Language selection

Search

Patent 3173356 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 3173356
(54) English Title: A TRIPLE PHARMACEUTICAL COMBINATION COMPRISING DABRAFENIB, AN ERK INHIBITOR AND A RAF INHIBITOR OR A PD-1 INHIBITOR.
(54) French Title: COMBINAISON PHARMACEUTIQUE TRIPLE COMPRENANT DABRAFENIB, UN INHIBITEUR D'ERK ET UN INHIBITEUR DE RAF OU UN INHIBITEUR DE PD-1.
Status: Compliant
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 31/4965 (2006.01)
  • A61K 31/506 (2006.01)
  • A61K 31/5377 (2006.01)
  • A61P 35/00 (2006.01)
(72) Inventors :
  • COOKE, VESSELINA (United States of America)
(73) Owners :
  • NOVARTIS AG (Switzerland)
(71) Applicants :
  • NOVARTIS AG (Switzerland)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2021-02-26
(87) Open to Public Inspection: 2021-09-02
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/IB2021/051641
(87) International Publication Number: WO2021/171260
(85) National Entry: 2022-08-26

(30) Application Priority Data:
Application No. Country/Territory Date
62/983,020 United States of America 2020-02-28

Abstracts

English Abstract

The present invention relates to a pharmaceutical combination comprising dabrafenib, an Erk-inhibitor and a RAF inhibitor; pharmaceutical compositions comprising the same; and methods of using such combinations and compositions in the treatment or prevention of conditions in which MAPK pathway inhibition is beneficial, for example, in the treatment of cancers.


French Abstract

La présente invention concerne une combinaison pharmaceutique comprenant de la dabrafenib, un inhibiteur d'Erk Et un inhibiteur de RAF; des compositions pharmaceutiques les comprenant ; et des procédés d'utilisation de telles combinaisons et compositions dans le traitement ou la prévention d'états dans lesquels l'inhibition de la voie MAPK est bénéfique, par exemple, dans le traitement de cancers.

Claims

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


What is claimed is:
1. A pharmaceutical combination comprising: N-(3-(5-(2-aminopyrimidin-4-y1)-2-
(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof; 4-(3-amino-6-((1S,3S,45)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof;
and N-(3-(2-
(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide (compound C), or a pharmaceutically acceptable salt thereof
2. The combination of claim 1, wherein N-(3-(5-(2-aminopyrimidin-4-y1)-2-(tert-

butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof, 4-(3-amino-6-((1S,3S,45)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof,
and N-(3-(2-
(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide (compound C), or a pharmaceutically acceptable salt thereof,
are
administered separately, simultaneously or sequentially, in any order.
3. The pharmaceutical combination according to claim 1 or 2, which is for oral

administration.
4. The pharmaceutical combination according to any one of claims 1 to 3,
wherein N-(3-(5-
(2-aminopyrimidin-4-y1)-2-(tert-butypthiazol-4-y1)-2-fluoropheny1)-2,6-
difluorobenzenesulfonamide (dabrafenib) is in an oral dosage form.
5. The pharmaceutical combination according to any one of claims 1 to 4,
wherein 4-(3-
amino-6-((1S,3S,45)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-
bromo-5-
fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide (compound A) is in an
oral dosage
form.

6. The pharmaceutical combination according to any one of claims 1 to 4,
wherein N-(3-(2-
(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide (compound C) is in an oral dosage form.
7. A pharmaceutical composition or a commercial package comprising the
pharmaceutical
combination according to any one of the preceding claims and at least one
pharmaceutically
acceptable carrier.
8. A pharmaceutical combination according to any one of claims 1 to 6 or the
pharmaceutical
composition or the commercial package according to claim 7 for use in the
treatment of
cancer.
9. The pharmaceutical combination or the pharmaceutical composition or the
commercial
package for use according to claim 8, wherein the cancer is selected from
breast cancer,
cholangiocarcinoma, colorectal cancer (CRC), melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer.
10. The pharmaceutical combination or the pharmacutical composition or the
commercial
package for use according to claim 8, wherein the cancer is advanced or
metastatic colorectal
cancer.
11. The pharmaceutical combination of claim 10 wherein the cancer is BRAF gain
of
function CRC or BRAF V600E, V600D or V600K CRC.
12. Use of the pharmaceutical combination according to any one of claims 1 to
6 or the
pharmaceutical composition or commercial package according to claim 7 for the
manufacture
of a medicament for the treatment of cancer.
13. The use of the pharmaceutical combination or the pharmaceutical
composition according
to claim 12 wherein the cancer is selected from breast cancer,
cholangiocarcinoma, colorectal
cancer, melanoma, non-small cell lung cancer, ovarian cancer and thyroid
cancer, optionally
51

wherein the cancer is advanced or metastatic colorectal cancer, optionally
wherein the cancer
is BRAF gain of function CRC or BRAF V600E, V600D or V600K CRC.
14. A method of treating a cancer selected from breast cancer,
cholangiocarcinoma,
colorectal cancer, melanoma, non-small cell lung cancer, ovarian cancer and
thyroid cancer
comprising administrating to a patient in need thereof a pharmaceutical
combination or
commercial package according to any one of claims 1 to 6 or the pharmaceutical
composition
according to claim 7.
15. The method of claim 14 wherein the colorectal cancer is advanced or
metastatic
colorectal cancer.
16. The method of claim 15 wherein the colorectal cancer is BRAF gain of
function CRC or
BRAF V600E, V600D or V600K CRC.
17. The method of claim 14, wherein N-(3-(5-(2-aminopyrimidin-4-yl)-2-(tert-
butyl)thiazol-
4-yl)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide (dabrafenib) is
administered orally at
a dose of about from about 1 to about 150 mg per day.
18. The method of claim 14, wherein 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-yl)-N-((S)-1-(3-bromo-5-fluorophenyl)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A) is administered orally at a dose of from about
50 to about
200 mg per day.
19. The method of claim 14, wherein N-(3-(2-(2-hydroxyethoxy)-6-
morpholinopyridin-4-yl)-
4-methylphenyl)-2-(trifluoromethyl)-isonicotinamide (compound C) is
adminstered orally at a
dose of from about 100 mg per day, or 200 mg per day, or 300 mg per day to
about 70 mg per
day.
52

Description

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


CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
A TRIPLE PHARMACEUTICAL COMBINATION COMPRISING DABRAFENIB,
AN ERK INHIBITOR AND A RAF INHIBITOR OR A PD-1 INHIBITOR.
FIELD OF THE INVENTION
[0001] The present invention relates to a pharmaceutical combination
comprising:
[0002] (i). dabrafenib, or a pharmaceutically acceptable salt thereof,
an Erk inhibitor
(ERKi) such as 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-
y1)-N-
((S)-1-(3-bromo-5-fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide
("Compound A"
or "compound A"), or a pharmaceutically acceptable salt thereof, and a RAF
inhibitor such as
N-(3-(2-(2-hydroxye thoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide ("Compound C") or a pharmaceutically acceptable salt thereof;
or
[0003] (ii). dabrafenib, or a pharmaceutically acceptable salt
thereof, an Erk inhibitor
(ERKi) such as 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-
y1)-N-
((S)-1-(3-bromo-5-fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide
("Compound A"
or "compound A"), or a pharmaceutically acceptable salt thereof, and a PD-1
inhibitor such as
Spartalizumab; and pharmaceutical compositions comprising the same; commercial
packages
comprising the same; and methods of using such combinations and compositions
in the
treatment or prevention of conditions in which MAPK pathway inhibition is
beneficial, for
example, in the treatment of cancers. The present invention also povides such
combinations
.. for use in the treatments of such conditions or cancers, including
colorectal cancer (CRC)
such as BRAF gain of function colorectal cancer.
BACKGROUND OF THE INVENTION
[0004] The MAPK pathway is a key signaling cascade that drives cell
proliferation,
differentiation, and survival. Dysregulation of this pathway underlies many
instances of
tumorigenesis. Aberrant signaling or inappropriate activation of the MAPK
pathway has been
shown in multiple tumor types and can occur through several distinct
mechanisms, including
activating mutations in RAS and BRAF. The MAPK pathway is frequently mutated
in human
1

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
cancer with KR/IS and BR/IF mutations being among the most frequent
(approximately 30%).
RAS mutations, particularly gain of function mutations, have been detected in
9-30% of all
cancers, with KR/IS mutations having the highest prevalence (86%).
[0005] The extracellular signal-regulated kinases (ERKs) are one class
of signaling
kinases that are involved in conveying extracellular signals into cells and
subcellular
organelles. ERK1 and ERK2 are involved in regulating a wide range of
activities and
dysregulation of the ERK1/2 cascade is known to cause a variety of pathologies
including
neurodegenerative diseases, developmental diseases, diabetes and cancer. The
role of
ERK1/2 in cancer is of special interest because activating mutations upstream
of ERK1/2 in
its signaling cascade are believed to be responsible for more than half of all
cancers.
Moreover, excessive ERK1/2 activity was also found in cancers where the
upstream
components were not mutated, suggesting that ERK1/2 signaling plays a role in
carcinogenesis even in cancers without mutational activations. The ERK pathway
has also
been shown to control tumor cell migration and invasion, and thus may be
associated with
.. metastasis.
[0006] The Programmed Death 1 (PD-1) protein is an inhibitory member
of the extended
CD28/CTLA-4 family of T cell regulators. Two ligands for PD-1 have been
identified, PD-Li
(B7-H1) and PD-L2 (B7-DC), that have been shown to downregulate T cell
activation upon
binding to PD-1. PD-Li is abundant in a variety of human cancers. PD-1 is
known as an
immunoinhibitory protein that negatively regulates TCR signals. The
interaction between PD-1
and PD-Li can act as an immune checkpoint, which can lead to, for example, a
decrease in tumor
infiltrating lymphocytes, a decrease in T-cell receptor mediated
proliferation, and/or immune
evasion by cancerous cells. Immune suppression can be reversed by inhibiting
the local
interaction of PD-1 with PD-Li or PD-L2; the effect is additive when the
interaction of PD-1 with
PD-L2 is blocked as well.
[0007] Given the importance of immune checkpoint pathways in
regulating an immune
response, the need exists for developing novel combination therapies that
activate the immune
system. The prognosis for patients suffering from certain cancers remains
poor. Resistance to
treatment occurs frequently and not all patients respond to available
treatments. For example,
the median survival for patients suffering from advanced colorectal cancer
with BRAF
mutation is less than 12 months. It is thus important to develop new therapies
for patients
2

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
suffering from cancer to achieve better clinical outcomes. Treatment options
which are better
tolerated and/or provide durable anti-tumor responses are also desired.
SUMMARY OF THE INVENTION
[0008] The triple combinations of the present invention (i) dabrafenib; an
Erk-
inhibitor such as Compound A, and a RAF-inhibitor such as compound C; or (ii)
dabrafenib,
an Erk-inhibitor such as Compound A, and a PD-1 inhibitor such as
Spartalizumab, can be
used as therapies for the treatment of diseases or disorders resulting from
the aberrant activity
of the MAPK pathway including, but not limited to, breast cancer,
cholangiocarcinoma,
salivary gland cancer, colorectal cancer, melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer. Triple combinations of dabrafenib, an Erk-inhibitor
such as
Compound A, and a RAF-inhibitor such as compound C, or dabrafenib, an Erk-
inhibitor such
as Compound A, and a PD-1 inhibitor such as Spartalizumab, are particularly
useful in the
treatment of colorectal cancer (CRC), including advanced or metastatic
colorectal cancer,
which is BRAF gain of function or BRAFV600E/D/K mutants.
[0009] The present invention provides a pharmaceutical combination
comprising:
(a) N-(3 -(5 -(2-aminopyrimidin-4-y1)-2-(tert-butypthiazol-4-y1)-2-
fluoropheny1)-2,6-
difluorobenzenesulfonamide (dabrafenib), or a pharmaceutically acceptable salt
thereof,
having the structure:
411 Fo
F N)L--
F HN
/ N
(b) 4-(3-amino-6-(( 1 S,3 S,4 5)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-y1)-N-
((S)- 1 -(3 -
bromo-5-fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), or
a
pharmaceutically acceptable salt thereof, having the structure:
3

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
F 0
F
NH2
N
I
Br
OH ;and
(c) N-(3-(2-(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-me thylpheny1)-2-
(trifluoromethyl)-isonicotinamide (Compound C), or a pharmaceutically
acceptable salt
thereof, having the structure:
0
N)<FF
N2 H I
OH
=
[0010] The present invention provides a pharmaceutical combination
comprising:
(a) N-(3-(5-(2-aminopyrimidin-4-y1)-2-(tert-butypthiazol-4-y1)-2-fluoropheny1)-
2,6-
difluorobenzenesulfonamide (dabrafenib), or a pharmaceutically acceptable salt
thereof,
having the structure:
Fp
F N
F HN
/ N
1_
1
""-N' -NH2
(b) 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-
1-(3-
bromo-5-fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), or
a
pharmaceutically acceptable salt thereof, having the structure:
4

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
F 0
7
F
NH2
N
I
Br
c.,'F
OH ;and
(c) a PD-1 inhibitor.
[0011] In a further aspect of the invention, the PD-1 inhibitor is
chosen from PDR001
(spartalizumab; Novartis), Nivolumab (Bristol-Myers Squibb), Pembrolizumab
(Merck & Co),
Pidilizumab (CureTech), MEDI0680 (Medimmune), REGN2810 (Regeneron), TSR-042
(Tesaro), PF-
06801591 (Pfizer), BGB-A317 (Beigene), BGB-108 (Beigene), INCSHR1210 (Incyte),
or AMP-224
(Amplimmune).
[0012] In a further aspect of teh invention, the PD-1 inhibitor is
PDR001 (spartalizumab).
[0013] Pharmaceutical combinations of (i) dabrafenib, or a
pharmaceutically
acceptable salt thereof, Compound A, or a pharmaceutically acceptable salt
thereof, and
Compound C, or a pharmaceutically acceptable salt thereof, or (ii) dabrafenib,
or a
pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable salt
thereof, and a PD-1 inhibitor such as Spartalizumab, will also be referred to
herein as a
"combination of the invention".
[0014] There is provided a combination of the invention for use in the
treatment of
cancer, e.g for use in a cancer which is selected from breast cancer,
cholangiocarcinoma,
salivary gland cancer, colorectal cancer, melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer.
[0015] There is provided a pharmaceutical combination of (i)
dabrafenib, or a
pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable salt
thereof, and Compound C, or a pharmaceutically acceptable salt thereof, or
(ii) dabrafenib, or
a pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable
salt thereof, and a PD-1 inhibitor such as spartaluzimab, e.g for use in a
cancer which is
5

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
selected from breast cancer, cholangiocarcinoma, salivary gland cancer,
colorectal cancer,
melanoma, non-small cell lung cancer, ovarian cancer and thyroid cancer.
[0016] There is also provided a combination of (i) dabrafenib, or a
pharmaceutically
acceptable salt thereof, Compound A, or a pharmaceutically acceptable salt
thereof, and
Compound C, or a pharmaceutically acceptable salt thereof, or (ii) dabrafenib,
or a
pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable salt
thereof, and a PD-1 inhibitor such as Spartalizumab for use in the treatment
of colorectal
cancer (which includes advanced or metastsatic colorectal cancer) which is
BRAF gain of
function or BRAFV600E/D/K mutants.
[0017] Also provided herein is a combination of the invention for use in
the treatment
of colorectal cancer (which includes advanced or metastsatic colorectal
cancer) which is
BRAF gain of function or BRAFV600E/D/K mutants.
[0018] In another embodiment of the combination of the invention,
dabrafenib, or a
pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable salt
thereof, and Compound C, or a pharmaceutically acceptable salt thereof, and
are in the same
formulation.
[0019] In another embodiment of the combination of the invention,
dabrafenib, or a
pharmaceutically acceptable salt thereof, Compound A, or a pharmaceutically
acceptable salt
thereof, and Compound C, or a pharmaceutically acceptable salt thereof, are in
separate
formulations.
[0020] In another embodiment, the combination of the invention is for
simultaneous
or sequential (in any order) administration.
[0021] In another embodiment, the present invention provides a method
for treating
cancer in a subject in need thereof comprising administering to the subject a
therapeutically
effective amount of the combination of the invention.
[0022] In a further embodiment of the method, the cancer is selected
from breast
cancer, cholangiocarcinoma, salivary gland cancer, colorectal cancer,
melanoma, non-small
cell lung cancer, ovarian cancer and thyroid cancer.
[0023] In a further embodiment, the present invention provides a
combination of the
invention for use in the manufacture of a medicament for treating a cancer
selected from
6

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
breast cancer, cholangiocarcinoma, salivary gland cancer, colorectal cancer,
melanoma, non-
small cell lung cancer, ovarian cancer and thyroid cancer.
[0024] In another embodiment there is provided a pharmaceutical
composition or
commercial package (e.g. a kit-of-parts) comprising the combination of the
invention.
[0025] In a further embodiment, the pharmaceutical composition further
comprises
one or more pharmaceutically acceptable excipients.
BRIEF DESCRIPTION OF THE DRAWINGS
[0026] Figure 1: Mice were randomized into treatment groups on Day 26
following
HT29 tumor cell implantation. Treatments were initiated on Day 26 and
continued until Day 39
post tumor cell implantation. Tumor dimensions and body weights were collected
at the time of
randomization and twice weekly thereafter for the study duration. Tumor
volumes (A) or percent
body weight change (B) from initial of treatment groups vs. days post
randomization are graphed.
Significant differences for tumor volume change were calculated using One-Way
Analysis of
variance (ANOVA) Tukey's multiple comparison test on Day 39 with N=9 mice per
group
(p<0.05, for Dabrafenib+Compound A+trametinib treated group versus vehicle,
single agents and
double combinations).
[0027] Figure 2: Mice were randomized into treatment groups on Day 28
following
HT29 tumor cell implantation. Treatments were initiated on Day 28 and
continued until Day 52
post tumor cell implantation. Tumor dimensions and body weights were collected
at the time of
randomization and twice weekly thereafter for the study duration. Tumor
volumes (A) or percent
body weight change (B) from initial of treatment groups vs. days post
randomization are graphed.
Significant differences were calculated using One-Way Analysis of variance
(ANOVA) Tukey's
multiple comparison test on Day 52 with N=7 mice per group, except untreated
control group
with n=4 (p<0. 0001, for both combinations treated groups versus non treated
controls).
[0028] Figure 3: Mice were randomized into treatment groups on Day 12
following
HCOX1329 tumor implantation. Treatments were initiated on Day 12 and continued
until Day 38
(vehicle), Day 62 (dabrafenib+ Compound A+trametinib), or Day 67
(dabrafenib+trametinib and
dabrafenib+trametinib+cetuximab) post tumor implantation. Tumor dimensions and
body
weights were collected at the time of randomization and twice weekly
thereafter for the study
7

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
duration. Tumor volumes (A) or percent body weight change (B) from initial of
treatment groups
vs. days post randomization are graphed. Significant differences were
calculated using One-Way
Analysis of variance (ANOVA) Tukey's multiple comparison test on Day 38 with
N=5 mice per
group, except dabrafenib+Compound A+trametinib group with n=4 (p =0.005 for
dabrafenib
+Compound A+trametinib vs dabrafenib+trametinib, and p=0.04 for dabrafenib +
Compound A +
trametinib vs dabrafenib+trametinib+cetiximab).
DESCRIPTION
[0029] The general terms used hereinbefore and hereinafter preferably
have within the
.. context of this disclosure the following meanings, unless otherwise
indicated, where more
general terms whereever used may, independently of each other, be replaced by
more specific
definitions or remain, thus defining more detailed embodiments of the
invention:
[0030] "Dabrafenib" is N-(3-(5-(2-aminopyrimidin-4-y1)-2-(tert-
butypthiazol-4-y1)-2-
fluoropheny1)-2,6-difluorobenzenesulfonamide, a selective inhibitor of mutated
BRAF at V600
capable of inhibiting BRAF(V600E), BRAF(V600K) and BRAF(V600G) mutations,
(also known
as: N-{345-(2-Amino-4-pyrimidiny1)-2-(1,1-dimethylethyl)-1,3-thiazol-4-yll -2-
fluorophenyl} -2,6-difluorobenzene sulfonamide ; Tafinlar ; & N- {3 [5 -(2-
Amino-4-
pyrimidiny1)-2-(1,1-dimethyle thyl)-1,3- thiazol-4-yll -2-fluorophenyl -2,6
difluorobenzene
sulfonamide, methane sulfonate salt).
[0031] "Cetuximab" is an epidermal growth factor receptor (EGFR) inhibitor
used for
the treatment of metastatic colorectal cancer, metastatic non-small cell lung
cancer and head
and neck cancer. Cetuximab is an epidermal growth factor receptor¨targetedIgG1
monoclonal
antibody that is approved for use in combination with irinotecan or as
monotherapy in the
treatment of metastatic CRC. Cetuximab is a chimeric (mouse/human) monoclonal
antibody
given by intravenous infusion.
[0032] "Compound A" is an inhibitor of extracellular signal-regulated
kinases (ERK)
1/2. "Compound A" is 4-(3-amino-6-((1S,3S,45)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-
y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-(methylamino)ethyl)-2-fluorobenzamide.
A
particularly preferred salt of Compound A is the hydrochloride salt thereof
8

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[0033] "Compound C", N-(3-(2-(2-hydroxyethoxy)-6-morpholinopyridin-4-
y1)-4-
methylpheny1)-2-(trifluoromethypisonicotinamide, is an ATP competitive
inhibitor of the
BRAF and CRAF protein kinases.
[0034] The term PD-1 inhibitors include PDR001. PDR001 is also known
as
spartalizumab, an anti-PD-1 antibody molecule described in US 2015/0210769,
published on July
30, 2015, entitled "Antibody Molecules to PD-1 and Uses Thereof," incorporated
by reference in
its entirety.
[0035] Further anti-PD-1 antibody molecules include the following:
[0036] Nivolumab (Bristol-Myers Squibb), also known as MDX-1106, MDX-
1106-04,
ONO-4538, BMS-936558, or OPDIVOO. Nivolumab (clone 5C4) and other anti-PD-1
antibodies are
disclosed in US 8,008,449 and WO 2006/121168, incorporated by reference in
their entirety;
[0037] Pembrolizumab (Merck & Co), also known as Lambrolizumab, MK-
3475, MK03475,
SCH-900475, or KEYTRUDAO. Pembrolizumab and other anti-PD-1 antibodies are
disclosed in
Hamid, 0. et al. (2013) New England Journal of Medicine 369 (2): 134-44, US
8,354,509, and WO
2009/114335, incorporated by reference in their entirety;
[0038] Pidilizumab (CureTech), also known as CT-011. Pidilizumab and
other anti-PD-1
antibodies are disclosed in Rosenblatt, J. et al. (2011)J Immunotherapy 34(5):
409-18, US 7,695,715,
US 7,332,582, and US 8,686,119, incorporated by reference in their entirety;
[0039] 1V1EDI0680 (Medimmune), also known as AMP-514. MEDI0680 and
other anti-PD-1
antibodies are disclosed in US 9,205,148 and WO 2012/145493, incorporated by
reference in their
entirety;
[0040] AMP-224 (B7-DCIg (Amplimmune), e.g., disclosed in WO
2010/027827 and WO
2011/066342, incorporated by reference in their entirety;
[0041] REGN2810 (Regeneron); PF-06801591 (Pfizer); BGB-A317 or BGB-108
(Beigene);
INCSHR1210 (Incyte), also known as INCSHR01210 or SHR-1210; TSR-042 (Tesaro),
also known
as ANB011; and further known anti-PD-1 antibodies including those described,
e.g., in WO
2015/112800, WO 2016/092419, WO 2015/085847, WO 2014/179664, WO 2014/194302,
WO
2014/209804, WO 2015/200119, US 8,735,553, US 7,488,802, US 8,927,697, US
8,993,731, and US
9,102,727, incorporated by reference in their entirety.
[0042] The term "subject" or "patient" as used herein is intended to
include animals,
which are capable of suffering from or afflicted with a cancer or any disorder
involving,
9

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
directly or indirectly, a cancer. Examples of subjects include mammals, e.g.,
humans, apes,
monkeys, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats,
and transgenic non-
human animals. In an embodiment, the subject is a human, e.g., a human
suffering from, at
risk of suffering from, or potentially capable of suffering from cancers.
[0043] The term "treating" or "treatment" as used herein comprises a
treatment
relieving, reducing or alleviating at least one symptom in a subject or
effecting a delay of
progression of a disease. For example, treatment can be the diminishment of
one or several
symptoms of a disorder or complete eradication of a disorder, such as cancer.
Within the
meaning of the present disclosure, the term "treat" also denotes to arrest,
delay the onset (i.e.,
the period prior to clinical manifestation of a disease) and/or reduce the
risk of developing or
worsening a disease.
[0044] The terms "comprising" and "including" are used herein in their
open-ended
and non-limiting sense unless otherwise noted.
[0045] The terms "a" and "an" and "the" and similar references in the
context of
describing the invention (especially in the context of the following claims)
are to be construed
to cover both the singular and the plural, unless otherwise indicated herein
or clearly
contradicted by context. Where the plural form is used for compounds, salts,
and the like, this
is taken to mean also a single compound, salt, or the like.
[0046] By "a combination" or "in combination with" or "co-administration
with" and
such like, it is not intended to imply that the therapy or the therapeutic
agents must be
physically mixed or administered at the same time and/or formulated for
delivery together,
although these methods of delivery are within the scope described herein. A
therapeutic agent
in these combinations can be administered concurrently with, prior to, or
subsequent to, one
or more other additional therapies or therapeutic agents. The therapeutic
agents or therapeutic
protocol can be administered in any order. In general, each agent will be
administered at a
dose and/or on a time schedule determined for that agent. It will further be
appreciated that
the additional therapeutic agent utilized in this combination may be
administered together in a
single composition or administered separately in different compositions. In
general, it is
expected that additional therapeutic agents utilized in combination be
utilized at levels that do

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
not exceed the levels at which they are utilized individually. In some
embodiments, the levels
utilized in combination will be lower than those utilized as single-agent
therapeutics.
[0047] When describing a dosage or dose herein as 'about' a specified
amount, the
actual dosage or dose can vary by up to 10%, e.g. 5%, from the stated amount:
this usage of
'about' recognizes that the precise amount in a given dose or dosage form may
differ slightly
from an intended amount for various reasons without materially affecting the
in vivo effect of
the administered compound. The skilled person will understand that where a
dose or dosage
of a therapeutic compound is quoted herein, that amount refers to the amount
of the
therapeutic compound in its free form or unsolvated form.
[0048] The phrase "therapeutically-effective amount" as used herein
means that
amount of a compound, material, or composition comprising a compound of the
present
invention which is effective for producing some desired therapeutic effect in
at least a sub-
population of cells in an animal (including a human) at a reasonable
benefit/risk ratio
applicable to any medical treatment.
[0049] 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
beings and
animals without excessive toxicity, irritation, allergic response, or other
problem or
complication, commensurate with a reasonable benefit/risk ratio.
[0050] The combinations of the invention, dabrafenib, compound A and
compound C
or spartalizumab, is also intended to represent unlabeled forms as well as
isotopically labeled
forms of the compounds. Isotopically labeled compounds have one or more atoms
replaced
by an atom having a selected atomic mass or mass number. Examples of isotopes
that can be
incorporated into dabrafenib, compound A and Compound C or spartalizumab
include
isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and
chlorine, such as
2H, 3H, 11C, 13C, 14C, 15N, 18F 31p, 32p, 35s, 36C1, 1231, 1241, 1251
respectively. The invention
includes isotopically labeled dabrafenib, compound A and compound C or
spartalizumab, for
example into which radioactive isotopes, such as 3H and '4C, or non-
radioactive isotopes,
such as 2H and '3C, are present. Isotopically labelled dabrafenib, compound A
and compound
C or spartalizumab are useful in metabolic studies (with '4C), reaction
kinetic studies (with,
11

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
for example 2H or 3H), detection or imaging techniques, such as positron
emission
tomography (PET) or single-photon emission computed tomography (SPECT)
including drug
or substrate tissue distribution assays, or in radioactive treatment of
patients. In particular,
dabrafenib, compound A or compound C or spartalizumab labeled with '8F may be
particularly desirable for PET or SPECT studies. Isotopically-labeled
compounds of the
invention can generally be prepared by conventional techniques known to those
skilled in the
art or by processes analogous to those described in the accompanying Examples
using
appropriate isotopically-labeled reagents.
[0051] Further, substitution with heavier isotopes, particularly
deuterium (i.e., 2H or
D) may afford certain therapeutic advantages resulting from greater metabolic
stability, for
example increased in vivo half-life or reduced dosage requirements or an
improvement in
therapeutic index. It is understood that deuterium in this context is regarded
as a substituent
of either dabrafenib, compound A or compound C or spartalizumab. The
concentration of
such a heavier isotope, specifically deuterium, may be defined by the isotopic
enrichment
factor. The term "isotopic enrichment factor" as used herein means the ratio
between the
isotopic abundance and the natural abundance of a specified isotope. If a
substituent
dabrafenib, compound A or compound C or spartalizumab is denoted deuterium,
such
compound has an isotopic enrichment factor for each designated deuterium atom
of at least
3500 (52.5% deuterium incorporation at each designated deuterium atom), at
least 4000 (60%
deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at
least 5000 (75%
deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at
least 6000 (90%
deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at
least 6466.7 (97%
deuterium incorporation), at least 6600 (99% deuterium incorporation), or at
least 6633.3
(99.5% deuterium incorporation).
Description of Preferred Embodiments
[0052] Dabrafenib is an orally bioavailable small molecule with RAF
inhibitory
activity. Compound A is an orally bioavailable small molecule with ERK
inhibitory activity.
It is an inhibitor of extracellular signal-regulated kinases 1 and 2 (ERK
1/2). Compound C is
an orally bioavailable small molecule with B/C-RAF inhibitory activity.
Spartalizumab is a
12

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
high-affinity, ligand-blocking, humanized anti-programmed death-1 (PD-1) IgG4
antibody
that blocks the binding of Programmed death-ligand 1 (PD-L1) and programmed
death-ligand
2 (PD-L2) to PD-1.
[0053] In one embodiment, with respect to the pharmaceutical
combination of the
invention, is a pharmaceutical combination comprising: N-(3-(5-(2-
aminopyrimidin-4-y1)-2-
(tert-butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof; 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3 -bromo-5 -fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof;
and N-(3-(2-
(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide (compound C), or a pharmaceutically acceptable salt thereof
[0054] In a further embodiment, N-(3-(5-(2-aminopyrimidin-4-y1)-2-
(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof, 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof,
and N-(3-(2-
(2-hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-
(trifluoromethyl)-
isonicotinamide (compound C), or a pharmaceutically acceptable salt thereof,
are
administered separately, simultaneously or sequentially, in any order.
[0055] In a further embodiment, the pharmaceutical combination is for oral
administration.
[0056] In a further embodiment of the pharmaceutical combination, N-(3-
(5-(2-
aminopyrimidin-4-y1)-2-(tert-butypthiazol-4-y1)-2-fluoropheny1)-2,6-
difluorobenzenesulfonamide (dabrafenib) is in an oral dosage form.
[0057] In a further embodiment of the pharmaceutical combination, 4-(3-
amino-6-
((lS,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-
fluoropheny1)-
2-(methylamino)ethyl)-2-fluorobenzamide (compound A) is in an oral dosage
form.
[0058] In a further embodiment of the pharmaceutical combination, N-(3-
(2-(2-
hydroxyethoxy)-6-morpholinopyridin-4-y1)-4-methylpheny1)-2-(trifluoromethyl)-
isonicotinamide (compound C) is in an oral dosage form.
13

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[0059] In another embodiment is a pharmaceutical composition or a
commercial
package comprising the pharmaceutical combination (as described in any of the
embodiments
above) and at least one pharmaceutically acceptable carrier.
[0060] In another embodiment is a pharmaceutical combination (as
described in any
of the embodiments above) or the pharmaceutical composition or the commercial
package (as
described in the embodiments above) for use in the treatment of cancer.
[0061] In a further embodiment, the cancer is selected from breast
cancer,
cholangiocarcinoma, colorectal cancer (CRC), melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer.
[0062] In a further embodiment, the cancer is advanced or metastatic
colorectal
cancer.
[0063] In a further embodiment, the cancer is BRAF gain of function
CRC or BRAF
V600E, V600D or V600K CRC.
[0064] In another embodiment is a use of the pharmaceutical
combination according
to any of the above embodiments or the pharmaceutical composition or
commercial package
according to the above embodiments for the manufacture of a medicament for the
treatment of
cancer.
[0065] In a further embodiment, the cancer is selected from breast
cancer,
cholangiocarcinoma, colorectal cancer, melanoma, non-small cell lung cancer,
ovarian cancer
and thyroid cancer, optionally wherein the cancer is advanced or metastatic
colorectal cancer,
optionally wherein the cancer is BRAF gain of function CRC or BRAF V600E,
V600D or
V600K CRC.
[0066] In another embodiment is a method of treating a cancer selected
from breast
cancer, cholangiocarcinoma, colorectal cancer, melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer comprising administrating to a patient in need
thereof a
pharmaceutical combination or commercial package according to any one of the
above
embodiemnts or the pharmaceutical composition according to the above
embodiments.
[0067] In a further embodiment, the colorectal cancer is advanced or
metastatic
colorectal cancer.
[0068] In a further embodiment, the colorectal cancer is BRAF gain of
function CRC
or BRAF V600E, V600D or V600K CRC.
14

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[0069] In a further embodiment, N-(3-(5-(2-aminopyrimidin-4-y1)-2-
(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide (dabrafenib)
is
administered orally at a dose of about from about 1 to about 150 mg per day
(for example, 1,
2, 5, 10, 50, 100 or 150 mg per day).
[0070] In a further embodiment, N-(3-(5-(2-aminopyrimidin-4-y1)-2-(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide (dabrafenib)
is
administered orally at a dose of 75mg BID.
[0071] In a further embodiment, 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A) is administered orally at a dose of from about
50 to about
200 mg per day (for example, at a dose of about 50, 75, 100, 125, 150, 175 or
200 mg per
day).
[0072] In a further embodiment, 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A) is administered orally at a dose of 100mg QD.
[0073] In a further embodiment, 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A) is administered orally at a dose of 200mg QD.
[0074] In a further embodiment, N-(3-(2-(2-hydroxyethoxy)-6-
morpholinopyridin-4-
y1)-4-methylpheny1)-2-(trifluoromethyl)-isonicotinamide (compound C) is
adminstered orally
at a dose of from about 100 mg per day, or 200 mg per day, or 300 mg per day
to about 400 mg
per day.
[0075] In a further embodiment, N-(3-(2-(2-hydroxyethoxy)-6-
morpholinopyridin-4-
y1)-4-methylpheny1)-2-(trifluoromethyl)-isonicotinamide (compound C) is
adminstered orally
at a dose of 200mg BID.
[0076] In one embodiment, with respect to the pharmaceutical
combination of the
invention, is a pharmaceutical combination comprising: N-(3-(5-(2-
aminopyrimidin-4-y1)-2-
(tert-butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof; 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-fluoropheny1)-2-
(methylamino)ethyl)-

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof;
and a PD-1
inhibitor, or a pharmaceutically acceptable salt thereof
[0077] In a further embodiment, N-(3-(5-(2-aminopyrimidin-4-y1)-2-
(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide
(dabrafenib), or a
pharmaceutically acceptable salt thereof, 4-(3-amino-6-((lS,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3 -bromo-5 -fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A), or a pharmaceutically acceptable salt thereof,
and a PD-1
inhibitor, or a pharmaceutically acceptable salt thereof, are administered
separately,
simultaneously or sequentially, in any order.
[0078] In another embodiment, the PD-1 inhibitor is an anti-PD-1 antibody
molecule.
[0079] In a further embodiment, the PD-1 inhibitor is an anti-PD-1
antibody molecule as
described in US 2015/0210769, published on July 30, 2015, entitled "Antibody
Molecules to PD-1 and
Uses Thereof," incorporated by reference in its entirety. In some embodiments,
the anti-PD-1
antibody molecule is BAP049-Clone E or BAP049-Clone B.
[0080] In a further embodiment, the anti-PD-1 antibody molecule is
Spartalizumab
(PDR001).
[0081] In one embodiment, the anti-PD-1 antibody molecule comprises at
least one, two,
three, four, five or six complementarity determining regions (CDRs) (or
collectively all of the
CDRs) from a heavy and light chain variable region comprising an amino acid
sequence shown in
Table 1 (e.g., from the heavy and light chain variable region sequences of
BAP049-Clone-E or
BAP049-Clone-B disclosed in Table 1), or encoded by a nucleotide sequence
shown in Table 1.
In some embodiments, the CDRs are according to the Kabat definition (e.g., as
set out in Table 1).
In some embodiments, the CDRs are according to the Chothia definition (e.g.,
as set out in Table
1). In some embodiments, the CDRs are according to the combined CDR
definitions of both
Kabat and Chothia (e.g., as set out in Table 1). In one embodiment, the
combination of Kabat and
Chothia CDR of VH CDR1 comprises the amino acid sequence GYTFTTYWMH (SEQ ID
NO:
541). In one embodiment, one or more of the CDRs (or collectively all of the
CDRs) have one,
two, three, four, five, six or more changes, e.g., amino acid substitutions
(e.g., conservative amino
acid substitutions) or deletions, relative to an amino acid sequence shown in
Table 1, or encoded
by a nucleotide sequence shown in Table 1.
16

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[0082] In one embodiment, the anti-PD-1 antibody molecule comprises a
heavy chain
variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO:
501, a
VHCDR2 amino acid sequence of SEQ ID NO: 502, and a VHCDR3 amino acid sequence
of
SEQ ID NO: 503; and a light chain variable region (VL) comprising a VLCDR1
amino acid
sequence of SEQ ID NO: 510, a VLCDR2 amino acid sequence of SEQ ID NO: 511,
and a
VLCDR3 amino acid sequence of SEQ ID NO: 512, each disclosed in Table 1.
[0083] In one embodiment, the antibody molecule comprises a VH
comprising a
VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 524, a VHCDR2 encoded
by the
nucleotide sequence of SEQ ID NO: 525, and a VHCDR3 encoded by the nucleotide
sequence of
SEQ ID NO: 526; and a VL comprising a VLCDR1 encoded by the nucleotide
sequence of SEQ
ID NO: 529, a VLCDR2 encoded by the nucleotide sequence of SEQ ID NO: 530, and
a
VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 531, each disclosed in
Table 1.
[0084] In one embodiment, the anti-PD-1 antibody molecule comprises a
VH comprising
the amino acid sequence of SEQ ID NO: 506, or an amino acid sequence at least
85%, 90%, 95%,
or 99% identical or higher to SEQ ID NO: 506. In one embodiment, the anti-PD-1
antibody
molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 520,
or an amino
acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO:
520. In one
embodiment, the anti-PD-1 antibody molecule comprises a VL comprising the
amino acid
sequence of SEQ ID NO: 516, or an amino acid sequence at least 85%, 90%, 95%,
or 99%
identical or higher to SEQ ID NO: 516. In one embodiment, the anti-PD-1
antibody molecule
comprises a VH comprising the amino acid sequence of SEQ ID NO: 506 and a VL
comprising
the amino acid sequence of SEQ ID NO: 520. In one embodiment, the anti-PD-1
antibody
molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 506
and a VL
comprising the amino acid sequence of SEQ ID NO: 516.
[0085] In one embodiment, the antibody molecule comprises a VH encoded by
the
nucleotide sequence of SEQ ID NO: 507, or a nucleotide sequence at least 85%,
90%, 95%, or
99% identical or higher to SEQ ID NO: 507. In one embodiment, the antibody
molecule
comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 521 or 517, or
a nucleotide
sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 521
or 517. In one
embodiment, the antibody molecule comprises a VH encoded by the nucleotide
sequence of SEQ
ID NO: 507 and a VL encoded by the nucleotide sequence of SEQ ID NO: 521 or
517.
17

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
[0086] In one embodiment, the anti-PD-1 antibody molecule comprises a
heavy chain
comprising the amino acid sequence of SEQ ID NO: 508, or an amino acid
sequence at least 85%,
90%, 95%, or 99% identical or higher to SEQ ID NO: 508. In one embodiment, the
anti-PD-1
antibody molecule comprises a light chain comprising the amino acid sequence
of SEQ ID NO:
522, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or
higher to SEQ ID
NO: 522. In one embodiment, the anti-PD-1 antibody molecule comprises a light
chain
comprising the amino acid sequence of SEQ ID NO: 518, or an amino acid
sequence at least 85%,
90%, 95%, or 99% identical or higher to SEQ ID NO: 518. In one embodiment, the
anti-PD-1
antibody molecule comprises a heavy chain comprising the amino acid sequence
of SEQ ID NO:
508 and a light chain comprising the amino acid sequence of SEQ ID NO: 522. In
one
embodiment, the anti-PD-1 antibody molecule comprises a heavy chain comprising
the amino
acid sequence of SEQ ID NO: 508 and a light chain comprising the amino acid
sequence of SEQ
ID NO: 518.
[0087] In one embodiment, the antibody molecule comprises a heavy
chain encoded by
the nucleotide sequence of SEQ ID NO: 509, or a nucleotide sequence at least
85%, 90%, 95%, or
99% identical or higher to SEQ ID NO: 509. In one embodiment, the antibody
molecule
comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 523
or 519, or a
nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ
ID NO: 523 or
519. In one embodiment, the antibody molecule comprises a heavy chain encoded
by the
nucleotide sequence of SEQ ID NO: 509 and a light chain encoded by the
nucleotide sequence of
SEQ ID NO: 523 or 519.
[0088] The antibody molecules described herein can be made by vectors,
host cells, and
methods described in US 2015/0210769, incorporated by reference in its
entirety.
Table 1. Amino acid and nucleotide sequences of exemplary anti-PD-1 antibody
molecules
BAP049-Clone-B HC
SEQ ID NO: 501 (Kabat) HCDR1 TYWMH
SEQ ID NO: 502 (Kabat) HCDR2 NIYPGTGGSNFDEKFKN
SEQ ID NO: 503 (Kabat) HCDR3 WTTGTGAY
SEQ ID NO: 504 HCDR1 GYTFTTY
(Chothia)
SEQ ID NO: 505 HCDR2 YPGTGG
(Chothia)
18

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
SEQ ID NO: 503 HCDR3 WTTGTGAY
(Chothia)
SEQ ID NO: 506 VH EVQLVQSGAEVKKPGESLRISCKGSGYTFTTYWMHWVRQ
ATGQGLEWMGNIYPGTGGSNEDEKEKNRVTITADKSTSTA
YMELSSLRSEDTAVYYCTRWTTGTGAYWGQGTTVTVSS
SEQ ID NO: 507 DNA VH GAGGTGCAGCTGGTGCAGTCAGGCGCCGAAGTGAAGAA
GCCCGGCGAGTCACTGAGAATTAGCTGTAAAGGTTCAG
GCTACACCTTCACTACCTACTGGATGCACTGGGTCCGCC
AGGCTACCGGTCAAGGCCTCGAGTGGATGGGTAATATC
TACCCCGGCACCGGCGGCTCTAACTTCGACGAGAAGTTT
AAGAATAGAGTGACTATCACCGCCGATAAGTCTACTAG
CACCGCCTATATGGAACTGTCTAGCCTGAGATCAGAGG
ACACCGCCGTCTACTACTGCACTAGGTGGACTACCGGCA
CAGGCGCCTACTGGGGTCAAGGCACTACCGTGACCGTG
TCTAGC
SEQ ID NO: 508 Heavy EVQLVQSGAEVKKPGESLRISCKGSGYTFTTYWMHWVRQ
chain ATGQGLEWMGNIYPGTGGSNEDEKEKNRVTITADKSTSTA
YMELSSLRSEDTAVYYCTRWTTGTGAYWGQGTTVTVSSA
STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWN
SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTC
NVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFP
PKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVE
VHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCK
VSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF
FLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLS
LG
SEQ ID NO: 509 DNA GAGGTGCAGCTGGTGCAGTCAGGCGCCGAAGTGAAGAA
heavy GCCCGGCGAGTCACTGAGAATTAGCTGTAAAGGTTCAG
chain GCTACACCTTCACTACCTACTGGATGCACTGGGTCCGCC
AGGCTACCGGTCAAGGCCTCGAGTGGATGGGTAATATC
TACCCCGGCACCGGCGGCTCTAACTTCGACGAGAAGTTT
AAGAATAGAGTGACTATCACCGCCGATAAGTCTACTAG
CACCGCCTATATGGAACTGTCTAGCCTGAGATCAGAGG
ACACCGCCGTCTACTACTGCACTAGGTGGACTACCGGCA
CAGGCGCCTACTGGGGTCAAGGCACTACCGTGACCGTG
TCTAGCGCTAGCACTAAGGGCCCGTCCGTGTTCCCCCTG
GCACCTTGTAGCCGGAGCACTAGCGAATCCACCGCTGCC
19

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
CTCGGCTGCCTGGTCAAGGATTACTTCCCGGAGCCCGTG
ACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGAGT
GCACACCTTCCCCGCTGTGCTGCAGAGCTCCGGGCTGTA
CTCGCTGTCGTCGGTGGTCACGGTGCCTTCATCTAGCCT
GGGTACCAAGACCTACACTTGCAACGTGGACCACAAGC
CTTCCAACACTAAGGTGGACAAGCGCGTCGAATCGAAG
TACGGCCCACCGTGCCCGCCTTGTCCCGCGCCGGAGTTC
CTCGGCGGTCCCTCGGTCTTTCTGTTCCCACCGAAGCCC
AAGGACACTTTGATGATTTCCCGCACCCCTGAAGTGACA
TGCGTGGTCGTGGACGTGTCACAGGAAGATCCGGAGGT
GCAGTTCAATTGGTACGTGGATGGCGTCGAGGTGCACA
ACGCCAAAACCAAGCCGAGGGAGGAGCAGTTCAACTCC
ACTTACCGCGTCGTGTCCGTGCTGACGGTGCTGCATCAG
GACTGGCTGAACGGGAAGGAGTACAAGTGCAAAGTGTC
CAACAAGGGACTTCCTAGCTCAATCGAAAAGACCATCT
CGAAAGCCAAGGGACAGCCCCGGGAACCCCAAGTGTAT
ACCCTGCCACCGAGCCAGGAAGAAATGACTAAGAACCA
AGTCTCATTGACTTGCCTTGTGAAGGGCTTCTACCCATC
GGATATCGCCGTGGAATGGGAGTCCAACGGCCAGCCGG
AAAACAACTACAAGACCACCCCTCCGGTGCTGGACTCA
GACGGATCCTTCTTCCTCTACTCGCGGCTGACCGTGGAT
AAGAGCAGATGGCAGGAGGGAAATGTGTTCAGCTGTTC
TGTGATGCATGAAGCCCTGCACAACCACTACACTCAGA
AGTCCCTGTCCCTCTCCCTGGGA
BAP049-Clone-B LC
SEQ ID NO: 510 (Kabat) LCDR1 KSSQSLLDSGNQKNFLT
................ + ..............................................
SEQ ID NO: 511 (Kabat) LCDR2 WASTRES
SEQ ID NO: 512 (Kabat) LCDR3 QNDYSYPYT
................ :-
SEQ ID NO: 513 L CDR1 SQSLLDSGNQKNF
(Chothia)
................ , ..............................................
SEQ ID NO: 514 ,, LCDR2 WAS
(Chothia)
SEQ ID NO: 515 LCDR3 DYSYPY
(Chothia)
SEQ ID NO: 516 VL EIVLTQSPATLSLSPGERATLSCKSSQSLLDSGNQKNFLTW
YQQKPGKAPKWYWASTRESGVPSRFSGSGSGTDFTFTISS
LQPEDIATYYCQNDYSYPYTFGQGTKVEIK

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
SEQ ID NO: 517 DNA VL GAGATCGTCCTGACTCAGTCACCCGCTACCCTGAGCCTG
AGCCCTGGCGAGCGGGCTACACTGAGCTGTAAATCTAG
TCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTTCCT
GACCTGGTATCAGCAGAAGCCCGGTAAAGCCCCTAAGC
TGCTGATCTACTGGGCCTCTACTAGAGAATCAGGCGTGC
CCTCTAGGTTTAGCGGTAGCGGTAGTGGCACCGACTTCA
CCTTCACTATCTCTAGCCTGCAGCCCGAGGATATCGCTA
CCTACTACTGTCAGAACGACTATAGCTACCCCTACACCT
TCGGTCAAGGCACTAAGGTCGAGATTAAG
SEQ ID NO: 518 Light EIVLTQSPATLSLSPGERATLSCKSSQSLLDSGNQKNFLTW
chain YQQKPGKAPKWYWASTRESGVPSRFSGSGSGTDFTFTISS
LQPEDIATYYCQNDYSYPYTFGQGTKVEIKRTVAAPSVFIF
PPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSG
NSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVT
HQGLSSPVTKSFNRGEC
SEQ ID NO: 519 DNA GAGATCGTCCTGACTCAGTCACCCGCTACCCTGAGCCTG
light AGCCCTGGCGAGCGGGCTACACTGAGCTGTAAATCTAG
chain TCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTTCCT
GACCTGGTATCAGCAGAAGCCCGGTAAAGCCCCTAAGC
TGCTGATCTACTGGGCCTCTACTAGAGAATCAGGCGTGC
CCTCTAGGTTTAGCGGTAGCGGTAGTGGCACCGACTTCA
CCTTCACTATCTCTAGCCTGCAGCCCGAGGATATCGCTA
CCTACTACTGTCAGAACGACTATAGCTACCCCTACACCT
TCGGTCAAGGCACTAAGGTCGAGATTAAGCGTACGGTG
GCCGCTCCCAGCGTGTTCATCTTCCCCCCCAGCGACGAG
CAGCTGAAGAGCGGCACCGCCAGCGTGGTGTGCCTGCT
GAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGA
AGGTGGACAACGCCCTGCAGAGCGGCAACAGCCAGGAG
AGCGTCACCGAGCAGGACAGCAAGGACTCCACCTACAG
CCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTACG
AGAAGCATAAGGTGTACGCCTGCGAGGTGACCCACCAG
GGCCTGTCCAGCCCCGTGACCAAGAGCTTCAACAGGGG
CGAGTGC
BAP049-Clone-E HC
................ :-
SEQ ID NO: 501 (Kabat) HCDR1 TYWMH
SEQ ID NO: 502 (Kabat) HCDR2 NIYPGTGGSNFDEKFKN
SEQ ID NO: 503 (Kabat) HCDR3 WTTGTGAY
__________________________________________________________________ ,
21

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
SEQ ID NO: 504 HCDR1 GYTFTTY
(Chothia)
SEQ ID NO: 505 HCDR2 YPGTGG
(Chothia)
________________ , ...............
SEQ ID NO: 503 HCDR3 WTTGTGAY
(Chothia)
SEQ ID NO: 506 VH EVQLVQSGAEVKKPGESLRISCKGSGYTFTTYWMHWVRQ
ATGQGLEWMGNIYPGTGGSNEDEKEKNRVTITADKSTSTA
YMELSSLRSEDTAVYYCTRWTTGTGAYWGQGTTVTVSS
SEQ ID NO: 507 DNA VH GAGGTGCAGCTGGTGCAGTCAGGCGCCGAAGTGAAGAA
GCCCGGCGAGTCACTGAGAATTAGCTGTAAAGGTTCAG
GCTACACCTTCACTACCTACTGGATGCACTGGGTCCGCC
AGGCTACCGGTCAAGGCCTCGAGTGGATGGGTAATATC
TACCCCGGCACCGGCGGCTCTAACTTCGACGAGAAGTTT
AAGAATAGAGTGACTATCACCGCCGATAAGTCTACTAG
CACCGCCTATATGGAACTGTCTAGCCTGAGATCAGAGG
ACACCGCCGTCTACTACTGCACTAGGTGGACTACCGGCA
CAGGCGCCTACTGGGGTCAAGGCACTACCGTGACCGTG
TCTAGC
SEQ ID NO: 508 Heavy EVQLVQSGAEVKKPGESLRISCKGSGYTFTTYWMHWVRQ
chain ATGQGLEWMGNIYPGTGGSNEDEKEKNRVTITADKSTSTA
YMELSSLRSEDTAVYYCTRWTTGTGAYWGQGTTVTVSSA
STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWN
SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTC
NVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFP
PKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVE
VHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCK
VSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQV
SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF
FLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLS
LG
SEQ ID NO: 509 DNA GAGGTGCAGCTGGTGCAGTCAGGCGCCGAAGTGAAGAA
heavy GCCCGGCGAGTCACTGAGAATTAGCTGTAAAGGTTCAG
chain GCTACACCTTCACTACCTACTGGATGCACTGGGTCCGCC
AGGCTACCGGTCAAGGCCTCGAGTGGATGGGTAATATC
TACCCCGGCACCGGCGGCTCTAACTTCGACGAGAAGTTT
AAGAATAGAGTGACTATCACCGCCGATAAGTCTACTAG
CACCGCCTATATGGAACTGTCTAGCCTGAGATCAGAGG
22

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
ACACCGCCGTCTACTACTGCACTAGGTGGACTACCGGCA
CAGGCGCCTACTGGGGTCAAGGCACTACCGTGACCGTG
TCTAGCGCTAGCACTAAGGGCCCGTCCGTGTTCCCCCTG
GCACCTTGTAGCCGGAGCACTAGCGAATCCACCGCTGCC
CTCGGCTGCCTGGTCAAGGATTACTTCCCGGAGCCCGTG
ACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGAGT
GCACACCTTCCCCGCTGTGCTGCAGAGCTCCGGGCTGTA
CTCGCTGTCGTCGGTGGTCACGGTGCCTTCATCTAGCCT
GGGTACCAAGACCTACACTTGCAACGTGGACCACAAGC
CTTCCAACACTAAGGTGGACAAGCGCGTCGAATCGAAG
TACGGCCCACCGTGCCCGCCTTGTCCCGCGCCGGAGTTC
CTCGGCGGTCCCTCGGTCTTTCTGTTCCCACCGAAGCCC
AAGGACACTTTGATGATTTCCCGCACCCCTGAAGTGACA
TGCGTGGTCGTGGACGTGTCACAGGAAGATCCGGAGGT
GCAGTTCAATTGGTACGTGGATGGCGTCGAGGTGCACA
ACGCCAAAACCAAGCCGAGGGAGGAGCAGTTCAACTCC
ACTTACCGCGTCGTGTCCGTGCTGACGGTGCTGCATCAG
GACTGGCTGAACGGGAAGGAGTACAAGTGCAAAGTGTC
CAACAAGGGACTTCCTAGCTCAATCGAAAAGACCATCT
CGAAAGCCAAGGGACAGCCCCGGGAACCCCAAGTGTAT
ACCCTGCCACCGAGCCAGGAAGAAATGACTAAGAACCA
AGTCTCATTGACTTGCCTTGTGAAGGGCTTCTACCCATC
GGATATCGCCGTGGAATGGGAGTCCAACGGCCAGCCGG
AAAACAACTACAAGACCACCCCTCCGGTGCTGGACTCA
GACGGATCCTTCTTCCTCTACTCGCGGCTGACCGTGGAT
AAGAGCAGATGGCAGGAGGGAAATGTGTTCAGCTGTTC
TGTGATGCATGAAGCCCTGCACAACCACTACACTCAGA
AGTCCCTGTCCCTCTCCCTGGGA
BAP049-Clone-E LC
SEQ ID NO: 510 (Kabat) LCDR1 KSSQSLLDSGNQKNFLT
SEQ ID NO: 511 (Kabat) LCDR2 WASTRES
SEQ ID NO: 512 (Kabat) LCDR3 QNDYSYPYT
................ .. ...............................................
SEQ ID NO: 513 L CDR1 SQSLLDSGNQKNF
(Chothia)
SEQ ID NO: 514 LCDR2 WAS
(Chothia)
SEQ ID NO: 515 LCDR3 DYSYPY
(Chothia)
........................ õ ........................................
23

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
SEQ ID NO: 520 VL EIVLTQSPATLSLSPGERATLSCKSSQSLLDSGNQKNFLTW
YQQKPGQAPRLLIYWASTRESGVPSRFSGSGSGTDFTFTISS
LEAEDAATYYCQNDYSYPYTFGQGTKVEIK
SEQ ID NO: 521 DNA VL GAGATCGTCCTGACTCAGTCACCCGCTACCCTGAGCCTG
AGCCCTGGCGAGCGGGCTACACTGAGCTGTAAATCTAG
TCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTTCCT
GACCTGGTATCAGCAGAAGCCCGGTCAAGCCCCTAGAC
TGCTGATCTACTGGGCCTCTACTAGAGAATCAGGCGTGC
CCTCTAGGTTTAGCGGTAGCGGTAGTGGCACCGACTTCA
CCTTCACTATCTCTAGCCTGGAAGCCGAGGACGCCGCTA
CCTACTACTGTCAGAACGACTATAGCTACCCCTACACCT
TCGGTCAAGGCACTAAGGTCGAGATTAAG
SEQ ID NO: 522 Light EIVLTQSPATLSLSPGERATLSCKSSQSLLDSGNQKNFLTW
chain YQQKPGQAPRLLIYWASTRESGVPSRFSGSGSGTDFTFTISS
LEAEDAATYYCQNDYSYPYTFGQGTKVEIKRTVAAPSVFIF
PPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSG
NSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVT
HQGLSSPVTKSFNRGEC
SEQ ID NO: 523 DNA GAGATCGTCCTGACTCAGTCACCCGCTACCCTGAGCCTG
light AGCCCTGGCGAGCGGGCTACACTGAGCTGTAAATCTAG
chain TCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTTCCT
GACCTGGTATCAGCAGAAGCCCGGTCAAGCCCCTAGAC
TGCTGATCTACTGGGCCTCTACTAGAGAATCAGGCGTGC
CCTCTAGGTTTAGCGGTAGCGGTAGTGGCACCGACTTCA
CCTTCACTATCTCTAGCCTGGAAGCCGAGGACGCCGCTA
CCTACTACTGTCAGAACGACTATAGCTACCCCTACACCT
TCGGTCAAGGCACTAAGGTCGAGATTAAGCGTACGGTG
GCCGCTCCCAGCGTGTTCATCTTCCCCCCCAGCGACGAG
CAGCTGAAGAGCGGCACCGCCAGCGTGGTGTGCCTGCT
GAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGA
AGGTGGACAACGCCCTGCAGAGCGGCAACAGCCAGGAG
AGCGTCACCGAGCAGGACAGCAAGGACTCCACCTACAG
CCTGAGCAGCACCCTGACCCTGAGCAAGGCCGACTACG
AGAAGCATAAGGTGTACGCCTGCGAGGTGACCCACCAG
GGCCTGTCCAGCCCCGTGACCAAGAGCTTCAACAGGGG
CGAGTGC
BAP049-Clone-B HC
SEQ ID NO: 524 (Kabat) :-
HCDR1 ACCTACTGGATGCAC
24

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
SEQ ID NO: 525 (Kabat) HCDR2 AATATCTACCCCGGCACCGGCGGCTCTAACTTCGACGAG
AAGTTTAAGAAT
SEQ ID NO: 526 (Kabat) HCDR3 TGGACTACCGGCACAGGCGCCTAC
......................... , ......................................
SEQ ID NO: 527 HCDR1 GGCTACACCTTCACTACCTAC
(Chothia)
......................... NS .....................................
SEQ ID NO: 528 HCDR2 TACCCCGGCACCGGCGGC
(Chothia)
SEQ ID NO: 526 HCDR3 TGGACTACCGGCACAGGCGCCTAC
(Chothia)
BAP049-Clone-B LC
......................... , ......................................
SEQ ID NO: 529 (Kabat) LCDR1 AAATCTAGTCAGTCACTGCTGGATAGCGGTAATCAGAA
GAACTTCCTGACC
SEQ ID NO: 530 (Kabat) LCDR2 TGGGCCTCTACTAGAGAATCA
SEQ ID NO: 531 (Kabat) LCDR3 CAGAACGACTATAGCTACCCCTACACC
......................... + ......................................
SEQ ID NO: 532 LCDR1 AGTCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTT
(Chothia) C
SEQ ID NO: 533 LCDR2 TGGGCCTCT
(Chothia)
SEQ ID NO: 534 LCDR3 GACTATAGCTACCCCTAC
(Chothia)
BAP049-Clone-E HC
......................... NS .....................................
SEQ ID NO: 524 (Kabat) HCDR1 ACCTACTGGATGCAC
SEQ ID NO: 525 (Kabat) HCDR2 AATATCTACCCCGGCACCGGCGGCTCTAACTTCGACGAG
AAGTTTAAGAAT
SEQ ID NO: 526 (Kabat) HCDR3 TGGACTACCGGCACAGGCGCCTAC
SEQ ID NO: 527 HCDR1 GGCTACACCTTCACTACCTAC
(Chothia)
SEQ ID NO: 528 HCDR2 TACCCCGGCACCGGCGGC
(Chothia)
SEQ ID NO: 526 HCDR3 TGGACTACCGGCACAGGCGCCTAC
(Chothia)
BAP049-Clone-E LC
SEQ ID NO: 529 (Kabat) LCDR1 AAATCTAGTCAGTCACTGCTGGATAGCGGTAATCAGAA
GAACTTCCTGACC
SEQ ID NO: 530 (Kabat) LCDR2 TGGGCCTCTACTAGAGAATCA
SEQ ID NO: 531 (Kabat) LCDR3 CAGAACGACTATAGCTACCCCTACACC
......................... ¨ ..........................

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
SEQ ID NO: 532 LCDR1 AGTCAGTCACTGCTGGATAGCGGTAATCAGAAGAACTT
(Chothia)
SEQ ID NO: 533 LCDR2 TGGGCCTCT
(Chothia)
SEQ ID NO: 534 LCDR3 GACTATAGCTACCCCTAC
(Chothia)
[0089] In a further embodiment of the pharmaceutical combination, N-(3-
(5-(2-
aminopyrimidin-4-y1)-2-(tert-butypthiazol-4-y1)-2-fluoropheny1)-2,6-
difluorobenzenesulfonamide (dabrafenib) is in an oral dosage form.
[0090] In a further embodiment of the pharmaceutical combination, 4-(3-
amino-6-
((1S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3-bromo-5-
fluoropheny1)-
2-(methylamino)ethyl)-2-fluorobenzamide (compound A) is in an oral dosage
form.
[0091] In another embodiment is a pharmaceutical composition or a
commercial
package comprising the pharmaceutical combination (as described in any of the
embodiments
above) and at least one pharmaceutically acceptable carrier.
[0092] In another embodiment is a pharmaceutical combination (as
described in any
of the embodiments above) or the pharmaceutical composition or the commercial
package (as
described in the embodiments above) for use in the treatment of cancer.
[0093] In a further embodiment, the cancer is selected from breast
cancer,
cholangiocarcinoma, colorectal cancer (CRC), melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer.
[0094] In a further embodiment, the cancer is advanced or metastatic
colorectal
cancer.
[0095] In a further embodiment, the cancer is BRAF gain of function
CRC or BRAF
V600E, V600D or V600K CRC.
[0096] In another embodiment is a use of the pharmaceutical
combination according
to any of the above embodiments or the pharmaceutical composition or
commercial package
according to the above embodiments for the manufacture of a medicament for the
treatment of
cancer.
[0097] In a further embodiment, the cancer is selected from breast cancer,
cholangiocarcinoma, colorectal cancer, melanoma, non-small cell lung cancer,
ovarian cancer
26

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
and thyroid cancer, optionally wherein the cancer is advanced or metastatic
colorectal cancer,
optionally wherein the cancer is BRAF gain of function CRC or BRAF V600E,
V600D or
V600K CRC.
[0098] In another embodiment is a method of treating a cancer selected
from breast
cancer, cholangiocarcinoma, colorectal cancer, melanoma, non-small cell lung
cancer, ovarian
cancer and thyroid cancer comprising administrating to a patient in need
thereof a
pharmaceutical combination or commercial package according to any one of the
above
embodiemnts or the pharmaceutical composition according to the above
embodiments.
[0099] In a further embodiment, the colorectal cancer is advanced or
metastatic
.. colorectal cancer.
[00100] In a further embodiment, the colorectal cancer is BRAF gain of
function CRC
or BRAF V600E, V600D or V600K CRC.
[00101] In a further embodiment, N-(3-(5-(2-aminopyrimidin-4-y1)-2-
(tert-
butypthiazol-4-y1)-2-fluoropheny1)-2,6-difluorobenzenesulfonamide (dabrafenib)
is
administered orally at a dose of about from about 1 to about 150 mg per day
(for example, 1,
2, 5, 10, 50, 100 or 150 mg per day).
[00102] In a further embodiment, 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-
hydroxycyclohexyl)pyrazin-2-y1)-N-((S)-1-(3 -bromo-5 -fluoropheny1)-2-
(methylamino)ethyl)-
2-fluorobenzamide (compound A) is administered orally at a dose of from about
50 to about
200 mg per day (for example, at a dose of about 50, 75, 100, 125, 150, 175 or
200 mg per
day).
[00103] In a further embodiment, the PD-1 inhibitor is administered at
a dose of about 300-
400 mg.
[00104] In a further embodiment, the PD-1 inhibitor is administered
once every 3 weeks or
once every 4 weeks.
[00105] In another embodiment, the PD-1 inhibitor is administered at a
dose of about 300 mg
once every 3 weeks.
[00106] In another embodiment, the PD-1 inhibitor is administered at a
dose of about 400 mg
once every 4 weeks.
27

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
Pharmacology and Utility
[00107] The RAS/RAF/MEK/ERK or mitogen activated protein kinase (MAPK)
pathway
is a key signaling cascade that integrates upstream cellular signals, such as
from growth factor
receptor tyrosine kinases, to orchestrate cell proliferation, differentiation,
and survival. The
MAPK signaling pathway is frequently dysregulated in human cancers, most
commonly through
mutation of members of the RAS family of genes. These mutations promote the
GTP-bound state
resulting in RAS activity leading in turn to activation of RAF, MEK, and ERK
proteins. RAS
mutations are found in multiple cancer types, including colorectal, lung, and
pancreatic cancers.
[00108] RAF (Rapidly Accelerated Fibrosarcoma) is a serine-threonine
protein kinase
discovered as a retroviral oncogene. The RAF family of proteins (ARAF, BRAF,
CRAF) signals
just downstream of activated RAS. Activated GTP-bound RAS recruits cytosolic
inactive RAF
monomers to the plasma membrane where RAF binds to GTP-RAS thereby promoting
homo- and
heterodimerization of RAF. The dimerization of RAF facilitates conformational
changes that lead
to catalytically activated RAF. Activated RAF dimers phosphorylate and
activate MEK1/2 (also
.. known as mitogen-activated protein kinase) proteins, which subsequently
phosphorylate and
activate extracellular signal-regulated kinases (ERK1/2). ERKs phosphorylate a
variety of
substrates, including multiple transcription factors, thereby regulating
several key cellular
activities, including proliferation, metabolism, migration, and survival. The
role of ERK1/2 in
cancer is of special interest because activating mutations upstream of ERK1/2
in its signaling
.. cascade are believed to be responsible for more than half of all cancers.
[00109] Dysregulated activation at any step in the MAPK pathway
contributes to
tumorigenesis. Activating BRAF mutations can be found in approximately 7% of
cancers, with
V600E accounting for greater than 90% of observed mutations in BRAF. The V600E
mutation
encodes a valine to glutamic acid substitution that exposes the active site of
BRAF, enabling its
constitutive activation as monomers or dimers independent of RAS. Inhibitors
of active RAF,
such as vemurafenib, dabrafenib, and encorafenib, have demonstrated dramatic
activity in BRAF
V600E metastatic melanoma with overall response rates (ORR) of 50-70%. The
success of these
inhibitors in V600E melanoma derives from the ability to bind to and inhibit
the mutant
monomeric form of RAF that is the oncogenic driver in cancer cells. However,
in cancer cells
that express wild-type BRAF, or in the normal cells of patients with V600E
driven cancers,
inhibitors such as vemurafenib paradoxically activate RAF signaling. The
complexity of MAPK
28

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
pathway signaling in the presence of monomeric RAF inhibitors is highlighted
in patients whose
BRAF V600E-dependent melanoma cells die while normal epidermal cells
containing wild-type
BRAF hyperproliferate. This paradoxical activation of RAF in wild-type cells
is precipitated by
the inhibitor's binding to one protomer of a RAF dimer. This leads to a
conformational change
that prevents inhibitor binding to the second protomer, and trans activation
of the second RAF
protomer of the dimer ensues. Inhibition at sequential nodes of the MAPK
pathway with RAF-
and MEK-directed combination therapy attenuates RAF dimer signaling in normal
cells, thereby
improving safety and clinical activity in metastatic BRAF V600 melanoma.
[00110] Single-agent RAF inhibitors or combination RAF/MEK inhibition
in BRAF
V600E colorectal cancer (CRC) demonstrate minimal activity; clinical benefit
is limited
compared to the activity seen in melanoma. Intrinsic and acquired resistance
to RAF inhibitors
and MEK inhibitors develop at multiple levels of the MAPK pathway. The
complexities of
signaling feedback and alternate pathways that circumvent BRAF inhibition are
central to the
challenge of targeting activated BRAF in CRC. Under physiologic conditions,
activated MAPK
.. signaling through mutant BRAF leads to ERK-dependent negative feedback on
signals generated
through activated RAS. Intrinsic resistance to RAF inhibition manifests
because drugs such as
vemurafenib or dabrafenib effectively inhibit BRAF V600E signaling through MEK
to ERK;
however, this in turn releases ERK-dependent negative feedback into RAS
signaling. Therefore,
upstream signals are able to activate RAS, leading to the induction of BRAF
V600E and wild-type
homo- and heterodimers. Because agents such as dabrafenib and vemurafenib
inhibit V600E
activated monomers in BRAF-dependent CRC cells, RAS-stimulated RAF dimer
signaling is
unopposed, leading to ERK reactivation to a greater degree than is seen in
BRAF V600E
melanoma, and thus limiting the effectiveness of therapy in CRC.
[00111] Under the pressure of BRAF and MEK inhibition in BRAF V600E
CRC, acquired
.. resistance quickly develops. For instance, in an analysis of nine tumor
samples from eight
patients experiencing disease progression after MAPK inhibition, genetic
alterations leading to
MAPK reactivation were uncovered. These included activating mutations in KRAS
or NRAS,
amplification of wild-type (WT)NRAS or KRAS or mutant BRAF V600E, and an
intragenic
deletion in BRAF V600E. Acquired genetic alterations have also been reported,
leading to
reactivation of ERK signaling in the face of MAPK inhibitors. Acquired
resistance may also arise
through complementary signaling in the tumor microenvironment.
29

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00112] Though previous therapeutic approaches to BRAF-mutant CRC have
focused on
chemotherapy and/or targeted therapy, there is also a role for immunotherapy.
During
tumorigenesis, cancer cells exploit immune checkpoint pathways to avoid
detection by the
adaptive immune system. Monoclonal antibody (mAb) inhibitors of the Programmed
Cell Death
Protein-1 (PD-1) and Programmed Death-Ligand 1 (PD-L1) immunological
checkpoints have
demonstrated significant antitumor activity in patients with various solid
tumors. PD-1 is a
particularly important immunological target, with inhibitors such as
pembrolizumab and
nivolumab demonstrating single-agent activity in melanoma, non-small cell lung
carcinoma
(NSCLC), and other solid tumors.
[00113] CRC, however, is generally unresponsive to PD-1 blockade with the
exception of
tumors possessing microsatellite instability. There is, however, rationale for
the use of small
molecule inhibitors to modulate the immune response. The same therapies that
inhibit genetic
dependencies on the MAPK pathway in cancer cells inhibit signaling cascades in
immune cells.
For instance, preclinical studies demonstrated that MAPK pathway inhibitors,
such as BRAF and
MEK inhibitors, could improve lymphocyte homing and function by increasing
tumor infiltrating
lymphocytes in tumors.
[00114] Therefore, RAF and MEK inhibitors may modulate the immune
response to
tumors, and the combination of such agents with checkpoint blockade may
increase the
susceptibility of "immune cold" tumors such as CRC to PD-1 inhibition.
Furthermore,
approximately 20% of BRAF-mutant CRCs are characterized by genetic
microsatellite instability
(MSI-H: microsatellite instability-high). In MSI-H CRC, irrespective of BRAF
genetic status,
single-agent anti-PD-1 therapy has been associated with response rates of 30-
50%. Furthermore,
targeted MAPK inhibition in tumor immune cells may complement the mechanism of
action of
anti-PD-1 antibodies in microsatellite stable and mismatch-repair deficient
CRC, thereby
potentially increasing anti-cancer immunomoduclation.
[00115] Lung cancer is a common type of cancer that affects men and
women around
the globe. NSCLC is the most common type (roughly 85%) of lung cancer with
approximately 70% of these patients presenting with advanced disease (Stage
IIIB or Stage
IV) at the time of diagnosis. About 30% of NSCLC tumors contain activating
KRAS
mutations, and these mutations are associated with resistance to EGFR tyrosine
kinase
inhibitors (TKIs). Activating KRAS mutations are also frequently found in
melanoma,

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
pancreatic cancer and ovarian cancer. BRAF mutations have been observed in up
to 3 % of
NSCLC and have also been described as a resistance mechanism in EGFR mutation
positive
NSCLC.
[00116] CRC is a common disease with more than 1.8 million new cases
estimated
worldwide in 2018, along with >800,000 deaths (World Health Organization,
Globocan 2018).
Mutations in genes encoding components of the MAPK pathway are common, with
RAS
mutations occurring in approximately 50% of CRC. Activating mutations in the
gene encoding
BRAF V600E are present in approximately 10-15% of CRC patients, and mutated
BRAF confers a
poor prognosis. The V600E mutation occurs in approximately 90% of BRAF-mutant
CRC,
though others, for example, V600D or V600K mutations are also seen.
[00117] Effective treatment options for BRAF-mutant CRC are limited.
Unlike melanoma,
where single-agent BRAF inhibitors yielded responses rates of approximately
70% in the
metastatic setting, single agent inhibition of metastatic BRAF-mutant CRC with
vemurafenib was
associated with an ORR of approximately 5%. Combination therapy with agents
targeting the
MAPK pathway have improved upon the effectiveness of BRAF inhibition, though
outcomes are
still poor. Dabrafenib combined with the MEK inhibitor trametinib was
associated with an ORR
of 12% and progression-free survival (PFS) of 3.5 months.
[00118] In CRC, stimulation of RAS through growth factor-mediated
receptor tyrosine
kinase activation supports the oncogenic milieu. Inhibitors of EGFR modestly
improved upon the
effectiveness of BRAF inhibition; BRAF inhibitors combined with EGFR
inhibitors were
associated with ORRs of 4-22% and PFS 3.2-4.2 months. Patients treated with
dabrafenib +
trametinib + panitumumab experienced an ORR of 21% and PFS of 4.2 months. In
the phase III
BEACON trial, patients were randomized to one of three arms in the 2nd-line of
treatment or
higher: encorafenib/binimetinib/cetuximab, encorafenib/cetuximab, versus
irinotecan/cetuximab
or FOLFIRI/cetuximab (control). Patients receiving triplet therapy achieved an
ORR of 26%,
PFS of 4.3 months, and overall survival (OS) of 9 months. Encorafenib plus
cetuximab was
associated with an ORR of 20% and PFS of 4.2 months, and OS of 8.4 months.
Both regimens
achieved statistically significant improvements over irinotecan or
FOLFIRI/cetuximab, which was
associated with an ORR of 2%, a PFS of 1.5 months, and OS of 5.4 months. The
improved
outcomes demonstrated by combined inhibition of RAF, MEK, and EGFR signaling
support the
31

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
concept that inhibition of multiple nodes within the MAPK pathway is required
for the treatment
of BRAF V600E CRC.
[00119] Dabrafenib (Tafinlar ) is an orally bioavailable, potent and
selective inhibitor of
RAF kinases, whose mechanism of action of is consistent with competitive
inhibition of
adenosine triphosphate (ATP) binding. The ability of dabrafenib to inhibit
some mutated forms of
BRAF kinases is concentration dependent, with in vitro IC50 values of 0.65,
0.5, and 1.84 nM for
BRAF V600E, BRAF V600K, and BRAF V600D enzymes, respectively. Inhibition of
wild-type
BRAF and CRAF kinases requires higher concentrations, with IC50 values of 3.2
and 5.0 nM,
respectively. Other kinases such as SIK1, NEK11, and LIMK1 may also be
inhibited at higher
concentrations. Dabrafenib inhibits cell growth of various BRAF V600 mutation-
positive tumors
in vitro and in vivo.
[00120] Dabrafenib was first approved by the FDA in 2013 as a single-
agent oral treatment
for unresectable or metastatic melanoma in adult patients with the BRAF V600
mutation and is
approved in various other countries for the same indication. Dabrafenib in
combination with
trametinib is also approved in multiple countries for the following
indications (approved
indications vary by country): treatment of patients with unresectable or
metastatic melanoma with
a BRAFV600 mutation; the adjuvant treatment of patients with Stage III
melanoma with a
BRAFV600 mutation, following complete resection; treatment of patients with
advanced non-
small cell lung cancer (NSCLC) with a BRAFV600 mutation; and treatment of
patients with
.. locally advanced or metastatic anaplastic thyroid cancer (ATC) with a
BRAFV600E mutation.
[00121] The recommended dose of dabrafenib is 150 mg BID (corresponding
to a total
daily dose of 300 mg).
[00122] Compound A is a potent, selective and orally bioavailable ATP-
competitive
ERK1/2 kinase inhibitor that exhibits physical chemical properties enabling
combinations with
RAF and MEK inhibitors, or other targeted therapeutic agents. Compound A
effectively inhibits
pERK signaling and has demonstrated tumor growth inhibition in multiple MAPK-
activated
cancer cells and xenograft models. Importantly, compound A demonstrated broad
efficacy
targeting multiple known mechanisms of resistance to BRAF and MEK inhibitors,
including RAS
mutations, BRAF splice variants and MEK 1/2 mutations, as shown in engineered
cell line models.
Compound A has been dosed in patients between 45 mg and 450 mg QD.
32

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00123] Clinical studies in BRAF V600E CRC have demonstrated that the
activity of
BRAF inhibitors alone or in combination with MEK EGFR inhibitors is limited
by insufficient
MAPK pathway suppression, and that in patients, mechanisms of resistance
quickly arise even in
the setting of initial clinical benefit. Acquired resistance mechanisms
leading to MAPK pathway
reactivation in patient tumors primarily involve activating genetic
alterations in RAS, BRAF or
MEK. This highlights the reliance of BRAF V600E CRC on MAPK signaling, and
suggests that
inhibition of ERK, the most downstream point of the signaling pathway, may
circumvent
resistance occurring at upstream nodes.
[00124] Preclinical models of RAS, RAF, or MEK resistance mutations
engineered into a
BRAF V600E cell line supported this concept. While the parental BRAF V600E
cell line was
sensitive to combinations of BRAF, MEK, EGFR, and/or ERK inhibitors, the
introduction of
KRAS, NRAS, MEK1, or MEK2 resistance mutations resulted in decreased
sensitivity of
engineered BRAF V600E cells to all inhibitor combinations, except for those
containing an ERK
inhibitor. Furthermore, the outgrowth of pre-existing, low-frequency pooled
resistant clones in
mouse xenografts was suppressed more effectively by treatment with drug
combinations
containing BRAF and ERK inhibitors, as compared to BRAF and MEK inhibitors.
[00125] The combination of Dabrafenib + Compound A was tested in vivo
in the BRAF
mutant human cell line xenograft HT29. Mice treated with Dabrafenib + Compound
A achieved
similar anti-tumor response as compared to Dabrafenib +Trametinib at
clinically relevant doses
(36% T/C vs 28% T/C, respectively). Single agent treatment led to progressive
disease, whereby
compound A achieved 54% T/C, Dabrafenib achieved 59% T/C, and Trametinib
achieved 48%
T/C. All regimens were tolerated as judged by lack of significant body weight
loss. These data
suggest that the combination of Dabrafenib + Compound A may achieve similar
anti-tumor
activity to Dabrafenib + Trametinib in patients with BRAF mutant colorectal
cancer, and provides
rationale for its use in the clinic.
[00126] The improved outcomes demonstrated by combined inhibition of
BRAF, MEK,
and EGFR signaling support the concept that inhibition of multiple nodes
within the MAPK
pathway is required for the treatment of BRAF V600 CRC.
[00127] Nonetheless, intrinsic and acquired resistance to therapy
remain important
challenges, and clinical outcomes are still poor. There is a role for
combination therapies that
provide more robust suppression of MAPK signaling and address the complexity
of mechanisms
33

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
of resistance both within and beyond the MAPK pathway. Given the adaptive
complexity of
signal transduction that characterizes BRA F-mut ant CRC, inhibition of
proteins beyond RAF and
ERK is required. As an illustration, one study of 218 BRAF-V600E mutated CRC
tumors
identified distinct subsets of tumors characterized by high
KRAS/mTOR/AKT/4EBP1/EMT
activation, while cell-cycle dysregulation characterized the other subset.
[00128] Despite the advances demonstrated by targeted therapy
combinations, such as
those studied in the BEACON trial (Kopetz et al. 2019), the ability to shut
down the BRAF V600
oncogenic drive in cancer cells is limited by 1.) the inability to fully
suppress BRAF activity due
the adaptive ability of RAF kinases to signal through ineffectively inhibited
dimers, and 2.)
ongoing ERK activation stimulated not only by adaptive mechanisms within the
MAPK pathway,
but also through parallel signaling pathways. Dabrafenib, vemurafenib, and
encorafenib
effectively suppress BRAF activity in BRAF-mutant cancer cells where monomeric
V600E is an
oncogenic driver. However, these drugs may also lead to the paradoxical
activation of ERK
through several mechanisms.
[00129] Combined inhibition of BRAF and MEK improves upon pathway
suppression;
however, the persistence of ERK signaling underlies the limitations of this
therapeutic approach.
Blockade of ERK, the ultimate signal of the MAPK pathway, may circumvent
adaptive upstream
signals and provide for improved efficacy and resilience to acquired
resistance.
[00130] BRAF-selective inhibitors are effective against constitutively
activated
monomeric BRAF V600; however, intrinsic and acquired resistance to RAF
inhibitors develop at
multiple levels of the MAPK pathway. Under steady-state conditions, activated
MAPK signaling
through BRAF V600E leads to ERK-dependent negative feedback on signals
generated through
activated RAS. In BRAF V600 CRC, intrinsic resistance to RAF inhibition
manifests because
drugs such as dabrafenib effectively inhibit monomeric BRAF V600E signaling
through MEK to
.. ERK; however, this in turn releases ERK-dependent negative feedback into
RAS signaling.
Therefore, upstream signals, such as through the epidermal growth factor
receptor (EGFR), are
able to activate RAS. This in turn leads to the induction of BRAF V600E and
wild-type homo-
and heterodimers, including homodimers and heterodimers of WT and BRAF-V600E,
CRAF and
BRAF-V600E and ARAF and BRAF-V600E that signal to MEK. Moreover, RAF
inhibitors such
as dabrafenib allosterically promote the homo- and hetero- dimerization of RAF
family members
34

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
such that inhibitor binds to only one RAF partner while the other unbound
dimer partner is
catalytically active in stimulating downstream signaling.
[00131] Therefore, agents such as dabrafenib target V600E monomers in
BRAF-dependent
CRC cells, leaving RAS-stimulated dimer signaling unopposed. This leads to ERK
reactivation to
a greater degree than is seen in BRAF V600E melanoma, thus limiting the
effectiveness of
therapy in CRC. In addition, CRAF plays an essential role in mediating
paradoxical activation
following BRAF-inhibitor treatment. Thus, RAF inhibitors, such as Compound C,
that potently
inhibit the activity of CRAF and BRAF can be effective in blocking BRAF-mutant
tumors and
RAS-driven adaptive MAPK activation. The role of CRAF in resistance to therapy
underlies the
rationale for the inclusion of an agent that inhibits both B- and CRAF in a
triple combination.
[00132] The triple combination of dabrafenib + Compound A + Compound C
can inhibit
the MAPK pathway in BRAF V600E/K/D colorectal cancer by leveraging the
potential to
uniquely target mechanisms of intrinsic and acquired resistance in BRAF V600-
driven cancer
cells.
[00133] Though single-agent checkpoint blockade is not effective in the
treatment of
microsatellite stable CRC, the treatment of MSI-H CRC with anti-PD-1
antibodies has been
associated with response rates of 31-50%. Though approximately 21% of BRAF
V600E CRC
may exhibit MSI-H status, microsatellite instability does not appear to modify
responsiveness to
MAPK-targeted therapy in this disease. Therefore, subjects with MSI-H BRAF
V600E CRC may
respond to either RAF/MEK/ERK-targeted therapy or checkpoint blockade. By
addressing the
co-occurring features of oncogenic BRAF and immunotherapy responsiveness in
MSI-H BRAF-
mutant CRC, the combination of MAPK pathway inhibition with checkpoint
blockade has the
potential to improve upon the outcomes achieved with either category of
therapy alone.
[00134] Furthermore, targeted small molecule inhibitors may modulate
the immune
microenvironment. For instance, preclinical studies demonstrated that MAPK
pathway inhibitors
could improve lymphocyte homing and function by increasing tumor infiltrating
lymphocytes in
tumors, decreasing upregulated immunosuppressive cytokines, and generally
counteracting
immune tolerance of cancer. Furthermore, the BRAF-MAPK signaling pathway is
essential for
cancer-immune evasion in human melanoma cells. Therefore, RAF and MEK
inhibitors can
modulate the immune response to tumors, and the combination of such agents
with checkpoint

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
blockade can even increase the susceptibility of "immune cold" tumors, such as
microsatellite
stable CRC, to PD-1 inhibition.
[00135] The triple combination of dabrafenib + Compound A +
Spartalizumab can inhibit
the MAPK pathway in BRAF V600E/K/D colorectal cancer by leveraging the
potential to
uniquely target mechanisms of intrinsic and acquired resistance in BRAF V600-
driven cancer
cells.
Pharmaceutical Compositions
[00136] In another aspect, the present invention provides
pharmaceutically acceptable
compositions which comprise a therapeutically-effective amount of dabrafenib,
compound A
and compound C, formulated together with one or more pharmaceutically
acceptable carriers
(additives) and/or diluents. As described in detail below, the pharmaceutical
compositions of
the present invention may be specially formulated for administration in solid
or liquid form,
including those adapted for oral administration, for example, drenches
(aqueous or non-
aqueous solutions or suspensions), tablets, e.g., those targeted for buccal,
sublingual, and
systemic absorption, boluses, powders, granules, pastes for application to the
tongue.
[00137] The phrase "pharmaceutically-acceptable carrier" as used herein
means a
pharmaceutically-acceptable material, composition or vehicle, such as a liquid
or solid filler,
diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium,
calcium or zinc
stearate, or steric acid), or solvent encapsulating material, involved in
carrying or transporting
the subject compound from one organ, or portion of the body, to another organ,
or portion of
the body. Each carrier must be "acceptable" in the sense of being compatible
with the other
ingredients of the formulation and not injurious to the patient. Some examples
of materials
which can serve as pharmaceutically-acceptable carriers include: (1) sugars,
such as lactose,
glucose and sucrose; (2) starches, such as corn starch and potato starch; (3)
cellulose, and its
derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and
cellulose acetate; (4)
powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as
cocoa butter and
suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower
oil, sesame oil, olive
oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11)
polyols, such as
glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as
ethyl oleate and ethyl
36

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and
aluminum
hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline;
(18) Ringer's
solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters,
polycarbonates
and/or polyanhydrides; and (22) other non-toxic compatible substances employed
in
pharmaceutical formulations.
[00138] As set out above, certain embodiments of the present compounds
may contain
a basic functional group, such as amino or alkylamino, and are, thus, capable
of forming
pharmaceutically-acceptable salts with pharmaceutically-acceptable acids. The
term
"pharmaceutically-acceptable salts" in this respect, refers to the relatively
non-toxic, inorganic
and organic acid addition salts of compounds of the present invention. These
salts can be
prepared in situ in the administration vehicle or the dosage form
manufacturing process, or by
separately reacting a purified compound of the invention in its free base form
with a suitable
organic or inorganic acid, and isolating the salt thus formed during
subsequent purification.
Representative salts include the hydrobromide, hydrochloride, sulfate,
bisulfate, phosphate,
nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate,
lactate, phosphate,
tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate,
mesylate, glucoheptonate,
lactobionate, and laurylsulphonate salts and the like.
[00139] The pharmaceutically acceptable salts of the subject compounds
include the
conventional nontoxic salts or quaternary ammonium salts of the compounds,
e.g., from non-
toxic organic or inorganic acids. For example, such conventional nontoxic
salts include those
derived from inorganic acids such as hydrochloride, hydrobromic, sulfuric,
sulfamic,
phosphoric, nitric, and the like; and the salts prepared from organic acids
such as acetic,
propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric,
ascorbic, palmitic, maleic,
hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic, sulfanilic, 2-
acetoxybenzoic,
fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic,
isothionic, and the like.
[00140] In other cases, the compounds of the present invention may
contain one or
more acidic functional groups and, thus, are capable of forming
pharmaceutically-acceptable
salts with pharmaceutically-acceptable bases. The term "pharmaceutically-
acceptable salts"
in these instances refers to the relatively non-toxic, inorganic and organic
base addition salts
of compounds of the present invention. These salts can likewise be prepared in
situ in the
administration vehicle or the dosage form manufacturing process, or by
separately reacting
37

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
the purified compound in its free acid form with a suitable base, such as the
hydroxide,
carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with
ammonia, or
with a pharmaceutically-acceptable organic primary, secondary or tertiary
amine.
Representative alkali or alkaline earth salts include the lithium, sodium,
potassium, calcium,
magnesium, and aluminum salts and the like. Representative organic amines
useful for the
formation of base addition salts include ethylamine, diethylamine,
ethylenediamine,
ethanolamine, diethanolamine, piperazine and the like.
[00141] A particularly preferred salt of dabrafenib is the mesylate
salt thereof A
particularly preferred solvate of compound A is the hydrochloride salt thereof
A particularly
preferred form of compound C is the free base crystalline form.
[00142] Wetting agents, emulsifiers and lubricants, such as sodium
lauryl sulfate and
magnesium stearate, as well as coloring agents, release agents, coating
agents, sweetening,
flavoring and perfuming agents, preservatives and antioxidants can also be
present in the
compositions.
[00143] Examples of pharmaceutically-acceptable antioxidants include: (1)
water
soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium
bisulfate, sodium
metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such
as ascorbyl
palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT),
lecithin, propyl
gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such
as citric acid,
ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric
acid, and the like.
[00144] Formulations of the present invention include those suitable
for oral, nasal,
topical (including buccal and sublingual), rectal, vaginal and/or parenteral
administration.
The formulations may conveniently be presented in unit dosage form and may be
prepared by
any methods well known in the art of pharmacy. The amount of active ingredient
which can
be combined with a carrier material to produce a single dosage form will vary
depending upon
the host being treated, the particular mode of administration. The amount of
active ingredient
which can be combined with a carrier material to produce a single dosage form
will generally
be that amount of the compound which produces a therapeutic effect. Generally,
out of one
hundred per cent, this amount will range from about 0.1 per cent to about
ninety-nine percent
of active ingredient, preferably from about 5 per cent to about 70 per cent,
most preferably
from about 10 percent to about 30 percent.
38

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00145] In certain embodiments, a formulation of the present invention
comprises an
excipient selected from the group consisting of cyclodextrins, celluloses,
liposomes, micelle
forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and
polyanhydrides;
and a compound of the present invention. In certain embodiments, an
aforementioned
formulation renders orally bioavailable a compound of the present invention.
[00146] Methods of preparing these formulations or compositions include
the step of
bringing into association a compound of the present invention with the carrier
and, optionally,
one or more accessory ingredients. In general, the formulations are prepared
by uniformly
and intimately bringing into association a compound of the present invention
with liquid
carriers, or finely divided solid carriers, or both, and then, if necessary,
shaping the product.
[00147] Formulations of the invention suitable for oral administration
may be in the
form of capsules, cachets, pills, tablets, lozenges (using a flavored basis,
usually sucrose and
acacia or tragacanth), powders, granules, or as a solution, suspension or
solid dispersion in an
aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid
emulsion, or as an
elixir or syrup, or as pastilles (using an inert base, such as gelatin and
glycerin, or sucrose and
acacia) and/or as mouth washes and the like, each containing a predetermined
amount of a
compound of the present invention as an active ingredient. A compound of the
present
invention may also be administered as a bolus, electuary or paste.
[00148] In solid dosage forms of the invention for oral administration
(capsules,
tablets, pills, dragees, powders, granules, trouches and the like), the active
ingredient is mixed
with one or more pharmaceutically-acceptable carriers, such as sodium citrate
or dicalcium
phosphate, and/or any of the following: (1) fillers or extenders, such as
starches, lactose,
sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for
example,
carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose
and/or acacia; (3)
humectants, such as glycerol; (4) disintegrating agents, such as agar-agar,
calcium carbonate,
potato or tapioca starch, alginic acid, certain silicates, and sodium
carbonate; (5) solution
retarding agents, such as paraffin; (6) absorption accelerators, such as
quaternary ammonium
compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7)
wetting agents,
such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic
surfactants; (8)
absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc,
calcium stearate,
magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, zinc
stearate, sodium
39

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
stearate, stearic acid, and mixtures thereof; (10) coloring agents; and (11)
controlled release
agents such as crospovidone or ethyl cellulose. In the case of capsules,
tablets and pills, the
pharmaceutical compositions may also comprise buffering agents. Solid
compositions of a
similar type may also be employed as fillers in soft and hard-shelled gelatin
capsules using
such excipients as lactose or milk sugars, as well as high molecular weight
polyethylene
glycols and the like.
[00149] A tablet may be made by compression or molding, optionally with
one or more
accessory ingredients. Compressed tablets may be prepared using binder (for
example,
gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent,
preservative, disintegrant
(for example, sodium starch glycolate or cross-linked sodium carboxymethyl
cellulose),
surface-active or dispersing agent. Molded tablets may be made by molding in a
suitable
machine a mixture of the powdered compound moistened with an inert liquid
diluent.
[00150] The tablets, and other solid dosage forms of the pharmaceutical
compositions
of the present invention, such as dragees, capsules, pills and granules, may
optionally be
scored or prepared with coatings and shells, such as enteric coatings and
other coatings well
known in the pharmaceutical-formulating art. They may also be formulated so as
to provide
slow or controlled release of the active ingredient therein using, for
example,
hydroxypropylmethyl cellulose in varying proportions to provide the desired
release profile,
other polymer matrices, liposomes and/or microspheres. They may be formulated
for rapid
release, e.g., freeze-dried. They may be sterilized by, for example,
filtration through a
bacteria-retaining filter, or by incorporating sterilizing agents in the form
of sterile solid
compositions which can be dissolved in sterile water, or some other sterile
injectable medium
immediately before use. These compositions may also optionally contain
opacifying agents
and may be of a composition that they release the active ingredient(s) only,
or preferentially,
in a certain portion of the gastrointestinal tract, optionally, in a delayed
manner. Examples of
embedding compositions which can be used include polymeric substances and
waxes. The
active ingredient can also be in micro-encapsulated form, if appropriate, with
one or more of
the above-described excipients.
[00151] Liquid dosage forms for oral administration of the compounds of
the invention
include pharmaceutically acceptable emulsions, microemulsions, solutions,
suspensions,
syrups and elixirs. In addition to the active ingredient, the liquid dosage
forms may contain

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
inert diluents commonly used in the art, such as, for example, water or other
solvents,
solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol,
ethyl carbonate,
ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene
glycol, oils (in
particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils),
glycerol,
tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of
sorbitan, and mixtures
thereof.
[00152] Besides inert diluents, the oral compositions can also include
adjuvants such as
wetting agents, emulsifying and suspending agents, sweetening, flavoring,
coloring,
perfuming and preservative agents.
[00153] Suspensions, in addition to the active compounds, may contain
suspending
agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene
sorbitol and sorbitan
esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-
agar and
tragacanth, and mixtures thereof
[00154] Examples of suitable aqueous and nonaqueous carriers which may
be
employed in the pharmaceutical compositions of the invention include water,
ethanol, polyols
(such as glycerol, propylene glycol, polyethylene glycol, and the like), and
suitable mixtures
thereof, vegetable oils, such as olive oil, and injectable organic esters,
such as ethyl oleate.
Proper fluidity can be maintained, for example, by the use of coating
materials, such as
lecithin, by the maintenance of the required particle size in the case of
dispersions, and by the
use of surfactants.
[00155] These compositions may also contain adjuvants such as
preservatives, wetting
agents, emulsifying agents and dispersing agents. Prevention of the action of
microorganisms
upon the subject compounds may be ensured by the inclusion of various
antibacterial and
antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid,
and the like. It
may also be desirable to include isotonic agents, such as sugars, sodium
chloride, and the like
into the compositions.
[00156] When the compounds of the present invention are administered as
pharmaceuticals, to humans and animals, they can be given per se or as a
pharmaceutical
composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%)
of active
ingredient in combination with a pharmaceutically acceptable carrier.
41

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00157] The compounds of the present invention and/or the
pharmaceutical
compositions of the present invention, are formulated into pharmaceutically-
acceptable
dosage forms by conventional methods known to those of skill in the art.
[00158] Actual dosage levels of the active ingredients in the
pharmaceutical
compositions of this invention may be varied so as to obtain an amount of the
active
ingredient which is effective to achieve the desired therapeutic response for
a particular
patient, composition, and mode of administration, without being toxic to the
patient.
[00159] The selected dosage level will depend upon a variety of factors
including the
activity of the particular compound of the present invention employed, or the
ester, salt or
amide thereof, the route of administration, the time of administration, the
rate of excretion or
metabolism of the particular compound being employed, the rate and extent of
absorption, the
duration of the treatment, other drugs, compounds and/or materials used in
combination with
the particular compound employed, the age, sex, weight, condition, general
health and prior
medical history of the patient being treated, and like factors well known in
the medical arts.
[00160] A physician or veterinarian having ordinary skill in the art can
readily
determine and prescribe the effective amount of the pharmaceutical composition
required.
For example, the physician or veterinarian could start doses of the compounds
of the
invention employed in the pharmaceutical composition at levels lower than that
required in
order to achieve the desired therapeutic effect and gradually increase the
dosage until the
desired effect is achieved.
[00161] In general, a suitable daily dose of the combination of the
invention will be
that amount of each compound which is the lowest dose effective to produce a
therapeutic
effect. Such an effective dose will generally depend upon the factors
described above.
[00162] In another aspect, the present invention provides
pharmaceutically acceptable
compositions which comprise a therapeutically-effective amount of one or more
of the subject
compounds, as described above, formulated together with one or more
pharmaceutically
acceptable carriers (additives) and/or diluents.
42

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
Examples
Example 1
Dabrafenib, Compound A and Compound C
[00163] Dabrafenib is synthesized according to example 58a of
W02009/137391.
Compound A is synthesized according to example 184 of W02015/066188. Compound
C is
synthesized according to example 1156 of W02014/151616. W02009/137391,
W02015/066188 and W02014/151616, are herein incorporated by reference in their
entirety.
The utility of a combination of Dabrafenib, Compound A and Compound C
described herein
can be evidenced by testing in the following examples.
Example 2
Combination efficacy of MAPK pathway inhibitors in the human BRAF mutant CRC
xenograft
model HT29 in nude mice
[00164] Dabrafenib (DRB436): selective inhibitor of mutated BRAF at
V600 capable of
inhibiting BRAF(V600E), BRAF(V600K) and BRAF(V600G) mutations. Compound A:
selective ATP-competitive ERK1 and ERK2 kinase inhibitor. Compound C: an ATP
competitive
inhibitor of BRAF and CRAF. Dabrafenib was dosed p.o. in vehicle: 0.5% HPMC +
0.2% Tween
80 in pH 8 DI water. Compound A was dosed p.o. in vehicle: 0.5% HPC / 0.5%
Pluronic F127 in
a pH 7.4 phosphate buffer, adjusted to pH 4.0 with acid. Compound C (free base
crystalline form,
in powder form) was dosed p.o. in MEPC4 vehicle (45% Cremophor RH40 + 27%
PEG400 +
18% Capmul MCM C8 + 10% ethanol).
[00165] The HT29 human colorectal cancer (CRC) tumor cell line was
purchased from
ATCC and was included in the Novartis Cell Line Encyclopedia (CLE) cell line
collection. The
line has been shown to be free ofMycoplasma sp. and murine viruses in the
IMPACT-VIII PCR
assay panel (Research Animal Diagnostic Laboratory (RADIL), University of
Missouri,
Columbia, MO). The cells were maintained in EMEM (Lonza #12-611F) plus 10% FBS
(Gibco
#26140-079) (56 C for 30 min. inactivated), at 37 C in a humidified atmosphere
containing 5%
carbon dioxide. Cells were harvested at 80-95% confluence with 0.25% trypsin-
EDTA (Gibco
#25200-056), neutralized with growth medium, after centrifugation for 5 min at
1200 rpm,
43

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
followed by resuspension of the cell pellet in cold HBSS (Gibco #14175-095)
and then mixed
with an equal volume of MatrigelTM Matrix (Corning #354234) to prepare a final
concentration of
10x106ce11s/mL. Then 200p.1 (2 x 106 cells) was implanted subcutaneously into
the right flank of
female nude mice. Tumor volume was determined by measurement with calipers and
calculated
using a formula, where tumor volume (TV) (rnm3) = (1 x w2)/2, where 1 is the
longest axis of the
tumor and w is perpendicular to 1. Mice were monitored for tumor growth and
body weight
twice/week. Animal well-being and behavior, including grooming and ambulation,
were
monitored twice weekly. General health of mice was monitored daily.
[00166] The HCOX1329 CRC patient-derived tumor xenograft (PDX) was
propagated by
serial passage of tumor slurry in nude mice. Briefly, fragments of fresh tumor
from a previous
passage were homogenized using gentleMACS Dissociator (MACS (Miltenyi Biotec,
#120-005-
331), passed through a tissue grinder (Chemglass lifeSciences # CLS-5020-085),
and diluted in
PBS. Then 4x10^6 cells in 100p.1 of tumor slurry was implanted subcutaneously
into the right
flank of female nude mice as passage 7. Tumor volume was determined by
measurement with
calipers and calculated using a formula, where tumor volume (TV) (rnm3) = (1 x
w2)/2, where 1 is
the longest axis of the tumor and w is perpendicular to 1. Mice were monitored
for tumor growth
and body weight twice/week. Animal well-being and behavior, including grooming
and
ambulation, were monitored twice weekly. General health of mice was monitored
daily.
[00167] The
efficacy study design for each model is described in Table 1, Table 2 and
table 3. Test agents were dosed at dose volume of 10 mL/kg, which was adjusted
according to
body weight. Tumor dimensions and body weights were collected at the time of
randomization
and twice weekly thereafter for the study duration.
Table 1
Number
Group of mice Compound Dose (mg/kg) Route Schedule
1 9 Vehicle p.o.
2 9 Dabrafenib 30 p.o. qd
3 9 Trametinib 0.3 p.o. qd
4 9 Compound A 75 p.o. qd
5 9 Dabrafenib+trametinib 30+0.3 p.o+p.o. qd+qd
6 9 Dabrafenib+Compound A 30+75 p.o+p.o. qd+qd
7 9 Dabrafenib+trametinib+Compound A 30+0.3+75
p.o.+p.o.+p.o qd+qd+qd
44

CA 03173356 2022-08-26
WO 2021/171260 PCT/IB2021/051641
Table 2
Number
Group of mice Compound Dose (mg/kg) Route
Schedule
1 7 Vehicle p.o.
2 7 Dabrafenib+trametinib+Compound A 30+0.15+75
p.o.+p.o.+p.o 0+0+0
3 7 Dabrafenib+ Compound A+Compound C 30+75+25
p.o.+p.o.+p.o cid+cid+bid
Table 3
Number of
Group mice Compound Dose (mg/kg) Route Schedule
1 5 Vehicle p.o.
2 5 Dabrafenib+trametinib 30+0.3 p.o.+p.o.
cid+cid
3 5 Dabrafenib+trametinib+ 30+0.3+20 p.o.
+p.o.+i.p. cid+cid+biw
cetuximab
4 5 Dabrafenib+trametinib+ 30+0.15+75
p.o.+p.o.+p.o 0+0+0
Compound A
p.o.: per os (oral gavage); i.p.: intraperitoneal; qd: once a day; bid: twice
a day; biw: twice a week.
The percent change in body weight was calculated as (BWctinent -
BWinitiai)/(BWinitiai) X 100%. Data
was presented as mean percent body weight change from the day of treatment
initiation SEM.
Tumor volume: Percent treatment/control (%T/C) values were calculated using
the following
formula: %T/C = 100 x AT/AC if AT > 0 ; % Regression = 100 X AT/Tinitial if AT
< 0; where:
T = mean tumor volume of the drug-treated group on the final day of the study;
AT = mean tumor volume of the drug-treated group on the final day of the study
¨ mean tumor
volume of the drug-treated group on initial day of dosing;
Tjniijai = mean tumor volume of the drug-treated group on initial day of
dosing;
C = mean tumor volume of the control group on the final day of the study; and
AC = mean tumor volume of the control group on the final day of the study ¨
mean tumor volume
of the control group on initial day of dosing.
All data were expressed as mean standard error of the mean (SEM). Percent
change in tumor
volume and body weight were used for statistical analysis. Between group
comparisons were
carried out using a one-way ANOVA followed by a Tukey's multiple comparisons
test. For all
statistical evaluations the level of significance was set at p <0.05.

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00168] The antitumor efficacy of MAPK pathway inhibitors was examined
in two studies
using the BRAF mutant HT29 human CRC xenograft model in athymic nude mice. In
the first
study (table 1), mice were treated with single agents or combinations of MAPK
pathway
inhibitors as described on Table 1 until vehicle-treated tumors achieved a
volume >1000 rnm3, 13
days post treatment initiation. Anti-tumor activity was determined by
assessing %T/C or %
regression on day 39 post implant (13 days post treatment initiation). Anti-
tumor activity, mean
change in tumor volume, mean change in body weight and survival 13 days post
treatment
initiation is reported in Table 3. Tumor volume and body weight change post
treatment are
plotted on Figure 1. Daily single agent treatment with Compound A, dabrafenib
or trametinib
achieved 54%T/C, 59%T/C, or 48%T/C, respectively, when compared to the vehicle
treated
group. The combined anti-tumor activity of Compound A+dabrafenib (35% T/C) was
similar to
the anti-tumor activity achieved by the trametinib+dabrafenib combination
(28%T/C), and the
anti-tumor activity of both combinations was significantly different when
compared to dabrafenib
treatment (p=0.0037 for dabrafenib+trametinib vs dabrafenib; p=0.03 for
dabrafenib+Compound
A vs dabrafenib), but not when compared to trametinib or Compound A
treatments. In
comparison, the triple combination of Compound A+dabrafenib+trametinib
achieved significant
(p<0.05) anti-tumor activity (3%T/C) when compared to the vehicle, all single
agents, and each
double combination groups (Table 1 and Figure 1). All treatment groups were
well tolerated with
minimal body weight loss (less than 10%) at the end of the two week study; no
signs of toxicity or
mortality were observed (Figure 1).
[00169] In the second study, the activity of the triple combination of
dabrafenib +
trametinib + Compound A was compared to the triple combination of dabrafenib +
Compound A
+ Compound C. Mice were treated with doses and regimens described in Table 2
until vehicle-
treated tumors achieved a volume >1000 mm3, 24 days post treatment initiation.
Anti-tumor
.. activity was determined by assessing %T/C or % regression on day 52 post
implant (24 days post
treatment initiation). Anti-tumor activity, mean change in tumor volume, mean
change in body
weight and survival 24 days post treatment initiation is reported in Table 4.
Table 4
Lays
Mgt" iiii,11111121211,i&mpbnt2211110iii "IMMO:SEMI"
46

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
iiiiirggiMgiMMMMEMMEiNiEMEZNITMEiNiiiiiiiiiiiiiiiOiiiiiiiiiiMilMTITIMgiMiMgITTq
i
.......................................................-
.........................................--
...............................................................................
........................
1073.95 1
Vehicle 39 13 -3.21 1.38
9/9
146.00
Compound A 75 qd 39 13 577.31 75.78 -5.61
1.58 9/9
trametinib 0.3 qd 39 13 511.85 68.46 -3.92
0.87 9/9
dabrafenib 30 qd 39 13 631.76 39.70 -4.88
1.25 9/9
7724/ Compound A 75 qd+
39 13 371.61 44.29 -5.95
0.91 9/9
HT29 +dabrafenib 30 qd
trametinib 0.3 qd+
39 13 302.45 39.97 -7.34
1.26 9/9
+dabrafenib 30 qd
Compound A + 75 qd+
trametinib+ 0.3 qd+ 39 13 29.47 17.55 -3.98
1.23 9/9
dabrafenib 30 qd 11
1
1157.33 1
Untreated control 52 24 5.57 2.91
4/7*
205.79
Compound A + 75 qd+ 166.09 33.35
8031/ trametinib+ 0.15 qd+ 52 24 0.45
0.93 7/7
HT29 dabrafenib 30 qd
Compound C + 25 bid+ 56.52 17.67
Compound A + 75 qd+ 52 24 -1.09 0.87
7/7
dabrafenib 30 qd ,
1 L
1685.22 f
7.65 1.10
Vehicle 38 26 238.87 5/5
trametinib+ 0.3 qd+ 288.00 55.77 1.74
0.90
38 26 5/5
8243/ dabrafenib 30 qd
HCOX trametinib+ 0.3 qd+ 222.98 96.84
2.83 1.07
1329 dabrafenib+ 30 qd+ 38 26 5/5
cetuximab 20biw
trametinib+ 0.15 qd+
-153.71 20.61 5.58 2.46
dabrafenib+ 30 qd+ 38 26 4/4
Compound A 75 qd ,
1 .
...............................................................................
...............................................................................
....................................................
*2 mice were sacrificed on day 49 and 1 mouse was sacrificed on day 45 due to
tumor
volume>1500mm3
47

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
[00170] Tumor volume and body weight change post treatment are plotted
on Figure 2.
The combined anti-tumor activity of Compound A+trametinib+dabrafenib (14% T/C)
was similar
and not-significantly different (p=0.38) when compared to the anti-tumor
activity achieved by
Compound A+Compound C+dabrafenib (5%T/C). The anti-tumor activity of both
combinations
was significantly improved when compared to the untreated control group
(p<0.0001 for both
groups against untreated control group) (Table 4 and Figure 2). All treatment
groups were well
tolerated with minimal body weight loss (less than 10%) at the end of the
study; no signs of
toxicity or mortality were observed (Figure 2).
[00171] Next, the antitumor efficacy of MAPK pathway inhibitors was
examined in the
BRAF mutant patient-derived CRC xenograft model HC0X1329 in athymic nude mice.
Mice
were treated with vehicle or combinations of MAPK pathway inhibitors as
described in Table 3
until vehicle-treated tumors achieved a volume >1000 mm3, or 62-67 days post
MAPK pathway
inhibitor treatment initiation. Anti-tumor activity was determined by
assessing %T/C or %
regression on day 38 post implant (26 days post treatment initiation), at
which point mice treated
with vehicle were sacrificed and mice treated with MAPK pathway inhibitors
were treated for an
additional 24-29 days and were sacrificed on day 62 (dabrafenib+Compound
A+trametinib) or
day 67 (dabrafenib+trametinib and dabrafenib+trametinib+cetuximab) post
implant. Anti-tumor
activity, mean change in tumor volume, mean change in body weight and survival
26 days post
treatment initiation is reported in Table 4. Tumor volume and body weight
change post 26-55
days of treatment are plotted on Figure 3. The combined anti-tumor activity of

dabrafenib+trametinib (17% T/C) was similar and not statistically significant
(p=0.68) when
compared to the anti-tumor activity achieved by
trametinib+dabrafenib+cetuximab (13%T/C). In
comparison, the triple combination of Compound A+dabrafenib+trametinib
achieved tumor
regression (70% Reg), which was significantly different compared to both the
dabrafenib+trametinib (p=0.005) or the dabrafenib+trametinib+cetuximab
combination (p=0.04)
(Table 4 and Figure 3). All treatment groups were well tolerated with minimal
body weight loss
(less than 10%) at the end of the two week study; no signs of toxicity or
mortality were observed
(Figure 3).
[00172] The in vivo activity of MAPK pathway inhibitor combinations was
profiled in
BRAF mutant CRC tumor xenografts. In the human BRAF mutant cell line derived
xenograft
48

CA 03173356 2022-08-26
WO 2021/171260
PCT/IB2021/051641
HT29, the combined activity of dabrafenib+trametinib was similar to that of
dabrafenib +
Compound A, and modestly better when compared to each single agent. In
comparison, the triple
combinations of dabrafenib+Compound A+trametinib or dabrafenib + Compound A +
Compound
C achieved significant anti-tumor activity in this xenograft model. In the
human patient derived
BRAF mutant CRC xenograft HC0X1329, the combination of dabrafenib +Compound
A+trametinib was also significantly more active than the combination of
dabrafenib+trametinib
and dabrafenib+trametinib+cetuximab. Collectively, these data indicate that
the triple
combinations of dabrafenib+Compound A+trametinib or dabrafenib+Compound
A+Compound C
can achieve greater and more durable responses in BRAF mutant CRC patients.
[00173] It is understood that the Examples and embodiments described
herein are for
illustrative purposes only and that various modifications or changes in light
thereof will be
suggested to persons skilled in the art and are to be included within the
spirit and purview of
this application and scope of the appended claims.
49

Representative Drawing

Sorry, the representative drawing for patent document number 3173356 was not found.

Administrative Status

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

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2021-02-26
(87) PCT Publication Date 2021-09-02
(85) National Entry 2022-08-26

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2023-01-18


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-02-26 $50.00
Next Payment if standard fee 2024-02-26 $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
Registration of a document - section 124 2022-08-26 $100.00 2022-08-26
Application Fee 2022-08-26 $407.18 2022-08-26
Maintenance Fee - Application - New Act 2 2023-02-27 $100.00 2023-01-18
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
NOVARTIS AG
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) 
Abstract 2022-08-26 1 61
Claims 2022-08-26 3 110
Drawings 2022-08-26 3 77
Description 2022-08-26 49 2,462
International Preliminary Report Received 2022-08-26 8 283
International Search Report 2022-08-26 4 111
Declaration 2022-08-26 2 30
National Entry Request 2022-08-26 9 590
Amendment 2022-10-24 10 441
Acknowledgement of National Entry Correction 2022-12-19 4 354
Cover Page 2023-02-01 1 31
Description 2022-10-24 49 3,524
Claims 2022-10-24 1 57

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.

BSL Files

To view selected files, please enter reCAPTCHA code :