Skip to main content
Archives of Medical Science : AMS logoLink to Archives of Medical Science : AMS
. 2020 Apr 14;16(3):485–489. doi: 10.5114/aoms.2020.94503

Lipid-lowering therapy and renin-angiotensin-aldosterone system inhibitors in the era of the COVID-19 pandemic

Niki Katsiki 1, Maciej Banach 2,3,, Dimitri P Mikhailidis 4,
PMCID: PMC7212217  PMID: 32399093

The novel coronavirus (severe acute respiratory syndrome coronavirus 2 – SARS-Cov-2) disease 2019 (COVID-19) pandemic has been associated with severe respiratory disease incidence and increased mortality [1]. Angiotensin converting enzyme (ACE) 2 is a homologue of ACE, but also a receptor for the coronaviruses [2]. ACE2 is highly expressed in the lungs, heart, gastrointestinal (GI) tract and kidney, thus affecting the cardiovascular system (CV) and the immune system [3]. The overexpression of ACE2 was reported to enhance viral entry and replication intracellularly [4]. COVID-19, also called SARS-CoV-2, may also use ACE2 as a receptor to initiate infection, leading to severe complications from the heart (acute coronary syndrome (ACS) and fulminant myocarditis), lungs (pneumonia and acute respiratory distress syndrome (ARDS)) and GI tract (diarrhoea syndrome) [5].

ACE2 gene expression is affected by several factors, including gender (ACE2 gene is X-linked), ACE2 gene polymorphisms, comorbidities (increased in the presence of CVD, hypertension, diabetes), and drug therapy [6]. With regard to drugs, angiotensin II receptor blockers (ARBs) and mineralocorticoid receptor antagonists (MRA) have been reported to raise ACE2 activity in human and animal studies [7]. There are only a few animal studies available showing that statins may also increase ACE2 activity [8, 9]. In the era of the COVID-19 pandemic, such a drug effect may be considered as potentially worrying [10]. In this context, it was recently even suggested that ARBs could be replaced with ACE inhibitors and that statin treatment may be discontinued during the pandemic, particularly in primary prevention settings [11].

However, before implementing such strategies, we should consider several issues. Firstly, as the COVID-19 infection progresses, ACE2 is downregulated, thus potentially generating an inflammatory response leading to impaired cardiac contractility and acute lung injury [5, 7, 12]. Therefore, reduced ACE2 expression is linked to worse outcomes. On the other hand, ACE2 overexpression has been associated with several beneficial effects, i.e. prevention of adverse cardiac remodelling and fibrosis, improvement of vascular endothelial dysfunction, reduction of blood pressure, and protection from ARDS [7, 12]. Both statins and ARBs were reported to exert these benefits.

Secondly, a combination of statins/ARBs were used during the 2014 Ebola virus disease epidemic in Sierra Leone, leading to improved outcomes and increased survival [13]. These drugs can affect the host response to infection, not the virus, especially by preventing endothelial dysfunction, a shared feature of several virus infections [14]. Their combination seemed to promote a return to homeostasis, allowing patients with Ebola virus infection to recover on their own [15].

Third, patients with cardiovascular disease (CVD) were shown to be more prone to COVID-19 infection and with worse prognosis [16, 17]. Elevated inflammatory markers, such as C-reactive protein (CRP) and interleukin-6 (IL-6), have been recognised as predictors of COVID-19 infection severity and mortality, suggesting a virus-activated “cytokine storm syndrome” [18, 19]. Therefore, as well as immunomodulation, COVID-19 treatment should also target reduction of inflammation. In this context, statins have been consistently reported to exert immunomodulatory and anti-inflammatory properties [2030]. Also, it was previously suggested that statins could enhance host defence and suppress inflammation, thus representing a practical and inexpensive adjunctive or alternative host-directed treatment for infections by viruses, fungi, protozoa, and bacteria [31]. Similarly, there are data supporting an anti-inflammatory role for ARBs [3234].

Fourth, statins may also prevent a viral-induced acute coronary syndrome (also in COVID-19 positive patients) by stabilising atherosclerotic plaques [35], as well as prevent acute kidney injury (AKI) [36]. Both acute cardiac injury and AKI are predictors of COVID-19-induced mortality [37]; statin therapy may prevent these complications and thus increase survival. Of note, statins can protect against contrast-induced AKI (CI-AKI) [3841]. This is of clinical importance, especially in hospitalised patients who undergo diagnostic or therapeutic procedures involving the administration of contrast media (e.g. computed tomography of the lungs).

Fifth, effective lipid-lowering therapy (LLT) and significant cholesterol reduction might significantly suppress coronavirus infection. It was show that for infectious bronchitis virus (IBV) coronavirus, drug-related cholesterol reduction disrupts lipid rafts (an important element for the cellular entry of coronavirus) that enable the binding of the coronavirus with the host cells and, consequently, further infection [42]. It was also observed, in the studies with porcine deltacoronavirus (PDCoV), that cholesterol present in the cell membrane and viral envelope (coronaviruses are positive-sense enveloped RNA viruses) contributes to PDCoV replication by acting as a key component in viral entry. Thus, the pharmacological sequestration of cellular or viral cholesterol with effective LLT significantly blocked both virus attachment and internalisation [43]. All these mechanisms might suggest a critical role of statins and LLTs in the inhibition of coronavirus infection.

In COVID-19-positive patients, the majority of baseline CVD is of atherosclerosis origin, with the worst prognosis for patients being at the high, and especially very high and extremely high, risk of CVD [16]; thus, intensive LLT with statins and/or fixed combination with ezetimibe and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors seems to be critical. Indeed, we should do our best to maximally improve therapy adherence and thus have a better prognosis for the infected CVD patients [44, 45]. In this context, there are no premises that PCSK9 inhibitors, because they are monoclonal antibodies (in relation to the above-mentioned high cytokine storm during infection), should be discontinued. In contrast, PCSK9 inhibitors should be continued to achieve further low-density lipoprotein cholesterol (LDL-C) lowering (based on “the lower, the better” principle), because then we might significantly stabilise atheroma plaque, reduce the risk of CVD events, and reduce inflammation [4648]. Recent available data have confirmed the role of PCSK9 inhibition in reducing the process of inflammation via decreasing main vascular inflammatory markers, reducing infiltration of monocytes into the subendothelial layer, and inhibiting monocyte migration. Apart from the reduction of pro-inflammatory mediators, PCSK9 inhibitors could ameliorate vascular inflammation [47]. Finally, a direct local anti-inflammatory action of PCSK9 inhibitors, independent of LDL-C reduction, has been shown in animal models; however, it still merits further investigation [47, 48].

It is of special interest now (due to the fact that coronavirus might also use different receptors to enter the host cell) that treatment with PCSK9 inhibitors has beneficial effects on LDL-C lowering via inhibition of LDL-receptors (LDL-R). This might exert an antiviral effect, among others, on hepatitis C viral (HCV) infection through down-regulation of the surface expression of LDL-R and cluster of differentiation (CD) 81 on hepatic cells, and a positive association with increased inflammatory responses, as well as with septic shock [48]. In a recent paper, we confirmed that there is no association between PCSK9 levels and resistance to antibiotics or the condition of patients hospitalised in intensive care units, a finding of clinical importance in the COVID-19 infection era [49].

Sixth, there are conflicting results regarding the possible effects of statins on ARDS development and outcomes [50, 51]. It was suggested that statins act beneficially in ‘hyper-inflammatory’ ARDS patients (defined by increased biomarkers of inflammation, coagulation and endothelial activation) [52], but not in ‘hypo-inflammatory’ patients [53, 54]. A potential benefit of ARBs on survival in ARDS patients has also been reported [55, 56]. Nevertheless, there is a paucity of data on this field, and thus further research is needed to elucidate the association between statin therapy, ARBs, and acute lung injury.

Of note, drug-drug interactions should also be considered. In this context, simvastatin and lovastatin are contraindicated in patients on lopinavir/ritonavir therapy due to an increased risk of rhabdomyolysis [57]. Atorvastatin, rosuvastatin and other statins can be used at the lowest possible dose, based on the instructions included in the summary of product characteristics (spc) [58]. Taking this into account, we should be careful while treating COVID-19 disease patients with statins being on antiviral drugs and some antibiotics (including macrolides), because they might increase the risk of statin-associated muscle symptoms (SAMS) [59, 60]. Therefore, their careful monitoring is highly recommended to avoid unnecessary drug-related side effects, and at the same time optimising LLT therapy to achieve the individual’s LDL-C goal. In this context, in patients at very high CVD risk, requiring intensive LLT, it is reasonable to initiate therapy with polypills/fixed combinations of statins (at lower doses) and ezetimibe, with or without PCSK9 inhibitors (as available), aimed at reducing the risk of SAMS [59, 60].

A position statement of the European Society (ESC) Council (on 13 March 2020) (as well as of other national and international societies) highlights the lack of evidence on harmful effects of ACE inhibitors and ARBs on the incidence and progression of COVID-19 infection and strongly supports the continuation of usual antihypertensive therapy [6, 61]. Regarding statins, their beneficial effects on inflammation, vascular, heart, and lung function strongly support the continuation of their use. Due to their significant effect on CVD prevention, PCSK9 inhibitors should also be continued, as available. Physicians should wait for strong evidence and recommendations from international scientific societies before altering their patients’ drug therapy in the COVID-19 era.

Acknowledgments

Dr Niki Katsiki and Maciej Banach contributed equally to this paper.

Conflict of interest

NK has given talks, attended conferences, and participated in trials sponsored by Angelini, Astra Zeneca, Bausch Health, Boehringer Ingelheim, Elpen, Mylan, NovoNordisk, Sanofi, and Servier. MB – speakers bureau: Abbott/Mylan, Abbott Vascular, Actavis, Akcea, Amgen, Biofarm, KRKA, MSD, Polpharma, Sanofi-Aventis, Servier and Valeant; consultant to Abbott Vascular, Akcea, Amgen, Daichii Sankyo, Esperion, Freia Pharmaceuticals, Lilly, MSD, Polfarmex, Resverlogix, Sanofi-Aventis; Grants from Sanofi and Valeant. DPM has given talks and attended conferences sponsored by Amgen, Novonordisk, and Libytec.

References

  • 1.McMichael TM, Currie DW, Clark S, et al. Epidemiology of Covid-19 in a long-term care facility in King County, Washington. N Engl J Med. 2020 doi: 10.1056/NEJMoa2005412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lazartigues E, Feng Y, Lavoie JL. The two fACEs of the tissue renin-angiotensin systems: implication in cardiovascular diseases. Curr Pharm Des. 2007;13:1231–45. doi: 10.2174/138161207780618911. [DOI] [PubMed] [Google Scholar]
  • 3.Turner AJ, Hiscox JA, Hooper NM. ACE2: from vasopeptidase to SARS virus receptor. Trends Pharmacol Sci. 2004;25:291–4. doi: 10.1016/j.tips.2004.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Li W, Moore MJ, Vasilieva N, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–4. doi: 10.1038/nature02145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hanff TC, Harhay MO, Brown TS, Cohen JB, Mohareb AM. Is there an association between COVID-19 mortality and the renin-angiotensin system-a call for epidemiologic investigations. Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. Renin-angiotensin-aldosterone system inhibitors in patients with Covid-19. N Engl J Med. 2020 doi: 10.1056/NEJMsr2005760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chamsi-Pasha MA, Shao Z, Tang WH. Angiotensin-converting enzyme 2 as a therapeutic target for heart failure. Curr Heart Fail Rep. 2014;11:58–63. doi: 10.1007/s11897-013-0178-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li YH, Wang QX, Zhou JW, et al. Effects of rosuvastatin on expression of angiotensin-converting enzyme 2 after vascular balloon injury in rats. J Geriatr Cardiol. 2013;10:151–8. doi: 10.3969/j.issn.1671-5411.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Suski M, Gębska A, Olszanecki R, et al. Influence of atorvastatin on angiotensin I metabolism in resting and TNF-alpha-activated rat vascular smooth muscle cells. J Renin Angiotensin Aldosterone Syst. 2014;15:378–83. doi: 10.1177/1470320313475907. [DOI] [PubMed] [Google Scholar]
  • 10.Diaz JH. Hypothesis: angiotensin-converting enzyme inhibitors and angiotensin receptor blockers may increase the risk of severe COVID-19. J Travel Med. 2020 doi: 10.1093/jtm/taaa041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Goldstein MR, Poland GA, Graeber CW. Are certain drugs associated with enhanced mortality in COVID-19? QJM. 2020 doi: 10.1093/qjmed/hcaa103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang H, Baker A. Recombinant human ACE2: acing out angiotensin II in ARDS therapy. Crit Care. 2017;21:305. doi: 10.1186/s13054-017-1882-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Fedson DS. Treating the host response to emerging virus diseases: lessons learned from sepsis, pneumonia, influenza and Ebola. Ann Transl Med. 2016;4:421. doi: 10.21037/atm.2016.11.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Steinberg BE, Goldenberg NM, Lee WL. Do viral infections mimic bacterial sepsis? The role of microvascular permeability: a review of mechanisms and methods. Antiviral Res. 2012;93:2–15. doi: 10.1016/j.antiviral.2011.10.019. [DOI] [PubMed] [Google Scholar]
  • 15.Fedson DS, Opal SM, Rordam OM. Hiding in plain sight: an approach to treating patients with severe COVID-19 infection. mBio. 2020;11(2) doi: 10.1128/mBio.00398-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Guo T, Fan Y, Chen M, et al. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Li B, Yang J, Zhao F, et al. Prevalence and impact of cardiovascular metabolic diseases on COVID-19 in China. Clin Res Cardiol. 2020 doi: 10.1007/s00392-020-01626-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020 doi: 10.1007/s00134-020-05991-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wan S, Xiang Y, Fang W, et al. Clinical features and treatment of COVID-19 patients in Northeast Chongqing. J Med Virol. 2020 doi: 10.1002/jmv.25783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Drapala A, Sikora M, Ufnal M. Statins, the renin-angiotensin-aldosterone system and hypertension – a tale of another beneficial effect of statins. J Renin Angiotensin Aldosterone Syst. 2014;15:250–8. doi: 10.1177/1470320314531058. [DOI] [PubMed] [Google Scholar]
  • 21.Katsiki N, Doumas M, Mikhailidis DP. Lipids, statins and heart failure: an update. Curr Pharm Des. 2016;22:4796–806. doi: 10.2174/1381612822666160701073452. [DOI] [PubMed] [Google Scholar]
  • 22.Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. The role of statins in the treatment of type 2 diabetes mellitus: an update. Curr Pharm Des. 2014;20:3665–74. doi: 10.2174/13816128113196660673. [DOI] [PubMed] [Google Scholar]
  • 23.Katsiki N, Reiner Ž, Tedeschi Reiner E, et al. Improvement of endothelial function by pitavastatin: a meta-analysis. Expert Opin Pharmacother. 2018;19:279–86. doi: 10.1080/14656566.2018.1428560. [DOI] [PubMed] [Google Scholar]
  • 24.Zeiser R. Immune modulatory effects of statins. Immunology. 2018;154:69–75. doi: 10.1111/imm.12902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ridker PM. Clinician’s guide to reducing inflammation to reduce atherothrombotic risk: JACC review topic of the week. J Am Coll Cardiol. 2018;72:3320–31. doi: 10.1016/j.jacc.2018.06.082. [DOI] [PubMed] [Google Scholar]
  • 26.Pirro M, Simental-Mendía LE, Bianconi V, Watts GF, Banach M, Sahebkar A. Effect of statin therapy on arterial wall inflammation based on 18F-FDG PET/CT: a systematic review and meta-analysis of interventional studies. J Clin Med. 2019;8:E118. doi: 10.3390/jcm8010118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bahrami A, Parsamanesh N, Atkin SL, Banach M, Sahebkar A. Effect of statins on toll-like receptors: a new insight to pleiotropic effects. Pharmacol Res. 2018;135:230–8. doi: 10.1016/j.phrs.2018.08.014. [DOI] [PubMed] [Google Scholar]
  • 28.Forouzanfar F, Butler AE, Banach M, Barreto GE, Sahbekar A. Modulation of heat shock proteins by statins. Pharmacol Res. 2018;134:134–44. doi: 10.1016/j.phrs.2018.06.020. [DOI] [PubMed] [Google Scholar]
  • 29.Sahebkar A, Kotani K, Serban C, et al. Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group Statin therapy reduces plasma endothelin-1 concentrations: a meta-analysis of 15 randomized controlled trials. Atherosclerosis. 2015;241:433–42. doi: 10.1016/j.atherosclerosis.2015.05.022. [DOI] [PubMed] [Google Scholar]
  • 30.Bielecka-Dabrowa A, Mikhailidis DP, Rizzo M, von Haehling S, Rysz J, Banach M. The influence of atorvastatin on parameters of inflammation left ventricular function, hospitalizations and mortality in patients with dilated cardiomyopathy: 5-year follow-up. Lipids Health Dis. 2013;12:47. doi: 10.1186/1476-511X-12-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Parihar SP, Guler R, Brombacher F. Statins: a viable candidate for host-directed therapy against infectious diseases. Nat Rev Immunol. 2019;19:104–17. doi: 10.1038/s41577-018-0094-3. [DOI] [PubMed] [Google Scholar]
  • 32.Di Raimondo D, Tuttolomondo A, Buttà C, Miceli S, Licata G, Pinto A. Effects of ACE-inhibitors and angiotensin receptor blockers on inflammation. Curr Pharm Des. 2012;18:4385–413. doi: 10.2174/138161212802481282. [DOI] [PubMed] [Google Scholar]
  • 33.Silva IVG, de Figueiredo RC, Rios DRA. Effect of different classes of antihypertensive drugs on endothelial function and inflammation. Int J Mol Sci. 2019;20 doi: 10.3390/ijms20143458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Del Fiorentino A, Cianchetti S, Celi A, Dell’Omo G, Pedrinelli R. The effect of angiotensin receptor blockers on C-reactive protein and other circulating inflammatory indices in man. Vasc Health Risk Manag. 2009;5:233–42. doi: 10.2147/vhrm.s4800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Madjid M, Safavi-Naeini P, Solomon SD, Vardeny O. Potential effects of coronaviruses on the cardiovascular system: a review. JAMA Cardiol. 2020 doi: 10.1001/jamacardio.2020.1286. [DOI] [PubMed] [Google Scholar]
  • 36.Yoshida T, Hayashi M. Pleiotropic effects of statins on acute kidney injury: involvement of Krüppel-like factor 4. Clin Exp Nephrol. 2017;21:175–81. doi: 10.1007/s10157-016-1286-4. [DOI] [PubMed] [Google Scholar]
  • 37.Deng Y, Liu W, Liu K, et al. Clinical characteristics of fatal and recovered cases of coronavirus disease 2019 (COVID-19) in Wuhan, China: a retrospective study. Chin Med J. 2020 doi: 10.1097/CM9.0000000000000824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Katsiki N, Tsioufis C, Hahalis G, Athyros VG. Contrast-induced acute kidney injury: beware of the risk after coronary angiography. Expert Rev Cardiovasc Ther. 2018;16:73. doi: 10.1080/14779072.2018.1421070. [DOI] [PubMed] [Google Scholar]
  • 39.Katsiki N, Fonseca V, Mikhailidis DP. Contrast-induced acute kidney injury in diabetes mellitus: clinical relevance and predisposing factors. Could statins be of benefit? J Diabetes Complications. 2018;32:982–4. doi: 10.1016/j.jdiacomp.2018.08.008. [DOI] [PubMed] [Google Scholar]
  • 40.Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. Contrast-Induced nephropathy: an “all or none” phenomenon? Angiology. 2015;66:508–13. doi: 10.1177/0003319714550309. [DOI] [PubMed] [Google Scholar]
  • 41.Mohammad S, Nguyen H, Nguyen M, et al. Pleiotropic effects of statins: untapped potential for statin pharmacotherapy. Curr Vasc Pharmacol. 2019;17:239–61. doi: 10.2174/1570161116666180723120608. [DOI] [PubMed] [Google Scholar]
  • 42.Guo H, Huang M, Yuan Q, et al. The important role of lipid raft-mediated attachment in the infection of cultured cells by coronavirus infectious bronchitis virus beaudette strain. PLoS One. 2017;12:e0170123. doi: 10.1371/journal.pone.0170123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jeon JH, Lee C. Cholesterol is important for the entry process of porcine deltacoronavirus. Arch Virol. 2018;163:3119–24. doi: 10.1007/s00705-018-3967-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Banach M, Stulc T, Dent R, Toth PP. Statin non-adherence and residual cardiovascular risk: there is need for substantial improvement. Int J Cardiol. 2016;225:184–96. doi: 10.1016/j.ijcard.2016.09.075. [DOI] [PubMed] [Google Scholar]
  • 45.Katsiki N, Athyros VG, Mikhailidis DP, Mantzoros C. Proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors: shaping the future after the further cardiovascular outcomes research with PCSK9 inhibition in subjects with elevated risk (FOURIER) trial. Metabolism. 2017;74:43–6. doi: 10.1016/j.metabol.2017.04.007. [DOI] [PubMed] [Google Scholar]
  • 46.Banach M, Penson PE. What have we learned about lipids and cardiovascular risk from PCSK9 inhibitor outcome trials: ODYSSEY and FOURIER? Cardiovasc Res. 2019;115:e26–31. doi: 10.1093/cvr/cvy301. [DOI] [PubMed] [Google Scholar]
  • 47.Momtazi-Borojeni AA, Sabouri-Rad S, Gotto AM, et al. PCSK9 and inflammation: a review of experimental and clinical evidence. Eur Heart J Cardiovasc Pharmacother. 2019;5:237–45. doi: 10.1093/ehjcvp/pvz022. [DOI] [PubMed] [Google Scholar]
  • 48.Khademi F, Momtazi-Borojeni AA, Reiner Ž, Banach M, Al-Rasadi KA, Sahebkar A. PCSK9 and infection: a potentially useful or dangerous association? J Cell Physiol. 2018;233:2920–7. doi: 10.1002/jcp.26040. [DOI] [PubMed] [Google Scholar]
  • 49.Jamialahmadi T, Panahi Y, Safarpour MA, et al. Association of serum PCSK9 levels with antibiotic resistance and severity of disease in patients with bacterial infections admitted to intensive care units. J Clin Med. 2019;8:E1742. doi: 10.3390/jcm8101742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.McAuley DF, Laffey JG, O’Kane CM, et al. Simvastatin in the acute respiratory distress syndrome. N Engl J Med. 2014;371:1695–703. doi: 10.1056/NEJMoa1403285. [DOI] [PubMed] [Google Scholar]
  • 51.National Heart, Lung, and Blood Institute ARDS Clinical Trials Network. Truwit JD, Bernard GR, Steingrub J, et al. Rosuvastatin for sepsis-associated acute respiratory distress syndrome. N Engl J Med. 2014;370:2191–200. doi: 10.1056/NEJMoa1401520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bos LD, Schouten LR, van Vught LA, et al. MARS consortium Identification and validation of distinct biological phenotypes in patients with acute respiratory distress syndrome by cluster analysis. Thorax. 2017;72:876–83. doi: 10.1136/thoraxjnl-2016-209719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Heijnen NFL, Bergmans DCJJ, Schnabel RM, Bos LDJ. Targeted treatment of acute respiratory distress syndrome with statins – a commentary on two phenotype stratified re-analysis of randomized controlled trials. J Thorac Dis. 2019;11(Suppl 3):S296–9. doi: 10.21037/jtd.2019.01.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Calfee CS, Delucchi KL, Sinha P, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med. 2018;6:691–8. doi: 10.1016/S2213-2600(18)30177-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kim J, Choi SM, Lee J, et al. Effect of renin-angiotensin system blockage in patients with acute respiratory distress syndrome: a retrospective case control study. Korean J Crit Care Med. 2017;32:154–63. doi: 10.4266/kjccm.2016.00976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Raiden S, Nahmod K, Nahmod V, et al. Nonpeptide antagonists of AT1 receptor for angiotensin II delay the onset of acute respiratory distress syndrome. J Pharmacol Exp Ther. 2002;303:45–51. doi: 10.1124/jpet.102.037382. [DOI] [PubMed] [Google Scholar]
  • 57.Driggin E, Madhavan MV, Bikdeli B, et al. Cardiovascular considerations for patients, health care workers, and health systems during the coronavirus disease 2019 (COVID-19) pandemic. J Am Coll Cardiol. 2020 doi: 10.1016/j.jacc.2020.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. https://www.ema.europa.eu/en/documents/product-information/kaletra-epar-product-information_en.pdf Last accessed 05 April 2020.
  • 59.Rosenson RS, Baker S, Banach M, et al. Optimizing cholesterol treatment in patients with muscle complaints. J Am Coll Cardiol. 2017;70:1290–301. doi: 10.1016/j.jacc.2017.07.752. [DOI] [PubMed] [Google Scholar]
  • 60.Banach M, Rizzo M, Toth PP, et al. Statin intolerance – an attempt at a unified definition. Position paper from an International Lipid Expert Panel. Arch Med Sci. 2015;11:1–23. doi: 10.5114/aoms.2015.49807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. https://www.escardio.org/Councils/Council-on-Hypertension-(CHT)/News/position-statement-of-the-esc-council-on-hypertension-on-ace-inhibitors-and-ang Last accessed 05 April 2020.

Articles from Archives of Medical Science : AMS are provided here courtesy of Termedia Publishing

RESOURCES