The largest database of trusted experimental protocols
> Chemicals & Drugs > Organic Chemical > Pitavastatin

Pitavastatin

Pitavastatin is a synthetic statin medication used to lower cholesterol levels in individuals with high blood lipid concentrations.
It functions by inhibiting the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which is a key step in cholesterol biosynthesis.
Pitavastatin has been shown to effectively reduce low-density lipoprotein (LDL) cholesterol, triglycerides, and increase high-density lipoprotein (HDL) cholesterol.
It is typicaly well-tolerated, with potential side effects including myopathy and elevations in liver enzymes.
Pitavastatin is an important pharmacotherpeutic option for the management of dyslipidemia and reduction of cardiovascular disease risk.

Most cited protocols related to «Pitavastatin»

The REAL-CAD study (Randomized Evaluation of Aggressive or Moderate Lipid Lowering Therapy With Pitavastatin in Coronary Artery Disease) is a prospective, multicenter, randomized, open-label, blinded end point, physician-initiated superiority trial to determine whether high-dose (4 mg/d) compared with low-dose (1 mg/d) pitavastatin therapy could reduce cardiovascular events in Japanese patients with stable CAD. Pitavastatin is a statin with potent LDL-C–lowering effects developed by Kowa Pharmaceutical Co Ltd (Tokyo, Japan). Pitavastatin doses of 1 and 4 mg were reported to reduce LDL-C by 33.6% and 47.2%, respectively, in Japanese patients.20 (link) A similar magnitude of LDL-C reduction was also reported in white and East Asian patients.21 (link)–23 (link) Pitavastatin 4 mg is the maximum approved dose in Japan and has demonstrated effects comparable to atorvastatin 20 mg in terms of both LDL-C reduction and coronary plaque regression assessed by intravascular ultrasound, whereas pitavastatin 1 mg has an LDL-C–lowering effect comparable to that of atorvastatin 5 mg.24 (link),25 (link)Eligible patients were men and women 20 to 80 years of age with stable CAD as defined by a history of acute coronary syndrome or coronary revascularization >3 months ago or a clinical diagnosis of CAD with angiographically documented coronary artery stenosis of at least 75% diameter narrowing according to the American Heart Association classification.26 (link) We excluded those patients with LDL-C <100 mg/dL without statin therapy before enrollment because the label in the instructions for pitavastatin restricted use to patients with hypercholesterolemia. Detailed inclusion and exclusion criteria are provided in the online-only Data Supplement. Patients were enrolled on an outpatient basis through academic and general hospitals and clinics across Japan. Eligible patients who provided informed consent were enrolled and received pitavastatin 1 mg once daily orally for a run-in period of at least 1 month. Patients were evaluated for secondary eligibility, excluding those patients with LDL-C ≥120 mg/dL after the run-in period, onset of acute coronary syndrome and/or coronary revascularization within the past 3 months, poor medication adherence to pitavastatin, occurrence of primary end point events, or adverse events prohibiting study continuation during the run-in period.
Patients who met the secondary eligibility criteria were randomized in a 1-to-1 fashion to oral pitavastatin, either 4 mg/d (high-dose group) or 1 mg/d (low-dose group), with an electronic data capture system and dynamic allocation stratified by facility, age (<65 or ≥65 years), sex, diabetes mellitus, and statin use before enrollment. The assignment algorithm was determined by the study statistician. This is an open-label trial. However, the independent event committee adjudicated all the end point events while blinded to the assigned group (online-only Data Supplement).
During follow-up, the patients’ visits dictated by the protocol were at 6 and 12 months in the first year and every 12 months thereafter. Serum lipid levels such as LDL-C, total cholesterol, triglycerides, and high-density lipoprotein cholesterol, as well as other blood tests such as creatine kinase, alanine aminotransferase, aspartate aminotransferase, creatinine, and hemoglobin A1c, were to be measured at baseline, at 6 and 12 months, and yearly thereafter, whereas high-sensitivity C-reactive protein (hsCRP) was to be measured at baseline and at 6 months.
The site investigators reported follow-up information through the web-based electronic data capturing system. Data were monitored by the data center, and the logical inconsistencies were resolved by queries. Final clinical follow-up data were collected through January to March 2016. From 2012 to 2016, site audits were performed for 3914 patients from 28 centers, and the independent data monitoring committee regularly assessed the safety aspect of study conduct.
Publication 2018

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2019
Patients pitavastatin Placebos Primary Prevention
Transport studies in HEK293 stable cell lines and in human SCH were conducted similarly to those described previously.41 (link),42 (link) HEK293-OATP1B1, -FLAG-OATP1B1, and -Mock cells were seeded at a density of 0.8–1.1 × 105 cells/well in 24-well culture plates precoated with poly-l-lysine and were cultured for 48–72 h. Cells from stable cell lines or human SCH were either pretreated with culture medium containing CQ, monensin, bafilomycin A1, or vehicle control for the designated times or not pretreated. 0.1% DMSO was used as vehicle control for monensin and bafilomycin A1 treatment. As CQ stock (50 mM) was resolved in water, CQ-free fresh medium was used as vehicle control for CQ pretreatment. To determine the effects of CQ on OATP1B1-mediated transport after prolonged treatment, HEK293-OATP1B1 cells were preincubated with CQ-free (CTL) or 25 μM CQ-containing medium for 2 h. At the end of pre-incubation, the culture medium was aspirated and CTL cells were cultured in CQ-free medium and cells pretreated with 25 μM CQ were cultured in medium containing 1.5 μM CQ, for the indicated time, up to 24 h.
At the time of uptake experiments, after rinsing with prewarmed (37 °C) HBSS buffer (pH 7.4) three times, cells were incubated with HBSS containing [3H]E217G (1 μM, 2 min) or [3H]pitavastatin (1 μM, 0.5 min) in the absence or presence of testing drugs (CQ or rifampicin). At the end of incubation, the buffer was aspirated rapidly, and the cells were rinsed with ice-cold HBSS three times and then lysed with Triton X-100 (0.5% v/v) in DPBS. An aliquot of the lysate was subjected to liquid scintillation counting (LS6500 scintillation counter, Beckman Coulter, Brea, CA). Substrate accumulation was normalized to protein concentration determined by BCA assay (Pierce Chemical, Rockford, IL) and corrected for nonspecific binding of the substrate by including a non-overlaid poly-l-lysine coated blank plate for uptake studies in stable cell lines and a Matrigel overlaid blank plate for human SCH, respectively.
To determine [3H]E217G transport kinetic parameters, the maximal transport velocity (Vmax) and the affinity constant (Km), [3H]E217G accumulation (0.1–40 μM, 2 min) was determined in HEK293-OATP1B1 and -Mock cells following pretreatment with CQ(25 μM) or CTL. Values of [3H]E217G accumulation in Mock cells were subtracted from those in HEK293-OATP1B1 cells. The Vmax and Km values of E217G transport were estimated by fitting the Michaelis–Menten equation (eq 2) to the data using Phoenix WinNonlin, v6.3 (Certara, St. Louis, MO), where ν is E217G transport velocity and S is E217G concentration.
Publication 2016

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2019
Acquired Immunodeficiency Syndrome Angina, Unstable Arteries Cardiovascular System Carotid Arteries Cerebrovascular Accident Congestive Heart Failure Diabetes Mellitus Heart High Density Lipoprotein Cholesterol HIV-1 Hospitalization Hydroxymethylglutaryl-CoA Reductase Inhibitors Inclusion Bodies Ischemia Myopathy pitavastatin Primary Prevention Safety Transient Ischemic Attack
PLGA with an average molecular weight of 20,000 and a copolymer ratio of lactide to glycolide of 75:25 (Wako Pure Chemical Industries Ltd, Osaka, Japan) was used as a matrix for the nanoparticle, whereas polyvinylalcohol (PVA-403; Kuraray, Osaka, Japan) was used as a dispersing agent. PLGA nanoparticle incorporating fluorescent marker fluorescein-isothiocyanate (FITC; Dojin Chemical, Tokyo, Japan) (FITC-NP) or pitavastatin (Kowa Pharmaceutical Co Ltd, Tokyo, Japan) (pitavastatin-NP) was prepared by an emulsion solvent diffusion method in purified water as previously described.[12 (link)–15 (link),18 (link)–21 ] The FITC-NP and pitavastatin-NP contained 4.2% (wt/vol) FITC and 12.0% (wt/vol) pitavastatin, respectively. A sample of nanoparticle suspension in distilled water was used for particle size analysis. The diameters of FITC-NP and pitavastatin-NP were 231 nm and 159 nm, respectively. Surface charge (zeta potential) was also analyzed by Zetasizer Nano (Sysmex, Hyogo, Japan) and was anionic [-16.7 mV (FITC-NP) and -4.1 mV (pitavastatin-NP)].
Full text: Click here
Publication 2015
Diffusion dilactide Emulsions Fluorescein Fluorescein-5-isothiocyanate isothiocyanate Pharmaceutical Preparations pitavastatin Polylactic Acid-Polyglycolic Acid Copolymer Polyvinyl Alcohol Solvents

Most recents protocols related to «Pitavastatin»

Pitavastatin was chosen to be the best hit for further in vitro investigation as it fulfills all the 5 features of the designed ligand-based pharmacophore model for hIDO1/hTDO2 dual inhibition with very good binding affinities to both enzymes and interacting with the main amino acids in both enzymes’ pockets mimicking the native ligand Trp. Accordingly, Pitavastatin calcium was obtained through extraction and purification from Lipidalon tablets 4 mg from (Mash Premiere for Pharmaceutical Industries, Egypt). First, the tablets were washed with distilled water to remove the yellow coat then dried and ground into powder. Solid-liquid extraction with chloroform was performed to extract Pitavastatin. Finally, the organic solvent was evaporated leaving a white solid powder of Pitavastatin calcium. It was purified by dissolving the impurities in a solution of n-hexanes and ethyl acetate. For validation purposes, the FT-IR and melting point were measured and compared with the corresponding reported data for Pitavastatin.
Full text: Click here
Publication 2024
PD-GSCs were harvested from a T75 flask and passaged into replicate T75 flasks for either pitavastatin (6μM) or vehicle (DMSO) treatment (2.0e6 cells/flask). Concomitantly, a portion of those PD-GSCs were used to inoculate laminin-treated 96 well plates for drug-dosing analysis (see IC50Analysis section). On D4, PD-GSCs were harvested using Accutase (1mL/25cm2) as described previously. Cell suspensions were spun at 1000rpm (193g) for five minutes. Cell pellets were then resuspended with serum-free culture media (200,000 cells/mL) to inoculate 96 well plates (100μL/well, 20,000 cells/well) for subsequent IC50 determination. PD-GSCs were incubated in serum-free culture media in 96 well plates for 48 hours to allow for cell attachment prior to replacing spent media with serum-free media with pitavastatin (or vehicle). Treated cells were incubated at 37°C for four days. Following the four-day treatment, cell viability was measured via MTT assay as described below.
Publication Preprint 2024
PD-GSCs were incubated in serum-free culture media (described above) with pitavastatin (6μM). Stock pitvastatin calcium (Selleck Chemicals LLC) was dissolved in DMSO to obtain a stock concentration of 10mg/mL and stored in aliquots at −80°C. Stock pitavastatin calcium solution was serially diluted in serum-free culture media to 100μM and then to the final concentration of 6μM with a final DMSO concentration of 0.053% (v/v).
To monitor longitudinally PD-GSC response to pitavastatin, we performed a reverse time-course treatment by adding pitavastatin to SN520 and SN503 cultures in a staggered fashion such that the longest (4-day) treatment would have drug added first. Subsequent addition of pitavastatin would occur on following days for 3- and 2-day treatment, respectively. This reverse time course design allowed us to collect all samples simultaneously on day four following the initial addition of pitavastatin. Because pitavastatin was added to PD-GSCs on different days, flasks were inoculated at slightly different cell densities to account for cell growth that would occur in between inoculation and time of pitavastatin addition. Consequently, scRNA-seq library preparation of all samples for a particular PD-GSC population occurred simultaneously to minimize batch effects due to individual sample processing (table S16)
Prior to T25 flask (BioLite) inoculation for pitavastatin treatment, PD-GSCs were first expanded in a T75 flask (BioLite). Once the culture was confluent, the culture was harvested and split into laminin-treated T25 flasks. Upon inoculation, cells were incubated in serum-free culture media at 37°C for 24 hours to allow cells to adhere to the interior surface of the flask. Following the first 24 hours, serum-free culture media was replaced with serum-free culture media with pitavastatin (6μM) in T25 flasks predetermined to receive a 4-day treatment. Spent culture media would then be replaced with fresh culture media with pitavastatin (6μM) on subsequent days for D3 and D2 treatment conditions.
Upon the completion of the 4-day treatment, spent media was removed and cells were harvested using Accutase (1mL/25cm2). To prevent any cell-free DNA/RNA from treatment-induced lysed cells contaminating single-cell samples, we first processed a portion of the cell harvest solution using the dead cell removal kit (Miltenyi Biotec 130–090-101) to remove any cell debris to avoid any free RNA from lysed cells from getting mixed in with mRNA to be extracted from live cells. Samples were processed per vendor’s specifications. The result was a cell suspension of the remaining live cells post vehicle- or pitavastatin-treatment. Cell suspension was then processed for scRNA-seq profiling per the 10X Chromium platform.
Publication Preprint 2024
Each patient in pitavastatin calcium group was orally treated with 2 mg pitavastatin calcium once daily before bedtime. Each patient in atorvastatin calcium group was orally treated with 10 mg of atorvastatin calcium once daily before bedtime. The treatment cycle lasted for 3 months.
Publication 2024
To prevent the photodegradation of CPI/III, all solutions were
prepared and stored in amber-colored tubes for 2 h. Stock solutions
of pitavastatin, CPI, and CPIII in the circulated perfusate (containing
1% DMSO) were prepared at 20 μM. Spiking solutions were prepared
from alternating stock solutions. The ranges for these spiking solutions
were 0.25–20 μM pitavastatin and 0.01–2 μM
CPI/III for the perfusate and liver. For perfusate standards, spiking
solutions and 6 M formic acid (100 μL) were mixed in a 1:1 (v/v)
ratio. For liver standards, spiking solutions, liver homogenate, and
6 M formic acid were mixed in a 1:1:2 (v/v) ratio.
For bile
standards, stock solutions of pitavastatin and CPI/III were prepared
in 6 M formic acid solution (1% DMSO) at 100 and 20 μM, respectively.
They are used in alternating volumes to prepare spiking standards
(10–100 μM pitavastatin and 1–20 μM CPI/III)
and added into blank bile in a 1:1 (v/v) ratio.
For sample preparation,
100 μL of 6 M formic acid solution
was added to 100 μL of perfusate and bile samples. Liver samples
(100 μL) were first diluted with blank Krebs buffer (100 μL)
and afterward acidified with 200 μL of 6 M formic acid solution.
After the agitation with a vortex (V1 Plus, Biosan, Lithuania)
at 1400 rpm for 5 min, 1 mL of methyl tertiary butyl ether (mtbe)
was added and the mixture was immediately centrifuged at 2325g for 10 min. After the collection of the supernatant (0.5
mL), this step was repeated for an additional two rounds and 1.5 mL
of supernatant was collected from each tube. The organic solvent was
evaporated within 30–60 min using the vacuum concentrator at
4 °C. After reconstitution in the mixture of acetonitrile and
water (65:35 (v/v), 0.1% formic acid) samples, calibration standards
and quality control samples (QCs) were obtained. The concentration
ranges are given in Table 1.
Full text: Click here
Publication 2024

Top products related to «Pitavastatin»

Sourced in United States
Pitavastatin is a laboratory reagent used in research and scientific experiments. It is a type of statin that functions as an inhibitor of HMG-CoA reductase, an enzyme involved in the production of cholesterol. This product is intended for use in controlled laboratory settings by trained professionals.
Sourced in United States, Germany, China, United Kingdom, France, Sao Tome and Principe, Japan, Slovenia, Sweden, Italy
Simvastatin is a laboratory instrument used for the analysis and measurement of chemical compounds. It is designed to accurately quantify the presence and concentration of specific substances in a given sample. The core function of Simvastatin is to provide precise and reliable data for research and scientific applications.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in United States
Mevalonate is a chemical compound used in laboratory settings for research and analysis purposes. It serves as a key intermediate in the biosynthesis of various molecules, including sterols, terpenes, and isoprenoids. Mevalonate plays a critical role in cellular metabolism and is essential for the proper functioning of various biological processes.
Sourced in United States
Pitavastatin is a laboratory equipment product manufactured by Merck Group. It is used to measure the levels of pitavastatin, a medication used to lower cholesterol levels. The core function of this product is to provide accurate and reliable measurements of pitavastatin concentrations in samples.
Sourced in United States, Switzerland, Germany
Mevalonolactone is a chemical compound used as a laboratory reagent. It is a precursor in the biosynthesis of various isoprenoids, such as cholesterol, steroids, and terpenes. Mevalonolactone is commonly used in biochemical and cell biology research applications.
Sourced in United States, Germany, United Kingdom, China, Italy, Sao Tome and Principe, France, Macao, India, Canada, Switzerland, Japan, Australia, Spain, Poland, Belgium, Brazil, Czechia, Portugal, Austria, Denmark, Israel, Sweden, Ireland, Hungary, Mexico, Netherlands, Singapore, Indonesia, Slovakia, Cameroon, Norway, Thailand, Chile, Finland, Malaysia, Latvia, New Zealand, Hong Kong, Pakistan, Uruguay, Bangladesh
DMSO is a versatile organic solvent commonly used in laboratory settings. It has a high boiling point, low viscosity, and the ability to dissolve a wide range of polar and non-polar compounds. DMSO's core function is as a solvent, allowing for the effective dissolution and handling of various chemical substances during research and experimentation.
Sourced in Japan
Pitavastatin is a synthetic statin drug developed by Kowa Pharmaceutical Company. It is a cholesterol-lowering medication that works by inhibiting the enzyme HMG-CoA reductase, which plays a key role in the production of cholesterol in the body.
Sourced in United States, United Kingdom, Canada, China, Germany, Japan, Belgium, Israel, Lao People's Democratic Republic, Italy, France, Austria, Sweden, Switzerland, Ireland, Finland
Prism 6 is a data analysis and graphing software developed by GraphPad. It provides tools for curve fitting, statistical analysis, and data visualization.
Sourced in United States, United Kingdom, Germany, China, Canada, Japan, Italy, France, Belgium, Australia, Uruguay, Switzerland, Israel, India, Spain, Denmark, Morocco, Austria, Brazil, Ireland, Netherlands, Montenegro, Poland
Matrigel is a solubilized basement membrane preparation extracted from the Engelbreth-Holm-Swarm (EHS) mouse sarcoma, a tumor rich in extracellular matrix proteins. It is widely used as a substrate for the in vitro cultivation of cells, particularly those that require a more physiologically relevant microenvironment for growth and differentiation.

More about "Pitavastatin"

Pitavastatin, a synthetic statin medication, is a highly effective drug used to manage dyslipidemia (abnormal blood lipid levels) and reduce the risk of cardiovascular disease.
This statin functions by inhibiting the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, a key step in the cholesterol biosynthesis pathway.
By blocking this enzyme, pitavastatin helps lower levels of low-density lipoprotein (LDL) cholesterol, triglycerides, and increase high-density lipoprotein (HDL) cholesterol.
This lipid-lowering effect makes pitavastatin an important pharmacotherapeutic option for individuals with high blood lipid concentrations, a condition known as hyperlipidemia or hypercholesterolemia.
Pitavastatin has been shown to be well-tolerated, with potential side effects including myopathy (muscle weakness) and elevations in liver enzymes.
Researchers often use related compounds like simvastatin, as well as cell culture models with fetal bovine serum (FBS), mevalonate, mevalonolactone, and DMSO to study the mechanisms of action and effects of statins like pitavastatin.
To enhance your pitavastatin research, tools like PubCompare.ai can help you identify and compare the best protocols from scientific literature, preprints, and patents.
This innovative platform, powered by artificial intelligence, can improve the reproducibility and accuracy of your studies, allowing you to make more informed decisions and advance your understanding of this important cardiovascular therapeutic.