Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

ORGANIC CHEMISTRY LABWORK

Dang Nhat Minh


Pham Hai Ha
FAS Department
University of Science and Technology of Hanoi (USTH)

A REVIEW ON REMDESIVIR’S
SYNTHESIS, A LONG-
July 27 th

2021
ESTABLISHED AND PROMISING
ANTIVIRAL AGENT

minhdn.bi10-114@st.usth.edu.vn

0705199988
INTRODUCTION
Remdesivir (trade name: Veklury (1), development code name: GS-5734 (2)), the structure of which is shown
below, (3) is an antiviral drug that was introduced by an American biopharmaceutical company named Gilead
Sciences, Inc. a few years ago (4) . This is a “prodrug” of a nucleotide or a “pro-tide”, which means that it can diffuse
into the cell and change into an active metabolite with the help of an enzyme called nucleoside-phosphate kinases.
(5)
This active form can inhibit viral RNA-dependent RNA polymerase (RdRP) and decrease the production of the
viral RNA. This can eventually lead to the death of the virus.

Figure 1: The structure of Remdesivir

Firstly, let’s ask ourselves the question: When was this drug invented? It all started when Gilead Sciences
began its development in 2009 (6), as a result of a collaboration between Gilead, the U.S. Centers for Disease Control
and Prevention (CDC), and the U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID). The
drug was created to treat hepatitis C but then was used as a potential treatment for other illnesses. (7)
So, what is its main purpose exactly? As already mentioned above, Remdesivir is an inhibitor to RdRP,
making it able to treat infections that are caused by RNA viruses, for example, filoviruses, pneumoviruses,
paramyxoviruses, and coronaviruses. (8) Let’s take a closer look at the drug’s usage with this list of illnesses that it
aims to cure.

• Hepatitis C & respiratory syncytial virus (RSV)


These diseases were the original targets of Remdesivir at the very beginning stages of its development in 2009,
according to Gilead Sciences, Inc. Unfortunately, the results of the trials were not very positive, and the research
came to a dead end. (6) This may be the reason why there is not much known publicly about this specific phase.

• Ebola virus
After failing to become the cure to hepatitis C, the drug got recognized once again in the fight against the Ebola
epidemic in Congo in 2014. (6) Indeed, the preclinical results showed that Remdesivir possessed “promising
antiviral activity” (9) and high potency (10) against the Ebola virus. Moreover, the drug showed unprecedentedly
good results in infected rhesus monkeys at a high dosage. Thus, the researchers suggested that further
investigation should be carried out for this potential antidote. (11)
The drug was then tested on a limited number of people infected with the disease in 2016-2017 with quite
satisfactory outcomes. (12) (13) Following this success, it was put on more clinical trials in 2019, competing
against other types of medications like “ZMapp (a triple monoclonal antibody), MAb114 (a single human
monoclonal antibody derived from an Ebola survivor), and REGN-EB3 (a mixture of three human
immunoglobulin G1 [IgG1] monoclonal antibodies)”. (14) Unfortunately, this time, Remdesivir proved to be the
worst option as its mortality rate was the highest compared to the other examined antibodies’. Hence, the hope
of using this medication as the primary cure for the Ebola virus disease was lost.

• Feline infectious peritonitis


This disease in cats is caused by a coronavirus, which makes it a potential target for Remdesivir. Certainly, a
study (15) was carried out in 2019 to evaluate its medicinal effects on this particular infection. The results
demonstrated that the drug’s main metabolite in non-primates, GS-441524, was very successful in treating the
ailment, keeping 24 out of 25 cats healthy at the end of the trial. In spite of this promising outcome, the use of
this drug in the treatment of the disease has sadly not been approved by the U.S Food and Drug Administration
(FDA). (16) However, as the metabolite has similar negative impacts on the coronavirus SARS-CoV-2, scientists
have since looked at it as a possible treatment for COVID-19. (17)

• COVID-19
Remdesivir had been known to be able to treat coronaviruses even before the COVID-19 pandemic in December
2019. Unsurprisingly, it was thus one of the first few drugs reported to be effective in the fight against the
disease in early 2020. (18) Moreover, as it had a long-established history in the medical field and the number of
people infected by SARS-CoV-2 kept increasing significantly, Remdesivir received a lot of interest from the
public as the primary potential remedy.
As a result, it was put on multiple clinical trials by the FDA in mid-2020, including 3 most important ones (Trial
1/ NCT04280705, trial 2/ NCT04292899, and trial 3/ NCT04292730). (19) These were conducted on 2043
hospitalized patients with mild to severe symptoms of COVID-19, 226 sites in 17 countries, and offered quite
satisfactory results. Indeed, according to the first trial, “the median time to recovery was 10 days in the
VEKLURY group compared to 15 days in the placebo group (recovery rate ratio 1.29 [95% CI 1.12 to 1.49], p
< 0.001)”. Moreover, trial 3 also showed there was a higher odd of recovery when the patient was treated with
the drug (odds ratio, 1.65; [95% CI, 1.09 to 2.48], p = 0.017).
Based on these promising results, Remdesivir was approved by the FDA in October 2020 with available
prescribing information such as side effects (nausea, pounding heartbeats, severe headache, fever, itching,
sweating, light-headedness…), dosage (200 mg IV as the loading dose, 100 mg IV as the maintenance dose)
and drug interactions (chloroquine or hydroxychloroquine can interfere with the treatment). (20) This made the
drug one of the very first officially recognized medications against the infectious virus. Since then, Remdesivir
has been approved to treat COVID-19 in an emergency in about 50 countries (Australia, Canada, Iran, Japan,
the European Union, Mexico, USA…). (21)
However, other studies suggested that the drug might not be as effective as one might have thought. For
example, this research in April 2020 concluded that Remdesivir “did not significantly improve the time to
clinical improvement, mortality, or time to clearance of virus in patients with serious COVID-19”. (22) Moreover,
another research even challenged the findings of the FDA’s trials by stating that some of the important criteria
for the drug’s efficacy were “unclear”. (23) And finally, the mid-October 2020 interim WHO Solidarity Trial
conducted on more than 10000 adults, a much larger population than that of the FDA’s trials, showed that
Remdesivir "had little or no effect on hospitalized patients with Covid-19, as indicated by overall mortality,
initiation of ventilation, and duration of hospital stay." (24) As a result, WHO eventually advised against the
usage of the drug in the treatment of Covid-19 in November 2020 (25). Hence, the chance of exploiting the pro-
tide to fight SARS-CoV-2 unfortunately diminished.

Figure 2: Remdesivir as a potential treatment for Covid-19


https://www.elsevier.com/about/press-releases/research-and-journals/remdesivir-induced-dangerously-low-heart-rate-in-covid-19-patient
SYNTHESIS
As Remdesivir has gained so much attention from scientists in the last year or so, it should not come as a
surprise that many studies have been carried out to find the most efficient and economical way to synthesize the
drug. Additionally, researchers are even trying to scale up the substance’s production to an industrial level so that
its high demands in the Covid-19 pandemic can be met. (26) In this part of the report, we will report some well-
established ways to synthesize this drug on a laboratory scale and also some of its potential industrial syntheses.

PROVIDED RESOURCES
Let’s start with the 3 synthesizing routes in the review (3) provided by our professor. After that, we will give our
opinions on which route is the simplest, has the highest yield, and is suitable for commercial production among the
three.

Figure 3: The provided review on the synthesis of some drug molecules, including Remdesivir
With that being said, let’s take a look at the first synthetic route.

Figure 4: Route 1 to synthesize Remdesivir

The figure above illustrates the first-generation synthesis of Remdesivir, which was proposed by Richard L.
Mackman, a scientist at Gilead Sciences, Inc. The synthesis started with the glycosylation reaction between the
lactone (2) and the bromo pyrrolotriazine nucleus (1a). This process was facilitated by the N-silyl protection of (1a)
with 1,2-bis(chlorodimethylsilyl)ethane or TMSCl generating a mixture of 1' isomers of nucleoside (3) after
lithium-halogen exchange of (2) with BuLi in tetrahydrofuran (THF). Next, the cyanation step: the hydroxyl group
on (3) was replaced by a cyano group using trimethylsilyl cyanide (TMSCN) in the presence of BF3.Et2O to produce
(4). After that, the desired (R) isomer of (4) was obtained after chromatographic purification. Then, we cleaved the
benzyl protecting groups using BCl3 in CH2Cl2 to get compound (5). The product in turn reacted with the
diastereomeric mixture (7) of phosphoramidyl chloride to synthesize (8a), a 1:1 mixture of Rp & Sp isomers.
Finally, Remdesivir (8b) was acquired through chiral HPLC (high-performance liquid chromatography).
The overall yield of this convergent synthesis is calculated by multiplying all the yields of the steps forming
the longest linear reaction sequence. (27) Hence, the yield for route 1 is given in two ways:
0.60 × 0.58 × 0.74 × 0.21 × 100% = 5.41% (60% yield for glycosylation step)
0.25 × 0.58 × 0.74 × 0.21 × 100% = 2.25% (25% yield for glycosylation step)
Either way, the yield of this process is very low, making it unsuitable for large-scale synthesis. This implies
that this first-generation synthesis still has a lot of flaws and disadvantages that can be improved. Some of these
imperfections (28) include:

• The frequent use of cryogenic temperatures, which made the reaction condition so much more difficult,
• The inconsistent yields,
• The dependency on the rate of addition of n-BuLi,
• The poor stereoselectivity,
• The need for chiral HPLC,
• The unselective reaction between (5) and (7) (all of the hydroxy groups on (5) can react with (7)),
• The use of (7), a mixture of isomers, as a reactant in the last reaction, which decreased the selectivity of
the conversion.

Now that we have examined carefully route 1, let’s move on to route 2 (the second-generation synthesis of
Remdesivir) to see whether there are any improvements.

Figure 5: Route 2 to synthesize Remdesivir


Even though the ideas (the glycosylation, the cyanation, and the combination with the phosphoramidyl moiety)
in route 2 are quite similar to route 1, this second-generation synthesis does involve more steps and is a more
complicated process. Now, let’s look into some of the differences in this new procedure and the changes that they
brought. (28)

• In the first glycosylation step, the inconsistent BuLi was replaced by the accelerating Turbo Grignard
reagent i-PrMgCl·LiCl. Moreover, PhMgCl and TMSCl could do a much better job at protecting the amino
group on (1b), allowing the reaction to move forward more quickly. Additionally, the iodo derivative of the
triazine (1b) made the metal-halogen exchange happen more easily, compared to its bromo counterpart in
route 1. And lastly, this step could now happen at -20 °C, a milder condition than in the first synthesis,
making the method more industrial-friendly.
• The cyanation step showed a much higher efficiency and was much more selective with an anomeric ratio
of 95:5 (28) favoring the desired β-anomer. These improvements, thanks to the use of TfOH (28), eliminated
the need for chiral HPLC separation and increased the overall yield significantly.
• The usage of the more stable p-nitrophenolate phosphoramide (10), instead of its chloridate counterpart in
route 1, generated a single Sp isomer (11a) after solvent crystallization. This undoubtedly increased the
stereoselective of the final product.
• In the combination reaction with the phosphoramide, compound (9) was fully protected with acetonide,
leaving the targeted primary hydroxy group free to react. This is why the yield of this step is so much higher
than in the first process. Moreover, this step could now occur at 50°C, which is a surprisingly milder
condition, compared to all the cryogenic temperatures we have seen so far. This makes route 2 somewhat
simpler than route 1.

Thanks to these improvements, the efficiency of the second route is expected to be higher than that of the first
one. Indeed, its yield is given by:
0.40 × 0.85 × 0.86 × 0.90 × 0.70 × 0.69 × 100% = 12.71%
All in all, this process proved to be a much better way to synthesize Remdesivir in many ways: scalability, yield,
and even stereoselectivity. However, it is not without flaws. Certainly, some of its drawbacks are: (29)
• The addition of the nucleophilic p-nitrophenol to stabilize the phosphoramidoyl chloridate,
• The lengthy steps of cleaving the benzyl protection groups (BnO) and re-protecting the OH groups with
acetonide, which decreased the overall yield significantly,
• The crystallizing purification of the 1:1 diastereomeric mixture (10), which resulted in a great loss (61%)
of materials,
• The last separated acetonide cleaving step that decreased the overall yield.

Now, let us take a look at the last method, the scheme of which is shown on the next page (figure 6), introduced
by Gilead Sciences. As we can see, route 3 is very similar to route 2. However, there are some changes in the
procedure that will be discussed below.
• Firstly, LaCl3.LiCl was introduced in the glycosylation step. The purpose of this addition was to decrease
the basicity of the Grignard reagent i-PrMgCl and in turn prevent the chances of side reactions when the
1,2-addition of Grignard reagent to ketone occurs. Besides, it was possible to conduct this step at 0°C and
-15°C, which are milder temperatures compared to those in route 2. For these reasons, we expect the yield
of this step to be improved.
• Secondly, the tert-Butyldimethylsilyl (TBDMS) group was used to protect the hydroxy groups on the lactone
and was only cleaved in the end. This is a big improvement from the lengthy second synthetic process.
• Thirdly, TFA was used in the cyanation instead of TfOH. This does not change the efficiency of the reaction
substantially as TFA still offers a very pure product and high diastereoselectivity, as one study suggested.
(30)
Additionally, this step can now be conducted at -30 °C, a milder condition than in route 2.
• Compound (15) could combine with either of the two derivatives of the phosphoramide moiety, “p-
nitrophenol (11a) in presence of MgCl2 and (i-Pr)2NEt in THF or with pentafluorophenyl protected
derivative (11b) in the presence of tert-butylmagnesium chloride (tBuMgCl) in THF”. This certainly gives
the process more flexibility and makes it simpler to some extent. It seems like the reaction with (11a) can
happen much more easily as the required temperature is much higher and not as harsh. However, the
reaction with (11b) could have a higher yield as its low temperature can prevent side reactions from
happening.

Figure 6: Route 3 to synthesize Remdesivir

Unfortunately, we could not find any more information about this specific process, including the rate of each
step or of the overall synthesis. However, we predict that the rate of route 3 should be higher than or at least, very
close to that of route 2. The reason for this is that route 3 is most likely an “improved version of route 2”, with the
addition of reagents (LaCl3.LiCl) or a change in hydroxy protection group (TBDMS) to enhance the reactions’
conditions and yields.
Now that we have examined closely each of these 3 routes, we can do some comparisons among them. Which
one is the simplest one, or the easiest to carry out? This could mean involving the least steps or requiring the mildest
conditions. Which one produces the highest amount of product? And finally, which one is suitable for mass
production to meet commercial demands? These questions can be answered more easily with the help of the figure
below. (31)
Figure 7: The summary of the three Remdesivir synthetic routes

So, which route is the simplest? As we discussed above, even though route 2 has improved so much from
route 1, it is undoubtedly a very lengthy and uneconomical process. That is why it should be the first one to be
eliminated here. As for the other two, route 1 and route 3 have a similar number of steps (route 3 has 1 fewer step).
However, the first route requires a very low temperature (-78 °C) for many of the steps and more importantly, the
need for chiral HPLC. The latter is the thing that sets route 1 apart from the other two and makes it become so much
more complicated. Indeed, chiral HPLC separation has been considered as one of the most complicated types of
separation techniques as the targeted stereoisomers have identical physical and chemical properties. (32) Moreover,
this method is also known for its unpredictability. Indeed, as stereoisomers interact very differently with even
slightly different chiral stationary phases (CSP), leading to a change in retention time and efficiency of the process,
there is no best universal choice for a separation column. That is why it is advisable to have chiral screening
conducted by experts beforehand in order to find the best column for a specific separation. (33) On the other hand,
route 3 is able to remove the need to use such a difficult technique and offer a much milder reaction condition. For
these reasons, we decided to choose route 3 as the simplest one.
Next question, which route has the highest yield? As we found out earlier, the yield of route 2 (12.71%) is
more than double that of route 1 (5.41%). Hence, route 1 is the first one to be ruled out. Moving on to routes 2 and
3, we can see that the latter retains the former’s essence very well and has even made some noticeable enhancements
to simplify the procedure and increase the synthesis’ effectiveness. Thus, although the exact yield of the third
process is not found, it is only reasonable to expect route 3 to be the one that is the most efficient.
And lastly, which route is suitable for industrial production? First of all, let’s look at route 1. As discussed
above, this method is inconsistent due to the usage of n-BuLi, employs very low temperatures too frequently, has
suboptimal stereoselectivity, generates too little product, and requires chiral HPLC separation. All of these reasons
are more than enough to justify that this particular method cannot be scaled up to an industrial level of production.
Consequently, it is once again the first one to be crossed out. Let’s move on to the other two methods. Both of
these routes offer much higher yields, improves the stereoselectivity considerably, and thus eliminates the need for
chiral separation. All of these certainly make them better candidates for mass production. However, as clearly stated
above, route 3 is “the improved version” of route 2 as it simplifies the process and presents an overall better way to
synthesize Remdesivir. That is why once again, we suspect that route 3 would be the most suitable for an
industrial synthesis among the three methods reported in the provided,
To sum up everything that has been stated so far, route 3 seems to be the superior method to the first two, in all
of the criteria in question: yield, simplicity, and scalability. This does not seem very surprising, considering that it
is the most recent process out of the three and hence, gets all the improvements from its predecessors. Nevertheless,
this method still employs chromatography to purify its intermediate (compound (15)), leading to a great loss of
materials and a decrease in the overall yield. Hence, it is still not the perfect way to produce the drug, especially on
an industrial level.

FURTHER READINGS
Now that the pandemic has gone on for over a year and shown no signs of slowing down, more methods need
to be developed to better accommodate the worldwide need for Remdesivir. As a result, researchers have come up
with different ways to synthesize the drug in a more economical way and on a much bigger scale. Thus, in this
section, we will report some advances made to the drug’s production since 2020.

1. Practical Remdesivir Synthesis through One-Pot Organocatalyzed Asymmetric (S)-P-


Phosphoramidation (29)
Gannedi et al. reported a one-pot synthesis of Remdesivir with the help of a bicyclic imidazole catalyst. This
method was inspired by the fact that imidazole derivatives can help greatly with stereoselectivity when it comes to
phosphoramidation, which can be observed in the mechanism of glucose 6-phosphatase catalyzing the hydrolysis
of glucose 6-phosphate. Moreover, studies (34) (35) have also shown that chiral bicycloimidazole-based catalysts
could generate high levels of stereoselectivity at phosphorus. And unsurprisingly, the new proposed catalyzed
reaction in this particular research is significantly more selective, favors the desired (S)-isomer, and eliminates the
need for p-nitrophenol as well as chiral separation. Moreover, the researchers were also able to remove the
separated cleaving step of the final product and hence, increase the overall efficiency. As a result, this novel method
can make the process much simpler and more efficient. The principal reaction is shown in the figure below.

Figure 8: Scheme of the Remdesivir one-pot synthesis


As we can see in figure 8, the researchers only added the catalyst, which is denoted 29 in figure 9, in the
combination stage of the two moieties of the drug. The steps to synthesize each moiety are left unchanged from the
procedures developed by Gilead. Next, we will take a closer look at this complete innovative method, step by step.

Figure 9: Synthesis of Remdesivir through one-pot organocatalyzed asymmetric (S)-P-Phosphoramidation

Firstly, the synthetic procedure suggested in route 1 was applied to prepare the phosphoramidoyl moiety.
Indeed, they stopped at the chlorodate derivative and never added the p-nitrophenol so as to avoid the low-yield
recrystallization step (the conversion from (10) to (11a) in route 2). Even though the chlorodate compound was
unstable and existed as a 1:1 isomeric mixture, it could still react quite well with the other moiety in the final
reaction thanks to the novel organo-catalyst.
Secondly, the cyano moiety was synthesized according to route 2’s procedure. In addition, the yields of the
steps in this part would count to the overall yield as they formed the longest reaction sequence in the synthesis.
Thirdly, the two moieties reacted with each other, giving the desired product with the help of the organic
catalyst, the structure of which is shown below. One important thing to point out is that the cleavage of acetonide
with p-TSA in MeOH and the recrystallization of the final product were both done right after the combination
reaction, in the same system. This could greatly reduce the loss of materials and increase the efficiency of the whole
synthesis. Certainly, the yield of this step is 73% with the diastereomeric ratio (d.r.) of 99.4/0.6 favoring our desired
Sp isomer on a 1-g scale. Moreover, the catalyst could also be recovered at the rate of 82% and used again in
another synthesis with similar efficacy, which was a truly astonishing improvement to reduce the cost of the process.
Now, let’s try calculating the yield of this whole process. Let’s assume that all the yields in figure 9 are true for
1-g scale production, the yield of this catalyzed synthesis would be given by:
0.40 × 0.85 × 0.86 × 0.90 × 0.73 × 100% = 19.21%
This is unsurprisingly higher than that of route 2 and is probably even higher than that of route 3 as well.
Furthermore, when conducted on a 10-g scale, the one-pot reaction showed very consistent results with the 1-g
scale the yield of product and catalyst recovery rate of 70% (99.3/0.7 d.r.) and 83%, respectively. This proves that
this method can be scaled up without too much problem and show real potential for mass production, even though
further research needs to be done to confirm this.
However, this method still presents a few drawbacks that need to be considered carefully if it is to be applied
to industrial production.

• Firstly, the reaction to prepare the phosphoramidoyl chlorodate moiety is not very effective, leaving a lot
of chemicals to go to waste.
• Secondly, the synthesis involves the additional preparation of the organic catalyst, which means that it
requires more materials and certainly adds more to an already expensive synthetic process.
• Thirdly, it still cannot get rid of the chromatographic purification during the cyanation step, which is one
of the biggest flaws of the three reported routes in the review.
• Finally, it still uses the benzyl group to protect the OH groups on the cyano moiety. This is exactly the
disadvantage that route 2 has, as discussed above.

(36)
2. Improvement of the C-glycosylation Step for the Synthesis of Remdesivir
Xue et al. came up with a process to improve the efficiency of the low-yield C-glycosylation reaction in the
Remdesivir synthesis, hoping that it would be applied to a larger production of the drug. This innovative method
involves the addition of a secondary amine as a catalyst, which is also what sets it apart from other glycosylation
steps of the 3 routes in the review. The reaction of this innovative method is shown in figure 10 below.

Figure 10: The amine-catalyzed C-glycosylation step


Next, figure 11 shows us all components and the mechanism of the reaction. First, we have two substrates: the
bromo pyrrolotriazine nucleus (2a) and the lactone (3). Next, 1,1,4,4-tetramethyl-1,4-dichloro disilethylene (5) is
introduced to protect the free amine group on (2a), facilitating the reaction. Moreover, n-BuLi is added for the
lithium-halogen exchange with (2a) in THF. The interesting thing to point out here, as discussed above, is the
employment of the amine. According to the researchers, this substance is used for two reasons. To start with, it
could activate the disilane (5) to form the disilyl substituted cyclic quaternary amine salt (8), which can in turn
promote the protection of the amine group on (2). Furthermore,the amine catalyst could stabilize the product of (7)
and n-BuLi by producing the intermediate (9), which would facilitate the reaction with lactone (3) to yield the
desired final product (4).
Figure 11: The mechanism of the catalyzed glycosylation step

Moreover, the scientists also experimented with different secondary amines to see whether that would lead to a
change in the reaction efficiency. As a result, they found out that diisopropylamine (i-PrNH2) gave the best yield of
74% on a 1-gram scale. Consequently, the reaction was scaled up to a 10-gram scale with i-PrNH2 with a consistent
yield of 75%. These are the highest known efficiency of this step, according to the authors of the study. In addition,
when it came to a 180-gram scale, the reaction was only 62% effective, which was still a very good yield compared
to the ones in the 3 synthetic routes introduced by Gilead. This proves that the method has a lot of potentials for
industrial production.
However, this process still possesses some disadvantages of its predecessors that have not been improved yet.

• Firstly, the use of the bromide substrate seems somewhat unwise as the iodide has proved to be more
efficient in the lithium-halogen exchange process,
• Secondly, the reaction might be affected by the addition rate of n-BuLi,
• Thirdly, it still uses the benzyl group to protect OH groups, the same drawback that the method of Gannedi
et al. has, as discussed above,
• And finally, it is dependent on a very low temperature (-78 °C).

Figure 12: The diisopropylamine-catalyzed C-glycosylation step


Now, let’s try finding the highest efficiency possible of the whole synthesis if this step is to be applied. This can
be done if we envision a synthetic process, illustrated in figure 13, on a 1-gram scale that involves different methods
that were discussed in this report. First of all, the C-glycosylation step would apply the method of Xue et al. with
the diisopropylamine catalyst (figure 12). This stage should have a yield of 74%. Next, the product will go through
the procedure of route 2 to yield the cyano moiety. This stage includes 3 steps, after excluding the first glycosylation
reaction, with yields of 85%, 86%, and 90% respectively. And lastly, the cyano moiety will react with the
phosphoramidoyl moiety in the presence of a bicyclic imidazole catalyst, which is the method introduced by
Gannedi et al. The efficiency of this should be near 73%. Furthermore, the conditions and reagent equivalents of
each reaction will be the same as reported in the studies. As a result, the overall yield of this synthesis would be
given by:
0.74 × 0.85 × 0.86 × 0.90 × 0.73 × 100% = 35.55%
This is the highest efficiency introduced in this report. However, this is only a theoretical yield of a suggested
small-scale synthetic route. The real value would likely be lower, although we do not expect it to differ too much.
However, this is not the simplest way in this report to synthesize the drug, as it involves the production of the
organic catalyst and the use of the benzyl group to protect OH groups on the lactone substrate.
All in all, thanks to the improvements made to different steps in the complete synthesis, hopefully, one day we
can get a synthetic route that offers an optimal condition for Remdesivir production and is efficient enough to be
applied to an industrial scale.

Figure 13: Improved synthesis of Remdesivir combining different methods


3. Development of a Large-Scale Cyanation Process Using Continuous Flow Chemistry En Route to the
Synthesis of Remdesivir (30)
Vieira et al. has come up with a cyanation process in the Remdesivir synthetic route that can be applied to a
mass-production scale with the help of continuous flow chemistry. This particular technology offers so many
advantages compared to traditional methods that can help us produce hundreds of kilograms of chemicals in a short
amount of time. These benefits will be discussed in detail below but first, let’s take a look at the targeted cyanation
step in figure 11. We have the product (1) of the C-glycosylation step, the desired isomer (2), and the by-product
(3).

Figure 11: Cyanation process


As shown above, before going into this cyanation step, we need to synthesize compound (1) first. For this, Vieira
et al. applied a procedure, the scheme of which is shown in figure 12, that is very similar to route 3. Indeed, TMSCl,
PhMgCl, and i-PrMgCl were still used. However, LaCl3.LiCl was replaced with NdCl3 and n-Bu4NCl, which would
not bring too much of a difference since both La and Nd are lanthanides and they share similar chemical
characteristics. Furthermore, the yield of this method was recorded to be 69% on a 280-kg scale, which is a very
impressive value for such a big production. This also proved that the researchers were able to produce enough
compound (1) to fuel a large-scale cyanation process.

Figure 12: Preparation of compound (1)


Next, let’s take a closer look at the continuous flow technology in figure 13 as we dissect some of its advantages.
Firstly, a fewer amount of hazardous cyanide will be produced after the cyanation as the reaction steam will be
quenched immediately in KOH after exiting the reactor. This could help decrease risks and create a safer working
environment, which is very important for industrial synthesis. Secondly, this method allows facilities to mix and
cool the reagents at the same time. This is quite an improvement to traditional methods where new reagents must
be cooled before being added to an already cold reactor. Moreover, this allows better temperature control as there
is no significant change in temperature in the reactor throughout the whole process. All in all, these benefits fit
perfectly in the mass-production-for-commercial-consumptions scenario and can help scale up the synthesis
significantly.
Figure 13: General scheme of continuous flow chemistry

After establishing basic conditions for the reaction vessel, the researchers would conduct experiments on
different scales (from gram to kilogram and finally to plant-manufacturing) and make necessary changes to the
system itself in order to meet the criteria for industrial synthesis. Some of these adjustments are as follows:

• The reactor needs to be made with stainless steel (SS). This durable material can help the reaction go more
smoothly and minimize the loss of containment. Moreover, this vessel also needs to be tested several times
under high pressure to make sure that it is ready for the real process,
• TFA and TMSOTf will now be added and cooled at the same time after the pre-cooling of compound 1,
• The temperature is adjusted to -30 °C and the precooling loop of TFA/TMSOTf was removed to prevent
TFA from crystallizing,
• The reactor has a tube-in-shell form to allow faster cooling and heat transfer,
• Three diaphragm pumps with back-pressure are added as feed lines to achieve the desired flow rates of the
substrates.
After a lot of trials and errors, the researchers were able to construct the optimally designed reactor, the scheme
of which is shown below, for production on the plant-manufacturing scale. This 2.5-L vessel could process up to
almost 2 kg of compound (1) per hour, making this method arguably the most productive synthetic route.

Figure 14: Plant-manufacturing-scale continuous flow reactor


They also found the appropriate reaction time of each loop (30 seconds for loop 1 and 2 minutes for loop 2) as
well as the reagent equivalents (Compound (1)/TFA/TMSOTf/TMSCN = 1/1/6/6) in order to have the best outcome.
Indeed, the final results of this process (shown below) are very impressive. For example, the reaction yield is quite
high (78%) with very high product purity (above 99%). Also, it is able to generate hundreds of kilograms of
products in a span of a few days. For all of these reasons, this method might very well be the best one for
industrial production.

Figure 15: Results of the continuous flow cyanation step on the plant-manufacturing scale
However, we need to remember that in this study, only the glycosylation and the cyanation steps were scaled
up. These are only a small part of the long and costly synthesis of Remdesivir. As a result, if we want to have a
complete optimal industrial synthetic route, other steps must be developed as well. For example, the catalyzed-
phosphoramidation step proposed by Gannedi et al., which has shown potentials for larger production, should be
modified and scaled up to a larger scale. All things considered, it seems like there is still so much left to do until
the day we can mass-produce Remdesivir in an economical and environment-friendly way.

CONCLUSION
In the light of the worldwide Covid-19 pandemic that started in 2020, Remdesivir, a long-established antiviral
drug, has gotten so much attention from the public as a potential cure for the infectious disease. This resulted in a
surge of researches and studies trying to find the optimal way to mass-produce the drug for commercial uses. Indeed,
Gilead Sciences, the enterprise that introduced Remdesivir in 2009, has successively come up with 3 different
synthetic routes, with the next one being the improved version of the one that came before. Furthermore, other
scientists have also joined in and tried to enhance the synthesis based on Gilead’s procedure. For example, one
study suggested the use of an organic catalyst to enhance the phosphoramidation’s efficiency in the hope that the
method can be applied to a larger scale. Likewise, another paper recommended using diisopropylamine to facilitate
the C-glycosylation step. This technique could provide very consistent yields when the production was scaled up.
And finally, the continuous flow technology was studied and applied to the drug’s cyanation step on a plant-
manufacturing production level. A suitable reactor was successfully constructed and able to produce more than 200
kg of the desired product. However, a lot more work still needs to be done so that we can have a complete optimal
synthesis of Remdesivir on an industrial scale.
REFERENCES

1. Gilead Announces Approval of Veklury® (remdesivir) in Japan for Patients With Severe COVID-19.
www.businesswire.com. Published May 7, 2020. Accessed August 1, 2021.
https://www.businesswire.com/news/home/20200507005738/en/

2. Pipeline | Gilead. www.gilead.com. Published April 29, 2021. Accessed August 1, 2021.
https://www.gilead.com/science-and-medicine/pipeline

3. Arora G, Shrivastava R, Kumar P, et al. Recent advances made in the synthesis of small drug molecules for
clinical applications: An insight. Current Research in Green and Sustainable Chemistry. 2021;4(100097):
doi:10.1016/j.crgsc.2021.100097

4. Amirian ES, Levy JK. Current knowledge about the antivirals remdesivir (GS-5734) and GS-441524 as
therapeutic options for coronaviruses. One Health. 2020;9(100128): doi:10.1016/j.onehlt.2020.100128

5. Wikipedia Contributors. Nucleoside-phosphate kinase. Wikipedia. Published October 8, 2017. Accessed


August 1, 2021. https://en.wikipedia.org/wiki/Nucleoside-phosphate_kinase

6. Stephens B. Opinion | The Story of Remdesivir. The New York Times.


https://www.nytimes.com/2020/04/17/opinion/remdesivir-coronavirus.html. Published April 18, 2020.
Accessed August 1, 2021.

7. Eastman RT, Roth JS, Brimacombe KR, et al. Remdesivir: A Review of Its Discovery and Development
Leading to Emergency Use
Authorization for Treatment of COVID-19. ACS Central Science. 2020;6(5):672-683.
doi:10.1021/acscentsci.0c00489

8. Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and
Paramyxoviruses. Scientific Reports. 2017;7(43395). doi:10.1038/srep43395

9. Detrick F. Wayback Machine. web.archive.org. Published December 24, 2016. Accessed August 1, 2021.
https://web.archive.org/web/20161224000431/http:/www.usamriid.army.mil/press_releases/Travis%20ID%2
0Week%20FINAL.pdf

10. Pardo J, Shukla AM, Chamarthi G, Gupte A. The journey of remdesivir: from Ebola to COVID-19. Drugs
in Context. 2020;9:1-9. doi:10.7573/dic.2020-4-14

11. Warren TK, Jordan R, Lo MK, et al. Therapeutic efficacy of the small molecule GS-5734 against Ebola
virus in rhesus monkeys. Nature. 2016;531(7594):381-385. doi:10.1038/nature17180

12. Jacobs M, Rodger A, Bell DJ, et al. Late Ebola virus relapse causing meningoencephalitis: a case report.
The Lancet. 2016;388(10043):498-503. doi:10.1016/S0140-6736(16)30386-5

13. Dörnemann J, Burzio C, Ronsse A, et al. First Newborn Baby to Receive Experimental Therapies
Survives Ebola Virus Disease. Journal of Infectious Diseases. 2017;215(2): doi:10.1093/infdis/jiw493
14. Mulangu S, Dodd LE, Davey RT, et al. A Randomized, Controlled Trial of Ebola Virus Disease
Therapeutics. New England Journal of Medicine. 2019;381(24):2293-2303. doi:10.1056/nejmoa1910993

15. Pedersen NC, Perron M, Bannasch M, et al. Efficacy and safety of the nucleoside analog GS-441524 for
treatment of cats with naturally occurring feline infectious peritonitis. Journal of Feline Medicine and
Surgery. 2019;21(4):271-281. doi:10.1177/1098612x19825701

16. Pedersen N. Black Market Production and Sale of GS-441524 and GC376. ; 2019. Accessed August 1,
2021. https://ccah.vetmed.ucdavis.edu/sites/g/files/dgvnsk4586/files/inline-
files/Black%20market%20production%20and%20sale%20of%20GS.pdf

17. Westgate J. Vet science “being ignored” in quest for COVID-19 drug. Vet Times. Published May 7, 2020.
Accessed August 1, 2021. https://www.vettimes.co.uk/news/vet-science-being-ignored-in-quest-for-covid-19-
drug/

18. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged
novel coronavirus (2019-nCoV) in vitro. Cell Research. 2020;30(3):269-271. doi:10.1038/s41422-020-0282-0

19. Center for Drug Evaluation and Research. Drug Trials Snapshots: VEKLURY. FDA. Published October
27, 2020. Accessed August 1, 2021. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-
snapshots-veklury

20. Office of the Commissioner. FDA Approves First Treatment for COVID-19. FDA. Published October 22,
2020. Accessed August 1, 2021. https://www.fda.gov/news-events/press-announcements/fda-approves-first-
treatment-covid-19

21. U.S. Food and Drug Administration Approves Gilead’s Antiviral Veklury® (remdesivir) for Treatment of
COVID-19. www.gilead.com. Published October 22, 2020. Accessed August 1, 2021.
https://www.gilead.com/news-and-press/press-room/press-releases/2020/10/us-food-and-drug-administration-
approves-gileads-antiviral-veklury-remdesivir-for-treatment-of-covid19

22. Wang Y, Zhang D, Du G, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind,
placebo-controlled, multicentre trial. The Lancet. 2020;395(1569–78). doi:10.1016/S0140-6736(20)31022-9

23. McCreary EK, Angus DC. Efficacy of Remdesivir in COVID-19. JAMA. 2020;324(11):1041-1042.
doi:10.1001/jama.2020.16337

24. WHO Solidarity Trial Consortium. Repurposed Antiviral Drugs for Covid-19 — Interim WHO Solidarity
Trial Results. New England Journal of Medicine. 2020;384:497-511. doi:10.1056/nejmoa2023184

25. World Health Organization. WHO recommends against the use of remdesivir in COVID-19 patients.
www.who.int. Published November 20, 2020. Accessed August 1, 2021. https://www.who.int/news-
room/feature-stories/detail/who-recommends-against-the-use-of-remdesivir-in-covid-19-patients

26. Jarvis L. Scaling up remdesivir amid the coronavirus crisis. Chemical & Engineering News. Published
April 20, 2020. Accessed August 1, 2021. https://cen.acs.org/biological-chemistry/infectious-disease/Scaling-
remdesivir-amid-coronavirus-crisis/98/web/2020/04

27. Synthetic Efficiency. Chemistry LibreTexts. Published November 11, 2014. Accessed August 1, 2021.
https://chem.libretexts.org/Courses/University_of_California_Davis/UCD_Chem_231A%3A_Methods_of_Or
ganic_Synthesis/Text/Synthetic_Efficiency
28. Bhat S. Synthesis of Remdesivir by Mackman. www.organic-chemistry.org. Published April 10, 2020.
Accessed August 1, 2021. https://www.organic-chemistry.org/totalsynthesis/totsyn07/remdesivir-
mackman.shtm

29. Gannedi V, Villuri BK, Reddy SN, Ku C-C, Wong C-H, Hung S-C. Practical Remdesivir Synthesis
through One-Pot Organocatalyzed Asymmetric (S)-P-Phosphoramidation. The Journal of Organic Chemistry.
2021;86(7):4977-4985. doi:10.1021/acs.joc.0c02888

30. Vieira T, Stevens AC, Chtchemelinine A, Gao D, Badalov P, Heumann L. Development of a Large-Scale
Cyanation Process Using Continuous Flow Chemistry En Route to the Synthesis of Remdesivir. Organic
Process Research & Development. 2020;24(10):2113-2121. doi:10.1021/acs.oprd.0c00172

31. Liang C, Tian L, Liu Y, et al. A promising antiviral candidate drug for the COVID-19 pandemic: A mini-
review of remdesivir. European Journal of Medicinal Chemistry. 2020;201(112527):
doi:10.1016/j.ejmech.2020.112527

32. Ward TJ, Oswald TM. Chiral Separations by High-Performance Liquid Chromatography. Encyclopedia of
Analytical Chemistry. Published online September 15, 2006. doi:10.1002/9780470027318.a5905

33. Regis Technologies. 6 Top Chiral Chromatography Questions. Regis Technologies. Published December
12, 2020. Accessed August 1, 2021. https://www.registech.com/blog/6-top-chiral-chromatography-questions/

34. Liu S, Zhang Z, Xie F, Butt NA, Sun L, Zhang W. First catalytic enantioselective synthesis of P-
stereogenic phosphoramides via kinetic resolution promoted by a chiral bicyclic imidazole nucleophilic
catalyst. Tetrahedron: Asymmetry. 2012;23(5):329-332. doi:10.1016/j.tetasy.2012.02.018

35. De Miranda AS, Gomes JC, Rodrigues MT, et al. Kinetic resolution of 5H-pyrrolo[1,2-a]imidazol-7-ol,
6,7-dihydro under continuous flow conditions: An intermediate for chiral ionic liquids synthesis. Journal of
Molecular Catalysis B: Enzymatic. 2013;91:77-80. doi:10.1016/j.molcatb.2013.02.008

36. Xue F, Zhou X, Zhou R, et al. Improvement of the C-glycosylation Step for the Synthesis of Remdesivir.
Organic Process Research & Development. 2020;24(9):1772-1777. doi:10.1021/acs.oprd.0c00310

You might also like