Language selection

Search

Patent 3200232 Summary

Third-party information liability

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

Claims and Abstract availability

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

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3200232
(54) English Title: CD73 INHIBITORS AND PHARMACEUTICAL USES THEREOF
(54) French Title: INHIBITEURS DE CD73 ET LEURS UTILISATIONS PHARMACEUTIQUES
Status: Application Compliant
Bibliographic Data
(51) International Patent Classification (IPC):
  • C7H 19/20 (2006.01)
  • A61K 31/7076 (2006.01)
(72) Inventors :
  • ZHANG, YUHUA (China)
  • HUANG, CHUANHAO (China)
  • CHEN, GANG (China)
  • LU, JIASHENG (China)
  • ZHANG, QIGUO (China)
  • CHEN, DAWEI (China)
  • ZHU, XINGWU (China)
  • KONG, XIANQI (Canada)
(73) Owners :
  • RISEN (SUZHOU) PHARMA TECH CO., LTD.
(71) Applicants :
  • RISEN (SUZHOU) PHARMA TECH CO., LTD. (China)
(74) Agent: BCF LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2021-11-25
(87) Open to Public Inspection: 2022-06-02
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: 3200232/
(87) International Publication Number: CA2021051688
(85) National Entry: 2023-05-25

(30) Application Priority Data:
Application No. Country/Territory Date
17/133,348 (United States of America) 2020-12-23
202011346141.0 (China) 2020-11-25

Abstracts

English Abstract

CD73 (also known as ecto-5?-nucleotidase) inhibitor compounds are provided, as well as compositions and uses thereof for treating or preventing CD73-associated or related diseases, disorders and conditions, including cancer- and immune-related disorders. CD73 inhibitor compounds include compounds having the structure set forth in Formula I': (I') and pharmaceutically acceptable esters or salts thereof.


French Abstract

L'invention concerne des composés inhibiteurs de la CD73 (également connue sous le nom d'ecto-5-nucléotidase) ainsi que des compositions et des utilisations de ceux-ci pour traiter ou prévenir des maladies, troubles et affections associés ou afférents à la CD73, y compris des troubles liés au cancer et à l'immunité. Les composés inhibiteurs de la CD73 comprennent des composés ayant La structure représentée par la formule I': (I') et des esters ou des sels pharmaceutiquement acceptables de ceux-ci.

Claims

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


Claims
What is claimed is:
1. A compound of Formula I', or a pharmaceutically acceptable salt
or ester thereof:
<IMG>
where:
W is oxygen; X' is ¨P(=0)(OR')¨, wherein R' is a hydrogen; Y is ¨PO3R'2,
wherein R' is
a hydrogen; R'' is a hydroxyl (¨OH); R2' is chlorine (¨C1); and
le and R4', together with the nitrogen atom to which they are attached, form a
monocyclic, bicyclic, tricyclic, spiral-cyclic, or fused-cyclic system,
wherein the cyclic
system is substituted or unsubstituted.
2. The compound of claim 1, wherein the compound is a compound of Formula I,
or a
pharmaceutically acceptable salt or ester thereof:
<IMG>
where:
R1- and R2 are independently selected from hydrogen, unsubstituted or
substituted aryl
group, unsubstituted or substituted heteroaryl group, unsubstituted or
substituted 4- to 8-
membered cyclic group, and unsubstituted or substituted 4- to 8-membered
heterocyclic
group; or,
212
CA 03200232 2023- 5- 25

R1 and R2, together with the carbon atom to which they are attached, form a 4-
to 8-
membered carbocyclic or heterocyclic ring, wherein the cyclic moiety is a
single-ring, a ring
fused with an aromatic ring, or a ring having a ketone functional group;
m and n are independently an integer from 0 to 4, provided that the sum of m
and n is 2
or greater; when m > 1, each R3 is the same or different, and when n > 1, each
R4 is the same
or different; and each R3 and each le are independently selected from
hydrogen, halide,
unsubstituted or substituted aryl group, unsubstituted or substituted
heteroaryl group, and 4-
to 8-membered carbocyclic or heterocyclic ring; or,
when m is 2, 3, or 4, two adjacent R3s, together with the carbon atoms to
which they are
attached, form an unsubstituted or substituted aromatic ring, and R4 and
another R3, if
present, are independently selected from hydrogen and halide; or,
when n is 2, 3, or 4, two adjacent les, together with the carbon atoms to
which they are
attached, form an unsubstituted or substituted aromatic ring, and R3 and
another R4, if
present, are independently selected from hydrogen and halide.
3. The compound of claim 2, wherein the compound is a compound of Formula II,
or a
pharmaceutically acceptable salt or ester thereof:
<IMG>
where:
p and q are independently an integer from 0 to 3, provided that p and q are
not 0 at the
same time, and that when p or q is 0, the carbon and the R group attached to
it are not
present;
r, s and t are independently an integer from () to 2;
R5 and R6 are independently selected from hydrogen, Ci to C6 alkyl,
substituted or
unsubstituted C4 tO C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or
213
CA 03200232 2023- 5- 25

R5 and R6, together with the carbon atoms to which they are attached, form an
unsubstituted or substituted aromatic ring;
R7, R8, R9 and R1- are independently selected from hydrogen, C1 to C6 alkyl,
substituted
or unsubstituted C4 10 C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or, R8 and R9, together with the carbon atoms to which
they are
attached, form a substituted or unsubstituted carbocy clic ring or an
unsubstituted or
substituted aromatic ring; or R7, together with the carbon to which it is
attached, forms a
carbonyl group.
4. The compound of claim 2, wherein the compound is a compound of Formula III,
or a
pharmaceutically acceptable salt or ester thereof:
<IMG>
where:
R5is selected from hydrogen, C1 to C6 alkyl, unsubstituted or substituted C4
tO C7 cyclic
unsubstituted or substituted aryl, and unsubstituted or substituted arylalkyl;
p and q are independently an integer from 0 to 3, provided that p and q are
not 0 at the
same time, and that when p or q is 0, the carbon and the R group attached to
it are not
present; and
R7, R8, R9, and R1 are independently selected from hydrogen, Ci to C6 alkyl,
substituted
or unsubstituted C4 to C7 cyclic alkyl, unsubstituted or substituted aiyl, and
unsubstituted or
substituted arylalkyl; or, R8 and R9, together with the carbon atoms to which
they are
attached, form a substituted or unsubstituted carbocyclic ring or an
unsubstituted or
substituted aromatic ring; or R7, together with the carbon to which it is
attached, forms a
carbonyl group.
214
CA 03200232 2023- 5- 25

5. The compound of claim 2, wherein the compound is a compound of Formula IV,
or a
pharmaceutically acceptable salt or ester thereof:
<IMG>
where:
X is selected from hydrogen, halide, amino group, hydroxyl group, and Ci to C6
alkyl
group;
p and q are independently an integer from 0 to 3, provided that p and q are
not 0 at the
same time, and that when p or q is 0, the carbon and the R group attached to
it are not
present;
r is an integer from 0 to 2; and
R7, R8, R9, and Rl are independently selected from hydrogen, Ci to C6 alkyl,
substituted
or unsubstituted C4 tO C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or, R8 and R9, together with the carbon atoms to which
they are
attached, form a substituted or unsubstituted carbocyclic ring or an
unsubstituted or
substituted aromatic ring; or R7, together with the carbon to which it is
attached, forms a
carbonyl group.
6. The compound of claim 2, wherein the compound is a compound of Formula V,
or a
pharmaceutically acceptable salt or ester thereof:
215
CA 03200232 2023- 5- 25

<IMG>
where:
r and s are independently an integer from 0 to 2, provided that r and s are
not 0 at the
same time;
p and q are independently an integer from 0 to 3, provided that p and q are
not 0 at the
same time, and that when p or q is 0, the carbon and the R group attached to
it are not
present;
R5 and R6 are independently selected from hydrogen, Ci to C6 alkyl,
substituted or
unsubstituted C4 to C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or
R5 and R6, together with the carbon atoms to which they are attached, form an
unsubstituted or substituted aromatic ring;
R7 and Itl are independently selected from hydrogen, C1 to C6 alkyl,
substituted or
unsubstituted C4 10 C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or R7, together with the carbon to which it is
attached, forms a carbonyl
group; and
X is selected from hydrogen, halide, amino group, hydroxyl group, and Cl to C6
alkyl
group.
7. The compound of claim 2, wherein the compound is a compound of Formula VI,
or a
pharmaceutically acceptable salt or ester thereof:
216
CA 03200232 2023- 5- 25

<IMG>
where:
r and s are independently an integer from 0 to 2, provided that r and s are
not 0 at the
same time;
R5 and R6 are independently selected from hydrogen, Ci to C6 alkyl,
substituted or
unsubstituted C4 tO C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or
R5 and R6, together with the carbon atoms to which they are attached, form an
unsubstituted or substituted aromatic ring;
and x ¨12
are independently selected from hydrogen, unsubstituted or substituted aryl
group, unsubstituted or substituted heteroaryl group, and unsubstituted or
substituted 4- to 8-
membered carbocyclic or heterocyclic group; or,
and Ri2, together with the carbon to which they are attached, form an
unsubstituted
or substituted 4- to 8-membered heterocyclic ring.
8. The compound of any one of claims 1 to 7, wherein the compound is:
<IMG>
217
CA 03200232 2023- 5- 25

<IMG>
218
CA 03200232 2023- 5- 25

<IMG>
219
CA 03200232 2023- 5- 25

<IMG>
or a pharmaceutically acceptable salt or ester thereof
9. A pharmaceutical composition comprising the compound or the
pharmaceutically
acceptable salt or ester thereof of any one of claims 1 to R and a ph
armacetiti cally acceptable
carrier.
10. The ph arm aceuti cal comp osi ti on of cl al m 9, wherein the ph arm
aceuti cal ly acceptabl e
carrier comprises a cream, an emulsion, a gel, a liposome, or a nanoparticle.
11. The pharmaceutical composition of claim 9 or 10, wherein the composition
is suitable for
oral administration.
12. The pharmaceutical composition of any one of claims 9 to 11, wherein the
composition is
in the form of a hard shell gelatin capsule, a soft shell gelatin capsule, a
cachet, a pill, a
tablet, a lozenge, a powder, a granule, a pellet, a pastille, or a dragee.
13. The pharmaceutical composition of any one of claims 9 to 11, wherein the
composition is
in the form of a solution, an aqueous liquid suspension, a non-aqueous liquid
suspension,
an oil-in-water liquid emulsion, a water-in-oil liquid emulsion, an elixir, or
a syrup.
14. The pharmaceutical composition of any one of claims 9 to 11, wherein the
composition is
enteric coated.
15. The pharmaceutical composition of any one of claims 9 to 14, wherein the
composition is
formulated for controlled release.
16. The pharmaceutical composition of claim 9 or 10, wherein the composition
is injectable.
220
CA 03200232 2023- 5- 25

17. The pharmaceutical composition of any one of claims 9 to 16, further
comprising at least
one additional therapeutic agent.
18. The pharmaceutical composition of claim 17, wherein the at least one
additional therapeutic
agent is a chemotherapeutic agent, an immune- and/or inflammation-modulating
agent, an
anti-hypercholesterolemia agent, an anti-infective agent, or an immune
checkpoint
inhibitor.
19. A method for treating or preventing a CD73-associated disease, disorder or
condition in a
subject in need thereof, comprising administering a therapeutically effective
amount of the
compound of any one of claims 1 to 8 or the pharmaceutical composition of any
one of
claims 9 to 18 to the subject, such that the CD73-associated disease, disorder
or condition
is treated or prevented in the subject.
20. The method of claim 19, wherein the CD73-associated disease, disorder or
condition is
cancer.
21. The method of claim 20, wherein said cancer is selected from the group
consisting of:
cancers of prostate, colon, rectum, pancreas, cervix, stomach, endometrium,
brain, liver,
bladder, ovary, testis, head, neck, skin, mesothelial membrane, leukocyte,
esophagus,
breast, muscle, connective tissue, lung, adrenal gland, thyroid, kidney and
bone.
22. The method of claim 20, wherein said cancer is selected from the group
consisting of:
glioblastoma, mesothelioma, renal cell carcinoma, gastric cancer, sarcoma,
choriocarcinoma, skin basal cell carcinoma and testicular seminoma.
23. The method of claim 20, wherein said cancer is selected from the group
consisting of:
melanoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer,
lung cancer,
leukemia, brain tumor, lymphoma, ovarian cancer and Kaposi's sarcoma.
24. The method of claim 19, wherein the CD73-associated disease, disorder or
condition is an
immune-related disease, disorder or condition selected from the group
consisting of:
rheumatoid arthritis, kidney failure, lupus, asthma, psoriasis, colitis,
pancreatitis, allergies,
fibrosis, anemia fibromyalgia, Alzheimer's disease, congestive heart failure,
stroke, aortic
valve stenosis, arteriosclerosis, osteoporosis, Parkinson's disease,
infections, Crohn's
disease, ulcerative colitis, allergic contact dermatitis, eczema, systemic
sclerosis and
multipl e sclerosis.
221
CA 03200232 2023- 5- 25

25. The method of any one of claims 19 to 24, further comprising
administration of at least one
additional therapeutic agent to the subject.
26. The method of claim 25, wherein the at least one additional therapeutic
agent and said
compound or composition are administered concomitantly or sequentially.
27. The method of claim 25 or 26, wherein the at least one additional
therapeutic agent is a
chemotherapeutic agent, an immune- and/or inflammation-modulating agent, an
anti-
hypercholesterolemia agent, or an anti-infective agent.
28. The method of claim 25 or 26, wherein the at least one additional
therapeutic agent is an
immune checkpoint inhibitor.
29. A kit comprising the compound or the pharmaceutically acceptable salt or
ester thereof of
any one of claims 1 to 8 and at least one additional therapeutic agent.
30. The kit of claim 29, wherein the at least one additional therapeutic agent
is a
chemotherapeutic agent, an immune- and/or inflammation-modulating agent, an
anti-
hypercholesterolemia agent, an anti-infective agent, or an immune checkpoint
inhibitor.
31. The kit of claim 29 or 30, further comprising a buffer or excipient,
and/or instructions for
use thereof
32. A method for treating cancer in a subject, comprising administering to the
subject an
effective amount of the compound of any one of claims 1 to 8 or the
pharmaceutical
composition of any one of claims 9 to 18 and an immune checkpoint inhibitor,
such that
cancer is treated in the subject.
33. The method of claim 32, wherein said administering is prior to, concurrent
with, or
subsequent to, radiation treatment.
34. The method of claim 32 or 33, wherein said compound and said immune
checkpoint
inhibitor are administered in combination.
35. The method of claim 32 or 33, wherein said compound and said immune
checkpoint
inhibitor are administered sequentially.
36. The method of claim 35, wherein said compound is administered after said
immune
checkpoint inhibitor.
37. The method of claim 35, wherein said compound is administered prior to
said immune
checkpoint inhibitor.
222
CA 03200232 2023- 5- 25

38. The kit of claim 30 or the method of any one of claims 32 to 37, wherein
said immune
checkpoint inhibitor is selected from the group consisting of ipulimumab,
nivolumab and
lambrolizumab.
223
CA 03200232 2023- 5- 25

Description

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


WO 2022/109741
PCT/CA2021/051688
CD73 INHIBITORS AND PHARMACEUTICAL USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001]
This application claims the benefit of priority from Chinese Patent
Application
No. 202011346141.0, filed November 25, 2020, and from U.S. Patent Application
No.
17/133,348, filed December 23, 2020, each of which is hereby incorporated by
reference in its
entirety.
FIELD
[0002] The present disclosure relates to compounds and
compositions that inhibit CD73
(ecto-5'-nucleotidase), and uses thereof for treating and/or preventing CD73-
associated or
related diseases, disorders and conditions, including cancer- and immune-
related disorders.
BACKGROUND
[0003]
Ecto-nucleotidases are a group of cell-surface located ecto-enzymes. The
members of the ecto-nucleotidase family include ecto-nucleotide
pyrophosphatase/
phosphodiesterases (E-NPPs), ecto-nucleoside triphosphate diphosphohydrolases
(E-
NTPDases), ecto-5'-nucleotidase (e5NT, also known as CD73) and alkaline
phosphatase (AP).
These enzymes hydrolyze a variety of extracellular nucleotides to nucleosides
including
adenosine. Extracellular nucleotides are important signaling molecules that
trigger cellular
responses by acting on their respective receptors (for example, adenosine
activates PI
receptors, and nucleotides thereof (ADP, ATP) activate P2 receptors).
Adenosine 5'-
monophosphate (AMP) is a major substrate of CD73 that is hydrolyzed to
adenosine.
Adenosine is ubiquitously present in the body and is an important regulator of
purinergic cell
signaling that is vital for many physiological and pathophysiological
processes.
[0004]
There is a wealth of data implicating CD73 enzymatic activity in promotion
and
metastasis of cancer. CD73 is up-regulated in many cancer cell-types and
tumors, and its
expression has been shown to be associated with tumor neovascularization,
invasiveness and
metastasis. The hydrolytic cascade from extracellular ATP to adenosine is an
important
immunosuppressive regulatory pathway in the tumor microenvironment. CD73
overexpression
impairs adaptive antitumor immune responses, and enhances tumor growth and
metastasis.
Extracellular adenosine is also implicated in regulating adaptive responses to
hypoxia.
Decreasing e5NT activity with monoclonal antibodies, siRNA, and small molecule
inhibitors
1
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
including AMPCP (adenosine [(a,[3)-methy1ene] diphosphate) has been shown to
attenuate the
growth and metastasis of tumors (see, e.g., Zhou et al., Oncol. Rep. 17
(2007): 1341-1346;
Stagg and Smyth, Oncogene, 29 (2010): 5346-5358). Tumor growth is also
impaired in CD73-
deficient mice and it has been established that these effects are largely
mediated by diminished
adenosine production in these mice. Inhibitors of CD73 have thus been actively
explored for
their therapeutic potential against cancer (see, e.g., M. al-Rashida et al.,
Eur. 1 Med. Chem.,
115 (2016): 484-494, and references cited therein).
[0005]
Tumor cells overcome anti-tumor responses in part through
immunosuppressive
mechanisms. There are several such immune modulatory mechanisms. Among them,
adenosine is a key factor which can be generated by both cancer and immune
cells in the tumor
microenvironment to suppress anti-tumor responses. The generation of adenosine
from
adenosine triphosphate (ATP) is catalyzed by two cell-surface proteins, CD73
and CD39, and
can be enhanced under metabolic stress, such as tumor hypoxic conditions.
Adenosine exerts
its immune-regulatory functions through four adenosine receptors (ARs), called
Al, A2A,
A2B, and A3, which are expressed on various immune cells. Overexpression of
adenosine-
generating enzymes such as CD73 and ARs has been correlated with tumor
progression in a
multitude of cancer types. Since the signaling of ARs enhances tumor
progression, their
modulation also represents a promising therapeutic approach for cancer (M. H.
Kazemi, et al.,
1 Cell. Physiol., 233 (2018): 2032-2057, and references cited therein).
[0006]
As mentioned above, ecto-nucleotidases are cell surface-located enzymes
that
regulate purinergic (and pyrimidinergic) signaling pathways. There are four
distinct families
of ecto-nucleotidases: ecto-nucl eosi de triphosphate diphosphohydrolasea
(CD39), ecto-
nucl eoti de pyropho sphatas es/phos phodi esteras es, alkaline phosphatas es,
and ecto-5 -
nucleotidase (e5NT, also known as CD73). CD73 is a glycophosphatidylinositol-
anchored di-
Zn metallophosphatase. CD73 catalyzes the dephosphorylation of extracellular
adenosine
monophosphate (AMP) to adenosine. This ecto-enzymatic cascade in tandem with
CD39
generates adenosine from ATP. The CD73-catalyzed conversion of AMP to
adenosine is
considered to be a major contributor to the elevated levels of extracellular
adenosine in the
tumor microenvironment (Stagg, J. et al., PrOC. Natl. Acad. Sci. USA.: 107
(2010): 1547-1552).
Expression of CD73 is directly upregulated by the hypoxia-inducible factor-la,
which explains
the observed increase in extracellular adenosine in hypoxic malignant tumors.
CD73 is also
expressed by T-regulatory cells (Tregs) and promotes Treg-mediated
immunosuppression
2
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
(Stagg J, et al., Cancer Res. 71 (2011): 2892-2900). In addition, CD73 is
induced by
transforming growth factor-I3 (TGF-13), tumor necrosis factor-a (TNF-a),
hepatocyte growth
factor (HGF), interleukin-6 (IL-6), mitogen-activated protein kinase (MAPK),
signal
transducers and activators of transcription 3 (STAT3), interleukin-2 (IL-2),
retinoic acid,
it/wingless (WNT), epithelial-to-mesenchymal transition, and p53 mutations.
CD73 is
overexpressed in a multitude of tumor types and promotes the invasion,
migration, and
adhesion of tumor cells. CD73 is also associated with immune tolerance and
poor prognosis in
cancer. CD73 is thus a promising target for the development of anti-cancer
drugs. Furthermore,
CD73 inhibitors have potential for the treatment of other diseases mediated by
adenosine and
its receptors (Y.-P. Gong, et al., Expert Op/n. Ther. Pat., 28 (2018): 167-
171).
[0007] The adenosine pathway is also known to be a major
immunosuppressive
component of many human tumors (for review, see Whiteside, T.L., Expert Rev.
Anticancer
Ther., 17 (2017): 527-535). Adenosine and inosine emerge as critical immune
checkpoints in
cancer. Cooperation of the adenosine and PGE2 pathways in the tumor
microenvironment
contributes to suppression of anti-tumor immune effector cells. Targeting of
the adenosine
pathway with pharmacologic inhibitors or antibodies is thus a promising
therapeutic strategy
in cancer.
[0008] Blocking activities of ecto-nucleotidases or of
adenosine receptor signaling in
preclinical in vivo studies has been successful in inhibiting tumor growth and
metastasis. The
adenosine pathway blockade alone or in combination with other immune
therapies. including
checkpoint inhibitors, is currently being implemented in initial phase I
clinical trials for
subjects with advanced malignancies.
[0009] Small-molecule inhibitors of CD73 have been reported.
For example, Adams et
al. (International PCT Application Publication No. W02017/098421) describe
substituted
benzothiadiazine derivatives that are inhibitors of CD73, pharmaceutical
compositions thereof,
and their use in the treatment of cancer, pre-cancerous syndromes and diseases
associated with
CD73 inhibition.
100101 Debien et al. (International PCT Application Publication
No. W02017/120508;
U.S. Patent Application Publication No. US2017/0267710) describe compounds
that modulate
the conversion of AMP to adenosine by 5'-nucleotidase, ecto, compositions
containing the
compounds, methods for synthesizing the compounds, and the use of such
compounds and
3
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
compositions for the treatment and/or prevention of a diverse array of
diseases that are
mediated by 5'-nucleotidase, ecto.
[0011]
Cacatian etal. (International PCT Application Publication No.
W02015/164573)
describe purine derivatives and pharmaceutical compositions thereof which are
inhibitors of
CD73 and are useful in the treatment of cancer.
[0012]
Chen et al. (International PCT Application Publication No. WO 2018/049145)
disclose preparation of nucleotides as ectonucleotidase inhibitors, and the
use of the
compounds in treating or preventing cancer.
SUMMARY
[0013]
The present disclosure relates to compounds and compositions comprising
the
compounds that inhibit the activity of ecto-5'-nucleotidase (also known as
e5NT, CD73, NT5E,
and 5NT). Inhibition of CD73 enzymatic activity leads to inhibition or
modulation of
extracellular adenosine levels and thus modulates the physiological
environment of cells and
tissues.
[0014]
The present disclosure also relates to the use of such compounds and
compositions for the treatment and/or prevention of diseases, disorders and
conditions
mediated, in whole or in part, by CD73. CD73 inhibitors have been linked to
the treatment of
many disorders, including cancer, fibrosis, neurological and neurodegenerative
disorders (e.g.,
depression and Parkinson's disease), cerebral and cardiac i schemi c diseases,
immune-related
disorders, and disorders with an inflammatory component. In particular
embodiments, the
CD73 inhibitor compounds and compositions described herein can act to inhibit
the
immunosuppressive activity and/or the anti-inflammatory activity of CD73, and
are useful as
therapeutics or prophylactic therapies when such inhibition is desirable.
[0015]
In a first broad aspect, there are provided compounds of Formula 1' and
pharmaceutically acceptable salts or esters thereof:
R3' R4'
'N-
N--/L
N
I
Y X-W R2'
'
OH Rt
4
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[0016] where: W is oxygen; X' is ¨P(=0)(OR')¨, wherein R' is a
hydrogen; Y is ¨P03R/2,
wherein R' is a hydrogen; RI' is hydroxyl (¨OH); R2' is chlorine (¨Cl); and
R3' and R4', together
with the nitrogen atom to which they are attached, form a monocyclic,
bicyclic, tricyclic, spiral-
cyclic, or fused-cyclic system, wherein the cyclic system is substituted or
unsubstituted.
[0017]
a second broad aspect, there are provided compounds of Formula I and
pharmaceutically acceptable salts or esters thereof:
R1 R2
( R3 ( R4 ) n
0 0
<
I I
N NI-C 1
HO
I
OH OH
OH OH (I)
[0018]
where: R3 and R2 are independently selected from hydrogen, unsubstituted
or
substituted aryl group, unsubstituted or substituted heteroaryl group,
unsubstituted or
substituted 4- to 8-membered cyclic group, and unsubstituted or substituted 4-
to 8-membered
heterocyclic group; or, 121 and 122, together with the carbon atom to which
they are attached,
form a 4- to 8-membered carbocyclic or heterocyclic ring, wherein the cyclic
moiety is a single-
ring, a ring fused with an aromatic ring, or a ring having a ketone functional
group; m and n
are independently an integer from 0 to 4, provided that the sum of m and n is
2 or greater; when
m> 1, each R3 is the same or different, and when n> 1, each R4 is the same or
different; and
each R3 and each R4 are independently selected from hydrogen, halide,
unsubstituted or
substituted aryl group, unsubstituted or substituted heteroaryl group, and 4-
to 8-membered
carbocyclic or heterocyclic ring; or, when m is 2, 3, or 4, two adjacent R3s,
together with the
carbon atoms to which they are attached, form an unsubstituted or substituted
aromatic ring,
and R4 and another R3, if present, are independently selected from hydrogen
and halide; or,
when n is 2, 3, or 4, two adjacent R4s, together with the carbon atoms to
which they are attached,
form an unsubstituted or substituted aromatic ring, and R3 and another R4, if
present, are
independently selected from hydrogen and halide.
[0019]
In another broad aspect, there are provided compounds of Formula II and
pharmaceutically acceptable salts or esters thereof:
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R7
Rs 126
)9 t
R5
R9
)s
RID( N
0 0
N Cl
HO
OH OH
OH OH (II)
100201
where: p and q are independently an integer from 0 to 3, provided that p
and q are
not 0 at the same time, and that when p or q is 0, the carbon and the R group
attached to it are
not present; r, s and t are independently an integer from 0 to 2; R5 and R6
are independently
selected from hydrogen, Ci to C6 alkyl, substituted or unsubstituted C4 to C7
cyclic alkyl,
unsubstituted or substituted aryl, and unsubstituted or substituted arylalkyl;
or R5 and R6,
together with the carbon atoms to which they are attached, form an
unsubstituted or substituted
aromatic ring; R7, R8, 1{9 and Rm are independently selected from hydrogen, Ci
to C6 alkyl,
substituted or unsubstituted C4 to C7 cyclic alkyl, unsubstituted or
substituted aryl, and
unsubstituted or substituted arylalkyl; or, le and R9, together with the
carbon atoms to which
they are attached, form a substituted or unsubstituted carbocyclic ring or an
unsubstituted or
substituted aromatic ring; or R7, together with the carbon to which it is
attached, forms a
carbonyl group.
[0021]
In another broad aspect, there are provided compounds of Formula III and
pharmaceutically acceptable salts or esters thereof:
6
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R9 R8
( Rio R7)
0 0
I 10
I I N Cl
OH OH 0
______________________________________________ /
OH OH (III)
[0022]
where: R5 is selected from hydrogen, C, to C6 alkyl, unsubstituted or
substituted
C4 to C7 cyclic alkyl, unsubstituted or substituted aryl, and unsubstituted or
substituted
arylalkyl; p and q are independently an integer from 0 to 3, provided that p
and q are not 0 at
the same time, and that when p or q is 0, the carbon and the R group attached
to it are not
present; and R7, R8, R9, and Rl are independently selected from hydrogen, CI
to Ch alkyl,
substituted or unsubstituted C4 to C7 cyclic alkyl, unsubstituted or
substituted aryl, and
unsubstituted or substituted arylalkyl; or, R8 and R9, together with the
carbon atoms to which
they are attached, form a substituted or unsubstituted carbocyclic ring or an
unsubstituted or
substituted aromatic ring; or R7, together with the carbon to which it is
attached, forms a
carbonyl group.
[0023]
In another broad aspect, there are provided compounds of Formula IV and
pharmaceutically acceptable salts or esters thereof:
R8
R7
R9
X
p
RIO ( ),
0 0 <
C
HO I
I I
OH OH
OH OH ( IV )
7
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[0024] where: Xis selected from hydrogen, halide, amino group,
hydroxyl group, and Ci
to Co alkyl group; p and q are independently an integer from 0 to 3, provided
that p and q are
not 0 at the same time, and that when p or q is 0, the carbon and the R group
attached to it are
not present; r is an integer from 0 to 2; and R7, R8, R9, and Rl are
independently selected from
hydrogen, C1 to C6 alkyl, substituted or unsubstituted C4 to C7 cyclic alkyl,
unsubstituted or
substituted aryl, and unsubstituted or substituted arylalkyl; or, le and R9,
together with the
carbon atoms to which they are attached, form a substituted or unsubstituted
carbocyclic ring
or an unsubstituted or substituted aromatic ring; or R7, together with the
carbon to which it is
attached, forms a carbonyl group.
[0025]
another broad aspect, there are provided compounds of Formula V and
pharmaceutically acceptable salts or esters thereof:
R7
)q R6
RI s R5
NN
0 0
<
11'1
Cl
OH OH 0
OH H (V )
[0026]
where: r and s are independently an integer from 0 to 2, provided that r
and s are
not 0 at the same time; p and q are independently an integer from 0 to 3,
provided that p and q
are not 0 at the same time, and that when p or q is 0, the carbon and the R
group attached to it
are not present; re and fe are independently selected from hydrogen, Ci to C6
alkyl, substituted
or unsubstituted C4 to C7 cyclic alkyl, unsubstituted or substituted aryl, and
unsubstituted or
substituted arylalkyl; or R5 and R6, together with the carbon atoms to which
they are attached,
form an unsubstituted or substituted aromatic ring; R7 and Rm are
independently selected from
hydrogen, Ci to Co alkyl, substituted or unsubstituted C4 to C7 cyclic alkyl,
unsubstituted or
substituted aryl, and unsubstituted or substituted arylalkyl; or R7, together
with the carbon to
8
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
which it is attached, forms a carbonyl group; and X is selected from hydrogen,
halide, amino
group, hydroxyl group, and CI_ to C6 alkyl group.
[0027]
In another broad aspect, there are provided compounds of Formula VI and
pharmaceutically acceptable salts or esters thereof:
RH R6
R1rN
s fe
0 0 <
I I
Cl
110 I I
OH OH
OH OH (VT)
[0028]
where: r and s are independently an integer from 0 to 2, provided that r
and s are
not 0 at the same time; R5 and R6 are independently selected from hydrogen,
CI_ to C6 alkyl,
substituted or unsubstituted C4 to C7 cyclic alkyl, unsubstituted or
substituted aryl, and
unsubstituted or substituted arylalk-yl, or R5 and R6, together with the
carbon atoms to which
they are attached, form an unsubstituted or substituted aromatic ring; R" and
R12 are
independently selected from hydrogen, unsubstituted or substituted aryl group,
unsubstituted
or substituted heteroaryl group, and unsubstituted or substituted 4- to 8-
membered carbocyclic
or heterocyclic group; or, R" and R12, together with the carbon to which they
are attached,
form an unsubstituted or substituted 4- to 8-membered heterocyclic ring.
[0029]
In another broad aspect, there are provided compounds of Formula P and
pharmaceutically acceptable salts or esters thereof:
N,R4'
1 I
-W N"N"-- R2.
Y X'
OH Rt (r)
where: W is oxygen, sulfur, nitrogen, or a methylene group; X' is a moiety
selected from
phosphonyl (-P(-0)(0R1)-), sulfonyl (-8(-0)2-), and carbonyl (¨C(-0)¨), where
R' is a
9
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
hydrogen, an ester-forming group, or a protecting group; or X' and W together
form ¨(CR7'R8').,
where n is an integer from 0 to 3, and R7' and le' are independently selected
from a hydrogen,
a halogen, a hydroxyl group and a lower alkyl group having 1 to 4 carbon
atoms; Y is selected
from phosphonate (¨P03R'2), sulfonate (¨SO3R'), and carboxylate (¨CO2R'),
where R' is a
hydrogen, an ester-forming group, or a protecting group; R1' is a hydroxyl
group or a hydrogen;
R2' is chlorine or a hydrogen; and R3' and R4' are independently selected from
a hydrogen, an
alkyl group, an alkenyl group and an alkynyl group, where at least one of R3'
and R4' has from
1 to 30 carbon atoms, such as without limitation from I to 10, from 11 to 20,
from 11 to 30, or
from 21 to 30, and wherein, when W is 0 or S, the carbon number is not 1 to 10
(i.e., when W
is 0 or S, at least one of R3' and R4' has from 11 to 30 carbon atoms).
[0030] In one embodiment of Formula I', R3', R4', and the
nitrogen atom to which they
are attached form a heterocyclic system which is independently selected from a
monocycle, a
bicycle, a tricycle, a spiral-ring, a fused-ring, and a bridged-ring system.
[0031] In one embodiment of Formula I', R3' and R4' are
independently selected from a
hydrogen and a ring system, the ring system being a monocycle, bicycle,
tricycle, spiral-ring,
fused-ring or bridged-ring containing carbocyclic (aromatic or non-aromatic)
or heterocyclic
ring system, and the ring system being substituted or non-substituted,
provided that R3' and R4'
are not both hydrogen at the same time, and provided that the ring system is
not a monocycle
when W is 0 or S.
[0032] In a further embodiment of Formula P, R3' is hydrogen or
a lower alkyl (e.g., Ci_
6) and R4' is ¨C(=0)R5' , ¨C(=0)NHR5' or ¨C(=0)0R5', where R5' is a C1-30
alkyl group, a C2-
30 alkenyl group or a C2-30 alkynyl group, wherein the C1_10 and C2-10 groups
are excluded when
W is 0 or S (i.e., R5' is a Cu-30 alkyl, C11-30 alkenyl or Cu-30 alkynyl group
when W is 0 or S).
100331 In some embodiments of Formula I', R3' is a hydrogen or
a lower alkyl and R4' is
¨C(=0)R5', ¨C(=0)NHR5' or ¨C(=0)0R5', where R5' is a ring system having a
monocycle, a
bicycle, a tricycle, a spiral-ring, a fused-ring or a bridged-ring containing
a carbocyclic
(aromatic or non-aromatic) or a heterocyclic ring system, the carbocyclic or
heterocyclic ring
system being substituted or non-substituted, provided that the R5' ring system
is not a
monocycle ring system when W is 0 or S.
[0034] In some embodiments of Formula I', R3' and R4' are not a
benzyl group.
[0035] In some embodiments of Formula P, the bicycle is not a
biphenyl ring.
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[0036] In some embodiments of
Formula I', R3' and are not a benzyl group, and the
bicycle is not a biphenyl ring.
[0037]
In one embodiment, there are provided compounds of Formula II' and/or
Formula
III', and pharmaceutically acceptable salts or esters thereof:
N,.R4'N,.R4'
N
I<NNCI
OH Rt (W), OH Rt
(III')
where W, X', Y, R1', R3', and R4' are as defined above.
[0038]
In another embodiment, there are provided compounds of Formula IV', and
pharmaceutically acceptable salts or esters thereof:
R3:N, R4NLN
I
Y X'-0
1C:1
OH Rt (IV')
where X', Y, R2', le', and R' are as defined above.
100391
In another embodiment, there are provided compounds of Formula IV'a, and
pharmaceutically acceptable salts or esters thereof:
R3:N R4'
R2'
1:?.)
OH R a V' a)
where X', Y, R2', le, and R' are as defined above.
[0040]
In another embodiment, there are provided compounds of Formula IV'b, and
pharmaceutically acceptable salts or esters thereof:
11
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R3' R4'
NI N
Fe2'

Y X'
OH RI (IV'b)
where X', Y, R2', R3', and R4' are as defined above.
100411
another embodiment, there are provided compounds of Formula IV'c, and
pharmaceutically acceptable salts or esters thereof:
R3' R4'
'N-
R7.
N N R2.
\(c Im'
R8 Ie1/4)
OH R1' (IV'c)
where m' is an integer from 0 to 3, X', Y, RP, R2', R3', and R4' are as
defined above, and R7' and
128' are independently selected from a hydrogen, a halogen, a hydroxyl group
and a lower alkyl
group which has from 1 to 4 carbon atoms.
[0042]
In yet another embodiment, there are provided compounds of Formulae V'
and/or
VI', and pharmaceutically acceptable salts or esters thereof:
R3'NI R4' R3' R4'
I I
Y X' Y X'
OH R1. (V'), OH R1.
(VI')
where X', Y, R1', R3', and R4' are as defined above.
[0043]
In another embodiment, there are provided compounds of Formulae V'a and/or
VI'a, and pharmaceutically acceptable salts or esters thereof:
12
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R3,'N,R4' R3:N,R4'
N---,..--"L---,N
N---_,--)=---..N
H2 2' H 2
N----'
.------. -C -:)- N R y ,-...,x ,C -Li)-?N -N -CI
Y X'
OH R1' (V'a), OH Rt
(VI'a)
where X', Y, R1', R3', and R4' are as defined above.
[0044]
In another embodiment, there are provided compounds of Formulae VII'
and/or
VIII', and pharmaceutically acceptable salts or esters thereof:
R3:N ..R4' WN
R4'
N---......--"L N N-----L.N
0 I 0 OR I
-A -
51,
RO-P CD ''''' N---"`-N". R2'
RO-C P ....' N.---" N R2'
OR 8 )_ 8 0
OH R1' (VW), OH R1'
(VIII')
where R' is hydrogen, an ester-forming group, or a protecting group; and 121',
R2', R3', and R4'
are as defined above.
[0045]
lit another embodiment, there are provided compounds of Formulae VII'a
and/or
VIII'a, and pharmaceutically acceptable salts or esters thereof:
R3:N _ RV R3:N ...
R4'
N ---,-J-- N
0 1 11 0 OR ,N N R2'
II ------, ------..õ N -----' N R2' R 0 - F)
RO-P C
OR 8 8
OH R1' (VII'a), OH R1' (VIII'a)
where R' is a hydrogen, an ester-forming group, or a protecting group; and
Itr, R2', R3', and R4'
are as defined above.
[0046]
In yet another embodiment, there are provided compounds of Formulae IX',
and
pharmaceutically acceptable salts or esters thereof:
13
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R3'N" R4'
-
0 OR_ I N
1,u
RO-P P
OR 8
OH R1. (Iv)
where R' is hydrogen, an ester-forming group, or a protecting group; and 10,
R2', R3', and R4'
are as defined above.
[0047]
In another embodiment, there are provided compounds of Formulae IX'a, and
pharmaceutically acceptable salts or esters thereof:
R3' R4'
-N-
NDCLN
0 OR I
RO¨P P N R2'
OR 0
OH R1' (IX'a)
where: R' is a hydrogen, an ester-forming group, or a protecting group; and
R1', R2', R3', and R4'
are as defined above.
[0048]
In an embodiment of Formula IX'a, R' is a hydrogen, an ester-forming
group, or
a protecting group; R1-' is a hydroxyl group or a hydrogen; R2' is a hydrogen
or a chlorine; and
R3' is a hydrogen or a lower alkyl, and R4' is an alkyl, an alkenyl, or an
alkynyl group having 1
to 30 carbon atoms; or R3' is a hydrogen or a lower alkyl, and R4' is a
substituent group
containing a monocyclic, bicyclic, tricyclic, or multi-cyclic ring system,
where the ring system
is fused, spiral, bridged, or parallel, and the ring system is carbocyclic,
aliphatic, aromatic,
heterocyclic, or a combination thereof; or R3' is a hydrogen or a lower alkyl,
and R4' is
¨C(=0)R5', -C(=0)NHR5' or ¨C(=0)0R5', where R5' is an alkyl group, an alkenyl
group or an
alkynyl group having 1 to 30 carbon atoms; or R3' is a hydrogen or a lower
alkyl, and R4' is
¨C(=0)R5' or ¨C(=0)0R5', where R5' is a substituent group containing a
bicycle, tricycle,
spiral-ring, fused-ring or bridged-ring containing a carbocyclic or a
heterocyclic ring system,
the carbocyclic ring system being aromatic or non-aromatic, the heterocyclic
ring system being
substituted or unsubstituted), where the ring is carbocyclic, aliphatic,
aromatic, heterocyclic,
or a combination thereof; or R3' is a hydrogen or a lower alkyl, and R4' is an
unsubstituted or
substituted 1-adamantyl, a-naphthylmethyl, or 13-naphthylmethyl; or R3', R4',
and the nitrogen
14
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
atom to which they are attached form a heterocyclic system independently
selected from a
monocycle, a bicycle, a tricycle, a spiral-ring, a fused-ring, and a bridged-
ring.
[0049]
In one embodiment of Formula IX'a, R' is a hydrogen, an ester-forming
group, or
a protecting group, R1' is a hydroxyl group or a hydrogen, R2' is a chlorine
or a hydrogen; R3'
is a hydrogen or a lower alkyl; and R' is a group containing an adamantyl
moiety. In some
such embodiments, R' is substituted or non-substituted 1-adamantyl or
substituted or non-
substituted 2-adamantyl. In some such embodiments, R' is substituted or non-
substituted 1-
adamantylmethyl. In some such embodiments, R' is substituted or non-
substituted 1-
adamantylethyl, substituted or non-substituted 1-adamantylpropyl, or
substituted or non-
substituted 1 -adamantylbutyl.
[0050]
In one embodiment of Formula IX'a, R' is a hydrogen, an ester-forming
group, or
a protecting group, R1-' is a hydroxyl group or a hydrogen, R2' is a chlorine
or a hydrogen; R3'
is a hydrogen or a lower alkyl; and R4' is a group containing a naphthyl
moiety. In some such
embodiments, R4' is substituted or non-substituted a-naphthyl or substituted
or non-substituted
(3-naphthyl. In some such embodiments, R' is substituted or non-substituted a-
naphthylmethyl
or substituted or non-substituted f3-naphthylmethyl. In some such embodiments,
R4' is selected
from substituted or non-substituted naphthylethyl, substituted or non-
substituted
naphthylpropyl, and substituted or non-substituted naphthylbutyl.
[0051]
In some embodiments of Formula IX'a, the fused tricycle structure is a
substituted
or unsubstituted carbazolyl moiety.
[0052]
In one embodiment, R1' is a hydroxyl group (i.e., the carbohydrate moiety
in the
compound is a D-ribosyl moiety). In another embodiment, 10. is hydrogen (i.e.,
the
carbohydrate moiety in the compound is a 2-deoxy-D-ribosyl moiety).
[0053]
another embodiment, R2' is hydrogen. In yet another embodiment, RI is
hydrogen and R1-' is a hydroxyl group (i.e., the compound is an adenosine
derivative). In another
embodiment, R2' is hydrogen and R1' is hydrogen (i.e., the compound is a
deoxyadenosine
derivative). In still another embodiment, R2' is hydrogen and both R3' and R4'
are not hydrogen
(i.e., the compound is an adenosine derivative or a deoxyadenosine derivative
with substituent
groups on the amino group of the adenine moiety). In another embodiment, R2'
is chlorine and
the compound is a 2-chloro-D-adenosine derivative or a 2-chloro-D-
deoxyadenosine
derivative.
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[0054]
In some embodiments, R3' is hydrogen or a lower alkyl (e.g., C1_6), and R"
is an
alkyl, alkenyl, or alkynyl group having 1 to 30 carbon atoms (i.e., a C1-30
alkyl group, a C2_30
alkenyl group, or a C2_30 alkynyl group) and R' has 11 to 30 carbon atoms when
W is 0 or S.
In some embodiments, R3' is a hydrogen or a lower alkyl, and R' is a group
containing an
adamantyl moiety. R' may be, for example, substituted or non-substituted 1-
adamantyl,
substituted or non-substituted 2-adamantyl, substituted or non-substituted 1-
adamantylmethyl,
substituted or non-substituted 1-adamantylethyl, substituted or non-
substituted 1-
adamantylpropyl, or substituted or non-substituted 1-adaman1ylbutyl. In some
embodiments,
R3' is a hydrogen or a lower alkyl, and R' is a group containing a naphthyl
moiety. R4' may be,
for example, substituted or non-substituted a-naphthyl, substituted or non-
substituted 13-
naphthyl, substituted or non-substituted a-naphthylmethyl, substituted or non-
substituted 13-
naphthylmethyl, substituted or non-substituted naphthylethyl, substituted or
non-substituted
naphthylpropyl, or substituted or non-substituted naphthylbutyl.
[0055]
another embodiment, R3' is hydrogen or a lower alkyl, and R4' is a
substituent
group containing a monocycle, bicyclic, tricyclic, or multicyclic ring system,
where the ring
system is fused, spiral, bridged, or parallel, and where the ring system is
carbocyclic, aliphatic,
aromatic, heterocyclic, or a combination thereof
[0056] Jr a further embodiment, R3' is hydrogen or a lower
alkyl, and R" is ¨C(=0)R5',
-C(=0)NHR5' or ¨C(=0)0R5', where R5' is an alkyl group or an alkenyl group or
an alkynyl
group haying 1 to 30 carbon atoms, wherein C1 to Cto groups are excluded when
W is 0 or S.
[0057]
h-1 some embodiments. R3' is hydrogen or a lower alkyl, and R' is
¨C(=0)R5', -
C(=0)NHR5' or ¨C(=0)0R5', where R5' is a substituent group containing a
monocyclic,
bicyclic, tricyclic, or multicyclic ring system, where the ring system is
fused, spiral, bridged,
or parallel, and where the ring system is carbocyclic, aliphatic, aromatic,
heterocyclic, or a
combination thereof, wherein R5' is not a monocyclic ring system when W is 0
or S.
[0058]
In one embodiment. R' is a group containing an adamantyl moiety. In
further
embodiment, R' is a substituted or non-substituted 1-adamantyl or 2-adamantyl.
In yet another
embodiment, R' is a substituted or non-substituted 1-adamantylmethyl. In some
embodiments,
R4' is 1-adamantylethyl, 1-adamantylpropyl, or 1-adamantylbutyl, where the
adamantyl moiety
can be substituted or non-substituted.
[0059]
In another embodiment, R" is a group containing naphthyl moiety. In
further
embodiment, R4' is substituted or non-substituted a-naphthy or P-naphthyl. In
other
16
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
embodiment, R4' is a-naphthylmethyl or f3-naphthylmethyl, without or with
further substitution.
In yet another embodiment, R4' is selected from naphthylethyl, naphthylpropyl,
and
naphthylbutyl, where the naphthyl moiety can be non-substituted or
substituted.
[0060]
Jr another embodiment, R3' and R4', together with the nitrogen to which
they are
attached, form a tricyclic fused ring system, such as without limitation a
substituted or
unsubstituted carbazolyl moiety.
[0061]
another embodiment, R3' and R4', together with the nitrogen to which they
are
attached, form a heterocyclic system independently selected from monocycle,
bicycle, tricycle,
spiral-ring, fused-ring, and bridged-ring.
[0062]
In some embodiments, there are provided compounds of Table 1 and
pharmaceutically acceptable salts or esters thereof
Table 1. Structures of example compounds.
Compound
Structure
No.
HN
NNO
0 I
1 H 0
,P P-0-
HO
N CI
OH OH
!Z:L1
OH OH
HN
N
0 0
2 ,P P-0-
HO N N CI
OH OH
0\ /0
HN
3
0 0 I 'r
H
,P P-0- N
HO N CI
OH OH
OH OH
17
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Boc,NBoc
N
9 o I
4 N CI
0 \
(0
Ow
HN
0 0II II
I
HO
N N CI
OH OH
OH OH
HN
NN
6 0 0 I
II FLO¨ CI
HO \'"-
OH OH
OH OH
HN
1\
0 0 I 11
7 H HNNCI
-P P-0¨
HO
OH OH
OH OH
0
HNNN
8 0 0 I
A 1A- o¨o N N CI
OH OH
OH OH
18
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
9
0 0
CI
HO
OH OH
OH OH
NN
0 0 I
N--"NI CI
HO
OH OH ._10
OH OH
0
HN
11 0 0 I
N N CI
HO \"--
OH OH
OH OH
HN
NN
12 0 0 I
II 11
N N CI
H 0 13\
OH OH 0
OH OH
HN
13 N
0 0 I
I! II
NNCI
,P Ho \-`-
P-0-
OH OH
OH OH
19
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
11
HN N
NA H
14
0 0 I NI
II II
,P HO P-0¨N CI
OH OH
OH OH
0
HNNN
AO
15 0 0
II II
N N CI
HO'P\
OH OH 0
OH OH
HN 0
16
0 0 NAII II
,P
HO P-0 N CI
OH OH
OH OH
HN
17 NN
9 9 I .1,
,P Ho P-0¨ CI
0
OH OH
OH OH
0
HNAN
18 o o INNCI
1
II II
HO-PVID\-(3
OH OH 0
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
N
19 9 9 I r
1\1---N CI
HO
OH OH
OH OH
HN
NN
20 9 9 I
,P P-0- N---1\1- CI
HO
OH OH
OH OH
HN
NN
21 0 0 I
,P P-0- CI
II II
HO
OH OH
OH OH
b9H
HN
22
9 I
P-0- NNCI
HO
OH OH
OH OH
HN1 0
23
0 0
II II
,P HO P-0- N N CI
OH OH ()
OH OH
21
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
24 N
0 0
II II I
,P HO P-0-NNCI
OH OH
OH OH
HN
25 NN
9 9
,P P-0- N N CI
HO
OH OH
OH OH
0 0
0 0 I y
26 ii ii
HO,P P-0- N -N CI
OH OH
OH OH
Oaf
NH
27 NN
0 0
II II
1\1----Nr CI
HO
OH OH
OH OH
0
HNANI. U
28
I
,P P-0- N----Th\r- CI
II II
HO
OH OH
OH OH
22
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
0
HN ¨
29 Nlejp.1
0 0II II
I ;1
-P HO P-0¨ N N CI
OH OH __?
OH OH
0
HN
0 0 1
I, I,
,P HO P-0 NNCI
OH OH
OH OH
31 N N
0 0
II II
,P
HO
OH OH
OH OH
NH2
NL
0 0 <NN1CI
32
HO \
OH
OH OH
HN
33
0 0 I
HO0¨ CI
OH
OH OH
23
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
34
O 0 I
II II
,S HO P-0-N CI
0 OH
OH OH
HN
O 0 I
CI
HO II
0
OH OH
36
O 0 IA I
H
N CI
HO-
OH OH 0
OH OH
CF3
HN
37
0 0 I
II II
,P HO P-0-N CI
OH OH
OH OH
38
0 0 I
II II
,P HO P-0- N N CI
0
OH OH
OH OH
24
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
39
0 0 I
II II
-P HO P-0- NNCI
OH OH
OH OH
HN
40 9 9 N fL
,P P-0- N N CI
HO \''=
OH OH
OH OH
,N1
HN H
41 N N
O 0 I
II II
-P P-0- N--TheLCI
HO
OH OH
OH OH
HN
42
O 0 I
CI
II II
HO
OH OH
OH OH
OH
HO
HN
43
O 0
< I
NNLCIII II
HO
OH OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
44
0 o
<
II II
,P HO P-0- N N CI
OH OH
OH OH
0
HNO3
I
9 9
NIII1,P HO P-0- N N CI
OH OH
OH OH
HN
46
N
9
HO I
,P P-0-
9 N CI
OH OH
OH OH
NLN
47 0 0
II II
,P HO P-0 NN -
CI
OH OH
OH OH
48
0 0
< I
II II
,P
HO P-0- Cl
\'"--
OH OH _10
OH OH
26
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
X
49
0 0
H II
,P N N CI
HO
OH OH
OH OH
HO
H2N
HN 0
0 0 I NI'
I, I,
,P HO P-0-N CI
OH OH
OH OH
OH
CjP1)
51
N
0 0 I I'
H H
,P P-0- CI
HO
OH OH
OH OH
HN49U
H
52
0 0
II II
,P N CI
HO 0
OH 0
OH OH
[0063]
some embodiments, there are provided compounds of Table la and
pharmaceutically acceptable salts or esters thereof
Table la. Structures of example compounds.
27
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Compound
Structure
Number
HN
0 0 H I NI
a-1 H 2
HO-P P¨C ¨N CI
OH OH
OH OH
HN
9 9 H2 I IN
-
a-2
HO N CI
OH OH
3K
HN
a-3 N
9 9 H2 I I
P HOC ¨ N CI
OH OH
OH OH
Boc,N,Boc
NN
0 0 H2 I
a-4 c _ CI
0 \
(0
0\ (0
HN
a-5 N
0
0 H I NI
H H
,P HOC ¨ N CI
OH OH
OH OH
28
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
NN
a-6 0 0 14 I
"2
P P-C - N N CI
HO-
OH OH
OH OH
HN
NN
H2 0 I
a-7 II 0 II "2
- N CI
HO
OH OH
OH OH
0
H N0
N
a-8 0 0 " I
II II "2
P HOC
" N CI
HO-
OH OH -2_1
OH OH
HN
H
a-9
0 0 w I
NN CI
HO- \
OH OH
LC)_?
OH OH
op
NN
a-10 0 0
I H2
P-C
" N CI
II HO
OH OH
OH OH
29
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
0
HN
a-11 0 0 1_4 I NI
H ..2
,P
HO P-C - N CI
OH OH
OH OH
cO
HN
NN
a-12
9 9 H2 I
õP HO P-C - N-.1\r- CI
OH OH
OH OH
HN
a-13 NN
9 9 H2 NNCI
,P HOC -
H OH
OH OH
HN N
a-14 NN H
0 0 1.4 I
,P HOC - CI
II II
HO (T
OH OH
OH OH
0
HNNN
a-15 9 9 H2 I
,P HOC CI
HO
OH OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN 0
a-16
0 0 14 I
II II
P P-C
HO NNCI
-
OH OH
OH OH
HN
a-17 N
0 0 H NNCI
II II 2
-P HO P-C -
OH OH
OH OH
0
HNAN
a-18 ';? H2 II
-P P-
HO
OH C -OH
OH OH
NH
10,
HO-P\
HO
a-19 HO \- N-TheLCI
OH OH
HN
NN
a-20 0 142 I
- -õ
N CI
HO-P\13 C
OH OH 0
OH OH
31
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
I
HN
a-21 9 9 H2 I
,P HO P-C - N CI
OH OH
OH OH
HN
a-22
9 9 H2 I NI
- N---N1 CI
HO \
OH OH
OH OH
jt), t7LH---
HN 0
a-23 N
9 9 I-12 I
,P HO P-C - N N CI
OH OH .2_?
OH OH
HN
a-24
N
0 0 1.4
H H
,P P-c - CI
HO \
OH OH ()
OH OH
HN
a-25 NN
0 0 1_4
II II 2 01\r--''N CI
HO -
\
OH OH
OH OH
32
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
NN
a-26 9 H2 I
,P HOC - N CI
HO
OH OH
OH OH
4111
*00
NH
a-27
II
0 0 H I 11
P-C2 N N CI
HO
OH OH
OH OH
0
HNAN
a-28
0 0 L4 I
VC - CI
HO
OH OH
OH OH
--1\11
HN H
a-29 N
1_4 0
0 I Nil
H H
,P HOC -
2 N'NCI
OH OH
OH OH
0 H
HN
a-30
0 0 1_4 N'
I, I, '2 I
,P P-
HO
OH OC -H
OH OH
33
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
cc)
a-31
0 0 H I NI
H 2
,P HO P-C
OH OH
OH OH
NH2
0 0 H_ I NI
a-32
NNCI
HO \
OH
OH OH
HN
a-33
0 OH I NI
HO \
N CI
OH
OH OH
a-34
0 H2 NII
II II
,S HO P-C
0 OH
OH OH
a-35
0 0 H I NI
HOC O LC2
OH OH
34
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
a-36 NNH
0
0 H I
H H 2 õ
-S HOC - " N CI
0 OH
C)_?
OH OH
UJ-CF3
HN
a-37
9 9 H2 I
P HOC - KNNCI
HO- k
OH OH
()
OH OH
1110
a-38 90 1.4 I
H
P P-C - - N CI
H0
OH OH
OH OH
HN
a-39 NN
0 0 1.4 I
N CI
HO -
OH OH
OH OH
HN
a-40 0 H
ii ii 2 õ ,
N CI
-P HOC - "
OH OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
--N1
HN
a-41
0 9H I Nil
õ2 NN CI
,P HOC -
OH OH
()
OH OH
HN
NL
a-42
0 0 H CI
I _LN
II II 2
,P P-C -
HO
OH OH
OH OH
OH
HO
HN
a-43
0 0 I
II II ..2
,P HOC - N CI
OH OH
OH OH
HN
a-44
9 9 H2 NNCI
,P P-C -
HO
OH OH
OH OH
0
cDHN
a-45
0 0 I Ni
14l
CI
II II ..2
,P HOC -
HO
OH OH
OH OH
36
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN /
NN
a-46
9 9 H2 I
,P HO P-C - Nr"N" CIOH OH
OH OH
HN N
NN
a-47 I NI
9 9 H2
,P P- -
HO C
OH OH
OH OH
a-48
0 0 H I
H 2
,P P-C - CI
HO
OH OH
OH OH
a-49
0 0 H I NI
H 2 NNCI
-
HO
OH OH
OH OH
HO
H2N
HN 0
a-50
1\1
0 0 IA I 1
II II
,P HOC - N CI
OH OH
D_?1
OH OH
37
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
OH
a-51
0 0 H I IN
H 2
1\1---N CI
-
HO
OH OH
OH OH
NN S
a-52 9 9 H2 I
- CI
HO
OH OH
OH OH
HNNN
JID
a-53 0 0 H
H 2
-P HOC
- CI
OH OH
OH OH
HN (s) 101
a-54 0 0 1.4
II II -2
,P HO P-C - N CI
1
OH OH
OH OH
HN (s)
,p
HO-P 0
a-55 HO \¨F' "
HO' NN
CI
OH OH
HN (s)
NN F
a-56 0 0 H2 I
II II
HO
-P P-C - 1\1----'N CI
OH OH
OH OH
38
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
NN
a-57 0 H I
õ õ2
-P HO P-C -
OH OH
OH OH
CI
HN
NNO
a-58 9 9' I-12
- NN CI
HO
OH OH
OH OH
a-59 0 H I Nil
õ õ2 NNCI
-P HO \C
OH OH
OH OH
NH2
N
0 0 I
Hil
a-60 H H 2
-P HO P-C -
OH OH
OH OH
a-61
0 0 H I
H 2 NNCI
-
HO
OH OH
OH OH
39
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
ççD
a-62 N----Lm
9 9
,P HO P-0¨ CI
0
OH OH
OH OH
100641
In some embodiments, there are provided compounds of Table lb and
pharmaceutically acceptable salts or esters thereof
Table lb. Structures of example compounds.
Compound
Structure
Number
HN
NO
0 0
b-1 ii ii H
N CI
,P HO P-N¨
OH OH
OH OH
HN
NO
0 0 ii ii H
b-2
HO
" N CI
OH OH
/0
o
HN
b-3
0 0 I
ii ii H
HO
OH OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Boc,N,Boc
______________________________________________________________________
J.N.,
b-4 _k;õ N N CI
0 \
(0
0\ (0
HN
b-5 N
0 0 H _1_1\i
õP 1"¨N¨
HO NNCI
OH OH SEL7
OH OH
HN
NL
b-6 o 0
N
ii ii H I
P HO P¨N¨ NN CI
OH OH
OH OH
HN
NLGO
N
0 0 H
HO
b-7 ii
A¨N¨
N¨Th\1 CI
OH OH
OH OH
0
HNAO-W
NN
b-8 o o ii H
HONA NN-CI
OH OH
OH OH
41
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN4r.&"
b-9
0 0 I Nil
,A
CI
ii H
HO
OH OH
OH OH
op
b-10
9 9 I
H N
HO,P P-N- CI
OH OH SF
OH OH
0
HN
b-11 9 9 H I NI
()1
CI
HO
OH OH
OH OH
JOQ
HN
NN
b-12 0 0
ii ii H I
,P HO P-N- N---N1 CI
OH OH
OH OH
HN
b-13
0 0 ii ii ,
NN-CI
HO
OH OH (,1)
OH OH
42
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN N
NA
b-14
0 0
ii ii H
C
HON N
I
OH OH
OH OH
0
HN
NI
b-15 9 9 H I _L
,P HON N
-_i----1\r
HO
OH CI
OH 19
OH OH
HN 0
b-16
0 0 I NI
ii ii H
HO, CI
OH OH
OH OH
HN
b-17
H 0
0 I NI
H H
,P NNCI
HO P-N-
OH OH
OH OH
0
HNAN
b-18 0 0
,P
HO 0
OH OH
OH OH
43
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
N
0 0 I I'
b-19 A4I
HO -
1\1----N1 CI
OH OH
OH OH
HN
b-20 0 0 I N
ii H NNCI
,P HO P-N-
OH OH
OH OH
I
N
b-21 0 0
ii H
,P P-- N N CI
HONOH OH
OH OH
HN
b-22
0 0 I N
II IIH
,P
C
HO I
OH OH
OH OH
t7LH's-
HN 0
b-23
9 9 H I NI
,P P-N- N N CI
HO
OH OH ()
OH OH
44
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
b-24
0 0 I NI
ii ii H
N CI
HO
OH OH
OH OH
HN
b-25 NLN
0 0 NNCI
ii ii H
,P P-N
HO
OH OH
OH OH
H N
N
II
b-26 H
HO P P - N N N CI
OH OH
OH OH
NH
b-27
0 0ii ii H I
ci
OH OH l2
OH OH
0
HNANifU,
b-28
0 0ii ii H
I
HO'
C1
OH OH LL?
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
--N1
0
HN H
b-29 NN
0 0
ii ii H
HO I
OH OH
OH OH
0
-H
HN
b-30
0 0 I NI
II H
NN-CI
HO
OH OH
OH OH
Ccii)
b-31
NNCI
9 9 H I IN
HO
OH OH 0
OH OH
NH2
NL
0 0 I NI
b-32NCI
HO \
OH
OH OH
b-33 NN H
0 c? H
CI
HO \
OH
OH OH
46
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HAY"
b-34
ii 0 0 I NI
A-FICI CII
HO 11'`=
0 OH
OH OH
HN496.--
b-35
0 0 I
HO 1\1---N CI
II
0
OH OH
HN
b-36
9 9 H I NI
HO,P P-N¨ N -N F
I
OH OH (3
OH OH
CF3
HN
b-37 NN
0 0
ii ii H I
,P HO P-N¨ 1\1--N CI
OH OH
OH OH
N
b-38
9 9 H I
,P P-N N-TheLCI
HO \s'
OH OH
OH OH
47
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
NRIIHN
b-39
9 9 H I
,P P-N- CI
HO
OH OH
OH OH
HN
b-40 9 9 H I
,P P-N CI
HO
OH OH
OH OH
HN
NN
b-41
0 0 I
ii H NNCI
,P HO P-N-
OH OH C?)
OH OH
--NI
HN
b-42
0 0 I N
ii ii H N'CI
õP N
HO
OH OH
OH OH
OH
HO
HN
b-43
0 0 I N
H H
CI
HO
OH OH
OH OH
48
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
b-44
0 0 I N
A A 411 N
HO, N CI
OH OH
OH OH
0
HN
b-45
0 0 N
ii ii H
,P P-N-
HO NNCI
OH OH
OH OH
HN
b-46 N
0 0 õ I
ii ii
N
HO
N N CI
OH OH
OH OH
NN
b-47 0 0 I
,A
II
HON NNCI
OH OH
OH OH
b-48
0 0 I
,P 1124
HO CI
OH OH
OH OH
49
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
X
b-49
Ni
0 0 I l
,P
CI
HO
OH OH
OH OH
HO
H2N
HN 0
b-50
0 0ii N H I
FLN CI
HO _N
OH
OH OH
OH
b-51
0 0 I 11
H
NNCI
HO F-N
OH OH l2
OH OH
0 0 I NI'
b-52 ii ii H N
HO
OH OH
OH OH
HN
b-53 0 0 I NI
II H
-P
1\1----'N CI
HO
OH OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN (S)
N N
b-54 0 0
H NNCI
õP HO P-N
OH OH
c04
OH OH
HN (s)
/;`)
HO-P\ ,0 Nx-L.N
b-55 \¨Pi I
H 0/1-1N N N CI
C)
OH OH
HN (s)
1
b-56 0 0 H
HOii
I Nil
1=-N-
\1---N CI
OH OH
OH OH
NN
b-57 0 0 I
II H
ci
HO
OH OH
OH OH
CI
HN
NO
9 9 H I NI
,P P-N¨ CI
HO
OH OH
OH OH
b-59 0 0 H NI'
II II
HO NN CI
OH OH 1()
OH OH
51
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
NH2
NN
O 0 I
b-60 H
-PCI
HONOH OH
OH OH
b-61
0
II II
H
-P
HO
OH H
O 0
PN
OH OH
[0065]
In some embodiments, there are provided compounds of Table 1 c and
pharmaceutically acceptable salts or esters thereof
Table lc. Structures of example compounds.
Compound
Structure
Number
HN
NS
O I
C-1 II 14 -2
CI
HO \
OH
OH OH
HN
O I jr\i,_
II H2
N CI
c-2 HO-P\-c
OH
0\ /0
/\
52
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
o
HN
c-3 NN
9H2
HOC - N CI
\
OH
OH OH
Boc,NBoc
NN
0 I
II
H2
C-4 C - CI
/0
0\ /0
/?\
HN
c-5
0 H I Nil
2
,P-C - N"NCI
HO \
OH sEfL?
OH OH
HN
c-6 I 0 IA
"2
-CI
HOC
OH 0
OH OH
HNC
0
"-OH
c-7 I NI'
HO-
CI
SFL7
OH OH
53
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
9NLN
C-8 ,P I
HO
OH OH
HN
N
C-9 H -
0 H2 I
-N CI
HO \
OH
OH OH
HN
0
-0H
,P I N
c-10 HO
CI
OH OH
HN
9 N
,P I N
c-11 HO \07:\---\
<NNCI
OH OH
0
HN)LO
N
c-12 0 1.4 I
H
P-C - CI
HO \
OH
OH OH
HNiefe115-"----
N N
c-13
0 14
II-2
N--N
CI
HO \
OH
OH OH
54
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
c-14 0 14 I
H
-P-C - CI
HO
OH
OH OH
0
HN
c-15 0 I
H
- NN"-CI
HO \
OH ()
OH OH
cQHN
NN
c-16 0 I
H
-P-C - NNCI
HO \
OH
OH OH
HN
c-17
i
I j
0 H NNCI
H -2
,P-C -
HO \
OH
OH OH
HN N
c-18
H2
0 I
H _
NNCI
,P-C -
HO \
OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
0
H N0
c-19 0 LA I I
H
HO' P\
OH
OH OH
HN 0
c-20
1_4
0 I
H
,P-C - ---1\r;LCI
HO \
OH
OH OH
HN
c-21
L4
0 I
H
HO'P\
OH 0
OH OH
0
HN N
0 H I NI
CI
_P-C - N
HO \
OH
OH OH
0 IA I y
c-23 H -2
HOC -
\
OH
OH OH
56
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
c-24 0 I
11 -2
_P-C - N^N"--
HO \
OH (?)
OH OH
I
HN
0 H I NI
H -2
_P-C - 1\1---N CI
HO \
OH ?
OH OH
HN
c-26 N
0 I Nil
H -2
,P-C - CI
HO \
OH
OH OH
HN AO
c-27
0 N
11
_P-C - I CI
HO \
OH
OH OH
HN
c-28
1_, N
0
HO \
OH
OH OH
57
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
c-29
N
I
0 IAI'
H "2
,P-C - CI
HO \
OH (?)
OH OH
0 I yc-30 H2
-
CI
HO \
OH
OH OH
*die
'VP NH
c-31
NI/L-NJ
0 w2 I
H "
1-10CN N CI
OH
OH OH
0
HN)LN4e.,
c-32
0 H
H _2
HO,POH-C -
\
OH OH
--NI
0
HN H
c-33
I
0 H "2
,P-C - N NNI CI
HO \
OH
OH OH
58
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
ON
HN
c-34 NN
IA2
0 I
H"
1\1---N- CI
HO \
OH
OH OH
c-35 NN
H
0 I
H -2 --j=L
,P-C - N N CI
HO \
OH
OH OH
0
N
c-36 "OH
I il
HO- \õ---N CI
OH OH
c-37 9N
,P I
HO \
OH 1\1--.1\r CI
C)
OH OH
NH2
0 NNCI
I y
c-38 H2
P\-C
OH
OH OH
59
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HNefd7
c-39
0 H2 NI
NN CI
HO \
OH
OH OH
0
C-40 ,p11-0H
HO I I
N kr -CI
OH OH
0
c-41 N
,P I
HO \0:\----N
N N CI
Cy,?)
OH OH
NJH
C-42
0 I NI
,g-C2 NN -
CI
HO %\
0 Y--?
OH OH
N---,/LN
c-43
0 I
II
H2
C N
HO 01
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HNiej&I
-H
c-44 0 I NI
II
H2
NNCI
C ¨
HO I
OH
OH OH
CF3
HN
c-45 NN
9H2 I
-P¨C ¨ N"N"--- CI
HO \
OH
OH OH
NN
c-46
9 H2 I
-P¨C CI
HO \
OH
OH OH
HN
c-47 N
0 H I
2
_P¨C ¨ NNCI
HO \
OH ._10
OH OH
HN
N
c-48 0 H
-2
- ¨
CI
HOP¨C \
OH
OH OH
61
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
N1
HN
c-49 N
H
0
H = .2
HOC - NI-ThNr CI
\
OH
OH OH
--N
HN
N
c-50
0 1_, NI
ci
,P-C -
HO \
OH
OH OH
OH
HO
HN
c-51
N
0 I
H
HO' PCC (?) ----N**LCI
OH
OH OH
HN
c-52
H
0 I NI
H -2
NNCI
HOC -
\
OH
OH OH
0
49
HN
C-53
0 Li2 I <NNLCI
"
,P-C -
HO \
OH
OH OH
62
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
NN
c-54
I
9H2
HOC - N----"-N" CI
\
OH
OH OH
HN N
c-55 I 9 H2 NI
-P-C - CI
HO \
OH
OH OH
c-56
N N
</
0 1.4
H "2 I
-P-C - CI
HO \
OH
OH OH
c-57
NI
I
0 "I
H "2
HO \
-P-C -
OH
OH OH
HO
H2N
HN 0
c-58
1_4 I N
0 I'
,P-C - N1\1*-CI
HO \
OH
OH OH
63
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
OH
c-59 NLN
0 I
"2
HO,P-C - N N CI
\
OH
OH OH
N N
0 I
HI'
C-60 ..2
,P-C - N CI
HO \
OH (?)
OH OH
HNNLN
X-D
c-61 0 IA
..2
,P-C -
HO \
OH
OH OH
HNL)
0
II OH N
c-62
HO \_N
N eLCI
OH OH
HN
II
c-63 ,P - NNCI
I NI
H 0 \oF-7\ =
OH OH
HN (S)
NO
c-64 0 L4
H ..2
,P-C - NN CI
HO \
OH
OH OH
64
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN 00
-OH N
I
c-65 HO- \---N CI
OH OH
HN 0
9
c-66 õP I NI
HO T"\-\
21_1
OH OH
HN (s)
,p
NO
c-67 HO-P\ <NN CI
I
\
OH OH
HN
0
II OH N N
c-68 õP-
HO I
NN CI
OH OH
HN
0
c-69 ,P I
HO c7\---\ KNNCI
OH OH
HN (s)
N N
c-70 0 I
H
,P-C - N N CI
HO \
OH
OH OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
HN
0
"..-OH
c-71 HO- \---N N CI
LCL?
OH OH
HN
0
c-72 ,P I
HO.-
\(:)F\----\
CI
C)_?
OH OH
NJLN
c-73 KI
II "2
,P-C - N N CI
HO \
OH
OH OH
CI
HN
NN S
c-74 0 LA
H
,P-C - N CI
HO \
OH
OH OH
NN
c-75 9 H2 I
,P-C - N---Th\I CI
HO \ 0
OH
OH OH
NH2
NN
c-76 H 0H 2
N N CI
HO \
OH
OH OH
66
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
c-77 H2 I
NIAN
0
-
,P-C - N N CI
HO \
OH
OH OH
c-78 0
" OH
P- I
HO- \õ----NNCI
01_?
OH OH
c-79 0
,P I
HO \01..-\7---\ CI
L01_?1
OH OH
[0066]
In some embodiments, there are provided compounds of Table id and
pharmaceutically acceptable salts or esters thereof
Table 1 d. Structures of example compounds.
Compound Compound
Structure
Structure
Number Number
d-1 d-2
0 0
-
HO' 0 N 0 0
1 N
OH OH ¨I (1) HO N
CI-
OH OH ¨1,12Rel_
OH OH
OH OH
67
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
N
c.-- F---"C
N N
d-3
N _c-1-----,. N d-4
NXL-N
0 0 0 0
-P HO P-0 N- 'N' 'CI ,i1= -,01
N N. 'CI
1"---"- 1
OH OH ¨1 HOc1.--(-7
OH OH-1 0
OH OH OH OH
/
:F.C--N
0 0 N
__k d-6
HO- (.---11-(3 N N CI 0
0õ11', ('
,, li,
OH OH ¨I ()
HO 6H 6,10-1õ441,-- N CI
OH OH OH OH
Q p
,N
N
d-7
NNI:-Ici
(3
9 9 d-8
HO- 1 (% -1_10 11 11
P-
HO 0 N N
CI
1 - 1
OH OH OH OH
¨
OH OH
F
N
d-9 d-10 N
NI-----t=1-N
0 0 N--_---k.--.
II II 0 0 I
"
-P P-0 N N CI II
II
HO P P-0¨ N---"N
CI
OH OH ¨1 I:) HO I 1
OH OH ..--0--...
OH OH
OH OH
F
N
N
d-11 d-12 N
/,1
N.--_---k--. N 0 0
0 0 KI
741 N CI
1 1 1 1
HO I "----
1
P P-0¨ N---N--- CI OH OH
HO
OH OH () OH OH
OH OH
r--\--- (z
F ,
N N
1
d-13 ,I N d-14
N_____
O 0
< _j 0 0 e/
Nil
1 1 II
,P P0 - N N CI II
II
HO1"----- 1 \N--"N 'CI
OH OH ¨I () 1 HO-01D1""-"Hi g-,_;)-,
OH OH OH OH
68
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
F, ()
c! F*, /
N N
d-15 N --,1,-- d-16 N- --
1-,
O 0
1 1 II II II
-P--. HO- -P-0 N --- N HO CI _P-0
N - "'N' CI
1 --- 1
OH OH -Y---rj_ OH OH -1s1--C-
-?J
OH OH OH
OH
7-0
\
N N
d-17
d-18 0
9 9 0 I
)
11 11
HO-P1'"---"Pl - HO -P P-0
N- --0
OH OH -1 (:.:? OH OH
OH OH OH
OH
0
N
N
d-19 d-20 o 0
N zz-N
I----) 11 11
N IV CI
9 9
'
P HO P-0 N N CI OH OH
OH OH -1. ..?:) I OH
011
OH OH
d-21 N d-22
----"" .. '- N
N--...CI,:=N 0 0
0 0 1 I 11 11
,P P N---- N''''-ci Ho 0 N----
-'N CI
HO 1 ',--- 1 0
.. - 'OH OH
OH OH ¨Isi-1--
OH OH OH
OH
1-\\
/
d-23 N --__ N d-24
i
O 0
/1S1
H H 0 0
- P--,,-P- 0 N ---""'N' CI II
II ,--L. 4-1.
HO1 1 - P---P-0-
N N ¨CI
OH OH --Isi- HO 1 1
OH OH .c:D--.;
OH OH
OH OH
_...----, -,---
I..----.
N N
d-25 1
N --_,--- -=-,-N d-26 N
N
O 0 9 9 1
1 1 1 1
N'N' -GI - P P- - N -
'N 'CI
HO'Pl""-"PI -C) HO 0 1 1
OH OH = -(.711- OH OH (1
OH OH OH
OH
69
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
,
VC
N
pd J.
d-27 N- ,--I-N d-28 o o
O 0 11 11Lo
II ii N
N CI
HO
-P P-0- N ---N1';' CI HO -' '
=
OH OH
LC'4 OH OH
OH
OH OH
C.
,..... i
N =--
1
N
N-..., --. N ...----
d-29 o o
//
d-30 o o
HO' 1 ----- 1 0 r\I----'N CI
jt, _k
OH OH -1,,i.õ (1,) HO 0 N
N CI
OH OH -1,4_7)
OH OH
OH OH
d-31
N- .--C, d-32
N
N- ---1-----
0 0 0 0 cJ
211
i= - N- N CI ,P P-
H0- -(-) HO --,..-- 0-
1c1.' 'N¨CI
OH OH 2 OH OH 0
OH OH OHI
OH
r)
N
N
d-33 d-34
N--- -_,-
N N
P-
CI
HO
N - --N----- CI HO H -
l'---'1 0
0 0
OH OH
-1_,C)_ OH On-1
4204
OH OH
OH OH
N
N
d-35 N-...),-N d-36
N 'L
0 0 1 0 0 --f
'_11 II 11
P-----P-0- N----'N' CI - -0 N
NI
."Cl
HO- HO 1 - 1
OH OH .LC:_o OH OH
-1_04
OH OH OH
OH
NH
N
d-37 N------t,--N d-38
9 9
P P-0- N-
Nr CI
,,,
HO '''' 1 -P--õ,,õ-P- 0
'' N CI
OH OH (.) HO I I
OH OH
OH OH OH
OH
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
N z
d-39
00
P N' N -- a
HO'
OH OH
OH OH
[0067]
In some embodiments, there is provided a compound as described herein
wherein
the C, H, 0, and N atoms in the compound are each independently selected from
atoms of
natural abundance and isotope-enriched atoms. Examples of isotope-enriched
atoms include,
without limitation, "C, 'SC, and mC for carbon; 'H, 2H, and sH for hydrogen;
160, 'TO, and 1'0
for oxygen; and i4N and 1-51\1 for nitrogen.
[0068]
In another broad aspect, there are provided pharmaceutical compositions
comprising a compound described herein, or a pharmaceutically acceptable salt
or ester thereof,
and a pharmaceutically acceptable carrier. In some embodiments, there are
provided
pharmaceutical compositions comprising a compound of any one of Formulae Ito
VI, I' to IX',
IV'a, IV'b, IV'c, Via, VI'a, Vila, or Villa, or a pharmaceutically acceptable
salt or ester thereof,
and a pharmaceutically acceptable carrier. In some embodiments, there are
provided
pharmaceutical compositions comprising a compound of any one of Formulae I to
VI, or a
pharmaceutically acceptable salt or ester thereof, and a pharmaceutically
acceptable carrier. In
some embodiments, there are provided pharmaceutical compositions comprising a
compound
of any one of Formulae I' to IX', or a pharmaceutically acceptable salt or
ester thereof, wherein,
in the compound, one of R3' and R4' is not hydrogen or a Ci to Cio alkyl
alkenyl, or alk-ynyl
group when W is 0 or S. In some embodiments, there are provided pharmaceutical
compositions comprising a compound shown in Tables 1, la, lb, lc, id, or a
pharmaceutically
acceptable salt or ester, and a pharmaceutically acceptable carrier. In some
embodiments, the
pharmaceutically acceptable carrier comprises a cream, an emulsion, a gel, a
liposome, or a
nanoparticle.
[0069] some embodiments, the pharmaceutical composition is
suitable for oral
administration. In some such embodiments, the composition is in the form of a
hard shell
gelatin capsule, a soft shell gelatin capsule, a cachet, a pill, a tablet, a
lozenge, a powder, a
granule, a pellet, a pastille, or a dragee. In some embodiments, the
composition is in the form
of a solution, an aqueous liquid suspension, a non-aqueous liquid suspension,
an oil-in-water
71
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
liquid emulsion, a water-in-oil liquid emulsion, an elixir, or a syrup. In
some embodiments, the
composition is enteric coated. In some embodiments, the composition is
formulated for
controlled release.
[0070] Jr some embodiments, the pharmaceutical composition is
injectable.
[0071] In some embodiments, the pharmaceutically acceptable
carrier further comprises
at least one additional therapeutic agent, such as, without limitation, a
chemotherapeutic agent,
an immune- and/or inflammation-modulating agent, an anti- hypercholesterolemia
agent, or an
anti-infective agent. In an embodiment, the at least one additional
therapeutic agent is an
immune checkpoint inhibitor. Non-limiting examples of immune checkpoint
inhibitors include
ipulimumab, ni volumab and lambroli zumab.
[0072] In another broad aspect, there are provided compounds,
compositions, and
methods of inhibiting CD73 activity in a subject in need thereof comprising
administering to
the subject an effective amount of a compound and/or a pharmaceutical
composition described
herein.
[0073] In particular embodiments, the compounds described
herein act to inhibit the
immunosuppressive activity and/or the anti-inflammatory activity of CD73, and
are useful as
therapeutic or prophylactic therapy when such inhibition is desired. Unless
otherwise indicated,
when uses of the compounds of the present invention are described herein, it
is to be understood
that such compounds may be in the form of a composition (e.g., a
pharmaceutical composition).
As used herein, the terms "CD73 inhibitor", "CD73 blocker", "adenosine by ecto-
5'-
nucleotidase inhibitor", "NT5E inhibitor", "5NT inhibitor" and all other
related art-accepted
terms are used interchangeably to refer to a compound capable of inhibiting,
either directly or
indirectly, the CD73 receptor in an in vitro assay, an in vivo model, and/or
other assay means
indicative of CD73 inhibition and potential therapeutic or prophylactic
efficacy. The terms also
refer to compounds that exhibit at least some therapeutic or prophylactic
benefit in a human
subj ect.
[0074] Although the compounds of the present invention are
believed to have effect by
inhibition of CD73, a precise understanding of the compounds' underlying
mechanism of
action is not required to practice the invention. For example, the compounds
may also have
effect, at least in part, through modulation (e.g., inhibition) of other
components of the
purinergic signaling pathway (e.g., CD39). The purinergic signaling system
consists of
transporters, enzymes and receptors responsible for the synthesis, release,
action, and
72
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
extracellular inactivation of (primarily) ATP and its extracellular breakdown
product
adenosine. Because inhibition of CD73 results in decreased adenosine
production, CD73
inhibitors can be used for the treatment of diseases or disorders mediated by
adenosine and its
actions on adenosine receptors, including Al, A2A, A2B and A3.
[0075]
For purposes of the present disclosure, the purinergic signaling process
can be
described as comprising the following components. The purinergic receptors
(P1, P2X and
P2Y), a first component, are membrane receptors that mediate various
physiological functions
(e.g., relaxation of gut smooth muscle) as a response to the release of ATP or
adenosine; in
general, all cells have the ability to release nucleotides into the
extracellular environment,
frequently through regulated exocytosis. The nucleoside transporters (NTs), a
second
component, are membrane transport proteins which transport nucleoside
substrates (e.g.,
adenosine) across cell membranes; the extracellular concentration of adenosine
can be
regulated by NTs, possibly in the form of a feedback loop connecting receptor
signaling with
transporter function. As previously described, the ecto-nucleotidases (CD73
and CD39)
hydrolyze nucleotides released into the extracellular environment and comprise
a further
component.
[0076]
In some embodiments, there are provided methods for treating or preventing
cancer in a subject (e.g., a human) comprising administering to the subject a
therapeutically
effective amount of at least one CD73 inhibitor compound or composition
described herein. In
some embodiments of such methods, the subject is administered at least one
CD73 inhibitor
compound or composition in an amount effective to reverse, slow or stop the
progression of
CD73-mediated immunos uppression. In some embodiments, the CD73-mediated
immunosuppression is mediated by an antigen-presenting cell (APC).
[0077]
The type of cancer or tumor that can be treated or prevented using the
compounds
and compositions described herein is not meant to be particularly limited.
Examples of cancers
and tumors that can be treated or prevented using the compounds and
compositions described
herein include, but are not limited to: cancers of the prostate, colorectum,
pancreas, cervix,
stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin
(including
melanoma and basal carcinoma), mesothelial lining, white blood cell (including
lymphoma and
leukemia), esophagus, breast, muscle, connective tissue, lung (including small-
cell lung
carcinoma and non-small-cell carcinoma), adrenal gland, thyroid, kidney, bone,
glioblastoma,
mesothelioma, renal cell carcinoma, gastric carcinoma, sarcoma,
choriocarcinoma, cutaneous
73
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
basocellular carcinoma, and testicular seminoma. In some embodiments of the
present
invention, the cancer is melanoma, colon cancer, pancreatic cancer, breast
cancer, prostate
cancer, lung cancer, leukemia, a brain tumor, lymphoma, sarcoma, ovarian
cancer, or Kaposi's
sarcoma.
[0078]
hi some embodiments, there are provided methods of treating a subject
receiving
a bone marrow transplant or peripheral blood stem cell transplant by
administering a
therapeutically effective amount of an CD73 inhibitor compound or composition
sufficient to
increase the delayed-type hypersensitivity reaction to tumor antigen, delay
the time-to-relapse
of post-transplant malignancy, increase relapse-free survival time post-
transplant, and/or
increase long-term post-transplant survival.
[0079]
In certain embodiments, there are provided methods for treating or
preventing an
infective disorder (e.g., a viral infection) in a subject (e.g., a human)
comprising administering
to the subject a therapeutically effective amount of at least one CD73
inhibitor compound or
composition provided herein. In some embodiments, the infective disorder is a
viral infection
(e.g., a chronic viral infection), a bacterial infection, a fungal infection,
or a parasitic infection.
In certain embodiments, the viral infection is human immunodeficiency virus or
cytomegalovirus.
100801
In still other embodiments, there are provided methods for treating and/or
preventing immune-related diseases, disorders and conditions; diseases having
an
inflammatory component; as well as disorders associated with the foregoing;
with at least one
CD73 inhibitor compound or composition provided herein.
[0081]
Other diseases, disorders and conditions that can be treated or prevented,
in whole
or in part, by inhibition of CD73 activity are candidate indications for the
CD73 inhibitor
compounds and compositions provided herein.
[0082]
hi some embodiments, there is further provided the use of the CD73
inhibitor
compounds and compositions described herein in combination with one or more
additional
agents. The one or more additional agents may have some CD73-modulating
activity and/or
they may function through distinct mechanisms of action. In some embodiments,
such agents
comprise radiation (e.g., localized radiation therapy or total body radiation
therapy) and/or
other treatment modalities of a non- pharmacological nature. When combination
therapy is
utilized, the CD73 inhibitor(s) and one additional agent(s) may be in the form
of a single
composition or multiple compositions, and the treatment modalities can be
administered
74
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
concurrently, sequentially, or through some other regimen. By way of example,
in some
embodiments there is provided a treatment regimen wherein a radiation phase is
followed by a
chemotherapeutic phase. A combination therapy can have an additive or
synergistic effect.
[0083] Jr some embodiments, there is provided the use of a CD73
inhibitor compound
or composition described herein in combination with bone marrow
transplantation, peripheral
blood stem cell transplantation, or other types of transplantation therapy.
[0084] In particular embodiments, there is provided the use of
the inhibitors of CD73
function described herein in combination with immune checkpoint inhibitors.
The blockade of
immune checkpoints, which results in the amplification of antigen-specific T
cell responses,
has been shown to be a promising approach in human cancer therapeutics. Non-
limiting
examples of immune checkpoints (ligands and receptors), some of which are
selectively
upregulated in various types of tumor cells, that are candidates for blockade
include PD1
(programmed cell death protein 1); PDL1 (PD1 ligand); BTLA (B and T lymphocyte
attenuator); CTLA4 (cytotoxic T-lymphocyte associated antigen 4); TIM3 (T-cell
membrane
protein 3); LAG3 (lymphocyte activation gene 3); A2aR (adenosine A2a receptor
A2aR); and
Killer Inhibitory Receptors. Non-limiting examples of immune checkpoint
inhibitors include
ipulimumab, nivolumab and lambrolizumab.
100851 In other embodiments, there are provided methods for
treating cancer in a subject,
comprising administering to the subject a therapeutically effective amount of
at least one CD73
inhibitor compound or composition thereof and at least one chemotherapeutic
agent, such
agents including, but not limited to alkylating agents (e.g., nitrogen
mustards such as
chlorambucil, cyclophosphamide, isofamide, mechlorethamine, melphalan, and
uracil mustard;
aziridines such as thiotepa; methanesulphonate esters such as busulfan;
nucleoside analogs
(e.g., gemcitabine); nitroso ureas such as carmustine, lomustine, and
streptozocin;
topoisomerase 1 inhibitors (e.g., irinotecan); platinum complexes such as
cisplatin and
carboplatin; bioreductive alkylators such as mitomycin, procarbazine,
dacarbazine and
altretamine); DNA strand-breakage agents (e.g., bleomycin); topoisomerase II
inhibitors (e.g.,
amsacrine, dactinomycin, daunorubicin, idarubicin, mitoxantrone, doxorubicin,
etoposide, and
teniposide); DNA minor groove binding agents (e.g., plicamydin);
antimetabolites (e.g., folate
antagonists such as methotrexate and trimetrexate; pyrimidine antagonists such
as fluorouracil,
fluorodeoxyuridine, CB3717, azacitidine, cytarabine, and floxuridine; purine
antagonists such
as mercaptopurine, 6-thioguanine, fludarabine, pentostatin; asparginase; and
ribonucleotide
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
reductase inhibitors such as hydroxyurea); tubulin interactive agents (e.g.,
vincristine,
estramustine, vinblastine, docetaxol, epothilone derivatives, and paclitaxel);
hormonal agents
(e. g. , estrogens; conjugated estrogens; ethinyl estradiol; di ethyl
stilbesterol ; chl ortri ani s en;
idenestrol; progestins such as hydroxyprogesterone caproate,
medroxyprogesterone, and
megestrol; and androgens such as testosterone, testosterone propionate,
fluoxymesterone, and
methyltestosterone); adrenal corticosteroids (e.g., prednisone, dexamethasone,
methylprednisolone, and prednisolone); leutinizing hormone releasing agents or
gonadotropin-
releasing hormone antagonists (e.g., leuprolide acetate and goserelin
acetate); and
antihormonal antigens (e.g., tamoxifen, antiandrogen agents such as flutamide;
and antiadrenal
agents such as mitotane and aminoglutethimide). There is also provided the use
of the CD73
inhibitors in combination with other agents known in the art (e.g., arsenic
trioxide) and other
chemotherapeutic agents that may be developed in the future.
[0086]
some embodiments drawn to methods of treating cancer, the administration
of
a therapeutically effective amount of a CD73 inhibitor in combination with at
least one
chemotherapeutic agent results in a cancer survival rate greater than the
cancer survival rate
observed by administering either agent alone. In further embodiments drawn to
methods of
treating cancer, the administration of a therapeutically effective amount of a
CD73 inhibitor in
combination with at least one chemotherapeutic agent results in a reduction of
tumor size or a
slowing of tumor growth greater than reduction of the tumor size or slowing of
tumor growth
observed by administration of either agent alone.
100871
In further embodiments, there are provided methods for treating or
preventing
cancer in a subject, comprising administering to the subject a therapeutically
effective amount
of at least one CD73 inhibitor compound or composition and at least one signal
transduction
inhibitor (STI). In a particular embodiment, the at least one STI is selected
from the group
consisting of bcr/abl kinase inhibitors, epidermal growth factor (EGF)
receptor inhibitors, her-
2/neu receptor inhibitors, and famesyl transferase inhibitors (FTIs).
[0088]
In other embodiments, there are provided methods of augmenting the
rejection of
tumor cells in a subject comprising administering an CD73 inhibitor compound
or composition
in conjunction with at least one chemotherapeutic agent and/or radiation
therapy, wherein the
resulting rejection of tumor cells is greater than that obtained by
administering either the CD73
inhibitor, the chemotherapeutic agent or the radiation therapy alone.
76
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[0089]
In further embodiments, there are provided methods for treating cancer in
a
subject, comprising administering to the subject a therapeutically effective
amount of at least
one CD73 inhibitor and at least one immunomodulator other than a CD73
inhibitor. It should
be understood that, as used herein, a "CD73 inhibitor" refers to compounds
provided herein,
e.g., a compound of any one of Formulae Ito VI, I' to IX', IV'a, IV'b, IV'c,
V'a, VI'a, VII'a, or
Villa, a compound of any one of Tables 1, la, lb, lc, and id, or a
pharmaceutically acceptable
salt or ester thereof, and to pharmaceutical compositions thereof
[0090]
In some embodiments, there are provided methods of treating or preventing
a
CD73-associated disease, disorder or condition in a subject in need thereof,
comprising
administering a therapeutically effective amount of at least one CD73
inhibitor or a
pharmaceutical composition thereof to the subject, such that the CD73-
associated disease,
disorder or condition is treated or prevented in the subject. In some
embodiments, the
compound is administered in an amount effective to reverse, slow or stop the
progression of
CD73-mediated immunosuppression in the subject.
[0091]
In some embodiments, the CD73-associated disease, disorder or condition is
cancer, such as, without limitation, a cancer of the prostate, colon, rectum,
pancreas, cervix,
stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin,
mesothelial lining,
white blood cell, esophagus, breast, muscle, connective tissue, lung, adrenal
gland, thyroid,
kidney, or bone. In some embodiments, the cancer is glioblastoma,
mesothelioma, renal cell
carcinoma, gastric carcinoma, sarcoma, choriocarcinoma, cutaneous bas
ocellular carcinoma,
or testicular seminoma. In some embodiments, the cancer is melanoma, colon
cancer,
pancreatic cancer, breast cancer, prostate cancer, lung cancer, leukemia, a
brain tumor,
lymphoma, ovarian cancer, or Kaposi's sarcoma.
[0092]
In some embodiments, the CD73-associated disease, disorder or condition is
an
immune-related disease, disorder or condition selected from the group
consisting of rheumatoid
arthritis, kidney failure, lupus, asthma, psoriasis, colitis, pancreatitis,
allergies, fibrosis, anemia
fibromyalgia, Alzheimer's disease, congestive heart failure, stroke, aortic
valve stenosis,
arteriosclerosis, osteoporosis, Parkinson's disease, infections, Crohn's
disease, ulcerative
colitis, allergic contact dermatitis, eczema, systemic sclerosis and multiple
sclerosis.
[0093]
In some emb o di ments, methods provided herein further comprise
administration
of at least one additional therapeutic agent to the subject. The at least one
additional therapeutic
agent may be administered concomitantly or sequentially with the compound or
composition
77
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
described herein. In some embodiments, the at least one additional therapeutic
agent is a
chemotherapeutic agent, an immune- and/or inflammation-modulating agent, an
anti-
hypercholesterolemia agent, or an anti-infective agent. In an embodiment, the
at least one
additinal therapeutic agent is an immune checkpoint inhibitor, such as,
without limitation,
ipulimumab, nivolumab or lambrolizumab.
[0094] In some embodiments, there are provided methods for
treating or preventing an
infective disorder (e.g., a viral infection) in a subject (e.g., a human)
comprising administering
to the subject a therapeutically effective amount of at least one CD73
inhibitor and a
therapeutically effective amount of an anti-infective agent(s), such as one or
more antimicrobial
agents.
[0095] In additional embodiments, treatment of an infective
disorder is effected through
the co-administration of a vaccine in combination with administration of a
therapeutically
effective amount of a CD73 inhibitor provided herein. In some embodiments, the
vaccine is an
anti-viral vaccine, including, for example, an anti-HIV vaccine. In other
embodiments, the
vaccine is effective against tuberculosis or malaria. In still other
embodiments, the vaccine is
a tumor vaccine (e.g., a vaccine effective against melanoma); the tumor
vaccine can comprise
genetically modified tumor cells or a genetically modified cell line,
including genetically
modified tumor cells or a genetically modified cell line that has been
transfected to express
granulocyte-macrophage stimulating factor (GM-CSF). In particular embodiments,
the vaccine
includes one or more immunogenic peptides and/or dendritic cells.
[0096] In certain embodiments drawn to treatment of an
infection by administering an
CD73 inhibitor and at least one additional therapeutic agent, a symptom of
infection observed
after administering both the CD73 inhibitor and the additional therapeutic
agent is improved
over the same symptom of infection observed after administering either alone.
In some
embodiments, the symptom of infection observed can be reduction in viral load,
increase in
CDzi-P T cell count, decrease in opportunistic infections, increased survival
time, eradication of
chronic infection, or a combination thereof
100971 In some embodiments, there are provided methods of
treating cancer in a subject,
comprising administering to the subject an effective amount of a compound or
composition
described herein and an immune checkpoint inhibitor, such that cancer is
treated in the subject.
The compound or composition described herein and the immune checkpoint
inhibitor may be
administered in combination or sequentially. The compound or composition may
be
78
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
administered after the immune checkpoint inhibitor or prior to administration
of the immune
checkpoint inhibitor. In some embodiments, the compound or composition and/or
the immune
checkpoint inhibitor are administered prior to, concurrent with, or subsequent
to, other anti-
cancer treatment such as, without limitation, radiation treatment. In some
embodiments, the
immune checkpoint inhibitor is selected from the group consisting of
ipulimumab, nivolumab
and lambrolizumab.
[0098] In another broad aspect, there are provided kits
comprising the compound or
composition described herein. Kits may further comprise a buffer or excipient,
and/or
instructions for use. In some embodiments, kits further comprise at least one
additional
therapeutic agent, such as without limitation a chemotherapeutic agent, an
immune- and/or
inflammation-modulating agent, an anti- hypercholesterolemia agent, an anti-
infective agent,
or an immune checkpoint inhibitor.
BRIEF DESCRIPTION OF THE DRAWINGS
[0099] For a better understanding of the invention and to show
more clearly how it may
be carried into effect, reference will now be made by way of example to the
accompanying
drawings, which illustrate aspects and features according to embodiments of
the present
invention, and in which:
[00100] F1G.1 is a graph showing the CD73 inhibition rate (%
Inhibition vs.
Log[Conc.]/nM) for compound 9;
[00101] FIG. 2 is a graph showing the CD73 inhibition rate for
compound 22;
[00102] FIG. 3 is a graph showing the CD73 inhibition rate for
compound d-1; and
[00103] FIG. 4 is a graph showing the CD73 inhibition rate for
compound a.
DETAILED DESCRIPTION
[00104] The number of subjects diagnosed with cancer and the
number of deaths
attributable to cancer continue to rise. Traditional treatment approaches
comprising
chemotherapy and radiotherapy are generally difficult for the subject to
tolerate and become
less effective as cancers (e.g., tumors) evolve to circumvent such treatments.
Recent
experimental evidence indicates that CD73 inhibitors may represent an
important new
treatment modality for cancer (e.g., breast cancer) treatment.
79
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00105] Promising data also support the role of inhibitors of
CD73 function to inhibit the
anti-inflammatory activity of CD73 and/or the immunosuppressive activity of
CD73, and thus
CD73 inhibitors may be useful to treat, for example, immunosuppressive
diseases (e.g., HIV
and AIDs). Inhibition of CD73 may also be an important treatment strategy for
subjects with
neurological or neuropsychiatric diseases or disorders such as depression.
[00106] There are provided herein, infer alia, small molecule
compounds having CD73
inhibitory activity, as well as compositions thereof, and methods of using the
compounds and
compositions for the treatment and prevention of the diseases, disorders and
conditions
described herein. Compounds provided herein are useful as inhibitors of CD73
and, therefore,
useful in the treatment of diseases, disorders, and conditions in which CD73
activity plays a
role. Additionally, the compounds provided herein may be useful as inhibitors
of adenosine
receptors such as, for example, the A2A receptor. Accordingly, the compounds
provided herein
are useful in the treatment of diseases, disorders, and conditions associated
with activity of one
or more adenosine receptors.
[00107] In an embodiment, there is provided herein a method of
treating a subject (e.g., a
human) with cancer or a disorder mediated by CD73 comprising the step of
administering to
the subject a therapeutically effective amount of an CD73 inhibitor provided
herein, e.g., a
compound provided herein or a pharmaceutically acceptable composition thereof
[00108] It should be understood that a pharmaceutical
composition comprises a compound
disclosed herein (or a pharmaceutically acceptable salt or ester thereof) and
a pharmaceutically
acceptable carrier, adjuvant, or vehicle. In certain embodiments, the amount
of a compound in
a composition is such that it is effective as an inhibitor of CD73 in a
biological sample (e.g., in
an in vitro assay, in an in vivo model, etc.) or in a subject. In certain
embodiments, the
composition is formulated for administration to a subject in need of such
composition. In some
embodiments, the composition is an injectable formulation. In other
embodiments, the
composition is formulated for oral administration to a subject.
[00109] There is also provided a method of treating a subject
(e.g., a human) with cancer
or a disorder mediated by an adenosine receptor (e.g., A2AR) comprising the
step of
administering to the subject a therapeutically effective amount of an CD73
inhibitor provided
herein, e.g., a compound provided herein or a pharmaceutically acceptable
composition
thereof In certain embodiments, the amount of a compound in a composition is
such that it is
effective as an inhibitor of an adenosine receptor (e.g., A2AR) in a
biological sample (e.g., in
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
an in vitro assay, in an in vivo model, etc.) or in a subject. In certain
embodiments, the
composition is formulated for administration to a subject in need of such
composition. In some
embodiments, the composition is an injectable formulation. In other
embodiments, the
composition is formulated for oral administration to a subject. In some
embodiments, the
composition is in the form of a hard shell gelatin capsule, a soft shell
gelatin capsule, a cachet,
a pill, a tablet, a lozenge, a powder, a granule, a pellet, a pastille, or a
dragee. In some
embodiments, the composition is in the form of a solution, an aqueous liquid
suspension, a
non-aqueous liquid suspension, an oil-in-water liquid emulsion, a water-in-oil
liquid emulsion,
an elixir, or a syrup. In some embodiments, the composition is enteric coated.
In some
embodiments, the composition is formulated for controlled release.
[00110]
In further embodiments, there are provided methods for treating or
preventing
cancer in a subject, comprising administering to the subject a therapeutically
effective amount
of at least one CD73 inhibitor and at least one signal transduction inhibitor
(STI). In a particular
embodiment, the at least one STI is selected from the group consisting of
bcr/abl kinase
inhibitors, epidermal growth factor (EGF) receptor inhibitors, her-2/neu
receptor inhibitors,
and farnesyl transferase inhibitors (FTIs). There are also provided methods of
augmenting the
rejection of tumor cells in a subject comprising administering an CD73
inhibitor in conjunction
with at least one chemotherapeutic agent and/or radiation therapy, wherein the
resulting
rejection of tumor cells is greater than that obtained by administering either
the CD73 inhibitor,
the chemotherapeutic agent or the radiation therapy alone. In further
embodiments, there are
provided methods for treating cancer in a subject, comprising administering to
the subject a
therapeutically effective amount of at least one CD73 inhibitor and at least
one
immunomodulator other than a CD73 inhibitor.
[00111]
In other embodiments, there are provided methods for treating or
preventing an
infective disorder (e.g., a viral infection) in a subject (e.g., a human)
comprising administering
to the subject a therapeutically effective amount of at least one CD73
inhibitor and a
therapeutically effective amount of an anti-infective agent(s), such as one or
more antimicrobial
agents.
1001121
In additional embodiments, treatment of an infective disorder is effected
through
the co-administration of a vaccine in combination with administration of a
therapeutically
effective amount of an CD73 inhibitor provided herein. In some embodiments,
the vaccine is
an anti-viral vaccine, including, for example, an anti-HIV vaccine. In other
embodiments, the
81
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
vaccine is effective against tuberculosis or malaria. In still other
embodiments, the vaccine is
a tumor vaccine (e.g., a vaccine effective against melanoma); the tumor
vaccine can comprise
genetically modified tumor cells or a genetically modified cell line,
including genetically
modified tumor cells or a genetically modified cell line that has been
transfected to express
granulocyte-macrophage stimulating factor (GM-CSF). In particular embodiments,
the vaccine
includes one or more immunogenic peptides and/or dendritic cells.
[00113]
In certain embodiments drawn to treatment of an infection by administering
an
CD73 inhibitor and at least one additional therapeutic agent, a symptom of
infection observed
after administering both the CD73 inhibitor and the additional therapeutic
agent is improved
over the same symptom of infection observed after administering either alone.
In some
embodiments, the symptom of infection observed can be reduction in viral load,
increase in
CD4+ T cell count, decrease in opportunistic infections, increased survival
time, eradication of
chronic infection, or a combination thereof
Definitions
[00114]
In order to provide a clear and consistent understanding of the terms used
in the
present specification, a number of definitions are provided below. Moreover,
unless defined
otherwise, all technical and scientific terms as used herein have the same
meaning as commonly
understood to one of ordinary skill in the art to which this invention
pertains.
[00115]
The use of the word "a" or "an" when used in conjunction with the term
"comprising- in the claims and/or the specification may mean "one-, but it is
also consistent
with the meaning of "one or more", "at least one", and "one or more than one".
Similarly, the
word "another" may mean at least a second or more.
[00116]
As used in this specification and claim(s), the words "comprising" (and
any form
of comprising, such as "comprise" and -comprises"), "having" (and any form of
having, such
as "have" and "has"), "including" (and any form of including, such as
"include" and
"includes-) or -containing- (and any form of containing, such as "contain- and
"contains-),
are inclusive or open-ended and do not exclude additional, unrecited elements
or process steps.
[00117]
The term "about- is used to indicate that a value includes an inherent
variation of
error for the device or the method being employed to determine the value.
[00118]
The term "derivative" as used herein, is understood as being a substance
similar
in structure to another compound but differing in some slight structural
detail.
82
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00119]
The present description refers to a number of chemical terms and
abbreviations
used by those skilled in the art. Nevertheless, definitions of selected terms
are provided for
clarity and consistency.
[00120]
As used herein, the term "alkyl" refers to saturated hydrocarbons having
from one
to thirty carbon atoms, including linear, branched, and cyclic alkyl groups.
Examples of alkyl
groups include, without limitation, methyl, ethyl, propyl, butyl, pentyl,
hexyl, heptyk octyl,
nonyl, decyl, isopropyl, tert-butyl, sec-butyl, isobutyl, cyclopropyl,
cyclopentyl, cyclohexyl,
cycloheptyl, cyclooctyl, and the like. The term alkyl includes both
unsubstituted alkyl groups
and substituted alkyl groups. The terms "Ci-Cnalkyl" and "Ci_n alkyl", wherein
n is an integer
from 2 to 30, are used interchangeably to refer to an alkyl group having from
1 to the indicated
"n" number of carbon atoms. Alkyl residues may be substituted or
unsubstituted. In some
embodiments, for example, alkyl may be substituted by hydroxyl, amino,
carboxyl, carboxylic
ester, amide, carbamate, or aminoalkyl. In some particular emodiments, -alkyl"
is modified by
a range of the number of carbon atoms and thus the size of the alkyl group is
defined
specifically. For example, a Cu-C30 alkyl specifies an alkyl group containing
at least 11 carbon
atoms and not more than 30 carbon atoms.
[00121]
As used herein, the term "acyclic" refers to an organic moiety without a
ring
system. The term "aliphatic group" includes organic moieties characterized by
straight or
branched-chains, typically having between 1 and 15 carbon atoms. Aliphatic
groups include
non-cyclic alkyl groups, alkenyl groups, and alkynyl groups.
[00122]
As used herein, the term "alkenyl" refers to unsaturated hydrocarbons
having
from two to thirty carbon atoms, including linear, branched, and cyclic non
aromatic alkenyl
groups, and comprising between one to six carbon-carbon double bonds. Examples
of alkenyl
groups include, without limitation, vinyl, allyl, 1-propen-2-yl, 1-buten-3-yl,
1-buten-4-yl, 2-
buten-4-yl, 1-penten-5-yl, 1,3 -pentadien-5-yl,
cyclopentenyl, cyclohexenyl,
ethylcyclopentenyl, ethylcylohexenyl, and the like. The term alkenyl includes
both
unsubstituted alkenyl groups and substituted alkenyl groups. The terms "C2-
Cnalkenyl" and
"C211 alkenyl-, wherein n is an integer from 3 to 30, are used interchangeably
to refer to an
alkenyl group having from 2 to the indicated "n" number of carbon atoms. In
some particular
embodiments, "alkenyl" is modified by a range of the number of carbon atoms
and thus the
size of the alkenyl group is defined specifically. For example, a Cii-C30
alkenyl specifies an
alkenyl group containing at least 11 carbon atoms and not more than 30 carbon
atoms.
83
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00123]
As used herein, the term "alkynyl" refers to unsaturated hydrocarbons
having
from two to thirty carbon atoms, including linear, branched, and cyclic non
aromatic alkynyl
groups, and comprising between one to six carbon-carbon triple bonds. Examples
of alkynyl
groups include, without limitation, ethynyl, 1-propyn-3-yl, 1-butyn-4-yl, 2-
butyn-4-yl, 1-
pentyn-5-yl, 1,3-pentadiyn-5-yl, and the like. The term alkynyl includes both
unsubstituted
alkynyl groups and substituted alkynyl groups. The terms "C2-Calkynyl" and
"C2_,, alkynyl",
wherein n is an integer from 3 to 30, are used interchangeably to refer to an
alkynyl group
having from 2 to the indicated "n" number of carbon atoms. In some particular
embodiments,
"alkynyl" is modified by a range of the number of carbon atoms and thus the
size of the alkynyl
group is defined specifically. For example, a Cii-C30 alkynyl specifies an
alkynyl group
containing at least I I carbon atoms and not more than 30 carbon atoms.
[00124]
Unless the number of carbons is otherwise specified, "lower" as in "lower
aliphatic," "lower alkyl," "lower alkenyl," and "lower alkylnyl", as used
herein means that the
moiety has at least one (two for alkenyl and alkynyl) and equal to or less
than 6 carbon atoms.
[00125]
The terms "cycloalkyl", "alicyclic", "carbocyclic" and equivalent
expressions
refer to a group comprising a saturated or partially unsaturated carbocyclic
ring in a single,
spiro (sharing one atom), or fused (sharing at least one bond) carbocyclic
ring system having
from three to fifteen ring members. Examples of cycloalkyl groups include,
without limitation,
cyclopropyl, cyclobutyl, cyclopentyl, cyclopenten-l-yl, cyclopenten-2-yl,
cyclopenten-3-yl,
cyclohexyl, cyclohexen- 1 -yl, cyclohexen-2-yl,
cyclohexen-3-yl, cycloheptyl,
bicyc1014,3,01nonanyl, norbomyl, and the like. The term cycloalkyl includes
both unsubstituted
cycloalkyl groups and substituted cycloalkyl groups. The terms "C3-
C11cycloalkyl" and "C3-n
cycloalkyl", wherein n is an integer from 4 to 15, are used interchangeably to
refer to a
cycloalkyl group having from 3 to the indicated "n" number of carbon atoms in
the ring
structure. Unless the number of carbons is otherwise specified, "lower
cycloalkyl" groups as
herein used, have at least 3 and equal to or less than 8 carbon atoms in their
ring structure.
[00126]
Cycloalkyl residues can be saturated or contain one or more double bonds
within
the ring system. In particular they can be saturated or contain one double
bond within the ring
system. In unsaturated cycloalkyl residues the double bonds can be present in
any suitable
positions. Monocycloalkyl residues are, for example, cyclopropyl, cyclobutyl,
cyclopentyl,
cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl,
cyclooctyl, cyclononyl,
cyclodecyl, cycloundecyl, cyclododecyl or cyclotetradecyl, which can also be
substituted, for
84
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
example by C1-4 alkyl. Examples of substituted cycloalkyl residues are 4-
methylcyclohexyl and
2,3-dimethylcyclopentyl. Examples of parent structures of bicyclic ring
systems are
norbornane, bi cy cl o [2. 2. 11heptane, bicyclo [2.2.2] octane and bi cy cl o
[3 .2. 1] octane.
[00127]
The term "heterocycloalkyl" and equivalent expressions refers to a group
comprising a saturated or partially unsaturated carbocyclic ring in a single,
Spiro (sharing one
atom), or fused (sharing at least one bond) carbocyclic ring system having
from three to fifteen
ring members, including one to six heteroatoms (e.g., N, 0, S, P) or groups
containing such
heteroatoms (e.g., NH, NRx (Rx is alkyl, acyl, aryl, heteroaryl or
cycloalkyl), PO2, SO, SO2,
and the like). Heterocycloalkyl groups may be C-attached or heteroatom-
attached (e.g., via a
nitrogen atom) where such is possible. Examples of heterocycloalkyl groups
include, without
limitation, pyrrolidino, tetrahydrofuranyl, tetrahydrodithienyl,
tetrahydropyranyl,
tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl,
piperazinyl,
azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl,
oxazepinyl, diazepinyl,
thiazepinyl, 1,2.3.6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl,
indolinyl, 2H-pyranyl, 4H-
pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl,
dihydropyranyl,
dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl,
azabicyclo[3,1,01hexanyl, 3-azabicyclo[4,1,01heptanyl, 311-indolyl,
quinolizinyl, and sugars,
and the like. The term heterocycloalkyl includes both unsubstituted
heterocycloalkyl groups
and substituted heterocycloalkyl groups. The terms "C3-C1.1heterocycloalkyl"
and "C3-n
heterocycloalkyl", wherein n is an integer from 4 to 15, are used
interchangeably to refer to a
heterocycloalkyl group having from 3 to the indicated "n" number of atoms in
the ring structure,
including at least one hetero group or atom as defined above. Unless the
number of carbons is
otherwise specified, "lower heterocycloalkyl" groups as herein used, have at
least 3 and equal
to or less than 8 carbon atoms in their ring structure.
1001281
The terms "aryl" and "aryl ring" refer to aromatic groups having "4n-F2"
(pi)
electrons, wherein n is an integer from 1 to 7, in a conjugated monocyclic or
polycyclic system
(fused or not) and having six to fourteen ring atoms. A polycyclic ring system
includes at least
one aromatic ring. Aryl may be directly attached, or connected via a Ci-C6
alkyl group (also
referred to as arylalkyl or aralkyl). Examples of aryl groups include, without
limitation, phenyl,
benzyl, phenetyl, 1-phenylethyl, tolyl, naphthyl, biphenyl, terphenyl,
indenyl,
ben zo cy cl oo cteny I , ben zo cy cl oh eptenyl , azul eny I , acenaphthyl
eny I, fl uorenyl, ph en anth emy I,
anthracenyl, and the like. The term aryl includes both unsubstituted aryl
groups and substituted
aryl groups. The terms "C6-C11aryl" and "C6-11 aryl", wherein n is an integer
from 6 to 30, are
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
used interchangeably to refer to an aryl group having from 6 to the indicated
"n" number of
atoms in the ring structure, including at least one hetero group or atom as
defined above.
[00129]
The terms "heteroaryl" and "heteroaryl ring" refer to an aromatic group
having
"4n+2"(pi) electrons, wherein n is an integer from 1 to 7, in a conjugated
monocyclic or
polycyclic system (fused or not) and having five to fourteen ring members,
including one to
six heteroatoms (e.g. N, 0, S) or groups containing such heteroatoms (e.g. NH,
NRx (Rx is
alkyl, acyl, aryl, heteroaryl or cycloalkyl). SO, and the like). A polycyclic
ring system includes
at least one heteroaromatic ring. Heteroaryls may be directly attached, or
connected via a Ci-
C3alkyl group (also referred to as heteroarylalkyl or heteroaralkyl).
Heteroaryl groups may be
C-attached or heteroatom-attached (e.g., via a nitrogen atom), where such is
possible. Examples
of heteroaryl groups include, without limitation, pyridyl, imidazolyl,
pyrimidinyl, pyrazolyl,
triazolyl, tetrazolyl, furyl, thienyl; isooxazolyl, thiazolyl, oxazolyl,
isothiazolyl, pyrrollyl,
quinolinyl, isoquinolinyl, indolyl, isoindolyl, chromenyl, isochromenyl,
benzimidazolyl,
benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl,
pyrazinyl, triazinyl,
isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl,
benzofurazanyl,
benzothiophenyl, benzothienyl, benzothiazolyl, benzoxazolyl, quinazolinyl,
quinolizinyl,
quinolonyl, isoquinolonyl, quinoxalinyl, naphthyridinyl, furopyridinyl,
carbazolyl,
phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenazinyl,
phenothiazinyl,
phenoxazinyl, dibenzofumayl, and the like. The term heteroaryl includes both
unsubstituted
heteroaryl groups and substituted heteroaryl groups. The terms "Cs-
Cnheteroaryl" and "C5-11
heteroaryl", wherein n is an integer from 6 to 29, are used interchangeably to
refer to a
heteroaryl group having from 5 to the indicated "n" number of atoms in the
ring structure,
including at least one hetero group or atom as defined above.
[00130]
The terms "heterocycle" or "heterocyclic" include heterocycloalkyl and
heteroaryl groups. Examples of heterocycles include, without limitation,
acridinyl, azocinyl,
benzimidazolyl, benzofuranyl, benzothiofuranyl,
benzothiophenyl, benzoxazoly 1,
benzthi azolyl , benztriazolyl, benztetrazolyl,
benzisoxazolyl, ben zi s otbi azoly 1 ,
benzimidazolinyl, carbazolyl, 412H-carbazolyl, carbolinyl, chromanyl,
chromenyl, cinnolinyl,
decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro12,3-
bitetrahydrofuran, furanyl,
furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl,
indolinyl,
indolizinyl, indolyl, 3H-indolyl, isobenzofuranyl, isochromanyl, isoindazolyl,
isoindobnyl,
isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl,
morpholinyl,
naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-
oxadiazolyl,
86
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl,
pyrimidinyl,
phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl,
phenoxazinyl,
phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl,
pteridinyl,
purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl,
pyridazinyl. pyridooxazole,
pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl,
pyrrolidinyl, pyrrolinyl, 2H-
pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl,
quinuclidinyl,
tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl,
6H-1,2,5-
thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl,
1,3,4-thiadiazolyl,
thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl,
thienoimidazolyl, thiophenyl,
triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl,
xanthenyl, and the like.
The term heterocycle includes both unsubstituted heterocyclic groups and
substituted
heterocyclic groups.
[00131] The term "amine" or "amino," as used herein, refers to
an unsubstituted or
substituted moiety of the formula --NRaRb, in which Ra and Rb are each
independently
hydrogen, alkyl, aryl, or heterocyclyl, or Ra and Rb, taken together with the
nitrogen atom to
which they are attached, form a heterocyclic ring. The term amino includes
compounds or
moieties in which a nitrogen atom is covalently bonded to at least one carbon
or heteroatom.
Thus, the terms "alkylamino" and "dialkylamino" as used herein mean an amine
group having
respectively one and at least two C1-C6alkyl groups attached thereto. The
terms "arylamino"
and "diarylamino" include groups wherein the nitrogen is bound to at least one
or two aryl
groups, respectively. The terms "amide" or "aminocarbonyl" include compounds
or moieties
which contain a nitrogen atom which is bound to the carbon of a carbonyl or a
thiocarbonyl
group. The term "acylamino" refers to an amino group directly attached to an
acyl group as
defined herein.
1001321 The term "bicycle" or "bicyclic" refers to a ring system
with two rings that has
two ring carbon atoms in common, and which can be located at any position
along either ring,
generally referring to bicyclic hydrocarbon radical, bicyclic aromatic carbon
atom ring
structure radical, and a saturated or partially unsaturated bicyclic carbon
atom ring structure
radical in which one or more carbon atom ring members have been replaced,
where allowed by
structural stability, with a heteroatom, such as an 0, S or N atom. The
bicyclic system can be
a fused-ring system, such as bi cycl o [4.4. 0] decan e or naphthalene, or a
bridged-ring system,
such as bicyclo[2.2.21octane.
87
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00133] The term "tricycle- or "tricyclic- refers to a ring
system with three rings that has
three ring carbon atoms in common, and which can be located at any position
along each ring;
generally referring to tricyclic hydrocarbon radical, tricyclic aromatic
carbon atom ring
structure radical, and a saturated or partially unsaturated tricyclic carbon
atom ring structure
radical in which one or more carbon atom ring members have been replaced,
where allowed by
structural stability, with a heteroatom, such as an 0, S or N atom. A
tricyclic system can have
three rings arranged as a fused ring, such as anthracene or
tetradecahydroanthracene, or a
bridged ring, such as in adamantine or tricycle[3.3.1.1]decane.
[00134] The term "multi-cycle", "multicycle", -multi-cyclic", or
"multi-cyclic" means a
ring system with more than three rings having more than three ring carbon
atoms in common,
and which can be located at any position along either ring. The term generally
refers to a
multicyclic hydrocarbon radical, a multicyclic aromatic carbon atom ring
structure radical, and
a saturated or partially unsaturated multicyclic carbon atom ring structure
radical in which one
or more carbon atom ring members have been replaced, where allowed by
structural stability,
with a heteroatom, such as an 0, S or N atom.
[00135] The term "fused ring" or -fused" refers to a polycyclic
ring system that contains
fused rings. Typically, a fused ring system contains 2 or 3 rings and/or up to
18 ring atoms. As
defined above, cycloalkyl radicals, aryl radicals and heterocyclyl radicals
may form fused ring
systems. Thus, a fused ring system may be aromatic, partially aromatic or not
aromatic and
may contain heteroatoms. A spiro ring system is not a fused-polycyclic by this
definition, but
fused polycyclic ring systems of the invention may themselves have spiro rings
attached thereto
via a single ring atom of the system. Examples of fused ring systems include,
but are not limited
to, naphthyl (e.g. 2-naphthyl), indenyl, fenanthryl, anthracyl, pyrenyl,
benzimidazole,
benzothiazole, etc. The terms -fused ring" and -fused-cyclic" are used
interchangeably herein.
[00136] The term "spiral ring" or "spiral" refers to an organic
compound, that presents a
twisted structure of two or more rings (a ring system), in which 2 or 3 rings
are linked together
by one common atom. Spiro compounds may be fully carbocyclic (all carbon),
such as without
limitation spiro[5.51undecane or heterocyclic (having one or more non-carbon
atom), including
but not limited to carbocyclic spiro compounds, heterocyclic spiro compounds
and polyspiro
compounds. The terms "spiral ring" and -spiral-cyclic" are used
interchangeably herein.
[00137] The term -bridged ring" or -bridged" refers to a
carbocyclic or heterocyclic
moiety where two or more atoms are shared between two or more ring structures,
where any
88
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
such shared atom is C, N, S, or other heteroatom arranged in a chemically
reasonable
substitution pattern. Alternatively, a "bridged" compound also refers to a
carbocyclic or
heterocyclic ring structure where one atom at any position of a primary ring
is bonded to a
second atom on the primary ring through either a chemical bond or atom (s)
other than a bond
which does (do) not comprise a part of the primary ring structure. The first
and second atom
may or may not be adjacent to one another in the primary ring. Illustrated
below are specific
non-limiting examples of bridged ring structures contemplated herein. Other
carbocyclic or
heterocyclic bridged ring structures are also contemplated, including bridged
rings wherein the
bridging atoms are C or heteroatom (s) arranged in chemically reasonable
substitution patterns,
as are known in the art.
[00138] The term "nitro" means ¨NO2; the terms "halo" and
"halogen" refer to bromine,
chlorine, fluorine or iodine substituents; the terms "thiol", "thio", and
"mercapto" mean ¨SH;
and the terms "hydroxyl" and "hydroxy" mean ¨OH. The term "alkylthio" refers
to an alkyl
group, having a sulfhydryl group attached thereto. Suitable alkylthio groups
include groups
having 1 to about 12 carbon atoms, preferably from 1 to about 6 carbon atoms.
The term
"alkylcarboxyl" as used herein means an alkyl group having a carboxyl group
attached thereto.
[00139] The terms "alkoxy" and "lower alkoxy" as used herein
mean an alkyl group
having an oxygen atom attached thereto. Representative alkoxy groups include
groups having
1 to about 6 carbon atoms, e.g., methoxy, ethoxy, propoxy, tert-butoxy and the
like. Examples
of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy,
pentoxy,
fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloromethoxy,
dichloromethoxy,
trichloromethoxy groups, and the like. The term "alkoxy" includes both
unsubstituted or
substituted alkoxy groups, etc., as well as perhalogenated alkyloxy groups.
[00140] The terms "carbonyl" and "carboxy" include compounds and
moieties which
contain a carbon connected with a double bond to an oxygen atom. Examples of
moieties which
contain a carbonyl include aldehydes, ketones, carboxylic acids, amides,
esters, anhydrides,
etc.
1001411 The term "acyl" refers to a carbonyl group that is
attached through its carbon atom
to a hydrogen (i.e., formyl), an aliphatic group (e.g., Ci-C29 alkyl, Ci-C29
alkenyl, Ci-C29
alkynyl, e.g., acetyl), a cycl alkyl group (C3-C8cycloalkyl), a heterocyclic
group (C3-
Csheterocycloalkyl and Cs-C6heteroary1), an aromatic group (C6aryl, e.g.,
benzoyl), and the
like. Acyl groups may be unsubstituted or substituted acyl groups (e.g.,
salicyloyl).
89
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00142]
It should be understood that "substitution" or "substituted with includes
the
implicit proviso that such substitution is in accordance with the permitted
valence of the
substituted atom and the substituent, and that the substitution results in a
stable compound, i.e.,
a compound which does not spontaneously undergo transformation such as by
rearrangement,
cyclization, elimination, etc. As used herein, the term "substituted" is meant
to include all
permissible substituents of organic compounds. In a broad aspect, the
permissible substituents
include acyclic and cyclic, branched and unbranched, carbocyclic and
heterocyclic, aromatic
and nonaromatic substituents of organic compounds. The permissible
substituents can be one
or more. The term "substituted", when used in association with any of the
foregoing groups
refers to a group substituted at one or more position with substituents such
as acyl, amino
(including simple amino, mono and dialkylamino, mono and diaryl amino, and
alkylarylamino),
acylamino (including carbamoyl, and ureido), alkylcarbonyloxy,
arylcarbonyloxy,
alkoxycarbonyloxy, alkoxycarbonyl, carboxy, carboxylate, aminocarbonyl, mono
and
dialkylaminocarbonyl, cyano, azido, halogen, hydroxyl, nitro, trifluoromethyl,
thio, alkylthio,
wylthio, alk-ylthiocarbonyl, thiocarboxylate, lower alkyl, lower alkenyl,
lower alk-ynyl,
cycloalkyl, heterocycloalkyl, aryl, heteroaryl, lower alkoxy, aryloxy,
aryloxycarbonylov,
benzyloxy, benzyl, sulfinyl, alkylsulfinyl, sulfonvl, sulfate, sulfonate,
sulfonamide, phosphate,
phosphonato, phosphinato, oxo, guanidine, imino, formyl and the like. Any of
the above
substituents can be further substituted if permissible, e.g., if the group
contains an alkyl group,
an aryl group, or other.
[00143]
The term "solvate" refers to a physical association of a compound with one
or
more solvent molecules, whether organic or inorganic. This physical
association includes
hydrogen bonding. In certain instances, a solvate will be capable of
isolation, for example when
one or more solvent molecules are incorporated in the crystal lattice of a
crystalline solid.
"Solvate" encompasses both solution-phase and isolable solvates. Exemplary
solvates include,
without limitation, hydrates, ethanolates, methanolates, hemiethanolates, and
the like.
[00144]
A "pharmaceutically acceptable salt" of a compound means a salt of a
compound
that is pharmaceutically acceptable. Desirable are salts of a compound that
retain or improve
the biological effectiveness and properties of the free acids and bases of the
parent compound
as defined herein or that take advantage of an intrinsically basic, acidic or
charged functionality
on the molecule and that are not biologically or otherwise undesirable.
Examples of
pharmaceutically acceptable salts are also described, for example, in Berge et
al.,
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
"Pharmaceutical Salts", J. Pharm. Sci. 66, 1-19 (1977). Non-limiting examples
of such salts
include:
[00145]
(1) acid addition salts, formed on a basic or positively charged
functionality, by
the addition of inorganic acids such as hydrochloric acid, hydrobromic acid,
hydroiodic acid,
sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, carbonate forming
agents, and the like;
or formed with organic acids such as acetic acid, propionic acid, lactic acid,
oxalic, glycolic
acid, pivalic acid, t-butylacetic acid, P-hydroxybutyric acid, valeric acid,
hexanoic acid,
cyclopentanepropionic acid, pyruvic acid, malonic acid, succinic acid, malic
acid, maleic acid,
fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-
hydroxybenzoyl)benzoic acid,
cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-
ethanedisulfonic
acid, 2-hydroxyethanesulfonic acid, cyclohexylaminosulfonic acid,
benzenesulfonic acid,
sulfanilic acid, 4-chlorobenzenesulfonic acid, 2-napthalenesulfonic acid, 4-
toluenesulfonic
acid, camphorsulfonic acid, 3-phenyl propionic acid, lauryl sulphonic acid,
lauryl sulfuric acid,
oleic acid, palmitic acid, stearic acid, lauric acid, embonic (pamoic) acid,
palmoic acid,
pantothenic acid, lactobionic acid, alginic acid, galactaric acid,
galacturonic acid, gluconic
acid, glucoheptonic acid, glutamic acid, naphthoic acid, hydroxynapthoic acid,
salicylic acid,
ascorbic acid, stearic acid, muconic acid, and the like;
[00146]
(2) base addition salts, formed when an acidic proton present in the
parent
compound either is replaced by a metal ion, including, an alkali metal ion
(e.g., lithium, sodium,
potassium), an alkaline earth ion (e.g., magnesium, calcium, barium), or other
metal ions such
as aluminum, zinc, iron and the like; or coordinates with an organic base such
as ammonia,
ethylamine, diethylamine, ethylenediamine, N,N1-dibenzylethylenediamine,
ethanolamine,
diethanolamine, triethanolamine, tromethamine, N-methylglucamine, piperazine,
chloroprocain, procain, choline, lysine and the like.
[00147]
Pharmaceutically acceptable salts may be synthesized from a parent
compound
that contains a basic or acidic moiety, by conventional chemical methods.
Generally, such salts
are prepared by reacting the free acid or base forms of compounds with a
stoichiometric amount
of the appropriate base or acid in water or in an organic solvent, or in a
mixture of the two.
Salts may be prepared in situ, during the final isolation or purification of a
compound or by
separately reacting a compound in its free acid or base form with the desired
corresponding
base or acid, and isolating the salt thus formed. The term "pharmaceutically
acceptable salts"
also include zwitterionic compounds containing a cationic group covalently
bonded to an
91
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
anionic group, as they are "internal salts". It should be understood that all
acid, salt, base, and
other ionic and non-ionic forms of compounds described herein are intended to
be
encompassed. For example, if a compound is shown as an acid herein, the salt
forms of the
compound are also encompassed. Likewise, if a compound is shown as a salt, the
acid and/or
basic forms are also encompassed.
[00148]
Compounds provided herein may contain unnatural proportions of atomic
isotopes at one or more of the atoms that constitute such compounds. Unnatural
proportions of
an isotope may be defined as ranging from the amount found in nature to an
amount consisting
of 100% of the atom in question. For example, compounds may incorporate
radioactive
isotopes, such as for example tritium (3H), iodine-125 (1250 (14C) ,
or carbon-14 or non-
radioactive isotopes, such as deuterium (2H) or carbon-13 (13C). Such isotopic
variations can
provide additional utilities to those described elsewhere within this
application. For instance,
isotopic variants of the compounds of the invention may find additional
utility, including but
not limited to, as diagnostic and/or imaging reagents, or as
cytotoxic/radiotoxic therapeutic
agents. Additionally, isotopic variants can have altered pharmacokinetic and
pharmacodynamic characteristics which can contribute to enhanced safety,
tolerability or
efficacy during treatment. All isotopic variations of compounds provided
herein, whether
radioactive or not, are intended to be encompassed herein.
[00149]
Isotopic enrichment is a process by which the relative abundance of the
isotopes
of a given element are altered, thus producing a form of the element that has
been enriched
(i.e., increased) in one particular isotope and reduced or depleted in its
other isotopic forms. As
used herein, an "isotope-enriched" compound or derivative refers to a compound
in which one
or more specific isotopic form has been increased, i.e., one or more of the
elements has been
enriched (i.e., increased) in one or more particular isotope. Generally, in an
isotope-enriched
compound or derivative, a specific isotopic form of an element at a specific
position of the
compound is increased. It should be understood however that isotopic forms of
two or more
elements in the compound may be increased. Further, an isotope-enriched
compound may be a
mixture of isotope-enriched forms that are enriched for more than one
particular isotope, more
than one element, or both. As used herein, an "isotope-enriched" compound or
derivative
possesses a level of an isotopic form that is higher than the natural
abundance of that form. The
level of isotope-enrichment will vary depending on the natural abundance of a
specific isotopic
form. In some embodiments, the level of isotope-enrichment for a compound, or
for an element
in a compound, may be from about 2 to about 100 molar percent (%), e.g., about
2%, about
92
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
5%, about 17%, about 30%, about 51%, about 83%, about 90%, about 95%, about
96%, about
97%, about 98%, greater than about 98%, about 99%, or 100%.
[00150]
As used herein, an "element of natural abundance" and an "atom of natural
abundance" refers to the element or atom respectively having the atomic mass
most abundantly
found in nature. For example, hydrogen of natural abundance is 1H (protium);
nitrogen of
natural abundance is 14N; oxygen of natural abundance is 160; carbon of
natural abundance is
and so on. A -non-isotope enriched- compound is a compound in which all the
atoms or
elements in the compound are isotopes of natural abundance, i.e., all the
atoms or elements
have the atomic mass most abundantly found in nature.
[00151]
The terms "patient" and "subject" are used interchangeably herein to refer
to a
human or a non-human animal (e.g., a mammal).
[00152]
The terms "administration", "administer" and the like, as they apply to,
for
example, a subject, cell, tissue, organ, or biological fluid, refer to contact
of, for example, an
inhibitor of CD73, a pharmaceutical composition comprising same, or a
diagnostic agent to the
subject, cell, tissue, organ, or biological fluid. In the context of a cell,
administration includes
contact (e.g., in vitro or ex vivo) of a reagent to the cell, as well as
contact of a reagent to a
fluid, where the fluid is in contact with the cell.
[00153]
The terms "treat", "treating", treatment" and the like refer to a course
of action
(such as administering an inhibitor of CD73 or a pharmaceutical composition
comprising same)
initiated after a disease, disorder or condition, or a symptom thereof, has
been diagnosed,
observed, and the like, so as to eliminate, reduce, suppress, mitigate, or
ameliorate, either
temporarily or permanently, at least one of the underlying causes of a
disease, disorder, or
condition afflicting a subject, or at least one of the symptoms associated
with a disease,
disorder, condition afflicting a subject. Thus, treatment includes inhibiting
(e.g., arresting the
development or further development of the disease, disorder or condition or
clinical symptoms
association therewith) an active disease.
[00154]
The term "in need of treatment" as used herein refers to a judgment made
by a
physician or other caregiver that a subject requires or will benefit from
treatment. This
judgment is made based on a variety of factors that are in the realm of the
physician's or
caregiver's expertise.
[00155]
The terms "prevent", "preventing", "prevention" and the like refer to a
course of
action (such as administering a CD73 inhibitor or a pharmaceutical composition
comprising
93
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
same) initiated in a manner (e.g., prior to the onset of a disease, disorder,
condition or symptom
thereof) so as to prevent, suppress, inhibit or reduce, either temporarily or
permanently, a
subject's risk of developing a disease, disorder, condition or the like (as
determined by, for
example, the absence of clinical symptoms) or delaying the onset thereof:
generally in the
context of a subject predisposed to having a particular disease, disorder or
condition. In certain
instances, the terms also refer to slowing the progression of the disease,
disorder or condition
or inhibiting progression thereof to a harmful or otherwise undesired state.
[00156]
The term "in need of prevention" as used herein refers to a judgment made
by a
physician or other caregiver that a subject requires or will benefit from
preventative care. This
judgment is made based on a variety of factors that are in the realm of a
physician's or
caregiver's expertise.
[00157]
The terms "therapeutically effective amount" and -effective amount" are
used
interchangeably herein to refer to the administration of an agent to a
subject, either alone or as
part of a pharmaceutical composition and either in a single dose or as part of
a series of doses,
in an amount capable of having any detectable, positive effect on any symptom,
aspect, or
characteristic of a disease, disorder or condition when administered to the
subject. The
therapeutically effective amount can be ascertained by measuring relevant
physiological
effects, and it can be adjusted in connection with the dosing regimen and
diagnostic analysis
of the subject's condition, and the like. By way of example, measurement of
the serum level of
a CD73 inhibitor (or, e.g., a metabolite thereof) at a particular time post-
administration may be
indicative of whether a therapeutically effective amount has been used. In
some embodiments,
the terms "therapeutically effective amount" and "effective amount" refer to
the amount or dose
of a therapeutic agent, such as a compound, upon single or multiple dose
administration to a
subject, which provides the desired therapeutic, diagnostic, or prognostic
effect in the subject.
An effective amount can be readily determined by an attending physician or
diagnostician using
known techniques and by observing results obtained under analogous
circumstances. In
determining the effective amount or dose of compound administered, a number of
factors are
considered including, but not limited to: the size, age, and general health of
the subject; the
specific disease involved; the degree of or involvement or the severity of the
disease or
condition to be treated; the response of the individual subject; the
particular compound
administered; the mode of administration; the bi availability characteristics
of the preparation
administered; the dose regimen selected; the use of concomitant medication(s);
and other
relevant considerations.
94
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00158]
The term "substantially pure" is used herein to indicate that a component
makes
up greater than about 50% of the total content of the composition, and
typically greater than
about 60% of the total content. More typically, "substantially pure" refers to
compositions in
which at least 75%, at least 85%), at least 90% or more of the total
composition is the
component of interest. In some cases, the component of interest will make up
greater than about
90%), or greater than about 95%) of the total content of the composition.
[00159]
As used herein, the terms "CD73-associated disease, disorder or condition-
and
"disease, disorder or condition mediated by CD73" are used interchangeably to
refer to any
disease, disorder or condition for which treatment with a CD73 inhibitor may
be beneficial. In
general, CD73-associated or mediated diseases, disorders and conditions are
those in which
CD73 activity plays a biological, mechanistic, or pathological role. Such
diseases, disorders
and conditions may also be associated with activity of one or more adenosine
receptors. Non-
limiting examples of CD73-associated diseases, disorders and conditions
include oncology-
related disorders (cancers, tumors, etc.), immune-related disorders, disorders
with an
inflammatory component, microbial-related disorders, CNS-related and
neurological disorders,
and other disorders (such as, without limitation, cardiovascular,
gastrointestinal, metabolic,
hepatic, pulmonary, ophthalmologic, and renal disorders).
[00160]
For example, a CD73 inhibitor may be used to prevent or treat a
proliferative
condition, cancer or tumor; to increase or enhance an immune response; to
improve
immunization, including increasing vaccine efficacy; and to increase
inflammation. Immune
deficiencies associated with immune deficiency diseases, immunosuppressive
medical
treatment, acute and/or chronic infection, and aging can be treated using CD73
inhibitors
disclosed herein. CD73 inhibitors can also be used to stimulate the immune
system of patients
suffering from iatrogenically-induced immune suppression, including those who
have
undergone bone marrow transplants, chemotherapy, or radiotherapy. In other
embodiments, a
CD73 inhibitor may be used to treat or prevent any viral, bacterial, fungal,
parasitic or other
infective disease, disorder or condition, including without limitation HIV and
AIDS.
[00161]
In some embodiments, a CD73 inhibitor may be used to prevent or treat an
immune-related disease, disorder or condition selected from the group
consisting of rheumatoid
arthritis, kidney failure, lupus, asthma, psoriasis, colitis, pancreatitis,
allergies, fibrosis, anemia
fibromyalgia, Alzheimer's disease, congestive heart failure, stroke, aortic
valve stenosis,
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
arteriosclerosis, osteoporosis, Parkinson's disease, infections, Crohn's
disease, ulcerative
colitis, allergic contact dermatitis, eczema, systemic sclerosis and multiple
sclerosis.
[00162] Pharmaceutical compositions provided herein can be
formulated to be compatible
with the intended method or route of administration; exemplary routes of
administration are set
forth herein. Furthermore, the pharmaceutical compositions may be used in
combination with
other therapeutically active agents or compounds as described herein in order
to treat or prevent
the CD73-associated diseases, disorders and conditions as contemplated herein.
1001631 Pharmaceutical compositions containing the active
ingredient (e.g., a CD73
inhibitor) may be in a form suitable for oral use, for example, as tablets,
capsules,
troches,lozenges, aqueous or oily suspensions, dispersible powders or
granules, emulsions,
hard or sofi: capsules, or syrups, solutions, microbeads or elixirs.
Pharmaceutical compositions
intended for oral use may be prepared according to any method known in the art
for the
manufacture of pharmaceutical compositions, and such compositions may contain
one or more
agents such as, for example, sweetening agents, flavoring agents, coloring
agents and
preserving agents in order to provide pharmaceutically acceptable
preparations. Tablets,
capsules and the like generally contain the active ingredient in admixture
with non-toxic
pharmaceutically acceptable carriers or excipients which are suitable for the
manufacture of
tablets. These carriers or excipients may be, for example, diluents, such as
calcium carbonate,
sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating
and
disintegrating agents, for example, corn starch, or alginic acid; binding
agents, for example
starch, gelatin or acacia, and lubricating agents, for example magnesium
stearate, stearic acid
or talc.
[00164] Tablets, capsules and the like suitable for oral
administration may be uncoated or
coated using known techniques to delay disintegration and absorption in the
gastrointestinal
tract and thereby provide a sustained action. For example, a time-delay
material such as
glyceryl monostearate or glyceryl distearate may be employed. They may also be
coated by
techniques known in the art to form osmotic therapeutic tablets for controlled
release.
Additional agents include biodegradable or biocompatible particles or a
polymeric substance
such as polyesters, polyamine acids, hydrogel, polyvinyl pyrrolidone,
polyanhydrides,
polygly colic acid, ethylenevinylacetate, methycellulose,
carboxymethylcellulose, protamine
sulfate, or lactide/glycolide copolymers, polylactide/glycolide copolymers, or
ethylenevinylacetate copolymers in order to control delivery of an
administered composition.
96
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
For example, the oral agent can be entrapped in microcapsules prepared by
coacervation
techniques or by interfacial polymerization, by the use of
hydroxymethylcellulose or gelatin-
microcapsules or poly (methylmethacrolate) microcapsules, respectively, or in
a colloid drug
delivery system. Colloidal dispersion systems include macromolecule complexes,
nano-
capsules, microspheres, microbeads, and lipid-based systems, including oil-in-
water
emulsions, micelles, mixed micelles, and liposomes. Methods for the
preparation of the above-
mentioned formulations will be apparent to those skilled in the art.
[00165] Formulations for oral use may also be presented as hard
gelatin capsules wherein
the active ingredient is mixed with an inert solid diluent, for example,
calcium carbonate,
calcium phosphate, kaolin or microcrystalline cellulose, or as soft gelatin
capsules wherein the
active ingredient is mixed with water or an oil medium, for example peanut
oil, liquid paraffin,
or olive oil. Aqueous suspensions contain the active materials in admixture
with excipients
suitable for the manufacture thereof Such excipients can be suspending agents,
for example
sodium carboxymethylcellulose, methykellulose, hydroxy-propylmethylcellulose,
sodium
alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or
wetting agents,
for example a naturally-occurring phosphatide (e.g., lecithin), or
condensation products of an
alkylene oxide with fatty acids (e.g., polyoxy-ethylene stearate), or
condensation products of
ethylene oxide with long chain aliphatic alcohols (e.g., for
heptadecaethyleneoxycetanol), or
condensation products of ethylene oxide with partial esters derived from fatty
acids and a
hexitol (e.g., polyoxyethylene sorbitol monooleate), or condensation products
of ethylene
oxide with partial esters derived from fatty acids and hexitol anhydrides
(e.g., polyethylene
sorbitan monooleate). The aqueous suspensions may also contain one or more
preservatives.
[00166] Oily suspensions may be formulated by suspending the
active ingredient in a
vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil,
or in a mineral oil
such as liquid paraffin. The oily suspensions may contain a thickening agent,
for example
beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set
forth above, and
flavoring agents may be added to provide a palatable oral preparation.
[00167] Dispersible powders and granules suitable for
preparation of an aqueous
suspension by the addition of water provide the active ingredient in admixture
with a dispersing
or wetting agent, suspending agent and one or more preservatives. Suitable
dispersing or
wetting agents and suspending agents are known in the art.
97
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00168]
Pharmaceutical compositions of the present invention may also be in the
form of
oil-in-water emulsions. The oily phase may be a vegetable oil, for example
olive oil or arachis
oil, or a mineral oil, for example, liquid paraffin, or mixtures of these.
Suitable emulsifying
agents may be naturally occurring gums, for example, gum acacia or gum
tragacanth; naturally
occurring phosphatides, for example, soy bean, lecithin, and esters or partial
esters derived
from fatty acids; hexitol anhydrides, for example, sorbitan monooleate; and
condensation
products of partial esters with ethylene oxide, for example, polyoxyethylene
sorbitan
monooleate.
[00169]
Pharmaceutical compositions typically comprise a therapeutically effective
amount of a CD84 inhibitor compound provided herein and one or more
pharmaceutically and
physiologically acceptable formulation agents. Suitable pharmaceutically
acceptable or
physiologically acceptable diluents, carriers or excipients include, but are
not limited to,
antioxidants (e.g., ascorbic acid and sodium bi sulfate), preservatives (e.g.,
benzyl alcohol,
methyl parabens, ethyl or n-propyl, p-hydroxybenzoate), emulsifying agents,
suspending
agents, dispersing agents, solvents, fillers, bulking agents, detergents,
buffers, vehicles,
diluents, and/or adjuvants. For example, a suitable vehicle may be
physiological saline solution
or citrate buffered saline, possibly supplemented with other materials common
in
pharmaceutical compositions for parenteral administration. Neutral buffered
saline or saline
mixed with serum albumin are further exemplary vehicles. Those skilled in the
art will readily
recognize a variety of buffers that can be used in the pharmaceutical
compositions and dosage
forms contemplated herein. Typical buffers include, but are not limited to,
pharmaceutically
acceptable weak acids, weak bases, or mixtures thereof As an example, the
buffer components
can be water soluble materials such as phosphoric acid, tartaric acids, lactic
acid, succinic acid,
citric acid, acetic acid, ascorbic acid, aspartic acid, glutamic acid, and
salts thereof Acceptable
buffering agents include, for example, a Tris buffer, N-(2-
Hydroxyethyl)piperazine-N'-(2-
ethanesulfonic acid) (I-TEPES), 2-(N-MoqJholino)ethanesulfonic acid (MES), 2-
(N-
Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-
Morpholino)propanesulfonic acid
(MOPS), and Ntris[Hydroxymethyllmethyl-3-arninopropanesulfonic acid (TAPS).
After a
pharmaceutical composition has been formulated, it may be stored in sterile
vials as a solution,
suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such
formulations may
be stored either in a ready-to-use form, a lyophilized form requiring
reconstitution prior to use,
a liquid form requiring dilution prior to use, or other acceptable form.
98
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00170]
In some embodiments, the pharmaceutical composition is provided in a
single-
use container (e.g., a single-use vial, ampoule, syringe, or autoinjector,
whereas a multi-use
container (e.g., a multi-use vial) is provided in other embodiments.
[00171]
Formulations can also include carriers to protect the composition against
rapid
degradation or elimination from the body, such as a controlled release
formulation, including
liposomes, hydrogels, and microencapsulated delivery systems. For example, a
time delay
material such as glyceryl monostearate or glyceryl stearate alone, or in
combination with a
wax, may be employed. Any drug delivery apparatus may be used to deliver a
CD73 inhibitor,
including implants (e.g., implantable pumps) and catheter systems, slow
injection pumps and
devices, all of which are well known to the skilled artisan.
[00172]
Pharmaceutical compositions may also be in the form of a sterile
injectable
aqueous or oleagenous suspension. This suspension may be formulated according
to the known
art using those suitable dispersing or wetting agents and suspending agents
mentioned herein.
The sterile injectable preparation may also be a sterile injectable solution
or suspension in a
non-toxic parenterally-acceptable diluent or solvent, for example, as a
solution in 1,3-butane
diol. Acceptable diluents, solvents and dispersion media that may be employed
include water,
Ringer's solution, isotonic sodium chloride solution, Cremophor ELTM (BASF,
Parsippany,
NJ) or phosphate buffered saline (PBS), ethanol, polyol (e.g., glycerol,
propylene glycol, and
liquid polyethylene glycol), and suitable mixtures thereof In addition,
sterile, fixed oils are
conventionally employed as a solvent or suspending medium. For this purpose
any bland fixed
oil may be employed, including synthetic mono- or diglycerides. Moreover,
fatty acids such as
oleic acid, find use in the preparation of injectables. Prolonged absorption
of particular
injectable formulations can be achieved by including an agent that delays
absorption (e.g.,
aluminum monostearate or gelatin).
[00173]
CD73 inhibitor compounds and compositions provided herein may be
administered to a subject in any appropriate manner known in the art. Suitable
routes of
administration include, without limitation: oral, parenteral (e.g.,
intramuscular, intravenous,
subcutaneous (e.g., injection or implant), intraperitoneal, intracistemal,
intraarticular,
intraperitoneal, intracerebral (intraparenchymal) and
intracerebroventricular), nasal, vaginal,
sublingual, intraocular, rectal, topical (e.g., transdermal), buccal and
inhalation. Depot
injections, which are generally administered subcutaneously or
intramuscularly, may also be
utilized to release the CD73 inhibitors disclosed herein over a defined period
of time. In certain
99
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
embodiments, CD73 inhibitor compounds and compositions are administered orally
to a
subject in need thereof
[00174]
CD73 inhibitor compounds and compositions provided herein may be
administered to a subject in an amount that is dependent upon, for example,
the goal of
administration (e.g., the degree of resolution desired); the age, weight, sex,
and health and
physical condition of the subject to which the formulation is being
administered; the route of
administration; and the nature of the disease, disorder, condition or symptom
thereof The
dosing regimen may also take into consideration the existence, nature, and
extent of any
adverse effects associated with the agent(s) being administered. Effective
dosage amounts and
dosage regimens can readily be determined from, for example, safety and dose-
escalation trials,
in vivo studies (e.g., animal models), and other methods known to the skilled
artisan. In general,
dosing parameters dictate that the dosage amount be less than an amount that
could be
irreversibly toxic to the subject (the maximum tolerated dose (MID)) and not
less than an
amount required to produce a measurable effect on the subject. Such amounts
are determined
by, for example, the pharmacokinetic and pharmacodynamic parameters associated
with
ADME, taking into consideration the route of administration and other factors.
[00175]
In some embodiments, an CD73 inhibitor may be administered (e.g., orally)
at
dosage levels of about 0.01 mg/kg to about 50 mg/kg, or about 1 mg/kg to about
25 mg/kg, of
subject body weight per day, one or more times a day, to obtain the desired
therapeutic effect.
For administration of an oral agent, the compositions can be provided in the
form of tablets,
capsules and the like containing from 1.0 to 1000 milligrams of the active
ingredient,
particularly 1, 3, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400,
500, 600, 750, 800,
900, or 1000 milligrams of the active ingredient.
[00176]
In some embodiments, the dosage of the desired CD73 inhibitor is contained
in a
"unit dosage form". The phrase "unit dosage form" refers to physically
discrete units, each unit
containing a predetemlined amount of the CD73 inhibitor, either alone or in
combination with
one or more additional agents, sufficient to produce the desired effect. It
will be appreciated
that the parameters of a unit dosage form will depend on the particular
agent(s) and the effect
to be achieved.
[00177]
There are also provided herein kits comprising a CD73 inhibitor compound
or
composition.Kits are generally in the form of a physical structure housing
various components
and may be used, for example, in practicing the methods provided herein. For
example, a kit
100
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
may include one or more CD73 inhibitor disclosed herein (provided in, e.g., a
sterile container),
which may be in the form of a pharmaceutical composition suitable for
administration to a
subject. The CD73 inhibitor can be provided in a form that is ready for use
(e.g., a tablet or
capsule) or in a form requiring, for example, reconstitution or dilution
(e.g., a powder) prior to
administration. When the CD73 inhibitors are in a form that needs to be
reconstituted or diluted
by a user, the kit may also include diluents (e.g., sterile water), buffers,
pharmaceutically
acceptable excipients, and the like, packaged with or separately from the CD73
inhibitors.
When combination therapy is contemplated, the kit may contain several
therapeutic agents
separately or they may already be combined in the kit. Each component of the
kit may be
enclosed within an individual container, and all of the various containers may
be within a single
package. A kit of the present invention may be designed for conditions
necessary to properly
maintain the components housed therein (e.g., refrigeration or freezing).
[00178] A kit may also contain a label or packaging insert
including identifying
information for the components therein and instructions for their use (e.g.,
dosing parameters,
clinical pharmacology of the active ingredient(s), including mechanism of
action,
pharmacokinetics and pharmacodynamics, adverse effects, contraindications,
etc.). Labels or
inserts can include manufacturer information such as lot numbers and
expiration dates. The
label or packaging insert may be, e.g., integrated into the physical structure
housing the
components, contained separately within the physical structure, or affixed to
a component of
the kit (e.g., an ampule, tube or vial).
EXAMPLES
[00179] The present invention will be more readily understood by
referring to the
following examples, which are provided to illustrate the invention and are not
to be construed
as limiting the scope thereof in any manner.
[00180] Unless defined otherwise or the context clearly dictates
otherwise, all technical
and scientific terms used herein have the same meaning as commonly understood
by one of
ordinary skill in the art to which this invention belongs. It should be
understood that any
methods and materials similar or equivalent to those described herein can be
used in the
practice or testing of the invention.
Compound Synthesis
[00181] Compounds provided herein can be prepared using
conventional methods and as
described in the Examples below.
101
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
R,NH R,NH
NN
Methylenediphosphonic dichloride 0 0
CI _______________________________________________
,P P-0¨ NNCI
Trimethyl phosphate HO
OH OH
OH OH
OH OH
S-x
[00182]
The amine compound RNH2 was obtained either from a commercial source, or
prepared according to methods described in the literature.
[00183]
Preparation of triethylammonium hydrogen carbonate buffer (TEAC). A 1 M
solution of TEAC was prepared by adding dry ice slowly to a 1 M triethylamine
solution in
water for several hours until the pH of the solution reached approximately 7.4-
7.6 (as measured
using a pH meter).
[00184]
The 2-chloropurine nucleoside derivative S-x (1 mmol, 1 eq.) was dissolved
in
trimethyl phosphate (10 mL). The solution was cooled with an ice-bath. To the
cold solution
was added a solution of bis(dichlorophosphoryOmethane (4.0 eq.) in trimethyl
phosphate (5
mL). The resulting mixture was stirred at 0 C for 2-4 h, and the reaction was
monitored by
thin-layer chromatography (TLC). The reaction was quenched by TEAC solution,
and the pH
of the reaction solution was adjusted to 7-8. The mixture was extracted with
dichloromethane
(DCM) and the aqueous phase was isolated and concentrated. The residual
material was
purified by reversed-phase chromatography with a C18-column, giving the
product as colorless
solid.
Example I. Synthesis of compound 1
[00185]
DIEA (diisopropylethylamine; 7.5 mmol, 969 mg, 1.5 eq.) was added dropwise
to a solution of 2,6-dichloro-9-(2',3',5'-tri-O-acetyl-3-D-
ribofuranosyl)purine (5.0 mmol, 2236
mg, 1.0 eq.) and benzylamine (5.0 mmol, 536 mg, 1.0 eq.) in dioxane (25 mL).
The reaction
mixture was stirred at room temperature overnight. The solvent was evaporated
in vacuo and
the residue was dissolved in DCM (100 mL), washed with water (2 x 30 mL). The
crude
product was purified by column chromatography. The intermediate was dissolved
in 50 mL
NH3/CH3OH solution and stirred at 35 C overnight. The solvent was evaporated
in vacuo; and
102
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
the residual material was purified by column chromatography, giving 2-
chloropurine
nucleoside derivative S-1 (1818 mg).
[00186] S-1 (1.0 mmol, 392 mg, 1.0 eq.) was dissolved in
trimethyl phosphate (10 mL),
cooled in an ice bath. To the cold solution was added a solution of
bis(dichlorophosphoryl)methane (4.0 eq.) in trimethyl phosphate (5 mL). The
reaction mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution, and the pH of the reaction solution was adjusted to
7-8. The
mixture was extracted with DCM and the aqueous phase was isolated, and
concentrated. The
residual material was purified by reversed-phase column chromatography (C18-
column),
providing compound 1 as colorless solid (369 mg): 1H NMR (500 MHz, CD30D-d4) 6
ppm
2.46 (t, 2H) , 4.23-4.74 (m, 7H) , 6.01 (d, 1H) , 7.19-7.38 (m, 5H) , 8.59 (s
, 1H) ; 13C NMR (125 MHz, CD3C1-d3) 6 ppm 40.12, 43.84, 63.92, 69.87, 74.64,
83.71, 88.50, 115.22, 126.92, 127.47, 128.13, 149.27, 154.08, 154.96, 160.55;
31P NMR (200 MHz, CD3C1-d3) 6 ppm 12.94, 18.11; m/z (ESL') 550.1.
Example 2. Synthesis of compound 6
[00187] DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a
solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acety1-13-D-ribofuranosyl)purine (5.0 mmol, 2.2 g,
1.0 eq.) and 1-
naphthalenemethanamine (5.0 mmol, 786 mg, 1.0 eq.) in dioxane (25 mL). The
reaction
mixture was stirred at room temperature overnight. The solvent was evaporated
in vacua and
the residue was dissolved in DCM (100 mL), washed with water (2 x 30 mL).
Solvent was
removed (using a rotary evaporator), and the residual material was purified by
column
chromatography. The intermediate was dissolved in 50-mL NH3/CH3OH solution and
stirred
at 35 C overnight. Solvent was evaporated in vacua and the residual material
was purified by
column chromatography, giving S-6 (1.3 g).
[00188] S-6 (1.0 mmol, 442 mg, 1.0 eq.) was dissolved in
trimethyl phosphate (10 mL),
cooled in an ice bath. To the cold solution was added a solution of
bis(dichlorophosphoryOmethane (4.0 eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution, and the pH of the reaction mixture was adjusted to
7-8. The
mixture was extracted with DCM. The aqueous phase was isolated, concentrated;
and the
residual material was purified by reversed-phase chromatography (C18-column),
giving
103
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
compound 6 as a colorless solid (110 mg): 1H NMR (500 MHz, D20) 6 ppm 0.88-
0.91 (m, 3
H), 1.34-1.43 (m, 4 H), 1.67-1.68 (m, 2H), 2.15-2.26 (m, 2H), 4.16-4.22 (m,
2H), 4.25-4.31
(m, 2H), 4.38-4.41 (m, 1H), 4.53-4.57 (m, 1H), 4.74-4.76 (m, H), 6.13-1.15 (m,
1H), 8.70-8.75
(m, 1H); 13C NMR (125 MHz, D20) 6 ppm 13.20, 21.60, 27.24, 27.55. 63.46,
67.12, 70.15,
74.34, 84.08, 84.14, 87.38, 120.98, 142.83, 150.24, 152.49, 153.22;31P NMR
(200 MHz, D20)
6 ppm 16.15, 18.97; rn/z (ES-) 571.8.
Example 3. Synthesis of compound 7
1001891 DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a
solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acetyl-13-D-ribofuranosyl)purine (5.0 mmol, 2.2 g,
1.0 eq.) and 2-
naphthalenemethanamine (5.0 mmol, 786 mg, 1.0 eq.) in dioxane (25 mL). The
reaction was
stirred at room temperature overnight. The solvent was evaporated in vacuo and
the residue
was dissolved in DCM (100 mL), washed with water (2 x 30 mL). The crude
product was
purified by column chromatography. The intermediate was dissolved in 50 mL
NH3/CH3OH
solution and stirred at 35 C overnight. The solvent was evaporated in vacuo
and the residual
material was purified by column chromatography, giving 2-chloropurine
nucleoside derivative
S-7 (1.15 g).
[00190] S-7 (1.0 mmol, 442 mg, 1.0 eq.) was dissolved in
trimethyl phosphate (10 niL),
and the solution was cooled with an ice-bath. To the cold solution was added a
solution of
bis(dichlorophosphoryl)methane (4.0 eq.) in trimethyl phosphate (5 mL). The
reaction mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution, and the pH of the reaction mixture was adjusted to
7-8. The
mixture was extracted with DCM; and the aqueous phase was isolated and
concentrated. The
residual material was purified by reversed-phase chromatography (C18-column),
providing
compound 7 as a colorless solid (105 mg): 1H NMR (500 MHz, D20) 6 ppm 2.11 (t,
J = 19.7
Hz, 2H), 4.10 (s, 2H), 4.29 (s, 1H), 4.42 (s, 1H), 4.55 (s, 1H), 4.88 (s, 2H),
5.79 (s, 1H), 7.36-
7.44(m, 4H), 7.71 (d, J = 7.9 Hz, 1H), 7.76 (d, J = 7.2 Hz, 1H), 7.87 (d, J =
7.8 Hz, 1H), 8.13
(s, 1H); 13C NMR (125 MHz, D20) 6 ppm 27.57, 42.23, 63.39, 70.05, 74.16,
83.59, 86.92,
117.78, 122.74, 125.48, 125.88, 126.05, 126.26, 128.24, 128.43, 130.48,
132.19, 133.08,
139.02, 148.62, 153.86, 154.44. 31P NMR (202 MHz, D20) 6 ppm 15.17, 19.58; m/z
(ES)
598.2.
Example 4. Synthesis of compound 8
104
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1001911 The 2-chloropurine nucleoside derivative S-8 (1.0 mmol,
415 mg, 1.0 eq.) was
dissolved in trimethyl phosphate (10 mL); and the solution was cooled with an
ice-bath. To the
cold solution was added a solution of bis(dichlorophosphoryl)methane (4.0 eq.)
in trimethyl
phosphate (5 mL), while the ice bath was applied. The resulting mixture was
stirred at 0 C for
2-4 h; and the reaction was monitored by TCL. The reaction was quenched by
TEAC solution;
and the pH of the reaction mixture was adjusted to 7-8. The mixture was
extracted with DCM;
and the aqueous phase was isolated and concentrated. The residual material was
purified
(reversed-phase column chromatography, C18-column), to obtain the product as
colorless solid
(58 mg). 1H NMR (500 MHz, D20) 6 ppm 2.12 (t, J = 19.7 Hz, 2H), 4.08 (s, 2H),
4.27 (s, 1H),
4.40 (s, 1H), 4.53 (s, 1H), 4.65 (s, 2H), 5.75 (s, 1H), 7.36 (s, 3H), 7.65 (s,
2H), 7.69 (d, J = 8.0
Hz, 2H), 8.28 (s, 1H); 13C NMR (125 MHz, D20) 6 ppm 44.04, 63.41, 70.06,
74.19, 83.73,
86.90, 125.50, 125.60, 126.00, 126.34, 127.38, 128.15, 132.13, 132.71, 135.18,
139.21, 153.96,
154.73; 31P NMR (202 MHz, D20) 6 ppm 15.86, 19.01; miz (ES-) 598.4.
Example 5. Synthesis of compound 9
1001921 DIEA (12.5 mmol, 1.6 g, 2.5 eq.) was added dropwise to a
solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acety1-13-D-ribofuranosyl)purine (5.0 mmol, 2.2g,
1.0 eq.) and
memantine hydrochloride (5.0 mmol, 1.0 g, 1.0 eq.) in 25-mL dioxane. The
reaction mixture
was stirred at room temperature overnight. The solvent was evaporated in vacuo
and the residue
was dissolved in DCM (100 mL), washed with water (2 x 30 mL), and
concentrated. The
residual material was purified by column chromatography, to give the
intermediate. The
intermediate was dissolved in 50 mL NH3/CH3OH solution and the mixture was
stirred at 35
C overnight. The solvent was evaporated in vacuo and the residual material was
purified by
column chromatography, giving 2-chloropurine nucleoside derivative S-9 (1.1
g).
1001931 S-9 (1.0 mmol, 463 mg, 1.0 eq.) was dissolved in
trimethyl phosphate (10 mL);
and the mixture was cooled with an ice-bath. To the cold mixture was added a
solution of
bis(dichlorophosphoryl)methane (4 eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. After
the reaction
finished, the reaction was quenched by TEAC solution. The pH of the mixture
was adjusted to
7-8. The mixture was extracted with DCM and the aqueous phase was isolated.
The aqueous
solution was concentrated; and the residue was purified by reversed-phase
column
chromatography (C18 column), providing compound 9 as a colorless solid (200
mg): 1H NMR
(500 MHz, D20) 6 ppm 0.76 (s, 6H), 0.98 -1.12 (m, 2H), 1.22 (s, 2H), 1.31 (d,
J = 11.3 Hz,
105
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
2H), 1.70 (dd, J = 29.4, 11.9 Hz, 4H), 1.90 (s, 2H), 2.11 (t, J = 19.9 Hz,
3H), 4.07 (s, 2H),4.28
(s, 1H) , 4.44 (dd, J = 6.5, 2.4 Hz, 1H), 4.67 - 4.63 (m, 1H), 5.91 (d, J =
5.7 Hz, 1H),8.33 (s,
1H) ; 13C NMR (125 MHz, D20) 6 ppm 25.96, 26.95, 29.97, 32.08, 39.23, 42.32,
50.32, 55.08,
63.73, 70.04, 74.33, 83.78, 87.45, 116.10, 138.34, 148.62, 153.57, 154.16; 31P
NMR (200 MHz,
D20) 6 ppm 18.12; m/z (ES-) 620.2.
Example 6. Synthesis of compound /0
[00194] The 2-chloropurine nucleoside derivative S-10 (1.0 mmol,
451 mg, 1.0 eq.) was
dissolved in trimethyl phosphate (10 mL), and the solution was cooled in an
ice-bath. To the
cold solution was added a solution of bis(dichlorophosphoryOmethane (4eq.) in
trimethyl
phosphate (5 mL). The resulting mixture was stirred at 0 C for 2-4 h, and the
reaction was
monitored by TLC. The reaction was quenched by TEAC solution; and the pH of
the reaction
solution was adjusted to 7-8. The mixture was extracted with DCM and the
aqueous phase was
isolated and concentrated. The residual material was purified by reversed-
phase column
chromatography (C18-column), giving compound 10 as a colorless solid (60 mg):
11-1 NMR
(500 MHz, D20) 6 ppm 2.27 (t, J = 19.4 Hz, 2H), 4.17 (s, 2H), 4.35 (s, 1H),
4.46 (s, 1H), 6.02
(s, 1H), 7.27 (s, 4H), 7.54 (s, 2H), 7.97 (s, 2H), 8.33 (s, 1H) ; 13C NMR (125
MHz, D20) .5
ppm 16.73, 25.47, 26.48 , 27.50, 57.39, 63.69, 70.09, 74.31, 83.84, 87.70,
113.16, 120.00,
122.84, 124.66, 126.42, 138.03, 143.08, 148.86, 152.70, 154.12 ; 31P NMR (200
MHz, D20) 6
ppm 17.43, 19.35- 19.97; miz (ES-) 608Ø
Example 7. Synthesis of compound //
[00195] The 2-Chloropurine nucleoside derivative S-11 (1.0 mmol,
564 mg, 1.0 eq.) was
dissolved in trimethyl phosphate (10 mL). The solution was cooled in an ice-
bath; and to the
cold solution was added a solution of bis(dichlorophosphoryl)methane (4 eq.)
in trimethyl
phosphate (5 mL). The resulting mixture was stirred at 0 C for 2-4 h; and the
reaction was
monitored by TLC. After being quenched by TEAC solution, the pH of the
reaction mixture
was adjusted to 7-8. This mixture was extracted with DCM and the aqueous phase
was isolated
and concentrated. The residual material was purified by reversed-phase column
chromatography (C18-column), providing compound 11 as a colorless solid (100
mg): 1H
NMR (500 MHz, D20) 6 ppm 0.77 (d, J = 6.5 Hz, 3H), 1.22 (s, 14H), 1.55 (s,
2H), 1.92 (s,
4H), 2.12 (t, J = 19.8 Hz, 2H), 2.67 (d, J = 38.2 Hz, 4H), 4.03 (s, 2H), 4.19
(s, 1H), 4.40 (s,
1H), 4.60 (s, 1H), 5.20 (d, J = 5.6 Hz, 4H), 5.88 (d, J = 4.4 Hz, 1H),8.51 (s,
1H) ; 13C NMR
(125 MHz, D20) 6 ppm 13.90 , 22.48 , 24.71 , 25.52, 27.10 , 29.26 , 29.64,
31.41, 36.86,
106
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
37.49, 63.89, 70.44 ,74.37, 84.15 , 86.83, 119.95, 127.82, 129.70, 149.34,
152.45, 152.86,
164.88 ,174.66; 31P NMR (200 MHz, D20) 6 ppm 16.04 ,18.65; m/z (ES-) 720.4.
Example 8. Synthesis of compound 12
1001961 DlEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a
solution of 2,6-
dichloro-9-(2',3',5'46-0-ace1yl-f3-D-ribofuranosyl)purine (5.0 mmol, 2.23 g,
1.0 eq.) and 2-
aminonaphthalene (5.0 mmol, 715 mg, 1.0 eq.) in 25-mL dioxane. The reaction
mixture was
stirred at room temperature overnight. The solvent was evaporated in vacuo;
and the residue
was dissolved in DCM (100 mL), washed with water (2 x 30 mL). The DCM solution
was
concentrated to dryness; and the residual material was purified by column
chromatography,
giving an intermediate. The intermediate was dissolved in 50 mL NH3/CH3OH
solution and
the mixture was stirred at 35 C overnight. The solvent was evaporated in
vacuo and the
residual material was purified by column chromatography, providing compound S-
12 (670
mg).
1001971 S-12 (1.0 mmol, 427 mg, 1.0 eq.) was dissolved in
trimethyl phosphate (10 mL),
and the solution was cooled in an ice-bath. To the cold solution was added a
solution of
bis(dichlorophosphoryl)methane (4 eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 'V for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution, and the pH of the quenched reaction mixture was
adjusted to 7-
8. The mixture was extracted with DCM; and the aqueous phase was isolated and
concentrated.
The residual material was purified by reversed-phase column chromatography
(C18-column),
giving compound 12 as a colorless solid (100 mg): 11-1NMR (D20, 500 MHz) 6 ppm
2.21-2.29
(m, 2 H), 4.20-4.22 (m, 2H), 4.40-4.44 (m, 1H), 4.56-4.60 (m, 1H), 4.78-4.80
(m, 1H), 6.09-
6.10(m, 1H), 7.58-7.64 (m, 3H), 7.71-7.73 (m, 1H), 7.97-7.99 (m, 1H) 8.02-
8.04(m, 2H), 8.55
(s, 1H); 13C NMR (D20, 125 MHz) 6 ppm 26.38, 27.37, 28.36, 63.61, 70.33,
84.01, 86.76,
118.06, 121.57, 122.25, 125.52, 126.23, 126.41, 139.82, 149.74, 153.39,
153.47; 31P NMR
(D20, 200 MHz) 6 ppm 16.14, 18.96; m/z (ES) 583.9.
Example 9. Synthesis of compound 15
1001981 The 2-Chloropurine nucleoside derivative S-15 (1.0 mmol,
513 mg, 1.0 eq.) was
dissolved in trimethyl phosphate (10 mL). The mixture was cooled in an ice-
bath, followed by
addition of a solution of bis(dichlorophosphoryOmethane (4eq.) in trimethyl
phosphate (5 mL).
The resulting mixture was stirred at 0 C for 2-4 h, and the reaction was
monitored by TLC.
The reaction was quenched by TEAC solution; and the pH of the reaction mixture
was adjusted
107
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
to 7-8. The mixture was extracted with DCM; and the aqueous phase was isolated
and
concentrated. The residual material was purified by reversed-phase column
chromatography
(C18-column), giving compound 15 as a colorless solid (30 mg): 1H NMR (500
MHz, D20) 6
ppm 0.70 (t, J = 6.3 Hz. 3H) ,1.06 (s, 16H), 1.27 (s, 2H), 1.58 (s. 2H), 2.10
(t, J = 19.7 Hz, 2H),
4.12 (d, J = 32.1 Hz, 4H), 4.27 (s, 1H), 4.46 (s, 1H), 4.66 (s, 1H), 6.00 (d,
J = 4.7 Hz, 1H),8.60
(s, 1H) ; 13C NMR (125 MHz, D20) 6 ppm 13.73, 22.40, 25.51, 27.55, 28.40,
29.07, 29.36,
31.66, 63.64, 66.70, 70.27, 74.36, 83.95, 87.18, 120.57, 142.65, 150.05,
152.38, 153.21; 31P
NMR (200 MHz, D20) 6 ppm 15.64,18.89; m/z (ES-) 670.1.
Example 10. ,Synthesis of compound 16
[00199] The 2-Chloropurine nucleoside derivative S-16 (1.0 mmol,
478 mg, 1.0 eq.) was
dissolved in trimethyl phosphate (10 mL); and the mixture was cooled in an ice-
bath, followed
by addition of a solution of bis(dichlorophosphoryl)methane (4 eq.) in
trimethyl phosphate (5
mL). The resulting mixture was stirred at 0 C for 2-4 h; and the reaction was
monitored by
TLC. The reaction was quenched by TEAC solution; and the pH of the mixture was
adjusted
to 7-8. The mixture was extracted with DCM; and the aqueous phase was isolated
and
concentrated. The residual material was purified by reversed-phase column
chromatography
(C18-column), providing compound 16 as a colorless solid (250 mg): 1H NMR (500
MHz,
D20) 6 ppm 1.64 (s, 11H), 1.91 (s, 4H), 2.16 (t, J = 19.8 Hz, 2H), 2.29 (s,
2H), 4.14 (dddd, J =
6.8, 5.9, 4.1, 1.6 Hz, 3H), 4.30-4.37 (m, 1H), 4.56 - 4.46 (m, 1H), 6.11 (d, J
= 5.1 Hz, 1H),8.72
(s, 1H) ;13C NMR (125 MHz, D20) 6 ppm 16.75 , 27.23 , 28.38, 33.41 , 36.07
42.03 , 51.44
, 63.47 , 70.17, 74.37, 84.19, 87.49, 115.91 , 121.90, 143.36, 149.28, 153.15
,173.66; 31P
NMR (200 MHz, D20) 6 ppm 16.37, 18.85; m/z (ES-) 634.1.
Example 11. Synthesis qf compound 17
1002001 DIEA (12.5 mmo1,1.6 g, 2.5 eq.) was added dropwise to a
solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acety1-13-D-ribofuranosyl)purine (5.0 mmol, 2.23 g,
1.0 eq.) and 2-
adamantanamine hydrochloride (5.0 mmol, 0.94 g, 1.0 eq.) in 25-mL dioxane. The
mixture was
stirred at room temperature overnight. The solvent was evaporated in vacua and
the residue
was dissolved in DCM (100 mL), washed with water (2 x 30 mL). The organic
layer was
concentrated; and the residual material was purified by column chromatography,
giving an
intermediate. This intermediate was dissolved in 50 mL NH3/CH3OH solution and
stirred at 35
'V overnight. After evaporation of the solvent, the residual material was
purified by column
chromatography, providing 2-chloropurine nucleoside derivative S-17 (880 mg).
108
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Compound S-17 (1.0 mmol, 435 mg, 1.0 eq.) was dissolved in trimethyl phosphate
(10 mL);
and the mixture was cooled in an ice-bath. To the cold mixture was added a
solution of
bis(dichlorophosphoryl)methane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution; and the pH of the mixture was adjusted to 7-8. The
mixture was
extracted with DCM; and the aqueous phase was isolated and concentrated. The
residual
material was purified by reversed-phase column chromatography (C18-column),
giving
compound 17 as a colorless solid (30 mg): 1H NMR (500 MHz, D20) 6 ppm 1.60 (d,
J = 12.7
Hz, 2H), 1.71 (s, 2H), 1.82 (d, J = 20.8 Hz, 7H), 1.96 (d, J = 19.6 Hz, 4H),
2.14 (t, J = 19.8 Hz,
2H), 4.10 (s, 2H), 4.20 (s, 1H), 4.31 (s, 1H), 4.47 (t, J = 4.2 Hz, 1H), 5.96
(d, J = 5.6 Hz,
I H),8.4 I (s, IH) ; 13C NMR (125 MHz, D20) 6 ppm 16.70, 26.77 , 30.81 , 31.44
, 36.48 , 36.86
, 57.36, 63.51 , 70.23 , 74.20, 83.97 , 86.73, 139.05 ,154.39 ; 31P NMR (200
MHz, D20) 6
ppm 15.85,19.04 ; m/z (ES) 592ØExample 12. Synthesis of compound 18
[00201]
The 2-Chloropurine nucleoside derivative S-18 (1.0 mmol, 512 mg, 1.0 eq.)
was dissolved in trimethyl phosphate (10 mL). The mixture was cooled in an ice-
bath, followed
by addition of a solution of bis(dichlorophosphoryl)methane (4eq.) in
trimethyl phosphate (5
mL). The resulting mixture was stirred at 0 C for 2-4 h; and the reaction was
monitored by
thin layer chromatography. The reaction was quenched by TEAC solution; and the
pH of the
mixture was adjusted to 7-8. The mixture was extracted with DCM; and the
aqueous phase was
isolated and concentrated. The residual material was purified by reversed-
phase column
chromatography (Cl 8-column), giving compound 18 as a colorless solid (70 mg):
1H NMR
(500 MHz, CD30D-d4) 6 ppm 0.93 (t, J = 6.8 Hz, 3H), 1.52 (s, 18H), 1.60 - 1.74
(m, 2H), 2.33
(t, J = 19.8 Hz, 2H), 3.43 (t, J = 6.6 Hz, 2H), 4.27 (d, J = 21.4 Hz, 3H),
4.47 - 4.61 (m, 1H),
4.71 (t, J = 5.3 Hz, 1H), 6.13 (d, J = 5.4 Hz, 1H),8.76 (s, 1H); 13C NMR (125
MHz, CD30D-
d4) 6 ppm 13.01 , 22.30 , 26.62, 28.98 , 29.28, 31.63 , 39.56, 63.95 , 7057,
74.91 , 84.44,
87.86, 118.78 , 142.32 , 150.79 , 151.78, 151.97 ,154.17; 31P NMR (200MHz,
CD30D-d4) 6
ppm 16.03, 20.25; m/z (ES), 669.2.
Example 13. Synthesis of compound 19
[00202]
DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a solution of 2,6-
dich1oro-9-(2',3',5'-tri-O-acety1-3-D-ribofuranosy1)purine (5.0 mmol, 2.23 g,
1.0 eq.) and di-N-
dodecylamine (5.0 mmol, 1.8 g, 1.0 eq.) in 25 mL dioxane. The reaction mixture
was stirred at
room temperature overnight. The solvent was evaporated in vacuo; and the
residue was
dissolved in DCM (100 mL), washed with water (2 x 30 mL). The organic layer
was
109
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
concentrated to dryness; and the residual material was purified by column
chromatography,
giving an intermediate compound. This intermediate was dissolved in 50 mL
NH3/CH3OH
solution and the mixture was stirred at 35 C overnight. After removal of
solvent in vacuo, the
residual material product was purified by column chromatography, giving 2-
chloropurine
nucleoside derivative S-19 (1.3 g).
1002031
S-19 (1.0 mmol, 637 mg, 1.0 eq.) was dissolved in trimethyl phosphate (10
mL). The mixture was cooled in an ice-bath, followed by addition of a solution
of
bis(dichlorophosphoryl)methane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution; and the pH of the reaction solution was adjusted to
7-8. The
mixture was extracted with DCM; and the aqueous phase was isolated and
concentrated. The
residual material was purified by reversed-phase column chromatography (C18-
column),
providing compound 19 as a colorless solid (150 mg): 1H NMR (500 MHz, CD30D-
d4) 6 ppm
0.93 (t, J = 6.6 Hz, 6H), 1.32 - 1.46 (m, 36H), 1.72 (s, 4H), 2.36 (t, J =
20.0 Hz, 2H), 3.70 (s,
2H), 4.11 - 4.32(m, 5H), 4.47 (s, 1H), 4.66 (t, J = 5.2 Hz, 1H), 6.04 (d, J =
5.4 Hz, 1H),8.37 (s,
1H); 13C NMR (125 MHz, CD30D-d4) 6 ppm 13.08, 22.34, 26.44, 29.08 , 29.34,
31.68, 64.24,
70.51, 74.51, 83.84, 87.46 ,118.27, 137.84, 151.64, 153.46, 154.25; 31P NMR
(200 MHz,
CD30D-d4) 6 ppm 16.20,19.99; m/z(ES-) 794.6.
Example 14. Synthesis of compound 20
[00204]
DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a solution of 2,6-
dich1oro-9-(2',3',5'-tri-O-acety1-3-D-ribofuranosy1)purine (5.0 mmol, 2.23 g,
1.0 eq.) and 2-
anthracenamine (5.0 mmol, 1.0 g, 1.0 eq.) in 25 mL dioxane. The reaction
mixture was stirred
at room temperature overnight. The solvent was evaporated in vacuo and the
residue was
dissolved in DCM (100 mL), washed with water (2 x 30 mL). The organic layer
was
concentrated to dryness; and the residual material was purified by column
chromatography,
giving an intermediate compound. This intermediate was dissolved in 50 mL
NH3/CH3OH
solution and stirred at 35 C overnight. The solvent was evaporated in vacuo
and the residual
material was purified by column chromatography, giving 2-chloropurine
nucleoside derivative
S-20 (770 mg).
[00205]
S-20 (1.0 mmol, 477 mg, 1.0 eq.) was dissolved in trimethyl phosphate (10
mL). The mixture was cooled in an ice-bath, followed by addition of a solution
of
bis(dichlorophosphoryl)methane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 'V for 2-4 h; and the reaction was monitored by TLC. The
reaction was
110
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
quenched by TEAC solution; and the pH of the reaction mixture was adjusted to
7-8. The
mixture was extracted with DCM; and the aqueous phase was isolated and
concentrated. The
residual material was purified by reversed-phase column chromatography (C18-
column),
providing compound 20 as a colorless solid (80 mg): 1H NMR (500 MHz, D20) 6
ppm 2.25 (1.,
J = 19.4 Hz, 2H), 4.28 (d, J = 58.1 Hz, 3H), 4.46 (s, 2H), 5.56 (s, 1H), 7.19
(s, 3H), 7.57 (d, J
= 73.7 Hz, 4H) ,7.85 (s, 2H), 8.21 (s, 1H); 31P NMR (200 MHz, D20) 6 ppm
18.42,19.15; m/z
(ES-) 633.9.
Example 15. Synthesis of compound 22
[00206]
DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acetyl-13-D-ribofuranosyl)purine (5.0 mmol, 2.23 g,
1.0 eq.) and 1-
adamantanamin (5.0 mmol, 756 mg, 1.0 eq.) in 25 mL dioxane. The reaction
mixture was
stirred at room temperature overnight. The solvent was evaporated in vacuo and
the residue
was dissolved in DCM (100 mL), washed with water (2 x 30 mL). After removal of
solvent,
the residual material was purified by column chromatography, providing an
intermediate
compound. This intermediate was dissolved in 50 mL NH3/CH3OH solution and the
mixture
was stirred at 35 'V overnight. The solvent was evaporated in vacuo and the
residual material
was purified by column chromatography, giving 2-chloropurine nucleoside
derivative S-22
(770 mg).
[00207]
S-22 (1.0 mmol, 435 mg, 1.0 eq.) was dissolved in trimethyl phosphate (10
mL). The mixture was cooled in an ice-bath, followed by addition of a solution
of
bis(dichlorophosphorypmethane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution; and the pH of the reaction solution was adjusted to
7-8. The
mixture was extracted with DCM; and the aqueous phase was isolated and
concentrated. The
residual material was purified by reversed phase column chromatography (C18-
column),
giving compound 22 as a colorless solid (100 mg): 1H NMR (500 MHz, D20) 6 ppm
1.64 (s,
6H),2.06 (d, J = 33.5 Hz, 9H),2.17 (d, J = 19.9 Hz, 2H), 4.09 (s, 2H), 4.30
(d, J = 0.9 Hz, 1H),
4.45 (ddd, J = 5.9, 3.0, 2.0 Hz, 1H), 4.68 - 4.65 (m, 1H), 5.94 (d, J = 5.2
Hz, 1H),8.38 (s, 1H)
;13C NMR (125 MHz, 1)20) 8 ppm 26.22, 27.22 .29.28 , 35.72 , 40.80 , 53.50 ,
63.57, 70.19,
74.20, 83.95, 86.81, 138.60, 148.74, 153.76,154.43 ; 31P NMR (200 MHz, D20) 6
ppm 16.38,
18.81; m/z (ES-) 592.2.
Example 16. Synthesis of compound 23
111
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00208]
The 2-Chloropurine nucleoside derivative S-23 (1.0 mmol, 481 mg, 1.0 eq.)
was dissolved in trimethyl phosphate (10 mL), and the mixture was cooled in an
ice-bath. To
the cold mixture was added a solution of bis(dichlorophosphoryl)methane (4
eq.) in trimethyl
phosphate (5 mL). The resulting mixture was stirred at 0 C for 2-4 h; and the
reaction was
monitored by TLC. The reaction was quenched by TEAC solution; and the pH of
the reaction
mixture was adjusted to 7-8. The mixture was extracted with DCM; and the
aqueous phase was
isolated and concentrated. The residual material was purified by reversed-
phase column
chromatography (C18-column), giving compound 23 as colorless solid (141 mg):
1I-1 NMR
(D20, 500 MHz) 6 ppm 0.90-0.97 (m, 9H), 1.18-1.20 (m, 1 H), 1.36-1.40 (m, 1
H), 1.75-1.80
(m, 1H), 1.90-1.97 (m, 1H), 2.00-2.30 (m, 2H), 2.40-2.45 (m, 1H), 4.20-4.25
(m, 2H), 4.40-
4.44 (m, I H), 4.44-4.57 (m, I H), 4.72-4.73 (m, I H), 4.91-5.05 (m, I H),
6.43-6.45 (m, I H),
9.02 (m, 1H); 13C NMR (D20, 125 MHz) 6 12.74, 18.03, 18.90, 35.91, 44.47,
47.41, 48.45,
63.50, 70.16, 74.34, 83.22, 84.06, 84.13, 87.37, 120.84, 142.72, 150.23,
152.41, 153.24, 153.45
ppm; 31P NMR (D20, 200 MHz) 6 ppm 16.49, 18.79; m/z (ES-) 638Ø
Example 17. Synthesis of compound 31
[00209]
DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acety1-13-D-ribofuranosyl)purine (5.0 mmol, 2.23 g,
1.0 eq.) and 3-
azaspiro[4.51decane (5.0 mmol, 696 mg, 1.0 eq.) in 25 mL dioxane. The reaction
mixture was
stirred at room temperature overnight. Solvent was evaporated in vacua and the
residue was
dissolved in DCM (100 mL), washed with water (2 x 30 mL). The organic layer
was evaporated
to dryness, and the residual material was purified by column chromatography,
providing an
intermediate compound. This intermediate was dissolved in 50 mL NH3/CH3OH
solution and
stirred at 35 `DC overnight. Solvent was evaporated in vacuo; and the residual
material was
purified by column chromatography, giving the corresponding 2-chloropurine
nucleoside
derivative, S-31 (1.1 g).
[00210]
S-31 (1.0 mmol, 423 mg, 1.0 eq.) was dissolved in trimethyl phosphate (10
mL); and the mixture was cooled in an ice-bath. To the cold mixture was added
a solution of
bis(dichlorophosphoryl)methane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TI.C. The
reaction was
quenched by TEAC solution; and the pH of the mixture was adjusted to 7-8. The
mixture was
extracted with DCM; and the aqueous phase was collected and concentrated. The
residual
material was purified by reversed-phase column chromatography (C18-column),
giving
compound 31 as a colorless solid (140 mg): 1H NMR (500 MHz, D20) 6 ppm 1.43
(t, J = 22.8
112
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Hz, 10H), 1.83 (d, J = 52.0 Hz, 2H), 2.15 (t, J = 19.7 Hz, 2H), 3.38 (s, 1H),
3.58 (s, 1H), 3.79
(s, 1H), 40.1 (s, 1H), 4.12(s, 2H), 4.32 (s, 1H), 4.49 (t, J = 4.4 Hz, 1H),
4.67 - 4.70 (m, 1H),
5.97-5.98 (d, J = 5.4 Hz, 1H), 8.36-8.37 (d, J = 6.8 Hz, 1H); 13C NMR (125
MHz, D20) 6 ppm
7.67 , 8.70, 9.72, 22.92 , 27.41,34.49 , 45.49 , 47.78 , 69.61 , 73.58 , 74.79
, 86.08 , 87.43,
118.15, 139.22, 150.08, 153.06 ,153.84; 31P NMR (200 MHz, D20) 6 ppm
15.71,19.11; miz
(ES-) 580Ø
Example 18. Synthesis of compound 51
[00211]
DIEA (7.5 mmol, 969 mg, 1.5 eq.) was added dropwise to a solution of 2,6-
dichloro-9-(2',3',5'-tri-O-acetyl-f3-D-ribofuranosyl)purine (5.0 mmol, 2.23 g,
1.0 eq.) and
nortropine (5.0 mmol, 636 mg, 1.0 eq.) in 25 mL dioxane. The reaction mixture
was stirred at
room temperature overnight. The solvent was evaporated in vacuo and the
residue was
dissolved in DCM (100 mL), washed with water (2 x 30 mL). The residual
material was
purified by column chromatography, giving an intermediate compound. This
intermediate was
dissolved in 50 mL NH3/CH3OH solution, and the mixture was stirred at 35 C
overnight. After
removal of solvent (in mew)), the residual material was purified by column
chromatography,
giving a derivative of 2-chloropurine nucleoside S-51 (1.2 g).
[00212]
Compound S-51 (1.0 mmol, 411 mg, 1.0 eq.) was dissolved in trimethyl
phosphate (10 mL). The mixture was cooled in an ice-bath, followed by addition
of
bis(dichlorophosphoryOmethane (4eq.) in trimethyl phosphate (5 mL). The
resulting mixture
was stirred at 0 C for 2-4 h; and the reaction was monitored by TLC. The
reaction was
quenched by TEAC solution; and the pH of the reaction solution was adjusted to
7-8. The
mixture was extracted then with DCM; and the aqueous phase was isolated and
concentrated.
The residual material was purified by reversed-phase column chromatography
(C18-column),
giving compound 51 as a colorless solid (160 mg): 1H NMR (500 MHz, D20) 6 ppm
1.78 -
2.42 (m, 11H), 4.06 (s, 1H), 4.12 (s, 2H), 4.28 - 4.37 (m, 1H), 4.45 -4.55 (m,
1H), 4.82 - 4.88
(m, 1H), 5.38 (dd, J = 3.4, 2.4 Hz, 1H), 5.99 (d, J = 5.6 Hz, 1H),8.40 (s,
1H); 13C NMR (125
MHz, D20) 6 ppm 26.05, 27.10, 27.68, 37.08, 38.20,53.79, 54.40, 64.38, 70.20,
74.09, 83.99,
86.71, 117.78, 138.54, 151.12 ,154.22; 31P NMR (200 MHz, D20) 6 ppm
16.90,18.63; miz
(ES-) 568Ø
Example 19. Synthesis of compound a-1
113
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
0 P
0
Et0 Eto_p8
\
NCO, HO
_o_ 1
NI 1-12S0,
2,2-DirnothoxyproppFc Trifluoromethenesultonic anhydn de 1
ABulA
D-Ribose
Acetone. Me0H. rt 2 5h DCM, -1050 4h 0 o THF -78 C 1 r FtO
0><0
lh
N 1-81018' 0 N '
TNmsoCTjf DIBEA
H OH 1,4-110X.e,
izonn, inn FNNCI
OAc OP. OAc OAc
O
OAc ONE
HN
NH,-MOH '-Pro P N
818f0 ¨P TMSBr ,o N
LtdN CI DMF, 50 C, 00 Fid
OH OH
1002131
In a 1000 ml round-bottomed flask was added D-Ribose (50 g, 333.05
mmol, 1 eq.), Acetone (400 mL), 2,2-dimethoxypropane (100 mL) and HC104 (25 g,
25.00 mL,
70% purity). The mixture was stirred at room temperature for 2.5 h, followed
by addition of a
solution of Me0H (30 mL) and the mixture was stirred overnight. After the
completion of the
reaction, it was cooled to -30 'V, then a solution of 30% Na2CO3 (75 mL) was
slowly added so
that the temperature did not exceed 10 'C. The Acetone (400 mL) was removed to
afford the
crude product. The precipitate was filtered and washed with ethyl acetate (50
mL). The filtrate
was concentrated. The resulting liquid was diluted with ethyl acetate (300
mL). The organic
layer was washed with brine (300 mL), dried (Na2SO4), filtered and evaporated
to dryness. The
residue was purified by column chromatography on silica gel eluted with (PE/EA
from 100:0
to 70:30) to give [(3aR,4R,6R,6aR)-4-methoxy-2,2-dimethy1-3aA6,6a-
tetrahydrofuro[3,4-
d][1,31dioxo1-6-yllmethanol (43 g, 63.22%). 1H NMR (500 MHz, CDC13) 6ppm 1.29
(s, 3H),
1.44 (d, J = 8.3 Hz, 3H), 3.36-3.45 (m, 3H), 3.52-3.72 (m, 2H), 4.39 (t, J =
2.8 Hz, 1H), 4.56
(d, J = 5.9 Hz, 1H), 4.80 (d, J = 5.9 Hz, 1H), 4.94 (s, 1H).
[00214]
To a solution of [(3aR,4R,6R,6aR)-4-methoxy-2,2-dimethy1-3a,4,6,6a-
tetrahydrofuro[3,4-d][1,31dioxol-6-yllmethanol (20 g, 97.93 mmol, 1 eq.) in
DCM (700
mL) was added pyridine (17.82 g, 225.25 mmol, 18.13 mL, 2.3 eq.). The mixture
was cooled
to about -10 C. Then trifluoromethanesulfonic anhydride (58.03 g, 205.66
mmol, 34.60 mL,
2.1 eq.) was added dropwise into the mixture at about -10 C. The mixture
become red and a
lot of solid was formed, and kept at -10 C for about 4 h. The organic layer
was washed with
water (400 mL), followed by brine (300 mL), dried with Na2SO4, filtered, and
evaporated to
dryness. The residue was purified by column chromatography on silica gel
eluted with (PE/EA
from 100:0 to 85:15) to give [(3aR,4R,6R,6aR)-4-methoxy-2,2-dimethy1-3a,4,6,6a-
114
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
tetrahydrofuro[3,4-d] [1,3] dioxo1-6-yllmethyl trifluoromethanesulfonate (21.6
g, 65.59%) as a
brown oil.
[00215] To a solution of
Hethoxy(methyl)phosphorylloxyethane (9.77 g, 64.23
mmol, 1.0 eq.) in THF (60 mL) was cooled to about -78 C under N2 protection.
Then n-BuLi
(2.5 M, 28.26 mL, 1.1 eq.) was added dropwise into the mixture at about -78
C. The mixture
was stirred at -78 C for 25 min. The solution of [(3aR,4R,6R,6aR)-4-methoxy-
2,2-dimethyl-
3a,4,6,6a-tetrahy drofuro [3,4-d] [1,3] di oxo1-6-yll methyl
trifluoromethanesulfonate (21.6 g,
64.23 mmol, 1 eq.) in THF (20 mL) was added dropwise into the mixture at about
-78 C. The
mixture was stirred at -78 C for 1 h and then quenched by aq. NH4C1 (60 mL)
at about -78 C.
H20 (60 mL) and Et0Ac (60 mL) were added into the mixture. The organic layer
was separated
and the water layer was extracted by Et0Ac ((i0 mL). Combined the organic
layer and
concentrated and the residue was purified by column chromatography on silica
gel eluted with
(PE/EA from 100:0 to 0:100) to afford (3aR,4R,6R,6aR)-6-(2-
diethoxyphosphorylethyl)-4-
methoxy -2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3,4- d] [1,3] di oxol e (11
g, 50.62%) as a yellow
oil. 1H NMR (500 MHz, CDC13) 6 ppm: 1.28-1.34 (m, 9H). 1.47 (s, 3H), 1.72-2.00
(m, 4H),
3.35 (t, J=10.7 Hz, 3H), 4.01-4.19 (m, 5H), 4.53 (d, J=5.9 Hz, 1H), 4.60 (d,
J=5.3 Hz, 1H), 4.94
(s, 1H).
[00216] A solution of 2-
[isopropoxy(methyl)phosphorylloxypropane (1.60 g, 8.87
mmol, 2 eq) in THF (6 mL) was cooled to about -78 C under N2 protection. n-
BuLi (2.5 M,
4.43 mL, 2.5 eq.) was added dropwise into the mixture at about -78 C. The
mixture was stirred
at -78 C for 1 h. (3aR,6R,6aR)-6-(2-diethoxyphosphorylethyl)-4-methoxy-2,2-
dimethyl-
3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]dioxole (1.5 g, 4.43 mmol, 1 eq) in THF
(5 mL) was added
into the mixture at about -78 'C. The mixture was stirred at -78 C for 10
minutes and room
temperature for 2 h. The mixture was quenched by aq.NH4C1 at room temperature.
The organic
layer was separated and the water layer was extracted by Et0Ac (50 mL).
Combined the
organic layer and concentrated to dry, the residue was purified by column
chromatography on
silica gel eluted with (EA/Me0H from 100:0 to 90:10) to afford (3aR,6R,6aR)-6-
12-
[diisopropoxyphosphorylmethyl (ethoxy)pho sphoryl] ethyl] -4-
methoxy
3a,4,6,6a-tetrahydrofuro[3,4-dl [1,3[dioxole (1 g, 47.74%). 1H NMR (500 MHz,
CDC13): 6 ppm
1.25-1.40 (m, 21H), 1.80-1.82 (m, 2H), 1.94-2.05 (m, 2H), 2.28-2.32 (m, 2H),
3.27-3.29 (m,
3H), 4.05-4.09 (m, 3H), 4.51-4.54 (m, 2H), 4.68-4.70 (m, 2H), 4.86-4.89 (m,
1H).
[00217] A solution
of (2R,3S,4R,5R)-2-12-
[diisopropoxyphosphorylmethyl(ethoxy) phos ph oryll ethy11-5-methoxy -tetrahy
drofuran-3,4-
115
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
diol (1.7 g, 3.93 mmol, 1 eq.) in 1,4-dioxane (7.6 mL) and aq. sulfuric acid
(1 M, 19 mL,
4.83 eq.) was heated to reflux for 2.5 h. After the reaction was cooled to
room temperature, it
was adjusted to pH 7 by saturated NaHCO3 (38 mL) and concentrated to dryness.
The residue
was evaporated with dry THF twice to afford the crude product, which was used
in the next
step without purification.
[00218] To the above solution
of (3R,4S,5R)-5-[2-
[dii s oprop oxy pho sphory 'methyl (ethoxy) phosphoryl] ethyl] tetrahy
drofuran-2,3,4-triol in dry
pyridine (25 mL) was added dropwise acetic anhydride (5.0 mL) at 0 'C. After
the reaction
mixture was stirred at room temperature for 16 h, it was evaporated to
dryness. The residue
was adjusted to pH 2 by aq. HC1 (1 M, 15 mL). It was treated with DCM (50 mL)
and the
organic layer was separated, washed with brine (10 mL), dried, and
concentrated. The residue
was purified by column chromatography on silica gel eluted with (DCM/Me0H from
100:0 to
90:10) to
give [(2R,3R,4R)-4,5-diacetoxy-242-[diisopropoxyphosphoryl
methyl(ethoxy)phosphoryllethylltetrahydrofuran-3-yl] acetate (1.5 g, 70.28%)
as a yellow oil.
1H NMR (500 MHz, Me0D) 6ppm 1.34 (dt, J=12.6, 6.4, 15H), 1.93-2.17 (m, 13H),
2.51-2.83
(m, 2H), 4.13 (dd, J=16.8, 7.2, 2H), 4.23 (d, J=15.6, 1H), 4.66-4.80 (m, 2H),
5.06-5.21 (m,
1H), 5.30 (t, J=15.2, 1H), 5.95-6.45 (m, 1H).
[00219] To a solution of
[(2R,3R,4R)-4,5 -di acetoxy -2-[2-
[diisopropoxyphosphorylmethyl (ethoxy)phosphoryllethylltetrahydrofuran-3-yll
acetate (2.5
g, 4.59 mmol, 1 eq.) and 2,6-dichloro-9H-purine (911.2 mg, 4.64 mmol, 1.05
eq.) in ACN (25
mL) was added DBU (768.7 mg, 5.05 mmol, 572.93 !IL, 1.1 eq.) and TMSOTf (2.25
g, 10.10
mmol, 1.83 mL, 2.2 eq.) at -10 C. The mixture was stirred at 50 C for lh.
The reaction
completion was detected by TLC (EA/Me0H=20:1). The mixture was concentrated
and purified by column chromatography on silica gel eluted with (EA/Me0H from
100:0 to
95:5) to
give [(2R,3R,4R,5R)-4-acetoxy -5 -(2,6-di chl oropurin-9-y1)-242-
[diisopropoxyphosphorylmethyl(ethoxy)phosphoryllethyll tetrahydrofuran-3-yll
acetate (1.5
g, 63.83%). 1H NMR (500 MHz, CDC13): 6 ppm 1.21-1.31 (m, 15H), 1.65-2.06 (m,
10H),
2.35-2.46 (m, 2H), 4.07-4.14 (m, 2H), 4.72-5.16 (m, 3H), 5.28-5.77 (m, 1H),
6.08-6.11 (m,
1H), 8.40-9.20 (m, 1H).
1002201
To a solution of [(2R,3R,4R,5R)-4-acetav-5-(2,6-dichloropurin-9-y1)-2-
[2-[diisopropoxy phosphorylmethyl(ethoxy)phosphoryllethylltetrahydrofuran-3-
yll acetate
(250 mg, 371.24 umol, 1 eq.) and phenylmethanamine (47.74 mg, 445.49 umol, 1.2
eq.) in 1,4-
dioxane (2.5 mL) was added DIPEA (143.94 mg, 1.11 mmol, 193.99 gL, 3 eq.). The
mixture
116
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
was stirred at 120 C for 16 h. The reaction was complete detected by TLC
(Et0Ac:
Me0H=10:1). The mixture was concentrated and purified by column chromatograph
on silica
gel eluted with (EA/Me0H from 100:0 to 90:10) to give [(2R,3R,4R,5R)-4-acetoxy-
546-
(benzylamino)-2-chloro-purin-9-y1]-2-[2-
[dii s oprop oxy pho sphory lmethyl (ethoxy)pho s phoryl] ethyl]
tetrahydrofuran-3-yll acetate (140
mg, 50.68%) as yellow semi-solid.
[00221]
A solution of [(2R,3R,4R,5R)-4-acetoxy-546-(benzylamino)-2-chloro-
purin-9-yll -2- [24 dii s oprop oxypho sphory lmethyl(ethoxy)pho sphoryl]
ethyl] tetrahy drofuran-3-
yl] acetate (140 mg, 188.14 nmol, 1 eq.) in NH3-Me0H (7M, 1.5 mL, 57.56 eq.)
was stirred at
room temperature for 2 h. The mixture was concentrated to remove the solvent
to afford the
crude product (2R,3R,4S,5R)-246-(benzylamino)-2-chloro-purin-9-
y11-5-[2-[diisopropoxy
phosphorylmethyl(ethoxy)phosphoryllethylltetrahydrofuran-3,4-diol as yellow
semi-solid. It
was used directly for the next step.
[00222]
To a solution of (2R,3R,4S,5R)-246-(benzylamino)-2-chloro-purin-9-y11-
5- [2-[dii s opro poxy pho sphorylmethyl(ethoxy)phos phoryllethylltetrahy
drofuran-3,4-di ol
(124.18 mg, 188.14 nmol, 1 eq.) in DMF (4 mL) was added bromo(trimethyl)silane
(432.05
nig, 2.82 nunol, 15 eq.). The mixture was stirred at 50 'V for 2 h. Then the
mixture was
quenched by TEAC solution until pH=8 at 0 C. The mixture was concentrated to
dry and
added H20 (5 mL) and purified by reversed-phase chromatography (C18-column)
(H20/ACN
from 100:0 to 90:10) to get a-1 (20 mg, 19.40%) as white solid. 1H NMR (500
MHz, CD30D)
6 ppm: 1.88-2.13 (m, 6H), 4.04-4.07 (m, 1H), 4.21-4.23 (m, 1H), 4.61-4.63 (m,
1H), 4.77 (s,
2H), 5.94 (d, J=2.5 Hz, 1H), 7.27-7.29 (m, 1H), 7.33-7.36 (m, 2H), 7.41-7.42
(m, 2H), 8.30 (s,
1H); 13C NMR (125 MHz, CD30D) 6 ppm: 27.85, 28.11, 28..89, 31.15, 31.75,
32.07, 32.66,
45.09, 74.36, 75.62, 86.30, 86.43, 69.41, 119.67, 128.33, 128.84, 129.56,
139.98, 141.01,
151.25, 155.72, 156.40; 31P NMR (203 MHz, CD30D) 6 ppm:14.99, 36.71; m/z
(ESL): 548.0
(M + H).
Example 20. Synthesis of compound a-9
117
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
H
Pr0-1,HN 0 N'lrL'N
I-Pr0 -PrC3-43 ea-)L.1 1-PrO-Fr 0
H
I-Prd
NH0
Etd N CI ci_ Pr9 EtdPH4 lc 'CI meol_ 0HNN
0
080 OAc '
4-dioxanc, 13000, 160Oh
0A0, OAc OH OH
OH
TMSBr ,0
DMF, 50 C 2h HO
Hdir:µ
OH CH
[00223]
To a solution of [(2R,3R,4R,5R)-4-acetoxy-5-(2,6-dichloropurin-9-y1)-2-
[2-[diisopropoxyphosphorylmethyl(ethoxy)phosphoryllethylltetrahydrofuran-3-yll
acetate
(660 mg, 980.08 mmol, 1 eq.) and 1(3R,5S)-3,5-dimethy1-1-
adamantyllammonium;chloride
(317.20 mg, 1.47 mmol, 1.5 eq.) in 1,4-dioxane (11 mL) was added DIPEA (380.00
mg, 2.94
mmol, 512.12 pL, 3 eq.). The mixture was stirred at 130 C for 16 h. The
reaction completion
was detected by TLC (Et0Ac: Me0H=10:1). The mixture was concentrated and
purified by
column chromatography on silica gel eluted with (EA/Me0H from 100:0 to 92:8)
to give
[(2R,3R,4R,5R)-4-acetoxy-5- [2-chl oro-6- [[(3R,5 S)-3,5 -dimethy1-1-
adamantyll amino] purin-
9-yl] -2-1-2-[di is oprop oxy pho sphorylmethyl(ethoxy)phosphoryl]
ethylltetrahydrofuran-3-yll
acetate (310 mg, 38.75%) as yellow semi-solid. 1H NMR (500 MHz, CD30D): 6 ppm:
0.90(s,
6H), 1.13-1.50 (m, 20H), 1.79-2.26(m, 14H), 2.64 (dd, J = 31.9, 15.0 Hz, 2H),
4.10 (qd, J=14.4,
7.1 Hz, 2H), 4.64-4.79 (m, 2H), 5.50-5.61 (m, 1H), 5.83-5.94 (m, 1H), 6.09 (d,
J=4.9 Hz, 1H),
6.99 (s, 1H), 8.18 (d, J=6.0 Hz, 1H).
[00224]
A solution of [(2R,3R,4R,5R)-4-acetoxy-5-[2-chloro-6-[[(3R,5S)-3,5-
di methyl -1-adamantyll ami no] purl n-9-yll -242-
[dii s prop oxy pho sphory 'methyl (ethoxy)pho s phoryl] ethyl]
tetrahydrofuran-3-yl] acetate (310
mg, 379.78 wnol. 1 eq.) in NI-13-Me0H (7M, 6 mL, 110.59 eq.) was stirred at
room
temperature for 3 h. The mixture was concentrated to remove the solvent. Et0Ac
(35 mL) was
added into the residue and washed by brine. The organic layer was dried with
Na2SO4 and
concentrated to afford the crude product (2R,3R,4S,5R)-2-12-chloro-6-[[(3R,5S)-
3,5-
dimethy1-1-adamantyll amino] p urin-9-yll -5 - [2-
[dii s oprop oxy pho sphory lmethyl (ethoxy)pho s phoryl] ethylltetrahy
drofuran-3,4-di ol (250 mg,
89.91%) as yellow semi-solid. The solid was used directly for the next step.
[00225]
To a solution of (2R,3R,4S,5R)-242-chloro-6-[[(3R,5S)-3,5-dimethy1-1-
adamantyl] amino] purin-9-yll -5 -[2-[dii s opropoxy pho sphory
lmethyl(ethoxy)pho sphoryl] ethyl]
tetrahydrofuran-3,4-diol (250 mg, 341.44 wnol, 1 eq.) in DMF (8 mL) was
118
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
added bromo(trimethyl)silane (993.18 mg, 6.49 mmol, 856.19 [IL, 19 eq.). The
mixture was
stirred at 50 C for 2 h. Then the mixture was quenched by TEAC solution until
pH=8 at 0 C.
The mixture was concentrated to dry and H20 (8 mL) was added, then compound
was purified
by reverse-phase chromatography (C18-column) (H20/ACN from 100:0 to 80:20) to
get a-9
(101 mg, 41.02%) as white solid. 1H NMR (500 MHz, CD30D) 6 ppm: 0.89 (s, 6H),
1.13-1.23
(m, 2H), 1.35 (d, J=12.3 Hz, 2H), 1.45 (d, J=12.0 Hz, 2H), 1.81 (d, J=11.8 Hz,
2H), 1.85-2.01
(m, 5H), 2.03-2.15 (m, 5H), 2.18 (d, J=2.9 Hz, 1H), 4.02 (dd, J=12.2, 5.3 Hz,
1H), 4.17 (t,
J=5.1 Hz, 1H), 4.57 (t, J=5.1 Hz, 1H), 5.89 (d, J=4.8 Hz, 1H), 8.26 (s, 1H);
13C NMR (125
MHz, CD30D) 6 ppm: 27.68, 27.94, 28.72, 30.75, 31.69, 33.44, 40.72, 43.75,
51.75, 55.91,
74.37, 75.65, 86.15, 86.29, 89.47, 119.70, 140.55, 150.78, 154.97, 155.88; 31P
NMR (203 MHz,
CD30D) 6 ppm:14.98, 38.21; 111/7 (ES-11): 620.1 (M + H).
Example 21. Synthesis of compound a-19
rf
CI HN HNf
P PrO 0 ,L
1-121,1 DIPEA d <1µhl '71,1
NH,Me0H TMS13r
otd N CI
1,4-dioxarle, 120 C, 16h Et0 \-41--''N CI 0, 2h PrO
Et0 \-141NCI DMF, 50 C, 2h
OAc OAc OA c OA c OH OH
HN:ri
HO N CI
OH OH
[00226]
To a solution of [(2R,3R,4R,5R)-4-acetoxy-5-(2,6-dichloropurin-9-y1)-2-
[2-Idiisopropoxyphosphorylmethyl(ethoxy)phosphoryllethylltetrahydrofuran-3-yll
acetate
(250 mg, 371.24 1.imol, 1 eq.) and dodecan-1-amine (82.57 mg, 445.49 1.imol,
1.2 eq.) in 1,4-
dioxane (2.5 mL) was added DIPEA (143.94 mg, 1.11 mmol, 193.99 [IL, 3 eq.).
The mixture
was stirred at 120 C. for 16 h. The reaction completion was detected by TLC
(Et0Ac:
Me0H=10:1). The mixture was concentrated and purified by column chromatography
on silica
gel eluted with (EA/Me0H from 100:0 to 90:10) to give [(2R,3R,4R,5R)-4-acetoxy-
542-
chloro-6-(dodecylamino)purin-9-y1]-2-12-
119
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[ dii s oprop oxy pho sphory lmethyl (ethoxy)pho s phoryl] ethyl]
tetrahydrofuran-3-yll acetate (150
mg, 49.14%) as yellow semi-solid.
[00227] To a solution
of [(2R,3R,4R,5R)-4-acetoxy -5- [2-chloro-6-
(dodecylamino)purin-9-y11-242-
[dii s prop oxy pho sphory lmethyl (ethoxy)pho s phoryl] ethylltetrahy
drofuran-3 -yl] acetate (150
mg, 182.41 itimol, 1 eq.) in NH3-Me0H (7M, 1.5 mL, 57.56 eq.) was stirred at
room
temperature for 2 h. The mixture was concentrated to remove the solvent to
afford the crude
product (2R,3R,4S,5R)-242-chloro-6-(dodecylamino)purin-9-y11-5-[24diisopropoxy
phosphorylmethyl(ethoxy)phosphoryllethylltetrahydrofuran-3,4-diol as yellow
semi-solid. It
was used directly for the next step.
1002281 To a solution of (2R,3R,4S,5R)-2-12-chloro-6-
(dodecylamino)purin-9-y11-
5- [2-[dii s opro poxy pho sphorylmethyl(ethoxy)phos phoryl] ethylltetrahy
drofuran-3,4-di ol
(134.66 mg, 182.41 1.imol, 1 eq.) in DMF (4 mL) was added
bromo(trimethyl)silane (418.90
mg, 2.74 mmol, 15 eq.). The mixture was stirred at 50 C for 2 h. Then the
mixture was
quenched by TEAC solution until pH 8 at 0 C. The mixture was concentrated to
dry and H20
(5 mL) was added, then compound was purified by reverse-phase chromatography
(C18-
column) (H20/ACN from 100:0 to 70:30) to get a-19 (20 mg, 17.51%) as white
solid. 1H NMR
(500 MHz, CD30D) 6 ppm: 0.86-0.089 (m, 3H), 1.26-1.30 (m, 18H), 1.64-L67 (m,
2H), 1.92-
2.17 (m, 6H), 1.81 (d, J=11.8 Hz, 2H), 1.85-2.01 (m, 5H), 2.03-2.15 (m, 5H),
3.51-3.54 (m,
2H), 4.05-4.06 (d, J=5.0, 1H), 4.19 (t, J=5.0 Hz, 1H), 4.59 (t, J=5.0 Hz, 1H),
5.91 (d, J=5.0 Hz,
1H), 8.25 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm: 13.13, 22.33, 26.17, 26.45,
28.87,
28.97, 29.04, 29.26, 29.32, 31.64, 40.20, 72.95, 74.25, 84.86, 87.88, 118.09,
139.24, 149.41,
154.44, 155.17; 31P NMR (203 MHz, CD30D) 6 ppm:15.01, 39.35; m/z (ESr): 626.2
(M + H).
Example 22. Synthesis of compound a-31
<N)
DO red'a 17p7OP\¨F,' rsi.i-isr'N NH
/ Me0H .. Pr'j .. AN i
HN--/ DIPEA Bui rel'a PrO
rt, Et0
0
1,4-clioxane, 130 C, 16h 16h -0-
OAc OAc
OAcOAe
OH OH
cD
0
TMSBr HO
<JLN
DMF, 50 C, 2h HO
OH OH
120
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00229]
To a solution of (2R,3R,4R,5R)-4-acetoxy-5-(2,6-dichloropurin-9-y1)-2-
[24diisopropoxyphosphorylmethyl(ethoxy)phosphory1l ethylltetrahydrofuran-3-yll
acetate
(1.5 g, 2.23 mmol, 1 eq) and 2-azaspir0[4.5]decane (465.22 mg, 3.34 mmol, 1.5
eq.) in 1,4-
dioxane (15 mL) was added DIPEA (1.01 g, 7.80 mmol, 1.36 mL, 3.0 eq.) . The
mixture was
stirred at 130 C for 16 h. The reaction completion was detected by TLC
(EA/Me0H=20:1).
The mixture was concentrated and purified by column chromatography on silica
gel eluted with
(EA/Me0H from 100:0 to 95:5) to give R2R,3R,4R,5R)-4-acetoxy-546-(2-
azaspiro [4. 5] decan-2-y1)-2-chloro-purin-9-y11-2424
diisopropoxyphosphorylmethyl(ethoxy)
phosphoryl] ethyl] tetrahydrofuran-3-yll acetate (950 mg, 54.95%). 1H NMR (500
MHz,
CD30D): 6 ppm 1.26-1.36 (m, 17H), 1.51-1.53 (m, 2H), 1.83-2.17 (m, 12H), 2.62-
2.69 (m,
2H), 3.39-3.53 (m, 1H), 3.71-3.73 (m, 1H), 3.93-3.95 (m, 1H), 4.13-4.27 (m,
4H), 4.69-4.71
(m, 2H), 5.58-5.59 (m, 1H), 5.91-5.93 (m, 1H), 6.10-6.12 (m, 1H), 6.15-6.17
(m, 1H).
[00230]
A solution of [(2R,3R,4R,5R)-4-acetoxy -5-[6-(2-azaspiro[4.51decan-2-
y1)-2-thloro-purin-9-yll -2-12-
[dii s prop oxy pho sphory 'methyl (ethoxy)pho s phoryl] ethyl]tetrahy dro
furan-3-yl] acetate (950
mg, 1.22 mmol, 1 eq.) in NH3-Me0H (7 M, 10 mL, 57.19 eq.) was prepared. The
mixture was
stirred at room temperature for 16 h. The mixture was concentrated and Et0Ac
(30 mL) was
added. The organic layer was washed with NaCl aqueous, dried (Na2SO4),
filtered, and
evaporated to dryness to give (2R,3R,4S,5R)-246-(2-azaspiro[4.5]decan-2-y1)-2-
chloro-purin-
9-yll -5424di is oprop oxy pho sphorylmethyl (ethoxy)pho sphoryl] ethyl]
tetrahy drofuran-3 ,4-di ol
(650 mg, 76.73%). 1H NMR (500 MHz, CD30D): 6 ppm 1.27-1.36 (m, 15H), 1.52-1.54
(m,
10H), 1.84-1.94 (m, 2H), 2.05-2.18 (m, 4H), 2.61-2.68 (m, 2H), 3.52-3.54 (m,
1H), 3.72-3.74
(m, 1H), 3.88-4.03 (m, 2H), 4.12-4.14 (m, 3H), 4.27-4.29 (m, 1H), 4.62-4.73
(m, 3H), 5.89 (s,
1H), 8.14-8.15 (m, 1H).
1002311 To a solution of (2R,3R,4S,5R)-2-1-6-(2-
azaspiro[4.51decan-2-y1)-2-
chloro-purin-9-y1]-5-12-
[diisopropoxyphosphorylmethyl(ethoxy)phosphoryl]ethylltetrahydrofuran-3,4-diol
(100 mg,
144.48 wnol, 1 eq.) in DMF (4 mL) was added bromo(trimethyl)silane (331.79 mg,
2.17 mmol,
15 eq.) at 0 C. The mixture was stirred at 50 C for 2h. Then the mixture
was quenched by
TEAC solution until pH 8 at 0 C. The mixture was concentrated to dry and H20
(10 mL) was
added, and purified by reverse-phase chromatography (C18-column) (H20/ACN from
100:0
to 90: 10) to get desired product a-31 (20 mg, 20.32%). 1H NMR (CD30D, 500
MHz): 6 ppm
121
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1.51-1.57 (m, 10H), 1.83-2.10 (m, 8H), 3.51-3.53 (m, 1H), 3.71-3.73 (m, 1H),
3.95-3.97 (m,
1H), 4.03-4.04 (m, 1H), 4.16-4.19 (m, 2H), 4.57-4.59 (m, 1H), 5.90-5.91 (d,
1H), 8.19-8.20
(m, 1H); 13C NMR (CD30D, 125 MHz): 6 ppm 23.02, 25.87, 26.18, 26.40, 27.18,
29.21,
29.80, 30.13, 30.73, 34.89, 40.37, 42.67, 72.94, 74.05, 84.70, 87.93, 118.71,
138.65, 150.96,
153.21, 153.83; 31P NMR (CD30D, 203 MHz): 6 15.12, 38.73; miz (ESI+):580.1
(M+H).
Example 23. Synthesis of compound c-13
CI
2 0
EtO-P Et04
0 ,N
Et0
H2s0h4 Etd
0- OH ______________ 0- ,,OAc Trisim' s
'oe_rfi
Pyri7e2 , 16h Etd \¨LCH-rf-
ctr4
OH OH OAc OAc DBU, Acetonitrile, 50 C, 1h
OAc OAc
AIL\
H.e
HN".
'KJ) N;67
7, õH
0 N¨
1:4
DIEA
TFM5S0Bcr 2h HEolc_5Fc_vroc_ j,re,Lei
9 11 ! Me0 11 Et0
1,4-dioxane, 130 C, 16hEtO\NCI 71._r
rt, 16h Et0 N CI om
Eld
OH OH OH OH
OAc OAc
1002321 To a mixture of (3aR,4R,6R,6aR)-6-(2-
diethoxyphosphorylethyl)-4-
methoxy-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3]dioxole (2.50 g, 7.39
mmol, 1
eq.) in 1,4-dioxane (10 mL) was added 1 N H2SO4 (1 M, 25 mL, 3.38 eq), and it
was heated to
reflux for 2.5 h. Then the pH was adjusted to 7 by aq. NaHCO3 (50 mL), and it
was
concentrated to dryness. To the residue in Pyridine (21 mL) was added Ac20
(836.84 mg, 8.20
mmol, 4.2 mL), and it was stirred at room temperature for 16 h. The resulting
solution
was evaporated to dryness. The residue was purified by column chromatography
on silica gel
eluted with (DCM/Me0H from 100:0 to 95:05) to give 1(2R,3R,4R)-4,5-diacetoxv-2-
(2-
diethoxyphosphorylethyl) tetrahydrofuran-3-yll acetate (1.9 g, 62.67%) . 1H
NMR (500 MHz,
CDC13): 6 ppm 1.36-1.54 (m, 6H), 1.81-2.30 (m, 13H), 1.94-2.05 (m, 2H), 4.13-
4.31 (m, 5H),
4.90-5.23 (m, 2H), 6.16-6.41 (m, 1H).
[00233] To a solution of
[(2R,3R,4R)-4,5-diacetoxy-2-(2-
diethoxyphosphorylethyl) tetrahydrofuran-3-yll acetate (1.9 g, 4.63 mmol, 1
eq) in ACN (19
mL) was added DBU (775.39 mg, 5.09 mmol, 760.18 pL, 1.1 eq) and TMSOTf (2.26
g, 10.19
mmol, 1.84 mL, 2.2 eq) at -10 C. The mixture was stirred at 50 C for 1 h.
The mixture was
concentrated and purified by column chromatography on silica gel eluted with
(EA/Me0H
from 100:0 to 95:5) to give [(2R,3R,4R,5R)-4-acetoxy-5-(2,6-dichloropurin-9-
y1)-2-(2-
diethoxyphosphoryl ethyl) tetrahydrofuran-3-y11 acetate (1.3 g, 52.06%). 1H
NMR (500 MHz,
122
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
CDC13): 6 ppm 1.24-1.34 (m, 6H), 2.01-2.14 (m, 10H), 4.09-4.12 (m, 5H), 5.56-
5.58 (m, 1H),
5.88-5.90 (m, 1H), 6.24-6.25 (d, 1H), 8.68 (s, 1H).
1002341
To a solution of [(2R,3R,4R,5R)-4-acetoxy-5-(2,6-dichloropurin-9-y1)-2-
(2-diethoxyphosphorylethyptetrahydrofuran-3-yll acetate (1.3 g, 2.41 mmol, 1
eq.) and
[(3R,5S)-3,5-dimethy1-1-adamantyl]ammonium;chloride (624.12 mg, 2.89 mmol, 1.2
eq.) in 1,4-dioxane (13 mL) was added DIPEA (934.61 mg, 7.23 mmol, 1.26 mL, 3
eq.). The
mixture was stirred at 130 C for 16 h. The reaction completion was detected
by TLC. The
mixture was concentrated and purified by column chromatography on silica gel
eluted with
(EA/Me0H from 100:0 to 90:10) to give [(2R,3R,4R,5R)-4-acetoxy-5-[2-chloro-6-
[[(3R,5S)-
3,5-dimethy1-1-adamantyll amino]purin-9-y11-2-(2-
diethoxyphosphorylethyl)tetrahydrofuran-
3-yll acetate (700 nig, 42.57%). 1H NMR (5001VI1-Iz, CD30D): 6 ppm 0.84-0.91
(m, 6H), 1.20-
1.32 (m, 9H), 1.36-1.52 (m, 4H), 1.82-2.01 (m, 6H), 2.07-2.14 (m, 10H), 4.08-
4.11 (m, 5H),
5.57-5.60 (m, 1H), 5.87-5.89 (m, 1H), 6.09-6.10 (m, 1H), 8.16 (m, 1H).
[00235]
To a solution of [(2R,3R,4R,5R)-4-acetoxy-542-chloro-6-[[(3R,5S)-3,5-
dimethy1-1-adamantyll amino] purin-9-yll -2-(2-
diethoxyphosphorylethyl)tetrahydrofuran-3-yll
acetate (700 mg, 1.03 mmol, 1 eq) was added NH3-Me0H (7 M, 7 mL, 47.75 eq.).
The mixture
was stirred at room temperature for 16 h. The mixture was concentrated and
Et0Ac (30 mL)
was added, and the organic layer was washed with aq. NaCl, dried (Na2SO4),
filtered, and
evaporated to
give (2R,3R,4S,5R)-2-[2-chloro-64[(3R,5S)-3,5-dimethy1-1-
adamantyl]amino]purin-9-y1]-5-(2-diethoxyphosphorylethyl) tetrahydrofuran-3,4-
diol (400
mg, 65.18%). 1H NMR (500 MHz, CD30D): 6 ppm 0.90-0.92 (m, 6H), 1.20-1.26 (m,
2H),
1.29-1.32 (m, 6H), 1.36-1.39 (m, 2H), 1.46-1.48 (m, 2H), 1.82-1.84 (m, 2H),
1.91-1.93 (m,
3H), 1.96-2.05 (m, 3H), 2.10-2.12 (m, 2H), 2.20-2.21 (m, 1H), 4.03-4.11 (m,
5H), 4.27-4.29
(m, 1H), 4.69-4.71 (m, 1H), 5.86-5.87 (m, 1H), 8.15 (s, 1H).
[00236]
To a solution of (2R,3R,4S,5R)-242-chloro-6-[[(3R,5S)-3,5-dimethy1-1-
adamantyl] amino] purin-9-y11-5-(2-diethoxyphosphorylethyptetrahydrofuran-3,4-
diol (100
mg, 167.20 1.trriol, 1 eq) in DMF (4 mL) was added bromo(trimethyOsilane
(383.97 mg, 2.51
mmol, 331.01 pt, 15 eq) at 0 C. The mixture was stirred at 50 C for 2 h.
Then the mixture
was quenched by TEAC solution until pH 8 at 0 C. The mixture was concentrated
and H20
(10mL) was added, then compound was purified by reverse-phase chromatography
(C18-
column) (H20/ACN from 100:0 to 70: 30) to get c-13 (40 mg, 37.20%). 1H NMR
(CD30D,
500 MHz): 6 ppm 1.19-1.26 (m, 2H), 1.36-1.38 (m, 2H), 1.45-1.48 (m, 2H),1.63-
1.74 (m, 2H),
1.82-1.84 (m, 2H), 1.90-1.95 (m, 2H), 1.98-2.10 (m,4H), 2.19-2.21 (m, 1H),
4.02-4.03 (m, 1H),
123
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
4.15-4.17 (m, 1H), 4.55-4.56(m, 1H), 5.90-5.91 (d, 1H), 8.22 (s, 1H) .13C NMR
(CD30D, 125
MHz): 6 ppm 25.41, 26.49, 29.21, 30.75, 31.70, 33.45, 40.71, 43.76, 51.76,
55.91, 74.60, 75.76,
85.99, 86.13, 89.71, 119.74, 140.42, 150.68, 154.98, 155.88; 31P NMR (CD30D,
203
MHz): oppm 24.12; m/z (ES1):540.2 (M-H).
Example 24. Synthesis of compound d-1
CI),r0 Mõe,C)H HCH C;i2r __ ' 'HO/ Aro
,_,
/ K2003,DMS0 HO HO EDCI HO MeCN I-120 ¨0 HORt T ir
-0
DIPEA, ONIF
Aco
< >
I I
0, 0 OAc OAc
_______________________________________ Ac0 N N¨ci NH3_Me01-
1,.. N¨ ci N N"Cl
H 700 0
OAc OAc OH CH
p-Ts0H CX
Ff)
07
0 0
\CI HC(OH OHC'43 N CI
P0(0Me)3
I-120 OH OH
1002371 A mixture of indane-1-carboxylic acid (5 g, 30.83 mmol,
1 eq.) and H2SO4 (3.02
g, 30.83 mmol, 1.64 mL, 1 eq.) in Me0H (100 mL) was heated at 65 C for 20 h.
The reaction
was complete as indicated by TLC analysis. The resulting solution was
evaporated and the
residue was diluted with Et0Ac (30 mL). Then it was washed with brine, dried
(Na2SO4),
filtered, and evaporated to dryness, giving methyl indane-1-carboxylate (4.7
g, 86.5%).
1002381 To a solution of methyl indane-l-carboxylate (4.7 g,
26.67 mmol,
1 eq.) in DMSO (47 mL) were added K2CO3 (11.06 g, 80.02 mmol, 3 eq.) and HCHO
(7.37 g,
80.02 mmol, 37% purity, 3 eq.) at 0 C under nitrogen atmosphere. The mixture
was stirred at
room temperature for 19 h. The resulting solution was quenched by H20 (50 mL).
The mixture
was extracted with Et0Ac (50 mL). Then the pH of the aqueous was adjusted to 3
with 2.5 N
HC1, extracted with Et0Ac (3 x 50 mL). The organic layer was washed with
brine, dried
(Na2SO4), filtered, and evaporated to dryness, giving 1-(hydroxymethypindane-1-
carboxylic
acid (3.3 g, 64.4%).
1002391 To a mixture of 1-(hydroxymethyl)indane- 1-carboxylic
acid (2 g, 10.41 mmol,
1 eq.) and (4-methoxyphenyl)methanamine (1.43 g, 10.41 mmol, leq.) in DMF (20
mL) were
added EDCI (2.99 g, 15.61 mmol, 1.5 eq.), HOBT (2.11 g, 15.61 mmol, 1.5 eq.)
and DIPEA
(2.02 g, 15.61 mmol, 2.72 mL, 1.5 eq.). The mixture was stirred at room
temperature for 18 h.
The reaction was complete as indicated by TLC. The resulting solution was
quenched by
124
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
water (10 mL), and diluted with Ft0Ac (80 mL). The organic layer was washed
with brine,
dried (Na2SO4), filtered, and evaporated to dryness. The residue was purified
by column
chromatograph on silica gel eluted with DCM/Me0H (from 100:0 to 95:5),
providing 1-
(hy dro xy methyl)-N-[(4-methoxy phenyl) methyl] indane-l-carb oxami de as an
oil, (2.4 g,
74.1%).
[00240] To a solution of 1-(hydroxymethyl)-N-[(4-
me1hoxyphenyl)me1hyllindane-1-
carboxamide (2.2 g, 7.07 mmol, 1 eq.) in THF (55 mL) under nitrogen atmosphere
were
added PPh3 (2.78 g, 10.60 mmol, 1.5 eq.), and DEAD (1.85 g, 10.60 mmol, 1.67
mL,
1.5 eq.) dropwise subsequently, while the temperature was maintained at 0 C.
The mixture
was stirred at room temperature for 2 h, and TLC analysis indicated completion
of the reaction.
The resulting solution was quenched by water (2 mL), followed by addition of
Et0Ac (20
mL). The organic layer was separated, washed with brine, dried (Na2SO4),
filtered, and
evaporated to dryness. The residue was purified by column chromatograph on
silica gel eluted
with DCM/Me0H (from 100:0 to 98:2-98:3), giving 1-[(4-methoxyphenyl)methyll-
spiro-
[azetidine-3,1'-indane1-2-one (1.7 g, 82.1 %) as a yellow solid.
[00241] A solution of 1-[(4-methoxyphenyl)methyl]spiro
[azetidine-3,1'-indane J -2-one
(1.4 g, 4.77 mmol, 1 eq.) in acetonitrile (36 mL) and H20 (4 mL) was added
Ceric ammonium
nitrate (9.16 g, 16.70 mmol, 3.5 eq.) at 0 C. The mixture was stirred at room
temperature for
16 h. The reaction was complete as indicated by TLC. The resulting solution
was diluted
with Et0Ac (50 mL). The organic layer was separated, washed with water, brine,
dried
(Na2SO4), filtered, and evaporated to dryness. The residue was purified by
column
chromatograph on silica gel eluted with PE/EA (from 100:0 to 45:55) to give
spiro[azetidine-
3,1'-indaneJ-2-one (160 mg, 19.4%) as a yellow solid.
[00242] To a solution of spiro[azetidine-3,1'-indane]-2-one
(180.00 mg, 1.04 mmol,
1 eq.) in THF (10 mL) was added LiA1H4 (39.44 mg, 1.04 mmol, 1 eq.) at 0 C.
The mixture
was stirred at 70 C overnight, quenched by H20 (1 mL), followed by addition
of Et0Ac (10
mL). The solid material was removed by filtration and the organic layer was
concentrated. The
residue was purified by column chromatography on silica gel eluted with
DCM/Me0H (100:0-
80:20) and afford the product spiro[azetidine-3,1'-indane] (80 mg, 48.4%) as
an oil.
1002431 To
a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (224.70 mg, 502.43
1 eq.) in 1,4-dioxane (8
mL) were added spiro[azetidine-3,1'-indane] (80 rug, 502.43 limo], 1 eq.) and
DIPEA (162.33
mg, 1.26 mmol, 218.78 !IL, 2.5 eq.). The mixture was stirred at 100 'V
overnight. TLC showed
125
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
that starting material was consumed and two new products were formed. The
mixture was
concentrated and the residue was purified by column chromatography on silica
gel (eluent,
PE/Et0Ac, from 100:0 to 60:40), affording [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-
chloro-6-
spiro [azetidine-3,1'-indanel -1-yl-p urin-9-y Otetrahy drofuran-2-yllmethyl
acetate (100 mg,
34.9%).
[00244]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy -5-(2-chloro-6-
spiro[azetidine-
3,1'-indane]-1-yl-purin-9-yl)tetrahy drofuran-2-yll methyl acetate (120 mg,
210.53 pmol,
1 eq.) in Me0H (2 mL) was added NH3-Me0H (7 M, 601.51 111,õ 20 eq.). The
mixture was
stirred at room temperature for 4 h. The solvent was removed by evaporation;
and the residue
was diluted with Et0Ac (30 mL), washed by brine (30 mL), dried with Na2SO4.
The organic
layer was concentrated to afford the crude product (2R,3R,4S,5R)-2-(2-chloro-6-
spiro [azetidine-3,1'-indanel -1-yl-purin-9-y1)-5-
(hydroxymethyptetrahydrofuran-3,4-diol (90
mg, 202.76 pmol, 96.31% yield) as light yellow solid, used without further
purification.
[00245]
To a solution of (2R,3R,4S,5R)-2-(2-ch1oro-6-spiro[azetidine-3,1'-indane1-
1-yl-
purin-9-y1)-5-(hydroxymethyptetrahydrofuran-3,4-diol (80 mg, 180.23 pmol, 1
eq.) in acetone
(20 mL) was added 2,2-dimethoxypropane (187.70 mg, 1.80 mmol, 10 eq.) and Ts0H-
H20
(35.69 mg, 180.23 pmol, 1.0 eq.). The mixture was stirred at room temperature
overnight. The
mixture was diluted with Et0Ac (70 mL), washed by aqueous NaHCO3 (40 mL), and
brine (40
mL). The organic layer was concentrated and the residue was purified by column
chromatography on silica gel eluted with PE/Et0Ac (100:0-45:55) to
afford [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro [azetidine-3,1'-indanel -1 -yl-
purin-9-y1)-2,2-
dimethy1-3a,4,6,6a-tetrahy drofuro[3,4-d] [1,31dioxo1-6-yll methanol (70 mg,
80.2%).
[00246]
A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[azetidine-3,1'-indane_1-
1-
yl-purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3] dioxo1-6-
yllmethanol (70 mg,
144.64 pmol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to about 0 C, followed by
addition a
solution of bis(dichlorophosphoryl)methane (72.26 mg, 289.29 pmol, 2.0 eq.) in
PO(Me0)3 (1
mL). The mixture was stirred at about 0 C for 6 h. H20 (3 mL) was added, and
mixture was
stirred further at room temperature overnight. Purification of the reaction
mixture (C18
reversed phase silica gel, 0-30% ACN in Water) afforded Compound d-1, (41 mg,
44.5%
yield): 1H NMR (500 MHz, CD30D) 6 ppm 2.47 (dt, J=31.3, 12.1 Hz, 4H), 2.96 (t,
J=7.1 Hz,
2H), 4.24 (s, 1H), 4.28 (dd, J=11.2, 6.5 Hz, 1H), 4.34 (d, J=3.6 Hz, 1H), 4.42
(t, J=4.8 Hz, 1H),
4.64(t, J=5.0 Hz, 1H), 6.00 (d, J=5.0 Hz, 1H), 7.16-7.31 (m, 3H), 7.48 (d,
J=7.3 Hz, 1H), 8.38
(s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 31.23, 39.74, 65.95, 71.42, 75.67,
84.73, 89.71,
126
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
119.06, 123.26, 125.49, 128.29, 128.80, 141.30, 144.69, 147.40, 151.90,
155.69; 31P NMR
(203 MHz, CD30D) 6 ppm 16.89, 19.83; m/z (ESP): 602.1(M + H).
Example 25. Synthesis of compound d-2
o H
d' o
CV I-6N\ õ..,
r
NaOH cr__-OH -I- L-E1u0K soci, .s., HCHO .-
TI-F/Et0H-1 1 rt lel, ..- 1120,100 "C' r- ----''Af e0H I ,
' IC2C 3.DMS 0H , > EDCI, HOElt [M.D.\ DMF
F
X
cbK;rsia , N
CO [¨Cf.? Ac0-124
N N DI
N.,..1.,, I-1 m
ecH,
DEAD,H7h3 N
DAIH;FP, DI, 06, H2 Pd(21-1)2 cr\ o
/(.:: ci ,,,
it õLci mec.,
,
r¨ Dilp.L.:7ne
Accisiõr4 ,-
F F OAc OAc
F FL,
1 1 N.)....N
,1, Or el
.,,, ':.CI :` ... ¨ 1
i HO N ' .
NC 'CI
PO(CiVie), i7, 5),_, N2 'reLCI
t-r
HOj U Hz0 HO 6H OH 1,124
Ox0
p-Ts0H OH OH
OH OH
[00247] To a solution of 4-fluoroindan-1-one (2 g, 13.32 mmol, 1 eq.) in
Et0H (20 mL)
and THF (20 mL), cooled at 0 C, was added t-BuOK (1 M, 26.64 mL, 2.0 eq.),
followed by
addition of TOSMIC (3.90 g, 19.98 mmol, 1.5 eq.) in Et0H/THF (1:1, 20 mL). The
mixture
was stirred at RT overnight, cooled to 0 C, followed by addition of brine.
The mixture was
extracted with Et0Ac, and the extract was concentrated. The residue was
purified by column
chromatograph on silica gel, eluted with PE/EA (100:0 to 83:17), affording 4-
fluoroindane-1-
carbonitrile (910 mg, 5.65 mmol, 42.39% yield)
[00248] To a solution of NaOH (677.52 mg, 16.94 mmol, 3.0 eq.) in H20 (15
mL) was
added 4-fluoroindane-1-carbonitrile (910 mg, 5.65 mmol, 1 eq.). The mixture
was stirred at
100 C for 16 h. The mixture was cooled to rt, diluted with water (30 mL), and
extracted with
Et0Ac (2 x 40 mL). The water layer was pH-adjusted (to 2) and extracted with
Et0Ac (2 x 50
mL). The organic layers were combined and concentrated to dryness, affording 4-
fluoroindane-
1-carboxylic acid (905 mg, 5.02 mmol, 88.96% yield) as brown solid.
[00249] To a solution of 4-fluoroindane-1-carboxylic acid (1 g, 5.55 mmol,
1 eq.) in
Me0H (20 mL) and DMF (0.1 mL) was added Thionyl chloride (3.30 g, 27.75 mmol,
2.02 mL,
eq.) at rt. After the addition completion, the reaction mixture was stirred at
rt for 3 h, and then
concentrated, quenched with ice water (50 mL), extracted with Et0Ac (2 x 50
mL). The
combined organic layers were washed with brine, dried over Na2SO4, and
concentrated to
dryness, affording methyl 4-fluoroindane-1-carboxylate (1.02 g, 5.25 mmol,
94.63% yield)
127
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00250]
To a mixture of methyl 4-fluoroindane-1-carboxylate (1.02 g, 5.25 mmol, 1
eq.)
in DMSO (25 mL) under nitrogen atmosphere were added K2CO3 (2.18 g, 15.76
mmol, 3 eq.)
and HCHO (941.54 mg, 10.50 mmol, 38% purity, 2 eq.) at 0 C. The mixture was
stirred at rt
for 16 h. The reaction was quenched with H20 (75 mL). The mixture was
extracted with Et0Ac
(50 mL). The pH of the aqueous solution was adjusted to 3 with 3 N HC1. The
mixture was
extracted with EA (3 x 80 mL). The combined organic layers were washed with
brine, dried
(Na2SO4), filtered, and evaporated to dryness, giving 4-fluoro-1-
(hydroxymethypindane-1-
carboxylic acid (1.02 g, 4.85 mmol, 92.39% yield) .
[00251]
To a mixture of 4-fluoro-1-(hydroxymethyl) indane-1-carboxylic acid (1.02
g,
4.85 mmol, 1 eq.) and phenylmethanamine (519.96 mg, 4.85 mmol, 1 eq.) in DMF
(15 mL)
were added EDCI (1.40 g, 7.28 mmol, 1.5 eq.), HOBT (983.50 mg, 7.28 mmol, 1.5
eq.) and
D1PEA (627.14 mg, 4.85 mmol, 845.20 nL, 1 eq.). The mixture was stirred at RT
for 18 h.
Solvent was removed by evaporation, followed by addition of water (50 mL) and
EA (60 mL).
The organic layer was washed with brine, dried (Na2SO4), filtered, and
evaporated to dryness.
The residue was purified by column chromatograph on silica gel (eluent: PE/EA
from 100:0 to
50:50), giving N-benzy1-4-fluoro-1-(hydroxymethyl)indane-1-carboxamide (1.25
g, 4.18
mmol, 86.06% yield).
[00252]
To a mixture of N-benzy1-4-fluoro-1-(hydroxymethyl)indane-1-carboxamide
(500 mg, 1.67 mmol, 1 eq.) in THF (10 mL) under nitrogen atmosphere were added
PPh3
(657.17 mg, 2.51 mmol, 1.5 eq.) and, dropwise, DEAD (436.35 mg, 2.51 mmol,
394.53 !IL,
1.5 eq.) at 0 'C. The mixture was stirred at rt for 2 h. The reaction was
quenched by water (20
mL), followed by addition of Et0Ac (60 mL). The organic layer was washed with
brine, dried
(Na2SO4), filtered, and evaporated to dryness. The residue was purified by
column
chromatography on silica gel (eluent, PE/EA, 100:0 to 67:33), giving 1-benzy1-
4'-fluoro-
spiro[azetidine-3,1'-indane1-2-one (380 mg, 1.35 mmol, 80.87% yield).
1002531
To a solution of A1C13 (360.22 mg, 2.70 mmol, 2 eq.) in THF (4 mL), cooled
at
0 C, was added LiA1H4 (153.78 mg, 4.05 mmol, 3 eq.) at 0 'C. The mixture was
kept at 0 C
for 30 min, followed by addition of 1-benzy1-4'-fluoro-spiro[azetidine-3,1'-
indane]-2-one (380
mg, 1.35 mmol, 1 eq.) in THF (3 mL). The mixture was stirred at rt for 16 h,
diluted with
Et0Ac (20 ml), followed by slow addition of 15% aqueous NaOH at 0 C to
adjusting the pH
to 10. The organic layer was separated, and dried over MgSO4. The insoluble
material was
removed by filtration, and filtrate was concentrated to dryness. The residue
was purified by
128
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
column chromatography on silica gel (eluent: PE/EA, 100:0-67:33), affording 1-
benzy1-41-
fluoro-spiro[azetidine-3,1'-indane] (280 mg, 1.05 mmol, 77.54% yield) .
[00254] To a solution of 1-benzy1-4'-fluoro-spiro[azetidine-3,1'-
indane] (280 mg, 1.05
mmol, 1 eq.) in Me0H (15 mL) were added ammonium formate (99.07 mg, 1.57 mmol,
1.5
eq.) and Pd(OH)2 (64.64 mg, 523.69 lamol, 0.5 eq.) .The mixture was stirred at
under H2-
atmosphere at 60 C overnight. The insoluble material was filtered off, and
washed with
Me0H. The filtrate and washing were combined and concentrated to dryness,
affording 4'-
fluorospirolazetidine-3,1'-indane] (148 mg, 835.13 umol, 79.74% yield).
[00255] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-yl]methyl acetate (340 mg, 760.24 mmol, 1.0 eq.) in 1,4-
dioxane (10 mL)
were added 4'-fluorospiro[azetidine-3,1'-indane] (148.20 mg, 836.27 'Limo],
1.1 eq.) and DIPEA
(442.14 mg, 3.42 mmol, 595.88 pL, 4.5 eq.). The mixture was stirred at 100 C
for 3 h.
[00256] The mixture was concentrated and the residue was
purified by column
chromatography on silica gel eluted with PE/EA (100:0-60:40), affording
[(2R,3R,4R,5R)-3,4-
diacetoxy-542-thloro-6-(4'-fluorospiro [azetidine-3,1'-indane] -1 -yl)purin-9-
yl[tetrahydrofuran-2-yl[methyl acetate (290 mg, 493.21 limo', 64.88% yield)
[00257] To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(4'-
fluorospiro[azetidine-3,1'-indane1-1-yl)purin-9-yl]tetrahydrofuran-2-yl]methyl
acetate (290
mg, 493.21 imol, 1 eq.) in Me0H (5 mL) was added NH3-Me0H (7 M, 1.76 mL, 25
eq.).The
mixture was stirred at room temperature for 2 h, and concentrated to dryness,
giving
(2R,3R,4S,5R)-242-chl oro-6-(4'-fluorospiro [azetidine-3, -indanel -1 -
yl)purin-9-y11-5-
(hydroxymethyl)tetrahydrofuran-3,4-diol (227 mg, 491.48 limo', 99.65% yield).
It was used in
the next step directly.
[00258] To a solution of (2R,3R,4S,5R)-242-chloro-6-(4'-
fluorospiro[azetidine-3,1-
indane1-1-yl)purin-9-y11-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (227 mg,
491.48 limo', 1
eq.) in acetone (10 mL) were added 2,2-dimethoxypropane (1.02 g, 9.83 mmol, 20
eq.) and
Ts0H-H20 (97.31 mg, 491.48 larnol, 1 eq). The mixture was stirred at rt. for 1
h. Solvent was
removed by evaporation. The residue was diluted with EtOAc (50 mL), washed
with aqueous
NaHCO3 and brine successively, and concentrated. The residue was purified by
column
chromatography on silica gel eluted with PE/EA (100:0-50:50), affording
[(3aR,4R,6R,6aR)-
4-[2-chloro-6-(4'-fluorospiro[azetidine-3,1'-indane]-1-yOpurin-9-y11-2,2-
dimethyl-3a,4,6,6a-
tetrahydrofuro[3,4-d][1,31dioxol-6-yl]methanol (170 mg, 338.69 [Imo], 68.91%
yield) .
129
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00259]
To a solution of [(3aR,4R,6R,6aR)-442-chloro-6-(4'-fluorospiro[azetidine-
3,1'-
indanel -1-yl)purin-9-yll -2,2-dimethy1-3a,4,6,6a-tetrahydrofuro [3,4-d] [1,3]
di oxo1-6-
yllmethanol (170 mg, 338.69 tunol, 1 eq.) in PO(Et0)3 (2.5 mL), cooled at 0
C, was added a
solution of bis(dichlorophosphoryl)methane (74.65 mg, 298.84 ttmol, 2.5 eq.)
in PO(E1.0)3 (2.5
mL). The mixture was stirred at 0 'V for 4 h, followed by introduction of H20
(2 mL) into the
reaction mixture. The mixture was stirred at room temperature overnight.
Purification of the
reaction mixture with C18 reversed phase silica gel (gradient eluent, 0 to 25%
ACN in water)
gave Compound d-2, (40.2 mg, 19.15% yield); 1H NMR (500 MHz, Me0D) 6 ppm 2.52
(dt, J
= 42.1, 14.0 Hz, 4H), 3.00 (t, J= 7.0 Hz, 2H), 4.21 -4.50 (m, 4H), 4.65 (s,
1H), 6.01 (d, J=
5.0 Hz, 1H), 6.96 (t, J= 8.4 Hz, 1H), 7.26 -7.38 (m, 2H), 8.40 (s, 1H). m/z
(ESI-1): 620.3(M+H).
Example 26. Synthesis of compound d-3
COOH
TH NHac 0H F õCo :I-1 s K C OH , HD M S
D 0 r
F E CI, HODI
Bt, PE4 DMF'
FO. T F:EMtO ICH % F,C13
AO
DEAD, PPM13
N 12,1-12, AICI, H2, Pc1C2H)2
F Fr
OAc OAc
THF THF
Me0H, NCWOOH
F ,o0
-124
OAc UA,
\
0 0
NH,-MOH N p-Te0H CI 'N>
Me0H
o J
P0(0Et),
HO Nv N'CI H,0 9 9 Cirrij
HO (1 N CI
"
OH OH OH OH
[00260]
To a solution of 5-fluoroindan-1 -one (6.0 g, 39.96 mmol, 1 eq.) in EtOH
(20
mL) and THF (20 mL), cooled at 0 C, was added t-BuOK (8.97 g, 79.92 mmol, 2.0
eq.) in
THF (40 mL), followed by addition of TOSMIC (11.70 g, 59.94 mmol, 1.5 eq.) in
1:1 Et0H
and THF (50 mL). The mixture was stirred at room temperature overnight, and
then cooled to
about 0 C, followed by addition of brine. The mixture was extracted with
Et0Ac (2 x 120
mL). The organic layers were combined, washed with brine, dried (Na2SO4),
filtered, and
concentrated. The residue was purified via column chromatograph on silica gel
eluted with
PE/EA (100:0 to 80:20), affording 5-fluoroindane-l-carbonitrile (2.15 g, 13.34
mmol, 33.38%
yield).
[00261]
To a mixture of 5-fluoroindane-1-carbonitrile (2.5 g, 15.51 mmol, 1 eq.)
in H20
(30 mL) was added NaOH (1.86 g, 46.53 mmol, 3 eq.). The mixture was stirred at
100 C for
16 h, and then cooled to room temperature, followed by addition of water (30
mL). The mixture
130
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
was extracted with Et0Ac (2 x 30 mL). pH of the aqueous layer was adjusted 2,
and the mixture
was extracted with Et0Ac (2 x 30 mL). The combined organic layers were washed
with brine,
dried (Na2SO4), filtered, and evaporated to dryness, giving 5-fluoroindane-1 -
carboxylic acid
(2.7 g, 14.99 mmol, 96.61% yield) as brown solid.
[00262] To a solution of 5-fluoroindane-1 -carboxylic acid (2.7
g, 14.99 mmol, 1
eq.) in CH3OH (40 mL), cooled at 0 C, was added 1 drop of DMF, followed by
addition of
S0C12 (7.13 g, 59.94 mmol, 4.35 mL, 4 eq.). The mixture was stirred at room
temperature for
3 h. Solvent was removed by evaporation at about 40 C bath temperature. To
the residue was
added were added water (60 mL) and Et0Ac (40 mL) and mixed well. The organic
layer was
separated, washed with NaHCO3 (aq.) and NaCl (aq.) successively, dried (sodium
sulfate),
filtered, and evaporated to dryness, giving methyl 5-fl uoroi n dan e-1-
carboxyl ate (2.8 g, 14.42
mmol, 96.21% yield) as brown oil.
[00263] To a solution of methyl 5-fluoroindane-1-carboxylate
(2.8 g, 14.42 mmol, 1
eq.) in DMSO (30 mL) were added K2CO3 (5.98 g, 43.25 mmol, 3 eq.) and HCHO
(3.98 g,
43.25 mmol, 37% purity, 3 eq.). The mixture was stirred at room temperature
for 16 h. The
reaction was quenched with water (100 mL). The mixture was extracted with
Et0Ac (2 x 80
mL2). pH of the aqueous layer was adjusted to 3 with 6 N HC1, followed by
extraction with
Et0Ac (2 x 60 mL). The organic layers were combined, washed with brine, dried
(Na2SO4),
filtered, and evaporated to dryness, giving 5-fluoro-1-(hydroxymethypindane-1-
carboxylic
acid (3.0 g, 14.27 mmol, 98.99% yield).
[00264] To a solution of 5-fluoro-1-(hydroxymethypindane-1-
carboxylic acid (3.0 g,
14.27 mmol, 1 eq.) in DMF (40.00 mL) were added phenylmethanamine (1.53 g,
14.27 mmol,
1.56 mL, 1 eq.) , DIPEA (3.32 g, 25.69 mmol, 4.47 mL, 1.8 eq.), HOBT (2.89 g,
21.41 mmol,
1.5 eq.) and EDCI (410g. 21.41 mmol, 1.5 eq.). The mixture was stirred at room
temperature
for 5 h. The reaction was quenched with water (20 mL), followed by addition of
Et0Ac (100
mL). The organic layer was separated, washed with brine, dried (Na2SO4),
filtered, and
evaporated to dryness. The residue was purified by column chromatography on
silica gel
eluted with PE/EA (100:0 to 50:50), giving N-benzy1-5-fluoro-1-
(hydroxymethypindane-1-
carboxamide (3.6 g, 12.03 mmol, 84.27% yield).
1002651 To a mixture of N-benzy1-5-fluoro-1 -(hy droxy methy
1)indane-l-carb oxami de
(3.6 g, 12.03 mmol, 1 eq.) in THF (40 mL) under nitrogen atmosphere were added
PPh3 (3.79
g, 14.43 mmol, 1.2 eq.) and, dropwise, DEAD (2.51 g, 14.43 mmol, 2.27 mL, 1.2
eq.) at 0 C.
The mixture was stirred at room temperature for 16 h. The reaction was
quenched with
131
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
water (20 mL)., followed by addition of Et0Ac (100 mL). The organic layer was
separated,
washed with brine, dried (Na2SO4), filtered, and evaporated to dryness. The
residue
was purified by column chromatograph on silica gel eluted with PE/EA (100:0 to
70:30),
affording 1 -b enzy1-5'-fl uoro-s piro [azeti dine-3,1'-indane] -2-one (3.0 g,
10.66 mmol, 88.67%
yield) as white solid.
[00266]
To a solution of A1C13 (473.97 mg, 3.55 mmol, 2 eq.) in THF (15 mL),
cooled at
0 C, was added LiA11-14 (202.35 mg, 5.33 mmol, 3 eq.). The mixture was
stirred at 0 C for 20
min, followed by addition of 1-benzy1-5'-fluoro-spiro[azetidine-3,1'-indane]-2-
one (500 mg,
1.78 mmol, 1 eq.; dissolved in 5 mL of THF). The mixture was stirred at room
temperature for
h, and diluted with THF (10 ml). The reaction was quenched by slow addition to
the mixture
of 15% aqueous NaOH at 0 C until the pH of the mixture reached 9. The organic
layer was
separated, dried over MgSO4. The insoluble material was removed by filtration,
and the filtrate
was concentrated to dryness, affording 1-benzy1-5'-fluoro-spiro[azetidine-3,1'-
indane] (475
mg, 1.78 mmol, 99.97% yield).
[00267]
To a solution of 1-benzy1-5'-fluoro-spiro[azetidine-3,1'-indane1 (475 mg,
1.42
mmol, 1 eq.) in Me0H (10 mL) were added ammonium formate (134.45 mg, 2.13
mmol, 1.5
eq.) and Pd(OH)2 (50 mg, 20% on carbon, wetted with ca.50% water). The mixture
was stirred
at under H2 atmosphere at 60 C overnight. The insoluble material was removed
by filtration,
and washed with Me0H. The filtrate and the washing were combined, and
concentrated to
dryness, affording 5'-fluorospiro[azetidine-3,1'-indane[ (250 mg, 99.25%) and
used directly
for the next step.
[00268]
To a mixture of 5'-fluorospiro[azetidine-3,1'-indane1 (86 mg, 97.056 wnol,
1 eq.)
in 1,4-dioxane (10 mL) were added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (217.03 mg, 485.28 iamol, 1 eq.) and
DIPEA (156.79
mg, 1.21 mmol, 211.31 !IL, 2.5 eq.). The mixture was stirred at 100 C for 4
h. Solvent was
removed by evaporation. The residue was diluted in Et0Ac (50 mL), washed with
water and
brine, successively. The organic layer was concentrated and the residue was
purified by column
chromatography on silica gel eluted with PE/EA (100:0-50:50), affording
[(2R,3R,4R,5R)-3,4-
di acetoxy-5- [2-chl oro-6-(5'-fl uorospi ro [azeti dine-3,1'-indane] -1-
yl)purin-9-
ylitetrahydrofuran-2-ylimethyl acetate (190 mg, 323.14 tamol, 66.59% yield) as
white solid.
[00269] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(51-
fl uoros pi ro [azeti di n e-3,1'-i n dan e1-1 -yl)puri n-9-yl[tetrahy
drofuran -2-yl] methyl acetate (190
mg, 323.14 wnol, 1 eq.) in Me0H (2.00 mL) was added NH3-Me0H (7 M, 1.38 mL, 30
eq.).
132
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
The mixture was stirred at room temperature overnight. Solvent was removed by
evaporation.
The residue was diluted with Et0Ac (30 mL), washed with brine (30 mL), dried
with Na2SO4,
and concentrated to dryness, affording (2R,3R,4S,5R)-242-chloro-6-(5'-
fluorospiro[azetidine-
3,1'-indane1-1 -yl)p urin-9-y 11 -5 -(hy droxymethyl)tetrahydrofuran-3,4-diol
(149 mg, 322.60
wriol, 99.83% yield) as white solid.
[00270]
To a solution of (2R,3R,4S,5R)-242-chloro-6-(5'-fluorospiro[azetidine-3,11-
indane1-1-y1)purin-9-y11-5-(hydroxymethyptetrahydrofuran-3,4-diol (149 mg,
322.60 limo', 1
eq.) in acetone (10 mL) were added p-Ts0H.H20 (61.36 mg, 322.60 mmol, 1 eq.)
and 2,2-
dimethoxypropane (671.96 mg, 6.45 mmol, 793.34 mL, 20 eq.). The mixture was
stirred at
room temperature for 2 h. The mixture was basified to pH 9 by slow addition of
aqueous
NaHCO3 (aq.) at 0 C. Solvent was removed by evaporation, and the residue was
extracted with
Et0Ac (2 x 30 mL). The organic layer was washed by brine, and concentrated.
The residue
was purified by column chromatography on silica gel eluted with PE/EA (100:0-
50:50),
giving [(3aR,4R,6R,6aR)-442-chloro-6-(5'-fluorospiro[azetidine-3,1'-indane1-1-
yl)purin-9-
y11-2,2-dimethyl-3 a,4,6,6a-tetrahy drofuro [3,4-d] [1,3] di oxol -6-yll
methanol (138 mg, 274.94
limo', 85.22% yield) as white solid.
[00271]
To a solution of [(3aR,4R,6R,6aR)-442-chloro-6-(5'-fluorospiro[azetidine-
3,1'-
indanel -1-yl)purin-9-yll -2,2-dimethy1-3a,4,6,6a-tetrahy drofuro [3,4-d]
[1,3] di oxo1-6-
yllmethanol (138 mg, 274.94 [imol, 1 eq.) in PO(OEt)3 (1.5 mL), cooled at 0
C, was added a
solution of bis(dichlorophosphoryl)methane (171.69 mg, 687.34 limo', 2.5 eq.)
in PO(OEt)3
(1.5 mL). The mixture was stirred at 0 C for 4 h, followed by addition of
water (2 mL), and
left stirred at 25 C overnight. Purification of the reaction mixture was
achieved using C18
reversed phase silica gel (0 to 30% ACN in water as gradient eluent), giving
Compound d-3,
(40 mg, 64.53 itimol, 23.55% yield). 1H NMR (500 MHz, Me0D) 6 ppm 2.47 ¨ 260
(m, 4H),
3.00 (t, J = 6.8 Hz, 2H), 4.25 ¨ 4.43 (m, 4H), 4.43 ¨ 4.52 (m, 2H), 4.68 (m,
3H), 6.04 (d, J =
4.8 Hz, 1H), 7.00 (d, J = 9.0 Hz, 2H), 7.53 (s, 1H), 8.45 (s, 1H); m/z (ESt):
620.0(M+H).
Example 27. Synthesis of compound d-4
133
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
..L--. > rrN "41--0-.
F' ', f.,:r \I> TOSMIC f ,'q NaOH , ilo WON , HUHO
F `-'" I H-1,3 F" "," uOK eN ,OSOCI, F `-' '---
--0K2CO3,DMS0
-t _cr-- HO EDCI, HOPI, Eld FIN, ,
DIPEA, DMF
HO
,-----__
CI
\ / r
1-17,a
ALO N N¨ci µ14'
riN'L
DEAD, PPh3 F ' ,/ LiAIH4, AlCla F,õ. - 0 H2,
Pd(OH)2F. ,,a, 1Z.,
¨IOAG O AGO Ac __11,
N -C1 "rMe H
THE isi .- .- 0-- 0 THF N ane McOH, NH4COOH
'14 DIPEA, Diox
) ¨I-?1 Me0H, a
H
\ ,,) 100 C, 16 h
./,----
\\ OAc OAc
Xilt, 0, 0 KPI-1):, Clc;zPP,1 0 0
¨ N GI P0(0Me rµiX1'''l
HO 'N N' -'CI X N'CI
¨04 HO'F,''''P!
0 )a OH uH --I--
11 I420
OH OH ,--- -,,, OH OH
p-Ts0H 02/0
RP02030-1
[00272] To a solution of 6-fluoroindan-1-one (2 g, 13.32 mmol,
1 eq.) in Et0H (9
mL) and THF (9 mL), cooled at 0 'V, was added a mixture of t-BuOK (2.99 g,
26.64 mmol,
2.0 eq.) in THF (25 mL), followed by addition of a mixture of TOSMIC (3.90 g,
19.98 mmol,
1.5 eq.) in Et0H/THF (1:1, 25 mL). The mixture was stirred at room temperature
for 16 h,
cooled to 0 C, followed by addition of brine. The mixture was extracted with
Et0Ac and the
extract was concentrated. The residue was purified (column chromatograph on
silica gel, eluted
with PE/Et0Ac from 100:0 to 85:15), affording 6-fluoroindane-1-carbonitrile
(1.3 g, 60.6%).
[00273] To a mixture of 6-fluoroindane-1-carbonitrile (2.6 g,
16.13 mmol, 1 eq.) in H20
(25 mL) was added NaOH (1.94 g, 48.39 mmol, 3.0 eq.). The mixture was stirred
at 100 0C for
h, then cooled to room temperature. The mixture was diluted with water (10
mL), and
extracted with Et0Ac (2 x 35 mL). pH of the aqueous layer was adjusted to 2,
and the layer
was extracted with Et0Ac (2 x 20 mL). The extracts were combined, washed by
brine, dried
with Na2SO4, and concentrated, affording 6-fluoroindane-1-carboxylic acid (2.7
g, 92.9%) as
light yellow solid.
[00274] To a solution of 6-fluoroindane-l-carboxylic acid (2.8
g, 15.54 mmol,
1 eq.) in Me0H (40 mL), cooled at 0 C, was added 1 drop of DMF, followed by
addition of
SOC12 (7.40 g, 62.16 mmol, 4.51 mL, 4.0 eq.). The mixture was stirred at room
temperature
overnight. The solvent was removed by evaporation, and the residue was treated
with water
(50 mL) and Et0Ac (100 mL). The organic layer was separated, washed first with
aqueous
NaHCO3 and then with brine, dried with Na2SO4, and evaporated, giving methyl 6-
fluoroindane-1-carboxylate (3 g, 99.4%) as brown oil.
134
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00275] To a solution of methyl 6-fluoroindane-1-carboxylate (3
g, 15.45 mmol,
1 eq.) in DMSO (30 mL) were added K2CO3 (7.05 g, 50.98 mmol, 3.3 eq.) and HCHO
(4.27 g,
46.34 mmol, 37% purity, 3.0 eq.). The mixture was stirred at room temperature
for 16 h. The
resulting solution was quenched with H20 (100 mL), extracted with Et0Ac (2 x
80 mL). pH
of the aqueous was adjusted to 3 with 6 N HC1, and the aqueous solution was
extracted with
Et0Ac (2 x 60 mL). The extracts were combined, washed with brine, dried
(Na2SO4), filtered,
and evaporated, giving 6-fluoro-1-(hydroxymethyl)indane-1-carboxylic acid
(2.37 g,
72.9%) as light yellow solid.
[00276] To a solution of 6-fluoro-1-(hydroxymethyl)indane-1-
carboxylic acid (2.37 g,
11.27 mmol, 1 eq.) in DMF (27 mL) were added phenylmethanamine (1.21 g, 11.27
mmol,
1.23 mL, 1.0 eq.), DIPEA (2.62 g, 20.29 mmol, 3.53 mL, 1.8 eq.), EDCI (3.24 g,
16.91 mmol,
1.5 eq.) and HOBT (2.29 g, 16.91 mmol, 1.5 eq.). The mixture was stirred at
room temperature
for 5 h. The mixture was diluted with Et0Ac (100 mL), and water (100 mL). The
organic layer
was separated, washed by brine (5 x 100 mL), dried with Na2SO4, and
concentrated,
affording N-benzy1-6-fluoro-1-(hydroxymethyl)indane-1-carboxamide (3.2 g,
94.8%) as
brown oil.
1002771 To a solution of N-benzy1-6-fluoro-1-
(hydroxymethyl)indane-1-carboxamide (1
g, 3.34 mmol, 1 eq.) in THF (15 mL) were added PPh3 (1.05 g, 4.01 mmol, 1.2
eq.), followed
by addition, at 0 C, of DEAD (698.14 mg, 4.01 mmol, 631.23 u.L, 1.2 eq.). The
mixture was
stirred at room temperature overnight, diluted with Et0Ac (40 mL). The organic
layer was
separated, washed with brine, and concentrated. The residue was purified on a
silica gel column
(eluted with PE/Et0Ac from 100:0 to 60:40), affording 1-benzy1-6'-fluoro-
spiro[azetidine-3,1'-
indaneJ-2-one (680 mg, 72.3%) as yellow solid.
[00278] To a solution of A1C13 (644.60 mg, 4.83 mmol, 2.0 eq.)
in THF (15 mL), cooled
at 0 C, was added LiA1H4 (275.19 mg, 7.25 mmol, 3.0 eq.). The mixture was
stirred at 0 C
for 15 min, followed by addition of 1-benzy1-6'-fluoro-spiro[azetidine-3,1'-
indane1-2-one (680
mg, 2.42 mmol, 1 eq.; in THF, 8 mL). The mixture was stirred at room
temperature overnight;
and the reaction was quenched with H20 (0.6 mL), followed by addition of 15%
aqueous NaOH
(2.5 mL) and Et0Ac (20 mL). The solid material was removed by filtration and
the filtrate was
concentrated, affording 1-benzy1-6'-fluoro-spiro[azetidine-3,1'-indanel (600
mg, 92.8%).
[00279] To a solution of 1-benzy1-6'-fluoro-spiro[azetidine-3,1'-
indane] (600 mg, 2.24
mmol, 1 eq.) in Me0H (12 mL) were added Pd(OH)2 (100 mg) and ammonium formate
(212.29 mg, 3.37 mmol, 1.5 eq.). The mixture was stirred at 60 C under H2
atmosphere for 5
135
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
h. The insoluble material was removed by filtration, washed with Me0H. The
filtrate and
washing were combined and concentrated to dryness, affording 6'-
fluorospiro[azetidine-3,11-
indanel (390 mg, 98.1%).
[00280]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yptetrahydrofuran-2-yll methyl acetate (500 mg, 1.12 mmol, 1 eq.) in 1,4-
dioxane (20
mL) were added 6'-fluorospiro [azetidine-3,1' -indane] (237.76 mg,
1.34 mmol,
1.2 eq.) and DIPEA (577.96 mg, 4.47 mmol, 778.93 tiL, 4.0 eq.). The mixture
was stirred at
100 C for 3 h. The mixture was concentrated and the residue was purified by
column
chromatography on silica gel eluted with (PE/Et0Ac from 100:0 to 60:40),
giving 1(2R,3R,4R,5R)-3 ,4-di acetoxy -5 - [2-chloro-6-(6'-fluorospiro [azeti
dine-3,1' -indane] -1 -
yl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (500 mg, 76.1%) as white
solid.
[00281] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-(6'-
fluorospiro[azetidine-3,1'-indane]-1-y1)purin-9-ylltetrahydrofuran-2-yllmethyl
acetate (500
mg, 850.36 ['mot, 1 eq.) in Me0H (2 mL) was added NH3-Me0H (7 M, 3.64 mL, 30
eq.). The
mixture was stin-ed at room temperature overnight. The solvent was removed by
evaporation.
The residue was diluted with Et0Ac, washed (first with water, and then with
brine), dried
(Na2SO4), and concentrated, affording (2R,3R,4S,5R)-2-[2-chloro-6-(6'-
fluorospiro [azetidine-
3,1 '-indane] -1 -y -5 -(hy droxymethyl)tetrahydrofuran-3,4-diol
(380 mg, 96.7%).
[00282]
To a solution of (2R,3R,4S,5R)-242-chloro-6-(6'-fluorospiro [azetidine-
3,1'-
indane[ -1-yl)purin-9-yl[ -5-(hydroxymethyl)tetrahy drofuran-3,4-diol (380 mg,
822.74 timol,
1 eq.) in acetone (15 mL) were added p-Ts0H (141.68 mg, 822.74 tunol, 1 eq.)
and 2,2-
dimethoxypropane (1.29 g, 12.34 mmol, 15 eq.). The mixture was stirred at room
temperature
for 3 h. The mixture was diluted with Et0Ac (25 mL), washed with aq. NaHCO3,
followed
washing with brine, and concentrated. The residue was purified by column
chromatography
on silica gel eluted with PE/Et0Ac from 100:0 to 60:40, giving
[(3aR,4R,6R,6aR)-442-chloro-
6-(6'-fluorospiro[azetidine-3,1' -indane] -1 -y 1)purin-9-yll -2,2-dimethy1-3
a,4, 6,6a-
1e trahy drofuro [3,4-d] [1,3] di oxo1-6-yll me thanol (200 mg, 48.4%) as
white solid.
[00283]
To a solution of 1(3aR,4R,6R,6aR)-442-chloro-6-(6'-fluorospiro[azetidine-
3,1'-
indanel -1-yl)purin-9-yll -2,2-dimethy1-3a,4,6,6a-tetrahydrofuro [3,4-d] [1,3]
di oxo1-6-
yllmethanol (200 mg, 398.46 ttmol, 1 eq.) in P0(0E03 (2 mL), cooled at 0 C,
was added
bis(dichlorophosphoryl)methane (248.82 mg, 996.14 ttmol, 2.5 eq.) in PO(OEt)3
(2 mL). The
mixture was stirred at about 0 C for 5 h. H20 (2 mL) was added into the
mixture at 0 C. The
mixture was stirred at about 25 'V overnight, and mixture was directly
injected into the column
136
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
for purification (C18 reversed phase silica gel, 0 to 30% ACN in water as
gradient eluent),
giving Compound d-4, (75 mg, 30.3% yield); 1HNMR (500 MHz, CD30D) 6 ppm 2.41-
2.57
(m, 4H), 2.93 (t, J=7.0 Hz, 2H), 4.21-4.39 (m, 3H), 4.42 (t, J=4.7 Hz, 1H),
4.64 (t, J=4.9 Hz,
1H), 6.00 (d, J=4.9 Hz, 1H), 6.95 (1, J=8.6 Hz, 1H), 7.23 (t, J=9.1 Hz, 2H),
8.39 (s, 1H); rniz
(ESI+):620.1 (M+H).
Example 28. Synthesis of compound d-5
HaN\
c
TOSMIC NaOH Me0H 2 pc; HCHO
THar eN N20 H -0 SOCl2 FOKZCODMSOHOf
HCi EDCI, HOBt,
DIPEA, DMF
coA NNLCI
DEAD, PPII3 0 7LiAIH,AICI. H2, Pd(OH)2 OAc OAc
70-Aco
THF THF I Me0H, NH4COON DIPEA,
Dioxano McOH, rt
Lo
100 C 16 h
OAc OAc
<N> F F F
-r(
0 0 N
0. o 0
HO N- 'le Ho __________ NNCI CI' 'CI
jo P0(0Me)2 HO OH OH
H0
OH OH 002 OH OH
p-1s0H RP02940-1
[00284] To a solution of 7-fluoroindan-1-one (4 g, 26.64 mmol, 1
eq.) in Et0H (15
mL) and THF (15 mL), cooled at 0 C, was added t-BuOK (5.98 g, 53.28 mmol, 2.0
eq.) in
THF (40 mL), followed by addition of TOSMIC (7.80 g, 39.96 mmol, 1.5 eq.) in
Et0H/THF
(1:1, 50 mL). The mixture was stirred at room temperature for 16 h, then
cooled to 0 C. After
addition of brine, the mixture was extracted with Et0Ac. The organic layer was
concentrated
and the residue was purified using column chromatography on silica gel, eluted
with
PE/Et0Ac from 100:0 to 85:15, affording 7-fluoroindane-1-carbonitrile (2.5 g,
58.2%) as
brown oil.
[00285] To a mixture of 7-fluoroindane-1-carbonitrile (2.5 g,
15.51 mmol, 1 eq.) in H20
(25 mL) was added NaOH (1.86 g, 46.53 mmol, 3.0 eq.). The mixture was stirred
at 110 C for
16 h, and then cooled to room temperature. To dilution with water (10 mL), the
mixture was
extracted with Et0Ac (2 x 35 mL). pH of the water layer was adjusted to, and
the layer was
extracted with Et0Ac (2 x 20 mL). The extracts were combined, washed by brine,
dried with
Na2SO4, and concentrated, affording 7-fluoroindane-1-carboxylic acid (2.6 g,
93.0%).
137
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00286] To a solution of 7-fluoroindane-1-carboxylic acid (2.7
g, 14.99 mmol,
1 eq.) in Me0H (25 mL), cooled at 0 C, was added 1 drop of DMF, followed by
addition of
S0C12 (7.13 g, 59.94 mmol, 4.35 mL, 4.0 eq.). The mixture was stirred at 25 C
for 4 h. Solvent
was removed by evaporation. The residue was diluted with water (50 mL), and
extracted with
Et0Ac (100 mL). The organic layer was washed (first with aq. NaHCO3, and then
with brine),
dried (Na2SO4), filtered, and evaporated to dryness, giving methyl 7-
fluoroindane-1-
carboxylate (2.7 g, 92.8%) as brown oil.
[00287] To a solution of methyl 7-fluoroindane-l-carboxylate
(2.7 g, 13.90 mmol,
1 eq.) in DMSO (27 mL) were added K2CO3 (6.34 g, 45.88 mmol, 3.3 eq.) and HCHO
(3.84 g,
41.71 mmol, 37% purity, 3.0 eq.). The mixture was stirred at room temperature
overnight. The
reaction was quenched with H20 (100 mL), and the mixture was extracted with
Et0Ac (2 x 80
mL). pH of the aqueous layer was adjusted to 3 with 6 N HC1, and the layer was
extracted with
Et0Ac (2 x 60 mL). The extracts were combined, washed with brine, dried
(Na2SO4), filtered,
and evaporated, giving 7-fluoro-1-(hydroxymethyl)indane-1-carboxylic acid (2.2
g, 75.3%).
[00288] To a solution of 7-fluoro-1-(hydroxymethyl)indane-1-
carboxylic acid (2.2 g,
10.47 mmol, 1 eq.) in DIVif (25 mL) were added phenylmethanamine (1.12 g,
10.47 mmol,
1.14 mL, 1.0 eq.), D1PEA (2.43 g, 18.84 mmol, 3.28 mL, 1.8 eq.), HOST (2.12 g,
15.70 mmol,
1.5 eq.) and EDCI (3.01 g, 15.70 mmol, 1.5 eq.). The mixture was stirred at
room temperature
for 16 h, and then diluted with Et0Ac (100 mL) and water (100 mL). The organic
layer was
separated, washed with brine (5 x 100 mL), dried with Na2SO4, and
concentrated, affording N-
benzy1-7-fluoro-1-(hydroxymethypindane-1-carboxamide (2.97 g, 94.8%) as yellow
solid.
[00289] To a solution of N-benzy1-7-fluoro-1-
(hydroxymethyl)indane-1-carboxamide (1
g, 3.34 mmol, 1 eq.) in THF (15 mL) were added PPh3 (1.14 g, 4.34 mmol, 1.3
eq.). DEAD
(756.32 mg, 4.34 mmol, 1.3 eq.) at about 0 C. The mixture was stirred at room
temperature
for 3 h, diluted with Et0Ac (40 mL). The organic layer was washed with brine,
and
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/EA from 100:0 to 60:40, affording 1-benzy1-7'-fluoro-spiro[azetidine-3,1'-
indane] -2-one
(880 mg, 93.6%) as light yellow solid.
[00290] To a solution of A1C13 (853.15 mg, 6.40 mmol, 2.0 eq.)
in THF (15 mL), cooled
at 0 C, was added LiA1H4 (364.22 mg, 9.60 mmol, 3.0 eq.). The mixture was
stirred at 0 C for
15 min, followed by addition of a solution of 1-benzy1-7'-fluoro-
spiro[azetidine-3,1'-indane]-
2-one (900 mg, 3.20 mmol, 1 eq.) in THF (10 mL). The mixture and stirred at
room temperature
overnight, and the reaction was quenched with H20 (0.6 mL), followed by
addition of 15% aq.
138
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
NaOH (3.5 mL) and Et0Ac (20 mL). The solid was removed by filtration and the
filtrate was
concentrated to dryness, affording 1 -benzy1-7'-fluoro-spiro [azetidine-3,1 '-
indanel (730 mg,
85.4%).
[00291] To a solution of 1-benzy1-7'-fluoro-spiro[azetidine-3,1'-
indanel (730 mg, 2.73
mmol, 1 eq.) in Me0H (15 mL) were added Pd(OH)2 (130 mg, 2.73 mmol,
1 eq.) and ammonium formate (258.29 mg, 4.10 mmol, 1.5 eq.). The mixture was
stirred at
60 C under H2 atmosphere for 5 h. The insoluble material was removed by
filtration, and
washed with Me0H. The filtrate and the washing were combined and concentrated
to dryness,
affording 7'-fluorospiro[azetidine-3,1'-indane] (480 mg, 99.2%).
[00292] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yl)tetrahydrofuran-2-yllmethyl acetate (500 mg, 1.12 mmol, 1 eq.) in 1,4-
dioxane (20
mL) were added 7'-fluorospiro[azetidine-3,1'-indane] (237.76
mg, 1.34 mmol,
1.2 eq.) and DIPEA (577.96 mg, 4.47 mmol, 778.93 !IL, 4.0 eq.). The mixture
was stirred at
100 C for 3 h. The mixture was concentrated and the residue was purified by
column
chromatography on silica gel eluted with PE/Et0Ac (100:0 to 60:40), giving
[(2R,3R,4R,5R)-
3,4-di acetoxy-5 42-chl oro-6-(7'-fluoro spi ro [azeti dine-3,1' -indane] -1-
yl)purin-9-
yl] tetrahydrofuran-2-yl] methyl acetate (300 mg, 45.6%) as white solid.
[00293] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-(71-
fluorospiro[azetidine-3,1'-indane1-1-y1)purin-9-ylltetrahydrofuran-2-yllmethyl
acetate (300
mg, 510.22 tmol, 1 eq.) in Me0H (2 mL) was added NH3-Me0H (7 M, 2.19 mL, 30
eq.). The
mixture was stirred at room temperature overnight. The solvent was removed by
concentration
and the residue was diluted with Et0Ac, washed with water and brine
successively, dried with
Na2SO4, and concentrated to dryness, affording (2R,3R,4S,5R)-2-[2-chloro-6-(7'-
fluorospiro [azeti dine-3,1 ' -indane] -1 -y Dpurin-9-yll -5 -(hy
droxymethyl)tetrahy drofuran-3,4-
diol (200 mg, 84.9%).
1002941 To a solution of (2R,3R,4S,5R)-2-1-2-chloro-6-(7'-
fluorospiro[azetidine-3,1'-
indane]-1-y1)purin-9-yll -5-(hy droxy me thyl) te trahy drofuran-3,4-diol (200
mg, 433.02 1.4mol,
1 eq.) in acetone (10 mL) were added p-Ts0H (74.57 mg, 433.02 pmol, 1 eq.) and
2,2-
dimethoxypropane (676.47 mg, 6.50 mmol, 15 eq.). The mixture was stirred at
room
temperature for 3 h. The mixture was diluted with Et0Ac (25 mL), washed with
aq. NaHCO3,
and
brine successively, and concentrated. The residue was purified by column
chromatography on silica gel, eluted with PE/Et0Ac from 100:0 to 60:40,
giving [(3aR,4R,6R,6aR)-4-]2-chloro-6-(7'-fluorospiro [az eti -1-
yl)purin-9-
139
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
yl] -2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3 ,4-d] [1,3] di oxol -6-yl]
methanol (140 mg,
64.4%) as white solid.
[00295] To a solution of 1(3aR,4R,6R,6aR)-442-chloro-6-(7'-
fluorospiro[azetidine-3,1'-
indanel -1-yl)p urin-9-yl] -2,2-dimethy1-3a,4,6,6a-tetrahy drofuro [3,4-d]
[1,3] di oxo1-6-
yllmethanol (140 mg, 278.92 umol, 1 eq.) in P0(0E03 (1.5 mL), cooled at 0 C,
was added a
solution of bis(dichlorophosphoryl)methane (174.17 mg, 697.30 [imol, 2.5 eq.)
in PO(OEt)3
(1.5 mL). The mixture was stirred at 0 C for 5 h, followed by addition of H20
(1.5 mL) at
0 C. The mixture was stirred at 25 C overnight, and the mixture was directly
injected into the
column for purification (C18 reversed phase silica gel, 0 to 25% ACN in water
gradient eluent),
giving Compound d-5, (60 mg, 34.7% yield); 1H NMR (500 MHz, CD30D) 6 ppm 2.53
(dt,
J=41.8, 14.0 Hz, 4H), 3.02 (t, J=7.2 Hz, 2H), 4.22-4.38 (m, 3H), 4.43 (d,
J=4.4 Hz, 2H), 4.65
(s, 2H), 4.76 (s, 1H), 4.97 (s, 1H), 6.00 (d, J=4.6 Hz, 1H), 6.92 (t, J=9.3
Hz, 1H), 7.06 (d, J=7.4
Hz, 1H), 7.24 (dd, J=12.9, 7.7 Hz, 1H), 8.39 (s, 1H); m/z (ESI-1):620.1 (M-
PH).
Example 29. Synthesis of compound d-6
OH
Br C- N git
0 THF H2
Me0H j LDA NaBH4, CoCI, ip _ SO4
C Et0H 70 C 0 õis,i
NH LJH
0' 0 0¨
1-0
N
/
AGO NNCI
<34 ItLN OAc OAc
N a NH 0 0
__________________________ AGO
,Me0H H0_14 N CI _________________________________________________ HO _14 N'
CI
DIPEA, Diosane Me01-1, rt ¨ )Dc -0-
lOO'C,lEh
OAc OAc OH OH 0 CI
p-TsON
C
0 0
N-
CI CI 0 0 2].
P0(0Me)2 N CI
H20 OH ON
OH OH
[00296] A mixture of tetralin-1 -carboxylic acid (0.5 g, 2.84
mmol, 1 eq.) and H2SO4
(278.30 mg, 2.84 mmol, 151.25 uL, 1 eq.) in Me0H (10 mL) was heated to 65 C
for 20 h. The
reaction was complete as indicated by TLC. The resulting solution was
evaporated. The residue
was diluted with Et0Ac (30 mL), washed with brine, dried (Na2SO4), filtered,
and evaporated
to dryness, give methyl tetralin-1-carboxylate (470 mg, 87.1%).
140
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00297] To a mixture of methyl tetralin-l-carboxyl ate (470 mg,
2.47 mmol, 1 eq.) in THF
(6 mL) under nitrogen atmosphere was added dropwise LDA (2 M, 1.48 mL, 1.2
eq.) at -78 C
and kept at-78 C, followed by addition of 2-bromoacetonitrile (592.69 mg,
4.94 mmol, 2 eq.)
was added (-78 C). The mixture was stirred at room temperature for 4 h. The
reaction was
complete as indicated by TLC analysis. The resulting solution was quenched by
1 N HC1 (3
mL), diluted with Et0Ac (30 mL). The organic layer was washed with brine,
dried (Na2SO4),
filtered, and evaporated to dryness. The residue was purified by column
chromatograph on
silica gel eluted with PE/EA (from 100:0 to 70:30), giving methyl 1-
(cyanomethyl) tetralin- 1 -
carboxylate (490 mg, 86.5%) as a yellow solid.
[00298] To a mixture of methyl 1-(cyanomethyl) tetralin-l-
carboxylate (200 mg, 872.32
'Limo], I eq.) in Et0H (30 mL) under nitrogen atmosphere were added dropwi se
CoC12 (226.52
mg, 1.74 mmol, 2 eq.) and NaBat (330.00 mg, 8.72 mmol, 10 eq.) at 0 C. The
mixture was
stirred at room temperature overnight. The reaction was complete as indicated
by TLC. The
reaction mixture was quenched by 1 N HC1 (30 mL), extracted with Et0Ac (2 x 50
mL).
The organic layer was washed with brine, dried (Na2SO4), filtered, and
evaporated to
dryness. The residue was purified by column chromatograph on silica gel eluted
with
DCM/Me0H (from 100:0 to 90:10), giving spiro[pyrrolidine-4,1'-tetralin1-2-one
(50 mg,
28.5%) as a solid.
[00299] To a mixture of spiro [pyn-olidine-4,1'-tetra1in1-2-one
(400 mg, 1.99 mmol,
1 eq.) in THE (30 mL) was added LiA1H4 (150.85 mg, 3.97 mmol, 2.0 eq.). The
mixture was
stirred at 70 'V overnight. TLC showed that one new product was formed. The
mixture was
quenched by H20 (1 mL), followed by addition of Et0Ac (20 mL). The solid
material was
removed by filtration, and the organic layer was concentrated to a residue.
The residual material
was purified by column chromatography on silica gel eluted with DCM/Me0H
(100:0-80:20),
affording spiro[pyrrolidine-3,1'-tetralin] (230 mg, 61.8%) as oil.
1003001 To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-y1)
tetrahydrofuran-2-yll methyl acetate (500 mg, 1.12 mmol, 1 eq.), Spiro
[pyrrolidine-3,1'-
tetralin] (230.32 mg, 1.23 mmol, 1.1 eq.) in 1,4-dioxane (20 mL) was added
DIEA (361.23 mg,
2.80 mmol, 486.83 L. 2.5 eq.). The mixture was stirred at 100 C overnight,
diluted with
Et0Ac (40 mL), washed with water and brine subsequently. The organic layer was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/Et0Ac (100:0-60:40),
affording 1(2R,3R,4R,5R)-3,4-di acetoxy -5 -(2-chl o ro-6-
141
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
spiro[pyrrolidine-3,11-tetralin]-1-yl-purin-9-yOtetrahydrofuran-2-yllmethyl
acetate (600 mg,
89.7%) as yellow solid.
[00301] To a solution
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro
[pyrrolidine-3,1'-tetralinl -1-yl-purin-9-y1) tetrahydrofuran-2-yl] methyl
acetate (600 mg, 1.00
mmol, 1 eq.) in Me0H (8 mL) was added NH3-Me0H (7 M, 4.30 mL, 30 eq.). The
mixture
was stirred at room temperature overnight. TLC showed starting material was
consumed and
one product was formed. The solvent was removed by evaporation; and the
residue was diluted
with Et0Ac (50 mL), washed water (30 mL) and brine (30 mL) subsequently, dried
with
Na2SO4, and concentrated, affording (2R,3R,4S,5R)-2-(2-chloro-6-
spiro[pyrrolidine-3,1'-
tetralin1 -1 -yl-purin-9-y1)-5 -(hy droxymethy Otetrahy drofuran-3,4-di ol
(450 mg, 95.1%).
1003021
To a solution of (2R,3R,4S,5R)-2-(2-chl oro-6-spiro [pyrroli din e-3,1'-
tetralin] - I -
yl-purin-9-y1)-5-(hydroxymethyl) tetrahydrofuran-3,4-diol (450 mg, 953.52
1.tinol,
1 eq.) in Acetone (25 mL) was added 2,2-dimethoxypropane (1.49 g, 14.30 mmol,
15 eq.) and Ts0H-H20 (188.80 mg, 953.52 lamol, 1 eq.). The mixture was stirred
at room
temperature for 3 h. The solvent was removed by evaporation; and the residue
was diluted with
Et0Ac (50 mL), washed with aqueous NaHCO3, and with brine subsequently. The
organic
layer was concentrated and the residue was purified by column chromatography
on silica gel
eluted with PE/Et0Ac (100:0-50:50),
affording [(3aR,4R,6R,6aR)-4-(2-chloro-6-
spiro[pyrrolidine-3,1'-tetralin] -1-yl-purin-9-y methy1-3a,4,6,6a-tetrahy
drofu ro [3,4-
d] [1,3[ dioxo1-6-yl[methanol (400 mg, 81.9%) as white solid.
[00303]
A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro [pyrrolidine-3,1'-
tetralinl -
1-yl-purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahy drofuro 13 ,4-d] [1,3] di oxo1-
6-yl]methanol (400
mg, 781.25 pmol, 1 eq.) in PO(Me0):3 (6 mL) was cooled to ¨0 'V, followed by
addition of a
solution of bis(dichlorophosphoryl) methane (390.29 mg, 1.56 mmol, 2.0 eq.) in
PO(Me0)3 (3
mL). The mixture was stirred at ¨0 C for 4 h, followed by addition of H20 (4
mL). and stirred
at 40 C for 1 h, and then stirred at room temperature overnight. Purification
of the reaction
mixture was (C18 reversed phase silica gel, 5-30% ACN in water) gave Compound
d-6 (321
mg, 64.2%) as white solid: 1H NMR (500 MHz, CD30D) 6 ppm 1.87 (ddd, J=34.9,
18.4, 7.5
Hz, 4H), 2.11 (d, J=47.6 Hz, 1H), 2.32-2.60 (m, 3H), 2.82 (d, J=5.5 Hz, 2H),
3.86 (dd, J=60.2,
34.8 Hz, 2H), 4.11-4.46 (m, 6H), 4.60 (d, J=19.6 Hz, 1H), 6.00 (s, 1H), 7.03-
7.19 (m, 3H), 7.30
(s, 1H), 8.37 (d, J=25.3 Hz, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 21.34, 26.47,
27.52,
28.57, 31.19, 35.75, 39.69, 41.72, 44.72, 46.89, 48.04, 62.29, 63.22, 65.91,
71.32, 75.76, 84.65,
142
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
89.81, 118.81, 127.37, 130.35, 138.74, 140.07, 141.21, 152.10, 154.03, 155.58;
31P NMR (203
MHz, CD30D) 6 ppm 16.82, 19.71; m/z (ESI ): 630.18 (M+H).
Example 30. Synthesis of compound d- 7
CI
Ac0 N-- N -CI
acii>
Q 1
c N.
T)11
L1A01: 0Ac OA
Ac0 N Pr C
1,11-13-Me0H
H:D-sTT 7 N DIPEA, Dioxane
Me0H, rt
100 C, 16 h
CAD OAc
/
\ 0 0
Isr
0><0CI CI
Ho ll P 7Me)'
<N:eNIC
I
p-Ts0H HO N CI H0
CX0
OH OH CHOP
[00304] To a mixture of indolin-2-one (2 g, 15.02 mmol, 1 eq.)
in THF (30 mL) under
nitrogen atmosphere was added dropwise LiHMDS (1 M, 33.05 mL, 2.2 eq.) at -78
C, and then
it was brought to -50 C for 30 min. The mixture was cooled to -78 C and 1,5-
dibromopentane
(3.45 g, 15.02 mmol, 2.05 mL, 1 eq.) in THF (15 mL) was added. The mixture was
stirred at
room temperature for 3 h, then at reflux for 5 h. The mixture was stirred at
room temperature
for 16 h. The mixture was evaporated under reduced pressure and partitioned
between ElOAc
and saturated NH4C1.'The organic layer was concentrated and the residue was
purified by
column chromatography on silica gel eluted with (PE/Et0Ac from 100:0 to 60:40)
to
give spiro[cyclohexane-1,3'-indoline1-2'-one (1.3 g, 43.0%) as a yellow solid.
[00305] To a solution of spiro[cyclohexane-1,3'-indoline]-2'-one
(500 mg, 2.48 mmol,
1 eq.) in THF (25 mL) was added Li AlH4 (188.56 mg, 4.97 mmol, 2.0 eq.). The
mixture was
stirred at 70 C overnight. The mixture was quenched by H20 (0.3 mL), and
Et0Ac (20 mL)
was added into the mixture. The solid was removed by filtration and the
organic layer was
concentrated, the residue was purified by column chromatography on silica gel
eluted with
PE/Et0Ac (100:0-80:20) and afforded spiro[cyclohexane-1,3'-indoline] (401 mg,
86.2%) as
white solid.
[00306] To a solution of 1(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-y1)
tetrahydrofuran-2-yllmethyl acetate (450 mg, 1.01 mmol, 1 eq.) in 1,4-dioxane
(25 mL) were
added spiro[cyclohexane-1,3'-indoline] (207.29 mg, 1.11 mmol, 1.1 eq.) and
DIPEA (325.10
mg, 2.52 mmol, 438.15 !IL, 2.5 eq.). The mixture was stirred at 100 C
overnight. The mixture
was concentrated and the residue was purified by column chromatography on
silica gel eluted
143
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
with PE/Et0Ac (100:0-60:40) and afford [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-
chloro-6-
Spiro 13 a,7a-dihy dro-2H-indol e-3,11-cy cl ohexane] -1 -y 1-purin-9-y
Otetrahy drofuran-2 -
yl] methyl acetate (300 mg, 49.7% yield).
[00307]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro [3 a, 7a-
dihy dro-2H-indol e-3,1'-cy clohexane] -1-yl-purin-9-yl)tetrahy drofuran-2-yll
methyl acetate
(300 mg, 499.95 pmol, 1 eq.) in Me0H (5 mL) was added Me0H-NH3 (7 M, 2.14 mL,
30 eq.).
The mixture was stirred at room temperature for 5 h. Large amount of solid was
formed. The
solvent was removed by evaporation, followed by addition of Et0Ac (100 mL) and
H20 (70
mL). After the aqueous layer was removed, the solid formed a suspension in
organic layer. The
suspension layer was concentrated to afford (2R,3R,4S,5R)-2-(2-chloro-6-
spiro[3a,7a-
di hy dro-2H-i n dol e-3,1'-cy cl ohex an e] n -9-y1)-5 -(hy droxy methyl)
tetrahydrofuran-
3,4-diol (236 mg, 99.6%).
[00308]
To a mixture of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[3a,7a-dihydro-2H-indole-
3,11-cyclohexanel -1-yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol
(236 mg,
497.94 pmol, 1 eq.) in acetone (20 mL) were added 2,2-dimethoxypropane (1.04
g, 9.96 mmol,
20 eq.) and p-Ts0H-H20 (98.59 mg, 497.94 pmol, 1.0 eq.). The mixture was
stirred at room
temperature overnight. The solvent was removed by evaporation, and the residue
was taken
into Et0Ac (50 mL). The organic solution was washed with aqueous NaHCO3, then
with brine.
The organic layer was concentrated and the residue was purified by column
chromatography
on silica gel eluted with PE/Et0Ac (100:0-50:50), afforded [(3aR,4R,6R,6aR)-4-
(2-chloro-6-
spiro
[3a,7 a-dihy dro-2H-indol e-3,1'- cy clohexanel -1-yl-purin-9-y 1)-2,2- di
methy1-3 a,4,6,6a-
tetrahydrofuro[3,4-d] [1,3] dioxo1-6-yll methanol (254 mg, 99.2%).
[00309] A solution
of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cyclohexane-1,3'-
indoline1-1'-yl-purin-9-y1)-2,2-dirnethyl-3a,4,6,6a-tetrahydrofuro [3,4-d]
[1,3] dioxo1-6-
yllmethanol (298.82 mg, 583.64 pinol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to
¨0 C,
followed by addition of a solution of bis(dichlorophosphoryl)methane (291.57
mg, 1.17 mmol,
2.0 eq.) in PO(Me0)3 (3mL). The mixture was stirred at 0 C for 5 h. Then H20
(4 mL) was
added into the reaction mixture. The mixture was stirred at 40 C for 40 min,
and then stirred
at room temperature overnight. The reaction mixture was purified via C18
reversed phase silica
gel (0-25% ACN in Water), giving Compound d-7, (301 mg, 80.7%): 1H NMR (500
MHz,
CD30D) 6 ppm 1.31-1.81 (m, 10H), 2.52 (t, J=20.9 Hz, 2H), 4.27 (s, 1H), 4.30-
4.42 (m, 2H),
4.45 (t, J=4.8 Hz, 1H), 4.53-4.61 (m, 2H), 4.66 (t, J=4.9 Hz, 1H), 6.05 (d,
J=4.7 Hz, 1H), 7.04
(t, J=7.3 Hz, 1H), 7.12-7.26 (m, 2H), 8.41-8.49 (m, 2H); 13C NMR (125 MHz,
CD30D) 6 ppm
144
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
24.12, 26.53, 27.51, 38.39, 45.84, 62.14, 66.16, 71.48, 75.72, 84.56, 89.72,
118.98, 120.30,
123.56, 128.51, 140.84, 142.88, 143.18, 152.54, 153.35, 154.56;31P NMR (203
MHz, CD30D)
6 ppm 16.61, 19.95; m/z (ESI11): 630.27 (M +H).
Example 31. Synthesis of compound d-8
_
0 - Br C- 1101...., CoCl2
LiAll 14 ),fl
0 HM2eS0 H4 j ' . - . 0 L D A -
-1- 70 C K_iI-1
0 Et0H, NaBH4
0_ \GEN NH
NH
OH 0
z
1 \
/
Ac0 N N CI ) CN
OAc OAc N-7 N-1/1,t3-N NH3-
Me0H N- ---1--,
_________________________ 33-
DIPEA, Dioxane Me0H, rtHO 0
N-- 'le"CI N- 'N''' 'CI
100 C, 16 h Ac0¨k_._-0,_ ¨I0_
HO ¨;-'N CI
p-Ts0H
OAc OAc OH OH 0^,
,,0
C0 0 I
CI¨P P-CI ,(
, \
CI CI
3.-
P0(0Me)3 N
1--,
H200 0 N,---,..N
,1= P
N
HO -
OH OHm _
¨I N CI ()-
OH OH
[00310] Methyl indane-1-carboxylate was prepared in the same way
as described in
Compound d-1 section.
[00311] To a mixture of methyl indane-1-carboxylate (2.1 g,
11.92 mrriol, 1 eq.) in THF
(25 mL) under nitrogen atmosphere was added dropwise LDA (2 M; 7.15 mL; 1.2
eq.) at -78
C, and 20 min later, a solution of 2-bromoacetonitrile (2.86 g, 23.84 mmol, 2
eq.) in THF (1
mL) was added to the reaction mixture at -78 C. The resulting mixture was
stirred at room
temperature for 4 h, quenched with 1 N HC1 (30 mL) and followed by addition of
Et0Ac (60
mL). The organic layer was washed with brine, dried (Na2SO4), filtered, and
evaporated to
dryness. The residue was purified by column chromatograph on silica gel,
eluted with
(PE/Et0Ac from 100:0 to 70:30), giving methyl 1-(cyanomethyl)indane-1-
carboxylate (2.4 g,
93.5%) as a yellow oil.
[00312] To a mixture of methyl 1-(cyanomethypindane-1 -
carboxylate (2.4 g, 11.15
mmol, 1 eq.) in Et0H (150 mL) under nitrogen atmosphere were added dropwise
CoC12 (2.90
g, 22.30 mmol, 2 eq.) and NaBH4 (4.22 g, 111.50 mmol, 10 eq.) while the
temperature was
145
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
maintained at 0 C. Then the reaction mixture was stirred at room temperature
overnight,
quenched with 1 N HC1 (150 mL), extracted with Et0Ac (2 x 200 mL). The organic
layer was
washed with brine, dried (Na2SO4), filtered, and evaporated to dryness. The
residue
was purified by column chromatograph on silica gel eluted with DCM/Me0H (100:0
to 95:5),
to giving spiro[indane-1,3'-pyrrolidine1-2'-one (1.05 g, 50.3%) as a yellow
solid.
[00313] To a solution of spiro[indane-1,3'-pyn-o1idine1-2'-one
(1 g, 5.34 mmol,
1 eq.) in THF (40 mL) was added LiA1H4 (405.37 mg, 10.68 mmol, 2.0 eq.). The
mixture was
stirred at 70 C overnight, quenched with H20 (1 mL), followed by addition of
15% NaOH
aqueous solution (1 mL), H20 (1 mL), and Et0Ac (30 mL). The solid was removed
by filtration
and the organic layer was concentrated to afford crude spiro[indane-1,3'-
pyrro1idine] (800 mg,
86.5%) as oil.
[00314] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yptetrahydrofuran-2-ylimethyl acetate (500 mg, 1.12 mmol, 1 eq.) in 1,4-
dioxane (25
mL) were added spiro[indane-1,3'-pyrrolidine] (232.44 mg, 1.34 mmol, 1.2 eq.)
and DIPEA
(361.23 mg, 2.80 mmol, 486.83 ILIL, 2.5 eq.). The mixture was stirred at 100
C for 2 h, and
then concentrated to a residue. The residual material was purified by column
chromatography
on silica gel, eluted with PE/Et0Ac (100:0-60:40), afforded [(2R,3R,4R,5R)-3,4-
diacetoxy-5-
(2-chloro-6-spiro[indane-1,31-pyrrolidine1-11-yl-purin-9-yOtetrahydrofuran-2-
yl]methyl
acetate (500 mg, 76.6%) as white solid.
[00315] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-
chloro-6-spiro[indane-1,3'-
pyn-olidine1-1'-yl-purin-9-yetetrahydrofuran-2-yllmethyl acetate (500 mg,
856.14 !Amok
1 eq.) in Me0H (6 mL) was added NH3-Me0H (7 M, 3.67 mL, 30 eq.). The mixture
was stirred
at room temperature for 4 h. The solvent was removed by evaporation, and the
residue was
taken into Et0Ac (100 mL), followed by addition of H20 (70 mL). The organic
layer was
separated, washed (brine), dried (Na2SO4), and concentrated, affording the
(2R,3R,4S,5R)-2-
(2-chloro-6-spiro[indane-1,3'-pyrrolidinel-1'-yl-purin-9-y1)-5-
(hy dro xy me thyl) te trahy drofuran-3,4-di ol (390 mg, 99.5%) as white
solid.
[00316] To a solution of (2R,3R,4S,5R)-2-(2-chloro-6-
spiro[indane-1,3'-pyrrolidinel-1'-
yl-purin-9-y1)-5-(hydroxymethyptetrahydrofuran-3,4-diol (390 mg, 851.70 wnol,
1 eq.) in acetone (20 mL) were added 2,2-dimethoxypropane (1.33 g, 12.78 mmol,
15 eq.) and p-Ts0H (146.66 mg, 851.70 [imol, 1 eq.). The mixture was stirred
at room
temperature overnight. The solvent was removed by evaporation. Tthe residue
was diluted with
Et0Ac (50 mL), washed by aqueous Naf1CO3, then with brine. The organic layer
was
146
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
concentrated and the residue was purified by column chromatography on silica
gel (PE/Et0Ac,
100:0-50:50), afforded [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro [indane-1,3'-
pyrrolidinel -1'-yl-
purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3]dioxo1-6-
yllmethanol (410 mg,
96.7%) as white solid.
1003171 To a solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-
spiro[indane-1,3'-pyrrolidinel-
V-yl-purin-9-y1)-2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-d][1,31dioxol-6-
yllmethanol (410
mg, 823.34 Lama 1 eq.) in PO(Me0)3 (5 mL), cooled to ¨0 C, was added a
solution
of bis(dichlorophosphoryOmethane (411.32 mg, 1.65 mmol, 2.0 eq.) in PO(Me0)3
(4 mL). The
mixture was stirred at 0 C for 5 h, followed by addition of H20 (6 mL). The
mixture was
stirred at 40 'V for 1 h and then stirred at room temperature overnight.
Purification of the
reaction mixture using C18 reversed phase silica gel (0-25% ACN in water)
provided
Compound d-8, (301 mg, 58.4%): NMR (500 MHz, CD30D) 6 ppm 1.96-2.35 (m, 4H),
2.44
(dd, J=37.0, 20.5 Hz, 2H), 2.94 (d, J=6.7 Hz, 2H), 3.64-4.19 (m, 3H), 4.21-
4.45 (m, 5H), 4.56
(d, J=20.9 Hz, 1H), 5.96 (s, 1H), 7.12-7.29 (m, 4H), 8.38 (d, J=33.4 Hz, 1H);
1-3C NMR (125
MHz, CD30D) 6 ppm 26.44, 27.50, 28.55, 31.11, 37.22, 38.81, 39.17, 53.92,
56.03, 59.67,
60.67, 65.91, 71.32, 75.73, 84.61, 89.81, 118.82, 123.32, 125.72, 127.84,
128.50, 140.10,
144.97, 147.27, 152.06, 154.06, 155.63; 31-13 NMR (203 MHz, CD30D) 6 ppm
16.96, 19.61;
m/z (ESL): 616.32 (M +H).
Example 32. Synthesis of compound d-9
NC 10
LHMDS
I n-BuLl 111 Pd/C,H,
HCI-EA
IP
)S" Br THF,-78 C,3h N40* THF,-78 C,2h CH3OH/CH3000H
rt,16h
NC n 3h
NH
CI
N
Ac0
OAc OAc NCTS NI-1,-Me0H N p-Ts0 -I F1,0 N
Co17 VCI
DIPEoxneN Me0H, rt,3h F111->t N acetone,t3h eNI-N.a,oi 1)
P0(0Me),, 0"C,5h
100 C, 16 hAce
N N*I'Cl N 2/1-rleeN0
C,40mIn OH OH ¨4?I
¨y24 OH OH
CA, OAc OH OH
[00318] A solution of tert-butyl 3-cyanoazetidine-1-carboxylate
(3.0 g, 16.46 mmol, 1
eq.) in 'THE (30 mL) was cooled to -78 C, followed by addition of LiHMDS (1
M, 20.58 mL,
147
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1.25 eq) . The mixture was stirred for 20 min., followed by addition of a
solution of 1-
(bromomethyl)-2-iodo-benzene (5.13 g, 17.29 mmol, 1.05 eq.) in THF (3 mL). The
mixture
was stirred at -78 C for 3 h, quenched with saturated NH4C1, and extracted
with Et0Ac (50
mL * 2). The organic layer was washed with brine, and concentrated. The
residue was
purified by column chromatographyony silica gel, eluted with PE/EA (100:0-
83:17),
affording t ert-b uty I 3-cy ano-3-[(2-iodophenyl)methyl] azeti dine-1 -
carboxy I ate (6.44 g, 16.17
mmol, 98.22% yield) as yellow oil.
[00319]
A solution of tert-butyl 3-cyano-3-[(2-iodophenyl)methyl]azetidine-1-
carboxylate (6.44 g, 16.17 mmol, 1 eq.) in THF (60 mL) was cooled to -78 C,
followed by
addition of n-BuLi (2.5 M, 12.94 mL, 2 eq.) dropwise at -78 'C. The mixture
was stirred at -
78 C for 2 h. TLC showed that starting material was consumed completely. The
mixture was
quenched with saturated NH4C1 and extracted with Et0Ac (75 mL * 2). The
organic layer was
washed with brine, and concentrated. The residue was purified by column
chromatographyon
silica gel, eluted with PE/EA (100:0-83:17), affording tert-butyl 1'-
oxospiro[azetidine-3,21-
indane1-1-carboxylate (3.4 g, 12.44 mmol, 76.92% yield) as light-yellow solid.
[00320]
To a solution of tert-butyl 1'-oxospiro[azetidine-3,2'-indaneJ-1-
carboxylate (1.0
g, 3.66 mmol, 1 eq.) in CH3OH/CH3COOH (5m1 +10 mL) was added Pd/C (100 mg, 10%
purity). The mixture was stirred at under H2 gas condition at rt overnight.
TLC showed that
starting material was mostly consumed. The mixture was filtered, washed with
Me0H. The
filtrate was concentrated to remove most of the solvent. Then the mixture was
neutralized with
saturated NaHCO3 (aq.) and extracted with Et0Ac (15 mL * 2). The organic layer
was washed
with brine, and concentrated. The residue was purified by column
chromatographyon silica gel,
eluted with PE/EA (100:0 to 90:10), giving tert-butyl spiro[azetidine-3,2'-
indaneJ-1-
carboxylate (360 mg, 1.39 mmol, 37.94% yield) as white solid.
[00321]
tert-butyl spiro[azetidine-3,2'-indane]-1-carboxylate (370 mg, 1.43 mmol,
1 eq.)
was dissolved in HC1-EA (5 mL), and the mixture was stirred at rt for 3 h. The
mixture was
concentrated and was used for the next step.
[00322] To
a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yptetrahydrofuran-2-ylimethyl acetate (523.74 mg, 1.17 mmol, 1 eq.) in 1,4-
dioxane (25
mL) were added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yl)tetrahydrofuran-2-
yllmethyl acetate (523.74 mg, 1.17 mmol, 1 eq.) and DIP EA (529.73 mg, 4.10
mmol, 713.92
uL, 3.5 eq.) . The mixture was stirred at 100 C overnight. TLC showed
starting material was
consumed. Solvent was removed by evaporation. The residue was taken into Et0Ac
(40 mL),
148
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
and the solution was washed with water, then with brine. The organic layer was
concentrated
and the residue was purified by column chromatography on silica gel eluted
with PE/EA
(100:0-65:35), affording [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-
spiro[azetidine-3,2'-
indane1-1-yl-purin-9-yOtetrahydrofuran-2-yllmethyl acetate (620 mg, 1.09 mmol,
92.88%
yield) as a yellow solid.
[00323]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro[azetidine-
3,2'-indane1-1-yl-purin-9-yptetrahydrofuran-2-yllmethyl acetate (620 mg, 1.09
mmol, 1 eq.)
in Me0H (6 mL) was added NH3-Me0H (7 M, 4.66 mL, 30 eq.). The mixture was
stirred at
room temperature for 3 h. LC-MS showed the starting material disappeared.
Solvent was
removed by evaporation, and the residue was taken into ethyl acetate (30 mL)
and H20 (50
mL). The organic layer was separated, washed with brine, and dried with
Na2SO4, and
concentrated, affording (2R,3R,4S,5R)-2-(2-chloro-6-spiro[azeti dine-3,2'-
indanel -1 -y 1-purin-
9-y1)-5-(hy droxy methy 1)tetrahy drofuran-3,4- di ol (459 mg, 1.03 mmol,
95.07% yield) as a
white solid.
[00324]
To a solution of (2R,3R,4S,5R)-2-(2-ch1oro-6-spiro[azetidine-3,2'-indane1-
1-yl-
purin-9-y1)-5-(hydroxymethyl) tetrahydrofuran-3,4-diol (459 mg, 1.03 mmol, 1
eq.) in acetone
(30 mL) were added p-Ts0H-H20 (197.76 mg, 1.03 mmol, 1 eq.) and 2,2-
dimethoxypropane
(2.15 g, 20.68 mmol, 20 eq.). The mixture was stirred at rt over a weekend.
Solvent was
removed by evaporation, and the residue was taken into ethyl acetate (50 mL),
washed by
aqueous NaHCO3, washed with brine. The organic layer was concentrated and the
residue
was purified by column chromatographyon silica gel (eluted with PE/EA, 100:0-
85:15),
affording [(3aR,4R,6R,6aR)-4-(2-chl oro-6-spiro [azeti dine-3,2'-indanel -1 -
yl-purin-9-y1)-2,2-
dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]dioxo1-6-yl]methanol (350 mg,
723.22 tumol,
69.94% yield) as white solid.
[00325]
A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[azetidine-3,2'-indane] -
1-
yl-purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahy drofuro [3 ,4-d] [1,3] di oxo1-6-
yl] methanol
(348.55 mg, 720.22 [tmol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to 0 C,
followed by addition
of bis(dichlorophosphoryl)methane (359.80 mg, 1.44 mmol, 2 eq.) in PO(Me0)3 (
4 mL). The
mixture was stirred at 0 C for 5 h. LC-MS showed that the starting material
was consumed.
H20 (7 mL) was added dropwise into the mixture, and the mixture was stirred at
40 C for 40
min, and then stirred at rt overnight. Purification of the reaction mixture
with C18 reversed
phase silica gel (0-25% ACN in Water) gave Compound d-9, (260 mg, 58.63%): 1-H
NMR
(500 MHz, Me0D) 6 ppm 2.48 (t, J = 20.9 Hz, 2H), 3.26 (s, 4H), 4.24 (s, 2H),
4.38 ¨ 4.26 (m,
149
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
3H), 4.42 (t, J = 4.7 Hz, 1H), 4.55 (s, 1H), 4.63 (t, J = 4.9 Hz, 1H), 6.00
(d, J = 4.8 Hz, 1H),
7.15 (dd, J = 5.1, 3.2 Hz, 2H), 7.25 -7.20 (m, 2H), 8.39 (s, 1H); 31P NMR (203
MHz, Me0D)
6 ppm 16.85, 19.68. 13C NMR (126 MHz, Me0D) 6 ppm 26.17, 43.38, 44.10, 61.81,
63.68,
64.53, 70.02, 74.35, 83.39, 88.33, 116.93, 124.21, 126.49, 139.71, 141.29,
150.27, 154.05,
154.37. m/z (ESL): 602.31 (M +H).
Example 33. Synthesis of compound d-10
NC
LIN
NBS r )C
____________________________________________ H-BuD I N'2He 4
EteSIIP
Fa-- 0014 THI-H777cS3h I DIS-1-,,21, Chl 1-1
( D7MTC`)Ic ish
CNI1
CI
r1)
õ
Ac0
`71 0 0
GAO GAO N Nh,.MeOh1 N _ 11.1, CI'
P\cr
DIPEA [hexane Pft-- Me0H, H a NON 0 1) PO(Ohle6 0
- C,5h
100 C 16 h.
'CI Hp rr e p-Ta0H -
Ho CI 2) %/,,,,47mI ,40n HO 6H 6H N CI
1,44
GAO GAO OH OH
X OH
OH
[00326] To a mixture of 4-fluoro-2-iodo-1-methyl-benzene (5 g,
21.18 mmol, 1 eq.) in
CC14 (50 mL) was added NBS (5.66 g, 31.78 mmol, 1.5 eq.). The mixture was
stirred at reflux
for 5 h. Solvent was removed by evaporation, and the residue was purified by
column
chromatography on silica gel eluted with PE/EA (100:0-99:1), affording 1-
(bromomethyl)-4-
fluoro-2-iodo-benzene (2.55 g, 8.10 mmol, 38.22% yield).
1003271 To a solution of tert-butyl 3-cyanoazetidine-1-
carboxylate (1.34g. 7.35 mmol, 1
eq.) in THF (20 mL), cooled at -78 C, was added LiHMDS (1 M, 9.19 mL, 1.25
eq.) dropwise.
The mixture was stirred at this temperature for 20 mm, followed by addition of
1-
(bromomethyl)-4-fluoro-2-iodo-benzene (2.55 g, 8.09 mmol, 1.1 eq., dissolved
in 5 mL of
THF). The mixture was stirred at -78 C for 3 h, and the reaction was quenched
with saturated
NH4C1. The reaction mixture was extracted with Et0Ac (2 x 25 mL). The organic
layer was
washed with brine and then concentrated. The residue was purified by column
chromatographyon silica gel (eluent: PE/EA, 100:0-75:25), affording tert-butyl
3-cyano-3-[(4-
fluoro-2-iodo-phenyl)methyllazetidine-1-carboxylate (2.41g, 5.79 mmol, 78.74%
yield).
[00328] To a solution of tert-butyl 3-cyano-34(4-fluoro-2-iodo-
phenyl)methyllazetidine-
1-carboxylate (2.41 g, 5.79 mmol, 1 eq.) in THF (25 mL), cooled at -78 C,was
added n-BuLi
(2.5 M, 4.63 mL, 2 eq.) dropwise. The mixture was stirred for 2 h at this
temperature, and the
reaction was quenched by saturated NH4C1. The mixture was extracted with Et0Ac
(2 x 100
mL). The organic layer was washed by brine, and concentrated. The residue was
purified by
150
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
column chromatography on silica gel eluted with PE/FA (100:0-70:30), affording
tert-butyl 61-
fluoro-11-oxo-spiro[azetidine-3,2'-indane1-1-carboxylate (1.21 g, 4.15 mmol,
71.74% yield).
[00329]
To a solution of tert-butyl 6'-fluoro-1'-oxo-spiro[azetidine-3,2'-indane1-
1-
carboxylate (1.21 g, 4.15 mmol, 1 eq.) in Me0H (5 mL) was added NaBH4 (392.82
mg, 10.38
mmol, 2.5 eq.) in portions at 0 'C. The mixture was stirred at the temperature
for 2 h. Solvent
was removed by evaporation. The residue was extracted with Et0Ac (2 x 100 mL),
washed
with brine (100 mL), and the organic layer was concentrated to dryness,
affording tert-butyl 6'-
fluoro-1'-hy droxy -s piro [azeti dine-3,2'-indane] -1-carboxyl ate (1.21 g,
4.13 mmol, 99.31%
yield) as white solid.
[00330]
To a mixture of tert-butyl 6'-fluoro-1'-hydroxy-spiro[azetidine-3,2'-
indane1-1-
carboxylate (305 mg, 1.04 mmol, 1 eq.) in DCM (10 mL) were added BF3- Et20 (4
mL) and
EhSiH (1.21 g, 10.40 mmol, 10 eq.) at -40 C. The mixture was stirred at 40 C
for 16 h, and
the reaction was quenched with water (20 mL) and NH4C1 (20 mL). pH of the
mixture was
adjusted to 10 with 15% NaOH. The mixture was extracted with DCM (2 x 60 mL).
The
combined organic layers were washed with brine (100 mL), dried with Na2SO4,
filtered, and
evaporated, giving 5'-fluorospiro[azetidine-3,2'-indane] (100 mg, 394.99 umol,
37.99% yield,
70% purity).
[00331]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (252 mg, 563.47umo1, leq.) in 1,4-
dioxane (5 mL) was
added 5'-fluorospiro[azetidine-3,2'-indane] (99.86 mg, 563.47 umol, 1 eq.) and
DIPEA (182.06
mg, 1.41 mmol, 245.36 uL, 2.5 eq.). The mixture was stirred at 100 C for 3 h.
The mixture
was concentrated and the residue was purified by column chromatographyon
silica gel eluted
with PE/EA (100:0-58:42) and afford [(2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-
(5'-
fluorospiro[azetidine-3,2'-indane1-1-yl)purin-9-ylltetrahydrofuran-2-ylimethyl
acetate (200
mg, 136.06 umol, 24.15% yield, 40% purity)
1003321 To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(5'-
fluorospiro[azetidine-3,2'-indane1-1-yl)purin-9-yll te trahy drofuran-2-yll me
thy 1 acetate
(200.00 mg, 136.06 pmol, 1 eq.) in Me0H (5 mL) was added Me0H-NH3 (7 M, 485.92
pt, 25
eq.) .The mixture was stirred at room temperature for 2 h. The residue was
purified by column
chromatography on silica gel eluted with (DCM/Me0H from 100:0 to 93:7), giving
(2R,3R,4S,5R)-242-chloro-6-(5'-fluorospiro [azetidine-3,2'-indane1-1 -yl)purin-
9-y11-5-
(hy droxy methyptetrahy drofuran -3,4-di ol (60 mg, 129.91 umol, 95.48% yield)
151
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00333] To a solution of (2R,3R,4S,5R)-242-chloro-6-(5'-
fluorospiro[azetidine-3,21-
indane1-1-yl)purin-9-y11-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (60 mg,
129.91 pmol, 1
eq.) in acetone (10 mL) were added p-Ts0H (22.37 mg, 129.91 mnol, 1 eq.) and
2,2-
dimethoxypropane (270.59 mg, 2.60 mmol, 20 eq.). The mixture was stirred at rt
for 1 h.
Solvent was removed by evaporation. The residue was diluted with EA (50 mL),
washed with
aqueous NaHCO3 and brine successively, and concentrated. The residue was
purified by
column chromatography on silica gel eluted with PE/EA (100:0-55:45) to afford
[(3aR,4R,6R,6aR)-4- [2-chl oro-6-(5'-fluorospiro [azetidine-3,2'-indane] -1-
yl)purin-9-yl] -2,2-
dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d1 11,31dioxo1-6-yllmethanol (60 mg,
119.54 mnol,
92.02% yield) .
1003341 To a solution off(3aR,4R,6R,6aR)-4-1-2-chloro-6-(5'-
fluorospiro[azetidine-3,2'-
indane] -1-yl)purin-9-yl] -2,2-dimethy1-3a,4,6,6a-tetrahy drofuro [3,4-d]
[1,3] di oxo1-6-
yl]methanol (60 mg, 119.54 1.imol, 1 eq) in PO(Et0)3 (2 mL), cooled at 0 C,
was added a
solution of bis(dichlorophosphoryl)methane (74.65 mg, 298.84 mmol, 2.5 eq.) in
PO(Et0)3 ( 2
mL). The mixture was stirred at 0 C for 4 h, followed by addition of H20 (3
mL). The mixture
was stirred at room temperature overnight. The reaction mixture was purified
using C18
reversed phase silica gel (gradient eluent, 0 to 25% ACN in water), giving
Compound d-10,
(7.1 mg, 9.58% yield); 1HNMR (500 MHz, Me0D) 6 ppm 2.47 (t, J= 20.8 Hz, 2H),
3.25 (d,
J = 19.8 Hz, 4H), 4.19 ¨ 4.37 (m, 5H), 4.42 (d, J = 8.2 Hz, 1H), 4.49 (d, J=
58.8 Hz, 2H), 4.64
(s, 1H), 6.00 (d, J= 4.8 Hz, 1H), 6.88 (t, J = 8.7 Hz, 1H), 6.97 (d, J 8.2 Hz,
1H), 7.14 ¨ 7.27
(m, 1H), 8.40 (s, 1H); m/z (ESL): 620.3(M+H).
Example 34. Synthesis of compound d-11
13F3 Et20
Br _____________________________________________________
Nal3Hq tt311-1
CH3OH L-t=
CH,GOOH
I NC
CI
AcONXNCI
Ox0
p-11:0H H 31--01
0, 0,
CAPE., Dioxena Me0H <Y.:1;1,c j
P7,7a eN:_eX
Ac0N UIHO "N UI 61-1
OH OH
152
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00335] To a mixture of 1-fluoro-3-iodo-2-methyl-benzene (5.00
g, 21.18 mmol, 1 eq.)
in CC14 (80 mL) were added NBS (5.66 g, 31.78 mmol, 1.5 eq.) and dibenzoyl
peroxide (256.57
mg, 1.06 mmol, 0.05 eq.). The mixture was reflux for 5 h. Solvent was removed
by evaporation.
The residue was extracted with DCM (2 x 50 mL). The organic layer was washed
with brine,
dried (Na2SO4), filtered, and concentrated. The residue was purified by column
chromatography on silica gel eluted with (PE/EA from 100:0 to 99:1), affording
2-
(bromomethyl)-1-fluoro-3-iodo-benzene (1.97 g, 6.26 mmol, 29.53% yield).
[00336] To a solution of tert-butyl 3-cyanoazetidine-l-
carboxylate (1.10 g, 6.05 mmol, 1
eq.) in THF (10 mL), cooled at -78 C, was added LiHMDS (1 M, 7.56 mL, 1.25
eq.) dropwise.
The mixture was stirred at -78 "V for 20 min, followed by addition of a
solution of 2-
(bromomethyl)- I -fluoro-3-iodo-benzene (2.0 g, 6.35 mmol, 1.05 eq.) in THF
(10 mL). The
mixture was stirred at -78 C for 3 h, and the reaction was quenched with
saturated NH4CI.
Extraction of the mixture was done using Et0Ac (2 x 50 mL). The combined
organic layers
were washed with brine, dried (Na2SO4), filtered, and concentrated. The
residue was
purified by column chromatography on silica gel eluted with (PE/EA from 100:0
to 80:20),
affording tert-butyl 3-cyano-3-[(2-fluoro-6-iodo-phenyl)methylJazetidine-1-
carboxylate (1.92
g, 4.61 mmol, 76.27% yield) as yellow oil.
[00337] To a solution of tert-butyl 3-cyano-3-[(2-fluoro-6-iodo-
phenyl)methyllazetidine-
1-carboxylate (1.92 g, 4.61 mmol, 1 eq.) in THF (30 mL), cooled at -78 C, was
added n-BuLi
(2.5 M, 3.69 mL, 2 eq.) dropvvise. The mixture was stirred at -78 C for 3 h,
and the reaction
was quenched with saturated NH4C1. Extraction of the mixture was performed
with Et0Ac (2
x 30 mL). The combined organic layers were washed with brine, dried (Na2SO4),
filtered, and
concentrated. The residue was dissolved in 1:1 mixture of THF and water (20
mL) and stirred
at room temperature for 4 days, followed by extracting the mixture was
extracted with Et0Ac
(2 x 30 mL). The organic layers were combined, washed with brine, dried
(Na2SO4), filtered,
and concentrated. The residue was purified by column chromatography on silica
gel eluted with
PE/EA (100:0 to 80:20), affording tert-butyl 4'-fluoro-V-oxo-spiro[azetidine-
3,2'-indane1-1-
carboxylate (430 mg, 1.48 mmol, 32.00% yield).
[00338] To a solution of tert-butyl 4'-fluoro-1'-oxo-spiro
[azetidine-3,2'-indane1-1-
carboxylate (430 mg, 1.48 mmol, 1 eq.) in CH3OH (10 mL) was added NaBH4
(167.52 mg,
4.43 mmol, 3 eq.) at 0 C. The mixture was stirred at the temperature for 2 h.
Solvent was
removed by evaporation, and the residue was extracted with Et0Ac (2 x 30 mL).
The combined
organic layer was washed with brine, dried (Na2SO4), filtered, and
concentrated, giving tert-
153
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
butyl 4'-fluoro-11-hydroxy-spiro[azetidine-3,2'-indane1-1-carboxylate (410 mg,
1.40 mmol,
94.69% yield) as white solid.
1003391
To a mixture of tert-butyl 4'-fluoro-1'-hydroxy-spiro[azetidine-3,2'-
indane]-1-
carboxylate (410 mg, 1.40 mmol. 1 eq.) in CH3COOH (10 mL) were added SF3.E120
(2.00
mL) and Et3SiH (1.30 g, 11.18 mmol, 1.79 mL, 8 eq.). The mixture was stirred
at 60 C for 16
h, and then cooled to 0 C, followed by slow addition of 15% NaOH (aqueous)
until the pH of
the mixture reached 9. The resultant mixture was extracted with Et0Ac (3 x 30
mL).
The organic layers were combined, washed with NH4C1(aq.) and with brine
successively, dried
(Na2SO4), filtered, and concentrated to dryness, affording 4'-
fluorospiro[azetidine-3,21-indane]
(245 mg, 1.38 mmol, 98.91% yield).
1003401
To a mixture of 4'-fluorospiro[azetidine-3,2'-indane] (72 mg, 406.28 umol,
1.2
eq.) in 1,4-dioxane (10 mL) were added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-
9-yl)tetrahydrofuran-2-yl[methyl acetate (151.42 mg, 338.57 timol, 1 eq.) and
DIPEA (109.39
mg, 846.42 timol, 147.43 tiL, 2.5 eq.). The mixture was stirred at 100 C
overnight. Solvent
was removed by evaporation. The residue was diluted with Et0Ac (50 mL), washed
with brine,
dried (Na2SO4), filtered, and concentrated. The residue was purified by column
chromatography on silica gel eluted with PE/EA (100:0 to 50:50), affording
[(2R,3R,4R,5R)-
3,4-di acetoxy-5 -[2-chl oro-6-(4'-fluoro spi ro [azeti dine-3 ,2'-indane] -1-
yl)purin-9-
yfltetrahydrofuran-2-y11 methyl acetate (100 mg, 170.07 wnol, 50.23% yield) as
white solid.
[00341] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(4'-
fluorospiro[azetidine-3,2'-indane1-1-yl)purin-9-yfltetrahydrofuran-2-yflmethyl
acetate (100
mg, 170.07 tunol, 1 eq.) in Me0H (2.00 mL) was added NH3-Me0H (7 M, 728.88
!AL, 30 eq.).
The mixture was stirred at room temperature for 2 h. Solvent was removed by
evaporation, and
the residue was extracted with Et0Ac (2 x 20 mL). The organic layers were
combined, washed
with brine, dried (Na2SO4), filtered, and concentrated to dryness, affording
(2R,3R,4S,5R)-2-
[2-chloro-6-(4'-fluorospiro [azeti dine-3 ,2'-indanel -1 -yl)purin-9-yl] -5-
(hy dro xy me thyl) te trahy drofuran-3,4-di ol (78 mg, 168.88 mol, 99.30%
yield) as white solid.
[00342]
To a solution of (2R,3R,4S,5R)-242-chloro-6-(4'-fluorospiro[azetidine-3,2'-
indane1-1-yl)purin-9-y11-5-(hydroxymethyptetrahydrofuran-3,4-diol (78 mg,
168.88 timol, 1
eq.) in acetone (10 mL) was added p-Ts0H-H20 (32.12 mg, 168.88 ttmol, 1 eq.),
2,2-
dimethoxypropane (351.76 mg, 3.38 mmol, 415.30 tiL, 20 eq.). The mixture was
stirred at
room temperature overnight. The mixture was pH-adjusted 9 by slow addition of
aqueous
NaHCO3 slowly at 0 'C. Solvent was removed by evaporation, and the residue was
extracted
154
CA 03200232 2023- 5- 25

WO 2022/109741 PCT/CA2021/051688
with Et0Ac (2 x 30 mL). The organic layers were combined, washed with brine,
dried
(Na2SO4), filtered, and concentrated. The residue was purified by column
chromatography on
silica gel eluted with PE/EA (100:0 to 50:50), affording [(3aR,4R,6R,6aR)-442-
chloro-6-(4'-
fluorospiro[azetidine-3,2'-indane1-1-yl)purin-9-y11-2,2-dimethyl-3a,4,6,6a-
tetrahydrofuro[3,4-
d][1,31dioxol-6-yl]methanol (57 mg, 113.56 umol, 67.24% yield) as white solid.
[00343] To a solution
of 1(3aR,4R,6R,6aR)-442-chloro-6-(4'-fluorospiro[azetidine-3,2'-
indane1-1-y1)purin-9-y11-2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-
d]11,31dioxol-6-
yl[methanol (57 mg, 113.56 umol, 1 eq.) in P0(0E03 (1 mL), cooled at 0 C, was
added a
solution of bis(dichlorophosphoryl)methane (70.91 mg, 283.90 umol, 2.5 eq.) in
PO(OEt)3 (1
mL). The mixture was stirred at 0 C for 4 h, followed by slow addition of
water (1.5 mL) at 0
C. The mixture was stirred at 40 C for 40 min, and then at 25 C overnight.
The reaction
mixture was purified using C18 reversed phase silica gel (gradient eluent, 0
to 25% ACN in
water), giving Compound d-11, (10 mg, 16.13 umol, 14.25% yield); 11-1 NMR (500
MHz,
Me0D) 6 ppm 2.53 (t, J = 20.6 Hz, 2H), 3.35 (s, 4H), 4.25 ¨ 4.41 (m, 5H), 4.45
(s, 1H), 4.60
(s, 2H), 4.67 (s, 1H), 6.04 (d, J = 4.0 Hz, 1H), 6.91 (t, J = 8.5 Hz, 1H),
7.09 (d, J = 7.1 Hz, 1H),
7.22 (d, J = 5.4 Hz, 1H), 8.47 (s, 1H); m/z (ESL): 620.1(M+H).
Example 35. Synthesis of compound d-12
7-'N1-12
HCHO I
H2SO4 , K2CO3 ," EDCI, HOBt , .. DEAD,PPh3
r r-
Me0H DMSO DIPEA, DMF --,, THF
\ OH 'OH
0
HO 0 0 0 HO 0 40 vi 0 N
I
CI
0
--
4\1 -alt,N_L
--. 1
Ac0 N 'N' 'CI
AICI3 Pd(OH)2 OAc OAc NN NH3-
Me0H
LiAIH4 L H2, Me0H DIPEA, Dioxane
Ac0 N N- CI Me 1-1' HTHF 0 HCOONH4 N
H 100"C, 16 h ¨112_,?1
OAc OAc
/ -
1
\
N I I N 0 0 N
CI¨P"¨'P,-CI N-
_),,,
lj
_4 HO¨ 1\1 ______ ..- I .,,
0 HO N CI P0(0Me)3 N--
l',,11'-0 .. N .. N' 'CI
4 ",_LCI - ¨
A 104 H20 HO" 1 - 1
OH OH ¨Isi----r-
p-Ts0H
OH OH (:) ,7\0
OH OH
155
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00344] A mixture of tetralin-l-carboxylic acid (2 g, 11.35
mmol, 1 eq.) and H2SO4 (1.11
g, 11.35 mmol, 605.00 nL, 1 eq.) in Me0H (20 mL) was heated at 65 C for 20 h.
The resulting
solution was concentrated. The residue was dissolved in Et0Ac (50 mL), and the
solution was
washed with brine, dried (Na2SO4), filtered, and concentrated to dryness,
giving methyl
tetralin-l-carboxylate (2.1 g, 97.2%).
1003451 To a mixture of methyl tetralin-1-carboxylate (2.1 g,
11.04 mmol, 1 eq.) and
in DMSO (25 mL) under nitrogen atmosphere were added K2CO3 (5.03 g, 36.43
mmol,
3.3 eq.) and HCHO (3.05 g, 33.12 mmol, 37% purity, 3 eq.) at 0 C. The mixture
was stirred
at room temperature for 19 h. TLC showed the starting material was consumed,
and the reaction
was quenched with H20 (75 mL). The mixture was extracted with Et0Ac (50 mL).
After pH
of the aqueous layer was adjusted to 3 with 3 N HCI, the aqueous layer was
extracted with
Et0Ac (3 x 80 mL). The organic layers were combined, washed with brine, dried
(Na2SO4),
filtered, and concentrated to dryness, giving 1-(hydroxymethyl) tetralin-1-
carboxylic acid (1.5
g, 65.9%).
[00346] To a mixture of 1 - (hy droxy methyl) tetralin- 1-
carboxylic acid (1.5 g, 7.27 mmol,
1 eq.) and phenylmethanamine (779.34 mg, 1 eq.) in DMF (15 mL) were added EDCI
(2.09 g,
10.91 mmol, 1.5 eq.), HOBT (1.47 g, 10.91 mmol, 1.5 eq.) and DIEA (1.41 g,
10.91 mmol,
1.90 mL, 1.5 eq.). The mixture was stirred at room temperature for 18 h.
Solvent was removed
by evaporation, followed by addition of water (50 mL) for quenching the
reaction, and
Et0Ac (60 mL). The organic layer was washed (brine), dried (Na2SO4), filtered,
and
evaporated to dryness. The residue was purified by column chromatography on
silica gel
eluted with PE/Et0Ac (100:0 to 70:30), giving N-benzy1-1-
(hydroxymethyptetralin-l-
carboxamide (1.3 g, 60.5%) as an oil.
[00347] To a mixture of N-benzy1-1-(hydroxymethyl) tetralin-1-
carboxamide (1.3 g, 4.40
mmol, 1 eq.) in THF (20 mL) under nitrogen atmosphere was added PPh3 (1.73 g,
6.60 mmol,
1.5 eq.), and then DEAD (1.15 g, 6.60 mmol, 1.04 mL, 1.5 eq.) dropwise at 0
C. The mixture
was stirred at room temperature for 2 h. The reaction was complete as
indicated by TLC
analysis. The resulting solution was quenched with water (20 mL), followed by
addition
of Et0Ac (60 mL). The organic layer was separated, washed (brine), dried
(Na2SO4), filtered,
and evaporated to dryness. The residue was purified by column chromatography
on silica gel
eluted with PE/Et0Ac (100:0 to 67:33), giving 1-benzylspiro [azetidine-3,1'-
tetralin1-2-one (1
g, 81.9%).
156
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00348]
A solution of A1C13 (961.50 mg, 7.21 mmol, 2.0 eq.) in THF (15 mL) was
cooled
to 0 C, followed by addition of LiA1H4 (410.48 mg, 10.82 mmol, 3.0 eq.). The
mixture was
stirred at 0 C for 30 min, followed by addition of a solution of 1-
benzylspiro[azetidine-3,1'-
tetralin1-2-one (1 g, 3.61 mmol, 1 eq.) in THF (4 mL). The mixture was stirred
at room
temperature overnight, and the reaction was quenched with H20 (1 mL), followed
by addition
of 15% aq. NaOH (4 mL) and Et0Ac (20 mL). The solid was removed by filtration
and the
organic layer was concentrated to affordl-benzylspiro [azetidine-3,1'-
tetralin] (800 mg,
84.2%).
[00349]
To a solution of 1-benzylspiro[azetidine-3,1'-tetralin] (800 mg, 3.04 mmol,
1 eq.) in Me0H (30 mL) were added ammonium formate (287.32 mg, 4.56 mmol,
1.5 eq.) and Pd(OH)2 (100 mg). The mixture was stirred under H2 gas atmosphere
and at 60 C
overnight. The mixture was filtered and the residual material was washed with
Me0H. The
filtrate and washing were combined and concentrated, affording crude
spiro[azetidine-3,1'-
tetralin] (500 mg, 95.0%).
[00350]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yl)tetrahydrofuran-2-yl] methyl acetate (500 mg, 1.12 mmol, 1 eq.) in 1,4-
dioxane (20
mL) were added spiro[azetidine-3,1'-tetralin] (251.81 mg, 1.45 mmol, 1.3 eq.)
and DIPEA
(577.96 mg, 4.47 mmol, 778.93 pt, 4 eq.). The mixture was stirred at 100 C
for 3 h. The
mixture was concentrated and the residue was purified by column chromatography
on silica
gel
eluted with PE/Et0Ac (100:0-50:50), affording [(2R,3R,4R,5R)-3,4-di ac
etoxy -5 -(2-
chl oro-6-spiro [azeti dine-3,1 ' -tetralin] -1 -yl-purin-9-y Otetrahy
drofuran-2-yll methyl acetate
(500 mg, 76.5%) as white solid.
[00351]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro[azetidine-
3,1'-tetralin1-1-yl-purin-9-yl)tetrahydrofuran-2-yllmethyl acetate (500 mg,
856_14 umol,
1 eq.) in Me0H (4 mL) was added NH3-Me0H (7 M, 3.67 mL, 30 eq.). The mixture
was
stirred at room temperature for 3 h. The solvent was removed by evaporation
and the residue
was diluted with Et0Ac (100 mL). The organic layer was washed with H20 (70
mL), and then
with brine (60 mL), and dried with Na2SO4, and concentrated, affording
(2R,3R,4S,5R)-2-(2-
chl oro-6-spiro [azeti dine-3, l' -tetralin] -1-yl-purin-9-y1)-5 -
(hydroxymethyl)tetrahydrofuran-
3,4-diol (390 mg, 851. 99.4%).
[00352]
To a solution of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[azetidine-3,1'-
tetralin1-1-yl-
purl n -9-y1)-5 -(hy droxy m ethy Otetrahy drofuran -3 ,4-di ol (391
mg, 853.88 umol,
1 eq.) in acetone (30 mL) were added 2,2-dimethoxypropane (1.78 g, 17.08 mmol,
157
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
20 eq.) and p-Ts0H (147.04 mg, 853.88 pmol, 1 eq.). The mixture was stirred at
room
temperature for 3 h. Solvent was removed by evaporation and the residue was
diluted with
Et0Ac (50 mL), washed with aqueous NaHCO3, then with brine. The organic layer
was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/Et0Ac (100:0-50:50), affording [(3 aR,4R,6R,6 aR)-4-(2-chl o ro-6-s pi ro
[azeti dine-3 ,1
tetralin1-1 -y 1-p urin-9-y1)-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3,4-d]
[1,3] dioxo1-6-
yllmethanol (350 mg, 82.3%) as white solid.
[00353]
A solution of 1(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[azetidine-3,1'-
tetralinl -1-
yl-purin-9-y1)-2,2-dimethy1-3 a,4,6,6 a-tetrahy drofuro [3 ,4-d] [1,3] di oxo1-
6-yll methanol (350
mg, 702.85 [imol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to 0 'V, followed by
addition of a
solution of bis(dichlorophosphoryl)methane (351.12 mg, 1.41 mmol, 2.0 eq.) in
PO(Me0)3 (3
mL). The mixture was stirred at 0 C for 5 h, followed by addition of H20 (5
mL) at 0 C. The
mixture was stirred at 40 C for 30 mm, and then stirred at room temperature
overnight.
Purification of the mixture using C18 reversed phase silica gel (0-25% ACN in
Water)
provided Compound d-12, (253 mg, 57.4%): 1H NMR (500 MHz, CD30D) 6 ppm 2.04
(s, 2H),
2.38 (s, 2H), 2.67 (t, J=20.9 Hz, 2H), 3.02 (t, J=6.1 Hz, 2H), 4.45 (s, 1H),
4.47-4.52 (m, 1H),
4.55 (dd, J=10.4, 7.0 Hz, 1H), 4.62 (t, J=4.8 Hz, 1H), 4.84 (t, J=5.0 Hz, 1H),
6.21 (d, J=4.8 Hz,
1H), 7.28 (d, J=7.5 Hz, 1H), 7.34 (t, J=7.4 Hz, 1H), 7.42 (t, J=7.5 Hz, 1H),
7.81 (d, J=7.9 Hz,
1H), 8.58 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 21.29, 26.46, 27.51, 28.56,
30.92,
37.23, 40.00, 65.91, 71.33, 75.71, 84.63, 89.84, 118.67, 127.05, 127.89,
130.14, 138.07,
140.81, 141.23, 151.77, 155.59, 155.76; 31P NMR (203 MHz, CD30D) 6 ppm 16.86,
19.80;
m/z (ESL): 616.3 (M +H).
Example 36. Synthesis of d-13
158
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Ac0 Isr CI NN
OAc OAc <
N 70 L LIAIH4 IHMDs,
DIFEA, Dioxane
100 C, 16 h __________________________________________________ 1.^ AGO
OAc OAc
Q õ.9 9 9
0 0
NH2-Me0H >(
CI CI
Me0H, rt 0 " P0(0Me)2 0 0
HO N a H -O
HO 41 N CI H20 N CI
H 0H
P-TSOH
0/K0
OH OH OH OH
[00354] To a mixture of indolin-2-one (2 g, 15.02 mmol, 1 eq.)
in THF (30 mL) under
nitrogen atmosphere was added dropwise LiHMDS (1 M, 33.05 mL, 2.2 eq.) at -78
C. The
reaction temperature was raised to -50 C, kept at -50 C for 30 min, and then
cooled back to -
78 C, followed by addition of 1,4-dibromobutane (3.24 g, 15.02 mmol, 1 eq.)
in THF (15 ml.).
The mixture was stirred at room temperature for 2 h, then at reflux for 2 h,
followed by being
stirred at room temperature for 16 h. The mixture was concentrated under
reduced pressure,
and the residue was partitioned between Et0Ac and saturated NH4C1. The organic
layer was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/Et0Ac (100:0 to 60:40). giving spiro[cyclopentane-1,3'-indolin]-2'-one
(1.29 g, 45.87%) as
a yellow solid.
[00355] To a solution of spiro[cyclopentane-1,3'-indoline]-2'-
one (1.29 g, 6.89 mmol,
leq.) in THF (30 mL) was added LiA1H4 (522.92 mg, 13.78 mmol, 2.0 eq.). The
mixture was
stirred at 70 C overnight, then the reaction was quenched with H20 (1 mL),
followed by
addition of Et0Ac (20 mL). The solid was removed by filtration and the organic
layer was
concentrated, the residue was purified by column chromatography on silica gel
eluted with
PE/Et0Ac. (100:0-80:20), affording spiro[cyclopentane-1,3'-indoline] (1.02 g,
85.45%) as
white solid.
[00356] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yl)tetrahydrofuran-2-yllmethyl acetate (1.2 g, 2.68 mmol, 1 eq.) in 1,4-
dioxane (25 mL) were
added spiro[cyclopentane-1,3'-indoline] (511 mg, 2.95 mmol, 1.10 eq.) and
DIPEA (866.94
mg, 6.71 mmol, 1.17 mL, 2.5 eq.). The mixture was stirred at 100 C overnight.
The mixture
was concentrated and the residue was purified by column chromatography on
silica gel eluted
159
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
with PE/Et0Ac (100:0-60:40), affording (2R,3R,4R,5R)-2-(acetoxymethyl)-5-(2-
chloro-6-
(spiro[cyclopentane-1,31-indolin] -11-y1)-9H-purin-9-yptetrahydrofuran-3,4-
diy1 diacetate
(1.49 g, 95.08% yield).
[00357] To a solution
of 1(2R,3R,4R,5R)-3, 4-di acetoxy -5 -(2-chl o ro-6-
Spiro [cy cl opentane-1,3'-indolinel -1'-yl-purin-9-yl)tetrahy drofuran-2-yll
methyl acetate (950
mg, 1.63 mmol, 1 eq.) in Me0H (15 mL) was added Me0H-NH3 (7 M, 5.81 mL, 25
eq.). The
mixture was stirred at room temperature for 4 h. The solid was collected by
filtration, and the
cake was washed with Me0H (20 mL), affording (2R,3R,4S,5R)-2-(2-chloro-6-
spiro [cyclopentane-1,31-indolinel-l'-yl-purin-9-y1)-5-
(hydroxymethyl)tetrahydrofuran-3,4-
diol (580 mg, 77.87% yield).
1003581
To a mixture of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[cyclopentane- I ,3'-
indolinel-
1'-yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (580 mg, 1.27
mmol, 1
eq.) in acetone (20 mL) were added 2,2-dimethoxypropane (1.98 g, 19.00 mmol,
15 eq.) and
p-Ts0H (218.11 mg, 1.27 mmol, 1 eq.). The mixture was stirred at room
temperature for 2 h.
The solvent was removed by evaporation and the residue was diluted with Et0Ac
(50 mL),
washed with aqueous NaHCO3 and then with brine. The organic layer was
concentrated and
the residue was purified by column chromatography on silica gel eluted with
PE/Et0Ac
(100:0-50:50), affording 1(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cyclopentane-
1,31-indolinel-
1 '-yl-purin-9-y1)-2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-d1[1,31dioxol-6-
yllmethanol (540
mg, 85.61% yield).
[00359] A solution
of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cyclopentane-1,3'-
indoline] -1'-y 1 -purin-9-y1)-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3,4-d]
[1,3] di oxo1-6-
yl[methanol (540 mg, 1.08 mmol, 1 eq) in PO(Me0)3 (3 mL) was cooled to 0 C,
followed by
addition of a solution of bis(dichlorophosphoryl) methane (812.60 mg, 3.25
mmol, 10 eq.) in
PO(Me0)3 (3 mL). The mixture was stirred at 0 C for 5 h. Then H20 (4 mL) was
added into
the reaction mixture. The mixture was stirred at room temperature overnight.
Purification of
the reaction mixture using C18 reversed phase silica gel (0-25% ACN in Water)
gave Compound d-13, (149.9 mg, 21.99% yield): 1H NMR (500 MHz, Me0D) 6ppm 1.90
(dd,
J = 17.7, 12.4 Hz, 8H), 2.52 (t, J = 20.9 Hz, 2H), 4.28 (s, 1H), 4.36 (dtd, J
= 15.0, 11.2, 4.7 Hz,
2H), 4.46 (t, J = 4.8 Hz, 1H), 4.55 (s, 2H), 4.66 (t, J = 4.9 Hz, 1H), 6.06
(d, J = 4.7 Hz, 1H),
7.07 (t, J = 7.4 Hz, 1H), 7.18¨ 7.27 (m, 2H), 8.40 ¨ 8.53 (m, 2H). 31P NMR
(203 MHz, Me0D)
6ppm 16.81, 19.86. 13C NMR (126 MHz, Me0D) 6ppm 24.48, 26.15, 27.21, 40.47,
51.52,
160
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
64.71, 64.80, 70.10, 74.33, 83.26, 88.31, 117.29, 119.01, 121.96, 123.89,
126.97, 139.49,
140.10, 142.36, 151.13, 152.00, 153.21. m/z (ESI+): 616.3(M +H).
Example 37. Synthesis of compound d-14
---?
c
N , c, Ill 0 Brõ------,,s g.,.., _
:._ l H 11
ij 0 THLFiAl7H0,0c so DIPEA ,..
_ 10 Dioxane, 0 C N 3 -11 M emOeHo- NH H 3 ..._
F F J F e, Ac0-1- N 'N "Cl
OAc OAc
F
¨_
N.T.k.,N p-Ts0H .
N, , N PO(Me0)9.- N -...N
acetone I
H20 OH OH ,I
HO N- 'N-<-'CI HO N- 'NI' -'CI H0,1 0 N Nr CI
0 0
OH OH 0, 0
--,'--- OH OH
[00360] A solution of 4-fluoroindolin-2-one (1 g, 6.62 mmol, 1
eq.) in THF (20 mL) was
cooled to -78 C, followed by addition of LiHMDS (1 M, 14.56 mL, 2.2 eq.). The
mixture was
stirred at -50 C for 30 min, and then cooled to -78 C. 1,4-dibromobutane
(1.43 g, 6.62 mmol,
1 eq.) in THF (10 mL) was added into the mixture at about -78 C. The mixture
was stirred at
room temperature for 2 h, at 70 'V for 3 h., and at room temperature
overnight. After
completion, the reaction was quenched with aqueous NH4C1 (50 mL) and diluted
with Et0Ac
(60 mL). The organic layer was separated, washed with brine, dried by Na2SO4,
filtered, and
purified by column chromatography on silica gel with (PE: EA=100:0 to 55:45),
giving 4'-
fluorospiro[cyclopentane-1,3'-indoline]-2'-one (750 mg, 55.23%) as a yellow
solid.
1003611
To a solution of 4'-fluorospiro[cyclopentane-1,3'-indoline1-2'-one (500
mg, 2.44
mmol, 1 eq.) inTHF (15 mL) was added LiA1H4 (369.83 mg, 9.75 mmol, 4.0 eq.).
The mixture
was stirred at 70 C for 2 h. The reaction was quenched by addition of H20
(0.3 mL) and 15%
aqueous NaOH (0.6 mL), followed by addition of Et0Ac (30 mL). The solid
material was
removed by filtration and the organic layer was concentrated, affording 4'-
fluorospiro[cyclopentane-1,3'-indoline] (440 mg, 94.43%).
[00362]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-di chloropuri n-9-
yptetrahydrofuran-2-y1Jmethyl acetate (800 mg, 1.79 mmol, 1 eq.) in 1,4-
dioxane (20
mL) were added DIPEA (693.56 mg, 5.37 mmol, 934.71 pL, 3.0 eq.) and 4'-
fluorospiro[cyclopentane-1,3'-indoline] (410.52 mg, 2.15 mmol, 1.2 eq.). The
mixture was
stirred at 100 C overnight, followed concentration. The residue was purified
by column
161
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
chromatography on silica gel eluted with (PE/EA from 100:0 to 60:40),
giving [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(4'-fluorospiro[cyclopentane-
1,3'-
indolinel-1'-yl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (0.9 g, 83.58%)
as light yellow
solid.
[00363] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(4'-
fluorospiro[cyclopentane-1,3'-indoline]-11-yl)purin-9-yll tetrahy drofuran-2-y
1] methy 1 acetate
(900 mg, 1.49 mmol, 1 eq.) in Me0H (6 mL) was added NH3-Me0H (7 M, 6.41 mL, 30
eq.).
The mixture was stirred at room temperature overnight. The solid was collected
by filtration,
affording (2R,3R,4S,5R)-2-[2-chloro-6-(4'-fluorospiro
[cy cl op entane-1,3' -indoline] -1'-y1)
purin-9-y1]-5-(hydroxymethyl) tetrahydrofuran-3,4-diol (700 mg, 98.39%) as
white solid.
1003641
To a solution of (2R,3R,4S,5R)-2-1-2-chloro-6-(4'-fluorospiro
[cyclopentane-
1,3'-indolinel-l'-y1) purin-9-y11-5-(hydroxymethyl) tetrahydrofuran-3,4-diol
(700 mg, 1.47
mmol, 1 eq.) in acetone (20 mL) were added 2,2-dimethoxypropane (2.30 g, 22.06
mmol, 2.71
mL, 15 eq.) and p-Ts0H (253.29 mg, 1.47 mmol, 1 eq.). The mixture was stirred
at room
temperature for 3 h. The mixture was diluted with Et0Ac (50 mL), and the
organic layer was
washed with aqueous NaHCO3 and then with brine, and concentrated. The residue
was purified
by column chromatograph on silica gel eluted with (PE/Et0Ac from 100:0 to
50:50), affording
[(3aR,4R,6R,6aR)-4- [2-chl oro-6-(4'-fluoros pi ro [cy cl op entane-1,3'-indol
ine] -11-y1)p urin-9-
yl] -2,2-dimethy1-3a,4,6,6a-tetrahydrofuro [3,4-dl[1,3]dioxo1-6-yll methanol
(600 mg,
79.06%).
[00365]
A solution of [(3aR,4R,6R,6aR)-442-chloro-6-(4'-fluorospiro[cyclopentane-
1,3 '-indolinel -2,2-dimethy1-3a,4,6,6a-tetrahydrofuro [3,4-
d] [1,3] di oxo1-6-
yl[methanol (300 mg, 581.44 nmol, 1 eq.) in PO(Et0)3 (3 mL) was cooled to 0
C, followed
by addition of a solution of bis(dichlorophosphoryl) methane (290.47 mg, 1.16
mmol,
2.0 eq.) in PO(Et0)3 (2 mL). The mixture was stirred at room temperature for 4
h, and then
H20 (3 ml) was added. The mixture was stirred at room temperature overnight.
Purification of
the reaction mixture using C18 reversed phase silica gel (0-30% ACN in Water)
afforded
Compound d-14, (195 mg, 52.9%): 1H NMR (500 MHz, CD30D) 6 ppm: 1.84 (d, J=20.7
Hz,
6H), 2.08 (d, J=31.3 Hz, 2H), 2.49 (t, J=20.3 Hz, 2H), 4.30 (s, 3H), 4.46 (d,
J=10.2 Hz, 3H),
4.65 (s, 1H), 6.04 (d, J=4.6 Hz, 1H), 6.73 (t, J=9.0 Hz, 1H), 7.15 (d, J=5.9
Hz, 1H), 8.19 (d,
J=8.1 Hz, 1H), 8.46(s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 25.99, 27.50,
40.04, 51.81,
65.57, 67.04, 71.73, 75.77, 88.96, 112.08, 114.79, 120.06, 130.02, 140.94,
145.47, 152.11,
162
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
152.26, 153.25, 154.30; 31P NMR (203 MHz, DMSO-d6) 6 ppm 14.72, 18.24; m/z
(ESI+):
634.2 (M +1-1).
Example 38. Synthesis of compound d-15
II H DIPEA, 1,4-dioxane,
T HLri Al7H040c QS:5/
100 uC
Ma0H-NHN CI
THF, LIHMDS Me0H --
F F =F 4111117 Ac0
OAc OAc
c-5 Q
N/L 9¨fiF
I
p-Ts0H
N
acetone D N PO(Me0)3 N
I H20 x-LN
HO N N CI HO N N 'CI H0,9EJõ9",, N N CI
p p -
O 0
OH OH OH OH
[00366] To a mixture of 5-fluoroindolin-2-one (500.00 mg, 3.31
mmol, 1 eq.) in THF (20
mL) under nitrogen atmosphere was added dropwise LiHMDS (1 M, 7.28 mL, 2.2
eq.) at -
78 C. The temperature of the mixture was raised to -50 C and kept at -50 'V
for 30 min. The
mixture then was cooled to -78 'V, followed by addition of 1,4-dibromobutane
(714.29 mg,
3.31 mmol, 1 eq.) in THF (15 mL). The mixture was stirred first at room
temperature for 1 h,
then at reflux for 3 h, and stirred further at room temperature for 16 h. The
mixture was
concentrated under reduced pressure, and the residual material was partitioned
between Et0Ac
and saturated NH4C1. The organic layer was concentrated and the residue was
purified by
column chromatography on silica gel eluted with PE/Et0Ac (100:0 to 60:40),
giving 5'-
fluorospiro[cyclopentane-1,3'-indoline1-2'-one (350 mg, 1.71 mmol, 51.55%
yield) as a yellow
solid.
[00367] To a solution of 5'-fluorospiro[cyclopentane-1,31-
indoline1-2'-one (350.00 mg,
1.71 mmol, 1 eq.) in THF (30 mL) was added LiA1H4 (161.80 mg, 4.26 mmol, 2.5
eq.). The
mixture was stirred at 70 C for 2 h. The reaction was quenched with H20 (1
mL) and 15%
NaOH (1 mL) at 0 C, followed by addition of Et0Ac (40 mL). The solid was
removed by
filtration and the organic layer was concentrated. Purification of the
residual material by
column chromatography on silica gel, eluted with PE/Et0Ac (100:0-78:22),
afforded 5'-
fluorospiro[cyclopentane-1,3'-indoline] (210 mg, 1.10 mmol, 64.39% yield) as
white solid.
[00368] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (447 mg, 999.49 nmol, 1 eq.) in 1,4-
dioxane (25
163
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mL) were added 5'-fluorospiro[cyclopentane-1,3'-indoline] (210.26 mg, 1.10
mmol, 1.1 eq.)
and DIPEA (322.94 mg, 2.50 mmol, 435.22 iaL, 2.5 eq.). The mixture was stirred
at 100 C for
4 h, and then concentrated. The residue was purified by column chromatography
on silica gel
eluted with PE/Et0Ac (100:0-58:42), affording [(2R,3R,4R,5R)-3,4-diacetoxy -5
42-chl o ro-6-
(5'-fluoros piro [ cy cl op entane-1,3'-indolinel -1'-y1) purin-9-yll
tetrahydrofuran-2-yll methyl
acetate (490 mg, 813.94 itimol, 81.44% yield).
[00369] To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(5'-
fluorospiro [cy cl opentane-1,3'-indoline] -1'-yl)purin-9-yll tetrahy drofuran-
2-yll methyl acetate
(490.00 mg, 813.94 p.mol, 1 eq.) in Me0H (15 mL) was added Me0H-NH3 (7 M, 2.91
mL, 25
eq.). The mixture was stirred at room temperature for 4 h. The solid material
thus formed was
collected by filtration, and washed with Me0H (10 mL), affording (2R,3R,4S,5R)-
2-[2-chloro-
6-(5'-fluorospiro[cyclopentane-1,3'-indoline1-1'-yl)purin-9-y1]-5-
(hydroxymethyptetrahydrofuran-3,4-diol (360 mg, 756.46111pol, 92.94% yield).
[00370]
To a mixture of (2R,3R,4S,5R)-2-[2-chloro-6-(5'-fluorospiro [cyclopentane-
1,31-
indolinel-l'-yOpurin-9-yll-5-(hydroxymethyptetrahydrofuran-3,4-diol (360 mg,
756.46 inmol,
1 eq.) in acetone (25 mL) were added 2,2-dimethoxypropane (1.18 g, 11.35 mmol,
15 eq.) and
p-Ts0H (130.26 mg, 756.46 pinol, 1 eq.) . The mixture was stirred at room
temperature for 2
h. The solvent was removed by evaporation and the residue was diluted with EA
(50 mL),
washed first with aqueous NaHCO3 and then with brine. The organic layer was
concentrated
and the residue was purified by column chromatographyon silica gel eluted with
PE/EA (100:0-
50:50), affording
1(3aR,4R,6R,6aR)-4- [2-chl oro-6-(5'-fluorospiro [ cycl opentane-1,3'-
indoline] -1'-yl)purin-9-yll -2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3 ,4-d]
[1,3] di oxo1-6-
yl[methanol (310 mg, 600.82 vino', 79.42% yield).
[00371]
A solution of [(3aR,4R,6R,6aR)-4-12-chloro-6-(5'-fluorospiro [cyclopentane-
1,3'-indolinel-l'-y1)purin-9-yll -2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3,4-
d] [1,3] di oxo1-6-
yllmethanol (310 mg, 600.82 mmol, 1 eq.) in PO(Me0)3 (3 mL) was cooled to 0
Cfollowed
by addition of a solution of bis(dichlorophosphoryl)methane (450.23 mg, 1.80
mmol, 3.0
eq.) in PO(Me0)3 (3 mL). The mixture was stirred at 0 C for 5 h, followed by
addition of H20
(4 mL). The mixture was stirred at room temperature overnight. Purification of
the reaction
mixture (C18 reversed phase silica gel, 0-25% ACN in Water) gave Compound d-
15, (129.9
mg, 33.99% yield, 99.65% purity): 11-1 NMR (500 MHz, Me0D) 6ppm 1.90 (d, J =
25.6 Hz,
8H), 2.52 (t, J = 20.9 Hz, 2H), 4.28 (s, 1H), 4.34 (dd, J = 22.5, 15.9 Hz,
2H), 4.46 (d, J = 4.6
Hz, 1H), 4.60 (s, 2H), 4.66 (s, 1H), 6.06 (d, J = 4.8 Hz, 1H), 6.95 (d, J =
2.5 Hz, 1H), 7.01 ¨
164
CA 03200232 2023- 5- 25

WO 2022/109741 PCT/CA2021/051688
7.08 (m, 1H), 8.43 (s, 1H), 8.49 (d, J = 4.1 Hz, 1H), 13C NMR (126 MHz, Me0D)
6ppm 24.40,
40.31, 51.52, 69.49, 70.68, 73.69, 74.89, 87.63, 88.95, 117.58, 118.92,
138.71, 140.45, 142.59,
150.89, 151.99, 153.16. 31P NMR (203 MHz, Me0D) 6ppm 16.76, 20.03. m/z (ESI-
1):634.2(M
+H).
Example 39. Synthesis of compound d-16
A00 N N CI
N
Br
Br OAc
111010 LiHMDS =(:) Dl
,
F N THF,-50 C,0.5h F N THF,0 C-rt,o/n F 'N
iPcE0Ac, D3ihoxane AcON N CI
rt,1h,70 C,2h OAc 0A0
C)
F N) F )
I I
0 0 0 0
1,N 9 NH3-Me0H CIJV'I%Ci I
p-Ts0H
xs- HO N N CI HO N CI _________ HO"
Nrk-C1
Me0H, H(,1) acetone
1) PO(OEt)3, 0 C,5h 01-1¨ OH -
2) H20,40'C,4Dmin 11
OH OH 0õ0 rt,O/N OH OH
7\
[00372] To a mixture of 6-fluoroindolin-2-one (500 mg, 3.31 mmol, 1 eq.) in
THF (5
mL) under nitrogen atmosphere was added dropwise LiHMDS (1 M, 7.28 mL, 2.2
eq.) at -78
C, and then it was brought to -50 C for 30 min. The mixture was cooled to -78
C, followed
by addition of 1,4-dibromobutane (714.30 mg, 3.31 mmol, 1 eq.) in THF (5 mL)
was added.
The mixture was stirred at RT for lh, then at reflux for 2 h. The mixture was
quenched with
saturated NH4C1 and extracted with Et0Ac (2 x 30 mL). The organic layer was
washed with
brine and concentrated. The residue was purified by column chromatography on
silica gel
eluted with PE/EA from 100:0 to 70:30), giving 6'-fluorospiro [cyclopentane-
1,3'-indoline1-2'-
one (282 mg, 1.37 mmol, 41.54% yield).
[00373] A solution of A1C13 (493.79 mg, 3.70 mmol, 2 eq.) in THF (10 mL)
was cooled
to 0 C, followed by addition of LiA1H4 (210.80 mg, 5.55 mmol, 3 eq.). The
mixture was stirred
at 0 C for 30 min, followed by addition of a solution of 6'-
fluorospiro[cyclopentane-1,3'-
indoline1-2'-one (380 mg, 1.85 mmol, 1 eq.) in THF (4 mL). the mixture was
stirred at rt
overnight. The mixture was diluted by THF (10 mL) and the reaction was
quenched with slow
addition of 15% NaOH (aq.) at 0 C until pH reached 9. The organic layer was
dried over
MgSO4. The solid was removed by filtration. The filtrate was concentrated, and
the residue
was purified by column chromatography on silica gel eluted with PE/EA (100:0-
90:10),
affording 6'-fluorospiro[cyclopentane-1,3'-indoline] (250 mg, 1.31 mmol,
70.60% yield).
165
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1003741
To a mixture of 6'-fluorospiro(cycloperitane-1,3'-indoline) (250 mg, 1.31
mmol,
1.2 eq.) in 1,4-dioxane
(15 mL) were added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-yptetrahydrofuran-2-yllmethyl acetate (487.19 mg, 1.09 mmol, 1
eq.) and
DIPEA (351.97 mg, 2.72 mmol, 474.36 L, 2.5 eq.) . The mixture was stirred at
100 C
overnight. TLC showed about 30% starting material was left and new product
formed. The
mixture was stirred at 120 C for 3 more hours, and then cooled to it. Solvent
was removed by
evaporation. The residue was diluted with Et0Ac (40 mL), washed first with
water and then
with brine. The organic layer was concentrated and the residue was purified by
column
chromatographyon silica gel eluted with PE/EA (100:0-60:40), affording
1(2R,3R,4R,5R)-3,4-
di acetoxy-5- [2-chl oro-6-(6'-fl uorospi ro [ cy cl op entane-1,3 '-indoline]
-1'-yOpurin-9-
ylltetrahydrofuran-2-yllmethyl acetate (520 mg, 863.77 [Imo], 79.29% yield) as
light yellow
solid.
1003751 To a mixture
of 1(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(6'-
fluorospiro[cyclopentane-1,3'-indolinel -11-yl)purin-9-yll tetrahy drofuran-2-
yll methyl acetate
(520 mg, 863.77 jtmol, 1 eq.) in Me0H (5 mL) was added NH3-Me0H (7 M, 3.70 mL,
30 eq.).
The mixture was stirred at room temperature for 4 h. LC-MS showed intermediate
was not
consumed, then it was stirred at room temperature overnight. At this point,
analysis (LC-MS)
indicated completion of the reaction. Solvent was removed by evaporation, and
the residue was
used for the next step without further purication.
1003761
To a solution of (2R,3R,4S,5R)-2-[2-chloro-6-(6'-fluorospiro[cyclopentane-
1,3'-
indolinel-1'-yl)purin-9-yll -5 -(hy droxy methyl)tetrahy drofuran-3,4-di ol
(410 mg, 861.53 nmol,
1 eq.) in acetone (20 mL) were added p-Ts0H-H20 (165.41 mg, 861.53 wnol, 1
eq.) , 2,2-
dimethoxypropane (1.79 g, 17.23 mmol, 2.12 mL, 20 eq.). The mixture was
stirred at RT for 2
h. LC-MS showed SM was consumed. The pH of the mixture was adjusted to 9 by
addition of
15% NaOH (aq.) slowly at 0 C. Solvent was removed by evaporation and the
residue was
extracted with Et0Ac (2 x 30 mL). The organic layer was washed with brine, and
concentrated.
The residue was purified by column chromatographyon silica gel eluted with
PE/EA (100:0-
50:50) to afford [(3aR,4R,6R,6aR)-442-chloro-6-(6'-fluorospiro[cyclopentane-
1,3'-indolinel-
1'-yl)purin-9-yll -2,2-dimethy1-3 a,4,6,6a-tetrahydrofuro [3,4-d] [1,31dioxo1-
6-yll methanol (410
mg, 794.63 nmol, 92.24% yield) as white solid.
1003771
A solution of [(3aR,4R,6R,6aR)-442-chloro-6-(6'-fluorospiro[cyclopentane-
1,3'-i ndol in el -1'-yl)purin -9-y1 -2,2-di methy1-3 a,4,6,6a-tetrahydrofuro
[3,4-d] [1,3] di ox 01-6-
yll methanol (380 mg, 736.49 nmol, 1 eq.) in P0(0E03 (3 mL) was cooled to 0
C, followed
166
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
by addition of a solution of bis(dichlorophosphoryl)methane (367.93 mg, 1.47
mmol, 2.0
eq.) in P0(0E03 (3 mL). The mixture was stirred at 0 C for 4 h. LC-MS showed
that about
very little starting material was left. H20 (3 mL) was slowly added into the
mixture at 0 C.
The mixture was stirred at 40 C for 40 min, then was stirred at rt overnight.
LC-MS showed
intermediate was consumed and product was detected. Purification of the
reaction mixture
using C18 reversed phase silica gel (0-30% ACN in Water) gave Compound d-16,
(255 mg,
54.24% yield with 99.29% purity): 1H NMR (500 MHz, Me0D) 6 ppm 1.91 (s, 8H),
2.57 (s,
2H), 4.31 (s, 1H), 4.34¨ 4.46 (m, 2H), 4.49 (s, 1H), 4.57 (s, 2H), 4.69 (s,
1H), 6.08 (d, J = 4.6
Hz, 1H), 6.77 (dd, J = 11.7, 5.2 Hz, 1H), 7.20 (dd. J = 8.0, 5.7 Hz, 1H), 8.20
(s, 1H), 8.46 (s,
1H); 31P NMR (203 MHz, Me0D) 6 ppm 16.85, 20.04; 13C NMR (126 MHz, Me0D) 6 ppm
24.31, 25.03, 26.09, 27.15, 40.61, 50.88, 64.73, 65.35, 70.07, 74.34, 83.17,
88.33, 104.93,
109.85, 118.96, 122.60, 135.54, 139.88, 143.43, 150.89,152.06, 153.00, 161.09,
162.99; miz
(ESI-1):634.3(M +H).
Example 40. Synthesis of compound d-1 7
4/Nr-(\
H Br,
N -Br N LiAIH4 ry
I 0 ______________ 0 ____________ DIPEA, NMP, C
Me0H-NH3
THF, LIHMDS THF, 70'e I
Me0H
3 Ac0 N CI
OAc OAc
rs1)(R QNP
N
F p_Ts0H F iorp(Me0)3
_______________________________________________ 3
<5'1 acetone I
H20
HO N HO N 91-1 Ner-1¨"Cl
0 0
OH OH 0 0
OH OH
[00378] A solution of 7-fluoroindolin-2-one (600 mg, 3.97 mmol,
1 eq.) in THF (25
mL) was cooled to about -78 C, followed by addition of LiHMDs (1 M, 8.73 mL,
2.2 eq.).
The mixture was stirred at about -50 'V for 30 min, then cooled to -78 C. To
the cold mixture
was added 1,4-dibromobutane (857.16 mg, 3.97 mmol, 1 eq.) in THF (5 mL) at -78
C. The
mixture was stirred at room temperature for 2 h, then stirred at 70 C for 3
h, and finally, stirred
at room temperature overnight. The reaction was quenched by addition of
aqueous NH4C1 (30
mL), followed by addition of Et0Ac (40 mL). The organic layer was separated,
washed with
brine, dried (Na2SO4), filtered, and evaporated to dryness. The residue was
purified by column
167
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
chromatography on silica gel eluted with (PE/Et0Ac from 100:0 to 75:25),
giving 7'-
fluorospiro[cyclopentane-1,3'-indolinel-2'-one (590 mg, 72.42%) as brown
solid.
[00379]
To a solution of 7'-fluorospiro[cyclopentane-1,3'-indolinel-2'-one (590
mg, 2.87
mmol, 1 eq.) in THF (20 mL) was added LiA1H4 (436.40 mg, 11.50 mmol, 4.0 eq.).
The
mixture was stirred at 70 'V for 2 h. The mixture was quenched by addition of
H20 (0.3 mL)
and 15% aqueous NaOH (0.6 mL). Et0Ac (40 mL) was added into the mixture. Solid
was
removed by filtration and the organic layer was concentrated and the residue
was purified by
column chromatography on silica gel eluted with (PE/Et0Ac from 100:0 to
75:25), giving 7'-
fluorospiro[cyclopentane-1,3'-indolinel (300 mg, 54.57%) as white solid.
[00380]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yl)tetrahydrofuran-2-yllmethyl acetate (350 mg, 782.60 nmol, 1 eq.) in NMP (20
mL) were
added 7'-fluorospiro[cyclopentane-1,3'-indolinel (149.67 mg, 782.60 wnol, 1
eq.) and D1PEA
(202.29 mg, 1.57 mmol, 272.62 nL, 2.0 eq.). The mixture was stirred at 140 C
overnight. The
mixture was diluted with Et0Ac (30 mL), washed first with water and then with
brine (3 x 30
mL). The organic layer was concentrated and the residue was purified by column
chromatography on silica gel eluted with (PE/Et0Ac from 100:0 to 60:40),
giving [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(7'-fluorospiro[cyclopentane-
1.3'-
indolinel-11-yl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (100 mg,
21.23%) as brown
solid.
[00381] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-(7'-
fluorospiro [cy clopentane-1,3'-indolinel -1'-yl)purin-9-yll tetrahy drofuran-
2-y methyl acetate
(350 mg, 581.39 wnol, 1 eq.) in Me0H (3 mL) was added NH3-Me0H (7 M, 2.49 mL,
30 eq.).
The mixture was stirred at room temperature overnight, and then concentrated.
The residue was
diluted with Et0Ac (30 mL), washed with water and brine subsequently, dried
with Na2SO4,
and concentrated, affording (2R,3R,4S,5R)-242-chloro-6-(7'-
fluorospiro[cyclopentane-1,3'-
indolinel-1'-yl)purin-9-yll-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (250 mg,
90.36%) as
brown solid.
[00382]
To a solution of (2R,3R,4S,5R)-2-[2-chloro-6-(7'-fluorospiro [cyclopentane-
1,3'-indolinel-l'-yl)purin-9-yll-5-(hydroxymethyl)tetrahydrofuran-3,4-diol
(270 mg, 567.35
tamol, 1 eq.) in acetone (10 mL) were added 2,2-dimethoxypropane (886.31 mg,
8.51 mmol,
1.05 mL, 15 eq.) and p-Ts0H (97.70 mg, 567.35 [tmol, 1 eq.). The mixture was
stirred at room
temperature for 3 h. The mixture was diluted with Et0Ac (50 mL), washed first
with aqueous
NaHCO3 and then with brine. The organic layer was concentrated and the residue
was purified
168
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
via column chromatograph on silica gel eluted with (PE/Et0Ac from 100:0 to
50:50),
afforded [(3aR,4R,6R,6aR)-442-chloro-6-(7'-fluorospiro[cyclopentane-1,31-
indolinel-V-
yl)purin-9-y11-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3] dioxo1-6-yll
methanol (290
mg, 99.07%)
[00383] A solution of [(3aR,4R,6R,6aR)-442-chloro-6-(71-
fluorospiro[cyclopentane-
1,31-indoline]-1'-yl)purin-9-yll -2,2-dimethy1-3 a,4,6,6a-tetrahydrofuro [3 ,4-
d1 [1,3] di oxo1-6-
yllmethanol (290 mg, 562.06 umol, 1 eq.) in PO(Et0)3 (3 mL) was cooled to 0
C, followed
by addition of a solution of bis(dichlorophosphoryl)methane (280.79 mg, 1.12
mmol,
2.0 eq.) in PO(Et0)3 (2 mL). The mixture was stirred at room temperature for 4
h, and H20 (3
ml) was added into the mixture. The mixture was stirred at room temperature
for overnight.
Purification of the reaction mixture using C I 8 reversed phase silica gel (0-
30% ACN in Water)
provided Compound d-17, (198 mg, 55.7%): 1H NMR (500 MHz, CD30D) 6 ppm 1.81
(dd,
J=36.0, 29.1 Hz, 8H), 2.51 (t, J=20.9 Hz, 2H), 4.27 (s, 1H), 4.32 (d, J=6.6
Hz, 1H), 4.37 (s,
1H), 4.45 (d, J=9.7 Hz, 3H), 4.68 (t, J=5.0 Hz, 1H), 6.07 (d, J=4.8 Hz, 1H),
7.01 (t, J=9.2 Hz,
1H), 7.09 (d, J=6.9 Hz, 1H), 7.15 (dd, J=7.8, 4.2 Hz, 1H), 8.51 (s, 1H); 13C
NMR (125 MHz,
CD30D) 6 ppm 25.94, 27.47, 39.77, 55.20, 65.92, 68.14, 71.50, 75.63, 84.72,
89.77, 116.28,
118.88, 121.40, 127.55, 130.21, 141.98, 146.35, 152.28, 153.71, 154.31,
154.75; 31P NMR
(203 MHz, CD30D) 6 ppm 16.73, 20.18; m/z (ESI-1): 634.1 (M+H).
[00384] Example 41. Synthesis of compound d-18
Ac0 N reLti QNc5
OAc OAc <
N.
1110:No _____________________ 410 Co LiAIH4 r _____________ c Ac0
N CI
LiHMDs, THF 70 C N
DIPEA, Dioxane
100 C, 16 h
OAc OAc
0 9
-
Q 9 9
0 0
NH,-Me0H Cl a
Me0H, rt NI/c _______
ci 0 P0(0Me), 9
c
HO1 NCI
p-Ts0H ¨1124 H20 H0Ho
I
0)<.0
OH OH OH OH
[00385] To a mixture of indolin-2-one (1.33 g, 9.99 mmol, 1 eq.)
in THF (20 mL) under
nitrogen atmosphere was added dropwise LiHMDS (1 M, 21.98 mL, 2.2 eq.) at -78
C. The
temperature was raised -50 C and kept at this temperature for 30 min. The
mixture was cooled
to -78 C, followed by addition of 1-bromo-2-(2-bromoethoxy)ethane (2.32 g,
9.99 mmol, 1
169
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
eq.) in THF (15 mL). The mixture was stirred at room temperature for 2 h, then
at reflux for 3
h, and finally at room temperature for 16 h. The mixture was concentrated
under reduced
pressure, and the residue was partitioned between Et0Ac and saturated NH4C1.
The organic
layer was concentrated and the residue was purified by column chromatography
on silica gel
eluted with (PE/Et0Ac from 100:0 to 60:40), giving spirofindoline-3,4'-
tetrahydropyran1-2-
one (420 mg, 20.69% yield) as a yellow solid.
[00386]
To a solution of spirofindoline-3,4'-tetrahydropyran1-2-one (410 mg, 2.02
mmol,
1 eq.) in THF (15 mL) was added (153.12 mg, 4.03 mmol, 2.0 eq.). The mixture
was stirred at
70 C overnight. The mixture was quenched with H20 (0.5 mL), followed by
addition of
Et0Ac (20 mL). The solid was removed by filtration and the organic layer was
concentrated.
Purification of the residue by column chromatography on silica gel (eluted
with PE/Et0Ac
from 100:0-75:25) afforded spiro[indoline-3,4'-tetrahydropyran] (310 mg,
81.20% yield) as
pink solid.
[00387]
To a solution of [(2R,3R,4R,5R)-3 ,4-di acetoxy-5-(2,6-di chl oropurin-9-
yOtetrahydrofuran-2-yll methyl acetate (666 mg, 1.49 mmol, 1 eq.) in 1,4-
dioxane (15
mL) were added spiro[indoline-3,4'-tetrahydropyran] (310.01 mg, 1.1 eq.) , and
DIPEA
(481.15 mg, 3.72 mmol, 2.5 eq.). The mixture was stirred at 100 C overnight,
and
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/Et0Ac (100:0-10:90), affording
[(2R,3R,4R,5R)-3,4-di acetoxy -5 -(2-chl o ro-6-
Spiro [indoline-3,4'-tetrahy dropy ran] -1 -yl-purin-9-yl)tetrahy drofuran-2-
yl] methyl acetate (550
mg, 61.55% yield).
[00388]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spirofindoline-
3,4'-tetrahydropyranJ-1-yl-purin-9-yptetrahydrofuran-2-y1Jmethyl acetate (550
mg, 916.64
[mot, 1 eq.) in Me0H (5 mL) was added Me0H-NH3(7 M, 3.27 mL, 25 eq.). The
mixture was
stirred at room temperature for 5 h. The solid was collected by filtration,
washed with Me0H
(15
mL), affording (2R,3R,4S,5R)-2-(2-chloro-6-spiro findoline-3,4'-
tetrahydropyrani -1 -yl-
purin-9-y1)-5-(hy droxy methy 1) te trahy drofuran-3,4-diol (310 mg, 71.36%
yield).
[00389] To a mixture
of (2R,3R,4S,5R)-2-(2-chloro-6-spiro [indoline-3,4'-
tetrahy dropy ran] -1-yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol
(310 mg,
654.13 [tmol, 1 eq.) in acetone (10 mL) were added 2,2-dimethoxypropane (1.02
g, 9.81 mmol,
15 eq.) and p-Ts0H (112.64 mg, 654.13 [tmol, 1 eq.). The mixture was stirred
at room
temperature for 2 h. The solvent was removed by evaporation and the residue
was diluted with
Et0Ac (50 mL), washed first with aqueous NafIC03 and then with brine. The
organic layer
170
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
was concentrated and the residue was purified by column chromatography on
silica gel eluted
with PE/Et0Ac (100:0-5:95), affording [(3aR,4R,6R,6aR)-4-(2-chloro-6-
spiro[indoline-3,41-
tetrahy dropy ran] -1 -yl-purin-9-y1)-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro
113 ,4-d] [1,3] di oxo1-6-
yllmethanol (252 mg, 74.95% yield).
[00390] A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-
spiro[indoline-3,4'-
letrahydropyran1-1-yl-purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahy drofuro 113
,4-d] [1,3] dioxo1-6-
yllmethanol (252 mg, 490.30 umol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to
about 0 C,
followed by addition of a solution of bis(dichlorophosphoryl)methane (306.17
mg, 1.23 mmol,
2.5 eq.) in PO(Me0)3 (3mL). The mixture was stirred at room temperature for 5
h, and then
H20 (4 mL) was added into the mixture, followed by stirring the mixture room
temperature
overnight. Purification of the reaction mixture using CI8 reversed phase
silica gel (0-25%
ACN in Water) gave Compound d-18, (39.0 mg, 12.43% yield): 1H NMR (500 MHz,
Me0D)
6ppm 1.61 (d, J = 13.5 Hz, 2H), 2.04 (t, J = 13.1 Hz, 2H), 2.53 (t, J = 21.0
Hz, 2H), 3.73 (dd, J
= 16.3, 8.0 Hz, 2H), 3.98 (d, J = 10.5 Hz, 2H), 4.29 (s, 1H), 4.31 ¨ 4.43 (m,
2H), 4.46 (t, J =
4.6 Hz, 1H), 4.66¨ 4.75 (m, 3H), 6.06 (d, J = 4.6 Hz, 1H), 7.10 (t, J = 7.4
Hz, 1H), 7.24 (t, J =
7.7 Hz, 1H), 7.29 (d, J = 7.4 Hz, 1H), 8.45 ¨ 8.54 (m, 2H). 31P NMR (203 MHz,
Me0D) oppm
16.77, 20.00; 13C NMR (126 MHz, Me0D) oppm 26.14, 37.03, 42.16, 60.27, 64.65,
70.13,
74.32, 83.28, 88.33, 117.70, 118.98, 122.34, 123.83, 127.62, 139.74, 142.05,
151.14, 152.05,
153.17. m/z (ESI+): 632.3(M +H).
Example 42. Synthesis of compound d-19
CI
N 40 Dei
NC
AO-1 N CI
Br N 24
LiHMDS "Or n-BuLl HCI-EA
0 OAc OAc
Br THF,-78 C,3h NC
( THF,-78"C,2h Nrt3h L DIPEA, Dioxane
NH HCI 100 oC, 4h
\NI
0
OAc OAc
qc,ii
.0
NH3-Me0H N p-To0H.H20 N .. CI' .. CI
Me0H, rt.3h acetone,rt,3h 1) P0(0M7),, O'C,5h
¨ Fic,Lko eNX::;11,ci
HO N lej'a H 7 N.,. CI c?,1 -- 2) HZ,: 0,40min -- OH OH ¨I
124
Oxf OH OH
OH OH
171
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00391] A solution of tert-butyl 3-cyanoazetidine-1-carboxylate
(3.0 g, 16.46 mmol, 1
eq.) in THF (30 mL) was cooled to about -78 C, followed by addition of LiHMDS
(1 M, 20.58
mL, 1.25 eq.) dropwise at -78 C. The mixture was stirred for 20 min at -78
C, followed by
addition of a solution of 1-(bromomethyl)-2-iodo-benzene (5.13 g, 17.29 mmol,
1.05 eq.) in
THF (3 mL). The mixture was stirred at -78 C for 3 h, and the reaction was
quenched by
saturated NH4C1. The mixture was extracted with Et0Ac (50 mL * 2). The organic
layer was
washed with brine, and concentrated. The residue was purified by column
chromatographyon
silica gel eluted with PE/EA (100:0-83:17) and afford tert-butyl 3-cyano-3-[(2-
iodophenyOmethyllazetidine-1-carboxylate (6.44 g, 16.17 mmol, 98.22% yield) as
yellow oil.
[00392] A solution of tert-butyl 3-cyano-3-[(2-
iodophenyl)methyl]azetidine-1-
carboxylate (6.44 g, 16.17 mmol, 1 eq.) in THF (60 mL) was cooled to about -78
C, followed
by addition of n-BuLi (2.5 M, 12.94 mL, 2 eq.) dropwise at about -78 C. The
mixture was
stirred at -78 C for 2 h, and the reaction was quenched by saturated NH4C1.
The mixture was
extracted with Et0Ac (75 mL * 2). The organic layer was washed with brine, and
concentrated.
The residue was purified by column chromatographyon silica gel eluted with
PE/EA (100:0-
83:17), affording tert-butyl 1'-oxospiro[azetidine-3,2'-indaneJ-1-carboxylate
(3.4 g, 12.44
mmol, 76.92% yield) as light-yellow solid.
[00393] Tert-butyl 1'-oxospiro[azetidine-3,2'-indane1-1-
carboxylate (350 mg, 1.28
mmol, 1 eq.) was dissolved in HC1-EA (4 mL). The mixture was stirred at RT for
3 h,
concentrated, and used directly for the next step.
[00394] To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yptetrahydrofuran-2-yll methyl acetate (408.31 mg, 912.99 [tmol, 1 eq.) in 1,4-
dioxane (20
mL) were added spiro[azetidin-1-ium-3,2'-indaneJ-1'-one chloride (268 mg, 1.28
mmol, 1.4
eq.) and DIEA (412.98 mg, 3.20 mmol, 556.58 iaL, 3.5 eq.). The mixture was
stirred at 100 C
for 4 h. Solvent was removed by evaporation. The residue was diluted with
Et0Ac (40 mL),
washed first with water, and then with brine. The organic layer was
concentrated and the
residue was purified by column chromatographyon silica gel eluted with PE/EA
(100:0-50:50),
affording [(2R,3R,4R,5R)-3,4-di acetoxy -5 - [2-chl o ro-6-(1'-oxo spiro
[azeti dine-3,2'-indane] -1 -
yl)purin-9-ylltetrahydrofuran-2-yll methyl acetate (440 mg, 753.46 lama 82.53%
yield) as
white solid.
[00395] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(11-
ox os pi ro [azeti di n e-3, 2'-i n dan el -1 -yl)puri n-9-ylltetrahy drofuran
-2-y I] methyl acetate (337 mg,
577.08 umol, 1 eq.) in Me0H (5.00 mL) was added NH3-Me0H (7 M, 2.47 mL, 30
eq.). The
172
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mixture was stirred at room temperature for 3h, and concentrated, affording
the crude product.,
which was extracted with Et0Ac (50 mL * 2) and washed with water (50 mL). The
organic
layer was washed with brine, dried with Na2SO4, and concentrated, affording 1-
[2-chloro-9-
[(2R,3R,4 S,5R)-3,4-dihy droxy -5-(hy droxy methy Otetrahy drofuran-2-yllpurin-
6-
yllspiro[azetidine-3,2'-indanel-F-one (264 mg, 576.59 pmol, 99.91% yield) as
white solid.
[00396] To a solution
of 1 -12-chloro-9- [(2R,3R,4S ,5R)-3,4-dihy droxy -5-
(hy droxymethyptetrahydrofuran-2-yl]purin-6-yllspiro [azetidine-3,2'-indane] -
1'-one (300 mg,
655.21 prnol, 1 eq.) in acetone (30 mL) were added 2,2-dimethoxypropane (1.36
g, 13.10
mmol, 1.61 mL, 20 eq.) and p-Ts0H-H20 (125.80 mg, 655.21111-ml, 1 eq.) at 0 C.
The mixture
was stirred at room temperature for 3 h, followed by removal of solvent. The
residue was
diluted with EA (50 mL), washed first with aqueous NaHCO3 (2 x 50 mL) and then
with (50
mL). The organic layer was concentrated. The residue was purified by column
chromatography
on silica gel eluted with PE/EA (100:0-60:40),
affording 1-19-[(3aR,4R,6R,6aR)-6-
(hy droxy methyl)-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro 13 ,4-d] [1,3] di
oxo1-4-yll -2-chl oro-
purin-6-yllspiro[azetidine-3,2'-indanel-l'-one (270 mg, 542.25 jamol, 82.76%
yield) as white
solid.
[00397] A solution
of 149-1(3 aR,4R,6R,6 aR)-6-(hy droxy methyl)-2,2-dimethyl-
3a,4,6,6a-tetrahy drofuro[3,4-d] [1,3] dioxo1-4-y11-2-chloro-purin-6-
yl]spiro[azetidine-3,2'-
indanel- 1' -one (270 mg, 542.25 ittmol, 1 eq.) in P0(0Me)3 (4 mL) was cooled
to 0 C,
followed by addition of bis(dichlorophosphoryl)methane (270.89 mg, 1.08 mmol,
2 eq.) in
PO(Me0)3 (4 mL). The mixture was stirred at 0 C for 5 h. LC-MS analysis
indicated that the
reaction did not progress much. Bis(dichlorophosphoryl)methane (135.4 mg, 0.54
mmol, 1
eq.) in PO(Me)03 (2 mL) was added into the mixture. The mixture was stirred at
RT overnight.
LCMS analysis indicated about 50% SM left. The mixture was used directly for
the next step.
[00398]
To the above mixture was added H20 (7 mL) at 0 C. The mixture was stirred
at
room temperature overnight. The mixture was purified by Prep-HPLC. H NMR
analysis
indicated impurity in the crude product (containing P0(0Me)3). The crude
product was
purified using C18 reversed phase silica gel (0-25% ACN in Water), giving
Compound d-19,
(60.6 mg, 98.11 limo', 22.38% yield, 99.71% purity): 1H NMR (500 MHz, Me0D) 6
ppm 2.52
(t, J = 20.9 Hz, 2H), 3.68 (s, 2H), 4.25 ¨ 4.43 (m, 4H), 4.47 (t, J = 4.7 Hz,
2H), 4.74 (d, J =
68.8 Hz, 3H), 6.04 (d, J = 4.7 Hz, 1H), 7.49 (t, J = 7.5 Hz, 1H), 7.61 (d, J =
7.7 Hz, 1H),7.74
(t, J = 7.5 Hz, 1H) ,7.80 (d, J = 7.6 Hz, 1H) ,8.44 (s, 1H). 31P NMR (203 MHz,
Me0D) 6 ppm
16.84, 19.73. 13C NMR (126 MHz, Me0D) 6 ppm 15.90, 26.14, 38.96, 46.25, 56.11,
58.97,
173
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
60.87, 64.58, 70.07, 74.31, 83.31, 88.34, 117.89, 123.72, 126.42, 127.67,
134.91, 135.65,
140.10, 150.63, 152.95, 154.15, 206.67. m/z (ESt): 616.3(M +H).
Example 43. Synthesis of compound d-20
CI
XLN
alL
Ac0
CI
=
ap = 111-
1
110
CAC OAc IlAr NI 1,1H,Me0H N oTH -H5M0 DCSO 5h L1AIH4
THF,70"C 3h N DIPEA, Dioxn ae
N Me0H, rt,
3h:
H rt,1h,70 C,3h H
100 C, 4 h I eLCI
Ac0
¨134 OH OH
OAc OAc
= =
0 0
* =
p-Ts0H <,N C I Y9
"----P I \C IC 0 0
N
acetone,rt,2h Ho ¨1411 N CI 1) P0(0Me)3, 0'C,5h HO ci
2) N20,40C,40min OH OH `-'.1.L:4)
rt,O/N
0><3
OH OH
[00399] To a mixture of indolin-2-one (1 g, 7.51 mmol, leq.) in
THF (8 mL) under
nitrogen atmosphere was added dropwise LiHMDS (1 M, 16.52 mL, 2.2 eq.) at -78
C, and the
temperature was raised to -50 C and kept at this temperature for 30 min. The
mixture was then
cooled to -78 C, followed by addition of 1,3-dibromopropane (1.52 g, 7.51
mmol, 1 eq.) in
THF (8 mL). The mixture was stirred at rt for 1 h, then at reflux for 3 h. The
mixture was
concentrated under reduced pressure and the residue was partitioned between
Et0Ac and
saturated NH4C1. The organic layer was concentrated and the residue was
purified by column
chromatography on silica gel eluted with PE/EA (100:0 to 85:15), giving
spiro[cyclobutane-
1,3'-indoline1-2'-one (345 mg, 1.99 mmol, 26.52% yield).
[00400] To a solution of spiro[cyclobutane-1,3'-indo1ine1-2'-one
(345 mg, 1.99 mmol,
leq) in THF (20 mL) was added LiA1H4 (151.18 mg, 3.98 mmol, 2 eq.). The
mixture was
stirred at 70 C for 4 h. The reaction was quenched with 15% NaOH (aq., 5 mL)
at 0 C,
followed by addition of Et0Ac (20 mL). The organic layer was dried over MgSO4.
After the
solid was removed by filtration, the filtrate was concentrated, affording
spiro[cyclobutane-1,3'-
indoline] (317 mg, 1.99 mmol, 99.95% yield).
[00401] To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-yll methyl acetate (593.58 mg, 1.33 mmol, leq.) in 1,4-
dioxane (30 mL)
were added spiro[cyclobutane-1,3'-indoline] (317 mg, 1.99 mmol, 1.5eq.) and
DIEA (428.83
mg, 3.32 mmol, 577.94 pL, 2.5eq.). The mixture was stirred at 100 'V for 5 h.
Solvent was
removed by evaporation. The residue was diluted with Et0Ac (50 mL), washed
first with water
and then with brine. The organic layer was concentrated and the residue was
purified by column
174
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
chromatographyon silica gel eluted with PE/EA (100:0-65:35), affording
[(2R,3R,4R,5R)-3,4-
di acetoxy-5-(2-chl oro-6-spiro [cy cl obutane-1,3 -1'-yl-purin-9-
yl)tetrahydrofuran-2-
yll methyl acetate (586 mg, 1.03 mmol, 77.46% yield), as light-yellow solid.
[00402] To a mixture of
[(2R,3R,4R,5R)-3,4-diacetoxy -5 -(2-chl oro-6-
spiro[cyclobutane-1,3'-indoline] -1'-yl-purin-9-yl)tetrahydrofuran-2-yllmethyl
acetate (586
mg, 1.03 mmol, leq.) in Me0H (5 mL) was added NH3-Me0H (7 M, 4.41 mL, 30 eq.).
The
mixture was stirred at room temperature for 3 h. LC-MS analysis indicated that
the starting
material was consumed, the molecular ion of the product was detected, and
certain amount of
intermediate was present. To the mixture was added NH3-Me0H (2 mL), and the
mixture was
stirred at rt overnight. Solvent was removed by evaporation, and the residue
was diluted with
Et0Ac (2 x 30 mL) and water (30 mL). The organic layer was washed with brine,
dried with
Na2SO4, and concentrated, affording (2R,3R,4S,5R)-2-(2-chloro-6-
spiro[cyclobutane-1,3'-
indolinel-1'-yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (450 mg,
1.01 mmol,
98.61% yield) as yellow solid.
[00403]
To a solution of (2R,3R,4S,5R)-2-(2-thloro-6-spiro[cyclobutane-1,3'-
indoline]-
1'-yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (450 mg, 1.01
mmol, leq.)
in acetone (30 mL) were added 2,2-dimethoxypropane (2.11 g, 20.28 mmol, 2.49
mL, 20 eq.)
and p-Ts0H H2O-
(193.92 mg, 1.01 mmol, 1 eq.) at 0 C. The mixture was stirred at room
temperature for 3 h. Solvent was removed by evaporation, and the residue was
diluted with EA
(50 mL), washed first with NaHCO3 (aq, 2 x 50 mL) and then with brine (50 mL),
and
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/EA (100:0-60:40), affording [(3aR,4R,6R,6aR)-4-(2-chloro-6-
spiro[cyclobutane-1,3'-
indoline] -1'-y 1 -purin-9-y1)-2,2-dimethy1-3 a,4,6,6 a-tetrahy drofuro [3,4-
d] [1,3] di oxo1-6-
yllmethanol (370 mg, 764.55 iumol, 75.42% yield) as light yellow solid.
[00404]
A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cyclobutane-1,3'-
indoline1-
1'-yl-purin-9-y1)-2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3]dioxol-6-
yl[methanol (370
mg, 764.55 iumol, 1 eq.) in PO(Me0)3 (4 mL) was cooled to 0 C, followed by
addition of a
solution of bis(dichlorophosphoryl)methane (381.95 mg, 1.53 mmol, 2 eq.) in
PO(Me0)3 ( 4
mL). The mixture was stirred at 0 C for 5 h. LC-MS indicated that
approximately 50% of
starting material was left. The reaction was continued, with addition of
bis(dichlorophosphoryOmethane (191 mg, 0.765 mmol, 1 eq.) in PO(Me0)3 (2 mL)
at 0 C, by
stirring the mixture at rt for 2 h. At this point, LC-MS analysis indicated
disappearance of the
starting material. H20 (7 mL) was added dropwise into the mixture at 0 'V, and
the mixture
175
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
was stirred first at 40 C for 40 min and then at rt overnight. Purification
of the reaction mixture
using C18 reversed phase silica gel (0-25% ACN in Water) provided Compound d-
20, (106
mg, 176.12 mmol, 23.04% yield, 98.33% purity). 1H NMR (500 MHz, Me0D) 6 ppm
2.10 ¨
2.22 (m, 2H), 2.38 (dd, J = 15.8, 9.9 Hz, 2H), 2.48 (d, J = 8.5 Hz, 2H), 2.56
(1, J = 20.9 Hz,
2H), 4.32 (s, 1H), 4.34 ¨ 4.39 (m, 1H), 4.40 ¨ 4.46 (m, 1H), 4.49 (t, J = 4.8
Hz, 1H), 4.70 (t, J
= 4.8 Hz, 1H), 4.84 (s, 2H), 6.08 (d, J = 4.6 Hz, 1H), 7.14 (t, J = 7.4 Hz,
1H), 7.23 (1, J = 7.8
Hz, 1H), 7.54 (d, J = 7.4 Hz, 1H), 8.46 (d, J = 8.2 Hz, 1H), 8.48 (s, 1H). 31P
NMR (203 MHz,
Me0D) 6 ppm 16.80, 19.93. 13C NMR (126 MHz, Me0D) 6 ppm 15.43, 26.15, 35.69,
46.27,
64.66, 70.07, 74.33, 83.21, 88.34, 117.15, 118.93, 122.13, 124.01, 127.29,
139.54, 139.80,
141.79, 150.91, 151.93, 153.18. m/z (ESI'): 602.2(M +H).
Example 44. Synthesis of compound d-21
, n ,
I
1,1_,*,......,....õ
N\\
.' 0-- N-
Nal3H, .._
<
___________________________________ 8 NY 0 Ch(--
CH3OH,0 C,2h N-
0-c,
0-
¨X --Ab
CI
11.2,1)I
,0 fl Ac0 N N CI
/Ai
01130H CH,COOH=1 4 HCI-EA OAc OAc
,J,,,
Pd/C,H2, rt,16h 0-13 rt 3h HN AGO N-
N-- CI
HCI I-_-01-
XOAc OAc
, i 410
,
r 1 1 ( 00
N N 0 0 CI¨P----P-C1 N
NH,Me0H I X CI' CI
Me0H, rl ' _____ e.--*L---,..t.,N PO(OMe),
H c, N N 1 p-Ts0H ...----,. 1-10-111 N
CI H20 F10-(17)(11,-HO
''¨I-_0
O0
OH OH x OH OH
[00405] A solution of tert-butyl 4-cyanopiperidine-1-carboxylate
(2.0 g, 9.51 mmol, 1
eq.) in THF (30 mL) was cooled to -78 C, followed by addition of LiHMDS (1 M,
11.89 mL,
1.25 eq.) dropvvise at -78 C and stirring the mixture for about 20 min. To
this mixture was
added a solution of 1-(bromomethyl)-2-iodo-benzene (3.11 g, 10.46 mmol, 1.1
eq.) in THF (5
mL). The mixture was stirred at -78 C for 3 h. The reaction was quenched with
saturated
NH4C1, and the mixture was extracted with Et0Ac (2 x 50 mL). The organic layer
was washed
with brine, and concentrated. The residue was purified by column
chromatographyon silica gel
176
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
eluted with PE/EA (100:0-83:17), affording
tert-butyl 4-cyano-4-[(2-
iodophenyOmethyll pip eridine-1-carboxylate (3.7 g, 91.25% yield).
[00406] A solution of tert-butyl 4-cyano-4-[(2-
iodophenyl)methy1lpiperidine-1-
carboxylate (3.7 g, 8.68 mmol, 1 eq.) in THF (30 mL) was cooled to about -78
C, followed by
addition of n-BuLi (2.5 M, 6.94 mL, 2 eq.) dropwise at about -78 C. The
mixture was stirred
for 2 h at this temperature, and the reaction mixture was quenched with
saturated NH4C1. The
mixture was extracted with EA (2 x 80 mL). The organic layer was washed with
brine, and
concentrated. The residue was purified by column chromatographyon silica gel
eluted with
PE/EA (100:0-75:25), affording tert-butyl 1-oxospiro[indane-2,4'-piperidine1-
1'-carboxylate
(1.55 g, 5.14 mmol, 59.25% yield).
1004071 To a solution of tert-butyl I -oxospiro[indane-2,4'-
piperidinel- l'-carboxylate
(1.55 g, 5.14 mmol, 1 eq.) in CH3OH (20 mL) was added NaBH4 (486.40 mg, 12.86
mmol, 2.5
eq.) in portions at 0 C. The mixture was stirred at the temperature for 2 h.
Solvent was removed
by evaporation, and the residue was extracted with Et0Ac (2 x 100 mL). The
organic layer was
washed with brine (100 mL), and concentrated by evaporation, affording tert-
butyl 1-
hydroxyspiro[indane-2,4'-piperidineJ-1'-carboxylate (1.54 g, 5.08 mmol, 98.69%
yield) as
white solid.
[00408] To a solution of tert-butyl 1-hydroxyspiro[indane-2,4'-
piperidine1-1'-carboxylate
(950 mg, 3.13 mmol, 1 eq.) in CH3OH/CH3COOH=1:4 (40 mL) was added Pd/C (200
mg, 1.65
mmol). The mixture was stirred at under H2 gas atmosphere at rt overnight. The
insoluble
material was removed by filtration and washed with Me0H. The filtrate and the
washing were
combined, concentrated by evaporation, neutralized with saturated NaHCO3
(aq.), and
extracted with DCM (2 x 80 mL). The organic layer was washed with brine, and
concentrated.
The residue was purified by column chromatography on silica gel, eluted with
(PE/EA from
100:0 to 90:10), affording tert-butyl spiro[indane-2,4'-piperidinel-r-
carboxylate (190 mg,
21.11% yield).
[00409] tert-Butyl spirofindane-2,4'-piperidinel-1'-carboxylate
(330 mg, 1.15 mmol, 1
eq.) was dissolved in HC1-dioxane (5 mL). After stirred at rt for 3 h, the
mixture was
concentrated and the residue was used directly for the next step.
1004101 To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (466.8 mg, 1.04 mmol, 1 eq.) in 1,4-
dioxane (10.00 mL)
were added spiro[indane-2,4'-piperidin-1 -ium] chloride (257 mg, 1.15 mmol,
1.10 eq.) and
D1PEA (472.14 mg, 3.65 mmol, 636.30 nL, 3.5 eq.). The mixture was stirred at
100 'V for 4
177
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
h, and concentrated by evaporation. The residue was purified by column
chromatography on
silica gel, eluted with PE/EA (100:0-55:45), affording [(2R,3R,4R,5R)-3,4-
diacetoxy-5-(2-
chloro-6-spiro [indane-2,4'-pi peri dine] -1'-y 1-purin-9-y l)tetrahy drofuran-
2-yl]methy 1 acetate
(590 mg, 94.52% yield).
[00411]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro[indane-
2,4'-
piperidine1-11-yl-purin-9-yl)tetrahydrofuran-2-yl]methyl acetate (590 mg,
986.55 [anol, 1 eq.)
in Me0H (8.38 mL) was added Me0H-NH3 (7 M, 3.52 mL, 25 eq.). The mixture was
stirred
at room temperature for 4 h. Solvent was removed by evaporation. The residue
was diluted
with Et0Ac (50 mL), followed by addition of water (40 mL). The organic layer
was
concentrated, giving (2R,3R,4S,5R)-2-(2-chloro-6-spiro [indane-2,4'-
piperidine]
y1)-5 -(hy droxy m ethy l)tetrahy drofuran -3,4-di ol (450 mg, 96.65% yield)
[00412]
To a mixture of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[indane-2,4'-piperidine1-
1-
yl-purin-9-y1)-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (450 mg, 953.52 gmol,
1 eq.) in
acetone (30 mL) were added 2,2-dimethoxypropane (1.49 g, 14.30 mmol, 15 eq.)
and p-Ts0H
(164.20 mg, 953.52 [unol, 1 eq.) . The mixture was stirred at room temperature
for 1 h. Solvent
was removed by evaporation, and the residue was diluted with Et0Ac (50 mL),
washed first
with aqueous Na1-1CO3 and then with brine. The organic layer was concentrated
and the residue
was purified by column chromatographyon silica gel eluted with PE/EA (100:0-
70:30),
affording [(3aR,4R,6R,6aR)-4-(2- chl oro-6-spiro Undane-2,4'-pi p dine] -1'-yl-
purin-9-y1)-2,2-
dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3]dioxol-6-yl[methanol (380 mg,
77.84% yield).
[00413] A solution of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro
yl-purin-9-y1)-2,2-dimethy1-3a,4,6,6a-tetrahy drofuro [3,4-d] [1,3] di oxo1-6-
yll methanol (380
mg, 742.19 gmol, 1 eq) in PO(Me0)3 (5 mL) was cooled to about 0 'C. To the
cold solution
was added a solution of bis(dichlorophosphoryl)methane (556.16 mg, 2.23 mmol,
3 eq.) in
PO(Me0)3 (4 mL). The mixture was stirred at rt for 5 h, followed by addition
of water (4 mL).
The stirring was continued at room temperature overnight. The mixture was
injected directly
into the column (C18 reversed phase silica gel) and eluted with ACN in water
(0 to 30%
gradient), giving Compound d-21, (66.7 mg, 14.01% yield, 98.23% purity): 11-1
NMR (500
MHz, Me0D) 6 ppm1.72 (d, J = 5.0 Hz, 4H), 2.50 (t, J = 20.9 Hz, 2H), 2.89 (s,
4H), 4.23 ¨
4.45 (m, 5H), 4.60 (t, J = 4.9 Hz, 1H), 6.00 (d, J = 4.8 Hz, 1H), 7.05 ¨ 7.13
(m, 2H), 7.14 ¨
7.22 (m, 2H), 8.29 (s, 1H). 31P NMR (203 MHz, Me0D) 6 ppm 16.87, 20.09. 13C
NMR (126
MHz, Me0D) 6 ppm 26.11, 36.57, 42.21, 44.08, 64.73, 70.07, 74.28, 83.17,
88.14, 118.20,
124.42, 126.01, 137.83, 141.79, 151.67, 153.65. m/z (ES1 ):630.3(M +H).
178
CA 03200232 2023- 5- 25

WO 2022/109741 PCT/CA2021/051688
Example 45. Synthesis of compound d-22
Act)
Br DLAIH4 is OAc OAc NH3-Me0H
THFU-H5M070 =..y 5h -
THF,70 C,3h DIPEA, Dioxane Me0H, r1,3h
rt,1h,70"C,3h 100 C, 4 HO
Ac0 NN CI
OH OH
OAc OAc
(0, =
0 0
p-Ts0H CI' CI
acetone,rt HO-1
,3h 4..N,,, NXt.NJ, 1) P0(0Me)3, 0'C,5h 9
2) H,0,40 C,40rnin HO 6H H-0 N CI
rt,O/N
Ox0
OH OH
[00414] To a mixture of indolin-2-one (3 g, 22.53 mmol, 1 eq.) in THF (50
mL) under
nitrogen atmosphere was added dropwise LiHMDS (1 M, 49.57 mL, 2.2 eq.) at -78
C. After
the temperature was raised to -50 C and kept at this temperature for 30 mm,
the mixture was
further cooled to -78 'V, followed by addition of 1,6-dibromohexane (5.50 g,
22.53 mmol, 1
eq.) in THF (20 ml). The mixture then was stirred at rt for lh, at reflux for
5 h, and at rt
overnight. The mixture was quenched and evaporated under reduced pressure and
the residue
was partitioned between Et0Ac and saturated NH4C1. The organic layer was
concentrated and
the residue was purified by column chromatography on silica gel eluted with
(PE/EA from
100:0 to 80:20), giving spiro[cycloheptane-1,3'-indoline]-2'-one (880 mg, 4.09
mmol, 18.14%
yield) as a yellow solid.
[00415] To a solution of spiro[cycloheptane-1,3'-indoline1-2'-one (1.06 g,
4.92 mmol, 1
eq.) in THF (20 mL) was added LiA1H4 (373.70 mg, 9.85 mmol, 2 eq.). The
mixture was stirred
at 70 C for 4 h, and then the reaction was quenched with 15% NaOH (aqueous, 5
mL) at 0 C,
followed by addition of Et0Ac (20 mL). The organic layer was separated, and
dried over
MgSO4. The insoluble material was removed by filtration, and the filtrate was
concentrated.
The residue was purified by column chromatography on silica gel eluted with
PE/EA (100:0 to
90:10), affording spiro[cycloheptane-1,3'-indoline] (210 mg, 1.04 mmol, 21.19%
yield).
[00416] To a mixture of spiro[cycloheptane-1,3'-indoline] (210 mg, 1.04
mmol, 1.2 eq.)
in 1,4-dioxane (20 mL) were added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yptetrahydrofuran-2-y1Jmethyl acetate (388.78 mg, 869.32 limo', 1 eq) and DIEA
(280.88 mg,
2.17 mmol, 378.54 !.IL, 2.5 eq.). The mixture was stirred at 100 C for 4 h.
Solvent was removed
by evaporation. The residue was diluted with Et0Ac (40 mL), washed first with
water and then
with brine. The organic layer was concentrated and the residue was purified by
column
179
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
chromatographyon silica gel eluted with PE/FA (100:0-60:40), affording
[(2R,3R,4R,5R)-3,4-
di acetoxy-5-(2-chl oro-6-spiro [cycloheptane-1,31-indolinel -11-y1 -purin-9-y
Otetrahy drofuran-2-
yll methyl acetate (310 mg, 506.48 mmol, 58.26% yield) as yellow solid.
[00417] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-
Spiro [cycloheptane-1,3'-indolinel -1'-yl-purin-9-yl)tetrahydrofuran-2-
yllmethyl acetate (310
mg, 506.48 iamol, 1 eq.) in Me0H (3 mL) was added NH3-Me0H (7 M, 2.17 mL, 30
eq.) . The
mixture was stirred at room temperature for 3 h. Solvent was removed by
evaporation, and the
residue was extracted with Et0Ac (30 mL * 2), followed by addition of H20 (30
mL). The
organic layer was separated, washed with brine, dried over Na2SO4, and
concentrated,
giving (2R,3R,4S ,5R)-2-(2-chloro-6-spiro [cycloheptane-1,3'-indolinel-l'-yl-
purin-9-y1)-5-
(hydroxymethyl) tetrahydrofuran-3,4-diol (246 mg, 506.21 tamol, 99.95% yield)
as white solid.
[00418] To a solution
of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[cycloheptane-1,3'-
indolinel-1'-yl-purin-9-y1)-5-(hydroxymethyptetrahydrofuran-3,4-diol (246 mg,
506.21 lAmol,
1 eq.) in acetone (20 mL) were added P-Ts0H-1-120 (97.19 mg, 506.21 mmol, 1
eq.) and 2,2-
dimethoxypropane (1.05 g, 10.12 mmol, 1.24 mL, 20 eq.). The mixture was
stirred at rt for 3
h. Solvent was removed by evaporation and the residue was diluted with EA (50
mL), washed
first with aqueous Na1-1CO3 and then with brine, and concentrated. The residue
was purified by
column chromatographyon silica gel eluted with PE/EA (100:0-70:30),
giving [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cycloheptane-1,3'-indoline1-1'-yl-
purin-9-y1)-
2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-d][1,3]dioxol-6-yl[methanol (213 mg,
404.92
mmol, 79.99% yield) as light green solid.
[00419] A solution
of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[cycloheptane-1,3'-
indoline] -1'-y 1 -purin-9-y1)-2,2-dimethy1-3 a,4,6,6 a-tetrahy drofuro [3,4-
d] [1,3] di oxo1-6-
yllmethanol (213 mg, 404.92 jamol, 1 eq.) in P0(0Me)3 (3 mL) was cooled to 0
C, followed
by addition of a solution of bis(dichlorophosphoryl)methane (303.43 mg, 1.21
mmol, 3 eq.) in
PO(Me0)3 ( 3 mL). The mixture was stirred at 0 C for 2.5 h. LCMS monitoring
indicated that
the mixture contained mainly starting material. The mixture was warmed to rt
and stirred for 2
h. LC-MS showed that about 65% starting material present. More
bis(dichlorophosphoryl)methane (101 mg, 0.403 mmol, 1 eq.) in PO(Me0)3 (1 mL)
was added
to the mixture at 0 C, and the mixture was stirred at rt overnight. At this
point, very little
starting material was observed by LC-MS monitoring. The mixture was cooled to
0 C,
followed by addition of water (5 mL), and continued with stirring at rt
overnight. Purification
of this reaction mixture using prep-HPLC provided sodium salt of the product.
The sodium salt
180
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mixture was acidified with resin (acidic form) and lyophilized, giving
Compound d-22, (76.6
mg, 117.26 [tmol, 29.00% yield, 98.58% purity): 11-1 NMR (500 MHz, Me0D) 6ppm
1.73 -
1.96 (m, 12H), 2.56 (t, J = 20.9 Hz, 2H), 4.30 - 4.39 (m, 2H), 4.41 (s, 1H),
4.50 (t, J = 4.6 Hz,
1H), 4.53 - 4.60 (m, 2H), 4.70 (1, J = 4.7 Hz, 1H), 6.10 (d, J = 4.8 Hz, 1H),
7.10 (1, J = 7.4 Hz,
1H), 7.23 (t, J = 7.8 Hz, 1H), 7.33 (d, J = 7.4 Hz, 1H), 8.45 (d, J = 8.1 Hz,
1H), 8.49 (s, 1H).
3113 NMR (203 MHz, Me0D) 6ppm 16.84, 19.95. NMR (126 MHz, Me0D) 6ppm
15.90,
23.47, 25.10, 26.16, 27.21, 29.19, 40.18, 56.14, 62.76, 64.68, 70.11, 74.36,
83.27, 88.21,
117.44, 122.19, 124.02, 126.92, 139.53, 141.39, 143.27, 27.21, 151.31, 152.09,
153.25. m/z
(ESL):644.3(M +H).
Example 46. Synthesis of compound d-23
cr.CN. = LDA CN LIAIH4 NH2
TosCl 41HTs 40 m en 10- CN
Pt BBAL , 012
, H35 C,16h KII-NTS
'Br THF.-78C THF,70"C EV,I,DCM
0
Ac0 N CI
N
1-113r/1-10Ac 0Ar OAc
0.Cro loxane F.11,iDcill
40 C,16h DIPEA D A00-1c1._ I mNe1-13-HMet03Hh
Ho
N CI
100 C 4 h
OAc
OH OH
0 0
p-Ts0H <201,01 -
acetone,rt,2h hOj12)) H0
P0(047)z,400 C:5h 'N CI
FT
Ox0 rt',0'/N OH OH
[00420] A solution of cyclohexanecarbonitrile (5.0 g, 45.80
mmol, 1 eq.) in 'THF (50
mL) was cooled to -78 C, followed by addition of LDA (1 M, 50.38 mL, 1.1
eq.). The mixture
was warmed to rt and stirred at RT for 1 h, and then cooled to -78 C. To this
cold mixture was
added 2-bromoethylbenzene (10.17 g, 54.96 mmol, 1.2 eq.) in THF (20 mL), and
the mixture
was stin-ed at rt overnight. The reaction was quenched with saturated NH4C1,
and the mixture
was extracted with Et0Ac (80 mL * 2). The organic layer was washed with brine
and
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/EA (100:0-95:5), affording 1-(2-phenylethyl)cyclohexanecarbonitrile (4.0 g,
18.75 mmol,
40.94% yield) as yellow oil.
[00421] To a solution of 1-(2-
phenylethyl)cyclohexanecarbonitrile (4.0 g, 18.75 mmol, 1
eq.) in THF (80 mL) was added. The mixture was cooled to 0 C, followed by
addition of
LiA1H4 (2.13 g, 56.25 mmol, 3 eq.). This mixture was stirred at 70 C
overnight, and the
181
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
reaction was quenched with 15% NaOH at 0 C until the pH reached about 10. The
organic
layer was dried over MgSO4. Solid material was removed by filtration, and the
filtrate was
concentrated, affording [1-(2-pheny1ethy1) cyclohexyllmethanamine (4.0 g,
18.40 mmol,
98.15% yield). The crude product was used directly in next step.
[00422] To a mixture of [1-(2-phenylethyl)cyclohexyllmethanamine
(2.0 g, 9.20 mmol,
1 eq.) in dry DCM (30 mL) were added Et3N (1.12g. 11.04 mmol, 1.53 mL, 1.2
eq.) and 4-
methylbenzenesulfonyl chloride (1.93 g, 10.12 mmol, 1.92 mL, 1.1 eq.) at 0 C.
The mixture
was stirred at room temperature overnight. The mixture was quenched with
saturated NH4C1,
and extracted with DCM (2 x 30 mL). The organic layer was washed with brine,
and
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/EA (100:0-90:10), affording 4-methyl-
N4[1-(2-
phenylethyl)cyclohexyl]methyl]benzenesulfonamide (2.4 g, 6.46 mmol, 70.20%
yield) as
white solid.
[00423] 4-Methyl-N-((1-
phenethylcyclohexyl)methyl)benzenesulfonamide (2.2 g, 5.92
mmol, 1 eq.), m-CPBA (1.53 g, 8.88 mmol, 1.5 eq.) and 12 (225.44 mg, 888.21
[imol, 0.15 eq.)
were dissolved in CH3CN/t-BuOH (1:1, 40 mL). The mixture was evacuated,
backfilled with
nitrogen, and then stirred at 35 C overnight. TLC indicated that starting
material was mostly
left. The stirring was continued at 50 C for 6 h, and the reaction was
quenched with Na2S203.
After the pH was adjusted to 8 with NaHCO3 (aq.), the mixture was extracted
with Et0Ac (2
x 30 mL). The organic layer was washed with brine and concentrated. The
residue was
purified by column chromatographyon silica gel eluted with PE/EA (100:0-
85:15), affording
3-phenyl-2-tosy1-2-azaspiro[4.51decane (900 mg, 2.44 mmol, 41.13% yield) with
recovery of
starting material (1.0 g).
[00424] A mixture of 3-pheny1-2-(p-tolylsulfony1)-2-
azaspirop.5]decane (1.0 g, 2.71
mmol, 1 eq.) in HBr/HOAc (15 mL) was stirred at 40 C for 20 h, followed by
removal of
HBr/HOAc on an oil pump. To the remaining mixture was slowly added 15% NaOH
(aq.) at 0
C until the pH of the mixture reached 10. The pH adjusted mixture was
extracted with ElOAc
(20 mL * 2). The extract was washed with brine and concentrated. The residue
was purified by
column chromatography on silica gel eluted with DCM/Me0H (100:0-90:10),
affording 3-
pheny1-2-azaspiro[4.51decane (250 mg, 1.16 mmol, 42.90% yield).
[00425] To a mixture of 3-phenyl-2-azaspiro[4.51decane (250 mg,
1.16 mmol, 1.4 eq.)
in 1,4-di ox an e (15 mL) were added [(2R,3R,4R,5R)-3,4-di acetoxy
chloropuri
yOtetrahydrofuran-2-yl]methyl acetate (370.88 mg, 829.28 umol, 1 eq.) and DlEA
(375.12 mg,
182
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
2.90 mmol, 505.55 p.L, 3.5 eq.). The mixture was stirred at 100 C for 4 h.
The solvent was
removed by evaporation. The residue was diluted with Et0Ac (30 mL), washed
first with water
and then with brine. The organic layer was concentrated and the residue was
purified by column
chromatographyon silica gel eluted with PE/EA (100:0-60:40), affording
[(2R.3R,4R,5R)-3,4-
di acetoxy-5- [2-chl oro-6-(3 -pheny1-2-azaspiro [4. 51 decan-2-yl)purin-9-
yl]tetrahydrofuran-2-
yl]methyl acetate (440 mg, 702.76 timol, 84.74% yield) as white solid.
[00426]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(3-phenyl-2-
azaspiro[4.51decan-2-yl)purin-9-yl]tetrahydrofuran-2-yl]methyl acetate (440
mg, 702.76
pmol, 1 eq.) in Me0H (4 mL) was added NH3-Me0H (7 M, 3.01 mL, 30 eq.) . The
mixture
was stirred at room temperature for 3 h. The solvent was removed and the
residue was extracted
with Et0Ac (20 mL * 2). The extract was washed with brine, dried with Na2SO4,
and
concentrated, affording (2R,3R,4 S,5R)-2- [2-chl oro-6-(3 -phenyl-2-az aspiro
[4. 51 decan-2-
yl)purin-9-y11-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (350 mg, 700.02
timol, 99.61%
yield) as white solid.
[00427]
To a solution of (2R,3R,4S,5R)-242-chloro-6-(3-phenyl-2-azaspiro[4.51decan-
2-yl)purin-9-y1[-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (350 mg, 700.02
pmol, 1 eq.)
in Acetone (20 mL) was added p-Ts0H-H20 (134.4 mg, 700.02 timol, 1 eq.) , 2,2-
climethoxypropane (1.46 g, 14.00 mmol, 1.72 mL, 20 eq.). The mixture was
stirred at 0 C for
2 h. TLC showed SM was consumed. The mixture was adjusted PH about 9 by 15%
NaOH
(aq.) slowly at 0 C. The solvent was removed by evaporation and the residue
was diluted with
Et0Ac (30 mL), washed by aq.NH4C1, followed by brine and the the organic layer
was
concentrated and the residue was purified by column chromatographyon silica
gel eluted with
PE/EA (100:0-50:50) to
afford [(3aR,4R,6R,6aR)-442-chl oro-6-(3 -pheny1-2-
azaspiro [4. 5] decan-2-yOpurin-9-y11-2,2-dimethyl-3 a,4,6,6a-tetrahy drofuro
[3,4-d] [1,3] di oxol-
6-yl]methanol (343 mg, 635.12 ttmol, 90.73% yield) as white solid.
1004281
A solution of [(3aR,4R,6R,6aR)-4-1-2-chloro-6-(3-phenyl-2-
azaspiro[4.51decan-
2-yOpurin-9-y11-2,2-dimethyl-3a,4,6,6a- tetrahy drofuro [3,4-d] [1,3] dioxo1-6-
yl]melhanol (343
mg, 635.12 timol, 1 eq.) in P0(0Me)3 (4 mL) was cooled to about 0 C, followed
by addition
of bis(dichlorophosphoryl)methane (317.29 mg, 1.27 mmol, 2 eq.) in PO(Me0)3 (
4 mL). The
mixture was stirred at 0 C for 4 h and at rt for 2 h, and cooled to 0 C,
followed by addition of
bis(dichlorophosphoryl)methane (158.65 mg, 0.635 mmol, 1 eq) in PO(Me0)3 ( 2
mL). The
mixture was stirred at rt overnight. Water (7 mL) was slowly added into the
mixture at 0 C,
and the mixture was stirred at 40 C for 40 min, then was stirred at rt
overnight. The reaction
183
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mixture was purified by prep-HPLC, affording sodium salt of the product. The
sodium salt
mixture was acidified (ionic resin, acidic form) and the resultant solution
was lyophilized,
giving a crude product in dark color. The crude material was purified using
C18 reversed phase
silica gel (0-50% ACN in Water), giving Compound d-23, (88.0 mg, 133.17 nmol,
21.01%
yield, 99.57% purity); 1-1-1 NMR (500 MHz, Me0D) oppm 1.47 ¨ 1.83 (m, 12H),
2.54 (t, J =
21.1 Hz, 2H), 4.36 (dd, J = 55.5, 27.1 Hz, 5H), 4.63 (s, 1H), 4.75 (d, J =
10.8 Hz, 1H), 5.37 (s,
1H), 6.02 (s, 1H), 7.24 (d, J = 41.5 Hz, 5H), 8.49 (s, 1H). 31-13 NMR (203
MHz, Me0D) 6ppm
17.09, 20.02. 1-3C NMR (126 MHz, Me0D) 6ppm 22.54, 23.49, 25.03, 25.87, 26.10,
27.15,
33.91, 35.90, 42.78, 61.74, 64.62, 69.99, 74.28, 83.20, 88.30, 118.13, 125.81,
126.25, 127.98,
138.87, 143.63, 151.07, 153.11, 153.67. m/z (ESI+): 658.3(M +H).
Example 47. Synthesis of compound d-26
ZOO'C 20h- 'N'7":0 THFL:DCSO Tõf. MeUH,60 o!
CX1H
OK-
I I
0
OAc OAc NN
N-
I,
NXLN NH,Me0H _Tr
AO FlOj N P-- ci_IL or :4,
17õ
WON, A, a HO N,L,CI rt 1) P0,0Etki3O C,5h HO 0-1 N CI
2) H:O4UCOr
OH OH rtOiN OHO
OAc OAc x0
[00429]
To a mixture of isochromane-1,3-di one (1 g, 6.17 mmol, 1 eq.) in toluene
(30
mL) was added phenylmethanamine (793.03 mg, 7.40 mmol, 1.2 eq.). The mixture
was stirred
at 110 C for 20 h, and then was concentrated. The residue was purified by
column
chromatography on silica gel eluted with PE/EA (100:0 to 75:25), giving 2-
benzy1-4H-
isoquinoline-1,3-dione (0.9 g, 3.58 mmol, 58.07% yield) as white solid.
[00430] To a mixture of 2-benzy1-4H-isoquinoline-1,3-dione (780
mg, 3.10 mmol, 1
eq.) in THF (8 mL) under nitrogen atmosphere was added dropwise LiHMDS (1 M,
6.83 mL,
2.2 eq.) at -78 C. The temperature of the reaction was raised to -50 C, kept
at this temperature
for 30 min, and then cooled to -78 C, followed by addition of 1,5-
dibromopentane (713.76
mg, 3.10 mmol, 1 eq.) in THF (8 mL). After completion of addition, the mixture
was stirred at
rt for 1 h, and then at reflux overnight. The mixture was quenched with
saturated NH4C1 and
extracted with Et0Ac (40 mL * 2). The organic layer was washed with brine and
concentrated.
The residue was purified by column chromatography on silica gel eluted with
(PE/EA from
100:0 to 90:10) to give 2'-benzylspirolcyclohexane-1,4'-isoquinoline]-1',3'-
dione (160 mg,
500.95 nmol, 16.14% yield).
184
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00431]
A solution of A1C13 (350.68 mg, 2.63 mmol, 4 eq.) in THF (10 mL) was
cooled
to 0 C, followed by addition of LiA1H4 (149.71 mg, 3.94 mmol, 6 eq.). The
mixture was stirred
at 0 C for 30 min, followed by addition a solution of 2'-
benzylspiro[cyclohexane-1,4'-
isoquinoline1-1',3'-dione (210 mg, 657.49 wnol, 1 eq.) in THF (2 mL). The
mixture was stirred
at ft for 6 h, diluted by THF (10 mL), and then quenched by through slow
addition of 15%
aqueous NaOH at 0 C until the pH of the mixture reached 10. The organic layer
was separated
and dried over MgSO4. Insoluble material was removed by filtration, and the
filtrate was
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/EA (100:0-90:10), affording 2-benzyl spiro [1,3-dihy droisoquinoline-4,1' -
cy clohexane]
(190 mg, 651.96 [imol, 99.16% yield).
1004321
To a solution of 2-ben zyl spiro[1,3-dihydroisoquinoline-4,1'-cycl
hexane] (190
mg, 651.96 wnol, 1 eq.) in Me0H (5 mL) were added HCOONH4 (61.67 mg, 977.94
wnol,
1.5 eq.) and Pd(OH)2 (20 mg, 134.51 wnol, 20% on carbon with 50% water). The
mixture was
stirred at under H2 atmosphere at 60 C overnight. Insoluble material was
removed by
filtration, and washed with Me0H. The filtrate and the washing was combined
and
concentrated to dryness, affording spiro[2,3-dihydro-1H-isoquinoline-4,1'-
cyclohexane] (120
mg, 596.11 timol, 91.43% yield), which was used directly in the next step.
[00433] To a mixture of s piro 12,3 -dihy dro-1H-i s o quinoline-
4,11- cy clohexane] (120 mg,
596.11 iumol, 1.3 eq.) in 1,4-dioxane (15 mL) were added [(2R,3R,4R,5R)-3,4-
diacetoxy-5-
(2,6-dichloropurin-9-y1) tetrahydrofuran-2-yl] methyl acetate (205.07 mg,
458.54 wnol, 1 eq.)
and DIPEA (148.16 mg, 1.15 mmol, 199.67 L, 2.5 eq.). The mixture was stirred
at 100 'V for
2 h. After removal of solvent (evaporation), the residual material was diluted
with Et0Ac (50
mL), washed first with water and then with brine. The organic layer was
concentrated and the
residue was purified by column chromatography on silica gel eluted with PE/EA
(100:0-50:50),
providing
[(2R,3R,4R,5R)-3,4-di acetoxy -5 -(2 -chloro-6-s piro [1,3-dihy droi s o
quinoline-4,1'-
cy cl ohexanel -2-yl-purin-9-yptetrahydrofuran-2-ylimethyl acetate (280 mg,
457.46 wnol,
99.76% yield) as white solid.
[00434]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2-chloro-6-spiro[1,3-
dihy droi s oquinoline-4,1'-cy cl ohexane] -2-yl-purin-9-yl)tetrahy drofuran-2-
yll methyl acetate
(280 mg, 457.46 tamol, 1 eq.) in Me0H (3 mL) was added NH3-Me0H (7 M, 1.96 mL,
30 eq.).
The mixture was stirred at room temperature for 2 h. Solvent was removed by
evaporation and
the residue was extracted with Et0Ac (30 mL * 2). The extract was washed with
brine, dried
with N a2S 04, and concentrated, affording (2R,3R,4 S,5R)-2-(2-chl oro-6-s
piro [1,3-
185
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
dihy droi s oquinoline-4,1'-cy cl ohexane] -2-yl-purin-9-y1)-5 -(hy
droxymethyl)tetrahydrofuran-
3,4-diol (220 mg, 452.71 nmol, 98.96% yield) as white solid.
[00435]
To a solution of (2R,3R,4S,5R)-2-(2-chloro-6-spiro[1,3-dihydroisoquinoline-
4,1'-cy cl hexane] -2-yl-purin-9-y1)-5-(hy droxy methyl) tetrahy drofuran-3,4-
diol (220 mg,
452.71 wnol, 1 eq.) in acetone (15 mL) were added p-Ts0H-H20 (86.11 mg, 452.71
wnol, 1
eq.) , 2,2-dimethoxypropane (942.97 mg, 9.05 mmol, 1.11 mL, 20 eq.). The
mixture was stirred
at rt for 2 h followed by pH adjustment to 9 with slow addition of 15% NaOH
(aq.) 0 C.
Solvent was removed by evaporation and the residue was extracted with Et0Ac
(30 mL * 2).
The extract was washed with brine, and concentrated to dryness. The residue
was purified by
column chromatography on silica gel eluted with PE/EA (100:0-50:50), giving
[(3aR,4R,6R,6aR)-4-(2-chl oro-6-spiro[ I ,3-dihydroi soquinol in e-4,1'-cy cl
oh exan el-2-y1
9-y1)-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro 113 ,4-d] [1,3] dioxo1-6-yl]
methanol (200 mg, 380.21
nmol, 83.99% yield) as white solid.
[00436]
To a cold solution (0 C) of [(3aR,4R,6R,6aR)-4-(2-chloro-6-spiro[1,3-
dihydroisoquinoline-4,1'-cyclohexane1-2-yl-purin-9-y1)-2,2-dimethyl-3a,4,6,6a-
tetrahydrofuro[3,4-d][1,3]dioxol-6-yl]methanol (200 mg, 380.21 nmol, 1 eq.) in
P0(0E03 (2
mL) was added bis(dichlorophosphoryl)methane (189.94 mg, 760.42 nmol, 2 eq.)
in P0(0E03
( 2 mL). The mixture was stirred at 0 C for 5 h, followed by slow addition of
water (3 mL) at
0 C. The mixture was stin-ed at 40 C for 40 min, and at rt overnight.
Purification of the
reaction mixture using C18 reversed phase silica gel (0-25% ACN in Water) gave
Compound
d-26, (82 mg, 126.21 nmol, 33.21% yield, 99.11% purity); 1H NMR (500 MHz,
Me0D) oppm
1.38 (s, 1H), 1.63 (s, 4H), 1.82 (dd, J = 31.4, 17.4 Hz, 5H), 2.54 (t, J =
20.2 Hz, 2H), 4.21-
4.43 (m, 4H),4.46 (s, 1H),4.66 (s, 1H), 4.79 (s, 2H), 5.64 (s, 1H), 6.05 (s,
1H), 7.17 ¨7.27 (m,
3H),7.48 (d, J = 7.5 Hz, 1H), 8.38 (s, 1H). 3113 NMR (203 MHz, Me0D) Sppm
17_04, 19.68.
1-3C NMR (126 MHz, Me0D) 6ppm 21.91, 25.01, 25.69, 26.05, 34.91, 35.30, 38.67,
64.73,
70.15, 74.27, 83.20, 88.08, 125.78, 126.56, 132.30, 138.23, 143.85, 151.78,
153.64. miz
(ESL): 644.20(M +H).
Example 48. Synthesis of compound d-29
186
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
fl
a [ 1 oõo
<,5,1 I '0 211' A
D,oxa,,eD, NI-12-Me0H N -sN
Ts0H-H0 2 <14-1jj
-at Aco Me0H, rt, 16h
HO MeCOMe, H,
leg¨ HO N-
OAc OAc
OAc OAc OH OH
0
CI
(CH20)200, 0"C, 16h
HO
OH OH
1004371 To
a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-y11 methyl acetate (1 g, 2.24 mmol, 1 eq.) and 2-
phenylpiperidine (432.65
mg, 2.68 mmol, 1.2 eq.) in 1,4-dioxane (10 mL) was added DIEA (722.45 mg, 5.59
mmol,
973.66 mL, 2.5 eq.). The mixture was reflux for 16 hours. After complete
conversion, the
reaction mixture was taken up in Et0Ac (30 mL). The mixture was washed with
brine,
concentrated in vacuo. The residual material was purified by column
chromatography on silica
gel (eluent, 0 to 30% EA in PE), giving [(2R,3R,4R,5R)-3,4-diacetoxy-5-1-2-
chloro-6-(2-
phenyl -1 -piperi dyl)purin-9-yl[tetrahy drofuran-2-yl[methyl acetate (1 g,
78.19%).
[00438] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-
chloro-6-(2-phenyl-l-
piperidyl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (1 g, 1.75 mmol, eq.)
in Me0H (5
mL) was added NH3-Me0H (7 M, 5 mL, 20.02 eq.). The mixture was stirred at rt
for 16 hours,
and concentrated by evaporation. The residue was purified by column
chromatography on
silica gel (eluent, 0 to 5% Me0H in DCM), affording (2R,3R,4S,5R)-242-chloro-6-
(2-phenyl-
1-pip eri dyl)purin-9-y11-5 -(hy droxy methyl)tetrahy drofuran-3,4-di ol (700
mg, 89.80%) .
[00439] To a mixture of (2R,3R,4S,5R)-242-chloro-6-(2-pheny1-1-
piperidyl)purin-9-
yll -5-(hydroxymethyl)tetrahydrofuran-3,4-diol (700 mg, 1.57 mmol, 1 eq.) and
Ts0H-H20
(358.31 mg, 1.88 mmol, 1.2 eq.) in acetone (10 mL) was added 2,2-
dimethoxypropane (1.63
g, 15.70 mmol, 10 eq.). The mixture was stirred at rt for 16 h. The mixture
was concentrated
and the residue was taken up in Et0Ac (50 mL), washed with aqueous NaHCO3 (2 x
20 mL).
The organic layer was concentrated and the residue was purified by column
chromatography on
silica gel eluted with (PE/EA from 100:0 to 70:30), affording [(3aR,4R,6R,6aR)-
4-[2-chloro-
642-phenyl-I -piperi dyl)purin-9-yll -2,2-dimethy1-3a,4,6,6a-tetrahy drofuro
[3,4-d] [1,3] di oxol-
6-yllmethanol (650 mg, 85.20%).
187
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00440] To a solution of 1(3aR,4R,6R,6aR)-442-chloro-6-(2-phenyl-
1-piperidyl)purin-
9-y11-2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro [3,4-d] [1,3] dioxo1-6-yl]
methanol (200 mg, 411.55
wnol, 1 eq.) in PO(Me0)3 (2 mL) at 0 C was added
bis(dichlorophosphoryl)methane (205.60
mg, 823.11 nmol, 2 eq.) in P0(0Me)3 (1 mL). The mixture was stirred for 16
hours at rt. The
reaction was quenched with H20 (5 mL), and the mixture was purified using C18
reversed
phase silica gel (0-20% ACN in water), giving Compound d-29, (60 mg, 24.14%),
1H NMR
(500 MHz, CD30D) 6 ppm 1.61-1.72 (m, 4H), 2.44-2.59 (m, 3H), 4.25-4.41 (m,
4H), 4.58-
4.62 (m, 1H), 6.01-6.02 (m, 1H), 7.23-7.24 (m, 1H), 7.27-7.28 (m, 2H)õ7.33-
7.35 (m. 2H),
8.25 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 19.31, 24.95, 25.56 ,26.05,
27.38, 64.78,
70.10, 74.26, 83.10, 88.10, 118.29, 126.36, 128.38, 137.87, 139.23, 151.95,
153.80, 154.68;
3 IP NMR (203 MHz, CD30D) 6 ppm 16.66, 20.04; miz (EST-): 601.9 (M - H).
Example 49. Synthesis of compound d-30
F T C-1
¨
0õ0
H
DIEA
AO DiOXane, 120 C, 16h Aco_4:NX.A1 :1,,, MN:03H-M,
H,1H6h (14NX:..:Al M e eTsZe H, rt, lb6 ' HO <:--C.-XCI
¨L.24
OAe
OAc OAc 0 0
OAc OH OH
CI CI
c.3
(CH 11 30),P0, 0 C, 16 9
14 CI
OH OH
[00441] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yl)tetrahydrofuran-2-yl]methyl acetate (1 g, 2.24 mmol, 1 eq.) and 3-
phenylpiperidine (432.65
mg, 2.68 mmol, 1.2 eq.) in 1,4-dioxane (10 mL) was added DIEA (722.45 mg, 5.59
mmol,
973.66 !IL, 2.5 eq.). The mixture was reflux for 16 hours. After completion of
the reaction, the
mixture was taken up in Et0Ac (30 mL), washed with brine, and concentrated in
vacuo. The
residual material was purified by column chromatography on silica gel (0 to
30% Et0Ac in
Pet-ether), giving [(2R,3R,4R,5R)-3,4-di acetoxy -5- [2-chl oro-6-(3 -phenyl-l-
piperi dyl)purin-
9-yl]tetrahydrofuran-2-yl]methyl acetate (1 g, 78.19%).
[00442] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-1_2-
chloro-6-(3-pheny1-1-
piperidyl)purin-9-yl]tetrahydrofuran-2-yl]methyl acetate (1 g, 1.75 mmol, 1
eq.) in Me0H (5
mL) was added NH3-Me0H (7 M, 5 mL, 20.02 eq.). The mixture was stirred at rt
for 16 hours.
After completion of the reaction, the mixture was concentrated (evaporation).
The residual
188
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
material was purified by column chromatography on silica gel (0 to 5% Me0H in
DCM),
giving (2R,3R,4S ,5 R)-2{2-chl oro-6-(2-pheny1-1 -pip eri dyl)purin-9-yl] -5 -
(hydroxymethyptetrahydrofuran-3,4-diol (600 mg, 76.97%) .
[00443]
To a mixture of (2R,3R,4S,5R)-242-chloro-6-(3-pheny1-1-piperidyl)purin-9-
y1]-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (600 mg, 1.35 mmol, 1 eq.) and
Ts0H-H20
(307.12 mg, 1.61 mmol, 1.2 eq.) in acetone (10 mL) was added 2,2-
dimethoxypropane (1.40
g, 13.46 mmol, 10 eq.). The mixture was stirred at rt for 16 h, and then
concentrated. The
residue was taken up in Et0Ac (50 mL), and washed with aqueous NaHCO3 (20 mL *
2). The
organic layer was concentrated and the residue was purified by column
chromatography on
silica gel (eluent, PE/EA, 100:0 to 70:30), affording [(3aR,4R,6R,6aR)-442-
chloro-6-(3-
ph enyl - I -pi peri dyl)purin-9-yl] -2,2-di m ethyl -3 a,4,6,6a-tetrahy
drofuro [3,4-d] [ I ,3] di oxol -6-
yllmethanol (550 mg, 84.11%).
[00444]
To a solution of 1(3aR,4R,6R,6aR)-4-[2-chloro-6-(3-pheny1-1-
piperidyl)purin-
9-yl] -2,2-dimethy1-3 a,4,6,6a-tetrahy drofuro 13 ,4-d] [1,3] dioxo1-6-yl]
methanol (200 mg, 411.55
1 eq.) in PO(Me0)3(2 mL), at 0 C, was added bis(dichlorophosphoryOmethane
(205.60
mg, 823.11 wriol, 2 eq.) in P0(0Me)3 (1 mL). The mixture was stirred for 16 h
at rt. The
reaction was quenched with H20 (5 mL), and purified using C18 reversed phase
silica gel (0
to 20% ACN in water), giving Compound d-30, (100 mg, 40.24%); 1H NMR (500 MHz,
CD30D) 6 ppm: 1.70-1.72 (m, 1H), 1.89-1.91 (m, 2H), 2.04-2.06 (m, 1H), 2.45-
2.54 (m, 2H),
2.77-2.79 (m, 1H), 4.24-4.41 (m ,4H), 4.59-4.61 (m, 1H), 6.00-6.02 (m, 1H),
7.21-7.31 (m,
5H), 8.27 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm: 25.01, 25.46, 26.05, 27.11,
31.58,
42.90, 64.76, 70.07, 75.25, 83.06, 88.12, 118.30, 126.33, 126.83, 128.19,
137.84, 143.19,
151.72, 153.55, 153.70; 31P NMR (203 MHz, CD30D) 6 ppm:16.70, 20.04; m/z (ESI-
): 602.0
(M-H).
Example 50. Synthesis of compound d-31
189
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
:.)
_ _ .= .;Ji
.D
1.=
ii
3[1.:
- -
_ . , .
:
[00445]
To a mixture of [(2R,3R,4R,5R)-3,4-di acetoxy -5 -(2,6-di chl oropurin-9-
yptetrahydrofuran-2-yll methyl acetate (1 g, 2.24 mmol, 1.0 eq.), 4-
phenylpiperidine (432.65
mg, 2.68 mmol, 1.2 eq.) in 1,4-dioxane (15 mL) was added DIPEA (722.45 mg,
5.59 mmol,
973.66 !IL, 2.5 eq.). The mixture was stirred at 100 C overnight. The mixture
was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/EA (100:0-50:50), affording 1(2R,3R,4R,5R)-3 ,4-di acetoxy -5- [2-chl oro-6-
(4 -phenyl-1-
piperidyl)purin-9-ylJtetrahydrofuran-2-y1Jmethyl acetate (1.1 g, 86.0% yield)
as brown solid.
[00446]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(4-pheny1-1-
piperidyl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (1.1 g, 1.0 eq.) in
Me0H (7 mL) was
added NH3-Me0H (7 M, 8.24 mL, 30 eq.). The mixture was stirred at room
temperature
overnight. The solvent was removed by evaporation, and the residue was diluted
with ethyl
acetate (80 mL), washed first with water (50 mL) and then with brine (50 mL),
dried with
Na2SO4. The solution was concentrated to dryness, gaving (2R,3R,4S,5R)-2-12-
chloro-6-(4-
phenyl-1 -piperi dy -5 -(hy
droxy methyl)tetrahy drofuran-3 ,4- di ol (810 mg,
94.4%) as brown solid.
[00447]
To a mixture of (2R,3R,4S,5R)-242-chloro-6-(4-pheny1-1-piperidyppurin-9-
y11-5-(hydroxymethyptetrahydrofuran-3,4-diol (810 mg, 1.82 mmol, 1 eq.) and
Ts0H-H20
(380.06 mg, 2.00 mmol, 1.1 eq.) in acetone (10 mL) was added 2,2-
dimethoxypropane (1.89 g,
18.17 mmol, 2.23 mL, 10 eq.). The mixture was stirred at room temperature for
16 h,
concentrated by evaporation. The residue was diluted with Et0Ac (100 mL),
washed (first with
aqueous NaHCO3 (2 x 50 mL), then with brine (50 mL)), dried (Na2SO4), and
concentrated.
The residue was purified by column chromatography on silica gel (eluted with
PE/EA, 100:0
to
50:50), affording [(3aR,4R,6R,6aR)-4- [2-chloro-6-(4-pheny1-1-
piperidyl)purin-9-yll -2,2-
190
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d][1,31dioxo1-6-yllmethanol (710 mg,
80.4%) as brown
solid.
[00448]
A solution of [(3aR,4R,6R,6aR)-4-[2-chloro-6-(4-pheny1-1-piperidyl)purin-9-
y11-2,2-dimethyl-3a,4,6,6a-1e1rahydr0fur0[3,4-d] [1,3] dioxo1-6-y llmethanol
(300 mg, 617.33
wnol, 1 eq.) in PO(Me0)3 (3 mL) was cooled to 0 C, followed by addition
of bis(dichlorophosphoryl) methane (308.40 mg in 1 mL P0(0Me)3, 1.23 mmol, 2
eq.). The
mixture was stirred for 6 h at 0 C, followed by addition of water (5 mL). The
mixture was
stirred at room temperature overnight, and then purified using C18 reversed
phase silica gel (0
to 30% ACN in water), giving Compound d-31, (189 mg, 50.1%) as white solid; 1H
NMR (500
MHz, CD30D) 6 ppm 1.74 (q, J=I2.1 Hz, 2H), 1.96 (d, J=12.0 Hz, 2H), 2.49 (t,
J=21.0 Hz,
2H), 2.91 (t, J=I2.1 Hz, IH), 3.18 (s, 2H), 4.24 (d, J=3.3 Hz, IH), 4.27-4.39
(rn, 2H), 4.42 (t,
J=4.9 Hz, 1H), 4.60 (t, J=5.0 Hz, 1H), 6.00 (d, J=4.8 Hz, 1H), 7.16 (t, J=7.1
Hz, 1H), 7.24 (dt,
J=8.1, 7.3 Hz, 4H), 8.29 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 26.40, 27.46,
28.52,
34.55, 43.85, 66.06, 71.37, 75.67, 84.44, 89.63, 119.20, 127.35, 127.78,
129.51, 139.26,
146.78, 152.93, 154.78, 155.16; 31P NMR (203 MHz, CD30D) 6 ppm 16.68, 20.03;
iniz
(ESI+): 604.0(M + H).
Example 51. Synthesis of compound d-32
AcO
N N CI
N- --"\IL--
0AcOAc
1 -N
N¨ 'CI NW13-Me0H y Ho Nr 'CI
DIPEA, Dioxane Ac0-10 Me0H, rt
100 C, 16 h
H HCI OAc OAc OH OH
0 0
0 0 CI¨P" 'N
__________________ 3 __ HO CI CI' CI ,
N N¨01
P0(0Me)3 HO
H20 OH OH
/K
p-Ts0H 0O OH OH
[00449]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (500 mg, 1.12
mmol, 1 eq.), 4-
cyclohexylpiperidine;hydrochloride (250.57 mg, 1.23 mmol, 1.1 eq.) in 1,4-
dioxane (20
191
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mL) was added DIEA (505.72 mg, 3.91 mmol, 681.56 pL, 3.5 eq.). The mixture was
stirred at
100 C overnight. After cooled to rt, the mixture was diluted with Et0Ac (40
mL), washed
with water and brine, subsequently. The organic layer was concentrated and the
residue
was purified by column chromatography on silica gel eluted with PE/Et0Ac
(100:0-60:40),
affording [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(4-cyclohexyl-l-
piperidyl)purin-9-
ylltetrahydrofuran-2-yllmethyl acetate (580 mg, 89.7%) as yellow solid.
[00450]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-12-chloro-6-(4-cyclohexy1-
1-
piperidyl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (580 mg, 1.00 mmol, 1
eq.) in Me0H
(8 mL) was added NH3-Me0H (7 M, 4.30 mL, 30 eq.). The mixture was stirred at
room
temperature overnight. Solvent was removed by evaporation; and the residue was
diluted with
Et0Ac (50 mL), washed subsequently with water (30 mL) and brine (30 mL), dried
with
Na2SO4, and concentrated, affording (2R,3R,4S,5R)-2-[2-chloro-6-(4-cyclohexy1-
1-
pip eri dyl)purin-9-yll -5 -(hy droxymethyl)t etrahy drofuran-3,4-di ol (420
mg, 92.6%)
[00451]
To a solution of (2R,3R,4 S,5R)-2- [2-ch1 oro-6-(4 -cy cl ohexyl-1 -pip
eri dyl)purin-
9-y1]-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (420 mg, 929.31 umol, 1 eq.)
in acetone (25
mL) were added 2,2-dimethoxypropane (1.45 g, 13.94 mmol, 15 eq.) and Ts0H-H20
(184.00
mg, 929.31 umol, 1 eq.) . The mixture was stirred at room temperature for 3 h.
After removal
of solvent by evaporation, the residue was diluted with Et0Ac (50 mL), washed
subsequently
with aqueous NaHCO3 and brine. The organic layer was concentrated and the
residue
was purified by column chromatography on silica gel eluted with PE/Et0Ac
(100:0-95:5),
giving [(3aR,4R,6R,6aR)-442-chl oro-6-(4-cy cl ohexyl -1 -piperi dy 1)purin-9-
yll -2,2-di methyl-
3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]dioxo1-6-yllmethanol (401 mg, 87.7%) as
white solid.
[00452] To a solution of [(3aR,4R,6R,6aR)-442-chloro-6-(4-cyclohexyl-l-
piperidyl)purin-9-y1]-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]
dioxo1-6-yll methanol
(400 mg, 812.99 umol, 1 eq.) in P0(0Me)3 (6 mL), at about 0 C, was
added bis(dichlorophosphoryl)methane (406.15 mg, 1.63 mmol, 2.0 eq., in 3 mL
of
P0(0Me)3). The mixture was stirred at 0 C for 5 h, followed by addition of
water (5 mL). The
mixture was stirred first at 40 C for 1 h, and then at room temperature
overnight. Purification
of the reaction mixture using C18 reversed phase silica gel (5 to 30% ACN in
water) gave
Compound d-32, (243 mg, 48.6%); 1H NMR (500 MHz, CD:30D) 6 ppm 0.98 (dd,
J=21.9,
11.8 Hz, 2H), 1.07-1.34 (m, 6H), 1.42 (s, 1H), 1.65 (d, J=11.6 Hz, 1H), 1.74
(d, J=10.7 Hz,
4H), 1.81 (s, 2H), 2.50 (t, J=21.0 Hz, 2H), 4.23 (d, J=3.4 Hz, 1H), 4.27-4.38
(m, 2H), 4.41 (s,
1H), 4.58 (s, 1H), 5.98 (d, J=4.8 Hz, 1H), 8.28 (s, 1H); 13C NMR (125 MHz,
CD30D) 6 ppm:
192
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
26.45,27.50, 27.68, 27.77, 28.56, 30.71, 31.19, 43.01, 43.92, 66.06, 71.41,
75.68, 84.46, 89.59,
119.27, 139.14, 152.92, 154.71, 155.15; 31P NMR (203 MHz, CD30D) 6 ppm 16.78,
19.91;
m/z (ESI+): 610.39(M+).
Example 52. Synthesis of compound d-33
ON CN
LDA NaBH4 COCl2 LIAIH4
0
E1OH 70 C
THF
'N
c,
0
N
(N
AGO N N CI
N
OAc OAc 0 0 <,tµi
______________________ 1.- Ac0 N CI NI-1,-Me0H... Ho N CI
Ho NI- N#LCI
DIPEA, Dioxene Me0H, rt
o
100 C, 16 h
OAc OAc OH OH 0 ,0
p-Ts0H 7\-
e
0
CI P'Fc CI
CI' CI , 0 0
P0(0Me)2 N CI
H20 OH OH ¨lc-4)4
OH OH
[00453] To a mixture of 2,2-diphenylacetonitrile (500 mg, 2.59
mmol, 1 eq.) in THF (5
mL) under nitrogen atmosphere was added dropwisc LDA (2 M, 1.55 mL, 1.2 eq.)
at -78 C,
and 20 min later, added ethyl 2-bromoacetate (518.52 mg, 3.10 mmol, 1.2 eq.)
in THF (2 mL)
at the same temperature. The mixture was stirred at room temperature for 5 h.
The reaction
was quenched by addition of 1 N HC1 (10 mL), and the mixture was extracted
with Et0Ac (50
mL). The organic layer was washed with brine, dried (Na2SO4), filtered, and
evaporated to
dryness. The residue was purified by column chromatography on silica gel
eluted with
(PE/Et0Ac from 100:0 to 75:25), giving ethyl 3-cyano-3,3-diphenyl-propanoate
(640 mg,
88.5%) as a yellow oil.
[00454]
To a mixture of ethyl 3-cyano-3,3-diphenyl-propanoate (640 mg, 2.29 mmol,
1 eq.) in Et0H (32 mL) under nitrogen atmosphere were added dropwise CoC12
(594.97 mg,
4.58 mmol, 2 eq.) and NaBH4 (866.75 mg, 22.91 mmol, 10 eq.) at 0 C. The
mixture was
stirred at room temperature overnight. The reaction was quenched with 1 N HC1
(10 mL), and
the mixture was extracted with Et0Ac (2 x 25 mL). The organic layer was washed
with brine,
dried (Na2SO4), filtered, and evaporated to dryness. The residue was purified
by column
193
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
chromatograph on silica gel eluted with (DCM/Me0H from 100:0 to 95:5), giving
4,4-
diphenylpyrrolidin-2-one (360 mg, 66.2%) as a yellow solid.
[00455] To a mixture of 4,4-diphenylpyrrolidin-2-one (320 mg,
1.35 mmol,
1 eq.) in THF (8 mL) was added LiA1H4 (102.35 mg, 2.70 mmol, 2.0 eq.). The
mixture was
stirred at 70 'V overnight. The reaction was quenched by H20 (1 mL), followed
by addition of
Et0Ac (20 mL). The solid material was removed by filtration, and the filtrate
was concentrated.
The residue was purified by column chromatography on silica gel eluted with
DCM/Me0H
(100:0-85:15), affording the product 3,3-diphenylpyrrolidine (200 mg, 66.4%)
as oil.
[00456] To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-
dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (380 mg, 849.68 jtmol, 1 eq.) in 1,4-
dioxane (10
mL) were added 3,3-di phenylpyrroli dine (208.72 mg, 934.65 itmol, I . I eq.)
and DIPEA
(274.53 mg, 2.12 mmol, 369.99 pL, 2.5 eq.). The mixture was stirred at 100 C
for 5 h, and
then diluted with Et0Ac (40 mL), washed first with water and then with brine.
The organic
layer was concentrated and the residue was purified by column chromatography
on silica gel
eluted with PE/Et0Ac (100:0-60:40), affording [(2R,3R,4R,5R)-3,4-diacetoxy-542-
chloro-6-
(3,3-dipheny 1py rroli din-1 -yl)purin-9-y1 Jtetrahy drofuran-2-y 1] methyl
acetate (401 .. mg,
74.4%).
[00457] To a solution
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(3,3-
diphenylpyrrolidin-1-yOpurin-9-ylltetrahydrofuran-2-yl]methyl acetate (401 mg,
632.41 itimol,
1 eq.) in Me0H (3 mL) was added NH3-Me0H (7 M, 2.71 mL, 30 eq.). The mixture
was
stirred at room temperature for 5 h. Solvent was removed by evaporation, and
the residue was
diluted with Et0Ac (50 mL), washed first with water (30 mL) and then with
brine (30 mL),
dried with Na2SO4, and concentrated, affording (2R,3R,4S,5R)-242-chloro-6-(3,3-
dipheny 1py rroli din-1 -y Opurin-9-yll -5 -(hydroxy methy Otetrahy drofuran-
3,4-diol (315 mg,
98.06%).
1004581 To a solution of (2R,3R,45,5R)-2-[2-chloro-6-(3,3-
diphenylpyrrolidin-1-
y urin-9-yll -5 -(hy droxy me thyl) le trahy drofuran-3,4-diol
(315 mg, 620.12 [tmol,
1 eq.) in acetone (15 mL) were added 2,2-dimethoxypropane (968.75 mg, 9.30
mmol,
15 eq.) and Ts0H-H20 (122.78 mg, 620.12 lirnol, 1 eq.). The mixture was
stirred at room
temperature for 4 h. Solvent was removed by evaporation. The residue was
diluted with Et0Ac
(50 mL), washed first with aqueous NaHCO3 and then with brine. The organic
layer was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/Et0Ac (100:0-60:40), affording 1(3aR,4R,6R,6aR)-4-[2-chloro-6-(3,3-
diphenylpyrrolidin-
194
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
1-yl)purin-9-y1]-2,2-dimethyl-3a,4,6,6a-tetrahydrofuro[3,4-d][1,31dioxol-6-
yl]methanol (320
mg, 94.16%) as white solid.
[00459]
To a solution of 1(3aR,4R,6R,6aR)-4-[2-chloro-6-(3,3-diphenylpyrrolidin-1-
yOpurin-9-y11-2,2-dimethyl-3a,4,6,6a-1e1rahydr0fur013,4-d] [1,31dioxo1-6-
yllmethanol (320
mg,
583.91 wnol, 1 eq.) in PO(Me0)3 (6 mL) at 0 C was added a solution
of bis(dichlorophosphoryl)methane (291.70 mg, 1.17 mmol, 2.0 eq.) in PO(Me0)3
( 3 mL).
The mixture was stirred at 0 C for 5 h, followed by addition of water (6 mL).
The mixture was
stirred at 40 C for 1 h, and at room temperature overnight. Purification of
the reaction mixture
using C18 reversed phase silica gel (5 to 30% ACN in water) gave Compound d-
33, (235 mg,
59.9%); 1H NMR (500 MHz, CD30D) 6 ppm 2.50 (td, J=20.9, 13.1 Hz, 2H), 2.69 (t,
J=6.8 Hz,
IH), 2.77 (d, J=6.8 Hz, IH), 3.63 (d, J=7.1 Hz, IH), 4.00 (d, J=6. I Hz, IH),
4. I 9-4.47 (m, 5H),
4.56-4.65 (m, 1H), 4.79 (q, J=12.2 Hz, 1H), 6.00 (dd, J=8.4, 4.7 Hz, 1H), 7.14
(s, 2H), 7.20-
7.35 (m, 8H), 8.40 (d, J=39.7 Hz, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 26.46,
27.52,
28.57, 36.54, 38.44, 47.49, 53.71, 55.80, 58.02, 59.05, 66.01, 71.41, 75.74,
84.61, 89.72,
119.03, 119.24, 127.63, 127.83, 129.59, 140.34, 146.49, 152.27, 153.87,
154.00, 155.50,
155.58; 31P NMR (203 MHz, CD30D) 6 ppm 16.89, 19.78; m/z (ESI+): 666.25(M+).
Example 53. Synthesis of compound d-34
cff
Ac0 IN- eLCI
Br t-IN / OH OJZX-
Boc 110 ; OAc OAc
N NH-Me0H Bac AICI ______________ DIPEA -
N Ac0 NI JD-
, , Me0H, rt
THF (CO2H)2 100 C, OAc OAc
16 h
/
\ \
0 0
.
0 0
I / 0 0
HO N N CI HO N C I
CI <1;I N
P0(0Me) DI1 8
3 HO-Y,--.1-0
H20 OH OH -I N CI
cL,
OH OH p- I sC)H Ox0 iL4
OH OH
[00460]
To a solution of bromobenzene (6 g, 38.21 mmol, 4.02 mL, 1 eq.) in THF (20
mL) was at -78 C, was added n-BuLi (2.5 M, 15.29 mL, 1 eq.). The mixture was
stirred for
min, followed by addition of tert-butyl 3-ox oazeti din e-1-carboxyl ate (3.27
g, 19.11 mmol,
0.5 eq.) in THF (15 mL). The mixture was warmed up to room temperature and
stirred at room
temperature overnight. The reaction was quenched with water (25 mL) at 0 C,
and the mixture
was diluted with Et0Ac (50 mL). The organic layer was separated, washed with
brine, and
195
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
concentrated. The residue was purified by column chromatography on silica gel
eluted with
PE/Et0Ac (100:0-70:30), affording tert-butyl 3 -hy droxy -3-phenyl-azeti dine-
1 -carboxy late
(3.9 g, 40.9%) as light brown solid
[00461]
A1C13 (1.60 g, 12.03 mmol, 3.0 eq.) was suspended in toluene (739.16 mg,
8.02
mmol, 853.53 uL, 2.0 eq.) and cooled to 0 'C. To the cold suspension, at 0 C,
a solution
of tert-butyl 3-hydroxy-3-phenyl-azetidine-1-carboxylate (1 g, 4.01 mmol, 1
eq.) in toluene
(731.82 [IL) was added. The mixture was stirred at 0 C for 2 h. The reaction
was quenched by
addition of ice water. The mixture was stirred for 0.5 h, followed by
addition, subsequently, of
saturated aqueous NaHCO3 and NH3-H20, until the pH of mixture reached 11. The
mixture
was extracted with Et0Ac. The extract was washed with brine, dried with
Na2SO4, filtered,
and concentrated. The residue was dissolved in a minimum amount of ethyl
acetate, followed
by addition of a solution of oxalic acid (361.12 mg, 4.01 mmol, 1 eq.) in
Et0Ac (1.5 mL). The
solid material thus formed was collected by filtration and dried, affording 3-
pheny1-3-(p-
tolyl)azetidine oxalate (1.05 g, 3.35 mmol, 83.54% yield).
[00462]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yl)tetrahydrofuran-2-yl]methyl acetate (500 mg, 1.12 mmol, 1.0 eq.) in 1,4-
dioxane (20
mL) were added 3-phenyl-3-(p-tolyl)azetidine oxalate (385.35 mg, 1.23 mmol,
1.1 eq.) and DIPEA (577.96 mg, 4.47 mmol, 778.93 pt, 4 eq.). The mixture was
stirred at
100 C overnight, and concentrated. The residue was purified by column
chromatography on
silica gel eluted with PE/Et0Ac (100:0-50:50), affording R2R,3R,4R,5R)-3,4-di
acetoxy -5 42-
chl oro-6- 113 -phenyl-3 -(p-toly pazeti din-1 -yl] purin-9-yll tetrahydro
furan-2-yll methyl acetate
(401 mg, 56.6%) as white solid
[00463]
To a solution of (2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-]3-pheny1-3-(p-
tolypazetidin-1-yl]purin-9-ylltetrahydrofuran-2-yllmethyl acetate (401 mg,
632.41 umol,
1 eq.) in Me0H (5 mL) was added Me0H-NH3 (7 M, 2.71 mL, 30 eq.). The mixture
was stirred
at room temperature for 4 h. Solvent was removed by evaporation, and the
residue was diluted
with Et0Ac (100 mL) and H20 (70 mL). The organic layer was separated, washed
with brine,
dried with Na2SO4, and concentrated to dryness, affording (2R,3R,45,5R)-2-[2-
chloro-643-
pheny1-3-(p-tolypazetidin-l-yll purin-9-y11-5-(hydroxymethyptetrahydrofuran-
3,4-diol (320
mg, 99.6%) .
[00464]
To a solution of (2R,3R,4S,5R)-242-chloro-643-pheny1-3-(p-tolyl)azetidin-1-
yll pun n -9-y1 -5 -(hy droxym ethy Otetrahy drofuran-3 ,4-di ol (320
mg, 629.96 umol,
1 eq.) in acetone (25 mL) was added 2,2-dimethoxypropane (1.31 g, 12.60 mmol,
196
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
20 eq.) and p-Ts0H (108.48 mg, 629.96 pmol, 1 eq.). The mixture was stirred at
room
temperature overnight. The solvent was removed by concentrated and the residue
was diluted
with Et0Ac (50 mL), washed by aq.NaHCO3, followed by brine and the organic
layer was
concentrated and the residue was purified by column chromatography on silica
gel eluted with
PE/Et0Ac (100:0-50:50) to
afford [(3aR,4R,6R,6aR)-442-chloro-643-pheny1-3-(p-
tolypazetidin-1-yllpurin-9-y11-2,2-dimethyl-3a,4,6,6a-tetrahy drofuro [3,4-d]
[1,31dioxo1-6-
yl] methanol (345 mg, 99.9%)
[00465]
To a solution of 1(3aR,4R,6R,6aR)-4-[2-chloro-6-[3-pheny1-3-(p-
tolyl)azetidin-
1-yll purin-9-y11-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro [3,4-d] [1,3] dioxo1-6-
yl[methanol (345
mg, 629.53 mnol, 1 eq.) in PO(Me0)3 (4 mL) at 0 CC was added a solution
of bis(dichlorophosphoryl)methane (314.49 mg, 1.26 mmol, 2.0 eq.) in PO(Me0)3
(4 mL). The
mixture was stirred at 0 C for 5 h, followed by addition of water (5 mL). The
mixture was
stirred at 40 C for 40 min, and then at room temperature overnight. The
reaction mixture was
purified via C18 reversed phase silica gel (0 to 25% ACN in water), giving
Compound d-34,
(259 mg, 60.0%); 1H NMR (500 MHz, CD30D) 6 ppm 2.23 (s, 3H), 2.55 (t, J=20.8
Hz, 2H),
4.23 (s, 1H), 4.26-4.38 (m, 2H), 4.41 (t, J=4.6 Hz, 1H), 4.61 (t, J=4.6 Hz,
1H), 4.81 (s, 2H),
5.14 (s, 2H), 5.97 (d, J=4.5 Hz, 1H), 7.06 (d, J=7.9 Hz, 2H), 7.14 (dd,
J=18.7, 7.2 Hz, 3H),7.19-
7.31 (m, 4H), 8.50 (s, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 21.04, 26.50,
27.55, 28.60,
65.09, 65.92, 66.99, 71.29, 75.75, 84.71, 89.87, 118.13, 127.37, 127.45,
127.70, 129.67,
130.26, 137.54, 141.28, 144.11, 147.29, 151.67, 155.08, 155.75;31P NMR (203
MHz, CD30D)
6 ppm 17.11, 19.54; m/z (ESL): 666.45 (M+H).
Example 54. Synthesis of compound d-35
ci
,
\
Ac0 N
Xci
Br (D't CH
OAc OAc
Boc
NH,Me0H
BuU h- BoT N Ac0
AICI, DIPEA, Do Me0H,
rt
THF (CO.,H) 100 C, 160
OAc OAc
/ /
\ \
Ok0
0 0
C1¨/P"---'P\-C1
HO N HO N leLCI CI 0, 9 N-
P0(0M03 P, P-0 N- reLCI
1120 Hr)-01-0H
OH OH P-Ts0H Ox0
OH OH
197
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00466]
To a solution of bromobenzene (6 g, 38.21 mmol, 4.02 mL, 1 eq.) in THF (20
mL), cooled at -78 C, was added n-BuLi (2.5 M, 15.29 mL, 1 eq.). The mixture
was stirred at
-78 C for 10 min, followed by addition of tert-butyl 3-oxoazetidine-1-
carboxylate (3.27 g,
19.11 mmol, 0.5 eq.) in THF (15 mL). The mixture was warmed up to room
temperature and
stirred at room temperature overnight. The reaction was quenched with water
(25 mL) at 0 'C.
After addition to the mixture of Et0Ac (50 mL), organic layer was separated,
washed with
brine, and concentrated. The residue was purified by column chromatography on
silica gel
eluted with PE/Et0Ac (100:0-70:30), affording tert-butyl 3-hy droxy -3 -phenyl-
az eti dine-1 -
carboxylate (3.9 g, 40.9%) as light brown solid.
[00467]
A1C13 (802.27 mg, 6.02 mmol, 3.0 eq.) was suspended in benzene (313.32 mg,
4.01 mmol, 358.49 itiL, 2.0 eq.) and cooled to about 0 C. To the cold
suspension at 0 C was
added tert- butyl 3-hy droxy -3 -phenyl-azeti dine-1 -carb oxyl ate (500 mg,
2.01 mmol,
1 eq.) in benzene (6 mL). The mixture was stirred at 0 C for 2 h. The
reaction was quenched
by adding ice water and stirring for 0.5 h. To the mixture was added
subsequently saturated
aqueous NaHCO3 and NH3-H20 until the pH of the mixture reached 11. The mixture
was
extracted with Et0Ac, and the extract was washed with brine, dried with
Na2SO4, filtered, and
concentrated. The residue was dissolved in a minimum amount of Et0Ac, followed
by addition
of oxalic acid (180.56 mg, 2.01 mmol, 1 eq.) in Et0Ac (8 mL). The solid, thus
formed, was
collected by filtration, and dried, affording 3,3-diphenylazetidine oxalate
(500 mg, 83.3%).
[00468]
To a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yptetrahydrofuran-2-ylimethyl acetate (500 mg, 1.12 mmol, 1 eq.) in 1,4-
dioxane (25
mL) were added 3,3-diphenylazetidine oxalate (401.57 mg, 1.34 mmol, 1.2 eq.)
and DIPEA
(577.96 mg, 4.47 mmol, 778.93 taL, 4.0 eq.). The mixture was stirred at 100 'V
overnight, and
then concentrated. The residue was diluted with Et0Ac (40 mL), washed
subsequently with
water and brine. The organic layer was concentrated and the residue was
purified by column
chromatography on silica gel, eluted with PE/Et0Ac (100:0-50:50), affording
[(2R,3R,4R,5R)-
3,4-di ace toxy -5 42-chl oro-6-(3,3-diphenylaze ti din-1 -y urin-9-yll
tetrahy drofuran-2-
yl] methyl acetate (300 mg, 43.3%).
[00469] To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(3,3-
diphenylazetidin-l-yl)purin-9-ylitetrahydrofuran-2-yllmethyl acetate (300 mg,
483.83 tamol,
1 eq.) in Me0H (4 mL) was added NH3-Me0H (7 M, 2.07 mL, 30 eq.). The mixture
was
stirred at room temperature for 3 h. After removal of solvent by evaporation,
the residual
material was diluted with Et0Ac (50 mL), washed subsequently with water (30
mL) and brine
198
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
(30 mL), dried with Na2SO4, and concentrated, giving (2R,3R,4S,5R)-2-[2-chloro-
6-(3,3-
diphenylazeti din-1 -yl)purin-9-yl] -5-(hy droxy methy Otetrahy drofuran-3,4-
diol (230 mg,
96.2%) as white solid.
[00470]
To a solution of (2R,3R,4S,5R)-2-[2-chloro-6-(3,3-diphenylaze1idin-1-
yl)purin-
9-y11-5-(hydroxymethy1)tetrahydrofuran-3,4-diol (230 mg, 465.64
1 eq.) in acetone (25
mL) were added 2,2-dimethoxypropane (727.43 mg, 6.98 mmol, 15 eq.) and p-Ts0H
(80.18
mg, 465.64 limo', 1 eq.). The mixture was stirred at room temperature for 2 h.
After removal
of the solvent by evaporation, the residue was diluted with Et0Ac (30 mL),
washed with
aqueous NaHCO3 and brine, subsequently. The organic layer was concentrated and
the residue
was purified by column chromatography on silica gel, eluted with PE/Et0Ac
(100:0-50:50),
affording [(3aR,4R,6R,6aR)-4 42-chloro-6-(3,3 -di ph enylazeti din- I -yl
)puri n -9-yl] -2,2-
dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]dioxo1-6-yl1methanol (220 mg,
88.5%) as white
solid.
[00471]
To a solution of [(3aR,4R,6R,6aR)-442-chloro-6-(3,3-diphenylazetidin-l-
yOpurin-9-y11-2,2-dimethyl-3a,4,6,6a-tetrahy drofuro [3 ,4-dl [1 ,31di oxo1-6-
y11 methanol (220
mg, 411.98 umol, 1 eq.) in PO(Me0)3 (4 mL) at
0 C was
added bis(dichlorophosphoryl)methane (205.81 mg, 823.96 pmol, 2.0 eq.) in
PO(Me0)3 (2
mL). The mixture was stirred at about 0 C for 4 h, followed by addition of
water (5 mL). The
mixture was stirred first at 40 C for 30 min, and then at room temperature
overnight.
Purification of the reaction mixture using C18 reversed phase silica gel (0 to
25% ACN in
water) gave Compound d-35, (150.4 mg, 54.9%);1H NMR (500 MHz, CD30D) 6 ppm
2.50 (t,
J=20.9 Hz, 2H), 4.24 (s, 1H), 4.30 (d, J=6.1 Hz, 1H), 4.33-4.38 (m, 1H), 4.42
(t, J=3.9 Hz, 1H),
4.62 (t, J=4.1 Hz, 1H), 4.89 (s, 2H), 5.22 (s, 2H), 5.89-6.03 (m, 1H), 7.23
(dt, J=8.1, 4.0 Hz,
2H), 7.34 (dd, J=8.7, 5.3 Hz, 8H), 8.41 (s, 1H); 13C NMR (125 MHz, CD30D) 6
ppm 26.47,
27.53, 28.58, 65.97, 71.39, 75.66, 84.63, 89.78, 119.17, 127.47, 127.84,
129.77, 141.49,
147.26, 152.00, 155.54, 155.60;31P NMR (203 MHz, CD30D) 6 ppm 16.79, 19.80;
m/z (ESE):
652.3 (M+H).
Example 55. Synthesis of compound d-36
199
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
0 0 0 Op phenol
Pd/C LAIH14,A1Clx cJ
112,Pd(OH)2
N NH,COOH
DIPEA,4A.,106 C 6h CHs0H,d,16h THF,rt,16h
N¨b) Me0H,60 C,O/N
NH
0 0
CI
I I
0 0
Acrp¨y24
NHs-Me0H p-Ts0H CI CI
OAc OAc N
DIPEA, Dioxane Me0H, rt, 16h N Hs0,4010,40min
Ac0-04 N CI HON Cl
05h N CI rt,o,N HOOCI
74
100"C 4h
IAc Ac HO HO Ox0 OH OH
[00472]
3,4-Diphenylfuran-2,5-dione (4.5 g, 17.98 mmol, 1 eq.), phenylmethanamine
(3.85 g, 35.96 mmol, 2 eq.), phenol (3.38 g, 35.96 mmol, 2 eq.), DIPEA
(18.59g. 143.86 mmol,
25.06 mL, 8 eq.) and molecular sieves 4A (500 mg) were placed in a sealed
tube. The mixture
was sin-ed at 100 'V for 6 h, and then cooled to rt. The pH of the mixture was
adjusted to 5
with 4% HC1. The pH adjusted mixture was extracted with Et0Ac (2 x 50 mL). The
extract
was washed with brine, and concentrated. The residue was purified by column
chromatography
on silica gel, eluted with PE/EA (100:0-90:10). The fraction containing
product was evaporated
to dryness, and the residue was triturated with PE/EA=50:1 (40 mL) overnight.
Solid material
thus formed was collected by filtration, washed with small amount of solvent
(PE/EA=50:1),
and dried, giving 1-benzy1-3,4-diphenyl-pyrrole-2,5-dione (5.85 g, 17.24 mmol,
95.86% yield).
[00473]
To a solution of 1-benzy1-3,4-diphenyl-pyrrole-2,5-dione (3.15 g, 9.28
mmol, 1
eq.) in CH3OH (60 mL) was added Pt02 (300 mg, 928.15 !Imo', 0.1 eq.). The
mixture was
stirred at under H2 atmosphere at rt overnight. Insoluble material was removed
by filtration and
washed with Me OH. The filtrate and washing were combined and concentrated,
affording 1-
benzy1-3,4-diphenyl-pyrrolidine-2,5-dione (3.0 g, 8.79 mmol, 94.68% yield).
[00474]
To a mixture of LiA1H4 (333.48 mg, 8.79 mmol, 6 eq.) in THF (15 mL) cooled
at 0 C was added Al C13 (781.13 mg, 5.86 mmol, 4 eq.). The mixture was
stirred at 0 C for 30
min, followed by addition of 1-benzy1-3,4-diphenyl-pyrrolidine-2,5-dione (500
mg, 1.46
mmol, 1 eq.) in THF (4 mL). The mixture and stirred at rt over a weekend. The
reaction was
quenched with water (1 mL), followed by addition of aqueous NaOH (15%, 4 mL)
and EA (20
mL). The mixture was dried over MgSO4. The insoluble material was removed by
filtration,
and the filtrate was concentrated, affording 1-benzy1-3,4-diphenyl-pyrrolidine
(490 mg).
[00475]
To a solution of 1-benzy1-3,4-diphenyl-pyrrolidine (490 mg, 1.56 mmol, 1
eq.)
in Me0H (15 mL) were added ammonium formate (147.87 mg, 2.34 mmol, 1.5
eq.) and Pd(OH)2 (44 mg, 312.00 lirnol, 0.2 eq.). The mixture was stirred at
under hydrogen
atmosphere at 60 'V overnight. The insoluble material was removed by
filtration and washed
200
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
with Me0H. The filtrate and washing were combined and concentrated to dryness,
affording
3,4-diphenylpyrrolidine (350 mg crude), which was used directly for the next
step.
[00476] To
a mixture of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-yllmethyl acetate (400.54 mg, 895.61 [unol, 1 eq.) in 1,4-
dioxane (20
mL) were added 3,4-diphenylpyrrolidine (350 mg, 1.57 mmol, 1.75 eq.) and DIPEA
(405.12
mg, 3.13 mmol, 545.98 [iL, 3.5 eq.). The mixture was stirred at 100 C for 4
h. Solvent was
removed by evaporation; and the residue was diluted with Et0Ac (40 mL), washed
with water
and brine, subsequently. The organic layer was concentrated and the residue
was purified by
column chromatography on silica gel eluted
with PE/EA (100:0-70:30),
giving [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(3,4-diphenylpyrrolidin-l-
yl)purin-9-
ylltetrahydrofuran-2-yllmethyl acetate (490 mg, 772.78 [Imo], 86.29% yield) as
white solid.
[00477] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(3,4-
diphenylpyrrolidin-1-yl)purin-9-ylltetrahydrofuran-2-yl]methyl acetate (490
mg, 772.78 1.tinol,
1 eq.) in Me0H (6 mL) was added NH3-Me0H (7 M, 3.31 mL, 30 eq.). The mixture
was stirred
at room temperature overnight. Solvent was removed by evaporation. The residue
was diluted
with Et0Ac (40 mL), washed with brine, dried with Na2SO4, and concentrated,
affording
(2R,3R,4S,5R)-2-[2-chloro-6-(3,4-diphenylpyrrolidin-1-yl)purin-9-yll -5 -
(hydroxymethyl)tetrahydrofuran-3,4-diol (364 mg, 716.58 limo', 92.73% yield)
as white solid.
[00478]
To a solution of (2R,3R,4 S,5R)-242-chl oro-6-(3,4-diphenylpyrroli din-1 -
y1)
purin-9-yl] -5 -(hy droxy methyl)tetrahy drofuran-3 ,4-di ol (364 mg, 716.58
mmol, 1
eq) in acetone (30 mL) were added 2,2-dimethoxypropane (1.49 g, 14.33 mmol,
1.76 mL, 20
eq) and p-Ts0H-H20 (137.58 mg, 716.58 wnol, 1 eq). The mixture was stirred at
room
temperature for 2 h. Solvent was removed by evaporation. The residue was
diluted with Et0Ac
(30 mL), washed with aqueous NaHCO3 (2 x 20 mL) and brine (30 mL)
subsequently, and
concentrated to dryness. The residue was purified by column chromatography on
silica gel,
eluted with PE/EA (100:0-60:40),
affording [(3 aR,4R,6R,6aR)-4- [2-chl oro-6-(3,4-
dipheny 1py rroli din-1 -y Opurin-9-y11-2,2-dimethy1-3 a,4,6,6a- tetrahy
drofuro [3,4-d] [1,3] dioxol-
6-yllmethanol (280 mg, 510.92 timol, 71.30% yield) as white solid.
[00479]
To a solution of [(3aR,4R,6R,6aR)-4-[2-chloro-6-(3,4-diphenylpyrrolidin-1-
yl)purin-9-yl] -2,2-dimethy1-3 a,4,6,6 a-tetrahy drofuro [3 ,4-d] [1,3] di
oxo1-6-yll methanol (280
mg,
510.92 1.imol, 1 eq.) in PO(Me0)3 (4 mL), cooled at 0 C, was added
bis(dichlorophosphoryOmethane (255.24 mg, 1.02 mmol, 2 eq.) in PO(Me0)3 (4
mL). The
mixture was stirred at 0 'V for 5 h. LC-MS monitoring indicated that the
reaction progressed
201
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
very little. Bis(dichlorophosphoryOmethane (127.62 mg, 0.51 mmol, 1 eq.) in
PO(Me0)3 (2
mL) was added into the mixture. The mixture was stirred at about 0 C for 3 h,
and used directly
for next step.
[00480] To the above
mixture at 0 C was added water (7 mL), following by stirring the
mixture at 40 'DC for 40 mm first and then at room temperature overnight. The
reaction mixture
was purified using C18 reversed phase silica gel (eluted with 0 to 30% ACN in
water), giving
Compound d-36, (75 mg, 112.62 iamol, 22.10% yield).1H NMR (500 MHz, Me0D,
CDC13)
6ppm 2.49 (t, J = 20.9 Hz, 2H), 3.77 (m, 3H), 4.15 (s, 1H), 4.23 - 4.38 (m,
4H), 4.45 (s, 2H),
4.65 (s, 1H), 6.05 (s, 1H), 7.22 (m, 2H), 7.26 - 7.37 (m, 8H), 8.40 (s, 1H).
31P NMR (203 MHz,
DMSO) oppm 14.57, 18.66. 13C NMR (126 MHz, DMSO) 6 ppm 27.99, 48.88, 50.88,
55.32,
56.96, 65.01, 70.68, 74.09, 83.76, 119.08, 127.38, 127.45, 128.26, 129.01,
139.55, 139.64,
139.71, 151.71, 153.02, 153.53. m/z (ESI ): 666.34(M+H).
Example 56. Synthesis of compound d-38
rsixj,ci N / \
_
_
\ 1
? 40 0 Ac0 1...4 Nj.--C1
I,.. j,..._N EN 17(),1 dioxane A:IP: 100 C ' Ac -
1, N ''' NH.
CF= O OMHeC:IH'. H 1
N'........CI ' ' '-
Ill
OA Ac 711-7:1H p-
Ts0H
-- ---/ ___ ¨
N CI ¨,P--- k-CI N"
<3.4f,1 9 CIPO(OM C) 1 2.-9
H 9 <'NXIILN
CI H20 ' -P,õ-P-0 N N- 'CI
¨I.LJ OH OH ---
CX) OH OH
[00481] To a solution
of 3-phenyl-1H-indole (200 mg, 1.03 mmol, 1 eq.) in CF3COOH
(2 mL) was added Et3SiH (240.69 mg, 2.07 mmol, 330.62 pt, 2.0 eq.). The
mixture was stirred
at 50 C overnight. pH of the mixture was adjusted 9 by adding aqueous Na2CO3,
followed by
addition of Et0Ac (25 mL). Organic layer was separated, dried with Na2SO4, and
concentrated.
The residue was purified by column chromatography on silica gel eluted with
(PE/Et0Ac from
100:0 to 80:20), to afford 3-phenylindoline (110 mg, 54.4%).
[00482] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yOtetrahydrofuran-2-ylimethyl acetate (229.04 mg, 512.14 [Imo', 1 eq.) in 1,4-
dioxane (15
mL) were added 3-phenylindoline (100 mg, 512.14 mmol, 1 eq.) and DIPEA (165.47
mg, 1.28
mmol, 223.01 4, 2.5 eq.). The mixture was stirred at 100 C for 4 h. After
cooled to rt, the
202
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mixture was diluted with Et0Ac (50 mL). The organic layer was separated,
washed
subsequently with water and brine, dried with Na2SO4, and concentrated. The
residue
was purified by column chromatography on silica gel, eluted with (PE/EA from
100:0 to
60:40), affording [(2R,3R,4R,5R)-3,4-diacetoxy -5- [2-chl oro-6-(3 -pheny
lindol in- 1-yl)p urin-
9-ylltetrahydrofuran-2-yllmethyl acetate (150 mg, 48.3%) as white solid.
[00483]
To a solution of [(2R,3R,4R,5R)-3,4-diacetoxy-542-chloro-6-(3-
phenylindolin-
1-yl)purin-9-ylltetrahydrofuran-2-yllmethyl acetate (150 mg, 247.51 iamol, 1
eq.) in Me0H (3
mL) was added Me0H-NH3 (7 M, 1.06 mL, 30 eq.). The mixture was stirred at room
temperature overnight. Solvent was removed by evaporation. The residue was
diluted with
Et0Ac (40 mL), washed with water and brine subsequently. The organic layer was
dried with
Na2SO4, and concentrated, giving (2R,3R,4S,5R)-2-1-2-chloro-6-(3-phenylindolin-
1 -yl)purin-
9-yll-5-(hydroxymethyl)tetrahydrofuran-3,4-diol (105 mg, 88.4%) as white
solid.
[00484]
To a solution of (2R,3R,4S,5R)-242-chloro-6-(3-phenylindolin-1-yl)purin-9-
yll -5-(hydroxymethyl)tetrahydrofuran-3,4-diol (105 mg, 218.79 lamol, 1 eq.)
in acetone (15
mL) were added 2,2-dimethoxypropane (227.86 mg, 2.19 mmol, 10 eq.) and p-Ts0H
(37.68
mg, 218.79 wriol, 1 eq.). The mixture was stirred at room temperature for 2 h,
and then diluted
with Et0Ac (30 mL). The mixture was washed with aqueous Naf1t03 and brine.
Organic layer
was separated and concentrated. The residue was purified by column
chromatography on silica
gel eluted with (PE/Et0Ac from 100:0 to 60:40), affording [(3aR,4R,6R,6aR)-442-
chloro-6-
(3-phenyl indolin-l-yl)purin-9-yl] -2,2-di methyl-3 a,4,6,6 a-tetrahy drofuro
13 ,4-d] [1,3] di oxo1-6-
yllmethanol (100 mg, 87.9%) as white solid.
[00485]
To a solution of 1(3aR,4R,6R,6aR)-4-12-chloro-6-(3-phenylindolin-1-
yl)purin-
9-y1]-2,2-dimethy1-3a,4,6,6a-tetrahydrofuro[3,4-d] [1,3]dioxo1-6-yl]methanol
(130 mg, 250.01
iamol, 1 eq.) in PO(Me0)3 (4 mL), cooled at 0 C, was added The solution
of bis(dichlorophosphoryl)methane (124.90 mg, 500.02 vino', 2.0 eq.) in
PO(Me0)3 (3 mL).
The mixture was stirred at about 0 C for 5 h. After addition of water (3 mL)
to the mixture at
0 C, the mixture was stirred at 40 C for 30 min first, and then at room
temperature overnight.
Purification of the reaction mixture was achieved by C18 reversed phase silica
gel
chromatography (gradient eluent 0 to 30% ACN in water), giving Compound d-38,
(89 mg,
54.1%). 1H NMR (500 MHz, CD30D) 6 ppm 2.27-2.59 (m, 2H), 4.25 (s, 1H), 4.30
(dd, J=21.5,
15.0 Hz, 2H), 4.43 (dt, J=9.6, 4.7 Hz, 1H), 4.56-4.70 (m, 3H), 5.15 (t, J=10.7
Hz, 1H), 6.03 (d,
J=4.6 Hz, 1H), 6.98 (dt, J=13.7, 7.1 Hz, 2H), 7.13-7.34 (m, 6H), 8.36 (s, 1H),
8.56 (d, J=8.2
Hz, 1H); 13C NMR (125 MHz, CD30D) 6 ppm 26.41, 27.47, 28.53, 47.79, 60.91,
66.02, 71.41,
203
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
75.73, 84.58, 89.65, 118.82, 120.37, 125.16, 126.19, 128.17, 128.81, 129.90,
137.19, 140.94,
144.32, 144.70, 144.77, 152.34, 153.36, 154.54; 31P NMR (203 MHz, CD30D) 6 ppm
16.79,
19.87; m/z (ESI+): 638.3 (M+H).
Example 57. Synthesis of compound d-39
0
NH
µ1:11 .11
AcD_NNCI
"
LC)_?
0 OAE 0 Au
H DIPEA PDhlOcg: IL%7E4
B%t,SEI,H BH,-THE20 a
THE
HO 0 _cm / Ae0 N -
eLCI
iciLO4
0Ac OAc
0)1
Isr ci
NI-13-Me0H J 0 p-T;OH.H2O, CI' CI
IJ Me0H, 4'1"
'cet''e HO NI -NeLCI PO(OEt) a-
0 0 j
H,0 N =Nt CI
HO-4- -124 OH OH -1cr, ?,
OH OH OH OH
1004861
To a mixture of isoindoline-1,3-dione (1 g, 6.80 mmol, 1 eq.) in DCM (30
mL) was added PhMgBr (1 M, 20.39 mL, 3 eq.) at 0 C. The mixture was stirred
for 4 h at 0
C, followed by quenching the reaction with water (30 mL). The mixture was
filtered. The
organic layer was separated, washed with brine, dried (Na2SO4), filtered, and
evaporated to
dryness. The residue was purified by column chromatography on silica gel with
(DCM:Me0H
=100:0 to 95:5 ), giving 3-hydroxy-3-phenyl-isoindolin-1 -one (1.3 g, 5.77
mmol, 84.92%
yield) as a solid.
[00487] To a mixture of 3-hydroxy-3-phenyl-isoindolin-1-one (980
mg, 4.35 mmol, 1
eq.) in DCM (15 mL) was added BF3-Et20 (3 mL) and Et3SiH (1.52 g, 13.05 mmol,
2.08 mL,
3 eq.) at -40 C, it was stirred at RT for 16 h. TLC showed the SM was
consumed. Then it was
quenched by water (20 mL) and NH4C1 (20 mL), another DCM (30 mL) was added,
solid
formed, the solid was removed by filtration. Then the filtrate was extracted
with DCM (25
mL*2). The organic layer was washed by brine, concentrated. It was merged with
the solid to
give 3-phenylisoindolin-1-one (900 mg, 4.30 mmol, 98.86% yield) as a white
solid.
[00488]
To a solution of 3-phenylisoindolin-1-one (700 mg, 3.35 mmol, 1 eq.) in
THF
(10 mL) was added BH3-THF (1 M, 30.11 mL, 9 eq.). The mixture and stirred at
70 C
overnight. The reaction was quenched by CH3OH (3 mL) and the mixture was
concentrated.
The residue was purified by column chromatography on silica gel eluted with
PE/EA (100:0-
90:10), affording 1-phenylisoindoline (185 mg, 947.46 iamol, 28.32% yield).
204
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
[00489]
To a mixture of 1-phenylisoindoline (185 mg, 947.46 pmol, 1.2 eq.) in 1,4-
dioxane (15 mL) were
added [(2R,3R,4R,5R)-3,4-diacetoxy-5-(2,6-dichloropurin-9-
yptetrahydrofuran-2-yl]methyl acetate (353.11 mg, 789.55 pmol, 1 eq.) and
DIPEA (255.10
mg, 1.97 mmol, 343.80 pL, 2.5 eq.). The mixture was stirred at 100 C for 4 h,
and at rt
overnight. Solvent was removed by evaporation. The residue was diluted with
Et0Ac (50 mL),
washed with water and brine successively. The organic layer was concentrated
and the residue
was purified by column chromatography on silica gel eluted with PE/EA (100:0-
50:50),
affording
[(2R,3R,4R,5R)-3,4-di acetoxy -5 - [2-chl oro-6-(1-pheny s oindolin-2-
yl)purin-9-
ylltetrahydrofuran-2-yllmethyl acetate (360 mg, 594.04 p,mol, 75.24% yield) as
green solid.
[00490] To a mixture
of [(2R,3R,4R,5R)-3,4-diacetoxy-5-[2-chloro-6-(1-
phenylisoindolin-2-yl)purin-9-ylitetrahydrofuran-2-ylimethyl acetate (420 mg,
693.04 jimol, 1
eq.) in Me0H (4 mL) was added NI-3-Me0H (7 M, 2.97 mL, 30 eq.). The mixture
was stirred
at room temperature overnight. Solvent was removed by evaporation. The residue
was diluted
with Et0Ac (40 mL), washed with brine (40 mL), dried with Na2SO4. The organic
layer was
concentrated to dryness, affording (2R,3R,4S,5R)-2-[2-chloro-6-(1-pheny-
lisoindolin-2-
yl)purin-9-y1]-5-(hydroxymethyptetrahydrofuran-3,4-diol (332 mg, 691.79 limo',
99.82%
yield).
[00491]
To a solution of (2R,3R,4S,5R)-2-12-chloro-6-(1-phenylisoindolin-2-
yl)purin-9-
y11-5-(hydroxymethyptetrahydrofuran-3,4-diol (332 mg, 691.79 jamol, 1 eq.) in
acetone (20
mL) were added p-Ts0H-H20 (131.58 mg, 691.79 limo', 1 eq.), 2,2-
dimethoxypropane (1.44
g, 13.84 mmol, 1.70 mL, 20 eq.). The mixture was stirred at rt for 2 h.
followed by pH
adjustment with slow addition of aqueous NaHCO3 at 0 C until the pH of the
mixture reached
9. Solvent was removed by evaporation. The residue was diluted with Et0Ac (50
mL), and the
mixture was washed with aqueous NH4C1 and brine, successively. The organic
layer was
concentrated, and the residue was purified by column chromatography on silica
gel eluted with
PE/EA (100:0-50:50), affording [(3aR,4R,6R,6aR)-4-[2-chloro-6-(1-
phenylisoindolin-2-
yOpurin-9-y11-2,2-dimethyl-3a,4,6,6a- tetrahy drofuro [3,4-d] [1,3] dioxo1-6-
yll methanol (197
mg, 378.86 pmol, 54.77% yield) as purple solid.
[00492]
To a solution of 1(3aR,4R,6R,6aR)-4-12-chloro-6-(1-phenylisoindolin-2-
yl)purin-9-y11-2,2-dimethy1-3 a,4,6,6 a-tetrahy drofuro [3 ,4-d] 11 ,31di oxo1-
6-yl] methanol (197
mg,
378.86 1.imol, 1 eq.) in P0(0E03 (2 mL), cooled at 0 C, was added
bis(dichlorophosphoryOmethane (208.19 mg, 833.50 pmol, 2.2 eq.) in P0(0E03 (2
mL). The
mixture was stirred at 0 'V for 5 h, followed by slow addition of water (3 mL)
at 0 'C. The
205
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
mixture was stirred at 40 C for 40 min., and at 20 C for overnight. The
reaction mixture was
purified using C18 reversed phase silica gel (gradient eluent, 0 to 30% ACN in
water), giving
Compound d-39, (14 mg, 21.66 lamol, 11.51% yield, 98.69% purity). 1H NMR (500
MHz,
Me0D) 6 ppm 2.55 (t, J = 20.6 Hz, 2H), 4.20¨ 4.51 (m, 4H), 7.37 ¨ 7.12 (m,
8H), 4.62 (t, J =
30.4 Hz, 1H), 5.27 (d, J = 16.8 Hz, 1H), 5.63 (t, J = 16.7 Hz, 1H), 6.00 (d, J
= 23.5 Hz, 1H),
6.50 (s, 1H), 7.12¨ 7.37 (m, 8H), 8.32 (dd, J = 138.5, 15.4 Hz, 1H); m/z (ESI
): 638.0(M+H).
Compound a
[00493]
Compound a is a compound reported in the literature (named AB680) with the
following structure:
HN
9 9 Rfl
0 N
, 0õ-.....0õ, CI
HO
HO
[00494]
This compound was prepared according to procedures described in
International
Application Publication No. W02019173682.
Compound b
[00495]
Compound b is a compound reported in the literature having the following
structure:
0 0
HO CI 0
OH OH
OH OH
[00496]
Compound b was prepared using procedures described in Chinese patent
application publication no. CN110885352.
Example 58. CD73 enzyme inhibition assay for selected compounds.
[00497] To evaluate the inhibitory effect of selected
compounds on CD73,
Malachite Green Phosphate Detection Kit (R&D, Cat# DY996) was used. Briefly,
compounds
were dissolved and diluted to the desired concentration using phosphate-free
buffer (Tris-HC1,
206
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
pH 7.3). 25 jiL of the compound solution was added to an equal volume of CD73
protein
solution (2 x concentration, 0.5 lig/mL, Novoprotein, Cat#C446), followed by a
5-minute
incubation at room temperature. 100_, of Malachite Green Reagent A was added
to each well,
mixed thoroughly and incubated for 10 minutes at room temperature. 10111_, of
Malachite Green
Reagent B was then added to each well, mixed thoroughly and incubated for 20
minutes at
room temperature. Finally, the optical density of each well was determined
using a microplate
reader set to 620 nm.
[00498]
The inhibitory activity of selected compounds is given in Table 2. In
addition, compounds such as a-1, a-9, and a-31 showed inhibitory activity in
the assay.
Table 2. Compound inhibitory activity in CD73 enzyme assay.
Compound No. Potency'
1 +++
2 ++
6 +++
7 +++
8 ++
9
++
11 ++
12 ++
+++
16
17 +++
18 ++
19
++
22 +++
23 ++
207
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
31 +++
51 +++
1 "+" denotes IC50> 100 nM; "++" denotes IC50 of 10-100 nM; "+++" denotes
IC50< 10 nM.
Example 59. Pharmacokinetic evaluation of selected compounds
1004991 A test
compound (1.0 mg/kg) was administered to fasted ICR male mice via
intravenous injection, and blood samples were collected at time points of
0.08, 0.25, 0.5, 1, 2,
4, 6, 8 and 24 h after the administration. Plasma sample was separated by
centrifugation (8000
rpm) and stored at -80 C until the sample was analyzed. Concentrations of the
test compound
in the plasma were determined by HPLC-MS/MS: the plasma was dispensed into
appropriate
tubes containing internal standard and methanol or acetonitrile. The tubes
were mixed
vigorously for 3 minutes to achieve deproteinization and then centrifuged at
8000 rpm for 5
minutes. The supernatant was transferred to an autosampler vial, injected into
the
chromatographic system, and quantified on MS/MS detector. Pharmacokinetic
parameters
including AUCo-i, Cmax, Tmax, tv2, MRT, Cl and Vd were calculated using
WinNonlin 6.3
software. Table 3 summarizes key PK parameters for test compounds (AUC(0_0:
the area under
the curve from time 0 to time t; AUC(0_.): the area under the curve from time
0 to infinity; t1i2:
Half-life; CL: clearance; Co: the concentration of drug in blood if the drug
molecules enter the
circulation and are distributed immediately (i.e., at time 0)).
Table 3. PK parameters for test compounds in ICR mice.
Compound t1/2 Cmax CL AUC(o_t) AUC(r)¨)
# (h) (u.g/L) (p g/L) (itg/L*hr)
(pg/L*hr)
d-1 0.556 8853 0.159 6357 6360
d-6 0.841 7837 0.153 6669 6681
d-7 21.3 12467 0.005 106797 196544
d-9 2.95 10673 0.045 19695 22535
d-12 0.949 8557 0.102 9865 9904
d-13 39.5 11600 0.003 108412 297675
d-20 12.2 15604 0.008 94011 121187
d-21 0.898 12227 0.137 7430 7450
d-22 28.9 8032 0.006 74120 166760
a 3.41 11718 0.042 24025 24169
b 0.85 15242 0.111 9110 9122
208
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
Example 60. CD73 enzyme inhibition assay for selected compounds.
[00500] Test compound solution preparation: Stock solution of a test
compound was
prepared in DMSO at 10 mM concentration. A series of preset 10-concentration
solutions of a
test compound were prepared by diluting the stock solution in 5-fold gradient
dilution with
DMSO.
[00501] Assay buffer (1x): 20 mm Tris, 25 mM NaCl, 1 mM MgC12, pH 7.5, 0.005%
Tween-20 (freshly prepared, and ready for use).
[00502] An aliquot of 0.25 !.IL of compound solution or DMSO (blank
control) was added
to each test well of a micro well plate. Recombinant human 5'- nucleotidase
(hCD73) was
diluted to I nM in assay buffer; and an aliquot of 25 iL hCD73 (1 nM) solution
was added to
the corresponding wells of the plate. The contents of the well were well-
mixed, and the plate
was covered with sealing film, centrifuged at 1000 rpm for 30 sec., followed
by incubation at
room temperature for 15 min. AMP, the substrate, was diluted to 60 uNI
concentration in the
assay buffer. To each reaction well was added 25 lit of AMP solution. After
mixing well, the
plate was covered with sealing film, centrifuged at 1000 rpm for 30 sec., and
incubated at 37
'V for 20 mM. The absorbance signal (end point) was measured on SPARK plate
reader at 635
nm (See PiColorLockTM Gold Phosphate Detection System (Abcam) test kit for
reference).
[00503] The reaction wells containing CD73 enzyme, substrate AMP and DMSO (no
compound) were used as positive control, and the reaction wells containing
substrate AMP and
DMSO but no CD73 enzyme were used as negative control. The percent (%)
inhibition at each
concentration of compound was calculated based on and relative to the signal
in the High and
Low control wells contained within each assay plate. The High control wells
were considered
as 0% inhibition, and the low control wells, which didn't contain any compound
but rather
DMSO (final concentration = 0.5%), as 100% inhibition. IC50 values were
obtained using
GraphPad Prism software. The calculated IC50 values are summarized in Table 4,
and the
CD73 inhibition curves for compounds d-1 and a are shown in FIGs. 3 and 4,
respectively.
Table 4. IC50 values in CD73 enzyme inhibition assay.
Compound IC50 (nM) Compound IC50 (nM) Compound IC50 (nM)
a 2.02 d-9 0.73 d-33 1.76
1.08 d-12 0.40 d-35 1.389
d-1 0.47 d-13 1.50 d-36 1.86
209
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
d-7 1.80 d-20 1.29 d-38 1.92
d-8 0.75 d-21 1.42
Example 61. CD73 cellular assay for selected compounds.
[00504] Procedures for T cell stimulation assay were the following: Dayl,
coating the plate:
diluting anti-CD3 to 1 j.ig/mL with sterile PBS; coating 96 well flat bottom
tissue culture plates
(Corning # 3599) with 50 uL of diluted anti-CD3, centrifuging (1000 rpm) for 1
min; sealing
the plate and incubating the plate at 4 C overnight for antibody
crosslinking.
[00505] Day 2, human T cell seeding: (a) washing the coated plate (from Day
1) with
sterilized PBS buffer (2 x 100 uL); (b) re-suspending T cells and adjusting
cell density to 0.5
x 106 cells/mL with growth media (X-VIVO 15 + 1% Pen/strep + 1% glutamine),
and seeding
100 uL/well (50,000 cells/well) in 96-well plates; (c) diluting CD28 antibody
for 8x working
concentration (8 vig/mL) with growth media, giving 1 vig/mL of the final
concentration,
followed by addition of CD28 antibody (25 pi) to cell plate; incubating the
plates under 5%
CO2 at 37 'V for 60 mm; (d) adding compound (25 uL, 8x dilution with growth
media) to cell
plate, and incubating the plates under 5% CO2 at 37 'DC for 60 min; (e) adding
AMP (25 litL)
and EHNA (25 uL, 8x dilution with growth media) to cell plate; (f) incubating
the plates under
5% CO2 at 37 C for 72 hours.
[00506] Day 5, collecting cell growth media and testing cell proliferation:
(a) centrifuging
the cell plate (1,000 rpm) for 5min; (b) harvesting 1501..1 of the cell
culture media from each
well to 96 well plate, followed by measurement of IFN-r expression by ELISA
test; (c) taking
a portion of the cell culture supernatant for cytokine analysis, removing the
remaining
supernatant, followed by addition of fresh growth media (X-VIVO 15 + 1%
Pen/strep + 1%
glutamine, 100 jut); (d) adding Celltiter-Glo solution (50 [IL) into each well
for the
measurement of cell proliferation by: (e) incubating the plates at room
temperature for 10 min
on a shaker; (I) transferring 1001.it solution into 96-well white plate and
taking the readout of
the Celltiter-Glo data using a TECAN Reader. Exemplary data for a selected
test compounds
are given in Table 5 (*CTG: CellTiter-Glo, Luminescent Cell Viability Assay;
#IFN-y:
Interferon Gamma, an indicator for T-cell activity).
Table 5. Test compound efficacy in CD73 cellular assay.
EC50 (nM)
Compound
CTG* IFN-y#
210
CA 03200232 2023- 5- 25

WO 2022/109741
PCT/CA2021/051688
a 17.65 34.88
7.531 14.23
d-1 3.128 1.445
d-33 4.066 2.532
[00507]
The contents of all documents and references cited herein are hereby
incorporated
by reference in their entirety.
1005081 Although this invention is described in detail with
reference to embodiments
thereof, these embodiments are offered to illustrate but not to limit the
invention. It is possible
to make other embodiments that employ the principles of the invention and that
fall within its
spirit and scope as defined by the claims appended hereto.
211
CA 03200232 2023- 5- 25

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
Compliance Requirements Determined Met 2023-06-14
Application Received - PCT 2023-05-25
National Entry Requirements Determined Compliant 2023-05-25
Request for Priority Received 2023-05-25
Letter sent 2023-05-25
Inactive: First IPC assigned 2023-05-25
Inactive: IPC assigned 2023-05-25
Inactive: IPC assigned 2023-05-25
Request for Priority Received 2023-05-25
Application Published (Open to Public Inspection) 2022-06-02

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2023-11-15

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 2023-05-25
MF (application, 2nd anniv.) - standard 02 2023-11-27 2023-11-15
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
RISEN (SUZHOU) PHARMA TECH CO., LTD.
Past Owners on Record
CHUANHAO HUANG
DAWEI CHEN
GANG CHEN
JIASHENG LU
QIGUO ZHANG
XIANQI KONG
XINGWU ZHU
YUHUA ZHANG
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) 
Cover Page 2023-08-28 1 35
Description 2023-05-24 211 8,558
Claims 2023-05-24 12 341
Representative drawing 2023-05-24 1 9
Drawings 2023-05-24 2 21
Abstract 2023-05-24 1 11
Maintenance fee payment 2023-11-14 1 26
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 64
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 1 34
Patent cooperation treaty (PCT) 2023-05-24 2 69
Patent cooperation treaty (PCT) 2023-05-24 1 36
International search report 2023-05-24 2 98
Patent cooperation treaty (PCT) 2023-05-24 1 36
Patent cooperation treaty (PCT) 2023-05-24 1 36
Patent cooperation treaty (PCT) 2023-05-24 1 36
Patent cooperation treaty (PCT) 2023-05-24 1 36
Patent cooperation treaty (PCT) 2023-05-24 1 36
National entry request 2023-05-24 12 276
Patent cooperation treaty (PCT) 2023-05-24 1 36
Courtesy - Letter Acknowledging PCT National Phase Entry 2023-05-24 2 51
Patent cooperation treaty (PCT) 2023-05-24 1 36