Your activity: 4022 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Intraoperative fluid management

Intraoperative fluid management
Author:
Girish P Joshi, MB, BS, MD, FFARCSI
Section Editor:
Michael F O'Connor, MD, FCCM
Deputy Editor:
Nancy A Nussmeier, MD, FAHA
Literature review current through: Feb 2022. | This topic last updated: Feb 07, 2022.

INTRODUCTION — Perioperative maintenance of adequate intravascular volume status is important to achieve optimal outcomes after surgery, but there are controversies regarding both composition and volume of intraoperative fluid therapy. This topic will review derangements and monitoring of intravascular volume status in this setting, as well as strategies for choosing appropriate composition, amount, and timing of intraoperative fluid administration.

Severe intravascular volume depletion in surgical patients is discussed in other topics. (See "Intraoperative management of shock in adults", section on 'Hypovolemic shock management' and "Massive blood transfusion".)

Routine management of maintenance and replacement fluids in nonsurgical settings is discussed separately. (See "Maintenance and replacement fluid therapy in adults".)

CAUSES OF INTRAVASCULAR VOLUME DERANGEMENTS

Preoperative factors

Preoperative fasting overnight for approximately 10 hours does not significantly reduce intravascular volume [1,2]. Nevertheless, preoperative dehydration should be avoided by limiting the period of fasting [3], and encouraging patients to consume clear oral liquids up to two hours before surgery. (See "Preoperative fasting in adults".)

Mechanical bowel preparation may be associated with fluid loss from the gastrointestinal tract, which may reduce preoperative intravascular volume.

Disorders such as bowel obstruction or pancreatitis may cause intravascular volume loss due to inflammation and interstitial edema. (See "Etiology, clinical manifestations, and diagnosis of volume depletion in adults", section on 'Etiology'.)

Ongoing bleeding typically requires surgical hemostasis in order to allow adequate volume repletion. (See "Initial management of NON-hemorrhagic shock in adult trauma".)

Anesthesia-related factors

Most anesthetic and adjuvant drugs cause dose-dependent vasodilation and myocardial depression that may lead to hypotension [4,5]. Avoiding unnecessarily deep anesthesia avoids hypotension, which in turn avoids otherwise unnecessary fluid administration. (See "Hemodynamic management during anesthesia in adults", section on 'Selection and dosing of anesthetic agents'.)

Hypotension that persists after reduction in anesthetic depth (if appropriate) and administration of fluid to replace known surgical losses can be treated with intravenous vasopressor/inotropic agents such as phenylephrine. (See 'Restrictive (zero-balance) strategy' below and "Hemodynamic management during anesthesia in adults", section on 'Vasopressor and positive inotropic agents'.)

Sympathetic blockade during neuraxial anesthesia can result in relative hypovolemia due to increased venous capacitance and dilation of arteriolar resistance vessels, with resultant hypotension. Rather than preloading with IV fluid, which is a traditional practice, use of a vasopressor such as phenylephrine or norepinephrine is preferred.  

Surgery-related factors

Hemorrhage.

Coagulopathy due to hemodilution and/or hypothermia, which aggravates blood loss.

Decreased venous return due to:

Abdominal insufflation during laparoscopy.  

Compression of the inferior vena cava or other major veins (eg, portal vein).

Positive pressure ventilation with large tidal volumes, recruitment maneuvers, or positive end-expiratory pressure (ie, ventilation with a high level of continuous positive airway pressure to reverse atelectasis) during mechanical ventilation. (See "Mechanical ventilation during anesthesia in adults", section on 'Recruitment maneuvers'.)

Prolonged operative time, particularly with an open abdominal cavity, may eventually lead to increased bowel edema and sequestration of fluid [6]. However, during short less invasive procedures there is minimal evaporative or insensible fluid loss from exposed body cavities or wounds and minimal sequestration of fluids in tissues (<1 mL/kg per hour) [7].

Factors preventing an early transition from intravenous to oral fluid therapy within 24 hours of surgery [3].

CONSEQUENCES OF INTRAVASCULAR VOLUME DERANGEMENTS — Maintenance of intravascular euvolemia throughout the perioperative period is ideal. Both hypovolemia and hypervolemia are associated with increased postoperative morbidity [8-11].

Hypovolemia — Absolute or relative hypovolemia is common in the perioperative period due to preoperative dehydration, vasodilation caused by anesthetic and adjuvant drugs, and surgical bleeding. (See 'Causes of intravascular volume derangements' above.)

Preoperative hypovolemia may increase the risk of significant decreases in blood pressure during induction of anesthesia [12]. Persistent hypovolemia leads to low cardiac output and decreased tissue perfusion and, if severe, can lead to shock and multiorgan failure [13,14].

Hypervolemia — The most common cause of perioperative hypervolemia is retention of fluid administered during surgery. Clinically significant postoperative fluid retention (ie, weight gain >10 percent above preoperative baseline) has been associated with increased morbidity, length of stay in the intensive care unit, and mortality [8,15]. In critically ill surgical patients, this association may be spurious since fluid may be administered to treat hemodynamic instability (from increased vascular permeability) or bleeding, factors that are independently associated with poor outcome. However, tissue edema has independent deleterious effects on various organ systems, even in patients who remain hemodynamically stable [16]. These include:

Respiratory effects – Increased extravascular fluid in the lung tissue can impair oxygen exchange and increase risk for postoperative respiratory failure and pneumonia [16]. Some patients develop frank pulmonary edema, particularly those with a history of heart failure [17]. (See "Overview of the management of postoperative pulmonary complications", section on 'Pulmonary edema'.)

Gastrointestinal effects – Increased extracellular fluid in the bowel can lead sequentially to gastrointestinal edema, decreased gastrointestinal motility, and possibly ileus [18]. In patients undergoing bowel surgery, intestinal edema can increase tension at bowel anastomoses and may contribute to anastomotic dehiscence [16].

Occasionally, massive fluid resuscitation may be associated with acute ascites [19]. Ascites and bowel edema can contribute to development of abdominal compartment syndrome [20]. (See "Abdominal compartment syndrome in adults".)

Coagulation – Excess intravascular fluid may dilute clotting factors, which can contribute to or cause coagulation abnormalities. (See "Massive blood transfusion", section on 'Alterations in hemostasis'.)

Wound healing – Marked tissue edema can impair wound healing. (See "Basic principles of wound healing".)

MONITORING INTRAVASCULAR VOLUME STATUS

Goals and challenges — The purpose of monitoring intravascular volume status is to guide fluid administration in order to maintain tissue perfusion. Hypovolemia typically reduces tissue perfusion. However, tissue edema can also reduce tissue perfusion (eg, excessive volume administration or compensatory fluid retention in a patient with severe myocardial dysfunction).

Intraoperative monitoring with standard hemodynamic measurements (eg, noninvasive blood pressure [BP] and heart rate [HR]) and in selected cases, with one or more invasive monitors of dynamic hemodynamic parameters, can be used to predict fluid responsiveness (ie, increase in stroke volume following intravenous fluid administration (figure 1)) [21]. Laboratory values may be obtained, but these do not immediately reflect acute changes in volume status. Other clinical methods routinely used to assess volume status in the awake state are not available in the anesthetized patient (eg, thirst, postural dizziness, lethargy, confusion).

Regardless of the monitors employed, intraoperative determination of intravascular volume status is challenging because of continuously changing cardiovascular responses to anesthetic drugs, variable surgical volume losses that are difficult to quantify, and a preoperative volume status that may be suboptimal or unknown.

Static (traditional) parameters — Static parameters (eg, measurements of BP, HR, urine output [UO], and central venous pressure [CVP]) have been traditionally used to provide supplemental data regarding intravascular volume status (see "Monitoring during anesthesia", section on 'Monitoring modalities'). However, sole use of these parameters to guide fluid therapy may result in either hypovolemia or hypervolemia, and significant intraoperative reduction in tissue perfusion may not be recognized even with continuous monitoring of these static parameters due to the following limitations [13,14,22]:

Blood pressure and heart rate – BP and HR responses to changes in intravascular volume status are not predictable in individual patients.

For example, a healthy young patient with a subclinical hypovolemic state often has normal BP and HR because the stress response to surgery activates the sympathetic nervous system and the renin-angiotensin system, with release of vasoconstrictor hormones that increase BP. Although the resulting peripheral vasoconstriction favors maintenance of adequate perfusion to the heart and the brain, perfusion to other organs such as the kidneys, gastrointestinal tract, and skin may be reduced. Furthermore, general or neuraxial anesthesia may blunt these compensatory vasoconstrictive responses to decreased perfusion.

Another example is a patient treated with beta-blockers who may not manifest tachycardia in response to hypovolemia. Thus, changes in volume status would not be detected by changes in HR.

Central venous pressure – CVP and pulmonary artery occlusion pressure are sometimes used to provide supplemental data regarding intravascular volume status, but these parameters are inaccurate surrogates to determine cardiac preload, are poor predictors of fluid responsiveness [23], and do not detect or predict impending pulmonary edema indicative of hypervolemia [17,21,24-30].

Urine output – Oliguria (UO <0.5 mL/kg per hour) is a commonly used indicator of hypovolemia. However, in patients undergoing anesthesia and surgery, oliguria alone is not a sufficient indication for fluid administration. Inhalation anesthetics, as well as surgical stress, may reduce UO. If the patient is actually euvolemic, administration of fluid to treat oliguria may lead to fluid overload [4,5].

Traditional thresholds for intraoperative oliguria do not predict acute kidney injury (AKI) [11,31-33]. In a 2016 systematic review and meta-analysis of 28 trials conducted in surgical and critically ill patients, less renal dysfunction was noted in patients receiving goal-directed fluid therapy without use of oliguria to guide fluid administration compared with patients who received conventional fluid management that targeted oliguria reversal (odds ratio [OR] 0.45, 95% CI 0.34-0.61) [33]. Different thresholds for UO were examined in a large retrospective study of 3560 patients undergoing major abdominal surgery [34]. Intraoperative UO <0.3 mL/kg per hour was associated with increased risk for AKI (OR 2.65, 95% CI 1.77-3.97) compared with UO 0.3 to 0.5 mL/kg per hour or higher. Another retrospective study in patients undergoing major surgery noted that duration of intraoperative oliguria (defined as UO <0.5 mL/kg) longer than 120 minutes was associated with increased risk for AKI compared with propensity-score matched patients with a shorter duration of oliguria (OR 2.2, 95% CI 1.8-2.7) [35]. Similarly, in a post hoc analysis of data from a trial of restrictive versus liberal fluid therapy (see 'Our approach to fluid management' below) [36], intraoperative UO <0.5 mL/kg per hour was associated with increased risk of AKI (risk ratio [RR] 1.38, 95% CI 1.14-1.44), although more severe oliguria with UO <0.3 mL/kg per hour did not increase risk further, and most patients with oliguria did not develop AKI [37]. Taken together, these data suggest that traditional targets that attempt to continuously maintain UO >0.5 mL/kg per hour are not warranted, but sustained oliguria may be associated with increased risk of renal injury, particularly if <0.3 mL/kg per hour [34,38,39].

Mixed venous oxygen saturation – Measurements intended to track global oxygen (O2) delivery have limited utility to guide fluid therapy [40]. These include measurements of mixed venous O2 saturation (SvO2; or central venous O2 saturation [ScvO2]), which may be obtained intermittently from blood gas analysis or continuously using a fiberoptic catheter. Although SvO2 and ScvO2 are proportional to cardiac output, tissue perfusion, and tissue O2 delivery, these measurements are also inversely proportional to tissue O2 consumption and do not reflect changes in tissue perfusion during the perioperative period when O2 consumption varies [40,41]. (See "Oxygen delivery and consumption".)

Dynamic hemodynamic parameters — Dynamic hemodynamic indices are used to guide goal-directed fluid therapy in patients undergoing major invasive surgery, particularly high-risk patients with large expected blood losses and fluid shifts [3]. Dynamic parameters provide superior assessment of response to a fluid challenge (ie, volume responsiveness) compared with traditional static parameters [25,40,42-46]. (See 'Goal-directed fluid therapy' below.)

Indices based on respiratory variation (arterial pressure waveform) — Variations in the arterial pressure waveform that occur during respiration (eg, pulse pressure variation [PPV], stroke volume variation [SVV], systolic blood pressure variation [SPV], or change in inferior vena cava diameter) can be observed or measured to assess responses to fluid challenges (figure 1) [46-50]. During controlled mechanical ventilation, inspiration increases intrathoracic pressure, which reduces venous return, right ventricular (RV) filling volume, and right and left ventricular stroke volume (SV). The opposite effects occur during expiration. These changes in venous return lead to variations in SV, as well as pulse pressure and systolic BP (figure 2), if arterial vasomotor tone and cardiac function remain constant [49]. Normal respiratory variations in these dynamic parameters are <10 percent [51]. Greater variations suggest fluid responsiveness and the likely need for fluid administration (figure 1) [23].

Although hemodynamic indices of respiratory variation can be computed (manually or automatically), visual estimation may be adequate to guide fluid therapy. In one study, determination of the need for a fluid bolus based on visual estimation of SPV in the arterial waveform was compared with computed values, with only 1 percent of treatment decisions based on visual estimation being incorrect (figure 1) [47].

Each of the dynamic indices based on respiratory variation has advantages and disadvantages, with limitations in sensitivity and specificity (table 1) [52-54]. In a 2018 systematic review (5017 patients; 68 studies), the pooled area under the curve (AUC) for PPV was 0.86 (95% CI 0.80-0.92), with a sensitivity of 80 percent (95% CI 74-85 percent) and specificity of 83 percent (95% CI 73-91 percent) [54]. In that study, the pooled AUC for SVV was 0.87 (95% CI 0.81-0.93), with a sensitivity of 82 percent (95% CI 75-89 percent) and specificity of 77 percent (95% CI 71-82 percent). Furthermore, these indices are not useful during open chest procedures [55,56], during mechanical ventilation with low tidal volumes <8 mL/kg or high positive end-expiratory pressure >15 cm H2O, or in patients who have elevated intraabdominal pressure, cardiac arrhythmias, RV failure, or a requirement for vasoactive infusions [50,57,58]. Thus, although superior to static parameters for assessment of fluid responsiveness (see 'Static (traditional) parameters' above), indices based on respiratory variation are interpreted with caution and should consider the clinical setting.

Stroke volume estimates — SV derived by using esophageal Doppler technology or arterial wave form analysis have been used to guide fluid therapy in an attempt to maintain intravascular volume [46,59-61]. Such devices may be useful when indices based on respiratory variation in the intra-arterial waveform cannot be used. (See 'Indices based on respiratory variation (arterial pressure waveform)' above.)

Left ventricular size estimates (transesophageal echocardiography) — With transesophageal echocardiography (TEE), intravascular volume status can be quickly assessed in the transgastric midpapillary short-axis view by qualitative visual assessment of LV cavity size. Underfilling of the left ventricle caused by acute hypovolemia is easily recognized in a patient with hyperdynamic systolic function and decreased end-diastolic and end-systolic LV cavity dimensions (movie 1). Also, quantitative measurements of the internal diameter or cross-sectional area of the LV at end-diastole can be made (image 1 and image 2 and table 2) [62,63]. Changes from baseline (normovolemia) are monitored using these qualitative and/or quantitative assessments. (See "Intraoperative transesophageal echocardiography for noncardiac surgery", section on 'Assessment of left ventricular volume'.).

Noninvasive technologies — Noninvasive commercially available technologies that measure cardiac output or assess fluid responsiveness have been studied (eg, pulse wave transit time, pulse contour analysis, carbon dioxide rebreathing, and thoracic electrical bioimpedance or bioreactance devices) [64]. A 2017 meta-analysis concluded that the percentage error of these devices for measurements of cardiac output is high compared with standard thermodilution techniques [65].

Measurement of laboratory values — Absent an ischemic insult to a specific organ, increased serum lactate levels or lactic acidosis on sequential arterial blood gases can be an important indicator of reduced global tissue perfusion. However, these laboratory values do not provide information regarding contemporaneous clinical intravascular volume status since they are measured intermittently and do not immediately reflect acute changes. (See "Etiology, clinical manifestations, and diagnosis of volume depletion in adults", section on 'Clinical manifestations'.)

CHOOSING FLUID: CRYSTALLOID, COLLOID, OR BLOOD

Crystalloid solutions — Crystalloids are solutions of electrolytes and sterile water that may be isotonic, hypotonic, or hypertonic with respect to plasma. Balanced electrolyte solutions (also termed buffered crystalloid solutions) that have an electrolyte composition similar to plasma with the addition of a buffer (eg, lactate) are most widely used [66]. Examples include Ringer's lactate (also termed Hartmann's solution) or Plasmalyte.

We typically select a balanced electrolyte crystalloid solution for routine perioperative fluid administration in order to maintain normovolemia and/or replace lost blood. Crystalloids are administered on a 1.5:1.0 volume basis until a transfusion threshold is met. We avoid dextrose-containing solutions due to the putative adverse effects of hyperglycemia [67].

We avoid administration of a large volume of normal saline (NS; 0.9 percent), as this has been associated with hyperchloremic acidosis [68-81]. Risk for other adverse outcomes, particularly acute kidney injury (AKI), has been associated with NS in several observational and randomized studies [69,70,82-85]. However, results are not consistent in patients who do not receive large volumes of NS or are not critically ill [86,87]. A meta-analysis of randomized trials that included 1096 surgical patients noted that perioperative administration of buffered (ie, balanced) electrolyte solutions was associated with a lower incidence of minor metabolic derangements, particularly mild metabolic acidosis compared with NS, but did not demonstrate an effect on mortality or renal replacement therapy (RRT); however, most of these patients were not critically ill [68]. Similarly, another meta-analysis of randomized trials in more than 3700 unselected critically ill or perioperative adult patients did not find a significant reduction in mortality (odds ratio [OR] 0.90, 95% CI 0.69-1.17) or incidence of RRT (OR 1.12, 95% CI 0.80-1.58) with administration of balanced electrolyte solutions rather than NS; however, most of these patients received a low fluid volume [88].

Colloid solutions — Colloids are human plasma derivatives (eg, human albumin, fresh frozen plasma [FFP]) or semisynthetic preparations (eg, hydroxyethyl starch [HES], gelatins) [89]. Colloids may be dissolved in isotonic saline or in a solution with a balanced electrolyte concentration similar to plasma [89].

There is little evidence that colloid solutions are superior to balanced electrolyte crystalloid solutions [90-94]. In one study, closed loop administration of balanced HES for elective major open abdominal surgery produced lower morbidity on postoperative day two and better disability-free survival at follow up one year after surgery, compared with balanced crystalloid [93,94]. Some clinicians prefer to use colloids in selected patients or situations in attempts to expand microvascular volume with minimal capillary leakage, thereby minimizing edema formation and the total quantity of administered fluid [95]. For example, during blood loss, colloids may be administered on a 1.0:1.0 volume basis until a transfusion threshold is met [95,96]. (See "Intraoperative transfusion of blood products in adults", section on 'Red blood cells'.)

Albumin — Human serum albumin is available in both 5 and 25 percent solutions. In some parts of the world, human serum albumin is available as 4 and 20 percent solutions. Human albumin 5 percent has a volume effect (ie, the percent of fluid administered that remains intravascular) of 70 percent, while albumin 25 percent solution is isosmotic with plasma (table 3). Human albumin is pasteurized and does not transmit any known infectious diseases. (See "Plasma derivatives and recombinant DNA-produced coagulation factors", section on 'Albumin'.)

Albumin is more expensive than other solutions, and may not be safer or more efficacious than synthetic colloids (eg, HES) or balanced crystalloid solutions [86,90,97-99]. (See 'Hydroxyethyl starches' below and 'Crystalloid solutions' above.) However, administration of 20 percent albumin during the intraoperative period during noncardiac surgery caused a long-lasting plasma volume expansion lasting into the postoperative period [100,101]. In one study, the intravascular half-life was 9.1 (5.7 to 11.2) hours [100]. A pilot study of administration of 20 percent albumin in the postoperative period after cardiac surgery resulted in less positive fluid balance and volume of fluid boluses, as well as decreased dosing of a vasopressor (norepinephrine) [102].

Hydroxyethyl starches — We do not use HES solutions for fluid therapy in surgical patients. HES solutions are synthetic colloids, identified by three numbers corresponding to concentration, molecular weight, and molar substitution (ie, the average hydroxyethyl groups per one glucose unit) (table 3). As an example, Hespan is HES 6 percent (600/0.75) with a volume effect of 100 percent and a high molar substitution of 0.75. Risk of HES-induced renal toxicity may be influenced by the molar substitution level in the specific product, with lower risk for low substituted HES products [103]. Due to concerns regarding renal toxicity and effects on hemostasis, administration of HES solutions is restricted in Europe and North America [86].

In a randomized trial of more than 800 patients undergoing major abdominal surgery, administration of a low molar-substituted HES 130/0.4 solution versus 0.9 percent saline to provide goal-directed fluid therapy (see 'Goal-directed fluid therapy' below) did not reduce the composite outcome of death or major postoperative complications, and may have had harmful effects (eg, increased of postoperative AKI [relative risk 1.34, 95% CI 1.00-1.80]) [104]. Similarly, a 2018 systematic review in critically ill patients with medical and surgical diagnoses noted a higher incidence of RRT in those receiving HES solutions compared with those receiving crystalloids (risk ratio [RR] 1.30, 95% CI 1.14-1.48; 8527 participants, nine studies) [90]. Also, a 2010 systematic review in mixed surgical and nonsurgical patient populations found an overall increased risk of author-defined kidney failure in surgical and medical patients receiving HES solutions (including some who received highly substituted HES products) compared with those receiving various other types of fluid therapy [105]. Furthermore, administration of HES solutions in cardiac surgical patients increases risk of postoperative bleeding and transfusion compared with albumin [106]. However, data are not consistent. Some randomized and observational studies have noted no differences in risk for AKI or other serious postoperative complications in patients receiving goal-directed fluid therapy with HES compared with other types of fluids (eg, balanced crystalloid solution, 5 percent albumin) [92,99,107-109].  

Since HES products impair platelet reactivity and decrease circulating plasma concentrations of coagulation factor VIII and von Willebrand factor, administration may result in weakening of clot formation and more transfusions of blood products including FFP, cryoprecipitate, and platelets compared with other solutions [90,110-112]. A 2018 systematic review in critically ill patients noted a higher incidence of transfusion in those receiving starch solutions compared with those receiving crystalloids (RR 1.19, 95% CI 1.02-1.39; 1917 participants, eight studies) [90]. HES products with low molar substitution (eg, pentastarch and tetrastarch) may have less effect on hemostasis [113]. One meta-analysis compared HES solutions with low (0.4) versus a somewhat higher (0.5) molar substitution, noting significantly less blood loss (404 mL) and red blood cell (RBC) transfusion (137 mL) in patients receiving the low substituted product (seven trials; 449 patients) [114].

Gelatins — Gelatins are not used in the United States because of their short duration of action (two to three hours) due to rapid excretion in the urine, possible effects on coagulation, and a relatively high incidence of anaphylaxis [86,89,115]. (See "Perioperative anaphylaxis: Clinical manifestations, etiology, and management", section on 'Colloids and plasma expanders'.)

Gelatins are used in some countries because they are inexpensive and have a volume effect of 70 to 80 percent (table 3) [116].

Blood

Transfusion of red blood cells — RBCs are used to replace intraoperative blood loss when a transfusion threshold is met, as discussed separately. (See "Intraoperative transfusion of blood products in adults", section on 'Red blood cells'.)

Transfusion of plasma derivatives — Decisions regarding transfusion of plasma derivatives of human blood (eg, FFP, cryoprecipitate) are based on estimates of the amount of current and expected ongoing blood loss and evidence of intractable microvascular bleeding indicating abnormal hemostasis, ideally with confirmation by diagnostic test results. These decisions are discussed separately. (See "Intraoperative transfusion of blood products in adults", section on 'Plasma' and "Intraoperative transfusion of blood products in adults", section on 'Cryoprecipitate'.)

OUR APPROACH TO FLUID MANAGEMENT — Our intraoperative fluid management strategy is based on the invasiveness of the surgical procedure, as well as whether invasive monitoring of dynamic hemodynamic parameters is employed. Expected blood loss and likelihood of nonhemorrhagic fluid shifts (eg, from open body cavities and wounds) are factors in decision-making regarding selection of noninvasive and/or invasive monitors for a planned surgical procedure (see 'Dynamic hemodynamic parameters' above). Other patient-specific and procedure-specific factors affecting fluid management include the patient's comorbidities (eg, anemia) and planned postoperative disposition (eg, home, ambulatory hospital ward, critical care unit).

Minimally/moderately invasive surgery — For most adult patients undergoing relatively brief minimally or moderately invasive surgery with planned early postoperative ambulation, we administer 1 to 2 L of a balanced electrolyte solution if the procedure does not incur significant fluid shifts or blood loss. This 1 to 2 L of fluid is typically administered during surgery, over a period of 30 minutes to two hours. Such empiric but limited fluid administration for less invasive surgery in ambulatory patients addresses the mild dehydration caused by preoperative fasting and is associated with less postoperative nausea and vomiting, as well as less postoperative pain [117,118]. A smaller fluid volume is appropriate in patients with a history of heart failure or chronic obstructive pulmonary disease.

Major invasive surgery — For adult patients undergoing major invasive surgical procedures, we employ a restrictive, zero-balance approach that minimizes fluid administration, or a goal-directed approach with fluid administration to achieve a pre-specified goal [8]. Evidence suggests that either a restrictive or goal-directed approach is associated with a lower incidence of perioperative morbidities (and possibly mortality) compared with traditional liberal or fixed-volume approaches that may result in fluid overload [10,13,14,22,59,60,119-122]. (See 'Avoid traditional liberal or fixed-volume approaches' below.)

We use a restrictive (zero-balance) approach to fluid therapy for invasive major surgery with expected blood loss <500 mL if there is no invasive monitoring of dynamic hemodynamic parameters via an intra-arterial catheter, esophageal Doppler probe, or transesophageal echocardiography (TEE) probe (see 'Restrictive (zero-balance) strategy' below). A potential disadvantage of this approach is that hypovolemia may not be clinically appreciated. Also, if hypotension occurs, it may be difficult to determine the etiology (eg, surgical volume losses or other causes such as cardiovascular responses to anesthetic agents).

For these reasons, we typically select a goal-directed approach for high-risk patients undergoing a major surgical procedure, particularly if significant blood losses (eg, >500 mL) and/or fluid shifts are expected. A goal-directed approach necessitates use of one or more invasive dynamic hemodynamic parameters, as noted below (see 'Goal-directed fluid therapy' below). Final decisions regarding selection of noninvasive versus invasive monitoring are also based on procedure-specific and patient-specific factors, as discussed in separate UpToDate topics.

Restrictive (zero-balance) strategy — With a restrictive zero-balance approach, only the fluid that is lost during surgery is replaced, including the following strategies [14]:

During the intraoperative period, patients receive a balanced electrolyte crystalloid solution administered at a rate of 1 to 3 mL/kg per hour to replace sensible and insensible losses [13].

For blood loss, additional fluid may be administered. Studies suggest that the optimal crystalloid-to-blood volume ratio is approximately 1.5:1.0, and that the optimal colloid-to-blood ratio is 1:1, until a threshold for red blood cell (RBC) transfusion is reached [123-126].

Patients do not receive "preloading" of crystalloids prior to a neuraxial block or induction of general anesthesia.

We avoid replacement of nonanatomic "third space" losses, since evidence suggests that this practice has no benefit, and may cause morbidity [6,127,128].

We avoid extremely deep anesthesia (eg, bispectral index values <40) that may result in hypotension. If necessary, vasopressor agents such as phenylephrine or ephedrine may be employed to treat hypotension caused by administration of anesthetic agents and/or neuraxial block [129,130]. (See "Accidental awareness after general anesthesia", section on 'Brain monitoring' and "Hemodynamic management during anesthesia in adults", section on 'Hypotension: Prevention and treatment'.)

Administration of a total volume of balanced electrolyte solutions that modestly exceeds zero fluid balance is appropriate in patients with evidence of hypovolemia [131].

In most studies, restrictive fluid therapy has been associated with better outcomes than traditional liberal or fixed-volume approaches for major elective surgical procedures [13,14,22,86,132,133]. However, variability in study design has resulted in inconsistent results. A 2012 meta-analysis found that standard or liberal approaches to fluid therapy in patients undergoing major abdominal procedures resulted in a higher risk for pneumonia (risk ratio [RR] 2.2, 95% CI 1.0-4.5) and pulmonary edema (RR 3.8, 95% CI 1.1-13), as well as longer hospital stay (mean difference two days, 95% CI 0.5-3.4), compared with a restrictive approach to fluid therapy (1160 patients; 12 randomized trials) [134]. In a subsequently published randomized trial of 3000 patients undergoing major abdominal surgery, a restrictive (zero-balance) fluid regimen was associated with a higher rate of acute kidney injury (AKI) compared with a liberal fluid regimen (8.6 versus 5.0 percent; RR 1.71, 95% CI 1.29-2.27) [36]. A limitation of this study was its pragmatic design in that perioperative care was not standardized and there was a wide variation in the anesthetic and analgesic techniques, including use of epidural analgesia, variable intraoperative hemodynamic management, and variable postoperative care. The total fluid volume administered during and up to 24 hours after surgery was 3.7 versus 6.1 L in the restrictive and liberal groups, respectively. These volumes were lower than traditional liberal fluid strategy totals, and balanced electrolyte solutions were used in both groups [131]. Similarly, a restrictive approach was associated with AKI in an observational trial of 769 patients undergoing cystectomy (odds ratio [OR] 0.79, 95% CI 0.68-0.91) [135]. Conversely, a randomized trial in 166 patients undergoing radical cystectomy employed a restrictive approach (1 mL/kg/h) combined with low-dose norepinephrine infusion during the initial portion of the surgery subsequently followed by hydration (3 mL/kg/h) during the last part of the surgical procedure [136]. This so-called "restrictive deferred hydration" approach was associated with fewer complications (RR 0.70, 95% CI 0.55-0.88) and a lower median duration of hospital stay (15 days, range 11 to 27) than a liberal approach with 6 mL/kg/h (17 days, range 11 to 95).

Taken together, these studies suggest that administration of a total volume of balanced electrolyte solutions that modestly exceeds zero fluid balance is not harmful [131]. Also, evidence suggesting hypovolemia should be recognized and appropriately treated with fluid, as noted below (see 'Goal-directed fluid therapy' below). However, excessive perioperative administration of intravenous fluid, which was common in traditional liberal or fixed-volume approaches to fluid therapy, should be avoided [10,86]. (See 'Avoid traditional liberal or fixed-volume approaches' below.)

Goal-directed fluid therapy — We typically select a goal-directed approach to fluid therapy (GDT) in patients undergoing major invasive surgery with expected blood loss >500 mL and/or other significant perioperative fluid shifts. With this approach, we ensure that intravascular volume status is optimal before adding vasopressor therapy to achieve optimal blood pressure [3].

While GDT appears superior to traditional liberal or fixed-volume approaches, there are limited data comparing GDT to the restrictive approach described above [134,137]. One disadvantage of GDT is that it requires invasive monitoring of dynamic hemodynamic parameters [3,14,59,60,120]. We consider the following factors in selection of monitoring modalities:

In most patients undergoing major surgery, we use the intra-arterial waveform tracing for automated measurements of pulse pressure variations (PPV) or stroke volume variation (SVV), or visually estimated or manually calculated PPV or systolic pressure variations (SPV), in order to determine responses to fluid boluses (typically 250 mL increments) (figure 1 and figure 2).

For high-risk patients undergoing a surgical procedure with expected blood loss >1000 mL, significant nonhemorrhagic fluid losses, and/or likely prolonged duration, we typically use a commercially available device that provides automated calculation of PPV, SVV, or SPV by analyzing the intra-arterial waveform tracing to assess responses to fluid challenges. An alternative is use of an esophageal Doppler device to estimate stroke volume (SV) [64]. TEE is another option, allowing visual qualitative evaluation or quantitative measurements of the left ventricular (LV) cavity size to monitor fluid responsiveness (movie 1 and image 1 and image 2 and table 2). (See 'Dynamic hemodynamic parameters' above.)

With a GDT approach, fluid is administered to achieve a pre-specified goal:

If respiratory variations in the arterial pressure waveform (PPV or SPV) are >10 to 15 percent, then the patient is assumed to be fluid responsive and we administer fluid boluses of a balanced electrolyte crystalloid solution (typically in 250 mL increments) [23,51,138]. Once change in the monitored dynamic parameter is <10 percent, fluid administration is stopped to avoid hypervolemia.

If SV estimates are used for dynamic hemodynamic monitoring, the typical goal of therapy is to achieve and maintain optimal intravascular volume with maximum SV. The new SV value after fluid administration has resulted in <10 percent change is recorded as the new baseline goal value (representing the maximum SV to be maintained). (See 'Indices based on respiratory variation (arterial pressure waveform)' above.).

If TEE is employed, hypovolemic and hypervolemic states can be quickly assessed by visual qualitative evaluation or quantitative measurements of LV cavity size. Fluid administration is stopped once normovolemia has been achieved (movie 1 and image 1 and image 2 and table 2). (See 'Left ventricular size estimates (transesophageal echocardiography)' above.)

Although most studies evaluating GDT have used boluses of colloid fluid [139,140], we typically select a balanced crystalloid solution for fluid boluses. Randomized trials in patients undergoing elective abdominal surgery have found little difference in postoperative complications or any clinical benefit with use of a 6 percent hydroxyethyl starch (HES) colloid solution for fluid boluses compared with a balanced crystalloid solution to provide GDT [92,94,123]. (See 'Choosing fluid: Crystalloid, colloid, or blood' above.)

GDT appears to be superior to traditional fixed-volume or liberal fluid approaches (see 'Avoid traditional liberal or fixed-volume approaches' below). In one meta-analysis, fluid administration with a GDT approach was associated with improved clinical outcomes, including lower risk of mortality (odds ratio [OR] 0.66, 95% CI 0.50-0.87), pneumonia (OR 0.69, 95% CI 0.51-0.92), acute kidney injury (OR 0.73, 95% CI 0.58-0.92), wound infection (OR 0.48, 95% CI 0.37-0.63), and shorter length of hospital stay (-0.9 days; 95% CI -1.3 to -0.5 days), compared with standard fluid management according to protocol (eg, maintenance of mean arterial pressure >65 mmHg) or the discretion of treating clinicians (11,659 patients; 95 randomized trials) [137]. Other randomized trials and meta-analyses have noted similar results with GDT compared with standard care (eg, lower risk of respiratory, renal, wound, and gastrointestinal complications, with shorter time to hospital discharge) [60,134,141-146]. While most studies reported lower complication rates with GDT, numerous limitations should be noted. These include clinical heterogeneity among trials with differing definitions for GDT, lack of well-defined endpoints, different types of fluid therapy, different devices used to monitor dynamic hemodynamic parameters, variations in management of the control group, different types of surgery, and small sample sizes [14,64,147-149]. In addition, most studies have included only limited information regarding anesthetic techniques and perioperative surgical care [147].

Whether GDT is superior to a restrictive fluid strategy is less certain. In a 2019 meta-analysis of randomized trials in patients undergoing major noncardiac surgery, very low-certainty evidence suggested that restrictive fluid therapy does not affect the risk of complications (RR 1.61, 95% CI 0.78-3.34; five studies; 484 participants), but may increase the risk of all-cause mortality (risk difference [RD] 0.03, 95% CI 0.00-0.06) [140]. Furthermore, GDT does not appear to offer additional benefits over a restrictive fluid approach for patients managed with protocols to achieve enhanced recovery after surgery (ERAS), [150-153]. The likely reason is that ERAS protocols implement multiple processes that each reduce the risk of perioperative fluid imbalances (eg, avoidance of preoperative dehydration, use of an intraoperative restrictive fluid approach, emphasis on early postoperative alimentation and ambulation). One meta-analysis assessing GDT in this setting found no benefit (or harm) with its use [143]. (See "Anesthetic management for enhanced recovery after major surgery (ERAS) in adults", section on 'Fluid management'.)

Clinical questions regarding GDT that remain unanswered include which types of patients are most likely to benefit, timing of the use of GDT (preoperative, intraoperative, and/or postoperative), which outcome measures or endpoints are optimal, which combinations of therapies constitute the best approach (eg, fluids with or without vasopressors or inotropic agents), and how long the regimen should be maintained during the postoperative period.

Avoid traditional liberal or fixed-volume approaches — We do not use traditional liberal or fixed-volume approaches to fluid therapy in patients undergoing any type of surgery, because these strategies result in administration of a large volume of crystalloid solution, with increased incidence or severity of tissue edema and associated adverse outcomes [13,14,16,22,59,60,119-122,154-156].

Traditional fixed-volume approaches were based upon predetermined calculations that included administration of fluid to account for presumed preoperative deficits, as well as intraoperative blood and urinary losses. In addition, fluid was typically administered to compensate for calculated non-anatomic "third space" fluid losses during the surgical procedure. This practice is inappropriate, as it has been well-established that such third space losses do not exist [6,127,128]. Furthermore, fixed volume regimens specified replacement of initial blood loss with crystalloid volume that was three times the amount of lost blood. For example, if blood loss was estimated to be 500 to 1000 mL, then 1500 to 3000 mL of crystalloid was typically administered. This calculation is not supported by available data. Rather, optimal volume ratios to compensate for lost blood are estimated to be 1.5:1.0 for crystalloid and 1.0:1.0 for colloid, as discussed above [123-125] (see 'Crystalloid solutions' above and 'Colloid solutions' above). Similarly, large volumes of fluid are unnecessary as preloading before a neuraxial block.

The overall result of use of traditional fixed volume calculations was administration of a large total volume of crystalloid solution and a high likelihood of increased perioperative tissue edema. For these reasons, such approaches have been abandoned.

SUMMARY AND RECOMMENDATIONS

Maintenance of tissue perfusion by maintaining euvolemia is the goal for intraoperative monitoring of intravascular fluid volume and administration of intraoperative fluid therapy. Both hypovolemia and hypervolemia are associated with postoperative morbidity. (See 'Consequences of intravascular volume derangements' above.)

Absolute or relative hypovolemia is common in the perioperative period due to preoperative dehydration, vasodilation caused by anesthetic and adjuvant drugs, and surgical bleeding. The most common cause of perioperative tissue edema is retention of fluid administered during surgery. (See 'Causes of intravascular volume derangements' above.)

While physiological parameters such as blood pressure (BP), heart rate (HR), central venous pressure (CVP), and urine output (UO) are monitored during surgery, significant intraoperative reduction in tissue perfusion may not be recognized even with continuous monitoring of these static parameters. (See 'Static (traditional) parameters' above.)

Monitoring of dynamic hemodynamic parameters based on respiratory variation uses the intra-arterial waveform tracing for automated analysis or visually estimated or manually calculated pulse pressure variations (PPV), stroke volume variation (SVV), or systolic pressure variations (SPV) (figure 1 and figure 2), or estimates of stroke volume (SV) in order to determine responses to fluid boluses (typically administered in 250 mL increments). An alternative is use of transesophageal echocardiography (TEE) to recognize hypovolemic and hypervolemic states by visual qualitative assessment or quantitative measurements of left ventricular (LV) cavity size (movie 1 and image 1 and image 2 and table 2). (See 'Dynamic hemodynamic parameters' above.)

We suggest the use of balanced electrolyte solutions (eg, Ringer's lactate, Plasmalyte), rather than normal saline or colloid as the standard intravenous fluid to maintain or replace intravascular volume in surgical patients (Grade 2C). In particular, we avoid hydroxyethyl starch (HES) colloid solutions due to possible harmful effects (eg, increased risk of postoperative acute kidney injury). We typically select a balanced crystalloid solution to be administered at 1 to 3 mL/kg per hour to replace sensible and insensible losses, and for fluid boluses (typically 250 mL) to optimize intravascular volume. (See 'Colloid solutions' above and 'Crystalloid solutions' above.)

We replace initial blood loss with either crystalloid administered in a volume that is 1.5 times the amount of lost blood, or colloid administered on a 1.0:1.0 volume basis, until a transfusion threshold is met. (See 'Choosing fluid: Crystalloid, colloid, or blood' above.)

For most adult patients undergoing minimally or moderately invasive surgical procedures with planned early postoperative ambulation, we administer 1 to 2 L of a balanced electrolyte solution to provide adequate intravascular hydration. (See 'Minimally/moderately invasive surgery' above.)

For major invasive surgical procedures, we suggest a restrictive (zero-balance) or goal-directed therapy (GDT) approach to fluid therapy, rather than traditional liberal or fixed-volume approaches (Grade 2B). We choose between these strategies based on anticipated significant blood loss (eg, >500 mL) and/or other significant perioperative fluid shifts. Both the restrictive and GDT strategies appear to be associated with reduced morbidity compared with more liberal fluid administration. (See 'Major invasive surgery' above.)

A restrictive (zero-balance) approach to fluid therapy replaces only fluid lost during the procedure. (See 'Restrictive (zero-balance) strategy' above.)

A GDT approach to fluid therapy requires invasive monitoring of dynamic hemodynamic parameters (ie, intra-arterial catheter). (See 'Goal-directed fluid therapy' above and 'Dynamic hemodynamic parameters' above.)

For most patients, we use visually estimated SPV in the intra-arterial waveform tracing to determine responsiveness to fluid boluses (typically 250 mL) (figure 1).

For high-risk patients undergoing a surgical procedure with expected blood loss >1000 mL, significant nonhemorrhagic fluid shifts, and/or likely prolonged duration, we typically select a commercially available device to determine responses to fluid challenges (eg, devices that provide automated calculations of respiratory variations in systolic BP, pulse pressure, or SV in the intra-arterial waveform tracing, esophageal Doppler device to estimate SV, TEE to assess LV cavity size).

REFERENCES

  1. Jacob M, Chappell D, Conzen P, et al. Blood volume is normal after pre-operative overnight fasting. Acta Anaesthesiol Scand 2008; 52:522.
  2. Danielsson EJD, Lejbman I, Åkeson J. Fluid deficits during prolonged overnight fasting in young healthy adults. Acta Anaesthesiol Scand 2019; 63:195.
  3. Miller TE, Myles PS. Perioperative Fluid Therapy for Major Surgery. Anesthesiology 2019; 130:825.
  4. Brauer KI, Svensén C, Hahn RG, et al. Volume kinetic analysis of the distribution of 0.9% saline in conscious versus isoflurane-anesthetized sheep. Anesthesiology 2002; 96:442.
  5. Connolly CM, Kramer GC, Hahn RG, et al. Isoflurane but not mechanical ventilation promotes extravascular fluid accumulation during crystalloid volume loading. Anesthesiology 2003; 98:670.
  6. Jacob M, Chappell D, Rehm M. The 'third space'--fact or fiction? Best Pract Res Clin Anaesthesiol 2009; 23:145.
  7. Lamke LO, Nilsson GE, Reithner HL. Water loss by evaporation from the abdominal cavity during surgery. Acta Chir Scand 1977; 143:279.
  8. Thacker JK, Mountford WK, Ernst FR, et al. Perioperative Fluid Utilization Variability and Association With Outcomes: Considerations for Enhanced Recovery Efforts in Sample US Surgical Populations. Ann Surg 2016; 263:502.
  9. Bellamy MC. Wet, dry or something else? Br J Anaesth 2006; 97:755.
  10. Shin CH, Long DR, McLean D, et al. Effects of Intraoperative Fluid Management on Postoperative Outcomes: A Hospital Registry Study. Ann Surg 2018; 267:1084.
  11. Zarbock A, Koyner JL, Hoste EAJ, Kellum JA. Update on Perioperative Acute Kidney Injury. Anesth Analg 2018; 127:1236.
  12. Myrberg T, Lindelöf L, Hultin M. Effect of preoperative fluid therapy on hemodynamic stability during anesthesia induction, a randomized study. Acta Anaesthesiol Scand 2019; 63:1129.
  13. Chappell D, Jacob M, Hofmann-Kiefer K, et al. A rational approach to perioperative fluid management. Anesthesiology 2008; 109:723.
  14. Brandstrup B. Fluid therapy for the surgical patient. Best Pract Res Clin Anaesthesiol 2006; 20:265.
  15. Lowell JA, Schifferdecker C, Driscoll DF, et al. Postoperative fluid overload: not a benign problem. Crit Care Med 1990; 18:728.
  16. Holte K, Sharrock NE, Kehlet H. Pathophysiology and clinical implications of perioperative fluid excess. Br J Anaesth 2002; 89:622.
  17. Arieff AI. Fatal postoperative pulmonary edema: pathogenesis and literature review. Chest 1999; 115:1371.
  18. Nisanevich V, Felsenstein I, Almogy G, et al. Effect of intraoperative fluid management on outcome after intraabdominal surgery. Anesthesiology 2005; 103:25.
  19. Mayberry JC, Welker KJ, Goldman RK, Mullins RJ. Mechanism of acute ascites formation after trauma resuscitation. Arch Surg 2003; 138:773.
  20. Balogh Z, McKinley BA, Cocanour CS, et al. Supranormal trauma resuscitation causes more cases of abdominal compartment syndrome. Arch Surg 2003; 138:637.
  21. Bentzer P, Griesdale DE, Boyd J, et al. Will This Hemodynamically Unstable Patient Respond to a Bolus of Intravenous Fluids? JAMA 2016; 316:1298.
  22. Joshi GP. Intraoperative fluid restriction improves outcome after major elective gastrointestinal surgery. Anesth Analg 2005; 101:601.
  23. Marik PE, Cavallazzi R, Vasu T, Hirani A. Dynamic changes in arterial waveform derived variables and fluid responsiveness in mechanically ventilated patients: a systematic review of the literature. Crit Care Med 2009; 37:2642.
  24. 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.
  25. Magder S. Fluid status and fluid responsiveness. Curr Opin Crit Care 2010; 16:289.
  26. Cecconi M, Aya HD. Central venous pressure cannot predict fluid-responsiveness. Evid Based Med 2014; 19:63.
  27. Gelman S. Venous function and central venous pressure: a physiologic story. Anesthesiology 2008; 108:735.
  28. Eskesen TG, Wetterslev M, Perner A. Systematic review including re-analyses of 1148 individual data sets of central venous pressure as a predictor of fluid responsiveness. Intensive Care Med 2016; 42:324.
  29. Mark JB. Central venous pressure monitoring: clinical insights beyond the numbers. J Cardiothorac Vasc Anesth 1991; 5:163.
  30. 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.
  31. Kheterpal S, Tremper KK, Englesbe MJ, et al. Predictors of postoperative acute renal failure after noncardiac surgery in patients with previously normal renal function. Anesthesiology 2007; 107:892.
  32. Alpert RA, Roizen MF, Hamilton WK, et al. Intraoperative urinary output does not predict postoperative renal function in patients undergoing abdominal aortic revascularization. Surgery 1984; 95:707.
  33. Egal M, Erler NS, de Geus HR, et al. Targeting Oliguria Reversal in Goal-Directed Hemodynamic Management Does Not Reduce Renal Dysfunction in Perioperative and Critically Ill Patients: A Systematic Review and Meta-Analysis. Anesth Analg 2016; 122:173.
  34. Mizota T, Yamamoto Y, Hamada M, et al. Intraoperative oliguria predicts acute kidney injury after major abdominal surgery. Br J Anaesth 2017; 119:1127.
  35. Shiba A, Uchino S, Fujii T, et al. Association Between Intraoperative Oliguria and Acute Kidney Injury After Major Noncardiac Surgery. Anesth Analg 2018; 127:1229.
  36. Myles PS, Bellomo R, Corcoran T, et al. Restrictive versus Liberal Fluid Therapy for Major Abdominal Surgery. N Engl J Med 2018; 378:2263.
  37. Myles PS, McIlroy DR, Bellomo R, Wallace S. Importance of intraoperative oliguria during major abdominal surgery: findings of the Restrictive versus Liberal Fluid Therapy in Major Abdominal Surgery trial. Br J Anaesth 2019; 122:726.
  38. Kunst G, Ostermann M. Intraoperative permissive oliguria - how much is too much? Br J Anaesth 2017; 119:1075.
  39. Md Ralib A, Pickering JW, Shaw GM, Endre ZH. The urine output definition of acute kidney injury is too liberal. Crit Care 2013; 17:R112.
  40. Renner J, Scholz J, Bein B. Monitoring fluid therapy. Best Pract Res Clin Anaesthesiol 2009; 23:159.
  41. Knotzer H, Hasibeder WR. Microcirculatory function monitoring at the bedside--a view from the intensive care. Physiol Meas 2007; 28:R65.
  42. Funk DJ, Moretti EW, Gan TJ. Minimally invasive cardiac output monitoring in the perioperative setting. Anesth Analg 2009; 108:887.
  43. Desebbe O, Cannesson M. Using ventilation-induced plethysmographic variations to optimize patient fluid status. Curr Opin Anaesthesiol 2008; 21:772.
  44. Cecconi M, Parsons AK, Rhodes A. What is a fluid challenge? Curr Opin Crit Care 2011; 17:290.
  45. Ansari BM, Zochios V, Falter F, Klein AA. Physiological controversies and methods used to determine fluid responsiveness: a qualitative systematic review. Anaesthesia 2016; 71:94.
  46. Thiele RH, Bartels K, Gan TJ. Inter-device differences in monitoring for goal-directed fluid therapy. Can J Anaesth 2015; 62:169.
  47. Thiele RH, Colquhoun DA, Blum FE, Durieux ME. The ability of anesthesia providers to visually estimate systolic pressure variability using the "eyeball" technique. Anesth Analg 2012; 115:176.
  48. Monnet X, Marik PE, Teboul JL. Prediction of fluid responsiveness: an update. Ann Intensive Care 2016; 6:111.
  49. Jozwiak M, Monnet X, Teboul JL. Pressure Waveform Analysis. Anesth Analg 2018; 126:1930.
  50. Si X, Xu H, Liu Z, et al. Does Respiratory Variation in Inferior Vena Cava Diameter Predict Fluid Responsiveness in Mechanically Ventilated Patients? A Systematic Review and Meta-analysis. Anesth Analg 2018; 127:1157.
  51. Cannesson M, Le Manach Y, Hofer CK, et al. Assessing the diagnostic accuracy of pulse pressure variations for the prediction of fluid responsiveness: a "gray zone" approach. Anesthesiology 2011; 115:231.
  52. Yang SY, Shim JK, Song Y, et al. Validation of pulse pressure variation and corrected flow time as predictors of fluid responsiveness in patients in the prone position. Br J Anaesth 2013; 110:713.
  53. Biais M, Bernard O, Ha JC, et al. Abilities of pulse pressure variations and stroke volume variations to predict fluid responsiveness in prone position during scoliosis surgery. Br J Anaesth 2010; 104:407.
  54. Messina A, Pelaia C, Bruni A, et al. Fluid Challenge During Anesthesia: A Systematic Review and Meta-analysis. Anesth Analg 2018; 127:1353.
  55. Jeong DM, Ahn HJ, Park HW, et al. Stroke Volume Variation and Pulse Pressure Variation Are Not Useful for Predicting Fluid Responsiveness in Thoracic Surgery. Anesth Analg 2017; 125:1158.
  56. Raphael J, Regali LA, Thiele RH. Hemodynamic monitoring in thoracic surgical patients. Curr Opin Anaesthesiol 2017; 30:7.
  57. Michard F, Biais M. Rational fluid management: dissecting facts from fiction. Br J Anaesth 2012; 108:369.
  58. Lansdorp B, Lemson J, van Putten MJ, et al. Dynamic indices do not predict volume responsiveness in routine clinical practice. Br J Anaesth 2012; 108:395.
  59. Abbas SM, Hill AG. Systematic review of the literature for the use of oesophageal Doppler monitor for fluid replacement in major abdominal surgery. Anaesthesia 2008; 63:44.
  60. Phan TD, Ismail H, Heriot AG, Ho KM. Improving perioperative outcomes: fluid optimization with the esophageal Doppler monitor, a metaanalysis and review. J Am Coll Surg 2008; 207:935.
  61. Bisgaard J, Gilsaa T, Rønholm E, Toft P. Optimising stroke volume and oxygen delivery in abdominal aortic surgery: a randomised controlled trial. Acta Anaesthesiol Scand 2013; 57:178.
  62. Porter TR, Shillcutt SK, Adams MS, et al. Guidelines for the use of echocardiography as a monitor for therapeutic intervention in adults: a report from the American Society of Echocardiography. J Am Soc Echocardiogr 2015; 28:40.
  63. Reeves ST, Finley AC, Skubas NJ, et al. Special article: basic perioperative transesophageal echocardiography examination: a consensus statement of the American Society of Echocardiography and the Society of Cardiovascular Anesthesiologists. Anesth Analg 2013; 117:543.
  64. Zhang C. Does the Device Matter in Goal-Directed Fluid Therapy? Anesth Analg 2016; 123:1061.
  65. Joosten A, Desebbe O, Suehiro K, et al. Accuracy and precision of non-invasive cardiac output monitoring devices in perioperative medicine: a systematic review and meta-analysis†. Br J Anaesth 2017; 118:298.
  66. Morgan TJ. The ideal crystalloid - what is 'balanced'? Curr Opin Crit Care 2013; 19:299.
  67. Duggan EW, Carlson K, Umpierrez GE. Perioperative Hyperglycemia Management: An Update. Anesthesiology 2017; 126:547.
  68. Bampoe S, Odor PM, Dushianthan A, et al. Perioperative administration of buffered versus non-buffered crystalloid intravenous fluid to improve outcomes following adult surgical procedures. Cochrane Database Syst Rev 2017; 9:CD004089.
  69. Krajewski ML, Raghunathan K, Paluszkiewicz SM, et al. Meta-analysis of high- versus low-chloride content in perioperative and critical care fluid resuscitation. Br J Surg 2015; 102:24.
  70. Shaw AD, Bagshaw SM, Goldstein SL, et al. Major complications, mortality, and resource utilization after open abdominal surgery: 0.9% saline compared to Plasma-Lyte. Ann Surg 2012; 255:821.
  71. Sen A, Keener CM, Sileanu FE, et al. Chloride Content of Fluids Used for Large-Volume Resuscitation Is Associated With Reduced Survival. Crit Care Med 2017; 45:e146.
  72. Scheingraber S, Rehm M, Sehmisch C, Finsterer U. Rapid saline infusion produces hyperchloremic acidosis in patients undergoing gynecologic surgery. Anesthesiology 1999; 90:1265.
  73. Wilkes NJ, Woolf R, Mutch M, et al. The effects of balanced versus saline-based hetastarch and crystalloid solutions on acid-base and electrolyte status and gastric mucosal perfusion in elderly surgical patients. Anesth Analg 2001; 93:811.
  74. Chowdhury AH, Cox EF, Francis ST, Lobo DN. A randomized, controlled, double-blind crossover study on the effects of 2-L infusions of 0.9% saline and plasma-lyte® 148 on renal blood flow velocity and renal cortical tissue perfusion in healthy volunteers. Ann Surg 2012; 256:18.
  75. Schäfer M, Von Ungern-Sternberg BS, Wight E, Schneider MC. Isotonic fluid absorption during hysteroscopy resulting in severe hyperchloremic acidosis. Anesthesiology 2005; 103:203.
  76. Gheorghe C, Dadu R, Blot C, et al. Hyperchloremic metabolic acidosis following resuscitation of shock. Chest 2010; 138:1521.
  77. Burdett E, Dushianthan A, Bennett-Guerrero E, et al. Perioperative buffered versus non-buffered fluid administration for surgery in adults. Cochrane Database Syst Rev 2012; 12:CD004089.
  78. Orbegozo Cortés D, Rayo Bonor A, Vincent JL. Isotonic crystalloid solutions: a structured review of the literature. Br J Anaesth 2014; 112:968.
  79. Hahn RG. Changing practices of fluid therapy. Acta Anaesthesiol Scand 2017; 61:576.
  80. Pfortmueller CA, Funk GC, Reiterer C, et al. Normal saline versus a balanced crystalloid for goal-directed perioperative fluid therapy in major abdominal surgery: a double-blind randomised controlled study. Br J Anaesth 2018; 120:274.
  81. Boer C, Bossers SM, Koning NJ. Choice of fluid type: physiological concepts and perioperative indications. Br J Anaesth 2018; 120:384.
  82. Yunos NM, Bellomo R, Hegarty C, et al. Association between a chloride-liberal vs chloride-restrictive intravenous fluid administration strategy and kidney injury in critically ill adults. JAMA 2012; 308:1566.
  83. Semler MW, Self WH, Wanderer JP, et al. Balanced Crystalloids versus Saline in Critically Ill Adults. N Engl J Med 2018; 378:829.
  84. Self WH, Semler MW, Wanderer JP, et al. Balanced Crystalloids versus Saline in Noncritically Ill Adults. N Engl J Med 2018; 378:819.
  85. Kellum JA. Abnormal saline and the history of intravenous fluids. Nat Rev Nephrol 2018; 14:358.
  86. Finfer S, Myburgh J, Bellomo R. Intravenous fluid therapy in critically ill adults. Nat Rev Nephrol 2018; 14:541.
  87. Maheshwari K, Turan A, Makarova N, et al. Saline versus Lactated Ringer's Solution: The Saline or Lactated Ringer's (SOLAR) Trial. Anesthesiology 2020; 132:614.
  88. Kawano-Dourado L, Zampieri FG, Azevedo LCP, et al. Low- Versus High-Chloride Content Intravenous Solutions for Critically Ill and Perioperative Adult Patients: A Systematic Review and Meta-analysis. Anesth Analg 2018; 126:513.
  89. Ertmer C, Rehberg S, Van Aken H, Westphal M. Relevance of non-albumin colloids in intensive care medicine. Best Pract Res Clin Anaesthesiol 2009; 23:193.
  90. Lewis SR, Pritchard MW, Evans DJ, et al. Colloids versus crystalloids for fluid resuscitation in critically ill people. Cochrane Database Syst Rev 2018; 8:CD000567.
  91. Heming N, Lamothe L, Jaber S, et al. Morbidity and Mortality of Crystalloids Compared to Colloids in Critically Ill Surgical Patients: A Subgroup Analysis of a Randomized Trial. Anesthesiology 2018; 129:1149.
  92. Kabon B, Sessler DI, Kurz A, Crystalloid–Colloid Study Team. Effect of Intraoperative Goal-directed Balanced Crystalloid versus Colloid Administration on Major Postoperative Morbidity: A Randomized Trial. Anesthesiology 2019; 130:728.
  93. Joosten A, Delaporte A, Ickx B, et al. Crystalloid versus Colloid for Intraoperative Goal-directed Fluid Therapy Using a Closed-loop System: A Randomized, Double-blinded, Controlled Trial in Major Abdominal Surgery. Anesthesiology 2018; 128:55.
  94. Joosten A, Delaporte A, Mortier J, et al. Long-term Impact of Crystalloid versus Colloid Solutions on Renal Function and Disability-free Survival after Major Abdominal Surgery. Anesthesiology 2019; 130:227.
  95. He H, Liu D, Ince C. Colloids and the Microcirculation. Anesth Analg 2018; 126:1747.
  96. McLean DJ, Shaw AD. Intravenous fluids: effects on renal outcomes. Br J Anaesth 2018; 120:397.
  97. Opperer M, Poeran J, Rasul R, et al. Use of perioperative hydroxyethyl starch 6% and albumin 5% in elective joint arthroplasty and association with adverse outcomes: a retrospective population based analysis. BMJ 2015; 350:h1567.
  98. Ryhammer PK, Tang M, Hoffmann-Petersen J, et al. Colloids in Cardiac Surgery-Friend or Foe? J Cardiothorac Vasc Anesth 2017; 31:1639.
  99. Kammerer T, Brettner F, Hilferink S, et al. No Differences in Renal Function between Balanced 6% Hydroxyethyl Starch (130/0.4) and 5% Albumin for Volume Replacement Therapy in Patients Undergoing Cystectomy: A Randomized Controlled Trial. Anesthesiology 2018; 128:67.
  100. Hasselgren E, Zdolsek M, Zdolsek JH, et al. Long Intravascular Persistence of 20% Albumin in Postoperative Patients. Anesth Analg 2019; 129:1232.
  101. Löffel LM, Hahn RG, Engel D, Wuethrich PY. Intraoperative Intravascular Effect of Lactated Ringer's Solution and Hyperoncotic Albumin During Hemorrhage in Cystectomy Patients. Anesth Analg 2021; 133:413.
  102. Wigmore GJ, Anstey JR, St John A, et al. 20% Human Albumin Solution Fluid Bolus Administration Therapy in Patients After Cardiac Surgery (the HAS FLAIR Study). J Cardiothorac Vasc Anesth 2019; 33:2920.
  103. McConnell M, Baisden J, Duncan AE. Pro: Third-Generation Hydroxyethyl Starch Solution Is Safe and Effective for Plasma Volume Expansion During Cardiac Surgery. J Cardiothorac Vasc Anesth 2018; 32:570.
  104. Futier E, Garot M, Godet T, et al. Effect of Hydroxyethyl Starch vs Saline for Volume Replacement Therapy on Death or Postoperative Complications Among High-Risk Patients Undergoing Major Abdominal Surgery: The FLASH Randomized Clinical Trial. JAMA 2020; 323:225.
  105. Dart AB, Mutter TC, Ruth CA, Taback SP. Hydroxyethyl starch (HES) versus other fluid therapies: effects on kidney function. Cochrane Database Syst Rev 2010; :CD007594.
  106. Navickis RJ, Haynes GR, Wilkes MM. Effect of hydroxyethyl starch on bleeding after cardiopulmonary bypass: a meta-analysis of randomized trials. J Thorac Cardiovasc Surg 2012; 144:223.
  107. Pagel JI, Rehm M, Kammerer T, et al. Hydroxyethyl Starch 130/0.4 and Its Impact on Perioperative Outcome: A Propensity Score Matched Controlled Observation Study. Anesth Analg 2018; 126:1949.
  108. Van Der Linden P, James M, Mythen M, Weiskopf RB. Safety of modern starches used during surgery. Anesth Analg 2013; 116:35.
  109. Gillies MA, Habicher M, Jhanji S, et al. Incidence of postoperative death and acute kidney injury associated with i.v. 6% hydroxyethyl starch use: systematic review and meta-analysis. Br J Anaesth 2014; 112:25.
  110. Tobey R, Cheng H, Gao M, et al. Postoperative Acute Kidney Injury and Blood Product Transfusion After Synthetic Colloid Use During Cardiac Surgery. J Cardiothorac Vasc Anesth 2017; 31:853.
  111. Hartog CS, Reuter D, Loesche W, et al. Influence of hydroxyethyl starch (HES) 130/0.4 on hemostasis as measured by viscoelastic device analysis: a systematic review. Intensive Care Med 2011; 37:1725.
  112. Sacchet-Cardozo F, Stoicea N, Joseph N, et al. Con: Hetastarch Should be Avoided for Volume Expansion in Cardiac Surgery Patients. J Cardiothorac Vasc Anesth 2018; 32:576.
  113. Kozek-Langenecker SA. Effects of hydroxyethyl starch solutions on hemostasis. Anesthesiology 2005; 103:654.
  114. Kozek-Langenecker SA, Jungheinrich C, Sauermann W, Van der Linden P. The effects of hydroxyethyl starch 130/0.4 (6%) on blood loss and use of blood products in major surgery: a pooled analysis of randomized clinical trials. Anesth Analg 2008; 107:382.
  115. Koponen T, Musialowicz T, Lahtinen P. Gelatin and the risk of bleeding after cardiac surgery. Acta Anaesthesiol Scand 2020; 64:1438.
  116. Thomas-Rueddel DO, Vlasakov V, Reinhart K, et al. Safety of gelatin for volume resuscitation--a systematic review and meta-analysis. Intensive Care Med 2012; 38:1134.
  117. Lambert KG, Wakim JH, Lambert NE. Preoperative fluid bolus and reduction of postoperative nausea and vomiting in patients undergoing laparoscopic gynecologic surgery. AANA J 2009; 77:110.
  118. Gan TJ, Diemunsch P, Habib AS, et al. Consensus guidelines for the management of postoperative nausea and vomiting. Anesth Analg 2014; 118:85.
  119. British Consensus Guidelines on Intravenous Fluid Therapy for Adult Surgical Patients (GIFTASUP). London: NHS National Library of Health. http://www.ics.ac.uk/intensive_care_professional/standards_and_guidelines/british_consensus_guidelines_on_intravenous_fluid_therapy_for_adult_surgical_patients__giftasup__2008 (Accessed on September 18, 2009).
  120. Bundgaard-Nielsen M, Holte K, Secher NH, Kehlet H. Monitoring of peri-operative fluid administration by individualized goal-directed therapy. Acta Anaesthesiol Scand 2007; 51:331.
  121. Bundgaard-Nielsen M, Secher NH, Kehlet H. 'Liberal' vs. 'restrictive' perioperative fluid therapy--a critical assessment of the evidence. Acta Anaesthesiol Scand 2009; 53:843.
  122. Hamilton MA, Cecconi M, Rhodes A. A systematic review and meta-analysis on the use of preemptive hemodynamic intervention to improve postoperative outcomes in moderate and high-risk surgical patients. Anesth Analg 2011; 112:1392.
  123. Yates DR, Davies SJ, Milner HE, Wilson RJ. Crystalloid or colloid for goal-directed fluid therapy in colorectal surgery. Br J Anaesth 2014; 112:281.
  124. Hartog CS, Kohl M, Reinhart K. A systematic review of third-generation hydroxyethyl starch (HES 130/0.4) in resuscitation: safety not adequately addressed. Anesth Analg 2011; 112:635.
  125. Orbegozo Cortés D, Gamarano Barros T, Njimi H, Vincent JL. Crystalloids versus colloids: exploring differences in fluid requirements by systematic review and meta-regression. Anesth Analg 2015; 120:389.
  126. Finfer S, Bellomo R, Boyce N, et al. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350:2247.
  127. Brandstrup B, Svensen C, Engquist A. Hemorrhage and operation cause a contraction of the extracellular space needing replacement--evidence and implications? A systematic review. Surgery 2006; 139:419.
  128. Myles PS, Andrews S, Nicholson J, et al. Contemporary Approaches to Perioperative IV Fluid Therapy. World J Surg 2017; 41:2457.
  129. Bellomo R. Noradrenaline: friend or foe? Heart Lung Circ 2003; 12 Suppl 2:S42.
  130. Sear JW. Kidney dysfunction in the postoperative period. Br J Anaesth 2005; 95:20.
  131. Brandstrup B. Finding the Right Balance. N Engl J Med 2018; 378:2335.
  132. Rahbari NN, Zimmermann JB, Schmidt T, et al. Meta-analysis of standard, restrictive and supplemental fluid administration in colorectal surgery. Br J Surg 2009; 96:331.
  133. Lobo SM, Ronchi LS, Oliveira NE, et al. Restrictive strategy of intraoperative fluid maintenance during optimization of oxygen delivery decreases major complications after high-risk surgery. Crit Care 2011; 15:R226.
  134. Corcoran T, Rhodes JE, Clarke S, et al. Perioperative fluid management strategies in major surgery: a stratified meta-analysis. Anesth Analg 2012; 114:640.
  135. Furrer MA, Schneider MP, Löffel LM, et al. Impact of intra-operative fluid and noradrenaline administration on early postoperative renal function after cystectomy and urinary diversion: A retrospective observational cohort study. Eur J Anaesthesiol 2018; 35:641.
  136. Wuethrich PY, Burkhard FC, Thalmann GN, et al. Restrictive deferred hydration combined with preemptive norepinephrine infusion during radical cystectomy reduces postoperative complications and hospitalization time: a randomized clinical trial. Anesthesiology 2014; 120:365.
  137. Chong MA, Wang Y, Berbenetz NM, McConachie I. Does goal-directed haemodynamic and fluid therapy improve peri-operative outcomes?: A systematic review and meta-analysis. Eur J Anaesthesiol 2018; 35:469.
  138. Biais M, de Courson H, Lanchon R, et al. Mini-fluid Challenge of 100 ml of Crystalloid Predicts Fluid Responsiveness in the Operating Room. Anesthesiology 2017; 127:450.
  139. Messina A, Longhini F, Coppo C, et al. Use of the Fluid Challenge in Critically Ill Adult Patients: A Systematic Review. Anesth Analg 2017; 125:1532.
  140. Wrzosek A, Jakowicka-Wordliczek J, Zajaczkowska R, et al. Perioperative restrictive versus goal-directed fluid therapy for adults undergoing major non-cardiac surgery. Cochrane Database Syst Rev 2019; 12:CD012767.
  141. Giglio MT, Marucci M, Testini M, Brienza N. Goal-directed haemodynamic therapy and gastrointestinal complications in major surgery: a meta-analysis of randomized controlled trials. Br J Anaesth 2009; 103:637.
  142. Grocott MP, Dushianthan A, Hamilton MA, et al. Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery. Cochrane Database Syst Rev 2012; 11:CD004082.
  143. Rollins KE, Lobo DN. Intraoperative Goal-directed Fluid Therapy in Elective Major Abdominal Surgery: A Meta-analysis of Randomized Controlled Trials. Ann Surg 2016; 263:465.
  144. Benes J, Giglio M, Brienza N, Michard F. The effects of goal-directed fluid therapy based on dynamic parameters on post-surgical outcome: a meta-analysis of randomized controlled trials. Crit Care 2014; 18:584.
  145. Pearse RM, Harrison DA, MacDonald N, et al. Effect of a perioperative, cardiac output-guided hemodynamic therapy algorithm on outcomes following major gastrointestinal surgery: a randomized clinical trial and systematic review. JAMA 2014; 311:2181.
  146. Calvo-Vecino JM, Ripollés-Melchor J, Mythen MG, et al. Effect of goal-directed haemodynamic therapy on postoperative complications in low-moderate risk surgical patients: a multicentre randomised controlled trial (FEDORA trial). Br J Anaesth 2018; 120:734.
  147. Kehlet H, Joshi GP. Systematic Reviews and Meta-Analyses of Randomized Controlled Trials on Perioperative Outcomes: An Urgent Need for Critical Reappraisal. Anesth Analg 2015; 121:1104.
  148. Gillies MA, Pearse R, Chew MS. Peri-operative goal-directed therapy: A definitive answer remains elusive. Eur J Anaesthesiol 2018; 35:467.
  149. Kaufmann T, Clement RP, Scheeren TWL, et al. Perioperative goal-directed therapy: A systematic review without meta-analysis. Acta Anaesthesiol Scand 2018; 62:1340.
  150. Joshi GP, Kehlet H. CON: Perioperative Goal-Directed Fluid Therapy Is an Essential Element of an Enhanced Recovery Protocol? Anesth Analg 2016; 122:1261.
  151. Minto G, Scott MJ, Miller TE. Monitoring needs and goal-directed fluid therapy within an enhanced recovery program. Anesthesiol Clin 2015; 33:35.
  152. Gómez-Izquierdo JC, Feldman LS, Carli F, Baldini G. Meta-analysis of the effect of goal-directed therapy on bowel function after abdominal surgery. Br J Surg 2015; 102:577.
  153. Miller TE, Roche AM, Mythen M. Fluid management and goal-directed therapy as an adjunct to Enhanced Recovery After Surgery (ERAS). Can J Anaesth 2015; 62:158.
  154. Lassen K. Intravenous fluid therapy. Br J Surg 2009; 96:123.
  155. Lobo DN. Fluid overload and surgical outcome: another piece in the jigsaw. Ann Surg 2009; 249:186.
  156. Hatton GE, Du RE, Wei S, et al. Positive Fluid Balance and Association with Post-Traumatic Acute Kidney Injury. J Am Coll Surg 2020; 230:190.
Topic 14942 Version 52.0

References

1 : Blood volume is normal after pre-operative overnight fasting.

2 : Fluid deficits during prolonged overnight fasting in young healthy adults.

3 : Perioperative Fluid Therapy for Major Surgery.

4 : Volume kinetic analysis of the distribution of 0.9% saline in conscious versus isoflurane-anesthetized sheep.

5 : Isoflurane but not mechanical ventilation promotes extravascular fluid accumulation during crystalloid volume loading.

6 : The 'third space'--fact or fiction?

7 : Water loss by evaporation from the abdominal cavity during surgery.

8 : Perioperative Fluid Utilization Variability and Association With Outcomes: Considerations for Enhanced Recovery Efforts in Sample US Surgical Populations.

9 : Wet, dry or something else?

10 : Effects of Intraoperative Fluid Management on Postoperative Outcomes: A Hospital Registry Study.

11 : Update on Perioperative Acute Kidney Injury.

12 : Effect of preoperative fluid therapy on hemodynamic stability during anesthesia induction, a randomized study.

13 : A rational approach to perioperative fluid management.

14 : Fluid therapy for the surgical patient.

15 : Postoperative fluid overload: not a benign problem.

16 : Pathophysiology and clinical implications of perioperative fluid excess.

17 : Fatal postoperative pulmonary edema: pathogenesis and literature review.

18 : Effect of intraoperative fluid management on outcome after intraabdominal surgery.

19 : Mechanism of acute ascites formation after trauma resuscitation.

20 : Supranormal trauma resuscitation causes more cases of abdominal compartment syndrome.

21 : Will This Hemodynamically Unstable Patient Respond to a Bolus of Intravenous Fluids?

22 : Intraoperative fluid restriction improves outcome after major elective gastrointestinal surgery.

23 : Dynamic changes in arterial waveform derived variables and fluid responsiveness in mechanically ventilated patients: a systematic review of the literature.

24 : Does central venous pressure predict fluid responsiveness? A systematic review of the literature and the tale of seven mares.

25 : Fluid status and fluid responsiveness.

26 : Central venous pressure cannot predict fluid-responsiveness.

27 : Venous function and central venous pressure: a physiologic story.

28 : Systematic review including re-analyses of 1148 individual data sets of central venous pressure as a predictor of fluid responsiveness.

29 : Central venous pressure monitoring: clinical insights beyond the numbers.

30 : Does the central venous pressure predict fluid responsiveness? An updated meta-analysis and a plea for some common sense.

31 : Predictors of postoperative acute renal failure after noncardiac surgery in patients with previously normal renal function.

32 : Intraoperative urinary output does not predict postoperative renal function in patients undergoing abdominal aortic revascularization.

33 : Targeting Oliguria Reversal in Goal-Directed Hemodynamic Management Does Not Reduce Renal Dysfunction in Perioperative and Critically Ill Patients: A Systematic Review and Meta-Analysis.

34 : Intraoperative oliguria predicts acute kidney injury after major abdominal surgery.

35 : Association Between Intraoperative Oliguria and Acute Kidney Injury After Major Noncardiac Surgery.

36 : Restrictive versus Liberal Fluid Therapy for Major Abdominal Surgery.

37 : Importance of intraoperative oliguria during major abdominal surgery: findings of the Restrictive versus Liberal Fluid Therapy in Major Abdominal Surgery trial.

38 : Intraoperative permissive oliguria - how much is too much?

39 : The urine output definition of acute kidney injury is too liberal.

40 : Monitoring fluid therapy.

41 : Microcirculatory function monitoring at the bedside--a view from the intensive care.

42 : Minimally invasive cardiac output monitoring in the perioperative setting.

43 : Using ventilation-induced plethysmographic variations to optimize patient fluid status.

44 : What is a fluid challenge?

45 : Physiological controversies and methods used to determine fluid responsiveness: a qualitative systematic review.

46 : Inter-device differences in monitoring for goal-directed fluid therapy.

47 : The ability of anesthesia providers to visually estimate systolic pressure variability using the "eyeball" technique.

48 : Prediction of fluid responsiveness: an update.

49 : Pressure Waveform Analysis.

50 : Does Respiratory Variation in Inferior Vena Cava Diameter Predict Fluid Responsiveness in Mechanically Ventilated Patients? A Systematic Review and Meta-analysis.

51 : Assessing the diagnostic accuracy of pulse pressure variations for the prediction of fluid responsiveness: a "gray zone" approach.

52 : Validation of pulse pressure variation and corrected flow time as predictors of fluid responsiveness in patients in the prone position.

53 : Abilities of pulse pressure variations and stroke volume variations to predict fluid responsiveness in prone position during scoliosis surgery.

54 : Fluid Challenge During Anesthesia: A Systematic Review and Meta-analysis.

55 : Stroke Volume Variation and Pulse Pressure Variation Are Not Useful for Predicting Fluid Responsiveness in Thoracic Surgery.

56 : Hemodynamic monitoring in thoracic surgical patients.

57 : Rational fluid management: dissecting facts from fiction.

58 : Dynamic indices do not predict volume responsiveness in routine clinical practice.

59 : Systematic review of the literature for the use of oesophageal Doppler monitor for fluid replacement in major abdominal surgery.

60 : Improving perioperative outcomes: fluid optimization with the esophageal Doppler monitor, a metaanalysis and review.

61 : Optimising stroke volume and oxygen delivery in abdominal aortic surgery: a randomised controlled trial.

62 : Guidelines for the use of echocardiography as a monitor for therapeutic intervention in adults: a report from the American Society of Echocardiography.

63 : Special article: basic perioperative transesophageal echocardiography examination: a consensus statement of the American Society of Echocardiography and the Society of Cardiovascular Anesthesiologists.

64 : Does the Device Matter in Goal-Directed Fluid Therapy?

65 : Accuracy and precision of non-invasive cardiac output monitoring devices in perioperative medicine: a systematic review and meta-analysis†.

66 : The ideal crystalloid - what is 'balanced'?

67 : Perioperative Hyperglycemia Management: An Update.

68 : Perioperative administration of buffered versus non-buffered crystalloid intravenous fluid to improve outcomes following adult surgical procedures.

69 : Meta-analysis of high- versus low-chloride content in perioperative and critical care fluid resuscitation.

70 : Major complications, mortality, and resource utilization after open abdominal surgery: 0.9% saline compared to Plasma-Lyte.

71 : Chloride Content of Fluids Used for Large-Volume Resuscitation Is Associated With Reduced Survival.

72 : Rapid saline infusion produces hyperchloremic acidosis in patients undergoing gynecologic surgery.

73 : The effects of balanced versus saline-based hetastarch and crystalloid solutions on acid-base and electrolyte status and gastric mucosal perfusion in elderly surgical patients.

74 : A randomized, controlled, double-blind crossover study on the effects of 2-L infusions of 0.9% saline and plasma-lyte®148 on renal blood flow velocity and renal cortical tissue perfusion in healthy volunteers.

75 : Isotonic fluid absorption during hysteroscopy resulting in severe hyperchloremic acidosis.

76 : Hyperchloremic metabolic acidosis following resuscitation of shock.

77 : Perioperative buffered versus non-buffered fluid administration for surgery in adults.

78 : Isotonic crystalloid solutions: a structured review of the literature.

79 : Changing practices of fluid therapy.

80 : Normal saline versus a balanced crystalloid for goal-directed perioperative fluid therapy in major abdominal surgery: a double-blind randomised controlled study.

81 : Choice of fluid type: physiological concepts and perioperative indications.

82 : Association between a chloride-liberal vs chloride-restrictive intravenous fluid administration strategy and kidney injury in critically ill adults.

83 : Balanced Crystalloids versus Saline in Critically Ill Adults.

84 : Balanced Crystalloids versus Saline in Noncritically Ill Adults.

85 : Abnormal saline and the history of intravenous fluids.

86 : Intravenous fluid therapy in critically ill adults.

87 : Saline versus Lactated Ringer's Solution: The Saline or Lactated Ringer's (SOLAR) Trial.

88 : Low- Versus High-Chloride Content Intravenous Solutions for Critically Ill and Perioperative Adult Patients: A Systematic Review and Meta-analysis.

89 : Relevance of non-albumin colloids in intensive care medicine.

90 : Colloids versus crystalloids for fluid resuscitation in critically ill people.

91 : Morbidity and Mortality of Crystalloids Compared to Colloids in Critically Ill Surgical Patients: A Subgroup Analysis of a Randomized Trial.

92 : Effect of Intraoperative Goal-directed Balanced Crystalloid versus Colloid Administration on Major Postoperative Morbidity: A Randomized Trial.

93 : Crystalloid versus Colloid for Intraoperative Goal-directed Fluid Therapy Using a Closed-loop System: A Randomized, Double-blinded, Controlled Trial in Major Abdominal Surgery.

94 : Long-term Impact of Crystalloid versus Colloid Solutions on Renal Function and Disability-free Survival after Major Abdominal Surgery.

95 : Colloids and the Microcirculation.

96 : Intravenous fluids: effects on renal outcomes.

97 : Use of perioperative hydroxyethyl starch 6% and albumin 5% in elective joint arthroplasty and association with adverse outcomes: a retrospective population based analysis.

98 : Colloids in Cardiac Surgery-Friend or Foe?

99 : No Differences in Renal Function between Balanced 6% Hydroxyethyl Starch (130/0.4) and 5% Albumin for Volume Replacement Therapy in Patients Undergoing Cystectomy: A Randomized Controlled Trial.

100 : Long Intravascular Persistence of 20% Albumin in Postoperative Patients.

101 : Intraoperative Intravascular Effect of Lactated Ringer's Solution and Hyperoncotic Albumin During Hemorrhage in Cystectomy Patients.

102 : 20% Human Albumin Solution Fluid Bolus Administration Therapy in Patients After Cardiac Surgery (the HAS FLAIR Study).

103 : Pro: Third-Generation Hydroxyethyl Starch Solution Is Safe and Effective for Plasma Volume Expansion During Cardiac Surgery.

104 : Effect of Hydroxyethyl Starch vs Saline for Volume Replacement Therapy on Death or Postoperative Complications Among High-Risk Patients Undergoing Major Abdominal Surgery: The FLASH Randomized Clinical Trial.

105 : Hydroxyethyl starch (HES) versus other fluid therapies: effects on kidney function.

106 : Effect of hydroxyethyl starch on bleeding after cardiopulmonary bypass: a meta-analysis of randomized trials.

107 : Hydroxyethyl Starch 130/0.4 and Its Impact on Perioperative Outcome: A Propensity Score Matched Controlled Observation Study.

108 : Safety of modern starches used during surgery.

109 : Incidence of postoperative death and acute kidney injury associated with i.v. 6% hydroxyethyl starch use: systematic review and meta-analysis.

110 : Postoperative Acute Kidney Injury and Blood Product Transfusion After Synthetic Colloid Use During Cardiac Surgery.

111 : Influence of hydroxyethyl starch (HES) 130/0.4 on hemostasis as measured by viscoelastic device analysis: a systematic review.

112 : Con: Hetastarch Should be Avoided for Volume Expansion in Cardiac Surgery Patients.

113 : Effects of hydroxyethyl starch solutions on hemostasis.

114 : The effects of hydroxyethyl starch 130/0.4 (6%) on blood loss and use of blood products in major surgery: a pooled analysis of randomized clinical trials.

115 : Gelatin and the risk of bleeding after cardiac surgery.

116 : Safety of gelatin for volume resuscitation--a systematic review and meta-analysis.

117 : Preoperative fluid bolus and reduction of postoperative nausea and vomiting in patients undergoing laparoscopic gynecologic surgery.

118 : Consensus guidelines for the management of postoperative nausea and vomiting.

119 : Consensus guidelines for the management of postoperative nausea and vomiting.

120 : Monitoring of peri-operative fluid administration by individualized goal-directed therapy.

121 : 'Liberal' vs. 'restrictive' perioperative fluid therapy--a critical assessment of the evidence.

122 : A systematic review and meta-analysis on the use of preemptive hemodynamic intervention to improve postoperative outcomes in moderate and high-risk surgical patients.

123 : Crystalloid or colloid for goal-directed fluid therapy in colorectal surgery.

124 : A systematic review of third-generation hydroxyethyl starch (HES 130/0.4) in resuscitation: safety not adequately addressed.

125 : Crystalloids versus colloids: exploring differences in fluid requirements by systematic review and meta-regression.

126 : A comparison of albumin and saline for fluid resuscitation in the intensive care unit.

127 : Hemorrhage and operation cause a contraction of the extracellular space needing replacement--evidence and implications? A systematic review.

128 : Contemporary Approaches to Perioperative IV Fluid Therapy.

129 : Noradrenaline: friend or foe?

130 : Kidney dysfunction in the postoperative period.

131 : Finding the Right Balance.

132 : Meta-analysis of standard, restrictive and supplemental fluid administration in colorectal surgery.

133 : Restrictive strategy of intraoperative fluid maintenance during optimization of oxygen delivery decreases major complications after high-risk surgery.

134 : Perioperative fluid management strategies in major surgery: a stratified meta-analysis.

135 : Impact of intra-operative fluid and noradrenaline administration on early postoperative renal function after cystectomy and urinary diversion: A retrospective observational cohort study.

136 : Restrictive deferred hydration combined with preemptive norepinephrine infusion during radical cystectomy reduces postoperative complications and hospitalization time: a randomized clinical trial.

137 : Does goal-directed haemodynamic and fluid therapy improve peri-operative outcomes?: A systematic review and meta-analysis.

138 : Mini-fluid Challenge of 100 ml of Crystalloid Predicts Fluid Responsiveness in the Operating Room.

139 : Use of the Fluid Challenge in Critically Ill Adult Patients: A Systematic Review.

140 : Perioperative restrictive versus goal-directed fluid therapy for adults undergoing major non-cardiac surgery.

141 : Goal-directed haemodynamic therapy and gastrointestinal complications in major surgery: a meta-analysis of randomized controlled trials.

142 : Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery.

143 : Intraoperative Goal-directed Fluid Therapy in Elective Major Abdominal Surgery: A Meta-analysis of Randomized Controlled Trials.

144 : The effects of goal-directed fluid therapy based on dynamic parameters on post-surgical outcome: a meta-analysis of randomized controlled trials.

145 : Effect of a perioperative, cardiac output-guided hemodynamic therapy algorithm on outcomes following major gastrointestinal surgery: a randomized clinical trial and systematic review.

146 : Effect of goal-directed haemodynamic therapy on postoperative complications in low-moderate risk surgical patients: a multicentre randomised controlled trial (FEDORA trial).

147 : Systematic Reviews and Meta-Analyses of Randomized Controlled Trials on Perioperative Outcomes: An Urgent Need for Critical Reappraisal.

148 : Peri-operative goal-directed therapy: A definitive answer remains elusive.

149 : Perioperative goal-directed therapy: A systematic review without meta-analysis.

150 : CON: Perioperative Goal-Directed Fluid Therapy Is an Essential Element of an Enhanced Recovery Protocol?

151 : Monitoring needs and goal-directed fluid therapy within an enhanced recovery program.

152 : Meta-analysis of the effect of goal-directed therapy on bowel function after abdominal surgery.

153 : Fluid management and goal-directed therapy as an adjunct to Enhanced Recovery After Surgery (ERAS).

154 : Intravenous fluid therapy.

155 : Fluid overload and surgical outcome: another piece in the jigsaw.

156 : Positive Fluid Balance and Association with Post-Traumatic Acute Kidney Injury.