Language selection

Search

Patent 3200814 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3200814
(54) English Title: TEAD INHIBITORS AND USES THEREOF
(54) French Title: INHIBITEURS DE TEAD ET UTILISATIONS ASSOCIEES
Status: Compliant
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 261/10 (2006.01)
  • A61K 31/444 (2006.01)
  • C07D 413/04 (2006.01)
  • C07D 413/10 (2006.01)
  • C07D 413/12 (2006.01)
  • C07D 413/14 (2006.01)
(72) Inventors :
  • CASTRO, ALFREDO C. (United States of America)
  • BURKE, MICHAEL (United States of America)
(73) Owners :
  • IKENA ONCOLOGY, INC. (United States of America)
(71) Applicants :
  • IKENA ONCOLOGY, INC. (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2021-12-02
(87) Open to Public Inspection: 2022-06-09
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2021/072684
(87) International Publication Number: WO2022/120354
(85) National Entry: 2023-05-31

(30) Application Priority Data:
Application No. Country/Territory Date
63/120,441 United States of America 2020-12-02

Abstracts

English Abstract

The present invention provides compounds, compositions thereof, and methods of using the same.


French Abstract

La présente invention concerne des composés, des compositions associées et des méthodes d'utilisation de ceux-ci.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
What is claimed is:
1. A compound of Formula 1:
Image
or a pharmaceutically acceptable salt thereof, wherein:
L1 is a covalent bound, or a C1.6 bivalent straight or branched hydrocarbon
chain wherein 1, 2, or
3 methylene units of the chain are independently and optionally replaced with -
N(R)-, -0-, or
-C(0)-;
Image
Ring A is selected from
Image
each of which is optionally substituted;
Image
Ring B is selected from
Image
Image
each Rw is independently selected from
165
CA 03200814 2023- 5- 31

each R2 is independently is selected from -OR, -C(0)NR2, optionally
substituted -C1_6 aliphatic,
Image
each Y is independently N or CR5;
each R3 is independently H, or optionally substituted -C1_6 aliphatic;
each R4 is independently -S(0)2NR2, -S(0)2R, -C(0)NR2, -C(0)R, or optionally
substituted -C1-6
aliphatic;
each R5 is independently R, -CN, -C(0)R, -C(0)NR2, or optionally substituted 5-
6 membered
heteroaryl having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur;
each m is independently 0, 1, or 2;
p is 0, 1, or 2; and
each R is independently H, optionally substituted -C1-6 aliphatic, optionally
substituted 3-8
membered saturated or partially unsaturated monocyclic carbocyclyl, or
optionally substituted
3-8 membered saturated or partially unsaturated monocyclic heterocyclyl having
1-2
heteroatoms independently selected from nitrogen, oxygen, or sulfur.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
wherein is Ci-
6 bivalent straight or branched hydrocarbon chain, wherein 1, 2, or 3
methylene units of the chain
are replaced with -N(R)-.
3. The compound of claims 1 or 2, or a pharmaceutically acceptable salt
thereof, wherein
Image
Ring A is , which is optionally substituted.
4. The compound of claims 1 or 2, or a pharmaceutically acceptable salt
thereof, wherein
Image
Ring A is , whi ch is optionally sub stituted.
166
CA 03200814 2023- 5- 31

5. The compound of claims 1 or 2, or a pharmaceutically acceptable salt
thereof, wherein
Image
Ring A is , which is optionally substituted.
6. The compound of claims 1 or 2, or a pharmaceutically acceptable salt
thereof, wherein
Image
Ring A is , which is optionally substituted.
7. The compound of any one of claims 1-6, or a pharmaceutically acceptable
salt thereof,
Image
wherein Ring B is
, or
Image
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable
salt thereof,
Image
wherein each 1r is independently
9. The compound of claim 1, which is a compound of formulas:
Image
167
CA 03200814 2023- 5- 31

Image
168
CA 03200814 2023- 5- 31

Image
or a pharmaceutically acceptable salt thereof, wherein each RI- is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
1 0. The compound of claim 1, which is a compound of formulas:
169
CA 03200814 2023- 5- 31

Image
170
CA 03200814 2023- 5- 31

Image
171
CA 03200814 2023- 5- 31

Image
or a pharmaceutically acceptable salt thereof, wherein each 10 is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
11 The compound of claim 1, which is a compound of formulas.
Image
172
CA 03200814 2023- 5- 31

Image
173
CA 03200814 2023- 5- 31

Image
174
CA 03200814 2023- 5- 31

Image
or a pharmaceutically acceptable salt thereof, wherein each
is independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
12. The compound of claim 1, which is a compound of formulas:
Image
175
CA 03200814 2023- 5- 31

Image
, or a pharmaceutically acceptable salt thereof, wherein each RI- is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R.
1 3 . The compound of claim 1, which is a compound of formulas:
Image
176
CA 03200814 2023- 5- 31

Image
177
CA 03200814 2023- 5- 31

or a pharmaceutically acceptable salt thereof, wherein each RI- is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
14. The compound of claim 1, which is a compound of formulas:
Image
178
CA 03200814 2023- 5- 31

Image
or a pharmaceutically acceptable salt thereof, wherein each Rl is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
15. The compound of claim 1, which is a compound of Formulas:
Image
or a pharmaceutically acceptable salt thereof, wherein each RI- is
independently R, halogen, -CN,
-C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is independently
0, 1, 2, or 3.
16. The compound of any one of the preceding claims, which is selected from
the compounds
of Table 1, or a pharmaceutically acceptable salt thereof
179
CA 03200814 2023- 5- 31

17. A pharmaceutical composition comprising the compound of any one of the
preceding
claims, or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable carrier,
adjuvant, or vehi cl e.
18. A method for treating cancer in a patient, comprising administering to
the patient the
compound of any one of claims 1-16, or a pharmaceutically acceptable salt
thereof, or the
pharmaceutical composition of claim 17.
19. The method of claim 18, wherein the cancer is associated with increased
TEAD expression.
20. The method of claims 18 or 19, wherein the cancer is associated with
increased TEAD
activity.
21. A method for inhibiting the progress of cancer in a patient, comprising
administering to
the patient the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt thereof,
or the pharmaceutical composition of claim 17.
22. The method of claim 21, wherein the cancer is associated with increased
TEAD expression.
23. The method of claims 21 or 22, wherein the cancer is associated with
increased TEAD
activity.
24. A method of treating a patient having a di sease or di sorder
associated with increased TEAD
expression comprising the step of administering to a patient in need thereof a
therapeutically
effective amount of the compound of any one of claims 1-16, or a
pharmaceutically acceptable salt
thereof, or the pharmaceutical composition of claim 17.
25. A method of treating a patient having a disease or disorder associated
with increased TEAD
activity comprising the step of administering to a patient in need thereof a
therapeutically effective
180
CA 03200814 2023- 5- 31

amount of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt thereof,
or the pharmaceutical composition of claim 17.
26. A method of treating a disease or disorder in which inhibition of TEAD
activity is
beneficial comprising the step of administering to a patient in need thereof a
therapeutically
effective amount of the compound of any one of claims 1-16, or a
pharmaceutically acceptable salt
thereof, or the pharmaceutical composition of claim 17.
27. A method of treating a di sease or disorder in which Hippo pathway
inhibition is beneficial
comprising the step of administering to a patient in need thereof a
therapeutically effective amount
of the compound of any one of claims 1-16, or a pharmaceutically acceptable
salt thereof, or the
pharmaceutical composition of claim 17.
28. The method of any one of claims 24-27, wherein the disease or disorder
is a cellular
proliferative disorder.
29. The method of claim 28, wherein the cellular proliferative disorder is
cancer.
30. The method of any one of claims 18-23 and 29, wherein the cancer is a
cancer in which
YAP is localized in the nucleus of cells of the cancer.
31. The method of any one of claims 19, 20, and 22-25, wherein the
increased TEAD
expression or increased TEAD activity is increased TEAD1 expression or
increased TEAD1
activity.
32. The method of any one of claims 19, 20, and 22-25, wherein the
increased TEAD
expression or increased TEAD activity is increased TEAD2 expression or
increased TEAD2
activity.
181
CA 03200814 2023- 5- 31

33. The method of any one of claims 19, 20, and 22-25, wherein the
increased TEAD
expression or increased TEAD activity is increased TEAD3 expression or
increased TEAD3
activity.
34. The method of any one of claims 19, 20, and 22-25, wherein the
increased TEAD
expression or increased TEAD activity is increased TEAD4 expression or
increased TEAD4
activity.
35. The method of any one of claims 19, 20, and 22-25, wherein the
increased TEAD
expression or increased TEAD activity is increased TEAD1 expression or
increased TEAD1
activity; increased TEAD2 expression or increased TEAD2 activity; increased
TEAD3 expression
or increased TEAD3 activity; increased TEAD4 expression or increased TEAD4
activity; or any
combination thereof
36. Use of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt
thereof, or the pharmaceutical composition of claim 17, for treating cancer in
a patient.
37. The use of claim 36, wherein the cancer is associated with increased
TEAD expression.
38. The use of claims 36 or 37, wherein the cancer is associated with
increased TEAD activity.
39. Use of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt
thereof, or the pharmaceutical composition of claim 17, for inhibiting the
progress of cancer in a
pati ent.
40. The use of claim 39, wherein the cancer is associated with increased
TEAD expression.
41. The use of claims 39 or 40, wherein the cancer is associated with
increased TEAD activity.
182
CA 03200814 2023- 5- 31

42. Use of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt
thereof, or a pharmaceutical composition of claim 17, for treating a disease
or disorder associated
with increased TEAD expression.
43. Use of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt
thereof, or the pharmaceutical composition of claim 17, for treating a disease
or disorder associated
with increased TEAD activity.
44. Use of the compound of any one of claims 1-16, or a pharmaceutically
acceptable salt
thereof, or the pharmaceutical composition of claim 17, for treating a disease
or disorder in which
inhibition of TEAD activity is beneficial.
45. Use of a compound of any one of claims 1-16, or a pharmaceutically
acceptable salt thereof,
or a pharmaceutical composition of claim 17, for treating a disease or
disorder in which Hippo
pathway inhibition is beneficial.
46. The use of any one of claims 42-45, wherein the disease or disorder is
a cellular
proliferative disorder.
47. The use of claim 46, wherein the cellular proliferative disorder is
cancer.
48. The use of any one of claims 36-41 and 47, wherein the cancer is a
cancer in which YAP
is localized in the nucleus of cells of the cancer.
49. The use of any one of claims 37, 38, and 40-43, wherein the increased
TEAD expression
or increased TEAD activity is increased TEAD1 expression or increased TEAD1
activity.
50. The use of any one of claims 37, 38, and 40-43, wherein the increased
TEAD expression
or increased TEAD activity is increased TEAD2 expression or increased TEAD2
activity.
183
CA 03200814 2023- 5- 31

51. The use of any one of claims 37, 38, and 40-43, wherein the increased
TEAD expression
or increased TEAD activity is increased TEAD3 expression or increased TEAD3
activity.
52. The use of any one of claims 37, 38, and 40-43, wherein the increased
TEAD expression
or increased TEAD activity is increased TEAD4 expression or increased TEAD4
activity.
53. The use of any one of claims 37, 38, and 40-43, wherein the increased
TEAD expression
or increased TEAD activity is increased TEAD1 expression or increased TEAD1
activity;
increased TEAD2 expression or increased TEAD2 activity; increased TEAD3
expression or
increased TEAD3 activity; increased TEAD4 expression or increased TEAD4
activity; or any
combination thereof.
184
CA 03200814 2023- 5- 31

Description

Note: Descriptions are shown in the official language in which they were submitted.


WO 2022/120354
PCT/US2021/072684
TEAD INHIBITORS AND USES THEREOF
SEQUENCE LISTING
100011
The instant application contains a Sequence Listing which has been
submitted
electronically in ASCII format and is hereby incorporated by reference in its
entirety. Said ASCII
copy, created on November 30, 2021, is named 187446 SL.txt and is 23,551 bytes
in size.
TECHNICAL FIELD OF THE INVENTION
100021
The present invention relates to compounds and methods useful for
inhibition of
Transcriptional Enhancer Associate Domain (TEAD).
The invention also provides
pharmaceutically acceptable compositions comprising compounds of the present
invention and
methods of using said compositions in the treatment of various diseases,
disorders, and conditions
as described herein.
BACKGROUND OF TIIE INVENTION
100031
Yes-associated protein (YAP) and transcriptional co-activator with PDZ-
binding motif
(TAZ) are transcriptional co-activators of the Hippo pathway network and
regulate cell
proliferation, migration, and apoptosis. Inhibition of the Hippo pathway
promotes YAP/TAZ
translocation to the nucleus, wherein YAP/TAZ interact with TEAD transcription
factors and
coactivate the expression of target genes and promote cell proliferation.
Hyperactivation of YAP
and TAZ and/or mutations in one or more members of the Hippo pathway network
have been
implicated in numerous cancers.
SUMMARY OF THE INVENTION
100041
The Hippo signaling cascade is an important pathway for cancer
biogenesis and tumor
maintenance. The Hippo pathway is heavily mutated across many cancer
indications through loss
of function mutations in genes such as NF2. These pro-tumor mutations lead to
the constitutive
activation of the downstream transcriptional coactivators YAP and TAZ that
drive the expression
of many pro-survival and proliferation genes through the essential interaction
with a TEAD protein
family member. In addition, this unrestrained transcriptional program drives
enhanced immune
suppression in the tumor microenvironment. As described herein, to target this
oncogenic pathway
1
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
novel small molecule inhibitors were identified that selectively bind to TEAD
and disrupt their
interaction with YAP and TAZ, thereby downregulating YAP- and TAZ-dependent
transcription.
As demonstrated herein, these TEAD inhibitors prevent TEAD palmitoylation,
which is critical
for the interaction between YAP and TEAD. Furthermore, the TEAD inhibitors
described herein
inhibit in vitro proliferation of YAP-dependent (i.e., Hippo pathway-deficient
cancer cell lines),
but not Hippo pathway wild type cancer cell lines. Importantly, as shown
herein, the TEAD
inhibitor compounds of the present invention did not affect survival of a
differentiated mouse
podocyte cell line or compromise mouse kidney histology. Subsequent
experiments in vivo
demonstrate the TEAD inhibitors described herein downregul ate YAP-dependent
genes in human
tumor xenografts after oral dosing. In addition, the TEAD inhibitors described
herein exhibit single
agent tumor growth inhibition of human tumor xenografts in mice at well
tolerated oral doses. The
data described herein demonstrate the ability of the small molecule TEAD
inhibitors provided
herein for targeting the Hippo pathway in cancers.
[0005] It has now been found that compounds of the present
invention, and pharmaceutically
acceptable compositions thereof, are effective as TEAD inhibitors. In one
aspect, the present
invention provides a compound of Formula I:
L1
A
or a pharmaceutically acceptable salt thereof, wherein each variable is
independently as defined
herein and as described in embodiments herein.
[0006] Compounds of the present invention, and pharmaceutically
acceptable salts and
compositions thereof, are useful for treating a variety of diseases, disorders
or conditions
associated with TEAD. Such diseases, disorders, or conditions include cellular
proliferative
disorders (e.g., cancer as described herein).
BRIEF DESCRIPTION OF FIGURES
100071 FIGURE 1 depicts a schematic of Hippo pathway signaling.
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
2
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1. General Description of Certain Embodiments of the Invention:
[0008] Compounds of the present invention, and pharmaceutical salts
and compositions
thereof, are useful as inhibitors of TEAD. Without wishing to be bound by any
particular theory,
it is believed that compounds of the present invention, and pharmaceutical
compositions thereof,
inhibit the activity of TEAD, and thus treat diseases, disorders or conditions
associated with
TEAD, such as cancer.
[0009] In one aspect, the present invention provides a compound of
Formula I:
L1
A
or a pharmaceutically acceptable salt thereof, wherein.
1_,1 is a covalent bound, or a C1_6 bivalent straight or branched hydrocarbon
chain wherein 1, 2, or
3 methylene units of the chain are independently and optionally replaced with -
N(R)-, -0-, or
-C(0)-;
FO H<>
Ring A is selected from
0¨\)
N,
1¨( .NH 41100 0 4111 N
110
H and
,
each of which is optionally substituted;
R2
R2
R2
I
R4 Rw Rw
Ring B is selected from Rw
R2
RA
EIIT> I
Rw¨e(CN-4
R4% N , and =
Br c tIN,16
nr-Ny
each IV' is independently selected from 0 , and =
3
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
each le is independently selected from -OR, -C(0)NR2, optionally substituted -
C1_6 aliphatic,
(R5)m (R5)
m (R5) (R5)m 0
HN+Y ____________________________ HNN(R5) (R5)m
HN
õ,õ ___________________________________________________________
N k=s, N N H
N
N
L.õ7.7,0
and
each Y is independently N or CR%
each le is independently H or optionally substituted -C1_6 aliphatic,
each R4 is independently -S(0)2NR2, -S(0)2R, -C(0)NR2, -C(0)R, or optionally
substituted -C1-6
aliphatic;
each Rs is independently R, -CN, -C(0)R, -C(0)NR2, or optionally substituted 5-
6 membered
heteroaryl having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur;
each m is independently 0, 1, or 2;
p is 0, 1, or 2; and
each R is independently H, optionally substituted -C1-6 aliphatic, optionally
substituted 3-8
membered saturated or partially unsaturated monocyclic carbocyclyl, or
optionally substituted
3-8 membered saturated or partially unsaturated monocyclic heterocyclyl having
1-2
heteroatoms independently selected from nitrogen, oxygen, or sulfur.
2. Compounds and Definitions:
100101 Compounds of the present invention include those described
generally herein, and are
further illustrated by the classes, subclasses, and species disclosed herein.
As used herein, the
following definitions shall apply unless otherwise indicated. For purposes of
this invention, the
chemical elements are identified in accordance with the Periodic Table of the
Elements, CAS
version, Handbook of Chemistry and Physics, '75th Ed. Additionally, general
principles of organic
chemistry are described in "Organic Chemistry", Thomas Sorrell, University
Science Books,
Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th E
a Ed.: Smith, M.B. and
March, J., John Wiley & Sons, New York: 2001, the entire contents of which are
hereby
incorporated by reference.
100111 The term "aliphatic" or "aliphatic group", as used herein,
means a straight-chain (i.e.,
unbranched) or branched, substituted or unsubstituted hydrocarbon chain that
is completely
saturated or that contains one or more units of unsaturation, or a monocyclic
hydrocarbon or
4
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
bicyclic hydrocarbon that is completely saturated or that contains one or more
units of
unsaturation, but which is not aromatic (also referred to herein as
"carbocycle," "cycloaliphatic"
or "cycloalkyl"), that has a single point of attachment to the rest of the
molecule. Unless otherwise
specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some
embodiments, aliphatic
groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic
groups contain 1-4
aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-
3 aliphatic carbon
atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic
carbon atoms. In some
embodiments, -cycloaliphatic" (or -carbocycle" or -cycloalkyl") refers to a
monocyclic C3-C6
hydrocarbon that is completely saturated or that contains one or more units of
unsaturation, but
which is not aromatic, that has a single point of attachment to the rest of
the molecule. Suitable
aliphatic groups include, but are not limited to, linear or branched,
substituted or unsubstituted
alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl,
(cycloalkenyl)alkyl
or (cycloalkyl)alkenyl.
100121 As used herein, the term "bicyclic ring- or "bicyclic ring
system- refers to any bicyclic
ring system, i.e. carbocyclic or heterocyclic, saturated or having one or more
units of unsaturation,
having one or more atoms in common between the two rings of the ring system.
Thus, the term
includes any permissible ring fusion, such as ortho-fused or spirocyclic. As
used herein, the term
"heterobicyclic" is a subset of "bicyclic" that requires that one or more
heteroatoms are present in
one or both rings of the bicycle. Such heteroatoms may be present at ring
junctions and are
optionally substituted, and may be selected from nitrogen (including N-
oxides), oxygen, sulfur
(including oxidized forms such as sulfones and sulfonates), phosphorus
(including oxidized forms
such as phosphates), boron, etc. In some embodiments, a bicyclic group has 7-
12 ring members
and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
As used herein, the
term "bridged bicyclic" refers to any bicyclic ring system, i . e. carbocycli
c or heterocyclic,
saturated or partially unsaturated, having at least one bridge. As defined by
IUPAC, a "bridge" is
an unbranched chain of atoms or an atom or a valence bond connecting two
bridgeheads, where a
"bridgehead" is any skeletal atom of the ring system which is bonded to three
or more skeletal
atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has
7-12 ring
members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or
sulfur. Such
bridged bicyclic groups are well known in the art and include those groups set
forth below where
each group is attached to the rest of the molecule at any substitutable carbon
or nitrogen atom.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Unless otherwise specified, a bridged bicyclic group is optionally substituted
with one or more
substituents as set forth for aliphatic groups. Additionally or alternatively,
any substitutable
nitrogen of a bridged bicyclic group is optionally substituted. Exemplary
bicyclic rings include:
111011 CO Cb
NOD
N H H
Exemplary bridged bicyclics include:
NH
H
HN
HN HN
110 H 02'1
0
NH NH G1NH
CS:I NHS
CC
o 1001
100131 The term "lower alkyl- refers to a C1-4 straight or branched
alkyl group. Exemplary
lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and
tert-butyl.
100141 The term "lower haloalkyl" refers to a C1-4 straight or
branched alkyl group that is
substituted with one or more halogen atoms.
100151 The term -heteroatom" means one or more of oxygen, sulfur,
nitrogen, phosphorus, or
silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or
silicon; the quaternized
form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic
ring, for example N (as in
3,4-dihydro-2H-pyrroly1), NH (as in pyrrolidinyl) or NR + (as in N-substituted
pyrrolidinyl)).
100161 The term "unsaturated," as used herein, means that a moiety
has one or more units of
unsaturation.
6
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[0017] As used herein, the term "bivalent C1-8 (or C1_6) saturated
or unsaturated, straight or
branched, hydrocarbon chain", refers to bivalent alkylene, alkenylene, and
alkynylene chains that
are straight or branched as defined herein.
[0018] The term "alkylene" refers to a bivalent alkyl group. An
"alkylene chain" is a
polymethylene group, i.e., ¨(CH2)n¨, wherein n is a positive integer,
preferably from 1 to 6, from
1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain
is a polymethylene
group in which one or more methylene hydrogen atoms are replaced with a
substituent. Suitable
substituents include those described below for a substituted aliphatic group.
[0019] The term "alkenylene" refers to a bivalent alkenyl group. A
substituted alkenylene
chain is a polymethylene group containing at least one double bond in which
one or more hydrogen
atoms are replaced with a substituent. Suitable substituents include those
described below for a
substituted aliphatic group.
[0020] As used herein, the term "cyclopropylenyl" refers to a
bivalent cyclopropyl group of
the following structure: .
[0021] The term "halogen" means F, Cl, Br, or I.
[0022] The term "aryl" used alone or as part of a larger moiety as
in "aralkyl," "aralkoxy," or
"aryloxyalkyl," refers to monocyclic or bicyclic ring systems having a total
of five to fourteen ring
members, wherein at least one ring in the system is aromatic and wherein each
ring in the system
contains 3 to 7 ring members. The term "aryl" may be used interchangeably with
the term "aryl
ring." In certain embodiments of the present invention, "aryl" refers to an
aromatic ring system
which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and
the like, which may
bear one or more substituents. Also included within the scope of the term
"aryl," as it is used
herein, is a group in which an aromatic ring is fused to one or more
non¨aromatic rings, such as
indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl,
and the like
[0023] The terms "heteroaryl" and "heteroar¨," used alone or as part
of a larger moiety, e.g.,
"heteroaralkyl," or "heteroaralkoxy," refer to groups having 5 to 10 ring
atoms, preferably 5, 6, or
9 ring atoms; having 6, 10, or 14 7C electrons shared in a cyclic array; and
having, in addition to
carbon atoms, from one to five heteroatoms. The term "heteroatom" refers to
nitrogen, oxygen, or
sulfur, and includes any oxidized form of nitrogen or sulfur, and any
quaternized form of a basic
nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl,
pyrrolyl, imidazolyl,
7
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl,
thiazolyl, isothiazolyl,
thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl,
purinyl, naphthyridinyl, and
pteridinyl. The terms "heteroaryl" and "heteroar¨", as used herein, also
include groups in which a
heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or
heterocyclyl rings, where the
radical or point of attachment is on the heteroaromatic ring. Nonlimiting
examples include indolyl,
isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl,
benzimidazolyl, benzthiazolyl,
quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl,
4H¨quinolizinyl,
carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl,
tetrahydroquinolinyl,
tetrahydroisoquinolinyl, and pyrido[2,3¨b]-1,4¨oxazin-3(4H)¨one. A heteroaryl
group may be
mono¨ or bicyclic. The term "heteroaryl" may be used interchangeably with the
terms "heteroaryl
ring," "heteroaryl group," or "heteroaromatic," any of which terms include
rings that are optionally
substituted. The term "heteroaralkyl" refers to an alkyl group substituted by
a heteroaryl, wherein
the alkyl and heteroaryl portions independently are optionally substituted.
100241 As used herein, the terms "heterocycle,- "heterocyclyl,"
"heterocyclic radical,- and
"heterocyclic ring" are used interchangeably and refer to a stable 5¨ to
7¨membered monocyclic
or 7-10¨membered bicyclic heterocyclic moiety that is either saturated or
partially unsaturated,
and having, in addition to carbon atoms, one or more, preferably one to four,
heteroatoms, as
defined above. When used in reference to a ring atom of a heterocycle, the
term "nitrogen" includes
a substituted nitrogen. As an example, in a saturated or partially unsaturated
ring having 0-3
heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N
(as in 3,4¨dihydro-
2H¨pyrroly1), NH (as in pyrrolidinyl), or 'Nit (as in N¨substituted
pyrrolidinyl).
100251 A heterocyclic ring can be attached to its pendant group at
any heteroatom or carbon
atom that results in a stable structure and any of the ring atoms can be
optionally substituted.
Examples of such saturated or partially unsaturated heterocyclic radicals
include, without
limitation, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl,
piperidinyl, pyrrolinyl,
tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl,
oxazolidinyl, piperazinyl,
dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and
quinuclidinyl. The
terms "heterocycle," "heterocyclyl," "heterocyclyl ring," "heterocyclic
group," "heterocyclic
moiety,- and "heterocyclic radical,- are used interchangeably herein, and also
include groups in
which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or
cycloaliphatic rings, such as
indolinyl, 3H¨indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A
heterocyclyl group
8
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
may be mono- or bicyclic. The term "heterocyclylalkyl" refers to an alkyl
group substituted by a
heterocyclyl, wherein the alkyl and heterocyclyl portions independently are
optionally substituted.
[0026] As used herein, the term "partially unsaturated" refers to a
ring moiety that includes at
least one double or triple bond. The term "partially unsaturated" is intended
to encompass rings
having multiple sites of unsaturation, but is not intended to include aryl or
heteroaryl moieties, as
herein defined.
[0027] As described herein, compounds of the invention may contain
"optionally substituted"
moieties. In general, the term -substituted," whether preceded by the term -
optionally" or not,
means that one or more hydrogens of the designated moiety are replaced with a
suitable substituent.
Unless otherwise indicated, an "optionally substituted" group may have a
suitable substituent at
each substitutable position of the group, and when more than one position in
any given structure
may be substituted with more than one substituent selected from a specified
group, the substituent
may be either the same or different at every position. Combinations of
substituents envisioned by
this invention are preferably those that result in the formation of stable or
chemically feasible
compounds. The term "stable," as used herein, refers to compounds that are not
substantially
altered when subjected to conditions to allow for their production, detection,
and, in certain
embodiments, their recovery, purification, and use for one or more of the
purposes disclosed
herein.
[0028] Each optional substituent on a substitutable carbon is a
monovalent substituent
independently selected from halogen; -(CH2)o_4R ; -(CH2)o_40R ; -0(CH2)0_4R , -
0-(CH2)o_
4C(0)01V; -(CH2)0_4CH(OR )2; -(CH2)0_4 SR ; -(CH2)0_4Ph, which may be
substituted with R ,
-(CH2)0_40(CH2)0_11311 which may be substituted with R`); -CH=CHPh, which may
be substituted
with R ; -(CH2)0_40(CH2)o-1-pyridyl which may be substituted with R ; -NO2; -
CN; -
N3; -(C112)0-4N(W)2; -(CH2)0-4N(R )C (0)R ; -N(R )C(S)R ;
-(CH2)o-
4N(R )C (0)NR 2 ; -N(R )C(S)NR 2; -(CH2)0_4N(R )C (0)0R
;
N(R )N(R )C(0)R ; -N(R )N(R )C(0)NR 2, -N(R )N(R )C(0)0R ; -(CH2)0_4C(0)R ; -
C(S)R ; -(CH2)o_4C(0)0R ; -(CH2)0_4C(0)SR ; -(CH2)o_4C(0)0SiR 3; -
(CH2)o_40C(0)R ; -
OC(0)(CH2)0_4SR-, SC(S)SR ; -(CH2)0_4SC(0)R ; -(CH2)0_4C(0)NR 2; -C(S)NR 2; -
C(S)SR ;
-SC(S)SR , -(CH2)0_40C(0)NR 2; -C(0)N(OR )R ; -C(0)C(0)R ; -C(0)CH2C(0)R ; -
C(NOR )R ; -(CH2)0_4 SSR ; -(CH2)0_4 S(0)2R ; -(CH2)0_4 S(0)20R ; -(CH2)0_40
S(0)2R ; -
S(0)2NR 2; -S(0)(NR )R ; -S(0)2N=C(NR 2)2; -(CH2)o-4 S (0)R ; -N(R )S(0)2NR
2;
9
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
N(R )S(0)2R ; -N(OR )R ; -C(NH)NR 2; -P(0)2R ; -P(0)R 2; -0P(0)R 2; -0P(0)(OR
)2;
SiR 3; -(C1_4 straight or branched alkylene)0-N(R )2; or -(C1_4 straight or
branched
alkyl ene)C (0)0-N(R )2.
100291 Each R is independently hydrogen, C1_6 aliphatic, -CH2Ph, -
0(CH2)0_113h, -CH2-(5-6
membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated,
or aryl ring having
0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or,
notwithstanding the
definition above, two independent occurrences of R , taken together with their
intervening atom(s),
form a 3-12-membered saturated, partially unsaturated, or aryl mono- or
bicyclic ring having 0-
4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which
may be substituted
by a divalent substituent on a saturated carbon atom of R selected from =0
and =S; or each R
is optionally substituted with a monovalent substituent independently selected
from halogen, -
(CH2)0_21e, -(haloR'), -(CH2)0_20H, -(CH2)0_20R', -(CH2)0_2CH(OR')2; -
0(haloR"), -CN, -N3,
-(CH2)0_2C(0)R", -(CH2)0_2C(0)0H, -(CH2)0_2C(0)0R", -(CH2)0_2SR", -(CH2)0_2SH,
-(CH2)o-
2NH2, -(CH2)0_2NE1fe, -(CH2)0_2NR"2, -NO2, -SiR"3, -0 SiR".3, -C(0)SR", -(C2_4
straight or
branched alkylene)C(0)0R", or -S SR'.
100301 Each R' is independently selected from C1-4 aliphatic, -
CH2Ph, -0(CH2)0_11311, or a 5-
6-membered saturated, partially unsaturated, or aryl ring having 0-4
heteroatoms independently
selected from nitrogen, oxygen, or sulfur, and wherein each R' is
unsubstituted or where preceded
by halo is substituted only with one or more halogens; or wherein an optional
substituent on a
saturated carbon is a divalent substituent independently selected from =0, =S,
=NNR*2,
=NNHC (0)R*, =NNHC (0)0R*, =NNHS (0)2R*, =NR', =NOR*, -0(C (R*2))2_30-, or -
S (C (R*2))2_3S-, or a divalent substituent bound to vicinal substitutable
carbons of an -optionally
substituted" group is -0(CR*2)2_30-, wherein each independent occurrence of R*
is selected from
hydrogen, C1_6 aliphatic or an unsubstituted 5-6-membered saturated, partially
unsaturated, or
aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen,
or sulfur.
100311 When R* is Cis aliphatic, R* is
optionally substituted with halogen, -
R', -(haloR'), -OH, -OR', -0(haloR'), -CN, -C(0)0H, -C(0)0R", -NH2, -NHR', -
NR'2, or -
NO2, wherein each le is independently selected from C11 aliphatic, -CH2Ph, -
0(CH2)0_21311, or a
5-6-membered saturated, partially unsaturated, or aryl ring having 0-4
heteroatoms independently
selected from nitrogen, oxygen, or sulfur, and wherein each R' is
unsubstituted or where preceded
by halo is substituted only with one or more halogens.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
100321 An optional substituent on a substitutable nitrogen is
independently -Rt, .. -
C(0)1e, -C(0)01e, -C(0)C(0)1e, -C(0)CH2C(0)1e, -S(0)21e, -S(0)2Nle2, -
C(S)Nlez, -
C(NH)NR1.2, or -N(Rt)S(0)2Rt; wherein each Rt is independently hydrogen, C1-6
aliphatic,
unsubstituted -0Ph, or an unsubstituted 5-6--membered saturated, partially
unsaturated, or aryl
ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or
sulfur, or, two
independent occurrences of R, taken together with their intervening atom(s)
form an unsubstituted
3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring
having 0-4
heteroatoms independently selected from nitrogen, oxygen, or sulfur; wherein
when RI. is C1_6
aliphatic, le is optionally substituted with halogen, -RI', -(haloR*), -OH, -
OR*, -0(haloR*), -
CN, -C(0)0H, -C(0)01e, -NH2, -NUR', -NR*2, or -NO2, wherein each R is
independently
selected from C1-4 aliphatic, -CH2Ph, -0(CH2)o-i_Ph, or a 5-6-membered
saturated, partially
unsaturated, or aryl ring having 0-4 heteroatoms independently selected from
nitrogen, oxygen,
or sulfur, and wherein each R. is unsubstituted or where preceded by halo is
substituted only with
one or more halogens.
100331 As used herein, the term "pharmaceutically acceptable salt"
refers to those salts which
are, within the scope of sound medical judgment, suitable for use in contact
with the tissues of
humans and lower animals without undue toxicity, irritation, allergic response
and the like, and
are commensurate with a reasonable benefit/risk ratio. Pharmaceutically
acceptable salts are well
known in the art. For example, S. M. Berge et al., describe pharmaceutically
acceptable salts in
detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by
reference.
Pharmaceutically acceptable salts of the compounds of this invention include
those derived from
suitable inorganic and organic acids and bases. Examples of pharmaceutically
acceptable,
nontoxic acid addition salts are salts of an amino group formed with inorganic
acids such as
hydrochloric acid, hydrobromi c acid, phosphoric acid, sulfuric acid and
perchlori c acid or with
organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid,
citric acid, succinic acid
or malonic acid or by using other methods used in the art such as ion
exchange. Other
pharmaceutically acceptable salts include adipate, alginate, ascorbate,
aspartate, benzenesulfonate,
benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate,
cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate,
fumarate,
glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate,
hexanoate, hydroiodide, 2-
hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate,
malate, maleate, malonate,
11
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
methanesulfonate, 2¨naphthalenesulfonate, nicotinate, nitrate, oleate,
oxalate, palmitate, pamoate,
pectinate, persulfate, 3¨phenylpropionate, phosphate, pivalate, propionate,
stearate, succinate,
sulfate, tartrate, thiocyanate, p¨toluenesulfonate, undecanoate, valerate
salts, and the like.
100341 Salts derived from appropriate bases include alkali metal,
alkaline earth metal,
ammonium and 1\r(Ci_aalky1)4 salts. Representative alkali or alkaline earth
metal salts include
sodium, lithium, potassium, calcium, magnesium, and the like. Further
pharmaceutically
acceptable salts include, when appropriate, nontoxic ammonium, quaternary
ammonium, and
amine cations formed using counterions such as halide, hydroxide, carboxylate,
sulfate, phosphate,
nitrate, loweralkyl sulfonate and aryl sulfonate.
100351 Unless otherwise stated, structures depicted herein are also
meant to include all
isomeric (e.g., enantiomeric, diastereomeric, and geometric (or
conformational)) forms of the
structure; for example, the R and S configurations for each asymmetric center,
Z and E double
bond isomers, and Z and E conformational isomers. Therefore, single
stereochemical isomers as
well as enantiomeric, diastereomeric, and geometric (or conformational)
mixtures of the present
compounds are within the scope of the invention. Unless otherwise stated, all
tautomeric forms of
the compounds of the invention are within the scope of the invention.
Additionally, unless
otherwise stated, structures depicted herein are also meant to include
compounds that differ only
in the presence of one or more isotopically enriched atoms. For example,
compounds having the
present structures including the replacement of hydrogen by deuterium or
tritium, or the
replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope
of this invention.
Such compounds are useful, for example, as analytical tools, as probes in
biological assays, or as
therapeutic agents in accordance with the present invention. In certain
embodiments, a warhead
moiety of a provided compound comprises one or more deuterium atoms.
100361 As used herein, the terms "inhibitor" or "TEAD inhibitor" or
"TEAD antagonist" are
defined as a compound that binds to and/or inhibits TEAD with measurable
affinity. In some
embodiments, inhibition in the presence of the inhibitor is observed in a dose-
dependent manner.
In some embodiments, the measured signal (e.g., signaling activity or
biological activity) is at least
about 5%, at least about 10%, at least about 15%, at least about 20%, at least
about 25%, at least
about 30%, at least about 35%, at least about 40%, at least about 45%, at
least about 50%, at least
about 55%, at least about 60%, at least about 65%, at least about 70%, at
least about 75%, at least
about 80%, at least about 85%, at least about 90%, at least about 95%, at
least about 96%, at least
12
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
about 97%, at least about 98%, at least about 99%, or at least about 100%
lower than the signal
measured with a negative control under comparable conditions. The potency of
an inhibitor is
usually defined by its IC50 value (half maximal inhibitory concentration or
concentration required
to inhibit 50% of the agonist response). The lower the IC50 value the greater
the potency of the
antagonist and the lower the concentration that is required to inhibit the
maximum biological
response. In certain embodiments, an inhibitor has an IC50 and/or binding
constant of less than
about 100 [tM, less than about 50 [tM, less than about 1 [IM, less than about
500 nM, less than
about 100 nM, less than about 10 nM, or less than about 1 nM.
100371 The terms "measurable affinity" and "measurably inhibit," as
used herein, means a
measurable change or inhibition in TEAD activity between a sample comprising a
compound of
the present invention, or composition thereof, and TEAD, and an equivalent
sample comprising
TEAD, in the absence of said compound, or composition thereof.
3. Description of Exemplar), Embodiments:
100381 In one aspect, the present invention provides a compound of
Formula I:
L1
A
or a pharmaceutically acceptable salt thereof, wherein:
L1 is a covalent bound, or a Ci.6 bivalent straight or branched hydrocarbon
chain wherein 1, 2, or
3 methylene units of the chain are independently and optionally replaced with -
N(R)-, -0-, or
-C(0)-,
1-0 H<>
Ring A is selected from
NN
,
H
N H = 0 1411 N , and 1410
,
each of which is optionally substituted;
13
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
R2
R2 Rw R2
I
N
Ring B is selected from Rw R4
R2
(R5)p
N
Rw¨<CN--1
N , 0 , and
Br CI t 16
nr1.4,71 **.c-11
each TV is independently selected from 0 , and ___________ ¨ =
each R2 is independently selected from -OR, -C(0)NR2, optionally substituted -
C1.6 aliphatic,
(R5)ni D5 3
(, )rn (R5)m (R)m 0
HN+Y
I HN)1-(R5)m 5 (R5)rn
NH FINN (R
N N
NW.. Aglow __________________________________ -,and
each Y is independently N or CR5;
each R3 is independently H or optionally substituted -C1-6 aliphatic;
each R4 is independently -S(0)2NR2, -S(0)2R, -C(0)NR2, -C(0)R, or optionally
substituted -C1-6
aliphatic;
each R5 is independently R, -CN, -C(0)R, -C(0)NR2, or optionally substituted 5-
6 membered
heteroaryl having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur;
each m is independently 0, 1, or 2,
p is 0, 1, or 2, and
each R is independently H, optionally substituted -C1_6 aliphatic, optionally
substituted 3-8
membered saturated or partially unsaturated monocyclic carbocyclyl, or
optionally substituted
3-8 membered saturated or partially unsaturated monocyclic heterocyclyl having
1-2
heteroatoms independently selected from nitrogen, oxygen, or sulfur.
100391 As defined generally above, L' is a covalent bound, or a C1-6
bivalent straight or
branched hydrocarbon chain wherein 1, 2, or 3 methylene units of the chain are
independently and
optionally replaced with -N(R)-, -0-, or -C(0)-.
14
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
100401 In some embodiments, LI is a covalent bond.
100411 In some embodiments, LI- is C1.6 bivalent straight or
branched hydrocarbon chain
wherein 1, 2, or 3 methylene units of the chain are independently and
optionally replaced with -
N(R)-
100421 In some embodiments, LI- is C1.6 bivalent straight or
branched hydrocarbon chain
wherein 1, 2, or 3 methylene units of the chain are independently and
optionally replaced with -0-
100431 In some embodiments, LI- is C1.6 bivalent straight or
branched hydrocarbon chain
wherein 1, 2, or 3 methylene units of the chain are independently and
optionally replaced with -
C(0)-
100441 In some embodiments, 1-1 is -NH-. In some embodiments, Ll is -
NH-CH2-. In some
embodiments, LI- is -NH-CH2-CH2-. In some embodiments, LI- is -CH2-. In some
embodiments,
,,.kil,)\= ,NH...?µ
H --i
LI- is \ a= . In some embodiments, LI- is \ . In some embodiments, LI-
is \
N
. In some embodiments, Ll- is -X
. In some embodiments, LI- is -CH=CH-. In some
..s.c......,..),õ
NC-2,õ..
embodiments, L' is . In some embodiments, L1 is
. In some embodiments, LI-
is -NH-C(0)-.
100451 In some embodiments, LI- is selected from those depicted in
Table 1, below.
100461 As defined generally above, Ring A is selected from -- .
M....,....)- 1-0
,
I-0
EX> HO 1 ( _________________________________ \
___________________________________________ 7H I (N 10 . 0
,
,
\ N
410
N' H
0 Ns
/ N
H , and , each of which is optionally substituted.
100471 In some embodiments, Ring A is optionally substituted ..
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
100481 .. In some embodiments, Ring A is optionally substituted ''`=."---- .
HO 100491 In some embodiments,
Ring A is optionally substituted .
100501 In some embodiments, Ring A is optionally substituted
100511 In some embodiments, Ring A is optionally substituted
He 100521 In some
embodiments, Ring A is optionally substituted .
I K _______________________________________________________________ \
N H
100531 In some embodiments,
Ring A is optionally substituted / .
100541 In some
embodiments, Ring A is optionally substituted .
S I--
100551 In some
embodiments, Ring A is optionally substituted N S.
0-
100561 In some
embodiments, Ring A is optionally substituted . 0
'fl\ N
,
N
100571 In some
embodiments, Ring A is optionally substituted H .
H
NI,
N
/
100581 In some
embodiments, Ring A is optionally substituted .
N
IS (R1)n 1 I
..,../.
100591 In
some embodiments, Ring A is selected from ,
(R1)n
(Ri)n
R1)n
1
(R1)n 1¨Ã31
(1¨\ 7H
1---C-
16
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US20211072684
iIII0
¨1¨(R1)n HSN (Ri)n
0
_______________________________________________________________________________
____ (Ri)n
\
N (R1 )n 1\11
, and
(Ri)n , wherein each RI- is independently R,
halogen, -CN, -C(0)R, -C(0)NR2, -OR, -SR, -S(0)2NR2, or -S(0)2R, and each n is
independently
0, 1, 2, or 3, wherein each R is independently as defined herein and as
described in embodiments
herein.
100601
In some embodiments, RI- is R. In some embodiments, It" is halogen. In
some
embodiments, R" is -CN. In some embodiments, R" is -C(0)R. In some
embodiments, R" is -
C(0)N16. In some embodiments, It" is -OR. In some embodiments,
is -SR. In some
embodiments, RI is -S(0)2NR2. In some embodiments, RI is -S(0)2R.
100611
In some embodiments, each It' is independently H, halogen, -C1_6
aliphatic optionally
substituted by 1-6 halogen, 3-8 membered saturated or partially unsaturated
monocyclic
carbocyclyl optionally substituted by 1-6 halogen, or 3-8 membered saturated
or partially
unsaturated monocyclic heterocyclyl having 1-2 heteroatoms independently
selected from
nitrogen, oxygen, or sulfur optionally substituted by 1-6 halogen.
100621
In some embodiments, each RI- is independently H, -CF3, -C(0)NH2, -CH3,
-CH2CH3,
-OCH3, -CI-IF2, -0CF3, -OCHF2, -SCF3, -Cl, -S(0)2-NH2, -OCH2CH3, -F, -
C(0)NHCH3, -CN, -
0
S(0)2-CH3, -OCH(C113)2, -CH(CH3)2, -C(CH3)3, T_T , or
100631
In some embodiments, each RI- is independently selected from those
depicted in Table
1, below.
100641
In some embodiments, n is 0. In some embodiments, n is 1. In some
embodiments, n
is 2. In some embodiments, n is 3.
17
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
,
100651 In some embodiments, Ring A is selected from IC-9¨R 1 1
DD /CO¨ , and
iCaR1, wherein each of Rl is as defined above and described in embodiments
herein, both
singly and in combination.
R1
1101 R1$
100661 In some embodiments, Ring A is selected from R1 , R1
R1, and
N ,--
R1, wherein each R1 is as defined above and described in embodiments herein,
both
singly and in combination.
100671 In some embodiments, Ring A is CF3, CF3, eF3,
OF3, cF3, CF3,
F
CHF2 CF3
.
0¨CF3
1 ( __________ >¨ = 0/
. .
0¨CF3 , 41, CI
0 µN---
4. II
=S¨N I-12 o1 I I
F
/
i 3CF
is, r_\4=5_cF3 = a = s
, ,
,
CI
0-CF3
= CI
F CF3
FF3 p F3
. F
0¨CF3, . 0 . 0
1, Cl
18
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
F CI
. CN S-CF3
Ilk
. 0
AA
-
CF3, =' a 0¨
. ¨
. 0¨K
=, ,
-N
'N---.,
0
CN 0-
-NH2
lik F
lik
F


\
0
CI
0 0
Ok
4. 4. CI
OH CI , NH2, 0-
F
F-(
F
0- F 0 F
11 F
* F . CI
. 4110 CN * F
F,
F
CF3 CI CN Cl
*
lik F
. Mk lik 41.* CI
F ,
,
CI
0 F
. .
,,. F\ C Ci
Ag
CN
CI 40 01-
F3F
,
0
--II-.0--.<
40 CF3 \s/\-) 1¨( NH
---
N
:
H
\ / \ CH F2 EX--
_________________________________________________________________ .
_____________
N N N N
, i__ / ,
,
19
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
...tr.-
Hµ ____________ --<> KS 01101 I-- )-( 00
N N N CF, cF37
,--5-
Aii= dib,h CF3
VIIIPI CF3 1 (N4
or C F3 .
,
,
100681 In some embodiments, Ring A is selected from 1-0¨cF3
,
0 c3
Ho_cF3 Ho...cF3 C F3
. C F3 1---µ _____>-CF3
, and N
100691 In some embodiments, Ring A is selected from those depicted
in Table 1, below.
R2
Rw
100701 As defined generally above, Ring B is selected from Rw 0
R4 7
R2 R2
R2 WI'
0 oie\ cYN'i :''N (R3)p
N _______________________________________________________________ 1
R RCw Rw R4-........- N w , 0 , and Rw-
<N-A
,
wherein each of R2, le, IV', p, and R4 is as defined herein and described in
embodiments herein,
both singly and in combination.
R2
1110
100711 In some embodiments, Ring B is Rw
, wherein each of R2 and It' is as
defined herein and described in embodiments herein, both singly and in
combination.
Rw
lb
100721 In some embodiments, Ring B is R4
, wherein each of le and It' is as
defined herein and described in embodiments herein, both singly and in
combination.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R2
[0073] In some embodiments, Ring B is Rw
, wherein each of R2 and It" is as defined
herein and described in embodiments herein, both singly and in combination.
R2
[0074] In some embodiments, Ring B is Rw
, wherein each of R2 and Rw is as
defined herein and described in embodiments herein, both singly and in
combination
Rw
100751 In some embodiments, Ring B is R4
, wherein each of R4 and It' is as
defined herein and described in embodiments herein, both singly and in
combination.
R2
N
100761 In some embodiments, Ring B is Rw
, wherein each of R2 and It' is as defined
herein and described in embodiments herein, both singly and in combination.
(R3) p
[0077] In some embodiments, Ring B is 0
, wherein each of R1 and p is as
defined herein and described in embodiments herein, both singly and in
combination. In some
embodiments, Ring B is 0 or 0
[0078] In some embodiments, Ring B is
, wherein R" is as defined herein and
Rwi
described in embodiments herein. In some embodiments, Ring B is H
, wherein It'
is as defined herein and described in embodiments herein.
[0079] In some embodiments, Ring B is selected from those depicted
in Table 1, below.
21
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Br
CI i,161
H
nr N y \i.L)
[0080] As defined generally above, IV' is selected from
0 , and .
_N
nr N y
Br
H.i.b
[0081] In some embodiments, IV' is 0
. In some embodiments, Rw is . In
Br
15 ,b ,

GI
-N
_N
some embodiments, IV' is
. . In some embodiments, It' is . In some
CI
Brtb iviNb
embodiments, It" is ...'"===^" . In some embodiments, IV is
. In some embodiments,
CI
R' is ---7--- .
[0082] In some embodiments, It" is selected from those depicted in
Table 1, below.
[0083]
As defined generally above, each R2 is independently selected from -OR, -
C(0)NR7,
(R )m (R5) (R5)m
, ,111 (R5)m
0
HN-I-Y
C, H-\ 111\1)(R5)m
...,õ NH HN.,,N
optionally substituted -CI-6 aliphatic, _I_ ' -.1
N
- - LI
-
R5 (R5),õ ,-,---- \
,, -
-.1
() N
--., .
\ N N
. ________________ , and
, wherein each of Y, m, and R5 is as defined herein and
described in embodiments herein, both singly and in combination..
22
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
100841
In some embodiments, R2 is -OR. In some embodiments, R2 is -C(0)NR2. In
some
(R5)m
HN-I-Y
Ylr N
embodiments, R2 is optionally substituted -C1_6 aliphatic. In some
embodiments, R2 is
(R5)m (R5)m
1
.N N ,NH
. In some embodiments, R2 is ¨1--- . In some embodiments, R2 is
. In some
(R5)m 0
H¨ \
HN
HN,,,,,,- N
embodiments, R2 is ________ 1 . In some
embodiments, R2 is 1,.."0
. In some embodiments,
(R5)m ,----\
R'-', _______________________________ -
(),,, QIN =-=-µ1,___.t..N.,1¨\\
R2 is . In some embodiments, R2 is ¨ .
R5 R5
HN-I-Y HN-FN
i µµ

YN, N
100851
In some embodiments, R2 is ¨ . In some embodiments, R2 is ¨ . In some
R5 R5
1-11-1¨\ N¨N
, 1 \ i µµ
N -,....,,N N .., N
embodiments, R2 is ¨ . In some embodiments, R2 is
. In some embodiments, R2
R5 R5 R5
\ \
HN-k\ N¨Y
i ¨\\
Y.,N
.,.,, N ..., N
is ¨ . In some embodiments, R2 is ¨ . In some embodiments, R2 is
. In
23
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
N¨N N¨%
N tr.\ IN
some embodiments, R2 is ¨ In some embodiments, R2 is
In some
R5¨ N N (R5)m-0
N
embodiments, R2 is . In some embodiments, R2 is
___________________________________ . In some
R5
R5
N
embodiments, R2 is . In some embodiments, R2 is
___________________________________ . In some embodiments, R2
R5
I4jN ,N
is ¨ In some embodiments, R2 is ¨I-
24
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
\O
HN
0
0
\ D3C\
Z
N¨\\ N-R\
N¨\\
.-=,,_.,,N '-cf,N
4y N
100861 In some embodiments, R2 is selected from:
_______________________
\ \c)
NH
H2N 0 OJ
0 0 0
0 0 0 ,N
Q
<1\ ---N\ ''
N¨\\ N--\\ N--\\ N-II HN¨\\ N¨\\
N¨\\
iN &.=,,,.,õN sceõN cr. i\I 4.yN
&-,µ,.õN 4N
, , , _________________________________ ¨ ,
\o
0
Q
CN
< \
H2N
----\
N¨\\ N¨\\ N-N
% t / N¨\\i2 tc, ..N -c)N ,N =., .NH
c.r.N
iN 0
N
.....1._
, -ci-13, -
, , , , , _____ 5
N_F)o o
NE12 ,7.Z\-OH \7,N
<1\
N-N N-N N-N N¨A j 01 N¨\\
0
4N 1'N µfN 1.k,õN
lyN
N
CH2CH3, OCH3, H
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
/
0
HO
N1 ,
N\
Alf
c;N j
N - µ( -N
0 H H H
Oy NH2 \----.
NH2 CN
0..'=-r NC
N -cl -I
...- N --y. N --...,..;,-õN .-.......N
...,r..N
_________________________________________________ and .
/
\
-\
N-\\ N¨\\ N¨\\ N-\\
(.-A.,,N 4k..,N
'c,N N
[0087] In some embodiments, R2 is selected from __ , ,
_
r--
HN CI
(
CF3
<
c..,N 4.-k.N 4...yN ,-NNI",... N
--1- , and -OCH3.
[0088] In some embodiments, R2 is selected from those depicted in
Table 1, below.
[0089] As defined generally above, each Y is independently N or CR5.
[0090] In some embodiments, Y is N. In some embodiments, Y is CR5.
In some embodiments,
Y is CH.
[0091] In some embodiments, both Y are N. In some embodiments, both
Y are CR5. In some
embodiments, one Y is N, and the other Y is CR5. In some embodiments, both Y
are CH. In some
embodiments, one Y is N, and the other Y is CH.
[0092] In some embodiments, Y is selected from those depicted in
Table 1, below.
[0093] As defined generally above, each R3 is independently H, -
C(0)R, or optionally
substituted -C1_6 aliphatic, wherein R is as defined herein and described in
embodiments herein.
[0094] In some embodiments, R3 is H.
[0095] In some embodiments, R3 is -C(0)R.
26
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
100961 In some embodiments, R3 is optionally substituted -C1_6
aliphatic.
100971 In some embodiments, R3 is selected from H, -CH3, -CH2CH3, -
C(0)CH3, and
S.
100981 In some embodiments, R3 is selected from those depicted in
Table 1, below.
100991 As defined generally above, each R4 is independently -
S(0)2NR2, -S(0)2R, -C(0)NR2,
-C(0)R, or optionally substituted -C1,6 aliphatic, wherein each R is
independently as defined herein
and as described in embodiments herein.
1001001 In some embodiments, R4 is -S(0)2NR2
1001011 In some embodiments, R4 is -S(0)2R.
1001021 In some embodiments, R4 is -C(0)NR2
1001031 In some embodiments, R4 is -C(0)R.
1001041 In some embodiments, R4 is -optionally substituted -C1-6
aliphatic.
s 0 D n 0 0
0
N¨S
ii ¨1 1 3¨"N¨S1 1 \ ii
N¨- SI H2N¨g-1
H II
u
1001051 In some embodiments, R4 is selected from 0 , 0 0 '
0
0 0 H , H
H
0 u ,.. N y--...õ-0,--...õ.. N li...\
.._,0 N -Ir.\
CN¨F1 >-H A---10
0 0 0 0 0
0 ,
,
0 , H
H2N 0¨g-1
0
0 H
)r\N F3C"\N_su 1
H 11 0 H 11 0 0
--- \N¨g
H2N
0 H 8
o
NH
0 H
N ....1(/ H ---
-N
\---\ N¨I ¨SI I ¨N1-1
NI¨I
0 8 0 H H 8 8
H 8
HON___\ 0
N¨g ¨I
, and H ,LI
0 .
27
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
\ 9 1 0
N¨S-1 fh N¨Sii ¨I
H II H II
1001061 In some embodiments, R4 is selected from: 0 0
, and
0
H
H
0 .
1001071 In some embodiments, R4 is selected from those depicted in Table 1,
below.
1001081 As defined generally above, each R5 is independently R, -CN, -C(0)R, -
C(0)NR2, or
optionally substituted 5-6 membered heteroaryl having 1-2 heteroatoms
independently selected
from nitrogen, oxygen, or sulfur, wherein each R is independently as defined
herein and as
described in embodiments herein.
1001091 In some embodiments, R5 is R.
1001101 In some embodiments, R5 is -CN.
1001111 In some embodiments, R5 is -C(0)R.
1001121 In some embodiments, R5 is -C(0)NR2.
1001131 In some embodiments, R5 is optionally substituted 5-6 membered
heteroaryl having 1-
2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
1001141 In some embodiments, each R5 is independently selected from: H, -CH3, -
CD3,
\O \
\
0
HN H2N 0 0
Z Z NH \
0 0 0 0 0
0) 0 0 0 0
N ¨ ,N,s.
N\ _s...c C/Z7,---N i N ##,
=,..)>" #0,
F,
,
0 0 0 ii31____
H2N NH2 ,7.0H CN HO
-CN,
,
0 0
..\./
\j-LNA NN
, -CH2CH3, -C(0)CH3, -CH2C(0)NHCH3, H , and H .
28
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00115] In some embodiments, each R5 is independently selected from: -CH3, -
CH2C1120CH3,
R\
c>. H N
-CH2CF3, -CH2CH2C1, , , and .
[00116] In some embodiments, R5 is selected from those depicted in Table 1,
below.
[00117] As defined generally above, each m is independently 0, 1, or 2.
[00118] In some embodiments, m is 0. In some embodiments, m is 1. In some
embodiments,
m is 2.
[00119] In some embodiments, m is selected from those depicted in Table 1,
below.
[00120] As defined generally above, p is 0, 1, or 2.
[00121] In some embodiments, p is 0. In some embodiments, p is 1. In some
embodiments, p
is 2.
[00122] Tn some embodiments, p is selected from those depicted in
Table 1, below.
[00123] As defined generally above, each R is independently H, optionally
substituted -C1_6
aliphatic, optionally substituted 3-8 membered saturated or partially
unsaturated monocyclic
carbocyclyl, or optionally substituted 3-8 membered saturated or partially
unsaturated monocyclic
heterocyclyl having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur.
[00124] In some embodiments, R is H.
1001251 In some embodiments, R is optionally substituted -C1-6 aliphatic.
In some
embodiments, R is unsubstituted -C1-6 aliphatic. In some embodiments, R is -
C1.6 aliphatic
substituted 1, 2, 3, 4, 5, or 6 times by -halogen, -CN, or -NO2. In some
embodiments, R is -C1-6
aliphatic substituted 1, 2, 3, 4, 5, or 6 times by -F. In some embodiments, R
is -C1.3 aliphatic
substituted 1, 2, 3, 4, 5, or 6 times by -F. In some embodiments, R is -CH3.
In some embodiments,
R is -CH2CH3. In some embodiments, R is -CF3. In some embodiments, R is -CHF2.
1001261 In some embodiments, R is optionally substituted 3-8 membered
saturated or partially
unsaturated monocyclic carbocyclyl. In some embodiments, R is unsubstituted 3,
4, 5, 6, 7, or 8
membered saturated or partially unsaturated monocyclic carbocyclyl. In some
embodiments, R is
3, 4, 5, 6, 7, or 8 membered saturated or partially unsaturated monocyclic
carbocyclyl substituted
1, 2, 3, 4, 5, or 6 times by -halogen, -CN, -NO2, or -C1_6 aliphatic, wherein
the -C1_6 aliphatic is
optionally substituted 1, 2, 3, 4, 5, or 6 times by -halogen, -CN, or -NO2. In
some embodiments,
29
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R is 3, 4, 5, 6, 7, or 8 membered saturated or partially unsaturated
monocyclic carbocyclyl
substituted 1, 2, 3, 4, 5, or 6 times by -halogen. In some embodiments, R is
3, 4, 5, 6, 7, or 8
membered saturated or partially unsaturated monocyclic carbocyclyl substituted
1, 2, 3, 4, 5, or 6
times by -C1.6 aliphatic, wherein the -C1.6 aliphatic is optionally
substituted 1, 2, 3, 4, 5, or 6 times
by -halogen. In some embodiments, R is 3, 4, 5, 6, 7, or 8 membered saturated
or partially
unsaturated monocyclic carbocyclyl substituted 1, 2, 3, 4, 5, or 6 times by -
F. In some
embodiments, R is 3, 4, 5, 6, 7, or 8 membered saturated or partially
unsaturated monocyclic
carbocyclyl substituted 1, 2, 3, 4, 5, or 6 times by -C1.6 aliphatic, wherein
the -C1.6 aliphatic is
optionally substituted 1, 2, 3, 4, 5, or 6 times by -F.
1001271 In some embodiments, R is optionally substituted 3-8 membered
saturated or partially
unsaturated monocyclic heterocyclyl having 1-2 heteroatoms independently
selected from
nitrogen, oxygen, or sulfur. In some embodiments, R is unsubstituted 3, 4, 5,
6, 7, or 8 membered
saturated or partially unsaturated monocyclic heterocyclyl having 1-2
heteroatoms independently
selected from nitrogen, oxygen, or sulfur. In some embodiments, R is 3, 4, 5,
6, 7, or 8 membered
saturated or partially unsaturated monocyclic heterocyclyl having 1-2
heteroatoms independently
selected from nitrogen, oxygen, or sulfur, which is substituted 1, 2, 3, 4, 5,
or 6 times by -halogen,
-CN, -NO2, or -C1.6 aliphatic, wherein the -C1_6 aliphatic is optionally
substituted 1, 2, 3, 4, 5, or
6 times by -halogen, -CN, or -NO2. In some embodiments, R is 3, 4, 5, 6, 7, or
8 membered
saturated or partially unsaturated monocyclic heterocyclyl having 1-2
heteroatoms independently
selected from nitrogen, oxygen, or sulfur substituted 1, 2, 3, 4, 5, or 6
times by -halogen. In some
embodiments, R is 3, 4, 5, 6, 7, or 8 membered saturated or partially
unsaturated monocyclic
heterocyclyl having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur
substituted 1, 2, 3, 4, 5, or 6 times by -C1.6 aliphatic, wherein the -C1.6
aliphatic is optionally
substituted 1, 2, 3, 4, 5, or 6 times by -halogen In some embodiments, R is 3,
4, 5, 6, 7, or 8
membered saturated or partially unsaturated monocyclic heterocyclyl having 1-2
heteroatoms
independently selected from nitrogen, oxygen, or sulfur substituted 1, 2, 3,
4, 5, or 6 times by -F.
In some embodiments, R is 3, 4, 5, 6, 7, or 8 membered saturated or partially
unsaturated
monocyclic heterocyclyl having 1-2 heteroatoms independently selected from
nitrogen, oxygen,
or sulfur substituted 1, 2, 3, 4, 5, or 6 times by -C1.6 aliphatic, wherein
the -C1.6 aliphatic is
optionally substituted 1, 2, 3, 4, 5, or 6 times by -F.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
\ \
NH 0
H2N 1 1
0 0 0
0 0 0
1001281 In some embodiments, R is selected from
\
0
CN 31 ___
/
0 0 0 0
CN H2N NH2 OH
0 ''
, , , OU >", , , , ,
,
HO
N-1 '''' 7 Y7 -CH3, -CD3, -CH2CH3,
-CH2C(0)NHCH3,
7
H 0 \ H
_., N y---,.,,,0
- ',õ-----0.--- 11
---- --N -.'-'-'*'-'.-0--""-\ r N F3C
0 H 0
,
H
H2N H2Nõ,,. N1r, ....,---/ H2N
CN-1 OH \,`" )\/1 0-1 1
0 -.,..5:-
\-----
0 (3>
..
-CH2CH2C1õ and .
1001291 In some embodiments, R is selected from those depicted in Table 1,
below.
1001301 In some embodiments, the present invention provides a compound of
Formulas:
(R5), (R5),
HN-hY HN-FY
"N l; "N
L Ll
4111 I=

I 110 (R1)n
4111 Rw Rw Rw
II-a II-b
31
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
(R5), (R5),
HN-hY HN Y
I %%
Y-N. N ` "N
1 Li
0 __R1......,10. 0 ----6---- R1
Rw Rw
, ,
II-c II-d
(R5)1Ti
HN-hc --õ
Y., 'N (R)mTI1
I. Lii:), Li
Ri 0 (Ri)n
Rw Fe'
7 7
II-e II-f
(R5),, (R5)m
-- N ,- N
Li Rw Li-,,
II ¨(Ri)n ____________________________________________________________ Ri
N,..,..-..--
Rw
7 7
II-g II-h
(R5)m
L1 L1-Ø_
------e--- R1 Ri
Rw WI'
II-i II-j
(R5), (R5)
\
Li Li
* (Ri)n
IT' Rw
1 7
II-k II-1
32
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
(R5)m (R5)
N¨\\
N¨\\
N N
Li
Lt_so ___________________________________ R1
R1
Rw Rw
II-m II-n
(R5)m (R5)rri
N (f, ,N
Li
Ri (Ri)n
Rw Rw
11-0 II-p
(R5), (R5),
I \\N \\N
Li
Ri
Rw Rw =
II-q II-r
(R5),
(R5)m
\ N
Li
1.11 Ri
Rw , or Rw
II-s II-t
or a pharmaceutically acceptable salt thereof, wherein each of le, Y, m, n,
and R5 is as
defined above and as described in embodiments herein, both singly and in
combination.
1001311 In some embodiments, the present invention provides a compound of
Formulas:
33
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5
R5 \
N¨Y N¨Y
I %\ I \\
Y- N lirs= N
L1 0 Lt.irõ, 1
0 0 (R1)n Rw ______ (R )n
N,,...---
Rw
III-a III-b
R5
R5
\ \
N¨Y N¨Y
I 1µ
I \\
)/--- N Y-N, N
0
0 LL__0._
R1
Rw Rw
III-c III-d
R5
\ R5
..,
N¨Y
I
Ysõ, N
L10,., L1
1410 R1 0 (R1)n
Rw Rw
III-e III-f
R5
R5 ..,, ..,
I N I
N
Ll L1,,,
---y------..,--,
II ¨(R1)n _________________________________________________________ R1
N,.-...-
Rw , Rw ,
III-g III-h
R5 R5
N
L1.3..__Ri LL.,0õ.....R1
Rw Rw
M-i M-j
34
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
N I N
Li Li
SI 0 (R1)n el )-r.
Rw
N,,,...,----
Rw
III-k III-1
R5 R5
-,õ
..,
I I
0 Li,,,,o,_ Li
W Rw, 0 ---194---Ri
Rw
111-m 11I-n
R5
I --- N-\\
\
N ,,. N
L1
101 R1 le (Ri)n
RA Rw 14111
III-o III-p
\ L \
N
l Ll.õ,0
-/--N.õ,,.
N.,,,. ___________________________________________________________ Ri
,.;
Rw , Rw ,
III-q III-r
N-\\
Li..,___e_.. Li.,,a
Ri R1
Fr Rw
Ill-s M-t
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5
R5
\\N ,\\N
Li Li
(Ri)n
Rw 4111 Rw
111-u M-v
R5 R5
Li
W
Rw Rw
III-w III-x
R5
,\\N
14111 Ri
or Rw
M-y
or a pharmaceutically acceptable salt thereof, wherein each of Y, n, and
R5 is as
defined above and as described in embodiments herein, both singly and in
combination.
1001321 In some embodiments, the present invention provides a compound of
Formulas:
R5 R5
sNù\\
Nù\\
Rw 1410 Li 1110 (Ri)n
Rw, (R1 n
36
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
\ \
N-\\ N-\\
\ N \ N
___________________________________________________________________ Ri 10 eRi
Fe Fe
IV-c W-d
R5 R5
\ ...,
N-\\
I .,, N
--, N
Li..a Li
14011 Ri =0 (R )n
Rw Rw
IV-e IV-f
R5 R5
-...,
I ,,, N I
Li Li,,0
--...y./
II ¨(Ri)n _________ R1
..,....
Rw
IR' N , ,
IV-g IV-h
R5 R5
-,., ,....,
I
,- N I N
L1
---).---R1 Ri
Rw Rw
IV-i IV-j
R5 R5
-...,.
I I
,- N
II
Rw
Li Li
N
411 0 (R1)n IR
'
IV-k IV-1
37
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
N
N
Li
R
14111
Rw Rw,
IV-m IV-n
R5
N N
Li
Rw
4111 IIIIIRi Rw = (Ri)n
IV-o IV-p
N N
Li L1-0
II ¨(R1)n
N '
Rw Rw
IV-q IV-r
N¨\\
N¨\\
N N
Li R1 L
R1
Rw Rw
R5
R5
,\\N
,\\N
Li 1
=(Ri)n 411) N )n
Rw
IV-u IV-v
38
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
R5 R5
\\N
,\\N
N N
Rw, _____________________________________ R1
Rw411 -------tes--- R1
IV-w IV-x
R5
\\
,N
N
R1
or Rw mill ,
IV-y
or a pharmaceutically acceptable salt thereof, wherein each of le, Ll, R", n,
and R5 is as defined
above and as described in embodiments herein, both singly and in combination.
1001331 In some embodiments, the present invention provides a compound of
Formulas:
R5
, R5
N-\\ R5 \
N, N N-\\ N-\\
Ll R1
0 I
0 Li
ilki 0
Rw R1
R1 Rw R1 R1 Rw R1
, ,
V-a V-b V-c
R5
.õ_
R5 R5
Ll R1
Rw lel Ri Li Li
R1 Rw R1 fel R1 Fe
V-d V-e V-f
39
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5
R5 R5
I
L1 W
01 I I 0 R1 Li
01 Li,,
Rw N
Ri Rw Ri Ri Rw
R1,
,
V-g V-h V-i
_.¨

N¨\\
N¨\\ --
N¨\\
\ \
=õ N N
l
RWZIX L R1 0 Ri Li L 1
Ri Rw R1 4110 R1 Rw
N..õ...,....R1
V-j V-k V-1
R5
R5
R5
,.\\N
R1
Rw L ,N N
l N
4111 0 R1 L1
10 0 L1
R1 Rw R1 R1Rw
R1
, ,or
V-m V-n V-o
, or a pharmaceutically acceptable salt thereof, wherein each of Rl, IV', Ll,
and R5 is as defined
above and as described in embodiments herein, both singly and in combination.
[00134] In some embodiments, the present invention provides a compound of
Formulas:
Rw Rw
Ll Ll
R\ IP 1110 (R1)n 0 0
N \ N \o
I I
R R
VI-a VI-b
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
Rw Rw
R lelL1-0 Ll
R1 (:)\\ lel -----ei"-- R1
RS RäS
N \ \ N \\
R R
7 7
VI-c VI-d
Rw
L,a R el
Rw
L1
R 4111 R1 0
(R1)n
R äs
I 0 N æ`
R H 0
7 ,
VI-e VI-f
Rw Rw
Li Li..,
0 n I 1)n R ù(R _______
411111 R1
\\ el Lt-
R, R,
7 7
VI-g VI-h
Rw Rw
Li Lio,,,
R\ I. -----E3L- R1 R el R1
R, R,
N \\(-)
7 7
VI-j
Rw Rw
R POP 11101
Ll R1 0\\ is L1 0
R. ,-S, R1 R, ,-S, R1 RI
0
R R
7 7
VI-k VI-1
Rw
L1
0 4111
R
W
N \\
I 0
R
VI-m
41
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
or a pharmaceutically acceptable salt thereof, wherein each of R, RI, LI, R",
and n is as defined
above and as described in embodiments herein, both singly and in combination.
1001351 In some embodiments, the present invention provides a compound of
Formulas:
Rw Rw

1
:& le L Ll
I
0 (R1)n 0
\\ \ N \\ \ N Nõ,...-
R, S R, ,,.S
I 0 I 0
R R
, ,
VH-a VH-b
Rw Rw
0
L1 L1
L1

R1 R\ LY õ.--..,-,......,N
R1
\\
R, .S R, _,S
N \\ N \\
I I 0
R R
, ,
VH-c VII-d
Rw
Rw
R\.1_1 0
0
\\ -=-:=..,,N R1
(R1)n
R õS.
N
I N \\,-µ
R H '
, ,
VH-e VH-f
Rw Rw
I
0 4Y -(Ri)n 0 & R1
\\ '., N N,../- \\ \ N
R, S R, S
N \\,-µ N \\,-µ
H ' H '
, ,
VII-g VH-h
Rw Rw
Lle),__ &L
N
la
0 0
\\ ,..-:.,.. R1
R, ,S R, ,S
N H ,-, \\ H ,-, N \\ ' '
, ,
VH-i VII-j
42
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
Rw Rw
R
R
L
0õ L=R1 0
N
R, Ri -111W- Ri
N N
0 R1 0
VII-k V11-1
Rw
Ll
R.S N
N \\0
VII-m
or a pharmaceutically acceptable salt thereof, wherein each of R, Ll, R",
and n is as defined
above and as described in embodiments herein, both singly and in combination.
1001361 In some embodiments, the present invention provides a compound of
Formulas:
Rw¨<CN¨L1 =¨(R1)n
N
V111-a V111-b
Rw<N_L1R1
Rw¨<CN¨L1¨¨R1
V111-c V111-d
Rw¨.<0 ¨
N L R1
Rw¨<C 1
N¨L ¨0¨R1 R1
R1
VIII-e VIII-f
Rw 1
N¨L Rw ¨<CN
N R1
R1 R1,
VHI-g VHI-h
or a pharmaceutically acceptable salt thereof, wherein each of IV', and n
is as defined
above and as described in embodiments herein, both singly and in combination.
1001371 In some embodiments, the present invention provides a compound of
Formulas:
43
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
(R5), (R5),
HN-hY HN-FY
, ,µ I µµ
Yy=N N Y,-,rN
L1 Ll
Rw N
0 (R1)n ________________ (R1 )n
..,-:.\.,
IR'--,' N N.,..,...;:-
IX-a IX-b
(R5),, (R5),õ
HN-hY HN-FY
I µN , IN
6
L1.0 L1
R1 1; --:...õ.., N R1
.
Rw N , Rw Y'k'N ,
IX-c IX-d
(R5)n,
HN-FYµ
Y...., \ N (R5)m
õIr.. 1_1,0, 0,... L1 0
I (R1 )n
RwN R1
Fe'
IX-e IX-f

(R5)m-0 (R56
,- N
Li L1-0
/ .
CY 1 )n (R _____________________________________________ I R1
Rw
-,,, N
Rw
IX-g IX-h
(R5)m--ON (R5)m-0
0 Li. ----- ------feL_ CY-
--. N R
Rw L1 R1 Rw 1
IX-i IX-j
44
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
(R5)m (R5)
\ V \ \\
Li Li
0 (R1 )n I ______ (Ri)n
Rw---.IN
Rw *---..-,...N N
IX-k IX-1
(R5), (R5)
N¨\\ N¨\\
\ \
Li
,
I Ri I
\ Rw---,N ----3).---R1
Rw N
IX-m IX-n
(R5), (R5),
N¨%\\N
\ V
s., N
N
L1Ø,. Li
..," ,
I & 0 (Ri)n
Rw\ N R1
Rw
IX-o IX-p
(R5), (R5),
e,\N
N N
Li
/Li......-).R1
J r
N ..s.
R,A,--_,I N
Rw-C-Lr--
IX-q IX-r
(R5),
(R5),
I \\N
1 \\N
N N
L1 ,
I
RwN -'s---Ã:;3\-----R1 \ N Ri
, or Rw ,
IX-s IX-t
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
or a pharmaceutically acceptable salt thereof, wherein each of RI, LI, IV', Y,
m, n, and R5 is as
defined above and as described in embodiments herein, both singly and in
combination.
1001381 In some embodiments, the present invention provides a compound of
Formulas:
R5
R5 \
N¨Y N¨Y
I \\ I 1µ
N lii-. N
,,.Li
,- ._-
1110 (R1)n r
Rw
\ N
Rw------N N,,,,-;:-
X-a X-b
R5
R5
\ \
NY N-Y
I µµ
I \\
N N
-..,-
-...,-
LL,0,_ L1
--16,._
R1
---,-.,,,,. R1
N
Rw ,
X-c X-d
R5
\ R5
--õ,
N¨Y
I" H N
Yy N
,....y.-1_1,,,c, Ll
I Rw 110 (R1)n
Rw-N R1 --. N
X-e X-f
R5
I
..= N
Ll L1,0
/
Cir '11(R1)n I _________ R1
--.. Rw N N.,.....--- Rw --.... N
X-g X-h
46
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
0 0\1
Li,õ,,c),_
-0 L1 R1 --'---e4--- -CC
R1
Rw , Rw ,
X-i X-j
R5 R5
I N
I ..-
,--
Li Li
I 10 (R1)n Rw I 'r
Rw
\ N \ N N ....5.:,
, ,
X-k X-1
R5 R5
,.
,..,
I I
L Rw N Rw\ Li
--- ,
I _________ Ri
\ N R '
X-m X-n
R5
-_
Rw 0 ____ Ri Li le
I .----Y (R1) n
N Rw'N.,.. N
X-o X-p
C7,11-\\
\ N N \ N N
Li o
Y .-(R 1 )n Cr Ri
--,,,N N ,,- \ N
Rw Rw Li
X-q X-r
47
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
N-\\
N-\\
N
N
Li
N RW R L--R1 N
X-S X-t
R5
R5
,N
.o N N N
r. Li
(R)n
RN
X-u X-v
R5 R5
,N
Li
RwN Rw N R
X-w X-x
R5
\\N
_or
N or Rw R
X-y
or a pharmaceutically acceptable salt thereof, wherein each of RI, LI, Rw, Y,
n, and R5 is as
defined above and as described in embodiments herein, both singly and in
combination.
1001391 In some embodiments, the present invention provides a compound of
Formulas:
48
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
N
\N¨

µ. \\
N¨\\
..,r.,1 'cl
Li Li
.-- .
I I '-il-

(R1 )n
Rw_
\ N (110 (Ri)n
Rw-z-s,...,. N N,.,,..--
XI-a XI-b
R5 R5
\ \
N¨\\ N¨\\
,=.õ.,N
LL..,0 ___________________________________ Ri Rw Li
_CrRw- N N --..-R1
XI-c XI-d
R5 R5
\N-\\ -..,..
I N
--, Nir.,,
Li Li
I 1 0 (R1)n
Rw'N a Rw N Ri \ ,
XI-e XI-f
R5 R5
.,..,
\ON
Li Liõ0
N R
r )n ________________ R1
\ N .,.:.5% \ N
Rw w I
XI-g XI-h
R5 R5
0 )0N
C Li r
N --Ãa--R1 -0(1-1.0"-R1
Rw Rw ,
XI-i XI-j
49
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R5 R5
I
N I *''
A\I
L (Ri)n
Li i
I 0 (Ri)n I )-r--
\ N \ N N,.,,-
Rw Rw
XI-k XI-1
R5 R5
-,õ
....,
-- N
Li...,0 Li
./
I ________ Ri I _1
\ N \ N ----.-C3L--R
Rw Rw
XI-m XI-n
R5
-..,
N
L1-.0_ Li
/
(Ri)n
I 0
\ N Ri
Rw--k N
Rw
XI-0 XI-p
\ N
Li',,o
Cr _______________________________________________________________
:a 1T-(Ri)n
\ N N_.,..õ-- \ N
Rw Li Ri
Rw
XI-q XI-r
N-k
N-µ V
N
Rw ---.=..,N R',
Rw--:-..N Ri
XI-s XI-t
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
R
R5 5
\NI ,\\N
N
N
\ N lb (Ri)n I 1
Rw, N N
Rw
XI-u XI-v
R5 R5
NN NN
1,0L Li
_________________________________________ Ri
Rw'......N
Rw--,.....,,, N Ri
XI-w XI-x
R5
N,\\N
1
_or L
-.., N '-'10-'.= R1
or Rw ,
XI-y
or a pharmaceutically acceptable salt thereof, wherein each of IV, L', R", n,
and It5 is as defined
above and as described in embodiments herein, both singly and in combination.
1001401 In some embodiments, the present invention provides a compound of
Formulas:
R5 R5
'N-\\ R \
Rw2Li Ri
/ 1
I Li L
/ C
\ N 0 I
Ri
Rw R ' Ri R1 RwY \ N N /
Ri
XII-a XII-b XII-c
51
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
R5
R5 R5
\ON 0
ti- Nis.-
R 1
Li 10 R 1 a Ll iiii Ll
N ,CY WI
\ N N /
Ri IR' Ri 111111AP Ri Rw W
, ,
XII-d XII-e XII-f
R5
R5 R5
,.- N
,-
Li Ri N N
/
1
,-' L1 L 1
./
\ N
Rw 0 Ri 1 1 )a
\ N 110 \ N N /
R 1 Rw R , ' R 1 Rw R 1
,
'
XII-g XII-h XII-i
Q¨\\ -- --
=,, N /N¨
L1
\ \
'.., N
C
R1 Y1110 L1
\ N ..-" ./
Rw Ri 1 1 I 1
' \
Ri Rw N R1 .1 R1, Rw
N
,
XII-j XII-k XII-1
R5
R
R5 5
N,\\N
\\N
r L1 0 R1 N
Rw_o R1 N
..---
..1y Li L
1
----..N \, N
W Rw R1 411}P
R1, or Rwe N.......
R1
,
XII-m XII-n XII-o
or a pharmaceutically acceptable salt thereof, wherein each of Rl, Ll, R", and
R5 is as defined
above and as described in embodiments herein, both singly and in combination.
1001411 Without wishing to be bound by any specific theory, the compounds
provided herein
are capable of covalently binding to an amino acid residue (such as cysteine,
lysine, histidine, or
52
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
other residues capable of being covalently modified) present in the binding
pocket of a target
protein, for example, TEAD, thereby irreversibly inhibiting the protein. In
some embodiments, a
warhead group is capable of covalently binding to cysteine. In some
embodiments, a warhead
group is capable of covalently binding to serine. In some embodiments, a
warhead group is capable
of covalently binding to lysine. In some embodiments, a warhead group is
capable of covalently
binding to Cys359 of hTEAD1, Cys405 of hTEAD1, Cys380 of hTEAD2, Cys368 of
hTEAD3,
and/or Cys367 of hTEAD4. In some embodiments, a warhead group is capable of
covalently
binding to Ser356 of hTEAD1, Ser345 and/or Ser377 of hTEAD2, Ser365 of hTEAD3,
and/or
Ser364 of hTEAD4. In some embodiments, a warhead group is capable of
covalently binding to
Lys336 of hTEAD1, Lys357 of hTEAD2, Lys345 of hTEAD3, and/or Lys344 of hTEAD4.

Representative reference amino acid sequences of human TEAD1, human TEAD2,
human
TEAD3, and human TEAD4 include UniProt KB ID P28347-1 (SEQ ID NO: I),
UniProtKB ID
Q15562 (SEQ ID NO: 2), UniProtKB ID Q99594 (SEQ ED NO: 3), and UniProtKB ID
Q15561
(SEQ ID NO: 4), respectively. Below is the sequence alignments of TEAD
coactivator binding
domains, which is shown in Table 1 of "Targeting Transcriptional Enhanced
Associate Domains
(TEADs)," J. Med. Chem. 2018, 61, 5057-5072, the entire content of which is
incorporated herein
by reference.
SEQ ID liTEADI
2a6WQGRSIGTTKEREVEFSAFLEQQRDPDSYNKEILEVHIGHANHSYSDELLESVDIRQ1YDKEPEKKGGLKEn5
NO: 5
SEQ hTEAD2
'''WQARGLGTARLQLVEFSAFVEPPDAVDSYQRHLEVH1SQHCPSPGAPPLESVDVRQIYDKEPEKKGGLRE'
NO: 6
SEQ IITEAD 3 'WQDRTIAS SRLRELEYSAFIVIEVQRD PDTY SKIILFV1I IGQINP
AFSD PPLEA VD VRQIYDKEPEKKGGEKE'
NO: 7
SEQ hTEAD4
2"WQGRSVASSKLWNILEFSAFLEQQQDPDTYNKHLEVHIGQSSPSYSDPYLEAVDIRQIYDKFF'EKKGGLKD283
NO: Si
SEQ liFEAD I 2761_,EGK.GPQN NEEL VKENN'AIDEN CN1Q-1313 A GA
FYGVTSQYESSENAITVICSTKVCSEGKQVVEK33'
NO: 5
SEQ 1iTEAD2
'"LYDRGEPHAFFLVKFWA_DENWGPSGEEAGAGGSISSGGFYGVSSQYESLEHMTLTCSSKVCSEGKQVVEIC"7
NO: 6
SEQ hTEAD3 2851_,YEKGPPNAFFLVI(_FWAJDENSTIQ-EGPGA
FYGVSSQYSSADSIVITISVSTKVCSEGKQVVEK315
NO: 7
SEQ hTEAD4 '"LFERGPSNAFFLVKFWADLNTNIE-DEGSS FY G
VSSQYESPENM1ITC STK VC SEG KQVVEK3"
NO: 8
SEQ ID liFEAD1
337vErEyARFENGREVYRINRSTMCEYMINFINKLKHLPEKYMNINSVLENFM,1_,VVTNROWET1.1_,CMACV"

NO: 5
53
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
SEQ ID hTEAD2
358VETERAQLEDGRFVYRLLRSPMCEYLVNITLIIKLRQLPERYIVIMNSVLENFTILQVVTNRDTQELLLCTAYV42
7
NO: 6
SEQ ID hTEAD3 '46VETEYARLENGREVYRIIIRSPNICEYMINFIIIKLKI-
ILPEKYMNINSVLENTTILQVVTSRDSQEILLVIAFV415
NO: 7
ST:,,Q ID liFEAD4 3I'VETEYA R YENGRYSYRHARSPLCEYMINFTH K L.KHLPEK
YMMNSVIENFTILQVVIN-RDTQEME,CIA \TV"
NO: 8
SEQ ID liTEAD I 4 7FEVSNSE,HGAQHHIYRLVKDI'6
NO: 5
SEQ ID hTEAD2 42817ENISTSERGAQHHIYRLVRD447
NO: 6
SEQ ID hTEAD3 416FEVSTSEHG AQI-IIIVYKINK D435
NO: 7
SEQ ID lirEAD4 41FEVSASEHGAQ141-1IYRLVIKEI 3'
NO: 8
1001421 Exemplary compounds of the invention are set forth in Table 1, below.
Table 1: Exemplary Compounds
I-1 1-2 1-3
H - -
H
(R) F )/
0
\-0
1-4 1-5 1-6
F _
` H
F -----,..."N
iNf.:- N H F - (R)
---........,....õ..)L N r:N. NH F \ (S)
F
N iv 0
F
F
1-7 1-8 1-9
\ F ,-.,%'-'ir = ,:,.1
F N N
CF,
0 N H
H' N
0
-.õ..N.,.).1.,
N
H
54
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
....._
NH
H ) = ' s;
L>====\i
N fik F
tr)
= , , o----- F
F
I-10 I-11 1-12
Br Br \
Z....N...7.N _NI
F
µ N----cµ
: H H
r
H
N N IS
N'ir.-"' 11110 F 0 P
N,,....,-;=====,..A. N.µ N,..,....õ--;.=-==,...A.
H 0 F H F
0 F
tC
¨ N
Br
1-13 1-14 1-15
\ \ a
t4-1 r.--
F
H . F ,N :
N
F N ==
F
=
Co
7:'----- N
at
1-16 1-17 1-18
CI Br Br
1
0 0 0
AN H 4#4 H (.-.
- -.&
N N
N -...., -..,
0 P 0 1 P 0 1
F
,,== N.,,,,;,.. ..---",--,)( µ'k ..-- N
kµ ....-' N
N =+, F N µ' r N \µ
F
1-19 1-20 1-21
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
/ / CI
N..,..... N ....,..-
H F
H E = X
....---..,
F
F
H
F
Br Br
1-22 1-23 1-24
CI Er Br
N
1 %
0 0 0
H
i4
.-N, .....-:?'..y N''.-.---. N-=:.:..,
0 I F 0 I I F 0 I I
F
N
kk..........,....õ....,,,,_ N .0 .........,;:,..õ.õ... N .=-=.õ
...õ-.1-4.... ,.. --...... `,.õ.... .-r."-..õ/...,
'"-... ---s-,õ, ..... S ....õ ......S
kk F N \N F .. N N`
F
H 0 F
H 0 F H
F
1-25 1-26 1-27
Br\ õ, \N---= F
==-=,,..., , 'Is : .F
l'. a

N...."
I H I H i
N.0 N . _
..--=
,.....,-?.,, N õ..-=,.
,.., r- =[--- T =µ-) t 11
-, J-. , Ni _-:)., F s...,......---
o I =_, ij N õ,,.. ...F
N. . ,.... , .....
%.....- ''''...-.
, ,.......
1 HN- b
1-F
F (...". 0 HN: b
F F
...;,..:Ni
Br'
1-28 1-29 1-30
Br.
..----N F CI,
1 ;...., N,......) -.--, . ,.r
'.?=N
\ ...,.., :. b
)- m .1: H !" r s....-
,N -3'... .:
" ' !
1.1
.1
Ni'
µ.8---,:-...- = .3, .N
,.. ....->s. .F
RN' s% = 'F ,---' HN = ',".
j
, o
Bf
1-31 1-32 1-33
Br\
,-----N ,¨

t....,...õ6
H
'
. ,....N., N.
,..., (---'..- ''k
E-IN' b f-F
P
56
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1-34
Br
H
111 1
N F F
H
1001431 In some embodiments, the present invention provides a compound set
forth in Table
1, above, or a pharmaceutically acceptable salt thereof.
1001441 In some embodiments, a compound of the present invention is not a
compound selected
from:
N .,3 7yN
0
P-1 P-2
1001451 The compounds of this invention can be prepared or isolated in general
by synthetic
and/or semi-synthetic methods known to those skilled in the art for analogous
compounds and by
methods described in detail in the Examples, herein. In some embodiments, the
present invention
provides an intermediate compound described in the Examples, or a salt
thereof.
4. Uses, Formulation and Administration:
Pharmaceutically acceptable compositions
1001461 According to another embodiment, the invention provides a
pharmaceutical
composition comprising a compound of this invention or a pharmaceutically
acceptable derivative
thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The
amount of compound
in compositions of this invention is such that is effective to measurably
inhibit TEAD, or a variant
or mutant thereof, in a biological sample or in a patient. In certain
embodiments, the amount of
compound in compositions of this invention is such that is effective to
measurably inhibit TEAD,
or a variant or mutant thereof, in a biological sample or in a patient. In
certain embodiments, a
composition of this invention is formulated for administration to a patient in
need of such
57
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
composition. In some embodiments, a composition of this invention is
formulated for oral
administration to a patient.
1001471 The terms "patient" or "subject" as used herein, means an animal,
preferably a
mammal, and most preferably a human.
1001481 The term "pharmaceutically acceptable carrier, adjuvant, or vehicle"
refers to a non-
toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological
activity of the
compound with which it is formulated. Pharmaceutically acceptable carriers,
adjuvants or vehicles
that may be used in the compositions of this invention include, but are not
limited to, ion
exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as
human serum albumin,
buffer substances such as phosphates, 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.
1001491 A "pharmaceutically acceptable derivative" means any non-toxic salt,
ester, salt of an
ester or other derivative of a compound of this invention that, upon
administration to a recipient,
is capable of providing, either directly or indirectly, a compound of this
invention or an inhibitorily
active metabolite or residue thereof
1001501 As used herein, the term "inhibitorily active metabolite or
residue thereof' means that
a metabolite or residue thereof is also an inhibitor of TEAD, or a variant or
mutant thereof
1001511 Compositions of the present invention canbe administered orally,
parenterally, by
inhalation spray, topically, rectally, nasally, buccally, vaginally or via an
implanted reservoir. The
term "parenteral" as used herein includes subcutaneous, intravenous,
intramuscular, intra-articular,
intra-synovial, intrastemal, intrathecal, intrahepatic, intralesional and
intracranial injection or
infusion techniques. Preferably, the compositions are administered orally,
intraperitoneally or
intravenously. Sterile injectable forms of the compositions of this invention
cancan be aqueous or
oleaginous suspension. These suspensions cancan be formulated according to
techniques known
in the art using suitable dispersing or wetting agents and suspending agents.
The sterile injectable
preparation cancan 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
58
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
vehicles and solvents that can be employed are water, Ringer's solution and
isotonic sodium
chloride solution. In addition, sterile, fixed oils are conventionally
employed as a solvent or
suspending medium.
1001521 For this purpose, any bland fixed oil can be employed including
synthetic mono- or di-
glycerides. 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 can also contain
a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or
similar dispersing
agents that are commonly used in the formulation of pharmaceutically
acceptable dosage forms
including emulsions and suspensions. Other commonly used surfactants, such as
Tweens, Spans
and other emulsifying agents or bioavailability enhancers which are commonly
used in the
manufacture of pharmaceutically acceptable solid, liquid, or other dosage
forms can also be used
for the purposes of formulation.
1001531 Pharmaceutically acceptable compositions of this invention can be
orally administered
in any orally acceptable dosage form including, but not limited to, capsules,
tablets, aqueous
suspensions or solutions. In the case of tablets for oral use, carriers
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 cornstarch. When
aqueous suspensions are required for oral use, the active ingredient is
combined with emulsifying
and suspending agents. If desired, certain sweetening, flavoring or coloring
agents can also be
added.
1001541 Alternatively, pharmaceutically acceptable compositions of this
invention can be
administered in the form of suppositories for rectal administration. These can
be prepared by
mixing the agent with a suitable non-irritating excipient that is solid at
room temperature but liquid
at rectal temperature and therefore will melt in the rectum to release the
drug. Such materials
include cocoa butter, beeswax and polyethylene glycols.
1001551 Pharmaceutically acceptable compositions of this invention can also be
administered
topically, especially when the target of treatment includes areas or organs
readily accessible by
topical application, including diseases of the eye, the skin, or the lower
intestinal tract. Suitable
topical formulations are readily prepared for each of these areas or organs.
59
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00156] Topical application for the lower intestinal tract can be
effected in a rectal suppository
formulation (see above) or in a suitable enema formulation. Topically-
transdermal patches can
also be used.
1001571 For topical applications, provided pharmaceutically acceptable
compositions can be
formulated in a suitable ointment containing the active component suspended or
dissolved in one
or more carriers. Carriers for topical administration of compounds of this
invention include, but
are not limited to, mineral oil, liquid petrolatum, white petrolatum,
propylene glycol,
polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
Alternatively,
provided pharmaceutically acceptable compositions can be formulated in a
suitable lotion or cream
containing the active components suspended or dissolved in one or more
pharmaceutically
acceptable carriers. 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.
1001581 For ophthalmic use, provided pharmaceutically acceptable compositions
can be
formulated as micronized suspensions in isotonic, pH adjusted sterile saline,
or, preferably, as
solutions in isotonic, pH adjusted sterile saline, either with or without a
preservative such as
benzylalkonium chloride. Alternatively, for ophthalmic uses, the
pharmaceutically acceptable
compositions can be formulated in an ointment such as petrolatum.
[00159] Pharmaceutically acceptable compositions of this invention can also be
administered
by nasal aerosol or inhalation. Such compositions are prepared according to
techniques well-
known in the art of pharmaceutical formulation and can be prepared as
solutions in saline,
employing benzyl alcohol or other suitable preservatives, absorption promoters
to enhance
bioavailability, fluorocarbons, and/or other conventional solubilizing or
dispersing agents.
[00160] Most preferably, pharmaceutically acceptable compositions of this
invention are
formulated for oral administration. Such formulations can be administered with
or without food.
In some embodiments, pharmaceutically acceptable compositions of this
invention are
administered without food. In other embodiments, pharmaceutically acceptable
compositions of
this invention are administered with food.
[00161] The amount of compounds of the present invention that can be combined
with the
carrier materials to produce a composition in a single dosage form varies
depending upon the host
treated, the particular mode of administration. Preferably, provided
compositions should be
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the
inhibitor can be
administered to a patient receiving these compositions.
1001621 It should also be understood that a specific dosage and treatment
regimen for any
particular patient depends upon a variety of factors, including the activity
of the specific compound
employed, the age, body weight, general health, sex, diet, time of
administration, rate of excretion,
drug combination, and the judgment of the treating physician and the severity
of the particular
disease being treated. The amount of a compound of the present invention in
the composition also
depends upon the particular compound in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions
The Hippo Signaling Network
1001631 The Hippo signaling network (also known as the Salvador/Warts/Hippo
(SWH)
pathway) is a master regulator of cell proliferation, death, and
differentiation. In some
embodiments, the main function of the Hippo signaling pathway is to regulate
negatively the
transcriptional co-activators Yes-associated protein (YAP) and its paralogue,
the transcriptional
co-activator with PDZ-binding motif (TAZ; also known as WWTR1). The Hippo
kinase cascade
phosphorylates and inhibits YAP/TAZ by promoting its cytoplasmic retention and
degradation,
thereby inhibiting the growth promoting function regulated under the YAP/TAZ
control. In an un-
phosphorylated/de-phosphorylated state, YAP, also known as YAP1 or YAP65,
together with
T AZ, are transported into the nucleus where they interact with TEAD family of
transcription
factors to upregulate genes that promote proliferation and migration, and
inhibit apoptosis. In some
instances, unregulated upregulation of these genes involved in proliferation,
migration, and anti-
apoptosis leads to development of cancer. In some instances, overexpression of
YAP/TAZ is
associated with cancer.
1001641 Additional core members of the Hippo signaling pathway comprise the
serine/threonine
kinases MST1/2 (homologues of Hippo/Hpo in Drosophila), Lats1/2 (homologues of
Warts/Wts),
and their adaptor proteins Sav 1 (homologue of Salvador/Say) and Mob (MOBKL1A
and
MOBKL1B; homologues of Mats), respectively. In general, MST1/2 kinase
complexes with the
scaffold protein Say 1, which in turn phosphorylates and activates Lats1/2
kinase. Lats1/2 is also
activated by the scaffold protein Mob. The activated Lats1/2 then
phosphorylates and inactivates
61
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
YAP or its paralog TAZ. The phosphorylation of YAP/TAZ leads to their nuclear
export, retention
within the cytoplasm, and degradation by the ubiquitin proteasome system.
1001651 In some instances, Lats1/2 phosphorylates YAP at the [HXRXXS] (SEQ ID
NO: 9)
consensus motifs. YAP comprises five [HXRXXS] (SEQ ID NO: 9) consensus motifs,
wherein X
denotes any amino acid residue. In some instances, Lats1/2 phosphorylates YAP
at one or more of
the consensus motifs. In some instances, Lats1/2 phosphorylates YAP at all
five of the consensus
motifs. In some instances, Lats1/2 phosphorylate at the S127 amino acid
position. The
phosphorylation of YAP S127 promotes 14-3-3 protein binding and results in
cytoplasmic
sequestration of YAP. Mutation of YAP at the S127 position thereby disrupts
its interaction with
14-3-3 and subsequently promotes nuclear translocation.
1001661 Additional phosphorylation occurs at the S381 amino acid position in
YAP.
Phosphorylation of YAP at the S381 position and on the corresponding site in
TAZ primes both
proteins for further phosphorylation events by CK1o/E in the degradation
motif, which then signals
for interaction with the I3-TRCP E3 ubiquitin ligase, leading to
polyubiquitination and degradation
of YAP.
1001671 In some instances, Lats1/2 phosphorylates TAZ at the [HXRXXS1 (SEQ ID
NO: 9)
consensus motifs. TAZ comprises four [HXRXXS] (SEQ ID NO: 9) consensus motifs,
wherein X
denotes any amino acid residues. In some instances, Lats1/2 phosphorylates TAZ
at one or more
of the consensus motifs. In some instances, Lats1/2 phosphorylates TAZ at all
four of the
consensus motifs. In some instances, Lats1/2 phosphorylate at the S89 amino
acid position. The
phosphorylation of TAZ S89 promotes 14-3-3 protein binding and results in
cytoplasmic
sequestration of TAZ. Mutation of TAZ at the S89 position thereby disrupts its
interaction with
14-3-3 and subsequently promotes nuclear translocation.
1001681 In some embodiments, phosphorylated YAP/TAZ accumulates in the
cytoplasm, and
undergoes SCFP-TR"-mediated ubiquitination and subsequent proteasomal
degradation. In some
instances, the Skp, Cullin, F-box containing complex (SCF complex) is a multi-
protein E3
ubiquitin ligase complex that comprises a F-box family member protein (e.g.,
Cdc4), Skpl, a
bridging protein, and RBX1, which contains a small RING Finger domain which
interacts with E2
-ubiquitin conjugating enzyme. In some cases, the F-box family comprises more
than 40 members,
in which exemplary members include F-box/WD repeat-containing protein lA
(FBXW1A,
I3TrCP1, Fbxwl, hsSlimb, plkappaBalpha-E3 receptor subunit) and S-phase kinase-
associated
62
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
proteins 2 (SKP2). In some embodiments, the SCF complex (e.g., SCFOrrcP1)
interacts with an El
ubiquitin-activating enzyme and an E2 ubiquitin-conjugating enzyme to catalyze
the transfer of
ubiquitin to the YAP/TAZ substrate. Exemplary El ubiquitin-activating enzymes
include those
encoded by the following genes: UBA1, UBA2, UB A3, UBA5, UBA5, UBA7, ATG7,
NAE1, and
SAEL Exemplary E2 ubiquitin-conjugating enzymes include those encoded by the
following
genes: UBE2A, UBE2B, UBE2C, UBE2D1, UBE2D2, UBE2D3, UBE2E1, UBE2E2, UBE2E3,
UBE2F, UBE2G1, UBE2G2, UBE2H, UBE2I, UBE2J1, UBE2J2, UBE2K, UBE2L3, UBE2L6,
UBE2M, UBE2N, UBE20, UBE2Q1, UBE2Q2, UBE2R1, UBE2R2, UBE2S, UBE2T, UBE2U,
UBE2V1, UBE2V2, UBE2Z, ATG2, BIRC5, and UTC1. In some embodiments, the
ubiquitinated
YAP/TAZ further undergoes the degradation process through the 26S proteasome.
1001691 In some embodiments, the Hippo pathway is regulated upstream by
several different
families of regulators. In some instances, the Hippo pathway is regulated by
the G-protein and its
coupled receptors, the Crumbs complex, regulators upstream of the MST kinases,
and the adherens
junction.
VAP/TAZ Interaction with TEAD
1001701 In some embodiments, un-phosphorylated and/or dephosphorylated YAP/TAZ

accumulates in the nucleus. Within the nucleus, YAP/TAZ interacts with the
TEAD family of
transcription factors (e.g., human TEAD1 (UniProt KB ID P28347-1 (SEQ ID NO:
1)), human
TEAD2 (UniProtKB ID Q15562 (SEQ ID NO: 2)), human TEAD3 (UniProtKB ID Q99594
(SEQ
ID NO: 3)), and human TEAD4 (UniProtKB ID Q15561 (SEQ ID NO: 4)) to activate
genes
involved in anti-apoptosis and proliferation, such as, for example, CTFG,
Cyr61, and FGF1.
1001711 Proteomic and biochemical studies have shown that the TEAD (TEA
Domain)
transcription factors are palmitoylated at evolutionarily conserved cysteine
residues. Three
cysteine residues were found that are evolutionarily conserved and mutated to
serine in human
TEAD1 (C53S, C327S and C359S) to test whether the mutation affects TEAD1
palmitoylation.
The C359S mutant showed the greatest loss of palmitoylation, and C327S and
C53S also showed
decreased palmitoylation. These results suggest that C359 plays a critical
role in TEAD1
palmitoylation. Furthermore, combination mutation of all three cysteine
residues, C53/327/359S
(3CS), completely ablated TEAD1 palmitoylation, indicating that these residues
are involved in
TEAD1 palmitoylation. It has been found that TEADs undergo PAT-independent
autopalmitoylation, under physiological concentrations of palmitoy 1-CoA.
Furthermore,
63
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
autopalmitoylation plays critical roles in regulating TEAD-YAP association and
their
physiological functions in vitro and in vivo. Chan, et at. Nature Chem. Biol.
12, pages 282-289
(2016); Noland, et at. Structure, 24, 1-8 (2016); Gibault et at. J. Med. Chem.
61, 5057-5072
(2018). Therefore, palmitoylation of TEADs play important roles in regulating
Hippo pathway
transcriptional complexes.
[00172] In some embodiments, the compounds disclosed herein modulate the
interaction
between YAP/TAZ and TEAD. In some embodiments, the compounds disclosed herein
bind to
TEAD, YAP, or TAZ and prevent the interaction between YAP/TAZ and TEAD.
[00173] In some embodiments, the compounds described herein irreversibly
inhibit a TEAD
transcription factor. In some embodiments, the transcription factor is TEAD1.
In some
embodiments, the transcription factor is TEAD2. In some embodiments, the
transcription factor is
TEAD3. In some embodiments, the transcription factor is TEAD4. In some
embodiments, the
compounds described herein covalently bind to the TEAD transcription factor
(e.g., TEAD1,
TEAD2, TEAD3, or TEAD4). In some embodiments, the compounds described herein
irreversibly
inhibit the activity of a TEAD transcription factor (e.g., TEAD1, TEAD2,
TEAD3, or TEAD4). In
some embodiments, the compounds described herein covalently inhibit the
activity of a TEAD
transcription factor (e.g., TEAD1, TEAD2, TEAD3, and/or TEAD4).
[00174] In some embodiments, the compounds disclosed herein bind to TEAD1 and
disrupt or
inhibit the interaction between YAP and IEAD1. In some embodiments, the
compounds disclosed
herein bind to TEAD2 and disrupt or inhibit the interaction between YAP and
TEAD2. In some
embodiments, the compounds disclosed herein bind to TEAD3 and disrupt or
inhibit the
interaction between YAP and TEAD3. In some embodiments, the compounds
disclosed herein
bind to TEAD4 and disrupt or inhibit the interaction between YAP and TEAD4.
[00175] In some embodiments, the compounds disclosed herein bind to TEAD1 and
disrupt or
inhibit the interaction between YAP and ItAD1. In some embodiments, the
compounds disclosed
herein bind to TEAD1 at C359, and disrupt or inhibit the interaction between
YAP and TEAD1.
In some embodiments, the compounds disclosed herein bind to TEAD1 at C53, and
disrupt or
inhibit the interaction between YAP and ItAD1. In some embodiments, the
compounds disclosed
herein bind to TEAD1 at C327, and disrupt or inhibit the interaction between
YAP and TEAD1.
In some embodiments, the compounds disclosed herein bind to TEAD1 at C405, and
disrupt or
inhibit the interaction between YAP and TEAD1. In some embodiments, the
compounds disclosed
64
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
herein bind to TEAD1 at C359 and C327, and disrupt or inhibit the interaction
between YAP and
TEAD1. In some embodiments, the compounds disclosed herein bind to TEAD1 at
C359 and C53,
and disrupt or inhibit the interaction between YAP and TEAD1. In some
embodiments, the
compounds disclosed herein bind to TEAD1 at C53 and C327, and disrupt or
inhibit the interaction
between YAP and TEAD1. In some embodiments, the compounds disclosed herein
bind to TEAD1
at C359 and C405, and disrupt or inhibit the interaction between YAP and
TEAD1. In some
embodiments, the compounds disclosed herein bind to TEAD1 at C53 and C405, and
disrupt or
inhibit the interaction between YAP and TEAD1. In some embodiments, the
compounds disclosed
herein bind to TEAD1 at C327 and C405, and disrupt or inhibit the interaction
between YAP and
TEAD1. In some embodiments, the compounds disclosed herein bind to TEAD1 at
C359, C327,
and C53, and disrupt or inhibit the interaction between YAP and TEAD1. In some
embodiments,
the compounds disclosed herein bind to TEAD1 at C359, C327, and C405, and
disrupt or inhibit
the interaction between YAP and TEAD 1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 at C359, C353, and C405, and disrupt or inhibit the interaction
between YAP and
TEAD1. In some embodiments, the compounds disclosed herein bind to TEAD1 at
C327, C53,
and C405, and disrupt or inhibit the interaction between YAP and TEAD1. In
some embodiments,
the compounds disclosed herein bind to TEAD1 at C359, C327, C53, and C405, and
disrupt or
inhibit the interaction between YAP and TEAD1.
1001761 In some embodiments, the compounds disclosed herein bind to TEAD,
prevent TEAD
palmitoylation, and disrupt or inhibit the interaction between YAP and TEAD.
In some
embodiments, the compounds disclosed herein bind to TEAD1 and prevent TEAD1
palmitoylation. In some embodiments, the compounds disclosed herein bind to
TEAD1 and
prevent TEAD1 palmitoylation at C359. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C53. In some embodiments,
the compounds
disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at C327. In
some
embodiments, the compounds disclosed herein bind to TEAD1 and prevent TEAD1
palmitoylation
at C405. In some embodiments, the compounds disclosed herein bind to TEAD1 and
prevent
TEAD1 palmitoylation at C359 and C327. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359 and C53. In some
embodiments, the
compounds disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at
C53 and C327.
In some embodiments, the compounds disclosed herein bind to TEAD1 and prevent
TEAD1
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
palmitoylation at C359 and C405. In some embodiments, the compounds disclosed
herein bind to
TEAD1 and prevent TEAD1 palmitoylation at C53 and C405. In some embodiments,
the
compounds disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at
C327 and
C405. In some embodiments, the compounds disclosed herein bind to TEAD1 and
prevent TEAD1
palmitoylation at C359, C327, and C53. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359, C327, and C405. In
some
embodiments, the compounds disclosed herein bind to TEAD1 and prevent TEAD1
palmitoylation
at C359, C353, and C405. In some embodiments, the compounds disclosed herein
bind to TEAD1
and prevent TEAD1 palmitoylation at C327, C53, and C405. In some embodiments,
the
compounds disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at
C359, C327,
C53, and C405.
1001771 In some embodiments, the compounds disclosed herein bind to TEAD1,
prevent
TEAD1 palmitoylation, and disrupt or inhibit the interaction between YAP and
TEAD1. In some
embodiments, the compounds disclosed herein bind to TEAD1 and prevent TEAD1
palmitoylation
at C359, and disrupt or inhibit the interaction between YAP and TEAD1. In some
embodiments,
the compounds disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation
at C53, and
disrupt or inhibit the interaction between YAP and TEAD1. In some embodiments,
the compounds
disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at C327, and
disrupt or inhibit
the interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C405, and disrupt or inhibit
the interaction
between YAP and TEAD1. In some embodiments, the compounds disclosed herein
bind to
TEAD1 and prevent TEAD1 palmitoylation at C359 and C327, and disrupt or
inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359 and C53, and disrupt or
inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C53 and C327, and disrupt or
inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359 and C405, and disrupt
or inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C53 and C405, and disrupt or
inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
66
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
bind to TEAD1 and prevent TEAD1 palmitoylation at C327 and C405, and disrupt
or inhibit the
interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359, C327, and C53, and
disrupt or inhibit
the interaction between YAP and TEAD1. In some embodiments, the compounds
disclosed herein
bind to TEAD1 and prevent TEAD1 palmitoylation at C359, C327, and C405, and
disrupt or
inhibit the interaction between YAP and l'EAD1. In some embodiments, the
compounds disclosed
herein bind to TEAD1 and prevent TEAD1 palmitoylation at C359, C353, and C405,
and disrupt
or inhibit the interaction between YAP and TEAD1. In some embodiments, the
compounds
disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at C327, C53,
and C405, and
disrupt or inhibit the interaction between YAP and TEAD1. In some embodiments,
the compounds
disclosed herein bind to TEAD1 and prevent TEAD1 palmitoylation at C359, C327,
C53, and
C405, and disrupt or inhibit the interaction between YAP and TEAD1.
[00178] In some embodiments, the compounds disclosed herein bind to TEAD2 at
C380, and
disrupt or inhibit the interaction between YAP and TEAD2.
1001791 In some embodiments, the compounds disclosed herein bind to TEAD2 and
prevent
TEAD2 palmitoylation. In some embodiments, the compounds disclosed herein bind
to TEAD2
and prevent TEAD2 palmitoylation at C380.
[00180] In some embodiments, the compounds disclosed herein bind to TEAD2,
prevent
TEAD2 palmitoylation, and disrupt or inhibit the interaction between YAP and
TEAD2. In some
embodiments, the compounds disclosed herein bind to TEAD2 and prevent TEAD2
palmitoylation
at C380, and disrupt or inhibit the interaction between YAP and TEAD2.
[00181] In some embodiments, the compounds disclosed herein bind to TEAD3 at
C371, and
disrupt or inhibit the interaction between YAP and TEAD3. In some embodiments,
the compounds
disclosed herein bind to TEAD3 at C368, and disrupt or inhibit the interaction
between YAP and
TEAD3. In some embodiments, the compounds disclosed herein bind to TEAD3 at
C371 and
C368, and disrupt or inhibit the interaction between YAP and TEAD3.
[00182] In some embodiments, the compounds disclosed herein bind to TEAD3 and
prevent
TEAD3 palmitoylation. In some embodiments, the compounds disclosed herein bind
to TEAD3
and prevent TEAD3 palmitoylation at C371. In some embodiments, the compounds
disclosed
herein bind to TEAD3 and prevent TEAD3 palmitoylation at C368. In some
embodiments, the
67
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
compounds disclosed herein bind to TEAD3 and prevent TEAD3 palmitoylation at
C368 and
C371.
[00183] In some embodiments, the compounds disclosed herein bind to TEAD3,
prevent
TEAD3 palmitoylation, and disrupt or inhibit the interaction between YAP and
TEAD3. In some
embodiments, the compounds disclosed herein bind to TEAD3 and prevent TEAD3
palmitoylation
at C371, and disrupt or inhibit the interaction between YAP and TEAD3. In some
embodiments,
the compounds disclosed herein bind to TEAD3 and prevent TEAD3 palmitoylation
at C368, and
disrupt or inhibit the interaction between YAP and TEAD3. In some embodiments,
the compounds
disclosed herein bind to TEAD3 and prevent TEAD3 palmitoylation at C371 and
C368, and disrupt
or inhibit the interaction between YAP and TEAD3.
[00184] In some embodiments, the compounds disclosed herein bind to TEAD4 at
C367, and
disrupt or inhibit the interaction between YAP and TEAD4.
[00185] In some embodiments, the compounds disclosed herein bind to TEAD4 and
prevent
TEAD4 palmitoylation. In some embodiments, the compounds disclosed herein bind
to
TEAD4and prevent TEAD4 palmitoylation at C367.
1001861 In some embodiments, the compounds disclosed herein bind to TEAD4,
prevent
TEAD4 palmitoylation, and disrupt or inhibit the interaction between YAP and
TEAD4. In some
embodiments, the compounds disclosed herein bind to TEAD4 and prevent TEAD4
palmitoylation
at C367, and disrupt or inhibit the interaction between YAP and TEAD4.
YAP/TAZ regulation mediated by G-proteins/GPCRs
[00187] In some embodiments, the Hippo pathway is regulated by the G protein-
coupled
receptor (GPCR) and G protein (also known as guanine nucleotide-binding
proteins) family of
proteins. G proteins are molecular switch es that transmit extracel I ul ar
stimuli into the cell through
GPCRs. In some instances, there are two classes of G proteins: monomeric small
GTPases and
heterotrimeric G protein complexes. In some instances, the latter class of
complexes comprise of
alpha (Go), beta (GO, and gamma (GO subunits. In some cases, there are several
classes of Go
subunits: Gqina, Gi2/13a, Geoa (G inhibitory, G other), and Gsa (G
stimulatory).
[00188] In some instances, Gia (G inhibitory), Goa (G other), GO la, and
G12/13a coupled
GPCRs activate YAP/TAZ and promote nuclear translocation. In other instances,
Gsa (G
stimulatory) coupled GPCRs suppress YAP/TAZ activity, leading to YAP/TAZ
degradation.
68
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00189] In some cases, Gia (G inhibitory), Goa (G other), Gqilla, and G12/13a
coupled GPCRs
activate YAP/TAZ through repression of Lats1/2 activities. In contrast, Gsa,
in some
embodiments, induces Lats1/2 activity, thereby promoting YAP/TAZ degradation.
Gq Family
[00190] Gqa (also known as Gq/11 protein), participates in the
inositol trisphosphate (IP3) signal
transduction pathway and calcium (Ca') release from intracellular storage
through the activation
of phospholipase C (PLC). The activated PLC hydrolyzes phosphatidylinositol
4,5-bisphosphate
(PIP2) to diacyl glycerol (DAG) and IP3. In some instances, IP3 then diffuses
through the cytoplasm
into the ER or the sarcoplasmic reticulum (SR) in the case of muscle cells,
and then binds to
inositol trisphosphate receptor (InsP3R), which is a Ca2 channel. In some
cases, the binding
triggers the opening of the Ca' channel, and thereby increases the release of
Ca' into the
cytoplasm.
[00191] In some embodiments, the GPCRs that interact with Gqa include, but are
not limited
to, 5-hydroxytryptamine receptor (5-HT receptor) types 5-HT2 and 5-HT3; alpha-
1 adrenergic
receptor; vasopressin type 1 receptors 1A and 1B; angiotensin II receptor type
1; calcitonin
receptor; histamine H1 receptor; metabotropic glutamate receptor, group I;
muscarinic receptors
Ml, M3, and M5; and trace amine-associated receptor 1.
[00192] In some instances, there are several types of Gqa: Gq, Gqiii,
Gq/14, and Gq/15. The Gq
protein is encoded by GNAQ. Gq/ii is encoded by GNA11. Gq/14 is encoded by
GNA14. Gq/15 is
encoded by GNA15.
[00193] In some instances, mutations or modifications of the Gqa genes have
been associated
with cancer. Indeed, studies have shown that mutations in Gqa promote uveal
melanoma (UM)
tumorigenesis. In some instances, about 80% of UM cases have been detected to
contain a mutation
in GNAQ and/or GNAll.
[00194] In some instances, mutations or modifications of the Gqa genes have
been associated
with congenital diseases. In some instances, mutations of Gqu have been
observed in congenital
diseases such as Port-Wine Stain and/or Sturge-Weber Syndrome. In some
instances, about 92%
of Port-Wine stain cases harbors a mutation in GNAQ. In some instances, about
88% of Sturge-
Weber Syndrome harbors a mutation in GNAQ.
G12/13 Family
69
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00195] G12/13a modulates actin cytoskeletal remodeling in cells and
regulates cell processes
through guanine nucleotide exchange factors (GEFs). GEFs participate in the
activation of small
GTPases which acts as molecular switches in a variety of intracellular
signaling pathways.
Examples of small GTPases include the Ras-related GTPase superfamily (e.g.,
Rho family such as
Cdc42), which is involved in cell differentiation, proliferation, cytoskeletal
organization, vesicle
trafficking, and nuclear transport.
[00196] In some embodiments, the GPCRs that interact with G12/13a include, but
are not limited
to, purinergic receptors (e.g., P2Y1, P2Y2, P2Y4, P2Y6); muscarinic
acetylcholine receptors M1
and M3; receptors for thrombin [protease-activated receptor (PAR)-1, PAR-2];
thromboxane
(TXA2); sphingosine 1-phosphate (e.g., S1P2, S1P3, S1P4 and S1P5);
lysophosphatidic acid (e.g.,
LPAi, LPA2, LPA3); angiotensin II (AT1); serotonin (5-HT2c and 5-HT4);
somatostatin (sst5);
endothelin (ETA and ETB), cholecystokinin (CCK1), Via vasopressin receptors,
D5 dopamine
receptors; fMLP formyl peptide receptors; GAL2 galanin receptors; EP3
prostanoid receptors; Ai
adenosine receptors; Gil adrenergic receptors; BB2 bombesin receptors; B2
bradykinin receptors;
calcium-sensing receptors; KSHV-0RF74 chemokine receptors; NI(' tachykinin
receptors; and
thyroid-stimulating hormone (TSH) receptors.
[00197] In some instances, Gi2/i3ci is further subdivided into G-12
and G13 types which are
encoded by GNA12 and GNA13, respectively.
GiA, Family
[00198] Goact (G inhibitory, G other) (also known as Gi/G0 or Gi protein)
suppresses the
production of 3', 5'-cyclic AMP (cAMP) from adenosine triphosphate (ATP)
through an inhibition
of adenylate cyclase activity, which converts ATP to cAMP.
[00199] In some embodiments, the GPCRs that interact with G. include, but are
not limited to,
5-hydroxytryptamine receptor (5-HT receptor) types 541T1 and 5-HT5; muscarinic
acetylcholine
receptors such as M2 and M4, adenosine receptors such as Ai and A3, adrenergic
receptors such as
ct2A, ct2B, and a2c, apelin receptors, calcium-sensing receptor, cannabinoid
receptors CB1 and CB2,
chemokine CXCR4 receptor, dopamines D2, D3, and D4, GABAB receptor, glutamate
receptors
such as metabotropic glutamate receptor 2 (mGluR2), metabotropic glutamate
receptor 3
(mGluR3), metabotropic glutamate receptor 4 (mGluR4), metabotropic glutamate
receptor 6
(mGluR6), metabotropic glutamate receptor 7 (mGluR7), and metabotropic
glutamate receptor 8
(mGluR8); histamine receptors such as H3 and H4 receptors; melatonin receptors
such as melatonin
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
receptor type 1 (MT1), melatonin receptor type 2 (MT2), and melatonin receptor
type 3 (MT3);
niacin receptors such as NIACR1 and NIACR2; opioid receptors such as 6, lc,
la, and nociceptin
receptors; prostaglandin receptors such as prostaglandin E receptor 1 (E131),
prostaglandin E
receptor 3 (EP3), prostaglandin F receptor (FP), and thromboxane receptor
(TP); somatostatin
receptors ssti, 55t2, s5t3, s5t4, and sst5; and trace amine-associated
receptor 8.
[00200] In some instances, there are several types of Gia: Ga1, Gia2, Gia3,
Gia4, Goa, Gt, GgEst,
and Gz. Gial is encoded by GNAT'. Gia2 is encoded by GNAI2. Gia3 is encoded by
GNAI3. Goa,
the ao subunit, is encoded by GNA01. Gt is encoded by GNAT1 and GNAT2. Gpist
is encoded by
GNAT3. Gz is encoded by GNAZ.
Gs Family
[00201] Gsa (also known as G stimulatory, Gs alpha subunit, or Gs protein)
activates the cAMP-
dependent pathway through the activation of adenylate cyclase, which convers
adenosine
triphosphate (ATP) to 3',5'-cyclic AMP (cAMP) and pyrophosphate. In some
embodiments, the
GPCRs that interact with Gsa include, but are not limited to, 5-
hydroxytryptamine receptor (5-HT
receptor) types 5-HT4, 5-HT6, and 5-HT7; adrenocorticotropic hormone receptor
(ACTH receptor)
(also known as melanocortin receptor 2 or MC2R); adenosine receptor types Aza
and A2b; arginine
vasopressin receptor 2 (AVPR2); 13-adrenergic receptors f31, 132, and (33;
calcitonin receptor;
calcitonin gene-related peptide receptor; corticotropin-releasing hormone
receptor; dopamine
receptor Dl-like family receptors such as Di and D5; follicle-stimulating
hormone receptor (FSH-
receptor); gastric inhibitory polypeptide receptor; glucagon receptor;
histamine H2 receptor;
luteinizing hormone/choriogonadotropin receptor; melanocortin receptors such
as MC1R, MC2R,
MC3R, MC4R, and MC5R; parathyroid hormone receptor 1; prostaglandin receptor
types D2 and
12; secretin receptor; thyrotropin receptor; trace amine-associated receptor
1; and box jellyfish
op si n .
[00202] In some instances, there are two types of Gsa: Gs and Goff. Gs is
encoded by GNAS.
Golf is encoded by GNAL.
Additional Regulators of the Hippo signaling network
[00203] In some embodiments, the additional regulator of the Hippo signaling
pathway is the
Crumbs (Crb) complex. The Crumbs complex is a key regulator of cell polarity
and cell shape. In
some instances, the Crumbs complex comprises transmembrane CRB proteins which
assemble
multi-protein complexes that function in cell polarity. In some instances, CRB
complexes recruit
71
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
members of the Angiomotin (AMOT) family of adaptor proteins that interact with
the Hippo
pathway components. In some instances, studies have shown that AMOT directly
binds to YAP,
promotes YAP phosphorylation, and inhibits its nuclear localization.
[00204] In some instances, the additional regulator of the Hippo signaling
pathway comprises
regulators of the MST kinase family. MST kinases monitor actin cytoskeletal
integrity. In some
instances, the regulators include TAO kinases and cell polarity kinase PAR-1.
[00205] In some instances, the additional regulator of the Hippo signaling
pathway comprises
molecules of the adherens junction. In some instances, E-Cadherin (E-cad)
suppresses YAP
nuclear localization and activity through regulating MST activity. In some
embodiments, E-cad-
associated protein a-catenin regulates YAP through sequestering YAP/14-3-3
complexes in the
cytoplasm. In other instances, Ajuba protein family members interact with
Lats1/2 kinase activity,
thereby preventing inactivation of YAP/TAZ.
[00206] In some embodiments, additional proteins that interact with YAP/TAZ
either directly
or indirectly include, but are not limited to, Merlin, protocadherin Fat 1,
MASK1/2, HIPK2,
PTPN14, RAS SF, PP2A, Salt-inducible kinases (SIKs), Scribble (SCRIB), the
Scribble associated
proteins Discs large (Dig), KIBRA, PTPN14, NPHP3, LKB1, Ajuba, and Z01/2.
[00207] In some embodiments, the compounds described herein are inhibitors of
transcriptional
coactivator with PDZ binding motif/Yes-associated protein transcriptional
coactivator
(TAZ/YAP). In some embodiments, the compounds described herein increase the
phosphorylation
of transcriptional coactivator with PDZ binding m otif/Yes-associ ate d
protein transcriptional
coactivator (TAZ/YAP) or decrease the dephosphorylation of transcriptional
coactivator with PDZ
binding motif/Yes-associated protein transcriptional coactivator (TAZ/YAP). In
some
embodiments, the compounds increase the ubiquitination of transcriptional
coactivator with PDZ
binding motif/Yes-associated protein transcriptional coactivator (TAZ/YAP) or
decrease the
deubiquitination of transcriptional coactivator with PDZ binding motif/Yes-
associated protein
transcriptional coactivator (TAZ/YAP).
[00208] In some embodiments, the compounds disclosed herein are inhibitors of
one or more
of the proteins encompassed by, or related to, the Hippo pathway. In some
embodiments, an
inhibitor of the Hippo pathway is an inhibitor of a G-protein and/or its
coupled GPCR. In some
embodiments, an inhibitor of the Hippo pathway is an inhibitor of a G-protein.
In some
embodiments, an inhibitor of the Hippo pathway is an inhibitor of the Gqa
family proteins such as
72
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Gq, Gq/11, Gq/14, and Gq/15; the G12/13a family of proteins such as G12 and
G13; or the Gia family of
proteins such as Gial, Gia2, Gia3, Gia4, Goa, Gt, Ggust, and G. In some
embodiments, an inhibitor
of the Hippo pathway is an inhibitor of Gq. In some embodiments, an inhibitor
of the Hippo
pathway is an inhibitor of Gqiii. In some embodiments, an inhibitor of the
Hippo pathway is an
inhibitor of Gq/14. In some embodiments, an inhibitor of the Hippo pathway is
an inhibitor of Gq/15.
In some embodiments, an inhibitor of the Hippo pathway is an inhibitor of G12.
In some
embodiments, an inhibitor of the Hippo pathway is an inhibitor of G13. In some
embodiments, an
inhibitor of the Hippo pathway is an inhibitor of Gial . In some embodiments,
an inhibitor of the
Hippo pathway is an inhibitor of G1a2. In some embodiments, an inhibitor of
the Hippo pathway
is an inhibitor of Gia3. In some embodiments, an inhibitor of the Hippo
pathway is an inhibitor of
Gia4. In some embodiments, an inhibitor of the Hippo pathway is an inhibitor
of Goa. In some
embodiments, an inhibitor of the Hippo pathway is an inhibitor of G. In some
embodiments, an
inhibitor of the Hippo pathway is an inhibitor of Ggust. In some embodiments,
an inhibitor of the
Hippo pathway is an inhibitor of G.
1002091 In some embodiments, an inhibitor of the Hippo pathway is an inhibitor
of a core
protein of the Hippo pathway. In some embodiments, an inhibitor of the Hippo
pathway is an
inhibitor of Say I . In some embodiments, an inhibitor of the Hippo pathway is
an inhibitor of Mob.
In some embodiments, an inhibitor of the Hippo pathway is an inhibitor of YAP.
In some
embodiments, an inhibitor of the Hippo pathway is an inhibitor of TAZ. In some
embodiments, an
inhibitor of the Hippo pathway is an inhibitor of TEAD.
[00210] In some embodiments, an inhibitor of the Hippo pathway is an inhibitor
of a protein
associated with the ubiquitination and proteasomal degradation pathway. In
some embodiments,
an inhibitor of the Hippo pathway is an inhibitor of a proteasomal degradation
pathway protein
(e.g., 26S proteasome).
[00211] In some embodiments, an inhibitor of the Hippo pathway is an inhibitor
of a protein of
the Ras superfamily of proteins. In some embodiments, an inhibitor of the
Hippo pathway is an
inhibitor of a protein of the Rho family of proteins. In some embodiments, an
inhibitor of the
Hippo pathway is an inhibitor of Cdc42.
[00212] Cdc42 is a member of the Ras superfamily of small GTPases.
Specifically, Cdc42
belongs to the Rho family of GTPases, in which the family members participate
in diverse and
critical cellular processes such as gene transcription, cell-cell adhesion,
and cell cycle progression.
73
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Cdc42 is involved in cell growth and polarity, and in some instances, Cdc42 is
activated by guanine
nucleotide exchange factors (GEFs). In some cases, an inhibitor of Cdc42 is a
compound disclosed
herein.
1002131 In some embodiments, an inhibitor of the Hippo pathway is an inhibitor
of a
deubiquitinating enzyme. In some embodiments, an inhibitor of the Hippo
pathway is an inhibitor
of a cysteine protease or a metalloprotease. In some embodiments, an inhibitor
of the Hippo
pathway is an inhibitor of an ubiquitin-specific protease. USP47 is a member
of the
ubiquitin-specific protease (USP/UBP) superfamily of cysteine proteases. In
some embodiments,
the compounds disclosed herein are inhibitors of USP47.
1002141 In some embodiments, the present invention provides a use of a
compound, or a
pharmaceutical salt or composition thereof, for treating one or more
disorders, diseases, and/or
conditions wherein the disorder, disease, or condition includes, but is not
limited to, a cellular
proliferative disorder.
1002151 The activity of a compound utilized in this invention as an inhibitor
of TEAD (e.g.,
TEAD1, TEAD2, TEAD3, and/or TEAD4), or a variant or mutant thereof, can be
assayed in vitro,
in vivo or in a cell line. In vitro assays include assays that determine
inhibition of TEAD (e.g.,
TEAD1, TEAD2, TEAD3, and/or TEAD4), or a variant or mutant thereof. Alternate
in vitro
assays quantitate the ability of the inhibitor to bind to TEAD (e.g., TEAD1,
TEAD2, TEAD3,
and/or TEAD4) or a variant or mutant thereof. Detailed conditions for assaying
a compound
utilized in this invention as an inhibitor of TEAD (e.g., TEAD1, TEAD2, TEAD3,
and/or TEAD4),
or a variant or mutant thereof, are set forth in the Examples below. See, for
example, Examples 2
and 5.
1002161 As used herein, the terms "treatment," "treat," and
"treating" refer to reversing,
alleviating, delaying the onset of, or inhibiting the progress of a disease or
disorder, or one or more
symptoms thereof, as described herein. In some embodiments, treatment can be
administered after
one or more symptoms have developed. In other embodiments, treatment can be
administered in
the absence of symptoms. For example, treatment can be administered to a
susceptible individual
prior to the onset of symptoms (e.g., in light of a history of symptoms and/or
in light of genetic or
other susceptibility factors). Treatment can also be continued after symptoms
have resolved, for
example, to prevent or delay their recurrence.
74
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00217] The provided compounds are inhibitors of TEAD (e.g., TEAD1, TEAD2,
TEAD3,
and/or TEAD4) and are therefore useful for treating one or more disorders
associated with activity
of TEAD (e.g., TEAD1, TEAD2, TEAD3, and/or TEAD4). Thus, in certain aspects
and
embodiments, the present invention provides a method for treating a TEAD-
mediated disorder
comprising the step of administering to a patient in need thereof a
therapeutically effective
compound of the present invention, or pharmaceutically acceptable composition
thereof.
[00218] As used herein, the term "TEAD-mediated" disorders, diseases, and/or
conditions as
used herein means any disease or other deleterious condition in which TEAD
(e.g., TEAD1,
TEAD2, TEAD3, and/or TEAD4), or a variant or mutant thereof, is known to play
a role
Accordingly, another aspect or embodiment of the present invention relates to
treating or lessening
the severity of one or more diseases in which TEAD (e.g., TEAD1, TEAD2, TEAD3,
and/or
TEAD4), or a variant or mutant thereof, are known to play a role.
[00219] As used herein, the term "a therapeutically effective amount of'
refers to the amount
of a TEAD inhibitor or a pharmaceutically acceptable salt thereof, which is
effective to reduce or
attenuate the biological activity of TEAD (e.g., TEAD1, TEAD2, TEAD3, and/or
TEAD4) or a
variant or mutant thereof, provide a therapeutic benefit in the treatment of a
condition, or to delay
or minimize one or more symptoms associated with the condition in a biological
sample or in a
patient. In some embodiments, "a therapeutically effective amount of' refers
to the amount of a
TEAD inhibitor or a pharmaceutically acceptable salt thereof that measurably
decreases the
binding or signaling activity of TEAD (e.g., TEAD1, TEAD2, TEAD3, and/or
TEAD4), or a
variant or mutant thereof or any TEAD-mediated activity. The term
"therapeutically effective
amount- can encompass, in some embodiments, an amount that improves overall
therapy, reduces
or avoids symptoms, signs, or causes of the condition, and/or enhances the
therapeutic efficacy of
another therapeutic agent In certain embodiments, a therapeutically effective
amount is an amount
sufficient for inhibition of a TEAD transcription factor. In certain
embodiments, a therapeutically
effective amount is an amount sufficient for treating a proliferative disease.
[00220] In some aspects and embodiments, provided herein are methods of
treating, reducing
the severity of, delaying the onset of, or inhibiting the progress of a
disease or disorder, or one or
more symptoms thereof of a disease or disorder characterized by or associated
with increased
TEAD (e.g., TEAD1, TEAD2, TEAD3, and/or TEAD4) expression and/or increased
TEAD (e.g.,
TEAD1, TEAD2, TEAD3, and/or TEAD4) activity comprising the step of
administering to a
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
patient in need thereof a therapeutically effective compound of the present
invention, or
pharmaceutically acceptable composition thereof. In some aspects and
embodiments, provided
herein are methods of treating, reducing the severity of, delaying the onset
of, or inhibiting the
progress of a disease or disorder, or one or more symptoms thereof of a
disease or disorder in
which inhibition or antagonizing of TEAD (e.g., TEAD, TEAD2, TEAD3, and/or
TEAD4)
activity is beneficial comprising the step of administering to a patient in
need thereof a
therapeutically effective compound of the present invention, or
pharmaceutically acceptable
composition thereof. In some aspects and embodiments, provided herein are
methods of treating,
reducing the severity of, delaying the onset of, or inhibiting the progress of
a disease or disorder,
or one or more symptms thereof of a disease or disorder in which inhibition or
antagonizing of the
Hippo pathway is beneficial comprising the step of administering to a patient
in need thereof a
therapeutically effective compound of the present invention, or
pharmaceutically acceptable
composition thereof
1002211 In some aspects and embodiments, the present invention provides a
method for treating
one or more disorders, diseases, and/or conditions wherein the disorder,
disease, or condition
includes, but is not limited to, a cellular proliferative disorder, comprising
administering to a
patient in need thereof, a TEAD inhibitor compound as described herein, or a
pharmaceutical salt
or composition thereof. In some embodiments, a cellular proliferative disorder
is cancer. In some
embodiments, the cancer is characterized by increased TEAD (e.g., TEAD1,
TEAD2, TEAD3,
and/or TEAD4) expression and/or increased TEAD (e.g., TEAD1, TEAD2, TEAD3,
and/or
TEAD4) activity.
1002221 As used herein, the terms "increased," -elevated,- or -enhanced,- are
used
interchangeably and encompass any measurable increase in a biological function
and/or biological
activity and/or a concentration. For example, an increase can be by at least
about 10%, about 15%,
about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%,
about 55%,
about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%,
about 95%,
about 96%, about 97%, about 98%, about 99%, about 100%, about 2-fold, about 3-
fold, about 4-
fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold,
about 10-fold, about 20-
fold, about 25-fold, about 50-fold, about 100-fold, or higher, relative to a
control or baseline
amount of a function, or activity, or concentration.
76
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002231 As used herein, the terms "increased expression" and/or "increased
activity" of a
substance, such as TEAD, in a sample or cancer or patient refers to an
increase in the amount of
the substance, such as TEAD, of about 5%, about 10%, about 15%, about 20%,
about 25%, about
30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about
65%, about
70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about
97%, about
98%, about 99%, about 100%, about 2-fold, about 3-fold, about 4-fold, about 5-
fold, about 6-fold,
about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20-fold, about
25-fold, about 50-
fold, about 100-fold, or higher, relative to the amount of the substance, such
as TEAD, in a control
sample or control samples, such as an individual or group of individuals who
are not suffering
from the disease or disorder (e.g., cancer) or an internal control, as
determined by techniques
known in the art. A subject can also be determined to have an "increased
expression" or "increased
activity" of TEAD if the expression and/or activity of TEAD is increased by
one standard
deviation, two standard deviations, three standard deviations, four standard
deviations, five
standard deviations, or more, relative to the mean (average) or median amount
of TEAD in a
control group of samples or a baseline group of samples or a retrospective
analysis of patient
samples. As practiced in the art, such control or baseline expression levels
can be previously
determined, or measured prior to the measurement in the sample or cancer or
subject, or can be
obtained from a database of such control samples.
1002241 As used herein, a "proliferative disease" refers to a disease that
occurs due to abnormal
growth or extension by the multiplication of cells (Walker, Cambridge
Dictionary of Biology,
Cambridge University Press: Cambridge, UK, 1990). A proliferative disease can
be associated
with: 1) the pathological proliferation of normally quiescent cells; 2) the
pathological migration of
cells from their normal location (e.g., metastasis of neoplastic cells); 3)
the pathological expression
of proteolytic enzymes, such as the matrix metalloproteinases (e.g.,
collagenases, gel atinases, and
elastases); or 4) the pathological angiogenesis as in proliferative
retinopathy and tumor metastasis.
Exemplary proliferative diseases include cancers (i.e., -malignant
neoplasms"), benign neoplasms,
angiogenesis, inflammatory diseases, and autoimmune diseases.
Cancer
1002251 The cancer or proliferative disorder or tumor to be treated using the
compounds and
methods and uses described herein include, but are not limited to, a
hematological cancer, a
77
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
lymphoma, a myeloma, a leukemia, a neurological cancer, skin cancer, breast
cancer, a
prostate cancer, a colorectal cancer, lung cancer, head and neck cancer, a
gastrointestinal cancer,
a liver cancer, a pancreatic cancer, a genitourinary cancer, a bone cancer,
renal cancer, and a
vascular cancer.
1002261 In some embodiments of the methods and uses described herein, a cancer
is mediated
by activation of transcriptional coactivator with PDZ binding motif/Yes-
associated protein
transcription coactivator (TAZ/YAP). In some embodiments of the methods and
uses described
herein, a cancer is mediated by modulation of the interaction of YAP/TAZ with
TEAD (e.g.,
TEAD1, TEAD2, TEAD3, and/or TEAD4). In some embodiments of the methods and
uses
described herein, the cancer is characterized by or associated with increased
TEAD (e.g., TEAD1,
TEAD2, TEAD3, and/or TEAD4) expression and/or increased TEAD (e.g., TEAD1,
TEAD2,
TEAD3, and/or TEAD4) activity. In some embodiments of the methods and uses
described herein,
the cancer is a cancer in which YAP is localized in the nucleus of the cancer
cells.
1002271 In some embodiments of the methods and uses described herein, the
cancer is
characterized or associated with a genetic alteration in one or more Hippo
pathway genes. As used
herein, the term "genetic alteration in one or more Hippo pathway genes"
refers to that certain
percentage of cells in a sample, such as a tumor sample, having a detectable
amount of genetic
alteration in one or more Hippo pathway genes. As used herein, a genetic
alteration in a gene, such
as a Hippo pathway gene, can refer, for example, to a loss-of-function
mutation in the gene
(including, for example, frameshifts, nonsense mutations and splicing
mutations), a change in gene
copy number (including, for example, copy gain, amplification, copy loss, or
deletion), or a fusion
of the gene with another gene, such as, for example, a TAZ-CAMTA1 fusion or
YAP1-TFE3
fusion. In some embodiments, genetic alteration in Hippo pathway genes refers
to that about 5%,
about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%,
about 45%,
about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%,
about 85%,
about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100% of
cells, such as
tumor cells, in a sample have at least about three copies of genetically
altered Hippo pathway
genes, at least about four copies of genetically altered Hippo pathway genes,
at least about five
copies of genetically altered Hippo pathway genes, at least about six copies
of genetically altered
Hippo pathway genes, at least about seven copies of genetically altered Hippo
pathway genes, at
least about eight copies of genetically altered Hippo pathway genes, at least
about nine copies of
78
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
genetically altered Hippo pathway genes, at least about ten copies of
genetically altered Hippo
pathway genes, at least about eleven copies of genetically altered Hippo
pathway genes, at least
about twelve copies of genetically altered Hippo pathway genes, at least about
nine copies of
genetically altered Hippo pathway genes, at least about ten copies of
genetically altered Hippo
pathway genes, at least about eleven copies of genetically altered Hippo
pathway genes, at least
about twelve copies of genetically altered Hippo pathway genes, at least about
thirteen copies of
genetically altered Hippo pathway genes, at least about fourteen copies of
genetically altered
Hippo pathway genes, at least about fifteen copies of genetically altered
Hippo pathway genes, at
least about twenty copies of genetically altered Hippo pathway genes, or more.
In some
embodiments, genetic alteration in Hippo pathway genes refers to that about
10% tumor cells in a
sample have at least about 15 copies of genetically altered Hippo pathway
genes. In some
embodiments, genetic alteration in Hippo pathway genes refers to that about
40% tumor cells in a
sample have at least about 4 copies of genetically altered Hippo pathway
genes. In some
embodiments, genetic alteration in Hippo pathway genes refers to that about
10% tumor cells in a
sample have at least about four copies of genetically altered Hippo pathway
genes. In some
embodiments, a Hippo pathway gene is NF2. In some embodiments, the genetic
alteration in the
one or more Hippo pathway genes is NF2 deficiency. In some embodiments, NF2
deficiency refers
to NF2 loss of function mutations. In some embodiments, NF2 deficiency refers
to NF2 copy losses
or deletions. In some embodiments, NF2 deficiency refers to absent or very low
NF2 mRNA
expression. In some embodiments, a Hippo pathway gene is YAP1. In some
embodiments, the
genetic alteration in the one or more Hippo pathway genes is YAP1
amplification. In some
embodiments, the genetic alteration in the one or more Hippo pathway genes is
a YAP1 fusion,
such as a YAP1-TFE3 fusion. In some embodiments, a Hippo pathway gene is TAZ.
In some
embodiments, the genetic alteration in the one or more Hippo pathway genes is
TAZ amplification.
In some embodiments, the genetic alteration in the one or more Hippo pathway
genes is a TAZ
fusion, such as a TAZ-CAMTA1 fusion. In some embodiments, a Hippo pathway gene
is LATS
1/2. In some embodiments, the genetic alteration in the one or more Hippo
pathway genes is LATS
1/2 copy number loss or deletion. In some embodiments, a Hippo pathway gene is
MST1/2. In
some embodiments, a Hippo pathway gene is BAP1.
1002281 In some embodiments, a cancer is characterized by a mutant Ga-protein.
In some
embodiments, a mutant Ga-protein is selected from G12, G13, Gq, G11, Gi, Go,
and Gs. In some
79
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
embodiments, a mutant Ga-protein is G12. In some embodiments, a mutant Ga-
protein is G13. In
some embodiments, a mutant Ga-protein is Gq. In some embodiments, a mutant Ga-
protein is Gil
. In some embodiments, a mutant Ga-protein is Gi. In some embodiments, a
mutant Ga-protein is
Go. In some embodiments, a mutant Ga-protein is Gs.
1002291 In some embodiments of the methods and uses described herein, a cancer
is treated by
inhibiting or reducing or decreasing or arresting further growth or spread of
the cancer or tumor.
In some embodiments of the methods and uses described herein, a cancer is
treated by inhibiting
or reducing the size (e.g., volume or mass) of the cancer or tumor by at least
5%, at least 10%, at
least 25%, at least 50%, at least 75%, at least 90%, at least 950/s, at least
96%, at least 97%, at least
98%, or at least 99% relative to the size of the cancer or tumor prior to
treatment. In some
embodiments of the methods and uses described herein, a cancer is treated by
reducing the quantity
of the cancers or tumors in the patient by at least 5%, at least 10%, at least
25%, at least 50%, at
least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least
98%, or at least 99%
relative to the quantity of the cancers or tumors prior to treatment.
1002301 In some embodiments, a patient treated using the methods or uses
described herein
exhibits progression-free survival of at least about one month, at least about
2 months, at least
about 3 months, at least about 4 months, at least about 5 months, at least
about 6 months, at least
about 7 months, at least about 8 months, at least about 9 months, at least
about 10 months, at least
about 11 months, at least about one year, at least about eighteen months, at
least about two years,
at least about three years, at least about four years, or at least about five
years after the treatment
is initiated. In some embodiments, a patient treated using the methods or uses
described herein
exhibits an overall survival of at least about one month, at least about 2
months, at least about 3
months, at least about 4 months, at least about 5 months, at least about 6
months, at least about 7
months, at least about 8 months, at least about 9 months, at least about 10
months, at least about
11 months, at least about one year, at least about 14 months, at least about
16 months, at least about
18 months, at least about 20 months, at least about 22 months, at least about
two years, at least
about three years, at least about four years, or at least about five years
after the treatment is
initiated.
[00231] In some embodiments, a patient treated using the methods or uses
described herein
exhibits an objective response rate (ORR) of at least about 15%, at least
about 20%, at least about
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
25%, at least about 30%, about 35%, about 40%, about 45%, about 50%, about
55%, about 60%,
about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%,
or about 100%.
1002321 In some embodiments of the methods and uses described herein, the
cancer is lung
cancer, thyroid cancer, ovarian cancer, colorectal cancer, prostate cancer,
cancer of the pancreas,
cancer of the esophagus, liver cancer, breast cancer, skin cancer, or
mesothelioma. In some
embodiments, the cancer is lung cancer, thyroid cancer, ovarian cancer,
colorectal cancer, prostate
cancer, cancer of the pancreas, cancer of the esophagus, liver cancer, breast
cancer, skin cancer,
or mesothelioma, sarcoma, or epithelioid hemangioendothelioma (EHE). In some
embodiments,
the cancer is mesothelioma, such as malignant mesothelioma In some
embodiments, the cancer is
EHE
1002331 In some embodiments, cancer includes, without limitation, leukemias
(e.g., acute
leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute
myeloblastic leukemia,
acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic
leukemia, acute
erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic
lymphocytic leukemia),
polycythemia vera, lymphoma (e.g., Hodgkin's disease or non-Hodgkin's
disease), Waldenstrom's
macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors
such as sarcomas
and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma,
osteogenic
sarcoma, chordoma, angiosarcoma, endotheliosarcoma,
lymphangiosarcoma,
lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor,
leiomyosarcoma,
rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian
cancer, prostate
cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat
gland carcinoma,
sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas,
cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell
carcinoma,
hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma,
Wilm's tumor,
cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell
lung carcinoma,
bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, glioblastoma
multifoline (GBM,
also known as glioblastoma), medulloblastoma, craniopharyngioma, ependymoma,
pinealoma,
hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma,
neurofibrosarcoma,
meningioma, melanoma, neuroblastoma, and retinoblastoma).
1002341 In some embodiments, the cancer is glioma, astrocytoma, glioblastoma
multiforme
(GBM, also known as glioblastoma), medulloblastoma, craniopharyngioma,
ependymoma,
81
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
pi neal om a, hem angi oblastom a, acoustic neurom a, ol i godendrogl i om a,
s chwannom a,
neurofibrosarcoma, meningioma, melanoma, neuroblastoma, or retinoblastoma.
[00235] In some embodiments, the cancer is acoustic neuroma, astrocytoma
(e.g., Grade I ¨
Pi 1 ocyti c A strocytom a, Grade II ¨ Low-grade A strocytom a, Grade III ¨ A
n apl asti c A strocytom a,
or Grade IV ¨ Glioblastoma (GBM)), chordoma, CNS lymphoma, craniopharyngioma,
brain stem
glioma, ependymoma, mixed glioma, optic nerve glioma, subependymoma,
medulloblastoma,
meningioma, metastatic brain tumor, oligodendroglioma, pituitary tumors,
primitive
neuroectodermal (PNET) tumor, or schwannoma. In some embodiments, the cancer
is a type
found more commonly in children than adults, such as brain stem glioma,
craniopharyngi om a,
ependymoma, juvenile pilocytic astrocytoma (SPA), medulloblastoma, optic nerve
glioma, pineal
tumor, primitive neuroectodermal tumors (PNET), or rhabdoid tumor. In some
embodiments, the
patient is an adult human. In some embodiments, the patient is a child or
pediatric patient.
[00236] Cancer includes, in another embodiment, without limitation,
mesothelioma,
hepatobilliary (hepatic and billiary duct), bone cancer, pancreatic cancer,
skin cancer, cancer of
the head or neck, cutaneous or intraocular melanoma, ovarian cancer, colon
cancer, rectal cancer,
cancer of the anal region, stomach cancer, gastrointestinal (gastric,
colorectal, and duodenal),
uterine cancer, carcinoma of the fallopian tubes, carcinoma of the
endometrium, carcinoma of the
cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease,
cancer of the
esophagus, cancer of the small intestine, cancer of the endocrine system,
cancer of the thyroid
gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma
of soft tissue, cancer
of the urethra, cancer of the penis, prostate cancer, testicular cancer,
chronic or acute leukemia,
chronic myeloid leukemia, lymphocytic lymphomas, cancer of the bladder, cancer
of the kidney
or ureter, renal cell carcinoma, carcinoma of the renal pelvis, non-Hodgkins's
lymphoma, spinal
axis tumors, brain stem glioma, pituitary adenoma, adrenocorti cal cancer,
gall bladder cancer,
multiple myeloma, cholangiocarcinoma, fibrosarcoma, neuroblastoma,
retinoblastoma, or a
combination of one or more of the foregoing cancers.
[00237] In some embodiments, the cancer is selected from hepatocellular
carcinoma, ovarian
cancer, ovarian epithelial cancer, or fallopian tube cancer, papillary serous
cystadenocarcinoma or
uterine papillary serous carcinoma (UPSC); prostate cancer; testicular cancer;
gallbladder cancer;
hepatocholangiocarcinoma; soft tissue and bone synovial sarcoma;
rhabdomyosarcoma;
osteosarcoma; chondrosarcoma; Ewing sarcoma; anaplastic thyroid cancer;
adrenocortical
82
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
adenoma; pancreatic cancer; pancreatic ductal carcinoma or pancreatic
adenocarcinoma;
gastrointestinal/stomach (GIST) cancer; lymphoma; squamous cell carcinoma of
the head and neck
(SCCHN); salivary gland cancer; glioma, or brain cancer; neurofibromatosis-1
associated
malignant peripheral nerve sheath tumors (MPNST); Wal den strom ' s m acrogl
obul inemi a; or
medulloblastoma.
[00238] In some embodiments, the cancer is selected from
hepatocellular carcinoma (HCC),
hepatoblastoma, colon cancer, rectal cancer, ovarian cancer, ovarian
epithelial cancer, fallopian
tube cancer, papillary serous cystadenocarcinoma, uterine papillary serous
carcinoma (UPSC),
hepatocholangiocarcinoma, soft tissue and bone synovi al sarcoma,
rhabdomyosarcoma,
osteosarcoma, anaplastic thyroid cancer, adrenocortical adenoma, pancreatic
cancer, pancreatic
ductal carcinoma, pancreatic adenocarcinoma, glioma, neurofibromatosis-1
associated malignant
peripheral nerve sheath tumors (MPNST), Waldenstrom's macroglobulinemia, or
medulloblastoma.
[00239] In some embodiments, a cancer is a solid tumor, such as a sarcoma,
carcinoma, or
lymphoma. Solid tumors generally comprise an abnormal mass of tissue that
typically does not
include cysts or liquid areas. In some embodiments, the cancer is selected
from renal cell
carcinoma, or kidney cancer; hepatocellular carcinoma (HCC) or hepatoblastoma,
or liver cancer;
melanoma; breast cancer; colorectal carcinoma, or colorectal cancer; colon
cancer; rectal cancer;
anal cancer; lung cancer, such as non-small cell lung cancer (NSCLC) or small
cell lung cancer
(SCLC); ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, or
fallopian tube cancer;
papillary serous cystadenocarcinoma or uterine papillary serous carcinoma
(UPSC); prostate
cancer; testicular cancer; gallbladder cancer; hepatocholangiocarcinoma; soft
tissue and bone
synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; Ewing
sarcoma;
anaplastic thyroid cancer; adrenocortical carcinoma; pancreatic cancer;
pancreatic ductal
carcinoma or pancreatic adenocarcinoma, gastrointestinal/stomach (GIST)
cancer, lymphoma,
squamous cell carcinoma of the head and neck (SCCHN), salivary gland cancer,
glioma, or brain
cancer, neurofibromatosis-1 associated malignant peripheral nerve sheath
tumors (MPNST),
Waldenstrom's macroglobulinemia, or medulloblastoma.
[00240] In some embodiments, the cancer is selected from renal cell carcinoma,
hepatocellular
carcinoma (HCC), hepatoblastoma, colorectal carcinoma, colorectal cancer,
colon cancer, rectal
cancer, anal cancer, ovarian cancer, ovarian epithelial cancer, ovarian
carcinoma, fallopian tube
83
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
cancer, papillary serous cystadenocarcinoma, uterine papillary serous
carcinoma (UPSC),
hepatocholangiocarcinoma, soft tissue and bone synovial sarcoma,
rhabdomyosarcoma,
osteosarcoma, chondrosarcoma, anaplastic thyroid cancer, adrenocortical
carcinoma, pancreatic
cancer, pancreatic ductal carcinoma, pancreatic adenocarcinoma, glioma, brain
cancer,
neurofibromatosis-1 associated malignant peripheral nerve sheath tumors
(MPNST),
Waldenstrom's macroglobulinemia, or medulloblastoma.
[00241] In some embodiments, the cancer is selected from hepatocellular
carcinoma (HCC),
hepatoblastoma, colon cancer, rectal cancer, ovarian cancer, ovarian
epithelial cancer, ovarian
carcinoma, fallopian tube cancer, papillary serous cystadenocarcinoma, uterine
papillary serous
carcinoma (UPSC), hepatocholangiocarcinoma, soft tissue and bone synovial
sarcoma,
rhabdomyosarcoma, osteosarcoma, anaplastic thyroid cancer, adrenocortical
carcinoma,
pancreatic cancer, pancreatic ductal carcinoma, pancreatic adenocarcinoma,
glioma,
neurofibromatosis-1 associated malignant peripheral nerve sheath tumors
(MPNST),
Waldenstrom's macroglobulinemia, or medulloblastoma.
1002421 In some embodiments, the cancer is hepatocellular carcinoma (HCC). In
some
embodiments, the cancer is hepatoblastoma. In some embodiments, the cancer is
colon cancer. In
some embodiments, the cancer is rectal cancer. In some embodiments, the cancer
is ovarian
cancer, or ovarian carcinoma. In some embodiments, the cancer is ovarian
epithelial cancer. In
some embodiments, the cancer is fallopian tube cancer. In some embodiments,
the cancer is
papillary serous cystadenocarcinoma. In some embodiments, the cancer is
uterine papillary serous
carcinoma (UPSC). In some embodiments, the cancer is hepatocholangiocarcinoma.
In some
embodiments, the cancer is soft tissue and bone synovial sarcoma. In some
embodiments, the
cancer is rhabdomyosarcoma. In some embodiments, the cancer is osteosarcoma.
In some
embodiments, the cancer is anaplastic thyroid cancer. In some embodiments, the
cancer is
adrenocortical carcinoma. In some embodiments, the cancer is pancreatic
cancer, or pancreatic
ductal carcinoma. In some embodiments, the cancer is pancreatic
adenocarcinoma. In some
embodiments, the cancer is glioma. In some embodiments, the cancer is
malignant peripheral
nerve sheath tumors (MPNST). In some embodiments, the cancer is
neurofibromatosis-1
associated IVfPNST. In some embodiments, the cancer is Waldenstrom's
macroglobulinemia. In
some embodiments, the cancer is medulloblastoma.
84
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002431 In some embodiments, a cancer is a viral-associated cancer, including
human
immunodeficiency virus (HIV) associated solid tumors, human papilloma virus
(EIPV)-16 positive
incurable solid tumors, and adult T-cell leukemia, which is caused by human T-
cell leukemia virus
type I (HTLV-I) and is a highly aggressive form of CD4+ T-cell leukemia
characterized by clonal
integration of HTLV-I in leukemic cells (See
https://clinicaltrials.gov/ct2/show/study/
NCT02631746); as well as virus-associated tumors in gastric cancer,
nasopharyngeal carcinoma,
cervical cancer, vaginal cancer, vulvar cancer, squamous cell carcinoma of the
head and neck, and
Merkel cell carcinoma. (See
https://clinicaltrials.govict2/show/study/NCT02488759; see also
http s : //cli ni cal trials .govict2/show/study/NCT0240886;
http s : //cli n i cal tri al s. gov/ct2/show/
NCT02426892)
1002441 In some embodiments, a cancer is melanoma cancer. In some embodiments,
a cancer
is breast cancer. In some embodiments, a cancer is lung cancer. In some
embodiments, a cancer
is small cell lung cancer (SCLC). In some embodiments, a cancer is non-small
cell lung cancer
(NSCLC).
1002451 The compounds and compositions, according to the methods of the
present invention,
can be administered using any amount and any route of administration effective
for treating or
lessening the severity of a cancer or tumor. The exact amount required varies
from subject to
subject, depending on the species, age, and general condition of the subject,
the severity of the
disease or condition, the particular agent, its mode of administration, and
the like. Compounds of
the invention are preferably formulated in dosage unit form for ease of
administration and
uniformity of dosage. The expression "dosage unit form" as used herein refers
to a physically
discrete unit of agent appropriate for the patient to be treated. It will be
understood, however, that
the total daily usage of the compounds and compositions of the present
invention is decided by the
attending physician within the scope of sound medical judgment. The specific
effective dose level
for any particular patient or organism depends upon a variety of factors,
including the disorder
being treated and the severity of the disorder; the activity of the specific
compound employed; the
specific composition employed; the age, body weight, general health, sex and
diet of the patient;
the time of administration, route of administration, and rate of excretion of
the specific compound
employed; the duration of the treatment; drugs used in combination or
coincidental with the
specific compound employed, and like factors well known in the medical arts.
The terms "patient"
or "subject", as used herein, means an animal, preferably a mammal, and most
preferably a human.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002461 Pharmaceutically acceptable compositions of this invention can be
administered to
humans and other animals orally, rectally, parenterally, intracisternally,
intravaginally,
intraperitoneally, topically (as by powders, ointments, or drops), bucally, as
an oral or nasal spray,
or the like, depending on the severity of the disease or disorder being
treated. In certain
embodiments, the compounds of the invention can be administered orally or
parenterally at dosage
levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg
to about 25
mg/kg, of subject body weight per day, one or more times a day, to obtain the
desired therapeutic
effect.
1002471 Liquid dosage forms for oral administration include, but are not
limited to,
pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions,
syrups and
elixirs. In addition to the active compounds, the liquid dosage forms may
contain inert diluents
commonly used in the art such as, for example, water or other solvents,
solubilizing agents and
emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl
acetate, benzyl alcohol,
benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide,
oils (in particular,
cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol,
tetrahydrofurfuryl
alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures
thereof Besides inert
diluents, the oral compositions can also include adjuvants such as wetting
agents, emulsifying and
suspending agents, sweetening, flavoring, and perfuming agents.
1002481 Injectable preparations, for example, sterile injectable aqueous or
oleaginous
suspensions may be formulated according to the known art using suitable
dispersing or wetting
agents and suspending agents. The sterile injectable preparation may also be a
sterile injectable
solution, suspension or emulsion in a nontoxic 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 water, Ringer's solution, U.S.P. 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 can be employed including synthetic mono- or diglycerides.
In addition, fatty
acids such as oleic acid are used in the preparation of injectables.
1002491 Injectable formulations can be sterilized, for example, by
filtration through a bacterial-
retaining filter, or by incorporating sterilizing agents in the form of
sterile solid compositions
which can be dissolved or dispersed in sterile water or other sterile
injectable medium prior to use.
86
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002501 In order to prolong the effect of a compound of the present invention,
it is often
desirable to slow the absorption of the compound from subcutaneous or
intramuscular injection.
This may be accomplished by the use of a liquid suspension of crystalline or
amorphous material
with poor water solubility. The rate of absorption of the compound then
depends upon its rate of
dissolution that, in turn, may depend upon crystal size and crystalline form.
Alternatively, delayed
absorption of a parenterally administered compound form is accomplished by
dissolving or
suspending the compound in an oil vehicle. Injectable depot forms are made by
forming
microencapsule matrices of the compound in biodegradable polymers such as
polylactide-
polyglycoli de. Depending upon the ratio of compound to polymer and the nature
of the particular
polymer employed, the rate of compound release can be controlled. Examples of
other
biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot
injectable
formulations are also prepared by entrapping the compound in liposomes or
microemulsions that
are compatible with body tissues.
1002511 Compositions for rectal or vaginal administration are preferably
suppositories which
can be prepared by mixing the compounds of this invention with suitable non-
irritating excipients
or carriers such as cocoa butter, polyethylene glycol or a suppository wax
which are solid at
ambient temperature but liquid at body temperature and therefore melt in the
rectum or vaginal
cavity and release the active compound.
1002521 Solid dosage forms for oral administration include capsules,
tablets, pills, powders, and
granules. In such solid dosage forms, the active compound is mixed with at
least one inert,
pharmaceutically acceptable excipient or carrier such as sodium citrate or
dicalcium phosphate
and/or a) fillers or extenders such as starches, lactose, sucrose, glucose,
mannitol, and silicic acid,
b) binders such as, for example, carboxymethylcellulose, alginates, gelatin,
polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol,
d) disintegrating
agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic
acid, certain silicates,
and sodium carbonate, e) solution retarding agents such as paraffin, f)
absorption accelerators such
as quaternary ammonium compounds, g) wetting agents such as, for example,
cetyl alcohol and
glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i)
lubricants such as
talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium
lauryl sulfate, and
mixtures thereof. In the case of capsules, tablets and pills, the dosage form
may also comprise
buffering agents.
87
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002531 Solid compositions of a similar type can also be employed as
fillers in soft and hard-
filled gelatin capsules using such excipients as lactose or milk sugar as well
as high molecular
weight polyethylene glycols and the like. The solid dosage forms of tablets,
dragees, capsules,
pills, and granules can be prepared with coatings and shells such as enteric
coatings and other
coatings well known in the pharmaceutical formulating art. They can optionally
contain
opacifying agents and can also be of a composition that they release the
active ingredient(s) only,
or preferentially, in a certain part of the intestinal tract, optionally, in a
delayed manner. Examples
of embedding compositions that can be used include polymeric substances and
waxes. Solid
compositions of a similar type can also be employed as fillers in soft and
hard-filled gelatin
capsules using such excipients as lactose or milk sugar as well as high
molecular weight
polethylene glycols and the like.
1002541 The active compounds can also be in micro-encapsulated form with one
or more
excipients as noted above. The solid dosage forms of tablets, dragees,
capsules, pills, and granules
can be prepared with coatings and shells such as enteric coatings, release
controlling coatings and
other coatings well known in the pharmaceutical formulating art. In such solid
dosage forms the
active compound may be admixed with at least one inert diluent such as
sucrose, lactose or starch.
Such dosage forms may also comprise, as is normal practice, additional
substances other than inert
diluents, e.g., tableting lubricants and other tableting aids such a magnesium
stearate and
microcrystalline cellulose. In the case of capsules, tablets and pills, the
dosage forms may also
comprise buffering agents. They may optionally contain opacifying agents and
can also be of a
composition that they release the active ingredient(s) only, or
preferentially, in a certain part of the
intestinal tract, optionally, in a delayed manner. Examples of embedding
compositions that can be
used include polymeric substances and waxes.
1002551 Dosage forms for topical or transderm al administration of a compound
of this invention
include ointments, pastes, creams, lotions, gels, powders, solutions, sprays,
inhalants or patches.
The active component is admixed under sterile conditions with a
pharmaceutically acceptable
carrier and any needed preservatives or buffers as may be required. Ophthalmic
formulation, ear
drops, and eye drops are also contemplated as being within the scope of this
invention.
Additionally, the present invention contemplates the use of transdermal
patches, which have the
added advantage of providing controlled delivery of a compound to the body.
Such dosage forms
can be made by dissolving or dispensing the compound in the proper medium.
Absorption
88
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
enhancers can also be used to increase the flux of the compound across the
skin. The rate can be
controlled by either providing a rate controlling membrane or by dispersing
the compound in a
polymer matrix or gel.
Co-Administration with One or More Other Therapeutic Agent(s)
1002561 Depending upon the particular condition, or disease, to be treated,
additional
therapeutic agents that are normally administered to treat that condition, can
also be present in the
compositions of this invention. As used herein, additional therapeutic agents
that are normally
administered to treat a particular disease, or condition, are known as -
appropriate for the disease,
or condition, being treated."
1002571 In some embodiments, the present invention provides a method of
treating a disclosed
disease or condition comprising administering to a patient in need thereof an
effective amount of
a compound disclosed herein or a pharmaceutically acceptable salt thereof and
co-administering
simultaneously or sequentially an effective amount of one or more additional
therapeutic agents,
such as those described herein. In some embodiments, the method includes co-
administering one
additional therapeutic agent. In some embodiments, the method includes co-
administering two
additional therapeutic agents. In some embodiments, the combination of the
disclosed compound
and the additional therapeutic agent or agents acts synergistically.
1002581 A compound of the current invention can also be used in combination
with known
therapeutic processes, for example, the administration of hormones or
radiation. In certain
embodiments, a provided compound is used as a radiosensitizer, especially for
the treatment of
tumors which exhibit poor sensitivity to radiotherapy.
1002591 A compound of the current invention can be administered alone or in
combination with
one or more other therapeutic compounds, possible combination therapy taking
the form of fixed
combinations or the administration of a compound of the invention and one or
more other
therapeutic compounds being staggered or given independently of one another,
or the combined
administration of fixed combinations and one or more other therapeutic
compounds. A compound
of the current invention can besides, or in addition, be administered
especially for tumor therapy
in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy,
surgical
intervention, or a combination of these. Long-term therapy is equally
possible, as is adjuvant
therapy in the context of other treatment strategies, as described above.
Other possible treatments
89
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
are therapy to maintain the patient's status after tumor regression, or even
chemopreventive
therapy, for example in patients at risk.
[00260] One or more other therapeutic agent(s) may be administered separately
from a
compound or composition of the invention, as part of a multiple dosage
regimen. Alternatively,
one or more other therapeutic agent(s) may be part of a single dosage form,
mixed together with a
compound of this invention in a single composition. If administered as a
multiple dosage regime,
one or more other therapeutic agent(s) and a compound or composition of the
invention can be
administered simultaneously, sequentially or within a period of time from one
another, for example
within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 18, 20,
21, 22, 23, or 24 hours from
one another. In some embodiments, one or more other therapeutic agent(s) and a
compound or
composition of the invention are administered as a multiple dosage regimen
within greater than 24
hours apart.
[00261] As used herein, the term "combination," "combined," and related terms
refers to the
simultaneous or sequential administration of therapeutic agents in accordance
with this invention.
For example, a compound of the present invention can be administered with one
or more other
therapeutic agent(s) simultaneously or sequentially in separate unit dosage
forms or together in a
single unit dosage form. Accordingly, the present invention provides a single
unit dosage form
comprising a compound of the current invention, one or more other therapeutic
agent(s), and a
pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00262] The amount of a compound of the invention and one or more other
therapeutic agent(s)
(in those compositions which comprise an additional therapeutic agent as
described above) that
may be combined with the carrier materials to produce a single dosage form
will vary depending
upon the host treated and the particular mode of administration. Preferably, a
composition of the
invention should be formulated so that a dosage of between 0.01 - 100 mg/kg
body weight/day of
a compound of the invention can be administered.
[00263] In those compositions which comprise one or more other therapeutic
agent(s), the one
or more other therapeutic agent(s) and a compound of the invention may act
synergistically.
Therefore, the amount of the one or more other therapeutic agent(s) in such
compositions may be
less than that required in a monotherapy utilizing only that therapeutic
agent. In such compositions
a dosage of between 0.01 ¨ 1,000 g/kg body weight/day of the one or more
other therapeutic
agent(s) can be administered.
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00264] The amount of one or more other therapeutic agent present in the
compositions of this
invention may be no more than the amount that would normally be administered
in a composition
comprising that therapeutic agent as the only active agent. Preferably the
amount of one or more
other therapeutic agent(s) in the presently disclosed compositions will range
from about 50% to
100% of the amount normally present in a composition comprising that agent as
the only
therapeutically active agent. In some embodiments, one or more other
therapeutic agent(s) is
administered at a dosage of about 50%, about 55%, about 60%, about 65%, about
70%, about 75%,
about 80%, about 85%, about 90%, or about 95% of the amount normally
administered for that
agent. As used herein, the phrase "normally administered" means the amount an
FDA approved
therapeutic agent is approved for dosing per the FDA label insert.
[00265] The compounds of this invention, or pharmaceutical compositions
thereof, may also be
incorporated into compositions for coating an implantable medical device, such
as prostheses,
artificial valves, vascular grafts, stents and catheters. Vascular stents, for
example, have been used
to overcome restenosis (re-narrowing of the vessel wall after injury).
However, patients using
stents or other implantable devices risk clot formation or platelet
activation. These unwanted
effects may be prevented or mitigated by pre-coating the device with a
pharmaceutically
acceptable composition comprising a kinase inhibitor. Implantable devices
coated with a
compound of this invention are another embodiment of the present invention.
Exemplary Other Therapeutic Agents
[00266] In some embodiments, one or more other therapeutic agent is a Poly ADP
ribose
polymerase (PARP) inhibitor. In some embodiments, a PARP inhibitor is selected
from olaparib
(LYNPARZA , AstraZeneca); rucaparib (RUBRACA , Clovis Oncology); niraparib
(ZEJULA , Tesaro); talazoparib (MDV3800/BMN 673/LT00673,
Medivation/Pfizer/Biomarin);
veliparib (ABT-888, AbbVie); and BGB-290 (BeiGene, Inc.).
[00267] In some embodiments, one or more other therapeutic agent is a histone
deacetylase
(HDAC) inhibitor. In some embodiments, an HDAC inhibitor is selected from
vorinostat
(ZOLINZA , Merck); romidepsin (ISTODAX , Celgene); panobinostat (FARYDAK ,
Novara s); belinostat (BELEODAQ , Spectrum Pharmaceuticals); entinostat (SNDX-
275,
Syndax Pharmaceuticals) (NCT00866333); and chidamide (EP1DAZA , HBI-8000,
Chipscreen
Biosciences, China).
91
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002681 In some embodiments, one or more other therapeutic agent is a CDK
inhibitor, such as
a CDK4/CDK6 inhibitor. In some embodiments, a CDK 4/6 inhibitor is selected
from palbociclib
(IBRANCE , Pfizer); ribociclib (KISQALI , Novartis); abemaciclib (Ly2835219,
Eli Lilly); and
trilaciclib (G1T28, G1 Therapeutics).
1002691 In some embodiments, one or more other therapeutic agent is a
phosphatidylinositol 3
kinase (PI3K) inhibitor. In some embodiments, a PI3K inhibitor is selected
from idelalisib
(ZYDELIG , Gilead), alpeli sib (BYL719, Novartis), taseli sib (GDC-0032,
Genentech/Roche);
pictilisib (GDC-0941, Genentech/Roche); copanlisib (BAY 806946, Bayer); duveli
sib (formerly
IPI-145, Infinity Pharmaceuticals); PQR309 (Piqur Therapeutics, Switzerland);
and TGR1202
(formerly RP5230, TG Therapeutics).
1002701 In some embodiments, one or more other therapeutic agent is a platinum-
based
therapeutic, also referred to as platins. Platins cause cross-linking of DNA,
such that they inhibit
DNA repair and/or DNA synthesis, mostly in rapidly reproducing cells, such as
cancer cells.
1002711 In some embodiments, a platinum-based therapeutic is selected from
cisplatin
(PLATINOL , Bristol-Myers Squibb); carboplatin (PARAPLATIN , Bristol-Myers
Squibb;
also, Teva; Pfizer); oxaliplatin (ELOXITIN Sanofi-Aventis); nedaplatin
(AQUPLAO,
Shionogi), picoplatin (Poniard Pharmaceuticals); and satraplatin (JIM-216,
Agennix).
1002721 In some embodiments, one or more other therapeutic agent is a taxane
compound,
which causes disruption of microtubules, which are essential for cell
division. In some
embodiments, a taxane compound is selected from paclitaxel (TAXOL , Bristol-
Myers Squibb),
docetaxel (TAXOTERE , Sanofi-Aventis; DOCEFREZ , Sun Pharmaceutical), albumin-
bound
paclitaxel (ABRAXANE ; Abraxis/Celgene), cabazitaxel (JEVTANA , Sanofi-
Aventis), and
SID530 (SK Chemicals, Co.) (NCT00931008).
1002731 In some embodiments, one or more other therapeutic agent is a
nucleoside inhibitor, or
a therapeutic agent that interferes with normal DNA synthesis, protein
synthesis, cell replication,
or will otherwise inhibit rapidly proliferating cells.
1002741 In some embodiments, a nucleoside inhibitor is selected from
trabectedin (guanidine
alkylating agent, YONDELIS , Janssen Oncology), mechlorethamine (alkylating
agent,
VALCHLOR , Aktelion Pharmaceuticals); vincristine (ONCOVIN , Eli Lilly;
VINCASAR ,
Teva Pharmaceuticals; MARQIBO , Talon Therapeutics); temozolomide (prodrug to
alkylating
agent 5-(3-methyltriazen-1-y1)-imidazole-4-carboxamide (MTIC) TEMODAR ,
Merck);
92
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
cytarabine injection (ara-C, antimetabolic cytidine analog, Pfizer); lomustine
(alkylating agent,
CEENU , Bristol-Myers Squibb; GLEOSTINE , NextSource Biotechnology);
azacitidine
(pyrimidine nucleoside analog of cytidine, VIDAZA , Celgene); omacetaxine
mepesuccinate
(cephalotaxine ester) (protein synthesis inhibitor, SYNRIBOR; Teva
Pharmaceuticals);
asparaginase Erwinia chrysanthemi (enzyme for depletion of asparagine, ELSPAR
, Lundbeck;
ERWINAZE , EUSA Pharma); eribulin mesylate (microtubule inhibitor, tubulin-
based
antimitotic, HALAVEN , Eisai); cabazitaxel (microtubule inhibitor, tubulin-
based antimitotic,
JEVTANA , Sanofi-Aventis); capacetrine (thymidylate synthase inhibitor, XELODA
,
Genentech); bendamustine (bifunctional mechlorethamine derivative, believed to
form interstrand
DNA cross-links, TREANDA , Cephalon/Teva); ixabepilone (semi-synthetic analog
of
epothilone B, microtubule inhibitor, tubulin-based antimitotic, IXEMPRA ,
Bristol-Myers
Squibb); nelarabine (prodrug of deoxyguanosine analog, nucleoside metabolic
inhibitor,
ARRANON , Novartis); clorafabine (prodrug of ribonucleotide reductase
inhibitor, competitive
inhibitor of deoxycytidine, CLOLAR , Sanofi-Aventis); and trifluridine and
tipiracil (thymidine-
based nucleoside analog and thymidine phosphorylase inhibitor, LONSURF , Taiho
Oncology).
1002751 In some embodiments, one or more other therapeutic agent is a kinase
inhibitor or
VEGF-R antagonist. In some embodiments, one or more other therapeutic agent is
a MEK
inhibitor. As used herein, a "IY11FK inhibitor" refers to any inhibitor or
blocker or antagonist that
binds to and/or inhibits mitogen-activated protein kinase enzymes MEK1 and/or
MEK2 In some
embodiments, an MEK inhibitor is selected from those as described in Clieng et
al., "Current
Development Status of MEK Inhibitors," Molecules 2017, 22, 1551, the contents
of which are
incorporated herein by reference in its entirety. In certain embodiments, the
MEK inhibitor is
selected from binimetinib (MEK162, ARRY-438162, ARRAY BIOPHARMA INC.),
cobimetinib(COTELLICR, Exelexis/Genentech/Roche), refametinib (BAY 86-9766,
RDEA119;
Bayer AG), selumetinib (AZD6244, ARRY-142886;
AS TRAZENEC A),
trametinib(MEKINIST , Novartis), mirdametinib(PD-0325901, Spring Works
Therapeutics),
pimasertib (AS703026, MSC1936369B, Merck KGaA)or a pharmaceutically acceptable
salt
and/or solvate of any of the foregoing. In certain embodiments, the second
anti-cancer agent is
binimetinib, cobimetinib, selumetinib, trametinib, mirdametinib, pimasertib,
or a pharmaceutically
acceptable salt and/or solvate of any of the foregoing. Other examples of MEK
inhibitors for use
as an other therapeutic agent in the methods and uses described herein
include, but are not limited
93
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
to, E6201 (Eisai Co Ltd./Strategia Theraputics), GDC-0623 (RG 7421, Genentech,
Inc.),
CH5126766 (R05126766, Chugai 232Pharmaceutica1 Co., Roche), 1-1L-085 (Shanghai
Kechow
Pharma, Inc.), SHR7390 (HENGRUI MEDICINE), TQ-B3234 (CHIATAITIANQING), CS-3006
(CSTON
______________________________________________________________________________
E Pharrn a.ceuti cal s), FCN-159 (FosunPha.rm a.ceutical s), VS-6766 (Verastem
Oncology),
and IMM-1-104 (Immuneering Corp.). Other examples of MEK inhibitors for use as
second anti-
cancer agents in the methods and uses described herein include, but are not
limited to, those
described in W02005/121142, W02014/169843, W02016/035008, W02016/168704,
W02020/125747, W02021/142144, W02021/142345, W02021/149776, the contents of
each of
which are herein incorporated by reference in their entireties.
1002761 In some embodiments, one or more other therapeutic agent is an F(IiFR
inhibitor. As
used herein, an "EGFR inhibitor" refers to any inhibitor or blocker or
antagonist that binds to
and/or inhibits epidermal growth factor receptor (EGFR). In soine embodiments,
an EGFR
inhibitor is selected from those as described in Ayati et al., "A review on
progression of epidermal
growth factor receptor (EGFR) inhibitors as an efficient approach in cancer
targeted therapy,"
Bioorganic Chemistry 2020, 99: 10'3811, the contents of which are incorporated
herein by
reference in its entirety. In some embodiments, an EGFR inhibitor is selected
from cetuximab,
necitumumab, panitumumab, zalutumumab, nimotuzumab, and matuzumab. In some
embodiments, an EGFR inhibitor is cetuximab. In some embodiments, an EGFR
inhibitor is
necitumumab. In some embodiments, an EGFR inhibitor is panitumumab. In some
embodiments,
an EGFR inhibitor is zalutumumab. In some embodiments, an EGFR inhibitor is
nimotuzumab. In
some embodiments, an EGFR inhibitor is matuzumab.
100011
In some embodiments, an EGFR inhibitor is selected from osimertinib,
gefitinib,
erlotinib, lapatinib, neratinib, vandetanib, afatinib, brigatinib,
dacomitinib, and icotinib. In some
embodiments, an EGFR inhibitor is Osimertinib. In some embodiments, an EGFR
inhibitor is
gefitinib. In some embodiments, an EGFR inhibitor is erlotinib. In some
embodiments, an EGFR
inhibitor is lapatinib. In some embodiments, an EGFR inhibitor is neratinib.
In some embodiments,
an EGFR inhibitor is vandetanib. In some embodiments, an EGFR inhibitor is
afatinib. In some
embodiments, an EGFR inhibitor is brigatinib. In some embodiments, an EGFR
inhibitor is
dacomitinib. In some embodiments, an EGFR inhibitor is icotinib.
100021
In some embodiments, an EGFR inhibitor is a "1st generation EGFR
tyrosine kinase
inhibitor" (1st generation TKI). A 18t. generation TKI refers to reversible
EGFR inhibitors, such as
94
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
gefitinib and erlotinib, which are effective in first-line treatment of NSCLC
harboring EGFR
activating mutations such as deletions in exon 19 and exon 21 L858R mutation.
[0003] In some embodiments, an EGFR inhibitor is a -2nd generation
EGFR tyrosine kinase
inhibitor" (2nd generation TKI) A 2' generation TKI refers to covalent
irreversible EGFR
inhibitors, such as afatinib and dacomitib, which are effective in first-line
treatment of NSCLC
harboring EGFR activating mutations such as deletions in exon 19 and exon 21
L858R mutation.
[00277] In some embodiments, an EGFR inhibitor is a "3rd generation EGFR
tyrosine kinase
inhibitor" (3rd generation TKI). A 3rd generation TKI refers to covalent
irreversible EGFR
inhibitors, such as osimertinib and lazertinib, which are selective to the
EGFR activating
mutations, such as deletions in exon 19 and exon 21 L858R, alone or in
combination with T790M
mutation, and have lower inhibitory activity against wild-type EGFR.
[00278] In some embodiment, one or more Whet therapeutic agent is selected
from approved
VEGF inhibitors and kinase inhibitors useful in the present invention include:
bevacizumab
(AVASTIN , Genentech/Roche) an anti-VEGF monoclonal antibody; ramucirumab
(CYRANIZA , Eli Lilly), an anti-VEGFR-2 antibody and ziv-aflibercept, also
known as VEGF
Trap (ZALTRAPO; Regeneron/Sanofi). VEGFR inhibitors, such as regorafenib
(STIVARGAO,
Bayer); vandetanib (CAPRELSA , Astra7eneca); axitinib (INLYTA , Pfizer); and
lenvatinib
(LENVIMA , Eisai); Raf inhibitors, such as sorafenib (NEXAVAR , Bayer AG and
Onyx);
dabrafenib (TAFINLAR , Novartis); and vemurafenib (ZELBORAFO,
Genentech/Roche); MEK
inhibitors, such as cobim etanib (COTELLIC , Exel exi s/Genentech/Roche);
tram eti nib
(MEKINIST , Novartis); Bcr-Abl tyrosine kinase inhibitors, such as imatinib
(GLEEVECO,
Novartis); nil otinib (TASIGNA , Novartis); dasatinib (SPRYCEL ,
BristolMyersSquibb);
bosutinib (BOSULIF , Pfizer); and ponatinib (INCLUSIG , Ariad
Pharmaceuticals); Her2 and
EGFR inhibitors, such as gefitinib (IRESSA , AstraZeneca); erlotinib
(TARCEEVAR,
Genentech/Roche/Astellas), lapatinib (TYKERB , Novartis), afatinib (GILOTRIF ,
Boehringer
Ingelheim), osimertinib (targeting activated EGFR, TAGRISSO , AstraZeneca),
and brigatinib
(ALUNBRIG , Ariad Pharmaceuticals), c-Met and VEGFR2 inhibitors, such as
cabozanitib
(COMETRIQ , Exelexis), and multikinase inhibitors, such as sunitinib (SUTENT ,
Pfizer),
pazopanib (VOTRIENTO, Novartis); ALK inhibitors, such as crizotinib (XALKORI ,
Pfizer);
ceritinib (ZYKADIA , Novartis); and alectinib (ALECENZa , Genentech/Roche);
Bruton's
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
tyrosine kinase inhibitors, such as ibrutinib (IIVIBRUVICA ,
Pharmacyclics/Janssen); and Flt3
receptor inhibitors, such as midostaurin (RYDAPT , Novartis).
1002791 Other kinase inhibitors and VEGF-R antagonists that are in development
and may be
used in the present invention include tivozanib (Aveo Pharm aecuti cal s);
vatal anib
(Bayer/Novartis); lucitanib (Clovis Oncology); dovitinib (TKI258, Novartis);
Chiauanib
(Chipscreen Biosciences); CEP-11981 (Cephalon); linifanib (Abbott
Laboratories); neratinib
(FIKI-272, Puma Biotechnology); radotinib (SUPECT , IY5511, 11-Yang
Pharmaceuticals, S.
Korea); ruxolitinib (JAKAFI , Incyte Corporation); P1C299 (PTC Therapeutics);
CP-547,632
(Pfizer); foretinib (Exelexis, GlaxoSmithKline); quizartinib (Daiichi Sankyo)
and motesanib
(Amgen/Takeda)
1002801 In some embodiments, one or more other therapeutic agent is an mTOR
inhibitor,
which inhibits cell proliferation, angiogenesis and glucose uptake. In some
embodiments, an
mTOR inhibitor is everolimus (AFINITOR , Novartis); temsirolimus (TORISEL ,
Pfizer); and
sirolimus (RAPAMUNE , Pfizer).
1002811 In some embodiments, one or more other therapeutic agent is a
proteasome inhibitor.
Approved proteasome inhibitors useful in the present invention include
bortezomib (VELCADE ,
Takeda); carfilzomib (KYPROLIS , Amgen); and ixazomib (NINLARO , Takeda).
1002821 In some embodiments, one or more other therapeutic agent is a growth
factor
antagonist, such as an antagonist of platelet-derived growth factor (PDGF), or
epidermal growth
factor (EGF) or its receptor (EGFR) Approved PDGF antagonists which may be
used in the
present invention include olaratumab (LARTRUVO ; Eli Lilly). Approved EGFR
antagonists
which may be used in the present invention include cetuximab (ERBITUX , Eli
Lilly);
necitumumab (PORTRAZZA , Eli Lilly), panitumumab (VECTIBIX , Amgen); and
osimertinib
(targeting activated EGFR, TAGRISSOR, AstraZeneca)
1002831 In some embodiments, one or more other therapeutic agent is an
aromatase inhibitor.
In some embodiments, an aromatase inhibitor is selected from exemestane
(AROMASIN ,
Pfizer), anastazole (ARIMIDEX , Astra7eneca) and letrozole (FEMARA ,
Novartis).
1002841 In some embodiments, one or more other therapeutic agent is an
antagonist of the
hedgehog pathway. Approved hedgehog pathway inhibitors which may be used in
the present
invention include sonidegib (ODOMZO , Sun Pharmaceuticals); and vismodegib
(ERIVEDGE , Genentech), both for treatment of basal cell carcinoma.
96
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00285] In some embodiments, one or more other therapeutic agent is a folic
acid inhibitor.
Approved folic acid inhibitors useful in the present invention include
pemetrexed (ALIMTA , Eli
Lilly).
[00286] In some embodiments, one or more other therapeutic agent is a CC
chemokine receptor
4 (CCR4) inhibitor. CCR4 inhibitors being studied that may be useful in the
present invention
include mogamulizumab (POTELIGEO , Kyowa Hakko Kirin, Japan).
[00287] In some embodiments, one or more other therapeutic agent is an
isocitrate
dehydrogenase (IDH) inhibitor. IDH inhibitors being studied which may be used
in the present
invention include AG120 (Celgene; NCT02677922); A6221 (Cel gene, NCT02677922,
NCT02577406); BAY1436032 (Bayer, NCT02746081); IDH305 (Novartis, NCT02987010)
[00288] In some embodiments, one or more other therapeutic agent is an
arginase inhibitor.
Arginase inhibitors being studied which may be used in the present invention
include AEB1102
(pegylated recombinant arginase, Aeglea Biotherapeutics), which is being
studied in Phase 1
clinical trials for acute myeloid leukemia and myelodysplastic syndrome
(NCT02732184) and
solid tumors (NCT02561234); and CB-1158 (Calithera Biosciences).
1002891 In some embodiments, one or more other therapeutic agent is a
glutaminase inhibitor.
Glutaminase inhibitors being studied which may be used in the present
invention include CB-839
(Calithera Biosciences).
[00290] In some embodiments, one or more other therapeutic agent is an
antibody that binds to
tumor antigens, that is, proteins expressed on the cell surface of tumor cells
Approved antibodies
that bind to tumor antigens which may be used in the present invention include
rituximab
(RITUXAN , Genentech/BiogenIdec); ofatumumab (anti-CD20, ARZERRA ,
GlaxoSmithKline); obinutuzumab (anti-CD20, GAZYVA , Genentech), ibritumomab
(anti-
CD20 and Yttrium-90, ZEVALINR, Spectrum Pharmaceuticals); daratumumab (anti-
CD38,
DARZALEX , Janssen Biotech), dinutuximab (anti-glycolipid GD2, UNITUXIN ,
United
Therapeutics), trastuzumab (anti-1-IER2, HERCEPTIN , Genentech), ado-
trastuzumab emtansine
(anti-HER2, fused to emtansine, KADCYLA , Genentech), and pertuzumab (anti-
HER2,
PERJETA , Genentech), and brentuximab vedotin (anti-CD30-drug conjugate,
ADCETRIS ,
Seattle Genetics).
1002911 In some embodiments, one or more other therapeutic agent is a
topoisomerase inhibitor.
Approved topoisomerase inhibitors useful in the present invention include
irinotecan
97
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
(ONIVYDE , Merrimack Pharmaceuticals); topotecan (HYCAMTIN , GlaxoSmithKline).

Topoisomerase inhibitors being studied which may be used in the present
invention include
pixantrone (PIXUVRIO, CTI Biopharma).
1002921 In some embodiments, one or more other therapeutic agent is an
inhibitor of anti-
apoptotic proteins, such as BCL-2. Approved anti-apoptotics which may be used
in the present
invention include venetoclax (VENCLEXTA , AbbVie/Genentech); and blinatumomab
(BLINCYTO , Amgen). Other therapeutic agents targeting apoptotic proteins
which have
undergone clinical testing and may be used in the present invention include
navitoclax (ABT-263,
Abbott), a BCL-2 inhibitor (NCT02079740).
1002931 In some embodiments, one or more other therapeutic agent is an
androgen receptor
inhibitor. Approved androgen receptor inhibitors useful in the present
invention include
enzalutamide (XTANDI , Astellas/Medivation), approved inhibitors of androgen
synthesis
include abiraterone (ZYTIGA , Centocor/Ortho); approved antagonist of
gonadotropin-releasing
hormone (GnRH) receptor (degaralix, FIRMAGON , Ferring Pharmaceuticals).
1002941 In some embodiments, one or more other therapeutic agent is a
selective estrogen
receptor modulator (SERM), which interferes with the synthesis or activity of
estrogens.
Approved SERMs useful in the present invention include raloxifene (EVISTA ,
Eli Lilly).
1002951 In some embodiments, one or more other therapeutic agent is an
inhibitor of bone
resorption. An approved therapeutic which inhibits bone resorption is
Denosumab (XGEVA ,
Amgen), an antibody that binds to RANKL, prevents binding to its receptor
RANK, found on the
surface of osteoclasts, their precursors, and osteoclast-like giant cells,
which mediates bone
pathology in solid tumors with osseous metastases. Other approved therapeutics
that inhibit bone
resorption include bisphosphonates, such as zoledronic acid (ZOMETAR,
Novartis).
1002961 In some embodiments, one or more other therapeutic agent is an
inhibitor of interaction
between the two primary p53 suppressor proteins, MDMX and MDM2. Inhibitors of
p53
suppression proteins being studied which may be used in the present invention
include ALRN-
6924 (Aileron), a stapled peptide that equipotently binds to and disrupts the
interaction of MDMX
and MDM2 with p53. ALRN-6924 is currently being evaluated in clinical trials
for the treatment
of AML, advanced myelodysplastic syndrome (MDS) and peripheral T-cell lymphoma
(PTCL)
(NCT02909972; NCT02264613).
98
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1002971 In some embodiments, one or more other therapeutic agent is an
inhibitor of
transforming growth factor-beta (TGF-beta or TGF-13). Inhibitors of TGF-beta
proteins being
studied which may be used in the present invention include NIS793 (Novartis),
an anti-TGF-beta
antibody being tested in the clinic for treatment of various cancers,
including breast, lung,
hepatocellular, colorectal, pancreatic, prostate and renal cancer (NCT
02947165). In some
embodiments, the inhibitor of TGF-beta proteins is fresolimumab (GC1008;
Sanofi-Genzyme),
which is being studied for melanoma (NCT00923169); renal cell carcinoma
(NCT00356460); and
non-small cell lung cancer (NCT02581787). Additionally, in some embodiments,
the additional
therapeutic agent is a TGF-beta trap, such as described in Connolly et al.
(2012) Int'l J. Biological
Sciences 8:964-978. One therapeutic compound currently in clinical trials for
treatment of solid
tumors is M7824 (Merck KgaA - formerly MSB0011459X), which is a bispecific,
anti-PD-
L1/TGF-13 trap compound (NCT02699515); and (NCT02517398). M7824 is comprised
of a fully
human IgG1 antibody against PD-Li fused to the extracellular domain of human
TGF-beta
receptor II, which functions as a TGF-13 "trap.-
1002981 In some embodiments, one or more other therapeutic agent is selected
from
glembatumumab vedotin-monomethyl auristatin E (MMAE) (Celldex), an anti-
glycoprotein NMB
(gpNMB) antibody (CR011) linked to the cytotoxic MMAE. gpNMB is a protein
overexpressed
by multiple tumor types associated with cancer cells' ability to metastasize.
1002991 In some embodiments, one or more other therapeutic agents is an
antiproliferative
compound. Such antiproliferative compounds include, but are not limited to
aromatase inhibitors;
antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors;
microtubule active
compounds; alkylating compounds; histone deacetylase inhibitors; compounds
which induce cell
differentiation processes; cyclooxygenase inhibitors; M_MP inhibitors; mTOR
inhibitors;
antineoplastic antimetabolites; platin compounds; compounds
targeting/decreasing a protein or
lipid kinase activity and further anti-angiogenic compounds; compounds which
target, decrease or
inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists;
anti-androgens,
methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors;
bisphosphonates;
biological response modifiers; antiproliferative antibodies; heparanase
inhibitors; inhibitors of Ras
oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds
used in the
treatment of hematologic malignancies; compounds which target, decrease or
inhibit the activity
of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin,
NSC330507), 17-
99
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-
504,
CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide
(TEMODALc));
kinesin spindle protein inhibitors, such as SB715992 or SB743921 from
GlaxoSmithKline, or
pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886
from
Array BioPharma, AZd6244 from AstraZeneca, PD181461 from Pfizer and
leucovorin.
[00300] The term "aromatase inhibitor" as used herein relates to a compound
which inhibits
estrogen production, for instance, the conversion of the substrates
androstenedione and
testosterone to estrone and estradiol, respectively. The term includes, but is
not limited to steroids,
especially atam estan e, ex em estane and form estane and, in particular, non-
steroids, especially
aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone,
ketokonazole,
vorozole, fadrozole, anastrozole and letrozole. Exemestane is marketed under
the trade name
AROMASINTm. Formestane is marketed under the trade name LENTARONTm. Fadrozole
is
marketed under the trade name AFEMATm. Anastrozole is marketed under the trade
name
ARIMIDEXTm. Letrozole is marketed under the trade names FEMARATm or FEMArTm.
Aminoglutethimide is marketed under the trade name ORIMETENTm. A combination
of the
invention comprising a chemotherapeutic agent which is an aromatase inhibitor
is particularly
useful for the treatment of hormone receptor positive tumors, such as breast
tumors.
[00301] The term "antiestrogen" as used herein relates to a compound which
antagonizes the
effect of estrogens at the estrogen receptor level. The term includes, but is
not limited to tamoxifen,
fulvestrant, ral oxi fen e and ral oxi fen e hydrochloride. Tam oxi fen is
marketed under the trade name
NOLVADEXTM. Raloxifene hydrochloride is marketed under the trade name
EVISTATm.
Fulvestrant can be administered under the trade name FASLODEXTmFulvestrant can
be
administered under the trade name FaslodexTM. A combination of the invention
comprising a
chemotherapeutic agent which is an anti estrogen is particularly useful for
the treatment of estrogen
receptor positive tumors, such as breast tumors.
[00302] The term "anti-androgen" as used herein relates to any substance which
is capable of
inhibiting the biological effects of androgenic hormones and includes, but is
not limited to,
bicalutamide (CASODEXTm). The term "gonadorelin agonist" as used herein
includes, but is not
limited to abarelix, goserelin, and goserelin acetate. Goserelin can be
administered under the trade
name ZOLADEXTM.
100
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00303] The term "topoisomerase I inhibitor" as used herein includes,
but is not limited to
topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-
nitrocamptothecin and the
macromolecular camptothecin conjugate PNU-1 66148. Irinotecan can be
administered, e.g., in the
form as it is marketed, e.g., under the trademark CAMPTOSARTm. Topotecan is
marketed under
the trade name HYCAMPTINTm.
[00304] The term "topoisomerase II inhibitor" as used herein
includes, but is not limited to the
anthracyclines such as doxorubicin (including liposomal formulation, such as
CAELYXTm),
daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones
mitoxantrone and
losoxantrone, and the podophillotoxines etoposide and teniposide. Etoposi de
is marketed under
the trade name ETOPOPHOSTm. Teniposide is marketed under the trade name VM 26-
Bristol
Doxorubicin is marketed under the trade name ACRIBLASTINTm or ADRIAMYCINTm.
Epirubicin is marketed under the trade name FARMORUBICINTm. Idarubicin is
marketed, under
the trade name ZAVEDOSTM. Mitoxantrone is marketed under the trade name
NOVANTRONTm.
[00305] The term "microtubule active agent" relates to microtubule
stabilizing, microtubule
destabilizing compounds and microtublin polymerization inhibitors including,
but not limited to
taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as
vinblastine or vinblastine
sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides;
cochicine and
epothilones and derivatives thereof. Paclitaxel is marketed under the trade
name TAXOLTm.
Docetaxel is marketed under the trade name TAXOTERETm. Vinblastine sulfate is
marketed under
the trade name VINBLASTIN R.PTM. Vincristine sulfate is marketed under the
trade name
FARMIS TINTm.
[00306] The term "alkylating agent" as used herein includes, but is
not limited to,
cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel).
Cyclophosphamide
is marketed under the trade name CYCLOSTINTm. Ifosfami de is marketed under
the trade name
HOLOXANTM.
[00307] The term "histone deacetylase inhibitors" or "HDAC inhibitors" relates
to compounds
which inhibit the histone deacetylase and which possess antiproliferative
activity. This includes,
but is not limited to, suberoylanilide hydroxamic acid (SAHA).
[00308] The term "antineoplastic antimetabolite" includes, but is not
limited to, 5-fluorouracil
or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-
azacytidine and
decitabine, methotrexate and edatrexate, and folic acid antagonists such as
pemetrexed.
101
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Capecitabine is marketed under the trade name XELODATM. Gemcitabine is
marketed under the
trade name GEMZARTm.
1003091 The term "platin compound" as used herein includes, but is not limited
to, carboplatin,
cis-platin, cisplatinum and oxaliplatin Carboplatin can be administered, e.g.,
in the form as it is
marketed, e.g., under the trademark CARBOPLATTm. Oxaliplatin can be
administered, e.g., in the
form as it is marketed, e.g. under the trademark ELOXATINTm.
[00310] The term "compounds targeting/decreasing a protein or lipid kinase
activity; or a
protein or lipid phosphatase activity; or further anti-angiogenic compounds"
as used herein
includes, but is not limited to, protein tyrosine kinase and/or serine and/or
threonine kinase
inhibitors or lipid kinase inhibitors, such as a) compounds targeting,
decreasing or inhibiting the
activity of the platelet-derived growth factor-receptors (PDGFR), such as
compounds which target,
decrease or inhibit the activity of PDGFR, especially compounds which inhibit
the PDGF receptor,
such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101,
SU6668 and GFB-
111; b) compounds targeting, decreasing or inhibiting the activity of the
fibroblast growth factor-
receptors (FGFR); c) compounds targeting, decreasing or inhibiting the
activity of the insulin-like
growth factor receptor I (IGF-IR), such as compounds which target, decrease or
inhibit the activity
of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I
receptor, or antibodies
that target the extracellular domain of IGF-I receptor or its growth factors;
d) compounds targeting,
decreasing or inhibiting the activity of the Trk receptor tyrosine kinase
family, or ephrin B4
inhibitors; e) compounds targeting, decreasing or inhibiting the activity of
the AxI receptor
tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the
activity of the Ret
receptor tyrosine kinase; g) compounds targeting, decreasing or inhibiting the
activity of the
Kit/SCFR receptor tyrosine kinase, such as imatinib; h) compounds targeting,
decreasing or
inhibiting the activity of the C-kit receptor tyrosine kinases, which are part
of the PDGFR family,
such as compounds which target, decrease or inhibit the activity of the c-Kit
receptor tyrosine
kinase family, especially compounds which inhibit the c-Kit receptor, such as
imatinib, i)
compounds targeting, decreasing or inhibiting the activity of members of the c-
Abl family, their
gene-fusion products (e.g., BCR-Abl kinase) and mutants, such as compounds
which target
decrease or inhibit the activity of c-Abl family members and their gene fusion
products, such as
an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib
(AMN107); PD180970;
AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j)
compounds
102
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
targeting, decreasing or inhibiting the activity of members of the protein
kinase C (PKC) and Raf
family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAX,
PDK1,
PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family, and/or members of the
cyclin-
dependent kinase family (CDK) including staurosporine derivatives, such as
midostaurin;
examples of further compounds include UCN-01, safingol, BAY 43-9006,
Bryostatin 1,
Perifosine; llmofosine; RO 318220 and RO 320432, GO 6976; lsis 3521;
LY333531/LY379196;
isochinoline compounds; FTIs; PD184352 or QAN697 (a P 13K inhibitor) or A17519
(CDK
inhibitor); k) compounds targeting, decreasing or inhibiting the activity of
protein-tyrosine kinase
inhibitors, such as compounds which target, decrease or inhibit the activity
of protein-tyrosine
kinase inhibitors include imatinib mesylate (GLEEVECTM) or tyrphostin such as
Tyrphostin
A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490,

Tyrphostin B44, Tyrphostin B44 (+) enantiomer, Tyrphostin AG 555, AG 494,
Tyrphostin AG
556, AG957 and adaphostin (4-{[(2,5- dihydroxyphenyl)methyl]aminol-benzoic
acid adamantyl
ester; NSC 680410, adaphostin); 1) compounds targeting, decreasing or
inhibiting the activity of
the epidermal growth factor family of receptor tyrosine kinases (EGFRIErbB2,
ErbB3, ErbB4 as
homo- or heterodimers) and their mutants, such as compounds which target,
decrease or inhibit
the activity of the epidermal growth factor receptor family are especially
compounds, proteins or
antibodies which inhibit members of the EGF receptor tyrosine kinase family,
such as EGF
receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, CP
358774, ZD 1839,
ZM 105180; trastuzumab (HERCEPTINTm), cetuximab (ERBITUXTm), Iressa, Tarceva,
OSI-774,
C1-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or
E7.6.3, and 7H-
pyrrolo-[2,3-d]pyrimidine derivatives; m) compounds targeting, decreasing or
inhibiting the
activity of the c-Met receptor, such as compounds which target, decrease or
inhibit the activity of
c-Met, especially compounds which inhibit the kinase activity of c-Met
receptor, or antibodies that
target the extracellular domain of c-Met or bind to HGF, n) compounds
targeting, decreasing or
inhibiting the kinase activity of one or more JAK family members
(JAK1/JAK2/JAK3/TYK2
and/or pan-JAK), including but not limited to PRT-062070, SB-1578,
baricitinib, pacritinib,
momelotinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxolitinib, o)
compounds
targeting, decreasing or inhibiting the kinase activity of PI3 kinase (PI3K)
including but not limited
to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib,
pictrelisib,
PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib; and; and q)
compounds
103
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
targeting, decreasing or inhibiting the signaling effects of hedgehog protein
(Hh) or smoothened
receptor (SMO) pathways, including but not limited to cyclopamine, vismodegib,
itraconazole,
erismodegib, and IPI-926 (saridegib).
[00311]
The term "PI3K inhibitor" as used herein includes, but is not limited
to compounds
having inhibitory activity against one or more enzymes in the
phosphatidylinosito1-3-kinase
family, including, but not limited to PI3Ka, P13K1, PI3Ko, PI3K13, PI3K-C2a,
PI3K-C213, PI3K-
C27, Vps34, p110-a, p110-13, p110-7, p110-6, p85-a,
p55-7, p150, p101, and p87. Examples
of PI3K inhibitors useful in this invention include but are not limited to ATU-
027, SF-1126, DS-
7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502,
BYL-719,
dactolisib, XL-147, XL-765, and idelalisib
[00312] The term "Bc1-2 inhibitor" as used herein includes, but is not limited
to compounds
having inhibitory activity against B-cell lymphoma 2 protein (Bc1-2),
including but not limited to
ABT-199, ABT-731, ABT-737, apogossypol, Ascenta's pan-Bc1-2 inhibitors,
curcumin (and
analogs thereof),
dual B c1-2/B cl-xL inhibitors (Infinity Pharm aceuti cal s/Novar ti s
Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see
W02008118802),
navitoclax (and analogs thereof, see US7390799), NH-1 (Shenayng Pharmaceutical
University),
obatoclax (and analogs thereof, see W02004106328), S-001 (Gloria
Pharmaceuticals), TW series
compounds (Univ. of Michigan), and venetoclax. In some embodiments the Bc1-2
inhibitor is a
small molecule therapeutic. In some embodiments the Bc1-2 inhibitor is a
peptidomimetic.
[00313] The term "BTK inhibitor" as used herein includes, but is not limited
to compounds
having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including,
but not limited to
AVL-292 and ibrutinib.
[00314] The term "SYK inhibitor" as used herein includes, but is not limited
to compounds
having inhibitory activity against spleen tyrosine kinase (SYK), including but
not limited to PRT-
062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib.
[00315] Further examples of BTK inhibitory compounds, and conditions treatable
by such
compounds in combination with compounds of this invention can be found in
W02008039218
and W02011090760, the entirety of which are incorporated herein by reference.
[00316] Further examples of SYK inhibitory compounds, and conditions treatable
by such
compounds in combination with compounds of this invention can be found in
W02003063794,
W02005007623, and W02006078846, the entirety of which are incorporated herein
by reference.
104
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00317] Further examples of PI3K inhibitory compounds, and conditions
treatable by such
compounds in combination with compounds of this invention can be found in
W02004019973,
W02004089925, W02007016176, US8138347, W02002088112, W02007084786,
W02007129161, W02006122806, W02005113554, and W02007044729 the entirety of
which
are incorporated herein by reference.
[00318] Further examples of JAK inhibitory compounds, and conditions treatable
by such
compounds in combination with compounds of this invention can be found in
W02009114512,
W02008109943, W02007053452, W02000142246, and W02007070514, the entirety of
which
are incorporated herein by reference
[00319] Further anti-angiogenic compounds include compounds having another
mechanism for
their activity, e.g., unrelated to protein or lipid kinase inhibition e.g.,
thalidomide (THALOMIDTm)
and TNP-470.
[00320] Examples of proteasome inhibitors useful for use in combination with
compounds of
the invention include, but are not limited to bortezomib, disulfiram,
epigallocatechin-3-gallate
(EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
1003211 Compounds which target, decrease or inhibit the activity of a protein
or lipid
phosphatase are e.g., inhibitors of phosphatase 1, phosphatase 2A, or CDC25,
such as okadaic acid
or a derivative thereof.
[00322] Compounds which induce cell differentiation processes include, but are
not limited to,
retinoic acid, cc- 7- or 6- tocopherol or a- 7- or 6-tocotrienol
[00323] The term cyclooxygenase inhibitor as used herein includes, but is not
limited to, Cox-
2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and
derivatives, such as celecoxib (
1003241 CELEBREXTm), rofecoxib (VIOXXTm), etoricoxib, valdecoxib or a 5-alkyl-
2-
aryl aminophenyl acetic acid, such as 5-methyl-2-(2'-chloro-6'-
fluoroanilino)phenyl acetic acid,
lumiracoxib.
1003251 The term "bisphosphonates" as used herein includes, but is
not limited to, etridonic,
clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and
zoledronic acid. Etridonic
acid is marketed under the trade name DIDRONELTM. Clodronic acid is marketed
under the trade
name BONEFOSTM. Tiludronic acid is marketed under the trade name SkelidTM.
Pamidronic acid
is marketed under the trade name AREDIATM. Alendronic acid is marketed under
the trade name
FOSAMAXTm. Ibandronic acid is marketed under the trade name BONDRANATTm.
Risedronic
105
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
acid is marketed under the trade name ACTONELTm. Zoledronic acid is marketed
under the trade
name ZOMETATm. The term "mTOR inhibitors" relates to compounds which inhibit
the
mammalian target of rapamycin (mTOR) and which possess antiproliferative
activity such as
sirolimus (RAPAMUNER), everolimus (CERTICANTm), CCI-779 and ABT578
1003261 The term "heparanase inhibitor" as used herein refers to compounds
which target,
decrease or inhibit heparin sulfate degradation. The term includes, but is not
limited to, PI-88. The
term "biological response modifier" as used herein refers to a lymphokine or
interferons.
1003271 The term "inhibitor of Ras oncogenic isoforms", such as H-Ras, K-Ras,
or N-Ras, as
used herein refers to compounds which target, decrease or inhibit the
oncogenic activity of Ras,
for example, a "farnesyl transferase inhibitor" such as L-744832, DK8G557 or
R115777
(ZARNESTRA TM). The term "telomerase inhibitor" as used herein refers to
compounds which
target, decrease or inhibit the activity of telomerase. Compounds which
target, decrease or inhibit
the activity of telomerase are especially compounds which inhibit the
telomerase receptor, such as
telomestatin.
1003281 The term "methionine aminopeptidase inhibitor" as used herein refers
to compounds
which target, decrease or inhibit the activity of methionine aminopeptidase.
Compounds which
target, decrease or inhibit the activity of methionine aminopeptidase include,
but are not limited
to, bengamide or a derivative thereof.
1003291 The term "proteasome inhibitor" as used herein refers to compounds
which target,
decrease or inhibit the activity of the proteasome Compounds which target,
decrease or inhibit the
activity of the proteasome include, but are not limited to, Bortezomib
(VELCADETM) and MLN
341.
1003301 The term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) as
used herein
includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic
inhibitors,
tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor
batimastat and its orally
bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat
(NSC 683551)
BMS-279251, BAY 12-9566, TAA211 , 1VIMI270B or AAJ996.
1003311 The term "compounds used in the treatment of hematologic malignancies"
as used
herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors,
which are compounds
targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase
receptors (Flt-3R);
106
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
interferon, 1-13-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK
inhibitors, which are
compounds which target, decrease or inhibit anaplastic lymphoma kinase.
[00332] Compounds which target, decrease or inhibit the activity of FMS-like
tyrosine kinase
receptors (Flt-3R) are especially compounds, proteins or antibodies which
inhibit members of the
Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine
derivative, SU11248
and MLN518.
[00333] The term "HSP90 inhibitors" as used herein includes, but is not
limited to, compounds
targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90;
degrading, targeting,
decreasing or inhibiting the HSP90 client proteins via the ubiquitin
proteosome pathway.
Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of
HSP90 are
especially compounds, proteins or antibodies which inhibit the ATPase activity
of HSP90, such as
17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative,
other
geldanamycin related compounds; radicicol and HDAC inhibitors.
[00334] The term "antiproliferative antibodies" as used herein
includes, but is not limited to,
trastuzumab (HERCEPTINTm), Trastuzumab-DM1, erbitux, bevacizumab (AVASTINTm),
rituximab (RITUXAN(9), PR064553 (anti-CD40) and 2C4 Antibody. By antibodies is
meant intact
monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed
from at least 2
intact antibodies, and antibodies fragments so long as they exhibit the
desired biological activity.
[00335] For the treatment of acute myeloid leukemia (AML), compounds of the
current
invention can be used in combination with standard leukemia therapies,
especially in combination
with therapies used for the treatment of AML. In particular, compounds of the
current invention
can be administered in combination with, for example, farnesyl transferase
inhibitors and/or other
drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-
C, VP-16,
Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
[00336] Other anti-leukemic compounds include, for example, Ara-C, a
pyrimidine analog,
which is the f-alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine.
Also included is
the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine
phosphate.
Compounds which target, decrease or inhibit activity of histone deacetylase
(HDAC) inhibitors
such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the
activity of the
enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275,
SAHA,
FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US
6,552,065 including,
107
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
but not limited to, N-hydroxy-3-14-1112-(2-methy1-1H-indo1-3-y1)-ethy11-
amino]methyllphenyl]-
2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-
3-[4-[(2-
hydroxyethy1){2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide,
or a
pharmaceutically acceptable salt thereof, especially the lactate salt
Somatostatin receptor
antagonists as used herein refer to compounds which target, treat or inhibit
the somatostatin
receptor such as octreotide, and S0M230. Tumor cell damaging approaches refer
to approaches
such as ionizing radiation. The term "ionizing radiation" referred to above
and hereinafter means
ionizing radiation that occurs as either electromagnetic rays (such as X-rays
and gamma rays) or
particles (such as alpha and beta particles). Ionizing radiation is provided
in, but not limited to,
radiation therapy and is known in the art. See Hellman, Principles of
Radiation Therapy, Cancer,
in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol.
1 , pp. 248-275 (1993).
[00337] Also included are EDG binders and ribonucleotide reductase inhibitors.
The term
"EDG binders" as used herein refers to a class of immunosuppressants that
modulates lymphocyte
recirculation, such as FTY720. The term "ribonucleotide reductase inhibitors-
refers to pyrimidine
or purine nucleoside analogs including, but not limited to, fludarabine and/or
cytosine arabinoside
(ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine
(especially in combination
with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase
inhibitors are especially
hydroxyurea or 2-hydroxy-1H-isoindole-1 ,3 -di one derivatives.
[00338] Also included are in particular those compounds, proteins or
monoclonal antibodies of
VEGF such as 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a
pharmaceutically
acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine
succinate;
ANGIOSTATINTm; ENDOSTATINTm; anthranilic acid amides; ZD4190; Zd6474; SU5416;
SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies,
such as
rhuMAb and RHUFab, VEGF aptamer such as Macugon; FLT-4 inhibitors, FLT-3
inhibitors,
VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (AVASTINTm).
[00339] Photodynamic therapy as used herein refers to therapy which uses
certain chemicals
known as photosensitizing compounds to treat or prevent cancers. Examples of
photodynamic
therapy include treatment with compounds, such as VISUDYNETM and porfimer
sodium.
[00340] Angiostatic steroids as used herein refers to compounds which block or
inhibit
angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-
epihydrocotisol,
108
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
cortexol one, 1 7a-hydroxyprogesterone, corticosterone, desoxycorticosterone,
testosterone,
estrone and dexamethasone.
1003411 Implants containing corticosteroids refers to compounds, such as
fluocinolone and
dexam ethasone.
1003421 Other chemotherapeutic compounds include, but are not limited to,
plant alkaloids,
hormonal compounds and antagonists; biological response modifiers, preferably
lymphokines or
interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA
or siRNA; or
miscellaneous compounds or compounds with other or unknown mechanism of
action.
1003431 The structure of the active compounds identified by code numbers,
generic or trade
names may be taken from the actual edition of the standard compendium "The
Merck Index" or
from databases, e.g., Patents International (e.g., IMS World Publications).
Exemplary Immuno-Oncology agents
1003441 In some embodiments, one or more other therapeutic agent is an immuno-
oncology
agent. As used herein, the term "an immuno-oncology agent" refers to an agent
which is effective
to enhance, stimulate, and/or up-regulate immune responses in a subject. In
some embodiments,
the administration of an immuno-oncology agent with a compound of the
invention has a synergic
effect in treating a cancer.
1003451 An immuno-oncology agent can be, for example, a small molecule drug,
an antibody,
or a biologic or small molecule. Examples of biologic immuno-oncology agents
include, but are
not limited to, cancer vaccines, antibodies, and cytokines. In some
embodiments, an antibody is a
monoclonal antibody. In some embodiments, a monoclonal antibody is humanized
or human.
1003461 In some embodiments, an immuno-oncology agent is (i) an agonist of a
stimulatory
(including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory
(including a co-
inhibitory) signal on T cells, both of which result in amplifying antigen-
specific T cell responses.
1003471 Certain of the stimulatory and inhibitory molecules are members of the

immunoglobulin super family (IgSF). One important family of membrane-bound
ligands that bind
to co-stimulatory or co-inhibitory receptors is the B7 family, which includes
B7-1, B7-2, B7-H1
(PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-
H6.
Another family of membrane bound ligands that bind to co-stimulatory or co-
inhibitory receptors
is the TNF family of molecules that bind to cognate TNF receptor family
members, which includes
109
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
CD40 and CD4OL, OX-40, OX-40L, CD70, CD27L, CD30, CD3OL, 4-1BBL, CD137 (4-
1BB),
TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL,
TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTr3R, LIGHT,
DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1,
Lymphotoxin cc/TNE13, TNFR2, INFoc, LTI3R, Lymphotoxin al132, FAS, FASL, RELT,
DR6,
TROY, NGFR.
[00348] In some embodiments, an immuno-oncology agent is a cytokine that
inhibits T cell
activation (e.g., IL-6, IL-10, TGF-13, VEGF, and other immunosuppressive
cytokines) or a cytokine
that stimulates T cell activation, for stimulating an immune response.
[00349] In some embodiments, a combination of a compound of the invention and
an immuno-
oncology agent can stimulate T cell responses. In some embodiments, an immuno-
oncology agent
is: (i) an antagonist of a protein that inhibits T cell activation (e.g.,
immune checkpoint inhibitors)
such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA,
CD69,
Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1,
and TIM-
4; or (ii) an agonist of a protein that stimulates T cell activation such as
B7-1, B7-2, CD28, 4-1BB
(CD137), 4-1BBL, ICOS, ICOS-L, 0X40, OX4OL, GITR, GITRL, CD70, CD27, CD40, DR3
and
CD28H.
[00350] In some embodiments, an immuno-oncology agent is an antagonist of
inhibitory
receptors on NK cells or an agonist of activating receptors on NK cells. In
some embodiments, an
immuno-oncology agent is an antagonist of KIR, such as lirilumab.
[00351] In some embodiments, an immuno-oncology agent is an agent that
inhibits or depletes
macrophages or monocytes, including but not limited to CSF-1R antagonists such
as CSF-1R
antagonist antibodies including RG7155 (W011/70024, W011/107553, W011/131407,
W013/87699, W013/119716, W013/132044) or FPA-008 (W011/140249; W013169264;
W014/036357).
[00352] In some embodiments, an immuno-oncology agent is selected from
agonistic agents
that ligate positive costimulatory receptors, blocking agents that attenuate
signaling through
inhibitory receptors, antagonists, and one or more agents that increase
systemically the frequency
of anti-tumor T cells, agents that overcome distinct immune suppressive
pathways within the
tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-
Ll/PD-1
interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal
antibody (e.g.,
110
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes
such as IDO, or
reverse/prevent T cell energy or exhaustion) and agents that trigger innate
immune activation
and/or inflammation at tumor sites.
1003531 In some embodiments, an immuno-oncology agent is a CTLA-4 antagonist.
In some
embodiments, a CTLA-4 antagonist is an antagonistic CTLA-4 antibody. In some
embodiments,
an antagonistic CTLA-4 antibody is YERVOY (ipilimumab) or tremelimumab.
1003541 In some embodiments, an immuno-oncology agent is a PD-1 antagonist. In
some
embodiments, a PD-1 antagonist is administered by infusion. In some
embodiments, an immuno-
oncology agent is an antibody or an antigen-binding portion thereof that binds
specifically to a
Programmed Death-1 (PD-1) receptor and inhibits PD-1 activity. In some
embodiments, a PD-1
antagonist is an antagonistic PD-1 antibody. In some embodiments, an
antagonistic PD-1 antibody
is OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514,
W02012/145493). In some embodiments, an immuno-oncology agent may be
pidilizumab (CT-
011). In some embodiments, an immuno-oncology agent is a recombinant protein
composed of
the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgGl,
called AMP-224.
1003551 In some embodiments, an immuno-oncology agent is a PD-Li antagonist.
In some
embodiments, a PD-L1 antagonist is an antagonistic PD-Li antibody. In some
embodiments, a
PD-Li antibody is MPDL3280A (RG7446; W02010/077634), durvalumab (MEDI4736),
BMS-
936559 (W02007/005874), and MSB0010718C (W02013/79174).
1003561 In some embodiments, an immuno-oncology agent is a LAG-3 antagonist.
In some
embodiments, a LAG-3 antagonist is an antagonistic LAG-3 antibody. In some
embodiments, a
LAG3 antibody is BMS-986016 (W010/19570, W014/08218), or IMP-731 or IMP-321
(W008/132601, W0009/44273).
1003571 In some embodiments, an immuno-oncology agent is a CD137 (4-1BB)
agonist. In
some embodiments, a CD137 (4-1BB) agonist is an agonistic CD137 antibody. In
some
embodiments, a CD137 antibody is urelumab or PF-05082566 (W012/32433).
1003581 In some embodiments, an immuno-oncology agent is a GITR agonist. In
some
embodiments, a GITR agonist is an agonistic GITR antibody. In some
embodiments, a GITR
antibody is BMS-986153, BMS-986156, TRX-518 (W0006/105021, W0009/009116), or
MK-
4166 (W011/028683).
111
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00359] In some embodiments, an immuno-oncology agent is an indoleamine (2,3)-
dioxygenase (IDO) antagonist. In some embodiments, an IDO antagonist is
selected from
epacadostat (INCB024360, Incyte); indoximod (NLG-8189, NewLink Genetics
Corporation);
capmanitib (INC280, Novartis); GDC-0919 (Genentech/Roche); PF-06840003
(Pfizer);
BMS:F001287 (Bristol-Myers Squibb); Phy906/KDI08 (Phytoceutica); an enzyme
that breaks
down kynurenine (Kynase, Ikena Oncology, formerly known as Kyn Therapeutics);
and NLG-919
(W009/73620, W0009/1156652, W011/56652, W012/142237).
[00360] In some embodiments, an immuno-oncology agent is an 0X40 agonist. In
some
embodiments, an 0X40 agonist is an agonistic 0X40 antibody. In some
embodiments, an 0X40
antibody is MEDI-6383 or MEDI-6469.
[00361] In some embodiments, an immuno-oncology agent is an OX4OL antagonist.
In some
embodiments, an OX4OL antagonist is an antagonistic 0X40 antibody. In some
embodiments, an
OX4OL antagonist is RG-7888 (W006/029879).
[00362] In some embodiments, an immuno-oncology agent is a CD40 agonist. In
some
embodiments, a CD40 agonist is an agonistic CD40 antibody. In some
embodiments, an immuno-
oncology agent is a CD40 antagonist. In some embodiments, a CD40 antagonist is
an antagonistic
CD40 antibody. In some embodiments, a CD40 antibody is lucatumumab or
dacetuzumab.
[00363] In some embodiments, an immuno-oncology agent is a CD27 agonist. In
some
embodiments, a CD27 agonist is an agonistic CD27 antibody. In some
embodiments, a CD27
antibody is varlilumab.
[00364] In some embodiments, an immuno-oncology agent is MGA271 (to B7H3)
(W011/109400).
1003651 In some embodiments, an immuno-oncology agent is abagovomab,
adecatumumab,
afutuzumab, al emtuzumab, an atum omab mafenatox, apoli zumab, atezolimab,
avel um ab ,
blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab,
indoximod,
inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab,
MED14736,
MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab,
pembrolizumab, pidilizumab, rituximab, ticilimumab, samalizumab, or
tremelimumab.
[00366] In some embodiments, an immuno-oncology agent is an immunostimulatory
agent. For
example, antibodies blocking the PD-1 and PD-Li inhibitory axis can unleash
activated tumor-
reactive T cells and have been shown in clinical trials to induce durable anti-
tumor responses in
112
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
increasing numbers of tumor histologies, including some tumor types that
conventionally have not
been considered immunotherapy sensitive. See, e.g., Okazaki, T. et at. (2013)
Nat. Immunol. 14,
1212-1218; Zou et al. (2016) Sci. Transl. Med. 8. The anti-PD-1 antibody
nivolumab (OPDIV0 ,
Bristol-Myers Squibb, also known as ONO-4538, MDX1106 and BMS-936558), has
shown
potential to improve the overall survival in patients with RCC who had
experienced disease
progression during or after prior anti-angiogenic therapy.
[00367] In some embodiments, the immunomodulatory therapeutic specifically
induces
apoptosis of tumor cells. Approved immunomodulatory therapeutics which may be
used in the
present invention include pomali domi de (POMALYST , Celgene); lenali domi de
(REVLIMID ,
Celgene); ingenol mebutate (PICATO , LEO Pharma).
[00368] In some embodiments, an immuno-oncology agent is a cancer vaccine. In
some
embodiments, the cancer vaccine is selected from sipuleucel-T (PROVENGE ,
DendreonNaleant Pharmaceuticals), which has been approved for treatment of
asymptomatic, or
minimally symptomatic metastatic castrate-resistant (hormone-refractory)
prostate cancer; and
talimogene laherparepvec (EVILYGIC , BioVex/Amgen, previously known as T-VEC),
a
genetically modified oncolytic viral therapy approved for treatment of
unresectable cutaneous,
subcutaneous and nodal lesions in melanoma. In some embodiments, an immuno-
oncology agent
is selected from an oncolytic viral therapy such as pexastimogene devacirepvec
(PexaVec/JX-594,
SillaJen/formerly Jennerex Biotherapeutics), a thymidine kinase- (TK-)
deficient vaccinia virus
engineered to express GM-CSF, for hepatocellular carcinoma (NCT02562755) and
melanoma
(NCT00429312); pelareorep (REOLYSIN , Oncolytics Biotech), a variant of
respiratory enteric
orphan virus (reovirus) which does not replicate in cells that are not RAS-
activated, in numerous
cancers, including colorectal cancer (NCT01622543); prostate cancer
(NCT01619813); head and
neck squamous cell cancer (NCT01166542); pancreatic adenocarcinoma
(NCT00998322); and
non-small cell lung cancer (NSCLC) (NCT 00861627), enadenotucirev (NG-348,
PsiOxus,
formerly known as ColoAd1), an adenovirus engineered to express a full length
CD80 and an
antibody fragment specific for the T-cell receptor CD3 protein, in ovarian
cancer (NCT02028117),
metastatic or advanced epithelial tumors such as in colorectal cancer, bladder
cancer, head and
neck squamous cell carcinoma and salivary gland cancer (NC102636036); ONCOS-
102
(Targovax/formerly Oncos), an adenovirus engineered to express GM-C SF, in
melanoma
(NCT03003676); and peritoneal disease, colorectal cancer or ovarian cancer
(NCT02963831); GL-
113
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
ONC1 (GLV-1h68/GLV-1h153, Genelux GmbH), vaccinia viruses engineered to
express beta-
galactosidase (beta-gal)/beta-glucoronidase or beta-gal/human sodium iodide
symporter (hNIS),
respectively, were studied in peritoneal carcinomatosis (NCT01443260);
fallopian tube cancer,
ovarian cancer (NCT 02759588); or CG0070 (Cold Genesys), an adenovirus
engineered to express
GM-CSF, in bladder cancer (NCT02365818).
[00369] In some embodiments, an immuno-oncology agent is selected from JX-929
(SillaJen/formerly Jennerex Biotherapeutics), a TK- and vaccinia growth factor-
deficient vaccinia
virus engineered to express cytosine deaminase, which is able to convert the
prodrug 5-
fluorocytosi ne to the cytotoxic drug 5-fluorouracil; TGO1 and T002
(Targovax/formerly Oncos),
peptide-based immunotherapy agents targeted for difficult-to-treat RAS
mutations; and TILT-123
(TILT Biotherapeutics), an engineered adenovirus designated: Ad5/3-E2F-de1ta24-
hTNFa-IRES-
hIL20; and VSV-GP (ViraTherapeutics) a vesicular stomatitis virus (VSV)
engineered to express
the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), which can
be further
engineered to express antigens designed to raise an antigen-specific CDS+ T
cell response.
1003701 In some embodiments, an immuno-oncology agent is a T-cell engineered
to express a
chimeric antigen receptor, or CAR. The T-cells engineered to express such
chimeric antigen
receptor are referred to as a CAR-T cells.
[00371] CARs have been constructed that consist of binding domains, which may
be derived
from natural ligands, single chain variable fragments (scFv) derived from
monoclonal antibodies
specific for cell-surface antigens, fused to endodomains that are the
functional end of the T-cell
receptor (TCR), such as the CD3-zeta signaling domain from TCRs, which is
capable of generating
an activation signal in T lymphocytes. Upon antigen binding, such CARs link to
endogenous
signaling pathways in the effector cell and generate activating signals
similar to those initiated by
the TCR complex.
[00372] For example, in some embodiments the CAR-T cell is one of those
described in U.S.
Patent 8,906,682 (June et al.; hereby incorporated by reference in its
entirety), which discloses
CAR-T cells engineered to comprise an extracellular domain having an antigen
binding domain
(such as a domain that binds to CD19), fused to an intracellular signaling
domain of the T cell
antigen receptor complex zeta chain (such as CD3 zeta). When expressed in the
T cell, the CAR
is able to redirect antigen recognition based on the antigen binding
specificity. In the case of
CD19, the antigen is expressed on malignant B cells. Over 200 clinical trials
are currently in
114
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
progress employing CAR-T in a wide range of
indications.
[https ://clini caltri al s . gov/ct2/re sults?term= chim eri c+anti
gen+receptors& pg=l] .
1003731 In some embodiments, an immunostimulatory agent is an activator of
retinoic acid
receptor-related orphan receptor y (RORyt). RORyt is a transcription factor
with key roles in the
differentiation and maintenance of Type 17 effector subsets of CD4+ (Th17) and
CD8+ (Tc17) T
cells, as well as the differentiation of IL-17 expressing innate immune cell
subpopulations such as
NK cells. In some embodiments, an activator of RORyt is LYC-55716 (Lycera),
which is currently
being evaluated in clinical trials for the treatment of solid tumors
(NCT02929862).
1003741 In some embodiments, an immunostimulatory agent is an agonist or
activator of a toll-
like receptor (TLR). Suitable activators of TLRs include an agonist or
activator of TLR9 such as
SD-101 (Dynavax). SD-101 is an immunostimulatory CpG which is being studied
for B-cell,
follicular and other lymphomas (NCT02254772). Agonists or activators of TLR8
which may be
used in the present invention include motolimod (VTX-2337, VentiRx
Pharmaceuticals) which is
being studied for squamous cell cancer of the head and neck (NCT02124850) and
ovarian cancer
(NCT02431559).
1003751 Other immuno-oncology agents that can be used in the present invention
include
urelumab (BMS-663513, Bristol-Myers Squibb), an anti-CD137 monoclonal
antibody; varlilumab
(CDX-1127, Celldex Therapeutics), an anti-CD27 monoclonal antibody; BMS-986178
(Bristol-
Myers Squibb), an anti-0X40 monoclonal antibody; lirilumab (IPH2102/BMS-
986015, Innate
Pharma, Bristol-Myers Squibb), an anti-KIR monoclonal antibody; monalizumab
(IPH2201,
Innate Pharma, AstraZeneca) an anti-NKG2A monoclonal antibody; andecaliximab
(GS-5745,
Gilead Sciences), an anti-MNIP9 antibody; MK-4166 (Merck & Co.), an anti-GITR
monoclonal
antibody.
1003761 In some embodiments, an immunostimulatory agent is selected from
elotuzumab,
mifamurti de, an agonist or activator of a toll-like receptor, and an
activator of RORyt
1003771 In some embodiments, an immunostimulatory therapeutic is recombinant
human
interleukin 15 (rhIL-15). rhIL-15 has been tested in the clinic as a therapy
for melanoma and renal
cell carcinoma (NCT01021059 and NCT01369888) and leukemias (NCT02689453). In
some
embodiments, an immunostimulatory agent is recombinant human interleukin 12
(rhIL-12). In
some embodiments, an IL-15 based immunotherapeutic is heterodimeric IL-15
(hetIL-15,
Novartis/Admune), a fusion complex composed of a synthetic form of endogenous
IL-15
115
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
complexed to the soluble IL-15 binding protein IL-15 receptor alpha chain
(IL15:sIL-15RA),
which has been tested in Phase 1 clinical trials for melanoma, renal cell
carcinoma, non-small cell
lung cancer and head and neck squamous cell carcinoma (NCT02452268). In some
embodiments,
a recombinant human inter] eukin 12 (rhIL-12) is NM-IL-12 (Neumedi ci n es,
Inc.), NCT02544724,
or NCT02542124.
[00378] In some embodiments, an immuno-oncology agent is selected from those
descripted in
Jerry L. Adams et at., "Big opportunities for small molecules in immuno-
oncology," Cancer
Therapy 2015, Vol. 14, pages 603-622, the content of which is incorporated
herein by reference in
its entirety. In some embodiments, an immuno-oncology agent is selected from
the examples
described in Table 1 of Jerry L. Adams et at. In some embodiments, an immuno-
oncology agent
is a small molecule targeting an immuno-oncology target selected from those
listed in Table 2 of
Jerry L. Adams et al. In some embodiments, an immuno-oncology agent is a small
molecule agent
selected from those listed in Table 2 of Jerry L. Adams et at.
[00379] In some embodiments, an immuno-oncology agent is selected from the
small molecule
immuno-oncology agents described in Peter L. Toogood, "Small molecule immuno-
oncology
therapeutic agents," Bioorganic & Medicinal Chemistry Letters 2018, Vol. 28,
pages 319-329, the
content of which is incorporated herein by reference in its entirety. In some
embodiments, an
immuno-oncology agent is an agent targeting the pathways as described in Peter
L. Toogood.
1003801 In some embodiments, an immuno-oncology agent is selected from those
described in
Sandra L. Ross et al., "Bispecific T cell engager (BITER ) antibody constructs
can mediate
bystander tumor cell killing", PLoS ONE 12(8): 00183390, the content of which
is incorporated
herein by reference in its entirety. In some embodiments, an immuno-oncology
agent is a
bispecific T cell engager (BITER) antibody construct. In some embodiments, a
bispecific T cell
engager (BITER) antibody construct is a CD19/CD3 bispecific antibody
construct. In some
embodiments, a bispecific T cell engager (BITER) antibody construct is an
EGFR/CD3 bispecific
antibody construct. In some embodiments, a bispecific T cell engager (BITER)
antibody construct
activates T cells. In some embodiments, a bispecific T cell engager (BITER)
antibody construct
activates T cells, which release cytokines inducing upregulation of
intercellular adhesion molecule
1 (ICAM-1) and FAS on bystander cells. In some embodiments, a bispecific T
cell engager
(BITER) antibody construct activates T cells which result in induced bystander
cell lysis. In some
embodiments, the bystander cells are in solid tumors. In some embodiments, the
bystander cells
116
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
being lysed are in proximity to the BITE -activated T cells. In some
embodiment, the bystander
cells comprises tumor-associated antigen (TAA) negative cancer cells. In some
embodiment, the
bystander cells comprise EGFR-negative cancer cells. In some embodiments, an
immuno-
oncology agent is an antibody which blocks the PD-L 1 /PD1 axis and/or CTLA4.
In some
embodiments, an immuno-oncology agent is an ex vivo expanded tumor-
infiltrating T cell. In
some embodiments, an immuno-oncology agent is a bispecific antibody construct
or chimeric
antigen receptors (CARs) that directly connect T cells with tumor-associated
surface antigens
(TAAs).
Exemplary Immune Checkpoint Inhibitors
[00381] In some embodiments, an immuno-oncology agent is an immune checkpoint
inhibitor
as described herein.
[00382] The term "checkpoint inhibitor" as used herein relates to agents
useful in preventing
cancer cells from avoiding the immune system of the patient. One of the major
mechanisms of
anti-tumor immunity subversion is known as "T-cell exhaustion," which results
from chronic
exposure to antigens that has led to up-regulation of inhibitory receptors.
These inhibitory
receptors serve as immune checkpoints in order to prevent uncontrolled immune
reactions.
1003831 PD-1 and co-inhibitory receptors such as cytotoxic T-lymphocyte
antigen 4 (CTLA-4,
B and T Lymphocyte Attenuator (BTLA; CD272), T cell Immunoglobulin and Mucin
domain-3
(Tim-3), Lymphocyte Activation Gene-3 (Lag-3; CD223), and others are often
referred to as a
checkpoint regulators. They act as molecular "gatekeepers" that allow
extracellular information
to dictate whether cell cycle progression and other intracellular signaling
processes should
proceed.
1003841 In some embodiments, an immune checkpoint inhibitor is an antibody to
PD-1. PD-1
binds to the programmed cell death 1 receptor (PD-1) to prevent the receptor
from binding to the
inhibitory ligand PDL-1, thus overriding the ability of tumors to suppress the
host anti-tumor
immune response.
[00385] In one aspect, the checkpoint inhibitor is a biologic
therapeutic or a small molecule. In
another aspect, the checkpoint inhibitor is a monoclonal antibody, a humanized
antibody, a fully
human antibody, a fusion protein or a combination thereof. In a further
aspect, the checkpoint
inhibitor inhibits a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1,
B7-H3, B7-H4,
117
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
BTLA, HVEM, TI1\43, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1,
CHK2,
A2aR, B-7 family ligands or a combination thereof. In an additional aspect,
the checkpoint
inhibitor interacts with a ligand of a checkpoint protein selected from CTLA-
4, PDL1, PDL2, PD1,
B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-
15049,
CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof In an aspect,
the checkpoint
inhibitor is an immunostimulatory agent, a T cell growth factor, an
interleukin, an antibody, a
vaccine or a combination thereof In a further aspect, the interleukin is IL-7
or IL-15. In a specific
aspect, the interleukin is glycosylated IL-7. In an additional aspect, the
vaccine is a dendritic cell
(DC) vaccine.
1003861 Checkpoint inhibitors include any agent that blocks or
inhibits in a statistically
significant manner, the inhibitory pathways of the immune system. Such
inhibitors may include
small molecule inhibitors or may include antibodies, or antigen binding
fragments thereof, that
bind to and block or inhibit immune checkpoint receptors or antibodies that
bind to and block or
inhibit immune checkpoint receptor ligands. Illustrative checkpoint molecules
that can be targeted
for blocking or inhibition include, but are not limited to, CTLA-4, PDL1,
PDL2, PD1, B7-H3, B7-
H4, BTLA, HVEM, GAL9, LAG3, TIIV13, VISTA, KIR, 2B4 (belongs to the CD2 family
of
molecules and is expressed on all NK, 76, and memory CD8+ (4) T cells), CD160
(also referred
to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, and various B-7 family
ligands.
B7 family ligands include, but are not limited to, B7- 1, B7-2, B7-DC, B7-H1,
B7-H2, B7-H3, B7-
H4, B7-H5, B7-H6 and B7-H7. Checkpoint inhibitors include antibodies, or
antigen binding
fragments thereof, other binding proteins, biologic therapeutics, or small
molecules, that bind to
and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1,
BTLA, HVEM,
T11V13, GAL9, LAG3, VISTA, KIR, 2B4, CD 160 and CGEN-15049. Illustrative
immune
checkpoint inhibitors include, but are not limited to, Tremelimumab (CTLA-4
blocking antibody),
anti-0X40, PD-Li monoclonal Antibody (Anti-B7-H1, MEDI4736), MK-3475 (PD-1
blocker),
Nivolumab (anti-PD1 antibody), CT-011 (anti-PD1 antibody), B Y55 monoclonal
antibody,
AlVIP224 (anti-PDL1 antibody), BMS- 936559 (anti-PDL1 antibody), MPLDL3280A
(anti-PDL1
antibody), MSB0010718C (anti-PDL1 antibody), and ipilimumab (anti-CTLA-4
checkpoint
inhibitor). Checkpoint protein ligands include, but are not limited to PD-L1,
PD-L2, B7-H3, B7-
H4, CD28, CD86 and TIM-3.
118
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1003871 In certain embodiments, the immune checkpoint inhibitor is selected
from a PD-1
antagonist, a PD-Li antagonist, and a CTLA-4 antagonist. In some embodiments,
the checkpoint
inhibitor is selected from the group consisting of nivolumab (
1003881 OPDIVOR), ipilimumab (YERVOYR), and pembrolizumab (K EYTRUD AR). In
some embodiments, the checkpoint inhibitor is selected from nivolumab (anti-PD-
1 antibody,
OPDIV00, Bristol-Myers Squibb); pembrolizumab (anti-PD-1 antibody, KEYTRUDA ,
Merck); ipilimumab (anti-CTLA-4 antibody, YERVOY , Bristol-Myers Squibb);
durvalumab
(anti-PD-Li antibody, IMFINZI , AstraZeneca); and atezolizumab (anti-PD-Li
antibody,
TECENTRIQR, Genentech).
1003891 In some embodiments, the checkpoint inhibitor is selected from the
group consisting
of lambrolizumab (MK-3475), nivolumab (BMS-936558), pidilizumab (CT-011), AMP-
224,
MDX-1105, MEDI4736, MPDL3280A, BMS-936559, ipilimumab, lirlumab, IPH2101,
pembrolizumab (KEYTRUDA ), and tremelimumab.
1003901 In some embodiments, an immune checkpoint inhibitor is REGN2810
(Regeneron), an
anti-PD-1 antibody tested in patients with basal cell carcinoma (NCT03132636);
NSCLC
(NCT03088540); cutaneous squamous cell carcinoma (NCT02760498); lymphoma
(NCT02651662); and melanoma (NCT03002376); pidilizumab (CureTech), also known
as CT-
011, an antibody that binds to PD-1, in clinical trials for diffuse large B-
cell lymphoma and
multiple myeloma; avelumab (BAVENCIO , Pfizer/Merck KGaA), also known as
MSB0010718C), a fully human IgG1 anti-PD-Li antibody, in clinical trials for
non-small cell lung
cancer, Merkel cell carcinoma, mesothelioma, solid tumors, renal cancer,
ovarian cancer, bladder
cancer, head and neck cancer, and gastric cancer; or PDR001 (Novartis), an
inhibitory antibody
that binds to PD-1, in clinical trials for non-small cell lung cancer,
melanoma, triple negative breast
cancer and advanced or metastatic solid tumors. Tremelimumab (CP-675,206;
Astrazeneca) is a
fully human monoclonal antibody against CTLA-4 that has been in studied in
clinical trials for a
number of indications, including. mesothelioma, colorectal cancer, kidney
cancer, breast cancer,
lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma,
pancreatic cancer,
germ cell cancer, squamous cell cancer of the head and neck, hepatocellular
carcinoma, prostate
cancer, endometrial cancer, metastatic cancer in the liver, liver cancer,
large B-cell lymphoma,
ovarian cancer, cervical cancer, metastatic anaplastic thyroid cancer,
urothelial cancer, fallopian
tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and
melanoma. AGEN-1884
119
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
(Agenus) is an anti-CTLA4 antibody that is being studied in Phase 1 clinical
trials for advanced
solid tumors (NCT02694822).
1003911 In some embodiments, a checkpoint inhibitor is an inhibitor of T-cell
immunoglobulin
mucin containing protein-3 (TIM-3). TIM-3 inhibitors that may be used in the
present invention
include TSR-022, LY3321367 and MBG453. TSR-022 (Tesaro) is an anti-TIM-3
antibody which
is being studied in solid tumors (NCT02817633). LY3321367 (Eli Lilly) is an
anti-TIM-3
antibody which is being studied in solid tumors (NCT03099109). MBG453
(Noyartis) is an anti-
TIM-3 antibody which is being studied in advanced malignancies (NC102608268).
1003921 In some embodiments, a checkpoint inhibitor is an inhibitor of T cell
immunoreceptor
with Ig and ITTM domains, or TIGIT, an immune receptor on certain T cells and
NK cells TIGIT
inhibitors that may be used in the present invention include BMS-986207
(Bristol-Myers Squibb),
an anti-TIGIT monoclonal antibody (NCT02913313), OMP-313M32 (Oncomed), and
anti-TIGIT
monoclonal antibody (NCT03119428).
1003931 In some embodiments, a checkpoint inhibitor is an inhibitor of
Lymphocyte Activation
Gene-3 (LAG-3). LAG-3 inhibitors that may be used in the present invention
include BMS-
986016 and REGN3767 and IMP321. BMS-986016 (Bristol-Myers Squibb), an anti-LAG-
3
antibody, is being studied in glioblastoma and gliosarcoma (NCT02658981).
REGN3767
(Regeneron), is also an anti-LAG-3 antibody, and is being studied in
malignancies
(NCT03005782). IMP321 (Immutep S.A.) is an LAG-3-Ig fusion protein, being
studied in
melanoma (NCT02676869); adenocarcinoma (NCT02614833); and metastatic breast
cancer
(NC 100349934).
1003941 Checkpoint inhibitors that may be used in the present invention
include 0X40 agonists.
0X40 agonists that are being studied in clinical trials include PF-04518600/PF-
8600 (Pfizer), an
agonistic anti-0X40 antibody, in metastatic kidney cancer (NCT03092856) and
advanced cancers
and neoplasms (NCT02554812, NCT05082566), GSK3174998 (Merck), an agonistic
anti-0X40
antibody, in Phase 1 cancer trials (NC102528357), MED10562
(Medimmune/AstraZeneca), an
agonistic anti-0X40 antibody, in advanced solid tumors (NCT02318394 and
NCT02705482),
MEDI6469, an agonistic anti-0X40 antibody (Medimmune/AstraZeneca), in patients
with
colorectal cancer (NC102559024), breast cancer (NC101862900), head and neck
cancer
(NC102274155) and metastatic prostate cancer (NCT01303705); and BMS-986178
(Bristol-
Myers Squibb) an agonistic anti-0X40 antibody, in advanced cancers
(NCT02737475).
120
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00395] Checkpoint inhibitors that may be used in the present invention
include CD137 (also
called 4-1BB) agonists. CD137 agonists that are being studied in clinical
trials include
utomilumab (PF-05082566, Pfizer) an agonistic anti-CD137 antibody, in diffuse
large B-cell
lymphoma (NCT02951156) and in advanced cancers and neoplasms (NCT02554812 and
NCT05082566); urelumab (BMS-663513, Bristol-Myers Squibb), an agonistic anti-
CD137
antibody, in melanoma and skin cancer (NCT02652455) and glioblastoma and
gliosarcoma
(NC102658981); and CTX-471 (Compass Therapeutics), an agonistic anti-CD137
antibody in
metastatic or locally advanced malignancies (NCT03881488).
[00396] Checkpoint inhibitors that may be used in the present invention
include CD27 agonists
CD27 agonists that are being studied in clinical trials include varlilumab
(CDX-1127, Celldex
Therapeutics) an agonistic anti-CD27 antibody, in squamous cell head and neck
cancer, ovarian
carcinoma, colorectal cancer, renal cell cancer, and glioblastoma
(NCT02335918), lymphomas
(NCT01460134); and glioma and astrocytoma (NCT02924038).
[00397] Checkpoint inhibitors that may be used in the present invention
include glucocorticoid-
induced tumor necrosis factor receptor (GITR) agonists. GITR agonists that are
being studied in
clinical trials include TRX518 (Leap Therapeutics), an agonistic anti-GITR
antibody, in malignant
melanoma and other malignant solid tumors (NCT01239134 and NCT02628574);
GWN323
(Novartis), an agonistic anti-GITR antibody, in solid tumors and lymphoma (NCT
02740270);
INCAGN01876 (Incyte/Agenus), an agonistic anti-GITR antibody, in advanced
cancers
(NCT02697591 and NCT03126110); MK-4166 (Merck), an agonistic anti -GITR
antibody, in solid
tumors (NCT02132754) and 1VIEDI1873 (Medimmune/AstraZeneca), an agonistic
hexameric
GITR-ligand molecule with a human IgG1 Fc domain, in advanced solid tumors
(NCT02583165).
1003981 Checkpoint inhibitors that may be used in the present invention
include inducible T-
cell co-stimulator (ICOS, also known as CD278) agonists ICOS agonists that are
being studied
in clinical trials include MEDI-570 (Medimmune), an agonistic anti-ICOS
antibody, in lymphomas
(NC102520791), GSK3359609 (Merck), an agonistic anti-ICOS antibody, in Phase 1

(NCT02723955), JTX-2011 (Jounce Therapeutics), an agonistic anti-ICOS
antibody, in Phase 1
(NCT02904226).
[00399] Checkpoint inhibitors that may be used in the present invention
include killer IgG-like
receptor (KIR) inhibitors. KIR inhibitors that are being studied in clinical
trials include lirilumab
(IPH2102/BMS-986015, Innate Pharma/Bristol-Myers Squibb), an anti-KIR
antibody, in
121
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
leukemias (NCT01687387, NCT02399917, NCT02481297, NCT02599649), multiple
myeloma
(NCT02252263), and lymphoma (NCT01592370); IPH2101 (1-7F9, Innate Pharma) in
myeloma
(NCT01222286 and NCT01217203); and IPH4102 (Innate Pharma), an anti-KIR
antibody that
binds to three domains of the long cytoplasmic tail (KIR3DL2), in lymphoma
(NCT02593045).
1004001 Checkpoint inhibitors that may be used in the present invention
include CD47
inhibitors of interaction between CD47 and signal regulatory protein alpha
(SlRPa). CD47/SlRPa
inhibitors that are being studied in clinical trials include ALX-148 (Alexo
Therapeutics), an
antagonistic variant of (SIRPa) that binds to CD47 and prevents CD47/SIRPa-
mediated signaling,
in phase 1 (NCT03013218); TTI-621 (SIRPa-Fc, Trillium Therapeutics), a soluble
recombinant
fusion protein created by linking the N-terminal CD47-binding domain of SlRPa
with the Fc
domain of human IgGl, acts by binding human CD47, and preventing it from
delivering its "do
not eat" signal to macrophages, is in clinical trials in Phase 1 (NCT02890368
and NCT02663518),
CC-90002 (Celgene), an anti-CD47 antibody, in leukemias (NCT02641002); and
Hu5F9-G4
(Forty Seven, Inc.), in colorectal neoplasms and solid tumors (NCT02953782),
acute myeloid
leukemia (NCT02678338) and lymphoma (NCT02953509).
1004011 Checkpoint inhibitors that may be used in the present invention
include CD73
inhibitors. CD73 inhibitors that are being studied in clinical trials include
MEDI9447
(Medimmune), an anti-CD73 antibody, in solid tumors (NCT02503774); and BMS-
986179
(Bristol-Myers Squibb), an anti-CD73 antibody, in solid tumors (NCT02754141).
1004021 Checkpoint inhibitors that may be used in the present
invention include agonists of
stimulator of interferon genes protein (STING, also known as transmembrane
protein 173, or
TMEM173). Agonists of STING that are being studied in clinical trials include
1'VIK-1454
(Merck), an agonistic synthetic cyclic dinucleotide, in lymphoma
(NCT03010176); and ADU-
S100 (MIW815, Aduro Biotech/Novartis), an agonistic synthetic cyclic
dinucleotide, in Phase 1
(NCT02675439 and NCT03172936)
1004031 Checkpoint inhibitors that may be used in the present invention
include CSF1R
inhibitors. CSF1R inhibitors that are being studied in clinical trials include
pexidartinib
(PLX3397, Plexxikon), a CSF1R small molecule inhibitor, in colorectal cancer,
pancreatic cancer,
metastatic and advanced cancers (NCT02777710) and melanoma, non-small cell
lung cancer,
squamous cell head and neck cancer, gastrointestinal stromal tumor (GIST) and
ovarian cancer
(NCT02452424); and IMC-CS4 (LY3022855, Lilly), an anti-CSF-1R antibody, in
pancreatic
122
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
cancer (NCT03153410), melanoma (NCT03101254), and solid tumors (NCT02718911);
and
BLZ945 (4-[2((1R,2R)-2-hydroxycyclohexylamino)-benzothiazol-6-
yloxyl]-pyridine-2-
carboxylic acid methylamide, Novartis), an orally available inhibitor of
CSF1R, in advanced solid
tumors (NCT02829723).
[00404] Checkpoint inhibitors that can be used in the present invention
include NKG2A
receptor inhibitors. NKG2A receptor inhibitors that are being studied in
clinical trials include
monalizumab (IPH2201, Innate Pharma), an anti-NKG2A antibody, in head and neck
neoplasms
(NC102643550) and chronic lymphocytic leukemia (NCT02557516).
[00405] In some embodiments, the immune checkpoint inhibitor is selected from
nivolumab,
pembrolizumab, ipilimumab, avelumab, durvalumab, atezolizumab, or pidilizumab.
EXEMPLIFICATION
[00406] The following examples are intended to illustrate the invention and
are not to be
construed as being limitations thereon. Unless otherwise stated, one or more
tautomeric forms of
compounds of the examples described hereinafter may be prepared in situ and/or
isolated. All
tautomeric forms of compounds of the examples described hereafter should be
considered to be
disclosed. Temperatures are given in degrees centigrade. If not mentioned
otherwise, all
evaporations are performed under reduced pressure, preferably between about 15
mm Hg and 100
mm Hg (= 20-133 mbar). The structure of final products, intermediates and
starting materials is
confirmed by standard analytical methods, e.g., microanalysis and
spectroscopic characteristics,
e.g., MS, JR, NMR. Abbreviations used are those conventional in the art.
[00407] All starting materials, building blocks, reagents, acids,
bases, dehydrating agents,
solvents, and catalysts utilized to synthesis the compounds of the present
invention are either
commercially available or can be produced by organic synthesis methods known
to one of
ordinary skill in the art. Further, the compounds of the present invention can
be produced by
organic synthesis methods known to one of ordinary skill in the art as shown
in the following
examples.
Example 1: Synthesis of Exemplary Compounds
[00408] Certain exemplary compounds are prepared following the following
schemes.
123
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
H
1-1K-INH
:11
LTIAHI HF 4 0
HO
r4t*FF
Et,NDCMK2C0s, KI, MeCN
HO Ts0
Step 1: 13-(Trifluoromethyl)-1-bicyclo11.1.11pentanyll methanol
HO
1004091 To a solution of LiA1H4 (84.28 mg, 2.22 mmol, 2 eq) in THE (5 mL) was
added 3-
(trifluoromethyl)bicyclo[1.1.1]pentane-1-carboxylic acid (200 mg, 1.11 mmol, 1
eq) at 0 C and
stirred at 25 C for 2 h. TLC (PE/Et0Ac = 1/1, Rf = 0.60) indicated the
starting material was
consumed completely and one new spot formed. The mixture was quenched by 10%
aq. NaOH
(0.5 mL) and the precipitated solid was filtered and the filtrate was
concentrated to yield [3-
(trifluoromethyl)-1-bicyclo[1.1.1]pentanyl]methanol (180 mg, 975.08 [tmol,
87.8% yield, 90%
purity) as colorless oil, which was used in the next step without further
purification. 1H NMR (500
MHz, CDC13) 6 ppm 3.67 (s, 2H), 1.92 (s, 6H).
Step 2: 13-(Trifluoromethyl)-1-bicyclo11.1.11pentanyllmethyl methanesulfonate
jc-FF
Ts0
1004101 To a solution of [3-(trifluoromethyl)-1-
bicyclo[1.1.1]pentanyl]methanol (180 mg,
975.08 mot, 90% purity, 1 eq) and EtiN (197.33 mg, 1.95 mmol, 271.44 [tL, 2
eq) in DCM (3
mL) was added MsC1 (170 mg, 1.48 mmol, 114.86 l_tL, 1.52 eq) dropwise at 0 C
and the mixture
stirred at 25 C for 2 h. TLC (PE/Et0Ac = 1/1, Rf = 0.67) indicated the
starting material was
consumed completely and one new spot formed. The mixture was quenched with
sal. aq.NaHCO3
(10 mL) and extracted with DCM (15 mL x 2). The combined organic phase was
dried over
anhydrous Na2SO4, filtered and concentrated to yield [3-(trifluoromethyl)-1-
bicyclo[1.1.11pentanylimethyl methanesulfonate (230 mg, 894.65 tmol, 91.8%
yield, 95.0%
purity) as a colorless gum, which was used in the next step without further
purification. 1H NMR
(400 MHz, CDC13) 6 ppm 4.25 (s, 2H), 3.02 (s, 3H), 2.02 (s, 6H).
124
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Step 3: N4(1S,5R)-3-113-(Trifluoromethyl)-1-bicyclo[1.1.11pentanyllmethyll-3-
azabicyclo[3.1.01hexan-6-Aprop-2-enamide
0
H
NFF
1004111 To a solution of N-[(1S,5R)-3-azabicyclo[3.1.0]hexan-6-
yliprop-2-enamide (120 mg,
360.61 mmol, 80% purity, 1 eq, TFA) and [3-(trifluoromethyl)-1-
bicyclo[1.1.1]pentanylimethyl
methanesulfonate (92.71 mg, 360.61 [tmol, 95% purity, 1 eq) in ACN (3 mL) were
added K2CO3
(149.52 mg, 1.08 mmol, 3 eq) and KI (5.99 mg, 36.06 mot, 0.1 eq). The mixture
was stirred at
60 C for 16 h. The solvent was removed to yield a residue which was purified
by preparative
HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (10
mMNH4HCO3)-
ACN]; B%: 48%-78%, 10min), followed by lyophilization to yield N-[(1S,5R)-34[3-

(trifluoromethyl)-1-bicyclo[1.1.1]pentanyl]methy1]-3-azabicyclo[3.1.0]hexan-6-
yl]prop-2-
enamide (18.28 mg, 60.87 timol, 16.9% yield, 100.0% purity) as a white solid.
1-H NTVIR (500
MHz, CDC13) 6 ppm 6.26 (d, J= 16.9 Hz, 1H), 6.01 (dd, J= 10.4, 16.9 Hz, 1H),
5.61 (d, J= 10.2
Hz, 1H), 5.54 (br s, 1H), 3.17 (d, J = 8.9 Hz, 2H), 3.02 (d, J = 2.0 Hz, 1H),
2.51 (s, 2H), 2.37 (d,
J= 8.4 Hz, 2H), 1.86 (s, 6H), 1.61 (s, 2H); ES-LCMS m/z 301.2 [M+H]t
1-8
H H
LAH4 C" HKINH 1
CF,
HO .(
THF HO NaBH,CN,Me0H 0
Step 1: 14-(Trifluoromethyl)-1-bieyelo12.2.21octanyllmethanol
HO
1004121 To a solution of 4-(trifluoromethyl)bicyclo[2.2.2]octane-1-
carboxylic acid (300 mg,
1.35 mmol, 1 eq) in THF (5 mL) was added LiA1H4 (102.49 mg, 2.70 mmol, 2 eq)
at 25 C. The
mixture was stirred at 25 C for 1 h. TLC (PE/Et0Ac = 3/1, Rf = 0.59)
indicated the starting
material was consumed completely and one new spot formed. The mixture was
quenched by 10%
aq. NaOH (0.5 mL) and the precipitated solid was filtered and the filtrate was
concentrated to yield
[4-(trifluoromethyl)-1-bicyclo[2.2.2]octanyl]methanol (280 mg, 1.28 mmol,
94.6% yield, 95.0%
purity) as colorless gum, which was used in the next step without further
purification. 111 NMR
125
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
(400 MHz, CDC13) 6 ppm 3.30 (s, 2H), 1.76-1.65 (m, 6H), 1.51-1.41 (m, 6H).
Step 2: 4-(Trifluoromethyl)bicyclo12.2.210ctane-1-carbaldehyde
F3
[00413] To a solution of[4-(trifluoromethyl)-1-
bicyclo[2.2.2]octanyl]methanol (50 mg, 228.12
mot, 95% purity, 1 eq) in DCM mL) was added PCC (98.35 mg, 456.25 pmol, 2 eq).
The
mixture was stirred at 25 C for 1 h. TLC (PE/Et0Ac = 3/1, Rf = 0.75)
indicated the starting
material was consumed completely and one new spot formed. The solvent was
removed to yield a
residue which was purified by flash silica gel chromatography (from PE/Et0Ac =
2/1, TLC:
PE/Et0Ac = 3/1, Rf = 0.75) to yield 4-(trifluoromethyl)bicyclo[2.2.2]octane-1-
carbaldehyde (50
mg, 223.08 mol, 97.8% yield, 92.0% purity) as yellow oil. 1-1-1 NMR (400 MHz,
CDC13) 6 ppm
9.47 (s, 1H), 1.76-1.69 (m, 12H).
Step 3: N-1(1R,5S)-3-114-(Trifluoromethyl)-1-bicyclo[2.2.21octanyllmethyl]-3-
azabicyclo[3.1.01hexan-6-Aprop-2-enamide
H H
0 K-IN
[00414] To a solution of N-[(1S,5R)-3-azabicyclo[3.1.0]hexan-6-
yl]prop-2-enamide (120 mg,
360.61 mol, 80% purity, 1 eq, TFA) and TEA (36.49 mg, 360.61 mol, 50.19 L,
1 eq) in Me0H
(10 mL) was added 4-(trifluoromethyl)bicyclo[2.2.2]octane-1-carbaldehyde
(47.43 mg, 211.62
mot, 92% purity). The mixture was stirred at 25 C for 2 h. NaBH3CN (67.98 mg,
1.08 mmol, 3
eq) was added and the mixture was stirred at 25 C for 16 h. The solvent was
removed to yield a
residue which was purified by preparative HPLC (column: Agela DuraShell C18
150*25mm*5um;mobile phase: [water(0.05% NH3H20+10 mM NE14HCO3)-ACN] ;B%: 51%-
81%,10min), followed by lyophilization to yield N-[(1R,5S)-34[4-
(trifluoromethyl)-1-
bicyclo[2.2.2]octanylimethyl]-3-azabicyclo[3.1.0]hexan-6-yl]prop-2-enamide
(24.64 mg, 71.96
mol, 20.0% yield, 100.0% purity) as a white solid. 1-1-1NMR (400 MHz, CDC13) 6
ppm 6.31-6.22
(m, 1H), 6.01 (dd, J= 10.3, 17.1 Hz, 1111), 5.61 (dd, J= 1.1, 10.1 Hz, 1H),
5.49 (br s, 1H), 3.13 (d,
J= 8.8 Hz, 2H), 3.02 (d, J= 1.7 Hz, 1H), 2.48 (d, J= 8.3 Hz, 2H), 2.13 (s,
2H), 1.68-1.59 (m, 6H),
126
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1.49 (s, 2H), 1.43-1.32 (m, 6H); ES-LCMS m/z 343.2 [M+Hr.
1-9
Br H Cr' Br Boc CF' Br I?. CF3
(Boc)20 Fe, NH4C1
N
01111
DIEA, DCM
02N 502N H2N
1
N-\\
CF, c N
CF,
N\ki N-\\
Br Boc
Sn(n-Bu), Boc CF3
TFA N140
5Lr 40
Pd(dppf)C12, DMF 410
Step 1: tert-Butyl (2-bromo-4-nitrophenyl)(4-(trifluoromethyl)benzyl)carbamate
CF3
Br Boc
41111
02N
[00415] To a solution of 2-bromo-4-nitro-N-[[4-
(trifluoromethyl)phenyl]methyl]aniline (1.10
g, 2.67 mmol, 91.1% purity, 1 eq) in DCM (10 mL) was added (Boc)20 (1.75 g,
8.00 mmol, 1.84
mL, 3 eq) and DMAP (325.66 mg, 2.67 mmol, 1 eq). The mixture was stirred at 25
C for 12 h.
The reaction mixture was concentrated to yield a residue which was purified by
flash silica gel
chromatography (from PE/Et0Ac = 1/0 to10/1, TLC: PE/Et0Ac =10/1, Rf = 0.51) to
yield tert-
butyl N-(2-bromo-4-nitro-phenyl)-N-114-
(trifluoromethyl)phenyl]methyl]carbamate (1.15 g, 2.40
mmol, 89.9% yield, 98.7% purity) as a white solid. I-H NMIR (400 MHz, CDC13) 6
ppm 8.49 (br s,
1H), 8.06 (d, J = 7.8 Hz, 1H), 7.57 (d, J = 7.4 Hz, 2H), 7.36 (d, J = 8.2 Hz,
2H), 5.20 (d, J = 14.1
Hz, 1H), 4.39 (d, J= 15.7 Hz, 1H), 1.57-1.40 (m, 9H); ES-LCMS m/z 375.3[M-
Boc+H]t
Step 2: tert-Butyl (4-amino-2-bromophenyl)(4-(trifluoromethyl)benzyl)carbam
ate
CF3
Br Boc 40,
H2N
[00416] To a solution of tert-butyl N-(2-bromo-4-nitro-pheny1)-N-[[4-
(trifluoromethyl)phenyl]methyl]carbamate (1.11 g, 2.31 mmol, 98.7%, 1 eq) in
Et0H (5 mL) and
H20 (5 mL) was added Fe (646.28 mg, 11.57 mmol, 5 eq) and NH4C1 (1.24 g, 23.15
mmol, 10 eq).
127
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
The mixture was stirred at 80 C for 1 h. The reaction mixture was quenched by
addition of water
(40 mL) and extracted with Et0Ac (30 mL x 3). The combined organic layers were
washed with
brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced
pressure to yield tert-
butyl /V-(4-am no-2-brom o-phenyl )-N--[[4-(tri fluorom ethyl )phenyl ]m
ethyl] carb am ate (1.01 g,
2.06 mmol, 89.1% yield, 91.0% purity) as a yellow solid, which was used in the
next step without
further purification. 1H NAIR (400 MHz, CDC13) 6 ppm 7.54 (d, J = 7.8 Hz, 2H),
7.39 (s, 2H),
6.95-6.88 (m, 1H), 6.58 (d, J = 8.6 Hz, 1H), 6.50-6.39 (m, 1H), 5.20 (d, J =
14.9 Hz, 1H), 4.27-
4.20 (m, 1H), 3.72 (s, 2H), 1.38 (s, 9H); ES-LCMS nilz 391.0 [M-Boc+Hr
Step 3: tert-Butyl (4-acrylamido-2-bromophenyl)(4-
(trifluoromethyl)benzyl)carbamate
Br Boc CF3
1004171 To a solution of tert-butyl /V-(4-amino-2-bromo-phenyl)-N-114-
(trifluoromethyl)phenyl]methyl]carbamate (250 mg, 539.00 [tmol, 96% purity, 1
eq) in DCM (10
mL) was added acryloyl chloride (73.18 mg, 808.49 p.mol, 65.92 L, 1.5 eq) and
DIEA (139.32
mg, 1.08 mmol, 187.77 iftL, 2 eq). The mixture was stirred at 25 C for 1 h.
The reaction mixture
was quenched by addition of water (50 mL) and extracted with DCM (50 mL x 3).
The combined
organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and
concentrated under
reduced pressure to yield a residue which was purified by flash silica gel
chromatography (from
PE/Et0Ac = 1/0 to 3/1, TLC: PE/Et0Ac = 3/1, Rf = 0.43) to yield tert-butyl N-
[2-bromo-4-(prop-
2-enoylamino)pheny1]-N-[[4-(trifluoromethyl)phenyl]methyl]carbamate (243 mg,
486.66 umol,
90.2% yield, 100% purity) as colorless oil. 1H NIVIR (400 MHz, CDC13) (5 ppm
7.60-7.53 (m, 2H),
7.40-7.29 (m, 3H), 7.26-7.18 (m, 1H), 6.92-6.75 (m, 1H), 6.46 (d, J ¨ 16.8 Hz,
1H), 6.21 (dd, J ¨
10.6, 16.8 Hz, 1H), 5.84-5.74 (m, 1H), 5.21 (d, J= 15.3 Hz, 1H), 4.30 (s, 1H),
4.26 (s, 1H), 4.13
(q, J = 7.2 Hz, 1H), 2.06 (s, 1H), 1.63-1.52 (m, 9H), 1.33-1.25 (m, 2H); ES-
LCMS rn/z 521.1
[M+H] .
Step 4: tert-Butyl (4-acrylamido-2-(1-methy1-1H-imidazol-4-yl)phenyl)(4-
(trifluoromethyl)benzyl)carbamate
128
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
\N¨\\
N C F3
Boc
NI
0
1004181 To a solution of tert-butyl N42-bromo-4-(prop-2-enoylamino)pheny1]-N-
[[4-
(trifluoromethyl)phenyl]methyl]carbamate (200 mg, 400.54 [imol, 100%, 1 eq)
and tributyl-(1-
methylimidazol-4-yl)stannane (234.73 mg, 600.82 lamol, 95%, 1.5 eq) in DMF (7
mL) was added
Pd(dppf)C12 (29.31 mg, 40.05 [tmol, 0.1 eq). The mixture was stirred under N2
atmosphere at 130
C for 1 h. The reaction mixture was quenched by addition of water (50 mL) and
extracted with
Et0Ac (50 mL x 3). The combined organic layers were washed with brine (20 mL),
dried over
Na2SO4, filtered and concentrated under reduced pressure to yield a residue
which was purified by
flash silica gel chromatography (from PE/Et0Ac = 1/0 to 4/5, TLC: PE/Et0Ac =
3/1, Rf = 0.20)
to yield tert-butyl N12-(1-methylimidazol-4-y1)-4-(prop-2-enoylamino)pheny1]-N-
[[4-
(trifluoromethyl)phenyl]methyl]carbamate (100 mg, 199.80 [tmol, 49.8% yield,
100.0% purity) as
a yellow solid. 1H NMR (400 MHz, CDC13) 6 ppm 7.86 (s, 1H), 7.55-7.43 (m, 4H),
7.34 (d, J ¨
7.8 Hz, 3H), 6.88 (br s, 1H), 6.73 (d, J = 7.0 Hz, 1H), 6.45-6.35 (m, 1H),
6.25-6.14 (m, 1H), 5.76
(d, J = 10.6 Hz, 1H), 5.18 (d, J = 14.5 Hz, 1H), 4.18-4.07 (m, 1H), 4.11 (d, J
= 13.7 Hz, 1H), 3.67
(s, 3H), 1.26 (s, 9H); ES-LCMS rniz 501.2 [M+Hr.
Step 5: N-(3-(1-Methy1-1H-imidazol-4-y1)-4-04-
(trifluoromethyl)benzyl)amino)phenyl)acrylamide
\N¨\\
N CF
1004191 To a solution of tert-butyl N42-(1-methylimidazol-4-y1)-4-(prop-2-
enoylamino)pheny1]-N-[[4-(trifluoromethyl)phenyl]methyl]carbamate (100 mg,
199.80 mol,
100%, 1 eq) in DCM (3 mL) was added TFA (3.08 g, 27.01 mmol, 2.00 mL, 135.20
eq). The
129
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
mixture was stirred at 25 C for 1 h. The reaction mixture was quenched by
addition of water (30
mL) and extracted with Et0Ac (30 mL x 3). The combined organic layers were
washed with brine
(10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure
to yield a residue
which was purified by preparative HPLC (column: Agel a DuraShell C18
150*25mm*5 m; mobile
phase: [water (0.05% NH3H20+10mM NE4EIC03)-ACN]; B%: 40%-70%, 10min), followed
by
lyophilization to yield N-[3 -(1-
methylimidazol-4-y1)-44 [4-
(trifluoromethyl)phenyl]methylamino]phenyl]prop-2-enamide (10.48 mg, 26.17
p.mol, 13.1%
yield, 100.0% purity) as a yellow solid. 1H NMR (400 MHz, CDC13) 6 ppm 8.35
(br s, 1H), 7.92
(s, 1H), 7.57-7.45 (m, 5H), 7.24 (hr s, 1H), 7.08-6.94 (m, 2H), 6.47-6.36 (m,
2H), 6.26-6.14 (m,
1H), 5.71 (d, J = 11.3 Hz, 1H), 4.53 (s, 2H), 3.74 (s, 3H); ES-LCMS nilz 401.2
[M+Hr.
I-10 and I-11 (isomers of 1-25)
Br
Bry,o,
NH
U\s N'INia,F3 pd(dppf) C-12B're,S0, R\, 141 N CF
TFA/DCM .
NaHCOr,, Et0Ac
s'N'S\s
PMB PMB H 0
1
Br Br Br
_N -N
t
SFC H
R\CF. R\ N N F'acF
101ENII2) 1,1 CF,
H A H 0
Step 1: N-[(4-Methoxyphenyl)methyll-N-methy1-4-115-(trifluoromethyl)-2-
pyridyllaminol-3-
vinyl-benzenesulfonamide
N
\\
,S CF3
N
0
PMB
[00420] To a solution of 3-bromo-N-[(4-methoxyphenyl)methy1]-N-methy1-41[5-
(trifluoromethyl)-2-pyridyl]aminoThenzenesulfonamide (600 mg, 1.13 mmol, 1 eq)
and 4,4,5,5-
tetramethy1-2-viny1-1,3,2-dioxaborolane (348.48 mg, 2.26 mmol, 383.79 pi, 2
eq) in 1,4-dioxane
(10 mL) and H20 (2 mL) was added Cs2CO3 (737.21 mg, 2.26 mmol, 2 eq) and
Pd(dppf)C12 (82.78
130
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
mg, 113.13 litmol, 0.1 eq). The mixture was stirred under N2 atmosphere at 100
C for 12 h. The
reaction mixture was quenched by addition of water (50 mL) and extracted with
Et0Ac (50 mL x
3). The combined organic layers were washed with brine (30 mL), dried over
Na2SO4, filtered and
concentrated under reduced pressure to yield a residue which was purified by
flash silica gel
chromatography (from PE/Et0Ac ¨ 1/0 to 2n, TLC: PE/Et0Ac ¨ 1/1, Rf ¨ 0.59) to
yield N-[(4-
methoxyphenyl)methyl]-N-methy1-44[5-(trifluoromethyl)-2-pyridyl] amino]-3 -
vinyl-
benzenesulfonamide (500 mg, 1.04 mmol, 91.6% yield, 98.9% purity) as a white
solid. 1H NMR
(400 MHz, CDC13) 6 ppm 8.53 (s, 1H), 7.98-7.90 (m, 2H), 7.76 (dt, J= 2.2, 8.9
Hz, 2H), 7.25 (d,
= 8.6 Hz, 2H), 6.93-6.82 (m, 5H), 5.83 (d, = 17.6 Hz, 1H), 5.58 (d, = 11.0 Hz,
1H), 4.13 (s,
2H), 3.81 (s, 3H), 2.63 (s, 3H); ES-LCMS nilz 478.6 [M+Hr.
Step 2: N-Methyl-4-115-(trifluoromethyl)-2-pyridyllaminol-3-vinyl-
benzenesulfonamide
0 N
\\ 410
,S CF3
N
H
1004211 To a solution of N-[(4-methoxyphenyl)methy1]-N-methyl-44[5-
(trifluoromethyl)-2-
pyridyl]amino]-3-vinyl-benzenesulfonamide (500 mg, 1.04 mmol, 98.9% purity, 1
eq) in DCM (2
mL) was added TFA (L52 g, 13.37 mmol, 989.70 L, 12.90 eq). The mixture was
stirred at 25 C
for 2 h. The reaction mixture was quenched by addition of water (50 mL) and
extracted with Et0Ac
(30 mL x 3). The combined organic layers were washed with brine (20 mL), dried
over Na2SO4,
filtered and concentrated under reduced pressure to yield a residue which was
purified by flash
silica gel chromatography (from PE/Et0Ac = 1/0 to 2/1, TLC: PE/Et0Ac = 2/1, Rf
= 0.49) to yield
N-methyl-4[[5-(trifluoromethyl)-2-pyridyl]amino]-3-vinyl-benzenesulfonamide
(300 mg, 805.09
litmol, 77.7% yield, 95.9% purity) as colorless oil. 1H N1VIR (400 MHz, CDC13)
6 ppm 8.50 (s, 1H),
7.97 (d, J = 2.0 Hz, 1H), 7.89 (d, J = 8.6 Hz, 1H), 7.77 (dt, J= 2.1, 8.5 Hz,
2H), 6.94 (s, 1H), 6.90-
6.80 (m, 2H), 5.83 (d, J= 17.4 Hz, 1H), 5.56 (d, J= 11.2 Hz, 1H), 4.51 (q, J=
5.1 Hz, 1H), 2.72
(d, J = 5.4 Hz, 3H); ES-LCMS m./z 358.1 [M+H]t
131
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Step 3: 3-1(5S)-3-Bromo-4,5-dihydroisoxazol-5-yll-N-methyl-4-115-
(trifluoromethyl)-2-
pyridyllaminolbenzenesulfonamide and 3-1(5R)-3-bromo-4,5-dihydroisoxazol-5-yll-
N-
methyl-4-[15-(trifluoromethyl)-2-pyridyllamino]benzenesulfonamide
Br Br
0 0
= H
0\ ih- N N
0\\
's

CF3 ,S CF3
N \µ, N
H H
1004221 To a stirred solution of N-methy1-44[5-(trifluoromethyl)-2-
pyridyliamino]-3-vinyl-
benzenesulfonamide (270 mg, 724.58 mol, 95.9%, 1 eq) and dibromomethanone
oxime (293.94
mg, 1.45 mmol, 2 eq) in Et0Ac (10 mL) was added NaHCO3 (608.70 mg, 7.25 mmol,
281.80 p.L,
eq). The reaction mixture was stirred at 25 C for 2 h. The reaction mixture
was quenched by
addition of water (50 mL) and extracted with Et0Ac (50 mL x 3). The combined
organic layers
were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated
under reduced
pressure to yield a residue which was purified by preparative HPLC (column:
Agela DuraShell
C18 150*25mm*.5p.m; mobile phase: [water (0.05% NH3.H20+10mM NH4HCO3)-ACN];
B%:
40%-70%, 10min), followed by lyophilization to yield the mixture which was
separated by chiral
SFC column: DAICEL CHIRALPAK IG (250mm*30mm, 10 m); mobile phase: [0.1%
NH3.H20
MEOH]; B%: 40%-40%) to yield Peak 1 and Peak 2. Peak 1 was concentrated under
reduced
pressure to yield a residue which was dissolved in MeCN (10 mL) and H20 (20
mL) and
lyophilized to yield 3-[(5S)-3-bromo-4,5-dihydroisoxazol-5-y1]-N-methy1-44[5-
(trifluoromethyl)-
2-pyridyl]aminoThenzenesulfonamide (44.51 mg, 92.87 mol, 12.8% yield, 100.0%
purity, SFC:
R1= 1.658, ee = 100%, [a]3'4D _44.4 (CH3OH, c = 0.054 g/100 mL)) as a yellow
solid. 1-E1 NMR
(400 MHz, CDC13) 6 ppm 8.51 (s, 1H), 8.19 (d, J= 8.6 Hz, 1H), 7.88 (dd, J=
2.1, 8.7 Hz, 1H),
7.84-7.76 (m, 2H), 7.40 (s, 1H), 6.81 (dõI = 8.8 Hz, 1H), 5.81 (tõ/ = 11.1 Hz,
1H), 4.41 (s, 1H),
3.62-3.52 (m, 1H), 3.49-3.38 (m, 1H), 2.71 (d, J= 5.4 Hz, 3H); ES-LCMS nilz
479.0 481.0
[M+H]. Peak 2 was concentrated under reduced pressure to yield a residue which
was dissolved
in MeCN (10 mL) and H20 (20 mL) and lyophilizedto yield 3-[(5R)-3-bromo-4,5-
dihydroisoxazol-5-y1]-N-methy1-4-[[5-(trifluoromethyl)-2-
pyridyl]aminoThenzenesulfonamide
(48.81 mg, 101.84 p.mol, 14.1% yield, 100.0% purity, SFC: Rt = 1.982, ee =
100%, [x]314D = +
40.0 (CH3OH, c = 0.050 g/100 mL)) as a yellow solid. 1HNMR (400 MHz, CDC13) 6
ppm 8.51
132
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
(s, 1H), 8.20 (d, J = 8.6 Hz, 1H), 7.89 (dd, J= 2.2, 8.6 Hz, 1H), 7.83-7.76
(m, 2H), 7.39 (s, 1H),
6.81 (d, J= 8.8 Hz, 1H), 5.81 (t, J= 11.1 Hz, 1H), 4.35 (d, J= 4.9 Hz, 1H),
3.62-3.51 (m, 1H),
3.49-3.39 (m, 1H), 2.71 (d, J= 5.4 Hz, 3H); ES-LCMS nilz 479.1, 481.1 [M-F1-
1]+.
1-12 and 1-13 (isomers of 1-26)
NN \ N
CF,
CF, Br N Boc,0 DMAP Sc(o Bu)
&
Br NH, NaBH4CN, HOAc H oc
Pd(dppf)CI,, DMF N-Boc
CF, CF,
Br
¨\\ N¨

N 0 F \\
N ifoo is CF, N CF,
TEA
Chiral SEC
Pd(dppf)Cl2, Cs2COP', N'13'¶ NaHCO,, Et0Ac DCM
¨N ¨N
Br Br
N_\\N op N¨\\ N CF, CF,
N
¨N
Br Br
Step 1: 5-Bromo-2-iodo-N-114-(trifluoromethyl)phenyllmethyllaniline
i
Br
cF,
1004231 To a solution of 5-bromo-2-iodo-aniline (2.5 g, 8.39 mmol, 1 eq) and 4-

(trifluoromethyl)benzaldehyde (4.38 g, 25.17 mmol, 3.37 mL, 3 eq) in Me0H (25
mL) was added
AcOH (50.39 mg, 839.16 mtnol, 47.99 L, 0.1 eq). The mixture was stirred at 60
C for 4 h.
NaBH3CN (2.64 g, 41.96 mmol, 5 eq) was added at 25 C. The mixture was stirred
at 60 C for
12 h. The solvent was removed and the residue was quenched by addition of
water (200 mL) and
extracted with Et0Ac (100 mL x 3). The combined organic layers were washed
with brine (50
mL), dried over Na2SO4, filtered and concentrated under reduced pressure to
yield a residue which
was purified by flash silica gel chromatography (from PE/Et0Ac = 1/0 to 5/1,
TLC: PE/Et0Ac =
133
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
5/1, Rf = 0.70) to yield 5-bromo-2-iodo-N-114-
(trifluoromethyl)phenyl]methyl]aniline (1.88 g, 3.67
mmol, 43.7% yield, 89.0% purity) as yellow oil. 1H NMR (400 MHz, CDC13) 6 ppm
7.64 (d, J =
8.1 Hz, 2H), 7.52 (d, J= 8.3 Hz, 1H), 7.47 (d, J = 7.8 Hz, 2H), 6.64-6.58 (m,
2H), 4.72 (s, 1H),
4.47 (dõI = 5.6 Hz, 2H); ES-LCMS nilz 455.9, 457.9 [M+H].
Step 2: tert-Butyl N-(5-bromo-2-iodo-phenyl)-N-114-
(trifluoromethyl)phenyllmethyl]carbamate
I
Br
Boc
CF3
[00424]
To a solution of 5-bromo-2-iodo-N[[4-
(trifluoromethyl)phenyl]methyl]aniline (1.88 g,
3.67 mmol, 89%, 1 eq) in THF (20 mL) was added DMAP (448.24 mg, 3.67 mmol, 1
eq) and
Boc20 (2.40 g, 11.01 mmol, 2.53 mL, 3 eq). The mixture was stirred at 20 C
for 12 h. The solvent
was removed to yield a residue which was purified by flash silica gel
chromatography (from
PE/Et0Ac = 1/0 to 10/1, TLC: PE/Et0Ac = 10/1, Rf = 0.8) to yield tert-buty1N-
(5-bromo-2-iodo-
pheny1)-N-[[4-(trifluoromethyl)phenylimethylicarbamate (2.23 g, crude) as a
green solid.
NMR (400 MHz, CDC13) (5 ppm 7.71 (d, .1= 8.3 Hz, 1H), 7.59 (d, .1= 7.3 Hz,
2H), 7.38 (d, .1= 7.8
Hz, 2H), 7.12 (d, J= 7.8 Hz, 1H), 6.97 (s, 1H), 5.16 (d, J= 15.2 Hz, 1H), 4.27
(d, J= 14.9 Hz,
1H), 1.57 (s, 9H); ES-LCMS nilz 499.9, 421.9 [M-t-Bu+Hr.
Step 3: tert-Butyl N-15-bromo-2-(1-methylimidazol-4-yl)phenyll-N-114-
(trifluoromethyl)phenyllmethylicarbamate
\N-\\ c3
N
N,Boc
Br
[00425] A mixture of tert-butyl
N-(5-bromo-2-iodo-pheny1)-N-[[4-
(trifluoromethyl)phenyl]methyl]carbamate (1 g, 1.73 mmol, 96.3% purity, 1 eq),
tributyl-(1-
methylimidazol-4-yl)stannane (662.53 mg, 1.73 mmol, 97% purity, 1 eq) and
Pd(dppf)C12 (126.70
134
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
mg, 173.15 tmo1, 0.1 eq) in DME (10 mL) was degassed and purged with N2 for 3
times and the
mixture was stirred under N2 atmosphere at 120 C for 4 h. The reaction
mixture was quenched by
addition of water (100 mL) and extracted with Et0Ac (50 mL x 3). The combined
organic layers
were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated
under reduced
pressure to yield a residue which purified by flash silica gel chromatography
(from PE/Et0Ac -
1/0 to 3/1, TLC: PE/Et0Ac = 3/1, Rf = 0.40) to yield tert-butyl N45-bromo-2-(1-
methylimidazol-
4-yl)phenyl]-N-[[4-(trifluoromethyl)phenyl]methyl]carbamate (530 mg, 934.66
mot, 54.0%
yield, 90.0% purity) as black brown oil. I-H NMR (400 MHz, CDC13) 6 ppm 7.97
(d, J = 7.6 Hz,
1H), 7.55 (d, = 8.1 Hz, 2H), 7.46 (s, 2H), 7.35 (d, ,/ = 6.8 Hz, 2H), 6.94 (s,
1H), 6.82 (s, 1H),
5.15 (d, J = 14.4 Hz, 1H), 4.18 (d, J = 14.9 Hz, 1H), 3.66 (s, 3H), 1.26 (s,
9H); ES-LCMS nilz
510.1, 512.1 [M+Hr.
Step 4: tert-Butyl N-12-(1-methylimidazol-4-y1)-5-vinyl-phenyll-N-114-
(trifluoromethyl)phenyllmethyllcarbamate
N-\\
N
N,Boc
1004261 A mixture of tert-butyl N45-bromo-2-(1-methylimidazol-4-yl)phenyll-
N4[4-
(trifluoromethyl)phenyl]methyl]carbamate (530 mg, 934.66 jimol, 90%, 1 eq),
4,4,5,5-
tetramethy1-2-viny1-1,3,2-dioxaborolane (287.90 mg, 1.87 mmol, 317.07 iL, 2
eq), Pd(dppf)C12
(68.39 mg, 93.47 [imol, 0.1 eq), Cs2CO3 (761.33 mg, 2.34 mmol, 2.5 eq) in 1,4-
dioxane (4.5 mL)
and H20 (1.5 mL) was degassed and purged with N2 for 3 times and the mixture
was stirred under
N2 atmosphere at 100 C for 12 h. The reaction mixture was quenched by
addition of water (50
mL) and extracted with Et0Ac (30 mL x 3). The combined organic layers were
washed with brine
(10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure
to yield a residue
which was purified by flash silica gel chromatography (from PE/Et0Ac = 1/0 to
3/1, TLC:
PE/Et0Ac =3/1, Rf = 0.40) to yield tert-butyl N42-(1-methylimidazol-4-y1)-5-
vinyl-pheny1]-N-
[[4-(trifluoromethyl)phenyl]methyl]carbamate (219 mg, 464.34 jimol, 49.7%
yield, 97.0% purity)
135
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
as yellow oil. 111 NMR (400 MHz, CDC13) 6 ppm 8.05 (d, J= 8.1 Hz, 1H), 7.54
(d, J= 7.8 Hz,
3H), 7.47 (s, 1H), 7.38 (d, J= 7.3 Hz, 3H), 6.85 (s, 1H), 6.74 (s, 1H), 6.57
(dd, J= 10.9, 17.5 Hz,
1H), 5.55 (d, J= 17.4 Hz, 1H), 5.18 (d, J= 10.8 Hz, 1H), 5.22-5.15 (m, 1H),
3.67 (s, 3H), 1.25 (s,
9H); ES-LCMS m/z 458.1 [M+H].
Step 5: tert-Butyl N-12-(1-methylimidazol-4-y1)-5-vinyl-phenyll-N-114-
(trifluoromethyl)phenyl]methyl]carbamate
N¨\\
N C F3
Boc
¨N
Br
1004271 To a solution of tert-butyl N42-(1-methylimidazol-4-y1)-5-
vinyl-phenyli-N-[[4-
(trifluoromethyl)phenylimethylicarbamate (169 mg, 358.33 [tmol, 97%, 1 eq) and

dibromomethanone oxime (109.02 mg, 537.49 iumol, 1.5 eq) in Et0Ac (3 mL) was
added NaHCO3
(301.02 mg, 3.58 mmol, 10 eq) The mixture was stirred at 25 C for 4 h The
reaction mixture
was quenched by addition of water (50 mL) and extracted with Et0Ac (30 mL x
3). The combined
organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and
concentrated under
reduced pressure to yield
tert-butyl /V-[5-(3 -brom o-4,5-di hydroi soxazol-5-y1)-2-(1-
methylimidazol-4-y1)phenyl]-N-[[4-(trifluoromethyl)phenyl]methyl]carbamate
(248 mg, crude) as
a yellow solid which was used in the next step without further purification. 1-
11 NMR (400 MHz,
CDC13) 6 ppm 8.10 (s, 1H), 7.64 (s, 2H), 7.60 (d, J= 8.3 Hz, 2H), 7.51-7.47
(m, 1H), 6.95-6.82
(m, 2H), 6.68 (s, 1H), 5.54 (d, J= 8.8 Hz, 1H), 5.30-5.08 (m, 2H), 3.71 (s,
3H), 3.59-3.45 (m, 2H),
1.54 (s, 9H).
Step 6: (S)-5-(3-Bromo-4,5-dihydroisoxazol-5-y1)-2-(1-methy1-1H-imidazol-4-y1)-
N-(4-
(trifluoromethyl)benzyl)aniline and (R)-5-(3-bromo-4,5-dihydroisoxazol-5-y1)-2-
(1-methy1-
1H-imidazol-4-y1)-N-(4-(trifluoromethyl)benzypaniline
136
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
N-\\
N-\\
N CF3 N
H CF3
4111 110
0 c0
-N
Br Brc
1004281 To a solution of tert-butyl N45-(3-bromo-4,5-dihydroisoxazol-5-y1)-2-
(1-
methylimidazol-4-yl)phenyl]-N-[[4-(trifluoromethyl)phenyl]methyl]carbamate
(248 mg, 428.02
p.mol, 1 eq) in DCM (3 mL) was added TFA (184.80 mg, 1.62 mmol, 120 p,L, 3.79
eq). The
mixture was stirred at 25 C for 1 h. The reaction mixture was quenched by
addition of water (100
mL) and extracted with Et0Ac (60 mL x 3). The combined organic layers were
washed with brine
(20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure
to yield a residue
which was purified by flash silica gel chromatography (from PE/Ft0Ac = 100/1
to 3/1, TLC:
PE/Et0Ac = 3/1, Rf= 0.05)to yield the compound which was separated by SFC
(column: DAICEL
CHIRALPAK AD (250mm*30mm, 10p.m); mobile phase: [0.1% NH3H20 ETOH]; B%: 50%-
50%) to yield Peak 1 and Peak 2. Peak 1 was purified by preparative HPLC
(column: Agela
DuraShell C18 150*25mm*5[tm; mobile phase: [water (0.05% NH3H20+10mM NH4HCO3)-
ACN]; B%: 52%-82%, 10min) and lyophilized to yield (S)-5-(3-bromo-4,5-
dihydroisoxazol-5-y1)-
-methy1-1H-imidazol-4-y1)-N-(4-(trifluoromethyl)benzypaniline (10.44 mg, 21.35
p.mol,
5.0% yield, 98.5% purity, SFC : Rt = 2.143, ee = 98.2%, [cd32- D= + 180.0
(Me0H, c = 0.02 g/100
mL)) as a white solid. 1H NMR (400 MHz, CDC13) (3 ppm 8.52 (s, 1H), 7.60-7.56
(m, 2H), 7.54-
7.50(m, 2H), 7.47 (s, 1H), 7.39 (d, J= 7.8 Hz, 1H), 7.18 (s, 1H), 6.61 (d, J=
7.8 Hz, 1H), 6.47 (s,
1H), 5.57-5.47 (m, 1H), 4.55 (s, 2H), 3.77 (s, 3H), 3.50 (dd, J= 10.9, 17.2
Hz, 1H), 3.07 (dd, J=
8.9, 17.2 Hz, 1H); ES-LCMS rn/z 478.8, 480.8 [M-H]. Peak 2 was purified by
preparative HPLC
(column: Agela DuraShell C18 150*25mm*5 m; mobile phase: [water (0.05%
NH3H20+10mM
NH4HCO3)-ACN]; B%: 51%-81%, 10 min) and lyophilized to yield (R)-5-(3-bromo-
4,5-
di hydroi soxazol -5-y1)-2-(1-m ethyl -1H-i mi dazol -4-y1)-N-(4-(tri fluorom
ethyl)benzyl)aniline (9.5
mg, 18.99 mot, 4.4% yield, 95.9% purity, SFC : Rt = 2.482, ee = 98.8%, ee =
98.8%, [a]32- D= -
187.5(Me0H, c = 0.016 g/100 mL)) as a white solid. 1-FI NIVIR (400 MHz, CDC13)
ppm 8.52 (s,
1H), 7.60-7.56 (m, 2H), 7.53-7.50 (m, 2H), 7.47 (s, 1H), 7.39 (d, J= 7.8 Hz,
1H), 7.18 (d, J= 1.2
137
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
Hz, 1H), 6.61 (d, J= 7.8 Hz, 1H), 6.47 (s, 1H), 5.52 (dd, J= 8.9, 10.9 Hz,
1H), 4.55 (s, 2H), 3.77
(s, 3H), 3.50 (dd, J = 10.9, 17.2 Hz, 1H), 3.07 (dd, J= 8.8, 17.1 Hz, 1H); ES-
LCMS m/z 479.0,
481.0 [M-F1-1]+.
1-14
"N-\\ \N-\\
Br
H2N-0-CF3 Br r4
N
N
NaH
\VD, cIrdpspZIB'DJ
0:N P
cF N'acE3 Et3N DCM 9
NO,
H:N 4141)-P CF,
Step 1: N-(2-Bromo-4-nitropheny1)-5-(trifluoromethyl)pyridin-2-amine
Br
1401
0 2 N CF3
1004291 To a solution of 5-(trifluoromethyl) pyridin-2-amine (1.84 g, 11.36
mmol, 1 eq) in THF
(30 mL) was added NaH (1.36 g, 34.09 mmol, 60%, 3 eq) at 0 C. The mixture was
stirred for 30
min. 2-Bromo-1-fluoro-4-nitro-benzene (2.5 g, 11.36 mmol, 1 eq) was added at 0
C and the
mixture was stirred at 25 C for 12 h. The reaction mixture was quenched with
water (50 mL) and
extracted with Et0Ac (50 mL x 3). The combined organic layers were dried over
Na2SO4, filtered
and concentrated under reduced pressure to yield a residue which was purified
by flash silica gel
chromatography (from PE/Et0Ac = 100/1 to 10/1, TLC: PE/Et0Ac = 5/1, Rf = 0.65)
to yield N-
(2-bromo-4-nitro-pheny1)-5-(trifluoromethyl)pyridin-2-amine (850 mg, 2.00
mmol, 17.6% yield,
85.0% purity) as a yellow solid. 11-INMIR (400 MHz, CDC13) 6 ppm 8.76 (d, J =
9.3 Hz, 1H), 8.63
(s, 1H), 8.52 (d, J = 2.4 Hz, 1H), 8.24 (dd, J= 2.4, 9.3 Hz, 1H), 7.87 (dd, J=
2.2, 8.8 Hz, 1H),
7.54 (s, 1H), 7.00 (d, J = 8.8 Hz, 1H); ES-LCMS nilz 363.9 [M-41] .
Step 2: 2-(1-Methy1-1H-imidazol-4-y1)-N1-(5-(trifluoromethyl)pyridin-2-
y1)benzene-1,4-
diamine
\N-A
N
N
N
H2N CF3
1004301 To a solution of N-(2-bromo-4-nitro-phenyl)-5-(trifluoromethyl)pyridin-
2-amine (700
138
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
mg, 1.93 mmol, 1 eq) and tributyl-(1-methylimidazol-4-y1)stannane (1.58 g,
3.87 mmol, 91%, 2
eq) in DMF (15 mL) was added Pd(dppf)C12 (70.73 mg, 96.66 p.mol, 0.05 eq)
under N2
atmosphere. The mixture was stirred under N2 atmosphere at 130 C for 12 h.
The reaction mixture
was partitioned between water (50 mL) and Et0Ac (100 mL x 3). The organic
phase was separated,
washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under
reduced pressure
to yield a residue which was purified by silica gel column chromatography
(from pure PE to
PE/Et0Ac = 0/1, TLC: PE/Et0Ac = 0/1, Rf = 0.34) to yield 2-(1-methylimidazol-4-
y1)-A7145-
(trifluoromethyl)-2-pyridylibenzene-1,4-diamine (300 mg, 648.04 mol, 33.5%
yield, 72.1%
purity) as a yellow oil. 1H NN4R (400 MHz, CDC13) 6 ppm 9.41 (s, 1H), 8.40 (s,
1H), 7.72 (d, J=
8.6 Hz, 1H), 7.59-7.43 (m, 2H), 7.27 (s, 1H), 7.18-7.01 (m, 2H), 6.74-6.57 (m,
2H), 3.71 (s, 3H);
ES-LCMS m/z 334.3 [M+H].
Step 3: N-(3-(1-Methy1-1H-imidazol-4-y1)-4-05-(trifluoromethyl)pyridin-2-
yDamino)phenyl)acrylamide
\N¨\\
N
0 N
CF3
1004311 To a solution of 2-(1-methylimidazol-4-y1)-N145-(trifluoromethyl)-2-
pyridyl]benzene-1,4-di amine (230 mg, 496.83 timol, 72%, 1 eq) and Et3N
(150.82 mg, 1.49 mmol,
207.46 pt, 3 eq) in DCM (3 mL) was added prop-2-enoyl chloride (58.46 mg,
645.88 litmol, 52.66
pL, 1.3 eq) under N2 atmosphere at 0 C. The mixture was stirred at 25 C for
2 h. The reaction
mixture was partitioned between water (30 mL) and Et0Ac (50 mL x 3). The
organic phase was
separated, washed with brine (30 mL), dried over Na2SO4, filtered and
concentrated under reduced
pressure to yield a residue which was purified by preparative HPLC (column:
Agela DuraShell
C18 150*25 mm*5 pm; mobile phase: [water (0.05% NH3H20 + 10 mM NH4HCO3)-ACN];
B%:
38%-68%, 10 min) to yield N43-(1-methylimidazol-4-y1)-4-[[5-(trifluoromethyl)-
2-
pyridyl]amino]phenyl]prop-2-enamide (85.15 mg, 215.43 pmol, 43.4% yield, 98.2%
purity) as a
white solid. 1H NMR (500 MHz, CDC13) 6 ppm 11.30 (s, 1H), 8.44 (s, 1H), 8.38
(d, J= 8.9 Hz,
1H), 8.17 (d, J= 2.0 Hz, 1H), 7.74 (s, 1H), 7.60 (dd, J= 2.0, 8.9 Hz, 1H),
7.47 (s, 1H), 7.18 (s,
139
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
2H), 6.82 (d, J= 8.7 Hz, 1H), 6.49-6.40 (m, 1H), 6.35-6.24 (m, 1H), 5.75 (d,
J= 10.4 Hz, 1H),
3.68 (s, 3H); ES-LCMS nilz 388.2 [M-41] .
1-15 and 1-16 (isomers of 1-27)
Br CI CI
-N -N
co ç
HCI SFC H
aiht. N
0
11011
c F3 3
CF3
N S S N S
H \\C) H
Step 1: 3-1(5S)-3-Chloro-4,5-dihydroisoxazol-5-y1]-N-methyl-4415-
(trifluoromethyl)-2-
pyridyllaminolbenzenesulfonamide
ci
H
R\ N
,S CF3
N
H
1004321 To a solution of 3 -(3 -bromo-4,5-dihydroi
soxazol-5-y1)-N-methy1-4-[[5-
(trifluoromethyl)-2-pyridyl] aminoThenzenesulfonamide (900 mg, 723.35 umol,
38.5% purity, 1
eq) in 1,4-dioxane (10 mL) was added aq. HC1 (2 mL). The mixture was stirred
at 40 C for 12 h.
The reaction mixture was quenched by addition of saturated NaHCO3 (100 mL) and
extracted with
Et0Ac (50 mL x 3). The combined organic layers were washed with brine (50 mL),
dried over
Na2SO4, filtered and concentrated under reduced pressure to yield a residue
which was purified by
preparative HPLC (column: Agela DuraShell C18 150*25mm*5um; mobile phase:
[water
(0.05%NH3.H20-F10mM NH4HCO3)-ACN]; B%: 40%-70%, 10 min), followed by
lyophilization
to yield the mixture which was separated by chiral SFC (column: column: DAICEL
CHIRALPAK
AS(250mm*30mm,10 m); mobile phase: [0.1% NH3-1-120 Et0H]; B%: 25%-20%) to
yield Peak
1 and Peak 2. Peak 1 was concentrated under reduced pressure to yield a
residue which was
dissolved in MeCN (10 mL) and H20 (20 mL) then lyophilized to yield 3-[(5S)-3-
chloro-4,5-
dihydroisoxazol-5-yli-N-methy1-4-[[5-(trifluoromethyl)-2-
pyridyliaminoThenzenesulfonamide
(48.36 mg, 111.22 timol, 15.4% yield, 100.0% purity, SFC: Rt = 1.382, cc =
99.84%, [a131-4D = -
46.15 (CH3OH, c = 0.052 g/100 mL)) as a white solid. 1H NMR (400 MHz, CDC13)
ri ppm 8.51
(s, 1H), 8.19 (d, J = 8.8 Hz, 1H), 7.89 (dd, J= 2.1, 8.7 Hz, 1H), 7.82-7.77
(m, 2H), 7.42 (s, 1H),
140
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
6.82 (d, J= 8.8 Hz, 1H), 5.88 (t, J= 11.1 Hz, 1H), 4.46-4.33 (m, 1H), 3.56-
3.37 (m, 2H), 2.71 (d,
J= 5.1 Hz, 3H); ES-LCMS m/z 435.1, 437.1 [M-41] .
1-17 and 1-18 (isomers of 1-28)
Br Br Br i Br H
N
CI -N
PMB NH3 H20 ---ANH' ______________
so
Et3N, THF s N Pd(OAc)2, Xarilphos N
CF3
ci 0 0 Cs2CO3, anisole 0
PMB PMB PMB
Br
¨N
,rN
TFA .OH
0 I NH
--pcm N _ NaHCO3 Pd(dppf)C12,
Cs2CO3 N
411111)" CF 3 CF;
Br,IN CZ,
IN NH SO
PMBC) H
CF,
Mo
Brtie)
Chiral SFC, H
)'N
CF3
µ0
Step 1: 5-13romo-6-chloro-N-1(4-methoxyphenyl)methyll-N-methyl-pyridine-3-
sulfonamide
Br
CI
0
.N
-S\
N
PMB
1004331 To a solution of 5-bromo-6-chloro-pyridine-3-sulfonyl
chloride (1.0 g, 3.44 mmol, 1
eq) and Et3N (695.58 mg, 6.87 mmol, 956.78 p.L, 2 eq) in THE (10 mL) was added
1-(4-
methoxypheny1)-N-methyl-methanamine (571.67 mg, 3.78 mmol, 1.1 eq). The
mixture was stirred
at -30 C for 1 h. TLC (PE/Et0Ac = 3/1, Rf = 0.56) indicated the starting
material was consumed
completely and one new spot formed. The mixture was poured into water (50 mL)
and the
precipitated solid was filtered and dried to yield 5-bromo-6-chloro-N-[(4-
methoxyphenyl)methyli-
N-methyl-pyridine-3-sulfonamide (1.1 g, 2.49 mmol, 72.6% yield, 92.0% purity)
as a light yellow
solid, which was used in the next step without further purification. 1H NMR
(500 MHz, CDC13)
ppm 8.71 (d, J = 2.1 Hz, 1H), 8.23 (d, J = 2.1 Hz, 1H), 7.21 (d, J = 8.7 Hz,
2H), 6.90-6.85 (m,
2H), 4.18 (s, 2H), 3.81 (s, 3H), 2.70 (s, 3H); ES-LCMS m/z 405.0, 407.0 [M+H]t
Step 2: 6-Amino-5-bromo-N-1(4-methoxyphenyl)methy1]-N-methyl-pyridine-3-
sulfonamide
141
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Br
NH 2
10µ
\S
N
0
PMB
[00434] To a solution of 5-bromo-6-chloro-N-[(4-methoxyphenyl)methyl]-N-methyl-
pyridine-
3-sulfonamide (950 mg, 2.15 mmol, 92% purity, 1 eq) in THF (5 mL) was added
NH3.H20 (1.48
mL, 28% purity, 5 eq). The mixture was stirred under microwave at 100 C for
12 h. TLC
(PE/Et0Ac = 1/1, Rf = 0.31) indicated the starting material was consumed
completely and one
new spot formed. The solvent was removed to yield 6-amino-5-bromo-N-[(4-
methoxyphenyl)methy1]-N-methyl-pyridine-3-sulfonamide (860 mg, 2.12 mmol,
98.2% yield,
95.0% purity) as a yellow solid, which was used in the next step without
further purification. '1-1
NMR (500 MHz, DMSO-d6) 6 ppm 8.33 (d, J= 2.0 Hz, 1H), 7.99 (d, J= 2.0 Hz, 1H),
7.31 (br s,
2H), 7.23 (d, J = 8.4 Hz, 3H), 6.92 (d, J = 8.5 Hz, 2H), 3.75 (s, 3H).
Step 3: 5-Bromo-N-1(4-methoxyphenypmethyll-N-methy1-644-
(trifluoromethyl)anilinolpyridine-3-sulfonamide
Br
N
0, N
CF3
PMB
[00435] To a solution of 6-amino-5-bromo-N-[(4-methoxyphenyl)methyll-N-methyl-
pyridine-
3-sulfonamide (510 mg, 1.25 mmol, 95% purity, 1 eq) and 1-iodo-4-
(trifluoromethyl)benzene
(409.42 mg, 1.51 mmol, 221.31 p.t, 1.2 eq) in anisole (20 mL) were added
Pd(OAc)2 (42.24 mg,
188.15 p.mol, 0.15 eq), xantphos (72.58 mg, 125.43 p.mol, 0.1 eq) and Cs2CO3
(613.02 mg, 1.88
mmol, 1.5 eq). The mixture was stirred under N2 atmosphere at 130 C for 16 h.
The solvent was
removed and the residue was treated with Et0Ac (30 mL). The mixture was
filtered and the filtrate
was concentrated to yield a residue which was purified by flash silica gel
chromatography (from
PE/Et0Ac = 1/0 to 3/1, TLC: PE/Et0Ac = 3/1, Rf = 0.57) to yield 5-bromo-N-[(4-
methoxyphenyl)methy1]-N-methy1-6- [4-(trifluoromethyl)anilino]pyri dine-3 -
sulfonamide (587
mg, 1.05 mmol, 83.8% yield, 95.0% purity) as a yellow solid.11-1 N1VIR (500
MHz, CDC13) 6 ppm
8.61 (d, J = 1.8 Hz, 1H), 8.12 (d, J = 1.8 Hz, 1H), 7.81 (d, J = 8.5 Hz, 2H),
7.64 (d, J = 8.4 Hz,
2H), 7.51 (s, 1H), 7.23 (d, I = 8.5 Hz, 2H), 6.88 (d, J = 8.5 Hz, 2H), 4.14
(s, 2H), 3.81 (s, 3H),
142
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
2.65 (s, 3H); ES-LCMS nilz 530.0, 532.0 [M+H]t
Step 4: N-1(4-Methoxyphenyl)methyll-N-methy1-6-14-(trifluoromethyl)anilino1-5-
vinyl-
pyridine-3-sulfonamide
0 1101
S C F3
N
0
PMB
1004361 To a solution of 5-brom o-/V-[(4-m ethoxyphenyl
)methyl ]-AT-methyl -644-
(trifluoromethyl)anilino]pyridine-3-sulfonamide (587 mg, 1.05 mmol, 95%
purity, 1 eq) and
4,4,5,5-tetramethy1-2-vinyl-1,3,2-dioxaborolane (323.88 mg, 2.10 mmol, 356.70
u-Lõ 2 eq) in 1,4-
dioxane (18 mL) and H20 (3 mL) were added Pd(dppf)C12 (76.94 mg, 105.15 umol,
0.1 eq) and
Cs2CO3 (685.17 mg, 2.10 mmol, 2 eq). The mixture was stirred under N2
atmosphere at 90 C for
16 h. The solvent was removed and the residue was treated with Et0Ac (20 inL).
The mixture was
filtered and the filtrate was concentrated to yield a residue which was
purified by flash silica gel
chromatography (from PE/Et0Ac = 1/0 to 3/1, TLC: PE/Et0Ac = 3/1, Rf = 0.49) to
yield N-[(4-
methoxyphenyl)methyl] -N-methyl-6-14-(trifluoromethyl)anilino]-5-vinyl-pyri
dine-3 -sulfonamide
(350 mg, 732.99 umol, 69.7% yield, 100.0% purity) as a yellow solid. 1H NMR
(500 MHz, CDC13)
6 ppm 8.62 (d, J= 2.3 Hz, 1H), 7.89 (d, J= 2.3 Hz, 1H), 7.75 (d, J= 8.5 Hz,
2H), 7.61 (d, J= 8.5
Hz, 2H), 7.23 (d, J= 8.5 Hz, 2H), 6.87 (d, J= 8.5 Hz, 3H), 6.75 (dd, J= 11.1,
17.3 Hz, 1H), 5.84
(d, J= 17.2 Hz, 1H), 5.71 (d, J= 11.1 Hz, 1H), 4.13 (s, 2H), 3.80 (s, 3H),
2.63 (s, 3H); ES-LCMS
nilz 478.2 [M+H].
Step 5: N-Methyl-644-(trifluoromethyl)anilinol-5-vinyl-pyridine-3-sulfonamide
0,
,S CF3
N
H
1004371 To a solution of N-1(4-methoxyphenyl)methy1]-N-methy1-6-14-
(trifluoromethyl)anilino]-5-vinyl-pyridine-3-sulfonamide (300 mg, 628.27 umol,
100% purity, 1
eq) in DCM (5 mL) was added TFA (1.5 mL). The mixture was stirred at 25 C for
16 h. TLC
143
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
(PE/Et0Ac = 1/1, Rf = 0.58) indicated the starting material was consumed
completely and one
new spot formed. The mixture was concentrated to yield N-methy1-644-
(trifluoromethyl)anilino]-
5-vinyl-pyridine-3-sulfonamide (330 mg, 616.07 umol, 98.1% yield, 88.0%
purity, TFA) as an
off-white solid, which was used in the next step without further purification.
I-HNN4R (500 MHz,
CD30D) 6 ppm 8.50 (d, J¨ 2.3 Hz, 1H), 8.09 (d, J¨ 2.1 Hz, 1H), 7.85 (d, J¨ 8.5
Hz, 2H), 7.62
(d, J= 8.5 Hz, 2H), 7.05 (dd, J= 10.9, 17.2 Hz, 1H), 5.91 (d, J= 17.1 Hz, 1H),
5.62 (d, J= 11.0
Hz, 1H), 2.59 (s, 3H).
Step 6: 5-(3-Bromo-4,5-dihydroisoxazol-5-y1)-N-rnethy1-6-14-
(trifluoromethyDanilinolpyridine-3-sulfonamide
Br
,Z3 H
N s\>, CF3
H
1004381 To a solution of N-methy1-6-[4-(trifluoromethyl)anilino]-5-vinyl-
pyridine-3-
sulfonamide (330 mg, 616.07 umol, 88% purity, 1 eq, TFA) and dibromomethanone
oxime
(249.92 mg, 1.23 mmol, 2 eq) in Et0Ac (20 mL) was added NaHCO3 (517.54 mg,
6.16 mmol, 10
eq). The mixture was stirred at 25 C for 3 h. TLC (PE/Et0Ac = 1/1, Rf = 0.78)
indicated the
starting material was consumed completely and one new spot formed. The mixture
was filtered
and the filtrate was concentrated to yield a residue which was purified by
flash silica gel
chromatography (from PE/Et0Ac = 1/0 to 2/1, TLC: PE/Et0Ac = 1/1, Rf = 0.78) to
yield 5-(3-
bromo-4,5-dihydroi soxazol -5-y1)-/V-m ethyl -6[4-(tri fluorom ethypani i
no]pyri dine-3 -
sulfonamide (290 mg, 574.83 litmol, 93.3% yield, 95.0% purity) as a yellow
solid. 1H NMR (400
MHz, CDC13) 6 ppm 8.71 (d, J= 2.2 Hz, 1H), 7.85 (d, J= 2.0 Hz, 1H), 7.74-7.66
(m, 2H), 7.64-
7.57 (m, 2H), 5.77 (t, J= 11.2 Hz, 1H), 4.39 (q, J= 5.4 Hz, 1H), 4.12 (q, J=
7.1 Hz, 1H), 3.56
(dd, J= 1.8, 11.4 Hz, 2H), 2.71 (d, J= 5.4 Hz, 3H); ES-LCMS nilz 479.0, 481.0
[M-Ffi].
Step 7: (S)-5-(3-Bromo-4,5-dihydroisoxazol-5-y1)-N-methy1-6-((4-
(trifluoromethyl)phenyl)amino)pyridine-3-sulfonamide
144
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Br
H
0N
N
CF3
H
1004391 The compound 5-(3-bromo-4,5-dihydroisoxazol-5-
y1)-N-methy1-6-[4-
(trifluoromethypanilino]pyridine-3-sulfonamide (100 mg, 198.22 pmol, 95%
purity, 1 eq) was
separated by chiral SFC (column: DATCEL CHTRALPAK AD(250mm*30mm,10um);mobile
phase: [0.1%NH3H20 Et0E1]; B%: 30%-30%) to yield Peak 1 and Peak 2. Peak 1 was
concentrated
under reduced pressure to yield a residue which was dissolved in MeCN (10 mL)
and H20 (20
mL) and lyophilized to yield (S)-5-(3-bromo-4,5-dihydroisoxazol-5-y1)-N-methy1-
64(4-
(trifluoromethyl)phenyl)amino)pyridine-3-sulfonamide (32.79 mg, 68.42 p.mol,
34.5% yield,
100.0% purity, SFC: R1 = 1.372, ee = 100%, [0(]28.6D _5.00 (Me0H, c = 0.08
g/100 mL) as a
white solid. 1H NMIR (500 MHz, CDCh) 6 ppm 8.71 (d, J= 2.1 Hz, 1H), 7.85 (d,
J= 2.1 Hz, 1H),
7.72-7.65 (m, 3H), 7.64-7.58 (m, 2H), 5.77 (t, J= 11.2 Hz, 1H), 4.44 (q, J=
5.2 Hz, 1H), 3.62-
3.51 (m, 2H), 2.71 (d, J = 5.3 Hz, 3H); ES-LCMS nilz 478.9, 480.9 [M+Hr and
Peak 2 was
concentrated under reduced pressure to yield a residue which was dissolved in
MeCN (10 mL) and
H20 (20 mT,) and lyophilized to yield (R)-5-(3-bromo-4,5-dihydroisoxazol-5-y1)-
1V-methyl-6-((4-
(trifluoromethyl)phenyl)amino)pyridine-3-sulfonamide (31.64 mg, 66.02 pmol,
33.3% yield,
100.0% purity, SFC: Rt = 1.596, ee = 100%, [c(]28-8D = +6.67 (Me0H, c = 0.09
g/100 mL) as a
white solid.1HNMR (500 MHz, CDC13) 6 ppm 8.70 (d, J= 2.3 Hz, 1H), 7.86 (d, J=
2.1 Hz, 1H),
7.72-7.65 (m, 3H), 7.63-7.59 (m, 2H), 5.77 (t, J = 11.1 Hz, 1H), 4.48 (q, J=
5.3 Hz, 1H), 3.62-
3.51 (m, 2H), 2.70 (d, J = 5.3 Hz, 3H); ES-LCMS m/z 479.0, 481.0 [M+Hr.
1-19 and 1-20 (isomers of 1-29)
145
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
uN/
N
/nN/ N
4),"N
/rN
Am NH, 5n(-Bu), N 7 N
Br:NH
NH2
Pd(dppf)C12 Br am NH2 Pd(dppf)C12, Cs200, NH, NaHCO,
"IIIU N \
1111
Br
uN/ Br "IP
uN/ irN/
Br
CF0 N CF, N - N
CE,
AMM
THF, DIEA, 25 C
Nic Nip N \
Br Br Br
Step 1: 4-Bromo-2-(1-methylimidazol-4-yl)aniline
N
NH2
Br
1004401 To a mixture of 4-bromo-2-iodo-aniline (2 g, 6.71 mmol, 1 eq) and
tributyl-(1-
methylimidazol-4-yl)stannane (2.72 g, 6.71 mmol, 91.5%, 1 eq) in DMF (20 mL)
was added
Pd(dppf)C12 (491.22 mg, 671.32 [Imo], 0.1 eq). The mixture was stirred under
N2 atmosphere at
130 C for 12 h. The mixture was diluted with water (100 mL) and extracted
with Et0Ac (100 mL
x 3). The combined organic phase was washed with brine (50 mL), dried over
anhydrous Na2SO4,
filtered and concentrated in vacuum to yield a residue which was purified by
flash silica gel
chromatography (from PE/Et0Ac = 100/1 to 0/1, TLC: PE/Et0Ac = 0/1, Rf = 0.30)
to yield 4-
bromo-2-(1-methylimidazol-4-yl)aniline (1 g, 3.25 mmol, 48.5% yield, 82.0%
purity) as yellow
oil. 1H NIVIR (400 MHz, CDC13) 6 ppm 7.47-7.42 (m, 2H), 7.13-7.06 (m, 2H),
6.58 (d, J= 8.6 Hz,
1H), 5.72-5.34 (m, 2H), 3.72 (s, 3H); ES-LCMS nilz 252.0, 254.0 [M+Hr.
Step 2: 2-(1-Methylimidazol-4-y1)-4-vinyl-aniline
N
NH2
1004411 To a solution of 4-bromo-2-(1-methylimidazol-4-yl)aniline (1
g, 3.25 mmol, 82.0%, 1
146
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
eq) and 4,4,5,5-tetramethy1-2-vinyl-1,3,2-dioxaborolane (1.00 g, 6.51 mmol,
1.10 mL, 2 eq) in
1,4-dioxane (30 mL) and H20 (6 mL) was added Pd(dppf)C12 (237.99 mg, 325.25
mol, 0.1 eq)
and Cs2CO3 (3.18 g, 9.76 mmol, 3 eq). The mixture was stirred under N2
atmosphere at 100 C for
2 h. The mixture was diluted with water (100 mL) and extracted with Et0Ac (100
mL x 3). The
combined organic phase was washed with brine (50 mL), dried over anhydrous
Na2SO4, filtered
and concentrated in vacuum to yield a residue which was purified by flash
silica gel
chromatography (from PE/Et0Ac = 100/1 to 0/1, TLC: PE/Et0Ac = 0/1, Rf = 0.35)
to yield 2-(1-
methylimidazol-4-y1)-4-vinyl-aniline (500 mg, 2.16 mmol, 66.4% yield, 86.0%
purity) as brown
oil. 1H NMR (400 MHz, DMSO-do) 6 ppm 7.68 (s, 1H), 7.56 (d, J= 1.2 Hz, 1H),
7.46 (d, J= 2.0
Hz, 1H), 7.06 (dd, J = 2.0, 8.2 Hz, 1H), 6.62 (d, J = 8.2 Hz, 1H), 6.55 (dd, J
= 11.0, 17.6 Hz, 1H),
6.43 (s, 2H), 5.53 (dd, J= 1.2, 17.6 Hz, 1H), 4.94 (dd, J= 1.0, 10.8 Hz, 1H),
3.70 (s, 3H); ES-
LCMS m/z 200.3 [M+H].
Step 3: 4-(3-Bromo-4,5-dihydroisoxazol-5-y1)-2-(1-methylimidazol-4-yl)aniline
N
ei NH2
Br
[00442] To a solution of 2-(1-methylimidazol-4-y1)-4-vinyl-aniline
(500 mg, 2.16 mmol,
86.0%, 1 eq) in Et0Ac (10 mL) was added NaHCO3 (1.81 g, 21.58 mmol, 839.35 L,
10 eq) and
dibromomethanone oxime (656.59 mg, 3.24 mmol, 1.5 eq). The mixture was stirred
at 25 C for
12 h. The reaction mixture was filtered and concentrated under reduced
pressure to yield a residue
which was purified by preparative TLC (PE/Et0Ac = 0/1, TLC: PE/Et0Ac = 0/1, Rf
= 0.20) to
yield 4-(3-bromo-4,5-dihydroisoxazol-5-y1)-2-(1-methylimidazol-4-yl)aniline
(150 mg, 434.35
mol, 20.1% yield, 93.0% purity) as yellow oi1.11-1 NMR (500 MHz, CDC13) 6 ppm
7.47 (s, 1H),
7.34 (d, J= 1.8 Hz, 1H), 7.17 (s, 1H), 7.01-6.99 (m, 1H), 6.71 (d, J= 8.2 Hz,
1H), 5.57 (t, J= 10.2
Hz, 2H), 3.80-3.71 (m, 3H), 3.51 (dd, J= 10.8, 17.3 Hz, 1H), 3.23 (dd, J= 9.8,
17.3 Hz, 1H), 2.98-
1.99 (m, 1H); ES-LCMS m/z 321.1, 323.1 [M-Ffi]t.
147
CA 03200814 2023- 5- 31

WO 2022/120354 PCT/US2021/072684
Step 4: 4-1(5S)-3-Bromo-4,5-dihydroisoxazol-5-y11-2-(1-methylimidazol-4-y1)-N-
R4-
(trifluoromethyl)phenyllmethyllaniline and 4-1(5R)-3-bromo-4,5-dihydroisoxazol-
5-y11-2-(1-
methylimidazol-4-y1)-N-114-(trifluoromethyl)phenyllmethyllaniline
N CF3 N 7 c,3
p .0% p
N).3
Br Br
1004431 To a solution of 4-(3-bromo-4,5-dihydroisoxazol-5-y1)-2-(1-
methylimidazol-4-
yl)aniline (120 mg, 347.48 prnol, 93.0%, 1 eq) in THF (5 mL) was added DIEA
(134.73 mg, 1.04
mmol, 181.57 !IL, 3 eq) and 1-(bromomethyl)-4-(trifluoromethyl)benzene (166.12
mg, 694.95
pinol, 107.17 pL, 2 eq). The mixture was stirred at 25 C for 12 h. The
mixture was diluted with
water (30 mL) and extracted with Et0Ac (30 mL x 3). The combined organic phase
was washed
with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in
vacuum to yield a
residue which was purified by preparative HPLC (column: Boston Prime C18
150*30 mm*5 p.m;
mobile phase: [water (0.05% NH3.H20+10 mM NH4HCO3)-ACN]; B%: 60%-90%, 10 min),

followed by lyophilization to yield a product. The product was separated by
SFC (column:
DAICEL CHlRALPAK IG (250 mm*50 mm, 10 pm); mobile phase: [0.1% NH3.H20 Et0H];
B%:
60%-60%) to yield peak 1 and peak 2. Peak 1 was concentrated under reduced
pressure to yield a
residue which was dissolved in MeCN (20 mL) and H20 (40 mL) and lyophilized to
yield 4-[(5S)-
3 -brom o-4,5-di hydroi sox azol -5-y1]-2-(1-m ethyl i mi dazol -4-y1)-N- [[4-
(trifluoromethyl)phenyl]methyl]aniline (24.61 mg, 50.72 p.mol, 14.6% yield,
98.8% purity, SFC:
Rt = 2.248, ee = 100%, [a]268D _ +140.000 (Me0H, c = 0.180 g/100 mL)) as a
white solid. 41
NMR (500 MHz, CDC13) 6 ppm 8.52 (s, 1H), 7.57 (d, J= 8.1 Hz, 2H), 7.49 (d, J=
10.2 Hz, 3H),
7.39 (d, J= 2.0 Hz, 1H), 7.23 (s, 1H), 7.00 (dd, J= 2.1, 8.5 Hz, 1H), 6.49 (d,
J= 8.4 Hz, 1H), 5.57
(t, J= 10.2 Hz, 1H), 4.56 (s, 2H), 3.77 (s, 3H), 3.50 (dd, J= 10.7, 17.4 Hz,
1H), 3.23 (dd, J= 9.8,
17.3 Hz, 1H); ES-LCMS nilz 478.9, 480.9 [M+Hr. Peak 2 was concentrated under
reduced
pressure to yield a residue which was dissolved in MeCN (20 mL) and H20 (40
mL) and
lyophilized to yield 4-[(5R)-3-bromo-4,5-dihydroisoxazol-5-y1]-2-(1-
methylimidazol-4-y1)-N-[[4-
(trifluoromethyl)phenyl]methyl]aniline (24.58 mg, 50.61 p.mol, 14.6% yield,
98.7% purity, SFC:
148
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Rt = 3.301, ee = 100%, [a]26'8D = -198.71 (Me0H, c = 0.155 g/100 mL)) as a
white solid. 1-1-1NMR
(500 MHz, CDC13) 6 ppm 8.54 (s, 1H), 7.58-7.55 (m, 2H), 7.49 (d, J= 9.2 Hz,
3H), 7.39 (d, J=
2.0 Hz, 1H), 7.23 (s, 1H), 7.00 (dd, J= 2.0, 8.4 Hz, 1H), 6.49 (d, J= 8.5 Hz,
1H), 5.57 (t, J= 10.3
Hz, 1H), 4.56 (s, 2H), 3.77 (s, 3H), 3.50 (d.dõ/ = 10.8, 17.3 Hz, 1H), 3.23
(d.dõ/ = 9.8, 17.3 Hz,
1H); ES-LCMS nilz 478.9, 480.9 [M+E-1] .
1-21 and 1-22 (isomers of 1-30)
Br CI CI t
aq HCI
H
0 N
N
0,
Thsl-Sss CF, CF 3 N CF,
H 0 H
Step 1: (S)-5-(3-Chloro-4,5-dihydroisoxazol-5-y1)-N-methy1-6-04-
(trifluoromethyl)phenyl)amino)pyridine-3-sulfonamide
CI ________________________________________ ,1,0
0
N
N CF3
H
1004441 To a solution of 5-(3-bromo-4,5-dihydroisoxazol-5-y1)-N-methyl-6-[4-
(trifluoromethyl)anilino]pyridine-3-sulfonamide (185 mg, 366.70 [tmol, 95%
purity, 1 eq) in 1,4-
dioxane (10 mL) was added HC1 (4 M, 0.5 mL). The mixture was stirred at 40 C
for 16 h. The
solvent was removed and the residue was treated with water (10 mL), adjusted
to pH 8 with sat.
aq. NaHCO3 and extracted with Et0Ac (20 mL x 2). The combined organic layers
were washed
with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced
pressure to yield
a residue which was purified by preparative TLC (PE/Et0Ac = 1/1, Rf = 0.71) to
yield a product
which was separated by chiral SFC (column: DAICEL CH1RALPAK
AD(250mm*30mm,10um);
mobile phase: 10.1%NH3H20 EtOHLF3%: 35%-35%) to yield Peak 1 and Peak 2. Peak
1 was
concentrated under reduced pressure to yield a residue which was dissolved in
MeCN (10 mL) and
H20 (20 mL) and lyophilized to yield (S)-5-(3-chloro-4,5-dihydroisoxazol-5-y1)-
N-methy1-64(4-
(trifluoromethyl)phenyl)amino)pyridine-3-sulfonamide (43.65 mg, 97.09 mot,
26.5% yield,
96.7% purity, SFC: Rt = 1.279, ee = 99.4%, [c]2.4.4D _ _24.24 (Me0H, c =
0.0825 g/100 mL) as a
white solid. 1H NMR (500 MHz, CDC13)6 ppm 8.71 (d, J= 2.4 Hz, 1H), 7.85 (d, J=
2.3 Hz, 1H),
149
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
7.70-7.68 (m, 3H), 7.64-7.58 (m, 2H), 5.84 (t, J= 11.2 Hz, 1H), 4.41 (br s,
1H), 3.62-3.41 (m, 2H),
2.71 (d, J= 5.3 Hz, 3H); ES-LCMS nilz 435.0 [M+H]t
1-23 and 1-24 (isomers of 1-31)
Br Br
CI YaC
N N
H2N NTL:1),' _______ 0 TFA
FNII N",
NaH, DMF CF3 PelOPPOCI, ,\\S N
CF, CF3 Sµs N
Cs,CO, -N
0 I 0 0
FMB PMB PMB
1
Br
BBr f
orr N
SFC
NaHCO, N N
0 I
N
CF, UcF,
H
H
Step 1: 5-Bromo-N-1(4-m ethoxyphenyl)m ethyl] -N-m ethy1-6-115-
(trifluoromethyl)-2-
pyridyllaminolpyridine-3-sulfonamide
Br
0
.\\
,S C F3
N
I 0
PMB
1004451 To a solution of 5-(trifluoromethyl)pyridin-2-amine (91.11 mg,
562.00 nmol, 1.5 eq)
in DMF (3 mL) was added NaH (59.94 mg, 1.50 mmol, 60% purity, 4 eq) and the
mixture was
stirred at 0 C for 0.5 h. 5-Bromo-6-chloro-N-[(4-methoxyphenyl)methyl]-/V-
methyl-pyridine-3-
sulfonamide (160 mg, 374.67 nmol, 95% purity, 1 eq) was added and the mixture
was stirred at
25 C for 3 h. The reaction mixture was diluted with H20 (20 mL) and extracted
with Et0Ac (40
mL x 3). The combined organic phases were washed with brine (20 mL), dried
over anhydrous
Na2SO4, filtered and concentrated under reduced pressure to yield a residue.
To the residue was
added Me0H (5 mL) and the mixture was stirred at 25 C for 2 h. The slurry was
filtered and the
cake was rinsed with Me0H (3 mL x 2). The solid was collected and dried in
vacuo to yield 5-
bromo-N-[(4-methoxyphenyl)methy1]-N-methy1-6-1115-(trifluoromethyl)-2-
pyridyl]amino]pyridine-3-sulfonamide (160 mg, 301.12 nmol, 80.8% yield, 100.0%
purity) as a
white solid. 1H NMR (400 MHz, DMSO-d6) 6 ppm 9.21 (s, 1H), 8.72 (s, 1H), 8.68
(d, J= 2.2 Hz,
1H), 8.41 (d, J= 2.2 Hz, 1H), 8.28-8.19 (m, 2H), 7.24 (d, J= 8.6 Hz, 2H), 6.93
(d, J= 8.6 Hz,
2H), 4.16 (s, 2H), 3.74 (s, 3H), 2.60 (s, 3H); ES-LCMS nilz 533.0 [M+H] .
150
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Step 2: N-1(4-MethoxyphenyOmethyll-N-methy1-6-115-(trifluoromethyl)-2-
pyridyllaminol-5-
vinyl-pyridine-3-sulfonamide
0
C F3
N
0
PMB
1004461 To a solution of 5-bromo-N-1(4-methoxyphenyl)methy1]-N-methy1-6-115-
(trifluoromethyl)-2-pyridyl]amino]pyridine-3-sulfonamide (260 mg, 489.32
[tmol, 100% purity, 1
eq) in 1,4-dioxane (6 mL) and H20 (1 mL) was added 4,4,5,5-tetramethy1-2-viny1-
1,3,2-
dioxaborolane (301.45 mg, 1.96 mmol, 331.99 p,L, 4 eq), Pd(dppf)C12 (35.80 mg,
48.93 litmol, 0.1
eq) and Cs2CO3 (318.86 mg, 978.64 [tmol, 2 eq). The mixture was stirred under
N2 atmosphere at
90 C for 12 h. The reaction mixture was diluted with H20 (20 mL) and
extracted with Et0Ac (40
mL x 3). The combined organic phases were washed with brine (20 mL), dried
over anhydrous
Na2SO4, filtered and concentrated under reduced pressure to yield a residue
which was purified by
flash silica gel chromatography (from PE/Et0Ac = 1/0 to 5/1, TLC: PE/Et0Ac =
5/1, Rr = 0.35)
to yield AT-[(4-methoxyphenyl)methyl]-AT-methy1-64[5-(trifl
uoromethyl)-2-pyridyl]amino]-5-
vinyl-pyridine-3-sulfonamide (200 mg, 409.62 limo', 83.7% yield, 98.0% purity)
as a yellow solid.
lEINMIR (400 MHz, CDC13) 6 ppm 8.69 (d, J= 2.3 Hz, 1H), 8.64 (d, J= 8.8 Hz,
1H), 8.55 (s, 1H),
8.01-7.91 (m, 2H), 7.79 (s, 1H), 7.24 (d, J= 8.8 Hz, 2H), 6.95-6.86 (m, 2H),
6.82 (dd, .1= 11.0,
17.3 Hz, 1H), 5.86 (d, J= 17.3 Hz, 1H), 5.74 (d, J= 11.0 Hz, 1H), 4.16(s, 2H),
3.81 (s, 3H), 2.66
(s, 3H); ES-LCMS nilz 479.6 [M-41] .
Step 3: N-Methyl-6-115-(trifluoromethyl)-2-pyridyllaminol-5-vinyl-pyridine-3-
sulfonamide
N N
0
N
,S CF3
N
H
[00447] To a solution of N-[(4-methoxyphenyl)methyl]-N-methyl-64[5-
(trifluoromethyl)-2-
151
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
pyridyl]amino]-5-vinyl-pyridine-3-sulfonamide (200 mg, 409.62 !amok 98%
purity, 1 eq) in DCM
(3 mL) was added TFA (1.51 g, 13.24 mmol, 980.00 L, 32.31 eq). The mixture
was stirred at 25
C for 3 h. The solvent was removed to yield N-methy1-64[5-(trifluoromethyl)-2-
pyridyl]amino]-
5-vinyl-pyridine-3-sulfonamide (140 mg, crude) as a yellow solid. I-HNIVIR
(400 MHz, CDC13) (-5
ppm 8.71 (d, J¨ 2.0 Hz, 1H), 8.52 (s, 1H), 8.37 (d, J¨ 8.8 Hz, 1H), 8.28 (s,
1H), 8.19 (d, J¨ 7.1
Hz, 1H), 6.75 (s, 1H), 6.61 (s, 1H), 5.94 (d, J= 16.9 Hz, 1H), 5.78 (d, J=
11.0 Hz, 1H), 3.99 (s,
3H); ES-LCMS m/z 359.2 [M+H].
Step 4: 5-1(5S)-3-Bromo-4,5-dihydroisoxazol-5-y11-N-methy1-6-R5-
(trifluoromethyl)-2-
pyridyllaminolpyridine-3-sulfonamide and 5-1(5R)-3-bromo-4,5-dihydroisoxazol-5-
y11-N-
methyl-641-5-(trifluoromethyl)-2-pyridyllaminolpyridine-3-sulfonamide
Br
0
H
N N
0
õS CF3
N
H 0
1004481 To a solution of N-methy1-64[5-(trifluoromethyl)-2-
pyridyl]amino]-5-vinyl-pyridine-
3-sulfonamide (140 mg, 390.69 [tmol, 1 eq) in Et0Ac (10 mL) was added NaHCO3
(328.22 mg,
3.91 mmol, 151.95 L, 10 eq) and dibromomethanone oxime (158.49 mg, 781.38
ttmol, 2 eq). The
mixture was stirred at 25 C for 6 h. The mixture was filtered and the
filtrate was concentrated to
yield a residue which was purified by flash silica gel chromatography (from
PE/Et0Ac = 1/0 to
1/1, TLC: PE/Et0Ac = 1/1, RE = 0.32) to yield a product which was separated by
chiral SFC
(column: DAICEL CHIRALPAK AD(250mm*30mm,10um);mobile phase: [0.1%NH3H20
Me0H]; B%: 55%-55%) to yield Peak 1 and Peak 2. Peak 2 was concentrated under
reduced
pressure to yield 5-[(5R)-3-bromo-4,5-dihydroisoxazol-5-y1]-N-methy1-64[5-
(trifluoromethyl)-2-
pyridyl]amino]pyridine-3-sulfonamide (39.4 mg, 81.48 mot, 20.9% yield, 99.3%
purity, SFC:
= 4.427, ee = 99.9%, [cd245D = +39.22 (Me0H, c = 0.051 g/100 mL) as a white
solid. 11-I NMR
(500 MHz, DMSO-d6) 6 ppm 9.73 (s, 1H), 8.68-8.55 (m, 2H), 8.14-8.07 (m, 1H),
8.06-7.98 (m,
2H), 7.63 (q, J= 4.8 Hz, 1H), 6.19 (dd, J= 7.9, 11.0 Hz, 1H), 3.92 (dd, J=
11.0, 17.5 Hz, 1H),
3.43-3.39 (m, 1H), 2.45 (d, J= 4.9 Hz, 3H); ES-LCMS miz 480.1 [M+H]t
152
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1-33 and 1-34 (isomers of 1-32)
Br ryCF3 Br
H11,1
TAN'OH
R\
2*--C"")
=1.410.
Pd(cippf)C12 N44.0
Br'
Br
NaHCO,
''CF3 =
H 0 \\O Cs2C10
H
Br Brt 11 o Br
¨N ¨
0
Chiral SFC
ENI
'CD
H 0 H 0 H
Step 1: 3-Bromo-N-methyl-4-114-
(trifluoromethyl)cyclohexyllaminolbenzenesulfonamide
Br
0\\ N4..0,,
CF3
N
H 0
1004491 To a solution of 4-(trifluoromethyl)cyclohexanamine (748.24 mg, 4.48
mmol, 1.5 eq)
in DMSO (25 mL) was added 3-bromo-4-fluoro-N-methyl-benzenesulfonamide (800
mg, 2.98
mmol, 100.0% purity, 1 eq). The reaction mixture was stirred under N2
atmosphere at 140 C for
3 h. The reaction mixture was diluted with H20 (30 mL) and extracted with
Et0Ac (60 mL x 3).
The combined organic layers were washed with brine (50 mL), dried over Na2SO4,
filtered and
concentrated under reduced pressure to yield a residue which was purified by
flash silica gel
chromatography (from PE/Et0Ac = 100/1 to 3/1, TLC: PE/Et0Ac = 3/1, Rf = 0.36)
to yield the
crude product which was further purified by preparative HPLC (column: Boston
Green ODS
150*30mm*5[1.m; mobile phase: [water (0.05% HC1)-ACN]; B%: 53%-83%, 10 min) to
yield 3-
bromo-N-methy1-44[4-(trifluoromethyl)cyclohexyliamino]benzenesulfonamide (250
mg, 571.92
mmol, 19.2% yield, 95.0% purity) as a white solid. 1-E1 NMIR (500 MHz, CDC13)
6 ppm 7.92 (d, J
= 2.1 Hz, 1H), 7.65 (dd, J= 2.1, 8.7 Hz, 1H), 6.65 (d, J= 8.7 Hz, 1H), 4.68
(s, 1H), 4.20 (s, 1H),
3.38-3.34 (m, 1H), 2.71-2.58 (m, 3H), 2.27 (d, J= 11.0 Hz, 2H), 2.15-2.03 (m,
3H), 1.55-1.47 (m,
2H), 1.34-1.25 (m, 2H); ES-LCMS nilz 415.2, 417.2 [M-41] .
Step 2: N-Methyl-4-114-(trifluoromethyl)cyclohexyllaminol-3-vinyl-
benzenesulfonamide
153
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
CI`µ
,S
N
H0
1004501 To a solution of
3-bromo-N-methy1-44[4-
(trifluoromethyl)cyclohexyl]amino]benzenesulfonamide (210 mg, 441.64 mol, 1
eq, HC1) and
4,4,5,5-tetramethy1-2-vinyl-1,3,2-dioxaborolane (102.03 mg, 662.45 mol, 112.36
[iL, 1.5 eq) in
1,4-dioxane (6 mL) and H20 (2 mL) was added Pd(dppf)C12 (32.31 mg, 44.16
p,mol, 0.1 eq) and
Cs2CO3 (359.73 mg, 1.10 mmol, 2.5 eq). The mixture was stirred under N2
atmosphere at 100 C
for 2 h. The mixture was diluted with water (30 mL) and extracted with Et0Ac
(30 mL x 3). The
organic layer was washed with brine (30 mL x 2), dried over Na2SO4, filtered
and concentrated
under reduced pressure to yield a residue which was purified by flash silica
gel chromatography
(from PE/Et0Ac = 100/1 to 1/1, TLC: PE/Et0Ac = 1/1, Rf = 0.60) to yield N-
methy1-44[4-
(trifluoromethyl)cyclohexyl]amino]-3-vinyl-benzenesulfonamide (160 mg, 435.75
tmol, 98.7%
yield, 98.7% purity) as yellow oil. N1VIR (400 MHz, CDC13) 6 ppm 7.72-7.56
(m, 2H), 6.71-
6.54 (m, 2H), 5.67 (d, J= 17.4 Hz, 1H), 5.44 (d, J= 11.0 Hz, 1H), 4.20-4.14
(m, 1H), 4.12 (d, J=
7.1 Hz, 1H), 3.35 (m, J= 4.0, 7.4, 11.2 Hz, 1H), 2.64 (d, J= 5.4 Hz, 3H), 2.26
(d, J= 12.0 Hz,
2H), 2.08 (s, 1H), 2.04 (s, 2H), 1.54-1.42 (m, 2H), 1.26-1.22 (m, 2H); ES-LCMS
nilz 363.2
[M+H] .
Step 3: 3-1(5S)-3-Bromo-4,5-dihydroisoxazol-5-yll-N-methyl-4-114-
(trifluoromethyl)cyclohexyllaminolbenzenesulfonamide
Br
H
cz\
,S
N
H
1004511 To a solution of N-methy1-44[4-(trifluoromethypcyclohexyl]amino]-3-
vinyl-
benzenesulfonamide (110 mg, 297.45 p,mol, 1 eq) and in Et0Ac (5 mL) was added
dibromomethanone oxime (90.50 mg, 446.18 limo', 1.5 eq) and NaHCO3 (249.88 mg,
2.97 mmol,
eq). The mixture was stirred at 40 C for 12 h. TLC (PE/Et0Ac = 1/1, Rf =
0.72) showed the
starting material was consumed completely and one new spot was detected. The
mixture was
154
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
diluted with water (50 mL) and extracted with Et0Ac (50 mL x 3). The organic
layer was washed
with brine (50 mL x 2), dried over Na2SO4, filtered and concentrated under
reduced pressure to
yield a residue which was purified by flash silica gel chromatography (from
PE/Et0Ac = 100/1 to
1/1, TLC: PE/Et0Ac = 1/1, Rf = 0.60) and by preparative SFC (column: DAICEL
CHIRALPAK
AD (250 mm*30 mm, 10 um); mobile phase: [0.1% NH3=1120 ETOH]; B%: 25%-25%) to
yield
Peak 1 and Peak 2. Peak 1 was concentrated under reduced pressure to yield a
residue which was
dissolved in MeCN (20 mL) and H20 (20 mL) and lyophilized to yield 3-[(55)-3-
bromo-4,5-
dihydroisoxazol-5-y1]-N-methy1-4-[[4-
(trifluoromethyl)cyclohexyl]amino]benzenesulfonamide
(60 mg, 122.40 imol, 41.1% yield, 98.8% purity, SFC: Rt = 1.414, ee = 100%,
[GL]24.4D _ _23.07
(Me0H, c = 0.026 g/100 mL)) as a yellow solid. 1-E1 NMR (400 MHz, CDC13) 6 ppm
7.71 (d, J =
8.6 Hz, 1H), 7.55 (s, 1H), 6.75 (d, J = 8.8 Hz, 1H), 5.66 (t, J = 11.2 Hz,
1H), 4.60 (d, J = 7.1 Hz,
1H), 4.22 (q, J= 4.9 Hz, 1H), 3.55-3.40 (m, 2H), 3.38-3.29 (m, 1H), 2.66-2.61
(m, 3H), 2.24 (s,
2H), 2.07 (d, = 12.5 Hz, 3H), 1.53-1.44 (m, 2H), 1.29-1.17 (m, 2H); ES-LCMS
m,/z 486.0
[M+H] .
Example 2. TEAD inhibition Assay
[00452] TEAD inhibition can be assayed using Hippo Pathway TEAD Reporter ¨
MCF7 Cell
Line (BPS Bioscience, Catalog #: 60618).
Background
[00453] The Hippo pathway regulates cell proliferation and cell death. It is
activated by high
cell density and cell stress to stop cell proliferation and induce apoptosis.
The mammalian Hippo
pathway comprises MST kinases and LATS kinases. When the Hippo pathway is
activated, MST
kinases phosphorylate LATS kinases, which phosphorylate transcriptional co-
activators YAP and
TAZ. Unphosphorylated YAP and TAZ can translocate to the nucleus and interact
with
TEAD/TEF transcriptional factors to turn on cell cycle-promoting gene
transcription. However,
when phosphorylated, YAP and TAZ are recruited from the nucleus to the
cytosol, so that the YAP
and TAZ-dependent gene transcription is turned off. Dysfunction of the Hippo
pathway is
frequently detected in human cancer and its down-regulation correlates with
the aggressive
properties of cancer cells and poor prognosis.
Description
[00454] The TEAD Reporter ¨ MCF7 cell line contains the firefly luciferase
gene under the
155
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
control of TEAD responsive elements stably integrated into the human breast
cancer cell line,
MCF7. Inside the cells, basal unphosphorylated YAP/TAZ remains in the nucleus
and induces the
constitutive expression of luciferase reporter. The cell line is validated for
the inhibition of the
expression of luciferase reporter by the activators of the Hippo pathway.
Application
= Monitor Hippo pathway activity.
= Screen for activators or inhibitors of the Hippo pathway.
Format
1004551 Each vial contains ¨1.5 X 106ce11s in 1 ml of 10% DMSO.
Storage
1004561 Immediately upon receipt, store in liquid nitrogen.
General Culture Conditions
1004571 Thaw Medium 1 (BPS Bioscience #60187) + 10 pg/ml of Insulin (Sigma-
Aldrich #
10516): MEM medium (Hyclone #SH30024.01) supplemented with 10% FBS (Invitrogen
#26140-
079), 1% non-essential amino acids (Hyclone #SH30238.01), 1 mM Na pyruvate
(Hyclone
#SH30239.01), 1% Penicillin/Streptomycin (Hyclone SV30010.01), plus 10 ug/m1
of insulin
(Sigma-Aldrich # 10516)
1004581 Growth Medium 1B (BPS Bioscience #79531) + 10 pg/ml of Insulin (Sigma-
Aldrich # 10516): Thaw Medium 1 (BPS Cat. #60187) + 10 jig/m1 of insulin
(Sigma-Aldrich #
10516), and 400 ug/m1 of Geneticin (Invitrogen #11811031).
1004591 Cells should be grown at 37 C with 5% CO2 using Growth Medium 1B with
10 ug/m1
of Insulin. It may be necessary to adjust the percentage of CO2 in the
incubator depending on the
NaHCO3 level in the basal medium.
1004601 To thaw the cells, it is recommended to quickly thaw the frozen cells
from liquid
nitrogen in a 37 C water-bath, transfer to a tube containing 10 ml of Thaw
Medium 1 + Insulin (no
Geneticin), spin down cells, resuspend cells in pre-warmed Thaw Medium 1 +
Insulin (no
Geneticin), transfer resuspended cells to a T25 flask and culture in a CO2
incubator at 37 C
overnight. The next day, replace the medium with fresh Thaw Medium 1 + Insulin
(no Geneticin),
and continue growing culture in a CO2 incubator at 37 C until the cells are
ready to be split. At
first passage, switch to Growth Medium 1B + 10 ps/m1 of Insulin (includes Thaw
Medium 1,
Insulin, and Geneticin). Cells should be split before they reach complete
confluence.
156
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
1004611 To passage the cells, rinse cells with phosphate buffered
saline (PBS), and detach cells
from the culture vessel with 0.25% Trypsin/EDTA. Add Growth Medium 1B + 10
mg/m1 of Insulin
(Includes Thaw Medium 1, Insulin, and Geneticin) and transfer to a tube, spin
down the cells, then,
resuspend cells and seed appropriate aliquots of cell suspension into new
culture vessels.
Subcultivation ration: 1:5 to 1:10 weekly.
[00462] To freeze down the cells, rinse cells with phosphate buffered saline
(PBS), and detach
cells from culture vessel with Trypsin/EDTA. Add Growth Medium 1B + 10 iig/m1
of Insulin
(Includes Thaw Medium 1, Insulin, and Geneticin) and transfer to a tube, spin
down cells, and
resuspend in freezing medium (10% DMSO + 90% FBS). Place at -80 C overnight
and place in
liquid nitrogen the next day. Alternatively, vials may be placed directly in
liquid nitrogen.
Functional Validation and Assay Performance
[00463] The following assays are designed for 96-well format. To perform the
assay in different
tissue culture formats, the cell number and reagent volume should be scaled
appropriately.
Materials Required but Not Supplied for Cell Culture
= Thaw Medium 1 (BPS Bioscience #60187) + 10 mg/m1 of insulin
= Growth Medium 1B (BPS Bioscience #79531) + 10 itg/ml of insulin
= Insulin Solution from Bovine Pancreas (Sigma-Aldrich #: 10516)
Materials Required but Not Supplied for Cellular Assay
= H202: activator of Hippo pathway (activate MST kinases)
= Insulin
= Assay Medium: Thaw Medium 1 (BPS Cat. #60187) + 10 [ig/m1 of insulin
= Insulin Solution from Bovine Pancreas (Sigma-Aldrich Cat#: 10516)
= Okadaic acid (BPS bioscience #27047): activator of Hippo pathway
(activate MST
kinases). Prepare 10 mM stock in DMSO.
= 96-well tissue culture plate or 96-well tissue culture-treated white
clear-bottom assay plate
= ONE-StepTM Luciferase Assay System (BPS, Cat. #60690)
= Luminometer
Mycoplasma testing
[00464] The cell line has been screened using the PCR-based VenorGeM
Mycoplasma
Detection kit (Sigma-Aldrich) to confirm the absence of Mycoplasma species.
157
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
Inhibition of TEAD reporter activity by activator of Hippo pathway in TEAD
Reporter ¨
MCF7 cells
1) Harvest TEAD Reporter ¨ MCF7 cells from culture in growth medium and seed
cells at a
density of 35,000 cells per well into white clear-bottom 96-well microplate in
45 [1.1 of assay
medium.
2) Incubate cells at 37 C in a CO2 incubator for overnight.
3) Dilute the activators (H202 or okadaic acid) stock in assay medium. Add 5
1 of diluted
activators to the wells. The final concentration of DMSO in assay medium is
0.1%.
4) Add 5 tl of assay medium with same concentration of DMSO without activator
to control
wells.
5) Add 50 pi of assay medium with DMSO to cell-free control wells (for
determining background
luminescence).
6) Set up each treatment in at least triplicate.
7) Incubate cells at 37 C in a CO2 incubator for 5-6 hours.
8) Perform luciferase assay using the ONE-StepTm Luciferase Assay System
following the
protocol provided: Add 100 IA of ONE-StepTM Luciferase reagent per well and
rock at room
temperature for ¨15 minutes. Measure luminescence using a luminometer.
9) Data Analysis: Obtain the background-subtracted luminescence by subtracting
the average
background luminescence (cell-free control wells) from the luminescence
reading of all wells.
[00465] Certain compounds were tested in TEAD reporter assay, and in H226 and
H28. The
data are listed in Table 2 below. A: EC50 <0.1 uM; B: 0.1 uM < EC50 < 0.5 uM;
C: EC50 > 0.5
uM.
Table 2. In vitro Data of Certain Exemplary Compounds.
I# H226 EC50 (uM) TEAD Reporter Assay EC50 (uM)
1
2
3
4
6
158
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
7
8 A A
9 A A
isomer 1 of 1-25 A
Isomer 2 of 1-25 C
isomer 1 of 1-26 A A
Isomer 2 of 1-26 B
14 A A
isomer 1 of 1-27 B
Isomer 2 of 1-27 C
isomer 1 of 1-28 B
Isomer 2 of 1-28 C
isomer 1 of 1-29 A
isomer 2 of 1-29 B
isomer 1 of I-30 A
isomer 2 of 1-30 C
isomer 1 of 1-31 C
isomer 2 of 1-31 B
isomer 1 of 1-32 C A
isomer 2 of 1-32 B
Example 3: Mouse Pharmacokinetics Study
1004661 Formulated compounds are administered intravenously or orally via
gavage to BALB/c
mice. Typically, at 0.167, 0.5, 1, 2, 4, 6, 12, and 24 hours post-dose, blood
is collected and
processed to plasma by centrifugation and stored at -80 C until analysis.
Internal standard is added
to each sample prior to protein precipitation with acetonitrile or TCA. The
precipitates are filtered
through a filter plate and the samples are analyzed by LC/MS/MS. A standard
curve is prepared in
plasma from typically from 1.0 ng/mL to 3000 ng/mL and processed in the same
manner as the
samples. Sample analysis is typically performed on a suitable LC/MS/MS system
fitted with an
analytical UPLC column and compounds eluted from the analytical column with a
gradient from
30-95% 0.1 % formic acid (v/v) in ACN: 0.1 % formic acid (v/v) in water. Mass
spectrometric
159
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
detection of test compound and the internal standard is performed by MRM in
positive mode. The
pharmacokinetics of each compound are analyzed by Phoenix WinNonlin software
(Pharsight, St.
Louis, MO) via noncompartmental analysis.
Example 4. CTGF Data Analysis
[00467] NU/NU nude female mice are obtained from Charles River Laboratories
and
subcutaneously injected with NCI-H226 (ATCC) human mesothelioma cells. Once
tumors grow
to an average size of 350-400 mm3, mice are randomized into each treatment
group. NCI-H226
tumor bearing mice are treated by oral gavage with Vehicle (5%DMS0/95% PEG
400) or a TEAD
inhibitor for a total of 3 administrations. 4 hours post-third administration,
mice are euthanized
and tumors collected for isolation of RNA for pharmacodynamic (PD) analysis.
[00468] RNA is extracted from the tumors utilizing the QIAZOL (Qiagen) lysis
reagent, tissues
are then homogenized for 10 minutes using TissueLyser II (Qiagen). Once sample
disruption and
digestion is complete, chloroform is added to each sample, the homogenate is
separated into
aqueous and organic phases by centrifugation.
1004691 RNA is then isolated from samples using the KingFisher Flex automated
extraction
system and MagMAX mirvana total RNA isolation kit. Manufacturer's recommended
protocol
for high-throughput isolation of RNA from tissue samples is followed for RNA
extraction.
[00470] Expression of the YAP/TEAD-regulated gene, CCN2 that encodes CTFG
(Connective
Tissue Growth Factor), and the housekeeping gene, human glyceraldehyde 3-
phosphate
dehydrogenase (GAPDH), are quantified by qRT-PCR analysis using the TaqMan
Gene
Expression Master Mix and TaqMan probes. CTGF and GAPDH cycle threshold (Ct)
values for
tumor cDNA samples are determined, and CTGF expression is normalized to GAPDH
as an
internal control.
[00471] The relative CTGF mRNA expression levels for each treatment group from
tumor
tissues are normalized to the vehicle control group. For comparisons between
vehicle control and
TEAD inhibitor treatment groups, an independent sample t-test is used for
statistical analysis.
Example 5. Anti-Proliferation Assay
1004721 Individual cell lines are grown in medium according to supplier
instructions and seeded
into 96-well plates at a density that ensures logarithmic growth over 72-96
hours. lEAD inhibitor
160
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
compounds are administered to cells at a top concentration of 10 lam and
subsequently a 10 point
3-fold serial dilution is conducted. After 72-96 hours, proliferation is
quantified using Cell
TITERGLOTm (Promega, Inc.) and compared to vehicle control. IC50 and EC50
values are
generated using Prism or XLFit curve fitting software.
Example 6. In vivo Inhibition of Tumor Growth
NCI-H226 in vivo Efficacy Studies
1004731 6-8 week old nu/nu nude mice (CRL) are inoculated subcutaneously with
5 x106 NCI-
H226 human mesothelioma tumor cells in the right flank. Tumor growth is
monitored twice per
week using vernier calipers and mean tumor volume (MTV) calculated using the
formula V= W2
x L/2.
1004741 When the MTV reaches approximately 150-200 mm3, animals are randomized
into
treatment groups (n=8-10/group) and dosed per os (PO) on a once everyday (QD)
schedule for 27-
40 days with either Vehicle (5% DMSO + 95% PEG 400) or TEAD inhibitors.
1004751 Randomization and treatments start on Day 0 and % Tumor Growth
Inhibition is
calculated on the last day of the study (when the control MTV reaches maximum
allowable tumor
volume), and the following calculation is performed.
1004761 %TGI= 100 ¨ [MTV treated / MTV control] x 100
1004771 Tumor growth and body weight change are measured twice per week.
1004781 For comparisons between vehicle control and TEAD inhibitor treatment
groups, an
independent sample t-test is used for statistical analysis.
MSTO-211H in vivo efficacy studies
1004791 6-8 week old SCID mice (CRL) are inoculated subcutaneously with 5 x106
MSTO-
211H human mesothelioma tumor cells in the right flank. Tumor growth is
monitored twice per
week using vernier calipers and mean tumor volume (MTV) is calculated using
the formula V=
W2 x L/2.
1004801 When the MTV reached approximately 150-200 mm3, animals are randomized
into
treatment groups (n=6-8/group) and dosed per os (PO) on a once everyday (QD)
schedule for 22-
25 days with either Vehicle (5% DMSO + 95% PEG 400) or TEAD inhibitors.
161
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
[00481] Randomization and treatments started on Day 0 and % Tumor Growth
Inhibition is
calculated on the last day of the study (when the control MTV reaches maximum
allowable tumor
volume), the following calculation is performed.
[00482] %TGI= 100 ¨ [MTV treated / MTV control] x 100
[00483] Tumor growth and body weight change are measured twice per week.
[00484] For comparisons between vehicle control and TEAD inhibitor treatment
groups, an
independent sample t-test is used for statistical analysis.
Example 7. TEAD Selectivity Assays
[00485] The TEAD targeting selectivity profiles of the TEAD inhibitor
compounds described
herein can be determined by any of the exemplary assays provided herein
designed to monitor the
interaction of TEAD isoforms or variants, e.g., human TEAD1 (UniProt KB ID
P28347-1 (SEQ
ID NO: 1)), human TEAD2 (UniProtKB ID Q15562 (SEQ ID NO: 2)), human TEAD3
(UniProtKB ID Q99594 (SEQ ID NO: 3)), and human TEAD4 (UniProtKB ID Q15561
(SEQ ID
NO: 4), and YAP1 or TAZ. While co-immunoprecipitation techniques can be used
to monitor
protein-protein interactions, it is difficult to increase the throughput based
on the basic
methodology required. Accordingly, alternative but complementary assays are
employed to
monitor the interaction of the different TEAD isoforms or variants, e.g.,
human TEAD1 (UniProt
KB ID P28347-1 (SEQ liD NO: 1)), human TEAD2 (UniProtKB ID Q15562 (SEQ ID NO:
2)),
human TEAD3 (UniProtKB ID Q99594 (SEQ ID NO: 3)), and human TEAD4 (UniProtKB
ID
Q15561 (SEQ ID NO: 4), and YAP1 (or TAZ).
[00486] The first exemplary assay is an in vitro biochemical
fluorescent polarization assay
using recombinantly expressed and purified YAP-binding domains of individual
TEAD isoforms
and a fluorescently labeled peptide derived from the primary sequence of YAP1.
(Bum-Erdene et
al., Cell Chem Biol. 2019 Mar 21;26(3):378-389.e13, the contents of which are
herein
incorporated by reference in their entireties). Compounds are incubated with
individual TEAD
isoform proteins and the fluorescent peptide and potency is determined by
quantifying the
displacement of the peptide.
[00487] The second exemplary assay is a cell-based assay employing the split
luciferase
reporter system (Hall et al., ACS Chem. Biol. 2012, 7, 11, 1848-1857, the
contents of which are
herein incorporated by reference in their entireties). Briefly, the YAP-
binding domain of each
162
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
TEAD isoform is transiently co-expressed with the TEAD-binding domain or
either YAP1 or TAZ
in HEK293 cells and the proximity of the two chimeric gene fusion products is
monitored by
luciferase activity (Noun i et at. Cancers (Basel). 2019 Oct 19;11(10), the
contents of which are
herein incorporated by reference in their entireties). Compounds that
interfere with the interaction
of a TEAD isoform and YAP1 (or TAZ) decrease the resulting luciferase activity
relative to vehicle
treated controls. Similar in process to the fluorescent polarization assay,
these chimeric gene
fusions are recombinantly expressed in bacteria or insect cells and employed
as an in vitro
biochemical assay with a similar luciferase readout as the cell-based assay.
1004881 Another exemplary assay is a thiol conjugation assay that monitors
prevention of
covalent binding of a fluorescence turn-on probe to a cysteine in the central
pocket of TEAD
isoforms by small molecules, as described in Karatas et at., "Discovery of
Covalent Inhibitors
Targeting the Transcriptional Enhanced Associate Domain (TEAD) Central
Pocket," J. Med.
Chem. 2020, the contents of which are herein incorporated by reference in
their entireties. Briefly,
the thiol reactive pro-fluorescent probe, N-(4-(7-diethylamino-4-
methylcoumarin-3-y1) phenyl)
maleimide (CPM) is used. The fluorescence in CPM is quenched due to the
maleimide substitution
on the phenyl group that modulates the resonance between the coumarin carbonyl
and 7-amino
groups. However, upon reaction with a thiol, CPM fluorescence increases
strongly. CPM can be
used to investigate TEAD inhibition because reaction of a free cysteine
residue in the TEAD
central pocket yields a fluorescence signal, such that small molecules that
bind to the TEAD central
pocket with appreciable potency prevents covalent labeling of the cysteine by
CPM. Thus, in thiol
conjugation assays, freshly prepared TEAD protein solutions are added and
incubated at room
temperature with test compounds in assay buffer, followed by the addition of a
CPM solution and
fluorescence is measured (Ex/Em: 380/470 nm). Any test compounds that bind the
TEAD central
pocket with appreciable potency show less fluorescence or inhibition of
fluorescence relative to
compounds that do not bind the TEAD central pocket.
Example 8. Inhibition of malignant mesothelioma tumor cell growth
1004891 The tumor cell growth inhibitory activity of the TEAD inhibitors
described herein is
evaluated in NCI-H2052 mesothelioma cell line harboring a NF2 mutation. This
cell line is
selected, in part, based on its mutational status and the ability of a siRNA
directed against YAP,
TAZ or TEAD1-TEAD4 to inhibit cell proliferation. The nuclear localization of
YAP at
163
CA 03200814 2023- 5- 31

WO 2022/120354
PCT/US2021/072684
confluence is also taken into account. 10,000 cells/well are plated in a 96-
well black plate with
clear flat bottom TC-Treated Imaging plate in regular medium with serum, which
is replaced the
day after with starvation medium containing 1% serum. After one day growth in
the starvation
medium, cells are incubated with TEAD inhibitor compounds. The starting
concentration is 30 ILIM
and serial dilutions in DMSO and medium are performed until 0.1 [tM to achieve
a final DMSO
concentration of 0.5%. The cells are then allowed to grow for 3 days, and
then, EdU (Invitrogen,
Molecular Probe) is added in each well at a final concentration of 10 mM and
the cells are returned
to the incubator for an additional 24h. The starvation medium is removed and
100 [1.1 of PFA 4%
containing Hoechst dye is added in each well to fix the cells. Plates are then
incubated at room
temperature for 15 min, washed twice with PBS, and the cells permeabilized by
adding 100 pi per
well of triton-100 containing 0.3% BSA. After 20 min, cells are washed with
PBS and EdU
detection is performed according to the instructions of the manufacturer.
Image acquisition is
performed, for example, using the ImageXpress Micro and analyzed using the
MetaXpress
software (Molecular Device).
1004901 While we have described a number of embodiments of this invention, it
is apparent that
our basic examples may be altered to provide other embodiments that utilize
the compounds and
methods of this invention. Therefore, it will be appreciated that the scope of
this invention is to
be defined by the application and appended claims rather than by the specific
embodiments that
have been represented by way of example.
164
CA 03200814 2023- 5- 31

Representative Drawing

Sorry, the representative drawing for patent document number 3200814 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2021-12-02
(87) PCT Publication Date 2022-06-09
(85) National Entry 2023-05-31

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2023-11-27


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2024-12-02 $125.00
Next Payment if small entity fee 2024-12-02 $50.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $421.02 2023-05-31
Maintenance Fee - Application - New Act 2 2023-12-04 $100.00 2023-11-27
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
IKENA ONCOLOGY, INC.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Declaration of Entitlement 2023-05-31 1 17
Voluntary Amendment 2023-05-31 18 331
Description 2023-05-31 164 7,146
Patent Cooperation Treaty (PCT) 2023-05-31 1 49
Claims 2023-05-31 20 410
Drawings 2023-05-31 1 55
International Search Report 2023-05-31 8 209
Patent Cooperation Treaty (PCT) 2023-05-31 1 63
Correspondence 2023-05-31 2 47
Abstract 2023-05-31 1 4
National Entry Request 2023-05-31 8 223
Cover Page 2023-09-01 1 25
Claims 2023-06-01 17 313

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

No BSL files available.