yingweiwo

Fondaparinux (Heparin pentasaccharide)

Alias: Fondaparinux free acid; Fondaparinux; 104993-28-4; Natural heparin pentasaccharide; Arixtra; CHEBI:61033; UNII-J177FOW5JL; J177FOW5JL; HSDB 7845;
Heparin pentasaccharide (Fondaparinux) is a chemically synthesized selective factor Xa inhibitor with anticoagulant activity.
Fondaparinux (Heparin pentasaccharide)
Fondaparinux (Heparin pentasaccharide) Chemical Structure CAS No.: 104993-28-4
Product category: Factor Xa
This product is for research use only, not for human use. We do not sell to patients.
Size Price
500mg
1g
Other Sizes
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text

 

  • Business Relationship with 5000+ Clients Globally
  • Major Universities, Research Institutions, Biotech & Pharma
  • Citations by Top Journals: Nature, Cell, Science, etc.
Top Publications Citing lnvivochem Products
Product Description
Heparin pentasaccharide (Fondaparinux) is a chemically synthesized, selective factor Xa inhibitor with anticoagulant activity. Heparin pentasaccharide can be used in studies related to venous thromboembolic events.
Biological Activity I Assay Protocols (From Reference)
Targets
Factor Xa
ln Vitro
The first novel anticoagulant that targets factor Xa specifically is fondaparinux sodium. The IC50 value (anti-Xa IU/ml) for fondaparinux is 0.59±0.05 for activated monocytes (ac-M) and 0.17±0.03 for MMPs (monocyte-derived microparticles) [2].
Effect of Fondaparinux, low molecular weight heparin and unfractionated heparin on thrombin generation supported by activated monocytes [2]
Values obtained at baseline were 2.1 ± 0.1 min for lag time, 181 ± 6 nmol/l for peak, 1624 ± 84 nmol/l min for ETP and 59 ± 4 nmol/l/min for rate index. Fondaparinux, enoxaparin and UFH inhibited ac-M-supported thrombin generation in a concentration-dependent manner (Fig. 1). Except for the lag time, the inhibitory effect of UFH was superior to enoxaparin, which was superior to fondaparinux, from 0.3 to 1.0 anti-Xa IU/ml. Whereas comparable inhibitory effect of fondaparinux and enoxaparin on peak and ETP was noted at the lowest anti-Xa concentration (0.1 IU/ml), the inhibitory effect on rate index of fondaparinux was more pronounced than that of enoxaparin at 0.1 IU/ml anti-Xa concentration.
Calculation of IC50 [2]
The inhibitory effect on thrombin generation was evaluated by calculating the inhibitory concentration (IC)50, which was defined for peak, ETP and rate index as the drug concentration yielding 50% reduction, and for lag time as the drug concentration resulting in doubling it. UFH was the only drug for which IC50 was calculated for all thrombin generation parameters in the two models. In contrast, enoxaparin IC50 values were calculated for ETP, peak and rate index, but not for lag time, in the two models. Furthermore, IC50 for Fondaparinux was reached only for rate index in the two models and for peak and ETP only in the MMP model. Rate index was the only parameter for which IC50 was reached in both models for the three drugs; UFH was the most effective, followed by enoxaparin and fondaparinux (Table 1).
ln Vivo
The pharmacokinetics of fondaparinux sodium are linear and dose-dependent, resulting in a very predictable response. With a quick start of action, a half-life of 14 to 16 hours, and continuous antithrombotic activity for 24 hours, fondaparinux sodium is 100% bioavailable. The medication has no effect on platelet function or aggregation, prothrombin time, or activated partial thromboplastin time [1].
Compared with the ex-vivo anti-Xa levels reported in Organization to Assess Strategies in Acute Ischemic Syndromes (OASIS)-5 substudy, our results demonstrate that anti-Xa values obtained in patients are either similar or even higher than those required in our in-vitro models used to reach rate index IC50. Fondaparinux anti-Xa level of 0.52 (±0.22) IU/ml in OASIS-5 study was sufficient to inhibit rate index in both in-vitro models tested. For enoxaparin, the anti-Xa level measured in OASIS-5 substudy patients was 1.2 (±0.45) IU/ml, which was markedly more superior than anti-Xa concentrations required to inhibit rate index in the two models of thrombin generation tested here (ac-M IC50: 0.27 ± 0.03 IU/ml, and MMPs IC50 0.19 ± 0.02 IU/ml).

The OASIS-5 substudy reported less inhibition of ETP in Fondaparinux-treated patients versus enoxaparin. Our in-vitro ETP results are in agreement with these data, taking into consideration that the Oasis-5 substudy ETP was performed in cell-free assay. Fondaparinux yielded lower inhibition than enoxaparin in both models, with anti-Xa concentrations ranging from 0.3 to 1.0 IU/ml. Fondaparinux ETP IC50 values were not always reached despite increased anti-Xa levels (fondaparinux tested at concentrations ranging from 0.1 to 1.0 anti-Xa IU/ml), and its ETP IC50 was reached only in the MMPs model. In contrast, enoxaparin reached ETP IC50 in the two in-vitro models tested here. As for rate index, the levels of anticoagulation in OASIS-5 enoxaparin-treated patients are much more superior to those required to inhibit ETP in our cell models.

Fondaparinux at 2.5 mg daily (corresponding to anti-Xa of 0.52 ± 0.22 IU/ml) in the Oasis-5 study, showed an optimal efficacy safety in ACS patients. This anti-Xa level for fondaparinux corresponded to the rate index IC50 of the ac-M model and to the ETP IC50 value of the MMP model. At this anti-Xa concentration, rate index was inhibited in the MMP model and ETP IC50 was not attained in ac-M model. For enoxaparin, rate index IC50 in ac-M model was similar to ETP IC50 in MMP model and was closely equal to 0.3 IU/ml. At this anti-Xa level, rate index in MMP model was largely inhibited and ETP IC50 in ac-M was not reached. Given this, it is assumed that an anti-Xa level of about 0.3 IU/ml for enoxaparin is sufficient in ACS for an optimal efficacy-safety profile. It should be noted that, of the three agents analysed here, fondaparinux is the only agent for which the daily dose has been rationally determined, as evidenced by the use of 2.5 mg dose on the basis of previous dose-finding studies [2].
Cell Assay
Monocyte activation [2]
Purified monocytes were washed in RPMI-1640 medium and then adjusted to 1.5 106 cells/ml in RPMI-1640 containing 5% (v/v) heat-inactivated foetal calf serum, 2 mmol/l L-glutamine and 100 ng/ml LPS (Escherichia coli serotype O55:B5, Sigma). Following incubation of the monocytes for 5 h at 37°C in a 5% CO2 humidified atmosphere, supernatants were removed by centrifugation for 5 min at 400g. The ac-M-containing pellet was resuspended in 150 μl phosphate-buffered saline.
Monocyte-derived microparticles preparation and quantification [2]
Monocytes were incubated for 18 h, as described above, and supernatants were collected by centrifugation for 5 min at 2200g. MMPs were recovered following an additional centrifugation at 17000g for 30 min and quantified by flow cytometry [5]. MMPs (100 μl) were incubated for 15 min in the dark with 1 μl annexin V-FITC, according to manufacturer's recommendations, followed by the addition of 400 μl annexin V-binding buffer and 100 μl flow-count Fluorospheres. Fluorescence was acquired for 60 s on Epics XL-MCL flow cytometer, using System 2 software. MMPs were quantified using the flow-count Fluorospheres and expressed as MMPs/μl.
Fluorogenic measurement of thrombin generation [2]
Thrombin generation test was performed according to the assay described by Hemker and modified by Poitevin; ac-Ms and MMPs being defined as cell models. PPP used for all experimental conditions was systematically supplemented with aprotinin at 200 kallikrein inhibitory units (KUI)/ml final concentration. PPP was spiked with increasing concentrations of Fondaparinux, LMWH or UFH; final concentrations being 0.0, 0.1, 0.3, 0.6 and 1.0 anti-FXa IU/ml. We previously optimized the concentrations of ac-Ms and MMPs included in the assay. Thus, 20 μl of ac-M (0.2 × 106 ac-M per well) or of MMPs (160 000 MMPs per well) was added to 80 μl of PPP. Results were expressed as percentage, taking the baseline measurements as 100%.

Fluorometric determination of thrombin generation was done on Fluoroskan Ascent plate reader, following addition of the fluorogenic substrate, Z-Gly-Gly-Arg-AMC; Thrombinoscope software used for calculating thrombin generation. Four parameters of the thrombin generation curve were analysed: lag time (min), thrombin peak (peak, nmol/l), endogenous thrombin potential (ETP, nmol/l min), rate index of propagation phase defined by the formula: peak/(time to peak – lag-time) (rate index, nmol/l/min).
Animal Protocol
Platelet-poor plasma preparation [2]
Venous blood samples were obtained from five healthy volunteers (mean age 25 ± 2 years). Volunteers were laboratory staff members and did not receive any medication for the last 2 weeks. Informed consents were obtained from all of them. Blood was withdrawn by antecubital venipuncture into Monovette tubes (0.106 mol/l citrate). A three-step centrifugation procedure including 10 min at 190g, 10 min at 1750g and 30 min at 13 000g was used. Platelet-poor plasma (PPP) supernatants were pooled, stored at −80°C and thawed immediately before use.
Monocyte purification [2]
Cytapheresis material was obtained from six healthy volunteers who were admitted for platelet donation in the blood transfusion unit of CHU Robert Debré. Informed consent was obtained from the participants. Monocytes were purified from cytapheresis residues by elutriation, as previously described. Monocyte purity was assessed by CD14 staining of isolated cells (>95% CD14-positive). Cell viability (>98%) was determined by the trypan blue exclusion principle. Purified monocytes were used for preparing ac-Ms and MMPs.
ADME/Pharmacokinetics
The pharmacology and mechanism of action of fondaparinux sodium are described. Fondaparinux sodium is the first agent of a new class of anticoagulants that selectively target factor Xa. It has a linear, dose-dependent pharmacokinetic profile, which provides a highly predictable response. It is 100% bioavailable, has a rapid onset of action, and has a half-life of 14 to 16 hours, allowing for sustained antithrombotic activity over a 24-hour period. The drug does not affect prothrombin time or activated partial thromboplastin time, nor does it affect platelet function or aggregation. Studies in patients with confirmed heparin-induced thrombocytopenia demonstrate that the drug is not associated with in vitro cross-reactivity to heparin antibodies. Fondaparinux sodium appears to meet the criteria for an ideal antithrombotic agent: equal or better effectiveness than currently available agents, a low bleeding risk, no need for laboratory monitoring, and once-daily administration.[1]
Toxicity/Toxicokinetics
Hepatotoxicity
Fondaparinux therapy is associated with a low rate of serum aminotransferase elevations during therapy, with levels above 3 times the upper limit of normal occurring in 1% to 3% of patients, a lower rate than occurs with heparin (~8%) or low molecular weight heparins (4% to 12%) but higher than occurs with placebo (
Likelihood score: E (unlikely cause of clinically apparent liver injury).
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation
Fondaparinux is considered to be acceptable to use during breastfeeding.

◉ Effects in Breastfed Infants
Relevant published information was not found as of the revision date.

◉ Effects on Lactation and Breastmilk
Relevant published information was not found as of the revision date.
Interactions
To investigate the in vitro metabolism of the antithrombotic agent fondaparinux sodium in mammalian liver fractions and to evaluate its potential inhibitory effect on human cytochrome P450 (CYP)-mediated metabolism of other drugs. Metabolism was evaluated by incubating radioisotope-labelled fondaparinux sodium with postmitochondrial liver fractions of rat, rabbit, monkey or human origin (three subjects). Human liver microsomal preparations and an NADPH-generating system were incubated with phenacetin, coumarin, tolbutamide, S-mephenytoin, bufuralol, chlorzoxazone or nifedipine. These are selectively metabolised by CYP isoforms: CYP1A2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1 or CYP3A4, respectively. Experiments were designed to determine apparent K(i) (inhibitory constant) values for fondaparinux sodium against each CYP isoform, by varying concentrations of fondaparinux sodium and the selective substrate. Each experiment included control reaction mixtures containing an isoform-selective inhibitor. After incubation, the mixtures were analysed by LC-MS/MS or with fluorometric detection. All liver fractions were enzymatically active, as demonstrated by degradation of [(14)C]testosterone. No metabolism of fondaparinux sodium was detectable in postmitochondrial liver fractions. Apparent K(i) values for fondaparinux sodium against the CYP isoforms could not be determined because the oxidative metabolism of the isoform-selective CYP substrates was not significantly inhibited in pooled microsomal reaction mixtures. In the presence of selective CYP inhibitors, metabolism of each substrate was significantly reduced, confirming that inhibition could be observed in these assays. The demonstrated lack of mammalian hepatic metabolism of fondaparinux sodium is consistent with animal and human studies. The absence of inhibition of the human CYP isoforms commonly involved in the metabolism of drugs suggests that clinical treatment with fondaparinux sodium is unlikely to interfere with the pharmacokinetics and metabolism of a wide range of other drugs which are associated with CYP inhibition. PMID:12383041

... Two separate studies assessed any possible interaction between fondaparinux sodium at steady state and aspirin (acetylsalicylic acid) or piroxicam in healthy volunteers. In the first study a single dose of aspirin 975mg was assessed initially, followed by single doses of aspirin or placebo on the fourth day of an 8-day regimen of subcutaneous fondaparinux sodium (10mg once daily). The second study was a three-way crossover, double-blind, randomised study which investigated fondaparinux sodium 10mg + placebo, fondaparinux sodium 10mg + piroxicam 20mg, or placebo + piroxicam 20mg. ... Neither aspirin nor piroxicam influenced the pharmacokinetics of fondaparinux sodium at steady state. Two hours after administration, prolongation of bleeding time with aspirin alone or with aspirin plus fondaparinux sodium was significantly greater than with fondaparinux sodium alone (p = 0.003 and p = 0.004, respectively). No significant differences were observed between aspirin alone or aspirin + fondaparinux sodium in effect on bleeding time. A small decrease in collagen-induced platelet aggregation was observed after administration of piroxicam alone or piroxicam + fondaparinux sodium. A small effect on aPTT was observed; it was similar for fondaparinux sodium whether administered alone or in combination with either aspirin or piroxicam. No serious adverse events were reported. PMID:12383043

... In this study /investigators/ assessed the possible pharmacokinetic and pharmacodynamic interaction of fondaparinux sodium with digoxin at steady state in healthy male volunteers. In a phase I randomised, crossover study, volunteers (n = 24) were treated in two periods. The first period was once-daily administration of fondaparinux sodium 10mg subcutaneously alone for 7 days; the second period was 7 days of digoxin 0.25mg orally alone followed by 7 days of coadministration with fondaparinux sodium 10mg. Each period was separated by a washout of 12 days. ... The pharmacokinetic profiles of both digoxin and fondaparinux sodium were unaffected by coadministration. Bioequivalence was concluded, based on the 90% confidence intervals of the ratio of adjusted geometric means calculated for the 2-by-2 comparison of peak concentration, area under the concentration-time curve and cumulative urinary excretion, which lay within the 0.80 to 1.25 reference interval. There were no clinically significant fluctuations in vital signs and ECG parameters. The coadministration of digoxin with fondaparinux sodium was well tolerated and no significant changes were observed in vital signs.
Protein Binding
94% in vitro protein binding specifically to ATIII
References

[1]. Fondaparinux sodium: a selective inhibitor of factor Xa. Am J Health Syst Pharm. 2001 Nov 1;58 Suppl 2:S14-7.

[2]. Differential coagulation inhibitory effect of fondaparinux, enoxaparin and unfractionated heparin in cell models of thrombin generation. Blood Coagul Fibrinolysis. 2011 Jul;22(5):369-73.

[3]. Fondaparinux Sodium: Recent Advances in the Management of Thrombosis. J Cardiovasc Pharmacol Ther. 2023 Jan-Dec;28:10742484221145010.

Additional Infomation
Fondaparinux is a synthetic pentasaccharide which, apart from the O-methyl group at the reducing end of the molecule, consists of monomeric sugar units which are identical to a sequence of five monomeric sugar units that can be isolated after either chemical or enzymatic cleavage of the polymeric glycosaminoglycans heparin and heparan sulfate. It has a role as an anticoagulant. It is an amino sugar, an oligosaccharide sulfate and a pentasaccharide derivative. It is functionally related to a normethylfondaparinux. It is a conjugate acid of a fondaparinux(10-).
Fondaparinux (Arixtra) is a synthetic anticoagulant agent consisting of five monomeric sugar units and a O-methyl group at the reducing end of the molecule. It is structurally similar to polymeric glycosaminoglycan heparin and heparan sulfate (HS) when they are cleaved into monomeric units. The monomeric sequence in heparin and HS is thought to form the high affinity binding site for the natural anti-coagulant factor, antithrombin III (ATIII). Once bound to heparin or HS, the anticoagulant activity of ATIII is potentiated by 1000-fold. Fondaparinux potentiates the neutralizing action of ATIII on activated Factor X 300-fold. Fondaparinux may be used: to prevent venous thromboembolism in patients who have undergone orthopedic surgery of the lower limbs (e.g. hip fracture, hip replacement and knee surgery); to prevent VTE in patients undergoing abdominal surgery who are are at high risk of thromboembolic complications; in the treatment of deep vein thrombosis (DVT) and pumonary embolism (PE); in the management of unstable angina (UA) and non-ST segment elevation myocardial infarction (NSTEMI); and in the management of ST segment elevation myocardial infarction (STEMI).

Fondaparinux is a Factor Xa Inhibitor. The mechanism of action of fondaparinux is as a Factor Xa Inhibitor.
Fondaparinux is a synthetic inhibitor of factor Xa which given by injection and is used as an anticoagulant to treat as well as prevent venous thromboembolism. Fondaparinux has been associated with a low rate of serum aminotransferase elevations during therapy, but has not been implicated in cases of clinically apparent, idiosyncratic liver injury.
Fondaparinux is a synthetic glucopyranoside with antithrombotic activity. Fondaparinux selectively binds to antithrombin III, thereby potentiating the innate neutralization of activated factor X (Factor Xa) by antithrombin. Neutralization of Factor Xa inhibits its activity and interrupts the blood coagulation cascade, thereby preventing thrombin formation and thrombus development.
FONDAPARINUX is a Oligosaccharide drug with a maximum clinical trial phase of IV (across all indications) that was first approved in 2001 and has 6 approved and 12 investigational indications. This drug has a black box warning from the FDA.
Synthetic pentasaccharide that mediates the interaction of HEPARIN with ANTITHROMBINS and inhibits FACTOR Xa; it is used for prevention of VENOUS THROMBOEMBOLISM after surgery.
Fondaparinux Sodium is the sodium salt form of fondaparinux, a synthetic glucopyranoside with antithrombotic activity. Fondaparinux sodium selectively binds to antithrombin III, thereby potentiating the innate neutralization of activated factor X (Factor Xa) by antithrombin. Neutralization of Factor Xa inhibits its activity and interrupts the blood coagulation cascade, thereby preventing thrombin formation and thrombus development. (NCI05)
Synthetic pentasaccharide that mediates the interaction of HEPARIN with ANTITHROMBINS and inhibits FACTOR Xa; it is used for prevention of VENOUS THROMBOEMBOLISM after surgery.
See also: Fondaparinux (has active moiety).
Drug Indication
1. 5-mg/0. 3-ml and 2. 5-mg/0. 5-ml solution for injectionPrevention of venous thromboembolic events (VTE) in adults undergoing major orthopaedic surgery of the lower limbs such as hip fracture, major knee surgery or hip-replacement surgery. Prevention of VTE in adults undergoing abdominal surgery who are judged to be at high risk of thromboembolic complications, such as patients undergoing abdominal cancer surgery. Prevention of VTE in adult medical patients who are judged to be at high risk for VTE and who are immobilised due to acute illness such as cardiac insufficiency and / or acute respiratory disorders, and / or acute infectious or inflammatory disease. Treatment of adults with acute symptomatic spontaneous superficial-vein thrombosis of the lower limbs without concomitant deep-vein thrombosis. 2. 5-mg/0. 5-ml solution for injectionTreatment of unstable angina or non-ST-segment-elevation myocardial infarction (UA/NSTEMI) in adult patients for whom urgent (< 120 mins) invasive management (PCI) is not indicated. infarction (STEMI) in adult patients who are managed with thrombolytics or who initially are to receive no other form of reperfusion therapy. 5-mg/0. 4-ml, 7. 5-mg/0. 6-ml and 10-mg/0. 8-ml solution for injectionTreatment of adults with acute deep-vein thrombosis (DVT) and treatment of acute pulmonary embolism (PE), except in haemodynamically unstable patients or patients who require thrombolysis or pulmonary embolectomy.
1. 5 mg/0. 3 ml and 2. 5 mg/0. 5 ml, solution for injection: , Prevention of Venous Thromboembolic Events (VTE) in patients undergoing major orthopaedic surgery of the lower limbs such as hip fracture, major knee surgery or hip replacement surgery. , Prevention of Venous Thromboembolic Events (VTE) in patients undergoing abdominal surgery who are judged to be at high risk of thromboembolic complications, such as patients undergoing abdominal cancer surgery (see section 5. 1). , Prevention of Venous Thromboembolic Events (VTE) in medical patients who are judged to be at high risk for VTE and who are immobilised due to acute illness such as cardiac insufficiency and/or acute respiratory disorders, and/or acute infectious or inflammatory disease. , , 2. 5 mg/0. 5 ml, solution for injection: , Treatment of unstable angina or non-ST segment elevation myocardial infarction (UA/NSTEMI) in patients for whom urgent (< 120 mins) invasive management (PCI) is not indicated (see sections 4. 4 and 5. 1). , Treatment of ST segment elevation myocardial infarction (STEMI) in patients who are managed with thrombolytics or who initially are to receive no other form of reperfusion therapy. , , 5 mg/0. 4 ml, 7. 5 mg/0. 6 ml and 10 mg/0. 8 ml solution for injection: , Treatment of acute Deep Vein Thrombosis (DVT) and treatment of acute Pulmonary Embolism (PE), except in haemodynamically unstable patients or patients who require thrombolysis or pulmonary embolectomy.
Anticoagulants, including unfractionated heparin (UFH), enoxaparin and fondaparinux, are approved drugs in acute coronary syndrome (ACS). Monocytes and monocyte-derived microparticles (MMPs) play an important procoagulant role in ACS by expressing high tissue factor (TF) levels, which in turn triggers thrombin generation. The objective of our study is to compare the in-vitro inhibitory effect of UFH, enoxaparin and fondaparinux in monocytes and MMP models. Human-elutriated monocytes were activated for 5 and 18 h by lipopolysaccharide to obtain activated monocytes (ac-M) or MMPs, respectively. Thrombin generation inhibition was assessed using ac-M or MMPs mixed with platelet-poor plasma containing increased concentrations of anticoagulants. Thrombin generation inhibition was dose-dependent with a differential effect according to the drug: the highest for UFH, the lowest for fondaparinux. Rate index was the most sensitive parameter. For fondaparinux, its IC50 values (anti-Xa IU/ml) were 0.59±0.05 for ac-M and 0.17±0.03 for MMPs. For enoxaparin, rate index IC50 values were 0.27±0.03 for ac-M and 0.19±0.02 for MMPs. Our data support the notion that cell-induced thrombin generation assay may be a reliable alternative to anti-Xa assessment in determining patient anticoagulation level. [2]
In conclusion, our study demonstrates a differential inhibitory effect of the three anticoagulants approved in ACS, the evaluation being performed using cell-induced thrombin generation models. Our results support the conclusion reached in OASIS-5 substudy that lower anticoagulation levels should be considered. Our results clearly show the limitation to answer the effect of an anticoagulant molecule using anti-Xa assays. Thrombin generation is an informative and sensitive assay, which remains difficult to standardize, although improvement of standardization is ongoing. Thrombin generation could be used to evaluate the pharmacodynamic effect of new anticoagulant drugs. Furthermore, thrombin generation assay could be a helpful tool to assess more accurately the level to reach in phase I and II studies.[2]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C31H53N3O49S8
Molecular Weight
1508.26
Exact Mass
1506.95133
Elemental Analysis
C, 24.69; H, 3.54; N, 2.79; O, 51.98; S, 17.01
CAS #
104993-28-4
Appearance
Typically exists as solids at room temperature
SMILES
S(N[C@H]1[C@@H](O[C@@H]2[C@H](C(=O)O)O[C@H]([C@@H]([C@H]2O)OS(=O)(=O)O)O[C@@H]2[C@@H](COS(=O)(=O)O)O[C@@H]([C@@H]([C@H]2O)NS(=O)(=O)O)OC)O[C@H](COS(=O)(=O)O)[C@H]([C@@H]1OS(=O)(=O)O)O[C@H]1[C@@H]([C@H]([C@@H]([C@@H](C(=O)O)O1)O[C@@H]1[C@@H]([C@H]([C@@H]([C@@H](COS(=O)(=O)O)O1)O)O)NS(=O)(=O)O)O)O)(=O)(=O)O
Synonyms
Fondaparinux free acid; Fondaparinux; 104993-28-4; Natural heparin pentasaccharide; Arixtra; CHEBI:61033; UNII-J177FOW5JL; J177FOW5JL; HSDB 7845;
HS Tariff Code
2934.99.9001
Storage

Powder      -20°C    3 years

                     4°C     2 years

In solvent   -80°C    6 months

                  -20°C    1 month

Shipping Condition
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
Solubility Data
Solubility (In Vitro)
May dissolve in DMSO (in most cases), if not, try other solvents such as H2O, Ethanol, or DMF with a minute amount of products to avoid loss of samples
Solubility (In Vivo)
Note: Listed below are some common formulations that may be used to formulate products with low water solubility (e.g. < 1 mg/mL), you may test these formulations using a minute amount of products to avoid loss of samples.

Injection Formulations
(e.g. IP/IV/IM/SC)
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution 50 μL Tween 80 850 μL Saline)
*Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH ₂ O to obtain a clear solution.
Injection Formulation 2: DMSO : PEG300Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL DMSO 400 μLPEG300 50 μL Tween 80 450 μL Saline)
Injection Formulation 3: DMSO : Corn oil = 10 : 90 (i.e. 100 μL DMSO 900 μL Corn oil)
Example: Take the Injection Formulation 3 (DMSO : Corn oil = 10 : 90) as an example, if 1 mL of 2.5 mg/mL working solution is to be prepared, you can take 100 μL 25 mg/mL DMSO stock solution and add to 900 μL corn oil, mix well to obtain a clear or suspension solution (2.5 mg/mL, ready for use in animals).
View More

Injection Formulation 4: DMSO : 20% SBE-β-CD in saline = 10 : 90 [i.e. 100 μL DMSO 900 μL (20% SBE-β-CD in saline)]
*Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution.
Injection Formulation 5: 2-Hydroxypropyl-β-cyclodextrin : Saline = 50 : 50 (i.e. 500 μL 2-Hydroxypropyl-β-cyclodextrin 500 μL Saline)
Injection Formulation 6: DMSO : PEG300 : castor oil : Saline = 5 : 10 : 20 : 65 (i.e. 50 μL DMSO 100 μLPEG300 200 μL castor oil 650 μL Saline)
Injection Formulation 7: Ethanol : Cremophor : Saline = 10: 10 : 80 (i.e. 100 μL Ethanol 100 μL Cremophor 800 μL Saline)
Injection Formulation 8: Dissolve in Cremophor/Ethanol (50 : 50), then diluted by Saline
Injection Formulation 9: EtOH : Corn oil = 10 : 90 (i.e. 100 μL EtOH 900 μL Corn oil)
Injection Formulation 10: EtOH : PEG300Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL EtOH 400 μLPEG300 50 μL Tween 80 450 μL Saline)


Oral Formulations
Oral Formulation 1: Suspend in 0.5% CMC Na (carboxymethylcellulose sodium)
Oral Formulation 2: Suspend in 0.5% Carboxymethyl cellulose
Example: Take the Oral Formulation 1 (Suspend in 0.5% CMC Na) as an example, if 100 mL of 2.5 mg/mL working solution is to be prepared, you can first prepare 0.5% CMC Na solution by measuring 0.5 g CMC Na and dissolve it in 100 mL ddH2O to obtain a clear solution; then add 250 mg of the product to 100 mL 0.5% CMC Na solution, to make the suspension solution (2.5 mg/mL, ready for use in animals).
View More

Oral Formulation 3: Dissolved in PEG400
Oral Formulation 4: Suspend in 0.2% Carboxymethyl cellulose
Oral Formulation 5: Dissolve in 0.25% Tween 80 and 0.5% Carboxymethyl cellulose
Oral Formulation 6: Mixing with food powders


Note: Please be aware that the above formulations are for reference only. InvivoChem strongly recommends customers to read literature methods/protocols carefully before determining which formulation you should use for in vivo studies, as different compounds have different solubility properties and have to be formulated differently.

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 0.6630 mL 3.3151 mL 6.6302 mL
5 mM 0.1326 mL 0.6630 mL 1.3260 mL
10 mM 0.0663 mL 0.3315 mL 0.6630 mL

*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

Calculation results

Working concentration mg/mL;

Method for preparing DMSO stock solution mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.

Method for preparing in vivo formulation:Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.

(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
             (2) Be sure to add the solvent(s) in order.

Clinical Trial Information
Treatment with Aspirin Alone Versus Aspirin in Combination with Fondaparinux Before Early Coronary Assessment in Patients with Non-ST-Elevation Myocardial Infarction
CTID: NCT06710184
Phase: Phase 4    Status: Not yet recruiting
Date: 2024-11-29
Endovenous Laser Ablation in Acute Thrombophlebitis of the Varicose Great Saphenous Vein
CTID: NCT05001776
Phase: Phase 4    Status: Completed
Date: 2023-12-20
Fondaparinux in Preventing Blood Clots in Patients Undergoing Surgery for Gynecologic Cancer
CTID: NCT00381888
Phase: Phase 2    Status: Completed
Date: 2017-12-28
The Effects of Arixtra on Wound Drainage Following Total Joint Arthroplasty
CTID: NCT00909064
Phase: Phase 4    Status: Completed
Date: 2017-05-15
Fondaparinux as an Anticoagulant in Haemofiltration in Patients With Acute Kidney Failure.
CTID: NCT00256100
Phase: N/A    Status: Terminated
Date: 2017-04-12
View More

Study of Arixtra (Fondaparinux Sodium) to Prevent Blood Clots in Women Undergoing Abdominopelvic Surgery for Likely Gynecologic Malignancy
CTID: NCT00539942
Phase: Phase 3    Status: Terminated
Date: 2017-03-27


The Treatment of Acute Pulmonary Thromboembolism (PE) of GSK576428 (Fondaparinux Sodium) in Japanese Patients
CTID: NCT00981409
Phase: Phase 3    Status: Completed
Date: 2016-12-16
The Treatment of Acute Deep Vein Thrombosis (DVT) of GSK576428 (Fondaparinux Sodium) in Japanese Patients
CTID: NCT00911157
Phase: Phase 3    Status: Completed
Date: 2016-11-23
A Study to Evaluate the Efficacy and Safety of Fondaparinux Sodium When Used With Intermittent Pneumatic Compression to Prevent Venous Thromboembolic (IPC) Versus IPC Alone for the Prevention of Venous Thromboembolic Events in Subjects at Increased Risk Undergoing Major Abdominal Surgery (APOLLO).
CTID: NCT00038961
Phase: Phase 3    Status: Completed
Date: 2016-09-01
A Study to Evaluate the Efficacy and Safety of Fondaparinux for the Prevention of Venous Blood Clots in Patients With a Plaster Cast or Other Type of Immobilization for a Below-knee Injury Not Ne
Superficial vein thrombosis (SVT) treated for forty-five days with Rivaroxaban versus Fondaparinux
CTID: null
Phase: Phase 3    Status: Completed
Date: 2012-04-11
SAFE-AF: Safety of Fondaparinux in electric cardioversion of atrial fibrillation.
CTID: null
Phase: Phase 2    Status: Completed
Date: 2009-05-04
(FONDACAST, del inglés FONDAparinux in patients with a plaster CAST)
CTID: null
Phase: Phase 3    Status: Completed
Date: 2008-11-25
Prospective, multicentre, open-label study evaluating Fondaparinux (1,5 mg/day) in venous thromboembolic events prevention in patients with renal impairment defined by a creatinin clearance between 20 and 50 ml/min and undergoing a major orthopaedic surgery (hip fracture, total hip replacement and total knee replacement)
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2008-09-15
Markers of hypercoagulability and risk of death and rehospitalization in heart failure patients: a pilot study on the effects of Fondaparinux
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2008-05-09
prospective randomized open study on the comparison of fondaparinux with the low-molecular-weight heparin enoxaparin in patients undergoing femoro-distal venous bypass operation
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2008-03-27
Pharmacodynamics of Fondaparinux subcutaneous versus continuous intravenous infusion in critically ill patients with or without vasopressors.
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2007-10-17
PHARMACOKINETICS OFDANAPAROID SODIUM (ORGARAN®), FONDAPARINUX SODIUM(ARIXTRA®) AND NADROPARIN CALCIUM (FRAXIPARIN®)
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2007-09-04
Fondaparinux vs Lepirudin in the treatment of heparin-induced thrombocytopenia, with or without thrombosis.
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2007-04-04
Comparison of ARIXTRA™ in lower LImb Superficial Thrombophlebitis with placebO (CALISTO).
CTID: null
Phase: Phase 3    Status: Completed
Date: 2007-03-14

Contact Us