Resuscitative Effect of Centhaquine (Lyfaquin®) in Hypovolemic Shock Patients: A Randomized, Multicentric, Controlled Trial

Anil Gulati, Dinesh Jain, Nilesh Radheshyam Agrawal, Prashant Rahate, Rajat Choudhuri, Soumen Das, Deba Prasad Dhibar, Madhav Prabhu, Sameer Haveri, Rohit Agarwal, Manish S Lavhale, Anil Gulati, Dinesh Jain, Nilesh Radheshyam Agrawal, Prashant Rahate, Rajat Choudhuri, Soumen Das, Deba Prasad Dhibar, Madhav Prabhu, Sameer Haveri, Rohit Agarwal, Manish S Lavhale

Abstract

Introduction: Centhaquine (Lyfaquin®) showed significant efficacy as a resuscitative agent in animal models of haemorrhagic shock. Its safety and tolerability were confirmed in healthy human volunteers. In this study, our primary objective was to determine the safety, and the secondary objective was to assess the efficacy of centhaquine in patients with hypovolemic shock.

Methods: A prospective, multicentre, randomized phase II study was conducted in male and female patients aged 18-70 years with hypovolemic shock having systolic BP ≤ 90 mmHg. Patients were randomized in a 1:1 ratio to either the control or centhaquine group. The control group received 100 ml of normal saline infusion over 1 h, while the centhaquine group received 0.01 mg/kg of centhaquine in 100 ml normal saline infusion over 1 h. Every patient received standard of care (SOC) and was followed for 28 days.

Results: Fifty patients were included, and 45 completed the trial: 22 in the control group and 23 in the centhaquine group. The demographics of patients in both groups were comparable. No adverse event related to centhaquine was recorded in the 28-day observation period. The baseline, Injury Scoring System score, haemoglobin, and haematocrit were similar in both groups. However, 91% of the patients in the centhaquine group needed major surgery, whereas only 68% in the control group (p = 0.0526). Twenty-eight-day all-cause mortality was 0/23 in the centhaquine group and 2/22 in the control group. The percent time in ICU and ventilator support was less in the centhaquine group than in the control group. The total amount of vasopressors needed in the first 48 h of resuscitation was lower in the centhaquine group than in the control group (3.12 ± 2.18 vs. 9.39 ± 4.28 mg). An increase in systolic and diastolic BP from baseline through 48 h was more marked in the centhaquine group than in the control group. Compared with the control group, blood lactate level was lower by 1.75 ± 1.07 mmol/l in the centhaquine group on day 3 of resuscitation. Improvements in base deficit, multiple organ dysfunction syndrome (MODS) score and adult respiratory distress syndrome (ARDS) were greater in the centhaquine group than in the control group.

Conclusion: When added to SOC, centhaquine is a well-tolerated and effective resuscitative agent. It improves the clinical outcome of patients with hypovolemic shock.

Trial registration: ClinicalTrials.gov identifier number: NCT04056065.

Keywords: Centhaquine; Haemorrhage; Hypovolemia; Resuscitative agent; Shock.

Figures

Fig. 1
Fig. 1
Patient enrolment, randomization, and trial completion
Fig. 2
Fig. 2
Total volume of fluid, blood products, and vasopressors administered during the first 48 h in the control and centhaquine group of patients. Data presented as the mean ± standard error. Each dot represents the amount administered to each patient
Fig. 3
Fig. 3
Systolic blood pressure during the first 48 h in the control and centhaquine groups of patients. The upper panel shows data as the mean ± standard error. The lower panel shows a change in each patient's systolic blood pressure with time. Two-way ANOVA showed a significant change in systolic blood pressure in the centhaquine (p < 0.0001) but less in the control (p = 0.0261) group during the first 12 h of resuscitation
Fig. 4
Fig. 4
Diastolic blood pressure during the first 48 h in the control and centhaquine group of patients. The upper panel shows data as the mean ± standard error. The lower panel shows a change in each patient's diastolic blood pressure with time. Two-way ANOVA showed a significant change in diastolic blood pressure in the centhaquine (p < 0.0001) but not in the control (p = 0.1812) group during the first 12 h of resuscitation
Fig. 5
Fig. 5
Blood lactate levels on days 1 and 3 of resuscitation in the control and centhaquine group of patients. The upper panel shows data as the mean ± standard error in the control and centhaquine groups. The lower panel shows a change in each patient's blood lactate levels on days 1 and 3 of the control (p = 0.0682) and centhaquine (p = 0.0007) groups

References

    1. Taghavi S, Jones G, Duchesne J, McGrew P, Guidry C, Schroll R, et al. Impact of trauma center volume on major vascular injury: an analysis of the National Trauma Data Bank (NTDB) Am J Surg. 2020;220(3):787–792.
    1. Cannon JW. Hemorrhagic shock. N Engl J Med. 2018;378(4):370–379.
    1. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–2128.
    1. Haut ER, Kalish BT, Cotton BA, Efron DT, Haider AH, Stevens KA, et al. Prehospital intravenous fluid administration is associated with higher mortality in trauma patients: a National Trauma Data Bank analysis. Ann Surg. 2011;253(2):371–377.
    1. Haut ER, Kalish BT, Cotton BA, Efron DT, Haider AH, Stevens KA, et al. Reply to letter: "Prehospital intravenous fluid administration is associated with higher mortality in trauma patients". Ann Surg. 2014;259(2):e20–e21.
    1. Holcomb JB, Tilley BC, Baraniuk S, Fox EE, Wade CE, Podbielski JM, et al. Transfusion of plasma, platelets, and red blood cells in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with severe trauma: the PROPPR randomized clinical trial. JAMA. 2015;313(5):471–482.
    1. Holcomb JB, Jenkins D, Rhee P, Johannigman J, Mahoney P, Mehta S, et al. Damage control resuscitation: directly addressing the early coagulopathy of trauma. J Trauma. 2007;62(2):307–310.
    1. Nederpelt CJ, El Hechi MW, Kongkaewpaisan N, Kokoroskos N, Mendoza AE, Saillant NN, et al. Fresh frozen plasma-to-packed red blood cell ratio and mortality in traumatic hemorrhage: nationwide analysis of 4,427 patients. J Am Coll Surg. 2020;230(6):893–901.
    1. Havel C, Arrich J, Losert H, Gamper G, Mullner M, Herkner H. Vasopressors for hypotensive shock. Cochrane Database Syst Rev. 2011;11(5):003709.
    1. Abid O, Akca S, Haji-Michael P, Vincent JL. Strong vasopressor support may be futile in the intensive care unit patient with multiple organ failure. Crit Care Med. 2000;28(4):947–949.
    1. Aoki M, Abe T, Saitoh D, Hagiwara S, Oshima K. Use of vasopressor increases the risk of mortality in traumatic hemorrhagic shock: a nationwide cohort study in Japan. Crit Care Med. 2018;46(12):e1145–e1151.
    1. Georgoff PE, Nikolian VC, Higgins G, Chtraklin K, Eidy H, Ghandour MH, et al. Valproic acid induces prosurvival transcriptomic changes in swine subjected to traumatic injury and hemorrhagic shock. J Trauma Acute Care Surg. 2018;84(4):642–649.
    1. Ganster F, Burban M, de la Bourdonnaye M, Fizanne L, Douay O, Loufrani L, et al. Effects of hydrogen sulfide on hemodynamics, inflammatory response and oxidative stress during resuscitated hemorrhagic shock in rats. Crit Care. 2010;14(5):R165.
    1. Wepler M, Merz T, Wachter U, Vogt J, Calzia E, Scheuerle A, et al. The mitochondria-targeted H2S-Donor AP39 in a murine model of combined hemorrhagic shock and blunt chest trauma. Shock. 2019;52(2):230–239.
    1. Thakral S, Wolf A, Beilman GJ, Suryanarayanan R. Development and in vivo evaluation of a novel lyophilized formulation for the treatment of hemorrhagic shock. Int J Pharm. 2018;537(1–2):162–171.
    1. Gulati A, Sen AP. Dose-dependent effect of diaspirin cross-linked hemoglobin on regional blood circulation of severely hemorrhaged rats. Shock. 1998;9(1):65–73.
    1. Gulati A, Sen AP, Sharma AC, Singh G. Role of ET and NO in resuscitative effect of diaspirin cross-linked hemoglobin after hemorrhage in rat. Am J Physiol. 1997;273(2 Pt 2):H827–H836.
    1. Sloan EP, Koenigsberg M, Gens D, Cipolle M, Runge J, Mallory MN, et al. Diaspirin cross-linked hemoglobin (DCLHb) in the treatment of severe traumatic hemorrhagic shock: a randomized controlled efficacy trial. JAMA. 1999;282(19):1857–1864.
    1. Sloan EP, Koenigsberg MD, Philbin NB, Gao W, Group DCTHSS, European HI. Diaspirin cross-linked hemoglobin infusion did not influence base deficit and lactic acid levels in two clinical trials of traumatic hemorrhagic shock patient resuscitation. J Trauma. 2010;68(5):1158–71.
    1. Vincent JL, Privalle CT, Singer M, Lorente JA, Boehm E, Meier-Hellmann A, et al. Multicenter, randomized, placebo-controlled phase III study of pyridoxalated hemoglobin polyoxyethylene in distributive shock (PHOENIX) Crit Care Med. 2015;43(1):57–64.
    1. Gulati A. Vascular endothelium and hypovolemic shock. Curr Vasc Pharmacol. 2016;14(2):187–195.
    1. Gulati A, Lavhale M, Giri R, Andurkar S, Xanthos T. Centhaquine citrate. Alpha2B-Adrenoceptor ligand, resuscitative agent for hypovolemic shock. Drugs Fut. 2020;45(3):153–163.
    1. Gulati A, Lavhale MS, Garcia DJ, Havalad S. Centhaquin improves resuscitative effect of hypertonic saline in hemorrhaged rats. J Surg Res. 2012;178(1):415–423.
    1. Gulati A, Zhang Z, Murphy A, Lavhale MS. Efficacy of centhaquin as a small volume resuscitative agent in severely hemorrhaged rats. Am J Emerg Med. 2013;31(9):1315–1321.
    1. Kontouli Z, Staikou C, Iacovidou N, Mamais I, Kouskouni E, Papalois A, et al. Resuscitation with centhaquin and 6% hydroxyethyl starch 130/04 improves survival in a swine model of hemorrhagic shock: a randomized experimental study. Eur J Trauma Emerg Surg. 2019;45(6):1077–1085.
    1. Lavhale MS, Havalad S, Gulati A. Resuscitative effect of centhaquin after hemorrhagic shock in rats. J Surg Res. 2013;179(1):115–124.
    1. Papapanagiotou P, Xanthos T, Gulati A, Chalkias A, Papalois A, Kontouli Z, et al. Centhaquin improves survival in a swine model of hemorrhagic shock. J Surg Res. 2016;200(1):227–235.
    1. Reynolds PS, Michael MJ, Cochran ED, Wegelin JA, Spiess BD. Prehospital use of plasma in traumatic hemorrhage (The PUPTH Trial): study protocol for a randomised controlled trial. Trials. 2015;30(16):321.
    1. Bulger EM, May S, Kerby JD, Emerson S, Stiell IG, Schreiber MA, et al. Out-of-hospital hypertonic resuscitation after traumatic hypovolemic shock: a randomized, placebo controlled trial. Ann Surg. 2011;253(3):431–441.
    1. Farrington CP, Manning G. Test statistics and sample size formulae for comparative binomial trials with null hypothesis of non-zero risk difference or non-unity relative risk. Stat Med. 1990;9(12):1447–1454.
    1. Langan NR, Eckert M, Martin MJ. Changing patterns of in-hospital deaths following implementation of damage control resuscitation practices in US forward military treatment facilities. JAMA Surg. 2014;149(9):904–912.
    1. Hayakawa K. Aggressive fluid management in the critically ill: Pro. J Intensive Care. 2019;7:9.
    1. Kobayashi L, Costantini TW, Coimbra R. Hypovolemic shock resuscitation. Surg Clin N Am. 2012;92(6):1403–1423.
    1. Gulati A, Voshtina E, Zhang Z, Murphy A. Alpha adrenergic receptors mediate resuscitative effect of centhaquin in hemorrhaged rats. Crit Care Med. 2012;40(12):U162–U163.
    1. Berlin DA, Bakker J. Understanding venous return. Intensive Care Med. 2014;40(10):1564–1566.
    1. Jansen JR, Maas JJ, Pinsky MR. Bedside assessment of mean systemic filling pressure. Curr Opin Crit Care. 2010;16(3):231–236.
    1. Chalkias A, Koutsovasilis A, Laou E, Papalois A, Xanthos T. Measurement of mean systemic filling pressure after severe hemorrhagic shock in swine anesthetized with propofol-based total intravenous anesthesia: implications for vasopressor-free resuscitation. Acute Crit Care. 2020;35(2):93–101.
    1. Shen T, Baker K. Venous return and clinical hemodynamics: how the body works during acute hemorrhage. Adv Physiol Educ. 2015;39(4):267–271.
    1. Jansen AJ, leNoble PJ, Steegers EA, van Rhenen DJ, Duvekot JJ. Relationship between haemoglobin change and estimated blood loss after delivery. BJOG. 2007;114(5):657.
    1. Yefet E, Yossef A, Suleiman A, Hatokay A, Nachum Z. Hemoglobin drop following postpartum hemorrhage. Sci Rep. 2020;10(1):21546.
    1. Hernandez G, Ospina-Tascon GA, Damiani LP, Estenssoro E, Dubin A, Hurtado J, et al. Effect of a resuscitation strategy targeting peripheral perfusion status vs serum lactate levels on 28-day mortality among patients with septic shock: the ANDROMEDA-SHOCK randomized clinical trial. JAMA. 2019;321(7):654–664.
    1. Chawla LS, Ostermann M, Forni L, Tidmarsh GF. Broad spectrum vasopressors: a new approach to the initial management of septic shock? Crit Care. 2019;23(1):124.
    1. Gazmuri RJ, Whitehouse K, Whittinghill K, Baetiong A, Shah K, Radhakrishnan J. Early and sustained vasopressin infusion augments the hemodynamic efficacy of restrictive fluid resuscitation and improves survival in a liver laceration model of hemorrhagic shock. J Trauma Acute Care Surg. 2017;82(2):317–327.
    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):509–518.
    1. Khanna A, English SW, Wang XS, Ham K, Tumlin J, Szerlip H, et al. Angiotensin II for the treatment of vasodilatory shock. N Engl J Med. 2017;377(5):419–430.
    1. Lat I, Coopersmith CM, De Backer D, Research Committee of the Surviving Sepsis C, Members of the Surviving Sepsis Campaign Research Committee contributing to this article are as f, Co-chair AGA, et al. The surviving sepsis campaign: fluid resuscitation and vasopressor therapy research priorities in adult patients. Crit Care Med. 2021;49(4):623–35.
    1. Gulati A, Zhang Z, Arshad K. Centhaquin decreases the requirement of norepinephrine, maintains blood pressure and improves survival following resuscitation of hemorrhaged rats. Crit Care Med. 2011;39(12):114.
    1. Odom SR, Howell MD, Silva GS, Nielsen VM, Gupta A, Shapiro NI, et al. Lactate clearance as a predictor of mortality in trauma patients. J Trauma Acute Care Surg. 2013;74(4):999–1004.
    1. Doran CM, Doran CA, Woolley T, Carter A, Male K, Midwinter MJ, et al. Targeted resuscitation improves coagulation and outcome. J Trauma Acute Care Surg. 2012;72(4):835–843.

Source: PubMed

3
Předplatit