each occurrence is independently selected from
halo; or C1-C6 alkoxy;
C1 -C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl, C1-C6
haloalkyl, -NH(C1-C6 alkyl),
N(C1-C6 alky1)2, and -NHC(0)(C1-C6 alkyl), each of which is optionally
substituted with from 1-3
independently selected R.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy; C1-C6
haloalkoxy; C1-C6 thioalkoxy; and C1-C6 thiohaloalkoxy, each of which is
optionally substituted with from 1-3
independently selected R.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy and C1-C6
haloalkoxy, each of which is optionally substituted with from 1-3
independently selected R.
In certain embodiments, Rb at each occurrence is independently selected from
C1-C6 alkoxy, each of
which is optionally substituted with from 1-3 independently selected R. In
embodiments, Rb is C1-C6 alkoxy
(e.g., OCH3).
In embodiments, Rb can further include halo.
In certain embodiments, RI2 is unsubstituted phenyl.
In certain embodiments, RI2 is phenyl that is substituted with 1 Rb. Rb can be
as defined anywhere
herein (e.g., Rb can be C1-C6 alkoxy, e.g., OCH3). For example, RI2 can be 3-
methoxyphenyl.
[W] In some embodiments, Z is -S(0)õR13, in which n can be 0, 1, or 2.
In some embodiments, RI3 is C6-C10 aryl that is optionally substituted with
from 1-4 (e.g., 1-3, 1-2, or
1) Rb.
In certain embodiments, Rb at each occurrence is independently selected from
halo; or C1-C6 alkoxy;
C1-C6 haloalkoxy; C1-C6 thioalkoxy; C1-C6 thiohaloalkoxy; C1-C6 alkyl, C1-C6
haloalkyl, -NH(C1-C6 alkyl),
89
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
N(C1-C6 alky1)2, and -NHC(0)(C1-C6 alkyl), each of which is optionally
substituted with from 1-3
independently selected Re.
In certain embodiments, Rb at each occurrence is independently selected from
Ci-C6 alkoxy; C1-C6
haloalkoxy; C1-C6 thioalkoxy; and C1-C6 thiohaloalkoxy, each of which is
optionally substituted with from 1-3
independently selected R.
In certain embodiments, Rb at each occurrence is independently selected from
Ci-C6 alkoxy and C1-C6
haloalkoxy, each of which is optionally substituted with from 1-3
independently selected R.
In certain embodiments, Rb at each occurrence is independently selected from
Ci-C6 alkoxy, each of
which is optionally substituted with from 1-3 independently selected R. In
embodiments, Rb is Ci-C6 alkoxy
(e.g., OCH3).
In embodiments, Rb can further include halo.
In certain embodiments, R13 is unsubstituted phenyl.
In certain embodiments, R13 is phenyl that is substituted with 1 Rb. Rb can be
as defined anywhere
herein (e.g., Rb can be C1-C6 alkoxy, e.g., OCH3). For example, R13 can be 3-
methoxyphenyl.
In embodiments, R12 and/or R13 cannot be substituted phenyl. In embodiments,
R12 and/or R13 cannot
be substituted phenyl when R and R' are defined according to definition (1);
and A is CRA1RA2 (e.g., CHOR9,
e.g., CHOH), and each of L1 and L2 is C1-C3 alkylene (e.g., each of L1 and L2
is CH2).
[V] In some embodiments, Z is heterocycloalkenyl containing from 5-
6 ring atoms, wherein from
1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl),
NC(0)(C1-C6 alkyl), 0, and S;
and wherein said heterocycloalkenyl is optionally substituted with from 1-4
independently selected R.
In certain embodiments, Z is heterocycloalkenyl containing 6 ring atoms,
wherein from 1-3 of the ring
atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6
alkyl), 0, and S; and wherein said
heterocycloalkenyl is optionally substituted with from 1-4 independently
selected R.
In certain embodiments, from 1-3 of the ring atoms is independently selected
from N, NH, N(C1-C6
alkyl), and NC(0)(C1-C6 alkyl).
In certain embodiments, le at each occurrence is, independently selected from
oxo, thioxo, =NH, and
=N(C1-C6 alkyl), e.g., =NH.
For example, Z can be:
NH
)sCs'N
1
[V]
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
In some embodiments, Z is heteroaryl containing from 5-14 ring atoms, wherein
from 1-6 of the ring
atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S; and
wherein said heteroaryl is
optionally substituted with from 1-4 Rb.
In certain embodiments, Z is heteroaryl containing from 5-10 ring atoms,
wherein from 1-4 of the ring
atoms is independently selected from N, NH, and N(C1-C3 alkyl); and wherein
said heteroaryl is optionally
substituted with from 1-2 Rb.
Variables R and R'
II]
In some embodiments, R and R' together with C2 and C3, respectively, form a
fused phenyl
ring having formula (II):
R6
R7
1
..--µ....---.C3 .....õ,"...............
.......,
C2 R8
I
µrtrtnf
I
(II)
in which each of R5, R6, R7, and R8 is independently selected from hydrogen,
halo, hydroxyl,
sulfhydryl, Ci-C6 alkoxy, Ci-C6 thioalkoxy, Ci-C6haloalkoxy, C1-C6
halothioalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, -NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6
alkyl), and nitro.
For purposes of clarification, it is understood that compounds in which R and
R' together with C2 and
C3, respectively, form a fused phenyl ring having formula (II) correspond to
compounds having the following
general formula:
R6
R5
R4
R3 111 R7
R2 1 N R8
\
R1 \ 2
L----...z
(III)
in which RI, R2, R3, R4, LI, L2, A, and Z can be as defined anywhere herein.
91
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In some embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g., R6)
is selected from halo,
hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6 thioalkoxy, CI-C6 haloalkoxy, C1-C6
thiohaloalkoxy, C1-C6 alkyl,
CI-C6 haloalkyl, cyano, -NH2, -NH(C1-C6 alkyl), N(C1-C6 alky1)2, -NHC(0)(Ci-C6
alkyl), and nitro; and the
others are hydrogen.
In certain embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g.,
R6) is selected from halo,
C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, cyano, and
nitro; and the others are hydrogen.
In certain embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g.,
R6) is selected from halo,
C1-C6 alkyl, and C1-C6 haloalkyl; and the others are hydrogen.
In certain embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g.,
R6) is selected from halo
and C1-C6 alkyl; and the others are hydrogen.
In certain embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g.,
R6) is halo (e.g., bromo or
chloro) and C1-C6 alkyl; and the others are hydrogen.
In certain embodiments, one or two of R5, R6, R7, and R8 (e.g., one of, e.g.,
R6) is bromo; and the
others are hydrogen.
In some embodiments, R6 is selected from halo, hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6 thioalkoxy,
C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, cyano, -
NH2, -NH(C1-C6 alkyl), N(C1-
C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; and each of R5, R7, and R8 can be
as defined anywhere herein.
In certain embodiments, R6 is selected from halo, hydroxyl, sulfhydryl, C1-C6
alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy, C1-C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, -NH2, -NH(C1-C6
alkyl), N(C1-C6 alky1)2, -NHC(0)(C1-C6 alkyl), and nitro; and each of R5, R7,
and R8 is hydrogen.
In some embodiments, R6 is selected from halo, C1-C6 alkoxy, C1-C6 haloalkoxy,
C1-C6 alkyl, C1-C6
haloalkyl, cyano, and nitro; and each of RI, R2, and R4 can be as defined
anywhere herein.
In certain embodiments, R6 is selected from halo, C1-C6 alkoxy, C1-C6
haloalkoxy, C1-C6 alkyl, C1-C6
haloalkyl, cyano, and nitro; and each of R5, R7, and R8 is hydrogen.
In some embodiments, R6 is selected from halo, C1-C6 alkyl, and C1-C6
haloalkyl; and each of R5, R7,
and R8 can be as defined anywhere herein.
In certain embodiments, R6 is selected from halo, C1-C6 alkyl, and C1-C6
haloalkyl; and each of R5, R7,
and R8 is hydrogen.
In some embodiments, R6 is selected from halo and C1-C6 alkyl; and each of R5,
R7, and R8 can be as
defined anywhere herein.
In certain embodiments, R6 is selected from halo and C1-C6 alkyl; and each of
R5, R7, and R8 is
hydrogen.
In some embodiments, R6 is halo (e.g., bromo or chloro); and each of R5, R7,
and R8 can be as defined
anywhere herein..
In certain embodiments, R6 is halo (e.g., bromo or chloro); and each of R5,
R7, and R8 is hydrogen.
92
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In some embodiments, R6 is bromo; and each of le, R7, and le can be as defined
anywhere herein..
In certain embodiments, R6 is bromo; and each of le, R7, and le is hydrogen.
In some embodiments, each of R5, R6, R7, and le is independently selected from
hydrogen, halo, and
CI-C6 alkyl.
In certain embodiments, each of R5, R6, R7, and le is independently selected
from hydrogen and
halo(e.g., bromo or chloro).
In some embodiments, each of R5, R6, R7, and le is hydrogen.
In some embodiments, when any one or more of R5, R6, R7, and le can be a
substituent other than
hydrogen, said substituent, or each of said substituents, is other than Ci-C6
alkyl (e.g., Ci-C3 alkyl, e.g., CH3).
Embodiments can include any one or more of the features described anywhere
herein, including (but
not limited to) those described below.
{A}
Each of R1, R2, R3, and R4 can be as defined anywhere herein.
R3 is selected from halo, hydroxyl, sulfhydryl, C1-C6 alkoxy, C1-C6
thioalkoxy, C1-C6 haloalkoxy, CI-
C6 thiohaloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, cyano, -NH2, -NH(C1-C6
alkyl), N(C1-C6 alky1)2, -
NHC(0)(C1-C6 alkyl), and nitro; and each of R1, R2, and R4 can be as defined
anywhere herein (e.g., each of
R1, R2, and R4 is hydrogen).
R3 is selected from halo and C1-C6 alkyl; and each of R1, R2, and R4 can be as
defined anywhere herein
(e.g., each of R1, R2, and R4 is hydrogen).
R3 is halo (e.g., bromo or chloro); and each of R1, R2, and R4 can be as
defined anywhere herein (e.g.,
each of le, R2, and R4 is hydrogen).
R3 is bromo; and each of R1, R2, and R4 can be as defined anywhere herein
(e.g., each of R1, R2, and R4
is hydrogen).
Each of R1, R2, R3, and R4 is independently selected from hydrogen and
halo(e.g., bromo or chloro).
Each of R1, R2, R3, and R4 is hydrogen.
{B}
Each of L1 and L2 is, independently, C1-C3 alkylene, which is optionally
substituted with from 1-2
independently selected Re.
Each of L1 and L2 is CH2.
One of L1 and L2 is CH2 (e.g., L1), and the other (e.g., L2) is methylene that
is substituted with 1 or 2
(e.g., 1) independently selected Re, in which Re can be as defined anywhere
herein.
Each of L1 and L2 is methylene that is substituted with 1 or 2 (e.g., 1)
independently selected Re, in
which Re can be as defined anywhere herein.
L1 is C1-C3 (e.g., C1-C2) straight chain alkylene, which is optionally
substituted with from 1-2
independently selected Re, and L2 is a bond that directly connects A in
formula (I) to Z in formula (I).
93
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
{C}
One of RA' and RA2 is OR9, and the other is hydrogen. In embodiments, R9 can
be hydrogen. R9 can
be Ci-C3 alkyl (e.g., CH3).
One of RA' and RA2 can be halo (e.g., fluoro), and the other is hydrogen.
Each of RA' and RA2 can be a substituent other than hydrogen. For example,
each of RA' and RA2 can
be halo (e.g., fluoro). As another example, one of RA' and RA2 can be OR9
(e.g., in which R9 is hydrogen), and
the other is Ci-C3 alkyl (e.g., CH3).
Each of RA' and RA2 is hydrogen.
A is CRA1RA2, wherein each of RA' and RA2 is independently selected from
hydrogen, halo, Ci-C3
alkyl, or OR9; and each of L' and L2 is, independently, C1-C3 alkylene, which
is optionally substituted with
from 1-2 independently selected Re.
{D}
Z is -NR10R11, in which le and R11 can be as defined anywhere herein.
One of le and R11 is C6-C10 aryl that is optionally substituted with from 1-4
Rb. In embodiments, the
other of le and R11 is hydrogen or C1-C3 alkyl (e.g., CH3). In embodiments,
the other of le and R11 is
hydrogen.
In certain embodiments, one of le and R11 is unsubstituted phenyl, and the
other is hydrogen.
In certain embodiments, one of le and R11 is phenyl that is substituted with
1 Rb, and the other is
hydrogen. In embodiments, Rb is C1-C6 alkoxy (e.g., C1-C3 alkoxy, e.g., OCH3).
For example, one of le and
R11 is 3-methoxyphenyl, and the other is hydrogen.
Z is -OR' or ¨S(0)õR13, in which R12 and R13 can be as defined anywhere
herein.
Embodiments can include features from any one, two, three, or four of {A},
{B}, {C}, and {D}; or any
combinations thereof
In some embodiments:
R3 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
R1, R2, and R4 can be as defined anywhere herein (e.g., each of R1, R2, and R4
is hydrogen); and
R6 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
R5, R7, and le can be as defined anywhere herein (e.g., each of le, R7, and le
is hydrogen).
In some embodiments:
R3 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
R1, R2, and R4 can be as defined anywhere herein (e.g., each of R1, R2, and R4
is hydrogen); and
R6 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
R5, R7, and le can be as defined anywhere herein (e.g., each of le, R7, and le
is hydrogen); and
94
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
A is CRAIRA2, wherein each of RAI and RA2 is independently selected from
hydrogen, halo, Ci-C3
alkyl, or OR9; and each of LI and L2 is, independently, C1-C3 alkylene, which
is optionally substituted with
from 1-2 independently selected Re.
Embodiments can include any one or more features described herein (e.g., as
described under {B} and
{C} above).
In some embodiments:
R3 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
RI, R2, and R4 can be as defined anywhere herein (e.g., each of RI, R2, and R4
is hydrogen); and
R6 is a substituent other than hydrogen (e.g., halo and C1-C6 alkyl; e.g.,
halo, e.g., bromo); and each of
R5, R7, and R8 can be as defined anywhere herein (e.g., each of le, R7, and R8
is hydrogen); and
A is CRAIRA2, wherein each of RAI and RA2 is independently selected from
hydrogen, halo, C1-C3
alkyl, or OR9; and each of LI and L2 is, independently, C1-C3 alkylene, which
is optionally substituted with
from 1-2 independently selected Re; and
Z is -NRI0R11, in which RI and RII can be as defined anywhere herein.
Embodiments can include any one or more features described herein (e.g., as
described under {B}, {C}
, and {D} above).
In some embodiments:
each of LI and L2 is CH2.;
A is CRAIRA2, wherein one of RAI and RA2 is OR9, and the other is hydrogen.;
Z is -NRI0R11; and
each of le and Ril is independently selected from
(a) hydrogen;
(b) C6-Cio aryl that is optionally substituted with from 1-4 Rb;
(d) C1-C6 alkyl or C1-C6 haloalkyl, each of which is optionally substituted
with from 1-3 Rd;
(0 C2-C6 alkenyl or C2-C6 alkynyl.
Embodiments can include any one or more features described herein (e.g., as
described under {A}, {C}
, and {D} above).
In some embodiments:
A is CRAIRA2, in which each of RAI and RA2 is, independently, hydrogen, halo,
or C1-C3 alkyl; or
A is CRAIRA2, in which one of RAI and RA2 is halo (e.g., fluoro), and the
other of RAI and RA2 is,
independently, hydrogen, halo, or C1-C3 alkyl (e.g., hydrogen); or
A is CRAIRA2, in which one of RAI and RA2 is halo (e.g., fluoro), and the
other of RAI and RA2 is
hydrogen; and
RI, R2, R3, R4, LI, L2, and Z can be as defined anywhere herein; or a salt
(e.g., pharmaceutically
acceptable salt) thereof
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Embodiments can include features from any one, two, three, or four of {A},
{B}, {C}, and {D}; or any
combinations thereof
In some embodiments:
one of RAI and RA2 can be OR9. In embodiments, the other of RAI and RA2 can be
as defined anywhere
herein; e.g., the other of RAI and RA2 can be hydrogen or Ci-C3 alkyl. For
example, one of RAI and RA2 can be
OR9, and the other of RAI and RA2 is hydrogen. In embodiments, R9 can be
hydrogen; and
RI, R2, R3, R4, LI, L2, and Z can be as defined anywhere herein; or a salt
(e.g., pharmaceutically
acceptable salt) thereof
In embodiments, one or more of the following apply, e.g., when A is CHOH and Z
is NRI0R11:
= each of R3 and R6 is CH3; and/or each of R3 and R6 is bromo; and/or each of
R3 and R6 is
chloro; and/or one of R3 and R6 is CH3 (e.g., R6), and the other is bromo
(e.g., R3);
= each of RI and RII is other than hydrogen;
= each of RI and RII is hydrogen;
= one of RI and RII is heteroaryl as defined anywhere herein;
= L1 and/or L2 is C2-C3 alkylene (optionally substituted);
= (B) and/or (C) applies.
Embodiments can include features from any one, two, three, or four of {A},
{B}, {C}, and {D}; or any
combinations thereof
In some embodiments, Z is other than NRI0R11; and RI, R2, R3, R4, LI, L2, Z,
and A can be as defined
anywhere herein; or a salt (e.g., pharmaceutically acceptable salt) thereof In
embodiments, (B) and/or (C)
applies. Embodiments can include features from any one, two, three, or four of
{A}, {B}, {C}, and {D}; or any
combinations thereof
In some embodiments, Z is -0R12 and/or ¨S(0)õR13; and RI, R2, R3, R4, LI, L2,
and A can be as defined
anywhere herein; or a salt (e.g., pharmaceutically acceptable salt) thereof In
embodiments, (B) and/or (C)
applies. Embodiments can include features from any one, two, three, or four of
{A}, {B}, {C}, and {D}; or any
combinations thereof
In some embodiments, A is (ii) C=0; and/or (iv) heterocycloalkylene containing
from 3-5 ring atoms,
wherein from 1-2 of the ring atoms is independently selected from N, NH, N(C1-
C3 alkyl), 0, and S; and
wherein said heterocycloalkylene is (a) substituted with 1 oxo; and (b) is
optionally further substituted with
from 1-4 independently selected le; and RI, R2, R3, R4, LI, L2, and Z can be
as defined anywhere herein; or a
salt (e.g., pharmaceutically acceptable salt) thereof Embodiments can include
features from any one, two,
three, or four of {A}, {B}, {C}, and {D}; or any combinations thereof
III] In some embodiments, each of R and R' is, independently,
hydrogen, C1-C6 alkyl, or Ci-C6
haloalkyl.
In embodiments, R and R' can each be the same or different.
96
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In certain embodiments, each of R and R' is, independently, C1-C6 alkyl, e.g.,
each of R and R' is CH3.
In other embodiments, each of R and R' is hydrogen.
Embodiments can include any one or more of the features described anywhere
herein, including (but
not limited to) those described in conjunction with Formula (III).
RI] In some embodiments, R and R' together with C2 and C3, respectively, form
a fused
heterocyclic ring containing from 5-6 ring atoms, wherein from 1-2 of the ring
atoms is independently selected
from N, NH, N(C1-C6 alkyl), NC(0)(C1-C6 alkyl), 0, and S; and wherein said
heterocyclic ring is optionally
substituted with from 1-3 independently selected R. For purposes of
clarification and illustration, a non-
limiting example of these compounds is provided below (formula (IV)):
R63
R4 /
N
R3
R2 101 N\
\
R1 \ ----..
L-2
Z
(IV)
in which RI, R2, R3, R4, LI, L2, A, and Z can be as defined anywhere herein.
Here, R and R' together with C2
and C3, respectively, form a fused heterocyclic ring containing 5-6 ring
atoms.
Embodiments can include any one or more of the features described anywhere
herein, including (but
not limited to) those described in conjunction with Formula (III). In certain
embodiments, R63 can be
hydrogen or C1-C3 alkyl (e.g., CH3).
In some embodiments, it is provided:
(i) each of LI and L2 must be C1-C3 alkylene, which is optionally substituted
with from 1-2
independently selected Re when A is CH2; or
(ii) Z must be other than heteroaryl containing from 5-14 (e.g., 5-6 or 6)ring
atoms, wherein from 1-6
of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and
S; and wherein said heteroaryl
is optionally substituted with from 1-4 independently selected Rb; e.g., other
than substituted pyridyl, e.g.,
other than pyridyl substituted with Ci-C3 alkyl (e.g., CH3), e.g., other than
2 or 6-methylpyridyl.
[IV] In some embodiments, R and R' together with C2 and C3, respectively, form
a fused C5-C6
cycloalkyl ring that is optionally substituted with from 1-4 independently
selected R. For purposes of
clarification and illustration, a non-limiting example of such compounds is
provided below (formula (V)):
97
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
R4
R3
411)
R2 10 N
\
W \
L-2
---....z
(V)
in which RI, R2, R3, R4, LI, L2, A, and Z can be as defined anywhere herein.
Here, R and R' together with C2
and C3, respectively, form a fused C6 cycloalkyl ring. Embodiments can include
any one or more of the
features described anywhere herein, including (but not limited to) those
described in conjunction with Formula
(III).
[V] In some embodiments, R and R' together with C2 and C3,
respectively, form a fused heteroaryl
ring containing from 5-6 ring atoms, wherein from 1-2 of the ring atoms is
independently selected from N,
NH, N(C1-C3 alkyl), 0, and S; and wherein said heteroaryl ring is optionally
substituted with from 1-3
independently selected Rb. See, e.g., the title compound of Example 13.
Embodiments can include any one or
more of the features described anywhere herein, including (but not limited to)
those described in conjunction
with Formula (III).
Any genus, subgenus, or specific compound described herein can include one or
more of the
stereochemistry features described herein (e.g., as delineated in the
Summary).
Compound Forms and Salts
The compounds of the presently disclosed embodiments may contain one or more
asymmetric centers
and thus occur as racemates and racemic mixtures, enantiomerically enriched
mixtures, single enantiomers,
individual diastereomers and diastereomeric mixtures. All such isomeric forms
of these compounds are
expressly included in the presently disclosed embodiments. The compounds of
the presently disclosed
embodiments may also contain linkages (e.g., carbon-carbon bonds, carbon-
nitrogen bonds such as amide
bonds) wherein bond rotation is restricted about that particular linkage, e.g.
restriction resulting from the
presence of a ring or double bond. Accordingly, all cis/trans and E/Z isomers
and rotational isomers are
expressly included in the presently disclosed embodiments. The compounds of
the presently disclosed
embodiments may also be represented in multiple tautomeric forms, in such
instances, the presently disclosed
embodiments expressly includes all tautomeric forms of the compounds described
herein, even though only a
single tautomeric form may be represented. All such isomeric forms of such
compounds are expressly
included in the presently disclosed embodiments.
98
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Optical isomers can be obtained in pure form by standard procedures known to
those skilled in the art,
and include, but are not limited to, diastereomeric salt formation, kinetic
resolution, and asymmetric synthesis.
See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions
(Wiley Interscience, New York,
1981); Wilen, S.H., et al., Tetrahedron 33:2725 (1977); Eliel, E.L.
Stereochemistry of Carbon Compounds
(McGraw-Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical
Resolutions p. 268 (E.L. Eliel,
Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972), each of which is
incorporated herein by reference in
their entireties. It is also understood that the presently disclosed
embodiments encompass all possible
regioisomers, and mixtures thereof, which can be obtained in pure form by
standard separation procedures
known to those skilled in the art, and include, but are not limited to, column
chromatography, thin-layer
chromatography, and high-performance liquid chromatography.
The compounds of the presently disclosed embodiments include the compounds
themselves, as well as
their salts and their prodrugs, if applicable. A salt, for example, can be
formed between an anion and a
positively charged substituent (e.g., amino) on a compound described herein.
Suitable anions include chloride,
bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate,
trifluoroacetate, and acetate. Likewise,
a salt can also be formed between a cation and a negatively charged
substituent (e.g., carboxylate) on a
compound described herein. Suitable cations include sodium ion, potassium ion,
magnesium ion, calcium ion,
and an ammonium cation such as tetramethylammonium ion. Examples of prodrugs
include C1_6 alkyl esters
of carboxylic acid groups, which, upon administration to a subject, are
capable of providing active compounds.
Pharmaceutically acceptable salts of the compounds of the presently disclosed
embodiments include
those derived from pharmaceutically acceptable inorganic and organic acids and
bases. As used herein, the
term "pharmaceutically acceptable salt" refers to a salt formed by the
addition of a pharmaceutically
acceptable acid or base to a compound disclosed herein. As used herein, the
phrase "pharmaceutically
acceptable" refers to a substance that is acceptable for use in pharmaceutical
applications from a toxicological
perspective and does not adversely interact with the active ingredient.
Examples of suitable acid salts include acetate, adipate, alginate, aspartate,
benzoate,
benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate,
digluconate, dodecylsulfate,
ethanesulfonate, formate, fumarate, glucoheptanoate, glycolate, hemisulfate,
heptanoate, hexanoate,
hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate,
maleate, malonate,
methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate,
pectinate, persulfate, 3-
phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate,
succinate, sulfate, tartrate, thiocyanate,
tosylate and undecanoate. Other acids, such as oxalic, while not in themselves
pharmaceutically acceptable,
may be employed in the preparation of salts useful as intermediates in
obtaining the compounds of the
presently disclosed embodiments and their pharmaceutically acceptable acid
addition salts. Salts derived from
appropriate bases include alkali metal (e.g., sodium), alkaline earth metal
(e.g., magnesium), ammonium and
N-(alkyl)4+ salts. The presently disclosed embodiments also envision the
quaternization of any basic nitrogen-
99
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
containing groups of the compounds disclosed herein. Water or oil-soluble or
dispersible products may be
obtained by such quaternization. Salt forms of the compounds of any of the
formulae herein can be amino acid
salts of carboxyl groups (e.g. L-arginine, -lysine, -histidine salts).
Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th
ed., Mack Publishing
Company, Easton, Pa., 1985, p. 1418; Journal of Pharmaceutical Science, 66, 2
(1977); and "Pharmaceutical
Salts: Properties, Selection, and Use A Handbook; Wermuth, C. G. and Stahl, P.
H. (eds.) Verlag Helvetica
Chimica Acta, Zurich, 2002 [ISBN 3-906390-26-8] each of which is incorporated
herein by reference in their
entireties.
The neutral forms of the compounds may be regenerated by contacting the salt
with a base or acid and
isolating the parent compound in the conventional manner. The parent form of
the compound differs from the
various salt forms in certain physical properties, such as solubility in polar
solvents, but otherwise the salts are
equivalent to the parent form of the compound for the purposes of the
presently disclosed embodiments.
In addition to salt forms, the presently disclosed embodiments provide
compounds which are in a
prodrug form. Prodrugs of the compounds described herein are those compounds
that undergo chemical
changes under physiological conditions to provide the compounds of the
presently disclosed embodiments.
Additionally, prodrugs can be converted to the compounds of the presently
disclosed embodiments by
chemical or biochemical methods in an ex vivo environment. For example,
prodrugs can be slowly converted
to the compounds of the presently disclosed embodiments when placed in a
transdermal patch reservoir with a
suitable enzyme or chemical reagent. Prodrugs are often useful because, in
some situations, they may be easier
to administer than the parent drug. They may, for instance, be more
bioavailable by oral administration than
the parent drug. The prodrug may also have improved solubility in
pharmacological compositions over the
parent drug. A wide variety of prodrug derivatives are known in the art, such
as those that rely on hydrolytic
cleavage or oxidative activation of the prodrug. An example, without
limitation, of a prodrug would be a
compound of the presently disclosed embodiments which is administered as an
ester (the "prodrug"), but then
is metabolically hydrolyzed to the carboxylic acid, the active entity.
Additional examples include peptidyl
derivatives of a compound of the presently disclosed embodiments.
The presently disclosed embodiments also include various hydrate and solvate
forms of the
compounds.
The compounds of the presently disclosed embodiments may also contain
unnatural proportions of
atomic isotopes at one or more of the atoms that constitute such compounds.
For example, the compounds may
be radiolabeled with radioactive isotopes, such as for example tritium (3H),
iodine-125 (1251) or carbon-14
(14C). All isotopic variations of the compounds of the presently disclosed
embodiments, whether radioactive or
not, are intended to be encompassed within the scope of the presently
disclosed embodiments.
SYNTHESIS
100
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The compounds of the presently disclosed embodiments can be conveniently
prepared in accordance
with the procedures outlined in the Examples section, from commercially
available starting materials,
compounds known in the literature, or readily prepared intermediates, by
employing standard synthetic
methods and procedures known to those skilled in the art. Standard synthetic
methods and procedures for the
preparation of organic molecules and functional group transformations and
manipulations can be readily
obtained from the relevant scientific literature or from standard textbooks in
the field. It will be appreciated
that where typical or preferred process conditions (i.e., reaction
temperatures, times, mole ratios of reactants,
solvents, pressures, etc.) are given, other process conditions can also be
used unless otherwise stated. Optimum
reaction conditions may vary with the particular reactants or solvent used,
but such conditions can be
determined by one skilled in the art by routine optimization procedures. Those
skilled in the art of organic
synthesis will recognize that the nature and order of the synthetic steps
presented may be varied for the
purpose of optimizing the formation of the compounds described herein.
Synthetic chemistry transformations (including protecting group methodologies)
useful in synthesizing
the compounds described herein are known in the art and include, for example,
those such as described in R.C.
Larock, Comprehensive Organic Transformations, 2d.ed., Wiley-VCH Publishers
(1999); P.G.M. Wuts and
T.W. Greene, Protective Groups in Organic Synthesis, 4th Ed., John Wiley and
Sons (2007); L. Fieser and M.
Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and
Sons (1994); and L. Paquette, ed.,
Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995),
and subsequent editions
thereof
The processes described herein can be monitored according to any suitable
method known in the art.
For example, product formation can be monitored by spectroscopic means, such
as nuclear magnetic resonance
spectroscopy (e.g., 1H or 13C), infrared spectroscopy (FT-IR),
spectrophotometry (e.g., UV-visible), or mass
spectrometry (MS), or by chromatography such as high performance liquid
chromatography (HPLC) or thin
layer chromatography (TLC).
Preparation of compounds can involve the protection and deprotection of
various chemical groups.
The need for protection and deprotection, and the selection of appropriate
protecting groups can be readily
determined by one skilled in the art. The chemistry of protecting groups can
be found, for example, in Greene,
et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991,
which is incorporated herein by
reference in its entirety.
The reactions of the processes described herein can be carried out in suitable
solvents which can be
readily selected by one of skill in the art of organic synthesis. Suitable
solvents can be substantially
nonreactive with the starting materials (reactants), the intermediates, or
products at the temperatures at which
the reactions are carried out, i.e., temperatures which can range from the
solvent's freezing temperature to the
solvent's boiling temperature. A given reaction can be carried out in one
solvent or a mixture of more than
101
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
one solvents. Depending on the particular reaction step, suitable solvents for
a particular reaction step can be
selected.
Resolution of racemic mixtures of compounds can be carried out by any of
numerous methods known
in the art. An example method includes preparation of the Mosher's ester or
amide derivative of the
corresponding alcohol or amine, respectively. The absolute configuration of
the ester or amide is then
determined by proton and/or 19F NMR spectroscopy. An example method includes
fractional recrystallization
using a "chiral resolving acid" which is an optically active, salt-forming
organic acid. Suitable resolving
agents for fractional recrystallization methods are, for example, optically
active acids, such as the D and L
forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid,
mandelic acid, malic acid, lactic acid or the
various optically active camphorsulfonic acids. Resolution of racemic mixtures
can also be carried out by
elution on a column packed with an optically active resolving agent (e.g.,
dinitrobenzoylphenylglycine).
Suitable elution solvent compositions can be determined by one skilled in the
art.
The compounds of the presently disclosed embodiments can be prepared, for
example, using the
reaction pathways and techniques as described below.
A series of carbazole 1,2-aminoalcohol compounds of formula 3 may be prepared
by the method
outlined in Scheme 1. The 9-oxiranylmethy1-9H-carbazole of formula 2 may be
prepared from an
appropriately substituted carbazole of formula 1 and epichlorohydrin in the
presence of a strong base such as
sodium hydride.
Scheme 1
R
R R 5 R4 R5 R4 R5
R2 4 R3 6 R3
R3 R6 CI 0
HNRi0Rii
Vs. R2 . # R7 _____________ 11.- R2 = 110 R6
R7
. 110 R7
N N
N
R1 H R8 R1 R8 R1 y 1 8
1 2 / 3
NRioRii
0 OH
9-oxiranylmethy1-9H-carbazole
The oxiranyl ring of formula 2 may be opened in the presence of a primary or
secondary amine to
produce the 1,2-amino alcohol of formula 3. Such reactive primary or secondary
amines can be, but are not
limited to, phenethylamine, 3-phenylally1 amine, and N-substituted piperazines
and the like.
Alternatively, a variety of carbazole 1,2-aminoalcohol compounds of formula 8
may be prepared by
the method outlined in Scheme 2. The epoxide of 9-oxiranylmethy1-9H-carbazole
of formula 2 may be opened
with a primary amine, H2NR10, to produce the secondary aminoalcohol of formula
4 and then protected with an
amine protecting group (P) such as tert-butoxycarbonyl (Boc) to afford the
protected aminoalochol of formula
5. Next, the hydroxyl group of formula 5 may be alkylated with a strong base
such as sodium hydride and an
alkylating agent (RX) such as an alkyl halide, tosylate, triflate or mesylate
to produce the ether of formula 6.
Removal of the amine protecting group in the presence of a suitable acid can
provide the desired OR ether
102
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
compounds of formula 7. Finally, reductive alkylation of the secondary amine
of formula 7 may be achieved
in the presence of an aldehyde and a reducing agent such as sodium cyano
borohydride (NaCNBH3) to provide
the tertiary 1,2-aminoalcohol of formula 8.
Scheme 2
R4 R5 R4 R5
R3 R6 R3 R6
H2NR1
R2 * * R7 -1P.- R2 * * R7
N N
R1 \_7R8 R1
L R8
NHRio
2 0 4 OH
R4 R5 R4 R5
R3 R6 R3 R6
amine protecting R2 * .
R2 * . R7
group (P) RX
N -).-- N
R7
R1 yI8 R10 R1 6
R10
N
N
OH 1
OR 1
P P
R4 R5 R4 R5
R3 R6 R3 R6
acid____ v.
R2 . . R7 R2 * * R7
(reductive amination)
N N
R1 L R8 Rlizi R1y!
7 8
N NRioRii
H
5 OR OR
A series of substituted indole compounds of formula 11 and 12 may be prepared
by the method
outlined below in Scheme 3. Compounds of formula 11 may be prepared by the
alkylation of an indole of
formula 9 with an epoxide A, for example with epichlorohydrin or
epibromohydrin, in the presence of a strong
base such as potassium hydroxide (KOH) or n-butyllithium (n-BuLi) to produce
the oxiranyl indole of formula
10. Next, opening of the epoxide of compounds of formula 10 with a primary
amine, substituted alcohol or
thiol in the presence of a strong base or a mild Lewis acid such as lithium
bromide (LiBr) or bismuth chloride
(BiC13) can provide the alcohol of formula 11. Additionally, compounds of
formula 12 may be prepared by
opening an epoxide B at the less hindered position with the indole nitrogen of
formula 9.
103
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Scheme 3
R5 R6 0
x A R5 R6
0 HYR8 R5
R6
----- -----
¨µ Rzi ,
0 H
R4 ,NH X = CI, Br R4 N---/ Y = 0, N, S
R3
*
-
R8
R3 Ri R3 Ri Ri
R2
R2 R2
9 1 0 11
0
I R7...õ...../..< j B
R5 R6 OH
......zr
____
R4 0 N
12
R3 Rij..y
R2
In addition, a variety of epoxide derivatives may be prepared by following the
methods outlined in
Scheme 4. The secondary alcohol of compounds of formula 11 may be oxidized
using an oxidizing agent or
under Swern-like oxidation conditions to provide the ketone of formula 13
which can further undergo
reductive amination to provide the amine of compound 14. Alternatively, the
secondary alcohol may be
converted into an ester using a carboxylic acid anhydride (where Z=R"C(0)) or
an ether (where Z=alkly1)
using standard alkylation conditions to produce compounds of formula 15.
Fluorine compounds of formula 16
may be prepared by reaction of the alcohol of formula 11 with a fluorinating
agent such as diethylaminosulfur
trifluoride (DAST). Nitrogen-heteroarylated compounds of formula 17 may be
prepared in the presence of a
catalytic amount of copper iodide and a heteroaryl iodide starting from
compounds of formula 11 (where
Y=N). Finally, sulfoxides and sulfones of formula 18 may be prepared under
oxidative conditions, for
example in the presence of m-chloroperoxybenzoic acid (m-CPBA), starting from
sulfides of formula 11
(where Y=S).
104
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Scheme 4
R5
R6
R4 --__ NR 9
. N )2(..... R8 R5
R5 R6
R6 OZ
R4 ---- 0 R3 R4 --._
. N Y-.... R2 R1
14 44,1k N R3
11'R8
R3
R8 Z =
13
Palkyl or A ff
R1 1) oxidation of OH R2 RC (0)
I'
R2
2) reductive annination
with R9NH
oxidation
capping with (RCO)20
R5 or alkyl halide
R6
R4--,_ OH
R3
. N 21'
R8
11 Y = 0, N, S
R
R2 1
m-CPBA
Y R8 = SR8
fluorination 1. CIC(0)0CH3
2. (HeteroaryI)-1,Cul
R5
3. NaOH R6
R5 R6 R4 --._ OH
R4 --__ F . N .(0)nR8
. NY,
R8
n=1 or2
R3 R5 R6 R3 -- 0 H R8 R1
R4 ¨.. R2 18
R1 16 . N II,(heteroaryl)
R2
R3
Ri
R2 17
PHARMACEUTICAL COMPOSITIONS
The term "pharmaceutically acceptable carrier" refers to a carrier or adjuvant
that may be administered
to a subject (e.g., a patient), together with a compound of the presently
disclosed embodiments, and which
does not destroy the pharmacological activity thereof and is nontoxic when
administered in doses sufficient to
deliver a therapeutic amount of the compound.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used
in the compositions of
the presently disclosed embodiments include, but are not limited to, ion
exchangers, alumina, aluminum
stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-
tocopherol polyethyleneglycol
1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens
or other similar polymeric
delivery matrices, serum proteins, such as human serum albumin, buffer
substances such as phosphates,
105
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of
saturated vegetable fatty acids, water,
salts, or electrolytes, such as protamine sulfate, disodium hydrogen
phosphate, potassium hydrogen phosphate,
sodium chloride, zinc salts, colloidal silica, magnesium trisilicate,
polyvinyl pyrrolidone, cellulose-based
substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates,
waxes, polyethylene-
polyoxypropylene-block polymers, polyethylene glycol and wool fat.
Cyclodextrins such as a-, 13-, and y-
cyclodextrin, or chemically modified derivatives such as
hydroxyalkylcyclodextrins, including 2- and 3-
hydroxypropy1-13-cyclodextrins, or other solubilized derivatives may also be
advantageously used to enhance
delivery of compounds of the formulae described herein.
The compositions for administration can take the form of bulk liquid solutions
or suspensions, or bulk
powders. More commonly, however, the compositions are presented in unit dosage
forms to facilitate accurate
dosing. The term "unit dosage forms" refers to physically discrete units
suitable as unitary dosages for human
subjects and other mammals, each unit containing a predetermined quantity of
active material calculated to
produce the desired therapeutic effect, in association with a suitable
pharmaceutical excipient. Typical unit
dosage forms include prefilled, premeasured ampules or syringes of the liquid
compositions or pills, tablets,
capsules, losenges or the like in the case of solid compositions. In such
compositions, the compound is usually
a minor component (from about 0.1 to about 50% by weight or preferably from
about 1 to about 40% by
weight) with the remainder being various vehicles or carriers and processing
aids helpful for forming the
desired dosing form.
The amount administered depends on the compound formulation, route of
administration, etc. and is
generally empirically determined in routine trials, and variations will
necessarily occur depending on the
target, the host, and the route of administration, etc. Generally, the
quantity of active compound in a unit
dose of preparation may be varied or adjusted from about 1, 3, 10 or 30 to
about 30, 100, 300 or 1000 mg,
according to the particular application. In a particular embodiment, unit
dosage forms are packaged in a
multipack adapted for sequential use, such as blisterpack, comprising sheets
of at least 6, 9 or 12 unit dosage
forms. The actual dosage employed may be varied depending upon the
requirements of the patient and the
severity of the condition being treated. Determination of the proper dosage
for a particular situation is within
the skill of the art. Generally, treatment is initiated with smaller dosages
which are less than the optimum
dose of the compound. Thereafter, the dosage is increased by small amounts
until the optimum effect under
the circumstances is reached. For convenience, the total daily dosage may be
divided and administered in
portions during the day if desired.
The following are examples (Formulations 1-4) of capsule formulations.
106
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Capsule Formulations
Capsule Formulation Formulation 1; Formulation 2;
Formulation 3; Formulation 4;
mg/capsule mg/capsule mg/capsule
mg/capsule
Carbazole (solid 100 400 400
200
solution)
Silicon Dioxide 0.625 2.5 3.75
1.875
Magnesium Stearate 0.125 0.5 0.125
0.625
NF2
Croscarmellose Sodium 11.000 44.0 40.0
20.0
NF
Pluronic F68 NF 6.250 25.0 50.0
25.0
Silicon Dioxide NF 0.625 2.5 3.75
1.875
Magnesium Stearate NF 0.125 0.5 1.25
0.625
Total 118.750 475.00 475.00
475.00
Capsule Size No.4 No.0 No.0
No.2
Preparation of Solid Solution
Crystalline carbazole (80 g/batch) and the povidone (NF K29/32 at 160 g/batch)
are dissolved in
methylene chloride (5000 mL). The solution is dried using a suitable solvent
spray dryer and the residue
reduced to fine particles by grinding. The powder is then passed through a 30
mesh screen and confirmed to be
amorphous by x-ray analysis.
The solid solution, silicon dioxide and magnesium stearate are mixed in a
suitable mixer for 10
minutes. The mixture is compacted using a suitable roller compactor and milled
using a suitable mill fitted
with 30 mesh screen. Croscarmellose sodium, Pluronic F68 and silicon dioxide
are added to the milled mixture
and mixed further for 10 minutes. A premix is made with magnesium stearate and
equal portions of the
mixture. The premix is added to the remainder of the mixture, mixed for 5
minutes and the mixture
encapsulated in hard shell gelatin capsule shells.
USE
In one aspect, methods for treating (e.g., controlling, relieving,
ameliorating, alleviating, or slowing
the progression of) or methods for preventing (e.g., delaying the onset of or
reducing the risk of developing)
one or more diseases, disorders, or conditions caused by, or associated with,
aberrant (e.g., insufficient)
neurogenesis or accelerated neuron cell death in a subject in need thereof are
featured. The methods include
administering to the subject an effective amount of a compound of formula (I)
(and/or a compound of any of
107
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
the other formulae described herein) or a salt (e.g., a pharmaceutically
acceptable salt) thereof as defined
anywhere herein to the subject.
In another aspect, the use of a compound of formula (I) (and/or a compound of
any of the other
formulae described herein) or a salt (e.g., a pharmaceutically acceptable
salt) thereof as defined anywhere
herein in the preparation of, or for use as, a medicament for the treatment
(e.g., controlling, relieving,
ameliorating, alleviating, or slowing the progression of) or prevention (e.g.,
delaying the onset of or reducing
the risk of developing) of one or more diseases, disorders, or conditions
caused by, or associated with, aberrant
(e.g., insufficient) neurogenesis or exacerbated neuronal cell death is
featured.
In embodiments, the one or more diseases, disorders, or conditions can include
neuropathies, nerve
trauma, and neurodegenerative diseases. In embodiments, the one or more
diseases, disorders, or conditions
can be diseases, disorders, or conditions caused by, or associated with
aberrant (e.g., insufficient) neurogenesis
(e.g., aberrant hippocampal neurogenesis as is believed to occur in
neuropsychiatric diseases) or accelerated
death of existing neurons. Examples of the one or more neuropsychiatric and
neurodegenerative diseases
include, but are not limited to, schizophrenia,major depression, bipolar
disorder, normal aging, epilepsy,
traumatic brain injury, post-traumatic stress disorder, Parkinson's disease,
Alzheimer's disease, Down
syndrome, spinocerebellar ataxia, amyotrophic lateral sclerosis, Huntington's
disease, stroke, radiation
therapy, chronic stress, and abuse of neuro-active drugs (such as alcohol,
opiates, methamphetamine,
phencyclidine, and cocaine), retinal degeneration, spinal cord injury,
peripheral nerve injury, physiological
weight loss associated with various conditions, and cognitive decline
associated with normal aging, radiation
therapy, and chemotherapy. The resultant promotion of neurogenesis or survival
of existing neurons ( i.e. a
resultant promotion of survival, growth, development, function and/or
generation of neurons) may be detected
directly, indirectly or inferentially from an improvement in, or an
amelioration of one or more symptoms of
the disease or disorder caused by or associated with aberrant neurogenesis or
survival of existing neurons.
Suitable assays which directly or indirectly detect neural survival, growth,
development, function and/or
generation are known in the art, including axon regeneration in rat models
(e.g. Park et al., Science. 2008 Nov
7; 322:963-6), nerve regeneration in a rabbit facial nerve injury models (e.g.
Zhang et al., J Transl Med. 2008
Nov 5;6(1):67); sciatic nerve regeneration in rat models (e.g. Sun et al.,
Cell Mol Neurobiol. 2008 Nov 6);
protection against motor neuron degeneration in mice (e.g. Poesen et al., J.
Neurosci. 2008 Oct
15;28(42):10451-9); rat model of Alzheimer's disease, (e.g. Xuan et al.,
Neurosci Lett. 2008 Aug
8;440(3):331-5); animal models of depression (e.g. Schmidt et al., Behav
Pharmacol. 2007 Sep;18(5-6):391-
418; Krishnan et al., Nature 2008, 455, 894-902); and/or those exemplified
herein.
ADMINISTRATION
The compounds and compositions described herein can, for example, be
administered orally,
parenterally (e.g., subcutaneously, intracutaneously, intravenously,
intramuscularly, intraarticularly,
108
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
intraarterially, intrasynovially, intrasternally, intrathecally,
intralesionally and by intracranial injection or
infusion techniques), by inhalation spray, topically, rectally, nasally,
buccally, vaginally, via an implanted
reservoir, by injection, subdermally, intraperitoneally, transmucosally, or in
an ophthalmic preparation, with a
dosage ranging from about 0.01 mg/kg to about 1000 mg/kg, (e.g., from about
0.01 to about 100 mg/kg, from
about 0.1 to about 100 mg/kg, from about 1 to about 100 mg/kg, from about 1 to
about 10 mg/kg) every 4 to
120 hours, or according to the requirements of the particular drug. The
interrelationship of dosages for
animals and humans (based on milligrams per meter squared of body surface) is
described by Freireich et al.,
Cancer Chemother. Rep. 50, 219 (1966). Body surface area may be approximately
determined from height
and weight of the patient. See, e.g., Scientific Tables, Geigy
Pharmaceuticals, Ardsley, New York, 537
(1970). In certain embodiments, the compositions are administered by oral
administration or administration by
injection. The methods herein contemplate administration of an effective
amount of compound or compound
composition to achieve the desired or stated effect. Typically, the
pharmaceutical compositions of the
presently disclosed embodiments will be administered from about 1 to about 6
times per day or alternatively,
as a continuous infusion. Such administration can be used as a chronic or
acute therapy.
Lower or higher doses than those recited above may be required. Specific
dosage and treatment
regimens for any particular patient will depend upon a variety of factors,
including the activity of the specific
compound employed, the age, body weight, general health status, sex, diet,
time of administration, rate of
excretion, drug combination, the severity and course of the disease, condition
or symptoms, the patient's
disposition to the disease, condition or symptoms, and the judgment of the
treating physician.
Upon improvement of a patient's condition, a maintenance dose of a compound,
composition or
combination of the presently disclosed embodiments may be administered, if
necessary. Subsequently, the
dosage or frequency of administration, or both, may be reduced, as a function
of the symptoms, to a level at
which the improved condition is retained when the symptoms have been
alleviated to the desired level.
Patients may, however, require intermittent treatment on a long-term basis
upon any recurrence of disease
symptoms.
In some embodiments, the compounds described herein can be coadministered with
one or more other
therapeutic agents. In certain embodiments, the additional agents may be
administered separately, as part of a
multiple dose regimen, from the compounds of the presently disclosed
embodiments (e.g., sequentially, e.g.,
on different overlapping schedules with the administration of one or more
compounds of formula (I)
(including any subgenera or specific compounds thereof)). In other
embodiments, these agents may be part of
a single dosage form, mixed together with the compounds of the presently
disclosed embodiments in a single
composition. In still another embodiment, these agents can be given as a
separate dose that is administered at
about the same time that one or more compounds of formula (I) (including any
subgenera or specific
compounds thereof) are administered (e.g., simultaneously with the
administration of one or more compounds
of formula (I) (including any subgenera or specific compounds thereof)). When
the compositions of the
109
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
presently disclosed embodiments include a combination of a compound of the
formulae described herein and
one or more additional therapeutic or prophylactic agents, both the compound
and the additional agent can be
present at dosage levels of between about 1 to 100%, and more preferably
between about 5 to 95% of the
dosage normally administered in a monotherapy regimen.
The compositions of the presently disclosed embodiments may contain any
conventional non-toxic
pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases,
the pH of the formulation may be
adjusted with pharmaceutically acceptable acids, bases or buffers to enhance
the stability of the formulated
compound or its delivery form.
The compositions may be in the form of a sterile injectable preparation, for
example, as a sterile
injectable aqueous or oleaginous suspension. This suspension may be formulated
according to techniques
known in the art using suitable dispersing or wetting agents (such as, for
example, Tween 80) and suspending
agents. The sterile injectable preparation may also be a sterile injectable
solution or suspension in a non-toxic
parenterally acceptable diluent or solvent, for example, as a solution in 1,3-
butanediol. Among the acceptable
vehicles and solvents that may be employed are mannitol, water, Ringer's
solution and isotonic sodium
chloride solution. In addition, sterile, fixed oils are conventionally
employed as a solvent or suspending
medium. For this purpose, any bland fixed oil may be employed including
synthetic mono- or diglycerides.
Fatty acids, such as oleic acid and its glyceride derivatives are useful in
the preparation of injectables, as are
natural pharmaceutically-acceptable oils, such as olive oil or castor oil,
especially in their polyoxyethylated
versions. These oil solutions or suspensions may also contain a long-chain
alcohol diluent or dispersant, or
carboxymethyl cellulose or similar dispersing agents which are commonly used
in the formulation of
pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
Other commonly used
surfactants such as Tweens or Spans and/or other similar emulsifying agents or
bioavailability enhancers
which are commonly used in the manufacture of pharmaceutically acceptable
solid, liquid, or other dosage
forms may also be used for the purposes of formulation.
The compositions of the presently disclosed embodiments may be orally
administered in any orally
acceptable dosage form including, but not limited to, capsules, tablets,
emulsions and aqueous suspensions,
dispersions and solutions. In the case of tablets for oral use, carriers which
are commonly used include lactose
and corn starch. Lubricating agents, such as magnesium stearate, are also
typically added. For oral
administration in a capsule form, useful diluents include lactose and dried
corn starch. When aqueous
suspensions and/or emulsions are administered orally, the active ingredient
may be suspended or dissolved in
an oily phase is combined with emulsifying and/or suspending agents. If
desired, certain sweetening and/or
flavoring and/or coloring agents may be added.
The compositions of the presently disclosed embodiments may also be
administered in the form of
suppositories for rectal administration. These compositions can be prepared by
mixing a compound of the
presently disclosed embodiments with a suitable non-irritating excipient which
is solid at room temperature
110
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
but liquid at the rectal temperature and therefore will melt in the rectum to
release the active components.
Such materials include, but are not limited to, cocoa butter, beeswax and
polyethylene glycols.
Topical administration of the compositions of the presently disclosed
embodiments is useful when the
desired treatment involves areas or organs readily accessible by topical
application. For application topically
to the skin, the composition should be formulated with a suitable ointment
containing the active components
suspended or dissolved in a carrier. Carriers for topical administration of
the compounds of the presently
disclosed embodiments include, but are not limited to, mineral oil, liquid
petroleum, white petroleum,
propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax
and water. Alternatively,
the composition can be formulated with a suitable lotion or cream containing
the active compound suspended
or dissolved in a carrier with suitable emulsifying agents. Suitable carriers
include, but are not limited to,
mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl
alcohol, 2-octyldodecanol, benzyl
alcohol and water. The compositions of the presently disclosed embodiments may
also be topically applied to
the lower intestinal tract by rectal suppository formulation or in a suitable
enema formulation.
In some embodiments, topical administration of the compounds and compositions
described herein
may be presented in the form of an aerosol, a semi-solid pharmaceutical
composition, a powder, or a solution.
By the term "a semi-solid composition" is meant an ointment, cream, salve,
jelly, or other pharmaceutical
composition of substantially similar consistency suitable for application to
the skin. Examples of semi-solid
compositions are given in Chapter 17 of The Theory and Practice of Industrial
Pharmacy, Lachman,
Lieberman and Kanig, published by Lea and Febiger (1970) and in Remington's
Pharmaceutical Sciences, 21st
Edition (2005) published by Mack Publishing Company, which is incorporated
herein by reference in its
entirety.
Topically-transdermal patches are also included in the presently disclosed
embodiments. Also within
the presently disclosed embodiments is a patch to deliver active
chemotherapeutic combinations herein. A
patch includes a material layer (e.g., polymeric, cloth, gauze, bandage) and
the compound of the formulae
herein as delineated herein. One side of the material layer can have a
protective layer adhered to it to resist
passage of the compounds or compositions. The patch can additionally include
an adhesive to hold the patch
in place on a subject. An adhesive is a composition, including those of either
natural or synthetic origin, that
when contacted with the skin of a subject, temporarily adheres to the skin. It
can be water resistant. The
adhesive can be placed on the patch to hold it in contact with the skin of the
subject for an extended period of
time. The adhesive can be made of a tackiness, or adhesive strength, such that
it holds the device in place
subject to incidental contact, however, upon an affirmative act (e.g.,
ripping, peeling, or other intentional
removal) the adhesive gives way to the external pressure placed on the device
or the adhesive itself, and allows
for breaking of the adhesion contact. The adhesive can be pressure sensitive,
that is, it can allow for
positioning of the adhesive (and the device to be adhered to the skin) against
the skin by the application of
pressure (e.g., pushing, rubbing,) on the adhesive or device.
111
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The compositions of the presently disclosed embodiments may be administered by
nasal aerosol or
inhalation. Such compositions are prepared according to techniques well-known
in the art of pharmaceutical
formulation and may be prepared as solutions in saline, employing benzyl
alcohol or other suitable
preservatives, absorption promoters to enhance bioavailability, fluorocarbons,
and/or other solubilizing or
dispersing agents known in the art.
A composition having the compound of the formulae herein and an additional
agent (e.g., a therapeutic
agent) can be administered using any of the routes of administration described
herein. In some embodiments,
a composition having the compound of the formulae herein and an additional
agent (e.g., a therapeutic agent)
can be administered using an implantable device. Implantable devices and
related technology are known in
the art and are useful as delivery systems where a continuous, or timed-
release delivery of compounds or
compositions delineated herein is desired. Additionally, the implantable
device delivery system is useful for
targeting specific points of compound or composition delivery (e.g., localized
sites, organs). Negrin et al.,
Biomaterials, 22(6):563 (2001). Timed-release technology involving alternate
delivery methods can also be
used in the presently disclosed embodiments. For example, timed-release
formulations based on polymer
technologies, sustained-release techniques and encapsulation techniques (e.g.,
polymeric, liposomal) can also
be used for delivery of the compounds and compositions delineated herein.
The presently disclosed embodiments will be further described in the following
examples. It should
be understood that these examples are for illustrative purposes only and are
not to be construed as limiting the
presently disclosed embodiments in any manner.
EXAMPLES
Example la and lb. P7C3-S16 and P7C3-S17: S- and R-1-(3,6-Dibromo-9H-carbazol-
9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol
Br Br
Br 0 # Br * *
N Ø_H
H
(R) 1a and 1b (s)
NH NH
* OMe * OMe
Representative Procedure 1.
Step 1. Synthesis of 3,6-Dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole (Epoxide 2-
A)
112
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 .
N
o
Following a literature procedure (Asso, V.; Ghilardi, E.; Bertini, S.;
Digiacomo, M.; Granchi, C.;
Minutolo, F.; Rapposelli, S.; Bort lato, A.; Moro, S. Macchia, M. ChemMedChem,
2008, 3, 1530-1534)
powdered KOH (0.103 g, 1.85 mmol) was added to a solution of 3,6-
dibromocarbazole (0.500 g, 1.54 mmol)
in DMF (1.5 mL) at ambient temperature and stirred for 30 min until dissolved.
Epibromohydrin (0.32 mL,
3.8 mmol) was added via syringe and the reaction was stirred at room
temperature overnight. Upon
completion, the solution was partitioned between Et0Ac and H20. The aqueous
layer was washed 3x with
Et0Ac, and the combined organics were washed with saturated aqueous NaC1,
dried over Na2504, filtered, and
concentrated in vacuo. The crude residue was recrystallized from Et0Ac/Hexane
to afford the desired product
(389 mg, 66%).
1H NMR (CDC13, 500 MHz) 6 8.10 (d, 2H, J = 2.0 Hz), 7.54 (dd, 2H, J = 2.0, 8.5
Hz), 7.31 (d, 2H, J =
8.5 Hz), 4.62 (dd, 1H, J = 2.5, 16.0 Hz), 4.25 (dd, 1H, J = 5.5, 16.0 Hz),
3.29 (m, 1H), 2.79 (dd, 1H, J = 4.0,
4.5 Hz), 2.46 (dd, 1H, J = 2.5, 5.0 Hz).
ESI m/z 381.0 ([M+1-1] , C15H12Br2NO requires 379.9)
Representative Procedure 2
Step 2. Synthesis of 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
Br
Br . =
NH
0 OMe
Following a literature procedure (Asso, V.; Ghilardi, E.; Bertini, S.;
Digiacomo, M.; Granchi, C.;
Minutolo, F.; Rapposelli, S.; Bortolato, A.; Moro, S. Macchia, M. ChemMedChem,
2008, 3, 1530-1534) m-
Anisidine (1.0 mL, 8.95 mmol) was added to a suspension of epoxide 2-A (3.02
g, 7.92 mmol) in cyclohexane
(73 mL). BiC13 (0.657 g, 2.08 mmol) was added and the mixture was heated to
reflux overnight. Upon
completion, the reaction was partitioned between Et0Ac and H20. The aqueous
layer was washed 3x with
Et0Ac, and the combined organics were washed with saturated aqueous NaC1,
dried over Na2504, filtered, and
113
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
concentrated in vacuo. The crude residue was purified by chromatography (Si02,
0-50% Et0Ac/Hexane) to
afford the desired alcohol as an opaque yellow solid (998 mg, 25%).
1H NMR (CDC13, 400 MHz) 6 8.12 (d, 2H, J = 1.6 Hz), 7.52 (dd, 2H, J = 2.0, 8.8
Hz), 7.32 (d, 2H, J =
8.8 Hz), 7.07 (dd, 1H, J = 8.0 Hz), 6.31 (dd, 1H, J = 2.4, 8.0 Hz), 6.21 (dd,
1H, J = 2.0, 8.0 Hz), 6.12 (dd, 1H, J
= 2.0, 2.4 Hz), 4.34-4.39 (m, 3H), 4.00 (br s, 1H), 3.71 (s, 3H), 3.30 (dd,
1H, J = 3.6, 13.2 Hz), 3.16 (dd, 1H, J
= 6.4, 13.2 Hz), 2.16 (br s, 1H).
13C NMR (CDC13, 100 MHz) 6 161.0, 149.2, 139.9 (2C), 130.4 (2C), 129.5 (2C),
123.8 (2C), 123.5
(2C), 112.8, 111.0 (2C), 106.7, 103.8, 99.8, 69.5, 55.3, 48.0, 47.4
ESI m/z 502.9 ([M+1-1] , C22H21Br2N202 requires 503.0)
Step 3. Synthesis of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-y1 3,3,3-trifluoro-
2-methoxy-2-phenylpropanoate
Br
Br 0 =
F3C ome
NL.504 Ph4-
0
HN
411 OMe
1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol (0.150 g,
0.298 mmol) was
dissolved in anhydrous dichloromethane (6 mL) and cooled to 0 C. Pyridine
(0.053 mL, 0.655 mmol) was
added, followed by S-(+)-a-methoxy-a-trifluoromethylphenylacetyl chloride (S-
Mosher's acid chloride, 0.083
mL, 0.446 mmol) and dimethylaminopyridine (0.004 g, 0.030 mmol). The reaction
was allowed to warm to
room temperature over 4 hours, after which it was quenched by addition of
saturated aqueous NaHCO3. The
mixture was extracted 3x with Et0Ac, and the combined organics were washed
with saturated aqueous NaC1,
dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue was
purified by chromatography
(Si02, 0-50% Et0Ac/Hexane) to afford a mixture of both possible esters and
both possible amides (-5:1
ester: amide ratio by 1H NMR, 132 mg, 64%). Separation of the mixture was
achieved using HPLC
(Phenomenex 5i02 Luna, 21x250 mm, 15% Et0Ac/Hexane, 16 mL/min; HPLC Retention
time: 25.6 min
(ester 1) and 41.2 min (ester 2).
Ester 1: 1H NMR (CDC13, 500 MHz) 6 8.11 (d, 2H, J= 2.0 Hz), 7.45 (dd, 2H, J=
8.5 Hz), 7.24 (m,
2H), 7.22 (m, 4H), 7.05 (t, 1H, J = 8.0 Hz), 6.32 (dd, 1H, J = 2.0, 8.0 Hz),
6.12 (dd, 1H, J = 2.0, 8.0 Hz), 6.05
(dd, 1H, J = 2.0, 2.5 Hz), 5.59 (m, 1H), 4.54 (d, 2H, J = 6.5 Hz), 3.71 (br s,
1H), 3.69 (s, 3H), 3.43 (m, 1H),
3.29 (ddd, 1H, J = 5.5, 13.5 Hz), 3.19 (s, 3H).
114
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Ester 2: 1H NMR (CDC13, 500 MHz) 6 8.08 (d, 2H, J = 2.0 Hz), 7.42 (dd, 2H, J =
2.0, 9.0 Hz), 7.28
(m, 2H), 7.24 (m, 4H), 7.04 (t, 1H, J = 8.0 Hz), 6.31 (dd, 1H, J = 2.0, 8.5
Hz), 6.11 (dd, 1H, J = 2.0, 8.0 Hz),
6.01 (dd, 1H, J = 2.0, 2.5 Hz), 5.63 (m, 1H), 4.49 (d, 2H, J = 6.5 Hz), 3.82
(dd, 1H, J = 5.5, 6.0 Hz), 3.66 (s,
3H), 3.42 (s, 3H), 3.39 (m, 1H), 3.28 (dd, 1H, J = 5.0, 13.5 Hz)
Step 4. Synthesis of S- and R-1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol
Br Br
Br 0 # Br 0 *
N
<0 Nv(:H
H
(R) la and lb (s)
NH NH
* OMe * OMe
Following a literature procedure (Abad, J-L.; Casas, J.; Sanchez-Baeza, F.;
Messeguer, A. J. Org.
Chem. 1995, 60, 3648-3656) ester 1 from example 3 (0.011 g, 0.015 mmol) was
dissolved in degassed Et20
(0.150 mL) and cooled to 0 C. Lithium aluminum hydride (1M in THF, 0.018 mL,
0.018 mmol) was added
via syringe and the reaction was stirred for 20 min. Upon completion by TLC
the reaction was quenched by
the addition of Me0H and stirred for 45 min. The mixture was partitioned
between Et0Ac and H20. The
aqueous layer was extracted 3x with Et0Ac, and the combined organics were
washed with saturated aqueous
NaC1, dried over Na2SO4, filtered, and concentrated in vacuo. The crude
residue was purified by
chromatography (Si02, 0-30% Et0Ac/Hexane) to afford the desired alcohol (4.7
mg, 64%).
(From Ester 1): [4) = +100 (c = 0.1, CH2C12); Example la
(From Ester 2): [a]r) = ¨14 (c = 0.1, CH2C12); Example lb
Example 2. P7C3-S5: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-iminopyridin-1(2H)-
yl)propan-2-ol
Br
Br * .
N
OH
,--N
----L/NH
Example 2 was prepared following Representative Procedure 2, except with a
reaction time of 2 days
at 80 C. The crude product was used without further purification.
115
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) d= 8.14 (2H, J = 1.9 Hz), 7.55 (dd, 2H, J = 1.9, 8.8
Hz), 7.35 (d, 2H, J =
8.7 Hz), 6.83 (t, 1H, J = 7.6 Hz), 6.37 (d, 1H, J = 6.8), 6.32 (d, 1H, J = 9.1
Hz) , 5.65 (t, 1H, J = 6.7 Hz), 4.39
(dm, 5H), 3.54 (d, 1H, J = 13.9 Hz). MS (ESI), m/z: found 473.9 (M+1) ([M+1]+
for C20H18Br2N30 requires
474.0)
Example 3a. P7C3-S7: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylthio)propan-2-
01
0
S
r(OH
N
* *
Br Br
Benzenethiol (30 Ti, 0.29 mmol) was added to a solution of 3,6-dibromo-9-
(oxiran-2-ylmethyl)-9H-
carbazole (epoxide 2-A, 101.6 mg, 0.27 mmol) in 5.0 ml Me0H at r.t. The
reaction mixture was heated to 80
C and stirred overnight at the same temperature. The reaction was monitored by
lc/ms for the consumption of
SM. The reaction was cooled, diluted with ethyl acetate and washed with water
and brine. The organic layer
was dried over Na2504, filtered and condensed.
1H NMR (CDC13, 400 MHz) A 8.03 (d, 2H, J = 2.1 Hz), 7.48 (dd, 2H, J = 2.0, 8.7
Hz), 7.33-7.20 (m,
7H), 4.33 (dd, 1H, J = 4.3, 14.9 Hz), 4.20 (dd, 1H, J = 6.9, 14.9 Hz), 4.00-
4.12 (m, 1H), 3.05 (dd, 1H, J = 5.3,
13.9 Hz), 2.93 (dd, 1H, J = 7.2, 13.9 Hz), 2.51 (bs, 1H); 13C NMR (CDC13, 126
MHz) 6139.9, 134.5, 130.4,
129.6, 129.4, 127.4, 123.8, 123.4, 112.7, 111.1, 69.3, 48.1, 39.4; MS (ESI),
m/z: found: 505.9 [M+0-1]-
([M+0-1]- for C21H17Br2NOS requires 504.9; (oxidation occurred under MS
conditions; NMR not consistent
with sulfoxide)
Example 3b. P7C3-S39: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-01
B
Br r
*4
N
yo =
OH
Following Representative Procedure 1, the title compound of Example 3b was
prepared from
dibromocarbazole and phenoxymethyloxirane in 61% yield.
1H NMR (CDC13, 400 MHz) 6 8.14 (d, 2H, J = 1.9 Hz), 7.51 (dd, 2H, J = 1.9, 8.7
Hz), 7.36 (d, 2H, J =
8.8 Hz), 7.127-7.32 (m, 2H), 7.00 (t, 1H, J = 7.3 Hz), 6.87 (dd, 2H, J = 0.8,
8.9 Hz), 4.58 (dd, 1H, J=7.9, 16.7
116
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Hz), 4.41-4.49 (m, 2H), 4.00 (dd, 1H, J-4.4, 9.6 Hz), 3.89 (dd, 1H, J=4.5, 9.5
Hz), 2.38 (d=1H, J=5.7Hz). MS
(ESI), m/z: 517.9 [M+HCOO] ([M+HCOO]- for C21H17Br2NO2 requires 518.0
Example 3c. P7C3-S27: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylsulfmyl)propan-
2-01
Br Br
crs
oil
OH
An aqueous solution of NaI04(5.14 g) was added to silica gel (20 g) and shaken
until a free-flowing
solid was obtained. Thio-ether (1-(3,6-dibromo-9H-carbazol-9-y1)-3-
(phenylthio)propan-2-ol, (0.0120 g,
0.0244 mmol) and NaI04/silica gel (0.1018 g NaI04, 0.122 mmol) were suspended
in CH2C12 (1 mL). The
white suspension was heated to 50 C in a sealed vial for 4 hours until TLC
showed complete disappearance of
starting material. The reaction mixture was subjected to silica gel
chromatography using Hexanes/Et0Ac (1:9)
to afford 0.0081g white solid as product, yield 65.4% as a 1:1 mixture of
diastereomers.
1H NMR (CDC13, 400 MHz) 6ppm = 2.39 (dd, J=13.7, 1.7 Hz, 1 H diastereomer A)
2.83 (dd, J=13.2,
2.9 Hz, 1 Dias. B) 2.97 (dd, J=13.2, 8.6 Hz, 1 H Diast. B) 3.15 (dd, J=13.7,
9.3 Hz, 1 H Diast. A) 3.90 (d, J=
1.7 Hz, 1 H Dias. B) 3.96 (d, J= 2.6 Hz, 1 H Diast. A), 4.24 (dd, J = 15.0,
6.3 Hz, 1H Dias A), 4.30 (dd, J =
15.2, 6.7, 1H Diast. B), 4.35 (dd, J = 15.2, 6.0 Hz, 1 H Diast. B), 4.45 (dd,
J = 15.1, 6.4 Hz, 1H Diast. B), 4.65
-4.55 (m, 1 H Diast. A) 4.87 -4.76 (m, 1 H Diast. B) 7.16 (d, J= 8.7 Hz, 2 H
Diast. A) 7.34 (d, J= 8.8 Hz,
2H Diast B) 7.60 -7.30 (m, 7 H Diast A + 7 H Dast. B) 8.08 (d, J= 1.9 Hz, 2 H
Diast. A) 8.13 (d, J= 1.9 Hz,
2 H Diast B). MS (ESI) m/z: 549.9 [M + HCOO] ([M+CH00]- for C21H17Br2NO2S
requires 549.9).
Example 3d. P7C3-S28: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-
(phenylsulfonyl)propan-2-01
Br Br
= 11*
cr, 410
0 1-10/1 1.1
To a solution of thio-ether (1-(3,6-dibromo-9H-carbazol-9-y1)-3-
(phenylthio)propan-2-ol, (0.0113 g,
0.0230 mmol) in 0.5 mL CH2C12, a solution of mCPBA (ca. 77% pure, 0.0129 g,
0.0575 mmol) in 0.5 mL
CH2C12 was added dropwise. The mixture was stirred at room temperature
overnight. The crude reaction
mixture was neutralized by 9 ml. Et3N and stirred for 30 min then diluted with
30 mL Et0Ac and washed with
saturated NaHCO3 3 x 30 mL and brine 1 x 30 mL. The organic layer was dried
over anhydrous Na2504 and
evaporated to afford the crude product, which was subjected to silica gel
chromatography using
Hexanes/Et0Ac (3:7) to afford white solid as product (0.0120 g, yield 99.7%).
117
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6ppm 3.15 (dd, J=14.2, 3.0 Hz, 1 H) 3.21 -3.31 (m, 2
H) 4.38 (d, J=6.3
Hz, 2 H) 4.60 - 4.76 (m, 1 H) 7.25 - 7.31 (m, 2 H) 7.47 - 7.56 (m, 4 H) 7.60 -
7.70 (m, 1H) 7.79 (dd, J=8.4, 1.2
Hz, 2 H) 8.11 (d, J=1.9 Hz, 2 H);MS (ESI) m/z: 565.9 [M + HC00]; 543.7 [M +
Na]' ([M+HCOOT for
C21H17Br2NO3S requires 595.9; [M+Na]+ requires 543.9).
Example 4. P7C3-S9: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(3-
methoxyphenypacetamide
1:::
N i& 1:::
r(OH
N
0 *
Br Br
Following a literature procedure (Morcuende et al., J. Org. Chem. 1996, 5264-
5270) triethylamine (14
T1, 0.10 mmol) and acetyl chloride (8 Tl, 0.11 mmol) were added to a
heterogeneous mixture of 143,6-
dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol (53 mg, 0.11
mmol) and dibutyltin oxide
(5.5 mg, 0.022 mmol) in anhydrous toluene (1.5 m1). The reaction vessel was
purged with nitrogen, sealed and
heated under microwave radiation to 150 C for 9 minutes. The reaction was
monitored by lc/ms and all SM
had been consumed. The heterogeneous solution was filtered under vacuum to
yield a white solid. The crude
product was used without purification.
1H NMR (CDC13, 500 MHz) A 8.09 (2H, J = 1.6 Hz), 7.52 (dd, 2H, J = 1.8, 8.7
Hz), 7.29 (d, 2H, J =
8.8 Hz), 7.26 (t, 1H, J = 8.2 Hz), 6.86 (dd, 1H, J = 2.5, 8.4 Hz), 6.68 (dd,
1H, J = 1.3, 7.7 Hz), 6.62 (s, 1H,),
4.33-4.40 (m, 1H), 4.29 (dd, 2H, J = 2.6, 6.0 Hz), 3.94 (d, 1H, J = 4.1 Hz),
3.76 (s, 3H), 3.51 (dd, 1H, J = 2.3,
14.0 Hz), 1.9 (s, 3H);
13C NMR (CDC13, 126 MHz) 6 173.6, 160.9, 144.5, 139.9, 131.0, 129.4, 123.8,
123.4, 119.7, 113.9,
113.5, 112.6, 111.1, 70.9, 55.7, 55.2, 46.0, 22.8.
MS (ESI), m/z: 544.9 (M+1) ([M+1] for C24H22Br2N203 requires 545.0)
Example 5. P7C3-S12: 5-((3,6-dibromo-9H-carbazol-9-yl)methyl)-3-(3-
methoxypheny1)-oxazolidin-2-
one
0
0-4
(L/N 411,
N O-
S .
Br Br
118
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Methyl chloroformate (10 Ti, 0.13 mmol) was added to a stirring solution of jn-
128-186 (55.0 mg,
0.11 mmol) and indium powder (3.5 mg, 0.030 mmol) in acetonitrile (3.0 ml),
and the reaction mixture was
stirred overnight at r.t. An additional 3.1 mg (0.027 mmol) of indium and 20
T1(2.6 eq.) of methyl
chloroformate were added. After several hours, the reaction was diluted with
ethyl actetate, and washed with
water and then brine. The organic layer was dried over Na2SO4, filtered and
concentrated. The methyl
carbonate was purified via flash chromatography in 20-40% ethyl
acetate/hexanes. Sodium methoxide (3.0
ml) was added to a solution of carbonate (21.3 mg, 0.038 mmol) and methanol
(1.0 m1). After an hour at
ambient temperature the solution was diluted with water and extracted with
ethyl acetate. The organic layer
was washed with water and brine and condensed.
1H NMR (CD3C0CD3, 500 MHz) A 8.40 (s, 2H), 7.78 (d, 2H, J = 8.5 Hz), 7.64 (d,
2H, J = 8.9 Hz),
7.23-7.28 (m, 2H), 7.05 (d, 1H, J = 8.3 Hz), 6.70 (d, 1H, J = 8.3 Hz), 5.24-
5.31 (m, 1H), 5.00 (dd, 1H, J = 7.9,
15.7 Hz), 4.91 (dd, 1H, J = 3.2, 15.8 Hz), 4.38 (t, 1H, J = 9.3 Hz), 4.05 (m,
1H), 3.78 (s, 3H);
13C NMR (CDC13, 126 MHz) 6 160.4, 153.9, 140.3, 140.2, 129.8, 129.4, 124.0,
123.5, 112.4,112.1,
110.3, 109.0, 104.4, 71.9, 54.9, 47.9, 46.6.
MS (ESI), m/z: 528.9 (M+1)'. ([M+1]+ for C23H19Br2N203 calculated 529.0)
Example 6a. P7C3-S10: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-
methoxyaniline (also
designated as "P7C3A20")
0
0
rENH
F
N
* .
Br Br
Representative Procedure 3: Epoxide opening with Ns-protected anilines.
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(3-methoxyphenyl)-4-
nitrobenzenesulfonamide
Br Br
e .
N
y,N1, elo OMe
OH /5-:: ¨
0/ =
NO2
119
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
A heterogeneous mixture of N-(4-methoxypheny1)-4-nitrobenzenesulfonamide
(100.2 mg, 0.32 mmol)
in toluene (2.5 ml, 0.13 M) under a N2 atmosphere was cooled in a dry
ice/acetone bath before dropwise
addition of n-butyllithium (200u1 of 1.78 M in hexanes, 0.36 mmol). The
reaction was stirred at -78 C for 10
minutes before addition of carbazole epoxide 2-A. The heterogeneous mixture
was stirred at room
temperature for 5 minutes before heating at 100 C for 48 hours. The cooled
reaction was diluted with Et0Ac
and washed three times with 5% acetic acid solution, followed by a brine wash.
The organic layer was dried
over Na2SO4, filtered and condensed. The crude mixture was purified in 100%
dichloromethane. Yield=88%.
1H NMR (CDC13, 400 MHz) 6 8.23(d, 2H, J= 8.5 Hz), 8.06 (d, 2H, J= 1.9 Hz),
7.65 (d, 2H, J=8.5 Hz),
7.46, (dd, 2H, J=8.6, 1.9 Hz), 7.22 (d, 2H, J=8.8 Hz), 6.94 (d, 2H, 8.8 Hz),
6.83 (d, 2H, 9.1 Hz), 4.44 (dd, 1H,
J=14.9, 3.6 Hz), 4.26-4.34 (m, 1H), 4.17-4.24 (bs, 1H), 3.81 (s, 3H), 3.62-
3.75 (m, 2H). MS (ESI), m/z: 732.0
[(M+HC00-); C28H23Br2N306S (M) requires 687]
Representative Procedure 4: Fluorination of Secondary Alcohol
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-N-(3-methoxyphenyl)-4-
nitrobenzenesulfonan
Br Br
* *
ILrN a
OMe
µS---
F //
0 *
NO2
An oven dried 20 ml scintillation vial containing N-(3-(3,6-dibromo-9H-
carbazol-9-y1)-2-
hydroxypropy1)-N-(3-methoxypheny1)-4-nitrobenzenesulfonamide (18.3 mg, 0.027
mmol; see representative
procedure 3 above) was purged with N2 and charged with anhydrous
dichloromethane (1.5 ml, 0.018 M). The
sealed vial was cooled in a dry ice acetone bath before the dropwise addition
of diethylaminosulfur trifluoride
(DAST, 7 ul, 0.053 mmol). The reaction temperature was maintained at -78 C
for an hour and then slowly
warmed to room temperature and stirred overnight. The reaction was quenched
with 2.0 ml of saturated
NaHCO3 solution and diluted with 6 ml CH2C12 and extracted three times. The
combined organics were dried
over Na2504, filtered and condensed. Crude product carried forward.
Quantitative yield.
Alternatively, morpholinosulfur trifluoride (MORPHO-DAST) can be used at rt.
1H NMR (CDC13, 400 MHz) 6 8.28 (d, 2H, J= 8.0 Hz), 8.13 (s, 2H), 7.72 (d, 2H,
J=8.7 Hz), 7.54, (d,
2H, J=8.0 Hz), 7.21 (d, 3H, J=8.1 Hz), 6.89 (dd,1H, 8.3, 2.4 Hz), 6.67 (t, 1H,
J=2.0 Hz), 6.55 (d, 1H, J=8.0
Hz) 4.93 (m, 1H), 4.43-4.68 (m, 2H), 4.20 (t, 1H, J=6.2 Hz), 3.81 -3.99(m,
2H), 3.75 (s, 3H).
MS (ESI), m/z: calculated 688.96, found 733.9 (M+HC00-) .
120
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Representative Procedure 5: nosyl group deprotection (see Fukuyama, T.; Jow,
C.-K.; Cheung, M. Tetrahedron
Lett. 1995, 36, 6373-6374)
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-methoxyaniline
Br Br
. *
N
H,N 411 ome
H
F
To a vial containing N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-N-(3-
methoxyphenyl)-4-
nitrobenzenesulfonamide (21.0 mg, 0.030 mmol; see representative procedure 4)
was added lithium hydroxide
(3.2 mg, 0.134 mmol), dimethylformamide (0.5 ml, 0.06 M) and mercaptoacetic
acid (4.2 ul 0.060 mmol).
After stirring at rt for lh the reaction mixture was diluted with Et0Ac and
washed sequentially with water,
saturated sodium bicarbonate solution, water (3x) and brine. The organic layer
was dried over Na2SO4, filtered
and condensed. The crude reaction mixture was purified in 30% Et0Ac/hexanes
(+0.2% TEA), with 13.6 mg
isolated. Yield=88%
Additional Representative Procedure
DAST [(Et2NSF3) 0.12 ml, 0.916 mmol] was added dropwise to a solution of 1-
(3,6-dibromo-9H-
carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-ol (0.102 g, 0.203 mmol) in
6.0 ml of anhydrous DCM at -
78 C. The reaction was stirred at -78 C for one hour before being slowly
warmed to 0 C over 5 hours. The
reaction was quenched by addition of phosphate buffer (pH=8) and extracted
with DCM. The aqueous phase
was extracted twice with 10 ml DCM. The combined organics were dried over
Na2SO4, filtered and
concentrated. The crude reaction material was purified by flash chromatography
on Si02 (20%
Et0Ac/hexanes/0.2%TEA). Fractions containing the desired fluorinated product
were further purified with
40% Et0Ac/hexanes (+ 0.1%TEA). Isolated 5.7 mg desired product.
Analytical Data for the title compound of Example 6a
1H NMR (CDC13, 500 MHz) A 8.16 (2H, J = 2.0 Hz), 7.56 (dd, 2H, J = 1.9, 8.7
Hz), 7.31 (d, 2H, J =
8.6 Hz), 7.11 (t, 1H, J = 8.1 Hz), 6.36 (dd, 1H, J = 2.2, 8.1 Hz), 6.23 (dd,
1H, J = 2.0, 8.0 Hz), 6.15 (t, 1H, J =
2.3 Hz), 5.11 (dddd, 1H, J= 4.6, 5.8, 10.4, 47.7 Hz), 4.60 (m, 2H), 4.39 (dm,
2H), 3.95 (t, 1H, J = 6.3 Hz),
3.75 (s, 3H)
MS (ESI), m/z: 504.9 (M+1) . ([M+1]+ for C22H19Br2FN20 calculated 505.0)
121
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Gram-scale synthesis of P7C3-S10, also known as P7C3A20
3,6-dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole
Br Br
fk 10
N
A duplicate set of reactions were set up. Solutions of 3,6-dibromocarbazole
(49.61 and 51.98 g, 152.6
and 159.9 mmol respectively) and crushed potassium hydroxide pellets (11.1 and
10.6 g, 197.8 and 188.9
mmol, respectively) in dimethylformamide (1 L each) were stirred for an hour
before the addition of
epibromohydrin (32 and 35 ml, 386.6 and 422.9 mmol respectively). The
reactions were stirred overnight.
Each reaction was worked up portionwise by dilution with Et0Ac, and washed
several times with water and
then brine. The organic layer was dried over Mg504, filtered and condensed.
The off-white solid was washed
with minimal Et0Ac to give 95.2 g of epoxide in 80% yield.
1H NMR (CDC13, 400 MHz) 6 8.15 (d, J= 1.9 Hz, 2H), 7.58 (dd, J= 8.6, 2.0 Hz,
2H), 7.35 (d, J= 8.7
Hz, 2H), 4.66 (dd, J= 16.0, 2.7 Hz, 1H), 4.29 (dd, J= 15.9, 5.1 Hz, 1H), 3.33
(ddd, J= 6.7, 5.2, 2.8 Hz, 1H),
2.82 (t, J= 4.3 Hz, 1H), 2.50 (dd, J= 4.7, 2.6 Hz, 1H).
1,1,1-trifluoro-N-(3-methoxyphenyl)methanesulfonamide
HNS02CF3
OMe
A solution of trifluoromethanesulfonic anhydride (45 ml, 26.7 mmol) in
methylene chloride (250 ml)
were added dropwise to an ice chilled solution of m-anisidine (25 ml, 22.3
mmol) and triethylamine (39 ml,
28.0 mmol) in methylene chloride (1.25 L). The reaction was stirred overnight
at rt. Workup was performed
portionwise. Each of the two portions was basified by addition of 250 ml of
2.5 N NaOH solution and 625 ml
Me0H. The aqueous was extracted thrice (100 ml each) with methylene chloride
to remove any unreacted
aniline or doubly triflated product. The aqueous phases were combined
,acidified to pH 2 with 18% HC1, and
again extracted with methylene chloride three times. The organic layer is
dried over Mg504, filtered and
condensed to give 17.69 g of brown solid in 77% yield.
1H NMR (CDC13, 400 MHz) 67.48-7.13 (m, 1H), 6.97-6.61 (m, 3H), 3.82 (s, 3H).
MS (ESI), m/z: calculated 255.21, found 255.9 (M+1) .
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide (P7C3-S244)
122
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br Br
= .
N
N lei OMe
OH µSO2CF3
N-butyllithium (2.5 M in hexanes, 48 ml) was added dropwise to an ice-cooled
solution of 1,1,1-
trifluoro-N-(3-methoxyphenyl)methanesulfonamide (22.07 g, 86.5 mmol) in dry
dioxane (145 ml) over a 40
minute period. The solution was then stirred at rt for 15 minutes before
addition of 3,6-dibromo-9-(oxiran-2-
ylmethyl)-9H-carbazole (25.05 g, 65.7 mmol), followed by heating at 90 C for
an hour. These conditions were
optimized to maximize conversion while minimizing formation of an aziridene by-
product. The reaction was
allowed to cool to rt then diluted with 1.2 L ethyl acetate and washed several
times with water and finally
brine. The organic layer was dried over MgSO4, filtered and condensed to give
an orange viscous mixture. To
this mixture was added 150 ml of 60% methylene chloride/hexanes, and the
solution was then concentrated to
generate a yellow foam (presumably this procedure helps remove residual ethyl
acetate and/or dioxane). A
further 150 ml of 60% methylene chloride/hexanes was added and stirred
overnight. The mixture was filtered
and washed several times with 60% methylene chloride/hexanes until the solid
was white giving 20.1 g of 99%
purity. A second crop gave 2.98 g in 91% purity. Filtrates and washings were
combined and found to contain a
2:2.6:1 mixture of SM: product: aziridene by-product. This mixture was
subjected to amination conditions by
heating the mixture (24 g in approximately 2 equal portions) in ammonia in
methanol (7N, 11 and 8 ml,
respectively) at 100 C in sealed pressure tubes overnight. Epoxide SM is
converted to 13-hydroxy amine
(MacMillan et al., J. Am. Chem. Soc. 2011, /33, 1428), which aids
chromatographic purification. Column
chromatography in 80% DCM/hexanes gave a further 9.7 g of product with an
overall yield of 32.78 g and
78%.
1H NMR (CDC13, 400 MHz) 6 8.13 (d, J= 1.9 Hz, 2H), 7.54 (dd, J= 8.7, 1.9 Hz,
2H), 7.33 (t, J = 8.1
Hz, 1H), 7.22 (d, J= 8.7 Hz, 2H), 6.95 (dd, J= 8.4, 2.3 Hz, 2H), 6.88 (s, 1H),
4.56 ¨4.10 (m, 4H), 3.99 (m,
1H), 3.81 (s, 3H), 1.98 (d, J= 4.2 Hz, 1H).
MS (ESI), m/z: calculated 633.94, found 678.6 (M+HC00)- .
3,6-dibromo-9-((1-(3-methoxyphenyl)aziridin-2-yl)methyl)-9H-carbazole
Br Br
O .
N
N 1 OMe
1H NMR (400 MHz, CDC13) 6 7.94 (d, J= 1.9 Hz, 2H), 7.40 (dd, J= 8.7, 1.9 Hz,
2H), 7.26 (d, J= 8.7
Hz, 2H), 6.84 (t, J= 8.1 Hz, 1H), 6.31 (dd, J= 8.2, 2.4 Hz, 1H), 6.12 ¨5.94
(m, 1H), 5.84 (t, J= 2.2 Hz, 1H),
4.42 (dd, J= 15.4, 2.8 Hz, 1H), 3.94 (dd, J= 15.4, 8.0 Hz, 1H), 3.33 (s, 3H),
2.22 (dq, J= 8.7, 3.0 Hz, 1H),
2.16 (d, J= 3.3 Hz, 1H), 2.02 (d, J= 6.3 Hz, 1H).
123
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
MS (ESI), m/z: calculated 483.98, found 484.7 (M+1) .
N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide (P7C3-S241)
Br Br
* IP
N
YN el OMe
F µSO2CF3
Morpho-Dast (14.0 ml, 115 mmol) was added to a solution of N-(3-(3,6-dibromo-
9H-carbazol-9-y1)-2-
hydroxypropy1)-1,1,1-trifluoro-N-(3-ethoxyphenyl)methanesulfonamide (20.6 g,
32.4 mmol) in anhydrous
methylene chloride (315 ml) and stirred overnight. The solution, in a water
bath at ambient temperature, was
neutralized by dropwise addition of 375 ml saturated bicarbonate solution. The
biphasic mixture was extracted
with methylene chloride twice. The combined organics were dried over Mg504,
filtered and condensed to give
21.5g of off-white foam in quantitative yield.
1H NMR (400 MHz, CDC13) 6 8.15 (d, J= 1.9 Hz, 2H), 7.56 (dd, J= 8.7, 1.9 Hz,
2H), 7.32 (t, J= 8.2
Hz, 1H), 7.21 (d, J= 8.6 Hz, 2H), 6.99 ¨ 6.90 (m, 2H), 6.86 (m, 1H), 5.08 ¨
4.86 (dm, 1H), 4.57 ¨ 4.44 (m,
2H), 4.09 (m, 2H), 3.79 (s, 3H).
MS (ESI), m/z: calculated 635.93, found 680.6 (M+HCOOT .
P7C3-S10 (P7C3A20): N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-
methoxyaniline
Br Br
= 0
N
HN lel OMe
H
F
A two neck flask containing N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropy1)-1,1,1-trifluoro-N-
(3-methoxyphenyl)methanesulfonamide (5.00 g, 7.83 mmol) was purged with N2
before addition of degassed
xylene (52.0 m1). The solution was cooled in a dry-ice acetone bath before the
dropwise addition of Red-Al
(sodium bis(2-methoxyethoxy)aluminum hydride solution, 65% wt in toluene, 11.0
ml, 36.1 mmol) during
which the internal temperature was maintained between -50 to -40 C. The cold
bath was removed
immediately upon completion of Red-Al addition. The reaction was allowed to
warm slowly to about -23 C at
which time the reaction flask was transferred to a heating block. The reaction
flask was heated until the
internal temperature was 59.3-62.0 C. Heating was continued for an hour, and
then the mixture was allowed
to cool to ambient temperature over 30 minutes. Analysis by HPLC/MS-ESI
revealed the following: 92%
consumption of SM, 75% product (N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropy1)-3-methoxyaniline),
124
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1% carbazole and 7% des-bromo decomposition products, 3% N-(3-(3,6-dibromo-9H-
carbazol-9-y1)-2-
hydroxypropy1)-1,1,1-trifluoro-N-(3-methoxyphenyl)methanesulfonamide and less
than 5% elimination
products (P7C3-S179). The reaction mixture was diluted with EtOAc and washed
with water until white solid
Al salts were no longer observed. The organic layer was then washed with 6M
HC1 a few times until yellow
precipitate formed. The hydrochloride salt was filtered to give 3.60 g (85%
yield). This salt formation removes
the carbazole decomposition product, unreacted SM and some elimination
products from the crude reaction
mixture. The salt was free-based by vigorously stirring in a 1:1 mixture of
methylene chloride and saturated
bicarbonate solution until a translucent two-phase mixture was obtained. The
organic layer was separated, and
the aqueous phase was extracted 3 times with methylene chloride. The combined
organics were dried over
Mg504, filtered and condensed to give a solid that contained the des-
brominated product as a minor impurity
(ca. 3%). The solid was washed several times by stirring successively
overnight in 40% Et20/hexanes and 30%
DCM/hexanes and filtering the solid. For the final purification, solids from 3
reactions (19.5 g triflate SM
total) were combined and assessed at 96% purity. The solid was stirred in 30%
DCM/hexanes overnight and
filtered to give 10.6 g of product in 98% purity and 69% yield.
1H NMR (CDC13, 500 MHz) 6 8.16 (d, J= 2.0 Hz, 2H), 7.56 (dd, J= 1.9, 8.7 Hz,
2H), 7.31 (d, J= 8.6
Hz, 2H), 7.11 (t, J= 8.1 Hz, 1H), 6.36 (dd, J= 2.2, 8.1 Hz, 1H), 6.23 (dd, J=
2.0, 8.0 Hz, 1H), 6.15 (t, J= 2.3
Hz, 1H), 5.11 (dddd, J= 4.6, 5.8, 10.4, 47.7 Hz, 1H), 4.60 (dm, 2H), 3.95 (t,
J= 6.3 Hz, 1H), 3.75 (s, 3H),
4.39 (dm, 2H).
13C NMR (CDC13, 100.5 MHz) 6 161.0, 148.6, 139.6, 130.4, 129.6, 123.9, 123.5,
112.9, 110.6 (d, 4J=
2.0 Hz), 106.5, 103.9, 99.7, 90.7 (d, 1J= 176.9 Hz), 55.3, 45.6 (d, 2J= 22.1
Hz), 45.1 (d, 2J= 25.1 Hz),
MS (ESI), m/z: calculated 503.98, found 504.9 (M+1) .
(E)-N-(3-(3,6-dibromo-9H-carbazol-9-yl)prop-1-en-1-y1)-1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide (P7C3-S179).
Br Br
fit IP
N
N el
OMe
µSO2CF3
1H NMR (CDC13, 400 MHz) 6 8.13 (d, J= 1.9 Hz, 2H), 7.55 (dd, J= 8.6, 2.0 Hz,
2H), 7.32 (t, J= 8.2
Hz, 1H), 7.21 (d, J= 8.7 Hz, 2H), 7.01 (d, J= 13.4 Hz, 1H [olefin CH]), 6.98 -
6.93 (m, 1H), 6.80 (dd, J= 7.9,
1.9 Hz, 1H), 6.73 (t, J= 2.3 Hz, 1H), 4.83 (d, J= 6.7 Hz, 2H), 4.76 (ddd, J=
12.8, 7.2, 5.4 Hz, 1H), 3.75 (s,
3H).
MS (ESI), m/z: calculated 615.93, found 660.5 (M+HC00)- .
125
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 6b. P7C3-S11: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-3-
methoxy-N-
methylaniline
B
Br r
*
y, OMe
F I
The title compound of Example 6b was prepared according to the procedure
described in
Representative Procedure 4 except using 1-(3,6-dibromo-9H-carbazol-9-y1)-3-((3-
methoxyphenyl)(methyl)-
amino)propan-2-ol (see Example 23)
1H NMR (CDC13, 500 MHz) 6 8.13 (d, 2H, J = 1.9 Hz), 7.54 (dd, 2H, J = 1.9, 8.8
Hz), 7.23 (d, 2H, J =
8.7 Hz), 7.12 (t, 1H, J = 8.2 Hz), 6.32 (dd, 1H, J = 2.2, 8.1 Hz), 6.26 (dd,
1H, J = 2.3, 8.0 Hz), 6.17 (t, 1H, J =
2.4 Hz), 5.10 (dddd, 1H, J= 4.6, 6.4, 10.7, 48.5 Hz), 4.37-4.48 (m, 2H), 3.72
(s, 3H), 3.60-3.71 (m, 1H), 3.53
(td, 1H, J= 6.9, 15.9 Hz), 2.99 (s, 3H).
MS (ESI), m/z: 518.9 [M+1] ([M+H]+ for C23H21Br2FN20 requires 519Ø)
Example 7a. P7C3-S3: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)-
propan-2-one
0
* *
Br Br
Trietheylamine (1.65 ml, 11.8 mmol ) was added to a stirring solution of 1-
(3,6-dibromo-9H-carbazol-
9-y1)-3-(3-methoxyphenylamino)propan-2-ol (1.02 g, 2.02 mmol) in DMSO (21 m1).
The solution was stirred
for 30 minutes before addition of sulfur trioxide pyridine complex (0.659 g,
4.14 mmol). After stirring
overnight, additional triethylamine (1.0 ml, 7.17 mmol) was added, followed by
sulfur trioxide pyridine
complex (0.663 mg, 4.17 mmol) an hour later. After stirring for 1 h, the
orange solution was diluted with - 150
ml ethyl acetate and washed several times with water and then brine. The
organic layer was dried over
Na2504, filtered and concentrated to yield brown foam. Flash chromatography on
5i02 100% (CH2C12+
0.2%TEA) provided a higher Rf ketone (thioether, 18%) and a lower Rf ketone
(Yield= 40%).
Major product: 1H NMR (CDC13, 400 MHz) 6 8.18 (2H, J = 1.9 Hz), 7.56 (dd, 2H,
J = 1.9, 8.7 Hz),
7.11 (d, 2H, J = 8.8 Hz), 7.06 (t, 1H, J = 8.1 Hz), 6.30 (dd, 1H, J = 2.3, 8.2
Hz), 6.07 (dd, 1H, J = 2.0, 8.0 Hz),
6.11 (t, 1H, J = 2.2 Hz), 5.08 (s, 2H,), 4.41 (t, 1H, J = 4.8 Hz), 3.90 (d,
2H, J = 5.1 Hz), 3.72 (s, 3H)
13C NMR (CDC13, 126 MHz) 6 = 202.9, 161.1, 147.9 (2 C), 139.5, 130.6 (2 C),
129.9 (2 C), 124.1(2
C), 123.9(2 C), 113.5, 110.1(2 C), 103.7, 99.3, 55.4, 51.9, 51Ø
MS (ESI), m/z: 500.9 (M+1) ([M+1]+ for C22H18Br2N202 requires 501.0)
126
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 7b. P7C3-S4: 3-(3,6-dibromo-9H-carbazol-9-y1)-1-(3-methoxyphenylamino)-
1-
(methylthio)propan-2-one
Br
101
Br *, Nj\....._
H
N
s\ * 0\
The title compound of Example 7b was obtained as a minor product in the
preparation of the title
compound of Example 7a.
1H NMR (CDC13, 400 MHz): 6 8.16 (d, 2H, J= 2.0 Hz), 7.55 (dd, 2H, J= 1.7, 8.8
Hz), 7.25 (d, J = 8.8
Hz, 2H), 7.12 (t, 1H, J= 8.4 Hz), 6.39 (dd, 1H, J= 2.2, 8.2 Hz), 6.33 (dd, 1H,
J= 2.2, 8.0 Hz), 6.29 (t, 1H, J =
2.2 Hz), 5.50 (d, 1H, J= 18.0 Hz), 5.22 (d, 1H, J= 18.4 Hz), 5.25 (d, J = 8.0
Hz, 1H), 4.50 (d, J= 8.0 Hz, 1H,
exchangeable), 3.76 (s, 3H), 1.74 (s, 3H)
13C NMR (CDC13, 126 MHz) 6 = 193.2, 160.9, 143.9 (2 C), 139.8(2C), 130.4,
129.8(2C), 124.1,
123.7(2C), 113.4(2C), 110.3(2C), 107.8, 104.7, 101.0, 60.3, 55.4, 48.9, 9.0
ESI m/z 498.9 [M-SMe+H]+ ([M-SMe+H]+ for C23H20Br2N202S requires 499Ø
HRMS m/z: 546.9675 [M+H]+ ([M+H]+ for C23H20Br2N202S requires 545.9612.
Example 8. P7C3-S13: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-methoxypropy1)-3-
methoxyaniline
0
(10
r(NH
0
N
* .
Br Br
Sodium hydride (9.0 mg, 0.23 mmol) was added to a stirring solution of 1-(3,6-
dibromo-9H-carbazol-
9-y1)-3-(3-methoxyphenylamino)propan-2-ol (99.3 mg, 0.20 mmol) in DMF 0.5 ml,
0.39 M). The solution was
stirred at room temperature for about 70 minutes before the dropwise addition
of a solution of methyl iodide
(14 ml. 0.22 mol) in DMF (1.0 m1). The reaction was monitored by lc/ms for the
consumption of SM and the
appearance of 0 and N-methyl products. After 2.5 hours of stirring at r.t,
conversion was about 30% and about
5% N-methyl product had formed. The reaction was stopped when an increase of N-
Me to 0-Me had been
observed and conversion was about 50%. The brown solution was diluted with
ethyl acetate and washed
several times with water and finally brine. The organic layer was dried over
Na2504, filtered and condensed.
The mixture was purified by preparative TLC 30% Et0Ac/hexanes.
127
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) A 8.13(s, 2H), 7.51 (dd, 2H, J = 1.8, 8.8 Hz), 7.31
(d, 2H, J = 8.7 Hz),
7.09 (t, 1H, J=8.2 Hz), 6.33 (dd, 1H, J= 2.3, 8.3 Hz), 6.21 (dd, 1H, J=2.1,
8.0 Hz), 6.12 (m, 1H), 4.42 (m, 1H),
4.03 (bs, 1H), 3.85 (m, 1H), 3.74(s, 3H), 3.29 (s, 3H), 3.09(m, 2H)
13C NMR (CDC13, 126 MHz) 6 161.0, 149.4, 139.8, 130.4, 129.5, 123.8, 123.5,
112.7, 110.9, 106.7,
103.6, 99.7, 78.2, 58.3, 55.3, 45.3, 44.3.
MS (ESI), m/z: 516.9 (M+1) ([M+1]+ for C23H22Br2N202 requires 517.0).
Example 9. P7C3-S2: 1-(3,6-Dimethy1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
0
0
NH
(OH
N
* *
Step 1. Synthesis of 3,6-Dimethy1-9-(oxiran-2-ylmethyl)-9H-carbazole
r0
N
0*
Following Representative Procedure 1, 3,6-dimethyl carbazole (Beyer etal., 0.
J. Org. Chem. 2003,
68, 2209-2215) was added to epichlorohydrin in 69% yield.
1H NMR (CDC13, 500 MHz) A 7.84 (d, 2H, J = 1.0 Hz), 7.30 (d, 2H, J = 8.5 Hz),
7.26 (dd, 2H, J =
1.0, 8.5 Hz), 4.54 (dd, 1H, J = 3.5, 16.0 Hz), 4.35 (dd, 1H, J = 4.5, 16.0
Hz), 3.30 (m, 1H), 2.76 (dd, 1H, J =
4.0, 5.0 Hz), 2.52 (s, 6H), 2.51 (m, 1H)
Step 2. Synthesis of 1-(3,6-Dimethy1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
0
I.
r(NH
OH
N
* *
128
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following Representative procedure 2, 1-(3,6-Dimethy1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol was prepared from 3,6-Dimethy1-9-(oxiran-2-
ylmethyl)-9H-carbazole in 22
% following purification by preparative TLC.
1H NMR (CDC13, 500 MHz) 6 7.84 (d, 2H, J = 0.5 Hz), 7.30 (d, 2H, J = 8.0 Hz),
7.23 (d, 2H, J = 8.0
Hz), 7.05 (t, 1H, J = 8.0 Hz), 6.28 (dd, 1H, J = 2.5, 8.0 Hz), 6.21 (dd, 1H, J
= 2.5, 8.0 Hz), 6.12 (dd, 1H, J =
2.0, 2.5 Hz), 4.39 (m, 3H), 4.01 (br s, 1H), 3.68 (s, 3H), 3.31 (dd, 1H, J =
3.0, 11.5 Hz), 3.17 (dd, 1H, J = 6.5,
13.0 Hz), 2.51 (s, 6H), 2.13 (br s, 1H)
13C NMR (CDC13, 125 MHz) 6 161.0, 149.5, 139.5 (2C), 130.3 (2C), 128.7, 127.3
(2C), 123.2 (2C),
120.5 (2C), 108.7 (2C), 106.7, 103.7, 99.5, 69.7, 55.2, 48.0, 47.4, 21.6 (2C).
ESI m/z 375.2 ([M+1-1] , C24H27N202 requires 375.2)
Example 10. P7C3-S14: 1-(3-Bromo-6-methy1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-
ol
Me
Br i& =
LW N
v........OH
HN
* OMe
Step 1. Synthesis of 3-Bromo-6-methyl-9-(oxiran-2-ylmethyl)-9H-carbazole
Me
Br
N
C)
Following Representative Procedure 2, Example 14 was prepared in 74% yield.
1H NMR (CDC13, 500 MHz) 6 8.13 (d, 1H, J= 1.5 Hz), 7.80 (d, 1H, J= 1.0 Hz),
7.50 (dd, 1H, J =
2.0, 8.5 Hz), 7.33-7.28 (m, 3H), 4.57 (dd, 1H, J = 3.0, 15.5 Hz), 4.29 (dd,
1H, J = 5.0, 15.5 Hz), 3.29 (m, 1H),
2.77 (dd, 1H, J = 4.0, 4.5 Hz), 2.51 (s, 3H), 2.48 (dd, 1H, J = 2.5, 4.5 Hz)
Step 2. Synthesis of 1-(3-Bromo-6-methy1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol
129
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Me
Br r& .
1W N0N
HN
* OMe
Following Representative Procedure 2, Example 15 was prepared from 3-Bromo-6-
methyl-9-(oxiran-
2-ylmethyl)-9H-carbazole in 41% yield.
1H NMR (CDC13, 500 MHz) 6 8.14 (d, 1H, J = 2.0 Hz), 7.81 (s, 1H), 7.48 (dd,
1H, J = 2.0, 8.5 Hz),
7.31 (d, 1H, J = 5.0 Hz), 7.29 (br s, 1H), 7.06(t, 1H, J = 8.5 Hz), 6.29 (dd,
1H, J = 2.0, 8.0 Hz), 6.21 (dd, 1H,
J = 2.0, 8.0 Hz), 6.11 (t, 1H, J = 2.0 Hz), 4.37 (m, 3H), 3.99 (br s, 1H),
3.70 (s, 3H), 3.30 (dd, 1H, J = 3.5,
13.5 Hz), 3.16 (dd, 1H, J= 6.5, 13.5 Hz), 2.51 (s, 3H), 2.14 (br s, 1H)
13C NMR (CDC13, 125 MHz) 6 161.0, 149.4, 139.8, 139.5, 130.3, 129.4, 128.5,
128.2, 124.7, 123.2,
122.3 120.7, 112.1, 110.6, 109.0, 106.7, 103.7, 99.6, 69.5, 55.3, 47.9, 47.4,
21.5.
ESI m/z 439.1 ([M+1-1] , C23H24BrN202 requires 439.1)
Example 11. P7C3-S15: 1-(3,6-Dichloro-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
a
a . .
N
HO--.
HN
it OMe
Step 1. Synthesis of 3,6-Dichloro-9-(oxiran-2-ylmethyl)-9H-carbazole
CI
CI 0 .
N
0
Following Representative Procedure 1, 3,6-Dichloro-9-(oxiran-2-ylmethyl)-9H-
carbazole was
prepared in 23% yield.
130
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 600 MHz) 6 7.92 (d, 2H, J = 1.8 Hz), 7.40 (dd, 2H, J = 1.8, 9.0
Hz), 7.32 (d, 2H, J
= 9.0 Hz), 4.59 (dd, 1H, J = 3.0, 16.2 Hz), 4.22 (dd, 1H, J = 5.4, 16.2 Hz),
3.27 (m, 1H), 2.78 (dd, 1H, J =
4.2, 4.8 Hz), 2.46 (dd, 1H, J = 2.4, 4.8 Hz)
Step 2. Synthesis of 1-(3,6-Dichloro-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
I
a r& =
l'W N
HO--.
HN
* OMe
Following Representative Procedure 2, 1-(3,6-dichloro-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol was prepared from 3,6-Dichloro-9-(oxiran-2-
ylmethyl)-9H-carbazole in
37% yield.
1H NMR (CDC13, 500 MHz) 6 7.95 (d, 2H, J= 2.0 Hz), 7.38 (dd, 2H, J = 2.0, 8.5
Hz), 7.33 (d, 2H, J
= 9.0 Hz), 7.06 (t, 1H, J = 8.0 Hz), 6.30 (dd, 1H, J = 2.0, 8.0 Hz), 6.20 (dd,
1H, J = 2.0, 8.0 Hz), 6.11 (dd,
1H, J = 2.0, 2.5 Hz), 4.30-4.35 (m, 3H), 3.70 (s, 3H), 3.28 (dd, 1H, J = 3.5,
13.0 Hz), 3.13 (dd, 1H, J = 6.5,
13.0 Hz)
13C NMR (CDC13, 150 MHz) 6 161.0, 149.3, 139.7, 130.4 (2C), 126.9 (2C), 125.5
(2C), 123.4 (2C),
120.4 (2C), 110.5 (2C), 106.7, 103.8, 99.8, 69.6, 55.3, 48.0, 47.5.
ESI m/z 415.0 ([M+1-1] , C22H20C12N202 requires 415.1)
Example 12. P7C3-S18: 1-(5-bromo-2,3-dimethy1-1H-indo1-1-y1)-3-
(phenylamino)propan-2-ol
Me
Br 0\ Me
N
HO---...._
NH
ilt
Step 1. Synthesis of 5-Bromo-2,3-dimethy1-1H-indole
Me
Br .
\ Me
N
H
131
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following a published procedure (Gundersen, E. G. U.S. Patent App. Publ. US
2005/070592) 2-
Butanone (0.11 mL, 1.278 mmol) was added to a solution of 4-
bromophenylhydrazine hydrochloride (0.300 g,
1.342 mmol in Et0H (3.8 mL). The mixture was heated to reflux for 22 h,
concentrated in vacuo, and
partitioned between Et0Ac and 1N HC1. The organic layer was washed with H20
and saturated aqueous
NaHCO3, dried over Na2SO4, filtered, and concentrated. The crude residue was
purified by chromatography
(Si02, 0-20% Et0Ac/Hexane) to afford the desired indole as a pink powder (200
mg, 67%).
1H NMR (CDC13, 500 MHz) 6 7.69 (br s, 1H), 7.55 (d, 1H, J = 2.0 Hz), 7.15 (dd,
1H, J = 2.0, 8.5 Hz),
7.09 (dd, 1H, J = 0.5, 8.5 Hz), 2.34 (s, 3H), 2.15 (d, 3H, J = 0.5 Hz). ESI
m/z 224.0 ([M+H]+, CioHliBrN
requires 224.0)
Step 2. Synthesis of 5-Bromo-2,3-dimethy1-1-(oxiran-2-ylmethyl)-1H-indole
Me
Br is\ Me
N
o\
Following Representative Procedure 1, 5-bromo-2,3-dimethy1-1-(oxiran-2-
ylmethyl)-1H-indole was
prepared from 5-Bromo-2,3-dimethy1-1H-indole in 48% yield.
1H NMR (CDC13, 500 MHz) 6 7.58 (d, 1H, J = 2.0 Hz), 7.20 (dd, 1H, J = 2.0, 8.5
Hz), 7.10 (d, 1H, J
= 8.5 Hz), 4.35 (dd, 1H, J= 3.0, 16.0 Hz), 4.09 (dd, 1H, J = 4.5, 16.0 Hz),
3.17 (m, 1H), 2.72 (t, 1H, J = 4.5
Hz), 2.35 (dd, 1H, J = 3.0, 5.0 Hz), 2.33 (s, 3H), 2.19 (s, 3H). ESI m/z 280.0
([M+H]+, C13H15BrNO requires
280.0)
Step 3. Synthesis of 1-(5-bromo-2,3-dimethy1-1H-indo1-1-y1)-3-
(phenylamino)propan-2-ol
Me
Br 0\ Me
N
HO---...._
NH
ilt
Following Representative Procedure 2, 1-(5-bromo-2,3-dimethy1-1H-indo1-1-y1)-3-
(phenylamino)propan-2-ol was prepared from 5-Bromo-2,3-dimethy1-1-(oxiran-2-
ylmethyl)-1H-indole in 39%
yield.
1H NMR (CDC13, 500 MHz) 6 7.58 (d, 1H, J = 2.0 Hz), 7.17 (dd, 2H, J = 7.0, 8.5
Hz), 7.11 (d, 1H, J
= 8.5 Hz), 6.75 (t, 1H, J = 7.0 Hz), 6.60 (d, 2H, J = 8.5 Hz), 4.17 (m, 1H),
4.15 (m, 2H), 3.27 (dd, 1H, J =
3.0, 8.5 Hz), 3.12 (dd, 1H, J = 7.0, 13.0 Hz), 2.34 (s, 3H), 2.19 (s, 3H)
132
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
13C NMR (CDC13, 125 MHz) 6 147.9, 135.1, 134.3, 130.6, 129.6(2C), 123.6,
120.9, 118.6, 113.7
(2C), 112.5, 110.5, 107.1, 69.9, 47.7, 47.4, 10.7, 9Ø ESI m/z 373.0 ([M+11]
, C19H22BrN20 requires 373.1).
Example 13. P7C3-S26: 1-(3,6-Dibromo-9H-pyrido13,4-blindol-9-y1)-3-
(phenylamino)propan-2-61
Br
¨
Br 0 N
\ /
NL....(0:
NH
0
Step 1. Synthesis of 3,6-Dibromo-fl-carboline
Br
Br, N
\ /
N
H
Following a literature procedure (Ponce, M. A.; Erra-Balsells, R. J.
Heterocyclic Chem. 2001, 38,
1087) 13-Carboline (0.100 g, 0.595 mmol) and Si02 (1.00 g) were suspended in
CH2C12 (15 mL). N-
Bromosuccinimde (0.212 g, 1.189 mmol) was dissolved in CH2C12 (15 mL) and the
solution was added to the
carboline mixture slowly via syringe in the absence of light. The reaction was
stirred at ambient temperature
for 2.5 h, after which the silica gel was filtered off and washed 3xCH2C12.
The combined organic layer was
extracted with 0.1 M NaOH and saturated aqueous NaC1, dried over Na2SO4,
filtered, and concentrated in
vacuo. The crude product was purified by chromatography (Si02, 0-100%
Et0Ac/Hexane) to afford the
desired 3,6-dibrominated carboline (25 mg, 13%) as well as 6,8-dibrominated
carboline (15 mg, 8%) and the
tribrominated carboline (36 mg, 19%).
1H NMR (d6-DMSO, 500 MHz) 6 8.72 (s, 1H), 8.58 (d, 1H, J = 1.5 Hz), 8.48 (s,
1H), 7.70 (dd, 1H, J
= 1.5, 9.0 Hz), 7.58 (d, 1H, J = 9.0 Hz). ESI m/z 326.9 ([M+H]+, C11H7Br2N2
requires 326.9).
Step 2. Synthesis of 3,6-Dibromo-9-(oxiran-2-ylmethyl)-9H-pyrido[3,4-1Vindole
Br
--
Br 0 N
\ /
N
o
133
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following Representative Procedure 1, 3,6-dibromo-9-(oxiran-2-ylmethyl)-9H-
pyrido[3,4-b]indole
was prepared from 3,6-dibromo-3-carboline in 73% yield.
1H NMR (CDC13, 400 MHz) 6 8.62 (d, 1H, J= 0.8 Hz), 8.17 (d, 1H, J= 2.0 Hz),
8.02 (d, 1H, J = 1.2
Hz), 7.69 (dd, 1H, J = 2.0, 8.8 Hz), 7.41 (d, 1H, J = 8.8 Hz), 5.34 (br s,
1H), 4.73 (dd, 1H, J = 2.4, 16.0 Hz),
4.27 (dd, 1H, J = 5.2, 16.0 Hz), 3.32 (m, 1H), 2.83 (dd, 1H, J = 4.0, 4.4 Hz),
2.49 (dd, 1H, J = 2.4, 4.4 Hz).
ESI m/z 382.9 ([M+H]+, C14H11Br2N20 requires 382.9).
Step 3. Synthesis of 1-(3,6-Dibromo-9H-pyrido[3,4-Nindo1-9-y1)-3-
(phenylamino)propan-2-ol
Br
--
Br 0 N
\ /
Nv.......(0:
NH
*
Following Representative Procedure 2, 1-(3,6-dibromo-9H-pyrido[3,4-b]indo1-9-
y1)-3-
(phenylamino)propan-2-ol was prepared from 3,6-dibromo-9-(oxiran-2-ylmethyl)-
9H-pyrido[3,4-b]indole in
14% yield after purification by preparative TLC.
1H NMR (CDC13, 500 MHz) 6 8.64 (s, 1H), 8.18 (d, 1H, J = 2.0 Hz), 7.99 (s,
1H), 7.66 (dd, 1H, J =
1.5, 9.0 Hz), 7.40 (d, 1H, J = 9.0 Hz), 7.18 (dd, 2H, J = 7.5 Hz), 6.76 (t,
1H, J = 7.5 Hz), 6.63 (d, 2H, J = 8.5
Hz), 5.33 (br s, 1H), 4.38-4.49 (m, 3H), 3.37 (dd, 1H, J = 4.0, 13.0 Hz), 3.21
(dd, 1H, J = 7.0, 13.0 Hz)
13C NMR (CDC13, 125 MHz) 6 147.7, 141.2, 137.0, 132.6, 132.5, 130.9, 130.1,
129.7 (2C), 125.0,
122.0, 119.0, 118.6, 113.8 (2C), 113.4, 111.9, 69.6, 48.1, 47.9. ESI m/z 475.9
([M+H]+, C20H18Br2N30
requires 476.0)
Example 14. P7C3-S36: 1-(3-Azidophenylamino)-3-(3,6-dibromo-9H-carbazol-9-
yppropan-2-61
Br
NkiØ....H
NH
0 N
3
Following Representative Procedure 2, Example 14 was prepared in 14% yield.
134
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 6 8.13 (d, 2H, J = 2.0 Hz), 7.53 (dd, 2H, J = 2.0, 8.5
Hz), 7.31 (d, 2H, J =
8.5 Hz), 7.12 (t, 1H, J = 8.0 Hz), 6.44 (dd, 1H, J = 1.5, 8.0 Hz), 6.36 (dd,
1H, J = 1.5, 8.0 Hz), 6.20 (dd, 1H, J
= 2.0 Hz), 4.35-4.41 (m, 3H), 4.10 (br s, 1H), 3.31 (dd, 1H, J = 3.0, 13.0
Hz), 3.17 (dd, 1H, J = 6.5, 13.0 Hz),
2.11 (br s, 1H)
ESI m/z 513.9 ([M+1-1] , C21H18l3r2N50 requires 514.0)
Example 15. P7C3-S34: 1,3-Bis(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
Br
Br t& .
l'W N
HO---
Br # N
Br
3,6-Dibromocarbazole (0.050 g, 0.154 mmol) was dissolved in DMF (1.5 mL) and
cooled to 0 C.
10 NaH (60% dispersion in mineral oil, 0.007 g, 0.169 mmol) was added and
the reaction was stirred for 45 min
at 0 C. 3,6-Dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole (0.059 g, 0.154 mmol)
was added and the reaction
was stirred at ambient temperature for 24 h. Upon consumption of the starting
material by TLC, the reaction
was partitioned between Et0Ac and H20. The aqueous layer was washed 3x with
Et0Ac, and the combined
organics were washed with saturated aqueous NaC1, dried over Na2SO4, filtered,
and concentrated in vacuo.
15 The crude residue was purified by chromatography (Si02, 0-50%
Et0Ac/Hexane) to afford the desired
product (37 mg, 34%).
1H NMR (acetone-d6, 400 MHz) 6 8.36 (d, 4H, J = 2.0 Hz), 7.64 (d, 4H, J = 8.8
Hz), 7.56 (dd, 4H, J
= 2.0, 8.8 Hz), 4.72 (m, 5H), 2.78 (br s, 1H)
13C NMR (acetone-d6, 100 MHz) 6 141.2 (4C), 129.8 (4C), 124.6 (4C), 124.1
(4C), 112.9 (4C), 112.7
20 (4C), 70.3, 48.3 (2C). ESI m/z 747.0 ([M+CO2H], C28H19Br4N203 requires
746.8)
Example 16. P7C3-S35: 1-(9H-Carbazol-9-y1)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br
Br, .
N
HO----
4. N
el
135
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following a procdure analogous to that used to prepare Example 15, Example 16
was prepared in 48%
yield.
1H NMR (acetone-d6, 400 MHz) 6 8.36 (m, 2H), 8.14 (d, 2H, J = 8.0 Hz), 7.63
(d, 2H, J = 8.4 Hz),
7.55 (s, 2H), 7.42 (dt, 2H, J = 1.2, 7.2 Hz), 7.20 (dt, 2H, J = 0.8, 7.2 Hz),
4.76(m, 1H), 4.64-4.72(m, 4H),
2.77 (br s, 1H).
13C NMR (acetone-d6, 100 MHz) 6 142.0 (2C), 141.0 (2C), 129.8 (2C), 126.6
(2C), 124.5 (2C), 124.1
(2C), 123.8 (2C), 121.0 (2C), 119.9 (2C), 112.7 (2C), 112.6 (2C), 110.5 (2C),
70.3, 48.4, 48.1.
ESI m/z 591.0 ([M+CO2H], C28H21Br2N203 requires 591.0).
Example 17. P7C3-S31: 3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxy-N-(3-
methoxypheny1)-
propanamide
Br
Br r& It
IW N
\.......(01-1
0
it OMe
Step 1. Synthesis of Methyl 3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
hydroxypropanoate
Br
Br* .
N
v......<0 H
MO e
3,6-Dibromocarbazole (0.300 g, 0.923 mmol) was dissolved in DMF (1.2 mL) and
cooled to 0 C.
NaH (60% dispersion in mineral oil, 0.074 g, 1.846 mmol) was added and the
reaction stirred for 1 h at 0 C.
Methyl glycidate (0.471 g, 4.615 mmol) was added and the reaction was stirred
and warmed to ambient
temperature over 3.5 h. Upon completion by TLC the reaction mixture was
partitioned between Et0Ac and
H20. The aqueous layer was extracted 3x with Et0Ac, and the combined organics
were washed with saturated
aqueous NaC1, dried over Na2SO4, filtered, and concentrated in vacuo. The
crude residue was purified by
chromatography (Si02, 0-30% Et0Ac/Hexane) to afford the desired product (125
mg, 32%).
1H NMR (CDC13, 500 MHz) 6 8.10 (d, 2H, J = 2.0 Hz), 7.53 (dd, 2H, J = 2.0, 9.0
Hz), 7.36 (d, 2H, J
= 9.0 Hz), 4.63-4.55 (m, 3H), 3.69 (s, 3H), 2.94 (d, 1H, J = 5.5 Hz).
ESI m/z 425.8 ([M+I-1] , C16H14Br2NO3requires 425.9)
136
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Step 2. Synthesis of 3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropanoic acid
Br
Br 0 *
N
\...........T
OH
0
NaOH (0.64 mL, 1M solution in H20) was added to a suspension of methyl 3-(3,6-
dibromo-9H-
carbazol-9-y1)-2-hydroxypropanoate (0.055 g, 0.129 mmol) in Et0H (2.6 mL) and
the reaction was stirred at
ambient temperature for 2.5 h. The reaction was concentrated in vacuo and the
residue was acidified with 1N
aqueous HC1. The mixture was extracted with Et0Ac (3x), and the combined
organics were washed with
saturated aqueous NaC1, dried over Na2SO4, filtered, and concentrated in vacuo
to afford the desired product as
a white solid (53 mg, 99%).
1H NMR (CDC13, 500 MHz) 6 8.10 (d, 2H, J= 1.5 Hz), 7.52 (dd, 2H, J= 1.5, 8.5
Hz), 7.40 (d, 2H, J
= 9.0 Hz), 4.68 (m, 2H), 4.60 (dd, 1H, J = 6.5, 15.5 Hz).
ESI m/z 411.9 ([M+1-1] , C151-112Br2NO3requires 411.9)
Step 3. Synthesis of 3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxy-N-(3-
methoxypheny1)-propanamide
Br
Br 0 *
N
\........)H
NH
0
41 15 OMe
3-(3,6-Dibromo-9H-carbazol-9-y1)-2-hydroxypropanoic acid (0.025 g, 0.061 mmol)
was suspended in
anhydrous CH2C12and cooled to 0 C. Thionyl chloride (0.005 mL, 0.073 mmol)
was added dropwise and the
reaction was stirred at 0 C for 1 h. m-Anisidine (0.008 mL, 0.073 mmol) and
Et3N (0.010 mL, 0.073 mmol)
were added and the reaction was allowed to warm to ambient temperature over
2.5 h. Upon completion, the
solution was partitioned between Et0Ac and H20. The aqueous layer was washed
3x with Et0Ac, and the
combined organics were washed with saturated aqueous NaC1, dried over Na2SO4,
filtered, and concentrated in
vacuo. The crude residue was purified by chromatography (Si02, 0-30%
Et0Ac/Hexane) to afford the desired
product (15 mg, 48%).
137
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (acetone-d6, 500 MHz) 6 9.22 (br s, 1H), 8.34 (d, 2H, J= 1.5 Hz), 7.65
(d, 2H, J = 8.5 Hz),
7.59 (dd, 2H, J = 4.0, 8.5 Hz), 7.42 (dd, 1H, J = 2.0 Hz), 7.24 (m, 1H), 7.20
(dd, 1H, J = 8.0 Hz), 6.67 (dd,
1H, J = 2.0, 8.0 Hz), 5.56 (br s, 1H), 4.82 (m, 1H), 4.73 (m, 2H), 3.77 (s,
3H)
13C NMR (CDC13, 100 MHz) 6 170.9, 161.1, 141.1, 140.3, 130.3 (2C), 129.8 (2C),
124.6 (2C), 124.0
(2C), 113.1 (2C), 112.8 (2C), 112.7, 110.5, 106.4, 72.7, 55.6, 48.4.
ESI m/z 514.9 ([M¨H], C22H17Br2N203 requires 515.0)
Example 18. Ethyl 5-(2-Hydroxy-3-(3-methoxyphenylamino)propy1)-8-methyl-3,4-
dihydro-1H-
pyrido[4,3-Mindole-2(5H)-carboxylate
0
,-0 Et
N
Me 0\
N
HO--?
HN
0
* /
Step 1. Synthesis of Ethyl 8-Methyl-3,4-dihydro-1H-pyrido[4,3-1Vindole-2(5H)-
carboxylate
0
,-0 Et
N
Me 0\
N
H
Following a literature procedure (Harbert et al., J. Med. Chem. 1980, 23, 635-
643)p-tolylhydrazine
hydrochloride (0.500 g, 3.15 mmol) and 1-carbethoxy-4-piperidone (0.18 mL,
1.17 mmol) were suspended in
Et0H (0.880 mL) and heated to reflux for 2 hours. The reaction mixture was
removed from heat and allowed
to stand overnight at ambient temperature. The resulting mixture was filtered
and washed with 50% aqueous
Et0H to afford the desired product as a beige powder (259 mg, 86%).
1H NMR (CDC13, 500 MHz) 6 7.73 (br s, 1H), 7.23 (s, 1H), 7.18 (d, 1H, J = 8.0
Hz), 6.96 (d, 1H, J =
8.0 Hz), 4.64 (br s, 2H), 4.18 (q, 2H, J = 7.0 Hz), 3.85 (m, 2H), 2.81 (br s,
2H), 2.42 (s, 3H), 1.28 (t, 3H, J =
7.0 Hz).
Step 2. Synthesis of Ethyl 8-Methy1-5-(oxiran-2-ylmethyl)-3,4-dihydro-1H-
pyrido[4,3-1Vindole-2(5H)-
carboxylate
138
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
0
¨0 Et
N
Me 0\
N
o
Ethyl 8-methy1-3,4-dihydro-1H-pyrido[4,3-b]indole-2(511)-carboxylate (0.025 g,
0.097 mmol) was
dissolved in anhydrous degassed THF and was cooled to ¨78 C. A solution of n-
BuLi (0.082 mL, 1.78 M in
hexanes) was added dropwise and the reaction was stirred at ¨78 C for 30 min.
Epibromohydrin (0.016 mL,
0.194 mmol) was added and the reaction was allowed to warm slowly to ambient
temperature. After 3.5 h,
epibromohydrin (0.008 mL, 0.097 mmol) was added and the reaction was stirred
overnight at ambient
temperature. Upon completion, saturated aqueous NH4C1 was added to quench the
reaction and the mixture
was extracted with Et0Ac (3x). The combined organic layers were washed with
brine, dried over Na2SO4,
filtered, and concentrated. The crude residue was purified by chromatography
(Si02, 0-50% Et0Ac/Hexane)
to afford the desired product (15 mg, 49%).
1H NMR (CDC13, 500 MHz) 6 7.19 (m, 1H), 7.00 (d, 1H, J = 8.5 Hz), 4.65 (br s,
2H), 4.32 (dd, 1H, J =
3.0, 15.5 Hz), 4.18 (q, 2H, J = 7.0 Hz), 4.08 (dd, 1H, J = 5.0, 15.5 Hz), 3.85
(m, 2H), 3.18 (m, 1H), 2.81 (br s,
2H), 2.73 (dd, 1H, J = 4.0, 4.5 Hz), 2.44 (s, 3H), 2.38 (br s, 1H), 1.29 (t,
3H, J = 7.0 Hz)
Step 3. Synthesis of Ethyl 5-(2-Hydroxy-3-(3-methoxyphenylamino)propy1)-8-
methyl-3,4-dihydro-1H-
pyrido[4,3-Nindole-2(5H)-carboxylate
0
)¨O Et
N
Me 0\
N
HO---
HN
* 0/
Following a literature procedure (Chakraborti et al., Eur. J. Org. Chem. 2004,
3597-3600) LiBr (0.001
g, 0.010 mmol) and m-anisidine (0.011 mL, 0.102 mmol) were added to ethyl 8-
Methy1-5-(oxiran-2-ylmethyl)-
3,4-dihydro-1H-pyrido[4,3-b]indole-2(5H)-carboxylate (0.032 g, 0.102 mmol) and
stirred vigorously at
ambient temperature overnight. Upon completion the reaction was partitioned
between Et0Ac/H20, and the
organic layer was concentrated to an orange oil. The crude residue was
purified by chromatography (Si02, 0-
50% Et0Ac/Hexane) to afford the desired product (30 mg, 67%).
139
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 6 7.23 (br s, 1H), 7.17 (d, 1H, J = 8.0 Hz), 7.05 (dd,
1H, J = 8.0 Hz), 6.97
(d, 1H, J = 8.5 Hz), 6.28 (dd, 1H, J = 1.5, 8.0 Hz), 6.19 (d, 1H, J = 8.0 Hz),
6.11 (br s, 1H), 4.64 (br s, 2H),
4.18 (m, 1H), 4.16 (q, 2H, J = 7.5 Hz), 4.12 (m, 1H), 3.80 (br s, 2H), 3.71
(s, 3H), 3.23 (dd, 1H, J = 3.5, 13.0
Hz), 3.07 (dd, 1H, J = 7.5, 13.0 Hz), 2.83 (m, 1H), 2.76 (m, 1H), 2.42 (s,
3H), 1.27 (t, 3H, J = 7.0 Hz).
ESI m/z 438.2 ([M+14] , C25H32N304 requires 438.2).
Example 19. P7C3-S26: 4-(3,6-dibromo-9H-carbazol-9-y1)-1-(phenylamino)butan-2-
ol
Br Br
. *
N
cOH
0
Step 1. Synthesis of 3,6-dibromo-9-(2-(oxiran-2-yOethyl)-9H-carbazole
Br Br
* 0
Nc j\
0
Crushed KOH (0.0054g, 0.0954mmo1, 1.2equiv) was added to 3,6-dibromocarbazole
(0.0258g,
0.0795mmo1, 1 equiv.) in 0.5mL DMF solution and the mixture was stirred for
30min. 1-Bromo-3,4-
epoxybutane (0.0300g, 0.199mmol) in 0.5mL DMF solution was dropwise added into
the mixture and it was
stirred at room temperature for overnight. Reaction crude was diluted with
20mL Et0Ac and washed with
water 5 x 10mL. The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford 31.2mg white
solid as product, yield 97.9%.
1H NMR (CDC13, 400 MHz) 6 ppm 1.65- 1.81 (m, 1H) 2.13 -2.27 (m, 1H) 2.34 (dd,
J=4.88, 2.64 Hz,
1H) 2.64 (dd, J=4.78, 4.05 Hz, 1H) 2.69 - 2.80 (m, 1H) 4.26 - 4.54 (m, 2H)
7.27 (d, J=8.69 Hz, 2H) 7.50 (dd,
J=8.69, 1.90 Hz, 2H) 8.08 (d, J=1.90 Hz, 2H)
Step 2. Synthesis of 4-(3,6-dibromo-9H-carbazol-9-y1)-1-(phenylamino)butan-2-
ol
Br N Br
fk #
il
0
According to Representative Procedure 2, Example 19 was isolated as a white
solid in 31% yield.
140
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6ppm 1.87- 1.98 (m, 1H) 2.05 -2.14 (m, 1H) 2.99 - 3.07
(dd, J=13.24,
3.43 Hz, 1H) 3.09 - 3.17 (dd, J=13.24, 8.27 Hz, 1H) 3.60 - 3.74 (m, 1H) 4.39 -
4.48 (m, 1H) 4.51 - 4.60 (m,
1H) 6.57 (d, J=7.71 Hz, 2H) 6.74 (t, J=7.34 Hz, 1H) 7.15 (dd, J=8.27, 7.59 Hz,
2H) 7.38 (d, J=8.69 Hz, 2H)
7.56 (dd, J=8.69, 1.90 Hz, 2H) 8.14 (d, J=1.85 Hz, 2H)
13C NMR (CDC13, 500 MHz) 6 = 148.1, 139.6, 129.6, 129.4, 123.8, 123.6, 118.7,
113.6, 112.4, 110.8,
67.7, 51.0, 39.9, 33.7.
m/z (ESI): 486.9 (M + II+) ([M+1] for C22H20Br2N20 requires 467.0)
Example 20. P7C3-S33: N-(3-(3,6-dibromo-9H-carbazol-9-yl)propypaniline
Br
*
H
Br
* NN., N 0
Step 1. Synthesis of 3,6-dibromo-9-(3-bromopropy1)-9H-carbazole
Br
r
N
* .
Br Br
Crushed KOH (0.0673g, 1.20mmol, 1.2equiv) was added to 3,6-dibromocarbazole
(0.3250 g, 1.00
mmol) in 2mL DMF solution and the mixture was stirred for 30min. 1,3-
dibromopropane (0.5047g, 2.50mmol,
2.5equiv) in 3mL DMF solution was added dropwise into the mixture and it was
stirred at room temperature
overnight. The crude reaction mixture was diluted with 30mL Et0Ac and washed
with 1M HC1 2 x 10mL and
water 3 x 10mL. The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford the crude
product, which was subjected to silica gel chromatography using Hexanes/Et0Ac
to afford 0.1275g colorless
oil as product, yield 28.6%.
1H NMR (CDC13, 400 MHz) 6ppm 2.24 - 2.44 (m, 2H) 3.29 (t, J=6.05 Hz, 2H) 4.33
(t, J=6.59 Hz, 2H)
7.26 (d, J=8.83 Hz, 2H) 7.51 (dd, J=8.69, 1.95 Hz, 2H) 8.02 (d, J=1.71 Hz, 2H)
Step 2. Synthesis of N-(3-(3,6-dibromo-9H-carbazol-9-Apropy1)-2-nitro-N-
phenylbenzenesulfonamide
141
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
0NO2
4 o=s=o
1
Br * NN 0
Crushed KOH (0.0024g, 0.0431 mmol) was added to 2-nitro-N-
phenylbenzenesulfonamide (0.0100 g,
0.0359 mmol) in 0.2mL DMF solution and the mixture was stirred for 30min. 3,6-
dibromo-9-(3-
bromopropy1)-9H-carbazole (Example 35, 0.0240g, 0.0538 mmol) in 0.3 mL DMF
solution was added
dropwise into the mixture and it was stirred at room temperature overnight.
The crude reaction mixture was
diluted with 20mL Et0Ac and washed with water 5 x 10mL. The organic layer was
dried over anhydrous
Na2SO4 and evaporated to afford the crude product, which was subjected to
silica gel chromatography using
Hexanes/Et0Ac to afford 0.0082g white solid as impure product, purity 66.9%
(impurity is starting Ns-aniline;
used without additional purification), yield 35.5%.
1H NMR (CDC13, 400 MHz) 6ppm 1.89 - 2.01 (m, 2H) 3.95 (t, J=6.61 Hz, 2H) 4.32 -
4.38 (m, 2H)
7.15 (s, 1H) 7.17 (s, 1H) 7.18 - 7.25 (m, 3H) 7.32 (d, J=3.66 Hz, 2H) 7.41 -
7.44 (m, 2H) 7.51 (dd, J=8.69,
1.95 Hz, 2H) 7.59 - 7.71 (m, 2H) 8.09 (d, J=1.90 Hz, 2H)
Step 3. Synthesis of N-(3-(3,6-dibromo-9H-carbazol-9-yl)propyl)aniline
Br
*
H
Br
N-(3-(3,6-dibromo-9H-carbazol-9-yl)propy1)-2-nitro-N-phenylbenzenesulfonamide
(0.0378g,
0.0588mmo1, lequiv), cesium carbonate (0.0574g, 0.176 mmol, 3equiv) and
benzenethiol (0.0194g, 0.176
mmol) were mixed in lmL anhydrous THF. The mixture was stirred at room
temperature for 3 hours. THF
was removed under vacuum and the residue was purified by silica gel
chromatography using Hexanes/Et0Ac
to afford 0.0164g colorless oil as product, yield 60.9%.
1H NMR (CDC13, 400 MHz) 6ppm 2.08 - 2.29 (m, 2H) 3.09 (t, J=6.56 Hz, 2H) 3.55
(br. s., 1H) 4.37 (t,
J=6.69 Hz, 2H) 6.53 (dd, J=8.56, 0.95 Hz, 2H) 6.73 (t, J=7.32 Hz, 1H) 7.16
(dd, J=8.49, 7.37 Hz, 2H) 7.25 (d,
J=8.69 Hz, 2H) 7.51 (dd, J=8.69, 1.95 Hz, 2H) 8.12 (d, J=1.85 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6 =148.0, 139.5, 129.6, 129.4, 123.7, 123.6, 118.2,
113.3, 112.4, 110.5,
41.4, 40.9, 28.9
MS (ESI) ,m/z: 456.9 [M+H]+ ([M+H]+ for C21H18Br2N2 requires 457.0)
142
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 21. P7C3-S32: 1-(3,6-dibromo-9H-carbazol-9-y1)-4-(phenylamino)butan-2-
ol
Br Br
. 11*
N
Cr.:1\1
OH 0
Step 1. Synthesis of N-(but-3-eny1)-2-nitro-N-phenylbenzenesulfonamide
*12
A is' N
No
I.
Crushed KOH (0.0484g, 0.862mmo1, 1.2equiv) was added to 2-nitro-N-
phenylbenzenesulfonamide
(0.200g, 0.719mmol) in lmL DMF, and the mixture was stirred for 30 min. 4-
Bromo-1-butene (0.2426g,
1.80mmol) in 2mL DMF solution was added dropwise into the mixture and it was
stirred at room temperature
overnight. The reaction mixture was diluted with 30mL Et0Ac and washed with 1M
HC1 2 x 10mL and water
3 x 10mL. The organic layer was dried over anhydrous Na2SO4 and evaporated to
afford the crude product,
which was subjected to silica gel chromatography using Hexanes/Et0Ac to afford
0.1546g white solid, yield
63.5%.
1H NMR (CDC13, 400 MHz) 6ppm 2.20 (q, J=6.90 Hz, 2H) 3.83 (t, J=7.15 Hz, 2H)
5.00 (d, J=4.39 Hz,
1H) 5.03 (s, 1H) 5.64 - 5.83 (m, 1H) 7.14 - 7.21 (m, 3H) 7.30 (d, J=1.85 Hz,
2H) 7.42 - 7.46 (m, 2H) 7.52 -
7.58 (m, 1H) 7.60 - 7.66 (m, 1H)
Step 2. Synthesis of 2-nitro-N-(2-(oxiran-2-yOethyl)-N-
phenylbenzenesulfonamide
*0 0
4S, N
NO p
I*
mCPBA (77%, 0.0550g, 0.246mmo1) was added to N-(but-3-eny1)-2-nitro-N-
phenylbenzenesulfonamide (0.0653 g, 0.196 mmol) in 1 mL CHC13 at 0 C. The
mixture was stirred at 0 C for
30 min, then gradually warmed up to room temperature and continued to stir for
18hr. After TLC showed the
disappearance of starting material, the reaction mixture was diluted with a
1:1 mixture of water and saturated
NaHCO3 (2 x 10mL) and water (10mL). The organic layer was dried over anhydrous
Na2SO4 and evaporated
to afford the crude product, which was subjected to silica gel chromatography
using Hexanes/Et0Ac to afford
0.0662 g colorless oil as product, yield 96.9%.
143
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6ppm 1.66- 1.79 (m, 2H) 2.46 (dd, J=4.95, 2.66 Hz, 1H)
2.70 - 2.80 (m,
1H) 2.93 - 3.03 (m, 1H) 3.87 - 4.07 (m, 2H) 7.19 - 7.23 (m, 2H) 7.28 - 7.34
(m, 3H) 7.43 - 7.47 (m, 2H) 7.57 -
7.66 (m, 2H).
MS (ESI) m/z: 371.0 (M + Na+) ([M+Na]+ for C16H16N2055 requires 371.1)
Step 3. Synthesis of N-(2-(oxiran-2-yOethyl)aniline
0
HN
I.
Prepared from 2-nitro-N-(2-(oxiran-2-yl)ethyl)-N-phenylbenzenesulfonamide
using an analogous
procedure as used to prepare the compound of Example 20.
1H NMR (CDC13, 400 MHz) 6ppm 1.64- 1.79 (m, 1H) 1.98 -2.15 (m, 1H) 2.55 (dd,
J=4.90, 2.71 Hz,
1H) 2.79 (t, J=4.44 Hz, 1H) 3.00 - 3.10 (m, 1H) 3.31 (t, J=6.64 Hz, 2H) 3.87
(br. s., 1H) 6.62 (d, J=7.71 Hz,
2H) 6.71 (t, J=7.32 Hz, 1H) 7.18 (dd, J=8.49, 7.37 Hz, 2H)
MS (ESI) m/z: 164.1 (M+H ) ([M+1]+ for C10H13N0 requires 164.1)
Step 4. Synthesis of 1-(3,6-dibromo-9H-carbazol-9-y1)-4-(phenylamino)butan-2-
ol
Br Br
* *
N
Cr Fil
OH 0
NaH (60% dispersed in mineral oil, 0.0018g, 0.0452mmo1) was added to a
solution of 3,6-
dibromocarbazole (0.0147g, 0.0452mmol) in 0.5 mL anhydrous THF and the mixture
was stirred for 15min.
N-(2-(oxiran-2-yl)ethyl)aniline (0.0067g, 0.0410mmol) in 1.5mL anhydrous THF
solution was added dropwise
and the resulting mixture was stirred at 60 C overnight. THF was removed
under vacuum and the residue was
dissolved in 10mL Et0Ac and washed with water 2 x 5mL. The organic layer was
dried over anhydrous
Na2504 and evaporated to afford the crude product, which was subjected to
silica gel chromatography using
Hexanes/Et0Ac to afford 0.0115g colorless oil; yield 57.5%.
1H NMR (CDC13, 400 MHz) 6 ppm 1.76 - 1.95 (m, 2H) 3.22 - 3.41 (m, 2H) 4.20 -
4.38 (m, 3H) 6.63
(d, J=8.49 Hz, 2H) 6.76 (t, J=7.32 Hz, 1H) 7.18 (t, J=7.95 Hz, 2H) 7.31 (d,
J=8.74 Hz, 2H) 7.54 (dd, J=8.69,
1.95 Hz, 2H) 8.12 (d, J=1.95 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6= 148.1, 139.9, 129.6, 129.5, 123.8, 123.5, 118.7,
113.9, 112.7, 111.1,
70.7, 50.0, 42.2, 34.1.
MS (ESI) m/z: 531.0 [M + HCOO] 486.9 [M + 1-1] ([M+1---]+ for C22H20Br2N20
requires 487.0)
144
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 22. P7C3-S38: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-2-
ylamino)propan-2-ol
Br
41 HO H
4it N.,.)N,t(
Br I\L,
Step 1. Synthesis of 1-amino-3-(3,6-dibromo-9H-carbazol-9-Apropan-2-ol
Br
it OH
= NNH2
Br!
A solution of NH3 (9.4mL of 7M in Me0H, 65.6mmol) was added to 3,6-dibromo-9-
(oxiran-2-
ylmethyl)-9H-carbazole (0.500 g, 1.31 mmol,). The vial was tightly sealed and
the reaction mixture was heated
to 100 C and stirred for 1 hour. Volatile components were removed under
vacuum. The residue was
suspended in CH2C12 and the white precipitate was filtered. The filtrate was
saved and CH2C12 was removed
under vacuum to afford 0.3413g white solid as crude product, which contained
about 50% unidentified side-
product. This crude product was used as is in next step without any further
purification. Purification by flash
chromatography on silica gel provided pure material.
1H NMR (CDC13, 400 MHz) 6 ppm 2.61 (dd, J=12.66, 7.78 Hz, 1H) 2.90 (dd,
J=12.52, 4.03 Hz, 1H)
3.96 - 4.06 (m, 1H) 4.32 (d, J=5.81 Hz, 2H) 7.36 (d, J=8.74 Hz, 2H) 7.55 (dd,
J=8.69, 1.95 Hz, 2H) 8.13 (d,
J=1.90 Hz, 2H)
MS (ESI) m/z: 396.9 (M+H ) ([M+H]+ for C15H14Br2N20 requires 397.0)
Step 2. Synthesis of 5-((3,6-dibromo-9H-carbazol-9-yOmethyl)oxazolidin-2-one
Br
* 0
04
Br lit, N.,.....L/NH
A solution of triphosgene (0.0890g, 0.300mmol, 0.35equiv) in 2mL anhydrous
CH2C12 was added
dropwise to a solution of 1-amino-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
(0.3413g, 0.857mmo1) and
Et3N (0.1909g, 1.886mmol) in lmL CH2C12 under N2 atmosphere at 4 C. The
reaction mixture was stirred for
15min at 4 C and then warmed to room temperature and stirred for 1 hour.
CH2C12 was removed under
vacuum. Saturated NH4C1 (5 mL) and 10 mL Et0Ac was added to the residue and
stirred for 20min. Then the
aqueous layer was separated and the organic layer was washed with water 2 x
10mL. The combined aqueous
145
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
layers were extracted with Et0Ac, dried over anhydrous Na2SO4 and evaporated
to afford the crude product,
which was subjected to silica gel chromatography using CH2C12/Et0Ac to afford
0.1173g white solid, yield
20.0% over 2 steps.
1H NMR (CDC13, 400 MHz) 6 ppm 3.37 (dd, J=8.98, 6.34 Hz, 1H) 3.67 (t, J=8.49
Hz, 1H) 4.54 (dd,
J=5.22, 1.81 Hz, 2H) 5.02 (br. s., 1H) 5.05 - 5.14 (m, 1H) 7.31 (d, J=8.69 Hz,
2H) 7.58 (dd, J=8.69, 1.85 Hz,
2H) 8.14 (d, J=1.85 Hz, 2H)
MS (ESI) m/z: 466.9 [M + HCOO] ([M+HC00]- for C16H12Br2N202 requires 466.9.
Step 3. Synthesis of 54(3,6-dibromo-9H-carbazol-9-yOmethyl)-3-(pyridin-2-
y0oxazolidin-2-one
Br
4 0
Br
04
. NN.......c./ N---0
N /
A mixture of 5-((3,6-dibromo-9H-carbazol-9-yl)methyl)oxazolidin-2-one
(0.0195g, 0.0460mmol), 2-
iodopyridine (0.0209g, 0.102mmol), CuI (0.0009g, 0. 00460mmol), and K2CO3
(0.0058g, 0.0418mmol,) in
0.5mL of DMSO was sealed tightly in a vial and heated at 130 C for 12 hours.
The reaction mixture was
cooled and diluted with 20mL Et0Ac and washed with water 5 x 10mL. The organic
layer was dried over
anhydrous Na2504 and evaporated to afford the crude product, which was
subjected to silica gel
chromatography using CH2C12/Et0Ac as elute to afford 0.0183g white solid as
product, yield 79.4%.
1H NMR (CDC13, 400 MHz) 6ppm 4.04 (dd, J=10.79, 7.08 Hz, 1H) 4.36 (dd,
J=10.69, 8.74 Hz, 1H)
4.60 (d, J=5.03 Hz, 2H) 5.02 - 5.16 (m, 1H) 7.02 (t, J=6.08 Hz, 1H) 7.35 (d,
J=8.69 Hz, 2H) 7.59 (dd, J=8.66,
1.73 Hz, 2H) 7.68 (t, J=7.88 Hz, 1H) 8.11 (s, 1H) 8.13 (d, J=1.32 Hz, 2H) 8.25
(d, J=4.93 Hz, 1H)
MS (ESI) m/z: 543.9 [M + HCOO] ([M+HC00]- for C21H15Br2N302 requires 544.0)
Step 4. Synthesis of 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-2-
ylamino)propan-2-ol
Br
41 HO H
. N .,.) N
Br I\L,
Li0H+120 (0.0076g, 0.182mmol, 10equiv) was added to 543,6-dibromo-9H-carbazol-
9-yl)methyl)-
3-(pyridin-2-yl)oxazolidin-2-one (0.0091g, 0.0182mmol) in a mixture of 208 L
THF and 23 L H20 (v/v =
9:1). The mixture was stirred at room temperature for 7 days. The reaction
mixture was purified by silica gel
chromatography using CH2C12/Et0Ac as elute to afford 0.0071g white solid as
product, yield 41.0%.
146
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6ppm 2.27 - 2.44 (m, 1H) 3.15 - 3.32 (m, 1H) 3.44 (dd,
J=15.23, 5.03 Hz,
1H) 4.26 - 4.41 (m, 3H) 4.52 (t, J=5.00 Hz, 1H) 6.46 (d, J=8.00 Hz, 1H) 6.66
(t, J=6.20 Hz, 1H) 7.37 (d,
J=8.74 Hz, 2H) 7.40 - 7.48 (m, 1H) 7.56 (dd, J=8.69, 1.90 Hz, 2H) 8.04 (d,
J=4.49 Hz, 1H) 8.14 (d, J=1.85 Hz,
2H)
13C NMR (CDC13, 400 MHz) 6 = 158.6, 146.7, 139.5, 138.1, 129.2, 123.6, 123.3,
113.9, 112.3, 110.9,
109.6, 70.5, 47.4, 46.8
MS (ESI) m/z: 518.0 [M + HCOO] ([M+HC00]- for C20H17Br2N30 requires 518Ø
Example 23. P7C3-S1: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-43-
methoxyphenyl)(methyl)-amino)propan-
2-ol
Br
Br i #
NIL.,(OH/
\--N
4 0 Me
Synthesized using a similar synthetic procedure analogous to Representative
Procedure 2.
Example 25. P7C3-S6: 3-amino-1-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyppyridinium
Br Br
. 0
N
OH
Example 25 was synthesized using a similar synthetic procedure analogous to
Representative
Procedure 2.
Example 26. P7C3-S8: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyrimidin-2-
ylamino)propan-2-ol
Br Br
. *
N
N 1
Ly.., A... ......
N N
OHH
To a 4 ml vial was added the corresponding primary amine (34.8 mg, 0.087
mmol), 2-
chloropyrimidine (10.3 mg, 0.090 mmol) and dimethylformamide (1.5 ml, 0.058
M). The reaction was heated
147
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
at 100 C overnight. The cooled reaction mixture was diluted with Et0Ac and
washed several times with water
and brine. The organic layer was dried over Na2SO4, filtered and condensed.
The crude mixture was subjected
to chromatography on silica gel (20% Me0H/ CH2C12).
1H NMR (CDC13, 400 MHz) 6 8.26 (d, 2H, J = 4.94 Hz), 8.14 (d, 2H, J = 1.88
Hz), 7.56 (dd, 2H,
J=6.7, 1.9 Hz), 7.37 (d, 2H, J=8.7 Hz), 6.63 (t, 1H, J = 4.9 Hz), 5,43 (t, 1H,
J=5.71 Hz), 4.36 (s, 3H), 3.56 (m,
1H), 3.30-3.38 (m, 1H).
13C NMR (CDC13, 126 MHz) 139.4, 29.5(2C), 129.3(2C), 123.7 (2C), 123.4(2C),
118.6(2) (2 C),
113.5(2C), 112.3, 110.7(2 C), 67.6 , 50.9, 33.6.
MS (ESI) m/z: 474.9 [(M+1) ; C19H16Br2N40 (M) requires 474)].
The title compound of Example 26 can also be synthesized using a procedure
analogous to that
described in Representative Procedure 2.
Example 28. P7C3-S19: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-methoxypropan-2-ol
Br
Br 0 .
NL....(0:
OM e
Following Representative Procedure 1, Example 28 was prepared from
dibromocarbazole and
methoxymethyloxirane.
Example 29. P7C3-S21: 1-(3,6-dibromo-9H-carbazol-9-y1)-4-phenylbutan-2-ol
Br
Br * .
N
OH
*
Following Representative Procedure 1, Example 29 was prepared from
dibromocarbazole and 2-
phenethyloxirane.
148
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 30. P7C3-S22: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(1H-indo1-1-yppropan-
2-ol
Br
Br 0 4.
N
\.......OH
W/ ,N
Following Representative Procedure 1, Example 30 was prepared from
dibromocarbazole and 1-
(oxiran-2-ylmethyl)-1H-indole.
Example 31. P7C3-S23: 3-(1-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-
1H-1,2,3-triazol-4-
yl)propan-1-ol
Br
Br 0 *
N
\.......(OH
\ N,
'N N
HO
Example 31 was synthesized using a similar synthetic procedure analogous to
Representative
Procedure 2.
Example 32. P7C3-S24: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-
ethoxyphenylamino)propan-2-ol
Br
Br, .
N
\...õ.(C.7 *
OEt
N
H
Example 32 was synthesized using a similar synthetic procedure analogous to
Representative
Procedure 2.
149
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 33. P7C3-S25: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,5-dimethy1-1H-
pyrazol-1-yppropan-2-01
Br
Br 0 it
Nv....Ø: N
N;X
Example 33 was synthesized using a similar synthetic procedure analogous to
Representative
Procedure 2.
Example 36. P7C3-S29: 1-(3-bromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-01
Br
4. .
N
N I. OCH3
H
OH
Step 1. 3-bromo-9-(oxiran-2-ylmethyl)-9H-carbazole
Br
. *
N
0
The title compound of Example 36, step 1 was prepared using a procedure
analogous to that described
in representative procedure 1.
1H NMR (CDC13, 400 MHz) 6 = 2.52 (dd, J= 4.6, 2.6 Hz, 1H) 2.80 (t, J= 4.3 Hz,
1H) 3.33 (td, J=
5.3, 2.2 Hz, 1H) 4.34 (dd, J= 15.9, 4.9 Hz, 1H) 4.64 (dd, J= 15.9, 2.9 Hz, 1H)
7.26 (t, J= 7.3 Hz, 1H) 7.35
(d, J= 8.7 Hz, 1H) 7.58 - 7.42 (m, 3H) 8.02 (d, J= 5.1 Hz, 1H) 8.19 (d, J= 1.7
Hz, 1H).
Step 2. The title compound was prepared from 3-bromo-9-(oxiran-2-ylmethyl)-9H-
carbazole using a
procedure similar to that described in representative procedure 2.
1H NMR (CDC13, 400 MHz) 6 = 2.13 (d, J= 3.0 Hz, 1H) 3.21 (dd, J= 13.0, 6.5 Hz,
1H) 3.35 (dd, J=
13.0, 3.2 Hz, 1H) 3.72 (s, 3H) 4.03 (s, br, 1H) 4.50 - 4.36 (m, 3H) 6.15 (t,
J= 2.3 Hz, 1H) 6.24 (dd, J = 8.0,
2.2 Hz, 1H) 6.32 (dd, J= 8.2, 2.3 Hz, 1H) 7.08 (t, J= 8.1 Hz, 1H) 7.30- 7.24
(m, 1H) 7.36 (d, J= 8.7 Hz, 1H)
7.51 -7.44 (m, 2H) 7.53 (dd, J= 8.7, 1.9 Hz, 1H) 8.05 (d, J= 7.9 Hz, 1H) 8.21
(d, J= 1.9 Hz, 1H)
150
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
13C NMR (CDC13, 400 MHz) 6 = 161.0, 149.4, 141.2, 139.6, 130.4, 128.8, 126.9,
125.0, 123.3, 122.2,
120.8, 120.1, 112.4, 110.7, 109.4, 106.7, 103.8, 99.7, 69.6, 55.3, 48.0, 47.4.
ESI m/z: 425.0 [(M + H ), C22H21BrN202 (M) requires 421.1].
Example 37. P7C3-S37: N-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylamino)phenoxy)penty1)-2-(7-(dimethylamino)-2-oxo-2H-chromen-4-
yl)acetamide
Br Br
* *
N 0
YN . ON
H H
OH 0
N
0 0
1
The coumarin was attached to Example 62 Compound using a known procedure
(Alexander, et al.,
ChemBioChem, 2006, 7, 409-416.
Example 39. P7C3-S43: N-(2-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropoxy)ethyl)-acetamide
Br
* j¨N HAc
N¨ /0
41104 H 0
Br
Step 1. 2-(2-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropoxy)ethyl)
isoindoline-1,3-dione
Br Br
e = 0
N
=
YO N
OH 0
Sodium hydride dispersion (31.6 mg, 0.79 mmol) was added to a solution of N-(2-
hydroxyethyl)-
phthalimide (153.7 mg, 0.80 mmol) in anhydrous THF (1.2 ml, 0.67 M). The
suspension is stirred for 15
minutes before the addition of carbazole epoxide 2-A. The reaction was stirred
at room temperature for five
minutes and then at 60 C for 1 hour. The cooled reaction was diluted with
Et0Ac and washed with water. The
aqueous layer was extracted and the combined organics were filtered over a
celite pad. The Crude product was
used without further purification. Yield=44 %
151
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 8.12 (s, 2H), 7.85 (s, 2H), 7.72 (m, 2H), 7.55 (d, 2H,
J=8.5 Hz), 7.33
(d, 2H, J=8.7 Hz), 4.64 (d, 1H, J=16.1 Hz), 4.27 (d, 1H), 3.88 (m, 4H), 3.31
(bs, 1H), 2.80 (m, 1H), 2.48 (m,
1H), 2.04 (s, 1H).
MS (ESI), m/z: 614.9 [(M+HC00)-; C25H20Br2N204 (M) requires 570].
Step 2. 1-(2-aminoethoxy)-3-(3,6-dibromo-9H-carbazol-9-Apropan-2-ol
Br Br
= =
N
N H2
OH
Hydrazine hydrate (400 ul, 8.22 mmol) was added to a solution of the
phthalimide prepared in step 1
above (53 mg, 0.093 mmol) in ethanol (2.0 ml, 0.046 M). The reaction was
stirred overnight, condensed and
purified in 5-10% Me0H/DCM.
1H NMR (CDC13, 500 MHz) 8.11 (s, 2H), 7.53 (dd, 2H, J=8.7, 1.8 Hz), 7.38 (d,
2H, J=8.5 Hz), 4.37
(dm, 5H), 4.05 (t, 1H, J=6.8 Hz), 2.84 (m, 2H), 2.62 (m, 1H)
MS (ESI), m/z: 440.9 [(M+1) ; C17H18Br2N202 (M) requires 440.0].
Step 3. The title compound of Example 39 was prepared as follows.
Triethylamine (33.5 ul, 0.26
mmol) and acetic anhydride (17 ul, 0.18 mmol) were added to a solution of
amine XIII (71 mg, 0.16 mmol) in
THF (3.0 ml, 0.053 M). The reaction was stirred overnight. The reaction
mixture was diluted with Et0Ac,
washed with water, dried over Na2504, filtered and condensed. The crude
mixture was subjected to flash
chromatography (5% Me0H/CH2C12).
1H NMR (CDC13, 500 MHz) 8.13 (d, 2H, J=1.7 Hz, 7.55 (dd, 2H, J=8.7, 1.8 Hz),
7.34 (d, 2H, 9.1
Hz), 5.78 (bs, 1H), 4.35 (ddd, 3H, J=6.2, 6.8 Hz), 4.22 (m, 1H), 3.46 (m, 4H),
3.33 (dd, 1H, J=9.7, 5.4 Hz),
2.80 (bs, 1H), 1.98 (s, 3H)
MS (ESI), m/z: 482.9 [(M+1) ; C19H20Br2N203 (M) requires 482.0]
Example 40. P7C3-S44: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-3-
ylamino)propan-2-ol
Br
* ¨C?
/ HO
Br HN
Step 1. 5-((3,6-dibromo-9H-carbazol-9-yOmethyl)-3-(pyridin-3-y0oxazolidin-2-
one
152
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
ilk 0
04
Br N N \ --;
N
A mixture of the corresponding N-H oxazolidinone (0.0390g, 0.0920mmol), 3-
iodopyridine (0.0419g,
0.204mmol), CuI (0.0018g, 0. 00920mmol), and K2CO3 (0.0116g, 0.0837mmo1) in
0.5mL of DMSO was
heated at 130 C for 12 hours in a sealed vial. The reaction mixture was cooled
and diluted with 20 mL Et0Ac
and washed with water 2 x 10 ml- and brine 2 x 10mL. The organic layer was
dried over anhydrous Na2SO4
and evaporated to afford the crude product (0.0383g white solid, yield 83.7%),
which was used without further
purification.
1H NMR (CDC13, 400 MHz) 6=3.82 (dd, J= 9.1, 6.6 Hz, 1H) 4.12 (dd, J= 10.0, 7.9
Hz, 1H) 4.72 -
4.55 (m, 2H) 5.15 (td, J= 11.8, 5.4 Hz, 1H) 7.27 (dd, J= 8.3, 4.9 Hz, 1H) 7.34
(d, J= 8.7 Hz, 2H) 7.59 (dd, J
= 8.7, 1.9 Hz, 2H) 8.03 (ddd, J= 8.5, 2.6, 1.2 Hz, 1H) 8.14 (d, J= 1.9 Hz, 2H)
8.37 (d, J= 4.2 Hz, 1H) 8.44 (s,
1H). ESI m/z: 543.9 [(M + HC00-); C21H15Br2N302 (M) requires 499].
Step 2. The title compound of Example 40 was prepared as follows. Li0H+120
(0.0097 g, 0.231
mmol) was added to 543,6-dibromo-9H-carbazol-9-yl)methyl)-3-(pyridin-3-
y1)oxazolidin-2-one (0.0116g,
0.0231mmol) in a mixture of 265 [LI., THF and 29 [LI., H20 (v/v = 9:1). The
mixture was stirred at room
temperature for 7 days. The reaction mixture purified by silica gel
chromatography using CHC13/Me0H as
elute to afford 0.0087 g white solid as product, yield 79.3%.
1H NMR (CDC13, 600 MHz) 6 = 3.15 (dd, J= 12.6, 6.2 Hz, 1H) 3.30 (d, J= 11.8
Hz, 1H) 4.45 -4.33
(m, 3H) 6.81 (d, J= 7.4 Hz, 1H) 7.02 (s, br, 1H) 7.32 (d, J= 8.7 Hz, 2H) 7.52
(dd, J= 8.7, 1.8 Hz, 2H) 7.83 (s,
br, 2H) 8.11 (d, J= 1.6 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6 = 139.8, 139.5, 136.2, 130.0, 129.5, 124.1, 123.8,
123.5, 119.7, 112.8,
110.9, 69.0, 47.6, 47.3
ESI m/z: 517.9 [(M + HC00-); C20H17Br2N30 (M) requires 473].
Example 41. P7C3-S45: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-4-
ylamino)propan-2-ol
Br
* _
N-)N-CN
* HO
Br
Step 1. 5-((3,6-dibromo-9H-carbazol-9-yOmethyl)-3-(pyridin-4-y0oxazolidin-2-
one
153
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
* 0
04
Br 4. N,c/N---C\N
A mixture of the corresponding N-H oxazolidinone (0.0195g, 0.0460mmo1), 4-
iodopyridine (0.0209g,
0.102mmol), CuI (0.0009g, 0. 00460mmo1), and K2CO3 (0.0058g, 0.0418mmol) in
0.5mL of DMSO was at
130 C for 12 hours in a sealed vial. The reaction mixture was cooled and
diluted with 20 mL Et0Ac and
washed with brine (3 x 10mL). The organic layer was dried over anhydrous
Na2SO4 and evaporated to afford
the crude product, which was further triturated from CH2C12 suspension by
hexane to afford 0.0187g white
solid as product, yield 74.6%.
1H NMR (CDC13, 400 MHz) 6= 3.77 (dd, J= 9.4, 6.8 Hz, 1H) 4.08 (t, J= 9.0 Hz,
1H) 4.64 (d, J= 4.6
Hz, 2H) 5.23 -5.10 (m, 1H) 7.34 (d, J= 8.7 Hz, 2H) 7.37 (s, br, 2H) 7.61 (dd,
J= 8.6, 1.8 Hz, 2H) 8.16 (d, J=
1.8 Hz, 2H) 8.55 (s, br, 2H).
ESI m/z: 544.0 [(M + HC00-); C21H15Br2N302 (M) requires 499].
Step 2. The title compound of Example 41 was prepared as follows. Li0H.H20
(0.0157 g, 0.373
mmol) was added to 54(3,6-dibromo-9H-carbazol-9-yl)methyl)-3-(pyridin-4-
y1)oxazolidin-2-one (0.0187g,
0.0373mmo1) in a mixture of 428 [LL THF and 48 [LL H20 (v/v = 9:1). The
mixture was stirred at room
temperature for 3 days. The reaction mixture was diluted with 30 mL Et0Ac and
washed with brine 3 x 30
mL. The organic layer was dried over anhydrous Na2SO4 and evaporated to afford
the crude product, which
did not require purification (0.0013 g white solid, 7.3%).
1H NMR (d6-Acetone, 400 MHz) 6 = 3.33 (dd, J= 13.1, 6.4 Hz, 1H) 3.49 (dd, J=
13.2, 4.4 Hz, 1H)
4.41 (td, J= 7.6, 4.1 Hz, 1H) 4.51 (dd, J= 15.0, 7.6 Hz, 1H) 4.61 (dd, J=
14.8, 3.4 Hz, 1H) 6.61 (s, 2H) 7.56
(d, J= 8.6 Hz, 2H) 7.62 (d, J= 8.7 Hz, 2H) 8.10 (s, br, 2H) 8.37 (s, 2H)
13C NMR (d6-Acetone, 400 MHz) 6= 179.0, 149.6, 140.4, 129.0, 123.8, 123.3,
112.1, 111.8, 107.8,
68.8, 47.6, 46.4
ESI m/z: 517.9 [(M + HC00-); C20H17Br2N30 (M) requires 473].
Example 42. P7C3-S46: 1-(2,8-dimethy1-3,4-dihydro-1H-pyrido[4,3-13]indol-5(2H)-
y1)-3-
(phenylamino)propan-2-ol
H3C,
N
...--
N- 1-17 11
110 HO
H3C
154
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 42 was synthesized using a similar synthetic procedure analogous to
Representative
Procedure 2.
Example 43. P7C3-S59: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2,2-difluoropropy1)-
3-methoxyaniline
Br
* OMe
N¨ HN *
/ F F
5 Br
Step 1. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-oxopropy1)-N-(3-methoxypheny1)-4-
nitrobenzenesulfonamide
Br Br
= =
N
0
y,14, n OMe
0 /5-:3-
0/ fa
NO2
10 The nosylate of the title compound of Example 62 (prepared according to
the procedures described
herein) was oxidized with Dess-Martin periodinane using a procedure similar to
that described in Example
103. Quantitative yield.
1H NMR (CDC13, 500 MHz) 6 8.24 (d, 2H, J=8.9 Hz), 8.14 (s, 2H), 7.68 (d, 2H,
J=9.1 Hz), 7.53 (d,
2H, J=8.6 Hz), 7.18 (t, 1H, J=8.7 Hz), 7.05 (t, 2H, J=8.1 Hz), 6.87 (dd, 1H,
J=8.3, 2.5 Hz) 5.21, (s, 2H), 4.30
(s, 2H), 2.48 (s, 3H). MS (ESI), m/z: 683.9 [(M-1)- ; C28H21Br2N306S (M)
require 685.0].
Step 2. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2,2-difluoropropyl)-N-(3-
methoxypheny1)-4-
nitrobenzenesulfonamide
Br Br
= =
N
el
N , OMe
F F N/S:.µj
0/ =
NO2
155
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The title compound of Example 43, step 2 was prepared from the ketone prepared
in step 1 above
using a procedure similar to that described in Example 103. Yield was
quantitative and crude product was
used without additional purification.
1H NMR (CDC13, 500 MHz) 6 8.31 (d, 2H, J=8.9 Hz), 8.11 (s, 2H), 7.77 (d, 2H,
J=8.9 Hz), 7.55 (dd,
2H, J=8.7, 1.8 Hz), 7.25 (m, 3H), 6.92 (dd, 1H, J=8.3, 2.0 Hz), 6.73 (m, 1H)
6.61, (d, 1H, J=7.7 Hz), 4.78 (t,
2H, T=14.7 Hz), 4.18 (t, 2H, J=11.2 Hz), 3.78 (s, 3H).
MS (ESI), m/z: 751.9 [(M+HC00)-; C28H21Br2F2N305S (M) requires 707.0].
Step 3. The title compound of Example 43 was prepared as follows. The nosyl
group on N-(3-(3,6-
dibromo-9H-carbazol-9-y1)-2,2-difluoropropy1)-N-(3-methoxyphenyl)-4-
nitrobenzenesulfonamide was
removed using the procedure described in Representative Procedure 5.
1H NMR (CDC13, 400 MHz) 6 8.11 (d, 2H, J=1.6 Hz), 7.49 (dd, 2H, J=8.7, 2.0
Hz), 7.32 (d, 2H, J=8.9
Hz), 7.11 (t, 1H, J=8.2 Hz) 6.39 (dd, 1H, J=8.2, 2.3 Hz), 4.68 (t, 2H, J=13.2
Hz), 3.89 (t, 1H, J=7.0 Hz), 3.74
(s, 3H), 3.47 (m, 2H)
MS (ESI), m/z: 566.9 [(M+HC00)-; C22H18Br2F2N20 (M) requires 522.0].
Example 45. P7C3: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol
Br
Br 0 *
N
\........(0...H
NH
ilk
This compound can be purchased from ChemBridge Corporation.
Example 46. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(o-tolylamino)propan-2-ol
Br
Br. illt
N
v......c...0H
NH
0
This compound can be purchased from ChemBridge Corporation.
156
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 47. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(m-tolylamino)propan-2-ol
Br
Br I. lit
N
v.......c.C.:H
NH
This compound can be purchased from ChemBridge Corporation.
Example 48. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-methoxyphenylamino)propan-2-
ol
Br
Br 0 4.
N
v.....c...OH
NH ome
0
This compound can be purchased from ChemBridge Corporation.
Example 50. 1-(4-bromophenylamino)-3-(3,6-dichloro-9H-carbazol-9-yl)propan-2-
ol
CI
CI if
IW N
L.(OH
4
1 0 Br
This compound can be purchased from ChemBridge Corporation.
Example 51. 1-(4-bromophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
157
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 =
N
\-- NH
*
Br
This compound can be purchased from ChemBridge Corporation.
Example 52. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(4-ethoxyphenylamino)propan-2-
ol
Br
Br 0 fe
N
v....,(OH
V- NH
4
0 Et
This compound can be purchased from ChemBridge Corporation.
Example 53. 1-(4-chlorophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-
ol
Br
Br 0 it
N
µ......(OH
\-- NH
*
CI
This compound can be purchased from ChemBridge Corporation.
Example 54. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenethylamino)propan-2-ol
158
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br
N
k.....f0H
\--NH
0
This compound can be purchased from ChemBridge Corporation.
Example 55. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2-hydroxyethylamino)propan-2-
01
Br
Br* fik
N
L...i0H
\--NH
LA
OH
This compound can be purchased from ChemBridge Corporation.
Example 56. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2,4-
dimethoxyphenylamino)propan-2-01
B r
Br r .
Nk......(0 H
\
4
0
This compound can be purchased from ChemBridge Corporation.
Example 57. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2,3-dimethylphenylamino)propan-
2-01
Br
Br 0 46
N
k....,e0H
\--NH
4
159
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
This compound can be purchased from ChemDiv, Inc.
Example 58. 1-(2-chlorophenylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-
ol
Br
Br 0 4,
N
L....(OH
\--NH ci
4
This compound can be purchased from ChemDiv, Inc.
Example 59. 1-(tert-butylamino)-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
Br
Br 0 .
Nv...../OH
\¨NH
k
This compound can be purchased from ChemDiv, Inc.
Example 60. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(isopropylamino)propan-2-ol
Br
Br 0 *
N\.......(0_H
N H
This compound can be purchased from ChemDiv, Inc.
Example 61. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(4-methoxyphenylamino)propan-2-
ol
160
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 *
N
L...e0H
\--NH
*
OMe
This compound can be purchased from ChemDiv, Inc.
Example 62. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-methoxyphenylamino)propan-2-
ol
Br Br
* 0
Nr N 0
0
H 1
OH
This compound can be purchased from ChemDiv, Inc.
Example 63. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(m-tolylamino)propan-2-ol
Br Br
* *
NHN 0
H
OH
This compound can be purchased from ChemDiv, Inc.
Example 64. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,5-dimethylphenylamino)propan-
2-ol
Br Br
* *
NHN 0
H
OH
This compound can be purchased from ChemDiv, Inc.
Example 65. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,4-dimethylphenylamino)propan-
2-ol
161
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br Br
* 0
Nr N 0
H
0 H
This compound can be purchased from ChemDiv, Inc.
Example 66. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3,4-dimethylphenylamino)propan-
2-01
Br Br
* *
NH N 0
H
OH
This compound can be purchased from ChemDiv, Inc.
Example 67. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(2,5-dimethylphenylamino)propan-
2-01
Br Br
* *
NH N 40)
H
0 H
This compound can be purchased from ChemDiv, Inc.
Example 68. 1-(4-bromophenylamino)-3-(2,3-dimethy1-1H-indo1-1-y1)propan-2-01
I. N\
L...s0H
Br .N
H
This compound can be purchased from ChemBridge Corporation.
Example 69. 1-(2,3-dimethy1-1H-indo1-1-y1)-3-(4-methoxyphenylamino)propan-2-01
162
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
01 N\
\1OH
0 .N
H
This compound can be purchased from ChemBridge Corporation.
Example 70. 1-(2,3-dimethy1-1H-indo1-1-y1)-3-(4-ethoxyphenylamino)propan-2-ol
L...s0H
0 .
--_,..--
N
H
This compound can be purchased from ChemBridge Corporation.
Example 71. 1-(2,3-dimethy1-1H-indo1-1-y1)-3-(p-tolylamino)propan-2-ol
* N\
OH
. N
H
This compound can be purchased from ChemBridge Corporation.
Example 72. 1-(2,3-dimethy1-1H-indo1-1-y1)-3-(phenylamino)propan-2-ol oxalate
HO \
N
\1OH
0
ri( ISI
OH HN
0,
This compound can be purchased from ChemBridge Corporation.
163
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 73. 1-(1H-indo1-1-y1)-3-(4-methoxyphenylamino)propan-2-ol
hydrochloride
0 H H
NI-N 0
IP H CI 0
This compound can be purchased from ChemBridge Corporation.
Example 74. 1-(1H-indo1-1-y1)-3-(phenylamino)propan-2-ol oxalate
0 H H
*
HO __________________________________________ A
o
This compound can be purchased from ChemBridge Corporation.
Example 75. 1-(3,4-dihydro-1H-carbazol-9(2H)-y1)-3-(m-tolylamino)propan-2-ol
. =
N
0 HH
This compound can be purchased from ChemBridge Corporation.
Example 76. P7C3-S229: 1-(9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol
* 0
N
H 0
NH
I.1
This compound can be purchased from ChemBridge Corporation.
A separate batch was also synthesized independently. Specifically, following
representative procedure
2, 1-(9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol was prepared in 80% yield.
1H NMR (500 MHz, CDC13) 6 8.09 (d, J= 7.7 Hz, 2H), 7.45 (q, J= 8.2 Hz, 4H),
7.24 (d, J= 6.6 Hz,
2H), 7.17 (t, J= 7.6 Hz, 2H), 6.80 (t, J= 7.5 Hz, 2H), 6.71 (d, J= 7.8 Hz,
2H), 4.49 (s, 1H), 4.46 (d, J= 5.1
164
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Hz, 2H), 3.40 (d, J= 12.9 Hz, 1H), 3.28 (dd, J= 12.3, 7.5 Hz, 1H). ESI m/z:
317.1 ([M+H]+, C21I-120N20
requires 317.16)
Example 77. 1-(3,6-dichloro-9H-carbazol-9-y1)-3-(phenylamino)propan-2-ol
CI CI
* I.
N
HOJ
NH
1401
This compound can be purchased from ChemBridge Corporation.
Example 78. 1-(9H-carbazol-9-y1)-3-(p-tolylamino)propan-2-ol
* *
N
HO
NH
CH3
10 This compound can be purchased from ChemBridge Corporation.
Example 79. 1-(3,6-dichloro-9H-carbazol-9-y1)-3-(p-tolylamino)propan-2-ol
CI CI
* *
N
HOJ
NH
lei
CH3
This compound can be purchased from ChemBridge Corporation.
165
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 80. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(p-tolylamino)propan-2-ol
Br Br
* *
N
HO)
NH
1.1
CH3
This compound can be purchased from ChemBridge Corporation.
Example 81. N-(4-(3-(9H-carbazol-9-y1)-2-hydroxypropoxy)phenypacetamide
* 4
N
HO
0
140)
H3Cy NH
0
This compound can be purchased from ChemBridge Corporation.
Example 82. 1-(9H-carbazol-9-y1)-3-phenoxypropan-2-ol
* *
N
HO
0
SI
This compound can be purchased from ChemBridge Corporation.
166
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 83. 1-(9H-carbazol-9-y1)-3-(4-methoxyphenylamino)propan-2-ol
* *
N
HO)
N H
0
n.__. 3L, õ,0
This compound can be purchased from ChemBridge Corporation.
Example 84. 1-(benzylamino)-3-(9H-carbazol-9-yl)propan-2-ol
*0
N
HI 0OH
This compound can be purchased from ChemBridge Corporation.
Example 85. methyl 4-(3-(9H-carbazol-9-y1)-2-hydroxypropoxy)benzoate
*0 o
N
0
HO 0 1
CH 3
0
This compound can be purchased from ChemBridge Corporation.
Example 86. 1-(9H-carbazol-9-y1)-3-(4-methoxyphenoxy)propan-2-ol
* *
N yH3
HO) o
VI
No
This compound can be purchased from ChemBridge Corporation.
167
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 87. P7C3-S20: 1-amino-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
Br
Br 0 4.
Nv.......Ø..H
NH 2
This compound can be purchased from ChemBridge Corporation.
Example 88a. P7C3-S40: (S)-1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-
ol
Br Br
= .
N
Hr0 SI
OH
Example 88b. P7C3-S41: (R)-1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-
ol
Br Br
. .
N
OH
The title compounds of Examples 88a and 88b were prepared according to the
procedure described in
Example 3b except using the appropriate commercially available optically
active phenoxymethyl oxirane as
the epoxide starting material.
Example 89. P7C3-S42: 3,6-dibromo-9-(2-fluoro-3-phenoxypropy1)-9H-carbazole
Br Br
= *
Ncr.c) 0
F
The title compound of Example 89 was prepared according to the procedure
described in
Representative Procedure 4 except using the title compound of Example 3b as
the starting material. The crude
mixture was purified in 100% DCM (+0.2% TEA). Isolated yield=97%.
1H NMR (CDC13, 400 MHz) 6 8.13(d, 2H, J=1.7 Hz), 7.51 (dd, 2H, J= 8.7, 1.9
Hz), 7.29-7.35 (m, 4H),
168
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
7.01 (t, 1H,J= 7.5 Hz), 6.91 (d, 1H, J= 7.8 Hz), 5.16 (dddd, 1H, J= 4.5, 5.4,
9.7, 46.0 Hz), 4.59-4.79 (m, 2H),
4.03-4.17 (m, 2H).
MS (ESI), m/z: 519.9 [(M+HC00)-; C21H16Br2FNO (M) requires 475.0].
Example 90. P7C3-S54: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-2-
methylpropan-2-ol
Br
it OH
Br
H
O N
l'W
Step 1. Chlorohydrin-19
OH ,
ClIr\11 1. 0
l'W
m-Anisidine (0.18 mL, 1.62 mmol) was added to 2-chloromethy1-2-methyl oxirane
(0.154 mL, 1.62
mmol) in acetic acid (2 mL) and the mixture was heated to 75 C. Upon
completion the reaction was
neutralized with saturated sodium bicarbonate to pH 7, then extracted 3x with
Et0Ac, washed with brine and
dried with Mg504 filtered, and concentrated in vacuo. The crude residue was
purified by chromatography
(5i02, 0-25% Et0Ac/Hexane) to afford the desired alcohol (332 mg, 89%).
1H NMR (CDC13, 400 MHz) 6 7.08 ( t, 1H, J= 8.1 Hz), 6.29 (m, 2H), 6.23 (t, 1H,
J= 2.3 Hz), 3.95 (s,
NH), 3.77 (s, 3H), 3.60 (dd, 2H, J= 35.1, 11.0 Hz), 3.25 (dd, 2H, J= 44.8,
13.0 Hz), 2.31 (apparent d, OH),
1.36 (s, 3H) ESI m/z 230.1 ([M+H]+).
Step 2. Epoxide-20
,)N
H
0
0 \
Chlorohydrin-19 (0.166g, 0.722 mmol) was dissolved in dioxane (1 mL) and added
to a solution of
KOH (0.168mgs, 3.0 mmol). The reaction was followed by TLC (20% Et0Ac/Hexane)
until the starting
material was consumed and the less polar product was obtained. After aqueous
workup, the crude product was
used without purification.
169
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 7.07 ( t, 1H, J= 8.1 Hz), 6.27 (dd, 1H, J= 8.2, 0.8
Hz), 6.22 (dd, 1H, J
= 8.2, 0.8 Hz), 6.16 (t, 1H, J= 2.3 Hz), 3.83 (s, NH), 3.32 (br s, 2H), 2.82
(d, 1H, J= 4.5 Hz), 2.63 (d, 1H, J=
4.5 Hz).
Reference: Chemistry of Heterocyclic Compounds volume 41, No 4, 2005, pg 426.
Step 3. The title compound of Example 90 was prepared in 83% yield using 3,6-
dibromocarbazole,
sodium hydride (NaH), and epoxide 20. See, e.g., the procedure described in
Example 21, step 4.
1H NMR (CDC13, 400 MHz): 6 8.14 (s, 2H), 7.53 (d, 2H, J= 8.9 Hz), 7.42 (d, 2H,
J= 8.4 Hz), 7.09 (t,
1H, J= 8.4 Hz), 6.33 (d, 1H, J= 6.3 Hz), 6.27 (d, 1H, J= 6.3 Hz), 6.18 (s,
1H), 4.41 (d, 1H, J= 15.3 Hz), 4.32
(d, 1H, J= 15.3 Hz) 3.74 (s, NH), 3.49 (s, 3H), 3.28 (d, 1H, 12.4 Hz), 3.22
(d, 1H, 12.4 Hz), 2.03 (s, OH),
1.33 (s, 3H) ESI m/z 518.9 ([M+H]+).
13C NMR (CDC13, 100 MHz) 6 161.0, 149.8, 140.6 (2C), 130.4 (2C), 129.4 (2C),
123.8 (2C), 123.2
(2C), 112.8, 111.8 (2C), 106.9, 103.8, 99.8, 75.0, 55.4, 52.5, 51.5, 25.1
ESI m/z 516.9 ([M+I-1] , C23H22Br2N202 requires 516.04
Example 91. 1-(2,8-dimethy1-3,4-dihydro-1H-pyrido14,3-blindol-5(2H)-y1)-3-(3-
methoxyphenylamino)propan-2-ol
Me
N
Me 0\
N
NH
. OMe
Following a literature procedure (Zoidis et al., Bioorg. Med. Chem. 2009, /7,
1534-1541), the title
compound of Example 18 (0.015 g, 0.034 mmol) was dissolved in anhydrous THF
(0.34 mL) and cooled to 0
C. A solution of LAH (0.10 mL, 1.0 M in THF) was added dropwise, and the
reaction was stirred for 2 hat 0
C. Me0H was added to quench the remaining LAH and after 45 min, the mixture
was partitioned between
Et0Ac/H20. The organic layer was separated and the aqueous layer was extracted
with Et0Ac (3x), and the
combined organic layers were washed with satd. aq. NaC1, dried over Na2504,
filtered, and concentrated. The
crude residue was purified by column chromatography (5i02, 0-20% Me0H/Acetone
+ 1% Et3N), followed
by PTLC (10% Me0H/Acetone + 1% Et3N) to afford the desired product (0.6 mg,
5%).
170
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 6 = 7.14 (m, 2H), 7.04 (dd, 1H, J = 8.0, 8.0 Hz), 6.98
(d, 1H, J = 8.5
Hz), 6.27 (d, 1H, J = 8.0 Hz), 6.18 (d, 1H, J = 8.0 Hz), 6.12 (s, 1H), 4.14
(m, 1H), 4.10 (m, 1H), 4.01 (m,
1H), 3.72 (s, 3H), 3.20 (m, 1H), 3.06 (m, 1H), 2.72 (s, 3H), 2.41 (s, 3H).
ESI m/z 380.2 ([M+I-1] , C23H30N302 requires 380.2).
Example 92. P7C3-S48: 1-(4-azidophenylamino)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br
Br 110N-.....\
H0?--A
HN 0N3
4-Azidoaniline (0.038 g, 0.283 mmol) was added to a solution of 3,6-dibromo-9-
(oxiran-2-ylmethyl)-
9H-carbazole (0.100 g, 0.262 mmol) in THF (0.10 mL). LiBr (0.001 g, 0.013
mmol) was added and the
reaction was stirred at room temperature for 3 days. The reaction was purified
directly by chromatography
(Si02, 0-25% Et0Ac/Hexane) to afford the desired product (31 mg, 23%).
1H NMR (d6-acetone, 500 MHz) 6 = 8.36 (d, 2H, J = 2.0 Hz), 7.61 (m, 2H), 7.55
(m, 2H), 6.85 (m,
2H), 6.74 (m, 2H), 5.19 (br s, 1H), 4.61 (dd, 1H, J = 4.0, 15.0 Hz), 4.56 (br
s, 1H), 4.50 (dd, 1H, J = 8.0,
15.0 Hz), 4.39 (m, 1H), 3.39 (dd, 1H, J = 4.5, 13.0 Hz), 3.25 (dd, 1H, J =
6.5, 13.0 Hz).
13C NMR (acetone-d6, 100 MHz) 6 = 147.7, 141.1, 129.8 (2C), 128.9, 124.5,
124.0 (2C), 120.7 (2C),
114.9 (2C), 112.8 (2C), 112.6, 111.9, 69.6, 48.5, 48.4.
ESI m/z 513.9 ([M+I-1] , C21H18Br2N50 requires 514.0).
Example 93. P7C3-S47: 1-(3-azido-6-bromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol
N3
Br 0 *
N
NH
110 20 OMe
Step 1. 3-azido-6-bromo-9H-carbazole
171
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
N3
Br 0 4.
N
H
3,6-Dibromocarbazole (0.500 g, 1.538 mmol), NaN3 (0.120 g, 1.846 mmol), CuI
(0.029 g, 0.154
mmol), L-proline (0.053 g, 0.461 mmol) and NaOH (0.019 g, 0.461 mmol) were
dissolved in 7:3 Et0H/H20
(3.0 mL) and heated to 95 C under a N2 atmosphere for 24 h. The completed
reaction was partitioned
between Et0Ac/H20 (3x) and the combined organics were washed with satd. aq.
NaC1, dried over Na2SO4,
filtered, and concentrated. The crude residue was purified by chromatography
(Si02, 0-15% Et0Ac/toluene),
followed by HPLC (Phenomenex Si02 Luna 10 , 250x21.2 mm column, 50%
Et0Ac/Hexane, 21 mL/min,
retention time = 48 min) to afford the desired product.
1H NMR (CDC13, 500 MHz) 6 8.14 (s, 1H), 8.08 (br s, 1H), 7.64 (s, 1H), 7.50
(d, 1H, J = 8.5 Hz),
7.38 (d, 1H, J = 9.0 Hz), 7.29 (d, 1H, J = 8.5 Hz), 7.10 (d, 1H, J = 9.0 Hz).
ESI m/z 285.0 ([M¨H], C12H6BrN4 requires 285.0).
Step 2. The title compound of Example 93 was synthesized from 3-azido-6-bromo-
9H-carbazole in
46% yield using a procedure analogous to that described in Example 90, step 3.
1H NMR (CDC13, 500 MHz) 6 8.14 (d, 1H, J = 1.5 Hz), 7.64 (d, 1H, J = 2.0 Hz),
7.52 (dd, 1H, J =
1.5, 8.5 Hz), 7.40(d, 1H, J = 9.0 Hz), 7.31 (d, 1H, J = 8.5 Hz), 7.12 (dd, 1H,
J = 2.0, 8.5 Hz), 7.07 (dd, 1H,
J= 8.0, 8.0 Hz), 6.31 (dd, 1H, J = 2.0, 8.0 Hz), 6.21 (dd, 1H, J = 1.5, 8.0
Hz), 6.13 (dd, 1H, J = 2.0, 2.5
Hz), 4.39-4.35 (m, 3H), 3.71 (s, 3H), 3.31 (dd, 1H, J= 3.5, 13.0 Hz), 3.16
(dd, 1H, J = 7.0, 13.0 Hz), 2.17
(br s, 1H).
ESI m/z 466.0 ([M+1-1] , C22H21BrN502 requires 466.1).
Example 94. P7C3-S49: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(4-methoxyphenoxy)
propan-2-ol
Br
111 H 0
N---.)---j . OM e
0
Br
The title compound of Example 93 was synthesized from dibromocarbazole and (p-
methoxypheny1)-
glycidyl ether in 47% yield using a procedure analogous to those described in
Example 90, step 3 and
Example 93, step 2.
172
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 6 8.12 (d, 2H, J= 2.0 Hz), 7.50 (dd, 2H, J= 2.0, 8.5
Hz), 7.34 (d, 2H, J
= 8.5 Hz), 6.81 (m, 2H), 6.79 (m, 2H), 4.56 (m, 1H), 4.42 (m, 3H), 3.93 (dd,
1H, J= 4.5, 9.5 Hz), 3.81 (dd,
1H, J= 4.5, 9.5 Hz), 3.76 (s, 3H), 2.39 (d, 1H, J= 6.0 Hz).
13C NMR (acetone-d6, 100 MHz) 6 155.2, 153.8, 141.2 (2C), 129.8 (2C), 124.5
(2C), 124.0 (2C),
116.4 (2C), 115.5 (2C), 112.9 (2C), 112.5 (2C), 71.1, 69.8, 55.9, 47.4.
ESI m/z 547.9 ([M+CO21-1] , C23H20Br2NO5requires 548.0).
Example 95. P7C3-S52: 1-(3,6-dichloro-9H-carbazol-9-y1)-3-
(phenylsulfonyl)propan-2-ol
CI
itOHO
CI*0
Step 1. 1-(3,6-dichloro-9H-carbazol-9-y1)-3-(phenylthio)propan-2-ol
CI
1110 OH
CI* N,)S 0
The title compound of Example 95, step 1 was prepared using a procedure
analogous to that described
in Example 3a (white solid, 0.0293 g, yield 99.0%).
1H NMR (CDC13, 400 MHz) 6 = 2.55 (s, 1H) 2.97 (dd, J= 13.8, 7.2 Hz, 1H) 3.09
(dd, J= 13.9, 5.2
Hz, 1H) 4.20 ¨4.06 (m, 1H) 4.28 (dd, J= 15.0, 7.0 Hz, 1H) 4.41 (dd, J= 15.0,
4.1 Hz, 1H) 7.46 ¨7.14 (m,
9H) 7.93 (d, J= 1.8 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6 = 139.7, 134.5, 130.3, 129.5, 127.3, 126.8, 125.4,
123.3, 120.4, 110.6,
69.3, 48.2, 39.4
ESI m/z: 446.0, 436.0 [(M + HC00-), (M + Cl); C21H17C12NOS (M) requires
401.0].
Step 2. The title compound of Example 95 was prepared as follows. To a
solution of 1-(3,6-dichloro-
9H-carbazol-9-y1)-3-(phenylthio)propan-2-ol (0.0081 g, 0.0201 mmol) in 0.2 mL
CH2C12, a solution of
mCPBA (77%, 0.0113 g, 0.0503 mmol) in 0.2 mL CH2C12 was added dropwise. The
mixture was sealed and
stirred at rt overnight. The crude was diluted with 30 mL Et0Ac and washed
with saturated NaHCO3 (3 x 30
mL) and brine 1 x 30 mL. The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford the
crude product, which was subjected to silica gel chromatography using
Hexanes/Et0Ac to afford white solid
as product (0.0080 g, yield 91.3%).
173
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 = 3.17 (dd, J= 14.2, 3.0 Hz, 1H) 3.28 (dd, J= 14.3,
8.3 Hz, 1H) 3.29
(d, J= 2.9 Hz, 1H) 4.39 (d, J= 6.3 Hz, 2H) 4.67 (dtt, J= 8.7, 5.9, 3.0 Hz, 1H)
7.31 (d, J= 8.7 Hz, 2H) 7.40
(dd, J= 8.7, 2.0 Hz, 2H) 7.52 (t, J= 7.9 Hz, 2H) 7.66 (t, J= 7.5 Hz, 1H) 7.80
(d, J= 7.3 Hz, 2H) 7.96 (d, J=
2.0 Hz, 2H).
13C NMR (CDC13, 400 MHz) 6= 139.6, 138.8, 134.5, 129.8, 128.0, 127.0, 125.7,
123.5, 120.5, 110.5,
65.8, 60.0, 48.5
ESI m/z: 477.9 [(M + HC00-); C21H17C12NO3S (M) requires 433.0].
Example 96. P7C3-S53: 3,6-dibromo-9-(2-fluoro-3-(phenylsulfonyl)propy1)-9H-
carbazole
Br
* F 0,, SI
* N Sõ
0
Br
Step 1. 3,6-dibromo-9-(2-fluoro-3-(phenylthio)propy1)-9H-carbazole
Br
* F
Br * N S 0
The title compound of Example 96, step 1 was prepared by fluorination of the
title compound of
Example 31 using a procedure similar to that described in Representative
Procedure 4.
1H NMR (CDC13, 400 MHz) 6 = 3.09 (ddd, J= 14.2, 11.3, 8.4 Hz, 1H) 3.37 - 3.23
(m, 1H) 4.53 (ddd,
J= 20.8, 15.9, 6.7 Hz, 1H) 4.66 (ddd, J= 26.6, 15.9, 2.8 Hz, 1H) 5.04 -4.81
(m, 1H) 7.36 - 7.27 (m, 5H) 7.42
(dt, J= 3.2, 2.0 Hz, 2H) 7.54 (dd, J= 8.7, 1.9 Hz, 2H) 8.13 (d, J= 1.9 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6= 139.8, 134.3, 129.6, 129.5, 127.6, 123.9, 123.4,
112.9, 110.91 (d, J=
2.1 Hz, 1C) 92.2, 90.4, 46.16 (d, J= 22.8 Hz, 1C) 35.63 (d, J= 23.3 Hz, 1C)
Step 2. The title compound of Example 96 was prepared as follows. To a
solution of 3,6-dibromo-9-
(2-fluoro-3-(phenylthio)propy1)-9H-carbazole (0.0143 g, 0.0290 mmol) in 0.5 mL
CH2C12, a solution of
mCPBA (77%, 0.0162 g, 0.0725 mmol) in 0.5 mL CH2C12 was added dropwise. The
mixture was sealed and
stirred at rt overnight. The crude was diluted with 30 mL Et0Ac and washed
with saturated NaHCO3 3 x 30
ml- and brine 1 x 30 mL. The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford the
crude product, which was subjected to silica gel chromatography using
Hexanes/Et0Ac as elute to afford
white solid as product (0.0114 g, yield 74.8%).
174
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6= 3.61 - 3.40 (m, 2H) 4.56 (ddd, J= 22.4, 16.0, 6.6
Hz, 1H) 4.72 (dd, J
= 26.8, 15.9 Hz, 1H) 5.38 (dd, J= 47.1, 5.9 Hz, 1H) 7.34 (d, J= 8.7 Hz, 2H)
7.63 -7.53 (m, 4H) 7.68 (t, J =
7.4 Hz, 1H) 7.90 (d, J= 8.0 Hz, 2H) 8.12 (s, J = 2.0 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6=139.8, 134.7, 129.84, 129.79, 128.2, 124.1, 123.5,
113.3, 110.91,
110.89, 88.1, 86.3, 58.4, 58.1, 47.3, 47.1
ESI m/z: 557.9 [(M + Cl); C21H16Br2FNO2S (M) requires 522.9].
Example 97a. P7C3-S50: (S)-1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylsulfonyl)
propan-2-ol
Br
* OHO
. N 0
Br OS
Example 97b. P7C3-S51: (R)-1-(3,6-dibromo-9H-carbazol-9-y1)-3-(phenylsulfonyl)
propan-2-ol
Br
* OHO
N
Br 41i C) 110
The title compounds of Examples 97a and 97b were prepared from (S)- or (R)-3,6-
dibromo-9-
(oxiran-2-ylmethyl)-9H-carbazole using a procedure similar to that described
in Example 3d.
Preparation of (S)-3,6-dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole [(S)-epoxide
A]
Br Br
* *
N
I, (S)
"\10
To a solution of 3,6-dibromocarbazole (0.2194 g, 0.675 mmol) and
triphenylphosphine (0.1770 g,
0.675 mmol) in THF (5.4 mL) was added S-(-) -glycidol (44.8 [tL, 0.0500 g,
0.675 mmol). The reaction
mixture was cooled in an ice bath and diethyl azodicarboxylate (106.3 [tL,
0.1175 g, 0.675 mmol) was added.
The reaction mixture was allowed to warm to room temperature and stir
overnight. THF was removed under
vacuum and the residue was dissolved in 30 mL Et0Ac and washed with brine (3 x
30 mL). The organic layer
was dried over anhydrous Na2SO4 and evaporated to afford the crude product,
which was subjected to silica
gel chromatography using Hexanes/Et0Ac to afford white solid as product
(0.0514 g, yield 20.0%).
175
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 98. P7C3-S62: 1-(3,6-dicyclopropy1-9H-carbazol-9-y1)-3-(phenylamino)
propan-2-ol
1111
* OH H
V
Step 1. tert-butyl 3,6-dibromo-9H-carbazole-9-carboxylate
Br Br
. #
N
Boc
A solution of 3,6-dibromocarbazole (0.8288 g, 2.55 mmol) in 20 mL THF was
added to a suspension
of NaH (60%, 0.1122 g, 2.81 mmol) in 10 mL THF at -78 C. After stirring for 1
h, a solution of (Boc)20
anhydride (0.6122 g, 2.81 mmol) in 20 mL THF was added dorpwise into the
mixture. The reaction was
allowed to warm to room temperature and stir overnight. THF was removed under
vacuum and the residue was
dissolved in 30 mL Et0Ac and washed with 1M HC1 (2 x 30 mL) and brine (1 x 30
mL). The organic layer
was dried over anhydrous Na2SO4 and evaporated and the crude product was
subjected to silica gel
chromatography using Hexanes/Et0Ac to afford white solid as product (0.9890 g,
yield 91.7%).
1H NMR (CDC13, 400 MHz) 6 = 1.75 (s, 9H) 7.58 (dd, J= 8.9, 2.0 Hz, 1H) 8.05
(d, J= 1.8 Hz, 1H)
8.16 (d, J= 8.9 Hz, 1H).
13C NMR (CDC13, 400 MHz) 6 = 150.5, 137.5, 130.5, 126.3, 122.6, 117.9, 116.4,
84.9, 28.5.
Step 2. tert-butyl 3,6-dicyclopropy1-9H-carbazole-9-carboxylate
44 lib-
. *
N
Boc
Following a literature procedure (Petit et al., ChemMedChem 2009, 4, 261-
275.), tert-butyl 3,6-
dibromo-9H-carbazole-9-carboxylate (0.0200 g, 0.0470 mmol), cyclopropyl
boronic acid (0.0202 g, 0.235
mmol), palladium acetate (10 mol%, 0.0011 g, 0.00470 mmol), potassium
phosphate tribasic (0.0350g, 0.165
mmol), tricyclohexylphosphine (0.0026 g, 0.00941 mmol), water (12.2 [LI-) and
a stir bar were combined in a
176
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
sealed vial. The vial was sparged with N2 and charged with 0.22 mL degassed
toluene. The mixture was stirred
at 100 C for 65 h. The crude reaction mixture was diluted with 10 mL Et0Ac
and washed with brine (3 x 10
mL). The organic layer was dried over anhydrous Na2SO4 and evaporated to
afford the crude product, which
was used as is without further purification.
1H NMR (CDC13, 400 MHz) 6 = 0.82 - 0.76 (m, 4H) 1.02 (ddd, J= 8.4, 6.4, 4.4
Hz, 4H) 1.74 (s, 9H)
2.11 -2.01 (m, 2H) 7.19 (dd, J= 8.6, 1.9 Hz, 2H) 7.65 (d, J= 1.7 Hz, 2H) 8.14
(d, J= 8.5 Hz, 2H)
Step 3. 3,6-dicyclopropy1-9H-carbazole
411
To a solution of the corresponding N-Boc carbazole (0.0163 g, 0.0469 mmol) in
1 mL CH2C12, TFA
(144.8 [LI-, 1.876 mmol) was added dropwise. The mixture was sealed and
stirred at rt for 6 h. CH2C12 and
TFA were removed under vacuum. The residue was diluted with 30 mL Et0Ac and
washed with saturated
NaHCO3 3 x 30 mL. The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford the crude
product, which was subjected to silica gel chromatography using Hexanes/Et0Ac
as elute to afford white solid
as product (0.0139 g).
1H NMR (CDC13, 400 MHz) 6 = 0.77 (dt, J= 6.4, 4.5 Hz, 4H) 0.99 (ddd, J= 8.4,
6.2, 4.4 Hz, 4H) 2.13
-2.03 (m, 2H) 7.16 (dd, J= 8.4, 1.7 Hz, 2H) 7.28 (d, J= 8.4 Hz, 2H) 7.76 (d,
J= 1.1 Hz, 2H) 7.83 (s, br, 1H).
Step 4. 3,6-dicyclopropy1-9-(oxiran-2-ylmethyl)-9H-carbazole
411P
0
*
The title compound of Example 98, step 4 was prepared from 3,6-dicyclopropy1-
9H-carbazole using a
procedure similar to that described in Representative Procedure 1.
1H NMR (CDC13, 400 MHz) 6 = 0.81 -0.74 (m, 4H) 1.03 - 0.96 (m, 4H) 2.09 (ddd,
J= 14.4, 8.9, 5.6
Hz, 2H) 2.53 (dd, J= 4.8, 2.6 Hz, 1H) 2.77 (t, J= 4.3 Hz, 1H) 3.30 (dt, J=
7.4, 3.9 Hz, 1H) 4.35 (dd, J= 15.8,
4.6 Hz, 1H) 4.54 (dd, J= 15.8, 3.4 Hz, 1H) 7.22 (dd, J= 8.4, 1.7 Hz, 2H) 7.31
(d, J= 8.4 Hz, 2H) 7.78 (d, J=
1.1 Hz, 2H).
177
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Step 5. The title compound of Example 98 was prepared from 3,6-dicyclopropy1-9-
(oxiran-2-
ylmethyl)-9H-carbazole using a procedure similar to that described in
Representative Procedure 2.
1H NMR (CDC13, 600 MHz) 6 = 0.79 - 0.75 (m, 4H) 0.99 (td, J= 6.2, 4.6 Hz, 4H)
2.08 (ddd, J= 13.6,
8.5, 5.1 Hz, 2H) 3.21 (dd, J= 12.9, 5.6 Hz, 1H) 3.35 (d, J= 13.8 Hz, 1H) 4.39
(s, J= 23.7 Hz, 3H) 6.62 (d, J=
8.4 Hz, 2H) 6.75 (t, J= 7.3 Hz, 1H) 7.17 (t, J= 7.9 Hz, 2H) 7.20 (dd, J= 8.4,
1.1 Hz, 2H) 7.32 (d, J= 8.4 Hz,
2H) 7.78 (s, 2H)
13C NMR (CDC13, 500 MHz) 6 = 148.2, 139.8, 134.9, 129.6, 124.8, 123.2, 118.5,
117.5, 113.7, 108.8,
69.8, 48.0, 47.6, 15.7, 9.1
ESI m/z: 441.2 [(M + HC00-); C27H28N20 (M) requires 396.2].
Example 99. P7C3-S63: 1-(3,6-diiodo-9H-carbazol-9-y1)-3-(phenylamino)propan-2-
ol
I
4 OH H
* NN 0
I
Step 1. 3,6-diiodo-9-(oxiran-2-ylmethyl)-9H-carbazole
i
*
0
* NI"
I
The title compound of Example 99, step 1 was prepared from 3,6-diiodo
carbazole (Maegawa et al.,
Tetrahedron Lett. 2006, 47, 6957-6960) using a procedure similar to that
described in Representative
Procedure 1.
1H NMR (CDC13, 400 MHz) 6 = 2.48 (dd, J= 4.6, 2.6 Hz, 1H) 2.80 (t, J= 4.3 Hz,
1H) 3.37 - 3.24 (m,
1H) 4.28 (dd, J= 16.0, 5.1 Hz, 1H) 4.64 (dd, J= 15.9, 2.7 Hz, 1H) 7.24 (d, J=
8.6 Hz, 2H) 7.73 (dd, J = 8.6,
1.6 Hz, 2H) 8.33 (d, J= 1.7 Hz, 2H)
13C NMR (CDC13, 500 MHz) 6 = 140.0, 135.0, 129.5, 124.3, 111.3, 82.6, 50.6,
45.2, 44.9
Step 2. The title compound of Example 99 was prepared from 3,6-diiodo-9-
(oxiran-2-ylmethyl)-9H-
carbazole using a procedure similar to that described in Representative
Procedure 1.
178
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 = 2.92 (s, br, 1H) 3.19 (dd, J= 12.8, 6.1 Hz, 1H)
3.33 (d, J= 10.9 Hz,
1H) 4.49 -4.29 (m, 3H) 6.63 (d, J= 8.3 Hz, 2H) 6.78 (t, J= 7.3 Hz, 1H) 7.20
(t, J= 7.7 Hz, 2H) 7.28 (d, J=
2.5 Hz, 2H) 7.72 (d, J= 8.6 Hz, 2H) 8.35 (s, 2H).
13C NMR (CDC13, 400 MHz) 6 = 147.9, 140.1, 135.1, 129.65, 129.63, 124.4,
118.9, 113.7, 111.5,
82.6, 69.6, 48.0, 47.3
ESI m/z: 613.0 [(M + HC00-); C21H18I2N20 (M) requires 568.0].
Example 100. P7C3-S64: 1-(3,6-diethyny1-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino) propan-2-ol
H
\\
*
OH H
* NN 0 OMe
--
H ----
Step 1. 1-(3,6-bis((triisopropylsily0ethyny1)-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-
ol
TIPS
\\
* OH H
* NN 0 OMe
----
TIPS
The title compound of Example 62 (0.0112 g, 0.0222 mmol),
bis(benzonitrile)dichloropalladium (3
mol%, 0.0003 g, 0.0007 mmol), [(tBu)3PH]BF4 (6.2 mol%, 0.0004 g, 0.0014 mmol),
copper(I) iodide (2
mol%, 0.0001g, 0.0004 mmol), DABCO (0.0060 g, 0.0533 mmol) were combined under
an N2 atmosphere.
Degassed dioxane (0.1 mL) was added, and the resulting solution was stirred at
room temperature for 10 min.
Trimethylsilylacetylene (11.8 [tt, 0.0533 mmoL) was added into the mixture via
microsyringe. The mixture
was then stirred at rt overnight. The crude reaction mixture was diluted with
10 mL Et0Ac and washed with
brine (3 x 10 mL). The organic layer was dried over anhydrous Na2SO4 and
evaporated to afford the crude
product, which was subjected to silica gel chromatography using Hexanes/Et0Ac
to afford colorless oil as
product (0.0152 g, yield 96.8%).
1H NMR (CDC13, 400 MHz) 6 = 1.22 - 1.13 (m, 42H) 2.24 (s, br, 1H) 3.17 (dd, J=
12.6, 6.7 Hz, 1H)
3.31 (d, J= 12.1 Hz, 1H) 3.71 (s, 3H) 4.48 -4.31 (m, 3H) 6.12 (t, J= 2.1 Hz,
1H) 6.22 (dd, J= 8.0, 1.8 Hz,
179
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H) 6.31 (dd, J= 8.1, 2.1 Hz, 1H) 7.07 (t, J= 8.1 Hz, 1H) 7.37 (d, J= 8.5 Hz,
2H) 7.58 (dd, J= 8.5, 1.5 Hz,
2H) 8.22 (d, J= 1.4 Hz, 2H)
13C NMR (CDC13, 400 MHz) 6 = 171.5, 161.0, 149.3, 140.9, 130.6, 130.4, 124.9,
122.7, 115.1, 109.3,
108.2, 106.7, 103.9, 99.7, 88.7, 69.5, 55.3, 47.4, 19.0, 11.6
Step 2. The title compound of Example 100 was prepared as follows. To a
solution of 143,6-
bis((triisopropylsilyl)ethyny1)-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)propan-2-ol (0.0152 g, 0.0215
mmol) in 200 [LI., anhydrous THF, a solution of TBAF in THF (1 M, 64.5 [tt,
0.0645 mmol) and acetic acid
(2.5 [tt, 0.0430 mmol) were added. The mixture was sealed and stirred under N2
atmosphere at rt for 27 h until
TLC showed the complete disappearance of starting material. The crude was
diluted with 10 mL Et0Ac and
washed with saturated NaHCO3 (3 x 10) mL. The organic layer was dried over
anhydrous Na2SO4 and
evaporated to afford the crude product, which was subjected to silica gel
chromatography using
Hexanes/Et0Ac to afford white solid as product (0.0061 g, yield 71.9%).
1H NMR (CDC13, 400 MHz) 6 = 2.24 (s, br, 1H) 3.09 (s, 2H) 3.20 (s, br, 1H)
3.32 (s, br, 1H) 3.72 (s,
3H) 4.48 -4.27 (m, 3H) 6.14 (s, 1H) 6.23 (dd, J= 8.0, 1.4 Hz, 1H) 6.32 (dd, J=
8.2, 1.8 Hz, 1H) 7.08 (t, J=
8.1 Hz, 1H) 7.40 (d, J= 8.5 Hz, 2H) 7.59 (dd, J= 8.5, 1.4 Hz, 2H) 8.21 (d, J=
1.1 Hz, 2H)
13C NMR (CDC13, 500 MHz) 6 = 161.1, 149.3, 141.2, 130.7, 130.4, 125.0, 122.7,
113.6, 109.6, 106.7,
103.8, 99.8, 84.7, 76.0, 69.6, 55.3, 48.0, 47.4
ESI m/z: 439.1 [(M + HC00-); C26H22N202 (M) requires 394.2].
Example 101. P7C3-S65: 9-(2-hydroxy-3-(3-methoxyphenylamino)propy1)-9H-
carbazole-3,6-
dicarbonitrile
NC
sit OH
H
. N N r OMe
NC
LW
Following a literature procedure (Weissman et al., J. Org. Chem. 2005, 70,
1508-1510), the title
compound of Example 62 (0.0252 g, 0.05 mmol), potassium hexacyanoferrate(II)
trihydrate (0.0106 g, 0.025
mmol), sodium bicarbonate (0.0106 g, 0.1 mmol) and palladium acetate (1 mol %,
0.0001 g) were combined
under a N2 atmosphere. Anhydrous dimethylacetamide (0.1 mL) was added, and the
reaction mixture was
stirred at 120 C overnight. The crude reaction mixture was diluted with 10 mL
Et0Ac and washed with water
(2 x 10 mL) and brine (1 x 30 mL). The organic layer was dried over anhydrous
Na2SO4 and evaporated to
180
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
afford the crude product, which was subjected to silica gel chromatography
using Hexanes/Et0Ac to afford
white solid as product (0.0110 g, yield 54.6%).
1H NMR (d6-acetone, 400 MHz) 6 = 2.81 (s, 1 H) 3.36 -3.28 (m, 1H) 3.50- 3.43
(m, 1H) 3.71 (s, 3
H) 4.44 (s, br, 1 H) 4.66 (dd, J= 15.0, 8.5 Hz, 1 H) 4.77 (dd, J= 15.1, 3.4
Hz, 1 H) 5.16 (t, J= 5.8 Hz, 1H)
6.22 (dd, J= 8.1, 2.1 Hz, 1H) 6.27 (t, J= 2.0 Hz, 1H) 6.31 (dd, J= 8.1, 1.2
Hz, 1H) 7.01 (t, J= 8.1 Hz, 1H)
7.84 (dd, J= 8.6, 1.2 Hz, 2H) 7.91 (d, J= 8.6 Hz, 2H) 8.74 (s, 2H)
13C NMR (d6-acetone, 500 MHz) 6 =161.3, 150.4, 143.9, 130.02, 129.95, 126.0,
122.4, 119.8, 112.0,
106.0, 103.3, 102.5, 98.9, 69.0, 54.5, 48.0, 47.7
ESI m/z: 441.1 [(M + HC00-); C24H20N402 (M) requires 396.2).
Example 102. P7C3-S55: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropypaniline
Br Br
. =
N
H
F
Step 1. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-4-nitro-N-
phenylbenzenesulfonamide
Br Br
= .
N
YThsl, el
F d',
NO2
The title compound of Example 102, step 1 was prepared from epoxide 2-A and Ns-
aniline using
procedures similar to those described in representative procedures 3 and 4.
The crude mixture was purified in
40% Et0Ac/hexanes(+0.1% TEA). The isolated yield was 60%.
1H NMR ((CD3)2C0)3, 400 MHz) 6 8.37(m, 2H), 7.90 (m, 2H), 7.68 (m, 1H), 7.53-
7.60 (m, 6H),
7.32-7.40 (m, 5H), 5.03 (dm, 1H), 4.71-4.93 (m, 2H), 4.27-4.41 (m, 2H).
MS (ESI), m/z: 703.9 [(M+HC00)-; C27H20Br2FN304S (M) requires 659.0]
Step 2. The title compound of Example 102 was prepared as follows. Cesium
carbonate (11.5 mg,
0.036 mmol), the nosylate prepared in step 1 above (7.9 mg, 0.012 mmol), THF
(0.7 ml, 0.017 M) and
benezenthiol (3.8 ul, 0.037 mmol) were combined and stirred overnight. The
crude reaction mixture was
181
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
diluted with Et0Ac, washed with water and brine. The organic layer was dried
over Na2SO4, filtered and
condensed. Chromatographic purification on Si02 (20% Et0Ac/hexanes (0.2% TEA))
provided 74% (4.2 mg).
1H NMR (CDC13, 500 MHz) 6 = 8.16 (s, 2H), 7.56 (d, 2H, J=8.5 Hz), 7.31 (d, 2H,
J=8.5 Hz), 7.21 (t,
2H, J=7.4 Hz), 6.80 (t, 1H, J=7.3 Hz), 6.62 (d, 2H, J=8.5 Hz), 5.11 (dddd, 1H,
J=5.4, 5.4, 10.4, 47.4 Hz), 4.52-
4.68 (m, 2H), 3.94 (t, 1H, J=6.02 Hz), 3.30-3.51, (dm, 2H).
MS (ESI), m/z: 475.0 [(M+1)-; C21H17Br2FN2 (M) requires 474.0].
Example 103. P7C3-S56: 3,6-dibromo-9-(2,2-difluoro-3-phenoxypropy1)-9H-
carbazole
Br Br
= =
1411
F F
Step 1. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-one
Br Br
=
H0
r
0
Dess-Martin periodinane (58.2 mg, 0.137 mmol) was charged to a solution of the
title compound of Exam
3b (45.0 mg, 0.095 mmol) in dichloromethane (1.0 ml, 0.095 M). After two hours
the reaction mixture was dilutec
with Et0Ac and washed with saturated sodium thiosulfate solution, water and
brine. The organic layer was dried c
Na2504, filtered and condensed. The crude product was used without additional
purification. Yield = 74%
1H NMR (CDC13, 400 MHz) 6 8.15 (d, 2H, J=1.9 Hz), 7.52 (dd, 2H, J=8.6, 1.9 Hz)
7.35 (m, 2H), 7.08
(t, 1H, J=7.3 Hz), 7.04 (d, 2H, J=8.9 Hz), 6.91 (m, 2H), 5.29 (s, 2H), 4.68
(m, 2H)
MS (ESI), m/z: 469.9 [(M-1)- ; C21H15Br2NO2 (M) requires 570.9].
Step 2. The title compound of Example 103 was prepared as follows.
Diethylaminosulfur trifluoride
(39 ul, 0.30 mmol) was added dropwise to a solution of 1-(3,6-dibromo-9H-
carbazol-9-y1)-3-phenoxypropan-
2-one (33.3 mg, 0.070 mmol) in anhydrous dichloromethane (1.5 ml, 0.047M). The
reaction was quenched
with saturated sodium bicarbonate solution, and then extracting three times
with dichloromethane. The organic
layer is dried over Na2504, filtered and condensed. The crude mixture was
purified on 5i02 (10%
Et0Ac/hexanes +0.2% TEA. Isolated yield was 69 %.
182
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 8.09 (d, 2H, J=1.9 Hz), 7.48 (dd, 2H, J=8.7, 1.8 Hz)
7.30-7.4 (m, 4H),
7.06 (t, 1H, J=7.3 Hz), 6.91 (d, 2H, J=7.9 Hz), 4.79 (t, 2H, J=12.4 Hz), 4.07
(t, 2H, J=11.1Hz).
MS (ESI), m/z: 537.9 [(M+HC00)-; C21H15Br2F2NO (M) requires 492.9].
Example 104. P7C3-S60: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-4-
methoxyaniline
Br Br
I. = OMe
YN
N
el
H
F
Step 1. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(4-
methoxypheny1)-4-
nitrobenzenesulfonamide
Br N Br
= = 0 OMe
N ,
OH ;Vµj
0/ =
NO2
The title compound of Example 104, step 1 was prepared from epoxide 2-A and Ns-
anisidine
according to Representative Procedure 3. Yield=71%
1H NMR (CDC13, 400 MHz) 6 8.29 (d, 2H, J=8.7 Hz), 8.11 (d, 2H, J=1.9 Hz), 7.71
(,2H, J=8.6 Hz),
7.52 (dd, 2H, J=8.6, 1.9 Hz), 7.23 (d, 2H, J=8.9 Hz), 6.94 (d, 2H, J=8.9 Hz),
6.82 (d, 2H, J=8.9 Hz), 4.44 (dd,
1H, J=14.8, 3.8 Hz), 4.30 (m, 1H), 4.21 (bs, 1H), 3.81 (s, 3H), 3.69 (m, 2H).
MS (ESI), m/z: 732.0 [(M+HC00-); C28H23Br2N306S (M) requires 687.0]
Step 2. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-N-(4-
methoxypheny1)-4-
nitrobenzenesulfonamide
Br N Br
. AP OMe
H-,N =
F0
or,e
NO2
183
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The title compound of Example 104, step 2 was prepared from the nosylate
prepared in step 1 above
according to General Procedure 4. Yield=61.5%
1H NMR (CDC13, 400 MHz) 6 8.27 (m, 2H), 8.09 (m, 2H), 7.71 (d, 2H, J=7.41 Hz),
7.53 (m, 2H), 7.19
(m, 2H), 6.95 (d, 2H, J=8.8 Hz), 6.82 (d, 2H, J=8.8 Hz), 4.92 (dm, 1H, Jd=48.3
Hz), 4.55 (m, 2H), 3.88 (m,
2H), 3.79 (s, 3H).
MS (ESI), m/z: 734.0 (M+HC00)-; C28H22Br2FN305S (M) requires 689.0]
Step 3. The title compound of Example 104 was prepared according to
Representative Procedure 5.
Isolated yield 70%.
1H NMR (CDC13, 400 MHz) 6 8.14 (m, 2H0, 7.53 (dt, 2H, J=8.8, 1.6 Hz), 7.30 (d,
2H, 8.6 Hz), 6.78
(d, 2H, J=7.9 Hz), 6.57 (d, 2H, J=7.9 Hz), 5.07 (dddd, 1H, J=4.7, 6.1, 9.4,
47.7), 4.58 (m, 2H), 3.75 (s, 3H),
3.32 (m, 2H).
MS (ESI), m/z: 549. 0 [(M+HC00)-; C22H19Br2FN20 (M) requires 505.0).
Example 105. P7C3-S67: N-(2-bromo-3-(3,6-dibromo-9H-carbazol-9-yl)propy1)-N-(4-
methoxypheny1)-
4-nitrobenzenesulfonamide
Br Br
= N .
N 0 OMe
Y.H
Br
Step 1. N-(2-bromo-3-(3,6-dibromo-9H-carbazol-9-Apropy1)-N-(4-methoxypheny1)-4-
nitrobenzenesulfonamide
Br Br
. N 40 OMe
YNI, in
Br /5----
01 fa,
NO2
A solution of the title compound Example 104, Step 1 (20.5 mg, 0.030 mmol) in
anhydrous
dichloromethane (1.0 ml, 0.03 M) was cooled in an ice bath before the addition
of BBr3 (7 ul, 0.074 mmol).
After lh the reaction was diluted with Et0Ac, washed twice with water,
saturated sodium bicarbonate solution
184
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
and brine. The organic layer was dried over Na2SO4, filtered and condensed.
The crude mixture was purified in
100% CH2C12 (+0.2% TEA). Isolated yield =56%.
1H NMR (CDC13, 500 MHz) 6 8.26 (d, 2H, J=8.9 Hz), 8.12 (d, 2H, J=1.7 Hz), 7.60
(d, 2H, J=8.8 Hz)
7.53 (dd, 2H, J=8.7, 1.9 Hz), 7.18 (d, 2H, J=8.7 Hz), 6.89 (d, 2H, J=8.9 Hz)
6.81 (d, 2.H, J=9.0 Hz), 4.86 (dd,
1H, J=15.6, 5.4 Hz), 4.57 (m, 1H), 4.44 (m, 1H), 3.92 (m, 2H), 3.82 (s, 3H).
MS (ESI), m/z: 747.9 [(M-1)-;
C28H22Br3N305S (M) requires 748.9]
Step 2. The title compound of Example 105 was prepared from the nosylate
prepared in step 1 above
according to Representative Procedure 5. Isolated yield = 43% in appoximately
90% purity.
1H NMR (CDC13, 400 MHz) 6 8.14 (d, 2H, J=1.7 Hz), 7.51 (dd, 2H, J=8.6, 1.9
Hz), 7.28 (d, 2H, J=8.7
Hz), 6.71 (d, 2H, J=8.9 Hz), 6.41 (d, 2H, J=8.8 Hz), 4.84 (m, 1H), 4.63 (m,
3H), 3.82 (m, 1H), 3.73 (s, 3H).
MS (ESI), m/z: 564.8 [(M+1) ; C22H19Br3N20 requires 563.9].
The title compounds of Examples 106-109 can be prepared using the methods
described herein and/or
using conventional synthesis methods.
Example 106. P7C3-S61: Ethyl 2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)acetate
Br Br
= = 10 0Et
N
y-,N I. 0
H
F
Example 107. P7C3-S66: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-4-
(2-(2-
methoxyethoxy)ethoxy)aniline
Br = Br
=
N 0 OcIO
H,N
H
F
Step 1. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(4-
methoxypheny1)-4-
nitrobenzenesulfonamide
185
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
r
OCH.
6A
s,
,
The title compound was prepared according to Representative Procedure 3.
Step 2. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-N-(4-
hydroxypheny1)-4-
nitrobenzenesulfonamide
Br Br
= = OH
y, 0
OH /P
0
NO2
Boron tribromide (290 ul, 3.06 mmol) was added to solution of the product of
Step 1 (598 mg, 0.87
mmol) in anhydrous dichloromethane (17.0 ml) at 0 C. The reaction mixture was
condensed, diluted with
ethyl acetate and washed with water, saturated sodium bicarbonate, water and
then brine. Pure product was
isolated from column chromatography of the crude mixture in 1% Me0H/DCM.
Yield=59%
1H NMR (CD3)2CO3 500 MHz) 6 8.42 (d, 2H, J= 8.8 Hz), 8.35 (s, 2H), 7.87 (d,
2H, J= 8.8 Hz), 7.56
(dd, 2H, J= 1.7, 8.8 Hz), 7.49 (d, 2H, J= 8.9 Hz) 7.05 (d, 2H, J= 8.7 Hz),
6.81 (d, 2H, J= 8.6 Hz), 4.59 (dd,
1H, J= 2.9, 15.2 Hz), 4.53 (d, 1H, J= 5.5 Hz), 4.15 (m, 1H), 3.87 (m, 1H).
Step 3. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-N-(4-
hydroxypheny1)-4-
nitrobenzenesulfonamide
Br Br
= = OH
y,
0
NO2
186
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The product of Step 2 was fluorinated according to Representative Procedure 4.
Pure product was
obtained after column chromatography in 1% Me0H/DCM (+0.2% TEA). Yield=89%.
1H NMR (CD3)2CO, 400 MHz) 6 8.48 (d, 2H, J= 9.0 Hz), 8.41 (d, 2H, J= 1.7 Hz),
7.94 (d, 2H, J =
8.6 Hz), 7.66 (dd, 2H, J= 1.9, 8.8 Hz), 7.60 (d, 2H, J= 8.8 Hz), 7.10 (d, 2H,
J= 9.0 Hz), 6.89 (d, 2H, J= 8.8
Hz), 5.10 (dm, 1H), 4.74-4.94 (m, 2H), 4.20-4.32 (m, 2H).
Step 4. N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropyl)-N-(4-(2-(2-
methoxyethoxy)ethoxy)pheny1)-4-nitrobenzenesulfonamide
Br . Br
. 0 Oc:10
N
y,N1, ,.....0
0 F/,
NO2
A solution of the product of Step 3 (15.9 mg, 0.023 mmol), potassium carbonate
(13.6 mg, 0.098
mmol) and 1-bromo-2-(2-methoxyethoxy)ethane (8.5 mg, 0.041 mmol) in
dimethylformamide (1.0 ml) was
heated at 70 C overnight. The reaction was diluted with Et0Ac and washed with
water several times, then
brine. Column chromatography in 100% DCM (+0.2% TEA) - 1% Me0H/DCM (+0.2% TEA)
gave the pure
product. Yield= 43%.
1H NMR (CDC13, 500 MHz) 6 8.30 (d, 2H, J= 8.9 Hz), 8.14 (d, 2H, J= 1.7 Hz),
7.72 (d, 2H, J = 8.8
Hz), 7.56 (dd, 2H, J= 1.8, 8.6 Hz), 7.23 (d, 2H, J= 8.8 Hz), 6.95 (d, 2H, J=
8.7Hz), 6.85 (d, 2H, J = 8.7 Hz),
4.93 (dm, 1H), 4.46-4.69 (m, 2H), 4.13 (t, 2H, J= 5.2 Hz), 3.85 ¨ 3.91 (m,3H),
3.72 (m, 2H), 3.58 (m, 2H),
3.46-3.50 (m, 1H), 3.39 (s, 3H). MS (ESI), m/z: 824.0 (M+HC00)-
Step 5. P7C3-S66: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropyl)-4-(2-(2-
methoxyethoxy)ethoxy)aniline
Br Br
e '0
N 0 00::0
Y.N
F H
The nitrosulfonyl group was removed from the product of Step 4 via
Representative Procedure 5. Pure
product was isolated following preparative TLC. Yield=92%
1H NMR (CDC13, 400 MHz) 6 8.15 (d, 2H, J= 1.8 Hz), 7.55 (dd, 2H, J= 1.9, 8.7
Hz), 7.30 (d, 2H, J =
8.6 Hz), 6.81 (d, 2H, J= 8.9 Hz), 6.57 (d, 2H, J= 9.2 Hz), 5.08 (dm, 1H, 1
JH_F = 47.8 Hz), 4.50-4.69 (m, 2H),
4.08 (m, 2H), 3.84 (m, 2H), 3.66-3.75 (m, 2H), 3.59 (m, 2H), 3.40 (s, 3H),
3.27-3.45 (m, 2H). MS (ESI), m/z:
calculated 594.31, found 595 (M+1) .
187
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Example 108. P7C3-S68: N-(2-(2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)acetamido)ethyl)-5-(2-oxohexahydro-1H-thieno[3,4-
d]imidazol-4-
yppentanamide
Br Br
. .
N OYH
N N)=,,,,
NH
0 H b OCH
H--N
H
F
The title compound of Example 108 (P7C3-S68) was prepared via alkylation of
the product of Step 3
in the synthesis of Example 107 Compound (P7C3-566) with iodoethyl acetate and
subsequent amidation and
desulfonylation. The product was purified by preparative TLC in 10%
Me0H/CH2C12 (+0.2% TEA). 1H NMR
(CD30D, 500 MHz) 6 = 8.23 (s, 2H), 7.51 (dd, 4H, J= 31.0, 8.8, Hz), 6.84 (d,
2H, J=8.9 Hz) 6.67 (d, 2H, J
=8.6 Hz), 5.04 (dm, 1H, J= 48.9 Hz), 4.69 (d, 1H, J= 5.2 Hz) 4.65 (m, 1H),
3.37-3.42 (m, 3H), 4.17 (m, 1H),
3.42-3.52 (m, 1H), 3.37 (m, 4H) 3.05 (m, 1H), 2.82 (dm, 1H), 2.69 (m, 1H),
2.63 (d, 1H, J= 12.7 Hz), 2.13-
2.18 (m, 2H), 1.15-1.69 (m, 6H). 13C NMR (CDC13, 126 MHz) 6 = 176.6, 166.0,
151.7, 144.6, 141.2, 130.3,
124.9, 124.1, 117.1, 115.5, 113.4, 112.4, 106.2, 92.6 (d, 1J= 176.7 Hz), 69.2,
63.3, 61.6, 56.9, 47.2 (d, 2J=
22.2 Hz), 46.1 (d, 2J= 24.1 Hz), 41.0, 40.2, 39.7, 36.8, 29.7, 29.4, 26.8. MS
(ESI), m/z: calculated 816.11,
found 817.1 (M+1) .
Example 109. P7C3-S57.
Br Br
e 10
N
0 lei
NHN
Example 110. P7C3-S70: 2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)-N,N-
dimethylacetamide
Br Br
= =0 0 0j-N
N
\
yN
F H
188
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The title compound was prepared analogously to P7C3-S66. 1H NMR (CDC13, 400
MHz) 6 = 8.04 (d,
2H, J= 8.6 Hz), 7.45 (dd, 2H, J=1.9, 8.6 Hz), 7.20 (d, 2H, J= 9.7 Hz), 6.75
(d, 2H, J= 8.8 Hz), 6.47 (d, 2H, J
= 9.1 Hz), 4.97 (dm, 1H, 1.TH_F= 47.2 Hz), 4.53 (s, 2H), 4.38-4.60 (m, 2H),
3.11-3.36 (m, 2H), 3.00 (s, 3H), 2.89
(s, 3H). 3C NMR (CDC13, 100 MHz) 6 = 184.0, 168.3, 151.4, 142.0, 139.6, 129.5,
123.4, 116.1, 112.9, 110.7(
d, 4J= 1.8 Hz), 90.8 ( d, 1J= 175.5 Hz), 68.4, 46.4 ( d, 2J= 24.7 Hz), 45.0 (
d, 2J= 92.3 Hz), 29.8, 32.9. MS
(ESI), m/z: calculated 575.02, found 622.0 (M+HC00)-.
Example 111. P7C3-S71: 2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropylamino)phenoxy)-N-(2-
hydroxyethypacetamide
Br Br
= '00 0 0j. 0 H
N N H
H- N
F H
The title compound was prepared analogously to P7C3-566 and was purified by
chromatography on silica gel
(5% Me0H/DCM +0.2%TEA).1H NMR (CDC13, 400 MHz) 6 = 12.07 (bs, 1H), 8.15 (d,
2H), 7.55 (dd, 2H, J=
2.0, 8.5 Hz), 7.31 (d, 2H, J= 8.8 Hz), 7.06 (bm, 1H), 6.80 (d, 2H, J= 9.1 Hz),
6.57 (d, 2H, 9.2 Hz), 5.09 (dm,
1H, 1JH_F= 47.2 Hz), 4.51-4.68 (m, 2H), 4.51-4.68 (m, 2H), 4.45 (s, 2H), 3.78
(t, 3H, J= 4.9 Hz), 3.53 (q, 2H, J
= 5.4 Hz), 3.22-3.45 (m, 2H), 2.57 (bs, 1H). 13C NMR (CDC13, 100 MHz).
6 = 169.9, 150.5, 142.5, 139.7, 129.6, 123.5, 116.2, 110.7 ( d, 4J= 1.2 Hz),
90.8 ( d, 1J= 176.5
Hz), 68.3, 62.4, 46.3 ( d, 2J= 21.8 Hz), 45.0 ( d, 2J= 25.7 Hz), 42.2. MS
(ESI), m/z: calculated 591.02, found
638.0 (M+HC00)-.
Example 112. P7C3-S72: 1-(bis(4-bromophenyl)amino)-3-(phenylamino)propan-2-ol
Br Br
= .
NyN 0
H
0 H
P7C3-572 was synthesized from di-(4-bromophenyl)amine, epibromohydrin and
aniline following
Representative Procedures 1 and 2. 1H NMR (CDC13, 400 MHz) 6 = 7.38 (d, 4H, J=
8.8 Hz), 7.19 (d, 2H, J=
7.4 Hz), 6.95 (d, 4H, J= 8.8 Hz), 6.76 (t, 1H, J= 7.4 Hz), 6.62 (d, 2H, J= 7.9
Hz), 4.17 (bm, 1H), 3.89 (dd,
1H, J= 4.3, 15.2 Hz), 3.72-3.81 (m, 1H), 3.32 (dd, 1H, J= 3.2, 12.8 Hz), 3.08-
3.18 (m, 1H). 13C NMR
189
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(CDC13, 100 MHz) 6 = 148.0, 147.0, 132.6, 129.5, 123.1, 118.4, 114.9, 113.5,
67.9, 56.6, 47.8. MS (ESI), m/z:
calculated 473.99, found 521 (M+HC00)-.
Example 113. P7C3-S73: (E)-3,6-dibromo-9-(3-phenoxyally1)-9H-carbazole and (E)-
3,6-dibromo-9-(3-
phenoxyprop-1-en-1-y1)-9H-carbazole.
Step 1. 3,6-dibromo-9-(2-bromo-3-phenoxypropy1)-9H-carbazole
Br
* Br
Br * NO,
To an ice-cold solution of P7C3-539 (95.0 mg, 0.20 mmol, 1 equiv) and
triphenylphosphine (78.7 mg,
0.30 mmol, 1.5 equiv) in dichloromethane (0.6 mL) was added tetrabromomethane
(73.0 mg, 0.22 mmol, 1.1
equiv). The mixture was stirred at rt for 3 hours. Dichloromethane was and the
crude residue was purified by
silica gel chromatography using 9% Et0Ac/Hex to afford 7.4 mg white solid as
product, yield 6.9%.1H NMR
(CDC13, 400 MHz) 6 = 4.22 - 4.11 (m, 2H) 4.61 (dt, J= 12.2, 6.2 Hz, 1H) 4.68
(dd, J= 15.2, 6.4 Hz, 1H) 4.98
(dd, J= 15.2, 7.1 Hz, 1H) 6.88 (d, J= 7.8 Hz, 2H) 7.02 (t, J= 7.4 Hz, 1H) 7.37-
7.26 (m, 4H) 7.49 (dd, J=
8.7, 1.8 Hz, 2H) 8.12 (d, J= 1.8 Hz, 2H)
Step 2. P7C3-573. (E)-3,6-dibromo-9-(3-phenoxyally1)-9H-carbazole and (E)-3,6-
dibromo-9-(3-
phenoxyprop-1-en-1-y1)-9H-carbazole.
Br Br
* and *
Br
* N 0 Br
0 * 1\10 0
To a 4-mL vial were added the product of Step 1, kryptofix 222 (4.8 mg, 0.0130
mmol, 1 equiv), KF
(0.5 mg, 0.0090 mmol, 0.7 equiv), K2CO3 (0.3 mg, 0.0019 mmol, 0.15 equiv) and
acetonitrile (0.15 mL). The
vial was tightly sealed and heated to 80 C for 20 min. The crude was purified
by silica gel chromatography
using 9% Et0Ac/Hex to afford 4.9 mg white solid in one fraction as a mixture
of these two olefins in a 45:55
ratio, total yield 83.6%. 1H NMR (CDC13, 400 MHz) 6 = 4.51 (dd, J = 6.5, 1.4
Hz, 0.45 x 1H) 4.83 (dd, J =
6.2, 1.2 Hz, 0.55 x 1H) 6.21 (dt, J= 8.0, 6.6 Hz, 0.45 x 1H) 6.31 (dt, J=
14.2, 6.1 Hz, 0.55 x 1H) 6.74 (d, J=
7.9 Hz, 1H) 6.94 - 6.85 (m, 1H) 7.05 - 6.98 (m, 2H) 7.38 - 7.15 (m, 4H) 7.49
(d, J= 8.7 Hz, 1H) 7.57 (ddd, J
= 8.6, 4.1, 1.9 Hz, 2H) 8.14 (dd, J= 13.0, 1.8 Hz, 2H).
190
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 114. P7C3-S75: 1-(3,6-bis(trilluoromethyl)-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Step 1. 4-(trifluoromethyl)phenyl trifluoromethanesulfonate
F 3C * OSO2CF3
To a solution of 4-trifluoromethylphenol (324.2 mg, 2.0 mmol, 1 equiv) in
dichloromethane (1.2 mL)
was added pyridine (194.1 [LL, 2.4 mmol, 1.2 equiv). A solution of triflic
anhydride (370.1 [LL, 2.2 mmol, 1.1
equiv) in dichloromethane (1.2 mL) was added dropwise at 0 C. The mixture was
stirred at 0 C for 1 hour,
and then rt for 2.5 hours. The reaction was quenched with lmL of water. The
organic phase was washed with
saturated NaHCO3, 1M HC1 and brine, then dried with MgSO4 and concentrated to
give crude product. It was
further purified by silica gel chromatography using 5% Et0Ac/Hex to afford
449.4 mg colorless oil as product,
yield 76.4%.
1H NMR (CDC13, 400 MHz) 6 = 7.42 (d, J= 8.8 Hz, 2H) 7.75 (d, J= 9.0 Hz, 1H).
Step 2. 3,6-bis(trifluoromethyl)-9H-carbazole
F3C CF3
* 110
N
H
Following methods in Watanabe et al., J. Org. Chem. 2009, 74, 4720-4726, to a
vial under argon
atmosphere containing the product of Step 1, (29.4 mg, 0.10 mmol, 1 equiv), 4-
(trifluoromethyl)aniline (17.7
mg, 0.11 mmol, 1.1 equiv), Pd(OAc)2 (2.2 mg, 0.01 mmol, 0.1 equiv), XPhos (7.2
mg, 0.015 mmol, 0.15
equiv) and Cs2CO3 (39.1 mg, 0.12 mmol, 1.2 equiv) was added toluene (0.2 mL).
The mixture was stirred at
100 C for 1.5 hour. After cooling, the crude mixture was diluted with ethyl
acetate and washed with brine. The
organic layer was dried with Mg504 and concentrated. The crude product was
further purified by silica gel
chromatography using 0-5% of Et0Ac/Hex to afford 22.2 mg of the diaryl amine
as a colorless oil as, yield
69.2%. To this intermediate was added acetic acid (0.8 mL) and Pd(OAc)2(2.5
mg). The mixture was heated to
90 C for 12 h under an oxygen balloon. Solid NaHCO3 was added to quench the
reaction. The mixture was
diluted with ethyl acetate and washed with NaHCO3. The organic layer was dried
with Mg504 and
concentrated to give crude product. It was further purified by silica gel
chromatography using 25%
Et0Ac/Hex to afford 9.2 mg white solid yield 41.7%. 1H NMR (CDC13, 400 MHz) 6
= 7.54 (d, J = 8.6 Hz, 2H)
7.72 (dd, J= 8.6, 1.5 Hz, 2H) 8.38 (s, 2H) 8.47 (s, br, 1H). ESI (m/z): 302.0
(M - H ).
Step 3. 1-chloro-3-(phenylamino)propan-2-ol
H OH
s N 7,C1
191
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Acetic acid (0.56 mL), aniline (456 [tt, 5.0 mmol, 1 equiv) and epichlorohydin
(469 [LL, 6.0 mmol,
1.2 equiv) were combined and stirred at 75 C for 3 h in a sealed vial. The
reaction was quenched with solid
NaHCO3 (0.8218 g) and the mixture was diluted with ethyl acetate and washed
with saturated NaHCO3. The
combined organic extracts were dried with MgSO4 and concentrated to give crude
product. It was further
purified by silica gel chromatography using 30% Et0Ac/Hex to afford 495.5 mg
colorless oil as product, yield
53.4%. 1H NMR (CDC13, 400 MHz) 6 = 2.10 (d, J= 0.9 Hz, 1H) 3.25 (dd, J= 13.3,
7.1 Hz, 1H) 3.39 (dd, J=
13.3, 4.5 Hz, 1H) 3.77 - 3.56 (m, 2H) 4.17 - 4.03 (m, 1H) 6.67 (dd, J= 8.6,
1.0 Hz, 2H) 6.76 (tt, J= 7.4, 1.0
Hz, 1H) 7.20 (dd, J= 8.5, 7.4 Hz, 2H). ESI (m/z): 186.1 (M + RP); 230.1 (M +
HC00-).
Step 4. N-(oxiran-2-ylmethyl)aniline
H 0
0 N,I
To a solution of the product of Step 3 (185.7 mg, 1.0 mmol, 1 equiv) in 1,4-
dioxane (3.3 mL) was
added KOH powder (67.3 mg, 1.2 mmol, 1.2 equiv). The mixture was stirred at
room temperature for 24
hours. The mixture was diluted with Et0Ac and washed with 1M HC1 and brine.
The organic layer was dried
with Mg504 and concentrated to give crude product. It was further purified by
silica gel chromatography using
20% Et0Ac/Hex to afford 141.8 mg colorless oil as product, yield 95.0%. 1H NMR
(CDC13, 400 MHz) 6 =
2.70 (dd, J= 4.9, 2.3 Hz, 1H) 2.87 -2.77 (m, 1H) 3.23 - 3.18 (m, 1H) 3.26 (t,
J= 4.9 Hz, 1H) 3.59 - 3.48 (m,
1H) 3.87 (s, 1H) 6.64 (d, J= 7.7 Hz, 2H) 6.73 (t, J= 7.3 Hz, 1H) 7.18 (dd, J=
8.3, 7.5 Hz, 2H).
Step 5. P7C3-S75: 1-(3,6-bis(trifluoromethyl)-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
F 3C
* OH H
* N, N 0
F3C
To a solution of the product of Step 2 (4.6 mg, 0.0152 mmol, 1 equiv) in THF
(0.25mL) was added
NaH (60% dispersion in mineral oil, 0.7 mg, 0.0167 mmol, 1.1 equiv) and the
mixture was stirred at room
temperature for 15 min. The product of Step 4 (2.7 mg, 0.0182 mmol, 1.2 equiv)
was added and the resulting
mixture was stirred at room temperature overnight and then heated at 65 C for
4 hours. Brine was added and
the crude reaction was extracted 3 times with Et0Ac. The combined organic
extracts were dried with Mg504
and concentrated to give crude product. It was further purified by silica gel
chromatography using 30%
Et0Ac/Hex to afford 4.1 mg white solid as product, yield 59.6%. 1H NMR (CDC13,
400 MHz) 6 = 2.33 (s, 1H)
3.25 (dd, J= 13.1, 7.1 Hz, 1H) 3.40 (dd, J= 13.1, 4.0 Hz, 1H) 4.43 (ddd, J=
11.3, 6.8, 4.6 Hz, 1H) 4.62 - 4.46
192
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(m, 2H) 6.64 (d, J= 8.3 Hz, 2H) 6.79 (t, J= 7.3 Hz, 1H) 7.23 ¨ 7.12 (m, 2H)
7.60 (d, J= 8.6 Hz, 2H) 7.75 (dd,
J= 8.6, 1.4 Hz, 2H) 8.41 (s, 2H). 13C NMR (CDC13, 400 MHz) 6 = 147.8, 143.1,
129.7, 123.9 (dd, J= 7.0, 3.5
Hz, 1C), 123.0, 122.7, 122.5, 119.0, 118.5 (q, J= 4.2 Hz, 1C), 113.8, 110.0,
69.7, 48.1, 47.5. ESI (m/z): 497.1
(M + HC00-).
Example 115. P7C3-S77: 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylthio)propan-2-01
Br
4.
N
41)1:0)---A
s .0Me
Br
Prepared analogously to Example 3a. Chromatography (0-50% Et0Ac in hexanes)
provided 242 mg
(88% yield) of an off-white foam. 1H NMR (CDC13, 500 MHz) 6 = 8.01 (d, J=1.5
Hz, 2H), 7.46 (dd, J=1.5,
8.5 Hz, 2H), 7.21 (d, J =9.0 Hz, 2H), 7.14 (dd, J=8.0, 8.0 Hz, 1H), 6.85 (d,
J=7.5 Hz, 1H), 6.80 (m, 1H),
6.72 (dd, J=2.0, 8.0 Hz, 1H), 4.32 (dd, J=4.0, 15.0 Hz, 1H), 4.20 (dd, J =7.0,
15.0 Hz, 1H), 4.09 (m, 1H),
3.69 (s, 3H), 3.03 (dd, J=5.0, 14.0 Hz, 1H), 2.91 (dd, J=7.5, 14.0 Hz, 1H),
2.55 (d, J =3.0 Hz, 1H). 13C NMR
(CDC13, 125 MHz) 6 = 160.1, 139.7, 135.7, 130.3, 129.3 (2C), 123.6, 123.3
(2C), 122.0, 115.4, 112.7, 112.6,
111.0 (2C), 69.2, 55.4, 48.0, 39Ø ESI m/z: 563.6 ([M+HCOO]).
Example 116. P7C3-S78: 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(4-
methoxyphenylthio)propan-2-01
Br
N
el
Br HO s =
OMe
Prepared analogously to Example 3a. Chromatography (0-50% Et0Ac in hexanes)
provided 263 mg
(96% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 = 8.02 (d, J=2.0
Hz, 2H), 7.47 (dd, J =2.0,
8.5 Hz, 2H), 7.28 (d, J =8.5 Hz, 2H), 7.22 (d, J=9.0 Hz, 2H), 6.77 (d, J =9.0
Hz, 2H), 4.31 (dd, J =4.0, 15.0
Hz, 1H), 4.18 (dd, J =7.0, 15.5 Hz, 1H), 4.01 (m, 1H), 3.75 (s, 3H), 2.93 (dd,
J=5.0, 14.0 Hz, 1H), 2.79 (dd, J
=7.5, 13.5 Hz, 1H), 2.6 (d, J=3.5 Hz, 1H). 13C NMR (CDC13, 125 MHz) 6 = 159.7,
139.8 (2C), 133.9 (2C),
129.3 (2C), 124.4, 123.6 (2C), 123.3 (2C), 115.1 (2C), 112.6 (2C), 111.0 (2C),
69.1, 555.5, 48.0, 41.3. ESI
m/z: 563.5 ([M+HCOO]).
193
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 117. P7C3-S79: 3,6-Dibromo-9-(2-fluoro-3-(3-methoxyphenylthio)propy1)-
9H-carbazole
Br
N
0Me
lel ---)-----\F s .
Br
Prepared analogously to Example 96 from P7C3-S77. Chromatography (0-5% Et0Ac
in hexanes)
provided 32 mg (32% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 =
8.07 (d, J =1.5 Hz, 2H),
7.50 (dd, J =1.5, 8.5 Hz, 2H), 7.26 (d, J =8.5 Hz, 2H), 7.21 (t, J =8.0 Hz,
1H), 6.96 (d, J =7.5 Hz, 1H), 6.92
(br s, 1H), 6.77 (dd, J =2.0, 8.5 Hz, 1H), 4.90 (dm, J =47.5 Hz, 1H), 4.59
(ddd, J =2.5, 16.0, 26.5 Hz, 1H),
4.45 (ddd, J =7.0, 16.0, 22.0 Hz, 1H), 3.76 (s, 3H), 3.26 (ddd, J =4.5, 15.0,
15.0 Hz, 1H), 3.06 (m, 1H). 13C
NMR (CDC13, 125 MHz) 6 = 160.2, 139.8 (2C), 135.5, 130.5, 129.5 (2C), 123.9
(2C), 123.4 (2C), 122.2 (2C),
115.8, 113.0, 112.9, 110.9 (d, J =2.1 Hz, 2C), 104.9, 91.3 (d, J =180 Hz),
55.5, 46.1 (d, J =22.9 Hz), 35.4 (d,
J =23 .9 Hz). ESI m/z: 565.7 ([M+HCOO]).
Example 118. P7C3-S80: 3,6-Dibromo-9-(2-fluoro-3-(4-methoxyphenylthio)propy1)-
9H-carbazole
Br
it
0
Br S .
OMe
Prepared analogously to Example 96 from P7C3-S78. Chromatography (0-5% Et0Ac
in hexanes)
provided 23 mg (23% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 =
8.08 (d, J =1.5 Hz, 2H),
7.52 (dd, J =1.5, 8.5 Hz, 2H), 7.39 (d, J =9.0 Hz, 2H), 7.28 (d, J =8.5 Hz,
2H), 6.84 (d, J =9.0 Hz, 2H), 4.83
(dm, J =48.0 Hz, 1H), 4.58 (ddd, J =2.5, 15.5, 27.0 Hz, 1H), 4.45 (ddd, J
=7.0, 16.0, 20.5 Hz, 1H), 3.78 (s,
3H), 3.13 (ddd, J=4.5, 14.5, 14.5 Hz, 1H), 2.96 (m, 1H). 13C NMR (CDC13, 125
MHz) 6 = 159.9, 134.2,
129.5, 124.4, 123.9, 123.4, 115.2, 112.9, 110.9 (d, J =2.1 Hz, 2C), 104.9,
91.5 (d, J =179.6 Hz), 55.6, 46.1 (d,
J =22.6 Hz), 37.6 (d, J =22.4 Hz). ESI m/z: 565.7 ([M+HCOO] 565.9).
Example 119. P7C3-S81: 3,6-Dibromo-9-(2-fluoro-3-(3-
methoxyphenylsulfonyppropy1)-9H-carbazole
194
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
411
N--\
Br W Fis¨A ¨0
0---S¨
. OMe
Prepared analogously to Example 96 from P7C3-S77. Chromatography (0-30% Et0Ac
in hexanes)
provided 17.7 mg (84% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6
= 8.11 (d, J =1.5 Hz, 2H),
7.55 (dd, J =1.5, 8.5 Hz, 2H), 7.43 (m, 2H), 7.34 (d, J =8.5 Hz, 2H), 7.33 (m,
1H), 7.16-7.14 (m, 1H), 5.34
(dm, J =49.0 Hz, 1H), 4.71 (ddd, J =2.5, 16.0, 26.5 Hz, 1H), 4.56 (ddd, J
=7.0, 16.0, 22.5 Hz, 1H), 3.81 (s,
3H), 3.48 (m, 2H). 13C NMR (CDC13, 125 MHz) 6 = 160.4, 140.0, 139.7 (2C),
130.9, 129.7 (2C), 124.0 (2C),
123.5 (2C), 121.1 (2C), 120.2, 113.2, 112.6, 110.9 (d, J =2.1 Hz, 2C), 87.1
(d, J =181.3 Hz), 58.1 (d, J =23.4
Hz), 56.0, 47.1 (d, J=22.0 Hz). ESI m/z: 531.7 ([M-H2F]).
Example 120. P7C3-S82: 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylsulfonyl)propan-2-ol
Br
N
Br
0--S¨
* OMe
Prepared analogously to Example 3d from P7C3-S77. Chromatography (0-25% Et0Ac
in hexanes)
provided 30 mg (94% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 =
8.06 (d, J =2.0 Hz, 2H),
7.49 (dd, J=2.0, 9.0 Hz, 2H), 7.36 (apparent t, J=8.0 Hz, 1H), 7.31 (m, 1H),
7.22 (d, J =9 .0 Hz, 2H), 7.20
(m, 1H), 7.10 (m, 1H), 4.61 (m, 1H), 4.33 (m, 2H), 3.78 (s, 3H), 3.32 (br s,
1H), 3.23 (dd, J =8.0, 14.0 Hz,
1H), 3.12 (dd, J =3 .0, 14.5 Hz, 1H). 13C NMR (CDC13, 125 MHz) 6 = 160.3,
139.7, 139.6 (2C), 130.8, 129.6
(2C), 123.8, 123.4 (2C), 120.8, 119.9, 113.0 (2C), 112.3 (2C), 110.9 (2C),
65.6, 59.9, 55.9, 48.2. ESI m/z:
595.6 ([M+HCOO]).
Example 121. P7C3-S83: 3,6-Dibromo-9-(2-fluoro-3-(4-
methoxyphenylsulfonyppropy1)-9H-carbazole
195
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
=N---\
2---- ,0 Asa
Br F ,,s.
0 lip OMe
Prepared analogously to Example 96 from P7C3-S78. Chromatography (0-30% Et0Ac
in hexanes)
provided 18.9 mg (89% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6
= 8.10 (d, J2.0 Hz, 2H),
7.78 (d, J =8.5 Hz, 2H), 7.54 (dd, J =1.5, 8.5 Hz, 2H), 7.32 (d, J =8.5 Hz,
2H), 6.96 (d, J =9.0 Hz, 2H), 5.32
(dm, J =47.5 Hz, 1H), 4.69 (ddd, J =2.5, 16.0, 27.0 Hz, 1H), 4.54 (ddd, J
=7.0, 16.0, 22.5 Hz, 1H), 3.85 (s,
3H), 3.49-3.42 (m, 2H). 13C NMR (CDC13, 125 MHz) 6 = 164.5, 139.7 (2C), 130.5
(2C), 130.3, 129.7 (2C),
124.0 (2C), 123.5 (2C), 114.9 (2C), 113.2 (2C), 110.9 (d, J =2.25 Hz, 2C),
87.4 (d, J =181.1 Hz), 58.5 (d, J
=23.1 Hz), 56.0, 47.2 (d, J=22.0 Hz). ESI m/z: 531.5 ([M¨H2F].
Example 122. P7C3-S84: 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(4-
methoxyphenylsulfonyl)propan-2-01
Br
it
N
Br I. H-0)Th -0
0---S-
it
OMe
Prepared analogously to example 3d from P7C3-S78. Chromatography (0-30% Et0Ac
in hexanes)
provided 27 mg (85% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 =
8.09 (d, J =2.0 Hz, 2H),
7.67 (d, J =9.0 Hz, 2H), 7.50 (dd, J =2.0, 9.0 Hz, 2H), 7.25 (d, J =8.0 Hz,
2H), 6.92 (d, J =9.0 Hz, 2H), 4.61
(m, 1H), 4.36 (d, J =6.0 Hz, 2H), 3.86 (s, 3H), 3.35 (d, J =2.5 Hz, 1H), 3.20
(dd, J =8.5, 14.0 Hz, 1H), 3.10
(dd, J =2.5, 14.0 Hz, 1H). 13C NMR (d6-acetone, 125 MHz) 6 = 164.7, 141.0
(2C), 132.8, 131.2 (2C), 129.8
(2C), 124.5 (2C), 124.0(2C), 115.2(2C), 112.74(2C), 112.68 (2C), 66.6, 61.0,
56.3, 49.7. ESI m/z: 595.6
([M+HCOOD.
Example 123. P7C3-S91: 34343 ,6-Dibromo-9H-c arbazol-9-y1)-2-hydroxyp
ropylthiMp henol
196
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br is .
N
v_....70H
\--S
.4 OH
Prepared analogously to example 3a. Silica chromatography (0-40% Et0Ac in
hexanes) followed by
HPLC purification (75% MeCN/H20 + 0.1% HCO2H, Phenomenex C18 Luna, 10x250 mm,
3 mL/min)
provided 9.9mg (21% yield) of an off-white solid. 1H NMR (d6-acetone, 400 MHz)
6 = 8.35 (br s, 2H), 7.56
(m, 4H), 7.13 (apparent t, J =8.0 Hz, 1H), 6.94 (br s, 1H), 6.88 (d, J =7.6
Hz, 1H), 6.69 (dd, J =1.6, 8.0 Hz,
1H), 4.66 (dd, J =3.2, 15.2 Hz, 1H), 4.47 (dd, J =8.4, 14.8 Hz, 1H), 4.26 (m,
1H), 3.22 (d, J =6.4 Hz). 13C
NMR (d6-acetone, 125 MHz) 6 = 158.8, 141.1 (2C), 138.2, 130.9, 129.7 (2C),
124.4 (2C), 124.0 (2C), 120.7
(2C), 116.5, 114.2, 112.8 (2C), 112.5, 70.2, 49.2, 38.5. ESI m/z: 549.7
([M+HCOO]).
Example 124. P7C3-S92: 4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
hydroxypropylthiMphenol
Br
1101
OH s
Br it N\.-c___
=
OH
Prepared analogously to example 3a. Chromatography (0-3% acetone in
dichloromethane) followed
by HPLC purification (75% MeCN/H20 + 0.1% HCO2H, Phenomenex C18 Luna, 10x250
mm, 3 mL/min)
provided 11.4 mg (25% yield) of an off-white solid. 1H NMR (d6-acetone, 500
MHz) 6 = 8.64 (br s, 1H), 8.34
(s, 2H), 7.56 (m, 4H), 7.36 (d, J =8.5 Hz, 2H), 6.82 (d, J =8.5 Hz, 2H), 4.62
(dd, J =3.5, 15.0 Hz, 1H), 4.54
(br s, 1H), 4.43 (dd, J =8.5, 15.0 Hz, 1H), 4.16 (m, 1H), 3.09 (d, J =6.5 Hz,
2H). 13C NMR (d6-acetone, 125
MHz) 6 = 158.0, 141.1 (2C), 134.3 (2C), 129.7 (2C), 125.3, 124.4 (2C), 124.0
(2C), 117.1 (2C), 112.9 (2C),
112.5 (2C), 70.3, 49.1, 41.2. ESI m/z: 503.6 ([M¨H], C2IHI6Br2NO2S requires
503.9).
Example 125. P7C3-S93: 3-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
hydroxypropylsulfonyl)phenol
197
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
. OHO ,,,
. N S ' u
Br
410 OH
Prepared analogously to example 3d from P7C3-S91. Chromatography (0-40% Et0Ac
in hexanes)
followed by HPLC purification (75% MeCN/H20 + 0.1% HCO2H, Phenomenex C18 Luna,
10x250 mm, 3
mL/min) provided 9.9 mg (46% yield) of an off-white solid. 1H NMR (d6-acetone,
500 MHz) 6 = 9.28 (br s,
1H), 8.36 (s, 2H), 7.59 (m, 4H), 7.44 (apparent t, J =8.0 Hz, 1H), 7.43 (m,
1H), 7.38 (br s, 1H), 7.16 (d, J =8.0
Hz, 1H), 4.72 (br s, 1H), 4.64 (dd, J =2.5, 14.0 Hz, 1H), 4.76 (m, 1H), 4.54
(dd, J =8.5, 14.0 Hz, 1H), 3.66
(dd, J =5.0, 14.5 Hz, 1H), 3.58 (dd, J =6.5, 14.5 Hz, 1H). 13C NMR (d6-
acetone, 125 MHz) 6 = 158.9, 142.5,
141.0 (2C), 131.4, 129.8 (2C), 124.5 (2C), 124.1 (2C), 121.7, 119.8, 115.3,
112.8 (2C), 112.7 (2C), 66.5, 60.7,
49.7. ESI m/z: 535.5 ([M¨H], C21H16Br2N04S requires 535.9).
Example 126. P7C3-S94: 4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
hydroxypropylsulfonyl)phenol
Br
AP OHO
Br N 131.g
= 101
OH
Prepared analogously to example 3d from P7C3-S92. Chromatography (0-40% Et0Ac
in hexanes)
provided 5.5 mg (23% yield) of an off-white solid. 1H NMR (d6-acetone, 500
MHz) 6 = 8.36 (s, 2H), 7.79 (d,
J =9 .0 Hz, 2H), 7.60 (m, 4H), 7.01 (d, J =9 .0 Hz, 2H), 4.66-4.50 (m, 3H),
3.61 (dd, J=5.0, 14.5 Hz, 1H),
3.52 (dd, J=6.0, 14.5 Hz, 1H). 13C NMR (d6-acetone, 125 MHz) 6 = 163.2, 141.0
(2C), 131.7, 131.4 (2C),
129.8 (2C), 124.5 (2C), 124.0 (2C), 116.7(2C), 112.8 (2C), 112.7 (2C), 66.6,
61.1, 49.7. ESI m/z: 535.5 ([M¨
H], C2IHI6Br2NO4S requires 535.9).
Example 127. P7C3-S95: 1-(3-Aminophenylthio)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br
. OH
. N S
Br
= N H2
198
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Prepared analogously to example 3a. Chromatography (0-50% Et0Ac in hexanes)
provided 5.5 mg
(23% yield) of an off-white solid. 1H NMR (CDC13, 400 MHz) 6 = 8.08 (s, 2H),
7.50 (d, J =8.8 Hz, 2H), 7.26
(d, J =8.8 Hz, 2H), 7.01 (apparent t, J =8.0 Hz, 1H), 6.66 (d, J =8.0 Hz, 1H),
6.49 (m, 2H), 4.39 (dd, J =4.8,
15.2 Hz, 1H), 4.27 (dd, J =6.8, 15.6 Hz, 1H), 4.13 (m, 1H), 3.58 (br s, 2H),
3.01 (dd, J =5.2, 14.0 Hz, 1H),
2.88 (dd, J =7.6, 14.0 Hz, 1H), 2.53 (br s, 1H). 13C NMR (CDC13, 125 MHz) 6 =
147.3, 139.8 (2C), 135.2,
130.3 (2C), 129.4 (2C), 123.7, 123.4 (2C), 120.0 (2C), 116.1, 114.0, 112.7,
111.1 (2C), 69.2, 48.1, 39Ø ESI
m/z: 504.6 ([M+H]+, C21H19Br2N2OS requires 505.0).
Example 128. P7C3-S96: 1-(4-Aminophenylthio)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br
= OH
41, N S
Br
.
NH2
Prepared analogously to example 3a. Chromatography (0-50% Et0Ac in hexanes)
provided 31 mg
(23% yield) of an off-white solid. 1H NMR (CDC13, 400 MHz) 6 = 8.09 (s, 2H),
7.50 (d, J =8.8, 2H), 7.28 (d,
J =8.4 Hz, 2H), 7.18 (d, J =8.4 Hz, 2H), 6.55 (d, J =8.4 Hz, 2H), 4.36 (dd, J
=4.0, 15.6 Hz, 1H), 4.23 (dd, J
=6.8, 15.2 Hz, 1H), 4.03 (m, 1H), 3.73 (br s, 2H), 2.91 (dd, J =5.2, 14.0 Hz,
1H), 2.75 (dd, J =8.0, 13.6 Hz,
1H), 2.59 (br s, 1H). 13C NMR (CDC13, 125 MHz) 6 = 146.9, 139.9 (2C), 134.6
(2C), 129.3 (2C), 123.7, 123.3
(2C), 121.0 (2C), 115.9 (2C), 112.6 (2C), 111.2 (2C), 69.1, 48.1, 41.9. ESI
m/z: 504.7 ([M+H]+,
C21H19Br2N2OS requires 505.0).
Example 129. P7C3-S97: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-
amine
Step 1. 1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-one
Br
it
0
Br * N.)0 0
To a solution of P7C3-S39 (87.2 mg, 0.1835 mmol, 1 equiv) in CHC13 (3 mL) was
added Dess-Martin
periodinane (DMP, 77.8 mg, 0.1835 mmol, 1 equiv). The mixture was stirred at
room temperature. After 1
hour, a second batch of DMP (31.1 mg, 0.0734 mmol, 0.4 mmol) was added to the
reaction mixture and
further stirred for another 4 hours. Solvent was removed on the vacuum and the
crude residue was purified by
silica gel chromatography using 28% Et0Ac to afford 31.7 mg white solid as
product, yield 36.9%. 1H NMR
199
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
(CDC13, 400 MHz) 6 = 4.69 (s, 2H) 5.30 (s, 2H) 6.92 (d, J= 8.7 Hz, 2H) 7.04
(d, J= 8.6 Hz, 2H) 7.08 (t, J=
8.7 Hz, 1H) 7.36 (t, J= 8.0 Hz, 2H) 7.53 (d, J= 8.7 Hz, 2H) 8.16 (s, 2H)
Step 2. (Z)-1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-one 0-benzyl
oxime
Br
101
41 N.0
BrN ,>1 0
* 0
To a solution of the product of Step 1 (17.7 mg, 0.0374 mmol, 1.0 equiv) in
THF (400 [tL) were added
2,6-lutidine (4.4 [tt, 0.0374 mmol, 1.0 equiv), 0-benzylhydroxylamine
hydrochloride (14.3 mg, 0.0898 mmol,
2.4 equiv) and 4A molecular sieves (15.8 mg). The mixture was stirred for 12 h
until TLC indicated complete
consumption of starting material. The reaction mixture was quenched with
saturated NaHCO3 and extracted 3
times with dichloromethane. The combined organic extracts were dried with
Mg504 and concentrated to give
crude product. It was further purified by silica gel chromatography (5-10%
Et0Ac/Hex) to afford 20.2 mg
white solid as product, yield 93.4%. 1H NMR (CDC13, 400 MHz) 6 = 4.68 (s, 2H)
5.00 (s, 2H) 5.14 (s, 2H)
6.72 (d, J= 8.2 Hz, 2H) 6.94 (t, J= 7.3 Hz, 1H) 7.47 - 7.16 (m, 11H) 8.06 (s,
2H)
Step 3. P7C3-S97: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-phenoxypropan-2-amine
Br
41 N H2
Br . N 0 0
To a stirred solution containing the product of Step 2 (5.8 mg, 0.01 mmol, 1
equiv) in anhydrous THF
(0.2 mL) at 0 C was added borane-THF complex (1M in THF, 150 [tL, 0.15 mmol,
15.0 equiv). The mixture
was stirred at rt overnight. The reaction mixture was quenched with methanol
and concentrated under vacuum.
10% Pd-C (4.0 mg) and anhydrous methanol were added and the mixture was
stirred at rt for 5 hours under a
hydrogen balloon. The mixture was filtered through a plug of silica-gel and
NaHCO3 was further purified by
silica gel chromatography (1-5% Me0H/0.2% Et3N/dichloromethane) to afford 4.1
mg white solid as product,
yield 58.1%.1H NMR (CD30D, 500 MHz) 6 = 3.61 (td, J= 9.7, 4.0 Hz, 1H) 3.72
(dd, J= 9.6, 4.0 Hz, 1H) 3.89
(dd, J= 9.5, 4.2 Hz, 1H) 4.39 (dd, J= 14.9, 5.9 Hz, 1H) 4.59 (dd, J= 14.9, 8.2
Hz, 1H) 6.88 (d, J= 8.0 Hz,
2H) 6.94 (t, J= 7.4 Hz, 1H) 7.26 (t, J= 8.0 Hz, 2H) 7.46 (dd, J= 8.8, 1.7 Hz,
2H) 7.49 (d, J= 8.7 Hz, 2H)
8.21 (s, 2H). 13C NMR (CD30D, 500 MHz) 6 = 159.8, 141.0, 130.5, 130.2, 124.9,
124.2, 122.2, 115.5, 113.3,
112.2, 69.8, 51.2, 46.9 ESI (m/z): 472.7 (M + H ).
200
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 130. P7C3-S98: N-Benzy1-2-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylthio)-
phenoxy)acetamide
Br
Br * .
S 414
.
NH
0/
0
Prepared analogously to P7C3-S66 from P7C3-S91. Chromatography (0-50% Et0Ac in
hexanes)
provided 6.6 mg (23% yield) of an off-white solid. 1H NMR (CDC13, 500 MHz) 6 =
8.05 (d, J =1.5 Hz, 2H),
7.47 (dd, J =1.5, 8.5 Hz, 2H), 7.30-7.23 (m, 5H), 7.18-7.15 (m, 2H), 6.92 (d,
J =7.5 Hz, 1H), 6.81 (br s, 1H),
6.72-6.69 (m, 2H), 4.43 (s, 2H), 4.41-4.35 (m, 3H), 4.28 (dd, J =7.0, 15.0 Hz,
1H), 4.12 (m, 1H), 3.04 (dd, J
=6.0, 14.0 Hz, 1H), 2.97 (dd, J =7.0, 14.0 Hz, 1H), 2.75 (br s, 1H). 13C NMR
(CDC13, 125 MHz) 6 = 169.3,
168.1, 157.7, 139.8, 137.7, 136.7, 130.6, 129.4, 129.0, 127.92, 127.90, 123.8,
123.4, 123.2, 115.5, 113.2,
112.7, 111.1, 69.3, 67.5, 48.1, 43.2, 38.7. ESI m/z: 696.6 ([M+HCOO].
Example 131. P7C3-S99: N-Benzy1-2-(4-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylthio)-
phenoxy)acetamide
Br
Br 40 *
N
\--S
IP
II 0
Prepared analogously to P7C3-S66 from P7C3-S92. Chromatography (0-70% Et0Ac in
hexanes,
followed by 0-10% EtOAC in dichloromethane) provided 8.7 mg (22% yield) of an
off-white solid. 1H NMR
(CDC13, 500 MHz) 6 = 8.10 (s, 2H), 7.50 (dd, J =1.5, 8.5 Hz, 2H), 7.32-7.26
(m, 8H), 6.79 (m, 3H), 4.51 (d, J
=6.0 Hz, 2H), 4.48 (s, 2H), 4.40 (dd, J =4.5, 15.0 Hz, 1H), 4.29 (dd, J =7.0,
15.5 Hz, 1H), 4.07 (m, 1H), 2.99
(dd, J =5.0, 14.0 Hz, 1H), 2.85 (dd, J =7.5, 13.5 Hz, 1H), 2.54 (br s, 1H).
13C NMR (CDC13, 125 MHz) 6 =
167.8, 157.0, 139.9, 133.7, 129.4, 129.0, 128.0, 127.9, 123.9, 123.8, 123.5,
115.8, 112.7, 111.1, 69.2, 67.6,
48.1, 43.2, 41.1. ESI m/z: 696.5 ([M+HCOO], C31H27Br2N205S requires 697.0).
Example 132. P7C3-S100
201
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Br Br
=
NY N 0 N
F¨ la
\
A solution of amine-terminated P7C3 analog (prepared via alkylation of the
phenol analogously to
P7C3-S66) (5.0 mg, 0.0087 mmol) in 300 I DMF was added to 4,4-difluoro-5,7-
dimethy1-4-bora-3a,4a-diaza-
s-indacene-3-propionic acid succinimidyl ester (Bodipy-OSu, 4.0 mg, 0.010
mmol), followed by the addition
of diisopropylethyl amine (25 1, 0.14 mmol). The reaction was stirred
overnight in the absence of light. The
reaction was diluted with Et0Ac and washed several times with water and then
brine. The organic layer was
dried over Na2SO4, filtered and condensed. The crude mixture was purified by
preparative TLC in the absence
of light in 100% Et0Ac to give the desired product. Yield = 54 %. MS (ESI),
m/z: calculated 848.18, found
848.7 (M+1) .
Example 133. P7C3-S101: 3-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
fluoropropylsulfonyl)phenol
Br
Br 40
µS/
= OH
Prepared analogously to example 96 from P7C3-591. Chromatography (0-50% Et0Ac
in hexanes)
followed by HPLC purification (30% Et0Ac/hexanes, Phenomenex Silica Luna,
10x250 mm, 3 mL/min)
provided 13.9 mg (14% yield) of a pale yellow solid. 1H NMR (d6-acetone, 500
MHz) 6 = 9.41 (br s, 1H),
8.38 (s, 2H), 7.60 (m, 4H), 7.45 (apparent t, J =8.0 Hz, 1H), 7.39 (d, J =8.0
Hz, 1H), 7.35 (br s, 1H), 7.16 (dd,
J =2.0, 8.0 Hz, 1H), 5.42 (dm, J =47.0 Hz, 1H), 4.89-4.78 (m, 2H), 3.92 (d, J
=5.5 Hz, 1H), 3.87 (m, 1H). 13C
NMR (d6-acetone, 125 MHz) 6 = 159.0, 142.2, 140.8, 131.5, 130.1, 124.7, 124.3,
122.0, 119.8, 115.4, 113.2,
112.5 (d, J =1.75 Hz), 88.6 (d, J =178.8 Hz), 58.5 (d, J =21.8 Hz), 47.1 (d, J
=21.1 Hz). ESI m/z: 537.7 ([M¨
H].
Example 134. P7C3-S102: N-Benzy1-2-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylsulfony1)-
phenoxy)acetamide
202
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 *
NH
= f::
0
Prepared analogously to P7C3-S66 from P7C3-S93. Chromatography (0-50% acetone
in hexanes)
provided 10.1 mg (20% yield) of an off-white solid. 1H NMR (d6-acetone, 500
MHz, 45 C) 6 = 8.32 (s, 2H),
8.00 (br s, 1H), 7.57 (s, 3H), 7.55-7.52 (m, 2H), 7.32-7.30 (m, 1H), 7.29 (m,
2H), 7.22 (m, 1H), 4.65 (s, 2H),
4.63-4.60 (m, 2H), 4.53 (m, 1H), 4.47 (d, J =6.0 Hz, 1H), 3.61 (m, 2H), 3.32
(d, J =5.5 Hz, 1H). 13C NMR
(d6-acetone, 125 MHz) 6 = 168.1, 159.0, 142.7, 141.0, 140.2, 131.5, 129.9,
129.2, 128.4, 127.8, 124.5, 124.1,
121.7, 121.0 115.2, 112.8, 112.7, 68.3, 66.5, 60.7, 49.6, 43.1. ESI m/z: 728.5
([M+HCOO].
Example 135. P7C3-S103: 4-(3-(3,6-Dibromo-9H-carbazol-9-y1)-2-
fluoropropylsulfonyl)phenol
Br
Br I. 4.
µS/
it
OH
Prepared analogously to example 96 from P7C3-S94. HPLC purification (40%
Et0Ac/hexanes,
Phenomenex Silica Luna, 21.2x250 mm, 13.5 mL/min) provided 11.4 mg (16% yield)
of an off-white solid.
1H NMR (d6-acetone, 500 MHz) 6 = 8.39 (s, 2H), 7.76 (d, J =8.5 Hz, 2H), 7.60
(m, 4H), 7.00 (d, J =8.5 Hz,
2H), 5.39 (dm, J =51.5 Hz, 1H), 4.89-4.81 (m, 2H), 3.85 (m, 1H), 3.80 (d, J
=5.5 Hz). 13C NMR (d6-acetone,
125 MHz) 6 = 163.5, 140.8 (2C), 131.5(2C), 131.3, 130.1 (2C), 124.7 (2C),
124.3 (2C), 116.8 (2C), 113.2
(2C), 112.5 (d, J=1.9 Hz, 2C), 88.8 (d, J=178.5 Hz), 58.8 (d, J=21.6 Hz), 47.2
(d, J=21.3 Hz). ESI m/z:
537.6 (EM¨H].
Example 136. P7C3-S104: 5-(5-(3-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropylamino)phenoxy)pentylcarbamoy1)-2-(6-hydroxy-3-oxo-3H-xanthen-9-
yl)benzoic acid
203
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Br Br
4. *
N0
H
YN ISI OWN 110 if& 0
H
OH
----VP
HOOC
* 0
HO
The title compound was synthesized analogously to P7C3-S100. MS (ESI), m/z:
calculated 931.1, found 931.6
(M) .
Example 137. P7C3-S105: 1-(8-bromo-3,4-dihydro-1H-pyrido14,3-blindo1-5(2H)-y1)-
3-phenoxypropan-
2-ol
Step 1. tert-butyl 8-bromo-3,4-dihydro-1H-pyrido[4,3-b]indole-2(5H)-
carboxylate
0
Br ,--0----
4410 \ N
N
H
A solution of 8-bromo-2,3,4,5-tetrahydro-1H-pyrido[4,3-b]indole (813 mg, 3.2
mmol),
dimethylaminopyridine (53.5 mg, 0.14 mmol) and di-tert butyl dicarbonate (1.46
g, 6.7 mmol) in methylene
chloride (10 ml) and methanol (5.0 ml) with triethlamine (0.95 ml, 6.8 mmol)
was stirred overnight. The
reaction was condensed to a dark red semi-solid before dilution with methylene
chloride. The organic layer
was washed twice with water and brine, then dried over Na2504, filtered and
condensed. The crude reaction
product was purified in 50% Et0Ac/hexanes to give 931.8 mg of product (82%).
1H NMR (CDC13, 500 MHz)
6 = 7.88 (bs, 1H), 7.58 (s, 1H), 7.22 (dd, 2H, J= 8.3, 28.1 Hz), 4.58 (s, 2H),
3.82 (s, 2H), 2.83 (s, 2H), 1.51 (s,
9H). (ESI (m/z): 350.8 (M+1) .
Step 2: tert-butyl 8-bromo-5-(2-hydroxy-3-phenoxypropy1)-3,4-dihydro-1H-
pyrido[4,3-b]indole-
2(5H)-carboxylate
204
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
0___0......
Br
44k \ N
N
YOPh
OH
A solution of tert-butyl 8-bromo-3,4-dihydro-1H-pyrido[4,3-b]indole-2(5H)-
carboxylate (449.7 mg,
1.28 mmol) and powdered potassium hydroxide (86.9 mg, 1.54 mmol) in acetone
(4.0 ml) was stirred for 15
minutes before the addition of 2-(phenoxymethyl)oxirane (254mg, 1.69 mmol).
After 1 h the reaction was
condensed, diluted with Et0Ac and washed twice with water and then brine. The
organic layer was then dried
over Na2SO4, filtered and condensed. The crude mixture was purified by silica
gel chromatography (1%
Me0H/CH2C12 +0.1% Et3N). Yield = 21%. ESI (m/z): 546.6 (M+CHC00-).
Step 3. P7C3-S105: 1-(8-bromo-3,4-dihydro-1H-pyrido[4,3-1Vindol-5(2H)-y1)-3-
phenoxypropan-2-ol
Br
NH
fk \
N
I.
0
OH
Trifluoroactetic acid (31 ul, 0.40 mmol) was added to a solution of the
product of Step 2 (20.1 mg,
0.04 mmol) in methylene chloride (0.3 m1). After 100 minutes the reaction
mixture was condensed and
purified by preparative TLC (10% Me0H/CH2C12). Yield= 96%. 1H NMR (CDC13, 400
MHz) ) 6 = 7.43 (s,
1H), 7.27 (s, 1H), 7.17 (dd, 2H, J= 8.5, 26.7 Hz), 6.97 (t, 1H, 4.58 J= 7.0
Hz), 6.86 (d, 2H, J= 6.9 Hz), 4.24
(dm, 5H), 4.06 (m, 1H), 3.88 (m, 2H), 3.34 (m, 2H), 3.16 (m, 1H), 2.96 (m,
1H). ESI (m/z): 400.8 (M+1) .
Example 138. P7C3-S106: 1-(8-bromo-2-cyclopropy1-3,4-dihydro-1H-pyrido[4,3-
b]indo1-5(2H)-y1)-3-
phenoxypropan-2-ol
Br
NA
fa \
N
YO el
OH
Following a literature procedure (Barta, Thomas E. et al. WO 2003/091247 A2),
ethoxycyclopropyl-
oxy trimethylsilane (30 IL, 0.15 mmol) was added to a solution of P7C3-5105
(45.9 mg, 0.114 mmol) in
205
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
methanol (1.0 ml) and acetic acid (70 1, 1.2 mmol). The reaction was stirred
for 10 minutes before the
addition of sodium cyanoborohydride (37.0 mg, 0.59 mmol). The sealed vial was
heated to reflux for 2.5 hours
after which it was condensed, diluted with Et0Ac, washed with 1 N NaOH
solution, water and brine. The
organic layer was then dried over Na2SO4, filtered and condensed. Purification
by preparative TLC (5%
Me0H/CH2C12) provided the product in 8% yield. 1H NMR (CDC13, 400 MHz) ) 6
7.54 (s, 1H), 7.30 (t, 1H, J
= 7.7 Hz), 7.18 (s, 2H), 7.00 (t, 1H, J= 7.3 Hz), 6.88 (d, 2H, J= 8.4 Hz),
4.29 (m, 2H), 4.15 (m, 1H), 3.92
(m, 4H), 3.00 (m, 4H), 1.98 (bs, 1H), 1.33 (m, 1H), 0.6 (m, 4H). 13C NMR
(CDC13, 126 MHz) 6158.1, 135.7,
125.2, 129.8, 127.6, 123.9, 121.7, 120.5, 114.6, 112.7, 110.7, 69.6, 38.8,
50.8, 49.6, 45.7, 45.7, 38.0, 8.7, 6.4.
ESI (m/z): calculated 440.11, found 440.9 (M+1) .
Example 139. P7C3-S107: 8-bromo-5-(2-hydroxy-3-phenoxypropy1)-3,4-dihydro-1H-
pyrido[4,3-
Mindole-2(5H)-carbonitrile
AN
Br
YO
OH
Following a literature procedure (Kong, Chan Chun et al..; W02004/52885)
cyanogen bromide (5.0 M
in CH3CN, 44 I) was added to a solution of P7C3-S105 (88.1 mg, 0.22 mmol) and
potassium carbonate (45.4
mg, 0.33 mmol) in methylene chloride (2.1 m1). The reaction was stirred at
ambient temperature then at reflux
overnight. The cooled reaction mixture was filtered through a small celite
plug directly into a separatory
funnel. The organic layer was washed with water and brine, dried over Na2SO4,
filtered and condensed.
Chromatography on silica gel (1% Me0H/CH2C12) provided the purified product.
Yield = 12% 1H NMR
(CDC13, 400 MHz) ) 6 = 7.52 (s, 1H), 7.32 (t, 1H, J= 8.2 Hz), 7.25 (m, 2H),
7.02 (t, 1H, J= 7.3 Hz), 6.90 (d,
2H, J= 7.8 Hz), 4.46 (s, 2H), 4.34 (m, 2H), 4.19 (m, 1H), 4.00 (dd, 1H, J=
4.4, 9.5 Hz), 3.87 (dd, 1H, J= 4.8,
9.7 Hz), 3.55 (m, 2H), 3.01 (m, 2H) 2.49 (bs, 1H). 13C NMR (CDC13, 126 MHz)
6160.0, 125.4, 133.9, 129.9,
124.9, 120.5, 118.2, 113.3, 111.0, 104.8, 69.5, 68.8, 46.7, 46.3, 45.9, 22.1.
ESI (m/z): calculated 425.07, found 471.8(M+CH3C00)-.
Example 140. P7C3-S108: 8-bromo-5-(2-fluoro-3-phenoxypropy1)-2,3,4,5-
tetrahydro-1H-pyrido[4,3-
Mindole
Step 1. tert-butyl 8-bromo-5-(2-fluoro-3-phenoxypropy1)-3,4-dihydro-1H-
pyrido[4,3-b]indole-2(5H)-
206
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
carboxylate
Br
NBoc
0
Following Representative Procedure 4, the title compound was synthesized from
the product of Step 2
in the synthesis of P7C3-S105. The crude reaction product used without
purification.
Step 2. P7C3-S108: 8-bromo-5-(2-fluoro-3-phenoxypropy1)-2,3,4,5-tetrahydro-1H-
pyrido[4,3-
b]indole
Br
'VA \ NH
o
Trifluoroactetic acid (15 IL, 0.20 mmol) was added to a solution of the
product of Step 1(20.6 mg,
0.04 mmol) in methylene chloride (0.4 m1). A further 25 I trifluoroactetic
acid (0.32 mmol) was added after
3 hours. The reaction was diluted with methylene chloride, washed with twice
with water and twice with 10%
NaC1 solution. The organic layer was dried over Na2504, filtered and
condensed. The crude was purified by
preparative TLC (7% Me0H/DCM +0.15% TEA) and isolated in quantitative yield.
1H NMR (CD30D, 500 MHz) ) 6 = 7.62 (m, 1H), 7.38 (d, 1H, J= 9.9 Hz), 7.25 (m,
3H), 6.92 (m, 2H),
5.06 (dm, 1H), 4.56 (m, 2H), 4.37 (s, 2H), 4.08-4.24 (m, 2H), 3.57 (m, 2H),
3.27 (m, 1H), 3.18 (m, 2H). 13C
NMR (CD30D, 126 MHz) 6 = 159.7, 137.1, 134.5, 130.7, 126.0, 121.4, 115.6,
114.3, 112.6, 103.2, 91.7 (d,
=177.1 Hz), 68.0 (d, 2J=23.5 Hz), 47.9, 45.0 (d, 2J=22.9 Hz), 42.9, 41.9,
20.8, 9.2. MS (ESI), m/z: calculated
402.07, found 402.8 (M+1) .
Example 141. P7C3-S109: 1-(cyclohexylamino)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br Br
=
NHNC)
OH
207
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Cyclohexylamine (152 IL, 1.3 mmol) was added to a heterogeneous solution of
3,6-dibromo-9-
(oxiran-2-ylmethyl)-9H-carbazole (102.5 mg, 0.27 mmol) in ethanol (2.6 m1).
The reaction mixture was heated
to reflux for 1 h and then condensed to yield pure desired product. Yield=
97%. 1H NMR (CDC13, 500 MHz) )
6 8.13 (d, 2H, J= 1.5 Hz), 7.55 (dd, 2H, J= 1.8, 8.6 Hz), 7.36 (d, 2H, J= 8.8
Hz), 4.28 (d, 2H, J= 5.5 Hz),
4.01 (m, 1H), 2.81 (dd, 1H, J= 3.5, 12.0 Hz), 2.50 (m, 1H), 2.29 (m, 1H), 1.77
(d, 2H, J= 11.4 Hz), 1.63 (m,
3H), 0.84- 1.28 (m, 6H). 13C NMR (CDC13, 500 MHz) 6140.0, 129.3, 123.7, 123.3,
112.4, 111.1, 69.2, 56.8,
50.0, 47.6, 34.1, 33.7, 26.0, 25.1 ESI (m/z): calculated 478.03, found 524.7
(M+CHC00)-.
Example 142. P7C3-S110: (9-(2-hydroxy-3-(phenylthio)propy1)-9H-carbazole-3,6-
dicarbonitrile
NC
* OH
* 0
NC NS
Prepared from P7C3-S7 5.3% yield analogously to Example 101. 1H NMR (d6-
Acetone, 400 MHz) 6 =
3.40 -3.24 (m, 2H) 4.30 (tdd, J= 9.0, 6.1, 2.9 Hz, 1H) 4.66 (dd, J= 15.1, 8.7
Hz, 1H) 4.74 (d, J= 5.1 Hz, 1H)
4.82 (dd, J= 15.1, 3.0 Hz, 1H) 7.22 (t, J= 7.4 Hz, 1H) 7.33 (t, J= 7.6 Hz, 2H)
7.47 (dd, J= 8.3, 1.0 Hz, 2H)
7.92 - 7.77 (m, 4H) 8.73 (s, 2H) 13C NMR (d6-Acetone, 500 MHz) 6 = 143.8,
136.3, 130.1, 129.4, 129.2,
126.4, 126.0, 122.4, 119.8, 111.9, 103.2, 69.4, 48.7, 37.9 ESI (m/z): 427.8 (M
+ HC00-).
Example 143. P7C3-S111: 9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3,6-
dicarbonitrile
NC
* OH
NC * N 0 0
Prepared from P7C3-S39 in 16.5% yield, analogously to Example 101. 1H NMR (d6-
Acetone, 400
MHz) 6 = 4.15 (d, J= 5.4 Hz, 2H) 4.56 (dt, J= 9.2, 5.1 Hz, 1H) 4.76 (dd, J=
15.1, 7.6 Hz, 1H) 4.86 (dd, J=
15.1, 3.9 Hz, 1H) 6.98 (dd, J= 16.4, 8.0 Hz, 3H) 7.31 (t, J= 8.0 Hz, 2H) 7.85
(dd, J= 8.6, 1.4 Hz, 2H) 7.96
(d, J= 8.6 Hz, 2H) 8.75 (s, 1H). 13C NMR (d6-Acetone, 500 MHz) 6 = 158.9,
143.9, 130.1, 129.7, 126.0,
122.5, 121.2, 119.7, 114.7, 112.0, 103.3, 69.7, 69.0, 46.9. ESI (m/z): 411.9
(M + HC00-).
Example 144a and 144b. P7C3-S113 and P7C3-S114: N-(3-(3,6-dibromo-9H-carbazol-
9-y1)-2-
fluoropropy1)-3-methoxyaniline (R- and S- enantiomers)
208
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Step 1: (2R)-N-(3-(3,6-dibromo-9H-carbazol-9-y0-2-fluoropropyl)-3,3,3-
trifluoro-2-methoxy-N-(3-
methoxyphenyl)-2-phenylpropanamide
Br
OMe
e0 CF3
N
Br 4,
a 0
To a solution of P7C3-S10 (20.0 mg, 0.0395 mmol, 1.0 equiv) in dichloromethane
(790 [tL) was
added NaH (60% dispersion in mineral oil, 0.9 mg, 0.0395 mmol, 1.0 equiv). The
mixture was stirred at room
temperature for 15 minutes. (S)-(+)-a-methoxy-a-trifluoromethyl-phenylacetyl
chloride (14.8 [LL, 0.0790
mmol, 2.0 equiv) was added dropwise into the reaction mixture. 4-
(dimethylamino)pyridine (DMAP, catalytic)
was added to the above mixture after 1 hour. The mixture was stirred at room
temperature overnight and then
quenched by water. The crude reaction was diluted with ethyl acetate and
washed with brine. The organic
layer was dried with MgSO4 and concentrated to give crude product. It was
further purified by silica gel
preparative HPLC (20-25% Et0Ac/Hex) to afford 10.1 mg white solid of the
faster eluting diastereomer and
6.8 mg white as the slower eluting diasteromer, yield 59.2%. 1H NMR (CDC13,
400 MHz) Faster eluting
diasteromer: 6 = 3.39 (s, 3H) 3.54 (s, 3H) 3.70 - 3.61 (m, 1H) 4.34 (dd, J=
30.0, 14.2 Hz, 1H) 4.61 -4.44 (m,
2H) 5.24 (d, J= 50.4 Hz, 1H) 6.66 (d, J= 8.1 Hz, 1H) 7.40 -7.23 (m, 10H) 7.54
(d, J= 8.6 Hz, 2H) 8.12 (s,
2H) Slower diastereomer: 6 = 3.25 (s, 3H) 3.50 (s, 3H) 3.61 -3.53 (m, 1H) 4.27
(dd, J = 32.4, 14.4 Hz, 1H)
4.61 -4.40 (m, 2H) 5.32 (d, J= 50.3 Hz, 1H) 6.65 (d, J= 7.9 Hz, 1H) 7.42 -
7.20 (m, 10H) 7.56 (d, J= 8.6
Hz, 2H) 8.12 (s, 2H). P7C3-S113 (see below) was derived from the diastereomer
that elutes faster on reverse
phase HPLC (C18 column) and elutes slower by normal phase (silica gel) HPLC.
Step 2. P7C3-S113 and P7C3-S114: N-(3-(3,6-dibromo-9H-carbazol-9-y0-
2-fluoropropyl)-3-
methoxyaniline (absolute stereochemistiy unassigned).
Br Br
N.,&..) OMe * NNH OMe
Br = Br
k's
s===`-?
and
To dry and nitrogen flushed vials containing the separated products of Step 1
(4.0 mg, 0.00554 mmol,
1 equiv) was added anhydrous and degassed diethyl ether (206 [LL). The
suspension was chilled to 0 C.
Lithium aluminum hydride solution (1M in THF, 60 [tL, 0.06 mmol, 3 equiv) was
added to the above chilled
suspension. The mixture was stirred in ice bath for 1 hour and further at room
temperature for another 1 hour.
Water (0.4 [tL), 15% NaOH (0.4 [tL) and water (1.2 [tL) were added
successively to the mixture to quench the
209
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
reaction. The crude was diluted with ethyl acetate and washed with brine. The
organic layer was dried with
MgSO4 and concentrated. It was further purified by silica gel chromatography
(30% EtOAC/Hex) to afford 1.5
mg white solid as product, yield 50-55%. P7C3-S113 and ¨S114 displayed
identical LC/MS chromtograms
and NMR spectra as P7C3-S10. P7C3-S113 was found to have >99% ee by HPLC
(Chiralcel OD-H, 1
mL/min, 100% Acetonitrile ti i3 = 5.45 min, tsii4 = 5.74 min). P7C3-5114 was
found to have 79% ee.
It should be appreciated by one skilled in the art, as generally known, that
different enantiomers may
have different activity. One enantiomer can be more active than another
enantiomer. Two enantiomers
combined can have another level of activity that is different than either
substantially pure enantiomer.
Preliminary experiments suggest P7C3-5113 is more active than P7C3-5114 in pro-
neurogenic and/or anti-
apoptotic activities in an in vivo assay where 12 week old adult male C57/B16
mice were treated with 10 [LM of
either compound. It should be noted that such difference in enantiomer
activity may also be observed in other
compounds of the presently disclosed embodiments. It should also be noted that
such activity may depend on
assay mode, compound concentration, compound purity, compound stability, as
well as other parameters. It is
possible that when tested at a different concentration, a less active
enantiomer may show increased activity,
and vice versa.
Example 145. P7C3-S115: N-(2-(3,6-dibromo-9H-carbazol-9-ypethypaniline
Step 1. ethyl 2-(3,6-dibromo-9H-carbazol-9-yOacetate
Br
* 0
Br
To a solution of 3,6-dibromocarbazole (325.0 mg, 1.0 mmol, 1 equiv) in
anhydrous IV,N-
dimethylformamide (5 mL) was added crushed KOH (67.3 mg, 1.2 mmol, 1.2 equiv).
The mixture was stirred
for 30 minutes. Ethyl bromoacetate (277.2 [LL, 2.5 mmol, 2.5 equiv) was added
into the mixture and it was
stirred at room temperature overnight. The reaction crude was diluted with
ethyl acetate (30 mL) and washed
with 1M HC1 and water. The organic layer was dried with Mg504 and the
concentrated to afford 396.3 mg
white solid as product (96.4%).
1H NMR (CDC13, 400 MHz) 6 = 1.22 (t, J= 7.1 Hz, 3H) 4.20 (q, J= 7.1 Hz, 2H)
4.94 (s, 2H) 7.21 (d,
J= 8.7 Hz, 2H) 7.57 (dd, J= 8.6, 1.1 Hz, 2H) 8.16 (s, 2H). ESI (m/z): 407.6 (M
- H ).
Step 2. 2-(3,6-dibromo-9H-carbazol-9-yOacetic acid
210
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
illt 0
Br * NOH
To a solution of the product of Step 1(41.1 mg, 0.1 mmol, 1 equiv) in THF-
methanol-water (3:2:1,
total 1.2 mL) was added LiOH (12.0 mg, 0.5 mmol, 5 equiv). The mixture was
stirred at room temperature for
1 hour. The reaction was diluted with 1M HC1 (10 mL) and extracted with ethyl
acetate (10 mL). The organic
layer was washed with water (10 mL) twice and dried with Mg504 to afford 38.3
mg white solid as product,
yield 99%. 1H NMR (CDC13, 400 MHz) 6 = 5.02 (s, 2H) 7.22 (d, J= 8.8 Hz, 2H)
7.58 (dd, J= 8.7, 1.2 Hz, 2H)
8.16 (d, J= 1.6 Hz, 2H). ESI (m/z): 379.6 (M - 1-111).
Step 3. 2-(3,6-dibromo-9H-carbazol-9-y1)-N-phenylacetamide
Br
di 0 0
N
* H ,AN
Br
To a solution of the product of Step 3 (9.6 mg, 0.025 mmol, 1 equiv) in
anhydrous dichloromethane
(1.5 mL) was added N-(3-Dimethylaminopropy1)-N'-ethylcarbodiimide
hydrochloride (EDC, 5.8 mg, 0.03
mmol, 1.2 equiv), 1-hydroxybenzotriazole hydrate (HOBt, 4.1 mg, 0.03 mmol, 1.2
equiv) and 4-
(dimethylamino)pyridine (DMAP, 1 crystal). After the mixture was stirred at rt
for 20 min, aniline (3.4 [LL,
0.0375 mmol, 1.5 equiv) was added. The resulting mixture was heated at 80 C
overnight. The reaction
mixture was diluted with ethyl acetate (20 mL) and washed successively with 1M
NaOH, 1M HC1 and water.
The organic layer was dried with Mg504 and the concentrated to give a poorly
soluble white solid, which was
pure enough to be used in the next step. 1H NMR (d6-DMSO, 400 MHz) 6 = 5.29
(s, 2H) 7.06 (t, J= 7.3 Hz,
1H) 7.31 (t, J= 7.8 Hz, 2H) 7.66- 7.55 (m, 6H) 8.50 (s, 2H) 10.55 (s, 1H). ESI
(m/z): 454.6 (M - H11).
Step 4. P7C3-S115. N-(2-(3,6-dibromo-9H-carbazol-9-yOethyl)aniline
Br
*
0
* N ,,z,
Br
211
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
To a dry and nitrogen flushed vial with the product of Step 3 (9.2 mg, 0.02
mmol, 1 equiv) was added
anhydrous and degassed diethyl ether (750 [LL). The suspension was chilled to
0 C. Lithium aluminum
hydride (1M in THF, 60 [LL, 0.06 mmol, 3 equiv) was added and the mixture was
stirred in ice bath for 1 hour
and at rt overnight. Water (3.6 [tL), 15% NaOH (3.6 [LL) and water (10.8 [LL)
were added successively to the
mixture to quench the reaction. The crude mixture was diluted with ethyl
acetate and washed with brine. The
organic layer was dried with Mg504 and concentrated to give crude product. It
was further purified by silica
gel chromatography (60% of dichloromethane/Hex) to afford 2.7 mg white solid
as product, yield 28.8%. 1H
NMR (CDC13, 400 MHz) 6 = 3.70 - 3.56 (m, 2H) 4.46 (t, J= 5.5 Hz, 2H) 6.55 (d,
J= 7.8 Hz, 2H) 6.76 (t, J=
7.4 Hz, 1H) 7.16 (d, J= 8.8 Hz, 2H) 7.20 (t, J= 7.9 Hz, 2H) 7.50 (dd, J= 8.7,
1.9 Hz, 2H) 8.14 (d, J= 1.7 Hz,
2H). 13C NMR (CDC13, 500 MHz) 6 = 146.8, 139.5, 129.7, 129.4, 123.7, 123.5,
118.4, 113.1, 112.6, 110.5,
42.7, 42.5. ESI (m/z): 486.7 (M + HC00-); 476.7 (M + Cl-).
Example 146. P7C3-S129: 2-(6-Amino-3-imino-3H-xanthen-9-y1)-4-(6-(5-(3-(3-(3,6-
dibromo-9H-
carbazol-9-y1)-2-hydroxypropylamino)phenoxy)pentylamino)-6-
oxohexylcarbamoyDbenzoic acid AND
2-(6- amino-3-imino-3H-xanth en-9-y1)-5-(6-(5-(3-(3-(3,6-dib romo-9H-c arbazol-
9-y1)-2-
hydroxypropylamino)phenoxy)pe ntylamino)-6-oxohexylc arbamoyDbe nzoic acid
H2N
it 0
Br
Br 04. / \ # NH
N 0--- COOH
NH
HO----
HN
.H
Prepared analogously to P7C3-S100. HPLC purification (45% MeCN/H20 + 0.1%
HCO2H,
Phenomenex C18 Luna, 10x250 mm, 3 mL/min) provided 1.7 mg (50% yield) as a
mixture of isomers. ESI
m/z: 1043.2 ([M+H]+, C53H53Br2N607 requires 1043.2).
Example 147. P7C3-S130:
212
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br fa'
\ pH
,0
0/
0
Prepared analogously to example P7C3-S66 from P7C3-S94. Chromatography (1%
Me0H in
dichloromethane) then trituration with hexanes provided 1.2 mg (5.3% yield) of
an off-white solid. 1H NMR
(CDC13, 500 MHz) d = 8.12 (s, 2H), 7.71 (d, J = 7.0 Hz, 2H), 7.54 (d, J = 9.0
Hz, 2H), 7.29 (m, 2H), 6.98 (d,
J = 7.0 Hz, 2H), 4.62 (br s, 1H), 4.39 (s, 2H), 4.19 (s, 2H), 3.88 (s, 2H),
3.72 (m, 11H), 3.42 (s, 1H), 3.23 (d, J
= 5.0 Hz, 1H), 3.16 (s, 1H), 2.49 (t, J = 14.0 Hz, 2H), 1.43 (s, 9H). ESI m/z:
841.6 ([M+HCOO],
C35H42Br2NOIIS requires 842.1).
Example 148. P7C3-S131: 1-(8-bromo-2-methyl-3,4-dihydro-1H-pyrido[4,3-blindol-
5(21/)-y1)-3-
phenoxypropan-2-ol
= N
B r
Powdered KOH (13.6 mg, 0.24 mmol) was added to a solution of 8-bromo-2-methy1-
2,3,4,5-
tetrahydro-1H-pyrido[4,3-Mindole (Boekelheide, V.; Ainsworth, C. J. Am. Chem.
Soc. 1950, 72, 2134) (52.5
mg, 0.20 mmol) in DMF (1.0 mL) at ambient temperature and stirred for 30 min
until dissolved. 2-
(Phenoxymethyl)oxirane was added via syringe and the reaction was stirred at
room temperature overnight.
Upon completion, the solution was diluted with Et0Ac. The mixture was washed
with H20 and brine. The
organic layer was dried over Na2SO4, filtered, and concentrated in vacuo. The
crude residue was purified by
flash column chromatography to afford the product as a white foam (35.3 mg,
43%). 1H NMR (CDC13) 6 =
7.49 (s, 1H), 7.27 (t, J= 7.9 Hz, 2H), 7.18-7.15 (m, 2H), 6.98 (t, J= 7.8 Hz,
1H), 6.81 (d, J= 8.0 Hz, 2H),
4.23 (dd, J= 14.6, 4.5 Hz, 1H), 4.15-4.08 (m, 1H), 4.03 (dd, J= 14.6, 7.1 Hz,
1H), 3.83-3.75 (m, 2H), 3.53-
3.43 (m, 2H), 2.85-2.63 (m, 4H), 2.47 (s, 3H). 13C NMR (CDC13, 126 MHz) 6 =
158.0, 135.4, 135.0, 123.6,
121.3, 114.4, 110.7, 107.7, 69.1, 68.9, 52.2, 51.3, 46.0, 45.6, 23Ø ESI m/z:
414.8 ([M + H]+, C21I-123BrN202
requires 415.0).
213
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 149. P7C3-S137: Synthesis of 1 -(3 -(3 ,6-dibromo -9H-c arb azol-9-y1)-
2-hydroxypropyl)pyridin-
2(1H)-one
Br
Br 401 4.
N
/
0
n-BuLi (2.5 M in hexanes, 80 [tl, 0.2 mmol) was added to a solution of 3,6-
dibromo-9H-carbazole
(32.1 mg, 0.10 mmol) in THF (1.0 mL) at -78 C and stirred for 30 min. 1-
(Oxiran-2-ylmethyl)pyridin-2(1H)-
one1 was added at -78 C and the reaction was stirred at room temperature
overnight. Upon completion, the
solution was quenched with H20. CH2C12 was added and the mixture was washed
with H20, and saturated
aqueous NaCl. The organic layer was dried over Na2504, filtered, and
concentrated in vacuo. The crude
residue was purified by flash column chromatography to afford the product as a
white solid (42.2 mg, 89%).
1H NMR (d6-DMSO, 500 MHz) 6 8.47 (s, 2H), 7.64 - 7.53 (m, 5H), 7.40 (t, 1H, J=
7.2 Hz), 6.35 (d,
1H, J= 9.0 Hz), 6.19 (t, 1H, J= 6.2 Hz), 5.33 (d, 1H, J= 5.5 Hz), 4.44 (d, 1H,
J= 14.8 Hz), 4.35 (dd, 1H, J=
7.9, 14.8 Hz), 4.28 (d, 1H, J = 13.0 Hz), 4.25 -4.17 (m, 1H), 3.74 (dd, 1H, J=
8.8, 12.5 Hz). MS (ESI) m/z:
474.6 ([M + 1-1] , C20I-117Br2N202 requires 475.0).
Example 150. P7C3-S138: 9-(2 -hydroxy-3 -phenoxypropy1)-9H-carb azo le-3 -carb
onitrile
Step 1. Synthesis of 9H-carbazole-3-carbonitrile
ON
0 N4*
H
A solution of 3-bromo-9H-carbazole (123.4 mg, 0.50 mmol) and CuCN (49.9 mg,
0.56 mmol) in N-
methyl-pyrrolidone (2 mL) was heated at 200 C for 5 h. The cooled reaction
mixture was poured into water
and the precipitate was filtered and washed with ethyl acetate. The filtrate
was extracted with ethyl acetate and
the combined ethyl acetate extracts were washed with water, brine, dried
(Na2504), filtered, and concentrated
in vacuo. The crude residue was purified by flash column chromatography to
afford the product as a white
solid (84.7 mg, 88%).
1H NMR (CD30D, 500 MHz) 6 8.46 (s, 1H), 8.13 (d, 1H, J= 7.9 Hz), 7.65 (d, 1H,
J= 8.4 Hz), 7.55
(d, 1H, J= 8.4 Hz), 7.51 (d, 1H, J= 8.1 Hz), 7.47 (t, 1H, J= 7.5 Hz), 7.24 (t,
1H, J= 7.5 Hz), 3.35 (s, 1H). MS
(ESI) m/z: 193.0 ([M + 1-1] , C13H9N2 requires 193.1).
Step 2. Synthesis of 9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carbonitrile
214
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
CN
SNOH
4*
OPh
P7C3-S138 was synthesized and isolated in 61% yield analogously to P7C3-S137
except 9H-
carbazole-3-carbonitrile and 2-(phenoxymethyl)oxirane were used.
1H NMR (CD30D, 400 MHz) 6 8.45 (s, 1H), 8.14 (d, 1H, J= 7.8 Hz), 7.73 - 7.56
(m, 3H), 7.47 (t, 1H,
J= 7.7 Hz), 7.27 (td, 3H, J= 2.0, 7.9 Hz), 6.94 (t, 3H, J= 8.6 Hz), 4.66 (dd,
1H, J= 5.0, 15.0 Hz), 4.52 (dd,
1H, J= 6.8, 15.0 Hz), 4.43 - 4.34 (m, 1H), 3.99 (dd, 1H, J= 5.4, 9.8 Hz), 3.93
(dd, 1H, J= 4.6, 9.8 Hz). MS
(ESI) m/z: 342.9 ([M + 1-1] , C22H19N202 requires 343.1).
Example 151. P7C3-S141: tert-butyl (5-(4-((3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)sulfonyl)phenoxy)pentyl)carbamate
Pr,
-- OH
The title compound was synthesized analogously to P7C3-598.
MS (ESI) m/z: 766.6 [M+formate], C31H36Br2N206S requires 722.1.
Example 152. P7C3-S142: 6-bromo -9 -(2-hydroxy-3 -phenoxypropy1)-9H-carb azo
le-3 -c arb nitrite
CN
Br 4.
OPh
N-bromosuccinimide (8.0 mg, 0.09 mmol) was added to a solution of P7C3-5138
(14.0 mg, 0.04
mmol) in toluene (0.7 mL) and ethyl acetate (0.3 mL) at room temperature. The
reaction was stirred at 70 C
for 2 days, and then cooled to room temperature at which point additional N-
bromosuccinimide (8.1 mg, 0.09
mmol) was added. The reaction was stirred at 70 C for another 2 days. Upon
completion of the reaction as
monitored by 1HNMR, the suspension was washed with water, and dried (Na2504),
filtered, and concentrated
in vacuo. The crude residue was purified by flash column chromatography to
afford the product as a white
solid (9.7 mg, 56%).
215
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 8.31 (s, 1H), 8.20 (d, 1H, J= 1.7 Hz), 7.66 (d, 1H,
J= 8.6 Hz), 7.63 -
7.53 (m, 2H), 7.43 (d, 1H, J= 8.7 Hz), 7.31 (t, 2H, J= 8.0 Hz), 7.02 (t, 1H,
J= 7.4 Hz), 6.89 (d, 2H, J = 8.6
Hz), 4.63 (dd, 1H, J= 5.0, 14.4 Hz), 4.57 - 4.41 (m, 2H), 4.04 (dd, 1H, J=
4.5, 9.4 Hz), 3.91 (dd, 1H, J = 4.5,
9.4 Hz), 2.49 (d, 1H, J= 5.6 Hz). MS (ESI) m/z: 420.8 ([M + H]+, C22H18BrN202
requires 421.1).
Example 153. P7C3-S146: 6-bromo -9-(2 -hydroxy-3 -phenoxypropy1)-9H-carb azo
le-3 -c arb oxamide
CON H2
Br, it
N
\---0Ph
A mixture of P7C3-5142 (4.5 mg, 0.01 mmol), 50% hydrogen peroxide (0.008 mL)
and 1N aqueous
NaOH (0.007 mL) in ethanol (1 mL) was stirred at 30 C for 30 h. Then
additional 50% hydrogen peroxide
(0.008 mL) and 1N aqueous NaOH (0.007 [tt) were added, and the reaction
completed after 15 hat 30 C. The
solution was concentrated and the crude residue was purified by preparative
thin layer chromatography to
afford the product (3.9 mg, 72%).
1H NMR (CDC13, 400 MHz) 6 8.48 (s, 1H), 8.19 (d, 1H, J= 1.6 Hz), 7.86 (dd, 1H,
J= 1.6, 8.6 Hz),
7.58 - 7.46 (m, 2H), 7.40 (d, 1H, J= 8.7 Hz), 7.33 - 7.28 (m, 2H), 7.01 (t,
1H, J= 7.4 Hz), 6.90 (d, 2H, J= 7.8
Hz), 6.08 (s, 1H), 5.61 (s, 1H), 4.61 (t, 1H, J= 8.5 Hz), 4.55 - 4.40 (m, 2H),
4.03 (dd, 1H, J= 4.6, 9.5 Hz),
3.93 (dd, 1H, J= 4.6, 9.5 Hz), 2.73 (s, 1H). MS (ESI) m/z: 438.8 ([M + H]+,
C22H20BrN203 requires 439.1).
Example 154. P7C3-S147: 1 -(3 ,6-dibromo-9H-c arb azol-9-y1)-3 -(pyridin-2 -
ylo xy)prop an-2-ol
Step 1. Synthesis of 3-(pyridin-2-yloxy)propane-1,2-diol
I
NOOH
OH
Following a literature procedure2, a solution of solketal (1.25 mL, 0.01 mol)
in THF (20 mL) was
stirred and cooled to 0 C under N2 and KO13u (1.349 g, 0.012 mol) was added.
The mixture was stirred 15
min, 2-bromopyridine (1.1 mL, 0.011 mol) was added, and the mixture was
stirred 18 h at room temperature,
diluted with H20, and extracted with CH2C12. The combined extracts were dried
(Na2504), filtered, and
evaporated to give the crude title product (288.1 mg, 17%).
1H NMR (CDC13, 400 MHz) 6 7.98 (d, 1H, J= 3.2 Hz), 7.50 (t, 1H, J= 6.8 Hz),
6.81 (t, 1H, J= 5.8
Hz), 6.69 (d, 1H, J= 8.3 Hz), 4.35 (d, 2H, J= 4.5 Hz), 4.09 (s, 1H), 3.95 -
3.83 (m, 1H), 3.63 - 3.46 (m, 2H),
2.81 (s, 1H). MS (ESI) m/z: 170.0 ([M + H]+, C8H12NO3 requires 170.1).
Step 2. 1-(mesityloxy)-3-(pyridin-2-yloxy)propan-2-ol
216
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
I
NOOMes
OH
2-Mesitylenesulfonyl chloride (120.8 mg, 0.55 mmol), Bu2SnO (63.1 mg, 0.25
mmol), DMAP (61.9
mg, 0.51 mmol) and Et3N (1 mL) were added to a solution of 3-(pyridin-2-
yloxy)propane-1,2-diol (83.7 mg,
0.50 mmol) in toluene (5 mL). The reaction mixture was stirred at room
temperature for 2 h. Following the
addition of water, the mixture was extracted with CH2C12. The organic layers
were dried with Na2SO4, and the
solvent was removed under reduced pressure. The residue was purified by flash
column chromatography to
give the title product (123.9 mg, 71%).
1H NMR (CDC13, 400 MHz) 6 8.05 (dd, 1H, J= 1.9, 5.1 Hz), 7.65 -7.56 (m, 1H),
7.01 -6.87 (m, 3H),
6.74 (d, 1H, J= 7.8 Hz), 4.98 (s, 1H), 4.43 (qd, 2H, J= 4.1, 12.2 Hz), 4.15
(s, 1H), 4.02 (d, 2H, J= 5.6 Hz),
2.61 (s, 6H), 2.27 (s, 3H). MS (ESI) m/z: 351.9 ([M + H]+, C17H225N05 requires
352.1).
Step 3. Synthesis of 1-(3,6-dibromo-9H-carbazol-9-y1)-3-(pyridin-2-
yloxy)propan-2-ol (P7C3-S147)
Br
Br 0 *
N
\---0
b
Following Representative Procedure 1, a solution of 1, 3,6-dibromo-9H-
carbazole (32.4 mg, 0.10
mmol) in dry DMF (0.5 mL), was treated KOH (10.2 mg, 0.15 mmol) and 1-
(mesityloxy)-3-(pyridin-2-
yloxy)propan-2-ol (47.5 mg, 0.14 mmol) in dry DMF (1.0 mL) to afford the title
product (34.1 mg, 72%).
1H NMR (d6-DMSO, 400 MHz) 6 8.46 (d, 2H, J= 1.8 Hz), 8.15 - 8.10 (m, 1H), 7.79
- 7.67 (m, 1H),
7.62 (d, 2H, J= 8.8 Hz), 7.56 (dd, 2H, J= 1.9, 8.7 Hz), 7.04 - 6.93 (m, 1H),
6.86 (d, 1H, J= 8.3 Hz), 5.39 (d,
1H, J= 3.6 Hz), 4.53 (dd, 1H, J= 3.6, 14.8 Hz), 4.44 (dd, 1H, J= 6.9, 14.8
Hz), 4.31 - 4.19 (m, 3H). MS (ESI)
m/z: 474.7 ([M + 11] , C20H17Br2N202 requires 475.0).
Example 155. P7C3-S150: 1 -(3 -bromo-9H-c arb azol-9-y1)-3 -phenoxyprop an-2-
ol
Br
lei =
Nv...._c_11.)H
OPh
217
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following Representative Procedure 1, a solution of bromocarbazole (24.7 mg,
0.10 mmol) in dry
DMF (0.6 mL), was treated KOH (10.0 mg, 0.15 mmol) and 2-
(phenoxymethyl)oxirane (30.8 mg, 0.21 mmol)
in dry DMF (0.4 mL) to afford the title product (10.5 mg, 31%).
1H NMR (CDC13, 400 MHz) 6 8.19 (d, 1H, J= 1.8 Hz), 8.03 (d, 1H, J= 7.8 Hz),
7.54 - 7.42 (m, 3H),
7.36 (d, 1H, J= 8.7 Hz), 7.34 - 7.21 (m, 3H), 7.00 (t, 1H, J= 7.4 Hz), 6.89
(d, 2H, J= 7.8 Hz), 4.60 (dd, 1H, J
= 8.1, 16.1 Hz), 4.53 - 4.43 (m, 2H), 4.01 (dd, 1H, J= 4.2, 9.5 Hz), 3.91 (dd,
1H, J= 4.5, 9.5 Hz), 2.44 (d, 1H,
J= 5.6 Hz). MS (ESI) m/z: 395.8 ([M + H]+, C2II-119BrNO2 requires 396.1).
Example 156. P7C3-S151: methyl 6-bromo-9-(2-hydroxy-3 -phenoxypropy1)-9H-c arb
azo le-3 -carb oxyl ate
Step 1. Synthesis of 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-
carboxylic acid
CO2H
Br 0 .
N
\---OPh
Concentrated HC1 (773 [LI-) was added to a solution of P7C3-5142 (10.8 mg,
0.026 mmol) in dioxane
(3.1 mL) at room temperature. The mixture was irradiated at 150 C for 4 h in
a microwave reactor. Upon
completion 1 N NaOH aqueous was added to make pH value about 4. The solution
was diluted with Et0Ac.
The mixture was washed with H20, and saturated aqueous NaCl. The organic layer
was dried over Na2504,
filtered, and concentrated in vacuo to give the crude title product.
Step 2. Synthesis of methyl 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-
3-carboxylate
(P7C3-S151)
CO2Me
Br 0 .
N
\.........(OH
\--0Ph
Concentrated H2504 (20 [tL) was added to a solution of the above crude acid in
Me0H (1 mL) at room
temperature. Then the solution was stirred at 70 C overnight. Upon
completion, the solution was concentrated
in vacuo. The crude residue was purified by flash column chromatography to
afford the product (5.8 mg, 50%
with two steps).
1H NMR (CDC13, 400 MHz) 6 8.75 (s, 1H), 8.25 (d, J= 1.7 Hz, 1H), 8.13 (dd, J=
8.7, 1.5 Hz, 1H),
7.54 (dd, J= 8.7, 1.9 Hz, 1H), 7.49 (d, J= 8.7 Hz, 1H), 7.40 (d, J= 8.7 Hz,
1H), 7.31 (t, J= 8.0 Hz, 2H), 7.01
(t, J= 7.3 Hz, 1H), 6.89 (d, J= 7.9 Hz, 2H), 4.70 - 4.57 (m, 1H), 4.56 - 4.40
(m, 2H), 4.04 (dd, J= 9.6, 4.4 Hz,
218
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H), 4.00 - 3.86 (m, 4H), 2.51 (d, J= 5.2 Hz, 1H). MS (ESI) m/z: 453.8 ([M +
H]+, C23H21BrN04 requires
454.1).
Example 157. P7C3-S153: 6-bromo -9-(2 -hydroxy-3 -phenoxypropy1)-9H-carb azo
le-3 -carboxylic acid
CO2H
Br 0 gr
N
\.......(OH
\--0Ph
LiORH20 was added to a solution of P7C3-5151 in 2 ml- THF/H20/Me0H (v/v/v =
3/1/1) at room
temperature and stirred at 60 C for 3 h. Upon completion, the solution was
treated with 1.0 N HC1 to make pH
value about 3. The mixture was extracted with ethyl acetate and the ethyl
acetate extracts were washed with
H20, and saturated aqueous NaCl. The organic layer was dried over Na2504,
filtered, and concentrated in
vacuo. The crude residue was purified by preparative thin layer chromatography
to afford the product as a
white solid (2.2 mg, 67%).
1H NMR (d6-DMSO, 400 MHz) 6 8.84 (d, 1H, J= 1.3 Hz), 8.54 (d, 1H, J= 1.8 Hz),
8.03 (dd, 1H, J=
1.6, 8.7 Hz), 7.68 (dd, 2H, J= 8.8, 14.4 Hz), 7.59 (dd, 1H, J= 1.9, 8.7 Hz),
7.33 - 7.26 (m, 2H), 6.99 - 6.91
(m, 3H), 5.45 (s, 1H), 4.61 (dd, 1H, J= 4.1, 14.7 Hz), 4.49 (dd, 1H, J= 4.1,
14.8 Hz), 4.31 -4.22 (m, 1H),
4.05 - 3.93 (m, 2H). MS (ESI) m/z: 437.8 ([M - H], C22H17BrN04 requires
438.0).
Examples 158a and 158b. P7C3-S154: 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-
pyrido [2,3-b] indo le-3 -
c arb nitrite and P7C3-S155: 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [2,3 -b]
indole-3 -c arb nitrite
Step 1. Synthesis of 6-amino-5-bromonicotinonitrile
CN
Br
N
H2N
Br2 (0.52 mL, 0.01 mol) was added to a solution of 6-aminonicotinonitrile
(1.1901 g, 0.01 mol) in
AcOH (10 mL) at room temperature. The mixture was stirred at room temperature
for 2 h. Then the mixture
was concentrated and the residue was purified by flash column chromatography
to give the title product (980.0
mg, 49%).
1H NMR (d6-DMSO, 400 MHz) 6 8.36 (s, 1H), 8.19 (s, 1H), 7.33 (bs, 2H). MS
(ESI) m/z: 197.9 ([M +
H]+, C6H5BrN3 requires 198.0).
Step 2. Synthesis of 5,6-dibromonicotinonitrile
219
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
CN
Br 5_N
Br
t-BuONO (77.4 mg, 0.75 mmol) was added to a solution of anhydrous CuBr2 (135.2
mg, 0.61 mmol)
in CH3CN (3 mL) at room temperature. The mixture was heated to 65 C and then
added a suspension of 6-
amino-5-bromonicotinonitrile (98.1 mg, 0.50 mmol) in CH3CN (2 mL). The mixture
was stirred at 65 C for 3
h. Then the mixture was poured into 3M HC1 and extracted with ethyl acetate.
The organic layer was dried
over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by
flash column chromatography
to give the title product (81.0 mg, 62%).
1H NMR (CD30D, 400 MHz) 6 8.70 (d, 1H, J = 2.0 Hz), 8.53 (d, 1H, J = 2.0 Hz).
MS (ESI) m/z:
260.7 ([M HT', C6H3Br2N2 requires 260.9).
Step 3. Synthesis of 5-bromo-6-((4-bromophenyl)amino)nicotinonitrile
ON
Br
Br \ N/1
N
H
Following a literature procedure3, a mixture of 2,4-bromoaniline (59.0 mg,
0.34 mmol), 5,6-
dibromonicotinonitrile (81.0 mg, 0.31 mmol), Pd(OAc)2 (3.6 mg, 0.016 mmol),
PPh3 (8.2 mg, 0.03 mmol), and
Na0t-Bu (36.1 mg, 0.38 mmol) in o-xylene (3 mL) was sparged with N2 for about
5 min at room temperature,
15 placed under N2 atmosphere, and heated at 120 C for 3 h in a screw-
capped sample vial. The reaction mixture
was allowed to cool to room temperature and concentrated in vacuo. The crude
residue was purified by flash
column chromatography to afford the product as a white solid (56.4 mg, 52%).
1H NMR (CDC13, 400 MHz) 6 8.41 (d, 1H, J= 1.7 Hz), 7.94 (d, 1H, J= 1.7 Hz),
7.55 - 7.46 (m, 4H),
7.34 (s, 1H). MS (ESI) m/z: 351.7 ([M + H]+, C12H8Br2N3 requires 351.9).
20 Step 4. Synthesis of the mixture bromo and des-bromo carbolines.
CN CN
Br
0 \ N/1 0 \ 1\11
N N
H H
Following a literature procedure3, Pd(OAc)2 (1.3 mg, 0.006 mmol), PCy3 (3.3
mg, 0.012 mmol), and
DBU (16.0 mg, 0.11 mmol were added to the solution of 5-bromo-6((4-
bromophenyl)amino)nicotinonitrile
(36.0 mg, 0.10 mmol) in DMA (2 mL) at room temperature. The reaction mixture
was sparged for about 5
25 min, placed under N2 atmosphere, and heated at 145 C for about 16 h.
The reaction mixture was concentrated
under reduced pressure. The residue was dissolved in ethyl acetate with
heating at 40-50 C. The mixture was
washed several times with water and then brine, dried over anhydrous Na2504,
filtered, and concentrated. The
220
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
residue was purified by flash column chromatography to give the mixture of 6-
bromo-9H-pyrido[2,3-b]indole-
3-carbonitrile and 9H-pyrido[2,3-b]indole-3-carbonitrile (5.9 mg) and
recovered 5-bromo-6-((4-
bromophenyl)amino)nicotinonitrile (19.8 mg).
Step 5. Synthesis of 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [2,3 -b]
indo le-3 -carb nitrite
(P7C3-S154) and 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [2,3-b] indo le-3 -c
arb onitrile (P7C3-S155)
CN CN
Br
\ 101 N/I
\--OPh \--0Ph
P7C3-S154 P7C3-S155
MeLi (1.6 M in Et20, 108 [tl, 0.17 mmol) was added to a solution of the
mixture of 6-bromo-9H-
carbazole-3-carbonitrile and 9H-carbazole-3-carbonitrile (24.1 mg) in THF (1.0
mL) at -78 C and stirred for
40 min. Phenyl glycidyl ether (24.1 mg, 0.16 mmol) was added at -78 C and the
reaction was stirred at 45 C
overnight. Upon completion, the solution was quenched with H20. Ethyl acetate
was added and the mixture
was washed with H20, and saturated aqueous NaCl. The organic layer was dried
over Na2504, filtered, and
concentrated in vacuo. A small fraction was purified by reverse HPLC to afford
the title products P7C3-S154
and P7C3-5155 .
P7C3-5154: 1H NMR (d6-DMSO, 400 MHz) 6 9.13 (d, 1H, J= 1.9 Hz), 8.91 (d, 1H,
J= 1.9 Hz), 8.55
(d, 1H, J= 1.7 Hz), 7.81 (d, 1H, J= 8.8Hz), 7.74 (dd, 1H, J=1.9, 8.8Hz), 7.27
(t, 2H, J= 7.9 Hz), 6.93 (t, 1H,
J= 7.3 Hz), 6.88 (d, 2H, J= 8.0 Hz), 5.42 (s, 1H), 4.68 (dd, 1H, J= 4.8, 14.3
Hz), 4.61 (dd, J= 1H, 7.7, 14.2
Hz), 4.41 - 4.32 (m, 1H), 4.04 (dd, 1H, J= 4.9, 9.9 Hz), 3.98 (dd, 1H, J= 5.5,
9.9 Hz). MS (ESI) m/z: 421.8
([M + H]+, C21H17BrN302 requires 422.1).
P7C3-5155: 1H NMR (d6-DMSO, 400 MHz) 6 9.11 (d, 1H, J= 2.0 Hz), 8.87 (d, 1H,
J= 2.0 Hz), 8.29
(d, 1H, J= 8.0 Hz), 7.82 (d, 1H, J= 8.3 Hz), 7.60 (dt, 1H, J= 0.8, 7.6 Hz),
7.38 (t, 1H, J= 7.5 Hz), 7.29 - 7.23
(m, 2H), 6.92 (t, 1H, J= 7.3 Hz), 6.88 (d, 2H, J= 7.8Hz), 5.41 (s, 1H), 4.68
(dd, 1H, J= 5.2, 14.3 Hz), 4.62
(dd, 1H, J= 7.4, 14.3 Hz), 4.44 - 4.34 (m, 1H), 4.04 (dd, 1H, J= 4.8, 9.9 Hz),
3.98 (dd, 1H, J= 5.4, 9.9 Hz).
MS (ESI) m/z: 343.8 ([M + H]+, C21H18N302 requires 344.1).
Example 159. P7C3-S157: tert-butyl 3 -(2424243 -((3 -(3 ,6-dibromo -9H-
carbazol-9-y1)-2-
hydroxypropyl)amino)phenoxy)ethoxy)ethoxy)ethoxy)propanoate
5r.,
.- =
OH
0
0 = = 0 0
Br
221
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The title compound was synthesized analogously to P7C3-S219 (see below).
MS (ESI) m/z: 748.7 [M+H]+, C34H42Br2N207requires 748.1.
Example 160. P7C3-S159: 1-(3-bromo-6-methoxy-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Representative procedure 6: Bromination of heterocycles.
Step 1. Synthesis of 3-bromo-6-methoxy-9H-carbazole
OMe
Br is .
NH
3-Methoxy-9H-carbazole (Bedford, R. B.; Betham, M. J. Org. Chem. 2006, 71,
9403-9410) (0.029 g,
0.147 mmol) was dissolved in dry DMF (0.28 mL) and NBS (0.026 g, 0.147 mmol)
was added to the solution.
The reaction mixture was stirred at room temperature for 2 h under absence of
light. The solution was poured
into water (2 mL), filtered and washed with water. The title compound was
isolated as a grey solid (0.033 g,
82%).
1H NMR (CDC13, 400 MHz) 6 8.13 (s, 1H), 7.91 (brs, 1H), 7.50-7.43 (m, 2H),
7.30 (d, 1H, J= 8.7
Hz), 7.25 (d, 1H, J= 5.6 Hz), 7.08 (d, 1H, J= 8.7 Hz), 3.91 (s, 3H). MS (ESI)
m/z 276.9 [M+H]+ ([M+H]+,
C13H11BrNO requires 276.0).
Representative procedure 7: Alkylation of carbazoles with NaH
Step 2. Synthesis of 1-(3-bromo-6-methoxy-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol (P7C3-
S159)
OMe
Br
NOH
\----NH
b
NaH (0.0023 g of 60% suspension (in oil), 0.093 mmol) was added to a solution
of 3-bromo-6-
methoxy-9H-carbazole (0.014 g, 0.052 mmol) in anhydrous THF (0.1 mL) at 0 C
and stirred for 30 min. A
solution of N-(oxiran-2-ylmethyl)aniline (0.0093 g, 0.062 mmol) in anhydrous
THF (0.1 mL) was added
dropwise to the reaction mixture. The reaction was warmed to room temperature
and stirred for 48 h. The
reaction was quenched with saturated aqueous NH4C1 and extracted with Et0Ac.
The combined organics were
concentrated and purified by chromatography (5i02, 0-30% Et0Ac/Hexane) to
afford the title compound
(0.007 g, 25%).
222
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 500 MHz) 6 8.15 (s, 1H), 7.56-7.44 (m, 2H), 7.33 (dd, 2H, J=
8.8, 22.6 Hz), 7.19 (t,
2H, J= 7.7 Hz,), 7.11 (dd, 1H, J= 2.2, 8.8 Hz), 6.80 (t, 1H, J= 7.3 Hz), 6.67
(d, 2H, J= 7.8 Hz), 4.41 (me,
1H), 4.39 (me, 2H), 3.91 (s, 3H), 3.34 (dd, 1H, J= 3.0, 12.9 Hz), 3.20 (dd,
1H, J= 6.9, 12.9 Hz). MS (ESI)
m/z 424.8 [MA-]P ([M+1-1] , C22H22BrN202 requires 425.0).
Example 161. P7C3-S160: 1-(3,6-dibromo-1,4-dimethoxy-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Me0 Br
Br tat *
N OMe
1111r
\--NH
b
Representative procedure 8: Synthesis of carbazoles via Consecutive Amination
and C-H Activation
Step 1. Synthesis of 1,4-dimethoxy-9H-carbazole
Me0
=
0
NH OMe
Following a published procedure (Bedford, R. B.; Betham, M. J. Org. Chem.
2006, 71, 9403-9410),
NaOtBu (0.754 g, 7.84 mmol), Pd(OAc)2 (0.014 g, 0.06 mmol), and [HPtBu3][BF4]
(0.023 g, 0.078 mmol)
were suspended in dry toluene (5 mL) in a microwave vial. 2-Chloroaniline
(0.165 mL, 1.567 mmol) and 2-
bromo-1,4-dimethoxybenzene (0.240 mL, 1.598 mmol) were then added, and the
vial was sealed. The reaction
was then heated in the microwave reactor at 170 C for 4 h, allowed to cool,
and then quenched by addition of
1M HC1. The aqueous phase was extracted with CH2C12, the organic layer was
dried over Na2504, filtered and
condensed. The crude mixture was purified by chromatography (5i02, 0-10%
Et0Ae/Hexane) to afford the
title product (0.193 g, 55%).
1H NMR (CDC13, 500 MHz) 6 8.19 (d, 1H, J= 7.8 Hz), 8.15 (brs, 1H), 7.29-7.22
(m, 2H), 7.10 (t, 1H,
J= 7.2 Hz), 6.63 (d, 1H, J= 8.4 Hz), 6.38 (d, 1H, J= 8.4 Hz), 3.88 (s, 3H),
3.81 (s, 3H). MS (ESI) m/z 228.0
[M+H]+ ([M+14] , C14H14NO2 requires 228.2).
Step 2. Synthesis of 3,6-dibromo-1,4-dimethoxy-9H-carbazole
Me0 Br
Br 10 .
NH OMe
223
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following Representative Procedure 6, 1,4-dimethoxy-9H-carbazole (0.050 g,
0.22 mmol) was treated
with NBS (0.078 g, 0.44 mmol) in dry CH2C12 (11 mL) to afford the title
compound (0.073 g, 88%).
1H NMR (CDC13, 400 MHz) 6 8.33 (brs, 1H), 8.31 (d, 2H, J= 1.8 Hz), 7.52 (dd,
1H, J= 1.8, 8.6 Hz),
7.32 (d, 1H, J= 8.6 Hz), 6.99 (s, 1H), 4.03 (s, 3H), 3.97 (s, 3H). MS (ESI)
m/z 383.7 [M-Hr (EM-HT,
C14H10Br2NO2 requires 384.0).
Step 3. Synthesis of 1-(3,6-dibromo-1,4-dimethoxy-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Me0 Br
Br
Mr
N OMe
\--NH
ö
Following Representative Procedure 7, the title compound was prepared in 46%
yield.
1H NMR (CDC13, 500 MHz) 6 8.32 (s, 1H), 7.54 (dd, 1H, J= 1.7, 8.8 Hz), 7.39
(d, 1H, J= 8.8 Hz),
7.19 (t, 2H, J= 7.8 Hz), 7.01 (s, 1H), 6.75 (t, 1H, J= 7.3 Hz), 6.64 (d, 2H,
J= 7.8 Hz), 4.76 (dd, 1H, J= 3.9,
14.7 Hz), 4.55 (dd, 1H, J= 7.8, 14.7 Hz), 4.37 (me, 1H), 4.01 (s, 3H), 3.94
(s, 3H), 3.35 (dd, 1H, J= 3.8, 13.0
Hz), 3.20 (dd, 1H, J= 7.4, 13.0 Hz). MS (ESI) m/z 534.7 [M+H]+ ([M+14] ,
C23H23Br2N203 requires 535.2).
Example 162. P7C3-S161: 1-(3,6-dibromo-1,8-dimethy1-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Br
Br
Mr N
\--NH
b
The title compound was prepared analogously to P7C3-S160.
1H NMR (CDC13, 500 MHz) 6 7.97 (s, 2H), 7.29 (s, 2H), 7.13 (t, 2H, J= 7.7 Hz),
6.73 (t, 1H, J= 7.3
Hz), 6.43 (d, 2H, J= 8.1 Hz), 4.85 (dd, 1H, J= 8.1, 15.6 Hz), 4.74 (dd, 1H, J=
4.7, 15.6 Hz), 3.92 (me, 1H),
3.86 (me, 1H), 3.00 (dd, 1H, J= 3.4, 13.2 Hz), 2.94 (dd, 1H, J= 7.2, 13.2 Hz),
2.73 (s, 6H). MS (ESI) m/z
502.7 [M+14] ([M+14] , C23H23Br2N20 requires 503.2).
Example 163. P7C3-S164: ethyl 2-(3,6-dibromo-9H-carbazol-9-yl)acetate
224
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 .
N
\...,..e
OEt
Sodium Hydride was added to a stirred solution of 3,6-dibromocarbazole (250
mg, 0.77 mmol) in
DMF (4 m1). The solution was stirred for 30 minutes before the dropwise
addition of ethyl chloroacetate.
After 12 hours water was added and a fine white precipitate formed which was
filtered and rinsed with water
and hexanes to afford the desired ethyl ester in 93% yield.
1H NMR (500 MHz, CDC13) 6 8.15 (s, 2H), 7.56 (d, J= 8.6 Hz, 2H), 7.21 (d, J=
8.5 Hz, 2H), 4.94 (s,
2H), 4.20 (q, J= 6.3 Hz, 2H), 1.26- 1.18 (m, 3H). ESI m/z: 409.7 ([M+1-1] ,
C16H13Br2NO2 requires 409.9)
Example 164. P7C3-S165: 2-(3,6-dibromo-9H-carbazol-9-yl)acetic acid
Br
Br r 4.
IW N
v.......e
OH
Ethyl 2-(3,6-dibromo-9H-carbazol-9-yl)acetate (50 mg, 0.12 mmol) was dissolved
in 0.6 ml of THF.
To this stirred solution was added 0.4 ml of methanol, 0.2 ml of water, and
lithium hydroxide (14.5 mg, 0.6
mmol). After 1 hour all starting material had been consumed. The solution was
acidified with 1N HC1. Upon
reaching a pH of about 4 precipitate had formed which was collected and rinsed
with fresh water to afford the
desired acid in 95% yield.
1H NMR (500 MHz, Acetone-d6) 6 8.41 (s, 2H), 7.62 (dt, J= 8.6, 1.7 Hz, 2H),
7.58 (dd, J= 8.7, 1.5
Hz, 2H), 5.31 (d, J= 1.6 Hz, 2H). ESI m/z: 381.7 ([M+H]+, C14H9Br2NO2 requires
381.9)
Example 165. P7C3-S166: 1-(6-bromo-3-methoxy-1-methy1-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
OMe
Br
IP N
\OH
\--NH
b
The title compound was prepared analogously to P7C3-S160.
1H NMR (CDC13, 400 MHz) 6 8.13 (d, 1H, J= 1.9 Hz), 7.47 (dd, 1H, J= 1.9, 8.7
Hz), 7.34 (d, 1H, J=
2.5 Hz), 7.30 (d, 1H, J= 8.7 Hz), 7.21-7.15 (m, 2H), 6.86 (d, 1H, J= 1.9 Hz),
6.75 (t, 1H, J= 7.3 Hz), 6.60 (d,
2H, J= 7.7 Hz), 4.65 (dd, 1H, J= 8.3, 15.4 Hz), 4.56 (dd, 1H, J= 4.4, 15.4
Hz), 4.31 (me, 1H), 3.89 (s, 3H),
225
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
3.30 (dd, 1H, J= 3.8, 13.0 Hz), 3.21 (dd, 1H, J= 7.2, 13.0 Hz), 2.76 (s, 3H).
MS (ESI) m/z 440.9 [M+H]+
([M+H]+, C23H24BrN202 requires 440.3).
Example 166. P7C3-S167: 1-(4,6-dibromo-3-methoxy-1-methy1-9H-carbazol-9-y1)-3-
(phenylamino)propan-
2-ol
Br OMe
Br tati .
IF N
o\----NH
The title compound was synthesized analogously to P7C3-S160.
1H NMR (CDC13, 400 MHz) 6 9.03 (d, 1H, J= 1.9 Hz), 7.53 (dd, 1H, J= 1.9, 8.8
Hz), 7.29 (d, 1H, J
= 8.8 Hz), 7.22-7.15 (m, H), 6.86 (s, 1H), 6.76 (t, 1H, J= 7.3 Hz), 6.60 (d,
2H, J= 7.7 Hz), 4.65 (dd, 1H, J=
8.4, 15.4 Hz), 4.53 (dd, 1H, J= 4.1, 15.4 Hz), 4.27 (me, 1H), 3.94 (s, 3H),
3.29 (dd, 1H, J= 3.9, 13.1 Hz), 3.20
(dd, 1H, J= 7.2, 13.1 Hz), 2.77 (s, 3H). MS (ESI) m/z 518.7 [M+H]+ ([M+H]+,
C23H23Br2N202 requires 519.2).
Example 167. P7C3-S168: 1-(3,6-dibromo-4-methoxy-9H-carbazol-9-y1)-3-
(phenylamino)propan-2-ol
Me0 Br
Br Ail 41k,
lir N
\----NH
b
The title compound was synthesized analogously to P7C3-5160.
1H NMR (CDC13, 500 MHz) 6 8.46 (s, 1H), 7.62-7.47 (m, 2H), 7.44 (d, 1H, J= 8.7
Hz), 7.18 (t, 2H, J
= 7.6 Hz), 6.75 (t, 1H, J= 7.2 Hz), 6.64 (d, 2H, J= 8.2 Hz), 6.60 (d, 1H, J=
8.5 Hz), 5.04 (dd, 1H, J= 4.1,
15.2 Hz), 4.68 (dd, 1H, J= 8.1, 15.2 Hz), 4.51 (me, 1H), 4.07 (s, 3H), 3.39
(dd, 1H, J= 3.1, 13.0 Hz), 3.24 (dd,
1H, J= 7.5, 13.0 Hz). MS (ESI) m/z 504.7 [M+H]+ ([M+H]+, C22H21Br2N202
requires 505.2).
Example 168. P7C3-S172: 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido13,4-1Aindole-3-
carboxylic acid
Step 1: Ethyl 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-
carboxylate
226
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
0
OEt
\
\--0
Following Representative Procedure 1, the title compound ethyl 9-(2-hydroxy-3-
phenoxypropy1)-9H-
pyrido[3,4-b]indole-3-carboxylate was prepared from Ethyl 9H-pyrido[3,4-
b]indole-3-carboxylate and
phenoxymethyloxirane in 67% yield.
1H NMR (500 MHz, DMSO-d6) d 9.12 (s, 1H), 8.92 (s, 1H), 8.43 (d, J = 7.7 Hz,
1H), 7.80 (d, J = 8.4
Hz, 1H), 7.66 (t, J = 7.4 Hz, 1H), 7.37 (t, J = 7.4 Hz, 1H), 7.30 (t, J = 7.6
Hz, 2H), 6.96 (dd, J = 12.0, 8.0 Hz,
3H), 5.50 (s, 1H), 4.75 (dd, J = 14.8, 3.4 Hz, 1H), 4.64 (dd, J = 14.8, 7.3
Hz, 1H), 4.38 (dd, J=14.2, 7.1 Hz,
1H), 4.31 (s, 1H), 4.03 (p, J = 9.7 Hz, 2H), 1.41-1.32 (m, 3H). ESI m/z: 390.9
([M+H]+, C23H22N204 requires
391.16)
Step 2: P7C3 -S172 : 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [3,4-b]indole-3 -
carboxylic acid
OH
\
Ethyl 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylate (42
mg, 0.107 mmol)
was suspended in 10% NaOH ) and heated to reflux for 3 hours. Upon completion
the reaction was cooled to
room temperature and acidified with ice cold HC1 (cone.). The temperature was
maintained at 0 C and stirred for
1 hour. The precipitate was filtered, rinsed with water, and dried under
vacuum to afford the desired compound
in 92% yield.
1H NMR (500 MHz, DMSO-d6) 6 9.16 (s, 1H), 9.00 (s, 1H), 8.48 (d, J= 7.7 Hz,
1H), 7.83 (d, J= 8.4
Hz, 1H), 7.66 (t, J= 7.4 Hz, 1H), 7.37 (t, J= 7.4 Hz, 1H), 7.30 (t, J= 7.6 Hz,
2H), 6.96 (dd, J= 12.0, 8.0 Hz,
3H), 5.52 (s, 1H), 4.78 (dd, J= 14.8, 3.4 Hz, 1H), 4.68 (dd, J= 14.8, 7.3 Hz,
1H), 4.31 (s, 1H), 4.03 (p, J= 9.7
Hz, 2H). ESI m/z: 362.9 ([M+H]+, C21H18N204 requires 363.13)
Example 169. P7C3-5173: 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-
b]indole-3-carboxylic
acid
Step 1: Synthesis of ethyl 6-bromo-9H-pyrido[3,4-b]indole-3-carboxylate
227
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
0
OEt
¨
Br N
0 \ /
N
H
Following a procedure in Chem.Bio.Chem, 2009, 10, 889-895, N-bromosuccinimide
(37 mg, 0.208
mmol) was added to a solution of ethyl 9H-pyrido[3,4-b]indole-3-carboxylate
(50 mg, 0.208 mmol) in acetic
acid (1.5 ml) and stirred at room temperature. After 45 minutes the acetic
acid was removed and the solid
material was partitioned between Et0Ac and NaHCO3. The organic layer was
washed with water, brine, and
dried over Na2SO4 to afford the desired compound in 98% yield.
1H NMR (500 MHz, DMSO-d6) 6 8.99 (d, J= 3.4 Hz, 2H), 8.70 (s, 1H), 7.67 (dd,
J= 24.8, 8.5 Hz,
2H), 4.37 (q, J= 7.1 Hz, 2H), 1.37 (t, J= 7.1 Hz, 3H). ESI m/z: 318.8 ([M+H]+,
C14H11BrN202 requires 319.0)
Step 2: ethyl 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-
carboxylate (P7C3-
S174)
0
OEt
-
Br N
0 \ /
N
v.......(OH
\-0
*
Following Representative Procedure 1, the title compound ethyl 6-bromo-9-(2-
hydroxy-3-
phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylate was prepared from ethyl 6-
bromo-9H-pyrido[3,4-
b]indole-3-carboxylate and phenoxymethyloxirane in 71% yield.
1H NMR (500 MHz, DMSO-d6) 6 9.26 (s, 1H), 9.19 (s, 1H), 8.87 (s, 1H), 7.90
¨7.80 (m, 2H), 7.30 (t,
J= 6.6 Hz, 2H), 6.96 (d, J= 7.5 Hz, 3H), 4.81 (d, J= 14.9 Hz, 1H), 4.73 (dd,
J= 15.7, 8.6 Hz, 1H), 4.44 (dd, J
= 13.8, 6.8 Hz, 2H), 4.29 (s, 1H), 4.03 (s, 2H), 1.40 (td, J= 7.0, 2.6 Hz,
3H). ESI m/z: 468.8 ([M+H]+,
C23H21BrN204 requires 469.07)
Step 3: P7C3 -S173 : 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [3,4-
b]indole-3 -carboxylic
acid
o
OH
-
Br N
SI \ /
N
µ........(OH
\-0
*
P7C3-S173 was synthesized analogously to P7C3-S172 except ethyl 6-bromo-9-(2-
hydroxy-3-
phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylate was used.
228
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (500 MHz, DMSO-d6) 6 9.35 (s, 1H), 9.27 (s, 1H), 8.91 (s, 1H), 7.90
(dd, J= 24.1, 9.5 Hz,
2H), 7.30 (t, J= 7.4 Hz, 2H), 6.96 (d, J= 7.4 Hz, 3H), 4.89 ¨ 4.73 (m, 3H),
4.29 (s, 1H), 4.04 (d, J= 4.2 Hz,
2H). ESI m/z: 440.8 ([M+H]+, C21H17BrN204 requires 441.0)
Example 170. P7C3-S174: ethyl 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-
pyrido[3,4-b]indole-3-
carboxylate
0
OEt
-
Br N
0 \ /
N
\-0
0
P7C3-S174 was an intermediate in the synthesis of P7C3-S173.
Example 171. P7C3-S175: 9-(2-fluoro-3-phenoxypropy1)-9H-carbazole-3,6-
dicarbonitrile
NC CN
. .
N
0
Y el
F
The title compound was prepared according to the procedure described in
Representative Procedure 4
with Morpho-Dast, except using P7C3-S111 as starting material. The crude
mixture was purified on silica gel
in 100% DCM (+0.2% TEA). Isolated yield=75%.
1H NMR (THF-d8, 400 MHz) 6 8.62 (s, 2H), 7.80 (s, 4H), 7.27 (t, J= 8.2 Hz,
2H), 6.84 (dm, 1H, JI-1-F
= 47.3 Hz), 4.84-5.04 (m, 2H), 4.16-4.34 (m, 2H). MS (ESI), m/z: calculated
369.13, found 413.9
(M+HC00-).
Example 172. P7C3-S176: 1-(cyclopentylamino)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
Br Br
. .
N
H
OH
A solution of 3,6-Dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole (153 mg,
0.40mmol) and cyclopentylamir
(200 [rl, 2.02 mmol) in ethanol (4.0 ml) was heated at 80 C for 3 hours. The
reaction was cooled and condensed tc
give desired in quantitative yield.
1H NMR (CDC13, 400 MHz) 6 8.15 (d, J= 1.9 Hz, 2H), 7.56 (dd, J= 8.7, 2.0 Hz,
2H), 7.38 (d, J= 8.7 Hz,
229
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
2H), 4.32 (d, J= 5.6 Hz, 2H), 4.11 ¨4.00 (m, 1H), 3.75 (q, J= 6.6 Hz,1H), 3.06
¨ 2.95 (m, 1H), 2.79 (dd, J= 12.1
Hz, 1H), 2.52 (dd, J= 12.0, 8.8 Hz, 1H), 1.90 ¨ 1.39 (m, 6H), 1.32¨ 1.17 (m,
2H). MS (ESI), m/z: calculated 464.
found 508.7 (M+HC00-).
Example 173. P7C3-S177: 9-(2-hydroxy-2-methyl-3-phenoxypropy1)-9H-carbazole-
3,6-dicarbonitrile
Step 1: 1-(3,6-dibromo-9H-carbazol-9-y1)-2-methy1-3-phenoxypropan-2-ol
Br Br
=
0
HO
Methylmagnesium bromide (92 IL, 3.0 M in THF) was added to an ice-cooled
solution of title
compound of example 103, step 1, 1-(3,6-dibromo-9H-carbazol-9-y1)-3-
phenoxypropan-2-one (86 mg, 0.18
mmol) in anhydrous THF (1.8 m1). The reaction was stirred in the gently
warming ice bath for 6 hours, and
then quenched by addition of water. The crude mixture was diluted with Et0Ac
and washed with saturated
sodium bicarbonate, water and then brine. The organic layer was dried over
Na2504, filtered and condensed.
Some unreacted starting material was precipitated from ¨50% acetone/hexanes.
The condensed filtrate was
carried forward. Yield=89%. MS (ESI), m/z: calculated 486.98, found 531.7
(M+HC00-).
Step 2: 9-(2-hydroxy-2-methyl-3-phenoxypropy1)-9H-carbazole-3,6-dicarbonitrile
(P7C3-S177)
NC CN
*
NL
0
An oven dried vial was charged with 1-(3,6-dibromo-9H-carbazol-9-y1)-2-methy1-
3-phenoxypropan-2-
ol (79.5 mg, 0.16 mmol), copper iodide (16 mg, 0.08 mmol), sodium cyanide (22
mg, 0.45 mmol) and
potassium iodide (16 mg, 0.10 mmol). The sealed vial was nitrogen filled and
evacuated three times before
addition of N, N-dimethy1-1,2-ethanediamine (22.5 IL, 0.21 mmol) and
anhydrous toluene (75 I). The
reaction was heated at 100 C over 60 hours. The cooled mixture was diluted
with EtOac, washed with water
and brine. The organic layer was dried over Na2504, filtered and condensed.
The crude mixture was purified
on silica gel in 40%THF/hexanes. Yield=34%.
1H NMR (THF-d8, 400 MHz) 6 8.49 (d, J= 1.0 Hz, 2H), 7.79 (d, J= 8.6 Hz, 2H),
7.60 (dd, J= 8.7, 1.5 IL
2H), 7.17 (d, J= 8.0 Hz, 2H), 6.89-6.74 (m, 3H), 4.46-4.70 (m, 2H), 371-3.89
(m, 2H), 1.32 (s, 3H). MS (ESI), mI
calculated 381.15, found 425.9 (M+HC00-).
Example 174. P7C3-S178: 1-(cyclohexyloxy)-3-(3,6-dibromo-9H-carbazol-9-
yl)propan-2-ol
230
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br Br
Nyo
OH
A spatula tip of sodium hydride (60% suspension in mineral oil) was added to
solution of 3,6-Dibromo-9-
(oxiran-2-ylmethyl)-9H-carbazole (Epoxide 2-A) (145 mg, 0.38 mmol) in
cyclohexanol (10 m1). The reaction was
stirred overnight at 60 C. The mixture was washed with water and then dried
over several days under vacuum.
Yield=45%
1H NMR (CDC13, 400 MHz) 6 8.14 (d, J= 1.9 Hz, 2H), 7.56 (dd, J= 8.7, 1.9 Hz,
2H), 7.39 (d, J= 8.7
Hz, 2H), 4.43 (dd, J= 15.0, 6.8 Hz, 1H), 4.32 (dd, J= 14.9, 5.7 Hz, 1H), 4.18
(m, 1H), 3.49 (dd, J= 9.4, 4.1
Hz, 1H), 3.27 (m, 2H), 2.53 (d, J= 6.0 Hz, 1H),1.89 (m, 2H), 1.79¨ 1.69 (m,
2H), 1.56 (m, 2H), 1.40¨ 1.17
(m, 4H). MS (ESI), m/z: calculated 479.01, found 523.7 (M+HC00-).
Example 175. P7C3-S179: (E)-N-(3 -(3,6-dibromo-9H-carbazol-9-yl)prop-1 -en-l-
y1)-1,1,1 -trifluoro-N-(3 -
methoxyphenyl)methanesulfonamide
Br Br
=
N
OMe
µSO2CF3
The title compound was isolated as a by-product from the reaction of N-(3-(3,6-
dibromo-9H-carbazol-9-3/1
fluoropropy1)-1,1,1-trifluoro-N-(3-methoxyphenyl)methanesulfonamide (P7C3-
S241) in toluene with Red-Al at 8(
It was purified with column chromatography in 10%Et0Ac/hexanes.
1H NMR (CDC13, 400 MHz) 6 8.13 (d, J= 1.9 Hz, 2H), 7.55 (dd, J= 8.6, 2.0 Hz,
2H), 7.32 (t, J= 8.2
Hz, 1H), 7.21 (d, J= 8.7 Hz, 2H), 7.01 (d, J= 13.4 Hz, 1H), 6.98 ¨6.93 (m,
1H), 6.80 (dd, J= 7.9, 1.9 Hz,
1H), 6.73 (t, J= 2.3 Hz, 1H), 4.83 (d, J= 6.7 Hz, 2H), 4.76 (ddd, J= 12.8,
7.2, 5.4 Hz, 1H), 3.75 (s, 3H). MS
(ESI), m/z: calculated 615.93, found 660.5 (M+HC00-).
Example 176. P7C3-S180: 1-(9H-carbazol-9-y1)-3-(naphthalen-1-ylamino)propan-2-
ol
=
OH
The title compound was prepared using representative procedure 2 and 9-(oxiran-
2-ylmethyl)-9H-carbazo
Chromatography in 10%Et0Ac/hexanes gave desired in 49% yield.
231
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 8.13 (d, J= 7.7 Hz, 2H), 7.83 (ddd, J= 9.7, 6.8, 3.0
Hz, 2H), 7.57 ¨
7.43 (m, 6H), 7.37 ¨ 7.27 (m, 4H), 6.58 (dd, J= 6.6, 2.0 Hz, 1H), 4.77 (s,
1H), 4.68 ¨ 4.50 (m, 3H), 3.55 (dd, J
= 12.6, 3.6 Hz, 1H), 3.41 (dd, J= 12.7, 6.9 Hz, 1H), 2.23 (s, 1H). MS (ESI),
m/z: calculated 366.17, found
367.0 (M+1).
Example 177. P7C3-S183: 1-(8-bromo-5H-pyrido[4,3-b]indo1-5-y1)-3-phenoxypropan-
2-ol
____N
Br ao, /
N
\--0
=
The tile compound was synthesized analogously to P7C3-S160 using phenyl
glycidyl ether and the
appropriate carboline (Sako, K. et al. Bioorg. Med. Chem. 2008, 16, 3780-3790)
1H NMR (CDC13-Me0D [4:2], 500 MHz) 6 8.98 (s, 1H), 8.25 (d, 1H, J= 5.9 Hz),
8.12 (d, 1H, J= 1.5
Hz), 7.46 (d, 1H, J= 8.6 Hz), 7.40-7.33 (m, 2H), 7.19 (t, 2H, J= 7.8 Hz), 6.88
(t, 1H, J= 7.3 Hz), 6.81 (d, 2H,
J= 8.5 Hz), 4.52 (dd, 1H, J= 4.9, 14.8 Hz), 4.36 (dd, 1H, J= 6.4, 14.8 Hz),
4.30 (me, 1H), 3.85 (me, 2H). MS
(ESI) m/z 398.8 [M+H]+ ([M+H]+, C20H18BrN202 requires 398.2).
Example 178. P7C3-S184: 1-(3,6-dichloro-9H-carbazol-9-y1)-3-(naphthalen-1-
ylamino)propan-2-ol
CI CI
* =
N
H'il Os
OH
The title compound was prepared analogously to P7C3-S180. Chromatography with
10% Et0Ac/hexanes
desired in 59% yield.
1H NMR (400 MHz, Acetone-d6) 6 8.22 (d, J= 2.0 Hz, 2H), 8.17 ¨ 7.96 (m, 1H),
7.88 ¨ 7.73 (m, 1H),
7.68 (d, J= 8.8 Hz, 2H), 7.59 ¨ 7.34 (m, 4H), 7.26 (m, 1H), 7.21 (d, J= 8.1
Hz, 1H), 6.61 (d, J= 7.4 Hz, 1H),
5.63 (d, J= 5.1 Hz, 1H), 4.71 (m, 1H), 4.60 (m, 2H), 3.58 (dm, 3H). MS (ESI),
m/z: calculated 434.10, found
434.9 (M-1).
Example 179. P7C3-S186: 1-(6-bromo-9H-pyrido[2,3-b]indo1-9-y1)-3-phenoxypropan-
2-ol
232
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br 40 4
The title compound was synthesized analogously to P7C3-S160 using a-carboline
(Sako, K. et al.
Bioorg. Med. Chem. 2008, 16, 3780-3790) and phenyl glycidyl ether.
1H NMR (CDC13, 500 MHz) 6 8.46 (d, 1H, J= 4.7 Hz), 8.29 (d, 1H, J= 7.6 Hz),
8.15 (s, 1H), 7.55 (d,
1H, J= 8.7 Hz), 7.42 (d, 1H, J= 8.7 Hz), 7.27 (t, 2H, J= 7.9 Hz), 7.23 (dd,
1H, J= 5.0, 7.5 Hz), 6.96 (t, 1H, J
= 7.3 Hz), 6.88 (d, 2H, J= 8.1 Hz), 4.73 (dd, 1H, J= 2.6, 14.9 Hz), 4.66 (dd,
1H, J= 5.6, 14.9 Hz), 4.53 (me,
1H), 4.07 (dd, 1H, J= 5.1, 9.1 Hz), 3.86 (dd, 1H, J= 7.6, 9.1 Hz). MS (ESI)
m/z 397.8 [M+H]+ ([M+H]+,
C20H18BrN202 requires 398.2).
Example 180. P7C3-S187: 1-(3,6-dibromo-9H-pyrido[2,3-b]indo1-9-y1)-3-
phenoxypropan-2-ol
Br
Br au N
o
The title compound was synthesized analogously to P7C3-5186, except excess NBS
was used in the
bromination.
1H NMR (CDC13-Me0D [4:2], 500 MHz) 6 8.39 (s, 1H), 8.33 (s, 1H), 8.05 (s, 1H),
7.52-7.43 (m, 1H),
7.43-7.32 (m, 1H), 7.18 (t, 2H, J= 7.1 Hz), 6.87 (t, 1H, J= 7.3 Hz), 6.78 (d,
2H, J= 7.9 Hz), 4.60 (dd, 1H, J=
4.6, 14.7 Hz), 4.53 (dd, 1H, J = 6.0, 14.7 Hz), 4.38 (me, 1H), 3.89 (me, 2H).
MS (ESI) m/z 476.7 [M+H]+
([1\4 H]+, C20H17Br2N202 requires 477.1).
Example 181. P7C3-S188: 1-(3,6-dibromo-9H-carbazol-9-y1)-3-((6-methoxypyridin-
2-yl)amino)propan-2-ol
',6r1
?
ChN
The title compound was prepared analogously to P7C3-S10, also known as
P7C3A20.
MS (ESI) m/z: 503.7 [M+H]+, C21H19Br2N302requires 503Ø
Example 182. P7C3-S190: 1-(6-bromo-3-methy1-9H-pyrido[3,4-b]indo1-9-y1)-3-
phenoxypropan-2-ol
233
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
CH3
¨
Br N
0 \ /
N
\......./=)H
\--0
0
Step 1: 6-bromo-9H-pyrido[3,4-b]indo1-3-yl)methanol(P7C3-S204)
OH
¨
Br N
0 \ /
N
H
Ethyl 6-bromo-9H-pyrido[3,4-b]indole-3-carboxylate (141 mg, 0.44 mmol) and
lithium borohydride
(19 mg, 0.88 mmol) were dissolved in THF (2 ml) and stirred at 60 C
overnight. Upon completion, the
reaction was quenched with 1N HC1. The mixture was extracted with CH2C12(3x),
washed with brine, H20,
and dried over Na2SO4. The material was used in the next step without
purification.
1H NMR (500 MHz, DMSO-d6) 6 12.05 (s, 1H), 8.99 (s, 1H), 8.74 (s, 1H), 8.65
(s, 1H), 7.77 (d, J=
8.7 Hz, 1H), 7.67 (d, J = 8.7 Hz, 1H), 4.90 (s, 2H). ESI m/z: 276.8 ([M+H]+,
C12H9BrN20 requires 276.99)
Step 2: 6-bromo-9H-pyrido[3,4-b]indole-3-carbaldehyde
0
H
¨
Br N
N
H
Following a published procedure (J.Med.Chem., 1982, 25, 1081) 6-bromo-9H-
pyrido[3,4-b]indo1-3-
yl)methanol (253 mg, 1.27 mmol) and Mn02 (300 mg, 3.44 mmol) were combined in
acetonitrile (5 ml) and
stirred at reflux overnight. The material was cooled to room temperature and
filtered over a pad of celite. This
was rinsed with hot CH3CN to give the desired aldehyde in 58% yield.
1H NMR (500 MHz, DMSO-d6) 6 12.36 (s, 1H), 10.11 (s, 1H), 9.10 (s, 1H), 8.90
(s, 1H), 8.73 (s, 1H),
7.75 (d, J= 8.8 Hz, 1H), 7.67 (d, J= 8.6 Hz, 1H). ESI m/z: 274.8 ([M+H]+,
C12H7BrN20 requires 274.97)
Step 3: 6-bromo-3-methy1-9H-pyrido[3,4-b]indole (P7C3-S226)
CH3
Br N
lei \ /
N
H
234
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following a published procedure (Tetrahedron, 2002, 5251) 6-bromo-9H-
pyrido[3,4-b]indole-3-
carbaldehyde (40 mg, 0.145 mmol), hydrazine hydrate (51 mg, 1.02 mmol) and KOH
(29 mg, 0.509 mmol)
were combined in 0.5 mL of ethylene glycol and heated to 150 C overnight. The
reaction was cooled to room
temperature. Ice chips were added and the mixture was stirred in an ice bath
for 1 hour. White precipitate
formed which was filtered and dried to afford the desired compound in 73%
yield.
1H NMR (500 MHz, DMSO-d6) 6 11.59(s, 1H), 8.77(s, 1H), 8.42(s, 1H), 7.97(s,
1H), 7.61 (d, J=
8.4 Hz, 1H), 7.51 (d, J= 8.0 Hz, 1H), 2.58 (s, 3H). ESI m/z: 260.8 ([M+H]+,
C12H9BrN2 requires 260.99)
Step 4: P7C3 -S190: 1-(6-bromo-3-methy1-9H-pyrido[3,4-b]indo1-9-y1)-3-
phenoxypropan-2-ol
CH3
Br
¨
N
lei N\
/
N
vs....(OH
\--0
411D
Following Representative Procedure 1, the title compound 1-(6-bromo-3-methy1-
9H-pyrido[3,4-
b]indo1-9-y1)-3-phenoxypropan-2-ol was prepared from 6-bromo-3-methy1-9H-
pyrido[3,4-b]indole and
phenoxymethyloxirane in 78% yield after a precipitation from isopropanol.
1H NMR (500 MHz, DMSO-d6) 6 8.93 (s, 1H), 8.47 (s, 1H), 8.01 (s, 1H), 7.70
¨7.61 (m, 2H), 7.30 (t,
J= 7.7 Hz, 2H), 6.95 (d, J= 7.1 Hz, 3H), 5.47 (s, 1H), 4.63 (d, J= 15.7 Hz,
1H), 4.51 (dd, J= 15.3, 6.7 Hz,
1H), 4.26 (s, 1H), 4.02 ¨ 3.91 (m, 2H), 2.60 (s, 3H). ESI m/z: 410.8 ([M+H]+,
C21H19BrN204 requires 411.06)
Example 183. P7C3-5191: 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-
b]indole-3-carboxamide
0
NH2
¨
Br N
Si \ /
N
\......,(OH
\--0
*
6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-carboxylic acid
(P7C3- S174) (30
mg, 0.068 mmol) was added to 0.5 ml. CH2C12 with a catalytic amount of DMF.
Oxalyl chloride (60 ul, 0.68
mmol) was added and the solution was stirred for 1 hour at room temperature.
The reaction was concentrated
to remove solvent, and NH3 in dioxane was added. Upon completion of the
reaction, the solvent was removed
under vacuum to give the desired compound in 92% yield.
235
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (500 MHz, DMSO-d6) 6 9.06 (s, 1H), 8.90 (s, 1H), 8.66 (s, 1H), 7.98 (s,
1H), 7.73 (dt, J=
8.8, 5.3 Hz, 2H), 7.34 (s, 1H), 7.33 ¨ 7.25 (m, 2H), 6.96 (d, J= 8.5 Hz, 3H),
5.41 (s, 1H), 4.73 (dd, J= 15.0,
3.9 Hz, 1H), 4.63 (dd, J= 15.0, 7.4 Hz, 1H), 4.31 (s, 1H), 4.03 (d, J= 5.4 Hz,
2H). ESI m/z: 439.8 ([M+H]+,
C21H1813rN303 requires 440.05)
Example 184. P7C3-S192: 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-
b]indole-3-carbonitrile
Step 1: 6-bromo-9H-pyrido[3,4-b]indole-3-carbonitrile (P7C3-S221)
CN
-
Br N
0 \ /
N
H
6-bromo-9H-pyrido[3,4-b]indole-3-carbaldehyde (30 mg, 0.11 mmol) was suspended
in 100 ul of
THF. Ammonium hydroxide (1 mL) and iodine (31 mg, 0.12 mmol) were added and
the mixture was stirred at
room temperature for 1 hour. Upon completion, Na2S203 (5 ml) was added and was
extracted 3X with diethyl
ether. Material was used in the next step without further purification.
1H NMR (500 MHz, DMSO-d6) 6 12.26 (s, 1H), 9.05 (s, 1H), 8.92 (s, 1H), 8.62
(s, 1H), 7.77 (d, J=
8.7 Hz, 1H), 7.68 (d, J= 8.7 Hz, 1H). ESI m/z: 271.8 ([M+H]+, C12H6BrN3
requires 271.97)
Step 2: 6-bromo-9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido[3,4-b]indole-3-
carbonitrile (P7C3 -S192)
CN
-
Br N
Si \ /
N
\---0
*
6-bromo-9H-pyrido[3,4-b]indole-3-carbonitrile (21.3mg, 0.785mmo1) was added to
a flask and purged
with nitrogen. THF was added and the solution was cooled to -78 C. Methyl
lithium (1.6 M in Et20) (1.1 eq)
was added and was stirred for 30 minutes. 1,2-epoxy-3-phenoxypropane (0.011
mL, 0.824 mmol) was added
with gradual warming to 45 C. Upon completion the reaction was quenched with
1 N HC1 and extracted 3X
with Et0Ac. The organic layer was washed with H20, brine, and dried over
Na2504. The crude mixture was
purified on 5i02 (0 to 50% Et0Ac/hexanes to afford the desired product in 90%
yield.
1H NMR (500 MHz, DMSO-d6) 6 9.24 (s, 1H), 8.94 (s, 1H), 8.66 (s, 1H), 7.86 ¨
7.77 (m, 2H), 7.30 (d,
J= 3.9 Hz, 2H), 6.95 (d, J= 3.4 Hz, 3H), 5.50 (s, 1H), 4.77 (d, J= 15.1 Hz,
1H), 4.71 ¨4.62 (m, 1H), 4.28 (s,
1H), 4.01 (s, 2H). ESI m/z: 421.8 ([M+H]+, C21H1813rN303 requires 422.04)
Example 185. P7C3-5194: 1-(8-bromo-5H-pyrido[3,2-b]indo1-5-y1)-3-phenoxypropan-
2-ol
236
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
N¨
Br .\ /
N
\---0
The title compound was synthesized analogously to e P7C3-S186.
1H NMR (CDC13, 500 MHz) 6 8.57 (d, 1H, J= 4.5 Hz), 8.51 (d, 1H, J= 1.6 Hz),
7.85 (d, 1H, J= 8.3
Hz), 7.59 (dd, 1H, J= 1.8, 8.7 Hz), 7.48 (d, 1H, J= 8.7 Hz), 7.38-7.31 (m,
1H), 7.31-7.26 (m, 1H), 7.23-7.17
(m, 1H), 7.04 (t, 1H, J= 7.3 Hz), 6.94 (d, 2H, J= 8.0 Hz), 5.50 (me, 1H), 4.76
(dd, 1H, J= 6.5, 15.2 Hz), 4.62
(dd, 1H, J= 5.9, 15.2 Hz), 4.07 (dd, 1H, J= 4.8, 10.4 Hz), 3.98 (dd, 1H, J=
3.5, 10.4 Hz). MS (ESI) m/z 398.8
[M+H]+ ([M+H]+, C20H18BrN202 requires 398.2).
Example 186. P7C3-S195: 8-bromo-5-(2-hydroxy-3-phenoxypropy1)-5H-pyrido[4,3-
b]indole 2-oxide
00
_NO
Br Au \ I
Illr N
\----0
b
To a stirred solution of P7C3-5183 (0.029 g, 0.073 mmol) in CHC13:Et0H (1:1)
(0.072 mL) was
added mCPBA (0.057 g, 0.252 mmol). The mixture was stirred under reflux for 30
min. The cooled reaction
was treated with 2M NaOH and the mixture stirred for 30 min at room
temperature. The aqueous phase was
extracted with CH2C12, the organic layer was dried over Na2504, filtered and
condensed. The crude mixture
was purified by chromatography (5i02, 0-10% Me0H/CH2C12) to afford the title
compound (0.029 g, 90%).
1H NMR (CDC13-Me0D [4:2], 500 MHz) 6 8.82 (s, 1H), 8.12 (d, 1H, J= 1.5 Hz),
8.09 (d, 1H, J= 6.9
Hz), 7.55 (dd, 1H, J= 1.7, 8.8 Hz), 7.52 (d, 1H, J= 7.1 Hz), 7.41 (d, 1H, J=
8.8 Hz), 7.26-7.21 (m, 2H), 6.93
(t, 1H, J= 7.3 Hz), 6.85 (d, 2H, J= 8.1 Hz), 4.57 (dd, 1H, J= 3.9, 15.1 Hz),
4.41 (dd, 1H, J= 6.7, 15.1 Hz),
4.21 (me, 1H), 3.93 (dd, 1H, J= 4.3, 9.5 Hz), 3.86 (dd, 1H, J= 7.0, 9.5 Hz).
MS (ESI) m/z 414.8 [M+H]+
([M+H]+, C20H18BrN203 requires 414.2).
Example 187. P7C3-S198: 8-bromo-5-(2-hydroxy-3-phenoxypropy1)-5H-pyrido[3,2-
b]indole 1-oxide
237
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
a
o, e
N-
Br lio \ /
1\1OH
\-0
P7C3-S198 was synthesized and isolated in 53% yield analogously to P7C3-S195
except P7C3-S194
was used.
1H NMR (CDC13-Me0D [4:2], 500 MHz) 6 8.68 (s, 1H), 7.99 (s, 1H), 7.57 (me,
1H), 7.48 (d, 1H, J=
8.7 Hz), 7.35 (d, 1H, J= 8.7 Hz), 7.18 (me, 1H), 7.14 (t, 2H, J= 7.6 Hz), 6.82
(t, 1H, J= 7.1 Hz), 6.76 (d, 2H,
J= 8.4 Hz), 4.51 (dd, 1H, J= 3.7, 15.1 Hz), 4.35 (dd, 1H, J= 6.5, 15.1 Hz),
4.24 (me, 1H), 3.86-3.74 (m, 2H).
MS (ESI) m/z 412.8 [M-Hr ([M-H], C20H18BrN203 requires 412.2).
Example 188. P7C3-S204: (6-bromo-9H-pyrido[3,4-b]indo1-3-yl)methanol
OH
-
Br N
N
H
The title compound was an intermediate in the synthesis of P7C3-S190.
Example 189. P7C3-S205: ethyl 6-bromo-9H-pyrido[3,4-b]indole-3-carboxylate
0
OEt
-
Br N
N
H
The title compound was an intermediate in the synthesis of P7C3-S173.
Example 190. P7C3-S208: tert-butyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropyl)carbamate
Step 1: 1-azido-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol
Br
Br 0 *
N3
3,6-dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole (500 mg, 1.3 mmol), NaN3 (111
mg, 1.7 mmol),
NH4C1 (91 mg, 1.7 mmol), were combined in 4 ml of Et0H and 1 ml of H20 and
heated to 80 C overnight.
Upon completion, the Et0H was evaporated and the mixture was partitioned
between Et0Ac and H20. The
238
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
organic layer was washed with brine and dried over Na2SO4 and concentrated.
The material was used in the
next step without further purification.
1H NMR (500 MHz, CDC13) 6 8.14 (s, 1H), 7.57 (d, J= 8.7 Hz, 1H), 7.34 (d, J=
8.7 Hz, 1H), 4.35 (d,
J= 5.9 Hz, 1H), 4.25 (dd, J= 9.9, 4.9 Hz, 1H), 3.49 (dt, J= 12.4, 4.5 Hz, 1H),
3.38 ¨3.30 (m, 1H), 2.15 (s,
1H). ESI m/z: 466.7 ([M+HCOO], C12H7BrN202 requires 424.1)
Step 2: 9-(3-azido-2-fluoropropy1)-3,6-dibromo-9H-carbazole
Br
Br 40 #
N
N3
Synthesized according to representative procedure 4, except 1-azido-3-(3,6-
dibromo-9H-carbazol-9-
yl)propan-2-ol was used.
1H NMR (500 MHz, CDC13) 6 8.15 (s, 2H), 7.59 (d, J= 8.7 Hz, 2H), 7.33 (d, J=
8.7 Hz, 2H), 5.08 ¨
4.90 (m, 1H), 4.55 (dt, J= 9.5, 5.1 Hz, 2H), 3.62 ¨ 3.52 (m, 1H), 3.39 (ddd,
J= 24.1, 13.7, 4.7 Hz, 1H). ESI
m/z: 469.6 ([M+HCOO], C20H11Br2FN4 requires 423.94)
Step 3: 3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropan-1-amine
Br
Br 0 4.
Nv....."
\--NH2
9-(3-azido-2-fluoropropy1)-3,6-dibromo-9H-carbazole (60 mg, 0.14 mmol) and
triphenylphosphine
(44 mg, 0.168 mmol) were combined in 0.5 m1_, of THF and stirred overnight at
60 C. Upon completion 1 ml
of H20 was added and the mixture was stirred for 1 hour. The mixture was
extracted with Et0Ac. The organic
layer was washed with H20, brine, and dried over Na2504 and concentrated to a
white foam. This material was
used in the next step without further purification.
1H NMR (500 MHz, Me0H-d4) 6 8.29 (s, 1H), 7.60 (d, J= 8.7 Hz, 1H), 7.53 (s,
1H), 5.16 (d, J= 50.4
Hz, 1H), 4.73 (dd, J= 23.1, 5.3 Hz, 1H), 3.43 (d, J= 14.2 Hz, 1H), 3.24 ¨ 3.11
(m, 1H). ESI m/z: 398.7
([M+1-1] , C15H13Br2FN2 requires 398.94)
Step 4: P7C3-S208: tert-butyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropyl)carbamate
239
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Br
Br 0 4.
NJ\_I 0 oy.....
----N
H
3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropan-1-amine (21 mg, 0.0524 mmol)
was dissolved in
500 ul of THF and cooled to 0 C. Boc anhydride (12.6 mg, 0.0576 mmol) was
dissolved in THF and added
dropwise. The reaction was stirred overnight at room temperature. Upon
completion the material was
concentrated to remove excess THF and partitioned between Et0Ac and H20. The
organic layer was washed
with brine and dried over Na2SO4. The crude mixture was purified on Si02 (0 to
50% Et0Ac/hexanes to afford
the desired product in 42% yield.
1H NMR (500 MHz, CDC13) 6 8.15 (s, 2H), 7.57 (d, J= 8.7 Hz, 2H), 7.30 (d, J=
8.7 Hz, 2H), 4.94 (d,
J= 57.3 Hz, 2H), 4.59 - 4.39 (m, 2H), 3.68 - 3.52 (m, 1H), 3.30 (ddd, J= 21.4,
13.4, 6.2 Hz, 1H), 1.46 (s,
9H). ESI m/z: 498.8 ([M+H]+, C20H21Br2FN202 requires 499.0)
Example 191. P7C3-S213: 2-(3,6-dibromo-9H-carbazol-9-yl)acetamide
Br
Br
N
NH2
Ethyl 2-(3,6-dibromo-9H-carbazol-9-yl)acetate (P7C3-S164) (10 mg,0.024 mmol)
and ammonium
hydroxide (100 ul) were stirred at 60 C overnight. Upon completion the
reaction was filtered and rinsed with
H20 to give the desired product in quantitative yield.
1H NMR (500 MHz, Acetone-d6) 6 8.40 (d, J= 1.9 Hz, 2H), 7.61 (dd, J= 8.7, 1.9
Hz, 2H), 7.54 (d, J=
8.7 Hz, 2H), 7.07 (s, 1H), 6.64 (s, 1H), 5.08 (s, 2H). ESI m/z: 424.7
([M+HCOO], C14H10Br2FN20 requires
378.92)
Example 192. P7C3-S214: 2-(3,6-dibromo-9H-carbazol-9-y1)-N-methylacetamide
Br
Br 0 fik
N
\........e
HN-cH3
2-(3,6-dibromo-9H-carbazol-9-yl)acetic acid (P7C3-S165) (50 mg, 0.13 mmol) was
suspended in 1 ml
of dichloromethane. Methylamine (2.0 M in THF) (0.144 mmol) was added followed
by a catalytic amount of
240
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
dimethylamino pyridine. The mixture was cooled to 0 C in an ice bath and a
solution of dicyclohexyl
carbodiimide in 1 ml of dichloromethane was added dropwise. The mixture was
stirred overnight at room
temperature. Upon completion, the DCM mixture was washed with H20, brine,
dried over Na2SO4, and
concentrated. The crude mixture was purified on Si02 (0 to 50% Et0Ac/hexanes)
followed by a precipitation
from Me0H/H20 to afford the desired product in 42% yield.
1H NMR (500 MHz, CDC13) 6 8.19 (s, 2H), 7.61 (d, J= 8.6 Hz, 2H), 7.23 (s, 2H),
5.37 (s, 1H), 4.89
(s, 2H), 2.72 (d, J= 4.9 Hz, 3H). ESI m/z: 439.7 ([M+HCOO], C15H12Br2N20
requires 392.93)
Example 193. P7C3-S215: 2-(3,6-dibromo-9H-carbazol-9-y1)-N,N-dimethylacetamide
Br
Br to #
No
H3C,N_cH3
Synthesized analogously to P7C3-S214, except dimethylamine was used.
1H NMR (500 MHz, CDC13) 6 8.15 (s, 2H), 7.54 (d, J= 8.7 Hz, 2H), 7.19 (d, J=
8.7 Hz, 2H), 5.01 (s,
2H), 3.09 (s, 3H), 2.99 (s, 3H). ESI m/z: 452.6 ([M+HCOO], C16H14Br2N20
requires 406.95)
Example 194. P7C3-S217: 3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropan-1-
amine hydrochloride
Br
Br . 4.
N
__Cc HCI
NH2
The free base of P7C3-S217 was an intermediate in the synthesis of P7C3-5208.
The HC1 salt was
formed by adding 1M HC1 to a solution of the free base in CH2C12 at 0 C. A
white solid was collected by
filtration and washed with cold CH2C12.
Example 195. P7C3-S218: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
fluoropropyl)acetamide
Br
Br = fa
N
\........c:NH
-----
0
241
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropan-1-amine hydrochloride (P7C3-
S217) (25 mg, 0.057
mmol) was dissolved in dichloromethane (2 mL) with triethylamine (12 mg, 0.12
mmol). The solution was
cooled to 0 C and acetyl chloride (4.49 mg, 0.057 mmol) was added. Desired
product precipitated out of
solution and within 10 minutes the reaction had gone to completion. The
precipitate was filtered, rinsed with
H20, and dried under vacuum.
1H NMR (500 MHz, CDC13) 6 8.15 (d, J= 1.7 Hz, 2H), 7.57 (dd, J= 8.7, 1.8 Hz,
2H), 7.30 (d, J= 8.7
Hz, 2H), 5.75 (s, 1H), 4.98 (dddd, J= 13.1, 9.5, 6.6, 2.6 Hz, 1H), 4.58 -4.41
(m, 2H), 3.83 (dddd, J= 29.0,
14.6, 6.9, 2.6 Hz, 1H), 3.36 - 3.23 (m, 1H), 2.03 (s, 3H). ESI m/z: 440.7
([M+14] , C17H1513r2FN20 requires
440.95)
Example 196. P7C3-S219: N-(5-(34(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)amino)phenoxy)penty1)-5-(2-oxohexahydro-1H-thieno[3,4-d]imidazol-
4-yl)pentanamide
Br Br
0
OH
HNyNH
0
The title compound was synthesized analogously to P7C3-S100 using biotin N-
succinimidyl ester. The
mixture was chromatographed in 5-10% Me0H/CH2C12 Isolated yield = 37%.
MS (ESI), m/z: calculated 799.1, found 799.7 (M+1) .
Example 197. P7C3-S220: 2-(3,6-dibromo-9H-carbazol-9-yl)propanamide
Br
Br
H3C
NH2
The title compound was prepared analogously to P7C3-5213.
1H NMR (500 MHz, CDC13) 6 8.18 (s, 2H), 7.58 (d, J= 8.7 Hz, 2H), 7.30 (d, J=
8.7 Hz, 2H), 5.58 (s,
1H), 5.42 (s, 1H), 5.27 (q, J= 7.1 Hz, 1H), 1.74 (d, J= 7.1 Hz, 3H). ESI m/z:
394.7 ([M+H]+, C15H12Br2N20
requires 394.93)
Example 198. P7C3-S221: 6-bromo-9H-pyrido[3,4-b]indole-3-carbonitrile
242
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
CN
-
Br I. N
\ /
N
H
The title compound was an intermediate in the synthesis of P7C3-S192.
Example 199. P7C3-S226: 6-bromo-3-methy1-9H-pyrido[3,4-b]indole
CH3
_
Br 0 N
\ /
N
H
The title compound was an intermediate in the synthesis of P7C3-S190.
Example 200. P7C3-S233: 94(1H-tetrazol-5-yl)methyl)-3,6-dibromo-9H-carbazole
Br
Br is *
N H
N-N
Following a published procedure (J.O.C., 1993, 4139-4141), a solution of 2-
(3,6-dibromo-9H-
carbazol-9-yl)acetonitrile (P7C3-S235) (75 mg, 0.123 mmol) and
azidotrimethylsilane (32 ul, 0.24 mmol) in
toluene (500 uL) was added to dibutyltin oxide (3 mg, 0.012 mmol) and heated
to reflux overnight. The
reaction was cooled to room temperature, concentrated to remove toluene, and
partitioned between Et0Ac and
10% NaHC0300. The aqueous layers were combined and acidified to pH 2 and
extracted with Et0Ac (2x).
The organic layers were combined and washed with brine, dried over Na2SO4 and
concentrated. The crude
material was purified on Si02 (0-50% Et0Ac/hexanes.)
1H NMR (500 MHz, Acetone-d6) 6 8.43 (s, 2H), 7.69 (d, J= 8.7 Hz, 2H), 7.64 (d,
J= 8.6 Hz, 2H),
6.11 (s, 2H). ESI m/z:403.5 ([M-H], C14H9Br2N5 requires 403.92)
Example 201. P7C3-S234: methyl (2-(3,6-dibromo-9H-carbazol-9-
yl)acetyl)carbamate
Br
Br 0 446
N0
HN---f0
OMe
243
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following a published procedure (PCT Int. App: 2011054851), 2-(3,6-dibromo-9H-
carbazol-9-
yl)acetamide (P7C3-S213) (50 mg, 0.13 mmol) was dissolved in 1 ml of THF and
cooled to 0 C.
Methylchloroformate (12 uL, 0.157 mmol) was added followed slow addition of
LiOtBu (25 mg, 0.31 mmol)
in 1 ml of THF. Upon completion, the mixture was partitioned between 2 N HC1
and Et0Ac. Upon adding
Et0Ac, a precipitate formed which was filtered, rinsed with H20 and hexanes
and dried under reduced
pressure.
1H NMR (500 MHz, THF-d8) 6 12.71 (s, 1H), 10.14 (s, 2H), 9.36 (d, J= 8.7 Hz,
2H), 9.22 (d, J= 8.7
Hz, 2H), 7.45 (s, 2H), 5.66 (s, 3H). ESI m/z: 436.6 ([M-H], C16H12Br2N203
requires 436.92)
Example 202. P7C3-S241: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-
1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide
Br Br
1001 OMe
F k2CF3
Following General Procedure 4, P7C3-S244 was fluorinated to give the title
compound.
1H NMR (400 MHz, CDC13) 6 8.15 (d, J= 1.9 Hz, 2H), 7.56 (dd, J= 8.7, 1.9 Hz,
2H), 7.32 (t, J= 8.2
Hz, 1H), 7.21 (d, J= 8.6 Hz, 2H), 6.99 -6.90 (m, 2H), 6.86 (m, 1H), 5.08 -4.86
(dm, 1H), 4.57 - 4.44 (m,
2H), 4.09 (m, 2H), 3.79 (s, 3H). MS (ESI), m/z: calculated 635.93, found 680.6
(M+HC00-)-
Example 203. P7C3-S243: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-fluoropropy1)-6-
methoxypyridin-2-amine
Br Br
=
yNNOMe
2-(Dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-propy1-1,1'-biphenyl
(BrettPhos, 35.6 mg,
0.066 mmol) and chloro[2-(dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-
propy1-1,1'-biphenyl][2-(2-
aminoethyl)phenyl]palladium(II) (52.8 mg, 0.066 mmol) were charged to a vial
containing the free amine of
P7C3-5217 (116.8 mg, 0.29 mmol). The vial was purged with nitrogen for ten
minutes before addition of 1,4-
dioxane (4.75 ml) followed by the addition of lithium bis(trimethylsilyl)amide
solution (1.0 M solution in
THF, 610 I). The reaction was heated at 100 C for 5.5 hours. The mixture was
cooled and centrifuged to
separate out solids. The filtrate was loaded directly onto a silica gel column
and purified in 80% DCM/hexanes
(+0.1% TEA). The purest fractions (80-90% pure) were treated with DCM/hexanes
and the solid pellet
contained the purified product.
244
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
1H NMR (CDC13, 400 MHz) 6 8.16 (d, J= 1.9 Hz, 2H), 7.56 (d, J= 1.9 Hz, 2H),
7.35- (t, J= 7.8
Hz, 1H), 7.30 (d, J= 8.7 Hz, 1H), 6.04 (dd, J= 32.7, 8.0 Hz, 2H), 5.29 - 5.02
(dm, 1H), 4.65 -4.46 (m, 3H),
3.87 - 3.74 (m, 1H), 3.70 (s,3H), 3.66 - 3.49 (m, 1H). MS (ESI), m/z:
calculated 504.98, found 506.7 (M+1).
Example 204. P7C3-S244: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-
1,1,1-trifluoro-N-(3-
methoxyphenyl)methanesulfonamide
Step 1: 1,1,1-trifluoro-N-(3-methoxyphenyl)methanesulfonamide
HN ,...S02CF3
OMe
A solution of trifluoromethanesulfonic anhydride (45 ml, 26.7 mmol) in
methylene chloride (250 ml)
was added dropwise to an ice chilled solution of m-anisidine (25 ml, 22.3
mmol) and triethylamine (39 ml,
28.0 mmol) in methylene chloride (1.25 L). The reaction was stirred overnight
at ambient temperature.
Workup was performed portionwise. Each of the two portions was basified by
addition of 250 ml of 2.5 N
NaOH solution and 625 ml Me0H. The aqueous was extracted thrice (100 ml each)
with methylene chloride.
The combined aqueous phases was acidified to pH 2 with 18% HC1 and again
extracted with methylene
chloride three times. The organic layer is dried over Mg504, filtered and
condensed to give 17.69 g of brown
solid in 77% yield.
1H NMR (CDC13, 400 MHz) 67.48-7.13 (m, 1H), 6.97-6.61 (m, 3H), 3.82 (s, 3H).
MS (ESI), m/z:
calculated 255.21, found 255.9 (M+1) .
Step 2: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-1,1,1-trifluoro-N-
(3-
methoxyphenyl)methanesulfonamide (P7C3-S244).
Br Br
fk .
N
N 1.1 OMe
OH k2CF3
N-butyllithium (2.5 M in hexanes, 48 ml) was added dropwise to an ice-cooled
solution of 1,1,1-
trifluoro-N-(3-methoxyphenyl)methanesulfonamide (22.07 g, 86.5 mmol) in dry
dioxane (145 ml) over a 40
minute period. The solution was then stirred at ambient temperature for 15
minutes before addition of 3,6-
dibromo-9-(oxiran-2-ylmethyl)-9H-carbazole (25.05 g, 65.7 mmol) followed by
heating at 90 C for an hour.
The reaction was cooled then diluted with 1.2 L ethyl acetate and washed
several times with water and finally
brine. The organic layer was dried over Mg504, filtered and condensed to give
an orange viscous mixture. The
residue was dissolved in 150 ml of 60% methylene chloride/hexanes, then
concentrated to yellow foam to
which a further 150 ml of 60% methylene chloride/hexanes was added and stirred
overnight. The mixture was
245
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
filtered and washed several times with 60% methylene chloride/hexanes until
the solid was white giving 20.1 g
of 99%
1H NMR (CDC13, 400 MHz) 6 8.13 (d, J= 1.9 Hz, 2H), 7.54 (dd, J= 8.7, 1.9 Hz,
2H), 7.33 (t, J= 8.1
Hz, 1H), 7.22 (d, J= 8.7 Hz, 2H), 6.95 (dd, J= 8.4, 2.3 Hz, 2H), 6.88 (s, 1H),
4.56 -4.10 (m, 4H), 3.99 (m,
1H), 3.81 (s, 3H), 1.98 (d, J= 4.2 Hz, 1H). MS (ESI), m/z: calculated 633.94,
found 678.6 (M+HC00)- .
Example 205. P7C3-S255: 1-(3,6-dibromo-9H-carbazol-9-y1)-344-methoxybenzyl)(3-
methoxyphenyl)amino)propan-2-ol
Br Br
= .
N
YN SI OMe
OH
101
OMe
P7C3-5255 was synthesized analogously to P7C3-5244 using representative
procedure 3 and 3-
methoxy-N-(4-methoxybenzyl)aniline. Yield = 31%
1H NMR (CDC13, 400 MHz) 6 8.07 (d, J= 2.0 Hz, 2H), 7.50 (dd, J= 8.7, 2.0 Hz,
2H), 7.19 (d, J= 8.7
Hz, 2H), 7.12- 7.04 (m, 3H), 6.81 (d, J= 8.6 Hz, 2H), 6.36 (ddd, J= 12.7, 8.1,
2.4 Hz, 2H), 6.28 (t, J= 2.4
Hz, 1H), 4.48 (d, J= 2.7 Hz, 2H), 4.35 -4.27 (m, 1H), 4.28 -4.10 (m, 2H), 3.78
(s, 3H), 3.65 (s, 3H), 3.54 -
3.34 (m, 2H), 2.22 (s, 1H). MS (ESI), m/z: calculated 622.05, found 666.7
(M+HC00)- .
Example 206. P7C3-S261: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-hydroxypropy1)-
2,2,2-trifluoroacetamide
Br.
N
H HN __ <
Br O
CF3
TFAA (78 [LL, 0.5526 mmol) was added to a solution of 1-amino-3-(3,6-dibromo-
9H-carbazol-9-
yl)propan-2-ol (100 mg, 0.2518 mmol) and pyridine (61 [tt, 0.7536 mmol) in 1.7
mL DCM. After thirty
minutes, TLC showed complete consumption of starting material. The reaction
mixture was separated using
NaHCO3. The aqueous layer was washed with DCM. The combined organic layers
were dried and
concentrated to give crude material as a tan solid. Crude material was
dissolved in hot CHC13 and triturated
with hexanes. 50.6 mg (40.8% yield) of clean product was collected by vacuum
filtration.
246
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
1H NMR (500 MHz, CDC13) 6 8.16 (d, J= 1.7 Hz, 2H), 7.59 (dd, J= 8.7, 1.8 Hz,
2H), 7.31 (d, J= 8.7
Hz, 2H), 6.71 (s, 1H), 4.41 -4.28 (m, 2H), 3.79 (dd, J= 13.1, 6.7 Hz, 1H),
3.49 (s, 1H), 3.46 - 3.39 (m, 1H).
MS (ESI) m/z = 492.6 ([M+H]+, C17H13Br2F3N202 requires 491.93)
Example 207. P7C3-S263: tert-butyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)carbamate
Br.
N
101 \ 0
H HN
Br O
0
X
Boc anhydride (82.4 mg, 0.3777 mmol) was added to a solution of 1-amino-3-(3,6-
dibromo-9H-
carbazol-9-yl)propan-2-ol (100 mg, 0.2518 mmol) and triethylamine (70 L, .5036
mmol) in 1.7 mL DCM.
The reaction stirred overnight at room temperature. The reaction mixture was
separated with brine and the
aqueous layer washed twice with DCM. The combined organic layers were dried
and concentrated to afford
crude material as an off-white solid. This was subjected to column
chromatography using DCM/Me0H to
afford 4.6 mg (3.7% yield) product as a white solid.
1H NMR (500 MHz, CDC13) 6 8.14 (d, J= 1.9 Hz, 3H), 7.56 (dd, J= 8.6, 1.9 Hz,
3H), 7.34 (d, J= 8.7
Hz, 3H), 4.86 (s, 1H), 4.31 (d, J= 6.2 Hz, 3H), 3.40 - 3.25 (m, 1H), 3.09 -
2.97 (m, 1H), 1.45 (s, 13H), 3.25 -
3.09 (m, 2H). MS (ESI) m/z = 440.7 ([M-Boc]+, C20H22Br2N203 requires 496.00
Example 208. P7C3-S271: 5-(2-hydroxy-3-phenoxypropy1)-5H-pyrimido[5,4-b]indole-
2-carboxylic acid
CO2H
N--:--(
N
110 \ 1
N
OH
\-----0
=
Following Representative Procedure 7, the title compound was synthesized from
P7C3-5262 (0.028 g,
0.117 mmol) and 2-(phenoxymethyl)oxirane (24 [tL, 0.175 mmol) (0.030 g, 70%).
1H NMR (CDC13-Me0D [4:2], 500 MHz) 6 9.02 (s, 1H), 8.35 (d, 1H, J= 7.9 Hz),
7.49 (d, 2H, J= 3.7
Hz), 7.22 (dt, 1H, J= 4.0, 7.9 Hz), 7.08 (t, 2H, J= 8.0 Hz), 6.77 (t, 1H, J=
7.4 Hz), 6.72 (d, 2H, J= 8.0 Hz),
4.59 (dd, 1H, J= 4.4, 15.1 Hz), 4.45 (dd, 1H, J= 6.5, 15.1 Hz), 4.25 (me, 1H),
3.81 (dd, 1H, J= 4.4, 9.5 Hz),
3.76 (dd, 1H, J= 6.5, 9.5 Hz). MS (ESI) m/z 363.9 [M+H]+ ([M+H]+, C20H18N304
requires 364.4).
247
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
Example 209. P7C3-S273: N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)acetamide
Br.
N __ \
104 2 __ \ ____ 0
Br HO
A solution of acetic anhydride (29 [LL, 0.3022 mmol) in 0.7 mL DCM was added
to a solutio of 1-
amino-3-(3,6-dibromo-9H-carbazol-9-yl)propan-2-ol (100 mg, 0.2518 mmol) and
triethylamine (42 [LL,
0.3022 mmol) ml mL DCM at 0 C. After stirring overnight, the reaction mixture
was diluted with DCM, then
washed with 18% HC1, brine, and NaHCO3. The organic layer was dried and
concentrated to give crude
material as a pink-tinted solid. Preparatory TLC using 5% Me0H/DCM as the
eluent resulted in 6.5 mg (5.9%
yield) product as a white solid.
1H NMR (500 MHz, CDC13) 6 8.15 (d, J= 2.5 Hz, 2H), 7.59 - 7.54 (m, 2H), 7.34
(dd, J= 8.9, 4.2 Hz,
2H), 5.77 (s, 1H), 4.31 (q, J= 5.5, 4.3 Hz, 2H), 4.26 (s, 2H), 3.57 (d, J= 3.8
Hz, 1H), 3.46- 3.26 (m, 3H),
2.01 (d, J= 4.3 Hz, 3H). MS (ESI) m/z = 438.7 ([M+1-1] , C17H16Br2N202
requires 437.96
Example 210. P7C3-S274: ethyl (3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)carbamate
Br.
N
lip \ 0
H HN
Br O
0
(
The title compound was prepared analogously to P7C3-5218. The crude material
was
chromatographed using 5% Me0H/DCM as the eluent. 24.5 mg (20.7% yield) product
was obtained as a white
solid.
1H NMR (500 MHz, CDC13) 6 8.15 (s, 2H), 7.56 (d, J= 8.6 Hz, 2H), 7.33 (dd, J=
8.6, 1.6 Hz, 2H),
4.98 (s, 1H), 4.31 (d, 2H), 4.26 (s, 1H), 4.14 (q, J= 7.2 Hz, 2H), 3.45 -3.36
(m, 1H), 3.29 -3.17 (m, 1H),
3.01 (s, 1H), 1.26 (t, J= 7.9, 3.7 Hz, 3H). MS (ESI) m/z = 468.7 ([M+1-1] ,
C18H18Br2N203 requires 467.97
Example 211. P7C3-S278: 6-bromo-9-(3-(4-bromophenoxy)-2-hydroxypropy1)-9H-
carbazole-3-carbonitrile
Step 1: 9H-carbazole-3-carbonitrile
248
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
CN
* =
N
H
To a solution of 3-bromo-9H-carbazole (4.00 g, 16.3 mmol, 1 equiv) in N-Methyl-
2-pyrrolidinone (40
mL) was added copper(I) cyanide (1.6012 g, 17.9 mmol, 1.1 equiv). The mixture
was sealed and heated at 200
C until TLC showed no starting material. The reaction solution was cooled and
60 mL of water was added.
The off-white precipitate was filtered off and washed with Et0Ac (3 x 20 mL).
This filtrate was extracted with
ethyl acetate (3 x 20 mL). The combined organic layers were washed with water
(20mL) and brine (20mL),
and then dried over Na2SO4. The solvent was removed under reduced pressure to
afford red-brown oil.
Methanol was added to the crude oil to afford 0.8251 g off-white precipitate
as pure product. The mother
liquid from the methanol precipitation was concentrated and purified by
chromatography (25% Et0Ac/Hex) to
provide 0.28 g off-white solid. The combined yield was 35.5%.
1H NMR (CD30D, 400 MHz) 6ppm 7.26 (ddd, J= 8.0, 6.9, 1.3 Hz, 1H) 7.54 - 7.45
(m, 2H) 7.57 (dd,
J= 8.4, 0.6 Hz, 1H) 7.67 (dd, J= 8.5, 1.6 Hz, 1H) 8.18- 8.12 (m, 1H) 8.49 (dd,
J= 1.5, 0.6 Hz, 1H). MS
(ESI) m/z = 192.9 ([M+H]+), C131-18N2 requires 192.07
Step 2: 9-(2-hydroxy-3-phenoxypropy1)-9H-carbazole-3-carbonitrile
NC * 0
N 0
.OH
To a solution of 3-cyano-9H-carbazole (0.4998 g, 2.6 mmol, 1 equiv) in THF
(40mL) was added
"BuLi (1.6 M in hexane, 3.25 mL, 5.2 mmol, 2 equiv). The mixture was stirred
at -78 C for 2 hours. 1,2-
epoxy-3-phenoxypropane (0.7809 g, 5.2 mmol, 2 equiv) was added and the mixture
was stirred at 45 C
overnight. Water (50 mL) was added and the mixture was extracted by
dichloromethane (3 x 30 mL). The
combined dichloromethane layers were washed by brine (2 x 30 mL) and then
dried over Na2504. The solvent
was removed under reduced pressure, and the product was chromatographed on
silica gel with
dichloromethane to yield 0.7736 g white foamed solid (86%).
1H NMR (CD30D, 400 MHz) 6ppm 3.95 (dd, J= 9.8, 4.6 Hz, 1H) 4.02 (dd, J= 9.8,
5.4 Hz, 1H) 4.46
-4.35 (m, 1H) 4.57 (dd, J= 15.0, 6.9 Hz, 1H) 4.70 (dd, J= 15.2, 5.0 Hz, 1H)
7.00 - 6.88 (m, 3H) 7.34 - 7.21
(m, 3H) 7.49 (t, J= 7.8 Hz, 1H) 7.65 (d, J= 8.3 Hz, 2H) 7.72 (d, J= 8.5 Hz,
1H) 8.17 (d, J= 7.8 Hz, 1H) 8.49
(s, 1H). MS (ESI) m/z = 342.9 ([M+H]+), 364.9 ([M+Na]), C22H18N202 requires
342.14
Step 3: 6-bromo-9-(3-(4-bromophenoxy)-2-hydroxypropy1)-9H-carbazole-3-
carbonitrile (P7C3-5278)
249
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
NC
Ai Br
la
411r7 N 0
. OH
Br
To a solution of the product of Step 2 (0.7736 g, 2.26 mmol, 1 equiv) in THF
(8mL) was added N-
Bromosuccinimide (0.8043 g, 4.52 mmol, 2 equiv). The mixture was stirred at
room temperature for 30
minutes. THF was removed on the vacuum and the crude residue was purified by
silica gel chromatography
(30% ethyl acetate in hexanes) afford 1.0230 g white solid as product, yield
90.5%.
1H NMR (CDC13, 400 MHz) 6ppm 3.88 (dd, J= 9.5, 4.7 Hz, 1H) 3.99 (dd, J= 9.5,
4.7 Hz, 1H) 4.69 -
4.38 (m, 3H) 6.81 - 6.70 (m, 2H) 7.44 - 7.35 (m, 3H) 7.54 (t, J= 6.5 Hz, 1H)
7.59 (dd, J= 8.7, 1.9 Hz, 1H)
7.66 (d, J = 8.6 Hz, 1H) 8.20 (d, J = 1.3 Hz, 1H) 8.31 (s, 1H). MS (ESI) m/z =
498.7 ([M+H]+),
C22H19Br2N302 requires 497.96
Example 212. P7C3-S279: methyl 9-(2-hydroxy-3-phenoxypropy1)-9H-pyrido [3 ,4-
1)] indo le-3 -c arb oxylate
0 N_
Me
.OH
To a solution of P7C3-5172 (10.9 mg, 0.03 mmol, 1 equiv) in DMF (0.5 mL) was
added iodomethane
(6.4 mg, 0.045 mmol, 1.5 equiv) and potassium carbonate (12.4 mg, 0.09 mmol, 3
equiv). The mixture was
sealed and stirred in a 4-mL vial at room temperature for overnight. The crude
reaction was diluted with 3 mL
of dichloromethane and washed with brine (3 x 3 mL). The organic layers were
dried over Na2504 and
dichloromethane was removed under vacuum to yield yellow crude oil. It was
further purified by silica gel
chromatography using 10% of dichloromethane in methanol as elute to afford 8.6
mg white solid as product,
yield 82.0%.
1H NMR (CDC13, 400 MHz) 6ppm 3.64 (s, 3H) 4.14 (ddd, J = 15.8, 9.3, 5.6 Hz,
2H) 4.58 (dd, J =
14.6, 7.3 Hz, 1H) 4.68 (d, J= 4.4 Hz, 1H) 4.76 (dd, J= 14.6, 3.3 Hz, 1H) 6.98
(dd, J= 13.9, 7.6 Hz, 3H) 7.31
(dd, J= 8.5, 7.5 Hz, 2H) 7.36 (td, J= 7.5, 3.4 Hz, 1H) 7.62 (d, J= 3.7 Hz, 2H)
8.14 (d, J= 7.8 Hz, 1H) 8.57 (s,
1H) 9.00 (s, 1H). MS (ESI) m/z = 377.1 ([M+H]+), C22H20N204 requires 376.14
Example 213. P7C3-S282: 1 -(3 ,6-dimethoxy-9H-c arb azol-9-y1)-3 -phenoxyprop
an-2 -ol
250
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
OMe
Me0 t dii 4 41,
N OH
----C--0
o
Following Representative Procedure 7, the title compound was synthesized from
3,6-dimethoxy-9H-
carbazole (Hsieh, B. B.; Litt, M. H. Macromolecules, 1986, 19, 516-520) (0.030
g, 0.0917 mmol) and 2-
(phenoxymethyl)oxirane (19 [tt, 0.128 mmol) in dry THF:DMF (1:1) (0.459 mL)
(0.028 g, 81%).
1H NMR (CDC13, 400 MHz) 6 7.51 (s, 2H), 7.35 (d, 2H, J= 8.6 Hz), 7.29 (d, 2H,
J= 8.6 Hz), 7.05
(dd, 2H, J= 2.4, 8.8 Hz), 6.98 (t, 1H, J= 7.4 Hz), 6.88 (d, 2H, J= 7.9 Hz),
4.52 (me, 1H), 4.46 (me, 2H), 4.00-
3.85 (m, 8H). MS (ESI) m/z 378.2 [M+1-1] ([M+14] , C23H24N04 requires 378.4).
Example 214. P7C3-S283: 2-chloro-N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)acetamide
Br
Br. .
N OH
\-------NH
----/CI
0
P7C3-538 (0.134 g, 0.337 mmol) was dissolved in CH2C12/MeCN (9:1, 5.6 mL) and
treated with Et3N
(70 [tL, 0.50 mmol) at 0 C. Chloroacetyl chloride (29 [tt, 0.370 mmol) was
then added dropwise. After being
stirred at room temperature for 45 min, the reaction was quenched with a
solution of 1N HC1. The organic
phase was separated, washed with brine, dried over Na2504, filtered and
condensed. The crude mixture was
purified by chromatography (5i02, 0-80% Et0Ac/Hexanes) to afford the title
compound (0.091 g, 60%).
1H NMR (acetone-d6, 400 MHz) 6 8.36 (s, 2H), 7.74-7.51 (m, 4H), 4.65 (d, 1H, J
= 4.7 Hz), 4.49 (dd,
1H, J= 3.4, 15.0 Hz), 4.41 (dd, 1H, J= 8.3, 15.0 Hz), 4.29 (me, 1H), 4.13 (s,
2H), 3.63-3.50 (m, 1H), 3.43 (dt,
1H, J= 5.7, 12.8 Hz). MS (ESI) m/z 474.6 [M+1-1] ([M+14] , C17H16Br2C1N202
requires 475.5).
Example 215. P7C3-S284: 2-chloro-N-(3-(3,6-dibromo-9H-carbazol-9-y1)-2-
hydroxypropyl)acetamide
Br
Br is .
NO
NH
251
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Following a reported procedure (Metro, T. X.; Pardo, D. G.; Cossy, J. J. Org.
Chem. 2008, 73, 707-
710), P7C3-S283 (0.091 g, 0.192 mmol) was dissolved in iPrOH (7.7 mL) and
treated with tBuOK (0.054 g,
0.48 mmol) in iPrOH (1.9 mL) at 0 C. The mixture was stirred at room
temperature for 4 h. The reaction was
then quenched with 1N HC1 and concentrated in vacuum. The residue was diluted
with water a then extracted
with Et0Ac. The organic layers were washed with brine, dried over Na2SO4,
filtered and condensed. The
crude mixture was purified by chromatography (Si02, Et0Ac) to afford the title
compound (0.060 g, 72%).
1H NMR (d6-DMSO, 400 MHz) 6 8.47 (d, 2H, J= 1.9 Hz), 7.67 (d, 2H, J= 8.7 Hz),
7.60 (dd, 2H, J=
1.9, 8.7 Hz), 4.55 (d, 2H, J= 5.5 Hz), 4.10 (me, 1H), 3.98-3.77 (m, 2H), 3.18
(t, 2H, J= 11.3 Hz). MS (ESI)
m/z 438.6 [M+H]+ ([M+H]+, C17H14Br2N202 requires 439.1), 460.6 [M+Na]+
([M+Na]+, C17H13Br2N2Na02
requires 460.1).
Example 216. P7C3-S287: 2-(3-bromo-6-methy1-9H-carbazol-9-y1)-N-
(phenylsulfonyl)acetamide
0 H
N---\___N_ =
ip, O 6
Br
The title compound was prepared analogously to P7C3-5232. Column
chromatography(1:4
THF:Hexanes) resulted in collection of 86.7 mg (35.5% yield) product as a
white solid.
1H NMR (400 MHz, Acetone-d6) 6 8.23 (d, J= 1.9 Hz, 1H), 7.97 (d, J= 1.1 Hz,
1H), 7.95 (d, J= 1.5
Hz, 1H), 7.94 (s, 1H), 7.65 ¨ 7.60 (m, 1H), 7.56 ¨ 7.50 (m, 2H), 7.45 (dd, J=
8.7, 2.0 Hz, 1H), 7.34 ¨ 7.28 (m,
1H), 7.26 (d, J= 0.7 Hz, 1H), 7.24 (dd, J= 8.4, 1.6 Hz, 1H), 5.12 (s, 2H),
2.47 (s, 3H). MS (ESI) m/z = 457.0
([M+H]+, C21H17BrN203S requires 456.01.)
Example 217. P7C3-S288: 2-(3-bromo-6-methy1-9H-carbazol-9-yl)acetamide
0 õ,
IN-)rNH2
lip 0
Br
The title compound was prepared analogously to P7C3-5213.
1H NMR (400 MHz, CDC13) 6 8.19 (d, J= 1.9 Hz, 1H), 7.86 (s, 1H), 7.57 (dd, J=
8.6, 1.9 Hz, 1H),
7.36 (dd, J= 8.3, 1.6 Hz, 1H), 7.28 (d, J= 8.4 Hz, 1H), 7.24 (s, 1H), 5.34 (d,
J= 31.8 Hz, 2H), 4.88 (s, 2H),
2.54 (s, 3H). MS (ESI) m/z = 317.0 ([M+H]+, C15H13BrN20 requires 316.02)
Example 218. P7C3-S294: 2-(3-bromo-6-methy1-9H-carbazol-9-yl)ethanol
252
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
0 õ,......,
11 \--OH
IP
Br
The title compound was prepared analogously to P7C3-S171.
1H NMR (400 MHz, CDC13) 6 8.17 (d, J= 1.9 Hz, 1H), 7.84 (s, 1H), 7.52 (dd, J=
8.6, 2.0 Hz, 1H),
7.38 -7.28 (m, 3H), 4.44 (t, J= 5.4 Hz, 2H), 4.05 (q, J= 5.5 Hz, 2H), 2.53 (s,
3H), 1.47 (t, J= 6.0 Hz, 1H).
MS (APCI) m/z = 304.0 ([M+1-1] , C15H14BrNO requires 303.03).
Example 219. P7C3-S295: N-(3-(3-bromo-6-methy1-9H-carbazol-9-y1)-2-
fluoropropy1)-6-methoxypyridin-2-
amine
Br
4* *
N
1
N N OMe
H
F
A vial containing S286 (50.5 mg, 0.13 mmol), 2-iodo-6-methoxypyridine (29.6
mg, 0.13 mmol),
chloro[2-(dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-propy1-1,1'-
biphenyl][2-(2-
aminoethyl)phenyl]palladium(II) (BrettPhos palladacycle, 10.8 mg, 0.014 mmol)
and 2-
(dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-propy1-1,1'-biphenyl
(BrettPhos, 6.8 mg, 0.013 mmol)
was purged with nitrogen for 20 minutes before the addition of dioxane (2.45
ml) followed by the dropwise
addition of LHMDS (1.0 M in THF, 0.5 mmol). The reaction was stirred for an
hour before being centrifuged.
The supernatant was chromoatographed on silica gel in 10-30% THF/hexanes.
Yield = 25%
1H NMR (400 MHz, (CD3)2C0) 6 8.27 (dd, J= 1.7, 0.9 Hz, 1H), 7.98 (dt, J= 1.9,
0.9 Hz, 1H), 7.53
(t, J= 1.4 Hz, 1H), 7.49 (d, J= 8.5 Hz, 1H), 7.35 - 7.26 (m, 2H), 6.92 (s,
1H), 6.14 (d, J= 8.0 Hz, 1H), 5.91
(dd, J= 7.8, 0.7 Hz, 1H), 5.33 - 5.04 (m, 1H), 4.77 (dd, J= 5.5, 3.8 Hz, 1H),
4.71 (d, J= 5.5 Hz, 1H), 3.86 -
3.76 (m, 1H), 3.74 -3.64 (m, 1H), 3.60 (s, 3H), 2.49 (s, 3H). MS (ESI), m/z:
calculated 441.09, found 442.0
(M+1).
Additional compounds of the presently disclosed embodiments can also be
synthesized via similar
schemes and methods as described above.
Pro-neurogenic Efficacy! Neuroprotection Activity of Various Compounds:
Compounds were tested in vivo for dose-responsive neurotrophic efficacy. The
results are shown in
Table 1.
253
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Table 1. In Vivo Activity
Vehicle 14.5 1.08
FGF-2: (fibroblast growth
factor 2) 28.4 2.12
Example la 29.8 2.0
((S)-P7C3-0Me)
Example lb 18.3 0.8
((R)-P7C3-0Me)
Example 2 24.4 1.4
Example 3a 30.9 3
Example 3b 29.6 1.3
Example 3c 16.1 1.74
Example 3d 27.1 1.34
Example 4 23.7 0.6
Example 5 21.5 2.18
Example 6a (P7C3A20) 38 2.4
Example 6b 25.5 (one
animal tested)
Example 7a 18.4 1.8
Example 7b 23.4 1.31
Example 8 23.2 0.8
Example 9 16.2 1.7
Example 10 27 1.3
Example 11 15.1 0.6
Example 12 21.7 2.9
Example 13 28.5 2.6
Example 14 17.8 1.9
Example 15 15.1 0.9
Example 16 17.1 0.9
Example 17 20.8 0.3
Example 19 15 0.5
Example 20 23.2 0.48
Example 21 27.6 3.4
Example 22 27.3 1.8
Example 23 21.5 2.2
Example 25 16.8 1.3
Example 26 15.6 1
Example 28 21 0.6
Example 29 17.6 2.3
Example 30 13.4 1.2
Example 31 14.7 1
Example 32 16 0.4
Example 33 14 0.2
Example 36 19 2.54
Example 39 23.4 1.1
Example 40 14.4 1.5
Example 41 16 1.1
Example 43 21.3 2.6
Example 45 (P7C3) 30 1.42
254
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Ptest Matenal ..---.. 410 U+
tells I min SEM. Otandard error of the
Example 76 (P7C3-S229) 26.5 3.7
Example 87 25.4 2.4
Example 88a 16.2 1
Example 88b 30.6 3.66
Example 89 23.4 0.26
Example 90 33.3 3.3
Example 91 18.3 2.9
Example 92 29 1.6
Example 93 20.1 2.5
Example 94 23.9 2.43
Example 95 21.5 1.2
Example 96 34.2 4.29
Example 97a 32.4 3.84
Example 97b 26.3 1.55
Example 101 25.8 2.6
Example 102 27.6 2.7
Example 103 16.8 1.13
Example 104 25.1 2
Example 105
P7C3-S67 17.7 1.4
Example 107 19.3 1.4
Example 108
P7C3-S68 14.6 0.84
Example 109 23.7 0.75
Example 110
P7C3-S70 14.7 0.6
Example 111
P7C3-S71 14.3 1.5
Example 112
P7C3-S72 23.3 2.2
Example 113
P7C3-S73 20.8 1.5
Example 114
P7C3-S75 20.6 3.5
Example 115
P7C3-S77 24 1.5
Example 116
P7C3-S78 28.1 1.71
Example 117
P7C3-S79 27.3 2.17
Example 118
P7C3-S80 25.9 1.1
Example 119
P7C3-S81 25.1 1.8
Example 120
P7C3-S82 23.6 0.74
Example 121
P7C3-S83 24.9 0.8
255
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
test .M OH
agaggaggagg211000#0111gymv
Example 122
P7C3-S84 25.6 1.4
Example 123
P7C3-S91 16.3 1.1
Example 124
P7C3-S92 16.8 2
Example 126
P7C3-S94 16.9 1.4
Example 127
P7C3-S95 17.2 0.9
Example 128
P7C3-S96 17.4 0.9
Example 129
P7C3-S97 15.1 1.6
Example 130
P7C3-S98 13.8 1.8
Example 131
P7C3-S99 15.2 0.9
Example 132
P7C3-S100 24 0.6
Example 133
P7C3-S101 19.8 1.4
Example 134
P7C3-S102 17.7 1.6
Example 135
P7C3-S103 13.9 0.8
Example 137
P7C3-S105 21.6 1.4
Example 138
P7C3-S106 21.7 0.8
Example 139
P7C3-S107 14.6 0.5
Example 140
P7C3-S108 15.2 0.4
Example 141
P7C4-S109 18.8 1.7
Example 142
P7C3-S110 21 1.2
Example 143
P7C3-S111 24.5 2.2
Example 144a
P7C3-S113 31.5 2
Example 144b
P7C3-S114 15.2 1.3
Example 145
P7C3-S115 13.2 2.1
Example 148
P7C3-S131 17.9 1.5
256
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
test *M OH 4171!!!!!!!!!!!!iliiiin110110.
agaggaggamail000tWiligyrgv
Example 150
P7C3-S138 20.8 4
Example 151
P7C3-S141 22.5 2.3
Example 152
P7C3-S142 26.4 3.3
Example 153
P7C3-S146 29 2.6
Example 154
P7C3-S147 26.5 1.1
Example 155
P7C3-S150 16.7 1.4
Example 156
P7C3-S151 29.8 2
Example 157
P7C3-S153 21.4 1.8
Example 158a
P7C3-S154 19.7 2.5
Example 158b
P7C3-S155 26.1 3.7
Example 159
P7C3-S157 20.3 0.4
Example 160
P7C3-S159 13.8 1.5
Example 161
P7C3-S160 26.2 3
Example 162
P7C3-S161 22.7 2.4
Example 163
P7C3-S164 15.2 1.8
Example 164
P7C3-S165 35.7 4.7
Example 165
P7C3-S166 35.6 2.4
Example 166
P7C3-S167 27.2 2.6
Example 167
P7C3-S168 29.1 1.1
Example 168
P7C3-S172 28.3 2.3
Example 169
P7C3-S173 25.5 3
Example 170
P7C3-S174 18.6 1.5
Example 171
P7C3-S175 24.6 2.6
Example 172
P7C3-S176 13.8 1
257
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
test M4 OH
agaggaggagg21100#0tOiigyrgv
Example 173
P7C3-S177 27.7 1.80
Example 174
P7C3-S178 27.5 2.3
Example 175
P7C3-S179 34.5 2.3
Example 177
P7C3-S183 15.4 1
Example 178
P7C3-S184 13.4 1.7
Example 179
P7C3-S186 16.1 1.3
Example 180
P7C3-S187 25.2 2.3
Example 181
P7C3-S188 26.1 1.3
Example 182
P7C3-S190 16.7 0.3
Example 183
P7C3-S191 21.4 0.8
Example 184
P7C3-S192 21 2.1
Example 185
P7C3-S194 16.2 1.2
Example 186
P7C3-S195 17.9 1.9
Example 187
P7C3-S198 22 1.0
Example 188
P7C3-S204 28.4 1.5
Example 189
P7C3-S205 29.1 2.7
Example 190
P7C3-S208 27 2.6
Example 191
P7C3-S213 13 0.7
Example 192
P7C3-S214 19.7 2
Example 193
P7C3-S215 16.6 0.9
Example 194
P7C3-S217 24.6 2.9
Example 195
P7C3-S218 18.9 1.6
Example 196
P7C3-S219 16.8 2.8
Example 197
P7C3-S220 21 2.5
258
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Example 198
P7C3-S221 29.5 2
Example 199
P7C3-S226 25.6 2
Example 200
P7C3-S233 14.5 0.7
Example 201
P7C3-S234 35.6 1.3
Example 202
P7C3-S241 30.9 1.5
Example 203
P7C3-S243 35.7 3.8
Example 204
P7C3-S244 23.2 1.9
Example 205
P7C3-S255 35.3 2.1
Example 206
P7C3-S261 19.3 2
Example 207
P7C3-S263 24.1 3.3
Example 208
P7C3-S271 34.6 1.7
Example 209
P7C3-S273 22.8 2.4
Example 210
P7C3-S274 25.7 2
Example 211
P7C3-S278 36.2 3.3
Example 212
P7C3-S279 25 0.9
Compounds were evaluated for pro-neurogenic efficacy / neuroprotection in our
standard in vivo assay
at 10 [tM concentration in four 12 week old adult male C57/B16 mice.
The (+) (dextrorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-propan-2-ol as described herein exhibited higher
activity.
The (-) (levorotatory) enantiomer of 1-(3,6-Dibromo-9H-carbazol-9-y1)-3-(3-
methoxyphenylamino)-
propan-2-ol as described herein exhibited lower activity.
Previously, a commercial sample of P7C3-S229 was tested in the neurogenesis
assay and found to be
indistinguishable from vehicle when dosed intracranially as a 10 micromolar
solution (MacMillan etal., J. Am.
Chem. Soc. ,2011; (133):1428-1437). Subsequently, a separate sample was
independently synthesized and
tested analogously, and found to be active. While there may be many reasons
for this discrepancy, without
wishing to be bound by theory, it has been hypothesized that possible
explanations include: 1) The identity of
the commercial sample was not rigorously confirmed; it may have been a
different substance. 2) The purity of
259
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
the commercial sample was not rigorously confirmed; an impurity may have
counteracted the pro-neurogenic
activity of the commercial sample. 3) P7C3-S229 displays low solubility.
Slight variation in the formulation of
different samples or the physical characteristics of the sample (e.g.
crystalline vs. amorphous solid) may have
resulted in differences in exposure in the in vivo experiment.
It is also believed that P7C3-5295 (Example 219), based on the structure, can
have high biological
activity (e.g., in the pro-neurogenic efficacy / neuroprotection assay in mice
models described herein).
Additional experiments (in vitro and in vivo) are underway to further
characterize the effect of P7C3-5295 in
disease models described herein.
Identification of pro-neurogenic or neuroprotective compounds:
In an effort to identify compounds that might stimulate the birth of new
neurons, or protect newborn
neurons from cell death, a library of 1,000 compounds was screened using an in
vivo assay. In the initial
screen, compounds were randomly pooled into groups of ten and administered
intracerebroventricularly at a
constant rate over seven days into the left lateral ventricle of living mice
via Alzet osmotic mini-pumps.
Compounds were administered at a concentration of 10[LM for each molecule,
making a total solute
concentration of 100[LM. After seven days of infusion at a constant rate of
0.5Whour, a total of 844, of
volume will have left the pump (0.00084 Moles) and entered the cerebrospinal
fluid. The average volume of
a brain from a 12 week old male, C57/B6 mouse in our study is 500mm3. The
maximal amount of drug was
estimated that could potentially be present in the brain, taking the extreme
and unlikely scenario of 100%
absorbance of the drug into brain tissue and 0% clearance throughout the seven
day infusion period. Under
these conditions, at the end of one week of infusion each compound would be
present at 1.7 Molar
concentration. Since the actual amount of chemical compound in the brain is
likely to be only a fraction of
this predicted level, it is reasonable to estimate that compounds were
administered at mid to low-nanomolar
concentrations.
During compound infusion, animals were intraperitoneally (IP) injected daily
with the thymidine
analog, bromodeoxyuridine (BrdU), as a means of scoring the birth and survival
of proliferating neural
precursor cells in the hippocampus. Because both social interaction and
voluntary exercise are known to
stimulate hippocampal neurogenesis, mice were housed individually without
access to running wheels
throughout the screening period. Following the week-long period of compound
administration, animals were
perfused and sacrificed. Dissected brain tissue was fixed, embedded,
sectioned, stained with antibodies to
BrdU, and evaluated by light microcopy as a means of quantifying neurogenesis
and survival of newborn
neural precursor cells localized to the subgranular layer of the dentate gyrus
on the brain hemisphere
contralateral to the side of mini-pump cannulation. Every fifth section
throughout the entire rostral-caudal
extent of the hippocampus was analyzed, and the total number of BrdU+ cells
was normalized against the
measured volume of the dentate gyms. Because both increased proliferation and
survival of newborn neurons
260
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
are important screening parameters, the screen was conducted over seven days
in order to cast a wide net to
detect molecules that might augment either process. The choice of parameters
for the screen was based on
pulse-chase experiments with a single injection of BrdU, under identical
conditions to those used in our screen,
which revealed that 40% of newborn cells in the dentate gyms die within the
first five days of their birth
(Figure 1). Intracranial infusions of either fibroblast growth factor 2 (FGF-
2) or artificial cerebral spinal fluid
(aCSF) vehicle via the same, week-long protocol were employed as positive and
negative controls. There was
no difference in the number of BrdU-labeled cells in the dentate gyms between
mice subjected to surgical
pump implantation and infusion with vehicle, and mice having had no surgery
(Figure 2). This confirmed the
validity of the in vivo approach to assess the ability of
intracerebroventricularly infused compounds to enhance
hippocampal neurogenesis in the contralateral hemisphere.
Considered to be important is that stimulation of neurogenesis triggered by
any compound be localized
to the exact region of the brain known to produce new neurons at an enhanced
level in response to healthy
activities such as wheel running, access to an enriched environment, or access
to social interaction. For this
reason attention was focused solely on compound pools that stimulated BrdU
incorporation only in the
subgranular zone of the dentate gyms. Prominent nonspecific incorporation of
BrdU in ectopic regions, such
as CA3, CA1, cortex, or striatum, was presumed to reflect pathological
inflammation, as proliferating cells
incorporate BrdU in DNA synthesis, or to indicate other forms of toxicity, as
cells also incorporate BrdU
during DNA repair. Any compound pools yielding ectopic BrdU incorporation were
eliminated from the
screen. For an example, see Figure 3.
Each of the 100 pools was tested on two independent mice. As shown in Figure
4, ten of the 100 test
pools were observed to enhance dentate gyms-specific neurogenesis to an extent
roughly equivalent to FGF-2.
Each pool that scored positive in the initial two test animals was
subsequently re-evaluated in two additional
mice, and all ten pools were found to exert their pro-neurogenic effect with
statistical significance (Figure 5).
In order to identify single, pro-neurogenic compounds, positive pools were
broken down into their ten
component molecules, each of which was infused individually at two
concentrations (10[LM and 100[LM) in
two mice per concentration. Figure 6A shows the results of break-down assays
on pool #7, wherein it was
discovered that neurogenesis was selectively stimulated by one of the
constituent chemicals of the pool
(compound #3), chemicals in the pool demonstrating no effect. This molecule
was designated as Example 45
Compound or P7C3. In breaking down the ten positive pools, eight pools yielded
a single pro-neurogenic
compound (Figure 6B). To ensure that the pro-proliferative or neuroprotective
effect on neural stem cells was
not an artifact of storage conditions in the UTSWMC chemical compound library,
re-supplied compounds
were verified to by 99% pure by mass spectrometry, evaluated in 4 mice each at
10 ,M concentration, and
shown to retain either pro-proliferative or neuroprotective properties in
neural stem cells (Figure 6C).
Pharmacokinetic analysis of Example 45 Compound in plasma and whole brain
tissue was undertaken
after single IV, IP and oral gavage administrations. Example 45 Compound was
noted to be orally
261
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
bioavailable, readily able to cross the blood-brain barrier, and endowed with
a plasma terminal half life of 6.7
hours after IP delivery. These favorable pharmacological properties
facilitated a dose response experiment
wherein daily oral administration of Example 45 Compound to adult mice was
monitored for both brain levels
of the chemical and pro-neurogenic efficacy (Figure 7). Maximal, pro-
neurogenic efficacy was observed at
oral doses of 5mg/kg and above, and graded reductions in efficacy were
observed at doses of 2.5 and lmg/kg.
Liquid chromatography-mass spectrometry analysis of the brain levels of
Example 45 Compound in the dose
ranges of 1, 2.5 and 5mg/kg revealed corresponding compound concentrations of
213 nM (101ng/g brain
tissue), 1.13 [tM (534ng/g brain tissue) and 1.35 [tM (640ng/g brain tissue)
five hours after dosing.
Enantiomer Selective Activity of Example 45 Compound Derivatives:
In order to further study Example 45 Compound, an in vivo structure activity
relationship (SAR) study
was conducted using 37 chemical derivatives of the compound for pro-neurogenic
activity via direct
administration into the brain of adult mice via Alzet minipumps. Compounds
were administered for one week
at 10uM into 4 mice per compound, along with daily IP injections of BrdU.
Following compound
administration, animals were perfused, sacrificed and subjected to sectioning,
staining and light microscopy in
order to monitor hippocampal neurogenesis localized to the subgranular layer
of the dentate gyms. Roughly
10% of the variant compounds retained pro-neurogenic activity
indistinguishable from the parent compound.
An approximately equal number of compounds yielded slightly diminished
activity, yet the majority of
variants were of significantly diminished activity (Figure 8). For example, a
variant of Example 45
Compound having a methoxy substitution on the aniline ring (Example 62
Compound) was re-tested for pro-
neurogenic activity via direct administration into the brain of adult mice via
Alzet minipumps. The compound
was administered for one week at 10[LM into 4 mice which were injected daily
with BrdU. Following
compound administration, animals were perfused, sacrificed and subjected to
sectioning, staining and light
microscopy in order to monitor hippocampal neurogenesis localized to the
subgranular layer of the dentate
gyrus. The methoxy derivative exhibited activity comparable to Example 45
Compound. Subsequently, the
(+) and (-) enantiomers of Example 62 Compound were prepared (Figure 9A). The
two enantiomers were
evaluated in the in vivo neurogenesis assay. The (+)-enantiomer of Example 62
Compound retained potent
pro-neurogenic activity, and the (-) enantiomer displayed diminished activity
(Figure 9B). Other derivatives
have also been resynthesized and retested, as described above.
Example 45 Compound Enhances the Survival of Newborn Neurons:
The nature of the cells produced in the subgranular zone of the dentate gyrus
was investigated when
Example 45 Compound was administered as follows. Animals were exposed to oral
administration of
Example 45 Compound for 30 days. Brain tissue was then prepared for
immunohistochemical staining with an
antibody to doublecortin (DCX), a microtubule-associated protein that serves
as a marker of neurogenesis in
262
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
the dentate gyrus by virtue of transient expression in newly formed neurons,
but not glial cells, between the
timing of their birth and final maturation (Brown et al., 2003). As shown in
Figure 10A, the relative
abundance of doublecortin-positive neurons increased dramatically as a
function of exposure to prolonged
administration of Example 45 Compound. Although this observation does not rule
out the possibility that the
compound might also enhance the formation of glial cells, it clearly shows
that Example 45 Compound
enhanced the formation of cells destined to become neurons.
Example 45 Compound-mediated neurogenesis was next investigated to see whether
it was
attributable to increased cell proliferation or protection of newborn cells
from cell death during the time
between their birth and eventual incorporation into the granular layer of the
dentate gyrus. This was
accomplished by comparing the ability of Example 45 Compound to enhance either
short- or long-term
increases in the incorporation of BrdU in the dentate gyrus (Figure 10B).
Animals exposed to orally-delivered
Example 45 Compound or vehicle for 30 days were administered a single pulse of
BrdU via IP injection.
Short-term effects on neuron birth were monitored by sacrificing animals one
hour post-BrdU injection,
followed by fixation of the tissue, sectioning and immunohistochemical
detection of BrdU incorporation into
cells localized in the subgranular layer of the dentate gyrus. Example 45
Compound administration did not
lead to an elevation in the level of BrdU-positive cells relative to vehicle
in this short-term assay. At one day
after BrdU administration both groups still showed no statistically
significant differences in number of BrdU+
cells in the dentate gyms. By contrast, at the 5 day time point, by which time
40% of newborn cells in our
assay normally die (Figure 1), animals that received Example 45 Compound
showed a statistically significant,
25% increase in BrdU+ cells compared to the vehicle-only control group. This
difference between groups
progressed with time such that mice that received a daily oral dose of Example
45 Compound for 30 days
starting 24 hours after the pulse treatment of BrdU exhibited a 5-fold
increase in the abundance of BrdU-
positive cells in the dentate gyrus relative to vehicle-only controls.
Notably, in this longer-term trial, BrdU-
positive cells were observed not only along the subgranular layer of the
dentate gyms where new neurons are
known to be born, but also within the granular layer itself It is hypothesized
that these cells represent mature
neurons that have migrated into the granular layer, completed the
differentiation process, and incorporated
themselves into the dentate gyms as properly wired neurons. Observations
supportive of this interpretation
will be presented in a subsequent section of this document. In summary, these
experiments give evidence that
Example 45 Compound enhances the formation of neurons in the mature
hippocampus, and that its mode of
action would appear to take place at some point subsequent to their birth.
It should be appreciated by one of ordinary skill in the art that the above
described cell proliferation
tests can also be used to test other compounds of presently disclosed
embodiments.
Example 45 Compound Normalizes Apoptosis and Ameliorates Morphological and
Electrophysiological
Deficits in the Dentate Gyrus of NPAS3-Deficient Mice:
263
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Mice lacking both copies of the gene encoding neuronal PAS domain protein 3
(NPAS3) suffer a
profound impairment in adult neurogenesis (Pieper et al., Proc. Natl. Acad.
Sci. USA 2005, 102, 14052-
14057). By evaluating BrdU incorporation in a short-term assay of neurogenesis
by sacrificing animals 1
hours after BrdU pulse, it was observed that NPAS3-deficient animals have no
detectable deficit in the birth of
neurons in the subgranular layer of the dentate gyms (Figure 11). This is in
contrast to our earlier
observations of profoundly diminished BrdU labeling in the dentate gyms of
NPAS3-deficient animals when
BrdU is administered for a longer period of time (12 days) (Pieper et al.,
Proc. Natl. Acad. Sci. USA 2005,
102, 14052-14057). Knowing that the NPAS3 transcription factor is required for
proper expression of the
fibroblast growth factor receptor 1 (FGFR1) in the hippocampus (Pieper et al.,
Proc. Natl. Acad. Sci. USA
2005, 102, 14052-14057), it is possible that impediments in growth factor
signaling might impair the trophic
environment critical for the survival of newborn neurons in the dentate gyms.
As an initial test of this
hypothesis, brain tissue prepared from NPAS3-deficient animals was compared
with that of wild type
littermates for the presence of cleaved caspase 3 (CCSP3)-positive cells in
the subgranular layer of the dentate
gyrus. A statistically significant, 2-fold increase in CCSP3-positive
(apoptotic) cells was observed in the
dentate gyms of NPAS3-deficient animals (Figure 11). This enhanced rate of
programmed cell death is likely
to account, at least in part, for the nearly complete elimination of adult
neurogenesis in mice lacking the
NPAS3 transcription factor (Pieper et al., Proc. Natl. Acad. Sci. USA 2005,
102, 14052-14057).
In addition to this quantitative deficit in adult neurogenesis, abnormalities
have been observed in both
the morphology and electrophysiology of granular neurons of the dentate gyrus
of NPAS3-deficient animals.
Relative to wild type animals, Golgi-Cox staining revealed severe attenuation
in dendritic branching and spine
density of dentate gyrus granular neurons of NPAS3-deficient animals (Figure
12A and 12B). By contrast, no
genotype-dependent differences in these measures were observed in pyramidal
cells of the CA1 region of the
hippocampus. Equivalently specific deficits were observed by
electrophysiologic recordings of NPAS3-
deficient animals compared with wild type littermates (Figure 13A and 13B).
Whole field recordings of
excitatory postsynaptic potentials (fEPSP) revealed significant deficits in
NPAS3-deficient animals, relative to
wild type littermates. In the dentate gyms, stimulating and recording
electrodes were positioned in the outer
molecular layer, which is innervated by axons of the perforant pathway
originating from the entorhinal cortex.
In the CA1 region of the hippocampus, stimulation and recording electrodes
were positioned in the stratum
radiatum, which is innervated by the Schaffer collateral axons of CA3
pyramidal cells. Stimulus intensity was
increased in 5 [LA increments, the slope of the decreasing part of field
potentials was measured, and fEPSP was
quantified relative to the amplitude of the fiber volley, which represents
firing of action potentials in pre-
synaptic axons. This analysis revealed aberrant hyper-excitability of synaptic
transmission in npas3-/- mice
both in the outer molecular layer of the dentate gyrus and in the CA1 region
(Figure 13A and 13B).
Armed with these genotype- and region-specific deficits in both neuron
morphology and
electrophysiological activity, whether prolonged administration of Example 45
Compound might favorably
264
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
repair either deficit in NPAS3-deficient animals was tested. Before embarking
on this effort, it was first
confirmed that Example 45 Compound was capable of enhancing hippocampal
neurogenesis in NPAS3-
deficient mice, by demonstrating that Example 45 Compound enhances both BrdU
incorporation as well as
expression of doublecortin in newborn neurons in the dentate gyrus of npas3-/-
mice (Figure 14). Knowing
that formation of the dentate gyms initiates in the late pre-natal mouse
embryo around embryonic day 14
(Stanfield and Cowan, 1988, The development of the hippocampal region. In
Cerebral Cortex, E.G. Jones and
A. Peters, eds.(New York: Plenum Press), pp. 91-131), animals were exposed to
Example 45 Compound for as
extended a period of time as possible in order to give the compound the best
possible chance for exhibiting
favorable effects. Following oral gavage of pregnant female mice, 14 day
embryos were recovered, dissected
and processed by acetonitrile:water extraction so that Example 45 Compound
levels could be measured in the
embryonic brain. Daily administration of 20mg/kg of Example 45 Compound to
pregnant females yielded
appreciable levels of the compound in the brain tissue of developing embryos.
It was similarly observed that
oral administration of the compound to lactating females led to delivery of
Example 45 Compound to the brain
tissue of weanling pups. In both cases, LC/MS-based quantitation of Example 45
Compound revealed levels
of compound accumulation at or above the 1.3504 limit required to support
adult neurogenesis (Figure 7).
Finally, it was observed that daily IP administration of Example 45 Compound
to weaned pups at 20 mg/kg
was sufficient to yield brain levels of Example 45 Compound at or above the
level required to enhance adult
neurogenesis.
Female mice heterozygous at the NPAS3 locus were mated to heterozygous males.
Two weeks post-
mating, females were given a daily oral gavage of either 20mg/kg of Example 45
Compound or vehicle-only
formula. Dosing was continued throughout the last trimester of pregnancy, as
well as the two week post-natal
period of lactation. Following weaning, pups were given a daily IP dose of
either 20 mg/kg Example 45
Compound or vehicle control. At about 7 weeks of age, mice were switched to
oral gavage delivery of the
same dose of Example 45 Compound. When mice were 3 months of age they were
sacrificed and brain tissue
was dissected and subjected to either Golgi-Cox staining or
electrophysiological recording. As shown in
Figure 15, prolonged exposure to Example 45 Compound robustly repaired
morphological deficits in the
dendritic branching of granular neurons of the dentate gyms in NPAS3-deficient
mice. Moreover, as shown in
Figure 13A, the electrophysiological deficit in the dentate gyms of NPAS3-
deficient mice was also corrected
following prolonged exposure of mice to Example 45 Compound. The corresponding
electrophysiological
deficit in CA1 region of the hippocampus, however, was not affected (Figure
13B), underscoring the
specificity of Example 45 Compound to improving functioning of the dentate
gyms in this animal model.
It is also notable that, relative to vehicle-only controls, administration of
Example 45 Compound did
not affect any aspect of the health of mothers, embryos, weanlings or young
adult mice. Gross histology of
brain tissue was normal in both compound- and vehicle-treated animals, and
there was no evidence of neuronal
cell loss or degenerative changes (cytoplasmic eosinophilia, vacuolization or
nuclear pyknosis). The only
265
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
morphological change, other than normalization of dendritic arborization of
granular neurons of the dentate
gyrus, was a compound-dependent increase in the thickness of the granular
layer of the dentate gyrus itself
(Figure 16). The thickness of the granular layer of the dentate gyrus is
roughly 40% less in NPAS3-deficient
animals than wild type littermates. Prolonged administration of Example 45
Compound through late
embryonic development, early post-natal development, and two months post-
weaning significantly corrected
this deficit without affecting the thickness of other hippocampal layers in
NPAS3-deficient mice (Figure 16).
Recognizing that the reduced thickness of the granular layer of the dentate
gyrus in NPAS3-deficient
animals could be attributed to elevated levels of apoptosis of newborn
hippocampal neural precursor cells, the
effect of Example 45 Compound treatment on apoptosis in the hippocampus of
NPAS3-deficient animals was
examined through immunohistochemical staining of cleaved caspase 3 (CCSP3). As
shown in Figure 17, 12
days of treatment with orally delivered Example 45 Compound (20 mg/kg) to
adult NPAS3-deficient animals
significantly reduced CCSP3 staining in the dentate gyms, whereas vehicle-
treatment had not effect. It is
thereby proposed that Example 45 Compound facilitated repair of the granular
layer of the dentate gyms in
NPAS3-deficient mice by ameliorating a genotype-specific exacerbation of
programmed cell death.
It should be appreciated by one of ordinary skill in the art that the above
described apoptosis tests can
also be used to test other compounds of presently disclosed embodiments.
Example 45 Compound Protects Mitochondrial Integrity:
Extensive evidence pioneered by the laboratory of Xiaodong Wang has shown that
an intrinsic
pathway leading to programmed cell death emanates from mitochondria (Liu et
al., Cell 1996, 86, 147-157;
Yang et al., Science 1997, 275, 1129-1132). With the help of the Wang lab,
assays were established to test
whether Example 45 Compound might protect mitochondria from calcium-induced
dissolution (Distelmaier et
al., Cytometry A 2008, 73, 129-138). Tetramethylrhodamine methyl ester (TMRM)
is a cell-permeant,
cationic red-orange fluorescent dye that is readily sequestered by active
mitochondria. When loaded with
TMRM dye, vehicle-only treated cells released the dye within 15 minutes of
exposure to the calcium
ionophore A23187. By contrast, dye release was prevented in cells exposed to
as little as lOng of Example 45
Compound (Figure 18A). As with in vivo neurogenesis assay, as well as the in
vitro protection from A13(25-35)-
mediated toxicity of cultured cortical neurons, preservation of mitochondrial
membrane potential in this assay
was observed only with the (+) enantiomer of Example 62 Compound (Figure 18B).
It should be appreciated by one of ordinary skill in the art that the above
described mitochondrial
integrity tests can also be used to test other compounds of presently
disclosed embodiments.
Comparison of Example 45 Compound and Dimebon:
A chemical compound sharing structural similarity to Example 45 Compound is
2,3,4,5-Tetrahydro-
2,8-dimethy1-5-(2-(6-methyl-3-pyridyl)ethyl)-1H-pyrido(4,3-b)indole (Figure
19A). An anti-histamine, trade
266
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
named Dimebon, was anecdotally noticed over the decades to ameliorate symptoms
of dementia (O'Brien,
Lancet Neurol. 2008, 7, 768-769; Burns and Jacoby, Lancet 2008, 372, 179-180).
More recently, an
American biotechnology company designated Medivation initiated clinical trials
to formally test whether
Dimebon might improve the symptoms of patients suffering from Alzheimer's
disease. The results of FDA-
sponsored, phase 2 clinical trials in Alzheimer's disease were recently
published, reporting favorable response
rates (Doody et al., Lancet 2008, 372, 207-215). Example 45 Compound and
Dimebon were compared in
three functional assays. The in vivo test for effects on hippocampal
neurogenesis revealed activity for both
compounds, with Example 45 Compound exhibiting between 10- and 30-fold higher
level of potency and a
ceiling of efficacy roughly 40% higher than the anti-histamine drug (Figure
19B). Dimebon has been
implicated in protecting mitochondria (Bachurin et al., Ann. NY Acad. Sci.
2001, 939, 425-435; Bachurin et
al., Ann. NY Acad. Sci. 2003, 993, 334-344, discussion 345-349). Therefore
Dimebon was compared with
Example 45 Compound in the calcium-induced mitochondrial dissolution assay.
Both compounds were
observed to be active, and it was again observed that the relative potency of
Example 45 Compound was
superior to Dimebon (Figure 19C). Protection of mitochondrial membrane
permeability was lost for Example
45 Compound between the 10 and 1nM doses, whereas that of Dimebon was lost
between 10 and 1[tM.
Example 45 Compound and Dimebon were tested for binding to the H1 histamine
receptor. While
Dimebon displayed high affinity for this receptor (IC50 < 100 nM), both
enantiomers of Example 45
Compound display low H1 affinity (1050> 10 ,M).
It should be appreciated by one of ordinary skill in the art that the above
described binding activity
tests can also be used to test other compounds of presently disclosed
embodiments.
Effect of Example 45 Compound on Aged Rats
Next, aged Fisher rats were used as a means of performing behavioral tests
capable of assessing the
potential benefits of Example 45 Compound on hippocampus-dependent learning.
It is well established that
normal rodent aging is associated with attenuation of hippocampal neurogenesis
(Kuhn et al., J. Neurosci.
1996, 16, 2027-2033; Driscoll et al., Neuroscience 2006, 139, 1173-1185).
Reduced neurogenesis in aged rats
is likely related to increased neuronal apoptosis in the aged rat brain
(Martin et al., J. Biol. Chem. 2002, 277,
34239-34246; Kim et al., Exp. Gerontol. 2010, 45, 357-365). These changes have
been hypothesized to
contribute to cognitive decline as a function of terminal aging.
It was first evaluated whether Example 45 Compound would enhance hippocampal
neurogenesis in
aged rats as it does in adult mice. Rats were injected with a daily, IP dose
of either 10 mg/kg of Example 45
Compound or vehicle, coinjected with a daily dose of BrdU, and then sacrificed
after 7 days for
immunohistochemistry. As shown in Figure 20A, compound-treated animals
revealed a 500% increase in
BrdU labeling in the dentate gyrus relative to vehicle-treated controls.
Immunohistochemical staining with
267
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
antibodies to doublecortin likewise revealed a robust, compound-specific
enrichment in this marker of
newborn neurons. Having observed proneurogenic efficacy of Example 45 Compound
in this short term assay,
it was then tested whether prolonged administration of Example 45 Compound
might ameliorate age-related
decline in cognition by subjecting 18-month-old rats to daily administration
of either i 0 mg/kg of Example 45
Compound or vehicle only for 2 months. Animals of both groups were further
subjected to weekly IP
administration of BrdU (50 mg/kg) for later immunohistochemical measurements
of hippocampal
neurogenesis. As a control, both Example 45 Compound- and vehicle-treated
groups were confirmed to
display equal ability to physically participate in the task, and learn the
task, as shown by decreased latency
times to find the hidden platform over the 5 day training period, both before
and after 2 months of treatment
(Figure 20B). Moreover, neither swim speed (Figure 20C) nor locomotor activity
(Figure 20D) varied with
age or treatment paradigm.
After 2 months of compound or vehicle administration, cognitive ability was
assessed blind to
treatment group by removing the goal platform. Animals of the Example 45
Compound-treated group retained
a statistically significant improvement in ability to navigate to the region
of the missing platform, as evidenced
by performance in the probe test. As shown in Figure 21A, when the platform
was removed from the maze,
rats treated with Example 45 Compound crossed the precise location previously
containing the platform
significantly more often than vehicle-treated rats. Furthermore, Example 45
Compound-treated rats spent a
higher percentage of time in the general goal area, defined as the quadrant
previously containing the platform,
than vehicle-treated rats (35.5% 2.2% for Example 45 Compound treated, 28.1%
2.6% for vehicle treated,
Student's t Test, p < 0.02).
After behavioral testing, animals were sacrificed for immunohistochemical
detection of BrdU and
CCSP3. As shown in Figure 21B, the dentate gyrus of rats exposed to Example 45
Compound showed a 3-
fold higher level of BrdU-positive neurons than that of the vehicle group.
Moreover, Example 45 Compound-
treated animals showed a statistically significant reduction in the number of
CCSP3-positive cells relative to
vehicle controls (Figure 21C). Unexpectedly, administration of Example 45
Compound helped rats maintain
stable body weight with aging, in contrast to vehicle-treated rats, whose
weight declined steadily with age
(Figure 21D). Example 45 Compound-mediated effects on body weight were
independent of food intake
(Figure 20E), and treatment of aged rats with Example 45 Compound had no
effect on postfasting blood
glucose levels (Figure 20E). Next it was tested whether Example 45 Compound-
mediated preservation of
body weight in aged rats operates via central or peripheral modes of action.
It should be appreciated by one of ordinary skill in the art that the above
described in vivo tests in rats
or other animal models can also be used to test other compounds of presently
disclosed embodiments.
Example 45 Compound Augments Hypothalamic Neurogenesis
268
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Positioned immediately below the thalamus and forming the floor and lower
lateral walls of the third
ventricle, the hypothalamus consists of multiple groups of cells that regulate
the autonomic nervous system
and also control motivational behaviors via extensive neuronal connections to
the pituitary gland, thalamus,
midbrain and cerebral cortex. These functions include water balance,
biological rhythms, feeding and drinking
drive, sexual activity, pituitary gland function and temperature regulation.
Neural stem cells in the adult brain
reside in the wall of the third ventricle and proliferate in response to
various stimuli, and formation of new
neurons in the hypothalamus has also been observed in the hypothalamic
parenchyma. Administration of
trophic factors such as brain-derived neurotrophic factor and ciliary
neurotrophic factor enhances neurogenesis
in the rodent hypothalamus. Furthermore, newborn neurons in the adult
hypothalamus integrate into existing
hypothalamic neural circuits and express neuronal markers such as POMC
(phosphorylated signal transducer
of activator of transcription), neuropeptide Y, ocytocin and vasopressin.
During hypothalamic development,
POMC-expressing progenitor cells differentiate into two populations of cells
with antagonistic roles,
expressing either POMC or neuropeptide Y, that exert opposite effects in
regulating energy balance. It is thus
proposed that differential regulation of postnatally-generated neurons in the
hypothalamus might form the
basis of developing new treatments to regulate food intake behavior. This
hypothesis is supported by
observations that acute ablation of new hypothalamic neurons leads to severe
anorexia and weight loss.
It was evaluated whether P7C3 might augment hypothalamic neurogenesis by
administering either
vehicle or P7C3 (10 mg/kg twice daily, i.p.) to nine week old male C57BL/6
mice, starting two days before
implantation of 7 day Alzet osmotic minipumps (model 1007d) loaded with BrdU
(lmg/kg). Pumps were
connected to a cannula that delivered BrdU at a constant rate into the left
lateral ventricle for the seven day
period, during which time animals continued to receive either vehicle or P7C3.
Pumps were surgically
removed at the conclusion of their 7 day operating period, and mice were
allowed to survive for 4 more weeks,
during which time they continued to receive either vehicle or P7C3. At the end
of the 4 week period, mice
were deeply anesthetized with intraperitoneal (i.p.) injection of mouse
anesthetic cocktail and transcardially
perfused with 4% paraformaldehyde (PFA) in phosphate buffered saline (pH 7.4).
Brains were then dissected
and post-fixed overnight at 4 degrees Celsius in 4% PFA, and cryoprotected in
30 % sucrose in PBS. Fixed
brains were embedded in 0.C.T and cut at 20 micrometer thickness with a
cryostat. Every third section was
immunohistochemically stained for BrdU (Accurate, rat anti-Brdu ,1:400) per
our standard procedures. Anti-
rat Dylight 596 was used to visualize BrdU incorporation. As can be seen from
Figure 27, treatment with
P7C3 markedly enhances hypothalamic neurogenesis in the rodent brain, with a
significantly increased amount
of BrdU positive staining.
It should be appreciated by one of ordinary skill in the art that the above
described hypothalamic
neurogenesis tests can also be used to test other compounds of presently
disclosed embodiments.
Because P7C3 (and its derivatives and analogs) can enhance hypothalamic
neurogenesis, compounds
of the presently disclosed embodiments can be useful for regulating
hypothalamic functions such as water
269
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
balance, biological rhythms, feeding and drinking drive, sexual activity,
pituitary gland function and
temperature regulation. For example, given P7C3's role in maintaining stable
body weight in aging rats,
compounds of the presently disclosed embodiments can provide therapeutic
benefits to patients experiencing
physiological weight loss for various reasons, such as normal aging, radiation
treatment, chemotherapy,
anorexia, cachexia, diabetes, stress, substance abuse, dementia, stroke,
cancer, infection, as well as other
diseases and/or conditions.
Example 45 Compound Protects Mitochondria
Since P7C3 ameliorates the death of newborn neurons in the dentate gyms in
living mice, it is possible
that its function might relate to mitochondrial integrity. Assays were
established to test whether P7C3 might
protect cultured U205 cells from calcium-induced mitochondrial dissolution
(Distelmaier et al., Cytometry A
2008, 73, 129-138). Tetramethylrhodamine methylester (TMRM) dye is sequestered
by active mitochondria,
and, when loaded with TMRM, vehicle-treated cells released the dye within 15
rain of exposure to the calcium
ionophor A23187. By contrast, dye release was fully prevented in cells exposed
to as little as lOnM of P7C3
(Figure 22A). Compounds known to be less active in vivo were also less active
in this assay (not shown).
Preservation of mitochondrial membrane potential in this assay was observed
for the R-enantiomer of P7C3-
0Me, Example lb, (Figure 22B), but not the S-enantiomer, Example la, (Figure
22C). Finally, protection of
mitochondrial membrane permeability was observed at an enhanced level for a
compound variant P7C3A20
(Example 6a), which also exhibited a high level of proneurogenic activity
(Figure 22D). Derivatives that have
less proneurogenic activity than P7C3 such as Example 33 (Figure 22E) and
Example 21 (Figure 22F),
displayed less protective effect in preserving mitochondrial integrity at the
tested doses in cultured primary
cortical neurons.
It was also examined whether Example 45 Compound preserves mitochondrial
membrane potential in
cultured primary cortical neurons (Figure 23). Cortical neurons cultures from
rats on embryonic day 14 were
loaded with tetramethylrhodamine methyl ester (TMRM) dye after 6 days of
maturation. The top panels (no
calcium ionophore) show that the dye alone did not affect the health of
neurons. The remaining panels are
from cells that were exposed to the calcium ionophore A23187 at time zero.
With vehicle-alone, cortical
neuron mitochondrial membrane potential was rapidly lost after exposure to the
ionophore. Escalating doses
of Example 45 Compound preserved mitochondrial membrane potential following
exposure to the calcium
ionophore A23187 in a dose dependent manner, with full protection achieved at
1 mM. The less active
compound (Example 33) was less effective in preserving mitochondrial membrane
potential at any dose tested.
Results shown are representative of 10 fields analyzed in each of 2
experimental runs for all conditions.
270
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
It should be appreciated by one of ordinary skill in the art that the above
described mitochondrial tests
can also be used to test other compounds of presently disclosed embodiments.
Example 45 Compound Normalizes Elevated Levels of Hippocampal Apoptosis in
npas3 -/-Mice
Recognizing that reduced thickness of the npas3 -I- dentate gyrus granular
layer could be attributed to
increased apoptosis of proliferating neural precursor cells, the effect of
Example 45 Compound (P7C3)
treatment on apoptosis in the hippocampus of npas3 -/-mice was examined
through immunohistochemical
staining of CCSP3. As shown in Figure 17, after 12 days of orally delivered
Example 45 Compound (20
mg/kg) to adult npas3-/- mice, a statistically significant reduction in CCSP3
staining was observed in the
dentate gyms. It is thereby proposed that Example 45 Compound facilitates
repair of the granular layer of the
dentate gyms in npas3 4- mice by overcoming a genotype-specific enhancement in
apoptosis.
It should be appreciated by one of ordinary skill in the art that the above
described mice model and
other animal model can also be used to test other compounds of presently
disclosed embodiments.
Example 45 Compound (P7C3) Provides Therapeutic Benefit in Animal Model of
Amyotrophic Lateral
Sclerosis (ALS)
ALS, also known as Lou Gehrig's disease, is an adult-onset (typically between
ages 40-70), rapidly
progressive and fatal disease caused by selective degeneration of upper
(cortical layer V within the primary
motor cortex) and lower (spinal cord) motor neurons, the nerve cells in the
central nervous system that control
voluntary muscle movement. An estimated 5000 people in the United States are
diagnosed with ALS every
year. This disorder causes muscle weakness and atrophy throughout the body,
and patients with ALS
ultimately lose their ability to initiate and control all voluntary movement.
The earliest parts of the body
affected in ALS reflect those motor neurons that are damaged first. About 75%
of patients experience onset of
symptoms in their arms or legs manifested as difficulty with manual dexterity
or ambulation, while about 25%
experience 'bulbar onset' of ALS ¨ difficulty speaking clearly or swallowing.
A small proportion of patients
have respiratory onset of ALS in the form of weakness of the intercostal
muscles that support breathing.
Regardless of the region of onset, muscle weakness and atrophy invariably
spread to other parts of the body as
the disease progresses. Most patients develop a constellation of symptoms that
includes difficulty moving,
dysphagia (difficulty swallowing), dysarthria (difficulty speaking or forming
words) and classical
manifestations of loss of upper motor neurons (muscular spasticity,
hyperreflexia and overactive gag reflex)
and lower motor neurons (muscular weakness, muscle atrophy, muscle cramps and
fasciculations). Sensory
nerves and the autonomic nervous system are usually spared, though may be
involved in some patients. About
20% of ALS patients also develop frontotemporal lobar dementia (FTLD), while
30-50% of patients develop
subtle cognitive changes that can be observed with detailed neuropsychological
testing. Around 15-45% of
patients with ALS also experience what is called "pseudobulbar affect" ¨ a
form of emotional lability in which
271
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
patients manifest intermittent bouts of uncontrollable laughter, crying or
smiling. This symptom domain is
thought to be related to degeneration of bulbar upper motor neurons, resulting
in exaggerated motor
expressions of emotion. Although disease progression varies between
individuals, most patients are eventually
unable to stand or walk, get in or out of bed on their own, or use their hands
and arms. Difficulty chewing and
swallowing further leads to progressive weight loss and increased risk of
choking and aspiration pneumonia.
Towards the end stages of disease, as the diaphragm and intercostal muscles
weaken, most patients require
ventilator support. Individuals with ALS most commonly die of respiratory
failure or pneumonia within 2-5
years of diagnosis.
Ninety-five percent of ALS cases occur sporadically (SALS), with no
identifiable cause or family
history of the disease. The remaining 5% of cases are inherited, known as
Familial ALS (FALS). Because
FALS and SALS are clinically and neuropathologically similar, the pathogenesis
of these forms of ALS may
converge on a common pathogenic pathway. Approximately 20% of FALS and 3% of
SALS cases are
associated with autosomal dominant mutations in the SOD1 gene on chromosome
21, and about 150 different
mutations dispersed throughout this gene have been identified in FALS. SOD1
encodes cytosolic Cu/Zn
superoxide dismutase, an antioxidant enzyme that protects cells by converting
superoxide (a toxic free radical
generated through normal metabolic activity of mitochondria) to hydrogen
peroxide. Unchecked, free radicals
accumulate and damage both mitochondrial and nuclear DNA, as well as proteins
within cells. In ALS linked
to mutations in SOD1, cytotoxicity of motor neurons appears to result from a
gain of toxic SOD1 function,
rather than from loss of dismutase activity. Although the exact molecular
mechanisms underlying toxicity are
unclear, mutation-induced conformational changes in SOD1 are known to lead to
misfolding and subsequent
cytotoxic aggregation of mutant SOD1 in cell bodies and axons. Aggregate
accumulation of mutant SOD1 is
thought to disrupt cellular functions and precipitate neuron death by damaging
mitochondria, proteasomes,
protein folding chaperones, or other proteins.
Transgenic animal models of mutant SOD1 are currently used for research into
the pathogenic
mechanisms thought to broadly underlie ALS, such as G93A SOD1 mutant mice.
Mice hemizygous for the
G93A-SOD1 transgene express 18 +/- 2.6 copies of a form of SOD1 found in some
patients with FALS (a
substitution of glycine to alanine at codon 93). This was the first mutant
form of SOD1 to be expressed in
mice, and is the most widely used and well-characterized mouse model of ALS.
Superoxide dismutase activity
in these mice is left intact such that the pathogenic effect of the mutant
transgene appears to be gain of
function, as is thought to occur in human patients. In these mice, death of
motor neurons in the ventral horn
of the spinal cord and loss of myelinated axons in ventral motor roots leads
to paralysis and muscle atrophy.
Upper cortical motor neurons in these mice also die as the disease progresses,
and protein aggregates of mutant
SOD1 are found only in diseased tissues, with greater amounts being detected
during motor neuron
degeneration. Around 100 days of age, G93A-SOD1 mice become paralyzed in one
or more limbs with
272
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
paralysis due to loss of motor neurons from the spinal cord. This paralysis
rapidly spreads throughout the
body, culminating in 50% death when mice are 128.9 +/- 9.1 days old.
P7C3 was intraperitoneally administered to female G93A-SOD1 transgenic mice
using a treatment
paradigm of 10 mg/kg P7C3 i.p. twice a day, compared to vehicle, starting at
40 days of age. This treatment
scheme was selected based on standard protocols for initial proof of concept
screens in these mice. To control
for transgene copy number, mice are sibling matched between treatment groups,
as per standard protocol.
After initiation of P7C3 or vehicle treatment, date of onset of illness is
determined by peak weight, and initial
progression of disease is defined as the day at which mice fall to 10% below
their maximum weight. Mice are
also assessed daily by a standard determination of neurological severity
score, with a score of 2 or worse for
two consecutive days serving as an additional marker of illness progression.
This score is determined blind to
treatment group with the scoring system described in the legend for the
figure. As shown in Figure 24A,
P7C3 treatment slows disease progression in G93A-SOD1 mice in terms of
delaying the time point at which
mice drop to 10% below their maximum weight. Treatment with P7C3 also
significantly delays the age at
which G93A-SOD1 mice attain a neurological severity score of 2, another marker
of disease progression, as
shown in Figure 24B. Furthermore, P7C3 treatment significantly improved
performance in the accelerating
rotarod task as the disease progressed in these mice, as shown in Figure 24C,
indicating a slowing of
progression of motor impairment in the disease process. This protective
effective of P7C3 on motor
performance in G93A-SOD1 mice is also observed in the ink footprint analysis
of walking gait, as shown in
Figure 24D.
It should be appreciated by one of ordinary skill in the art that the above
described ALS model and
other animal model can also be used to test other compounds of presently
disclosed embodiments.
Example 6a Compound (P7C3A20) Provides Therapeutic Benefit in Animal Model of
Parkinson's
Disease
Parkinson's disease (PD) is a progressive neurodegenerative disease
characterized by the death of
dopaminergic neurons in the substantia nigra, which project to the striatum to
control normal movement.
Though it is one of the most common nervous system disorders of the elderly,
the cause of PD remains
uncertain. Symptoms early in the disease are movement-related, including
shaking, rigidity, slowness of
movement, and difficulty with walking gait. More advanced stages of the
disease are typically associated with
cognitive and behavioral problems, including dementia. The early motor
symptoms are partially managed by
administration of drugs that enhance dopaminergic signaling. However, as the
disease progresses and the
dopaminergic neurons in the substantia nigra continue to die, patients reach a
point at which these drugs
become ineffective at treating the symptoms and additionally produce the
complication of dyskinesia.
Effectively preventing the death of dopaminergic neurons in the substantia
nigra would therefore be an ideal
treatment approach for patients with PD.
273
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
MPTP (1-methy1-4-pheny1-1,2,3,6-tetrahydropyridine) is a potent neurotoxin
that selectively kills
dopaminergic neurons in the substantia nigra of both mice and monkeys, causing
a clinical picture resembling
PD. The MPTP toxicity model can therefore be used to study the death of
dopaminergic neurons with the goal
of developing new treatments for PD based on neuroprotective strategies found
to be effective in these
neurons. To determine if P7C3A20 might be neuroprotective in the substantia
nigra, the well-characterized
and popular MPTP administration regimen was employed, as developed by Tatton
and Kish (1997),
Neuroscience 77: 1037-1048, and Jakson-Lewis et al. (2007), Nature Protocols
2: 141-151. Here, 12 week old
wild type male C57BL/6 mice were treated for 3 days with P7C3A20 (10 mg/kg
i.p. twice daily) or vehicle,
and on the fourth day a five day regimen of 30 mg/kg/day i.p. free base MPTP
was initiated. During this five
day period of MPTP administration the mice continued to receive P7C3A20 or
vehicle. Mice continued to
receive the same dose of P7C3A20 or vehicle every day for 21 more days, at
which point they were sacrificed
by transcardial perfusion with 4% paraformaldehyde. Brains were post-fixed in
4% paraformaldehyde at 4
degrees Celsius overnight and then cryoprotected with 30% sucrose in phosphate-
buffered saline. Fixed brains
were cut at 30 microns with a sliding microtone, and every 4th section (spaced
120 microns apart) was stained
with antibodies directed against tyrosine hydroxylase (TH) (Abcam, rabbit anti-
TH, 1:2500). TH-positive
cells were counted in the substantia nigra area. As shown in Figure 25A and
Figure 25B, treatment with
P7C3A20 significantly attenuates MPTP-mediated killing of substantia nigra
dopaminergic neurons. These
observations suggest that P7C3A20 and related compounds may form the basis of
new neuroprotective
strategies for preventing or slowing the progression of Parkinson's disease.
It should be appreciated by one of ordinary skill in the art that the above
described PD model and
other animal model can also be used to test other compounds of presently
disclosed embodiments.
Example 45 Compound Provides Therapeutic Benefit in Animal Model of
Huntington's Disease
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease
characterized by the
insidious and progressive development of mood disturbances, behavioral
changes, involuntary choreiform
movements (ceaseless and complex writhing movements of the limbs) and
cognitive impairment. HD has a
prevalence of about 1 in 10,000 people in the U.S., and is caused by a
polyglutamine expansion of greater than
36 repeats in the N terminus of the protein huntingtin (Htt). There are
currently no treatments that delay the
appearance or progression of this disease. HD is pathologically characterized
by a dramatic loss of neurons in
the striatum and cerebral cortex, and therapeutic strategies to protect these
neurons from dying might provide
new treatment options for patients. The physical symptoms of HD typically have
their onset between 35-44
years of age, though onset has been reported to occur at times ranging from
infancy to old age. The exact way
in which HD affects an individual varies and can differ even between members
of the same family, but
symptoms progress predictably in most cases. The earliest symptoms include a
general lack of coordination
and unsteady gait, and as the disease advances uncoordinated and jerky body
movements become more
274
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
apparent. More advanced stages are typically accompanied by an observable
decline in mental abilities,
associated with behavioral and psychiatric problems, such as anxiety, severe
depression, blunted affect,
egocentrism, aggression, and compulsive behaviors such as alcoholism, gambling
or hypersexuality. Over
time, physical abilities are gradually impeded until coordinated movement
becomes very difficult, and mental
abilities generally decline into dementia. Complications such as pneumonia,
heart disease, eating difficulties
leading to weight loss and malnutrition, and physical injury from falls reduce
life expectancy to around twenty
years after onset of symptoms. There is no cure for HD, and full-time care is
required in later stages of disease.
Htt is a large cytoplasmic protein that interacts with over 100 other
proteins, and appears to have
multiple biological functions. The behavior of mutated Htt (mHtt) protein is
not completely understood, but it
is known to be toxic to neurons. Damage mainly occurs in the striatum, but in
later stages other areas of the
brain are also attacked, such as the cerebral cortex. As neuronal cell death
progresses, symptoms associated
with the functions of the affected brain areas appear. For example, planning
and modulating movement are the
main functions of the striatum, and difficulties with these tasks are frequent
initial symptoms of HD. Disease
initiation and progression are thought to involve in large part a
conformational change in the mHtt protein due
to the polyglutamine expansion, altered protein-protein interactions, abnormal
protein aggregation in both the
nucleus and cytoplasm and proteolysis, which in turn may lead to
transcriptional dysregulation, excitotoxicity,
mitochondrial dysfunction, and neuronal apoptosis. In addition to a role for a
gain of new toxic properties of
mHtt in HD pathology, there is increasing evidence that loss of wild-type Htt
function also contributes to
pathogenesis. For example, an essential role of Htt in mitotic spindle
formation and mammalian neurogenesis
has recently been identified.
One animal model of HD that can be employed for screening potential
therapeutic agents is R6/2
transgenic mice. These mice express a mutant exon 1 of the human huntingtin
gene, engineered to include an
approximately 145-155 CAG repeat expansion. R6/2 mice phenocopy much of the
neuropathology (striatal
and cortical neuron cell death) and behavioral manifestations of clinical HD.
They display progressive motor
and cognitive impairments, ubiquitinated nuclear and cytoplasmic inclusions of
mutant Htt, weight loss,
decreased striatal and brain size, altered levels of neurotransmitters and
their receptors, and premature death.
They exhibit motor deficits as early as 5-6 weeks of age, display overt
behavioral abnormalities at 8-9 weeks,
and typically die between 11 and 13 weeks of age. R6/2 mice also display
significantly lower levels of adult
hippocampal neurogenesis relative to wild-type littermates, even before onset
of symptoms.
In one hypothesis, P7C3 (and its derivates) may enhance the formation of
neurons in the mature
hippocampus by preventing death rather than promoting proliferation of these
cells. As such, P7C3 is
"proneurogenic" by virtue of its neuroprotective activity. It is also possible
that P7C3 (and its derivates)
prevents cell death and promotes cell proliferation. It was evaluated whether
P7C3 might provide therapeutic
benefit in R6/2 mice. P7C3 (10 mg/kg i.p. twice daily starting at 6 weeks of
age) or vehicle were administered
to 40 female R6/2 mice. As shown in Figure 26A, 50% of vehicle-treated R6/2
mice die at approximately 15
275
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
weeks of age, and treatment with P7C3 delays animal death by about three
weeks. At 14 weeks of age, R6/2
mice treated with P7C3 showed improved general condition score and appearance
as shown in Figure 26B, as
compared to vehicle-treated littermates. General condition score was
determined by a 3 point scoring system
that was conducted blind to genotype and treatment group (score of 0 = fur
looks groomed, normal posture (no
hunch), clear eyes, alert; score of 1 = fur beginning to stick up, slight
hunch; score of 2 = piloerection (fur
sticking up), unkempt fur, hunch in back or neck area, crusty eyes). Death was
monitored twice daily, and
defined as either when animals were found dead, or when they were unable to
right themselves after being
placed on their backs with movement subsequently initiated by gentle prodding
for 30 seconds. By general
appearance of coat condition, grooming and spontaneous activity in the home
cage, R6/2 mice treated with
P7C3 also appear qualitatively better than VEH-treated R6/2 mutant mice (not
shown).
It should be appreciated by one of ordinary skill in the art that the above
described HD model and
other animal model can also be used to test other compounds of presently
disclosed embodiments.
Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of
Parkinson's disease
Parkinson's disease (PD) is an incurable and progressive neurodegenerative
disorder of predominantly
idiopathic origin that is characterized by the death of dopaminergic neurons
in the substantia nigra pars
compacta (SNc), a region of the brain that controls motor activity by
projecting dopaminergic axons to the
striatum (Lees A.J., Hardy J., Revesz T (2012) Parkinson's disease. Lancet
373:2055-2066). Early symptoms
in PD are primarily movement-related, including shaking, rigidity, brady- and
hypo-kinesia, tremor and
difficulty walking. More advanced stages of PD are associated with cognitive
and behavioral problems,
including dementia. Current treatment strategies for PD consist primarily of
partial management of early
motor symptoms with drugs that enhance dopaminergic signaling, such as L-DOPA
or dopamine receptor
agonists. Unfortunately, as greater numbers of dopaminergic neurons in the SNc
die, these drugs fail to
alleviate symptoms and additionally produce dyskinesia. There is thus a
significant unmet need for new
pharmacologic strategies to slow the progression of PD, such as drugs capable
of blocking the death of SNc
dopaminergic neurons.
We have previously reported the identification of an aminopropyl carbazole
(P7C3) discovered via an
unbiased, in vivo screen for small molecules capable of enhancing postnatal
hippocampal neurogenesis. P7C3
displays enantiomeric-selective stabilization of mitochondrial membrane
potential, and enhances neurogenesis
by blocking apoptosis of newborn neurons in the dentate gyrus (Pieper A.A., et
al. (2010) Discovery of a
Proneurogenic, Neuroprotective Chemical. Cell 142:39-51). Prolonged oral or
intraperitoneal (i.p.)
administration of P7C3 to rodents safely improves hippocampal functioning. For
example, administration of
P7C3 to mice suffering from pathologically high levels of neuronal apoptosis
in the dentate gyms, neuronal
PAS domain protein 3 (NPAS3) ¨ deficient mice (Pieper A.A., et al. (2005) The
neuronal PAS domain protein
276
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
3 transcription factor controls FGF-mediated adult hippocampal neurogenesis in
mice. Proc Natl Acad Sci
USA 102:14052-14057), restored hippocampal structure and function with no
obvious physiologic side effects
(Pieper A.A., et al. (2010) Discovery of a Proneurogenic, Neuroprotective
Chemical. Cell 142:39-51). In
addition, extended administration of P7C3 to aged rats safely impeded
hippocampal cell death and preserved
cognitive ability as a function of terminal aging (Pieper A.A., et al. (2010)
Discovery of a Proneurogenic,
Neuroprotective Chemical. Cell 142:39-51).
Through an in vivo structure-activity relationship (SAR) study, we have
identified analogs of P7C3
displaying either increased or decreased activity. In particular, a chemical
variant known as P7C3A20 was
observed to have greater potency and efficacy than P7C3. P7C3A20 differs from
P7C3 by virtue of replacing
the hydroxyl group at the chiral center of the linker with a fluorine, and the
addition of a methoxy group to the
aniline ring. This analog displays a more favorable toxicity profile than
P7C3, with no hERG channel binding,
histamine receptor binding or toxicity to HeLa cells. We have also found that
Dimebon, an antihistaminergic
drug long deployed in Russia that is claimed to have anti-apoptotic and
mitochondrial protective properties,
displays modest efficacy in the same biologic assays employed to discover and
characterize P7C3 and
P7C3A20. The chemical structure of Dimebon is related to the P7C3 class of
aminopropyl carbazoles, yet its
rank order of activity relative to chemical derivatives of P7C3 is very low.
Here, we report that the
neuroprotective activity of these agents extends beyond promoting long-term
survival of newborn cells in the
adult hippocampus. Specifically, we show that the most active variants of P7C3
exhibit robust protection of
mature dopaminergic neurons in both mouse and worm models of
neurodegeneration, and propose that
substituted carbazoles may represent attractive chemical scaffolds for the
optimization of therapeutic agents
for the treatment of Parkinson's disease.
Results
Neuroprotective efficacy of P7C3, P7C3A20 and Dimebon for newborn hippocampal
neurons.
Adult hippocampal neurogenesis in mice is an approximately month-long process,
during which time
the majority of newborn cells die as they transition through a
"differentiation gauntlet" lasting about 1 month
before surviving cells become functionally wired into the central nervous
system. We have previously found
that approximately 40% of these newborn cells die within the first week
following their birth in the
subgranular zone of the dentate gyms. P7C3 was originally discovered through a
week-long in vivo screen
designed to identify small molecules that might enhance either proliferation
or survival of newborn
hippocampal neural precursor cells. Subsequent bromodeoxyuridine (BrdU) pulse-
chase labeling studies
revealed that P7C3 does not affect neural precursor proliferation, but instead
augments survival of newborn
cells by blocking apoptosis. P7C3A20 was found to be active at lower doses and
to have a higher ceiling of
efficacy (CoE) than P7C3 in this 7 day in vivo assay, whereas Dimebon was
shown to be substantially less
potent and efficacious than P7C3.
277
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
In order to more carefully compare the neuroprotective efficacy of these
compounds in abetting
hippocampal neurogenesis, we conducted dose-response studies in a 30 day BrdU
pulse-chase survival assay
(Figure 28). Briefly, newborn cells were labeled with a single intraperitoneal
(i.p.) injection of BrdU (150
mg/kg), followed by daily treatment with the three test compounds beginning
the following day. After 30 days
of compound administration, mice were transcardially perfused with 4%
paraformaldehyde, brains were
dissected, and immunohistochemical detection of BrdU followed by standard
microscopic imaging and
normalization for dentate gyrus volume was used to quantify the number of
surviving cells. As shown in
Figure 28, P7C3A20 increased neuron survival by almost 100% at the lowest dose
tested (5 mg/kg/day). By
contrast, neither P7C3 nor Dimebon exhibited any effect at this dose. At the
next higher dose (10 mg/kg/day),
P7C3 and Dimebon showed 65% and 15% neuroprotective efficacy, respectively,
whereas P7C3A20 showed
neuroprotective ceiling of efficacy (CoE) of about 175%. All three compounds
exhibited their maximal
individual CoEs at the two highest doses tested (20 or 40 mg/kg/day), with
P7C3A20's CoE being highest
(;:t 230% increase in survival), P7C3's CoE being intermediate (:t 130%
increase in survival) and Dimebon's
CoE being the lowest (:t 30% increase in survival).
Efficacy assays for P7C3, P7C3A20 and Dimebon for protection from MPTP-
toxicity to dopaminergic neurons
in mice.
The protective efficacy of the three test compounds for newborn hippocampal
neurons in the 30 day
survival assay prompted us to investigate whether they might also have
neuroprotective efficacy in mature
neurons outside of the hippocampus. To investigate this hypothesis, we
utilized the 1-methy1-4-phenyl-
1,2,3,6-tetrahydropyridine (MPTP) model of neuronal cell death. MPTP is a
potent toxin that selectively kills
neurons in the substantia nigra of both rodents and primates, causing clinical
manifestations resembling PD
(Fukuda T. (2001) Neurotoxicity of MPTP. Neuropathology 21:323-332). MPTP is
lipophilic and readily
crosses into the brain, where it is metabolized by monoamine oxidase B in
glial cells into the highly toxic
cation 1-methy1-4-phenylpyridinium (MPP ). MPP is selectively concentrated in
SNc dopaminergic neurons
by virtue of its high affinity for the plasma membrane dopamine transporter,
and toxicity is further potentiated
by binding of MPP to melanin in these cells, creating a depot mechanism that
maintains prolonged high
intracellular concentrations of MPP . MPTP toxicity is routinely used to study
the death of dopaminergic
neurons as a possible means of discovering new treatments for PD based on
neuroprotective strategies found to
be effective in this model.
We compared the protective efficacy of our agents in the Tatton and Kish model
of MPTP
administration, which induces prolonged apoptotic death of SNc dopaminergic
neurons lasting about 3 weeks
after a short course of daily MPTP administration. Mice were treated daily for
5 days with 30 mg/kg/day free
base MPTP. On the sixth day, 24 hours after receiving the fifth and final dose
of MPTP, daily treatment with
P7C3, P7C3A20, Dimebon or vehicle was initiated. This testing paradigm ensured
that any observed activity
278
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
of P7C3 or its analogs could be attributed to neuroprotective effects, and not
to disruption of MPTP uptake or
metabolism. Dose-response studies were conducted in which mice received twice
daily doses of each
compound (or vehicle) for the ensuing 21 days (Figure 29A). Treatment groups
consisted of 15 animals each.
At the end of the 21 day treatment period, mice were sacrificed by
transcardial perfusion with 4%
paraformaldehyde, and fixed brains were sectioned through the striatum and SNc
at 30 ,M intervals. Every
fourth section (spaced 120 ,M apart) was stained with antibodies specific to
tyrosine hydroxylase (TH)
(Abcam, rabbit anti-TH, 1:2500). The TH enzyme catalyzes the conversion of the
amino acid L-tyrosine to L-
3,4-dihydroxyphenylalanine (L-DOPA), which serves as the precursor for
dopamine. TH staining thus
provides a means to immunohistochemically identify dopaminergic neurons. By
counting the number of TH+
cells in the SNc, we were able to assess the neuroprotective efficacy of the
three chemicals following MPTP
exposure. All microscopic analysis was performed by two investigators, blind
to treatment group.
As has been observed by others, MPTP administration reduced the number of TH+
neurons in the SNc
by about 50% (VEH) (Figure 29A). This neurotoxicity was blocked to varying
degrees by both P7C3 and
P7C3A20. P7C3 enhanced survival by about 40% over VEH at a dose of 5
mg/kg/day, and the highest dose of
P7C3 (20 mg/kg/day) afforded almost 60% protection relative to vehicle. By
contrast, the 20 mg/kg/day dose
of P7C3A20 preserved the number of dopaminergic neurons in the SNc to about
85% of that seen in normal
mice not exposed to MPTP. At every dose tested, P7C3A20 provided superior
protection to P7C3. Both CoE
and potency of P7C3A20 were greater than P7C3, with the onset of P7C3A20
efficacy (30% over VEH) at a
dose of 1 mg/kg/day. Dimebon failed to confer any measurable degree of
protection from MPTP at any dose.
In addition to allowing quantification of dopaminergic cells in the SNc, TH
staining is also routinely
employed to visualize the integrity of dopaminergic axonal protections from
SNc cell bodies into the striatum.
Figure 29B shows that the highest dose of P7C3A20 (20 mg/kg/day) almost
completely blocked depletion of
dopaminergic axons in the striatum after MPTP exposure. The highest dose of
P7C3 also revealed
qualitatively notable protection. By contrast, Dimebon offered no protection
of dopaminergic axons in the
striatum, corresponding to its lack of neuroprotective efficacy in the SNc. As
shown in Figure 30, LC/MS/MS
quantification of brain and blood levels of P7C3A20 and P7C3 confirmed that
the neuroprotective efficacy for
each compound correlated with brain and blood levels for each chemical.
Notably, P7C3A20 displayed
significantly greater protective efficacy than P7C3 in spite of the fact that
P7C3A20 accumulated in brain
tissue at less than one-tenth the concentration of P7C3. Dimebon, which
displayed no neuroprotective efficacy
in the MPTP model of dopaminergic neuron cell death, showed comparable levels
of brain accumulation to
P7C3A20.
Efficacy assays of P7C3, P7C3A20 and Dimebon for protection from MPP -
toxicity to dopaminergic neurons
in C. elegans.
279
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Genes, metabolic signaling pathways, neurotransmitters and receptor
pharmacology are highly
conserved between C. elegans and vertebrates, and exposure of C. elegans to
MPP has been reported to
selectively kill dopaminergic neurons and impair mobility. To investigate the
neuroprotective efficacy of
P7C3A20, P7C3 and Dimebon from MPP toxicity in C. elegans, we monitored
dopaminergic cell death in a
transgenic strain of worms in which dopaminergic neurons fluoresce green by
virtue of GFP expression driven
by the dopaminergic neuron-specific promoter dat-1. As shown in Figure 31,
incubation of synchronized Li
larvae for 40 hours with 5 mM MPP elicited virtually complete destruction of
all four cephalic sensilla
dopaminergic dendrites. For this assessment, GFP fluorescence was observed in
20 worms per group, and was
performed in triplicate. Cephalic sensilla dopaminergic dendrites were
observed under 40X magnification
(AMG, Evos fl microscope), and GFP signal was followed from the nerve ring to
the tip of the nose, following
established protocols. If any part of a dendrite was absent, as evidenced by
loss of GFP signal, it was counted
as degraded. All analyses were performed blind to treatment group.
Co-treatment of MPP -exposed worms with 10 .1\4 P7C3A20 conferred 80%
protection. By
comparison, the neuroprotective efficacy of the same dose of P7C3 was only
about 50%. Neuroprotective
efficacy of both agents was diminished as the chemical dose was gradually
reduced. By these measures,
P7C3A20 showed greater potency than P7C3, with an onset of neuroprotective
efficacy of 30% at the 0.1 M
dose. By comparison, P7C3 did not show efficacy (35%) until administered at
1.0 M. Administration of
Dimebon at the highest dose (10 .1\4) failed to protect dopaminergic neurons
in worms from MPP -induced
toxicity.
Efficacy assays of P7C3, P7C3A20 and Dimebon for protection from MPP -induced
mobility deficits in C.
elegans.
As a behavioral measure of toxicity, worm mobility was assessed 32 hours after
MPP -exposure. To
quantify worm locomotion, video representations were recorded for 10 seconds
at 4X magnification using a
Nikon Eclipse 80i microscope. Each video segment consisted of 160 frames, and
the head of each worm (10-
20 worms per group, repeated in triplicate) was manually tracked in each frame
using Imera software. The
body length of each worm was also measured by Imera software. The ratio of
distance traveled to body length
was used to determine the movement index, defined as locomotion, as previously
established (Wang J., et al.
(2009) An ALS-linked mutant SOD1 produces a locomotor defect associated with
aggregation and synaptic
dysfunction when expressed in neurons of Caenorhabditis elegans. PLoS Genetics
e10003350). As shown in
Figure 32, locomotion was reduced in C. elegans by 50% after 32 hours of
exposure to 5mM MPP . Co-
incubation of MPP -exposed worms with 10 .1\4 P7C3A20 conferred 80%
preservation of locomotion, while
280
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
,M P7C3 protected to about 60% of normal levels. Dimebon offered no protective
efficacy in this
behavioral assay.
Correlation of efficacy of new analogs of P7C3 in the in vivo hippocampal
neurogenesis assay with
5 neuroprotective efficacy in MPTP-mediated dopaminergic cell death.
Over the past two years, we have conducted a comprehensive structure-activity
relationship (SAR)
study in order to improve the potency, efficacy and physical properties of the
P7C3 series of molecules, as
well as to eliminate real or perceived chemical liabilities. To date, we have
synthesized over 300 analogs of
P7C3, all of which have been evaluated by primary screening in the in vivo
hippocampal neurogenesis assay.
10 Our efforts include, but are not limited to, eliminating the bromines
and aniline ring, increasing biologic
activity, decreasing lipophilicity, eliminating toxicities such as hERG
channel binding, increasing solubility
and reducing molecular weight. Here, we show the results of evaluation of 8 of
these new analogs (Figure
33A) in both the hippocampal neurogenesis assay (4 mice for each compound) and
the MPTP protection assay
(10 mice for each compound) (Figure 33B). All analyses were conducted blind to
treatment group.
With respect to the original P7C3 scaffold, P7C3-S7 differs by replacing the
aniline NH with a sulfide
linker, P7C3-S8 differs by replacing the aniline phenyl ring with a
pyrimidine, and P7C3-S25 differs by
replacing the aniline moiety with a dimethyl pyrazole (Figure 33A). As shown
in Figure 33B, all of the eight
test molecules crossed the blood brain barrier. P7C3-S7 and P7C3-S25 were
active in both the hippocampal
neurogenesis assay and the MPTP protection assay. By contrast, P7C3-S8 was
devoid of activity in both
assays. We also compared the efficacy of members of a new enantiomeric pair in
these assays. P7C3-S40 and
P7C3-S41 differ from P7C3 by replacing the aniline NH with an oxygen linker
(Figure 33A). P7C3S40 and
P7C3S41 are the R and S single enantiomers, respectively, and Figure 33B shows
that neuroprotective activity
in both assays resides exclusively in the S enantiomer. P7C3-S54 differs from
P7C3 mainly by the addition of
a methyl group to the central carbon of the propyl linker, and additionally
has an OMe group on the aniline
ring (Figure 33A). This analog was observed to retain neuroprotective activity
in both assays. P7C3-S165
differs from P7C3 by replacing the aniline and carbinol fragments with a
carboxylic acid, a change that
dramatically increases polarity (Figure 33A). Encouragingly, neuroprotection
was observed in both assays
(Figure 33B).
Finally, P7C3-S184 differs from P7C3 by replacing the bromines on the
carbazole with chlorines, and
by replacing the aniline with a naphthyl amine (Figure 33A). This molecule was
inactive in both in vivo
assays. P7C3-S184 has been reported as a 13-secretase (BACE1) inhibitor (Asso
V., et al.(2008) alpha-
naphthylaminopropan-2-ol derivatives as BACE1 inhibitors. ChemMedChem 3:1530-
1534). BACE1 is an
aspartate proteolytic enzyme that catalyzes the formation of A13 peptide from
amyloid precursor protein, which
281
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
has been proposed as a therapeutic target for Alzheimer's disease. By
analyzing BACE1 inhibition with
multiple molecules in our P7C3 series, we have found no correlation between
neuroprotective efficacy in our
in vivo models and BACE1 inhibition (data not shown).
Discussion
The results of a target-agnostic, unbiased screen of 1,000 chemically diverse,
drug-like compounds led
to the identification of an aminopropyl carbazole endowed with the capacity to
enhance adult neurogenesis.
This compound, designated P7C3, was found to act by blocking the death of
newborn neurons in the dentate
gyrus of adult mice. Here we have sought to answer a simple question. If P7C3
is capable of preventing the
death of newborn neurons during hippocampal neurogenesis in adult mice, this
compound may also prevent
death of existing neurons in animal models of neurodegenerative disease. More
specifically, we have
administered MPTP to mice as a means of killing dopamine neurons. Fully 24
hours after removal of the
toxin, mice were treated with varying doses of one of three compounds for a
period of three weeks. Thereafter
mice were sacrificed and assayed for evidence of neuroprotective efficacy. As
a second, related animal model
of neuron death, C. elegans worms were co-treated with MPP and varying doses
of P7C3, P7C3A20 and
Dimebon as a means of assessing neuroprotective activity.
The three compounds chosen for extensive testing, P7C3, its structurally
related analog P7C3A20, and
Dimebon, were selected by virtue of the knowledge that they demonstrate
distinct pro-neurogenic activities.
Among dozens of chemical analogs of P7C3 evaluated in the study first
reporting this category of pro-
neurogenic compounds, P7C3A20 displayed the highest potency and ceiling of pro-
neurogenic efficacy. In
addition to the P7C3 and P7C3A20 chemicals, we included Dimebon in the present
study for two reasons.
First, even though P7C3 and Dimebon both contain three-ring heterocycles,
Dimebon was found to display
significantly diminished levels of potency and efficacy relative to P7C3 in
our original study. Its pro-
neurogenic activity in the hippocampus of adult mice was observed only at
relatively high doses, and its
ceiling of efficacy was clearly diminished relative to both P7C3 and P7C3A20.
Likewise, when tested for its
ability to protect mitochondrial membrane integrity following exposure of
cultured cells to a calcium
ionophore, Dimebon exhibited a protective potency between 100- and 1,000-fold
lower than P7C3. Second,
Dimebon has been the subject of extensive clinical studies in both Alzheimer's
disease and Huntington's
disease. Despite early indications of efficacy in a phase 2 trial for
Alzheimer's disease (Doody R.S., et al.
(2008) Effect of Dimebon on cognition, activities of daily living, behaviour,
and global function in patients
with mild-to-moderate Alzheimer's disease: a randomised, double-blind, placebo-
controlled study. Lancet
372:207-215), Dimebon failed in two independent phase 3 trials. By testing the
properties of these three
compounds in the present study of neuroprotective efficacy, one having a very
favorable efficacy profile as a
pro-neurogenic chemical (P7C3A20), another having an intermediate efficacy
profile (P7C3), and a third
having a far more modest efficacy profile (Dimebon), we sought to determine
whether this hierarchy of
activities might be preserved.
282
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Encouragingly, we observe that the A20 chemical variant of P7C3 displays
significant neuroprotective
efficacy in the MPTP model of dopaminergic neuron cell death in both rodents
and worms. P7C3, the original
compound discovered in the unbiased screen of 1,000 drug-like chemicals for
pro-neurogenic activity,
displayed a lower level of neuroprotective activity in the mouse and worm
models of dopaminergic neuron
death than P7C3A20. By contrast, Dimebon showed no protective activity in
either assay. Although others
have recently demonstrated that treatment of an Alzheimer's disease mouse
model (TgCRND8 mice) with
Dimebon improves memory and lowers accumulation of insoluble A1342 in the
brain, Dimebon appears too
weakly active to afford neuroprotection in the models of Parkinson's disease
that we have utilized.
We conclude that P7C3 and P7C3A20 protect dopaminergic neurons from MPTP-
induced cell death
with a hierarchy of activity that is analogous to their abilities to protect
newborn hippocampal neurons from
cell death. If correct, this interpretation offers the possibility that the
relatively straightforward assay we have
employed to monitor the activities of hundreds of chemical variants of P7C3,
wherein adult neurogenesis is
monitored over a seven day period following direct administration of test
compounds into the adult mouse
brain, may represent a trusted surrogate for the refinement of drug-like
chemicals having broad
neuroprotective activity. Indeed, we hereby observe that an unbiased, blinded
analysis of nine analogs of
P7C3 confirms this correlation. Five of these molecules showed significant
efficacy in our standard in vivo
hippocampal neurogenesis assay, and these same five molecules also showed
significant neuroprotective
efficacy from MPTP-mediated neurotoxicity to dopaminergic neurons. The four
analogs that were inactive in
the in vivo neurogenesis likewise showed no efficacy in the in vivo MPTP
neurotoxicity assay. Taken
together, these results show that the relatively rapid evaluation of new
molecules in the in vivo neurogenesis
assay is predictive of their neuroprotective efficacy in the MPTP assay.
Ongoing SAR efforts with our P7C3 series of molecules using the in vivo
neurogenesis assay appears
to qualify as a rapid and accurate way to guide the refinement of this series
of molecules into a neuroprotective
drug for Parkinson's disease. In this context, several of the analogs shown in
Figure 33 represent potential
improvements to P7C3. We have been primarily concerned with the presence of an
aniline ring, as this
functionality can be associated with toxicity. Encouragingly, three of the
analogs (P7C3S7, -S41, and -S165)
lack aniline rings and display potency equal to or greater than P7C3 in both
in vivo assays. We further note
that P7C3 was originally identified as a racemic mixture, and that racemic
mixtures may require additional
characterization for clinical development. P7C3 S41 and P7C3 S165 address this
limitation because the former
is a single enantiomer while the latter lacks stereochemistry altogether.
Finally, we have sought to reduce the
polarity and molecular weight of these neuroprotective compounds. P7C3 S165 is
significantly lighter than
P7C3 (mw = 383 Da, vs 474 Da for P7C3) and, by virtue of the carboxylic acid,
substantially more polar.
These results suggest that it should be possible to further improve the
physical properties of these analogs in
efforts to optimize derivatives suitable for clinical testing.
283
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Consistent with the interpretation that the activity of P7C3 analogs in the in
vivo neurogenesis assay
correlates with neuroprotective efficacy in mature neurons are the results of
assays of P7C3, P7C3A20 and
Dimebon in a mouse model of amyotrophic lateral sclerosis. In this model,
using mice expressing a high level
of a human transgene encoding a mutated variant of the gene encoding human
Cu,Zn superoxide dismutase,
we have observed the same hierarchy of activities wherein P7C3A20 is active,
P7C3 is intermediately active
and Dimebon is inactive. If the more active variants of this class of
compounds indeed possess
neuroprotective properties, and if we can rely on the relatively rapid in vivo
assay of enhanced neurogenesis in
order to rank order compounds for structure-activity relationship (SAR)
scoring, it should be possible to
optimize variants with the goal of selecting an appropriately qualified
chemical to advance for human testing.
To date, no safely tolerated, neuroprotective chemical is available for the
treatment of any of a wide range of
neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease
and amyotrophic lateral
sclerosis. Based upon the observations reported herein, we propose that a
properly optimized variant of the
P7C3 class of pro-neurogenic, neuroprotective chemicals may offer promise for
the treatment of
neurodegenerative disease.
Materials and Methods
Approval for the animal experiments described herein was obtained by the
University of Texas
Southwestern Medical Center Institutional Animal Care and Use Committee.
Statistics: All p values were obtained with the Student's t test.
30 Day Survival Assay of Newborn Hippocampal Neurons: Because both social
activity and
voluntary exercise enhance hippocampal neurogenesis, mice were individually
housed without access to
running wheels throughout the entire procedure, beginning one week prior to
bromodeoxyuridine (BrdU,
Sigma-Aldrich) labeling of newborn cells. Throughout the study, mice had ad
libitum access to food and
water. BrdU was injected intraperitoneally at 150 mg/kg i.p., and 24 hours
later administration of test
compounds or vehicle was initiated. P7C3 and P7C3A20 were dissolved in 5%
dextrose (pH 7.0) with 2.5%
DMSO and 10% Cremaphor EL (Sigma, C5135). Dimebon was dissolved in normal
saline. Compounds were
compared to their respective controls, and were tested at 2.5, 5, 10 and 20
mg/kg twice daily (i.p.) for 30 days.
The injection site was alternated between right and left sides. Each group
consisted of six 12-week-old adult
male C57 B1/6 mice. Animals were monitored daily for general health and weight
loss. Cage changes were
performed per routine scheduling. After 30 days of compound administration,
mice were sacrificed by
transcardial perfusion with 4% paraformaldehyde at pH 7.4, and their brains
were processed for
immunohistochemical detection of incorporated BrdU in the dentate gyrus.
Dissected brains were immersed
in 4% paraformaldehyde overnight at 4 degrees Celsius, then cryoprotected in
sucrose before being sectioned
with a Leica SM2000R sliding microtome coronally into 40 [tM thick free-
floating sections. Unmasking of
BrdU antigen was achieved through incubating tissue sections for two hours in
50% formamide / 2X SSC at 65
284
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
degrees Celsius, followed by five minute wash in 2X SSC and subsequent
incubation for thirty minutes in 2M
HC1 at 37 degrees Celsius. Sections were processed for immunohistochemical
staining with mouse monoclonal
anti-BrdU (1:100, Roche). Diaminobenzidine was used as a chromagen, and tissue
was counter-stained with
hematoxylin to aid in visualization of the neuroanatomy. Images were analyzed
with a Nikon Eclipse 90i
motorized research microscope with Plan Apo lenses coupled with Metamorph
Image Acquisition software
(Nikon). Quantification of all staining was done blind to treatment group. The
number of BrdU+ cells in the
entire dentate gyrus was quantified by counting BrdU+ cells within the dentate
gyms in every fifth section
throughout the entire hippocampus and then normalizing for dentate gyms
volume.
P7C3-57, -S8, -S40, -S41, -S54, -S165 and -A20 were synthesized as previously
described.
P7C3-S184 was synthesized as previously described by Asso et al.(2008) alpha-
naphthylaminopropan-2-ol
derivatives as BACE1 inhibitors. ChemMedChem 3:1530-1534.
Pharmacokinetic Analysis: C57BL/6 mice treated with MPTP and then dosed IP
with compound for
21 days were utilized for pharmacokinetic (PK) analysis of total P7C3, P7C3A20
and Dimebon levels in
plasma and brain. In a separate set of experiments designed to test the
ability of new P7C3 analogs to cross
the blood brain barrier, C57BL/6 mice were dosed IP a single time with
compounds at 10 mg/kg. P7C3,
P7C3A20 and Dimebon were formulated for administration as described above.
Analogs were formulated in
5% Dextrose, pH 7.4, containing 5% DMSO and 10% Cremophor EL with the
exception of P7C3-58 which
required 10% DMSO and 20% Cremophor EL dissolved in 5% Dextrose for delivery.
Six hours after the final
compound dose, animals were given an inhalation overdose of CO2 and whole
blood and brain collected.
Plasma was prepared from blood and was stored along with the brain tissue at -
80 C until analysis. Brain
homogenates were prepared by homogenizing the tissues in a 3-fold volume of
PBS. Total brain homogenate
volume was estimated as volume of PBS added plus volume of brain in mL. One
hundred mL of either plasma
or brain homogenate was processed by addition of a two- or four-fold excess of
methanol or acetonitrile
containing formic acid and an internal standard (IS), N-benzylbenzamide (Sigma-
Aldrich, lot #02914LH) to
precipitate plasma or tissue protein and release bound drug. The final formic
acid concentration was 0.1%,
and the final IS concentration was 25 ng/ml. Extraction conditions were
optimized prior to PK analysis for
efficient and reproducible recovery over a three log range of concentrations.
The samples were vortexed 15
sec, incubated at room temp for 10' and spun 2 x 16,100 g in a standard
refrigerated microcentrifuge. The
supernatant was then analyzed by LC/MS/MS. Standard curves were prepared by
addition of the appropriate
compound to plasma or brain homogenate. A value of 3x above the signal
obtained in the blank plasma or
brain homogenate was designated the limit of detection (LOD). The limit of
quantitation (LOQ) was defined
as the lowest concentration at which back calculation yielded a concentration
within 20% of the theoretical
value and above the LOD signal. LOQ values for plasma and brain ranged from
0.5 to 500 ng/ml but were
well-below the concentrations measured at 6 hours for all of the compounds.
Compound levels were
285
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
monitored by LC/MS/MS using an AB/Sciex (Framingham, MA) 3200 Qtrap mass
spectrometer coupled to a
Shimadzu (Columbia, MD) Prominence LC. The compounds were detected with the
mass spectrometer in
MRM (multiple reaction monitoring) mode by following the precursor to fragment
ion transition 474.9 ¨>
337.8 for P7C3 (pos. mode; M+H ), 507.0 ¨> 204.1 for P7C3A20 (pos. mode; M+H
), 320.3 ¨> 277.3 for
Dimebon (pos. mode; M+H ), 381.9¨> 80.7 for P7C3-S165 (neg. mode; M-H+),
519.0¨> 338.0 for P7C3-S54
(pos. mode; M+H ), 536.0¨> 536.0 (redundant MRM) for P7C3-S7 (neg mode; M+
HC00-), 520.0¨> 520.0
for P7C3-S41 (neg mode; M+HC00-); 520.1¨> 520.1 for P7C3-S40 (neg mode; M+HC00-
), 477.1¨> 138.2
for P7C3-S8 (pos. mode; M+H ), 478.0¨> 153.2 for P7C3-S25 (pos. mode; M+H ),
and 435.2¨> 248.2 for
P7C3-S184 (pos. mode; M+H ). The IS, N-benzylbenzamide, was monitored using a
212.1 ¨> 91.1transition
(pos. mode; M+H ). An Agilent (Santa Clara, CA) XDB C18 column (50 X 4.6 mm, 5
micron packing) was
used for chromatography with the following conditions: Buffer A: dH20 + 0.1%
formic acid, Buffer B:
methanol + 0.1% formic acid, 0 ¨ 1. 5 min 0% B, 1.5 ¨ 2.5 min gradient to 100%
B, 2.5 ¨ 3.5 min 100% B,
3.5 to 3.6 min gradient to 0% B, 3.6 to 4.5 min 0% B. Chromatography
conditions were identical for all
compounds, except the initial and final concentration of Buffer B, which was
set to 0% for P7C3 and
P7C3A20 and 3% for Dimebon and all of the other P7C3 analogs.
Maintenance of C. elegans: C. elegans were grown at 20 degrees Celsius on
nematode growth
medium (NGM) agar in 60mm Petri plates according to standard protocols. Worms
were fed the Escherichia
coli nutrient-rich strain HB101. All experiments were performed using BZ555
[Pdat-1::GFP], obtained from
the Ceanorhabditis Genetics Center at the University of Minnesota, USA. BZ555
is an integrated transgenic
strain (chromosome IV) that expresses GFP under the control of the dopamine
neuron specific promoter dat-1.
To obtain first-stage synchronous larvae (Li 's), gravid adults were treated
with alkaline hypochlorite solution,
rinsed three times in M9 buffer, suspended in 6m1 of M9 and shaken for 12-
14hrs at room temperature,
according to standard protocols. Compound tests were performed in 500111
solution of PBS, compounds, and a
bacterial density of HB101 at 0D600 of 2 at 20 degrees Celsius in 12-well
plates (BD Falcon; Thermo Fisher
Scientific Inc.).
Assessment of MPTP-mediated neurotoxicity to murine SNc neurons: 15 adult male
C57B1/6
mice were individually housed for one week and then injected daily for 5 days
with 30 mg/kg/day (i.p.) free
base MPTP (Sigma). On day 6, 24 hours after receiving the fifth and final dose
of MPTP, daily treatment with
P7C3, P7C3A20, Dimebon or vehicle was initiated. Mice were housed in
disposable caging, and protective
gear and precautions were implemented for handling MPTP in accordance with UT
Southwestern Medical
Center policy. Dose response studies were conducted in which mice received
twice daily doses of each
compound (or vehicle) by intraperitoneal injection for the following 21 days,
after which mice were sacrificed
by transcardial perfusion with 4% paraformaldehyde. Brains were dissected,
fixed overnight in 4%
paraformaldehyde, and cryoprotected in sucrose for freezing by standard
procedures. Frozen brains were
286
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
sectioned through the striatum and SNc at 30 uM intervals, and every fourth
section (spaced 120 ,M apart)
was stained with antibodies directed against tyrosine hydroxylase (TH) (Abcam,
rabbit anti-TH, 1:2500).
Diaminobenzidine was used as a chromagen, and tissue was counter-stained with
hematoxylin to aid in
visualization of the neuroanatomy. Images were analyzed with a Nikon Eclipse
90i motorized research
microscope with Plan Apo lenses coupled with Metamorph Image Acquisition
software (Nikon). TH+
neurons were counted with Image J software (NIH) in every section by 2 blinded
investigators and results
were averaged and multiplied by the sectioning interval to determine the total
number of TH+ neurons per
SNc.
Assessment of MPP dopaminergic neuron toxicity in C. elegans: Synchronized Li
larvae were
plated into each well of a 12-well plate (approximately 400 larvae per well)
containing PBS, Vehicle or
compounds, with or without 5mM MPP iodide (Sigma) freshly diluted in PBS.
DMSO was used as vehicle
(VEH) and the concentration in treatment groups was maintained below 1%. The
assay solution (500m1) was
incubated for 40 hrs at 20 degrees Celsius. The worms were then washed in dH20
and supernatant was
aspirated. To examine dopaminergic neuron toxicity, worms were anesthetized
(0.1% tricaine, 0.01%
tetramizole) for 5 min and then transferred to microscope slides and
coverslipped. Pictures were taken at 40X
magnification (AMG, Evos fl microscope). Each experiment was conducted in
triplicate with 10-20 worms
counted per condition. For quantification, investigators were blind to
treatment condition. Quantification was
done by observing all four cephalic sensilla (CEP) dendrites, per standard
protocol. Briefly, GFP fluorescence
was visualized from the nerve ring to the tip of the nose, and if any portion
of a dendrite was absent then it was
counted as being degenerated.
Locomotion analysis of C. elegans: A video-based assay was used to assess the
swim speed,
distance traveled and length of worms. After exposure to MPP for 32 hrs,
worms were washed, resuspended
in M9 buffer (500u1) and transferred to microscope slides. A 10 second movie
was recorded at 4X
magnification using a Nikon Eclipse 80i microscope. Each movie consisted of
160 frames and the distance
traveled by the head of each worm was manually tracked in each frame using
Imera software. This software
was also used to measure the length of the worm body. The ratio of movement
distance to body length was
used as a movement index, and defined as locomotion per standard protocols.
Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of
amyotrophic lateral sclerosis
Amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig's disease, is a
relatively rare, adult-
onset, rapidly progressive and fatal disease that involves degeneration of
spinal cord motor neurons (Tandan
R, Bradley W.G. (1985) Amyotrophic lateral sclerosis. Part 1. Clinical
features, pathology, and ethical issues
in management. Ann Neurol 18:271-280). This disorder causes muscle weakness
and atrophy throughout the
body, and patients with ALS ultimately lose all voluntary movement. The
earliest parts of the body affected in
287
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
ALS reflect those motor neurons that are damaged first. Regardless of the
region of onset, however, muscle
weakness and atrophy invariably spread to other parts of the body as the
disease progresses. Although disease
progression varies between individuals, most patients are eventually unable to
stand or walk, get in or out of
bed on their own, or use their hands and arms. Difficulty with chewing,
swallowing and breathing leads to
progressive weight loss and increased risk of choking and aspiration
pneumonia. Towards the end stages of
disease, as the diaphragm and intercostal muscles weaken, most patients
require ventilator support.
Individuals with ALS most commonly die of respiratory failure or pneumonia
within 2-5 years of diagnosis.
There are no current treatments for ALS.
Approximately 20% of inherited cases of ALS, and 3% of sporadic cases, are
associated with
autosomal dominant mutations in the SOD] gene on chromosome 21, and about 150
different mutations
dispersed throughout the gene have been identified thus far. SOD] encodes
cytosolic Cu/Zn superoxide
dismutase, an antioxidant enzyme that protects cells by converting superoxide
(a toxic free radical generated
through normal metabolic activity of mitochondria) to hydrogen peroxide.
Unchecked, free radicals damage
both mitochondrial and nuclear DNA, as well as proteins within cells. In ALS
linked to mutations in SOD],
cytotoxicity of motor neurons appears to result from a gain of toxic SOD1
function, rather than from loss of
dismutase activity. Although the exact molecular mechanisms underlying
toxicity are unclear, mutation-
induced conformational changes in SOD1 lead to misfolding and subsequent
aggregation of mutant SOD1 in
cell bodies and axons. Aggregate accumulation of mutant SOD1 is thought to
disrupt cellular functions and
precipitate neuron death by damaging mitochondria, proteasomes, protein
folding chaperones, or other
proteins.
Transgenic animal models of mutant SOD1, such as G93A-SOD1 mutant mice, are
currently used for
research into the pathogenic mechanisms thought to broadly underlie ALS. Mice
hemizygous for the G93A-
SOD1 transgene express 18 +/- 2.6 copies of a form of SOD] found in some
patients with inherited ALS (a
substitution of glycine to alanine at codon 93). This was the first mutant
form of SOD] to be expressed in
mice, and is the most widely used and well-characterized mouse model of ALS.
Superoxide dismutase activity
in these mice is intact, and the pathogenic effect of the mutant transgene
appears to be gain of function, as is
thought to occur in human patients. Death of motor neurons in these mice
occurs in the ventral horn of the
spinal cord and is associated with paralysis and muscle atrophy. Around 100
days of age, G93A-SOD1 mice
characteristically experience the onset of paralysis in one or more limbs, due
to loss of spinal cord motor
neurons. Paralysis spreads rapidly throughout the body, culminating in death
of 50% of the mice within seven
weeks of disease onset.
We have previously reported the identification of a proneurogenic,
neuroprotective aminopropyl
carbazole (P7C3) discovered through a target-agnostic in vivo screen of
postnatal hippocampal neurogenesis
(Pieper et al. (2010) Discovery of a Proneurogenic, Neuroprotective Chemical.
Cell 142:39-51). Prolonged
administration of P7C3 to mice suffering from pathologically high levels of
neuronal apoptosis in the dentate
288
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
gyrus safely restored hippocampal structure and function with no observable
physiologic side effects.
Furthermore, extended administration of P7C3 to aged rats impeded hippocampal
cell death and preserved
cognitive ability as a function of terminal aging.
We have synthesized and characterized a variant of P7C3, known as P7C3A20,
which has greater
potency and pro-neurogenic efficacy than the parent compound. P7C3A20 differs
structurally by replacement
of the hydroxyl group at the chiral center of the linker with a fluorine, and
the addition of a methoxy group to
the aniline ring. P7C3A20 also displays a more favorable toxicity profile than
P7C3, with no hERG channel
binding, histamine receptor binding or toxicity to HeLa cells. We have also
found that Dimebon, an
antihistaminergic drug that is chemically related to P7C3 and reported to have
anti-apoptotic and
mitochondrial protective properties, displays modest efficacy in the same
biologic assays employed to discover
and characterize P7C3 and P7C3A20. However, it does so with substantially less
potency and ceiling of
efficacy (CoE).
Armed with three related chemicals, one having very high pro-neurogenic
activity (P7C3A20), one
having intermediate activity (P7C3), and one having only modest activity
(Dimebon), we initiated efficacy
studies in two animal models of neurodegenerative disease. We report above
evidence of significant
neuroprotective activity of P7C3A20 in a rodent model of Parkinson's disease
(PD). P7C3 exhibited
intermediate activity in the PD animal model, and Dimebon showed no evidence
of efficacy. The correlative
activities of chemicals tested in the neurogenesis and PD assays were extended
to eight additional analogs of
P7C3. In every case, derivatives of P7C3 that were active in the neurogenesis
assay were also active in the
animal model of PD, and inactive variants were inactive in both assays.
Here, we have employed the same approach to score the activities of P7C3A20,
P7C3 and Dimebon in
a model of neuron death outside of the brain. To address this question, we
utilized G93A-SOD1 mutant mice,
a model of amyotrophic lateral sclerosis (ALS) characterized by spinal motor
neuron death associated with
decreased motor functioning. As was observed for the rodent model of PD, we
hereby report robust activity
of P7C3A20 in the G93A-SOD1 mouse model of ALS, intermediate activity for
P7C3, and no activity for
Dimebon.
Results
Efficacy of early administration of P7C3 to G93A-SOD1 mutant mice before
disease onset.
As an initial test of efficacy in this disease model, we intraperitoneally
administered P7C3 to female
G93A-SOD1 transgenic mice using a treatment paradigm of 20 mg/kg/day P7C3,
with vehicle administered to
siblings, starting at 40 days of age. This treatment scheme was selected based
on standard protocols for initial
proof of concept screens in G93A-SOD1 mutant mice. To control for transgene
copy number, mice were
289
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
sibling matched between treatment groups, as per standard protocol, and
quantitative PCR was performed to
ensure that the copy number was maintained within the normal range. After
initiation of P7C3 or vehicle
treatment, date of onset of illness was determined by peak weight, and initial
progression of disease was
defined as the day at which mice fell to 10% below their maximum weight. Mice
were also assessed daily by
a standard measure of neurological severity score ranging from 0-4, with a
higher number reflecting greater
neurologic impairment. In addition to weight loss, a score of 2 or greater for
two consecutive days was also
employed as an indication of disease progression.
P7C3 treatment slowed disease progression in G93A-SOD1 mice, as indicated by
delaying the time
point at which mice dropped to 10% below their maximum weight. Treatment with
P7C3 also delayed the age
at which G93A-SOD1 mice advanced to a neurological severity score of 2.
Furthermore, P7C3 treatment
improved performance in the accelerating rotarod task as a function of disease
progression, indicating a
slowing of progression of motor impairment. This effect of slowing disease
progression did not translate into
increased survival of the animals, which is consistent with other
interventions that have ameliorated disease
symptoms in rodent models of ALS without improving survival.
Comparison of the efficacy of administration of P7C3A20, P7C3 and Dimebon at
disease onset for blocking
spinal motor neuron cell death in G93A-SOD1 mutant mice.
Based on the promising results of early (day 40) administration of P7C3 to
G93A-SOD1 mutant mice,
we next sought to determine whether P7C3A20, P7C3 or Dimebon could protect
ventral horn spinal motor
neurons when administered at the expected time of disease onset (day 80). We
initiated administration of
either P7C3, P7C3A20 or Dimebon, each at a dose of 20 mg/kg/day, and analyzed
motor neuron cell survival
by staining lumbar spinal sections for choline acetyltransferase (ChAT). ChAT,
the enzyme that synthesizes
the neurotransmitter acetylcholine, serves as a marker for spinal cord motor
neurons. All sections were
counted blind to treatment group in order to quantify motor neuron survival,
and five mice for each treatment
group were analyzed at 90, 100, 110 and 120 days. Each treatment group was
compared to its own sibling-
matched group that received the corresponding vehicle.
As shown in Figure 34, the wild type bar represents the average number of
spinal motor neurons in
110 day old, vehicle-treated, wild-type littermate mice. Because survival of
motor neurons did not differ
between the various vehicle treatment groups within any given time point, the
results were combined for ease
of presentation. For animals expressing the G93A-SOD1 transgene, the number of
spinal cord motor neurons
steadily declined between days 90 and 120 (Figure 34). At every time point,
treatment with P7C3A20
provided significant protection from spinal motor neuron cell death (Figure
34). Treatment with Dimebon
revealed a rate of motor neuron loss indistinguishable from vehicle treatment
groups. P7C3, by contrast,
290
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
provided intermediate protection on days 100 (p=.048) and 110 (p=.01). By the
time mice reached 120 days of
age, however, the P7C3-treated group showed the same degree of motor neuron
cell loss as vehicle and
Dimebon-treated groups. Representative immunohistological staining of spinal
cord sections is shown in
Figure 34 from each of the five mice examined on day 110. Taken together,
these results demonstrate that
daily administration of P7C3A20 starting at disease onset effectively blocks
spinal motor neuron cell death in
G93A-SOD1 mutant mice. P7C3 was active by these measures, but to a lesser
extent than P7C3A20, whereas
Dimebon was completely devoid of neuroprotective activity.
Comparison of the efficacy of administration of P7C3A20, P7C3 and Dimebon at
disease onset for preserving
rotarod performance in G93A-SOD1 mutant mice.
Having observed evidence of compound-mediated protection of spinal cord motor
neurons, we next
sought to determine whether motor performance might also be protected in these
mice. Motor performance
was monitored by the accelerating rotarod task standardly employed for
evaluation of rodent models of ALS.
We again initiated administration of P7C3, P7C3A20 or Dimebon on day 80, at 20
mg/kg/day, starting with no
less than 20 mice per treatment group. Each animal in each group had its own
sibling-matched vehicle control,
and testing was conducted blind to treatment group. Rotarod training was
initiated on day 50 for 2 days, and
repeated weekly testing was conducted every 7 days thereafter. Each mouse was
subjected to four trials of 600
seconds each, with a 20 minute recovery break between each trial. The latency
time to fall was averaged
across all 4 trials.
As shown in Figure 35, performance in all treatment groups was equal at weeks
10 and 11. Treatment
with the test compounds was initiated midway between weeks 11 and 12, on day
80, and at week 12 there were
no significant differences between groups. By week 13, however, P7C3A20-
treated mice showed significantly
better performance (p=.019) than the corresponding vehicle treatment group. In
subsequent weeks, both P7C3
and Dimebon, as well as all vehicle groups, continued to decline at a steady
pace in performance in this task,
with P7C3A20-treated mice performing significantly better at each time point.
Rotarod data were not
collected beyond week 16 because too few animals survived to this time point
for valid comparison across
groups.
As noted with early initiation of administration (day 40) of P7C3, this
intervention improved rotarod
performance but did not extend survival of the mice. Also, despite improvement
in rotarod performance in
P7C3A20-treated mice when daily treatment was initiated on day 80, we did not
observe any delay in other
measures of disease progression (neurological score or weight loss). This
observation may reflect the
increased challenge for efficacy associated with administering compounds at
the time of disease onset. Taken
together, our results show that administration at the time of disease onset of
the most potent member of the
291
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
P7C3 series of neuroprotective drugs, P7C3A20, significantly improves
performance of G93A-SOD1 mice in
the accelerating rotarod test. Both P7C3 and Dimebon were insufficiently
active to preserve motor function in
the accelerating rotarod task at 20 mg/kg/day when administration was
initiated at the time of disease onset.
Comparison of the efficacy of administration of P7C3A20, P7C3 and Dimebon at
disease onset for preserving
walking gait in G93A-SOD1 mutant mice.
Analysis of walking gait offers a second means of assessing motor limb
strength and coordination in
rodent models of ALS. We conducted this analysis in the same mice used for the
accelerating rotarod task, at
three time points: 90, 118 and 132 days. Briefly, the front paws of each test
mouse were dipped in orange
tempera paint, and the back paws in blue tempera paint. Mice were then
directed into a bisected PVC-tube
placed on top of artists easel paper, such that the mouse was prompted to walk
through the tunnel for a
distance of 30 inches, leaving a trail of pawprints on the paper. Key
parameters of the pawprints were then
manually measured, as described in Methods. These parameters included front
and back stride length, front
and back width, and front-to-back paw distance (Figure 36A).
Twenty total measurements (10 on each side) for each parameter were recorded
per mouse, and 20
mice per group were evaluated at the 90 and 118 day time points. All
measurements were conducted blind to
treatment group. Front and back widths showed no difference as a function of
treatment group or disease
progression until day 132, at which point P7C3A20 treatment was observed to
preserve back width. Three of
the measured parameters (back stride, back-front distance and front stride)
showed significant improvement as
a function of treatment with P7C3A20 earlier in the disease process, whereas
none of these parameters in the
walking gait analysis at 20 mg/kg/day were significantly improved by treatment
with P7C3 or Dimebon
(Figure 36B).
Back stride is defined as the distance between each successive back paw print
on a single side, and one
of the first features of disease in G93ASOD1 mutant mice is the onset of hind
limb muscle weakness. As the
disease progresses, mice are unable to move their hind limbs as much with each
step, and back stride distance
decreases. This was evident on day 118, in which P7C3, Dimebon and all vehicle
treatment groups showed
reduced back stride length (Figure 36B). Back stride measure was significantly
(p=.0016) preserved to a near-
normal level in P7C3A20-treated mice (Figure 36B). Front stride is analogously
defined as the distance
between each successive front paw print on a single side, and as the disease
progresses this measure also
shortens as a consequence of the reduced hind limb stride that prevents the
mouse from moving as great a
distance with each step. Compromised front stride length thus confirms the
deficit associated with back stride
length, and we observed that on day 118 this measure was indeed reduced in
P7C3, Dimebon and all vehicle
treatment groups, yet preserved to almost normal levels in P7C3A20-treated
mice (Figure 36B).
292
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
On day 132, there were insufficient numbers of mice in the P7C3-VEH and
Dimebon-VEH groups
that could participate in the task, due to complete paralysis of one or more
limbs in the majority of the original
test group. In the A20-VEH group, however, there were still ten P7C3A20 mice
that were able to walk across
the paper. Here, we observed that improvements in back stride and front stride
were preserved, but there was
no longer a difference in back-front distance. Back-front distance is defined
as the distance between a back
pawprint and the front pawprint on the same side. Early on, as the disease
progresses in this animal model of
ALS, the back-front distance steadily increases because the front limbs are
able to extend normally, but the
hindlimbs are not strong enough to formulate proper steps that should result
in the back paw landing on top of
the front paw print. It is evident in Figure 36B that as assayed on day 118,
treatment with P7C3A20 attenuated
this increase in back-front distance. On day 132, however, the differences
between VEH and P7CA20 treated
mice in back-front distance were lost. At this stage, the disease was
sufficiently advanced that this measure
reflects the additional complication of front limb weakness, such that the
mice were unable to extend their
front limbs normally. As a result, the back-front distance declined, and there
were no differences between
P7C3A20 and its sibling-matched vehicle group. Taken together, our results of
gait analysis demonstrate that
treatment with P7C3A20 at the time of disease onset helps preserve walking
gait in the G93A-SOD1 mouse
model of ALS.
Analysis of plasma, brain and spinal cord levels of P7C3, P7C3A20 and Dimebon.
LC/MS/MS quantification of brain and blood levels of P7C3, P7C3A20 and Dimebon
confirmed that
all three compounds were able to enter both the brain and spinal cord (Figure
37). Notably, P7C3A20
displayed significantly greater protective efficacy compared to the other two
compounds, despite the fact that
P7C3A20 accumulated in spinal cord tissue at less than one-twentieth the
concentration of P7C3. Dimebon,
which displayed no protective efficacy in G93A-SOD1 mice, showed comparable
levels of spinal cord
accumulation to P7C3A20. These results parallel findings observed in
evaluation of the neuroprotective
efficacy of these same three compounds in MPTP-treated mice.
Discussion
The results of an unbiased screen of 1,000 chemically diverse, drug-like
compounds led to the
identification of an aminopropyl carbazole endowed with the capacity to
enhance adult neurogenesis. This
compound, designated P7C3, was found to act by blocking the death of newborn
neurons in the dentate gyms
of adult mice. We have also found that P7C3, P73A20 and other active analogs
protect dopamingergic
neurons of the substantia nigra from MPTP-induced neurotoxicity. Here we have
sought to determine whether
this class of pro-neurogenic compounds might also block nerve cell death
outside of the brain.
293
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
We selected P7C3, P7C3A20 and Dimebon for testing because they display
distinct levels of pro-
neurogenic, neuroprotective activity when assayed for protection from
apoptotic cell death of either newborn
hippocampal neurons or following MPTP-toxicity to mature dopaminergic neurons.
P7C3A20 displays the
highest potency and ceiling of efficacy amongst these three molecules. We
evaluated Dimebon because of
extensive studies in human clinical trials, and its relative similarity in
chemical structure to P7C3. When
tested for its ability to protect mitochondrial membrane integrity following
exposure of cultured cells to a
calcium ionophore, Dimebon exhibited a protective potency between 100- and
1,000-fold lower than P7C3.
Similarly modest activity was observed when Dimebon was assayed in our
standard model of hippocampal
neurogenesis. The reduced potency and efficacy of Dimebon has been further
revealed in its inability to
protect dopaminergic neurons in the substantia nigra from MPTP-toxicity.
Finally, Dimebon has been
extensively studied in human clinical trials of both Alzheimer's disease and
Huntington's disease. Although
early indications in a phase 2 trial suggested that Dimebon might be
efficacious for Alzheimer's disease, the
drug failed in two independent phase 3 trials. By testing the properties of
these three related compounds in the
present study of protective efficacy in an animal model of ALS, we sought to
determine whether the hierarchy
of activities amongst these three molecules might be preserved.
Encouragingly, we observe that P7C3A20 significantly blocks death of spinal
motor neurons in the
G93A-SOD1 mouse model of ALS. Importantly this protective effect is observed
when administration of the
compound is initiated at the time of disease onset, and it correlates with
preservation of muscle strength and
coordination as assessed through the accelerating rotarod test and analysis of
walking gait. P7C3 offered
intermediate protection from cell death when administered at the time of
disease onset. Administration of
P7C3 for a prolonged period of time by initiating treatment much earlier (day
40) did preserve motor function
as assayed by the accelerating rotarod task. Dimebon offered no protection in
any of these measures.
Although efficacy of Dimebon in an animal model of Alzheimer's disease
(TGCRND8) mice has recently
been reported, this drug appears too weakly active to afford any protection in
the G93A-SOD1 mutant mouse
model of ALS.
We conclude that P7C3A20 and P7C3 display a hierarchy of activities analogous
to their abilities to
protect newborn hippocampal neurons from cell death, to block MPTP-mediated
killing of mature
dopaminergic neurons in the substantia nigra, and to protect spinal motor
neurons from dying in G93A-SOD1
mutant mice. These collective observations give evidence that the relatively
straightforward assay of
monitoring adult hippocampal neurogenesis over a seven day period following
direct administration of new
analogs of P7C3 into the adult mouse brain may represent a trusted surrogate
for the refinement of drug-like
chemicals having neuroprotective activity. Over the past two years we have
conducted a comprehensive
structure-activity relationship (SAR) study in order to improve the chemical
scaffold of the P7C3 series of
molecules. To date, we have synthesized over 250 analogs of P7C3, all of which
have been evaluated in the in
vivo hippocampal neurogenesis assay. Our goal in these efforts to foster the
discovery of a neuroprotective
294
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
drug is to maximize neuroprotective efficacy and alleviate real or perceived
vulnerabilities in the chemical
structures. These efforts include, but are not limited to, eliminating the
carbazole bromines, eliminating the
aniline ring, increasing biologic activity, decreasing lipophilicity,
eliminating any toxicities including hERG
channel binding, increasing solubility and reducing molecular weight. By use
of the in vivo hippocampal
neurogenesis assay, these ongoing SAR efforts with P7C3 analogs may offer an
effective way to guide
optimization of this series of molecules towards a neuroprotective drug
candidate.
No safely tolerated, neuroprotective chemical is available for the treatment
of any form of
neurodegenerative disease, including Parkinson's disease, Alzheimer's disease
and amyotrophic lateral
sclerosis. Based upon the observations reported herein, we propose that a
properly optimized variant of the
P7C3 class of pro-neurogenic, neuroprotective chemicals may represent a viable
candidate for the treatment of
neurodegenerative disease.
Materials and Methods
Approval for the animal experiments described herein was obtained by the
University of Texas
Southwestern Medical Center Institutional Animal Care and Use Committee.
Statistics: All p values were obtained with the Student's t test, by comparing
treatment groups to their
individual sibling-matched vehicle treatment groups.
Analysis of motor neuron survival in the spinal cord: After transcardial
perfusion with 4%
paraformaldehyde (PFA), lumbar spinal cord was dissected and post-fixed
overnight in 4% PFA, cryo-
protected in 30% sucrose at 4 degrees Celsius, and then embedded in OCT and
sectioned on a Thermo-Fisher
cryostat (HM550) at 30 [LM thickness. Every seventh section was
immunohistochemically stained with goat
anti-choline acetyltransferase (ChAT) (Millipore). Briefly, sections were
incubated in 1% H202 for 45
minutes at room temperature, rinsed in tris-buffered saline (TBS), treated
with 0.1% Triton-TBS and then
blocked for 60 minutes in 3% BSA, 5% donkey serum, 0.3% triton-100 in TBS.
Sections were then incubated
in goat anti-ChAT (1:100) in the same blocking solution overnight at 4 degrees
Celsius. The next day,
sections were rinsed in TBS and incubated with donkey anti-goat biotin (1:200,
Jackson Immune). Signal was
amplified with an ABC kit from Vector Labs, and diaminobenzidine was used as a
chromagen.
Immunostained tissue was then photographed at 4X using a Nikon Eclipse 90i
motorized microscope, and the
number of ChAT-positive neurons was counted in a blinded manner by two
investigators, followed by
normalization for ventral horn volume.
Rotarod: Beginning on day 50, mice were trained on the accelerating rotarod
using Colombia
Instruments Rotamex-5. Training consisted of mice being placed on a rotarod
moving at 5 rpm for 300
seconds. Mice were trained to stay on the rotarod for the entire 300 seconds.
If a mouse fell, it was placed
295
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
back on the rotarod and the 300 second trial was started again. Training took
place on two consecutive days.
On day 52, mice ran their first full rotarod test, as described in Current
Protocols for Neuroscience. The
rotarod began at 4 rpm and accelerated to 40 rpm over 600 seconds, increasing
by 1.25 rpm every 20 seconds.
The time to fall was automatically recorded. Each run was separated by 20
minutes to allow the mice to rest,
and each mouse participated in 4 runs. Mice were run every seven days until
the time at which they were
unable to stay on the rotarod for more than 10 seconds for 3 trials.
Pawprint analysis: Five measurements were taken: front and back stride, front
and back width, and
front to back distance, as described in Current Protocols for Neuroscience. 20
total measures were taken for
each measurement. Front and back stride were collected as a straight line from
paw print to the following paw
print. Front to back distance was collected as a straight line from back paw
print to corresponding front paw
print. Correspondence was based on closest front footprint. Width from paw
print was measured by drawing a
line at a 90 degree angle from the line connecting the stride previous and the
paw being analyzed. The distance
was recorded as length of line from paw to the stride line opposite the paw
print. Mice attaining a score of
three were eliminated from analysis as measurements could not be taken for a
foot not being used in forward
motion.
Pawprints were recorded at 90, 118 and 132 days of age. A 6 inch by 42 inch
PVC pipe cut in half
lengthwise was placed on top of a piece of easel paper (27"x30 1/4"). Each
mouse had paws covered in non-
toxic tempera paint (orange front, blue back) and was placed at one end of the
pipe. The mouse ran quickly to
the other end of the pipe when released, and the procedure was repeated until
10 clear back and front prints
were made for each side while the subject was running. Pawprints were scanned
using a hand scanner, and
then visualized for measurement in Nikon Metamorph software. Measurements were
based on established
guidelines.
Neurological scoring: Neurological score was performed every day starting with
compound treatment
at 80 days, and was determined as follows: '0' = full extension of hind legs
away from lateral midline when
the test mouse was suspended by its tail, and could hold this for 2 seconds,
suspended 2-3 times; '1' = collapse
or partial collapse of leg extension towards lateral midline (weakness) or
trembling of hind legs during tail
suspension; '2' = toes curl under at least twice during walking of 12 inches,
or any part of foot drags along
cage bottom / table; '3' = rigid paralysis or minimal joint movement, foot not
being used for forward motion;
and '4' = mouse cannot right itself within 30 seconds from either side. Upon
reaching a score of 2, animals
were given a fresh Petri dish with wet food in the dish daily. When mice
achieved a score of 4 for two
consecutive days they were euthanized.
Weight data: Mice were weighed daily starting on the day of initiation of
compound administration in
order to assess disease progression and readjust compound dosage. A digital
balance with a step of .01 g was
used, and mice were placed in a small plastic container on the scale when
weighed. Weight was taken between
11 am and 1 pm every day.
296
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
Quantitative PCR: Quantitative PCR done in accordance with guidelines
established by Jackson
Laboratory protocol for SOD1-G93A mice.
Synthesis and Preparation of P7C3A20: Preparation of compound as described
above.
Pharmacokinetic Analysis of P7C3, P7C3A20 and Dimebon: Analysis of compound as
described
above.
OTHER EMBODIMENTS
This application claims the benefit of U.S. Provisional Application No.
61/143,755, which is
incorporated herein by reference in its entirety. The disclosure of U.S.
Provisional Application No.
61/143,755 includes, but is not limited to:
methods for promoting postnatal mammalian neurotrophism in a patient
determined to be in need thereof,
comprising administering to the patient an effective amount of a neurotrophic
carbazole compound of
formula 1:
R4 R5
R3 R6
R2
. 10 R7
N
Ri R8
NRi9Rii
OR9
wherein:
RI ¨ Rg are each independently selected hydrogen, heteroatom, heteroatom
functional group, and
optionally-substituted, optionally heteroatom lower (C1-C6) alkyl;
R9 is hydrogen or optionally-substituted, optionally heteroatom lower (C1-C6)
alkyl; and
R10 and R11 are each independently selected hydrogen, optionally-substituted,
optionally heteroatom Cl -
C6 alkyl, optionally-substituted, optionally heteroatom C2-C6 alkenyl,
optionally-substituted, optionally
heteroatom C2-C6 alkynyl, and optionally-substituted, optionally heteroatom C6-
C14 aryl, including
tautomers, stereoisomers and pharmaceutically-acceptable salts thereof
Unless otherwise noted, all structures depicted herein encompass
interconvertable tautomers as if each
were separately depicted.
297
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The presently disclosed embodiments encompass all alternative combinations of
particular embodiments:
-wherein R1 ¨ Rg are each independently selected hydrogen and halide;
- wherein RI, R2, R4, R5, R7 and Rg are hydrogen, and R3 and R6 are halide,
such as Cl, Br, I and F;
- wherein R9 is hydrogen;
- wherein R10 is hydrogen and R11 is optionally-substituted, optionally
heteroatom C6-C14 aryl;
- wherein R10 and R11 are joined to form a 5-7 membered, optionally
substituted heterocyclic ring;
- wherein R10 and R11 are joined to form an optionally substituted
pyrrolidine or a piperidine;
- wherein R10 is hydrogen and R11 is substituted phenyl, such as halide-or
C 1-C6 alkoxy-phenyl, including
para-, meta-, or ortho positions;
- wherein R10 is hydrogen and R11 is napthyl;
- wherein the compound has a formula of Table 1 (herein) or Table 2
(herein);
- wherein the compound has formula 2:
Br Br
41 10
N
N
H
OH
-wherein (a) at least one of R1 ¨ Rg is heteroatom, optionally-substituted, or
optionally heteroatom
lower (C1-C6) alkyl, and at least one of RI-R.4 or at least one of R5-R8 is
different; or (b) R9 is optionally-
substituted, optionally heteroatom lower (C 1-C6) alkyl;
-further comprising the step of detecting a resultant neurotrophism,
particularly neurogenesis; and/or
- further comprising the antecedent step of determining that the patient
has aberrant neurotrophism,
particularly aberrant neurogenesis, particularly aberrant hippocampal and/or
hypothalamic neurogenesis, or a
disease or disorder associated therewith, particularly by detecting and/or
diagnosing the same.
The presently disclosed embodiments also provide novel pharmaceutical,
particularly novel
neurogenic, compositions in unit dosage comprising a disclosed neurotrophic
carbazole not previously known
or suggested to provide pharmacological, particularly neurogenic, activity, or
a pharmaceutically-acceptable
salt thereof, and a pharmaceutically acceptable excipient.
The presently disclosed embodiments also provide disclosed novel neurotrophic
carbazoles and
pharmaceutically-acceptable salts thereof
298
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
U.S. Provisional Application No. 61/143,755 further discloses:
The term "heteroatom" as used herein generally means any atom other than
carbon, hydrogen or
oxygen. Preferred heteroatoms include oxygen (0), phosphorus (P), sulfur (S),
nitrogen (N), silicon (S),
arsenic (As), selenium (Se), and halogens, and preferred heteroatom functional
groups are hatoformyl,
hydroxyl, aldehyde, amine, azo, carboxyl, cyanyl, thocyanyl, carbonyl, halo,
hydroperoxyl, imine, aldimine,
isocyanide, iscyante, nitrate, nitrite, nitrite, nitro, nitroso, phosphate,
phosphono, sulfide, sulfonyl, sulfo, and
sulfhydryl.
The term "alkyl," by itself or as part of another substituent, means, unless
otherwise stated, a straight
or branched chain, or cyclic hydrocarbon radical, or combination thereof,
which is fully saturated, having the
number of carbon atoms designated (i.e. C1-C8 means one to eight carbons).
Examples of alkyl groups include
methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl,
cyclohexyl, (cyclohexyl)methyl,
cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-
heptyl, n-octyl and the like.
The term "alkenyl", by itself or as part of another substituent, means a
straight or branched chain, or
cyclic hydrocarbon radical, or combination thereof, which may be mono- or
polyunsaturated, having the
number of carbon atoms designated (i.e. C2-C8 means two to eight carbons) and
one or more double bonds.
Examples of alkenyl groups include vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-
(butadienyl), 2,4-pentadienyl,
3-(1,4-pentadienyl) and higher homologs and isomers thereof
The term "alkynyl", by itself or as part of another substituent, means a
straight or branched chain
hydrocarbon radical, or combination thereof, which may be mono- or
polyunsaturated, having the number of
carbon atoms designated (i.e. C2-C8 means two to eight carbons) and one or
more triple bonds. Examples of
alkynyl groups include ethynyl, 1- and 3-propynyl, 3-butynyl and higher
homologs and isomers thereof
The term "alkylene" by itself or as part of another substituent means a
divalent radical derived from
alkyl, as exemplified by -CH2-CH2-CH2-CH2-. Typically, an alkyl (or alkylene)
group will have from 1 to 24
carbon atoms, with those groups having 10 or fewer carbon atoms being
preferred in the presently disclosed
embodiments. A "lower alkyl" or "lower alkylene" is a shorter chain alkyl or
alkylene group, generally having
eight or fewer carbon atoms.
The terms "alkoxy," "alkylamino" and "alkylthio" (or thioalkoxy) are used in
their conventional sense,
and refer to those alkyl groups attached to the remainder of the molecule via
an oxygen atom, an amino group,
or a sulfur atom, respectively.
The term "heteroalkyl," by itself or in combination with another term, means,
unless otherwise stated,
a stable straight or branched chain, or cyclic hydrocarbon radical, or
combinations thereof, consisting of the
stated number of carbon atoms and from one to three heteroatoms selected from
the group consisting of 0, N,
Si and S, wherein the nitrogen and sulfur atoms may optionally be oxidized and
the nitrogen heteroatom may
optionally be quaternized. The heteroatom(s) 0, N and S may be placed at any
interior position of the
heteroalkyl group. The heteroatom Si may be placed at any position of the
heteroalkyl group, including the
299
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
position at which the alkyl group is attached to the remainder of the
molecule. Examples include -CH2-CH2-0-
CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, -CH2-S-CH2-CH3, -CH2-CH2,-S(0)-CH3,
-CH2-CH2-S(0)2-
CH3, -CH=CH-O-CH3, -Si(CH3)3, -CH2-CH=N-OCH3, and -CH=CH-N(CH3)-CH3. Up to two
heteroatoms
may be consecutive, such as, for example, -CH2-NH-OCH3 and -CH2-0-Si(CH3)3.
Similarly, the term "heteroalkylene," by itself or as part of another
substituent means a divalent radical
derived from heteroalkyl, as exemplified by -CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-
CH2-NH-CH2-. For
heteroalkylene groups, heteroatoms can also occupy either or both of the chain
termini (e.g., alkyleneoxy,
alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further,
for alkylene and heteroalkylene
linking groups, no orientation of the linking group is implied.
The terms "cycloalkyl" and "heterocycloalkyl", by themselves or in combination
with other terms,
represent, unless otherwise stated, cyclic versions of "alkyl" and
"heteroalkyl", respectively. Accordingly, a
cycloalkyl group has the number of carbon atoms designated (i.e., C3-C8 means
three to eight carbons) and
may also have one or two double bonds. A heterocycloalkyl group consists of
the number of carbon atoms
designated and from one to three heteroatoms selected from the group
consisting of 0, N, Si and S, and
wherein the nitrogen and sulfur atoms may optionally be oxidized and the
nitrogen heteroatom may optionally
be quaternized. Additionally, for heterocycloalkyl, a heteroatom can occupy
the position at which the
heterocycle is attached to the remainder of the molecule. Examples of
cycloalkyl include cyclopentyl,
cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
Examples of heterocycloalkyl include
1-(1,2,5,6-tetrahydropyrid- yl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl,
4-morpholinyl, 3-morpholinyl,
tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl,
tetrahydrothien-3-yl, 1-piperazinyl, 2-
piperazinyl, and the like.
The terms "halo" and "halogen," by themselves or as part of another
substituent, mean, unless
otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally,
terms such as "haloalkyl," are
meant to include alkyl substituted with halogen atoms, which can be the same
or different, in a number ranging
from one to (2m'+1), where m' is the total number of carbon atoms in the alkyl
group. For example, the term
"halo(C1-C4)alkyl" is mean to include trifluoromethyl, 2,2,2-trifluoroethyl, 4-
chlorobutyl, 3-bromopropyl, and
the like. Thus, the term "haloalkyl" includes monohaloalkyl (alkyl substituted
with one halogen atom) and
polyhaloalkyl (alkyl substituted with halogen atoms in a number ranging from
two to (2m'+1) halogen atoms,
where m' is the total number of carbon atoms in the alkyl group). The term
"perhaloalkyl" means, unless
otherwise stated, alkyl substituted with (2m'+1) halogen atoms, where m' is
the total number of carbon atoms
in the alkyl group. For example the term "perhalo(C1-C4)alkyl" is meant to
include trifluoromethyl,
pentachloroethyl, 1,1,1-trifluoro-2-bromo-2-chloroethyl and the like.
The term "acyl" refers to those groups derived from an organic acid by removal
of the hydroxy portion
of the acid. Accordingly, acyl is meant to include, for example, acetyl,
propionyl, butyryl, decanoyl, pivaloyl,
benzoyl and the like.
300
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
The term "aryl" means, unless otherwise stated, a polyunsaturated, typically
aromatic, hydrocarbon
substituent which can be a single ring or multiple rings (up to three rings)
which are fused together or linked
covalently. Non-limiting examples of aryl groups include phenyl, 1-naphthyl, 2-
naphthyl, 4-biphenyl and
1,2,3,4-tetrahydronaphthalene.
The term heteroaryl," refers to aryl groups (or rings) that contain from zero
to four heteroatoms
selected from N, 0, and S, wherein the nitrogen and sulfur atoms are
optionally oxidized and the nitrogen
heteroatom are optionally quaternized. A heteroaryl group can be attached to
the remainder of the molecule
through a heteroatom. Non-limiting examples of heteroaryl groups include 1-
pyrrolyl, 2-pyrrolyl, 3-pyrrolyl,
3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-
phenyl-4-oxazolyl, 5-oxazolyl, 3-
isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl,
2-furyl, 3-furyl, 2-thienyl, 3-
thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-
benzothiazolyl, purinyl, 2-benzimidazolyl,
5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-
quinoly1 and 6-quinolyl.
For brevity, the term "aryl" when used in combination with other terms (e.g.,
aryloxy, arylthioxy,
arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the
term "arylalkyl" is meant to
include those radicals in which an aryl group is attached to an alkyl group
(e.g., benzyl, phenethyl,
pyridylmethyl and the like) including those alkyl groups in which a carbon
atom (e.g., a methylene group) has
been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-
pyridyloxymethyl, 3-(1-
naphthyloxy)propyl, and the like).
Each of the above terms (e.g., "alkyl," "heteroalkyl," "aryl" and
"heteroaryl") is meant to include both
substituted and unsubstituted forms of the indicated radical. Preferred
substituents for each type of radical are
provided below.
Substituents for the alkyl and heteroalkyl radicals (as well as those groups
referred to as alkylene,
alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl,
cycloalkenyl and
heterocycloalkenyl) can be a variety of groups selected from: -OR', =0, =NR',
=N-OR', -NR'R", -SR', halogen,
-SiR'R"R"', -0C(0)R', -C(0)R', -CO2R', -CONR'R", -0C(0)NR'R", -NR"C(0)R', -NR'-
C(0)NR"R"', -NR'-
SO2NR"', -NR"CO2R', -NH-C(NH2)=NH, -NR'C(NH2)=NH, -NH-C(NH2)=NR', -S(0)R', -
SO2R', -SO2NR'R",
-NR"SO2R, -CN and -NO2, in a number ranging from zero to three, with those
groups having zero, one or two
substituents being particularly preferred. R', R" and R" each independently
refer to hydrogen, unsubstituted
(C1-C8)alkyl and heteroalkyl, unsubstituted aryl, aryl substituted with one to
three halogens, unsubstituted
alkyl, alkoxy or thioalkoxy groups, or ary1-(C1-C4)alkyl groups. When R' and
R" are attached to the same
nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6- or
7-membered ring. For
example, -NR'R" is meant to include 1-pyrrolidinyl and 4-morpholinyl.
Typically, an alkyl or heteroalkyl
group will have from zero to three substituents, with those groups having two
or fewer substituents being
preferred in the presently disclosed embodiments. More preferably, an alkyl or
heteroalkyl radical will be
unsubstituted or monosubstituted. Most preferably, an alkyl or heteroalkyl
radical will be unsubstituted. From
301
CA 02882923 2015-02-24
WO 2014/031125
PCT/US2012/052283
the above discussion of substituents, one of skill in the art will understand
that the term "alkyl" is meant to
include groups such as trihaloalkyl (e.g., -CF3 and -CH2CF3).
Preferred substituents for the alkyl and heteroalkyl radicals are selected
from: -OR', =0, -NR'R", -SR',
halogen, -SiR'R"R"', -0C(0)R', -C(0)R', -CO2R', -CONR'R", -0C(0)NR'R", -
NR"C(0)R', -NR"CO2R', -NR'-
SO2NR"R"', -S(0)R', -SO2R', -SO2NR'R", -NR"SO2R, -CN and -NO2, where R' and R"
are as defined above.
Further preferred substituents are selected from: -OR', =0, -NR'R", halogen, -
0C(0)R', -CO2R', -CONR'R", -
OC(0)NR'R", -NR"C(0)R', -NR"CO2R', -NR'-SO2NR"R"', -SO2R', -SO2NR'R", -NR"
SO2R, -CN and -NO2.
Similarly, substituents for the aryl and heteroaryl groups are varied and
selected from: halogen, -OR', -
OC(0)R', -NR'R", -SR', -R', -CN, -NO2, -CO2R', -CONR'R", -C(0)R', -0C(0)NR'R",
-NR"C(0)R', -
NR"CO2R', -NR'-C(0)NR"R"', -NR'-SO2NR"R"', -NH-C(NH2)=NH, -NR'C(NH2)=NH, -NH-
C(NH2)=NR', -
S(0)R', -502R', -SO2NR'R", -NR" 502R, -N3, -CH(Ph)2, perfluoro(C1-C4)alko- xy
and perfluoro(C1-C4)alkyl,
in a number ranging from zero to the total number of open valences on the
aromatic ring system; and where R',
R" and R" are independently selected from hydrogen, (C1-C8)alkyl and
heteroalkyl, unsubstituted aryl and
heteroaryl, (unsubstituted aryl)-(C1-C4)alkyl and (unsubstituted aryl)oxy-(C1-
C4)alkyl. When the aryl group
is 1,2,3,4-tetrahydronaphthalene, it may be substituted with a substituted or
unsubstituted (C3-
C7)spirocycloalkyl group. The (C3-C7)spirocycloalkyl group may be substituted
in the same manner as
defined herein for "cycloalkyl". Typically, an aryl or heteroaryl group will
have from zero to three
substituents, with those groups having two or fewer substituents being
preferred in the presently disclosed
embodiments. In one embodiment, an aryl or heteroaryl group will be
unsubstituted or monosubstituted. In
another embodiment, an aryl or heteroaryl group will be unsubstituted.
Preferred substituents for aryl and heteroaryl groups are selected from:
halogen, -OR', -0C(0)R', -
NR'R", -SR', -R', -CN, -NO2, -CO2R', -CONR'R", -C(0)R',-0C(0)NR'R", -
NR"C(0)R', -S(0)R', -502R', -
SO2NR'R", -NR"502R, -N3, -CH(Ph)2, perfluoro(C1-C4)alkoxy and perfluoro(C1-
C4)alkyl, where R' and R"
are as defined above. Further preferred substituents are selected from:
halogen, -OR', -0C(0)R', -NR'R", -R', -
CN, -NO2, -CO2R', -CONR'R", -NR"C(0)R', -502R', -SO2NR'R", -NR"502R,
perfluoro(C1-C4)alkoxy and
perfluoro(C1-C4)alkyl.
The substituent -CO2H, as used herein, includes bioisosteric replacements
therefor; see, e.g., The
Practice of Medicinal Chemistry; Wermuth, C. G., Ed.; Academic Press: New
York, 1996; p. 203.
Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may
optionally be replaced
with a substituent of the formula -T-C(0)-(CH2)q-U-, wherein T and U are
independently -NH-, -0-, -CH2- or
a single bond, and q is an integer of from 0 to 2. Alternatively, two of the
substituents on adjacent atoms of the
aryl or heteroaryl ring may optionally be replaced with a substituent of the
formula -A-(CH2)r-B-, wherein A
and B are independently -CH2-, -0-, -NH-, -S-, -5(0)-, -S(0)2-, -S(0)2NR'- or
a single bond, and r is an integer
of from 1 to 3. One of the single bonds of the new ring so formed may
optionally be replaced with a double
bond. Alternatively, two of the substituents on adjacent atoms of the aryl or
heteroaryl ring may optionally be
302
CA 02882923 2015-02-24
WO 2014/031125 PCT/US2012/052283
replaced with a substituent of the formula -(CH2)s-X-(CH2)t- -, where s and t
are independently integers of
from 0 to 3, and X is -0-, -NR'-, -S-, -S(0)-, -S(0)2-, or -S(0)2NR'-. The
substituent R' in -NR'- and -
S(0)2NR'- is selected from hydrogen or unsubstituted (C1-C6)alkyl.
303