Skip main navigation

Part 9: Post–Cardiac Arrest Care

2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care
Originally publishedhttps://doi.org/10.1161/CIRCULATIONAHA.110.971002Circulation. 2010;122:S768–S786

There is increasing recognition that systematic post–cardiac arrest care after return of spontaneous circulation (ROSC) can improve the likelihood of patient survival with good quality of life. This is based in part on the publication of results of randomized controlled clinical trials as well as a description of the post–cardiac arrest syndrome.13 Post–cardiac arrest care has significant potential to reduce early mortality caused by hemodynamic instability and later morbidity and mortality from multiorgan failure and brain injury.3,4 This section summarizes our evolving understanding of the hemodynamic, neurological, and metabolic abnormalities encountered in patients who are initially resuscitated from cardiac arrest.

The initial objectives of post–cardiac arrest care are to

  • Optimize cardiopulmonary function and vital organ perfusion.

  • After out-of-hospital cardiac arrest, transport patient to an appropriate hospital with a comprehensive post–cardiac arrest treatment system of care that includes acute coronary interventions, neurological care, goal-directed critical care, and hypothermia.

  • Transport the in-hospital post–cardiac arrest patient to an appropriate critical-care unit capable of providing comprehensive post–cardiac arrest care.

  • Try to identify and treat the precipitating causes of the arrest and prevent recurrent arrest.

Subsequent objectives of post–cardiac arrest care are to
  • Control body temperature to optimize survival and neurological recovery

  • Identify and treat acute coronary syndromes (ACS)

  • Optimize mechanical ventilation to minimize lung injury

  • Reduce the risk of multiorgan injury and support organ function if required

  • Objectively assess prognosis for recovery

  • Assist survivors with rehabilitation services when required

Systems of Care for Improving Post–Cardiac Arrest Outcomes

Post–cardiac arrest care is a critical component of advanced life support (Figure). Most deaths occur during the first 24 hours after cardiac arrest.5,6 The best hospital care for patients with ROSC after cardiac arrest is not completely known, but there is increasing interest in identifying and optimizing practices that are likely to improve outcomes (Table 1).7 Positive associations have been noted between the likelihood of survival and the number of cardiac arrest cases treated at any individual hospital.8,9 Because multiple organ systems are affected after cardiac arrest, successful post–cardiac arrest care will benefit from the development of system-wide plans for proactive treatment of these patients. For example, restoration of blood pressure and gas exchange does not ensure survival and functional recovery. Significant cardiovascular dysfunction can develop, requiring support of blood flow and ventilation, including intravascular volume expansion, vasoactive and inotropic drugs, and invasive devices. Therapeutic hypothermia and treatment of the underlying cause of cardiac arrest impacts survival and neurological outcomes. Protocolized hemodynamic optimization and multidisciplinary early goal-directed therapy protocols have been introduced as part of a bundle of care to improve survival rather than single interventions.1012 The data suggests that proactive titration of post–cardiac arrest hemodynamics to levels intended to ensure organ perfusion and oxygenation may improve outcomes. There are multiple specific options for acheiving these goals, and it is difficult to distinguish between the benefit of protocols or any specific component of care that is most important.

Figure.

Figure. Post–cardiac arrest care algorithm.

Table 1. Multiple System Approach to Post–Cardiac Arrest Care

Ventilation Hemodynamics Cardiovascular Neurological Metabolic
  • Capnography

    • Rationale: Confirm secure airway and titrate ventilation

    • Endotracheal tube when possible for comatose patients

    • Petco2∼35–40 mm Hg

    • Paco2∼40–45 mm Hg

  • Frequent Blood Pressure Monitoring/Arterial-line

    • Rationale: Maintain perfusion and prevent recurrent hypotension

    • Mean arterial pressure ≥65 mm Hg or systolic blood pressure ≥90 mm Hg

  • Continuous Cardiac Monitoring

    • Rationale: Detect recurrent arrhythmia

    • No prophylactic antiarrhythmics

    • Treat arrhythmias as required

    • Remove reversible causes

  • Serial Neurological Exam

    • Rationale: Serial examinations define coma, brain injury, and prognosis

    • Response to verbal commands or physical stimulation

    • Pupillary light and corneal reflex, spontaneous eye movement

    • Gag, cough, spontaneous breaths

  • Serial Lactate

    • Rationale: Confirm adequate perfusion

  • Chest X-ray

    • Rationale: Confirm secure airway and detect causes or complications of arrest: pneumonitis, pneumonia, pulmonary edema

  • Treat Hypotension

    • Rationale: Maintain perfusion

    • Fluid bolus if tolerated

    • Dopamine 5–10 mcg/kg per min

    • Norepinephrine 0.1–0.5 mcg/kg per min

    • Epinephrine 0.1–0.5 mcg/kg per min

  • 12-lead ECG/Troponin

    • Rationale: Detect Acute Coronary Syndrome/ST-Elevation Myocardial Infarction; Assess QT interval

  • EEG Monitoring If Comatose

    • Rationale: Exclude seizures

    • Anticonvulsants if seizing

  • Serum Potassium

    • Rationale: Avoid hypokalemia which promotes arrhythmias

    • Replace to maintain K >3.5 mEq/L

  • Pulse Oximetry/ABG

    • Rationale: Maintain adequate oxygenation and minimize Fio2

    • Spo2 ≥94%

    • Pao2∼100 mm Hg

    • Reduce Fio2 as tolerated

    • Pao2/Fio2 ratio to follow acute lung injury

  • Treat Acute Coronary Syndrome

    • Aspirin/heparin

    • Transfer to acute coronary treatment center

    • Consider emergent PCI or fibrinolysis

  • Core Temperature Measurement If Comatose

    • Rationale: Minimize brain injury and improve outcome

    • Prevent hyperpyrexia >37.7°C

    • Induce therapeutic hypothermia if no contraindications

    • Cold IV fluid bolus 30 mL/kg if no contraindication

    • Surface or endovascular cooling for 32°C–34°C×24 hours

    • After 24 hours, slow rewarming 0.25°C/hr

  • Urine Output, Serum Creatinine

    • Rationale: Detect acute kidney injury

    • Maintain euvolemia

    • Renal replacement therapy if indicated

  • Mechanical Ventilation

    • Rationale: Minimize acute lung injury, potential oxygen toxicity

    • Tidal Volume 6–8 mL/kg

    • Titrate minute ventilation to Petco2∼35–40 mm Hg Paco2∼40–45 mm Hg

    • Reduce Fio2 as tolerated to keep Spo2 or Sao2 ≥94%

  • Echocardiogram

    • Rationale: Detect global stunning, wall-motion abnormalities, structural problems or cardiomyopathy

  • Consider Non-enhanced CT Scan

    • Rationale: Exclude primary intracranial process

  • Serum Glucose

    • Rationale: Detect hyperglycemia and hypoglycemia

    • Treat hypoglycemia (<80 mg/dL) with dextrose

    • Treat hyperglycemia to target glucose 144–180 mg/dL

    • Local insulin protocols

  • Treat Myocardial Stunning

    • Fluids to optimize volume status (requires clinical judgment)

    • Dobutamine 5–10 mcg/kg per min

    • Mechanical augmentation (IABP)

  • Sedation/Muscle Relaxation

    • Rationale: To control shivering, agitation, or ventilator desynchrony as needed

  • Avoid Hypotonic Fluids

    • Rationale: May increase edema, including cerebral edema

A comprehensive, structured, multidisciplinary system of care should be implemented in a consistent manner for the treatment of post–cardiac arrest patients (Class I, LOE B). Programs should include as part of structured interventions therapeutic hypothermia; optimization of hemodynamics and gas exchange; immediate coronary reperfusion when indicated for restoration of coronary blood flow with percutaneous coronary intervention (PCI); glycemic control; and neurological diagnosis, management, and prognostication.

Overview of Post–Cardiac Arrest Care

The provider of CPR should ensure an adequate airway and support breathing immediately after ROSC. Unconscious patients usually require an advanced airway for mechanical support of breathing. It may be necessary to replace a supraglottic airway used for initial resuscitation with an endotracheal tube, although the timing of replacement may vary. Methods for securing an advanced airway are discussed in Part 8.1: “Airway Management,” but several simple maneuvers deserve consideration. For example, rescuers and long-term hospital providers should avoid using ties that pass circumferentially around the patient's neck, potentially obstructing venous return from the brain. They should also elevate the head of the bed 30° if tolerated to reduce the incidence of cerebral edema, aspiration, and ventilatory-associated pneumonia. Correct placement of an advanced airway, particularly during patient transport, should be monitored using waveform capnography as described in other sections of the 2010 AHA Guidelines for CPR and ECC. Oxygenation of the patient should be monitored continuously with pulse oximetry.

Although 100% oxygen may have been used during initial resuscitation, providers should titrate inspired oxygen to the lowest level required to achieve an arterial oxygen saturation of ≥94%, so as to avoid potential oxygen toxicity. It is recognized that titration of inspired oxygen may not be possible immediately after out-of-hospital cardiac arrest until the patient is transported to the emergency department or, in the case of in-hospital arrest, the intensive care unit (ICU). Hyperventilation or “overbagging” the patient is common after cardiac arrest and should be avoided because of potential adverse hemodynamic effects. Hyperventilation increases intrathoracic pressure and inversely lowers cardiac output. The decrease in Paco2 seen with hyperventilation can also potentially decrease cerebral blood flow directly. Ventilation may be started at 10 to 12 breaths per minute and titrated to achieve a Petco2 of 35 to 40 mm Hg or a Paco2 of 40 to 45 mm Hg.

The clinician should assess vital signs and monitor for recurrent cardiac arrhythmias. Continuous electrocardiographic (ECG) monitoring should continue after ROSC, during transport, and throughout ICU care until stability has been achieved. Intravenous (IV) access should be obtained if not already established and the position and function of any intravenous catheter verified. IV lines should be promptly established to replace emergent intraosseous access achieved during resuscitation. If the patient is hypotensive (systolic blood pressure <90 mm Hg), fluid boluses can be considered. Cold fluid may be used if therapeutic hypothermia is elected. Vasoactive drug infusions such as dopamine, norepinephrine, or epinephrine may be initiated if necessary and titrated to achieve a minimum systolic blood pressure of ≥90 mm Hg or a mean arterial pressure of ≥65 mm Hg.

Brain injury and cardiovascular instability are the major determinants of survival after cardiac arrest.13 Because therapeutic hypothermia is the only intervention demonstrated to improve neurological recovery, it should be considered for any patient who is unable to follow verbal commands after ROSC. The patient should be transported to a facility that reliably provides this therapy in addition to coronary reperfusion (eg, PCI) and other goal-directed postarrest care therapies.

Overall the most common cause of cardiac arrest is cardiovascular disease and coronary ischemia.14,15 Therefore, a 12-lead ECG should be obtained as soon as possible to detect ST elevation or new or presumably new left bundle-branch block. When there is high suspicion of acute myocardial infarction (AMI), local protocols for treatment of AMI and coronary reperfusion should be activated. Even in the absence of ST elevation, medical or interventional treatments may be considered for treatment of ACS14,16,17 and should not be deferred in the presence of coma or in conjunction with hypothermia. Concurrent PCI and hypothermia are safe, with good outcomes reported for some comatose patients who undergo PCI.

Patients who are unconscious or unresponsive after cardiac arrest should be directed to an inpatient critical-care facility with a comprehensive care plan that includes acute cardiovascular interventions, use of therapeutic hypothermia, standardized medical goal-directed therapies, and advanced neurological monitoring and care. Neurological prognosis may be difficult to determine during the first 72 hours, even for patients who are not undergoing therapeutic hypothermia. This time frame for prognostication is likely to be extended in patients being cooled.18 Many initially comatose survivors of cardiac arrest have the potential for full recovery such that they are able to lead normal lives.1,2,19 Between 20% and 50% or more of survivors of out-of-hospital cardiac arrest who are comatose on arrival at the hospital may have good one-year neurological outcome.1,2,11 Therefore, it is important to place patients in a hospital critical-care unit where expert care and neurological evaluation can be performed and where appropriate testing to aid prognosis is available and performed in a timely manner.

Attention should be directed to treating the precipitating cause of cardiac arrest after ROSC. The provider should initiate or request studies that will further aid in evaluation of the patient. It is important to identify and treat any cardiac, electrolyte, toxicological, pulmonary, and neurological precipitants of arrest. The clinician may find it helpful to review the H's and T's mnemonic to recall factors that may contribute to cardiac arrest or complicate resuscitation or postresuscitation care: hypovolemia, hypoxia, hydrogen ion (acidosis of any etiology), hyper-/hypokalemia, moderate to severe hypothermia, toxins, tamponade (cardiac), tension pneumothorax, and thrombosis of the coronary or pulmonary vasculature. For further information on treating other causes of cardiac arrest, see Part 12: “Special Resuscitation Situations.”

Targeted Temperature Management

Induced Hypothermia

For protection of the brain and other organs, hypothermia is a helpful therapeutic approach in patients who remain comatose (usually defined as a lack of meaningful response to verbal commands) after ROSC. Questions remain about specific indications and populations, timing and duration of therapy, and methods for induction, maintenance, and subsequent reversal of hypothermia. One good randomized trial1 and a pseudorandomized trial2 reported improved neurologically intact survival to hospital discharge when comatose patients with out-of-hospital ventricular fibrillation (VF) cardiac arrest were cooled to 32°C to 34°C for 12 or 24 hours beginning minutes to hours after ROSC. Additional studies with historical control groups show improved neurological outcome after therapeutic hypothermia for comatose survivors of VF cardiac arrest.20,21

No randomized controlled trials have compared outcome between hypothermia and normothermia for non-VF arrest. However, 6 studies with historical control groups reported a beneficial effect on outcome from use of therapeutic hypothermia in comatose survivors of out-of-hospital cardiac arrest associated with any arrest rhythm.11,2226 Only one study with historical controls reported better neurological outcome after VF cardiac arrest but no difference in outcome after cardiac arrest associated with other rhythms.27 Two nonrandomized studies with concurrent controls28,29 indicate a possible benefit of hypothermia after in- and out-of-hospital cardiac arrest associated with non-VF initial rhythms.

Case series have reported the feasibility of using therapeutic hypothermia after ROSC in the setting of cardiogenic shock23,30,31 and therapeutic hypothermia in combination with emergent PCI.3236 Case series also report successful use of fibrinolytic therapy for AMI after ROSC,37,38 but data are lacking about interactions between fibrinolytics and hypothermia in this population.

The impact of the timing of initiating hypothermia after cardiac arrest is not completely understood. Studies of animal models of cardiac arrest showed that short-duration hypothermia (≤1 hour) achieved <10 to 20 minutes after ROSC had a beneficial effect that was lost when hypothermia was delayed.3941 Beyond the initial minutes of ROSC and when hypothermia is prolonged (>12 hours), the relationship between the onset of hypothermia and the resulting neuroprotection is less clear.42,43 Two prospective clinical trials in which hypothermia was achieved within 2 hours2 or at a median of 8 hours (interquartile range [IQR] 4 to 16 hours)1 after ROSC both demonstrated better outcomes in the hypothermia-treated than the normothermia-treated subjects. Subsequent to these studies, one registry-based case series of 986 comatose post–cardiac arrest patients35 suggested that time to initiation of cooling (IQR 1 to 1.8 hours) and time to achieving target temperature (IQR 3 to 6.7 hours) were not associated with improved neurological outcome after discharge. A case series of 49 consecutive comatose post–cardiac arrest patients44 cooled intravascularly after out-of-hospital cardiac arrest also documented that time to target temperature (median 6.8 hours [IQR 4.5 to 9.2 hours]) was not an independent predictor of neurological outcome.

The optimal duration of induced hypothermia is at least 12 hours and may be >24 hours. Hypothermia was maintained for 122 or 24 hours1 in the studies of out-of-hospital patients presenting in VF. Most case series of adult patients have reported 24 hours of hypothermia. The effect of a longer duration of cooling on outcome has not been studied in adults, but hypothermia for up to 72 hours was used safely in newborns.45,46

Although there are multiple methods for inducing hypothermia, no single method has proved to be optimal. Feedback-controlled endovascular catheters and surface cooling devices are available.4749 Other techniques (eg, cooling blankets and frequent application of ice bags) are readily available and effective but may require more labor and closer monitoring. As an adjunct, iced isotonic fluid can be infused to initiate core cooling but must be combined with a follow-up method for maintenance of hypothermia.5052 Although a theoretical concern is that rapid fluid loading could have adverse cardiopulmonary effects such as pulmonary edema, 9 case series indicate that cooling can be initiated safely with IV ice-cold fluids (500 mL to 30 mL/kg of saline 0.9% or Ringer's lactate).5159 One human case series56 showed that the deterioration in oxygenation that often occurs after ROSC was not significantly affected by the infusion of cold fluids (3427 mL ± 210 mL). Two randomized controlled trials,60,61 one study with concurrent controls,62 and 3 case series63,64 indicate that cooling with IV cold saline can be initiated safely in the prehospital setting.

Clinicians should continuously monitor the patient's core temperature using an esophageal thermometer, bladder catheter in nonanuric patients, or pulmonary artery catheter if one is placed for other indications.1,2 Axillary and oral temperatures are inadequate for measurement of core temperature changes, especially during active manipulation of temperature for therapeutic hypothermia,65,66 and true tympanic temperature probes are rarely available and often unreliable. Bladder temperatures in anuric patients and rectal temperatures may differ from brain or core temperature.66,67 A secondary source of temperature measurement should be considered, especially if a closed feedback cooling system is used for temperature management.

A number of potential complications are associated with cooling, including coagulopathy, arrhythmias, and hyperglycemia, particularly with an unintended drop below target temperature.35 The likelihood of pneumonia and sepsis may increase in patients treated with therapeutic hypothermia.1,2 Although these complications were not significantly different between groups in the published clinical trials, infections are common in this population, and prolonged hypothermia is known to decrease immune function. Hypothermia also impairs coagulation, and any ongoing bleeding should be controlled before decreasing temperature.

In summary, we recommend that comatose (ie, lack of meaningful response to verbal commands) adult patients with ROSC after out-of-hospital VF cardiac arrest should be cooled to 32°C to 34°C (89.6°F to 93.2°F) for 12 to 24 hours (Class I, LOE B). Induced hypothermia also may be considered for comatose adult patients with ROSC after in-hospital cardiac arrest of any initial rhythm or after out-of-hospital cardiac arrest with an initial rhythm of pulseless electric activity or asystole (Class IIb, LOE B). Active rewarming should be avoided in comatose patients who spontaneously develop a mild degree of hypothermia (>32°C [89.6°F]) after resuscitation from cardiac arrest during the first 48 hours after ROSC. (Class III, LOE C).

Hyperthermia

After resuscitation, temperature elevation above normal can impair brain recovery. The etiology of fever after cardiac arrest may be related to activation of inflammatory cytokines in a pattern similar to that observed in sepsis.68,69 There are no randomized controlled trials evaluating the effect of treating pyrexia with either frequent use of antipyretics or “controlled normothermia” using cooling techniques compared to no temperature intervention in post–cardiac arrest patients. Case series7074 and studies7580 suggest that there is an association between poor survival outcomes and pyrexia ≥37.6°C. In patients with a cerebrovascular event leading to brain ischemia, studies7580 demonstrate worsened short-term outcome and long-term mortality. By extrapolation this data may be relevant to the global ischemia and reperfusion of the brain that follows cardiac arrest. Patients can develop hyperthermia after rewarming posthypothermia treatment. This late hyperthermia should also be identified and treated. Providers should closely monitor patient core temperature after ROSC and actively intervene to avoid hyperthermia (Class I, LOE C).

Organ-Specific Evaluation and Support

The remainder of Part 9 focuses on organ-specific measures that should be included in the immediate post–cardiac arrest period.

Pulmonary System

Pulmonary dysfunction after cardiac arrest is common. Etiologies include hydrostatic pulmonary edema from left ventricular dysfunction; noncardiogenic edema from inflammatory, infective, or physical injuries; severe pulmonary atelectasis; or aspiration occurring during cardiac arrest or resuscitation. Patients often develop regional mismatch of ventilation and perfusion, contributing to decreased arterial oxygen content. The severity of pulmonary dysfunction often is measured in terms of the Pao2/Fio2 ratio. A Pao2/Fio2 ratio of ≤300 mm Hg usually defines acute lung injury. The acute onset of bilateral infiltrates on chest x-ray and a pulmonary artery pressure ≤18 mm Hg or no evidence of left atrial hypertension are common to both acute lung injury and acute respiratory distress syndrome (ARDS). A Pao2/Fio2 ratio <300 or <200 mm Hg separates acute lung injury from ARDS, respectively.81 Positive end-expiratory pressure (PEEP), a lung-protective strategy for mechanical ventilation, and titrated Fio2 are strategies that can improve pulmonary function and Pao2 while the practitioner is determining the pathophysiology of the pulmonary dysfunction.

Essential diagnostic tests in intubated patients include a chest radiograph and arterial blood gas measurements. Other diagnostic tests may be added based on history, physical examination, and clinical circumstances. Evaluation of a chest radiograph should verify the correct position of the endotracheal tube and the distribution of pulmonary infiltrates or edema and identify complications from chest compressions (eg, rib fracture, pneumothorax, and pleural effusions) or pneumonia.

Providers should adjust mechanical ventilatory support based on the measured oxyhemoglobin saturation, blood gas values, minute ventilation (respiratory rate and tidal volume), and patient-ventilator synchrony. In addition, mechanical ventilatory support to reduce the work of breathing should be considered as long as the patient remains in shock. As spontaneous ventilation becomes more efficient and as concurrent medical conditions allow, the level of support may be gradually decreased.

The optimal Fio2 during the immediate period after cardiac arrest is still debated. The beneficial effect of high Fio2 on systemic oxygen delivery should be balanced with the deleterious effect of generating oxygen-derived free radicals during the reperfusion phase. Animal data suggests that ventilations with 100% oxygen (generating Pao2 >350 mm Hg at 15 to 60 minutes after ROSC) increase brain lipid peroxidation, increase metabolic dysfunctions, increase neurological degeneration, and worsen short-term functional outcome when compared with ventilation with room air or an inspired oxygen fraction titrated to a pulse oximeter reading between 94% and 96%.8287 One randomized prospective clinical trial compared ventilation for the first 60 minutes after ROSC with 30% oxygen (resulting in Pao2=110±25 mm Hg at 60 minutes) or 100% oxygen (resulting in Pao2=345±174 mm Hg at 60 minutes).88 This small trial detected no difference in serial markers of acute brain injury, survival to hospital discharge, or percentage of patients with good neurological outcome at hospital discharge but was inadequately powered to detect important differences in survival or neurological outcome.

Once the circulation is restored, monitor systemic arterial oxyhemoglobin saturation. It may be reasonable, when the appropriate equipment is available, to titrate oxygen administration to maintain the arterial oxyhemoglobin saturation ≥94%. Provided appropriate equipment is available, once ROSC is achieved, adjust the Fio2 to the minimum concentration needed to achieve arterial oxyhemoglobin saturation ≥94%, with the goal of avoiding hyperoxia while ensuring adequate oxygen delivery. Since an arterial oxyhemoglobin saturation of 100% may correspond to a Pao2 anywhere between ∼80 and 500 mm Hg, in general it is appropriate to wean Fio2 when saturation is 100%, provided the oxyhemoglobin saturation can be maintained ≥94% (Class I, LOE C).

Because patients may have significant metabolic acidosis after cardiac arrest, there is a temptation to institute hyperventilation to normalize blood pH. However, metabolic acidosis is likely to be reversed once adequate perfusion is restored, and there are several physiological reasons why hyperventilation may be detrimental. Minute ventilation alters the partial pressure of carbon dioxide (Paco2), which in turn can affect cerebral blood flow. In a normal brain a 1-mm Hg decrease in Paco2 results in a decrease in cerebral blood flow of approximately 2.5% to 4%; cerebral blood flow remains CO2-reactive after cardiac arrest,89,90 although the magnitude of the CO2 reactivity (magnitude of change in cerebral blood flow per millimeters of mercury [mm Hg] change in Pco2) may be diminished or suppressed for 1 to 3 hours after reperfusion,91,92 especially after prolonged ischemia (≥15 minutes).93,94 After ROSC there is an initial hyperemic blood flow response that lasts 10 to 30 minutes, followed by a more prolonged period of low blood flow.95,96 During this latter period of late hypoperfusion, a mismatch between blood flow (as a component of oxygen delivery) and oxygen requirement may occur. Hyperventilation at this stage may lower Paco2, cause cerebral vasoconstriction, and exacerbate cerebral ischemic injury.

Physiological data in humans suggests that hyperventilation could cause additional cerebral ischemia in the post–cardiac arrest patient because sustained hypocapnia (low Pco2) may reduce cerebral blood flow.97,98 Transcranial Doppler measurements of the middle cerebral artery and jugular bulb oxygen saturation measurements in 10 comatose subjects after cardiac arrest revealed that hyperventilation with hypocapnia did not affect median flow velocity but did decrease jugular bulb oxygen saturation below the ischemic threshold (55%). Conversely, hypoventilation with hypercapnia produced the opposite effect.99 In one study, controlled ventilation with specific goals to keep Paco2 37.6 to 45.1 mm Hg (5 to 6 kPa) and Sao2 95% to 98% as part of a bundle with multiple other goals (including hypothermia and blood pressure goals) increased survival from 26% to 56%.11 In that study it was impossible to ascertain an independent effect of controlled ventilation separate from all other components of the bundle.

Hyperventilation also may compromise systemic blood flow because of occult or auto-PEEP and is deleterious in all low-flow states, including cardiopulmonary resuscitation (CPR)100,101 and hypovolemia.102,103 Auto-PEEP, also known as intrinsic PEEP or gas trapping, occurs preferentially in patients with obstructive lung disease and is aggravated by hyperventilation that does not allow sufficient time for complete exhalation. A gradual increase in end-expiratory volume and pressure in the lung (hyperinflation) is transmitted to the great veins in the thorax and depresses both venous return and cardiac output.104,105 Similar effects may occur after cardiac arrest, suggesting that hyperventilation should be avoided, especially in hypotensive patients.

Other ventilatory parameters may affect the outcome of patients on mechanical ventilation after cardiac arrest, particularly when acute lung injury or ARDS develops. Over the last decade attention has focused on low-volume/high-rate ventilation. In a comparison of high- and low-tidal-volume ventilation, the death rate of patients with ARDS was reduced from 40% to 31% in the group with reduced tidal volume (Vt).106 This and subsequent studies recommend ventilating patients to maintain Vt of 6 to 8 mL/kg predicted body weight and inspiratory plateau pressure ≤30 cm H2O to reduce ventilator-associated lung injury.107 Because low Vt ventilation (6 mL/kg) is associated with an increased incidence of atelectasis, PEEP and other lung “recruitment maneuver” procedures may be warranted.108 However, one study reported no difference in the rate of discharge or survival between ARDS patients receiving high- or low-PEEP regimens.109 Furthermore, a recent historical comparison of ventilation practice after cardiac arrest reported no differences in pneumonia, oxygenation, lung compliance, and ventilator days when a low Vt strategy versus a more liberal “old practice” Vt was applied.110

In conclusion, post–cardiac arrest patients are at risk of acute lung injury and ARDS, but refractory hypoxemia is not a frequent mode of death after cardiac arrest. There is no reason to recommend hyperventilation and “permissive hypercapnia” (hypoventilation) for these patients, and normocapnia should be considered the standard. There is also no data to recommend unique ventilation strategies in this population different from usual care of other mechanically ventilated patients at risk for acute lung injury and ARDS.

Routine hyperventilation with hypocapnia should be avoided after ROSC because it may worsen global brain ischemia by excessive cerebral vasoconstriction (Class III, LOE C). Hyperventilation or excessive tidal volumes resulting in increased intrathoracic pressure may also contribute to hemodynamic instability in certain patients. Ventilation rate and volume may be titrated to maintain high-normal Paco2 (40 to 45 mm Hg) or Petco2 (35 to 40 mm Hg) while avoiding hemodynamic compromise (Class IIb, LOE C).

Treatment of Pulmonary Embolism After CPR

Fibrinolytic use may benefit patients with massive pulmonary emboli who have not had CPR,111 and use of fibrinolytics to treat pulmonary embolism after CPR has been reported.112 The use of fibrinolytics during CPR has been studied, and CPR itself does not appear to pose an unacceptable risk of bleeding.113121 Alternatively, surgical embolectomy has also been used successfully in some patients after PE-induced cardiac arrest.117,122125 Mechanical thrombectomy was employed in a small case series and only one of seven patients died and pulmonary perfusion was restored in the majority (85.7%).115 In post–cardiac arrest patients with arrest due to presumed or known pulmonary embolism, fibrinolytics may be considered (Class IIb, LOE C).

Sedation After Cardiac Arrest

Patients with coma or respiratory dysfunction after ROSC are routinely intubated and maintained on mechanical ventilation for a period of time, which results in discomfort, pain, and anxiety. Intermittent or continuous sedation and/or analgesia can be used to achieve specific goals. Patients with post–cardiac arrest cognitive dysfunction may display agitation or frank delirium with purposeless movement and are at risk of self-injury. Opioids, anxiolytics, and sedative-hypnotic agents can be used in various combinations to improve patient-ventilator interaction and blunt the stress-related surge of endogenous catecholamines. Other agents with sedative and antipsychotic-tranquilizer properties, such as α2-adrenergic agonists,126 and butyrophenones127 are also used based on individual clinical circumstances.

If patient agitation is life-threatening, neuromuscular blocking agents can be used for short intervals with adequate sedation. Caution should be used in patients at high risk of seizures unless continuous electroencephalographic (EEG) monitoring is available. In general sedative agents should be administered cautiously with daily interruptions and titrated to the desired effect. A number of sedation scales128133 and motor activity scales134 were developed to titrate these pharmacological interventions to a clinical goal.

Shorter-acting medications that can be used as a single bolus or continuous infusion are usually preferred. There is little evidence to guide sedation/analgesia therapy immediately after ROSC. One observational study135 found an association between use of sedation and development of pneumonia in intubated patients during the first 48 hours of therapy. However, the study was not designed to investigate sedation as a risk factor for either pneumonia or death in patients with cardiac arrest.

Although minimizing sedation allows a better clinical estimate of neurological status, sedation, analgesia, and occasionally neuromuscular relaxation are routinely used to facilitate induced hypothermia and to control shivering. The duration of neuromuscular blocker use should be minimized and the depth of neuromuscular blockade should be monitored with a nerve twitch stimulator.

It is reasonable to consider the titrated use of sedation and analgesia in critically ill patients who require mechanical ventilation or shivering suppression during induced hypothermia after cardiac arrest (Class IIb, LOE C). Duration of neuromuscular blocking agents should be kept to a minimum or avoided altogether.

Cardiovascular System

ACS is a common cause of cardiac arrest.14,16,34,35,134147 The clinician should evaluate the patient's 12-lead ECG and cardiac markers after ROSC. A 12-lead ECG should be obtained as soon as possible after ROSC to determine whether acute ST elevation is present (Class I, LOE B). Because it is impossible to determine the final neurological status of comatose patients in the first hours after ROSC, aggressive treatment of ST-elevation myocardial infarction (STEMI) should begin as in non–cardiac arrest patients, regardless of coma or induced hypothermia. Because of the high incidence of acute coronary ischemia, consideration of emergent coronary angiography may be reasonable even in the absence of STEMI.14,16 Notably, PCI, alone or as part of a bundle of care, is associated with improved myocardial function14 and neurological outcomes.11,16 Therapeutic hypothermia can be safely combined with primary PCI after cardiac arrest caused by AMI.11,31,3335 Other details of ACS treatment are discussed in Part 10.

Patients with cardiac arrest may receive antiarrhythmic drugs such as lidocaine or amiodarone during initial resuscitation. There is no evidence to support or refute continued or prophylactic administration of these medications.7,148152

Vasoactive Drugs for Use in Post–Cardiac Arrest Patients

Vasopressors

Vasoactive drugs may be administered after ROSC to support cardiac output, especially blood flow to the heart and brain. Drugs may be selected to improve heart rate (chronotropic effects), myocardial contractility (inotropic effects), or arterial pressure (vasoconstrictive effects), or to reduce afterload (vasodilator effects). Unfortunately many adrenergic drugs are not selective and may increase or decrease heart rate and afterload, increase cardiac arrhythmias, and increase myocardial ischemia by creating a mismatch between myocardial oxygen demand and delivery. Myocardial ischemia, in turn, may further decrease heart function. Some agents may also have metabolic effects that increase blood glucose, lactate, and metabolic rate. There is a paucity of data about which vasoactive drug to select first, although providers should become familiar with the differing adverse effects associated with these drugs, which might make a particular agent more or less appropriate for a specific patient.153

Specific drug infusion rates cannot be recommended because of variations in pharmacokinetics (relation between drug dose and concentration) and pharmacodynamics (relation between drug concentration and effect) in critically ill patients,154,155 so commonly used initial dose ranges are listed in Table 2. Vasoactive drugs must be titrated at the bedside to secure the intended effect while limiting side effects. Providers must also be aware of the concentrations delivered and compatibilities with previously and concurrently administered drugs.

Table 2. Common Vasoactive Drugs

Drug Typical Starting Dose (Then Titrate to Effect)
Epinephrine 0.1–0.5 mcg/kg/min (In 70-kg adult, 7–35 mcg/min)
  • Useful for symptomatic bradycardia if atropine and transcutaneous pacing fail or if pacing is not available

  • Used to treat severe hypotension (eg, systolic blood pressure <70 mm Hg)

  • Useful for anaphylaxis associated with hemodynamic instability or respiratory distress158

Norepinephrine 0.1–0.5 mcg/kg/min (In 70-kg adult, 7–35 mcg/min)
  • Used to treat severe hypotension (eg, systolic blood pressure <70 mm Hg) and a low total peripheral resistance

  • Relatively contraindicated in patients with hypovolemia. It may increase myocardial oxygen requirements, mandating cautious use in patients with ischemic heart disease

  • Usually induces renal and mesenteric vasoconstriction; in sepsis, however, norepinephrine improves renal blood flow and urine output159,160

Phenylephrine 0.5–2.0 mcg/kg/min (In 70-kg adult, 35–140 mcg/min)
  • Used to treat severe hypotension (eg, systolic blood pressure <70 mm Hg) and a low total peripheral resistance

Dopamine 5–10 mcg/kg/min
  • Used to treat hypotension, especially if it is associated with symptomatic bradycardia

  • Although low-dose dopamine infusion has frequently been recommended to maintain renal blood flow or improve renal function, more recent data have failed to show a beneficial effect from such therapy161,162

Dobutamine 5–10 mcg/kg/min
  • The (+) isomer is a potent beta-adrenergic agonist, whereas the (–) isomer is a potent alpha-1-agonist163

  • The vasodilating beta2-adrenergic effects of the (+) isomer counterbalance the vasoconstricting alpha-adrenergic effects, often leading to little change or a reduction in systemic vascular resistance

Milrinone Load 50 mcg/kg over 10 minutes then infuse at 0.375 mcg/kg/min
  • Used to treat low cardiac output

  • May cause less tachycardia than dobutamine

In general, adrenergic drugs should not be mixed with sodium bicarbonate or other alkaline solutions in the IV line because there is evidence that adrenergic agents are inactivated in alkaline solutions.156,157 Norepinephrine (levarterenol) and other catecholamines that activate α-adrenergic receptors may produce tissue necrosis if extravasation occurs. Therefore, administration through a central line is preferred whenever possible. If extravasation develops, infiltrate 5 to 10 mg of phentolamine diluted in 10 to 15 mL of saline into the site of extravasation as soon as possible to prevent tissue death and sloughing.

Use of Vasoactive Drugs After Cardiac Arrest

Hemodynamic instability is common after cardiac arrest.6 Death due to multiorgan failure is associated with a persistently low cardiac index during the first 24 hours after resuscitation.6,164 Vasodilation may occur from loss of sympathetic tone and from metabolic acidosis. In addition, the ischemia/reperfusion of cardiac arrest and electric defibrillation both can cause transient myocardial stunning and dysfunction165 that can last many hours but may improve with use of vasoactive drugs.158 Echocardiographic evaluation within the first 24 hours after arrest is a useful way to assess myocardial function in order to guide ongoing management.14,17

There is no proven benefit or harm associated with administration of routine IV fluids or vasoactive drugs (pressor and inotropic agents) to patients experiencing myocardial dysfunction after ROSC. Although some studies found improved outcome associated with these therapies, the outcome could not be solely ascribed to these specific interventions because they were only one component of standardized treatment protocols (eg, PCI and therapeutic hypothermia).6,11,12,166 Invasive monitoring may be necessary to measure hemodynamic parameters accurately and to determine the most appropriate combination of medications to optimize perfusion.

Fluid administration as well as vasoactive (eg, norepinephrine), inotropic (eg, dobutamine), and inodilator (eg, milrinone) agents should be titrated as needed to optimize blood pressure, cardiac output, and systemic perfusion (Class I, LOE B). Although human studies have not established ideal targets for blood pressure or blood oxygenation,11,12 a mean arterial pressure ≥65 mm Hg and an ScvO2 ≥70% are generally considered reasonable goals.

Although mechanical circulatory support improves hemodynamics in patients not experiencing cardiac arrest,167171 it has not been associated with improved clinical outcome and routine use of mechanical circulatory support after cardiac arrest is not recommended.

Modifying Outcomes From Critical Illness

Cardiac arrest is thought to involve multiorgan ischemic injury and microcirculatory dysfunction.68,69,172 Implementing a protocol for goal-directed therapy using fluid and vasoactive drug administration along with monitoring of central venous oxygen saturation may improve survival from sepsis,173 suggesting that a similar approach may benefit post–cardiac arrest patients. By analogy, studies have explored several other interventions believed to be beneficial in sepsis or other critical illness.

Glucose Control

The post–cardiac arrest patient is likely to develop metabolic abnormalities such as hyperglycemia that may be detrimental. Evidence from several retrospective studies7,73,174176 suggests an association of higher glucose levels with increased mortality or worse neurological outcomes. Variable ranges for optimum glucose values were suggested, and the studies do not provide evidence that an interventional strategy to manage glucose levels will alter outcomes. Only one study examined patients with induced hypothermia.175

The optimum blood glucose concentration and interventional strategy to manage blood glucose in the post–cardiac arrest period is unknown. A consistent finding in clinical trials of glucose control177185 is that intensive therapy leads to more frequent episodes of severe hypoglycemia (usually defined as blood glucose level ≤40 mg/dL [2.2 mmol/L]). Hypoglycemia may be associated with worse outcomes in critically ill patients.186,187

Strategies to target moderate glycemic control (144 to 180 mg/dL [8 to 10 mmol/L]) may be considered in adult patients with ROSC after cardiac arrest (Class IIb, LOE B). Attempts to control glucose concentration within a lower range (80 to 110 mg/dL [4.4 to 6.1 mmol/L]) should not be implemented after cardiac arrest due to the increased risk of hypoglycemia (Class III, LOE B).

Steroids

Corticosteroids have an essential role in the physiological response to severe stress, including maintenance of vascular tone and capillary permeability. In the post–cardiac arrest phase, several authors report a relative adrenal insufficiency compared with the metabolic demands of the body.188,189 Relative adrenal insufficiency in the post–cardiac arrest phase was associated with higher rates of mortality.188190

At present there are no human randomized trials investigating corticosteroid use after ROSC. One investigation combined steroid therapy with use of vasopressin, which made interpretation of results specific to steroids impossible.191 The post–cardiac arrest syndrome has similarities to septic shock, but the efficacy of corticosteroids remains controversial in patients with sepsis as well.68,192194 Whether the provision of corticosteroids in the post–cardiac arrest phase improves outcome remains unknown and the value of the routine use of corticosteroids for patients with ROSC following cardiac arrest is uncertain.

Hemofiltration

Hemofiltration has been proposed as a method to modify the humoral response to the ischemic-reperfusion injury that occurs after cardiac arrest. In a single randomized controlled trial there was no significant difference in 6-month survival among the groups.195 Future investigations are required to determine whether hemofiltration will improve outcome in post–cardiac arrest patients.

Central Nervous System

Brain injury is a common cause of morbidity and mortality in post–cardiac arrest patients. Brain injury is the cause of death in 68% of patients after out-of-hospital cardiac arrest and in 23% after in-hospital cardiac arrest.13 The pathophysiology of post–cardiac arrest brain injury involves a complex cascade of molecular events that are triggered by ischemia and reperfusion and then executed over hours to days after ROSC. Events and conditions in the post–cardiac arrest period have the potential to exacerbate or attenuate these injury pathways and impact ultimate outcomes. Clinical manifestations of post–cardiac arrest brain injury include coma, seizures, myoclonus, various degrees of neurocognitive dysfunction (ranging from memory deficits to persistent vegetative state), and brain death.3

Seizure Management

Whether there is any disease-specific management of seizures after cardiac arrest remains unknown and the true incidence of post–cardiac arrest electrographic seizures may be higher as the clinical diagnosis of seizures may not be readily apparent. It is accepted in other settings that prolonged, untreated seizures are detrimental to the brain, and seizures are common after ROSC, occurring in 5% to 20% of comatose cardiac arrest survivors with or without therapeutic hypothermia.11,196,197

An EEG for the diagnosis of seizure should be performed with prompt interpretation as soon as possible and should be monitored frequently or continuously in comatose patients after ROSC (Class I, LOE C). More clinical data are needed to define the diagnosis and management of seizures after cardiac arrest. Neuroprotective agents with anticonvulsant properties such as thiopental196 and single-dose diazepam or magnesium or both198 given after ROSC have not improved neurological outcome in survivors. No studies have addressed whether anticonvulsant therapy improves outcome after cardiac arrest, and several studies demonstrated that post–cardiac arrest seizures were refractory to traditional anticonvulsant agents.199201 The same anticonvulsant regimens for the treatment of seizures used for status epilepticus caused by other etiologies may be considered after cardiac arrest. (Class IIb, LOE C).

Neuroprotective Drugs

The molecular events that cause neurodegeneration after cardiac arrest occur over hours to days after ROSC. This time course suggests a potentially broad therapeutic window for neuroprotective drug therapy. However, the number of clinical trials performed to date is limited and has failed to demonstrate improved neurological outcome with potential neuroprotective drugs given after cardiac arrest.

Few neuroprotective drugs have been tested in clinical trials, and only one published randomized trial202 was performed in which a neuroprotective drug was combined with therapeutic hypothermia. No neuroprotection benefit was observed when patients (without hypothermia) were treated with thiopental, glucocorticoids, nimodipine, lidoflazine, diazepam, and magnesium sulfate. One trial using coenzyme Q10 in patients receiving hypothermia failed to show improved survival with good neurological outcome.202 The routine use of coenzyme Q10 in patients treated with hypothermia is uncertain (Class IIb, LOE B).

Prognostication of Neurological Outcome in Comatose Cardiac Arrest Survivors

The goal of post–cardiac arrest management is to return patients to their prearrest functional level. However, many patients will die, remain permanently unresponsive, or remain permanently unable to perform independent activities. Early prognostication of neurological outcome is an essential component of post–cardiac arrest care. Most importantly, when decisions to limit or withdraw life-sustaining care are being considered, tools used to prognosticate poor outcome must be accurate and reliable with a false-positive rate (FPR) approaching 0%. Poor outcome is defined as death, persistent unresponsiveness, or the inability to undertake independent activities after 6 months.203 No prearrest or intra-arrest parameters (including arrest duration, bystander CPR, or presenting rhythm) alone or in combination accurately predict outcome in individual patients who achieve ROSC.

A thorough neurological evaluation is needed to obtain accurate prognostic findings. No postarrest physical examination finding or diagnostic study has as yet predicted poor outcome of comatose cardiac arrest survivors during the first 24 hours after ROSC. After 24 hours somatosensory evoked potentials (SSEPs) and select physical examination findings at specific time points after ROSC in the absence of confounders (such as hypotension, seizures, sedatives, or neuromuscular blockers) are the most reliable early predictors of poor outcome in patients not undergoing therapeutic hypothermia. However, the decision to limit care should never be made on the basis of a single prognostic parameter, and expert consultation may be needed.

Neurological Assessment

The neurological examination is the most widely studied parameter to predict outcome in comatose post–cardiac arrest patients. Prognostication of functional outcome has not been established in noncomatose patients. Neurological examination for this purpose can be reliably undertaken only in the absence of confounding factors (hypotension, seizures, sedatives, or neuromuscular blockers). On the basis of existing studies, no clinical neurological signs reliably predict poor outcome <24 hours after cardiac arrest.204,205 Among adult patients who are comatose and have not been treated with hypothermia, the absence of both pupillary light and corneal reflexes at ≥72 hours after cardiac arrest predicted poor outcome with high reliability.204 The absence of vestibulo-ocular reflexes at ≥24 hours (FPR 0%, 95% CI 0% to 14%)205,206 or Glasgow Coma Scale (GCS) score <5 at ≥72 hours (FPR 0%, 95% CI 0% to 6%)204,207,208 are less reliable for predicting poor outcome or were studied only in limited numbers of patients. Other clinical signs, including myoclonus,209213 are not recommended for predicting poor outcome (Class III, LOE C).

EEG

No electrophysiological study reliably predicts outcome in comatose patients during the first 24 hours after ROSC. In normothermic patients without significant confounders (sedatives, hypotension, hypothermia, neuromuscular blockade, or hypoxemia), an EEG pattern showing generalized suppression to <20 μV, burst-suppression pattern associated with generalized epileptic activity, or diffuse periodic complexes on a flat background is associated with a poor outcome (FPR 3%, 95% CI 0.9% to 11%).203 One week after the initial arrest event, specific EEG findings may be useful for predicting poor outcomes in comatose cardiac arrest survivors,161,203,204,206,214221 The prognostic accuracy of malignant EEG patterns appears to be less reliable in patients treated with hypothermia. Status epilepticus in post-ROSC patients treated with hypothermia has an FPR of 7% (95% CI 1% to 25%) to 11.5% (95% CI 3% to 31%) for predicting poor outcome.218,222

In the absence of confounding factors such as sedatives, hypotension, hypothermia, neuromuscular blockade, seizures, or hypoxemia, it may be helpful to use an unprocessed EEG interpretation observed ≥24 hours after ROSC to assist with the prediction of a poor outcome in comatose survivors of cardiac arrest not treated with hypothermia (Class IIb, LOE B).

Evoked Potentials

Abnormalities in evoked potentials are associated with poor outcomes. Bilateral absence of the N20 cortical response to median nerve SSEPs predicts poor outcome (FPR 0%, 95% CI 0% to 3%).161,203 Although other evoked potential measurements (for example, Brain stem Auditory Evoked Potentials) have been associated with poor outcomes in comatose cardiac arrest survivors, they are either less reliable predictors of poor outcome than SSEPs or have not been studied in enough patients to establish their reliability. Bilateral absence of the N20 cortical response to median nerve stimulation after 24 hours predicts poor outcome in comatose cardiac arrest survivors not treated with therapeutic hypothermia (Class IIa, LOE A).

The impact of therapeutic hypothermia on the prognostic accuracy of SSEPs has not been adequately studied.

Neuroimaging

The most studied neuroimaging modalities are magnetic resonance imaging (MRI) and computed tomography (CT) of the brain. Extensive cortical and subcortical lesions on MRI are associated with poor neurological outcome.223253 These studies varied widely in the MRI parameters used, sample size, and interval after arrest when testing occurred.

CT imaging to detect brain injury and predict functional outcome is supported by several studies.233,244,245,247,248,254267 The timing of CT in these studies varied widely. CT parameters associated with poor outcome were varied and included quantitative measure of gray matter:white matter Hounsfield unit ratio and qualitative description of brain structures. A nonenhanced CT scan can also provide information about structural lesions, stroke, or intracranial hemorrhage that may have contributed to cardiac arrest.268,269

Other less utilized and investigated neuroimaging modalities have included single-photon emission computed tomography,253,267,270 cerebral angiography244 and transcranial Doppler240 A nuclear imaging study observed that abnormal tracer uptake in the cerebral cortices was associated with poor outcome in one case report.248

Despite tremendous potential, neuroimaging has yet to be proved as an independently accurate modality for prediction of outcome in individual comatose survivors of cardiac arrest and specific neuroimaging modalities cannot be recommended for predicting poor outcome after cardiac arrest.

Blood and Cerebrospinal Fluid Biomarkers

There has been extensive clinical research exploring biomarkers in the blood (plasma or serum) and cerebrospinal fluid (CSF) as early predictors of poor outcome in comatose cardiac arrest survivors. Biomarkers that are predictive of neurological outcome are typically released from dying neurons or glial cells in the brain (eg, neuron-specific enolase [NSE], S100B, GFAP, CK-BB) and can be measured in the blood or CSF. The primary advantage of biomarkers is that levels are unlikely to be confounded by sedation or neuromuscular blockade, which are commonly used in the first few days after cardiac arrest. However, for most biomarkers, only an association with outcome has been reported. When using a cutoff value that results in an FPR of 0% for predicting poor outcome, the 95% CI is unacceptably high due to the small number of patients studied.

The most promising and extensively studied biomarker is serum NSE, which has been reported to have a 0% FPR (95% CI 0% to 3%) for predicting poor outcome when measured between 24 and 72 hours after cardiac arrest.203,204 Other guidelines have recommended the use of serum NSE to predict poor outcome in patients after ROSC.203 However, the primary limitation of serum NSE is the variability among studies in both the assays used and the cutoff value that results in an FPR of 0% for predicting poor outcome.271282 Furthermore, interventions such as therapeutic hypothermia appear to variably alter the NSE cutoff value that is predictive of poor outcome.283285 Finally a number of clinical disorders, such as abdominal organ injury, have been associated with elevated NSE levels independent of cardiac arrest.286

The routine use of any serum or CSF biomarker as a sole predictor of poor outcome in comatose patients after cardiac arrest is not recommended (Class III, LOE B).

Changes in Prognostication With Hypothermia

There is a paucity of data about the utility of physical examination, EEG, and evoked potentials in patients who have been treated with induced hypothermia. Physical examination (motor response, pupillary light and corneal reflexes), EEG, SSEP, and imaging studies are less reliable for predicting poor outcome in patients treated with hypothermia. Durations of observation greater than 72 hours after ROSC should be considered before predicting poor outcome in patients treated with hypothermia (Class I, Level C).

Organ Donation After Cardiac Arrest

Despite maximal support and adequate observation, some patients will be brain-dead after cardiac arrest. Studies suggest that there is no difference in functional outcomes of organs transplanted from patients who are brain-dead as a consequence of cardiac arrest when compared with donors who are brain-dead due to other causes.287290 Adult patients who progress to brain death after resuscitation from cardiac arrest should be considered for organ donation (Class I, LOE B).

Summary

The goal of immediate post–cardiac arrest care is to optimize systemic perfusion, restore metabolic homeostasis, and support organ system function to increase the likelihood of intact neurological survival. The post–cardiac arrest period is often marked by hemodynamic instability as well as metabolic abnormalities. Support and treatment of acute myocardial dysfunction and acute myocardial ischemia can increase the probability of survival. Interventions to reduce secondary brain injury, such as therapeutic hypothermia, can improve survival and neurological recovery. Every organ system is at risk during this period, and patients are at risk of developing multiorgan dysfunction.

The comprehensive treatment of diverse problems after cardiac arrest involves multidisciplinary aspects of critical care, cardiology, and neurology. For this reason, it is important to admit patients to appropriate critical-care units with a prospective plan of care to anticipate, monitor, and treat each of these diverse problems. It is also important to appreciate the relative strengths and weaknesses of different tools for estimating the prognosis of patients after cardiac arrest.

Disclosures

Guidelines Part 9: Post–Cardiac Arrest Care: Writing Group Disclosures

Writing Group Member Employment Research Grant Other Research Support Speakers' Bureau/Honoraria Ownership Interest Consultant/Advisory Board Other
Mary Ann Peberdy Virginia Commonwealth University–Professor of Medicine & Emergency Medicine None None None None None None
Clifton W. Callaway University of Pittsburgh School of Medicine–Associate Professor; UPMC Health System–Physician Grants to University of Pittsburgh: NHLBI-Resuscitation Outcomes Consortium HRSA-Development and Dissemination of Program Tools for Uncontrolled Donation After Cardiac Death (UDCD) *Loan of an Arctic Sun cooling device (without disposables) to human physiology laboratory for experiments on hypothermia by Medivance, Inc. None Co-inventor on patent about ventricular fibrillation waveform analysis, licensed by University of Pittsburgh to Medtronic ERS, Inc. None None
Robert W. Neumar University of Pennsylvania–Associate Professor of Emergency Medicine Funding Source: NIH/NINDS Grant Number: R21 NS054654 Funding Period 06/01/07 to 06/31/2010 Role on Project: Principal Investigator Title: Optimizing Therapeutic Hypothermia After Cardiac Arrest Description: The goal of this project is to evaluate how the onset and duration of therapeutic hypothermia after cardiac arrest impacts survival and neuroprotection None None None None None
Romergryko G. Geocadin Johns Hopkins University School of Medicine–Associate Professor of Neurology, Anesthesiology-Critical Care Medicine and Neurosurgery NIH RO1 Grant: “Consequence of Cardiac Arrest: Brain Injury” *NIH R44 Grant: “Cortical Injury Monitor Phase IIB” None *Academic Grand Rounds American Academy of Neurology None None *Guest Editor: Neurology Clinics, Emergency Medicine Clinics, and Seminars in Neurology
Janice L. Zimmerman The Methodist Hospital Physician Organization–Head, Critical Care Division and Director, MICU None None *Society of Critical Care Medicine American College of Chest Physicians Center for Biomedical Communications None None *Callaway and Associates Andrews and Kurth
Michael Donnino Harvard Medical Faculty Physicians–Physician Corticosteroids in Post-cardiac Arrest Patients [Scientist Development Grant, American Heart Association] Thiamine as a Metabolic Resuscitator in Septic Shock [Pending] *Thiamine Deficiency in Septic Shock [completed, NIH through Harvard Medical School] Statin Therapy in Sepsis [Eleanor Shores Grant-nonindustry] None None None None None
Andrea Gabrielli University of Florida–Professor of Anesthesiology and Surgery NIH-Biomarkers and Traumatic Brain Injury None None None None None
Scott M. Silvers Mayo Clinic Florida–Chair, Department of Emergency Medicine None None None None None None
Arno L. Zaritsky Children's Hospital of the King's Daughters–Sr. VP for Clinical Services None None None None *Data Safety Monitoring Board for NIH-sponsored clinical trial of therapeutic hypothermia after pediatric cardiac arrest None
Raina Merchant University of Pennsylvania–Research fellow None None None None None None
Terry L. Vanden Hoek University of Chicago–Associate Professor *Principal Investigator 09/06/04–04/30/10 DOD/Office of Naval Research $885,639 Proteomic Development of Molecular Vital Signs: Mapping a Mitochondrial Injury Severity Score to Triage and Guide Resuscitation of Hemorrhagic Shock. This research grant is awarded to the University of Chicago None None *Hypothermia Induction Patents (3 approved, 3 pending) no income None None
Steven L. Kronick University of Michigan Health System Healthcare institution Assistant Professor None None None None None *Expert Witness: Reviewed a single case for an attorney. Less than 4 hours work total

This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person's gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.

*Modest.

Significant.

Footnotes

The American Heart Association requests that this document be cited as follows: Peberdy MA, Callaway CW, Neumar RW, Geocadin RG, Zimmerman JL, Donnino M, Gabrielli A, Silvers SM, Zaritsky AL, Merchant R, Vanden Hoek TL, Kronick SL. Part 9: post–cardiac arrest care: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation. 2010;122(suppl 3):S768–S786.

*Co-chairs and equal first co-authors.

References

  • 1. HACA. Hypothermia After Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med. 2002; 346:549–556.CrossrefMedlineGoogle Scholar
  • 2. Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, Smith K . Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med. 2002; 346:557–563.CrossrefMedlineGoogle Scholar
  • 3. Neumar RW, Nolan JP, Adrie C, Aibiki M, Berg RA, Bottiger BW, Callaway C, Clark RS, Geocadin RG, Jauch EC, Kern KB, Laurent I, Longstreth WT, Merchant RM, Morley P, Morrison LJ, Nadkarni V, Peberdy MA, Rivers EP, Rodriguez-Nunez A, Sellke FW, Spaulding C, Sunde K, Vanden Hoek T . Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication. A consensus statement from the International Liaison Committee on Resuscitation (American Heart Association, Australian and New Zealand Council on Resuscitation, European Resuscitation Council, Heart and Stroke Foundation of Canada, InterAmerican Heart Foundation, Resuscitation Council of Asia, and the Resuscitation Council of Southern Africa); the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; and the Stroke Council. Circulation. 2008; 118:2452–2483.LinkGoogle Scholar
  • 4. Safar P . Resuscitation from clinical death: Pathophysiologic limits and therapeutic potentials. Crit Care Med. 1988; 16:923–941.CrossrefMedlineGoogle Scholar
  • 5. Negovsky VA . The second step in resuscitation–the treatment of the ‘post-resuscitation disease.’Resuscitation. 1972; 1:1–7.CrossrefMedlineGoogle Scholar
  • 6. Laurent I, Monchi M, Chiche JD, Joly LM, Spaulding C, Bourgeois B, Cariou A, Rozenberg A, Carli P, Weber S, Dhainaut JF . Reversible myocardial dysfunction in survivors of out-of-hospital cardiac arrest. J Am Coll Cardiol. 2002; 40:2110–2116.CrossrefMedlineGoogle Scholar
  • 7. Skrifvars MB, Pettila V, Rosenberg PH, Castren M . A multiple logistic regression analysis of in-hospital factors related to survival at six months in patients resuscitated from out-of-hospital ventricular fibrillation. Resuscitation. 2003; 59:319–328.CrossrefMedlineGoogle Scholar
  • 8. Carr BG, Kahn JM, Merchant RM, Kramer AA, Neumar RW . Inter-hospital variability in post-cardiac arrest mortality. Resuscitation. 2009; 80:30–34.CrossrefMedlineGoogle Scholar
  • 9. Callaway CW, Schmicker R, Kampmeyer M, Powell J, Rea TD, Daya MR, Aufderheide TP, Davis DP, Rittenberger JC, Idris AH . Nichol G Receiving hospital characteristics associated with survival after out-of-hospital cardiac arrest. Resuscitation; 2010; 81:524–529.CrossrefMedlineGoogle Scholar
  • 10. Kirves H, Skrifvars MB, Vahakuopus M, Ekstrom K, Martikainen M, Castren M . Adherence to resuscitation guidelines during prehospital care of cardiac arrest patients. Eur J Emerg Med. 2007; 14:75–81.CrossrefMedlineGoogle Scholar
  • 11. Sunde K, Pytte M, Jacobsen D, Mangschau A, Jensen LP, Smedsrud C, Draegni T, Steen PA . Implementation of a standardised treatment protocol for post resuscitation care after out-of-hospital cardiac arrest. Resuscitation. 2007; 73:29–39.CrossrefMedlineGoogle Scholar
  • 12. Gaieski DF, Band RA, Abella BS, Neumar RW, Fuchs BD, Kolansky DM, Merchant RM, Carr BG, Becker LB, Maguire C, Klair A, Hylton J, Goyal M . Early goal-directed hemodynamic optimization combined with therapeutic hypothermia in comatose survivors of out-of-hospital cardiac arrest. Resuscitation. 2009; 80:418–424.CrossrefMedlineGoogle Scholar
  • 13. Laver S, Farrow C, Turner D, Nolan J . Mode of death after admission to an intensive care unit following cardiac arrest. Intensive Care Med. 2004; 30:2126–2128.CrossrefMedlineGoogle Scholar
  • 14. Spaulding CM, Joly LM, Rosenberg A, Monchi M, Weber SN, Dhainaut JF, Carli P . Immediate coronary angiography in survivors of out-of-hospital cardiac arrest. N Engl J Med. 1997; 336:1629–1633.CrossrefMedlineGoogle Scholar
  • 15. Anyfantakis ZA, Baron G, Aubry P, Himbert D, Feldman LJ, Juliard JM, Ricard-Hibon A, Burnod A, Cokkinos DV, Steg PG . Acute coronary angiographic findings in survivors of out-of-hospital cardiac arrest. Am Heart J. 2009; 157:312–318.CrossrefMedlineGoogle Scholar
  • 16. Reynolds JC, Callaway CW, El Khoudary SR, Moore CG, Alvarez RJ, Rittenberger JC . Coronary angiography predicts improved outcome following cardiac arrest: propensity-adjusted analysis. J Intensive Care Med. 2009; 24:179–186.CrossrefMedlineGoogle Scholar
  • 17. Dumas F, Cariou A, Manzo-Silberman S, Grimaldi D, Vivien B, Rosencher J, Empana J-P, Carli P, Mira J-P, Jouven X, Spaulding C . Immediate percutaneous coronary intervention is associated with better survival after out-of-hospital cardiac arrest: insights from the PROCAT (Parisian Region Out of Hospital Cardiac Arrest) Registry. Circ Cardiovasc Interv. 2010; 3:200–207.LinkGoogle Scholar
  • 18. Booth CM, Boone RH, Tomlinson G, Detsky AS . Is this patient dead, vegetative, or severely neurologically impaired? Assessing outcome for comatose survivors of cardiac arrest. JAMA. 2004; 291:870–879.CrossrefMedlineGoogle Scholar
  • 19. Bunch TJ, White RD, Gersh BJ, Meverden RA, Hodge DO, Ballman KV, Hammill SC, Shen WK, Packer DL . Long-term outcomes of out-of-hospital cardiac arrest after successful early defibrillation. N Engl J Med. 2003; 348:2626–2633.CrossrefMedlineGoogle Scholar
  • 20. Belliard G, Catez E, Charron C, Caille V, Aegerter P, Dubourg O, Jardin F, Vieillard-Baron A . Efficacy of therapeutic hypothermia after out-of-hospital cardiac arrest due to ventricular fibrillation. Resuscitation. 2007; 75:252–259.CrossrefMedlineGoogle Scholar
  • 21. Castrejon S, Cortes M, Salto ML, Benittez LC, Rubio R, Juarez M, Lopez de Sa E, Bueno H, Sanchez PL, Fernandez Aviles F . Improved prognosis after using mild hypothermia to treat cardiorespiratory arrest due to a cardiac cause: comparison with a control group. Rev Esp Cardiol. 2009; 62:733–741.MedlineGoogle Scholar
  • 22. Bernard SA, Jones BM, Horne MK . Clinical trial of induced hypothermia in comatose survivors of out-of-hospital cardiac arrest. Ann Emerg Med. 1997; 30:146–153.CrossrefMedlineGoogle Scholar
  • 23. Oddo M, Schaller MD, Feihl F, Ribordy V, Liaudet L . From evidence to clinical practice: effective implementation of therapeutic hypothermia to improve patient outcome after cardiac arrest. Crit Care Med. 2006; 34:1865–1873.CrossrefMedlineGoogle Scholar
  • 24. Busch M, Soreide E, Lossius HM, Lexow K, Dickstein K . Rapid implementation of therapeutic hypothermia in comatose out-of-hospital cardiac arrest survivors. Acta Anaesthesiol Scand. 2006; 50:1277–1283.CrossrefMedlineGoogle Scholar
  • 25. Storm C, Steffen I, Schefold JC, Krueger A, Oppert M, Jorres A, Hasper D . Mild therapeutic hypothermia shortens intensive care unit stay of survivors after out-of-hospital cardiac arrest compared to historical controls. Crit Care. 2008; 12:R78.CrossrefMedlineGoogle Scholar
  • 26. Don CW, Longstreth WT, Maynard C, Olsufka M, Nichol G, Ray T, Kupchik N, Deem S, Copass MK, Cobb LA, Kim F . Active surface cooling protocol to induce mild therapeutic hypothermia after out-of-hospital cardiac arrest: a retrospective before-and-after comparison in a single hospital. Crit Care Med. 2009; 37:3062–3069.CrossrefMedlineGoogle Scholar
  • 27. Bro-Jeppesen J, Kjaergaard J, Horsted TI, Wanscher MC, Nielsen SL, Rasmussen LS, Hassager C . The impact of therapeutic hypothermia on neurological function and quality of life after cardiac arrest. Resuscitation. 2009; 80:171–176.CrossrefMedlineGoogle Scholar
  • 28. Arrich J . Clinical application of mild therapeutic hypothermia after cardiac arrest. Crit Care Med. 2007; 35:1041–1047.CrossrefMedlineGoogle Scholar
  • 29. Holzer M, Mullner M, Sterz F, Robak O, Kliegel A, Losert H, Sodeck G, Uray T, Zeiner A, Laggner AN . Efficacy and safety of endovascular cooling after cardiac arrest: cohort study and Bayesian approach. Stroke. 2006; 37:1792–1797.LinkGoogle Scholar
  • 30. Skulec R, Kovarnik T, Dostalova G, Kolar J, Linhart A . Induction of mild hypothermia in cardiac arrest survivors presenting with cardiogenic shock syndrome. Acta Anaesthesiol Scand. 2008; 52:188–194.CrossrefMedlineGoogle Scholar
  • 31. Hovdenes J, Laake JH, Aaberge L, Haugaa H, Bugge JF . Therapeutic hypothermia after out-of-hospital cardiac arrest: experiences with patients treated with percutaneous coronary intervention and cardiogenic shock. Acta Anaesthesiol Scand. 2007; 51:137–142.CrossrefMedlineGoogle Scholar
  • 32. Batista LM, Lima FO, Januzzi JL, Donahue V, Snydeman C, Greer DM . Feasibility and safety of combined percutaneous coronary intervention and therapeutic hypothermia following cardiac arrest. Resuscitation. 2010; 81:398–403.CrossrefMedlineGoogle Scholar
  • 33. Wolfrum S, Pierau C, Radke PW, Schunkert H, Kurowski V . Mild therapeutic hypothermia in patients after out-of-hospital cardiac arrest due to acute ST-segment elevation myocardial infarction undergoing immediate percutaneous coronary intervention. Crit Care Med. 2008; 36:1780–1786.CrossrefMedlineGoogle Scholar
  • 34. Knafelj R, Radsel P, Ploj T, Noc M . Primary percutaneous coronary intervention and mild induced hypothermia in comatose survivors of ventricular fibrillation with ST-elevation acute myocardial infarction. Resuscitation. 2007; 74:227–234.CrossrefMedlineGoogle Scholar
  • 35. Nielsen N, Hovdenes J, Nilsson F, Rubertsson S, Stammet P, Sunde K, Valsson F, Wanscher M, Friberg H . Outcome, timing and adverse events in therapeutic hypothermia after out-of-hospital cardiac arrest. Acta Anaesthesiol Scand. 2009; 53:926–934.CrossrefMedlineGoogle Scholar
  • 36. Sunde K, Pytte M, Jacobsen D, Mangschau A, Jensen LP, Smedsrud C, Draegni T, Steen PA . Implementation of a standardised treatment protocol for post resuscitation care after out-of-hospital cardiac arrest. Resuscitation. 2007; 73:29–39.CrossrefMedlineGoogle Scholar
  • 37. Voipio V, Kuisma M, Alaspaa A, Manttari M, Rosenberg P . Thrombolytic treatment of acute myocardial infarction after out-of-hospital cardiac arrest. Resuscitation. 2001; 49:251–258.CrossrefMedlineGoogle Scholar
  • 38. Weston CF, Avery P . Thrombolysis following pre-hospital cardiopulmonary resuscitation. Int J Cardiol. 1992; 37:195–198.CrossrefMedlineGoogle Scholar
  • 39. Kuboyama K, Safar P, Radovsky A, Tisherman SA, Stezoski SW, Alexander H . Delay in cooling negates the beneficial effect of mild resuscitative cerebral hypothermia after cardiac arrest in dogs: a prospective, randomized study. Crit Care Med. 1993; 21:1348–1358.CrossrefMedlineGoogle Scholar
  • 40. Abella BS, Zhao D, Alvarado J, Hamann K, Vanden Hoek TL, Becker LB . Intra-arrest cooling improves outcomes in a murine cardiac arrest model. Circulation. 2004; 109:2786–2791.LinkGoogle Scholar
  • 41. Takata K, Takeda Y, Sato T, Nakatsuka H, Yokoyama M, Morita K . Effects of hypothermia for a short period on histologic outcome and extracellular glutamate concentration during and after cardiac arrest in rats. Crit Care Med. 2005; 33:1340–1345.CrossrefMedlineGoogle Scholar
  • 42. Hicks SD, DeFranco DB, Callaway CW . Hypothermia during reperfusion after asphyxial cardiac arrest improves functional recovery and selectively alters stress-induced protein expression. J Cereb Blood Flow Metab. 2000; 20:520–530.CrossrefMedlineGoogle Scholar
  • 43. Colbourne F, Li H, Buchan AM . Indefatigable CA1 sector neuroprotection with mild hypothermia induced 6 hours after severe forebrain ischemia in rats. J Cereb Blood Flow Metab. 1999; 19:742–749.CrossrefMedlineGoogle Scholar
  • 44. Wolff B, Machill K, Schumacher D, Schulzki I, Werner D . Early achievement of mild therapeutic hypothermia and the neurologic outcome after cardiac arrest. Int J Cardiol. 2009; 133:223–228.CrossrefMedlineGoogle Scholar
  • 45. Gluckman PD, Wyatt JS, Azzopardi D, Ballard R, Edwards AD, Ferriero DM, Polin RA, Robertson CM, Thoresen M, Whitelaw A, Gunn AJ . Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet. 2005; 365(9460):663–670.CrossrefMedlineGoogle Scholar
  • 46. Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF, Fanaroff AA, Poole WK, Wright LL, Higgins RD, Finer NN, Carlo WA, Duara S, Oh W, Cotten CM, Stevenson DK, Stoll BJ, Lemons JA, Guillet R, Jobe AH . Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med. 2005; 353:1574–1584.CrossrefMedlineGoogle Scholar
  • 47. Heard KJ, Peberdy MA, Sayre MR, Sanders A, Geocadin RG, Dixon SR, Larabee TM, Hiller K, Fiorello A, Paradis NA, O'Neil BJ . A randomized controlled trial comparing the Arctic Sun to standard cooling for induction of hypothermia after cardiac arrest. Resuscitation. 81:9–14.CrossrefMedlineGoogle Scholar
  • 48. Flint AC, Hemphill JC, Bonovich DC . Therapeutic hypothermia after cardiac arrest: performance characteristics and safety of surface cooling with or without endovascular cooling. Neurocrit Care. 2007; 7:109–118.CrossrefMedlineGoogle Scholar
  • 49. Pichon N, Amiel JB, Francois B, Dugard A, Etchecopar C, Vignon P . Efficacy of and tolerance to mild induced hypothermia after out-of-hospital cardiac arrest using an endovascular cooling system. Crit Care. 2007; 11:R71.CrossrefMedlineGoogle Scholar
  • 50. Kliegel A, Losert H, Sterz F, Kliegel M, Holzer M, Uray T, Domanovits H . Cold simple intravenous infusions preceding special endovascular cooling for faster induction of mild hypothermia after cardiac arrest–a feasibility study. Resuscitation. 2005; 64:347–351.CrossrefMedlineGoogle Scholar
  • 51. Kliegel A, Janata A, Wandaller C, Uray T, Spiel A, Losert H, Kliegel M, Holzer M, Haugk M, Sterz F, Laggner AN . Cold infusions alone are effective for induction of therapeutic hypothermia but do not keep patients cool after cardiac arrest. Resuscitation. 2007; 73:46–53.CrossrefMedlineGoogle Scholar
  • 52. Kim F, Olsufka M, Carlbom D, Deem S, Longstreth WT, Hanrahan M, Maynard C, Copass MK, Cobb LA . Pilot study of rapid infusion of 2 L of 4 degrees C normal saline for induction of mild hypothermia in hospitalized, comatose survivors of out-of-hospital cardiac arrest. Circulation. 2005; 112:715–719.LinkGoogle Scholar
  • 53. Kliegel A, Losert H, Sterz F, Kliegel M, Holzer M, Uray T, Domanovits H . Cold simple intravenous infusions preceding special endovascular cooling for faster induction of mild hypothermia after cardiac arrest–a feasibility study. Resuscitation. 2005; 64:347–351.CrossrefMedlineGoogle Scholar
  • 54. Bernard S, Buist M, Monteiro O, Smith K . Induced hypothermia using large volume, ice-cold intravenous fluid in comatose survivors of out-of-hospital cardiac arrest: a preliminary report. Resuscitation. 2003; 56:9–13.CrossrefMedlineGoogle Scholar
  • 55. Virkkunen I, Yli-Hankala A, Silfvast T . Induction of therapeutic hypothermia after cardiac arrest in prehospital patients using ice-cold Ringer's solution: a pilot study. Resuscitation. 2004; 62:299–302.CrossrefMedlineGoogle Scholar
  • 56. Jacobshagen C, Pax A, Unsold BW, Seidler T, Schmidt-Schweda S, Hasenfuss G, Maier LS . Effects of large volume, ice-cold intravenous fluid infusion on respiratory function in cardiac arrest survivors. Resuscitation. 2009; 80:1223–1228.CrossrefMedlineGoogle Scholar
  • 57. Kilgannon JH, Roberts BW, Stauss M, Cimino MJ, Ferchau L, Chansky ME, Dellinger RP, Parrillo JE, Trzeciak S . Use of a standardized order set for achieving target temperature in the implementation of therapeutic hypothermia after cardiac arrest: a feasibility study. Acad Emerg Med. 2008; 15:499–505.CrossrefMedlineGoogle Scholar
  • 58. Spiel AO, Kliegel A, Janata A, Uray T, Mayr FB, Laggner AN, Jilma B, Sterz F . Hemostasis in cardiac arrest patients treated with mild hypothermia initiated by cold fluids. Resuscitation. 2009; 80:762–765.CrossrefMedlineGoogle Scholar
  • 59. Larsson IM, Wallin E, Rubertsson S . Cold saline infusion and ice packs alone are effective in inducing and maintaining therapeutic hypothermia after cardiac arrest. Resuscitation. 2010; 81:15–19.CrossrefMedlineGoogle Scholar
  • 60. Kim F, Olsufka M, Longstreth WT, Maynard C, Carlbom D, Deem S, Kudenchuk P, Copass MK, Cobb LA . Pilot randomized clinical trial of prehospital induction of mild hypothermia in out-of-hospital cardiac arrest patients with a rapid infusion of 4 degrees C normal saline. Circulation. 2007; 115:3064–3070.LinkGoogle Scholar
  • 61. Kamarainen A, Virkkunen I, Tenhunen J, Yli-Hankala A, Silfvast T . Prehospital therapeutic hypothermia for comatose survivors of cardiac arrest: a randomized controlled trial. Acta Anaesthesiol Scand. 2009; 53:900–907.CrossrefMedlineGoogle Scholar
  • 62. Hammer L, Vitrat F, Savary D, Debaty G, Santre C, Durand M, Dessertaine G, Timsit JF . Immediate prehospital hypothermia protocol in comatose survivors of out-of-hospital cardiac arrest. Am J Emerg Med. 2009; 27:570–573.CrossrefMedlineGoogle Scholar
  • 63. Kamarainen A, Virkkunen I, Tenhunen J, Yli-Hankala A, Silfvast T . Prehospital induction of therapeutic hypothermia during CPR: a pilot study. Resuscitation. 2008; 76:360–363.CrossrefMedlineGoogle Scholar
  • 64. Kamarainen A, Virkkunen I, Tenhunen J, Yli-Hankala A, Silfvast T . Induction of therapeutic hypothermia during prehospital CPR using ice-cold intravenous fluid. Resuscitation. 2008; 79:205–211.CrossrefMedlineGoogle Scholar
  • 65. Imamura M, Matsukawa T, Ozaki M, Sessler DI, Nishiyama T, Kumazawa T . The accuracy and precision of four infrared aural canal thermometers during cardiac surgery. Acta Anaesthesiol Scand. 1998; 42:1222–1226.CrossrefMedlineGoogle Scholar
  • 66. Pujol A, Fusciardi J, Ingrand P, Baudouin D, Le Guen AF, Menu P . Afterdrop after hypothermic cardiopulmonary bypass: the value of tympanic membrane temperature monitoring. J Cardiothorac Vasc Anesth. 1996; 10:336–341.CrossrefMedlineGoogle Scholar
  • 67. Akata T, Setoguchi H, Shirozu K, Yoshino J . Reliability of temperatures measured at standard monitoring sites as an index of brain temperature during deep hypothermic cardiopulmonary bypass conducted for thoracic aortic reconstruction. J Thorac Cardiovasc Surg. 2007; 133:1559–1565.CrossrefMedlineGoogle Scholar
  • 68. Adrie C, Adib-Conquy M, Laurent I, Monchi M, Vinsonneau C, Fitting C, Fraisse F, Dinh-Xuan AT, Carli P, Spaulding C, Dhainaut JF, Cavaillon JM . Successful cardiopulmonary resuscitation after cardiac arrest as a “sepsis-like” syndrome. Circulation. 2002; 106:562–568.LinkGoogle Scholar
  • 69. Adrie C, Laurent I, Monchi M, Cariou A, Dhainaou JF, Spaulding C . Postresuscitation disease after cardiac arrest: a sepsis-like syndrome?Curr Opin Crit Care. 2004; 10:208–212.CrossrefMedlineGoogle Scholar
  • 70. Takino M, Okada Y . Hyperthermia following cardiopulmonary resuscitation. Intensive Care Med. 1991; 17:419–420.CrossrefMedlineGoogle Scholar
  • 71. Zeiner A, Holzer M, Sterz F, Schorkhuber W, Eisenburger P, Havel C, Kliegel A, Laggner AN . Hyperthermia after cardiac arrest is associated with an unfavorable neurologic outcome. Arch Intern Med. 2001; 161:2007–2012.CrossrefMedlineGoogle Scholar
  • 72. Hickey RW, Kochanek PM, Ferimer H, Graham SH, Safar P . Hypothermia and hyperthermia in children after resuscitation from cardiac arrest. Pediatrics. 2000; 106(pt 1):118–122.CrossrefMedlineGoogle Scholar
  • 73. Langhelle A, Tyvold SS, Lexow K, Hapnes SA, Sunde K, Steen PA . In-hospital factors associated with improved outcome after out-of-hospital cardiac arrest. A comparison between four regions in Norway. Resuscitation. 2003; 56:247–263.CrossrefMedlineGoogle Scholar
  • 74. Takasu A, Saitoh D, Kaneko N, Sakamoto T, Okada Y . Hyperthermia: is it an ominous sign after cardiac arrest?Resuscitation. 2001; 49:273–277.CrossrefMedlineGoogle Scholar
  • 75. Wang Y, Lim LL, Levi C, Heller RF, Fisher J . Influence of admission body temperature on stroke mortality. Stroke. 2000; 31:404–409.LinkGoogle Scholar
  • 76. Diringer MN . Treatment of fever in the neurologic intensive care unit with a catheter-based heat exchange system. Crit Care Med. 2004; 32:559–564.CrossrefMedlineGoogle Scholar
  • 77. Diringer MN, Reaven NL, Funk SE, Uman GC . Elevated body temperature independently contributes to increased length of stay in neurologic intensive care unit patients. Crit Care Med. 2004; 32:1489–1495.CrossrefMedlineGoogle Scholar
  • 78. Reith J, Jorgensen HS, Pedersen PM, Nakayama H, Raaschou HO, Jeppesen LL, Olsen TS . Body temperature in acute stroke: relation to stroke severity, infarct size, mortality, and outcome. Lancet. 1996; 347(8999):422–425.CrossrefMedlineGoogle Scholar
  • 79. Hanchaiphiboolkul S . Body temperature and mortality in acute cerebral infarction. J Med Assoc Thai. 2005; 88:26–31.MedlineGoogle Scholar
  • 80. Kammersgaard LP, Jorgensen HS, Rungby JA, Reith J, Nakayama H, Weber UJ, Houth J, Olsen TS . Admission body temperature predicts long-term mortality after acute stroke: the Copenhagen Stroke Study. Stroke. 2002; 33:1759–1762.LinkGoogle Scholar
  • 81. Bernard GR, Artigas A, Brigham KL, Carlet J, Falke K, Hudson L, Lamy M, Legall JR, Morris A, Spragg R . The Am-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med. 1994; 149(3 Pt 1): 818–824.CrossrefMedlineGoogle Scholar
  • 82. Marsala J, Marsala M, Vanicky I, Galik J, Orendacova J . Post cardiac arrest hyperoxic resuscitation enhances neuronal vulnerability of the respiratory rhythm generator and some brainstem and spinal cord neuronal pools in the dog. Neurosci Lett. 1992; 146:121–124.CrossrefMedlineGoogle Scholar
  • 83. Zwemer CF, Whitesall SE, D'Alecy LG . Cardiopulmonary-cerebral resuscitation with 100% oxygen exacerbates neurological dysfunction following nine minutes of normothermic cardiac arrest in dogs. Resuscitation. 1994; 27:159–170.CrossrefMedlineGoogle Scholar
  • 84. Liu Y, Rosenthal RE, Haywood Y, Miljkovic-Lolic M, Vanderhoek JY, Fiskum G . Normoxic ventilation after cardiac arrest reduces oxidation of brain lipids and improves neurological outcome. Stroke. 1998; 29:1679–1686.LinkGoogle Scholar
  • 85. Vereczki V, Martin E, Rosenthal RE, Hof PR, Hoffman GE, Fiskum G . Normoxic resuscitation after cardiac arrest protects against hippocampal oxidative stress, metabolic dysfunction, and neuronal death. J Cereb Blood Flow Metab. 2006; 26:821–835.CrossrefMedlineGoogle Scholar
  • 86. Richards EM, Fiskum G, Rosenthal RE, Hopkins I, McKenna MC . Hyperoxic reperfusion after global ischemia decreases hippocampal energy metabolism. Stroke. 2007; 38:1578–1584.LinkGoogle Scholar
  • 87. Richards EM, Rosenthal RE, Kristian T, Fiskum G . Postischemic hyperoxia reduces hippocampal pyruvate dehydrogenase activity. Free Radic Biol Med. 2006; 40:1960–1970.CrossrefMedlineGoogle Scholar
  • 88. Kuisma M, Boyd J, Voipio V, Alaspaa A, Roine RO, Rosenberg P . Comparison of 30 and the 100% inspired oxygen concentrations during early post-resuscitation period: a randomised controlled pilot study. Resuscitation. 2006; 69:199–206.CrossrefMedlineGoogle Scholar
  • 89. Safar P, Xiao F, Radovsky A, Tanigawa K, Ebmeyer U, Bircher N, Alexander H, Stezoski SW . Improved cerebral resuscitation from cardiac arrest in dogs with mild hypothermia plus blood flow promotion. Stroke. 1996; 27:105–113.LinkGoogle Scholar
  • 90. Kagstrom E, Smith ML, Siesjo BK . Cerebral circulatory responses to hypercapnia and hypoxia in the recovery period following complete and incomplete cerebral ischemia in the rat. Acta Physiol Scand. 1983; 118:281–291.CrossrefMedlineGoogle Scholar
  • 91. Krep H, Brinker G, Pillekamp F, Hossmann KA . Treatment with an endothelin type A receptor-antagonist after cardiac arrest and resuscitation improves cerebral hemodynamic and functional recovery in rats. Crit Care Med. 2000; 28:2866–2872.CrossrefMedlineGoogle Scholar
  • 92. Krep H, Brinker G, Schwindt W, Hossmann KA . Endothelin type A-antagonist improves long-term neurological recovery after cardiac arrest in rats. Crit Care Med. 2000; 28:2873–2880.CrossrefMedlineGoogle Scholar
  • 93. Nemoto EM, Snyder JV, Carroll RG, Morita H . Global ischemia in dogs: cerebrovascular CO2 reactivity and autoregulation. Stroke. 1975; 6:425–431.LinkGoogle Scholar
  • 94. Vanicky I, Marsala M, Murar J, Marsala J . Prolonged postischemic hyperventilation reduces acute neuronal damage after 15 min of cardiac arrest in the dog. Neurosci Lett. 1992; 135:167–170.CrossrefMedlineGoogle Scholar
  • 95. Wolfson SK, Safar P, Reich H, Clark JM, Gur D, Stezoski W, Cook EE, Krupper MA . Dynamic heterogeneity of cerebral hypoperfusion after prolonged cardiac arrest in dogs measured by the stable xenon/CT technique: a preliminary study. Resuscitation. 1992; 23:1–20.CrossrefMedlineGoogle Scholar
  • 96. Fischer M, Hossmann KA . No-reflow after cardiac arrest. Intensive Care Med. 1995; 21:132–141.CrossrefMedlineGoogle Scholar
  • 97. Ausina A, Baguena M, Nadal M, Manrique S, Ferrer A, Sahuquillo J, Garnacho A . Cerebral hemodynamic changes during sustained hypocapnia in severe head injury: can hyperventilation cause cerebral ischemia?Acta Neurochir Suppl. 1998; 71:1–4.MedlineGoogle Scholar
  • 98. Yundt KD, Diringer MN . The use of hyperventilation and its impact on cerebral ischemia in the treatment of traumatic brain injury. Crit Care Clin. 1997; 13:163–184.CrossrefMedlineGoogle Scholar
  • 99. Buunk G, van der Hoeven JG, Meinders AE . Cerebrovascular reactivity in comatose patients resuscitated from a cardiac arrest. Stroke. 1997; 28:1569–1573.LinkGoogle Scholar
  • 100. Yannopoulos D, Aufderheide TP, Gabrielli A, Beiser DG, McKnite SH, Pirrallo RG, Wigginton J, Becker L, Vanden Hoek T, Tang W, Nadkarni VM, Klein JP, Idris AH, Lurie KG . Clinical and hemodynamic comparison of 15:2 and 30:2 compression-to-ventilation ratios for cardiopulmonary resuscitation. Crit Care Med. 2006; 34:1444–1449.CrossrefMedlineGoogle Scholar
  • 101. Yannopoulos D, Tang W, Roussos C, Aufderheide TP, Idris AH, Lurie KG . Reducing ventilation frequency during cardiopulmonary resuscitation in a porcine model of cardiac arrest. Respir Care. 2005; 50:628–635.MedlineGoogle Scholar
  • 102. Herff H, Paal P, von Goedecke A, Lindner KH, Severing AC, Wenzel V . Influence of ventilation strategies on survival in severe controlled hemorrhagic shock. Crit Care Med. 2008; 36:2613–2620.CrossrefMedlineGoogle Scholar
  • 103. Pepe PE, Raedler C, Lurie KG, Wigginton JG . Emergency ventilatory management in hemorrhagic states: elemental or detrimental?J Trauma. 2003; 54:1048–1055.CrossrefMedlineGoogle Scholar
  • 104. Pepe PE, Marini JJ . Occult positive end-expiratory pressure in mechanically ventilated patients with airflow obstruction: the auto-PEEP effect. Am Rev Respir Dis. 1982; 126:166–170.MedlineGoogle Scholar
  • 105. Franklin C, Samuel J, Hu TC . Life-threatening hypotension associated with emergency intubation and the initiation of mechanical ventilation. Am J Emerg Med. 1994; 12:425–428.CrossrefMedlineGoogle Scholar
  • 106. The 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; 342:1301–1308.CrossrefMedlineGoogle Scholar
  • 107. Tremblay LN, Slutsky AS . Ventilator-induced lung injury: from the bench to the bedside. Intensive Care Med. 2006; 32:24–33.CrossrefMedlineGoogle Scholar
  • 108. Borges JB, Okamoto VN, Matos GF, Caramez MP, Arantes PR, Barros F, Souza CE, Victorino JA, Kacmarek RM, Barbas CS, Carvalho CR, Amato MB . Reversibility of lung collapse and hypoxemia in early acute respiratory distress syndrome. Am J Respir Crit Care Med. 2006; 174:268–278.CrossrefMedlineGoogle Scholar
  • 109. Brower RG, Lanken PN, MacIntyre N, Matthay MA, Morris A, Ancukiewicz M, Schoenfeld D, Thompson BT . Higher versus lower positive end-expiratory pressures in patients with the acute respiratory distress syndrome. N Engl J Med. 2004; 351:327–336.CrossrefMedlineGoogle Scholar
  • 110. Wongsurakiat P, Pierson DJ, Rubenfeld GD . Changing pattern of ventilator settings in patients without acute lung injury: changes over 11 years in a single institution. Chest. 2004; 126:1281–1291.CrossrefMedlineGoogle Scholar
  • 111. Wan S, Quinlan DJ, Agnelli G, Eikelboom JW . Thrombolysis compared with heparin for the initial treatment of pulmonary embolism: a meta-analysis of the randomized controlled trials. Circulation. 2004; 110:744–749.LinkGoogle Scholar
  • 112. Scholz KH, Hilmer T, Schuster S, Wojcik J, Kreuzer H, Tebbe U . Thrombolysis in resuscitated patients with pulmonary embolism. Dtsch Med Wochenschr. 1990; 115:930–935.CrossrefMedlineGoogle Scholar
  • 113. Bottiger BW, Bode C, Kern S, Gries A, Gust R, Glatzer R, Bauer H, Motsch J, Martin E . Efficacy and safety of thrombolytic therapy after initially unsuccessful cardiopulmonary resuscitation: a prospective clinical trial. Lancet. 2001; 357(9268):1583–1585.CrossrefMedlineGoogle Scholar
  • 114. Fatovich DM, Dobb GJ, Clugston RA . A pilot randomised trial of thrombolysis in cardiac arrest (The TICA trial). Resuscitation. 2004; 61:309–313.CrossrefMedlineGoogle Scholar
  • 115. Fava M, Loyola S, Bertoni H, Dougnac A . Massive pulmonary embolism: percutaneous mechanical thrombectomy during cardiopulmonary resuscitation. J Vasc Interv Radiol. 2005; 16:119–123.CrossrefMedlineGoogle Scholar
  • 116. Janata K, Holzer M, Kurkciyan I, Losert H, Riedmuller E, Pikula B, Laggner AN, Laczika K . Major bleeding complications in cardiopulmonary resuscitation: the place of thrombolytic therapy in cardiac arrest due to massive pulmonary embolism. Resuscitation. 2003; 57:49–55.CrossrefMedlineGoogle Scholar
  • 117. Konstantinov IE, Saxena P, Koniuszko MD, Alvarez J, Newman MA . Acute massive pulmonary embolism with cardiopulmonary resuscitation: management and results. Tex Heart Inst J. 2007; 34:41–45; discussion 45–46.MedlineGoogle Scholar
  • 118. Lederer W, Lichtenberger C, Pechlaner C, Kroesen G, Baubin M . Recombinant tissue plasminogen activator during cardiopulmonary resuscitation in 108 patients with out-of-hospital cardiac arrest. Resuscitation. 2001; 50:71–76.CrossrefMedlineGoogle Scholar
  • 119. Lederer W, Lichtenberger C, Pechlaner C, Kinzl J, Kroesen G, Baubin M . Long-term survival and neurological outcome of patients who received recombinant tissue plasminogen activator during out-of-hospital cardiac arrest. Resuscitation. 2004; 61:123–129.CrossrefMedlineGoogle Scholar
  • 120. Zahorec R . Rescue systemic thrombolysis during cardiopulmonary resuscitation. Bratisl Lek Listy. 2002; 103(7–8):266–269.MedlineGoogle Scholar
  • 121. Spohr F, Bottiger BW . Thrombolytic therapy during or after cardiopulmonary resuscitation: efficacy and safety of a new therapeutic approach. Minerva Anestesiol. 2003; 69:357–364.MedlineGoogle Scholar
  • 122. Schmid C, Zietlow S, Wagner TO, Laas J, Borst HG . Fulminant pulmonary embolism: symptoms, diagnostics, operative technique, and results. Ann Thorac Surg. 1991; 52:1102–1105.CrossrefMedlineGoogle Scholar
  • 123. Dauphine C, Omari B . Pulmonary embolectomy for acute massive pulmonary embolism. Ann Thorac Surg. 2005; 79:1240–1244.CrossrefMedlineGoogle Scholar
  • 124. Doerge HC, Schoendube FA, Loeser H, Walter M, Messmer BJ . Pulmonary embolectomy: review of a 15-year experience and role in the age of thrombolytic therapy. Eur J Cardiothorac Surg. 1996; 10:952–957.CrossrefMedlineGoogle Scholar
  • 125. Ullmann M, Hemmer W, Hannekum A . The urgent pulmonary embolectomy: mechanical resuscitation in the operating theatre determines the outcome. Thorac Cardiovasc Surg. 1999; 47:5–8.CrossrefMedlineGoogle Scholar
  • 126. Hall JE, Uhrich TD, Barney JA, Arain SR, Ebert TJ . Sedative, amnestic, and analgesic properties of small-dose dexmedetomidine infusions. Anesth Analg. 2000; 90:699–705.CrossrefMedlineGoogle Scholar
  • 127. Milbrandt EB, Kersten A, Kong L, Weissfeld LA, Clermont G, Fink MP, Angus DC . Haloperidol use is associated with lower hospital mortality in mechanically ventilated patients. Crit Care Med. 2005; 33:226–229, discussion 263–225.CrossrefMedlineGoogle Scholar
  • 128. De Jonghe B, Cook D, Griffith L, Appere-de-Vecchi C, Guyatt G, Theron V, Vagnerre A, Outin H . Adaptation to the Intensive Care Environment (ATICE): development and validation of a new sedation assessment instrument. Crit Care Med. 2003; 31:2344–2354.CrossrefMedlineGoogle Scholar
  • 129. Weinert C, McFarland L . The state of intubated ICU patients: development of a two-dimensional sedation rating scale for critically ill adults. Chest. 2004; 126:1883–1890.CrossrefMedlineGoogle Scholar
  • 130. Ramsay MA, Savege TM, Simpson BR, Goodwin R . Controlled sedation with alphaxalone-alphadolone. Br Med J. 1974; 2(5920):656–659.CrossrefMedlineGoogle Scholar
  • 131. Sessler CN, Gosnell MS, Grap MJ, Brophy GM, O'Neal PV, Keane KA, Tesoro EP, Elswick RK . The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med. 2002; 166:1338–1344.CrossrefMedlineGoogle Scholar
  • 132. Riker RR, Fraser GL, Cox PM . Continuous infusion of haloperidol controls agitation in critically ill patients. Crit Care Med. 1994; 22:433–440.CrossrefMedlineGoogle Scholar
  • 133. de Lemos J, Tweeddale M, Chittock D . Measuring quality of sedation in adult mechanically ventilated critically ill patients: the Vancouver Interaction and Calmness Scale. Sedation Focus Group. J Clin Epidemiol. 2000; 53:908–919.CrossrefMedlineGoogle Scholar
  • 134. Devlin JW, Boleski G, Mlynarek M, Nerenz DR, Peterson E, Jankowski M, Horst HM, Zarowitz BJ . Motor Activity Assessment Scale: a valid and reliable sedation scale for use with mechanically ventilated patients in an adult surgical intensive care unit. Crit Care Med. 1999; 27:1271–1275.CrossrefMedlineGoogle Scholar
  • 135. Rello J, Diaz E, Roque M, Valles J . Risk factors for developing pneumonia within 48 hours of intubation. Am J Respir Crit Care Med. 1999; 159:1742–1746.CrossrefMedlineGoogle Scholar
  • 136. Bendz B, Eritsland J, Nakstad AR, Brekke M, Klow NE, Steen PA, Mangschau A . Long-term prognosis after out-of-hospital cardiac arrest and primary percutaneous coronary intervention. Resuscitation. 2004; 63:49–53.CrossrefMedlineGoogle Scholar
  • 137. Engdahl J, Abrahamsson P, Bang A, Lindqvist J, Karlsson T, Herlitz J . Is hospital care of major importance for outcome after out-of-hospital cardiac arrest? Experience acquired from patients with out-of-hospital cardiac arrest resuscitated by the same Emergency Medical Service and admitted to one of two hospitals over a 16-year period in the municipality of Goteborg. Resuscitation. 2000; 43:201–211.CrossrefMedlineGoogle Scholar
  • 138. Gorjup V, Radsel P, Kocjancic ST, Erzen D, Noc M . Acute ST-elevation myocardial infarction after successful cardiopulmonary resuscitation. Resuscitation. 2007; 72:379–385.CrossrefMedlineGoogle Scholar
  • 139. Garot P, Lefevre T, Eltchaninoff H, Morice MC, Tamion F, Abry B, Lesault PF, Le Tarnec JY, Pouges C, Margenet A, Monchi M, Laurent I, Dumas P, Garot J, Louvard Y . Six-month outcome of emergency percutaneous coronary intervention in resuscitated patients after cardiac arrest complicating ST-elevation myocardial infarction. Circulation. 2007; 115:1354–1362.LinkGoogle Scholar
  • 140. Lettieri C, Savonitto S, De Servi S, Guagliumi G, Belli G, Repetto A, Piccaluga E, Politi A, Ettori F, Castiglioni B, Fabbiocchi F, De Cesare N, Sangiorgi G, Musumeci G, Onofri M, D'Urbano M, Pirelli S, Zanini R, Klugmann S . Emergency percutaneous coronary intervention in patients with ST-elevation myocardial infarction complicated by out-of-hospital cardiac arrest: early and medium-term outcome. Am Heart J. 2009; 157:569–575.CrossrefMedlineGoogle Scholar
  • 141. Kahn JK, Glazier S, Swor R, Savas V, O'Neill WW . Primary coronary angioplasty for acute myocardial infarction complicated by out-of-hospital cardiac arrest. Am J Cardiol. 1995; 75:1069–1070.CrossrefMedlineGoogle Scholar
  • 142. Marcusohn E, Roguin A, Sebbag A, Aronson D, Dragu R, Amikam S, Boulus M, Grenadier E, Kerner A, Nikolsky E, Markiewicz W, Hammerman H, Kapeliovich M . Primary percutaneous coronary intervention after out-of-hospital cardiac arrest: patients and outcomes. Isr Med Assoc J. 2007; 9:257–259.MedlineGoogle Scholar
  • 143. McCullough PA, Prakash R, Tobin KJ, O'Neill WW, Thompson RJ . Application of a cardiac arrest score in patients with sudden death and ST segment elevation for triage to angiography and intervention. J Interv Cardiol. 2002; 15:257–261.CrossrefMedlineGoogle Scholar
  • 144. Nagao K, Hayashi N, Kanmatsuse K, Arima K, Ohtsuki J, Kikushima K, Watanabe I . Cardiopulmonary cerebral resuscitation using emergency cardiopulmonary bypass, coronary reperfusion therapy and mild hypothermia in patients with cardiac arrest outside the hospital. J Am Coll Cardiol. 2000; 36:776–783.CrossrefMedlineGoogle Scholar
  • 145. Peels HO, Jessurun GA, van der Horst IC, Arnold AE, Piers LH, Zijlstra F . Outcome in transferred and nontransferred patients after primary percutaneous coronary intervention for ischaemic out-of-hospital cardiac arrest. Catheter Cardiovasc Interv. 2008; 71:147–151.CrossrefMedlineGoogle Scholar
  • 146. Pleskot M, Babu A, Hazukova R, Stritecky J, Bis J, Matejka J, Cermakova E . Out-of-hospital cardiac arrests in patients with acute ST elevation myocardial infarctions in the East Bohemian region over the period 2002–2004. Cardiology. 2008; 109:41–51.CrossrefMedlineGoogle Scholar
  • 147. Quintero-Moran B, Moreno R, Villarreal S, Perez-Vizcayno MJ, Hernandez R, Conde C, Vazquez P, Alfonso F, Banuelos C, Escaned J, Fernandez-Ortiz A, Azcona L, Macaya C . Percutaneous coronary intervention for cardiac arrest secondary to ST-elevation acute myocardial infarction. Influence of immediate paramedical/medical assistance on clinical outcome. J Invasive Cardiol. 2006; 18:269–272.MedlineGoogle Scholar
  • 148. Investigators A . A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular arrhythmias. The Antiarrhythmics versus Implantable Defibrillators (AVID) Investigators. N Engl J Med. 1997; 337:1576–1583.CrossrefMedlineGoogle Scholar
  • 149. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G . A randomized study of the prevention of sudden death in patients with coronary artery disease. Multicenter Unsustained Tachycardia Trial Investigators. N Engl J Med. 1999; 341:1882–1890.CrossrefMedlineGoogle Scholar
  • 150. Connolly SJ, Gent M, Roberts RS, Dorian P, Roy D, Sheldon RS, Mitchell LB, Green MS, Klein GJ, O'Brien B . Canadian implantable defibrillator study (CIDS): a randomized trial of the implantable cardioverter defibrillator against amiodarone. Circulation. 2000; 101:1297–1302.LinkGoogle Scholar
  • 151. Kuck KH, Cappato R, Siebels J, Ruppel R . Randomized comparison of antiarrhythmic drug therapy with implantable defibrillators in patients resuscitated from cardiac arrest: the Cardiac Arrest Study Hamburg (CASH). Circulation. 2000; 102:748–754.LinkGoogle Scholar
  • 152. Wever EFD, Hauer RNW, Van Capelle FJL, Tijssen JGP, Crijns HJGM, Algra A, Wiesfeld ACP, Bakker PFA, Robles de Medina EO . Randomized study of implantable defibrillator as first-choice therapy versus conventional strategy in postinfarct sudden death survivors. Circulation. 1995; 91:2195–2203.LinkGoogle Scholar
  • 153. De Backer D, Biston P, Devriendt J, Madl C, Chochrad D, Aldecoa C, Brasseur A, Defrance P, Gottignies P, Vincent JL . Comparison of dopamine and norepinephrine in the treatment of shock. N Engl J Med. 2010; 362:779–789.CrossrefMedlineGoogle Scholar
  • 154. Kellum JA, Pinsky MR . Use of vasopressor agents in critically ill patients. Curr Opin Crit Care. 2002; 8:236–241.CrossrefMedlineGoogle Scholar
  • 155. Zaritsky AL . Catecholamines, inotropic medications, and vasopressor agents. In: , Chernow B ed. The Pharmacologic Approach to the Critically Ill Patient. III ed. Baltimore, MD: Williams & Wilkins; 1994:387–404.Google Scholar
  • 156. Grillo JA, Gonzalez ER, Ramaiya A, Karnes HT, Wells B . Chemical compatibility of inotropic and vasoactive agents delivered via a multiple line infusion system. Crit Care Med. 1995; 23:1061–1066.CrossrefMedlineGoogle Scholar
  • 157. Bonhomme L, Benhamou D, Comoy E, Preaux N . Stability of epinephrine in alkalinized solutions. Ann Emerg Med. 1990; 19:1242–1244.CrossrefMedlineGoogle Scholar
  • 158. Vasquez A, Kern KB, Hilwig RW, Heidenreich J, Berg RA, Ewy GA . Optimal dosing of dobutamine for treating post-resuscitation left ventricular dysfunction. Resuscitation. 2004; 61:199–207.CrossrefMedlineGoogle Scholar
  • 159. Gisvold SE, Sterz F, Abramson NS, Bar-Joseph G, Ebmeyer U, Gervais H, Ginsberg M, Katz LM, Kochanek PM, Kuboyama K, Miller B, Obrist W, Roine RO, Safar P, Sim KM, Vandevelde K, White RJ, Xiao F . Cerebral resuscitation from cardiac arrest: treatment potentials. Crit Care Med. 1996; 24(2 Suppl):S69–80.CrossrefMedlineGoogle Scholar
  • 160. del Zoppo GJ, Mabuchi T . Cerebral microvessel responses to focal ischemia. J Cereb Blood Flow Metab. 2003; 23:879–894.CrossrefMedlineGoogle Scholar
  • 161. Zandbergen EG, de Haan RJ, Stoutenbeek CP, Koelman JH, Hijdra A . Systematic review of early prediction of poor outcome in anoxic-ischaemic coma. Lancet. 1998; 352(9143):1808–1812.CrossrefMedlineGoogle Scholar
  • 162. Rothstein TL . Recovery from near death following cerebral anoxia: A case report demonstrating superiority of median somatosensory evoked potentials over EEG in predicting a favorable outcome after cardiopulmonary resuscitation. Resuscitation. 2004; 60:335–341.CrossrefMedlineGoogle Scholar
  • 163. Kaplan PW, Genoud D, Ho TW, Jallon P . Etiology, neurologic correlations, and prognosis in alpha coma. Clin Neurophysiol. 1999; 110:205–213.CrossrefMedlineGoogle Scholar
  • 164. Mullner M, Domanovits H, Sterz F, Herkner H, Gamper G, Kurkciyan I, Laggner AN . Measurement of myocardial contractility following successful resuscitation: quantitated left ventricular systolic function utilising non-invasive wall stress analysis. Resuscitation. 1998; 39:51–59.CrossrefMedlineGoogle Scholar
  • 165. Weaver WD, Cobb LA, Copass MK, Hallstrom AP . Ventricular defibrillation: a comparative trial using 175-J and 320-J shocks. N Engl J Med. 1982; 307:1101–1106.CrossrefMedlineGoogle Scholar
  • 166. Mayr V, Luckner G, Jochberger S, Wenzel V, Ulmer H, Pajk W, Knotzer H, Friesenecker B, Lindner K, Hasibeder W, Dunser M . Arginine vasopressin in advanced cardiovascular failure during the post-resuscitation phase after cardiac arrest. Resuscitation. 2007; 72:35–44.CrossrefMedlineGoogle Scholar
  • 167. Stevenson LW, Miller LW, Desvigne-Nickens P, Ascheim DD, Parides MK, Renlund DG, Oren RM, Krueger SK, Costanzo MR, Wann LS, Levitan RG, Mancini D . Left ventricular assist device as destination for patients undergoing intravenous inotropic therapy: a subset analysis from REMATCH (Randomized Evaluation of Mechanical Assistance in Treatment of Chronic Heart Failure). Circulation. 2004; 110:975–981.LinkGoogle Scholar
  • 168. Thiele H, Sick P, Boudriot E, Diederich KW, Hambrecht R, Niebauer J, Schuler G . Randomized comparison of intra-aortic balloon support with a percutaneous left ventricular assist device in patients with revascularized acute myocardial infarction complicated by cardiogenic shock. Eur Heart J. 2005; 26:1276–1283.CrossrefMedlineGoogle Scholar
  • 169. Burkhoff D, Cohen H, Brunckhorst C, O'Neill WW . A randomized multicenter clinical study to evaluate the safety and efficacy of the TandemHeart percutaneous ventricular assist device versus conventional therapy with intraaortic balloon pumping for treatment of cardiogenic shock. Am Heart J. 2006; 152:469 e461–e468.CrossrefGoogle Scholar
  • 170. Greenberg SB, Deshur M, Khavkin Y, Karaikovic E, Vender J . Successful resuscitation of a patient who developed cardiac arrest from pulsed saline bacitracin lavage during thoracic laminectomy and fusion. J Clin Anesth. 2008; 20:294–296.CrossrefMedlineGoogle Scholar
  • 171. Seyfarth M, Sibbing D, Bauer I, Frohlich G, Bott-Flugel L, Byrne R, Dirschinger J, Kastrati A, Schomig A . A randomized clinical trial to evaluate the safety and efficacy of a percutaneous left ventricular assist device versus intra-aortic balloon pumping for treatment of cardiogenic shock caused by myocardial infarction. J Am Coll Cardiol. 2008; 52:1584–1588.CrossrefMedlineGoogle Scholar
  • 172. Dellinger RP, Carlet JM, Masur H, Gerlach H, Calandra T, Cohen J, Gea-Banacloche J, Keh D, Marshall JC, Parker MM, Ramsay G, Zimmerman JL, Vincent JL, Levy MM . Surviving Sepsis Campaign guidelines for management of severe sepsis and septic shock. Crit Care Med. 2004; 32:858–873.CrossrefMedlineGoogle Scholar
  • 173. Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M . Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001; 345:1368–1377.CrossrefMedlineGoogle Scholar
  • 174. Nolan JP, Laver SR, Welch CA, Harrison DA, Gupta V, Rowan K . Outcome following admission to UK intensive care units after cardiac arrest: a secondary analysis of the ICNARC Case Mix Programme Database. Anaesthesia. 2007; 62:1207–1216.CrossrefMedlineGoogle Scholar
  • 175. Losert H, Sterz F, Roine RO, Holzer M, Martens P, Cerchiari E, Tiainen M, Mullner M, Laggner AN, Herkner H, Bischof MG . Strict normoglycaemic blood glucose levels in the therapeutic management of patients within 12h after cardiac arrest might not be necessary. Resuscitation. 2008; 76:214–220.CrossrefMedlineGoogle Scholar
  • 176. Mullner M, Sterz F, Binder M, Schreiber W, Deimel A, Laggner AN . Blood glucose concentration after cardiopulmonary resuscitation influences functional neurological recovery in human cardiac arrest survivors. J Cereb Blood Flow Metab. 1997; 17:430–436.CrossrefMedlineGoogle Scholar
  • 177. Oksanen T, Skrifvars MB, Varpula T, Kuitunen A, Pettila V, Nurmi J, Castren M . Strict versus moderate glucose control after resuscitation from ventricular fibrillation. Intensive Care Med. 2007; 33:2093–2100.CrossrefMedlineGoogle Scholar
  • 178. van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R . Intensive insulin therapy in the critically ill patients. N Engl J Med. 2001; 345:1359–1367.CrossrefMedlineGoogle Scholar
  • 179. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R . Intensive insulin therapy in the medical ICU. N Engl J Med. 2006; 354:449–461.CrossrefMedlineGoogle Scholar
  • 180. Arabi YM, Dabbagh OC, Tamim HM, Al-Shimemeri AA, Memish ZA, Haddad SH, Syed SJ, Giridhar HR, Rishu AH, Al-Daker MO, Kahoul SH, Britts RJ, Sakkijha MH . Intensive versus conventional insulin therapy: a randomized controlled trial in medical and surgical critically ill patients. Crit Care Med. 2008; 36:3190–3197.CrossrefMedlineGoogle Scholar
  • 181. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K . Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008; 358:125–139.CrossrefMedlineGoogle Scholar
  • 182. Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Henderson WR, Hebert PC, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ . Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009; 360:1283–1297.CrossrefMedlineGoogle Scholar
  • 183. Preiser JC, Devos P, Ruiz-Santana S, Melot C, Annane D, Groeneveld J, Iapichino G, Leverve X, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chiolero R . A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Med. 2009; 35:1738–1748.CrossrefMedlineGoogle Scholar
  • 184. Griesdale DE, de Souza RJ, van Dam RM, Heyland DK, Cook DJ, Malhotra A, Dhaliwal R, Henderson WR, Chittock DR, Finfer S, Talmor D . Intensive insulin therapy and mortality among critically ill patients: a meta-analysis including NICE-SUGAR study data. CMAJ. 2009; 180:821–827.CrossrefMedlineGoogle Scholar
  • 185. Wiener RS, Wiener DC, Larson RJ . Benefits and risks of tight glucose control in critically ill adults: a meta-analysis. JAMA. 2008; 300:933–944.CrossrefMedlineGoogle Scholar
  • 186. Krinsley JS, Grover A . Severe hypoglycemia in critically ill patients: risk factors and outcomes. Crit Care Med. 2007; 35:2262–2267.CrossrefMedlineGoogle Scholar
  • 187. Arabi YM, Tamim HM, Rishu AH . Hypoglycemia with intensive insulin therapy in critically ill patients: predisposing factors and association with mortality. Crit Care Med. 2009; 37:2536–2544.CrossrefMedlineGoogle Scholar
  • 188. Schultz CH, Rivers EP, Feldkamp CS, Goad EG, Smithline HA, Martin GB, Fath JJ, Wortsman J, Nowak RM . A characterization of hypothalamic-pituitary-adrenal axis function during and after human cardiac arrest. Crit Care Med. 1993; 21:1339–1347.CrossrefMedlineGoogle Scholar
  • 189. Kim JJ, Lim YS, Shin JH, Yang HJ, Kim JK, Hyun SY, Rhoo I, Hwang SY, Lee G . Relative adrenal insufficiency after cardiac arrest: impact on postresuscitation disease outcome. Am J Emerg Med. 2006; 24:684–688.CrossrefMedlineGoogle Scholar
  • 190. Pene F, Hyvernat H, Mallet V, Cariou A, Carli P, Spaulding C, Dugue MA, Mira JP . Prognostic value of relative adrenal insufficiency after out-of-hospital cardiac arrest. Intensive Care Med. 2005; 31:627–633.CrossrefMedlineGoogle Scholar
  • 191. Mentzelopoulos SD, Zakynthinos SG, Tzoufi M, Katsios N, Papastylianou A, Gkisioti S, Stathopoulos A, Kollintza A, Stamataki E, Roussos C . Vasopressin, epinephrine, and corticosteroids for in-hospital cardiac arrest. Arch Intern Med. 2009; 169:15–24.CrossrefMedlineGoogle Scholar
  • 192. Minneci PC, Deans KJ, Banks SM, Eichacker PQ, Natanson C . Corticosteroids for septic shock. Ann Intern Med. 2004; 141:742–743.CrossrefMedlineGoogle Scholar
  • 193. Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, Weiss YG, Benbenishty J, Kalenka A, Forst H, Laterre PF, Reinhart K, Cuthbertson BH, Payen D, Briegel J . Hydrocortisone therapy for patients with septic shock. N Engl J Med. 2008; 358:111–124.CrossrefMedlineGoogle Scholar
  • 194. Annane D, Sebille V, Charpentier C, Bollaert PE, Francois B, Korach JM, Capellier G, Cohen Y, Azoulay E, Troche G, Chaumet-Riffaud P, Bellissant E . Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA. 2002; 288:862–871.CrossrefMedlineGoogle Scholar
  • 195. Laurent I, Adrie C, Vinsonneau C, Cariou A, Chiche JD, Ohanessian A, Spaulding C, Carli P, Dhainaut JF, Monchi M . High-volume hemofiltration after out-of-hospital cardiac arrest: a randomized study. J Am Coll Cardiol. 2005; 46:432–437.CrossrefMedlineGoogle Scholar
  • 196. Brain Resuscitation Clinical Trial I Study Group. Randomized clinical study of thiopental loading in comatose survivors of cardiac arrest. N Engl J Med. 1986; 314:397–403.CrossrefMedlineGoogle Scholar
  • 197. Holzer M, Sterz F, Behringer W, Oschatz E, Kofler J, Eisenburger P, Kittler H, Konschitzky R, Laggner AN . Endothelin-1 elevates regional cerebral perfusion during prolonged ventricular fibrillation cardiac arrest in pigs. Resuscitation. 2002; 55:317–327.CrossrefMedlineGoogle Scholar
  • 198. Longstreth WT, Fahrenbruch CE, Olsufka M, Walsh TR, Copass MK, Cobb LA . Randomized clinical trial of magnesium, diazepam, or both after out-of-hospital cardiac arrest. Neurology. 2002; 59:506–514.CrossrefMedlineGoogle Scholar
  • 199. Krumholz A, Stern BJ, Weiss HD . Outcome from coma after cardiopulmonary resuscitation: relation to seizures and myoclonus. Neurology. 1988; 38:401–405.CrossrefMedlineGoogle Scholar
  • 200. Wijdicks EF, Parisi JE, Sharbrough FW . Prognostic value of myoclonus status in comatose survivors of cardiac arrest. Ann Neurol. 1994; 35:239–243.CrossrefMedlineGoogle Scholar
  • 201. Hui AC, Cheng C, Lam A, Mok V, Joynt GM . Prognosis following Postanoxic Myoclonus Status epilepticus. Eur Neurol. 2005; 54:10–13.CrossrefMedlineGoogle Scholar
  • 202. Damian MS, Ellenberg D, Gildemeister R, Lauermann J, Simonis G, Sauter W, Georgi C . Coenzyme Q10 combined with mild hypothermia after cardiac arrest: a preliminary study. Circulation. 2004; 110:3011–3016.LinkGoogle Scholar
  • 203. Wijdicks EF, Hijdra A, Young GB, Bassetti CL, Wiebe S . Practice parameter: prediction of outcome in comatose survivors after cardiopulmonary resuscitation (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2006; 67:203–210.CrossrefMedlineGoogle Scholar
  • 204. Zandbergen EG, Hijdra A, Koelman JH, Hart AA, Vos PE, Verbeek MM, de Haan RJ . Prediction of poor outcome within the first 3 days of postanoxic coma. Neurology. 2006; 66:62–68.CrossrefMedlineGoogle Scholar
  • 205. Edgren E, Hedstrand U, Nordin M, Rydin E, Ronquist G . Prediction of outcome after cardiac arrest. Crit Care Med. 1987; 15:820–825.CrossrefMedlineGoogle Scholar
  • 206. Young GB, Doig G, Ragazzoni A . Anoxic-ischemic encephalopathy: clinical and electrophysiological associations with outcome. Neurocrit Care. 2005; 2:159–164.CrossrefMedlineGoogle Scholar
  • 207. Bassetti C, Bomio F, Mathis J, Hess CW . Early prognosis in coma after cardiac arrest: a prospective clinical, electrophysiological, and biochemical study of 60 patients. J Neurol Neurosurg Psychiatry. 1996; 61:610–615.CrossrefMedlineGoogle Scholar
  • 208. Edgren E, Hedstrand U, Kelsey S, Sutton-Tyrrell K, Safar P . Assessment of neurological prognosis in comatose survivors of cardiac arrest. BRCT I Study Group. Lancet. 1994; 343(8905):1055–1059.CrossrefMedlineGoogle Scholar
  • 209. Arnoldus EP, Lammers GJ . Postanoxic coma: good recovery despite myoclonus status. Ann Neurol. 1995; 38:697–698.CrossrefMedlineGoogle Scholar
  • 210. Celesia GG, Grigg MM, Ross E . Generalized status myoclonicus in acute anoxic and toxic-metabolic encephalopathies. Arch Neurol. 1988; 45:781–784.CrossrefMedlineGoogle Scholar
  • 211. Datta S, Hart GK, Opdam H, Gutteridge G, Archer J . Post-hypoxic myoclonic status: the prognosis is not always hopeless. Crit Care Resusc. 2009; 11:39–41.MedlineGoogle Scholar
  • 212. English WA, Giffin NJ, Nolan JP . Myoclonus after cardiac arrest: pitfalls in diagnosis and prognosis. Anaesthesia. 2009; 64:908–911.CrossrefMedlineGoogle Scholar
  • 213. Morris HR, Howard RS, Brown P . Early myoclonic status and outcome after cardiorespiratory arrest. J Neurol Neurosurg Psychiatry. 1998; 64:267–268.CrossrefMedlineGoogle Scholar
  • 214. Rundgren M, Rosen I, Friberg H . Amplitude-integrated EEG (aEEG) predicts outcome after cardiac arrest and induced hypothermia. Intensive Care Med. 2006; 32:836–842.CrossrefMedlineGoogle Scholar
  • 215. Shibata S, Imota T, Shigeomi S, Sato W, Enzan K . Use of the bispectral index during the early postresuscitative phase after out-of-hospital cardiac arrest. J Anesth. 2005; 19:243–246.CrossrefMedlineGoogle Scholar
  • 216. Stammet P, Werer C, Mertens L, Lorang C, Hemmer M . Bispectral index (BIS) helps predicting bad neurological outcome in comatose survivors after cardiac arrest and induced therapeutic hypothermia. Resuscitation. 2009; 80:437–442.CrossrefMedlineGoogle Scholar
  • 217. Ajisaka H . Early electroencephalographic findings in patients with anoxic encephalopathy after cardiopulmonary arrest and successful resusitation. J Clin Neurosci. 2004; 11:616–618.CrossrefMedlineGoogle Scholar
  • 218. Rossetti AO, Logroscino G, Liaudet L, Ruffieux C, Ribordy V, Schaller MD, Despland PA, Oddo M . Status epilepticus: an independent outcome predictor after cerebral anoxia. Neurology. 2007; 69:255–260.CrossrefMedlineGoogle Scholar
  • 219. Berkhoff M, Donati F, Bassetti C . Postanoxic alpha (theta) coma: a reappraisal of its prognostic significance. Clin Neurophysiol. 2000; 111:297–304.CrossrefMedlineGoogle Scholar
  • 220. Thomke F, Brand A, Weilemann SL . The temporal dynamics of postanoxic burst-suppression EEG. J Clin Neurophysiol. 2002; 19:24–31.CrossrefMedlineGoogle Scholar
  • 221. Fatovich DM, Jacobs IG, Celenza A, Paech MJ . An observational study of bispectral index monitoring for out of hospital cardiac arrest. Resuscitation. 2006; 69:207–212.CrossrefMedlineGoogle Scholar
  • 222. Rossetti AO, Oddo M, Liaudet L, Kaplan PW . Predictors of awakening from postanoxic status epilepticus after therapeutic hypothermia. Neurology. 2009; 72:744–749.CrossrefMedlineGoogle Scholar
  • 223. Allen JS, Tranel D, Bruss J, Damasio H . Correlations between regional brain volumes and memory performance in anoxia. J Clin Exp Neuropsychol. 2006; 28:457–476.CrossrefMedlineGoogle Scholar
  • 224. De Volder AG, Michel C, Guerit JM, Bol A, Georges B, de Barsy T, Laterre C . Brain glucose metabolism in postanoxic syndrome due to cardiac arrest. Acta Neurol Belg. 1994; 94:183–189.MedlineGoogle Scholar
  • 225. Fujioka M, Nishio K, Miyamoto S, Hiramatsu KI, Sakaki T, Okuchi K, Taoka T, Fujioka S . Hippocampal damage in the human brain after cardiac arrest. Cerebrovasc Dis. 2000; 10:2–7.CrossrefMedlineGoogle Scholar
  • 226. Tommasino C, Grana C, Lucignani G, Torri G, Fazio F . Regional cerebral metabolism of glucose in comatose and vegetative state patients. J Neurosurg Anesthesiol. 1995; 7:109–116.CrossrefMedlineGoogle Scholar
  • 227. Lovblad K, Senn P, Walpoth BH, Walpotth BN, Mattle HP, Radanov BP, Ozdoba C, Schroth G . Increased brain tolerance for ischemia in accidental deep hypothermia and circulatory arrest. Riv Neuroradiol1998; 11(SUPPL 2):224–226.CrossrefGoogle Scholar
  • 228. Edgren E, Enblad P, Grenvik A, Lilja A, Valind S, Wiklund L, Hedstrand U, Stjernstrom H, Persson L, Ponten U, Langstrom B . Cerebral blood flow and metabolism after cardiopulmonary resuscitation. A pathophysiologic and prognostic positron emission tomography pilot study. Resuscitation. 2003; 57:161–170.CrossrefMedlineGoogle Scholar
  • 229. Grubb NR, Fox KA, Smith K, Best J, Blane A, Ebmeier KP, Glabus MF, O'Carroll RE . Memory impairment in out-of-hospital cardiac arrest survivors is associated with global reduction in brain volume, not focal hippocampal injury. Stroke. 2000; 31:1509–1514.LinkGoogle Scholar
  • 230. Gut E, Fritz R, Leyhe T, et al . MRT after cerebral hypoxia. Correlation of imaging findings with clinical outcome and functional rehabilitation. Klin Neuroradiol. 1999; 9:147–152.CrossrefGoogle Scholar
  • 231. Els T, Kassubek J, Kubalek R, Klisch J . Diffusion-weighted MRI during early global cerebral hypoxia: a predictor for clinical outcome?Acta Neurol Scand. 2004; 110:361–367.CrossrefMedlineGoogle Scholar
  • 232. Kano H, Houkin K, Harada K, Koyanagi I, Nara S, Itou Y, Imaizumi H, Asai Y, Saitou M . Neuronal cell injury in patients after cardiopulmonary resuscitation: evaluation by diffusion-weighted imaging and magnetic resonance spectroscopy. Neurosurg Rev. 2006; 29:88–92.CrossrefMedlineGoogle Scholar
  • 233. Nogami K, Fujii M, Kato S, Nishizaki T, Suzuki M, Yamashita S, Oda Y, Sadamitsu D, Maekawa T . Analysis of magnetic resonance imaging (MRI) morphometry and cerebral blood flow in patients with hypoxic-ischemic encephalopathy. J Clin Neurosci. 2004; 11:376–380.CrossrefMedlineGoogle Scholar
  • 234. Wijdicks EF, Campeau NG, Miller GM . MR imaging in comatose survivors of cardiac resuscitation. AJNR Am J Neuroradiol. 2001; 22:1561–1565.MedlineGoogle Scholar
  • 235. Wijman CA, Mlynash M, Caulfield AF, Hsia AW, Eyngorn I, Bammer R, Fischbein N, Albers GW, Moseley M . Prognostic value of brain diffusion-weighted imaging after cardiac arrest. Ann Neurol. 2009; 65:394–402.CrossrefMedlineGoogle Scholar
  • 236. Wu O, Sorensen AG, Benner T, Singhal AB, Furie KL, Greer DM . Comatose patients with cardiac arrest: predicting clinical outcome with diffusion-weighted MR imaging. Radiology. 2009; 252:173–181.CrossrefMedlineGoogle Scholar
  • 237. Arbelaez A, Castillo M, Mukherji SK . Diffusion-weighted MR imaging of global cerebral anoxia. AJNR Am J Neuroradiol. 1999; 20:999–1007.MedlineGoogle Scholar
  • 238. Barrett KM, Freeman WD, Weindling SM, Brott TG, Broderick DF, Heckman MG, Crook JE, Divertie GD, Meschia JF . Brain injury after cardiopulmonary arrest and its assessment with diffusion-weighted magnetic resonance imaging. Mayo Clin Proc. 2007; 82:828–835.CrossrefMedlineGoogle Scholar
  • 239. Berek K, Lechleitner P, Luef G, Felber S, Saltuari L, Schinnerl A, Traweger C, Dienstl F, Aichner F . Early determination of neurological outcome after prehospital cardiopulmonary resuscitation. Stroke. 1995; 26:543–549.LinkGoogle Scholar
  • 240. Iida K, Satoh H, Arita K, Nakahara T, Kurisu K, Ohtani M . Delayed hyperemia causing intracranial hypertension after cardiopulmonary resuscitation. Crit Care Med. 1997; 25:971–976.CrossrefMedlineGoogle Scholar
  • 241. Ettl A, Felber S, Birbamer G, Daxer A . Corticol blindness following cerebral hypoxia. Proton nuclear magnetic resonance imaging and spectroscopy observations. Neuroophthalmology. 1994; 14:259–263.CrossrefGoogle Scholar
  • 242. Greer DM . MRI in anoxic brain injury. Neurocrit Care. 2004; 1:213–215.CrossrefMedlineGoogle Scholar
  • 243. Kuoppamaki M, Bhatia KP, Quinn N . Progressive delayed-onset dystonia after cerebral anoxic insult in adults. Mov Disord. 2002; 17:1345–1349.CrossrefMedlineGoogle Scholar
  • 244. Arishima H, Ishii H, Kubota T, Maeda H, Shigemori K . [Angiographic features of anoxic encephalopathy in the acute phase: a case report]. No To Shinkei. 2003; 55:977–982.MedlineGoogle Scholar
  • 245. Verslegers W, Crols R, van den Kerchove M, de Potter W, Appel B, Lowenthal A . Parkinsonian syndrome after cardiac arrest: radiological and neurochemical changes. Clin Neurol Neurosurg. 1988; 90:177–179.CrossrefMedlineGoogle Scholar
  • 246. Bolouri MR, Small GA . Neuroimaging of hypoxia and cocaine-induced hippocampal stroke. J Neuroimaging. 2004; 14:290–291.CrossrefMedlineGoogle Scholar
  • 247. Fujioka M, Okuchi K, Sakaki T, Hiramatsu K, Miyamoto S, Iwasaki S . Specific changes in human brain following reperfusion after cardiac arrest. Stroke. 1994; 25:2091–2095.LinkGoogle Scholar
  • 248. Hung GU, Lee JD, Lee JK . Bilateral cranial Tc-99m MDP uptake due to hypoxic-ischemic encephalopathy. Clin Nucl Med. 2007; 32:328–329.CrossrefMedlineGoogle Scholar
  • 249. Johkura K, Naito M . Wernicke's encephalopathy-like lesions in global cerebral hypoxia. J Clin Neurosci. 2008; 15:318–319.CrossrefMedlineGoogle Scholar
  • 250. Konaka K, Miyashita K, Naritomi H . Changes in diffusion-weighted magnetic resonance imaging findings in the acute and subacute phases of anoxic encephalopathy. J Stroke Cerebrovasc Dis. 2007; 16:82–83.CrossrefMedlineGoogle Scholar
  • 251. Singhal AB, Topcuoglu MA, Koroshetz WJ . Diffusion MRI in three types of anoxic encephalopathy. J Neurol Sci. 2002; 196(1–2):37–40.CrossrefMedlineGoogle Scholar
  • 252. Wartenberg KE, Patsalides A, Yepes MS . Is magnetic resonance spectroscopy superior to conventional diagnostic tools in hypoxic-ischemic encephalopathy?J Neuroimaging. 2004; 14:180–186.CrossrefMedlineGoogle Scholar
  • 253. Zhang YX, Liu JR, Jiang B, Liu HQ, Ding MP, Song SJ, Zhang BR, Zhang H, Xu B, Chen HH, Wang ZJ, Huang JZ . Lance-Adams syndrome: a report of two cases. J Zhejiang Univ Sci. 2007; 8:715–720.CrossrefGoogle Scholar
  • 254. Choi SP, Park HK, Park KN, Kim YM, Ahn KJ, Choi KH, Lee WJ, Jeong SK . The density ratio of grey to white matter on computed tomography as an early predictor of vegetative state or death after cardiac arrest. Emerg Med J. 2008; 25:666–669.CrossrefMedlineGoogle Scholar
  • 255. De Reuck J, Decoo D, Vienne J, Strijckmans K, Lemahieu I . Significance of white matter lucencies in posthypoxic-ischemic encephalopathy: comparison of clinical status and of computed and positron emission tomographic findings. Eur Neurol. 1992; 32:334–339.CrossrefMedlineGoogle Scholar
  • 256. Inoue Y, Shiozaki T, Irisawa T, Mohri T, Yoshiya K, Ikegawa H, Tasaki O, Tanaka H, Shimazu T, Sugimoto H . Acute cerebral blood flow variations after human cardiac arrest assessed by stable xenon enhanced computed tomography. Curr Neurovasc Res. 2007; 4:49–54.CrossrefMedlineGoogle Scholar
  • 257. Nunes B, Pais J, Garcia R, Magalhaes Z, Granja C, Silva MC . Cardiac arrest: long-term cognitive and imaging analysis. Resuscitation. 2003; 57:287–297.CrossrefMedlineGoogle Scholar
  • 258. Yanagawa Y, Un-no Y, Sakamoto T, Okada Y . Cerebral density on CT immediately after a successful resuscitation of cardiopulmonary arrest correlates with outcome. Resuscitation. 2005; 64:97–101.CrossrefMedlineGoogle Scholar
  • 259. Della Corte F, Barelli A, Giordano A, Iacobucci T, Valente MR, Pennisi MA . CBF determination in post-ischemic-anoxic comatose patients. Minerva Anestesiol. 1993; 59:637–641.MedlineGoogle Scholar
  • 260. Kjos BO, Brant-Zawadzki M, Young RG . Early CT findings of global central nervous system hypoperfusion. AJR. 1983; 141:1227–1232.CrossrefMedlineGoogle Scholar
  • 261. Morimoto Y, Kemmotsu O, Kitami K, Matsubara I, Tedo I . Acute brain swelling after out-of-hospital cardiac arrest: pathogenesis and outcome. Crit Care Med. 1993; 21:104–110.CrossrefMedlineGoogle Scholar
  • 262. Torbey MT, Geocadin R, Bhardwaj A . Brain arrest neurological outcome scale (BrANOS): predicting mortality and severe disability following cardiac arrest. Resuscitation. 2004; 63:55–63.CrossrefMedlineGoogle Scholar
  • 263. Torbey MT, Selim M, Knorr J, Bigelow C, Recht L . Quantitative analysis of the loss of distinction between gray and white matter in comatose patients after cardiac arrest. Stroke. 2000; 31:2163–2167.LinkGoogle Scholar
  • 264. Imaizumi H, Tsuruoka K, Ujike Y, Kaneko M, Namiki A . [Hypoxic brain damage after prolonged cardiac arrest during anesthesia–changes in CT and serum NSE concentration]. Masui. 1994; 43:1256–1260.MedlineGoogle Scholar
  • 265. Kelsen J, Obel A . Images in clinical medicine. Fatal cerebral hypoxemia after cardiac arrest. N Engl J Med. 2003; 348:817.CrossrefMedlineGoogle Scholar
  • 266. Schwab SA, Richter G, Bautz WA, Uder M, Alibek S . [Hypoxic injury of all deep nuclei of the brain–a case report from computed tomography]. Rontgenpraxis. 2008; 56:245–248.CrossrefMedlineGoogle Scholar
  • 267. Tanaka H, Masugata H, Fukunaga R, Mandai K, Sueyoshi K, Abe H . Sequential change of heterogeneous cerebral blood blow patterns after diffuse brain ischemia. Resuscitation. 1992; 24:273–281.CrossrefMedlineGoogle Scholar
  • 268. Inamasu J, Miyatake S, Tomioka H, Suzuki M, Nakatsukasa M, Maeda N, Ito T, Arai K, Komura M, Kase K, Kobayashi K . Subarachnoid haemorrhage as a cause of out-of-hospital cardiac arrest: a prospective computed tomography study. Resuscitation. 2009; 80:977–980.CrossrefMedlineGoogle Scholar
  • 269. Naples R, Ellison E, Brady WJ . Cranial computed tomography in the resuscitated patient with cardiac arrest. Am J Emerg Med. 2009; 27:63–67.CrossrefMedlineGoogle Scholar
  • 270. Heckmann JG, Lang CJ, Pfau M, Neundorfer B . Electrocerebral silence with preserved but reduced cortical brain perfusion. Eur J Emerg Med. 2003; 10:241–243.CrossrefMedlineGoogle Scholar
  • 271. Grubb NR, Simpson C, Sherwood RA, Abraha HD, Cobbe SM, O'Carroll RE, Deary I, Fox KA . Prediction of cognitive dysfunction after resuscitation from out-of-hospital cardiac arrest using serum neuron-specific enolase and protein S-100. Heart. 2007; 93:1268–1273.CrossrefMedlineGoogle Scholar
  • 272. Reisinger J, Hollinger K, Lang W, Steiner C, Winter T, Zeindlhofer E, Mori M, Schiller A, Lindorfer A, Wiesinger K, Siostrzonek P . Prediction of neurological outcome after cardiopulmonary resuscitation by serial determination of serum neuron-specific enolase. Eur Heart J. 2007; 28:52–58.CrossrefMedlineGoogle Scholar
  • 273. Prohl J, Rother J, Kluge S, de Heer G, Liepert J, Bodenburg S, Pawlik K, Kreymann G . Prediction of short-term and long-term outcomes after cardiac arrest: a prospective multivariate approach combining biochemical, clinical, electrophysiological, and neuropsychological investigations. Crit Care Med. 2007; 35:1230–1237.CrossrefMedlineGoogle Scholar
  • 274. Rech TH, Vieira SR, Nagel F, Brauner JS, Scalco R . Serum neuron-specific enolase as early predictor of outcome after in-hospital cardiac arrest: a cohort study. Crit Care. 2006; 10:R133.CrossrefMedlineGoogle Scholar
  • 275. Pfeifer R, Borner A, Krack A, Sigusch HH, Surber R, Figulla HR . Outcome after cardiac arrest: predictive values and limitations of the neuroproteins neuron-specific enolase and protein S-100 and the Glasgow Coma Scale. Resuscitation. 2005; 65:49–55.CrossrefMedlineGoogle Scholar
  • 276. Meynaar IA, Straaten HM, van der Wetering J, Verlooy P, Slaats EH, Bosman RJ, van der Spoel JI, Zandstra DF . Serum neuron-specific enolase predicts outcome in post-anoxic coma: a prospective cohort study. Intensive Care Med. 2003; 29:189–195.CrossrefMedlineGoogle Scholar
  • 277. Zingler VC, Krumm B, Bertsch T, Fassbender K, Pohlmann-Eden B . Early prediction of neurological outcome after cardiopulmonary resuscitation: a multimodal approach combining neurobiochemical and electrophysiological investigations may provide high prognostic certainty in patients after cardiac arrest. Eur Neurol. 2003; 49:79–84.CrossrefMedlineGoogle Scholar
  • 278. Rosen H, Sunnerhagen KS, Herlitz J, Blomstrand C, Rosengren L . Serum levels of the brain-derived proteins S-100 and NSE predict long-term outcome after cardiac arrest. Resuscitation. 2001; 49:183–191.CrossrefMedlineGoogle Scholar
  • 279. Schoerkhuber W, Kittler H, Sterz F, Behringer W, Holzer M, Frossard M, Spitzauer S, Laggner AN . Time course of serum neuron-specific enolase. A predictor of neurological outcome in patients resuscitated from cardiac arrest. Stroke. 1999; 30:1598–1603.LinkGoogle Scholar
  • 280. Fogel W, Krieger D, Veith M, Adams HP, Hund E, Storch-Hagenlocher B, Buggle F, Mathias D, Hacke W . Serum neuron-specific enolase as early predictor of outcome after cardiac arrest. Crit Care Med. 1997; 25:1133–1138.CrossrefMedlineGoogle Scholar
  • 281. Martens P, Raabe A, Johnsson P . Serum S-100 and neuron-specific enolase for prediction of regaining consciousness after global cerebral ischemia. Stroke. 1998; 29:2363–2366.LinkGoogle Scholar
  • 282. Dauberschmidt R, Zinsmeyer J, Mrochen H, Meyer M . Changes of neuron-specific enolase concentration in plasma after cardiac arrest and resuscitation. Mol Chem Neuropathol. 1991; 14:237–245.CrossrefMedlineGoogle Scholar
  • 283. Tiainen M, Roine RO, Pettila V, Takkunen O . Serum neuron-specific enolase and S-100B protein in cardiac arrest patients treated with hypothermia. Stroke. 2003; 34:2881–2886.LinkGoogle Scholar
  • 284. Oksanen T, Tiainen M, Skrifvars MB, Varpula T, Kuitunen A, Castren M, Pettila V . Predictive power of serum NSE and OHCA score regarding 6-month neurologic outcome after out-of-hospital ventricular fibrillation and therapeutic hypothermia. Resuscitation. 2009; 80:165–170.CrossrefMedlineGoogle Scholar
  • 285. Rundgren M, Karlsson T, Nielsen N, Cronberg T, Johnsson P, Friberg H . Neuron specific enolase and S-100B as predictors of outcome after cardiac arrest and induced hypothermia. Resuscitation. 2009; 80:784–789.CrossrefMedlineGoogle Scholar
  • 286. Pelinka LE, Hertz H, Mauritz W, Harada N, Jafarmadar M, Albrecht M, Redl H, Bahrami S . Nonspecific increase of systemic neuron-specific enolase after trauma: clinical and experimental findings. Shock (Augusta, Ga.)2005; 24:119–123.CrossrefMedlineGoogle Scholar
  • 287. Adrie C, Haouache H, Saleh M, Memain N, Laurent I, Thuong M, Darques L, Guerrini P, Monchi M . An underrecognized source of organ donors: patients with brain death after successfully resuscitated cardiac arrest. Intensive Care Med. 2008; 34:132–137.CrossrefMedlineGoogle Scholar
  • 288. Ali AA, Lim E, Thanikachalam M, Sudarshan C, White P, Parameshwar J, Dhital K, Large SR . Cardiac arrest in the organ donor does not negatively influence recipient survival after heart transplantation. Eur J Cardiothorac Surg. 2007; 31:929–933.CrossrefMedlineGoogle Scholar
  • 289. Matsumoto CS, Kaufman SS, Girlanda R, Little CM, Rekhtman Y, Raofi V, Laurin JM, Shetty K, Fennelly EM, Johnson LB, Fishbein TM . Utilization of donors who have suffered cardiopulmonary arrest and resuscitation in intestinal transplantation. Transplantation. 2008; 86:941–946.CrossrefMedlineGoogle Scholar
  • 290. Mercatello A, Roy P, Ng-Sing K, Choux C, Baude C, Garnier JL, Colpart JJ, Finaz J, Petit P, Moskovtchenko JF, et al . Organ transplants from out-of-hospital cardiac arrest patients. Transplant Proc. 1988; 20: 749–750.MedlineGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.