CC BY-NC-ND 4.0 · Journal of Child Science 2017; 07(01): e60-e75
DOI: 10.1055/s-0037-1603897
Review Article
Georg Thieme Verlag KG Stuttgart · New York

Pediatric Sepsis: Clinical Considerations

G. Balázs
1   Clinical Center, Institute of Pediatrics, University of Debrecen, Debrecen, Hungary
,
S. Szima
2   Department of Pediatrics, Medical Center Coburg, Academic Hospital of the University of Split, Coburg, Germany
,
N. Elek
1   Clinical Center, Institute of Pediatrics, University of Debrecen, Debrecen, Hungary
,
P. Dahlem
2   Department of Pediatrics, Medical Center Coburg, Academic Hospital of the University of Split, Coburg, Germany
› Author Affiliations
Further Information

Address for correspondence

G. Balázs, MD
Clinical Center, Institute of Pediatrics, University of Debrecen
Nagyerdei krt 98, H-4032 Debrecen
Hungary   

Publication History

14 January 2017

25 April 2017

Publication Date:
26 July 2017 (online)

 

Abstract

Sepsis remains one of the leading causes of childhood mortality today, although survival has substantially improved, thanks to an ever deeper and more thorough understanding of sepsis pathophysiology, developments in vaccination and intensive therapy, and goal-directed therapeutic approaches. The key to successful therapy is early recognition and antibiotic treatment, as well as supportive therapy aimed at correcting circulatory, respiratory, and metabolic derangements as soon as possible. Diagnosis and management of childhood sepsis mainly evolves along the path of studies conducted in adult patients; however, their applicability is limited due to a variability of cardiovascular and immune responses in each age group. In addition to childhood characteristics of hemodynamic responses in sepsis, this review looks at major areas of pediatric sepsis therapy, with the intention of providing bedside clinicians with pointers useful in their day-to-day work.


#

Introduction

Sepsis is a systemic inflammatory response syndrome (SIRS) triggered by a suspected or verified pathogen or its product;[1] our understanding of its pathomechanism has been on the rise.[2] [3] The septic process is an extremely complex, simultaneous interplay of proinflammatory and anti-inflammatory responses.[4] [5] [6] The direction, extent, and magnitude of the host response are determined, on the one hand, by patient-specific factors (e.g., genetic predisposition, age, vaccination status, comorbidity, severe injury, medication taken, use of invasive equipment) and, on the other hand, by virulence factors of the pathogen (mostly bacteria, also viruses, fungi, parasites).[7] [8] [9] The disease process has an effect on practically all organ systems; sepsis is often accompanied by the disruption of cardiovascular function, intravascular volume status, respiratory function, immune/inflammatory regulation, renal function, coagulation, and hepatic function.[10] Childhood sepsis is a global health problem, with incidence and outcomes varying across regions.[11] Sepsis is one of the leading causes of death in children;[12] its mortality is between 2 and 10%,[13] [14] [15] which is much lower than in adults.[16] Hospital mortality in the United States was found to be 2% in healthy children, and 8% in patients suffering from chronic diseases.[12] For more than two-thirds of the cases, airway or bloodstream infections are identified in the background.[11] [17] [18] The most vulnerable pediatric groups include preterm newborns, infants, sufferers from hereditary or acquired immune deficiencies, chronic debilitating medical conditions, severe trauma victims, and those recovering from major surgery or implanted with invasive medical equipment.[19] [20] Early detection and adequate first line treatment are essential: every single hour spent in hypotension and with capillary refill longer than 3 seconds doubles the chance of mortality.[21] Thanks to a therapeutic strategy that emphasizes early diagnosis, aggressive fluid therapy, and vasoactive support, as well antibiotic treatment as early as possible, the mortality of childhood sepsis is gradually decreasing;[22] however, a growing number of children with predisposing comorbidities and a rise in sepsis prevalence due to the spread of multidrug resistant pathogens continue to challenge health care providers worldwide.[11] The objective of this review is to provide a summary of the clinical management of childhood sepsis and septic shock.


#

Diagnosis

Historically, the term sepsis was widely used in relation to a diverse group of syndromes, which often confused clinicians.[23] In 1992, as part of standardization efforts for the definition, the American College of Chest Physicians and the Society of Critical Care Medicine introduced the term systemic inflammatory response syndrome, defined as a widespread inflammatory response that may or may not be associated with infection.[24] In 2005, the International Pediatric Consensus Conference adapted the definitions of SIRS, sepsis, and other related terms ([Tables 1] [2]) to pediatric age groups ([Table 3]) by introducing age-specific heart rate, respiratory rate, and white blood cell count cutoffs ([Table 4]). In addition to facilitating observational study design and the assessment of various therapeutic interventions in clinical trials, this set of distinct definitions (sepsis, severe sepsis and septic shock, organ failure) serves as a guide to clinicians in severity assessment, progression monitoring, and treatment response evaluation.[25] [26] However, it is important to note that the diagnosis of childhood sepsis should not be confined to a predefined set of pathophysiological and laboratory deviations: in an observational study at an academic medical center, only two-thirds of patients under the age of 18 years treated for sepsis or septic shock satisfied the strict requirements of the consensus system of criteria.[27] Lately, more and more clinicians question the usefulness of SIRS criteria in sepsis and call for separate sets of definitions for clinical versus research use.[28] [29] In February 2016, new definitions and clinical criteria of adult sepsis and septic shock were introduced at the 45th Annual Critical Care Congress.[30]

Table 1

Definitions of SIRS, infection, sepsis, severe sepsis, and septic shock

SIRS

The presence of at least two of the following four criteria, one of which must be abnormal temperature or leukocyte count:

 Core temperature of >38.5°C or <36°C

 Tachycardia, defined as a mean heart rate >2 SD above normal for age in the absence of external stimulus, chronic drugs, or painful stimuli; or otherwise unexplained persistent elevation over a 0.5- to 4-h period or for children <1 y old: bradycardia, defined as a mean heart rate <10th percentile for age in the absence of external vagal stimulus, beta-blocker drugs, or congenital heart disease; or otherwise unexplained persistent depression over a 0.5-h time period

 Mean respiratory rate >2 SD above normal for age or mechanical ventilation for an acute process not related to underlying neuromuscular disease or the receipt of general anesthesia

 Leukocyte count elevated or depressed for age (not secondary to chemotherapy-induced leukopenia) or >10% immature neutrophils

Infection

A suspected or proven (by positive culture, tissue stain, or polymerase chain reaction test) infection caused by any pathogen or a clinical syndrome associated with a high probability of infection. Evidence of infection includes positive findings on clinical exam, imaging, or laboratory tests (e.g., white blood cells in a normally sterile body fluid, perforated viscus, chest radiograph consistent with pneumonia, petechial or purpuric rash, or purpura fulminans)

Sepsis

SIRS in the presence of or as a result of suspected or proven infection

Severe sepsis

Sepsis plus one of the following: cardiovascular organ dysfunction or acute respiratory distress syndrome or two or more other organ dysfunctions; organ dysfunctions are defined in [Table 2]

Septic shock

Sepsis and cardiovascular organ dysfunction as defined in [Table 2]

Abbreviations: SD, standard deviation; SIRS, systemic inflammatory response syndrome.


Source: Data from Goldstein et al.[1]


Table 2

Organ dysfunction criteria

Cardiovascular dysfunction

Despite administration of isotonic intravenous fluid bolus ≥40 mL/kg in 1 h:

Decrease in BP (hypotension) 5th percentile for age or systolic BP <2 SD below normal for age

or

Need for vasoactive drug to maintain BP in normal range (dopamine >5 µg/kg/min or dobutamine, epinephrine, or norepinephrine at any dose)

or two of the following

 Unexplained metabolic acidosis: base deficit >5 mEq/L

 Increased arterial lactate more than two times upper limit of normal

 Oliguria: urine output <0.5 mL/kg/h

 Prolonged capillary refill: >5 s

 Core to peripheral temperature gap >3°C

Respiratory

PaO2/FIO2 < 300 in the absence of cyanotic heart disease or preexisting lung disease

or

PaCO2 > 65 Torr or 20 mm Hg over baseline PaCO2

or

Proven need or >50% FiO2 to maintain saturation >92%

or

Need for nonelective invasive or noninvasive mechanical ventilation

Neurologic

Glasgow Coma Score 11

or

Acute change in mental status with a decrease in Glasgow Coma Score ≥3 points from abnormal baseline

Hematological

Platelet count of 80,000/mm3 or a decline of 50% in platelet count from highest value recorded over the past 3 d (for chronic hematology/oncology patients)

or

International normalized ratio > 2

Renal

Serum creatinine more than two times upper limit of normal for age or twofold increase in baseline creatinine

Hepatic

Total bilirubin < 4 mg/dL (not applicable for newborn)

or

ALT two times upper limit of normal for age

Abbreviations: ALT, alanine aminotransferase; BP, blood pressure; SD, standard deviation.


Source: Data from Goldstein et al.[1]


Table 3

Pediatric age groups for severe sepsis definitions

Newborn

0 d to 1 wk

Neonate

1 wk to 1 mo

Infant

1 mo to 1 y

Toddler and preschool

2–5 y

School age child

6–12 y

Adolescent and young adult

13 to <18 y

Source: Data from Goldstein et al.[1]


Table 4

Age-specific vital signs and laboratory variables[a]

Heart rate (beats/min)

Respiratory rate (breaths/min)

Leukocyte count (leukocytes 103/mm3)

Systolic blood pressure (mm Hg)

Age group

Tachycardia

Bradycardia

0 d to 1 wk

>180

<100

>50

>34

<65

1 wk to 1 mo

>180

<100

>40

>19.5 or <5

<75

1 mo to 1 y

>180

<90

>34

>17.5 or <5

<100

2–5 y

>140

NA

>22

>15.5 or <6

<94

6–12 y

>130

NA

>18

>13.5 or <4.5

<105

13 to <18 y

>110

NA

>14

>11 or <4.5

<117

a Lower values for heart rate, leukocyte count, and systolic blood pressure are for the fifth and upper values for heart rate, respiration rate, or leukocyte count for the 95th percentile.


Source: Data from Goldstein et al.[1]


Medical conditions elicited by any sort of proven or presumed pathogens are defined as infections. Infection can be confirmed by positive bacterial culture, tissue stain, or polymerase chain reaction (PCR) test. Infection is presumable in certain clinical syndromes (e.g., fever and coughing, appearance of petechiae and purpurae accompanied by hemodynamic instability, meningism, abscess, organ perforation); it should also be suspected when consistent with laboratory or imaging findings (e.g., pulmonary infiltration on chest X-ray, presence of leukocytes in normally sterile body fluids).[1]

In a great majority of sepsis cases, the site of infection is likely to be identified through medical history, physical examination, laboratory tests, and various imaging studies. To identify the pathogen, it is recommended to collect two peripheral blood cultures (both aerobic and anaerobic) prior to treatment, and an additional sample from any intravascular tubes in place for more than 48 hours.[31] [32] [33] The positive predictive value of testing can be improved, and contamination rates reduced, through proper sampling of at least 1 mL or, for older children, 3–10 mL of blood[33] [34] [35] [36] by a trained phlebotomist or experienced blood culture team[37] [38] [39] using sterile latex gloves and best-practice antisepsis.[40] In an effort to not cause pain or more discomfort, pediatricians often use intravenous catheters for blood culture sampling;[41] however, avoiding this practice might significantly reduce the number of false-positive blood cultures.[42] Sampling should include other cultures (e.g., urine, wound exudate, airway secretions, cerebrospinal fluid) as appropriate for the primary infection site.[16]

Culture growth becomes apparent within a few days; however, a negative result can only be confirmed after 5 days.[43] [44] In addition to time issues, another major limiting factor of the technique is low sensitivity, which can be aggravated by prior antibiotic treatment and, especially with pediatric patients, sample volume limitations.[45] Challenges posed by growth-inhibiting bacteriostatic substances, low pathogen concentrations, and long incubation times of methods with high detection thresholds can be overcome using molecular amplification techniques.[46] [47] [48] PCR assays capable of detecting up to 25 types of bacteria and fungi within 4 to 6 hours are available today;[49] [50] [51] [52] [53] their application may open new dimensions in sensitivity and speed, providing treatment targets at early stages of life-threatening infections.


#

Developmental Differences in the Hemodynamic Response to Sepsis in Children and Adults

One of the major challenges of pediatric critical care medicine is dealing with developmental differences that greatly impact the pathophysiology of sepsis.[7] Age-related changes in heart rate, stroke volume (SV), blood pressure, systemic vascular resistance (SVR), hemoglobin concentration, metabolic rate, glycogen stores, and protein mass all contribute to differences in disease progress between children and adults and to the heterogeneity of cardiovascular and metabolic responses across pediatric age groups.[7] [10] [54] [55] An essential requirement for successful treatment of childhood sepsis is the awareness of developmental differences and understanding of their effects on the function of various organ systems.

In septic shock, adult hemodynamic response is initially characterized by hyperdynamic (high-output) physiology, including low SVR, tachycardia, and normal or increased cardiac output (CO), as manifested by flash capillary refill, bounding pulses, warm and dry extremities, and wide pulse pressures (“warm” shock).[56] Myocardial dysfunction and reduced CO are compensated in adults through two mechanisms: elevation of heart rate and reduction of SVR.[7] [54] [55] [57] In the absence of adequate intervention, progressive SVR reduction might result in maldistribution of CO and organ hypoperfusion. It is a sign of poor prognosis when compensation fails or is insufficient; the risk of death is also higher in patients whose SVR reduction does not respond to vasopressors.[56] [58] [59] Pediatric septic shock is often accompanied by hypovolemia; children therefore respond well to aggressive fluid therapy.[55] [57] In contrast with the high-CO, low-SVR adult response, children's pathophysiology is dominated by increased SVR and myocardial dysfunction.[60] [61] [62] Children's cardiac reserves are limited. While adults tolerate even doubling of the resting heart rate to offset a decreased SV and maintain a sufficient CO fairly well, children do not have heart rate reserves for a similar increase. Although tachycardia is an important compensatory mechanism in children as well, the younger the patient, the more likely it is for the response to become inadequate with a critical heart rate increase; with the shortening of diastolic filling time and a consequential decrease of coronary perfusion, CO may continue to decline.[10] [55] In addition to tachycardia, newborns, infants, and children maintain blood pressure by increasing SVR; in this case, clinical appearance is characterized by hypodynamic physiology with tachycardia, delayed capillary refill, diminished pulses, cold and mottled extremities (“cold” shock).[10] [55] [57] Because vasoconstriction intensifies, hypotension comes as a late-onset sign in childhood septic shock;[61] however, in lack of adequate intervention, further increase in SVR becomes counterproductive as the disease progresses since, combined with increased afterload, it may lead to further reduction in CO, which, in turn, may cause septic shock decompensation, cardiac failure, and death.[55] Ceneviva et al classified 50 children with fluid- and dopamine-resistant septic shock on hemodynamic data acquired through a pulmonary artery catheter. Most of the examined patients (58%) was hypodynamic (low CO, high SVR), 20% showed a hyperdynamic response similar to that of adults (high CO, low SVR), whereas 22% was detected with a combination of low CO and low SVR. Four children showed a complete change in hemodynamic status.[61] In summary, contrary to adults, children produce a hypodynamic response to septic shock more often; in addition, their hemodynamic response as a whole is often heterogeneous and changes frequently with progress of the disease.


#

Management of Sepsis in Children

Apart from antimicrobial therapy, management of sepsis is almost exclusively limited to supportive therapeutic interventions. The goal-directed approach to septic shock treatment emphasizes rapid restoration of vascular functions, oxygen delivery, and a therapeutic strategy aimed at early start antibiotics. Initial resuscitation endpoints should be reached as soon as possible, preferably within an hour ([Table 5]).[16] [21] [57] [63] [64] [65] [66] [67] In therapy-resistant shock, constant advanced hemodynamic monitoring is essential in an intensive care setting. Numerous alternative approaches exist (SVR, SVR index, SV variation, global end-diastolic volume), but to date, only measurement of CO and venous oximetry have been adequately validated and incorporated in clinical guidelines.[68] [69] [70] [71] [72] [73] [74] [75] [76] Therapeutic interventions of hemodynamic support should be directed at maintaining mixed venous oxygen saturation at ≥70%, cardiac index between 3.3 and 6 L/minute/m2, and a normal perfusion pressure for age.[16] [57]

Table 5

Therapeutic goals during management of children with septic shock

Initial therapeutic endpoints:

 • Threshold heart rate associated with increased mortality in critically ill (not necessarily septic) infants are a HR < 90 bpm or > 160 bpm, and in children are a HR < 70 bpm or >150 bpm)

 • Capillary refill time < 2 s

 • Strong distal pulses equal to central pulses

 • Warm extremities

 • Normal blood pressure for age

 • Normal mental status

 • Urine output >1 mL/kg/h once effective circulating volume is restored

 • Lactate < 4 mmol/L or ≥10% decrease per every 1–2 h

Central venous oxygen saturation greater than or equal to 70%

Cardiac index between 3.3 and 6 L/min/m2

Abbreviations: HR, heart rate.


Source: Data from Brierley et al.[57]


Airway Management and Respiratory Support

In septic children, airways and respiration are to be closely observed since lung compliance and work of breathing might change rapidly and acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are common complications.[77] Respiratory alkalosis of central origin is often detected in early stage sepsis. With the progress of the disease, metabolic acidosis, itself a consequence of progressive peripheral circulatory failure, may further deteriorate owing to respiratory acidosis caused by a consequential disturbance of consciousness and possible pulmonary complications.[57] Due to their generally lower functional residual capacity (FRC) and limited compensation reserves, septic children, especially young infants, may require endotracheal intubation even in early stage disease.[16] Respiratory muscles can demand up to 40% of the total CO; this can be decreased for the benefit of vital organs through adequate oxygen therapy, respiratory support and ventilation, or administration of muscle relaxants;[57] however, increased intrathoracic pressure caused by invasive ventilation might lead to further deterioration by impeding venous return, especially if fluid status of the patient has not been stabilized prior to the intervention.[16]

Upon detection of respiratory distress in pediatric sepsis, and for all children with septic shock, 100% oxygen must be promptly given to improve peripheral oxygen delivery, with oxygenation constantly monitored by pulse oximetry.[78] In children receiving any oxygen therapy, once peripheral circulation has been restored and pulse oximetry is reliable, hyperoxia should be avoided to prevent potential harm caused by the production of free radicals.[79] In children, especially infants, whose normal oxygenation cannot be maintained by oxygen supply, nasal continuous positive airway pressure can be applied to increase FRC and reduce work of breathing[80] or, as appropriate, bag-valve-mask ventilation should be used while securing venous access before endotracheal intubation and also during volume resuscitation and peripheral inotropic treatment.[16] [81]

Judgment on the indication for endotracheal intubation should primarily consider age-specific respiratory rate, work of breathing, and level of consciousness; situations that may require intubation include ARDS, consciousness disorder, increased intracranial pressure, convulsion persisting after repeated benzodiazepine administration, central line insertion procedures, invasive hemodynamic monitoring, and metabolic acidosis expected to respond to slight mechanical hyperventilation.[16] [57] [81]

Mechanical ventilation can be life-saving, but low lung compliance and/or high peak pressures might lead to ventilator-induced lung injury through alveolar overstretching (volutrauma), repeated alveolar collapse–expansion cycles (atelectrauma), and oxygen toxicity.[82] During ventilation, lung protective strategies established in adult practice should be followed.[16] In sepsis accompanied by ALI/ARDS, FiO2 and positive end expiratory pressure should be titrated for SpO2 to be above 90%,[83] while still avoiding hyperoxia for reasons outlined previously. Similar to adult recommendations, pH should be maintained within the range of 7.3 to 7.45; in certain cases, permissive hypercapnia might by advantageous unless contraindicated.[84] [85] Optimal peak pressure is below 30 cm H2O; the recommended tidal volume is 4 to 6 mL/kg, with values above 10 mL/kg avoided.[16] [83] [84] [86] In case conventional ventilation fails, high-frequency oscillatory ventilation can be applied as rescue treatment.[87] [88] [89]


#

Sedoanalgesia

In the treatment of septic children, sedoanalgesia might be necessary during endotracheal intubation, mechanical ventilation, invasive interventions (e.g., securing a central line), and hemodynamic monitoring. Aggressive fluid and vasoactive therapy for early stabilization significantly decreases the risk of hemodynamic collapse during intubation.[90] Introduction and maintenance of sedoanalgesia requires attention to vasodilatory, direct myocardial depressive, and endogenous catecholamine suppressor effects of candidate substances (propofol, thiopental, benzodiazepines, opioids) because these might cause a deterioration of the patient's cardiovascular status.[57] Etomidate inhibits 11-β-hydroxylase, an enzyme involved in adrenal steroid hormone synthesis, in a concentration-dependent and reversible manner.[91] [92] A single dose alone can cause adrenal suppression lasting beyond 24 hours;[93] [94] increased mortality in childhood meningococcal septicemia has been reported in relation to etomidate use,[94] which is therefore not recommended in the management of sepsis and septic shock.[16] [90] [95] [96] Experimental data suggest a benefit of using ketamine in sepsis because it decreases interleukin (IL)-6 and tumor necrosis factor-α synthesis[97] [98] and increases endogenous norepinephrine release, helping to maintain cardiovascular stability as part of a treatment strategy to increase heart rate and SVR.[99] Based on these characteristics, the use of ketamine for induction is recommended unless contraindications exist.[57] Continuous infusion of ketamine with benzodiazepine not only ensures proper sedation and analgesia but may also carry definite benefits in hemodynamically unstable patients.[57] [99] To facilitate emergency endotracheal intubation and to reduce failed laryngoscopy and complication rates, properly trained and experienced practitioners may consider using short-acting muscle relaxants in addition to sedatives;[2] [78] [100] however, continuous use should be avoided, except in ARDS cases.[16]


#

Fluid Therapy

Assessment and management of airways and respiration is followed by evaluation and optimization of the circulation. For the treatment of critically ill children, establishing vascular access as early as possible is essential;[78] [101] [102] [103] however, it often poses a challenge. If executable quickly, peripheral venous access is acceptable, but the largest and best accessible vein should be selected.[104] The intraosseous route is a quick and secure alternative also in children and should be first choice in patients with shock.[105] [106] [107] [108] [109] All intravenous drugs can be used through the intraosseous route including infusions, vasoactive and inotropic drugs, and blood products.

If justified and not contraindicated, volume resuscitation must be initiated following placement of vascular access. Numerous trials have been performed with fluid therapy. In children with dengue shock syndrome, a survival rate of almost 100% was observed following fluid resuscitation, irrespective of composition of the fluid administered.[110] [111] [112] During the Saline versus Albumin Fluid Evaluation (SAFE) study conducted for fluid therapy of adults in the intensive care unit, no difference was found between the use of 4% albumin and normal saline in 28-day mortality, requirements of mechanical ventilation and renal replacement therapy, time at the intensive care unit and in hospital.[113] Given the lack of data in favor of colloid use, crystalloids should be used first line owing to their availability and better price.[110] [111] [112] [113] [114] [115] [116]

While previously, the primary question in fluid therapy was the choice between colloid and crystalloid solutions, today the focus has shifted to finding the ideal composition for crystalloids.[117] Sodium overload, hyperchloremic acidosis, and consequential renal perfusion disorder, each observed during normal saline use, all potentially worsen prognosis in critically ill patients, especially in those with associated acute kidney injury.[117] [118] The use of so-called balanced crystalloids seems to be more advantageous,[117] [118] [119] [120] [121] a common feature of which is a cation composition more closely matching that of plasma, and a lower chloride concentration compared with normal saline. Their in vitro isotonicity is ensured by other anions such as lactate, gluconate, and acetate.[117] [120] [121] Owing to their potassium content and lower osmolarity, their use may be harmful in hyperkalemia, anuria, and elevated intracranial pressure.[117] [122] [123] To date, no clinical trial has confirmed superiority of the childhood use of balanced solutions. As a summary, with possible complications of sodium or chloride overload and contraindications of balanced solutions in mind, the ideal crystalloid choice must be based on an awareness of the patient's risk factors and consideration of expected responses during the use of each solution type.

Current recommendations suggest initiation of fluid resuscitation with the administration of crystalloids (or colloid equivalent) at 20 mL/kg by push or a pressure bag.[16] [57] [124] Titrated to achieve therapeutic goals ([Table 5]), 40 to 60 mL/kg and sometimes even 200 mL/kg dose might be necessary in the initial phase.[16] [21] [57] [125] [126] [127] Children with persistently elevated heart rate unresponsive to fluid resuscitation should be evaluated for cardiac dysfunction. During acute stabilization and the use of large-volume fluid, no increase was detected in the incidence of ARDS and cerebral edema;[57] [124] [128] however, special attention must be devoted to signs of fluid overload during fluid resuscitation (increased work of breathing, new-onset rales, gallop rhythm, hepatomegaly), for which termination of fluid therapy and administration of inotropic agents and diuretics are recommended.[16] [57] [129]

After shock resuscitation, in cases of fluid overload exceeding 10% when native urine output fails to restore fluid balance even with the use of diuretics, peritoneal dialysis or continuous renal replacement therapy must be performed.[57] [130] [131] [132] Fluid loss and hypovolemia due to diffuse capillary leak may persist for days; maintenance fluid requirements are largely variable, and ongoing fluid replacement must be performed under monitoring of perfusion, central venous pressure, CO, and echocardiographic control.[57]


#

Cardiovascular Drug Therapy

Several factors need to be considered during selection and dosing of cardiovascular drugs; their delayed use significantly increases mortality.[133] [134] Each drug may facilitate restoration of adequate organ perfusion acting on various targets, that is, by increasing contractility (inotropy) and heart rate (chronotropy), or modifying SVR (vasopressor or vasodilator effect). Pharmacokinetics and pharmacodynamics of individual drugs are influenced not only by patient age but also by organ failure (liver and kidney) developing during sepsis.[57] Again, it is important to note that sepsis and septic shock are dynamically changing pathologies, often necessitating modification in the dose, or replacement of medications used, or adding agents with other targets, due to changes in hemodynamic status of patients.[61]

Dopamine titrated to 5 to 10 µg/kg/minute is the first-choice inotropic agent in fluid-refractory shock. At doses above 10 µg/kg/minute, its α-adrenergic effect predominates, making it beneficial for use in low SVR setting.[57] [135] Low-dose usage was previously recommended to improve renal and splanchnic blood flow and renal function; however, more recent studies and meta-analyses failed to reinforce this benefit.[136] [137] [138] More recent data reported about increased mortality related to dopamine administration,[139] [140] [141] presuming a complex underlying mechanism. Reduced gastrointestinal motility, mucosal pH, and oxygen consumption have been reported about its use,[142] [143] in some cases potentially intensifying ventilation–perfusion mismatch,[144] causing multiple anomalies in the thyroid axis[145] [146] or resulting in immune dysfunction by reducing prolactin hormone secretion and attenuating the chemotactic effect of IL-8 on neutrophil granulocytes.[145] [147] [148]

At doses of 3 to 20 µg/kg/minute, dobutamine increases myocardial contractility and heart rate while decreasing SVR; therefore, it can be used instead of or with mid-dose dopamine in low CO associated with high or normal SVR.[57] [149] [150] [151] [152] [153]

Low CO shock refractory to dopamine and/or dobutamine may be reversed with epinephrine initiated at doses of 0.02 µg/kg/minute and titrated up to 1 µg/kg/minute,[61] [154] [155] [156] [157] with some clinicians using it as first-line inotrope in hypodynamic shock due to the unfavorable side-effects of dopamine.[57] At low (<0.3 µg/kg/min) or high (>3 µg/kg/min) doses, it dominantly has β and α adrenergic effect, respectively. Ideally, epinephrine should be given through central venous access, but in emergency cases, intraosseous and peripheral venous administration is also feasible.[57] For a therapeutic response, it should be titrated to achieve desired effect due to its wide interpatient variability.[158] [159] Epinephrine intensifies gluconeogenesis and glycogenolysis, with its hyperglycemic effect likely resulting in adverse outcomes. By stimulating gluconeogenesis, epinephrine enhances lactate shuttle to the liver, and plasma lactate concentration rises and becomes independent of organ perfusion, possibly impeding interpretation of the measured value.[57]

In the less prevalent dopamine-resistant high-CO and low-SVR setting, norepinephrine should be titrated from an initial dose of 0.02 µg/kg/minute to reach hemodynamic support endpoints.[57] [160] In vasodilatory shock treatment, vasopressin and terlipressin, both acting independent from catecholamine receptor stimulation, might be an alternative.[161] [162] [163] [164] [165] [166] During vasopressor use, close hemodynamic monitoring is recommended; increased SVR might lead to deterioration of myocardial contractility and consequential reduction of CO, potentially necessitating the use of inotropic agents.[57] [157]

In normotensive patients refractory to inotropic treatment and having low CO and high SVR, nitrovasodilators can be used to reduce ventricular afterload.[57] Nitroprusside can be titrated for the desired effect (0.5–10 µg/kg/minute).[157] Type III phosphodiesterase inhibitor (milrinone, inamrinone) use may be an alternative to increase myocardial contractility and reduce SVR.[167] [168] [169] Their main advantage is a sustained effect even under β-adrenergic receptor downregulation and desensitization.[57] Having a long half-life, they reach steady-state serum concentrations in up to several hours, driving some clinicians to give a bolus loading dose. Arrhythmia and hypotension owing to their use requires withdrawal, with the latter potentially necessitating crystalloid or colloid fluid bolus or vasopressor treatment.[57] [78] In intractable myocardial dysfunction, levosimendan may be considered as a last resort.[170] [171] [172]


#

Corticosteroids

In sepsis, hypothalamic–pituitary–adrenal axis affects inflammation through leukocytes, cytokines, and nitric-oxide production;[173] however, inflammatory cytokines might cause simultaneous insufficient adrenal output by suppression of the cortisol response to adrenocorticotropin and may result in peripheral glucocorticoid resistance by reducing the number and affinity of receptors.[174] [175] [176] In sepsis, corticosteroid treatment suppresses cytokine production and increases sensitivity of the cardiovascular system to endogenous and exogenous catecholamines, thereby improving myocardial contractility, SV, SVR, and effective circulating blood volume.[177] Although hydrocortisone treatment might be life-saving,[178] [179] [180] yet data on the exact effect on mortality are still controversial.[57] [181] [182] [183] As per current recommendations, steroid treatment should be reserved for children with catecholamine-resistant septic shock and suspected (e.g., severe septic shock with purpura, prior steroid use due to chronic illness, pituitary or adrenal abnormalities) or proven (defined as random total cortisol level less than 18 µg/dL) adrenal insufficiency.[16] [57]


#

Elimination of Pathogens

Early antibiotic therapy is crucial in the treatment of septic patients, possibly initiated within an hour.[16] In adults with septic shock, each hour delay from detection increased mortality by 7.6%,[184] whereas a retrospective study reported significantly higher mortality rates in children receiving antibiotic therapy initiated beyond 3 hours.[185]

Empirical antibiotic therapy should cover sensitivity of the most probable pathogens; well-selected therapy significantly reduces the risk of mortality.[186] Selection of the active ingredient is complex. Patient age, history (underlying illnesses, immune status, drug hypersensitivity), where primary infection was acquired (community-acquired or nosocomial), results of previous cultures (sensitivity of pathogens inducing colonization or confirmed infection), prior antibiotic treatment (last 3 months), and local antibiotic resistance should be considered during decision-making. Additionally, it is important for antibiotics to penetrate in adequate amounts into the target organ or tissue.[16] In sepsis treatment, bactericidal agents should be preferred over bacteriostatic ones, given that former drugs kill pathogens independent of the patient's immune system.[23] There are no specific guidelines on empirical antibiotic therapy; the most frequent combinations are listed in [Table 6]. If possible, consultation with a pediatric infectologist is recommended.[16]

Table 6

Suggested initial empiric antimicrobial regimens

Situation

Initial empiric antimicrobial regimen

Alternative regimen

Children without comorbidities (>2–3 mo)

Piperacillin (or ampicillin) + β-lactamase inhibitor

Ampicillin + cefotaxime

Cefotaxime or ceftazidime ± aminoglycoside

Carbapenem

Nosocomial infection

Piperacillin + β-lactamase inhibitor

Ceftazidime ± aminoglycoside ± glycopeptide (in case of possible MRSA or MRSE infection)

Carbapenem

Immunosuppression, neutropenia

Piperacillin + β-lactamase inhibitor

Ceftazidime + aminoglycoside ± glycopeptide (in case of possible MRSA or MRSE infection)

Ceftazidime

Meropenem

Suspected urosepsis or pyelonephritis

Ampicillin + aminoglycoside

Cefotaxime or ceftazidime + ampicillin (in case of possible enterococcal infection) ± aminoglycoside

Possible GI source

Cefotaxime + metronidazole

Piperacillin + β-lactamase inhibitor

Ampicillin + β-lactamase inhibitor

Meropenem

Catheter-related infection

Cefuroxime

Consider adding rifampicin

Ceftazidime or carbapenem + glycopeptide (in case of possible MRSA or MRSE infection)

Possible MRSA infection

Add vancomycin to the empiric antibiotic regimen

Linezolid or teicoplanin

Possible MRGN infection

Meropenem

Imipenem

Severe sepsis or septic shock

Meropenem ± vancomycin ± aminoglycoside

Cefotaxime (in case of confirmed meningococcal meningitis)

Possible fungal infection

Echinocandin (micafungin or caspofungin)

Liposomal amphotericin

Abbreviations: GI, gastrointestinal; MRGN, multiresistant gram-negative bacteria; MRSA, methicillin-resistant Staphylococcus aureus; MRSE, methicillin-resistant Staphylococcus epidermidis.


Source: Adapted from Sepsis (außer neonatale Early-onset-Sepsis). In: Berner R, Forster J, Bialek R, Borte M, DGPI - Deutsche Gesellschaft für Pädiatrische Infektiologie. DGPI Handbuch. Infektionen bei Kindern und Jugendlichen. Stuttgart: Thieme; 2013:776–784.


Difficulties during or contraindications to culture collection (e.g., hemodynamic instability and/or coagulopathy during lumbar puncture) must not delay treatment initiation; nevertheless, successful sampling is indispensable for de-escalation of subsequent antibiotic therapy. During initial fluid resuscitation and combined antibiotic therapy, drugs given as bolus must be prioritized over infused active ingredients regarding administration sequence; concurrent use of antibiotics and fluid therapy can be facilitated through a second vascular access.[16]

De-escalation of empirical treatment should be done within 3 to 5 days after obtaining culture results; for subsequent infection management, the most effective monotherapy must be selected (except aminoglycoside monotherapy). Antibiotic therapy should be maintained for 7 to 10 days, but in cases of slow clinical response, immunodeficiency, source control problems, or Staphylococcus aureus sepsis, longer treatment periods might be necessary. During antibiotic therapy of septic patients, hepatic and renal dysfunction, fluid compartments, and redistribution altered by fluid resuscitation may necessitate dose modification. Also, improving efficacy and minimizing toxicity risk can be promoted by measuring serum concentrations.[16] [23]

Source control is an important component of sepsis treatment,[16] including debridement of various infected, necrotic tissues, drainage of infected fluids, rapid removal of objects, and foreign bodies identified as the infection source, and, through restoration of anatomic integrity and function, it aims to eliminate further contamination.[187] During the intervention, along with benefit from reducing the infectious inoculum, potential hazards and complications of the intervention and associated transport must be considered.[188] [189]


#

Immunoglobulins

Adjuvant treatment of sepsis with intravenous immunoglobulin (IVIG) seemed highly promising earlier; however, more recent results are controversial.[190] [191] [192] [193] [194] [195] [196] [197] [198] Most of the available studies have small sample size and some have methodological flaws. A large randomized, controlled trial (RCT) for the treatment of sepsis in adults and a larger one in newborns found no benefit for IVIG.[199] [200] Due to its potential toxin-neutralizing effect and an influence on facilitating opsonization of Streptococci, the use of polyclonal immunoglobulin was recommended previously in invasive group A Streptococcal infections.[198] [201] [202] Based on our current knowledge, the benefit of IVIG is doubtful in this case and its use may be considered in refractory toxic shock syndrome.[16]


#

Red Blood Cell Transfusion and Treatment of Disseminated Intravascular Coagulation

Restoration of massive cytokine release, myocardial depression, capillary leak, and abnormal tissue oxygen delivery due to acidosis, each associated with sepsis, by normalizing CO and hemoglobin levels is a cornerstone of therapy.[16] [57] [203] No optimal hemoglobin concentration is known for critically ill children.[16] [203] [204] For adults, efficacy analyses of goal-directed therapy showed better outcomes if central venous oxygen saturation could be maintained above 70% in the first 6 hours following presentation (protocol included transfusion to attain hematocrit levels of ≥30).[205] Earlier recommendations for intensive therapy suggested higher hemoglobin goals for septic children requiring intensive care.[206] [207] In hemodynamically stable, critically ill children, restrictive (hemoglobin threshold of 7 g/dL for red blood cell transfusion) transfusion strategy was as safe as liberal strategy (hemoglobin threshold of 9.5 g/dL for red blood cell transfusion).[204] [208] An RCT on early goal-directed therapy of childhood septic shock confirmed better survival if 10 g/dL was selected as transfusion cutoff.[68] Accordingly, hemoglobin goal is 10 g/L in hemodynamically unstable septic children during resuscitation, with this threshold being lower at 7 g/dL in stable patients.[16]

Disseminated intravascular coagulation is commonly observed in severe sepsis and septic shock.[209] Therapeutic options are controversial and lack validation.[210] Routine use of replacement therapy is not recommended, neither in case of mild clinical signs of bleeding,[211] but to stop clinically significant bleeding, the use of platelet, fresh frozen plasma, and cryoprecipitate infusions might be necessary.[212] [213]

Based on the few data available, protein C concentrate might be effective in severe sepsis associated with purpura fulminans. Improved microcirculation, moreover, lower amputation and skin graft rates were reported about its use with no adverse reactions.[214] [215] [216]


#

Glucose Control

In septic children, disorders of glucose homeostasis are frequent. Due to peripheral insulin resistance and enhanced gluconeogenesis, there is high risk of hyperglycemia, which might be further enhanced by excessive sugar intake through infusions and total parenteral nutrition.[217] Neutrophil and macrophage function, as well as wound healing are deteriorated by hyperglycemia, while increasing infection and thrombosis risk.[218] [219] Higher mortality rates and longer hospitalization were reported for hyperglycemia in critically ill children,[220] [221] [222] with few and controversial available data on the treatment of hyperglycemia developed during sepsis.[203] [223] [224] [225] Until results of more recent trials become available, insulin therapy should be initiated only for blood glucose levels above 180 mg/dL, along with close blood glucose monitoring to avoid hypoglycemia, in accordance with recommendations for treatment of adults.[16] For note, insulin demand usually declines rapidly 18 hours from shock initiation.[57] Hypoglycemia can have devastating neurologic consequences and should be diagnosed early and treated immediately. Once initial hypoglycemia is resolved, children should receive 10% dextrose in normal saline ensuring a glucose intake of 2 to 8 mg/kg/minute depending on age to maintain euglycemia.[16] [57]


#

Refractory Shock

In children with refractory shock, potential associated conditions negatively influencing outcome must be revealed. Pneumothorax, pericardial effusion, and intra-abdominal hypertension may equally be mechanic causes of shock persistence, and their treatment must not be delayed after diagnosis. In adrenal insufficiency or hypothyroidism, adequate hormone substitution should be performed. In refractory shock, expansion of empirical antibiotic coverage, drainage, and debridement of the necrotic areas might be necessary. Immunosuppressive treatment should be withheld; in neutropenia, granulocyte colony-stimulating factor should be used where appropriately indicated. Excessive bleeding most frequently developed due to disseminated intravascular coagulation might indicate use of RBC and blood products.[16] [57] If hemodynamic support endpoints cannot be reached by treating these reversible causes and in associated refractory respiratory failure, extracorporeal membrane oxygenation may be used as a last resort.[226] [227] [228] [229] [230]


#
#

Conclusion

Therapeutic algorithm of childhood sepsis and septic shock is summarized in [Fig. 1]. Success of sepsis care is improved by properly organized emergency care and compliance with internationally accepted treatment guidelines. It is important to note that most pediatric sepsis cases can be prevented; therefore, effective prevention based on screening programs, vaccination, and infection control provisions is an exceedingly important strategic move.

Zoom Image
Fig. 1Stepwise management of hemodynamic support in infants and children with septic shock.

#
#

No conflict of interest has been declared by the author(s).

  • References

  • 1 Goldstein B, Giroir B, Randolph A. ; International Consensus Conference on Pediatric Sepsis. International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005; 6 (01) 2-8
  • 2 Cinel I, Dellinger RP. Advances in pathogenesis and management of sepsis. Curr Opin Infect Dis 2007; 20 (04) 345-352
  • 3 Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S, Remick DG. The pathogenesis of sepsis. Annu Rev Pathol 2011; 6: 19-48
  • 4 Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med 2003; 348 (02) 138-150
  • 5 Lederer JA, Rodrick ML, Mannick JA. The effects of injury on the adaptive immune response. Shock 1999; 11 (03) 153-159
  • 6 Payen D, Faivre V, Lukaszewicz AC, Villa F, Goldberg P. Expression of monocyte human leukocyte antigen-DR in relation with sepsis severity and plasma mediators. Minerva Anestesiol 2009; 75 (09) 484-493
  • 7 Wynn J, Cornell TT, Wong HR, Shanley TP, Wheeler DS. The host response to sepsis and developmental impact. Pediatrics 2010; 125 (05) 1031-1041
  • 8 Wheeler DS, Zingarelli B, Wheeler WJ, Wong HR. Novel pharmacologic approaches to the management of sepsis: targeting the host inflammatory response. Recent Pat Inflamm Allergy Drug Discov 2009; 3 (02) 96-112
  • 9 Hazelzet J, Driessen GJA, Abboud P, Wheeler DS, Shanley TP, Wong HR. Sepsis. In: Wheeler DS, Wong HR, Shanley TP. , eds. Pediatric Critical Care Medicine: Basic Science and Clinical Evidence. London: Springer-Verlag; 2007: 1421-1444
  • 10 Wheeler DS, Wong HR, Zingarelli B. Pediatric sepsis - part I: “children are not small adults!”. Open Inflamm J 2011; 4: 4-15
  • 11 Weiss SL, Fitzgerald JC, Pappachan J. , et al; Sepsis Prevalence, Outcomes, and Therapies (SPROUT) Study Investigators and Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network. Global epidemiology of pediatric severe sepsis: the sepsis prevalence, outcomes, and therapies study. Am J Respir Crit Care Med 2015; 191 (10) 1147-1157
  • 12 Carcillo JA. Reducing the global burden of sepsis in infants and children: a clinical practice research agenda. Pediatr Crit Care Med 2005; 6 (3, Suppl): S157-S164
  • 13 Odetola FO, Gebremariam A, Freed GL. Patient and hospital correlates of clinical outcomes and resource utilization in severe pediatric sepsis. Pediatrics 2007; 119 (03) 487-494
  • 14 Typpo KV, Petersen NJ, Hallman DM, Markovitz BP, Mariscalco MM. Day 1 multiple organ dysfunction syndrome is associated with poor functional outcome and mortality in the pediatric intensive care unit. Pediatr Crit Care Med 2009; 10 (05) 562-570
  • 15 Kissoon N, Carcillo JA, Espinosa V. , et al; Global Sepsis Initiative Vanguard Center Contributors. World Federation of Pediatric Intensive Care and Critical Care Societies: Global Sepsis Initiative. Pediatr Crit Care Med 2011; 12 (05) 494-503
  • 16 Dellinger RP, Levy MM, Rhodes A. , et al; Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41 (02) 580-637
  • 17 Hartman ME, Linde-Zwirble WT, Angus DC, Watson RS. Trends in the epidemiology of pediatric severe sepsis*. Pediatr Crit Care Med 2013; 14 (07) 686-693
  • 18 Ruth A, McCracken CE, Fortenberry JD, Hall M, Simon HK, Hebbar KB. Pediatric severe sepsis: current trends and outcomes from the Pediatric Health Information Systems database. Pediatr Crit Care Med 2014; 15 (09) 828-838
  • 19 Butt W. Septic shock. Pediatr Clin North Am 2001; 48 (03) 601-625 , viii
  • 20 Gaines NN, Patel B, Williams EA, Cruz AT. Etiologies of septic shock in a pediatric emergency department population. Pediatr Infect Dis J 2012; 31 (11) 1203-1205
  • 21 Han YY, Carcillo JA, Dragotta MA. , et al. Early reversal of pediatric-neonatal septic shock by community physicians is associated with improved outcome. Pediatrics 2003; 112 (04) 793-799
  • 22 Kutko MC, Calarco MP, Flaherty MB. , et al. Mortality rates in pediatric septic shock with and without multiple organ system failure. Pediatr Crit Care Med 2003; 4 (03) 333-337
  • 23 Simmons ML, Durham SH, Carter CW. Pharmacological management of pediatric patients with sepsis. AACN Adv Crit Care 2012; 23 (04) 437-448 , quiz 449–450
  • 24 Bone RC, Balk RA, Cerra FB. , et al; The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest 1992; 101 (06) 1644-1655
  • 25 Levy MM, Fink MP, Marshall JC. , et al; International Sepsis Definitions Conference. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Intensive Care Med 2003; 29 (04) 530-538
  • 26 Marshall JC. SIRS and MODS: what is their relevance to the science and practice of intensive care?. Shock 2000; 14 (06) 586-589
  • 27 Weiss SL, Parker B, Bullock ME. , et al. Defining pediatric sepsis by different criteria: discrepancies in populations and implications for clinical practice. Pediatr Crit Care Med 2012; 13 (04) e219-e226
  • 28 Argent AC. Recognizing pediatric sepsis: do the concepts help us to focus appropriately?. Pediatr Crit Care Med 2016; 17 (05) 460-461
  • 29 Vincent JL, Opal SM, Marshall JC, Tracey KJ. Sepsis definitions: time for change. Lancet 2013; 381 (9868): 774-775
  • 30 Singer M, Deutschman CS, Seymour CW. , et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016; 315 (08) 801-810
  • 31 Agyeman P, Aebi C, Hirt A. , et al. Predicting bacteremia in children with cancer and fever in chemotherapy-induced neutropenia: results of the prospective multicenter SPOG 2003 FN study. Pediatr Infect Dis J 2011; 30 (07) e114-e119
  • 32 Struthers S, Underhill H, Albersheim S, Greenberg D, Dobson S. A comparison of two versus one blood culture in the diagnosis and treatment of coagulase-negative staphylococcus in the neonatal intensive care unit. J Perinatol 2002; 22 (07) 547-549
  • 33 Isaacman DJ, Karasic RB, Reynolds EA, Kost SI. Effect of number of blood cultures and volume of blood on detection of bacteremia in children. J Pediatr 1996; 128 (02) 190-195
  • 34 Kaditis AG, O'Marcaigh AS, Rhodes KH, Weaver AL, Henry NK. Yield of positive blood cultures in pediatric oncology patients by a new method of blood culture collection. Pediatr Infect Dis J 1996; 15 (07) 615-620
  • 35 Schelonka RL, Chai MK, Yoder BA, Hensley D, Brockett RM, Ascher DP. Volume of blood required to detect common neonatal pathogens. J Pediatr 1996; 129 (02) 275-278
  • 36 Buttery JP. Blood cultures in newborns and children: optimising an everyday test. Arch Dis Child Fetal Neonatal Ed 2002; 87 (01) F25-F28
  • 37 Surdulescu S, Utamsingh D, Shekar R. Phlebotomy teams reduce blood-culture contamination rate and save money. Clin Perform Qual Health Care 1998; 6 (02) 60-62
  • 38 Weinbaum FI, Lavie S, Danek M, Sixsmith D, Heinrich GF, Mills SS. Doing it right the first time: quality improvement and the contaminant blood culture. J Clin Microbiol 1997; 35 (03) 563-565
  • 39 Weinstein MP. Blood culture contamination: persisting problems and partial progress. J Clin Microbiol 2003; 41 (06) 2275-2278
  • 40 Calfee DP, Farr BM. Comparison of four antiseptic preparations for skin in the prevention of contamination of percutaneously drawn blood cultures: a randomized trial. J Clin Microbiol 2002; 40 (05) 1660-1665
  • 41 Hall KK, Lyman JA. Updated review of blood culture contamination. Clin Microbiol Rev 2006; 19 (04) 788-802
  • 42 Norberg A, Christopher NC, Ramundo ML, Bower JR, Berman SA. Contamination rates of blood cultures obtained by dedicated phlebotomy vs intravenous catheter. JAMA 2003; 289 (06) 726-729
  • 43 Hardy DJ, Hulbert BB, Migneault PC. Time to detection of positive BacT/Alert blood cultures and lack of need for routine subculture of 5- to 7-day negative cultures. J Clin Microbiol 1992; 30: 2743-2745
  • 44 Huang AH, Yan JJ, Wu JJ. Comparison of five days versus seven days of incubation for detection of positive blood cultures by the Bactec 9240 system. Eur J Clin Microbiol Infect Dis 1998; 17 (09) 637-641
  • 45 Connell TG, Rele M, Cowley D, Buttery JP, Curtis N. How reliable is a negative blood culture result? Volume of blood submitted for culture in routine practice in a children's hospital. Pediatrics 2007; 119 (05) 891-896
  • 46 Dark P, Dunn G, Chadwick P. , et al. The clinical diagnostic accuracy of rapid detection of healthcare-associated bloodstream infection in intensive care using multipathogen real-time PCR technology. BMJ Open 2011; 1 (01) e000181
  • 47 Tsalik EL, Jones D, Nicholson B. , et al. Multiplex PCR to diagnose bloodstream infections in patients admitted from the emergency department with sepsis. J Clin Microbiol 2010; 48 (01) 26-33
  • 48 Lucignano B, Ranno S, Liesenfeld O. , et al. Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in newborns and children with suspected sepsis. J Clin Microbiol 2011; 49 (06) 2252-2258
  • 49 Dark PM, Dean P, Warhurst G. Bench-to-bedside review: the promise of rapid infection diagnosis during sepsis using polymerase chain reaction-based pathogen detection. Crit Care 2009; 13 (04) 217
  • 50 Lehmann LE, Hunfeld KP, Emrich T. , et al. A multiplex real-time PCR assay for rapid detection and differentiation of 25 bacterial and fungal pathogens from whole blood samples. Med Microbiol Immunol (Berl) 2008; 197 (03) 313-324
  • 51 von Lilienfeld-Toal M, Lehmann LE, Raadts AD. , et al. Utility of a commercially available multiplex real-time PCR assay to detect bacterial and fungal pathogens in febrile neutropenia. J Clin Microbiol 2009; 47 (08) 2405-2410
  • 52 Wellinghausen N, Kochem AJ, Disqué C. , et al. Diagnosis of bacteremia in whole-blood samples by use of a commercial universal 16S rRNA gene-based PCR and sequence analysis. J Clin Microbiol 2009; 47 (09) 2759-2765
  • 53 Westh H, Lisby G, Breysse F. , et al. Multiplex real-time PCR and blood culture for identification of bloodstream pathogens in patients with suspected sepsis. Clin Microbiol Infect 2009; 15 (06) 544-551
  • 54 Watson RS, Carcillo JA. Scope and epidemiology of pediatric sepsis. Pediatr Crit Care Med 2005; 6 (3, Suppl): S3-S5
  • 55 Aneja R, Carcillo J. Differences between adult and pediatric septic shock. Minerva Anestesiol 2011; 77 (10) 986-992
  • 56 Parker MM, Shelhamer JH, Natanson C, Alling DW, Parrillo JE. Serial cardiovascular variables in survivors and nonsurvivors of human septic shock: heart rate as an early predictor of prognosis. Crit Care Med 1987; 15 (10) 923-929
  • 57 Brierley J, Carcillo JA, Choong K. , et al. Clinical practice parameters for hemodynamic support of pediatric and neonatal septic shock: 2007 update from the American College of Critical Care Medicine. Crit Care Med 2009; 37 (02) 666-688
  • 58 Practice parameters for hemodynamic support of sepsis in adults with sepsis. Task force of the American College of Critical Care Medicine, Society of Critical Care Medicine. Crit Care Med 1999; 27: 639-660
  • 59 Parker MM, Shelhamer JH, Bacharach SL. , et al. Profound but reversible myocardial depression in patients with septic shock. Ann Intern Med 1984; 100 (04) 483-490
  • 60 Pollack MM, Fields AI, Ruttimann UE. Sequential cardiopulmonary variables of infants and children in septic shock. Crit Care Med 1984; 12 (07) 554-559
  • 61 Ceneviva G, Paschall JA, Maffei F, Carcillo JA. Hemodynamic support in fluid-refractory pediatric septic shock. Pediatrics 1998; 102 (02) e19
  • 62 Feltes TF, Pignatelli R, Kleinert S, Mariscalco MM. Quantitated left ventricular systolic mechanics in children with septic shock utilizing noninvasive wall-stress analysis. Crit Care Med 1994; 22 (10) 1647-1658
  • 63 Pollack MM, Ruttimann UE, Getson PR. Pediatric risk of mortality (PRISM) score. Crit Care Med 1988; 16 (11) 1110-1116
  • 64 Dugas MA, Proulx F, de Jaeger A, Lacroix J, Lambert M. Markers of tissue hypoperfusion in pediatric septic shock. Intensive Care Med 2000; 26 (01) 75-83
  • 65 Nguyen HB, Rivers EP, Knoblich BP. , et al. Early lactate clearance is associated with improved outcome in severe sepsis and septic shock. Crit Care Med 2004; 32 (08) 1637-1642
  • 66 Arnold RC, Shapiro NI, Jones AE. , et al; Emergency Medicine Shock Research Network (EMShockNet) Investigators. Multicenter study of early lactate clearance as a determinant of survival in patients with presumed sepsis. Shock 2009; 32 (01) 35-39
  • 67 Scott HF, Brou L, Deakyne SJ, Fairclough DL, Kempe A, Bajaj L. Lactate clearance and normalization and prolonged organ dysfunction in pediatric sepsis. J Pediatr 2016; 170: 149-55.e1 , 4
  • 68 de Oliveira CF, de Oliveira DS, Gottschald AF. , et al. ACCM/PALS haemodynamic support guidelines for paediatric septic shock: an outcomes comparison with and without monitoring central venous oxygen saturation. Intensive Care Med 2008; 34 (06) 1065-1075
  • 69 Schiffmann H, Erdlenbruch B, Singer D. , et al. Assessment of cardiac output, intravascular volume status, and extravascular lung water by transpulmonary indicator dilution in critically ill neonates and infants. J Cardiothorac Vasc Anesth 2002; 16 (05) 592-597
  • 70 Deep A, Goonasekera CD, Wang Y, Brierley J. Evolution of haemodynamics and outcome of fluid-refractory septic shock in children. Intensive Care Med 2013; 39 (09) 1602-1609
  • 71 Brierley J, Peters MJ. Distinct hemodynamic patterns of septic shock at presentation to pediatric intensive care. Pediatrics 2008; 122 (04) 752-759
  • 72 Martin GS, Eaton S, Mealer M, Moss M. Extravascular lung water in patients with severe sepsis: a prospective cohort study. Crit Care 2005; 9 (02) R74-R82
  • 73 Boehne M, Baustert M, Paetzel V. , et al. Determination of cardiac output by ultrasound dilution technique in infants and children: a validation study against direct Fick principle. Br J Anaesth 2014; 112 (03) 469-476
  • 74 Wongsirimetheekul T, Khositseth A, Lertbunrian R. Non-invasive cardiac output assessment in critically ill paediatric patients. Acta Cardiol 2014; 69 (02) 167-173
  • 75 Crittendon III I, Dreyer WJ, Decker JA, Kim JJ. Ultrasound dilution: an accurate means of determining cardiac output in children. Pediatr Crit Care Med 2012; 13 (01) 42-46
  • 76 Fernandez EG, Green TP, Sweeney M. Low inferior vena caval catheters for hemodynamic and pulmonary function monitoring in pediatric critical care patients. Pediatr Crit Care Med 2004; 5 (01) 14-18
  • 77 Dahlem P, van Aalderen WM, Hamaker ME, Dijkgraaf MG, Bos AP. Incidence and short-term outcome of acute lung injury in mechanically ventilated children. Eur Respir J 2003; 22 (06) 980-985
  • 78 Kleinman ME, Chameides L, Schexnayder SM. , et al. Part 14: pediatric advanced life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122 (18) (Suppl. 03) S876-S908
  • 79 Asfar P, Calzia E, Huber-Lang M, Ignatius A, Radermacher P. Hyperoxia during septic shock--Dr. Jekyll or Mr. Hyde?. Shock 2012; 37 (01) 122-123
  • 80 Cam BV, Tuan DT, Fonsmark L. , et al. Randomized comparison of oxygen mask treatment vs. nasal continuous positive airway pressure in dengue shock syndrome with acute respiratory failure. J Trop Pediatr 2002; 48 (06) 335-339
  • 81 Khilnani P, Singhi S, Lodha R. , et al. Pediatric Sepsis Guidelines: summary for resource-limited countries. Indian J Crit Care Med 2010; 14 (01) 41-52
  • 82 International consensus conferences in intensive care medicine: ventilator-associated Lung Injury in ARDS. Am J Respir Crit Care Med 1999; 160 (06) 2118-2124
  • 83 Randolph AG. Management of acute lung injury and acute respiratory distress syndrome in children. Crit Care Med 2009; 37 (08) 2448-2454
  • 84 Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A. ; Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 2000; 342 (18) 1301-1308
  • 85 Rotta AT, Steinhorn DM. Is permissive hypercapnia a beneficial strategy for pediatric acute lung injury?. Respir Care Clin N Am 2006; 12 (03) 371-387
  • 86 Santschi M, Jouvet P, Leclerc F. , et al; PALIVE Investigators; Pediatric Acute Lung Injury and Sepsis Investigators Network (PALISI); European Society of Pediatric and Neonatal Intensive Care (ESPNIC). Acute lung injury in children: therapeutic practice and feasibility of international clinical trials. Pediatr Crit Care Med 2010; 11 (06) 681-689
  • 87 Ben Jaballah N, Khaldi A, Mnif K. , et al. High-frequency oscillatory ventilation in pediatric patients with acute respiratory failure. Pediatr Crit Care Med 2006; 7 (04) 362-367
  • 88 Arnold JH, Hanson JH, Toro-Figuero LO, Gutiérrez J, Berens RJ, Anglin DL. Prospective, randomized comparison of high-frequency oscillatory ventilation and conventional mechanical ventilation in pediatric respiratory failure. Crit Care Med 1994; 22 (10) 1530-1539
  • 89 Randolph AG, Meert KL, O'Neil ME. , et al; Pediatric Acute Lung Injury and Sepsis Investigators Network. The feasibility of conducting clinical trials in infants and children with acute respiratory failure. Am J Respir Crit Care Med 2003; 167 (10) 1334-1340
  • 90 Fengler BT. Should etomidate be used for rapid-sequence intubation induction in critically ill septic patients?. Am J Emerg Med 2008; 26 (02) 229-232
  • 91 Wagner RL, White PF, Kan PB, Rosenthal MH, Feldman D. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med 1984; 310 (22) 1415-1421
  • 92 Dörr HG, Kuhnle U, Holthausen H, Bidlingmaier F, Knorr D. Etomidate: a selective adrenocortical 11 β-hydroxylase inhibitor. Klin Wochenschr 1984; 62 (21) 1011-1013
  • 93 den Brinker M, Joosten KF, Liem O. , et al. Adrenal insufficiency in meningococcal sepsis: bioavailable cortisol levels and impact of interleukin-6 levels and intubation with etomidate on adrenal function and mortality. J Clin Endocrinol Metab 2005; 90 (09) 5110-5117
  • 94 den Brinker M, Hokken-Koelega AC, Hazelzet JA, de Jong FH, Hop WC, Joosten KF. One single dose of etomidate negatively influences adrenocortical performance for at least 24h in children with meningococcal sepsis. Intensive Care Med 2008; 34 (01) 163-168
  • 95 Jackson Jr WL. Should we use etomidate as an induction agent for endotracheal intubation in patients with septic shock?: a critical appraisal. Chest 2005; 127 (03) 1031-1038
  • 96 Bloomfield R, Noble DW. Etomidate and fatal outcome--even a single bolus dose may be detrimental for some patients. Br J Anaesth 2006; 97 (01) 116-117
  • 97 Taniguchi T, Shibata K, Yamamoto K. Ketamine inhibits endotoxin-induced shock in rats. Anesthesiology 2001; 95 (04) 928-932
  • 98 Song XM, Li JG, Wang YL. , et al. Effects of ketamine on proinflammatory cytokines and nuclear factor kappaB in polymicrobial sepsis rats. World J Gastroenterol 2006; 12 (45) 7350-7354
  • 99 Mencía SB, López-Herce JC, Freddi N. Analgesia and sedation in children: practical approach for the most frequent situations. J Pediatr (Rio J) 2007; 83 (2, Suppl): S71-S82
  • 100 Sagarin MJ, Chiang V, Sakles JC. , et al; National Emergency Airway Registry (NEAR) investigators. Rapid sequence intubation for pediatric emergency airway management. Pediatr Emerg Care 2002; 18 (06) 417-423
  • 101 Sun CY, Lee KC, Lin IH. , et al. Near-infrared light device can improve intravenous cannulation in critically ill children. Pediatr Neonatol 2013; 54: 194-197
  • 102 Lillis KA, Jaffe DM. Prehospital intravenous access in children. Ann Emerg Med 1992; 21 (12) 1430-1434
  • 103 Stovroff M, Teague WG. Intravenous access in infants and children. Pediatr Clin North Am 1998; 45 (06) 1373-1393 , viii
  • 104 Idris AH, Melker RJ. High-flow sheaths for pediatric fluid resuscitation: a comparison of flow rates with standard pediatric catheters. Pediatr Emerg Care 1992; 8 (03) 119-122
  • 105 Rosetti VA, Thompson BM, Miller J, Mateer JR, Aprahamian C. Intraosseous infusion: an alternative route of pediatric intravascular access. Ann Emerg Med 1985; 14 (09) 885-888
  • 106 Seigler RS, Tecklenburg FW, Shealy R. Prehospital intraosseous infusion by emergency medical services personnel: a prospective study. Pediatrics 1989; 84 (01) 173-177
  • 107 Glaeser PW, Hellmich TR, Szewczuga D, Losek JD, Smith DS. Five-year experience in prehospital intraosseous infusions in children and adults. Ann Emerg Med 1993; 22 (07) 1119-1124
  • 108 Fiorito BA, Mirza F, Doran TM. , et al. Intraosseous access in the setting of pediatric critical care transport. Pediatr Crit Care Med 2005; 6 (01) 50-53
  • 109 Horton MA, Beamer C. Powered intraosseous insertion provides safe and effective vascular access for pediatric emergency patients. Pediatr Emerg Care 2008; 24 (06) 347-350
  • 110 Ngo NT, Cao XT, Kneen R. , et al. Acute management of dengue shock syndrome: a randomized double-blind comparison of 4 intravenous fluid regimens in the first hour. Clin Infect Dis 2001; 32 (02) 204-213
  • 111 Wills BA, Nguyen MD, Ha TL. , et al. Comparison of three fluid solutions for resuscitation in dengue shock syndrome. N Engl J Med 2005; 353 (09) 877-889
  • 112 Dung NM, Day NP, Tam DT. , et al. Fluid replacement in dengue shock syndrome: a randomized, double-blind comparison of four intravenous-fluid regimens. Clin Infect Dis 1999; 29 (04) 787-794
  • 113 Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R. ; SAFE Study Investigators. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350 (22) 2247-2256
  • 114 Upadhyay M, Singhi S, Murlidharan J, Kaur N, Majumdar S. Randomized evaluation of fluid resuscitation with crystalloid (saline) and colloid (polymer from degraded gelatin in saline) in pediatric septic shock. Indian Pediatr 2005; 42 (03) 223-231
  • 115 Akech S, Ledermann H, Maitland K. Choice of fluids for resuscitation in children with severe infection and shock: systematic review. BMJ 2010; 341: c4416
  • 116 Ford N, Hargreaves S, Shanks L. Mortality after fluid bolus in children with shock due to sepsis or severe infection: a systematic review and meta-analysis. PLoS One 2012; 7 (08) e43953
  • 117 Mendes PV, Zampieri FG, Park M. Is There a Role for Balanced Solutions in Septic Patients?. Shock 2017; 47 (1S, Suppl 1): 30-34
  • 118 Guidet B, Soni N, Della Rocca G. , et al. A balanced view of balanced solutions. Crit Care 2010; 14 (05) 325
  • 119 Vincent JL, Orbegozo Cortés D, Acheampong A. Current haemodynamic management of septic shock. Presse Med 2016; 45 (4 Pt 2): e99-e103
  • 120 Allen SJ. Fluid therapy and outcome: balance is best. J Extra Corpor Technol 2014; 46 (01) 28-32
  • 121 Orbegozo Cortés D, Rayo Bonor A, Vincent JL. Isotonic crystalloid solutions: a structured review of the literature. Br J Anaesth 2014; 112 (06) 968-981
  • 122 Roquilly A, Loutrel O, Cinotti R. , et al. Balanced versus chloride-rich solutions for fluid resuscitation in brain-injured patients: a randomised double-blind pilot study. Crit Care 2013; 17 (02) R77
  • 123 Williams EL, Hildebrand KL, McCormick SA, Bedel MJ. The effect of intravenous lactated Ringer's solution versus 0.9% sodium chloride solution on serum osmolality in human volunteers. Anesth Analg 1999; 88 (05) 999-1003
  • 124 Stoner MJ, Goodman DG, Cohen DM, Fernandez SA, Hall MW. Rapid fluid resuscitation in pediatrics: testing the American College of Critical Care Medicine guideline. Ann Emerg Med 2007; 50 (05) 601-607
  • 125 Carcillo JA, Davis AL, Zaritsky A. Role of early fluid resuscitation in pediatric septic shock. JAMA 1991; 266 (09) 1242-1245
  • 126 Maitland K, Kiguli S, Opoka RO. , et al; FEAST Trial Group. Mortality after fluid bolus in African children with severe infection. N Engl J Med 2011; 364 (26) 2483-2495
  • 127 Santhanam I, Sangareddi S, Venkataraman S, Kissoon N, Thiruvengadamudayan V, Kasthuri RK. A prospective randomized controlled study of two fluid regimens in the initial management of septic shock in the emergency department. Pediatr Emerg Care 2008; 24 (10) 647-655
  • 128 Zadrobilek E, Hackl W, Sporn P, Steinbereithner K. Effect of large volume replacement with balanced electrolyte solutions on extravascular lung water in surgical patients with sepsis syndrome. Intensive Care Med 1989; 15 (08) 505-510
  • 129 Cruz AT, Perry AM, Williams EA, Graf JM, Wuestner ER, Patel B. Implementation of goal-directed therapy for children with suspected sepsis in the emergency department. Pediatrics 2011; 127 (03) e758-e766
  • 130 Goldstein SL, Somers MJ, Baum MA. , et al. Pediatric patients with multi-organ dysfunction syndrome receiving continuous renal replacement therapy. Kidney Int 2005; 67 (02) 653-658
  • 131 Askenazi D. Evaluation and management of critically ill children with acute kidney injury. Curr Opin Pediatr 2011; 23 (02) 201-207
  • 132 Gulla KM, Sachdev A, Gupta D, Gupta N, Anand K, Pruthi PK. Continuous renal replacement therapy in children with severe sepsis and multiorgan dysfunction - a pilot study on timing of initiation. Indian J Crit Care Med 2015; 19 (10) 613-617
  • 133 Ninis N, Phillips C, Bailey L. , et al. The role of healthcare delivery in the outcome of meningococcal disease in children: case-control study of fatal and non-fatal cases. BMJ 2005; 330 (7506): 1475
  • 134 Thompson MJ, Ninis N, Perera R. , et al. Clinical recognition of meningococcal disease in children and adolescents. Lancet 2006; 367 (9508): 397-403
  • 135 Ushay HM, Notterman DA. Pharmacology of pediatric resuscitation. Pediatr Clin North Am 1997; 44 (01) 207-233
  • 136 Bellomo R, Chapman M, Finfer S, Hickling K, Myburgh J. ; Australian and New Zealand Intensive Care Society (ANZICS) Clinical Trials Group. Low-dose dopamine in patients with early renal dysfunction: a placebo-controlled randomised trial. Lancet 2000; 356 (9248): 2139-2143
  • 137 Kellum JA, M Decker J. Use of dopamine in acute renal failure: a meta-analysis. Crit Care Med 2001; 29 (08) 1526-1531
  • 138 Zacharias M, Mugawar M, Herbison GP. , et al. Interventions for protecting renal function in the perioperative period. Cochrane Database Syst Rev 2013; (09) CD003590
  • 139 De Backer D, Aldecoa C, Njimi H, Vincent JL. Dopamine versus norepinephrine in the treatment of septic shock: a meta-analysis*. Crit Care Med 2012; 40 (03) 725-730
  • 140 Avni T, Lador A, Lev S, Leibovici L, Paul M, Grossman A. Vasopressors for the treatment of septic shock: systematic review and meta-analysis. PLoS One 2015; 10 (08) e0129305
  • 141 Ventura AM, Shieh HH, Bousso A. , et al. Double-blind prospective randomized controlled trial of dopamine versus epinephrine as first-line vasoactive drugs in pediatric septic shock. Crit Care Med 2015; 43 (11) 2292-2302
  • 142 Jakob SM, Ruokonen E, Takala J. Effects of dopamine on systemic and regional blood flow and metabolism in septic and cardiac surgery patients. Shock 2002; 18 (01) 8-13
  • 143 Dive A, Foret F, Jamart J, Bulpa P, Installé E. Effect of dopamine on gastrointestinal motility during critical illness. Intensive Care Med 2000; 26 (07) 901-907
  • 144 Shoemaker WC, Appel PL, Kram HB, Duarte D, Harrier HD, Ocampo HA. Comparison of hemodynamic and oxygen transport effects of dopamine and dobutamine in critically ill surgical patients. Chest 1989; 96 (01) 120-126
  • 145 Van den Berghe G, de Zegher F, Lauwers P. Dopamine suppresses pituitary function in infants and children. Crit Care Med 1994; 22 (11) 1747-1753
  • 146 Van den Berghe G, de Zegher F. Anterior pituitary function during critical illness and dopamine treatment. Crit Care Med 1996; 24 (09) 1580-1590
  • 147 Sookhai S, Wang JH, Winter D, Power C, Kirwan W, Redmond HP. Dopamine attenuates the chemoattractant effect of interleukin-8: a novel role in the systemic inflammatory response syndrome. Shock 2000; 14 (03) 295-299
  • 148 Schilling T, Gründling M, Strang CM, Möritz KU, Siegmund W, Hachenberg T. Effects of dopexamine, dobutamine or dopamine on prolactin and thyreotropin serum concentrations in high-risk surgical patients. Intensive Care Med 2004; 30 (06) 1127-1133
  • 149 Perkin RM, Levin DL, Webb R, Aquino A, Reedy J. Dobutamine: a hemodynamic evaluation in children with shock. J Pediatr 1982; 100 (06) 977-983
  • 150 Ruffolo Jr RR. The pharmacology of dobutamine. Am J Med Sci 1987; 294 (04) 244-248
  • 151 Habib DM, Padbury JF, Anas NG, Perkin RM, Minegar C. Dobutamine pharmacokinetics and pharmacodynamics in pediatric intensive care patients. Crit Care Med 1992; 20 (05) 601-608
  • 152 Harada K, Tamura M, Ito T, Suzuki T, Takada G. Effects of low-dose dobutamine on left ventricular diastolic filling in children. Pediatr Cardiol 1996; 17 (04) 220-225
  • 153 Berg RA, Donnerstein RL, Padbury JF. Dobutamine infusions in stable, critically ill children: pharmacokinetics and hemodynamic actions. Crit Care Med 1993; 21 (05) 678-686
  • 154 Bollaert PE, Bauer P, Audibert G, Lambert H, Larcan A. Effects of epinephrine on hemodynamics and oxygen metabolism in dopamine-resistant septic shock. Chest 1990; 98 (04) 949-953
  • 155 Mackenzie SJ, Kapadia F, Nimmo GR, Armstrong IR, Grant IS. Adrenaline in treatment of septic shock: effects on haemodynamics and oxygen transport. Intensive Care Med 1991; 17 (01) 36-39
  • 156 Wilson W, Lipman J, Scribante J. , et al. Septic shock: does adrenaline have a role as a first-line inotropic agent?. Anaesth Intensive Care 1992; 20 (04) 470-474
  • 157 Irazuzta J, Sullivan KJ, Garcia PC, Piva JP. Pharmacologic support of infants and children in septic shock. J Pediatr (Rio J) 2007; 83 (2, Suppl): S36-S45
  • 158 Berg RA, Padbury JF. Sulfoconjugation and renal excretion contribute to the interpatient variation of exogenous catecholamine clearance in critically ill children. Crit Care Med 1997; 25 (07) 1247-1251
  • 159 Fisher DG, Schwartz PH, Davis AL. Pharmacokinetics of exogenous epinephrine in critically ill children. Crit Care Med 1993; 21 (01) 111-117
  • 160 Martin C, Papazian L, Perrin G, Saux P, Gouin F. Norepinephrine or dopamine for the treatment of hyperdynamic septic shock?. Chest 1993; 103 (06) 1826-1831
  • 161 Klinzing S, Simon M, Reinhart K, Bredle DL, Meier-Hellmann A. High-dose vasopressin is not superior to norepinephrine in septic shock. Crit Care Med 2003; 31 (11) 2646-2650
  • 162 Delmas A, Leone M, Rousseau S, Albanèse J, Martin C. Clinical review: vasopressin and terlipressin in septic shock patients. Crit Care 2005; 9 (02) 212-222
  • 163 Matok I, Vard A, Efrati O. , et al. Terlipressin as rescue therapy for intractable hypotension due to septic shock in children. Shock 2005; 23 (04) 305-310
  • 164 Liedel JL, Meadow W, Nachman J, Koogler T, Kahana MD. Use of vasopressin in refractory hypotension in children with vasodilatory shock: five cases and a review of the literature. Pediatr Crit Care Med 2002; 3 (01) 15-18
  • 165 Vasudevan A, Lodha R, Kabra SK. Vasopressin infusion in children with catecholamine-resistant septic shock. Acta Paediatr 2005; 94 (03) 380-383
  • 166 Biban P, Gaffuri M. Vasopressin and terlipressin in neonates and children with refractory septic shock. Curr Drug Metab 2013; 14 (02) 186-192
  • 167 Lindsay CA, Barton P, Lawless S. , et al. Pharmacokinetics and pharmacodynamics of milrinone lactate in pediatric patients with septic shock. J Pediatr 1998; 132 (02) 329-334
  • 168 Irazuzta JE, Pretzlaff RK, Rowin ME. Amrinone in pediatric refractory septic shock: an open-label pharmacodynamic study. Pediatr Crit Care Med 2001; 2 (01) 24-28
  • 169 Bishara T, Seto WT, Trope A, Parshuram CS. Use of milrinone in critically ill children. Can J Hosp Pharm 2010; 63 (06) 420-428
  • 170 Papoff P, Caresta E, Versacci P, Pinto R, Moretti C, Midulla F. Beneficial effects of levosimendan in infants with sepsis-associated cardiac dysfunction: report of 2 cases. Pediatr Emerg Care 2012; 28 (10) 1062-1065
  • 171 Morelli A, De Castro S, Teboul JL. , et al. Effects of levosimendan on systemic and regional hemodynamics in septic myocardial depression. Intensive Care Med 2005; 31 (05) 638-644
  • 172 Namachivayam P, Crossland DS, Butt WW, Shekerdemian LS. Early experience with Levosimendan in children with ventricular dysfunction. Pediatr Crit Care Med 2006; 7 (05) 445-448
  • 173 Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med 1995; 332 (20) 1351-1362
  • 174 Jäättelä M, Ilvesmäki V, Voutilainen R, Stenman UH, Saksela E. Tumor necrosis factor as a potent inhibitor of adrenocorticotropin-induced cortisol production and steroidogenic P450 enzyme gene expression in cultured human fetal adrenal cells. Endocrinology 1991; 128 (01) 623-629
  • 175 Huang ZH, Gao H, Xu RB. Study on glucocorticoid receptors during intestinal ischemia shock and septic shock. Circ Shock 1987; 23 (01) 27-36
  • 176 Molijn GJ, Koper JW, van Uffelen CJ. , et al. Temperature-induced down-regulation of the glucocorticoid receptor in peripheral blood mononuclear leucocyte in patients with sepsis or septic shock. Clin Endocrinol (Oxf) 1995; 43 (02) 197-203
  • 177 Wynn JL, Wong HR. Pathophysiology and treatment of septic shock in neonates. Clin Perinatol 2010; 37 (02) 439-479
  • 178 Hatherill M, Tibby SM, Hilliard T, Turner C, Murdoch IA. Adrenal insufficiency in septic shock. Arch Dis Child 1999; 80 (01) 51-55
  • 179 Matot I, Sprung CL. Corticosteroids in septic shock: resurrection of the last rites?. Crit Care Med 1998; 26 (04) 627-630
  • 180 Briegel J, Forst H, Kellermann W, Haller M, Peter K. Haemodynamic improvement in refractory septic shock with cortisol replacement therapy. Intensive Care Med 1992; 18 (05) 318
  • 181 Markovitz BP, Goodman DM, Watson RS, Bertoch D, Zimmerman J. A retrospective cohort study of prognostic factors associated with outcome in pediatric severe sepsis: what is the role of steroids?. Pediatr Crit Care Med 2005; 6 (03) 270-274
  • 182 Atkinson SJ, Cvijanovich NZ, Thomas NJ. , et al. Corticosteroids and pediatric septic shock outcomes: a risk stratified analysis. PLoS One 2014; 9 (11) e112702
  • 183 Zimmerman JJ, Williams MD. Adjunctive corticosteroid therapy in pediatric severe sepsis: observations from the RESOLVE study. Pediatr Crit Care Med 2011; 12 (01) 2-8
  • 184 Kumar A, Roberts D, Wood KE. , et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med 2006; 34 (06) 1589-1596
  • 185 Weiss SL, Fitzgerald JC, Balamuth F. , et al. Delayed antimicrobial therapy increases mortality and organ dysfunction duration in pediatric sepsis. Crit Care Med 2014; 42 (11) 2409-2417
  • 186 Leibovici L, Shraga I, Drucker M, Konigsberger H, Samra Z, Pitlik SD. The benefit of appropriate empirical antibiotic treatment in patients with bloodstream infection. J Intern Med 1998; 244 (05) 379-386
  • 187 Marshall JC, Maier RV, Jimenez M, Dellinger EP. Source control in the management of severe sepsis and septic shock: an evidence-based review. Crit Care Med 2004; 32 (11, Suppl): S513-S526
  • 188 Knight PH, Maheshwari N, Hussain J. , et al. Complications during intrahospital transport of critically ill patients: focus on risk identification and prevention. Int J Crit Illn Inj Sci 2015; 5 (04) 256-264
  • 189 Foronda C, VanGraafeiland B, Quon R, Davidson P. Handover and transport of critically ill children: an integrative review. Int J Nurs Stud 2016; 62: 207-225
  • 190 El-Nawawy A, El-Kinany H, Hamdy El-Sayed M, Boshra N. Intravenous polyclonal immunoglobulin administration to sepsis syndrome patients: a prospective study in a pediatric intensive care unit. J Trop Pediatr 2005; 51 (05) 271-278
  • 191 Tugrul S, Ozcan PE, Akinci O. , et al. The effects of IgM-enriched immunoglobulin preparations in patients with severe sepsis [ISRCTN28863830]. Crit Care 2002; 6 (04) 357-362
  • 192 Carlet J. ; International Sepsis Forum. Immunological therapy in sepsis: currently available. Intensive Care Med 2001; 27 (Suppl. 01) S93-S103
  • 193 Rieben R, Roos A, Muizert Y, Tinguely C, Gerritsen AF, Daha MR. Immunoglobulin M-enriched human intravenous immunoglobulin prevents complement activation in vitro and in vivo in a rat model of acute inflammation. Blood 1999; 93 (03) 942-951
  • 194 Kreymann KG, de Heer G, Nierhaus A, Kluge S. Use of polyclonal immunoglobulins as adjunctive therapy for sepsis or septic shock. Crit Care Med 2007; 35 (12) 2677-2685
  • 195 Hentrich M, Fehnle K, Ostermann H. , et al. IgMA-enriched immunoglobulin in neutropenic patients with sepsis syndrome and septic shock: a randomized, controlled, multiple-center trial. Crit Care Med 2006; 34 (05) 1319-1325
  • 196 Rodríguez A, Rello J, Neira J. , et al. Effects of high-dose of intravenous immunoglobulin and antibiotics on survival for severe sepsis undergoing surgery. Shock 2005; 23 (04) 298-304
  • 197 Mouthon L, Lortholary O. Intravenous immunoglobulins in infectious diseases: where do we stand?. Clin Microbiol Infect 2003; 9 (05) 333-338
  • 198 Shah SS, Hall M, Srivastava R, Subramony A, Levin JE. Intravenous immunoglobulin in children with streptococcal toxic shock syndrome. Clin Infect Dis 2009; 49 (09) 1369-1376
  • 199 Werdan K, Pilz G, Bujdoso O. , et al; Score-Based Immunoglobulin Therapy of Sepsis (SBITS) Study Group. Score-based immunoglobulin G therapy of patients with sepsis: the SBITS study. Crit Care Med 2007; 35 (12) 2693-2701
  • 200 Brocklehurst P, Farrell B, King A. , et al; INIS Collaborative Group. Treatment of neonatal sepsis with intravenous immune globulin. N Engl J Med 2011; 365 (13) 1201-1211
  • 201 Kaul R, McGeer A, Norrby-Teglund A. , et al; The Canadian Streptococcal Study Group. Intravenous immunoglobulin therapy for streptococcal toxic shock syndrome--a comparative observational study. Clin Infect Dis 1999; 28 (04) 800-807
  • 202 Norrby-Teglund A, Stevens DL. Novel therapies in streptococcal toxic shock syndrome: attenuation of virulence factor expression and modulation of the host response. Curr Opin Infect Dis 1998; 11 (03) 285-291
  • 203 El-Wiher N, Cornell TT, Kissoon N, Shanley TP. Management and Treatment Guidelines for Sepsis in Pediatric Patients. Open Inflamm J 2011; 4 (Suppl 1-M11): 101-109
  • 204 Karam O, Tucci M, Ducruet T, Hume HA, Lacroix J, Gauvin F. ; Canadian Critical Care Trials Group; PALISI Network. Red blood cell transfusion thresholds in pediatric patients with sepsis. Pediatr Crit Care Med 2011; 12 (05) 512-518
  • 205 Rivers E, Nguyen B, Havstad S. , et al; Early Goal-Directed Therapy Collaborative Group. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med 2001; 345 (19) 1368-1377
  • 206 Laverdière C, Gauvin F, Hébert PC. , et al; Canadian Critical Care Trials Group. Survey on transfusion practices of pediatric intensivists. Pediatr Crit Care Med 2002; 3 (04) 335-340
  • 207 Nahum E, Ben-Ari J, Schonfeld T. Blood transfusion policy among European pediatric intensive care physicians. J Intensive Care Med 2004; 19 (01) 38-43
  • 208 Lacroix J, Hébert PC, Hutchison JS. , et al; TRIPICU Investigators; Canadian Critical Care Trials Group; Pediatric Acute Lung Injury and Sepsis Investigators Network. Transfusion strategies for patients in pediatric intensive care units. N Engl J Med 2007; 356 (16) 1609-1619
  • 209 Levi M. Disseminated intravascular coagulation. Crit Care Med 2007; 35 (09) 2191-2195
  • 210 Bick RL. Disseminated intravascular coagulation current concepts of etiology, pathophysiology, diagnosis, and treatment. Hematol Oncol Clin North Am 2003; 17 (01) 149-176
  • 211 Barbui T, Falanga A. Disseminated intravascular coagulation in acute leukemia. Semin Thromb Hemost 2001; 27 (06) 593-604
  • 212 Williams MD, Chalmers EA, Gibson BE. ; Haemostasis and Thrombosis Task Force, British Committee for Standards in Haematology. The investigation and management of neonatal haemostasis and thrombosis. Br J Haematol 2002; 119 (02) 295-309
  • 213 Franchini M, Manzato F. Update on the treatment of disseminated intravascular coagulation. Hematology 2004; 9 (02) 81-85
  • 214 de Kleijn ED, de Groot R, Hack CE. , et al. Activation of protein C following infusion of protein C concentrate in children with severe meningococcal sepsis and purpura fulminans: a randomized, double-blinded, placebo-controlled, dose-finding study. Crit Care Med 2003; 31 (06) 1839-1847
  • 215 Veldman A, Fischer D, Wong FY. , et al. Human protein C concentrate in the treatment of purpura fulminans: a retrospective analysis of safety and outcome in 94 pediatric patients. Crit Care 2010; 14 (04) R156
  • 216 Ettingshausen CE, Veldmann A, Beeg T, Schneider W, Jäger G, Kreuz W. Replacement therapy with protein C concentrate in infants and adolescents with meningococcal sepsis and purpura fulminans. Semin Thromb Hemost 1999; 25 (06) 537-541
  • 217 McCowen KC, Malhotra A, Bistrian BR. Stress-induced hyperglycemia. Crit Care Clin 2001; 17 (01) 107-124
  • 218 Carr ME. Diabetes mellitus: a hypercoagulable state. J Diabetes Complications 2001; 15 (01) 44-54
  • 219 Turina M, Fry DE, Polk Jr HC. Acute hyperglycemia and the innate immune system: clinical, cellular, and molecular aspects. Crit Care Med 2005; 33 (07) 1624-1633
  • 220 Branco RG, Garcia PC, Piva JP, Casartelli CH, Seibel V, Tasker RC. Glucose level and risk of mortality in pediatric septic shock. Pediatr Crit Care Med 2005; 6 (04) 470-472
  • 221 Faustino EV, Apkon M. Persistent hyperglycemia in critically ill children. J Pediatr 2005; 146 (01) 30-34
  • 222 Day KM, Haub N, Betts H, Inwald DP. Hyperglycemia is associated with morbidity in critically ill children with meningococcal sepsis. Pediatr Crit Care Med 2008; 9 (06) 636-640
  • 223 Brunkhorst FM, Engel C, Bloos F. , et al; German Competence Network Sepsis (SepNet). Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med 2008; 358 (02) 125-139
  • 224 Klein GW, Hojsak JM, Schmeidler J, Rapaport R. Hyperglycemia and outcome in the pediatric intensive care unit. J Pediatr 2008; 153 (03) 379-384
  • 225 Garcia Branco R, Tasker RC, Ramos Garcia PC, Piva JP, Dias Xavier L. Glycemic control and insulin therapy in sepsis and critical illness. J Pediatr (Rio J) 2007; 83 (5, Suppl): S128-S136
  • 226 Meyer DM, Jessen ME. ; The Extracorporeal Life Support Organization. Results of extracorporeal membrane oxygenation in children with sepsis. Ann Thorac Surg 1997; 63 (03) 756-761
  • 227 Goldman AP, Kerr SJ, Butt W. , et al. Extracorporeal support for intractable cardiorespiratory failure due to meningococcal disease. Lancet 1997; 349 (9050): 466-469
  • 228 Maclaren G, Butt W, Best D, Donath S, Taylor A. Extracorporeal membrane oxygenation for refractory septic shock in children: one institution's experience. Pediatr Crit Care Med 2007; 8 (05) 447-451
  • 229 MacLaren G, Butt W, Best D, Donath S. Central extracorporeal membrane oxygenation for refractory pediatric septic shock. Pediatr Crit Care Med 2011; 12 (02) 133-136
  • 230 Butt W, Maclaren G. Extracorporeal membrane oxygenation. F1000Prime Rep 2013; 5: 55

Address for correspondence

G. Balázs, MD
Clinical Center, Institute of Pediatrics, University of Debrecen
Nagyerdei krt 98, H-4032 Debrecen
Hungary   

  • References

  • 1 Goldstein B, Giroir B, Randolph A. ; International Consensus Conference on Pediatric Sepsis. International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005; 6 (01) 2-8
  • 2 Cinel I, Dellinger RP. Advances in pathogenesis and management of sepsis. Curr Opin Infect Dis 2007; 20 (04) 345-352
  • 3 Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S, Remick DG. The pathogenesis of sepsis. Annu Rev Pathol 2011; 6: 19-48
  • 4 Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med 2003; 348 (02) 138-150
  • 5 Lederer JA, Rodrick ML, Mannick JA. The effects of injury on the adaptive immune response. Shock 1999; 11 (03) 153-159
  • 6 Payen D, Faivre V, Lukaszewicz AC, Villa F, Goldberg P. Expression of monocyte human leukocyte antigen-DR in relation with sepsis severity and plasma mediators. Minerva Anestesiol 2009; 75 (09) 484-493
  • 7 Wynn J, Cornell TT, Wong HR, Shanley TP, Wheeler DS. The host response to sepsis and developmental impact. Pediatrics 2010; 125 (05) 1031-1041
  • 8 Wheeler DS, Zingarelli B, Wheeler WJ, Wong HR. Novel pharmacologic approaches to the management of sepsis: targeting the host inflammatory response. Recent Pat Inflamm Allergy Drug Discov 2009; 3 (02) 96-112
  • 9 Hazelzet J, Driessen GJA, Abboud P, Wheeler DS, Shanley TP, Wong HR. Sepsis. In: Wheeler DS, Wong HR, Shanley TP. , eds. Pediatric Critical Care Medicine: Basic Science and Clinical Evidence. London: Springer-Verlag; 2007: 1421-1444
  • 10 Wheeler DS, Wong HR, Zingarelli B. Pediatric sepsis - part I: “children are not small adults!”. Open Inflamm J 2011; 4: 4-15
  • 11 Weiss SL, Fitzgerald JC, Pappachan J. , et al; Sepsis Prevalence, Outcomes, and Therapies (SPROUT) Study Investigators and Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network. Global epidemiology of pediatric severe sepsis: the sepsis prevalence, outcomes, and therapies study. Am J Respir Crit Care Med 2015; 191 (10) 1147-1157
  • 12 Carcillo JA. Reducing the global burden of sepsis in infants and children: a clinical practice research agenda. Pediatr Crit Care Med 2005; 6 (3, Suppl): S157-S164
  • 13 Odetola FO, Gebremariam A, Freed GL. Patient and hospital correlates of clinical outcomes and resource utilization in severe pediatric sepsis. Pediatrics 2007; 119 (03) 487-494
  • 14 Typpo KV, Petersen NJ, Hallman DM, Markovitz BP, Mariscalco MM. Day 1 multiple organ dysfunction syndrome is associated with poor functional outcome and mortality in the pediatric intensive care unit. Pediatr Crit Care Med 2009; 10 (05) 562-570
  • 15 Kissoon N, Carcillo JA, Espinosa V. , et al; Global Sepsis Initiative Vanguard Center Contributors. World Federation of Pediatric Intensive Care and Critical Care Societies: Global Sepsis Initiative. Pediatr Crit Care Med 2011; 12 (05) 494-503
  • 16 Dellinger RP, Levy MM, Rhodes A. , et al; Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41 (02) 580-637
  • 17 Hartman ME, Linde-Zwirble WT, Angus DC, Watson RS. Trends in the epidemiology of pediatric severe sepsis*. Pediatr Crit Care Med 2013; 14 (07) 686-693
  • 18 Ruth A, McCracken CE, Fortenberry JD, Hall M, Simon HK, Hebbar KB. Pediatric severe sepsis: current trends and outcomes from the Pediatric Health Information Systems database. Pediatr Crit Care Med 2014; 15 (09) 828-838
  • 19 Butt W. Septic shock. Pediatr Clin North Am 2001; 48 (03) 601-625 , viii
  • 20 Gaines NN, Patel B, Williams EA, Cruz AT. Etiologies of septic shock in a pediatric emergency department population. Pediatr Infect Dis J 2012; 31 (11) 1203-1205
  • 21 Han YY, Carcillo JA, Dragotta MA. , et al. Early reversal of pediatric-neonatal septic shock by community physicians is associated with improved outcome. Pediatrics 2003; 112 (04) 793-799
  • 22 Kutko MC, Calarco MP, Flaherty MB. , et al. Mortality rates in pediatric septic shock with and without multiple organ system failure. Pediatr Crit Care Med 2003; 4 (03) 333-337
  • 23 Simmons ML, Durham SH, Carter CW. Pharmacological management of pediatric patients with sepsis. AACN Adv Crit Care 2012; 23 (04) 437-448 , quiz 449–450
  • 24 Bone RC, Balk RA, Cerra FB. , et al; The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest 1992; 101 (06) 1644-1655
  • 25 Levy MM, Fink MP, Marshall JC. , et al; International Sepsis Definitions Conference. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Intensive Care Med 2003; 29 (04) 530-538
  • 26 Marshall JC. SIRS and MODS: what is their relevance to the science and practice of intensive care?. Shock 2000; 14 (06) 586-589
  • 27 Weiss SL, Parker B, Bullock ME. , et al. Defining pediatric sepsis by different criteria: discrepancies in populations and implications for clinical practice. Pediatr Crit Care Med 2012; 13 (04) e219-e226
  • 28 Argent AC. Recognizing pediatric sepsis: do the concepts help us to focus appropriately?. Pediatr Crit Care Med 2016; 17 (05) 460-461
  • 29 Vincent JL, Opal SM, Marshall JC, Tracey KJ. Sepsis definitions: time for change. Lancet 2013; 381 (9868): 774-775
  • 30 Singer M, Deutschman CS, Seymour CW. , et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016; 315 (08) 801-810
  • 31 Agyeman P, Aebi C, Hirt A. , et al. Predicting bacteremia in children with cancer and fever in chemotherapy-induced neutropenia: results of the prospective multicenter SPOG 2003 FN study. Pediatr Infect Dis J 2011; 30 (07) e114-e119
  • 32 Struthers S, Underhill H, Albersheim S, Greenberg D, Dobson S. A comparison of two versus one blood culture in the diagnosis and treatment of coagulase-negative staphylococcus in the neonatal intensive care unit. J Perinatol 2002; 22 (07) 547-549
  • 33 Isaacman DJ, Karasic RB, Reynolds EA, Kost SI. Effect of number of blood cultures and volume of blood on detection of bacteremia in children. J Pediatr 1996; 128 (02) 190-195
  • 34 Kaditis AG, O'Marcaigh AS, Rhodes KH, Weaver AL, Henry NK. Yield of positive blood cultures in pediatric oncology patients by a new method of blood culture collection. Pediatr Infect Dis J 1996; 15 (07) 615-620
  • 35 Schelonka RL, Chai MK, Yoder BA, Hensley D, Brockett RM, Ascher DP. Volume of blood required to detect common neonatal pathogens. J Pediatr 1996; 129 (02) 275-278
  • 36 Buttery JP. Blood cultures in newborns and children: optimising an everyday test. Arch Dis Child Fetal Neonatal Ed 2002; 87 (01) F25-F28
  • 37 Surdulescu S, Utamsingh D, Shekar R. Phlebotomy teams reduce blood-culture contamination rate and save money. Clin Perform Qual Health Care 1998; 6 (02) 60-62
  • 38 Weinbaum FI, Lavie S, Danek M, Sixsmith D, Heinrich GF, Mills SS. Doing it right the first time: quality improvement and the contaminant blood culture. J Clin Microbiol 1997; 35 (03) 563-565
  • 39 Weinstein MP. Blood culture contamination: persisting problems and partial progress. J Clin Microbiol 2003; 41 (06) 2275-2278
  • 40 Calfee DP, Farr BM. Comparison of four antiseptic preparations for skin in the prevention of contamination of percutaneously drawn blood cultures: a randomized trial. J Clin Microbiol 2002; 40 (05) 1660-1665
  • 41 Hall KK, Lyman JA. Updated review of blood culture contamination. Clin Microbiol Rev 2006; 19 (04) 788-802
  • 42 Norberg A, Christopher NC, Ramundo ML, Bower JR, Berman SA. Contamination rates of blood cultures obtained by dedicated phlebotomy vs intravenous catheter. JAMA 2003; 289 (06) 726-729
  • 43 Hardy DJ, Hulbert BB, Migneault PC. Time to detection of positive BacT/Alert blood cultures and lack of need for routine subculture of 5- to 7-day negative cultures. J Clin Microbiol 1992; 30: 2743-2745
  • 44 Huang AH, Yan JJ, Wu JJ. Comparison of five days versus seven days of incubation for detection of positive blood cultures by the Bactec 9240 system. Eur J Clin Microbiol Infect Dis 1998; 17 (09) 637-641
  • 45 Connell TG, Rele M, Cowley D, Buttery JP, Curtis N. How reliable is a negative blood culture result? Volume of blood submitted for culture in routine practice in a children's hospital. Pediatrics 2007; 119 (05) 891-896
  • 46 Dark P, Dunn G, Chadwick P. , et al. The clinical diagnostic accuracy of rapid detection of healthcare-associated bloodstream infection in intensive care using multipathogen real-time PCR technology. BMJ Open 2011; 1 (01) e000181
  • 47 Tsalik EL, Jones D, Nicholson B. , et al. Multiplex PCR to diagnose bloodstream infections in patients admitted from the emergency department with sepsis. J Clin Microbiol 2010; 48 (01) 26-33
  • 48 Lucignano B, Ranno S, Liesenfeld O. , et al. Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in newborns and children with suspected sepsis. J Clin Microbiol 2011; 49 (06) 2252-2258
  • 49 Dark PM, Dean P, Warhurst G. Bench-to-bedside review: the promise of rapid infection diagnosis during sepsis using polymerase chain reaction-based pathogen detection. Crit Care 2009; 13 (04) 217
  • 50 Lehmann LE, Hunfeld KP, Emrich T. , et al. A multiplex real-time PCR assay for rapid detection and differentiation of 25 bacterial and fungal pathogens from whole blood samples. Med Microbiol Immunol (Berl) 2008; 197 (03) 313-324
  • 51 von Lilienfeld-Toal M, Lehmann LE, Raadts AD. , et al. Utility of a commercially available multiplex real-time PCR assay to detect bacterial and fungal pathogens in febrile neutropenia. J Clin Microbiol 2009; 47 (08) 2405-2410
  • 52 Wellinghausen N, Kochem AJ, Disqué C. , et al. Diagnosis of bacteremia in whole-blood samples by use of a commercial universal 16S rRNA gene-based PCR and sequence analysis. J Clin Microbiol 2009; 47 (09) 2759-2765
  • 53 Westh H, Lisby G, Breysse F. , et al. Multiplex real-time PCR and blood culture for identification of bloodstream pathogens in patients with suspected sepsis. Clin Microbiol Infect 2009; 15 (06) 544-551
  • 54 Watson RS, Carcillo JA. Scope and epidemiology of pediatric sepsis. Pediatr Crit Care Med 2005; 6 (3, Suppl): S3-S5
  • 55 Aneja R, Carcillo J. Differences between adult and pediatric septic shock. Minerva Anestesiol 2011; 77 (10) 986-992
  • 56 Parker MM, Shelhamer JH, Natanson C, Alling DW, Parrillo JE. Serial cardiovascular variables in survivors and nonsurvivors of human septic shock: heart rate as an early predictor of prognosis. Crit Care Med 1987; 15 (10) 923-929
  • 57 Brierley J, Carcillo JA, Choong K. , et al. Clinical practice parameters for hemodynamic support of pediatric and neonatal septic shock: 2007 update from the American College of Critical Care Medicine. Crit Care Med 2009; 37 (02) 666-688
  • 58 Practice parameters for hemodynamic support of sepsis in adults with sepsis. Task force of the American College of Critical Care Medicine, Society of Critical Care Medicine. Crit Care Med 1999; 27: 639-660
  • 59 Parker MM, Shelhamer JH, Bacharach SL. , et al. Profound but reversible myocardial depression in patients with septic shock. Ann Intern Med 1984; 100 (04) 483-490
  • 60 Pollack MM, Fields AI, Ruttimann UE. Sequential cardiopulmonary variables of infants and children in septic shock. Crit Care Med 1984; 12 (07) 554-559
  • 61 Ceneviva G, Paschall JA, Maffei F, Carcillo JA. Hemodynamic support in fluid-refractory pediatric septic shock. Pediatrics 1998; 102 (02) e19
  • 62 Feltes TF, Pignatelli R, Kleinert S, Mariscalco MM. Quantitated left ventricular systolic mechanics in children with septic shock utilizing noninvasive wall-stress analysis. Crit Care Med 1994; 22 (10) 1647-1658
  • 63 Pollack MM, Ruttimann UE, Getson PR. Pediatric risk of mortality (PRISM) score. Crit Care Med 1988; 16 (11) 1110-1116
  • 64 Dugas MA, Proulx F, de Jaeger A, Lacroix J, Lambert M. Markers of tissue hypoperfusion in pediatric septic shock. Intensive Care Med 2000; 26 (01) 75-83
  • 65 Nguyen HB, Rivers EP, Knoblich BP. , et al. Early lactate clearance is associated with improved outcome in severe sepsis and septic shock. Crit Care Med 2004; 32 (08) 1637-1642
  • 66 Arnold RC, Shapiro NI, Jones AE. , et al; Emergency Medicine Shock Research Network (EMShockNet) Investigators. Multicenter study of early lactate clearance as a determinant of survival in patients with presumed sepsis. Shock 2009; 32 (01) 35-39
  • 67 Scott HF, Brou L, Deakyne SJ, Fairclough DL, Kempe A, Bajaj L. Lactate clearance and normalization and prolonged organ dysfunction in pediatric sepsis. J Pediatr 2016; 170: 149-55.e1 , 4
  • 68 de Oliveira CF, de Oliveira DS, Gottschald AF. , et al. ACCM/PALS haemodynamic support guidelines for paediatric septic shock: an outcomes comparison with and without monitoring central venous oxygen saturation. Intensive Care Med 2008; 34 (06) 1065-1075
  • 69 Schiffmann H, Erdlenbruch B, Singer D. , et al. Assessment of cardiac output, intravascular volume status, and extravascular lung water by transpulmonary indicator dilution in critically ill neonates and infants. J Cardiothorac Vasc Anesth 2002; 16 (05) 592-597
  • 70 Deep A, Goonasekera CD, Wang Y, Brierley J. Evolution of haemodynamics and outcome of fluid-refractory septic shock in children. Intensive Care Med 2013; 39 (09) 1602-1609
  • 71 Brierley J, Peters MJ. Distinct hemodynamic patterns of septic shock at presentation to pediatric intensive care. Pediatrics 2008; 122 (04) 752-759
  • 72 Martin GS, Eaton S, Mealer M, Moss M. Extravascular lung water in patients with severe sepsis: a prospective cohort study. Crit Care 2005; 9 (02) R74-R82
  • 73 Boehne M, Baustert M, Paetzel V. , et al. Determination of cardiac output by ultrasound dilution technique in infants and children: a validation study against direct Fick principle. Br J Anaesth 2014; 112 (03) 469-476
  • 74 Wongsirimetheekul T, Khositseth A, Lertbunrian R. Non-invasive cardiac output assessment in critically ill paediatric patients. Acta Cardiol 2014; 69 (02) 167-173
  • 75 Crittendon III I, Dreyer WJ, Decker JA, Kim JJ. Ultrasound dilution: an accurate means of determining cardiac output in children. Pediatr Crit Care Med 2012; 13 (01) 42-46
  • 76 Fernandez EG, Green TP, Sweeney M. Low inferior vena caval catheters for hemodynamic and pulmonary function monitoring in pediatric critical care patients. Pediatr Crit Care Med 2004; 5 (01) 14-18
  • 77 Dahlem P, van Aalderen WM, Hamaker ME, Dijkgraaf MG, Bos AP. Incidence and short-term outcome of acute lung injury in mechanically ventilated children. Eur Respir J 2003; 22 (06) 980-985
  • 78 Kleinman ME, Chameides L, Schexnayder SM. , et al. Part 14: pediatric advanced life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122 (18) (Suppl. 03) S876-S908
  • 79 Asfar P, Calzia E, Huber-Lang M, Ignatius A, Radermacher P. Hyperoxia during septic shock--Dr. Jekyll or Mr. Hyde?. Shock 2012; 37 (01) 122-123
  • 80 Cam BV, Tuan DT, Fonsmark L. , et al. Randomized comparison of oxygen mask treatment vs. nasal continuous positive airway pressure in dengue shock syndrome with acute respiratory failure. J Trop Pediatr 2002; 48 (06) 335-339
  • 81 Khilnani P, Singhi S, Lodha R. , et al. Pediatric Sepsis Guidelines: summary for resource-limited countries. Indian J Crit Care Med 2010; 14 (01) 41-52
  • 82 International consensus conferences in intensive care medicine: ventilator-associated Lung Injury in ARDS. Am J Respir Crit Care Med 1999; 160 (06) 2118-2124
  • 83 Randolph AG. Management of acute lung injury and acute respiratory distress syndrome in children. Crit Care Med 2009; 37 (08) 2448-2454
  • 84 Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A. ; Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 2000; 342 (18) 1301-1308
  • 85 Rotta AT, Steinhorn DM. Is permissive hypercapnia a beneficial strategy for pediatric acute lung injury?. Respir Care Clin N Am 2006; 12 (03) 371-387
  • 86 Santschi M, Jouvet P, Leclerc F. , et al; PALIVE Investigators; Pediatric Acute Lung Injury and Sepsis Investigators Network (PALISI); European Society of Pediatric and Neonatal Intensive Care (ESPNIC). Acute lung injury in children: therapeutic practice and feasibility of international clinical trials. Pediatr Crit Care Med 2010; 11 (06) 681-689
  • 87 Ben Jaballah N, Khaldi A, Mnif K. , et al. High-frequency oscillatory ventilation in pediatric patients with acute respiratory failure. Pediatr Crit Care Med 2006; 7 (04) 362-367
  • 88 Arnold JH, Hanson JH, Toro-Figuero LO, Gutiérrez J, Berens RJ, Anglin DL. Prospective, randomized comparison of high-frequency oscillatory ventilation and conventional mechanical ventilation in pediatric respiratory failure. Crit Care Med 1994; 22 (10) 1530-1539
  • 89 Randolph AG, Meert KL, O'Neil ME. , et al; Pediatric Acute Lung Injury and Sepsis Investigators Network. The feasibility of conducting clinical trials in infants and children with acute respiratory failure. Am J Respir Crit Care Med 2003; 167 (10) 1334-1340
  • 90 Fengler BT. Should etomidate be used for rapid-sequence intubation induction in critically ill septic patients?. Am J Emerg Med 2008; 26 (02) 229-232
  • 91 Wagner RL, White PF, Kan PB, Rosenthal MH, Feldman D. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med 1984; 310 (22) 1415-1421
  • 92 Dörr HG, Kuhnle U, Holthausen H, Bidlingmaier F, Knorr D. Etomidate: a selective adrenocortical 11 β-hydroxylase inhibitor. Klin Wochenschr 1984; 62 (21) 1011-1013
  • 93 den Brinker M, Joosten KF, Liem O. , et al. Adrenal insufficiency in meningococcal sepsis: bioavailable cortisol levels and impact of interleukin-6 levels and intubation with etomidate on adrenal function and mortality. J Clin Endocrinol Metab 2005; 90 (09) 5110-5117
  • 94 den Brinker M, Hokken-Koelega AC, Hazelzet JA, de Jong FH, Hop WC, Joosten KF. One single dose of etomidate negatively influences adrenocortical performance for at least 24h in children with meningococcal sepsis. Intensive Care Med 2008; 34 (01) 163-168
  • 95 Jackson Jr WL. Should we use etomidate as an induction agent for endotracheal intubation in patients with septic shock?: a critical appraisal. Chest 2005; 127 (03) 1031-1038
  • 96 Bloomfield R, Noble DW. Etomidate and fatal outcome--even a single bolus dose may be detrimental for some patients. Br J Anaesth 2006; 97 (01) 116-117
  • 97 Taniguchi T, Shibata K, Yamamoto K. Ketamine inhibits endotoxin-induced shock in rats. Anesthesiology 2001; 95 (04) 928-932
  • 98 Song XM, Li JG, Wang YL. , et al. Effects of ketamine on proinflammatory cytokines and nuclear factor kappaB in polymicrobial sepsis rats. World J Gastroenterol 2006; 12 (45) 7350-7354
  • 99 Mencía SB, López-Herce JC, Freddi N. Analgesia and sedation in children: practical approach for the most frequent situations. J Pediatr (Rio J) 2007; 83 (2, Suppl): S71-S82
  • 100 Sagarin MJ, Chiang V, Sakles JC. , et al; National Emergency Airway Registry (NEAR) investigators. Rapid sequence intubation for pediatric emergency airway management. Pediatr Emerg Care 2002; 18 (06) 417-423
  • 101 Sun CY, Lee KC, Lin IH. , et al. Near-infrared light device can improve intravenous cannulation in critically ill children. Pediatr Neonatol 2013; 54: 194-197
  • 102 Lillis KA, Jaffe DM. Prehospital intravenous access in children. Ann Emerg Med 1992; 21 (12) 1430-1434
  • 103 Stovroff M, Teague WG. Intravenous access in infants and children. Pediatr Clin North Am 1998; 45 (06) 1373-1393 , viii
  • 104 Idris AH, Melker RJ. High-flow sheaths for pediatric fluid resuscitation: a comparison of flow rates with standard pediatric catheters. Pediatr Emerg Care 1992; 8 (03) 119-122
  • 105 Rosetti VA, Thompson BM, Miller J, Mateer JR, Aprahamian C. Intraosseous infusion: an alternative route of pediatric intravascular access. Ann Emerg Med 1985; 14 (09) 885-888
  • 106 Seigler RS, Tecklenburg FW, Shealy R. Prehospital intraosseous infusion by emergency medical services personnel: a prospective study. Pediatrics 1989; 84 (01) 173-177
  • 107 Glaeser PW, Hellmich TR, Szewczuga D, Losek JD, Smith DS. Five-year experience in prehospital intraosseous infusions in children and adults. Ann Emerg Med 1993; 22 (07) 1119-1124
  • 108 Fiorito BA, Mirza F, Doran TM. , et al. Intraosseous access in the setting of pediatric critical care transport. Pediatr Crit Care Med 2005; 6 (01) 50-53
  • 109 Horton MA, Beamer C. Powered intraosseous insertion provides safe and effective vascular access for pediatric emergency patients. Pediatr Emerg Care 2008; 24 (06) 347-350
  • 110 Ngo NT, Cao XT, Kneen R. , et al. Acute management of dengue shock syndrome: a randomized double-blind comparison of 4 intravenous fluid regimens in the first hour. Clin Infect Dis 2001; 32 (02) 204-213
  • 111 Wills BA, Nguyen MD, Ha TL. , et al. Comparison of three fluid solutions for resuscitation in dengue shock syndrome. N Engl J Med 2005; 353 (09) 877-889
  • 112 Dung NM, Day NP, Tam DT. , et al. Fluid replacement in dengue shock syndrome: a randomized, double-blind comparison of four intravenous-fluid regimens. Clin Infect Dis 1999; 29 (04) 787-794
  • 113 Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R. ; SAFE Study Investigators. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350 (22) 2247-2256
  • 114 Upadhyay M, Singhi S, Murlidharan J, Kaur N, Majumdar S. Randomized evaluation of fluid resuscitation with crystalloid (saline) and colloid (polymer from degraded gelatin in saline) in pediatric septic shock. Indian Pediatr 2005; 42 (03) 223-231
  • 115 Akech S, Ledermann H, Maitland K. Choice of fluids for resuscitation in children with severe infection and shock: systematic review. BMJ 2010; 341: c4416
  • 116 Ford N, Hargreaves S, Shanks L. Mortality after fluid bolus in children with shock due to sepsis or severe infection: a systematic review and meta-analysis. PLoS One 2012; 7 (08) e43953
  • 117 Mendes PV, Zampieri FG, Park M. Is There a Role for Balanced Solutions in Septic Patients?. Shock 2017; 47 (1S, Suppl 1): 30-34
  • 118 Guidet B, Soni N, Della Rocca G. , et al. A balanced view of balanced solutions. Crit Care 2010; 14 (05) 325
  • 119 Vincent JL, Orbegozo Cortés D, Acheampong A. Current haemodynamic management of septic shock. Presse Med 2016; 45 (4 Pt 2): e99-e103
  • 120 Allen SJ. Fluid therapy and outcome: balance is best. J Extra Corpor Technol 2014; 46 (01) 28-32
  • 121 Orbegozo Cortés D, Rayo Bonor A, Vincent JL. Isotonic crystalloid solutions: a structured review of the literature. Br J Anaesth 2014; 112 (06) 968-981
  • 122 Roquilly A, Loutrel O, Cinotti R. , et al. Balanced versus chloride-rich solutions for fluid resuscitation in brain-injured patients: a randomised double-blind pilot study. Crit Care 2013; 17 (02) R77
  • 123 Williams EL, Hildebrand KL, McCormick SA, Bedel MJ. The effect of intravenous lactated Ringer's solution versus 0.9% sodium chloride solution on serum osmolality in human volunteers. Anesth Analg 1999; 88 (05) 999-1003
  • 124 Stoner MJ, Goodman DG, Cohen DM, Fernandez SA, Hall MW. Rapid fluid resuscitation in pediatrics: testing the American College of Critical Care Medicine guideline. Ann Emerg Med 2007; 50 (05) 601-607
  • 125 Carcillo JA, Davis AL, Zaritsky A. Role of early fluid resuscitation in pediatric septic shock. JAMA 1991; 266 (09) 1242-1245
  • 126 Maitland K, Kiguli S, Opoka RO. , et al; FEAST Trial Group. Mortality after fluid bolus in African children with severe infection. N Engl J Med 2011; 364 (26) 2483-2495
  • 127 Santhanam I, Sangareddi S, Venkataraman S, Kissoon N, Thiruvengadamudayan V, Kasthuri RK. A prospective randomized controlled study of two fluid regimens in the initial management of septic shock in the emergency department. Pediatr Emerg Care 2008; 24 (10) 647-655
  • 128 Zadrobilek E, Hackl W, Sporn P, Steinbereithner K. Effect of large volume replacement with balanced electrolyte solutions on extravascular lung water in surgical patients with sepsis syndrome. Intensive Care Med 1989; 15 (08) 505-510
  • 129 Cruz AT, Perry AM, Williams EA, Graf JM, Wuestner ER, Patel B. Implementation of goal-directed therapy for children with suspected sepsis in the emergency department. Pediatrics 2011; 127 (03) e758-e766
  • 130 Goldstein SL, Somers MJ, Baum MA. , et al. Pediatric patients with multi-organ dysfunction syndrome receiving continuous renal replacement therapy. Kidney Int 2005; 67 (02) 653-658
  • 131 Askenazi D. Evaluation and management of critically ill children with acute kidney injury. Curr Opin Pediatr 2011; 23 (02) 201-207
  • 132 Gulla KM, Sachdev A, Gupta D, Gupta N, Anand K, Pruthi PK. Continuous renal replacement therapy in children with severe sepsis and multiorgan dysfunction - a pilot study on timing of initiation. Indian J Crit Care Med 2015; 19 (10) 613-617
  • 133 Ninis N, Phillips C, Bailey L. , et al. The role of healthcare delivery in the outcome of meningococcal disease in children: case-control study of fatal and non-fatal cases. BMJ 2005; 330 (7506): 1475
  • 134 Thompson MJ, Ninis N, Perera R. , et al. Clinical recognition of meningococcal disease in children and adolescents. Lancet 2006; 367 (9508): 397-403
  • 135 Ushay HM, Notterman DA. Pharmacology of pediatric resuscitation. Pediatr Clin North Am 1997; 44 (01) 207-233
  • 136 Bellomo R, Chapman M, Finfer S, Hickling K, Myburgh J. ; Australian and New Zealand Intensive Care Society (ANZICS) Clinical Trials Group. Low-dose dopamine in patients with early renal dysfunction: a placebo-controlled randomised trial. Lancet 2000; 356 (9248): 2139-2143
  • 137 Kellum JA, M Decker J. Use of dopamine in acute renal failure: a meta-analysis. Crit Care Med 2001; 29 (08) 1526-1531
  • 138 Zacharias M, Mugawar M, Herbison GP. , et al. Interventions for protecting renal function in the perioperative period. Cochrane Database Syst Rev 2013; (09) CD003590
  • 139 De Backer D, Aldecoa C, Njimi H, Vincent JL. Dopamine versus norepinephrine in the treatment of septic shock: a meta-analysis*. Crit Care Med 2012; 40 (03) 725-730
  • 140 Avni T, Lador A, Lev S, Leibovici L, Paul M, Grossman A. Vasopressors for the treatment of septic shock: systematic review and meta-analysis. PLoS One 2015; 10 (08) e0129305
  • 141 Ventura AM, Shieh HH, Bousso A. , et al. Double-blind prospective randomized controlled trial of dopamine versus epinephrine as first-line vasoactive drugs in pediatric septic shock. Crit Care Med 2015; 43 (11) 2292-2302
  • 142 Jakob SM, Ruokonen E, Takala J. Effects of dopamine on systemic and regional blood flow and metabolism in septic and cardiac surgery patients. Shock 2002; 18 (01) 8-13
  • 143 Dive A, Foret F, Jamart J, Bulpa P, Installé E. Effect of dopamine on gastrointestinal motility during critical illness. Intensive Care Med 2000; 26 (07) 901-907
  • 144 Shoemaker WC, Appel PL, Kram HB, Duarte D, Harrier HD, Ocampo HA. Comparison of hemodynamic and oxygen transport effects of dopamine and dobutamine in critically ill surgical patients. Chest 1989; 96 (01) 120-126
  • 145 Van den Berghe G, de Zegher F, Lauwers P. Dopamine suppresses pituitary function in infants and children. Crit Care Med 1994; 22 (11) 1747-1753
  • 146 Van den Berghe G, de Zegher F. Anterior pituitary function during critical illness and dopamine treatment. Crit Care Med 1996; 24 (09) 1580-1590
  • 147 Sookhai S, Wang JH, Winter D, Power C, Kirwan W, Redmond HP. Dopamine attenuates the chemoattractant effect of interleukin-8: a novel role in the systemic inflammatory response syndrome. Shock 2000; 14 (03) 295-299
  • 148 Schilling T, Gründling M, Strang CM, Möritz KU, Siegmund W, Hachenberg T. Effects of dopexamine, dobutamine or dopamine on prolactin and thyreotropin serum concentrations in high-risk surgical patients. Intensive Care Med 2004; 30 (06) 1127-1133
  • 149 Perkin RM, Levin DL, Webb R, Aquino A, Reedy J. Dobutamine: a hemodynamic evaluation in children with shock. J Pediatr 1982; 100 (06) 977-983
  • 150 Ruffolo Jr RR. The pharmacology of dobutamine. Am J Med Sci 1987; 294 (04) 244-248
  • 151 Habib DM, Padbury JF, Anas NG, Perkin RM, Minegar C. Dobutamine pharmacokinetics and pharmacodynamics in pediatric intensive care patients. Crit Care Med 1992; 20 (05) 601-608
  • 152 Harada K, Tamura M, Ito T, Suzuki T, Takada G. Effects of low-dose dobutamine on left ventricular diastolic filling in children. Pediatr Cardiol 1996; 17 (04) 220-225
  • 153 Berg RA, Donnerstein RL, Padbury JF. Dobutamine infusions in stable, critically ill children: pharmacokinetics and hemodynamic actions. Crit Care Med 1993; 21 (05) 678-686
  • 154 Bollaert PE, Bauer P, Audibert G, Lambert H, Larcan A. Effects of epinephrine on hemodynamics and oxygen metabolism in dopamine-resistant septic shock. Chest 1990; 98 (04) 949-953
  • 155 Mackenzie SJ, Kapadia F, Nimmo GR, Armstrong IR, Grant IS. Adrenaline in treatment of septic shock: effects on haemodynamics and oxygen transport. Intensive Care Med 1991; 17 (01) 36-39
  • 156 Wilson W, Lipman J, Scribante J. , et al. Septic shock: does adrenaline have a role as a first-line inotropic agent?. Anaesth Intensive Care 1992; 20 (04) 470-474
  • 157 Irazuzta J, Sullivan KJ, Garcia PC, Piva JP. Pharmacologic support of infants and children in septic shock. J Pediatr (Rio J) 2007; 83 (2, Suppl): S36-S45
  • 158 Berg RA, Padbury JF. Sulfoconjugation and renal excretion contribute to the interpatient variation of exogenous catecholamine clearance in critically ill children. Crit Care Med 1997; 25 (07) 1247-1251
  • 159 Fisher DG, Schwartz PH, Davis AL. Pharmacokinetics of exogenous epinephrine in critically ill children. Crit Care Med 1993; 21 (01) 111-117
  • 160 Martin C, Papazian L, Perrin G, Saux P, Gouin F. Norepinephrine or dopamine for the treatment of hyperdynamic septic shock?. Chest 1993; 103 (06) 1826-1831
  • 161 Klinzing S, Simon M, Reinhart K, Bredle DL, Meier-Hellmann A. High-dose vasopressin is not superior to norepinephrine in septic shock. Crit Care Med 2003; 31 (11) 2646-2650
  • 162 Delmas A, Leone M, Rousseau S, Albanèse J, Martin C. Clinical review: vasopressin and terlipressin in septic shock patients. Crit Care 2005; 9 (02) 212-222
  • 163 Matok I, Vard A, Efrati O. , et al. Terlipressin as rescue therapy for intractable hypotension due to septic shock in children. Shock 2005; 23 (04) 305-310
  • 164 Liedel JL, Meadow W, Nachman J, Koogler T, Kahana MD. Use of vasopressin in refractory hypotension in children with vasodilatory shock: five cases and a review of the literature. Pediatr Crit Care Med 2002; 3 (01) 15-18
  • 165 Vasudevan A, Lodha R, Kabra SK. Vasopressin infusion in children with catecholamine-resistant septic shock. Acta Paediatr 2005; 94 (03) 380-383
  • 166 Biban P, Gaffuri M. Vasopressin and terlipressin in neonates and children with refractory septic shock. Curr Drug Metab 2013; 14 (02) 186-192
  • 167 Lindsay CA, Barton P, Lawless S. , et al. Pharmacokinetics and pharmacodynamics of milrinone lactate in pediatric patients with septic shock. J Pediatr 1998; 132 (02) 329-334
  • 168 Irazuzta JE, Pretzlaff RK, Rowin ME. Amrinone in pediatric refractory septic shock: an open-label pharmacodynamic study. Pediatr Crit Care Med 2001; 2 (01) 24-28
  • 169 Bishara T, Seto WT, Trope A, Parshuram CS. Use of milrinone in critically ill children. Can J Hosp Pharm 2010; 63 (06) 420-428
  • 170 Papoff P, Caresta E, Versacci P, Pinto R, Moretti C, Midulla F. Beneficial effects of levosimendan in infants with sepsis-associated cardiac dysfunction: report of 2 cases. Pediatr Emerg Care 2012; 28 (10) 1062-1065
  • 171 Morelli A, De Castro S, Teboul JL. , et al. Effects of levosimendan on systemic and regional hemodynamics in septic myocardial depression. Intensive Care Med 2005; 31 (05) 638-644
  • 172 Namachivayam P, Crossland DS, Butt WW, Shekerdemian LS. Early experience with Levosimendan in children with ventricular dysfunction. Pediatr Crit Care Med 2006; 7 (05) 445-448
  • 173 Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med 1995; 332 (20) 1351-1362
  • 174 Jäättelä M, Ilvesmäki V, Voutilainen R, Stenman UH, Saksela E. Tumor necrosis factor as a potent inhibitor of adrenocorticotropin-induced cortisol production and steroidogenic P450 enzyme gene expression in cultured human fetal adrenal cells. Endocrinology 1991; 128 (01) 623-629
  • 175 Huang ZH, Gao H, Xu RB. Study on glucocorticoid receptors during intestinal ischemia shock and septic shock. Circ Shock 1987; 23 (01) 27-36
  • 176 Molijn GJ, Koper JW, van Uffelen CJ. , et al. Temperature-induced down-regulation of the glucocorticoid receptor in peripheral blood mononuclear leucocyte in patients with sepsis or septic shock. Clin Endocrinol (Oxf) 1995; 43 (02) 197-203
  • 177 Wynn JL, Wong HR. Pathophysiology and treatment of septic shock in neonates. Clin Perinatol 2010; 37 (02) 439-479
  • 178 Hatherill M, Tibby SM, Hilliard T, Turner C, Murdoch IA. Adrenal insufficiency in septic shock. Arch Dis Child 1999; 80 (01) 51-55
  • 179 Matot I, Sprung CL. Corticosteroids in septic shock: resurrection of the last rites?. Crit Care Med 1998; 26 (04) 627-630
  • 180 Briegel J, Forst H, Kellermann W, Haller M, Peter K. Haemodynamic improvement in refractory septic shock with cortisol replacement therapy. Intensive Care Med 1992; 18 (05) 318
  • 181 Markovitz BP, Goodman DM, Watson RS, Bertoch D, Zimmerman J. A retrospective cohort study of prognostic factors associated with outcome in pediatric severe sepsis: what is the role of steroids?. Pediatr Crit Care Med 2005; 6 (03) 270-274
  • 182 Atkinson SJ, Cvijanovich NZ, Thomas NJ. , et al. Corticosteroids and pediatric septic shock outcomes: a risk stratified analysis. PLoS One 2014; 9 (11) e112702
  • 183 Zimmerman JJ, Williams MD. Adjunctive corticosteroid therapy in pediatric severe sepsis: observations from the RESOLVE study. Pediatr Crit Care Med 2011; 12 (01) 2-8
  • 184 Kumar A, Roberts D, Wood KE. , et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med 2006; 34 (06) 1589-1596
  • 185 Weiss SL, Fitzgerald JC, Balamuth F. , et al. Delayed antimicrobial therapy increases mortality and organ dysfunction duration in pediatric sepsis. Crit Care Med 2014; 42 (11) 2409-2417
  • 186 Leibovici L, Shraga I, Drucker M, Konigsberger H, Samra Z, Pitlik SD. The benefit of appropriate empirical antibiotic treatment in patients with bloodstream infection. J Intern Med 1998; 244 (05) 379-386
  • 187 Marshall JC, Maier RV, Jimenez M, Dellinger EP. Source control in the management of severe sepsis and septic shock: an evidence-based review. Crit Care Med 2004; 32 (11, Suppl): S513-S526
  • 188 Knight PH, Maheshwari N, Hussain J. , et al. Complications during intrahospital transport of critically ill patients: focus on risk identification and prevention. Int J Crit Illn Inj Sci 2015; 5 (04) 256-264
  • 189 Foronda C, VanGraafeiland B, Quon R, Davidson P. Handover and transport of critically ill children: an integrative review. Int J Nurs Stud 2016; 62: 207-225
  • 190 El-Nawawy A, El-Kinany H, Hamdy El-Sayed M, Boshra N. Intravenous polyclonal immunoglobulin administration to sepsis syndrome patients: a prospective study in a pediatric intensive care unit. J Trop Pediatr 2005; 51 (05) 271-278
  • 191 Tugrul S, Ozcan PE, Akinci O. , et al. The effects of IgM-enriched immunoglobulin preparations in patients with severe sepsis [ISRCTN28863830]. Crit Care 2002; 6 (04) 357-362
  • 192 Carlet J. ; International Sepsis Forum. Immunological therapy in sepsis: currently available. Intensive Care Med 2001; 27 (Suppl. 01) S93-S103
  • 193 Rieben R, Roos A, Muizert Y, Tinguely C, Gerritsen AF, Daha MR. Immunoglobulin M-enriched human intravenous immunoglobulin prevents complement activation in vitro and in vivo in a rat model of acute inflammation. Blood 1999; 93 (03) 942-951
  • 194 Kreymann KG, de Heer G, Nierhaus A, Kluge S. Use of polyclonal immunoglobulins as adjunctive therapy for sepsis or septic shock. Crit Care Med 2007; 35 (12) 2677-2685
  • 195 Hentrich M, Fehnle K, Ostermann H. , et al. IgMA-enriched immunoglobulin in neutropenic patients with sepsis syndrome and septic shock: a randomized, controlled, multiple-center trial. Crit Care Med 2006; 34 (05) 1319-1325
  • 196 Rodríguez A, Rello J, Neira J. , et al. Effects of high-dose of intravenous immunoglobulin and antibiotics on survival for severe sepsis undergoing surgery. Shock 2005; 23 (04) 298-304
  • 197 Mouthon L, Lortholary O. Intravenous immunoglobulins in infectious diseases: where do we stand?. Clin Microbiol Infect 2003; 9 (05) 333-338
  • 198 Shah SS, Hall M, Srivastava R, Subramony A, Levin JE. Intravenous immunoglobulin in children with streptococcal toxic shock syndrome. Clin Infect Dis 2009; 49 (09) 1369-1376
  • 199 Werdan K, Pilz G, Bujdoso O. , et al; Score-Based Immunoglobulin Therapy of Sepsis (SBITS) Study Group. Score-based immunoglobulin G therapy of patients with sepsis: the SBITS study. Crit Care Med 2007; 35 (12) 2693-2701
  • 200 Brocklehurst P, Farrell B, King A. , et al; INIS Collaborative Group. Treatment of neonatal sepsis with intravenous immune globulin. N Engl J Med 2011; 365 (13) 1201-1211
  • 201 Kaul R, McGeer A, Norrby-Teglund A. , et al; The Canadian Streptococcal Study Group. Intravenous immunoglobulin therapy for streptococcal toxic shock syndrome--a comparative observational study. Clin Infect Dis 1999; 28 (04) 800-807
  • 202 Norrby-Teglund A, Stevens DL. Novel therapies in streptococcal toxic shock syndrome: attenuation of virulence factor expression and modulation of the host response. Curr Opin Infect Dis 1998; 11 (03) 285-291
  • 203 El-Wiher N, Cornell TT, Kissoon N, Shanley TP. Management and Treatment Guidelines for Sepsis in Pediatric Patients. Open Inflamm J 2011; 4 (Suppl 1-M11): 101-109
  • 204 Karam O, Tucci M, Ducruet T, Hume HA, Lacroix J, Gauvin F. ; Canadian Critical Care Trials Group; PALISI Network. Red blood cell transfusion thresholds in pediatric patients with sepsis. Pediatr Crit Care Med 2011; 12 (05) 512-518
  • 205 Rivers E, Nguyen B, Havstad S. , et al; Early Goal-Directed Therapy Collaborative Group. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med 2001; 345 (19) 1368-1377
  • 206 Laverdière C, Gauvin F, Hébert PC. , et al; Canadian Critical Care Trials Group. Survey on transfusion practices of pediatric intensivists. Pediatr Crit Care Med 2002; 3 (04) 335-340
  • 207 Nahum E, Ben-Ari J, Schonfeld T. Blood transfusion policy among European pediatric intensive care physicians. J Intensive Care Med 2004; 19 (01) 38-43
  • 208 Lacroix J, Hébert PC, Hutchison JS. , et al; TRIPICU Investigators; Canadian Critical Care Trials Group; Pediatric Acute Lung Injury and Sepsis Investigators Network. Transfusion strategies for patients in pediatric intensive care units. N Engl J Med 2007; 356 (16) 1609-1619
  • 209 Levi M. Disseminated intravascular coagulation. Crit Care Med 2007; 35 (09) 2191-2195
  • 210 Bick RL. Disseminated intravascular coagulation current concepts of etiology, pathophysiology, diagnosis, and treatment. Hematol Oncol Clin North Am 2003; 17 (01) 149-176
  • 211 Barbui T, Falanga A. Disseminated intravascular coagulation in acute leukemia. Semin Thromb Hemost 2001; 27 (06) 593-604
  • 212 Williams MD, Chalmers EA, Gibson BE. ; Haemostasis and Thrombosis Task Force, British Committee for Standards in Haematology. The investigation and management of neonatal haemostasis and thrombosis. Br J Haematol 2002; 119 (02) 295-309
  • 213 Franchini M, Manzato F. Update on the treatment of disseminated intravascular coagulation. Hematology 2004; 9 (02) 81-85
  • 214 de Kleijn ED, de Groot R, Hack CE. , et al. Activation of protein C following infusion of protein C concentrate in children with severe meningococcal sepsis and purpura fulminans: a randomized, double-blinded, placebo-controlled, dose-finding study. Crit Care Med 2003; 31 (06) 1839-1847
  • 215 Veldman A, Fischer D, Wong FY. , et al. Human protein C concentrate in the treatment of purpura fulminans: a retrospective analysis of safety and outcome in 94 pediatric patients. Crit Care 2010; 14 (04) R156
  • 216 Ettingshausen CE, Veldmann A, Beeg T, Schneider W, Jäger G, Kreuz W. Replacement therapy with protein C concentrate in infants and adolescents with meningococcal sepsis and purpura fulminans. Semin Thromb Hemost 1999; 25 (06) 537-541
  • 217 McCowen KC, Malhotra A, Bistrian BR. Stress-induced hyperglycemia. Crit Care Clin 2001; 17 (01) 107-124
  • 218 Carr ME. Diabetes mellitus: a hypercoagulable state. J Diabetes Complications 2001; 15 (01) 44-54
  • 219 Turina M, Fry DE, Polk Jr HC. Acute hyperglycemia and the innate immune system: clinical, cellular, and molecular aspects. Crit Care Med 2005; 33 (07) 1624-1633
  • 220 Branco RG, Garcia PC, Piva JP, Casartelli CH, Seibel V, Tasker RC. Glucose level and risk of mortality in pediatric septic shock. Pediatr Crit Care Med 2005; 6 (04) 470-472
  • 221 Faustino EV, Apkon M. Persistent hyperglycemia in critically ill children. J Pediatr 2005; 146 (01) 30-34
  • 222 Day KM, Haub N, Betts H, Inwald DP. Hyperglycemia is associated with morbidity in critically ill children with meningococcal sepsis. Pediatr Crit Care Med 2008; 9 (06) 636-640
  • 223 Brunkhorst FM, Engel C, Bloos F. , et al; German Competence Network Sepsis (SepNet). Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med 2008; 358 (02) 125-139
  • 224 Klein GW, Hojsak JM, Schmeidler J, Rapaport R. Hyperglycemia and outcome in the pediatric intensive care unit. J Pediatr 2008; 153 (03) 379-384
  • 225 Garcia Branco R, Tasker RC, Ramos Garcia PC, Piva JP, Dias Xavier L. Glycemic control and insulin therapy in sepsis and critical illness. J Pediatr (Rio J) 2007; 83 (5, Suppl): S128-S136
  • 226 Meyer DM, Jessen ME. ; The Extracorporeal Life Support Organization. Results of extracorporeal membrane oxygenation in children with sepsis. Ann Thorac Surg 1997; 63 (03) 756-761
  • 227 Goldman AP, Kerr SJ, Butt W. , et al. Extracorporeal support for intractable cardiorespiratory failure due to meningococcal disease. Lancet 1997; 349 (9050): 466-469
  • 228 Maclaren G, Butt W, Best D, Donath S, Taylor A. Extracorporeal membrane oxygenation for refractory septic shock in children: one institution's experience. Pediatr Crit Care Med 2007; 8 (05) 447-451
  • 229 MacLaren G, Butt W, Best D, Donath S. Central extracorporeal membrane oxygenation for refractory pediatric septic shock. Pediatr Crit Care Med 2011; 12 (02) 133-136
  • 230 Butt W, Maclaren G. Extracorporeal membrane oxygenation. F1000Prime Rep 2013; 5: 55

Zoom Image
Fig. 1Stepwise management of hemodynamic support in infants and children with septic shock.