Pitavastatin

Pitavastatin − pharmacological profile from early phase studies

Alberico L. Catapano*
Professor of Pharmacology and Director, Centre for the Study of Atherosclerosis, Department of Pharmacological Sciences, University of Milan, and IRCSS Multimedica, Sesto S. Giovanni, Milan, Italy

Abstract

Pitavastatin has been designed as a synthetic 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor with a novel cyclopropyl moiety that results in several differences compared to other statins. These include effective inhibition of cholesterol synthesis and increased lipoprotein lipase expression at lower doses than other statins, and significant high-density lipoprotein-cholesterol and apolipoprotein A1-elevating activity that persists with time.

The safety, tolerability and pharmacokinetics of pitavastatin and its major metabolite, pitavastatin lactone, have been investigated in a variety of patient groups with similar results, which suggests dosage adjustments are not required for gender, age or race. In healthy subjects, pitavastatin is well tolerated at the approved doses with no serious adverse events. The bioavailability of pitavastatin is, at 60%, higher than that of any other statin and the majority of the bioavailable fraction of an oral dose is excreted unchanged in the bile. The entero-hepatic circulation of unchanged drug contributes to the prolonged duration of action and allows once-daily, any-time dosing.
Pitavastatin is only slightly metabolised by cytochrome P450 (CYP) 2C9 and not at all by CYP3A4. Neither pitavastatin nor its lactone form, have inhibitory effects on CYP, and CYP3A4 inhibitors have no effect on pitavastatin concentrations. Moreover, P-glycoprotein- mediated transport does not play a major role in the drug’s disposition and pitavastatin does not inhibit P-glycoprotein activity. Pitavastatin is transported into the liver by several hepatic transporters but OATP1B1 inhibitors have relatively little effect on plasma concentrations compared with other statins. In general, interactions, except with multi-transporter inhibitors like ciclosporin, are not clinically significant. Consequently, pitavastatin has minimal drug–food and drug–drug interactions making it a treatment option in the large group of dyslipidaemic people that require multidrug therapy.

Keywords: Pitavastatin; Statins; Dyslipidaemia; Hypercholesterolaemia; Drug–drug interactions

1. Introduction

Pitavastatin calcium (NK-104) was designed as a novel 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) re- ductase inhibitor (statin) with a synthetic cyclopropyl side group (Fig. 1). This unique structure contributes to a number of pharmacological benefits compared to other statins, including inhibition of cholesterol synthesis at lower doses than the commonly-used statins, minimal metabolism leading to increased bioavailability and an extended duration of action, and a unique metabolic profile that reduces the risk of drug–food and drug–drug interactions (DDIs) [1−4].

2. Mechanism of action

Statins are structurally similar to HMG-CoA and generally bind to HMG-CoA reductase with a several thousand-fold greater affinity than the natural substrate [5]. This inhibits mevalonate production from HMG-CoA and reduces the level of intracellular cholesterol, thereby stimulating low- density lipoprotein- (LDL-) receptor activity and increasing the uptake of non-high-density lipoprotein (non-HDL) particles from the systemic circulation.

In vitro studies show that pitavastatin binds to HMG- CoA reductase with a 1.6- and 3.5-fold greater affinity than simvastatin or pravastatin, respectively [4], and that it in- hibits HMG-CoA reductase in a competitive, concentration- dependent manner that is 2.4 times more potent than simvastatin and 6.8 times more potent than pravastatin [3]. Moreover, studies in HepG2 cells using radio-labelled LDL show that pitavastatin enhances LDL-receptor binding and LDL-internalisation in a dose-dependent manner, with a 2.7-fold increase in binding at 10−6 M (P < 0.01 vs. no pitavastatin) and a 2.4-fold increase in internalisation at 10−6 M (P < 0.01) [6]. Similarly, a further study showed that pitavastatin was associated with increased LDL-receptor mRNA expression in HepG2 cells – an effect that was significantly more potent than that produced by either atorvastatin or simvastatin (P < 0.01 for both) [4]. Overall, these results are consistent with those from clinical trials, which demonstrate that pitavastatin effectively reduces LDL-C levels at lower doses than other statins [7−16]. In addition to reducing the level of cholesterol synthesis and increasing LDL uptake from the circulation, in vitro studies in HepG2 cells show that pitavastatin promotes the degradation of newly synthesised apolipoprotein B (apoB) [17], reduces very low-density lipoprotein-C (VLDL-C) and apoB secretion [18], and increases lipoprotein lipase activity by 30% more than other statins [19]. Moreover, pitavastatin appears to have a consistent beneficial effect on HDL-C levels [20]. An ELISA-based study in HepG2 cells, for example, showed that pitavastatin 3 mM dose- dependently induced expression of apoAI – the major pro- tein component of HDL-C – to a greater extent than either simvastatin 10 mM or atorvastatin 30 mM [20]. This effect was inhibited by the addition of mevanolate, indicating an HMG-CoA reductase inhibition-dependent mechanism. Furthermore, pitavastatin increased ATP-binding cassette transporter ABCA1 (cholesterol efflux regulatory protein) mRNA expression, thereby promoting apoA1 and apoE lipidation and protecting apoA1 from catabolism, and suppressed Rho and Rho kinase activity, thereby increasing apoA1 levels via a third pathway. Since low levels of HDL-C are associated with a higher risk of coronary heart disease, even in patients with ‘normal’ (<1.8 mmol/L; <70 mg/dL) LDL-C [21−23], pitavastatin has the potential to reduce CV risk by increasing HDL-C levels as well as by reducing total cholesterol (TC), LDL-C, apoB, and triglyceride (TG) concentrations. 3. Pharmacokinetic profile of pitavastatin The pharmacokinetic profiles of statins vary considerably between drugs. For example, systemic bioavailability ranges from 5% with simvastatin and lovastatin to >60% with pitavastatin (Table 1), depending on the extent of first-pass metabolism and variations in the activity of intestinal and hepatic transport proteins [2]. Pitavastatin’s oral absorption is, at 80%, second only to fluvastatin and protein binding varies from >95% for pitavastatin, simvastatin, atorvastatin and lovastatin, to 50% for pravastatin [24,25]. However, the most important differences between pitavastatin and other statins are due to metabolism. Whereas lovastatin, simvastatin and atorvastatin are metabolised predominantly by cytochrome P450 (CYP)3A4 and fluvastatin and rosuvas- tatin are metabolised by CYP2C9, pitavastatin’s cyclopropyl group diverts the drug away from metabolism by CYP3A4 and allows only a small amount of clinically insignificant metabolism by CYP2C9 (Fig. 2) [25−29]. As a result, most of the bioavailable fraction of an oral dose of pitavastatin is excreted unchanged in the bile and is then ready for entero- hepatic circulation by reabsorption in the small bowel. Less than 5% of a dose of pitavastatin is excreted in the urine.

Fig. 2. Pitavastatin and pitavastatin lactone: Distribution, metabolism and mechanisms of potential drug–drug interactions (modified from [30]). The drugs listed are substrates for the pathways indicated and therefore have the potential to interact with pitavastatin. OATP, organic anion transporting polypeptide; MDR1, multidrug resistance associated protein 1; NTCP, sodium taurocholate cotransporting polypeptide; BCRP, breast cancer resistance protein.

Most statins are administered orally as an active acidic form that is biotransformed by glucuronosyltransferase (UGT) to a very unstable compound. This compound is then quickly converted to a lactone metabolite, which is rapidly metabolised by CYP450 isoenzymes [25,27,30]. In human hepatic microsomes the metabolic clearance of the lactone metabolites of atorvastatin, simvastatin, and rosuvastatin is between 64- and 73-fold higher than the biotransformation of the acid forms of these statins. The metabolic clearance of the acid and lactone forms of atorvastatin and simvas- tatin, for example, are 26 and 1892 ml/min/mg protein, re- spectively, and 28 and 1959 ml/min/mg protein. In contrast, both pitavastatin acid and lactone show little metabolism in human hepatic microsomes (metabolic clearance 3 and 5 ml/min/mg protein, respectively) [27]. Thus, pitavastatin has a unique metabolic profile compared to other statins that contributes to an increased bioavailability, a longer duration of action and a lower probability of drug–food or drug–drug interactions.

4. Safety and tolerability

The safety, tolerability and pharmacokinetics of pitavastatin and pitavastatin lactone have been investigated in a number of different patient groups with similar results [31]. In healthy Caucasian subjects (N = 71), pitavastatin 1−64 mg or placebo was administered under fasted conditions on Day 1, followed by a 7-day washout period and then once daily dosing under fed conditions for 2 weeks. After multiple
dosing, pitavastatin was rapidly absorbed with a median Tmax of 1.0−1.8 hours. The Cmax and AUC for pitavastatin and pitavastatin lactone increased in an approximately dose- proportional manner for both single and multiple once-daily administrations and there was no relevant drug accumula- tion (mean AUC0−24 Day 21 : AUC0−24 Day 8 ratios ranged from 1.0 to 1.5). The mean t1/ 2 was 8−9 hours after a single-dose administration and 9−12 hours after multiple dosing, suggesting there was no relevant accumulation after repeated, once-daily doses. A steady-state was reached after approximately 4 days. In this study, pitavastatin 1−64 mg was well tolerated with no serious adverse events (AEs) and no significant changes in safety or laboratory parameters, including alanine transaminase, aspartate transaminase, and creatinine kinase.

5. Pitavastatin has a low potential for DDIs

To attain their recommended LDL-C targets, patients with dyslipidaemia and high CV risk often require combinations of lipid-lowering agents that include statins, ezetimibe, niacin, and/or bile acid sequestrants. Furthermore, since dyslipidaemia frequently coexists with conditions such as hypertension, diabetes, and renal disease, additional agents for the treatment of chronic conditions are often used alongside these drugs. Combinations of certain drugs can increase the plasma concentration of statins [32] and increase the risk of statin-related AEs, including myopa- thy and rhabdomyolysis [33]. Whereas most statins are rapidly metabolised by CYP isoenzymes – in particular CYP3A4 – pitavastatin acid undergoes only a small amount of metabolism by CYP2C9 and UGT and none at all by CYP3A4 (Fig. 2) [26,27]. Consequently, pitavastatin at the recommended doses for clinical use is likely to have a better DDI profile than other statins.

In general, substrates for CYP2C9 and UGT, such as warfarin [34], fenofibrate, and rifampicin do not interact significantly with pitavastatin [24]. Although fenofibrate increases pitavastatin’s steady state AUC0−24 by 18%, concurrent administration is well-tolerated and the interac- tion is not clinically significant [35]. Furthermore, whereas the co-administration of CYP3A4 inhibitors with other statins causes significant clinical effects, co-administration with pitavastatin does not. Concurrent administration of atorvastatin with grapefruit juice (a CYP3A4 inhibitor), for example, increases the mean AUC0−24 by 83% for atorvastatin and by only 13% for pitavastatin [36].

Pitavastatin is transported from the circulation into the liver by organic anion-transporting polypeptides (OATPs) and from the liver to the bile duct by Breast Cancer Resistance Protein (BCRP), and for the lactone, Multi- drug Resistance Associated Protein 1 (MDR1) (Fig. 2) [28,29,37]. Whereas interactions between MDR and sim- vastatin/atorvastatin are thought to be responsible for the increased risk of rhabdomyolysis in people taking these statins with digoxin [38,39], drugs that either inhibit or compete with MDR do not interact significantly with pitavastatin [24]. In contrast, pitavastatin has limited inter- actions with a number of OATP1B1 inhibitors. Gemfibrozil, for example, inhibits OATP1B1 and increases the AUC of pitavastatin 1.4-fold [40]. However, given the interactions between gemfibrozil and lovastatin (3.8-fold increase in AUC), simvastatin (1.9-fold), pravastatin (2.2-fold) and rosuvastatin (1.9-fold) [41], pitavastatin appears to be less dependent on OATP1B1 transportation than other statins. This observation is also reflected by the interaction between ciclosporin and statins. Co-administration of pitavastatin and ciclosporin, for example, increased the mean AUC0−24 of pitavastatin by 4.6-fold compared to 8.7- and 7-fold for atorvastatin and rosuvastatin, respectively [40]. Whilst the pharmacokinetic interaction between ciclosporin and pitavastatin is less marked than with other statins, there is not enough clinical data to demonstrate safety and the co- administration of ciclosporin with pitavastatin is currently contraindicated. Nevertheless, most studies demonstrate that pitavastatin has a lower potential for DDIs than the most commonly used statins, making it an excellent choice for dyslipidaemic patients that require polypharmacy.

6. Conclusions

Early phase studies suggest that pitavastatin’s unique cyclo- propyl side-group offers a number of pharmacokinetic bene- fits compared to other statins [1−4]. These include a greater inhibition of HMG-CoA reductase [3] and cholesterol syn- thesis [4], increased LDL-receptor activity, and increased HDL-C production in HepG2 cells [20]. These observations, together with pitavastatin’s minimal metabolism and high bioavailability, explain why pitavastatin achieves significant LDL-C reductions and HDL-C increases at doses lower than many other statins. Unlike other statins, pitavastatin’s cyclopropyl group diverts the drug away from metabolism by CYP isoenzymes and, although pitavastatin undergoes a small amount of clinically insignificant metabolism by CYP2C9, it is not metabolised by CYP3A4 and therefore has a reduced risk of DDIs. This metabolic stability is likely to have significant benefits in the vast numbers of dyslipidaemic patients that require additional medications for concomitant conditions, such as hypertension, diabetes, and acute coronary syndromes. Thus, early phase studies suggest that pitavastatin has potential as a useful addition to the statin family.

Conflict of interest statement

Prof. Catapano has received speaker fees from AstraZeneca, Merck, Schering-Plough, Pfizer, sanofi-aventis, and Kowa.

References

[1] Saito Y. Critical appraisal of the role of pitavastatin in treating dyslipidemias and achieving lipid goals. Vasc Health Risk Manag 2009;5:921−36.
[2] Mukhtar RY, Reid J, Reckless JP. Pitavastatin. Int J Clin Pract 2005;59:239−52.
[3] Aoki T, Nishimura H, Nakagawa S, et al. Pharmacological profile of a novel synthetic inhibitor of 3-hydroxy-3-methylglutaryl- coenzyme A reductase. Arzneimittelforschung 1997;47:904−9.
[4] Morikawa S, Umetani M, Nakagawa S, et al. Relative induction of mRNA for HMG CoA reductase and LDL receptor by five different HMG-CoA reductase inhibitors in cultured human cells. J Atheroscler Thromb 2000;7:138−44.
[5] Corsini A, Bellosta S, Baetta R, Fumagalli R, Paoletti R, Bernini F. New insights into the pharmacodynamic and pharmacokinetic properties of statins. Pharmacol Ther 1999;84:413−28.
[6] Tamaki T, Nakagawa S, Tanabe S. Effect of NK-104 on lipid metabolism in HepG2 vcells: inhibition of cholesterol synthesis and enhancement of LDL receptor activity. Tokyo Research Laboratories, editor. R99045. 1994. Tokyo, Japan, Kowa Company Ltd. 1−1–2010.
[7] Budinski D, Arneson V, Hounslow N, Gratsiansky N. Pitavastatin compared with atorvastatin in primary hypercholesterolemia or combined dyslipidemia. Clin Lipidol 2009;4:291–302.
[8] Study of pitavastatin 4 mg vs. simvastatin 40 mg (following up-titration) in patients with primary hypercholesterolemia or
combined dyslipidemia and 2 or more risk factors for coronary heart disease. NK-104-304 clinical study report. Data on file (Kowa).
[9] Ose L, Budinski D, Hounslow N, Arneson V. Comparison of pitavastatin with simvastatin in primary hypercholesterolaemia or combined dyslipidaemia. Curr Med Res Opin 2009;25:2755−64.
[10] Ose L, Budinski D, Hounslow N, Arneson V. Long-term treatment with pitavastatin is effective and well tolerated by patients
with primary hypercholesterolemia or combined dyslipidemia. Atherosclerosis 2010;210:202−8.
[11] Hounslow N, Budinski D, Eriksson M. Pitavastatin 4 mg shows comparable LDL-C and superior triglyceride reduction to simvastatin 40 mg in high-risk primary hypercholesterolemia or combined dyslipidaemia. EAS 2010 (poster).
[12] Stender S, Hounslow N. Robust efficacy of pitavastatin and comparable safety to pravastatin. Atheroscler Suppl 2009;10:P770.
[13] Yokote K, Bujo H, Hanaoka H, et al. Multicenter collaborative randomized parallel group comparative study of pitavastatin and atorvastatin in Japanese hypercholesterolemic patients: collaborative study on hypercholesterolemia drug intervention and their benefits for atherosclerosis prevention (CHIBA study). Atherosclerosis 2008;201:345−52.
[14] Sasaki J, Ikeda Y, Kuribayashi T, et al. A 52-week, randomized, open-label, parallel-group comparison of the tolerability and effects of pitavastatin and atorvastatin on high-density lipoprotein
cholesterol levels and glucose metabolism in Japanese patients with elevated levels of low-density lipoprotein cholesterol and glucose intolerance. Clin Ther 2008;30:1089–101.
[15] Kurihara Y, Douzono T, Kawakita K, Nagasaka Y. A large-scale, long-term prospective post-marketing surveillance of pitavastatin (Livalo) – Livalo effectiveness and safety study (LIVES). Jpn Pharmacol Ther 2008;36:709−31.
[16] Betteridge J. Pitavastatin − results from phase III & IV. Atheroscl Suppl 2010;11(3):8−14.
[17] Ooyen C, Zecca A, Bersino AM, Catapano AL. NK-104, a potent 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, decreases apolipoprotein B-100 secretion from Hep G2 cells. Atherosclerosis 1999;145:87−95.
[18] Pitavastatin. Itavastatin, Nisvastatin, Nk 104, Nks 104, P 872441. Drugs R D 2002;3:58−60.
[19] Saiki A, Murano T, Watanabe F, Oyama T, Miyashita Y, Shirai K. Pitavastatin enhanced lipoprotein lipase expression in 3T3-L1 preadipocytes. J Atheroscler Thromb 2005;12:163−8.
[20] Maejima T, Yamazaki H, Aoki T, et al. Effect of pitavastatin on apolipoprotein A-I production in HepG2 cell. Biochem Biophys Res Commun 2004;324:835−9.
[21] Barter P, Gotto AM, LaRosa JC, et al. HDL cholesterol, very low levels of LDL cholesterol, and cardiovascular events. N Engl J Med 2007;357:1301−10.
[22] Teramoto T, Shimano H, Yokote K, Urashima M. Effects
of pitavastatin (LIVALO Tablet) on high density lipoprotein cholesterol (HDL-C) in hypercholesterolemia. J Atheroscler Thromb 2009;16:654−61.
[23] Goldenberg I, Boyko V, Tennenbaum A, Tanne D, Behar S,
Guetta V. Long-term benefit of high-density lipoprotein cholesterol- raising therapy with bezafibrate: 16-year mortality follow-up
of the bezafibrate infarction prevention trial. Arch Intern Med 2009;169:508−14.
[24] Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid- lowering drugs: mechanisms and clinical relevance. Clin Pharmacol Ther 2006;80:565−81.
[25] Fujino H, Yamada I, Shimada S, Nagao T, Yoneda M. Metabolic fate of pitavastatin (NK-104), a new inhibitor of 3-hydroxy-
3-methyl-glutaryl coenzyme A reductase. Effects on drug- metabolizing systems in rats and humans. Arzneimittelforschung 2002;52:745−53.
[26] Fujino H, Yamada I, Shimada S, Yoneda M, Kojima J. Metabolic fate of pitavastatin, a new inhibitor of HMG-CoA reductase: human UDP-glucuronosyltransferase enzymes involved in lactonization. Xenobiotica 2003;33:27−41.
[27] Fujino H, Saito S, Tsunenari Y, Kojima J, Sakaeda T. Metabolic properties of the acid and lactone forms of HMG-CoA reductase inhibitors. Xenobiotica 2004;34:961−71.
[28] Hirano M, Maeda K, Shitara Y, Sugiyama Y. Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther 2004;311:139−46.
[29] Hirano M, Maeda K, Matsushima S, Nozaki Y, Kusuhara H, Sugiyama Y. Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol Pharmacol 2005;68:800−7.
[30] Ose L. Pitavastatin: A distinctive lipid lowering drug. Clin Lipidol 2010;5(3):309−23.
[31] NK-104 Pitavastatin Investigators’ Brochure. 2007. Tokyo, Japan, Kowa Company, Ltd.
[32] Williams D, Feely J. Pharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitors. Clin Pharmacokinet 2002;41:343−70.
[33] Evans M, Rees A. Effects of HMG-CoA reductase inhibitors on skeletal muscle: are all statins the same? Drug Saf 2002;25:649−63.
[34] Inagaki Y, Hunt T, Arana B, Gosho M, Morgan R. Drug– drug interaction study to assess the effects of multiple dose of
pitavastatin on steady state warfarin in healthy adult volunteers. Atheroscler Suppl 2009;10:e806.
[35] Mathew P, Cuddy T, Tracewell WG, Salazar D. An open-label study on the pharmacokinetics (PK) of pitavastatin (NK-104) when administered concomitantly with fenofibrate or gemfibrozil in healthy volunteers. Clin Pharmacol Ther 2004;75:P33.
[36] Ando H, Tsuruoka S, Yanagihara H, et al. Effects of grapefruit juice on the pharmacokinetics of pitavastatin and atorvastatin. Br J Clin Pharmacol 2005;60:494−7.
[37] Fujino H, Saito T, Ogawa S, Kojima J. Transporter-mediated influx and efflux mechanisms of pitavastatin, a new inhibitor of HMG- CoA reductase. J Pharm Pharmacol 2005;57:1305−11.
[38] Bizzaro N, Bagolin E, Milani L, Cereser C, Finco B. Massive rhabdomyolysis and simvastatin. Clin Chem 1992;38:1504.
[39] Boyd RA, Stern RH, Stewart BH, et al. Atorvastatin coadministration may increase digoxin concentrations by inhibition of intestinal P-glycoprotein-mediated secretion. J Clin Pharmacol 2000;40:91−8.
[40] Hasunuma T, Masahiko N, Takashi Y, et al. The drug–drug Interactions of pitavastatin (NK-104), a novel HMG-CoA reductase Inhibitor and cyclosporine. J Clin Ther Med 2003;19:381−9.
[41] Shitara Y, Hirano M, Sato H, Sugiyama Y. Gemfibrozil and its glucuronide inhibit the organic anion transporting polypeptide 2 (OATP2/OATP1B1:SLC21A6)-mediated hepatic uptake and CYP2C8-mediated metabolism of cerivastatin: analysis of the mechanism of the clinically relevant drug–drug interaction between cerivastatin and gemfibrozil. J Pharmacol Exp Ther 2004;311:228−236.