Selepressin, a novel selective vasopressin V1A agonist, is an effective substitute for norepinephrine in a phase IIa randomized, placebo-controlled trial in septic shock patients

James A Russell, Jean-Louis Vincent, Anne Louise Kjølbye, Håkan Olsson, Allan Blemings, Herbert Spapen, Peder Carl, Pierre-Francois Laterre, Lars Grundemar, James A Russell, Jean-Louis Vincent, Anne Louise Kjølbye, Håkan Olsson, Allan Blemings, Herbert Spapen, Peder Carl, Pierre-Francois Laterre, Lars Grundemar

Abstract

Background: Vasopressin is widely used for vasopressor support in septic shock patients, but experimental evidence suggests that selective V1A agonists are superior. The initial pharmacodynamic effects, pharmacokinetics, and safety of selepressin, a novel V1A-selective vasopressin analogue, was examined in a phase IIa trial in septic shock patients.

Methods: This was a randomized, double-blind, placebo-controlled multicenter trial in 53 patients in early septic shock (aged ≥18 years, fluid resuscitation, requiring vasopressor support) who received selepressin 1.25 ng/kg/minute (n = 10), 2.5 ng/kg/minute (n = 19), 3.75 ng/kg/minute (n = 2), or placebo (n = 21) until shock resolution or a maximum of 7 days. If mean arterial pressure (MAP) ≥65 mmHg was not maintained, open-label norepinephrine was added. Co-primary endpoints were maintenance of MAP >60 mmHg without norepinephrine, norepinephrine dose, and proportion of patients maintaining MAP >60 mmHg with or without norepinephrine over 7 days. Secondary endpoints included cumulative fluid balance, organ dysfunction, pharmacokinetics, and safety.

Results: A higher proportion of the patients receiving 2.5 ng/kg/minute selepressin maintained MAP >60 mmHg without norepinephrine (about 50% and 70% at 12 and 24 h, respectively) vs. 1.25 ng/kg/minute selepressin and placebo (p < 0.01). The 7-day cumulative doses of norepinephrine were 761, 659, and 249 μg/kg (placebo 1.25 ng/kg/minute and 2.5 ng/kg/minute, respectively; 2.5 ng/kg/minute vs. placebo; p < 0.01). Norepinephrine infusion was weaned more rapidly in selepressin 2.5 ng/kg/minute vs. placebo (0.04 vs. 0.18 μg/kg/minute at 24 h, p < 0.001), successfully maintaining target MAP and reducing norepinephrine dose vs. placebo (first 24 h, p < 0.001). Cumulative net fluid balance was lower from day 5 onward in the selepressin 2.5 ng/kg/minute group vs. placebo (p < 0.05). The selepressin 2.5 ng/kg/minute group had a greater proportion of days alive and free of ventilation vs. placebo (p < 0.02). Selepressin (2.5 ng/kg/minute) was well tolerated, with a similar frequency of treatment-emergent adverse events for selepressin 2.5 ng/kg/minute and placebo. Two patients were infused at 3.75 ng/kg/minute, one of whom had the study drug infusion discontinued for possible safety reasons, with subsequent discontinuation of this dose group.

Conclusions: In septic shock patients, selepressin 2.5 ng/kg/minute was able to rapidly replace norepinephrine while maintaining adequate MAP, and it may improve fluid balance and shorten the time of mechanical ventilation.

Trial registration: ClinicalTrials.gov, NCT01000649 . Registered on September 30, 2009.

Keywords: Fluid balance; Mechanical ventilation; Norepinephrine; Selepressin; Septic shock; V1A agonist.

Conflict of interest statement

Ethics approval and consent to participate

The study protocol and informed consent documents were approved by the independent ethics committees or research ethics boards of all participating institutions, and written informed consent was obtained from all patients, their next of kin, or another surrogate decision maker, as appropriate prior to enrollment. The study was also approved by the competent regulatory authorities of each country participating in the trial.

Consent for publication

Informed written consent was received for publication of the manuscript and figures.

Competing interests

JAR reports patents owned by the University of British Columbia (UBC) that are related to PCSK9 inhibitor(s) and sepsis and related to the use of vasopressin in septic shock. JAR is an inventor on these patents. JAR is a founder, director, and shareholder in Cyon Therapeutics Inc. (developing a sepsis therapy). JAR has share options in Leading BioSciences Inc. JAR is a shareholder in Molecular You Corp. JAR reports receiving consulting fees from Cubist Pharmaceuticals (now owned by Merck; formerly was Trius Pharmaceuticals; developing antibiotics), Leading BioSciences Inc. (developing a sepsis therapeutic), Ferring Pharmaceuticals (manufacturer of vasopressin and developing selepressin), Grifols (seller of albumin), La Jolla Pharmaceutical Co. (developing angiotensin II; JAR chairs the data safety and monitoring board of a trial of angiotensin II), CytoVale Inc. (developing a sepsis diagnostic), and Asahi Kasei Pharma America (AKPA; developing recombinant thrombomodulin). JAR reports having received an investigator-initiated grant from Grifols that is provided to and administered by UBC. HO, ALK, LG, and AB are employees of Ferring Pharmaceuticals A/S. JLV is editor-in-chief of Critical Care. JLV, HS, PC, and PFL declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Consolidated Standards of Reporting Trials (CONSORT) diagram of patient flow through the study
Fig. 2
Fig. 2
Proportion of patients maintaining a mean arterial pressure >60 mmHg without any open-label norepinephrine support at the indicated time points. The difference between 2.5 ng/kg/minute and placebo was statistically significant (p 

Fig. 3

Mean cumulative amount ( a…

Fig. 3

Mean cumulative amount ( a ) and infusion rate ( b ) of…

Fig. 3
Mean cumulative amount (a) and infusion rate (b) of open-label norepinephrine over time in septic shock patients. Selepressin and placebo were infused at a constant rate as indicated, whereas norepinephrine was weaned as fast as possible while still keeping the target treatment mean arterial pressure ≥65 mmHg. Numbers at the bottom of the figure indicate number of patients at each time point. Bars indicate SD. Pl Placebo, 1.25 1.25 ng/kg/minute, 2.5 2.5 ng/kg/minute

Fig. 4

Mean cumulative net fluid balance…

Fig. 4

Mean cumulative net fluid balance over 7 days from start of selepressin/placebo treatment.…

Fig. 4
Mean cumulative net fluid balance over 7 days from start of selepressin/placebo treatment. Selepressin and placebo were infused at a constant rate as indicated. Numbers at the bottom of the figure indicate number of patients at each time point. Bars indicate SD. Pl Placebo, 1.25 1.25 ng/kg/minute, 2.5 2.5 ng/kg/minute
Fig. 3
Fig. 3
Mean cumulative amount (a) and infusion rate (b) of open-label norepinephrine over time in septic shock patients. Selepressin and placebo were infused at a constant rate as indicated, whereas norepinephrine was weaned as fast as possible while still keeping the target treatment mean arterial pressure ≥65 mmHg. Numbers at the bottom of the figure indicate number of patients at each time point. Bars indicate SD. Pl Placebo, 1.25 1.25 ng/kg/minute, 2.5 2.5 ng/kg/minute
Fig. 4
Fig. 4
Mean cumulative net fluid balance over 7 days from start of selepressin/placebo treatment. Selepressin and placebo were infused at a constant rate as indicated. Numbers at the bottom of the figure indicate number of patients at each time point. Bars indicate SD. Pl Placebo, 1.25 1.25 ng/kg/minute, 2.5 2.5 ng/kg/minute

References

    1. Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit. Care Med. 2017;45(3):486–552. doi: 10.1097/CCM.0000000000002255.
    1. Holmes CL, Walley KR, Chittock DR, Lehman T, Russell JA. The effects of vasopressin on hemodynamics and renal function in severe septic shock: a case series. Intensive Care Med. 2001;27(8):1416–21. doi: 10.1007/s001340101014.
    1. Landry DW, Levin HR, Gallant EM, Ashton RC, Jr, Seo S, D’Alessandro D, et al. Vasopressin deficiency contributes to the vasodilation of septic shock. Circulation. 1997;95(5):1122–5. doi: 10.1161/01.CIR.95.5.1122.
    1. Patel BM, Chittock DR, Russell JA, Walley KR. Beneficial effects of short-term vasopressin infusion during severe septic shock. Anesthesiology. 2002;96(3):576–82. doi: 10.1097/00000542-200203000-00011.
    1. Russell JA, Walley KR, Singer J, Gordon AC, Hébert PC, Cooper DJ, et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N. Engl. J. Med. 2008;358(9):877–87. doi: 10.1056/NEJMoa067373.
    1. Gordon AC, Mason AJ, Thirunavukkarasu N, Perkins GD, Cecconi M, Cepkova M, et al. Effect of early vasopressin vs norepinephrine on kidney failure in patients with septic shock: the VANISH randomized clinical trial. JAMA. 2016;316(5):509–18. doi: 10.1001/jama.2016.10485.
    1. Hsu JL, Liu V, Patterson AJ, Martin GS, Nicolls MR, Russell JA. Potential for overuse of corticosteroids and vasopressin in septic shock. Crit. Care. 2012;16(5):447. doi: 10.1186/cc11460.
    1. He X, Su F, Taccone FS, Laporte R, Kjølbye AL, Zhang J, et al. A selective V1A receptor agonist, selepressin, is superior to arginine vasopressin and to norepinephrine in ovine septic shock. Crit. Care Med. 2016;44(1):23–31. doi: 10.1097/CCM.0000000000001380.
    1. Maybauer MO, Maybauer DM, Enkhbaatar P, Laporte R, Wiśniewska H, Traber LD, et al. The selective vasopressin type 1a receptor agonist selepressin (FE 202158) blocks vascular leak in ovine severe sepsis. Crit. Care Med. 2014;42(7):e525–33. doi: 10.1097/CCM.0000000000000300.
    1. Rehberg S, Ertmer C, Vincent JL, Morelli A, Schneider M, Lange M, et al. Role of selective V1a receptor agonism in ovine septic shock. Crit. Care Med. 2011;39(1):119–25. doi: 10.1097/CCM.0b013e3181fa3898.
    1. Rehberg S, Yamamoto Y, Sousse L, Bartha E, Jonkam C, Hasselbach AK, et al. Selective V1a agonism attenuates vascular dysfunction and fluid accumulation in ovine severe sepsis. Am. J. Physiol. Heart Circ. Physiol. 2012;303(10):H1245–54. doi: 10.1152/ajpheart.00390.2012.
    1. National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network Comparison of two fluid-management strategies in acute lung injury. N. Engl. J. Med. 2006;354(24):2564–75. doi: 10.1056/NEJMoa062200.
    1. Boyd JH, Forbes J, Nakada TA, Walley KR, Russell JA. Fluid resuscitation in septic shock: a positive fluid balance and elevated central venous pressure are associated with increased mortality. Crit. Care Med. 2011;39(2):259–65. doi: 10.1097/CCM.0b013e3181feeb15.
    1. Vincent JL, Sakr Y, Sprung CL, Ranieri VM, Reinhart K, Gerlach H, et al. Sepsis in European intensive care units: results of the SOAP study. Crit. Care Med. 2006;34(2):344–53. doi: 10.1097/01.CCM.0000194725.48928.3A.
    1. Murphy CV, Schramm GE, Doherty JA, Reichley RM, Gajic O, Afessa B, et al. The importance of fluid management in acute lung injury secondary to septic shock. Chest. 2009;136(1):102–9. doi: 10.1378/chest.08-2706.
    1. Ball SG. Vasopressin and disorders of water balance: the physiology and pathophysiology of vasopressin. Ann. Clin. Biochem. 2007;44(Pt 5):417–31. doi: 10.1258/000456307781646030.
    1. Kaufmann JE, Vischer UM. Cellular mechanisms of the hemostatic effects of desmopressin (DDAVP) J. Thromb. Haemost. 2003;1(4):682–9. doi: 10.1046/j.1538-7836.2003.00190.x.
    1. Kaufmann JE, Iezzi M, Vischer UM. Desmopressin (DDAVP) induces NO production in human endothelial cells via V2 receptor- and cAMP-mediated signaling. J. Thromb. Haemost. 2003;1(4):821–8. doi: 10.1046/j.1538-7836.2003.00197.x.
    1. O’Connor PM, Cowley AW., Jr Vasopressin-induced nitric oxide production in rat inner medullary collecting duct is dependent on V2 receptor activation of the phosphoinositide pathway. Am. J. Physiol. Renal Physiol. 2007;293(2):F526–32. doi: 10.1152/ajprenal.00052.2007.
    1. Morelli A, Ertmer C, Pietropaoli P, Westphal M. Terlipressin: a promising vasoactive agent in hemodynamic support of septic shock. Expert Opin. Pharmacother. 2009;10(15):2569–75. doi: 10.1517/14656560903257808.
    1. Morelli A, Ertmer C, Rehberg S, Lange M, Orecchioni A, Cecchini V, et al. Continuous terlipressin versus vasopressin infusion in septic shock (TERLIVAP): a randomized, controlled pilot study. Crit. Care. 2009;13(4):R130. doi: 10.1186/cc7990.
    1. Laporte R, Kohan A, Heitzmann J, Wisniewska H, Toy J, La E, et al. Pharmacological characterization of FE 202158, a novel, potent, selective, and short-acting peptidic vasopressin V1a receptor full agonist for the treatment of vasodilatory hypotension. J. Pharmacol. Exp. Ther. 2011;337(3):786–96. doi: 10.1124/jpet.111.178848.
    1. Wisniewski K, Galyean R, Tariga H, Alagarsamy S, Croston G, Heitzmann J, et al. New, potent, selective, and short-acting peptidic V1a receptor agonists. J. Med. Chem. 2011;54(13):4388–98. doi: 10.1021/jm200278m.
    1. The ProCESS Investigators A randomized trial of protocol-based care for early septic shock. N. Engl. J. Med. 2014;370(18):1683–93. doi: 10.1056/NEJMoa1401602.
    1. Asfar P, Russell JA, Tuckermann J, Radermacher P. Selepressin in septic shock: a step toward decatecholaminization? Crit. Care Med. 2016;44(1):234–6. doi: 10.1097/CCM.0000000000001441.
    1. Edwards RM, Trizna W, Kinter LB. Renal microvascular effects of vasopressin and vasopressin antagonists. Am. J. Physiol. 1989;256(2 Pt 2):F274–8.
    1. Stolk RF, van der Poll T, Angus DC, van der Hoeven JG, Pickkers P, Kox M. Potentially inadvertent immunomodulation: norepinephrine use in sepsis. Am. J. Respir. Crit. Care Med. 2016;194(5):550–8. doi: 10.1164/rccm.201604-0862CP.
    1. Gordon AC, Wang N, Walley KR, Ashby D, Russell JA. The cardiopulmonary effects of vasopressin compared with norepinephrine in septic shock. Chest. 2012;142(3):593–605. doi: 10.1378/chest.11-2604.

Source: PubMed

3
Abonneren