Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SMALL MOLECULE INHIBITORS OF GALECTIN-3
Document Type and Number:
WIPO Patent Application WO/2021/226002
Kind Code:
A1
Abstract:
The present disclosure relates to compounds of Formula (I) or Formula (II), which inhibit Gal-3, and include pharmaceutically acceptable salts, compositions comprising such compounds, and methods using and making such compounds and compositions.

Inventors:
JALAGAM PRASADA RAO (IN)
NAIR SATHEESH KESAVAN (IN)
NARA SUSHEEL JETHANAND (IN)
PANDA MANORANJAN (IN)
DEVASTHALE PRATIK (US)
REGUEIRO-REN ALICIA (US)
Application Number:
PCT/US2021/030540
Publication Date:
November 11, 2021
Filing Date:
May 04, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRISTOL MYERS SQUIBB CO (US)
International Classes:
C07D407/14; A61K31/4192; A61P29/00; C07H17/02
Domestic Patent References:
WO2018209276A12018-11-15
WO2005113568A12005-12-01
WO2005113569A12005-12-01
WO2014067986A12014-05-08
WO2017080973A12017-05-18
WO2016120403A12016-08-04
WO2018209255A12018-11-15
Foreign References:
US20140099319A12014-04-10
Other References:
IMMUNOLOGICAL REVIEWS, vol. 230, pages 160 - 171
CIRCULATION, vol. 110, pages 3121 - 3128
J PHARMACOL EXP, vol. 351, pages 336 - 343
AM J. RESPIR. CRIT. CARE MED., vol. 185, pages 537 - 546
PNAS, vol. 103, pages 5060 - 5065
AM. J. PATHOL., vol. 172, pages 288 - 298
CURR. HEART FAIL. REP., vol. 7, pages 1 - 8
"Human Lectin for Treatment of Cancer", ACS SYMPOSIUM SERIES, vol. 1115, pages 195 - 23
"Galectin Therapeutics", 7 February 2017, PRESS RELEASE
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
SYNTHESIS, vol. 6, 2007, pages 845 - 852
Attorney, Agent or Firm:
SUN, Jing G. et al. (US)
Download PDF:
Claims:
CLAIMS

We claim:

1 A compound of Formula (I) or Formula (II): or a pharmaceutically acceptable salt thereof, wherein:

X is independently selected from -C(O)-, -CH2-, and -CH2C(0)-;

Ar1 is independently phenyl or naphthyl; and wherein each ring moiety is substituted with 1 to 5 substituents selected from cyano, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, and Ci-4 haloalkoxy;

R1 is independently selected fromH, Ci-4 alkyl, Ci-4 haloalkyl, and -CH2C(0)0H;

R2 is independently selected from H, Ci-4 alkyl substituted with 0 to 1 OH, Ci-4 haloalkyl, -(CH2)O-2-C3-6 cycloalkyl, and -(CH2)o-2-phenyl substituted with 0 to 3 halogen;

R3 is independently C3-6 cycloalkyl or heterocycloalkyl including from 4 to 7 ring atoms, wherein from 1 to 2 ring atoms are each independently selected from N(, N(RB), and O, and S; wherein said ring moiety is substituted with 0 to 1 R5 and 1 R5A; R5 is independently OH, cyano, halogen, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy, and Ci-4 alkyl substituted with 0 to 1 OH;

R is independently selected from: , a bicyclic ring selected from X , wherein said bicyclic ring is substituted 0 to 2 R5C;

R5B is independently or phenyl substituted with 0 to 2 substituents selected from cyano, halogen, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy;

R5C is independently selected from: cyano, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, and Ci-4 haloalkoxy;

R5D is independently selected from R5C, . phenyl, naphthyl, pyridinyl, and primidinyl, wherein each ring moiety is substituted with 0 to 2 substituents selected from cyano, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy;

R5E is independently selected from: H, Ci-4 alkyl, Bn, -C(0)(Ci-4 alkyl),

-C(0)0(Ci-4 alkyl), R5F is independently -NH-phenyl, wherein said phenyl is substituted with 0 to 2 substituents selected from cyano, halogen, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy.

2. The compound of claim 1, wherein:

X is -C(O)-; and Ar1 is phenyl substituted with 1 to 3 halogen.

3. The compound of claim 1 or claim 2, wherein:

R3 is independently C5-6 cycloalkyl or heterocycloalkyl including from 4 to 6 ring atoms, wherein from 1 to 2 ring atoms are each independently selected from N(RB), N(RE), and O; wherein each said ring moiety is substituted with 0 to 1 R5 and 1 R5A.

4. A compound of any one of claims 1 to 3, wherein:

5. A compound of any one of claims 1 to 4, wherein:

R3 is independently selected from:

6. A compound of any one of claims 1 to 4, wherein:

7. A compound of any one of claims 1 to 4, wherein: R1 is independently H or C1-4 alkyl; and

R2 is independently selected from H, C1-4 alkyl substituted with 0 to 1 OH, C1-4 haloalkyl, -(CH2)o-i-cyclopropyl, and -CH2-(phenyl substituted with 0 to 2 halogen).

8. A compound of any one of claims 1 to 5, wherein:

R1 is independently H or G¾; and

R2 is independently selected from: H, Cfb, -CH2CH3, -CH2CH2OH, -CH2CHF2, cyclopropyl and cyclopryopylmethyl.

9. A compound of claim 1, wherein the compound is selected from the exemplified Examples or a pharmaceutically acceptable salt thereof.

10. A composition comprising a therapeutically effective amount of a compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 9, and one or more pharmaceutically acceptable carriers.

11. A compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 9, or a composition according to claim 10, for use as a medicament.

12. A use for treating fibrosis of organs (including liver, kidney, lung, heart and skin), liver diseases and conditions (including acute hepatitis, chronic hepatitis, liver fibrosis, liver cirrhosis, portal hypertension, regenerative failure, non-alcoholic steatohepatitis (NASH), liver hypofunction, and hepatic blood flow disorder), cell proliferative diseases, cancers, and conditions (including solid tumor, solid tumor metastasis, vascular fibroma, myeloma, multiple myeloma, Kaposi's sarcoma, leukemia, chronic lymphocytic leukemia (CLL)) and invasive metastasis of cancer cell), inflammatory diseases and conditions (including psoriasis, nephropathy, and pneumonia), gastrointestinal tract diseases and conditions (including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and abnormal pancreatic secretion), renal diseases and conditions, urinary tract-associated diseases and conditions (including benign prostatic hyperplasia or symptoms associated with neuropathic bladder disease, spinal cord tumor, hernia of intervertebral disk, spinal canal stenosis, and symptoms derived from diabetes), lower urinary tract diseases and conditions (including obstruction of lower urinary tract), inflammatory diseases and conditions of lower urinary tract (including dysuria and frequent urination), pancreatic diseases and conditions, abnormal angiogenesis-associated diseases and conditions (including arterial obstruction), scleroderma, brain-associated diseases and conditions (including cerebral infarction and cerebral hemorrhage), neuropathic pain and peripheral neuropathy, ocular diseases and conditions (including age-related macular degeneration (AMD), diabetic retinopathy, proliferative vitreoretinopathy (PVR), cicatricial pemphigoid, and glaucoma filtration surgery scarring) comprising administering a therapeutically effective amount of a compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 9, to a patient.

13. The use of claim 12 where the disease or condition is renal fibrosis, pulmonary fibrosis, hepatic fibrosis, arterial fibrosis, or systemic sclerosis.

Description:
SMALL MOLECULE INHIBITORS OF GALECTIN-3

CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the priority benefit of U.S. Provisional Application No. 63/020,041, filed May 05, 2020; the content of which is herein incorporated by reference in its entirety.

BACKGROUND OF THE INVENTION

Galectin-3 (Gal-3) is a b-galactoside binding lectin of about 30 KDa(Cell 76: 597- 598), that is involved in the regulation of inflammatory and fibrotic processes. (Immunological Reviews 230: 160-171). Under uncontrolled inflammation and pro- fibrotic condition, Gal-3 promotes fibroblast proliferation and transformation and mediates collagen production (Circulation 110:3121-3128).

Gal-3 is localyzed in many cellular location such as cytoplasm, nucleus, and cell surface. Gal-3 is also secreted by various cell types, mainly macrophages and monocytes into the blood stream (J Pharmacol Exp Ther 351:336-343). There are multiple lines of evidence in the literature supporting the involment of Gal-3 in the development of fibrotic process in multiple organs such as lung (Am J. Respir. Crit. Care Med. 185: 537-546), liver (PNAS 103:5060-5065) and kidney (Am. J. Pathol. 172:288-298). Gal-3 has also been identified as a biomarker for heart failure indicating that modulation of Gal-3 has potential uses in the treatment of heart failure (Curr. Heart Fail. Rep. 7:1-8). Modulation of Gal-3 can be used in the treatment of cancer since Gal-3 is involved in cell growth and differentiation playing a critical role in angiogenic, apoptotic, and metastatic pathways (Galectin-3C: Human Lectin for Treatment of Cancer. ACS Symposium Series, Vol.

1115. Chapter 12, 195-23). Recently, Gal-3 inhibitors have proven to have positive effects when used in combination immunotherapy (Galectin Therapeutics. Press Release, February 7, 2017).

Several publications and patent applications describe synthetic inhibitors of Gal-3 that are being explored as antifibrotic agents. Recent examples of these approach are W02005113568, W02005113569, WO2014067986, W02017080973, W02016120403, US20140099319 and WO2018209255.

DESCRIPTION OF THE INVENTION

The present disclosure relates to compounds of the present invention, which inhibit Gal-3, and include pharmaceutically acceptable salts, compositions comprising such compounds, and methods using and making such compounds and compositions.

In a 1st aspect, the present invention provides, inter alia, a compound of Formula (I) or Formula (II): or a pharmaceutically acceptable salt thereof, wherein:

X is independently selected from -C(O)-, -CH2-, and -CH2C(0)-;

Ar 1 is independently phenyl or naphthyl; and wherein each ring moiety is substituted with 1 to 5 substituents selected from cyano, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 haloalkyl, and Ci-4 haloalkoxy;

R 1 is independently selected fromH, C1-4 alkyl, C1-4 haloalkyl, and -CH2C(0)OH;

R 2 is independently selected from H, C1-4 alkyl substituted with 0 to 1 OH, C1-4 haloalkyl, -(CH2)O-2-C3-6 cycloalkyl, and -(CH2)o-2-phenyl substituted with 0 to 3 halogen; R 3 is independently C3-6 cycloalkyl or heterocycloalkyl including from 4 to 7 ring atoms, wherein from 1 to 2 ring atoms are each independently selected from N(, N(R B ), and O, and S; wherein said ring moiety is substituted with 0 to 1 R 5 and 1 R 5A ;

R 5 is independently OH, cyano, halogen, C1-4 haloalkyl, C1-4 alkoxy, C1-4 haloalkoxy, and Ci-4 alkyl substituted with 0 to 1 OH;

R is independently selected from: bicyclic ring selected from , wherein said bicyclic ring is substituted 0 to 2 R 5C ;

R 5B is independently or phenyl substituted with 0 to 2 substituents selected from cyano, halogen, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy;

R 5C is independently selected from: cyano, halogen, C1-4 alkyl, C1-4 alkoxy, Ci-4 haloalkyl, and C1-4 haloalkoxy; R 5D is independently selected from R 5C , . phenyl, naphthyl, pyridinyl, and primidinyl, wherein each ring moiety is substituted with 0 to 2 substituents selected from cyano, halogen, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy; R 5E is independently selected from: H, C1-4 alkyl, Bn, -C(0)(Ci-4 alkyl),

-C(0)0(Ci-4 alkyl), R 5F is independently -NH-phenyl, wherein said phenyl is substituted with 0 to 2 substituents selected from cyano, halogen, halogen, C1-4 alkyl, C1-4 alkoxy, C1-4 haloalkyl, Ci-4 haloalkoxy.

In a 2nd aspect, within the scope of the 1st aspect, wherein: X is -C(O)-; and Ar 1 is phenyl substituted with 1 to 3 halogen.

In another aspect, within the scope of the 1st or 2nd aspect, wherein: Ar 1 is phenyl substituted with 1 to 3 F.

In another aspect, within the scope of the 1st or 2nd aspect, wherein the compound is of Formula (I).

In another aspect, within the scope of the 1st or 2nd aspect, wherein the compound is of Formula (II).

In a 3rd aspect, within the scope of the 1st or 2nd aspect, wherein:

R 3 is independently C5-6 cycloalkyl or heterocycloalkyl including from 4 to 6 ring atoms, wherein from 1 to 2 ring atoms are each independently selected from N(R B ), N(R E ), and O; wherein each said ring moiety is substituted with 0 to 1 R 5 and 1 R 5A .

In a 4th aspect, within the scope of the 1st to 3rd aspects, wherein: R 3 is independently selected from: In a 5th aspect, within the scope of the 1st to 4th aspects, wherein:

In a 6th aspect, within the scope of the 1st to 4th aspects, wherein: R 3 is independently selected from:

In another aspect, within the scope of the 1st to 4th aspects, wherein: R 3 is independently selected from: and

In a 7th aspect, within the scope of the 1st to 4th aspects, wherein: R 1 is independently H or Ci-4 alkyl; and

R 2 is independently selected from H, Ci-4 alkyl substituted with 0 to 1 OH, Ci-4 haloalkyl, -(CH2)o-i-cyclopropyl, and -CH2-(phenyl substituted with 0 to 2 halogen). In an 8th aspect, within the scope of the 1st to 5th aspect, wherein:

R 1 is independently H or CH3; and

R 2 is independently selected from: H, CH3, -CH2CH3, -CH2CH2OH, -CH2CHF2, cyclopropyl and cyclopryopylmethyl.

In another aspect, within the scope of any of the 1st to 6th aspects, wherein R 1 is H.

In another aspect, within the scope of any of the 1st to 6th aspects, wherein R 1 is CH3.

In annother aspect, the invention provides a compound selected from the exemplified examples or a pharmaceutically acceptable salt thereof.

In annother aspect, the invention provides a compound selected from the exemplified Examples or a pharmaceutically acceptable salt thereof.

Unless specified otherwise, these terms have the following meanings. “Alkyl” means a straight or branched alkyl group composed of 1 to 6 carbons. “Cycloalkyl” means a monocyclic ring system composed of 3 to 7 carbons. Terms with a hydrocarbon moiety (e.g. alkoxy) include straight and branched isomers for the hydrocarbon portion which are composed of 1 to 6 carbons. “Halo” includes fluoro, chloro, bromo, and iodo. “Haloalkyl” and “haloalkoxy” include all halogenated isomers from monohalo to perhalo. “Aryl” means a monocyclic or bicyclic aromatic ring system having 5 to 12 carbon atoms wherein one or both of the rings are aromatic. Representative examples of aryl groups include, but are not limited to, indanyl, indenyl, naphthyl, phenyl, and tetrahydronaphthyl. “Heteroaryl” means a 5 to 7 membered monocyclic or 8 to 11 membered bicyclic aromatic ring system with 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Where a bonding attachment location is not specified, the bonding may be attached at any appropriate location as understood by practitioners in the art. Combinations of substituents and bonding patterns are only those that result in stable compounds as understood by practitioners in the art. Parenthetic and multiparenthetic terms are intended to clarify bonding relationships to those skilled in the art. For example, a term such as ((R)alkyl) means an alkyl substituent further substituted with the substituent R.

The invention includes all pharmaceutically acceptable salt forms of the compounds. Pharmaceutically acceptable salts are those in which the counter ions do not contribute significantly to the physiological activity or toxicity of the compounds and as such function as pharmacological equivalents. These salts can be made according to common organic techniques employing commercially available reagents. Some anionic salt forms include acetate, acistrate, besylate, bromide, chloride, citrate, fumarate, glucouronate, hydrobromide, hydrochloride, hydroiodide, iodide, lactate, maleate, mesylate, nitrate, pamoate, phosphate, succinate, sulfate, tartrate, tosylate, and xinofoate. Some cationic salt forms include ammonium, aluminum, benzathine, bismuth, calcium, choline, diethylamine, diethanolamine, lithium, magnesium, meglumine, 4-phenylcyclohexylamine, piperazine, potassium, sodium, tromethamine, and zinc.

Some of the compounds of the invention exist in stereoisomeric forms. The invention includes all stereoisomeric forms of the compounds including enantiomers and diastereomers. Methods of making and separating stereoisomers are known in the art.

The invention includes all tautomeric forms of the compounds. The invention includes atropisomers and rotational isomers. The invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include deuterium and tritium. Isotopes of carbon include 13 C and 14 C. Isotopically- labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed. Such compounds may have a variety of potential uses, for example as standards and reagents in determining biological activity. In the case of stable isotopes, such compounds may have the potential to favorably modify biological, pharmacological, or pharmacokinetic properties.

BIOLOGICAL METHODS

Gal 3 HTRF Assay

ASSAY BUFFER Composition: 25 mM HEPES, 100 mM NaCl, 0.005% Tween 20, 0.05% BSA prepared in sterile water (all reagents from Sigma).

CONTROLS:

Positive Control: 100% DMSO (lpL) + His-tagged hGal-3 (20 pL)+ B-ASF (20pL)+ Anti-His Terbium Antibody (5pL) + Strep d2 Antibody (5pL).

Negative Control: 100% DMSO ( 1 pL) + His-tagged hGal-3(20pL) +Anti His Terbium Antibody (5pL) + Strep d2 Aantibody (5pL).

STOCKS PREPARATION:

PROTOCOL: The Gal-3 assays were performed in 384 white Opti plates in three replicates at room temperature with gentle shaking at 250-300 rpmFrom the original stocks, 2.525X working stock concentrations of His- tagged recombinant human Gal-3 (hGal-3) and that of B-ASF were prepared. From the working stock, 20 mE of hGal-3 (15 nM) and 20 mE B-ASF (15 nM) were added to the plates. In Negative Control, only hGal- 3 was added. A concentration range of 50x working stocks were prepared for the compounds in 100% DMSO. Aliquots of ImE of the compounds were added to the wells and pre-incubated with 20 pL hGal-3 per well for 30 minutes Then 20 pL B-ASF wereadded and incubated for another 1 hour. To detect the signal, 5pL (final cone of 1.0 nM) terbium labelled Anti-His antibody was added and incubated for 30 min followed by adding 5pL (final cone of 20 nM) Streptavidin d2 and incubation for another 1 hour.

The assay signal was detected using HTRF screen protocol (Excitation wavelength = 340 nm, emission wavelength = 615 nm/665 nm) on Envision 2104 Multilabel Reader. Data analysed using Toolset and Curve Master. Results are reported in the experimental section (IC50 in mM). All IC50 reported were generated using the HTRF assay except when specifically indicated. Gal-3 ELISA Assay Materials: 1. Coating Buffer: Phosphate Buffered Saline (lx) - PBS

The Solution was prepared by dissolving the PBS packets procured from Sigma Aldrich (Catalogue No.: P3813-5x1 OPak) - 1 Pack in ILiter of Milli-Q water.

2. Asialofetuin from fetal Calf Serum, Type-II. Sigma Aldrich (Catalogue No. : A1908-50MG).

3. Fetal Bovine Serum. Invitrogen (Catalogue No.: 26400-044-500mL).

4. Tween-20. Sigma Aldrich (Catalogue No.: P1379-250mL).

5. BD OptEIA Enzyme Reagent Streptavidin-HRP (Catalogue No.: 554066).

6. Sulphuric Acid. Sigma Aldrich (Catalogue No.: 25,810-5). 7. Paraformaldehyde. Sigma Aldrich (Catalogue No.: P6148-500G).

8. TMB Substrate. BD Biosciences (Catalogue No.: 555214).

9. Biotin-tagged hGalectin-3 - A 0.82mg/mL stock solution (28.6kDa, 28.6713uM) of biotin tagged hGal-3 in-house synthesized by the proteomic group was used for the titration. 10. TD-139 (EXT-001109-01-001): A small molecule synthesized in-house, used as an internal standard for the small molecule screening in hGalectin-3 neutralization binding assay.

A. Protocol a. Coating of Plate: The ASF at concentration 15nM was prepared in lx PBS and was plated in the 96 well flat-bottom nunc plates (Nunc immuno plate, Maxisorp, Catalogue No.: 439454) according to the plate-map and was incubated overnight at 4°C after sealing the plates with a top-seal b. Fixing and Blocking of Plate: On the assay day, the coating solution was drained and the plates were fixed by addition of 100pL of 2%

Paraformaldehyde solution and incubating at 37°C for 30min. and washed with 300pL of wash buffer (PBS with 0.05% Tween-20) for 3 times, spin dried and taken for blocking.

The plates were later blocked with 10% FBS and incubated for lh at room temperature. Later the plate was washed with 300pL of wash buffer (PBS with 0.05% Tween-20) for 3 times. B. Incubation: After spin drying the plates from previous washing, 100pL of test compounds, at various concentrations as specified in the plate-map (pre-incubated with the hGalectin-3 or mGalectin-3 at concentration 15nM for lh at Room Temperature-RT) were added onto the plate as per the plate map. The plates were run in duplicates for data duplication and reproducibility.

These plates were incubated at RT for lh and were washed for 5 times with wash buffer, spin dried and 100pL of Streptavidin HRP (1:1000 dilutions) was added and incubated for lh at room temperature and washed for 7 times with wash buffer.

C. Detection: After spin drying the plate from previous washing, 100pL of TMB Substrate was added to each well and incubated for 15min. at room temperature. Later the reaction was stopped with 2N sulphuric acid and the plate was read in spectramax at 450nm

Results: The read out (OD) obtained were plotted against the control wells after normalization with averaged controls and analyzed for the Log of Inhibitory concentration 50 (Log ICso) values for program compounds.

Summary: The ICso values of the program compounds were as presented in the report (attached in excel format from Curve master compilation). The Plate control TD-139 had an ICso value of 10.3nM and 108.12nM for human and mouse Galectin-3 respectively.

The same was plotted on the semi-log graph.

PHARMACEUTICAL COMPOSTION AND METHODS OF USE The compounds of this invention inhibit Gal-3. Accordingly, another aspect of the invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

Another aspect of the invention is a method for treating a patient afflicted with a disease or condition selected from fibrosis of organs (including liver, kidney, lung, heart and skin), liver diseases and conditions (including acute hepatitis, chronic hepatitis, liver fibrosis, liver cirrhosis, portal hypertension, regenerative failure, non-alcoholic steatohepatitis (NASH), liver hypofunction, and hepatic blood flow disorder), cell proliferative diseases, cancers, and conditions (including solid tumor, solid tumor metastasis, vascular fibroma, myeloma, multiple myeloma, Kaposi's sarcoma, leukemia, chronic lymphocytic leukemia (CLL)) and invasive metastasis of cancer cell), inflammatory diseases and conditions (including psoriasis, nephropathy, and pneumonia), gastrointestinal tract diseases and conditions (including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and abnormal pancreatic secretion), renal diseases and conditions, urinary tract-associated diseases and conditions (including benign prostatic hyperplasia or symptoms associated with neuropathic bladder disease, spinal cord tumor, hernia of intervertebral disk, spinal canal stenosis, and symptoms derived from diabetes), lower urinary tract diseases and conditions (including obstruction of lower urinary tract), inflammatory diseases and conditions of lower urinary tract (including dysuria and frequent urination), pancreatic diseases and conditions, abnormal angiogenesis-associated diseases and conditions (including arterial obstruction), scleroderma, brain-associated diseases and conditions (including cerebral infarction and cerebral hemorrhage), neuropathic pain and peripheral neuropathy, ocular diseases and conditions (including age-related macular degeneration (AMD), diabetic retinopathy, proliferative vitreoretinopathy (PVR), cicatricial pemphigoid, and glaucoma filtration surgery scarring) with a compound of the present invention.

Another aspect of the invention is a method of treating renal fibrosis, pulmonary fibrosis, hepatic fibrosis, arterial fibrosis and systemic sclerosis comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating fibrosis of organs (including liver, kidney, lung, heart and skin) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating liver diseases and conditions (including acute hepatitis, chronic hepatitis, liver fibrosis, liver cirrhosis, portal hypertension, regenerative failure, non-alcoholic steatohepatitis (NASH), liver hypofunction, and hepatic blood flow disorder) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating cell proliferative diseases, cancers, and conditions (including solid tumor, solid tumor metastasis, vascular fibroma, myeloma, multiple myeloma, Kaposi's sarcoma, leukemia, chronic lymphocytic leukemia (CLL)) and invasive metastasis of cancer cell) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating inflammatory diseases and conditions (including psoriasis, nephropathy, and pneumonia) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating gastrointestinal tract diseases and conditions (including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and abnormal pancreatic secretion) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating renal diseases and conditions comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating urinary tract-associated diseases and conditions (including benign prostatic hyperplasia or symptoms associated with neuropathic bladder disease, spinal cord tumor, hernia of intervertebral disk, spinal canal stenosis, and symptoms derived from diabetes) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating lower urinary tract diseases and conditions (including obstruction of lower urinary tract), inflammatory diseases and conditions of lower urinary tract (including dysuria and frequent urination) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating pancreatic diseases and conditions comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating abnormal angiogenesis- associated diseases and conditions (including arterial obstruction) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating brain-associated diseases and conditions (including cerebral infarction and cerebral hemorrhage) comprising administering to a compound of the present invention to a patient.

Another aspect of the invention is a method for treating neuropathic pain and peripheral neuropathy comprising administering to a compound of the present invention to a patient. Another aspect of the invention is a method for treating ocular diseases and conditions (including age-related macular degeneration (AMD), diabetic retinopathy, proliferative vitreoretinopathy (PVR), cicatricial pemphigoid, and glaucoma filtration surgery scarring) comprising administering to a compound of the present invention to a patient.

The compounds of the invention may be used in the treatment and/or prophylaxis of conditions in which Gal-3 plays a role.

The compounds of the present invention may be used for the manufacture of a medicament for the treatment and/or prophylaxis of a condition in which inhibition of the physiological activity of Gal-3 is useful, such as diseases in which a Gal-3 receptor participates, is involved in the etiology or pathology of the disease, or is otherwise associated with at least one symptom of the disease.

The compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more, preferably one to two other agent(s).

“Therapeutically effective” means the amount of agent required to provide a meaningful patient benefit as understood by practitioners in the field of pain.

“Patient” means a person afflicted with pain and suitable for therapy as understood by practitioners in the field.

“Treatment,” “therapy,” “regimen,” and related terms are used as understood by practitioners in the field.

The compounds of this invention are generally given as pharmaceutical compositions comprised of a therapeutically effective amount of a compound of the present invention or its pharmaceutically acceptable salt and a pharmaceutically acceptable carrier and may contain conventional excipients. A therapeutically effective amount is that which is needed to provide a meaningful patient benefit. Pharmaceutically acceptable carriers are those conventionally known carriers having acceptable safety profiles. Compositions encompass all common solid and liquid forms including capsules, tablets, losenges, and powders as well as liquid suspensions, syrups, elixers, and solutions. Compositions are made using common formulation techniques, and conventional excipients (such as binding and wetting agents) and vehicles (such as water and alcohols) are generally used for compositions. See, for example, Remington ’s Pharmaceutical Sciences , 17th edition, Mack Publishing Company, Easton, PA (1985).

Solid compositions are normally formulated in dosage units and compositions providing from about 1 to 1000 mg of the active ingredient per dose are preferred. Some examples of dosages are 1 mg, 10 mg, 100 mg, 250 mg, 500 mg, and 1000 mg.

Generally, other antiretroviral agents will be present in a unit range similar to agents of that class used clinically. Typically, this is 0.25-1000 mg/unit.

Liquid compositions are usually in dosage unit ranges. Generally, the liquid composition will be in a unit dosage range of 1-100 mg/mL. Some examples of dosages are 1 mg/mL, 10 mg/mL, 25 mg/mL, 50 mg/mL, and 100 mg/mL.

The invention encompasses all conventional modes of administration; oral and parenteral methods are preferred. Generally, the dosing regimen will be similar to other agents used clinically. Typically, the daily dose will be 1-100 mg/kg body weight daily. Generally, more compound is required orally and less parenterally. The specific dosing regime, however, will be determined by a physician using sound medical judgement.

CHEMICAL METHODS

It will be evident to one skilled in the art that the present disclosure is not limited to the foregoing illustrative examples, and that it can be embodied in other specific forms without departing from the essential attributes thereof. The examples therefore should be considered in all respects as illustrative and not restrictive, reference being made to the appended claims, rather than to the foregoing examples, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced.

SECTION A

LCMS analyses were performed on Waters Acquity UPLC system coupled with Waters TUV and SQ mass detector (Column: BEH Cl 8 2.1 x 50 mm; Mobile Phase A: water with 0.05% TFA; Mobile Phase B: acetonitrile with 0.05% TFA; Gradient: 2-98% B over 1.6 minutes; Flow: 0.8 mL/min); HPLC analyses were performed on Shimadzu LC10-AT HPLC system coupled with SPD-10AV UV detector (Column YMC S5 Combiscreen ODS 4.6 x 50 mm; Mobile Phase A: 5:95 acetonitrile:water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile:water with 0.1% TFA; Gradient: 0-100% B over 40 minutes, then a 1 -minute hold at 100% B; Flow: 1 mL/min); Preparative HPLC purifications were conducted on Shimadzu LC-8 preparative HPLC system coupled with SPD 20 UV detector. Detailed conditions are described in experimental procedures.

METHODS OF PREPARATION

Analytical LC-MS/HPLC retention time reported for each example and intermediate uses one of the following general analytical LC-MS/HPLC conditions:

LCMS Conditions:

Method A: Column: Ascentis Express C18 (50x2. lmm), 2.7pm; Mobile Phase A: 5:95 acetonitrile: water with 10 mM UOAc; Mobile Phase B: 95:5 acetonitrile:water with 10 mMMUOAc; Temperature: 50°C; Gradient: 0- 100% B over 3 minutes.

Method B: Column: Ascentis Express C18 (50x2.1mm), 2.7pm; Mobile Phase A: 5:95 acetonitrile:water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile: water with 0.1% TFA; Temperature: 50°C; Gradient: 0-100% B over 3 minutes; Flow: l.lml/min.

Method C: Column-KINETEX- XB-C18 (75 X 3mm- 2.6pm); Mobile Phase A: lOmM NH4COOH IN WATER: ACN(98:02); Mobile Phase B : lOmM NH4COOH in WATER:ACN(02:98); Gradient = 20-100% B over 4 minutes; Flow rate: 1.1 mL/min; Detection: UV at 254 nm.

Method D: Column: Waters Acquity UPLC BEH C18 (2.1 x 50 mm), 1.7 p; Mobile phase A: 0.1% TFA in water; Mobile phase B: 0.1% TFA in ACN; Gradient = 20-90% B over 1.1 minute, then a 0.6 minute hold at 90% B; Temperature: 50°C; Flow rate: 0.7 mL/min; Detection: UV at 220 nm.

Method E: Column: Waters Acquity UPLC BEH C18 (2.1 x 50 mm) 1.7 p, Mobile phase A: 5mM UOAc, Acetonitrile (95:5); Mobile phase B: 5mM MUOAc: ACN (5:95), Gradient = 20-90% B over 1.1 minute, then a 0.6 minute hold at 90% B; Temperature: 50°C; Flow rate: 0.7 mL/min; Detection: UV at 220 nm

Method F: Column- ZORBAX SB-C18 (50 X 4.6mm- 5.0pm); M.phase A: lOmM NH4COOH IN WATER: ACN(98: 02); M.phase B : lOmM NH4COOH IN WATER:ACN(02:98); Gradient = 30-100% B over 4 minutes; Flow rate: 1.5 mL/min; Detection: UV at 254 nm. Prep-HPLC conditions:

Method A: Column: Waters XBridge Cl 8, 19 x 150 mm, 5-pmparticles; Mobile Phase A: 10-mM NPBOAc; Mobile Phase B: acetonitrile; Gradient: 15-50% B over 20 minutes, then a 5-minute hold at 100% B; Flow: 15 mL/min.

Method B: Column: Inertsil ODS(250*19)mm-5pm particles; Mobile Phase A: 10-mM NFBOAc -pH 4.5; Mobile Phase B: ACN; Gradient: 30-50% B over 27minutes, then a 5- minute hold at 100% B; Flow: 17 mL/min.

Method C: Column Symmetry C8 (300mm x 19mm)-7pm particles; Mobile Phase A: 10- mM NH4OAC -pH 4.5; Mobile Phase B: ACN; Gradient: 50-70% B over 24 minutes, then a 5-minute hold at 100% B; Flow: 17 mL/min.

Method D: Column: Inertsil ODS(250*19)mm-5pm particles; Mobile Phase A: 10-mM NH4OAC -pH 4.5; Mobile Phase B: ACN; Gradient: 25-60% B over 20 minutes, then a 5- minute hold at 100% B; Flow: 30 mL/min. Method E: Column: Lux-cellulose C4(250 X21.2)mm , 5pm particles; Mobile Phase A: 0.1%DEA in MeOH ; Gradient: 100% A over 20 minutes, then a 5-minute hold at 100% B; Flow: 19 mL/min.

Method F: Column: Lux-cellulose C2(250 X21.2)mm, 5pm particles; Mobile Phase A: lOmM Ammonium formate; Mobile.Phase B: ACN:MeOH(l:l); Gradient: 80% B over 20 minutes, then a 5-minute hold at 100% B; Flow: 19 mL/min.

Synthesis of carboxylic acid intermediate:

i) Tf 2 0, Py, DCM, tetrabutylammonium nitrite, DMF, 50 °C Step 4

Step 3 OTf OH

Step 1: Synthesis of (2R,3R,4S,5R,6R)-methyl 3,4,5-trihydroxy-6- (hydroxymethyl)tetrahydro-2H-pyran-2-carboxylate: Synthesized from b-D-galactose pentaacetate by following literature procedure (Ref: Synthesis, 2007, 6, 845 - 852 and references cited therein).

Step 2: Synthesis of (2S,4aR,6R,7R,8R,8aR)-methyl 7,8-dihydroxy-2- phenylhexahydropyrano[3,2-d] [l,3]dioxine-6-carboxylate: / Toluenesul Tonic acid monohydrate (1.199 g, 6.30 mmol) was added to a stirred suspension of

(2R,3R,4S,5R,6R)-methyl 3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2- carboxylate (29 g, 90 mmol) and benzaldehyde dimethyl acetal (33.8 mL, 225 mmol) in acetonitrile (563 mL) at rt under Ar atmosphere. The mixture was degassed with Ar three times and sonicated for 2 min. Then, the reaction mixture was stirred at rt for 4 h, quenched with TEA (5.77 mL, 41.4 mmol) and stirred for 10 min. The mixture was filtered and the filtrate was concentrated under reduced pressure to get the crude product which was purified via chromatography in silica gel (50-100 % EtOAc in n-hexane) to yield the title compound (16.3 g, 52.5 mmol, 58 %) as a white solid. 1H NMR (400 MHz, CHLOROFORM-d): d 7.53 - 7.49 (m, 2 H), 7.40 - 7.36 (m, 3 H), 5.57 (s, 1 H), 4.39 (dd, J= 12.5, 1.5 Hz, 1 H), 4.28 (dd, J= 4.0, 1.0 Hz, 1 H), 4.15 - 4.05 (m, 2 H), 3.87 - 3.84 (m, 4 H), 3.73 (td, .7= 9.0, 4.0 Hz, 1 H), 3.56 (q, J = 1.5 Hz, 1 H), 3.24 (d, = 2.5 Hz, 1 H), 2.63 (d, = 8.5 Hz, 1 H).

Step 3: Synthesis of (2S,4aR,6R,7S,8S,8aS)-methyl-7-acetoxy-2-phenyl-8- (((trifluoromethyl)sulfonyl)oxy)hexahydropyrano[3,2-d][l,3]d ioxine-6-carboxylate: To a solution of (2S,4aR,6R,7R,8R,8aR)-methyl 7,8-dihydroxy-2-phenylhexahydropyrano[3,2- d] [l,3]dioxine-6-carboxylate (17.3 g, 55.8 mmol) in DCM (180 mL), pyridine (18.04 mL, 223 mmol) was added at -15 °C and the mixture was stirred for 10 min. Triflic anhydride (8.48 mL, 50.2 mmol) was added drop-wise over a period of 15 min under argon and the mixture was stirred for 1 h at - 15°C. The reaction mixture was allowed to reach rt over a period of 2 h. Acetyl chloride (4.76 mL, 66.9 mmol) was added at 0 °C, and the mixture was allowed to warm to rt and stirred for 10 h. DCM (300 mL) was added, and the solution was washed with 0.7 N HC1 (150 mL), saturated sodium bicarbonate (2x 100 mL) and brine solution. The organic layer was separated and dried over sodium sulfate. The solvent was removed under reduced pressure and purified via chromatography in silica gel (30-80 % EtOAc in n-hexane) to yield the title comopund (14 g, 28.9 mmol, 52 %) as a white solid. 1H NMR (400 MHz, CHLOROFORM-d): d 7.53 (dd, J= 7.4, 2.1 Hz, 2 H), 7.44 - 7.36 (m, 3 H), 5.64 (d, J= 9.9 Hz, 1 H), 5.60 (s, 1 H), 5.00 (dd, .7= 9.9, 3.6 Hz, 1 H), 4.53 (d, J= 3.6 Hz, 1 H), 4.42 (dd, J= 12.8, 1.5 Hz, 1 H), 4.08 (dd, J= 12.8, 1.5 Hz, 1 H), 4.03 (d, J= 9.9 Hz, 1 H), 3.77 (s, 3 H), 3.59 (d, J = 1.0 Hz, 1 H), 2.10 (s, 3 H).

Step 4: Synthesis of (2S,4aR,6R,7R,8R,8aR)-methyl-7-acetoxy-8-hydroxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate: To a solution of (2S,4aR,6R,7S,8S,8aS)-methyl-7-acetoxy-2-phenyl-8-(((trifluo romethyl)sulfonyl)oxy) hexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (32 g, 66.1 mmol) in DMF (320 mL), tetrabutylammonium nitrate (50.3 g, 165 mmol) was added and degasified twice with argon and the mixture was heated at 50 °C for 6 h. Then the reaction mixture was diluted with EtOAc (500 mL), washed with water (4 x 200 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified via chromatography in silica gel (60 - 100 % EtOAc in n-hexane) to yield the title compound (15 g, 42.6 mmol, 64 %) as a yellow solid. 1H NMR (400 MHz, CHLOROFORM-d): d 7.55 - 7.51 (m, 2 H), 7.42 - 7.36 (m, 3 H), 5.55 (s, 1 H), 5.39 (dd, J= 10.3, 2.8 Hz, 1 H), 4.45 (d, J= 10.3 Hz, 1 H), 4.37 (dd, J= 12.8, 1.5 Hz, 1 H), 4.23 (t, .7= 3.1 Hz, 1 H), 4.16 - 4.13 (m, 1 H), 4.05 (dd, J= 12.8, 2.0 Hz, 1 H), 3.79 (d, J= 1.5 Hz, 1 H), 3.75 (s, 3 H), 2.10 (s, 3 H).

Step 5: Synthesis of (2S,4aR,6R,7S,8R,8aS)-methyl-7-acetoxy-2-phenyl-8- (((trifluoromethyl)sulfonyl)oxy)hexahydropyrano[3,2-d][l,3]d ioxine-6-carboxylate: To a solution of (2S,4aR,6R,7R,8R,8aR)-methyl 7-acetoxy-8-hydroxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (2.3 g, 6.53 mmol) in DCM (20 mL), pyridine (2.112 mL, 26.1 mmol) was added and the mixture was cooled to -15 °C followed by drop wise addition of triflic anhydride (1.654 mL, 9.79 mmol) under argon and stirred for 1 h at - 15°C. The reaction mixture was allowed to warm to rt and stirred for 2 h. Then, the reaction mixture was diluted with DCM (200 mL), washed with aq. 0.7 N HC1 (50 mL), aq.NaHCCh (2x 50 mL), brine solution and dried over sodium sulphate. The solvent was removed under reduced pressure and the residue was purified via chromatography in silica gel (30 - 80 % EtOAc in n-hexane) to yield the title compound (1.2 g, 2.477 mmol, 38 %) as a solid. 1H NMR (400 MHz, CHLOROFORM-d): d 7.54 - 7.49 (m, 2 H), 7.43 - 7.38 (m, 3 H), 5.60 (s, 1 H), 5.54 (dd, J= 10.5, 3.0 Hz, 1 H), 5.28 (t, J= 3.3 Hz, 1 H), 4.43 - 4.37 (m, 2 H), 4.30 (dd, J= 3.5, 1.0 Hz, 1 H), 4.11 (dd, J= 12.8, 1.5 Hz, 1 H), 3.80 (s, 3 H), 3.78 (d, J= 1.5 Hz, 1 H), 2.10 (s, 3 H).

Step 6: Synthesis of (2S,4aR,6R,7R,8S,8aR)-methyl-7-acetoxy-8-azido-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate: To a solution of (2S,4aR,6R,7S,8R,8aS)-methyl-7-acetoxy-2-phenyl-8-(((trifluo romethyl)sulfonyl)oxy) hexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (1.8 g, 41.3 mmol) in DMF (18 mL), tetrabutyl ammonium azide (3.17 g, 11.15 mmol) was added in a single portion. The mixture was degasified with Ar and heated at 50 °C for 5 h. The reaction mixture was diluted with EtOAc (200 mL), washed with water (3x 100 mL), dried over sodium sulfate and concentrated. The residue was purified via chromatography in silica gel (50-90 % EtOAc in n-hexane) to yield the title compound (1.2 g, 3.18 mmol, 86%) as a off-white solid. LC-MS, [M+18] + = 395.2, {Method C : fe = 2.37 min}. 1H NMR (300 MHz, CHLOROFORM-d): d 7.53 (dd, J= 7.2, 2.3 Hz, 2 H), 7.42 - 7.33 (m, 3 H), 5.60 (s, 1 H), 5.58 - 5.51 (m, 1 H), 4.40 - 4.33 (m, 2 H), 4.06 (dd, J= 12.7, 1.7 Hz, 1 H), 3.99 (d, J =

9.8 Hz, 1 H), 3.76 (s, 3 H), 3.50 (s, 1 H), 3.41 (dd, J= 10.4, 3.2 Hz, 1 H), 2.11 (s, 3 H).

Step 7: Synthesis of (4aR,6R,7R,8S,8aR)-methyl 7-acetoxy-8-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-phenylhexahydropyrano [3,2-d][l,3]dioxine-6- carboxylate: To a solution of (4aR,6R,7R,8S,8aR)-methyl 7-acetoxy-8-azido-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (1.2 g, 3.18 mmol) in DMF (50 mL) and water (10.00 mL). l-ethynyl-3-fluorobenzene (1.146 g, 9.54 mmol), Sodium ascorbate (0.693 g, 3.50 mmol) and copper(II) sulfate pentahydrate (0.715 g, 2.86 mmol) were added sequentially. Reaction mixture was degassed with nitrogen for 10 min and heated to 80 °C for lh. The reaction mixture was cooled to RT and diluted with water (60 ml) and DCM (50 ml) and stir for 1 h. Reaction mixture was filtered through celite pad, washed with DCM (100 ml), filtrate taken for further workup. The organic layer separated out and aqueous layer was re-extracted with DCM (2 x 100 ml), combined organic layer was washed with water (400 ml), brine (100 ml), dried the organic layer over sodium sulfate and concentrated under reduced pressure. To the crude residue, diethyl ether was added and solid was filtered through Buchner funnel and dried for lh to the title compound (1.1 g, 2.211 mmol, 69.5 % yield) as a white solid. LC-MS, [M+H] + =

498.2, {Method C : t R = 2.71 min}. ¾ NMR (400MHz, CDCh): d ppm 8.07 (s, 1H), 7.54 - 7.50 (m, 2H), 7.48 - 7.35 (m, 6H), 7.05 - 6.99 (m, 1H), 5.90 (dd, .7=11.1, 9.6 Hz, 1H), 5.52 (s, 1H), 5.21 (dd, .7=11.1, 3.4 Hz, 1H), 4.51 - 4.47 (m, 2H), 4.23 (d, .7=9.5 Hz, 1H), 4.12 (dd, .7=12.8, 1.8 Hz, 1H), 3.81 (s, 3H), 3.80 - 3.78 (m, 1H), 1.87 (s, 3H).

Step-8: Synthesis of (4ai?,6i?,7i?,8i?,8ai?)-8-(4-(3-fluorophenyl)-177-l,2,3-tria zol- l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2-<7][l,3]dioxi ne-6-carboxylic acid: To a stirred solution of (4a/76/ri7/ri85'.8a//)-methyl 7-acetoxy-8-(4-(3-fluorophenyl)-177-l,2,3- triazol- 1 -yl)-2-phenylhe\ahydropyrano| 3.2- < 7| 1 1.3 |dio\ine-6-carbo\ylate (2 g, 4.02 mmol) in Tetrahydrofuran (20 mL) and Water (10 mL) was added lithium hydroxide (0.48 g, 20.10 mmol) and stirred the mixture at rt for 2 h. After confirmation of completion of reaction with LCMS, tetrahydrofuran was removed under reduced pressure. The residue was diluted with water (100 mL) and pH adjusted to approx 2-3 using 1.5N HC1 solution. The precipitated solid was filtered and washed with water and dried under reduced pressure to yield the title compound (1.8 g, quantitative). LC-MS, [M+H] + =

442.2, {Method C : t R = 3.31 min}. ¾ NMR (400MHz, MEOH-ώ) d ppm 8.46 (s, 1H), 7.56 (dt, .7=10.2, 2.2 Hz, 2H), 7.49 - 7.40 (m, 3H), 7.37 - 7.30 (m, 3H), 7.09 (td, .7=8.4,

2.3 Hz, 1H), 5.56 (s, 1H), 5.12 (dd, .7=10.5, 3.5 Hz, 1H), 4.62 (t, .7=10.0 Hz, 1H), 4.54 (d, .7=3.5 Hz, 1H), 4.37 (d, .7=12.5 Hz, 1H), 4.18 (dd, .7=12.5, 1.5 Hz, 1H), 4.06 (d, .7=9.5 Hz, 1H), 3.89 (s, 1H). Synthesis of C2-Methoxy carboxylic acid intermediate:

Step-1: Synthesis of (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)-lH-

1.2.3-triazol-l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2 -d][l,3]dioxine-6- carboxylate: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-

1.2.3-triazol-l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2 -d][l,3]dioxine-6-carboxylic acid (1.55 g, 3.51 mmol) in DMF (10 mL), was added K2CO3 (4.85 g, 35.1 mmol) followed by Mel (1.976 mL, 31.6 mmol) and stirred at rt for 16 h. After confirmation of completion of reaction by LCMS, the reaction mass was quenched into ice water (100 mL) and stirred for 10 minutes. The solid was filtered and washed with water and dried under reduced pressure to yield the title compound as an off white solid (1.45 g, 91%). LC-MS, [M+H] + = 456.2, {Method F : t R = 1.95 min}. ¾ NMR (400MHz, MEOH-ώ) d ppm 8.41 (s, 1H), 7.58 (d, J=8.0 Hz, 1H), 7.55 - 7.50 (m, 1H), 7.47 - 7.40 (m, 3H), 7.37 - 7.32 (m, 3H), 7.07 (td, J=8.3, 2.5 Hz, 1H), 5.56 (s, 1H), 5.11 (dd, .7=11.0, 3.5 Hz, 1H), 4.68 - 4.61 (m, 1H), 4.53 (d, J=2.5 Hz, 1H), 4.32 - 4.26 (m, 1H), 4.20 - 4.13 (m, 2H), 3.89 (s, 1H), 3.82 (s, 3H).

Step-2: Synthesis of (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d] [l,3]dioxine-6- carboxylate: To a stirred solution of (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2 -d][l,3]dioxine -6- carboxylate (1.45 g, 3.18 mmol) in DMF (20 mL), was added 4A MS (1 g) and stirred for 10 minutes at rt. Then, silver oxide (3.69 g, 15.92 mmol) and Mel (0.1 mL, 15.92 mmol) were added sequentially and stirred at rt for 16 h. Reaction mass was filtered through a celite pad, washed with excess DCM (20 mL) and filtrate was concentrated under reduced pressure to afford (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-c arboxylate as an off white solid (1.3 g, 87%) which was as such taken for next step without further purification. LC-MS, [M+H] + = 470.2, {Method F : t R = 2.15 min}. ¾ NMR (400MHz, MEOH-ώ) d ppm 8.59 (s, 1H), 7.62 - 7.58 (m, 1H), 7.55 (dt, =10.0, 2.0 Hz, 1H), 7.50 - 7.42 (m, 3H), 7.40 - 7.35 (m, 3H), 7.08 (td, =8.4, 2.3 Hz, 1H), 5.58 (s, 1H), 5.19 (dd, =10.5, 3.5 Hz, 1H), 4.50 (d, J= 2.5 Hz, 1H), 4.47 - 4.43 (m, 1H), 4.29 (dd, .7=12.8, 1.8 Hz, 1H), 4.19 - 4.13 (m, 2H), 3.87 (s, 1H), 3.84 (s, 3H), 3.12 (s, 3H).

Step-3: Synthesis of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]d ioxine-6-carboxylic acid: To a stirred solution of (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]d ioxine-6-carboxylate (1.3 g, 2.8 mmol) in Tetrahydrofuran (50 mL) and Water (50 mL) was added lithium hydroxide (0.33 g, 13.85 mmol) and stirred at rt for 1 h. After confirmation of completion of reaction with LCMS, solvent was removed under reduced pressure. Then the residue was diluted with water (100 mL) and pH adjusted to approx 2-3 using aq.l.5N HC1 solution. The precipitated solid was filtered, washed with water and dried under reduced pressure to yield the title compound as an off white solid (1.1 g, 85 %). LC-MS, [M+H] + = 456.2, {Method F: t R = 0.64 min}. 1H NMR (400MHz, MEOH-d4) d ppm 8.58 (s, 1H), 7.61 (d, J=6.5 Hz, 1H), 7.55 (dd, J=10.0, 2.5 Hz, 1H), 7.50 - 7.41 (m, 3H), 7.36 (d, J=3.5 Hz, 3H), 7.11 - 7.04 (m, 1H), 5.58 (s, 1H), 5.16 (dd, J=11.0, 3.5 Hz, 1H), 4.50 (d, J=3.0 Hz, 1H), 4.42 - 4.35 (m, 1H), 4.34 - 4.29 (m, 1H), 4.16 (dd, J=12.8, 1.8 Hz, 1H), 4.06 (d, J=9.0 Hz, 1H), 3.84 (s, 1H), 3.18 (s, 3H).

Synthesis of C2-deoxy carboxylic acid intermediate (e.g., (4aR,6R,8R,8aR)-8-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-phenylhexahydropyrano [3,2-d][l,3]dioxine-6- carboxylic acid):

Step-1: Synthesis methyl (4aR,6R,7R,8S,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-7-((methylsulfonyl)oxy)-2-phenylhexahydropyran o[3,2-d][l,3]dioxine-6- carboxylate: To a stirred solution of (4aR,6R,7R,8R,8aR)-methyl 8-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2 -d][l,3]dioxine-6- carboxylate (300 mg, 0.66 mmol) in Pyridine (4 mL) was added Mesyl-Cl (0.13 mL, 1.71 mmol) at 0 °C and the reaction mixture was stirred for 6 h. The reaction mixture was quenched with ice water and stirred for 5 minutes. The obtained solid was filtered, washed with excess water and dried to afford the title compound as an off-white solid (0.26 g, 72%). LC-MS, [M+H] + = 534.2, {Method C : t R = 1.990 min}.

Step-2: Synthesis of methyl (4aR,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-2-phenyl-4,4a,8,8a-tetrahydropyrano[3,2-d][l,3 ]dioxine-6-carboxylate: To a solution of (4aR,6R,7R,8S,8aR)-methyl 8-(4-(3-fhiorophenyl)-lH-l,2,3-triazol-l-yl)-7- ((methylsulfonyl)oxy)-2-phenylhexahydropyrano[3,2-d][l,3]dio xine-6-carboxylate (260 mg, 0.487 mmol) in 2,6-Lutidine (25 mL) was added aluminum oxide (Basic) (2.5 g, 24.37 mmol) at rt and the reaction mixture was heated at 50 °C for 16 h. The solvent was removed under reduced pressure and crude residue was purified via chromatography in silica gel (2 - 3 % MeOH in DCM) to yield the title compound (0.19 g, 83%) as a pale yellow solid. LC-MS, [M+l] + = 438.2, {Method C : t R = 1.966 min}. 1H NMR (400 MHz, CHLOROFORM-d) d ppm 7.93 (s, 1 H), 7.50 - 7.56 (m, 2 H), 7.28 - 7.41 (m, 6 H), 7.00 - 7.05 (m, 1 H), 6.00 - 6.04 (m, 1 H), 5.96 - 5.99 (m, 1 H), 5.57 (s, 1 H), 4.64 (dd, J=12.8, 1.8 Hz, 1 H), 4.50 - 4.54 (m, 1 H), 4.29 - 4.34 (m, 1 H), 4.12 - 4.20 (m, 1 H), 3.85 (s, 3 H).

Step-3: Synthesis of methyl (4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-c arboxylate: To a degassed solution (4aR,8R,8aR)-methyl 8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-phenyl- 4,4a,8,8atetrahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (0.22 g, 0.503 mmol) in EtOAc (8 mL), was added palladium on carbon (10% w/w, 50% wet) (54 mg, 0.05 mmol) and stirred the mixture at rt under hydrogen pressure (~1 atm) for 12 h. The reaction mixture was filtered through Celite pad, washed with excess EtOAc/MeOH (1:1, 30 mL) and filtrate was concentrated under reduced pressure to give the title compound (0.2 g, 90%) as an off-white solid. LC/MS [M+H] + = 440.2, {Method B : tR = 1.25 min }. 1H NMR (400 MHz, CHLOROFORM-d) d ppm 7.94 (s, 1 H), 7.48 - 7.56 (m, 2 H), 7.34 - 7.44 (m, 6 H), 6.96 - 7.11 (m, 1 H), 5.54 (s, 1 H), 5.13 - 5.26 (m, 1 H), 4.50 (dd, J=12.5, 1.5 Hz, 1 H), 4.27 - 4.42 (m, 2 H), 4.03 - 4.20 (m, 1 H), 3.82 (s, 3 H), 3.70 (d, J=1.5 Hz, 1 H), 2.58 - 2.69 (m, 1 H), 2.34 - 2.42 (m, 1 H).

Step-4: Synthesis of (4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylic acid: To a stirred solution of (4aR,6R,8R,8aR)-methyl8-(4-(3-fluorophenyl)-lH-l,2,3-triazol -l-yl)-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylate (0.2 g, 0.455 mmol) in THF (8 mL), Water (2.0 mL), LiOH (0.044 g, 1.821 mmol) was added at rt and the stirring continued for 2 h. After confirmation of completion of reaction with LCMS, tetrahydrofuran was removed under reduced pressure. The residue was diluted with water (100 mL) and pH adjusted to approx 2-3 using 1.5N HC1 solution. The precipitated solid was filtered and washed with water and dried under reduced pressure to yield the title compound (0.17 g, 88%) as an off-white solid. LC/MS [M+H] + = 426.2, {Method A : tR = 0.79 min }.

Example la and lb: Synthesis of (27?, 3R, 4S, 5R, 6R)-4-(4-(3-fluorophenyl)- 1//- 1.2.3- triazol- 1 -y l )-/V-(( 15'.2/?.3/?)-3-(4-(3-fluorophenyl)- \ H- \ 2.3-triazol- 1 -yl)-2- hydroxycyclohexyl)-5-hydroxy-6-(hydroxymethyl)-3-methoxytetr ahydro-277-pyran-2- carboxamide (Isomer 1 and Isomer 2). Step 1 a er, racemate relative stereochemistry ste P 3

Step-1: Synthesis of 3-azidocyclohex-l-ene: To a stirred solution of 3- bromocyclohex-l-ene (7.1 mL, 62.1 mmol) in mixture of CCU (100 mL)/water (100 mL) was added sodium azide (14. lg, 217 mmol) at rt and stirred for 48 h. Aq. layer was separated and extracted with DCM (2X50 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated to give 3- azidocyclohex-l-ene (6.6 g, 86 %) as pale yellow liquid. ¾ NMR (400 MHz, CHLOROF ORM-ύG) d ppm 5.94 - 6.07 (m, 1 H), 5.66 - 5.75 (m, 1 H), 3.83 - 3.92 (m, 1 H), 1.96 - 2.17 (m, 2 H), 1.84 - 1.94 (m, 1 H), 1.69 - 1.80 (m, 2 H), 1.57 - 1.68 (m, 1 H).

Step-2: Synthesis of (lR,2R,6S)-2-azido-7-oxabicyclo[4.1.0]heptane (racemate): To a solution of 3-azidocyclohex-l-ene (5.5 g, 44.7 mmol) in dichloromethane (200 mL) was added mCPBA (15.4 g, 67.0 mmol) (dissolved in 20 mL DCM) at 0 °C. The reaction mixture was allowed to reach rt and stirred for 16 h. The reaction mixture was cooled to 0 °C, precipitated solid was filtered and washed with DCM (50 mL). The filtrate was washed with sat. Na2S03 solution, aq.10% NaHCCb solution, brine solution, dried over sodium sulphate and concentrated to give crude product. The crude residue was purified by flash chromatography (0-2% EtOAc in n-hexane) to afford (lR,2R,6S)-2-azido-7- oxabicyclo[4.1.0]heptane (1.9 g, 30%, cis-isomer) and (lS,2R,6R)-2-azido-7- oxabicyclo[4.1.0]heptane (2.9 g, 46%, trans isomer).

Cis-isomer: ¾ NMR (400 MHz, CHLOROF ORM-ri) d ppm 3.50 - 3.66 (m, 1 H), 3.32 (dd, =4.0, 2.0 Hz, 1 H), 3.25 - 3.28 (m, 1 H), 1.77 - 1.97 (m, 2 H), 1.58 - 1.74 (m, 3 H), 1.19 - 1.38 (m, 1 H).

Trans-isomer: ¾ NMR (400 MHz, CHLOROF ORM-ri) d ppm 3.83 (t, J=6.8 Hz,

1 H), 3.19 - 3.28 (m, 1 H), 3.09 (d, .7=3.5 Hz, 1 H), 1.97 - 2.08 (m, 1 H), 1.76 - 1.93 (m, 2 H), 1.45 - 1.53 (m, 1 H), 1.28 - 1.40 (m, 2 H).

Step-3: Synthesis of l-((lS,2R,6R)-7-oxabicyclo[4.1.0]heptan-2-yl)-4-(3- fluorophenyl)-lH- 1,2, 3-triazole (racemate): To a solution of (lS,2R,6R)-2-azido-7- oxabicyclo[4.1.0]heptane (0.5 g, 3.59 mmol) in DMF (5 mL) and water (1.5 mL) was added sodium ascorbate (0.71 g, 3.59 mmol), copper(II) sulfate pentahydrate (0.81 g, 3.23 mmol) and 3-fluorophenylacetylene (1.7 mL, 14.37 mmol) at rt. The reaction mixture was heated at 85 °C for 30 min. The reaction mixture was cooled to rt, diluted with 1 : 1 DCM (50 mL) and water (50 mL) and stirred at rt for 15 min. The reaction mixture was filtered through Celite pad and washed with DCM (20 mL). From the filtrate, organic layer was separated and aqueous layer re-extracted with DCM (2x20 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated to give crude residue. The crude residue was purified by flash chromatography (40-50% EtOAc in n-hexane) to afford l-((lS,2R,6R)-7- oxabicyclo[4.1.0]heptan-2-yl)-4-(3-fluorophenyl)-lH-l, 2, 3-triazole (0.53 g, 57%) as pale yellow solid. LC/MS [M+H] + = 260.2, tR= 2.266 min (Method C).

Step-4: Synthesis of (lR,2R,6S)-2-azido-6-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)cyclohexanol (racemate): To a solution of l-((lR,2S,6S)-7-oxabicyclo[4.1.0]heptan- 2 -yl)-4-(3 -fluorophenyl)- lH-1, 2, 3-triazole (200 mg, 0.771 mmol) in MeOH (16 mL) and water (4.00 mL) was added ammonium chloride (103 mg, 1.928 mmol) and sodium azide (251 mg, 3.86 mmol) at rt. The reaction mixture was heated at 75 °C for 16 h. The reaction mixture was cooled to rt, MeOH was removed under reduced pressure and extracted with EtOAc (3X25 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated under reduced pressure to give crude residue. The crude residue was purified by flash chromatography (20 - 25% EtOAc in n-hexane) to afford (lR,2R,6S)-2-azido-6-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)cyclohexanol (0.22 g, 98% ) as an off-white solid. LC/MS [M+H] + = 303.2, tR = 1.699 min (Method F).

Step-5: Synthesis of (lS,2R,6S)-2-amino-6-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)cyclohexanol (racemate): To a degassed solution of (lR,2R,6S)-2-azido-6-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)cyclohexanol (210 mg, 0.695 mmol) in MeOH (20 mL) was added palladium on carbon (10% w/w, 50% wet) (74 mg, 0.069 mmol) and stirred the mixture at rt under hydrogen pressure (~1 atm) for 2 h. The reaction mixture was filtered through Celite pad, washed with excess MeOH (10 mL) and filtrate was concentrated under reduced pressure to give (lS,2R,6S)-2-amino-6-(4-(3-fluorophenyl)- 1 H- 1 ,2,3-triazol- 1 -yl)cy clohexanol (170 mg, 71%). LC/MS [M+H] + = 277.0, t R = 1.255 min (Method F).

Step-6: Synthesis of ((2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-N-((lR,2S, 3S)-3-(4-(3-fluorophenyl)-lH-l, 2, 3-triazol-l-yl)-2-hydroxy cyclohexyl)- 5-hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-c arboxamide: To a solution of (2S,4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol -l-yl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxy lic acid (50 mg, 0.110 mmol) (50 mg, 0.110 mmol) in DMF (2 mL) was added DIPEA (0.06 mL, 0.329 mmol) and HATU (62.6 mg, 0.165 mmol) at rt and stirred for 15 min. Then (lS,2R,6S)-2- amino-6-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)cyclohexan ol (36.4 mg, 0.132 mmol) was added and stirred the mixture at rt for 1 h. The reaction mixture was quenched with ice cold water (10 mL) and stirred for 15 min. The resultant solid was filtered, washed with excess water and residue was dried to afford diastereomeric mixture of crude residue. The crude residue was further purified by prep-HPLC [Method F] to obtain two isomers: Isomer 1: 20 mg, 21 % yield; LC/MS [M+H] + = 714.2, tR = 2.897 min (Method C). Isomer 2: 15 mg, 19 % yield; LC/MS [M+H] + = 714.2, t R = 2.878 min (Method C).

Step-7: A solution of ((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-2- hydroxycyclohexyl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][ l,3]dioxine-6- carboxamide (20 mg, 0.028 mmol, isomer 1) in 70% aqueous acetic acid (5 mL) was stirred overnight at 70 °C. The reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude product was purified by preparative LCMS (Method A) to yield Example la as a white solid (4.5 mg, 25% yield). LC-MS, [M+H] + = 626.2, [ tR = 1.688 min, Method A] and & [ tR = 1.710 min, Method B] 1H NMR (400MHz, MEOH-d4) d ppm 8.59 (s, 1H), 8.39 (s, 1H), 7.66 (dd, J=7.8, 4.2 Hz, 2H), 7.62 - 7.56 (m, 2H), 7.49 - 7.41 (m, 2H), 7.08 (m, 2H), 4.91 (dd, J=10.8, 2.7 Hz, 1H), 4.57 - 4.44 (m, 1H), 4.28 - 4.21 (m, 1H), 4.11 (d, J=2.7 Hz, 1H),

4.03 - 3.89 (m, 3H), 3.82 - 3.77 (m, 2H), 3.75 - 3.69 (m, 1H), 3.19 - 3.12 (m, 3H), 2.24 - 2.12 (m, 2H), 2.05 (d, J=11.7 Hz, 1H), 1.95 (d, J=13.2 Hz, 1H), 1.68 - 1.54 (m, 2H). hGal3 ICso (ELISA)= 0.7 mM; hGal3 ICso (HTRF)= 0.29 mM.

Example lb: A solution of ((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-2- hydroxycyclohexyl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][ l,3]dioxine-6- carboxamide (15 mg, 0.021 mmol, isomer 2) in 70% aqueous acetic acid (5 mL) was stirred overnight at 70 °C. The reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude product was purified by preparative LCMS (Method A) to yield Example lb as a white solid (1.1 mg, 8 % yield). LC-MS, [M+H] + = 626.2, [ tR = 1.690 min, Method A] and & [ tR = 1.688 min, Method B] 1H NMR (400MHz, MEOH-d4) d ppm 8.63 (s, 1H), 8.40 (s, 1H), 7.72 - 7.57 (m, 4H), 7.51 - 7.42 (m, 2H), 7.13 - 7.05 (m, 2H), 4.93 (dd, J=10.8, 2.7 Hz, 1H), 4.58 -

4.45 (m, 1H), 4.27 (t, J=9.9 Hz, 1H), 4.12 (d, J=2.4 Hz, 1H), 4.02 - 3.89 (m, 3H), 3.85 - 3.79 (m, 2H), 3.77 - 3.71 (m, 1H), 3.20 (s, 3H), 2.26 - 2.14 (m, 2H), 2.12 (br. s., 1H), 1.98 (d, J= 11.5 Hz, 1H), 1.71 - 1.59 (m, 2H). hGal3 ICso (ELISA)= 16 mM.

The Examples in Table 1 were prepared in an analogous fashion to Examples la and lb, substituting l-fluoro-3-ethynylbenzene with the appropriate acetylenes in the synthetic sequence

Table 1

Example 6a and 6b: Synthesis of (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((lR,2S,3S)-3-(5-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-2- hydroxycyclohexyl)-5-hydroxy-6-(hydroxymethyl)-3-methoxytetr ahydro-2H-pyran-2- carboxamide (isomer 1 and isomer 2)

Step-1: Synthesis of l-((lR,2S,6S)-7-oxabicyclo[4.1.0]heptan-2-yl)-5-(3- fluorophenyl)-lH-l,2,3-triazole(racemate): To a stirred solution of (lR,2S,6S)-2-azido-7- oxabicyclo[4.1.0]heptane (100 mg, 0.719 mmol) and 3-fluorophenylacetylene (0.34 mL, 2.87 mmol) in toluene (3 mL) was added chloro(l,5-cyclooctadiene)(pentamethyl cyclopentadienyl)ruthenium (8.19 mg, 0.022 mmol). The reaction mixture was heated at 85 °C for 4 h. The reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was purified by flash chromatography (40 - 50% EtOAc in n-hexane) to afford the title compound (175 mg, 92%) as an off-white solid. LC/MS [M+H] + = 260.2, te = 2.12 min (Method C).

Step-2: Synthesis of (lR,2R,6S)-2-azido-6-(5-(3-fluorophenyl)-lH-l,2,3-triazol-

1-yl)cyclohexanol(racemate): To a solution of l-((lR,2S,6S)-7-oxabicyclo[4.1.0]heptan-

2-yl)-5 -(3 -fluorophenyl)- lH-1, 2, 3-triazole (170 mg, 0.656 mmol) in MeOH (8 mL) and water (2 mL) was added ammonium chloride (88 mg, 1.639 mmol) and sodium azide

(213 mg, 3.28 mmol) at rt. The reaction mixture was heated at 75 °C for 16 h. The reaction mixture was cooled to rt, MeOH was removed under reduced pressure and extracted with EtOAc (3X25 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated under reduced pressure to give crude residue. The crude residue was purified by flash chromatography (20 - 25% EtOAc in n-hexane) to afford the title compound (0.15 g, 72% ) as a brown solid. LC/MS [M+H] + = 303.5, t R = 1.10 min (Method E).

Step-3: Synthesis of (lS,2R,6S)-2-amino-6-(5-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)cyclohexanol(racemate): To a degassed solution of (lR,2R,6S)-2-azido-6-(5-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)cyclohexanol (150 mg, 0.496 mmol) in MeOH (10 mL) was added palladium on carbon (10% w/w, 50% wet) (53 mg, 0.050 mmol) and stirred the reaction mixture at rt under hydrogen pressure (~1 atm) for 2 h. The reaction mixture was filtered through Celite pad, washed with excess MeOH (10 mL) and filtrate was concentrated under reduced pressure to give the title compound (68 mg, 40%).

LC/MS [M+H] + = 277.2, tR = 1.18 min (Method F).

Step-4: Synthesis of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-N-((lR,2S, 3S)-3-(5-(3-fluorophenyl)-lH-l, 2, 3-triazol-l-yl)-2-hydroxy cyclohexyl)- 7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carbo xamide: To a solution of ((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l -yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylic acid (50 mg, 0.110 mmol) in DMF (2 mL) was added DIPEA (0.06 mL, 0.329 mmol) and HATU (62.6 mg, 0.165 mmol) at rt and stirred for 15 min. Then (lS,2R,6S)-2-amino-6-(5-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)cyclohexanol (36.4 mg, 0.132 mmol) was added and stirred the mixture at rt for 1 h. The reaction mixture was quenched with ice cold water (10 mL) and stirred for 15 min. The resultant solid was filtered, washed with excess water and the residue was dried to afford diastereomeric mixture of crude residue. The crude residue was further purified by prep-HPLC [Method E] to obtain isomer 1 and isomer 2.

Isomer 1: 10 mg, 13% yield; LC/MS [M+H] + = 714.2, tR = 2.461 min (Method F).

Isomer 2: 15 mg, 17% yield; LC/MS [M+H] + = 714.2, tR = 2.457 min (Method F).

Step-5: ((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l -yl)-N- ((lR,2S,3S)-3-(5-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-h ydroxycyclohexyl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxa mide (Isomer 1) (10 mg, 0.014 mmol) was suspended in aq. 70% acetic acid (3 mL, 52.4 mmol) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford Example 6a (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl )- N-((1R, 2S, 3S)-3-(5-(3-fluorophenyl)-lH-l, 2, 3-triazol-l-yl)-2 -hydroxy cyclohexyl)-5- hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-car boxamide Isomer 1 (2.9 mg, 33%). LC/MS [M+H] + = 626.1, t R = 1.667 min (Method A); 1H NMR (400MHz, MEOH-d4) d ppm 8.63 (s, 1H), 7.80 (s, 1H), 7.70 (d, J=7.8 Hz, 1H), 7.66 -

7.54 (m, 2H), 7.51 - 7.38 (m, 3H), 7.29 (td, J=8.6, 2.4 Hz, 1H), 7.10 (td, J=8.6, 2.4 Hz, 1H), 4.93 (dd, J=10.6, 2.8 Hz, 1H), 4.38 - 4.30 (m, 1H), 4.29 - 4.16 (m, 2H), 4.12 (d, J=2.7 Hz, 1H), 3.94 (d, J=9.3 Hz, 1H), 3.92 - 3.79 (m, 3H), 3.77 - 3.69 (m, 1H), 3.19 (s, 3H), 2.28 - 2.16 (m, 1H), 2.06 (d, J=10.8 Hz, 2H), 1.89 (d, J=13.4 Hz, 1H), 1.67 - 1.44 (m, 2H). hGal3 IC50 = 27 mM.

Example 6b: (2S,4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol -l- yl)-N-((lR,2S,3S)-3-(5-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-2-hydroxycyclohexyl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxa mide (Isomer 2) (15 mg, 0.021 mmol) was suspended in 70% Aq. acetic acid (5 mL, 87 mmol) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl )-N- ((lR,2S,3S)-3-(5-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-h ydroxycyclohexyl)-5- hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-car boxamide Isomer 2 (5.5 mg, 42%). LC/MS [M+H] + = 626.1, t R = 1.683 min (Method A); Ή NMR (400MHz, MEOH-d4) d ppm 8.61 (s, 1H), 7.81 (s, 1H), 7.68 (d, J=8.1 Hz, 1H), 7.65 -

7.55 (m, 2H), 7.51 - 7.40 (m, 3H), 7.32 - 7.26 (m, 1H), 7.14 - 7.07 (m, 1H), 4.92 (dd, J=10.9, 3.1 Hz, 1H), 4.39 - 4.31 (m, 1H), 4.29 - 4.16 (m, 2H), 4.12 (d, J=2.7 Hz, 1H), 3.94 (d, J=9.3 Hz, 1H), 3.90 (br. s., 1H), 3.85 - 3.79 (m, 2H), 3.77 - 3.72 (m, 1H), 3.17 (s, 3H), 2.28 - 2.16 (m, 1H), 2.11 - 1.98 (m, 2H), 1.88 (d, J=13.7 Hz, 1H), 1.63 - 1.47 (m, 2H). hGal3 ICso = 0.90 mM.

Synthetic Scheme for Indazole derivatives:

Step-1: Synthesis of (lR,2R,6S)-2-azido-6-(4-fluoro-lH-indazol-l- yl)cyclohexanol and (lR,2R,6S)-2-azido-6-(4-fluoro-2H-indazol-2- yl)cyclohexanol(racemate): To a solution of (lS,2R,6R)-2-azido-7- oxabicyclo[4.1.0]heptane (300 mg, 2.156 mmol) and 4-fluoro-lH-indazole (308 mg, 2.264 mmol) in DMSO (3 mL) was added DBU (0.975 mL, 6.47 mmol) at rt. The reaction mixture was heated at 100 °C for 16 h. The reaction mixture was cooled to rt, extracted with EtOAc (3x 30 mL), washed with water, brine, dried over sodium sulphate and concentrated under reduced pressure to give crude product. The crude residue was purified by prep-HPLC (Method B) to afford Nl-regioisomer (lR,2R,6S)-2-azido-6-(4- fluoro-lH-indazol-l-yl)cyclohexanol (120 mg, 20%) and N2-regioisomer (lR,2R,6S)-2- azido-6-(4-fluoro-2H-indazol-2-yl)cyclohexanol (150 mg, 20%).

Nl-regioisomer: ¾NMR (400 MHz, CHLOROFORM-ri) d ppm 8.12 (d, .7=0 8 Hz, 1 H), 7.16 - 7.39 (m, 2 H), 6.76 - 6.83 (m, 1 H), 4.29 - 4.38 (m, 1 H), 4.15 - 4.23 (m,

1 H), 3.48 - 3.57 (m, 1 H), 2.39 (d, .7=3.5 Hz, 1 H), 2.13 - 2.20 (m, 1 H), 2.04 - 2.11 (m, 1 H), 1.94 - 2.00 (m, 1 H), 1.43 - 1.68 (m, 3 H). LC/MS [M+H] + = 276.2, t R = 1.98 min (Method F).

N2-regioisomer: 'H NMR (400 MHz, CHLOROFORM-ri) d ppm 8.06 (d, .7=0 8 Hz, 1 H), 7.43 (s, 1 H), 7.11 - 7.25 (m, 1 H), 6.66 - 6.73 (m, 1 H), 4.19 - 4.36 (m, 1 H), 4.06 (t, J= 9.6 Hz, 1 H), 3.46 - 3.57 (m, 1 H), 2.20 - 2.26 (m, 1 H), 2.09 - 2.17 (m, 2 H), 1.96 - 2.02 (m, 1 H), 1.50 - 1.59 (m, 2 H). LC/MS [M+H] + = 276.2, t R = 2.12 min (Method C).

Step-2: Synthesis of (lS,2R,6S)-2-amino-6-(4-fluoro-lH-indazol-l- yl)cyclohexanol(racemate): To a degassed solution of (lR,2R,6S)-2-azido-6-(4-fluoro- lH-indazol-l-yl)cyclohexanol (100 mg, 0.363 mmol) in MeOH (5 mL) was added palladium on carbon (10% w/w) (39 mg, 0.036 mmol) and stirred the reaction mixture at rt under hydrogen pressure (~1 atm) for 2 h. The reaction mixture was filtered through Celite pad, washed with excess MeOH (10 mL) and filtrate was concentrated under reduced pressure to give (lS,2R,6S)-2-amino-6-(4-fluoro-lH-indazol-l-yl)cyclohexanol (65 mg, 62 %). LC/MS [M+H] + = 250.0, t R = 1.26 min (Method F).

Step-3: Synthesis of (4aR,6R,7R,8R,8aR)-N-((lR,2S,3S)-3-(4-fluoro-lH-indazol-l-yl )-2- hydroxycyclohexyl)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamide: To a solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylic acid (50 mg, 0.110 mmol) in DMF (5 mL) was added DIPEA (0.19 mL, 1.098 mmol) and HATU (83 mg, 0.220 mmol) at rt and stirred for 15 min. Then (lS,2R,6S)-2-amino-6-(4-fluoro-lH-indazol-l- yl)cyclohexanol (racemate) (28 mg, 0.110 mmol) was added and stirred the mixture at rt for 1 h. The reaction mixture was quenched with ice cold water (10 mL) and stirred for 15 min. The resultant solid was filtered, washed with excess water and residue was dried to afford diastereomeric mixture of crude residue. The crude residue was purified by prep-HPLC [Method E] to obtain Isomerl and Isomer 2.

Isomer 1: 21 mg, 27% yield; LC/MS [M+H] + = 687.2, tR = 2.54 min (Method F).

Isomer 2: 20 mg, 26% yield; LC/MS [M+H] + = 6872, tR = 2.55 min (Method F).

Step-4: Synthesis of (2R,3R,4S,5R,6R)-N-((lR,2S,3S)-3-(4-fluoro-lH-indazol-l- yl)-2-hydroxycyclohexyl)-4-(4-(3-fluorophenyl)-lH-l,2,3-tria zol-l-yl)-5-hydroxy-6- (hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-carboxamide (Isomer 1 and 2): (4aR,6R,7R,8R,8aR)-N-((lR,2S,3S)-3-(4-fluoro-lF[-indazol-l-y l)-2 -hydroxy cyclohexyl)- 8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-methoxy-2-phe nylhexahydropyrano[3,2- d][l,3]dioxine-6-carboxamide (Isomer-1) (20 mg, 0.029 mmol) was suspended in aq.70% acetic acid (5 mL) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford Example 7a, (2R,3R,4S,5R,6R)-N- ((lR,2S,3S)-3-(4-fluoro-lH-indazol-l-yl)-2-hydroxycyclohexyl )-4-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxy tetrahydro-2H-pyran-2- carboxamide Isomer 1 (7.8 mg, 45%). LC/MS [M+H] + = 599.1, tR = 1.77 min (Method A); 1H NMR (400MHz, MEOH-d4) d = 8.60 (s, 1H), 8.14 (s, 1H), 7.68 (d, J=8.1 Hz,

1H), 7.62 (d, J=10.3 Hz, 1H), 7.49 - 7.43 (m, 2H), 7.39 - 7.34 (m, 1H), 7.12 - 7.07 (m, 1H), 6.83 - 6.81 (m, 1H), 4.93 (dd, J=10.9, 2.8 Hz, 1H), 4.57 (br.s, 1H), 4.27 - 4.12 (m, 1H), 4.13 - 4.05 (m, 3H), 4.03 (d, J=9.3 Hz, 1H), 3.93 - 3.73 (m, 3H), 3.16 (s, 3H), 2.16 - 1.92 (m, 4H), 1.71 - 1.52 (m, 2H). hGal3 IC50 = 0.64 mM.

Example 7b: Prepared in a similar fashion as described for Example 7a, Isomer 1 using (4aR,6R,7R,8R,8aR)-N-((lR,2S,3S)-3-(4-fluoro-lH-indazol-l-yl )-2- hydroxycyclohexyl)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamide (Isomer 2) (20 mg, 0.029 mmol). The crude residue was purified by prep-HPLC [Method A] to afford (2R,3R,4S,5R,6R)-N-((lR,2S,3S)-3-(4-fluoro-lH-indazol-l-yl)- 2-hydroxycyclohexyl)-4- (4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5-hydroxy-6-(hydr oxymethyl)-3- methoxytetrahydro-2H-pyran-2-carboxamide Isomer 2 (8.1 mg, 46%). LC/MS [M+H] + = 599.1, t R = 1.747 min (Method A); ¾ NMR (400MHz, MEOH-d4) d ppm 8.63 (s, 1H),

8.13 (s, 1H), 7.70 (d, J=7.8 Hz, 1H), 7.64 (d, J=10.3 Hz, 1H), 7.50 - 7.43 (m, 2H), 7.38 - 7.33 (m, 1H), 7.12 - 7.07 (m, 1H), 6.82 - 6.80 (m, 1H), 4.93 (dd, J=10.6, 2.8 Hz, 1H),

4.60 - 4.47 (m, 1H), 4.28 (t, J=10.0 Hz, 1H), 4.15 - 3.98 (m, 3H), 3.94 (d, J=9.3 Hz, 1H), 3.87 - 3.76 (m, 2H), 3.76 - 3.67 (m, 1H), 3.18 (s, 3H), 2.22 - 1.89 (m, 4H), 1.77 - 1.56 (m, 2H). hGab ICso = 23 mM.

Example 8: Synthesis of (2R,3R,4S,5R,6R)-N-((lR,2S,3S)-3-(4-fluoro-2H-indazol-2-yl)- 2-hydroxycyclohexyl)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-5-hydroxy-6-

(hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-carboxamid e.

Example 8

Step 1: Synthesis of (lS,2R,6S)-2-amino-6-(4-fluoro-2H-indazol-2- yl)cyclohexanol(racemate): To a degassed solution of (lR,2R,6S)-2-azido-6-(4-fluoro- 2H-indazol-2-yl)cyclohexanol (150 mg, 0.545 mmol)) in MeOH (10 mL) was added palladium on carbon (10% w/w) (58 mg, 0.054 mmol) and stirred the reaction mixture at rt under hydrogen pressure (~1 atm) for 2 h. The reaction mixture was filtered through Celite pad, washed with excess MeOH (10 mL) and filtrate was concentrated under reduced pressure to give the title compound (80 mg, 45%) as an off white solid. LC/MS [M+H] + = 250.2, t R = 0.74 min (Method C).

Step-2: Synthesis of (4aR,6R,7R,8R,8aR)-N-((lR,2S,3S)-3-(4-fluoro-2H-indazol- 2-yl)-2-hydroxycyclohexyl)-8-(4-(3-fluorophenyl)-lH-l,2,3-tr iazol-l-yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamide: To a solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylic acid (50 mg, 0.110 mmol) in DMF (5 mL) was added DIPEA (0.19 mL, 1.098 mmol) and HATU (83 mg, 0.220 mmol) at rt and stirred for 15 min. Then (lS,2R,6S)-2-amino-6-(4-fluoro-2H-indazol-2- yl)cyclohexanol(racemate) (27.4 mg, 0.110 mmol) was added at rt and stirred for 1 h.

The reaction mixture was quenched with ice cold water (10 mL) and stirred for 15 min. The resultant solid was filtered, washed with excess water and residue was dried to afford diastereomeric mixture of crude residue. The crude residue was further purified by prep- HPLC [Method E] to obtain Isomerl and Isomer 2.

Isomer 1: 10 mg, 13% yield; LC/MS [M+H] + = 687.0, tR = 2.318 min (Method F).

Isomer 2: 12 mg, 15% yield; LC/MS [M+H] + = 687.2, t R = 2.328 min (Method F).

Step-3: (4aR,6R,7R,8R,8aR)-N-((lR,2S,3S)-3-(4-fluoro-2H-indazol-2-yl )-2- hydroxycyclohexyl)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamide (Isomer 1) (10 mg, 0.015 mmol) was suspended in aq.70% acetic acid (5 mL) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford Example 8 (2R,3R,4S,5R,6R)-N-((lR,2S,3S)-3-(4-fluoro-2H-indazol-2-yl)- 2-hydroxycyclohexyl)-4- (4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5-hydroxy-6-(hydr oxymethyl)-3- methoxytetrahydro-2H-pyran-2-carboxamide (1.6 mg, 18%). LC/MS [M+H] + = 599.1, tR = 1.685 min (Method A); ¾ NMR (400MHz, MEOH-d 4 ) d = 8.60 (s, 1H), 8.37 (s, 1H), 7.67 (d, =7.6 Hz, 1H), 7.61 (d, J=10.0 Hz, 1H), 7.50 - 7.41 (m, 2H), 7.29 - 7.21 (m, 1H), 7.09 (t, =8.4 Hz, 1H), 6.72 (dd, J=10.5, 7.6 Hz, 1H), 4.92 (dd, J=10.8, 2.4 Hz, 1H), 4.46 (br. s., 1H), 4.25 (t, J= 9.9 Hz, 1H), 4.12 (d, J= 2.2 Hz, 1H), 4.07 - 3.91 (m, 3H), 3.86 - 3.67 (m, 3H), 3.17 (s, 3H), 2.31 - 2.13 (m, 2H), 2.10 - 1.90 (m, 2H), 1.72 - 1.55 (m, 2H). hGal3 IC50 = 0.57 mM.

Example 9a and 9b: Synthesis of (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((lS,2R,3R)-3-(3-(3-fluorophenyl)-lH-l,2,4-t riazol-l-yl)-2- hydroxycyclohexyl)-5-hydroxy-6-(hydroxymethyl)-3-methoxytetr ahydro-2H-pyran-2- carboxamide Isomers 1 and 2.

Step-1: Synthesis of (lR,2R,6S)-2-azido-6-(3-bromo-lH-l,2,4-triazol-l- yl)cyclohexanol(racemate): To a solution of (lS,2R,6R)-2-azido-7-oxabicyclo [4.1.0]heptane (200 mg, 1.437 mmol) and 3-bromo-lH-l, 2, 4-triazole (213 mg, 1.437 mmol) in DMSO (2 mL) was added DBU (0.650 mL, 4.31 mmol) and heated at 100 °C for 16 h. After confirmation of completion of reaction by LCMS, the reaction mass was diluted with EtOAc (20 mL) and Water (20 mL). The organic layer was separated and aqueous layer was re-extracted with EtAOc (2xl0mL). Combined organic extracts were washed with brine (20 mL) and dried over Na2SC>4. The solvent was removed under reduced pressure and the crude was purified by flash chromatography (60-120 mesh silica column, 50 - 60 % ethyl acetate in pet ether). The concentrate after purification yielded (lR,2R,6S)-2-azido-6-(3-bromo-lH-l,2,4-triazol-l-yl)cyclohex anol (143 mg, 0.5 mmol, 35%) as an off-white solid. LC-MS, [M+2] + = 289.0, {Method F, tR: 1.030 min, ELSD detector}.

Step-2: Synthesis of (lR,2R,6S)-2-azido-6-(3-(3-fluorophenyl)-lH-l,2,4-triazol- l-yl)cyclohexanol(racemate): To a solution of (lR,2R,6S)-2-azido-6-(3-bromo-lH-l,2,4- triazol-l-yl)cyclohexanol (110 mg, 0.383 mmol), (3-fluorophenyl)boronic acid (64.3 mg, 0.460 mmol) in 1,4-Dioxane (3 mL) and Water (0.450 mL) was added K2CO3 (116 mg, 0.843 mmol). The reaction mixture was degassed with N2 and added Pd(Ph3P)4 (22.14 mg, 0.019 mmol) under N2. The vial was sealed and heated at 95 °C for 16 h. The reaction mixture was cooled to rt and solvent was removed under reduced pressure to get the crude residue which was purified by flash chromatography (60-120 silicagel, 80 - 90% ethyl acetate in pet ether) to afford (lR,2R,6S)-2-azido-6-(3-(3-fluorophenyl)-lH- l,2,4-triazol-l-yl)cyclohexanol (58 mg, 47%) as an off-white solid. LC-MS, [M+H] + = 303.2, tR:2.256 min {Method C}.

Step-3: Synthesis of (lS,2R,6S)-2-amino-6-(3-(3-fluorophenyl)-lH-l,2,4-triazol- l-yl)cyclohexanol(racemate): Prepared in a similar fashion as described in Example la, Step-5 using ( 1 R,2R,6S)-2-azido-6-(3 -(3 -fluorophenyl)- 1 H- 1 ,2,4-triazol- 1 - yl)cyclohexanol to afford (lS,2R,6S)-2-amino-6-(3-(3-fluorophenyl)-lH-l,2,4-triazol-l- yl)cyclohexanol (0.048 g, 87 % yield) as an off-white solid. LC-MS, [M+H] + = 277.2 {tR:0.730 min, Method E}.

Step-4: Synthesis of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-N-((lR,2S, 3S)-3-(3-(3-fluorophenyl)-lH-l, 2, 4-triazol-l-yl)-2-hydroxy cyclohexyl)- 7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carbo xamide: Prepared in a similar fashion as described in Example la, Step-6 using (lS,2R,6S)-2-amino-6-(3-(3- fluorophenyl)-lH-l,2,4-triazol-l-yl)cyclohexanol (33.4 mg, 0.121 mmol) and (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-7-methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxylic acid (0.05 g, 0.110 mmol). The crude was purified by flash chromatography (60-120 silicagel, 6 - 10% MeOH in CHCb) to afford (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N- ((lR,2S,3S)-3-(3-(3-fluorophenyl)-lH-l,2,4-triazol-l-yl)-2-h ydroxycyclohexyl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxa mide (Isomer 1) (0.020 g, 24 % yield) and (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N- ((lR,2S,3S)-3-(3-(3-fluorophenyl)-lH-l,2,4-triazol-l-yl)-2-h ydroxycyclohexyl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxa mide (Isomer 2) (0.024 g, 22 % yield). LC-MS, [M+H] + = 714.2 {Method F, t R : 2.375 -Isomer- 1 /2.350 for Isomer-2}.

Step-5: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-N-((lR,2S,3S)-3-(3-(3-fluorophenyl)-lH-l ,2,4-triazol-l-yl)-2- hydroxycyclohexyl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][ l,3]dioxine-6- carboxamide (Isomer 1) (20 mg, 0.028 mmol) in dichloromethane (1 mL) was added trifluoroacetic acid (0.15 mL, 1.947 mmol) and stirred the reaction mixture at rt for 2h. Then the solvent was removed under reduced pressure and crude was purified through Prep HPLC {Method A}. Desired fractions were concentrated under reduced pressure to afford Example 9a:(2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l -yl)-N- ((lS,2R,3R)-3-(3-(3-fluorophenyl)-lH-l,2,4-triazol-l-yl)-2-h ydroxycyclohexyl)-5- hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro-2H-pyran-2-car boxamide (Isomer 1) (4.8 mg, 27.4 % yield) as an off-white solid. LC-MS, [M+H] + = 626.1, [ tR = 1.682 min, Method A] and & [ tR = 1.691 min, Method B] 'H NMR (400MHZ, MEOH-d4) d ppm 8.61 (s, 1H), 8.50 (s, 1H), 7.88 (d, J= 7.8 Hz, 1H), 7.76 (d, .7=10.3 Hz, 1H), 7.67 (d, J= 7.8 Hz, 1H), 7.62 (d, .7=10.0 Hz, 1H), 7.53 - 7.42 (m, 2H), 7.17 (td, .7=8.4, 2.3 Hz, 1H), 7.13 - 7.06 (m, 1H), 4.92 (dd, .7=10.8, 2.9 Hz, 1H), 4.36 - 4.22 (m, 2H), 4.12 (d, .7=2.9 Hz, 1H), 4.02 - 3.88 (m, 3H), 3.85 - 3.78 (m, 2H), 3.76 - 3.70 (m, 1H), 3.18 (s, 3H), 2.22 - 2.12 (m, 2H), 2.08 - 2.02 (m, 1H), 1.98 - 1.92 (m, 1H), 1.68 - 1.52 (m, 2H). hGal3 IC50 = 0.5 mM.

Example 9b: Prepared in a similar fashion as described in Example 9a using (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N-((lR,2S,3S)-3-(3-(3- fluorophenyl)- 1H- 1 ,2,4-triazol- 1 -y l)-2-hy droxy cy clohexy l)-7 -methoxy-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamide (Isomer 2) in step 5 (0.024 g, 0.034 mmol). The crude was purified through Prep HPLC {Method A}. Desired fractions were concentrated under reduced pressure to afford (2R,3R,4S,5R,6R)-4-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((lS,2R,3R)-3-(3-(3-f luorophenyl)-lH-l,2,4- triazol-l-yl)-2-hydroxycyclohexyl)-5-hydroxy-6-(hydroxymethy l)-3-methoxytetrahydro- 2H-pyran-2-carboxamide (Isomer 2) (1.01 mg, 4.5 % yield) as an off-white solid. LC- MS, [M+H] + = 626.1, [ tR = 1.661 min, Method A] and & [ tR = 1.668 min, Method B] ¾ NMR (400MHz, MEOH-d 4 ) d ppm 8.63 (s, 1H), 8.49 (s, 1H), 7.87 (d, J= 7.6 Hz, 1H), 7.75 (d, .7=10.3 Hz, 1H), 7.70 (d, .7=7.3 Hz, 1H), 7.63 (d, .7=11.0 Hz, 1H), 7.51 - 7.44 (m, 2H), 7.20 - 7.07 (m, 2H), 4.94 - 4.90 (m, 1H), 4.35 - 4.24 (m, 2H), 4.12 (d, J=3.4 Hz,

1H), 3.97 - 3.86 (m, 3H), 3.84 - 3.79 (m, 2H), 3.76 - 3.70 (m, 1H), 3.20 (s, 3H), 2.22 - 2.06 (m, 4H), 1.94 (br. s., 1H), 1.65 - 1.57 (m, 1H). hGal3 ICso = >100 mM.

Example 10a &10b

Step-1: Synthesis of l-((lS,2R,6R)-7-oxabicyclo[4.1.0]heptan-2-yl)-4-(3- fluorophenyl)-lH-l,2,3-triazole(racemate): To a solution of (lS,2R,6R)-2-azido-7- oxabicyclo[4.1.0]heptane (0.5 g, 3.59 mmol) in DMF (5 mL) and water (1.5 mL) was added sodium ascorbate (0.71 g, 3.59 mmol), copper(II) sulfate pentahydrate (0.81 g, 3.23 mmol) and 3-fluorophenylacetylene (1.7 mL, 14.37 mmol) at rt. The reaction mixture was heated at 85 °C for 30 min. The reaction mixture was cooled to rt, diluted with 1 : 1 DCM (50 mL) and water (50 mL) and stirred at rt for 15 min. The reaction mixture was filtered through Celite pad and washed with DCM (20 mL). From the filtrate, organic layer was separated and aqueous layer re-extracted with DCM (2x20 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated to give crude residue. The crude residue was purified by flash chromatography (40-50% EtOAc in n-hexane) to afford l-((lS,2R,6R)-7- oxabicyclo[4.1.0]heptan-2-yl)-4-(3-fluorophenyl)-lH-l, 2, 3-triazole (0.53 g, 57%) as pale yellow solid. LC/MS [M+H] + = 260.2, tR = 2.266 min (Method C).

Step-2: Synthesis of (lR,2R,6S)-2-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-6- (methylamino)cyclohexanol(racemate): In sealed tube l-((lS,2R,6R)-7- oxabicyclo[4.1.0]heptan-2-yl)-4-(3-fluorophenyl)-lH-l, 2, 3-triazole (110 mg, 0.424 mmol) and methanamine (33% solution in ethanol) (5 mL, 0.424 mmol) was heated to 65 °C for 16 h. The reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was triturated with n-pentane and dried to afford (lR,2R,6S)-2-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-6- (methylamino)cyclohexanol (109 mg, 88%). LC/MS [M+H] + = 291.0, tR = 1.372 min (Method F).

Step-3: Synthesis of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-N-((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l, 2, 3-triazol-l-yl)-2-hydroxy cyclohexyl)- 7-methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxin e-6-carboxamide: To a solution of ((2S,4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazo l-l-yl)-7- methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxy lic acid (50 mg, 0.11 mmol) in DMF (2 mL) was added DIPEA (0.06 mL, 0.33 mmol) and HATU (63 mg, 0.16 mmol) at rt and stirred for 15 min. Then (lR,2R,6S)-2-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-6-(methylamino)cyclohexanol (35.1 mg, 0.121 mmol) was added and stirred the mixture at rt for 1 h. The reaction mixture was quenched with ice cold water (10 mL) and stirred for 15 min. The resultant solid was filtered, washed with excess water and residue was dried to afford diastereomeric mixture of crude residue. The crude residue was further purified by prep-SFC to obtain Isomer 1 and Isomer 2.

Preparative SFC Conditions

Column/dimensions: Chiralcel OD-H(250 X 21)mm,5u % C02: 60% % Co solvent: 40% of 0.2 % DEA in MEOH Total Flow: 80.0g/min Back Pressure: 100 bar Temperature: 25°C UV: 242 nm

Isomer 1: 35 mg, 37% yield; LC/MS [M+H] + = 728.2, tR = 3.178 min (Method C).

Isomer 2: 25 mg, 31% yield; LC/MS [M+H] + = 728.0, tR = 2.635 min (Method F).

Example 10a: (2S,4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol -l- yl)-N-((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-2-hydroxycyclohexyl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamide (Isomer

1) (35 mg, 0.048 mmol) was suspended in aq.70% acetic acid (5 mL) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford ((2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-N-((lS,2R,3R)-3- (4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-hydroxycyclohex yl)-5-hydroxy-6-

(hy droxymethyl)-3-methoxy-N-methyltetrahy dro-2H-pyran-2-carboxamide Isomer 1 (9.3 mg, 30%) as an off-white solid. LC/MS [M+H] + = 640.2, tR = 1.788 min (Method A); 'H NMR (400MHz, MEOH-d4) d ppm 8.71, 8.70 (two singlets, 1H), 8.53, 8.42 (two singlets, 1H), 7.76 - 7.57 (m, 4H), 7.53 - 7.42 (m, 2H), 7.15 - 7.05 (m, 2H), 5.03 - 4.96 (m, 1H), 4.76 - 4.66 (m, 1H), 4.58 (m, 1H), 4.55 - 4.35 (m, 3H), 4.34 - 4.04 (m, 4H), 3.21, 3.00 (two singlets, 3H), 3.13, 3.11 (two singlets, 3H), 2.27 - 2.09 (m, 2H), 2.04 - 1.90 (m, 2H), 1.85 (m, 2H) {Rotameric mixture}. hGal3 ICso = 5.5 mM.

Example 10b: ((2S,4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazo l-l- yl)-N-((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-2-hydroxycyclohexyl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamide (Isomer

2) (25 mg, 0.034 mmol) was suspended in 70% Aq. acetic acid (5 mL, 87 mmol) and heated at 70 °C for 16 h. The reaction mixture was cooled to rt and concentrated under reduced pressure to give crude residue. The crude residue was purified by prep-HPLC [Method A] to afford ((2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-N- ((lS,2R,3R)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-2-h ydroxycyclohexyl)-5- hydroxy-6-(hydroxymethyl)-3-methoxy-N-methyltetrahydro-2H-py ran-2-carboxamide Isomer 2 (2.2 mg, 10%). LC/MS [M+H] + = 640.2, t R = 1.78 min (Method A); ¾ NMR (400MHz, MEOH-d4) d ppm 8.72, 8.71 (two singlets, 1H), 8.47, 8.45 (two singlets, 1H), 7.75 - 7.57 (m, 4H), 7.53 - 7.43 (m, 2H), 7.10 (t, J=8.4 Hz, 2H), 5.03 - 4.95 (m, 1H), 4.65 - 4.49 (m, 3H), 4.47 - 4.40 (m, 1H), 4.21 - 4.00 (m, 2H), 3.94 - 3.84 (m, 1H), 3.83 - 3.64 (m, 2H), 3.21, 2.98 (two singlets, 3H), 3.14, 3.11(two singlets, 3H), 2.28 - 2.11 (m, 2H), 2.10 - 1.95 (m, 2H), 1.94 - 1.64 (m, 2H). [Rotameric mixture]; hGab IC50 = 0.091 mM.

The Examples in the table 2 were prepared in an analogous fashion to Example 10a and 10b, substituting methyl amine with the appropriate alkyl amines in the synthetic sequence.

Table 2

Example 16a and 16b: Synthesis of (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxypiperidin-3-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxy -N-methyltetrahydro- 2H-pyran-2-carboxamide (Isomer 1 and 2).

Step-1: Synthesis oftert-butyl (lS,5R,6R)-5-hydroxy-7-oxa-3- azabicyclo[4.1.0]heptane-3-carboxylate(racemate): To a solution of N-Boc-3-hydroxy- 1,2,3,6-tetrahydropyridine (500 mg, 2.51 mmol) in DCM (25 mL) was added sodium bicarbonate (211 mg, 2.51 mmol) followed by mCPBA (928 mg, 3.76 mmol) in DCM (3 mL) at 0 °C under N2. The reaction mixture was allowed to warm to rt and was stirred for 24 h. The reaction mixture was diluted with DCM (100 mL), filtered through a Celite pad and the filtrate was washed with saturated Na2SC>3 (2x50 mL), saturated NaHCCb (2x50 mL) and saturated NaCl (25 mL). The organic layer was dried over Na2SC>4, filtered, and concentrated and the crude residue was purified by silica gel chromatography (40% 50% ethyl acetate in hexanes) to yield the title compound (430 mg, 80% yield) as a white solid. 1H NMR (400MHz, CHLOROFORM-d) d 4.01 (br.s, 1 H), 3.92 - 3.44 (m, 5 H), 3.10 (dd, J = 12.8, 7.2 Hz, 1 H), 1.29 (s, 9 H) (Rotameric mixture). Step-2: Synthesis oftert-butyl (lS,5S,6R)-5-azido-7-oxa-3-azabicyclo[4.1.0] heptane-3 -carboxylate(racemate): To a stirred solution of (lS,5R,6R)-tert-butyl 5- hydroxy-7-oxa-3-azabicyclo[4.1.0]heptane-3-carboxylate (325 mg, 1.510 mmol) in THF (50 mL) was added triphenylphosphine (792 mg, 3.02 mmol). The reaction mixture was cooled 0 °C. DEAD (0.478 mL, 3.02 mmol) and DPPA (0.651 mL, 3.02 mmol) were added sequentially under N2. The reaction mixture was allowed to warm to rt and was stirred for 16 h. The solvent was removed under reduced pressure and the crude residue was purified by silica gel chromatography (10% 15% ethyl acetate in hexanes) to yield the title compound (230 mg, 64% yield) as pale a yellow oil. 1H NMR (400MHz, DMSO-d6) d 4.09 - 3.82 (m, 2 H), 3.62 - 3.38 (m, 4 H), 3.17 (dd, J = 13.1, 3.3 Hz, 1 H), 1.40 (s, 9 H) (Rotameric mixture).

Step-3: Synthesis oftert-butyl (lS,5S,6R)-5-(4-(3-fhiorophenyl)-lH-l,2,3-triazol- l-yl)-7-oxa-3-azabicyclo[4.1.0]heptane-3-carboxylate(racemat e): To a solution of (lR,5R,6S)-tert-butyl 5-azido-7-oxa-3-azabicyclo[4.1.0]heptane-3-carboxylate (1.6 g,

6.66 mmol) in DMF (30 mL) and water (7.50 mL), was added sodium ascorbate (1.319 g, 6.66 mmol), copper(II) sulfate pentahydrate (1.496 g, 5.99 mmol) and 3- fluorophenylacetylene (3.08 mL, 26.6 mmol) sequentially at rt. The reaction mixture was degassed with N2 for 5 min and heated to 85 °C for 30 min. The mixture was cooled to rt, diluted with ice cold water (100 mL) and stirred for 15 min to get a solid. The solid was filtered, suspended in DCM (100 mL), filtered through celite pad and washed with excess DCM. The filtrate was dried over sodium sulfate and concentrated. The residue was purified by silica gel (40% 80% ethyl acetate in hexanes) to afford the title compound (1.1 g, 46% yield) as a white solid. 'H NMR (400MHz, CHLOROFORM-d) d 7.83 (br.s,

1 H), 7.59 - 7.55 (m, 2 H), 7.42 - 7.38 (m, 1 H), 7.10 - 7.07 (m, 1 H), 5.18 - 5.06 (m, 1 H), 4.34 - 4.29 (m, 1 H), 3.93 - 3.45 (m, 5 H), 1.47 - 1.11 (m, 9 H) (rotameric mixture); LC/MS, [M+H] + = 361.0, /R = 2.01 min (Method E).

Step-4: Synthesis of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)-4-hydroxy-5-(methylamino)piperidine-l-carboxylate(race mate): (lS,5S,6R)-tert- butyl5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-oxa-3-az abicyclo[4.1.0]heptane-3- carboxylate (100 mg, 0.277 mmol) in methanamine (33% solution in Ethanol) (5 mL, 0.277 mmol) was heated in a sealed tube at 65 °C for 16 h. Reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was triturated with pentane and dried under reduced pressure to give (3S,4S,5R)- tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxy-5- (methylamino)piperidine-l-carboxylate (105 mg, 94%). LC-MS, [M+H] + = 392.2, {Method C: tR = 1.855 min}.

Step-5: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d] [l,3]dioxine-6-carboxylic acid (50 mg, 0.110 mmol) in DMF (2 mL), DIPEA (0.058 mL, 0.329 mmol) and HATU (62.6 mg, 0.165 mmol) were added sequentially at rt and stirred for 15 min. Then (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxy-5- (methylamino)piperidine-l-carboxylate (47.3 mg, 0.121 mmol) was added and stirred the reaction mixture at rt for 1 h. The reaction mixture was quenched with ice water and stirred for 15 minutes. The resultant solid was filtered and dried to afford crude residue. The crude residue was purified by silica gel (5-10% MeOH in chloroform) to afford tert- butyl (3S,4S,5R)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5-(( 4aR,6R,7R,8R,8aR)-8- (4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-methoxy-N-methy l-2- phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxamido)-4-hy droxypiperidine-l- carboxylate as the diastereomeric mixture which was further purified by chiral SFC to afford the two isomers:

Prep SFC Method info:

Column/dimensions: Chiralcel OJ-H(250 X 21)mm,5u % C02: 70%

% Co solvent: 30 % of IPA +ACN Total Flow: 70.0 g/min Back Pressure: 100 bar Temperature: 25°C UV: 244nm

Analytical chiral SFC conditions:

Analytical Column: ChiralCel OJH (250 X 4.6)mm,5u

BPR pressure: 100 bars Temperature: 22.3 °C

Flow rate: 2.8 g/min

Mobile Phase: C0 2 / IPA+ACN (70/30) Detector Wavelength: UV 200-400 nm Isomer 1: (30 mg, 31% yield); chiral SFC tR = 1.8 min; LC-MS, [M+H] + = 829.0, {Method F : tR = 2.855 min};

Isomer 2: (28 mg, 31% yield); chiral SFC /R = 5.15 min; LC-MS, [M+H] + = 829.0, {Method F : tR = 3.0 min};

Step-6: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-5-((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH -l,2,3-triazol-l-yl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamido)-4- hydroxypiperidine-l-carboxylate Isomer 1(30 mg, 0.036 mmol) in dichloromethane (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added and stirred the mixture at rt for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative HPLC Method A to afford Example 16a: (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl )-N-((3R,4S,5S)-5-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxypiperidin-3-yl )-5-hydroxy-6- (hydroxymethyl)-3-methoxy-N-methyltetrahydro-2H-pyran-2-carb oxamide Isomer 1 (10.6 mg, 47% yield) as an off-white solid. LC-MS, [M+H] + = 641.3, {Method A: tR = 1.520}. 1H NMR (400MHz, MEOH-d4) d ppm 8.72, 8.68 (two singlets, 1H), 8.45 (m, 1H), 7.74 - 7.55 (m, 4H), 7.51 - 7.41 (m, 2H), 7.14 - 7.04 (m, 2H), 4.97 (dd, J=10.6, 2.8 Hz, 1H), 4.67 - 4.49 (m, 2H), 4.48 - 4.24 (m, 3H), 4.10 (dd, J=7.9, 2.6 Hz, 1H), 3.95 (dd, J=8.4, 3.8 Hz, 1H), 3.89 - 3.76 (m, 2H), 3.74 - 3.68 (m, 1H), 3.43 - 3.35 (m, 1H), 3.23, 3.00 (two singlets, 3H), 3.22-3.16 (m, 1H), 3.14 - 3.10 (m, 3H), 3.09 - 3.01 (m, 1H) [ *rotameric mixture]; hGal3 IC50 = 3.5 mM.

Example 16b: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-5-((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophen yl)-lH-l,2,3-triazol-l-yl)- 7-methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxin e-6-carboxamido)-4- hydroxypiperidine-l-carboxylate Isomer 2 (28 mg, 0.034 mmol) in dichloromethane (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added at rt and stirred for 2 h.

Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative LC/MS Method A to afford (2R,3R,4S,5R,6R)-4-(4- (3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-( 3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-4-hydroxypiperidin-3-yl)-5-hydroxy-6-(hydroxym ethyl)-3-methoxy-N- methyltetrahydro-2H-pyran-2-carboxamide Isomer 2 (12.4 mg, 0.019 mmol, 57%) as an off-white solid. LC-MS, [M+H] + = 641.1, {Method A: ta = 1.517}. 1HNMR (400MHz, MEOH-d4) d ppm 8.71, 8.68 (two singlets, , 1H), 8.53, 8.51 (two singlets, 1H), 7.73 - 7.57 (m, 4H), 7.51 - 7.42 (m, 2H), 7.14 - 7.05 (m, 2H), 4.97 (dt, J=10.3, 3.4 Hz, 1H), 4.57 - 4.37 (m, 2H), 4.30 - 4.21 (m, 1H), 4.08 (dd, J=7.5, 2.3 Hz, 1H), 4.01 - 3.96 (m, 1H),

3.90 - 3.67 (m, 5H), 3.50 - 3.41 (m, 2H), 3.27, 3.00 (two singlets, 3H), 3.13 - 3.10 (m, 3H), 3.04 (m, 1H) {Rotameric Mixture}. hGal3 ICso = 0.044 mM.

Example 17a and 17b: Synthesis of (2R,4R,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxypiperidin-3-yl)-5-hydroxy-6-(hydroxymethyl)-N-methylt etrahydro-2H-pyran-2- carboxamide (Isomer 1 and 2)

Example 17a & 17b Step-1 : Synthesis of tert-butyl (3S,4S,5R)-3-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-5-((4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l, 2,3-triazol-l-yl)-N-methyl- 2-phenylhexahydropyrano[3,2-d] [ 1 ,3] dioxine-6-carboxamido)-4-hydroxypiperidine- 1 - carboxylate: To a stirred solution of (4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-c arboxylic acid (50 mg, 0.118 mmol) in DMF (2 mL), DIPEA (0.062 mL, 0.353 mmol) and HATU (67.0 mg,

0.176 mmol) were added sequentially at rt and stirred for 15 min. Then (3S,4S,5R)-tert- butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxy-5-(me thylamino)piperidine- 1-carboxylate (50.6 mg, 0.129 mmol) was added and stirred the reaction mixture at rt for 1 h. The reaction mixture was quenched with ice water and stirred for 15 minutes. The resultant solid was filtered and dried to afford crude residue. The crude residue was purified by silica gel (5-10% MeOH in chloroform) to afford (3S,4S,5R)-tert-butyl 3-(4- (3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5-((4aR,6R,8R,8aR)-8 -(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-N-methyl-2-phenylhexahydropyrano[3,2-d][ l,3]dioxine-6- carboxamido)-4-hydroxypiperidine-l-carboxylate as the diastereomeric mixture which was further purified by chiral SFC to afford the two isomers:

Prep SFC Method info:

Column/dimensions: Chiralcel OJ-H(250 X 21)mm,5u % C02: 80%

% Co solvent: 20 % of IPA +ACN Total Flow: 70.0g/min Back Pressure: 100 bar Temperature: 25°C UV: 244nm

Analytical chiral SFC conditions:

Analytical Column: ChiralCel OJH (250 X 4.6)mm,5u

BPR pressure: 100 bars Temperature: 22.3 °C

Flow rate: 3 g/min

Mobile Phase: CO2 / IPA+ACN (75/25) Detector Wavelength: UV 200-400 nm

Isomer 1: (28 mg, 0.035 mmol, 30%); chiral SFC /R = 3.74 min; LC-MS, [M+H] + =

799.1, {Method F: tR = 3.888 min};

Isomer 2: (29 mg, 0.034 mmol, 29%); chiral SFC R = 5.91 min; LC-MS, [M+H] + =

799.2, {Method F: tR = 3.505 min};

Step-2: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-5-((4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l, 2,3-triazol-l-yl)-N-methyl- 2-phenylhexahydropyrano[3,2-d] [ 1 ,3] dioxine-6-carboxamido)-4-hydroxypiperidine- 1 - carboxylate (Isomer 1) (28 mg, 0.035 mmol) in DCM (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added at rt and stirred for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative HPLC Method A to afford Example 17a: (2R,4R,5R,6R)-4-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-f luorophenyl)-lH-l,2,3- triazol-1 -yl)-4-hy droxypiperi din-3-yl)-5-hydroxy-6-(hydroxymethyl)-N- methyltetrahydro-2H-pyran-2-carboxamide Isomer 1 (10.9 mg, 50 %) as an off-white solid. LC-MS, [M+H] + = 611.3, {Method A: ta = 1.257}. 1HNMR (400MHz, MEOH- d4) d ppm 8.53 - 8.46 (m, 2H), 7.71 - 7.59 (m, 4H), 7.52 - 7.42 (m, 2H), 7.15 - 7.06 (m, 2H), 5.13 (d, J=12.5 Hz, 1H), 4.81 - 4.74 (m, 1H), 4.68 (dd, J=9.2, 6.5 Hz, 1H), 4.56 - 4.35 (m, 2H), 4.14 (s, 1H), 4.01 - 3.56 (m, 5H), 3.48 - 3.36 (m, 1H), 3.25, 3.00 (two singlets, 3H), 3.06 (q, J=7.3 Hz, 1H), 2.91 - 2.79 (m, 1H), 2.25 - 2.14 (m, 1H) {Rotameric Mixture}; hGal3 IC50 = 5.2 mM.

Example 17b: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-5-((4aR,6R,8R,8aR)-8-(4-(3-fluorophenyl) -lH-l,2,3-triazol-l-yl)-N- methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6-carboxam ido)-4- hy droxypiperi dine- 1 -carboxylate Isomer 2 (29 mg, 0.036 mmol) in DCM (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added at rt and stirred for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative LC/MS Method A to afford ((2R,4R,5R,6R)-4-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-f luorophenyl)-lH-l,2,3- triazol-1 -yl)-4-hy droxypiperi din-3-yl)-5-hy droxy-6-(hy droxymethyl)-N- methyltetrahydro-2H-pyran-2-carboxamide Isomer 2 (6.7 mg, 0.011 mmol, 30%) as an off-white solid. LC-MS, [M+H] + = 611.3, {Method A: t R = 1.255}. 1HNMR (400MHz, MEOH-d4) d ppm 8.54 - 8.46 (m, 2H), 7.70 - 7.57 (m, 4H), 7.51 - 7.41 (m, 2H), 7.14 - 7.05 (m, 2H), 5.15 - 5.06 (m, 1H), 4.95 - 4.86 (m, 1H), 4.74 - 4.61 (m, 1H), 4.48 - 4.41 (m, 1H), 4.32 (td, J=10.9, 4.3 Hz, 1H), 4.10 (s, 1H), 4.01 - 3.70 (m, 5H), 3.57 - 3.43 (m, 1H), 3.24, 2.99 (two singlets, 3H), 3.05 (q, J=7.4 Hz, 1H), 2.85 - 2.69 (m, 1H), 2.32 - 2.15 (m, 1H) {Rotameric Mixture}. hGal3 IC50 = 0.13 mM. Example 18a and 18b: Synthesis of(2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxypiperidin-3-yl)-3,5-dihydroxy-6-(hydroxymethyl)-N-met hyltetrahydro-2H-pyran- 2-carboxamide (Isomer 1 and 2)

Example 18a &18b

Step-1 : To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-7-hydroxy-2-phenylhexahydropyrano[3,2-d] [l,3]dioxine-6-carboxylic acid (50 mg, 0.113 mmol) in DMF (2 mL), DIPEA (0.059 mL, 0.340 mmol) and HATU (64.6 mg, 0.170 mmol) were added sequentially at rt and stirred for 15 min. Then

(3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxy-5- (methylamino)piperidine-l-carboxylate (48.8 mg, 0.125 mmol) was added and stirred the reaction mixture at rt for 1 h. The reaction mixture was quenched with ice water and stirred for 15 minutes. The resultant solid was filtered and dried to afford crude residue. The crude residue was purified by silica gel (5-10% MeOH in Chloroform) to afford (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5- ((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l -yl)-7-hydroxy-N-methyl- 2-phenylhexahydropyrano[3,2-d] [ 1 ,3] dioxine-6-carboxamido)-4-hydroxypiperidine- 1 - carboxylate as the diastereomeric mixture which was further purified by prep-HPLC Method C to afford the two isomers:

Analytical HPLC conditions: Analytical Column: Symmetry C9 (250 X 4.6)mm,5u Mobile Phase A: 0.1% TFA in Water Mobile Phase B: ACN

Flow rate: 1 mL/min Gradient: 20-100% B over 20minutes, then a 10-minute hold at 100% B

Detector Wavelength: UV 200-400 nm

Isomer 1: (25 mg, 0.030 mmol, 27%); HPLC to = 16.083 min; LC-MS, [M+H] + = 815.0, {Method F: to = 2.683 min}.

Isomer 2: (21 mg, 0.025 mmol, 22%); HPLC to = 16.700 min; LC-MS, [M+H] + = 815.2, {Method F: tR = 3.98 min}.

Step-2: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-5-((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH -l,2,3-triazol-l-yl)-7- hydroxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamido)-4- hydroxypiperidine-l-carboxylate Isomer 1(25 mg, 0.031 mmol) in DCM (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added at rt and stirred for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative HPLC Method D to afford Example 18a: (2R,3R,4S,5R,6R)-4-(4- (3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-( 3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-4-hydroxypiperidin-3-yl)-3,5-dihydroxy-6-(hydr oxymethyl)-N- methyltetrahydro-2H-pyran-2-carboxamide Isomer 1 (8 mg, 41% yield) as an off-white solid. LC-MS, [M+H] + = 627.2, {Method C: t R = 1.500}. 1H NMR (400MHz, MEOH- d4) d ppm 8.44, 8.43 (two singlets, 1H), 8.36, 8.34 (two singlets, 1H), 7.70 - 7.56 (m,

4H), 7.50 - 7.41 (m, 2H), 7.13 - 7.04 (m, 2H), 4.98 - 4.90 (m, 1H), 4.80 - 4.75 (m, 1H), 4.65 - 4.50 (m, 2H), 4.44 (d, J=9.0 Hz, 1H), 4.37 - 4.28 (m, 1H), 4.23 (td, J=10.7, 4.8 Hz, 1H), 4.14 (d, J=2.5 Hz, 1H), 4.01 (dd, J=8.3, 3.8 Hz, 1H), 3.93 - 3.77 (m, 2H), 3.75 - 3.69

(m, 2H), 3.43 - 3.34 (m, 1H), 3.24 (s, 1H), 3.22 - 3.12 (m, 2H) [ *rotameric mixture]; hGal3 IC50 = 1.8 mM.

Example 18b: To a solution of (3S,4S,5R)-tert-butyl 3-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-5-((4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophen yl)-lH-l,2,3-triazol-l-yl)- 7-hydroxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxin e-6-carboxamido)-4- hydroxypiperidine-l-carboxylate Isomer 2(16 mg, 0.020 mmol) in DCM (1 mL), trifluoroacetic acid (0.15 mL, 1.947 mmol) was added at rt and stirred for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via preparative LC/MS Method A to afford (2R,3R,4S,5R,6R)-4-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-f luorophenyl)-lH-l,2,3- triazol-l-yl)-4-hydroxypiperidin-3-yl)-3,5-dihydroxy-6-(hydr oxymethyl)-N- methyltetrahydro-2H-pyran-2-carboxamide Isomer 2 (2.8 mg, 22%) as an off-white solid. LC-MS, [M+H] + = 627.1, {Method A: t R = 1.517}. 1HNMR (400MHz, MEOH-d4) d ppm 8.55 (d, J=2.9 Hz, 1H), 8.53 (d, J=2.4 Hz, 1H), 7.72 - 7.66 (m, 2H), 7.64 (d, J=7.6 Hz, 2H), 7.53 - 7.44 (m, 2H), 7.16 - 7.07 (m, 2H), 4.97 (dd, J=10.6, 2.8 Hz, 1H), 4.80 - 4.73 (m, 1H), 4.48 - 4.39 (m, 2H), 4.16 (d, J=3.4 Hz, 1H), 4.05 (d, J=5.4 Hz, 1H), 3.94 - 3.70 (m, 6H), 3.55 - 3.46 (m, 1H), 3.29, 3.02 (two singlets, 3H), 3.06 (d, J=7.8 Hz, 1H),

{Rotameric mixture}. hGal3 IC50 = 0.05 mM.

Example 19. Synthesis of(2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N-((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-4-hydroxy-l- methylpiperidin-3-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxy- N-methyltetrahydro-2H- pyran-2-carboxamide Step-1: Synthesis of (lR,5R,6S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7- oxa-3-azabicyclo[4.1.0]heptane(homochiral): To a stirred solution of tert-butyl (lR,5R,6S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-ox a-3- azabicyclo[4.1.0]heptane-3-carboxylate (100 mg, 0.277 mmol) in dichloromethane (5 mL), 100 mg of 4A molecular sieves and BF3.0Et2 (0.105 mL, 0.832 mmol) were added sequentially at rt and stirred for 30 min. The reaction mixture was filtered through a Celite pad, washed with excess DCM (20 mL) and the filtrate was concentrated under reduced pressure to give (lR,5R,6S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-ox a- 3-azabicyclo[4.1.0]heptane (70 mg, 83% yield) as a pale yellow oil; LC-MS, [M+H] + = 261.4, {Method E: tR = 0.90 min}.

Step-2: Synthesis of (lR,5R,6S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-3- methyl-7-oxa-3-azabicyclo[4.1.0]heptane: To a stirred solution of (lR,5R,6S)-5-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-7-oxa-3-azabicyclo[4.1. 0]heptane (70 mg, 0.269 mmol) in MeOH (5 mL), paraformaldehyde (40.4 mg, 1.345 mmol) and 0.1 mL of AcOH was added and stirred for 10 min at rt. Then sodium cyanoborohydride (16.90 mg, 0.269 mmol) was added and stirred at rt for 16 h. MeOH was removed under reduced pressure and the crude residue was extracted with 10% MeOH in DCM (2X30 mL) washed with water, brine, dried over sodium sulphate and concentrated. The residue was purified via silica gel chromatography (20-50 % EtOAc in n-hexane) to yield (lR,5R,6S)-5-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-3-methyl-7-oxa-3-azabic yclo[4.1.0]heptane (70 mg, 92% yield) as an off-white solid. LC-MS, [M+H] + = 275.2, {Method C: tR = 1.828 min}.

Step-3: Synthesis of (3R,4R,5S)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-l- methyl-5-(methylamino)piperidin-4-ol: (lR,5R,6S)-5-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-3-methyl-7-oxa-3-azabicyclo[4.1.0]heptane (70 mg, 0.255 mmol) in methanamine (33% solution in Ethanol) (5 mL, 0.255 mmol) was heated in a sealed tube at 65 °C for 16 h. Reaction mixture was cooled to rt and solvent was removed under reduced pressure to give the title compound (65 mg, 81 % yield) as brown solid. LC-MS, [M+H] + = 306.2, {Method C: t R = 0.833 min}.

Step-4: Synthesis of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4-hydroxy-l- methylpiperidin-3-yl)-7-methoxy-N-methyl-2-phenylhexahydropy rano[3,2- d][l,3]dioxine-6-carboxamide: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3- fluorophenyl)- lH-1, 2, 3-triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3, 2- d][l,3]dioxine-6-carboxylic acid (30 mg, 0.066 mmol) in DMF (2 mL), DIPEA (0.035 mL, 0.198 mmol) and HATU (37.6 mg, 0.099 mmol) were added sequentially at rt and stirred for 15 min. Then (3R,4R,5S)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-l- methyl-5-(methylamino)piperidin-4-ol (24.14 mg, 0.079 mmol) was added and stirred the reaction mixture at rt for 1 h. The reaction mixture was quenched with ice water and stirred for 15 minutes. The resultant solid was filtered and dried to afford the title compound (35 mg, 55% yield) as brown solid. LC-MS, [M+H] + = 743.2, {Method C: te = 2.88 min}.

Step-5: To a solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4-hydroxy-l- methylpiperidin-3-yl)-7-methoxy-N-methyl-2-phenylhexahydropy rano[3,2- d][l,3]dioxine-6-carboxamide (30 mg, 0.040 mmol) in dichloromethane (3 mL), trifluoroacetic acid (0.5 mL, 6.49 mmol) was added at rt and stirred for 2 h. Then, the solvent was removed under reduced pressure to get crude residue. The crude material was purified via prep-HPLC Method D to afford Example 19: (2R,3R,4S,5R,6R)-4-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)-N-((3S,4R,5R)-5-(4-(3-f luorophenyl)-lH-l,2,3- triazol-l-yl)-4-hydroxy-l-methylpiperidin-3-yl)-5-hydroxy-6- (hydroxymethyl)-3- methoxy-N-methyltetrahydro-2H-pyran-2-carboxamide (18.7 mg, 0.029 mmol, 71%) as an off-white solid. LC-MS, [M+H] + = 655.1, {Method A: tR = 1.685 min}. 'HNMR (400MHz, MEOH-d4) d ppm 8.62, 8.59 (two singlets, 1H), 8.41, 8.38 (two singlets, 1H), 7.63 - 7.48 (m, 4H), 7.42 - 7.33 (m, 2H), 7.05 - 6.96 (m, 2H), 4.88 (dd, .7=10.1, 3.1 Hz, 1H), 4.54 - 4.47 (m, 1H), 4.41 - 4.23 (m, 3H), 4.01 (d, J= 2.9 Hz, 1H), 3.89 (d, J= 3.2 Hz, 1H), 3.81 - 3.74 (m, 2H), 3.71 - 3.59 (m, 4H), 3.17 - 3.15 (m, 3H), 3.05 - 2.86 (m, 6H), 2.75 (s, 1H) {Rotameric mixture}. hGal3 IC50 = 0.02 mM.

Example 20a and 20b: Synthesis of (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxytetrahydro-2H-pyran-3-yl)-5-hydroxy-6-(hydroxymethyl) -3-methoxytetrahydro- 2H-pyran-2-carboxamide (Isomer 1 and 2)

Example 20a & 20b

Step-1: Synthesis of 3-azido-3,6-dihydro-2H-pyran: To a stirred solution of 3,6- dihydro-2H-pyran-3-ol (1 g, 9.99 mmol) in DCM (50 mL), triethylamine (2.78 mL, 19.98 mmol) and mesyl-Cl (0.934 mL, 11.99 mmol) were added sequentially at 0 °C under N2 and stirred for 30 min. Then the reaction mixture was extracted with DCM (100 mL), washed with water, brine, dried over sodium sulphate and concentrated. The crude mesylate was dissolved in DMSO (10 mL), sodium azide (2.74 g, 42.1 mmol) was added and stirred at rt for 16 h. The reaction mixture was extracted with EtOAc (3x50 mL), washed with water, brine and dried over sodium sulphate. Solvent was removed under reduced pressure to get crude residue which was purified by flash chromatography (15 - 20% EtOAc in n-hexane) to afford 3-azido-3,6-dihydro-2H-pyran (0.75 g, 5.95 mmol, 71%). ¾ NMR (400 MHz, CHLOROF ORM-ύG) d ppm 6.07 - 6.22 (m, 1 H), 5.84 - 5.97 (m, 1 H), 4.18 - 4.25 (m, 1 H), 4.07 - 4.15 (m, 1 H), 3.96 (ddd, .7=12.0, 3.0, 1.0 Hz, 1 H), 3.81 - 3.86 (m, 1 H), 3.55 (br.s, 1 H).

Step-2: Synthesis of l-(3,6-dihydro-2H-pyran-3-yl)-4-(3-fluorophenyl)-lH-l, 2,3- triazole: To a solution of 3-azido-3,6-dihydro-2H-pyran (0.74 g, 5.91 mmol) in DMF (10 mL) and water (3 mL) was added sodium ascorbate (1.172 g, 5.91 mmol), copper(II) sulfate pentahydrate (1.329 g, 5.32 mmol) and 3-fluorophenylacetylene (2.73 mL, 23.66 mmol). The reaction mixture was degassed N2 for 10 min and heated at 85 °C for 15 min. Then the reaction mixture was cooled to rt, diluted with DCM (50 mL)/water (50 mL) and stirred for 30 minutes. The organic layer was separated and aqueous layer was re extracted with DCM (2x3 OmL). Combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated. The crude residue was purified by flash chromatography (35 - 50% EtOAc in n-hexane) to afford the title compound (1.36 g, 5.45 mmol, 92%) as pale yellow solid. LC-MS, [M+H] + = 246.2, {Method C: ta = 2.006 min}.

Step-3: Synthesis of (3S,4R,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)tetrahydro-2H-pyran-3,4-diol(racemate):To a stirred solution of l-(3,6-dihydro-2H- pyran-3-yl)-4-(3-fluorophenyl)-lH-l, 2, 3-triazole (0.4 g, 1.631 mmol) in acetone (4 mL) and water (1 mL), was added 4-methylmorpholine-N-oxide (0.287 g, 2.446 mmol) and osmium tetroxide (2.048 mL, 0.163 mmol, 2.5% w/v in t-butanol) at rt and stirred for 14 h. The reaction mixture was quenched with sat.Na2S03 solution and acetone was removed under reduced pressure to give crude residue. The crude residue was extracted with EtOAc (2X100 mL), washed with water, brine, dried over sodium sulphate and concentrated. The crude residue was purified by flash chromatography (80 - 100%

EtOAc in n-hexane) to afford the title compound (0.25 g, 0.895 mmol, 55%). LC-MS, [M+H] + = 280.2, {Method C: t R = 1.025 min}.

Step-4: Synthesis of (3aS,7S,7aR)-7-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)tetrahydro-3aH-[l,3,2]dioxathiolo[4,5-c]pyran 2,2-dioxide(racemate): To a stirred solution of (3S,4R,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)tetra hydro-2H-pyran- 3,4-diol (0.25 g, 0.895 mmol) in DCM (10 mL) was added TEA (0.250 mL, 1.790 mmol) at rt and stirred for 5 min. Then the reaction mixture was cooled 0°C and SOCh (0.131 mL, 1.790 mmol) was added under N2. Reaction mixture was allowed to reach rt and stirred at rt for 1 h. Solvent was removed under reduced pressure, crude was extracted with EtOAc (2 X 50 mL), washed with water, brine and dried over sodium sulphate. The solvent was removed under reduced pressure and crude residue was purified by flash chromatography (60-95% of EtOAc in n-hexane) to afford the title compound (0.23 g, 0.707 mmol, 79 %). LC-MS, [M+H] + = 326.4, {Method E: t R = 1.08 min}.

Step-5: Synthesis of (3aS,7S,7aR)-7-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)tetrahydro-3aH-[l,3,2]dioxathiolo[4,5-c]pyran 2,2-dioxide(racemate): To a stirred solution of (3aS,7S,7aR)-7-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)tet rahydro-3aH- [l,3,2]dioxathiolo[4,5-c]pyran 2-oxide (0.2 g, 0.615 mmol) in acetonitrile (3 mL) and water (1 mL), was added sodium periodate (0.263 g, 1.230 mmol) and ruthenium(III) chloride hydrate (0.014 g, 0.061 mmol) at rt and stirred for 12 h. Solvent was removed under reduced pressure, crude was extracted with EtOAc (2 X 50 mL), washed with water, brine and dried over sodium sulphate. The solvent was removed under reduced pressure and crude residue was purified by flash chromatography (80-100% of EtOAc in n-hexane) to afford the title compound (160 mg, 0.469 mmol, 76 %). LC-MS, [M+H] + = 342.2, {Method F: tR = 2.028 min}.

Step-6: Synthesis of (3R,4R,5S)-3-azido-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)tetrahydro-2H-pyran-4-ol(racemate): To a stirred solution of (3aS,7S,7aR)-7-(4-(3- fluorophenyl)-lH-l,2,3-triazol-l-yl)tetrahydro-3aH-[l,3,2]di oxathiolo[4,5-c]pyran 2,2- dioxide (160 mg, 0.469 mmol) in DMF (4 mL) was added sodium azide (122 mg, 1.875 mmol) at rt. Then the reaction mixture was heated at 60 °C for 3 h. Reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was dissolved in THF (2 mL), 1 mL of stock solution [1 mL H2SO4 + 0.4 mL of water + 8.6 mL of THF] was added at 0°C and stirred for 30 min. The reaction mixture was basified with NaHCCh, filtered thorough Celite pad and washed with excess EtOAc. The filtrate was concentrated under reduced pressure to give crude residue which was purified by flash chromatography (60-95% of EtOAc in n-hexane) to afford the title compound (90 mg, 0.296 mmol, 63%). LC-MS, [M+H] + = 305.2, {Method C: tR = 1.94 min}.

Step-7: Synthesis of (3R,4S,5S)-3-amino-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol- l-yl)tetrahydro-2H-pyran-4-ol(racemate): To a stirred solution of (3R,4R,5S)-3-azido-5- (4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)tetrahydro-2H-pyra n-4-ol (90 mg, 0.296 mmol) in MeOH (4 mL),was added 10% Pd/C (6.30 mg, 0.030 mmol) and stirred at rt under hydrogen atmosphere for 16 h. The reaction mixture filtered through celite, washed with excess MeOH and the filtrate was concentrated to afford the title compound (60 mg,

0.216 mmol, 73%). LC-MS, [M+H] + = 279.5, {Method E: t R = 0.72min}

Step-8: To a stirred solution of (3R,4S,5S)-3-amino-5-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)tetrahydro-2H-pyran-4-ol (45.8 mg, 0.165 mmol) in DMF (2 mL), was added DIPEA (0.192 mL, 1.098 mmol) at rt. After 5 min, (4aR,6R,7R,8R,8aR)-8-(4-(3- fluorophenyl)- 1E1- 1 ,2,3-triazol-l -yl)-7-methoxy-2-phenylhexahy dropyrano[3,2- d][l,3]dioxine-6-carboxylic acid (50 mg, 0.110 mmol) and HATU (104 mg, 0.274 mmol) were added and the reaction mixture was stirred at rt for 12 h. Then the reaction mixture was diluted with ice cold water and stirred for 10 min. The obtained solid was filtered and dried to afford crude residue containing (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-l H-l,2,3-triazol-l-yl)-4- hydroxytetrahydro-2H-pyran-3-yl)-7-methoxy-2-phenylhexahydro pyrano[3,2- d][l,3]dioxine-6-carboxamide as a diasteromeric mixture. The crude residue was purified by prep-HPLC to separate the two diastereomers.

Prep HPLC Method info: Column : Lux-cellulose C4(250 X21.2)mm , 5 micron; Mobile.Phase A: -Mobile.Phase B: 0.1%DEA IN MeOH; Flow : 19 mL/min; Time(min)/%B: 0/100, 20/100;

Isomer 1: (20 mg, 0.028 mmol, 25.5 % yield) LC-MS, [M+H] + = 716.0, {Method F: tR = 2.287 min}

Isomer 2: (18 mg, 0.025 mmol, 22.91 % yield). LC-MS, [M+H] + = 716.0, {Method F: t R = 2.30 min}

Step-9: (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N- ((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-h ydroxytetrahydro-2H-pyran- 3-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d][l,3]dioxine-6 -carboxamide Isomer 1(20 mg, 0.028 mmol) was suspended in 80% Aq.AcOH (1 mL) and heated at 70 °C for 14 h. Then, reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude material was purified via preparative HPLC [Method A] to afford Example 20a: (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxytetrahydro-2H-pyran-3-yl)-5-hydroxy-6-(hydroxymethyl) -3-methoxytetrahydro- 2H-pyran-2-carboxamide Isomer 1 (5 mg, 7.89 pmol, 28%). LC-MS, [M+H] + = 628.1, {Method A: t R = 1.618 min}; 1H NMR (400MHz, MEOH-d4) d = 8.63 (s, 1H), 8.48 (s, 1H), 7.70 (dd, J=7.7, 3.1 Hz, 2H), 7.66 - 7.61 (m, 2H), 7.53 - 7.46 (m, 2H), 7.13 (td, J=8.1, 4.0 Hz, 2H), 4.95 (dd, J=10.6, 2.8 Hz, 1H), 4.67 (d, J=4.4 Hz, 1H), 4.35 - 4.25 (m, 3H), 4.21 (dd, J=10.3, 4.9 Hz, 1H), 4.15 (d, J=2.7 Hz, 1H), 4.11 - 4.05 (m, 1H), 4.03 - 3.94 (m, 2H), 3.87 - 3.81 (m, 2H), 3.79 - 3.73 (m, 1H), 3.47 (t, J=10.9 Hz, 1H), 3.20 (s, 3H). hGal3 IC 5 o = 36 pM.

Example 20b: (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl )-N- ((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-h ydroxytetrahydro-2H-pyran- 3-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro-2H-pyr an-2-carboxamide Isomer 2 (5.5 mg, 8.76 pmol, 34.8 % yield) was prepared following the same procedure used in step 9 for example 20a but using (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l ,2,3-triazol-l-yl)-4- hydroxytetrahydro-2H-pyran-3-yl)-7-methoxy-2-phenylhexahydro pyrano[3,2- d][l,3]dioxine-6-carboxamide Isomer 2 as starting material. LC-MS, [M+H] + = 628.1, {Method A: tR = 1.610 min}. ¾ NMR (400MHz, MEOH-dr) d = 8.63 (s, 1H), 8.48 (s, 1H), 7.70 (dd, .7=7.7, 3.1 Hz, 2H), 7.66 - 7.61 (m, 2H), 7.53 - 7.46 (m, 2H), 7.13 (td, .7=8.1, 4.0 Hz, 2H), 4.95 (dd, =10.6, 2.8 Hz, 1H), 4.67 (d, =4.4 Hz, 1H), 4.35 - 4.25 (m, 3H), 4.21 (dd, =10.3, 4.9 Hz, 1H), 4.15 (d, =2.7 Hz, 1H), 4.11 - 4.05 (m, 1H), 4.03 - 3.94 (m, 2H), 3.87 - 3.81 (m, 2H), 3.79 - 3.73 (m, 1H), 3.47 (t, .7=10.9 Hz, 1H), 3.20 (s, 3H). hGal3 ICso = 0.09 pM.

Example 21: Synthesis of (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N-((3R,4S,5S)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-y l)-4-hydroxytetrahydro-2H- pyran-3-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxytetrahydro- 2H-pyran-2- carboxamide

Step-1: Synthesis of N-(3,6-dihydro-2H-pyran-3-yl)-N-methyl-4- nitrobenzenesulfonamide(racemate): To a stirred solution of 3,6-dihydro-2H-pyran-3-ol (1.667 g, 16.65 mmol), N-methyl-4-nitrobenzenesulfonamide (3.0 g, 13.88 mmol) in

THF (30 mL), was added triphenylphosphine (7.28 g, 27.8 mmol) and DIAD (5.40 mL, 27.8 mmol) at 0 oC. The mixture was allowed to warm to rt and stirred for 18 h. The reaction mixture was extracted with EtOAc (3X 50 mL), washed with water, brine, dried over sodium sulphate and concentrated. The crude residue was purified by flash chromatography (0-30% EtOAc in n-hexane) to afford the title compound (4 g, 13.41 mmol, 97%) as an off-white solid. 1H NMR (400 MHz, CHLOROFORM-d) d ppm 8.34 - 8.41 (m, 2 H), 7.97 - 8.08 (m, 2 H), 6.02 - 6.09 (m, 1 H), 5.36 - 5.42 (m, 1 H), 4.37 - 4.43 (m, 1 H), 4.08 - 4.15 (m, 1 H), 3.96 - 4.04 (m, 1 H), 3.76 (d, J=4.0 Hz, 2 H), 2.92 (s, 3 H). Step-2: Synthesis of N-((3S,4R,5S)-4,5-dihydroxytetrahydro-2H-pyran-3-yl)-N- methyl-4-nitrobenzenesulfonamide(racemate): To a stirred solution of N-(3,6-dihydro- 2H-pyran-3-yl)-N-methyl-4-nitrobenzenesulfonamide (4.0 g, 13.41 mmol) in acetone (50 mL) and water (13.33 mL) was added 4-methylmorpholine-N-oxide (2.356 g, 20.11 mmol) and osmium tetroxide (5.05 mL, 0.402 mmol, 2.5% w/v in t-butanol) at rt and stirred for 16 h. The reaction mixture was quenched with sat.Na2S03 solution and acetone was removed under reduced pressure to give crude residue. The crude was extracted with EtOAc (2x100 mL), washed with water, brine, dried over sodium sulphate and concentrated. The residue was purified by flash chromatography (80 - 100% EtOAc in n-hexane) to afford the title compound (3.5 g, 10.53 mmol, 79%) as an off-white solid. ¾ NMR (300 MHz, DMSO-rie) d ppm 8.36 (d, J= 8.7 Hz, 2 H), 8.06 (d, J= 8.7 Hz, 2 H), 4.75 (d, =4.2 Hz, 1 H), 4.53 (d, .7=6.4 Hz, 1 H), 3.98 (br td, .7=10.6, 4.9 Hz, 1 H), 3.50 - 3.70 (m, 4 H), 3.24 - 3.37 (m, 2 H, merged with moisture peak), 2.79 (s, 3 H).

Step-3: Synthesis ofN-((3aS,7S,7aR)-2,2-dioxidotetrahydro-3aH-

[1.3.2]dioxathiolo[4,5-c]pyran-7-yl)-N-methyl-4-nitrobenz enesulfonamide(racemate): To a stirred solution of N-((3S,4R,5S)-4,5-dihydroxytetrahydro-2H-pyran-3-yl)-N-methy l-4- nitrobenzenesulfonamide (3.5 g, 10.53 mmol) in DCM (30 mL), TEA (2.94 mL, 21.06 mmol) and thionyl chloride (1.537 mL, 21.06 mmol) were added sequentially at 0 °C and stirred for 1 h. Then the reaction mixture was extracted with DCM (2x75 mL), washed with water, brine and dried over sodium sulphate. Solvent was removed under reduced pressure to give a residue that was taken for the next step without further purification.

To a stirred solution of N-methyl-4-nitro-N-((3aS,7S,7aR)-2-oxidotetrahydro-3aH-

[1.3.2]dioxathiolo[4,5-c]pyran-7-yl)benzenesulfonamide (3.8 g, 10.04 mmol) in acetonitrile (100 mL)/water (66.7 mL), sodium periodate (4.30 g, 20.09 mmol) and ruthenium(III) chloride hydrate (0.163 g, 0.036 mmol) were added sequentially at 0 °C. Then the reaction mixture was allowed to reach rt and stirred for 12 h. The solvent was removed under reduced pressure to give crude residue, crude was extracted with EtOAc (150 mL), washed with water (100 mL), sat NaHS04 solution (3x 50mL) and dried over Na2S04. The solvent was removed under reduced pressure to give crude residue which was purified by flash chromatography (70 - 100% EtOAc in n-hexane) to afford N- ((3aS,7S,7aR)-2,2-dioxidotetrahydro-3aH-[l,3,2]dioxathiolo[4 ,5-c]pyran-7-yl)-N-methyl- 4-nitrobenzenesulfonamide (3.5 g, 8.87 mmol, 88%) as an off-white solid. 'H NMR (400 MHz, DMSO-rie) d ppm 8.42 (d, J= 9.0 Hz, 2 H), 8.06 - 8.10 (m, 2 H), 5.55 - 5.61 (m, 1 H), 5.38 - 5.42 (m, 1 H), 4.23 - 4.32 (m, 2 H), 3.76 (dd, .7=14.8, 1.8 Hz, 1 H), 3.62 - 3.68 (m, 1 H), 3.48 - 3.55 (m, 1 H), 2.89 (s, 3 H). Step-4: Synthesis of N-((3S,4S,5R)-5-azido-4-hydroxytetrahydro-2H-pyran-3-yl)- N-methyl-4-nitrobenzenesulfonamide(racemate): To a stirred solution ofN- ((3aS,7S,7aR)-2,2-dioxidotetrahydro-3aH-[l,3,2]dioxathiolo[4 ,5-c]pyran-7-yl)-N-methyl- 4-nitrobenzenesulfonamide (0.7 g, 1.775 mmol) in DMF (15 mL) was added sodium azide (0.462 g, 7.10 mmol) at rt. Then the reaction mixture was heated at 60 °C for 2 h. Reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was dissolved in THF (8.6 mL), 0.4 mL of water and 1 mL of H2SO4 was added at 0°C and stirred for 30 min. The reaction mixture was basified with solid NaHCCh, filtered thorough celite pad and washed with excess EtOAc. The filtrate was concentrated under reduced pressure to give crude residue which was purified by flash chromatography (30-50% of EtOAc in n-hexane) to afford the title compound (0.5 g, 1.352 mmol, 76%). LC-MS, [M+18] + = 375.2, {Method C: te = 1.917min}.

Step-5: Synthesis of N-((3S,4S,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)- 4-hydroxytetrahydro-2H-pyran-3-yl)-N-methyl-4-nitrobenzenesu lfonamide(racemate):

To a solution of N-((3S,4S,5R)-5-azido-4-hydroxytetrahydro-2H-pyran-3-yl)-N-m ethyl- 4-nitrobenzenesulfonamide (2.2 g, 6.16 mmol) in DMF (20 mL) and water (5.00 mL) was added sodium ascorbate (1.220 g, 6.16 mmol), copper(II) sulfate pentahydrate (1.383 g, 5.54 mmol) and l-ethynyl-3-fluorobenzene (2.85 mL, 24.63 mmol) at rt. The reaction mixture was degassed N2 for 10 min and heated at 80 °C for 30 min. Then the reaction mixture was cooled to rt, diluted with DCM (100 mL)/water (100 mL) and stirred for 30 minutes. The organic layer was separated and aqueous layer was re-extracted with DCM (2x50mL). Combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated. The crude residue was purified by flash chromatography (0 - 15% MeOH in DCM) to afford N-((3S,4S,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-4-hydroxytetrahydro-2H-pyran-3-yl)-N-methyl-4-nitrobenze nesulfonamide (2.1 g, 4.40 mmol, 71%) as an off-white solid. LC-MS, [M+H] + = 478.2, {Method C: tR = 2.453 min}.

Step-6: Synthesis of (3R,4R,5S)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5- (methylamino)tetrahydro-2H-pyran-4-ol: To a solution of N-((3S,4S,5R)-5-(4-(3- fluorophenyl)- 1H- 1 ,2,3-triazol- 1 -yl)-4-hy droxytetrahydro-2H-pyran-3-yl)-N-methyl-4- nitrobenzenesulfonamide (1 g, 2.094 mmol) in acetone (10 mL), potassium carbonate (0.868 g, 6.28 mmol) and benzenethiol (0.346 g, 3.14 mmol) were added sequentially at rt and stirred for 16 h. Then, the solvent was removed under reduced pressure to give crude residue which was purified by flash chromatography (0-20% MeOH(10% ammonia)/DCM) to afford (3R,4R,5S)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5- (methylamino)tetrahydro-2H-pyran-4-ol as a racemic mixture. The racemic mixture was further purified by SFC to separate the enantiomers.

Preparative chiral HPLC conditions:

Preparative Column: Chiralpak IA (250 X 30)mm,5um BPR pressure: 100 bars

Temperature: 25 °C

Flow rate: 60 g/min

Mobile Phase: C0 2 / 0.2% DEA in MeOH (70/30) Detector Wavelength: 245 nm Sample preparation: 10 mg / 1 mL MeOH:

Analytical chiral HPLC conditions:

Analytical Column: Chiralpak IA (250 X 30)mm,5um BPR pressure: 100 bars Temperature: 30 °C

Flow rate: 3 g/min

Mobile Phase: CO2/ 0.2% DEA in MeOH (70/30) Detector Wavelength: UV 200-400 nm Enantiomer 1 (undesired): 0.15 g: chiral HPLC /R = 3.63 min; LC-MS, [M+H] + = 293.2, {Method C: tR = 0.934 min};

Enantiomer 2 (desired): 0.18 g: chiral HPLC R = 5.77 min; LC-MS, [M+H] + = 293.1, {Method C: tR = 0.941 min}. Step-7: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d] [l,3]dioxine-6-carboxylic acid (200 mg, 0.439 mmol) in DMF (5 mL), HATU (334 mg, 0.878 mmol), DIPEA (0.767 mL, 4.39 mmol) and (3R,4R,5S)-3-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-5- (methylamino)tetrahydro-2H-pyran-4-ol Enantiomer 2 (128 mg, 0.439 mmol) were added sequentially at rt and stirred for 16 h. The reaction mixture was poured into ice cold water (100 mL) and stirred for 10 min. The obtained solid was filtered and dried to afford crude residue which was further purified by flash chromatography (0-10% MeOH in DCM) to afford (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N-((3S,4R,5R)- 5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-hydroxytetrah ydro-2H-pyran-3-yl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamide (0.195 g, 0.259 mmol, 59%) as an off-white solid. LC-MS, [M+H] + = 730.2, {Method C: te = 3.051 min};

Step-8: (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l- yl)-N- ((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-triazol-l-yl)-4-h ydroxytetrahydro-2H-pyran- 3-yl)-7-methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3] dioxine-6-carboxamide (195 mg, 0.267 mmol) was suspended in 70% Aqueous AcOH (10 mL) and heated at 70 °C for 16 h. Then reaction mixture was cooled to rt and solvent was removed under reduced pressure to give crude residue. The crude residue was purified by Prep-HPLC Method B to afford Example 21: (2R,3R,4S,5R,6R)-4-(4-(3-fluorophenyl)-lH-l,2,3- triazol-l-yl)-N-((3S,4R,5R)-5-(4-(3-fluorophenyl)-lH-l,2,3-t riazol-l-yl)-4- hydroxytetrahydro-2H-pyran-3-yl)-5-hydroxy-6-(hydroxymethyl) -3-methoxy-N- methyltetrahydro-2H-pyran-2-carboxamide (0.095 g, 0.148 mmol, 55%) as an off-white solid. LC-MS, [M+H] + = 642.2, {Method C: t R = 1.95}. ¾ NMR (400 MHz, MEOH- dt) d ppm 8.70 (d, J=8.5 Hz, 1 H), 8.49 (d, J=8.0 Hz, 1 H), 7.57 - 7.71 (m, 4 H), 7.43 - 7.49 (m, 2 H), 7.06 - 7.12 (m, 2 H), 4.98 (br d, J=8.0 Hz, 1 H), 4.62 - 4.78 (m, 1 H), 4.41 - 4.60 (m, 3 H), 4.06 - 4.26 (m, 3 H), 3.65 - 4.01 (m, 6 H), 2.99 - 3.26 (m, 6 H)

[*rotameric mixture]; hGal3 IC50 = 0.03 mM.

Example 22: Synthesis of (2R,3R,4S,5R,6R)-N-((3S,4R,5R)-5-(5-fluoro-lH- pyrazolo[3,4-b]pyridin-l-yl)-4-hydroxytetrahydro-2H-pyran-3- yl)-4-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-5-hydroxy-6-(hydroxymethyl)-3-methoxy -N-methyltetrahydro-2H- pyran-2-carboxamide (Isomer 1 and 2)

Example 22a & 22b

Step-1: To a stirred solution of 5-fluoro-lH-pyrazolo[3,4-b]pyridine (67.8 mg, 0.494 mmol) in THF (3 mL), 18-crown-6 (171 mg, 0.647 mmol) and NaH (25.9 mg, 0.647 mmol, 60% w/w) were added at rt and refluxed at 80 °C for 10 min. Then N-

((3aS,7S,7aR)-2,2-dioxidotetrahydro-3aH-[l,3,2]dioxathiol o[4,5-c]pyran-7-yl)-N-methyl-

4-nitrobenzenesulfonamide (150 mg, 0.380 mmol) in THF (3 mL) was added dropwise over a period of 5 min and refluxed at 80 °C for 16 h. Reaction mixture was cooled to 0 ° C, acidified with con.HCl (around pH=l) and further refluxed at 80 °C for 2 h. The reaction mixture was cooled to rt, neutralized with aq.10% NaHC03 solution and extracted with EtOAc (2 X 25 mL). The combined organic extracts were washed with water, brine, dried over sodium sulphate and concentrated. The crude residue was purified by flash chromatography (60-80% EtOAc in n-hexane) to afford N-((3S,4S,5R)-

5-(5-fluoro-lH-pyrazolo[3, 4-b]pyri din-1 -yl)-4-hy droxytetrahy dro-2H-pyran-3-yl)-N- methyl-4-nitrobenzenesulfonamide as a racemate. The racemate was further purified by chiral SFC to afford pure enantiomer- 1 and enantiomer-2. Preparative chiral HPLC conditions:

Preparative Column: Chiralpak ADH (250 X 30)mm,5um

BPR pressure: 100 bars Temperature: 25 °C

Flow rate: 60 g/min

Mobile Phase: C0 2 / 0.4% DEA in EtOH (60/40) Detector Wavelength: 245 nm Sample preparation: 10 mg / 1 mL MeOH:

Analytical chiral HPLC conditions:

Analytical Column: Chiralpak ADH (250 X 4.6)mm,5um

BPR pressure: 100 bars Temperature: 25 °C

Flow rate: 3 g/min

Mobile Phase: CO2/ 0.4% DEA in EtOH (60/40) Detector Wavelength: UV 200-400 nm

Enantiomer 1: (50 mg, 0.111 mmol, 29.1 % yield); chiral HPLC /R = 5.59 min; LC-MS, [M+H] + = 452.2, {Method C: t R = 2.352 min};

Enantiomer 2: (50 mg, 0.111 mmol, 29.1 % yield); chiral HPLC /R = 8.23 min; LC-MS, [M+H] + = 452.2, {Method C: t R = 2.353 min}.

Step-2: To a stirred solution of N-((3S,4S,5R)-5-(5-fluoro-lH-pyrazolo[3,4- b]pyridin-l-yl)-4-hydroxytetrahydro-2H-pyran-3-yl)-N-methyl- 4- nitrobenzenesulfonamide Enantiomer 1 (50 mg, 0.111 mmol) in acetone (2 mL) was added K2CO3 (45.9 mg, 0.332 mmol) followed by thiophenol (0.031 mL, 0.299 mmol) at rt and stirred for 2 h. Then the solvent was removed under reduced pressure to give crude residue and the crude was purified by flash chromatography (5-15% MeOH(5% aq.NH3) in CHCh) to afford (3R,4R,5S)-3-(5-fluoro-lH-pyrazolo[3,4-b]pyridin-l-yl)-5-

(methylamino)tetrahydro-2H-pyran-4-ol (20 mg, 0.075 mmol, 68%). LC-MS, [M+H] + = 267.2, {Method D: tR = 0.57 min}. Step-3: To a stirred solution of (4aR,6R,7R,8R,8aR)-8-(4-(3-fluorophenyl)-lH- l,2,3-triazol-l-yl)-7-methoxy-2-phenylhexahydropyrano[3,2-d] [l,3]dioxine-6-carboxylic acid (25 mg, 0.055 mmol) and (3R,4R,5S)-3-(5-fluoro-lH-pyrazolo[3,4-b]pyridin-l-yl)- 5-(methylamino)tetrahydro-2H-pyran-4-ol (14.62 mg, 0.055 mmol) in DMF (0.6 mL), DIPEA (0.048 mL, 0.274 mmol) and HATU (31.3 mg, 0.082 mmol) were added sequentially at rt and stirred for 16 h. Then the reaction mixture was diluted with 20 mL of ice cold water and stirred for 10 min. The obtained solid was filtered and dried to afford (4aR,6R,7R,8R,8aR)-N-((3S,4R,5R)-5-(5-fluoro-lH-pyrazolo[3,4 -b]pyridin-l-yl)-

4-hydroxytetrahydro-2H-pyran-3-yl)-8-(4-(3-fluorophenyl)- lH-l,2,3-triazol-l-yl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamide (25 mg, 0.036 mmol, 65%). LC-MS, [M+H] + = 704.4, {Method D: t R = 1.28 min}.

Step-4: (4aR,6R,7R,8R,8aR)-N-((3S,4R,5R)-5-(5-fluoro-lH-pyrazolo[3,4 -b]pyridin-l- yl)-4-hydroxytetrahydro-2H-pyran-3-yl)-8-(4-(3-fluorophenyl) -lH-l,2,3-triazol-l-yl)-7- methoxy-N-methyl-2-phenylhexahydropyrano[3,2-d][l,3]dioxine- 6-carboxamide (30 mg, 0.043 mmol) was suspended in 70% aq.AcOH (10 mL) and heated at 75 °C for 16 h. Reaction mixture was cooled to rt and purified by prep-HPLC Method A to afford Example 22a: (2R,3R,4S,5R,6R)-N-((3S,4R,5R)-5-(5-fluoro-lH-pyrazolo[3,4-b ]pyridin- l-yl)-4-hydroxytetrahydro-2H-pyran-3-yl)-4-(4-(3-fluoropheny l)-lH-l,2,3-triazol-l-yl)-

5-hydroxy-6-(hydroxymethyl)-3-methoxy-N-methyltetrahydro- 2H-pyran-2-carboxamide Isomer 1 (13.4 mg, 0.022 mmol, 51%). LC-MS, [M+H] + = 616.1, {Method A: t R = 1.54 min}. ¾ NMR (400MHz, MEOH-d 4 ) d = 8.71 (d, J=7.0 Hz, 1H), 8.52 (dt, J= 6.5, 2.0 Hz, 1H), 8.15 (d, =3.0 Hz, 1H), 7.97 (dt, .7=8.4, 3.1 Hz, 1H), 7.70 (d, J=8.0 Hz, 1H), 7.67 - 7.60 (m, 1H), 7.47 (td, J= 7.9, 5.8 Hz, 1H), 7.09 (td, .7=8.5, 2.5 Hz, 1H), 5.16 - 4.95 (m, 2H), 4.79 - 4.65 (m, 1H), 4.58 - 4.51 (m, 1H), 4.46 - 4.41 (m, 1H), 4.25 - 4.13 (m, 2H), 4.09 - 3.88 (m, 3H), 3.88 - 3.65 (m, 4H), 3.26 (s, 1H), 3.12 (s, 2H), 3.09 (s, 2H), 3.01 (s, 1H). hGal3 % = >10 mM.

Example 22b: Prepared in a similar fashion as described for Example 22a, by using enantiomer 2 in Step-2 as the starting material. LC-MS, [M+H]+ = 616.1,

{Method A : tR = 1.54 min}. ¾ NMR (400 MHz, MEOH-ώ) d ppm 8.67 - 8.75 (m, 1 H), 8.39 - 8.53 (m, 1 H), 8.13 - 8.22 (m, 1 H), 7.98 (d, J= 2.5 Hz, 1 H), 7.38 - 7.80 (m, 3 H), 7.02 - 7.17 (m, 1 H), 5.18 - 5.32 (m, 1 H), 4.92 - 5.12 (m, 2 H), 4.71 - 4.82 (m, 2 H), 4.38 - 4.49 (m, 1 H), 3.66 - 4.15 (m, 8 H), 3.27 (s, 1 H), 3.09 - 3.12 (m, 3 H), 3.04 (s, 2 H) (rotameric mixture). hGal3 ICso = 0.04.