Language selection

Search

Patent 3151322 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 3151322
(54) English Title: COMBINATION THERAPY WITH IMMUNE STIMULATORY CONJUGATES
(54) French Title: POLYTHERAPIE COMPRENANT DES CONJUGUES IMMUNOSTIMULANTS
Status: Compliant
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 47/68 (2017.01)
  • A61K 45/06 (2006.01)
  • A61P 35/00 (2006.01)
(72) Inventors :
  • ODEGARD, VALERIE (United States of America)
  • BAUM, PETER R. (United States of America)
  • THOMPSON, PETER A. (United States of America)
(73) Owners :
  • SILVERBACK THERAPEUTICS, INC. (United States of America)
(71) Applicants :
  • SILVERBACK THERAPEUTICS, INC. (United States of America)
(74) Agent: ROBIC
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2020-10-01
(87) Open to Public Inspection: 2021-04-08
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2020/053870
(87) International Publication Number: WO2021/067644
(85) National Entry: 2022-03-15

(30) Application Priority Data:
Application No. Country/Territory Date
62/908,881 United States of America 2019-10-01
62/935,789 United States of America 2019-11-15
63/018,615 United States of America 2020-05-01

Abstracts

English Abstract

Combination therapies using a conjugate comprising a TLR agonist and an antibody that binds a tumor associated antigen or liver cell antigen, and a second therapeutic agent, such as an immune checkpoint inhibitor, are provided for treating cancer or viral infections. Combination therapies using a conjugate comprising a TLR agonist and an antibody that binds HER2, and a second therapeutic agent are also provided for treating cancer.


French Abstract

La présente invention concerne des polythérapies, faisant appel à un conjugué comprenant un agoniste de TLR et un anticorps qui se lie à un antigène associé à une tumeur ou à un antigène de cellule hépatique ainsi qu'un second agent thérapeutique, tel qu'un inhibiteur de point de contrôle immunitaire, qui sont destinées à traiter le cancer ou des infections virales. L'invention concerne également des polythérapies, faisant appel à un conjugué comprenant un agoniste de TLR et un anticorps qui se lie à HER2 ainsi qu'un second agent thérapeutique, qui sont destinées à traiter le cancer.

Claims

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


WO 2021/067644
PCT/1JS2020/053870
CLAIMS
WHAT IS CLAIMED IS:
1. A method of treating cancer or viral
infection, comprising administering
to a subject with cancer or viral infection a conjugate and an immune
checkpoint
inhibitor that inhibits PD-1 and/or PD-L1; wherein the conjugate is
represented by
Formula (I):
= 4> Dx
(j);
wherein:
A is an antibody that binds a tumor associated antigen or a liver cell
antigen,
L is a linker;
Dx is a TLR8 agonist, wherein the TL118 agonist is a benzazepine compound;
n is selected from 1 to 20; and
z is selected from 1 to 20.
2. The method of claim 1, wherein the immune checkpoint inhibitor is an
antibody.
3. The method of claim 1 or 2, wherein the immune checkpoint inhibitor is
an anti-PD-1 antibody or an anti-PD-L1 antibody.
4. The method of claim 3, wherein the anti-PD-1 antibody is selected from
pembrolizumab, nivolumab, cemiplimab, and biosimilars thereof.
5. The method of claim 3, wherein the anti-PD-L1 antibody is selected
from avelumab, durvalumab, atezolizumab, and biosimilars thereof.
6. The method of any one of claims 1-5, wherein n is 1 and z is from 1 to
8.
297
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
7. The method of any one of claims 1-6,
wherein the TLR8 agonist is a
compound of Category A, Formula (IIB):
R1
R2
N-R2
R5'1_1
R25
R24
R21
L2-R4
R22 R--
(IIB)
or a pharmaceutically acceptable salt thereof, wherein:
12 is -Xt -;
L2 is selected from -X2-, -x2-c 1-6 alkylene-X2-, -X2-C2-6 alkeny1ene-X2-, and
-
X2-C 2-6 alkynylene-X2-, each of which is optionally substituted on alkylene,
alkenylene
or alkynylene with one or more Rt2;
Xt is selected from -C(0)-, and -C(0)N(Rm)-*, wherein * represents where Xth
is bound to R5;
X2 at each occurrence is independently selected from a bond, -0-, -S-, -N(Rw)-
,
-C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(114 )-, -C(C)N(R10)C(0)-,
-C(0)N(Rm)C(0)N(Itn, -N(R"))C(0)-, -N(Rw)C(0)N(Rn-, -
Not tow(0)0_,
-0C(0)N(Rn-, -N(Rw)C(NRw)-, -C(NR' )N(Rt )-, -N(R4 )C(NR')N(R4 )-
, -S(0)2-, -0S(0)-, -S(0)0-, -S(0), -OS(0)2-, -S(0)20, -N(RnS(0)2-, -
S(0)2N(Rt0)-,
-N(Rm)S(0)-, -S(0)N(R1o)_, Ncr.ts. k iS(0)2N(Rw)-, and -N(Rth)S(0)N(R1 )-;
RI- and R2 are independently selected from hydrogen; and Ct-to alkyl, C2-to
alkenyl, and C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -OW ,
-C(0)N(R10)2,
-N(114- )2, -S(0)Rm, -S(0)2114- , -C(0)R1 , -C(0)0Rt , -0C(0)10 , -NO2, =0,
=S,
=N(Rw), and -CN,
R4 is selected from: -OR', -N(Rm)2, -C(0)N(Rw)2, -C(0)Rm, -C(0)0Rw,
-S(0)Rm, and -S(0)2101); C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, -Coltm, -C(0)N(R0)2, -N(Rm)C(0)Ftw,
frk.,(0)N(R10)2, -N(R10)2,
298
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
-C(0)Rm, -C(0)0R1-0, -0C(0)R", -NO2, =0, =S, =MR"), -CN, C3-12 carbocycle, and

3- to 12-membered heterocycle; and C3-12 carbocycle, and 3- to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in R4 is
optionally substituted with one or more substituents independently selected
from
halogen, -OR", -SR', -C(0)N(Rto)2,
-N(RICI)C (0 )R1-", -Ncle )C(0)N(R")2, -N(Rn2,
-C(D)RM, -C(C)ORKI, -0C(0)10 , -NO2, =0, =S, =N(R"), -CN, Ct-salkyl, C2-6
alkenyl,
and C2-6 alkynyl;
R5 is selected from unsaturated C4-8 carbocycle; bicyclic carbocycle; and
fused
5-5, fused 5-6, and fused 6-6 bicyclic heterocycle, wherein R5 is optionally
substituted
and wherein substituents are independently selected at each occurrence from:
halogen, -OR", -SR", -C(0)N(RI-0)2, _ N(10 )C(0)R",
-N(R")C(0)N(Rto)2, _N(t10)2, -C(0)R", -C(0)0R", -0C(0)R", -NO2, 0,=S,
=N(R"), and -CN;
CI-toalkyl, C2-10 alkenyl, C2-to alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-OR",
-SR", -C(0)N(R")2, -
m(Rto),c(o)Rto, --is(Rto),c(c)Nt-Rict,2, _
N(R")2, -C(0)R",
-C(0)0R", -0C(0)R", -NO2, =0, =S, =N(R"), -CN, C3-12 carbocycle, and 3- to 12-
membered heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, wherein each C3-12
carbocycle, and 3- to 12-membered heterocycle in R5 is optionally substituted
with one
or more substituents independently selected from halogen, -OR", -SR", -
C(0)N(R")2,
-N(IVIC(0)R", -
N(Rto)C(c)mR10)2, _mato)2, _Cor
K
- C(0)0R", -0C(0)Rth,
-NO2, =0, =S, =N(R"), -CN, C1.6alkyl, C2-6alkenyl, and C2-6alkynyl;
R" is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C61-15; and CI-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0CH2C6Els, -NHC(0)0CH2C6H5, Ci.to alkyl, -Ct-to haloalkyl, -0-C 1-to
alkyl, C2.10
alkenyl, C2-to alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle, and
habalkyl;
and
102 is independently selected at each occurrence from halogen, -OR",
299
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
_sRio, _N(Rio)2,
-C(0)R10, -C(0)N(R10)2, -N(R1 )C(Or - 10, K 20)0e), -0C(0)R1-0,
-SOW , -S(0)2Rio., -P(0)(011.10)2, -0P(0)(010)2, -NO2, =0, =S, =N(Rm), and -
CN;
Ct-to alkyl, C2-10 alkenyl, C2-11) alkynyl, each of which is optionally
substituted with one
or more substituents independently selected from halogen, -ORM, _SRI ,
_N(R10)2,
-C(0)Rm, -C(C)N(R1 lc
(0)
0)2, _mR10-)cor 10, _lc ORM, _ocoor _ 10, S(0)0-1), -S(0)2Ri ,
-P(0)(0R112, -0P(0)(ORIM)2, -NO2, =0, =S, =N(111 ), -CN, C3-10 carbocycle and
3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-to carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-OW ,
_sRio, _N(Rio)2,
-C(0)'K 10, - C(0)N(R10)2, _ot_l In Ngorto, _
ts. C(0)0R10,
-0C(0)R1 , -S(0)Rw, -S(0)2Rm, -P(0)(010 )2, -0P(0)(010 )2, -NO2, =0, =S,
=N(R1 ), -CN, C1-6 alkyl, C24 alkenyl, C2-6 alkynyl;
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from Ry or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle;
R20, R21, R22, and n23
n are independently selected from hydrogen, halogen,
-OW , -SR10, -N(Rm)2, -S(0)10, -S(0)21tm, -C(0)Rm, -C(0)011m, -0C(0)Rm, -NO2,
1:0, =S, =N(Rm), -CN, Ci-io alkyl, C2-10 alkenyl, and C2-10 alkynyl; and
R24 and Kn25
are independently selected from hydrogen, halogen, -ORm, -SRI',
-N(Rm)2, -S(0)11.1 , -S(0)2Rio, _icor , _ lO
it.
C(0)0111 , -0C(0)1110, -NO2, =0, =S,
=N(Rick _
), CN, Ci-io alkyl, C2-io alkenyl, and C2-io alkynyl; or R24 and R25 taken
together
form an optionally substituted saturated C3-7 carbocycle.
8. The method of any one of claims 1-7,
wherein the TLR8 agonist is a
compound of Category A, Formula IIC:
R1
N
co N -R2,
R5-- 0
..---
R4
L2-
(IIC)
or a pharmaceutically acceptable salt thereof, wherein:
300
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
RI- and R2 are hydrogen;
L2 is -C(0)-;
R4 is -N(R1 )2;
is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0C1tC6E15, -NHC(0)0CH2C6H5, Ci-to alkyl, -Ct-to haloalkyl, -0-C1-to
alkyl, C2-to
alkenyl, C2-10 alkynyl, C3-12 carbocycle, 3- to
12-membered heterocycle, and haloalkyl;
L10 is _C(c)mR10%_*,
wherein * represents where L11 is bound to R5; and
R5 is a fused 5-5, fused 5-6, or fused 6-6 bicyclic heterocycle, wherein R5 is

optionally substituted and wherein substituents are independently selected at
each
occurrence from:
halogen, -OR', -SR', -C(0)N(10 )2, -N(R' )C(0)R1- ,
-N(10 )C(0)N(Rw)2, -N(10 )2, -C(0)10 , -C(0)0Rw, -0C(0)Rw, -NO2, =0, =S,
=N(11.10), and -CN;
CI-toalkyl, C2-10 alkenyl, C2-10 alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-010 ,
-C(0)N(11.112, -N(R10)C(0)Rw, -
mR10)Coosi(Rto)2, 4,(R10)2, _Cow,
-C(0)0Rm, -0C(0)Rw, -NO2, =0, =S, =N(10 ), -CN, C3-12 carbocycle, and 3- to 12-

membered heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
halogen, -OW ,
-C(0)N(Rm)2, -N(R1 )C(0)R1 , -
N(R1 )C(0)N(Rw)2, -N(Rw)2,
-C(0)10 , -C(0)010 , -0C(0)10 , -NO2, =0, =S, =N(R1. ), -CN, 0.-6 alkyl, C2-6
alkenyl,
and C2-6 alkynyl.
9. The method of claim 8, wherein R4 is -
N(Ct4 alky1)2 and 12 is
-C(0)N(H)-*.
301
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
10. The method of claim 8 or 9, wherein:
CCH3
v.NI
R4 is CH3.
11. The method of any one of claims 1 to 10, wherein the TLR8 agonist is
selected from:
oo os 0
0 SID o ..... NH2
NH2
N.....
M N
* N
H
0 -- T----\
N
N
0
OH
0011 N N..... NH2 0 4
0 NI, NH2
H IP N
H
OH -- /----\
N
N
0 \--\ ,
0 \--\ ,
H
N
0 0 NH2 HN 110 0
NH2
N,
N
II
so -- r-N
11 *-- r-=
N
N
0 \¨\
0 \--\
SI 0 0
010 N 0
NH2
NH2
N
N
HN
,
N N
H
1101
- r"-\
- r"-\
N
N
0 \---\
0 µ-----\
,
,
As 0 N, NH2
0
A.
ift- EiN toN*.
NH2
N,
-- r--\.
11 10 , ,
N
0 N
0 \--\
,
,
4 0
cci N 0
NH2
NH2
_...N N,
N._
Cbz"
N *
H
rm.\ NH2
--- r¨\sõ
N
N
0 \---\
0 \----\
302
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
ci.......,:_cx 0 ....N
ciIL 0
N
NH2 NH2
N..,
õ
N---
N
H H2N
H
NHCbz --- r---
\ --- r---\
N
N
,
,
caN 0
N H2 0 NH2
N.õ
0
N N
CbzHN
HILIH
re \\ H2N
---
h(----\
N
,
N
O. 0
NH2
= Nai, N N NH2
N
HN,. H
H
az ---- f---\
--- NrTh
N
,
,
H
ouN 0
CbleNaiN
..-= , 0
NH2
I NH2
HN -====._ N__
N,
N
N
H
H
.--- /---\
---- NT- \
N
c .....ra 0
HN
NH2
,
az N
N
H
--- Nr--\
,
H
N
ilk 0--(
NH2
N
4N (3
t
HO H
--- /Th
N
0 \----\
,
NH2
Si
o Nõ
aNteCILIXN N
.
H H *0
.----
Nr- \
,
CbzHN
\ 4coLN 0
NH2
HN *
' Nr-\
0 \----\
,
303
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N,..1
0
LN * CCX N NH2
N ...e ,
N
H
..---
rs,
N
1
1 rt.. 0
NH2 NH:
Mail N, HiraiN N
11 ill
H Sil
H3C0 a..... r--\
F --- re-\
N N
0 \Th
0 \Th
0
H2N------CN so ,a5s, 0
NH2
F F " N ' ----
m N,
ii 0---- 1---\
N
and
0 \--\\ ,and
pharmaceutically acceptable salts thereof.
12. The method of any one of claims 1-11,
wherein Dx is a compound of
Formula (IVB):
R1
R20
1
N--R2
1 12
N.õ
R8--I- R25
,)LçfR24
R21 1 22 _R4
R22 R23 1-
(IVB)
or a pharmaceutically acceptable salt thereof, wherein:
I22 is selected from -V-, -V-C1-6alkylene-r-, -V-C2-salkenylene-X3-, and
-V-C2-6alkynylene-X3-, each of which is optionally substituted on alkylene,
alkenylene, or alkynylene with one or more substituents independently selected
from
R'2,
L22 is independently selected from -X4-, -X4-Ci-6alkylene-r-, -X4-C2-6
alkenylene-X4-, and -r-C2-6alkynylene-r-, each of which is optionally
substituted on
304
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
alkylene, alkenylene, or alkynylene with one or more substituents
independently
selected from R";
X3 and X4 are independently selected at each occurrence from a bond, -0-, -S-,
-N(R")-, -C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(RION_
),
C(0)N(R")C(0)-,
-C(0)N(Rm)C(0)N(R' )-, -N(R")C(0)-, -N(Rt )C(0)N(RtG)-, -N(R")C(0)0-,
-0C(0)N(R")-, -g-NRtoy, 41/44(t10),G(NR10)_, -,c(NR Joy,4(Rto)-,
-N(R")C(NRIIN(R")-, -S(0)2-, -OS(0)-, -S(0)0-, -S(0)-, -OS(0)2-, -S(0)20-,
-N(RnS(0)2-, -S(0)2N(R10)_,
NatnS(C)-, -S(0)N(Rm)-, -N(RnS(0)2N(Rm)-, and
-N(RnS(0)N(R")-;
RI- and R2 are independently selected from 1,3, and hydrogen; and Ci-to alkyl,

C2-10 alkenyl, and C2-t0 alkynyl, each of which is optionally bound to 1-3 and
each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -ORD, -SR", -C(0)N(R")2, -N(Rw)2, -S(C)R", -S(0)2R", -C(0)R",
-C(0)0R"), -0C(0)Rm, -NO2, =0, =S, =N(R"), and -CN;
R.4 and le are independently selected from: -OR", -N(141 )2, -C(C)N(Rm)2,
-C(0)R", -C(0)0R", -S(0)R", and -S(0)2R"; Ci-to alkyl, C2-10 alkenyl, C2-to
alkynyl,
each of which is optionally bound to I-3 and each of which is optionally
substituted with
one or more substituents independently selected from halogen, -OR", -SR",
-C(0)N(R1 )2, -N(R1 )C(0)Rt , -N(10 )C(C)N(Rm)2, -N(R")2, -C(0)Rm, -C(0)0R",
-0C(0)Rm, -NO2, =0, =S, =N(R")), -CN, C3-12 carbocycle, and 3- to 12-membered
heterocycle; and C3-12 carbocycle, and 3- to 12-membered heterocycle, wherein
each
C3-12 carbocycle, and 3- to 12-membered heterocycle in le and le is optionally
bound
to L3 and each C3-12 carbocycle, and 3- to 12-membered heterocycle in R4 and
le is
optionally substituted with one or more substituents independently selected
from
halogen, -OR', -SR", -C(0)N(Rm)2, -N(RnC(0)R", -N(Rm)C(0)N(R")2,
-N(R")2, -C(Otto,
) -C(0)010 , -0C(0)10 , -NO2, =0,
=S, =N(R"), -CN, CI-6alkyl,
C2-6alkenyl, and C2-6alkynyl;
Rm is independently selected at each occurrence from L3, hydrogen, -N112,
-C(0)0CH2C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NH2, =0, =S,
305
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, C1-10 alkyl, C2-143 alkenyl, C2-to alkynyl, C3-
12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
L3 iS a linker moiety, wherein there is at least one occurrence of L3; and
R12 is independently selected at each occurrence from halogen, -0R1 , -SW ,
-N(Rm)2, -C(0)Rto,
-C(0)N(Rm) - 2 , N(Rm)C(0)R1 1-C(0)0Rto, -0,c(c)Rtio, _so:ow ,
2n 10, -
-S(0) K P(0)(010 )2, -0P(0)(0R112, -NO2, =0, =S, =N(Rm), and -CN; C1-10 alkyl,

C2-10 alkenyl, C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -ORD),

)
_ C(0)R1 ,
-C(0)N(R1 13)2, -N(R1G)C(0)tc.n 1 , -C(0)0R1 , -0C(0)141 , -S(0)11.1 , -
S(0)211.10,
_powaRto)2,
-0P(0)(010 )2, -NO2, =0, =S, -N(Rm), -CN, C3-10 carbocycle and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-ORR',
mwo)2, to, _ -C(0)Rm, -C(0)N(Rto)2, -N(R1 )C(0)R1
, -C(0)0R10, -0C(0)Rm,
-S(0)Rm, -S(0)2R1o, _p(:woRto)2, -0P(OXORm)2, -NO2, =0, =S, =N(Rm), -CN, C1-6
alkyl, C2-6 alkenyl, C2-6 alkynyl;
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from R12 or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle;
R20, R21, K n22,
and R23 are independently selected from hydrogen, halogen,
-OW , - N(Rm)2, -S(0)1110, -S(0)2R1 , -C(0)Rm, -C(0)0Rm, -0C(0)Rm, -NO2,
=0, =S, =N(Rm), -CN, Ct-to alkyl, C2-10 alkenyl, and C2-10 alkynyl; and
R24, and R25 are independently selected from hydrogen, halogen, _Ow , am,
-N(Rm)2, -S(0)Rm, -S(0)2Rm, -C(0)Rm, -C(0)0Rm, -0C(0)Rm, -NO2, =0, =S,
=Novo% _
CN, Ci-to alkyl, C2-to alkenyl, and C2-to alkynyl; or R24 and R25 taken
together
form an optionally substituted saturated C3-7 carbocycle.
306
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
13. The method of any one of claims 1-12,
wherein Dx is a compound of
Formula (1VC):
N¨R2
RBL12
L22-R4
(IVC)
or a pharmaceutically acceptable salt thereof, wherein:
RI and R2 are hydrogen;
L22 is _C(0)-
R4 is -N(Rm)2;
RR' is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C6H5; and Ci-loalkyl, C2-10 alkenyl, C2-to alkynyl, C3-12 carbocycle,
and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NH2, =0, =S,
-C(0)0CH2C6H5, -NHC(0)0CH2C61-15, Ci-to alkyl, C2-143alkenyl, C2-113alkynyl,
C3-12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
L1-2 is -C(0)N(Rto)*_,
wherein * represents where L'2 is bound to le;
R8 is an optionally substituted fused 5-5, fused 5-6, or fused 6-6 bicyclic
heterocycle bound to linker moiety L3,
and wherein optional substituents are independently selected at each
occurrence
from:
halogen, -ORto, _situ% -,c(c)mRto)2, 4.4(Rto),c(c)Rto,
-N(RnC(0)N(Rm)2, _mato)2, -C(0)Rm, -C(0)ORt , -0C(0)Rt0, -NO2, 0,=S,
=N(R1 ), and -CN;
CI-toalkyl, C2-10 alkenyl, C2-to alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-OR',
-C(0)N(Rm)2, -N(Rm)C(0)Rm,
2
- mRto)Con-Rto,), - MR112, -C(0)R1171,
-C(0)0Rt0, -0C(0)R1 , -NO2, =0, =S, =N(R11"), -CN, C3-12 carbocycle, and 3- to
12-
membered heterocycle; and
307
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
C3-i2 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
halogen, -Ole , -SRI , -C(0)N(11.1 )2, -N(11.1 )C(0)Ri , -N(R1 )C(0)N(RIO)2, -
N(R10)2,
_Cow , _C(0)0Rw, -0C(0)R1 , -NO2, =0, =S, =N(R11, -CN, C 1-6alkyl, C2-6
alkenyl,
and C2-6 alkynyl.
14. The method of claim 13, wherein R4 is -N(Ci4 alky1)2 and L12 is
-C(0)N(1-1)-*.
15. The method of claim 13 or 14, wherein:
CCH3
v.NI
R4 is CH3.
16. The method of any one of claims 1-15, wherein L and Dx together have
a structure selected from:
H2N y0
HN
H H
IsiCIRri N N N
H NH2
0 0 1. 0..õtr
ill
H
N
H
0
---- r---=
N
H2N y0
HN
0
issCFOCIFNIIAN Fri
: H NH2
00 0
...v., Ell jiji..; M,
II
N
H
0
N
308
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
N
0
1 0
H2
0
AN
licilõ.......1j
N, N
0 41
Thle -. N
N NN H 0 H
=---." r\
H H
N
0
0 NI-a\
Hi y
H2N0
,
N
0
NH2
H
N;:(J 0 N,
0 Xllii 0 0 0A NThrN
N
.v.RX:õ........,,,,,...,,,..A
N N-yeit-N H 0 H
H H
N
Of
0 \-"---\
HN
H2NA0
i
* N
0
0 Nii2
A
kilm.jc...11
N,
0 4"%riiii 0 1. 0 N
N
0 H
N
--"--. r---\
H H
N
0
0 \Th
NH
OgH2
,
. N
0
A rill.,..../U1
0 Y H 0 00 0 ri
N
H
VRX:õ......................",.....A
NThrINK:AN
H i H
N
0 .=,..1...
0 \.-*\
NH
CAN H2
,
1-12Ny0
HN
H H
FRX-1111;1a14;-N
0 40 0
H
0
1 yl'il -%--e---Iil
* re.N*1:-11 0 NH2
0
W,N N.
H
,
309
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
02Nyo
liN
0
- N
0 IS

N
= 0
H
Oa 0 0
O N
y
NH2
0
,
HLJNr-Ths.
0
H2Ny0
FIN
0
Nt\
11-Rn
0
0 0 OS
N
0
Hit n 0
NH2
--- re\
0 5
H2N Nei
HN
0
jk\ 111
N
0
H
0 0 140
0
H 0
LLNN, NH2
0 \ 5
H2N tO
HN
H
RTN
0
0 111
0
N
C1NU o
NH2
\--Th
310
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N
tO
HN
1 0
1 )
11(
tsittcr N
= H
0 411 0
0
N
CINU
NH2
N,
0 \---\
H2Ny.0
HN
0
'R A
0 H 0 N 40 0.....e,NOLIN
NH2
0
reN,
0 NI
H2N tO
HN
0
caN 0
NH2
- H 0 Nõ
0
0
\---rN
o 0
0 0
* re o NH2
141i HFFH
H H
HIjfJ0 rsk
HN
0 \---\
H2N--%0
and
o o
oIN^-2CN * o NH,
F F
N
H - H
re \
HN
H2VLO
and salts thereof,
311
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
wherein the RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide
moiety bound to a residue of an antibody,
wherein on RX* represents the point of
attachment to the residue of the
antibody.
17. The method of claim 16, wherein L and
Dx together have a structure
selected from:
H2Ny0
HN
H 0
Oy N * 0
NH2
0
N__
- rm.
0 \
H2Ny0
HN
1 ,1Aki
_ N
0 141)
0
o Jr\
NH2
0
Nt,
-
LJÇ
0
\Th r
H2N y0
HN
NH2
0 * 0
0
NC\
0
312
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N y0
HN
0
Rnj13y
b- N
0 NH2
_ H 0 1010
0
0 \¨\
o
* 0
= NH2
VRXIC.---A-X-ry-N HFF H
H H
HN
0
H2N
0
0
=
0 0 Ns.--)C-N
NH2
F F
H E H
Ill 0 al
111111' fTh
HN
0 \Th
and H2W--LO
7
and salts thereof,
wherein the RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide
moiety bound to a residue of an antibody,
wherein on RX* represents the point of
attachment to the residue of the
antibody.
18. The method of claim 16 or 17, wherein RX* comprises a succinamide
moiety and is bound to a cysteine residue of the antibody.
19. The method of claim 16 or 17, wherein RX* comprises a hydrolyzed
succinamide moiety and is bound to a cysteine residue of the antibody.
20. The method of any one of claims 1-19, wherein the tumor antigen is
selected from YIER2, Nectin4, MSLN, LIV-1, MUC16, CEACAM1, CEACAM3,
CEACAM4, CEACAM5, CEACAM6, CEACAM7, CEACAM8, CEACAM16,
CEACAM18, CEACAMI9, CEACAM20, CEACAM2I, VEGFRI, VEGFR2, MUCI,
PSMA, PSA, MUC-2, and LRRC15
313
CA 03151322 2022- 3- 15

WO 2021(067644
PCT/1JS2020/053870
21. The method of any one of claims 1-20,
wherein the tumor associated
antigen is selected from:
a) an antigen present on breast cancer, wherein the antigen is optionally
selected from ITER2, LIV-I, CDH3 (p-cadherin), MUC I, Sialo-epitope CA6, PTK7,

GPNMB, LAMP-1, LRRC15, ADAM12, EPHA2, TNC, LYPD3, EFNA4, Nectin-4,
and CLDN6;
b) an antigen present on lung cancer, wherein the antigen is optionally
selected from mesothelin, HER2, EGFR, PD-L1, MSLN, LY6K, CD56, PTK7, FOLR1,
DLL3, SLC34A2, CECAM5, MUC16, LRRCI5, ADAM12, EGFRvIII, LYPD3,
EFNA4, Nectin-4, and MUCl;
c) an antigen present on stomach/gastric cancer, wherein the antigen is
optionally selected from HER2, EPHB2, TMEM238, CECAM5, and EFNA4;
d) an antigen present on colorectal cancer, wherein the antigen is
optionally
selected from EPHB2, TMEM238, CECAM5, LRRC15, ADAM12, EFNA4, Nectin-4,
and GPA33;
e) an antigen present on liver cancer, wherein the antigen is optionally
selected from GPC3, EPCAM, and CECAM5;
0 an antigen present on lcidney cancer,
wherein the antigen is optionally
selected from HAVCR1, ENPP3, CD116, CD70, and cMET;
g) an antigen present on ovarian cancer, wherein the antigen is optionally
selected from MUC16, MUC I, MSLN, FOLRI, sTN, VTCN1, HER2, PTK7, FAP,
TMEM238, LRRC15, CLDN6, SLC34A2, and EFNA4;
h) an antigen present on head and neck cancer, wherein the antigen is
optionally selected from LY6K, PTK7, LRRC15, ADAM12, LYPD3, EFNA4, Nectin-
4, and TNC;
i) an antigen present on bone cancer, wherein the antigen is optionally
selected from EPHA2, LRRCI5, ADAM12, GPNMB, TP-3, and CD248;
I) an antigen present on mesothelioma,
wherein the antigen is optionally
MSLN;
314
CA 03151322 2022-3-15

WO 2021(067644
PCT/1JS2020/053870
k) an antigen present on bladder cancer, wherein the antigen is optionally
selected from LY6K, PTK7, UPK1B, UPK2, TNC, Nectin4, SLITRK6, LYPD3,
EFNA4, and 1-IER2;
l) an antigen present on prostate cancer, wherein the antigen is optionally

selected from PSMA, FOLH1, PTK7, STEAP, TMEFF2 (TENB2), OR51E2,
SLC30A4, and EFNA4;
m) an antigen present on thyroid cancer, wherein the antigen is optionally
PTK7;
n) an antigen present on uterine cancer, wherein the antigen is optionally
selected from LY6K, PTK7, EPFIB2, FOLR1, ALPPL2, MUC16, Nectin-4, and
EFNA4;
o) an antigen present on cervical/endometrial cancer, wherein the antigen
is
optionally selected from LY6K, PTK7, MUC16, LYPD3, EFNA4, Nectin4, and muc 1;
and
13) an antigen present on pancreatic
cancer, wherein the antigen is
optionally selected from PTK7, MUC16, MSLN, LRRC15, ADAM12, EFNA4,
MUC5A, Nectin-4, and MUC1.
22. The method of any one of claims 1-21, wherein the antibody is an anti-
HER2 antibody.
23. The method of any one of claims 1-22, wherein the antibody is
pertuzumab, trastuzumab, or ladiratuzumab, or a biosimilar thereof; or
comprises the
CDRs of pertuzumab, trastuzumab, or ladiratuzumab; or comprises an antigen
binding
fragment of pertuzumab, trastuzumab, or ladiratuzumab.
24. The method of any one of claims 1-23, wherein the antibody comprises
HC-CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ 1:13
NOs: 1-6, respectively; or the antibody comprises a heavy chain variable
region
comprising the amino acid sequence of SEQ ID NO: 7 and a light chain variable
region
comprising the amino acid sequence of SEQ ID NO: 8; or the antibody comprises
a
315
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
heavy chain comptising the amino acid sequence of SEQ ID NO: 9 and a light
chain
comprising the amino acid sequence of SEQ ID NO: 10.
25. The method of any one of claims 1-24, wherein the method comprises
administering an additional therapeutic agent.
26. The method of claim 25, wherein the additional therapeutic agent
comprises a chemotherapy agent, a kinase inhibitor, a therapeutic antibody, or
any
combination thereof.
27. The method of claim 25 or 26, wherein the additional therapeutic agent
comprises a therapeutic agent targeting HER2.
28. The method of any one of claims 25-27, wherein the additional
therapeutic agent is selected from trastuzumab, trastuzumab-qyyp, trastuzumab-
pkrb,
trastuzumab-dttb, trastuzumab-anns, trastuzumab-dkst, trastuzumab, deruxtecan-
nxki,
ado-trastuzumab emtansine, trastuzumab duocarmazine, margetuximab,
zenocutuzumab
, tucatinib, cabozantinib, pyrotinib, and poziotinib.
29. The method of any one of claims 25-28, wherein the additional
therapeutic agent comprises an anti-IIER2 antibody, optionally wherein the
therapeutic
agent comprising the anti-HER2 antibody is selected from trastuzumab,
trastuzumab-
qyyp, trastuzumab-pkrb, trastuzumab-dtth, trastuzumab-anns, trastuzumab-dkst,
trastuzumab deruxtecan-nxki, ado-trastuzumab emtansine, trastuzumab
duocarmazine,
margetuximab, and zenocutuzumab
30. The method of claim 26, wherein the kinase inhibitor is a tyrosine
kinase
inhibitor or a serine/threonine kinase inhibitor.
31. The method of claim 30, wherein the kinase inhibitor is a tyrosine
kinase
inhibitor selected from tucatinib, cabozantinib, afatinib, erlotinib,
pyrotinib, poziotinib,
316
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
dacomitinib, gefitinib, lapatinib, osimertinib, larotrectinib, axitinib,
lenvatinib,
pazopanib, regorafenib, and sunitinib.
32. The method of claim 30, wherein the kinase inhibitor is a
serine/threonine kinase inhibitor that is a CDK4/6 inhibitor selected from
abemaciclib,
palbociclib, and ribociclib.
33. The method of claim 26, wherein the chemotherapy agent comprises:
(a) a hormone-based compound, optionally wherein the hormone
based compound comprises anastrozole, exemestane, letrozole, fulvestrant,
bicalutamide, flutamide, nilutamide, enzalutamide, apalutamide, darolutamide,
degarelix, toremifene, goserelin, triptorelin, histrelin, leuprolide, or
tamoxifen citrate;
(b) a platinum coordination complex, optionally wherein the
platinum coordination complex comprises cisplatin, oxaliplatin, or
carboplatin;
(c) a folic acid analog, optionally wherein the folic acid analog
comprises pemetrexed, methotrexate, or trimetrexate;
(d) a primidine analog, optionally wherein the pyrimidine analog
comprises fluorouracil, capecitabine, fluorodeoxyuridine, tezacitabine,
gemcitabine,
cytosine arabinoside, cytarabine, 5-azacytidine, or 2,2'-
difluorodeoxycytidine;
(e) a microtubule inhibitor, optionally wherein the microtubule
inhibitor comprises paclitaxel, docetaxel, and eribulin;
or any combination thereof.
34. The method of claim 25, wherein the additional therapeutic agent
comprises a chemotherapy protective agent, optionally wherein the chemotherapy

protective agent comprises leucovorin.
35. The method of any one of claims 1-4 and 6-34, wherein:
317
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(a) L and Dx together have a stnrcture selected from:
FI2N yO
HN
H
x11-11\-14
0
H"
0 O 0,1,4
õLa....N.-p 0
i
8
NH2
N__
0
and
H2Ny0
HN
0
1/1A1:11
0
0 H 0 1. ONCLJLN NH2,
11
and salts thereof;
(b) the immune checkpoint inhibitor is pembrolizumab;
(c) optionally comprising an additional therapeutic agent, wherein
the additional therapeutic agent comprises fam-trastuzumab deruxtecan-nxki or
ado-
trastuzumab emtansine; and
(d) optionally, wherein the antibody comprises HC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
36, The method of claim 1-4 and 6-34,
wherein:
318
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(a) L and Dx together have a structure selected from:
H2Ny0
HN
H 0\ 0
hRXr H H
0
0 NYLN 0 lkit OyN..,N *
N
H
--.
0
NH2
0
MN N,
H
¨ it----\
hi
and
H2N tO
HN
0
kli)yl 411) ill
N
1---roc<s------------Thr N
1 0
H
0 0
0.õ..NOLIN NNH2,
11
H
N
and salts thereof
(b) the immune checkpoint inhibitor is nivolumab;
(c) optionally, wherein the antibody comprises HC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ [13 NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
37. The method of any one of claims 25-29,
and 33, wherein:
319
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(a) L and Dx together have a structure selected from:
H2Ny0
HN
H
1--RXINX1111%N
*

0 O OyN.N

0
....1µ1õzsi
NH2
LJÇ
0
r¨ss,
0
and
H2N yO
HN
ARINXTL)INH
0 40/
NH2
0
14,
11
0
0 \---\
and salts thereof
(b) optionally wherein the immune checkpoint inhibitor that inhibits
PD-1 and/or PD-L1 is pembrolizumab, nivolumab, or atezolizumab;
(c) the additional therapeutic agent comprises:
trastuzumab,
a pyrimidine analog, optionally wherein the pyrmidine analog is
fluorouracil or capecitabine, and
a platinum coordination complex, optionally wherein the
platinum coordination complex is oxaliplatin, or cisplatin; and
(d) optionally wherein the cancer is gastric cancer;
(e) optionally, wherein the antibody comprises HC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
320
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
38. The method of any one of claims 25-29,
and 33, wherein:
(a) L and Dx together have a
structure selected from:
H2N,f0
HN
H 0
0
0 H 0 OyNN *
0
NH2
0
N
0 \
and
H2Ny0
HN
NH2
0 11;11?\11 0 PI 40

0
0
;
and salts thereof;
(b) the immune checkpoint
inhibitor that inhibits PD-1 and/or PD-Ll
is pembrolizumab, nivolumab, or atezolizumab;
(c) the additional therapeutic
agent comprises:
(i) trastuzumab; and
(ii) a chemotherapy regimen comprising lencovorin,
fluorouracil, and oxaliplatin (FOLFOX);
(c) optionally, wherein the cancer
is gastric cancer; and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
321
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
39. The method of any one of claims 25-29,
and 33, wherein:
(a) L and Dx together have a
structure selected from:
HaN y.0
HN
0
1.-1RX1141;CS1(11:11
0
HN 411
0 O
8
N
roC1
I-12 N
N,
/Th
0 \----\\
and
H2Nto
HN
0
Alt\ 111
N,
NH2
0
0 41 o
0 r-\
0 ;
and salts thereof;
(b) the immune checkpoint
inhibitor that inhibits PD-1 and/or PD-L1
is pembrolizumab, nivolumab, or atezolizumab;
(c) the additional therapeutic
agent comprises:
trastuzumab;
(ii) leucovorin,
(iii) fluorouracil; and
(iv) cisplatin;
(d) optionally, wherein the cancer
is gastric cancer; and
322
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(e) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
40. The method of any one of claims 25-29,
and 33, wherein:
(a) L and Dx together have a structure selected from:
H2Ny0
HN
0
AyL\irril
FRXtr11
0
HN
0
CLIIN-*%="----"N
rall 0 NH2
0
N I N,
ra\
and
FI2N tO
HN
ÚRÇLÇH
N
0 H 0 10/
it_ NH2
0
rC
0 ;
and salts thereof;
(b) the immune checkpoint inhibitor that inhibits PD-1 and/or PD-L1
is pembrolizumab, nivolumab, or atezolizumab;
(c) the additional therapeutic agent comprises:
trastuzumab;
(ii) capecitabine;
323
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(iii) oxaliplatin; and
(c) optionally, wherein the cancer is gastric cancer; and
(d) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
41. The method of any one of claims 25-29
and 33, wherein:
(a) L and Dx together have a structure selected from:
H2Nr
HN
H
1"-RX
0 0 le
0
0
NH2
jt
0
N,
N
and
H2N tO
HN
H
A in N
0
0 0 SIN
NH2
0
r-Thõ
0 \
and salts thereof;
(b) the immune checkpoint inhibitor that inhibits PD-1 and/or PD-Ll
is pembrolizumab, nivolumab, or atezolizumab;
(c) the additional therapeutic agent comprises:
324
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
trastuzumab; and
(ii) optionally capecitabine;
(d) optionally, wherein the cancer is gastric cancer; and
(e) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
42. The method of any one of claims 25-29
and 33, wherein:
(a) L and Dx together have a stmcture selected from:
H2N y0
HN
H
hR>6.1-N N N sah
"pi 0
0 O OyNNio
NH2
HJ
r--
0 N\-Th r
and
H2N tO
HN
rj?\ri
0 1411 0 M..N
N
NH2
0
,
and salts thereof;
(b) the immune checkpoint inhibitor that inhibits PD-1 and/or PD-Ll
comprises pembrolizumab, and optionally atezolizumab;
325
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(c) the additional therapeutic
agent comprises:
(i) trastuzumab; and
(ii) docetaxel or paclitaxel;
(d) optionally, wherein the cancer
is breast cancer; and
(e) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
43. The method of any one of claims 25-29
and 33, wherein:
(a) L and Dx together have a
structure selected from:
H2NtO
HN
H 0
ERXrdsadlir4,)Ci::1)rN sit
mpi
0
0
o
NH2
0
NI&NATINK,
o
and
H2Nto
HN
0
NH2
0 10
1.N 0 N,
0
0 N\Th;
and salts thereof;
326
CA 03151322 2022-3-15

WO2021/067644
PCT/1JS2020/053870
(b) the immune checkpoint inhibitor that inhibits PD-1 and/or PD-L1
is atezolizumab or durvalumab;
(c) the additional therapeutic agent comprises fam-trastuzumab
deruxtecan-nxki or ado-trastuzumab emtansine;
(d) optionally wherein the cancer is breast cancer; and
(e) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
44. The method of any one of claims 25, 26,
and 33, wherein:
(a) L and Dx together have a
structure selected from:
H2Ny.0
HN
0
11
0
" 140
0 -õle o 0
so Ca 0 N NH2
8
- r-
and
327
CA 03151322 2022-3-15

WO2021/067644
PCMJS2020/053870
H2NyO
HN
HN:11LaH
NH2
0 X Hlie 0 110N
0
0 \-- rTh.
-\
and salts thereof;
(b) the immune checkpoint inhibitor is pembrolizumab;
(c) the additional therapeutic agent comprises:
pemetrexed; and
(ii) carboplatin or
cisplatin;
(c) optionally, wherein the cancer
is non-small cell lung cancer;
and/or
(d) optionally, wherein the antibody comprises HC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
45. The method of any one of claims 25, 26,
and 33, wherein:
(a) L and Dx together have a
structure selected from:
H2N
tO
HN
0
14:2111111
cp
0
0
r..,N,2:71
NH2
111N)L,40A.-N
N,
0
and
328
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
H2Ny.0
HN
H
H 140 ONCLIì
NH2
N.,
0
0
=
and salts thereof;
(b) the immune checkpoint
inhibitor is atezolizumab;
(c) the additional therapeutic
agent comprises:
paclitaxel;
(ii) carboplatin; and
(iii) bevacizumab;
(c) optionally, wherein the cancer
is non-small cell lung cancer;
and/or
(d) optionally, wherein the
antibody comprises FIC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
46. The method of claim 25-29, wherein the conjugate comprises
pertuzumab or a biosimilar thereof, or comprises the CDRs of pertuzumab, or
comprises an antigen binding fragment of pertuzumab, and the additional
therapeutic
agent comprises trastuzumab or comprises the CDRs of trastuzumab, or comprises
an
antigen binding fragment of trastuzumab.
47. The method of any one of claims 1-46, wherein the cancer expresses
HER2 at a level of 2+ or 3+ as determined by immunohistochemistry.
329
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
48. The method of claim 47, wherein the cancer expresses HER2 at a level
of 3+ as determined by immunohistochemistry.
49. The method of any one of claims 1-48, wherein the antibody of the
conjugate comprises an antigen binding domain and an Fc domain.
50. The method of claim 49, wherein the Fc domain exhibits the same or
substantially similar binding affinity to at least one Fcgamma receptor and/or
an FcRn
receptor as compared to a wild-type IgG1 Fc domain.
51. The method of claim 49, wherein the Fc domain has increased binding
affinity to at least one Fcgamma receptor as compared to a wild-type IgG Fc
domain.
52. The method of any one of claims 49-51, wherein the Fc domain is an
IgG1 Fc domain.
53. The method of any one of claims 1-52, wherein the method comprises
administering an effective regimen of the conjugate, wherein the effective
regimen
results in a Tmax of the conjugate in the subject of greater than 4 hours
following each
administration of the conjugate.
54. The method of any one of claims 1-53, wherein the method comprises
administering an effective regimen of the conjugate, wherein the effective
regimen
comprises at least two cycles of administration of the conjugate to the
subject and a
total dose of greater than 0.4 mg/kg of the conjugate per cycle.
55. The method of any one of claims 1-54, wherein the conjugate is
administered subcutaneously.
56. The method of any one of claims 1-54, wherein the conjugate is
administered intravenously.
330
CA 03151322 2022-3-15

WO 2021(067644
PCT/1JS2020/053870
57. A method of treating a HER2-expressing cancer, comprising
administering to a subject with a HER2-expressing cancer a conjugate and an
additional
therapeutic agent comprising a kinase inhibitor, a therapeutic agent
comprising an anti-
HER2 antibody, or both; wherein the conjugate is represented by Formula (I):
4) 4>
11:0õ )n
(I);
wherein:
A is an antibody that binds HER2,
L is a linker;
Dx is a TLR8 agonist, wherein the TLR8 agonist is a benzazepine compound;
n is selected from 1 to 20; and
z is selected from 1 to 20.
58. The method of claim 57, wherein n is 1 and z is from 1 to 8.
59. The method of claim 57 or claim 58, wherein the antibody of the
conjugate is pertuzumab or trastuzumab, or a biosimilar thereof; or comprises
the CDRs
of pertuzumab or trastuzumab; or comprises an antigen binding fragment of
pertuzumab
or trastuzumab.
60. The method of any one of claims 57-59, wherein the antibody of the
conjugate comprises HC-CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and
LC-CDR3 of SEQ ID NOs: 1-6, respectively; or the antibody comprises a heavy
chain
variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light
chain
variable region comprising the amino acid sequence of SEQ ID NO: 8; or the
antibody
comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and
a
light chain comprising the amino acid sequence of SEQ lD NO: 10.
331
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
61. The method of any one of claims 57-60, wherein the additional
therapeutic agent comprises an anti-HER2 antibody that binds to a different
epitope as
compared to the conjugate antibody that binds HER2.
62. The method of any one of claims 57-61, wherein the additional
therapeutic agent is selected from trastuzumab, trastuzumab-qyyp, trastuzumab-
pkrb,
trastuzurnab-dttb, trastuzumab-anns, trastuzumab-dkst, trastuzumab deruxtecan-
nxki,
ado-trastuzumab emtansine, trastuzumab duocarmazine, margetuximab,
zenocutuzumab
, tucatinib, cabozantinib, pyrotinib, and ponotinib.
63. The method of any one of claims 57-60, wherein the kinase inhibitor is
a
tyrosine kinase inhibitor, optionally wherein the tyrosine kinase inhibitor is
tucatinib,
cabozantinib, afatinib, erlotinib, pyrotinib, poziotinib, dacomitinib,
gefitinib, lapatinib,
osimertinib, larotrectinib, axitinib, lenvatinib, pazopanib, regorafenib, or
sunitinib.
64 The method of any one of claims 57-60,
wherein the kinase inhibitor is a
CDK4/6 kinase inhibitor, optionally wherein the CDK4/6 kinase inhibitor is
palbociclib, abemaciclib, or ribociclib.
65. The method of any one of claims 57-64, the additional therapeutic agent

further comprises a chemotherapy agent, an immune checkpoint inhibitor,
chemotherapy protective agent, or any combination thereof
66. The method of any one of claims 57-65, wherein the conjugate
comprises pertuzumab or comprises the CDRs of pertuzumab, or comprises an
antigen
binding fragment of pertuzumab; and the additional therapeutic agent comprises

trastuzumab, or a biosimilar thereof, or comprises the CDRs or trastuzumab; or

comprises an antigen binding fragment of trastuzumab.
67. The method of any one of claims 57-66, wherein the cancer expresses
HER2 at a level of 2+ or 3+ as determined by immunohistochemistry.
332
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
68. The method of claim 67, wherein the cancer expresses HER2 at a level
of 3+ as determined by immunohistochemistry.
69. The method of any one of claims 57-68, wherein the TLR8 agonist is a
compound of Category A, Formula (IlB):
R1
R2
%
N-R2
R5--1-1 N
-- R25
R24
---
R21
in L2-R4
R22 R-
(116)
or a pharmaceutically acceptable salt thereof, wherein:
Li() is _xto_;
L2 is selected from -X2-, -X2-C1-6 alkylene-X2-, -X2-C2-6 alkenylene-X2-, and
-X2-C2-6 alkynylene-X2-, each of which is optionally substituted on alkylene,
alkenylene
or alkynylene with one or more 102;
XI is selected from -C(0)-, and -C(C)N(Rm)-*, wherein * represents where X1
is bound to R5;
X2 at each occurrence is independently selected from a bond, -0-, -S-, -N(R10)-
,
-C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(R10)-, -C(0)N(Rw)C(0)-,
-C(0)N(Rm)C(0)N(It'o), _N(tto)ccv.)-,_, _
N(R1 )C(0)N(Ri )-, -N(R')C(0)0-,
-0C(0)N(IV,:)_, _(Nitio)_, _N(Rio),G(NRKI,_
),
-C(NR"))N(Rm)-,
-N(Rw)C(NR"))N(Rm)-, -S(0)2-, -0S(0)-, -S(0)0-, -S(0), -OS(0)2-, -S(0)20,
-N(Rt5S(0)2-, -S(0)2N(Rl0)-, _Nattowc.)_, -S(0)N(10- )-, -N(R10)S(0)2N(R10)-,
and
-N(RnS(0)N(R1 )-;
RI and R2 are independently selected from hydrogen; and CI-toalkyl, C2-to
alkenyl, and C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -0100, -SR', -C(0)N(R10)2,
_N(tt0)2, -S(0)Rm, -S(0)2Itt , -C(0)Rw, -C(0)0Rm, -0C(0)10- , -NO2, =0, =S,
=N(11.1 ), and -CN;
It4 is selected from: -OW , -N(11."))2, -C(0)N(R10)2, -C(0)R1 , -C(0)0Rw,
333
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
-S(0)Rm, and -S(0)2R10; Ci-to alkyl, C2-lo alkenyl, C2-10a1kyny1, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, 10
-OR, -SR", -C(0)N(R")2, -N(R")C(0)R", -N(R1 )C(0)N(R10)2, -N(R10)2,
_C(D)on 10, _
OC(0)R", -NO2, =0, =S, =N(R"), -CN, C3-12 carbocycle, and
3- to 12-membered heterocycle; and C3-12 carbocycle, and 3- to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in R4 is
optionally substituted with one or more substituents independently selected
from
halogen, -OR',

ts_
_ C(0)N(Rm)2, -N(Rw)C(0)R"), -
N(R1 )C(0)N(R10)2, _m(R10)2,
_C(D)R1o, -,c(c)Or.K lo, _
OC(0)R", -NO2, =0, =S, =MR"), -CN, C1-6alkyl, C2-6 alkenyl,
and C2-6 alkynyl;
R5 is selected from unsaturated C4-8 carbocycle; bicyclic carbocycle; and
fused
5-5, fused 5-6, and fused 6-6 bicyclic heterocycle, wherein R5 is optionally
substituted
and wherein substituents are independently selected at each occurrence from:
halogen, _ORto, _sn to, _
C(0)N(R112, -N(R")C(0)R",
-NOVIC(C)N(Rto)2, _
C(0)Rm, -C(0)011", -0C(0)11", -NO2, 0,-S,
=TAR"), and -CN;
Ct-w alkyl, C2-10 alkenyl, C2-10 alkynyl, each of which is optionally
substituted with one or more substiments independently selected from halogen, -
OR",
-SR` , 2 -C(0)N(R113)µ,
TARM)C(C)R1- , - N(WIC(0)N(R10)2,
2
_N(R10)µ, _ C(0)Rt ,
-C(0)0R", -0C(0)Rm, -NO2, =0, =S, =N(11"), -CN, C3-12 carbocycle, and 3- to 12-

membered heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, wherein each C3-12
carbocycle, and 3- to 12-membered heterocycle in R5 is optionally substituted
with one
or more substituents independently selected from halogen, -OR", -SR", -
C(0)N(Rt )2,
_mRtoycor 10, _
N(Rm)C(0)N(Rn2, -N(R")2, -C(0)R", -C(0)0R", -00(c)Rto,
-NO2, =0, =S, =N(R"), -CN, Cboalkyl, C2-6 alkenyl, and C2-6 alkynyl;
It" is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
334
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, C1-10 alkyl, -C t- to haloalkyl, -0-C 1-10
alkyl, C2-10
alkenyl, C2-to alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle, and
haloalkyl;
and
R12 is independently selected at each occurrence from halogen, -Or,
_sRto, _N(Rio)2, _icor , _ to C(0)N(R10)2, _N(R10)Cor _ 10,
K
C(C)OR10, -0C(0)Rm,
-S(0)Rm, -S(0)2R10, -P(0)(0111 )2, -0P(0)(0141 )2, -NO2, =0, =S, =N(Rm), and -
CN;
Ct-to alkyl, C2-10 alkenyl, C2-to alkynyl, each of which is optionally
substituted with one
or more substituents independently selected from halogen, -ORm,
-N(R1 )2,
_C(D)Rto, -,c(c)mR10)2, _NRIonor , _ to
K
0(0)ORW, _Gc(0)R10, _soy.K _
S(D)2R10,
2
_PoxiaR10,),
-0P(0)(010 )2, -NO2, =0, =S, -N(R1 ), -CN, C3-10 carbocycle and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-OW ,
mR)2,to _ -C(0)R1 , -C(0)N(Rto)2, -N(R1 )C(0)R1
, -C(0)0R10,
-0C(0)R1o, _sor to, _
S(0)21:01, -P(0)(0R1 )2, -0P(0)(01e)2, -NO2, =0, =S,
=N(R1 ), -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl;
wherein any substitutable carbon on the benzanpine core is optionally
substituted by a substituent independently selected from R12 or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle;
R20, R21, R22, and E. n23
are independently selected from hydrogen,
halogen, at , _(Rt0)2, _s(0)Rto, _s(0)2Rto,
-,c(c)Rto, _c(c)loRto,
-0C(0)R1 , -NO2, =0, =S, =N(111 ), -CN, Ct-to alkyl, C2-10 alkenyl, and C2-10
alkynyl;
and
24
n
K. and R25 are independently selected from hydrogen, halogen, -ORD',
-SRm, - N(R1 )2, -S(0)Rm, -S(0)2R10, _C(D)R10, _C(o)on 10, _
OC(0)R1 , -NO2, =0, -S,
_Notto,, _
CN, Ci-to alkyl, C2-to alkenyl, and C2-to alkynyl; or R24 and R25 taken
together
form an optionally substituted saturated C3-7 carbocycle.
335
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
70.
The method of any one of claims
57-69, wherein the TLR8 agonist is a
compound of Category A, Formula IIC:
R1
N¨R2
co N ,
R5 0
-----
L2-R4
(IIC)
or a pharmaceutically acceptable salt thereof, wherein:
RI- and R2 are hydrogen;
L2 is -C(0)-;
R4 is -N(R1. )2;
RR' is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0C1tC6H5; and Chto alkyl, C2-10 alkenyl, C2-io alkynyl, C312 carbocycle,
and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0CH2C6H5, -NI-1C(0)0CH2C6H5, Ci-io alkyl, -Ct-io haloalkyl, -0-C 1-10
alkyl, C2-lo
alkenyl, C2-10 alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle, and
haloalkyl;
Lio
is -C(0)N(R'-
) *, wherein * represents where Lm is bound to R5; and
R5 is a fused 5-5, fused 5-6, or fused 6-6 bicyclic heterocycle, wherein R5 is

optionally substituted and wherein substituents are independently selected at
each
occurrence from:
halogen, -ORM, _SRI , _C(D)TARICI)2, _NotiowecoRio,
-N(R10)C(0)N(R10)2, -
N(R.....,)2, _
1n C(0)R10, -0(0)0R1-
(1, -00(0)R1n, -NO2, 1:7:0, =S,
=N(11.1 ), and -CN;
CI-io alkyl, C2-10 alkenyl, C2-10 alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-OW ,
am, -,c(c)N(Rio)2, _N(Rio),c(0)Rio, 2
_TARilicopk-Rio,), _ N(R"))2, -C(0)R1 ,
-C(0)0Rm, -0C(0)10 , -NO2, =0, =S, =MR"), -CN, C3-12 carbocyde, and 3- to 12-
membered heterocycle; and
336
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
C342 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
halogen, -Ole, -SRI , -C(0)N(11.1 )2, -N(11.1 )C(0)Ri , -N(R1 )C(0)N(RIO)2, -
N(R10)2,
_Cow , _Cowl , -0C(0)R1 , -NO2, =0, =S, =N(R11, -CN, C 1-6alkyl, C2-6 alkenyl,

and C2-6 alkynyl.
71. The method of claim 70, wherein R4 is -N(Ci4 alky1)2 and LI is
-C(0)N(1-1)-*.
72. The method of claim 70 or 71, wherein:
CCH3
v.NI
R4 is CH3.
73. The method of any one of claims 57-72, wherein the TLR8 agonist is
selected from:
o
els .
N
N
NH2 040
0
N NH,
0 N *
H
-- r--\
N
OH
es . N NH2 00
0 Nõ. NH2
OH M *
-- /-----\
N
H
' iTh
0 N\---\\
0
NH * 0
NH2 HN 41) 0
NH2
11
N
N
*1-1
--- re\
N *
--
r-s..
N
N
0 \----\
0 \----\
337
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
. 0
40 0
N.

NH2 NH2
HN
N 1101
N
H
N
H
__.- cs,
--- NiTh
N
0 \Th
0 \Th
, ,
=
o AO N
N__ NH2
0
. 1 * 0A N 011
0 NH2
N,
--- .----\\
ri
--- Nr---\
0
0
4 0 NH2
crsi 10
N H2
.õN _ N,
Cbz N N.
*
N
H
H N r-,,N,
NH2 --- NC\
0 µ---\
0 \--\
,
,
cp N
NH2
ca. 0
N NH2
N
N
H H2N
H
NHCbz ---
N/Th --- r---\
N
,
,
c: C.).,
0.1
N.__ NH2
N
N
CbzHN H H
r--\\ H2N
--
r--s\
N N
,
raN....x
es, 0 N___ NH2
* N ".... 1 N NH2
N
N
HNs. H
H
Cbz -- /Th
--- Nr\
N
,
,
H
oCi
CbleN
N
se I NH2 I CI N
H2
HN --...
N
N
H
H
--- 1--\
--- r's\
N
N
N
ad -,._.
_C ..Ø,
HN I
N N
H
---
Nr---\
0 \----\
,
338
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H
e
NH2
s 0,
RB *
N,
N
HO H
---
rThsi
N
,
NH2
0 0 ....1 rAMN Cl N__
u 11 *
0
--- r---\
N
0
\---\ ,
CbzliN,i o
t.... pi so
......N.,......õ ,
ri,11........4...ri
* N, NH2
---
r---\
0 N\--\ ,
H2N...1
0
LN
* OCX H CI NH2
N
...."' 0 N,
...õ
r--\
N
N
N
C., 0
1 '%. 0
NH2 NH2
HIWN. N Ha:1.N N,
VI lb
H 001
H3C0 r---\
F --- Nr-\
N
0 \--\
0 \---\
0
H2N-----r N = ratil 0
NH2
F F H N
....-- pd N,
ii 0
---
ra- \
N
\
0 --\
and
, and
pharmaceutically acceptable salts thereof.
339
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
74.
The method of any one of claims
57-73, wherein Dx is a compound of
Formula (IVB):
R1
R2o
x
N-R2
R8,c2 N--
-- R25
--- R24
R21
22
L22-R4
R R23
(IVB)
or a pharmaceutically acceptable salt thereof, wherein:
Lu is selected from -X3-, -X3-C1-6alkylene-X3-, -X3-C2-6alkenylene-X3-, and -
X3-C2-6 alkynylene-X3-, each of which is optionally substituted on alkylene,
alkenylene,
or alkynylene with one or more substituents independently selected from R12;
L22 is independently selected from -r-, -r-Ci.-6 alkylene-r-, -X4-C2-6
alkenylene-r-, and -X4-C2-6 alkynylene-r-, each of which is optionally
substituted on
alkylene, alkenylene, or alkynylene with one or more substituents
independently
selected from Rw;
X3 and X4 are independently selected at each occurrence from a bond, -0-, -S-,
-N(Rm)-, -C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(Rn-, -C(0)N(It'c)c(0)_,
-C(C)N(Rm)C(C)N(R"))-, -N(R')C(0)-, -N(Rt5C(0)N(R")-, -N(t' )C(0)0-,
-0C(0)N(Rw)-, -C(NR` )-, -N(Rw)C(NIV )-, -C(NR"))N(R")-,
-N(Rw)C(NRI1N(Rw)-, -S(0)2-, -0S(0)-, -S(0)0-, -S(0)-, -OS(0)2-, -S(0)20-,
-N(RnS(0)2-, -S(0)2N(Rth)-, -N(RnS(0)-, -S(0)N(Rm)-, -N(Rm)S(0)2N(R'o)_, and
-N(RnS(0)N(10 )-;
RI- and R2 are independently selected from L3, and hydrogen; and Chio alkyl,
C2-
alkenyl, and C2-10 alkynyl, each of which is optionally bound to L3 and each
of which
is optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SRI , -C(0)N(Rm)2, -N(Rn2, -S(0)Rw, -S(0)2Rm, -C(0)Rw,
-C(0)0Rm, -0C(0)11.1 , -NO2, =0, =S, =N(R1 ), and -CN;
R4 and R8 are independently selected from: -Ole , -N(R10)2, -C(C)N(R"))2,
340
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
-C(0)Rm, -C(0)0R1- , -S(0)R'', and -S(0)2Rm; C1-10 alkyl, C2-10 alkenyl, C2-to
alkynyl,
each of which is optionally bound to L3 and each of which is optionally
substituted with
one or more substituents independently selected from halogen, -OW , -SRI ,
-C(0)N(R10)2, 4õ4(R10),c(0)Rto,
-N(R10)C(0)N(R10)2, -Nen ttc10)2, -C(0)11.1 , -C(0)0Rm,
-0C(C)RY), -NO2, =0, =S, =N(Rw), -CN, C3-12 carbocycle, and 3- to 12-membered
heterocycle; and C3-12 carbocycle, and 3- to 12-membered heterocycle, wherein
each
C3-12 carbocycle, and 3- to 12-membered heterocycle in R4 and R8 is optionally
bound
to L3 and each C3-12 carbocycle, and 3- to 12-membered heterocycle in re and
11.8 is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SR', -C(0)N(R"))2, -N(RnC(0)Rw, -N(R'')C(0)N(Rt )2,
,n10
-N(R"))2, -C(0)Kõ -C(0)0R11), -0C(0)Rffi, -NO2, =0, =S, =N(R'"), -CN, 0.-6
alkyl,
C2-6 alkenyl, and C2-6 alkynyl;
Ril) is independently selected at each occurrence from L3, hydrogen, -NI12,
-C(0)0CH2C6H5; and Chia alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12 carbocycle,
and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NH2, =0, =S,
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, Ci-toalkyl, C2-10 alkenyl, C2-to alkynyl, C3-
12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
L3 is a linker moiety, wherein there is at least one occurrence of
and
RI-2 is independently selected at each occurrence from halogen, -OR', -SR',
-N(Rw)2, -C(0)Rto, -C(0)N(R11)2, -N(Rw)C(0)10.0, -C(0)0Rw, -0C(0)Rw, -S(0)Rw,
-S(0) K10, - P(0)(0R10)2, -OP(0)(OR112, -NO2, =0, =S, =MR"), and -CN; C1-10
alkyl,
C2-10alkenyl, C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -ORM, _SR10,
2
_N(R113µ), _ C(0)Rm,
-C(0)N(Rm)2, -N(R'orroyn 10,
-C(0)ORKI, -0C(0)Rw, -S(0)Rm, -S(0)2R"),
2
_p(p)(0R10µ),
-0P(0)(0R10)2, -NO2, =0, =S, =N(RID), -CN, C3-10 carbocycle and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocyde and 3- to 10-membered heterocycle init.' is
optionally
substituted with one or more substituents independently selected from halogen,
-OW ,
341
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
-SW , -MR112, -C(0)Rt0, -C(0)N(R10)2, -N(11 )C(0)R1-0, -20)0R11),
-0C(0)RM, -SOW"), -S(0)2R10, -P(0)(0R10)2, -0P(0)(0R10)2, -NO2, =0, =S,
=N(1.1 ), -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl;
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from RI' or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle;
R20, R21, lc .22,
and R23 are independently selected from hydrogen, halogen,
-SR' , -N(Rm)2, -S(0)Rw, -S(0)2Rm, -C(0)Rm, -C(0)0Rw, -0C(0)Rm, -NO2,
=S, =N(Rm), -CN, Ct-to alkyl, C2-to alkenyl, and C2-10 alkynyl; and
R24, and R25 are independently selected from hydrogen, halogen, -OR', -Sle ,
-N(Rn2, -S(0)2Rt , -C(0)Rt , -C(0)OR', -0C(0)Rm, -NO2, =0, =S,
=Me), -CN, Ci-to alkyl, C2-to alkenyl, and C2-to alkynyl; or R24 and R25 taken
together
form an optionally substituted saturated C3-7 carbocycle.
75.
The method of any one of claims
57-74, wherein Dx is a compound of
Formula (1VC).
R1
N
L12 N -R2,
R8 =
-
a--
L22--R4
(IVC)
or a pharmaceutically acceptable salt thereof, wherein:
IV and R2 are hydrogen;
L22 is _C(0)-
R4 is -N(Rm)2;
Rm is independently selected at each occurrence from hydrogen, -Nth,
-C(0)0CH2C6H5; and CI-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NH2, =0, =S,
-C(0)0C1tC6H5, -NHC(0)0CH2C6H5, C1-10 alkyl, C2-to alkenyl, C2-to alkynyl, C3-
12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
342
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
L1-2 is -C(0)N(Rto)*_,
wherein * represents where lan is bound to RK;
Rg is an optionally substituted fused 5-5, fused 5-6, or fused 6-6 bicyclic
heterocycle bound to linker moiety 12 ,
and wherein optional substituents are independently selected at each
occurrence
from:
halogen, -OR", -SRI , -,c(c)mRtc)2p., _
N(R1 )C(0)R10,
-N(Rm)C(0)N(Rm)2, -N(R10)2, -C(0)R10, -C(0)0R11 , -0C(0)11.1 , -NO2, :20, =S,
=MR"), and -CN;
Ci-io alkyl, C2-10 alkenyl, C2-10alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-OR",
am, _Conkto)z, _Nothc(0)Rio, _mkto)C(3)Nt-Rto.,2, _
N(R")2., -C(0)R",
-C(0)0R", -0C(0)R", -NO2, =0, =S, =N(R"), -CN, C3-12 carbocycle, and 3- to 12-
membered heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
halogen, -OR", -SR", -C(0)N(R")2, -N(10)C(0)R", -N(R1 )C(0)N(R")2, -N(R113)2,
-C(D)RM, -C(0)0R10, -0C(0)Rto, -NO2, =0, =S, =MR"), -CN, C1-6alkyl, C2-6
alkenyl,
and C2-6alkynyl.
76. The method of claim 75, wherein R4 is -N(C1-1 alky1)2 and I,' is
-C(0)N(1-1)-*.
77. The method of claim 75 or 76, wherein:
CCH3
R4 is CH3.
343
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
78.
The method of any one of
claims 57-77, wherein L and Dx together have
a structure selected from:
FuN ,,,r0
HN
0
caõ.....õ,....a....õ...õ...Thr NHxitiliril
IssCRX N
N..... 0
H
NH2
0 0 00
0,.....,[=11 .,,...1)..... N N,
11
H
N
0 \----\ ,
H2N tO
HN
0
N
RX*Wir 111 JL- r%)Y[sl
NH2
- H
N,
0 ......7..õ... 0 410
11
H
0
---
N
0
(Ni 0
0
N ciF1 0 0Aflr" ' H
N
H 0
Hy ri
H2N--.0
,
N
0
0 NH2
0 0N-----yG
A
N N.__
0 Xllii 0
----
U
---
vRX:`,...............õ,.....õ.õ..A. N......",
H 0 H
N - N
H - H
is¨\ .
N
Of
HN
H2W-LO
,
Ill .,.14,.1/4
0
0
--11--.
11-4LA , NH2
V
0 yiiii 0 40 0 N
N NI
N N...,N
H 0 H
-
H H
N
0
NH
0.4-NH2
,
344
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
0
= N
0
A.
II ..... _LI' NH2
0 0 0 0 N
N
H
H
VRilerisL}L
ri 0
- N
C\
N H
E H
0 -s......L
NH
OA N H2
,
H 2N tO
H N
1:11ily,11
1.-Rir%----...--%---.Thre5
1;1
N
0
H
0 0
0 = yr41,...........\ %II 0 1,...aaN 0
NH2
0
N ,--- N,
N
H
- r- \
N
H2NtO
HN
0
RX iril
ll
r H 0 4 H
0 õAõ.....
OyN,,,---,N so n 1µ1,-41 0
H
0
IN N ......../A, N NH2
H
-=-" rThs.
N
H2Ny0
HN
0
1(11xy
11:5iRli
ARXC..............hµ'''...Thr N
0
H H
0 0 mit OyN....,....Thq
HAD N 0
.VI 0
NH2
c.f....A.' ..N
N,
H
345
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
H2Ny0
HN
=
N.
0
H

0
N
I), 0
NI-12
0 \¨\
I-12N tO
HN
H
es" NXLIN I
0 0 *
0
N
NU N
NH2
---
re-\
0
H2Ny0
HN
0
z H
- N 0 411 0 0
0
N
'%1 0
NNH2
N,
H2N-f0
HN
H
Nxillik
NH2
0 le 0.õ,e.NOUN
N

,
0
/Th
0
346
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N y0
HN
N= co% 0
NH2
H_ 0
0
0
/---\\
o
\-\
o H o
o Rt1 o NH2
F F
07111
HN
0 N\--Ths1/4
Fl2tr1/40
and
=
NJL
rXrri op) Ainn
CC) NH2
F F
N
H H
o
HN
H2NA0
and salts thereof,
wherein the RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide
moiety bound to a residue of an antibody,
wherein )4 on RX* represents the point of attachment to the residue of the
antibody.
347
CA 03151322 2022-3-15

WO 2021(067644
PCT/1JS2020/053870
79. The method of claim 78, wherein L and
Dx together have a structure
selected from:
H2N,r0
HN
H H
tLIRX"Wir NY% )YN0 H 0
0 . 1 0
Oy N .,......,..-NN * rõ... Nõ,...ti 0
H NH2
0
W. N Nõ
H
----
Csµ,
N
H2Ny0
HN
_Ai,:
iscR,,,N. N
0 411
o
z H H
0 ......A......
OyN..........--.1,4 * r,..,L N,.AN...., 0
H N,NH2
0
Nj
H
N
H2NtO
HN
H.2:111( H
N N
N
ARX<--.%-------------ir N
N NH2
H
0 s 0
,
II
H
0
/---\
.--
N
H2N yp
HN
H H
/....Rn N 4.N N 5
r....m.....i 0
I
NH2
0 ,,..5...... H 0
0............ NI j4-,--,..AN N,
H
H
.---
N
0
o
o
A
crii 0 ri2c11 * CO, N NH2
N aN.d.AN
N ...-- N,
H H
H
0
rTh---- .
N
11
H2N 0
2
348
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
and
0
0
Rxz_eõ....õ%.õ....õ..)fir NH it, 40 0 friC
so , 0 NH2
F F
- N
H E H
Nrs
HN
0 \
H N
and salts thereof,
wherein the RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide
moiety bound to a residue of an antibody,
wherein
on RX* represents the point of
attachment to the residue of the
antibody.
80. The compound or salt of claim 78 or 79, wherein RX* comprises a
succinamide moiety and is bound to a cysteine residue of the antibody.
81. The compound or salt of claim 78 or 79, wherein RX* comprises a
hydrolyzed succinamide moiety and is bound to a cysteine residue of the
antibody.
82. The method of any one of claims 57-81, wherein:
(a) L and Dx together have a structure selected from:
H2N,re
HN
H
Rxcwir Nt4\ N
0 IS
0
0 = y
NH,
0N
14_
=
LJÇJ
0
and
349
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
H2NTO
HN
0
ARACWY
0 140 0Nal. .N
NI, NH2
0
0
0
\--Ths =
and salts thereof;
(b) the additional therapeutic agent comprises trastuzumab; and
(c) optionally, wherein the antibody of the conjugate comprises 11C-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
83. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
H2NtO
HN
H
0
0
NH2
0
OyN....õ====,N *
(Yi 0
N
0
and
350
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N
tO
HN
0
15ck\111
0 0
0 SO 0..._,NIOLIN N NH2,
11
0
fe-\\
0 \¨\\
and salts thereof;
(b) the additional therapeutic agent comprises fam-trastuzumab
deruxtecan-nxki; and
(c) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
84. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
H2Ny0
HN
H oH
O hR)CiN N
0
0 H 0.,1e-N4
0
NH2
N._
rThs.
and
351
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2N TO
HN
0
0 40/ONOL3LN
NçN H2
0
11
0
0
=
and salts thereof;
(b) the additional therapeutic agent comprises ado-trastuzumab
emtansine; and
(c) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ 1D NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
85. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
stmcture selected from:
HAtp
HN
H
411
0
O 0 Oy
(...1114t.:1
NH2
LJJ
0
0 \
and
352
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
H2Ny0
HN
H
Rr X
H Wy NIL N
NH2
0 0 411
N.,
0
and salts thereof;
(b) the additional therapeutic agent comprises tucatinib; and
(c) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, 11C-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
86. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
1-12Ny0
HN
0
ERX'r!xi\ 1:11
HN 0 Fl
0 O OyN.aN
N'", 0
NH2
N,
r¨Ths,
0
and
353
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
H2Ny0
HN
H
NH2
0 HNi 0 411
NL_
o
and salts thereof;
(b) the additional therapeutic agent comprises palbociclib,
abemaciclib, or ribociclib;
(c) optionally wherein the HER2-expressing cancer is breast cancer;
and
(d) optionally, wherein the antibody comprises HC-CDR1, HC-
CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ LD NOs: 1-6,
respectively; or the antibody comprises a heavy chain variable region
comprising the
amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising the
amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
87.
The method of any one of claims 57-81, wherein:
(a) L and Dx together have a
structure selected from:
1-12Ny0
HN
0
0 HN 0 = y
so ,
0N
N,
r`ss,
0
and
354
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
H2Ny0
HN
H
H
N H2
0 411 N.,
0
0
and salts thereof;
(b) the additional therapeutic
agent comprises:
(i) trastuzumab;
(ii) a pyrimidine analog, optionally wherein the pyrimidine
analog is fluorouracil or capecitabine;
(iii) a platinum coordination complex, optionally wherein the
platinum coordination complex is oxaliplatin, or cisplatin;
(iii) a PD-1 and/or PD-Ll
inhibitor, optionally wherein the
PD-1 and/or PD-Ll inhibitor is pembrolizumab, nivolumab, or atezolizumab;
(c) optionally wherein the HER2-
expressing cancer is gastric cancer;
and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
88. The method of any one of claims 57-81,
wherein:
355
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(a) L and Dx together have a
stnrcture selected from:
H2NtO
HN
H o
hRX
0
0 o len
iso 0
NH2
0
Nre\
and
H2N yO
HN
0
lilt\ 11
A Rn
0 401
NI1

2
0
0
NI
0
\----\\ =
and salts thereof;
(b) the additional therapeutic
agent comprises:
(i) trastuzumab;
(ii) a chemotherapy regimen comprising lencovorin,
ftuorouracil, and oxatiplann (FOLFOX); and
(iii) optionally pembrolizumab, nivolumab, or atezolizumab;
(c) optionally wherein the HER2-
expressing cancer is gastric cancer;
and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
356
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
89. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
H2Ny0
FlN
11 411
0
0 kitil\O-1 0-õwe.R.,--
.,N
-..
NH2
and
H2Ny0
HN
mx\
0 S oNOLì 0
NH,
N,
0
rTh,
0
\---\ =
and salts thereof;
(b) the additional therapeutic
agent comprises:
trastuzumab;
(ii) leucovorin,
(iii) fluorouracil:
(iv) cisplatin; and
(iii) optionally
pembrolizumab, nivolumab, or atezolizumab;
(c) optionally, wherein the HER2-
expressing cancer is gastric
cancer; and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
357
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
90. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
H2N
tO
HN
H
hR)C<WIt\i
N."
m 0 u OyNN*
NH2
0N
N,
0
and
H2Ny0
HN
LitO\
0
NH2
0
NCO,
0
\---\ -
and salts thereof;
(b) the additional therapeutic
agent comprises:
trastuzumab;
(ii) capecitabine;
(iii) oxaliplatin; and
(iv) optionally pembrolizumab, nivolumab, or atezolizumab;
(c) optionally, wherein the HER2-
expressing cancer is gastric
cancer; and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
358
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
91. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
H2N
tO
HN
H
x11:5-y.11 ries,
iF0err)r-N
0 W oy
*
NH2
0
LÇÇN
0
and
H2Ny0
HN
H
0
NH2
0
and salts thereof;
(b) the additional therapeutic
agent comprises:
trastuzumab;
(ii) pembrolizumab, nivolumab, or atezolizumab; and
(iii) optionally capecitabine;
(c) optionally, wherein the HER2-
expressing cancer is gastric
cancer; and
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
359
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
92. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
structure selected from:
1-12Nto
HN
H
l'IRXWYNYC\
O " 0
* nti,,zi
NH2
0
N,
0
and
112N tO
HN
Hi\iõH
0
NH2
0 H 0
0õ.Nia.-1/4.,1,N N,
0
r---
0
\--\ =
and salts thereof;
(b) the additional therapeutic
agent comprises:
trastuzumab;
(ii) pembrolizumab;
(iii) docetaxel or paclitaxel; and
(iv) optionally, atezolizumab;
(c) optionally, wherein the HER2-
expressing cancer is breast cancer;
and
360
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(d) optionally, wherein the
antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
93. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a structure selected from:
H2N tO
HN
H
*
0
0 0
* 14.zzl 0
NH2
N
N,
0
and
H2Ny0
HN
H H
0 H 0 le
N__ N H2
11
and salts thereof;
(b) the additional therapeutic agent comprises one of the following
combinations:
fam-trastuzumab deruxtecan-nxki and durvalumab,
(ii) fam-trastuzumab deruxtecan-nxIci and atezolizumab,
(iii) ado-trastuzumab emtansine and durvalumab; and
361
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/1JS2020/053870
(iv) ado-trastuzumab emtansine and atezolizumab;
(c) optionally wherein the HER2-expressing cancer is breast cancer;
and
(d) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
94. The method of any one of claims 57-81,
wherein:
(a) L and Dx together have a
stmcture selected from:
H2N,r0
HN
H
0
0 o
Ca.
NH
0
N 2,
\
0 NI\
and
H2Ny0
HN
HAH
ess,,RnN
= 0
0 H 0
,,,,NOLI N,
0
NH2
11
0
0
-
and salts thereof;
(b) the additional therapeutic
agent comprises:
(i) trastuzumab;
362
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
(ii) a hormone-based
compound; and
(ii) palbociclib or
abemaciclib;
(c) optionally, wherein the HER2-expressing cancer is breast cancer;
and
(d) optionally, wherein the antibody of the conjugate comprises HC-
CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs:
1-6, respectively; or the antibody comprises a heavy chain variable region
comprising
the amino acid sequence of SEQ ID NO: 7 and a light chain variable region
comprising
the amino acid sequence of SEQ ID NO: 8; or the antibody comprises a heavy
chain
comprising the amino acid sequence of SEQ ID NO: 9 and a light chain
comprising the
amino acid sequence of SEQ ID NO: 10.
95. The method of any one of claims 57-94, wherein the antibody of the
conjugate comprises an antigen binding domain and an Fc domain.
96. The method of claim 95, wherein the Fc domain exhibits the same or
substantially similar binding affinity to at least one Fcgamma receptor and/or
an FcRn
receptor as compared to a wild-type IgG1 Fc domain.
97. The method of claim 95, wherein the Fc domain has increased binding
affinity to at least one Fcgamma receptor as compared to a wild-type IgG Fc
domain.
98. The method of any one of claims 95-97, wherein the Fc domain is an
IgG1 Fc domain.
99. The method of any one of claims 57-98, wherein the method comprises
administering an effective regimen of the conjugate, wherein the effective
regimen
results in a Tmax of the conjugate in the subject of greater than 4 hours
following each
administration of the conjugate.
363
CA 03151322 2022-3-15

WO 2021/067644
PCT/1JS2020/053870
100. The method of any one of claims 57-99, wherein the method comprises
administering an effective regimen of the conjugate, wherein the effective
regimen
comprises at least two cycles of administration of the conjugate to the
subject and a
total dose of greater than 0.4 mg/kg of the conjugate per cycle.
101. The method of any one of claims 57-100, wherein the conjugate is
administered subcutaneously.
102. The method of any one of claims 57-100, wherein the conjugate is
administered intravenously.
103. The method of any one of claims 57-102, wherein the HER2-expressing
cancer is selected from breast cancer, stomach/gastric cancer, colorectal
cancer, non-
small cell lung cancer (NSCLC), urothelial cancer, endometrial cancer, and
ovarian
cancer.
364
CA 03151322 2022-3-15

Description

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


WO 2021/067644
PCT/US2020/053870
COMBINATION THERAPY WITH IMMUNE STIMULATORY CONJUGATES
STATEMENT REGARDING SEQUENCE LISTING
The Sequence Listing associated with this application is provided in text
5 format in lieu of a paper copy, and is hereby incorporated by reference
into the
specification. The name of the text file containing the Sequence Listing is
860234 412W0 SEQUENCE LISTINGixt. The text file is 9 KB, was created on
September 17, 2020, and is being submitted electronically via EFS-Web.
BACKGROUND
Technical Field
100011 The present application relates to combination therapies comprising
immune-stimulatory conjugates and methods for treating cancer or viral
infections.
Description of the Related Art
100021 One of the leading causes of death in the United States is cancer.
15 Conventional methods of cancer treatment, like chemotherapy, surgery, or
radiation
therapy, tend to be highly toxic and/or nonspecific to a cancer, resulting in
limited
efficacy and harmful side effects. The immune system has the potential to be a

powerful, specific tool in fighting cancers. This observation has led to the
development
of immunotherapeutics as drug candidates for clinical trials.
Immunotherapeutics can
20 act by boosting a specific immune response and have the potential to be
a powerful
anti-cancer treatment. Such immunotherapeutics may comprise benzazepine
compounds, which in some instances, act as toll-like receptor (e.g., TLR8)
agonists.
INCORPORATION BY REFERENCE
100031 All publications, patents, and patent applications mentioned in this
25 specification are herein incorporated by reference to the same extent as
if each
individual publication, patent, or patent application was specifically and
individually
indicated to be incorporated by reference.
1
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
BRIEF SUMMARY
[0004] The present disclosure provides tumor-associated antigen- or liver cell

antigen-dependent uptake of a Toll-like receptor (TLR) agonist, which allows
for
systemic delivery of an innate immune agonist that generates tumor- or liver-
localized
5 immune cell activation while avoiding systemic toxicity. In some
embodiments, a
method of treating cancer or viral infection is provided, comprising
administering to a
subject with cancer or viral infection a conjugate comprising a TLR agonist
and an
antibody that binds a tumor-associated or liver cell antigen, and an immune
checkpoint
inhibitor. In some embodiments, the immune checkpoint inhibitor inhibits PD-1
and/or
10 PD-L1. In some embodiments, the TLR agonist is a TLR8 agonist.
[0005] In certain embodiments, a method of treating cancer or viral infection
is
provided, comprising administering to a subject with cancer or viral infection
a
conjugate and an immune checkpoint inhibitor that inhibits PD-1 and/or PD-L1;
wherein the conjugate is represented by Formula (I):
= 4> Dx
15 Z
(0;
wherein:
A is an antibody that binds a tumor associated antigen or a liver cell
antigen,
L is a linker;
Dx is a TLR8 agonist, wherein the TLR8 agonist is a benzazepine compound;
20 n is selected from 1 to 20; and
z is selected from 1 to 20.
[0006] In further embodiments, a method of treating a HER2-expressing cancer
is provided, comprising administering to a subject with a HER2-expressing
cancer a
conjugate and an additional therapeutic agent comprising a HER2-targeted agent
such
25 as a kinase inhibitor, a therapeutic agent comprising an anti-RER2
antibody, or both;
wherein the conjugate is represented by Formula (I):
2
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
4) = Dõ 1 ]
(0;
wherein:
A is an antibody that binds HER2,
L is a linker;
5 Dx is a TLR8 agonist, wherein the TLR8 agonist is a benzazepine
compound;
n is selected from 1 to 20; and
z is selected from 1 to 20.
BRIEF DESCRIPTION OF THE DRAWINGS
[0007] Certain features of the disclosure are set forth with particularity in
the
10 appended claims. A better understanding of the features and advantages
of the present
disclosure will be obtained by reference to the following detailed description
that sets
forth illustrative aspects, in which the principles of the disclosure are
utilized, and the
accompanying drawings of which:
[0008] FIGS. IA-1B show increased expression levels of CD86 (FIG. 1A) and
15 PD-Li (FIG. 1B) in dendritic cells following exposure to HER2-TLR8 in
the presence
of HER2-expressing tumor cells, as described in Example 1.
[0009] FIG. 2 shows IFN-y expression in human PBMCs co-cultured with 5IC-
BR-3 (3+ HER2+ tumor cell line), MDA-MB-453 (2+ HER2+ tumor cell line), or
MDA-MB-468 (HER2- tumor cell line) in the presence of HER2-TLR8 for 24 hours,
as
20 described in Example 2.
[0010] FIG. 3 shows the similarity in expression between TLR7 in mouse and
TLR8 in human dendritic cells and macrophages, as described in Example 3.
[0011] FIGS. 4A-4K show single agent efficacy of a HER2-TLR7 (mouse
surrogate for human HER2-TLR8) in an in vivo xenograft mouse model, as
described in
25 Example 4. The arrows indicate the day a dose was administered to the
mice. Slopes
of growth curves contrasting unconjugated HER2 mAb against HER2-TLR7 are
significantly different (p < 0.001) at each dose level.
3
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0012] FIGS. SA-5B show the upregulation of PD-Li on the surface of HER2-
expressing tumor cells following treatment with the HER2-TLR8 surrogate, HER2-
TLR7, 48 hours after: (FIG. 5A) a single dose or (FIG. 5B) the third of three
doses, as
described in Example 5.
5 100131 FIGS. 6A-6C show (FIG. 6A) 1P10 expression and 1FN-y
expression at
day 2 (FIG. 6B) and day 7 (FIG. 6C) in EMT6 tumors from mice treated once with

HER2-TLR7 mouse surrogate at 10mg/kg, unconjugated anti-HER2 antibody at
10mg/kg, isotype control antibody at 10mg/kg, each alone or in combination
with anti
PD-1 antibody, as described in Example 6.
10 [0014] FIGS. 7A-7F show spider plots of the anti-tumor response in
mice with
HER2+ tumors after treatment with (FIG. 7F) HER2-TLR7 in combination with an
anti-PD1 antibody, as compared to treatment with (FIG. 7A) combinations of
isotype
control antibodies (r1gG2a and mIgG2a, (FIG. 7B) mIgG2a/HER2 mAb, (FIG. 7C)
mIgG2a/HER2-TLR7, (FIG. 7D) anti-PD1/mIgG2a, or (FIG. 7E) anti-PDI/HER2 mAb,
15 as described in Example 7.
00151 FIGS. 8A-8B show the interactions of HER2-TLR8 and hz4D5 with
monomeric human HER2 ECD were evaluated using Octet Red 96TM in 2 orientations

FIG. 8A shows monomeric human HER2 ECD with 10x histidine tag immobilized to
penta-his sensor used to capture HER2-TLR8 until saturation and then tested
for
20 additive binding against itself, HER2-TLR8 (solid line), or hz4D5
(broken line).
Increased signal of hz4D5 is observed after ITER2-TLR8 has saturated HER2 ECD
indicates HER2-TLR8 and hz4D5 occupy different epitopes on HER2 EGO_ FIG. 8B
is
the same assay set-up as in FIG. 8A, except with hz4D5 captured until
saturation and
then tested for additive binding against itself, hz4D5 (broken line), or HER2-
TLR8
25 (solid line). Increased signal of HER2-TLR8 is observed after hz4D5 has
saturated
HER2 ECD additionally indicates HER2-TLR8 and hz4D5 occupy different epitopes
on
HER2 ECD.
[0016] FIG. 9 shows that the HER2-TLR8 agonist conjugate does not impede
the function of the trastuzumab-like antibody, hz4D5, of reducing tumor cell
viability in
30 vitro, as described in Example 9.
4
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0017] FIGS. 10A-10F. FIGS. 10A-10D show TNF-a production in co-
cultures of various HER2+ (FIGS. 10A-10C) or HER2- (FIG. 10D) tumor cell lines
and
PBMCs contacted with HER2-TLR8, unconjugated trastuzumab-like anti-HER2
antibody, and a combination of both, as described in Example 10. FIGS. 10E-10F
show
5 IFN-y expression in co-cultures of a HER2+ (FIG. 10E) or HER2- (FIG. 10F)
tumor
cell line and PBMCs contacted with HER2-TLR8, unconjugated trastuzumab-like
anti-
HER2 antibody, and a combination of both, as described in Example 10.
[0018] FIGS. 11A-11F show spider plots of an anti-tumor response in SC1D
mice with HER2+ NCI N87 tumors following treatment with 10 mg/kg HER2-TLR7,
10 matched unconjugated HER2 mAb, or isotype control antibody, alone or in
combination with the trastuzumab-like antibody, hz4D5. The arrows indicate
dose
administration.
[0019] FIGS. 12A-12F show that HER2-TLR7 surrogate in combination with
trastuzumab-like monoclonal antibody, hz4D5, results in enhanced efficacy over
the
15 single agents alone, as described in Example 12. Arrows indicate day
dose was
administered.
DETAILED DESCRIPTION
[0020] Immune checkpoint inhibition therapy has been largely ineffective as
single agent therapies in tumors, in part due to the absence of a T cell
infiltrate. An
20 abundant myeloid cell population within checkpoint refractory tumors
provides an
attractive target with the potential to expand and recruit tumor-specific
cytotoxic T
lymphocytes (CTLs). Single agent therapies for chronic viral infections, such
as
Hepatitis B virus (HBV) and Hepatitis C virus (HCV), have been equally
ineffective.
The present disclosure demonstrates for the first time that in vivo treatment
with a
25 TLR8 agonist, when conjugated to an antibody, drives anti-tumor or anti-
viral
immunity, which in turn results in PD-Li upregulation, a negative regulator of
T cell
activation. This PD-Li induction is expected to be in the tumor
microenvironment or in
infected liver. The combination of immune checkpoint inhibitor and TLR8
agonist
antibody conjugate results in a more profound T cell response as demonstrated
by
30 enhanced 1FN-y production and ultimately enhanced efficacy compared to
either agent
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
alone. In addition, in the context of an immune competent tumor-bearing mouse,
a
TLR7 agonist (used as a TLR8 agonist surrogate) conjugated to an anti-HER2
antibody
caused an expansion in intra-tumoral, neo-antigen specific CD8+ T cells and
robust
myeloid cell activation. In the case of HER2+ disease and given the current
standard of
5 care for patients with HER2+ disease often includes HER2-directed agents
that show
clinical benefit, the ability of a TLR8 agonist conjugated to an anti-HER2
antibody to
combine with HER2 directed agents to further lower tumor burden, as provided
in the
present disclosure, is expected to be clinically useful.
[0021] Additional aspects and advantages of the present disclosure will become
10 apparent to those skilled in this art from the following detailed
description, wherein
illustrative aspects of the present disclosure are shown and described. As
will be
appreciated, the present disclosure is capable of other and different aspects,
and its
several details are capable of modifications in various respects, all without
departing
from the disclosure. Accordingly, the descriptions are to be regarded as
illustrative in
15 nature, and not as restrictive.
[0022] As used herein, a "tumor associated antigen" or "tumor antigen" refers
to
an antigen present on a cancer cell that can be recognized by an antibody and
is
preferentially present on a cancer cell as compared to normal (non-cancerous)
cells.
[0023] As used herein, the term "antibody" refers to an immunoglobulin
20 molecule that specifically binds to, or is immunologically reactive
toward, a specific
antigen The portion of the antibody that binds a specific antigen may be
referred to as
an "antigen binding domain." The term antibody can include, for example,
polyclonal,
monoclonal, genetically engineered, and antigen binding fragments thereof An
antibody can be, for example, murine, chimeric, humanized, a heteroconjugate,
25 bispecific, diabody, triabody, or tetrabody. An antigen binding fragment
can include,
for example, a Fab', F(a13')2, Fab, Fv, rIgG, scFv, hcAbs (heavy chain
antibodies), a
single domain antibody, Vim, VNAR, sdAbs, or nanobody.
[0024] As used herein, "recognize" refers to the specific association or
specific
binding between an antigen binding domain and an antigen. Specific association
or
30 specific binding does not require that the antigen binding domain does
not associate
6
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
with or bind to any other antigen, but rather that it preferentially
associates with or
binds to the antigen, as compared to association with or binding to an
unrelated antigen.
[0025] As used herein, "specifically binds" and the like refers to the
specific
association or specific binding between the antigen binding domain and the
antigen, as
5 compared with the interaction of the antigen binding domain with a
different antigen
(i.e., non-specific binding). In some embodiments, an antigen binding domain
that
recognizes or specifically binds to an antigen has a dissociation constant
(ICD) of <100
nM, <10 nM, <1 nM, <0.1 nM, <0.01 nM, or <0.001 nM (e.g. 104 M or less, e.g.
from
M to 10' M, e.g., from 104 M to 1043 M). Specific binding does not require
that
10 the antigen binding domain does not associate with or bind to any other
antigen, but
rather that it preferentially associates with or binds to the antigen, as
compared to
association with or binding to an unrelated antigen.
[0026] As used herein, an "Fc domain" refers to a domain from an Fc portion of

an antibody that can specifically bind to an Fc receptor, such as a Fcgamma
receptor or
15 an FcRn receptor.
00271 As used herein, a "myeloid cell" refers to a dendritic cell, a
macrophage,
a monocyte, a myeloid derived suppressor cell (VIDSC).
[0028] As used herein, an "antigen presenting cell" or "APC" refers to a cell
that can present antigen to a T-, or B-cell, in a productive manner leading to
activation
20 and/or expansion of T-, or B-cell clones specific for said antigen.
Nonlimiting
exemplary APCs include dendritic cells, macrophages, monocytes, and B cells.
In
some embodiments, an antigen presenting cell is a dendritic cell, a
macrophage, or a
monocyte.
[0029] As used herein, an "immune stimulatory compound" is a compound that
25 activates or stimulates an immune cell, such as a myeloid cell or an
APC.
[0030] As used herein, a "myeloid cell agonist" refers to a compound that
activates or stimulates an immune response by a myeloid cell.
[0031] As used herein, a "benzazepine compound" refers to small molecule
chemical compound comprising a benzazepine moiety, where the benzazepine
moiety is
30 a benzene ring fused to a 7-membered ring that comprises one or two
nitrogen ring
members. In addition to the bond where the ring is fused to the benzene ring,
the 7-
7
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
membered ring includes two double bonds (e.g., an azepine or diazepine ring),
one
double bond (e.g., a dihydroazepine or dihydro-diazepine ring), or no double
bonds
(e.g., a tetrahydroazepine, azepane, tetrahydrodiazepine, or diazepane ring).
The
benzazepine moiety is optionally substituted. In some embodiments, the
benzazepine
5 moiety is an optionally substituted 4,5-dihydro-3H-benzo[b]azepine. In
some
embodiments, the benzazepine moiety has the structure:
NH2
L2 -R
wherein = is a double bond or a single bond;
L2 is selected from -X2-, -X2-CI-6 alkylene-X2-, -X2-C24 alkenylene-X2-, and -
10 X2-C24 alkynylene-X2-, each of which is optionally substituted on
alkylene, alkenylene
or alkynylene with one or more Itn;
X2 at each occurrence is independently selected from a bond, -0-, -S-, -N(R10)-
,
-C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C( ) _ C(0)N(R1 )C(0)-,
-C(0)N(Rio)C(o)N(aio), _
ty) N(R1 )C(0)N(R1 )-, -N(R1 )C(0)0-,
15 -0C(0)N(Rio)Th _0(NRio)-,
) -C(NR")N(R1 )-,
-N(IVIC(NWIN(R1 )-, -S(0)2-, -0S(0)-, -S(0)0-, -S(0), -OS(0)2-, -S(0)20,
-N(R")S(0)2-, -S(0)2N(10 )-, -N(RnS(0)-, -S(0)N(R")-, -N(11.1 )S(0)2N(R1 )-,
and
le2 is independently selected at each occurrence from halogen, -OR", -SR",
20 -N(R1 )2, -C(0)R1 , -C(0)N(R1 )2, -N(R1 )C(0)R", -C(0)0R", -0C(0)R", -
S(0)R",
-S(0)210 , -P(0)(0R1 )2, -0P(0)(010 )2, -NO2, =0, =S, =N(R"), and -CN; Ci.-to
alkyl,
C2-10 alkenyl, C2-lo alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -OR", -SR", -N(R10)2, -
C(0)10 ,
-C(0)N(12.1 )2, -N(R")C(0)R1 , -C(0)011", -0C(0)R1 , -S(0)11.1 , -S(0)211.1 ,
25 -P(0)(oRich2
), -0P(0)(010 )2, -NO2, =0, =S, =N(R"), -CN, C3-10 carbocycle and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-OR",
-S111 , -N(R1 )2, -C(0)R10, -C(0)N(12.10)2, -N(11.1 )C(0)R", -C(0)011", -
0C(0)11.1 ,
8
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-S(0)Rm, -S(0)2R10, -P(0)(0111 3)2, -0P(0)(012.1 )2, -NO2, =0, =S, =N(Rm), -
CN, CI-6
alkyl, C2-6 alkenyl, C2.6 alkynyl;
R4 is selected from: -OW , -
N(Rto)2, _c(0)N(R10)2, _goAto, _C(0)0R1 ,
-S(0)11.1 , and -S(0)2R1 ; Ct-to alkyl, C2-10 alkenyl, C2-to alkynyl, each of
which is
5 optionally substituted with one or more substituents independently
selected from
halogen, -ORm, -C(0)N(R1 )2, -N(11.1
)C(0)Rm, -N(Rm)C(0)N(R1 )2, -
Nita 0)2,
-C(0)R1 , -C(0)0R1 , -0C(0)R1 , -NO2, =0, =S, =N(R10), -CN, C3-12 carbocycle,
and
3-to 12-membered heterocycle; and C3-12 carbocycle, and 3-to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in le is
10 optionally substituted with one or more substituents independently
selected from
halogen, -Ole , -Ste , -C(0)N(R111)2, -N(le )C(0)R10, -N(Rm)C(0)N(Rm)2, -
Ni(Ro)2,
-C(0)R1 , -C(0)0R1 , -0C(0)R1 , -NO2, =0, =S, =N(R10), -CN, C2-6 alkenyl,
and C2-6 alkynyl; and
le is independently selected at each occurrence from hydrogen, -N112,
15 -C(0)0CH2C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0C1tC6H5, -NHC(0)0CH2C6H5, Ci-to alkyl, -Ct-to haloalkyl, -0-C1-to alkyl,
C2-to
alkenyl, C2-to alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle, and
haloalkyl;
20 and the moiety is optionally substituted at any position.
[0032] A "small molecule" is an organic compound with a molecular weight of
less than 1500, or 100, or 900, or 750, 01 600, or 500 Daltons. In some
embodiments, a
small molecule agonist has an octanol-water partition coefficient (logP) in
the range of
from 3 to 6, or from 4 to 5, or from 2 to 4. In some embodiments, a small
molecule
25 agonist has a polar surface area of less than 200, or less than 150 A2
In some
embodiments, the small molecule agonist has not more than five, or not more
than
three, hydrogen bond donors, and not more than 10, or not more than three
hydrogen
bond acceptors. A small molecule myeloid cell agonist is not a protein, a
polysaccharide, or a nucleic acid. In some embodiments, a small molecule is an
30 immune checkpoint inhibitor, such as a PD-1 or PD-Li inhibitor, or a
HER2 inhibitor.
9
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
100331 As used herein, the term "conjugate" refers to a polypeptide attached
to
at least one compound, optionally via a linker(s). In some embodiments, the
polypeptide is an antibody or antigen binding fragment thereof.
100341 As used herein, an "immune-stimulatory conjugate" refers to a conjugate
5 that activates or stimulates the immune system or a portion thereof, as
determined by an
in vitro or in vivo assay.
100351 As used herein, an "immune cell" refers to a T cell, B cell, MC cell,
NKT cell, or an antigen presenting cell. In some embodiments, an immune cell
is a T
cell, B cell, NK cell, or NKT cell. In some embodiments, an immune cell is an
antigen
10 presenting cell. In some embodiments, an immune cell is not an antigen
presenting cell.
100361 The terms "salt" or "pharmaceutically acceptable salt" refer to salts
derived from a variety of organic and inorganic counter ions well known in the
art.
Pharmaceutically acceptable acid addition salts can be formed with inorganic
acids and
organic acids. Inorganic acids from which salts can be derived include, for
example,
15 hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid,
phosphoric acid, and the
like. Organic acids from which salts can be derived include, for example,
acetic acid,
propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic
acid,
succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid,
cinnamic acid,
mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic
acid,
20 salicylic acid, and the like. Pharmaceutically acceptable base addition
salts can be
formed with inorganic and organic bases. Inorganic bases from which salts can
be
derived include, for example, sodium, potassium, lithium, ammonium, calcium,
magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic
bases
from which salts can be derived include, for example, primary, secondary, and
tertiary
25 amines, substituted amines including naturally occurring substituted
amines, cyclic
amines, basic ion exchange resins, and the like, specifically such as
isopropylamine,
trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
In
some embodiments, the pharmaceutically acceptable base addition salt is chosen
from
ammonium, potassium, sodium, calcium, and magnesium salts.
30 100371 The term "Cx-y" when used in conjunction with a chemical
moiety, such
as alkyl, alkenyl, or alkynyl is meant to include groups that contain from x
to y carbons
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
in the chain. For example, the term "CL-6 alkyl" refers to substituted or
unsubstituted
saturated hydrocarbon groups, including straight-chain alkyl and branched-
chain alkyl
groups that contain from 1 to 6 carbons. The term ¨Cx-y alkylene- refers to a
substituted
or unsubstituted alkylene chain with from x to y carbons in the alkylene
chain. For
5 example ¨Ci-6 alkylene- may be selected from methylene, ethylene,
propylene,
butylene, pentylene, and hexylene, any one of which is optionally substituted.

100381 The terms "Cx-y alkenyl" and "Cx-y alkynyl" refer to substituted or
unsubstituted unsaturated aliphatic groups analogous in length and possible
substitution
to the alkyls described above, but that contain at least one double or triple
bond,
10 respectively. The term ¨Cx-y alkenylene- refers to a substituted or
unsubstituted
alkenylene chain with from x to y carbons in the alkenylene chain. For
example, ¨C24
alkenylene- may be selected from ethenylene, propenylene, butenylene,
pentenylene,
and hexenylene, any one of which is optionally substituted. An alkenylene
chain may
have one double bond or more than one double bond in the alkenylene chain. The
term
15 ¨C1-y alkynylene- refers to a substituted or unsubstituted alkynylene
chain with from x
to y carbons in the alkenylene chain. For example, ¨C2-6 alkenylene- may be
selected
from ethynylene, propynylene, butynylene, pentynylene, and hexynylene, any one
of
which is optionally substituted. An alkynylene chain may have one triple bond
or more
than one triple bond in the alkynylene chain.
20 100391 "Alkylene" refers to a divalent hydrocarbon chain linking
the rest of the
molecule to a radical group, consisting solely of carbon and hydrogen,
containing no
unsaturation, and preferably having from one to twelve carbon atoms, for
example,
methylene, ethylene, propylene, butylene, and the like. The alkylene chain is
attached
to the rest of the molecule through a single bond and to the radical group
through a
25 single bond. The points of attachment of the alkylene chain to the rest
of the molecule
and to the radical group are through the terminal carbons respectively. In
other
embodiments, an alkylene comprises one to five carbon atoms (i.e., Ci-Cs
alkylene). In
other embodiments, an alkylene comprises one to four carbon atoms (i.e., C t-
C4
alkylene). In other embodiments, an alkylene comprises one to three carbon
atoms (i.e.,
30 Ci-C3 alkylene). In other embodiments, an alkylene comprises one to two
carbon atoms
(i.e., C1-C2 alkylene). In other embodiments, an alkylene comprises one carbon
atom
11
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(i.e., Ci alkylene). In other embodiments, an alkylene comprises five to eight
carbon
atoms (1.e., C5-C8 alkylene). In other embodiments, an alkylene comprises two
to five
carbon atoms C2-05 alkylene). In other
embodiments, an alkylene comprises three
to five carbon atoms (La, C3-CS alkylene). Unless stated otherwise
specifically in the
5 specification, an alkylene chain is optionally substituted by one or more
substituents
such as those substituents described herein.
100401 "Alkenylene" refers to a divalent hydrocarbon chain linking the rest of

the molecule to a radical group, consisting solely of carbon and hydrogen,
containing at
least one carbon-carbon double bond, and preferably having from two to twelve
carbon
10 atoms. The alkenylene chain is attached to the rest of the molecule
through a single
bond and to the radical group through a single bond. The points of attachment
of the
alkenylene chain to the rest of the molecule and to the radical group are
through the
terminal carbons respectively. In other embodiments, an alkenylene comprises
two to
five carbon atoms (La, C2-05 alkenylene). In other embodiments, an alkenylene
15 comprises two to four carbon atoms (i.e., C2-C4 alkenylene). In other
embodiments, an
alkenylene comprises two to three carbon atoms (La, C2-C3 alkenylene). In
other
embodiments, an alkenylene comprises two carbon atoms (La, C2 alkenylene). In
other
embodiments, an alkenylene comprises five to eight carbon atoms (i.e., C5-Cs
alkenylene). In other embodiments, an alkenylene comprises three to five
carbon atoms
20 (i.e., C3-05 alkenylene). Unless stated otherwise specifically in the
specification, an
alkenylene chain is optionally substituted by one or more substituents such as
those
substituents described herein.
[0041] "Alkynylene" refers to a divalent hydrocarbon chain linking the rest of

the molecule to a radical group, consisting solely of carbon and hydrogen,
containing at
25 least one carbon-carbon triple bond, and preferably having from two to
twelve carbon
atoms. The alkynylene chain is attached to the rest of the molecule through a
single
bond and to the radical group through a single bond. The points of attachment
of the
alkynylene chain to the rest of the molecule and to the radical group are
through the
terminal carbons respectively. In other embodiments, an alkynylene comprises
two to
30 five carbon atoms (La, C2-Cs alkynylene). In other embodiments, an
alkynylene
comprises two to four carbon atoms (La, C2-C4 alkynylene). In other
embodiments, an
12
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
alkynylene comprises two to three carbon atoms (La, C2-C3 alkynylene). In
other
embodiments, an alkynylene comprises two carbon atoms (La, C2 alkynylene). In
other
embodiments, an alkynylene comprises five to eight carbon atoms (i.e., Cs-Cs
alkynylene). In other embodiments, an alkynylene comprises three to five
carbon atoms
5 (i.e., C3-05 alkynylene). Unless stated otherwise specifically in the
specification, an
alkynylene chain is optionally substituted by one or more substituents such as
those
substituents described herein.
[0042] "Heteroalkylene" refers to a divalent hydrocarbon chain including at
least one heteroatom in the chain, containing no unsaturation, and preferably
having
10 from one to twelve carbon atoms and from one to 6 heteroatoms, e.g., -0-
, -NH-, -S-.
The heteroalkylene chain is attached to the rest of the molecule through a
single bond
and to the radical group through a single bond. The points of attachment of
the
heteroalkylene chain to the rest of the molecule and to the radical group are
through the
terminal atoms of the chain. In other embodiments, a heteroalkylene comprises
one to
15 five carbon atoms and from one to three heteroatoms. In other
embodiments, a
heteroalkylene comprises one to four carbon atoms and from one to three
heteroatoms
In other embodiments, a heteroalkylene comprises one to three carbon atoms and
from
one to two heteroatoms. In other embodiments, a heteroalkylene comprises one
to two
carbon atoms and from one to two heteroatoms. In other embodiments, a
20 heteroalkylene comprises one carbon atom and from one to two
heteroatoms. In other
embodiments, a heteroalkylene comprises five to eight carbon atoms and from
one to
four heteroatoms. In other embodiments, a heteroalkylene comprises two to five
carbon
atoms and from one to three heteroatoms. In other embodiments, a
heteroalkylene
comprises three to five carbon atoms and from one to three heteroatoms. Unless
stated
25 otherwise specifically in the specification, a heteroalkylene chain is
optionally
substituted by one or more substituents such as those substituents described
herein.
[0043] The term "carbocycle" as used herein refers to a saturated, unsaturated
or
aromatic ring in which each atom of the ring is carbon. Carbocycle includes 3-
to 10-
membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-
membered
30 bridged rings. Each ring of a bicyclic carbocycle may be selected from
saturated,
unsaturated, and aromatic rings. In an exemplary embodiment, an aromatic ring,
e.g.,
13
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane,
cyclopentane, or cyclohexene. A bicyclic carbocycle includes any combination
of
saturated, unsaturated and aromatic bicyclic rings, as valence permits. A
bicyclic
carbocycle includes any combination of ring sizes such as 4-5 fused ring
systems, 5-5
5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7
fused ring
systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring
systems.
Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl,
adamantyl,
phenyl, indanyl, and naphthyl. The term "unsaturated carbocycle" refers to
carbocycles
with at least one degree of unsaturation and excluding aromatic carbocycles.
Examples
10 of unsaturated carbocycles include cyclohexadiene, cyclohexene, and
cyclopentene.
100441 The term "heterocycle" as used herein refers to a saturated,
unsaturated
or aromatic ring comprising one or more heteroatoms. Exemplary heteroatoms
include
N, 0, Si, P, B, and S atoms. Heterocycles include 3- to 10-membered monocyclic

rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings.
A
15 bicyclic heterocycle includes any combination of saturated, unsaturated
and aromatic
bicyclic rings, as valence permits. In an exemplary embodiment, an aromatic
ring, e.g.,
pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane,
cyclopentane, morpholine, piperidine or cyclohexene. A bicyclic heterocycle
includes
any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring
systems, 5-
20 6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-
7 fused ring
systems, 5-8 fused ring systems, and 6-8 fused ring systems. The term
"unsaturated
heterocycle" refers to heterocycles with at least one degree of unsaturation
and
excluding aromatic heterocycles. Examples of unsaturated heterocycles include
dihydropyrrole, dihydrofuran, oxazoline, pyrazoline, and dihydropyridine.
25 100451 The term "heteroaryl" includes aromatic single ring
structures,
preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings,
whose ring
structures include at least one heteroatom, preferably one to four
heteroatoms, more
preferably one or two heteroatoms. The term "heteroaryl" also includes
polycyclic ring
systems having two or more rings in which two or more carbons are common to
two
30 adjoining rings wherein at least one of the rings is heteroaromatic,
e.g., the other rings
can be aromatic or non-aromatic carbocyclic, or heterocyclic. Heteroaryl
groups
14
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
include, for example, pyrrole, fman, thiophene, imidazole, oxazole, thiazole,
pyrazole,
pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
[0046] The term "substituted" refers to moieties having substituents replacing
a
hydrogen on one or more carbons or substitutable heteroatoms, e.g., -NH-, of
the
5 structure. It will be understood that "substitution" or "substituted
with" includes the
implicit proviso that such substitution is in accordance with permitted
valence of the
substituted atom and the substituent, and that the substitution results in a
stable
compound, i.e., a compound which does not spontaneously undergo transformation

such as by rearrangement, cyclization, elimination, etc. In certain
embodiments,
10 substituted refers to moieties having substituents replacing two
hydrogen atoms on the
same carbon atom, such as substituting the two hydrogen atoms on a single
carbon with
an oxo, imino or thioxo group. As used herein, the term "substituted" is
contemplated
to include all permissible substituents of organic compounds. In a broad
aspect, the
permissible substituents include acyclic and cyclic, branched and unbranched,
15 carbocyclic and heterocyclic, aromatic and non-aromatic substituents of
organic
compounds. The permissible substituents can be one or more and the same or
different
for appropriate organic compounds. For purposes of this disclosure, the
heteroatoms
such as nitrogen may have hydrogen substituents and/or any permissible
substituents of
organic compounds described herein which satisfy the valences of the
heteroatoms.
20 [0047] In some embodiments, substituents may include any
substituents
described herein, for example: halogen, hydroxy, oxo (=0), thioxo (=S), cyano
(-CN),
nitro (-NO2), imino (=N-H), oximo (=N-OH), hydrazino (=N-NH2), -Rb-011.a, -
11.11-
0C(0)-1e, -RP-OC(0)-0W, -1e-OC(0)-N(Ra)2, -PP-N(Ril)2, -le-C(0)1e, 4e-
C(0)01e, -Rb-C(0)N(R12, -Rb-O-Itc-C(0)N(Ra)2., -Rb-N(Ra)C(0)0Ra, -R1-
25 N(Ra)C(0)Ra, -Rb-N(Ra)S(0)tRa (where t is 1 or 2), -ltb-S(0),Ra (where t
is 1 or 2), -Rb-
S(0),011a (where t is 1 or 2), and -R1'-S(0)iN(Ita)2 (where t is 1 or 2); and
alkyl, alkenyl,
alkynyl, aryl, aralkyl, aralkenyl, aralkynyl, cycloalkyl, cycloalkylalkyl,
heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, and heteroarylalkyl any
of which
may be optionally substituted by alkyl, alkenyl, alkynyl, halogen, haloalky1,
30 haloalkenyl, haloalkynyl, oxo (=0), thioxo (=S), cyano (-CN), nitro (-
NO2), imino (=N-
H), oximo (=N-OH), hydrazine (=N-NH2), -Rb-011.a, -Rb-OC(0)-1e, -Rb-OC(0)-
01ta,
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
-11.6-0C(0)-N(R12, -170-N(R12, -Rb-C(0)W, -Rb-C(0)0170, -Rb-C(0)N(1112, -Rb-O-
Itc-
C(0)N(Ra)2, -Rb-N(RIC(0)01e, -Rb-N(RIC(0)re, -Rb-NatiS(0)tRa (where t is 1 or
2), -R1'-S(0)rita (where t is 1 or 2), -Rb-S(0)rOW (where t is 1 or 2) and -Rb-
S(0)tN(W)2
(where t is 1 or 2); wherein each 11.3 is independently selected from
hydrogen, alkyl,
cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl,
heteroaryl, or heteroarylalkyl, wherein each W, valence permitting, may be
optionally
substituted with alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl,
haloalkynyl,
oxo (0), thioxo (=S), cyano (-CN), nitro (-NO2), imino (=N-H), oximo (=N-011),

hydrazine (=N-NH2), -Rb-OW, -Rb-OC(0)-W, -Rb-OC(0)-011.a, -Rb-OC(0)-N(W)2,
-Rb-N(Ra)2, -Rb-C(0)Ra, -10-C(0)0Ra, -Rb-C(0)N(Ra)2, -Rb-O-Re-C(0)N(Ra)2, -Rb-
N(W)C(0)0Ra, -Rb-N(W)C(0)10, -le-N(10)8(0)tR' (where t is 1 or 2), -Rb-S(0)rR3

(where t is 1 or 2), -Rb-S(0)tOR3 (where t is 1 or 2) and RP-S(0)tN(Ra)2
(where t is 1 or
2); and wherein each Itb is independently selected from a direct bond or a
straight or
branched alkylene, alkenylene, or alkynylene chain, and each RC is a straight
or
branched alkylene, alkenylene or alkynylene chain.
100481 It will be understood by those skilled in the art that substituents can

themselves be substituted, if appropriate. Unless specifically stated as
"unsubstituted,"
references to chemical moieties herein are understood to include substituted
variants.
For example, reference to a "heteroaryl" group or moiety implicitly includes
both
substituted and unsubstituted variants.
[0049] In addition, it should be understood that the individual compounds
(e.g.,
proteins), or groups of compounds, derived from the various combinations of
the
structures and substituents (e.g., domains, regions or peptide components)
described
herein, are disclosed by the present application to the same extent as if each
compound
or group of compounds was set forth individually. Thus, selection of
particular
structures or particular substituents is within the scope of the present
disclosure.
100501 Chemical entities having carbon-carbon double bonds or carbon-nitrogen
double bonds may exist in Z- or E- form (or cis- or trans- form). Furthermore,
some
chemical entities may exist in various tautomeric forms. Unless otherwise
specified,
chemical entities described herein are intended to include all Z-, E- and
tautomeric
forms as well.
16
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0051] A "tautomer" refers to a molecule wherein a proton shift from one atom
of a molecule to another atom of the same molecule is possible. The compounds
presented herein, in certain embodiments, exist as tautomers. In circumstances
where
tautomerization is possible, a chemical equilibrium of the tautomers will
exist. The
5 exact ratio of the tautomers depends on several factors, including
physical state,
temperature, solvent, and pH. Some examples of tautomeric equilibrium include.
v ar1; \AAA 9
V1/4NA
H H
µ3,
N H2
H
\ NH 2 \ NH \NA H
\CA-NA
H
cr's tie
--- :ra
N I
N,N
HN¨Nõ1\1---1 NH
N
,sst N '1/41 s e`r4 t e
E
OH
0
[0052] The phrases "intravenous administration" and "administered
intravenously" as used herein refer to injection or infusion of a conjugate
into a vein of
a subject.
[0053] The phrases "intravenous slow infusion" and "IV slow infusion" as used
here refer to an intravenous infusion that results in a Tmax of about 4 hours
or more.
[0054] The phrases "subcutaneous administration," "subcutaneously
administering" and the like refer to administration of a conjugate into the
subcutis of a
15 subject. For clarity, a subcutaneous administration is distinct from an
intratumoral
injection into a tumor or cancerous lesion located in the subcuta.
[0055] The phrase "pharmaceutically acceptable" is employed herein to refer to

those compounds, materials, compositions, and/or dosage forms which are,
within the
scope of sound medical judgment, suitable for use in contact with the tissues
of human
17
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
beings and animals without excessive toxicity, irritation, allergic response,
or other
problem or complication, commensurate with a reasonable benefit/risk ratio.
[0056] As used herein, "identical" or "identity" refer to the similarity
between a
DNA, RNA, nucleotide, amino acid, or protein sequence to another DNA, RNA,
5 nucleotide, amino acid, or protein sequence. Identity can be expressed in
terms of a
percentage of sequence identity of a first sequence to a second sequence.
Percent (%)
sequence identity with respect to a reference DNA sequence can be the
percentage of
DNA nucleotides in a candidate sequence that are identical with the DNA
nucleotides
in the reference DNA sequence after aligning the sequences. "Percent (%)
sequence
10 identity with respect to a reference amino acid sequence can be the the
percentage of
amino acid residues in a candidate sequence that are identical with the amino
acid
residues in the reference polypeptide sequence after aligning the sequences
and
introducing gaps, if necessary, to achieve the maximum percent sequence
identity, and
not considering any conservative substitutions as part of the sequence
identity. The
15 percentage sequence identity values can be generated using the NCBI
BLAST 2.0
software as defined by Altschul et a/. (1997) "Gapped BLAST and PSI-BLAST. a
new
generation of protein database search programs," Nucleic Acids Res. 25:3389-
3402,
with the parameters set to default values.
[0057] The term "about" as used herein in the context of a number refers to a
20 range centered on that number and spanning 10% less than that number and
10% more
than that number. The term "about" used in the context of a range refers to an
extended
range spanning 10% less than that the lowest number listed in the range and
10% more
than the greatest number listed in the range.
[0058] In the present description, any concentration range, percentage range,
25 ratio range, or integer range is to be understood to include the value
of any integer
within the recited range and, when appropriate, fractions thereof (such as one
tenth and
one hundredth of an integer), unless otherwise indicated. Also, any number
range
recited herein relating to any physical feature, such as polymer subunits,
size or
thickness, are to be understood to include any integer within the recited
range, unless
30 otherwise indicated. It should be understood that the terms "a" and "an"
as used herein
refer to "one or more" of the enumerated components. The use of the
alternative (e.g.,
18
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
"of') should be understood to mean either one, both, or any combination
thereof of the
alternatives. As used herein, the terms "include" and "comprise" are used
synonymously.
100591 The phrase "at least one of" when followed by a list of items or
5 elements refers to an open ended set of one or more of the elements in
the list, which
may but does not necessarily include more than one of the elements.
Exemplary Antibodies
[0060] In some embodiments, a conjugate as described herein comprises an
antibody. In some such embodiments, the antibody comprises one or more antigen
10 binding domains and an Fc domain, wherein each antigen binding domain
specifically
binds to an antigen. An antibody can have, for example, a first antigen
binding domain
that specifically binds to a first antigen, a second antigen binding domain
that
specifically binds to a second antigen, and an Fc domain. In various
embodiments, an
antibody can include two antigen binding domains, in which each antigen
binding
15 domain recognizes the same epitope on the antigen. An antibody can
include two
antigen binding domains in which each antigen binding domain recognizes a
different
epitope of the same antigen. An antibody can include two antigen binding
domains in
which each antigen binding domain recognizes different antigens. In various
embodiments, an antibody has one antigen binding domain. In various
embodiments,
20 an antigen binding domain may comprise, for example, a heavy chain
variable domain
(VH) and a light chain variable domain (VL), or in the case of a heavy-chain
only
antibody, a VHH
[0061] Nonlimiting exemplary tumor antigens that may be bound by a
polypeptide, such as an antibody, include CD5, CD25, CD37, CD33, CD45, BCMA,
25 CS-1, PD-L1, 87-H3, B7-DC (PD-L2), HLD-DR, carcinoembryonic antigen
(CEA),
TAG-72, EpCAM, MUD, folate-binding protein (FOLR1), A33, G250 (carbonic
anhydrase IX), prostate-specific membrane antigen (PSMA), G02, GD3, GM2, Ley,
CA-125, CA19-9 (MUC1 sLe(a)), epidermal growth factor, HER2, IL-2 receptor,
EGFRvIll (de2-7 EGFR), fibroblast activation protein (FAP), a tenascin, a
30 metalloproteinase, endosialin, avB3, LMP2, EphA2, PAP, AFP, ALK,
polysialic acid,
19
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
TRP-2, fucosyl GM1, mesothelin (MSLN), PSCA, sLe(a), GM3, BORIS, Tn, TF,
GloboH, STn, CSPG4, AKAP-4, SSX2, Legumain, Tie 2, Tim 3, VEGFR2, PDGFR-B,
ROR2, TRA1L1, MUC16, EGFR, CMET, HER3, MUC1, MUC15, CA6, NAPI2B,
CLDN182, RON, LY6E, FRAlpha, DLL3, PTK7, LIV1, ROR1, CLDN6, GPC3,
5 ADAM12, LRRC15, CDH6, TMEFF2, TMEM238, GPNMB, ALPPL2, UPK1B,
UPK2, LAMP-1, LY6K, EphB2, STEAP, ENPP3, CDH3, Nectin4, LYPD3, EFNA4,
GPA33, SLITRK6, and HAVCRL
[0062] In certain embodiments, the tumor antigen is selected from HER2,
Nectin4, MSLN, LIV-1, MUC16, CEACAM1, CEACAM3, CEACAM4, CEACAM5,
CEACAM6, CEACAM7, CEACAM8, CEACAM16, CEACAM18, CEACAM19,
CEACAM20, CEACAM21, VEGFR1, VEGFR2, MUC1, PSMA, PSA, M1JC-2, and
LARC15
[0063] In certain embodiments, a polypeptide, such as an antibody,
specifically
binds to a non-proteinaceous or glycoantigen, such as GD2, GD3, GM2, Ley,
polysialic
15 acid, fucosyl GM1, GM3, Tn, STn, sLe(animal), or GloboH.
[0064] In certain embodiments, a polypeptide, such as an antibody,
specifically
binds to a solid tumor antigen. In some embodiments, the solid tumor antigen
is
preferentially present on sarcoma or carcinoma cell(s). In some embodiments,
the solid
tumor antigen is preferentially present on a sarcoma cell(s). In some
embodiments, the
20 solid tumor antigen is preferentially present on a carcinoma cell(s).
[0065] In some embodiments, the solid tumor antigen is present on cells of a
brain, breast, lung, liver, kidney, pancreatic, colorectal, ovarian, head and
neck, bone,
skin, mesothelioma, bladder, stomach, prostate, thyroid, uterine or
cervical/endometrial
cancer.
25
[0066] In some embodiments, the solid tumor antigen
is an antigen present on
breast cancer, such as HER2, LIV-1, CDH3 (p-cadherin), MUC1, Sialo-epitope
CA6,
PTX.7, GPNMB, LAMP-1, LRRC15, ADAM12, EPHA2, TNC, LYPD3, EFNA4,
Nectin-4, and CLDN6.
[0067] In some embodiments, the solid tumor antigen is an antigen present on
30 brain cancer, such as EGFRAII, TNC and DLL-3.
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0068] In some embodiments, the solid tumor antigen is an antigen present on
lung cancer, such as mesothelin, HER2, EGFR, PD-L1, MSLN, LY6K, CD56, PTK7,
FOLR1, DLL3, SLC34A2, CECAM5, MUC16, LRRC15, ADAM12, EGFRAII,
LYPD3, EFNA4, Nectin-4, and MUCl.
5
[0069] In some embodiments, the solid tumor antigen
is an antigen present on
liver cancer, such as GPC3, EPCAM, and CECAM5.
[0070] In some embodiments, the solid tumor antigen is an antigen present on
kidney cancer, such as HAVCR1, ENPP3, CDH6, CD70, and cMET.
100711 In some embodiments, the solid tumor antigen is an antigen present on
10 pancreatic cancer, such as PTK7, MUC16, MSLN, LRRC15, ADAM12, EFNA4,
MUC5A, Nectin-4, and MUCl.
[0072] In some embodiments, the solid tumor antigen is an antigen present on
colorectal cancer, such as EPHB2, TMEM238, CECAM5, LRRC15, Nectin4,
ADAM12, EFNA4 and GPA33.
15
[0073] In some embodiments, the solid tumor antigen
is an antigen present on
ovarian cancer, such as M1JC16, MUC1, MSLN, FOLR1, sTN, VTCN1, HER2, PTK7,
FAP, TMEM238, LRRC15, CLDN6, SLC34A2 and EFNA4.
[0074] In some embodiments, the solid tumor antigen is an antigen present on
head and neck cancer, such as LY6K, PTK7, LRRC15, ADAM12, LYPD3, Nectin4,
20 EFNA4 and TNC.
[0075] In some embodiments, the solid tumor antigen is an antigen present on
bone cancer, such as EPHA2, LRRC15, ADAM12, GPNMB, TP-3 and CD248.
[0076] In some embodiments, the solid tumor antigen is an antigen present on
mesothelioma, such as MSLN.
25
[0077] In some embodiments, the solid tumor antigen
is an antigen present on
bladder cancer, such as LY6K, PTK7, UPK1B, UPK2, TNC, Nectin4, SLITRK6,
LYPD3, EFNA4 and HER2.
[0078] In some embodiments, the solid tumor antigen is an antigen present on
stomach/gastric cancer, such as HER2, EPHB2, TMEM238, CECAM5 and EFNA4.
21
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0079] In some embodiments, the solid tumor antigen is an antigen present on
prostate cancer, such as PSMA, FOLH1, PTK7, STEAP, T1M-EFF2 (TENB2), 0R51E2,
SLC30A4 and EFNA4.
100801 In some embodiments, the solid tumor antigen is an antigen present on
5 thyroid cancer, such as PTK7.
[0081] In some embodiments, the solid tumor antigen is an antigen present on
uterine cancer, such as present on uterine cancer such as LY6K, PTK7, EPHB2,
FOLR1, ALPPL2, M1JC16, Nectin-4, and EF1sTA4.
[0082] In some embodiments, the solid tumor antigen is an antigen present on
10 cervical/endometrial cancer, such as LY6K, PTK7, MUC16, LYPD3, EFNA4,
Nectin-
4, and MUCl.
[0083] In some embodiments, the solid tumor antigen is an antigen present on a

sarcoma, such as LRRC15.
[0084] In some embodiments, the tumor antigen is HER2. In some aspects, the
15 HER2 antigen is expressed for example, on a lung, colorectal, ovarian,
bladder,
stomach/gastric, or breast cancer cell.
[0085] In some aspects, the antigen is a liver cell antigen. In some aspects,
the
liver cell antigen is expressed on a canalicular cell, Kupffer cell,
hepatocyte, or any
combination thereof. In some aspects, the liver cell antigen is a hepatocyte
antigen. In
20 some aspects, the liver cell antigen is selected from the group
consisting of ASGR1
(asialoglycoprotein receptor 1), ASGR2 (asialoglycoprotein receptor 2), TRF2,
UGT1A1, SLC22A7, SLC13A5, SLC22A1, and C9. In some aspects, the liver cell
antigen is selected from the group consisting of ASGR1, ASGR2, and TRF2. In
some
aspects, the liver cell antigen is expressed on a liver cell infected with a
virus selected
25 from the group consisting of HBV and HCV.
[0086] In some aspects, the antigen is a viral antigen from a virus selected
from
the group consisting of HBV and HCV. In some aspects, the viral antigen is an
HBV
antigen. In some aspects, the viral antigen is HBsAg, HBcAg, or HBeAg. In some

aspects, the viral antigen is HBsAg.
30 100871 In some embodiments, an antibody comprises an antigen
binding domain
and an Fc domain. In some embodiments, an antibody comprises two light chain
22
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
polypeptides (light chains) and two heavy chain polypeptides (heavy chains),
held
together covalently by disulfide linkages. The heavy chain typically comprises
a heavy
chain variable region (VH) and a heavy chain constant region. The heavy chain
constant region comprises three domains, CHI, CH2, and CH3. An Fc domain
5 typically comprises heavy chain CH2 and CH3 domains. The light chain
typically
comprises a light chain variable region (VL) and a light chain constant
region_ The
antigen-recognition regions of the antibody variable domains typically
comprise six
complementarity determining regions (CDRs), or hypervariable regions, that lie
within
the framework of the heavy chain variable region and light chain variable
region at the
10 N-terminal ends of the two heavy and two light chains. The constant
domains provide
the general framework of the antibody and may not be involved directly in
binding the
antibody to an antigen, but can be involved in various effector functions,
such as
participation of the antibody in antibody-dependent cellular cytotoxicity
(ADCC).
100881 An antibody can be any class, e.g., IgA, IgD, IgE, IgG, and Ig.M.
15 Certain classes can be further divided into isotypes, e.g., IgGl, IgG2,
IgG3, IgG4, IgAl ,
and IgA2. The heavy-chain constant regions that correspond to the different
classes of
immunoglobulins can be a, 6, e, y, and p., respectively. The light chains can
be either
kappa (or K) or lambda (or X).
100891 In some embodiments an antigen binding domain comprises a light chain
20 complementary determining region 1 (LCDRI), a light chain complementary
determining region 2 (LCDR2), a light chain complementary determining region 3

(LCDR3), a heavy chain complementary determining region 1 (HCDR1), a heavy
chain
complementary determining region 2 (HCDR2), and a heavy chain complementary
determining region 3 (HCDR3). In some embodiments, an antibody may be a heavy-
25 chain only antibody, in which case the antigen binding domain comprises
HCDRI,
HCDR2, and HCDR3, and the antibody lacks a light chain. Unless stated
otherwise, the
CDRs described herein can be defined according to the 'MGT (the international
ImMunoGeneTics information) system.
100901 An antibody can be chimeric or humanized. Chimeric and humanized
30 forms of non-human (e.g., murine) antibodies can be intact (full length)
chimeric
immunoglobulins, immunoglobulin chains or antigen binding fragments thereof
(such
23
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
as Fv, Fab, Fab', F(abl2 or other target-binding subdomains of antibodies),
which can
contain sequences derived from non-human immunoglobulin. In general, the
humanized antibody can comprise substantially all of at least one, and
typically two,
variable domains, in which all or substantially all of the CDR regions
correspond to
5 those of a non-human immunoglobulin and all or substantially all of the
framework
(FR) regions are those of a human immunoglobulin sequence. A humanized
antibody
can also comprise at least a portion of an immunoglobulin constant region
(Fc), an Fc
domain, typically that of a human immunoglobulin sequence.
[0091] An antibody described herein can be a human antibody. As used herein,
10 "human antibodies" can include antibodies having, for example, the amino
acid
sequence of a human immunoglobulin and include antibodies isolated from human
immunoglobulin libraries or from animals transgenic for one or more human
immunoglobulins and that typically do not express endogenous immunoglobulins.
Human antibodies can be produced using transgenic mice which are incapable of
15 expressing functional endogenous immunoglobulins, but which can express
human
immunoglobulin genes. Completely human antibodies that recognize a selected
epitope
can be generated using guided selection. In this approach, a selected non-
human
monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of
a
completely human antibody recognizing the same epitope
20 100921 An antibody described herein can be a bispecific antibody
or a dual
variable domain antibody (DVD). Bispecific and DVD antibodies are monoclonal,
often human or humanized, antibodies that have binding specificities for at
least two
different antigens.
[0093] An antibody described herein can be derivatized or otherwise modified.
25 For example, derivatized antibodies can be modified by glycosylation,
acetylation,
pegylation, phosphorylation, amidation, derivatization by known
protecting/blocking
groups, proteolytic cleavage, or the like.
[0094] An antibody described herein can specifically bind to a cancer antigen.

An antibody can specifically bind to a solid tumor antigen.
30 100951 In some embodiments, the antibody may comprise the CDRs
(such as
LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to thellVIGT
24
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
system), antigen binding fragment (e.g., the variable regions), or the entire
heavy and
light chains of an antibody selected from trastuzumab, cetuximab, panitumumab,

ofatumumab, belimumab, ipilimumab, pertuzumab, tremelimumab, nivolumab,
pembrolizumab, atezolizumab, MDX-1105 (PCT Publication No. WO 2007/005874),
5 dacetuzumab, urelumab, MPDL3280A, lambroliz-umab, blinatumomab,
nimotuzumab,
zalutumumab, onartuzumab, patritumab, clivatuzumab, sofituzumab, edrecolomab,
adecatumumab, anetumab, huDS6, lifastuzumab, PRI A3, humanized PRI A3,
humanized Ab2-3, claudiximab, AMG595, ABT806, sibrotuzumab, DS-8895a variant
1, DS-8895a variant 2, MEDI-547, namatumab, RG7841, farletuzumab,
mirvetuximab,
10 J591 variant 1, J591 variant 2, rovalpituzumab, PF-06647020,
ladiratuzumab,
cirmtuzumab, ladiratuzumab, huLiv1-14 (PCT Publication No. WO 2012/078688),
Liv1-1.7A4 (US Patent Publication No. 2011/0117013), huLiv1-22 (PCT
Publication
No. WO 2012078688), 4H11 (US Patent Publication No. 2013/0171152), 4115 (US
Patent Publication No. 2013/0171152), glembatumumab, oportuzumab, enfortumab,
15 depatuxizumab, the antibody of ASG-15ME, huM25 (PCT Publication No.
W02017/095808A1), and codrituzumab.
100961 In some embodiments, the antibody may comprise the CDRs (such as
LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to the [MGT
system), the antigen binding fragment (e.g., variable regions), or the entire
heavy and
20 light chains of an antibody selected from pertuzumab, trastuzumab, and
ladiratuzumab,
or biosimilar thereof.
100971 In some such embodiments, the anti-HER2 antibody of the conjugate
comprises heavy chain (LC)-CDR1, HC-CDR2, HC-CDR3, light chain (LC)-CDR1,
LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6, respectively. In further embodiments,
25 the anti-HER2 antibody of the conjugate comprises a heavy chain and
light chain,
wherein: (a) the heavy chain comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ
ID NOs: 1-3, respectively, and comprises a heavy chain variable region (VH)
having an
amino acid sequence that has at least 90%, at least 91%, at least 92%, at
least 93%, at
least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least
99%, at least
30 99.5%, or 100% identity to the VH amino acid sequence of SEQ ID NO: 7;
and (b) the
light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ED NOs: 4-6,
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
respectively, and a light chain variable region (VI.) having an amino acid
sequence that
has at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at
least 95%, at
least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100%
identity to
the VL amino acid sequence of SEQ ID NO: 8. In still further embodiments, the
anti-
5 HER2 antibody of the conjugate comprises a Vir comprising or consisting
of the amino
acid sequence of SEQ ID NO: 7 and a N/L, comprising or consisting of the amino
acid
sequence of SEQ ID NO: 8. In yet further embodiments, the anti-HER2 antibody
of the
conjugate comprises a heavy chain and light chain, wherein: (a) the heavy
chain
comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ ID NOs: 1-3, respectively,
10 and comprises an amino acid sequence that has at least 90%, at least
91%, at least 92%,
at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least
98%, at least
99%, at least 99.5%, or 100% identity to the heavy chain amino acid sequence
of SEQ
ID NO: 9; and (b) the light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3 of
SEQ ID NOs: 4-6, respectively, and an amino acid sequence that has at least
90%, at
15 least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at
least 96%, at least
97%, at least 98%, at least 99%, at least 99.5%, or 100% identity to the light
chain
amino acid sequence of SEQ ID NO: 10. In more embodiments, the anti-HER2
antibody of the conjugate comprises a heavy chain comprising or consisting of
the
amino acid sequence of SEQ ID NO: 9 and a light chain comprising or consisting
of the
20 amino acid sequence of SEQ ID NO: 10.
100981 In some embodiments, the antibody comprises a heavy chain variable
region comprising the amino acid sequence of SEQ ID NO: 7 and a light chain
variable
region comprising the amino acid sequence of SEQ ID NO: 8; or the antibody
comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and
a
25 light chain comprising the amino acid sequence of SEQ ID NO: 10.
00991 In some embodiments, an antibody specifically binds to a breast cancer
antigen. In some such embodiments, the antibody may comprise the CDRs (such as

LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to the IMGT
system), antigen binding fragment (e.g., the variable regions), or the entire
heavy and
30 light chains of an antibody selected from trastuzumab, pertuzumab,
ladiratuzumab,
huLiv1-14 (PCT Publication No. WO 2012/078688), Liv1-1,7A4 (US Patent
26
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Publication No. 2011/0117013), huLiv1-22 (PCT Publication No. WO 2012078688),
huDS6, glembatumumab, PF-0664720, MEDI-547, DS-8895a variant 1, and DS-
08895a variant 2.
101001 In some embodiments, an antibody specifically binds to an antigen
5 present on brain cancer cells. In some such embodiments, the antibody may
comprise
the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
according to the IMGT system), antigen binding fragment (e.g., the variable
regions), or
the entire heavy and light chains of an antibody selected from 4MG595, A8T806,

rovalpituzumab or depatuxizumab.
10 01011 In some embodiments, an antibody specifically binds to an
antigen
present on lung cancer cells. In some such embodiments, the antibody may
comprise
the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
according to the IMGT system), antigen binding fragment (e.g., the variable
regions), or
the entire heavy and light chains of an antibody selected from panitumumab,
cetuximab,
15 pembrolizumab, nivolumab, atezolizumab, nimotuzumab, lifastuzumab,
anetumab, PF-
0664720, farletuzumab, rovalpituzumab, lifastuzumab, sofituzumab, huDS6,
ABT806,
AMG595, and huM25 (PCT Publication No. W02017/095808A1).
[0102] In some embodiments, an antibody specifically binds to an antigen
present on liver cancer. In some such embodiments, the antibody may comprise
the
20 CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
heavy and light chains of an antibody selected from codrituzumab, oportuzumab,
and
humanized PR1A3.
[0103] In some embodiments, an antibody specifically binds to an antigen
25 present on kidney cancer. In some such embodiments, the antibody may
comprise the
CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
heavy and light chains of an antibody selected from AGS-16M8F, AGS-16C3, the
antibody of CDX-014, and onartuzumab.
30 101041 In some embodiments, an antibody specifically binds to an
antigen
present on pancreatic cancer. In some such embodiments, the antibody may
comprise
27
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
according to the IMGT system), antigen binding fragment (e.g., the variable
regions), or
the entire heavy and light chains of an antibody selected from PF-0664720,
clivatuzumab, 4HI1 (US Patent Publication No. 2013/0171152), 4H5 (US Patent
5 Publication No. 2013/0171152), anetumumab, huDS6, sofituzumab, huM25 (PCT
Publication No. W02017/095808A1), and RG7841.
101051 In some embodiments, an antibody specifically binds to an antigen
present on colorectal cancer. In some such embodiments, the antibody may
comprise
the CDRs (such as LCDRI, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
10 according to the IMGT system), antigen binding fragment (e.g., the
variable regions), or
the entire heavy and light chains of an antibody selected from huM25 (PCT
Publication
No. W02017/095808A1), PR1A3, humanized PR1A3, pantumumab, cetuximab,
nimotuzumab, and zalutumumab.
[0106] In some embodiments, an antibody specifically binds to an antigen
15 present on ovarian cancer. In some such embodiments, the antibody may
comprise the
CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
heavy and light chains of an antibody selected from sofituzumab, 41111 (U.S.
Patent
Publication No. 2013/0171152, 4H5 (U.S. Patent Publication No. 2013/0171152),
20 huDS6, farletuzumab, anetumab, trastuzumab, pertuzumab, PF-0664720,
sibrotuzumab,
huM25 (PCT Publication No. W02017/095808A1), and lifastuzumab.
[0107] In some embodiments, an antibody specifically binds to an antigen
present on head and neck cancer. In some such embodiments, the antibody may
comprise the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
25 according to the IMGT system), antigen binding fragment (e.g., the
variable regions), or
the entire heavy and light chains of an antibody selected from cetuximab,
panitumumab,
nimtuzumab, PF-0664720, pantumumab, cetuximab, nimotuzumab, and zalutumumab.
[0108] In some embodiments, an antibody specifically binds to an antigen
present on bone cancer. In some such embodiments, the antibody may comprise
the
30 CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
28
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
heavy and light chains of an antibody selected from huM25 (PCT Publication No.

W02017/095808A1), DS-8895a variant 1, DS-8895a variant 2, and glembatumab.
[0109] In some embodiments, an antibody specifically binds to an antigen
present on skin cancer.
5 [0110] In some embodiments, an antibody specifically binds to an
antigen
present on mesothelioma.
[0111] In some embodiments, an antibody specifically binds to an antigen
present on cervical/endometrial cancer. In some such embodiments, the antibody
may
comprise the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
10 according to the IMGT system), antigen binding fragment (e.g., the
variable regions), or
the entire heavy and light chains of an antibody selected from PF-0664720,
anetumumab, 4H11(US Patent Publication No. 2013/0171152), 4H5 (US Patent
Publication No. 2013/0171152), huDS6, and sofituzumab.
[0112] In some embodiments, an antibody specifically binds to an antigen
15 present on bladder cancer. In some such embodiments, the antibody may
comprise the
CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
heavy and light chains of an antibody selected from enfortumab, trastuz-umab,
pertuzumab and SLITRK6.
20 [0113] In some embodiments, an antibody specifically binds to an
antigen
present on stomach/gastric cancer. In some such embodiments, the antibody may
comprise the CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3,
according to the [MGT system), antigen binding fragment (e.g., the variable
regions), or
the entire heavy and light chains of an antibody selected from sofituzumab,
anetumab,
25 pertuzumab, trastuzumab, and humanized PR1A3.
[0114] In some embodiments, an antibody specifically binds to an antigen
present on prostate cancer. In some such embodiments, the antibody may
comprise the
CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the [MGT system), antigen binding fragment (e.g., the variable regions), or
the entire
30 heavy and light chains of an antibody selected from mirvetuximab, J591
variant 1, and
J591 variant 2.
29
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0115] In some embodiments, an antibody specifically binds to an antigen
present on thyroid cancer.
[0116] In some embodiments, an antibody specifically binds to an antigen
present on uterine cancer. In some such embodiments, the antibody may comprise
the
5 CDRs (such as LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3, according to
the IMGT system), antigen binding fragment (e.g., the variable regions), or
the entire
heavy and light chains of an antibody selected from PF-0664720, farletuzumab,
sofituzumab, 41111(US Patent Publication No. 2013/0171152, and 4115 (US Patent

Publication No. 2013/0171152).
10 [0117] In some embodiments, an antibody specifically binds to an
antigen
present on a sarcoma,
[0118] In some embodiments, an antibody specifically binds to an antigen
present on a liver cell and the subject has a viral infection (e.g., HBV or
HCV). The
antibody can be, for example, an antibody that binds to ASGR1 or ASGR2.
15 Exemplary Fc domains
[0119] A polypeptide, such as a fusion protein or an antibody, may comprise an

Fc domain. An Fc domain is a structure that can bind to one or more Fe
receptors
(FcRs). In various embodiments, an Fc domain is from an IgG antibody, such as
an
IgGl, IgG2, or IgG4 antibody. An Fc domain typically comprises CH2 and CH3
20 domains of a heavy chain constant region, but may comprise more or less
of the heavy
chain constant region as well.
[0120] An Fc domain can be a domain of an antibody that can bind to an
FcR(s). FcRs are organized into classes (e.g., gamma (y), alpha (a) and
epsilon (H))
based on the class of antibody that the FcR recognizes The FcaR class binds to
IgA
25 and includes several isoforms, Fecal (CD89) and Fca R. The FcyR class
binds to IgG
and includes several isoforms, FcyRI (CD64), Fc-yRIIA (CD32a), FcyRIIB
(CD32b),
FcyRIIIA (CD16a), and FcyRIBB (CD16b). An Fc-yRIIIA (CD16a) can be an FcyRIRA
(CD16a) F158 variant or a V158 variant. FcRs also can be FcRn receptors.
[0121] Each FcyR isoform can differ in binding affinity to the Fc domain of
the
30 IgG antibody. For example, FcyRI can bind to IgG with greater affinity
than FcyRII or
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
FcyRIII. The affinity of a particular FeyR isoform to an IgG can be
controlled, in part,
by a glycan (e.g., oligosaccharide) at position CH2 84.4 of the IgG antibody.
For
example, fucose containing CH2 84.4 glycans can reduce IgG affinity for
FeyRBIA. In
addition, GO glucans can have increased affinity for FcyRIIIA due to the lack
of
5 galactose and terminal GIcNAc moiety.
101221 Binding of an Fc domain to an Felt can enhance an immune response.
FcR-mediated signaling that can result from an Fc domain binding to an FcR and
can
lead to the maturation of immune cells. FcR-mediated signaling that can result
from an
Fc domain binding to an FcR can lead to the maturation of dendritic cells
(DCs). FcR-
10 mediated signaling that can result from an Fc domain binding to an FcR
can lead to
antibody dependent cellular cytotoxicity. FcR-mediated signaling that can
result from
an Fc domain binding to an FcR can lead to more efficient immune cell antigen
uptake
and processing. FcR-mediated signaling that can result from an Fc domain
binding to
an FcR can promote the expansion and activation of T cells. FcR-mediated
signaling
15 that can result from an Fc domain binding to an FcR can promote the
expansion and
activation of CD8-F T cells FcR-mediated signaling that can result from an Fc
domain
binding to an FcR can influence immune cell regulation of T cell responses.
FcR-
mediated signaling that can result from an Fc domain binding to an FcR can
influence
dendritic cell regulation of T cell responses. Felt-mediated signaling that
can result
20 from an Fc domain binding to an FcR can influence functional
polarization of T cells
(e.g., polarization can be toward a THI cell response).
01231 An Fc domain can be modified, such as by a modification of the amino
acid sequence, to alter the recognition of an FcR for the Fc domain. Such
modification(s) may still allow for Felt-mediated signaling, depending on the
25 modification. A modification can be a substitution of an amino acid at a
residue of an
Fc domain for a different amino acid at that residue. A modification can be an
insertion
or deletion of an amino acid at a residue of an Fc domain. A modification can
permit
binding of an FcR to a site on the Fc domain to which the FcR may not
otherwise bind.
A modification can increase binding affinity of an FcR to the Fe domain. A
30 modification can decrease binding affinity of an FcR to the Fc domain.
31
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
101241 An Fe domain can be a variant of a naturally occurring Fc domain (e.g.,

a wild type Fc domain) and can comprise at least one amino acid change as
compared to
the sequence of a wild-type Fc domain. An amino acid change in an Fc domain
can
allow the antibody or conjugate to bind to at least one Fc receptor with
greater affinity
5 compared to a wild-type Fc domain. An amino acid change in an Fc domain
can allow
the antibody or conjugate to bind to at least one Fc receptor with lessor
affinity
compared to a wild-type Fc domain.
101251 In some embodiments, an Fc domain exhibits increased binding affinity
to one or more Fc receptors. In some embodiments, an Fc domain exhibits
increased
10 binding affinity to one or more Fcgamma receptors. In some embodiments,
an Fc
domain exhibits increased binding affinity to FcRn receptors. In some
embodiments, an
Fc domain exhibits increased binding affinity to Fcgamma and FcRn receptors.
In other
embodiments, an Fc domain exhibits the same or substantially similar binding
affinity
to Fcgamma and/or FeRn receptors as compared to a wild-type Fc domain from an
IgG
15 antibody (e.g., IgG1 antibody).
101261 In some embodiments, an Fc domain exhibits decreased binding affinity
to one or more Fc receptors. In some embodiments, an Fe domain exhibits
decreased
binding affinity to one or more Fcgamma receptors. In some embodiments, an Fc
domain exhibits decreased binding affinity to FcRn receptors. In some
embodiments,
20 an Fc domain exhibits decreased binding affinity to Fcgamma and FcRn
receptors. In
some embodiments, an Fc domain is an Fc null domain. In some embodiments, an
Fc
domain exhibits decreased binding affinity to FcRn receptors, but exhibits the
same or
increased binding affinity to one or more Fcgamma receptors as compared to a
wildtype
Fe domain. In some embodiments, an Fc domain exhibits increased binding
affinity to
25 FeRn receptors, but exhibits the same or decreased binding affinity to
one or more
Fcgamma receptors.
01271 An Fc domain may have one or more, two or more, three or more, or
four or more amino acid substitutions that decrease binding of the Fc domain
to an Fc
receptor. In certain embodiments, an Fc domain has decreased binding affinity
for one
30 or more of FeTRI (CD64), FeyRIIA (CD32), FeyRIIIA (CD16a), FeyRuIB
(CD16b), or
any combination thereof. In order to decrease binding affinity of an Fc domain
to an Fc
32
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
receptor, the Fc domain may comprise one or more amino acid substitutions that

reduces the binding affinity of the Fc domain to an Fc receptor. In other
embodiments,
an Fc domain exhibits the same or substantially similar binding affinity to
one or more
of Fc7RI (CD64), FcyRIIA (CD32), FcyRIIIA (CD16a),
(CD16b), or any
5 combination thereof as compared to a wild-type Fc domain from an IgG
antibody (e.g.,
IgG1 antibody). In some embodiments, an Fc domain can comprise a sequence of
an
IgG isoform that has been modified from the wild-type IgG sequence. In some
embodiments, the Fc domain can comprise a sequence of the IgG1 isoform that
has
been modified from the wild-type IgG1 sequence. In some embodiments, the
10 modification comprises substitution of one or more amino acids that
reduce binding
affinity of an IgG Fe domain to all Fey receptors.
[0128] A modification can be substitution of E233, L234 and L235, such as
E233P/L234V/L235A or E233P/L234V/L235A1AG236, according to the EU index of
Kabat. A modification can be a substitution of P238, such as P238A, according
to the
15 EU index of Kabat. A modification can be a substitution of D265, such as
D265A,
according to the EU index of Kabat. A modification can be a substitution of
N297,
such as N297A, according to the EU index of Kabat. A modification can be a
substitution of A327, such as A327Q, according to the EU index of Kabat. A
modification can be a substitution of P329, such as P239A, according to the EU
index
20 of Kabat.
[0129] In some embodiments, an IgG Fc domain comprises at least one amino
acid substitution that reduces its binding affinity to Fc7R1, as compared to a
wild-type
or reference IgG Fc domain. A modification can comprise a substitution at
F241, such
as F241A, according to the EU index of Kabat. A modification can comprise a
25 substitution at F243, such as F243A, according to the EU index of Kabat.
A
modification can comprise a substitution at V264, such as V264A, according to
the EU
index of Kabat. A modification can comprise a substitution at D265, such as
D265A
according to the EU index of Kabat.
101301 In some embodiments, an IgG Fc domain comprises at least one amino
30 acid substitution that increases its binding affinity to FeyR1, as
compared to a wild-type
33
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
or reference IgG Fc domain. A modification can comprise a substitution at A327
and
P329, such as A327Q/P329A, according to the EU index of Kabat.
[0131] In some embodiments, the modification comprises substitution of one or
more amino acids that reduce binding affinity of an Igo Fe domain to FcyRII
and
5 FcyRIIIA receptors. A modification can be a substitution of D270, such as
D270A,
according to the EU index of Kabat. A modification can be a substitution of
Q295,
such as Q295A, according to the EU index of Kabat. A modification can be a
substitution of A327, such as A237S, according to the EU index of Kabat.
[0132] In some embodiments, the modification comprises substitution of one or
10 more amino acids that increases binding affinity of an IgG Fe domain to
FcyRII and
FcyRIIIA receptors. A modification can be a substitution of T256, such as
T256A,
according to the EU index of Kabat. A modification can be a substitution of
K290,
such as K290A, according to the EU index of Kabat.
[0133] In some embodiments, the modification comprises substitution of one or
15 more amino acids that increases binding affinity of an IgG Fe domain to
FicyRII
receptor. A modification can be a substitution of R255, such as R255A,
according to
the EU index of Kabat. A modification can be a substitution of E258, such as
E258A,
according to the EU index of Kabat. A modification can be a substitution of
S267, such
as S267A, according to the EU index of Kabat. A modification can be a
substitution of
20 E272, such as E272A, according to the EU index of Kabat. A modification
can be a
substitution of N276, such as N276A, according to the EU index of Kabat. A
modification can be a substitution of D280, such as D280A, according to the EU
index
of Kabat. A modification can be a substitution of 11285, such as H285A,
according to
the EU index of Kabat. A modification can be a substitution of N286, such as
N286A,
25 according to the EU index of Kabat. A modification can be a substitution
of T307, such
as T307A, according to the EU index of Kabat. A modification can be a
substitution of
L309, such as L309A, according to the EU index of Kabat. A modification can be
a
substitution of N315, such as N3 15A, according to the EU index of Kabat. A
modification can be a substitution of K326, such as K326A, according to the EU
index
30 of Kabat. A modification can be a substitution of P331, such as P33 IA,
according to
the EU index of Kabat. A modification can be a substitution of S337, such as
S337A,
34
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
according to the EU index of Kabat. A modification can be a substitution of
A378,
such as A378A, according to the EU index of Kabat. A modification can be a
substitution of E430, such as E430, according to the EU index of Kabat.
101341 In some embodiments, the modification comprises substitution of one or
5 more amino acids that increases binding affinity of an IgG Fc domain to
FicyRII
receptor and reduces the binding affinity to FcyRIIIA receptor. A modification
can be a
substitution of H268, such as H268A, according to the EU index of Kabat. A
modification can be a substitution of R301, such as R301A, according to the EU
index
of Kabat. A modification can be a substitution of K322, such as K322A,
according to
10 the EU index of Kabat.
101351 In some embodiments, the modification comprises substitution of one or
more amino acids that decreases binding affinity of an IgG Fc domain to
Fcy11.11
receptor but does not affect the binding affinity to FcyRIIIA receptor. A
modification
can be a substitution of R292, such as R292A, according to the EU index of
Kabat. A
15 modification can be a substitution of K414, such as K414A, according to
the EU index
of Kabat.
[0136] In some embodiments, the modification comprises substitution of one or
more amino acids that decreases binding affinity of an IgG Fc domain to FcyRII

receptor and increases the binding affinity to FcyRIIIA receptor. A
modification can be
20 a substitution of S298, such as S298A, according to the EU index of
Kabat. A
modification can be substitution of S239, 1332 and A330, such as
S239D/1332E/A330L
A modification can be substitution of S239 and 1332, such as S239D/I332E.
[0137] In some embodiments, the modification comprises substitution of one or
more amino acids that decreases binding affinity of an IgG Fc domain to
FcyRIIIA
25 receptor. A modification can be substitution of F241 and F243, such as
F241S/F243S
or F241I/F2431, according to the EU index of Kabat.
[0138] In some embodiments, the modification comprises substitution of one or
more amino acids that decreases binding affinity of an IgG Fc domain to
FcyRIIIA
receptor and does not affect the binding affinity to FcyRII receptor. A
modification can
30 be a substitution of S239, such as S239A, according to the EU index of
Kabat. A
modification can be a substitution of E269, such as E269A, according to the EU
index
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
of Kabat. A modification can be a substitution of E293, such as E293A,
according to
the EU index of Kabat. A modification can be a substitution of Y296, such as
Y296F,
according to the EU index of Kabat. A modification can be a substitution of
V303,
such as V303A, according to the EU index of Kabat. A modification can be a
5 substitution of A327, such as A327G, according to the EU index of Kabat.
A
modification can be a substitution of K338, such as K338A, according to the EU
index
of Kabat. A modification can be a substitution of D376, such as D376A,
according to
the EU index of Kabat.
[0139] In some embodiments, the modification comprises substitution of one or
10 more amino acids that increases binding affinity of an IgG Fc domain to
FcyRITEA
receptor and does not affect the binding affinity to FcyRII receptor. A
modification can
be a substitution of E333, such as E333A, according to the EU index of Kabat.
A
modification can be a substitution of K334, such as K334A, according to the EU
index
of Kabat. A modification can be a substitution of A339, such as A339T,
according to
15 the EU index of Kabat. A modification can be substitution of S239 and
1332, such as
5239D/1332E.
[0140] In some embodiments, the modification comprises substitution of one or
more amino acids that increases binding affinity of an IgG Fc domain to
FcyRIIIA
receptor. A modification can be substitution of L235, F243, R292, Y300 and
P396,
20 such as L235V/F243L/R292P/Y300L/P396L (IgG1VLPLL) according to the EU
index
of Kabat. A modification can be substitution of S298, E333 and K334, such as
5298A/E333A/K334A, according to the EU index of Kabat. A modification can be
substitution of K246, such as K246F, according to the EU index of Kabat.
[0141] Other substitutions in an IgG Fc domain that affect its interaction
with
25 one or more Fey receptors are disclosed in U.S. Patent Nos. 7,317,091
and 8,969,526
(the disclosures of which are incorporated by reference herein).
[0142] In some embodiments, an IgG Fc domain comprises at least one amino
acid substitution that reduces the binding affinity to FcRn, as compared to a
wild-type
or reference IgG Fc domain. A modification can comprise a substitution at
H435, such
30 as H435A according to the EU index of Kabat. A modification can comprise
a
substitution at 1253, such as I253A according to the EU index of Kabat. A
modification
36
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
can comprise a substitution at H310, such as H310A according to the EU index
of
Kabat. A modification can comprise substitutions at 1253, H310 and H435, such
as
I253A/H310A/H435A according to the EU index of Kabat.
[0143] A modification can comprise a substitution of one amino acid residue
5 that increases the binding affinity of an IgG Fc domain for FcRn,
relative to a wildtype
or reference IgG Fc domain. A modification can comprise a substitution at
V308, such
as V308P according to the EU index of Kabat. A modification can comprise a
substitution at M428, such as M428L according to the EU index of Kabat. A
modification can comprise a substitution at N434, such as N434A according to
the EU
10 index of Kabat or N43411 according to the EU index of Kabat. A
modification can
comprise substitutions at T250 and M428, such as T250Q and M428L according to
the
EU index of Kabat. A modification can comprise substitutions at M428 and N434,
such
as M428L and N434S, N434A or N434H according to the EU index of Kabat_ A
modification can comprise substitutions at M252, S254 and T256, such as
15 M252Y/S254T/T256E according to the EU index of Kabat. A modification can
be a
substitution of one or more amino acids selected from P257L, P257N, P2571,
V279E,
V279Q, V279Y, A281S, E283F, V284E, L306Y, T307V, V308F, Q311V, D376V, and
N434H. Other substitutions in an IgG Fc domain that affect its interaction
with FeRn
are disclosed in U.S. Patent No. 9,803,023 (the disclosure of which is
incorporated by
20 reference herein).
[0144] In some embodiments, an antibody is a human IgG2 antibody, including
an IgG2 Fc region. In some embodiments, the heavy chain of the human IgG2
antibody
can be mutated at cysteines at positions 127, 232, or 233. In some
embodiments, the
light chain of a human IgG2 antibody can be mutated at a cysteine at position
214. The
25 mutations in the heavy and light chains of the human IgG2 antibody can
be from a
cysteine residue to a serine residue.
Immune-Stimulatory Compounds
[0145] The antibodies are attached to immune stimulatory compounds, typically
via a linker(s) to form immune-stimulatory conjugates. An antibody can be
attached to
37
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
one or more immune-stimulatory compounds, typically from about 1 to about 10
compounds per antibody.
[0146] In some embodiments, an immune stimulatory compound activates
human immune cells, including but not limited to dendritic cells, macrophages,
5 monocytes, myeloid-derived suppressor cells, NK cells, B cells, T cells,
or tumor cells,
or a combination thereof. In some embodiments, an immune-stimulatory compound
is
a myeloid cell agonist. A myeloid cell agonist is a compound that activates or

stimulates an immune response by a myeloid cell. For example, a myeloid cell
agonist
can stimulate an immune response by causing the release of cytokines by
myeloid cells,
10 which results in the activation of immune cells. The stimulation of an
immune response
by a myeloid cell agonist can be measured in vitro by co-culturing immune
cells (e.g.,
peripheral blood mononuclear cells (PBMCs)) with cells targeted by the
conjugate and
measuring cytokine release, chemokine release, proliferation of immune cells,
upregulation of immune cell activation markers, ADCC, or any combination
thereof.
15 Exemplary assays are described in the Examples. ADCC can be measured by
determining the percentage of remaining target cells in the co-culture after
administration of the conjugate with the target cells and PBMCs.
[0147] In general, an immune stimulatory compound acts on toll like receptors
(TLRs), nucleotide-oligomerization domain-like receptors (NOD), RIG-I-Like
receptors
20 (RLR), C-type lectin receptors (CLR), or cytosolic DNA Sensors (CDS), or
a
combination thereof.
[0148] In some embodiments, an immune stimulatory compound comprises a
ligand of one or more TLRs selected from the group consisting of: TLR2, TLR3,
TLR4,
TLR5, TLR7, TLR8, TLR7/TLR8, TLR9, and TLR10.
25
[0149] In some embodiments, an immune-stimulatory
compound is a myeloid
cell agonist. In some embodiments, a myeloid cell agonist is a ligand of TLR2
selected
from the group consisting of: (a) a heat killed bacteria product, preferably
HKAL,
HKEB, HK_HP, HKLM, HKLP, HKLR, HKMF, HKPA, HKPG, or HKSA, HKSP, and
(b) a cell-wall components product, preferably LAM, LM, LPS, LIA, LIA, PGN,
FSL,
30 Pam2CSK4, Pam3CSK4, or Zymosan.
38
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
101501 In some embodiments, a myeloid cell agonist is a ligand of TLR3
selected from the group consisting of: rintatolimod, poly-ICLC, RIBOXXONO,
Apoxxim, RIBOXXIMO, MCT-465, MCT-
475, and ND-1.1.
101511 In some embodiments, a myeloid cell agonist is a ligand of TLR4
5 selected from the group consisting of LPS, MPLA or a pyrimido[5,4-
b]indole such as
those described in PCT Publication No. WO 2014/052828 (U of Cal).
101521 In some embodiments, the myeloid cell agonist is a ligand of TLR5
selected from the group consisting of: FLA and Flagellin.
101531 In some embodiments, the myeloid cell agonist is a ligand of TLR6.
10 101541 In certain embodiments, a myeloid cell agonist is a TLR7
agonist and/or
a TLR8 agonist. In certain embodiments, the myeloid cell agonist is a TLR7
agonist.
In certain embodiments, the myeloid cell agonist is a TLR8 agonist. In some
embodiments, the myeloid cell agonist selectively agonizes TLR7 and not TLR8.
In
other embodiments, the myeloid cell agonist selectively agonizes TLR8 and not
TLR7.
15 101551 In certain embodiments, a myeloid cell agonist is a TLR7
agonist. In
certain embodiments, the TLR7 agonist is selected from an imidazoquinoline, an

imidazoquinoline amine, a thiazoquinoline, an aminoquinoline, an
aminoquinazoline, a
pyrido[3,2-d]pyrimidine-2,4-diamine, a pyrimidine-2,4-diamine, a 2-
aminoimidazole,
an 1-alkyl-1H-benzimidazol-2-amine, a tetrahydropyridopyrimidine, a
20 heteroarothiadiazide-2,2-dioxide, a benzonaphthyridine, a thieno[3,2-
d]pyrimidine, a 4-
amino-imidazoquinoline, an imidazo-ppidinone, an imidazo-pyrimidinone, a
purine, a
fused pyrimidine-lactam, an imidazo[4,5-c]quinoline-4-amine, an imidazo[4,5-
c]quinoline, a pyrimidine, a ben7.a7epine, an imidazo-pyridine, a pyrrolo-
pyrimidine, a
2-amino-quinazoline, a guanosine analog, an adenosine analog, a thymidine
25 homopolymer, an ssRNA, CpG-A, PolyG10, and PolyG3. In certain
embodiments, the
TLR7 agonist is selected from an imidazoquinoline, an imidazoquinoline amine,
a
thiazoquinoline, an aminoquinoline, an aminoquinazoline, a pyrido[3,2-
d]primidine-
2,4-diamine, a pyrimidine-2,4-diamine, a 2-aminoimidazole, a 1-alky1-1H-
benzimidazol-2-amine, a tetrahydropyridopyrimidine, a heteroarothiadiazide-2,2-

30 dioxide, a benzonaphthyridine, a thieno[3,2-d]pyrimidine, a 4-amino-
imidazoquinoline,
an imidazo-pyridinone, an imidazo-pyrimidinone, a purine, a fused pyrimidine-
lactam,
39
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
an imidazo[4,5-c]quinoline-4-amine, an imidazo[4,5-c]quinoline, a pyrimidine,
a
benzazepine, an imidazo-pyridine, a pyrrolo-pyrimidine, and a 2-amino-
quinazoline,
but is other than a guanosine analog, an adenosine analog, a thymidine
homopolymer,
an ssRNA, CpG-A, PolyG10, and PolyG3. In some embodiments, a TLR7 agonist is a
5 non-naturally occurring compound. Examples of TLR7 modulators include GS-
9620,
GSK-2245035, imiquimod, resiquimod, DSR-6434, DSP-3025, IMO-4200, MCT-465,
MEDI-9197, 3M-051, SB-9922, 3M-052, Limtop, TMX-30X, TMX-202, RG-7863,
RG-7795, and the TLR7 modulator compounds disclosed in US Patent Publication
No.
2016/0168164 (Janssen, thieno[3,2-d]pyrimicline derivatives), US Patent
Publication
10 No. 2015/0299194 (Roche, 4-amino-imidazoquinoline derivatives), US
Patent
Publication No. 2011/0098248 (Gilead Sciences, imidazo-pyridinone, imidazo-
pyrimidinone, and purine derivatives), US Patent Publication No. 2010/0143301
(Gilead Sciences, fused pyrimidine-lactam derivatives), and US Patent
Publication No.
2009/0047249 (Gilead Sciences, purine derivatives), and these publications are
15 incorporated by reference herein. Further examples of TLR7 modulators
include
compounds disclosed in PCT Publication No. W02018/009916 (Stanford
University/Bolt Biotherapeutics, imidazo[4,5-c]quinolin-4-amine derivatives),
PCT
Publication No. W02018/112108 (Bolt Biotherapeutics, imidazo[4,5-c]quinoline,
pyrimidine, benzazepine, imidazo-pyridine, pyrrolo-pyrimidine, and purine
20 derivatives), US Patent Publication No. 2019/0055247 (Bristol-Myers
Squibb, purine
derivatives), PCT Publication No. PCT Publication No. W02018/198091 (Novartis,

pyrrolo-pyrimidine derivatives), US Patent Publication No. 2017/0121421
(Novartis,
pyrrolo-pyrimidine derivatives), US Patent No. 10,253,003 (Janssen, 2-amino-
quinazoline derivatives), and US Patent No. 10,233,184 (Roche, imidazo-
pyrimidinone
25 derivatives), and these publications are incorporated by reference
herein. In some
embodiments, a TLR7 agonist has an EC50 value of 500 nM or less by PBMC assay
measuring TNFalpha or IFNgammaproduction. In some embodiments, a TLR7 agonist
has an EC50 value of 100 nM or less by PBMC assay measuring TNFalpha or
IFNgamma production. In some embodiments, a TLR7 agonist has an EC50 value of
30 50 TIM or less by PBMC assay measuring TNFalpha or IFNgamma production.
In some
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
embodiments, a TLR7 agonist has an EC50 value of 10 tiM or less by PBMC assay
measuring TNFalpha or IFNgamma production.
[0156] In certain embodiments the myeloid cell agonist is a TLR8 agonist. In
certain embodiments, the TLR8 agonist is selected from the group consisting of
a
5 benzazepine, an imidazoquinoline, a thiazoloquinoline, an aminoquinoline,
an
aminoquinazoline, a pyrido[3,2-d]pyrimidine-2,4-diamine, a pyrimidine-2,4-
diamine, a
2-aminoimidazole, an 1-allcyl-1H-benzimidazol-2-amine, a
tetrahydropyridopyrimidine,
a pyrido[3,2-d]pyrimidine, a dihydropyrimidinyl benzazepine carboxamide, a
benzo[b]azepine, benzazepine dicarboxamide derivatives with a tertiary amide,
10 benzazepine dicarboxamide derivatives with a secondary amide, a
quinazoline, a
pyrido[3,2-d]pyrimidine, a diamino-pyrimidine, an amino-quinazoline, a
heterocyclic-
substituted 2-amino-quinazoline, a diamino-pyrimidine, a piperidino-
pyrimidine, an
alkylamino-pyrimidine, an 8-substitued benzoazepine, an amino-diazepine, an
amino-
benzo-diazepine, an amido-indole, an amido-benzimidazole, a phenyl
sulfonamide, a
15 dihydropteridinone, a fused amino-pyrimidine, a quinazoline, a pyrido-
pyrimidine, an
amino-substituted benzazepine, a pyrrolo-pyridine, an imidazo-pyridine
derivatives, an
amino-benzazepine, and a ssRNA. In certain embodiments, a TLR8 agonist is
selected
from the group consisting of a benzazepine, an imidazoquinoline, a
thiazoloquinoline,
an aminoquinoline, an aminoquinazoline, a pyrido[3,2-d]pyrimidine-2,4-diamine,
a
20 pyrimidine-2,4-diamine, a 2-aminoimidazole, an 1-alkyl-1H-benzimidazol-2-
amine, a
tetrahydropyridopyrimidine, a pyrido[3,2-d]pyrimidine, a dihydropyrimidinyl
benzazepine carboxamide, a benzo[b]azepine, benzazepine dicarboxamide
derivatives
with a tertiary amide, benzazepine dicarboxamide derivatives with a secondary
amide, a
quinazoline, a pyrido[3,2-d]pyrimidine, a diamino-pyrimidine, an amino-
quinazoline, a
25 heterocyclic-substituted 2-amino-quinazoline, a diamino-pyrimidine, a
piperidino-
pyrimidine, an alkylamino-pyrimidine, an 8-substitued benzoazepine, an amino-
diazepine, an amino-benzo-diazepine, an amido-indole, an amido-benzimidazole,
a
phenyl sulfonamide, a dihydropteridinone, a fused amino-pyrimidine, a
quinazoline, a
pyrido-pyrimidine, an amino-substituted benzazepine, a pyrrolo-pyridine, an
imidazo-
30 pyridine derivatives, and an amino-benzazepine, and is other than a
ssRNA. In some
embodiments, a TLR8 agonist is a non-naturally occurring compound. Examples of
41
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
TLR8 agonists include motolimod, selgantolimod, resiquimod, 3M-051, 3M-052,
MCT-465, IMO-4200, VTX-763, VTX-1463, and the TLR8 modulator compounds
disclosed in US Patent Publication No. 2018/0086755 (Gilead, pyrido[3,2-
d]pyrimidine
derivatives), PCT Publication No. W02017/216054 (Roche, dihydropyrimidinyl
5 benzazepine carboxamide derivatives), PCT Publication No. PCT Publication
No.
W02017/190669 (Shanghai De Novo Pharmatech, benzo[b]azepine derivatives), PCT
Publication No. W02016/142250 (Roche, benzazepine dicarboxamide derivatives),
PCT Publication No. W02017/202704 (Roche, benzazepine dicarboxamide
derivatives
with a tertiary amide), PCT Publication No. W02017/202703 (Roche, benzazepine
10 dicarboxamide derivatives with a secondary amide), US Patent Publication
No.
2017/0071944 (Gilead, quinazoline and pynidop,2-d]pyrimdine derivatives), US
Patent
Publication No. 2014/0045849 (Janssen, diamino-pyrimidine derivatives), US
Patent
Publication No. 2014/0073642 (Janssen, amino-quinazoline derivatives), PCT
Publication No. W02014/056953 (Janssen, pyrrolo[3,2-d]pyrimidine derivatives),
PCT
15 Publication No. W02014/076221 (Janssen, heterocyclic substituted 2-amino-

quinazoline derivatives), PCT Publication No. W02014/128189 (Janssen, diamino-
pyrimidine derivatives), US Patent Publication No. 2014/0350031 (Janssen,
piperidino-
pyrimidine derivatives), PCT Publication No. W02014/023813 (Janssen, alkyl-
aminopyrimidine derivatives), US Patent Publication No. 2008/0234251 (Array
20 Biophanna, 8-substituted benzoazepine derivatives), US Patent
Publication No.
2008/0306050 (Array Biophartna, amino-diazepine derivatives), US Patent
Publication
No. 2010/0029585 (VentiRx Pharma, amino-benzazepine derivatives), US Patent
Publication No. 2011/0092485 (VentiRx Pharma, amino-benzazepine derivatives),
US
Patent Publication No. 2011/0118235 (VentiRx Pharma, amino-benzazepine
25 derivatives), US Patent Publication No. 2012/0082658 (VentiRx Pharma,
amino-
benzazepine VTX-378), US Patent Publication No. 2012/0219615 (VentiRx Pharma),

US Patent Publication No. 20140066432 (VentiRx Pharma, amino-benzazepine VTX-
2337), US Patent Publication No. 2014/0088085 (VentiRx Pharma, amino-
benzazepine
and amino-benzo-diazepine derivatives), US Patent Publication No. 2014/0275167
30 (Novira Therapeutics, amido-indole and amido-benzimidazole derivatives),
and US
Patent Publication No. 2013/0251673 (Novira Therapeutics, phenyl sulfonamide
42
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
derivatives), and these publications are incorporated by reference herein.
Further
examples of TLR8 modulators include compounds disclosed in U52016/0108045
(Gilead, dihydropteridinone derivatives), US Patent Publication No.
2018/0065938
(Gilead, fused amino-pyrimidine derivatives), US Patent Publication No.
2018/0263985
5 (Gilead, quinazoline and pyrido-pyrimidine derivatives), PCT Publication
No.
W02017/046112 (Roche, amino-substituted benzazepine derivatives), PCT
Publication
No. W02016/096778 (Roche, amino-substituted benzazepine derivatives), US
Patent
Publication No. 2019/0016808 (Birdie Biopharmaceuticals, pyrrolo- or imidazo-
pyridine derivatives or amino-benzazepine derivatives), and these publications
are
10 incorporated by reference herein. In some embodiments, the TLR8 agonist
comprises
NH2
L.
1.1 )
the structure:
, wherein the structure is
optionally substituted at any
position other than the -NM position. In some embodiments, a TLR8 agonist has
an
EC50 value of 500 n.M or less by PBMC assay measuring TNFalpha production. In
some embodiments, a TLR8 agonist has an EC50 value of 100 nNI or less by PBMC
15 assay measuring TNFalpha production. In some embodiments, a TLR8 agonist
has an
EC50 value of 50 tilvl or less by PBMC assay measuring TNFalpha production. In

some embodiments, a TLR8 agonist has an EC50 value of 10 nM or less by PBMC
assay measuring TNFalpha production.
[0157] In some embodiments, a TLR8 agonist is a benzazepine selected from
20 compounds provided herein.
101581 In some embodiments, a myeloid cell agonist is a ligand of TLR9
selected from the group consisting of: ODN1585, ODN1668, ODN1826, PF-3512676
(0DN2006), 0DN2007, 0DN2216, 0DN2336, 0DN2395, BB-001, BB-006, CYT-
003, IM0-2055, IM0-2125, IMO-3100, IMO-8400, IR-103, IM0-9200, agatolimod,
25 DIMS-9054, DV-1079, DV-1179, AZD-I419, leftolimod (MGN-1703), litenimod,
and
CYT-003-QbGl0
[0159] In other embodiments, the myeloid agonist selectively agonizes TLR9,
TLR3, TLR4, TLR2, TLR5, MG-I, STING, cGAS, NOD1, NOD2, NOD1/NOD2,
NRLP3, ALPK I, MDA5 AIN42,1RE1, or PERK.
30 [0160] In some embodiments, a myeloid cell agonist is a ligand of
TLR10.
43
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0161] In some embodiments, a myeloid cell agonist is a ligand of a ligand of
nucleotide-oligomerization domain (NOD)-like selected from the group
consisting of:
NOD! agonist (C12-iE-DAP, iE-DAP, Tri-DAP), NOD2 agonist (L18-MDP, MDP, M-
TriLYS, M-TriLYS-D-ASN, Murabutide, N-Glycolyl-MDP), and NOD1/NOD2
5 agonists (M-TriDAP, PGN).
[0162] In some embodiments, a myeloid cell agonist is a ligand of one or more
RIG-I-Like receptors (RLR) selected from the group consisting of: Sippp-dsRNA,
Poly
(dA:dT), Poly(dG:dC), and Poly (I:C).
[0163] In some embodiments, a myeloid cell agonist is a ligand of one or more
10 C-type Iectin receptors (CLR) selected from the group consisting of:
Cnrdlan AL,
RICCA, HKSC, WGP, Zymosan, and Trehalose-6,6-dibehenate.
[0164] In some embodiments, a myeloid cell agonist is a ligand of one or more
Cytosolic DNA Sensors (CDS) selected from the group consisting of: ADU-S100, c-

GMP, c-G-AMP, c-G-GMP, c-A-AMP, c-di-AMP, c-di-IMP, c-di-GMP, c-di-LTMP,
15 HSV-60, ISD, pCpG, Poly (dA:dT), Poly( dG:dC), Poly (dA),VACV-70 and a-
mangostin and the compounds disclosed in PCT Publication No. W02018/156625 (U
of Texas), PCT Publication No. WO 2018/152453 (Eisai), PCT Publication No.
W02018/138685 (Janssen), PCT Publication No. W02018/100558 (Takeda), PCT
Publication No. W02018/098203 (Janssen), PCT Publication No. W02018/065360
20 (Biolog Life Sciences), PCT Publication No, W02018/060323 (Boehringer
Ingelheim),
PCT Publication No. PCT Publication No. W02018/045204 (1FM Therapeutics), PCT
Publication No. PCT Publication No. W02018/009466 (Aduro), PCT Publication No.

WO 2017/161349 (Immune Sensor), PCT Publication No. W02017/123669, PCT
Publication No. W02017/123657, PCT Publication No. W02017/027646 (Merck),
25 PCT Publication No. W02017/027645 (Merck), PCT Publication No,
W02016/120305
(GSK), PCT Publication No. W02016/096174 (InvivoGen), and US Patent
Publication
No. 2014/0341976 (Aduro).
[0165] In some embodiments, the myeloid cell agonist is a ligand of an
inflammasome inducer selected from the group consisting of: (a) NLRP3
30 inflammasome protein complex, preferably alum Crystals, ATP, CPPD
Crystals,
44
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Hennozoin, MSU Crystals, Nano-Si 02, Nigericin, and (b) AIM2 inflammasome
protein
complex, such as Poly (dA:dT).
[0166] In certain aspects, a TLR8 agonist or a TLR7 agonist is selected from
Category A or Category B, respectively, as further described herein. Variables
and
5 Formula of the Compounds of Category A (TLR8 agonists) are described in
the section
entitled Compounds of Category A, and variables and Formula of the Compounds
of
Category B (TLR7 agonists) are described in the subsequent section, entitled
Compounds of Category B. Formulas and variables of the Compounds of Category A

and the Compounds of Category B may overlap in nomenclature, e.g., Formula IA
for
10 both Compounds of Category A and Category B; however, variables and
Formula
descriptions are not intended to be interchangeable between the catagories.
[0167] In some aspects, the myeloid cell agonist is a benzazepine-4-
carboxamide compound. In some aspects, the benzazepine-4-carboxamide compound
has the structure of Formula X-1:
H2N
=
* teR4
0
R11-R2
15 X-1
wherein:
RE is C3-7 alkyl;
112 is C3-7 alkyl or C3-7 cycloalkyl-CI-7a1ky1;
it is hydrogen;
20 it is selected from the group consisting of
Cl-7 alkyl, said C1-7 alkyl being unsubstituted or substituted by one or two
groups selected from the group consisting of phenyl and heteroaryl, said
heteraryl being
an aromatic 5- or 6-membered ring which comprises one, two, or three atoms
selected
from nitrogen, oxygen, and/or sulfur;
25 C3-7 cycloalkyl, said C3-7 cycloalkyl being unsubstituted or
substituted by phenyl
or phenylamino-C14 alkyl, and
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
heterocyclyl, said heterocyclyl being a saturated 3- to 7-membered ring
containing one heteroatom selected from N and 0 and being unsubstituted or
substituted by phenyl.
Structures of Formula X-1 are described, for example, in PCT Publication No.
W02017/202703.
101681 In some aspects, the the myeloid cell agonist is a benzazepine-
dicarboxamide compound. In some aspects, the benzazepine-dicarboxamide
compound
has the structure of Formula X-2:
H2N 0
ioi R3
0
14-~R2
R1
X-2
wherein:
RI is C3-7 alkyl;
R2 is C3-7 alkyl or C3-7 cycloalkyl-CI-7 alkyl;
R3 is a heterocycle selected from
xi x3
¨ise/ X \ N¨Z1
NX2 a,
X4
a)
wherein
Xi is (CH2)rn wherein m is 1 or 2;
X2 is (CH2)n wherein n is 1 or 2;
X3 is (CHOO wherein o is 1 or 2;
X4 is (CH2)p wherein p is 1 or 2; and
Z1 is phenyl, wherein phenyl is unsubstituted or substituted by one or
two groups selected from the group consisting of Ci.-7 alkyl, halogen, halogen-
C1-7
alkyl, CI-7 alkoxy, hydroxy-Ct-7 alkyl, amino-C1-7 alkyl, CI-7 alkyl-amino-C1-
7 alkyl,
and di-C1-7 alkyl-amino-C1-7 alkyl; or
46
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
/X\ õed.'22
¨N Y1
xe
b)
wherein
X5 is (CH2)q wherein q is 1 or 2;
X6, is (CHOr wherein r is 1 or 2;
5 Y1 is a carbon or nitrogen atom;
Z2 is hydrogen; and
Z3 is selected from the group consisting of hydrogen, C1-7 alkoxy, C2-7
alkenyloxy, phenyl, phenyl-C1-7 alkyl, phenyl-C1-7 alkyloxy, phenyl-C1-7
alkylamino,
phenylamino-C1-7 alkyl, phenylamino, wherein phenyl is unsubstituted or
substituted by
10 one or two groups selected from the group consisting of C1-7 alkyl,
halogen, halogen-Ci-
7 alk-yl, C1-7 alkoxy, hydroxy-CI-7 alkyl, amino-C1-7 alkyl, C1-7 alkyl-amino-
C1-7 alkyl,
and di-C1-7 alkyl-amino-C1-7 alkyl; or
/\ki
X7 Z4
\ ______________________________________________________
wherein
15 X7 is (CH2)s wherein s is 1 or 2; and
.14 is phenyl, wherein phenyl is unsubstituted or substituted by one or
two groups selected from the group consisting of Ct-7 alkyl, halogen, halogen-
C1-7
alkyl, C1-7 alkoxy, hydroxy-Ct-7 alkyl, amino-C1-7 alkyl, CI-7 alkyl-amino-C1-
7 alkyl,
and di-C1-7 alkyl-amino-C1-7 alkyl; or
1106
20 d)
wherein
Xs is (CH2)t wherein t is 1 or 2; and
47
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Z5 is phenyl, wherein phenyl is unsubstituted or substituted by one or
two groups selected from the group consisting of C1-7 alkyl, halogen, halogen-
CL-7
alkyl, C1-7 alkoxy, hydroxy-Ct-7 alkyl, amino-C1-7 alkyl, C I -7 alkyl-amino-
C1-7 alkyl,
and di-C1-7 alkyl-amino-C1-7 alkyl.
5 Compounds of Formula X-2 are described, for example, in
PCT
Publication No. W02017/202704.
101691 In some aspects, the the myeloid cell agonist is a benzazepine
sulfonamide compound. In some aspects, the benzazepine sulfonamide compound
has
the structure of Formula X-3:
R3
R4
H2N
I
6
Re
0
R2
RV
10 X-
3
wherein
R' and R2 are the same or different and are selected from the group consisting
of
CI-7 alkyl, hydroxy-C2-7 alkyl, amino-C2-7 alkyl, C2-7 alkenyl, and C3-7
alkynyl;
R5 is hydrogen or C1-7 alkyl;
15 R6 is hydrogen or C1-7 alkyl;
one of R4 and R5 is selected from the group consisting of hydrogen, Ct-7
alkyl,
halogen-C1-7 alkyl, and C1-7 alkoxy,
01
R
if
and the other one of R4 and R5 is
wherein R7 and Rs are the same or different and are selected from the group
20 consisting of hydrogen, C1-7 alkyl, halogen-C1-7 alkyl, hydroxy-Ct-7
alkyl, hydroxy-Ct-7
alkoxy-CI-7 alkyl, amino-C1-7 alkyl, CI-7 alkyl-amino-C1-7 alkyl, amino-CI-7
alkoxy-CI-7
alkyl, C1-7 alkyl-amino-Ct-7 alkoxy-C1-7 alkyl, amino-C1-7 alkyl-carbonyl, and
C1-7
alkyl-xamino-C 1-7 alkyl-carbonyl; or
R7 and Rs together with the nitrogen atom they are attached to form a 4- to 6-
25 membered heterocycle which is unsubstituted or substituted with a group
selected from
48
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
the group consisting of amino, C1-7 alkyl-amino, hydroxy, and hydroxy-C1-7
alkyl, and
which may contain an additional N-R1 group, wherein 10 is selected from the
group
consisting of hydrogen, amino-C1-7 alkyl, and C1-7 alkyl-amino-C1-7 alkyl; and
Y is N or CR9;
5 wherein R9 is selected from the group consisting of hydrogen, C1-7
alkyl, and
halogen-C1-7 alkyl.
Compounds of Formula X-3 are described, for example, in PCT Publication No.
W02016/096778.
01701 In some aspects, the myeloid cell agonist is a dihydropyrimidinyl
10 benzazepine carboxamide compound. In some aspects, the
dihydropyrimidinyl
benzazepine carboxamide compound has the structure of Formula X-4:
X
Re
H N
R5
H2N
õ 411
R4
Rs
0
RI/ n
X4
wherein
RE is C3-7 alkyl;
15 R2 is C3-7 alkyl or C3-7 cycloalkyl-CI-7 alkyl;
R3 is hydrogen or C1-7 alkyl;
R4 is hydrogen or C1-7 alkyl;
R5 is selected from the group consisting of hydrogen, halogen, C1-7 alkyl, and

CI-7 alkoxy;
20 Rs is selected from the group consisting of hydrogen, halogen, C1-
7 alkyl, and
CI-7 alkoxy; and
X is N or CR7, wherein R7 is selected from the group consisting of hydrogen,
halogen, CI-7 alkyl, and C1-7 alkoxy.
Compounds of Formula X-4 are described, for example, in PCT Publication No.
25 W02017/216054.
49
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
101711 In some aspects, the myeloid cell agonist is a sulfinylphenyl or
sulfonimidoylphenyl benzazepine compound. In some aspects, the sulfinylphenyl
or
sulfonimidoylphenyl benzazepine compound has the structure of Formula X-5:
R6 X Ra
H2N
-tee
R4
0
R
X-5
wherein
X is CR' or N;
IV is C3-7alkyl or C3-7cyc10a1ky1;
R2 is selected from the group consisting of C3-7 alkyl, hydroxy-C 1-7 alkyl,
C3-7-
alkynyl, amino-C1-7 alkoxy-Ct-7 alkoxy-C1-7 alkyl, halogen-C1-7 alkyl, and C3-
7
0 cycloalkyl-C1-7 alkyl;
0
II a
I a
one of R3 and It4 is , and
the other one of R3 and R4 is selected
from the group consisting of hydrogen, C14 alkyl, and halogen;
R5, 1(6, and R" are independently from each other selected from hydrogen, C1-7
alkyl, and halogen;
15 R.8 is C1-7 alkyl; and
R9 is absent or is =N-R' , wherein 1(10 is selected from the group consisting
of
hydrogen, C14 alkyl, halogen-CI-7 alkyl, hydroxy-C1-7 alkyl, and hydroxy-C1-7
alkoxy-
C 1-7 alkyl.
Compounds of Formula X-5 are described, for example, in PCT Publication No,
20 W02017/046112.
101721 In some aspects, the myeloid cell agonist is a TLR modulator compound
that has the structure of Formula X-6:
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
()
itz
So113
...-, :
4 1
Rb
X-6
wherein
(1) is a double bond or a single bond;
= (2) is a single bond or is double bond and RI is absent;
5 R2 and R3 are independently selected from H and lower alkyl, or R2
and 1(3 are
connected to form a saturated carbocycle haying from 3 to 7 ring members;
I N X
one of R7 and Rs is -NRfRg, 0
,or ,
and the other is hydrogen;
where Rf and Rg are lower alkyl or Rf and Rg together with the nitrogen to
which
10 they are attached form a saturated heterocyclic ring having 4 to 6 ring
members;
R4 is -NReRd or -ORro;
Re and Rd are lower alkyl, where the alkyl is optionally substituted with one
or
more -OH;
Rio is alkyl, where the alkyl is optionally substituted with one or more -OH;
15 Z is C and = (1) is a double bond, or Z is N and = (1) is a single
bond;
Ra and Rb are independently selected from H, alkyl, alkenyl, alkynyl, and Re,
wherein the alkyl is optionally substituted with one or more -0R10, or Re;
W is selected from -1\11-12, -NH(alkyl), and -N(alkyl)2;
RI is absent when = (2) is a double bond, or when = (2) is a single bond,
20 W and one of W or BY are taken together with the atoms to which they are
attached to
form a saturated, partially unsaturated, or unsaturated heterocycle having 5-7
ring
members, and the other of W or Rb is hydrogen or is absent as necessary to
accommodate ring unsaturation.
[0173] In some aspects, the myeloid cell agonist is a TLR modulator compound
25 that has the structure of Formula X-7:
51
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
P.Sv
los
le
R4
NFL,
X-7
wherein
Y is CF2CF3, CF2CF7R6, or an aryl or heteroaryl ring, wherein said aryl and
heteroaryl rings are substituted with one or more groups independently
selected from
5 alkenyl, alkynyl, Br, CN, OH, N1161V, C(3)R8, NR6S02R7, (CI-C6
alkyl)amino,
R60C(=0)CH=CH2¨, SR6 and 50211.6, and wherein the aryl and heteroaryl rings
are
optionally further substituted with one or more groups independently selected
from F,
Cl, CF, CF30-, HCF20-, alkyl, heteroalkyl and Ar0-;
RI-, R3 and R4 are independently selected from H, alkyl, alkenyl, alkynyl,
10 heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl and
heteroaryl, wherein the
alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, cycloalkenyl,
heterocycloalkyl, aryl and
heteroaryl are optionally substituted with one or more groups independently
selected
from alkyl, alkenyl, alkynyl, F, Cl, Br, I, CN, OR6, NR6R7, C(=0)R6, C(0)0R6,
OC(=0)R6, C(=0)NR6117, (C t-C6 alkyl)amino, CH3OCH20-, R60C(0)CH=CH2-,
15 NR6S02R7, SR6 and SO2R6,
or R3 and le together with the atom to which they are attached form a
saturated
or partially unsaturated carbocyclic ring, wherein the carbocyclic ring is
optionally
substituted with one or more groups independently selected from alkyl,
alkenyl,
alkynyl, F, Cl, Br, I, CN, OR6, NR6R7, C(0)R6, C(=0)0R6, OC(3)R6,
20 C(=O)NR6R7, (Ci-Cis alkyl)amino, CH3OCH20¨,11.60C(=0)CHH2-, NR6S02R7,
SR6 and S02R6;
1(2 and 1(8 are independently selected from H, OR6, NR6R7, alkyl, alkenyl,
alkynyl, heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl and
heteroaryl,
wherein the alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, cycloalkenyl,
25 heterocycloalkyl, aryl and heteroaryl are optionally substituted with
one or more groups
independently selected from alkyl, alkenyl, alkynyl, F, Cl, Br, I, CN, 01(6,
NR6R7,
52
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
4C(3)R6, g=0)0R6, OC(3)1;t6, C(0)NR61e, (Ci-C6 alkyl)amino, CH3OCH20-,
R60C(3)CHH2-, NR6S02R7, SR6 and SO2R6;
R5a, R51', and R5' are independently H, F, Cl, Br, I, OMe, CH3, CH2F, CHF2 or
CF3; and
5 R6 and le are independently selected from H, alkyl, alkenyl,
alkynyl,
heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl and heteroaryl,
wherein
said alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, cycloalkenyl,
heterocycloalkyl, aryl
and heteroaryl are optionally substituted with one or more groups
independently
selected from alkyl, alkenyl, alkynyl, F, Cl, Br, I, CN, OR6, NR6117, CD)R6,
10 C(3)0R6, OC(3)1t6, C(3)NR6R7, (CI-C6 alkyeamino, CH3OCH20-,
R60C(0)CHH2-, NR6S02Ie, SR6 and SO2R6,
or R6 and IV together with the atom to which they are attached form a
saturated
or partially unsaturated heterocyclic ring, wherein said heterocyclic ring is
optionally
substituted with one or more groups independently selected from alkyl,
alkenyl,
15 alkynyl, F, Cl, Br, I, CN, OW, NR6R7, C(=0)1e, C(=0)0R6, OC(3)R6,
C(D)NR6R7, (Ci-C6alkyl)amino, CH3OCH20-, R6OC(=D)CHCH2-, NR6S02R7, SR6
and SO2R6.
[0174] In some aspects, the myeloid cell agonist is a TLR modulator compound
that has the structure of Formula X-8:
ay_ R4 N,N'
twin-
.N
20 X-
8
wherein
W is -C(0)-;
Z is H, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, heterocycloalkyl,
aryl,
heteroaryl, Ole or NR6F2, wherein the alkyl, alkenyl, alkynyl, heteroalkyl,
cycloalkyl,
25 heterocycloalkyl, aryl and heteroaryl are optionally substituted with
one or more groups
independently selected from alkyl, alkenyl, alkynyl. F, Cl, Br, I, CN, OR',
NR6R7,
53
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
4C(3)R6, g=0)0R6, OC(3)1;t6, C(3)NR61e, (Ci-C6 alkyl)amino, CH30CH20-,
R6OCCD)CHH2-, NR6S021V, SR6 and SO2R6;
RI, R2, R3 and R4 are independently selected from H, alkyl, alkenyl, alkynyl,
heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl and heteroaryl,
wherein
5 said alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, cycloalkenyl,
heterocycloalkyl,
aryl, and heteroaryl are optionally substituted with one or more groups
independently
selected from alkyl, alkenyl, alkynyl, F, CI, Br, I, CN, OR', NR6R7, C(D)R6,
C(3)0R6, OC(3)116, C(3)NR6R7, (CI-C6 alkyl)amino, CH3OCH20-,
R60C(C)CHH2-, NR6S02117, 51(6 and 502R6,
10 or le and R2 together with the atom to which they are attached
form a saturated
Of partially unsaturated carbocyclic ring, wherein said carbocyclic ring is
optionally
substituted with one or more groups independently selected from alkyl,
alkenyl,
alkynyl, F, Cl, Br, I, CN, OR6, NR6R7, C(=0)R6, C(=0)0R6, OC(=3)R6,
OHLONR6R7, (C1-C6 alkyl)amino, CH3OCH20-, R60C(=0)CHH2-, NR6S02R7,
15 SR6 and 502R6,
or R3 and R4 together are oxo;
R5 is H, F, Cl, Br, I, OMe, CH3, CH2F, CHF2, CF3 or CF2CF3;
re and R7 are independently selected from H, alkyl, alkenyl, alkynyl,
heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl, and heteroaryl,
wherein
20 said alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl cycloalkenyl,
heterocycloalkyl, aryl,
and heteroaryl are optionally substituted with one or more groups
independently
selected from alkyl, alkenyl, alkynyl, F, Cl, Br, I, CN, OR', NR612.7, C(3)R6,

C(:)0126, OC(1)R6, C(1)NR6R7, (CI-C6 alkyl)amino, CH3OCH20-,
R60C(=0)CHH2-, NR6S021C, SR6 and SO2R6;
25 or R6 and R7 together with the atom to which they are attached
form a saturated
or partially unsaturated heterocyclic ring, wherein said heterocyclic ring is
optionally
substituted with one or more groups independently selected from alkyl,
alkenyl,
alkynyl, F, Cl, Br, I, CN, OR6, NR6R7, C(=0)1e, C(=0)0R6, OC(3)R6,
C(=0)NR6R7, (Ci-C6 alkyl)amino, CH3OCH20-, R60C(1)CHC112-, NR6S02R7,
30 SR6 and SO2R6; and
n is 0, 1, 2,3 or 4.
54
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
101751 Compounds of Formula X-6, X-7, and X-8 are described, for example, in
U.S. Publication Nos. US2019/0016808 and U.S. Publication No. 2014/0088085.
[0176] In some aspects, the myeloid cell agonist is a TLR modulator compound
that has the structure of Formula X-9:
H2N
04
we
Al
0
N,R2
5 R1
X-9
wherein
RI is C3-7alkyl or C3-7cycloalkyl;
R2 is selected from the group consisting of C1-7a1ky1, hydroxy-C1-7alkyl, C2-
7a1keny1, C3-7a11cyny1, amino-C1-7alkoxy-C1-7alkyl, amino-C1-7alkoxy-C1-
7alkoxy-Ci-
10 7a1ky1, halogen-C1-7allcyl, C3-7cycloalkyl-CL-7alkyl, and phenyl-CL-
7alkyl, wherein
phenyl is unsubstituted or substituted by amino-C1-7alkyl;
R3 is hydrogen;
le is selected from the group consisting of
phenyl, said phenyl being unsubstituted or substituted by one or two groups
15 selected from the group consisting of C1-7alkyl, halogen, halogen-C1-
7alkyl, C1-7a1k0xy,
hydroxy-Ci_7alkyl, amino-C1-7a1ky1, CE-7a1ky1-amino-C1-7a1ky1,
amino-C2-7alkenyl, Ci-7alkyl-amino-C2-7alkenyl, di-C1-7alkyl-amino-C2-
7alkenyl,
amino-C2-7alkynyl, C1-7alkyl-amino-C2-7alkynyl, di-C1-7alkyl-amino-C2-
7alkynyl,
benzyloxycarbonylamino-C1-7alkyl, amino-C1-7a1k0xy, amino-C1-7alkoxy-C1-
7alkoxy,
20 amino-C1-7alkoxy-C1-7alkyl, amino-C1-7alkoxy-C1-7alkoxy-C1-7alkyl,
Cialkylsulfonyl,
heterocyclylcarbonyl, and phenyl-C1-7alkyl, wherein phenyl is unsubstituted or

substituted by Ci-7alkoxy or amino-C1-7alkyl; or
heteroaryl, said heteroaryl being a 5- or 6-membered aromatic ring containing
one, two, or three heteroatoms selected from N, 0, or S. and being
unsubstituted or
25 substituted by one or two groups selected from the group consisting of
C1-7 alkyl,
halogen, halogen-C1-7 alkyl, C1-7 alkoxy, hydroxy-C1-7 alkyl, amino-C1-7
alkyl, C1-7
alkyl-amino-C1-7 alkyl, di-C1-7 alkyl-amino-Ci-7 alkyl, amino-C2-7 alkenyl, C1-
7 alkyl-
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
amino-C2-7 alkenyl, alkyl-amino-C2-7
alkenyl, amino-C2-7 alkynyl, 0-7 alkyl-
amino-C2-7 alkynyl, di-C1-7 alkyl-amino-C2-7 alkynyl, benzyloxycarbonylamino-0-
7
alkyl, amino-0-7 alkoxy, amino-C1-7 alkoxy-0-7 alkoxy, amino-CI-7 alkoxy-C1-7
alkyl,
amino-C1-7 alkoxy-0-7 alkoxy-C1-7 alkyl, CI-7 alkylsulfonyl,
heterocyclylcarbonyl, and
5 phenyl-CI-7 alkyl, wherein phenyl is unsubstituted or substituted by CI-7
alkoxy or
amino-C14 alkyl.
Compounds of Formula X-9 are described, for example, in PCT Publication No.
W02016/142250.
Compounds of Category A, TLR8 Agonists
10 101771 In some aspects, the present disclosure provides a TLR8
agonist
represented by the structure of Formula (IA):
R1
N-R2
N
Li ,
R5
L2
(IA)
or a pharmaceutically acceptable salt thereof, wherein:
¨ represents an optional double bond;
15 LIO is _xl0-;
L2 is selected from -X2-, -X2-CI-6alkylene-X2-, -X2-C2-6alkenylene-X2-, and -
X2-C2-&alkynylene-X2-, each of which is optionally substituted on alkylene,
alkenylene
or alkynylene with one or more R12;
X10 is selected from -C(0)-, and -C(0)N(10 )-*, wherein * represents where X1
20 is bound to Its;
X2 at each occurrence is independently selected from a bond, -0-, -S-, -N(R10)-
,
-C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(10 )-, -C(0)N(R1 )C(0)-,
C(0)N(R1 )C(0)N(Rio), _Nazto-Kok, _N(io)c(0)mai0) _N(IC )n% 10,
C(0)0-,
-0C(0)N(Rio)_, _c(NRio)_, 44-Rio)c(NRio,_,
) -C(NR1 )N(R.1 )-,
25 -N(R1 )C(NR1 )N(R1 )-, -S(0)2-, -0S(0)-, -S(0)0-, -S(0), -OS(0)2-, -S(0)20,
56
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-N(10)S(0)2-, -S(0)2N(Rio)-, _N(zio)s(0)_, -S(0)N(Rm)-, -N(RnS(0)2N(10)-, and
-N(R1 )S(0)N(10)-;
R' and 11.2 are independently selected from hydrogen; and Ci-io alkyl, C2-11)
alkenyl, and C2-10 alkynyl, each of which is optionally substituted with one
or more
5 substituents independently selected from halogen, -OR',
-C(0)N(10)2,
-N(10)2, -S(0)10, -S(0)210, -C(0)10, -C(0)0R' , -0C(0)10, -NO2, =0, =S,
=N(R1 ), and -CN;
is selected from: -OR lo, _
N(10)2, -C(0)N(R1 )2, -C(0)10 , -C(0)0R'

,
-S(0)R1 , and -S(0)2R1 ; Ci-io alkyl, C2-lo alkenyl, C2-lo alkynyl, each of
which is
10 optionally substituted with one or more substituents independently
selected from
halogen, -OR',
= -C(0)N(Rt )2, -N(10 )C(0)R10, -
N(10)C(0)N(R1 )2, -N(R1 )2,
-C(0)R' , -C(0)0R1 , -0C(0)R1 , -NO2, =0, =S, =N($1 ), -CN, C3-12 carbocycle,
and
3-to 12-membered heterocycle; and C3-12 carbocycle, and 3-to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in fe is
15 optionally substituted with one or more substituents independently
selected from
halogen, -OR',
= -C(0)N(10)2, -N(10)C(0)R' , -
N(R1 )C(0)N(R1 )2, -N(R10)2,
-C(0)R1 , -C(0)0R10, -0C(0)R' , -NO2, =0, =S, =N(F0), -CN, C1-6alkyl,
C24alkenyl,
and C2-6 alkynyl;
12.5 is selected from unsaturated C4-8 carbocycle; bicyclic carbocycle; and
fused
20 5-5, fused 5-6, and fused 6-6 bicyclic heterocycle, wherein R5 is
optionally substituted
and wherein substituents are independently selected at each occurrence from:
halogen, -OR', -S10, -C(0)N(10)2, -N(10)C(0)R10

,
-N(R1 )C(0)N(R1 )2, -N(10)2, -C(0)10, -C(0)0R1 , -0C(0)10, -NO2, =S,
=N(Rm), and -CN;
25
Ci-to alkyl, C2-10 alkenyl, C2-10alkynyl, each of which
is optionally
substituted with one or more substituents independently selected from halogen,
-010,
-SW , -C(0)N(Rio)2, -N(R10)C(0)R10, 2
4,4(Rio)ceD0A-Rto,), _ NOV12, -C(0)10,
-C(0)0R' , -0C(0)10, -NO2, =0, =S, =N(10), -CN, C3-12 carbocycle, and 3- to 12-

membered heterocycle;
57
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
and C3-I2 carbocycle, and 3- to 12-membered heterocycle, wherein each
C3-12 carbocycle, and 3- to 12-membered heterocycle in R5 is optionally
substituted with
one or more substituents independently selected from halogen, -0R1 , -SR1 ,
-C(0)N(R10)2, _N(Rio),c(o)Rto,
-N(R10)C(c)N(R10)2,
-iNkicen1 )2, -C(0)R1 , -C(0)0R1 ,
5 -0C(0)R' , -NO2, =0, =S, =N(Rw), -CN, CI-6 alkyl, C2-6 alkenyl, and C2-6
alkynyl;
le is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
10 -C(0)0CH2C6H5, -NHC(0)0CH2C6H5, C1-10 alkyl, -Ci-io haloalkyl, -0-C 1-10
alkyl, C2-10
alkenyl, C2-10 alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle, and
haloalkyl;
and
R12 is independently selected at each occurrence from halogen, -OW',
Nio _sr, to, _(t)2,-C(0)R1 , -C(0)N(R10)2, -N(R1 )C(0)R1 , -C(0)0R1 , -0C(0)R1
,
15 -S(0)R1 , -S(0)2Rto, _p(0)(OR to)2, -0P(0)(0R1 )2, -NO2, =0, =S, =N(Rm),
and -CN;
Ci-io alkyl, C2-10 alkenyl, C2-to alkynyl, each of which is optionally
substituted with one
or more substituents independently selected from halogen, -OW , -SRm, -N(Rm)2,
-C(0)Rm, -C(0)N(R10)2,
)1.(0)R10, -C(0)0R1 , -0C(0)R10, -S(0)Rm, -S(0)2R10

,
_p(0)(oRio)2,
-0P(0)(010 )2, -NO2, =0, =S, =N(R1 ), -CN, C3-10 carbocycle and 3- to
20 10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-ORm,
-SW , -N(R10)2, -C(0)R1 , -C(0)N(R10)2, -N(Rm)C(0)R1 , -C(0)0R1 , -0C(0)R1 ,
_s(0)Rto, _s(0)2Rto, _p(0)(oRto)2,
OP(0)(0R1 )2, -NO2, =0, =S, =N(Rm), -CN,
25 Ct-o alkyl, C2-6 alkenyl, C2-6 alkynyl;
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from Ry or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle.
101781 In some embodiments, the compound of Formula (HA) is represented by
30 Formula (HB):
58
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
R1
R2
N-R2
R25
n. 42
R21
1-2-R4
R22 R-9-
,1
(IIB)
or a pharmaceutically acceptable salt thereof, wherein:
R20, 1(21, n22,
lc and R23 are independently
selected from hydrogen,
halogen, -OW , -SRP, -N(Rw)2, -S(0)R' , -S(0)2111 , -C(0)R1 , -C(0)010 ,
5 -0C(0)R1 , -NO2, =0, =S, =N(R1 ), -CN, C,-to alkyl, C2-10 alkenyl, and C2-
10 alkynyl;
and
R24 and Kn25
are independently selected from hydrogen, halogen, -OR', -SR10

,
-N(Rm)2, -S(0)11.1 , -S(0)2R10, -C(0)R1 , -C(0)011.1 , -0C(0)R1 , -NO2, 0, =S,

=Novo.% -CN, Ci-to alkyl, C2-to alkenyl, and C2-10 aIkynyl; or 1(24 and R25
taken together
10 form an optionally substituted saturated C3-7 carbocycle.
[0179] In some embodiments, R20, R21, R22, and tc. n 23
are independently selected
from hydrogen, halogen, -OH, -0R' , -NO2, -CN, and Ct-to alkyl. R20, R21, R22,
and R23
may be each hydrogen. In certain embodiments, R21 is halogen. In certain
embodiments, 1(21 is hydrogen. In certain embodiments, 1(21 is -0R10. For
example,
15 R21 may be -OCH3.
[0180] In some embodiments, R24 and R25 are independently selected from
hydrogen, halogen, -OH, -NO2, -CN, and Ci-to alkyl, or R24 and R25 taken
together form
an optionally substituted saturated C3-7 carbocycle. In certain embodiments,
R24 and R25
are each hydrogen. In other embodiments, 1(24 and R25 taken together form an
20 optionally substituted saturated C3-5 carbocycle, wherein substituents
are selected from
halogen, -010, -SR1 , 2 -
C(0)N(R1o),,
N(R1 )C(0)R1 , -N(R15C(0)N(R1 )2, -N(R10)2,
-C(0)R1 , -C(0)0R' , -0C(0)R' , -NO2, =0, =S, =N(R1 ), and -CN; and Ci-to
alkyl, C2-
alkenyl, C2-10 alkynyt, each of which is independently optionally substituted
with one
or more substituents independently selected from halogen, -0R1 ,
-C(0)N(R1 )2,
25 -N(Rio)cor _
N(Rm)C(0)-N(R1 )2, - t-
tAR ) _
C(0)R1 , -0(0)0RE , -0C(0)R10

,
-NO2, =0, =S, =WW1, -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle.
59
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
[0181] In some embodiments, Ri is hydrogen. In some embodiments, R2 is
hydrogen. In some embodiments, R2 is¨C(0)-.
[0182] In some embodiments, LI is selected from -C(0)N(R1 )-*. In certain
embodiments, Rif) r _
or C(0)N(10 )-* is selected from hydrogen and CI-6 alkyl. For
5 example, 12 may be -C(0)NH-t.
[0183] In some embodiments, 11.5 is an optionally substituted bicyclic
carbocycle. In certain embodiments, R5 is an optionally substituted 8- to 12-
membered
bicyclic carbocycle. R5 may be an optionally substituted 8- to 12- membered
bicyclic
carbocycle substituted with one or more substituents independently selected
from
10 halogen, -OR', -N(Rw)2, -C(0)R' , -C(0)0R1", -0C(0)10- , -NO2,
=0, =S, -CN,
CL-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl. In certain embodiments, 12.5 is an
optionally
substituted 8- to 12- membered bicyclic carbocycle substituted with one or
more
substituents independently selected from -OR', -N(R1 )2, and =O. In some
embodiments, R5 is an optionally substituted indane, and optionally
substituted
04
15 tetrahydronaphthalene. R5 may be selected from:
and , any
one of which is optionally substituted. For example, the R5 is selected from:
0
OH
0
OH
41141:1 =140
, and CbzHN
[0184] In some embodiments, R5 is an optionally substituted unsaturated C4-8
20 carbocycle. In certain embodiments, le is an optionally substituted
unsaturated C4-6
carbocycle. In certain embodiments, leis an optionally substituted unsaturated
C4-6
carbocycle with one or more substituents independently selected from
optionally
substituted C342 carbocycle, and optionally substituted 3- to 12-membered
heterocycle.
It..5 may be an optionally substituted unsaturated C4-6 carbocycle with one or
more
25 substituents independently selected from optionally substituted phenyl,
optionally
substituted 3-to 12- heterocycle, optionally substituted Ci-to alkyl,
optionally
substituted C2-ioalkenyl, and halogen.
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
101851 In some embodiments, 11.5 is selected from an optionally substituted
fused
5-5, fused 5-6, and fused 6-6 bicyclic heterocycle. In certain embodiments, R5
is an
optionally substituted fused 5-5, fused 5-6, and fused 6-6 bicyclic
heterocycle with one
or more substituents independently selected from -C(0)0R'

,
Nit 1)2, -OW , and
5 optionally substituted Ci-io alkyl. In certain embodiments, le is an
optionally
substituted fused 5-5, fused 5-6, and fused 6-6 bicyclic heterocycle
substituted with
-C(0)0R' . In certain embodiments, R5 is an optionally substituted fused 6-6
bicyclic
heterocycle. For example, the fused 6-6 bicyclic heterocycle may be an
optionally
substituted pyridine-piperidine. In some embodiments,
is bound to a carbon atom
10 of the pyridine of the fused pyridine-piperidine. In certain
embodiments, Rs is selected
from tetrahydroquinoline, tetrahydroisoquinoline, tetrahydronaphthyridine,
cyclopentapyridine, and dihydrobenzoxaborole, any one of which is optionally
substituted. Pi may be an optionally substituted tetrahydronaphthyridine. In
some
embodiments, R5 is selected from:
15 N N
Olt FIN
FIN 110 N
0
craiN gal
so N 411
Cbz, N
NI-I2
NHCbz
Cbze N
_3/4.,
.ra)y
N
I
H
0
1411:1 C bre LI
N
I I
N
"====.,
0
61
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
0
H2N...3/4õ.."-..N
N
ratIsai H2W-...)CN
F F H
HO
c :LAIN
CbzHN-Ca
01
H2N CbzHN
and CbzHN
101861 In some embodiments, when R5 is substituted, substituents on R5 are
independently selected at each occurrence from: halogen, -OR"), -SR1 , -
C(0)N(R1 )2,
_mato-icor
K _ Witt )C(C)MRIC)2, _N(t10)2, _c(0)R10, -C(0)0R1 , -0c(c)R10

,
-NO2, =0, =S, =N(R1 ), and -CN; Ci-to alkyl, C2-10 alkenyl, C2-10 alkynyl,
each of which
is optionally substituted with one or more substituents independently selected
from
halogen, -ORM, _sr.K 10, _ C(0)N(Rio)2, _malaK(0)R10

,
p.,(0)N(R1 )2, -N(R1 )2,
-C(0)R1 , -C(0)010 , -0C(0)10, -NO2, =0, =S, =N(I0), -CN, C3-12 carbocycle,
and
3-to 12-membered heterocycle; and C3-12 carbocycle, and 3-to 12-membered
heterocycle, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R10, _SRIO, _c(c)N(Rto)2, _matoie,
(0)R1 ,
-N(R1 )C(0)N(R1 )2, -N(R1 )2, -C(0)R1 , -C(0)0111 , -0C(0)11.1 , -NO2, 0,=S,
=N(R1 ), -CN, C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl. In certain
embodiments, the
substituents on R5 are independently selected at each occurrence from:
halogen, -ORm,
-SR1 , -C(0)N(R10)2, -N(10)C(0)R10, -N(R10)C(0)N(R10)2,

-N(10)2, -C(0)10,
-C(0)010, -0C(0)R1 , -NO2, =0, =S, =N(R1 ), and -CN; Ci-to alkyl, C2-10
alkenyl,
C2-10 alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R' ,
-C(0)N(11.10)2, -N(R1 )C(0)R1 ,
-N(R1 )C(0)N(Rio)2, _Nc-R10)2, _cor _
C(0)010, -0C(0)11.10, -NO2, =0, =S,
=N(R1 ), -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle. In certain
embodiments, the substituents on R5 are independently selected at each
occurrence
from: halogen, -0R10, -C(0)N(11.10)2, -
N(Rto)c(0)Rio, _N(R10)2, _c(o)Rio,
-C(0)010, -0C(0)R1 , -NO2, =0, and -CN; and Ci-m alkyl optionally substituted
with
one or more substituents independently selected from halogen, -OR',
-N(I0)2,
-C(0)R' , -C(0)011.10, -NO2, =0, and -CN. In some embodiments, R5 is not
substituted.
62
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
[0187] In some embodiments, L2 is selected from -C(0)-, and -C(0)NR`o_. In
some embodiments, 0 is -C(0)-. In some embodiments, L2 is selected from
-C(0)NR1t. R1 of -C(0)NRIo_ may be selected from hydrogen and C1-6 alkyl. For

example, L2 may be -C(0)NH-.
5 [0188] In some embodiments, le is selected from: -01t1 , -N(R1 )2,
-C(0)N(R1 )2, -C(0)R1 , -C(0)0111 , -S(0)R1 , and -S(0)2R1 .; Ct-to alkyl, C2-
to alkenyl,
C2-10 alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R' , -SR1 , _c (c)N(Rto)2, 4.4(R1
)C(0)111 ,
-N(Rw)C(0)N(Rto)2, _N(t10)2, -C(0)R1 , -C(0)0R1 , -0C(0)R1 , -NO2, :), =S,
10 =N(1410), -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle; and
C3-12
carbocycle and 3- to 12-membered, each of which is optionally substituted with
one or
more substituents independently selected from halogen, -0R10, -SR1 , -
C(0)N(R10)2,
-N(Rw)C(0)10 , -N(R1 )C(0)N(Rto)2, _ in
)2
1,,(R__,, _ C(0)R1 , -C(0)0R' , -0C(0)R1 ,
-NO2, =0, =S, =N(R1 ), -CN, Ct-6alkyl, C2-6 alkenyl, and C2-6 alkynyl. In some
15 embodiments, R.4 is selected from: -0R1 , and -N(R1")2; and CLAD alkyl,
C2-10 alkenyl,
C2-10 alkynyl, C3-12 carbocycle, and 3- to 12-membered heterocycle, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, -OR', -SR1 , - N(R1 )2, -S(0)R10, -S(0)211.10, -C(0)12.1 , -C(0)0100

,
-0C(0)R1 , -NO2, =0, =S, =N(R1 ), -CN, Ct-to alkyl, C2-10 alkenyl, and C2-to
alkynyl.
20 In certain embodiments, R4 is _Ts(R10)2. .tc, n10
of -N(R10)2 may be independently selected
at each occurrence from optionally substituted C1-6 alkyl. In certain
embodiments, R1
of -N(R1 )2is independently selected at each occurrence from methyl, ethyl,
propyl, and
CCH3
tveN 1
butyl, any one of which is optionally substituted. For example, R4 may be
CH3.
rcH3
A.,ii_NI.._
0
In certain embodiments, -L2-R4 is
cH3.
25 [0189] In some embodiments, R12 is independently selected at each
occurrence
from halogen, -0R10, _SR10, _ma10)2, _
C(0)1e , -C(0)N(Rio)2, _N(Rto)cowto,
63
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-C(0)0R' , -0C(0)R10, -S(0)R10, -S(0)2R1 , -P(0)(0R10)2, -0P(0)(0R1 )2, -NO2,
=0,
=S, =N(R1- ), and -CN; Ct-io alkyl, C2-10 alkenyl, C2-lo alkynyl, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SR ' , _
N(Rw)2, -C(0)Rio, -C(0)N(R10)2, -N(RP)C(0)R1 , -C(0)0111 ,
5 -0C(0)R' , -S(0)Rt0, -S(0)2R10, -P(0)(0R1 )2, -0P(0)(010- )2, -NO2, =0,
=S,
=N(R10), -CN, C3-10 carbocycle and 3- to 10-membered heterocycle; and C3-10
carbocycle and 3- to 10-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -OR', -SR',
-N(R10)2, -C(0)Rio,
-C(0)N(R1 )2, -N(R1 )C(0' - )K 1 , C(0)0R1 , -0C(0)R1 , -S(0)11.1 ,
10 -S(0)2R1 , 13(0)(0R1 )2, -0P(0)(0R1 )2, -NO2, =0, =S, -N(R1 ), -CN, C1-6
alkyl, C2-6
alkenyl, and C2-6 alkynyl. In certain embodiments, R'2 is independently
selected at each
occurrence from halogen, -ORlo, _SR10, -MR10)2, -C(0)R10, -C(0)N(R10)2,
-N(R1 )C(0)R1 , -C(0)0R1 , -0C(0)R1 ., -S(0)R1 , -S(0)2R1 , -13(0)(0R1 )2,
-0P(0)(0R1 )2, -NO2, =0, =S, =N(R1 ), and -CN; and Ct-io alkyl, C2-to alkenyl,
C2-10
15 alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R' , _sty , _JAREN, _0(0)Rio,
_c(c)mRio)2,
-N(RE )C(0)R10, -C(0)0R10, -0C(0)R' , -S(0)R"), -S(0)21e, -P(0)(ORE )2,
-0P(0)(0RE0)2, -NO2, =0, =S, =N(R1 ), -CN, C3-10 carbocycle and 3- to 10-
membered
heterocycle.
20 101901 In some embodiments, the compound of Formula (IIB) is a
compound of
Formula (TC).
IR:
L1 N
N-R2
Rs- 0
L2_.4
(iic)
or a pharmaceutically acceptable salt thereof, wherein:
10- and R2 are hydrogen;
25 L2 is -C(0)-;
IV is -N(R1 )2;
RI is independently selected at each occurrence from hydrogen, -NI-12,
64
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-C(0)0CH2C6H5.; and Ci-to alkyl, C2-to alkenyl, Ca-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, Ct-to alkyl, -C I- to haloalkyl, -0-Ci-to
alkyl, C2-io
5 alkenyl, C2-10alkynyl, C3-12 carbocycle, 3- to 12-membered heterocycle,
and haloalkyl;
is _c(o)N(Rto)_*,
wherein * represents where 1,1 is bound to R5; and
R5 is a fused 5-5, fused 5-6, or fused 6-6 bicyclic heterocycle, wherein R5 is

optionally substituted and wherein substituents are independently selected at
each
occurrence from:
10 halogen, -0R10, -S100, -C(0)N(R1 )2, -N(W1C(0)R1 ,
-N(R1 )C(0)N(10 )2, -N(1012, -C(0)10- , -C(0)01013, -0C(0)10 , -NO2, =0, =S,
=N(10 ), and -CN;
CI-toalkyl, C2-to alkenyl, C2-to alkynyl, each of which is optionally
substituted with one or more substituents independently selected from halogen,
-OR',
15 -SR1 , -C(0)N(P..1 )2, -N(10 )C(0)Rw, -N(10 )C(0)N(R1 )2, -N(R112, -
COW),
-C(0)0R' , -0C(0)10 , -NO2, =0, =S, =N(101, -CN, C3-12 carbocycle, and 3- to
12-membered heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
20 halogen, OW , -S111 , -C(0)N(1012, -N(11.1 )C(0)10 , -
mitto)c(0)N-Rio)2, _N(Rto)2,
-C(0)R1 , -c(o)aRi , -0C(0)R' , -NO2, =0, =S, =N(Rw), -CN, C1-6alkyl, C2-
6alkertyl,
and C2-6alkynyl.
[0191] In certain embodiments, 10 of -N(I0 )2is independently selected at
each
occurrence from methyl, ethyl, propyl, and butyl, any one of which is
optionally
25 substituted, and/or 10 of -C(0)N(10 )-* is hydrogen.
[0192] In certain embodiments, R4 is -N(C14 alky1)2 and 1,1 is -C(0)N(H)-*.
CCH3
[0193] In certain embodiments, R4 is
CH3; and/or R1 of -
C(0)N(Rlo)_*
is hydrogen.
[0194] In some embodiments, the compound is selected from:
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
0 NH2
N, no
0 N
N..... NH2
----- re-\
iii *
N 0 --- Nre\
0
o \---\
o
, ,
OS 0
O. o
NH2
NH2
N,

N N
H
H
---- r---\ OH
---- is--\
N
N
OH
H
O.
N 0
* 0
NH2
NH2
N,
N,
N
* N
H
H
N
N
HN 0 0
NH2 , HN
4 0
NH2
,
N.,
)LcN
N
* N
H
H
--- rTh
--- /Th
N
N
,
0 ,
*
N 0
= N,H2
NH2
N.õ 4.
N N
N
0
0 \Th
0
A
* ON * 0
NH2
N,
11 * ....-
rms\
N
0 \---\
go% ,
40) 0
0
NH2
,N N,
N NH2
Cbz N Ili
N
H fei\ NH2
H ---- r"\
N
N
0
czill
cLii 0
,
NH2
NH2
N.,

N *NHCbz
r__\ H2N ...õ f---\
N N
0 \Th
0 \Th
66
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
21
NH2
N, NH2 40 te 0
cOs, I0
N,
N ..
N
CbzHN H
H
r-\ H2N
Ac
- r-\
N N
N
000 0 NH2
100 Nia.,-1
N._ NH2
N
N
HN% H
H
Cbz --- 11----
S\ /---\
N
0 \----\ 0 \--\
H
o uN
NH2 0
Cbz,, N co...N
--- 1 0
I
N..... NH2
HN --N., N,
N
N
H
H
--- r---\
--- Nf----\
N
,
N 0
HN
NH2
/ -41%,cu N,
Cbz N
H
N
0 \--\
,
H
0 0_.?
0 , * 0
NH2
N
B N
----
r H
HO
N
,
NH2
0 0 ....n,ACUN N 0
N
II H *
0
__.- r \
N
,
CbzHN
A.tri 40 ouN 0

NH2
N ----

---
r--\
N
67
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N
1 * rouN 0
NH2
HcI
N,
-% 0 0
NH2
=
40) N,N
NNH2
H3C0
0 \Th
0 \Th
H2N------CN it r-Dao
NH2
F F ^ N
N,
N
r-\
and
and a salt of any one thereof.
5 101951 In some aspects, the present disclosure provides a compound
represented
by the structure of Formula (IIIA):
R1
N¨R2
N
L11ICYTS
R4 R6
(IlIA)
or a pharmaceutically acceptable salt thereof, wherein:
¨ represents an optional double bond;
10 L" is -X"-;
L2 is selected from -X2-, -X2-C1-6 alkylene-X2-, -X2-C2-6 alkenylene-X2-, and
-X2-C2-6 alkynylene-X2-, each of which is optionally substituted on alkylene,
alkenylene
or alkynylene with one or more Rn;
X" is selected from -C(0)- and -C(0)N(11')-*, wherein * represents where X"
15 is bound to R6;
68
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
X2 at each occurrence is independently selected from a bond, -0-, -S-, -N(Rm)-
,
-C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(R10)-, -C(0)N(R1 )C(0)-,
C(0)N(R1 )C(0)N(R1 )-, -N(R1 )C(0)-, -N(R1 )C(0)N(R1 )-, -
tm-R ow(0)0_,
OC(0)N(R1 )-, -C(NRm)-, -
mato)c(NRio)_, _0(NRio)N(Rto)_, _mktonNRtox,4(Rio)_,
-S(0)2-, -0S(0)-, -S(0)0-, -S(0)-, -OS(0)2-, -S(0)20-, -N(1tm)S(0)2-, -
S(0)2N(Rm)-,
-N(144 )S(0)-, -S(0)N(141 )-, -N(141 )S(0)2N(Rm)-, and -N(1410)S(0)N(R1 )-;
141 and R2 are independently selected from hydrogen; Chi alkyl, C2-10
alkenyl,
and C2-to alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -OW , -SR'', -C(0)N(14.1 )2, -
11,w o)2, _s(D)Rto,
-S(0)2141 , -C(0)R10, -C(0)0141 , -OC(0)141 , -NO2, =0, =S, =N(R1 ), and -CN;
R4 is selected from: -Ole , -N(R1 )2, -C(0)N(R1 )2, -C(0)10 , -C(0)0R1 ,
-S(0)R1 , and -S(0)2R1 ; Ci-to alkyl, C2-to alkenyl, C2-10alkynyl, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SRI , -C(0)N(14.1 )2, -N(R1 )C(0)R1 , -N(R1 )C(0)N(R1 )2, -
N(R10)2,
-C(0)R' , -C(0)0R10, -0C(0)R' , -NO2, =0, =S, =N(Rm), -CN, C3-12 carbocycle,
and
3-to 12-membered heterocycle; and C3-12 carbocycle, and 3--to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in R4 is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SRI , -C(0)N(R1 )2, -N(R1 )C(0)R1 , -N(R1 )C(0)N(R1 )2, -N(R
112,
-C(0)141 , -C(0)0R10, -0C(0)14.1 , -NO2, =0, =S, =N(R10), -CN, C1-6 alkyl, C2-
6 alkenyl,
and C2-6alkynyl;
146 is selected from phenyl and 5- or 6- membered heteroaryl, any one of which

is substituted with one or more substituents selected from R7 and R6 is
further
optionally substituted by one or more additional substituents independently
selected
from R12;
R7 is selected from -C(0)NIINH2, -C(0)NH-CI-3alkylene-NH(R1 ), -C(0)CH3,
-C1-3 alkylene-NHC(0)01411, -C1-3 alkylene-NHC(0)141 , -C1-3 alkylene-
NHC(0)NHRto, -Ct-3a1ky1ene-NHC(0)-C 1-3 alkylene-1410, and a 3- to 12-membered

heterocycle optionally substituted with one or more substituents independently
selected
from R12;
Rm is independently selected at each occurrence from hydrogen, -NH2,
69
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-C(0)0CH2C6H5; and Ci-io alkyl, C2-io alkenyl, Ca-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S,
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, -Ci-io alkyl, -Ci-to haloalkyl, -0-C i-to
alkyl,
5 C2-m alkenyl, C2-10 alkynyl, C3-12 carbocycle, and 3- to 12-membered
heterocycle;
R" is selected from C3-12 carbocycle and 3- to 12-membered heterocycle, each
of which is optionally substituted with one or more substituents independently
selected
from R12; and
R12 is independently selected at each occurrence from halogen, -0R1 ,
-N(R1 )2, -C(0)R10, -C(0)N(R)2,
-N(R.1 )C(Ort , -C K
(0)0R10, _oc(0oR10, _s(o)RlO,
_s(0)2n _
P(0)(0R1 )2, -0P(0)(01V0)2, -NO2, =0, =S, =N(R1 ), and -CN; Ci-io alkyl,
C2-10 alkenyl, C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -OR',
-N(R1 )2, -C(0)R1 ,
2 -C(0)N(R1 0)µ, _ N(R1 )C(0)1t1 , -C(0)0R1 , -0C(0)11.1 , -S(0)R1 , -S(0)210
,
15 -P(0)(0R1 )2, -0P(0)(01:0 )2, -NO2, =0, ,s, _N(Rio), -CN, C3-10
carbocycle and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-OR',
am, _mRich2 _
), C(0)100, -C(0)N(R10)2, - N(R1 )C(0)101, -C(0)0R1 , -0C(0)R10

,
-S(0)R1 , -S(0)2Rio,
P(0)(01;012, -0P(0)(0111 )2, -NO2, =0, =S, =N(R10), -CN, C1-6
alkyl, C2-6 alkenyl, and C2-6 alkynyl; and
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from R12 or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle.
25
101961 In some embodiments, the compound of Formula
(IIIA) is represented
by Formula (MB):
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
R1
R2
N¨R2
R25
n. 42
R21
1-2-R4
R22 R-9-
,1
(IIIB)
or a pharmaceutically acceptable salt thereof, wherein:
it20, 1(21, lc n22,
and R23 are independently selected from hydrogen, halogen,
-0R' , -SR10, -N(R1 )2, -S(0)R' , -S(0)21e , -C(0)R' , -C(0)011.1 , -0C(0)R' ,
-NO2,
5 =S, =N(R1 ), -CN, Ct-to alkyl, C2-10 alkenyl, and C2-10 alkynyl; and
R24 and R25 are independently selected from hydrogen, halogen, -OR', -SRI',
-N(Rn2, -S(0)11.1 , -S(0)2R1 , -Maw, -C(0)0R1 , -0C(0)11.1 , -NO2, =0, =S,
=N(10), -CN, Ci-to alkyl, C2-to alkenyl, and C2-10 alkynyl; or R24 and R25
taken together
form an optionally substituted saturated C3-7 carbocycle.
10 [0197] In some embodiments, R20

., R21, R22, and R23 are independently selected
from hydrogen, halogen, -OH, -NO2, -CN, and Ci-io alkyl. In certain
embodiments, R20,
R2i, Kn22,
and R23 are each hydrogen. In some embodiments, RN and R25 are
independently selected from hydrogen, halogen, -OH, -NO2, -CN, and C1-10
alkyl, or R24
and R25 taken together form an optionally substituted saturated C3-7
carbocycle. In
15 certain embodiments, R24 and R25 are each hydrogen. In certain
embodiments, R24 and
R25 taken together form an optionally substituted saturated C3-5 carbocycle.
[0198] In some embodiments, R1 is hydrogen. In some embodiments, R2 is
hydrogen.
[0199] In some embodiments, L" is selected from ¨C(0)N(R1 )- *. In some
20 embodiments, 12.1 of -C(0)N(Rw)-* is selected from hydrogen and C1-6
alkyl. For
example, L11 may be ¨C(0)NH-t.
[0200] In some embodiments, 12.6 is phenyl substituted with R7 and R6 is
further
optionally substituted with one or more additional substituents independently
selected
from R12. In some embodiments, 11.6 is selected from phenyl substituted with
one or
25 more substituents independently selected from -C(0)NI-INH2, -C(0)NH-C1-3
alkylene-
mwo%
) C 1-3 alkylene-NHC(0)R1 , and -C(0)CH3; and
3- to 12-membered
71
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
heterocycle, which is optionally substituted with one or more substituents
selected from
¨OH, -N(R10)2, ¨NHC(0)(Rt ), -NHC(0)0(R1 ), -NHC(0)N(R10)2, -C(0)R10

,
-C(0)N(R1 )2, -C(0)2R1 , and -CL-3 alkyleneaw) and R6 is further optionally
substituted with one or more additional substituents independently selected
from Rn.
5 For example, le may be selected from:
HO
H2NHN HOLD 40)
Si
0
0
H3CA-0 0 0
H3C 110
H3C
meo2c.
0
H 140 H 4111 CbzHNN
1/11 0
,CO2Me
140 H
Olt
0 , and =
10 102011 In some embodiments, 11.6 is selected from a 5- and 6-
membered
heteroaryl substituted with one or more substituents independently selected
from R7,
and R6 is further optionally substituted with one or more additional
substituents selected
from 102. In certain embodiments, R6 is selected from 5- and 6-membered
heteroaryl
substituted with one or more substituents independently selected from -
C(0)CH3, -C1-3
15 alkylene-NHC(0)0Rt , -CI-3 alkylene-NHC(0)Rt , -CI-3 alkylene-NHC(0)NHR10,
and
-C1-3 alkylene-NHC(0) -C1-3 alkylene-(R' ); and 3- to 12-membered heterocycle,
which
is optionally substituted with one or more substituents selected from ¨OH, -
N(R1 )2,
,
¨NHC(0),(Rio), NHC(0)0(R1 ), -NHC(0)N(R10)2, -C(0)R' , -C(0)N(RE )2,
-C(0)2R' , and -C1-3 allcylene-(R' ), and PP is optionally further substituted
with one or
20 more additional substituents independently selected from R12. R6 may be
selected from
72
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
substituted pyridine, pyrazine, pyrimidine, pyridazine, furan, pyran, oxazole,
thiazole,
imidazole, pyrazole, oxadiazole, oxathiazole, and triazole, and R6 is
optionally further
substituted with one or more additional substituents independently selected
from R12.
In some embodiments, le is substituted pyridine and re is optionally further
substituted
with one or more additional substituents independently selected from Rn. 116
may be
R7 N
.....N1
70.1
represented as follows: R or N-LIMI
. In some embodiments, R6 is
substituted pyridine, and wherein 117 is -CI-3 alkylene-NHC(0)-Ci-3 alkylene-
r. In
certain embodiments, le is -Ci alkylene-NHC(0)-Ci alkylene-10 . In certain
embodiments, R7 is -Ci alkylene-NHC(0)-Ci alkylene-NH2. In some embodiments,
R6
4 N oiN *
H H H N
N.......,.N......Xj.,"
I I
is selected from: 0 , cp
,
4111 N
N
H %%a 4 NH He...,..ni
N 4%....
N
N
H
0 0
N N
00) NH2 H .......ni 4 NH2 H .fil
2 N --...
N N.,
0 0
N
....14
--- ,
iio aior NHõ..L1, IL 411
ri.....õ0.1 CbzHN -'....tliN
I
-....
NH 0
H2N
il
Cbzhl , 0 JON N
0 .o.N
-laN H2N i i
iji
\ I
0
0
oxiiN
N
H3Cit,.... CjI
õ.õ..,-..NACINUI
I
\
H2N H HN
N
OS
H2XH..,._ I H
,.._
.ir N."--"Lie)-)ss C bz H 4-Ntire N...."---"9-11/4" ebzHN
0 0
0
73
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Cbz 0 , and 0
. In certain embodiments, le is
Jcio,N
H2N-Thr
0
[0202] In some embodiments, L2 is selected from -C(0)-, and -C(0)NR1 -. In
some embodiments, L2 is selected from -C(0)NRio_. Rio of _c(o)NRio_ may be
5 selected from hydrogen and Cho alkyl. For example, 12 may be -C(0)NH-. In
some
embodiments, L2 is -C(0)-.
[0203] In some embodiments, 11.4 is selected from: -OR', -N(R1 )2,
-C(0)N(R10)2, -C(0)R1 , -C(0)011.1 , -S(0)R' , and -S(0)2R1'; Ci-io alkyl, C2-
10 alkenyl,
C2-10 alkynyl, each of which is optionally substituted with one or more
substituents
10 independently selected from halogen, -0R' , -SRn, -C(0)N(Rn)2,
_N(Rio)c(0)R10

,
-N(Rt )C(C)N(Rm)2, - (), FT Rio,2 _
C(0)R1 , -C(0)0R' , -0C(0)R1 , -NO2,
=S,
=N(R1 ), -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12
carbocycle and 3- to 12-membered, each of which is optionally substituted with
one or
more substituents independently selected from halogen, -Ole , -SRI , -
C(0)N(RI0)2,
15 -N(Rio)C(or _
N(Rm)C(0)N(Rw)2, _maio)2, _c(0)Rio, -0(0)01?-m, -0C(0)R10, -
NO2, 0,=S, =N(Rw), -CN, Ci-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl_ In some
embodiments, 10 is selected from: -OR' and -N(RE )2; and Ci-io alkyl, C2-10
alkenyl, C2-
alkynyl, C3-12 carbocycle and 3- to 12-membered heterocycle, each of which is
independently optionally substituted at each occurrence with one or more
substituents
20 selected from halogen, -010, -N(RE )2, -S(0)11"), -S(0)2R10
_c(D)Rio, _
C(0)0R' , -0C(0)R1 , -NO2, =0, =S, =N(R1 ), -CN, Ci-io alkyl, C2-10 alkenyl,
and C2-10
alkynyl. In certain embodiments, R4 is _Is(R10)2 R10 of _N(tc.. n-- io
)2 may be independently
selected at each occurrence from optionally substituted Ci-6 alkyl. In some
embodiments, RI of -N(R10)2 is independently selected at each occurrence from
methyl,
25 ethyl, propyl, and butyl, any of which are optionally substituted. For
example, R4 may
('CH3
(CH3
EVe
ArNI
0
be CH3 In some embodiments, -L2-le is
CH3
74
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
102041 In some embodiments, R12 is independently selected at each occurrence
from halogen, -ORth, -SR1 , -N(R10)2, _coy.n -C(0)N(R10)2,-N(Rt )C(0)R1 ,
-C(0)0R' , -0C(0)R1 , -S(0)R1 , -S(0)2R1 , -P(0)(0R1 )2, -0P(0)(0R1 )2, -NO2,
=0,
=S, =N(R1 ), and -CN; Ct-to alkyl, C2-10 alkenyl, C2-to alkynyl, each of which
is
5 independently optionally substituted at each occurrence with one or more
substituents
selected from halogen, -01e , -S11.1 , -N(11.1 )2, -C(0)11.10, -C(0)N(R1 )2,
-N(R1 )C(0)R1 , -C(0)0R10, -0C(0)R' , -S(0)R1 , -S(0)2R10, -P(0)(0R10)2,
-0P(0)(ORP)2, -NO2, =0, =S, =N(Rw), -CN, C3-to carbocycle and 3- to 10-
membered
heterocycle; and C3-10 carbocycle and 3- to 10-membered heterocycle, each of
which is
10 independently optionally substituted at each occurrence with one or more
substituents
selected from halogen, -OR', = -N(I011)2, -
C(0)R' , -C(0)N(R1 )2,
-N(R1 )C(0)R1 , -C(0)0R10, -0C(0)R' , -S(0)R1 , -S(0)2R10, -P(0)(01120)2,
-0P(0)(0R1 )2, -NO2, =0, =S, =N(Rw), -CN, Ct-6 alkyl, C2-6 alkenyl, and C2-6
allcynyl.
In certain embodiments, R12 is independently selected at each occurrence
15 from halogen, -0R1 , -SR1 , -N(R1 )2, -C(0)12.1 , -C(0)N(Rm)2, -N(R1
)C(0)R1 , -C(0)0
R10, -0C(0)10 , -S(0)10 , -S(0)2R' , _13(0)(0R1 )2, -0P(0)(01tw)2, -NO2, =0,
=S,
=N(11.10), and -CN; Ct-to alkyl, C2-to alkenyl, C2-loalkynyl, each of which is

independently optionally substituted at each occurrence with one or more
substituents
selected from halogen, -0R1 , = -N(R1 )2, -
C(0)Rw, -C(0)N(R1 )2, -N(100)C(0)R1
20 0, -C(0)0R'0, -0C(0)11.1 , -S(0)111 , -S(0)2111 , -P(0)(013.1 )2, -
0P(0)(0R1 )2, -NO2,
=0, =S, =N(R1 ), -CN, C3-10 carbocycle and 3- to 10-membered heterocycle.
102051 In some embodiments, the compound is selected from:
HO
CN
H S0 NH2
NI-I2
2NHN
0 *
*
--
0 issTh
0 'NM
5
HOo Si
0
NH2
N,
ags
0
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
Cbz,,., CIN N
r, --iu 0
NH2
....... i
11,_
iH AO- r---\
N
0 \---\
,
H2N...ON N
y ) 0
Na._. NH2
H
N
0
H2N
ON N
0
NH2
N
ril *
N
0 \----\
,
0
H2N __ter, N
HACIN1 N
n 0
NH2
N,
N (00
H
--- Nr-\
0
.....N
0.,%.1 0
NH2
0

VI *HN
....- rm,
N
0 \---\
,
0
H3CAC1
N . 0
NH2
N
N 0
-- r,
N
,
76
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
40 N 0 NH2
H3C N,
H so0
NC\....õ
0 z,
,
0
CbzHN..õ.õ...-,N 0
0
H N, NH2
N
H
---- Nr- \
0 Z,
0 NH2
HN
N,
*
--- C\
N
CbzHNI"---NEI *
0 \Th
0
,
* H
N 0
o N..... NH2
N
H
N
0
4 H H j xN N
NH2
N......e.N "-... N.,
I I
*
H
raj
N
5
0
le ....N N
0
Ic....õ..c.....1,N * N, NH2
H H
0
N
0 \Th
,
N
*
NH H ..n 0
NI-12
N
N
H
N
0
\---\ 5
*.
H %........a NH2
N
H
0
--- r-1
N
0
77
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
N
4 NH2 H ri- ) 0
NH2
N.,.....&.õ..A..N
N,
H
--- ri
0
N
N
H re1/4.n1
N H2
0... N .õic. ...............A . I ,N
0 N
*,
H
8
N
0 \---\
,
1101 N
H j lei
N -,..õ
N
N, NH2
NH2 0 H
N
,
CbzHN-...---till 0
NH2
--õ,.
K.,
N *
H
--- Nr\
ihuuN 0
NH2
N___
110 0
N *
N
0
to
H3c 40 0
NH2
N.....
M *
N
0
H CAC: )1 1
3 0
NH2
N,
M SI
N
78
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
ri.., IN 0
NH2
N
CbzHIXt N
H
0
--- CI\
N
0 \----\
,
JuN 0
NH2
N
H2 ...N)Cir N
H
0
11101
N/Th
,
0 0
NH2
N
CbzHN N
H
N
N
N
H
NH2
0
N
-_.
H
Cbz 0
/Th
N
CO2 Me
%.;
N
Me02C. 40 0 N NH2
0 NH2 N
--.
It
le N ,
H
H
N N --- /--\
--- /Th
N
N
N
tx
H
0 NH2
H2N....õ..T.N \
0 H I

--- re-\
N
, and 0 \-
1\ and a salt of any one thereof.
[0206] In some aspects, the present disclosure provides a compound represented

by the structure of Formula (IA):
79
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
R1
1
N-R2
N,
L1
R4
R3
L2--
(IA)
or a pharmaceutically acceptable salt thereof, wherein:
- represents an optional double bond;
12 is selected from -XI-, -X2-C1-6 alkylene-X2-Ci-6 alkylene-, -X2-C2-6
5 alkenylene-X2-, and -X2-C2-6 alkynylene-X2-, each of which is optionally
substituted on
alkylene, alkenylene or alkynylene with one or more RP,
L2 is selected from -X2-, -X2-C 1-6 alkylene-X2-, -X2-C2-6 alkenylene-X2-, and

-X2-C2-e alkynylene-X2-, each of which is optionally substituted on alkylene,
alkenylene or alkynylene with one or more Rt2;
10 r is selected from -S-*, -N(Rm)-*, -C(0)0-*, -0C(0)-*, -0C(0)0-*, -

C(0)N(R1 )C(0)- *, -C(0)N(R1 )C(0)N(Rw)*, -N(RnC(0)-*, -CRt 2N(R1 )C (0)-*,
-N(1V )C(0)N(R1 )-*, -N(1V )C(0)0-*, -0C(0)N(R15-*, -C(NR1 )-*,
-N(Rt )C(NRI )-*, -C(N10 )N(R1 )-*, -N(R1 )C(NR1 )N(Rn-*, -S(0)2-*, -0S(0)-*,
-S(0)0-*, -S(0), -0S(0)2-*, -S(0)20*, -N(Rw)S(0)2-*, -S(0)2N(R1 )-*,
15 -N(RnS(0)-*, -S(0)N(R1 )-*, -N(Rm)S(0)2N(R1 )-*, and -N(10 )S(0)N(R1 )-
*,
wherein * represents where X1 is bound to R3;
X2 is independently selected at each occurrence from -0-, -S-, -N(101)-, -C(0)-
,
-C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(100)-, -C(0)N(Rt )C(0)-,
-C(0)N(R1 )C(0)N(R1 ), -N(R1 )C(0)-, -N(R1 )C(0)N(R1 )-, -N(Rt )C(0)0-,
20 -0C(0)N(Rm)-, -C(NRw)-, -N(R1 )C(NR1 )-, -C(N11.1 )N(Rm)-, -N(10 )C(NR1
)N(Rm)-
, -S(0)2-, -0S(0)-, -S(0)0-, -S(0), -OS(0)2-, -S(0)20, -N(Rw)S(0)2-, -
S(0)2N(111 )-
, -N(Rt )S(0)-, -S(0)N(11.11-, -N(11.1 )S(0)2N(R1 )-, and -N(Rw)S(0)N(Rw)-;
R1 and R2 are independently selected from hydrogen; Ci-io alkyl, C2-10
alkenyl,
and C2-10 alkynyl, each of which is optionally substituted with one or more
substituents
25 independently selected from halogen, -01e , -SRt , -C(0)N(R10)2, -
N(Rm)2, -S(0)10 ,
-S(0)212-1 , -C(0)10 , -C(0)0R1 , -0C(0)R' , -NO2, =0, =S, =N(R1 ), and -CN;
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
IV is selected from optionally substituted C3-12 carbocycle, and optionally
substituted 3- to 12-membered heterocycle, wherein substituents on R3 are
independently selected at each occurrence from: halogen, -OW , -SW , -C(0)N(R1
)2,
-N(RP)C(0)R' , -N(R40)C(0)N(R1)2, -N(R112, -C(0)11P, -C(0)01e , -00potio,
5 -Nth, =0, =S, =N(101, and -CN; Ct-to alkyl, C2-10 alkenyl, C2-toalkynyl,
each of which
is optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SW , -C(0)N(R40)2, -N(Rw)C(0)RP, -N(LOC(0)N(R10)2, -N(R1 )2,
-C(0)R1 , -C(0)0R1 , -0C(0)11P, -NO2, =0, =S, =N(R1 ), -CN, C3-12 carbocycle,
and
3-to 12-membered heterocycle; and C3-12 carbocycle, and 3-to 12-membered
10 heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered
heterocycle in 112 is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SW , -C(0)N(R40)2, -N(Rw)C(0)R4 , -N(100)C(0)N(R10)2, -
N(R10)2,
-C(0)R1 , -C(0)0R1 , -0C(0)11P, -NO2, =0, =S, =N(R1 ), -CN, Ct-is alkyl, C2-6
alkenyl,
and C2-6alkynyl;
15 le is selected from: -01V , -N(W12, -C(0)N(R10)2,
-C(0)0R1 ,
-S(0)R' , and -S(0)2R1 ; Ci-to alkyl, C2-10 alkenyl, C2-loalkynyl, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, -OW , -SW , -C(0)N(R40)2, -N(11.1 )C(0)Rw, -N(R1 )C(0)N(R10)2, -
N(R10)2,
-C(0)10- , -C(0)0R10, -0C(0)R4 , -NO2, =0, =S, =N(R1"), -CN, C3-12 carbocycle,
and
20 3-to 12-membered heterocycle; and C3-12 carbocycle, and 3-to 12-membered
heterocycle, wherein each C3-12 carbocycle, and 3- to 12-membered heterocycle
in R4 is
optionally substituted with one or more substituents independently selected
from
halogen, -OR', -SW , -C(0)N(R4 )2, -N(R1 )C(0)Rm, -N(R1 )C(0)N(R10)2, -
N(1110)2, -
C(0)R1 , -C(0)0R4 , -0C(0)R4 , -NO2, =0, =S, =N(R10), -CN, C1-6 alkyl, C2-
6alkenyl,
25 and C2-6alkynyl;
Itm is independently selected at each occurrence from:
hydrogen, -NH2, -C(0)0CH2C61-15; and Ci-to alkyl, C2-to alkenyl, C2-to
alkynyl, C3-12
carbocycle, and 3- to 12-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -CN, -NO2, -
NI-12,
30
=S, -C(0)0CH2C6H5, -NHC(0)0CH2C6H5, C1-10alkyl,
C2-to alkenyl, C2-toalkynyl,
C3-12 carbocycle, 3- to 12-membered heterocycle, and haloalkyl; and
81
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
R12 is independently selected at each occurrence from halogen, -OR", -SR",
-N(R")2, -C(0)R1 , -C(0)N(R10)2, -N(Rm)C(0)1V- , -C(0)0R", -0C(0)R10, -
S(0)11P,
-S(0)21e , -P(0)(OR")2, -0P(0)(011.1 )2, -NO2, =0, =S, =N(R1 ), and -CN; C'4)
alkyl,
C2-10 alkenyl, C2-10 alkynyl, each of which is optionally substituted with one
or more
5 substituents independently selected from halogen, -OR10, _SR", _N(RE0)2, -
C(0)R' ,
-C(0)N(Rio)2, _mato)coynio,
-C(0)0R", -0C(0)R", -s(o)Rio., _s(0)2Rto,
-P(0)(0R10)2, -0P(0)(0R10)2, -NO2, =0, =S, =N(11"), -CN, C3-10 carbocycle and
3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in 111-2 is
optionally
10 substituted with one or more substituents independently selected from
halogen, -OR",
_swop, _N(Rio)2, _icor to, _
K C(0)N(Rio)2,
N(Rio)cor to, _
K
C(0)0R", -0C(0)R",
-S(0)R", -S(0)2R'

,
P(:WOR")2, -0P(0X0R10)2, -NO2, =0, =S, =N(W-1), -CN, C1-6
alkyl, C2-6 alkenyl, and C2-6 alkynyl; and
wherein any substitutable carbon on the benzazepine core is optionally
15 substituted by a substituent independently selected from RI' or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle
102071 In some embodiments, the compound of Formula (IA) is represented by
Formula OB):
Ri
R2
N-R2
R25
R24
R21
1-2-R4
R22 R-=>-
n
(1B)
20 or a pharmaceutically acceptable salt thereof, wherein:
R20, R21, R22, and n n 23
are independently selected from hydrogen, halogen,
-OR", -sinK _ 10, WRILe)2, -S(0)R1 , -S(0)2R' , -C(0)R' , -C(0)0R1 , -0C(0)R1
, -NO2,
=S, =N(Itn, -CN, Cr-to alkyl, C2-10 alkenyl, and C2-to alkynyl; and
R24 and K-25
are independently selected from hydrogen, halogen, -OR', -SR",
25 -N(W1)2, -S(0)Rm, -S(0)2R", -C(0)R", -C(0)0R1 , -0C(0)RP, -NO2, =0, =S,
82
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
=Noce), -CN, C1-to alkyl, C2-to alkenyl, and C2-to alkynyl; or R24 and 1125
taken together
form an optionally substituted saturated C3-7 carbocycle.
[0208] In some embodiments, R20, R21, R22, and ts. r. 23
are independently selected
from hydrogen, halogen, -OH, -NO2, -CN, and Ci-to alkyl. In certain
embodiments, 1120

,
5 1121, 11 22,
and 1123 are each hydrogen.
[0209] In some embodiments, R24 and R25 are independently selected from
hydrogen, halogen, -OH, -NO2, -CN, and Ci-to alkyl, or R24 and 1125 taken
together form
an optionally substituted saturated C3-7 carbocycle. In some embodiments, R24
and 1125
are each hydrogen. In some embodiments, R24 and 1125 taken together form an
10 optionally substituted saturated C3-5 carbocycle.
[0210] In some embodiments, R1 is hydrogen. In some embodiments, R2 is
hydrogen.
[0211] In some embodiments, is selected from -N(RO)C(0)-*, -S(0)2N(Rn-
*, _cRio2N(Rto)c
*and -X2-C1-6 alkylene-X2-Cl-6 alk-ylene-. In some embodiments,
15 1.2 is selected from -N(R1 )C(0)-*. In certain embodiments, le of -N(R1
)C(0)-* is
selected from hydrogen and C1-6 alkyl. For example, may be -N1HC(0)-*. In some

embodiments, 1,1 is selected from -S(0)2N(Rm)-*. In certain embodiments, Rio
of
-S(0)2N(R1 )-* is selected from hydrogen and C1-6 alkyl. For example, I2 is -
S(0)2N1-I-
*. In some embodiments, 12 is -CR1 2N(R10)C(0)-*. In certain embodiments, 1,1
is
20 selected from -CH2N(H)C(0)-t and -CH(CH3)N(H)C(0)-4.
[0212] In some embodiments, 10 is selected from optionally substituted
C3-12 carbocycle, and optionally substituted 3- to 12-membered heterocycle,
wherein
substituents on R3 are independently selected at each occurrence from:
halogen, -OR',
-C(0)N(R1 ) _ N(R1 )C(0)10 , -N(R1 )C(0)N(R10)2,
2
_N(R10ftµ, _ C(0)R1 ,
25 -C(0)0R1 , -0C(0)R' , -NO2, =0, ¨S, ¨114(R10), and -CN; Ci-to alkyl, C2-
10 alkenyl, C2-
alkynyl, each of which is optionally substituted with one or more substituents
independently selected from halogen, -0R10,
_c(o)N(Rto)2, _NR10),c(o)Rio,
-N(12.1 )C(0)N(R 1)2, - ) _ C(0)R1 , -
C(0)01011, -0C(0)100, -NO2, =S,
=N(R1 ), -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12
30 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted
with one or more substituents independently selected from halogen, -OR', -
Sltm,
83
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-C(0)N(Rlo)2, _
in Notic(0)Rto, _N(R1
)µ..`"(0)N(Rw)2, -N(Rw)2, -C(0)11.10, -C(0)0R' , -
0C(0)R' , -NO2, =0, =S, =N(Rw), -CN, C1-6 alkyl, C2-6 alkenyl, and C2-6
alkynyl. In
certain embodiments, R3 is selected from optionally substituted C3-12
carbocycle, and
optionally substituted 3- to 12-membered heterocycle, wherein substituents on
R3 are
5 independently selected at each occurrence from: halogen, 0R' ,-
_snK, _ 10 C(0)N(Rw)2, -
mitio)c(o)Rto, _mg.to)c onRio)2, _r,4(Rio)2,
-C(0)R' ,

- C(0)0R1 , -0C(0)R1 , -
NO2, =S, =N(Rw), and -CN; Ct-to alkyl, C2-10
alkenyl, C2-10 alkynyl, each of which
is optionally substituted with one or more substituents independently selected
from
halogen, -OR', _SRI , _C(0)N(R10)2, -N(lut)c(c)Rio, _mat )µ...7(0)N(11.192, -
N(R10)2, -
10 C(0)R' , -C(0)0R' , -0C(0)R' , -NO2, =0, =S, -N(Rw), -CN, C3-12
carbocycle, and 3-
to 12-membered heterocycle.
[0213] In some embodiments, R3 is selected from an optionally substituted aryl

and an optionally substituted heteroaryl. In some embodiments, R3 is an
optionally
substituted heteroaryl. R3 may be an optionally substituted heteroaryl
substituted with
15 one or more substituents independently selected from halogen, -ORto, am,
_N(Rio)2, _
C(0)10), -C(0)0R' , -0C(0)R' , -NO2, =0, =S, -CN, C1-6 alkyl, C2-6 alkenyl,
and C2-6
alkynyl. In certain embodiments, R3 is selected from an optionally substituted
6-
membered heteroaryl. For example, R3 may be an optionally substituted
pyridine. In
some embodiments, R3 is an optionally substituted aryl. In certain
embodiments, R3 is
20 an optionally substituted aryl substituted with one or more substituents
independently
selected from halogen, -ORIO, _SR10, _NR10)2, _c(or _ 10,
K C(0)0R1 , -0C(0)R' , -NO2,
=0, =S, -CN, C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl. R3 may be an
optionally
substituted phenyl. In certain embodiments, R3 is selected from pyridine,
phenyl,
tetrahydronaphthalene, tetrahydroquinoline, tetrahydroisoquinoline, indane,
25 cyclopropylbenzene, cyclopentapytidine, and dihydrobenzoxaborole, any
one of which
oisN Sop
is optionally substituted. R3 may be selected from:
I-1
4111 HN 40
HN
.4111
84
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
HO
oat N
B
I col se . 0 NA 6
OP
H
/4
, any one of which is optionally substituted. For example, IV may be
selected from:
0
OH
N
,
cl 0 0 0 all
el
0
OH
H
N,
HN 40)
Iii
filli
HN 410 N
5 , , , H
0
N
A 101 011n CAN
.--....... 1
III
CbzeN le NE-I2
* ,
1
g )011 N
I
c Ø1 N
cal 40 411
= 411
NHCbz , H2N CbzHN H2N CbzHN
,
, .
HOlo
HOCIN oi
N 0
0 N .µ H2NHN le
0
0
N 0
0
--)HON le (-Tiy
lir CbzHN---.N 0
H
CH3
s
s s
N
N
H
H H
N .,_____ -=-Ø. IMI
lie N.,,e, N.,..X.)../"....
C bzHN ---..' I:1 11. 411
I I
10 0 0
0
,
, ,
SI N _N
NH
N
H
0
0
CA 03151322 2022-3-15

WO 2021/067644
PC T/US2020/053870
NH2
0
N 1111 1-12 H .....X.11 ilin
-z N "...,
Irl jt,.*ey 1t
0
0
N '
N
,
N
H N H2
il,,,,,a 01
11,,Jai
1101 8 0
NH2 0
,
0
N
N......niess
H
H2Neet.MN 1 CbzHN --11 1
I -Atli CbzHN-
ItrN ...--- I ...õ. 1 %,.. -.....
0
,
,
IS tuai t.,0,,es,,N
II
k4e02c.
4111
CbzHN 0
N õ
,CO2Me
N
N
i
ii# . ili
Cbz 0
HO,B HR C bZ
i
HO
B N H
v
6 411 01 SI
0/13 40
5 , and CbzHN
.
102141 In some embodiments, L2 is selected from -C(0)-, and -C(0)NRio_. In
certain embodiments, L2 is -C(0)-. In certain embodiments, L2 is selected from
-C(0)NRio_. RID of _c(0)NR10_ may be selected from hydrogen and Ci-6 alkyl.
For
example, L2 may be -C(0)NH-.
10
102151 In some embodiments, R4 is selected from: -0R10, -
N(Rw)2,
-C(0)N(10)2, -C(0)R' , -C(0)0100, -S(0)R' , and -S(0)210; Ci-io alkyl, C2-10
alkenyl,
C2-10alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -01tw, -SRM, _coysst(t10)2,
_N(R1th)..."(0)R1 ,
-N(R1 )C(C)N(RECI)2, -N(R10)2, -C(0)R10, -C(0)0R1 , -0C(0)R10, -NO2, =0, =S,
15 =N(Rw), -CN, C3-I2 carbocycle, and 3-to 12-membered heterocycle; and C3-
12
carbocycle, and 3- to 12-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -0R' , -
Sltw,
-C(0)N(Rl0)2, _N(Rio)c(0)Rio, _
N(R1 )C(0)N(R1 )2, 'T kn..'" J-N4 1 )2, -C(0)R' , C(0)0R1 ,
-0C(0)R' , -NO2, =0, =S, =N(Rw), -CN, CI-6 alkyl, C2-6 alkenyl, and C2-6
alkynyl.
20
102161 In some embodiments, 114 is selected from: -OW , -
N(Rw)2,
n io,
-C(0)N(R10)2, -C(0)R1 , -C(0)0k -S(0)R' , and -S(0)2R' ; Ci-io alkyl, C2-10
alkenyl,
86
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
C2-10alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R1 , _sin to,
ts. -C(0)N(Rm)2, -N(R1 )C(0)R1 ,
-N(R1 )C(0)N(R113)2,

)
_ C(0)R1 , -C(0)0R1 , -0C(0)R1 , -NO2,
=0, =S,
=N(10), -CN, C3-12 carbocycle, and 3- to 12-membered heterocycle. In some
5 embodiments, 11.4 is selected from: -Ole , and -N(Rm)2; and Ci-io alkyl,
C2-10 alkenyl,
C2-10alkynyl, C3-12 carbocycle, and 3- to 12-membered heterocycle, each of
which is
optionally substituted with one or more substituents independently selected
from
halogen, _caw, _SRI ,

) _ S(0)10 , -
S(0)210, -C(0)R1 , -C(0)010 ,
-0C(0)R' , -NO2, =0, =S, =N(11.11), -CN, Ct-to alkyl, C2-10 alkenyl, and C2-10
alkynyl.
10 In certain embodiments, R4

is -MR10)2. tic -=-= 10
of -N(Rm)2 may be independently selected
at each occurrence from optionally substituted C1-6 alkyl. In certain
embodiments, IV
of-N(R10)2 is independently selected at each occurrence from methyl, ethyl,
propyl, and
CCH3
butyl, any one of which is optionally substituted. For example, 10 may be
CH3.
cscr.N1,1
0 1,
In certain embodiments, L2-R4 is CH3.
15 102171 In some embodiments, R12 is independently selected at each
occurrence
from halogen, -OR10, _SRN, _N(R10)2, _garb 10, _
tr.
C(0)N(Rm)2, -N(11.1 )C(0)R1 , -
C(0) R' , _Oc(0)Rio, _spy. io, _
S(0)2Rm, -P(0)(0R10)2, -0P(0)(0R10)2, -NO2, =0,
=S, =N(Rm), and -CN; Ct-io allcyl, C2-10 alkenyl, C2-w alkynyl, each of which
is
optionally substituted with one or more substituents independently selected
from
20 halogen, -0R' , 2
_N(Rto,), _ C(0)R1 , -C(0)N(R10)2, _We:W(0)R', -C(0)011.1 , -
0C(0)11.1 , -S(0)R10, -S(0)2R10, -P(0)(0R10)2, -0P(0)(0R10)2, -NO2, =0, =s,
=N(Rio),
-CN, C3-10 carbocycle and 3- to 10-membered heterocycle; and C3-10 carbocycle
and 3-
to 10-membered heterocycle, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -0R10, _SRM, - N(R112, -
C(0)11.1 ,
25 -C(0)N(Rio,
) N(R1 )C(0)R1 ,-C(0)0R1 , -0C(0)R1 , -S(0)11.1 , -S(0)211.10

,
-P(0)(0R1 )2, -0P(0)(01k10)2, -NO2, =0, =S, =N(R1 ), -CN, Ci-s alkyl, C2-6
alkenyl,
87
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
C2-6 alkynyl. In some embodiments, R" is independently selected at each
occurrence
from halogen, -ORm, -SRm, -N(R10)2, _c(o)Rto, _c(0)N(R10)2, _Nottonowto, _
C(0)0R1 , -0C(0)R1 , -S(0)1e, -S(0)2R' , -P(0)(0R1 )2, -0P(0)(011.1 )2, -NO2,
=0,
=S, =N(111 ), and -CN; Ct-to alkyl, C2-10 alkenyl, C2-to alkynyl, each of
which is
5 optionally substituted with one or more substituents independently
selected from
halogen, -OR'", _N(t to)2, -C(0)11-10, -
C(0)N(R10)2, -N(R.1 7)C(0)r-C(0)011."), -
0C(0)11.1 , -S(0)R1 , -S(0)2100, -11(0)(01;t10)2, -0P(0)(010 )2., -NO2, =0,
=S, =N(11.1 ),
-CN, C3-io carbocycle and 3- to 10-membered heterocycle.
102181 In some embodiments, the compound is selected from:
0
NH2
I-1
cy N, NH:
110
11 1111
0
r-\
0
10 0
,and
a salt of any one thereof.
102191 In some aspects, the present disclosure provides a compound represented
by the structure of Formula (IVA):
N¨R2
L12
R4
Ra
(IVA)
15 or a pharmaceutically acceptable salt thereof, wherein:
¨ represents an optional double bond;
L12 is selected from -30-, -X3-C1-6
-V-C2-6a11eny1ene-X3-, and
alkynylene-V-, each of which is optionally substituted on alkylene,
alkenylene, or allcynylene with one or more substituents independently
selected from
20 Rt2;
L22 is independently selected from -X4-, -X4-C1-6 alkylene-r-, -r-C2-6
alkenylene-r-, and -r-C2-6 alkynylene-r-, each of which is optionally
substituted on
88
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
alkylene, alkenylene, or alkynylene with one or more substituents
independently
selected from Rix%
X3 and X4 are independently selected at each occurrence from a bond, -0-, -S-,
-N(R1 )-, -C(0)-, -C(0)0-, -0C(0)-, -0C(0)0-, -C(0)N(RIc)_r,, _
C(0)N(R1 )C(0)-,
-C(0)N(R1 )C(0)N(R1 )-, -N(Rw)C(0)-, -N(R1 )C(0)N(R1 )-, -N(Rm)C(0)0-,
-0C(0)N(10)-, -gmtio)_, _mitio)c(NRKI)_, _gNRio)N(Rio)-,
-N(R1 )C(N10)N(Rm)-, -S(0)2-, -0S(0)-, -S(0)0-, -S(0)-, -OS(0)2-, -S(0)20-,
-N(Rm)S(0)2-, _
-S(0)2N(Rio,), _ N(RnS(0)-, -S(0)N(Rm)-, -N(10)S(0)2N(Rm)-, and
-N(R1 )S(0)N(R")-;
It1 and R2 are independently selected from 1,3, and hydrogen; and Ci-io alkyl,
C2-
10 alkenyl, and C2-10 alkynyl, each of which is optionally bound to 1_,3 and
each of which
is optionally substituted with one or more substituents independently selected
from
halogen, -OR', -S10, -C(0)N(Rm)2, -N(10)2, -S(0)10, -S(0)2R' , -C(0)R1 ,
-C(0)010, -0C(0)10, -NO2, =0, =S, =N(R"), and -CN;
le and le are independently selected from: -0R1 , -N(R1 )2, -C(0)N(10)2,
-C(0)R' , -C(0)0R", -S(0)R1 , and -S(0)2R' ; Ci-to alkyl, C2-10 alkenyl, C2-to
alkynyl,
each of which is optionally bound to I-3 and each of which is optionally
substituted with
one or more substituents independently selected from halogen, -OR", -SRm,
-C(0)N(R1 )2, -N(10)C(0)R1 , -N(10)C(0)N(10 )2, -N(R1 )2, -C(0)10, -C(0)010,
-0C(0)10, -NO2, =0, =S, =N(R1 ), -CN, C3-12 carbocycle, and 3- to 12-membered
heterocycle; and C3-12 carbocycle, and 3- to 12-membered heterocycle, wherein
each
C3-12 carbocycle, and 3- to 12-membered heterocycle in 14.4 and le is
optionally bound
to L3 and each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R1 , -SRI', -C(0)N(R1 )2, - N(R1
)C(0)10, -N(
10)C(0)N(R1 )2, -N(Rm)2, -C(0)Rm, -C(0)010 , -0C(0)11.10, -NO2, =0, =S, -
N(Rm),
-CN, C1-6alkyl, C2-6 alkenyl, and C2-6 alkynyl;
10 is independently selected at each occurrence from 1,3, hydrogen, -Nib,
-C(0)0C112C6H5; and Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NI-12, =0, =S,
89
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
-C(0)0CH2C6H5, -NHC(0)0CH2C6H5, C i-io alkyl, C2-io alkenyl, C2-to alkynyl, C3-
12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
L3 is a linker moiety, wherein there is at least one occurrence of L3; and
R12 is independently selected at each occurrence from halogen, -0R1 ,
-N(R1 )2, -C(0)R' ,
-C(0)N(R1 µ , - p N(R10)C(0)R1 , -C(0)0R' , _oc(D)Rio, _s(0)Rio,
)2n to, _
-S(0 K P(0)(0R"))2, -0P(0)(0R1)2, -NO2, =0, =S, =N(R1 ), and -CN; C1-10 alkyl,

C2-10 alkenyl, C2-10 alkynyl, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -OR'),

)
_ C(0)R1 ,
-C(0)N(R10)2, -N(RIG)C(0'fttc. 1 , -C(0)0R1 , -0C(0)11.1 , -S(0)11.1 , -
S(0)211.10

,
-P(0)(0R152, -013(0)(010 )2, -NO2, =0, =S, -N(11.1 ), -CN, C3-10 carbocycle
and 3- to
10-membered heterocycle; and C3-10 carbocycle and 3- to 10-membered
heterocycle,
wherein each C3-10 carbocycle and 3- to 10-membered heterocycle in R12 is
optionally
substituted with one or more substituents independently selected from halogen,
-0R1 ,
am, _
C(0)R1 , -C(0)N(R1 0-.)2 _, MR1 )C(0)R1 , -C(0)0R1 ., -0C(0)R1 ,
-S(0)11.1 , -S(0)2Rto, _
P(OX0111 )2, -0P(OX0R1 )2, -NO2, =0, =S, =N(11.1 ), -CN, C1-6
alkyl, C2-6 alkenyl, C24 alkynyl;
wherein any substitutable carbon on the benzazepine core is optionally
substituted by a substituent independently selected from R12 or two
substituents on a
single carbon atom combine to form a 3- to 7- membered carbocycle.
102201 In some embodiments, the compound of Formula (WA) is represented
by Formula (P/13):
Ri
R2o
N-R2
R8,L12 N'
R25
R24
R21
L22-R4
R22 R23
(IVB)
or a pharmaceutically acceptable salt thereof, wherein:
R20, R21, R22, and R23 are independently selected from hydrogen, halogen,
-OR', -sinK _ 10, MR113)2, -S(0)R1 , -S(0)2R10,
-C(0)0R1 , -0C(0)R1 , -NO2,
=S, =N(R1 ), -CN, Ct-to alkyl, C2-lo alkenyl, and C2-10 alkynyl; and
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
1&24, and R25 are independently selected from hydrogen, halogen, -OR', -SRm,
-N(R1e)2, -S(0)114- , -S(0)2R10, -C(0)R' , -C(0)011410, -0C(0)R' , -NO2, =0,
=S,
=N(Rio), _
CN, Ci-to alkyl, C2-to alkenyl, and C2-10 alkynyl; or R24 and R25 taken
together
form an optionally substituted saturated C3-7 carbocycle.
5 [0221] In some embodiments, le is L3. In some embodiments, 112 is
L3.
[0222] In some embodiments, 1_,12 is _c(o)mR10)_.
In some embodiments, Rrn
of -C(0)N(R1 )- is selected from hydrogen, C1-6 alkyl, and L3. For example,
L12 may be
¨C(0)NH-.
[0223] In some embodiments, 118 is an optionally substituted 5- or 6-membered
10 heteroaryl. le may be an optionally substituted 5- or 6- membered
heteroaryl, bound to
L3. In some embodiments, le is an optionally substituted pyridine, bound to
L3.
[0224] In some embodiments, L22 is selected from -C(0)-, and -C(0)NR10-. In
certain embodiments, L22 is -C(0)-. In certain embodiments, L22 is -C(0)Nr-.
Rw of
-C(0)NRio_ may be selected from hydrogen, C1-6 alkyl, and ¨L3. For example,
L22 may
15 be-C(0)NH-.
[0225] In some embodiments, R4 is selected front -010 , and -N(RI0)2; and Cl-
io alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-12 carbocycle, 3- to 12-membered
heterocycle,
aryl, and heteroaryl, each of which is optionally substituted with one or more

substituents independently selected from halogen, -ORM, _SRM, -
N(R10)2t _
S(0)1e ,
20 -S(0)211m, _0(0)Rio, _
C(0)0114- , -0C(0)R' , -NO2, =0, =S, =N(R1 ), -CN, Ci-io alkyl,
C2-10 alkenyl, and C2-to alkynyl and each of which is further optionally bound
to L3 In
some embodiments, R4 is -N(Rn2 and 124- of -N(11412 is selected from L3 and
hydrogen,
and wherein at least one le of -N(R1 )2 is L3.
[0226] In some aspects, the compound of Formula (IVB) is a compound of
25 Formula (IVC).
R1
N¨R2
R8-
L12 N,
(Iv.)
or a pharmaceutically acceptable salt thereof, wherein:
91
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
le and R2 are hydrogen;
L22 is _c(o)_;
R4 -N(R1 )2;
RR) is independently selected at each occurrence from hydrogen, -NI-12,
5 -C(0)0C1FC6H5; and Ci-io alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -CN, -NO2, -NH2, =0, =S,
-C(0)0C1tC6F15, -NHC(0)0CH2C6H5, Ci-io alkyl, C2-io alkenyl, C2-to alkynyl, C3-
12
carbocycle, 3- to 12-membered heterocycle, and haloalkyl;
10 Li2 is _c(0)*Rici,_
) *, wherein * represents where L12 is bound to Rs;
Rs is an optionally substituted fused 5-5, fused 5-6, or fused 6-6 bicyclic
heterocycle bound to linker moiety, 1,3, and wherein optional substituents are
independently selected at each occurrence from:
halogen, -0R1 , -C(0)N(R10)2, -N(R1
)C(0)R1 , -N(14.1 )C(0)N(R10)2,
15 -N(R11)2, -C(0)R' , -C(0)0R1 , -0C(0)R1 , -NO2, =0, =S, =N(R1 ), and -
CN;
Ci-to alkyl, C2-10 alkenyl, C2-to alkynyl, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -01e, -
S11.1 ,
-C(0)N(R10)2, -N(R1 )C(0)R1 , -N(R1 )C(0)N(R10)2, -N(R10)2, -C(0)R10, -C(0)0R'


,
-0C(0)R1 , -NO2, =0, =S, =N(R1 ), -CN, C3-12 carbocycle, and 3- to 12-membered

20 heterocycle; and
C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is
optionally substituted with one or more substituents independently selected
from
halogen, -010 , -SR1 , -C(0)N(R1 )2, -N(R1 )C(0)Rm, -N(R1 )C(0)N(R10)2, -
N(1110)2,
-C(0)1011, -C(0)0R'0, -0C(0)R1 , -NO2, =0, =S, =N(R10), -CN, C1-6alkyl, C2-6
alkenyl,
25 and C2-6alkynyl.
102271 In certain embodiments: Rl of -N(R1 )2is independently selected at
each
occurrence from methyl, ethyl, propyl, and butyl, any one of which is
optionally
substituted. In certain embodiments, le of -C(0)N(Rm)-* is hydrogen.
102281 In certain embodiments, R4 is -N(C14alkyl)2 and L12 is -C(0)N(H)-*.
92
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
CCH3
veNI
102291 In certain embodiments, R4 is
CH3; and/or Rm of -C(0)N(R10)_*
is hydrogen.
02301 In some embodiments, the compound is further covalently bound to a
linker, L3. In some embodiments, L3 is a noncleavable linker. In some
embodiments,
5 L3 is a cleavable linker. L3 may be cleavable by a lysosomal enzyme. In
some
embodiments, the compound is covalently attached to an antibody. In some
embodiments, the antibody specifically binds to a tumor antigen. In some
embodiments, the antibody comprises a target binding domain.
102311 In some embodiments, L3 is represented by the formula:
0
l
-kto
L4.--peptide-,L5-RX e We-
wherein:
L4 represents the C-terminus of the peptide and L5 is selected from a bond,
alkylene and heteroalkylene, wherein L5 is optionally substituted with one or
more
groups independently selected from R32, and RX is a reactive moiety; and
15 R32 is independently selected at each occurrence from halogen, -
OH, -CN,
-0-alkyl, -SH, =0, =S, -Nth, -NO2; and CI-to alkyl, C2-to alkenyl, C2-to
alkynyl, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -0-alkyl, -SH, =0, =S, -NH2, -NO2.
102321 In some embodiments, RX comprises a leaving group. In some
20 embodiments, RX comprises a maleimide. In some embodiments, L3 is
further
covalently bound to an antibody. In some embodiments, the antibody is directed

against a tumor antigen. In some embodiments, the antibody comprises a target
binding
domain.
102331 In some embodiments, L3 is represented by the formula:
93
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
X-ILO
wherein
L4 represents the C-terminal of the peptide and
L5 is selected from a bond, alkylene and heteroalkylene, wherein 1.2 is
optionally
5 substituted with one or more groups independently selected from R32;
Rrcomprises a bond, a succinimide moiety, or a hydrolyzed succinimide
moiety bound to a residue of an antibody, wherein -S. on RX* represents the
point of
attachment to the residue of the antibody; and,
R32 is independently selected at each occurrence from halogen, -OH, -CN,
10 alkyl, -SH, =0, =S, -NH2, -NO2; and Ct-io alkyl, C2-10 alkenyl, C2-10
alkynyl, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -0-alkyl, -SH, =0, =S, -N112, -NO2. In some
embodiments,
the peptide of L3 comprises Val¨Cit or Val¨Ala.
102341 In some aspects, the present disclosure provides a compound or salt
15 selected from:
FI2N TO
HN
o
H.21-11.(11
triN
0
NH2
0 4i
kiJ N N,
0
o
0 \--\
H2N.f.0
HN
o
H
\rENI
N
H 0 Oil
H2
N,
0
r¨s\
o
94
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
N
0
H
rfat ) N H2
0
N,
40
..
H
il 0 H
N
0
HN
H2NA 0
,
N
0 H in' N H2
0 AW-
y111.--'.-SN N,
. 0 ..
H
ri 0
H i H
N
0 Of
HN
H2NA 0
,
IIP
0 r ...i.,1 o
ce.0 A.
111 Nõ NH2
0 XrrEi 0 0 0 N **--"-se .C.A.%=N
H H *
/ N ,........-..,.............A N ...N
0
N
H H
N
o o
NH
A-
_iL
0 NH2
,
0
1 0
c.NO N )criii N *
o --
N, N H2
0 0
H
H
rj
-
ID H 0 --I H
N
NH
='---
0 NH2
,
H2N y0
HN
0 H
*
H H
0 0
N,.,,,..,%,
0
0 ty N 0 niNkl
0
H
NH2
0
N
N
H
-- r-=
N
95
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0 H
0
kk_k H
0 0 *
0 = y
ri 041. 0
NH2
0
N
0
H2Ny0
FIN
0 H 0
)LicewliNix2NkN
0
0 0 *
0
0 HAOuN 0
NH2
N,
-
0 \ \
H2Ny0
FIN
0 H 0
N
0
0 0
0
H
N
0
NH2
N,
r¨\\
0
H2N tO
HN
0 0
H
0 0
re. N
0
0
N
NH2
Nr
0 \
96
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0 H 0 H
N....}.. N
r t X ill 0 Ill 0-ITA
0
0 n
0
N
NU
N
H
1k_4? N"'

H2Nt0
HN
0 1-111141y1Icil
N
err N N 1 0
H NNH2
0 0 40 0,.....eNaiN
õ
0
II H
0 \Th,
H2Ny0
HN
0 0
11;11 J.:111(111
N
err i NI 0 s
0rais 0
NH2
0 .............õ
,,,...eN ---...i N,
0
H N
H
N
0 \Th
,
o o
o ON -)0 0 ouN 0
A.
cl....N.............301... "--rir rii 0 .
N._ NH2
N N
N
H H
H
0 0
--- C\
N
Hy
0 \Th.
H2N--%0
)
0
0
ic0 -1.toc ,r,i * ouN 0
ri,..............õõ. ..... a II: CR 4 0 NH2
N ..---
N,
N
H - H
H
0 Of
--- r--\
N
HN
0 \Th
H2NAo
,
and a salt of any one thereof
[0235] In some aspects, the present disclosure provides a compound or salt
selected from:
97
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
1 \II
ARXCW11 11)LN
H
NH2
0
N,
U
N
0
H --- r---\
N
40 \ ¨ \
H2N y.0
s
HN
0
a...,....õ..y H :Lliy 11
FRX* "---f-- - N
N
H
r 1 0 NH2
H_ 0 0
,
0 .,...---....
0......e.N
N
U
H
N
N ,
0
0
N
ist., NH2
O
0 0 A N ry *--"-
H 0
H
--- NiTh
H 0
HN
H2N-e1/40
N ,
0 1i,, kii 0
NH2
N,
O
---rirNõ 0õ..)1.. 0 0A H 0 H N
=Tr. RW:La....,...............}1.. ....
N - N
--- r--\\
H " H
N
Of-
HN
H2N'--LO
0 N ,
0 0
ri N.... kii N 0
NH2
O --tiry 0 is -ekri
H
IC
.11r-RX.,.............õ....}..,N N
0
N
---- r---- \
H H
N
0
0 µ--- \
NH
OA N H2
,
98
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
lie 0
N0
0 0 * 0 N
N
11
H
VIV(iNJL
ri 0
- N
r---\
H E H
N
0 -.10 teN
NH
CYAN H2
,
H2N tO
HN
NFIxtt-iiirlrl
1-1:01- NN 0 0
0
0 . yli ......-
----rii 0 r-- =ki 0 NH2
0
i(14....õ..-1.,N N,
H
-- r---\
N
H2N ,r0
HN
H ?
tRX.WIIN Aril N. H 0
H2
OyN.,,,.--...N 0 i.,...t.õ N,,sti 0
N
H
0
l!l....)-- ..N N,
HLJN:- Nr\
H2N,,,r0
HN.....
0
Hxit__ H
15.--RX IN N N
0
H H
HAC1N N 0
LL.,--- 0
NH2
N.,
N
H
-- r---\
N
99
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2N
yO
HN
H (311
N.:11TV' =
_ N
o
" H
0
0
-Ka N
N__ NH2
r-\\
0 \Th
H2N tO
HN
SH
0 0
0C.-1 N
U
NH2
reN
o
\Th
H2N
tO
HN
0
.NA\111
1- H
- N 0 0 'RI
0
TON
0
NH2
N
/Th
0 \Th
H2N y0
HN
0
iscnyLr 11\1
NH2
0 \ 0 le
0 /Th
0 \
100
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
H
1'14' 0 N N
ouN 0
NH2 _ 0
0
0
0
0
4
110 N
H H
0
0 \
H2W%
0
0
0 xtr.At 0 0-1-N-Th\----
N =
H2
tsi ri F F
ri
N
H a H
Of
rTh.
H2NAO
, and a salt of any one thereof,
5 wherein the RX* is a bond, a succinimide moiety, or a hydrolyzed
succinimide
moiety bound to a residue of an antibody,
wherein on RX* represents the point of
attachment to the residue of the
antibody.
[0236] In some embodiments, L3 is represented by the formula:
Vi\1/4111:9%)101.-CL
10 n RX , wherein RX comprises a
reactive moiety, and n =
0-9. In some embodiments, RX comprises a leaving group. In some embodiments,
RX
comprises a maleimide. In some embodiments, I: is represented as follows:
Sag,
47.(1\ltero-N%-9
=
µ3- , wherein RX* comprises a bond, a succinimide moiety,
or a hydrolyzed succinimide moiety bound to a residue of an antibody, wherein -
NI on
15 RX* represents the point of attachment to the residue of the antibody,
and n = 0-9.
101
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
102371 In some aspects, the present disclosure provides a compound or salt
selected from:
N
0
H NH2
cfixrio
isii,.õT.N N,
H
---- if-j.
N 0
N
0
0
0
N
.sekr......01i ii\ii ........õ....õ..............j. H
_.......õU 0 NI-I2
N
"%., N,
0 N
1-1
0 0
II *
-- r
0
0 Lnair Li
0
H
N
0 NAC1NVL N
N H2
N,
H
----
r¨\\
N
5
0
0
oN 0
cuN 0
H
N
0
.===="- N H2LjL,
N
H
---
r--\
N
and a salt of any one thereof
102381 In some aspects, the present disclosure provides a compound or salt
o
H
1 IN 0 NI-I2
N,
v Rxz. jai" N N
H H
0
--- r---N,
N
\ ---- \
selected from:
0 ,
4111) N
e(RXInGly ti 0 H I 1
N NI-12
N-_,..--.......õ----,_,AN N ,.,=.,..;ra.---**N ,
H H
N
10
o
102
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1.--Rknair 0
0 H N
)1".-CINU
NH2
N 40
r\
0 \---\
'1st-misty H 0
reõ.N..zzei
0
I
NH
H
2
rTh,
, and a salt of
any one thereof, wherein the RX*comprises a bond, a succinimide moiety, or a
hydrolyzed succinimide moiety bound to a residue of an antibody, wherein -IS
on RX*
5 represents the point of attachment to the residue of the antibody.
[0239] In some embodiments, RX* comprises a succinamide moiety and is
bound to a cysteine residue of an antibody. In some embodiments, RX* comprises
a
hydrolyzed succinamide moiety and is bound to a cysteine residue of an
antibody.
[0240] In some aspects, the present disclosure provides a conjugate
represented
(D¨L3fAntibody
10 by the formula: IS , wherein Antibody is an
antibody, D is a Category
A compound or salt disclosed herein, and 1-3 is a linker moiety.
[0241] In some aspects, the present disclosure provides a conjugate
represented
(D¨Lal-Antibody
by the formula: 1-8 , wherein
Antibody is an antibody and D-1_,3 is a
Category A compound or salt disclosed herein.
15 [0242] In some aspects, the present disclosure provides a
pharmaceutical
composition, comprising the conjugate disclosed herein and at least one
pharmaceutically acceptable excipient.
[0243] In some embodiments, the average DAR of the conjugate is from about 2
to about 8, or about 1 to about 3, or about 3 to about 5.
103
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Compounds of Category B. TLR7 Agonists
102441 In some aspects, the present disclosure provides a compound represented
by the structure of Formula (IA):
(R15)w
R1
N Nõ
=-=-=
R3
R4 R11 R12 I
Xi
N--/(
,N
R5-
---1Ati\CX2 N IA); IAC1 R6
Ri 3 R14 R9 R10
R7 R8
(IA),
5 or a pharmaceutically acceptable salt thereof, wherein:
R', R2, R3, R4, and R5 are independently selected from hydrogen; and Cl4
alkyl,
C2-6 alkenyl, and C2-6 alkynyl, each of which is optionally substituted with
one or more
substituents independently selected from halogen, -0R20, -SR20, -C(0)N(R20)2,
-N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0, =S,
10 =N(R20), and -CN; or R3 and It" taken together form a 5- to 10-membered
heterocycle
optionally substituted with one or more substituents independently selected
from
halogen, -OR', -SR20, -C(0)N(R20)2, -N(R2 )2, -S(0)R20, -S(0)2R20, -C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), and -CN;
R6 is selected from halogen, -OR", -N(R20)2, -C(0)N(R20)2, -C(0)R20

,
15 -C(0)0R20, -S(0)R20, and -S(0)2R20; and C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(1t20), and -CN;
R7, R8, R9, and IV are independently selected at each occurrence from
hydrogen
20 and halogen; and C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of
which is optionally
substituted with one or more substituents independently selected from halogen;
R" and 10-2 are independently selected from hydrogen, halogen, -OR', -SR20

,
-C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R2 , -C(0)R20, -C(0)0R20, -0C(0)R20, -
NO2,
and -CN; and C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which is
optionally
25 substituted with one or more substituents independently selected from
halogen, -0R20

,
104
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
-SR20, -C(C)N(R20)2, -N(R20)2, _s(0)-r20, _
S(0)2R2 , -C(0)R20, -C(0)0R20, -0C(0)R20

,
-NO2, =0, =S, =N(R20), -CN, C3-12 carbocycle, and 3- to 12-membered
heterocycle; or
R11 and R12 taken together form a C3-6 carbocycle optionally substituted with
one or
more substituents independently selected from halogen, -0R20, -SR20, -
C(0)N(R20)2,
5 -N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0,
=S,
=N(R20), and -CN;
R13 and R14 are independently selected at each occurrence from hydrogen,
halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, and -CN; C1-6 alkyl, C2-6 alkenyl, and C2-6
alkynyl, each
10 of which is optionally substituted with one or more substituents
independently selected
from halogen, -OR', -SR", -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20, -
C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), -CN, C3-n carbocycle, and 3- to 12-

membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle,
each
of which is optionally substituted with one or more substituents independently
selected
15 from halogen, -OR', -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20, -
C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), -CN, CI-6 alkyl, C2-6 alkenyl, and
C2-6
alkynyl;
R15 is independently selected at each occurrence from halogen, -OR', -SR20

,
-C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R2 , -C(0)R20, -C(0)0R20, -0C(0)R20, -
NO2,
20 0, =S, =N(R20), -CN, CI-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-12
carbocycle, and 3- to
12-membered heterocycle, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -OH, -CN, -NO2, -NH2, =0,
=S, -CI-6
alkyl, -C1-6 haloalkyl, -0-C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-12
carbocycle, and 3-to
12-membered heterocycle;
25 R16 is selected from hydrogen, and CI-6 alkyl, C2-6 alkenyl, C24
alkynyl, C3-L2
carbocycle, and 3- to 12-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -OH, -CN, -
NO2,
-NH2, =0, =S, C1-6 alkyl, -C1-6 haloalkyl, -0-C1-6 alkyl, C2-6 alkenyl, C2-6
allcynyl, C3-12
carbocycle, and 3- to 12-membered heterocycle;
30 R2 is independently selected at each occurrence from hydrogen;
and CI-6 alkyl,
C2-6 alkenyl, C2-6 alkynyl, C3-I2 carbocycle, and 3- to 12-membered
heterocycle, each of
105
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -NO2, -NH2, =0, =S, -C(0)0CH2C6H5, -NHC(0)0CH2C6Hs,
C1-6 alkyl, -C1-6 haloalkyl, -0-CE-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-12
carbocycle, and
3- to 12-membered heterocycle;
5 X` is 0, S, or NR16;
X2 is C(0) or S(0)2;
n is 1, 2, or 3;
x is 1, 2, or 3,
w is 0, 1, 2, 3, or 4; and
10 z is 0, 1, or 2.
[0245] In certain embodiments, for a compound of Formula (IA), wherein is
0. In certain embodiments, for a compound of Formula (IA), n is 2. In certain
embodiments, for a compound of Formula (IA), x is 2. In certain embodiments,
for a
compound of Formula (IA), z is 0. In certain embodiments, for a compound of
Formula
15 (IA), z is 1.
[0246] In certain embodiments, a compound of Formula (IA) is represented by
Formula (113):
(R15)w
R1
N
N,R 2
I
R11 R12 R3 R9' R1 Cr .__7=1 _8.1
Izt
X Xi R6
R4 R9" R10"
REr
(113),
or a pharmaceutically acceptable salt thereof, wherein RT, Rr, R8 Rr, R9.,
R9", ler,
20 and RR' are independently selected at each occurrence from hydrogen and
halogen; and
Ci-isalkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which is optionally
substituted with
one or more substituents independently selected from halogen.
[0247] In certain embodiments, a compound of Formula (IA) is represented by
Formula (IC):
106
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(R15)w
R1
N N
R2
Ra R11 R12 R3 R9I Rty R7õ R8õ
N--
R5¨N -7\--)C LA---\CX1)C(1( R6
Ri3Ria Rg" RI
R7' Rs. (IC),
or a pharmaceutically acceptable salt thereof, wherein R7I, R7", By, Ran, R9r,
R9n, R10',
and WIT are independently selected at each occurrence from hydrogen and
halogen; and
C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which is optionally
substituted with
5 one or more substituents independently selected from halogen.
[0248] In certain embodiments, for a compound or salt of any one of Formulas
(LA), (B3), or (IC), le, R2, R3, n4,
K and R5 are independently selected from hydrogen and
C1-6 alkyl optionally substituted with one or more substituents independently
selected
from halogen, -OR', -SR20, -C(0)N(R20)2, _
)2
N(R20+, _ S(0)R2 , -S(0)2R20

,
10 -C(0)R20, _c(0)0R20, _oc(0)R20, -NO2, =0, =S, =N(t20), and ¨CN_
102491 In certain embodiments, for a compound or salt of any one of Formulas
(IA), (B3), or (IC), le and R2 are independently selected from hydrogen and C1-
6 alkyl.
In certain embodiments, for a compound or salt of any one of Formulas (IA),
(IB), or
(IC), le and R2 are each hydrogen.
15 [0250] In certain embodiments, for a compound or salt of any one
of Formulas
(IA), (B3), or (IC), R3 is selected from hydrogen and C1-6 alkyl optionally
substituted
with one or more halogens.
[0251] In certain embodiments, for a compound or salt of any one of Formulas
(IA), ([13), or (IC), R3 is hydrogen.
20 [0252] In certain embodiments, for a compound or salt of any one
of Formulas
(IA), (B3), or (IC), le is selected from hydrogen and C1-6 alkyl optionally
substituted
with one or more halogens.
[0253] In certain embodiments, for a compound or salt of any one of Formulas
(IA), (B3), or (IC), le is hydrogen.
25 [0254] In certain embodiments, for a compound or salt of any one
of Formulas
(IA), (B3), or (IC), R5 is selected from hydrogen and C1-6 alkyl optionally
substituted
with one or more substituents independently selected from halogen, -0R20, -
SR20, -
107
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
C(0)N(R2o)2, _mR20)2, _se:01-nIt2o, _ S(0)2R2o, _c(0)-nIt2o, _ C(0)011.2 , -
0C(0)12.20, -NO2,
=S, =N(R20), and -CN. In certain embodiments, for a compound or salt of any
one
of Formulas (IA), (JIB), or (IC), R5 is hydrogen.
102551 In certain embodiments, for a compound or salt of any one of Formulas
5 (IA), (IB), or (IC), R6 is selected from halogen, -OR', and -N(R20)2; and
CI-6alk-0, C2-6
alkenyl, C24allcynyl, each of which is optionally substituted with one or more
substituentsindependently selected from halogen, -0R20
--
_sR20, _c(o)m-R2o.) 2,N(R9n)2, _
_s(0)R20, _s(0)2R20, _C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0, =S, =1\la20),
and -
CN; and
10 R2 is independently selected at each occurrence from hydrogen;
and C1-6 alkyl,
C2-6 alkenyl, C2-6 alkynyl, C3-12 carbocycle, and 3- to 12-membered
heterocycle, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -NO2, -NW, =0, =S, -C(0)0CH2C6145, -NHC(0)0CH2C6Hs,
C1-6 alkyl, -C1-6 haloalkyl, -0-CE-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-12
carbocycle, and
15 3- to 12-membered heterocycle.
102561 In certain embodiments, for a compound or salt of any one of Formulas
(IA), (LB), or (IC),
It.6 is C1-6 alkyl optionally substituted with one or more substituents
independently selected from halogen, -0R20 , -SR", -C(0)N(R20)2, _N(R20) _
S(0)R2 ,
20 -S(0)2R20, _corm, _
K C(0)0R20, -0C(0)R20; and
R2 is independently selected at each occurrence from hydrogen; C1-6 alkyl, C3-
12
carbocycle, and 3- to 12-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -OH, -CN, -
NO2,
-NH2, =0, =S, -C(0)0CH2C6if5, -NHC(0)0CH2C6H5, C1-6 alkyl, -Ci-6haloalkyl,
25 -0-C14 alkyl, C24 alkenyl, C24 alkynyl, C3-12 carbocycle, and 3- to 12-
membered
heterocycle.
102571 In certain embodiments, for a compound or salt of any one of Formulas
(IA), (1B), or (IC), R6 is Ci4 alkyl substituted with -0R20, and R20 is
selected from
hydrogen and C1-6 alkyl optionally substituted with one or more substituents
30 independently selected from halogen, -OH, and -N1-12.
108
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
[0258] In certain embodiments, for a compound or salt of any one of Formulas
(IA), (JIB), or (IC), R71, R7", Rw, R8", R91, R9", Rlil, and Rlip" are
independently selected at
each occurrence from hydrogen and halogen; and C1-6 alkyl, optionally
substituted with
one or more substituents independently selected from halogen.
5 [0259] In certain embodiments, for a compound or salt of any one
of Formulas
(LB) or (IC), wherein R7' and Rs' are each hydrogen. In certain embodiments,
for a
compound or salt of any one of Formulas (JIB) or (IC), wherein R7" and Rs" are
each C1-6
alkyl. In certain embodiments, for a compound or salt of any one of Formulas
(LB) or
(IC), RT" and Rs" are each methyl.
10 102601 In certain embodiments, for a compound or salt of any one
of Formulas
(JIB) or (IC), R9P, R9", Riot, and Rm.' are independently selected at each
occurrence from
hydrogen and C1-6 alkyl.
[0261] In certain embodiments, for a compound or salt of any one of Formulas
([13) or (IC), R9', R9, Rill, and R1 " are each hydrogen.
15 [0262] In certain embodiments, for a compound or salt of any one
of Formulas
(IA), (113), or (IC), R11 and R12 are independently selected from hydrogen,
halogen, -OR', _sR20, _C(0)N(R20)2, _TAR2o)2, _C(0)R20, -C(0)0R20, -0C(0)R20;
and
Cbealkyl, optionally substituted with one or more substituents independently
selected
from halogen, -0R20, _sR20, -C(0)N(R20)2,
N(R20)2, -C(0)R20, -C(0)0R20, _OC(0)R20

,
20 C3-12 carbocycle, and 3- to 12-membered heterocycle.
[0263] In certain embodiments, for a compound or salt of any one of Formulas
(IA) or (IC), Rn and R" are independently selected from hydrogen, halogen, -
0R20

,
-SR20, -C(0)N(R20)z, 2
_ Nat2o),, _
C(0)R2 , -C(0)0R20, -0C(0)R20.; and Ci-6 alkyl
optionally substituted with one or more substituents independently selected
from
25 halogen, -0R20, -5R20, -C(0)N(R20)2, 2
ick ())2, -C(0)R20, -C(C)0R20, -0C(0)R20, C3-12
carbocycle, and 3- to 12-membered heterocycle.
[0264] In certain embodiments, for a compound or salt of any one of Formulas
(IA), (1B), or (IC), le and R" taken together form an optionally substituted 5-
to 6-
membered heterocycle.
109
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
102651 In certain embodiments, for a compound or salt of any one of Formulas
(IA), (B3), or (IC), R" and R12 taken together form an optionally substituted
C3-6
carbocycle.
102661 In certain embodiments, for a compound or salt of any one of Formulas
5 (IA), (lE), or (IC), X2 is C(0).
102671 In certain embodiments, the compound is represented by:
4.1 R., NH2
../
H N
0
H2N....õ...----irN-...,.................-----........0
O /
õI N..... NH2
---."
N
jr,-N-,..õ...........õ--...,,,0
0
H2N
O /
0, N..... NH2
...,"
H N
Isy_yert¨c_ C Xr-N-....õ.............--...õ,
H2N 0 0
O /
0 Ns, N H2
I _..,.
H - N
X.....y_N-.......õ.õ,..-.....Th
0
H2N
10 0 /
N,... NH2
H
oxyl*N
I
CN....,.............,..--õ,õ
0
H2N
0
/
HO op
N NH2
I
H
. "--- N
I)4,7N-4 0ir N...,..õ..,.....--õ..õ
0
, or
HN *
NH2
H
N
I
H2N - N
-..,...........-----...õõ0
\-0
0 3
110
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
or a pharmaceutically acceptable salt of any one thereof
102681 In certain aspects, the disclosure provides a pharmaceutical
composition
of a compound or pharmaceutically acceptable salt of any one of Formulas (IA),
(B3),
or (IC), and a pharmaceutically acceptable excipient.
5 102691 In certain embodiments, for a compound or salt of any one
of Formulas
(IA), (B3), or (IC), the compound or salt is further covalently bound to a
linker,
102701 In certain aspects the disclosure provides a compound represented by
Formula (HA):
(R15),õ
R21
N NõR-
R23
R4 R11 R12 I
N_1(
R26e#N--)c;Xlyi R6
R13 Ria Rs Rlo
R7 Rs (HA),
10 or a pharmaceutically acceptable salt thereof, wherein:
R2 and le are independently selected from hydrogen; and C1-6 alkyl, C2-6
alkenyl, and C2-6 alkynyl, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -0R20, -SR20, -C(0)N(R20)2,
-N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0, =S,
15 =N(R20), and -CN;
R21, .k. n23,
and R25 are independently selected from hydrogen; C1-6 alkyl, C2-6
alkenyl, and C24 alkynyl, each of which is optionally substituted with one or
more
substituents independently selected from halogen, -0R20, -SR20, -C(0)N(R20)2,
-N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0, =S,
20 =N(Rm), and -CN; and or R23 and R11 taken together form a 5- to
10-membered
heterocycle optionally substituted with one or more substituents independently
selected
from halogen, -0R20, _sR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), and -CN; and wherein one of R21,
R23,
and R25 is L3;
25 R6 is selected from halogen, -0R20, _N(R20)2,
(0 )N(R2 )2, C A2 ,
ill
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-C(0)0R20, -S(0)R20, and -S(0)2R20; and C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)21k20, -
C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), and -CN;
5 le, le, R9, and It1 are independently selected at each occurrence
from hydrogen
and halogen; and CI-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which is
optionally
substituted with one or more substituents independently selected from halogen;
R1". and R12 are independently selected from hydrogen, halogen, -OR', -SR20

,
-C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R2 , -C(0)R20, -C(0)0R20, -0C(0)R20, -
NO2,
10 and -CN; and C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which
is optionally
substituted with one or more substituents independently selected from halogen,
-0R20

,
-SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, eS(0)2R20, -C(0)R20, -C(0)0R20, -
0C(0)R20

,
-NO2, =0, =S, =N(R20), -CN, C3-12 carbocycle, and 3- to 12-membered
heterocycle; or
It" and R12 taken together form a C3-6 carbocycle optionally substituted with
one or
15 more substituents independently selected from halogen, -0R20, -SR20, -
C(0)N(R20)2,
-N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -0C(0)R20, -NO2, =0, =S,
=N(R20), and -CN;
R" and RH are independently selected at each occurrence from hydrogen,
halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20

,
20 -C(0)0R20, -0C(0)R20, -NO2, -CN, C1-6 alkyl, C2-6 alkenyl, and C2-6
alkynyl, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OR', -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), -CN, C3-12 carbocycle, and 3- to
12-
membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle,
each
25 of which is optionally substituted with one or more substituents
independently selected
from halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -
C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), -CN, C1-6 alkyl, C2-6 alkenyl,
and C2-6
alkynyl;
R15 is independently selected at each occurrence from halogen, -0R20, -SR20

,
30 -C(0)N(R20)2, -N(R20)2, -S(0)R20, -S(0)2R20, -C(0)R20, -C(0)0R20, -
0C(0)R20, -NO2,
=S, =N(R20), -CN, CI-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, C3-12
carbocycle, and 3-
112
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
to 12-membered heterocycle, each of which is optionally substituted with one
or more
substituents independently selected from halogen, -OH, -CN, -NO2, -NI-b, =0,
=S, -C1-6
alkyl, -C1-6haloalkyl, -0-Ct-6alkyl, C2-6 alkenyl, C2-6a1kynyl, C3-12
carbocycle, and 3-to
12-membered heterocycle;
5 It' is selected from hydrogen; and CI-fl alkyl, C2-6 alkenyl, C2-6
alkynyl, C3-12
carbocycle, and 3- to 12-membered heterocycle, each of which is optionally
substituted
with one or more substituents independently selected from halogen, -OH, -CN, -
NO2,
-NH2, =0, =S, C1-6 alkyl, -Cl-flhaloalkyl, -0-C1-6 alkyl, C2-6 alkenyl, C2-
6alkynyl, C3-12
carbocycle, and 3- to 12-membered heterocycle;
10 R2 is independently selected at each occurrence from hydrogen; C1-
6 alkyl, C2-6
alkenyl, C2-6 alkynyl, C3-12 carbocycle, and 3- to 12-membered heterocycle,
each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -NO2, -NW, =0, =S, -C(0)0CH2C6145, -NHC(0)0CH2C6Hs,
CI-6 alkyl, -C1-6 haloalkyl, -0-C t-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-12
carbocycle, and
15 3- to 12-membered heterocycle;
L3 is a linker;
X' is 0, S, or NR16;
X2 is C(0) or S(0)2;
n is 1, 2, or 3;
20 x is 1, 2, or 3;
w is 0, 1, 2, 3, or 4; and
z is 0, 1, or 2.
[0271] In certain embodiments, for a compound or salt of Formula (IIA), X' is
0. In certain embodiments, for a compound or salt of Formula (BA), n is 2. In
certain
25 embodiments, for a compound or salt of Formula (11A), x is 2. In certain
embodiments,
for a compound or salt of Formula (HA), z is 0. In certain embodiments, for a
compound or salt of Formula (HA), z is 1.
[0272] In certain embodiments, the compound of Formula (IA) is represented
by (BB) or (IC):
113
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(R15)w
R21
N N,
R2
0 1
Rii R12 R23 049 010
,
R25 \
R1SzN _2(
R6
1
R4 Ro" Ri 0"
R7. R8
MBA
(R16)w
R21
N
N,R 2
23 R11 R R9' R10'
I
R25 R12
RT. R8õ
1
R4¨ N
X1 \R6
R13 R14 R9" Rio"
RT R81
(IIC),
or a pharmaceutically acceptable salt thereof, wherein R7', R7n, Rw, R8, R9t,
R9,
and RI' are independently selected at each occurrence from hydrogen and
halogen; and
5 Ct-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl, each of which is optionally
substituted with
one or more substituents independently selected from halogen.
102731 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (IIC), R2 and le are independently selected from hydrogen and
C1-6 alkyl
optionally substituted with one or more substituents independently selected
from
10 halogen, -0R20, _sR20, _c(o)N(R20)2, _mR2o)2., _
S(0)R2 , -S(0)2R20, -C(0)R20

,
-C(0)0R20, -0C(0)R20, -NO2, =0, =S, =N(R20), and ¨CN.
102741 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (IIC), R2 and R4 are independently selected from hydrogen and
C1-6
alk-yl. In certain embodiments, for a compound or salt of any one of Formulas
(IA),
15 (IIB), or (11C), R2 and R4 are each hydrogen.
102751 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (IIC), R23 is selected from hydrogen and C1-6 alkyl optionally
substituted
with one or more halogens. In certain embodiments, for a compound or salt of
any one
of Formulas (IA), (IfB), or (IIC), R23 is hydrogen.
114
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0276] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IM), or (HC), R2' is selected from hydrogen and C1-6 alkyl optionally
substituted
with one or more halogens. In certain embodiments, for a compound or salt of
any one
of Formulas (HA), (I1B), or (IIC), R2' is hydrogen.
5 [0277] In certain embodiments, for a compound or salt of any one
of Formulas
(HA), (ID3), or (IIC), R2' is L3.
102781 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (ID!), or (IIC), R25 is selected from hydrogen and C1-6 alkyl,
optionally
substituted with one or more substituents independently selected from halogen,
-0R20

,
10 _sR20, _qcomR20)2, _NR-R20µft2, _
S(0)R2 , -S(0)2R20, -C(0).K.n20, _C(0)0R20, -0C(0)R20

,
-NO2, =0, =S, =N(R20), and -CN. In certain embodiments, for a compound or salt
of
any one of Formulas (HA), (BB), or (IIC), R25 is hydrogen.
[0279] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (ID3), or (IIC), R25 is L3.
15 [0280] In certain embodiments, for a compound or salt of any one
of Formulas
(HA), (IH3), or (HC),
1(6 is selected from halogen, -OR', and eN(R20)2; and C1-6 alkyl, C24 alkenyl,

C2-6 alkynyl, each of which is optionally substituted with one or more
substituents
independently selected from halogen, -0R20, _sR20, _0(Q)N(R20)2, _N(R20)2, _
S(0)R2 ,
20 -S(0)2R20, _cor2o, _
.K. C(0)0R20, -0C(0)R20, -NO2, =0,
=S, =N(R20), and -CN; and
1(20 is independently selected at each occurrence from hydrogen; and C1-6
alkyl,
C2-6 alkenyl, C2-6 alkynyl, C3-12 carbocycle, and 3- to 12-membered
heterocycle, each of
which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -NO2, -NH2, =0, =S, -C(0)0CH2C6H5, -NHC(0)0CH2C6Hs,
25 C14 alkyl, -C14 haloalkyl, -0-CI-6 alkyl, C24 alkenyl, C24 alkynyl, C3-
12 carbocycle, and
3- to 12-membered heterocycle.
[0281] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (ID3), or (IIC),
1t6 is C1-6 alkyl optionally substituted with one or more substituents
30 independently selected from halogen, -OR', -SR20, -C(0)N(R20)2,
_TAR20)2, _so0ooR20

,
-S(0)2R20, _cor20, _
C(0)0R2 , -0C(0)R20; and
115
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
1t20 is independently selected at each occurrence from hydrogen, -NH2,
-C(0)0CH2C6H5; CI-6 alkyl, C3-12 carbocycle, and 3-to 12-membered heterocycle,
each
of which is optionally substituted with one or more substituents independently
selected
from halogen, -OH, -CN, -NO2, -NH2, =0, =S, -C(0)0CH2C6H5, -NHC(0)0CH2C6Hs,
5 CI-6 alkyl, -C1-6 haloalkyl, -0-C r-6 alkyl, C2-6 alkenyl, C2-6 alkynyl,
C3-12 carbocycle, and
3- to 12-membered heterocycle.
102821 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (ID3), or (HC),
R6 is C1-6 alkyl substituted with -0R20, and
10 R2 is selected from hydrogen and CI-6 alkyl, which is optionally
substituted
with one or more substituents independently selected from halogen, -OH, and -
NH2.
102831 In certain embodiments, for a compound or salt of any one of Formulas
(ITB) or (IIC), Rr, R7", IV', le", R91., R911, ler, and Rw" are independently
selected at
each occurrence from hydrogen and halogen; and CI-6 alkyl optionally
substituted with
15 one or more substituents independently selected from halogen.
102841 In certain embodiments, for a compound or salt of any one of Formulas
(Ill) or (IIC), R7' and BY are hydrogen.
102851 In certain embodiments, for a compound or salt of any one of Formulas
(Ill) or (IIC), R7" and RP are CI-6 alkyl.
20 102861 In certain embodiments, for a compound or salt of any one
of Formulas
(DB) or (LIC), R7" and R8" are methyl.
102871 In certain embodiments, for a compound or salt of any one of Formulas
(nil) or (HC), R9r, Rw, and Rw" are independently
selected at each occurrence from
hydrogen and CI-6 alkyl.
25 102881 In certain embodiments, for a compound or salt of any one
of Formulas
(DB) or (HC), R9', R9", 10 , and Rw" are each hydrogen.
102891 In certain embodiments, for a compound or salt of any one of Formulas
(HA), (ID3), or (HC), R" and 1:0-2 are independently selected from hydrogen,
halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -C(0)R20, -C(0)0R20, and -
0C(0)R20;
30 and C14alkyl optionally substituted with one or more substituents
independently
selected from halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -C(0)R20, -
C(0)0R20

,
116
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
-0C(0)R20, C3-12 carbocycle, and 3- to 12-membered heterocycle.
[0290] In certain embodiments, for a compound or salt of any one of Formulas
(HA) or (HC), R13 and R14 are independently selected from hydrogen,
halogen, -OR', -SR20, -C(0)N(R20)2, -N(R20)2, -C(0)1120, -C(0)0R20, and -
0C(0)R20;
5 and CL-6 alkyl optionally substituted with one or more substituents
independently
selected from halogen, -0R20, -SR20, -C(0)N(R20)2, -N(R20)2, -C(0)R20, -
C(0)0R20

,
-0C(0)R20, C3-12 carbocycle, and 3- to 12-membered heterocycle.
[0291] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (HC), R23 and R" taken together form an optionally substituted
10 5- to 6-membered heterocycle.
[0292] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (HC), R" and R12 taken together form an optionally substituted
C34
carbocycle.
[0293] In certain embodiments, for a compound or salt of any one of Formulas
15 (HA), (IIB), or (IIC), X2 is C(0).
[0294] In certain embodiments, for a compound or salt of any one of Formulas
(HA), (IIB), or (IIC), L3 is a cleavable linker. In certain embodiments, for a
compound
or salt of any one of Formulas (IA), (BB), or (IIC), L3 is cleavable by a
lysosomal
enzyme.
20 [0295] In certain embodiments, for a compound or salt of any one
of Formulas
(HA), (BB), or (LIC), L3 is represented by the formula:
0
1 itepeptide,, L5 rRX
N---1
-
H
wherein:
L4 represents the C-terminus of the peptide and L5 is selected from a bond,
25 alkylene and heteroalkylene, wherein 12 is optionally substituted with
one or more
groups independently selected from RTh, and RX is a reactive moiety; and
1(30 is independently selected at each occurrence from halogen, -OH, -CN, -0-
alkyl, -SH, =0, =S, -NH2, -NO2; and Ci-Cio alkyl, C2-Cio alkenyl, and C2-Cio
alkynyl,
117
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
each of which is independently optionally substituted at each occurrence with
one or
more substituents selected from halogen, -OH, -CN, -0-alkyl, -SH, =0, =S, -NI-
12, and
-NO2.
[0295] In certain embodiments, for a compound or salt of any one of Formulas
5 (HA), (BB), or (HC), RX comprises a leaving group. In certain
embodiments, for a
compound or salt of any one of Formulas WAX (I113), or (IIC), RX is a
maleimide or an
alpha-halo carbonyl. In certain embodiments, for a compound or salt of any one
of
Formulas (HA), (LIE), or (HC), the peptide of L3 comprises Val-Cit or Val-Ala.
[0297] In certain embodiments, for a compound or salt of any one of Formulas
10 (HA), (ID3), or (LIC), L3 is represented by the formula:
iss(litc eticc
-9 n
RX
wherein:
RX comprises a reactive moiety; and
n is 0-9.
15 [0298] In certain embodiments, for a compound or salt of any one
of Formulas
(HA), (I1.13), or (HC), RX comprises a leaving group. In certain embodiments,
for a
compound or salt of any one of Formulas (hA), (DB), or (IIC), RX is a
maleimide or an
alpha-halo carbonyl. In certain embodiments, for a compound or salt of any one
of
Formulas (HA), (1113), or (HC), L3 is further covalently bound to an antibody
to form a
20 conjugate.
[0299] In certain embodiments, the disclosure provides a conjugate represented

by the formula:
(D L3_)
wherein:
25 n is 1 to 20;
D is a compound or salt of any one of a Category B compound of Formulas
(IA), (I13), or (IC); and L3 is a linker moiety; or
118
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
D-L3 is a compound or salt of any one of a Category B compound of Formulas
(HA), (DB), or (IIC).
[0300] In certain embodiments, for a conjugate of a compound or salt of any
one of
Formulas (IA), (D3), (IC), (HA), (JIB), and (IIC), n is selected from 1 to 8.
In certain
5 embodiments, for a conjugate of a compound or salt of any one of Formulas
(IA), (1113),
(IC), (IA), (DB), and (IIC), n is selected from 2 to 5. In certain
embodiments, for a
conjugate of a compound or salt of any one of Formulas (IA), (1B), (IC), (IA),
(DB),
and (IIC), n is 2.
103011 In certain embodiments, for a compound or salt of any one of Formulas
10 (HA), (ID3), and (IIC), -L3 is represented by the formula:
0
VILO
4,peptide,, 5-RX,/
wherein:
L4 represents the C-terminus of the peptide and L5 is selected from a bond,
alkylene and heteroalkylene, wherein L5 is optionally substituted with one or
more
15 groups independently selected from R30;
RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide moiety bound
to a residue of an antibody, wherein 7 on RX* represents the point of
attachment to
the residue of the antibody; and
R3 is independently selected at each occurrence from halogen, -OH, -CN, -0-
20 alkyl, -SH, =0, =S, -NH2, -NO2; and CE-Ctoalkyl, C2-Cmalkenyl, and C2-
Cioalkynyl,
each of which is independently optionally substituted at each occurrence with
one or
more substituents selected from halogen, -OH, -CN, -0-alkyl, -SH, =0, =S, -
NH2, and
-NO2.
[0302] In certain embodiments, for a compound or salt of any one of Formulas
25 (HA), (ID3), or (IIC), RX* is a succinamide moiety, hydrolyzed
succinamide moiety or a
mixture thereof and is bound to a cysteine residue of an antibody.
[0303] In certain embodiments for a compound of Formulas (ILA), (JIB) and
(DC), -L3 is represented by the formula:
119
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
wherein:
Rris a bond, a succinimide moiety, or a hydrolyzed succinimide moiety bound
to a residue of an antibody, wherein 7 on RX* represents the point of
attachment to
5 the residue of the antibody; and
n is 0-9.
103041 In some aspects, the present disclosure provides a conjugate
represented
by the following structure:
a
0
G;
-=Vt"*.Te.%%4%.-AN
a
/1
10 or a pharmaceutically acceptable salt thereof, wherein Ab comprises an
antibody, D is a
compound or salt of a Category B compound of Formula (HD):
'NH
1>-
1
\
(I1D)
wherein R4 is alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl,
heteroaryl,
arylalkyl, or heteroarylalkyl group comprising from 1 to 8 carbons, each J is
hydrogen,
15 each U is N, each t is 2, Q is not present, the dashed line represents a
point of
attachment of the adjuvant to G1 , and G1 is a bond; subscript a is an integer
from 1 to
40; and subscript r is an integer from 1 to 10. In certain embodiments, D has
the
following structure:
120
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
NH2
N 44,N>art
traL.,õ
H
CN)
-4- . In further embodiments, the
conjugate has the following
structure:
0
At
N
k
H2NAN irr
Linkers
5 [0305] The conjugates include a linker(s) that attaches an
antibody to at least
one immune-stimulatory compound, such as a myeloid cell agonist. A linker can
be,
for example, a cleavable or a non-cleavable linker. A conjugate can comprise
multiple
linkers. The linkers in a conjugate can be the same linkers or different
linkers.
[0306] As will be appreciated by skilled artisans, a linker connects an immune-

10 stimulatory compound(s), such as a myeloid cell agonist, to the antibody
by forming a
covalent linkage to the compound at one location and a covalent linkage to the
antibody
at another location. The covalent linkages can be formed by reaction between
functional groups on the linker and functional groups on the immune-
stimulatory
compound and on the antibody. As used herein, the expression "linker" can
include (i)
15 unattached forms of the linker that can include a functional group
capable of covalently
attaching the linker to an immune-stimulatory compound and a functional group
capable of covalently attached the linker to an antibody; (ii) partially
attached forms of
the linker that can include a functional group capable of covalently attaching
the linker
to an antibody and that can be covalently attached to an immune-stimulatory
20 compound, or vice versa; and (iii) fully attached forms of the linker
that can be
covalently attached to both an immune stimulatory compound and to an antibody.
In
some specific embodiments, the functional groups on a linker and covalent
linkages
121
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
formed between the linker and an antibody can be specifically illustrated as
Rx and Rx',
respectively.
[0307] A linker can be short or long, and cleavable or non-cleavable. A linker

can contain segments that have different characteristics, such as segments of
flexibility
5 or segments of rigidity, segments of hydrophilicity, and/or segments of
hydrophobicity.
A linker can be chemically stable to extracellular environments, for example,
chemically stable in the blood stream, and/or may include linkages that are
not stable.
A linker can include linkages that are designed to cleave and/or immolate or
otherwise
breakdown specifically or non-specifically inside cells. A cleavable linker
can be
10 sensitive to enzymes at a specific site, such as the lysosome or the
extracellar space
adjacent cancer cells.
[0308] A cleavable linker can include a valine-citrulline peptide, a valine-
alanine peptide, a phenylalanine-lysine or other peptide, such as a peptide
that forms a
protease recognition and cleavage site. Such a peptide-containing linker can
contain a
15 pentafluorophenyl group. A peptide-containing linker can include a
succimide or a
maleimide group A peptide-containing linker can include a para aminobenzoic
acid
(PABA) group. A peptide-containing linker can include an
aminobenzyloxycarbonyl
(PABC) group. A peptide-containing linker can include a PABA or PABC group and
a
pentafluorophenyl group. A peptide-containing linker can include a PABA or
PABC
20 group and a succinimide group. A peptide-containing linker can include a
PABA or
PABC group and a maleimide group.
[0309] A non-cleavable linker is generally protease-insensitive and
insensitive
to intracellular processes. A non-cleavable linker can include a maleimide
group. A
non-cleavable linker can include a succinimide group. A non-cleavable linker
can be
25 maleimido-alkyl-C(0)- linker. A non-cleavable linker can be
maleimidocaproyl linker.
A maleimidocaproyl linker can be N-maleimidomethylcyclohexane-1-carboxylate. A

maleimidocaproyl linker can include a succinimide group. A maleimidocaproyl
linker
can include pentafluorophenyl group.
[0310] A linker can be a combination of a maleimidocaproyl group and one or
30 more polyethylene glycol molecules. A linker can be a maleimide-PEG4
linker. A
linker can be a combination of a maleimidocaproyl linker containing a
succinimide
122
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
group and one or more polyethylene glycol molecule& A linker can be a
combination
of a maleimidocaproyl linker containing a pentafluorophenyl group and one or
more
polyethylene glycol molecules. A linker can contain a ma1eimide(s) linked to
polyethylene glycol molecules in which the polyethylene glycol can allow for
more
5 linker flexibility or can be used lengthen the linker.
03111 A linker can be a (maleimidocaproy1)-(valine-alanine)-(para-
aminobenzyloxycarbonyl) linker. A linker can be a (maleimidocaproy1)-(valine-
citrulline)-(para-aminobenzyloxycarbonyl) linker. A linker can be a
(maleimidocaproyl)-(phenylalanine-lysine)-(para-aminobenzyloxycarbonyl)
linker.
10 103121 A linker can also contain segments of alkylene, alkenylene,
alkynylene,
polyether, polyester, polyamide, polyamino acids, peptides, polypeptides,
cleavable
peptides, and/or aminobenzyl-carbamates. A linker can contain a maleimide at
one end
and an N-hydroxysuccinimidyl ester at the other end. A linker can contain a
lysine with
an N-terminal amine acetylated, and a valine-citrulline, valine-alanine or
phenylalanine-
15 lysine cleavage site. A linker can be a link created by a microbial
transglutaminase,
wherein the link can be created between an amine-containing moiety and a
moiety
engineered to contain glutamine as a result of the enzyme catalyzing a bond
formation
between the acyl group of a glutamine side chain and the primary amine of a
lysine
chain. A linker can contain a reactive primary amine. A linker can be a
Sortase A
20 linker. A Sortase A linker can be created by a Sortase A enzyme fusing
an LXPTG
recognition motif (SEQ 111) NO: 11) to an N-terminal GGG motif to regenerate a
native
amide bond. The linker created can therefore link to a moiety attached to the
LXPTG
recognition motif (SEQ ID NO: 11) with a moiety attached to the N-terminal COG

motif. A linker can be a link created between an unnatural amino acid on one
moiety
25 reacting with oxime bond that was formed by modifying a ketone group
with an
alkoxyamine on another moiety. A moiety can be part of a conjugate. A moiety
can be
part of an antibody, such as an antibody. A moiety can be part of an immune-
stimulatory compound, such as a myeloid cell agonist. A moiety can be part of
a
binding domain. A linker can be unsubstituted or substituted, for example,
with a
30 substituent. A substituent can include, for example, hydroxyl groups,
amino groups,
nitro groups, cyano groups, azido groups, carboxyl groups, carboxaldehyde
groups,
123
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
imine groups, alkyl groups, alkenyl groups, alkynyl groups, alkoxy groups,
acyl groups,
acyloxy groups, amide groups, and ester groups.
[0313] A linker can be polyvalent such that it covalently links more than one
immune-stimulatory compound to a single site on the antibody, or monovalent
such that
5 it covalently links a single immune-stimulatory compound to a single site
on the
antibody.
103141 Exemplary polyvalent linkers that may be used to attach many immune-
stimulatory compounds to an antibody of the conjugate are described. For
example,
Fleximer linker technology has the potential to enable high-DAR conjugate
with good
10 physicochemical properties. As shown below, the Fleximer linker
technology is
based on incorporating molecules into a solubilizing poly-acetal backbone via
a
sequence of ester bonds. The methodology renders highly-loaded conjugates (DAR
up
to 20) whilst maintaining good physicochemical properties. This methodology
can be
utilized with an immune-stimulatory compound as shown in the scheme below,
where
15 Drug' refers to the immune-stimulatory compound.
irrHOy -,TrOH OH)
to-oto
mtroT 0
40
n
add Fieximer loket o 0 HO
E
MN
1-ÃN FIN
0-Drug
O¨Dfug` 0-Ortig'
103151 To utilize the Fleximer linker technology depicted in the scheme
above, an aliphatic alcohol can be present or introduced into the immune-
stimulatory
compound. The alcohol moiety is then attached to an alanine moiety, which is
then
20 synthetically incorporated into the Fleximer linker. Liposomal
processing of the
conjugate in vitro releases the parent alcohol-containing drug.
[0316] By way of example and not limitation, some cleavable and noncleavable
linkers that may be included in the conjugates described herein are described
below.
124
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0317] Cleavable linkers can be cleavable in vitro and in vivo. Cleavable
linkers can include chemically or enzymatically unstable or degradable
linkages.
Cleavable linkers can rely on processes inside the cell to liberate an immune-
stimulatory compound, such as reduction in the cytoplasm, exposure to acidic
5 conditions in the lysosome, or cleavage by specific proteases or other
enzymes within
the cell. Cleavable linkers can incorporate one or more chemical bonds that
are
chemically or enzymatically cleavable while the remainder of the linker can be
non-
cleavable.
[0318] A linker can contain a chemically labile group such as hydrazone and/or
10 disulfide group. Linkers comprising chemically labile groups can exploit
differential
properties between the plasma and some cytoplasmic compartments. The
intracellular
conditions that can facilitate immune-stimulatory compound release for
hydrazine-
containing linkers can be the acidic environment of endosomes and lysosomes,
while
disulfide-containing linkers can be reduced in the cytosol, which can contain
high thiol
15 concentrations, e.g., glutathione. The plasma stability of a linker
containing a
chemically labile group can be increased by introducing steric hindrance using

substituents near the chemically labile group.
[0319] Acid-labile groups, such as hydrazones, can remain intact during
systemic circulation in the blood's neutral pH environment (pH 7.3-7.5) and
can
20 undergo hydrolysis and can release an immune-stimulatory compound once
the
conjugate is internalized into mildly acidic endosomal (pH 5.0-6.5) and
lysosomal (pH
4.5-5.0) compartments of the cell. This pH dependent release mechanism can be
associated with nonspecific release of the immune-stimulatory compound. To
increase
the stability of the hydrazone group of the linker, the linker can be varied
by chemical
25 modification, e.g., substitution, allowing tuning to achieve more
efficient release in the
lysosome with a minimized loss in circulation.
[0320] Hydrazone-containing linkers can contain additional cleavage sites,
such
as additional acid-labile cleavage sites and/or enzymatically labile cleavage
sites.
Conjugates including exemplary hydrazone-containing linkers can include, for
example,
30 the following structures:
125
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
(Ia) N "N
)1 0
0
(Ib) N.. =õfirw.).5_st
)1 0 Ab
0
0
(1."--)11.-NtAb
N
(Ic)
010
CH3
wherein D is an immune-stimulatory compound and Ab is an antibody,
respectively,
5 and n represents the number of compound-bound linkers (LP) bound to the
antibody. In
certain linkers, such as linker (Ia), the linker can comprise two cleavable
groups, a
disulfide and a hydrazone moiety. For such linkers, effective release of the
unmodified
free immune-stimulatory compound can require acidic pH or disulfide reduction
and
acidic pH. Linkers such as (Ib) and (Ic) can be effective with a single
hydrazone
10 cleavage site.
[0321] Other acid-labile groups that can be included in linkers include cis-
aconityl-containing linkers. cis-Aconityl chemistry can use a carboxylic acid
juxtaposed to an amide bond to accelerate amide hydrolysis under acidic
conditions.
103221 Cleavable linkers can also include a disulfide group. Disulfides can be
15 thermodynamically stable at physiological pH and can be designed to
release an
immune-stimulatory compound upon internalization inside cells, wherein the
cytosol
can provide a significantly more reducing environment compared to the
extracellular
environment. Scission of disulfide bonds can require the presence of a
cytoplasmic
thiol cofactor, such as (reduced) glutathione (GSH), such that disulfide-
containing
20 linkers can be reasonably stable in circulation, selectively releasing
the immune-
stimulatory compound in the cytosol. The intracellular enzyme protein
disulfide
isomerase, or similar enzymes capable of cleaving disulfide bonds, can also
contribute
to the preferential cleavage of disulfide bonds inside cells. GSH can be
present in cells
in the concentration range of 0.5-10 inM compared with a significantly lower
25 concentration of GSH or cysteine, the most abundant low-molecular weight
thiol, in
126
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
circulation at approximately 5 M. Tumor cells, where irregular blood flow can
lead to
a hypoxic state, can result in enhanced activity of reductive enzymes and
therefore even
higher glutathione concentrations. The in vivo stability of a disulfide-
containing linker
can be enhanced by chemical modification of the linker, e.g., use of steric
hindrance
5 adjacent to the disulfide bond.
[0323] Immune-stimulatory conjugates including disulfide-containing linkers
can include the following structures:
R R 0
ci))(S--S-..2r}LNI-Ab
(Ha) H
R R
n
(lib) D----------"S'S Ab
n
(tft)
R R
10 n
wherein D is an immune-stimulatory compound and Ab is an antibody,
respectively, n
represents the number of compounds bound to linkers bound to the antibody and
R is
independently selected at each occurrence from hydrogen or alkyl, for example.

Increasing steric hindrance adjacent to the disulfide bond can increase the
stability of
15 the linker. Structures such as (ha) and (IIc) can show increased in vivo
stability when
one or more R groups is selected from a lower alkyl such as methyl.
[0324] Another type of linker that can be used is a linker that is
specifically
cleaved by an enzyme. For example, the linker can be cleaved by a lysosomal
enzyme.
Such linkers can be peptide-based or can include peptidic regions that can act
as
20 substrates for enzymes. Peptide based linkers can be more stable in
plasma and
extracellular milieu than chemically labile linkers.
[0325] Peptide bonds can have good serum stability, as lysosomal proteolytic
enzymes can have very low activity in blood due to endogenous inhibitors and
the
unfavorable pH value of blood compared to lysosomes. Release of an immune-
25 stimulatory compound from an antibody can occur due to the action of
lysosomal
proteases, e.g., cathepsin and plasmin. These proteases can be present at
elevated levels
127
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
in certain tumor tissues. A linker can be cleavable by a lysosomal enzyme. The

lysosomal enzyme can be, for example, cathepsin B, cathepsin S, P-
glucuronidase, orp-
galactosidase.
103261 The cleavable peptide can be selected from tetrapeptides such as Gly-
5 Phe-Leu-Gly, Ala-Leu-Ala-Leu, dipeptides such as Val-Cit, Val-Ala, and
Phe-Lys, or
other peptides. Dipeptides can have lower hydrophobicity compared to longer
peptides,
depending on the composition of the peptide.
[0327] A variety of dipeptide-based cleavable linkers can be used in the
immune-stimulatory conjugates described herein.
10 [0328] Enzymatically cleavable linkers can include a self-
immolative spacer to
spatially separate the immune-stimulatory compound from the site of enzymatic
cleavage. The direct attachment of an immune-stimulatory compound to a peptide

linker can result in proteolytic release of the immune-stimulatory compound or
of an
amino acid adduct of the immune-stimulatory compound, thereby impairing its
activity.
15 The use of a self-immolative spacer can allow for the elimination of the
fully active,
chemically unmodified immune-stimulatory compound upon amide bond hydrolysis
[0329] One self-immolative spacer can be a bifunctional para-aminobenzyl
alcohol group (PABA), which can link to the peptide through the amino group,
forming
an amide bond, while amine containing immune-stimulatory compounds can be
20 attached through carbamate functionalities to the benzylic hydroxyl
group of the linker
(to give ap-amidobenzylcarbaniate, PA13C). The resulting pro-immune-
stimulatory
compound can be activated upon protease-mediated cleavage, leading to a 1,6-
elimination reaction releasing the unmodified immune-stimulatory compound,
carbon
dioxide, and remnants of the linker. The following scheme depicts the
fragmentation of
25 p- amidobenzyl carbamate and release of the immune-stimulatory compound:
0 so 0 X¨D protease Op 0
\,t(-13 1,6-eliminatior!
CO2
HN
+
peptide N H2
X-D
wherein X-D represents the unmodified immune-stimulatory compound and the
carbonyl group adjacent "peptide" is part of the peptide. Heterocyclic
variants of this
self-immolative group have also been described.
128
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0330] An enzymatically cleavable linker can be a f3-g,lucuronic acid-based
linker. Facile release of an immune-stimulatory compound can be realized
through
cleavage of the 13-glucuronide glycosidic bond by the lysosomal enzyme 13-
glucuronidase. This enzyme can be abundantly present within lysosomes and can
be
5 overexpressed in some tumor types, while the enzyme activity outside
cells can be low.
13-glucuronic acid-based linkers can be used to circumvent the tendency of an
immune-
stimulatory conjugate to undergo aggregation due to the hydrophilic nature of
B-
glucuronides. In certain embodiments, 13-glucuronic acid-based linkers can
link an
antibody to a hydrophobic immune-stimulatory compound. The following scheme
10 depicts the release of an immune-stimulatory compound (D) from an immune-

stimulatory conjugate containing a 13-glucuronic acid-based linker:
HO
HO
HO oCi .1D voucuronidase HO sap At).
1,6-elimination
HO
Op 1 CO2
HO 0
FiNyMkb
HNyAb
0
0
OH OH
wherein Ab indicates the antibody.
[0331] A variety of cleavablef3-glucuronic acid-based linkers useful for
linking
15 drugs such as auristatins, camptothecin and doxorubicin analogues, CBI
minor-groove
binders, and psymberin to antibodies have been described. These it-glucuronic
acid-
based linkers may be used in the conjugates described herein. In certain
embodiments,
the enzymatically cleavable linker is a13-galactoside-based linker. I3-
Galactoside is
present abundantly within lysosomes, while the enzyme activity outside cells
is low.
20 [0332] Additionally, immune-stimulatory compounds containing a
phenol group
can be covalendy bonded to a linker through the phenolic oxygen. One such
linker
relies on a methodology in which a diamino-ethane "Space Link" is used in
conjunction
with traditional "PABO"-based self-immolative groups to deliver phenols.
[0333] Cleavable linkers can include non-cleavable portions or segments,
and/or
25 cleavable segments or portions can be included in an otherwise non-
cleavable linker to
render it cleavable. By way of example only, polyethylene glycol (PEG) and
related
polymers can include cleavable groups in the polymer backbone. For example, a
129
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
polyethylene glycol or polymer linker can include one or more cleavable groups
such as
a disulfide, a hydrazone or a dipeptide.
[0334] Other degradable linkages that can be included in linkers can include
ester linkages formed by the reaction of PEG carboxylic acids or activated PEG
5 carboxylic acids with alcohol groups on an immune-stimulatory compound,
wherein
such ester groups can hydrolyze under physiological conditions to release the
immune-
stimulatory compound. Hydrolytically degradable linkages can include, but are
not
limited to, carbonate linkages; imine linkages resulting from reaction of an
amine and
an aldehyde; phosphate ester linkages formed by reacting an alcohol with a
phosphate
10 group; acetal linkages that are the reaction product of an aldehyde and
an alcohol;
orthoester linkages that are the reaction product of a formate and an alcohol;
and
oligonucleotide linkages formed by a phosphoramidite group, including but not
limited
to, at the end of a polymer, and a 5' hydroxyl group of an oligonucleotide.
[0335] A linker can contain an enzymatically cleavable peptide moiety, for
15 example, a linker comprising structural formula (Ma), (Mb), (Inc), or
(Md):
RY
is q 0
011a)
N T
peptide..õN
y _ X
RY 0
0
OAA
Lpeptide-õN 101
Fe
RY
0
0
ii-TiLpeptide"...N
RY
0
Rz 0
(Ind) It q
Or s'1/4-
TeAThepticle....N
20 or a pharmaceutically acceptable salt thereof, wherein: "peptide"
represents a peptide
(illustrated in N¨>C orientation, wherein peptide includes the amino and
carboxy
"termini") that is cleavable by a lysosoma1 enzyme; T represents a polymer
comprising
one or more ethylene glycol units or an alkylene chain, or combinations
thereof; Ra is
130
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
selected from hydrogen, alkyl, sulfonate and methyl sulfonate; KY is hydrogen
or C1-4
alkyl-(0)14C14 alkylene)s-GI or C14 alkyl-(N)-[(C14 alkylene)-G12; Rz is C14
alkyl-
(0)r-(Cr-4 allcylene)s-G2; GI is SO3H, CO2H, PEG 4-32, or a sugar moiety; G2
is SO3H,
CO2H, or a PEG 4-32 moiety; r is 0 or I; s is 0 or 1; p is an integer ranging
from 0 to 5;
5 q is 0 or 1; xis 0 or 1; y is 0 or 1; 1 represents the point of
attachment of the linker to
an immune-stimulatory compound; and * represents the point of attachment to
the
remainder of the linker.
103361 In certain embodiments, the peptide can be selected from natural amino
acids, unnatural amino acids or combinations thereof In certain embodiments,
the
10 peptide can be selected from a tripeptide or a dipeptide. In particular
embodiments, the
dipeptide can comprise L-amino acids and be selected from Val-Cit; Cit-Val;
Ala-Ala;
Ma-Cit; Cit-Ma; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser;
Lys-
Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ma-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys;
Lys-Val;
Ma-Lys; Lys-Ala; Phe-Cit; Cit-Phe; Leu- Cit; Cit-Leu; Ile-Cit, Cit-Ile; Phe-
Arg; Mg-
15 Phe; Cit-Trp; and Trp-Cit, or salts thereof.
103371 Exemplary embodiments of linkers according to structural formula (IIIa)
are illustrated below (as illustrated, the linkers include a reactive group
suitable for
covalently linking the linker to an antibody):
0
0 0
0 H 111) 01 CAA
(IIIa.1) trk N 0
0 -N-jcieN'''!"."14
H
H
HN
1-12W-LO
0
0 0
0 0
N
411
H 0 E H
-
20 0
0
XicatNH
H (f? S0--11A
N
(IIIa.3)
etWir ---fe-11/4%N
- H
H
0 0 :-
....803H 0 -
131
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
0
(IIIa.4) CKA
H
H
0
0 0
0 0'1/4
(IIIa.5) õõ)OcNir:uL 101
-
N
H 0 E H
( NH2
N
0
NH
(IIIa.6)
_
H
0
0 cNH2
NA-0
0
0 0
0 0A1(
* (IIIa.7)
_ N
H 0 _ H
NH2
L.NA0
0
0 0
H o011
_ H
0
0
( ONE
N
wherein -04" indicates an attachment site of a linker to an immune-stimulatory

compound.
103381 Exemplary embodiments of linkers according to structural formula
(11%), (Mc), or (1114) that can be included in the conjugates described herein
can
include the linkers illustrated below (as illustrated, the linkers can include
a reactive
group suitable for covalendy linking the linker to an antibody):
132
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
0 0 0 ON
H
(Mb. 1) t_LNAHNorNYLIFµil 0
a
( NH2
NAO
H
0
0
(IIIb.2)
crc.........õ.õ,,,,A0 N
- isir.. 1/4õ,N 0
....)
0 ON-
O
H i H
0( x2
N 0
H
0
0
ON 0 H 0
(IIIb.3) cr.õ.........---...õ...--j--, Xr. N
.,....õ1,_ N 0
O H 0 = H
0
O 0
1.4 0 ON
iq......A 41110
(IIIb.4) seCAN
E N
" "
o
o c NH2
NO
H
NH2
0
O 0 4 0 410 ON
H
(IIIb.5)
\ H 0
H
0
( NH2
140
H
0
0 0 0
0'11A
(IIIb.6) " 0 =
0
133
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H
H2N .....e.. N
I I
0
H.......A
N
(IIIb.7) tze---"---AN H 0 _ N
\ z
H
0 (
X-I2
N 0
H
0
0
ON 0
0
(IIIb.8) cirl)LicrilljL SI
-
N
H _ H
0 CX
0 OH
0
0 OH
0 H 0
ON
(IIIb.9) ..,..KN
- cr .........õ----...õ Sy N jib..
N 410
H
H .7.
0 0 c x2
N 0
H
H2N 0
0
OejV 0
H 0
(IIIb.10)
cr..........--..õ...-..........X N N '....A-N le
0
H 0 - H
C.- X2
N 0
H
0
cr............õ........A0
)criii.......AN 0
ON 0 0
(IIIb.11)
= H 0
H
SO3H ' n C NH2
N AO
H
0
criniA0 i)icriiN
0 0 410 ON
(IIIb.12)
H i
H
0
803H 0 c NH2
N AO
H
134
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
0
CO2H
0
H 0 ON
(IIIb.13)
N
O
H 0 E H
1-12
N 0
0
0
(Illb.14)
N)cr'r;LAN
0'
H 0 E H
X2
N 0
0
o
H vj 4110
(IIIb.15)
-
N
o
= H
E H
0
SO3H
C NH2
0
N
0
O
I I
(IIIb.16) NILO 0
H
0 0
N N
H
0
0
OH
HO 7 CO2H
Hot.
(IIIb.17)
NILO 10 0
0 0
NLH
NrNAJ
HI
H
0 0
135
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
HO3S.L.,....0
0
(HIM 8) 1/2110 IS 0
H -%--'',
0 0
Nili.N.,..õ---...N.A.õ---...õ.."........)?
H
II H
0
0
OH
HO,,..
0 H 0
t
H 0 CO2H
(IIIb.19)
itirµk-Ari-1-11-N
\ 0 ......z..., 0 41111 0.1,µ
0
0
0
0
0 0 0 ON
H
(IIk. 1 )
cri....õ......Ø--..õ..Ø...}...X.rN.."...
.
N
H 0 _ H
(--
0
NH2
IA AO
H
0
el?
H
H2N N.,.
0 ....)
......1/4 0
HN 0
(IIIc.2)
11101 0
N 3/4"ICH N
H
yff,0
0
0
H
H2N,..e..N
Oil ) 0
0 0
(IIIc.3)
is
fr.0
0
0
0 OMe
136
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
0 0
H
0
(IIIc.4)
0
H 02C
0
-1/2L0
0
N j, lir N
NH
0 0
0
ish
HO
HO2C t = .
0 e NH z.--
,,7\70 e-c 0
0 HN/
NH N
/ 0
6
o 0
o sen 0)IA
N
N
0
_ H
o r NH2
N
-fro
ono
(IIId. I) 401 17
- CI).-
Oy.1
)-11
0
0 0
137
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
-fro
0 H2N..f0
NH n
(IIId.2) 0 I
_ 0 O

yi
H
1-
0)
H 1 1
0 0
Nn
0.1)
H
CI 11
()y) 0110
IIId.3) 0
0 24
0
o
.0H
-%
Ho2c : OH
OH
H2N---fo
(NH
ono
N
2 0
H H
(IIId.4) 00
H
frO 0
0
0
wherein sr"' indicates an attachment site to an immune-stimulatory compound.
103391 The linker can contain an enzymatically cleavable sugar moiety, for
example, a linker comprising structural formula (WO, (IVb), (IVO, (IVA or
(IVe):
0 i
--- xi o
(IVa) N 0i
H AOH
0 -
H02C z OH
6H
138
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
OH
HO2Cõ, µõ riOHi%
0
(IVb) - OH
0 b
q *X1 4- *
(IVC) 0
AOH
HO2C : OH
OH
OH
H02Cõ,(3 õ,OH
0
(IVd) _ OH
0 6
AL-0 -re-' q 410
)0
-...*
*
0 I
40 xi0
(IVe) Wit
H r OH
0 "µ
OH
OH OH
5 or a pharmaceutically acceptable salt thereof, wherein: q is 0 or 1; r is
0 or 1; X' is CH2,
0 or NH; 'represents the point of attachment of the linker to an immune-
stimulatory
compound; and * represents the point of attachment to the remainder of the
linker
103401 Exemplary embodiments of linkers according to structural formula (IVa)
that may be included in the immune-stimulatory conjugates described herein can
10 include the linkers illustrated below (as illustrated, the linkers
include a group suitable
for covalently linking the linker to an antibody):
139
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
-frO
0
--...,
0
(IVa.1) 0 Nyt...,..õ..,__.
yLe.õ.õ,..õ,õ j?
H N
Ho2c,(07 0
HO'. 41`0H
OH
-le
0
-.....
0
0
(IVa.2)
. NA-#"--"N'IL)13
H
H /
HO2C 0 0
HON 0H V.1i 0
OH
Ot
0
0
0
(IVa.3)
* ,--,..,-0-...õ-----.N..A._õ---...õ...---,,,,h11?
0
H
HO2C4670yNH 0
HO . ..10H
OH
frO
0
110 0 0
0
(IVa.4)
N'jl-'-'--MI)L''e--"---rl?\
HO2CtrOTO H H
0
HO'. -e'OH
OH
140
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0 al
0
NA0 0
N
------%"N"A 7-----0N Nr.,,,.1.-?
(IVa.5)
H H k -H
HO2C 0 0
0 0
H 090 H
OH
fro
0
(IVa.6) 0 0 N
NA__t
____.N ?
H H
HO2C 00
0
HO'lty
.110H
OH
lir.0
0
0
0
0
(IVa.7) is
74?
NA--------N-JO
H H N
HO2C 0 0
0
H:gitH
OH
fr0
0 0
0
(IVa.8) 0
m 0
..s.--t.t.k.,
H H
HO2C 0 0
8
HOVOH
OH
0 0 0 0
R j

NA------%NA------------N
(IVa.9) H H 0
HO2C 0 0
HOVOH
OH
141
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Sir-0
0 ....,
0
(IVa.10)
H H
H 02C 0 0
00
HO''' t H
V'e
OH
Sr0
0 -....õ
HO3S H 0
I.
N
(IVa.11) 101
ins
NA"-----44%N"--%0 0
HO2C47 H H
HO"( '"OH
OH
frO
0
0
--õ, H037
H
iy3R\
(IVa.12) 0 Ciii
0
0
N --i4Nr 0
HO2C(02#0 H
H
HO'µ. d"OH
OH
,
wherein sir' represents the point of attachment of a linker to an immune-
stimulatory.
103411 Exemplary embodiments of linkers according to structural formula (IVb)
that may be included in the conjugates described herein include the linkers
illustrated
below (as illustrated, the linkers include a group suitable for covalently
linking the
linker to an antibody).
Sr0
0
(IVb.1)
Ho2ci070 0
HO"( "OH
0
H
OH 0..õ..----Ø-----,..N.N).5
0
/
0
142
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
--sro
o
HO2C 0 0
(IVb.2)
0
HOVOHe
H
OH 0=01µ1)(j1?
0
0
1f0
0
HO2C 0 0
(IVb.3)
HO:VOHS
HO3S 0 0
OH 0,...--,.,_,NH
H
0
0
-frO
0
HO2CiOt 0
HO3S 0
(IVb.4)
HO'''ritH
Hyt ,õ)?
OH
N
0-,----...o,.....õ..N
H
0 0
-frO
0
HO2C 4,70 0
(IVb.5)
HO'. .#'0H
0 0
OH
NA--------)5
H
/
0
yt0
0
(IVb.6)
HO2C 000 is
HOC(../H
H
OH0,.--,,aõ--,,a,,-,,,N
oil?
0
0
143
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
(IVb.7) HO2CiOrTO
0
HO . .40H SLII?
OH
0
0
(IVb.8)
HO2CirOt *
0
'''OH
OH
/1
Aro
0
HO2C.õ1/4(01Dro0
(IVb.9)
HO"0101
4"/OH
0
OH
0
to
0
HO 0
(IVb.10) 2C=CLI.- µµµ
HO _ OH
0
OH
0
0
5 wherein isid represents the point of attachment of a linker to an immune-
stimulatory
compound.
103421 Exemplary embodiments of linkers according to structural formula (IVc)
that may be included in the conjugates described herein include the linkers
illustrated
below (as illustrated, the linkers include a group suitable for covalently
linking the
10 linker to an antibody).
144
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
OH
,,,A
HOD

H
0 0 CO2H
0
0
(IVc.1)
1.1 ir`-e '''IrrIL--,'"%'-'11?
H
0
0
Co
OH
HO,n ,. .00H
0 0 CO2H
0 0
(IVc.2)
1111
H
i
0
0
)<LO
OH
..
Ho, ,,,On H
0 0 CO2H
0
0
(IVc.3)
II ON)7%1-?
H
0
0
ICLO
OH
oria .,,OH
---4.,,
0 0 CO2H
(IVc.4) 0 H
0
0 cf-s- II ii P=Lire
j
o H 0 3 S 0
0
XL0
145
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
OH
HO.õ OH n.,µ
0 0 .02H
0
(Ivc.,) loi 0,0 )5, H -
1---11? --'-'.-.-N 1 1
H
0 HO3S 0 0
OH
H0H0.8..,µOH
18
0 CO2H
(IVc.6) 0 H
0
le Or."---AX---------- vi ill N1/21C-r------Thsfj,
0 HO3S 0
0
)<L0
OH
HO,,. OH
n,,,
0 0 CO2H
(IVc.7) 0 H
0
I. (:)......õ..-0...,..---...eN
x.iremji
0 H038
0
"CLO
OH
A
0 0 .02H
0
(ive.8)
is
H
0
0
NLO
146
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
OH
HO,õ .,NOH
n0 0 CO2H
(IVc.9) --
a%µ""--.%-t0 H 0
. 0iliLiN,y-wiji
0
0 HO3S
0
XL0
OH
HOF,. 0H
A.0
0 0 CO2H
0
0
(IVc.10)
le
H 0
0
)(LO
OH
HO,,. OH
A,,,,
0 0 c02H
(Iwen)
* 0 H
N NyWNJ5
0 H ) 0
i
HO3S
0
wherein sir' represents the point of attachment of a linker to an immune-
stimulatory
compound.
[0343] Exemplary embodiments of linkers according to structural formula (IVd)
that may be included in the conjugates described herein include the linkers
illustrated
below (as illustrated, the linkers include a group suitable for covalently
linking the
linker to an antibody):
cr0 Li
OH
si HO,,.
.00H
------Thr
0 0 CO2H
(IVd.1) ell 0 0
0

147
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
OH
c--CmiH
N õ,%se.---....0 0
HOAOH
(IVd.2) 0 0
0 0 CO2H
0
XL0
0
OH
H
(IVd.3) cr..õ..õ..---T.N..õ..--,0,--..õ..0õ-----
....0 0 HOAOH
0 0
0 0 CO2H
0
OH
n
Ha,.
.,\OH rE11 -----------0"--------a"-------0
SO (IVd.4) NH 0 0 CO2H
OA-a-el 0
Co
0 0 OH
0
HOAOH
ttr-""--AN----1- -------0
. .
(IVd.5) \ H
0
0
0 0 CO2H
0
XL0
0 OH
H
or N...........--.0,----....õ,0,...e.----t
ati HO,,,. .t.OHO2
(IVd.6) \ 0
0
0 0 CH
0
5
X 0
,
wherein siss' represents the point of attachment of a linker to an immune-
stimulatory
compound.
103441 Exemplary embodiments of linkers according to structural formula (lye)
10 that may be included in the conjugates described herein include the
linkers illustrated
below (as illustrated, the linkers include a group suitable for coyalently
linking the
linker to an antibody):
148
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
frO
0
0
(IVe.1) 0 OH
lix070 (1101 0 0
HO etIOH
OH
0
0 0
0 0
(IVe.2) OH 101
N N
0
4
H
00
0
HO3S 16
HOLipx '2.''OH
OH
wherein stra represents the point of attachment of a linker to an immune-
stimulatory
compound.
[0345] Although cleavable linkers can provide certain advantages, the linkers
comprising the conjugate described herein need not be cleavable. For non-
cleavable
linkers, the immune-stimulatory compound release may not depend on the
differential
properties between the plasma and some cytoplasmic compartments. The release
of the
immune-stimulatory compound can occur after internalization of the immune-
stimulatory conjugate via antigen-mediated endocytosis and delivery to
lysosomal
compartment, where the antibody can be degraded to the level of amino acids
through
intracellular proteolytic degradation. This process can release an immune-
stimulatory
compound derivative, which is formed by the immune-stimulatory compound, the
linker, and the amino acid residue or residues to which the linker was
covalently
attached. The immune-stimulatory compound derivative from immune-stimulatory
conjugates with non-cleavable linkers can be more hydrophilic and less
membrane
permeable, which can lead to less bystander effects and less nonspecific
toxicities
compared to immune-stimulatory conjugates with a cleavable linker. Immune-
stimulatory conjugates with non-cleavable linkers can have greater stability
in
149
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
circulation than immune-stimulatory conjugates with cleavable linkers. Non-
cleavable
linkers can include alkylene chains, or can be polymeric, such as, for
example, based
upon polyalkylene glycol polymers, amide polymers, or can include segments of
alkylene chains, polyalkylene glycols and/or amide polymers. The linker can
contain a
5 polyethylene glycol segment having from 1 to 6 ethylene glycol units.
[0346] The linker can be non-cleavable in vivo, for example, a linker
according
to the formulations below:
0
0
(Va) 1/21LHOto>""a"--- N Rx
0-9
0
(Vb)
0-7
0-9
0 0
(W) NAILrN A(4 Rx
0-9 H 0-9
0
(Vd) Rx
1/2Y140-8
0
(Ye) -9 N
A H Rx
C1191-0-9
)0Laer,
frekilett: N
(Vf) RX
or salts thereof, wherein: Ra is selected from hydrogen, alkyl, sulfonate and
methyl
15 sulfonate; it' is a reactive moiety including a functional group capable
of covalently
linking the linker to an antibody; and ,041# represents the point of
attachment of the linker
to an immune-stimulatory compound.
[0347] Exemplary embodiments of linkers according to structural formula (Va)-
(Vf) that may be included in the conjugates described herein include the
linkers
20 illustrated below (as illustrated, the linkers include a group suitable
for covalently
linking the linker to an antibody, and =Pri" represents the point of
attachment of the linker
to an immune-stimulatory compound:
150
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0
0 0
(Va.1)
0
0
(Vc.1)
=1/2-1Le
0
(Vc.2)
0
0
(Vd.1)
0
0
0
(Vd.2)
5 SO3H 0
0 O.,
(Vd.3)
j,Lr01,1?
0
(Vd.4)
SO3H 0
0
0
H TOTAN
We.!)
0
103481 Attachment groups that are used to attach the linkers to an antibody
can
10 be electrophilic in nature and include, for example, maleimide groups,
alkynes,
alkynoates, allenes and allenoates, activated disulfides, active esters such
as NHS esters
and HOBt esters, haloformates, acid halides, alkyl, and benzyl halides such as

haloacetamides. There are also emerging technologies related to "self-
stabilizing"
maleimides and "bridging disulfides" that can be used in accordance with the
15 disclosure.
103491 Maleimide groups are frequently used in the preparation of conjugates
because of their specificity for reacting with thiol groups of, for example,
cysteine
groups of the antibody of a conjugate. The reaction between a thiol group of
an
antibody and a drug with a linker including a maleimide group proceeds
according to
151
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
the following scheme:
O 0
(
e..
Antibod
..-S
SH Antibodyy-- 1-
NTh
¨I.-
%.....4NTh
Linking Group
Linking Group
O
/ 0 /
Drug
Drug
[0350] The reverse reaction leading to maleimide elimination from a thio-
substituted succinimide may also take place. This reverse reaction is
undesirable as the
5 maleimide group may subsequently react with another available thiol group
such as
other proteins in the body having available cysteines. Accordingly, the
reverse reaction
can undermine the specificity of a conjugate. One method of preventing the
reverse
reaction is to incorporate a basic group into the linking group shown in the
scheme
above. Without wishing to be bound by theory, the presence of the basic group
may
10 increase the nucleophilicity of nearby water molecules to promote ring-
opening
hydrolysis of the succinimide group. The hydrolyzed form of the attachment
group is
resistant to deconjugation in the presence of plasma proteins. So-called "self-

stabilizing" linkers provide conjugates with improved stability. A
representative
schematic is shown below:
Drug S
AntibodyeSH +
Antibody--
¨N.- %.1N¨KDrug
: Base !Base
0
0
O 0
¨(
Ant body--S Drug ¨
_Sl Drug ...- Antibodyy(--
0
HN¨<
-Base
: Base+
O 1
H
:0-
1
H
.
[0351] The hydrolysis reaction schematically represented above may occur at
either carbonyl group of the succinimide group. Accordingly, two possible
isomers
may result, as shown below:
152
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
o
HO
Antibody"-4Drug
Antibody-3"r
Drug
0
HN¨K
HN¨(
Base
Base
Base+
OH
0
103521 The identity of the base as well as the distance between the base and
the
maleimide group can be modified to tune the rate of hydrolysis of the thio-
substituted
succinimide group and optimize the delivery of a conjugate to a target by, for
example,
5 improving the specificity and stability of the conjugate.
103531 Bases suitable for inclusion in a linker described herein, e.g., any
linker
described herein with a maleimide group prior to conjugating to an antibody,
may
facilitate hydrolysis of a nearby succinimide group formed after conjugation
of the
antibody to the linker. Bases may include, for example, amines (e.g., -
N(R26)(R27),
10 where R26 and R27 are independently selected from H and C1-6 alkyl),
nitrogen-
containing heterocycles (e.g., a 3- to 12-membered heterocycle including one
or more
nitrogen atoms and optionally one or more double bonds), amidines, guanidines,
and
carbocycles or heterocycles substituted with one or more amine groups (e.g., a
3- to 12-
membered aromatic or non-aromatic cycle optionally including a heteroatom such
as a
15 nitrogen atom and substituted with one or more amines of the type -
N(R26)(R27), where
R26 and n27
are independently selected from H or C1-6 alkyl). A basic unit may be
separated from a maleimide group by, for example, an alkylene chain of the
form
¨(CH2)0r, where m is an integer from 0 to 10. An alkylene chain may be
optionally
substituted with other functional groups as described herein.
20 103541 A linker described herein with a maleimide group may
include an
electron withdrawing group such as, but not limited to, -C(0)R, =0, -CN, -NO2,
-CX3,
-X, -COOR, -CONR2, -COR, -COX, -SO2R, -S020R, -SO2NFIR, -SO2NR2, P03R2,
-P(0)(CH3)NITR, -NO, -NR.3 , -CR=CR2, and -CCR, where each R is independently
selected from H and C I-6 alkyl and each X is independently selected from F,
Br, Cl, and
25 I. Self-stabilizing linkers may also include aryl, e.g., phenyl, or
heteroaryl, e.g.,
pyridine, groups optionally substituted with electron withdrawing groups such
as those
described herein.
153
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
[0355] Examples of self-stabilizing linkers are provided in, e.g., U.S. Patent

Publication Number 2013/0309256, the linkers of which are incorporated by
reference
herein. It will be understood that a self-stabilizing linker useful in
conjunction with
immune-stimulatory compounds may be equivalently described as unsubstituted
5 maleimide-including linkers, thio-substituted succinimide-including
linkers, or
hydrolyzed, ring-opened thio-substituted succinimide-including linkers.
[0356] In certain embodiments, a linker comprises a stabilizing linker moiety
selected from.
HOH0
0 0
"bbct NLOHN*1%0H
0 0
¨rC) oiyN 0
0
0
0 0 0 K 40
0
c tie
N H2
NH
0
N H2 =
10 [0357] In the scheme provided above, the bottom structure may be
referred to as
(maleimido)-DPR-Val-Cit-PAB, where DPR refers to diaminopropinoic acid, Val
refers
to valine, Cit refers to citrulline, and PAB refers to para-
aminobenzylcarbonyl. 4-re"
represents the point of attachment to an immune-stimulatory compound.
103581 A method for bridging a pair of sulfhydryl groups derived from
15 reduction of a native hinge disulfide bond has been disclosed and is
depicted in the
schematic below. An advantage of this methodology is the ability to synthesize

homogenous DAR4 conjugates by full reduction of IgGs (to give 4 pairs of
sulfhydryls
from interchain disulfides) followed by reaction with 4 equivalents of the
alkylating
agent. Conjugates containing "bridged disulfides" are also claimed to have
increased
20 stability.
154
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
reduoe disulfide 1
-4"
T
it

\ - 6¨SH HS¨.0 .
= SH
0
0
C1/4 \--ii.
S NA
________________________________________________________________ A1025
9
op
0
1
ors HA
H in situ elimination
' [
4k 11µ __________________ , * 11
soSO2 0 o
0
o
_____________________________________ . : s att NA
W "
de 0
"bridged disulfide"
[0359] Similarly, as depicted below, a maleimide derivative that is capable of

bridging a pair of sulthydryl groups has been developed.
S
:
41
_______________________________________________________________________________
_ .
0
NO

[0360] A linker can contain the following structural formulas (Via), (VIb), or

(Vic):
0
0
VI
(VIa) 0
la
Rq
0
crirr*
(VIb) 0 ( )y 0
tj \
N,
"
G2
155
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
0
cfrNO 0, *
(Vic) 0
Rw
or salts thereof, wherein: Rq is H or-0-(CH2CH20)ii-CH3; x is 0 or 1; y is 0
or 1; G2 is
-CH2CH2CH2S03H or¨CH2CH20-(CH2CH20)11-CH3; Rw is¨O-CH2CH2S03H or
¨NH(C0)-CH2CH20-(CH2CH20)12-CH3; and * represents the point of attachment to
the remainder of the linker.
103611 Exemplary embodiments of linkers according to structural formula (VIa)
and (V1b) that can be included in the conjugates described herein can include
the linkers
illustrated below (as illustrated, the linkers can include a group suitable
for covalently
linking the linker to an antibody):
OH
HO2C11, att0H
a%
_
1 1
(VIa.1)
110
0 op 1:1 H III- 0
'cc
H i H
OH
HO. .00H
,,n
0 0 C 0H
(VIa.2) 2 0
? 0 0
)4.?
X% N
H
0
ii
156
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
\-o)
N
(VIa.3) 1
H2N
H H

N yezizrity:
yre0 0
0
0
0
40 A-0 0 _de
H.7- 0
7
N
(VIa.4) frO Ili 0 H
0
0
0
00H
0 =
HO2C , OH
OH
0/0)¨
/11
r
N N
( Vth .1 )
H2N
N:14 1 11?
- 0
H
rsil 0
Sr0= 0 0
0
01(¨P t
0 N N
(VIb.2)
H2N
Netk...1?
0 -
F H
0
Ny,
fro 0
0
0
157
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
4/
_______________________________________________________________________________
_______ /803H
C
N
Kisi I si,si?
= 0
H 11
0
(VIb.3) is Ni---..N
H
0
0
0 ,,,OH
HO2C : OH
OH
SO3H
/
(
N
Niõ IN)?
N
N
H H
0
yr
401 Nyc....HN Ny-
(VIb.4) 0=0
0
0
µ,0H
0 .
HO2C , OH
OH
act /0 i
CO
N
N:.
i.."?.
1
N
r 0
H I 1
0
(VIb.6)
H
lir-0 101 0 0
0
NOH
0 '
HO2C : OH
OH
158
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
pot
r0
N 0
N
N
z 0
H
(VIIb.7) "T.0 IP 0
0
0
0 skOH
HO2C , OH
OH
,N,
1
N N ri - --N,....---L--"--4-
¨/
\ ii
= 0
H
0
H
(VIIb.8) --l.r.0 IP
0
0
.0H
HO2C , OH
OH
,
wherein sirs' represents the point of attachment of the linker to an immune-
stimulatory
compound.
103621 Exemplary embodiments of linkers according to structural formula (VIc)
that can be included in the immune-stimulatory conjugates described herein can
include
the linkers illustrated below (as illustrated, the linkers can include a group
suitable for
covalently linking the linker to an antibody).
z 0
=.12>
-Ari3O IP 0
0
(Vic.1)
0
(0 0
0 .00H
H038)
HO2C : OH
OH
159
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
= 0
II ! y. $ c 3)
ICN
'"ye'N
H
N I
(VIc.2) frO al 0
0
0 I I (:)4
(0 0
HO3S)
-(
16
= 0
H i
III 00
(VIc.3)
frO IP
0
(0 0
/
H 03S)
H
H2N ,,..e.. N .1
I I
1---- 0 0
00
(VIc.4) ).(ii.0 µtir---N
H
=NH 0 7
_______________________________________________________________________________
_____ rlircro N)
0
(õ0 0
H 03S)
H
00
(VIc.5) lii0 so
a 0 Nr(7.5oN
(
0
HO
0
0
HO2C
ale-g "OH
)
..-: HO3S
OH
H ! iL H
00
(VIc.6) yro SI
0
07NH
0
%OH
0 ',
0---.
1
I-102C : OH
0-cr" or
1 1
OH
wherein ire' represents the point of attachment of the linker to an immune-
stimulatory
compound.
160
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
103631 A linker can be attached to an antibody at any suitable position.
Factors
to be considered in selecting an attachment site include whether the linker is
cleavable
or non-cleavable, the reactive group of the linker for attachment to the
antibody, the
chemical nature of the immune-stimulatory compound and compatabiltity with
reactive
5 sites on the linker and the antibody, and the effect of the attachment
site on functional
activities of the Fc domain. A linker may be attached to a terminus of an
amino acid
sequence of an antibody or can be attached to a side chain of an amino acid of
an
antibody, such as the side chain of a lysine, serine, threonine, cysteine,
tyrosine,
aspartic acid, glutamine, a non-natural amino acid residue, or glutarnic acid
residue. A
10 linker may be bound to a terminus of an amino acid sequence of an Fc
domain or Fc
region of an antibody, or may be bound to a side chain of an amino acid of an
Fc
domain of an antibody, such as the side chain of a lysine, setine, threonine,
cysteine,
tyrosine, aspartic acid, glutamine, a non-natural amino acid residue, or
glutamic acid
residue.
15 103641 In some embodiments, a linker is attached to a hinge
cysteine of an
antibody Fc domain. A linker can be attached to an antibody at a light chain
constant
domain lysine. A linker can be attached to an antibody at an engineered
cysteine in the
light chain. A linker can be attached to an antibody at an engineered light
chain
glutamine. A linker can be attached to an antibody at an unnatural amino acid
20 engineered into the light chain. A linker can be attached to an antibody
at a heavy chain
constant domain lysine A linker can be attached to an antibody at an
engineered
cysteine in the heavy chain. A linker can be attached to an antibody at an
engineered
heavy chain glutamine. A linker can be attached to an antibody at an unnatural
amino
acid engineered into the heavy chain. Amino acids can be engineered into an
amino
25 acid sequence of an antibody as described herein or as known to the
skilled artisan and
can be connected to a linker of a conjugate. Engineered amino acids can be
added to a
sequence of existing amino acids. Engineered amino acids can be substituted
for one or
more existing amino acids of a sequence of amino acids.
103651 A linker can be attached to an antibody via a sulfhydryl group. A
linker
30 can be attached to an antibody via a primary amine. A linker can be a
link created
between an unnatural amino acid on an antibody by reacting with oxime bond
that was
161
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
formed by modifying a ketone group with an alkoxyamine on an immune
stimulatory
compound.
103661 As is known by skilled artisans, the linker selected for a particular
conjugate may be influenced by a variety of factors, including but not limited
to, the
5 site of attachment to the antibody (e.g., lys, cys or other amino acid
residues), structural
constraints of the drug pharmacophore and the lipophilicity of the drug. The
specific
linker selected for a conjugate should seek to balance these different factors
for the
specific antibody/drug combination.
103671 For example, conjugates have been observed to effect killing of
10 bystander antigen-negative cells present in the vicinity of the antigen-
positive tumor
cells. The mechanism of bystander cell killing by conjugates has indicated
that
metabolic products formed during intracellular processing of the conjugates
may play a
role. Neutral cytotoxic metabolites generated by metabolism of the conjugates
in
antigen-positive cells appear to play a role in bystander cell killing while
charged
15 metabolites may be prevented from diffusing across the membrane into the
medium, or
from the medium across the membrane, and therefore cannot affect bystander
killing
In certain embodiments, the linker is selected to attenuate the bystander
effect caused
by cellular metabolites of the conjugate. In certain embodiments, the linker
is selected
to increase the bystander effect.
20 103681 The properties of the linker, or linker-compound, may also
impact
aggregation of the conjugate under conditions of use and/or storage.
Typically,
conjugates reported in the literature contain no more than 3-4 drug molecules
per
antibody molecule. Attempts to obtain higher drug-to-antibody ratios ("DAR")
often
failed, particularly if both the drug and the linker were hydrophobic, due to
aggregation
25 of the conjugate. In many instances, DARs higher than 3-4 could be
beneficial as a
means of increasing potency. In instances where an immune-stimulatory compound
is
more hydrophobic in nature, it may be desirable to select linkers that are
relatively
hydrophilic as a means of reducing conjugate aggregation, especially in
instances where
DARs greater than 3-4 are desired. Thus, in certain embodiments, a linker
incorporates
30 chemical moieties that reduce aggregation of the conjugates during
storage and/or use.
A linker may incorporate polar or hydrophilic groups such as charged groups or
groups
162
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
that become charged under physiological pH to reduce the aggregation of the
conjugates. For example, a linker may incorporate charged groups such as salts
or
groups that deprotonate, e.g., carboxylates, or protonate, e.g., amines, at
physiological
pH.
5 [0369] In particular embodiments, the aggregation of the
conjugates during
storage or use is less than about 40% as determined by size-exclusion
chromatography
(SEC). In particular embodiments, the aggregation of the conjugates during
storage or
use is less than 35%, such as less than about 30%, such as less than about
25%, such as
less than about 20%, such as less than about 15%, such as less than about 10%,
such as
10 less than about 5%, such as less than about 4%, or even less, as
determined by size-
exclusion chromatography (SEC).
Conjugates
[0370] A conjugate as described herein comprises an antibody and at least one
linker attached to at least one immune-stimulatory compound, such as a myeloid
cell
15 agonist or other agonist TLR8 agonist, TLR7 agonist, other TLR
agonist, STING
agonist, RIG-I-Like receptor agonist, C-type lectin receptors agonist, or
cytosolic DNA
Sensors agonist). In some aspects, the present disclosure provides a conjugate

represented by Formula I:
= = Dx I
(D;
20 wherein:
A is an antibody,
L is the linker;
Dx is the immune-stimulatory compound;
n is selected from 1 to about 20 or 2 to about 10 or 3 to about 8; and
25 z is selected from 1 to 20.
[0371] In further aspects, the present disclosure provides an antibody
conjugate
represented by the formula:
(D L3_)
163
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
wherein:
n is selected from 1 to about 20 or 2 to about 10 or 3 to about 8;
L3 is a linker; and
D is selected from a compound or salt of a compound of any one of Category A
5 Formulas (IA), (B3), (HA), (DB), (IIC), (IBA), (IBB), (IVA), (IVB), and
(IVC), or any
one of Category B Formulas (IA), (JIB), (IC), (IA), (BB), and (RC).
103721 In some embodiments, the immune-stimulatory compound is a myeloid
cell agonist. In some embodiments, the immune-stimulatory compound is a TLR8
agonist. In some embodiments, the immune-stimulatory compound is a TLR7
agonist.
10 In some embodiments, the immune-stimulatory compound is a TLR3 agonist.
In some
embodiments, the immune-stimulatory compound is a TLR4 agonist. In some
embodiments, the immune-stimulatory compound is a TLR5 agonist. In some
embodiments, the immune-stimulatory compound is a TLR9 agonist. In some
embodiments, the immune-stimulatory compound is a STING agonist. In some
15 embodiments, the immune-stimulatory compound is a RIG-I-Like receptor
agonist. In
some embodiments, the immune-stimulatory compound is a C-type lectin receptors

agonist. In some embodiments, the immune-stimulatory compound is a cytosolic
DNA
Sensors agonist.
103731 In some embodiments, the TLR8 agonist compound is a compound
20 selected from any one of Category A Formulas (IA), (113), (HA), (IB3),
(IIC), (IIIA),
(HID), (IVA), (IVB), and (IVC). In some embodiments, TLR7 agonist is a
compound
selected from any one of Category B Formulas (IA), (IB), (IC), (IA), (IB3),
and (IIC).
103741 In some aspects, the present disclosure provides a conjugate comprising

at least one immune-stimulatory compound (e.g., a compound or salt thereof),
an
25 antibody, and at least one linker, wherein each immune-stimulatory
compound is
linked, i.e., covalently bound, to the antibody through a linker. The linker
can be
selected from a cleavable or non-cleavable linker. In some embodiments, the
linker is
cleavable. In other embodiments, the linker is non-cleavable. Linkers are
further
described in the present application in the preceeding section, any one of
which can be
30 used to connect an antibody to an immune-stimulatory compound.
164
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0375] In a conjugate, the drug loading is represented by z, the number of
immune-stimulatory compound-linker molecules per antibody, or the number of
immune-stimulatory compounds per antibody, depending on the particular
conjugate.
Depending on the context, z can represent the average number of immune-
stimulatory
5 compound(-linker) molecules per antibody, also referred to the average
drug loading. z
can range from 1 to 20, from 1-50 or from 1-100. In some conjugates, z is
preferably
from 1 to 8. In some preferred embodiments, when z represents the average drug

loading, z ranges from about 2 to about 5. In some embodiments, z is about 2,
about 3,
about 4, or about 5. The average number of immune-stimulatory compounds per
10 antibody in a preparation of conjugate may be characterized by
conventional means
such as mass spectroscopy, liquid chromatography/mass spectrometry (LC/MS),
UHC,
ELISA assay, and HPLC.
103761 A number of conjugates are consistent with the disclosure herein. The
conjugates generally comprise an immune-stimulatory compound covalently bound
to
15 an antibody that localizes the conjugate to a target tissue, cell
population or cell. The
antibody is covalently attached to each immune-stimulatory compound, either
directly
or through a linker that tethers the immune-stimulatory compound to the
antibody.
Antibodies listed herein as well as antibodies to antigens or epitiopes
thereof listed
herein or otherwise known to one of skill in the art are consistent with the
conjugates as
20 disclosed herein.
[0377] Some exemplary conjugates are as follows. A conjugate can comprise
an antibody, at least one immune-stimulatory compound, and optionally at least
one
linker. A conjugate can comprise an antibody, at least one TLR7 agonist, and
at least
one linker. A conjugate can comprise an antibody, at least one TLR8 agonist,
and at
25 least one linker. A conjugate can comprise an antibody, at least one
Compound A
TLR8 agonist, and at least one linker. A conjugate can comprise an antibody,
at least
one Compound B TLR7 agonist, and at least one linker. A conjugate can comprise
an
antibody, at least one TLR3 agonist, and at least one linker. A conjugate can
comprise
an antibody, at least one TLR4 agonist, and at least one linker. A conjugate
can
30 comprise an antibody, at least one TLR5 agonist, and at least one
linker. A conjugate
can comprise an antibody, at least one TLR9 agonist, and at least one linker.
A
165
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
conjugate can comprise an antibody, at least one STING agonist, and at least
one linker.
A conjugate can comprise an antibody, at least one RIG-I agonist, and at least
one
linker. A conjugate can comprise an antibody, at least one C-type lectin
receptor
agonist, and at least one linker. A conjugate can comprise an antibody, at
least one
5 cytosolic DNA Sensors agonist, and at least one linker.
03781 In some embodiments, the immune stimulatory compound is a myeloid
cell agonist. A number of myeloid cell agonists are consistent with the
disclosure
herein such as a TLR8 agonist. Exemplary TLR8 agonists are provided herein. In

some embodiments, a myeloid cell agonist-linker compound (Linker-Payload) is
10 selected from any of Linker-Payloads provided herein.
Category A and Category B Conjugates
103791 In certain embodiments, the disclosure provides an immune-stimulatory
conjugate (or conjugate) of a or an antibody and at least one compound of any
one of
15 Category A Formulas (IA), (D3), (IA), (DB), (DC), (DA), (IDB), (WA),
(IVB), and
(IVC), each compound optionally attached to the antibody via a linker. In
certain
embodiments, the disclosure provides an immune-stimulatory conjugate of an
antibody
and at least one compound of any one of Category B Formulas (IA), (113), (IC),
(HA),
(DB), and (IIC), each compound optionally attached to the antibody via a
linker. In
20 certain embodiments, the average Drug-to-Antibody Ratio (DAR) of the
pharmaceutical
composition is selected from 1 to about 8, 2 to about 6, about 3 to about 5,
or about 4.
103801 In certain embodiments, the disclosure provides a pharmaceutical
composition suitable for intravenous or subcutaneous administration,
comprising an
immune stimulatory conjugate of a compound of any one of Category A Formulas
(IA),
25 (D3), (ILA), (DB), (IIC), (DIA), (MB), (WA), (IVB), and (IVC) and a
pharmaceutically
acceptable excipient. In certain embodiments, the disclosure provides a
pharmaceutical
composition suitable for intravenous or subcutaneous administration,
comprising an
immune stimulatory conjugate of a compound of any one of Category B Formulas
(IA),
(D)), (IC), (IIA), (DB), and (IIC), and a pharmaceutically acceptable
excipient. In
30 certain embodiments, the average Drug-to-Antibody Ratio (DAR) of the
pharmaceutical
composition is selected from 1 to about 8, 2 to about 6, about 3 to about 5,
or about 4.
166
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
103811 In certain embodiments, the disclosure provides a method for the
treatment of a disease treatable by a TLR agonist (e.g., cancer, viral
disease) comprising
subcutaneously administering an effective amount of a conjugate of a compound
of any
one of Category A Formulas (IA), (JIB), (IA), (JIB), (BC), (IIIA), (IIIB),
(IVA), (IVB),
5 and (IVC), or a pharmaceutical composition thereof suitable for
intravenous or
subcutaneous administration to a subject in need thereof. In certain
embodiments, the
disclosure provides a method for the treatment of cancer (e.g., breast, lung,
stomach/gastric, colorectal) or viral infection (e.g., HBV, HCV), comprising
intravenously or subcutaneously administering an effective amount of the
conjugate of
10 a compound of any one of Category B Formulas (IA), (B3), (IC), (IIA),
(IIB), and (IW),
or a pharmaceutical composition thereof suitable for subcutaneous
administration to a
subject in need thereof. In any of the embodiments herein, the conjugate may
be
administered by slow infusion.
103821 The disclosure provides a method of preparing an antibody conjugate of
15 the formula:
( D¨L3+Antibody
n
wherein:
n is selected from 1 to 20;
L3 is a linker; and
20 D is selected from a compound or salt of a compound of any one of
Category A
Formulas (IA), (M), (HA), (IIB), (IIC), (IIIA), (IBB), (IVA), (IVB), and
(IVC), and
Category B Formulas (IA), (IB), (IC), (IIA), (IIB), and (11C),
comprising contacting D-L3 with an antibody.
[0383] The disclosure provides a method of preparing an antibody conjugate of
25 the formula:
( D¨L3+Antibody
n
wherein:
n is selected from 1 to 20;
1,3 is a linker; and
167
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
D is selected from a compound of any one of Category A Formulas (IA), (1B),
(HA), (BB), (IIC), (IIIA), (IBB), (WA), (IVB), and (IVC), and Category B
Formulas
(IA), ([13), (IC), (HA), (JIB), and (IIC),
comprising contacting L3 with the antibody to form L3-antibody and contacting
5 L3 antibody with D to form the conjugate.
103841 In some embodiments, L3 and D together have a structure selected from:
H2N
yO
HN
H 0
IN.YLHN
NH2
N,
0
H2Nyo
HN
0
isss,Rxr. NH
H
N
0 H
H2 0 Oa
N,
0
0 \ea\
0
NH2
\RXZACcrH 4, 0 [µiimi
H
N
0
H 0
0
HN
10 H2N
AO
0
N, NH2
%tRXJ0 ---fr H 0 Sp 0 N'Thr
H 0
- H
o
HN
H2NAo
168
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
,
0 N
0
,A.NH2
0 --tn.:, 0
H
0 0 [zi N
0
H
N
--- r--\
H N
0

0 \----\
NH
0.)---NH2
,
00 N
o o
hi õ......¶ N, NH2
0 XrieH 0 * 0-AN N
,1/4.-RXI..:õ....-õ.õ...........,...õA
H 0 H
N N ..":-AN
--- Nr---\
H ! H
o A,
o \---- \
NH
0.).--NH2
,
FI2N y0
HN
11
14 0 H
0
0 o oy rtN *
H NH2
0
H
---- Nr--\
0 \----\
,
Hp tip
HN
H
,killirE4
.itcRx,-,r-N:II H
0 ..õ,..-7.õ..
Oy N ....,....--,N * r... Iczb 0 o
H NH2
0
11..õ.AN
H
--- /Th
N
169
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2N TO
HN
H
N N
0 40)
0
0
0 oTNNAc

_in_ 0
NH2
N,
\
0 \
H2N tO
HN
H
N 1.41
kRn - N 0 4,1 0y 14 0
H
HA N 0
`NI 0 NH2
N,
Nr-\
0
H2NyO
Amic-ro 1111ri
-r le
0
0
N
0
NH2
170
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
0
H
N
0 0 1. OyN
0 N
LL
N H2
N,
H2Ny0
HN
0
yiLIlir
A MC-WY
0 NH2
=
0
NrTh
0 \--\
H2N,f0
HN
0
j=L\ r
IM)C11111 N
õI 0
H
N H2
0 0
11 N,
0 \--\
coLN 0
0
r
_
H
FF H N N, N H2
0
/Th
HN 0 \
H2NAo
171
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
RXzJ%is/ 40 crAN.---y-, is
ratii 0 NH2
F F
rt,
H H
HN
H2N 0
and a salt of any one thereof, wherein the RX* is a bond, a succinimide
moiety, or a hydrolyzed succinimide moiety bound to a residue of an antibody,
wherein
5 on RX* represents the point of attachment to the residue of the
antibody.
103851 In some embodiments, RX* comprises a succinamide moiety and is
bound to a cysteine residue of an antibody. In some embodiments, RX* comprises
a
hydrolyzed succinamide moiety and is bound to a cysteine residue of an
antibody.
[0386] The compounds disclosed herein, in some embodiments, are used in
10 different enriched isotopic forms, e.g., enriched in the content of 2H,
3H, 11C, 13C and/or
14C.
In one particular embodiment, the compound is deuterated in at least one
position.
Such deuterated forms can be made by the procedure described in U.S Patent
Nos.
5,846,514 and 6,334,997. As described in U.S. Patent Nos. 5,846,514 and
6,334,997,
deuteration can improve the metabolic stability and or efficacy, thus
increasing the
15 duration of action of drugs.
[0387] Unless otherwise stated, structures depicted herein are intended to
include compounds which differ only in the presence of one or more
isotopically
enriched atoms. For example, compounds having the present structures except
for the
replacement of a hydrogen by a deuterium or tritium, or the replacement of a
carbon by
20 13C- or 14C-enriched carbon are within the scope of the present
disclosure.
[0388] The compounds of the present disclosure optionally contain unnatural
proportions of atomic isotopes at one or more atoms that constitute such
compounds.
For example, the compounds may be labeled with isotopes, such as for example,
deuterium (214), tritium (3H), iodine-125 (1251) or carbon-14 (4C). Isotopic
substitution
25 with 2H, 13c, 14c, 15c, 12N, l3N, 15N, 16N, 160, 170, 14F, 15F,
16F, 17F, 18F, 33s, 34s,
"S, 36S, "Cl, "Cl, "Br, 'Br, 125I are all contemplated. All isotopic
variations of the
172
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
compounds of the present disclosure, whether radioactive or not, are
encompassed
within the scope of the present disclosure.
[0389] In certain embodiments, the compounds disclosed herein have some or
all of the III atoms replaced with 21-I atoms. The methods of synthesis for
deuterium-
5 containing compounds are known in the art and include, by way of non-
limiting
example only, the following synthetic method&
103901 Deuterium substituted compounds are synthesized using various
methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the
Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and
10 Development. [In: Curr., Phann. Des., 2000; 6(10)] 2000, 110 pp; George
W.; Vanma,
Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic
Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony.
Synthesis
of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32.
[0391] Deuterated starting materials are readily available and are subjected
to
15 the synthetic methods described herein to provide for the synthesis of
deuterium-
containing compounds. Large numbers of deuterium-containing reagents and
building
blocks are available commercially from chemical vendors, such as Aldrich
Chemical
Co.
[0392] Compounds of the present disclosure also include crystalline and
20 amorphous forms of those compounds, pharmaceutically acceptable salts,
and active
metabolites of these compounds having the same type of activity, including,
for
example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated
polymorphs
(including anhydrates), conformational polymorphs, and amorphous forms of the
compounds, as well as mixtures thereof.
25 103931 Included in the present disclosure are salts, particularly
pharmaceutically
acceptable salts, of the compounds described herein. The compounds of the
present
disclosure that possess a sufficiently acidic, a sufficiently basic, or both
functional
groups, can react with any of a number of inorganic bases, and inorganic and
organic
acids, to form a salt. Alternatively, compounds that are inherently charged,
such as
30 those with a quaternary nitrogen, can form a salt with an appropriate
counterion, e.g., a
halide such as bromide, chloride, or fluoride.
173
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0394] The compounds described herein may in some cases exist as
diastereomers, enantiomers, or other stereoisomeric forms. The compounds
presented
herein include all diastereomeric, enantiomeric, and epimeric forms as well as
the
appropriate mixtures thereof. Separation of stereoisomers may be performed by
5 chromatography or by forming diastereomers and separating by
recrystallization, or
chromatography, or any combination thereof (Jean Jacques, Andre Collet, Samuel
H.
Wilen, "Enantiomers, Racemates and Resolutions," John Wiley and Sons, Inc.,
1981,
herein incorporated by reference for this disclosure). Stereoisomers may also
be
obtained by stereoselective synthesis.
10 [0395] The methods and compositions described herein include the
use of
amorphous forms as well as crystalline forms (also known as polymorphs). The
compounds described herein may be in the form of pharmaceutically acceptable
salts.
As well, active metabolites of these compounds having the same type of
activity are
included in the scope of the present disclosure. In addition, the compounds
described
15 herein can exist in unsolvated as well as solvated forms with
pharmaceutically
acceptable solvents such as water, ethanol, and the like. The solvated forms
of the
compounds presented herein are also considered to be disclosed herein.
[0396] In certain embodiments, compounds or salts of the compounds described
herein may be prodrugs attached to antibodies to form conjugates. The term
"prodrug"
20 is intended to encompass compounds which, under physiologic conditions,
are
converted into active compounds, e.g., TLR8 agonists, TLR7 agonists, other TLR

agonists, STING agonist, RIG-I-Like receptor agonists, C-type lectin receptors

agonists, or cytosolic DNA Sensors agonists. One method for making a prodrug
is to
include one or more selected moieties which are hydrolyzed or otherwise
cleaved under
25 physiologic conditions to reveal the desired molecule. In other
embodiments, the
prodrug is converted by an enzymatic activity of the host animal such as
specific target
cells in the host animal.
[0397] Prodrug forms of the herein described compounds, wherein the prodrug
is metabolized in vivo to produce a compound described herein are included
within the
30 scope of the claims. In some cases, some of the herein-described
compounds may be a
prodrug for another derivative or active compound
174
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
103981 In certain embodiments, an immune-stimulatory compound, such as a
TLR8 agonist or TLR7 agonist, is modified as a prodrug with a masking group,
such
that the TLR8 agonist, TLR7 agonist or other agonist, has limited activity or
is inactive
until it reaches an environment where the masking group is removed to reveal
the active
5 compound. For example, a TLR8 agonist or TLR7 agonist can be covalently
modified
at an amine involved in binding to the active site of a TLR8 receptor such
that the
compound is unable to bind the active site of the receptor in its modified
(prodrug)
form. In such an example, the masking group is removed under physiological
conditions, e.g., enzymatic or acidic conditions, specific to the site of
delivery, e.g.,
10 intracellular or extracellular adjacent to target cells. Masking groups
may be removed
from the amine of the compound or salt described herein due to the action of
lysosomal
proteases, e.g., cathepsin and plasmin. These proteases can be present at
elevated levels
in certain tumor tissues. The masking group may be removed by a lysosomal
enzyme.
The lysosomal enzyme can be, for example, cathepsin B, cathepsin S,13-
glucuronidase,
15 or13-galactosidase.
103991 In certain embodiments, an amine masking group inhibits binding of the
amine group of the compound with residues of a TLR8 receptor. The amine
masking
group may be removable under physiological conditions within a cell but
remains
covalently bound to the amine outside of a cell. Masking groups that may be
used to
20 inhibit or attenuate binding of an amine group of a compound with
residues of a TLR8
receptor include, for example, peptides and carbamates.
104001 Synthetic chemistry transformations and methodologies useful in
synthesizing the compounds described herein are known in the art and include,
for
example, those described in R. Larock, Comprehensive Organic
25 Transformations (1989); T. W. Greene and P. G. M Wuts, Protective Groups
in
Organic Synthesis, 2d. Ed. (1991); L. Fieser and M. Fieser, Fieser and Fieser
's
Reagents for Organic Synthesis (1994); and L. Paquette, ed., Encyclopedia of
Reagents
for Organic Synthesis (1995).
104011 The activation, stimulation or augmentation of an immune response by
30 an immune-stimulatory conjugate, such as a myeloid cell agonist, can be
measured in
vitro by co-culturing immune cells (e.g., myeloid cells) with cells targeted
by the
175
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
conjugate and measuring cytokine release, chemokine release, proliferation of
immune
cells, upregulation of immune cell activation markers, ADCC, or any
combination
thereof. ADCC can be measured by determining the percentage of remaining
target
cells in the co-culture after administration of the conjugate with the target
cells, myeloid
5 cells, and other immune cells. In some embodiments, an immune-stimulatory
conjugate
can activate or stimulate immune cell activity, as determined by in vitro
assay, such as a
cytokine release assay, by detection of activation markers (e.g., MEC class II
markers)
or other assays known in the art. In some embodiments, an immune-stimulatory
conjugate has an EC50 of 100 n1V1 or less, as determine by cytokine release
assay. In
10 some embodiments, an immune-stimulatory conjugate has an EC50 of 50 nlv1
or less, as
determine by cytokine release assay. In some embodiments, an immune-
stimulatory
conjugate has an EC50 of 10 nlY1 or less, as determine by cytokine release
assay. In
some embodiments, an immune-stimulatory conjugate has an EC50 of 1mM or less.
Exemplary Combination Therapy
15 104021 Conjugates described herein can be administered in
combination with
one or more other modes of treatment in a combination therapy. The other mode
or
modes of treatment can be provided before, substantially contemporaneous with,
and/or
after the immune-stimulatory conjugate. Exemplary combination therapies
comprise an
immune-stimulatory conjugate described herein and a second anti-cancer agent.
In
20 some embodiments, the second anti-cancer agent is a small molecule
chemotherapy
and/or a biologic therapy, such as a therapeutic antibody. In some
embodiments, the
second anti-cancer agent is or comprises an immune checkpoint inhibitor, a
therapeutic
comprising a HER2-targeting agent, a kinase inhibitor, or a molecularly
targeted
therapy. In some embodiments, a combination therapy comprises surgery and/or
25 radiation therapy in addition to an immune-stimulatory conjugate
described herein, with
or without a second anti-cancer agent.
[0403] Second anti-cancer agents useful in the methods provided herein
include,
for example, chemotherapy agents, ionizing radiation, chemotherapy protective
agents,
molecularly targeted therapy, anti-cancer biologic therapy, immune checkpoint
30 inhibitor, therapeutic agents targeting HERZ kinase inhibitors, and
other anti-cancer
176
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
drugs. Examples of chemotherapy agents contemplated as further therapeutic
agents
include alkylating agents, such as nitrogen mustards (e.g., mechlorethamine,
cyclophosphamide, ifosfamide (IFEX0), melphalan (Alkeran ), and chlorambucil);

bifunctional chemotherapeutics (e.g., bendamustine); nitrosoureas (e.g.,
carmustine
5 (BCNU, BiCNUO; polifeprosan 20 implant (Gliadel )), lomustine (CCNU), and
semustine (methyl-CCNU)); ethyleneimines and methyl-melamines (e.g.,
triethylenemelamine (TEM), triethylene thiophosphoramide (thiotepa), and
hexamethylmelamine (HMM, altretamine)); alkyl sulfonates (e.g., busulfan
(Mylerane), busulfan injection (Busulfexe)); and triazines (e.g., dacabazine
(DTIC));
10 antimetabolites, such as folic acid analogues (e.g., methotrexate
(Folexe), trimetrexate,
and pemetrexed (multi-targeted antifolate)); pyrimidine analogues (such as 5-
fluorouracil (5-FU, Adrucil , Efudex0), capecitabine (Xeloda0),
fluorodeoxyuridine,
tezacitabine, gemcitabine, cytosine arabinoside (AraC, cytarabine (Cytosar-U
);
cytarabine Liposome injection (DepoCyte)), 5-azacytidine, and 2,2'-
15 difluorodeoxycytidine); purine analogues (e.g., 6-mercaptopurine
(Purinethol0), 6-
thioguanine, azathioprine, 2'-deoxycoformycin (pentostatin),
erythrohydroxynonyladenine (EHNA), fludarabine phosphate (Fludara0), 2
chlorodeoxyadenosine (cladribine, 2-CdA)); Type I topoisomerase inhibitors
such as
camptothecin (CPT), topotecan (Hycamptine), and irinotecan (Camptosar0);
natural
20 products, such as epipodophylotoxins (e.g., etoposide (Vepeside) and
teniposide
(Vumon )); vinca alkaloids (e.g., vinblastine (Velban ), vincristine
(Oncovin0), and
vinorelbine (Navelbine0)); anti-tumor antibiotics such as actinomycin D
(dactinomycin, Cosmegan0), doxorubicin hydrochloride (Adriamycin , Rubex0),
mitoxantrone (Novantronee), and bleomycin sulfate (Blenoxane0),
radiosensitizers
25 such as 5-bromodeozyuridine, 5-iododeoxyuridine, and bromodeoxycytidine;
platinum
coordination complexes such as cisplatin (Platinol*), carboplatin
(Paraplatin0), and
oxaliplatin (Eloxatin0); substituted ureas, such as hydroxyurea (Hydrea0);
microtubule
inhibitors such as paclitaxel (Taxo10), docetaxel (Taxotere0), and eribulin
(Halavene);
FOLFOX (a chemotherapy regimen comprising Folinic acid (leucovorin) "FOL",
30 Fluorouracil (5-F1.1) "F", and Oxaliplatin (E(oxatin) "OX"),
immunosuppressive agents
such as cyclophosphamide (Cytoxan or Neosar0); hormone-based compound such as
177
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
anastrozole (Arimidexe), exemestane (Aromasin*), letrozole (Femara*),
fulvestrant
(Faslodexe), bicalutamide (Casodexe), Flutamide (Eulexine), Nilutamide
(Nilandrone), Enzalutamide (Xtandi0), apalutamide (Erleada0), darolutamide
(Nubega.0), Degarelix (Firmagone), Toremifene (Farestone), goserelin
(Zoladexe),
5 Triptorelin (Trelstar0), Histrelin (Vantase), leuprolide (Luprone) and
tamoxifen
citrate (Nolvadexe); an anti-inflammatory agent such as dexamethasone; an anti-

androgen compound such as flutamide (Eulexine); an anthracycline compound such
as
idarubicin (Idamycin , Zavedosit) and epirubicin (Ellence8); bioreductive anti-
cancer
agent such as tirapazamine (Tirazonee); serine/threonine kinase inhibitors
such as
10 CDK4/6 inhibitors abemaciclib (Verzenio0), palbociclib (Ibrance0), and
ribociclib
(Kisciali0); tyrosine kinase inhibitors such as tucatinib, cabozantinib,
afatinib, erlotinib,
pyrotinib, neratinib, poziotinib, dacomitinib, gefitinib, lapatinib,
osimertinib,
larotrectinib, axitinib, lenvatinib, pazopanib, regorafenib, and sunitinib;
and
methylhydrazine derivatives such as N methylhydrazine (WEI) and procarbazine.
15 104041 Anticancer molecularly targeted therapy target specific
molecules
involved in the development, growth and spread of cancer cells. In some
embodiments,
a molecularly targeted therapy is a small molecule or comprises an antibody or
antigen
binding fragment. Examples of molecularly targeted include ALK inhibitors such
as
crizotinib, ceritinib, and alectinib; BCL2 inhibitors such as venetoclax,
obatoclax,
20 navitoclax, and gossypol; PARP inhibitors such as iniparib and olaparib;
BRAF
inhibitors such as vernurafenib and dabrafenib; and angiogenesis inhibitors
such as
VEGF and/or VEGFR inhibitors, including axitinib, sunitinib, pazopanib,
cabozantinib,
vandetanib, motesanib, regorafenib, cediranib, lenvatinib, sorafenib, ziv-
aflibercept,
aflibercept, ranibizumab, ramucirumab, and bevacizumab.
25 104051 Examples of anti-cancer biologic therapy include biologic
therapy
comprising antibodies (e.g., therapeutic antibodies or antigen binding
fragments
thereof, antibody conjugates, cellular immunotherapy (e.g., chimeric antigen
receptor
(CAR) T cells, T cell receptor (TCR) immunotherapy, dendritic cell therapy),
and Fc-
fusion proteins. In some embodiments, an anti-cancer biologic therapy targets
a tumor
30 antigen, or a molecule in involved in development, growth, and/or spread
of cancer
cells (molecularly targeted immunotherapy). Examples of antibody conjugate
therapy
178
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
contemplated for use as therapeutic agents include, for example, sacituzumab
govitecan
and enfortumab vedotin.
[0406] In some embodiments, additional anti-cancer agents useful in the
methods provided herein include chemotherapy protective agents that prevent or
5 descrease toxic side effects of chemotherapy agents. Examples of
chemotherapy
protect agents contemplated for use as further therapeutic agents include, for
example,
leucovorin (folinic acid), allpuronol (Zyloprim0), rasburicase (Fasturtece),
amifostine
(Ethyo10), dexrazoxane (Zinecard0), and mesna (Mesnexa).
[0407] In certain embodiments, the combination therapy comprises a conjugate
10 described herein and palbociclib. In further embodiments, the
combination therapy
comprises a conjugate described herein and abemaciclib. In still further
embodiments,
the combination therapy comprises a conjugate described herein and ribociclib.
Immune Checkpoint Inhibitors
[0408] In some embodiments, combination therapies comprise a conjugate
15 described herein and a second anti-cancer agent, wherein the second anti-
cancer agent
is or comprises an immune checkpoint inhibitor. In some embodiments, the
immune
checkpoint inhibitor is an antagonist of PD-1, PD-L1, and/or PD-L2. In some
embodiments, the antagonist of PD-1, PD-L1, and/or PD-L2 is a small molecule,
such
as INCB 86550, 1,3,4- and 1,2,4-oxadiazole and 1,3,4- and 1,2,4-thiadiazole
20 compounds (see WO 2015/033301 WO 2016/142894; WO 2016/142833,
WO 2018/051255; WO 2018/07375, and Guzik etal., Molecules 24:2071, 2019, the
compounds from which are incorporated herein by reference), and GS-4224;
peptides
and peptidomimetics such as AUNP-12 and heptapeptide SNTSEFS-NH2 (SEQ ID NO:
12)), macrocyclic peptides such as BMS-57, BMS-71, and BMS99; ansamycin class
of
25 antibiotics such as geldanamycin and rifabutin; Fc-fusion proteins, such
as AMP-224;
and antibodies, such as pembrolizumab (CEYTRUDA ); pembrolizumab biosimilar;
nivolumab (OPDIVO ); nivolumab biosimilar; pidilizumab; pidilizumab
biosimilar;
cemiplimab; AMP-514 (MEDI0608, see U.S. Patent No. 8,609,089,B2); MDX-1105;
dostarlimab; tislelizumab; ICD-033, AGEN2034; STI-A1010; STI-A1110;
durvalumab;
30 atezolizumab; avelumab; BMS-936559; or a PD-1 antibody disclosed in U.S.
Patent
179
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
No. 8,008,449; 6,808,710; 7,488,802; 8,168,75; or 8,354,509; or PCT
Publication No.
WO 2012/145493. In some embodiments, the immune checkpoint inhibitor is an
anti-
PD1 antibody selected from pembrolizumab (KEYTRUDA0); pembrolizumab
biosimilar; nivolumab (OPDIV00); nivolumab biosimilar; pidilizumab;
pidilizumab
5 biosimilar; toripalimab; cemiplimab; cemiplimab biosimilar, dostarlimab;
tislelizumab;
BMS-936559; AMP-514 (MEDI0608, see U.S. Patent No. 8,609,089,B2); AGEN2034;
and STI-A1110. In some embodiments, the immune checkpoint inhibitor is an anti-
PD-
L1 antibody selected from atezolizumab; atezolizumab biosimilar; durvalumab;
durvalumab biosimilar; avelumab; avelumab biosimilar; MDX-1105; BMS-936559;
KD-033; and STI-A1010.
[0409] In certain embodiments, the immune checkpoint inhibitor is an inhibitor

of TIGIT, e.g., an anti-TIGIT antibody such as tiragolumab.
[0410] In some embodiments, the combination therapy comprises a conjugate
described herein and an additional therapeutic agent comprising pembrolizumab,
15 nivolumab, pidilizumab, or a biosimilar thereof. In certain embodiments,
the
combination therapy comprises a conjugate described herein and pembrolizumab
or a
biosimilar thereof. In certain further embodiments, the combination therapy
comprises
a conjugate described herein and nivolumab or a biosimilar thereof
Therapeutic Agents Targeting HER2
20 [0411] In some embodiments, combination therapies comprise a
conjugate
described herein and an additional therapeutic agent targeting HER2, such as
kinase
inhibitors and/or therapeutic agents comprising an anti-HER2 antibody. In some

embodiments, a kinase inhibitor is a tyrosine kinase inhibitor or a
serine/threonine
kinase inhibitor, including a CDK4/6 inhibitor. In some embodiments, a CDK4/6
25 inhibitor is selected from abemaciclib (Verzenio0), palbociclib
(Ibrance0), and
ribociclib (Kisqa1i0). Nonlimiting exemplary tyrosine kinase inhibitors
include
tucatinib, cabozantinib, afatinib, erlotinib, pyrotinib, neratinib,
poziotinib, dacomitinib,
gefitinib, lapatinib, osimertinib, larotrectinib, axitinib, lenvatinib,
pazopanib,
regorafenib, and sunitinib. In some embodiments, the tyrosine kinase inhibitor
is
30 selected from tucatinib, cabozantinib, pyrotinib, neratinib, and
poziotinib. In some
180
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
embodiments, an additional therapeutic is an adenosine receptor antagonist,
such as
AB928. Therapeutic agents comprising an anti-HER2 antibody include anti-HER2
antibodies or antigen binding fragments thereof and anti-HER2 antibody
conjugates. In
some embodiments, the therapeutic agent comprising an anti-HER2 antibody is
selected
5 from trastuzumab, a trastuzumab biosimilar (such as trastuzumab-qyyp
(TRAZIMERATm), trastuzumab-plcrb (HERZUMA0), trastuzumab-dttb
(ONTRUZANTO), trastuzumab-anns (KANJINTITm), trastuzumab-dkst (OgivriTm),
fam-trastuzumab deruxtecan-nxki (ENHERTUO, DS-8201), ado-trastuzumab
emtansine (KADCYLA0), trastuzumab duocarmazine (SYD985), PF-06804103,
10 trastuzumab auristatin-0101, margetuximab, zenocutuzumab (MCLA-128),
ZW25
(Zymeworks); ZW49 (Zymeworks); RC48-ADC (RemeGen); RG6148 (DHES0815A);
or RG6194 (BTRC4017A).
[0412] In some embodiments, the combination therapy comprises a conjugate
described herein and an additional therapeutic agent selected from any one of
15 trastuzumab, trastuzumab-qyyp, trastuzumab-pkrb, trastuzumab-dttb,
trastuzumab-anns,
trastuzumab-dkst, and margetuximab. In certain embodiments, the combination
therapy
comprises a conjugate described herein and trastuzumab. In certain further
embodiments, the combination therapy comprises a conjugate described herein
and
trastuzumab-qyyp.
20 [0413] In further embodiments, the combination therapy comprises a
conjugate
described herein and an additional therapeutic agent selected from any one of
fam-
trastuzumab deruxtecan-nxki, ado-trastuzumab emtansine, and trastuzumab
duocarmazine. In certain embodiments, the combination therapy comprises a
conjugate
described herein and fam-trastuzumab deruxtecan-nxki. In certain embodiments,
the
25 combination therapy comprises a conjugate described herein and ado-
trastuzumab
emtansine.
[0414] In some embodiments, the combination therapy comprises a conjugate
described herein and an additional therapeutic agent selected from any one of
tucatinib,
pyrotinib, neratinib, and poziotinib. In certain embodiments, the combination
therapy
30 comprises a conjugate described herein and tucatinib.
181
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Pharmaceutical Formulations
[0415] The conjugates described herein are useful as pharmaceutical
compositions for administration to a subject in need thereof Pharmaceutical
compositions can comprise the conjugates described herein and one or more
5 pharmaceutically acceptable excipients, suitable for administration by a
selected route.
A pharmaceutical composition can comprise any conjugate described herein. A
pharmaceutical composition can further comprise buffers, carbohydrates, and/or

preservatives, as appropriate. Pharmaceutical compositions comprising a
conjugate can
be manufactured, for example, by lyophilizing the conjugate, mixing,
dissolving,
10 emulsifying, encapsulating or entrapping the conjugate. The
pharmaceutical
compositions can also include the conjugates described herein in a free-base
form or
pharmaceutically-acceptable salt form.
[0416] Methods for formulation of the pharmaceutical compositions can include
formulating any of the conjugates described herein with one or more inert,
15 pharmaceutically-acceptable excipients or carriers to form a solid, semi-
solid, or liquid
composition for intravenous or subcutaneous administration. Solid compositions
can
include, for example, powders, and in some aspects, the solid compositions
further
contain nontoxic, auxiliary substances, for example wetting or emulsifying
agents, pH
buffering agents, and other pharmaceutically-acceptable additives.
Alternatively, the
20 compositions described herein can be lyophilized or in powder form for
re-constitution
with a suitable vehicle, e.g., sterile pyrogen-free water, before use
[0417] The pharmaceutical compositions and formulations can be sterilized.
Sterilization can be accomplished by filtration through sterile filtration.
[0418] The pharmaceutical compositions described herein can be formulated for
25 administration as an injection, e.g., an intravenous or subcutaneous
injection Non-
limiting examples of formulations for injection can include a sterile
suspension,
solution or emulsion in oily or aqueous vehicles. Suitable oily vehicles can
include, but
are not limited to, lipophilic solvents or vehicles such as fatty oils or
synthetic fatty acid
esters, or liposomes. Aqueous injection suspensions can contain substances
which
30 increase the viscosity of the suspension. The suspension can also
contain suitable
stabilizers. Alternatively, the pharmaceutical compositions described herein
can be
182
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
lyophilized or in powder form for reconstitution with a suitable vehicle,
e.g., sterile
pyrogen-free water, before use.
[0419] The conjugates can be formulated for administration in a unit dosage
form in association with a pharmaceutically acceptable vehicle. Such vehicles
can be
5 inherently nontoxic, and non-therapeutic. A vehicle can be water, saline,
Ringer's
solution, dextrose solution, and 5% human serum albumin. Nonaqueous vehicles
such
as fixed oils and ethyl oleate can also be used. The vehicle can contain minor
amounts
of additives such as substances that enhance isotonicity and chemical
stability (e.g.,
buffers and preservatives).
10 [0420] In some embodiments, an aqueous formulation of a conjugate
provided
herein, such as for subcutaneous administration, has a pH from 4-5.2. The
aqueous
formulation may comprise one or more excipients, such as, for example, one or
more
buffering agents, one or more lyoprotectants, and the like. In some
embodiments, the
pH of the formulation is from 4-5.1, 4.1-5.1, 4.2-5.1, 4.3-5.1, 4.4-5.1, 4.5-
5.1, 4-5, 4.1-
15 5, 4.2-5, 4.3-5, 4.4-5, or 4.5-5. In some embodiments, the formulation
comprises at
least one buffer. In various embodiments, the buffer may be selected from
histidine,
citrate, aspartate, acetate, phosphate, lactate, tromethamine, gluconate,
glutamate,
tartrate, succinate, malic acid, fumarate, a-ketoglutarate, and combinations
thereof In
some embodiments, the buffer is at least one buffer selected from histidine,
citrate,
20 aspartate, acetate, and combinations thereof In some embodiments, the
buffer is a
combination of histidine and aspartate. In some embodiments, the total
concentration
of the buffer in the aqueous formulation is 10mM to 40mIv1, such as 15mM-30mM,

15mM-25mM, or 20 mM.
[0421] In some embodiments, the aqueous formulation comprises at least one
25 lyoprotectant. In some such embodiments, the at least one lyoprotectant
is selected
from sucrose, arginine, glycine, sorbitol, glycerol, trehalose, dextrose,
alpha-
cyclodextrin, hydroxypropyl beta-cyclodextrin, hydroxypropyl gamma-
cyclodextrin,
proline, methionine, albumin, mannitol, maltose, dextran, and combinations
thereof In
some embodiments, the lyoprotectant is sucrose. In some embodiments, the total
30 concentration of lyoprotectant in the aqueous formulation is 3-12%, such
as 5-12%, 6-
10%, 5-9%, 7-9%, or 8%.
183
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0422] In some embodiments, the aqueous formulation comprises at least one
surfactant. Exemplary surfactants include polysorbate 80, polysorbate 20,
poloxamer
88, and combinations thereof In some embodiments, the aqueous formulation
comprises polysorbate 80. In some embodiments, the total concentration of the
at least
5 one surfactant is 0.01%-0.1%, such as 0.01%43.05%, 0.01%4).08%, or 0.01%-
0.06%,
0.01%-0.04%, 0.01%-0.03%, or 0.02%.
[0423] In some embodiments, the concentration of the conjugate in the aqueous
formulation is 1 mg/mL-200 mg/mL, such as 10 mg/mL-160 mg/mL, 10 mg/mL-140
mg/mL, 10 mg/mL-120 mg/mL, 20 mg/mL-120 mg/mL, or 30 mg/mL-120 mg/mL, or
10 40 mg/mL-120 mg/mL, or 40 mg/mL-100 mg/mL. In some embodiments, the
concentration of the conjugate in the aqueous formulation is 10 mg/mL-I40
mg/mL or
40 mg/mL-140 mg/mL.
Therapeutic Applications
[0424] The combination therapies provided herein are useful for treating
15 plurality of different subjects including, but not limited to, a mammal,
human, non-
human mammal, a domesticated animal (e.g., laboratory animals, household pets,
or
livestock), non-domesticated animal (e.g., wildlife), clog, cat, rodent,
mouse, hamster,
cow, bird, chicken, fish, pig, horse, goat, sheep, rabbit, and any combination
thereof. In
various embodiments, the subject is a human.
20
[0425] In various embodiments, methods of treating
cancer (e.g., breast, lung,
stomach/gastric, colorectal) or viral infection (e.g., HBV, HCV) are provided,

comprising administering to a subject with cancer or viral infection an immune

checkpoint inhibitor and a conjugate provided herein, wherein the conjugate
comprises
an antibody that binds a tumor associated antigen for cancer, or a liver cell
antigen for
25 viral infection. In some such embodiments, the immune checkpoint
inhibitor is an
antibody. In some such embodiments, the immune checkpoint inhibitor is an
inhibitor
of PD-1 and/or PD-L1, e.g., an anti-PD-1 and/or anti-PD-Li antibody. In
certain
embodiments, the immune checkpoint inhibitor is an inhibitor of TIGIT, e.g.,
an anti-
TIGIT antibody such as tiragolumab.
184
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0426] Nonlimiting exemplary inhibitors of PD-1 and/or PD-Li include
pembrolizumab, nivolumab, cemiplimab, atezolizumab, avelumab, and durvalumab,
or
a biosimilar thereof. In some embodiments, the immune checkpoint inhibitor is
an anti-
PD1 antibody selected from pembrolizumab, nivolumab, zimberelimab, and
5 cemiplimab, or a biosimilar thereof In some embodiments, the immune
checkpoint
inhibitor is an anti.-PD-L1 antibody selected from atezolizumab, avelumab, and

durvalumab, or a biosimilar thereof.
[0427] In some embodiments, the conjugate comprises an antibody that binds
HERI In some embodiments, the anti-HER2 antibody of the conjugate comprises
the
10 CDRs of pertuzumab, antigen binding fragment (e.g., variable regions) of
pertuzumab,
light and heavy chains of pertuzumab. In some embodiments, the anti-HER2
antibody
of the conjugate comprises the CDRs of trastuzumab, antigen binding fragment
(e.g.,
variable regions) of trastuzumab, light and heavy chains of trastuzumab.
[0428] In some such embodiments, the anti-HER2 antibody of the conjugate
15 comprises heavy chain (HC)-CDR1, HC-CDR2, HC-CDR3, light chain (LC)-
CDR1,
LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6, respectively. In further embodiments,

the anti-HER2 antibody of the conjugate comprises a heavy chain and light
chain,
wherein: (a) the heavy chain comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ
ID NOs: 1-3, respectively, and comprises a heavy chain variable region (YR)
having an
20 amino acid sequence that has at least 90%, at least 91%, at least 92%,
at least 93%, at
least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least
99%, at least
99.5%, or 100% identity to the Vii amino acid sequence of SEQ ID NO: 7; and
(b) the
light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 4-6,
respectively, and a light chain variable region (VL) having an amino acid
sequence that
25 has at least 90%, at least 91%, at least 92%, at least 93%, at least
94%, at least 95%, at
least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100%
identity to
the Vt, amino acid sequence of SEQ ID NO: 8. In still further embodiments, the
anti-
HER2 antibody of the conjugate comprises a NTH comprising or consisting of the
amino
acid sequence of SEQ ID NO: 7 and a Vt, comprising or consisting of the amino
acid
30 sequence of SEQ ID NO: 8. In yet further embodiments, the anti-HER2
antibody of the
conjugate comprises a heavy chain and light chain, wherein: (a) the heavy
chain
185
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ ID NOs: 1-3, respectively,
and comprises an amino acid sequence that has at least 90%, at least 91%, at
least 92%,
at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least
98%, at least
99%, at least 99.5%, or 100% identity to the heavy chain amino acid sequence
of SEQ
5 ID NO: 9; and (b) the light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3
of
SEQ ID NOs: 4-6, respectively, and an amino acid sequence that has at least
90%, at
least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least
96%, at least
97%, at least 98%, at least 99%, at least 99.5%, or 100% identity to the light
chain
amino acid sequence of SEQ ID NO: 10. In yet further embodiments, the anti-
HER2
10 antibody of the conjugate comprises a heavy chain comprising or
consisting of the
amino acid sequence of SEQ ID NO: 9 and a light chain comprising or consisting
of the
amino acid sequence of SEQ ID NO: 10.
[0429] In some such embodiments, the method comprises administering an
additional therapeutic agent. In some such embodiments, the additional
therapeutic
15 agent comprises a chemotherapy agent, a kinase inhibitor, a therapeutic
agent targeting
HER2, a biologic therapy such as a therapeutic antibody, a molecularly
targeted
therapy, a chemotherapy protective agent, or any combination thereof.
[0430] In some such embodiments, the therapeutic agent is a therapeutic agent
targeting HER-2, such as kinase inhibitors and/or therapeutic agents
comprising an anti-
20 HER2 antibody. In some such embodiments, the therapeutic agent targeting
HER2 is
selected from trastuzumab, trastuzumab-qyyp, trastuzumab-pkrb, trastuzumab-
dttb,
trastuzumab-anns, trastuzumab-dkst, trastuzumab deruxtecan-nxki, ado-
trastuzumab
emtansine, trastuzumab, duocarmazine, margetuximab, zenocutuzumab (MCLA-128),
tucatinib, cabozantinib, pyrotinib, neratinib, and poziotinib. In some such
25 embodiments, the therapeutic agent targeting HER2 comprises an anti-HER2
antibody,
e.g., an anti-HER2 antibody or antigen binding fragment thereof, or anti-HER2
antibody conjugate. In some such embodiments, the therapeutic agent comprising
an
anti-HER2 antibody is selected from trastuzumab, trastuzumab-qyyp, trastuzumab-
pkrb,
trastuzumab-dttb, trastuzumab-anns, trastuzumab-dkst, trastuzumab deruxtecan-
nxki,
30 ado-trastuzumab emtansine, trastuzumab, duocarmazine, margetuximab,
zenocutuzumab (MCLA-128).
186
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0431] In some such embodiments, the kinase inhibitor is a tyrosine kinase
inhibitor or a serine/threonine kinase inhibitor, such as a CDK4/6 inhibitor.
In some
embodiments, a tyrosine kinase inhibitor is tucatinib, cabozantinib, afatinib,
erlotinib,
pyrotinib, neratinib, poziotinib, dacomitinib, gefitinib, lapatinib,
osimertinib,
5 larotrectinib, axitinib, lenvatinib, pazopanib, regorafenib, or
sunitinib. In some
embodiments, the tyrosine kinase inhibitor is tucatinib, cabozantinib,
pyrotinib,
neratinib, or poziotinib. In some embodiments the CDK4/6 inhibitor is
abemaciclib,
palbociclib, or ribociclib.
[0432] In some such embodiments, the chemotherapy agent comprises a
10 hormone-based compound, optionally wherein the hormone based compound
comprises
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
enzalutamide, apalutamide, darolutamide, degarelix, torernifene, goserelin,
triptorelin,
histrelin, leuprolide, or tamoxifen citrate.
[0433] In some such embodiments, the chemotherapy agent comprises a
15 platinum coordination complex, optionally wherein the platinum
coordination complex
comprises cisplatin, oxaliplatin, or carboplatin.
[0434] In some such embodiments, the chemotherapy agent comprises a folic
acid analog, optionally wherein the folic acid analog comprises pemetrexed,
methotrexate, or trimetrexate.
20 [0435] In some such embodiments, the chemotherapy agent comprises
a
pyrimidine analog, optionally wherein the pyrimidine analog comprises
fluorouracil,
capecitabine, fluorodeoxyufidine, tezacitabine, gemcitabine, cytosine
arabinoside,
cytarabine, 5-azacytidine, or 2,2'-difluorodeoxycytidine.
[0436] In some such embodiments, the chemotherapy agent comprises a
25 microtubule inhibitor, optionally wherein the microtubule inhibitor
comprises
paclitaxel, docetaxel, and eribulin.
[0437] In some such embodiments, the chemotherapy protective agent
comprises leucovorin (folinic acid), allpuronol, rasburicase, amifostine,
dexrazoxane, or
mesna.
30 104381 In any of the aforementioned embodiments, the linker-
payload
compound of the conjugate has a structure selected from:
187
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N TO
HN
0
myliplyi
N
405---Rwre N
H
0 0 0.,...õNi -a31.,,N
N
NH2
0
,
I I
H
,
FI2N tO
HN
0
RXt
ts "rqlYL\11 rµl
NH2
H
0 0 0
H
N
0
N
A0 N__
NH2
. H
N ,....-
o -õ--
01 -..,1
Xff.,H o . n
o
rt-Thr ri
tztrRX:I.,...,.........,......%__A_
N NIAN
H H
N
0

0 \---"\
HN
H2NA0
/
OSr ..iN1 o 0
JI,N..., NH2
0 Xiiii 0 is 0 [1
H
N ---- i----\
H H
N
0
NH
Cd%.--NH2
H2 N yel
HN
H H
FRx,wyNxiltily N4
0
H H
0
a a Oy N......õ..---N * 0 NH2
H
0
N,
N
H
0
¨ r---...
5
188
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
0 rIXII\H" 0 11
Oy
0
ii
I II NH2
N,
-
H2Ny0
HN
H
0 0 * 031
YTh
0
NH2
reN.,
0
0
0
0 0 Wer5CN
N-% 0
N H F F
11101 rajN N, NH2
H H
reN,
HN
0 \--Th
H2NAb
and a salt of any one thereof, wherein the RX* is a bond, a succinimide
moiety, or a
5 hydrolyzed succinimide moiety bound to a residue of an antibody, wherein
on RX*
represents the point of attachment to the residue of the antibody. In some
embodiments,
RX* comprises a succinamide moiety and is bound to a cysteine residue of an
antibody.
In some embodiments, RX* comprises a hydrolyzed succinamide moiety and is
bound
to a cysteine residue of an antibody.
10 [0439] In certain embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by Formula (I):
4:0
Dx 1
z 0);
189
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
5 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N tO
HN
H
(LyN
0 01
N N, NH2
0
0
rTh
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
10 succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen; and
(b) pembrolizumab.
[0440] In certain embodiments, a method of
treating cancer comprises
15 administering:
(a) an antibody conjugate represented by
Formula (I):
= = DX)]
z (J);
wherein:
A is an antibody;
20 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
190
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
L and Dx have a structure of:
H2N y0
HN
H H
H
0 . Ckõ_õNrDcj,N
NH2
C)
N.__
11
H
N
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein ) on RX*
represents
5
the point of attachment to the residue of the
antibody, wherein the antibody binds to a
tumor associated antigen;
(b) pembrolizumab; and
(c) fam-trastuzumab deruxtecan-nx1d.
[04411 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4, 4, Dx i 1
z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
191
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
\111
NH2
0 H 0 I.
N,
/Th
or a salt thereof, wherein the RX+ is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(13) pembrolizumab; and
(c) ado-trastuzumab emtansine.
[0442] In further embodiments, a method of
treating cancer comprises
administering:
10 (a) an antibody conjugate represented by Formula (I):
4) = DX)]
z (i);
wherein:
A is an antibody;
n is one;
15 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
192
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2NtO
HN
oL
NH2
0 411
0 N,
11
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) pembrolizumab; and
(c) a chemotherapy agent comprising pemetrexed; and
(d) a chemotherapy agent comprising carboplatin; and
optionally wherein the cancer is a non-small cell lung cancer (NSCLC).
10 104431 In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = x 1
z 0);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
193
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2NtO
HN
oL
ARX1111-
NH2
0 411
0 N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) pembrolizumab; and
(c) a chemotherapy agent comprising pemetrexed; and
(d) a chemotherapy agent comprising cisplatin; and
optionally wherein the cancer is a NSCLC.
10 104441 In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = x 1
z 0);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
194
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1-12Ny0
HN
H
ARc irwNiNli.N
0 H 0 SIOYNOOLN
NH2
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RIC* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) atezolizumab;
(c) bevacizumab;
(d) a chemotherapy agent comprising carboplatin; and
(e) a chemotherapy agent comprising paclitaxel; and
10 optionally wherein the cancer is a NSCLC.
[0445] In certain embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
z (J);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
195
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
NH2
0 H 0 le 01/2.,õ,NalN
N,
0
0 \--I\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein "1- on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen; and
(b) nivolumab.
[04461 In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= =
x
1
10 z (I);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
15 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
N2Ny0
HN
0
y-3.1(
oaN 0
ISC
NH2
0 H 0 *
N,
o
0
196
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
5 (13) trastuzumab.
[04471 In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
15 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure Of:
H2 Ny0
HN
ARx<yHNA H
NIH2
0 le 0.õ,e,Nraj 0
N,
1,4
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -44 on RX*
represents
20
the point of attachment to the residue of the
antibody, wherein the antibody binds to a
HER2 antigen; and
(b) fam-trastuzumab deruxtecan-nxki.
197
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0448] In certain embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
4) = x 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N tO
HN
H
0
r.....31.õ1
0
N, N H2
0 H
0
\---\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein t on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
(b) ado-trastuzumab emtansine.
104491 In certain embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
x 1 1
z (J);
198
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
5 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny.0
HN
0
1111;\,11
NH2
0 H 0140
0
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
10 succinimide moiety bound to a residue of an antibody, wherein .-st on
RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
(b) tucatinib.
104501 In certain embodiments, a method of
treating a HER2-expressing cancer
15 comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1
z (J);
wherein:
A is an antibody;
20 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
199
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
L and Dx have a structure of:
FuNto
HN
H
0
N NH2
11
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
5
the point of attachment to the residue of the
antibody, wherein the antibody binds to a
HER2 antigen; and
(b) palbociclib.
104511 In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
10 (a) an antibody conjugate represented by Formula (I):
1
z 0);
wherein:
A is an antibody;
n is one;
15 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
200
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N tO
HN
0
0 0
0 10 OGL1 N N, NH2
11
0
0 \---\
or a salt thereof, wherein the RX+ is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen; and
(b) abemaciclib.
[04521 In certain embodiments, a method of
treating HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
4) = 1
10 z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
15 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny0
HN
H
Nts%11 N
raj N
ON
, NH2
0
0
201
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --A on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
5 (b) ribociclib.
[04531 In further embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
15 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2NtO
HN
H
NH
0 0 40
N, 2
0
0 \Th
or a salt thereof, wherein the RS is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --A on RX*
represents
20
the point of attachment to the residue of the
antibody, wherein the antibody binds to a
HER2antigen;
(b) trastuzurnab; and
202
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(c) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
[0454] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
1
z 0);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Nyo
HN
ARx<
H
0 41) NH2
0
N,
o
0
r¨\\

or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
203
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
optionally wherein the cancer is a gastric cancer.
[0455] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= =
1
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N tO
HN
H
0 411ONOLA N, NH2
0
11
0
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
204
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[04561 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny0
HN
H
0 0 40OYFIOL1N

0
NH2
N,
/Th
0 \---\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
1104571
In further embodiments, a method
of treating HER2-expressing cancer
comprises administering:
205
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(a) an antibody conjugate represented by
Formula (I):
= 1
Z (J):
wherein:
A is an antibody;
5 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N
yO
HN
0
(111-n-
0, NaIN
NH2
0
N,
0
10 0
o
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
FIER2 antigen;
15 (b) trastuzumab; and
(c) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
cisplatin; and
optionally wherein the cancer is a gastric cancer.
[0458] In further embodiments, a method of
treating cancer comprises
20 administering:
(a) an antibody conjugate represented by
Formula (I):
206
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
z (J);
wherein:
A is an antibody;
n is one;
5 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N y0
HN
0
ARC IN,,A111
0
NH2
0 0 40
N,
0
10 or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
15 (c) pembrolizumab; and
(d) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
cisplatin; and
optionally wherein the cancer is a gastric cancer.
[04591 In further embodiments, a method of
treating cancer comprises
20 administering:
(a) an antibody conjugate represented by
Formula (I):
207
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
z (J);
wherein:
A is an antibody;
n is one;
5 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
1-12N
HN
Ha\ H
NH
0 H 0
N, 2
0
0
10 or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -S. on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
15 (c) nivolumab; and
(d) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
cisplatin; and
optionally wherein the cancer is a gastric cancer.
[0460] In further embodiments, a method of
treating cancer comprises
20 administering:
(a) an antibody conjugate represented by
Formula (I):
208
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
z (J);
wherein:
A is an antibody;
n is one;
5 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
1-0 y0
HN
0
ThCY11;11
NH2
0 _pt.-L.__ H 0
N,
11
0
f-Th
0
10 or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
15 (c) atezolizumab; and
(d) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
cisplatin; and
optionally wherein the cancer is a gastric cancer.
1104611 In further embodiments, a method of
treating a HER2-expressing cancer
20 comprises administering:
(a) an antibody conjugate represented by
Formula (I):
209
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
z (J);
wherein:
A is an antibody;
n is one;
5 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
ii2N y0
HN
H
Rn0 NH2
0 11LT 110
N,
n
0
0
10 or a salt thereof, wherein the RX+ is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to
ITER2 antigen;
(b) trastuzumab; and
15 (c) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
104621 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= =
DX

1
20 z (I):
wherein:
210
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
5 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N y0
HN
0 140
NHz
0
N,
0
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
10
the point of attachment to the residue of the
antibody, wherein the antibody binds to a
tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
optionally wherein the cancer is a gastric cancer.
15 [0463] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
=
z 0);
wherein:
20 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
211
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N y0
HN
N
i.HA,,,õ
1 0
NH2
H
0 X
0 1110 0.........NratC---.N N,
ll
H
0
N
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
5
succinimide moiety bound to a residue of an
antibody, wherein Y on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
10 optionally wherein the cancer is a gastric cancer.
[04641 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = Dx 1 1
z (J);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
212
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
H
ARdcwyNiNir.N
0 H 0 SI oyNiN
NH2
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RIC* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolumab; and
optionally wherein the cancer is a gastric cancer.
1104651 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) =
1
z (J);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure Of:
213
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N yO
HN
Ha\rH
NH2
0 H 0 le
NOLAN N,
0
LkN0 \--- re\
\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0466] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = DX)]
Z (i);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
214
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N tO
HN
ARIY
0
INI
mN 0
NH2
0 0 00
N,
0 \-\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -)4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0467] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = x 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
215
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N tO
HN
0
IR lit\ 011
m INI N 0
.,N H2
0 0
N N
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -)4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0468] In further embodiments, a method of treating a FIER2-
expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
4) = Dx 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
216
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N tO
HN
ARIY
0
INI
mN 0
NH2
0 0 00
.N
11
0
0 \-\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -)4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2antigen;
(b) trastuzumab; and
(c) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
[0469] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= Dm /1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
217
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
ARC
H
0 H 1411
0OyN1lLiflycyNNH2

,
0
0 \-- re\
\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(13) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 1104701 In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
=
x
1
z 0);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
218
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
RAH
N N
N
1--n N
NH2
0 A H 0 SO
11
H
0
N
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -$ on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0471] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = Dx / 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and D. have a structure of:
219
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny.0
HN
0
rsly:1)Yri
cuN 0
NH2
0 0
0
0 \---\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --A on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0472] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
Dx 1 1
Z (0;
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
220
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N tO
HN
ARIY
0
INI
mN 0
NH2
0 0 00
N,
0 \-\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -)4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising docetaxel; and
optionally wherein the cancer is a breast cancer.
10 [0473] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = x 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
221
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N yO
HN
Ha\ H
NH2
0 H 0 le
NOLAN N,
0
LkN0 \--- re\
\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising paclitaxel; and
optionally wherein the cancer is a breast cancer
10 [0474] In further embodiments, a method of treating a HER2-
expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
4) = DX)]
Z (i);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
222
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
RCY
RAH
NH2
0 A H 0 SIP
N,
11
0
r-\\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -$ on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 BER2 antigen;
(b) trastuzumab;
(c) palbociclib; and
(d) a hormone-based compound selected from any one or more of
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
10 enzalutamide, apalutamide, darolutamide, degarelix, toremifene,
goserelin, triptorelin,
histrelin, leuprolide, and tamoxifen citrate; and
optionally wherein the cancer is a breast cancer.
[0475] In further embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
15 (a) an antibody conjugate represented by Formula (I):
1
z 0);
wherein:
A is an antibody;
n is one;
20 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
223
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
0 ky N, NH2
140N 0
0
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen;
(b) trastuzumab;
(c) abemaciclib; and
(d) a hormone-based compound selected from any one or more of
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
10 enzalutamide, apalutamide, darolutamide, degarelix, toremifene,
goserelin, triptorelin,
histrelin, leuprolide, and tamoxifen citrate; and
optionally wherein the cancer is a breast cancer.
[0476] In further embodiments, a method of
treating cancer comprises
administering:
15 (a) an antibody conjugate represented by Formula (I):
4) = DX)]
Z (0;
wherein:
A is an antibody;
n is one;
20 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
224
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
.,ilN
0
0 H 0 I.
N1N12
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein "1- on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab;
(d) atezolizumab; and
(e) a chemotherapy agent comprising docetaxel; and
10 optionally wherein the cancer is a breast cancer.
[0477] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
=
z (J);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
225
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
ARc
H
0 H
0
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(13) trastuzumab;
(c) pembrolizumab;
(d) atezolizumab; and
(e) a chemotherapy comprising paclitaxel; and
10 optionally wherein the cancer is a breast cancer.
[0478] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1
z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
226
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
H
H
NH2
0 0 *
N,
0
0 \-\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --1( on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) fam-trastuzumab deruxtecan-nxki; and
(c) durvalumab; and
optionally wherein the cancer is a breast cancer.
1104791 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) = DX)]
z (J);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
227
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N y0
HN
=
n
0 NH2
0 H 0
OyNXNN,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RIC* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) fam-trastuzumab deruxtecan-nxki; and
(c) atezolizumab; and
optionally wherein the cancer is a breast cancer
1104801 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) =
1
z (J);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
228
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N yO
HN
Ha\ H
NH2
0 H 0 le
NOLAN N,
0
LkN0 \--- re\
\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) ado-trastuzumab emtansine; and
(c) durvalumab; and
optionally wherein the cancer is a breast cancer
[0481] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1
z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
229
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N TO
HN
0
0
N H2
0 0 140
N ,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) ado-trastuzumab emtansine; and
(c) atezolizumab; and
optionally wherein the cancer is a breast cancer.
[0482] In any of the aforementioned
embodiments having a linker-payload (L-
10 Dx) structure of
H2Ny0
HN
H
NH2
0 H 0
N,
0
Nc
0
the antibody comprises HC-CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and
LC-CDR3 of SEQ ID NOs: 1-6, respectively; or the antibody comprises a heavy
chain
variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light
chain
15 variable region comprising the amino acid sequence of SEQ ID NO: 8; or
the antibody
comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and
a
light chain comprising the amino acid sequence of SEQ ID NO: 10. In some
embodiments, RX* comprises a succinamide moiety and is bound to a cysteine
residue
of an antibody construct. In some embodiments, RX* comprises a hydrolyzed
20 succinamide moiety and is bound to a cysteine residue of an antibody
construct.
230
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0483] In certain embodiments, a method of treating cancer comprises
administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny0
HN
H
1-1RXCINAr"
0 40
0
11
0
o
*
NH2
0
N,
rTh
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --44 on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen; and
(b) pembrolizumab.
[0484] In certain embodiments, a method of treating cancer comprises
administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z 0);
231
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
5 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H,Nto
HN-I
H
1%1RXIN)NTI
LIN
0
0 H 40
rxiN
0 OyNõ,...õ,--..N
NH2
0
N I N,
0 \
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
10 succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) pembrolizumab; and
(c) fam-trastuzumab deruxtecan-nxId.
15 [04851 In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
1
z 0);
wherein:
20 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
232
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
L and Dx have a structure of:
H,Nto
HN
ERXIEllYLYN
0
0 H le 0y rl
110 0 NH
0N
0 N\----\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
5 the point of attachment to the residue of the antibody, wherein the
antibody binds to a
tumor associated antigen;
(b) pembrolizumab; and
(c) ado-trastuzumab emtansine.
[04861 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1
z imp;
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
233
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
511 ::11:11rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) pembrolizumab; and
(c) a chemotherapy agent comprising pemetrexed; and
(d) a chemotherapy agent comprising carboplatin; and
optionally wherein the cancer is a NSCLC.
10 [0487] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
234
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) pembrolizumab; and
(c) a chemotherapy agent comprising pemetrexed; and
(d) a chemotherapy agent comprising cisplatin; and
optionally wherein the cancer is a NSCLC.
10 [0488] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
235
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
511 ::11:11rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) atezolizumab;
(c) bevacizumab;
(d) a chemotherapy agent comprising carboplatin; and
(e) a chemotherapy agent comprising paclitaxel; and
10 optionally wherein the cancer is a NSCLC.
[04891 In certain embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
236
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
::11:11HN1re1ll
0
OP
0 0 OyN..N
0
NH
2
0
=N N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen; and
(b) nivolumab.
104901 In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1 1
10 z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
15 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H,Nto
HN
O H
-.ru 1.. tri 40
0
0
NH2
0
===,..,.AN N,
ra\
0
237
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
5 (13) trastuzumab.
[0491] In certain embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
15 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny0
HN
H1 H
FRXIN
0
HN 40
0 0 Oy
so a t iN
NH2
0
N,
rTh
0
I\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -$ on RX*
represents
20 the point of attachment to the residue of the antibody, wherein the
antibody binds to a
ITER2 antigen; and
(b) fam-trastuzumab deruxtecan-nxki.
[0492] In certain embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
238
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
(a) an antibody conjugate represented by
Formula (I):
=
Z (J):
wherein:
A is an antibody;
5 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2Ny0
HNI
H
"--RXWYNXLINIIIrN
0 0
NH2
8=N,
10
o
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
15 (b) ado-trastuzumab emtansine.
[0493] In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1 1
z imp;
20 wherein:
A is an antibody;
239
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
5 L and Dx have a structure of:
H2NtO
HN
oL
H
0 410
0
0
0
NH2
N
rThs%
0 \
or a salt thereof, wherein the RX+ is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --A on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
10 HER2 antigen; and
(b) tucatinib.
[0494] In certain embodiments, a method of
treating a BER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= DX)]
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
20 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and D. have a structure of:
240
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
::11:11HN1re1ll
0
OP
0 0 OyN..N
0
NH
2
0
=N N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen; and
(b) palbociclib.
104951 In certain embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
= Dx 1 1
10 z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
15 L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H,Nto
HN
O H
-.ru 1.. tri 40
0
0
NH2
0
===,..,.AN N,
ra\
0
241
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen; and
5 (b) abemaciclib.
[0496] In certain embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z (J);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4; and
L and Dx have a structure of:
H2Ny0
HN
H
1RX(14)\'N
0
0
0
0
NH
2
0
rTh

or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein --A on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to
HER2 antigen; and
20 (b) ribociclib.
[0497] In further embodiments, a method of treating a HER2-expressing
cancer
comprises administering:
(a) an antibody conjugate represented by Formula (I):
242
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
1
z (J);
wherein:
A is an antibody;
n is one;
5 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N tO
HN
0
0
HN 0
Oy N
nN.z.1 0
NH
N
2
0
N,
r---
0 N
10 or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen;
(b) trastuzumab; and
15 (c) a chemotherapy regimen comprising leucovorin,
fluorouracil, and
oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
[0498] In further embodiments, a method of
treating cancer comprises
administering:
20 (a) an antibody conjugate represented by Formula (I):
411
x 1
z (J);
wherein:
243
CA 03151322 2022- 3- 15

WO 2021/067644
PCT/US2020/053870
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
5 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H,Nto
HN
H
O H
Oy N
so ijazsa.
I
NI12
0
- r-\
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein )4 on RX*
represents
10 the point of attachment to the residue of the antibody, wherein the
antibody binds to a
tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
15 oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
[0499] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
=
1
20 z (I);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
244
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
H2N tO
HN
H
N:S
FRXC iryW Ny 0 40
0
0 OT
0
NH2
0 \
5 or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein -$ on RX*
represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
tumor associated antigen;
(b) trastuzumab;
10 (c) nivolumab; and
(d) a chemotherapy regimen comprising
leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
optionally wherein the cancer is a gastric cancer.
105001 In further embodiments, a method of
treating cancer comprises
15 administering:
(a) an antibody conjugate represented by
Formula (I):
= 40 Dx 1
z 0);
wherein:
A is an antibody;
20 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
245
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
511 ::11:11rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
oxaliplatin (FOLFOX); and
10 optionally wherein the cancer is a gastric cancer.
[05011 In further embodiments, a method of
treating a HER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
246
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen;
(b) trastuzumab; and
(c) a chemotherapy regimen comprising leucovorin, fluorouracil, and
cisplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0502] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
247
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
cisplatin; and
10 optionally wherein the cancer is a gastric cancer.
[0503] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
248
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
cisplatin; and
10 optionally wherein the cancer is a gastric cancer.
[0504] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
249
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
511 ::11:11rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy regimen comprising leucovorin, fluorouracil, and
cisplatin; and
10 optionally wherein the cancer is a gastric cancer.
[0505] In further embodiments, a method of
treating a FIER2-expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
250
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
suecinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen;
(b) trastuzumab; and
(c) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
[0506] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
251
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
optionally wherein the cancer is a gastric cancer.
[0507] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
252
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
optionally wherein the cancer is a gastric cancer.
[0508] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
17),, is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
253
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolumab; and
optionally wherein the cancer is a gastric cancer.
[0509] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
254
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0510] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
255
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0511] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
256
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy comprising capecitabine; and
optionally wherein the cancer is a gastric cancer.
10 [0512] In further embodiments, a method of treating a HER2-
expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
257
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen;
(b) trastuzumab; and
(c) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
[0513] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
258
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0514] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
259
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) nivolumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0515] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
260
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) atezolizumab; and
(d) a chemotherapy regimen comprising capecitabine and oxaliplatin; and
optionally wherein the cancer is a gastric cancer.
10 [0516] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
261
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising docetaxel; and
optionally wherein the cancer is a breast cancer.
10 [0517] In further embodiments, a method of treating cancer
comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
262
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2N to
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab; and
(d) a chemotherapy comprising paclitaxel; and
optionally wherein the cancer is a breast cancer.
10 [0518] In further embodiments, a method of treating a HER2-
expressing cancer
comprises administering:
(a) an antibody conjugate represented by
Formula (I):
4) 1
z (J);
wherein:
15 A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
20 L and Dx have a structure of:
263
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
::11:11HN1re1ll
0
OP
0 0 OyN..N
0
NH
2
0
=N N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
HER2 antigen;
(b) trastuzumab;
(c) palbociclib; and
(d) a hormone-based compound selected from any one or more of
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
enzalutamide, apalutamide, darolutamide, degarelix, toremifene, goserelin,
niptorelin,
histrelin, leuprolide, and tamoxifen citrate; and
optionally wherein the cancer is a breast cancer.
[05191 In further embodiments, a method of
treating a HER2-expressing cancer
comprises administering
(a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z 0);
wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
264
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
::11:11HN1re1ll
0
OP
0 0 OyN..N
0
NH
2
0
=N N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 HER2 antigen;
(b) trastuzumab;
(c) abemaciclib; and
(d) a hormone-based compound selected from any one or more of
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
10 enzalutamide, apalutamide, darolutamide, degarelix, toremifene,
goserelin, triptorelin,
histrelin, leuprolide, and tamoxifen citrate; and
optionally wherein the cancer is a breast cancer.
1105201 In further embodiments, a method of
treating cancer comprises
administering:
15 (a) an antibody conjugate represented by Formula (I):
4) = Dx 1
z 0);
wherein:
A is an antibody;
n is one;
20 z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound, wherein:
L and Dx have a structure of:
265
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab;
(d) atezolizumab; and
(e) a chemotherapy agent comprising docetaxel; and
10 optionally wherein the cancer is a breast cancer.
[05211 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
266
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) trastuzumab;
(c) pembrolizumab;
(d) atezolizumab; and
(e) a chemotherapy comprising paclitaxel; and
10 optionally wherein the cancer is a breast cancer.
[05221 In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
DX)]
Z 0);
15 wherein:
A is an antibody;
n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
20 Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
267
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) fam-trastuzumab deruxtecan-nxki; and
(c) durvalumab; and
optionally wherein the cancer is a breast cancer.
[0523] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
268
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) fam-trastuzumab deruxtecan-nxki; and
(c) atezolizumab; and
optionally wherein the cancer is a breast cancer.
[0524] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
269
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Nto
HN
0
5::11:1111rell
0
HN 0 410
0
NH
0
2
0
N,
0
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) ado-trastuzumab emtansine; and
(c) durvalumab; and
optionally wherein the cancer is a breast cancer.
[0525] In further embodiments, a method of
treating cancer comprises
administering:
(a) an antibody conjugate represented by
Formula (I):
4) 4> DX)]
Z 0);
wherein:
A is an antibody;
15 n is one;
z is selected from 2 to about 8 or from 2 to about 4;
L is the linker; and
Dx is the immune-stimulatory compound; wherein:
L and Dx have a structure of:
270
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0
PlyLiJy.
0
HN 0 0 OP
OyN.N is
0
NH2
0
N,
or a salt thereof, wherein the RX* is a bond, a succinimide moiety, or a
hydrolyzed
succinimide moiety bound to a residue of an antibody, wherein
on RX* represents
the point of attachment to the residue of the antibody, wherein the antibody
binds to a
5 tumor associated antigen;
(b) ado-trastuzumab emtansine; and
(c) atezolizumab; and
optionally wherein the cancer is a breast cancer.
[0526] In any of the aforementioned
embodiments having a linker-payload
10 structure of
H2N y0
HN
0
irly HN L1)1( -s
ERX.1 0 so
Oy
NH2
0
the antibody comprises HC-CDR1, HC-CDR2, HC-CDR3, LC-CDR1, LC-CDR2, and
LC-CDR3 of SEQ ID NOs: 1-6, respectively; or the antibody comprises a heavy
chain
variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light
chain
15 variable region comprising the amino acid sequence of SEQ ID NO: 8; or
the antibody
comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and
a
light chain comprising the amino acid sequence of SEQ ID NO: 10. In some
embodiments, RX* comprises a succinamide moiety and is bound to a cysteine
residue
of an antibody construct. In some embodiments, RX* comprises a hydrolyzed
20 succinamide moiety and is bound to a cysteine residue of an antibody
construct.
271
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
105271 In any of the aforementioned embodiments, the cancer may be a HER2-
expressing cancer, such as breast cancer, stomach/gastric cancer, colorectal
cancer, non-
small cell lung cancer (NSCLC), urothelial cancer, endometrial cancer, or
ovarian
cancer.
5 105281 In various embodiments, methods of treating HER2-expressing
cancer
are provided, comprising administering a conjugate provided herein, wherein
the
conjugate comprises an antibody that binds HER2, and an additional therapeutic
agent
comprising a kinase inhibitor, a therapeutic agent comprising an anti-HER2
antibody,
or both the tyrosine kinase inhibitor and therapeutic agent comprising an anti-
HER2
10 antibody. In some embodiments, the anti-HER2 antibody of the conjugate
comprises
the CDRs of pertuzumab, antigen binding fragment (e.g., variable regions) of
pertuzumab, light and heavy chains of pertuzumab. In some embodiments, the
anti-
HER2 antibody of the conjugate comprises the CDRs of trastuzumab, antigen
binding
fragment (e.g., variable regions) of trastuzumab, light and heavy chains of
trastuzumab.
15 105291 In some such embodiments, the anti-HER2 antibody of the
conjugate
comprises heavy chain (HC)-CDR1, HC-CDR2, HC-CDR3, light chain (LC)-CDR1,
LC-CDR2, and LC-CDR3 of SEQ ID NOs: 1-6, respectively. In further embodiments,

the anti-HER2 antibody of the conjugate comprises a heavy chain and light
chain,
wherein: (a) the heavy chain comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ
20 ID NOs: 1-3, respectively, and comprises a heavy chain variable region
(VH) having an
amino acid sequence that has at least 90%, at least 91%, at least 92%, at
least 93%, at
least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least
99%, at least
99.5%, or 100% identity to the VH amino acid sequence of SEQ ID NO: 7; and (b)
the
light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3 of SEQ ID NOs: 4-6,
25 respectively, and a light chain variable region (VI.) having an amino
acid sequence that
has at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at
least 95%, at
least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100%
identity to
the VL amino acid sequence of SEQ ID NO: 8. In still further embodiments, the
anti-
HER2 antibody of the conjugate comprises a VH comprising or consisting of the
amino
30 acid sequence of SEQ ID NO: 7 and a NIL comprising or consisting of the
amino acid
sequence of SEQ ID NO: 8. In yet further embodiments, the anti-HER2 antibody
of the
272
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
conjugate comprises a heavy chain and light chain, wherein: (a) the heavy
chain
comprises HC-CDR1, HC-CDR2, and HC-CDR3 of SEQ ID NOs: 1-3, respectively,
and comprises an amino acid sequence that has at least 90%, at least 91%, at
least 92%,
at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least
98%, at least
5 99%, at least 99.5%, or 100% identity to the heavy chain amino acid
sequence of SEQ
ID NO: 9; and (b) the light chain comprises LC-CDR1, LC-CDR2, and LC-CDR3 of
SEQ ID NOs: 4-6, respectively, and an amino acid sequence that has at least
90%, at
least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least
96%, at least
97%, at least 98%, at least 99%, at least 99.5%, or 100% identity to the light
chain
10 amino acid sequence of SEQ ID NO: 10. In yet further embodiments, the
anti-HER2
antibody of the conjugate comprises a heavy chain comprising or consisting of
the
amino acid sequence of SEQ ID NO: 9 and a light chain comprising or consisting
of the
amino acid sequence of SEQ ID NO: 10.
[0530] In some such embodiments, the method comprises administering a
15 conjugate comprising an antibody that binds HER2 and an additional
therapeutic agent
comprising an anti-HER2 antibody, wherein the anti-HER2 antibody of the
conjugate
and the anti-HER2 antibody of the additional therapeutic agent bind different
epitopes
of HER2. For example, the anti-HER2 antibody of the conjugate may comprise
pertuzumab or the CDRs or antigen binding fragment (e.g., variable regions of
20 pertuzumab), while the anti-HER2 antibody of the additional therapeutic
agent
comprises trastuzumab or a biosimilar thereof, or the CDRs or antigen binding
fragment
(e.g., variable regions of trastuzumab).
[0531] In any of the aforementioned embodiments, the additional therapeutic
agent is selected from trastuzumab, trastuzumab-qyyp, trastuzumab-pkrb,
trastuzumab-
25 dub, trastuzumab-anns, trastuzumab-dkst, trastuzumab deruxtecan-nxki,
ado-
trastuzumab emtansine, trastuzumab duocarmazine, margetuximab, and
zenocutuzumab
(MCLA-128 (Merus)).
[0532] In some embodiments, the additional therapeutic agent is a kinase
inhibitor selected from a tyrosine kinase inhibitor and a serinSthreonine
kinase
30 inhibitor. In some embodiments, the tyrosine kinase inhibitor is
tucatinib, cabozantinib,
afatinib, erlotinib, pyrotinib, neratinib, poziotinib, dacomitinib, gefitinib,
lapatinib,
273
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
osimertinib, larotrectinib, axitinib, lenvatinib, pazopanib, regorafenib, or
sunitinib. In
some embodiments, the serine/threonine kinase inhibitor is a CDK4/6 inhibitor,
such as
abemaciclib (Verzenio0), palbociclib (Ibrance0), or ribociclib (Kisqali0).
105331 In some embodiments, the additional therapeutic agent is a small
5 molecule tyrosine kinase inhibitor that inhibits HER2. In some
embodiments, the
additional therapeutic agent is selected from tucatinib, cabozantinib,
pyrotinib,
neratinib, and poziotinib.
[0534] In some embodiments, the additional therapeutic agents further
comprises a chemotherapy agent, an immune checkpoint inhibitor, a biologic
therapy
10 such as a therapeutic antibody, a molecularly targeted therapy,
chemotherapy protective
agent, or any combination thereof.
[0535] In some such embodiments, the immune checkpoint inhibitor is an
antibody. In some such embodiments, the immune checkpoint inhibitor is an
inhibitor
of PD-1 and/or PD-L1, e.g., an anti-PD-1 and/or anti-PD-Li antibody. Examples
of
15 anti-PD-1 and/or anti-PD-Li antibodies include pembrolizumab, nivolumab,
cerniplimab, avelumab, durvalumab, and atezolizumab, or a biosimilar thereof.
In some
embodiments, the immune checkpoint inhibitor is an anti-PD1 antibody selected
from
pembrolizumab, nivolumab, and cemiplimab, or a biosimilar thereof In some
embodiments, the immune checkpoint inhibitor is an anti-PD-Ll antibody
selected from
20 avelumab, durvalumab, and atezolizumab, or a biosimilar thereof.
[0536] In some such embodiments, the chemotherapy agent comprises a
hormone-based compound, optionally wherein the hormone based compound
comprises
anastrozole, exemestane, letrozole, fulvestrant, bicalutamide, flutamide,
nilutamide,
enzalutamide, apalutamide, darolutamide, degarelix, toremifene, goserelin,
triptorelin,
25 histrelin, leuprolide, or tamoxifen citrate.
[0537] In some such embodiments, the chemotherapy agent comprises a
platinum coordination complex, optionally wherein the platinum coordination
complex
comprises cisplatin, oxaliplatin, or carboplatin.
[0538] In some such embodiments, the chemotherapy agent comprises a folic
30 acid analog, optionally wherein the folic acid analog comprises
pemetrexed,
methotrexate, or trimetrexate.
274
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
[0539] In some such embodiments, the chemotherapy agent comprises a
pyrimidine analog, optionally wherein the pyrimidine analog comprises
fluorouracil,
capecitabine, fluorodeoxyuridine, tezacitabine, gemcitabine, cytosine
arabinoside,
cytarabine, 5-azacytidine, or 2,2'-difluorodeoxycytidine.
5 [0540] In some such embodiments, the chemotherapy agent comprises
a
microtubule inhibitor, optionally wherein the microtubule inhibitor comprises
paclitaxel, docetaxel, and eribulin.
[0541] In some such embodiments, the chemotherapy protective agent
comprises leucovorin (folinic acid), allpuronol, rasburicase, amifostine,
dexrazoxane, or
mesna.
[0542] In any of the aforementioned embodiments, the linker-payload
compound of the conjugate has a structure selected from:
H2Ny.0
HN
H
N__ NH2
0 401 0.1/4__CUN
0
\
0 \
H2N tO
HN
ti 0
N
N 0
N
0 0 40 0 NI
N, H2
o
/Th
0
0
ti4
I N, NH2
0 NxIA
'X/ 0 OANThr
n 0
N
r--
H H
0
=
15 H2No
275
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
0 N
0
11
A %Li,
0 yr. H 0 0 0 N
N
H
lar RXIAN H
0 H r N N ' N¨ \
H 0
0 \ ---- \
N H
A
0 NH2
H2Ny0
'
HN
0
INIAIRII
0
t'RXii N
o 0
H H
0
Na.õN
a..
Oy N.....õ,...Th 0
I 1/2% 0 NH2
0
H N,
N
H
---- Nr-\\
0 \Thl
H2N y0
HN
0
Mt\ II
is---Rx.--:--------------y N
o
0 o
H H
0 OyN.,...-
...N
H ACN N 0
I),

N
H
çJ
- r-=
IN

0 \--"N
H2N 1/4,r0
HN
0
kilx13y M
e(RX'<WY N
H
0 0 0 0 rl
0
N
tli U
N,
N
HLJ-- r-\\
N
0
0
0
4 4AnCri 0 OC., 0 .. NH2
VR).(1.'NAN reN N F F
N ,-- IC
N L3E

H 0H H
-- Nr---\
HN
0 \Th
H2NAo
,
276
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
and a salt of any one thereof, wherein the RX* is a bond, a succinimide
moiety, or a
hydrolyzed succinimide moiety bound to a residue of an antibody, wherein "A on
RX*
represents the point of attachment to the residue of the antibody. In some
embodiments,
RX* comprises a succinamide moiety and is bound to a cysteine residue of an
antibody.
5 In some embodiments, RX* comprises a hydrolyzed succinamide moiety and is
bound
to a cysteine residue of an antibody.
105431 In some embodiments, RX* comprises a succinamide moiety and is
bound to a cysteine residue of an antibody construct.
105441 In some embodiments RX* comprises a hydrolyzed succinamide moiety
10 and is bound to a cysteine residue of an antibody construct.
105451 In certain aspects, the instant disclosure provides a method of
treating
HER2-expressing cancer comprising administering:
(a) a conjugate having the following
structure:
0
0
r
Orsji.1/244. N
I-IN
15 or a pharmaceutically acceptable salt thereof, wherein:
Ab is an antibody, D is a TLR7 agonist of formula:
NT-1
N
or%-
If
wherein le is an alkyl, heteroallcyl, cycloalkyl, heterocycloalkyl, aryl,
heteroaryl,
arylalkyl, or heteroarylalkyl group comprising from 1 to 8 carbons, each J is
hydrogen,
277
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
each U is N, each t is 2, Q is not present, the dashed line represents a point
of
attachment of D to GI, and GI is a bond;
subscript a is an integer from 1 to 40; and
subscript r is an integer from 1 to 10; and
5 (b) a therapeutic antibody, a kinase inhibitor, a
chemotherapy agent, or any
NH2
N g Nt)__rj
11 1-1
N
(N)
combination thereof. In certain embodiments, D is -4-
.
105461 In certain embodiments, the instant disclosure provides a method of
treating HER2-expressing cancer comprising administering:
(a) a conjugate having the following structure:
r
Q
0
Tht--N mt.t ..\\, tirren-----o-iaNNArialeNe-AN
H Ab
1
112N N.-.Nte
I
i
4 r
10
,
wherein Ab is trastuzumab; and
(b) a PD-1 and/or PD-L1 inhibitor, optionally wherein the PD-1 and/or PD-
Li inhibitor comprises one or more of pembrolizumab, atezolizumab, and
durvalumab.
105471 The combination therapies provided herein can be used in the methods
15 described herein as a therapeutic, for example, as a treatment that can
be administered
in an effective regimen to a subject in need thereof to achieve a therapeutic
effect. A
therapeutic effect can be obtained in a subject by reduction, suppression,
remission,
alleviation or eradication of a disease state, including, but not limited to,
one or more
symptoms thereof A therapeutic effect in a subject having a disease or
condition, or
20 exhibiting an early symptom thereof or exhibiting or otherwise suspected
of being in or
approaching an early stage of a disease or condition, can be obtained by a
reduction, a
suppression, a prevention, a delay, a remission, an alleviation or an
eradication of the
condition or disease, or pre-condition or pre-disease state. In various
embodiments, the
278
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
method comprises administering an effective regimen that results in a Tmax of
the
conjugate of greater than 4 hours following each administration of the
conjugate. In
some embodiments, the effective regimen results in a Tmax greater than 6
hours,
greater than 8 hours, greater than 10 hours, greater than 12 hours, or greater
than 15
5 hours following each administration of the conjugate.
[0548] In certain embodiments, the methods include administration of an
immune-stimulatory conjugate, or a pharmaceutical composition thereof, to a
subject in
need thereof in an effective regimen to activate, stimulate or augment an
immune
response against a disease treatable with a TLR agonist (e.g., cancer or a
viral disease).
10 The polypeptide of the conjugate recognizes an antigen associated with
the disease or
disease state, such as a tumor-associated antigen or liver cell antigen.
[0549] In certain embodiments, the cancer to be treated with the therapeutic
combinations provided herein is a solid tumor, such as a sarcoma, a carcinoma
or
lymphoma. In some such embodiments, the antibody of the conjugate recognizes
an
15 antigen on the target cells, such as a tumor-associated antigen. In some
embodiments,
the cancer is a HER2-expressing cancer and the antibody of the conjugate binds
HER2.
In some embodiments, the HER2-expressing cancer is selected from breast
cancer,
stomach/gastric cancer, colorectal cancer, non-small cell lung cancer (NSCLC),

urothelial cancer, endometrial cancer, and ovarian cancer. In some aspects,
the HER2
20 expressing cancer expresses HER2 at a level of 2+ or 3+ as determined by
immunohistochemistry
105501 In certain embodiments, the methods include administration of a
combination therapy comprising a conjugate provided herein to a subject in
need
thereof to activate, stimulate or augment an immune response against tumor
cells of a
25 solid tumor, such as brain, breast, lung, liver, kidney, pancreatic,
colorectal, ovarian,
head and neck, bone, skin, mesothelioma, bladder, stomach/gastric, prostate,
thyroid,
uterine or cervical/endometrial cells. In some such embodiments, the
polypeptide of the
conjugate recognizes an antigen on the tumor cells.
105511 In some cases, treatment comprises reduced tumor growth. In some
30 cases, treatment comprises tumor arrest. In some cases, combination
therapy
279
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
comprising a conjugate provided herein allows for dose reduction of an
accompanying
chemotherapy agent.
[0552] In certain embodiments, the methods include administration of a
combination therapy comprising a conjugate provided herein (e.g., including a
TLR8
5 agonist antibody conjugate specific for a liver cell antigen, such as a
liver cell antigen
expressed on a liver cell infected with a virus) to a subject in need thereof
to activate,
stimulate or augment an immune response against viral infection, such as a
chronic viral
infection (e.g., HBV, HCV). In some embodiments, the liver cell antigen is a
hepatocyte antigen. In further embodiments, the liver cell antigen is ASGR1
10 (asialoglycoprotein receptor 1), ASGR2 (asialoglycoprotein receptor 2),
TRF2,
UGT1A1, SLC22A7, SLC13A5, SLC22A1, or C9. In particular embodiments, the liver

cell antigen is ASGR1, ASGR2, or TRF2.
[0553] One of ordinary skill in the art would understand that the amount,
duration and frequency of administration of a combination therapy described
herein to a
15 subject in need thereof depends on several factors including, for
example but not
limited to, the health of the subject, the specific disease or condition of
the subject, the
grade or level of a specific disease or condition of the subject, the
additional
therapeutics the subject is being or has been administered, and the like.
[0554] In some aspects of practicing the methods described herein, the
20 conjugates are administered in an effective regimen of at least two or
at least three
cycles. Each cycle can optionally include a resting stage between cycles.
Cycles of
administration can be of any suitable length. In some embodiments, each cycle
is a
week (7 days), 10 days, every two weeks (14 days or biweekly), every three
week (21
days) or every four weeks (28 days). In some embodiments, each cycle is a
month. In
25 some embodiments, at least two doses of the immune-stimulatory conjugate
are
administered more than 7 days apart, or more than 10 days apart. In some
embodiments, at least one dose of the conjugate is administered more than 7
days, or
more than 10 days, after the initial dose of the conjugate.
[0555] In certain embodiments, the total dose of the conjugate within a cycle
is
30 from about 0.1 to about 10 mg/kg. In some embodiments, the total dose is
from about
0.5 to about 7.5 mg/kg. In some embodiments, the total dose is from about 0.5
to about
280
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
mg/kg. In some embodiments, the total dose is from about 0.5 to about 4 mg/kg.
In
some embodiments, the total dose is from about 0.5 to about 3.5 mg/kg. In some

embodiments, the total dose is from about 0.5 to about 2 mg/kg.
105551 In some embodiments, an effective regimen comprises at least two
5 cycles of administration of the conjugate to the subject and a total dose
of greater than
0.4 mg/kg of the conjugate per cycle.
05571 Application of immune-stimulatory conjugates described herein shows
substantial benefit in directing a subject's own immune response to cells of a
particular
site of disease or disorder, such as cells associated with the disease or
disorder.
10 Activating or stimulating an immune response directed to targeted cells
facilitates the
reduction, inhibition of proliferation, inhibition of growth, inhibition of
progression,
inhibition of metastasis or otherwise inhibition up to and including in some
cases
clearance of the targeted cells. Thus, in some cases a targeted immune
response
activation or stimulation leads to inhibition of disease progression, or
alleviation of at
15 least one symptom of a manifest disease in a patient, up to and in some
cases including
complete elimination of from one symptom to an entire disease state in a
subject.
05581 In particular, the methods disclosed herein are well suited for use with

immune stimulatory conjugates, such as immune stimulatory conjugates that
direct an
immune response in a subject to a particular disorder or disease location,
cell type or
20 cell, Accordingly, practice of some methods herein comprises selection
of a suitable
subject such as a subject to be subjected to or undergoing a treatment with a
conjugate
that directs a ben7a7epine or benzazepine-like compound of the conjugate to a
particular disorder or disease site, cell type or cell. Often, the subject is
selected for
practice of the method due to having at least one symptom of a disease or
disorder, or
25 projected to develop at least one symptom of a disease or disorder (such
as a subject in
remission and at risk for relapse), suitable for treatment by a conjugate as
disclosed
herein. Some diseases are selected not based upon or not based solely on
disease type,
but upon detection or presence of a suitable epitope on a tumor, cell type or
particular
cell that facilitates localization of an immune-stimulatory conjugate to the
epitope.
281
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
EXAMPLES
105591 The following examples are included to further describe some
embodiments of the present disclosure and should not be used to limit the
scope of the
disclosure. The examples are not intended to represent that the experiments
below are
5 all or the only experiments performed. Efforts have been made to ensure
accuracy with
respect to numbers used (for example, amounts, temperature, etc.) but some
experimental errors and deviations should be accounted for. Unless indicated
otherwise, parts are parts by weight, molecular weight is average molecular
weight,
temperature is in degrees Centigrade, and pressure is at or near atmospheric.
10 105601 While aspects of the present disclosure have been shown and
described
herein, it will be apparent to those skilled in the art that such aspects are
provided by
way of example only. Numerous variations, changes, and substitutions will now
occur
to those skilled in the art without departing from the disclosure. It should
be understood
that various alternatives to the aspects of the disclosure described herein
may be
15 employed in practicing the disclosure. It is intended that the following
claims define
the scope of the disclosure and that methods and structures within the scope
of these
claims and their equivalents be covered thereby.
EXAMPLE 1-
HER2-TLR8 CONJUGATE ACTIVATES DENDRITIC CELLS AS MEASURED BY UPREGULATION
20 OF SURFACE MARKERS SUCH AS PD-L1 AND CD86
105611 The expression of activation markers, including CD86, CD40, CD80 and
PD-L1, on human dendritic cells (DCs) following contact with a HER2-TLR8
conjugate in the presence of HER2-expressing tumor cells was determined. HER2-
TLR8 comprises an anti-HER2 antibody comprising the heavy and light chains of
SEQ
25 ID NOs: 9 and 10, respectively, conjugated to a TLR8-linker having the
structure
282
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
H2Ny0
HN
0 0
0
NH2
0 0
0.,w,NOUNN
0
0
NiTh
\--\
Briefly, myeloid DCs were isolated from human blood using the EasySepTh Human
Myeloid DC Enrichment Kit (StemCell Technologies, Catalog #19061). Isolated
dendritic cells were plated with HER2-expressing BT474 tumor cells (ATCC) at a
2:1
5 ratio in the presence of titrating concentrations of HER2-TLR8 conjugate
or HER2
mAb. After 24 hours, the cells were collected, washed and stained on ice with
a
collection of commercially available antibodies conjugated to fluorophores
directed
against PD-L1, CD86, CD80, CD40, CD45, and HLA-DR per the manufacturer's
recommended protocol. After washing to remove unbound antibody-fluorophore
10 molecules, the stained cells are subjected to FACS analysis using a
Celesta flow
cytometer (BD Biosciences) with gating on live cells. The output was analyzed
by
FlowJo v10.2 software (FlowJo LLC).
[0562] The addition of HER2-TLR8 conjugate to the co-culture assays
containing BT474 cells resulted in the upregulation of all activation markers,
including
15 PD-L1, on the DC (see Figures 1A and 1B). Activation marker expression,
including
PD-L1, was not increased above baseline in co-cultures with the unconjugated
HER2
mAb. These data demonstrate that agonism of TLR8 by the HER2-TLR8 composition
result in the activation of DCs, including the upregulation of PD-Li. PD-Li is
the
ligand for the inhibitory receptor PD-1, a receptor expressed on T cells and
other
20 immune cells.
EXAMPLE 2-
HER2-TLR8 INDUCES THE PRODUCTION OF IFN-y A POTENT INDUCER OF PD-Li
UPREGULATION
[0563] Numerous studies have shown the upregulation of PD-L1, a T cell
25 checkpoint molecule, by on tumor cells in the tumor
microenvironment. (Abiko,
283
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Br J Cancer. 2015 Apr 28;112(9):1501-9; Garcia-Diaz A, Cell Rep. 2017 May
9;19(6):1189-1201; Mimura, Cancer Sci. 2018 Jan;109(1):43-53). To determine
whether a HER2-TLR8 conjugate can induce production of IFN-y by human PBMCs in

the presence of HER2 expressing tumor cells, the following experiment was
conducted.
5 Human whole blood was obtained from Bloodworks Northwest (Seattle, WA)
and
collected in 10mL EDTA tubes. Human PBMCs were then isolated from the whole
blood by Ficoll gradient centrifugation and resuspended in assay media (RPMI-
1640
Medium supplemented with 10% Fetal Bovine Serum, 1mM Sodium Pyruvate, lx
GlutaMAX-1, 1X Non-Essential Amino Acids, 10m/vI HEPES and 0.5%
10 Penicillin/Streptomycin; all from Gibco). Isolated PBMCs were
resuspended in assay
media and plated in 96-well flat bottom microtiter plates (125,000/well). HER2-

expressing tumor cells were removed from tissue culture flasks with HyQTASE
(Hyclone), washed twice, resuspended in assay media and were added to the
microtiter
plates (25,000/well) along with titrating concentrations of HER2-TLR8
conjugate.
15 After overnight culture, supernatants were harvested, and [EN-y levels
were determined
by a Meso Scale Discovery) analyte detection kit.
105641 As shown in Figure 2, HER2-TLR8 conjugate stimulated IFN-y
production from huPBMCs in the presence of HER2 3+ (SK-BR-3) and HER2 2+
(MDA-453) expressing cells. In contrast, there was no IF N-7 production from
20 huPBMCs in the presence of tumor cells lacking the expression of HER2
(MDA-468).
These data indicate that the combination of HER2-TLR8 with a T cell checkpoint

inhibitor, such as an anti-PD-1 small molecule or antibody, should provide
enhanced
efficacy.
EXAMPLE 3.
25 RATIONALE FOR HER2-TLR7 CONJUGATE AS A MOUSE SURROGATE FOR HER2-TLR8
105651 Rodents do not express a functional homolog of TLR8 and attempts by
multiple groups to generate relevant human TLR8 transgenic mice have been
unsuccessful (Wang, J Biol Chem. 2006 Dec 8;281(49):37427-34; Guiducci, J Exp
Med. 2013 Dec 16;210(13):2903-19). Like TLR8 in human, and in contrast to TLR7
in
30 human, TLR7 in mouse is expressed in myeloid cells. Using RNA expression
data
284
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
from the publicly available database Haemopedia, Figure 3 demonstrates the
similarity
in expression between TLR7 in mouse and TLR8 in human in dendritic cells and
macrophages. Further, and as anticipated by similarity in expression and
structure
between mouse TLR7 and human TLR8, stimulation of TLR7 in murine myeloid cells
5 results in activation that mirrors that observed with TLR8 stimulation in
human myeloid
cells (Clarke, J. Cytokine Interferon Res. 2009, Feb;29(2):113-26; Gordon, J.
Immunol.
2005 Feb 1;174(3):1259-68). A HER2-TLR7 mouse surrogate consisting of a TLR7
agonist conjugated to an anti-human HER2 antibody of the mouse IgG2a isotype
(HER2-TLR7) was generated for use in in vivo pharmacology studies. In vitro
studies
10 with mouse myeloid cells derived from bone marrow demonstrate that the
HER2-TLR7
mouse surrogate has an EC50 of 0.5nM, equivalent to the potency of HER2-TLR8
conjugates on human myeloid cells (data not shown). These studies and findings

collectively support 1-IER2-TLR7 conjugate as a surrogate molecule for HER2-
TLR8 in
mouse. As such, HER2-TLR7 was evaluated for activity in syngeneic mouse
models.
15 EXAMPLE 4-
TREATMENT WITH HER2-TLR7 SURROGATE DISPLAYS DOSE-DEPENDENT SINGLE
AGENT EFFICACY MI A XENOGRAFT MOUSE MODEL
105661 The single agent efficacy in mice bearing HER2-expressing tumors with
the 1-IER2-TLR7 agonist conjugate (i.e., surrogate for human HER2-TLR8
conjugate)
20 was examined. Briefly, SC1D mice (Taconic Biosciences, Albany, NY) were
inoculated subcutaneously with about 1 x 106 NCI-N87 cells (HER2+ tumorigenic
human epithelial cells) to generate the NCI-N87 xenograft mouse model_ Once
palpable, tumor dimensions were measured by calipers and volume was calculated

using the equation: Volume = xWxHx 0.5). When tumors reached approximately
25 90 mm3 (Day 0), mice were administered 5 mg/kg, 2 mg/kg, or 1 mg/kg HER2-
TLR7 or
unconjugated HER2-IgG2a mAb every 7 days for 4 doses. The HER2-TLR7 comprises
anti-HER2 antibody variable domains comprising the heavy chain CDRs of SEQ ID
NOs: 1-3 and light chain CDRs of SEQ ID NOs: 4-6, conjugated to a TLR7 agonist-

linker having the structure:
285
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
FI2N yo
HN
0 0
11:LA\ INI
0 r0Et
ris
0 0 411
N
11
0
* / Ni-I2
An isotype control mAb (BioXcell, West Lebanon, NH), matched to the
unconjugated
HER2-IgG2a mAb, was dosed at 5 mg/kg on the same schedule. Tumor volumes were
recorded 3 times per week.
5 105671 Treatment with the HER2-TLR7 surrogate resulted in
significant
inhibition of tumor growth at all doses tested (see Figures 4E, 4F, and 4G) as
compared
to the isotype antibody or unconjugated HER2-IgG2a antibody controls (see
Figures
4A, 4B, 4C, and 4D). Like unconjugated HER2-IgG2a antibody treated group, the
mice
treated with IgG2a-TLR7 showed no delay in tumor growth (see Figures 4H, 4J,
and
10 4K). These results demonstrate that the HER2-TLR7 surrogate is
efficacious across a
range of dose levels with robust single agent efficacy at dose levels as low
as 1-2 mg/kg
in viva
EXAMPLES:
IN VIVO TREATMENT WITH A HER2-TLR7 SURROGATE RESULTS IN PD-Li
15 UPREGULATION ON THE SURFACE OF TUMOR CELLS
105681 The effect of treatment of mice bearing HER2-expressing tumors with
the IIER2-TLR7 conjugate surrogate, on PD-L1 expression on the surface of
tumor
cells was examined. Briefly, female BALB/c mice (Jackson Laboratory) were
inoculated subcutaneously with 5x105 human HER2-expressing CT26 tumor cells.
20 Once palpable, tumor dimensions were measured by calipers and volume was
calculated using the following equation: Volume = (L xWxHx 0.5). When tumors
reached an average of approximately 200 mm3 (Day 0), mice were administered
mg/kg HER2-TLR7 or HER2 mAb (matched, unconjugated antibody control)
subcutaneously on days 0, 2, and it At 48 hours post-doses one and three,
tumors were
286
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
harvested and dissociated using the Miltenyi mouse tumor digest kit. Single
cell
suspensions were assayed for PD-Li expression using flow cytometry.
105691 As shown in Figures 5A and 5B, tumor cells (identified as CD45
negative, forward scatter high) showed a significant increase in PD-Li
expression at
5 both time points evaluated. These findings provide evidence that
combination with
immune checkpoint inhibition could enhance efficacy.
EXAMPLE 6,
COMBINATION OF HER2-TLR7 SURROGATE AND IMMUNE CHECKPOINT INHIBITOR
LEADS TO INCREASED TH1 CYTOKINES AND CHEMOKINES.
10 105701 The effect of treating tumor bearing mice with a
combination of HER2-
TLR7 conjugate surrogate and anti PD-1 on IFN-y and IF-10 expression was
determined. Briefly, female BALB/c mice (Jackson Laboratory) were inoculated
subcutaneously in the mammary fat with lx 105 human HER2-expressing EMT6 tumor

cells. Once palpable, tumor dimensions were measured by calipers and volume
was
15 calculated using the equation: Volume = (L xWxHx 0.5). When tumors
reached
approximately 97mm3 (Day 0), mice were given one dose of 10 mg/kg HER2-TLR7,
HER2 mAb (matched, unconjugated antibody control), rat anti-PD1 (clone RMP1-
14)
or isotype controls, alone or in combination. Two or seven days later, tumors
were
excised, weighed placed in 500mL RPMI (Gibco) and mechanically dissociated on
ice.
20 The resulting supernatants were analyzed by Luminex (Millipore) for
intratumoral
cytokines and chemokines. Data is expressed as picogram of analyte per gram of

starting tissue.
105711 Figure 6A shows that IP10 production was significantly increased in the

combination treated group at Day 2 compared to HER2-TLR7 or anti PD-1 treated
25 groups. Similarly, IFN-y production was augmented at Day 2 in tumors of
mice treated
with combination of HER2-TLR7 with anti-PD-1 compared to HER2-TLR7 treated
mice (Figure 6B). This amplification of IFN-y was sustained at Day 7 after
treatment
These data indicate that the combination of an immune checkpoint inhibitor
with
HER2-TLR8 is expected to lead to enhanced chemolcine and cytolcine production
by
30 immune cells.
287
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
EXAMPLE 7.
ANTI-TUMOR RESPONSE OF HER2-TLR7 SURROGATE IS ENHANCED WHEN COMBINED
WITH ANTI-PD1
105721 The anti-tumor efficacy induced by single agent HER2-TLR7 conjugate
5 mouse surrogate (HER2-TLR7) was examined in combination with anti-PD1
therapy.
Female BALB/c mice (Jackson Laboratory) were inoculated subcutaneously with lx
105
human HER2-expressing EMT6 tumor cells. Once palpable, tumor dimensions were
measured by calipers and volume was calculated using the following equation:
Volume = (L x Wx Hx 0.5). When tumors reached approximately 90 mm3 (Day 0),
10 mice were given 10 mg/kg HER2-TLR7, HER2 mAb (naked antibody control),
rat anti-
PD1 (clone RMP1-14) or isotype controls, alone or in combination, every 7 days
for a
total of 3 doses. Tumor volume was recorded 3 times per week and mice were
euthanized when tumors reached 1,500 MM3.
105731 Figure 7D spider plots show that this model is refractory to single
agent
15 anti-PD1 therapy with no survival benefit or growth inhibition over the
isotype control
group (see Figure 7A). In contrast, treatment with HER2-TLR7 surrogate
inhibited
tumor growth with 60% of mice surviving to day 42 (see Figure 7C). Moreover,
the
combination of HER2-TLR7 with anti-PD1 therapy greatly enhanced the effect of
either agent alone with 90% of mice surviving to day 42 (see Figure 7F).
20 EXAMPLE 8.
HER2-TLR8 AND A TRASTUZUMAB-LTICE MONOCLONAL ANTIBODY, HUMANIZED 4D5,
BIND TO DIFFERENT HER2 EFITOFES
105741 The trastuzumab-like monoclonal antibody, humanized 405 mAb
(hz4D5), was produced using the ExpiCHO expression system as described in the
25 manufacturer's protocol (Thermo Fisher Scientific). Briefly,
6x106ExpiCHO-S cells
per ml were cultured in ExpiCHO Expression Medium followed by addition of
ExpiFectamine CHO/plasmid DNA complexes Ogg DNA/ml) directly to cells.
ExpiCHO Feed and Enhancer were added the day after transfection to support
long-
term, high-density transient transfections and to enhance protein production_
The cell
30 culture supemantants were harvested on Day 7 following transfection.
Hz4D5 from the
288
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
ExpiCHO supernatants were purified to homogeneity over HiScreen MabSelect SuRe

protein A column on GE AKTA Pure ml system. It was confirmed for purity on
Size
Exclusion Chromatography using Agilent 1260 Infinity machine.
105751 Analysis of simultaneous versus competitive binding of HER2-TLR8
5 and hz4D5 to human HER2 extra-cellular domain (ECD) was performed using
an Octet
Red 96TM instrument (ForteBio). The experiments were performed using the
following steps: (1) Regeneration and neutralization of penta-his sensor in
10mM
glycine-HC1 (pH 1.5) and PBS/0.1%BSA/0.02%Tween20 (pH 7.4), respectively in
triplicate; (2) baseline acquisition (30s); (3) immobilization of monomeric
human
10 HER2 ECD with 10x histidine tag to penta-his sensor at a concentration
of 5gg/mL in
PBS/0.1%BSA/0.02%Tween20 (180s); (4) second baseline acquisition (60s);
(5) association of test article #1 (HER2-TLR8 or hz4D5) at 25 gg/mL until
saturation
(300s); (6) third baseline acquisition (15s); (7) association of test article
#2 (HER2-
TLR8 or hz4D5) at 25 gg/mL (300s); (8) dissociation in baseline buffer (60s).
The data
15 were analyzed using Octet Data Analysis Software HT 9.0TM (ForteBio).
105761 The interactions of HER2-TLItit and hz4D5 with monomeric human
HER2 ECD were evaluated using Octet Red 96TM in 2 orientations. Figure 8A
shows
Orientation #1 whereby HER2-TLR8 binds to HER2 ECD until saturation and then
either HER2-TLR8 or hz4D5 is added. Figure 8B shows Orientation #2 whereby
20 hz4D5 binds to HER2 ECD until saturation and then either hz4D5 or HER2-
TLR8 is
added. Affinity of either test article to immobilized HER2 ECD is subject to
2:1
binding avidity and no dissociation of test article #1 is observed in the
third baseline
step.
105771 Figure 8A shows that additional mass is observed when hz4D5 is added
25 to a HER2-TLR8 saturated surface, indicating additional binding sites
are available,
whereas no additional mass is observed when HER2-TLR8 is added to an already
HER2-TLR8 saturated surface, indicating all HER2-TLR8 binding sites are taken
up.
Figure 8B shows that additional mass is observed when HER2-TLR8 is added to a
hz4D5 saturated surface, indicating additional binding sites are available,
whereas no
30 additional mass is observed when hz4D5 is added to an already hz4D5
saturated
surface, indicating all hz4D5 binding sites are taken up. Together, these two
289
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
orientations confirm that HER2-TLR8 and hz4D5 do not compete for binding space
on
HER2 ECD when either site is saturated with the other test article.
Furthermore, this
data confirmed that HER2-TLR8 and hz4D5 are able to bind simultaneously to
human
HER2 ECD.
5 EXAMPLE 9.
1-MR2-TLR8 AGONIST CONJUGATE DOES NOT IMPEDE FUNCTION OF TRASTUZUMAB-
LIKE ANTIBODY, HZ4D5, IN VITRO
[0578] The ability of trastuzumab to inhibit HER2-dependent tumor cell growth
in vitro has been extensively described in the literature (see, e.g.,
Yamashita-Kashima et
10 al., Clin. Cancer Res. 17:5060, 2011; Ko et al., Mot Oncol. 9:398, 2015:
Nami et al ,
Cancers /0:342, 2018). SBT6050 and hz4D5 (trastuzumab binding domain)
recognize
distinct, non-overlapping epitopes on HER2. Briefly, BT-474 tumor cells were
cultured
in the presence of HER2-TLR8 conjugate, negative control mAb conjugated to
TLR8
agonist (IgG1-TLR8), or isotype control mAb alone or in combination with 1:1
ratio of
15 trastuzumab-like monoclonal antibody hz4D5. Cells were incubated for 3
days and
tumor cell viability was measured using Cell Titer-Glo Luminescent Cell
Viability Kit
(Promega). Results are expressed as a percentage decrease in viability
compared to BT-
474 tumor cells grown in media alone.
[0579] Antibody hz4D5 showed significant anti-proliferative effects on the
20 growth of BT-474 tumor cells, whereas the combination of hz4D5 and HER2-
TLR8
agonist conjugate increased this effect (Figure 9), HER2-TLR8 agonist
conjugate alone
had no effect on the growth of BT474 tumor cells (Figure 9). In short, HER2-
TLR8
agonist conjugate does not affect the ability of hz4D5 to inhibit the growth
of HER2
positive tumor cells in vitro. These results indicate that trastuzumab
function will likely
25 be maintained, and even enhanced, when used in combination with HER2-
TLR8
agonist conjugate in vivo.
290
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
EXAMPLE 10.
ComBrtoNG HER2-TLR8 CONJUGATE WITH A TRASTUZUMAB-LIKE MONOCLONAL
ANTIBODY AUGMENTS RELEASE OF PBMC TNF-A AND IFN-r PRODUCTION IN THE
PRESENCE OF HER2 EXPRESSING TUMOR CELLS
5 105801 Production of TNF-a and IFN-y from PBMC co-cultured with
HER2
positive tumor cell lines contacted with a HE.R2-TLR8 conjugate and hz4D5, a
trastuzumab-like monoclonal antibody, was examined. The HER2-TLR8 conjugate
does not cross-block binding or the function of the trastuzumab-like
monoclonal
antibody (see Examples 8 and 9). Peripheral blood mononuclear cells (PBMC)
were
10 isolated from normal human donor peripheral blood using SepMaten1-50
PBMC
Isolation Tubes (STEMCELL Technologies) according to manufacturers
instructions.
Isolated PBMC were cultured with the HER2-expressing tumor cell lines NCI-N87,

BT-474, HCC-1954 (ATCC) or the HER2-negative tumor cell line MDA-MB-468
(ATCC) at a 5:1 ratio in the presence of titrated concentrations of a HER2-
TLR8
15 conjugate, the matched unconjugated HER2 mAb control, a trastuzumab-like
humanized monoclonal antibody (hz4D5) or an isotype control monoclonal
antibody.
After 24 hours the cell-free supernatants were collected and stored at -80 C
prior to
analysis. TNF-a levels in the cell-free supernatants were quantified using the
TNF-a
(human) AlphaLISA Detection Kit (Perkin Elmer) according to manufacturer's
20 instructions. IFN-y levels in the cell-free supernatants were quantified
using the U-
PLEX Biomarker Group 1 (Human) Multiplex Assay (Mesa Scale Diagnostics)
according to manufacturer's instructions.
105811 Representative data is shown in Figure 10A for TNF-a and Figure 10B
for IFN-y. Figure 10A shows HER2-TLR8 induced TNF-a production is augmented in
25 PBMC cultured with the HER2-expressing tumor cell lines NCI-N87, BT-474
or HCC-
1954 when combined with the trastuzumab-like monoclonal antibody hz4D5
compared
to the combination of HER2-TLR8 and an isotype control. No TNT-a production
was
observed with the combination of hz4D5 and the matched unconjugated HER mAb
indicating that TLR8 agonism is required for TNF-a release. TNF-a production
was
30 observed with the HER2-negative tumor cell line MDA-M13-468 only in
cultures
containing HER2-TLR8 at the highest concentrations tested indicating that the
activity
291
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
is also dependent upon HER2. As observed for TNF-a, Figure 10B shows IFN-y
production is augmented in PBMC cultured with the HER2-expressing tumor cell
line
NCI-N87 when HER2-TLR8 is combined with the trastuzumab-like monoclonal
antibody hz4D5 compared to the combination of HER2-TLR8 and an isotype control
monoclonal antibody.
105821 Taken together these data demonstrate augmented TNF-a and LFN-y
production by the combination of HER2-TLR8 and the trastuzumab-like monoclonal

antibody hz4D5 over the effects observed with HER2-TLR8 alone. These effects
are
mediated by the TLR8 agonist conjugate rather than by HER2-TLR8 binding to a
different epitope than 4D5 and suggest combining HER2-TLR8 with a clinical
HER2-
targeting antibody, such as trastuzumab, may increase therapeutic efficacy in
certain
tumor settings.
EXAMPLE 11:
COMBINATION OF HER2-TLR7 SURROGATE WITH A TRASTUZUMAB-LIKE MONOCLONAL
ANTIBODY, HUMANIZED 4D5 (HZ4D5), IS EFFICACIOUS IN A XENOGRAFT MOUSE MODEL
105831 This example shows that the robust single agent efficacy, including an
80% cure rate, achieved with the HER2-TLR7 mouse surrogate conjugate at 10
mg/kg
is not impeded by combination with the trastuzumab-like monoclonal antibody,
hz4D5.
The trastuzumab-like monoclonal antibody, humanized 4D5 mAb (hz4D5), does not
cross-block HER2-TLR7 binding, suggestive of binding to different regions of
HER2.
SCID mice (Charles River) were inoculated subcutaneously with 1 x 106 NCI-N87
(HER2+) tumor cells. Once palpable, tumor dimensions were measured by calipers
and
volume was calculated using the equation: Volume = (L xWxHx 0.5). When tumors
reached approximately 90 mm3 (Day 0), mice were given 10 mg/kg 1-IER2-TLR7,
HER2 mAb (matched, unconjugated antibody control), or isotype controls
(BioXcell),
alone or in combination with 10 mg/kg hz4D5, every 7 days for a total of 4
doses.
Tumor volume was recorded 3 times per week.
105841 As shown in Figures 11A-11F, mice treated with isotype controls, the
unconjugated HER2 mAb, or hz4D5 showed no delay in tumor growth. Combination
of HER2 mAb with hz4D5 initially showed tumor growth inhibition, but the
tumors
292
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
eventually grew out. However, HER2-TLR7 as a single agent resulted in profound

efficacy with 8/10 mice completely clearing their tumors by Day 66 at this
dose level.
Importantly, the combination of HER2-TLR7 with hz4D5 had no adverse effect on
the
single agent efficacy of HER2-TLR7 at this dose level.
5 EXAMPLE 12.
COMBINATION OF A TRASTUZUMAI3-LIKE MONOCLONAL ANTIBODY, HUMANIZED 4D5
mAs (Hz4D5), AND LOW DOSE HER2-TLR7 SURROGATE CONJUGATE DEMONSTRATES
ENHANCED EFFICACY OVER SINGLE AGENTS IN A XENOGRAFT MOUSE MODEL
[0585] To examine whether the efficacy of HER2-TLR7 mouse surrogate
10 conjugate combined with the trastuzumab-like monoclonal antibody, hz4D5,
was
increased as compared to either agent administered alone, a low dose of the
HER2-
TLR7 mouse surrogate conjugate was used. Briefly, SCID mice (Charles River) (n
=
10) were inoculated subcutaneously with about lx106NCI-N87 (HER2+) tumor
cells.
Once palpable, tumor dimensions were measured by calipers and volume and
calculated
15 using the equation: Volume = (L xWxHx 0.5). When tumors reached
approximately
100 mm3 (Day 0), mice were dosed with HER2-TLR7 mouse surrogate conjugate (1
mg/kg), matched unconjugated HER2-IgG2a mAb (1 mg/kg), or isotype mAb control
matched to unconjugated HER2-IgG2a mAb (5 mg/kg) alone or in combination with
the trastuzumab-like monoclonal antibody hz4D5 (5 mg/kg) every 7 days for a
total of 3
20 doses. Tumor volume was recorded 3 times per week.
105861 HER2-TLR7 administered at the low dose in this example allowed for
evaluation of efficacy in combination with the trastuzumab-like monoclonal
antibody,
hz4D5, as compared to the single agents alone. Mice treated with unconjugated
HER2-
IgGa mAb, hz4D5, or HER2-TLR7 surrogate showed some delay in tumor growth
25 compared to the isotype control group, but the tumors eventually grew.
However, mice
treated with the HER2-TLR7 surrogate in combination with hz4D5 showed a
significant reduction in tumor growth (Figure 12F) compared to all other
treated groups
(Figures 12A-E). These data show that the HER2-TLR7 surrogate in combination
with
trastuzumab-like monoclonal antibody, hz4D5 results in entranced efficacy over
the
30 single agents alone. In addition, these data indicate that clinical
evaluation of HER2-
293
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
TLR8 agonist conjugates of this disclosure as a single agent and in
combination with
trastuzumab in relevant HER2-expressing tumor types is warranted.
[0587] While aspects of the present disclosure have been shown and described
herein, it will be apparent to those skilled in the art that such aspects are
provided by
5 way of example only. Numerous variations, changes, and substitutions will
now occur
to those skilled in the art without departing from the disclosure. It should
be understood
that various alternatives to the aspects of the disclosure described herein
may be
employed in practicing the disclosure. It is intended that the following
claims define
the scope of the disclosure and that methods and structures within the scope
of these
10 claims and their equivalents be covered thereby.
294
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
Table of Anti-HER2 Antibody Sequences
SEQ ID Description Sequence
NO
1 Anti-HER2 GFTFTDYTMD
antibody heavy
chain (HC) CDR1
2 Anti-HER2 DVNPNSGGSIYNQRFKG
antibody HC CDR2
3 Anti-HER2 NLGPSFYFDY
antibody HC CDR3
4 Anti-HER2 KASQDVSIGVA
antibody light chain
(LC) CDR1
Anti4lER2 SASYRYT
antibody LC CDR2
6 Anti-HER2 QQYYIYPYT
antibody LC CDR3
7 1&nti-HER2 EVQLVESGGG
LVQPGGSLRL SCAASGFTFT DYTMDWVRQA
antibody heavy PGKGLEWVAD
VNPNSGGSIY NQRFEGRFTL SVDRSKNTLY
chain variable LQMNSLRAED
TAVYYCARNL GPSFYFDYWG QGTLVTVSS
region (VH)
8 Anti-HER2 DIQMTQSPSS
LSASVGDRVT ITCKASQDVS IGVAWYQQKP
antibody light chain GKAPKLLIYS ASYRYTGVPS RFSGSGSGTD FTLTISSLQP
variable region EDFATYYCQQ
YYIYPYTFGQ GTKVEIK
(VL)
9 Anti-HER2 EVQLVESGGG
LVQPGGSLRL SCAASGFTFT DYTMDWVRQA
antibody heavy PGKGLEWVAD
VNPNSGGSIY NQRFEGRFTL SVDRSKNTLY
chain LQMNSLRAED
TAVYYCARNL GPSFYFDYWG QGTLVTVSSA
STKGPSVFPL APSSKSTSGG TAALGCLVKD YFPEPVTVSW
NSGALTSGVE TFPAVLQSSG LYSLSSVVTV PSSSLGTQTY
ICNVNHKPSN TKVDKKVEPK SCDKTHTCPP CPAPELLGGP
SVFLFPPKPK DTLMISRTPE VTCVVVDVSH EDPEVKFNWY
VDGVEVHNAK TKPREEQYNS TYRVVSVITV LHQDWLNGKE
YKCKVSNKAL PAPIEKTISK AKGQPREPQV YTLPPSREEM
TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVI
DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ
KSLSLSPG
Anti-HER2 DIQMTQSPSS LSASVGDRVT ITCKASQUVS IGVAWYQQKP
antibody light chain GKAPKLLIYS ASYRYTGVPS RFSGSGSGTD FTLTISSLQP
EDFATYYCQQ YYIYPYTFGQ GTKVEIKRTV AAPSVFIFPP
SDEQLKSGTA SVVCLLNNFY PREAKVQWKV DNALQSGNSQ
ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG
LSSPVTKSFN RGEC
The various embodiments described above can be combined to provide further
embodiments. All of the U.S. patents, U.S. patent application publications,
U.S. patent
5 applications, foreign patents, foreign patent
applications and non-patent publications
295
CA 03151322 2022-3-15

WO 2021/067644
PCT/US2020/053870
referred to in this specification and/or listed in the Application Data Sheet,
including
U.S. Patent Application No. 62/908,881, filed October 1, 2019, U.S. Patent
Application
No. 62/935,789, filed November 15, 2019, and U.S. Patent Application No.
63/018,615,
filed May 1, 2020, are incorporated herein by reference, in their entirety.
Aspects of the
5 embodiments can be modified, if necessary to employ concepts of the
various patents,
applications and publications to provide yet further embodiments.
These and other changes can be made to the embodiments in light of the above-
detailed description. In general, in the following claims, the terms used
should not be
construed to limit the claims to the specific embodiments disclosed in the
specification
10 and the claims, but should be construed to include all possible
embodiments along with
the full scope of equivalents to which such claims are entitled. Accordingly,
the claims
are not limited by the disclosure.
296
CA 03151322 2022-3-15

Representative Drawing
A single figure which represents the drawing illustrating the invention.
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 2020-10-01
(87) PCT Publication Date 2021-04-08
(85) National Entry 2022-03-15

Abandonment History

Abandonment Date Reason Reinstatement Date
2023-04-03 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Maintenance Fee


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2022-10-03 $50.00
Next Payment if standard fee 2022-10-03 $125.00

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 $407.18 2022-03-15
Registration of a document - section 124 $100.00 2022-03-15
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
SILVERBACK THERAPEUTICS, 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 2022-03-15 1 15
National Entry Request 2022-03-15 2 65
Assignment 2022-03-15 5 95
Patent Cooperation Treaty (PCT) 2022-03-15 1 56
Priority Request - PCT 2022-03-15 215 8,180
International Search Report 2022-03-15 6 169
Priority Request - PCT 2022-03-15 221 8,150
Priority Request - PCT 2022-03-15 220 8,329
Description 2022-03-15 296 9,401
Declaration 2022-03-15 3 55
Claims 2022-03-15 68 1,747
Drawings 2022-03-15 24 338
Patent Cooperation Treaty (PCT) 2022-03-15 2 61
Correspondence 2022-03-15 2 46
National Entry Request 2022-03-15 10 203
Abstract 2022-03-15 1 10
Representative Drawing 2022-05-09 1 3
Cover Page 2022-05-09 1 35

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.