You are on page 1of 11

European Journal of Medicinal Chemistry 234 (2022) 114256

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Chemical synthesis and pharmacological properties of heparin


pentasaccharide analogues
Zhipeng Zhou a, 1, Linlin Zhang b, 1, Xin Wu a, c, Lan Luo a, Jian Wu b, Dan Xu b, **,
Mingyi Wu a, c, *
a
State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201,
China
b
Nanjing Chia Tai Tianqing Pharmaceutical Co., Ltd, Nanjing, 210046, China
c
University of Chinese Academy of Sciences, Beijing, 100049, China

a r t i c l e i n f o a b s t r a c t

Article history: The pentasaccharide fondaparinux is a synthetic anticoagulant based on heparin antithrombin-binding
Received 30 January 2022 sequence. Fondaparinux improves safety and predictable pharmacodynamics compared with heparins;
Received in revised form however, it requires a complicate synthesis process which contain more than 50 steps of synthesis.
2 March 2022
Herein, we designed and synthesized four fondaparinux analogues (compounds 1, 2, 3, 4) using a [2þ3]
Accepted 3 March 2022
convergent synthetic method, which greatly simplified the synthetic process, improved the product
Available online 5 March 2022
yield, and curtailed the expenditures. These synthesized compounds showed stronger anticoagulant
activities by factor Xa inhibition (IC50 725e1126 nM vs. 1909 nM for fondaparinux) in the AT-dependent
Keywords:
Heparin
manner. After subcutaneous (s.c.) administration to rats, the compounds displayed long-lasting anti-
Pentasaccharide factor Xa activities and inhibition of thrombin generation ex vivo. Compared with fondaparinux, these
Anticoagulant compounds were slowly eliminated after s.c. administration to rats, the half-lies (t1/2) were more than 2-
Structure-activity relationships fold of that of fondaparinux. These results suggested the pentasaccharide analogues may exhibit better
Synthesis pharmacokinetic and predictable pharmacodynamic characteristics.
© 2022 Elsevier Masson SAS. All rights reserved.

1. Introduction therapy of venous thromboembolism (VTE) and the prevention of


stroke in patients with atrial fibrillation, namely heparins (paren-
Thrombosis is the leading cause of death worldwide, being teral), vitamin K antagonists (VKAs; oral), direct thrombin in-
responsible for one in four deaths [1]. The global burden is likely to hibitors (DTIs; parenteral and oral) and direct factor Xa (FXa)
increase with the ageing population because thrombosis risk in- inhibitors (oral) [2,3].
creases with age. Anticoagulants are used to treat a wide variety of The first anticoagulant, heparin (also referred to as unfractio-
conditions that involve arterial or venous thrombosis, including nated heparin (UFH)), has been a successful anticoagulant for over
prevention of venous thromboembolism and long-term prevention 80 years and is on the WHO list of essential drugs [4]. Despite the
of ischaemic stroke in patients with atrial fibrillation. There are four availability of other anticoagulants, it remains a major therapeutic
crucial classes of anticoagulants applied to the prevention and choice especially in surgical interventions. Unfractionated heparin
binds to the protein antithrombin (AT) and markedly increases the
ability of this protein to inhibit factor Xa and thrombin [2e5],
Abbreviations: APTT, activated partial thromboplastin time; AT, antithrombin;
which is currently used for cardiovascular surgery and for the
Fpx, fondaparinux sodium; FX, coagulation factor X; HCII, heparin cofactor II; prevention of venous thromboembolism. Fractionated heparin, in
LMWH, low-molecular-weight heparin; PT, prothrombin time; TT, thrombin time. the form of low-molecular-weight heparins (LMWHs), is intro-
* Corresponding author. State Key Laboratory of Phytochemistry and Plant Re- duced to improve the pharmacodynamics. These molecules also
sources in West China, Kunming Institute of Botany, Chinese Academy of Sciences,
target both factor Xa and thrombin, but their administration results
Kunming, 650201, China.
** Corresponding author.
in a lower incidence of bleeding than does unfractionated heparin
E-mail addresses: xiaoxux@gmail.com (D. Xu), wumingyi@mail.kib.ac.cn (1.4% for LMWHs versus 2.3% for UFH) [6]. Heparin suffers from
(M. Wu). several noteworthy limitations, including heparin-induced
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ejmech.2022.114256
0223-5234/© 2022 Elsevier Masson SAS. All rights reserved.
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

thrombocytopenia (HIT), a limb-threatening and life-threatening because of the difficulty in regio- and stereo-selective glycosidic
severe adverse event in which antibodies form against complexes bonds among the glucosamine, glucuronic acid, and iduronic acid
of heparin and platelet factor 4 and induce platelet activation and a building blocks, and the strategic installation of OSO 3 and NHSO3


prothrombotic state [7,8]. Fortunately, the incidence of heparin- groups [15]. Glucosaminylation mainly relies on the anomeric ef-
induced thrombocytopenia is reduced when low molecular- fect, which is insufficient in curbing production of the unwanted b
weight heparins are used [9]. isomer. The 1,2-trans glycosylation involving the uronic acid pre-
Synthetic pentasaccharides, such as fondaparinux, have been cursors, although feasible through neighboring group participation,
designed with a structure based on the antithrombin-binding is confounded by the different C2 functionalizations in the final
sequence of heparin [4,5]. Fondaparinux with the sequence D- product. So far, in order to ameliorate the synthetic efficiency, main
GlcNS6S-a-(1,4)-D-GlcA-b-(1,4)-D-GlcNS3,6S-a-(1,4)-L-IdoA2S-a- efforts have been made to synthesize heparin-like oligosaccharides
(1,4)-D-GlcNS6S-OMe (DEFGH) was identified as the AT-binding chemically or enzymatically by several groups, including Boons
sequence and later has been commercialized since 2002 with the [16], Huang [17], Hung [18,19], Linhardt [20], Liu [21], Petitou
trade name “Arixtra” (Fig. 1) [10]. Fondaparinux is shown to have [14,22], Qin [23], Seeberger [24,25], and Wong [15,26], but the
more predictable pharmacodynamics, non-existent risk of HIT, and procedures still encounter problems such as a long stepwise pro-
better biosafety like those of LMWH [5,8,10,11]. Total synthesis of cess, non-stereoselective glycosylation and low yield and efficiency.
fondaparinux has allowed to eliminate inherent limitations of Further researches led to develop a non-glycosaminoglycan
animal-sourced heparin and LMWHs, and avoided the contami- analogue, idraparinux, a synthetic pentasaccharide analogue of
nation in naturally occurring heparins that caused several deaths in fondaparinux, in which the N-sulfates are replaced by O-sulfates
2008 [12]. and the hydroxyl groups are methylated [27e29] (Fig. 1). Although
Chemical synthesis of fondaparinux, the most successful idraparinux is considerably easier to synthesize than fondaparinux,
example to date for preparing a synthetic heparin [10,13], entails it still poses some challenges such as the synthesis of the L-idosyl,
more than 50 steps with an overall yield of ~0.1% [14]; as such, the stereoselectivity of glycosylation between D and E, and the
fondaparinux is the most expensive drug among heparins. In stereoselectivity of glycosylation between G and H. Since its first
particular, the total synthesis of fondaparinux is very challenging synthesis, several research groups have developed alternate

Fig. 1. Structures and pharmacological properties of fondaparinux, idraparinux and compounds 1e4.

2
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

synthetic routes to idraparinux [27,30e33]. For example, Yu et al. D-glucofuranose via intramolecular epoxide formation, in 38% yield.
[27] used DEGþGH coupling strategy in total 51 steps from D- The glycosylation between donor 10 and acceptor 11 produced
glucose and methyl D-glucopyranoside. Borba s et al. used DEþFGH the protected disaccharide 9, in 46% yield. The yield (46%) is not
([2 þ 3]) coupling strategy in 39 steps [30,31] and the L-idose- good enough because part of donor 10 quickly turned to other
containing GH fragment was obtained by a short and straightfor- impurities under this condition, which might also be further opti-
ward synthesis whereby a 4,6-cyclic-acetal-protected L-idosyl thi- mized. We suggested that 1,2-trans a-selective glycosylation may
oglycoside bearing a C2-nonparticipating group was used as the a- be due to the formation of 1, 2-cis-nitrilium ions with low steric
selective glycosyl donor [33]. resistance from donor 10 to acetonitrile at low temperatures.
Idraparinux binds to antithrombin significantly stronger than Demeter et al. applied a 4,6-acetal-protected L-idose donor without
fondaparinux through the additional interaction of the extra sulfate a C2 participating group to ensure 1,2-trans a-selective glycosyla-
group at C3 of the H glucose unit as well as through hydrophobic tion [33]. Subsequently, the trisaccharide acceptor 6 (F-G-H) was
interactions [34,35]. Due to the increased sulfation, this agent ex- synthesized by the glycosylation between 4-OH-glucopyranoside
hibits a 30-fold higher binding affinity to AT than fondaparinux, derivative 9 and donor 7b. Additionally, the disaccharide donor 5
and a higher anti-factor Xa potency. Idraparinux exhibits a signifi- (D-E) was produced by linking donor 7 and acceptor 8, beyond 80%
cantly longer elimination half-life of about 120 h in clinical trial, yield. When TMSOTf was used as a catalyst at low temperature,
which allows for once-weekly dosing [35]. Unfortunately, idrapar- donor 7 without neighboring group participation was firstly con-
inux did not obtain FDA regulatory approval due to significant verted into oxonium ion, and then oxonium ion was attacked by
bleeding complications in the clinical trial III [36]. receptor 8 under the influence of dichloromethane which tended to
Approaches to establish structure-activity relationships form a -D-glycoside [27]. Finally, the target product b-linked pen-
applying modified heparin-like oligosaccharides have been proved tasaccharide analogues 1e4 were formed by the glycosylation
feasible [5,10,37]. Accordingly, continuous research may lead to [2 þ 3] between the trisaccharide 6 and the disaccharide 5 (Scheme
develop more potent pentasaccharide derivatives. According to the 2).
synthesis methods of non-glycosaminoglycan analogues [27e33], Differing from the traditional synthesis of idraparinux, in this
we would expect to obtain some new pentasaccharide analogues work, the carboxyl and hydroxyl groups were protected by benzyl
via changing degree of methylation, which might exhibit better groups, which were stable and susceptible to be removed. As a
pharmacological properties mostly including proper half-life and result, the conventional two pressurized hydrogenation steps
then less bleeding risk. In this work, we adopted a distinctive were simplified to one. And, due to the strong neighboring group
[2 þ 3] convergent method to chemically synthesize four fully O- participation of the acetyl group at C2 of E ring, almost no a-iso-
sulfated, partially O-methylated or O-acetylated analogues (1, 2, 3, mer was produced during the [D-E] þ [F-G-H] glycosylation, thus
4). Their anticoagulant activities were evaluated by coagulation optimizing the impurity control which is very important for in-
time assays, calibrated automated thrombogram (CAT) and dustrial production. When the protected disaccharide 9 were
thrombin generation test (TGT). AT-binding affinity and anti-FXa produced, trichloroacetyl imide lipides were used as a protective
activity were detected to confirm the action mechanism. Further- base for C-1 of donor 10, avoiding the use of highly toxic ethyl
more, their pharmacokinetics and pharmacodynamics were also mercaptan and very expensive silver trifluoroacetate. In G ring
investigated in rats. Our results indicated that the pure synthetic synthesis, p-methoxy phenyl ether was used to protect the C-1
pentasaccharide 3 may be not only much easier to synthesize than site, making compounds 46e49 easy to crystallize. By the [2 þ 3]
fondaparinux, but also displays excellent anti-FXa activity, a longer route, we were also able to avoid the secondary sulfonation and
duration of action and lower risk of bleeding. the mass impurities produced by the de-methyl ester in strong
base.
2. Results In the preparation of a-glycosylation between D ring and E ring,
b-isomer is an accompanied by-product (2%e20%), resulting in low
2.1. Synthesis procedure for compounds 1-4 yield of D-E building block. In the conventional [3 þ 2] route [27],
the preparation of D-E-F building block further reduces the yield of
To provide an efficient and concise access to these new heparin final product, it's also very time-consuming and high-cost. As for
pentasaccharide analogues, we conceived a [2 þ 3] convergent the [1 þ 4] strategy [28,29], it's more complex to synthesize the E-F-
synthetic route through the building blocks 7, 8, 10 and 11 (Scheme G-H building block, as well as thornier to remove the higher level of
1), all of which are readily attainable from commercially available isomer impurities. Additionally, the raw material of iduronic acid
starting materials. The approach followed the formation of the residue (G ring) is derived by conformational transform of other
disaccharide donors 5a, 5b, 5c, 5d and their subsequent coupling saccharide units, which is expensive, and its stereo-selective link-
age with other building block can result in further loss [18]. Borba s
with the reducing end trisaccharide acceptor 6 to generate the fully
protected precursors of the target compounds 1e4 (Fig. 1). et al. used a [2 þ 3] block synthesis utilizing a 6-O-silyl-protected L-
The monosaccharide 11 was synthesized using methyl a-D-glu- idose-containing trisaccharide acceptor. The unique strategy
copyranoside as the starting material (Scheme S1). Donor 7a was involved triple methylation followed by oxidation of the glucose
derived from compound 11 by the methylation of 5-OH, selective and the idose precursors into D-glucuronic and L-iduronic acids in
removal of the methyl group in 1-OH, and reacted with CCl3CN and one step, and provided the target pentasaccharide through a 39-
DBU, in 59% yield (Scheme S1). The synthesis of donor 7b was also step synthesis starting from D-glucose and methyl a-D-glucopyr-
derived from compound 11 by selective demethylation, acetylation, anoside [31]. In this work, four new pentasaccharides were syn-
and selective deacetylation, etc., in 27% yield (Scheme S2). Donor 7c thesized in overall 37 steps from commercially available starting
was stemmed from 1,2:5,6-di-O-isopropylidene-a-D-glucofuranose materials and in 1.2% overall yield over 20 linear steps from methyl
after eleven reactions, in 37% yield. Donor 7d was obtained from a-D-glucopyranoside (building block D). The [2 þ 3] convergent
methyl a-D-glucopyranoside with seven steps, in 40% yield. synthesis strategy showed excellent stereoselectivity in all glyco-
Acceptor 8 was derived from 22 through five steps with the yield of sylation steps, while avoiding rigorous reaction conditions and
71%. According to the strategy of Chen and Yu [27], the L-idopyr- expensive reagents, which could largely benefit the large-scale
anose donor 10 was synthesized by the epimerization of 5-OH in a- industrial production.

3
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

Scheme 1. Retrosynthetic analysis of the heparin pentasaccharide analogues 1e4. Ac ¼ acetyl, Bn ¼ benzyl, Ph ¼ benzylidene.

2.2. Anticoagulant activities of the pentasaccharide compounds assays, and thrombin generation (TG) inhibition activities were also
in vitro tested to further study the action mechanism (Fig. 2 and Table 1).
Compounds 1e4 showed potent FXa inhibitory activity in the
The anticoagulant activities of synthetic pentasaccharide com- presence of AT, with IC50 values of 88.8e120.6 nM, which was
pounds 1e4 and fondaparinux (Fpx) were evaluated by plasma comparable with that of Fpx (IC50, 99.6 nM) (Fig. 2B, Table 1).
clotting assays. These include using activated partial thrombo- Compounds 1e4 could strongly inhibit the binding of FXa to the
plastin time (APTT), prothrombin time (PT) and thrombin time (TT) immobilized heparin (IC50 values of 725e1126 nM), slightly lower
of plasma clotting assays. Such assays are routinely used to deter- than that of Fpx (IC50, 1909 nM) (Fig. 2C, Table 1). Additionally,
mine the ability of products to inhibit blood clotting through the compounds 1e4 strongly inhibited thrombin generation (TG) in
intrinsic, extrinsic and common pathways of the coagulation human plasma in a dose-dependent manner with IC50 values of
cascade, respectively [38e40]. The concentrations of these com- 11e19 mM (Table 1 and Fig. 2D, Fig. S44), whereas Fpx also inhibited
pounds 1e4 needed to double APTT (EC2.0) were 32.47e48.78 mM, TG (IC50, 14 mM) as previously described for this compound
which were slightly lower than that of Fpx (EC2.0, 51.20 mM) while [22,34,41]. Taken together, compounds 2e4 exhibited slightly
significantly higher than LMWH (EC2.0, 2.19 mM) (Fig. 2A, Table 1), stronger anticoagulant activity than Fpx.
indicating that these compounds have potent intrinsic anticoagu-
lant activities. As for PT and TT, no significant influences were 2.3. Pharmacokinetics and pharmacodynamics of the
observed in 1e4 and Fpx at their concentrations up to 128 mg/mL pentasaccharide compounds in vivo
compared with LMWH, which mean that they have no or little ef-
fect on the extrinsic and common coagulation pathways [10,40]. After administrating subcutaneously to rates, the plasma con-
The AT-dependent anti-FXa activities of compounds 1e4 and centrations of compounds 1e4 and Fpx were determined based on
their AT binding affinities were evaluated by competitive binding standard curves between the plasma drug concentration and anti-
4
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

compound 3 was comparable to Fpx (56.94 ± 8.12% for 3 vs.


55.66 ± 6.23% for fondaparinux) (Fig. 3B). Notably, compounds 2, 4
and Fpx showed the strongest antithrombotic activity at 1 h, while
1 and 3 were the most potent after injection 3 h. Moreover, at the
same dose, all the four oligosaccharides exhibited stronger
thrombosis inhibitions than Fpx after administration 3 h, consistent
with the results of pharmacokinetics (Fig. 3A). The results of
pharmacodynamics of oligosaccharides showed the linear kinetics,
suggesting the predictable pharmacodynamic characteristics.

2.4. Antithrombotic activity and hemorrhagic risk of the


pentasaccharide compounds in vivo

The dose-antithrombotic activity relationships of compounds


1e4 were further examined using the rat model of deep venous
thrombosis at three doses of 30, 100, 300 nmol/kg after 2h subcu-
taneously administration. The results indicated that all compounds
displayed potent inhibitions of thrombosis the same as Fpx in the
dose-dependent manner (Fig. 4A). At 30 nmol/kg, compound 3
exhibited the highest thrombosis inhibitory activity (inhibition rate
48.58 ± 11.5%), 1 and 2 also showed markedly antithrombotic activity
(33.19 ± 6.52% and 39.48 ± 13.69), whereas 4 and Fpx showed lower
inhibitory activity (less than 15%). At the doses of 100 and 300 nmol/
kg, the four oligosaccharides all showed stronger antithrombotic
activity than Fpx (p < 0.01). Particularly, compound 3 and 4 at
100 nmol/kg had the thrombosis inhibition rates of more than 70%,
moreover at 300 nmol/kg they inhibited thrombosis by
89.54% ± 2.4% and 93.12% ± 1.23%, respectively, whereas the inhi-
bition rate of Fpx was 59.31% ± 6.29%. These results suggested that
our synthesized oligosaccharides exhibited stronger antithrombotic
activities after administration subcutaneously into rats in vivo.
Bleeding risk has been the main problem in the application of
anticoagulants [2,3]. Herein, the bleeding risk of the synthetic
compounds was evaluated using mice tail-cut model [39,42]. Mice
were treated with compounds 1, 2, 3 and 4 at approximately 10
times antithrombosis dose (2000 nmol/kg) (Fig. 4B). The blood loss
Scheme 2. Synthesis of compounds 1e4. Reagents and conditions: a) dichloro- of negative control (normal saline) group was 33.7 ± 8.2 mL. In
methane, TMSOTf; b) methanol, palladium carbon; c) N, N-dimethylformamide, sulfur contrast to the negative control and Fpx treated group, both LMWH
trioxide trimethylamine complex; d) sodium hydroxide, ammonium acetate. Detailed and compound 1 significantly increased the blood loss (P < 0.05).
reaction conditions are described in Experimental Section.
Compounds 2e4 slightly increased the blood loss, but displayed no
statistical significance (P > 0.05) due to their larger individual
variations. As shown in pharmacokinetic results (Fig. 3A and
FXa activity (Fig. 3A), and the pharmacokinetics parameters were Table 2), Tmax for Fpx was 0.6 h, while the compounds were
calculated according to noncompartmental procedure model [42] 1.0e2.0 h, which implied that these compounds may exhibit more
(Table 2). After administrating each compound at the dose of exposures than Fpx at 1.5 h in the blood of mice after subcutane-
100 nmol/kg, the maximum times (Tmax) to reach the maximum ously administration. Nevertheless, risk of bleeding for compounds
drug concentration (Cmax) were 2.0 ± 0.45 h, 1.0 ± 0.27 h, 1.5 ± 0.29 2e4 are probably acceptable as anticoagulants compared with Fpx
and 2.0 ± 0.90 h, respectively, and their half-lives (t1/2) were and low molecular weight heparin in clinic. Further investigation
15.1 ± 1.14 h, 4.8 ± 1.11 h, 6.2 ± 1.10 h and 6.1 ± 1.11 h for compounds may be required for the hemorrhagic effect of these compounds,
1, 2, 3, and 4, respectively, which were more than 2-fold of that of such as more animal groups and detection at different time points
Fpx (1.2 ± 0.25 h) (Fig. 3A, Table 2). Moreover, other parameters of after administration.
compounds 1e4, including the Cmax and AUC0e32h (the area under
the time-drug concentration profiles within 32 h), displayed much 3. Discussion and conclusions
higher than those of Fpx, while the clearance rates of these com-
pounds were lower compared to Fpx. Above results indicated that Although many anticoagulants are available in clinic, their ap-
our four synthesized compounds may exhibit the stronger and/or plications are still limited out of the safety concern, and great ef-
longer drug exposures than fondaparinux mostly because these forts have been made in developing ideal anticoagulant drugs [43].
compounds were partially O-methylated or O-acetylated analogues Fondaparinux has been widely used in treating venous thrombo-
of Fpx. embolic events, deep vein thrombosis, and acute coronary syn-
The antithrombotic effects of 1e4 (100 nmol/kg) at different drome [44,45], etc. Fondaparinux is a highly selective FXa inhibitor
time points were evaluated using the rat model of deep venous by binding to AT, and as a potent anticoagulant due to it exhibits
thrombosis induced by stasis and thromboplastin [39]. The results fewer side effects and more predictable pharmacodynamics than
showed that after 1 h subcutaneously administration, the throm- heparin or LMWH in clinic [11,45]. However, the high cost of fon-
bosis inhibition rate of compound 4 was 77.78 ± 8.55%, indicating daparinux treatment results from the high-cost and time-
the best antithrombotic activity than others compounds, as well as consuming manufacturing process, limiting its availability.
5
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

A 100 B
Fpx 100
90 1
2

FXa activity (% of control)


3 Fpx
80 4 75 1
2
APTT (s)

70 3
50
4

60

50 25

40
0
0 20 40 60 80 100 120 140 0.1 1 10 100 1000
Concentration ( g/mL) Concentration ( g/L)

C 1.2 D 350 Compound 3


AT-heparin binding (% of control)

300
1.0

250
0.8 Fpx
1 Thrombin (nM)
2 200
0.6 3
4 150

0.4
100

0.2
50

0.0 0
10-1 100 101 102 103 104 105 0 10 20 30 40 50
Concentration (nM) Time (min)

Fig. 2. Anticoagulant activities of Fpx and compounds 1e4. Effects of Fpx and compounds 1e4 on human plasma APTT (A), AT-heparin binding (B), FXa activity in the presence of AT
(C), and inhibition of thrombin generation in human plasma (D). Experiments were repeated triplicate, data were expressed as mean ± SD in A-C, and the representative graph was
shown in D. Fpx, fondaparinux; APTT, activated partial thromboplastin time; AT, antithrombin; FXa, activated coagulation factor X.

Table 1
In vitro anticoagulant activities and effects of compounds 1e4 on coagulation factors.

Compd. APTT (EC2.0, mM)a PT (mg/mL)b TT (mg/mL)b Anti-FXa activity (IC50, nM)c Competitive AT-binding (IC50, nM)c TG inhibition (IC50, mM)

1 32.47 >128 >128 88.8 ± 7.00 1126.0 ± 79.18 14.82 ± 1.59


2 33.05 >128 >128 94.7 ± 9.34 778.7 ± 64.71 12.39 ± 0.50
3 48.78 >128 >128 108.1 ± 7.10 787.9 ± 68.30 12.53 ± 0.58
4 40.83 >128 >128 141.08 ± 10.2 724.7 ± 53.34 12.17 ± 1.71
LMWH 2.19 28.4a 1.12a 26.6d >4000 NDe
Fpx 51.20 >128 >128 99.6 ± 10.80 1909.5 ± 290.32 14.51 ± 0.62
a
EC2.0, the activity of agents to prolong APTT, PT or TT is expressed by the concentration of each agent (mM) that is required to double the APTT, PT or TT.
b
No significant influences of compounds on PT or TT were observed at concentration as high as 128 mg/mL.
c
IC50 value, the concentration of each agent required to inhibit 50% of protease activity.
d
Data from ref. 39.
e
ND, not determined.

Idraparinux, a synthetic pentasaccharide analogue of fonda- and less bleeding risk, a distinctive [2 þ 3] convergent synthetic
parinux, binds to antithrombin significantly stronger than fonda- strategy was reported to synthesize idraparinux analogues (com-
parinux [34] and is synthesized in shorter steps relative to pounds 1e4). Compared with the conventional [1 þ 4] or [3 þ 2]
fondaparinux [30,31]. Due to the increased sulfation, this agent strategies [19,27,47], the synthetic process was remarkably
exhibits a 30-fold higher binding affinity to AT than fondaparinux, simplified by [2 þ 3] route according to the synthetic process of
and a higher anti-factor Xa potency. Idraparinux exhibits a signifi- idraparinux [30,31]. Particularly, since the acetyl group at E-ring
cantly longer elimination half-life, which allows for once-weekly has a strong neighboring group participation on DEþFGH glucosi-
dosing [34,35]. But in phase III clinical the long-term (more than dation, by-products of isomer impurity were effectively reduced,
6 months) use of idraparinux also led to intracranial bleeding in and almost without alpha product. The expensive G-ring raw ma-
elderly patients and those with renal impairment [36]. terial was less required, and then the product yield increased, thus
In this study, in order to obtain non-glycosaminoglycan fonda- the same products can be obtained by this method in a cost-
parinux analogues as better anticoagulants with moderate half-life effective way. Previously, the structure-activity relationship study

6
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

A B 100
Fpx Fpx
2400
1 1

Inhibition of thrombosis (%)


2 2
75 ***
3
Concentration ( g/L)
3
1800 4 4
**
**
**
50
**
1200
* **
25
600

*
0
0
0 5 10 15 20 25 30 35 0 5 10 15 20 25
Time (h) Time (h)

Fig. 3. Pharmacokinetics and pharmacodynamics of the pentasaccharide compounds 1e4 and Fpx (s.c.) in rats. The plasma concentrations (A) and thrombosis inhibition rates (B) of
pentasaccharide compounds at indicated time points, after they were administered sc to rats at the doses of 100 nmol/kg. Data were expressed as mean ± SEM (n ¼ 6). *P < 0.05,
**P < 0.01, ***P < 0.01 vs. control group.

Table 2
Pharmacokinetic parameters of the synthetic pentasaccharide compounds in rats.

Compd. Pharmacokinetics, [Means ± SEM] (n ¼ 4 to 6).

Tmax (h) Cmax (mg/L) t1/2 (h) AUC(0-32) AUC(0-∞) Clearance (L/kg$h) Distribution volume (L/kg)
(h$mg/L) (h$mg/L)

1 2.0 ± 0.45 1141.3 ± 147.98 15.1 ± 1.14 12798.6 ± 2675.16 15057.8 ± 3665.62 0.017 ± 0.004 0.136 ± 0.024
2 1.0 ± 0.27 1247.1 ± 171.97 4.8 ± 1.11 6351.2 ± 1416.71 6754.4 ± 1428.73 0.038 ± 0.011 0.183 ± 0.047
3 1.5 ± 0.29 2016.5 ± 478.28 6.2 ± 1.10 14176.2 ± 4132.44 15536.1 ± 4890.33 0.021 ± 0.010 0.131 ± 0.031
4 2.0 ± 0.90 2041.0 ± 453.78 6.1 ± 1.11 17623.6 ± 4881.77 19053.2 ± 5467.44 0.031 ± 0.020 0.058 ± 0.005
Fpx 0.6 ± 0.12 519.2 ± 131.59 1.2 ± 0.25 1071.1 ± 315.58 1116.4 ± 324.78 0.239 ± 0.077 0.097 ± 0.055

A B *
600
100 *
***
Inhibition of thrombosis (%)

***
Blood loss ( L)

75 *** 450

*** ***
*** **
***
50 300
*** *
** **
** Fpx
25 1 150
2
3
0
4
0
0 50 100 150 200 250 300 Control Fpx LMWH 1 2 3 4
Dose (nmol/kg) Compounds

Fig. 4. Effects of the compounds 1e4 on venous thrombosis and hemostatic function. Dose-antithrombotic activity relationships (A) and bleeding effect (B) of compounds 1e4 after
administrated. (A) The thrombus weight was detected after 2 h administration (s.c.) of each compound to rats. Results are expressed as % thrombus weight (mean ± SEM, n ¼ 10,
**p < 0.01, ***p < 0.001 vs. saline), 100% representing absence of any thrombosis inhibition (thrombus weight in the absence of compound administration). (B) The blood loss was
determined after 1.5 h administration in mice tail-cut model. Data were expressed in mL of blood loss as Mean ± SEM, n  6; compared with the negative control group, *P < 0.05.

demonstrated that sulfate groups of the fondaparinux analogues Anticoagulant activities of synthetic pentasaccharide com-
are essential for biological activity [10,22], whereas the degrees of pounds were tested in contrast to fondaparinux in vitro (Fig. 2 and
methylation are correlateed with elimination half-life [28,37]. Table 1). The results indicated that these pentasaccharide ana-
Accordingly, our synthetic route introduced more sulfuric acid logues 1e4 showed high anti-FXa selectivity by binding to AT and
groups instead of amino groups and appropriate methylated with no obvious anti-thrombin activity, and exhibited APTT pro-
modification (Fig. 1), which enhanced the activities of synthetic longing activities without significant effect on PT and TT. Moreover,
compounds and extended the half-lives, while avoided the use of the synthetic analogues showed comparable anti-FXa activities and
explosive azide groups and further optimized the synthesis of slightly stronger thrombin generation inhibition. Since they exhibit
fondaparinux analogues. strong anticoagulation, including prolonging APTT, anti-FXa

7
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

activity by AT, binding to AT and TG inhibition, the abilities of these or a p-anisaldehyde solution (EtOH/p-anisaldehyde/AcOH/H2SO4,
oligosaccharides to inhibit thrombosis were closely related to their 135:5:4:1.5), followed by heating on a hot plate. Column chroma-
anticoagulant activities. tography was conducted using silica gel (200e300 mesh) with
Next, the pharmacokinetic results suggested that the fonda- EtOAc and petroleum ether or EtOAc (or CH2Cl2) and MeOH as
parinux analogues had longer half-life (t1/2), larger AUC and Cmax eluent. Procedures for chemical synthesis of 1e4 are complex and
than fondaparinux (Fig. 3A and Table 2). In contrast, fondaparinux tedious, and the description is very lengthy, thus are shown in
has a short Tmax and higher clearance [34], which consists with its detail in Supporting information.
shorter half-life and AUC. The findings should result from methyl- The structure analysis of compounds 1e4 by NMR analyses were
ation or acetylation of hydroxyl groups in compounds (Fig. 1), performed in D2O at 298 K with a Bruker Advance 400 or 800 MHz
which may increase their lipid solubility and prolong the metabolic spectrometer equipped with a13C/1H dual probe in FT model [28].
time in vivo. These pharmacokinetic properties also aligned with All spectra were recorded with HOD suppression by presaturation.
1
their pharmacodynamics (Fig. 3B). At 3 h after subcutaneously H-1H COSY, TOCSY, NOESY, 1H-13C HSQC, and HMBC spectra were
administration, compounds 1e4 showed stronger thrombosis in- recorded using stateetime proportional phase incrementation for
hibition than fondaparinux, and their inhibition rates could main- quadrature detection in the indirect dimension. All chemical shifts
tain high levels for 12 h. Compounds 1 and 3 showed the highest were relative to internal trimethylsilyl-propionic acid sodium (TSP).
antithrombotic activity at 3 h. All the compounds showed strong All samples were previously dissolved in deuterium oxide (D2O,
thrombosis inhibition in rats in dose-dependent manner. At the 99.9% D) and lyophilized three times to replace exchangeable
same dosage of fondaparinux as is used clinically into rats protons with D2O. The lyophilized samples were then dissolved in
(100 nmol/kg), the synthetic pentasaccharide compounds showed D2O at a concentration of 20 g/L. The desalted, pure oligosaccha-
much stronger antithrombotic effects than fondaparinux. The rides obtained were water-soluble white powders (HPLC purity
reduced dose requirements and prolonged effective duration could >98%) (compounds 1e4).
further benefit clinical application. In contrast, the half-lives for Methyl [4-O-methyl-2,3,6-tri-O-sulfonato-a-D-glucopyr-
compounds 2e4 in rats were much shorter than that for idrapar- anosyl]-(1 / 4)-[2-O-acetyl-3-O-methyl-b-D-glucopyranosyl
inux (4.8e6.2 h vs 14.7 h) [34], which may imply that their bleeding uronate]-(1 / 4)-[2,3,6-tri-O-sulfonato-a-D-glucopyranosyl]-
risks might less than that of idraparinux. The results of mice tail-cut (1 / 4)-[3-O-methyl-2-O-sulfonato-a-L-idopyranosyluronate]-
model confirmed that risk of bleeding for compounds 2e4 were (1 / 4)-2,3,6-tri-O-sulfonato-a-D-glucopyranoside sodium salt
low and acceptable as anticoagulants compared with fondaparinux (compound 1): 1H NMR (800 MHz, D2O) d 5.57 (dd, J ¼ 9.8, 3.5 Hz,
and low molecular weight heparin in clinic (Fig. 4B). Taken 1H), 5.51 (d, J ¼ 3.7 Hz, 1H), 5.16e5.12 (m, 2H), 4.93e4.81 (m, 1H),
together, the fondaparinux analogues may exhibit better pharma- 4.60 (dd, J ¼ 11.4, 7.7 Hz, 2H), 4.52e4.46 (m, 2H), 4.42 (dd, J ¼ 9.4,
cokinetic and predictable pharmacodynamic characteristics. 5.0 Hz, 1H), 4.40e4.28 (m, 5H), 4.26 (t, J ¼ 4.1 Hz, 1H), 4.24 (dd,
In conclusion, the synthesized pentasaccharide compounds may J ¼ 10.1, 3.6 Hz, 1H), 4.18 (dd, J ¼ 16.9, 10.5 Hz, 3H), 4.05e3.92 (m,
be potentially superior to fondaparinux and idraparinux, consid- 5H), 3.85 (d, J ¼ 9.7 Hz,1H), 3.78 (d, J ¼ 9.0 Hz, 2H), 3.59 (d, J ¼ 3.2 Hz,
ering their lower cost, higher yield, stronger antithrombotic activ- 8H), 3.55 (s, 3H), 3.46 (d, J ¼ 9.2 Hz, 4H), 2.27 (d, J ¼ 5.7 Hz, 3H). 13C
ity, longer half-life, and low bleeding risk. Thus, these compounds NMR (201 MHz, D2O) d 176.06, 175.98, 103.95, 102.95, 102.46,
(particularly 3 or 4) may be promising candidates in the treatment 102.43, 102.34, 101.46, 100.03, 99.88, 99.64, 99.46, 99.25, 97.85,
and prevention of thrombotic diseases. 97.50, 97.26, 96.89, 87.92, 87.12, 86.68, 86.64, 86.35, 84.28, 84.05,
83.91, 82.10, 81.35, 80.93, 80.86, 80.55, 80.49, 80.19, 80.13, 80.10,
4. Experimental section 79.88, 79.57, 79.41, 79.04, 78.87, 78.69, 78.58, 78.33, 78.14, 78.01,
77.92, 77.84, 77.81, 77.77, 77.74, 77.71, 77.66, 77.11, 76.71, 76.57,
4.1. Materials 76.29, 76.25, 76.21, 75.79, 75.31, 74.89, 74.80, 74.53, 73.31, 72.73,
72.36, 72.30, 72.03, 71.97, 71.88, 71.85, 71.82, 71.58, 71.55, 71.50,
Fondaparinux sodium injection (Arixtra®, 0.5 mL: 5 mg/mL) 71.36, 69.47, 68.93, 68.83, 68.69, 68.35, 68.29, 68.22, 63.24, 63.19,
was offered by GSK (Britain). the thromboplastin time (APTT), 62.98, 62.96, 62.86, 62.75, 62.04, 61.96, 61.59, 61.54, 58.12, 58.05,
prothrombin time (PT), thrombin time (TT) reagents, and standard 57.93, 57.26, 23.60, 23.47. MS(ESI): 2018.7 [MþH]þ, 2040.7
human plasma were all from TECO GmbH (Germany). LMWH [(M þNa]þ, 1960.7 [M-SO3þNa]þ
(Enoxaparin, 0.4 ml  4000 AXaIU) was from Sanofi-Aventis Methyl [2,3,6-tri-O-sulfonato-a-D-glucopyranosyl]-(1 / 4)-
(France). AT-dependent anti-factor Xa test kit (Heparin, ANTI-Xa) [3-O-methyl-b-D -glucopyranosyluronate]-(1 / 4)-[2,3,6-tri-O-
and antithrombin (AT) were from HYPHEN BioMed (France). sulfonato-a-D-glucopyranosyl]-(1 / 4)-[3-O-methyl-2-O-sulfo-
Normal human plasma was provided by Bedford (France). nato-a-L-idopyranosyluronate]-(1 / 4)-2,3,6-tri-O-sulfonato-a-
1
Thrombin Generation test kit (Thrombin Calibrator, Flu Ca Kit and D-glucopyranoside sodium salt (compound 2): H NMR (800 MHz,
PPP Reagent) were purchased from Stago Co. (Germany). One-time D2O) d 5.66 (d, J ¼ 3.5 Hz, 1H), 5.53 (d, J ¼ 3.5 Hz, 1H), 5.19 (s, 1H),
use of automatic quantitative intravenous blood vessels (sodium 5.15 (d, J ¼ 3.5 Hz, 1H), 4.64 (d, J ¼ 7.5 Hz, 1H), 4.61 (t, J ¼ 9.2 Hz, 1H),
citrate anti-coagulation tube) and blood collection of 4 mL con- 4.57 (t, J ¼ 9.6 Hz, 1H), 4.52 (s, 1H), 4.49 (d, J ¼ 11.3 Hz, 1H), 4.44 (t,
taining sodium citrate were purchased from Wuhan Zhiyuan J ¼ 11.2 Hz, 2H), 4.41e4.34 (m, 4H), 4.33e4.26 (m, 3H), 4.24 (dd,
Technology Co. Ltd (China). All other chemicals were of reagent J ¼ 10.1, 3.5 Hz, 1H), 4.19 (d, J ¼ 10.9 Hz, 1H), 4.03 (q, J ¼ 10.3, 9.6 Hz,
grade and obtained commercially. 4H), 3.95 (q, J ¼ 10.2 Hz, 2H), 3.88e3.75 (m, 3H), 3.65 (s, 3H), 3.60 (s,
4H), 3.53 (d, J ¼ 8.5 Hz, 1H), 3.46 (s, 3H).13C NMR (201 MHz, D2O)
4.2. General procedures for compounds 1e4 d 103.92, 100.04, 99.30, 96.64, 88.05, 83.76, 80.88, 80.46, 78.98,
78.67, 78.12, 77.67, 77.64, 77.52, 77.39, 76.25, 75.27, 74.78, 72.70,
All reagents were purchased from commercially sources and 72.45, 71.79, 70.81, 68.96, 68.88, 68.71, 62.82, 61.98, 58.13. MS(ESI):
used without further purification. All solvents were available 1962.7[MþH]þ, 1984.6 [MþNa]þ
commercially dried or freshly dried and distilled prior to use. Re- Methyl [3-O-methyl-2,6-di-O-sulfonato-a-D-glucopyr-
actions were monitored by thin-layer chromatography (TLC) using anosyl]-(1 / 4)-[3-O-methyl-b-D-glucopyranosyluronate]-
silica gel GF254 plates with detection using short wave UV light (1 / 4)-[2,3,6-tri-O-sulfonato-a-D-glucopyranosyl]-(1 / 4)-[3-
(l ¼ 254 nm) and staining with 10% phosphomolybdic acid in EtOH O-methyl-2-O-sulfonato-a-L-idopyranosyluronate]-(1 / 4)-
8
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

2,3,6-tri-O-sulfonato-a-D-glucopyranoside sodium salt (com- method (Thrombinoscope BV®, Maastricht, Netherlands). The total
pound 3): 1H NMR (800 MHz, D2O) d 5.59 (d, J ¼ 3.7 Hz, 1H), amount of thrombin generated was quantified by computing the
5.55e5.49 (m, 1H), 5.32 (s, 1H), 5.15 (d, J ¼ 3.6 Hz, 1H), 4.92 (s, 1H), area under the TG curve (AUC). The TGT was performed in normal
4.63 (ddd, J ¼ 36.0, 21.9, 9.4 Hz, 3H), 4.49e4.00 (m, 13H), 3.96e3.77 control plasma by the CAT method (ThrombinoscopeBV®, Maas-
(m, 4H), 3.66 (s, 4H), 3.62 (s, 3H), 3.62e3.59 (m, 5H), 3.56 (s, 2H), tricht, Netherlands), the thrombinoscope® software was used to
3.53 (t, J ¼ 8.1 Hz, 1H), 3.47 (s, 3H).13C NMR (201 MHz, D2O) construct the curve of time (min) versus thrombin concentration
d 104.53, 103.92, 101.77, 99.99, 99.25, 96.86, 88.57, 86.85, 84.76, (nM) and to calculate the TG parameters. The formation of this
82.80, 82.52, 82.41, 81.49, 79.85, 79.44, 79.40, 79.21, 79.09, 78.76, curve could be observed kinetically on the computer screen. The
78.53, 78.16, 78.04, 77.56, 76.98, 76.55, 76.49, 76.41, 76.15, 76.03, following TG parameters were measured and analyzed: (A) lag time
75.89, 75.83, 75.48, 74.47, 74.08, 72.78, 72.42, 72.38, 71.68, 71.27, (min), which is the time between addition of the trigger and the
70.93, 69.65, 69.08, 68.90, 68.76, 68.67, 63.32, 63.28, 63.20, 63.13, initiation of the TG; (B) peak (nM) refers to the maximum of
62.72, 61.96, 61.94, 60.10, 58.14, 49.53.MS(ESI):913.8[(M  2Na)/2]- thrombin concentration generated; (C) the endogenous thrombin
Methyl [2,6-di-O-sulfonato-a-D-glucopyranosyl]-(1 / 4)-[3- potential (ETP) (nM  min) represents the amount of thrombin
O-methyl-b-D-glucopyranosyluronate]-(1 / 4)-[2,3,6-tri-O-sul- formed over 60 min. The total amount of free thrombin produced is
fonato-a-D-glucopyranosyl]-(1 / 4)-[3-O-methyl-2-O-sulfo- referred to the area under the curve (AUC) or ETP, as described by
nato-a-L-idopyranosyluronate]-(1 / 4)-2,3,6-tri-O-sulfonato-a- Wolberg and Campbell [48].
1
D-glucopyranoside sodium salt (compound 4): H NMR (800 MHz,
D2O) d 5.62 (d, J ¼ 3.4 Hz, 1H), 5.53 (s, 1H), 5.15 (d, J ¼ 3.2 Hz, 2H), 4.6. Antithrombotic properties and bleeding effects in vivo
4.69e4.26 (m, 13H), 4.20e4.11 (m, 2H), 4.12e3.72 (m, 6H),
3.71e3.42 (m, 15H).13C NMR (201 MHz, D2O) d 103.87, 101.61, 4.6.1. Animals
100.04, 99.04, 96.70, 88.52, 84.63, 83.61, 82.69, 80.34, 80.09, 79.08, Sprague Dawley male rats (of body mass 250e300 g) from
79.05, 78.71, 78.55, 78.11, 77.63, 77.50, 77.31, 76.76, 76.44, 76.29, Hunan Shrek Jingda Experimental Animal Co., Ltd (Hunan, PR
75.74, 75.21, 74.97, 73.87, 73.04, 72.69, 72.47, 72.43, 71.82, 71.78, China) were used for animal experiments. Rats were caged and fed
71.52, 69.50, 68.98, 68.72, 68.65, 62.97, 61.99, 59.88, 58.13. MS(ESI): a regular diet for at least 1 week before use. These experiments
1904.6 [Mþ2Na-H]-. were reviewed and approved by the Animal Ethics Committee of
Kunming Institute of Botany, Chinese Academy of Sciences.
4.3. Determination of anticoagulant activities in vitro
4.6.2. Stasis-induced venous thrombosis
APTT, PT, and TT were determined with a coagulometer (TECO Thrombus formation was induced by promoting a combination
MC-4000, Germany) using APTT, PT and TT reagents and standard of stasis and hypercoagulability using a modification of the method
human plasma as previously described [39,46]. In brief, 5 mL sample of Vogel et al. [39,46,49]. Normal saline, compounds 1e4 and fon-
and 45 mL plasma were pipetted to cuvette; incubated at 37  C for daparinux sodium were administered dorsally and subcutaneously
2 min, then 50 mL APTT reagent was added; after incubating for at 100 nmol/kg. At the indicated time (1, 3, 12 and 24 h, respec-
3 min, 50 mL CaCl2 reagent was added, and clotting time was tively), rats were anesthetized by 10% chloral hydrate (0.3 mL/kg).
recorded. The compound plasma concentrations clotting times Two loose sutures were prepared about 2.0 cm apart on the inferior
were fitted linearly by Origin 2017 software (OriginLab, USA), and vena cava and all the collateral veins were ligated. Rabbit powder
the EC2.0 (concentration required for doubling clotting time) were leaching agent (2%) was injected intravenously. At a time of 20 s
calculated from the fitting curves. Determinations were performed following the injection, stasis was established by tightening the
in duplicate. two sutures: the proximal and then the distal. The abdominal cavity
was provisionally closed and stasis was maintained for 20 min. The
4.4. Inhibition of factor Xa by AT cavity was then reopened, the ligated segment was opened longi-
tudinally, and the thrombus formed was removed, rinsed, blotted
Antithrombin and anti-factor Xa activities in the presence of AT on filter paper, dried for 24 h at 50  C, and weighed.
were measured with Biophen Heparin Anti-IIIa kits and Biophen To test the dose-antithrombotic activity relationship, three
Antithrombin 2.5 kits. A mixture of 30 mL sample and 30 mL 1 IU/mL different doses 30, 100, 300 nmol/kg were administered for 2 h. For
AT was incubated at 37  C for 2 min. Then 30 mL of 24 NIH/mL each group (n ¼ 8), the mean thrombus mass was determined and
thrombin (or 8 mg/mL bovine factor Xa) was added. After incubation then expressed as a percentage of thrombus mass. The absence of
for 2 min (or 1 min for factor Xa), the residual thrombin or factor Xa any inhibition of thrombus formation (compared to the mean
activity was measured by the addition of 30 mL of 1.25 mM thrombus mass in the group administered normal saline) was
thrombin chromogenic substrate (CS-11(65)) or 1.2 mM factor Xa represented as 100%.
chromogenic substrate SXa-11. The absorbance of the reaction
mixture was read at 405 nm and were tested in twice. The reaction 4.7. Ex vivo anti FXa activities in rats
was stopped 7 min later. The percentage of inhibition was then
calculated as [% Inhibition ¼ 100  (Optical Density [OD] Blank - OD Three Sprague-Dawley rats were anesthetized by chloral hy-
Sample)/OD Blank]. The activity per milligram of each compound drate (10%, 0.3 mL/kg; intraperitoneal). Arterial blood samples
was determined by comparison with a calibrated standard (FXa) (4 mL) were withdrawn in a 3.8% trisodium citrate solution (1/9, v/
using an Origin 2017 progress function (OriginLab, USA). v). Each blood sample was centrifuged (1800 g  10 min) and the
platelet-poor plasmas were mixed in equal volume. Gradient con-
4.5. Thrombin generation assay centration of Fpx or compounds 1e4 were mixed with plasmas (1/
9, v/v), then the anti FXa activities of mixtures were measured and
The thrombin generation (TG) methodology was adapted from prepared the standard curve.
Hemker as previously reported [47]. TG was triggered by PPP- Sprague-Dawley rats were randomly divided into 6 groups
Reagent contains a mixture of phospholipids and tissue Factor in (n ¼ 5e8% group), and treated by subcutaneous of Fpx or com-
human platelet-poor plasma. Aliquots were taken every 15 s and pounds 1e4 at 100 nmol/kg, and the normal saline was injected as
the concentration of thrombin was measured using the CAT control. The venous blood (100 mL/time) were harvested in 15 min
9
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

before injection, and 30 min, 1 h, 2 h, 3 h, 4 h, 6 h, 8 h, 10 h, 24 h, 2864e2872.


[9] T.E. Warkentin, M.N. Levine, J. Hirsh, P. Horsewood, R.S. Roberts, M. Gent,
32 h after injection. The anti-FXa activities were examined and each
J.G. Kelton, Heparin-induced thrombocytopenia in patients treated with low-
concentration of compounds in the blood was calculated according molecular-weight heparin or unfractionated heparin, N. Engl. J. Med. 332
to the preceding curve. (1995) 1330e1336.
[10] M. Petitou, C.A. van Boeckel, A synthetic antithrombin III binding penta-
saccharide is now a drug! what comes next? Angew. Chem. Int. Ed. Engl. 43
4.8. Statistical analysis (2004) 3118e3133.
[11] J.W. Cheng, Fondaparinux: a new antithrombotic agent, Clin. Therapeut. 24
(2002) 1757e1769.
The results of the coagulation assays were calculated as sigmoid
[12] D.B. lossom, A.J. Kallen, P.R. Patel, A. Elward, L. Robinson, G. Gao, R. Langer,
or linear regressions using an Origin 2017 progress function (Ori- K.M. Perkins, J.L. Jaeger, K.M. Kurkjian, M. Jones, S.F. Schillie, N. Shehab,
ginLab, USA). The results of the venous thrombosis and bleeding D. Ketterer, G. Venkataraman, T.K. Kishimoto, Z. Shriver, A.W. McMahon,
effect experiments were expressed as means ± SD and were K.F. Austen, S. Kozlowski, A. Srinivasan, G. Turabelidze, C.V. Gould,
M.J. Arduino, R.N. Sasisekharan, Outbreak of adverse reactions associated with
analyzed by SPSS statistics version 16.0 (IBM, USA), and the sta- contaminated heparin, N. Engl. J. Med. 359 (2008) 2674.
tistical significance of results was determined using one-way [13] M.D. Kort, R.C. Buijsman, C.A.A.V. Boeckel, Synthetic heparin derivatives as
analysis of variance (ANOVA), followed by Levene's test or the new anticoagulant drugs, Drug Discov. Today 10 (2005) 769e779.
[14] M. Petitou, J.C. Jacquinet, P. Sinay, J. Choay, J.C. Lormeau, M. Nassr, Process for
chi-square test. Data were considered significantly different when: the Organic Synthesis of Oligosaccharides and Derivatives Thereof, U.S. Patent,
*p < 0.05, **p < 0.01 or ***p < 0.001. 1989, 4818816.
[15] S. Dey, H.J. Lo, C.H. Wong, Programmable one-pot synthesis of heparin pen-
tasaccharide fondaparinux, Org. Lett. 22 (2020) 4638e4642.
Credit author contribution statement [16] S. Arungundram, K. Al-Mafraji, J. Asong, F.E. Leach, I.J. Amster, A. Venot,
J.E. Turnbull, G.J. Boons, Modular synthesis of heparan sulfate oligosaccharides
Zhipeng Zhou: Data curation, Investigation, Writing-Original for structure-activity relationship studies, J. Am. Chem. Soc. 131 (2009)
17394e17405.
draft; Linlin Zhang: Methodology, Data curation, Writing- [17] G. Tiruchinapally, Z.J. Yin, M. El-Dakdouki, Z. Wang, X.F. Huang, Divergent
Reviewing and Editing; Xin Wu:Data curation, Investigation, heparin oligosaccharide synthesis with preinstalled sulfate esters, Chem. Eur J.
Writing-Reviewing and Editing; Lan Luo: Investigation, Writing- 17 (2011) 10106e10112.
[18] J.C. Lee, X.A. Lu, S.S. Kulkarni, Y.S. Wen, S.C. Hung, Synthesis of heparin oli-
Reviewing and Editing; Jian Wu: Investigation; Dan Xu and Min- gosaccharides, J. Am. Chem. Soc. 126 (2004) 476e477.
gyi Wu: Conceptualization, Supervision, Writing-Reviewing and [19] C.H. Chang, L.S. Lico, T.Y. Huang, S.Y. Lin, C.L. Chang, S.D. Arco, S.C. Hung,
Editing, Funding acquisition, Data curation. Synthesis of the heparin-based anticoagulant drug fondaparinux, Angew.
Chem. Int. Ed. Engl. 53 (2014) 9876e9879.
[20] Z.Q. Zhang, S.A. McCallum, J. Xie, L. Nieto, F. Corzana, J. Jimenez-Barbero,
Declaration of competing interest M. Chen, J. Liu, R.J. Linhardt, Solution structures of chemoenzymatically syn-
thesized heparin and its precursors, J. Am. Chem. Soc. 130 (2008) 12998.
[21] Y. Xu, S. Masuko, M. Takieddin, H. Xu, R. Liu, J. Jing, S.A. Mousa, R.J. Linhardt,
The authors declare that they have no known competing J. Liu, Chemoenzymatic synthesis of homogeneous ultralow molecular weight
financial interests or personal relationships that could have heparins, Science 334 (2011) 498e501.
appeared to influence the work reported in this paper. [22] M. Petitou, J.P. He rault, A. Bernat, P.A. Driguez, P. Ducjaussoy, J.C. Lormeau,
J.M. Herbert, Synthesis of thrombin-inhibiting heparin mimetics without side
effects, Nature 398 (1999) 417e422.
Acknowledgements [23] X. Dai, W. Liu, Q. Zhou, C. Cheng, C. Yang, S. Wang, M. Zhang, P. Tang, H. Song,
D. Zhang, Y. Qin, Formal synthesis of anticoagulant drug fondaparinux so-
dium, J. Org. Chem. 81 (2016) 162e184.
This work was supported in part by the National Natural Science [24] J.L. de Paz, C. Noti, P.H. Seeberger, Microarrays of synthetic heparin oligo-
Foundation of China (NO. 81872774 and 81930097), Youth Inno- saccharides, J. Am. Chem. Soc. 128 (2006) 2766e2767.
vation Promotion Association CAS (Y2021104), CAS "Light of West [25] C. Noti, J.L. de Paz, L. Polito, P.H. Seeberger, Preparation and use of microarrays
containing synthetic heparin oligosaccharides for the rapid analysis of
China" Program and the Ten-thousand Talents Program of Yunnan heparin-protein interactions, Chem. Eur J. 12 (2006) 8664e8686.
Province (YNWR-QNBJ-2018-271), and Yunnan Fundamental [26] T. Polat, C.H. Wong, Anomeric reactivity-based one-pot synthesis of heparin-
Research Projects (grant NO. 202101AS070292 and like oligosaccharides, J. Am. Chem. Soc. 129 (2007) 12795e12800.
[27] C. Chen, B. Yu, Efficient synthesis of Idraparinux, the anticoagulant penta-
202101AT070152).
saccharide, Bioorg. Med. Chem. Lett 19 (2009) 3875e3879.
[28] P. Westerduin, C.A. van Boeckel, J.E. Basten, M.A. Broekhoven, H. Lucas,
Appendix B. Supplementary data A. Rood, H. van der Heijden, R.G. van Amsterdam, T.G. van Dinther,
D.G. Meuleman, et al., Feasible synthesis and biological properties of six 'non-
glycosamino' glycan analogues of the antithrombin III binding heparin pen-
Supplementary data to this article can be found online at tasaccharide, Bioorg. Med. Chem. 2 (1994) 1267e1280.
https://doi.org/10.1016/j.ejmech.2022.114256. [29] P. Westerduin, J.E. Basten, M.A. Broekhoven, V. Kimpe, W.H. Kuijpers,
C.A. Boeckel, Synthesis of tailor made glycoconjugates showing AT III-
mediated inhibition of blood coagulation factors Xa and thrombin, Angew.
References Chem. Int. Ed. Engl. 35 (1996) 331e333.
[30] M. Herczeg, E. Mezo } , L. La
z € ve
ar, A. Fekete, K.E. Ko r, S. Antus, A. Borb
as, Novel
[1] A.M. Wendelboe, G.E. Raskob, Global burden of thrombosis, Circ. Res. 118 syntheses of Idraparinux, the anticoagulant pentasaccharide with indirect
(2016) 1340e1347. selective factor Xa inhibitory activity, Tetrahedron 69 (2013) 3149e3158.
[2] N. Mackman, Triggers, targets and treatments for thrombosis, Nature 451 [31] M. Herczeg, E. Mezo } , D. Eszenyi, S. Antus, A. Borb as, New synthesis of idra-
(2008) 914e918. parinux, the non-glycosaminoglycan analogue of the antithrombin-binding
[3] J.I. Weitz, J. Harenberg, New developments in anticoagulants: past, present, domain of heparin, Tetrahedron 70 (2014) 2919e2927.
and future, Thromb. Haemostasis 117 (2017) 1283e1288. [32] G. Łopatkiewicz, S. Buda, J. Mlynarski, Application of the EF and GH fragments
[4] G. Spadarella, A.D. Minnob, M.B. Donatic, M. Mormiled, I. Ventred, to the synthesis of Idraparinux, J. Org. Chem. 82 (2017) 12701e12714.
G.D. Minnod, From unfractionated heparin to pentasaccharide: paradigm of [33] F. Demeter, F. Veres, M. Herczeg, A. Borba s, Short synthesis of Idraparinux by
rigorous science growing in the understanding of the in vivo thrombin gen- applying a 2-O-methyl 4, 6-O-arylmethylene thioidoside as a 1, 2-trans-a-
eration, Blood Rev. 39 (2020) 100613. selective glycosyl donor, Eur. J. Org Chem. 48 (2018) 6901e6912.
[5] S.N. Baytas, R.J. Linhardt, Advances in the preparation and synthesis of heparin [34] J.M. Herbert, J.P. Herault, A. Bernat, R. Amsterdam, J.C. Lormeau, M. Petitou,
and related products, Drug Discov. Today 25 (2020) 2095e2109. C. van Boeckel, P. Hoffmann, D.G. Meuleman, Biochemical and pharmacolog-
[6] N. Mackman, New insights into the mechanisms of venous thrombosis, J. Clin. ical properties of SANORG 34006, a potent and long-acting synthetic penta-
Invest. 122 (2012) 2331e2336. saccharide, Blood 91 (1998) 4197e4205.
[7] D.J. Quinlan, A. McQuillan, J.W. Eikelboom, Low-molecular-weight heparin [35] The Persist Investigators, A novel long-acting synthetic factor Xa inhibitor
compared with intravenous unfractionated heparin for the treatment of (SanOrg34006) to replace warfarin for secondary prevention in deep vein
pulmonary embolism: a meta-analysis of randomized, controlled trials, Ann. thrombosis. A phase II evaluation, J. Thromb. Haemostasis 2 (2004) 47e53.
Intern. Med. 140 (2004) 175e183. [36] S. Apostolakis, Y. Guo, D.A. Lane, H. Buller, G.Y. Lip, Renal function and out-
[8] G.M. Arepally, Heparin-induced thrombocytopenia, Blood 129 (2017) comes in anticoagulated patients with non-valvular atrial fibrillation: the

10
Z. Zhou, L. Zhang, X. Wu et al. European Journal of Medicinal Chemistry 234 (2022) 114256

AMADEUS trial, Eur. Heart J. 34 (2013) 3572e3579. [44] X. Zhao, C. Gao, Y. Chu, L. Yang, X. Yang, W. Xu, W. He, P. Zhang, X. Liu, L. Tian,
[37] E. Caputo, E. Straub, W. Grinstaff, Design, synthesis, and biomedical applica- Fondaparinux vs. enoxaparin in patients with non-ST elevation acute coro-
tions of synthetic sulphated polysaccharides, Chem. Soc. Rev. 48 (2019) nary syndromes (NSTE-ACS) treated with percutaneous coronary intervention
2338e2365. and tirofiban: an exploratory study in China, J. Clin. Pharm. Therapeut. 40
[38] C.H. Chang, L.S. Lico, T.Y. Huang, S.Y. Lin, C.L. Chang, S.D. Arco, S.C. Hung, (2015) 584e589.
Synthesis of the heparin-based anticoagulant drug fondaparinux, Angew. [45] I. Fifth, , Organization to Assess Strategies in Acute Ischemic Syndromes,
Chem. Int. Ed. Engl. 53 (2014) 9876e9879. S. Yusuf, S.R. Mehta, S. Chrolavicius, R. Afzal, J. Pogue, C.B. Granger, A. Budaj,
[39] M. Wu, D. Wen, N. Gao, C. Xiao, L. Yang, L. Xu, W. Lian, W. Peng, J. Jiang, J. Zhao, R.J. Peters, J.P. Bassand, L. Wallentin, C. Joyner, K.A. Fox, Comparison of fon-
Anticoagulant and antithrombotic evaluation of native fucosylated chon- daparinux and enoxaparin in acute coronary syndromes, N. Engl. J. Med. 354
droitin sulfates and their derivatives as selective inhibitors of intrinsic factor (2006) 1464e1476.
Xase, Eur. J. Med. Chem. 92 (2015) 257e269. [46] L. Zhao, M. Wu, C. Xiao, L. Yang, W. Lian, L. Zhou, N. Gao, Z. Li, J. Chen, J. Chen,
[40] L. Lin, L. Zhao, N. Gao, R. Yin, S. Li, H. Sun, L. Zhou, G. Zhao, S.W. Purcell, J. Zhao, J. Liu, H. Qin, J. Zhao, Discovery of an intrinsic tenase complex inhibitor: pure
From multi-target anticoagulants to DOACs, and intrinsic coagulation factor nonasaccharide from fucosylated glycosaminoglycan, Proc. Natl. Acad. Sci. U.
inhibitors, Blood Rev. 39 (2020) 100615. S. A. 112 (2015) 8284e8289.
[41] G. Zhang, H. Jin, Y. Zhao, L. Guo, X. Gao, X. Wang, S. Tie, J. Shen, P.G. Wang, [47] H.C. Hemker, P. Giesen, R. Al Dieri, V. Regnault, E. de Smedt, R. Wagenvoord,
H. Gan, H. Cui, W. Zhao, An efficient anticoagulant candidate: characterization, T. Lecompte, S. Beguin, Calibrated automated thrombin generation measure-
synthesis and in vivo study of a fondaparinux analogue Rrt1.17, Eur. J. Med. ment in clotting plasma, Pathophysiol. Haemostasis Thrombosis 33 (2003)
Chem. 126 (2017) 1039e1055. 4e15.
[42] H. Sun, Na Gao, L. Ren, S. Liu, L. Lin, W. Zheng, L. Zhou, R. Yin, J. Zhao, The [48] A.S. Wolberg, R.A. Campbell, Thrombin generation, fibrin clot formation and
components and activities analysis of a novel anticoagulant candidate dHG-5, hemostasis, Transfus, Apher. Sci. 38 (2008) 15e23.
Eur. J. Med. Chem. 207 (2020) 112796. [49] G.M. Vogel, D.G. Meuleman, F.G. Bourgondien, P.M. Hobbelen, Comparison of
[43] N. Mackman, W. Bergmeier, G.A. Stouffer, J.I. Weitz, Therapeutic strategies for two experimental thrombosis models in rats effects of four glycosaminogly-
thrombosis: new targets and approaches, Nat. Rev. Drug Discov. 19 (2020) cans, Thromb. Res. 54 (1989) 399e410.
333e352.

11

You might also like