Abstract
The availability of highly purified recombinant human relaxin, serelaxin, has allowed clinical trials to be conducted in several indications and the elucidation of its pharmacology in human subjects. These studies have demonstrated that serelaxin has unique haemodynamic properties that are likely to contribute to organ protection and long‐term outcome benefits in acute heart failure. Clinical observations support its consideration for therapeutic use in other patient populations, including those with chronic heart failure, coronary artery disease, portal hypertension and acute renal failure.
Linked Articles
This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc
Abbreviations
- AHF
acute heart failure
- Ang II
angiotensin II
- BUN
blood urea nitrogen
- ET‐1
endothelin‐1
- FF
filtration fraction
- HF
heart failure
- HFpEF
heart failure with preserved ejection fraction
- MRSS
modified Rodnan skin score
- PCWP
pulmonary capillary wedge pressure
- RAAS
renin‐angiotensin‐aldosterone system
- RAP
right atrial pressure
- RBF
renal blood flow
- RPF
renal plasma flow
- SBP
systolic blood pressure
- SSc
systemic sclerosis
- SVR
systemic vascular resistance
- WHF
worsening heart failure
Tables of Links
TARGETS |
---|
GPCRs |
ETA receptors |
ETB receptors |
RXFP1 receptors |
LIGANDS | |
---|---|
Ang II, angiotensin II | Relaxin 2 |
Cinaciguat | Rolofylline |
Dopamine | TGF‐β |
ET‐1, endothelin 1 | TNF‐α |
Nesiritide | Ularitide |
NO |
These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Southan et al., 2016), and are permanently archived in the Concise Guide to PHARMACOLOGY 2015/16 (Alexander et al., 2015).
Introduction
Relaxin‐2 is a naturally occurring peptide hormone which was first investigated clinically in the 1950s as a partially purified extract of porcine ovaries (Casten and Boucek, 1958; Casten et al., 1960). In the intervening decades, large strides have been made in relaxin research, including the ability to produce highly purified recombinant human relaxin, known as serelaxin. The availability of serelaxin has enabled the study of its human pharmacology and unique mechanism of action in randomized, placebo‐controlled trials.
As is the case with other peptide hormones, serelaxin is believed to have temporal‐ and spatial‐specific effects, which require expression of the relaxin RXFP1 receptor, in relevant cell types (see Bathgate et al., 2013; Halls et al., 2015). In addition, the determination of which downstream signalling pathways are elicited in target cells and the magnitude of effect size appear to be highly dependent upon the status of other ligand–receptor systems, which are themselves tightly regulated, such as those for endothelin (Danielson et al., 2000; Dschietzig et al., 2003), angiotensin II (Ang II) (Chow et al., 2014) and TGF‐β (Heeg et al., 2005; Mookerjee et al., 2009). These relationships are likely to lead to a physiologically complex integrated system regulating both the vasculature and the interstitium. From a clinical development perspective, these interrelationships provide the potential for serelaxin to intervene in the progression of diseases in which dysregulation of these systems is part of the underlying pathology.
In women, endogenous relaxin‐2 is released into the circulation by the corpus luteum, initially rising to a peak approximating 1 ng·mL−1 during the first trimester of pregnancy and subsequently decreasing slightly over the course of gestation (Szlachter et al., 1982). In contrast, relaxin in the systemic circulation in rodents first rises mid‐pregnancy and demonstrates a pre‐partum surge in circulating concentrations (Sherwood, 1994). This difference in expression pattern may form the basis for the finding that certain aspects of the physiology of relaxin, such as peri‐partum cervical ripening, are clearly relaxin‐dependent in rodents (see Sherwood, 1994) but apparently not significantly affected by serelaxin in humans (Weiss et al., 2016). Because species‐specificity may be relevant with regard to other pharmacological responses to serelaxin, it is useful to take note of the responses that have been observed in completed human clinical studies using serelaxin, including those trials in which the primary endpoints have not been met. This review will describe responses observed in past clinical studies, current indications being pursued and indications that could prove fruitful in the future.
Observations in completed clinical trials
To date, serelaxin has been administered systemically in completed trials to more than 1500 human subjects, including healthy volunteers (Chen et al., 1993; Smith et al., 2006; Dahlke et al., 2015b), patients with systemic sclerosis (SSc) (Seibold et al., 1998; Seibold et al., 2000; Khanna et al., 2009), patients with acute (Teerlink et al., 2009; Teerlink et al., 2013; Ponikowski et al., 2014; Sato et al., 2015) and chronic (Dschietzig et al., 2009; Voors et al., 2014) heart failure (HF), women with post‐date pregnancies (Weiss et al., 2016), patients with hepatic impairment (Kobalava et al., 2015) and renal impairment (Dahlke et al., 2015a), and patients with compensated cirrhosis (Lachlan et al., 2015a,b) (Table 1). The pharmacokinetics of serelaxin in humans are well studied (Chen et al., 1993; Seibold et al., 1998; Dahlke et al., 2015a,b; Kobalava et al., 2015). Following i.v. infusion, steady state concentrations are attained by 4 h, the AUC is dose‐proportional over a wide range of doses (10–100 μg·kg−1·day−1), with terminal half‐lives from 7–16 h (Dahlke et al., 2015a).
Table 1.
Study population | clinicaltrials.gov or EudraCT identifier | Phase of study | Number of subjects | Serelaxin dose (μg·kg−1·day−1) | Duration/route of dosing | References |
---|---|---|---|---|---|---|
AHF patients | NCT00520806 | Ph II (Pre‐RELAX‐AHF) | 234 | 0, 10, 30, 100, 250 | 48 h/i.v. | Teerlink et al., 2009 |
NCT01870778 | Ph III (RELAX‐AHF‐1) | 1161 | 0, 30 | 48 h/i.v. | Metra et al., 2013; Teerlink et al., 2013; Filippatos et al., 2014 | |
NCT01543854 | Ph II | 71 | 0, 30 | 20 h/i.v. | Ponikowski et al., 2014 | |
NCT02002702 | Ph II | 46 | 0, 10, 30 | ≤48 h/i.v. | Sato et al., 2015 | |
Chronic HF patients | NCT00259116 | Ph II | 16 | Group A: 10, 30, 100; Group B: 240, 480, 960; Group C: 960 | Groups A and B: 8 h sequential infusions of increasing doses; Group C: 24 h/i.v. | Dschietzig et al., 2009 |
NCT01546532 | Ph II | 65 | 0, 30 | 24 h/i.v. | Voors et al., 2014 | |
NCT01982292 | Ph II | 320 | 0, 30 | 3 sequential 48 h/i.v. | Teerlink et al., 2016 | |
Systemic sclerosis patients | NA | Ph I | 30 | 0, 6, 12, 50, 100, 200 | 28 d/s.c. | Seibold et al., 1998 |
NA | Ph II | 68 | 0, 25, 100 | 24 wk/s.c. | Seibold et al., 2000 | |
NCT00704665 | Ph III | 231 | 0, 10, 25 | 24 wk/s.c. | Khanna et al., 2009; Teichman et al., 2009 | |
Cirrhosis patients | NCT01640964 | Ph II | 46 | 0, 80 followed by 30 | 1 h/i.v. for each dose | Lachlan et al., 2015a,b |
Healthy volunteers | NA | Ph I | 25 | 10 | Bolus / i.v. | Chen et al., 1993 |
NA | Ph II | 11 | 0.2 μg/kg bolus followed by 0.5 μg/kg/h | Bolus/i.v. followed by 5 h/i.v. | Smith et al., 2006 | |
EudraCT #2010–022528‐58 | Ph II | 40 | 0, 10, 30, 100 | 48 h/i.v. | Dahlke et al., 2015b | |
Healthy volunteers and subjects with hepatic impairment | NCT01433458 | Ph II | 49 | 30 | 24 h/i.v. | Kobalava et al., 2015 |
Subjects with renal impairment | NCT01875523 | Ph I | 36 | 10 | 4 h/i.v. | Dahlke et al., 2015a |
Post‐date pregnant women | NCT00259103 | Ph II | 72 | 0, 7.5, 25,75 | 24 h/i.v. | Weiss et al., 2016 |
Listed are all completed Phase (Ph) I, II, and III clinical trials in which serelaxin was systemically administered. clinicaltrials.gov or EudraCT identifiers are listed except when not available (NA). Data include type of subjects enrolled, number of subjects treated in each trial, doses administered, duration of treatment in hours (h), days (d) or weeks (wk), route of administration (i.v. or s.c.) and publications in which data are summarized.
The clinical trials of serelaxin mentioned above provide good evidence for serelaxin‐induced, receptor‐mediated, systemic and renovascular dilation. Additional but limited evidence, which will not be discussed in this review, has suggested that serelaxin can induce endometrial angiogenesis that may be related to local stimulation of VEGF (Unemori et al., 1999). Based on these and other observations (Unemori et al., 2009; Conrad and Shroff, 2011) two clinical trials were initiated in preeclampsia (clinicaltrials.gov identifiers NCT00333307 and NCT01566630), but enrolling patients into these trials proved difficult and the studies were terminated. Additionally, a modest natriuretic effect has been observed in some (Smith et al., 2006) but not all (Voors et al., 2014) subjects.
Systemic vasodilation
The vasodilatory effects of serelaxin have been measured invasively in clinical studies as a decrease in cardiac filling pressures, such as pulmonary capillary wedge pressure (PCWP) and right atrial pressure (RAP), and non‐invasively as a decrease in systolic blood pressure (SBP). In a double‐blind haemodynamic study in male and female patients with acute heart failure (AHF) with an entry PCWP ≥18 mmHg and SBP ≥115 mmHg, serelaxin administration caused a significant reduction in systemic vascular resistance, RAP, PCWP, pulmonary vascular resistance and pulmonary artery pressure with most of the changes evident 30 min after the start of the i.v. infusion and maintained throughout the 20 h infusion period (Ponikowski et al., 2014). No significant change in cardiac index was observed, consistent with the interpretation that a reduction in preload accompanied the decrease in afterload. A serelaxin‐dependent decrease in SBP was statistically significant at several time points from 30 min following the start of the infusion through to 24 h after the infusion was terminated. Despite differences in experimental design and study subject, these data are generally consistent with those suggesting serelaxin‐mediated systemic vasodilation at comparable doses in an earlier open‐label haemodynamic study in patients with chronic HF (Dschietzig et al., 2009). Unlike nitrate vasodilators (Munzel et al., 2014), no tolerance developed. The results from these invasive haemodynamic studies suggest that, at least in HF patients, serelaxin reduces preload and afterload via rapid systemic arterial and venous vasodilation with potential effects on the pre‐capillary pulmonary bed, as well.
In the Phase III RELAX‐AHF‐1 study, which enrolled AHF patients of the ‘vascular failure’ type (Gheorghiade et al., 2005) with a mean baseline SBP of 142 mmHg, 48 h treatment with serelaxin resulted in greater decreases in SBP from baseline compared with placebo during the infusion period through to 24 h after the infusion was terminated (Teerlink et al., 2013). In the Phase II Pre‐RELAX‐AHF study of similar AHF patients (mean baseline SBP of 147 mmHg), 48 h serelaxin treatment was also associated with a drop in SBP and in a post hoc analysis, the magnitude of the decrease in SBP was dependent upon the baseline SBP measurement (Teerlink et al., 2009). In patients with a starting SBP ≤140 mmHg, a non‐significant serelaxin treatment effect of −0.7 mmHg relative to placebo was observed. In contrast, the treatment difference was −4.9 mmHg (P = 0.04 by ANOVA) in patients with a starting SBP >140 mmHg.
The SBP‐lowering effect of serelaxin was also evident in patients with SSc, with the effect persisting across the 6 month continuous treatment period (Teichman et al., 2009). The magnitude of the change in these patients was also dependent upon SBP at baseline as subjects with a SBP of ≥140 mmHg at baseline had a much greater fall in SBP than the patient population as a whole. These studies suggest that, as has been observed in nonclinical studies (Debrah et al., 2005), serelaxin may induce more potent systemic receptor‐mediated vasodilation in the setting of pre‐constricted vessels.
Because of serelaxin's potential to interact with multiple pathways known to regulate vascular tone, including endothelin‐1 (ET‐1), Ang II (see Baccari and Bani, 2008; Du et al., 2014; Halls et al., 2015) and bradykinin (see Leo et al., 2016), the precise mechanisms underlying the observed vasodilation in these trials remain speculative. However, based on the importance of ET‐1 in the vasoconstrictive pathophysiology of HF and SSc, an attractive hypothesis is that serelaxin antagonizes the actions of this potent vasoconstrictor. Circulating ET‐1 is elevated in HF and is believed to be at least partly responsible for the vasoconstriction and associated symptoms manifested in HF patients (Kelly and Whitworth, 1999). Patients with SSc also have elevated circulating levels of ET‐1, which are believed to contribute to hypertension and renal crisis in these patients (Vancheeswaran et al., 1994; Penn et al., 2013). In non‐clinical studies, serelaxin up‐regulated the expression of the endothelial endothelin ETB receptor (Dschietzig et al., 2003) and/or increased local enzymic conversion of Big ET to bioactive ET‐1, thus enabling its binding to the endothelial ETB receptor (Jeyabalan et al., 2003). As ET‐1 modulates vascular tone through a balance between ET‐1‐mediated constriction via vascular smooth muscle cell ETA and ETB receptors and dilation and clearance transduced by the endothelial ETB receptor (Schneider et al., 2007), a serelaxin‐mediated preferential increase in the latter receptor would favour ET‐1‐mediated vasodilation, via both NO production and sequestration of ET‐1.
Of further interest is the concept that this pathway may be relevant to the observed reliance of the magnitude of systemic haemodynamic response to serelaxin on baseline haemodynamic status. Because the degree of vasodilation transduced by the endothelial ETB receptor would theoretically depend not only on the extent of target cell expression of the endothelial ETB receptor but also on the availability of its ligand (ET‐1), serelaxin effect size (magnitude of SBP decrease) could conceptually be more marked in subjects with elevated ET‐1 levels (vessel preconstriction) at baseline.
The straightforward clinical readout of a decrease in SBP, however,probably belies a complex and interesting series of serelaxin‐induced changes that may involve systemic and local ET‐1 concentrations and differential expression of the ET‐1 receptors in different vascular beds, as well as contributions by other vasodilator pathways, such as bradykinin (Leo et al., 2016). In addition, the observations of a serelaxin‐associated decrease in aldosterone levels, both in AHF (Teerlink et al., 2016) and chronic HF patients (Serelaxin Briefing Document, 2014), hint at additional interactions between serelaxin and components of the renin‐angiotensin‐aldosterone system (RAAS).
Renovascular dilation
Clinical studies that measured renal function indirectly by assessing serum markers such as cystatin C, serum creatinine, blood urea nitrogen (BUN) or uric acid suggest that serelaxin improves GFR. (i) In healthy individuals, an increase from baseline in estimated creatinine clearance was observed following 48 h serelaxin infusion (10, 30 and 100 μg·kg−1·day−1) compared with the placebo group (Dahlke et al., 2015b). (ii) In hospitalized AHF patients, serelaxin treatment prevented renal worsening (Metra et al., 2013), which is highly predictive of adverse long‐term outcomes in these patients (Gottlieb et al., 2002). In accordance with the decline in renal function in hospitalized AHF patients reported in other studies (Gottlieb et al., 2002; Klein et al., 2008; Lassus et al., 2010), the placebo treatment group in the RELAX‐AHF‐1 trial showed increases from baseline in mean serum creatinine, BUN and uric acid by 24 h after the start of infusion of placebo and in cystatin C by the earliest time point tested (48 h) (Metra et al., 2013). Serelaxin significantly attenuated the elevation in these serum markers to at least Day 5 and if assessed by cystatin C concentrations, to Day 14 (12 days following termination of infusion). This renal benefit occurred concomitantly with a larger mean decrease in BP in the serelaxin group compared with the placebo group (Teerlink et al., 2013). (iii) Similar creatinine results were found following 20 h serelaxin infusion in AHF patients (Ponikowski et al., 2014). Creatinine clearance increased by 20% by the end of the infusion period in the serelaxin treatment group compared with a 24% decrease in the placebo group (39% treatment difference, P = 0.0143 by ANCOVA) (Ponikowski et al., 2014). (iv) Serum creatinine and BUN were also decreased following short‐term i.v. administration of 30 and 100 μg·kg−1·day−1 doses of serelaxin in a small, open‐label study in stable HF patients (Dschietzig et al., 2009). (v) In patients with SSc, estimated creatinine clearance showed persistent increases during 6 months of continuous serelaxin administration (Khanna et al., 2009; Teichman et al., 2009), concomitant with small decreases in SBP (Teichman et al., 2009). (vi) In the recently completed RELAX‐REPEAT study conducted in chronic HF patients (Teerlink et al., 2016), plasma cystatin C levels were lower and estimated GFR was higher for the serelaxin group compared with placebo group after each of the three sequential infusions administered 4 weeks apart.
In these clinical studies, it is possible that the reported changes in serum renal markers could be due to factors other than the proposed serelaxin‐mediated augmentation of glomerular filtration. For example, changes in uric acid depend on status of the xanthine oxidase pathway (Doehner et al., 2007), and creatinine is subject to changes in tubular secretion (Perrone et al., 1992) and extrarenal influences (Sandilands et al., 2013). However, cystatin C is believed to be an accurate measure of GFR (Taub et al., 2012), and the data in aggregate strongly suggest that serelaxin improves GFR. Unlike the observations regarding serelaxin‐mediated decreases in SBP, a reliance of effect size on baseline renal status has not been apparent in human studies conducted to date.
The renal vasodilatory effect of serelaxin has been demonstrated in three clinical trials that measured either renal plasma flow (RPF) using clearance of para‐aminohippurate or renal artery flow in response to serelaxin infusion. Firstly, in a small, open‐label study of healthy volunteers (Smith et al., 2006), a 47% increase from baseline in RPF, with no change in GFR (measured using clearance of inulin), was recorded following a 5 h infusion of serelaxin. Filtration fraction (FF), the ratio of GFR/RPF, was reduced at the end of the infusion period relative to baseline. Secondly, in a placebo‐controlled study of stable chronic HF patients, a 13% increase in RPF, relative to placebo, with no change in GFR (measured using clearance of iothalamate) was observed following serelaxin administration (Voors et al., 2014). Creatinine clearance was unchanged. FF was reduced 16% in the serelaxin group compared to placebo. Finally, in an open‐label study of patients with stable alcohol‐related cirrhosis, renal artery flow measured using magnetic resonance imaging increased 65% relative to baseline following a 2 h serelaxin infusion (30 and 80 μg·kg−1·day−1); GFR was not measured in this study (Lachlan et al., 2015b).
In general, these data demonstrating a serelaxin‐mediated enhancement of RPF inform the underlying basis of the increases in GFR reported in clinical studies. Reasons for the dissociation of RPF and GFR in the Smith et al. (2006) and Voors et al. (2014) studies remain speculative. During early pregnancy, both afferent and efferent glomerular arterioles dilate, resulting in a RPF increase greater than the increase in GFR and a reduction in FF (Helal et al., 2012). In the two trials studying healthy volunteers (Smith et al., 2006; Dahlke et al., 2015b), it is possible that the discordant results (no increase in GFR in the former, improvement in estimated GFR in the latter) reveal a difference in sensitivity of the afferent and efferent arterioles to serelaxin such that the magnitude of serelaxin exposure (dose X duration of treatment) studied by Smith et al. (2006) did not enable sufficient dilation of afferent vessels and a GFR rise while the larger exposure studied by Dahlke et al. (2015b) did. In the HF patients (Voors et al., 2014), it is possible that the relatively small increase in RPF was insufficient to enable GFR augmentation.
In AHF, a decline in renal function, measured as an increase in serum creatinine, occurs in 20–40% of hospitalized AHF patients (Metra et al., 2012) and is a greater predictor of adverse outcomes than measurements of intrinsic cardiac function, such as left ventricular ejection fraction (Fonarow et al., 2005). While the primary mechanism underlying the serelaxin‐mediated improvement of renal function in the AHF studies cited above is likely to reflect a direct effect on intrarenal haemodynamics (improved RPF), other serelaxin‐mediated haemodynamic effects may also have contributed. A reduction in preload, which is associated with renal venous pressure, and a reduction in afterload without hypotension are known to positively affect renal perfusion and GFR (Verbrugge et al., 2014) and may also have contributed to the observed serelaxin effect.
The fact that a two day infusion of serelaxin was associated with improved intermediate to long‐term outcomes in AHF patients (Teerlink et al., 2009; Teerlink et al., 2013) is in keeping with the hypothesis that effectively preventing the acute decline in renal perfusion during hospitalization attenuates the progressive deterioration in renal function described by the cardiorenal syndrome (Gheorghiade et al., 2005). It may be worth noting that while the detoxification aspect of improved glomerular filtration is beneficial, increased renal perfusion in the absence of an elevation in GFR could help to preserve tubular function, including volume homeostasis, in patients with AHF (Verbrugge et al., 2014). In addition, the observed serelaxin‐induced reduction in FF may also have salutary effects on renal function (Mullens and Nijst, 2016). Complete understanding of the protective effect of serelaxin in AHF is partly constrained by limitations in understanding the pathophysiological mechanisms tethering renal worsening during hospitalization to long‐term adverse outcomes in AHF. The precise molecular mechanisms underlying the increase in renal function conferred by serelaxin in AHF patients, as well as in other human subjects, have yet to be defined but may involve antagonism of the renal vasoconstrictive effects of ET‐1 and Ang II via up‐regulation of NO (see Conrad and Shroff, 2011).
That serelaxin mediates an improvement in renal function is notable in the context of clinical experience with other vasodilators tested in AHF. In completed Phase II and III, placebo‐controlled, randomized trials, the vasodilators cinaciguat (Erdmann et al., 2013), tezosentan (McMurray et al., 2007), nesiritide (O'Connor et al., 2011) and ularitide (Mitrovic et al., 2006) have had, at best, a neutral effect on renal function (no different from placebo). In addition, agents in other pharmacological classes that have been tested in AHF, such as rolofylline (Massie et al., 2010) and dopamine (Chen et al., 2013), have also failed to show a favourable effect on renal function.
The observed coincidence of serelaxin‐mediated systemic and renal vasodilation in human subjects appears to be pharmacologically unique as the state of arterial underfilling that occurs as a consequence of peripheral vasodilation typically stimulates sympathetic and RAAS activation, which can lead to renal vasoconstriction (Schrier and Briner, 1991). Indeed, these untoward compensatory mechanisms have been associated with the use of other systemic vasodilators, such as nitrates, in HF (Elkayam et al., 2004; Munzel et al., 2011).
Anti‐fibrosis
To date, evidence suggestive of a serelaxin effect on modification of the extracellular matrix in humans, a robust aspect of its pharmacology in rodent models (reviewed in Samuel et al., 2007), is lacking when studied following systemic serelaxin administration in trials of SSc (Seibold et al., 1998; Seibold et al., 2000; Khanna et al., 2009) and cervical ripening (Weiss et al., 2016). The cervical ripening trial was the first in this indication to test the efficacy of serelaxin systemically administered at the end of pregnancy to advance ripening and labour (Weiss et al., 2016). Unlike the potent activity observed in rodents and pigs, serelaxin failed to accelerate cervical ripening in post‐date pregnant women, a difference that may be related to the dissimilarity in the secretion patterns of endogenous relaxin (see Sherwood, 1994) and its role during pregnancy among these species.
The two efficacy studies conducted in patients with SSc tested the ability of serelaxin to reverse, halt or delay progressive fibrosis using serial measurements of skin thickening, a metric that is predictive of visceral fibrotic involvement, and is the key clinical criterion used in SSc diagnosis and is correlated with decreased survival (Krieg and Takehara, 2009). A continuous 6 month systemic treatment regimen with serelaxin was considered necessary to allow manifestation of the putative anti‐fibrotic effects of serelaxin, including inhibition of interstitial collagen deposition and increased collagen degradation by MMPs, in patients with early diffuse SSc (Unemori and Amento, 1990; Unemori et al., 1992). While the small Phase II study was encouraging (Seibold et al., 2000), the larger pivotal trial failed to meet its primary endpoint, a clinically meaningful reduction in the Modified Rodnan Skin Score (MRSS), a palpation‐based measurement of skin thickness, compared to placebo (Khanna et al., 2009). Subgroup analysis by duration of disease (≤2.5 and >2.5 years) yielded no difference in MRSS results. While limitations of the MRSS, including susceptibility to intra‐ and inter‐observer variability, are acknowledged, assessors were routinely trained and the MRSS still remains the gold standard for quantitation of cutaneous fibrosis. Serelaxin‐associated improvements in lung function tests, such as forced vital capacity, which would be suggestive of pulmonary fibrosis amelioration, were also not observed. Serial pharmacokinetic measurements confirming the presence of serum serelaxin and observations of serelaxin‐associated menorrhagia and changes in estimated creatinine clearance and blood pressure confirmed continuous systemic exposure to serelaxin. For these reasons, the data in the pivotal study are a reasonable basis upon which to conclude that chronic exposure to a pharmacological dose of serelaxin sufficient to cause systemic and renal haemodynamic and uterine effects does not produce a measurable reduction in cutaneous fibrosis in patients with early diffuse SSc.
While these negative data comprise an important component of the database on human experience with serelaxin, they do not preclude the potential utility of serelaxin in other clinical indications in which fibrosis plays a pathophysiological role. A complex and varied array of cell types and soluble mediators can shape the dysregulated wound healing process that culminates in fibrosis (Wynn and Ramalingam, 2013), and it seems unlikely that serelaxin would be equipotent as an anti‐fibrotic in all circumstances. Translational rodent studies, while they may or may not faithfully represent serelaxin's anti‐fibrotic pharmacology in humans, serve to highlight the importance of the underlying basis of fibrosis in determining susceptibility to amelioration by serelaxin (Wong et al., 2013; Dschietzig et al., 2015).
Current and future trials
Acute heart failure
The clinical development programme in AHF is currently continuing with the 6800‐patient randomized, placebo‐controlled RELAX‐AHF‐2 trial (clinicaltrials.gov identifier NCT01870778). This study is designed to demonstrate an improvement in cardiovascular and all‐cause mortality, consistent with observations in the Phase III RELAX‐AHF‐1 (Teerlink et al., 2013) and Phase II Pre‐RELAX‐AHF trials (Teerlink et al., 2009) (Metra et al., 2013). It will also examine serelaxin's effect on worsening heart failure (WHF), which is strongly associated with increased mortality (Torre‐Amione et al., 2009; Metra et al., 2010); serelaxin reduced WHF in the Phase III RELAX‐AHF‐1 study (Teerlink et al., 2013) and showed similar trends in the Phase II Pre‐RELAX‐AHF study (Teerlink et al., 2009). RELAX‐AHF‐2 should also be instructive regarding causes of death in this AHF subpopulation in general and should also inform mechanisms specifically pertaining to serelaxin's apparent effect on mortality, including those related to stroke (Felker et al., 2014). The AHF programme has also expanded to Asia with the completion of a Phase II (Sato et al., 2015) and initiation of a Phase III AHF study (clinicaltrials.gov identifier NCT02007720). A Phase II study is also currently recruiting to assess the safety and pharmacokinetics of serelaxin in paediatric heart failure patients (clinicaltrials.gov identifier NCT02151383).
As AHF is an episodic disease with patients repeatedly hospitalized following decompensation, sequential dosing with serelaxin over time in these patients is envisioned. Although serelaxin is identical to the naturally occurring hormone relaxin‐2, antibody development to human proteins is known to occur following repeated or extended dosing (Schellekens, 2008). An additional study (RELAX‐REPEAT) has been completed, therefore, to examine the safety and antibody response to three sequential 48 h i.v. infusions of serelaxin 4 weeks apart in chronic HF patients (clinicaltrials.gov identifier NCT01982292). Results indicated that the repeat doses of serelaxin were safe and well tolerated with a favourable immunogenicity profile, including no confirmed hypersensitivity or infusion‐related reactions and only one patient of the 200 receiving serelaxin transiently developing antibodies (Teerlink et al., 2016). The antibodies were non‐neutralizing and not associated with adverse events.
Chronic heart failure
Fifty per cent of HF patients are in the subset of heart failure with preserved ejection fraction (HFpEF) whose 3 year mortality prognosis approximates to 50% (Loffredo et al., 2014). While patients with reduced ejection fraction have been successfully treated, HFpEF patients have not shown similar benefit in clinical trials after treatment with ACE inhibitors, Ang II receptor blockers, β‐blockers, digoxin, aldosterone antagonists or sildenafil (Jeong and Dudley, 2015). A post hoc analysis of data from RELAX‐AHF‐1 suggested that the serelaxin‐mediated clinical responses and favourable changes in HF‐relevant biomarkers of organ damage were similar in the HFpEF subgroup (approximating 25% of the patient cohort) compared with that observed in patients with reduced ejection fraction (Filippatos et al., 2014).
The reduction in mortality observed following short‐term serelaxin treatment during hospitalization in Pre‐RELAX‐AHF and RELAX‐AHF‐1 has led to the hypothesis that serelaxin may be interrupting pathophysiological cascades of events that, if unmodified, lead to cumulative organ damage and subsequent adverse outcomes (Metra et al., 2013; Diez, 2014; Dschietzig, 2014; Du et al., 2014). This hypothesis could be envisioned as encompassing a beneficial effect of short‐term serelaxin infusion on the attenuation of myocardial fibrosis characteristic of HFpEF patients, as well. In these patients, left ventricular diastolic dysfunction is believed to be the result of dysregulated remodelling of myocardial architecture, including cardiomyocyte hypertrophy and interstitial fibrosis (Loffredo et al., 2014). It has been proposed that cardiac fibroblasts, once activated along the pro‐fibrotic cascade to become myofibroblasts, are long‐lived, surviving for months to years following an inciting insult, and continuously stimulated to produce a collagen matrix in autocrine fashion by the generation of Ang II and TGF‐β in a persistent ‘secretome’ (Weber et al., 2013). Non‐clinical studies suggest that short‐term serelaxin treatment can block myofibroblast activation induced by both Ang II and TGF‐β, (Samuel et al., 2004). Thus, a 48 h infusion of serelaxin during a period of exacerbation of pro‐fibrotic stimuli, such as acute decompensation, could theoretically have incremental benefits in preventing progression of ventricular stiffness in the intermediate term.
A recent hypothesis regarding pathophysiology in HFpEF suggests that coronary microvascular inflammation, rather than afterload mismatch, is the primary driver of fibrosis and cardiomyocyte hypertrophy (Paulus and Tschope, 2013). Co‐morbidities such as obesity and ageing contribute to a chronic systemic proinflammatory state, resulting in coronary microvascular inflammation, NO dysregulation and oxidative stress. This process ultimately stimulates cardiomyocyte stiffness due to both hypertrophy and titin hypophosphorylation, as well as triggering myofibroblast activation and interstitial fibrosis. Non‐clinical studies suggest that serelaxin can potentially interfere at a number of steps in the progression of the disorder, including reduction of oxidative stress and attenuation of Ang II‐ and TNF‐α‐induced endothelial inflammation (see Bani, 2008; Conrad and Schroff, 2011; Du et al., 2014; Halls et al., 2015). For these reasons, chronic serelaxin treatment of HFpEF patients following hospital discharge may also have benefits in preventing worsening in these patients. In addition, serelaxin theoretically has the ability to reduce established cardiac fibrosis by stimulating a pro‐matrix degrading fibroblast phenotype (see Samuel et al., 2007). Chronic serelaxin treatment could also be surmised to effect a decrease in vascular stiffness (see Conrad and Shroff, 2011), thus potentially improving compliance and cardiac performance in HFpEF patients (Ooi et al., 2008; Butler et al., 2014).
Requirements for a drug development programme in this indication include (i) establishment of a schedule and route of serelaxin administration that is not only efficacious but also amenable to both patients and physicians to facilitate adherence to the chronic treatment regimen; (ii) the identification of quantifiable, clinically meaningful direct and surrogate endpoints; and (iii) demonstration of safety in the setting of the disease and concomitant medications. The safety data from the RELAX‐REPEAT study (Teerlink et al., 2016) are a requisite and encouraging component of development in this indication.
Coronary artery disease
In AHF patients, release of cytoplasmic cardiac troponin into the circulation (‘troponin leak’) reflects myocyte apoptosis, necrosis or lack of membrane integrity. This can occur as a consequence of increased wall stress, myocardial ischaemia, oxygen supply–demand inequity/oxidative stress or exposure to inflammatory cytokines or neurohormones (ET‐1 and Ang II) (Kociol et al., 2010; Januzzi et al., 2012). In RELAX‐AHF‐1, serelaxin, added to standard care, blunted the troponin increase at Day 2, suggesting a cardioprotective effect that may have contributed to increased survival at 180 days (Metra et al., 2013). Because elevated cardiac filling pressures have been associated with increased troponin release in HF (Horwich et al., 2003; Takashio et al., 2013), it is hypothesized that the observed serelaxin‐mediated rapid decrease in filling pressures (Dschietzig et al., 2009; Ponikowski et al., 2014) may have played a role in the observed decrease in troponin. In addition to the salutary effect of systemic haemodynamic correction induced by serelaxin, a direct vasodilatory, anti‐oxidative stress and anti‐inflammatory effect on coronary vessels, as demonstrated in non‐clinical studies (see Du et al., 2010), may also have contributed to the potential cardioprotective effect.
An exploratory double‐blind, randomized study has been initiated to examine the effect of 48 h infusion of serelaxin (30 μg·kg−1·day−1) on coronary perfusion reserve and pulse wave velocity in patients with coronary artery disease (clinicaltrials.gov identifier NCT01979614). Myocardial perfusion will be assessed using adenosine stress cardiac magnetic resonance imaging and augmentation index will be evaluated at several time points during infusion.
Portal hypertension
Portal hypertension, which is defined as an increase in the pressure gradient between the portal vein and inferior vena cava, is associated with serious complications, including variceal bleeding and the formation of ascites, and is the leading cause of death or liver transplantation in patients with cirrhosis (Bosch et al., 2015). While structural changes such as fibrosis and nodule formation are in part responsible for the increase in pressure, augmentation of hepatic vascular tone following stimulation by ET‐1, Ang II and other vasoconstrictors is believed to account for 30% of the increase in vascular resistance. Therefore, this component is theoretically amenable to rapid modulation (Reynaert et al., 2008). Because portal hypertension can result from increased hepatic artery blood flow, as well as increased intrahepatic vascular resistance, the ideal vasodilator would not contribute to the former and inhibit the latter. The challenge of developing such a therapeutic agent is highlighted by the limitations of simvastatin, which was shown to reduce portal resistance but concomitantly increased hepatic blood flow, resulting in no decrease in portal pressure in patients with cirrhosis (Zafra et al., 2004). NO donors, such as isosorbide 5‐mononitrate, reduced portal pressure (Navasa et al., 1989) but can also decrease renal function (Salmeron et al., 1993).
In non‐clinical models of cirrhosis, serelaxin administration rapidly decreases portal vein pressure, through mechanisms involving an increase in intrahepatic NO signalling and a reduction in contractile filament expression in hepatic stellate cells (Fallowfield et al., 2014). In RELAX‐AHF‐1, salutary effects of 48 h serelaxin infusion on serum liver injury markers were noted (Metra et al., 2013), perhaps suggestive of haemodynamic correction of hepatic congestion and/or impaired liver perfusion (Samsky et al., 2013).
A Phase II study of the effects of serelaxin (i.v. infusion of 80 μg·kg−1·day−1 for 60 min followed by 30 μg·kg−1·day−1 for at least 60 min) on portal hypertension and the hepatic and renal circulation in patients with compensated alcohol‐related cirrhosis was recently completed (Lachlan et al., 2015a; Lachlan et al., 2015b). In an open‐label portion of the study (n = 6), the mean portal pressure gradient, which measured 8.2 mmHg at baseline, was reduced by 31.3% after 120 min of serelaxin infusion. Portal vein pressure declined 30 min following the start of infusion and dropped progressively over time by 33.9% compared with baseline at 135 min. In another group of similar patients (n = 40), serelaxin increased blood flow in the hepatic artery slightly, while no effect on superior mesenteric artery flow was measured. Serelaxin infusion was associated with a 65% increase from baseline in renal blood flow (RBF). These encouraging results support additional larger studies on the acute effects of serelaxin in reducing portal hypertension in cirrhosis (clinicaltrials.gov identifier NCT02669875). If these studies are successful, studies on the potential of extended dosing of serelaxin on amelioration of the fibrotic component of the disease, as has been observed in non‐clinical studies (Williams et al., 2001; Bennett et al., 2014), would be of clinical interest.
Renal failure
Acute injury resulting in renal ischaemia often leads to further renal vasoconstriction (loss of autoregulation), progressing to endothelial damage due to an increase in ROS, a decrease in endothelial NOS and vasodilatory prostaglandins, and an increase in ET‐1 (Schrier et al., 2004). An influx of inflammatory cells contributes to tubular structural changes, such as shedding of epithelial cells, loss of polarity, aberrant fibronectin deposition and luminal obstruction, which lead to impaired tubular function. Prolongation of the insult or inflammation, in which ET‐1 and TNF‐α are implicated, can lead to renal failure, including fibrosis. Theoretically, if vasoconstriction could be reversed or prevented prior to tubular damage, therapy could blunt progression to acute renal failure (Schrier et al., 2004; Singh et al., 2013). Pharmacological agents with the potential to increase RBF that have been tested in acute renal failure include dopamine, fenoldopam and theophylline, but their efficacy remains unproven (Rosner and Okusa, 2006). Because of serelaxin's specific renal vasodilating ability, early therapeutic intervention in diseases of renal failure may be worth considering. From a clinical development perspective, short‐term treatment in clinical settings in which the initial ischaemic insult is known, such as in contrast media‐induced nephropathy (ten Dam and Wetzels, 2008) or cardiac surgery‐associated nephropathy (Mao et al., 2013), would enable the attractive scenario of early or prophylactic intervention.
In conclusion, clinical trials using serelaxin have provided data regarding its systemic and renal vasodilatory ability and organ protective effects in humans, potentially leading to long‐term outcome benefits in AHF. This information has led to additional hypotheses regarding its mechanisms of action and its consideration for future use in additional clinical indications. This ‘bench to bedside and back again’ basis of serelaxin research fosters the continuing evolution of understanding of its pharmacology and will help determine the full potential of this unique molecule in the treatment of human disease.
Conflict of interest
The author declares no conflicts of interest.
Acknowledgements
The author thanks Dr. Marion Dahlke (Novartis Pharmaceuticals Corporation) and Dr. Sam Teichman for their contribution in reviewing this manuscript and would also like to acknowledge the subjects and investigators who participated in the serelaxin studies reported in this paper.
Unemori, E. (2017) Serelaxin in clinical development: past, present and future. British Journal of Pharmacology, 174: 921–932. doi: 10.1111/bph.13695.
References
- Alexander SPH, Davenport AP, Kelly E, Marrion N, Peters JA, Benson HE et al. (2015). The Concise Guide to PHARMACOLOGY 2015/16: G protein‐coupled receptors. Br J Pharmacol 172: 5744–5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baccari MC, Bani D (2008). Relaxin and nitric oxide signalling. Curr Protein Pept Sci 9: 638–645. [DOI] [PubMed] [Google Scholar]
- Bani D (2008). Relaxin as a natural agent for vascular health. Vasc Health Risk Manag 4: 515–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bathgate RA, Halls ML, Westhuizen ET, Callander GE, Kocan M, Summers RJ (2013). Relaxin family peptides and their receptors. Physiol Rev 93: 405–480. [DOI] [PubMed] [Google Scholar]
- Bennett RG, Heimann DG, Singh S, Simpson RL, Tuma DJ (2014). Relaxin decreases the severity of established hepatic fibrosis in mice. Liver Int 34: 416–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bosch J, Groszmann RJ, Shah VH (2015). Evolution in the understanding of the pathophysiological basis of portal hypertension: how changes in paradigm are leading to successful new treatments. J Hepatol 62 (1 Suppl): S121–S130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butler J, Fonarow GC, Zile MR, Lam CS, Roessig L, Schelbert EB et al. (2014). Developing therapies for heart failure with preserved ejection fraction: current state and future directions. JACC Heart Fail 2: 97–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Casten GG, Boucek RJ (1958). Use of relaxin in the treatment of scleroderma. JAMA 166: 319–324. [DOI] [PubMed] [Google Scholar]
- Casten GG, Gilmore HR, Houghten FE, Samuels SS (1960). A new approach to the management of obliterative peripheral arterial disease. Angiology 11: 408–414. [DOI] [PubMed] [Google Scholar]
- Chen HH, Anstrom KJ, Givertz MM, Stevenson LW, Semigran MJ, Goldsmith SR (2013). Low dose dopamine or low‐dose nesiritide in acute heart failure with renal dysfunction: the ROSE acute heart failure randomized trial. JAMA 310: 2533–2543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SA, Perlman AJ, Spanski N, Peterson CM, Sanders SW, Jaffe R et al. (1993). The pharmacokinetics of recombinant human relaxin in nonpregnant women after intravenous, intravaginal, and intracervical administration. Pharm Res 10: 834–838. [DOI] [PubMed] [Google Scholar]
- Chow BS, Kocan M, Bosnyak S, Sarwar M, Wigg B, Jones ES et al. (2014). Relaxin requires the angiotensin II type 2 receptor to abrogate renal interstitial fibrosis. Kidney Int 86: 75–85. [DOI] [PubMed] [Google Scholar]
- Conrad KP, Shroff SG (2011). Effects of relaxin on arterial dilation, remodeling, and mechanical properties. Curr Hypertens Rep 13: 409–420. [DOI] [PubMed] [Google Scholar]
- Dahlke M, Halabi A, Canadi J, Tsubouchi C, Machineni S, Pang Y (2015a). Pharmacokinetics of serelaxin in patients with severe renal impairment or end‐stage renal disease requiring hemodialysis: a single‐dose, open‐label, parallel‐group study. J Clin Pharmacol 56: 474–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dahlke M, Ng D, Yamaguchi M, Machineni S, Berger S, Canadi J et al. (2015b). Safety and tolerability of serelaxin, a recombinant human relaxin‐2 in development for the treatment of acute heart failure, in healthy Japanese volunteers and a comparison of pharmacokinetics and pharmacodynamics in healthy Japanese and Caucasian populations. J Clin Pharmacol 55: 415–422. [DOI] [PubMed] [Google Scholar]
- Dam MA, Wetzels JF (2008). Toxicity of contrast media: an update. Neth J Med 66: 416–422. [PubMed] [Google Scholar]
- Danielson LA, Kercher LJ, Conrad KP (2000). Impact of gender and endothelin on renal vasodilation and hyperfiltration induced by relaxin in conscious rats. Am J Physiol Integr Comp Physiol 279: R1298–R1304. [DOI] [PubMed] [Google Scholar]
- Debrah DO, Conrad KP, Danielson LA, Shroff SG (2005). Effects of relaxin on systemic arterial hemodynamics and mechanical properties in conscious rats: sex dependency and dose response. J Appl Physiol (1985) 98: 1013–1020. [DOI] [PubMed] [Google Scholar]
- Diez J (2014). Serelaxin: a novel therapy for acute heart failure with a range of hemodynamic and non‐hemodynamic actions. Am J Cardiovasc Drugs 14: 275–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doehner W, Haehling S, Anker SD (2007). Uric acid in CHF: marker or player in a metabolic disease? Int J Cardiol 115: 156–158. [DOI] [PubMed] [Google Scholar]
- Dschietzig T, Bartsch C, Richter C, Laule M, Baumann G, Stangl K (2003). Relaxin, a pregnancy hormone, is a functional endothelin‐1 antagonist: attenuation of endothelin‐1‐mediated vasoconstriction by stimulation of endothelin type‐B receptor expression via ERK‐1/2 and nuclear factor‐kappaβ. Circ Res 92: 32–40. [DOI] [PubMed] [Google Scholar]
- Dschietzig T, Teichman S, Unemori E, Wood S, Boehmer J, Richter C et al. (2009). Intravenous recombinant human relaxin in compensated heart failure: a safety, tolerability, and pharmacodynamic trial. J Card Fail 15: 182–190. [DOI] [PubMed] [Google Scholar]
- Dschietzig TB (2014). Recombinant human relaxin‐2: (how) can a pregnancy hormone save lives in acute heart failure? Am J Cardiovasc Drugs 14: 343–355. [DOI] [PubMed] [Google Scholar]
- Dschietzig TB, Krause‐Relle K, Hennequin M, Websky K, Rahnenfuhrer J, Ruppert J et al. (2015). Relaxin‐2 does not ameliorate nephropathy in an experimental model of type 1 diabetes. Kidney Blood Press Res 40: 77–88. [DOI] [PubMed] [Google Scholar]
- Du X‐J, Hewitson TD, Ngugen M‐N, Samuel CS (2014). Therapeutic effects of serelaxin in acute heart failure – necessity for bilateral research translation. Circ J 78: 542–552. [DOI] [PubMed] [Google Scholar]
- Du XJ, Bathgate RA, Samuel CS, Dart AM, Summers RJ (2010). Cardiovascular effects of relaxin: from basic science to clinical therapy. Nat Rev Cardiol 7: 48–58. [DOI] [PubMed] [Google Scholar]
- Elkayam U, Bitar F, Akhter MW, Khan S, Patrus S, Derakhshani M (2004). Intravenous nitroglyercin in the treatment of decompensated heart failure: potential benefits and limitations. J Cardiovasc Pharmacol Ther 9: 227–241. [DOI] [PubMed] [Google Scholar]
- Erdmann E, Semigran MJ, Nieminen MS, Gheorghiade M, Agrawal R, Mitrovic V et al. (2013). Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute decompensated heart failure. Eur Heart J 34: 57–67. [DOI] [PubMed] [Google Scholar]
- Fallowfield JA, Hayden AL, Snowdon VK, Aucott RL, Stutchfield BM, Mole DJ et al. (2014). Relaxin modulates human and rat hepatic myofibroblast function and ameliorates portal hypertension in vivo. Hepatology 59: 1492–1504. [DOI] [PubMed] [Google Scholar]
- Felker GM, Teerlink JR, Butler J, Hernandez AF, Miller AB, Cotter G et al. (2014). Effect of serelaxin on mode of death in acute heart failure: results from the RELAX‐AHF study. J Am Coll Cardiol 64: 1591–1598. [DOI] [PubMed] [Google Scholar]
- Filippatos G, Teerlink JR, Farmakis D, Cotter G, Davison BA, Felker GM et al. (2014). Serelaxin in acute heart failure patients with preserved left ventricular ejection fraction: results from the RELAX‐AHF trial. Eur Heart J 35: 1041–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonarow GC, Adams KF Jr, Abraham WT, Yancy CW, Boscardin WJ, Adhere Scientific Advisory Committee SG et al. (2005). Risk stratification for in‐hospital mortality in acutely decompensated heart failure: classification and regression tree analysis. JAMA 293: 572–580. [DOI] [PubMed] [Google Scholar]
- Gheorghiade M, De Luca L, Fonarow GC, Filippatos G, Metra M, Francis GS (2005). Pathophysiologic targets in the early phase of acute heart failure syndromes. Am J Cardiol 96: 11G–17G. [DOI] [PubMed] [Google Scholar]
- Gottlieb SS, Abraham W, Butler J, Forman DE, Loh E, Massie BM et al. (2002). The prognostic importance of different definitions of worsening renal function in congestive heart failure. J Card Fail 8: 136–141. [DOI] [PubMed] [Google Scholar]
- Halls ML, Bathgate RA, Sutton SW, Dschietzig TB, Summers RJ (2015). International union of basic and clinical pharmacology. XCV. Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1‐4, the receptors for relaxin family peptides. Pharmacol Rev 67: 389–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heeg MH, Koziolek MJ, Vasko R, Schaefer L, Sharma K, Muller GA et al. (2005). The antifibrotic effects of relaxin in human renal fibroblasts are mediated in part by inhibition of the Smad2 pathway. Kidney Int 68: 96–109. [DOI] [PubMed] [Google Scholar]
- Helal I, Fick‐Brosnahan GM, Reed‐Gitomer B, Schrier RW (2012). Glomerular hyperfiltration: definitions, mechanisms and clinical implications. Nat Rev Nephrol 8: 293–300. [DOI] [PubMed] [Google Scholar]
- Horwich TB, Patel J, MacLellan WR, Fonarow GC (2003). Cardiac troponin I is associated with impaired hemodynamics, progressive left ventricular dysfunction, and increased mortality rates in advance heart failure. Circulation 108: 833–838. [DOI] [PubMed] [Google Scholar]
- Januzzi JL Jr, Filippatos G, Nieminen M, Gheorghiade M (2012). Troponin elevation in patients with heart failure: on behalf of the third universal definition of myocardial infarction global task force: heart failure section. Eur Heart J 33: 2265–2271. [DOI] [PubMed] [Google Scholar]
- Jeong EM, Dudley SC Jr (2015). Diastolic dysfunction. Circ J 79: 470–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeyabalan A, Novak J, Danielson LA, Kerchner LJ, Opett SL, Conrad KP (2003). Essential role for vascular gelatinase activity in relaxin‐induced renal vasodilation, hyperfiltration, and reduced myogenic reactivity of small arteries. Circ Res 93: 1249–1257. [DOI] [PubMed] [Google Scholar]
- Kelly JJ, Whitworth JA (1999). Endothelin‐1 as a mediator in cardiovascular disease. Clin Exp Pharmacol Physiol 26: 158–161. [DOI] [PubMed] [Google Scholar]
- Khanna D, Clements PJ, Furst DE, Korn JH, Ellman M, Rothfield N et al. (2009). Recombinant human relaxin in the treatment of systemic sclerosis with diffuse cutaneous involvement: a randomized, double‐blind, placebo‐controlled trial. Arthritis Rheum 60: 1102–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein L, Massie BM, Leimberger JD, O'Connor CM, Pina IL, Adams KF Jr et al. (2008). Admission or changes in renal function during hospitalization for worsening heart failure predict postdischarge survival: results from the outcomes of a prospective trial of intravenous milrinone for exacerbations of chronic heart failure (OPTIME‐CHF). Circ Heart Fail 1: 25–33. [DOI] [PubMed] [Google Scholar]
- Kobalava Z, Villevalde S, Kotovskaya Y, Hinrichsen H, Petersen‐Sylla M, Zaehringer A et al. (2015). Pharmacokinetics of serelaxin in patients with hepatic impairment: a single‐dose, open‐label, parallel group study. Br J Clin Pharmacol 79: 937–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kociol RD, Pang PS, Gheorghiade M, Fonarow GC, O'Connor CM, Felker GM (2010). Troponin elevation in heart failure prevalence, mechanisms, and clinical implications. J Am Coll Cardiol 56: 1071–1078. [DOI] [PubMed] [Google Scholar]
- Krieg T, Takehara K (2009). Skin disease: a cardinal feature of systemic sclerosis. Rheumatology 48: iii14–iii18. [DOI] [PubMed] [Google Scholar]
- Lachlan NJ, Masson N, Ireland H, Weir G, Hay C, Severin T et al. (2015a). Serelaxin reduced portal pressure gradient and portal vein pressure in patients with cirrhosis and portal hypertension. Hepatology 62 (Suppl): 585a. [Google Scholar]
- Lachlan NJ, Semple SIK, Patel D, Severin T, Dongre N, Saini R et al. (2015b). Serelaxin increased renal blood flow in patients with cirrhosis and portal hypertension. Hepatology 62 (Suppl): 345a. [Google Scholar]
- Lassus JP, Nieminen MS, Peuhkurinen K, Pulkki K, Siirila‐Waris K, Sund R et al. (2010). Markers of renal function and acute kidney injury in acute heart failure: definitions and impact on outcomes of the cardiorenal syndrome. Eur Heart J 31: 2791–2798. [DOI] [PubMed] [Google Scholar]
- Leo CH, Jelinic M, Ng HH, Tare M, Parry LJ (2016). Serelaxin: a novel therapeutic for vascular diseases. Trends Pharmacol Sci 37: 498–507. [DOI] [PubMed] [Google Scholar]
- Loffredo FS, Nikolova AP, Pancoast JR, Lee RT (2014). Heart failure with preserved ejection fraction: molecular pathways of the aging myocardium. Circ Res 115: 97–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao H, Katz N, Ariyanon W, Blanca‐Martos L, Adybelli Z, Giuliani A et al. (2013). Cardiac surgery‐associated acute kidney injury. Cardiorenal Med 3: 178–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massie BM, O'Connor CM, Metra M, Ponikowski P, Teerlink JR, Cotter G (2010). Rolofylline, an adenosine A1‐rectpor antagonist, in acute heart failure. N Engl J Med 363: 1419–1428. [DOI] [PubMed] [Google Scholar]
- McMurray JJ, Teerlink JR, Cotter G, Bourge RC, Cleland JG, Jondeau G et al. (2007). Effects of tezosentan on symptoms and clinical outcomes in patients with acute heart failure: the VERITAS randomized controlled trials. JAMA 298: 2009–2019. [DOI] [PubMed] [Google Scholar]
- Metra M, Cotter G, Davison BA, Felker GM, Filippatos G, Greenberg BH et al. (2013). Effect of serelaxin on cardiac, renal, and hepatic biomarkers in the relaxin in acute heart failure (RELAX‐AHF) development program: correlation with outcomes. J Am Coll Cardiol 61: 196–206. [DOI] [PubMed] [Google Scholar]
- Metra M, Cotter G, Gheorghiade M, Dei Cas L, Voors AA (2012). The role of the kidney in heart failure. Eur Heart J 33: 2135–2142. [DOI] [PubMed] [Google Scholar]
- Metra M, Teerlink JR, Felker GM, Greenberg BH, Filippatos G, Ponikowski P et al. (2010). Dyspnoea and worsening heart failure in patients with acute heart failure: results from the Pre‐RELAX‐AHF study. Eur J Heart Fail 12: 1130–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitrovic V, Seferovic PM, Simeunovic D, Ristic AD, Miric M, Moiseyev VS et al. (2006). Haemodynamic and clinical effects of ularitide in decompensated heart failure. Eur Heart J 27: 2823–2832. [DOI] [PubMed] [Google Scholar]
- Mookerjee I, Hewitson TD, Halls ML, Summers RJ, Mathai ML, Bathgate RA et al. (2009). Relaxin inhibits renal myofibroblast differentiation via RXFP1, the nitric oxide pathway, and Smad2. FASEB J 23: 1219–1229. [DOI] [PubMed] [Google Scholar]
- Mullens W, Nijst P (2016). Cardiac output and renal dysfunction. J Am Coll Cardiol 67: 2209–2212. [DOI] [PubMed] [Google Scholar]
- Munzel T, Daiber A, Gori T (2011). Nitrate therapy: new aspects concerning molecular action and tolerance. Circulation 123: 2132–2144. [DOI] [PubMed] [Google Scholar]
- Munzel T, Steven S, Daiber A (2014). Organic nitrates: update on mechanisms underlying vasodilation, tolerance and endothelial dysfunction. Vascul Pharmacol 63: 105–113. [DOI] [PubMed] [Google Scholar]
- Navasa M, Chesta J, Bosch J, Rodes J (1989). Reduction in portal pressure by isosorbide‐5‐mononitrate in patients with cirrhosis. Effects on splanchnic and systemic hemodynamics and liver function. Gastroenterology 96: 1110–1118. [DOI] [PubMed] [Google Scholar]
- O'Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V et al. (2011). Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med 365: 32–43. [DOI] [PubMed] [Google Scholar]
- Ooi H, Chung W, Biolog A (2008). Arterial stiffness and vascular load in heart failure. Congest Heart Fail 14: 31–36. [DOI] [PubMed] [Google Scholar]
- Paulus WJ, Tschope C (2013). A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol 62: 263–271. [DOI] [PubMed] [Google Scholar]
- Penn H, Quillinan N, Khan K, Chakravarty K, Ong VH, Burns A et al. (2013). Targeting the endothelin axis in scleroderma renal crisis: rationale and feasibility. QJM 106: 839–848. [DOI] [PubMed] [Google Scholar]
- Perrone RD, Madias NE, Levey AS (1992). Serum creatinine as an index of renal function: new insights into old concepts. Clin Chem 38: 1933–1953. [PubMed] [Google Scholar]
- Ponikowski P, Mitrovic V, Ruda M, Fernandez A, Voors AA, Vishnevsky A et al. (2014). A randomized, double‐blind, placebo‐controlled, multicentre study to assess haemodynamic effects of serelaxin in patients with acute heart failure. Eur Heart J 35: 431–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reynaert H, Urbain D, Geerts A (2008). Regulation of sinusoidal perfusion in portal hypertension. Anat Rec (Hoboken) 291: 693–698. [DOI] [PubMed] [Google Scholar]
- Rosner MH, Okusa MD (2006). Acute kidney injury associated with cardiac surgery. Clin J Am Soc Nephrol 1: 19–32. [DOI] [PubMed] [Google Scholar]
- Salmeron JM, Ruiz del Arbol L, Gines A, Garcia‐Pagan JC, Gines P, Feu F et al. (1993). Renal effects of acute isosorbide‐5‐mononitrate administration in cirrhosis. Hepatology 17: 800–806. [PubMed] [Google Scholar]
- Samsky MD, Patel CB, DeWald TA, Smith AD, Felker GM, Rogers JG et al. (2013). Cardiohepatic interactions in heart failure. An overview and clinical implications. J Am Coll Cardiol 61: 2397–2405. [DOI] [PubMed] [Google Scholar]
- Samuel CS, Hewitson TD, Unemori EN, Tang ML‐K (2007). Drugs of the future: the hormone relaxin. Cell Mol Life Sci 64: 1539–1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samuel CS, Unemori EN, Mookerjee I, Bathgate RAD, Layfield SL, Mak J et al. (2004). Relaxin modulates cardiac fibroblast proliferation, differentiation, and collagen production and reverses cardiac fibrosis in vivo. Endocrinology 145: 4125–4133. [DOI] [PubMed] [Google Scholar]
- Sandilands EA, Dhaun N, Dear JW, Webb DJ (2013). Measurement of renal function in patients with chronic kidney disease. Br J Clin Pharmacol 76: 504–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato N, Takahashi W, Hirayama A, Ajioka M, Takahashi N, Okishige K et al. (2015). Multicenter, randomized, double‐blinded, placebo‐controlled phase II study of serelaxin in Japanese patients with acute heart failure. Circ J 79: 1237–1247. [DOI] [PubMed] [Google Scholar]
- Schellekens H (2008). How to predict and prevent the immunogenicity of therapeutic proteins. Biotechnol Annu Rev 14: 191–202. [DOI] [PubMed] [Google Scholar]
- Schneider MP, Boesen EI, Pollock DM (2007). Contrasting actions of endothelin ETA and ETB receptors in cardiovascular disease. Annu Rev Pharmacol Toxicol 47: 731–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schrier RW, Briner VA (1991). Peripheral arterial vasodilation hypothesis of sodium and water retention in pregnancy: implications for pathogenesis of preeclampsia‐eclampsia. Obstet Gynecol 77: 632–639. [PubMed] [Google Scholar]
- Schrier RW, Wang W, Poole B, Mitra A (2004). Acute renal failure: definitions, diagnosis, pathogenesis, and therapy. J Clin Invest 114: 5–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seibold JR, Clements PJ, Furst DE, Mayes MD, McCloskey DA, Moreland LW et al. (1998). Safety and pharmacokinetics of recombinant human relaxin in systemic sclerosis. J Rheumatol 25: 302–307. [PubMed] [Google Scholar]
- Seibold JR, Korn JH, Simms R, Clements PJ, Moreland LW, Mayes MD et al. (2000). Recombinant human relaxin in the treatment of scleroderma. A randomized, double‐blind, placebo‐controlled trial. Ann Intern Med 132: 871–879. [DOI] [PubMed] [Google Scholar]
- Serelaxin Briefing Document , prepared by Novartis Pharmaceuticals Corporation for the Cardiovascular and Renal Drugs Advisory Committee Meeting, page 45, February 26, 2014. [Online] Available at: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/CardiovascularandRenalDrugsAdvisoryCommittee/UCM390444.pdf (accessed 12/1/2015)
- Sherwood OD (1994). Relaxin In: Knobil E, Neill JD. (eds). The physiology of reproduction, Second edn. Raven Press, Ltd: New York, pp. 862–1009. [Google Scholar]
- Singh P, Ricksten SE, Bragadottir G, Redfors B, Nordquist L (2013). Renal oxygenation and haemodynamics in acute kidney injury and chronic kidney disease. Clin Exp Pharmacol Physiol 40: 138–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith MC, Danielson LA, Conrad KP, Davison JM (2006). Influence of recombinant human relaxin on renal hemodynamics in healthy volunteers. J Am Soc Nephrol 17: 3192–3197. [DOI] [PubMed] [Google Scholar]
- Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SPH et al. (2016). The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucl Acids Res 44 (Database Issue): D1054–D1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szlachter BN, Quagliarello J, Jewelewicz R, Osathanondh R, Spellacy WN, Weiss G (1982). Relaxin in normal and pathogenic pregnancies. Obstet Gynecol 59: 167–170. [PubMed] [Google Scholar]
- Takashio S, Yamamuro M, Izumiya Y, Sugiyama S, Kojima S et al. (2013). Coronary microvascular dysfunction and diastolic load correlate with cardiac troponin T release measured by a highly sensitive assay in patients with nonischemic heart failure. J Am Coll Cardiol 62: 632–640. [DOI] [PubMed] [Google Scholar]
- Taub PR, Borden KC, Fard A, Maisel A (2012). Role of biomarkers in the diagnosis and prognosis of acute kidney injury in patients with cardiorenal syndrome. Expert Rev Cardiovasc Ther 10: 657–667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teerlink JR, Cotter G, Davison BA, Felker GM, Filippatos G, Greenberg BH et al. (2013). Serelaxin, recombinant human relaxin‐2, for treatment of acute heart failure (RELAX‐AHF): a randomised, placebo‐controlled trial. Lancet 381: 29–39. [DOI] [PubMed] [Google Scholar]
- Teerlink JR, Metra M, Felker GM, Ponikowski P, Voors AA, Weatherley BD et al. (2009). Relaxin for the treatment of patients with acute heart failure (Pre‐RELAX‐AHF): a multicentre, randomised, placebo‐controlled, parallel‐group, dose‐finding phase IIb study. Lancet 373: 1429–1439. [DOI] [PubMed] [Google Scholar]
- Teerlink JR, Saini S, Gullestad L, Descotes J, Masior T et al. (2016). RELAX‐REPEAT: a multicenter, prospective, randomized, double‐blind study evaluating the safety and tolerability of repeat doses of serelaxin in patients with chronic heart failure. J Card Fail 22: S14–S15. [Google Scholar]
- Teichman SL, Unemori E, Dschietzig T, Conrad K, Voors AA, Teerlink JR et al. (2009). Relaxin, a pleiotropic vasodilator for the treatment of heart failure. Heart Fail Rev 14: 321–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torre‐Amione G, Milo‐Cotter O, Kaluski E, Perchenet L, Kobrin I, Frey A et al. (2009). Early worsening heart failure in patients admitted for acute heart failure: time course, hemodynamic predictors, and outcome. J Card Fail 15: 639–644. [DOI] [PubMed] [Google Scholar]
- Unemori EN, Amento EP (1990). Relaxin modulates synthesis and secretion of procollagenase and collagen by human dermal fibroblasts. J Biol Chem 265: 10681–10685. [PubMed] [Google Scholar]
- Unemori EN, Bauer EA, Amento EP (1992). Relaxin alone and in conjunction with interferon‐gamma decreases collagen synthesis by cultured human scleroderma fibroblasts. J Invest Dermatol 99: 337–342. [DOI] [PubMed] [Google Scholar]
- Unemori EN, Erikson ME, Rocco SE, Sutherland KM, Parsell DA, Mak J et al. (1999). Relaxin stimulates expression of vascular endothelial growth factor in normal human endometrial cells in vitro and is associated with menometrorrhagia in women. Hum Reprod 14: 800–806. [DOI] [PubMed] [Google Scholar]
- Unemori E, Sibai B, Teichman SL (2009). Scientific rationale and design of a phase I safety study of relaxin in women with severe preeclampsia. Ann N Y Acad Sci 1160: 381–384. [DOI] [PubMed] [Google Scholar]
- Vancheeswaran R, Magoulas T, Efrat G, Wheeler‐Jones C, Olsen I, Penny R et al. (1994). Circulating endothelin‐1 levels in systemic sclerosis subsets – a marker of fibrosis or vascular dysfunction? J Rheumatol 21: 1838–1844. [PubMed] [Google Scholar]
- Verbrugge FH, Grieten L, Mullens W (2014). Management of the cardiorenal syndrome in decompensated heart failure. Cardiorenal Med 4: 176–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voors AA, Dahlke M, Meyer S, Stepinska J, Gottlieb SS, Jones A et al. (2014). Renal hemodynamic effects of serelaxin in patients with chronic heart failure: a randomized, placebo‐controlled study. Circ Heart Fail 7: 994–1002. [DOI] [PubMed] [Google Scholar]
- Weber KT, Sun Y, Bhattacharya SK, Ahokas RA, Gerling IC (2013). Myofibroblast‐mediated mechanisms of pathological remodelling of the heart. Nat Rev Cardiol 10: 15–26. [DOI] [PubMed] [Google Scholar]
- Weiss G, Teichman S, Stewart D, Nader D, Wood S, Breining P et al (2016). Recombinant human relaxin versus placebo for cervical ripening: a double‐blind randomized trial in pregnant women scheduled for induction of labor. BMC Pregnancy Childbirth 16: 260–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams EJ, Benyon RC, Trim N, Hadwin R, Grove BH, Arthur MJ et al. (2001). Relaxin inhibits effective collagen deposition by cultured hepatic stellate cells and decreases rat liver fibrosis in vivo. Gut 49: 577–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong SE, Samuel CS, Kelly DJ, Zhang Y, Becker GJ, Hewitson TD (2013). The anti‐fibrotic hormone relaxin is not reno‐protective, despite being active, in an experimental model of type 1 diabetes. Protein Pept Lett 20: 1029–1038. [DOI] [PubMed] [Google Scholar]
- Wynn TA, Ramalingam TR (2013). Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 18: 1028–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zafra C, Abraldes JG, Turnes J, Berzigotti A, Fernandez M, Garca‐Pagan JC et al. (2004). Simvastatin enhances hepatic nitric oxide production and decreases the hepatic vascular tone in patients with cirrhosis. Gastroenterology 126: 749–755. [DOI] [PubMed] [Google Scholar]