Statins and PCSK9 Inhibitors: Defining the Correct Patients

A332563_1_En_9_Fig1b_HTML.gif



Fig. 9.1
Role of PCSK9 in the regulation of LDL-C concentrations. (a) PCSK9 binds to LDL Receptor (LDLR) and, upon internalization of the LDL/LDLR complex, directs LDLR to lysosomal degradation, decreasing the number of LDLR at the surface of hepatocyte. (b) Statin therapy, via SERBP2 activation, stimulates both LDLR and PCSK9 expression. Anti-PCSK9 mAb prevents binding of PCSK9 to the LDL/LDLR complex



Beyond the regulation of LDLR concentrations, data support an effect of PCSK9 on lipoprotein assembly and secretion, with emerging evidence of a role in the metabolism of triglyceride-rich lipoproteins and triglyceride accumulation in visceral adipose tissue [23, 24]. The function of PCSK9 in the intestine is not completely known: PCSK9 null mice are protected from postprandial hypertriglyceridemia [25]. PCSK9 can enhance chylomicron secretion and participate in the control of enterocyte cholesterol balance [2628].



Impact of PCSK9 on Atherosclerosis


In mice fed high-fat high-cholesterol diet, gene inactivation of PCSK9 significantly reduced the accumulation of cholesteryl esters in aortas, which was markedly increased by overexpression of PCSK9 resulting in accelerated development of atherosclerotic plaques [29]. Interestingly, in LDLR-deficient mice lacking or overexpressing PCSK9, no significant differences were observed in cholesteryl ester accumulation and plaque size, strongly suggesting that the process by which PCSK9 enhances atherosclerosis is primarily mediated through its action on the LDLR [29].

Cloned minipigs created by transposition of a human PCSK9 gain-of-function mutant – a model for familial hypercholesterolemia (FH) – had a significant increase in aortic atherosclerosis compared with wild-type minipigs [30]. At the opposite, inhibition of PCSK9 by a mAb, alirocumab, reduced atherosclerosis development in ApoE3Leiden. CETP mice model, and enhanced the beneficial effects of atorvastatin [31], providing an argument for statin and PCSK9 combination therapy.

The impact of PCSK9 inhibition on atherosclerotic plaques is currently evaluated by intravascular ultrasound in a large phase III trial with evolocumab, GLAGOV (GLobal Assessment of plaque reGression with a PCSK9 antibOdy as measured by intraVascular ultrasound) [32].


Rationale of Statin and PCSK9 Inhibitor Combination Therapy


PCSK9 gene expression is mainly modulated by intracellular cholesterol concentrations and consequent activation of the transcription factor sterol-responsive element-binding protein 2 (SREBP2) [21] (Fig. 9.1b), similarly to other genes involved in cholesterol homeostasis, such as LDLR. The relationship between statin treatment and PCSK9 secretion has been investigated in animals and humans. In hepatic cell lines, statins upregulated the mRNA expression of LDLR and PCSK9 [33]. Statin administration to PCSK9 knockout mice enhanced LDL clearance from plasma [21]. In humans, statins induced a dose-dependent increase in the concentration of plasma PCSK9 [34, 35]. This concomitant regulation of both PCSK9 and LDLR by cholesterol via SREBP2 helps to explain the paradoxical effect of statin therapy [36], potentially limiting the pharmacological effect of statin on LDL-C concentration. This was confirmed in patients with LOF mutations in PCSK9 gene who are more responsive to statin therapy [37]. These data support PCSK9 inhibition as a very attractive target for lowering LDL-C and enhancing the efficacy of statin treatment.


Strategies for PCSK9 Inhibition


Several therapeutic approaches to the inhibition of PCSK9 have been proposed [38], including inhibition of PCSK9 synthesis by gene-silencing agents, such as antisense oligonucleotides (ASOs) or small interfering RNA (siRNA); inhibition of PCSK9 binding to LDLR by mAbs, small peptides, or adnectins; and inhibition of PCSK9 autocatalytic processing by small molecule inhibitors. These strategies targeting either extracellular or intracellular PCSK9 have been extensively described in recent reviews [3943].

Preclinical studies on inhibition of PCSK9 synthesis by ASOs were promising, but the development of two ASOs by BMS/ISIS (BMS-84421) and Santaris Pharma (SPC5001) was stopped in Phase I [43]. siRNA is another approach [44, 45]. In rats, siRNA targeting PCSK9 reduced LDL-C level by around 30 %, and in nonhuman primates, single-dose administration of 5 mg of the drug decreased LDL-C by 56–70 % [44]. In a phase I trial of ALN-PCS, an siRNA developed by Alnylam Pharmaceuticals, a dose-dependent reduction in LDL-C was observed, with a 40 % reduction with the highest dose, associated with a 70 % reduction in plasma PCSK9 concentrations [46]. Inhibition of PCSK9 binding to LDLR by small peptide inhibitors such as SX-PCK9 (Serometrix, East Syracuse, NY, USA) or adnectins such as BMS-962476 (BMS/Adnexus, Waltham, MA, USA) are in preclinical development or phase I [3941]. On the basis of the discovery of a LOF mutation in the autocatalytic cleavage site of PCSK9 [47], inhibition of PCSK9 autocatalytic processing is the approach chosen by Cadila Healthcare and Shifa Biomedical [39] with molecules in preclinical development phase. Finally, mAbs [48] are the most studied and advanced approach in terms of clinical development with published phase I, II, and III human trials. An alternative approach for PCSK9 inhibition could be a peptide-based anti-PCSK9 active vaccination approach providing the opportunity for long-term LDL-C management [49].


Efficacy of PCSK9 Inhibition with mAbs


Several mAbs targeting PCSK9 have been tested in preclinical studies to assess their disruption of the PCSK9-LDLR interaction or inhibition of PCSK9 internalization [39, 43]. Human data are mainly available for three mAbs: Alirocumab (SAR236553/REGN727) and evolocumab (AMG 145), two fully human mAbs developed by Sanofi/Regeneron and Amgen, respectively, and bococizumab (RN316/PF04950615), a humanized mAb developed by Pfizer/Rinat. The data obtained in phase II have been already extensively described [11, 42, 43]: globally, in combination with a statin – and also in monotherapy – mAbs induced dramatic significant decreases in all the atherogenic lipoproteins.

Three large phase III programs have been developed with alirocumab, evolocumab, and bococizumab. The lipid-lowering phase III trials [5073] with these three mABs are summarized in Tables 9.1, 9.2, and 9.3. Current data indicate that mAbs are very effective at lowering concentrations of atherogenic lipoproteins, with significant decreases in LDL-C, apoB, non-HDL-C, and also Lp(a) concentrations. So far, in the phase III programs, efficacy has been demonstrated


Table 9.1
Lipid-lowering phase III trials with Alirocumab (ODYSSEY Programme)
























































































Study acronym (NCT number)

Study population (number of subjects)

Duration; dosage; comparator

LDL-C reductiona (% change vs placebo1 or from BL2)

Reference/status

ODYSSEY FH I (NCT 01623115)

Heterozygous FH not controlled on maximally tolerated statin ± other LLT (n = 486)

78-weeks DB; 75/150 mg Q2W; placebo

−57.91

Presented at the ESC congress 2014 [50]

ODYSSEY FH II (NCT 01709500)

Heterozygous FH not controlled on maximally tolerated statin ± other LLT (n = 249)

78-weeks DB; 75/150 mg Q2W; placebo

−51.41

Presented at the ESC congress 2014 [51]

ODYSSEY HIGH FH (NCT 01617655)

Heterozygous FH on maximally tolerated statin ± other LLT and LDL-C >160 mg/dL (n = 105)

78-weeks DB; 150 mg Q2W; placebo

−39.11

Presented at the AHA congress 2014 [52]

ODYSSEY LONG TERM (NCT 01507831)

Heterozygous FH or high CV risk patients on maximally tolerated statin ± other LLT and LDL-C ≥70 mg/dL (n = 2341)

78-weeks DB; 150 mg Q2W; placebo

−61.91

Robinson et al. [53]

ODYSSEY COMBO I (NCT 01644175)

High CV risk patients not controlled on maximally tolerated statin ± other LLT (n = 316)

52-weeks DB; 75/150 mg Q2W; placebo

−45.91

Presented at the AHA congress 2014 [54]

ODYSSEY COMBO II (NCT 01644188)

High CV risk patients not controlled on maximally tolerated statin (n = 720)

104-weeks DB; 75/150 mg Q2W; ezetimibe

−50.62

Cannon et al. [55]

ODYSSEY MONO (NCT 01644174)

Hypercholesterolemic patients with moderate CV risk, not receiving any LLT (n = 103)

24-weeks DB; 75/150 mg Q2W; ezetimibe

−47.22

Roth et al. [56]

ODYSSEY ALTERNATIVE (NCT 01709513)

Statin intolerant patients (with statin rechallenge arm) (n = 314)

24-weeks DB; 75/150 mg Q2W; ezetimibe

−45.02

Presented at the AHA congress 2014 [57]

ODYSSEY OPTIONS I (NCT 01730040)

Patients not controlled on ATV 20 or 40 mg (n = 355)

24-weeks DB; 75/150 mg Q2W; addition of ezetimibe or doubling ATV dose or switching from ATV 40 mg to RSV 40 mg

−44.12 (entry ATV 20 mg) −54.02 (entry ATV 40 mg)

Presented at the AHA congress 2014 [58]

ODYSSEY OPTIONS II (NCT 01730053)

Patients not controlled on RSV 10 or 20 mg (n = 305)

24-weeks DB; 75/150 mg Q2W; addition of ezetimibe or doubling RSV dose

−50.62 (entry RSV 10 mg) −36.32 (entry RSV 20 mg)

Presented at the AHA congress 2014 [59]

ODYSSEY CHOICE I (NCT 01926782)

Patients (1) not controlled on maximally tolerated statin, (2) with moderate CV risk not receiving statin, or (3) with statin intolerance (n = 803)

48-weeks DB; 300 mg Q4W (or 150 mg Q2W after week 12); placebo

−52.41 (no statin group) −58.71 (statin group)

Presented at the ACC congress 2015 [60]

ODYSSEY CHOICE II (NCT 02023879)

Patients not receiving statin, but ezetimibe, fenofibrate or diet alone, with (1) statin intolerance or (2) moderate CV risk (n = 233)

24-weeks DB; 150 mg Q4W (or 150 mg Q2W after week 12); placebo

−56.41

Presented at the ACC congress 2015 [61]


aResults at week 24 (primary efficacy endpoint)

ATV atorvastatin, BL baseline, CV cardiovascular, DB double-blind, FH familial hypercholesterolemia, LLT lipid lowering therapy, Q2W once every 2 weeks, Q4W once every 4 weeks, RSV rosuvastatin



Table 9.2
Lipid-lowering phase III trials with Evolocumab (PROFICIO Programme)


























































Study acronym (NCT number)

Study population (number of subjects)

Duration; dosage; comparator

LDL-C reductiona (% change vs placebo1 or from BL2)

Reference/status

RUTHERFORD-2 (NCT 01763918)

Heterozygous FH not controlled (LDL-C ≥100 mg/dL) on statin ± other LLTb (n = 331)

12-weeks DB; 140 mg Q2W or 420 mg Q4W; placebo

−59.21 (Q2W)

−61.31 (Q4W)

Raal et al. [62]

DESCARTES (NCT 01516879)

Hypercholesterolemic patients on LLT with diet alone or diet plus ATV 10 mg, ATV 80 mg or ATV 80 mg plus EZE 10 mg, and LDL-C ≥75 mg/dL (n = 905)

52-weeks DB; 420 mg Q4W; placebo

−55.71 (diet alone)

−61.61 (entry ATV 10 mg)

−56.81 (entry ATV 80 mg)

−48.51 (entry ATV 80 mg + EZE 10 mg)

Blom et al. [63]

MENDEL −2 (NCT 01763827)

Patients with LDL-C ≥100 and <190 mg/dL and Framingham risk score ≤10% (n = 614)

12-weeks DB; 140 mg Q2W or 420 mg Q4W; placebo or ezetimibe

−56.51 (Q2W)

−57.41 (Q4W)

Koren et al. [64]

GAUSS-2 (NCT 01763905)

Statin intolerant patients (n = 307)

12-weeks DB; 140 mg Q2W or 420 mg Q4W; ezetimibe

−56.12 (Q2W)

−52.62 (Q4W)

Stroes et al. [65]

LAPLACE-2 (NCT 01763866)

Hypercholesterolemic patients on statin therapy (moderate- or high-intensity) (n = 1899)

12-weeks DB; 140 mg Q2W or 420 mg Q4W; placebo or ezetimibe

−76.31 (Q2W)

−70.51 (Q4W) (entry ATV 80 mg)

−68.31 (Q2W)

−55.01 (Q4W) (entry RSV 40 mg)

−71.41 (Q2W)

−59.21 (Q4W) (entry ATV 10 mg)

−70.61 (Q2W)

−60.41 (Q4W) (entry SIM 40 mg)

−68.21 (Q2W)

−64.51 (Q4W) (entry RSV 5 mg)

Robinson et al. [66]

TESLA (Part B) (NCT 01588496)

Homozygous FH on LLT and not receiving LDL-apheresis (n = 50)

12-weeks DB; 420 mg Q4W; placebo

−30.91

Raal et al. [67]

TAUSSIG (NCT 01624142)

Homozygous and heterozygous FH (n = 310)

Open-label trial; 140 mg Q2W or 420 mg Q4W

NA

Ongoing [68]


aResults at week 12 (primary efficacy endpoint), excepted for DESCARTES (week 52)

bexclusion of fibrates

ATV atorvastatin, BL baseline, DB double-blind, EZE ezetimibe, FH familial hypercholesterolemia, LLT lipid lowering therapy, NA not available, Q2W once every 2 weeks, Q4W once every 4 weeks, RSV rosuvastatin, SIM simvastatin



Table 9.3
Lipid-lowering phase III trials with Bococizumab (SPIRE Programme)







































Study acronym (NCT number)

Study population (number of subjects)

Duration; dosage; comparator

Reference/status

SPIRE-FH (NCT 01968980)

Heterozygous FH receiving highly effective statins (n = 300)a

52-weeks DB; 150 mg Q2W; placebo

Ongoing [69]

SPIRE-HR (NCT 01968954)

Hypercholesterolemic patients receiving highly effective statins (n = 600)a

52-weeks DB; 150 mg Q2W; placebo

Ongoing [70]

SPIRE-LDL (NCT 01968967)

Hypercholesterolemic patients receiving highly effective statins (n = 1932)a

52-weeks DB; 150 mg Q2W; placebo

Ongoing [71]

SPIRE-LL (NCT 02100514)

Hyperlipidemic patients receiving background statin therapy (n = 690)a

52-weeks DB; 150 mg Q2W; placebo

Ongoing [72]

SPIRE-SI (NCT 02135029)

Statin intolerance (n = 150)a

24-weeks DB; 150 mg Q2W; placebo or atorvastatin

Ongoing [73]


aestimated number

DB double-blind, FH familial hypercholesterolemia




  • In patients with heterozygous FH [5052, 62] and with homozygous FH [67, 74]


  • In high-risk patients not controlled by maximally tolerated statin and other lipid-lowering therapies [5355]


  • In combination with statins in patients with high LDL cholesterol [58, 59, 63, 66]


  • In patients as monotherapy [56, 64]


  • In patients who could not tolerate statins due to muscle-related side effects [57, 65]

These mAbs are administered as subcutaneous (SC) injections, with various doses and strategies: in the ODYSSEY program, alirocumab SC injections are mainly realized every 2 weeks (Q2W). Two doses, 75 and 150 mg, have been tested in the majority of phase III trials, with the possibility to uptitrate alirocumab from 75 to 150 mg Q2W depending on LDL-C goal achievement. The frequency of injections every 4 weeks (Q4W) and the dose of 300 mg Q4W have been recently evaluated in CHOICE trials [60, 61]. In the PROFICIO program, two doses of evolocumab have been tested, 140 mg Q2W and 420 mg Q4W [6268]. Finally, the SPIRE program is conducted with bococizumab 150 mg Q2W.

Globally, in combination with a statin, mAbs decrease LDL-C levels by 39–61 % in heterozygous FH and by 46–76 % in other hypercholesterolemic patients.

In the TESLA trial, the mean decrease in LDL-C is – as expected – less in patients with homozygous FH [67], with a greater response on evolocumab (−40.8 % in LDL-C) in patients defective in one or both alleles, providing a new complementary therapeutic strategy to treat these very high patients.

Moreover, treatment with mAbs induces a significant decrease in Lp(a) levels. A pooled analysis from 1359 patients in 4 phase II trials showed a dose-dependent reduction of Lp(a) levels with evolocumab (−29.5 and −24.5 % with 140 mg Q2W and 420 mg Q4W respectively) [75]. Complementary studies are needed to characterize the mechanism underlying this effect and to determine the clinical relevance of the Lp(a) reducing effect. However, especially considering that current therapies effective in reducing Lp(a) are limited to mipomersen (an anti-apoB antisense oligonucleotide) or lomitapide (an MTP inhibitor) [76], PCSK9 inhibitors might be an effective option to improve the CVD risk of patients with elevated Lp(a) plasma levels.


Safety and Tolerability of PCSK9 Inhibition with mAbs


Overall, the mAbs tested so far have been generally safe and well tolerated, with no major safety issues and no differences in the rate of adverse events (AEs) between treatment and placebo groups from completed phase II and III studies.

In all of the phase 2 studies, alirocumab was generally well tolerated over the treatment period [8–12 weeks]. Injection-site reactions were the most common AEs in two of the phase II trials but were generally mild in severity and transient. However, in the phase II study assessing alirocumab for treatment of FH [77], one patient in the group of 300 mg dose Q4W discontinued treatment after the first dose due to injection-site reaction and generalized pruritus. In another phase II trial [78], one patient receiving atorvastatin 80 mg plus alirocumab 150 mg Q2W discontinued treatment due to a hypersensitivity reaction and rash occurring 12 days after the second injection of mAb. There was a single case of cutaneous leukocytoclastic vasculitis reported in one patient, 9 days after initiation of alirocumab 300 mg [79]. The patient responded rapidly to withdrawal of the drug and initiation of steroid therapy.

Evolocumab was also generally well tolerated throughout the phase II trials, with a similar incidence of drug-related AEs across treatment groups and no evidence of a relationship between the incidence of any AEs and evolocumab dose [42]. Small numbers of serious adverse events (SAEs) occurred, but none was considered related to the treatment. Injection-site reactions were generally infrequent and mild. Of the 1359 randomized patients in the evolocumab phase II parent studies, 1104 (81 %) elected to enroll in the OSLER study, open trial designed to evaluate mainly longer-term safety. Patients were randomized 2:1 to receive either open-label SC evolocumab 420 mg Q4W with standard of care or standard of care alone [80]. In the OSLER trial, AE occurred in 81.4 % of evolocumab-treated patients and in 73.1 % of patients in the standard of care group. SAE occurred in 7.1 % of patients in the evolocumab group and 6.3 % in the standard of care group. An injection-site reaction was reported in 5.2 % of patients in the evolocumab group. In a specific analysis of the frequency of AEs by LDL-C value on evolocumab treatment, an imbalance appeared in memory impairment in patients with LDL-C <50 mg/dL: four patients (1 %) compared to one patient (0.3 %) in evolocumab subgroup with LDL-C ≥50 mg/dL and zero in the standard of care group. This finding seems at the origin of the request from FDA to make an assessment of potential neurocognitive AEs across the phase III development program for all the mAbs against PCSK9, especially in the longer-term studies.

Indeed, more information on safety and tolerability has been obtained from phase III programs of alirocumab [5061] and evolocumab [6267], especially from the longer-term ODYSSEY LONG TERM [53] and DESCARTES [63] trials. In the DESCARTES trial conducted in 901 patients (599 on evolocumab 420 mg Q4W, 302 on placebo) during 52 weeks, the overall incidence of AEs was similar in the evolocumab group and the placebo group. The most common AEs in the evolocumab group were nasopharyngitis, upper respiratory tract infection, influenza, and back pain. Injection-site reactions were reported in 5.7 % of patients in the evolocumab group and 5.0 % of patients in the placebo group [63].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 1, 2017 | Posted by in CARDIOLOGY | Comments Off on Statins and PCSK9 Inhibitors: Defining the Correct Patients

Full access? Get Clinical Tree

Get Clinical Tree app for offline access