Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 1.
Published in final edited form as: Circ Cardiovasc Imaging. 2017 Jul;10(7):e006676. doi: 10.1161/CIRCIMAGING.117.006676

Lipid-lowering and Anti-inflammatory Benefits of Statin Therapy: More Than Meets the Plaque

Viviany R Taqueti *, Paul M Ridker +
PMCID: PMC5757307  NIHMSID: NIHMS883568  PMID: 28679526

The benefits of statin therapy have proven so effective that patients with atherosclerosis are counseled to remain on treatment indefinitely.1 Because the precise mechanisms by which statins exert a survival benefit are incompletely explained by their effect on serum lipids,2 intense efforts have focused on inflammatory effects, both systemically and locally at the plaque itself.

In this issue of Circulation: Cardiovascular Imaging, Kwon and Kang et al.3 report a post-hoc analysis of the prospective, single-center STABLE (Statin and Atheroma Vulnerability Evaluation) trial,4 in which patients in Seoul, Korea underwent invasive coronary angiography with intravascular ultrasound (IVUS) at baseline and 12 months following 1:2 randomization to rosuvastatin 10 mg vs. 40 mg daily. Only those patients (n=312) with coronary stenoses defined by virtual histology (VH)-IVUS as fibroatheroma-containing lesions were enrolled, and 225 patients completed the protocol. The primary report of the STABLE trial concluded that patients on either dose of rosuvastatin demonstrated lesions with reduced frequencies of thin-cap fibroatheroma (TCFA), decreased percent necrotic core volume, and increased percent fibrofatty volume on follow-up evaluation.4

In the current analysis, investigators explored the relationship between the changes observed in VH-IVUS-defined characteristics of index plaque with two critical biomarkers, low-density lipoprotein cholesterol (LDLC) and high-sensitivity C-reactive protein (hsCRP). As would be anticipated in a statin trial, levels of LDLC and hsCRP decreased robustly (105.7 to 67.1 mg/dl and 2.2 to 1.2 mg/l, respectively) from baseline to follow-up. In analyses stratified by quartiles of change in hsCRP and LDLC, linear associations with changes in plaque characteristics (percent necrotic core and dense calcium volumes, as well as percent fibrous and fibrofatty volumes) were found to be statistically significant with hsCRP, but less so with LDL. In multivariable logistic regression, existing diabetes and TCFA at baseline, rather than baseline or change in serum biomarkers, demonstrated the strongest associations with the presence of TCFA at follow-up (adjusted OR 3.17, 95% CI 1.62–9.97 for diabetes and adjusted OR 8.82, 95% CI 3.04–27.92 for TCFA at baseline). The authors report that a greater decrease in hsCRP (but not LDLC) was observed in those without, as compared to those with TCFA at follow-up. They cite these data as supportive of the anti-inflammatory effects of statin, specifically on plaque stabilization.

What is new? Using serial IVUS, Nissen et al.5 previously reported in the multicenter REVERSAL trial (n=502) that patients randomized to intensive atorvastatin (80 mg daily) versus moderate pravastatin (40 mg daily) treatment over 18 months demonstrated reduced progression of coronary atherosclerosis by change in percent atheroma volume. Patients with slower rates of plaque progression demonstrated greater reductions in levels of both LDLC and CRP, even though the correlation between the reduction in LDLC and CRP levels was modest in the cohort as a whole.6 Additional investigations710 using serial IVUS, including the SATURN trial of patients receiving only high-intensity statin therapy,10 reported regression of plaque atheroma volume, but results were relatively modest.

As intracoronary imaging evolved, efforts expanded from simply measuring plaque volume using gray-scale IVUS to focusing on candidate markers of plaque stabilization. With the advent of radiofrequency or VH-IVUS, investigators began to correlate certain plaque characteristics at baseline with downstream response to therapy11, 12 and future cardiovascular events.13 In the PROSPECT trial, the presence of a TCFA was independently associated with the occurrence of a major adverse cardiovascular event at the site of a previously defined angiographically mild, nonculprit lesion (HR 3.35, 95% CI 1.77–6.36)13. The current study adds to the existing literature an analysis of serial VH-IVUS-defined plaque characteristics and serum lipid and inflammatory biomarkers in a cohort of Asian patients on a contemporary statin for secondary prevention.

Care should be taken when making direct comparisons between STABLE and earlier trials such as REVERSAL, which utilized gray-scale IVUS and demonstrated that changes in both hsCRP and LDLC correlate with change in plaque volume. STABLE was a single center experience with smaller sample size and shorter study duration. It is possible that the STABLE trial, despite use of VH-IVUS, was underpowered to detect consistent associations between differences in serum biomarkers and plaque characteristics Relatively low power may also explain why little correlation was observed between serum biomarkers and percent atheroma volume using traditional gray-scale IVUS, and why little difference was observed between high- and low-dose rosuvastatin.

Despite limitations, the current study by Kwon and Kang et al. is important because it ultimately makes the point that the dramatic LDLC and hsCRP lowering seen on average in patients on a later-generation statin (with proven survival benefit in large outcomes trials2, 14, 15), is not fully captured using either gray-scale or VH-IVUS evaluation of any given plaque. Indeed, the strongest marker associated with presence of TCFA post-statin in this study was the presence of TCFA at baseline. Even TCFA, which as defined by VH-IVUS has gained popularity as a surrogate for unstable plaque, lacks specificity; of 595 TCFA identified in PROSPECT, only 26 were actually associated with sites of future events over 3 years of follow-up.13 We must thus acknowledge that the quest for focal endpoint evidence (i.e. at the level of individual plaque anatomy) for what is almost certainly a systemic biological effect (e.g. the lipid-lowering and anti-inflammatory effects of statins) may never be fully realized.

Recent work has called attention to the fact that patients with “residual inflammatory risk” are biologically distinct from those with “residual cholesterol risk.”16 In this regard, the current data also underscore the need to separate lipid-lowering from anti-inflammatory effects of atherosclerotic therapies, as is now underway in cardiovascular outcomes trials of anti-inflammatory therapy for secondary prevention.1719 Embedded within these trials are innovative imaging substudies utilizing FDG-PET assessment of plaque inflammation to correlate with changes in plaque morphology20 at the local level, as well as cardiac PET quantification of coronary flow reserve to correlate global vasomotor and microvascular function with cardiac structure21 and serum biomarkers22 systemically. When complete, these concomitant imaging and outcome data may help to more precisely phenotype patients with residual risk and to determine best approaches for the care of our patients.

Footnotes

Disclosures: None.

References

  • 1.Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM, McBride P, Schwartz JS, Shero ST, Smith SC, Jr, Watson K, Wilson PW, Eddleman KM, Jarrett NM, LaBresh K, Nevo L, Wnek J, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Curtis LH, DeMets D, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Smith SC, Jr, Tomaselli GF. American College of Cardiology/American Heart Association Task Force on Practice G. 2013 acc/aha guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: A report of the american college of cardiology/american heart association task force on practice guidelines. Circulation. 2014;129:S1–45. doi: 10.1161/01.cir.0000437738.63853.7a. [DOI] [PubMed] [Google Scholar]
  • 2.Ridker PM, Cannon CP, Morrow D, Rifai N, Rose LM, McCabe CH, Pfeffer MA, Braunwald E. Pravastatin or Atorvastatin E, Infection Therapy-Thrombolysis in Myocardial Infarction I. C-reactive protein levels and outcomes after statin therapy. The New England journal of medicine. 2005;352:20–28. doi: 10.1056/NEJMoa042378. [DOI] [PubMed] [Google Scholar]
  • 3.Kwon O, Kang SJ, Kang SH, Lee PH, Yun SC, Ahn JM, Park DW, Lee SW, Kim YH, Lee CW, Han KH, Park SW, Park SJ. Relationship between serum inflammatory marker levels and the dynamic changes in coronary plaque characteristics following statin therapy. Circulation. Cardiovascular imaging. 2017 doi: 10.1161/CIRCIMAGING.116.005934. XX:XXX. [DOI] [PubMed] [Google Scholar]
  • 4.Park SJ, Kang SJ, Ahn JM, Chang M, Yun SC, Roh JH, Lee PH, Park HW, Yoon SH, Park DW, Lee SW, Kim YH, Lee CW, Mintz GS, Han KH, Park SW. Effect of statin treatment on modifying plaque composition: A double-blind, randomized study. Journal of the American College of Cardiology. 2016;67:1772–1783. doi: 10.1016/j.jacc.2016.02.014. [DOI] [PubMed] [Google Scholar]
  • 5.Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN Investigators R. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: A randomized controlled trial. Jama. 2004;291:1071–1080. doi: 10.1001/jama.291.9.1071. [DOI] [PubMed] [Google Scholar]
  • 6.Nissen SE, Tuzcu EM, Schoenhagen P, Crowe T, Sasiela WJ, Tsai J, Orazem J, Magorien RD, O'Shaughnessy C, Ganz P. Reversal of Atherosclerosis with Aggressive Lipid Lowering I. Statin therapy, ldl cholesterol, c-reactive protein, and coronary artery disease. The New England journal of medicine. 2005;352:29–38. doi: 10.1056/NEJMoa042000. [DOI] [PubMed] [Google Scholar]
  • 7.Jensen LO, Thayssen P, Pedersen KE, Stender S, Haghfelt T. Regression of coronary atherosclerosis by simvastatin: A serial intravascular ultrasound study. Circulation. 2004;110:265–270. doi: 10.1161/01.CIR.0000135215.75876.41. [DOI] [PubMed] [Google Scholar]
  • 8.Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif JC, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM Investigators A. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: The asteroid trial. Jama. 2006;295:1556–1565. doi: 10.1001/jama.295.13.jpc60002. [DOI] [PubMed] [Google Scholar]
  • 9.Okazaki S, Yokoyama T, Miyauchi K, Shimada K, Kurata T, Sato H, Daida H. Early statin treatment in patients with acute coronary syndrome: Demonstration of the beneficial effect on atherosclerotic lesions by serial volumetric intravascular ultrasound analysis during half a year after coronary event: The establish study. Circulation. 2004;110:1061–1068. doi: 10.1161/01.CIR.0000140261.58966.A4. [DOI] [PubMed] [Google Scholar]
  • 10.Nicholls SJ, Ballantyne CM, Barter PJ, Chapman MJ, Erbel RM, Libby P, Raichlen JS, Uno K, Borgman M, Wolski K, Nissen SE. Effect of two intensive statin regimens on progression of coronary disease. The New England journal of medicine. 2011;365:2078–2087. doi: 10.1056/NEJMoa1110874. [DOI] [PubMed] [Google Scholar]
  • 11.Hong MK, Park DW, Lee CW, Lee SW, Kim YH, Kang DH, Song JK, Kim JJ, Park SW, Park SJ. Effects of statin treatments on coronary plaques assessed by volumetric virtual histology intravascular ultrasound analysis. JACC. Cardiovascular interventions. 2009;2:679–688. doi: 10.1016/j.jcin.2009.03.015. [DOI] [PubMed] [Google Scholar]
  • 12.Nasu K, Tsuchikane E, Katoh O, Tanaka N, Kimura M, Ehara M, Kinoshita Y, Matsubara T, Matsuo H, Asakura K, Asakura Y, Terashima M, Takayama T, Honye J, Hirayama A, Saito S, Suzuki T. Effect of fluvastatin on progression of coronary atherosclerotic plaque evaluated by virtual histology intravascular ultrasound. JACC. Cardiovascular interventions. 2009;2:689–696. doi: 10.1016/j.jcin.2009.04.016. [DOI] [PubMed] [Google Scholar]
  • 13.Stone GW, Maehara A, Lansky AJ, de Bruyne B, Cristea E, Mintz GS, Mehran R, McPherson J, Farhat N, Marso SP, Parise H, Templin B, White R, Zhang Z, Serruys PW Investigators P. A prospective natural-history study of coronary atherosclerosis. The New England journal of medicine. 2011;364:226–235. doi: 10.1056/NEJMoa1002358. [DOI] [PubMed] [Google Scholar]
  • 14.Bohula EA, Giugliano RP, Cannon CP, Zhou J, Murphy SA, White JA, Tershakovec AM, Blazing MA, Braunwald E. Achievement of dual low-density lipoprotein cholesterol and high-sensitivity c-reactive protein targets more frequent with the addition of ezetimibe to simvastatin and associated with better outcomes in improve-it. Circulation. 2015;132:1224–1233. doi: 10.1161/CIRCULATIONAHA.115.018381. [DOI] [PubMed] [Google Scholar]
  • 15.Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ, Group JS. Rosuvastatin to prevent vascular events in men and women with elevated c-reactive protein. The New England journal of medicine. 2008;359:2195–2207. doi: 10.1056/NEJMoa0807646. [DOI] [PubMed] [Google Scholar]
  • 16.Ridker PM. Residual inflammatory risk: Addressing the obverse side of the atherosclerosis prevention coin. European heart journal. 2016;37:1720–1722. doi: 10.1093/eurheartj/ehw024. [DOI] [PubMed] [Google Scholar]
  • 17.Ridker PM. Testing the inflammatory hypothesis of atherothrombosis: Scientific rationale for the cardiovascular inflammation reduction trial (cirt) Journal of thrombosis and haemostasis : JTH. 2009;(7 Suppl 1):332–339. doi: 10.1111/j.1538-7836.2009.03404.x. [DOI] [PubMed] [Google Scholar]
  • 18.Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1beta inhibition and the prevention of recurrent cardiovascular events: Rationale and design of the canakinumab anti-inflammatory thrombosis outcomes study (cantos) American heart journal. 2011;162:597–605. doi: 10.1016/j.ahj.2011.06.012. [DOI] [PubMed] [Google Scholar]
  • 19.Ridker PM. From c-reactive protein to interleukin-6 to interleukin-1: Moving upstream to identify novel targets for atheroprotection. Circulation research. 2016;118:145–156. doi: 10.1161/CIRCRESAHA.115.306656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Taqueti VR, Di Carli MF, Jerosch-Herold M, Sukhova GK, Murthy VL, Folco EJ, Kwong RY, Ozaki CK, Belkin M, Nahrendorf M, Weissleder R, Libby P. Increased microvascularization and vessel permeability associate with active inflammation in human atheromata. Circulation. Cardiovascular imaging. 2014;7:920–929. doi: 10.1161/CIRCIMAGING.114.002113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Taqueti VR, Shaw LJ, Cook NR, Murthy VL, Shah NR, Foster CR, Hainer J, Blankstein R, Dorbala S, Di Carli MF. Excess cardiovascular risk in women relative to men referred for coronary angiography is associated with severely impaired coronary flow reserve, not obstructive disease. Circulation. 2017;135:566–577. doi: 10.1161/CIRCULATIONAHA.116.023266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Taqueti VR, Everett BM, Murthy VL, Gaber M, Foster CR, Hainer J, Blankstein R, Dorbala S, Di Carli MF. Interaction of impaired coronary flow reserve and cardiomyocyte injury on adverse cardiovascular outcomes in patients without overt coronary artery disease. Circulation. 2015;131:528–535. doi: 10.1161/CIRCULATIONAHA.114.009716. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES