Language selection

Search

Patent 3140231 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: (11) CA 3140231
(54) English Title: 3-ARYLOXYL-3-FIVE-MEMBERED HETEROARYL-PROPYLAMINE COMPOUND, AND CRYSTAL FORM AND USE THEREOF
(54) French Title: COMPOSE PROPYLAMINE HETEROARYLE 3-ARYLOXYL-3 A CINQ CHAINONS, FORME CRISTALLINE ET UTILISATION ASSOCIEE
Status: Granted and Issued
Bibliographic Data
(51) International Patent Classification (IPC):
  • C7D 409/12 (2006.01)
  • A61K 31/381 (2006.01)
  • A61K 31/397 (2006.01)
  • A61K 31/4025 (2006.01)
  • A61K 31/4525 (2006.01)
  • A61K 31/496 (2006.01)
  • A61K 31/5377 (2006.01)
  • A61P 1/00 (2006.01)
  • A61P 1/04 (2006.01)
  • A61P 11/00 (2006.01)
  • A61P 11/06 (2006.01)
  • A61P 13/02 (2006.01)
  • A61P 17/04 (2006.01)
  • A61P 29/00 (2006.01)
  • C7D 343/00 (2006.01)
  • C7D 407/12 (2006.01)
(72) Inventors :
  • WANG, YOUXIN (China)
  • ZHANG, LINGLING (China)
  • DING, QIANG (China)
(73) Owners :
  • SHANGHAI LEADO PHARMATECH CO. LTD.
(71) Applicants :
  • SHANGHAI LEADO PHARMATECH CO. LTD. (China)
(74) Agent: BENOIT & COTE INC.
(74) Associate agent:
(45) Issued: 2024-06-04
(86) PCT Filing Date: 2020-05-14
(87) Open to Public Inspection: 2020-11-19
Examination requested: 2021-11-12
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/CN2020/090354
(87) International Publication Number: CN2020090354
(85) National Entry: 2021-11-12

(30) Application Priority Data:
Application No. Country/Territory Date
201910411311.X (China) 2019-05-16
202010093736.3 (China) 2020-02-14
PCT/CN2019/100846 (China) 2019-08-15

Abstracts

English Abstract

The present invention relates to a 3-aryloxyl-3-five-membered heteroaryl propylamine compound, and crystal form and use thereof. Specifically, the present invention provides a compound, or a pharmaceutically acceptable salt or a prodrug thereof, the compound having a structure of formula I. The compound, or pharmaceutically acceptable salt or prodrug thereof of the present invention has an excellent inhibition function for a transient receptor potential channel protein (TPR), and has a good treatment function for diseases associated with the TPR.


French Abstract

La présente invention concerne un composé propylamine hétéroaryle 3-aryloxyl-3 à cinq chaînons, une forme cristalline et une utilisation associée. Plus particulièrement, la présente invention concerne un composé, ou un sel pharmaceutiquement acceptable ou un promédicament de celui-ci, le composé ayant une structure de formule I. Le composé, ou un sel pharmaceutiquement acceptable ou un promédicament de celui-ci selon la présente invention a une excellente fonction d'inhibition pour une protéine de canal à potentiel de récepteur transitoire (TPR), et a une bonne fonction de traitement pour des maladies associées au TPR.

Claims

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


Claims
1. A compound, or a phafinaceutically acceptable salt thereof, wherein the
compound
has a structure of Formula Z:
ti(R3 ) _______________________________
I /
R1
0
R2
wherein:
ring A is a substituted or unsubstituted furan ring;
R1 and R2 are each independently hydrogen, substituted or unsubstituted C1-C6
alkyl,
or substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, or
substituted or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
"*" represents that the configuration of the compound is racemic;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms on the
group are
each independently substituted by a substituent selected from the group
consisting of
Ci-C6 alkyl, C3-C7 cycloalkyl, C1 -C3 haloalkyl, halogen, nitro, cyano,
hydroxyl, Ci-C4
carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Ci-C6 haloalkoxy, benzyl, six-
membered
aryl, and five- or six-membered heteroaryl; and
wherein, the heteroaryl have 1 to 3 heteroatoms selected from N, 0 and S.
2. The compound of claim 1, wherein the compound is selected from the group
consisting of:
C\S
0
ON` 0 NH2 141'= 0
0 A
o o o
\ \ W W
s
0s 0 0 Ikr
and
H N'
0
0 0 /
0
=
3. A compound, or a pharmaceutically acceptable salt thereof, wherein the
compound
has a structure of formula I:
¨ 55 ¨
Date Reeue/Date Received 2023-09-21

n(113)--fJX\
,R1
0
R2
01.1
wherein:
ring A is a substituted or unsubstituted furan ring;
R1 and R2 are each independently hydrogen, substituted or unsubstituted C1-C6
alkyl,
or substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, or
substituted or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms on the
group are
each independently substituted by a substituent selected from the group
consisting of:
Ci-C6 alkyl, C3-C7 cycloalkyl, Ci-C3 haloalkyl, halogen, nitro, cyano,
hydroxyl, C1-C4
carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Ci-C6haloalkoxy, benzyl, six-
membered
aryl, and five- or six-membered heteroaryl; and
wherein, the heteroaryl have 1 to 3 heteroatoms selected from N, 0 and S.
4. The compound of claim 3, wherein the compound is selected from the group
consisting of:
Cs
-N -NH2 0 N'"'
0 0 0 0 0
1-2 1-3 1-4
1-5
CI
C(S
R)
_AR) 0 Pr
C) 0 Nctx and
H 0
0 0
0
ll-6 1-7 1-8 I-11.
5. A hydrochloride of the compound of formula I-1, or a crystal form A
thereof,
¨ 56¨
Date Revue/Date Received 2023-09-21

(R)
I-1
the X-ray powder diffraction pattern of the crystal form A of the
hydrochloride of the
compound of formula I-1 has characteristic peaks at 20 angles of 18.173 0.2 ,
22.084 0.2 , and 22.794 0.2 .
6. The crystal form A of the hydrochloride of the compound of formula I-1
according
to claim 5, wherein the crystal form A of the hydrochloride of the compound of
formula
I-1 further has characteristic peaks at one or more of 20 value selected from
the group
consisting of: 10.003 0.2 , 11.171 0.2 , 15.987 0.2 , 16.734 0.2 , 17.092 0.2
,
18.849 0.2 , 20.681 0.2 , 21.156 0.2 , 21.649 0.2 , 23.761 0.2 , 25.298 0.2 ,
25.967 0.2 , 26.640+0.2 , 27.273 0.2 , 28.099 0.2 , 28.615+0.2 , 28.813 0.2 ,
29.501 0.2 , 30.118 0.2 , 30.513 0.2 , 32.522 0.2 , 33.274 0.2 , 34.081 0.2 ,
35.815 0.2 , 37.553 0.2 , 40.018 0.2 , 42.927 0.2 , and 44.129 0.2.
7. The crystal form A of the hydrochloride of the compound of Formula I-1
according
to claim 5, wherein the X-ray powder diffraction pattern of the crystal form A
of the
hydrochloride has characteristic peaks and peak intensity at 20va1ue selected
from the
group consisting of:
relative
20va1ue d value
intensity %
10.003 8.8355 28.3
11.171 7.9137 12.8
15.987 5.5392 15.2
16.734 5.2936 51.9
17.092 5.1836 35.7
18.173 4.8774 100.0
18.849 4.7041 12.7
20.681 4.2913 13.6
21.156 4.1961 65.2
21.649 . 4.1017 35.3
22.084 4.0217 71.9
22.794 3.8981 91.8
23.761 3.7416 65.0
25.298 . 3.5177 46.3
25.967 3.4285 11.5
26.640 , 3.3433 29.2
27.273 3.2672 16.9
28.099 3.1730 35.7
28.615 3.1170 33.1
28.813 , 3.0959 25.2
29.501 3.0253 10.4
30.118 . 2.9647 12.1
30.513 2.9272 13.2
- 57 -
Date Revue/Date Received 2023-09-21

32.522 2.7508 18.4
33.274 2.6904 12.4
34.081 2.6285 13.2
35.815 2.5051 13.4
37.553 2.3931 9.6
40.018 2.2512 8.2
42.927 2.1051 10.6
and 44.129 2.0505 9.0 .
8. The crystal form A of thse hydrochloride of the compound of formula 1-1
according to
claim 5, wherein the crystal form A of the hydrochloride of the compound of
formula I-1
comprises one or more features selected from the group consisting of:
(i) the crystal form A of the hydrochloride of the compound of formula I-1 has
X-ray
powder diffraction characteristic peaks substantially as shown in Fig. 7;
(ii) the differential scanning calorimetry (DSC) pattern of the crystal form A
of the
hydrochloride of the compound of formula I-1 begins to appear endothermic
peaks upon
being heated to 142.30 C 1 C;
(iii) the differential scanning calorimetry (DSC) pattern of the crystal form
A of the
hydrochloride of the compound of formula I-1 is substantially as shown in Fig.
8;
(iv) the theimogravimetric analysis (TGA) pattern of the crystal form A of the
hydrochloride of the compound of formula I-1 has a weight loss of 0.9827% 0.5%
upon
being heated to 168.01 C; and
(v) the thermogravimetric analysis (TGA) pattern of the crystal form A of the
hydrochloride is substantially as shown in Fig. 9.
9. Use of the compound of formula Z according to claim 1, or the
pharmaceutically
acceptable salt thereof, or the compound of formula 1 according to claim 3, or
the
pharmaceutically acceptable salt thereof, or the crystal form A of the
hydrochloride of the
compound of formula I-1 according to claim 5 for preparing a transient
receptor potential
channel protein (TRPA1) inhibitor.
10. Use of the compound of formula Z according to claim 1, or the
pharmaceutically
acceptable salt thereof, or the compound of formula I according to claim 3, or
the
pharmaceutically acceptable salt thereof, or the crystal form A of the
hydrochloride of the
compound of formula I-1 according to claim 5 for manufacturing a medicament
for
preventing or treating diseases related to transient receptor potential
channel protein
(TRPA1).
11. The use according to claim 10, wherein the diseases related to transient
receptor
potential channel protein are selected from the group consisting of pain,
epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder,
inflammatory bowel
disease, and combinations thereof.
12. The use according to claim 11, wherein the pain is selected from the group
consisting of acute pain, inflammatory pain, visceral pain, neurogenic pain,
fibromyalgia,
headache, nerve pain, mixed pain, cancer-induced pain, and combinations
thereof.
13. The use according to claim 12, wherein the acute pain is injury pain or
postoperative pain; and
the inflammatory pain is osteoarthritis pain or rheumatoid arthritis pain.
14. A pharmaceutical composition, wherein the pharmaceutical composition
comprises the compound of formula Z according to claim 1, or the
pharmaceutically
acceptable salt thereof, or the compound of formula I according to claim 3, or
the
pharmaceutically acceptable salt thereof, or the crystal form A of the
hydrochloride of the
compound of formula I-1 according to claim 5; and a pharmaceutically
acceptable carrier.
¨ 58 ¨
Date Revue/Date Received 2023-09-21

Description

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


CA 03140231 2021-11-12
3-ARYL OXY-3 -FIVE-ME MBERED HE TEROARYL-PROPYLAMINE
COMPOUND, AND CRYSTAL FORM AND USE THEREOF
FIELD OF THE INVENTION
The invention relates to the field of pharmacochemistry and
pharmacotherapeutics,
in particular to a 3-aryloxy-3-five-membered heteroaryl-propylamine compounds
and
crystal form and use thereof.
BACKGROUND OF THE INVENTION
Pain is known as the fifth vital sign and is an alarming sign of damage to the
body's
tissues. Pain is one of the most common reasons for patients to seek medical
treatment.
According to its duration, pain can be divided into acute pain (acute onset,
short duration
or continuous state) and chronic pain (slow onset or transformed from acute
pain, long
duration, or intermittent onset, and many chronic pains can not find obvious
damage).
Acute pain is mostly nociceptive pain caused by tissue trauma, including
postoperative
pain, trauma, post-burn pain, childbirth pain, visceral pain such as angina
pectoris, biliary
colic and renal colic, etc, fracture pain, toothache and cancer pain.
Postoperative and
post-traumatic pain are the most common and urgent acute pain in clinic.
Chronic pain
mainly includes neuropathic pain, painful osteoarthritis, chronic back and low
back pain
and vascular pain, etc. Main types of neuropathic pain includes trigeminal
neuralgia,
diabetic pain, sciatica or postherpetic neuralgia. The global prevalence rate
of neuropathic
pain is about 10%, with a high incidence and a large number of patients.
Chronic pain
affects 10%-30% of the population in the United States, causing about $635
billion in
annual social spending, more than the combination of cancer and heart disease.
Chronic
pain has complex etiology and is a refractory disease. Only less than 50% of
patients can
achieve effective analgesia through drug treatment. It is estimated that the
total market
size of neuralgia drugs in China will be close to 26 billion yuan in 2026, and
the market
size of ion channel neuralgia drugs will exceed 20 billion yuan.
Traditional analgesics mainly include opioids and non-steroidal anti-
inflammatory
drugs. Opioids have strong analgesic effect, but long-term use can easily lead
to tolerance,
dependence and addiction, and have adverse reactions such as respiratory
depression and
central sedation. Non-steroidal anti-inflammatory drugs have only moderate
analgesic
effect, and have adverse reactions such as gastrointestinal bleeding and
cardiotoxicity.
Recently, the National Security Council of the United States released a report
on
preventable deaths, which showes that for the first time in American history,
the
proportion of deaths caused by opioid overdose exceeded that caused by car
accidents.
According to the commission's analysis of data on accidental deaths in 2017,
one in 96
Americans died from an opioid overdose, compared with one in 103 deaths from
car
accidents. Opioid abuse has caused a serious social crisis sweeping across the
United
States, so the market needs analgesics having new mechanisms.
TRPA1 is a member of the TRP ion channel superfamily and the only member of
the
TRPA subfamily. TRPA1 is a non-selective cation channel and can permeate Na,
K+, Ca2
+ and Mg'. TRPA1 is mainly distributed in the primary sensory neurons of
dorsal root
nerve (DRG), trigeminal nerve (TG) and vagus nerve (VG). From the distribution
of
human system, TRPA1 is highly expressed in peripheral nervous system,
respiratory
system, gastrointestinal system and urinary system. When these organs and
tissues are
dysfunctional, the expression and function of TRPA1 channel are usually
abnomtal
simultaneously. TRPA1 can transform cold stimulation, chemical stimulation and
mechanical stimulation into inward current, which triggers a series of
physiological
functions and participates in the formation of various pain senses.
Inflammatory pain is a
common problem of some chronic diseases, and still lack of effective treatment
in clinic.
¨1 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
Animal studies have shown that TRPA1 participates in inflammatory reaction and
plays an
important role in inflammatory pain. By using TRPA1 specific blocker,
inflammatory pain
reaction in rats can obviously be alleviated. From the current research, TRPA1
plays an
important role in the occurrence of asthma and cough, and the compounds that
induce
asthma and cough, whether endogenous or exogenous, can activate TRPA1. TRPA1
antagonists can alleviate asthma symptoms and block airway
hyperresponsiveness.
Through different animal models of visceral hypersensitivity, such as colitis,
colorectal
dilatation or stress, it is confirmed that TRPA1 is involved in the regulation
of visceral
hypersensitivity and plays an important role in visceral pain. Neurogenic pain
is a pain
syndrome caused by central or peripheral nervous system damage or disease,
which is
mainly manifested as hyperalgesia, abnormal hyperalgesia and spontaneous pain.
In recent
years, more and more studies have shown that TRPA1 channel plays an important
role in
different neurogenic pain, such as diabetic neuropathy and neuropathy caused
by
chemotherapy drugs. Recent studies have also shown that TRPA1 also plays a
mediating
role in toothache, migraine and other pain, and TRPA1 antagonist can obviously
relieve
the occurrence of pain symptoms.
TRPA1 is widely distributed and expressed in human system. In addition to the
above physiological functions involved by TRPA1, the reported indications for
TRPA1
inhibitors also involve inflammatory bowel disease, chronic obstructive
pulmonary disease,
antitussive, antipruritic, allergic rhinitis, ear diseases, anti-diabetes,
urinary incontinence
and so on. TRPA1 is a proven new target for the treatment of many diseases.
Therefore, considering that pain treatment is an unmet clinical demand at
present,
and many problems existing in existing therapeutic drugs, it is urgent to
develop a
therapeutic drug for TRP targets (especially TRPA1 targets) in the field, so
as to improve
the therapeutic effect of diseases.
SUMMARY OF THE INVENTION
The purpose of the present invention is to provide a compound with a novel
structure
that targets the TRP channel (especially the TRPA1 target), and crystal form
and use
thereof.
In the first aspect of the present invention, it provides a use of a compound,
or a
pharmaceutically acceptable salt thereof, or a prodrug thereof, for (a)
preparing a transient
receptor potential channel protein (TRPA1) inhibitor; (b) manufacturing a
medicament for
preventing and/or treating diseases related to transient receptor potential
channel protein
(TRPA1);
wherein the compound has a structure of formula Z:
r--X
n(R3 )
N
R2
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
¨ 2
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
IV and R2 are each independently hydrogen, substituted or unsubstituted Ci-Co
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
"*" represents that the configuration of the compound is racemic;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: Ci-C6 alkyl, C3-C7 cycloalkyl, Ci-C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, Cl-C6 alkoxy, Ci-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a C5
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently
have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N, 0 and S.
In another preferred embodiment, the compound of formula Z has a structure of
formula Z-1:
,S1(3)
0
R2
A el
Z-1
In another preferred embodiment, ring A is a substituted or unsubstituted 5-7
membered carbocyclic ring and a 5-7 membered heteroaromatic ring.
In another preferred embodiment, X is S or 0.
In another preferred embodiment, R' and R2 are each independently hydrogen or
substituted or unsubstituted Ci-C3 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted C1-C6 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted C i-Ca alkyl.
In another preferred embodiment, the halogen is F, Cl, Br or I.
In another preferred embodiment, when n>2, each R3 is the same or different.
In another preferred embodiment, ring A is a substituted or unsubstituted
5-membered carbocyclic ring, a substituted or unsubstituted 6-membered
carbocyclic ring,
or a substituted or unsubstituted furan ring.
In another preferred embodiment, ring A is not a benzene ring.
In another preferred embodiment, ring A is
0
\ , or
-3 -
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
=
3
nat ) /
In another preferred embodiment, the structure of is:
I .
In another preferred embodiment, le and R2 are each independently hydrogen,
methyl
or ethyl.
In another preferred embodiment, It3 is a hydrogen atom, a chlorine atom or a
methyl.
In another preferred embodiment, n is 1.
In another preferred embodiment, A is a 5-membered carbocyclic ring, a 6-
membered
carbocyclic ring or a furan ring.
In another preferred embodiment, n is 1, 2 or 3.
In another preferred embodiment, X is S.
In another preferred embodiment, ring A is a furan ring.
In another preferred embodiment, the ring containing X is a thiophene ring.
s
In another preferred embodiment, the structure of the thiophene ring is ¨ .
In another preferred embodiment, the structure of the ring A acene ring is
aulvv
1\(,)
In the present invention, " unArtf" is the linking site of the groups.
In another preferred embodiment, the compound of formula Z is selected from
the
group consisting of
0
0 NH 2
N
0 0 0 0 0
¨4¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
C I
ON-
H
H 0
o
0 0
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRPA1) is selected from the group consisting of pain,
epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder,
inflammatory bowel
disease, and combinations thereof. In another preferred embodiment, the pain
is selected
from the group consisting of acute pain, inflammatory pain, visceral pain,
neurogenic pain,
fibromyalgia, headache, nerve pain, mixed pain, cancer-induced pain, and
combinations
thereof.
In another preferred embodiment, the pain is postoperative pain.
In another preferred embodiment, the postoperative pain is postoperative pain
following a surgical procedure.
In another preferred embodiment, the postoperative pain is post-operative
wound
pain.
In another preferred embodiment, the post-operative wound pain is selected
from the
group consisting of post-operative skin wound pain, post-operative muscle
wound pain,
and combinations thereof.
In another preferred embodiment, the post-operative wound pain is skin and
muscle
post-operative wound pain.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In another preferred embodiment, the inflammatory pain is chronic inflammatory
pain.
In another preferred embodiment, the inflammatory pain is osteoarthritis pain
or
rheumatoid arthritis pain.
In another preferred embodiment, the headache is migraine or muscular tension
pain.
In another preferred embodiment, the neuralgia is trigeminal neuralgia,
diabetic pain,
sciatica, or postherpetic neuralgia.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In the second aspect of the present invention, it provides a use of a
compound, or a
pharmaceutically acceptable salt thereof, or a prodrug thereof, for (a)
preparing a transient
- 5 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
receptor potential channel protein (TRP) inhibitor; (b) manufacturing a
medicament for
preventing and/or treating diseases related to transient receptor potential
channel protein
(TRP);
wherein, the compound has the structure of follinula I:
n(R3)
I
Ale) R1
R2
A,
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
R' and R2 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: Ci-C6 alkyl, C3-C7 cycloalkyl, CI -C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Ci-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a Cs
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently
have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N, 0 and S.
In another preferred embodiment, the compound of formula I has a structure of
formula I-1:
11(R3)--õ1õ,_ _________________________
(R)
111
o
I ,
12-
I- 1.
In another preferred embodiment, ring A is a substituted or unsubstituted 5-7
- 6
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
membered carbocyclic ring and a 5-7 membered heteroaromatic ring.
In another preferred embodiment, X is S or 0.
In another preferred embodiment, R1 and R2 are each independently hydrogen or
substituted or unsubstituted C i-C3 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted Ci-C6 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted C i-C4 alkyl.
In another preferred embodiment, the halogen is F, Cl, Br or I.
In another preferred embodiment, when n>2, each R3 is the same or different.
In another preferred embodiment, ring A is a substituted or unsubstituted
5-membered carbocyclic ring, a substituted or unsubstituted 6-membered
carbocyclic ring,
or a substituted or unsubstituted furan ring.
In another preferred embodiment, ring A is not a benzene ring.
In another preferred embodiment, ring A is
0
F1( \ , or
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
%AWN
~Inn,
0140.
n(R3) ________________________________________________
1/
In another preferred embodiment, the structure of , is:
(R3)õ
X
I .
In another preferred embodiment, IV and R2 are each independently hydrogen,
methyl
or ethyl.
In another preferred embodiment, R3 is a hydrogen atom, a chlorine atom or a
methyl.
In another preferred embodiment, n is 1.
In another preferred embodiment, A is a 5-membered carbocyclic ring, a 6-
membered
carbocyclic ring or a furan ring.
In another preferred embodiment, n is 1, 2 or 3.
In another preferred embodiment, X is S.
In another preferred embodiment, ring A is a furan ring.
In another preferred embodiment, the ring containing X is a thiophene ring.
In another preferred embodiment, the structure of the thiophene ring is
- 7
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, the structure of the ring A acene ring is
vutiv
0 Oln
In another preferred embodiment, the compound is selected from the group
consisting
of:
0 1=1'NH2 ()
0 0 0 I 0 0
\
I-1 1-2 1-3 1-4
1-5
ci
FZ)
0
H 0
0 0
1-8 1-9 I-10
1-6 1-7
o
I-11.
In another preferred embodiment, the transient receptor potential channel
protein
(TRP) is TRPAL
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRP) is selected from the group consisting of pain, epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder,
inflammatory bowel
disease, and combinations thereof.
In another preferred embodiment, the pain is selected from the group
consisting of
acute pain, inflammatory pain, visceral pain, neurogenic pain, muscle fiber
pain, headache,
nerve pain, mixed pain, cancer-induced pain, and combinations thereof.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In another preferred embodiment, the postoperative pain is postoperative pain
following a surgical procedure.
In another preferred embodiment, the postoperative pain is post-operative
wound
pain.
In another preferred embodiment, the post-operative wound pain is selected
from the
group consisting of post-operative skin wound pain, post-operative muscle
wound pain,
and combinations thereof.
In another preferred embodiment, the post-operative wound pain is skin and
muscle
post-operative wound pain.
In another preferred embodiment, the inflammatory pain is chronic inflammatory
¨ 8
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
pain.
In another preferred embodiment, the inflammatory pain is osteoarthritis pain
or
rheumatoid arthritis pain.
In another preferred embodiment, the headache is migraine or muscular tension
pain.
In another preferred embodiment, the neuralgia is trigeminal neuralgia,
diabetic pain,
sciatica, or postherpetic neuralgia.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In the third aspect of the present invention, it provides a compound, or a
pharmaceutically acceptable salt thereof, or a prodrug thereof, wherein the
compound has
a structure of formula Z:
r--X
n(R3 ) Li
'co)
N
R2
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
R1 and R2 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
R3 is hydrogen, halogen, substituted or unsubstituted C1-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
"*" represents that the configuration of the compound is racemic;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: Ci-C6 alkyl, C3-C7 cycloalkyl, Ci -C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Ci-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a Cs
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently
have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N, 0 and S.
In another preferred embodiment, the pharmaceutically acceptable salt of the
compound of formula Z is a salt formed by the compound of formula Z and an
acid
selected from the group consisting of hydrochloric acid, mucic acid, D-
glucuronic acid,
hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric
acid, formic
acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid,
fumaric acid,
maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid,
methanesulfonic
acid, benzyl sulfonic acid, benzenesulfonic acid, aspartic acid, glutamic
acid, or
combinations thereof.
In another preferred embodiment, the compound of formula Z has a structure of
formula Z-1:
¨ 9 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
n(R3
iks=,N\r, X
0
A
R2
I.
Z-1
In another preferred embodiment, ring A is a substituted or unsubstituted 5-7
membered carbocyclic ring and a 5-7 membered heteroaromatic ring.
In another preferred embodiment, X is S or 0.
In another preferred embodiment, R1 and R2 are each independently hydrogen or
substituted or unsubstituted CI-C3 alkyl.
In another preferred embodiment, 113 is hydrogen, halogen, substituted or
unsubstituted Ci-C6 alkyl.
In another preferred embodiment, 123 is hydrogen, halogen, substituted or
unsubstituted CI-Ca alkyl.
In another preferred embodiment, the halogen is F, Cl, Br or I.
In another preferred embodiment, when n>2, each R3 is the same or different.
In another preferred embodiment, ring A is a substituted or unsubstituted
5-membered carbocyclic ring, a substituted or unsubstituted 6-membered
carbocyclic ring,
or a substituted or unsubstituted furan ring.
In another preferred embodiment, ring A is not a benzene ring.
In another preferred embodiment, ring A is
0
Prrr< \ , or
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
In another preferred embodiment, the structure of I is:
(R3)11 _____________________________________
X
I .
In another preferred embodiment, RI and R2 are each independently hydrogen,
methyl or ethyl.
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, IV is a hydrogen atom, a chlorine atom or a
methyl.
In another preferred embodiment, n is 1.
In another preferred embodiment, A is a 5-membered carbocyclic ring, a 6-
membered
carbocyclic ring or a furan ring.
In another preferred embodiment, n is 1, 2 or 3.
In another preferred embodiment, X is S.
In another preferred embodiment, ring A is a furan ring.
In another preferred embodiment, the ring containing X is a thiophene ring.
In another preferred embodiment, the structure of the thiophene ring is ¨ .
In another preferred embodiment, the structure of the ring A acene ring is
010
In the present invention, " aVVV" is the linking site of the groups.
In another preferred embodiment, the compound is selected from the group
consisting
of:
ON 0 NH
0 0 0 0 0
CI
*r
Ot'N'
H 0
0 0
0
In the fourth aspect of the present invention, it provides a compound, or a
¨11 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
pharmaceutically acceptable salt thereof, or a prodrug thereof, the compound
has a
structure of formula I:
n(R3
A01.!! Ri
-`,-"" N
R2
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
IV and R2 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
IV is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: C1-C6 alkyl, C3-C7 cycloalkyl, CI -C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, CI-C6 alkoxy, Ci-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a Cs
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently
have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N, 0 and S.
In another preferred embodiment, the pharmaceutically acceptable salt of the
compound of formula I is a salt foliated by the compound of formula I and an
acid selected
from the group consisting of hydrochloric acid, mucic acid, D-glucuronic acid,
hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric
acid, formic
acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid,
fumaric acid,
maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid,
methanesulfonic
acid, benzyl sulfonic acid, benzenesulfonic acid, aspartic acid, glutamic
acid, or
combinations thereof.
In another preferred embodiment, the compound of foimula I has a structure of
formula I-1:
ii(1z3) ______________________________
(\z,x
(R)
0
R2
1- 1,
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, A is a substituted or unsubstituted 5-7
membered
carbocyclic ring and a 5-7 membered heteroaromatic ring.
In another preferred embodiment, X is S or 0.
In another preferred embodiment, R1 and R2 are each independently hydrogen or
substituted or unsubstituted C i-C3 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted C -C 6 alkyl.
In another preferred embodiment, R3 is hydrogen, halogen, substituted or
unsubstituted Ci-C4 alkyl.
In another preferred embodiment, the halogen is F, Cl, Br or I.
In another preferred embodiment, when n>2, each R3 is the same or different.
In another preferred embodiment, A is a substituted or unsubstituted 5-
membered
carbocyclic ring, a substituted or unsubstituted 6-membered carbocyclic ring,
or a
substituted or unsubstituted furan ring.
In another preferred embodiment, ring A is not a benzene ring.
In another preferred embodiment, ring A is
0
risrr< \ , or
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
=
¨x
not3 r-
I
In another preferred embodiment, the structure of I is:
(R3).
X
I .
In another preferred embodiment, R1 and R2 are each independently hydrogen,
methyl or ethyl.
In another preferred embodiment, R3 is a hydrogen atom, a chlorine atom or a
methyl.
In another preferred embodiment, n is 1.
In another preferred embodiment, A is a 5-membered carbocyclic ring, a 6-
membered
carbocyclic ring or a furan ring;
In another preferred embodiment, n is 1, 2 or 3.
In another preferred embodiment, X is S.
In another preferred embodiment, ring A is a furan ring.
In another preferred embodiment, the ring containing X is a thiophene ring.
- 13 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
9
In another preferred embodiment, the structure of the thiophene ring is 5 .
In another preferred embodiment, the structure of the ring A acene ring is
\C:
In the present invention, " %/VW" is the linking site of the groups.
In another preferred embodiment, the compound is selected from the group
consisting
of:
(7)
0
0 illf-Q-NH2 C)N
0 0 0 I 0
\ 0
I-1 1-2 1-3 1-4
1-5
ci
R)
O Ofe 0
H 0
0 0
1-8 1-9 I-10
1-6 1-7
o
I-11.
In the fifth aspect of the present invention, it provides a compound of
formula A, or a
pharmaceutically acceptable salt thereof, or a prodrug thereof,
(R3) ______________________________ I/ ,
9
12_1
R7 R8 1.2
R4
A
R5
R6
A
¨ 14 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
R' and R2 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
W is 0 or S;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3 -C7 cycloalkyl;
R4, R5, R6, R7, R8, R9 and R19 are each independently hydrogen, substituted or
unsubstituted Ci-C6 alkyl, substituted or unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
"*" represents that the configuration of the compound is racemic;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: Ci-C6 alkyl, C3-C7 cycloalkyl, Ci -C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Ci-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a C5
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N,
0 and S.
In another preferred embodiment, the pharmaceutically acceptable salt of the
compound of formula A is a salt formed by the compound of formula A and an
acid
selected from the group consisting of hydrochloric acid, mucic acid, D-
glucuronic acid,
hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric
acid, formic
acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid,
fumaric acid,
maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid,
methanesulfonic
acid, benzyl sulfonic acid, benzenesulfonic acid, aspartic acid, glutamic
acid, or
combinations thereof.
In another preferred embodiment, in the compound of formula A, ring A, 10, R2,
R3,
X, and n are each independently as described in the third aspect of the
present invention.
In another preferred embodiment, W is 0.
In another preferred embodiment, R4, R5, R6, R7, R8, R9 and R1 are each
independently hydrogen, substituted or unsubstituted CI-Ca alkyl, substituted
or
unsubstituted C3-C6 cycloalkyl.
In another preferred embodiment, R4, R5, R6, R7, R8, R9 and R1-9 are each
independently hydrogen.
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
NOWLIV
R4 R4 R4
R5 R5 R5
R6 R6 , Or R6
In another preferred embodiment, the compound of formula A is a compound of
formula Z:
- 15 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
n(R3 )
/
N
R2
In another preferred embodiment, the compound of Formula Z is as described in
the
third aspect of the present invention.
In the sixth aspect of the present invention, it provides a compound of
formula B, or
a pharmaceutically acceptable salt thereof, or a prodrug thereof,
n( R3 ) ____________________________
R9 io
(R)
NR1
R7 R8 ilz2
R4
Ck R5
R6
wherein:
ring A is a substituted or unsubstituted 5-7 membered carbocyclic ring, a
substituted
or unsubstituted 5-7 membered heterocyclic ring, a substituted or
unsubstituted 5-7
membered heteroaromatic ring;
R1 and R2 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl;
X is an oxygen atom, a sulfur atom or a nitrogen atom;
W is 0 or S;
R3 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
R4, R5, R6, R7, R8, R9 and R1 are each independently hydrogen, substituted or
unsubstituted CI-C6 alkyl, substituted or unsubstituted C3-C7 cycloalkyl;
n is 1, 2 or 3;
"*" represents that the configuration of the compound is racemic;
wherein, any one of the "substituted" means that 1-4 hydrogen atoms
(preferably 1, 2,
3, or 4) on the group are each independently substituted by a substituent
selected from the
group consisting of: Ci-C6 alkyl, C3-C7 cycloalkyl, CI -C3 haloalkyl, halogen,
nitro,
cyano, hydroxyl, CI-Ca carboxy, C2-C4 ester, C2-C4 amide, Ci-C6 alkoxy, Cl-C6
haloalkoxy, benzyl, six-membered aryl, five- or six-membered heteroaryl
(preferably a C5
heteroaryl);
wherein, the heterocyclic ring, heteroaromatic ring and heteroaryl each
independently have 1 to 3 (preferably 1, 2 or 3) heteroatoms selected from N,
0 and S.
¨ 16 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, the phannaceutically acceptable salt of the
compound of formula B is a salt formed by the compound of formula B and an
acid
selected from the group consisting of hydrochloric acid, mucic acid, D-
glucuronic acid,
hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric
acid, formic
acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid,
fumaric acid,
maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid,
methanesulfonic
acid, benzyl sulfonic acid, benzenesulfonic acid, aspartic acid, glutamic
acid, or
combinations thereof.
In another preferred embodiment, in the compound of formula A, ring A, R1, R2,
IV,
X, and n are each independently as described in the fourth aspect of the
present invention.
In another preferred embodiment, W is 0.
In another preferred embodiment, R4, R5, R6, 127, R8, R9 and R1 are each
independently hydrogen, substituted or unsubstituted C i-C4 alkyl, substituted
or
unsubstituted C3-C6 cycloalkyl.
In another preferred embodiment, R4, R5, R6, R7, R8, R9 and R1 are each
independently hydrogen.
In another preferred embodiment, the connection structure between ring A and
the
adjacent benzene ring is:
.111VilW
R4 R4
R5 R5 R5
R6 R6 , or R6
In another preferred embodiment, the compound of Formula B is a compound
having
a structural of Formula I:
41e) __________________________________
0).e.õ
R2
In another preferred embodiment, the compound of Formula I is as described in
the
fourth aspect of the present invention.
In the seventh aspect of the present invention, it provides a pharmaceutical
composition comprising the compound as described in the third aspect of the
present
invention, or the pharmaceutically acceptable salt thereof, or the prodrug
thereof, and/or
the compound as described in the fourth aspect of the present invention, or
the
pharmaceutically acceptable salt thereof, or the prodrug thereof; and
pharmaceutically
acceptable carriers.
In the eighth aspect of the present invention, it provides a pharmaceutical
composition comprising the compound as described in the fifth aspect of the
present
invention, or the pharmaceutically acceptable salt thereof, or the prodrug
thereof, and/or
the compound as described in the sixth aspect of the present invention, or the
pharmaceutically acceptable salt thereof, or the prodrug thereof; and
pharmaceutically
acceptable carriers.
- 17
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In the ninth aspect of the present invention, it provides a use of the
compound of
formula A as described in the fifth aspect of the present invention, or the
pharmaceutically
acceptable salt thereof, or the prodrug thereof, or the compound of formula B
as described
in the sixth aspect of the present invention, or the pharmaceutically
acceptable salt thereof,
or the prodrug thereof for (a) preparing a transient receptor potential
channel protein
(TRPA1) inhibitor; (b) manufacturing a medicament for preventing and/or
treating
diseases related to transient receptor potential channel protein (TRPA1). In
another
preferred embodiment, the transient receptor potential channel protein (TRP)
is TRPA1.
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRP) is as described in the second aspect of the present
invention.
In the tenth aspect of the present invention, it provides a method for
preparing the
compound as described in the fourth aspect of the present invention, or the
pharmaceutically acceptable salt thereof, or the prodrug thereof, wherein the
method
comprises the steps of: in an inert solvent, reacting intermediate II with the
R'-NH-R2
compound to form the compound:
n( n(R3)
U., 1
0 Ri R2 0 N-
-N-
ft2
CIO ________________________________________ )" A
11
wherein X, A, R1, R2, le and n are as described in the fourth aspect of the
present
invention.
In another preferred embodiment, the method comprises the steps of:
, x x
diisopropyl
121 R2 n(R3)__f1
OH 3X n(R ) azodicarboxylate sodium
(R)
j triphenylphosphine N-R1
R2HOCI Anhydrous acetone
Tetrahydro-
tetrahydrofuran C141 furan
.4, a)
N
2 2
5
.4 4 5
phthalimide
n(R )3)[
potassium salt 0
sodium iodide (R)
N
N,N-dimethylformamide
CIO
wherein X, A, le, R2, le and n are as described in the fourth aspect of the
present
invention:
OH
(a) reacting
compound with (S)-1-(11(R3)-five-membered
heteroary1)-3-chloro-propanol in the presence of a condensation agent in an
inert solvent
to form Intermediate II;
(b) performing any reaction selected from the follows to form compound I:
¨ 18 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
(b-1) reacting intermediate II with 10-NH-R2 in an inert solvent to form
compound I;
(b-2) in an inert solvent, the intermediate II is reacted with the potassium
salt of
phthalimide to form intermediate III, which undergoes hydrazinolysis to form
compound I.
In the eleventh aspect of the present invention, it provides an intermediate,
wherein
the intermediate has the structure of formula II or formula III:
3 rr"X
X
n(R )
n(R3
0
0 CI OV N
11 co 0
III
or
wherein X, A, R3 and n are as described in the fourth aspect of the present
invention.
In the twelfth aspect of the present invention, it provides a method for
preparing the
intermediate as described in the seventh aspect of the present invention,
noR3)A) n(R3)_-0
OH noR3)11---x
CI 0 'N
CO 4-HO sa.
wherein X, A, R3 and n are as described in the fourth aspect of the present
invention:
(1) the method comprises the steps of:
OH
(i) reacting the compound 4210 with (S)-1-((R3)-five-membered
heteroary1)-3-chloro-propanol in an inert solvent in the presence of a
condensation agent
to foim Intermediate II;
or (2) the method comprises the steps of:
OH
(i) reacting the compound 4200 with (S)-
1 -(e (R3 )-fi v e-m embered
heteroary1)-3-chloro-propanol in an inert solvent in the presence of a
condensation agent
to form Intermediate II; and
(ii) reacting Intermediate II with the potassium salt of phthalimide in an
inert solvent
to form Intermediate III.
In another preferred embodiment, the intemiediate is selected from the group
consisting of:
0
(R) (R) II
001
0 0
Th
In the thirteenth aspect of the present invention, it provides a non-
therapeutic and
non-diagnostic in vitro method for inhibiting transient receptor potential
channel protein
¨ 19 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
activity, wherein the method comprises the steps of: in a culture system in
vitro,
contacting the transient receptor potential channel protein or the cell
expressing the
protein with the compound as described in the third, fourth, fifth and/or
sixth aspect of the
present invention, or the pharmaceutically acceptable salt thereof, or the
prodrug thereof,
thereby inhibiting the activity of the transient receptor potential channel
protein.
In the fourteenth aspect of the present invention, it provides a method for
inhibiting
transient receptor potential channel protein or preventing and/or treating
diseases related
to transient receptor potential channel protein (TRP), wherein the method
comprises the
steps of: administrating the compound as described in the third, fourth, fifth
and/or sixth
aspect of the present invention, or the pharmaceutically acceptable salt
thereof, or the
prodrug thereof to a subject in need thereof.
In another preferred embodiment, the subject includes humans and non-human
mammals (rodents, rabbits, monkeys, domestic animals, dogs, cats, etc.).
In the fifteenth aspect of the present invention, it provides a compound, or a
pharmaceutically acceptable salt thereof, or a prodrug thereof, wherein the
compound has
a structure of formula G:
F-
( R8 ) I
in 1
Y
0
A1
R6
wherein:
s>3
ODAi is or
OD group; wherein, ring B is substituted or unsubstituted
5-7 membered carbocyclic ring, substituted or unsubstituted 5-7 membered
heterocyclic
ring, substituted or unsubstituted 5-7 membered heteroaryl, substituted or
unsubstituted
C6-C12 aryl; ring D is substituted or unsubstituted 5-7 membered heteroaryl,
substituted or
unsubstituted C6-C12 aryl; and when Al is a substituted or unsubstituted
aromatic
structure, Al contains 1-3 heteroatoms selected from N, 0 and S;
wherein the heterocyclic ring or heteroaryl contains 1-3 heteroatoms selected
from N,
0 and S;
R6 and R7 are each independently hydrogen, substituted or unsubstituted Ci-C6
alkyl,
substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted C2-
C4 acyl,
substituted or unsubstituted C2-C6 ester, or R6, R7 and their linking N atom
form a
substituted or unsubstituted C3 -C7 heterocycloalkyl; wherein, the
heterocycloalkyl
contains 1-2 N atoms and 0-1 0 or S atom;
Xi is a carbon atom, an oxygen atom, a sulfur atom or a nitrogen atom;
Yi is a carbon atom or a nitrogen atom;
at least one of Xi and Yi is a heteroatom;
R8 is hydrogen, halogen, substituted or unsubstituted Ci-C6 alkyl, substituted
or
unsubstituted C3-C7 cycloalkyl;
m is 1, 2, 3,4 or 5;
- 2 0 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
"*" represents a chiral carbon atom, and the absolute configuration of the
chiral
carbon atom is R-type and S-type;
wherein, any one of the "substituted" means that one to four (preferably 1, 2,
3)
hydrogen atoms on the group are substituted by a substituent selected from the
group
consisting of: C i-C6 alkyl, C3-C7 cycloalkyl, Ci-C3 haloalkyl, halogen,
nitro, cyano,
hydroxyl, Ci-Ca carboxyl, C2-C4 ester, C2-C4 amide, C i-C6 alkoxy, Ci-C6
haloalkyloxy,
benzyl, five- or six-membered aryl or heteroaryl (preferably C6 aryl or C5
heteroaryl).
In the present invention, it should be understood that in the compound of
formula G,
"*" represents a chiral carbon atom, and the absolute configuration of the
chiral carbon
atom being R-type and S-type refers to the racemic form.
.1<
In another preferred embodiment, Ai is OD
In another preferred embodiment, Ai is not a naphthalene ring.
In another preferred embodiment, Ai is a substituted or unsubstituted C6-C12
bicyclic
heteroaryl, a substituted or unsubstituted 5-6 membered heterocycle phenyl, a
substituted
or unsubstituted 5-6 membered heterocycle5-6 membered heteroaryl, or
substituted or
unsubstituted C6-C12 benzoalicyclic group.
In another preferred embodiment, the C6-C12 bicyclic heteroaryl is quinolinyl,
isoquinolinyl, phthalimidyl, benzofuranyl, benzothienyl, indolyl,
benzooxazolyl,
benzothiazolyl, quinoxalinyl, imidazopyridyl or benzimidazolone.
In another preferred embodiment, the C6-C12 benzoalicyclic group includes
indanyl,
tetrahydronaphthyl or dihydronaphthyl.
In another preferred embodiment, Ai is substituted or unsubstituted
benzofuranyl,
benzothienyl, or indanyl.
In another preferred embodiment, at least one of Xi and Yi is a heteroatom.
In another preferred embodiment, Xi is S or 0.
In another preferred embodiment, Xi is S.
In another preferred embodiment, the heteroaryl contains 1-3 heteroatoms
selected
from N, 0, or S.
In another preferred embodiment, the substituted refers to being substituted
by one to
four substituents (preferably 1, 2, or 3) selected from the group consisting
of: C i-C3 alkyl,
C3-C7 cycloalkyl, C i-C3 haloalkyl, halogen, nitro, cyano, hydroxyl, carboxy,
C2-C4 ester,
C2-C4 amide, CI-Ca alkoxy, Ci-C6 haloalkoxy, benzyl, five-membered or six-
membered
aryl or heteroaryl (preferably C6 aryl or C5 heteroaryl).
In another preferred embodiment, Ai is a substituted or unsubstituted C6-Ci2
bicyclic
heteroaryl, a substituted or unsubstituted 5-6 membered heterocycle phenyl, a
substituted
or unsubstituted 5-6 membered heterocycle 5-6 membered heteroaryl, or
substituted or
unsubstituted C6-C12 benzoalicyclic group.
In another preferred embodiment, R6 and R7 are each independently a hydrogen
atom,
a C i-C3 alkyl, a C2-C4 acyl; or R6, R7 and their linking N atom form a
tetrahydropyrrolyl
substituted with carboxyl or a C2-C4 ester.
In another preferred embodiment, R8 is a hydrogen atom, halogen, substituted
or
unsubstituted C -C3 alkyl.
In another preferred embodiment, Ai is quinolinyl, isoquinolinyl,
phthalimidyl,
benzofuranyl, benzothienyl, indolyl, benzoxazolyl, benzothiazolyl,
quinoxalinyl,
imidazopyridyl, benzimidazolone, indanyl, tetrahydronaphthyl or
dihydronaphthyl.
In another preferred embodiment, R6 and R7 are each independently hydrogen
atom,
methyl, acetyl, or R6, R7 and their linking N atom (bun a proline group or a
proline methyl
ester group.
In another preferred embodiment, R8 is a hydrogen atom, a chlorine atom, or a
methyl.
-21 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, the compound of formula G is selected from
the
group consisting of:
0 NH
0 0 0 0 0
CI
H 0
0 0
=
0
In the sixteenth aspect of the present invention, it provides a use of the
compound of
formula G as described in the fifteenth aspect of the present invention for
(a) preparing a
transient receptor potential channel protein (TRP) inhibitors; (b)
manufacturing a
medicament for preventing and/or treating diseases related to transient
receptor potential
channel protein (TRP).
In another preferred embodiment, the transient receptor potential channel
protein
(TRP) is TRPAl.
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRP) is selected from the group consisting of pain, epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder, or
inflammatory bowel
disease.
In another preferred embodiment, the pain includes acute inflammatory pain,
chronic
inflammatory pain, visceral pain, neurogenic pain, fibromyalgia, headache,
neuralgia, or
cancer-induced pain.
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRP) is selected from the group consisting of pain, epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder,
inflammatory bowel
disease, and combinations thereof.
In another preferred embodiment, the pain is selected from the group
consisting of
acute pain, inflammatory pain, visceral pain, neurogenic pain, fibromyalgia,
headache,
nerve pain, mixed pain, cancer-induced pain, and combinations thereof.
-22 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In another preferred embodiment, the postoperative pain is postoperative pain
following a surgical procedure.
In another preferred embodiment, the postoperative pain is post-operative
wound pain.
In another preferred embodiment, the post-operative wound pain is selected
from the
group consisting of post-operative skin wound pain, post-operative muscle
wound pain,
and combinations thereof.
In another preferred embodiment, the post-operative wound pain is skin and
muscle
post-operative wound pain.
In another preferred embodiment, the inflammatory pain is chronic inflammatory
pain.
In another preferred embodiment, the inflammatory pain is osteoarthritis pain
or
rheumatoid arthritis pain.
In another preferred embodiment, the headache is migraine or muscular tension
pain.
In another preferred embodiment, the neuralgia is trigeminal neuralgia,
diabetic pain,
sciatica, or postherpetic neuralgia.
In the seventeenth aspect of the present invention, it provides a method for
preparing a
compound of formula G, or a pharmaceutically acceptable salt thereof, or a
prodrug
thereof as described in the fifteenth aspect of the present invention, wherein
the method
comprises the steps: in an inert solvent, reacting intermediate G-1 with R6-NH-
R7
compound to form the compound:
R7 6
m( R8 ) I I m(R8) __
Yi-
Yi-
-R7
0* Cl 0
A, A,
R6
G-1
wherein Xi, Yi, Ai, R6, R7, R8 and "*" are as described in the fifteenth
aspect of the
present invention.
In the eighteenth aspect of the present invention, it provides a hydrochloride
of the
compound of formula I-1 or a crystal form A thereof,
(R)
0
I.- 1
In another preferred embodiment, in the crystal form A of the hydrochloride of
the
compound I-1, the molecular molar ratio of the compound of Formula I-1 to
hydrochloric
acid is 4: 1,3: 1,2: 1, 1: 1, 1: 2, 1: 3 or 4: 1.
In another preferred embodiment, the crystal form A of the hydrochloride of
the
-23
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
compound of Formula I-1 is an anhydrous crystal form.
In another preferred embodiment, the X-ray powder diffraction pattern of the
crystal
form A of the hydrochloride of the compound of Formula I-1 has characteristic
peaks at 20
angles of 18.173+0.2 , 22.084+0.2 , and 22.794+0.2 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 further has characteristic peaks at one or more 20
values
selected from 16.734+0.2 , 21.156+0.2 , and 23.761+0.2 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 further has characteristic peaks at one or more 20
values
selected from 17.092+0.2 , 21.649+0.2 , 25.298+0.2 , 28.099+0.2 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Foimula I-1 further has characteristic peaks at one or more 20
values
selected from 10.003+0.2 , 26.640+0.2 , 28.615+0.2 , 28.813+0.2 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 further has characteristic peaks at one or more 20
values
selected from 10.003+0.2 , 16.734+0.2 , 17.092+0.2 , 18.173+0.2 , 21.156+0.2 ,
21.649+0.2 , 22.084+0.2 , 26.640+0.2 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 further has characteristic peaks at one or more 20
values
selected from 11.171+0.2 , 15.987+0.2 , 18.849+0.2 , 20.681+0.2 , 25.967+0.2 ,
27.273+0.2 , 29.501+0.2 , 30.118+0.2 , 30.513+0.2 , 32.522+0.2 , 33.274+0.2 ,
34.081+0.2 , 35.815+0.2 , 37.553+0.2 , 40.018+0.2 , 42.927+0.2 , 44.129+0.2.
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 further has characteristic peaks at one or more 20
values
selected from 10.003+0.2 , 11.171+0.2 , 15.987+0.2 , 16.734+0.2 , 17.092+0.2 ,
18.849+0.2 , 20.681+0.2 , 21.156+0.2 , 21.649+0.2 , 23.761+0.2 , 25.298+0.2 ,
25.967+0.2 , 26.640+0.2 , 27.273+0.2 , 28.099+0.2 , 28.615+0.2 , 28.813+0.2 ,
29.501+0.2 , 30.118+0.2 , 30.513+0.2 , 32.522+0.2 , 33.274+0.2 , 34.081+0.2 ,
35.815+0.2 , 37.553+0.2 , 40.018+0.2 , 42.927+0.2 , 44.129+0.2.
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 has characteristic peaks at one or more 20 values
selected from
10.003+0.2 , 11.171+0.2 , 15.987+0.2 , 16.734+0.2 , 17.092+0.2 , 18.173+0.2 ,
18.849+0.2 , 20.681+0.2 , 21.156+0.2 , 21.649+0.2 , 22.084+0.2 , 22.794+0.2 ,
23.761+0.2 , 25.298+0.2 , 25.967+0.2 , 26.640+0.2 , 27.273+0.2 , 28.099+0.2 ,
28.615+0.2 , 28.813+0.2 , 29.501+0.2 , 30.118+0.2 , 30.513+0.2 , 32.522+0.2 ,
33.274+0.2 , 34.081+0.2 , 35.815+0.2 , 37.553+0.2 , 40.018+0.2 , 42.927+0.2 ,
44.129+0.2.
In another preferred embodiment, the X-ray powder diffraction pattern of the
crystal
form A of the hydrochloride has characteristic peaks and peak intensities at
one or more
20 values selected from the group consisting of:
Relative
20value
value Intensity %
10.003 8.8355 28.3
11.171 7.9137 12.8
15.987 5.5392 15.2
16.734 5.2936 51.9
17.092 5.1836 35.7
18.173 4.8774 100.0
18.849 4.7041 12.7
20.681 4.2913 13.6
21.156 4.1961 65.2
¨ 24
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
21.649 4.1017 35.3
22.084 4.0217 71.9
22.794 3.8981 91.8
23.761 3.7416 65.0
25.298 3.5177 46.3
25.967 3.4285 11.5
26.640 3.3433 29.2
27.273 3.2672 16.9
28.099 3.1730 35.7
28.615 3.1170 33.1
28.813 3.0959 25.2
29.501 3.0253 10.4
30.118 2.9647 12.1
30.513 2.9272 13.2
32.522 2.7508 18.4
33.274 2.6904 12.4
34.081 2.6285 13.2
35.815 2.5051 13.4
37.553 2.3931 9.6
40.018 2.2512 8.2
42.927 2.1051 10.6
44.129 2.0505 9.0 .
In another preferred embodiment, the crystal form A of the hydrochloride of
the
compound of Formula I-1 has X-ray powder diffraction characteristic peaks
substantially as
shown in Fig. 7.
In another preferred embodiment, the differential scanning calorimetry (DSC)
pattern
of the crystal fonn A of the hydrochloride of the compound of Formula I-1
begins to
appear endothermic peaks upon being heated to 142.30 C (preferably 4 C, 3 C,
2 C or
1 C).
In another preferred embodiment, the differential scanning calorimetry (DSC)
pattern
of the crystal form A of the hydrochloride of the compound of Formula I-1 is
substantially
as shown in Fig. 8.
In another preferred embodiment, the thermogravimetric analysis (TGA) pattern
of
the crystal form A of the hydrochloride of the compound of Fonnula I-1 has a
weight loss
of about 0.9827% (preferably 0.1%, 0.2%, 0.3%, 0.4%, or 0.5%) upon being
heated
to 168.01 C .
In another preferred embodiment, the thermogravimetric analysis (TGA) pattern
of
the crystal form A of the hydrochloride is substantially as shown in Fig. 9.
In the nineteenth aspect of the present invention, it provides a method for
preparing
the crystal form A of the hydrochloride of the compound of Formula I-1 as
described in
the eighteenth aspect of the present invention, wherein the method comprises
the steps of:
(a) after mixing the compound of formula I-1 with an organic solvent, adding
hydrochloric acid dropwise at 5-15 C to adjust the pH of the system to 6-8, a
solid was
precipitated during the reaction, and filtering to obtain the crystal form A
of the
hydrochloride of the compound of Formula I-1.
In another preferred embodiment, in step (a), the organic solvent comprises
ethyl
acetate.
In another preferred embodiment, in step (a), the hydrochloric acid is
concentrated
hydrochloric acid.
-25 -
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
In another preferred embodiment, in step (a), the pH of the system is 6.5-7.5,
preferably 7Ø
In another preferred embodiment, in step (a), the reaction time is 3-8min,
preferably
5min.
In another preferred embodiment, in step (a), the reaction is reacted under
stirring
conditions.
In another preferred embodiment, in step (a), the hydrochloric acid is slowly
added.
In another preferred embodiment, in step (a), the weight-volume ratio (kg: L)
of the
compound of Fonnula I-1 to the organic solvent is 0.2-2: 2-30, preferably 0.4-
1.0: 5-18,
more preferably 0.5-0.9: 8-15.
In another preferred embodiment, in step (a), after the solid was
precipitated, the
crystal form A of the hydrochloride of the compound of Formula I-1 was
obtained by
drying at 40-45 C.
In the twentieth aspect of the present invention, it provides a pharmaceutical
composition comprising the crystal form A of the hydrochloride of the compound
of
Formula I-1 as described in the eighteenth aspect of the present invention;
and
pharmaceutically acceptable carriers.
In the twenty-first aspect of the present invention, it provides a use of the
crystal
form A of the hydrochloride of the compound of Formula I-1 as described in the
eighteenth aspect of the present invention for (a) preparing transient
receptor potential
channel protein (TRP) inhibitors; (b) manufacturing a medicament for
preventing and/or
treating diseases related to transient receptor potential channel protein
(TRP).
In another preferred embodiment, the transient receptor potential channel
protein
(TRP) is TRPAL
In another preferred embodiment, the disease related to transient receptor
potential
channel protein (TRP) is selected from the group consisting of pain, epilepsy,
inflammation, respiratory disorder, pruritus, urinary tract disorder,
inflammatory bowel
disease, and combinations thereof.
In another preferred embodiment, the pain is selected from the group
consisting of
acute pain, inflammatory pain, visceral pain, neurogenic pain, muscle fiber
pain, headache,
nerve pain, mixed pain, cancer-induced pain, and combinations thereof.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In another preferred embodiment, the postoperative pain is postoperative pain
following a surgical procedure.
In another preferred embodiment, the postoperative pain is post-operative
wound pain.
In another preferred embodiment, the post-operative wound pain is selected
from the
group consisting of post-operative skin wound pain, post-operative muscle
wound pain,
and combinations thereof.
In another preferred embodiment, the post-operative wound pain is skin and
muscle
post-operative wound pain.
In another preferred embodiment, the inflammatory pain is chronic inflammatory
pain.
In another preferred embodiment, the inflammatory pain is osteoarthritis pain
or
rheumatoid arthritis pain.
In another preferred embodiment, the headache is migraine or muscular tension
pain.
In another preferred embodiment, the neuralgia is trigeminal neuralgia,
diabetic pain,
sciatica, or postherpetic neuralgia.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
In the twenty-second aspect of the present invention, it provides a non-
therapeutic and
- 26 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
non-diagnostic in vitro method for inhibiting the activity of transient
receptor potential
channel proteins, which comprises the steps of: contacting the transient
receptor potential
channel proteins or cells expressing the proteins with the compound as
described in the
third aspect of the present invention, or the pharmaceutically acceptable salt
thereof, or the
prodrug thereof, the compound as described in the fourth aspect of the present
invention,
or the pharmaceutically acceptable salt thereof, or the prodrug thereof, the
compound as
described in the fifth aspect of the present invention, or the
pharmaceutically acceptable
salt thereof, or the prodrug thereof, the compound as described in the sixth
aspect of the
present invention, or the pharmaceutically acceptable salt thereof, or the
prodrug thereof,
or the crystal form A of the hydrochloride of the compound of Formula I-1 as
described in
the eighteenth aspect of the present invention, thereby inhibiting the
activity of the
transient receptor potential channel protein.
In the twenty-third aspect of the present invention, it provides a method for
inhibiting
transient receptor potential channel protein or preventing and/or treating
diseases related
to transient receptor potential channel protein (TRP), administering the
compound as
described in the third aspect of the present invention, or the
pharmaceutically acceptable
salt thereof, or the prodrug thereof, the compound as described in the fourth
aspect of the
present invention, or the pharmaceutically acceptable salt thereof, or the
prodrug thereof,
the compound as described in the fifth aspect of the present invention, or the
pharmaceutically acceptable salt thereof, or the prodrug thereof, the compound
as
described in the sixth aspect of the present invention, or the
pharmaceutically acceptable
salt thereof, or the prodrug thereof, or the crystal form A of the
hydrochloride of the
compound of Formula I-1 as described in the eighteenth aspect of the present
invention to
a subject in need thereof.
It should be understood that within the scope of the present invention, the
above-described technical features of the present invention and the technical
features
described in detail below (e.g., embodiments) may be combined with each other
to
constitute a new or preferred technical solution. Limited by space, it will
not be repeated
here.
DESCRIPTION OF THE DRAWINGS
Fig. 1 shows the TRPA1 inhibitory activity IC50 of compound I-1 tested by
automatic
patch clamp.
Fig. 2 shows the TRPA1 inhibitory activity IC50 of compound I-1 tested by
manual
patch clamp.
Fig. 3 shows the ED50 results of compound I-1 of the present invention, S-
duloxetine
and comparative compound Cl in the formalin pain model in mice.
Fig. 4 shows the statistical graph of MPE% of compound I-1 of the present
invention,
S-duloxetine, comparative compound Cl and gabapentin in the hot plate induced
pain
model in rat at different times.
Fig. 5 shows the results of analgesic activity of compound I-1 of the present
invention, S-duloxetine, indomethacin and anisodamine in the acetic acid
writhing pain
model in mice.
Fig. 6 shows the results of analgesic activity of compound I-1 of the present
invention, S-duloxetine and gabapentin in the SNL model in rat.
Fig. 7 is an X-ray powder diffraction pattern of the crystal form A of the
hydrochloride of the compound I-1.
Fig. 8 is a differential scanning calorimetry (DSC) pattern of the crystal
form A of
the hydrochloride of the compound I-1.
Fig. 9 is a thermogravimetric analysis (TGA) pattern of the crystal form A of
the
-27 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
hydrochloride of the compound I-1.
Fig. 10 shows the results of analgesic activity of compound I-I of the present
invention in a postoperative pain model in rat.
DETAILED DESCRIPTION OF THE INVENTION
Through extensive and in-depth research, the present inventors have
unexpectedly
developed, for the first time, a compound, or a pharmaceutically acceptable
salt thereof, or
a prodrug thereof, the compound has a structure of Formula I, Foimula Z,
Foimula G,
Formula A or Formula B. Experiments have shown that the compounds of the
present
invention have a significant inhibitory effect on TRP channels. The compound
of the
present invention can effectively treat pain related to TRP (especially TRPA1)
targets. In
addition, the present invention also provided a crystal form A of the
hydrochloride of the
compound of Foimula I-I in solid form, the crystal form A of the hydrochloride
of the
compound of Formula I-I is convenient for storage, transportation, and has
strong
druggability and strong stability (especially with excellent thermal stability
and high
humidity stability). The present invention has been completed on this basis.
Terms
As used herein, the terms "include," "comprise" and "contain" are used
interchangeably to include not only closed definitions, but also semi-closed,
and open
definitions. In other words, the term includes "consist of' and "substantially
consist of'.
As use herein, "RI", "Ri" and "Rl" have the same meaning and can be replaced
with
each other, and other similar definitions have the same meaning.
As use herein, the term "Ci-C6 alkyl", "Ci-C3 alkyl" or "Ci-C4 alkyl" refers
to a
linear or branched alkyl with 1 to 6, 1 to 3 or 1 to 4 carbon atoms, such as
methyl, ethyl,
propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, or the like.
As use herein, the term "Ci-C6 alkoxy" refers to a linear or branched alkoxy
with 1-6
carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy,
sec-butoxy, tert-butoxy, pentyloxy, hexyloxy, or the like.
As use herein, the term "C6-C12 benzo aliphatic ring" refers to a group with 6-
12
carbon atoms, including indanyl, tetrahydronaphthyl or dihydronaphthyl and the
like.
As use herein, the term "CC 6 haloalkoxy" refers to a linear or branched
alkoxy
with 1-6 carbon atoms in which one or more hydrogen atoms are substituted by a
halogen,
such as chloromethoxy, chloroethoxy, or the like.
As use herein, the term "C3-C7 cycloalkyl", "C3-C6 cycloalkyl" refers to a
cycloalkyl
(including monocyclic, dicyclic or polycyclic) with 3-7 or 3-6 carbon atoms,
such as
cyclopropyl, cyclobutyl, methylcyclobutyl, cyclopentyl, cycloheptyl, or the
like.
As use herein, the term "C2-C4 ester" refers to a group having structure of Ci-
C3
alkyl-OC(0)- or -OC (0)-Ci-05 alkyl, in which the alkyl can be linear or
branched, such
as CH3C00-, C2H5C00-, C3118C00-, (CH3) 2CHC00-, -COOCH3, -CO0C2H5,
-CO0C3118, or the like.
As use herein, the term "C2-C4 amide" refers to a group having structure of
Ci-C3alkyl-CO-NH- or -NH-CO-Ci-C3 alkyl, in which the alkyl can be linear or
branched,
such as CH3-CO-NH-, C2H5-CO-NH-, -
COOCH3, -CO-NH-C2H5,
-CO-NH-C3H8, or the like.
As use herein, the term "C2-C4 acyl" refers to a group having structure of
Ci-C3alkyl-00-, in which the alkyl can be linear or branched, such as CH3-00-,
C2H5-00-, C3118-00-, or the like.
As use herein, the term "C3-C7 heterocycloalkyl" refers to a monocyclic or
polycyclic heterocycles (preferably monocyclic heterocycles) having 3-7 ring
carbon
atoms and 1-3 heteroatoms (preferably contains 1 nitrogen atom, that is, the
nitrogen atom
adjacent to 12_1 and R2), such as piperidinyl, tetrahydropyrrolyl, or the
like.
¨28 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
As used herein, the tem' "5-7-membered carbocyclic" refers to any stable 5, 6,
or
7-membered monocyclic, bicyclic, or polycyclic ring, and the carbocyclic may
be
saturated, partially unsaturated, unsaturated, but cannot be aromatic.
Examples of the
carbocyclic rings include, but are not limited to, cyclopropyl rings,
cyclobutyl rings,
cyclobutene rings, cyclopentyl rings, cyclopentene rings, cyclohexyl rings,
cyclohexene
rings, cycloheptyl rings, cycloheptene rings, or the like.
As used herein, the term "5-7 membered heterocyclic" refers to any stable
monocyclic, bicyclic or polycyclic ring containing one or more (preferably 1,
2 or 3)
heteroatoms selected from N, 0 and S, and the number of ring atoms in the
heterocyclic
ring is 5-7, the heterocyclic ring can be a saturated, partially unsaturated,
or unsaturated
ring, but cannot be an aromatic ring. It should be understood that when there
are multiple
heteroatoms, the heteroatoms can be identical, partially identical, or
completely different.
As used herein, the term "C1-C3 haloalkyl" refers to a linear or branched
alkyl
having 1 to 3 carbon atoms in which one or more hydrogen atoms are substituted
by
halogen groups, such as monochloromethyl, dichloroethyl, trichloropropyl, or
the like.
As used herein, the term "C1-C4 carboxy" refers to a group having structure of
C1-C3 alkyl-COOH, in which the alkyl can be linear or branched, such as
CH3C0OH,
C2H5COOH, C3H8COOH, (CH3)2CHCOOH, or the like.
As used herein, the term "C6-C12 aryl" refers to a monocyclic or bicyclic
aromatic
hydrocarbon group having 6 to 12 carbon atoms in the ring portion, such as
phenyl,
naphthyl, biphenyl, or the like.
As used herein, the term "5-7 membered heteroaromatic ring" refers to an
aromatic
heterocyclic ring system having one to more (preferably 1, 2, or 3)
heteroatoms selected
from N, 0, and S, and having 5- 7 ring atoms. It should be understood that
when there are
multiple heteroatoms, the heteroatoms can be identical, partially identical,
or completely
different. For example, examples of 5-membered heteroaromatic rings include
(but are not
limited to): pyrrole ring, furan ring, thiophene ring, imidazole ring, oxazole
ring, thiazole
ring, examples of 6-membered heteroaromatic ring include (but are not limited
to)
pyridine ring, pyrazine ring, pyridazine ring, pyrimidine ring, or the like.
As used herein, the term "five- or six-membered heteroaryl" refers to an
aromatic
group having one to more (preferably 1, 2, or 3) heteroatoms selected from N,
0, and S.
and having 5 or 6 ring atoms. It should be understood that when there are
multiple
heteroatoms, the heteroatoms can be identical, partially identical, or
completely different.
For example, examples of 5-membered heteroaryl include (but are not limited
to): pyrrolyl,
furyl, thienyl, imidazolyl, oxazolyl, thiazolyl, or the like.
As used herein, the term "six-membered aryl" refers to an aromatic group
having 6
ring atoms, and the ring atoms are all carbon atoms, such as a phenyl, or the
like.
As use herein, that term "halogen" refers to F, Cl, Br and I.
X
nue ,
As used herein, I and
have the same meaning, and both
represent a unsubstituted heteroaryl or a heteroaryl substituted with 1 to 5
(preferably 1 to
3) R3 substituents.
As used herein, the term "substituted" means that the hydrogen atom on the
group is
substituted by a non-hydrogen atom group, but the valence requirements must be
met and
a chemically stable compound is generated by the substitution. In the
specification, it
should be construed that all substituents are unsubstituted, unless expressly
described as
"substituted" herein. In a preferred embodiment, any of the "substituted"
means that 1-4
(preferably 1, 2, 3, or 4) hydrogen atoms on the group are each independently
substituted
by a substituent selected from the group consisting of: Ci -C6 alkyl, C3-c7
cycloalkyl,
C -C3 haloalkyl, halogen, nitro, cyano, hydroxyl, C1-C4 carboxy, C2-C4 ester,
C2-C4
¨29 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
amide, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, benzyl, six-membered aryl, five- or
six-membered heteroaryl (preferably C5 heteroaryl).
It should be understood that in the present invention, substituents can be
connected
to the parent group or substrate on any atom, unless the connection violates
the valence
requirement; the same or different substituents can be on the same atom or on
different
atoms.
Similarly, it should be understood that those ordinary skilled in the art can
select the
substituents and substitution on the compounds of the present invention to
produce
chemically stable compounds, which can be synthesized by techniques known in
the art
and method described below. If substituted by more than one substituent, it
should be
understood that the multiple groups may be on the same carbon or on different
carbons, as
long as a stable structure is produced.
In the present invention, the structures of R-duloxetine and S-duloxetine are
as
follows:
(R) (õS)
R-duloxetine S-duloxetine
Active ingredient
As used herein, the compound of formula I of the present invention refers to a
compound having the structure of formula I, or a pharmaceutically acceptable
salt thereof,
or a prodrug thereof. It should be understood that the term also includes
mixtures of the
aforementioned components.
As used herein, the compound of formula Z of the present invention refers to a
compound having the structure of formula Z, or a pharmaceutically acceptable
salt thereof,
or a prodrug thereof. It should be understood that the term also includes
mixtures of the
aforementioned components.
As used herein, the compound of formula G of the present invention refers to a
compound having the structure of foimula G, or a pharmaceutically acceptable
salt thereof,
or a prodrug thereof. It should be understood that the term also includes
mixtures of the
aforementioned components.
The compound of the present invention not only has an inhibitory effect on
TRPA1,
but also has a certain inhibitory effect on other members of the TRP family.
The term "pharmaceutically acceptable salt" refers to a salt formed by the
compound
of the present invention and an acid or a base suitable for use as a medicine.
Pharmaceutically acceptable salts include inorganic salts and organic salts. A
preferred
type of salt is the salt formed by the compound of the present invention and
an acid, acids
suitable for salt formation include (but are not limited to): hydrochloric
acid, hydrobromic
acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid and other
inorganic acids,
formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic
acid, fumaric
acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric
acid,
methanesulfonic acid, benzenemethanesulfonic acid, benzenesulfonic acid and
other
organic acids; and acidic amino acids such as aspartic acid and glutamic acid,
etc. A
preferred type of salt is a metal salt formed by the compound of the present
invention and
a base, suitable bases for salt formation include (but are not limited to):
inorganic bases
such as sodium hydroxide, potassium hydroxide, sodium carbonate, sodium
bicarbonate,
sodium phosphate, etc., organic bases such as ammonia, triethylamine,
diethylamine, etc.
In the present invention, a preferred pharmaceutically acceptable salt of
compound of
¨ 30 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
formula Z, compound of formula I, compound of formula A or compound of formula
B is
a salt formed by compound of formula Z, compound of formula I, compound of
formula A
or compound of formula B and acids selected from the group consisting of:
hydrochloric
acid, mucic acid, D-glucuronic acid, hydrobromic acid, hydrofluoric acid,
sulfuric acid,
nitric acid, phosphoric acid, formic acid, acetic acid, propionic acid, oxalic
acid, malonic
acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid,
tartaric acid, citric
acid, picric acid, methanesulfonic acid, benzenemethanesulfonic acid,
benzenesulfonic
acid, aspartic acid, glutamic acid, or combinations thereof.
Preferably, the pharmaceutically acceptable salt of compound of foimula Z,
compound of foimula I, compound of formula A or compound of formula B is a
salt
formed by compound of formula Z, compound of formula I, compound of formula A
or
compound of foimula B and acids selected from the group consisting of:
hydrochloride,
maleate, oxalate, mucate, fumarate, D-glucuronate, or combinations thereof.
Preferred compounds of the present invention include any one compound selected
from Table 1 below:
Table 1
Number Name
Structural formula
(R)-3 - (benzofuran-7-y loxy )-N
o?)/N
Compound I-1 -methyl-3-(thiophen-2-yl)prop
an-1-amine 0
(R)-3-(benzofuran-7-yloxy)-3-
Compound 1-2 (thiophen-2-yl)propan-l-amin 0 NH2
(R)-3 - (benzofuran-7-y loxy )-N
Compound 1-3 ,N-dimethy1-3-(thiophen-2-y1)
propan-1 -amine 0
(R)-3-(benzofuran-7-yloxy)-N-e
Compound 1-4 thy1-3-(thiophen-2-yl)propan-1- o`N
amine oL
(R)-3-(benzofuran-7-yloxy )-N
Compound 1-5 -methyl-3-(thiophen-3-y 1)prop
an-1-amine
-31 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
(R)-3-(benzofuran-7-yloxy)-N
Compound 1-6 -methy1-3-(5-methylthiophen-
2-yl)propan- I-amine
0
CI
(R)-3-(benzofuran-7-yloxy)-3-
Compound 1-7 (5-chlorothiophen-2-y1)-N-met
hylpropan-l-amine
0
(R)-3-(benzofuran-7-yloxy)-3-
R)
Compound 1-8 (furan-2-y1)-N-methylpropan- 0)Cz-Nz
1-amine
(R)-3-((2,3-dihydro-1H-inden-
z
Compound 1-9 4-yl)oxy)-N-methyl-3-(thioph ON
en-2-yl)propan-l-amine
0
(R)-N-methyl-3-((5,6,7,8-tetra
R)
Compound 1-10 hydronaphthalen- 1 -yl)oxy)-3-(
thiophen-2-yl)propan-1-amine
(R)-3-(benzofuran-4-yloxy)-N
Nz
Compound I-11 -methyl-3-(thiophen-2-yl)prop O
an- 1-amine
Preparation method
The present invention also provided a preparation method of
(R)-3-aryloxy-3-five-membered heteroaryl-propylamine compound represented by
Formulal.
The present invention also provided a preparation method of intermediates II
to III,
which can be used for preparing the above-mentioned compounds.
The specific synthesis strategies are as follows:
Synthesis of the compound represented by formula I:
¨32 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
-
diisopropyl n(R)ff.)
R1 R2 n(12)¨V.
OH 3 azodicarboxylate sodium (R) R1
N'
"(R triphenylphosphine 0 _________________ 01 iodide 0
Anhydrous
HO CI tetrahydrofuran CIO acetone Tetrahydro-
furan
.g
F:1 g
t
.4 E
phthalimide rX
potassium salt n(R3) __ ii o
sodium iodide (R)
N,N-dimethylformamide
CIO
In
wherein A, X, R2, le and n are as defined above.
1) dissolving a fused furan ring phenol or fused aliphatic ring phenol,
(S)-1-(n(R3)-five-membered heteroary1)-3-chloro-propanol and
triphenylphosphine into
anhydrous tetrahydrofuran, slowly adding diisopropyl azodicarboxylate dropwise
to the
system under ice bath conditions, after the dropwise addition, transfering the
system to
20-25 C for overnight reaction. After completion of the reaction, spin-drying
the system
directly, and separating and purifing the residue by column chromatography to
obtain the
intermediate II.
2) dissolving the intermediate II into a saturated sodium iodide in acetone,
and
reacting overnight at a temperature of 50-70 C. After completion of the
reaction,
spin-drying the solvent, adding water into the system, extracting three times
with ethyl
acetate, washing with saturated brine, drying with anhydrous sodium sulfate,
filtering, and
concentrating; then dissolving the residue in tetrahydrofuran, and adding an
aqueous
solution or alcohol solution of amine, and reacting overnight at 20-25 C.
After
completion of the reaction, spin-drying the solvent, and adding a sodium
hydroxide
aqueous solution to the system, extracting with ethyl acetate for three times,
washing with
saturated brine, drying with anhydrous sodium sulfate, filtering, and
concentrating, then
separating the residue by column chromatography to obtain the compound I.
3) dissolving the intermediate II, phthalimide potassium salt and sodium
iodide in
N,N-dimethylformamide solution, and reacting overnight at 70-90 C. After
completion of
the reaction, adding water to the system, extracting with ethyl acetate for
three times,
washing with water, washing with saturated brine, drying with anhydrous sodium
sulfate,
filtering, and concentrating, then separating the residue by column
chromatography to
obtain the Intermediate III.
4) dissolving the Intermediate III in a methanol solution, adding hydrazine
hydrate,
and reacting overnight at 20-25 C. After completion of the reaction, spin-
drying the
solvent, and separating the residue by column chromatography to obtain the
compound I.
Synthesis of salts of compound
The compounds as shown in Formula I, Fonnula Z, Founula G, Formula A or
Formula B of the present invention can be converted into pharmaceutically
acceptable
salts by conventional methods, for example, the corresponding acid solution
can be added
into the solution of the above compounds, and the corresponding salts of the
compounds
of the present invention can be obtained by removing the solvent under reduced
pressure
after the salt formation is complete.
Transient receptor potential channel protein (TRP)
¨ 33 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
Transient receptor potential channel proteins are a protein superfamily
consisted of
important cation channels existing on the cell membrane. Transient receptor
potential
channel proteins include several subgroups, such as TRPA, TRPC, TRPM, TRPV,
TRPML
and TRPP subgroups.
TRPA1 is a member of TRPA subfamily, TRPA1 is also called as transient
receptor
potential anchor protein 1. Studies have found that TRPA1 channel protein is
related to
diseases such as pain, epilepsy, inflammation, respiratory disorders,
pruritus, urinary tract
disorders, inflammatory bowel disease and other diseases. TRPA1 is the target
for treating
pain, epilepsy, inflammation, respiratory disorders, pruritus, urinary tract
disorders,
inflammatory bowel disease and other diseases.
Typically, the disease related to transient receptor potential channel protein
(TRP) is
pain. The compound of Formula I, Formula Z, Formula G or the crystal form A of
the
hydrochloride of the compound of Formula I has an effective therapeutic effect
on pain.
Typically, the pain includes (but is not limited to): acute pain, inflammatory
pain,
visceral pain, neurogenic pain, fibromyalgia, headache, nerve pain, mixed
pain,
cancer-induced pain, inflammation pain, and combinations thereof.
Typically, the acute pain is injury pain or postoperative pain.
As used herein, the terms "postoperative pain" and "post-surgical pain" are
used
interchangeably.
Typically, the postoperative pain is postoperative pain following a surgical
procedure.
Typically, the postoperative pain is post-operative wound pain.
Typically, the post-operative wound pain is selected from the group consisting
of
post-operative skin wound pain, post-operative muscle wound pain, and
combinations
thereof.
Typically, the post-operative wound pain is skin and muscle post-operative
wound
pain.
Typically, the inflammatory pain is chronic inflammatory pain.
Typically, the inflammatory pain is osteoarthritic pain or rheumatoid
arthritic pain.
Typically, the headache is migraine or muscle tension pain.
Typically, the neuralgia is trigeminal neuralgia, diabetic pain, sciatica, or
postherpetic neuralgia.
In another preferred embodiment, the acute pain is injury pain or
postoperative pain.
Use
The invention also provided a method of inhibiting transient receptor
potential
channel protein (TPR) and a method of treating diseases related to TPR.
The compound of Formula I, Formula Z, Formula G or the crystal form A of the
hydrochloride of the compound of Formula I of the present invention can be
used to
inhibit the transient receptor potential channel protein, thereby preventing
or treating
diseases related to the transient receptor potential channel protein.
The invention provided a use of the compound of Formula Z, or the
pharmaceutically
acceptable salt thereof, or the prodrug thereof, the compound of Formula I, or
the
pharmaceutically acceptable salt thereof, or the prodrug thereof, the compound
of Formula
G, or the pharmaceutically acceptable salt thereof, or the prodrug thereof, or
the crystal
form A of the hydrochloride of the compound of Formula I-1 for (a) preparating
a
transient receptor potential channel protein (TRPA1) inhibitors; (b)
manufacturing a
medicament for the prevention and/or treatment of diseases associated with
transient
receptor potential channel protein (TRPA1).
In the present invention, the transient receptor potential channel protein
(TPR) is
TPR1
In the present invention, examples of diseases related to transient receptor
potential
- 34
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
channel proteins include (but are not limited to): pain, epilepsy,
inflammation, respiratory
disorders, pruritus, urinary tract disorders, inflammatory bowel disease, or
combinations
thereof. Typically, the pain includes (but is not limited to): acute
inflammatory pain,
inflammatory pain (such as chronic inflammatory pain, osteoarthritis pain or
rheumatoid
arthritis pain), visceral pain, neurogenic pain, fibromyalgia, headache (such
as migraine,
muscular tension pain, etc.), nerve pain (such as trigeminal neuralgia,
diabetic pain,
postherpetic neuralgia, etc.), or cancer-induced pain.
In a preferred embodiment, the present invention provided a non-therapeutic
and
non-diagnostic in vitro method for inhibiting transient receptor potential
channel protein
activity, includes, for example, in a culture system in vitro, contacting a
transient receptor
potential channel protein or a cell expressing the protein with the compound
of Formula I,
Formula Z or Formula G, or the pharmaceutically acceptable salt thereof, or
the prodrug
thereof, or the crystal form A of the hydrochloride of the compound of Foimula
I-1
according to the present invention, thereby inhibiting the activity of the
transient receptor
potential channel protein.
In the present invention, the non-therapeutic and non-diagnostic in vitro
method for
inhibiting the activity of transient receptor potential channel proteins can
be used for drug
screening, quality control and other purposes. For example, in a culture
system in vitro, by
contacting the compound of formula I, formula Z, or formula G, or the
pharmaceutically
acceptable salt thereof, or the prodrug thereof, or the crystal fotin A of the
hydrochloride
of the compound of Formula I-1 of the present invention with transient
receptor potential
channel protein or cells expressing the protein, and the compounds that can
inhibit
transient receptor potential channel protein are selected as candidate drugs.
Then, the
therapeutic effect of the candidate compounds can be further studied through
animal
experiments and clinical trials on transient receptor potential channel
protein and the
related diseases.
The invention also provided a method for inhibiting transient receptor
potential
channel proteins, which may be therapeutic or non-therapeutic. Generally, the
method
comprises the steps of: administering the compound of formula I, formula Z,
formula G,
formula A, or formula B, or the pharmaceutically acceptable salt thereof, or
the prodrug
thereof, or the crystal form A of the hydrochloride of the compound of Formula
I-1 of the
present invention to a subject in need thereof.
Preferably, the subject includes humans and non-human mammals (rodents,
rabbits,
monkeys, domestic animals, dogs, cats, etc.).
Crystal form
The present invention also provided a crystal form A of the hydrochloride of
the
compound of Formula I-1,
(R)
0
I-1
The crystal form A of the hydrochloride of the compound of Formula I-1 of the
present invention is in solid form. Compared with the oily substance of the
free compound
of formula I-1, the solid form of the salt crystal form of the compound of
formula I-1 is
convenient for storage, transportation and has a strong druggability. The
crystal form A of
¨35 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
the hydrochloride of the compound of Formula I-1 as described herein also has
excellent
stability, especially excellent thermal stability and high humidity stability.
As used herein, the terms "crystal form A of the hydrochloride of the compound
of
Formula I-1", "crystal form A of the hydrochloride " and "crystal form A" can
be used
interchangeably.
The crystal form A of the hydrochloride of the compound of Formula I-I as
described
herein has characteristic peaks at one or more 20 values selected from the
group consisting
of 10.003+0.2 , 11.171 0.2 , 15.987+0.2 , 16.734 0.2 , 17.092 0.2 , 18.173 0.2
,
18.849 0.2 , 20.681 0.2 , 21.156 0.2 , 21.649 0.2 , 22.084 0.2 , 22.794 0.2 ,
23.761 0.2 , 25.298 0.2 , 25.967 0.2 , 26.640 0.2 , 27.273 0.2 , 28.099 0.2 ,
28.615 0.2 , 28.813 0.2 , 29.501 0.2 , 30.118 0.2 , 30.513 0.2 , 32.522 0.2 ,
33.274 0.2 , 34.081+0.2 , 35.815 0.2 , 37.553 0.2 , 40.018+0.2 , 42.927 0.2 ,
44.129 0.2.
Typically, the crystal form A of the hydrochloride of the compound of Formula
I-1
has X-ray powder diffraction characteristic peaks substantially as shown in
Fig. 7.
In another preferred embodiment, the differential scanning calorimetry (DSC)
pattern
of the crystal foim A of the hydrochloride of the compound of Formula I-1
begins to
appear endothermic peaks upon being heated to 142.30 C (preferably 4 C, 3
C, 2 C
or 1 C).
Typically, the differential scanning calorimetry (DSC) pattern of the crystal
form A
of the hydrochloride of the compound of Formula I-1 is substantially as shown
in Fig. 8.
In another preferred embodiment, the thermogravimetric analysis (TGA) pattern
of
the crystal form A of the hydrochloride of the compound of Formula I-1 has a
weight loss
of about 0.9827% (preferably 0.1%, 0.2%, 0.3%, 0.4%, or 0.5%) upon being
heated
to 168.01 C.
Typically, the thermogravimetric analysis (TGA) pattern of the crystal form A
of the
hydrochloride of the compound of Formula I-1 is substantially as shown in Fig.
9.
Preferably, a method for preparing the crystal form A of the hydrochloride of
the
compound of Formula I-1 as described in the invention, comprising the steps
of:
(a) after mixing the compound of formula I-1 with ethyl acetate, adding
hydrochloric
acid dropwise at 5-15 C to adjust the pH of the system to 6-8, reacting and
precipitating a
solid, and filtering to obtain the crystal form A of the hydrochloride of the
compound of
Formula I-1.
In another preferred embodiment, in the step (a), the reaction time is 3-8
min,
preferably 5 min.
In another preferred embodiment, in the step (a), the weight-volume ratio (kg:
L) of
the compound of Formula I-1 to the organic solvent is 0.2-2: 2-30, preferably
0.4-1.0: 5-18,
more preferably 0.5-0.9: 8-15.
The crystal form A of the hydrochloride of the compound of Formula I-1 of the
present invention can inhibit TRPAl.
Compositions and methods of administration
The invention provided a composition for inhibiting the activity of transient
receptor
potential channel proteins. The compositions include, but are not limited to,
pharmaceutical compositions, food compositions, dietary supplements, beverage
compositions, etc.
Typically, the composition is a pharmaceutical composition comprising the
compound of Foimula I, Formula Z, Foimula G, Formula A or Formula B, or the
crystal
form A of the hydrochloride of the compound of Formula I-1 as described in the
present
invention; and pharmaceutically acceptable carriers.
¨ 36 ¨
Date Recue/Date Received 2021-11-12

In the present invention, the dosage forms of pharmaceutical compositions
include
(but are not limited to) oral preparations, injections and topical
preparations.
Typically, the dosage forms comprise (but are not limited to): tablets,
capsules,
injections, infusions, ointments, gels, solutions, microspheres and films.
The term "pharmaceutically acceptable carrier" means one or more compatible
fillers
in solid, semi-solid, liquid or gel form, which is suitable for human or
animal use and has
sufficient purity and low enough toxicity. The "compatible" means the
components and
the active ingredient of a pharmaceutical composition can be blended with each
other
without significantly reducing the efficacy.
It should be understood that in the present invention the carrier is not
particularly
limited, and materials commonly used in the field can be selected, or it can
be
manufactured by conventional methods, or it is commercially available.
Examples of
pharmaceutically acceptable carriers include cellulose and its derivatives
(such as methyl
cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, sodium
carboxymethyl
cellulose, etc.), gelatin, talc, and solid lubricants (such as stearic acid,
magnesium
stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil,
peanut oil, olive
oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol,
etc.), emulsifiers
(such as TweenTm), wetting agents (such as sodium lauryl sulfate), buffering
agents,
chelating agents, thickening agents, pH adjusters, penetration enhancers,
coloring agents,
flavoring agents, stabilizers, antioxidants, preservatives, bacteriostatic
agent, pyrogen-free
water, etc.
Typically, in addition to the active pharmaceutical ingredients, the liquid
dosage
form may contain inert diluents conventionally used in the art, such as water
or other
solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol,
ethyl carbonate,
ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide and oils,
especially
cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame
oil, or mixtures
thereof. In addition to these inert diluents, the composition may also contain
auxiliaries
such as wetting agents, emulsifiers, suspending agents, and the like.
Pharmaceutical preparations should match the mode of administration. The
medicament of the present invention may also be used with other synergistic
therapeutic
agents (including before, during or after use). When a pharmaceutical
composition or
preparation is used, a safe and effective amount of the drug is administered
to a subject in
need (such as a human or non-human mammal). The specific dose should consider
the
route of administration, patient's health and other factors, which are within
the skill range
of skilled doctors.
The main advantages of the present invention include:
(a) The present invention provided a class of compounds of Formula I, Formula
Z,
Formula G, Formula A or Formula B with novel structures and excellent TRP
channel (in
particular TRPA1) inhibitory activity.
(b) The compounds of the present invention exhibit potent analgesic effects in
variety
of animal models.
(c) The compounds of the present invention have less toxicity and higher
activity, and
therefore have a larger safety window.
(d) The compounds of the present invention have good druggability.
(e) The compounds of the present invention have excellent pharmacokinetic
properties.
(f) The compounds of the present invention are suitable for oral
administration.
(g) The invention also provided a crystal form A of the hydrochloride of the
compound of Formula I-1, which is in solid form, and the salt crystal form of
the
compound of Formula I-1 in solid form is convenient for storage,
transportation, and has
good druggability and stability (especially excellent thermal stability and
high humidity
¨37¨
Date Recite/Date Received 2023-04-03

CA 03140231 2021-11-12
stability) compared to the oil form of the free compound of Formula I-1.
The present invention will be further explained below in conjunction with
specific
embodiments. It should be understood that these embodiments are only used to
illustrate
the present invention and not to limit the scope of the present invention. In
the following
examples, the test methods without specific conditions are usually in
accordance with
conventional conditions or the conditions recommended by the manufacturer.
Unless
otherwise specified, percentages and parts are percentages by weight and parts
by weight.
Example 1
(R)-7-(3-chloro-1-(thiophen-2-yl)propoxy)benzofuran (Intermediate II-1)
0 CI
Io
528 mg of (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol, 400 mg of
7-hydroxybenzofuran and 862 mg of triphenylphosphine were dissolved in 30 ml
of
anhydrous tetrahydrofuran, and 667 mg of diisopropyl azodicarboxylate was
added
dropwise slowly to the system under ice bath, after the addition, then the
reaction system
was transfered to room temperature to react overnight. After completion of the
reaction,
the system was spin-dried directly, and the residue was separated and purified
by column
chromatography to obtain 685 mg Intermediate II-1 as a colorless oil, yield
78.46%.
1H NMR (500 MHz, CDC13) 6 7.62 (t, J= 3.2 Hz, 1H), 7.41 (dd, J= 1.8, 0.6 Hz,
1H), 7.24
(dt, J= 8.1, 1.8 Hz, 1H), 7.15 -7.11 (m, 1H), 6.91 (d, J= 7.7 Hz, 1H), 6.77
(dd, J= 8.0, 2.2 Hz,
1H), 6.35 (d, J= 3.3 Hz, 1H), 6.33 (dd, J= 3.3, 1.9 Hz, 1H), 5.75 (dd, J= 8.4,
5.1 Hz, 1H), 3.93
(dd, J= 11.1, 8.2, 5.4 Hz, 1H), 3.77- 3.70 (m, 1H), 2.85 - 2.74 (m, 1H), 2.54 -
2.48 (m, 1H).
MS (ESI, m/z): 292.93 (M+H)+.
Example 2
(R)-3-(benzofirran-7-yloxy)-N-methyl-3-(thiophen-2-yl)propan-1-amine (Compound
I-1)
0
(R)
0
I-1
685 mg of Intermediate II-1 was dissolved in a saturated sodium iodide
solution in acetone
and refluxed overnight. After completion of the reaction, the solvent was spin-
dried. Then water
was added into the reaction system, extracted with ethyl acetate for three
times, washed with
saturated salt water, dried over anhydrous sodium sulfate, filtered, and
concentrated. The
residue was dissolved in 30 mL tetrahydrofuran, 3 mL 40% methylamine aqueous
solution was
added to react overnight. After completion of the reaction, the solvent was
spin-dried, sodium
hydroxide solution was added into the system, extracted with ethyl acetate for
three times,
washed with saturated salt water, dried over anhydrous sodium sulfate,
filtered and
concentrated. The residue was separated by column chromatography
(methanol/dichloromethane= 1:15) to obtain 336 mg Compound I-I as a colorless
oil, yield
49.97%.
1H NMR (500 MHz, DMSO) 6 7.97 (d, J = 2.1 Hz, 1H), 7.49 (dd, J = 5.0, 1.1 Hz,
1H),
7.25 -7.18 (m, 2H), 7.08 (t, J = 7.9 Hz, 1H), 6.99 (dd, J = 5.0, 3.5 Hz, 1H),
6.96 (d, J = 7.9 Hz,
1H), 6.94 (d, J = 2.1 Hz, 1H), 6.05 (dd, J = 7.9, 5.2 Hz, 1H), 3.15 - 2.96 (m,
2H), 2.57 (s, 3H),
2.49 - 2.43 (m, 1H), 2.33 - 2.25 (m, 1H). MS (ESI, m/z): 288.0 (M+H)+.
-38 -
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
Example 3
(R)-2-(3-(benzofuran-7-yloxy)-3-(thiophen-2-y 1)propyl)i soindoline-1,3 -di
one
(Intermediate III)
o
R=== =N
0 0
iuiic
425 mg of intermediate II-1, 807 mg of phthalimide potassium salt and 100 mg
of sodium
iodide were dissolved in 15 ml of N,N-dimethylfoiiiiamide, and reacted at 90
C under nitrogen
overnight. After completion of the reaction, water was added to the system,
extracted with ethyl
acetate for three times, washed with water, washed with saturated brine, dried
over anhydrous
sodium sulfate, filtered, and concentrated. The residue was separated by
column
chromatography (ethyl acetate/petroleum ether = 1: 5) to obtain Intermediate
III, 412 mg of
yellow solid, with a yield of 70.35%.
111 NMR (500 MHz, CDC13) 6 7.84 ¨ 7.80 (m, 2H), 7.70 (dd, J= 5.4, 3.1 Hz, 2H),
7.50 (d,
J= 2.0 Hz, 1H), 7.21 (dd, J= 5.1, 1.4 Hz, 1H), 7.18 (dd, J= 7.9, 0.9 Hz, 1H),
7.09(d, J= 3.1
Hz, 1H), 7.08 ¨7.03 (m, 1H), 6.91 (dd, J= 10.1, 5.1 Hz, 1H), 6.82 (d, Jr 7.3
Hz, 1H), 6.71 (d,
J= 2.2 Hz, 1H), 5.85 (dd, J= 7.7, 5.4 Hz, 1H), 4.11 ¨ 3.92 (m, 2H), 2.68 (dd,
J= 14.4, 7.3 Hz,
1H), 2.53 ¨2.39 (m, 1H). MS (ESI, m/z): 403.99 (M+H) .
Example 4
(R)-3-(benzofuran-7-y loxy)-3-(thiophen-2-y 1)propan-1-amine (compound 1-2)
CI=ATNE12
0
1-2
412 mg of Intermediate III and 270 mg of hydrazine hydrate were dissolved in
15 ml of
methanol solution and reacted at room temperature overnight. After completion
of the reaction,
the solvent was spin-dried, and the residue was separated by column
chromatography
(methanol/dichloromethane = 1:15) to obtain Compound 1-2, 124 mg of colorless
oil, with a
yield of 44.42%.
1H NMR (500 MHz, DMSO) 8 7.87 (d, J = 2.3 Hz, 1H), 7.40 (dd, J = 5.5, 1.6 Hz,
1H),
7.19 ¨7.15 (m, 2H), 7.07 (t, J = 7.9 Hz, 1H), 6.94 (dd, J = 5.5, 3.8 Hz, 1H),
6.91 (d, J = 7.7 Hz,
1H), 6.87 (d, J = 2.4 Hz, 1H), 6.04 (m, 1H), 2.94 ¨ 2.80 (m, 2H), 2.37 ¨2.30
(m, 1H), 2.18 (dtd,
J= 11.8, 9.8, 5.1 Hz, 1H). MS (ESI, m/z): 273.98 (M+H)+.
Example 5
(R)-3-(benzofuran-7-y loxy)-N,N-dimethy1-3-(thi oph en-2-yl)propan-1 -amine
(compound
1-3)
(R)
N
0
T1 I
1-3
Except that the methylamine aqueous solution was replaced with dimethylamine,
the other
required raw materials, reagents and preparation methods were the same as
those in Example 2,
and 327 mg of colorless oily compound 1-3 was obtained, with a yield of
44.76%.
¨ 39 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
1H NMR (500 MHz, CDC13) 6 7.74 (d, J = 2.5 Hz, 1H), 7.31 (dt, J = 12.8, 6.4
Hz, 1H),
7.19 (dd, J = 7.8, 0.9 Hz, 1H), 7.05 ¨ 7.01 (m, 2H), 6.91 (dd, J = 5.0, 3.5
Hz, 1H), 6.83 (dd, J =
6.8, 6.1 Hz, 1H), 6.73 (d, J = 4.2 Hz, 1H), 5.85 ¨ 5.77 (m, 1H), 2.53 ¨ 2.48
(m, 2H), 2.48 ¨ 2.40
(m, 1H), 2.26 (s, 6H), 216 (dt, J = 10.1, 4.9 Hz, 1H).MS (ESI, m/z): 302M1
(M+H) .
Example 6
(R)-3-(benzofuran-7-yloxy)-N-ethyl-3-(thiophen-2-yl)propan-1 -amine (Compound
1-4)
0
1-4
Except that the methylamine aqueous solution was replaced with ethylamine, the
other
required raw materials, reagents and preparation methods were the same as
those in Example 2,
and 478 mg of colorless oily compound 1-4 was obtained, with a yield of
45.90%.
1H NMR (500 MHz, CDC13) 6 7.67 (d, J = 2.4 Hz, 1H), 7.25 (t, J = 7.6 Hz, 2H),
7.15 ¨
6.89 (m, 2H), 6.85 (dd, J = 5.5, 3.9 Hz, 1H), 6.74 (d, J = 8.4 Hz, 1H), 6.66
(d, J =1.8 Hz, 1H),
5.81 (dd, J =9.2, 6.4 Hz, 1H), 3.25 (t, J = 8.7 Hz, 2H), 3.15 (q, J = 7.3 Hz,
2H), 2.78 ¨2.66 (m,
1H), 2.60 ¨ 2.49 (m, 1H), 1.51 (t, J = 7.3 Hz, 3H). MS (ESI, m/z): 302.10
(M+H) .
Example 7
(R)-3-(benzofuran-7-yloxy)-N-methyl-3-(thiophen-3-yl)propan-1-amine (compound
1-5)
(,)
C/N
0
1-5
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(S)-3-chloro-1-(thiophen-3-yl)propan-1-ol, the other required raw materials,
reagents and
preparation methods were the same as those in Example 1-2, and 313 mg of
compound 1-5 was
obtained as a colorless oil, with a yield of 32.71%.
1H NMR (500 MHz, CDC13) 6 7.63 (d, J = 2.7 Hz, 1H), 7.31 (dd, J = 5.5, 3.1 Hz,
1H),
7.27 (d, J = 2.7 Hz, 1H), 7.20 ¨ 7.14 (m, 2H), 7.07 (t, J = 8.4 Hz, 1H), 6.76
(t, J = 4.2 Hz, 1H),
6.73 (d, J = 8.1 Hz, 1H), 5.66 (dd, J = 7.7, 5.6 Hz, 1H), 2.95 ¨2.83 (m, 2H),
2.50 (s, 3H), 2.44 ¨
2.33 (m, 1H), 2.26 ¨ 2.14 (m, 1H). MS (ESI, m/z): 287.97(M+H) .
Example 8
(R)-3-(benzofuran-7-yloxy)-N-methy1-3-(5-methylthiophen-2-yl)propan-1-amine
(compound 1-6)
0
1-6
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(S)-3-chloro-1-(5-methylthiophen-2-yl)propan-1-ol, the other required raw
materials, reagents
and preparation methods were the same as those in Example 1-2, and 222 mg of
compound 1-6
was obtained as a colorless oil, with a yield of 26.42%.
1H NMR (500 MHz, CDC13) 6 7.61 (d, J = 2.0 Hz, 1H), 7.16 (dt, J = 10.7, 5.4
Hz, 1H),
¨ 40 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
7.09 (d, J = 7.9 Hz, 1H), 6.87 (d, J = 7.7 Hz, 1H), 6.81 (d, J = 4.4 Hz, 1H),
6.77 (dd, J = 7.0, 2.2
Hz, 1H), 6.60 ¨ 6.53 (m, 1H), 5.74 (dd, J = 7.7, 5.6 Hz, 1H), 2.94 ¨ 2.80 (m,
2H), 2.55 (s, 3H),
2.46 ¨ 2.37 (m, 4H), 2.26 ¨2.17 (m, 1H). MS (ESI, m/z): 302.01 (M+H)+.
Example 9
(R)-3-(benzofuran-7-yloxy)-3-(5-chlorothiophen-2-y1)-N-methylpropan-l-amine
(Compound 1-7)
0
1-7
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(S)-3-chloro-1-(5-chlorothiophen-2-yl)propan-1-ol, the other required raw
materials, reagents
and preparation methods were the same as those in Example 1-2, and 275 mg of
compound 1-7
was obtained as a colorless oil, with a yield of 30.15%.
1H NMR (500 MHz, CDC13) 8 7.63 (d, J = 2.1 Hz, 1H), 7.22 (d, J =8.1 Hz, 1H),
7.11 (d, J
= 7.7 Hz, 1H), 6.91 (dd, J = 13.4, 6.4 Hz, 2H), 6.82 (d, J = 4.2 Hz, 1H), 6.76
(d, J = 4.3 Hz, 1H),
5.70 (dd, J = 8.7, 6.0 Hz, 111), 3.11 ¨3.02 (m, 2H), 2.53 (s, 3H), 2.48 (dt, J
= 21.6, 7.2 Hz, 1H),
2.27 (ddd, J = 13.7, 12.0, 6.7 Hz, 1H).MS (ESI, m/z): 321.96 (M+H)+.
Example 10
(R)-3-(benzofuran-7-y loxy)-3-(furan-2-y1)-N-methylpropan-1-amine (Compound 1-
8)
cb
R)
0 N.--
0
1-8
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(S)-3-chloro-1-(furan-2-yl)propan-1-ol, the other required raw materials,
reagents and
preparation methods were the same as those in Example 1-2, and 167 mg of
compound 1-8 was
obtained as a colorless oil, with a yield of 15.55%.
1H NMR (500 MHz, CDC13) 8 7.56 (d, J = 2.8 Hz, 1H), 7.33 (d, J = 1.9 Hz, 1H),
7.17 (d,
J = 8.7 Hz, 1H), 7.01 (dd, J = 11.5, 6.3 Hz, 1H), 6.74 (d, J = 7.9 Hz, 1H),
6.67 (d, J = 2.5 Hz,
1H), 6.26 (d, J = 4.3 Hz, 1H), 6.15 (dd, J = 4.2, 2.1 Hz, 1H), 5.56 (dd, J =
8.4, 6.7 Hz, 1H), 3.11
¨ 2.96 (m, 2H), 2.64 ¨ 2.55 (m, 4H), 2.40 (dd, J =12.4, 7.0 Hz, 1H). MS (ESI,
m/z): 272.02
(M+H)+.
Example 11
(R)-3-((2,3-dihydro-1H-inden-4-ypoxy)-N-methyl-3-(thiophen-2-y ppropan-l-amine
(Compound 1- 9)
R)
0
1-9
Except that 7-hydroxybenzofuran was replaced with 4-indanol, the other
required raw
materials, reagents and preparation methods were the same as those in Example
1-2, and 116
mg of compound 1-9 was obtained as a colorless oil, with a yield of 20.75%. 1H
NMR (500
¨41 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
MHz, CDC13) ö 7.27 (dd, J = 4.0, 2.1 Hz, 1H), 7.19 ¨ 7.16 (m, 2H), 6.99 (t, J
= 8.4 Hz, 1H),
6.88 (d, J = 7.7 Hz, 1H), 6.69 (d, J = 8.5 Hz, 1H), 5.68 (dd, J = 7.9, 4.8 Hz,
1H), 2.94 (dd, J =
18.69, 9.1 Hz, 4H), 2.87 (ddd, J =10.0, 8.5 Hz, 4.0 Hz ,2H), 2.47 (s, 3H),
2.35 ¨ 2.240 (m, 1H),
2.22 ¨ 2.16 (m, 1H), 2.12-2.04(m, 2H). MS (ESI, m/z): 288.03 (M+H)+.
Example 12
(R)-N-methyl-3 -((5,6,7,8-tetrahy dronaphthalen-1-yl)oxy )-3-(thi ophen-2-
yl)propan-1-ami n
e (Compound I -10)
0
1-10
Except that the 7-hydroxybenzofuran was replaced with tetrahydronaphthol, the
other
required raw materials, reagents and preparation methods were the same as
those in Example
1-2, and 103 mg of compound I-10 was obtained as a colorless oil, with a yield
of 25.17%.
1H NMR (500 MHz, CDC13) Ei 7.32 (d, J = 2.2 Hz, 1H), 7.22 ¨ 7.18 (m, 1H), 6.99
¨
6.95(m, 2H), 6.74 (d, J = 8.5 Hz, 1H), 6.63 (d, J = 8.8Hz, 1H), 4.88 (dd, J =
12.9, 4.4 Hz, 1H),
2.98 ¨ 2.74 (m, 5H), 2.66 ¨ 2.43 (m, 5H), 2.15-2.09 (m, 1H), 1.91 ¨ 1.76 (m,
4H).MS (ESI,
m/z): 302.0 (M+H) .
Example 13
(R)-3-(benzofuran-4-yloxy)-N-methy1-3-(thiophen-2-yl)propan-1-amine (compound
I-11)
I-11
Except that the 7-hydroxybenzofuran was replaced with 4-hydroxybenzofuran, the
other
required raw materials, reagents and preparation methods were the same as
those in Example
1-2, and 95 mg of compound 1-11 was obtained as a colorless oil, with a yield
of 14.06%.
1H NMR (500 MHz, CDC13) 7.49 (d, J = 2.8 Hz, 1H), 7.16 (d, J= 5.0, 1H), 7.11
¨7.04
(m, 3H), 6.89 (dd, J= 5.2, 3.2 Hz, 2H), 6.74 ¨ 6.65 (m, 1H), 5.81 (dd, J= 7.5,
5.0 Hz, 1H), 3.16
(t, J= 7.4 Hz, 2H), 2.76 ¨ 2.67 (m, 1H), 2.65 (s, 3H), 2.62 ¨ 2.51 (m, 1H). MS
(ESI, m/z):
287.97 (M+H)+.
Comparative example 1
(S)-3-(benzofuran-7-yloxy)-N-methy1-3-(thi oph en-2-yl)propan -1-amine
(compound
Cl)
(s)
0
Cl
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(R)-3-chloro-1-(thiophen-2-yl)propan-l-ol, the other required raw materials,
reagents and
preparation methods were the same as those in Example 1-2, and 199 mg of
compound Cl
was obtained, with a yield of 39.96%.
NMR (400 MHz, CDC13) 6 7.63 (d, J= 2.0 Hz, 1H), 7.20 (t, J= 6.6 Hz, 2H), 7.08
¨
6.99 (m, 2H), 6.88 (dd, J= 4.9, 3.6 Hz, 1H), 6.80 (d, J= 7.9 Hz, 1H), 6.75 (d,
J= 2.0 Hz, 1H),
¨42 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
5.93 (dd, J = 8.2, 4.4 Hz, 1H), 3.30 (t, J = 7.0 Hz, 2H), 2.82¨ 2.69 (m, 4H),
2.65 ¨2.54 (m, 1H).
MS (ESI, m/z): 287.87 (M+H)+.
Comparative example 2
(R)-3-(benzofuran-7-yloxy)-N-methy1-3-phenylpropan-1-amine (compound C2)
(R)
0
0
C2
Except that the (S)-3-chloro-1-(thiophen-2-yl)propan-1-ol was replaced with
(S)-3-chloro-1-phenylpropane-1-ol, the other required raw materials, reagents
and
preparation methods were the same as those in Example 1-2, and 147 mg of
compound C2
was obtained, with a yield of 24.42%.
1H NMR (500 MHz, CDC13) 6 7.64 (d, J= 2.1 Hz, 1H), 7.45 ¨ 7.40 (m, 2H), 7.32
(dd, J=
10.3, 4.8 Hz, 2H), 7.24 (dt, J = 2.4, 1.6 Hz, 1H), 7.12 (dd, J = 7.8, 0.8 Hz,
1H), 6.96 (dd, J =
10.4, 5.3 Hz, 1H), 6.74 (d, J= 2.1 Hz, 1H), 6.63 (d, J = 7.7 Hz, 1H), 5.49
(dd, J = 8.3, 4.8 Hz,
1H), 2.90 ¨ 2.80 (m, 2H), 2.46 (s, 3H), 2.39 ¨2.29 (m, 1H), 2.16 ¨2.06 (m,
1H). MS (ESI, m/z):
282.26 (M+H)+.
Example 14
TRPA1 inhibitory activity test
In this example, the compounds prepared in some examples of the present
invention
(shown in Table 1) were tested for their inhibitory activity on the transient
receptor
potential channel protein TRPA1. Wherein, compound of formula A (W02010075353)
was used as positive control:
0
NH¨i4
NLD
Formula A
The method was as follows:
Test method by using IonWorks Barracuda (IWB) automatic patch clamp detection:
HEK293 cells stably expressing TRPA1 were placed in a 37 C, 5% CO2 incubator
and
incubated with DMEM medium containing 15 ig/mL Blasticidin S HC1, 200 [tg/mL
Hygromycin B and 10% FBS serum in a T175 culture flask. When the cell density
grew to
¨80%, the culture medium was removed, the cells were washed with calcium and
magnesium free phosphate buffered saline (PBS), and 3 mL of Trypsin was added
and
digested for 2 minutes, and 7 mL of culture medium was added to terminate the
digestion.
The cells were collected into a 15 mL centrifuge tube and centrifuged at 800
r/min for 3
minutes, after removing the supernatant, the cells were added into the
extracellular fluid
with appropriate volume to re-suspend, so that the cell density was controlled
at 2-3 X
106/mL for IWB experiments. Extracellular fluid formulation (in mM):140 NaCl,
5 KCl,
1 MgCl2, 10 HEPES, 0.5 EGTA, 10 Glucose (pH 7.4); Intracellular fluid
formulation (in
mM):140 CsCI, 10 HEPES, 5 EGTA, 0.1 CaCl2, 1 MgCl2 (pH 7.2). Amphotericin B
was
¨43 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
freshly prepared with DMSO to 28 mg/mL on the day of the experiment, and then
formulated into a final concentration of 0.1 mg/mL with the intracellular
fluid.
The IWB experiment used a population patch clamp (PPC) plate, and the entire
detection process was automatically completed by the instrument, that is,
extracellular
fluid was added into the 384 wells of the PPC plate, and the intracellular
fluid was added
into the plenum which was under the PPC plate, and then 6 L of cell fluid was
added for
sealing test, and finally the intracellular fluid in plenum was replaced with
amphotericin
B-containing intracellular fluid, so that the sealed cells were perforated to
form a
whole-cell recording mode. The sampling frequency for recording the TPRA1
current was
10 kHz, the cell was clamped at 0 mV, and the voltage stimulation command
(channel
protocol) was a ramp voltage from -100 mV to +100 mV for 300 ms, the voltage
stimulation was given every 10 s, and the mTRPA1 current was induced via 300 M
AITC.
Data recording and current amplitude measurement and export were completed by
IWB Software (version 2.5.3, Molecular Devices Corporation, Union City, CA).
The
statistics of the well with a sealing impedance lower than 20 M SI were not
recorded. The
original current data was corrected by software, the TRPA1 current amplitude
was
measured in +100 mV. Every PPC plate of the experiment would have one HC030031
dose-effect data as positive control, for example, when IC50 value of a
HC030031 was
higher than 3 times of the average of that obtained on each preceding plate, a
retest would
be carried out. The dose-effect curve and IC50 of compounds were fitted and
calculated by
GraphPad Prism 5.02 (GraphPad Software, San Diego, CA).
Experimental results
A part of the compounds prepared in the examples of the present invention were
tested for IC50 inhibitory activity by the test method of IonWorks Barracuda
(IWB)
automatic patch clamp detection. The activity data was shown in Table 2.
Table 2. Inhibitory activity data (IC50, [tM) of part of compounds of the
present invention
against TRPA1 in automatic patch clamp detection
Number IC so (gM) Number IC so( M) Number IC so (11M)
Compound Compound
+++++++++ Compound 1-3 +++++
I-1 1-2
Compound Compound
+++++ +++++ Compound 1-6 ++++
1-4 1-5
Compound Compound Compound
+++++++++ ++++
1-8 1-9 1-11
Compound
C2 Formula A
+++ R-Duloxetine
(Comparative Compound
Example 2)
In which, the activity: IC5o(p. M):
51-100: +
21-50: ++
11-20: +++
6-10: ++++
1-5: +++++
The results showed that the compounds of the present invention showed a strong
inhibitory activity against TRPA1. Among them, 5 compounds have a half
effective
inhibitory concentration IC50 against TRPA1 between 1-5 [tM, and 4 compounds
have a
half effective inhibitory concentration IC50 against TRPA1 between 6-10[tM. As
shown in
Fig. 1, the compound I-1 of the present invention has an inhibitory activity
IC50 of 2.06
¨ 44
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
1tN1 against TRPA1. Therefore, it can be concluded that the compound of
formula I-1 of
the present invention has a strong inhibitory activity against TRPA1.
In addition, the activity ratio of compound I-1 (containing heteroaryl) and
comparative compound C2 (containing phenyl) (IC50 of compound C2/IC50 of
compound
I-1) is about 6.3, which shows that the compound containing heteroaryl of the
present
invention (such as I-1) has higher inhibitory activity against TRPA1.
Compared with compounds in which the A group is a benzene ring (such as
R-duloxetine), IC50 value of compound I-1, compound 1-3, compound 1-4,
compound 1-5,
compound 1-8 and compound 1-9 are significantly lower. The ratio of the IC50
of
R-duloxetine to the IC50 of any one of compound I-1, compound 1-3, compound 1-
4,
compound 1-5, compound 1-8 and compound 1-9 is about 9.3¨ 23.5. This indicates
that the
compounds of the present invention in which the A group is an alicyclic or
heteroaryl have
higher inhibitory activity against TRPA1.
The inventors measured the TRPA1 inhibitory activity of compound 1-1 via the
manual patch clamp detection method, as shown in Fig. 2, which is similar to
the results of
the automatic patch clamp detection method. In the manual patch clamp
detection method,
the IC50 of compound I-1 is 0.421.tM, which shows a strong inhibitory activity
against
TRPA1.
Example 15
Cytotoxicity test
In this example, the hepatocyte toxicity and neurocytotoxicity of compound I-1
and
S-duloxetine were tested.
1. Hepatocyte toxicity and neurocytotoxicity of compound 1-1.
HepG-2 and SH-SY5Y cells were prepared and placed in 10 cm dish and incubated
at 37 C, 5% CO2 in a cell incubator; the cells were digested with trypsinize,
resuspended
and then counted. Based on a system of 100111/well, 8000 cells were
transferred to a
96-well plate. The cells were incubated in a 37 C, 5% CO2 cell incubator for
24 hours; a
gradient concentration system of compound I-1 (prepared in Example 2) was
prepared, 2
times diluted, the system was 100 p1/well. The supernatant in the 96-well
plate cell culture
system on the first day was removed, and a fresh prepared drug concentration
system was
added to the corresponding culture plate wells culturing cells (set up
duplicate wells). The
cells were cultured in 5% CO2 incubator at 37 C for 72 hours. After
completion of the
culture, the supernatant was removed from the cell culture system of the 96-
well plate, 100
pi of detection solution (medium containing 10% CCK-8) was add to each well,
and
incubated in a cell incubator at 37 C and 5% CO2 for 1 hour, then taken it out
and
measured the absorbance at 450 nm with a microplate reader. Data were
processed,
cytotoxicity was calculated, and IC50 was calculated by GraphPad Prism. The
equation for
calculating cytotoxicity is as follows: cytotoxicity (%) = [A (0 dosing)-A
(dosing)14A (0
dosing)-A (blank)] x 100
A (dosing): absorbance of the well having cells, CCK-8 solution and drug
solution
A (blank): absorbance of the well having medium and CCK-8 solution, without
cells.
A (0 dosing): absorbance of the well having cells and CCK-8 solution, without
drug
solution
2. Hepatocyte toxicity and neurocytotoxicity of S-duloxetine.
The test method was similar to the above hepatocyte toxicity and
neurocytotoxicity
of the compound I-1, except that compound I-1 was replaced with S-duloxetine.
Results
The results of the hepatotoxicity (HepG2 cells) and neurocytotoxicity (SH-
SY5Y) of
compound I-1 are as follows:
¨45 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
The hepatotoxicity and neurocytotoxicity (IC50, [NI) of S-duloxetine are 33.33
p,M
and 28.59 NI, respectively, while the hepatotoxicity and neurotoxicity (IC5o,
uM) of
compound I-1 of the present invention are about 113.80 uM and 100.70 M,
indicating
that the compounds of the present invention have a significantly lower toxic
effect and
excellent safety.
EXAMPLE 16
The therapeutic effect of compound I-1 on acute pain and inflammatory pain
were
investigated through formalin pain model in mice
Experimental method
150 C57BL/6 mice (male, 9 weeks) were randomly divided into 15 groups with 10
mice for each group, and were used for the analgesic activity test of 3
compounds in the
formalin pain model in mice: the compound I-1 group (compound I-1 prepared in
Example
2, its hydrochloride was used in the experiment), the S-duloxetine group (its
hydrochloride
was used in the experiment) and the compound Cl group (compound Cl prepared in
Comparative Example 1, its hydrochloride was used in the experiment),
respectively.
Before the start of the experiment, the mice were allowed to adapt to the
experimental
environment for 72 hours, during which there was no need to fast for food or
water. The
tested drugs were administrated by intraperitoneal injection, and the doses
were as
follows:
The compound I-1 group: blank Vehicle (blank normal saline control), 0.1
mg/kg, 0.5
mg/kg, 1 mg/kg, 5 mg/kg and 10 mg/kg;
The S-duloxetine group: blank Vehicle (blank normal saline control), 1 mg/kg,
5
mg/kg, 10 mg/kg and 20 mg/kg;
The Compound Cl group: blank Vehicle (blank normal saline control), 0.1 mg/kg,
0.5 mg/kg, 1 mg/kg, 5 mg/kg and 10 mg/kg.
After the administration, the mice were placed in a transparent, ventilated
plexiglass
cylinder, and 1 hour later, 20 ul of 4% formalin solution was injected into
the left hind
foot plantar of each group of mice with a micro-injector, and foot pain
response in mice
were recorded in real time with a miniature camera. The length of time of
licking the left
foot was used as an indicator of pain response, the licking time were observed
and
recorded during two periods of 0-10 min (phase I) and 10-60 min (phase II)
respectively,
for statistical analysis, and the half effective dose (ED50) of the 3
compounds were
calculated: ED50 refers to the drug dose that reduces the licking time by half
compared
with the blank control group. The smaller the ED50 value is, the lower the
effective
analgesic dose of the compound is and the better its analgesic effect is.
Results
The test results of formalin pain model in mice are shown in Table 3 and Fig.
3. As
can be seen in Table 3 and Fig. 3, the analgesic activity of all three
compounds showed a
dose dependence. The compound I-1 of the present invention had a phase 11 (10-
60 min)
licking time that was already reduced by more than 50% compared to the blank
Vehicle at
an administered dose of 0.5 mg/kg, with an analgesic potency ED50 of 0.26
mg/kg in phase
II pain. The ED50 of S-duloxetine in phase II pain was 8.00 mg/kg, and the
ED50 of
comparative compound Cl in phase II pain was 2.22 mg/kg. From the above data,
it can be
seen that the compound I-1 of the present invention exhibits extremely strong
analgesic
activity in the formalin pain model in mice, and its ED50 is 30.8 times
stronger than that of
S-duloxetine, and 8.5 times stronger than that of Cl compound. The fonnalin
model in
mice is a classic model for evaluating the efficacy of drugs for acute pain
and
inflammatory pain, and therefore the compound I-1 of the present invention has
excellent
therapeutic effects on acute pain and inflammatory pain.
Table 3. statistical results of the licking time of the compound I-1 of the
present invention,
¨46 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
S-duloxetine and comparative compound Cl in the formalin model in mice at
different
doses in phase II (10-60min)
D statistics of the licking time (s)
ose
Compound I-I S-duloxetine Compound Cl
Blank Vehicle 349.55 43.09 349.55
43.09 349.55 43.09
0.1 mg/kg 321.55 47.75 339.35
27.90
0.5 mg/kg 161.25+30.83
368.16+44.46
1 mg/kg 131.28+23.99
259.97+64.44 171.57+39.69
mg/kg 87.85 25.15 230.49
41.31 158.02 25.18
mg/kg 99.08 19.73 162.83
36.27 35.68 11.77
mg/kg 72.27+17.29
Example 17
5 The
therapeutic effect of compounds I-1 on acute pain was investigated through hot
plate induced pain model in rat
Experimental method:
Male, mature and unmated healthy Sprague-Dawley rats were selected, and the
10 temperature of the cold/hot plate (product model: PE34, US IITC) was
adjusted to a
constant temperature of 53+0.1 C, and rats with painful response such as foot
licking,
foot shaking or slight jumping within 5-10 s were screened (abandoned those
who evaded
and jumped). The 50 animals screened were weighed and randomly divided into 5
groups
(10 rats in each group): normal saline control group (Vehicle, blank control),
S-duloxetine
15 group
(its hydrochloride salt was used in the experiment), gabapentin group,
comparative
compound Cl group (compound Cl prepared in Comparative Example 1, and its
hydrochloride salt was used in the experiment) and compound I-1 group
(compound I-1
prepared in Example 2, and its hydrochloride salt was used in the experiment).
The tested
compound was freshly formulated on the day of administration. A 0.9% NaCl
normal
20 saline
solution was prepared as a vehicle, an appropriate amount of the test compound
was
added to the required volume of normal saline, and the mixture was fully
suspended to
prepare the drug at a concentration of 1 mg/ml. The standard of dose in volume
to rats was
10 ml/kg via intraperitoneal administration, animals did not need to fast food
or water
before administration. The dose of S-duloxetine, compound Cl and compound I-I
were 30
mg/kg, and the dose of gabapentin was 100 mg/kg. The latency of thermal pain
was
measured at 0.5 h, 1 h and 2 h after drug administration. To avoid scalding
the animals on
the hot plate, the maximum latency was set to 30 s. The analgesic effect of
each compound
was evaluated using the maximum possible effect (MPE), i.e. MPE%=[(Post drug
latency-baseline latency)/(30- baseline latency)] x 100. Statistics of MPE% at
different
time points. The higher the value of MPE% is, the more potent the analgesic
efficacy of
the compound is.
Results
The results of analgesic activity of compounds in hot plate induced pain model
in rat
are shown in Table 4 and Fig.4. It can be seen from the results in Table 4 and
Fig.4,
compound I-1 of the present invention exhibits a very potent analgesic effect
at an
administered dose of 30 mg/kg compared with the normal saline control group,
with
significant differences. Compared with the positive control, the analgesic
activity within 2
hours of compound I-1 of the present invention is significantly superior to
that of
gabapentin at 100 mg/kg, and superior to that of S-duloxetine and comparison
compound
Cl at 30 mg/kg. The hot plate induced pain model is a classic model for
evaluating the
efficacy of drugs for acute pain, and therefore the compound I-1 of the
present invention
¨47 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
has excellent therapeutic effect on acute pain.
Table 4. MPE% statistics at different times for compound I-1 of the present
invention,
S-duloxetine, comparison compound Cl and gabapentin in the hot plate induced
pain
model in rat
MPE%
Compound Dose
0.5h 1 h 2h
normal saline group 5.15+4.31 -2.31+1.95 -
3.39+2.47
Compound
30 mg/kg 14.51+6.87 38.03+5.66
31.92+6.79
I-1
S-duloxetine 30 mg/kg 5.75+5.74 7.44+5.12
13.53+4.99
Compound
30 mg/kg 5.29+1.56 14.20+5.71
21.83+2.45
Cl
Gabapentin 100 mg/kg 6.47+5.49 14.56+6.18
22.66+4.93
Example 18
The therapeutic effect of compound I-1 on visceral pain and inflammatory pain
was
investigated through acetic acid writhing pain model in mice
Experimental method
ICR mice, male, 22-25g, were fasted for food 2h before administration, but can
have
water. All ICR mice were weighed and randomly grouped with the number of
animals >10
per group. The negative control group was a normal saline group (Vehicle,
blank control),
and the positive control groups were set to a dose of 10 mg/kg indomethacin (a
non-steroidal anti-inflammatory drug), a dose of 10 mg/kg Anisodamine (an
antispasmodic drug with clinically analgesic activity), a dose of 10 mg/kg S-
duloxetine
(its hydrochloride was used in the experiment) and a dose of 20 mg/kg S-
duloxetine (its
hydrochloride was used in the experiment). Test compound I-1 (compound I-1
prepared in
Example 2, its hydrochloride was used in the experiment, in a dose of 5 mg/kg
and 10
mg/kg). The mice were administered via intragastric administration according
to the
weight of mice. 1.5% acetic acid solution (0.1m1/10g) was intraperitoneally
injected at 1
hour after administration, and the number of times of visceral pain in each
group within 30
minutes was observed, the mice were counted once when the abdomen was concave,
the
trunk and hind legs were extended and the hips were elevated, and the number
of times of
these phenomena occurred within 30 min was finally counted. The less visceral
pain
occurred in mice after administration, which indicated that the analgesic
effect of the
compound was stronger.
Results
The acetic acid writhing pain model in mice was tested as shown in Fig. 5,
from
which it can be seen that by single intragastric administration of compound I-
1 (5 mg/kg
and 10 mg/kg) of the present invention significantly reduced the number of
acetic
acid-induced writhing reactions in mice, with a significant difference
compared with the
normal saline group (vehicle, blank control) (49 times). The number of
visceral pains in
mice at a dose of 5 mg/kg administered with compound I-1 was 19, which was 50%
lower
than the 49 in the normal saline control group, suggesting that the half
effective dose
(ED50) of compound I-1 in this model is less than 5 mg/kg. The analgesic
effect of
compound I-1 at a dose of 10 mg/kg (16 times) is superior to that of the
positive drugs
indomethacin (28 times), anisodamine (27 times) and S-duloxetine (27 times) at
the same
dose, and the analgesic effect of compound I-1 at a dose of 5 mg/kg (19 times)
is
¨48 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
comparable to that of S-duloxetine at 20 mg/kg (21 times). This experiment
showed that
the analgesic activity of compound I-1 of the present invention was
significantly better
than that of the positive control drug in acetic acid writhing pain model in
mice. The acetic
acid writhing pain model in mice is a classic model for evaluating the
efficacy of drugs in
the treatment of visceral pain and inflammatory pain, and therefore the
compound I-1 of
the present invention has excellent therapeutic effect on visceral pain and
inflammatory
pain.
Example 19
The therapeutic effect of compound I-1 on nerve pain was investigated through
rat
SNL model.
Experimental method
1. Surgery
SD rats were taken for surgery, male, SPF grade, mass 150 g-180 g. The
surgical
procedure was performed aseptically. The animals were anesthetized with sodium
pentobarbital (50 mg/kg, intraperitoneal injection). The surgical area of the
waist of
animals was shaved, and the skin was disinfected three times with iodophor and
70%
ethanol. Start the operation after the skin was dry. A longitudinal incision
was made in the
posterior part of the sacral bone of the animal's waist with a scalpel to
expose the left
paravertebral muscles, and the muscle tissue was separated with a distractor
to expose the
vertebra. The left spinal nerves L5 and L6 were separated, ligated with 6-0
silk thread, and
the wounds were sutured. After the surgery, the animals were placed on an
electric blanket
and injected subcutaneously with 5mL normal saline to prevent dehydration.
When the
animal was completely awake (free to move), put the animal back into the cage.
2. Grouping and mechanical hyperalgesia test
After the surgery, the animals were adapted in the experimental environment
for 15
minutes/day for 3 days. On the day before the administration, the baseline
test of
mechanical hyperalgesia was carried out on rats, and the animals without
mechanical
hyperalgesia (paw withdrawal threshold greater than 5g) were abandoned, then
the animals
.. were randomly divided into one control group and three experimental groups.
Administration:
The animal were weighed, in terms of administered dose, the three experimental
groups were given 100 mg/kg gabapentin, 10 mg/kg S-duloxetine (its
hydrochloride was
used in the experiment) and 10 mg/kg Compound I-1 (compound I-1 prepared in
Example
2, and its hydrochloride was used in the experiment) via intragastric
administration,
respectively, and the control group was given an equal volume of nonnal saline
via
intragastric administration. After administration, mechanical hyperalgesia
test was
performed. The rats were placed individually in a plexiglas box with a grid at
the bottom
of the box to ensure that the feet of the rats can be tested. Rats would adapt
for 15 minutes
before the test. After the adaptation was completed, the test fiber was used
to test rat at the
center of the sole of the left hind foot. The test fibers included 8 test
strengths: 3.61 (0.4 g),
3.84 (0.6 g), 4.08 (1g), 4.31 (2g), 4.56 (4g), 4.74 (6g), 4.93 (8g) and 5.18
(15g). For the
test, the test fiber were pressed vertically against the skin and force was
applied to bend
the fiber for 6-8 seconds, with a 5 seconds interval between each test. The
rapid paw
withdrawal of the animal during the test was recorded as a pain response. The
paw
withdrawal of the animal when the test fiber left the animal's skin was also
recorded as a
pain response. If the animal moved or walked, it will not be recorded as a
pain response,
the test should be repeated. The test was firstly performed with 4.31 (2 g),
and if the
animal had a pain response, the next test was performed with a test fiber of a
lower
.. strength; if the animal did not have a pain response, the next test was
performed with a test
fiber of a higher strength. The maximum strength of the tested fiber was 5.18
(15 g).
Mechanical hyperalgesia was expressed as the paw withdrawal threshold (PWT) in
¨49 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
the behavioral test of rats, which was calculated according to the following
equation:
50% response threshold (g) = (10(xf' k611))/10,000
Xf = the final test fiber value used in the test
K = table value
8 = Average difference
Data were collected using Excel software and analyzed using Prism 6.01 (Graph
pad
software, Inc.) software. Higher values of the paw withdrawal threshold (PWT)
indicate
that the compound is more potent in analgesia.
Results
The results of analgesic activity in the SNL model in rat are shown in Table 5
and Fig.
6. It can be seen from the results of Table 5 and Fig. 6, the compound I-1 of
the present
invention exhibits a very potent analgesic effect at an administration dose of
10 mg/kg,
with a significant difference compared with the normal saline control group.
Compared
with the positive control group, the analgesic activity of the compound I-1 of
the present
invention is superior to that of 100 mg/kg gabapentin and 10 mg/kg S-
duloxetine at 1 hour
after administration. The SNL model in rat is a classic model for evaluating
the efficacy of
drugs in the treatment of nerve pain, and therefore the compound I-1 of the
present
invention has excellent therapeutic effect on nerve pain.
Table 5. Paw withdrawal threshold (PWT) statistics data of compound I-1 of the
present
invention, S-duloxetine and gabapentin in the SNL model in rat, 1 hour after
administration
Compound Dose PWT (g),
Control group 3.967 0.775
Compound I-1 10 mg/kg 7.869 2.846
S-duloxetine 10 mg/kg 6.519 2.226
Gabapentin 100 mg/kg 6.352 1.897
Example 20
Preparation and characterization of the hydrochloride A of the compound of
Formula
I-1 of the present invention.
XRPD: X-ray powder diffraction; DSC: Differential scanning calorimetry; TGA:
Thermogravimetric analysis; DVS: Dynamic water adsorption;
X-ray powder diffraction analysis method: PANalytical X-ray powder diffraction
analyzer, working voltage: 40kV, working current: 40mA, using Cu target to
obtain X-ray
powder diffraction pattern.
Differential scanning calorimetry (DSC): The instrument was DSC Q2000;
Scanning
speed: 10 C/min; Protective gas, nitrogen.
Thermogravimetric analysis (TGA): TGA Q500; Scanning speed: 10 C/min;
Protective gas: nitrogen.
Preparation method of the crystal form A of the hydrochloride of the compound
of Formula I-1:
0.73 kg of the free base of the compound of formula I-1 prepared in Example 2
was
weighed and added into 11L of ethyl acetate, stired, and cooled to 5-15 C with
an ice
water bath, slowly added 37% concentrated hydrochloric acid dropwise to adjust
the pH of
the system to 7, and the reaction was stirred for 5 minutes. A solid
precipitated out,
filtered, the filter cake was washed with ethyl acetate and then placed in an
oven (40-45 C)
and dried to a constant weight to obtain 0.45 kg the crystal form A of the
hydrochloride of
the compound of formula I-1, with a yield of 54.70%.
Identification of the crystal form A of the hydrochloride of the compound of
¨ 50 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
Formula I-1
The X-ray powder diffraction data of the crystal form A of the hydrochloride
of the
compound of Formula I-1 are shown in Table 6, and the XRPD pattern is shown in
Fig. 7,
having the following characteristic peaks expressed with 2 theta: 10.003,
11.171, 15.987,
16.734, 17.092, 18.173, 18.849, 20.681, 21.156, 21.649, 22.084, 22.794,
23.761, 25.298,
25.967, 26.640, 27.273, 28.099, 28.615, 28.813, 29.501, 30.118, 30.513,
32.522, 33.274,
34.081, 35.815, 37.553, 40.018, 42.927, 44.129.
Table 6
2 theta Relative
d value
value Intensity %
10.003 8.8355 28.3
11.171 7.9137 12.8
15.987 5.5392 15.2
16.734 5.2936 51.9
17.092 5.1836 35.7
18.173 4.8774 100.0
18.849 4.7041 12.7
20.681 4.2913 13.6
21.156 4.1961 65.2
21.649 4.1017 35.3
22.084 4.0217 71.9
22.794 3.8981 91.8
23.761 3.7416 65.0
25.298 3.5177 46.3
25.967 3.4285 11.5
26.640 3.3433 29.2
27.273 3.2672 16.9
28.099 3.1730 35.7
28.615 3.1170 33.1
28.813 3.0959 25.2
29.501 3.0253 10.4
30.118 2.9647 12.1
30.513 2.9272 13.2
32.522 2.7508 18.4
33.274 2.6904 12.4
34.081 2.6285 13.2
35.815 2.5051 13.4
37.553 2.3931 9.6
40.018 2.2512 8.2
42.927 2.1051 10.6
44.129 2.0505 9.0
The differential scanning calorimetry (DSC) pattern of the crystal form A of
the
hydrochloride is substantially as shown in Fig. 8, and the endothermic peak
begins to
appear upon being heated to 142.30 C.
The thermogravimetric analysis (TGA) pattern of the crystal form A of the
hydrochloride is substantially as shown in Fig. 9, with a weight loss of about
0.9827%
upon being heated to 168.01 C.
Investigation on the characteristics of the crystal form A of the
hydrochloride of
-51 -
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
the compound of formula I-1
(1) The hygroscopicity experiment of the crystal form A of the hydrochloride
of the
compound of formula I-1:
Hygroscopicity was tested by dynamic moisture absorption (DVS) instrument. The
crystal form A of the hydrochloride of the compound of formula I-1 has a
weight change
of less than 0.2% under 25 C/80RH conditions, indicating that it has no
hygroscopicity
and has excellent high humidity stability, which eliminates the need for
storage and
transportation under harsh dry conditions, thereby reducing storage and
transportation
costs. (Refer to the 2015 edition of the Chinese Pharmacopoeia (Guiding
Principles for
Drug Hygroscopicity Test)).
(2) Stability study of the crystal form A of the hydrochloride of the compound
of
formula I-1:
About 10 mg of the crystal form A of the hydrochloride of the compound of
formula
I-1 was weighed and added into an HPLC vial, the bottle was sealed with a
parafilm, 10
small holes were pierced in the film, and the vial was placed at 25 C/60%RH
or 40
C/75%RH environment for 4 weeks, sampled in the first and fourth weeks
respectively.
Purity (using HPLC analysis) and crystal form (using X-ray powder diffraction
analysis)
of the samples were investigated. The results are shown in Table 7. It can be
seen from
Table 7 that after the crystal form A of the hydrochloride of the compound of
foimula I-1
was placed for 1 week and 4 weeks, the HPLC (high performance liquid
chromatography)
purity does not decrease significantly and no change in crystal form is
observed,
indicating the crystal form A of the hydrochloride of the compound of formula
I-1 has a
good thermal and other stability in physical and chemical.
Table 7
25 C/60% RH 40 C/75% RH
Initial
Experimental Change Crystal
Crystal form purity Purity Purity
duration of crystal form
(area%) (area%) (area%)
form change
Crystal form 1 week 100 100
A of the 100 NO NO
4 weeks 100 100
hydrochloride
Example 21
The therapeutic effect of compound I-1 on postoperative pain was investigated
through postoperative pain model in rat
Experimental method
1. Drug preparation
Injection of compound I-1: 36.13 mg of compound I-1 prepared in Example 2 was
weighed and added into 3.140 mL of normal saline, and well mixed by vortex;
Injection of Bupivacaine: purchased from Shanghai Zhaohui Pharmaceutical Co.,
Ltd..
The solvent of the test compound I-1 was normal saline, the administration
dose was
10 mg/kg, and the administration method was intramuscular injection. 36.13 mg
of the
compound 1-1 was weighed and added into 3.140 mL normal saline, and well mixed
by
vortex. The reference compound used in the experiment was bupivacaine
injection,
purchased from Shanghai Zhaohui Pharmaceutical Co., Ltd., in a dose of 10
mg/kg via
intramuscular injection.
2. Surgery
Aseptic operation was performed during surgery, surgical instruments
(scissors,
¨52 ¨
Date Recue/Date Received 2021-11-12

CA 03140231 2021-11-12
forceps, scalpel, surgical cotton, suture thread) were disinfected before the
operation. The
rats were anesthetized with sodium pentobarbital (50 mg/kg, intraperitoneal
injection), and
the toes of the rats were squeezed to confirm that the animals were completely
anesthetized before the surgery. Ophthalmic ointment was applied to the
animal's eyes to
prevent the animal's cornea from drying out. Iodophor and 70% ethanol were
used to
disinfect the skin of the surgery area on the sole of the left hind foot three
times, and the
surgery was started after the skin was dried. Starting at 0.5 cm from the
heel, a 1 cm long
incision was made longitudinally toward the toe. After cutting the skin, the
flexor
digitorum brevis was lifted to make longitudinal blunt injury. After pressing
to stop the
bleeding, the wound was sutured. The surgical instruments werw cleaned and
sterilized
with hot bead sterilizer. After the surgery, the animals were placed on an
electric blanket
and injected subcutaneously with 5mL normal saline to prevent dehydration.
When the
animal was completely awake (free to move), put the animal back into the cage.
3. Grouping and administration
At 24 hours after the surgery, the baseline test of mechanical hyperalgesia
was
performed on rats. The animals that did not exhibit mechanical hyperalgesia
(PWT > 5 g)
were abandoned, then the animals were randomly divided into groups according
to PWT.
The experiment was divided into three groups: model control group (normal
saline group,
intragastric administration of normal saline), bupivacaine group
(intramuscular injection
of bupivacaine at a dose of 10 mg/kg) and compound I-1 group (intramuscular
injection of
compound I-1 at a dose of 10 mg/kg). The rats in each group were tested for
mechanical
hyperalgesia at 1 hour and 2 hours after the administration.
4. mechanical hyperalgesia test
The rat was placed alone in a plexiglass box with a grid on the bottom of the
box to
ensure that the rat's feet can be tested. Rats would adapt for 15 minutes
before the test.
After the adaptation was completed, the test fiber was used to test in the
center of the left
hind foot of the rat. The test fibers included 8 test strengths: 3.61 (0.4 g),
3.84 (0.6 g), 4.08
(1g), 4.31 (2g), 4.56 (4g), 4.74 (6g), 4.93 (8g) and 5.18 (15g). During the
test, the test
fiber was pressed vertically to the skin and force was applied to bend the
fiber for 6-8
seconds, with a test interval of 5 seconds. During the test, the animal's
rapid foot shrinkage
was recorded as a pain response. The animal's foot shrinkage when the test
fiber left the
animal's skin was also recorded as a pain response. If the animal moved or
walked, it
would not be recorded as the pain response, the test should be repeated. For
the test, fiber
4.31 (2 g) was used at first. If the animal had a pain response, the test
fiber with a lower
strength was used in the next test; if the animal had no pain response, the
test fiber with a
higher strength was used in the next test. The maximum strength of the tested
fiber was
5.18 (15 g). The test results are recorded in the table, with pain response
records x and no
pain response records o.
3.61
3.84
4.08
4.3111,"I'
4.56o o o
474x x x ¨
4.93
5.18
Mechanical hyperalgesia was expressed as the paw withdrawal threshold (PWT) in
the behavioral test of rats, which was calculated according to the following
equation:
50% response threshold (g) = (10 (Xf + k))/10,000
Xf = the final test fiber value used in the test
K = table value (Chaplan et al. 1994, page 62)
0= Average difference
¨ 53 ¨
Date Recue/Date Received 2021-11-12

5. Data collection and analysis
Excel software was used to collect data. Prism 6.01 (Graph pad software, Inc.)
software was used to analyze the data (two-way ANOVA plus Bonferroni multiple
comparison test).
Results
The results of the analgesic activity of compound I-1 in the postoperative
pain model
in rats are shown in Table 8 and Fig. 10. As can be seen from the results in
Table 8 and
Figure 10, compound I-1 of the present invention exhibits a very potent
analgesic effect at
a dose of 10 mg/kg at 1 h and 2 h after administration, with significant
differences
compared with the normal saline control group. Compared with the positive drug
bupivacaine, bupivacaine shows a stronger analgesic activity at 1 hour after
administration,
but no significant analgesic effect at 2 hours after administration,
suggesting that
compound I-1 has a longer analgesic effect in the postoperative pain model.
The
postoperative pain model in rat is a classic model for evaluating the efficacy
of drugs in
the treatment of postoperative pain, and therefore the compound I-1 of the
present
invention has excellent therapeutic effect on postoperative pain.
Table 8. paw withdrawal threshold (PWT) statistical data of Compound I-1 of
the present
invention and bupivacaine in the postoperative pain model in rat, 1 hour and 2
hours after
administration
PWT (g)
Compound
1 hour 2 hour
Normal Saline 3.561 3.382
Bupivacaine 11.587 4.448
Compound I-1 5.885 5.135.
Further, it should be understood that upon reading the above teaching of the
present
invention, various modifications or modifications may be made to the present
invention by
those skilled in the art, and those equivalents also fall within the scope
defined by the
appended claims of the present application.
¨54¨
Date Recite/Date Received 2023-04-03

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

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 , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Inactive: Grant downloaded 2024-06-04
Inactive: Grant downloaded 2024-06-04
Letter Sent 2024-06-04
Grant by Issuance 2024-06-04
Inactive: Cover page published 2024-06-03
Pre-grant 2024-04-25
Inactive: Final fee received 2024-04-25
4 2024-03-14
Letter Sent 2024-03-14
Notice of Allowance is Issued 2024-03-14
Inactive: Q2 passed 2024-03-07
Inactive: Approved for allowance (AFA) 2024-03-07
Amendment Received - Voluntary Amendment 2023-09-21
Amendment Received - Response to Examiner's Requisition 2023-09-21
Examiner's Report 2023-07-04
Inactive: Report - No QC 2023-06-07
Amendment Received - Voluntary Amendment 2023-04-03
Amendment Received - Response to Examiner's Requisition 2023-04-03
Examiner's Report 2022-12-06
Inactive: Report - No QC 2022-11-25
Inactive: Cover page published 2022-01-11
Request for Priority Received 2021-12-01
Priority Claim Requirements Determined Compliant 2021-12-01
Priority Claim Requirements Determined Compliant 2021-12-01
Letter sent 2021-12-01
Letter Sent 2021-12-01
Priority Claim Requirements Determined Compliant 2021-12-01
Application Received - PCT 2021-12-01
Inactive: First IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Inactive: IPC assigned 2021-12-01
Request for Priority Received 2021-12-01
Request for Priority Received 2021-12-01
Request for Examination Requirements Determined Compliant 2021-11-12
All Requirements for Examination Determined Compliant 2021-11-12
National Entry Requirements Determined Compliant 2021-11-12
Application Published (Open to Public Inspection) 2020-11-19

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2024-04-29

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.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2021-11-12 2021-11-12
MF (application, 2nd anniv.) - standard 02 2022-05-16 2021-11-12
Request for examination - standard 2024-05-14 2021-11-12
MF (application, 3rd anniv.) - standard 03 2023-05-15 2023-05-10
Final fee - standard 2024-04-25
MF (application, 4th anniv.) - standard 04 2024-05-14 2024-04-29
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
SHANGHAI LEADO PHARMATECH CO. LTD.
Past Owners on Record
LINGLING ZHANG
QIANG DING
YOUXIN WANG
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 (Temporarily unavailable). 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) 
Representative drawing 2024-02-29 1 3
Representative drawing 2024-05-07 1 3
Cover Page 2024-05-07 2 48
Claims 2023-09-20 4 257
Claims 2023-04-02 5 267
Description 2021-11-11 54 3,269
Abstract 2021-11-11 1 15
Claims 2021-11-11 5 211
Drawings 2021-11-11 5 168
Cover Page 2022-01-10 2 44
Description 2023-04-02 54 4,582
Final fee 2024-04-24 4 170
Maintenance fee payment 2024-04-28 1 26
Electronic Grant Certificate 2024-06-03 1 2,527
Courtesy - Letter Acknowledging PCT National Phase Entry 2021-11-30 1 596
Courtesy - Acknowledgement of Request for Examination 2021-11-30 1 434
Commissioner's Notice - Application Found Allowable 2024-03-13 1 578
Examiner requisition 2023-07-03 3 151
Amendment / response to report 2023-09-20 14 558
Amendment - Abstract 2021-11-11 1 74
National entry request 2021-11-11 10 482
International search report 2021-11-11 8 263
Examiner requisition 2022-12-05 5 262
Amendment / response to report 2023-04-02 30 1,211