Lovastatin clinical pharmacology: Difference between revisions

Jump to navigation Jump to search
(Created page with "__NOTOC__ {{Lovastatin}} {{CMG}}; {{AE}} {{SS}} ==Clinical Pharmacology== ===Mechanism of Action=== Lovastatin is a lactone that is readily hydrolyzed in vivo to the corres...")
 
(Redirected page to Lovastatin#Pharmacology)
Line 1: Line 1:
__NOTOC__
#redirect [[Lovastatin#Pharmacology]]
{{Lovastatin}}
{{CMG}}; {{AE}} {{SS}}
 
==Clinical Pharmacology==
 
===Mechanism of Action===
 
Lovastatin is a lactone that is readily hydrolyzed in vivo to the corresponding β-hydroxyacid, a strong inhibitor of [[HMG-CoA]]reductase, the enzyme that catalyzes the conversion of [[HMG-CoA]]to mevalonate. The conversion of [[HMG-CoA]]to mevalonate is an early step in the biosynthetic pathway for cholesterol.
 
===Pharmacodynamics===
 
Lovastatin, as well as some of its metabolites, are pharmacologically active in humans. The liver is the primary site of action and the principal site of cholesterol synthesis and LDL clearance.
 
The involvement of low-density lipoprotein cholesterol ([[LDL-C]]) in atherogenesis has been well documented in clinical and pathological studies, as well as in many animal experiments. Epidemiological and clinical studies have established that high [[LDL-C]] and low high-density lipoprotein cholesterol ([[HDL-C]]) levels are both associated with coronary heart disease. However, the risk of developing coronary heart disease is continuous and graded over the range of cholesterol levels and many coronary events do occur in patients with total cholesterol (Total-C) and [[LDL-C]] levels in the lower end of this range.
 
Altoprev® has been shown to reduce [[LDL-C]], and Total-C. Across all doses studied, treatment with Altoprev® has been shown to result in variable reductions in triglycerides (TG), and variable increases in [[HDL-C]] [see Table 6 under Clinical Studies (14)].
 
Lovastatin immediate-release tablets have been shown to reduce both normal and elevated [[LDL-C]] concentrations. LDL is formed from very low-density lipoprotein ([[VLDL]]) and is catabolized predominantly by the high-affinity LDL receptor. The mechanism of the LDL-lowering effect of lovastatin immediate-release may involve both reduction of V[[LDL-C]] concentration, and induction of the LDL receptor, leading to reduced production and/or increased catabolism of [[LDL-C]]. Apolipoprotein B (Apo B) also falls substantially during treatment with lovastatin immediate-release. Since each LDL particle contains one molecule of Apo B, and since little Apo B is found in other lipoproteins, this strongly suggests that lovastatin immediate-release does not merely cause cholesterol to be lost from LDL, but also reduces the concentration of circulating LDL particles. In addition, lovastatin immediate-release can produce increases of variable magnitude in [[HDL-C]], and modestly reduces V[[LDL-C]] and plasma TG [see Table 7 under Clinical Studies (14)]. The independent effect of raising HDL or lowering TG on the risk of coronary and cardiovascular morbidity and mortality has not been determined. The effects of lovastatin immediate-release on lipoprotein (a) [Lp(a)], fibrinogen, and certain other independent biochemical risk markers for coronary heart disease are unknown.
 
===Pharmacokinetics===
 
Absorption: The appearance of lovastatin in plasma from an Altoprev® extended-release tablet is slower and more prolonged compared to the lovastatin immediate-release formulation.
 
A pharmacokinetic study carried out with Altoprev® involved measurement of the systemic concentrations of lovastatin (pro-drug), lovastatin acid (active-drug) and total and active inhibitors of [[HMG-CoA]]reductase. The pharmacokinetic parameters in 12 hypercholesterolemic subjects at steady state, after 28 days of treatment, comparing Altoprev® 40 mg to lovastatin immediate-release 40 mg, are summarized in Table 3.{|
|[[File:Lovastatin04.jpg|thumb|800px]]
|}
 
L=lovastatin, LA=lovastatin acid, TI=total inhibitors of [[HMG-CoA]]reductase, AI=active inhibitors of [[HMG-CoA]]reductase, Cmax=highest observed plasma concentration, Cmin=trough concentration at t=24 hours after dosing, Tmax=time at which the Cmax occurred, AUC0-24hr=area under the plasma concentration-time curve from time 0 to 24 hr after dosing, calculated by the linear trapezoidal rule.
 
* Administered at bedtime.
** Administered with the evening meal.
 
The mean plasma concentration-time profiles of lovastatin and lovastatin acid in patients after multiple doses of Altoprev® or lovastatin immediate-release at day 28 are shown in Figure 1.
 
{|
|[[File:Lovastatin05.jpg|thumb|800px]]
|}
 
The extended-release properties of Altoprev® are characterized by a prolonged absorptive phase, which results in a longer Tmax and lower Cmax for lovastatin (pro-drug) and its major metabolite, lovastatin acid, compared to lovastatin immediate-release.
 
The bioavailability of lovastatin (pro-drug) as measured by the AUC0-24hr was greater for Altoprev® compared to lovastatin immediate-release (as measured by a chemical assay), while the bioavailability of total and active inhibitors of [[HMG-CoA]]reductase were equivalent to lovastatin immediate-release (as measured by an enzymatic assay).
 
With once-a-day dosing, mean values of AUCs of active and total inhibitors at steady state were about 1.8-1.9 times those following a single dose. Accumulation ratio of lovastatin exposure was 1.5 after multiple daily doses of Altoprev® compared to that of a single dose measured using a chemical assay. Altoprev® appears to have dose linearity for doses from 10 mg up to 60 mg per day.
 
When Altoprev® was given after a meal, plasma concentrations of lovastatin and lovastatin acid were about 0.5 - 0.6 times those found when Altoprev® was administered in the fasting state, indicating that food decreases the bioavailability of Altoprev®. There was an association between the bioavailability of Altoprev® and dosing after mealtimes. Bioavailability was lowered under the following conditions, (from higher bioavailability to lower bioavailability) in the following order: under overnight fasting conditions, before bedtime, with dinner, and with a high fat breakfast. In a multicenter, randomized, parallel group study, patients were administered 40 mg of Altoprev® at three different times; before breakfast, after dinner and at bedtime. Although there was no statistical difference in the extent of lipid change between the three groups, there was a numerically greater reduction in [[LDL-C]] and TG and an increase in [[HDL-C]] when Altoprev® was administered at bedtime. Results of this study are displayed in Table 4.
{|
|[[File:Lovastatin06.jpg|thumb|800px]]
|}
 
At steady state in humans, the bioavailability of lovastatin, following the administration of Altoprev®, was 190% compared to lovastatin immediate-release.
 
Lovastatin Immediate-Release
Absorption of lovastatin, estimated relative to an intravenous reference dose in each of four animal species tested, averaged about 30% of an oral dose. Following an oral dose of 14C-labeled lovastatin in man, 10% of the dose was excreted in urine and 83% in feces. The latter represents absorbed drug equivalents excreted in bile, as well as any unabsorbed drug. In a single dose study in four hypercholesterolemic patients, it was estimated that less than 5% of an oral dose of lovastatin reaches the general circulation as active inhibitors.
 
Distribution: Both lovastatin and its β-hydroxyacid metabolite are highly bound (>95%) to human plasma proteins. Animal studies demonstrated that lovastatin crosses the blood-brain and placental barriers.
 
In animal studies, after oral dosing, lovastatin had high selectivity for the liver, where it achieved substantially higher concentrations than in non-target tissues.
 
Lovastatin undergoes extensive first-pass extraction in the liver, its primary site of action, with subsequent excretion of drug equivalents in the bile. As a consequence of extensive hepatic extraction of lovastatin, the availability of drug to the general circulation is low and variable.
 
Metabolism: Metabolism studies with Altoprev® have not been conducted.
 
Lovastatin
Lovastatin is a lactone that is readily hydrolyzed in vivo to the corresponding β-hydroxyacid, a strong inhibitor of [[HMG-CoA]]reductase. Inhibition of [[HMG-CoA]]reductase is the basis for an assay in pharmacokinetic studies of the β-hydroxyacid metabolites (active inhibitors) and, following base hydrolysis, active plus latent inhibitors (total inhibitors) in plasma following administration of lovastatin.
 
The major active metabolites present in human plasma are the β-hydroxyacid of lovastatin, its 6’-hydroxy derivative, and two additional metabolites. The risk of [[myopathy]] is increased by high levels of [[HMG-CoA]]reductase inhibitory activity in plasma. Strong inhibitors of CYP3A can raise the plasma levels of [[HMG-CoA]]reductase inhibitory activity and increase the risk of [[myopathy]] [see Warnings and Precautions (5.1), Drug Interactions (7)].
 
Lovastatin is a substrate for CYP3A4 [see Drug Interactions (7)]. Grapefruit juice contains one or more components that inhibit CYP3A and can increase the plasma concentrations of drugs metabolized by CYP3A4. In one study1, 10 subjects consumed 200 mL of double-strength grapefruit juice (one can of frozen concentrate diluted with one rather than 3 cans of water) three times daily for 2 days and an additional 200 mL double-strength grapefruit juice together with and 30 and 90 minutes following a single dose of 80 mg lovastatin on the third day. This regimen of grapefruit juice resulted in mean increases in the concentration of lovastatin and its beta-hydroxyacid metabolite (as measured by the area under the concentration-time curve) of 15-fold and 5-fold respectively (as measured using a chemical assay – liquid chromatography/tandem mass spectrometry). In a second study, 15 subjects consumed one 8 oz glass of single-strength grapefruit juice (one can of frozen concentrate diluted with 3 cans of water) with breakfast for 3 consecutive days and a single dose of 40 mg lovastatin in the evening of the third day. This regimen of grapefruit juice resulted in a mean increase in the plasma concentration (as measured by the area under the concentration-time curve) of active and total [[HMG-CoA]]reductase inhibitory activity [using a validated enzyme inhibition assay different from that used in the first study, both before (for active inhibitors) and after (for total inhibitors) base hydrolysis] of 1.34-fold and 1.36-fold, respectively, and of lovastatin and its β-hydroxyacid metabolite (measured using a chemical assay – liquid chromatography/tandem mass spectrometry) of 1.94-fold and 1.57-fold, respectively. The effect of amounts of grapefruit juice between those used in these two studies on lovastatin pharmacokinetics has not been studied.
 
{|
|[[File:Lovastatin07.jpg|thumb|800px]]
|}
 
*      Results based on a chemical assay.
 
†      Lovastatin acid refers to the β-hydroxyacid of lovastatin.
‡      The mean total AUC of lovastatin without itraconazole phase could not be determined accurately. Results could be representative of strong CYP3A inhibitors such as ketoconazole, posaconazole, clarithromycin, telithromycin, HIV protease inhibitors, and nefazodone.
§      Estimated minimum change.
¶      The effect of amounts of grapefruit juice between those used in these two studies on lovastatin pharmacokinetics has not been studied.
#       Double-strength: one can of frozen concentrate diluted with one can of water. Grapefruit juice was administered TID for 2 days, and 200 mL together with single dose lovastatin and 30 and 90 minutes following single dose lovastatin on Day 3.
Þ      Single-strength: one can of frozen concentrate diluted with 3 cans of water. Grapefruit juice was administered with breakfast for 3 days, and lovastatin was administered in the evening on Day 3.
ß      Cyclosporine-treated patients with psoriasis or post kidney or heart transplant patients with stable graft function, transplanted at least 9 months prior to study.
à      ND = Analyte not determined.
è      Lactone converted to acid by hydrolysis prior to analysis. Figure represents total unmetabolized acid and lactone.
 
Excretion: In a single-dose study with Altoprev®, the amounts of lovastatin and lovastatin acid excreted in the urine were below the lower limit of quantitation of the assay (1.0 ng/mL), indicating that negligible excretion of Altoprev® occurs through the kidney.
 
Lovastatin
Lovastatin undergoes extensive first-pass extraction in the liver, its primary site of action, with subsequent excretion of drug equivalents in the bile.
 
====Specific Populations====
 
Geriatric: Lovastatin Immediate-Release
 
In a study with lovastatin immediate-release which included 16 elderly patients between 70-78 years of age who received lovastatin immediate-release 80 mg/day, the mean plasma level of [[HMG-CoA]]reductase inhibitory activity was increased approximately 45% compared with 18 patients between 18-30 years of age [see Use in Specific Populations (8.5)].
 
Pediatric: Pharmacokinetic data in the pediatric population are not available.
 
Gender: In a single dose pharmacokinetic study with Altoprev®, there were no statistically significant differences in pharmacokinetic parameters between men (n=12) and women (n=10), although exposure tended to be higher in men than women.
 
In clinical studies with Altoprev®, there was no clinically significant difference in [[LDL-C]] reduction between men and women.
 
Renal Impairment: In a study of patients with severe renal impairment (creatinine clearance 10-30 mL/min), the plasma concentrations of total inhibitors after a single dose of lovastatin were approximately two-fold higher than those in healthy volunteers.
 
Hemodialysis: The effect of [[hemodialysis]] on plasma levels of lovastatin and its metabolites have not been studied.
 
Hepatic Impairment: No pharmacokinetic studies with Altoprev® have been conducted in patients with hepatic impairment.<ref name="dailymed.nlm.nih.gov">{{Cite web  | last =  | first =  | title = ALTOPREV (LOVASTATIN) TABLET, EXTENDED RELEASE [SHIONOGI INC.] | url = http://dailymed.nlm.nih.gov/dailymed/lookup.cfm?setid=e8b6ccc2-e6e7-45fd-ab66-c312bcbe2b01 | publisher =  | date =  | accessdate = 13 February 2014 }}</ref>
 
==References==
{{Reflist|2}}
 
{{Statins}}


[[Category:Statins]]
[[Category:Statins]]
[[Category:Carboxylate esters]]
[[Category:Carboxylate esters]]
[[Category:Lactones]]
[[Category:Lactones]]
[[Category:Cardiovascular Drug]]
[[Category:Cardiovascular Drugs]]
[[Category:Drug]]
[[Category:Drug]]

Revision as of 14:09, 21 July 2014