Skip to main content
Erschienen in: BMC Anesthesiology 1/2015

Open Access 01.12.2015 | Research article

Anion gap as a prognostic tool for risk stratification in critically ill patients – a systematic review and meta-analysis

verfasst von: Stella Andrea Glasmacher, William Stones

Erschienen in: BMC Anesthesiology | Ausgabe 1/2015

Abstract

Background

Lactate concentration is a robust predictor of mortality but in many low resource settings facilities for its analysis are not available. Anion gap (AG), calculated from clinical chemistry results, is a marker of metabolic acidosis and may be more easily obtained in such settings. In this systematic review and meta-analysis we investigated whether the AG predicts mortality in adult patients admitted to critical care settings.

Methods

We searched Medline, Embase, Web of Science, Scopus, The Cochrane Library and regional electronic databases from inception until May 2016. Studies conducted in any clinical setting that related AG to in-hospital mortality, in-intensive care unit mortality, 31-day mortality or comparable outcome measures were eligible for inclusion. Methodological quality of included studies was assessed using the Quality in Prognostic Studies tool. Descriptive meta-analysis was performed and the I2 test was used to quantify heterogeneity. Subgroup analysis was undertaken to identify potential sources of heterogeneity between studies.

Results

Nineteen studies reporting findings in 12,497 patients were included. Overall, quality of studies was poor and most studies were rated as being at moderate or high risk of attrition bias and confounding. There was substantial diversity between studies with regards to clinical setting, age and mortality rates of patient cohorts. High statistical heterogeneity was found in the meta-analyses of area under the ROC curve (I2 = 99 %) and mean difference (I2 = 97 %) for the observed AG. Three studies reported good discriminatory power of the AG to predict mortality and were responsible for a large proportion of statistical heterogeneity. The remaining 16 studies reported poor to moderate ability of the AG to predict mortality. Subgroup analysis suggested that intravenous fluids affect the ability of the AG to predict mortality.

Conclusion

Based on the limited quality of available evidence, a single AG measurement cannot be recommended for risk stratification in critically ill patients. The probable influence of intravenous fluids on AG levels renders the AG an impractical tool in clinical practice. Future research should focus on increasing the availability of lactate monitoring in low resource settings.

PROSPERO registration number

CRD42015015249. Registered on 4th February 2015.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12871-016-0241-y) contains supplementary material, which is available to authorized users.
Abkürzungen
AG
Anion gap
APACHE II
Acute physiology and chronic health evaluation ii
AUC
Area under the ROC curve
AVPU
Alert, verbal, pain, unresponsive
CIs
Confidence intervals
DKA
Diabetic ketoacidosis
ENT
Ear nose and throat
GCS
Glasgow coma scale
ICCU
Intensive cardiac care unit
ICU
Intensive care unit
IQR
Interquartile range
ISS
Injury severity score
Mdn
Median
MI
Myocardial infarction
OR
Odds ratio
PCI
Percutaneous coronary intervention
PRISMA
Preferred reporting items for systematic reviews and meta-analyses
Pro
Prospective
QUIPS
Quality in prognostic studies
Retro
Retrospective
SAPS
Simplified acute physiology score
SD
Standard deviation
SE
Standard error
SOFA
Sequential organ failure assessment
STEMI
ST-elevation myocardial infarction

Background

Much research has focussed on the prognostic value of serum lactate estimation in critically ill patients [1]; however, in the context of work in low resource settings we have noted that facilities for lactate and blood gas analysis are frequently not available, prompting a search for alternative risk stratification tools. The anion gap (AG) is an easily calculated marker of metabolic acidosis based on analytes typically available from routine chemistry analysis. It may have potential as a risk stratification tool to identify sick patients at risk of deterioration, who would benefit from further management whilst pathophysiological processes are still reversible. The AG reflects the concentration of unmeasured anions as calculated by the formula Na+ - (Cl + HCO3). Inclusion of potassium in the formula is recommended where its concentration is abnormally high or low [2]. In healthy subjects, the unmeasured anions or “gap” is mostly made up of albumin; however, hypoalbuminaemia, commonly observed in critically ill patients, can lower the AG and mask an acidosis. Feldman and colleagues therefore recommended that the AG should be corrected for albumin [3].
In metabolic acidosis, addition of fixed acids leads to a rise of the AG: while the proton within the acid combines with bicarbonate, the conjugate base contributes to the unmeasured anions. Metabolic acidosis is common in critically ill patients and is a strong predictor of prognosis [4]. Maciel and Park observed that unmeasured anions accounted for the majority of metabolic acidosis in both intensive care unit (ICU) survivors and non-survivors, whereas lactate accounted for only a quarter of acidosis [5]. AG may thus have potential as a risk stratification tool, especially if corrected for albumin.
The validity of the AG as a predictor of mortality has been studied and has been compared to other indices of acid–base balance, especially Stewart’s strong ion gap [6]. However, the strong ion gap is more cumbersome and expensive to measure than the AG and is thus less suitable as a risk stratification tool in low resource settings. In studies with contrasting findings, AG was noted to be a very strong predictor of mortality [7] or of limited value with neither the AG nor the strong ion gap effective as predictors of in-hospital mortality [8]. Furthermore, it has been noted that studies conducted in countries where gelatin-based intravenous fluids are routinely used, such as the UK and Australia, failed to show an association between the strong ion gap and mortality whereas studies conducted in settings where such fluids are not routinely used, especially the USA were able to demonstrate an association [9, 10]. Gelatins are an exogenous source of unmeasured anions [11] and an increase in AG after gelatin infusion has been demonstrated in animal experimental studies [12].
Recently, a large study of 18,985 patients found that ∆AG, defined as the difference in AG between pre-hospital admission and critical care admission, was a robust predictor of all-cause mortality, where the pre-hospital AG was determined between seven and 365 days before admission [13]. However, this approach requires adequate documentation and a laboratory database, which are unlikely to be available in resource-limited settings.
In the present systematic review and meta-analysis we therefore aimed to determine the validity of a single AG measurement as a risk stratification tool predicting 31-day mortality in-hospital mortality, in-ICU mortality and comparable outcome measures in adult patients admitted to critical care settings. We also aimed to compare the prognostic validity of the observed and albumin-corrected AG. Although the AG as a risk stratification tool would be mainly applicable to low income countries, this systematic review and meta-analysis does not limit itself to studies conducted in such countries as the main focus lies on the scientific validity of the AG as a risk stratification tool.

Methods

Protocol registration

This systematic review and meta-analysis adheres to the “preferred reporting items for systematic reviews and meta-analyses” (PRISMA) standards [14]. A protocol was registered with PROSPERO, registration number CRD42015015249.

Search strategy, study selection and data extraction

We searched the electronic databases of Medline, Embase, Scopus, Medion, The Cochrane Library, Web of Science and regional bibliographic databases including African Index Medicus, Latin America and the Caribbean (LILACS), IndMed, Index Medicus for South East Asia Region (IMSEAR) and Western Pacific Region Index Medicus (WPRIM). In addition, journals specialising in the fields of critical care, anaesthetics, emergency medicine and intensive care medicine were searched electronically. Searches were performed for studies that were conducted on humans and published in English, German or French using the search terms “anion gap”, “unmeasured anions” and “unidentified acids”. The initial search was performed in January 2015 and the search was subsequently updated in May 2016. All search results were initially screened by abstract and title and those considered relevant subsequently underwent full-text screening. To identify further relevant studies, reference lists were reviewed, citation searches were performed and citation alerts were set up for all articles considered relevant after full-text screening.
Studies, conducted in any acute care clinical setting, were eligible for inclusion if they were published within the last 15 years, reported measurement of the observed and/or corrected serum AG in adult patients and mortality defined as “in-hospital mortality”, “in ICU mortality” or, if a time-frame was stated, death within up to 31-days of hospital admission. The latter definition was chosen where both outcomes were reported within a single study. Case studies, case–control studies and studies whose main focus was a hyperglycaemic emergency, poisoning or renal failure were excluded.
Data extraction was performed by a single reviewer, SG. A second reviewer, WS, independently extracted data from 10 % of the studies selected using a random number generator. Corresponding authors were contacted where necessary to discuss missing or unclear data.

Assessment of methodological quality and risk of bias

Two reviewers (SG and WS) independently graded the methodological quality and risk of bias of included studies using a modified version of the Quality In Prognostic Studies (QUIPS) tool [15]. This tool assesses the risk of bias of prognostic studies in six domains: study participation, study attrition, prognostic factor measurement, outcome measurement, confounding, and statistical analysis and reporting. Each study was rated as being at high, moderate or low risk of bias in each domain. Disagreements were resolved by discussion between the two reviewers.

Statistical analysis

Area under the ROC curve (AUCs), odds ratios (ORs) and mean differences were pooled in random or fixed effects generic inverse variance models for the observed and corrected AG. The I2 test was used to quantify heterogeneity. A fixed-effects model was used where the I2 was below 30 %; otherwise, a random-effects model was used. Meta-analysis of ORs and mean differences was undertaken in Review Manager version 5.3 (The Cochrane Collaboration, 2014, Copenhagen), while AUCs were pooled in StatsDirect version 2.8.0 (England: StatsDirect Ltd. 2013); an AUC of ≥0.8 was considered to denote good discriminatory power. Pooled estimates were not presented in forest plots due to high heterogeneity in the meta-analyses of AUC and mean difference; in the results section the pooled estimates are reported together with their respective 95 % confidence intervals (CIs). Subgroup analysis was undertaken to assess whether heterogeneity between studies could be explained by the following study characteristics: patient age, study setting, quantity of intravenous fluids received, determination of the AG before the initiation of hospital-based treatment, the routine use of gelatin-based intravenous fluids in the study country, choice of outcome measure, publication date and overall mortality.
Statistical significance testing for subgroup differences employed the unpaired t-test in GraphPad® QuickCalc Web Calculator (La Jolla California USA) [16]. Probabilities were two-tailed and a probability of less than 0.05 was considered statistically significant. No adjustments were made for multiple comparisons. Sensitivity analysis was undertaken to assess the effect of including retrospective studies and studies at high risk of attrition bias in the meta-analysis. Funnel plots were visually inspected for evidence of publication bias.

Results

Study selection and characteristics of included studies

The study selection process is summarised in Fig. 1. In total, the search yielded 2688 non-duplicate publications; 2630 articles were excluded after title and abstract screening thus 58 articles were retrieved in full-text. Twenty-nine articles were excluded during full-text screening, leaving 29 studies that were subjected to data extraction. Ten studies were excluded during data extraction and thus 19 studies were included in the systematic review, of which 18 were included in one or more quantitative syntheses.
Table 1 illustrates the characteristics of included studies. A majority of studies were conducted in high income countries [7, 8, 1626] while three studies were conducted in middle income countries [2729] and one in a low income country [30]. Studies were conducted in the following settings: ICU (10 studies), trauma centre (5 studies), coronary care unit/intensive cardiac care unit (3 studies) and Accident and Emergency department (1 study). Five studies accounted for the effect of intravenous fluids on AG levels: in one study no patient received more than 400 ml of any intravenous fluid before the AG was measured [7], in two studies patients receiving more than 250 ml or 500 ml of intravenous fluids respectively were excluded from the analysis [16, 30]. Two studies stated that the AG was determined before hospital based management, including intravenous fluids, was initiated [21, 29]. The remaining studies did not report on the quantity of intravenous fluids received by their study cohorts. No study reported both in-hospital and a time-frame specific mortality. One study [29] failed to define the outcome measure, reporting it as “mortality”. Sensitivity analysis was carried out to determine whether inclusion of this study affected the pooled effect measure. The risks of bias ratings are displayed in Table 2. Risk of attrition bias and confounding were the most poorly rated domains. There were no disagreements between review authors on data extraction.
Table 1
Characteristics of included studies
First author/year
Nr
Country
Setting and most frequent reasons for admission
Study design
Age (mean or mdn)
Sample size
Male (%)
Outcome (mortality)
Total mortality (%)
Severity of illness (mean score ± SD or mdn and range or IQR)
Antonini 2008 [17]
1
Italy
General ICU admissions: 36 % trauma; 26 % cerebrovascular disease; 14 % sepsis
Pro
Mean: 53
136
71
28-day
27
SOFA: 6 (range 0–18)
SAPS II: 40 (range 6–76)
Attanà 2013 [18]
2
Italy
STEMI patients with persistent cardiogenic shock after primary PCI admitted to ICCU
Pro
Mean: 73
63
62
In-ICCU
49
APACHE II: 20.6 ± 12.4
Boniatti 2011 [27]
3
Brazil
General ICU admissions: 64 % medical admissions; 27 % sepsis; 24 % elective surgery; 12 % emergency surgery
Pro
Mean: 56
175
53
In-hospital
37
APACHE II: 20.8 ± 8.0
SOFA score: 6.2 ± 3.8
Cusack 2002 [19]
4
UK
General ICU admissions: 17 % respiratory failure; 11 % post-cardiac arrest; 8 % trauma
Pro
Mean: 61
100
NA
28-day
31
APACHE II: 20.5
Dondorp 2004 [29]
5
Vietnam
Patients with severe falciparum malaria admitted to ICU
Pro
Mdn: 31
268
80
Not defined
17
GCS < 11: 51 %
8 % Haemodynamic shocka
Dubin 2007 [37]
6
Argentina
General ICU admissions: 56 % medical admissions; 35 % elective surgery; 9 % emergency surgery
Pro
Mean: 65
935
49
30-day
11
APACHE II: 13 ± 7
SOFA: 3 ± 3
FitzSullivan 2005 [20]
7
USA
Trauma ICU admissions: 60 % blunt trauma
Retro
Mean: 36
3102
81
In-hospital
17
APACHE II: 26.1 ± 10.5
ISS: 20.4 ± 12.9
Hucker 2005 [21]
8
UK
A&E admissions: 46 % medical admissions; 17 % elderly care; 16 % discharged
Pro
Mean: 67
672
NA
In-hospital
12
93 % alert on AVPU scale
Kaplan 2004 [7]
9
USA
Trauma patients requiring vascular repair of torso or extremities, trauma centre: 83 % penetrating trauma
Retro
Mean: 32
282
NA
28-day
23
ISS: 15.8 ± 11.0
Kaplan 2008 [16]
10
USA
Major trauma patients, trauma centre: 59 % blunt trauma
Retro
Mean: 33
78
44
28-day in hospital
33
ISS: 8.9 ± 7.3
Lazzeri 2010 [22]
11
Italy
STEMI patients admitted to ICCU at tertiary centre undergoing primary PCI
Pro
Mdn: 67
445
75
In-hospital
10
92 % Killip class I-II
8 % Killip class II-IV
41 % complications in ICCU
Lipnick 2013 [13]
12
USA
General ICU admissions: 57 % medical; 44 % surgical; 16 % sepsis
Retro
Mean: 65
664
55
30-day
15
33 % no organ failure
53 % 1–2 organs failed
14 %% ≥ 3 organs failedb
Martin 2013 [23]
13
Germany
Surgical ICU admissions: 17 % maxillofacial surgery; 13 % ENT; 12 % neurosurgery
Retro
Mean: 59
1551
54
In-hospital
9
Average length of stay in ICU: 4.2 days
Martin 2005a [25]
14
USA
Surgical ICU admissions: 56 % abdominal; 18 % vascular; 10 % thoracic
Retro
Mean: 52
2291
61
In-ICU
8
APACHE II: 21.8 ± 9.7
SAPS: 16.8 ± 8.8
Martin 2005b [24]
15
USA
Trauma patients, trauma centre: 65 % blunt trauma
Retro
Mean: 38
427
79
In-hospital
10
ISS: 23 ± 23
Novovic 2014 [28]
16
Serbia
ICU patients requiring mechanical ventilation
Retro
Mean: 60
142
47
28-day
52
APACHE II: 16.2 ± 6.4
Rocktaeschel 2003 [8]
17
Australia
General ICU admissions: 91 % respiratory; 54 % gastrointestinal; 51 % cardiovascular
Retro
Mdn: 65
300
58
In-hospital
28
APACHE II: 17 (IQR 14 – 22)
Sahu 2006 [26]
18
USA
Patients with acute MI admitted to coronary care unit: 65 % STEMI
Retro
Mean: 63
773
62
In-hospital
11
5 % cardiogenic shock
Shane 2014 [30]
19
Uganda
Major trauma patients, trauma centre: 65 % road traffic accidents; 35 % assault
Pro
Mean: 26
93
81
In-hospital
34
ISS: 25.4 ± 8.3
APACHE II Acute Physiology and Chronic Health Evaluation, AVPU alert, verbal, pain, unresponsive, ENT ear, nose and throat, GCS Glasgow coma scale, ICCU intensive cardiac care unit, ICU intensive care unit, IQR interquartile range, ISS Injury Severity Score, Mdn median, MI myocardial infarction, PCI percutaneous coronary intervention, Pro prospective, Retro retrospective, SAPS simplified acute physiology score, SD standard deviation, SOFA sequential organ failure assessment, STEMI ST-elevation myocardial infarction
aBased on data of previously published original study including 346 patients [38]
bBased on data from entire study cohort of 18,995 patients
Table 2
Risk of bias rating
https://static-content.springer.com/image/art%3A10.1186%2Fs12871-016-0241-y/MediaObjects/12871_2016_241_Tab2_HTML.gif
Green, Yellow and Red refer to low, moderate and high risk of bias respectively

Prognostic ability of the AG to predict mortality

Owing to the high heterogeneity identified in the meta-analyses of AUC and mean difference, the pooled effect measures reported in this section should not be interpreted. Overall, three studies reported good discriminatory ability of the AG to predict mortality, of which two studies were included in meta-analysis [7, 16] and one study allowed the calculation of an OR for a specific AG threshold [22]. The former two studies were responsible for a large proportion of the statistical heterogeneity; both studies were conducted in young patients in the same trauma centre and only patients receiving less than a specified volume of intravenous fluids were included in the analysis. The latter study was conducted in patients with ST-elevation myocardial infarction undergoing percutaneous coronary intervention. The remaining 16 studies reported poor to moderate ability of the AG to predict mortality.
Nine studies reported AUCs for the observed AG (Fig. 2). Meta-analysis yielded a summary AUC of 0.72 (95 % CI 0.59 to 0.86). Heterogeneity was very high (I2 = 99 %) but reduced to I2 = 68 % when the two studies by Kaplan and Kellum were excluded from the analysis [7, 16]. Six studies reported AUCs for the corrected AG (Additional file 1: Figure S1). The summary AUC was estimated as 0.67 (95 % CI 0.62 to 0.71) and heterogeneity was high (I2 = 67 %).
Six studies reported ORs derived by logistic regression modelling for the observed AG (Fig. 3); five reported univariate logistic regression ORs whilst one study reported an OR adjusted for age. The summary OR was 1.08 (95 % CI 1.06 to 1.11); results were homogenous (I2 = 0 %) but it should be noted that the two studies by Kaplan and Kellum were not included in this analysis as neither study reported the OR. Four studies reported ORs derived by univariate logistic regression for the corrected AG (Additional file 2: Figure S2). The summary OR was 1.10 (95 % CI 1.07 to 1.13) and heterogeneity was very low (I2 = 5 %). Data reported in the study of Lazzeri and colleagues allowed the calculation of an OR for a specified AG positivity threshold. This yielded an OR of 2.8 (95 % CI 1.5 to 5.5) for an AG positivity threshold of 11 mEq/L [22].
Mean difference was the most frequently reported effect measure with ten studies reporting it for the observed AG (Fig. 4). The summary mean difference was 3.55 mEq/L (95 % CI 1.08 to 6.02). Heterogeneity was high (I2 = 97 %); however, excluding the study by Kaplan and Kellum [7] completely eliminated heterogeneity (I2 = 0 %). The mean difference for corrected AG was reported by three studies (Additional file 3: Figure S3) and the summary mean difference was estimated as 3.25 mEq/L (95 % CI 1.53 to 4.96) with homogeneous results (I2 = 0 %).
Sensitivity analysis showed that including retrospective studies and studies at risk of attrition bias did not affect the summary AUC. Including prospective studies only (six studies) yielded a summary AUC of 0.73 (95 % CI 0.69 to 0.78). Similarly, excluding studies rated at high risk of attrition bias (four studies) yielded a summary AUC of 0.75 (95 % CI 0.54 to 0.96). Excluding the study by Dondorp and colleagues [31] with an undefined outcome measure (“mortality”) yielded an AUC of 0.72 (95 % CI 0.56 to 0.88).
AUC of observed AG was chosen for subgroup analysis; results are shown in Table 3. The quantity of intravenous fluids given to a patient had the strongest influence on the summary AUC. Studies excluding patients who received more than a specified volume of intravenous fluids [7, 16] reported a significantly higher summary AUC than studies not excluding patients for this reason (P = 0.0008), but heterogeneity remained high in both subgroups. For the subsequent analysis, studies restricting intravenous fluids [7, 16] and studies measuring the AG before initiation of hospital-based management [21, 29] were combined in a subgroup and compared to studies that did not account in any way for the effect of intravenous fluids on the AG. The former subgroup yielded a significantly higher summary AUC than the latter subgroup (P <0.0001). Heterogeneity remained high in the former subgroup (I2 = 98 %), though results in the latter subgroup were homogenous (I2 = 0 %). The summary AUC of studies conducted in countries where gelatin-based resuscitation fluids are routinely used [8, 19, 28] is not significantly different to that of studies conducted in countries where gelatins are not routinely used [13, 24] (P = 0.33). Studies in which intravenous fluids were restricted or in which the AG was measured before initiation of hospital-based treatment were not included in the latter comparison. The observed AG appears to be a slightly better predictor of mortality among younger patients (P = 0.011) and those admitted to trauma centre settings (P = 0.0235). Subgroup analysis showed no significant difference between studies reporting in-hospital mortality and those reporting a time-framed mortality (P = 0.65); similarly, no significant difference was found between studies in which overall mortality was below 30 % and above 30 % (P = 0.89) or between studies published before and after the year 2005 (P = 0.43); and heterogeneity remained high in all subgroups. Funnel plots did not show evidence of publication bias (Additional file 4: Figure S4 and Additional file 5: Figure S5).
Table 3
Results of subgroup analysis
Study characteristic
Groups
Studies (nr)
Total sample size
Pooled AUC (95 % CIs)
I2 test
P-value
Study setting
Trauma patients
9, 10, 15
787
0.83 (0.68, 0.99)
97 %
0.0235
ICU patients
4, 5, 12, 16, 17
1474
0.66 (0.59, 0.73)
69 %
Age
Mean/Median age 30–40 years
5, 9, 10, 15
1055
0.81 (0.69, 0.93)
97 %
0.0114
Mean/Median age 60–70 years
4, 8, 12, 16, 17
1878
0.66 (0.60, 0.71)
70 %
Intravenous fluids restriction
Restriction
9, 10
356
0.91 (0.8, 1.0)
95 %
0.0008
No restriction
4, 5, 8, 12, 15, 16, 17
2573
0.67 (0.62, 0.72)
68 %
Intravenous fluids restriction and AG measured before treatment initiation
Restriction and AG measurement before hospital treatment initiation
5, 8, 9, 10
1296
0.83 (0.73, 0.93)
98 %
<0.0001
No restriction or AG measured after treatment was commenced
4, 12, 15, 16, 17
1633
0.63 (0.60, 0.66)
0 %
Routine use of gelatin-based intravenous fluids in study country
Gelatins not routinely used
12, 15
1091
0.62 (0.58, 0.65)
0 %
0.3344
Gelatins routinely used
4, 16, 17
542
0.65 (0.6, 0.7)
0 %
Outcome measure
Time frame stated e.g. 31-day or 28-day mortality
4, 9, 10, 12, 16
1266
0.74 (0.52, 0.96)
99 %
0.6518
In-hospital mortality
8, 15, 17
1399
0.69 (0.64, 0.75)
44 %
Overall mortality in study population
Below 30 %
5, 8, 9, 12, 15, 17
2613
0.74 (0.55, 0.93)
99 %
0.8856
Above 30 %
4, 10, 16
320
0.70 (0.57, 0.84)
83 %
Date of publication
Before and including 2005
4, 5, 8, 9, 15
1710
0.76 (0.59, 0.94)
98 %
0.4325
2006 and after
10, 12, 16, 17
1184
0.67 (0.58, 0.77)
87 %
CIs confidence intervals, ICU intensive care unit

Discussion

This systematic review and meta-analysis does not support the use of a single AG measurement for risk stratification in critically ill patients. Quantitative synthesis was limited by significant statistical heterogeneity, which, following a series of subgroup analyses, could be partially explained by the quantity of intravenous fluids received by study patients. Studies differed substantially with regards to setting, presumed use of gelatin-based intravenous fluids as well as the age and mortality rate of their patient cohorts; however, in our analysis none of these factors fully accounted for the high degree of heterogeneity. Disease severity was not consistently characterised across studies and could therefore not be analysed in subgroup analysis. Overall, the high degree of unexplained heterogeneity, poor quality of primary studies and poor to moderate discriminatory power of the AG reported by the majority of studies suggest that there is insufficient evidence to recommend the use of the AG in clinical practice. Owing to the small number of studies that calculated a corrected AG, we were unable to determine whether correction of the AG for albumin improves its predictive ability.
In subgroup analysis, a highly statistically significant difference was seen between studies accounting for intravenous fluids by means of fluid restriction or by measuring the AG before the initiation of hospital-based management and studies that did not account for quantity of intravenous fluids by any means. This indicates that intravenous fluids may have blurred the association between AG and mortality. Administration of normal saline lowers the AG because addition of NaCl to the plasma increases the baseline chloride concentration proportionately more than the baseline sodium concentration, owing to the differences in volume of distribution between the two ions [4]. This effect may not be seen with more balanced fluids such as Hartman’s solution but, given that normal saline is commonly used in clinical practice, a risk stratification tool that is considerably affected by saline infusion is impractical. However, the validity of this subgroup analysis is limited by its observational nature and relatively small number of studies contained in each subgroup. Therefore, other confounders may have accounted for these results. Furthermore, the study by Shane and colleagues [30] also employed intravenous fluid restriction but found no predictive effect of the AG; however, only mean difference in AG between survivors and non-survivors but not AUC was reported. Further research would be required to determine more conclusively the extent to which the AG is affected by intravenous fluids.
Other than intravenous fluid restriction, our subgroup analysis did not identify factors accounting for the high statistical heterogeneity in the meta-analysis of AUC. A small effect of study setting and mean/median age on the pooled AUC was observed; however, the associated probabilities were >0.01 where no adjustments were made for multiple comparisons and in both analyses one subgroup contained the two studies by Kaplan and Kellum [7, 16]. Therefore, we consider the observed differences most likely to have arisen due to chance or confounding. Notably, no significant effect of outcome measure or publication date was observed on the pooled AUC. This supports the appropriateness of including studies reporting a time-framed mortality and in-hospital mortality and studies published at different times over the past 15 years. As parameters denoting disease severity were not consistently reported across studies, we divided studies according to overall mortality in subgroup analysis. No difference in pooled AUC was seen between studies reporting mortality rates above and below 30 %; however, overall mortality is a suboptimal indicator of severity of illness. Therefore, the contribution of disease severity to the observed statistical heterogeneity remains unclear.
Another important factor limiting the ability of the AG to predict clinical outcomes is its considerable baseline variability amongst healthy people. To address this, Kraut and Nagami suggested comparing the AG during an acute admission to the “personal AG” measured when the individual was in good health [2]. Dynamic AG indices, describing not only the magnitude of acid–base disturbance but also trends over time, are better predictors of mortality, as shown by the large study by Lipnick and colleagues including 18,995 patients [13]. For a subset of patients in this study (n = 664), the predictive ability of a single AG measurement was also reported and was shown to be poor (AUC 0.61). However, implementation of the systems required to support a “personal AG” would be challenging in low resource settings.
A rise in the AG in critically ill patients was long thought to be predominantly due to lactic acidosis, yet several studies reported poor sensitivity of the AG in detecting hyperlactataemia defined by a lactate threshold of 2.5 mmol/l [3134]. The AG was an excellent predictor of severe hyperlactataemia defined as lactate above 4 mmol/l [32] or 5 mmol/l [8]; however, Nichol and colleagues found that a higher lactate concentration even within a normal reference range of 2 mmol/l independently predicts mortality [35]. The AG may thus miss patients at risk of mortality, as a considerable degree of hyperlactataemia is required to push the AG outside its normal reference range if the baseline AG is low [2]. This is in keeping with the extreme difference in lactate levels between survivors and non-survivors observed in the study reporting the highest predictive value of AG [7]. Other studies reported smaller, albeit mostly statistically significant, differences in lactate levels between survivors and non-survivors.

Limitations

The methodological quality of primary studies was generally poor. Most studies were rated at moderate or high risk of attrition bias and sampling bias as a result of failure to quantify missing outcome or prognostic data, especially in retrospective studies, where case notes with missing information are less likely to have been available. This may have affected our results where information was missing in a non-random manner. Similarly, risk of confounding was moderate to high in the majority of studies. Only one study explored the influence of age on AG levels, and stratification was not employed by any study; the risk of confounding affecting the review outcome is therefore high. Several studies did not report relevant effect measures, such as OR, AUC or mean difference or failed to provide confidence intervals, leading to exclusion from this review. Furthermore, the overall severity of illness in the study cohort was sometimes not quantified by means of an accepted disease severity score, such as the Acute Physiology and Chronic Health Evaluation II (APACHE II), Sequential Organ Failure Assessment (SOFA) score or Injury Severity Score (ISS) in trauma patients. No studies reported short-term mortality outcomes, which may have been more appropriate as naturally the prognosis in critical care patients is heavily influenced by clinical interventions undertaken during the inpatient stay. Shapiro and colleagues found that a single lactate level drawn on admission has good discriminatory power to predict 3-day mortality (AUC = 0.8) but poor discriminatory power to predict 28-day mortality (AUC = 0.67) [36]. Lastly, few studies stated the types of intravenous fluids used and no study included the quantity of intravenous fluid used as a variable in multivariate analysis.

Conclusion

The high degree of unexplained statistical heterogeneity, considerable diversity between patient cohorts and poor quality of primary studies, in particular the high risk of attrition bias and confounding, impact on the overall strength of evidence of this systematic review and meta-analysis. The majority of studies reported here do not support the use of the AG as a predictor of 31-day mortality, in-ICU mortality or in-hospital mortality. Therefore, based on the available evidence, the use of a single AG measurement for risk stratification in critically ill patients cannot be recommended. Further high quality research would be required to conclusively determine the validity of the AG as a predictor of mortality. However, the probable influence of intravenous fluids on AG levels and the substantial baseline variability between AG levels among healthy individuals may render the use of the AG problematic in clinical practice. In light of the growing body of evidence supporting the use of lactate concentration for monitoring of critically ill patients, it may be more worthwhile to focus efforts on increasing the capacity for lactate measurement in low resource settings.

Acknowledgements

We would like to thank our University of St Andrews colleagues Dr Jennifer Burr for critically reviewing the protocol and results and Dr Ruth Cruickshank for critically reviewing the protocol.

Funding

No funding was obtained to undertake this systematic review and meta-analysis.

Availability of data and materials

This review is solely based on secondary data reported by published research studies.

Authors’ contribution

SG participated in study design, drafted the protocol, performed the literature search, performed the quality assessment, data extraction, data analysis and drafted manuscript. WS participated in study design, critically revised the protocol, performed the quality assessment, independently extracted data from 10 % of the studies and critically revised the manuscript. Both authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interest.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Kruse O, Grunnet N, Barfod C. Blood lactate as a predictor for in-hospital mortality in patients admitted acutely to hospital: a systematic review. Scand J Trauma Resusc Emerg Med. 2011;19:74.CrossRefPubMedPubMedCentral Kruse O, Grunnet N, Barfod C. Blood lactate as a predictor for in-hospital mortality in patients admitted acutely to hospital: a systematic review. Scand J Trauma Resusc Emerg Med. 2011;19:74.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Kraut JA, Nagami GT. The Serum Anion Gap in the Evaluation of Acid–base Disorders: What Are Its Limitations and Can Its Effectiveness Be Improved? Clin J Am Soc Nephrol. 2013;8:2018–24.CrossRefPubMedPubMedCentral Kraut JA, Nagami GT. The Serum Anion Gap in the Evaluation of Acid–base Disorders: What Are Its Limitations and Can Its Effectiveness Be Improved? Clin J Am Soc Nephrol. 2013;8:2018–24.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Feldman M, Soni N, Dickson B. Influence of hypoalbuminemia or hyperalbuminemia on the serum anion gap. J Lab Clin Med. 2005;146:317–20.CrossRefPubMed Feldman M, Soni N, Dickson B. Influence of hypoalbuminemia or hyperalbuminemia on the serum anion gap. J Lab Clin Med. 2005;146:317–20.CrossRefPubMed
4.
Zurück zum Zitat Gunnerson KJ. Clinical review: the meaning of acid–base abnormalities in the intensive care unit part I - epidemiology. Crit Care. 2005;9:508–16.CrossRefPubMedPubMedCentral Gunnerson KJ. Clinical review: the meaning of acid–base abnormalities in the intensive care unit part I - epidemiology. Crit Care. 2005;9:508–16.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Maciel AT, Park M. Unmeasured anions account for most of the metabolic acidosis in patients with hyperlactatemia. Clinics. 2007;62:55–62.CrossRefPubMed Maciel AT, Park M. Unmeasured anions account for most of the metabolic acidosis in patients with hyperlactatemia. Clinics. 2007;62:55–62.CrossRefPubMed
6.
Zurück zum Zitat Stewart PA. Modern quantitative acid–base chemistry. Can J Physiol Pharmacol. 1983;61:1444–61.CrossRefPubMed Stewart PA. Modern quantitative acid–base chemistry. Can J Physiol Pharmacol. 1983;61:1444–61.CrossRefPubMed
7.
Zurück zum Zitat Kaplan LJ, Kellum JA. Initial pH, base deficit, lactate, anion gap, strong ion difference, and strong ion gap predict outcome from major vascular injury. Crit Care Med. 2004;32:1120–4.CrossRefPubMed Kaplan LJ, Kellum JA. Initial pH, base deficit, lactate, anion gap, strong ion difference, and strong ion gap predict outcome from major vascular injury. Crit Care Med. 2004;32:1120–4.CrossRefPubMed
8.
Zurück zum Zitat Rocktaeschel J, Morimatsu H, Uchino S, Bellomo R. Unmeasured anions in critically ill patients: can they predict mortality? Crit Care Med. 2003;31:2131–6.CrossRefPubMed Rocktaeschel J, Morimatsu H, Uchino S, Bellomo R. Unmeasured anions in critically ill patients: can they predict mortality? Crit Care Med. 2003;31:2131–6.CrossRefPubMed
11.
Zurück zum Zitat Hayhoe M, Bellomo R, Liu G, McNicol L, Buxton B. The aetiology and pathogenesis of cardiopulmonary bypass-associated metabolic acidosis using polygeline pump prime. Intensive Care Med. 1999;25:680–5.CrossRefPubMed Hayhoe M, Bellomo R, Liu G, McNicol L, Buxton B. The aetiology and pathogenesis of cardiopulmonary bypass-associated metabolic acidosis using polygeline pump prime. Intensive Care Med. 1999;25:680–5.CrossRefPubMed
12.
Zurück zum Zitat Sumpelmann R, Schurholz T, Marx G, Thorns E, Zander R. Alteration of anion gap during almost total plasma replacement with synthetic colloids in piglets. Intensive Care Med. 1999;25:1287–90.CrossRefPubMed Sumpelmann R, Schurholz T, Marx G, Thorns E, Zander R. Alteration of anion gap during almost total plasma replacement with synthetic colloids in piglets. Intensive Care Med. 1999;25:1287–90.CrossRefPubMed
13.
Zurück zum Zitat Lipnick MS, Braun AB, Cheung JT, Gibbons FK, Christopher KB. The difference between critical care initiation anion gap and prehospital admission anion gap is predictive of mortality in critical illness. Crit Care Med. 2013;41:49–59.CrossRefPubMed Lipnick MS, Braun AB, Cheung JT, Gibbons FK, Christopher KB. The difference between critical care initiation anion gap and prehospital admission anion gap is predictive of mortality in critical illness. Crit Care Med. 2013;41:49–59.CrossRefPubMed
14.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151:264–9. W264.CrossRefPubMed Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151:264–9. W264.CrossRefPubMed
15.
Zurück zum Zitat Hayden JA, van der Windt DA, Cartwright JL, Cote P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med. 2013;158(4):280–6.CrossRefPubMed Hayden JA, van der Windt DA, Cartwright JL, Cote P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med. 2013;158(4):280–6.CrossRefPubMed
16.
Zurück zum Zitat Kaplan LJ, Kellum JA. Comparison of acid–base models for prediction of hospital mortality after trauma. Shock. 2008;29:662–6.PubMed Kaplan LJ, Kellum JA. Comparison of acid–base models for prediction of hospital mortality after trauma. Shock. 2008;29:662–6.PubMed
17.
Zurück zum Zitat Antonini B, Piva S, Paltenghi M, Candiani A, Latronico N. The early phase of critical illness is a progressive acidic state due to unmeasured anions. Eur J Anaesthesiol. 2008;25:566–71.CrossRefPubMed Antonini B, Piva S, Paltenghi M, Candiani A, Latronico N. The early phase of critical illness is a progressive acidic state due to unmeasured anions. Eur J Anaesthesiol. 2008;25:566–71.CrossRefPubMed
18.
Zurück zum Zitat Attana P, Lazzeri C, Chiostri M, Picariello C, Gensini GF, Valente S. Strong-ion gap approach in patients with cardiogenic shock following ST-elevation myocardial infarction. Acute Card Care. 2013;15:58–62.CrossRefPubMed Attana P, Lazzeri C, Chiostri M, Picariello C, Gensini GF, Valente S. Strong-ion gap approach in patients with cardiogenic shock following ST-elevation myocardial infarction. Acute Card Care. 2013;15:58–62.CrossRefPubMed
19.
Zurück zum Zitat Cusack R, Rhodes A, Lochhead P, Jordan B, Perry S, Ball J, Grounds R, Bennett E. The strong ion gap does not have prognostic value in critically ill patients in a mixed medical/surgical adult ICU. ICM. 2002;28:864–9. Cusack R, Rhodes A, Lochhead P, Jordan B, Perry S, Ball J, Grounds R, Bennett E. The strong ion gap does not have prognostic value in critically ill patients in a mixed medical/surgical adult ICU. ICM. 2002;28:864–9.
20.
Zurück zum Zitat FitzSullivan E, Salim A, Demetriades D, Asensio J, Martin MJ. Serum bicarbonate may replace the arterial base deficit in the trauma intensive care unit. Am J Surg. 2005;190:941–6.CrossRefPubMed FitzSullivan E, Salim A, Demetriades D, Asensio J, Martin MJ. Serum bicarbonate may replace the arterial base deficit in the trauma intensive care unit. Am J Surg. 2005;190:941–6.CrossRefPubMed
21.
Zurück zum Zitat Hucker TR, Mitchell GP, Blake LD, Cheek E, Bewick V, Grocutt M, Forni LG, Venn RM. Identifying the sick: can biochemical measurements be used to aid decision making on presentation to the accident and emergency department. Br J Anaesth. 2005;94:735–41.CrossRefPubMed Hucker TR, Mitchell GP, Blake LD, Cheek E, Bewick V, Grocutt M, Forni LG, Venn RM. Identifying the sick: can biochemical measurements be used to aid decision making on presentation to the accident and emergency department. Br J Anaesth. 2005;94:735–41.CrossRefPubMed
22.
Zurück zum Zitat Lazzeri C, Valente S, Chiostri M, Picariello C, Gensini GF. Evaluation of acid–base balance in ST-elevation myocardial infarction in the early phase: a prognostic tool? Coron Artery Dis. 2010;21:266–72.CrossRefPubMed Lazzeri C, Valente S, Chiostri M, Picariello C, Gensini GF. Evaluation of acid–base balance in ST-elevation myocardial infarction in the early phase: a prognostic tool? Coron Artery Dis. 2010;21:266–72.CrossRefPubMed
23.
Zurück zum Zitat Martin J, Blobner M, Busch R, Moser N, Kochs E, Luppa PB. Point-of-care testing on admission to the intensive care unit: lactate and glucose independently predict mortality. Clin Chem Lab Med. 2013;51:405–12.CrossRefPubMed Martin J, Blobner M, Busch R, Moser N, Kochs E, Luppa PB. Point-of-care testing on admission to the intensive care unit: lactate and glucose independently predict mortality. Clin Chem Lab Med. 2013;51:405–12.CrossRefPubMed
24.
Zurück zum Zitat Martin M, Murray J, Berne T, Demetriades D, Belzberg H. Diagnosis of acid–base derangements and mortality prediction in the trauma intensive care unit: the physiochemical approach. J Trauma. 2005;58:238–43.CrossRefPubMed Martin M, Murray J, Berne T, Demetriades D, Belzberg H. Diagnosis of acid–base derangements and mortality prediction in the trauma intensive care unit: the physiochemical approach. J Trauma. 2005;58:238–43.CrossRefPubMed
25.
Zurück zum Zitat Martin MJ, FitzSullivan E, Salim A, Berne TV, Towfigh S. Use of serum bicarbonate measurement in place of arterial base deficit in the surgical intensive care unit. Arch Surg. 2005;140(8):745–51.CrossRefPubMed Martin MJ, FitzSullivan E, Salim A, Berne TV, Towfigh S. Use of serum bicarbonate measurement in place of arterial base deficit in the surgical intensive care unit. Arch Surg. 2005;140(8):745–51.CrossRefPubMed
26.
Zurück zum Zitat Sahu A, Cooper HA, Panza JA. The initial anion gap is a predictor of mortality in acute myocardial infarction. Coron Artery Dis. 2006;17:409–12.CrossRefPubMed Sahu A, Cooper HA, Panza JA. The initial anion gap is a predictor of mortality in acute myocardial infarction. Coron Artery Dis. 2006;17:409–12.CrossRefPubMed
27.
Zurück zum Zitat Boniatti MM, Cardoso PRC, Castilho RK, Vieira SRR. Is hyperchloremia associated with mortality in critically ill patients? A prospective cohort study. J Crit Care. 2011;26:175–9.CrossRefPubMed Boniatti MM, Cardoso PRC, Castilho RK, Vieira SRR. Is hyperchloremia associated with mortality in critically ill patients? A prospective cohort study. J Crit Care. 2011;26:175–9.CrossRefPubMed
28.
Zurück zum Zitat Novovic MN, Jevdjict J. Prediction of mortality with unmeasured anions in critically ill patients on mechanical ventilation. Vojnosanit Pregl. 2014;71:936–41.CrossRefPubMed Novovic MN, Jevdjict J. Prediction of mortality with unmeasured anions in critically ill patients on mechanical ventilation. Vojnosanit Pregl. 2014;71:936–41.CrossRefPubMed
29.
Zurück zum Zitat Dondorp AM, Chau TT, Phu NH, Mai NT, Loc PP, Chuong LV, Sinh DX, Taylor A, Hien TT, White NJ, et al. Unidentified acids of strong prognostic significance in severe malaria. Crit Care Med. 2004;32:1683–8.CrossRefPubMed Dondorp AM, Chau TT, Phu NH, Mai NT, Loc PP, Chuong LV, Sinh DX, Taylor A, Hien TT, White NJ, et al. Unidentified acids of strong prognostic significance in severe malaria. Crit Care Med. 2004;32:1683–8.CrossRefPubMed
30.
Zurück zum Zitat Shane I, Robert W, Arthur K, Patson M, Moses G. Acid–base disorders as predictors of early outcomes in major Trauma in a resource limited setting: An observational prospective study. Pan Afr Med J. 2014;17:2.PubMedPubMedCentral Shane I, Robert W, Arthur K, Patson M, Moses G. Acid–base disorders as predictors of early outcomes in major Trauma in a resource limited setting: An observational prospective study. Pan Afr Med J. 2014;17:2.PubMedPubMedCentral
31.
Zurück zum Zitat Adams BD, Bonzani TA, Hunter CJ. The anion gap does not accurately screen for lactic acidosis in emergency department patients. Emerg Med J. 2006;23:179–82.CrossRefPubMedPubMedCentral Adams BD, Bonzani TA, Hunter CJ. The anion gap does not accurately screen for lactic acidosis in emergency department patients. Emerg Med J. 2006;23:179–82.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Chawla LS, Shih S, Davison D, Junker C, Seneff MG. Anion gap, anion gap corrected for albumin, base deficit and unmeasured anions in critically ill patients: implications on the assessment of metabolic acidosis and the diagnosis of hyperlactatemia. BMC Emerg Med. 2008;8:18.CrossRefPubMedPubMedCentral Chawla LS, Shih S, Davison D, Junker C, Seneff MG. Anion gap, anion gap corrected for albumin, base deficit and unmeasured anions in critically ill patients: implications on the assessment of metabolic acidosis and the diagnosis of hyperlactatemia. BMC Emerg Med. 2008;8:18.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Dinh CH, Ng R, Grandinetti A, Joffe A, Chow DC. Correcting the anion gap for hypoalbuminaemia does not improve detection of hyperlactataemia. Emerg Med J. 2006;23:627–9.CrossRefPubMedPubMedCentral Dinh CH, Ng R, Grandinetti A, Joffe A, Chow DC. Correcting the anion gap for hypoalbuminaemia does not improve detection of hyperlactataemia. Emerg Med J. 2006;23:627–9.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Levraut J, Bounatirou T, Ichai C, Ciais JF, Jambou P, Hechema R, Grimaud D. Reliability of anion gap as an indicator of blood lactate in critically ill patients. Intensive Care Med. 1997;23:417–22.CrossRefPubMed Levraut J, Bounatirou T, Ichai C, Ciais JF, Jambou P, Hechema R, Grimaud D. Reliability of anion gap as an indicator of blood lactate in critically ill patients. Intensive Care Med. 1997;23:417–22.CrossRefPubMed
35.
Zurück zum Zitat Nichol AD, Egi M, Pettila V, Bellomo R, French C, Hart G, Davies A, Stachowski E, Reade MC, Bailey M, et al. Relative hyperlactatemia and hospital mortality in critically ill patients: a retrospective multi-centre study. Crit Care. 2010;14:R25.CrossRefPubMedPubMedCentral Nichol AD, Egi M, Pettila V, Bellomo R, French C, Hart G, Davies A, Stachowski E, Reade MC, Bailey M, et al. Relative hyperlactatemia and hospital mortality in critically ill patients: a retrospective multi-centre study. Crit Care. 2010;14:R25.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Shapiro NI, Howell MD, Talmor D, Nathanson LA, Lisbon A, Wolfe RE, Weiss JW. Serum lactate as a predictor of mortality in emergency department patients with infection. Ann Emerg Med. 2005;45:524–8.CrossRefPubMed Shapiro NI, Howell MD, Talmor D, Nathanson LA, Lisbon A, Wolfe RE, Weiss JW. Serum lactate as a predictor of mortality in emergency department patients with infection. Ann Emerg Med. 2005;45:524–8.CrossRefPubMed
37.
Zurück zum Zitat Dubin A, Menises MM, Masevicius FD, Moseinco MC, Kutscherauer DO, Ventrice E, Laffaire E, Estenssoro E. Comparison of three different methods of evaluation of metabolic acid–base disorders. Crit Care Med. 2007;35:1264–70.CrossRefPubMed Dubin A, Menises MM, Masevicius FD, Moseinco MC, Kutscherauer DO, Ventrice E, Laffaire E, Estenssoro E. Comparison of three different methods of evaluation of metabolic acid–base disorders. Crit Care Med. 2007;35:1264–70.CrossRefPubMed
38.
Zurück zum Zitat Day NP, Phu NH, Mai NT, Chau TT, Loc PP, Chuong LV, Sinh DX, Holloway P, Hien TT, White NJ. The pathophysiologic and prognostic significance of acidosis in severe adult malaria. Crit Care Med. 2000;28:1833–40.CrossRefPubMed Day NP, Phu NH, Mai NT, Chau TT, Loc PP, Chuong LV, Sinh DX, Holloway P, Hien TT, White NJ. The pathophysiologic and prognostic significance of acidosis in severe adult malaria. Crit Care Med. 2000;28:1833–40.CrossRefPubMed
Metadaten
Titel
Anion gap as a prognostic tool for risk stratification in critically ill patients – a systematic review and meta-analysis
verfasst von
Stella Andrea Glasmacher
William Stones
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Anesthesiology / Ausgabe 1/2015
Elektronische ISSN: 1471-2253
DOI
https://doi.org/10.1186/s12871-016-0241-y

Weitere Artikel der Ausgabe 1/2015

BMC Anesthesiology 1/2015 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.