yingweiwo

Varespladib Methyl

Alias: A002; LY-333013; S-3013; A002; A-002; LY333013; Varespladib methyl; 172733-08-3; LY-333,013; Varespladib methyl [USAN]; LY333,013; Varespladib methyl ester; S3013
Cat No.:V27947 Purity: ≥98%
Varespladib Methyl (LY-333013;A-002; S-3013)is a novel and potent sPLA2 (secretory phospholipase A2) inhibitorwith the potential for treating acute coronary syndrome (ACS).
Varespladib Methyl
Varespladib Methyl Chemical Structure CAS No.: 172733-08-3
Product category: Phospholipase
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
5mg
10mg
25mg
50mg
Other Sizes

Other Forms of Varespladib Methyl:

  • Varespladib (LY 315920)
  • Varespladib Sodium
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
Top Publications Citing lnvivochem Products
Product Description

Varespladib Methyl (LY-333013; A-002; S-3013) is a novel and potent sPLA2 (secretory phospholipase A2) inhibitor with the potential for treating acute coronary syndrome (ACS). It is an orally bioavailable prodrug of varespladib, which inhibits the IIA, V and X isoforms of sPLA2 to reduce inflammation, lower and modulate lipid levels, and reduce levels of C-reactive protein (CRP) and interleukin-6 (IL-6), both indicators of inflammation.

Biological Activity I Assay Protocols (From Reference)
Targets
sPLA2/secretory phospholipase A2 (IC50 = 9 nM)
ln Vitro
Varespladibmethyl, also known as LY333013, is a prodrug of methanol [1]. When used against 28 medically poisonous snake venom species from six continents, varespladib and its sidewall bioavailable prodrug methylvarespladib (methyl-Varespladib) exhibit high levels of fatal PLA2 (sPLA2) at nanomolar and picomolar concentrations. inhibiting impact [2].
Inhibition of sPLA2 Activities in Vitro [2]
In examining drugs that could be repurposed for snakebite, we found that varespladib (LY315920) and methyl-varespladib (LY333013) inhibited the sPLA2 activity of large arrays of snake venoms in vitro using chromogenic assays, but did not show great activity against bee venom used as a standard, positive control (Table 1 and Figure 1). Additionally surprising was the observation that the IC50 of varespladib and methyl-varespladib for essentially all snake venoms tested was significantly lower than values ever reported for inhibition of mammalian, including human, sPLA2 [2].
ln Vivo
Efficacy. At week 1 of treatment methyl-varespladib/LY333013, 1000 mg/day, was superior to placebo in the proportion of patients achieving an ACR20 response (p = 0.064) and a dose-response relationship was observed (p = 0.058) (Figure 1). A statistically significant dose-response relationship was observed for CRP at week 1 (p = 0.046). However, ACR20 response rates increased in all treatment groups at study weeks 4 and 8, with loss of difference from placebo. Exploratory analyses showed a trend towards a dose-dependent relationship for ACR20 response (p = 0.077) among patients older than 65 years. Analyses of secondary endpoints, including time to achieve an ACR20 response, time to treatment failure, and time to achieve a 20% improvement in patient-reported pain or global arthritis status did not differ among treatment groups. An ACR50 response was achieved by 9/56 (16.1%) patients treated with placebo, and by 10/60 (16.7%), 5/59 (8.5%), and 5/61 (8.2%) patients treated with methyl-varespladib/LY333013 50, 250, and 1000 mg/day, respectively. Changes among the treatment groups in ACR response components were generally small or inconsistent and not statistically significant. TJC increased at weeks 8 and 12 in the LY333013, 250 and 1000 mg/day groups, and SJC did so at week 12. The investigators’ global assessment reflected these findings, but the patients’ global and pain assessments did not. Exploratory analyses did not suggest a treatment response in terms of presence of RF, number of DMARD used, or specific DMARD used at study entry. Notably, only 18% of patients were taking NSAID at study entry, in contrast to 48% using low-dose corticosteroids. Corticosteroid use appeared to reduce achievement of ACR20 in the LY333013 1000 mg/day group [1].
The demographic characteristics of the treatment groups were similar. Dose-response relationships were found for ACR20 responses (p = 0.058) and reductions in C-reactive protein (p = 0.058) at week 1. The proportions of patients with an ACR20 response subsequently increased in all study groups including the placebo group at weeks 4 and 8, and the initial treatment benefit was lost. Adverse events were generally mild in severity and not associated with treatment. Conclusion: Treatment with methyl-varespladib/LY333013 for 12 weeks was well tolerated but ineffective as an adjunct to DMARD treatment of active RA [1].
ApoE−/− Standard Model [3]
The body weights of animals in control or A-002 (varespladib methyl/LY333013/S-3013-treated groups were similar at time 0 (vehicle: 27.2 ± 2.6, 30 mg/kg A-002: 26.7 ± 2.3, and 90 mg/kg A-002: 27.3 ± 2.5). Body weight increased over the 16 weeks on a Western diet by approximately 155%, 150%, and 141%, for the vehicle, 30 mg/kg A-002 (varespladib methyl/LY333013/S-3013, and 90 mg/kg A-002 groups, respectively (Fig. 2). There was no statistically significant difference in body weights between the groups using a repeated-measures 2-way ANOVA where time and treatment were the variables.

Plasma cholesterol levels at the beginning of the study were not significantly different between groups. However, after 1 month of twice a day treatment with A-002 (varespladib methyl/LY333013/S-3013, either 30 or 90 mg/kg doses, total cholesterol was significantly decreased (Fig. 3) compared with the control group. This effect remained consistent throughout the 4 months of treatment. Plasma cholesterol changed after 4 months of diet per treatment by +15%, −10%, and −12% for vehicle, 30 mg/kg A-002, and 90 mg/kg A-002 groups, respectively (Fig. 3). There was no apparent dose-response effect on plasma cholesterol concentrations.

Treatment with A-002 (varespladib methyl/LY333013/S-3013 had a significant effect on plaque content expressed as percent occupancy of the aortic luminal surface by atherosclerotic plaques. Vehicle-treated mice had approximately 12.6% ± 0.7% plaque coverage, whereas mice treated with 30 mg/kg A-002 twice a day had 6.3% ± 0.6% plaque coverage and mice treated with 90 mg/kg A-002 twice a day had 6.7% ± 0.8% plaque coverage. These represent significant decreases in plaque content in each of the A-002 treatment groups compared with the treatment group receiving only the formulation vehicle (P < 0.05) (Fig. 4).
Angiotensin II ApoE−/− Model [3]
Angiotensin II formulated in water twice a day or in 5% acacia twice a day resulted in similar aortic plaque coverage (18% ± 3.3% and 14.4% ± 4.8%, respectively, Fig. 5). A-002 (varespladib methyl/LY333013/S-3013 (30 mg/kg) significantly reduced the plaque coverage of the aorta (8% ± 3% vs. 18% ± 3.3% observed with angiotensin II infusion without drug treatment, P < 0.025). The background amount of atherosclerosis in the absence of angiotensin II (3.8% ± 0.6%, subcutaneous saline pump and water twice a day) was significantly lower than that with angiotensin II infusion (18% ± 3.3%, P < 0.025). Representative in face images are shown in figure 6.

Aortic aneurysm rate was assessed in each group of mice. In the absence of angiotensin II infusion, no aneurysms were observed. Infusion of angiotensin II formulated in water resulted in a 25% incidence of aneurysm and infusion of angiotensin II formulated in the acacia vehicle caused a 22.2% incidence of aneurysm. A-002 (varespladib methyl/LY333013/S-3013 treatment (30 mg/kg twice a day) in the mice infused with angiotensin II formulated in acacia prevented aneurysm formation completely (Prob>ChiSq = 0.0096) (Table 1).
Enzyme Assay
In Vitro Experiments [2]
Experiments were performed to assess sPLA2 activity using the 1,2-dithio analog of diheptanoyl phosphatidylcholine. sPLA2 catalyzes the hydrolysis of phospholipids at the sn-2 position yielding a free fatty acid and a lysophospholipid. The release of arachidonic acid from membrane phospholipids by PLA is believed to be a key step in the control of eicosanoid production within the cell. The Bee Venom PLA2 Control was a 100 μg/mL solution of bee venom PLA2 was supplied as a positive control from kits. Assay optimization, screening and dose response measurements were performed at the Yale Center for Molecular Discovery. Experiments were performed in an assay buffer containing 25 mM Tris-HCl, pH 7.5, 10 mM CaCl2, 100 mM KCl, 0.3% Triton X-100 (Fluka) and 454 µM DTNB and plated into clear, Non-Treated 384-well plates. Venoms were reconstituted in 1× phosphate-buffered saline to a concentration of 10,000 µg/mL. Crude, unfractionated lyophilized venom purchased from Sigma (E. carinatus and D. russelli) or the Miami Serpentarium (all others) was used in all cases. Varespladib and methyl-varespladib/LY333013 were purchased from Chemietek and dissolved in DMSO for in vitro experiments and bicarbonate/dextrose for in vivo experiments. The activity of venoms with 0.375 mM 1,2-bis(heptanoyl) Glycerophosphocholine, the sPLA2 substrate, was selected based on kinetic enzymatic assays conducted at room temperature. Concentrations of venom was selected for screening and potency studies in which high sPLA2 activity was observed relative to any background activity of no venom control wells, and for which there was negligible substrate depletion at 60 min. The range in final concentrations for venoms used in assays was 0.0037–5 µg/mL, demonstrating large differences among venoms in the proportion or relative sPLA2 activity for this substrate. For 13 elapid venoms final concentrations ranged from 0.0037 µg/mL for M. fulvius to 100 µg/mL for Dendroaspis polylepis (mean concentration 7.8 ± 28 µg/mL; median concentration 0.1 µg/mL) and for 15 viper venoms a range of 0.033 µg/mL, e.g., V. berus and 25 µg/mL Calloselasma rhodostoma (mean concentration 2.47 ± 6.35 µg/mL; median concentration 1 µg/mL). Piloting collections used in the first phases of screening were selected by the inventor or from libraries of known compounds and natural products available at the time of experiments on selected venoms including: NIH Clinical Collection, GenPlus, Pharmakon, Bioactive lipids, Protease Inhibitors, Procured Drugs and FDA Approved Drugs libraries. The GenPlus (the NINDS Custom Collection) from MicroSource Discovery Systems contains 960 compounds. Varespladib outperformed all compounds in the library confirming (within the limits of the screening libraries) its superior potency as an sPLA2 inhibitor. No molecule in these collections totaling more than 4000 distinct chemical entities was found to be within two orders of magnitude potency of varespladib and results are not within the scope of this manuscript. For inhibitor and dose-response testing, 10 µL of snake venom or bee venom (+control) was added to assay plates using a multichannel pipetman or a multidrop dispenser. Compounds from the chemical libraries or prepared serial dilution master plates dissolved in DMSO were added to assay plates using a pin tool to transfer 20 nL of compounds. Final DMSO concentrations in the assay are 0.1%. Substrate was then added in 10 µL for a final assay volume of 20 µL. Controls populations were included on each plate in replicate wells. The negative control wells were vehicle (DMSO-only) with no small molecule compound. The positive control to simulate full venom activity inhibition were wells in which no venom was added, and assay buffer added in its place. Assay signals were measured at initiation and after 60 min of reaction time at room temperature. Signals were quantified on the Tecan infiniTe M1000 plate reader measuring absorbance at 405 nm. Signals at initiation were subtracted from the signals at 60 min. These background-corrected values were normalized to the mean of replicate negative and positive control wells within the plate. To define the normalization scale, the mean of the negative control well signals, representing full venom activity, was normalized to 100% effect and the mean of positive control well signals, representing complete inhibition of venom activity, was normalized to 0% effect. And wells within the plate were scaled accordingly. These calculations were performed in MicroSoft Excel. Data were transferred to GraphPad Prism (6th edition, 2014, La Jolla, CA, USA) plotted and fit to models, such that IC50 or EC50 values could be determined. Tests of significance were calculated by Student’s t and all others were descriptive.
sPLA2 Inhibition [3]
The intrinsic activity of A-001 on the inhibition of sPLA2 group V and X enzymes was measured according to a chromogenic method described elsewhere.
Animal Protocol
Two hundred and fifty-one patients with active RA despite treatment with one or more disease modifying antirheumatic drugs (DMARD) received oral doses of LY333013 (50, 250, and 1000 mg) or placebo once daily for 12 weeks. Concomitant low-dose glucocorticoids (< or = 10 mg/day prednisone equivalent) were allowed. Clinical improvement was assessed using the response criteria of the American College of Rheumatology (ACR20), and safety was evaluated with respect to adverse events and laboratory test abnormalities. [1]
Animal in Vivo Studies [2]
M. fulvius and V. berus venoms had the highest sPLA2 activity in vivo and and after successful pilot survival studies was chosen for the Non-GLP study in rats and V. berus for mouse studies. CD-1 mice and Sprague-Dawley Rats with implanted jugular venous catheters were used. 18 Sprague-Dawley rats weighing between 183 and 214 g at the time of the study had jugular vein cannulas surgically implanted by the supplier. Rats were randomly assigned to six treatment groups (n = 3 each) and received snake venom with and without varespladib. Animals were monitored for signs of toxicity for approximately 24 h. Blood samples (without anticoagulant) were collected from each rat prior to dose administration and post dose administration at approximately 30 min, 1 h and 4 h. Per protocol, nominal blood collection times were pre-dose administration and post-varespladib administration at 30 min ± 1 min., 1 h ± 1 min., and 4 h ± 5 min. There were no deviations from these specifications except for animals that died prior to the last scheduled blood collect ion at 4 h. Blood was processed to serum and analyzed by the AILAC certified contract research organization to determine sPLA2 activity validated beforehand with rat serum for quality control. Surviving animals were euthanized following the 24-h observation. Tissues were grossly examined but not collected for further processing. Justification for the use of the mouse in this study is based on the premise that animal testing is an appropriate and ethical prerequisite to testing new drugs in humans, and that data obtained from nonclinical animal models will have relevance to the behavior of the test material in humans. Because of the complex interactions that occur in vivo, an in vitro system does not provide sufficient information for evaluation of a compound’s in vivo activities. It was expected that the number of animals used in this study would provide a large enough sample for scientifically meaningful results while using the fewest possible animals to achieve that result. The intravenous route was chosen to maximize the bioavailability of varespladib. All experiments were designed to insure 100% mortality in control animals within the expected ½ life of the test drug in order to produce clear results using the lowest number of animal.

sPLA2 Group IIA Transgenic Model [3]
A transgenic mouse model with human sPLA2 group IIA under inducible mouse metallothionein promoter control in a C57BL/6J ApoE+/+ background was used to evaluate the inhibition of phospholipase A2 activity in serum from animals dosed with methyl-varespladib/LY333013/A-002 as described by Fox et al.21 Animals were administered a single oral dose of methyl-varespladib/LY333013/A-002 (1 or 3 mg/kg) or vehicle (5% acacia), and enzymatic activity22 was measured in samples at baseline before treatment and 0.5, 2, and 4 hours after dosing.
ApoE−/− Standard Model [3]
The ApoE−/− model was developed as described elsewhere.23 Male ApoE−/− mice (129/Ola × C57BL/6J, multiple generations) were used at an age of 6-8 weeks. Mice were fed ad libitum a high-fat diet (21% fat: 0.15% cholesterol and 19.5% casein) for 2 weeks to allow time to adjust to the diet. Mice were randomized into groups based on plasma triglyceride and total cholesterol levels using the GroupOptimizer V211.xls program. Animals were treated for 16 weeks with 30 mg/kg twice a day or 90 mg/kg twice a day methyl-varespladib/LY333013/A-002. Compounds were formulated in 0.2 mL of 5% acacia (10 mL/kg body weight) and administered by oral gavage twice a day at 7 am and 3 pm.
ApoE−/− Angiotensin II Accelerated Model [3]
Male ApoE−/− mice (129/Ola × C57BL/6J, multiple generations) were used at an age of 6-8 weeks. In this model, angiotensin II was administered by 0.7 mg kg−1 d−1 subcutaneous infusion to induce faster atherosclerosis development associated with aneurysm formation.24 Mice were fed ad libitum a high-fat diet (21% fat: 0.15% cholesterol and 19.5% casein) for 2 weeks to allow time to adjust to the diet. The animals were then randomized into groups based on plasma triglyceride and total cholesterol levels using the GroupOptimizer V211.xls program and were treated for 4 weeks with 30 mg/kg twice a day of methyl-varespladib/LY333013/A-002 as described in the standard ApoE−/− model.
Procedures. Patients meeting the inclusion and exclusion criteria at an initial entry visit were reassessed at their baseline visit to assure that they continued to meet the entry criteria for active RA. The patients completed the Stanford Health Assessment Questionnaire (HAQ)20, including a 100 mm horizontal visual analog scale (VAS) for pain (0 = no pain; 100 = extreme pain), and assessed the global activity of their arthritis on a 100 mm VAS (0 = very well; 100 = very poorly) at each visit. The investigators were instructed in a standardized 28-joint technique21 for counting the number of swollen (SJC) and tender joints (TJC) at each visit, and they judged global arthritis activity on a 100 mm VAS (0 = very well; 100 = very poorly). Safety was assessed on the basis of adverse events reported at each visit (baseline and after 1, 4, 8, and 12 weeks of treatment), findings on physical examination, and laboratory evaluations. Samples for population pharmacokinetics and/or CRP were obtained at baseline and subsequent visits. Study medications. The study medications were prepared in identicalappearing white tablets containing 0, 50, 125, or 250 mg of methyl-varespladib/LY333013. Patients took 2 tablets twice daily from blister cards that corresponded to assigned dose levels of 0 (placebo), 50, 250, and 1000 mg/day of methyl-varespladib/LY333013. The 50 and 250 mg/day dose groups received active medication only in the morning dose. Randomization to treatment was blocked and stratified by site. Treatment assignments were managed by an interactive telephone voice response system. Treatment identities were provided to the investigators in a sealed envelope, but except for emergencies warranting it, unblinding did not occur until after the last study visit. Unblinded patients were discontinued from study participation unless there was a compelling ethical reason for their continuation in the study. Compliance to the treatment regimen was monitored by counts of medication in the returned blister cards. In the event that the blister cards were lost, medication use was assessed by patient report. Statistical analysis. The p values for the demographic, RA baseline disease features and medications, and adverse event data (Tables 1-4) were computed for continuous variables from the rank-transformed analysis of variance model with terms for dose and pooled investigative site; p values for categorical variables were computed from the Cochran-Mantel-Haenszel test stratified by pooled investigative site. The primary efficacy evaluation utilized the ACR Definition of Improvement22 at the 20% level (ACR20). Treatment was deemed a failure if, at the end of 4 weeks of treatment, the patient’s condition had worsened by > 20% relative to baseline or, after 8 weeks, there had been < 10% improvement or, after 12 weeks, there had been < 20% improvement. These patients were discontinued from study treatment and were classified as non-responders with respect to any definition of improvement. Efficacy analysis was performed on all randomized patients who received at least one dose of study medication and who had at least one efficacy assessment at the 4, 8, or 12 week visits [intention-totreat (ITT) group]. Missing data were handled using a last observation carried forward analysis. Adverse event analysis was performed on all patients who received at least one dose of study medication. Assuming that 30% of patients receiving placebo would achieve an ACR20 response compared to 50% of patients receiving LY333013, 1000 mg/day, 59 patients per treatment group would provide 80% probability of detecting a linear doseresponse relationship at a one-sided α = 0.1 using a logistic regression model with dose as the explanatory variable. Pharmacokinetic measurements. High performance liquid chromatography/mass spectroscopic methods were used to measure the active metabolite of LY333013. Results were evaluated according to the reported time of the last 4 doses of study medication and the assigned dose. A 2- compartment distribution model was used with assumption of first-order elimination. Based on previous studies, it was known that plasma clearance is affected by bioavailability, which in turn is reduced by increasing dose. These factors were included in the model evaluating individual apparent oral clearance and inter-individual variability in this parameter. Systemic exposure was estimated based on dose and apparent oral clearance for all patients with available pharmacokinetics data.[1]
ADME/Pharmacokinetics
Treatment exposure. The mean plasma steady-state concentration of the active metabolite of methyl-varespladib/LY333013 (LY315920) was estimated to be 222, 627, and 1445 ng/ml for the 50, 250, and 1000 mg/day dose groups, respectively. There were no apparent interactions between LY315920 concentrations and co-therapy with MTX, sulfasalazine, and hydroxychloroquine. Ninety-one percent of the efficacy analysis patients took 80-119% of their prescribed doses of study drug. [1]
Methyl-varespladib/LY333013, a rapidly absorbed, orally bioavailable prodrug of varespladib could be administered orally (e.g., as an elixir) so that a person with no or very limited skill could potentially initiate treatment outside a hospital setting. Methyl-varespladib/LY333013is metabolized to varespladib, so the parent compound was the focus of our proof-of-concept studies though field use of an IV formulation is an unlikely scenario. The use of 3-substituted indoles for snake venom sPLA2 inhibition represents a possible springboard for the genesis of effective field treatments for snakebites; either could be rapidly developed with programmatic support and industry cooperation. Our findings warrant further investigation into the efficacy of veraspladib and methyl-varespladib in an even wider diversity of snakes to determine if either could be an essential component of the long sought-after venom antagonistic, first-line field-treatment for snakebite.[2]
Plasma Levels and Dose Selection [3]
Figure 1 shows plasma levels of A-001 in serum after a single oral dose of methyl-varespladib/LY333013/A-002. Levels of A-001 were detectable in all samples from dosed mice at 10, 30, and 90 mg/kg. The 2 highest doses, 30 and 90 mg/kg A-002, reached concentrations of A-001 in plasma that were greater than the IC50 values for sPLA2 groups V and X, respectively, throughout the dosing period.
The family of secretory phospholipase A2 (sPLA2) enzymes has been associated with inflammatory diseases and tissue injury including atherosclerosis. A-001 is a novel inhibitor of sPLA2 enzymes discovered by structure-based drug design, and A-002 (varespladib methyl/LY333013/S-3013 is the orally bioavailable prodrug currently in clinical development. A-001 inhibited human and mouse sPLA2 group IIA, V, and X enzymes with IC50 values in the low nM range. A-002 (1 mg/kg) led to high serum levels of A-001 and inhibited PLA2 activity in transgenic mice overexpressing human sPLA2 group IIA in C57BL/6J background. In addition, the effects of A-002 on atherosclerosis in 2 ApoE−/− mouse models were evaluated using en face analysis. (1) In a high-fat diet model, A-002 (30 and 90 mg/kg twice a day for 16 weeks) reduced aortic atherosclerosis by 50% (P < 0.05). Plasma total cholesterol was decreased (P < 0.05) by 1 month and remained lowered throughout the study. (2) In an accelerated atherosclerosis model, with angiotensin II-induced aortic lesions and aneurysms, A-002 (30 mg/kg twice a day) reduced aortic atherosclerosis by approximately 40% (P < 0.05) and attenuated aneurysm formation (P = 0.0096). Thus, A-002 was effective at significantly decreasing total cholesterol, atherogenesis, and aneurysm formation in these 2 ApoE−/− mouse models.[3]
Toxicity/Toxicokinetics
Safety. Of the 251 patients who received at least one dose of study medication, 182 reported at least one adverse event (AE). The most commonly reported AE were: rhinitis (21.9%), headache (19.5%), cough (13.5%), diarrhea (9.2%), nausea (8.8%), fever (8.4%), dizziness (6.4%), abdominal pain (6.0%), sinusitis (6.0%), and pharyngitis (5.6%), with no statistically significant differences across treatment groups for any of these events (Table 4). In general, the type, severity, and distribution of AE were similar across all treatment groups. The overall proportion of patients with AE was somewhat lower in the placebo group (60.3%) than in the methyl-varespladib/LY333013 50 (69.4%), 250 (79.0%), or 1000 mg/day groups (81.3%, overall p = 0.043), but an analysis of AE by organ system indicated that only the cardiovascular system showed a statistical difference among treatment groups (p = 0.033). Cardiovascular AE did not appear to be dose-related; they occurred in more patients in the methyl-varespladib/LY333013 50 and 1000 mg/day treatment groups (16.1%; 15.6%, respectively) than in the methyl-varespladib/LY333013 250 mg/day (3.2%) and placebo groups (4.3%). Upper respiratory tract infections were more common in LY333013-treated patients, whereas urinary tract infections were more common among patients receiving placebo. These differences were not thought to be clinically significant. Six patients with non-serious AE discontinued study treatment: all were receiving LY333013 (Table 5). These AE were vertigo (50 mg); dyspepsia, nausea, rash (250 mg); asthenia and somnolence (1000 mg). Serious AE were observed in 3 placebo- and 4 LY333013-treated patients; these included septic arthritis, pneumonia, and stroke in the placebo group, and chest pain (50, 250, 1000 mg groups) and perforated colonic diverticulum (50 mg) in the LY333013 groups. The latter patient had a history of diverticulitis, and all 3 patients with chest pain had prior diagnoses of cardiovascular disease. One patient (250 mg group) underwent coronary angioplasty. Study treatment was discontinued in patients with serious AE, i.e., pneumonia and colonic perforation. Only the latter serious AE was assessed by the investigator to be possibly related to study treatment. No patients died during study participation. There were no clinically relevant changes in laboratory variables, vital signs, or electrocardiogram recordings. Using the National Cancer Institute Common Toxicity Criteria (CTC) grades, only serum AST and calcium differed between placebo and LY333013 (p = 0.049). No patient had a CTC grade 4 abnormality of any analyte; the highest CTC grade for serum calcium was 2. Serum AST was more frequently elevated in the LY333013 1000 mg/day group; the highest value in this group was 122 IU. [1]
References

[1]. A randomized, double-blinded, placebo-controlled clinical trial of LY333013, a selective inhibitor of group II secretory phospholipase A2, in the treatment of rheumatoid arthritis. J Rheumatol. 2005 Mar;32(3):417-23.

[2]. Varespladib (LY315920) Appears to Be a Potent, Broad-Spectrum, Inhibitor of Snake Venom Phospholipase A2 and a Possible Pre-Referral Treatment for Envenomation. Toxins (Basel). 2016 Aug 25;8(9). pii: E248.

[3]. Varespladib (A-002), a Secretory Phospholipase A2 Inhibitor, Reduces Atherosclerosis and Aneurysm Formation in ApoE−/− Mice J Cardiovasc Pharmacol. 2009 Jan;53(1):60-5.

Additional Infomation
Varespladib methyl or methyl-varespladib/LY333013 is a methyl ester resulting from the formal condensation of the carboxy group of varespladib with methanol. It is a potential therapy for the treatment of snakebite envenomings in which toxicity depends on the action of PLA2s. It has a role as a prodrug, an anti-inflammatory drug, an antidote and an EC 3.1.1.4 (phospholipase A2) inhibitor. It is a methyl ester, an aromatic ether, a member of benzenes, a member of indoles and a primary carboxamide. It is functionally related to a varespladib.
Varespladib methyl has been investigated for the treatment of Acute Coronary Syndrome. Studies showed that Varespladib methyl treatment resulted in significant positive changes on lipoproteins and inflammation.
Drug Indication
Investigated for use/treatment in atherosclerosis and coronary artery disease.
Mechanism of Action
A–002 is an orally administered‚ potent inhibitor of secretory phospholipase spla2(spla2)‚ including groups IIA‚ V‚ and X. Atherosclerosis is a disease of the arteries that results from inflammation and the build-up of plaque under the lining of the blood vessel. This build-up can cause vascular swelling and eventual rupture. spla2 levels have been shown to be elevated in patients with both stable and unstable coronary artery disease. Higher levels of the enzyme have been shown to predict an increased risk for future cardiovascular events such as heart attacks and stroke.
Varespladib methyl is a methyl ester resulting from the formal condensation of the carboxy group of varespladib with methanol. It is a potential therapy for the treatment of snakebite envenomings in which toxicity depends on the action of PLA2s. It has a role as a prodrug, an anti-inflammatory drug, an antidote and an EC 3.1.1.4 (phospholipase A2) inhibitor. It is a methyl ester, an aromatic ether, a member of benzenes, a member of indoles and a primary carboxamide. It is functionally related to a varespladib.
Varespladib methyl has been investigated for the treatment of Acute Coronary Syndrome. Studies showed that Varespladib methyl treatment resulted in significant positive changes on lipoproteins and inflammation.
VARESPLADIB METHYL is a small molecule drug with a maximum clinical trial phase of III (across all indications) and has 3 investigational indications.
Snakebite remains a neglected medical problem of the developing world with up to 125,000 deaths each year despite more than a century of calls to improve snakebite prevention and care. An estimated 75% of fatalities from snakebite occur outside the hospital setting. Because phospholipase A2 (PLA2) activity is an important component of venom toxicity, we sought candidate PLA2 inhibitors by directly testing drugs. Surprisingly, varespladib and its orally bioavailable prodrug, methyl-varespladib/LY333013 showed high-level secretory PLA2 (sPLA2) inhibition at nanomolar and picomolar concentrations against 28 medically important snake venoms from six continents. In vivo proof-of-concept studies with varespladib had striking survival benefit against lethal doses of Micrurus fulvius and Vipera berus venom, and suppressed venom-induced sPLA2 activity in rats challenged with 100% lethal doses of M. fulvius venom. Rapid development and deployment of a broad-spectrum PLA2 inhibitor alone or in combination with other small molecule inhibitors of snake toxins (e.g., metalloproteases) could fill the critical therapeutic gap spanning pre-referral and hospital setting. Lower barriers for clinical testing of safety tested, repurposed small molecule therapeutics are a potentially economical and effective path forward to fill the pre-referral gap in the setting of snakebite.[2]
The family of secretory phospholipase A2 (sPLA2) enzymes has been associated with inflammatory diseases and tissue injury including atherosclerosis. A-001 is a novel inhibitor of sPLA2 enzymes discovered by structure-based drug design, and methyl-varespladib/LY333013/A-002 is the orally bioavailable prodrug currently in clinical development. A-001 inhibited human and mouse sPLA2 group IIA, V, and X enzymes with IC50 values in the low nM range. A-002 (1 mg/kg) led to high serum levels of A-001 and inhibited PLA2 activity in transgenic mice overexpressing human sPLA2 group IIA in C57BL/6J background. In addition, the effects of A-002 on atherosclerosis in 2 ApoE−/− mouse models were evaluated using en face analysis. (1) In a high-fat diet model, A-002/methyl-varespladib/LY333013 (30 and 90 mg/kg twice a day for 16 weeks) reduced aortic atherosclerosis by 50% (P < 0.05). Plasma total cholesterol was decreased (P < 0.05) by 1 month and remained lowered throughout the study. (2) In an accelerated atherosclerosis model, with angiotensin II-induced aortic lesions and aneurysms, A-002 (30 mg/kg twice a day) reduced aortic atherosclerosis by approximately 40% (P < 0.05) and attenuated aneurysm formation (P = 0.0096). Thus, A-002 was effective at significantly decreasing total cholesterol, atherogenesis, and aneurysm formation in these 2 ApoE−/− mouse models.[3]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C22H22N2O5
Molecular Weight
394.427
Exact Mass
394.153
Elemental Analysis
C, 66.99; H, 5.62; N, 7.10; O, 20.28
CAS #
172733-08-3
Related CAS #
Varespladib;172732-68-2; 172733-42-5 (Varespladib sodium)
PubChem CID
9886917
Appearance
White to off-white solid powder
LogP
3.172
Hydrogen Bond Donor Count
1
Hydrogen Bond Acceptor Count
5
Rotatable Bond Count
9
Heavy Atom Count
29
Complexity
604
Defined Atom Stereocenter Count
0
SMILES
CCC1=C(C2=C(N1CC3=CC=CC=C3)C=CC=C2OCC(=O)OC)C(=O)C(=O)N
InChi Key
VJYDOJXJUCJUHL-UHFFFAOYSA-N
InChi Code
InChI=1S/C22H22N2O5/c1-3-15-20(21(26)22(23)27)19-16(24(15)12-14-8-5-4-6-9-14)10-7-11-17(19)29-13-18(25)28-2/h4-11H,3,12-13H2,1-2H3,(H2,23,27)
Chemical Name
methyl 2-(1-benzyl-2-ethyl-3-oxamoylindol-4-yl)oxyacetate
Synonyms
A002; LY-333013; S-3013; A002; A-002; LY333013; Varespladib methyl; 172733-08-3; LY-333,013; Varespladib methyl [USAN]; LY333,013; Varespladib methyl ester; S3013
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

Note: Please store this product in a sealed and protected environment (e.g. under nitrogen), avoid exposure to moisture.
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)
DMSO : ~100 mg/mL (~253.54 mM)
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 5 mg/mL (12.68 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 50.0 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL.
Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution.

Solubility in Formulation 2: ≥ 2.5 mg/mL (6.34 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.5353 mL 12.6765 mL 25.3530 mL
5 mM 0.5071 mL 2.5353 mL 5.0706 mL
10 mM 0.2535 mL 1.2677 mL 2.5353 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
Broad-spectrum Rapid Antidote: Varespladib Oral for Snakebite
CTID: NCT04996264
Phase: Phase 2
Status: Completed
Date: 2023-06-22
FRANCIS-ACS Trial: A Study of the Safety and Efficacy of A 002 in Subjects With Acute Coronary Syndromes
CTID: NCT00743925
Phase: Phase 2
Status: Completed
Date: 2014-03-04
A Study of the Pharmacokinetics and Safety of Varespladib in Subjects With Normal or Impaired Renal Function
CTID: NCT01359579
Phase: Phase 1
Status: Terminated
Date: 2012-03-20
Study to Assess the Metabolism and Elimination of [14C]-Labeled Varespladib Methyl in Healthy Male Subjects
CTID: NCT01359605
Phase: Phase 1
Status: Completed
Date: 2011-12-20
PLASMA Trial: A Dose-Response Study of A-002 In Subjects With Stable Coronary Artery Disease
CTID: NCT00455546
Phase: Phase 2
Status: Completed
Date: 2010-02-02
Contact Us