2020, OPR&D - Improvement of The C-Glycosylation Step For The Synthesis of Remdesivir PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Subscriber access provided by CARLETON UNIVERSITY

Communication
Improvement of the C-glycosylation Step for the Synthesis of Remdesivir
Fei Xue, Xinbo Zhou, Ruijie Zhou, Xiaohan Zhou, Dian Xiao, Wu Zhong,
Eric Gu, Xiaowen Guo, Ji Xiang, Ke Wang, Likai Yang, and Yong Qin
Org. Process Res. Dev., Just Accepted Manuscript • DOI: 10.1021/acs.oprd.0c00310 • Publication Date (Web): 05 Aug 2020
Downloaded from pubs.acs.org on August 5, 2020

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted
online prior to technical editing, formatting for publication and author proofing. The American Chemical
Society provides “Just Accepted” as a service to the research community to expedite the dissemination
of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in
full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully
peer reviewed, but should not be considered the official version of record. They are citable by the
Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore,
the “Just Accepted” Web site may not include all articles that will be published in the journal. After
a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web
site and published as an ASAP article. Note that technical editing may introduce minor changes
to the manuscript text and/or graphics which could affect content, and all legal disclaimers and
ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or
consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W.,


Washington, DC 20036
Published by American Chemical Society. Copyright © American Chemical Society.
However, no copyright claim is made to original U.S. Government works, or works
produced by employees of any Commonwealth realm Crown government in the course
of their duties.
Page 1 of 21 Organic Process Research & Development

1
2
3
4
5
6
7
8
Improvement of the C-glycosylation Step for the
9
10
11
12
Synthesis of Remdesivir
13
14
15
16 Fei Xue,†,‣ Xinbo Zhou,‡,‣ Ruijie Zhou,† Xiaohan Zhou,† Dian Xiao,‡ Wu Zhong,*, ‡ Eric Gu,§
17
18
19 Xiaowen Guo,§ Ji Xiang,§ Ke Wang,# Likai Yang,# Yong Qin*,†
20
21
22 AUTHOR ADDRESS
23
24
25 † Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and
26
27
28
Sichuan Province, and Sichuan Research Center for Drug Precision Industrial Technology, West
29
30 China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan Province, China.
31
32
33 ‡ National Engineering Research Center for the Emergence Drugs, Beijing Institute of
34
35 Pharmacology and Toxicology, Beijing 100850, China.
36
37
38
39 § Shanghai Syncores Technology Inc. Ltd., Shanghai 201203, China.
40
41
42
43 # Chengdu Open Pharmaceutical Co., Ltd., Qionglai 611500, Sichuan Province, China.
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59 1 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 2 of 21

1
2
3 For Table of Contents Only
4
5
6
NH2
7 Si Si
8 H
N H Cl Cl N
9 O O N
10 BnO i-Pr2NH, n-BuLi O N
N
11 N + BnO OH
12 Br N BnO OBn THF, –78 ºC
13 BnO OBn
10.0 g scale: 74% (chromatography)
14
180.0 g scale: 62% (recrystallization)
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59 2 Environment
60 ACS Paragon Plus
Page 3 of 21 Organic Process Research & Development

1
2
3 ABSTRACT: The bulk supply of the antiviral C-nucleoside analogue remdesivir is largely
4
5
6 hampered by a low-yielding C-glycosylation step in which the base is coupled to the pentose
7
8 unit. Here we disclose a significantly improved methodology for this critical transformation. By
9
10 utilizing diisopropylamine as a cost-effective additive, the addition reaction furnishes an optimal
11
12
13
yield of 75% of the desired ribofuranoside adduct, representing the highest yield obtained thus
14
15 far for this key step. The method proved suitable for hectogram scale synthesis without column
16
17 chromatographic operations.
18
19
20
21 KEYWORDS: Diisopropylamine, C-glycosylation, Remdesivir, COVID-19
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59 3 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 4 of 21

1
2
3 INTRODUCTION
4
5
6 The global outbreak of Corona Virus Disease 2019 (COVID-19) caused by severe acute
7
8 respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a severe threat to human health.1−5
9
10 Compared to its relatives (e.g., SARS-CoV and MERS-CoV), the new coronavirus exhibits a
11
12
13
more significant person-to-person transmission tendency. Most importantly, no drugs or vaccines
14
15 have thus far been approved to treat infections of SARS-CoV-2, which seems to render this
16
17 pandemic uncontrollable, with the number of infected cases worldwide exceeding ten million.
18
19
Under such circumstances, several existing antiviral drugs and drug candidates are actively
20
21
22 evaluated for their therapeutic potential for this disease.6,7,8 Among them, remdesivir (GS-5734,
23
24 1; Figure 1) represents one of the most promising compounds.8,9,10
25
26
27 NH2
28
29 N
30 O
O N
31 O N
32 O HN P O CN
33 O
34 HO OH
35
36 Remdesivir
37
38
39 Figure 1. Structure of remdesivir.
40
41
42
43 Remdesivir is an investigational small-molecule drug originally developed to treat Ebola
44
45 virus infections and later found to exhibit broad-spectrum activity against various RNA
46
47 viruses.11−17 In particular, a recent study (published on February 4th, 2020) clearly demonstrated
48
49
50
that remdesivir was effective against SARS-CoV-2 in vitro with an EC50 of 0.77 μM in Vero E6
51
52 cells.17 Of note, approximately one week prior to this report, Holshue et al disclosed that the first
53
54 COVID-19 patient in the USA had gradually recovered after receiving a compassionate
55
56
57
58
59 4 Environment
60 ACS Paragon Plus
Page 5 of 21 Organic Process Research & Development

1
2
3 administration of remdesivir.18 Subsequently, a number of clinical trials have been launched
4
5
6 worldwide to evaluate the efficacy and safety of remdesivir, 19 and other compassionate use of
7
8 this molecule has also been initiated,20 in the hope that it would benefit the treatment of the
9
10 epidemic COVID-19. Although the earliest clinical trials conducted in China did not yield
11
12
13
positive results in terms of reducing the death rate of severely affected patients due to
14
15 insufficient data,21 preliminary data from a clinical trial led by the National Institute of Allergy
16
17 and Infectious Diseases (NIAID) suggest a promising therapeutic effect of remdesivir.22 On May
18
19
1st 2020, the Food and Drug Administration (FDA) issued an Emergency Use Authorization
20
21
22 (EUA) for remdesivir to treat COVID-19 patients.22 Once further evidence supports the broad
23
24 clinical use of remdesivir, the bulk supply of the active pharmaceutical ingredients required for
25
26 the production of remdesivir would be an urgent concern.23
27
28
29
30 Structurally, remdesivir consists of three fragments: an adenine analogue (base), a pentose
31
32 unit, and a phosphoramidate side chain. A key challenge in the preparation of this C-nucleoside
33
34 analogue is the coupling of the ribose and base moieties. Previously, two-generation syntheses of
35
36
37
remdesivir, including three different methods for accessing ribofuranoside 4, have been
38
39 developed by chemists at Gilead Sciences (2 + 3 to 4; Figure 2).11,12,24-27 Temporary silyl
40
41 protection of the free primary amine group in the base fragment 2 was required in the addition
42
43
reaction between 2 and 3. Two silyl reagents (i.e., trimethylchlorosilane and 1,1,4,4-tetramethyl-
44
45
46 1,4-dichloro disilethylene (5)) were employed to generate the corresponding protected
47
48 intermediates 6 and 7,28 respectively. In the first generation synthesis, subjection of bromide 2a
49
50 and ribolactone 3 to the conditions of TMSCl and n-BuLi at –78 º C furnished the desired C-
51
52
53 glycosylation product 4 in a low yield of 25% (method A; Figure 2).24,25 Alternatively, the use of
54
55 disilane 5 and NaH in this transformation could deliver 4 in a reported yield of 60% on 4.4 g
56
57
58
59 5 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 6 of 21

1
2
3 scale (method B; Figure 2).12,26,27 The authors of the original studies added that “the efficiency of
4
5
6 both conditions (in the first generation synthesis) was suboptimal as the yields were capricious
7
8 and highly dependent on the cryogenic temperatures and the rate of n-BuLi addition required for
9
10 the transformation”.12 In the second generation synthesis (method C; Figure 3), iodide 2b was
11
12
13
employed as the source of the base unit, with a combination of TMSCl, PhMgCl, and i-
14
15 PrMgCl·LiCl reagents, which resulted in a 40% yield of 4.11,25,26 We attempted to reproduce
16
17 these documented procedures on a decagram scale11,25,26 which resulted in the isolation of 4 with
18
19
yields in the range of 21–42% (see Table S1 in the Supporting Information). Evidently, none of
20
21
22 the three above approaches efficiently addressed this problematic synthetic step, and the
23
24 aforementioned techniques are not suitable for scale-up preparation of remdesivir. Until very
25
26 recently, during the course of our manuscript preparation, the Gilead scientists reported an
27
28
29 optimization of method C for the critical C-glycosylation step, in which NdCl3 and n-Bu4NCl
30
31 were employed to facilitate the addition of lactone 3 with TMS-protected amine 2b (Method D;
32
33 Figure 2).29 The 282 kg scale-up synthesis of compound 4 with 69% yield proved the reliability
34
35
36
of this method. In this report, we describe an alternative improvement for the C-glycosylation
37
38 step, which renders the synthesis of remdesivir simple and efficient, with the aim of meeting the
39
40 demand for its large-scale synthesis.
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59 6 Environment
60 ACS Paragon Plus
Page 7 of 21 Organic Process Research & Development

1
2
3
the 1st generation synthesis
4
5 NH2 method A24,25: 25% yield method B26,27: 60% yield NH2
6 2a, TMSCl, THF, rt; 2a, 5, NaH, THF, rt;
N N
7 n-BuLi, 3, –78 ºC n-BuLi, 3, –78 ºC
N N
8 the 2nd generation synthesis O N
N
9 X 11,12 method D29: 69% yield BnO OH
method C : 40% yield
10 2a X = Br 2b, TMSCl, PhMgCl, BnO OBn 4
2b, TMSCl, THF, rt; PhMgCl,
11 2b X = I i-PrMgCl, THF, –20 ºC;
i-PrMgCl•LiCl, 0 ºC; 3, –20 ºC
12 3, NdCl3, n-Bu4NCl, THF, –20 ºC
13
14
15 TMS TMS Si Si
16 O N N
O
17 BnO
Si Si N N
18
BnO OBn Cl Cl N N
19 N N
20 3 5 X 6 X 7
21
22
23
24
Figure 2. Reported methods for accessing a key intermediate (4) in the remdesivir synthesis.
25
26
27 RESULTS AND DISCUSSION
28
29 Efficient protection of the free amine group in 2 would be essential for the successful
30
31
addition reaction of base 2 to lactone 3. We envisioned that the addition of a secondary amine
32
33
34 might be helpful for improving the reaction efficiency (Figure 3). First, the secondary amine
35
36 could serve to activate disilane 5 through the formation of a disilyl substituted cyclic quaternary
37
38 amine salt 8,30 thereby promoting the exchange of the silyl groups between the inactive primary
39
40
41 amine in 2 and the secondary amine to form cyclic disilazane 7. Second, a stablized lithium
42
43 intermediate 9 could be formed in the presence of the secondary amine after lithium-halogen
44
45 exchange with 7, which would subsequently facilitate the addition of the base motif to lactone 3
46
47
48
to yield the desired ribofuranoside 4.
49
50
51
52
53
54
55
56
57
58
59 7 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 8 of 21

1
2
3 O NH2
NH2 O
4 BnO
5 N Si Si N
6 Cl Cl 5 BnO OBn 3 N
N O N
7 N
X BnO OH
8 2 R2NH, n-BuLi
4
9 X = Br or I BnO OBn
10 5
11
R2NLi
12 n-BuLi
13
14 Si Si
15 N
exchange R R Cl 8
16 +3
17 Si Si
18 N
Si Si
19 N
R2NH, n-BuLi N
20
N N
21 N
22 N Li
N
23 N Li
X 7 9
R R
24
25
26
27 Figure 3. Working hypothesis for an improved synthesis of 4.
28
29
30 With the above working hypothesis in mind, we initiated our studies by performing the
31
32
33 reactions with bromide 2a (1.0 gram scale) and lactone 3 employing disilane 5 in the presence of
34
35 n-BuLi with various secondary amines (Table 1). When secondary amines bearing two linear
36
37 alkyl groups (such as diethylamine, dipropylamine, and dibutylamine) were used, the addition
38
39
reactions proceeded to afford 4 in moderate yields (41–54%, entries 1–3). To our delight, bulkier
40
41
42 alkyl substituents in the secondary amine were beneficial for improving the reaction efficiency.
43
44 As a result, conducting the reaction in the presence of diisopropylamine led to the generation of
45
46 4 in 74% yield (entry 4). Notably, similar results were obtained with other bulky secondary
47
48
49 amines: diisobutylamine (71%, entry 5), dicyclohexylamine (70%, entry 6), and 2,2,6,6-
50
51 tetramethylpiperidine (74%, entry 7). For the purposes of comparison, the use of tertiary amines
52
53 was likewise evaluated. For instance, we performed the addition reaction of 2a and 3 under the
54
55
56
57
58
59 8 Environment
60 ACS Paragon Plus
Page 9 of 21 Organic Process Research & Development

1
2
3 identical conditions of entry 4, except for the replacement of diisopropylamine with
4
5
6 diisopropylethylamine, which resulted in a lower yield (49%, entry 8) of product 4.
7
8
9 Table 1. Effect of Organic Amines in the Addition Reactiona
10
11
12 O NH2
13 NH2 O
BnO N
14 N Si Si
15 Cl Cl 5 BnO OBn 3 N
N O N
16 N
BnO OH
17 Br 2a R2NH, n-BuLi, THF, –78 ºC, 2 h 4
18 BnO OBn
[1.0 gram scale]
19
20
21
22 Entry R2NH Yield (%) Entry R2NH Yield (%)
23
24
25 1
H
N 41 5 H
71
N
26
27
28
H
29 H N
30 2 N 45 6 70
31
32
33
34
H
35 3 N 54 7 74
N
36 H
37
38
39 H
40 4
N
74 8 N 49
41
42
43
44
45 a Reactions were conducted on 1.0 gram scale of 2a with 1.1 equiv. of 5, 1.1 equiv. of R2NH, 4.3
46
47
48 equiv. of n-BuLi, and 2.0 equiv. of 3 at 0.08 mol/L concentration unless otherwise stated. Yields
49
50 were determined according to the isolated materials through column chromatography.
51
52
53 Based on the above preliminary results, we then carried out scale-up experiments
54
55
56
employing diisopropylamine as an additive (Table 2). A 10 g-scale addition of 2a to lactone 3
57
58
59 9 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 10 of 21

1
2
3 was performed according to the conditions described in Table 1, leading to the generation of
4
5
6 adduct 4 in 75% yield (entry 1). To further improve the practicality of the addition process, we
7
8 sought to reduce the amount of solvent and increase the reaction concentration. Specifically, a
9
10 74% yield was observed when conducting the reaction at the concentration of 0.20 mol/L (entry
11
12
13
2), comparable to that of 0.08 mol/L (entry 1), while the reaction at 0.25 mol/L led to a slightly
14
15 diminished yield (70%, entry 3). Moreover, effect of the equivalents of lactone 3 was
16
17 investigated. Lowering the loading of 3 delivered inferior results (i.e., 1.8 equiv., 62%, entry 4;
18
19
1.6 equiv., 59%, entry 5) compared to that obtained with 2.0 equiv. of 3. With the optimized
20
21
22 reaction conditions (entry 2), a 180 g-scale experiment was conducted, which occurred smoothly
23
24 to provide 4 with 62% yield and 99.3% purity after recrystallization without the need of column
25
26 chromatographic operations (entry 6).
27
28
29
30 Table 2. Further optimization studies and scale-up synthesisa
31
32
33 NH2 O O NH2
34 BnO
N Si Si N
35 Cl Cl 5 BnO OBn 3
N N
36 N O N
37 Br BnO OH
i-Pr2NH, n-BuLi, THF, –78 ºC, 2 h
38 2a 4
BnO OBn
39
40
41
42
Entry Scale of 2a Equiv. of 3 Concentration (mol/L) Yield (%)
43
44
45 1 10.0 g 2.0 0.08 75b
46
47
48 2 10.0 g 2.0 0.20 74b
49
50
51
52
3 10.0 g 2.0 0.25 70b
53
54
55
56
57
58
59 10 Environment
60 ACS Paragon Plus
Page 11 of 21 Organic Process Research & Development

1
2
3 4 10.0 g 1.8 0.20 62b
4
5
6
7 5 10.0 g 1.6 0.20 59b
8
9
10 6 180.0 g 2.0 0.20 62c
11
12
13
a Reactions were conducted with 1.1 equiv. of 5, 1.1 equiv. of i-Pr2NH, and 4.3 equiv. of n-BuLi
14
15
16 unless otherwise stated. b Yields were calculated based on the materials isolated through column
17
18 chromatography. c The yield was obtained after recrystallization.
19
20
21
22 CONCLUSION
23
24 In summary, the crucial C-glycosylation step that ordinarily limits the overall efficiency of
25
26 remdesivir synthesis has been significantly improved by the use of a cost-effective secondary
27
28
29 amine, diisopropylamine. Thus, subjecting the base unit 2a and ribolactone 3 to the optimal
30
31 reaction conditions delivered up to 75% yield of the anticipated ribofuranoside 4, which, to the
32
33 best of our knowledge, represents the highest yield obtained for this critical transformation thus
34
35
36
far. Furthermore, the procedure is amenable to hectogram scale synthesis without column
37
38 chromatographic operations, and as such would be highly advantageous for the large-scale
39
40 manufacture of the antiviral, remdesivir.
41
42 EXPERIMENTAL DETAILS
43
44
45 General Information. All reagents were purchased from commercial suppliers and were used
46
47 without further purification. The product of the decagram scale synthesis was purified by flash
48
49 column chromatography on silica gel (200 – 300 meshes) from the Anhui Liangchen Silicon
50
51
52 Material Company in China. 1H NMR and 13C NMR spectra were recorded on Varian INOVA-
53
54 400/54, Agilent DD2-600/54 and calibrated by using deuterated dimethyl sulfoxide (DMSO-d6,
55
56
57
58
59 11 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 12 of 21

1
2
3 1H NMR: δ = 2.50, 13C NMR: δ = 39.5) unless otherwise noted. High-resolution mass spectra
4
5
6 (HRMS) were recorded on Agilent LC-MSD TOF ESI mass spectrometers.
7
8
9 General Procedure for Synthesis of Compound 4 by Using Different Amines. To an oven-
10
11 dried round-bottom flask equipped with a stir bar was added 7-bromopyrrolo[2,1-f][1,2,4]triazin-
12
13
14
4-amine (2a, 1.00 g, 4.694 mmol, 1.0 equiv.). The flask was evacuated and backfilled with argon
15
16 for three times. Under the protection of argon, anhydrous THF (40 mL) was added and the
17
18 mixture was stirred at room temperature for 5 min before a solution of 1,2-
19
20
bis(chlorodimethylsilyl) ethane (5, 1.111 g, 5.163 mmol, 1.1 equiv.) in anhydrous
21
22
23 tetrahydrofuran (5 mL) was added. The resulting mixture was stirred for 10 min. Then amine
24
25 (5.163 mmol, 1.1 equiv.) was added. After stirring at room temperature for 5 min, the resulting
26
27 mixture was cooled to ‒78 °C over 20 min. At this point, n-BuLi (2.5 M in n-hexane, 8.1 mL,
28
29
30 20.18 mmol, 4.3 equiv.) was added dropwise over 30 min through a syringe to the mixture and
31
32 the reaction was allowed to stir at ‒78 °C for additional 15 min. Next, a solution of lactone 3
33
34 (3.930 g, 9.388 mmol, 2.0 equiv.) in anhydrous THF (5 mL) was added dropwise over 10 min
35
36
37
under the argon atmosphere through a syringe. After the reaction was stirred at ‒78 °C for 2 h, it
38
39 was allowed to warm slowly to 0 to 10 °C. The reaction was quenched by adding 1 M citric acid
40
41 (50 mL) and stirred vigorously for 15 min. Then the layers were separated and the aqueous layer
42
43
was extracted with EtOAc (3 × 30 mL). The organic layers were combined and washed
44
45
46 sequentially with water (1 × 50 mL), saturated aqueous NaHCO3 (1 × 50 mL) and brine (1 × 50
47
48 mL). The organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated
49
50 under reduced pressure. The residue was subjected to silica gel column chromatography (50%
51
52
53 EtOAc in petroleum ether to 100% EtOAc to 10% MeOH in EtOAc) to give 4 as white foam.
54
55
56
57
58
59 12 Environment
60 ACS Paragon Plus
Page 13 of 21 Organic Process Research & Development

1
2
3 Decagram Scale Synthesis of Compound 4 Under the Optimized Reaction Conditions. An
4
5
6 oven-dried three-neck round-bottom flask equipped with a stir bar and an additional funnel was
7
8 charged with 7-bromopyrrolo[2,1-f][1,2,4]triazin-4-amine (2a, 10.0 g, 46.94 mmol, 1.0 equiv.).
9
10 The above flask was evacuated and backfilled with argon for three times. Under the protection of
11
12
13
argon, anhydrous THF (80 mL) was added and the mixture was stirred at room temperature for 5
14
15 min before a solution of 1,2-bis(chlorodimethylsilyl) ethane (5, 11.1 g, 51.63 mmol, 1.1 equiv.)
16
17 in anhydrous tetrahydrofuran (20 mL) was added. The resulting mixture was stirred for 10 min.
18
19
Then diisopropylamine (7.3 mL, 51.63 mmol, 1.1 equiv.) was added. After stirring at room
20
21
22 temperature for 5 min, the resulting mixture was cooled to ‒78 °C over 20 min. At this point, n-
23
24 BuLi (2.5 M in n-hexane, 81 mL, 201.8 mmol, 4.3 equiv.) was added dropwise over 45 min
25
26 through the additional funnel to the mixture. Note: the dropwise addition of n-BuLi is essential
27
28
29 for this exothermic step. The addition time depends on the volume of n-BuLi, during which the
30
31 reaction temperature should be kept below ‒78 °C. In the decagram scale case, a 45 min period
32
33 could guarantee the complete formation of lithium intermediate. After the additional funnel was
34
35
36
rinsed by anhydrous THF, a solution of lactone 3 (39.3 g, 93.88 mmol, 2.0 equiv.) in anhydrous
37
38 THF (50 mL) was added dropwise over 20 min under the argon atmosphere, and the reaction was
39
40 allowed to stir at ‒78 °C for 2 h. Then the cold bath was removed and the mixture was slowly
41
42
warmed to 0 to 10 °C. The reaction was quenched by adding 1 M citric acid (200 mL) and stirred
43
44
45 vigorously for 15 min. Then the layers were separated and the aqueous layer was extracted with
46
47 EtOAc (3 × 200 mL). The organic layers were combined and washed sequentially with water (1
48
49 × 250 mL), saturated aqueous NaHCO3 (1 × 250 mL) and brine (1 × 250 mL). The organic phase
50
51
52 was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure.
53
54 The residue was subjected to silica gel column chromatography (50% EtOAc in petroleum ether
55
56
57
58
59 13 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 14 of 21

1
2
3 to 100% EtOAc to 10% MeOH in EtOAc ) to give the mixture of isomers 4 (19.2 g, 74%) as
4
5
6 white foam. The analytical data were consistent with data previously reported in the literature.11
7
8 1H-NMR (400 MHz, DMSO-d6): δ 8.10 (br s, 2H), 8.02 (s,1H), 7.37 – 7.23 (m, 11H), 7.17 –
9
10 7.12 (m, 3H), 7.03 – 7.01 (m, 2H), 6.98 (d, J = 4.0 Hz, 1H), 5.41 (d, J = 4.0 Hz, 1H), 5.10 (d, J =
11
12
13
8.0 Hz, 1H), 4.61 – 4.56 (m, 2H), 4.54 – 4.47 (m, 4H), 4.08 – 3.99 (m, 1H), 3.98 – 3.95 (m, 1H),
14
15 3.71 (dd, J = 8.0, 4.0 Hz, 1H), 3.50 (dd, J = 8.0, 4.0 Hz, 1H); 13C-NMR (100 MHz, DMSO-d6):
16
17 δ 187.97, 155.86, 148.98, 138.63, 138.45, 138.42, 138.15, 128.63, 128.25, 128.22, 128.16,
18
19
128.15, 127.83, 127.77, 127.73, 127.64, 127.61, 127.55, 127.49, 127.46, 127.42, 127.28, 127.22,
20
21
22 127.11, 127.04, 118.57, 117.52, 103.12, 102.27, 81.89, 80.85, 72.48, 72.31, 71.70, 71.40, 69.37;
23
24 HRMS (ESI): m/z calcd. for C32H32N4NaO5 [M + Na]+ 575.2265; found 575.2260.
25
26
27 Hectogram Scale Synthesis of Compound 4 Under the Optimized Reaction Conditions.
28
29
30 An oven-dried 20 L reactor was charged with 7-bromopyrrolo[2,1-f][1,2,4]triazin-4-amine (2a,
31
32 180.0 g, 0.845 mol, 1.0 equiv.) and anhydrous THF (1.44 L) under an atmosphere of nitrogen at
33
34 20 °C. The mixture was stirred for 10 min before a solution of 1,2-bis(chlorodimethylsilyl)
35
36
37
ethane (5, 200.0 g, 0.929 mol, 1.1 equiv.) in anhydrous tetrahydrofuran (360 mL) was added at
38
39 the same temperature. The resulting mixture was stirred for 15 min. Then diisopropylamine (94.1
40
41 g, 131 mL, 0.929 mol, 1.1 equiv.) was added. After stirring for 10 min, the reaction was cooled
42
43
to ‒85 to ‒78 °C. At this point, n-BuLi (4, 2.5 M in n-hexane, 1.45 L, 3.634 mol, 4.3 equiv.) was
44
45
46 added dropwise over 4 h while maintaining the reaction temperature below ‒78 °C during the
47
48 exothermic addition, and the resulting mixture was allowed to stir at ‒85 to ‒78 °C for 30 min.
49
50 Next, a solution of lactone 3 (707.2 g, 1.69 mol, 2.0 equiv.) in anhydrous THF (900 mL) was
51
52
53 added dropwise over 3 h while maintaining the temperature between ‒85 to ‒78 °C. After the
54
55 reaction was stirred at the same temperature for another 2 h, it was allowed to warm slowly to 0
56
57
58
59 14 Environment
60 ACS Paragon Plus
Page 15 of 21 Organic Process Research & Development

1
2
3 to 10 °C. The reaction was quenched by the addition of 1 M citric acid (3.6 L) and the
4
5
6 temperature was maintained below 25 °C while vigorously stirring for 10 min. Then the layers
7
8 were separated and the aqueous layer was extracted with EtOAc (3 × 3.6 L). The organic layers
9
10 were combined and washed sequentially with water (1 × 3.6 L), saturated aqueous NaHCO3 (1 ×
11
12
13
3.6 L) and brine (1 × 3.6 L). The organic phase was then concentrated in vacuo at 30 °C to
14
15 remove the majority of organic solvents. The residual solvents were removed by adding tert-
16
17 butyl methyl ether (3.6 L) and evaporated at 30 °C under reduced pressure. The mixture was
18
19
finally concentrated in vacuo to a total batch volume of 540 mL, whereupon n-heptane (3600
20
21
22 mL) was added. The resulting mixture was heated to 50 °C and stirred at this temperature for 30
23
24 min. Then it was allowed to cool to 0 °C. The resulting pale yellow solid was collected by
25
26 filtration. The filter cake was rinsed with n-heptane (540 mL) and dried under vacuum at 30 °C
27
28
29 to afford pale yellow solid of product 4 (291.5 g, 62%). Purity: 99.3% (Flow rate: 1.0 mL/min;
30
31 Column: Waters XTERRA MS C18 Column, 4.6 × 150 mm, 3.5 μm; Temperature: 45 °C;
32
33
34
Mobile phase A: water with 0.1% (v/v) trifluoroacetic acid; Mobile phase B: acetonitrile with
35
36 0.1% (v/v) trifluoroacetic acid; Gradient elution: mobile phase A/B = 95:5 (0-5 min) to mobile
37
38 phase A/B = 5:95 (5-32 min) to mobile phase A/B = 95:5 (32-40 min), tR = 19.272 min).
39
40
41
42
43
44
45 ASSOCIATED CONTENT
46
47
48 Supporting Information.
49
50 The Supporting Information is available free of charge at DOI:
51
52
53 NMR spectra of compounds; (PDF)
54
55
56
57
58
59 15 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 16 of 21

1
2
3 AUTHOR INFORMATION
4
5
6 Corresponding Authors
7
8
9 Wu Zhong - National Engineering Research Center for the Emergence Drugs, Beijing
10
11 Institute of Pharmacology and Toxicology, Beijing 100850, China;
12
13
14 Email: zhongwu@bmi.ac.cn
15
16 Yong Qin - Key Laboratory of Drug-Targeting and Drug Delivery System of the Education
17
18
19
Ministry and Sichuan Province, and Sichuan Research Center for Drug Precision Industrial
20
21 Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041,
22
23 Sichuan Province, China;
24
25 Email: yongqin@scu.edu.cn
26
27
28 Authors
29
30
31 Fei Xue - Key Laboratory of Drug-Targeting and Drug Delivery System of the Education
32
33 Ministry and Sichuan Province, and Sichuan Research Center for Drug Precision Industrial
34
35
36 Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041,
37
38 Sichuan Province, China
39
40 Xinbo Zhou - National Engineering Research Center for the Emergence Drugs, Beijing
41
42
43 Institute of Pharmacology and Toxicology, Beijing 100850, China
44
45 Ruijie Zhou - Key Laboratory of Drug-Targeting and Drug Delivery System of the
46
47
48 Education Ministry and Sichuan Province, and Sichuan Research Center for Drug
49
50 Precision Industrial Technology, West China School of Pharmacy, Sichuan University,
51
52 Chengdu 610041, Sichuan Province, China
53
54
55
56
57
58
59 16 Environment
60 ACS Paragon Plus
Page 17 of 21 Organic Process Research & Development

1
2
3
Xiaohan Zhou - Key Laboratory of Drug-Targeting and Drug Delivery System of the
4
5
6 Education Ministry and Sichuan Province, and Sichuan Research Center for Drug
7
8 Precision Industrial Technology, West China School of Pharmacy, Sichuan University,
9
10
11
Chengdu 610041, Sichuan Province, China
12
13 Dian Xiao - National Engineering Research Center for the Emergence Drugs, Beijing
14
15
Institute of Pharmacology and Toxicology, Beijing 100850, China
16
17
18 Xiaowen Guo - Shanghai Syncores Technology Inc. Ltd., Shanghai 201203, China
19
20
Ji Xiang - Shanghai Syncores Technology Inc. Ltd., Shanghai 201203, China
21
22
23 Ke Wang - Chengdu Open Pharmaceutical Co., Ltd., Qionglai 611500, Sichuan Province,
24
25
26
China
27
28 Likai Yang - Chengdu Open Pharmaceutical Co., Ltd., Qionglai 611500, Sichuan Province,
29
30
China
31
32
33 Author Contributions
34
35
36 ‣These authors contributed equally.
37
38
39 Notes
40
41
42 The authors declare no competing financial interest.
43
44
45 ACKNOWLEDGMENT
46
47
48 Wu Zhong thanks National Science and Technology Major Projects for “Major New Drugs
49
50 Innovation and Development” (2018ZX09711003), Yong Qin thanks financial support from
51
52
53 National Natural Science Foundation of China (21921002 and 21991114) and Special Funds for
54
55 Prevention and Control of COVID-19 of Sichuan University, and Likai Yang thanks Chengdu
56
57
58
59 17 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 18 of 21

1
2
3 Municipal Science and Technology Bureau (2020-YF08-00002-GX). We are grateful to Prof.
4
5
6 Chunming Cui (Nankai University) for helpful discussions.
7
8
9
10
11 REFERENCES
12
13
14 (1) Chan, J. F.-W.; Yuan, S.; Kok K.-H.; To, K. K.-W.; Chu, H.; Yang, J.; Xing, F.; BNurs, J.
15
16
Liu.; Yip C. C.-Y.; Poon, R. W.-S.; et al. A familial cluster of pneumonia associated with the
17 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster.
18
19 Lancet 2020, 395, 514‒523.
20
21 (2) Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu,
22
R.; et al. A novel coronavirus from patients with pneumonia in China. N. Engl. 2020, 382,
23
24 727‒733.
25
26 (3) Li, Q.; Guan, X.; Wu, P.; Wang, X.; Zhou, L.; Tong, Y.; Ren, R.; Leung, K. S. M.; Lau, E.
27
28 H. Y.; Wong, J. Y.; et al. Early transmission dynamics in Wuhan, China, of novel coronavirus–
29 infected pneumonia. N. Engl. 2020, 382, 1199‒1207.
30
31 (4) Zhou, P.; Yang. X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li,
32
33 B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat
34
35
origin. Nature 2020, 579, 270‒273.
36 (5) Wu, F.; Zhao, S.; Yu, B.; Chen, Y.-M.; Wang, W.; Song, Z.-G.; Hu, Y.; Tao, Z.-W.; Tian,
37
38 J.-H.; Pei, Y.-Y.; et al. A new coronavirus associated with human respiratory disease in China.
39
40 Nature 2020, 579, 265‒269.
41
(6) Li, G.; Clercq, E. D. Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nat.
42
43 Rev. Drug Dis. 2020, 19, 149‒150.
44
45 (7) Jean, S.-S.; Lee, P.-L.; Hsueh, P.-R. Treatment options for COVID-19: The reality and
46
47 challenges. J. Microbiol., Immunol. Infect. 2020, 53, 436‒443.
48 (8) Savi, C. D.; Hughes, D. L.; Kvaerno, L. Quest for a COVID-19 Cure by Repurposing Small-
49
50 Molecule Drugs: Mechanism of Action, Clinical Development, Synthesis at Scale, and Outlook
51
52 for Supply. Org. Process Res. Dev. 2020, 24, 940‒976.
53
54
55
56
57
58
59 18 Environment
60 ACS Paragon Plus
Page 19 of 21 Organic Process Research & Development

1
2
3 (9) Ko, W.-C.; Rolain, J.-M.; Lee, N.-Y.; Chen, P.-L.; Huang, C.-T.; Lee, P.-I.; Hsueh, P.-R.
4
5 Arguments in favour of remdesivir for treating SARS-CoV-2 infections. Int. J. Antimicrob.
6
7 Agents. 2020, 55, 105933.
8
9
(10) Amirian, E. S.; Levy, J. K. Current knowledge about the antivirals remdesivir (GS-5734)
10 and GS-441524 as therapeutic options for coronaviruses. One Health 2020, 9, 100128.
11
12 (11) Warren, T. K.; Jordan, R.; Lo, M. K.; Ray, A. S.; Mackman, R. L.; Soloveva, V.; Siegel,
13
14 D.; Perron, M.; Bannister, R.; Hui, H. C.; et al. Therapeutic efficacy of the small molecule GS-
15
5734 against Ebola virus in rhesus monkeys. Nature 2016, 531, 381‒385.
16
17 (12) Siegel, D.; Hui, H. C.; Doerffler, E.; Clarke, M. O.; Chun, K.; Zhang, L.; Neville, S.;
18
19 Carra, E.; Lew, W.; Ross, B.; et al. Discovery and synthesis of a phosphoramidate prodrug of a
20
21 pyrrolo[2,1-f ][triazin-4-amino] adenine C-nucleoside (GS-5734) for the treatment of Ebola and
22 emerging viruses. J. Med. Chem. 2017, 60, 1648‒1661.
23
24 (13) Lo, M. K.; Jordan, R.; Arvey, A.; Sudhamsu, J.; Shrivastava-Ranjan, P.; Hotard, A. L.;
25
26 Flint, M.; McMullan, L. K.; Siegel, D.; O. Clarke, M.; et al. GS-5734 and its parent nucleoside
27
28
analog inhibit filo-, Pneumo-, and paramyxoviruses. Sci. Rep. 2017, 7, 43395.
29 (14) Sheahan, T. P.; Sims, A. C.; Graham, R. L.; Menachery, V. D.; Gralinski, L. E.; Case, J.
30
31 B.; Leist, S. R.; Pyrc, K.; Feng, J. Y.; Trantcheva, I.; et al. Broad-spectrum antiviral GS-5734
32
33 inhibits both epidemic and zoonotic coronaviruses. Sci. Transl. Med. 2017, 9, eaal3653.
34
(15) Brown, A. J.; Won, J. J.; Graham, R. L.; Dinnon III, K. H.; Sims, A. C.; Feng, J. Y.;
35
36 Cihlar, T.; Denison, M. R.; Baric, R. S.; Sheahan, T. P. Broad spectrum antiviral remdesivir
37
38 inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA
39
40 dependent RNA polymerase. Antivir. Res. 2019, 169, 104541.
41 (16) Sheahan, T. P.; Sims, A. C.; Leist, S. R.; Schäfer, A.; Won, J.; Brown, A. J.; Montgomery,
42
43 S. A.; Hogg, A.; Babusis, D.; Clarke, M. O.; et al. Comparative therapeutic efficacy of
44
45 remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat.
46
Commun. 2020, 11, 222.
47
48 (17) Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao,
49
50 G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-
51
52 nCoV) in vitro. Cell Res. 2020, 30, 269‒271.
53
54
55
56
57
58
59 19 Environment
60 ACS Paragon Plus
Organic Process Research & Development Page 20 of 21

1
2
3 (18) Holshue, M. L.; DeBolt, C.; Linduist, S.; Lofy, K. H.; Wiesman, J.; Bruce, H.; Spitters, C.;
4
5 Ericson, K.; Wilkerson, S.; Tural, A.; et al. First case of 2019 novel coronavirus in the United
6
7 States. N. Engl. J. Med. 2020, 382, 929‒936.
8
9
(19) www. clinicaltrials.gov
10 (20) Grein, J.; Ohmagari, N.; Shin, D.; Diaz, G.; Asperges, E.; Castagna, A.; Feldt, T.;
11
12 Green, G.; Green, M. L.; Lescure, F.-X. Compassionate Use of Remdesivir for Patients with
13
14 Severe Covid-19. N. Engl. J. Med. 2020, 382, 2327‒2336.
15
(21) Wang, Y.; et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind,
16
17 placebo-controlled, multicentre trial. Lancet 2020, 395, 1569‒1578.
18
19 (22) http://www.gilead.com/news-and-press/press-room/press-releases/2020/5/gileads-
20
21 investigational-antiviral-remdesivir-receives-us-food-and-drug-administration-emergency-use-
22 authorization-for-the-treatment-of-covid19
23
24 (23) Jarvis, L. M. Scaling up remdesivir amid the coronavirus crisis. Chem. Eng. News
25
26 Published on April 20, 2020, https://cen.acs.org/biological-chemistry/infectious-disease/Scaling-
27
28
remdesivir-amid-coronavirus-crisis/98/web/2020/04
29 (24) Butler, T.; Cho, A.; Kim, C. U.; Saunders, O. L.; Zhang, L. 1′-Substituted carba-nucleoside
30
31 analogs for antiviral treatment. Patent US2010/0021425, 2010.
32
33 (25) Butler, T.; Cho, A.; Graetz, B. R.; Kim, C. U.; Metobo, S. E.; Saunders, O. L.; Waltman,
34
A. W.; Xu, J.; Zhang, L. Processes and intermediates for the preparation of 1′-substituted carba-
35
36 nucleoside analogues. US Patent 10023600B2, 2018.
37
38 (26) Metobo, S. E.; Xu, J.; Saunders, O. L.; Butler, T.; Aktoudianakis, E.; Cho, A.; Kim, C. U.
39
40 Practical synthesis of 1′-substituted tubercidin C-nucleoside analogs. Tetrahedron Lett. 2012, 53,
41 484‒486.
42
43 (27) Cho, A.; Saunders, O. L.; Butler, T.; Zhang, L.; Xu, J.; Vela, J. E.; Feng, J. Y.; Ray, A. S.;
44
45 Kim, C. U. Synthesis and antiviral activity of a series of 1′-substituted 4-aza-7,9-
46
dideazaadenosine C-nucleosides. Bioorg. Med. Chem. Lett. 2012, 22, 2705–2707
47
48 (28) Djuric, S.; Venit, J.; Magnus, P. Silicon in synthesis: stabase adducts - a new primary
49
50 amine protecting group: alkylation of ethyl glycinate. Tetrahedron Lett. 1981, 22, 1787‒1790.
51
52 (29) Vieira, T.; Stevens, A.; Chtchemelinine, A.; Gao, D.; Badalov, P.; Heumann, L.
53 Development of a Large-Scale Cyanation Process Using Continuous Flow Chemistry En Route
54
55 to the Synthesis of Remdesivir. Org. Process Res. Dev. 2020, DOI: 10.1021/acs.oprd.0c00172.
56
57
58
59 20 Environment
60 ACS Paragon Plus
Page 21 of 21 Organic Process Research & Development

1
2
3 (30) The disilyl substituted quaternary amine salt 8 proved to be very unstable. A related cationic
4
5 species was confirmed by X-ray crystallography when complexed with a hexameric cage like
6
7 anionic moiety, see: Lorber, C.; Vendier, L. Tight encapsulation of a “naked” chloride in an
8
9
imidotitanium hexanuclear host. Inorg. Chem. 2013, 52, 4756−4758.
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59 21 Environment
60 ACS Paragon Plus

You might also like