| Home | E-Submission | Sitemap | Contact Us |  
Search
Clin Exp Emerg Med Search

CLOSE

Clin Exp Emerg Med > Volume 10(3); 2023 > Article
Suh, Shin, Kwon, Kim, Jo, Choi, Chung, Kim, and for the Korean Shock Society Investigators: Hemodynamic management of septic shock: beyond the Surviving Sepsis Campaign guidelines

Abstract

Although the Surviving Sepsis Campaign guidelines provide standardized and generalized guidance, they are less individualized. This review focuses on recent updates in the hemodynamic management of septic shock. Monitoring and intervention for septic shock should be personalized according to the phase of shock. In the salvage phase, fluid resuscitation and vasopressors should be given to provide life-saving tissue perfusion. During the optimization phase, tissue perfusion should be optimized. In the stabilization and de-escalation phases, minimal fluid infusion and safe fluid removal should be performed, respectively, while preserving organ perfusion. There is controversy surrounding the use of restrictive versus liberal fluid strategies after initial resuscitation. Fluid administration after initial resuscitation should depend upon the patient’s fluid responsiveness and requires individualized management. A number of dynamic tests have been proposed to monitor fluid responsiveness, which can help clinicians decide whether to give fluid or not. The optimal timing for the initiation of vasopressor agents is unknown. Recent data suggest that early vasopressor initiation should be considered. Inotropes can be considered in patients with decreased cardiac contractility associated with impaired tissue perfusion despite adequate volume status and arterial blood pressure. Venoarterial extracorporeal membrane oxygenation should be considered for refractory septic shock with severe cardiac systolic dysfunction.

INTRODUCTION

Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Septic shock is a subset of sepsis in which underlying circulatory, cellular, and metabolic abnormalities are profound enough to substantially increase mortality [1]. Despite advances in sepsis care, sepsis remains a major cause of morbidity and mortality worldwide, including in Korea. In 2017, an estimated 48.9 million sepsis cases were recorded worldwide, and 11.0 million sepsis-related deaths were reported globally, representing 19.7% of all global deaths [2,3].
To reduce the morbidity and mortality associated with sepsis, the Surviving Sepsis Campaign (SSC) guidelines were first launched in 2002 by the European Society of Intensive Care Medicine (ESICM), the International Sepsis Forum (ISF), and the Society of Critical Care Medicine (SCCM). Since then, these guidelines have been regularly updated based on new research findings and clinical experiences [4]. While the guidelines provide standardized and generalized guidance, they are supported by evidence mainly based on randomized controlled trials (RCTs) that investigated patient response to a single intervention. However, recent large RCT studies have failed to show a difference in mortality [57]. The reason for this is that these RCT studies did not consider the characteristics of individual patients that may affect their response to specific interventions. Given that sepsis is a complex condition with variable clinical courses, patient phenotypes, and treatment responses, a “one-size-fits-all” management approach may not be appropriate for all patients. In this respect, the evidence-based SSC guidelines appear to be less individualized. Future sepsis treatment should be individualized based on the diversity of sepsis. This narrative review focuses on recent updates in the personalized hemodynamic management of septic shock not covered in the SSC guidelines.

PERSONALIZED HEMODYNAMIC MANAGEMENT

Hemodynamic support remains a cornerstone in the management of septic shock. Different phases exist in the management of shock, including the salvage, optimization, stabilization, and de-escalation phases [8], and monitoring and intervention should be personalized and tailored according to the phase of shock (Fig. 1) [9].

Personalized hemodynamic monitoring

Salvage phase

In the salvage phase, the goal of treatment is to provide life-saving tissue perfusion. A mean arterial pressure (MAP) of ≥65 mmHg and diastolic arterial pressure (DAP) of ≥45 mmHg should be achieved. Clinical assessment can identify patients who may respond to fluids and assess their response [10]. Altered clinical signs, including hypotension, tachycardia or bradycardia, cold extremities, skin mottling, increased capillary refill time (CRT), and oliguria, are important warning signals indicating that tissue hypoperfusion is occurring, but these signs cannot reliably indicate whether the cardiac output (CO) is low or high nor indicate the source of the hemodynamic alteration [11]. For this purpose, physicians should perform additional evaluations, such as lactate measurements and echocardiography. If cardiac impairment is suspected or the patient fails to respond to fluid therapy, bedside echocardiography is the only useful tool for rapid estimation of cardiac dysfunction along with the identification of the cause of low CO. Blood lactate level measurement is also useful for identifying impairments in tissue perfusion [9].

Optimization phase

The primary goal during the optimization phase is to optimize tissue perfusion. In addition to the monitoring tools used in the salvage phase, central venous oxygen saturation (ScvO2) or mixed venous oxygen saturation (SvO2) and venous-to-arterial carbon dioxide difference (Pv-aCO2) measurement may be used to estimate tissue perfusion [9]. ScvO2 or SvO2 reflects the balance between the actual oxygen consumption and tissue oxygen delivery. A low ScvO2 indicates inadequate oxygen delivery if hemoglobin and arterial oxygen saturation values are within normal ranges [9]. Pv-aCO2, defined as the difference between the venous and arterial carbon dioxide partial pressures, is inversely related to CO. Increased Pv-aCO2 reflects decreased microvascular blood flow during early phases of resuscitation in septic shock [12]. It is important to note that there are differences in the normalization rate between monitoring tools. In an observational study, monitoring tools such as ScvO2, Pv-aCO2, and CRT were already normal in >70% of survivors at 6 hours, whereas lactate showed a much slower normalization rate, decreasing significantly at 6 hours compared to baseline but with only 52% of patients achieving normality at 24 hours [12]. Therefore, it is preferable to use several monitoring tools in combination rather than just a single one.
Transpulmonary thermodilution, an advanced monitoring tool, allows continuous and real-time monitoring of CO. It estimates the end-diastolic volume and systolic function of the four cardiac chambers. It also measures extravascular lung water (EVLW), which quantifies the volume of pulmonary edema, and pulmonary vascular permeability, which quantifies the degree of a pulmonary capillary leak [13,14]. Transpulmonary thermodilution should be considered in patients with severe septic shock.

Stabilization phase

In the stabilization phase, the goal is to preserve organ perfusion and prevent organ dysfunction. Cardiac dysfunction and volume overload are common in this stage, and hemodynamic tools already in use can continue to be used. In particular, repeated echocardiography may be helpful to uncover the development of right ventricular dysfunction [9].

De-escalation phase

Finally, in the de-escalation phase, the goal is to achieve a negative fluid balance by weaning patients off vasoactive drugs and promoting spontaneous polyuria or by inducing fluid clearance using diuretics or ultrafiltration. Monitoring can be minimized. Tissue perfusion and fluid responsiveness should be evaluated prior to fluid removal. When hypoperfusion occurs, de-escalation should be stopped [9].

Fluid management after initial resuscitation

For patients with sepsis-induced hypoperfusion or septic shock, the SSC guidelines suggest that ≥30 mL/kg of intravenous crystalloid fluid should be given within the first 3 hours of resuscitation [4]. This fixed volume during initial resuscitation was chosen mainly based on the results of several large RCT trials [57,1517].
However, the SSC guidelines suggest no recommendation for fluid administration in patients with sepsis and septic shock who still have signs of hypoperfusion and volume depletion after initial resuscitation and that fluid resuscitation should be given only if patients present with signs of hypoperfusion. The guidelines emphasize that fluid administration after the initial fluid bolus should be guided by perfusion parameters as well as a response in hemodynamic variables [4]. Liberal fluid administration may have detrimental effects by causing edema in vital organs, leading to organ dysfunction and impairment of oxygen delivery, but the restrictive fluid strategy primarily relies on vasopressors to reverse hypotension and maintain perfusion while limiting fluid administration [18]. Observational clinical studies and randomized trials have reported harmful effects, including kidney injury, respiratory failure, or high mortality. These studies suggest that a restrictive fluid strategy is potentially superior to a liberal fluid strategy [1923]. Recently, the results of two RCT studies related to restrictive versus liberal fluid strategies after initial resuscitation have been published. In the CLASSIC (Conservative vs. Liberal Approach to fluid therapy of Septic Shock in Intensive Care) trial [24], the restrictive fluid group received an intravenous fluids bolus of 250 to 500 mL if the patient had severe hypoperfusion, which was defined as a plasma lactate value of ≥4 mmol/L, a MAP of <50 mmHg despite infusion of a vasopressor or an inotropic agent, a mottling score >2 points (on a scale of 0–5 points, with higher scores indicating a greater area of mottling), or a urinary output of <0.1 mL/kg/hr during the first 2 hours after randomization. In the standard fluid group, no upper limit of fluid administration was set. The study found that intravenous fluid restriction did not cause fewer deaths at 90 days than standard intravenous fluid therapy. Separately, in the CLOVERS (Crystalloid Liberal or Vasopressors Early Resuscitation in Sepsis) trial [25], patients with sepsis-induced hypotension refractory to initial treatment with 1 to 3 L of intravenous fluid were enrolled. There was no difference in 90-day mortality or adverse outcomes between groups receiving the restrictive fluid strategy (prioritizing vasopressors and lower intravenous fluid volumes) and the liberal fluid strategy (prioritizing higher volumes of intravenous fluids before vasopressor use), respectively. These studies showed that restrictive fluid therapy is not superior to liberal fluid therapy. This means that fluid administration after initial resuscitation may vary depending on the patient’s fluid responsiveness and require individualized management. A comprehensive evaluation, including tissue perfusion monitoring, benefits and risks of fluid infusion, and fluid responsiveness, should be completed to achieve individualized fluid management, which should be preferred over a restrictive or liberal fluid strategy [13].

Tests to predict fluid responsiveness

The goal of fluid administration in patients with septic shock is to increase CO and tissue perfusion. However, fluid infusion can cause deleterious effects of fluid overload without an increase in CO. In an observational cohort study [26], only two-thirds of patients with septic shock were fluid-responders. Therefore, patients not responding to volume expansion may experience fluid overload [27]. Fluid overload has been shown to cause enhanced shedding of the endothelial glycocalyx, whose disruption increases vascular permeability, leading to tissue edema [28]. To prevent harmful effects of fluid overload, predicting fluid responsiveness should be the first step of a fluid strategy. Fluid responsiveness refers to a set of bedside tests that reversibly increase the preload status of the heart, allowing the clinician to assess whether this manipulation determines a significant increase in CO [29]. Fluid responsiveness is commonly defined as a stroke volume (SV) increase of ≥10% following a fluid bolus of 200 to 500 mL in 10 to 15 minutes [30,31]. For this purpose, static measurements of preloads, including central venous pressure, inferior vena cava diameter, and arterial pressure, have been used for decades but are unreliable. Strong evidence suggests that these traditional uses should be abandoned [3034]. In the last two decades, a number of dynamic tests have been proposed to establish and monitor fluid responsiveness (Table 1). These dynamic tests use heart-tolung interactions, passive leg raising, or mini-fluid challenges to induce short-term changes in cardiac preloads and reveal their effects on CO [30,35]. All have some limitations, but they are frequently complementary, which helps clinicians to make the decision to give fluid or not [30,35].
In 2018, an expert statement [36] proposed an individualized fluid treatment based on a repeated bolus of 250 to 500 mL of intravenous crystalloids with the continuous monitoring of fluid responsiveness and the early administration of vasopressors if circulation fails to improve. Since it is impractical to standardize the amount of fluid according to each patient, an individualized strategy of resuscitation based on fluid responsiveness is preferable.
On the other hand, fluid unresponsiveness could be used to safely remove fluids in the hemodynamically stable patient [37].

TIMING OF VASOACTIVE AGENT INITIATION IN SEPTIC SHOCK

Septic shock results in shedding of the vascular endothelial glycocalyx and endothelial damage, which leads to increased permeability, diffuse alterations in microvascular perfusion, and vasodilation due to a marked decrease in vascular tone [28]. Hypotension in patients with septic shock is known to be associated with increased mortality [37,38]. Vasoactive agents play a crucial role in septic shock management by modulating vascular tone and enhancing myocardial contractility. Vasoactive agents possess varying abilities to constrict or dilate blood vessels and enhance myocardial contractility (Fig. 2) [39]. The selection of vasoactive agents is tailored to the individual patient’s hemodynamic profile and specific needs to achieve optimal cardiovascular stability and tissue perfusion. The SSC guidelines recommend norepinephrine as a first-line vasopressor to maintain a target MAP of 65 mmHg for initial resuscitation [4]. Norepinephrine is both an α-1 and β-1 adrenergic agonist that predominantly enhances vascular filling pressure and redistributes blood flow via its vasoconstrictive effect and myocardial contractility [40]. In septic shock patients, a decrease in norepinephrine dose led to a more significant decrease in mean systemic pressure than a decrease in resistance to venous return, leading to a reduction in venous return [41].
Timely initiation of vasopressors with fluid resuscitation is a key component in the management of septic shock. However, the optimal timing for the initiation of vasopressors has not been known. There are no recommendations on the timing of vasoactive agent initiation for septic shock treatment in the SSC guidelines. Recent data showed an association between delayed therapy and increased mortality and suggested that early initiation of vasopressors should be considered [37,42,43]. The 2018 SSC hour-1 bundle, which recommends vasopressor therapy within the first hour during or after volume resuscitation if blood pressure is not restored after initial fluid resuscitation to achieve a MAP of ≥65 mmHg [44]. In a retrospective study, every 1-hour delay in norepinephrine initiation during the first 6 hours after septic shock onset was associated with a 5.3% increase in mortality. Mortality rates at 28 days were significantly higher when norepinephrine administration was started ≥2 hours after septic shock onset compared to <2 hours [42]. A very early start of vasopressors within/before the next hour of the first resuscitative fluid load was related to a significant lower net fluid balance and also with a significant reduction in the risk of death at 28 days [45].
Early high-dose vasopressor within the first 6 hours of shock is associated with lower mortality [46]. In a systematic review and meta-analysis, early initiation of norepinephrine in patients with septic shock was associated with decreased short-term mortality, a shorter time to achieve the target MAP, and a smaller volume of intravenous fluids within 6 hours [47]. In the CENSER (Early Use of Norepinephrine in Septic Shock Resuscitation) trial [48], a single-center, prospective, double-blind, placebo-controlled trial, the early vasopressor group received norepinephrine at 1.5 hours compared to 3 hours in the standard treatment group. The shock control rate at 6 hours, which was the primary endpoint, was met in 76.1% of patients in the early vasopressor group compared to 48.4% of patients in the standard group (P<0.001), while there was no difference in 28-day mortality between these groups.
In contrast, earlier vasopressor use with a restrictive fluid strategy compared to later vasopressor use with a liberal fluid strategy did not result in significantly lower (or higher) mortality before discharge home by day 90 [25]. Similarly, vasopressor initiation within 1 hour of fluid loading was associated with higher 28-day mortality in patients with septic shock [49].
DAP and the diastolic shock index (DSI), defined as the ratio between heart rate and DAP, may be used to guide the timing of vasopressor initiation in septic shock. It seems logical to initiate vasopressors when DAP <45 mmHg or DSI >2, which indicates severe vasodilation [50]. A retrospective observational study showed that in patients with high DSIs (≥2.0) and high lactate levels (≥2.5 mmol/L), early initiation of vasopressor therapy was associated with decreased 28-day mortality [51]. These data suggest that norepinephrine should be initiated early, ideally within 1 hour of shock onset, but after adequate fluid resuscitation. DSI and lactate measurement can help guide the appropriate time to initiate vasopressor therapy in septic shock [52].
The SSC guidelines suggest adding vasopressin instead of escalating the dose of norepinephrine in adults with septic shock on norepinephrine with inadequate MAP levels. However, the timing of vasopressin initiation is not well-described in the literature. In VASST (Vasopressin and Septic Shock Trial) [53], there was no difference in 28-day mortality, but subgroup analyses identified a mortality benefit with the use of vasopressin in patients with less severe septic shock, i.e., those with a norepinephrine dose at randomization of ≤15 μg/min and those with a lactate concentration at randomization of ≤1.4 mmol/L. In a retrospective, observational study, a greater norepinephrine-equivalent dose at vasopressin initiation and a higher lactate concentration at vasopressin initiation were each associated with higher in-hospital mortality in patients with septic shock [54]. These data indicate that vasopressin should be initiated when patients are on low norepinephrine-equivalent doses or have low lactate concentrations. While the SSC guidelines suggest vasopressin initiation when the norepinephrine dose is in the range of 0.25 to 0.5 μg/kg/min [4], vasopressin initiation may be considered before norepinephrine-equivalent doses exceed 0.1 to 0.2 μg/kg/min (10–15 μg/min) [52].
Epinephrine should be considered as a third-line treatment for septic shock, and its use should be limited to those patients with inadequate MAP levels despite norepinephrine and vasopressin administration [4]. The specific norepinephrine-equivalent dose at which epinephrine should be administered in septic shock is unknown. One study [55] identified the optimal norepinephrine-equivalent dose range for initiating epinephrine as 37 to 133 μg/min. In this dose range, 29% of patients achieved hemodynamic stability with the initiation of epinephrine, while 15% of patients who had epinephrine initiated outside of this dose range achieved hemodynamic stability (P=0.03).

INOTROPES

Sepsis-induced cardiomyopathy (SCM) is a reversible myocardial dysfunction caused by sepsis. The prevalence of SCM varies from 10% to 70%, although studies defining SCM as an ejection fraction of <45% have generally reported a prevalence of 30% to 50% [56]. Inotropes can be considered in patients with decreased cardiac contractility associated with impaired tissue perfusion. The SSC guidelines suggest either adding dobutamine to norepinephrine or administering epinephrine alone for adults with septic shock and cardiac dysfunction with persistent hypoperfusion despite adequate volume status and arterial blood pressure [4]. Adverse effects (tachyarrhythmia, increased heart rate, hypotension, and myocardial oxygen consumption) and specific risks (hypertrophic cardiomyopathy, myocardial ischemia) should be carefully investigated, and the risk/benefit profile of intervention should be evaluated [9]. Milrinone is a phosphodiesterase inhibitor that increases intracellular cyclic adenosine monophosphate, leading to inotropic effects independent of β-adrenergic receptors [15]. Milrinone may be an effective therapeutic in patients recently on β-blockers [57]. Experts suggest adopting a stepwise approach to administering inotropics, as follows. First, begin with a limited dose of dobutamine (2.5–5.0 μg/kg/min) and evaluate efficacy and tolerance. If there is still severe contractility impairment, higher doses (≤20 μg/kg/min) may be considered. Second, substitute or add enoximone or milrinone and evaluate the efficacy and tolerance. Third, substitute or add levosimendan in cases of severe impairment. At each step, improvements in cardiac function and CO as well as resolution of tissue hypoperfusion and tolerance (e.g., lack of tachycardia, arrhythmias, etc.) should be evaluated. As soon as the situation improves, weaning off inotropics should be attempted [9]. However, the SSC guidelines suggest against using levosimendan, as it was not superior to dobutamine in adults with sepsis in terms of mortality [4,58].

TIMING OF CORTICOSTEROID INITIATION IN SEPTIC SHOCK

Sepsis results in disruption of the hypothalamic-pituitary-adrenal axis, which may translate into cardiovascular and other organ dysfunction and eventually an increased risk of death. Corticosteroids are known to improve cardiovascular function via sodium and water retention, restore systemic vascular resistance, and decrease organ failure [59]. Three recent large RCTs [6062] showed that corticosteroids accelerate the resolution of shock, but there was no clear effect on short- or long-term mortality. The SSC guidelines suggest administering intravenous corticosteroids in patients with septic shock and ongoing requirements for vasopressor therapy [4].
Although there is no clear recommendation with regard to the time of initiation of corticosteroids in septic shock patients, the early initiation of corticosteroid therapy in sepsis, specifically within 24 hours of shock, despite adequate fluid resuscitation and vasopressor administration (norepinephrine-equivalent dose of 0.5–1 μg/kg/min) is reasonable [52]. A retrospective cohort study reported decreased intensive care unit mortality when hydrocortisone was administered within 0 to 6 hours after shock onset compared to >48 hours after shock onset (odds ratio, 0.6; 95% confidence interval 0.4–0.8) and suggested that hydrocortisone should be started within the first 12 hours after shock onset [63].
A recent multicenter, propensity score-weighted observational cohort study (n=198) [64] evaluated early (≤12 hours of vasopressor initiation) versus late (>12 hours of vasopressor initiation) low-dose corticosteroid initiation in septic shock and determined that early initiation was associated with a shorter time to vasopressor discontinuation (40.7 hours vs. 60.6 hours, P=0.0002). The SSC guidelines suggest that corticosteroid administration is commenced ≥4 hours after vasopressor initiation and at norepinephrine or epinephrine doses of ≥0.25 μg/kg/min [4].

VA ECMO IN SEPTIC SHOCK

While the SSC guidelines suggest using venovenous extracorporeal membrane oxygenation (ECMO) when conventional mechanical ventilation fails for sepsis-induced severe acute respiratory distress syndrome, there is no suggestion of deploying venoarterial (VA) ECMO in septic shock complicated by SCM [4]. Most early studies of VA ECMO for refractory septic shock complicated by SCM reported low survival rates and poor outcomes [65]. A recent retrospective, multicenter study [66] showed that patients with severe sepsis-induced cardiogenic shock treated with VA ECMO experienced a large and significant improvement in survival compared to controls not receiving ECMO (60% vs. 25%, P<0.0001). A meta-analysis of 468 patients placed on VA ECMO for refractory septic shock [67] reported an overall survival rate of 36% and a significantly higher survival rate among patients with ejection fractions of <20% compared to those with ejection fractions of >35% (62% vs. 32.1%, P=0.05). Therefore, VA ECMO should be considered as a bridge therapy to recovery for patients with refractory septic shock with severe cardiac systolic dysfunction and end-organ hypoperfusion. However, VA ECMO should not be used to manage patients with isolated vasodilatory septic shock without significant myocardial dysfunction [65].

CONCLUSION

Sepsis is a complex condition with variable clinical courses, patient phenotypes, and treatment responses. Personalized hemodynamic monitoring and fluid responsiveness based on the phase of septic shock are essential in septic shock management to assess the patient’s cardiovascular status, guide fluid resuscitation, determine the need and timing for vasopressors and inotropic agents, and optimize tissue perfusion, which leads to improved outcomes in septic shock.

NOTES

ETHICS STATEMENT
Not applicable.
CONFLICT OF INTEREST
No potential conflict of interest relevant to this article was reported.
FUNDING
None.
AUTHOR CONTRIBUTIONS
Conceptualization: GJS; Data curation: GJS; Methodology: all authors; Visualization: all authors; Writing–original draft: GJS; Writing–review & editing: all authors. All authors have read and approved the final manuscript.

REFERENCES

1. Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA 2016; 315:801-10.
pmid pmc
2. Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet 2020; 395:200-11.
crossref pmid pmc
3. Namgung M, Ahn C, Park Y, Kwak IY, Lee J, Won M. Mortality among adult patients with sepsis and septic shock in Korea: a systematic review and meta-analysis. Clin Exp Emerg Med 2023; 10:157-71.
crossref pmid pmc pdf
4. Evans L, Rhodes A, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Intensive Care Med 2021; 47:1181-247.
pmid pmc
5. Mouncey PR, Osborn TM, Power GS, et al. Trial of early, goal-directed resuscitation for septic shock. N Engl J Med 2015; 372:1301-11.
crossref pmid
6. ARISE Investigators; ANZICS Clinical Trials Group, Peake SL, et al. Goal-directed resuscitation for patients with early septic shock. N Engl J Med 2014; 371:1496-506.
crossref pmid
7. ProCESS Investigators, Yealy DM, Kellum JA, et al. A randomized trial of protocol-based care for early septic shock. N Engl J Med 2014; 370:1683-93.
crossref pmid pmc
8. Vincent JL, De Backer D. Circulatory shock. N Engl J Med 2013; 369:1726-34.
crossref pmid
9. De Backer D, Cecconi M, Chew MS, et al. A plea for personalization of the hemodynamic management of septic shock. Crit Care 2022; 26:372.
crossref pmid pmc pdf
10. Cecconi M, Hernandez G, Dunser M, et al. Fluid administration for acute circulatory dysfunction using basic monitoring: narrative review and expert panel recommendations from an ESICM task force. Intensive Care Med 2019; 45:21-32.
crossref pmid pdf
11. De Backer D, Vieillard-Baron A. Clinical examination: a trigger but not a substitute for hemodynamic evaluation. Intensive Care Med 2019; 45:269-71.
crossref pmid pdf
12. Hernandez G, Luengo C, Bruhn A, et al. When to stop septic shock resuscitation: clues from a dynamic perfusion monitoring. Ann Intensive Care 2014; 4:30.
crossref pmid pmc pdf
13. De Backer D, Aissaoui N, Cecconi M, et al. How can assessing hemodynamics help to assess volume status? Intensive Care Med 2022; 48:1482-94.
crossref pmid pmc pdf
14. Monnet X, Teboul JL. Transpulmonary thermodilution: advantages and limits. Crit Care 2017; 21:147.
crossref pmid pmc pdf
15. Rhodes A, Evans LE, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med 2017; 43:304-77.
pmid
16. PRISM Investigators, Rowan KM, Angus DC, et al. Early, goal-directed therapy for septic shock: a patient-level meta-analysis. N Engl J Med 2017; 376:2223-34.
crossref pmid
17. Kuttab HI, Lykins JD, Hughes MD, et al. Evaluation and predictors of fluid resuscitation in patients with severe sepsis and septic shock. Crit Care Med 2019; 47:1582-90.
crossref pmid pmc
18. Self WH, Semler MW, Bellomo R, et al. Liberal versus restrictive intravenous fluid therapy for early septic shock: rationale for a randomized trial. Ann Emerg Med 2018; 72:457-66.
crossref pmid pmc
19. Hjortrup PB, Haase N, Bundgaard H, et al. Restricting volumes of resuscitation fluid in adults with septic shock after initial management: the CLASSIC randomised, parallel-group, multicentre feasibility trial. Intensive Care Med 2016; 42:1695-705.
crossref pmid pdf
20. Richard JC, Bayle F, Bourdin G, et al. Preload dependence indices to titrate volume expansion during septic shock: a randomized controlled trial. Crit Care 2015; 19:5.
crossref pmid pmc pdf
21. Andrews B, Semler MW, Muchemwa L, et al. Effect of an early resuscitation protocol on in-hospital mortality among adults with sepsis and hypotension: a randomized clinical trial. JAMA 2017; 318:1233-40.
crossref pmid pmc
22. Silversides JA, Major E, Ferguson AJ, et al. Conservative fluid management or deresuscitation for patients with sepsis or acute respiratory distress syndrome following the resuscitation phase of critical illness: a systematic review and meta-analysis. Intensive Care Med 2017; 43:155-70.
crossref pmid pdf
23. Maitland K, Kiguli S, Opoka RO, et al. Mortality after fluid bolus in African children with severe infection. N Engl J Med 2011; 364:2483-95.
crossref pmid
24. Meyhoff TS, Hjortrup PB, Wetterslev J, et al. Restriction of intravenous fluid in ICU patients with septic shock. N Engl J Med 2022; 386:2459-70.
pmid
25. National Heart, Lung, and Blood Institute Prevention and Early Treatment of Acute Lung Injury Clinical Trials Network, Shapiro NI, Douglas IS, et al. Early restrictive or liberal fluid management for sepsis-induced hypotension. N Engl J Med 2023; 388:499-510.
crossref pmid pmc
26. Leisman DE, Doerfler ME, Schneider SM, Masick KD, D’Amore JA, D’Angelo JK. Predictors, prevalence, and outcomes of early crystalloid responsiveness among initially hypotensive patients with sepsis and septic shock. Crit Care Med 2018; 46:189-98.
crossref pmid
27. Guarracino F, Bertini P, Pinsky MR. Heterogeneity of cardiovascular response to standardized sepsis resuscitation. Crit Care 2020; 24:99.
crossref pmid pmc pdf
28. Chappell D, Bruegger D, Potzel J, et al. Hypervolemia increases release of atrial natriuretic peptide and shedding of the endothelial glycocalyx. Crit Care 2014; 18:538.
crossref pmid pmc pdf
29. Kattan E, Castro R, Miralles-Aguiar F, Hernandez G, Rola P. The emerging concept of fluid tolerance: a position paper. J Crit Care 2022; 71:154070.
crossref pmid
30. Monnet X, Marik PE, Teboul JL. Prediction of fluid responsiveness: an update. Ann Intensive Care 2016; 6:111.
crossref pmid pmc pdf
31. Monnet X, Teboul JL. My patient has received fluid. How to assess its efficacy and side effects? Ann Intensive Care 2018; 8:54.
crossref pmid pmc pdf
32. Marik PE, Cavallazzi R. Does the central venous pressure predict fluid responsiveness? An updated meta-analysis and a plea for some common sense. Crit Care Med 2013; 41:1774-81.
crossref pmid
33. Marik PE, Baram M, Vahid B. Does central venous pressure predict fluid responsiveness?: a systematic review of the literature and the tale of seven mares. Chest 2008; 134:172-8.
crossref pmid
34. Reddi B, Finnis M, Udy AA, et al. The relationship between the change in central venous pressure and intravenous fluid volume in patients presenting to the emergency department with septic shock. Intensive Care Med 2018; 44:1591-2.
crossref pmid pdf
35. Monnet X, Shi R, Teboul JL. Prediction of fluid responsiveness: what’s new? Ann Intensive Care 2022; 12:46.
crossref pmid pmc pdf
36. Perner A, Cecconi M, Cronhjort M, et al. Expert statement for the management of hypovolemia in sepsis. Intensive Care Med 2018; 44:791-8.
crossref pmid pdf
37. Bakker J, Kattan E, Annane D, et al. Current practice and evolving concepts in septic shock resuscitation. Intensive Care Med 2022; 48:148-63.
crossref pmid pdf
38. Maheshwari K, Nathanson BH, Munson SH, et al. The relationship between ICU hypotension and in-hospital mortality and morbidity in septic patients. Intensive Care Med 2018; 44:857-67.
crossref pmid pmc pdf
39. Jentzer JC, Coons JC, Link CB, Schmidhofer M. Pharmacotherapy update on the use of vasopressors and inotropes in the intensive care unit. J Cardiovasc Pharmacol Ther 2015; 20:249-60.
crossref pmid pdf
40. Hernandez G, Teboul JL, Bakker J. Norepinephrine in septic shock. Intensive Care Med 2019; 45:687-9.
crossref pmid pdf
41. Persichini R, Silva S, Teboul JL, et al. Effects of norepinephrine on mean systemic pressure and venous return in human septic shock. Crit Care Med 2012; 40:3146-53.
crossref pmid
42. Bai X, Yu W, Ji W, et al. Early versus delayed administration of norepinephrine in patients with septic shock. Crit Care 2014; 18:532.
crossref pmid pmc pdf
43. Vincent JL, Nielsen ND, Shapiro NI, et al. Mean arterial pressure and mortality in patients with distributive shock: a retrospective analysis of the MIMIC-III database. Ann Intensive Care 2018; 8:107.
crossref pmid pmc pdf
44. Levy MM, Evans LE, Rhodes A. The surviving sepsis campaign bundle: 2018 update. Intensive Care Med 2018; 44:925-8.
crossref pmid pdf
45. Ospina-Tascon GA, Hernandez G, Alvarez I, et al. Effects of very early start of norepinephrine in patients with septic shock: a propensity score-based analysis. Crit Care 2020; 24:52.
crossref pmid pmc pdf
46. Roberts RJ, Miano TA, Hammond DA, et al. Evaluation of vasopressor exposure and mortality in patients with septic shock. Crit Care Med 2020; 48:1445-53.
pmid
47. Li Y, Li H, Zhang D. Timing of norepinephrine initiation in patients with septic shock: a systematic review and meta-analysis. Crit Care 2020; 24:488.
crossref pmid pmc pdf
48. Permpikul C, Tongyoo S, Viarasilpa T, Trainarongsakul T, Chakorn T, Udompanturak S. Early use of norepinephrine in septic shock resuscitation (CENSER): a randomized trial. Am J Respir Crit Care Med 2019; 199:1097-105.
crossref pmid
49. Yeo HJ, Lee YS, Kim TH, et al. Vasopressor initiation within 1 hour of fluid loading is associated with increased mortality in septic shock patients: analysis of national registry data. Crit Care Med 2022; 50:e351-60.
crossref pmid
50. Ospina-Tascon GA, Teboul JL, Hernandez G, et al. Diastolic shock index and clinical outcomes in patients with septic shock. Ann Intensive Care 2020; 10:41.
crossref pmid pmc pdf
51. Kim DS, Park JE, Hwang SY, et al. Prediction of vasopressor requirement among hypotensive patients with suspected infection: usefulness of diastolic shock index and lactate. Clin Exp Emerg Med 2022; 9:176-86.
crossref pmid pmc pdf
52. Ammar MA, Ammar AA, Wieruszewski PM, et al. Timing of vasoactive agents and corticosteroid initiation in septic shock. Ann Intensive Care 2022; 12:47.
crossref pmid pmc pdf
53. Russell JA, Walley KR, Singer J, et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N Engl J Med 2008; 358:877-87.
crossref pmid
54. Sacha GL, Lam SW, Wang L, Duggal A, Reddy AJ, Bauer SR. Association of catecholamine dose, lactate, and shock duration at vasopressin initiation with mortality in patients with septic shock. Crit Care Med 2022; 50:614-23.
crossref pmid
55. Ammar MA, Limberg EC, Lam SW, et al. Optimal norepinephrine-equivalent dose to initiate epinephrine in patients with septic shock. J Crit Care 2019; 53:69-74.
crossref pmid
56. Hollenberg SM, Singer M. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol 2021; 18:424-34.
crossref pmid pdf
57. Russell JA. Vasopressor therapy in critically ill patients with shock. Intensive Care Med 2019; 45:1503-17.
crossref pmid pdf
58. Bhattacharjee S, Soni KD, Maitra S, Baidya DK. Levosimendan does not provide mortality benefit over dobutamine in adult patients with septic shock: a meta-analysis of randomized controlled trials. J Clin Anesth 2017; 39:67-72.
crossref pmid
59. Annane D. The role of ACTH and corticosteroids for sepsis and septic shock: an update. Front Endocrinol (Lausanne) 2016; 7:70.
crossref pmid pmc
60. Annane D, Renault A, Brun-Buisson C, et al. Hydrocortisone plus fludrocortisone for adults with septic shock. N Engl J Med 2018; 378:809-18.
crossref pmid
61. Gordon AC, Mason AJ, Thirunavukkarasu N, et al. Effect of early vasopressin vs norepinephrine on kidney failure in patients with septic shock: the VANISH randomized clinical trial. JAMA 2016; 316:509-18.
crossref pmid
62. Venkatesh B, Finfer S, Cohen J, et al. Adjunctive glucocorticoid therapy in patients with septic shock. N Engl J Med 2018; 378:797-808.
crossref pmid
63. Sacha GL, Chen AY, Palm NM, Duggal A. Evaluation of the initiation timing of hydrocortisone in adult patients with septic shock. Shock 2021; 55:488-94.
crossref pmid
64. Ragoonanan D, Allen B, Cannon C, Rottman-Pietrzak K, Bello A. Comparison of early versus late initiation of hydrocortisone in patients with septic shock in the ICU setting. Ann Pharmacother 2022; 56:264-70.
crossref pmid pdf
65. Helwani MA, Lim A. Is venoarterial extracorporeal membrane oxygenation an option for managing septic shock. Curr Opin Anaesthesiol 2023; 36:45-9.
crossref pmid
66. Brechot N, Hajage D, Kimmoun A, et al. Venoarterial extracorporeal membrane oxygenation to rescue sepsis-induced cardiogenic shock: a retrospective, multicentre, international cohort study. Lancet 2020; 396:545-52.
crossref pmid
67. Ling RR, Ramanathan K, Poon WH, et al. Venoarterial extracorporeal membrane oxygenation as mechanical circulatory support in adult septic shock: a systematic review and meta-analysis with individual participant data meta-regression analysis. Crit Care 2021; 25:246.
crossref pmid pmc pdf

Fig. 1.
Hemodynamic monitoring, targets, and interventions at the different phases of shock. CRT, capillary refill time; MAP, mean arterial pressure; DAP, diastolic arterial pressure; SvO2, mixed venous oxygen saturation; ScvO2, central venous oxygen saturation; Pv-aCO2, venous-to-arterial carbon dioxide difference; TPTD, transpulmonary thermodilution; EVLW, extravascular lung water.
ceem-23-065f1.jpg
Fig. 2.
Vasoactive agents and their effects.
ceem-23-065f2.jpg
Table 1.
Tests predicting fluid responsiveness
Test Advantage Limitation Threshold
PPV/SVV Requires no maneuver Cannot be used in case of spontaneous breathing, cardiac arrhythmias, low Vt/lung compliance ≥ 12%
PLR No fluid infusion Requires a direct measurement of CO/SV CO ≥ 10%
Works regardless of breathing activity, cardiac rhythm, Vt, lung compliance
EEO test Easy to perform Requires a direct measurement of CO/SV CO ≥ 5%
Works regardless of breathing activity, cardiac rhythm, Vt, lung compliance Requires mechanical ventilation
Cannot be used in patients who cannot tolerate a 15-sec respiratory hold
Vt challenge Requires no measurement in CO/SV Requires mechanical ventilation PPV ≥ 1.0%–3.5%
Reliable in prone position and in spontaneous breathing under mechanical ventilation
IVC diameter variation Requires no measurement in CO/SV Cannot be used in spontaneous breathing, low Vt/lung compliance ≥ 12%
SVC diameter variation Requires no measurement in CO/SV Cannot be used in spontaneous breathing, low Vt/lung compliance ≥ 12%–36%
Requires transesophageal Doppler
Mini-fluid challenge Easy to perform Requires a precise technique for measuring CO CO ≥ 5%
Works regardless of breathing activity, cardiac rhythm, Vt, lung compliance, IAH Requires fluid infusion VTI ≥ 10%
Trendelenburg maneuver No fluid infusion Possible gastric reflux CO ≥ 8%–10%
Possible in prone position, ECMO Requires more validation

PPV, pulse pressure variation; SVV, stroke volume variation; Vt, tidal volume; PLR, passive leg raising; CO, cardiac output; SV, stroke volume; EEO, end-expiratory occlusion; IVC, inferior vena cava; SVC, superior vena cava; IAH, intra-abdominal hypertension; VTI, velocity time integral; ECMO, extracorporeal membrane oxygenation.

TOOLS
PDF Links  PDF Links
PubReader  PubReader
ePub Link  ePub Link
XML Download  XML Download
Full text via DOI  Full text via DOI
Download Citation  Download Citation
  Print
Share:      
METRICS
1
Crossref
0
Scopus
13,561
View
6,571
Download
Editorial Office
The Korean Society of Emergency Medicine
101-3104, Brownstone Seoul, 464 Cheongpa-ro, Jung-gu, Seoul 04510, Korea
TEL: +82-31-709-0918   E-mail: office@ceemjournal.org
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Copyright © by The Korean Society of Emergency Medicine.                 Developed in M2PI