A Multicentric, Randomized, Controlled Phase III Study of Centhaquine (Lyfaquin®) as a Resuscitative Agent in Hypovolemic Shock Patients

Anil Gulati, Rajat Choudhuri, Ajay Gupta, Saurabh Singh, S K Noushad Ali, Gursaran Kaur Sidhu, Parvez David Haque, Prashant Rahate, Aditya R Bothra, Gyan P Singh, Sanjiv Maheshwari, Deepak Jeswani, Sameer Haveri, Apurva Agarwal, Nilesh Radheshyam Agrawal, Anil Gulati, Rajat Choudhuri, Ajay Gupta, Saurabh Singh, S K Noushad Ali, Gursaran Kaur Sidhu, Parvez David Haque, Prashant Rahate, Aditya R Bothra, Gyan P Singh, Sanjiv Maheshwari, Deepak Jeswani, Sameer Haveri, Apurva Agarwal, Nilesh Radheshyam Agrawal

Abstract

Introduction: Centhaquine (Lyfaquin®) showed significant safety and efficacy in preclinical and clinical phase I and II studies.

Methods: A prospective, multicentric, randomized phase III study was conducted in patients with hypovolemic shock, systolic blood pressure (SBP) ≤ 90 mmHg, and blood lactate levels ≥ 2 mmol/L. Patients were randomized in a 2:1 ratio to the centhaquine group (n = 71) or the control (saline) group (n = 34). Every patient received standard of care (SOC) and was followed for 28 days. The study drug (normal saline or centhaquine 0.01 mg/kg) was administered in 100 mL of normal saline infusion over 1 h. The primary objectives were to determine changes (mean through 48 h) in SBP, diastolic blood pressure (DBP), blood lactate levels, and base deficit. The secondary objectives included the amount of fluids, blood products, and vasopressors administered in the first 48 h, duration of hospital stay, time in intensive care units, time on ventilator support, change in acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), and the proportion of patients with 28-day all-cause mortality.

Results: The demographics of patients and baseline vitals in both groups were comparable. The cause of hypovolemic shock was trauma in 29.4 and 47.1% of control group and centhaquine group patients, respectively, and gastroenteritis in 44.1 and 29.4%, respectively. Shock index (SI) and quick sequential organ failure assessment at baseline were similar in the two groups. An equal amount of fluids and blood products were administered in both groups during the first 48 h of resuscitation. A lesser amount of vasopressors was needed in the first 48 h of resuscitation in the centhaquine group. An increase in SBP from baseline was consistently higher up to 48 h (12.9% increase in area under the curve from 0 to 48 h [AUC0-48]) in the centhaquine group than in the control group. A significant increase in pulse pressure (48.1% increase in AUC0-48) in the centhaquine group compared with the control group suggests improved stroke volume due to centhaquine. The SI was significantly lower in the centhaquine group from 1 h (p = 0.032) to 4 h (p = 0.049) of resuscitation. Resuscitation with centhaquine resulted in a significantly greater number of patients with improved blood lactate (control 46.9%; centhaquine 69.3%; p = 0.03) and the base deficit (control 43.7%; centhaquine 69.8%; p = 0.01) than in the control group. ARDS and MODS improved with centhaquine, and an 8.8% absolute reduction in 28-day all-cause mortality was observed in the centhaquine group.

Conclusion: Centhaquine is an efficacious resuscitative agent for treating hypovolemic shock. The efficacy of centhaquine in distributive shock is being explored.

Trial registration: Clinical Trials Registry, India; ctri.icmr.org.in, CTRI/2019/01/017196; clinicaltrials.gov, NCT04045327.

Conflict of interest statement

Anil Gulati has issued and pending patents and is an employee and stockholder of Pharmazz, Inc. Rajat Choudhuri, Ajay Gupta, Saurabh Singh, S.K. Noushad Ali, Gursaran Kaur Sidhu, Parvez David Haque, Prashant Rahate, Aditya R. Bothra, Gyan P. Singh, Sanjiv Maheshwari, Deepak Jeswani, Sameer Haveri, Apurva Agarwal, and Nilesh Radheshyam Agrawal have no conflicts of interest that are directly relevant to the content of this article.

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 groups. Total urine output in the first 48 h in the control and centhaquine groups. Data are presented as mean ± standard error
Fig. 3
Fig. 3
Systolic BP during the first 48 h in the control and centhaquine groups. The upper panel shows data as the mean ± SEM. The lower panel indicates the number of patients with improved systolic BP at 12, 24, and 48 h of resuscitation. BP blood pressure, SEM standard error of the mean
Fig. 4
Fig. 4
Diastolic BP during the first 48 h in the control and centhaquine groups. The upper panel shows data as the mean ± SEM. The lower panel indicates the number of patients with improved diastolic BP at 12, 24, and 48 h of resuscitation. BP blood pressure, SEM standard error of the mean
Fig. 5
Fig. 5
Mean difference from baseline to 48 h at various time intervals plotted to determine the AUC for systolic blood pressure, diastolic blood pressure, and pulse pressure. Compared with the control group, the AUC0–48 for systolic blood pressure was higher by 12.99%, diastolic blood pressure was lower by 7.44%, and pulse pressure was higher by 48.14% in the centhaquine group. A significant increase in pulse pressure in the centhaquine group strongly suggests increased stroke volume. AUC area under the curve
Fig. 6
Fig. 6
Shock index (HR/SBP), an important indicator of cardiac performance (left ventricular stroke work) in early hemorrhage, was significantly improved by centhaquine in the first 4 h of resuscitation. CI confidence interval, Diff. difference, HR heart rate, SBP systolic blood pressure
Fig. 7
Fig. 7
Blood lactate levels in the control and centhaquine groups on day 3 of resuscitation (upper panel). Changes in blood lactate levels following resuscitation of patients with hypovolemic shock in control and centhaquine groups (lower panel)
Fig. 8
Fig. 8
Base deficit in control and centhaquine groups on day 3 of resuscitation (upper panel). Changes in base deficit following resuscitation of patients with hypovolemic shock in control and centhaquine groups of individual patients (lower panel)
Fig. 9
Fig. 9
ARDS was compared between day 1 (before resuscitation) and day 3 of resuscitation. Centhaquine treatment significantly improved ARDS following resuscitation, whereas improvement was minor in the control group. MODS was compared between day 3 and day 7 of resuscitation. In the control group, MODS worsened from 1.138 to 1.727, whereas it improved from 1.367 to 0.8182 in the centhaquine group. ARDS acute respiratory distress syndrome, MODS multiple organ dysfunction syndrome

References

    1. Gulati A. Vascular endothelium and hypovolemic shock. Curr Vasc Pharmacol. 2016;14(2):187–195.
    1. Kobayashi L, Costantini TW, Coimbra R. Hypovolemic shock resuscitation. Surg Clin North Am. 2012;92(6):1403–1423.
    1. Cannon JW. Hemorrhagic shock. N Engl J Med. 2018;378(4):370–379.
    1. Kaufman EJ, Richmond TS, Wiebe DJ, Jacoby SF, Holena DN. Patient experiences of trauma resuscitation. JAMA Surg. 2017;152(9):843–850.
    1. Perel P, Roberts I, Ker K. Colloids versus crystalloids for fluid resuscitation in critically ill patients. Cochrane Database Syst Rev. 2013;2:CD000567.
    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. Causey MW, Miller S, Hoffer Z, Hempel J, Stallings JD, Jin G, et al. Beneficial effects of histone deacetylase inhibition with severe hemorrhage and ischemia-reperfusion injury. J Surg Res. 2013;184(1):533–540.
    1. Xu Y, Dai X, Zhu D, Xu X, Gao C, Wu C. An exogenous hydrogen sulphide donor, NaHS, inhibits the apoptosis signaling pathway to exert cardio-protective effects in a rat hemorrhagic shock model. Int J Clin Exp Pathol. 2015;8(6):6245–6254.
    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. Wolf A, Lusczek ER, Beilman GJ. Hibernation-based approaches in the treatment of hemorrhagic shock. Shock. 2018;50(1):14–23.
    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):1158–1171.
    1. Williams AT, Lucas A, Muller CR, Munoz C, Bolden-Rush C, Palmer AF, et al. Resuscitation from hemorrhagic shock with fresh and stored blood and polymerized hemoglobin. Shock. 2020;54:464–473.
    1. Gould SA, Moore EE, Moore FA, Haenel JB, Burch JM, Sehgal H, et al. Clinical utility of human polymerized hemoglobin as a blood substitute after acute trauma and urgent surgery. J Trauma. 1997;43(2):325–331.
    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. Levy JH, Goodnough LT, Greilich PE, Parr GV, Stewart RW, Gratz I, et al. Polymerized bovine hemoglobin solution as a replacement for allogeneic red blood cell transfusion after cardiac surgery: results of a randomized, double-blind trial. J Thorac Cardiovasc Surg. 2002;124(1):35–42.
    1. Moore EE, Moore FA, Fabian TC, Bernard AC, Fulda GJ, Hoyt DB, et al. Human polymerized hemoglobin for the treatment of hemorrhagic shock when blood is unavailable: the USA multicenter trial. J Am Coll Surg. 2009;208(1):1–13.
    1. Gulati A, Barve A, Sen AP. Pharmacology of hemoglobin therapeutics. J Lab Clin Med. 1999;133(2):112–119.
    1. Spahn DR, Kocian R. Artificial O2 carriers: status in 2005. Curr Pharm Des. 2005;11(31):4099–4114.
    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;5:CD003709.
    1. Al-Hesayen A, Parker JD. The effects of dobutamine on renal sympathetic activity in human heart failure. J Cardiovasc Pharmacol. 2008;51(5):434–436.
    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. Santry HP, Alam HB. Fluid resuscitation: past, present, and the future. Shock. 2010;33(3):229–241.
    1. Link RE, Desai K, Hein L, Stevens ME, Chruscinski A, Bernstein D, et al. Cardiovascular regulation in mice lacking alpha2-adrenergic receptor subtypes b and c. Science. 1996;273(5276):803–805.
    1. Muszkat M, Kurnik D, Solus J, Sofowora GG, Xie HG, Jiang L, et al. Variation in the alpha2B-adrenergic receptor gene (ADRA2B) and its relationship to vascular response in vivo. Pharmacogenet Genom. 2005;15(6):407–414.
    1. Gulati A, Jain D, Agrawal NR, Rahate P, Choudhuri R, Das S, et al. Resuscitative effect of centhaquine (Lyfaquin®) in hypovolemic shock patients: a randomized, multicentric, controlled trial. Adv Ther. 2021 doi: 10.1007/s12325-021-01760-4.
    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. Gulati A, Hussain G, Srimal RC. Effect of repeated administration of centhaquin, a centrally acting hypotensive drug, on adrenergic, cholinergic (Muscarinic), dopaminergic, and serotonergic receptors in brain-regions of rat. Drug Dev Res. 1991;23(4):307–323.
    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. 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. Kontouli Z, Staikou C, Iacovidou N, Mamais I, Kouskouni E, Papalois A, et al. Resuscitation with centhaquin and 6% hydroxyethyl starch 130/0.4 improves survival in a swine model of hemorrhagic shock: a randomized experimental study. Eur J Trauma Emerg Surg. 2019;45(6):1077–1085.
    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. Birkhahn RH, Gaeta TJ, Terry D, Bove JJ, Tloczkowski J. Shock index in diagnosing early acute hypovolemia. Am J Emerg Med. 2005;23(3):323–326.
    1. Rady MY, Rivers EP, Martin GB, Smithline H, Appelton T, Nowak RM. Continuous central venous oximetry and shock index in the emergency department: use in the evaluation of clinical shock. Am J Emerg Med. 1992;10(6):538–541.
    1. Huang YS, Chiu IM, Tsai MT, Lin CF, Lin CF. Delta shock index during emergency department stay is associated with in hospital mortality in critically ill patients. Front Med (Lausanne). 2021;8:648375.
    1. Goulden R, Hoyle MC, Monis J, Railton D, Riley V, Martin P, et al. qSOFA, SIRS and NEWS for predicting inhospital mortality and ICU admission in emergency admissions treated as sepsis. Emerg Med J. 2018;35(6):345–349.
    1. Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, et al. Assessment of clinical criteria for sepsis: for the third international consensus definitions for sepsis and septic shock (sepsis-3) JAMA. 2016;315(8):762–774.
    1. Peek GJ, Mugford M, Tiruvoipati R, Wilson A, Allen E, Thalanany MM, et al. Efficacy and economic assessment of conventional ventilatory support versus extracorporeal membrane oxygenation for severe adult respiratory failure (CESAR): a multicentre randomised controlled trial. Lancet. 2009;374(9698):1351–1363.
    1. Murray JF, Matthay MA, Luce JM, Flick MR. An expanded definition of the adult respiratory distress syndrome. Am Rev Respir Dis. 1988;138(3):720–723.
    1. Marshall JC, Cook DJ, Christou NV, Bernard GR, Sprung CL, Sibbald WJ. Multiple organ dysfunction score: a reliable descriptor of a complex clinical outcome. Crit Care Med. 1995;23(10):1638–1652.
    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. Jacob M, Sahu S, Singh YP, Mehta Y, Yang KY, Kuo SW, et al. A prospective observational study of rational fluid therapy in Asian intensive care units: another puzzle piece in fluid therapy. Indian J Crit Care Med. 2020;24(11):1028–1036.
    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. 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. El-Menyar A, Goyal P, Tilley E, Latifi R. The clinical utility of shock index to predict the need for blood transfusion and outcomes in trauma. J Surg Res. 2018;227:52–59.
    1. Marenco CW, Lammers DT, Morte KR, Bingham JR, Martin MJ, Eckert MJ. Shock index as a predictor of massive transfusion and emergent surgery on the modern battlefield. J Surg Res. 2020;16(256):112–118.
    1. Guyette F, Suffoletto B, Castillo JL, Quintero J, Callaway C, Puyana JC. Prehospital serum lactate as a predictor of outcomes in trauma patients: a retrospective observational study. J Trauma. 2011;70(4):782–786.
    1. Pan J, Peng M, Liao C, Hu X, Wang A, Li X. Relative efficacy and safety of early lactate clearance-guided therapy resuscitation in patients with sepsis: a meta-analysis. Medicine (Baltimore) 2019;98(8):e14453.
    1. Giancarelli A, Birrer KL, Alban RF, Hobbs BP, Liu-DeRyke X. Hypocalcemia in trauma patients receiving massive transfusion. J Surg Res. 2016;202(1):182–187.
    1. Ditzel RM, Jr, Anderson JL, Eisenhart WJ, Rankin CJ, DeFeo DR, Oak S, et al. A review of transfusion- and trauma-induced hypocalcemia: Is it time to change the lethal triad to the lethal diamond? J Trauma Acute Care Surg. 2020;88(3):434–439.
    1. Santacruz CA, Pereira AJ, Celis E, Vincent JL. Which multicenter randomized controlled trials in critical care medicine have shown reduced mortality? A systematic review. Crit Care Med. 2019;47(12):1680–1691.
    1. Divatia JV, Amin PR, Ramakrishnan N, Kapadia FN, Todi S, Sahu S, et al. Intensive care in India: the Indian intensive care case mix and practice patterns study. Indian J Crit Care Med. 2016;20(4):216–225.
    1. Bhandarkar P, Patil P, Soni KD, O'Reilly GM, Dharap S, Mathew J, et al. An analysis of 30-day in-hospital trauma mortality in four urban university hospitals using the Australia india trauma registry. World J Surg. 2021;45(2):380–389.
    1. Jiang S, Wu M, Lu X, Zhong Y, Kang X, Song Y, et al. Is restrictive fluid resuscitation beneficial not only for hemorrhagic shock but also for septic shock?: a meta-analysis. Medicine (Baltimore) 2021;100(12):e25143.
    1. Vignon P, Laterre PF, Daix T, Francois B. New agents in development for sepsis: any reason for hope? Drugs. 2020;80(17):1751–1761.
    1. Kalkwarf KJ, Cotton BA. Resuscitation for hypovolemic shock. Surg Clin N Am. 2017;97(6):1307–1321.
    1. Das SK, Choupoo NS, Ray S. Reducing vasopressor exposure in patients with vasodilatory hypotension. JAMA. 2020;324(9):897–898.

Source: PubMed

3
Sottoscrivi