Review Article

Levosimendan Efficacy and Safety: 20 years of SIMDAX in Clinical Use

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Levosimendan was first approved for clinic use in 2000, when authorisation was granted by Swedish regulatory authorities for the haemodynamic stabilisation of patients with acutely decompensated chronic heart failure. In the ensuing 20 years, this distinctive inodilator, which enhances cardiac contractility through calcium sensitisation and promotes vasodilatation through the opening of adenosine triphosphate-dependent potassium channels on vascular smooth muscle cells, has been approved in more than 60 jurisdictions, including most of the countries of the European Union and Latin America. Areas of clinical application have expanded considerably and now include cardiogenic shock, takotsubo cardiomyopathy, advanced heart failure, right ventricular failure and pulmonary hypertension, cardiac surgery, critical care and emergency medicine. Levosimendan is currently in active clinical evaluation in the US. Levosimendan in IV formulation is being used as a research tool in the exploration of a wide range of cardiac and non-cardiac disease states. A levosimendan oral form is at present under evaluation in the management of amyotrophic lateral sclerosis. To mark the 20 years since the advent of levosimendan in clinical use, 51 experts from 23 European countries (Austria, Belgium, Croatia, Cyprus, Czech Republic, Estonia, Finland, France, Germany, Greece, Hungary, Italy, the Netherlands, Norway, Poland, Portugal, Russia, Slovenia, Spain, Sweden, Switzerland, UK and Ukraine) contributed to this essay, which evaluates one of the relatively few drugs to have been successfully introduced into the acute heart failure arena in recent times and charts a possible development trajectory for the next 20 years.

Disclosure:PP, TS and MK are full- or part-time employees of Orion Pharma. In the past 5 years, all other authors have received honoraria from Orion Pharma for educational lectures and/or unrestricted grants for investigator-initiated studies.

Received:

Accepted:

Published online:

Acknowledgements:The authors acknowledge Hughes Associates, Oxford, UK, for assistance in the editing of the manuscript. They thank Shrestha Roy for the initial graphic renditions of figures.

Correspondence Details:Piero Pollesello, Orion Pharma, PO Box 65, FIN-02101 Espoo, Finland. E: piero.pollesello@orionpharma.com

Open Access:

This work is open access under the CC-BY 4.0 License which allows users to copy, redistribute and make derivative works, provided the original work is cited correctly.

Origins of a Unique Cardiovascular Agent

Before the 1980s, therapy to enhance cardiac contractility in heart failure (HF) substantially meant oral digitalis glycosides, supplemented by beta-adrenergic agonists such as dopamine or dobutamine (introduced in the middle of the 1970s) in acute situations.1 It was therefore a matter of some note when the US Food and Drug Administration approved a new agent as a short-term IV therapy for patients with refractory HF. Amrinone was the product of a widespread research initiative that recognised the limitations of existing inotropic therapy and which, equipped with a new understanding of the cellular mechanisms of cardiac contractility, set out to develop what respected commentators of the time referred to as “non-glycoside, non-sympathomimetic positive inotropic agents”.2–4 Amrinone was the first agent to reach clinical use from the small but important family of phosphodiesterase (PDE) inhibitors, which would later include milrinone and enoximone.5,6 However, despite being non-sympathomimetic positive inotropic agents, all PDE inhibitors, in common with the catecholamines, were shown to be calcium mobilisers, probably due to their limited selectivity towards specific key PDE isoforms, and shared with catecholamines some unwanted effects intrinsic to any drug that raises intracellular calcium. In fact, all calcium mobilisers, by definition, exert an inotropic effect by providing increased ionic calcium levels for the contractile protein machinery, a process that may ultimately prove detrimental to individual cardiomyocytes and therefore also to patients.7

At about the same time, a new concept was proposed by the independent groups of J Caspar Rüegg in Heidelberg and R. John Solaro in Chicago, namely the potential of new agents to enhance the sensitivity to calcium of key targets in the contractile apparatus instead of increasing the intracellular calcium transient to augment contractility.8,9 In 1984, Rüegg et al. described the pharmacology of a new agent, later known as pimobendan, which combined PDE inhibitor activity with a direct calcium-sensitising effect.10

It was in this climate of innovation that the new chemical entity R-((4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl)hydrazono)propanedinitrile, known by the identifier OR-1259 at the time, appeared in the published records. An abstract was published in 1992 describing “a positive inotropic and vasodilatory compound with antiarrhythmic properties”.11 This preliminary report noted that OR-1259 exerted a positive inotropic effect despite a reduction in the voltage-sensitive Ca2+ current. As is not uncommon in abstract reports, the authors advised that “further studies… are in progress”.

In 1995, Heimo Haikala reported the findings of in-depth research into the mechanism of action of this agent in his pioneering paper.12 At the same time, a paper describing the binding of a new Ca2+ sensitiser, levosimendan, to recombinant human cardiac troponin C was also published.13 Those first descriptions may be regarded as foundation publications in the chronology of this drug, and a starting point for the PubMed-cited literature on levosimendan, which had expanded to almost 1,500 reports by the end of 2019.

Levosimendan was described as “a calcium sensitiser rationally designed and screened to act through its calcium-dependent binding to cardiac troponin C”, and the experimental basis for this description was set out in detail.12 From the beginning, clear mechanistic differences were spotted between levosimendan and several other drugs then in development, including pimobendan, MCI-154 and EMD 53998. Levosimendan was a first-in-class agent at the time of its emergence, promoting inotropy mainly through calcium sensitisation of cardiac troponin C (cTnC). More than 20 years later it remains, remarkably, an only-in-class drug, with a mechanism of action that clearly differentiates it from adrenergic agents.

Levosimendan, as reported by Pollesello et al. in 1994, binds to calcium-saturated human cTnC “in a hydrophobic patch of the N-domain near the site where the B helix is located when the protein is in its apo-form”.13 Figure 1 shows an original diagram from the 1994 paper proposing a molecular model of the drug–ligand complex.

That interaction leads to a stabilisation of the calcium-bound conformation of the regulatory (or N) domain of cTnC, which in turn causes a change in the conformation of the ‘switch’ region of cardiac troponin I (cTnI) and detachment of cTnI from actin filaments.14 Removal of the inhibitory effects of cTnI facilitates the formation of actin–myosin cross-bridges and the dis-inhibition of acto-myosin adenosine triphosphate (ATP) synthase, resulting in enhanced cardiac contractility.12,13,15–18 These findings, confirming that the binding of levosimendan to TnC is linked to calcium sensitisation, proved to be the first stage of what has since matured into a long-lasting research trail.19–22

The calcium-sensitising action of levosimendan is manifested as a leftward shift in the curve describing the relation between contractile force and calcium concentration, achieved via a direct effect on cTnC. That augmentation of contractility is not associated with increases in calcium transients, intracellular calcium or myocardial oxygen consumption and is not compromised by pre-treatment with beta-blockers. It should also be noted that the interaction between levosimendan and cTnC was shown to be more intense at high, systolic ionic calcium levels than at low, diastolic calcium levels, thus avoiding impairment of myocardial relaxation upon levosimendan administration.

In addition to its principal action as a calcium-sensitising agent, levosimendan was found in the course of its development programme to mediate the opening of ATP-dependent potassium channels (KATP channels) in vascular smooth muscle cells in various vascular beds.23 By this mechanism of action, levosimendan induces an increase in blood perfusion in key organs and a systemic vasodilatation when levosimendan is used at doses within the recognised therapeutic range, which means that the drug must be considered and used as an inodilator and not simply as an inotrope. An essential aspect of the pharmacology and clinical profile of levosimendan is that its perfusion enhancement and systemic vasodilation effects are mediated through different mechanisms and may therefore be disentangled from each other. Levosimendan – acting on KATP channels – has a different regional/peripheral versus systemic effect when compared with drugs such as the PDE inhibitors.24

Separate emphasis must be placed on the discovery that levosimendan also opens the KATP channels on the mitochondrial inner membrane.25,26 This effect has been associated with cardioprotection, infarct size reduction and mitigation of ischaemia/reperfusion injuries in a range of in vitro, ex vivo and in vivo studies in non-human species,27–31 and in clinical studies.32

The aforementioned effects deriving from calcium sensitisation and vasodilation are shared by the long-acting levosimendan metabolite OR-1896, which is formed in the intestine via a reduction–acetylation pathway.33–35 Free plasma concentrations of the parent drug and the metabolite are similar, but clinically meaningful plasma concentrations of pharmacologically active OR-1896 are detectable for days after an infusion of levosimendan and contribute to persistence of the therapeutic effect after administration of the parent drug is stopped.36

Early Molecular Model of the Levosimendan–cTnC Complex

Article image

Beyond these primary mechanisms, levosimendan has been identified as having a range of ancillary actions (often described as pleiotropic effects) that do not involve an enhancement of cardiac function, but which may be implicated in some of the clinical effects of and responses to levosimendan.37 These include anti-inflammatory, anti-oxidative and anti-apoptotic actions that may be exerted in non-cardiac organs, including the kidneys, liver, gut and splanchnic vasculature, lungs and/or respiratory muscles (Figure 2).

Levosimendan inhibits only one isoform of intracellular PDE enzymes (PDE-III) and in a highly selective manner. Of note, the PDE-III over PDE-IV isoform selectivity of levosimendan is the highest known to date, with a ratio of 10,000, compared with 14 for milrinone.38,39 It was proposed that inhibition of only the PDE-III isoform, not that of PDE-IV, would be insufficient to increase intracellular levels of cyclic adenosine monophosphate (cAMP) to the same levels as by simultaneous inhibition of the two isozymes.38 This lack of PDE dependence further differentiates levosimendan from non-selective PDE inhibitors, such as milrinone, and provides an explanation of their different pharmacological behaviours, as in the case of the oxygen consumption to force production ratios.38,40–42

Mode of Actions and Pharmacological Effects of Levosimendan

Article image

However, this interpretation is not unanimous. Maack et al. have proposed that PDE-III inhibition by levosimendan may indeed play a relevant role in the pharmacological effects of levosimendan.43 In that interpretation, PDE-III inhibition by levosimendan synergises with Ca2+ sensitisation for the resulting inotropic action. Interestingly, from this synergy, the authors predict that the more beta-adrenergic receptors are pre-activated by endogenous or exogenous catecholamines, the more pronounced will be the inotropic effect of levosimendan, and the more this effect would be mediated by PDE-III inhibition rather than by Ca2+ sensitisation. Conversely, at low pre-activation of beta-adrenergic receptors (such as during pharmacological beta-blockade), the Ca2+-sensitisation effect of levosimendan would become more important for inotropy. The take-home message of a consensus paper from the Translational Working Group of the Heart Failure Association of the European Society of Cardiology (ESC) is that long-term use of drugs that exclusively target adrenergic signalling (e.g. catecholamines and PDE inhibitors) is associated with adverse outcomes, while levosimendan, with its hybrid calcium sensitisation and PDE-III inhibition properties, should be given the benefit of the doubt and further attention.43

The effect of levosimendan has also been studied in the presence of beta-blockers and/or inopressors. Xanthos et al. reported that the combination of epinephrine, atenolol and levosimendan, when given during cardiac arrest and resuscitation in a pig model, resulted in improved 48-hour survival and post-resuscitation cardiac function.44 Concurrently, Lochner et al. reported that the effects of levosimendan were not blunted by the presence of beta-blockers, as in the case of adrenergic inotropes.45

Levosimendan entered formal clinical evaluation and development in acute heart failure (AHF) in the mid-1990s.46–48 Initially, it was established that IV levosimendan produced dose-dependent increases in cardiac output (CO) and decreases in pulmonary capillary wedge pressure (PCWP; Figure 3). Those effects were not accompanied by significant increases in myocardial energy consumption, thus confirming the paradigm envisioned on the basis of the preclinical data.28,49–51

From Bench to Bedside

Levosimendan entered clinical trials profiled as a novel inotrope with potential for the short-term treatment of acutely decompensated chronic HF. The regulatory studies programme devised to evaluate it in this indication enrolled almost 4,000 patients (Table 1), and produced the following key insights.

Clinical Effects

Haemodynamic Effects

The haemodynamic effects of levosimendan seen in preclinical studies were confirmed. In patients with AHF, levosimendan achieves significant dose-dependent increases in CO and stroke volume and decreases in PCWP, mean blood pressure, mean pulmonary artery pressure, mean right atrial pressure and total peripheral resistance.52

In line with preclinical data, clinical studies have confirmed that levosimendan does not have a negative effect on diastolic function. In contrast, levosimendan has lusitropic effects.53,54 Inodilation is not only seen in the left side of the heart; right ventricular contractility is also improved and pulmonary vascular resistance is decreased.55–57

Pharmacokinetics in Clinical Trials

As anticipated in non-clinical studies, in humans the haemodynamic effects of a 24-hour infusion of levosimendan are protracted for several days in patients with AHF due to the presence of an active metabolite (Figure 4A).58–60

Effects on Neurohormones

Rapid and sustained reductions in levels of natriuretic peptides were characteristic of levosimendan in its regulatory clinical trials.61–63 The effect on natriuretic peptides closely follows the haemodynamic effects; both are evident for at least 1 week after the levosimendan infusion period (Figure 4B).58 In the trial Survival of Patients with Acute Heart Failure in Need of IV Inotropic Support (SURVIVE), in patients with acute decompensated HF, changes in brain natriuretic peptide (BNP) levels up to 5 days after the start of infusion of levosimendan could be seen, which was not the case after 48 hours of treatment with dobutamine.61

Impact on Signs and Symptoms in AHF

Levosimendan induces a rapid and sustained improvement in symptoms, as evidenced by Packer et al. and Slawsky et al.62,64 In the second of those studies, relief of dyspnoea was reported in 29% of levosimendan-treated patients compared with 15% of the placebo-treated patients 6 hours after starting the infusion (p=0.037).64 Improvement in symptoms was evident for up to 5 days.62 Data on the use of rescue medications in the Randomised Evaluation of IV Levosimendan Efficacy (REVIVE) programme further confirm the effectiveness of levosimendan for symptom relief (Table 2).62 Dyspnoea and fatigue symptoms also responded better to levosimendan than to dobutamine in the Levosimendan Infusion versus Dobutamine (LIDO) trial, although not to the level required for statistical significance.63

Clinical Outcomes

Hospitalisations

Patients treated with levosimendan in the LIDO study spent significantly more days alive and out of hospital than dobutamine-treated patients in a retrospective 180-day follow-up analysis (median 157 versus 133 days; p=0.027).63 In the Randomised Study on Safety and Effectiveness of Levosimendan in Patients with Left Ventricular Failure After an Acute Myocardial Infarction (RUSSLAN), the combined risk of death and worsening HF was significantly lower in patients treated with levosimendan than in the control group during the infusion period (2% versus 6%; p=0.033) and at 24 hours (4% versus 9%; p=0.044).65 In the REVIVE II study, a greater percentage of patients treated with levosimendan than placebo were released within 5 days (46% versus 37%) and the mean duration of the initial hospitalisation was almost 2 days shorter (7.0 versus 8.9 days).62 No significant intergroup difference was recorded in the SURVIVE trial (p=0.3).61

Mortality

Thirty-one-day mortality in the LIDO trial indicated a survival advantage from levosimendan (mortality rate 8%, versus 17% with dobutamine, HR 0.43, p=0.049).63 This was corroborated in a retrospective extension of follow-up to 180 days (mortality rate 26%, versus 38% with dobutamine, HR 0.57, p=0.029). In RUSSLAN, a survival benefit from levosimendan persisted at 180-day follow-up (23% versus 31%; p=0.053).65

In the REVIVE and SURVIVE trials there were no significant differences in 3- and 6-month overall survival between the study groups.61,62 However, there was evidence of a survival gain from levosimendan treatment in SURVIVE patients who had a history of chronic decompensated HF or who were using beta-blockers.66 In patients with existing chronic HF (88% of the study population), mortality was lower in the levosimendan group than in the dobutamine group at day 5 (3.4% versus 5.8%, HR 0.58, 95% CI [0.33–1.01], p=0.05) and at day 14 (7.0% versus 10.3%, HR 0.67, 95% CI [0.45–0.99], p=0.045). In patients who used beta-blockers (50% of the study population), mortality was significantly lower for levosimendan than for dobutamine at day 5 (1.5% versus 5.1%, HR 0.29, 95% CI [0.11–0.78], p=0.01).

Change in Cardiac Output and Pulmonary Capillary Wedge Pressure

Article image

Safety

A safety summary prepared by Orion Pharma in its capacity as sponsor of the regulatory studies found no difference in the proportion of patients with reduction in arterial blood pressure in response to treatment (23.1% versus 23.1%), although REVIVE II, considered as a single study, diverged from this overall trend by showing more hypotension in the levosimendan arm.62 In 2012, Landoni et al. collected data from 5,480 patients in 45 randomised clinical trials and also carried out a meta-analysis of the adverse events.67 No signals were seen for MI (data from 25 studies, RR 0.789, 95% CI [0.522–1.185], p=0.3), ventricular arrhythmias (data from nine studies, RR 0.885, 95% CI [0.611–1.281], p=0.5) or supraventricular arrhythmias (data from 19 studies, RR 1.005, 95% CI [0.782–1.291], p=0.9), but a numerical trend was seen for hypotension (data from 22 studies, RR 1.389, 95% CI [0.996–1.936], p=0.53). There are some contradictory or indirect and inconclusive reports related to the impact of levosimendan on platelet function, but a recent meta-analysis of nine randomised controlled trials (RCTs) found that levosimendan did not increase postoperative bleeding risk.68,69 Moreover, in the supplementary appendix of the large regulatory trial Levosimendan in Patients with Left Ventricular Systolic Dysfunction Undergoing Cardiac Surgery Requiring Cardiopulmonary Bypass (LEVO-CTS) no signs of increase in peri- or post-procedural haemorrhage were seen after treatment with levosimendan.70

Regulatory Clinical Trials of Levosimendan

Article image

Pharmacokinetics of Levosimendan

Article image

Conversely, a later, independent meta-analysis of data from more than 5,000 patients indicated increased risks of extrasystoles (RR 1.88, 95% CI [1.26–2.81]), hypotension (RR 1.33, 95% CI [1.15–1.53]) and headache or migraine (RR 1.94, 95% CI [1.54–2.43]) when compared with reference therapies.71 Retrospective analyses of the REVIVE II dataset identified low blood pressure at baseline as a possible risk factor for the use of levosimendan, and the current, approved Summary of Product Characteristics reflects that finding.72

Dosing

Levosimendan is given as a continuous infusion of 0.05 or 0.1 or 0.2 
µg/kg/min for 24 hours, which may be preceded by a loading dose (bolus) of 6–12 µg/kg in 10 minutes. The loading dose was used in the active-controlled regulatory studies LIDO and SURVIVE, in which dobutamine served as comparator. Given that the elimination half-life of dobutamine is a few minutes while that of levosimendan is approximately 1 hour, the haemodynamic effects of dobutamine are seen almost immediately after the infusion is started, whereas a bolus of levosimendan is needed to see immediate effects. For consistency, all other studies in the regulatory clinical programme were designed to include a bolus dose, followed by a maintenance infusion. It was later found that, in the case of hypovolaemia or initial low blood pressure, a levosimendan bolus could be associated with hypotension or arrhythmias. Therefore, the use of an initial bolus of levosimendan is now generally not recommended, and it has often been avoided in clinical practice and used only if an instant effect is sought and the systolic blood pressure (SBP) is adequate.73,74

Into Regular Clinical Use

The experience gained in regulatory studies provided the basis for the first approval of IV levosimendan, which was introduced in Sweden in 2000 for the management of AHF with the name SIMDAX. Since then, more than 60 jurisdictions have approved the drug, including most of the countries of the EU and Latin America. Levosimendan is currently in active clinical evaluation in the US.

Use of Rescue Medications in the REVIVE Programme

Article image

In the 20 years since its first introduction, IV levosimendan has been one of the notably few successful drugs entering the market in an underserved area of cardiovascular medicine; attempts at drug innovation in AHF have been characterised by repeated disappointments (either partial or total) or contradictory findings that have hindered progress.75

Levosimendan itself has not been immune to some of the frustrations of research in this area; in particular, the non-univocal findings on 6-month mortality in its regulatory studies complicated the process of establishing its therapeutic niche. Innovation in this area may have been poorly served by a regulatory emphasis on longer-term survival effects. This was perhaps misaligned with clinical realities and led to an emphasis on large trials which, by aggregating data from patients with different underlying pathophysiologies plus variations in both pharmacological and non-pharmacological treatments, may have generated signal-to-noise ratios that precluded the identification of a meaningful effect on the central endpoint of all-cause mortality/survival. The unsuitability of all-cause long-term mortality as an index of therapeutic effect was acknowledged by experts in the field of HF about a decade ago, but that realisation came too late to influence the conduct of the regulatory trials of levosimendan.76,77

These obstacles notwithstanding, pooled analysis of the outcomes of the levosimendan regulatory trials provided strong indications, albeit not always statistically conclusive proof, of an overall survival benefit (Figure 5). Extensive experience with levosimendan has been accrued in smaller, often single-centre, non-regulatory studies. Many of those studies indicate a survival benefit from levosimendan, a finding affirmed in meta-analysis.67

Levosimendan has been evaluated in more than 200 clinical trials during its lifetime, in an extensive range of therapeutic settings. Experience in all those areas has been evaluated in meta-analyses, 31 of which have been conducted in the past 3 years (Figure 6). In every instance, levosimendan was associated with a favourable impact on the outcomes under consideration but, depending on the data selected, statistical significance in some cases remained elusive. Key therapeutic areas analysed in this way have included AHF, advanced HF (AdHF), cardiac surgery and sepsis, all of which have provided indications of benefit from levosimendan therapy.

The broadly affirmative findings of these exercises may be compared with similar appraisals of dobutamine and PDE inhibitors, which have been associated with overall worse mid-to-long-term prognosis.78–80 These contrasting findings highlight the distinction between inotropes that act, via either adrenergic or PDE-targeted pathways, to increase intracellular cAMP levels in cardiomyocytes and levosimendan, realising the ambitions of its inventors by promoting cardiac contractility without compromising the longer-term viability of cardiac muscle cells. This distinction is also illuminated by findings from the Acute Heart Failure Global Survey of Standard Treatment (ALARM-HF) registry, data from which were strongly indicative of survival benefit from levosimendan vis-à-vis adrenergic/calcium-mobilising inotropes, such as dobutamine.81

Effect of Levosimendan on Survival in the Regulatory Clinical Trials

Article image

Levosimendan in Current Use

The non-univocal findings of long-term survival benefit from short-term treatment with IV levosimendan have not prevented the drug from establishing itself in the therapeutic repertoire; it has been used in almost 2 million patients since 2000, when its first market authorisation was granted by the Swedish regulatory authorities on the basis of the data available at that time.

Its favourable, rapid and sustained impact on haemodynamics, neurohormone levels and symptoms in acute decompensated HF are undisputed and of clear therapeutic value. Formal acknowledgement of that value emerged in 2005, when it was mandated in the ESC guidelines.82 In the subsequent European guidelines (2008, 2012), the endorsements of levosimendan were more cautious, reflecting a general dissatisfaction of the HF medical community with the concept of inotropy.83,84 Levosimendan is currently recommended in the acute treatment of HF to reverse the effect of beta-blockade, if beta-blockade is thought to be contributing to hypotension with subsequent hypoperfusion.85

Results of 64 Meta-analyses of Levosimendan Clinical Trials

Article image

Common Concomitant Conditions in Acute Heart Failure

Article image

Due to the large therapeutic field they encompass, the European guidelines on acute and chronic HF are not as detailed as they could be and, in recommending therapeutic agents, ignore some of the different aetiologies and manifestations of AHF. Supplementary information and recommendations can be found in more than 20 expert consensus papers co-authored by more than 180 clinicians from 30 countries who have discussed when and how to use levosimendan in different therapeutic settings, including AHF and cardiogenic shock,74,81,86–88 AdHF,89–92 perioperative use,93–95 and use in the intensive care unit (ICU),96 and who have described its cardiorenal effects,88,97 its effects on quality of life,98,99 exercise performance,100 lung function,101 and pharmaco-economic considerations.102

In the context of a 20-year retrospective it is worth noting at this point that its complex mode of action might have had the potential to disadvantage levosimendan both in fact and in perception. In fact, that plurality of effects has emerged as both an important aspect of the drug’s clinical versatility and usefulness and as a stimulant to informed speculation among experts and to medical research.73,92,94,96,103

Levosimendan in Acute Settings

The most recent ESC guidelines, issued in 2016, identify short-term treatment with IV levosimendan (along with adrenergic inotropes or PDE inhibitors) as an option in the acute-phase management of “patients with hypotension (SBP <90 mmHg) and/or symptoms of hypoperfusion despite adequate filling status, to increase CO, increase blood pressure, improve peripheral perfusion and maintain end-organ function”.85 The ESC statement further endorses the short-term use of levosimendan to circumvent the effects of beta-blockade “if beta-blockade is thought to be contributing to hypotension with subsequent hypoperfusion”. The high proportion of patients now receiving beta-blockers as part of the treatment repertoire for chronic HF means that levosimendan has become an important resource in the management of acute decompensations in those patients.

The vasodilator dimension of levosimendan’s pharmacology is pertinent to the drug’s use in low-output states such as AHF, in which a key pathology is organ hypoperfusion. A drug that both augments CO and improves vasodilatation may be expected to have a more favourable impact in some cases than an agent that acts on CO alone.104

In this context, it is important to register that, in many acute settings, hypoperfusion and hypotension may not necessarily be only an effect of inadequate myocardial contractility but may also be related to shock-specific changes in vascular tone and vasodilatation. Thus, besides the need to exclude or correct inadequate volume status, concomitant treatment with a vasopressor agent before embarking on a course of inotropic therapy must also be considered. The importance of correcting inadequate volume before embarking on a course of vasodilatory or inotropic therapy must also be considered and monitored during therapy. These observations may be usefully contextualised into a ‘right patient, right drug’ schema that can be used to guide vasoactive and/or inotropic therapy (Table 3).105 The first phase of this schema examines whether or not an inotrope is needed at all and includes exclusion of otherwise treatable causes or the availability of viable alternatives. The next step is to identify the most suitable inotrope; levosimendan figures prominently in several categories, including cardiogenic shock, cardiac surgery and right ventricular failure.

Cardiogenic Shock

The purpose of inotropic support in cardiogenic shock secondary to left ventricular dysfunction is to aid the failing left ventricle by unloading it, increasing left ventricular output and improving coronary blood flow, and hence myocardial perfusion, at the same time decreasing pulmonary oedema. The inodilator profile of levosimendan provides a possibly more complete response to those needs than pure inotropic agents and its ability to promote inotropy with little or no adverse effect on metabolic rate, energy demand or oxygen consumption may be a bonus. The use of levosimendan may be beneficial in part via a substitution effect in which it reduces the need for catecholaminergic agents, which have less favourable effects on oxygen and energy consumption at the cellular level and a propensity to increase mortality.106

Formal experience with levosimendan in cardiogenic shock is limited, but it appears to be generally well-tolerated, to improve multiple indices of cardiac function and to reduce systemic vascular resistance.106–111 Levosimendan has also been reported to restore ventriculoarterial coupling and improve left ventricular function in various settings; this may be a further benefit in cardiogenic shock, but this conjecture is currently untested.112,113

Takotsubo Syndrome

An example of cardiogenic shock in which treatment with levosimendan presents unique benefits is takotsubo syndrome, or stress cardiomyopathy.114 Takotsubo-induced HF and/or cardiogenic shock is commonly treated with aggressive diuresis, haemodynamic support and inotropic drugs. The fact that catecholamines may be implicated in its pathogenesis suggests that catecholamine inotropes may be contraindicated, because these drugs increase cAMP within the cell, increase myocardial oxygen consumption and may worsen myocardial stunning. Levosimendan, in contrast, as a non-catecholamine inotrope that does not increase myocyte cAMP or oxygen consumption, is a rational therapeutic option in takotsubo-related cardiogenic shock.115–117

Cardiac Surgery

Levosimendan has been studied in more than 40 clinical trials in cardiac surgery, with indications emerging that it can reduce the risk of low-output cardiac syndrome (LCOS) or be effective in treating postoperative LCOS. The scale of this benefit (derived from a meta-analysis of 14 studies) is moderate but tangible, and is more marked in patients with baseline low left ventricular ejection fraction.118

A recalculation incorporating data from three recent large RCTs (Levosimendan in Coronary Artery Revascularisation [LICORN], Levosimendan to Reduce Mortality in High Risk Cardiac Surgery Patients [CHEETAH] and LEVO-CTS) causes dilution of the effect size of that estimate.119–122 Expert advice derived from these new data is that “levosimendan cannot be at the moment recommended for routine use in all cardiac surgery settings”.122 However, there appears to be potential for significant mortality benefit in some subgroups of patients, such as those with low ejection fraction or those undergoing isolated coronary artery bypass grafting (CABG) procedures.123,124 Whether these findings may be interpreted as a lack of efficacy of levosimendan in patients undergoing valve surgery due to differences in the underlying pathophysiology or dosing, or as a result of procedure-specific differences in surgical and perfusion management (i.e. too low a dose applied before cardiopulmonary bypass or use of crystalloid cardioplegia solutions), needs to be addressed in future studies.

The emphasis on postoperative mortality in these recent studies was substantially driven by regulatory requirements in the design of LEVO-CTS. Whether that outcome is the most relevant or revealing for evaluation of an intervention is an open question. Levosimendan exhibited efficacy in other measures, including a lower incidence of LCOS, less need for rescue catecholamines for inotropic support and augmentation of the cardiac index.118 Methodological difficulties also affected the interpretation of the LICORN and CHEETAH trials.

These caveats notwithstanding, mortality was numerically lower in levosimendan-treated patients in LEVO-CTS.118 In addition, the safety profile of levosimendan revealed in all these recent trials identifies it as arguably the safest agent among the broad grouping of inotropes and inodilators. There was no significant excess of arrhythmias or hypotension and no increase in mortality in levosimendan-treated patients.118

Encouragement for further evaluation of levosimendan in this area comes from other recent investigations. Wang et al. analysed data from 21 randomised trials (n=1,727) and calculated that IV levosimendan in patients undergoing CABG was associated with significant reductions in mortality rate (p=0.001) and postoperative AF (p=0.04), with benefit mostly restricted to patients pre-treated in advance of an isolated CABG procedure; on-pump status also affected outcomes.123 Levosimendan was associated with a higher incidence of hypotension (OR 2.26). These data are consistent with the findings from LEVO-CTS and offer indications of future lines of clinical appraisal.118,124 See also Weber et al.125

Right Ventricular Failure

Determinative randomised trials of levosimendan in right ventricular failure (with or without pulmonary hypertension) have yet to be conducted, but a recent meta-analysis of 10 studies of levosimendan in acute right-sided HF identified statistically robust benefits over placebo, with increases in tricuspid annular plane systolic excursion and ejection fraction, plus reductions in systolic pulmonary artery pressure (p=0.0001) and pulmonary vascular resistance (p=0.003).126 Adverse events were reported not to differ significantly between groups.

Effects on Renal Function in HF and Critical Illness

HF is a systemic syndrome involving the kidneys, lungs and liver, with a great impact on prognosis, and effects of cardiovascular drugs on non-cardiac organs are of the utmost importance.127 Evidence for a renal-protective action of levosimendan has been reported from preclinical experiments.128–130 It has been proposed that levosimendan may cause selective vasodilation on the afferent arterioles of the renal glomeruli, thus improving renal filtration.97 This suggestion is compatible with findings from the LIDO trial, in which levosimendan treatment was associated with an increase in estimated glomerular filtration rate (eGFR) but treatment with dobutamine was not, even though both drugs increased cardiac index and urine output.63 It is also consistent with recent reports by Fedele et al. and by Lannemyr et al.131,132 The substantial enhancement of eGFR observed in the second of those studies was not accompanied by impairment of renal oxygenation, given that renal oxygen delivery increased in proportion to the increase in eGFR. Non-impairment of the renal oxygen supply–demand relationship despite eGFR enhancement during levosimendan exposure has also been reported by Bragadottir et al.133 A recent report shows that the eGFR enhancement effect of levosimendan is not shared with milrinone.134

Current Clinical Applications of IV Levosimendan

Article image

Data on the effects of levosimendan on renal function in various clinical situations, including cardiac surgery and critical illness, have been collated and the results support a renal-protective effect, making levosimendan the inotrope of choice in the case of worsening cardio-renal syndrome.135–138 However, in all these situations, specifically designed prospective trials of adequate statistical power will be needed to confirm the effects and their clinical consequences.

Levosimendan in AdHF

Adoption of repeated intermittent cycles of IV levosimendan for the treatment of AdHF has been a significant milestone in both the lifecycle of the drug and the management of a complex aspect of HF. Patients with AdHF are on a trajectory ultimately either to a definitive intervention through heart transplantation or the implantation of a left ventricular assist device (LVAD), or to a palliative care pathway. Goals of therapy in AdHF include haemodynamic stabilisation and preservation of functional capacity, mitigation of symptoms and preservation of health-related quality of life. Prevention of HF-related hospitalisation is another key goal, both as a desirable outcome per se and as a way of averting the markedly worsened mortality that accompanies hospitalisation.139,140

All of the pharmacological properties of levosimendan outlined earlier – notably its metabolite-mediated persistence of effect – make it well-suited for repeated or intermittent use in the management of AdHF.

Three randomised, placebo-controlled, double-blind clinical trials, Randomised Trial Investigating the Efficacy and Safety of Pulsed Infusions of Levosimendan in Outpatients with Advanced Heart Failure (Levo-Rep; NCT01065194), Levosimendan Intermittent Administration in Outpatients: Effects on Natriuretic Peptides in Advanced Chronic Heart Failure (LION-HEART; NCT01536132) and Long-term Intermittent Administration of Levosimendan in Patients with Advanced Heart Failure (LAICA; NCT00988806), have examined the application of repeated cycles of levosimendan therapy in this setting.141–143 All these studies demonstrated that repeat-cycle levosimendan reduces N-terminal pro-BNP (NT-proBNP) levels and there were repeated and clear demonstrations of trends towards reductions in HF readmissions and mortality that are consistent with, and corroborate, the findings of meta-analyses.144,145 A recognised overall conclusion from these studies is that repetitive application of levosimendan is feasible and safe in an outpatient setting.141–143 Notably, onset destabilisation is not invariably an immediate-onset event, however, and it may be possible to identify opportunities when timely recognition of – and intervention on – signs and symptoms of decompensation may avoid unplanned/urgent hospitalisations due to haemodynamic crises.

The need for a larger randomised study (or studies) in this area is being addressed by the Repetitive Levosimendan Infusion for Patients with Advanced Chronic Heart Failure trial (LEODOR; NCT03437226), a multicentre, randomised, double-blind, placebo-controlled, three-arm trial, which will examine the impact and safety of intermittent levosimendan therapy, started during the vulnerable phase after a recent hospitalisation for HF.146 Treatment effectiveness will be assessed using a hierarchical composite clinical endpoint consisting of time to death or urgent heart transplantation or implantation of a ventricular assist device; time to non-fatal HF hospitalisation requiring IV vasoactive therapy; and time-averaged proportional change in NT-proBNP. Basing the trial on such an outcome measure should enhance the power to examine whether, compared with placebo, repeated use of levosimendan is associated with greater clinical stability over the course of subsequent weeks.

In addition to its use in maintaining haemodynamic stability in patients with AdHF, preoperative use of IV levosimendan in patients undergoing implantation of an LVAD, or identification of LVAD candidates, has been reported to be “generally well-tolerated and not interrupted because of side effects” and associated with significant improvements in end-organ function, although with similar early mortality rates.147 This is an application where a substantial expansion in the use of levosimendan may be anticipated.

The current clinical applications of IV levosimendan are summarised in Table 4.

The Next 20 Years of Levosimendan

In addition to the LEODOR study, there are currently more than 20 investigator-initiated clinical trials in progress in which levosimendan is being evaluated for possible therapeutic benefits. These include a study designed to examine the effect of a single 24-hour infusion of levosimendan to prevent rehospitalisation in patients with severe systolic HF (NCT03764722); Effect of Levosimendan or Placebo on Exercise in Advanced Chronic Heart Failure (LOCO-CHF; NCT03576677), a placebo-controlled appraisal of the effects of IV levosimendan on exercise capacity in patients with advanced chronic HF; and research into the effects of levosimendan in acute kidney injury after cardiac surgery (LEVOAKI; NCT02531724). The Early Management Strategies of Acute Heart Failure for Patients with NSTEMI study (EMSAHF; NCT03189901) is exploring whether early use of levosimendan in patients with acute MI combined with elevated BNP/NT-proBNP may reduce the risk of emergent AHF and improve outcome.

Developments in the technology and science of telemedicine and telemonitoring may make this a practical proposition in the foreseeable future. The ambition (already under active exploration and with progress further accelerated by the introduction of artificial intelligence into the diagnostic loop) is to develop patient monitoring to such a degree of immediacy and accuracy that overt decompensations may be wholly avoided by prompt, appropriate clinical responses to the first signs of deterioration.148,149 Intermittent IV levosimendan may be an appropriate intervention in this ‘acute but non-hospitalised’ scenario, depending on the clinical circumstances of an individual patient. Investigations in this direction can be expected, although these are likely to be driven primarily by developments in telemedicine technologies, rather than by any focus on specific medical interventions.150

A range of ICU situations has been identified in which levosimendan may offer clinical benefits, either as an adjunct to existing interventions or as an alternative to conventional therapies. These situations include haemodynamic support in cardiac critical care,151 haemodynamic support in septic cardiomyopathy,152,153 weaning from the ventilator,154 weaning from venoarterial extracorporeal membrane oxygenation after cardiac surgery,155–159 and renal failure and kidney protection in cardiorenal syndrome.97,130,132,160–166

In several of these areas, notably low CO syndrome, cardiogenic shock, takotsubo cardiomyopathy and sepsis, a substantial element of any benefit accruing from use of levosimendan may be attributable to the substitution of a non-adrenergic stimulant for conventional catecholaminergic agents, such as dobutamine, thereby averting some of the potential toxic complications of adrenergic overstimulation.167

‘Decatecholaminisation’ of the medical armamentarium is a developing line of practice in the management of critically ill patients.168–176 As an established non-adrenergic vasoactive agent that offers positive cardiovascular effects (e.g. ventriculoarterial recoupling, decongestion), as well as potentially advantageous ancillary effects on kidney function and cellular-protective actions, levosimendan is both a therapeutic resource and experimental tool for investigating this new approach.161,177–184

Developments in these areas, although already pioneered by promising exploratory studies will, inevitably, require well-designed and properly powered RCTs.151–153,185,186

Central to future investigations will be the identification of robust and relevant endpoints. An overemphasis on crude mortality may not be the most informative metric by which to judge outcomes. Trials directed towards establishing the best overall therapeutic strategy may be more progressive than studies framed to position any one agent as the best for a particular purpose or situation. Various commentaries have examined the challenges of conducting clinical trials in critically ill patients.187–190 Of notable significance in this respect is the work of Mebazaa et al., who have sought to apply lessons learned from trials in AHF to the more flexible and less prescriptive design of Phase III studies in sepsis.191

Principal among those lessons is the need to move towards composite primary endpoints, with isolated all-cause mortality addressed as a safety signal, not a primary marker of effectiveness. From that perspective, the model of HF as a complex condition of multifactorial causes and pathophysiology, and one therefore unlikely to be moderated by a single intervention on a simple outcome measure, such as total mortality, is an example of a needlessly restrictive approach to clinical trials design that has had a dampening effect on the development of needed therapies. In addition, there has been recent discussion about how syndrome-attributable risks of critical illness-associated diagnoses have often likely been overestimated using common statistical methods, contributing to very low success rates in this field.192 The authors of this article are unanimously supportive of that view, which we feel has frustrated advances in HF care. It is to be hoped that our colleagues in cardiac critical care are able to benefit in the design of their own clinical trials by examining some of the methodological missteps and misplaced emphases that have hampered some aspects of cardiology research in recent decades.

Levosimendan has been approved by Chinese authorities on the basis of several local corroborative regulatory clinical trials, and SIMDAX will enter that important market this year.161,193–195

The use of levosimendan in paediatric patients is currently contraindicated due to a lack of regulatory studies. A few investigator-initiated studies have been performed. The largest published study included retrospectively gathered data on 484 levosimendan infusions delivered to 293 patients at a single paediatric ICU; the majority of the patients (65%) were aged 12 months or younger.196 Levosimendan postponed or reduced the need for mechanical cardiac support in children with cardiomyopathy or who were undergoing cardiac surgery. In other reports, levosimendan was compared to the PDE inhibitor milrinone and found either to be similarly efficacious or superior.197–199 In a randomised double-blind study in children younger than 4 years of age undergoing cardiac surgery, patients receiving levosimendan had significantly higher cardiac index and lower pulmonary artery pressure than children receiving dobutamine.200 There is a strong rationale to perform properly powered clinical regulatory trials on the paediatric use of levosimendan in the near future.

Research and Development Outside Cardiology

Respiratory Function

Respiratory muscle dysfunction may develop in the course of several diseases, including chronic obstructive pulmonary disease (COPD), HF and critical illness.201,202 In addition to atrophy, reduced calcium sensitivity of contraction plays an important role in respiratory muscle weakness in these conditions.203,204 Levosimendan has been studied in non-cardiac muscle, especially in respiratory muscles. In vitro studies have demonstrated that levosimendan improves calcium sensitivity of force generation in diaphragm fibres from healthy subjects and patients with COPD.203 In a physiological study, levosimendan at a clinically used dose improved diaphragm contractile efficiency by 21% and reversed diaphragm fatigue induced by inspiratory muscle loading.205 In a placebo-controlled randomised study, the effects of levosimendan were evaluated in critically ill patients being weaned from mechanical ventilation.206 There was no difference in the primary endpoint of contractile efficiency between groups, while tidal volume and minute ventilation were both significantly higher in the levosimendan group, and arterial CO2 tension significantly lower.206 Improved respiratory mechanics may facilitate liberation from mechanical ventilation, although this requires further clinical studies.

Pulmonary Hypertension

A Phase II regulatory clinical trial is currently underway in the US on the repeated use of IV levosimendan for pulmonary hypertension in patients with HF and preserved ejection fraction (NCT03541603). The results are expected in 2020.

Motor Neurone Disease

There is interest in the potential of oral levosimendan in the management of amyotrophic lateral sclerosis (ALS). ALS is characterised by a progressive muscular paralysis arising from motor neurone degeneration.207 The disease eventually involves most skeletal muscles, plus the diaphragm and other respiratory muscles, leading to death from respiratory failure.208,209 It is the most common neurodegenerative disorder of midlife, with incidence and prevalence increasing with age.210 ALS is currently incurable and the medical options are limited. No treatment is currently approved to enhance motor function in ALS and recent clinical experiences have produced mixed results.211–214

The physiological and pharmacological rationale for levosimendan in ALS rest on the fact that both the diaphragm and skeletal muscle express genes for the slow-twitch (or cardiac) isoform of TnC (the diaphragm consists of approximately 50% slow-twitch fibres).215 As a calcium sensitiser with cTnC as molecular target, levosimendan can thus strengthen contractility also in the diaphragm and skeletal muscle. The multifaceted pharmacology of levosimendan may also provide supplementary clinical impact in patients with ALS through the already-mentioned range of pharmacological effects not directly related to the drug’s calcium-sensitising action.37,216

Positive effects of short-term oral levosimendan in patients with ALS were seen in the Effects of ODM-109 on Respiratory Function in Patients With ALS (LEVALS; NCT02487407) study, a Phase II trial that used a randomised, double-blind, placebo-controlled, cross-over design to evaluate the efficacy and safety of oral levosimendan in patients with definite or probable ALS. The 66 patients enrolled had experienced symptoms of ALS for between 12 and 48 months and had an early decline in respiratory function. Therapy consisted of 2 weeks of oral levosimendan at doses of 1 or 2 mg/day or placebo, administered in random order during three study periods separated by a washout period.217

The Phase III Effects of Oral Levosimendan (ODM-109) on Respiratory Function in Patients with ALS trial (REFALS; NCT03505021) is ongoing in North America, Europe and Australia. An open-extension phase of REFALS should offer important insights into the long-term safety and efficacy of oral levosimendan in ALS.

Translational and Early Development Phases

Other trials are looking at the effects of IV levosimendan on cellular metabolic alterations in patients with septic shock (NCT02963454) and the possibility that IV levosimendan may improve the prognosis in acute respiratory distress syndrome (NCT04020003).

Other recent research on the effect of levosimendan on oxidative stress in a mouse model of diabetes showing an effect of the drug in preventing memory impairment has opened up a new possible development path.218,219 A separate report, again on mice, highlighted the protective effects elicited by levosimendan against liver ischaemia/reperfusion injury.220

Of particular interest, new pharmaceutical agents are currently being developed with levosimendan and cTnC as a pharmacophore model in unexpected fields such as oncology.221

Conclusion

In the field of short-term haemodynamic treatments for acute cardiac care, levosimendan represents a rare case of an inotrope approved by regulatory authorities in the past 20 years. The approval was based on data from three Phase III clinical studies in which the endpoints were reached (LIDO, RUSSLAN and REVIVE).62,63,65 The safety data collected during those 20 years of clinical use are unprecedented and superior to those for any of the other inotropes or inodilators.222 In a recent consensus paper, the authors evaluated whether “the nearly total absence of evidence of benefit with some of the traditional IV drugs used in AHF and AdHF (such as the catecholamines or the PDE inhibitors) would warrant their elimination from routine use in favour of treatments where such evidence has been accrued (e.g. for levosimendan)”.75 With regard to posology, both a frequent use in the therapy of AdHF and an earlier use of levosimendan in the therapy of AHF have been shown to be of benefit.91,223 Beyond that, 20 years after its initial approval for clinical use, levosimendan remains an important resource in cardiovascular medicine and a valuable tool for clinical research, investigation and innovation in that and other areas of medicine.20 The clinical programme for the development of oral levosimendan as a treatment for ALS shows how retargeting a safe drug, even in a different formulation, is a rational strategy in pharmaceutical development.224

Click here to view supplementary material.

References

  1. Beregovich J, Bianchi C, D’Angelo R, et al. Haemodynamic effects of a new inotropic agent (dobutamine) in chronic cardiac failure. Br Heart J 1975;37:629–34.
    Crossref | PubMed
  2. Fabiato A, Fabiato F. Calcium and cardiac excitation-contraction coupling. Ann Rev Physiol 1979;41:473–84.
    Crossref | PubMed
  3. Wohlfart B, Noble MI. The cardiac excitation-contraction cycle. Pharmacol Ther 1982;16:1–43.
    Crossref | PubMed
  4. Colucci WS, Wright RF, Braunwald E. New positive inotropic agents in the treatment of congestive heart failure. Mechanisms of action and recent clinical developments. 1. N Engl J Med 1986;314:290–9.
    Crossref | PubMed
  5. Maskin CS, Sinoway L, Chadwick B, et al. Sustained hemodynamic and clinical effects of a new cardiotonic agent, WIN 47203, in patients with severe congestive heart failure. Circulation 1983;67:1065–70.
    Crossref | PubMed
  6. Dage RC, Roebel LE, Hsieh CP, et al. Cardiovascular properties of a new cardiotonic agent: MDL 17,043 (1.3-dihydro-4-methyl-5-[4-(methylthio)-benzoyl]-2H-imidazol-2-one). J Cardiovasc Pharmacol 1982;4:500–8.
    PubMed
  7. Nagy L, Pollesello P, Papp Z. Inotropes and inodilators for acute heart failure: sarcomere active drugs in focus. J Cardiovasc Pharmacol 2014;64:199–208.
    Crossref | PubMed
  8. Herzig JW, Feile K, Rüegg JC. Activating effects of AR-L 115 BS on the Ca2+ sensitive force, stiffness and unloaded shortening velocity (Vmax) in isolated contractile structures from mammalian heart muscle. Arzneimittelforschung 1981;31:188–91.
    PubMed
  9. Solaro RJ, Rüegg JC. Stimulation of Ca++ binding and ATPase activity of dog cardiac myofibrils by AR-L 115BS, a novel cardiotonic agent. Circ Res 1982;51:290–4.
    PubMed
  10. Rüegg JC, Pfitzer G, Eubler D, et al. Effect on contractility of skinned fibres from mammalian heart and smooth muscle by a new benzimidazole derivative, 4,5-dihydro-6-[2-(4-methoxyphenyl)-1H-benzimidazol-5-yl]-5-methyl-3(2H)-pyridazinone. Arzneimittelforschung 1984;34:1736–8.
    PubMed
  11. Raasmaja A, Talo A, Haikala H, et al. Biochemical properties of OR-1259: a positive inotropic and vasodilatory compound with an antiarrhythmic effect. Adv Exp Med Biol 1992;311:423.
    Crossref | PubMed
  12. Haikala H, Kaivola J, Nissinen E, et al. Cardiac troponin C as a target protein for a novel calcium sensitizing drug, levosimendan. J Mol Cell Cardiol 1995;27:1859–66.
    Crossref | PubMed
  13. Pollesello P, Ovaska M, Kaivola J, et al. Binding of a new Ca2+ sensitizer, levosimendan, to recombinant human cardiac troponin C. A molecular modelling, fluorescence probe, and proton nuclear magnetic resonance study. J Biol Chem 1994;269:28584–90.
    PubMed
  14. Pääkkönen K, Annila A, Sorsa T, et al. Solution structure and main chain dynamics of the regulatory domain (residues 1-91) of human cardiac troponin C. J Biol Chem 1998;273:15633–8.
    Crossref | PubMed
  15. Haikala H, Linden IB. Mechanisms of action of calcium-sensitizing drugs. J Cardiovasc Pharmacol 1995;26(Suppl 1):S10–9.
    PubMed
  16. Sorsa T, Pollesello P, Solaro RJ. The contractile apparatus as a target for drugs against heart failure: interaction of levosimendan, a calcium sensitiser, with cardiac troponin C. Mol Cell Biochem 2004;266:87–107.
    Crossref | PubMed
  17. Haikala H, Pollesello P. Calcium sensitivity enhancers. iDrugs 2000;3:1199–205.
    PubMed
  18. Sorsa T, Heikkinen S, Abbott MB, et al. Binding of levosimendan, a calcium sensitizer, to cardiac troponin C. J Biol Chem 2001;276:9337–43.
    Crossref | PubMed
  19. Levijoki J, Pollesello P, Kaivola J, et al. Further evidence for the cardiac troponin C mediated calcium sensitization by levosimendan: structure-response and binding analysis with analogs of levosimendan. J Mol Cell Cardiol 2000;32:479–91.
    Crossref | PubMed
  20. Pineda-Sanabria SE, Robertson IM, Sun YB, et al. Probing the mechanism of cardiovascular drugs using a covalent levosimendan analog. J Mol Cell Cardiol 2016;92:174–84.
    Crossref | PubMed
  21. Lindert S, Li MX, Sykes BD, et al. Computer-aided drug discovery approach finds calcium sensitizer of cardiac troponin. Chem Biol Drug Des 2015;85:99–106.
    Crossref | PubMed
  22. Robertson IM, Sun YB, Li MX, et al. A structural and functional perspective into the mechanism of Ca2+-sensitizers that target the cardiac troponin complex. J Mol Cell Cardiol 2010;49:1031–41.
    Crossref | PubMed
  23. Yokoshiki H, Katsube Y, Sunagawa M, et al. Levosimendan, a novel Ca2+ sensitizer, activates the glibenclamide-sensitive K+ channel in rat arterial myocytes. Eur J Pharmacol 1997;333:249–59.
    Crossref | PubMed
  24. Pagel PS, Hettrick DA, Warltier DC. Influence of levosimendan, pimobendan, and milrinone on the regional distribution of cardiac output in anaesthetized dogs. Br J Pharmacol 1996;119:609–15.
    Crossref | PubMed
  25. Kopustinskiene DM, Pollesello P, Saris NE. Potassium-specific effects of levosimendan on heart mitochondria. Biochem Pharmacol 2004;68:807–12.
    Crossref | PubMed
  26. Kopustinskiene DM, Pollesello P, Saris NE. Levosimendan is a mitochondrial K(ATP) channel opener. Eur J Pharmacol 2001;428:311–4.
    Crossref | PubMed
  27. Maytin M, Colucci WS. Cardioprotection: a new paradigm in the management of acute heart failure syndromes. Am J Cardiol 2005;96:26G–31G.
    Crossref | PubMed
  28. Nieminen MS, Pollesello P, Vajda G, et al. Effects of levosimendan on the energy balance: preclinical and clinical evidence. J Cardiovasc Pharmacol 2009;53:302–10.
    Crossref | PubMed
  29. Metzsch C, Linnér R, Steen S, et al. Levosimendan cardioprotection in acutely beta-1 adrenergic receptor blocked open chest pigs. Acta Anaesthesiol Scand 2010;54:103–10.
    Crossref | PubMed
  30. Papp JG, Pollesello P, Varró AF, et al. Effect of levosimendan and milrinone on regional myocardial ischemia/reperfusion-induced arrhythmias in dogs. J Cardiovasc Pharmacol Ther 2006;11:129–35.
    Crossref | PubMed
  31. du Toit EF, Genis A, Opie LH, et al. A role for the RISK pathway and K(ATP) channels in pre- and post-conditioning induced by levosimendan in the isolated guinea pig heart. Br J Pharmacol 2008;154:41–50.
    Crossref | PubMed
  32. Tritapepe L, De Santis V, Vitale D, et al. Levosimendan pre-treatment improves outcomes in patients undergoing coronary artery bypass graft surgery. Br J Anaesth 2009;102:198–204.
    Crossref | PubMed
  33. Takahashi R, Talukder MA, Endoh M. Inotropic effects of OR-1896, an active metabolite of levosimendan, on canine ventricular myocardium. Eur J Pharmacol 2000;400:103–12.
    Crossref | PubMed
  34. Takahashi R, Talukder MA, Endoh M. Effects of OR-1896, an active metabolite of levosimendan, on contractile force and aequorin light transients in intact rabbit ventricular myocardium. J Cardiovasc Pharmacol 2000;36:118–25.
    Crossref | PubMed
  35. Erdei N, Papp Z, Pollesello P, et al. The levosimendan metabolite OR-1896 elicits vasodilation by activating the K(ATP) and BK(Ca) channels in rat isolated arterioles. Br J Pharmacol 2006;148:696–702.
    Crossref | PubMed
  36. Kivikko M, Antila S, Eha J, et al. Pharmacokinetics of levosimendan and its metabolites during and after a 24-hour continuous infusion in patients with severe heart failure. Int J Clin Pharm Ther 2002;40:465–71.
    Crossref | PubMed
  37. Farmakis D, Alvarez J, Gal TB, et al. Levosimendan beyond inotropy and acute heart failure: evidence of pleiotropic effects on the heart and other organs: an expert panel position paper. Int J Cardiol 2016;222:303–12.
    Crossref | PubMed
  38. Szilágyi S, Pollesello P, Levijoki J, et al. The effects of levosimendan and OR-1896 on isolated hearts, myocyte-sized preparations and phosphodiesterase enzymes of the guinea pig. Eur J Pharmacol 2004;486:67–74.
    Crossref | PubMed
  39. de Cheffoy de Courcelles D, de Loore K, Freyne E, Janssen PA. Inhibition of human cardiac cyclic AMP-phosphodiesterases by R 80122, a new selective cyclic AMP-phosphodiesterase III inhibitor: a comparison with other cardiotonic compounds. J Pharmacol Exp Ther 1992;263:6–14.
    PubMed
  40. Szilagyi S, Pollesello P, Levijoki J, et al. Two inotropes with different mechanisms of action: contractile, PDE-inhibitory and direct myofibrillar effects of levosimendan and enoximone. J Cardiovasc Pharmacol 2005;46:369–76.
    Crossref | PubMed
  41. Hasenfuss G, Pieske B, Castell M, et al. Influence of the novel inotropic agent levosimendan on isometric tension and calcium cycling in failing human myocardium. Circulation 1998;98:2141–7.
    Crossref | PubMed
  42. Kaheinen P, Pollesello P, Levijoki J, et al. Effects of levosimendan and milrinone on oxygen consumption in isolated guinea-pig heart. J Cardiovasc Pharmacol 2004;43:555–61.
    Crossref | PubMed
  43. Maack C, Eschenhagen T, Hamdani N, et al. Treatments targeting inotropy: a position paper of the Committees on Translational Research and Acute Heart Failure of the Heart Failure Association of the European Society of Cardiology. Eur Heart J 2019;40:3626–44.
    Crossref | PubMed
  44. Xanthos T, Bassiakou E, Koudouna E, et al. Combination pharmacotherapy in the treatment of experimental cardiac arrest. Am J Emerg Med 2009;27:651–9.
    Crossref | PubMed
  45. Lochner A, Colesky F, Genade S. Effect of a calcium-sensitizing agent, levosimendan, on the postcardioplegic inotropic response of the myocardium. Cardiovasc Drugs Ther 2000;14:271–81.
    Crossref | PubMed
  46. Antila S, Eha J, Heinpalu M, et al. Haemodynamic interactions of a new calcium sensitizing drug levosimendan and captopril. Eur J Clin Pharmacol 1996;49:451–8.
    Crossref | PubMed
  47. Sundberg S, Lilleberg J, Nieminen MS, et al. Hemodynamic and neurohumoral effects of levosimendan, a new calcium sensitizer, at rest and during exercise in healthy men. Am J Cardiol 1995;75:1061–6.
    Crossref | PubMed
  48. Lilleberg J, Sundberg S, Nieminen MS. Dose-range study of a new calcium sensitizer, levosimendan, in patients with left ventricular dysfunction. J Cardiovasc Pharmacol 1995;26(Suppl 1):S63–9.
    PubMed
  49. Ukkonen H, Saraste M, Akkila J, et al. Myocardial efficiency during levosimendan infusion in congestive heart failure. Clin Pharmacol Ther 2000;68:522–31.
    Crossref | PubMed
  50. Lilleberg J, Nieminen MS, Akkila J, et al. Effects of a new calcium sensitizer, levosimendan, on haemodynamics, coronary blood flow and myocardial substrate utilization early after coronary artery bypass grafting. Eur Heart J 1998;19:660–8.
    Crossref | PubMed
  51. Ukkonen H, Saraste M, Akkila J, et al. Myocardial efficiency during calcium sensitization with levosimendan: a noninvasive study with positron emission tomography and echocardiography in healthy volunteers. Clin Pharmacol Ther 1997;61:596–607.
    Crossref | PubMed
  52. Nieminen MS, Akkila J, Hasenfuss G, et al. Hemodynamic and neurohumoral effects of continuous infusion of levosimendan in patients with congestive heart failure. J Am Coll Cardiol 2000;36:1903–12.
    Crossref | PubMed
  53. Jörgensen K, Bech-Hanssen O, Houltz E, et al. Effects of levosimendan on left ventricular relaxation and early filling at maintained preload and afterload conditions after aortic valve replacement for aortic stenosis. Circulation 2008;117:1075–81.
    Crossref | PubMed
  54. Fredholm M, Jörgensen K, Houltz E, et al. Inotropic and lusitropic effects of levosimendan and milrinone assessed by strain echocardiography: a randomised trial. Acta Anaesthesiol Scand 2018;62:1246–54.
    Crossref | PubMed
  55. Parissis JT, Paraskevaidis I, Bistola V, et al. Effects of levosimendan on right ventricular function in patients with advanced heart failure. Am J Cardiol 2006;98:1489–92.
    Crossref | PubMed
  56. Yilmaz MB, Yontar C, Erdem A, et al. Comparative effects of levosimendan and dobutamine on right ventricular function in patients with biventricular heart failure. Heart Vessels 2009;24:16–21.
    Crossref | PubMed
  57. Russ MA, Prondzinsky R, Carter JM, et al. Right ventricular function in myocardial infarction complicated by cardiogenic shock: improvement with levosimendan. Crit Care Med 2009;37:3017–23.
    Crossref | PubMed
  58. Lilleberg J, Laine M, Palkama T, et al. Duration of the haemodynamic action of a 24-h infusion of levosimendan in patients with congestive heart failure. Eur J Heart Fail 2007;9:75–82.
    Crossref | PubMed
  59. Kivikko M, Lehtonen L, Colucci WS. Sustained hemodynamic effects of intravenous levosimendan. Circulation 2003;107:81–6.
    Crossref | PubMed
  60. Antila S, Kivikko M, Lehtonen L, et al. Pharmacokinetics of levosimendan and its circulating metabolites in patients with heart failure after an extended continuous infusion of levosimendan. Br J Clin Pharmacol 2004;57:412–5.
    Crossref | PubMed
  61. Mebazaa A, Nieminen MS, Packer M, et al. Levosimendan vs dobutamine for patients with acute decompensated heart failure: the SURVIVE randomized trial. JAMA 2007;297:1883–91.
    Crossref | PubMed
  62. Packer M, Colucci W, Fisher L, et al. Effect of levosimendan on the short-term clinical course of patients with acutely decompensated heart failure. JACC Heart Fail 2013;1:103–11.
    Crossref | PubMed
  63. Follath F, Cleland JG, Just H, et al. Efficacy and safety of intravenous levosimendan, a novel calcium sensitiser, in severe low output heart failure: results of a randomised, double-blind comparison with dobutamine (LIDO Study). Lancet 2002;360:196–202.
    Crossref | PubMed
  64. Slawsky MT, Colucci WS, Gottlieb SS, et al. Acute hemodynamic and clinical effects of levosimendan in patients with severe heart failure. Circulation 2000;102:2222–7.
    Crossref | PubMed
  65. Moiseyev VS, Põder P, Andrejevs N, et al. Safety and efficacy of a novel calcium sensitiser, levosimendan, in patients with left ventricular failure due to an acute myocardial infarction: a randomized, placebo-controlled, double-blind study (RUSSLAN). Eur Heart J 2002;23:1422–32.
    Crossref | PubMed
  66. Mebazaa A, Nieminen MS, Filippatos GS, et al. Levosimendan vs. dobutamine: outcomes for acute heart failure patients on beta-blockers in SURVIVE. Eur J Heart Fail 2009;11:304–11.
    Crossref | PubMed
  67. Landoni G, Biondi-Zoccai G, Greco M, et al. Effects of levosimendan on mortality and hospitalization. A meta-analysis of randomized controlled studies. Crit Care Med 2012;40:634–46.
    Crossref | PubMed
  68. Sikora J, Pstragowski K, Skibinska N, et al. Impact of levosimendan on platelet function. Thromb Res 2017;159:76–81.
    Crossref | PubMed
  69. Yan SB, Wang XY, Shang GK, et al. Impact of perioperative levosimendan administration on risk of bleeding after cardiac surgery: a meta-analysis of randomized controlled trials. Am J Cardiovasc Drugs 2020;20:149–60.
    Crossref | PubMed
  70. Mehta RH, Leimberger JD, van Diepen S, et al. LEVO-CTS Investigators. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery. N Engl J Med 2017;376:2032–42.
    Crossref | PubMed
  71. Gong B, Li Z, Yat Wong PC. Levosimendan treatment for heart failure: a systematic review and meta-analysis. J Cardiothorac Vasc Anesth 2015;29:1415–25.
    Crossref | PubMed
  72. SIMDAX. Summary of Product Characteristics. https://www.simdax.com/siteassets/simdax-spc.pdf (accessed 23 April 2020).
  73. Nieminen MS, Buerke M, Cohen-Solál A, et al. The role of levosimendan in acute heart failure complicating acute coronary syndrome: a review and expert consensus opinion. Int J Cardiol 2016;218:150–7.
    Crossref | PubMed
  74. Harjola VP, Giannakoulas G, von Lewinski D, et al. Use of levosimendan in acute heart failure. Eur Heart J Suppl 2018;20(Suppl I):I2–10.
    Crossref | PubMed
  75. Pollesello P, Ben Gal T, Bettex D, et al. Short-term therapies for treatment of acute and advanced heart failure: why so few drugs available in clinical use, why even fewer in the pipeline? J Clin Med 2019;8:e1834.
    Crossref | PubMed
  76. Gheorghiade M, Adams KF, Cleland JG, et al. Phase III clinical trial end points in acute heart failure syndromes: a virtual roundtable with the Acute Heart Failure Syndromes International Working Group. Am Heart J 2009;157:957–70.
    Crossref | PubMed
  77. Zannad F, Garcia AA, Anker SD, et al. Clinical outcome endpoints in heart failure trials: a European Society of Cardiology Heart Failure Association consensus document. Eur J Heart Fail 2013;15:1082–94.
    Crossref | PubMed
  78. Tacon CL, McCaffrey J, Delaney A. Dobutamine for patients with severe heart failure: a systematic review and meta-analysis of randomised controlled trials. Intensive Care Med 2012;38:359–67.
    Crossref | PubMed
  79. Nony P, Boissel JP, Lievre M, et al. Evaluation of the effect of phosphodiesterase inhibitors on mortality in chronic heart failure patients. A meta-analysis. Eur J Clin Pharmacol 1994;46:191–6.
    Crossref | PubMed
  80. Pollesello P, Parissis J, Kivikko M, et al. Levosimendan meta-analyses: is there a pattern in the effect on mortality? Int J Cardiol 2016;209:77–83.
    Crossref | PubMed
  81. Mebazaa A, Parissis J, Porcher R, et al. Short-term survival by treatment among patients hospitalized with acute heart failure: the global ALARM-HF registry using propensity scoring methods. Intensive Care Med 2011;37:290–301.
    Crossref | PubMed
  82. Nieminen MS, Böhm M, Cowie MR, et al. Executive summary of the guidelines on the diagnosis and treatment of acute heart failure: the Task Force on Acute Heart Failure of the European Society of Cardiology. Eur Heart J 2005;26:384–416.
    Crossref | PubMed
  83. Dickstein K, Cohen-Solal A, Filippatos G, et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2008: the Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2008 of the European Society of Cardiology. Eur Heart J 2008;29:2388–442.
    Crossref | PubMed
  84. McMurray JJ, Adamopoulos S, Anker SD, et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: the Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Eur Heart J 2012;33:1787–847.
    Crossref | PubMed
  85. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 2016;37:2129–200.
    Crossref | PubMed
  86. Farmakis D, Agostoni P, Baholli L, et al. A pragmatic approach to the use of inotropes for the management of acute and advanced heart failure: an expert panel consensus. Int J Cardiol 2019;297:83–90.
    Crossref | PubMed
  87. Agostoni P, Farmakis DT, Garcia-Pinilla JM, et al. Hemodynamic balance in acute and advanced heart failure: an expert perspective on the role of levosimendan. Card Fail Rev 2019;5:155–61.
    Crossref | PubMed
  88. Bouchez S, Fedele F, Giannakoulas G, et al. Levosimendan in acute and advanced heart failure: an expert perspective on posology and therapeutic application. Cardiovasc Drugs Ther 2018;32:617–24.
    Crossref | PubMed
  89. Oliva F, Comin-Colet J, Fedele F, et al. Repetitive levosimendan treatment in the management of advanced heart failure. Eur Heart J Suppl 2018;20(Suppl I):I11–20.
    Crossref | PubMed
  90. Delgado JF, Oliva F, Reinecke A. The inodilator levosimendan in repetitive doses in the treatment of advanced heart failure. Eur Heart J Suppl 2017;19(Suppl C):C8–14.
    Crossref | PubMed
  91. Pölzl G, Altenberger J, Baholli L, et al. Repetitive use of levosimendan in advanced heart failure: need for stronger evidence in a field in dire need of a useful therapy. Int J Cardiol 2017;243:389–39.
    Crossref | PubMed
  92. Nieminen MS, Altenberger J, Ben-Gal T, et al. Repetitive use of levosimendan for treatment of chronic advanced heart failure: clinical evidence, practical considerations, and perspectives: an expert panel consensus. Int J Cardiol 2014;174:360–7.
    Crossref | PubMed
  93. Toller W, Algotsson L, Guarracino F, et al. Perioperative use of levosimendan: best practice in operative settings. J Cardiothorac Vasc Anesth 2013;27:361–6.
    Crossref | PubMed
  94. Toller W, Heringlake M, Guarracino F, et al. Preoperative and perioperative use of levosimendan in cardiac surgery: European expert opinion. Int J Cardiol 2015;184:323–36.
    Crossref | PubMed
  95. Shi WY, Li S, Collins N, Cottee DB, et al. Peri-operative levosimendan in patients undergoing cardiac surgery: an overview of the evidence. Heart Lung Circ 2015;24:667–72.
    Crossref | PubMed
  96. Herpain A, Bouchez S, Girardis M, et al. Use of levosimendan in intensive care unit settings: an opinion paper. J Cardiovasc Pharmacol 2019;73:3–14.
    Crossref | PubMed
  97. Yilmaz MB, Grossini E, Silva Cardoso JC, et al. Renal effects of levosimendan: a consensus report. Cardiovasc Drugs Ther 2013;27:581–90.
    Crossref | PubMed
  98. Nieminen MS, Dickstein K, Fonseca C, et al. The patient perspective: quality of life in advanced heart failure with frequent hospitalisations. Int J Cardiol 2015;191:256–64.
    Crossref | PubMed
  99. Nieminen MS, Fonseca C, Brito D, et al. The potential of the inodilator levosimendan in maintaining quality of life in advanced heart failure. Eur Heart J Suppl 2017;19(Suppl C):C15–21.
    Crossref | PubMed
  100. Mushtaq S, Andreini D, Farina S, et al. Levosimendan improves exercise performance in patients with advanced chronic heart failure. ESC Heart Fail 2015;2:133–41.
    Crossref | PubMed
  101. Campodonico J, Mapelli M, Spadafora E, et al. Surfactant proteins changes after acute hemodynamic improvement in patients with advanced chronic heart failure treated with Levosimendan. Respir Physiol Neurobiol 2018;252–253:47–51.
    Crossref | PubMed
  102. Nieminen MS, Buerke M, Parissis J, et al. Pharmaco-economics of levosimendan in cardiology: a European perspective. Int J Cardiol 2015;199:337–41.
    Crossref | PubMed
  103. Papp Z, Édes I, Fruhwald S, et al. Levosimendan: molecular mechanisms and clinical implications: consensus of experts on the mechanisms of action of levosimendan. Int J Cardiol 2012;159:82–7.
    Crossref | PubMed
  104. Pollesello P, Papp Z, Papp JG. Calcium sensitizers: what have we learned over the last 25 years? Int J Cardiol 2016;203:543–8.
    Crossref | PubMed
  105. Bistola V, Arfaras-Melainis A, Polyzogopoulou E, et al. Inotropes in acute heart failure: from guidelines to practical use: therapeutic options and clinical practice. Card Fail Rev 2019;5:133–9.
    Crossref | PubMed
  106. Fuhrmann JT, Schmeisser A, Schulze MR, et al. Levosimendan is superior to enoximone in refractory cardiogenic shock complicating acute myocardial infarction. Crit Care Med 2008; 36:2257–66.
    Crossref | PubMed
  107. García-Gonzáles MJ, Domínguez-Rodríguez A, Ferrer-Hita JJ, et al. Cardiogenic shock after primary percutaneous coronary intervention: effects of levosimendan compared with dobutamine on haemodynamics. Eur J Heart Fail 2006;8:723–8.
    Crossref | PubMed
  108. Dominguez-Rodriguez A, Samimi-Fard S, Garcia-Gonzalez MJ, et al. Effects of levosimendan versus dobutamine on left ventricular diastolic function in patients with cardiogenic shock after primary angioplasty. Int J Cardiol 2008;128:214–17.
    Crossref | PubMed
  109. Samimi-Fard S, García-González MJ, Domínguez-Rodríguez A, et al. Effects of levosimendan versus dobutamine on long-term survival of patients with cardiogenic shock after primary coronary angioplasty. Int J Cardiol 2008; 127:284–7.
    Crossref | PubMed
  110. Christoph A, Prondzinsky R, Russ M, et al. Early and sustained haemodynamic improvement with levosimendan compared to intraaortic balloon counterpulsation (IABP) in cardiogenic shock complicating acute myocardial infarction. Acute Card Care 2008;10:49–57.
    Crossref | PubMed
  111. Omerovic E, Råmunddal T, Albertsson P, et al. Levosimendan neither improves nor worsens mortality in patients with cardiogenic shock due to ST-elevation myocardial infarction. Vasc Health Risk Manag 2010;6:657–63.
    Crossref | PubMed
  112. Guarracino F, Cariello C, Danella A, et al. Effect of levosimendan on ventriculo-arterial coupling in patients with ischemic cardiomyopathy. Acta Anaesthesiol Scand 2007;51:1217–24.
    Crossref | PubMed
  113. Guarracino F, Baldassarri R, Pinsky MR. Ventriculo-arterial decoupling in acutely altered hemodynamic states. Crit Care 2013;17:213.
    Crossref | PubMed
  114. Hering D, Jaguszewski M. Levosimendan: new hope therapy for takotsubo syndrome. Cardiol J 2016;23:616–17.
    Crossref | PubMed
  115. De Santis V, Vitale D, Tritapepe L, et al. Use of levosimendan for cardiogenic shock in a patient with the apical ballooning syndrome. Ann Intern Med 2008;149:365–7.
    Crossref | PubMed
  116. Paur H, Wright PT, Sikkel MB, et al. High levels of circulating epinephrine trigger apical cardiodepression in a β2-adrenergic receptor/Gi-dependent manner: a new model of Takotsubo cardiomyopathy. Circulation 2012;126:697–706.
    Crossref | PubMed
  117. Lyon AR, Bossone E, Schneider B, et al. Current state of knowledge on Takotsubo syndrome: a position statement from the Taskforce on Takotsubo Syndrome of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2016;18:8–27.
    Crossref | PubMed
  118. Harrison RW, Hasselblad V, Mehta RH, et al. Effect of levosimendan on survival and adverse events after cardiac surgery: a meta-analysis. J Cardiothorac Vasc Anesth 2013;27:1224–32.
    Crossref | PubMed
  119. Cholley B, Caruba T, Grosjean S, et al. Effect of levosimendan on low cardiac output syndrome in patients with low ejection fraction undergoing coronary artery bypass grafting with cardiopulmonary bypass: the LICORN Randomized Clinical Trial. JAMA 2017;318:548–56.
    Crossref | PubMed
  120. Landoni G, Lomivorotov VV, Alvaro G, et al. Levosimendan for hemodynamic support after cardiac surgery. N Engl J Med 2017;376:2021–31.
    Crossref | PubMed
  121. Lee CT, Lin YC, Yeh YC, et al. Effects of levosimendan for perioperative cardiovascular dysfunction in patients receiving cardiac surgery: a meta-analysis with trial sequential analysis. Intensive Care Med 2017;43:1929–30.
    Crossref | PubMed
  122. Guarracino F, Heringlake M, Cholley B, et al. Use of levosimendan in cardiac surgery: an update after the LEVO-CTS, CHEETAH, and LICORN trials in the light of clinical practice. J Cardiovasc Pharmacol 2018;71:1–9.
    Crossref | PubMed
  123. Wang W, Zhou X, Liao X, et al. The efficacy and safety of prophylactic use of levosimendan on patients undergoing coronary artery bypass graft: a systematic review and meta-analysis. J Anesth 2019;33:543–50.
    Crossref | PubMed
  124. van Diepen S, Mehta RH, Leimberger JD, et al. Levosimendan in patients with reduced left ventricular function undergoing isolated coronary or valve surgery. J Thorac Cardiovasc Surg 2019; epub ahead of press.
    Crossref | PubMed
  125. Weber C, Esser M, Eghbalzadeh K, et al. Levosimendan reduces mortality and low cardiac output syndrome in cardiac surgery. Thorac Cardiovasc Surg 2019; epub ahead of press.
    Crossref | PubMed
  126. Qiu J, Jia L, Hao Y, et al. Efficacy and safety of levosimendan in patients with acute right heart failure: a meta-analysis. Life Sci 2017;184:30–6.
    Crossref | PubMed
  127. Fedele F, Severino P, Calcagno S, et al. Heart failure: TNM-like classification. J Am Coll Cardiol 2014;63:1959–60.
    Crossref | PubMed
  128. Zager RA, Johnson AC, Lund S, et al. Levosimendan protects against experimental endotoxemic acute renal failure. Am J Physiol Renal Physiol 2006;290:F1453–62.
    Crossref | PubMed
  129. Rehberg S, Ertmer C, Vincent JL, et al. Effects of combined arginine vasopressin and levosimendan on organ function in ovine septic shock. Crit Care Med 2010;38:2016–23.
    Crossref | PubMed
  130. Grossini E, Molinari C, Pollesello P, et al. Levosimendan protection against kidney ischemia/reperfusion injuries in anesthetized pigs. J Pharmacol Exp Ther 2012;342:376–88.
    Crossref | PubMed
  131. Fedele F, Bruno N, Brasolin B, et al. Levosimendan improves renal function in acute decompensated heart failure: possible underlying mechanisms. Eur J Heart Fail 2014;16:281–8.
    Crossref | PubMed
  132. Lannemyr L, Ricksten S-E, Rundqvist B, et al. Differential effects of levosimendan and dobutamine on glomerular filtration rate in patients with heart failure and renal impairment: a randomized double-blind controlled trial. J Am Heart Assoc 2018;7:e008455.
    Crossref | PubMed
  133. Bragadottir G, Redfors B, Ricksten SE. Effects of levosimendan on glomerular filtration rate, renal blood flow, and renal oxygenation after cardiac surgery with cardiopulmonary bypass: a randomized placebo-controlled study. Crit Care
    Med 2013;41:2328–35.
    Crossref | PubMed
  134. Lannemyr L, Bragadottir G, Redfors B, et al. Effects of milrinone on renal perfusion, filtration and oxygenation in patients with acute heart failure and low cardiac output early after cardiac surgery. J Crit Care 2020;57:225–30.
    Crossref | PubMed
  135. Sanfilippo F, Knight JB, Scolletta S, et al. Levosimendan for patients with severely reduced left ventricular systolic function and/or low cardiac output syndrome undergoing cardiac surgery: a systematic review and meta-analysis. Crit Care 2017;21:252.
    Crossref | PubMed
  136. Putzu A, Clivio S, Belletti A, et al. Perioperative levosimendan in cardiac surgery: a systematic review with meta-analysis and trial sequential analysis. Int J Cardiol 2018;251:22–31.
    Crossref | PubMed
  137. Niu ZZ, Wu SM, Sun WY, et al. Perioperative levosimendan therapy is associated with a lower incidence of acute kidney injury after cardiac surgery: a meta-analysis. J Cardiovasc Pharmacol 2014;63:107–12.
    Crossref | PubMed
  138. Bove T, Matteazzi A, Belletti A, et al. Beneficial impact of levosimendan in critically ill patients with or at risk for acute renal failure: a meta-analysis of randomized clinical trials. Heart Lung Vessel 2015;7:35–46.
    PubMed
  139. Solomon SD, Dobson J, Pocock S, et al. Influence of nonfatal hospitalization for heart failure on subsequent mortality in patients with chronic heart failure. Circulation 2007;116:1482–7.
    Crossref | PubMed
  140. Setoguchi S, Stevenson LW, Schneeweiss S. Repeated hospitalizations predict mortality in the community population with heart failure. Am Heart J 2007;154:260–6.
    Crossref | PubMed
  141. Altenberger J, Parissis JT, Costard-Jaeckle A, et al. Efficacy and safety of the pulsed infusions of levosimendan in outpatients with advanced heart failure (LevoRep) study: a multicentre randomized trial. Eur J Heart Fail 2014;16:898–906.
    Crossref | PubMed
  142. Comín-Colet J, Manito N, Segovia-Cubero J, et al. Efficacy and safety of intermittent intravenous outpatient administration of levosimendan in patients with advanced heart failure: the LION-HEART multicentre randomised trial. Eur J Heart Fail 2018;20:1128–36.
    Crossref | PubMed
  143. LAICA Study Investigators. Efficacy and security of intermittent repeated levosimendan administration in patients with advanced heart failure: a randomized, double-blind, placebo controlled multicenter trial: LAICA study. Presented at the European Society of Cardiology–Heart Failure Association Congress, Florence, Italy, 21 May 2016.
  144. Silvetti S, Nieminen MS. Repeated or intermittent levosimendan treatment in advanced heart failure: an updated meta-analysis. Int J Cardiol 2016;202:138–43.
    Crossref | PubMed
  145. Silvetti S, Greco T, Di Prima AL, et al. Intermittent levosimendan improves mid-term survival in chronic heart failure patients: meta-analysis of randomised trials. Clin Res Cardiol 2014;103:505–13.
    Crossref | PubMed
  146. Pölzl G, Allipour Birgani S, Comín-Colet J, et al. Repetitive levosimendan infusions for patients with advanced chronic heart failure in the vulnerable post-discharge period. ESC Heart Fail 2019;6:174–81.
    Crossref | PubMed
  147. Kocabeyoglu SS, Kervan U, Sert DE, et al. Optimization with levosimendan improves outcomes after left ventricular assist device implantation. Eur J Cardiothorac Surg 2020;57:176–82.
    Crossref | PubMed
  148. Sim I. Mobile devices and health. N Engl J Med 2019;381:956–68.
    Crossref | PubMed
  149. Andrès, E, Talha S, Zulfiqar A.-A., et al. Current research and new perspectives of telemedicine in chronic heart failure: narrative review and points of interest for the clinician. J Clin Med 2018;7:544.
    Crossref | PubMed
  150. Wells R, Stockdill ML, Dionne-Odom JN, et al. Educate, Nurture, Advise, Before Life Ends Comprehensive Heartcare for Patients and Caregivers (ENABLE CHF-PC): study protocol for a randomized controlled trial. Trials 2018;19:422.
    Crossref | PubMed
  151. Schumann J, Henrich EC, Strobl H, et al. Inotropic agents and vasodilator strategies for the treatment of cardiogenic shock or low cardiac output syndrome. Cochrane Database Syst Rev 2018;(1):CD009669.
    Crossref | PubMed
  152. Morelli A, De Castro S, Teboul JL, et al. Effects of levosimendan on systemic and regional hemodynamics in septic myocardial depression. Intensive Care Med 2005;31:638–44.
    Crossref | PubMed
  153. Zangrillo A, Putzu A, Monaco F, et al. Levosimendan reduces mortality in patients with severe sepsis and septic shock: a meta-analysis of randomized trials. J Crit Care 2015; 30:908–13.
    Crossref | PubMed
  154. Ouanes-Besbes L, Ouanes I, Dachraoui F, et al. Weaning difficult-to-wean chronic obstructive pulmonary disease patients: a pilot study comparing initial hemodynamic effects of levosimendan and dobutamine. J Crit Care 2011;26:15–21.
    Crossref | PubMed
  155. Aso S, Matsui H, Fushimi K, et al. In-hospital mortality and successful weaning from venoarterial extracorporeal membrane oxygenation: analysis of 5,263 patients using a national inpatient database in Japan. Crit Care 2016;20:80.
    Crossref | PubMed
  156. Affronti A, di Bella I, Carino D, et al. Levosimendan may improve weaning outcomes in venoarterial ECMO patients. ASAIO J 2013;59:554–7.
    Crossref | PubMed
  157. Distelmaier K, Roth C, Schrutka L, et al. Beneficial effects of levosimendan on survival in patients undergoing extracorporeal membrane oxygenation after cardiovascular surgery. Br J Anaesth 2016;117:52–8.
    Crossref | PubMed
  158. Sangalli F, Avalli L, Laratta M, et al. Effects of levosimendan on endothelial function and hemodynamics during weaning from veno-arterial extracorporeal life support. J Cardiothorac Vasc Anesth 2016;30:1449–53.
    Crossref | PubMed
  159. Jacky A, Rudiger A, Krüger B, et al. Comparison of levosimendan and milrinone for ECLS weaning in patients after cardiac surgery – a retrospective before and after study. J Cardiothorac Vasc Anesth 2018;32:2112–19.
    Crossref | PubMed
  160. Yilmaz MB, Yalta K, Yontar C, et al. Levosimendan improves renal function in patients with acute decompensated heart failure: comparison with dobutamine. Cardiovasc Drugs Ther 2007;21:431–5.
    Crossref | PubMed
  161. Hou ZQ, Sun ZX, Su CY, et al. Effect of levosimendan on estimated glomerular filtration rate in hospitalized patients with decompensated heart failure and renal dysfunction. Cardiovasc Ther 2013;31:108–14.
    Crossref | PubMed
  162. Zemljic G, Bunc M, Yazdanbakhsh AP, et al. Levosimendan improves renal function in patients with advanced chronic heart failure awaiting cardiac transplantation. J Card Fail 2007;13:417–21.
    Crossref | PubMed
  163. Silva-Cardoso J, Ferreira J, Oliveira-Soares A, et al. Effectiveness and safety of levosimendan in clinical practice. Rev Port Cardiol 2009;28:143–53.
    PubMed
  164. Zangrillo A, Alvaro G, Belletti A, et al. Effect of levosimendan on renal outcome in cardiac surgery patients with chronic kidney disease and perioperative cardiovascular dysfunction: a substudy of a multicenter randomized trial. J Cardiothorac Vasc Anesth 2018;32:2152–9.
    Crossref | PubMed
  165. Damman K, Voors AA. Levosimendan improves renal function in acute decompensated heart failure: cause and clinical application. Cardiovasc Drugs Ther 2007;21:403–4.
    Crossref | PubMed
  166. Singh P, Ricksten SE, Bragadottir G, et al. Renal oxygenation and haemodynamics in acute kidney injury and chronic kidney disease. Clin Exp Pharmacol Physiol 2013;40:138–47.
    Crossref | PubMed
  167. Santoro F, Ieva R, Ferraretti A, et al. Safety and feasibility of levosimendan administration in takotsubo cardiomyopathy: a case series. Cardiovasc Ther 2013;31:e133–7.
    Crossref | PubMed
  168. Schulz R, Rose J, Martin C, et al. Development of short-term myocardial hibernation. Its limitation by the severity of ischemia and inotropic stimulation. Circulation 1993;88:684–95.
    Crossref | PubMed
  169. Beohar N, Erdogan AK, Lee DC, et al. Acute heart failure syndromes and coronary perfusion. J Am Coll Cardiol 2008;52:13–16.
    Crossref | PubMed
  170. Duncker DJ, Koller A, Merkus D, et al. Regulation of coronary blood flow in health and ischemic heart disease. Prog Cardiovasc Dis 2015;57:409–22.
    Crossref | PubMed
  171. Pelliccia F, Kaski JC, Crea F, et al. Pathophysiology of Takotsubo syndrome. Circulation 2017;135:2426–41.
    Crossref | PubMed
  172. Rudiger A, Singer M. Mechanisms of sepsis-induced cardiac dysfunction. Crit Care Med 2007;35:1599–608.
    Crossref | PubMed
  173. Suzuki T, Suzuki Y, Okuda J, et al. Sepsis-induced cardiac dysfunction and β-adrenergic blockade therapy for sepsis. J Intensive Care 2007;5:22.
    Crossref | PubMed
  174. Belletti A, Benedetto U, Biondi-Zoccai G, et al. The effect of vasoactive drugs on mortality in patients with severe sepsis and septic shock. A network meta-analysis of randomized trials. J Crit Care 2017;37:91–8.
    Crossref | PubMed
  175. Wang Q, Yokoo H, Takashina M, et al. Anti-inflammatory profile of levosimendan in cecal ligation-induced septic mice and in lipopolysaccharide-stimulated macrophages. Crit Care Med 2015;43:e508–20.
    Crossref | PubMed
  176. Tsao CM, Li KY, Chen SJ, et al. Levosimendan attenuates multiple organ injury and improves survival in peritonitis-induced septic shock: studies in a rat model. Crit Care 2014;18:1683–12.
    Crossref | PubMed
  177. Morelli A, Donati A, Ertmer C, et al. Levosimendan for resuscitating the microcirculation in patients with septic shock: a randomized controlled study. Crit Care 2010;14:R232.
    Crossref | PubMed
  178. Torraco A, Carrozzo R, Piemonte F, et al. Effects of levosimendan on mitochondrial function in patients with septic shock: a randomized trial. Biochimie 2014;102:166–73.
    Crossref | PubMed
  179. Hajjej Z, Meddeb B, Sellami W, et al. Effects of levosimendan on cellular metabolic alterations in patients with septic shock: a randomized controlled pilot study. Shock 2017;48:307–12.
    Crossref | PubMed
  180. Singer M. Catecholamine treatment for shock: equally good or bad? Lancet 2007;370:636–7.
    Crossref | PubMed
  181. Andreis DT, Singer M. Catecholamines for inflammatory shock: a Jekyll-and-Hyde conundrum. Intensive Care Med 2016;42:1387–97.
    Crossref | PubMed
  182. Coquerel D, Sainsily X, Dumont L, et al. The apelinergic system as an alternative to catecholamines in low-output septic shock. Crit Care 2018;22:10.
    Crossref | PubMed
  183. He X, Su F, Taccone FS, et al. A selective V(1A) receptor agonist, selepressin, is superior to arginine vasopressin and to norepinephrine in ovine septic shock. Crit Care Med 2016;44:23–31.
    Crossref | PubMed
  184. Khanna A, English SW, Wang XS, et al. Angiotensin II for the treatment of vasodilatory shock. N Engl J Med 2017;377:419–30.
    Crossref | PubMed
  185. Creteur J, Bouckaert Y, Mélot C, et al. Effects of levosimendan on systemic and regional hemodynamics in septic myocardial depression. Intensive Care Med 2006;32:790, author reply 791–2.
    Crossref | PubMed
  186. Gordon AC, Perkins GD, Singer M, et al. Levosimendan for the prevention of acute organ dysfunction in sepsis. N Engl J Med 2016;375:1638–48.
    Crossref | PubMed
  187. Angus DC, Mira JP, Vincent JL. Improving clinical trials in the critically ill. Crit Care Med 2010;38:527–32.
    Crossref | PubMed
  188. Hodgson C, Cuthbertson BH. Improving outcomes after critical illness: harder than we thought! Intensive Care Med 2016;42:1772–4.
    Crossref | PubMed
  189. Marshall JC. Global collaboration in acute care clinical research: opportunities, challenges, and needs. Crit Care Med 2017;45:311–20.
    Crossref | PubMed
  190. Iwashyna TJ, Deane AM. Individualizing endpoints in randomized clinical trials to better inform individual patient care: the TARGET proposal. Crit Care 2016;20:218.
    Crossref | PubMed
  191. Mebazaa A, Laterre PF, Russell JA, et al. Designing phase 3 sepsis trials: application of learned experiences from critical care trials in acute heart failure. J Intensive Care 2016;4:24.
    Crossref | PubMed
  192. Girbes ARJ, de Grooth H-J. Time to stop randomized and large pragmatic trials for intensive care medicine syndromes: the case of sepsis and acute respiratory distress syndrome. J Thorac Dis 2020;12(Suppl 1):S101–9.
    Crossref | PubMed
  193. Zhang YH, Zhang J, Qing EM, et al. Comparison on efficacy and safety between domestic levosimendan versus dobutamine for patients with acute decompensated heart failure. Zhonghua Xin Xue Guan Bing Za Zhi 2012;40:153–6 [in Chinese].
    PubMed
  194. Zhang YH, Qing EM, Zhang J, et al. Hemodynamic and efficacies of domestic levosimendan versus dobutamine in patients with acute decompensated heart failure. Zhonghua Yi Xue Za Zhi 2012;92:555–8 [in Chinese]. PMID: 22490161" target="_blank">PubMed
  195. Wang L, Cui L, Wei JP, et al. Efficacy and safety of intravenous levosimendan compared with dobutamine in decompensated heart failure. Zhonghua Xin Xue Guan Bing Za Zhi 2010;38:527–30 [in Chinese]. PMID: 21033135" target="_blank">PubMed
  196. Suominen PK. Single-center experience with levosimendan in children undergoing cardiac surgery and in children with decompensated heart failure. BMC Anesthesiol 2011;11:18.
    Crossref | PubMed
  197. Lechner E, Moosbauer W, Pinter M, et al. Use of levosimendan, a new inodilator, for postoperative myocardial stunning in a premature neonate. Pediatr Crit Care Med 2007;8:61–3.
    Crossref | PubMed
  198. Momeni M, Rubay J, Matta A, et al. Levosimendan in congenital cardiac surgery: a randomized, double-blind clinical trial. J Cardiothorac Vasc Anesth 2011;25:419–24.
    Crossref | PubMed
  199. Pellicer A, Riera J, Lopez-Ortego P, et al. Phase 1 study of two inodilators in neonates undergoing cardiovascular surgery. Pediatr Res 2013;73:95–103.
    Crossref | PubMed
  200. Ebade AA, Khalil MA, Mohamed AK. Levosimendan is superior to dobutamine as an inodilator in the treatment of pulmonary hypertension for children undergoing cardiac surgery. J Anesth 2013;27:334–9.
    Crossref | PubMed
  201. Ottenheijm CA, Heunks LM, Hafmans T, et al. Titin and diaphragm dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2006;173:527–34.
    Crossref | PubMed
  202. Dres M, Dubé BP, Mayaux J, et al. Coexistence and Impact of limb muscle and diaphragm weakness at time of liberation from mechanical ventilation in medical intensive care unit patients. Am J Respir Crit Care Med 2017;195:57–66.
    Crossref | PubMed
  203. Van Hees HW, Dekhuijzen PN, Heunks LM. Levosimendan enhances force generation of diaphragm muscle from patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2009;179:41–7.
    Crossref | PubMed
  204. Hooijman PE, Beishuizen A, de Waard MC, et al. Diaphragm fiber strength is reduced in critically ill patients and restored by a troponin activator. Am J Respir Crit Care Med 2014;189:
    863–5.
    Crossref | PubMed
  205. Doorduin J, Sinderby CA, Beck J, et al. The calcium sensitizer levosimendan improves human diaphragm function. Am J Respir Crit Care Med 2012;185:90–5.
    Crossref | PubMed
  206. Roesthuis L, van der Hoeven H, Sinderby C, et al. Effects of levosimendan on respiratory muscle function in patients weaning from mechanical ventilation. Intensive Care Med 2019;45:1372–81.
    Crossref | PubMed
  207. Brown RH, Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med 2017;377:162–72.
    Crossref | PubMed
  208. Kurian KM, Forbes RB, Colville S, et al. Cause of death and clinical grading criteria in a cohort of amyotrophic lateral sclerosis cases undergoing autopsy from the Scottish Motor Neurone Disease Register. J Neurol Neurosurg Psychiatry 2009;80:84–7.
    Crossref | PubMed
  209. Paulukonis ST, Roberts EM, Valle JP, et al. Survival and cause of death among a cohort of confirmed amyotrophic lateral sclerosis cases. PLoS One 2015;10:e0131965.
    Crossref | PubMed
  210. Gowland A, Opie-Martin S, Scott KM, et al. Predicting the future of ALS: the impact of demographic change and potential new treatments on the prevalence of ALS in the United Kingdom, 2020-2116. Amyotroph Lateral Scler Frontotemporal Degener 2019;20:264–74.
    Crossref | PubMed
  211. Andrews JA, Cudkowicz ME, Hardiman O, et al. VITALITY-ALS, a phase III trial of tirasemtiv, a selective fast skeletal muscle troponin activator, as a potential treatment for patients with amyotrophic lateral sclerosis: study design and baseline characteristics. Amyotroph Lateral Scler Frontotemporal Degener 2018;19:259–66.
    Crossref | PubMed
  212. de Jongh AD, van Eijk RPA, van den Berg LH. Evidence for a multimodal effect of riluzole in patients with ALS? J Neurol Neurosurg Psychiatry 2019;90:1183–4.
    Crossref | PubMed
  213. Khairoalsindi OA, Abuzinadah AR. Maximizing the survival of amyotrophic lateral sclerosis patients: current perspectives. Neurol Res Int 2018;2018:6534150.
    Crossref | PubMed
  214. Writing Group of the Edaravone (MCI-186) ALS 19 Study Group. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2017;16:505–12.
    Crossref | PubMed
  215. Christensen TH, Kedes L. The myogenic regulatory circuit that controls cardiac/slow twitch troponin C gene transcription in skeletal muscle involves E-box, MEF-2, and MEF-3 motifs. Gene Expr 1999;8:247–61.
    PubMed
  216. Al-Chalabi A, Heunks LMA, Papp Z, et al. Potential of the cardiovascular drug levosimendan in the management of amyotrophic lateral sclerosis: an overview of a working hypothesis. J Cardiovasc Pharmacol 2019;74:389–99.
    Crossref | PubMed
  217. Al-Chalabi A, Shaw P, Leigh PN, et al. Oral levosimendan in amyotrophic lateral sclerosis: a phase II multicentre, randomised, double-blind, placebo-controlled trial. J Neurol Neurosurg Psychiatry 2019;90:1165–70.
    Crossref | PubMed
  218. Rababa’h AM, Alzoubi KH, Baydoun S, et al. Levosimendan prevents memory impairment induced by diabetes in rats: role of oxidative stress. Curr Alzheimer Res 2019;16:1300–8.
    Crossref | PubMed
  219. Rababa’h AM, Alzoubi KH, Atmeh A. Levosimendan
    enhances memory through antioxidant effect in rat model: behavioral and molecular study. Behav Pharmacol 2018;29:344–50.
    Crossref | PubMed
  220. Grossini E, Pollesello P, Bellofatto K, et al. Protective effects elicited by levosimendan against liver ischemia/reperfusion injury in anesthetized rats. J Liver Transpl 2014;20:361–75.
    Crossref | PubMed
  221. Lim H, He D, Qiu Y, et al. Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology. PLoS Comput Biol 2019;15(6):e1006619.
    Crossref | PubMed
  222. Nieminen MS, Fruhwald S, Heunks LM, et al. Levosimendan: current data, clinical use and future development. Heart Lung Vessel 2013;5:227–45.
    Crossref | PubMed
  223. Kivikko M, Pollesello P, Tarvasmäki T, et al. Effect of baseline characteristics on mortality in the SURVIVE trial on the effect of levosimendan vs dobutamine in acute heart failure: sub-analysis of the Finnish patients. Int J Cardiol 2016;215:26–31.
    Crossref | PubMed
  224. Sandner P, Ziegelbauer K. Product-related research: how research can contribute to successful life-cycle management. Drug Discov Today 2008;13:457–63.
    Crossref | PubMed