Skip to main content
Intended for healthcare professionals
Open access
Review article
First published online October 19, 2018

Vitamin C supplementation in the critically ill: A systematic review and meta-analysis

Abstract

Background:

Low plasma levels of vitamin C are associated with adverse outcomes, including increased mortality, in critically ill patients. Several trials have suggested that the administration of intravenous vitamin C in this setting may have beneficial effects, such as reducing the incidence of organ failure and improving survival. However, these studies have generally involved combination therapies consisting of vitamin C along with other antioxidants, confounding the effects of vitamin C alone. The primary objective of this meta-analysis is to investigate the effects of isolated intravenous supplementation of vitamin C in adults with critical illness.

Methods:

A database search was conducted for studies on the use of intravenous vitamin C in adult patients with critical illness. The primary outcome assessed was mortality at the longest follow-up time available. Secondary outcomes were the duration of mechanical ventilation, duration of vasopressor support, fluid requirements, and urine output in the first 24 h of intensive care unit admission.

Results:

Five studies (four randomized controlled trials and one retrospective review) enrolling a total of 142 patients were included in this meta-analysis. Compared with controls, the administration of intravenous vitamin C was associated with a decreased need for vasopressor support (standardized mean difference −0.71; 95% confidence interval (−1.16 to −0.26); p = 0.002) and decreased duration of mechanical ventilation (standardized mean difference −0.5; 95% confidence interval (−0.93 to −0.06); p = 0.03), but no difference was found in mortality (odds ratio 0.76; 95% confidence interval (0.27 to 2.16); p = 0.6). Trends were also noted toward decreased fluid requirements and increased urine output. No adverse effects were reported.

Conclusion:

The administration of intravenous vitamin C may lead to vasopressor sparing effects and a reduced need for mechanical ventilation in the critically ill, without affecting overall mortality. However, these results should be interpreted in light of the limitations of the primary literature and should serve as a preview of upcoming trials in this area.

Introduction

Vitamin C is a water-soluble vitamin with a variety of antioxidant,1 anti-inflammatory,2 and microvascular3 effects. Widely used in over-the-counter formulations for the common cold and general well-being,4 in recent years, there has been an expanding role for the use of vitamin C in the hospital setting. Although the overall prevalence of hypovitaminosis C is around 7.1% in the general population,5 up to 47.3% of undifferentiated hospitalized patients are deficient in vitamin C.6
Vitamin C levels are known to be decreased in critical illness710 and are associated with severity of illness.7,11 Although vitamin C requirements are greater in this population due to increased oxidative stress,12 levels may be restored to normal,8,13 or even brought to supra-normal,14 with parenteral supplementation.
Supplemental vitamin C has shown promise in both animal models of sepsis3,1521 and human trials in the intensive care unit (ICU) setting.2231 Although a simple vitamin, a variety of biological mechanisms have been postulated to account for the beneficial actions of vitamin C in the context of sepsis and organ failure. These include the prevention and restoration of micro-circulatory flow impairment due to reactive oxygen species, the preservation of vascular responsiveness to vasoconstrictors, the preservation of endothelial barrier function, and augmentation of anti-bacterial defense, leading to an overall mitigation of organ injury and dysfunction in critically ill patients.32
However, the largest studies with the most promising results have investigated vitamin C as part of combination (“cocktail”) therapies administered together with vitamin E or thiamine plus hydrocortisone.31 The last study is particularly impressive, demonstrating a substantial (8.5% versus 40.4%) reduction in mortality as well as decreased Sequential Organ Failure Assessment (SOFA) scores and length of vasopressor support compared to controls, but it is unclear which, if any, of the three constituents was responsible for these effects, obscuring the true effect of vitamin C. Furthermore, questions have been raised regarding the methodology of this study with respect to its small sample size, lack of randomization, and retrospective design.33
Few studies have focused specifically on the benefits of isolated administration of vitamin C in critically ill patients. Generally, these studies have been small and have yielded few conclusive results, and a comprehensive synthesis of the data has not been conducted heretofore. Hence, we aim in this review to provide a comprehensive meta-analysis of all studies involving isolated vitamin C administration in critically ill patients and to examine the effects upon overall mortality in addition to common clinical parameters in this setting, such as vasopressor requirements, the duration of mechanical ventilation, and resuscitation fluid requirements.

Methods and materials

Data sources

This study was conducted and prepared per the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines.34 We aimed to collect all studies which assessed the use of intravenous vitamin C in adult patients with critical illness. The following databases were searched for English language articles published from inception to September 2017: MEDLINE, Cochrane Central Register of Controlled Trials and the Cochrane Database of Systematic Reviews, and Elsevier. The following keywords were used along with MeSH terms: “vitamin C” or “ascorbic” or “ascorbate” and “sepsis” or “septic” or “shock” or “respiratory distress” or “ARDS” or “lung injury” or “critical” or “critically” or “intensive” or “ICU” or “trauma” or “burn.” All search terms were exploded and no restrictions or limitations were placed in our search strategy.

Study selection criteria

We included trials with the following characteristics:
1.
Type of studies: prospective and retrospective clinical trials;
2.
Population: adult ICU patients;
3.
Intervention: intravenous vitamin C supplementation versus placebo or no intervention, with no minimum dose.
4.
Study outcome: The primary outcome examined was cause mortality in the longest duration provided by the studies. Secondary outcomes were the duration of mechanical ventilation, duration of vasopressor support, fluid requirements, and urine output in the first 24 h of ICU admission.
Trials with these characteristics were excluded:
1.
They were not published as original studies;
2.
They were not published in English;
3.
They did not use adult patients;
4.
They did not use vitamin C alone without other interventions;
5.
Full-text articles not available;
6.
Lack of data on mortality;
7.
Case reports;
8.
Animal studies.

Data extraction

Data from included studies were extracted independently by both authors, and discrepancies were resolved through discussion until consensus was reached. The primary outcome was all cause mortality in the longest duration provided by the studies. Secondary outcomes were the duration of mechanical ventilation, duration of vasopressor support, and fluid requirements and urine output in the first 24 h of ICU admission. When appropriate, standard deviations were estimated using previously described methods.35 For standardized mean difference (SMD) analysis, scales of differing directionality were standardized when necessary with multiplication by −1.

Risk of bias

We used the Cochrane Collaboration’s tool for assessing risk of bias to evaluate the quality of the included randomized controlled trials.36,37 Domains assessed were random sequence generation (selection bias), allocation concealment (selection bias), blinding of participants and personnel (performance bias), blinding of outcome assessment (detection bias), incomplete outcome data (attrition bias), selective reporting (reporting bias), and other sources of bias. The remaining observational study was similarly assessed with the ROBINS-I tool.38 Studies were considered to be low risk only if every domain was individually adjudicated as such; otherwise, any domain rated as unclear or high risk brought the overall risk to that respective stratification.

Statistical analysis

Data were analyzed by Review Manager 5.3 (The Nordic Cochrane Center, Rigshospitalet, Copenhagen, Denmark). Results are presented with forest plots using odds ratios (ORs) with 95% confidence intervals (CIs) for dichotomous data and SMD with 95% CI for continuous data. The statistical heterogeneity among the studies was calculated and assessed with the I2 test based on previously recommended stratifications (low, moderate, and high to I2 values of 25%, 50%, and 75%, respectively).39 A Mantel–Haenszel (MH) method was used for the dichotomous primary outcome of mortality. For all continuous outcomes, inverse variance methods were used. In addition, the random effects model of DerSimonian and Laird was used if heterogeneity was observed; otherwise, a fixed effects model was used. The p value of <0.05 was considered statistically significant. Tests for publication bias, such as the funnel plot and Egger’s linear regression, were not utilized due to the low number of included studies.

Results

Study selection

The literature search strategy outlined above returned 3961 citations (Figure 1). After duplicates were expunged, 3478 articles were scanned by titles and abstracts, and 3425 non-relevant studies were removed. The remaining 53 articles were assessed for eligibility. Notable exclusions were due to pre- or perioperative administration for scheduled cardiac (n = 14),4053 gastrointestinal (n = 1),54 vascular (n = 1),55 or gynecologic (n = 1)56 surgery, administration as part of combination therapy with other agents (n = 27),2731,5778 or case reports (n = 3).7981 Many of the above were also excluded due to enteral administration. One study was excluded because the investigators sought to normalize plasma ascorbic acid by administering varying doses depending on baseline levels; this study also did not report clinical outcomes.82 Finally, five studies (Table 1) were included in this meta-analysis.2226
Figure 1. Study selection flow chart.
Table 1. Characteristics of included studies.
Study Design Setting Patients (n intervention/n control) Vitamin C dose in intervention group Overall mortality n (%) Mortality follow-up
Tanaka et al.22 Quasi-RCT Trauma/critical care unit Burn > 30% TBSA (total body surface area) (19/19) 66 mg/kg/h IV in Ringer’s lactate (RL) for 24 h 16 (43) Unspecified
Ferrón-Celma et al.23 RCT Surgical Septic patients post-abdominal surgery (10/10) 450 mg IV × 6 days post-op 10 (50) 6 days
Kahn et al.24 Observational Burn/trauma unit Burn > 20% TBSA (17/16) 66 mg/kg/h IV in RL for 24 h 7 (21) 2 weeks
Fowler et al.25 RCT Medical ICU Severe sepsis (16 (8 high dose, 8 low dose)/8) 50 mg (low dose) or 200 mg (high dose)/kg/24 h IV for 96 hours 12 (50) 28 days
Zabet et al.26 RCT Surgical ICU Surgical patients with septic shock (14/14) 25 mg/kg IV q6 h × 72 h 11 (39) 28 days
RCT: randomized controlled trial.

Study characteristics and quality

These five studies (four RCTs and one retrospective review) enrolled a total of 142 patients, of whom 76 received intravenous vitamin C, compared to 66 controls. Isovolumic placebo was administered in three studies,23,25,26 while in the remaining two studies, vitamin C was incorporated into resuscitation fluids in the intervention group.22,24 The average age was 54 years, and 72% of participants were male. Study settings were medical,25 surgical,23,26 and burn/trauma.22,26 All studies were single-center. The risk of bias summary figure is presented in Figure 2. Three of the four RCTs described some type of random sequence allocation, and these same three were also double-blinded. The Tanaka trial, despite its heading, should more accurately be considered as a quasi-randomized control trial, as the study participants therein were allocated per month of admission.22 Because blinding of outcome assessment was not specified, no trial was considered low risk. Across all domains, three studies were considered to be of unclear risk and the remaining two as high risk.
Figure 2. Risk of bias summary for RCTs (left) and the remaining observational study (right).

Meta-analysis results

Intravenous vitamin C administration and ICU/hospital mortality

Meta-analysis of the five included trials totaling 142 patients shows that intravenous vitamin C administration does not significantly reduce ICU and hospital mortality in critically ill patients (OR 0.76; 95% CI (0.27 to 2.16); p = 0.6) (Figure 3). Data were taken from the longest available follow-up, ranging from 6 to 28 days.23,25,26 Because study heterogeneity was moderate (I2 = 50%), a random effects model was used for this analysis. A sensitivity analysis was performed by removing each study singly and reanalyzing the remaining studies; no change in the effect size was noted.
Figure 3. Forest plot of the effect of intravenous vitamin C administration on mortality at the longest available follow-up of all studies.

Duration of vasopressor support

Three studies2426 enrolling a total of 85 patients evaluated the requirements for vasopressor support needed to maintain adequate hemodynamics. Compared to controls, the administration of intravenous vitamin C was associated with a reduced duration of vasopressor use (SMD −0.71; 95% CI (−1.16 to −0.26); p = 0.002) (Figure 4). Vasopressors administered include norepinephrine,26 vasopressin, dopamine, and phenylephrine.24 Because the data among these studies were homogeneous, a fixed effects model was used.
Figure 4. Forest plot of the effect of intravenous vitamin C administration on vasopressor requirements.

Duration of mechanical ventilation

Administration of vitamin C was associated with a reduction in the duration of mechanical ventilation in the three studies totaling 89 patients which investigated this outcome (SMD −0.5; 95% CI (−0.93 to −0.06); p = 0.03) (Figure 5). Data were reported as either time spent on ventilator support22,26 or ventilator free time.25 Again, a fixed effects model was used for this analysis.
Figure 5. Forest plot of the effect of intravenous vitamin C administration on the duration of mechanical ventilation.

Fluid requirements and urine output

Trends were noted toward both decreased fluid requirements (SMD −0.9; 95% CI (−1.86 to 0.06); p = 0.07) and urine output (SMD −0.5; 95% CI (−1.12 to 0.12); p = 0.11) in the three studies which investigated this outcome in the first 24 h of ICU admission22,24,26 (Figure 6). A random effects model was used due to high study heterogeneity. Also, in Zabet et al.26 study, while there was no difference in the first 24 h, there was a trend toward increased urine output when the entire duration of the study is considered, and this would result in overall statistical significance.
Figure 6. Forest plots of the effect of intravenous vitamin C administration on fluid requirements (top) and urine output (bottom) in the first 24 h of ICU admission.

Discussion

Although several meta-analyses have explored the effect of combination antioxidants in critical illness, our study is the first to focus exclusively on the role of vitamin C. The main results from our analysis of five studies are that vitamin C administration is not associated with decreased mortality but is associated with decreased vasopressor requirements and duration of mechanical ventilation.
Vitamin C is an essential cofactor for the production of endogenous vasopressors.83 Through its actions on tyrosine hydroxylase, the rate-limiting enzyme of catecholamine synthesis,84,85 and its role as a cofactor for dopamine β-hydroxylase,86 vitamin C is involved in the biosynthesis of norepinephrine at physiologic concentrations.84 Vitamin C also acts as a cofactor for peptidylglycine α-amidating monooxygenase (PAM),87 an enzyme that catalyzes the formation of arginine vasopressin.88 On the basis of these observations, Carr et al.89 hypothesized that “the administration of high-dose ascorbate in conditions of hypovitaminosis C (e.g. severe sepsis and septic shock) may support the endogenous synthesis of these vasoactive compounds and thus ameliorate the need for exogenously administered vasopressors.”
It is noteworthy that, although we were able to demonstrate decreased vasopressor requirements and trends toward reduced fluid resuscitation needs and increased urine output, we did not find an overall difference in mortality. One possible reason for this discrepancy is the variance in baseline patient characteristics across the included studies. In general, the studies which did not demonstrate a numerically less mortality rate in the treatment group featured patients with relatively less severe hemodynamic derangements upon study commencement. In Tanaka et al.’s22 study, the average mean arterial pressure of enrolled patients was around 90 mm Hg, and all deaths occurred after the 96-h fluid resuscitation phase, by which time plasma vitamin C levels in the treatment group had already declined to match controls. In Kahn et al.’s study, only 4 of 17 patients in the treatment group required vasopressors compared to 9 of 16 controls. The authors note that “because one group had more than 50% more patients on vasopressors, the results would have been misleading.” In Ferrón-Celma et al.’s23 study, only 5 of 20 patients even required vasopressor support. Conversely, Zabet et al.26 did demonstrate decreased mortality with vitamin C administration, but the average mean arterial pressure (MAP) upon enrollment was under 70 mm Hg. Taken together, these observations support the hypothesis that vitamin C indeed exerts a vasopressor sparing effect, but the magnitude of this effect may depend upon the initial need for vasopressor support. The benefits of supplemental vitamin C would be expected to be more pronounced in those with refractory vasopressor-dependent shock compared to patients with stable hemodynamics with little or no need for vasopressor support, and this difference in expected benefit might explain the difference in mortality among the analyzed studies. A similar line of reasoning applied to the VASST (Vasopressin and Septic Shock) trial, where the administration of supplemental vasopressin reduced the need for norepinephrine support without affecting mortality; the authors therein speculated that the reason for this was due to the high average MAP of enrolled patients (72–73 mm Hg).90
The significance of the decreased requirements for mechanical ventilation is unknown, as is the question of whether this is actually reflective of a real improvement in pulmonary status. Across all studies, only Tanaka et al.22 demonstrated an improvement in any parameter of pulmonary function (increased PaO2/FiO2 ratio in the treatment group), but the average duration of mechanical ventilation in the treatment group was 12.1 days, far longer than the initial 24-h period of vitamin C administration. Furthermore, it is unclear whether this effect is dependent upon a decreased need for resuscitation fluids and hence a concomitant reduction in the incidence and severity of pulmonary edema or is an independent effect upon the lung parenchyma. Animal research has demonstrated that vitamin C exerts a protective effect upon the pulmonary tissues in both ischemia–reperfusion91 and lipopolysaccharide-induced models of lung injury15,92,93 through a variety of mechanisms, including increased alveolar fluid clearance, enhanced epithelial barrier function, and the attenuation of pro-inflammatory and pro-coagulant states accompanying sepsis.94 The same group has reported encouraging results in a recent series of case reports in the setting of acute respiratory distress syndrome (ARDS)7981 and has recently concluded a phase II multicenter trial investigating Vitamin C Infusion for Treatment in Sepsis Induced Acute Lung Injury (CITRIS-ALI, NCT02106975).95 This study should further clarify the possible role of vitamin C in the treatment of acute lung injury and ARDS.
The optimal dose and target plasma concentration of vitamin C in the setting of critical illness are unknown. The recommended daily oral dose in healthy subjects needed to maintain normal plasma levels above 50 µmol/L is 95–110 mg/day.96 However, physiological requirements are increased during times of increased oxidative stress and metabolic turnover,12 and plasma levels have been found to be correspondingly lower in the ICU population.7 Intravenous dosing has been shown to produce higher plasma concentrations than oral administration97 due to saturable intestinal absorption,98 and 3 g/day is required to maintain plasma levels in the normal range in ICU patients.8
The five studies included in this meta-analysis featured doses as low as 450 mg/day23 to 1584 mg/kg/day,22,24 a 250-fold difference in a 70 kg subject. The difference in dosages given may result in differing effects. As previously mentioned, vitamin C is an essential cofactor for the production of endogenous vasopressors.83 It has been suggested that vitamin C is released from the adrenal cortex in response to adrenocorticotropic hormone (ACTH) to ensure that “norepinephrine synthesis [in the medulla] always proceeds at maximum velocity (Vmax).”99 Indeed, this study noted that vitamin C levels in the adrenal veins of patients with hyperaldosteronism who were administered ACTH were found to be 176 ± 71 μmol/L, over four times higher than concentrations within plasma. The authors speculated that this allows vitamin C to act locally as a paracrine mediator to stimulate norepinephrine and epinephrine synthesis. If the Carr hypothesis89 is correct, and given the suppression of corticotropin and the overall dysregulation of the hypothalamic–pituitary–adrenal (HPA) axis during critical illness,100,101 it can be surmised that the minimum plasma concentration of vitamin C required in critical illness to support endogenous norepinephrine synthesis should be at least as great as the maximum intraadrenal concentration needed for this function in healthy controls. Analogous reasoning with hypophyseal vein concentrations can be extended for endogenous vasopressin synthesis in the setting of sepsis-related endocrine dysfunction. This level might be slightly higher given the higher pituitary concentrations of vitamin C.102
In the Fowler study, patients administered “low-dose” vitamin C at 50 mg/kg/24 h achieved an average steady-state plasma concentration of 331 μmol/L,25 above what would be expected to support endogenous norepinephrine synthesis. However, there was a further reduction in SOFA scores among patients given a higher dose of vitamin C at 200 mg/kg/24 h. Patients in this latter group achieved an average steady-state plasma concentration of 3082 μmol/L. This may suggest additional, non-hemodynamic benefits at higher, supra-physiologic doses, possibly through the antioxidant, anti-inflammatory, and microvascular actions of vitamin C, and these may be responsible for the aforementioned pulmonary benefits. Furthermore, serum levels of C-reactive protein,25 procalcitonin,25 poly(ADP-ribose) polymerase,23 and malondialdehyde22 were decreased in the intervention groups across multiple studies. Several of these markers of cellular damage have been found to correlate with organ failure beyond the cardiovascular and pulmonary systems, suggesting an overall bodily cytoprotective effect in the context of systemic inflammation.103,104
High-dose intravenous vitamin C is generally regarded as safe even in gram doses.105 No significant side effects were reported across any of these five studies. Nevertheless, caution should be exercised in patients with renal impairment.106 Although rare, oxalate nephropathy has been documented in burn patients using the same 66 mg/kg/h dose used in the Tanaka and Kahn studies.107 Patients with glucose 6-phosphate dehydrogenase deficiency and paroxysmal nocturnal hemoglobinuria should be excluded from treatment due to the risk of intravascular hemolysis.13 Although not studied in the critically ill population, potential pro-oxidant effects are largely of academic interest only and do not appear to be of concern in noncancerous cells.108 Interestingly, one recent study in the outpatient setting suggests that intravenous vitamin C administered at gram doses may cause an acute decrease in MAP of around 7 mm Hg.109 Although this effect was limited to patients with prehypertension at baseline, vitamin C does indeed have acute vasodilatory properties;110 whether this may precipitate a paradoxical collapse in hemodynamic function in the setting of critical illness is unknown.
A major strength of this meta-analysis is that we investigated only the administration of isolated vitamin C on clinical outcomes in the critically ill, as opposed to vitamin C in combination with other agents as part of an antioxidant cocktail, the effects of which cannot be attributed properly to any particular agent. Also, a varied mix of medical, surgical, and burn patients were included, and each study contributed relatively equally in weight.
This meta-analysis has several weaknesses that should be kept in mind. First, only five studies were included due to the paucity of research on isolated vitamin C administration. Study sizes were small, with the largest study consisting of only 37 subjects, the doses used between studies was disparate, and the risk of bias was generally judged to be uncertain or high. Sample sizes for secondary outcomes were even sparser. Heterogeneity among the studies and the patient populations therein was not insignificant. In particular, mortality data had to be taken from the longest available time point in each study due to the non-uniform duration of follow-up across studies. In addition, through the inclusion of patients across many diverse settings, the gain in statistical power resulting therefrom must be weighed against the weakening association between the fixed intervention and the growing list of conditions, all under the umbrella of “critical illness,” on which the agent acts in thematic relation to. Whereas the mechanisms by which vitamin C exerts its actions have been studied mostly on animal models of sepsis and septic shock, such pathways may not pertain in likewise fashion to the hypovolemic shock of burn patients; the inclusion of the latter population and the application of meta-analytic techniques thereto may result in a conclusion that dilutes or otherwise obfuscates the effects of vitamin C when applied only to sepsis. One study utilized a dose of vitamin C far below the others, and this was not adjusted for patient weight;23 however, the exclusion of this study did not affect the primary outcome of mortality. Finally, focusing on the isolated administration of vitamin C ignores any summative or synergistic benefits to be had when used in combination with other agents. Without even knowing the specific nature of vitamin C, it does seem a priori implausible that a single substance would have a substantial effect upon a parameter as global as mortality given the complex biochemical and pathophysiological milieu of critical illness. Many essential vitamin and mineral deficiencies can occur in this setting, including iron, selenium,111 magnesium,112 thiamine,113 and vitamin D,114 and repletion of several of these may be required before substantial clinical improvements are seen.
In view of these deficiencies in the primary literature, this meta-analysis should be considered as an orienting endeavor to guide future studies. Indeed, further trials are in various stages of completion in this area, exploring the role of vitamin C within the context of acute lung injury,95 severe sepsis,115,116 and septic shock.117,118 In addition, spurred by the promising results of the Marik study, trials are underway to explore the effects of vitamin C combined with thiamine and hydrocortisone.119123 Taken together, these studies should elucidate both the role of isolated vitamin C and its synergistic effects.

Conclusion

Vitamin C deficiency is common in the context of critical illness and is associated with negative outcomes. Based on the current available evidence, the intravenous administration of vitamin C produces vasopressor sparing effects, possibly through the support of endogenous vasopressor synthesis, and reduces dependency on mechanical ventilation, possibly through the amelioration of lung injury, without affecting overall mortality. However, these conclusions should be tempered by the inherent limitations of the primary literature, particularly with respect to study heterogeneity, and forthcoming trials should further clarify the role of vitamin C in the management of the critically ill.

Declaration of conflicting interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) received no financial support for the research, authorship, and/or publication of this article.

ORCID iD

References

1. Padayatty SJ, Katz A, Wang Y, et al. Vitamin C as an antioxidant: evaluation of its role in disease prevention. J Am Coll Nutr 2003; 22: 18–35.
2. Sorice A, Guerriero E, Capone F, et al. Ascorbic acid: its role in immune system and chronic inflammation diseases. Mini Rev Med Chem 2014; 14: 444–452.
3. Tyml K. Vitamin C and microvascular dysfunction in systemic inflammation. Antioxidants 2017; 6: E49.
4. Hemila H, Chalker E. Vitamin C for preventing and treating the common cold. Cochrane Database Syst Rev 2013; 2013: CD000980.
5. Schleicher RL, Carroll MD, Ford ES, et al. Serum vitamin C and the prevalence of vitamin C deficiency in the United States: 2003-2004 National Health and Nutrition Examination Survey (NHANES). Am J Clin Nutr 2009; 90: 1252–1263.
6. Fain O, Paries J, Jacquart B, et al. Hypovitaminosis C in hospitalized patients. Eur J Intern Med 2003; 14: 419–425.
7. Schorah CJ, Downing C, Piripitsi A, et al. Total vitamin C, ascorbic acid, and dehydroascorbic acid concentrations in plasma of critically ill patients. Am J Clin Nutr 1996; 63: 760–765.
8. Long CL, Maull KI, Krishnan RS, et al. Ascorbic acid dynamics in the seriously ill and injured. J Surg Res 2003; 109: 144–148.
9. Metnitz PG, Bartens C, Fischer M, et al. Antioxidant status in patients with acute respiratory distress syndrome. Intensive Care Med 1999; 25: 180–185.
10. Polidori MC, Mecocci P, Frei B. Plasma vitamin C levels are decreased and correlated with brain damage in patients with intracranial hemorrhage or head trauma. Stroke 2001; 32: 898–902.
11. Galley HF, Davies MJ, Webster NR. Ascorbyl radical formation in patients with sepsis: effect of ascorbate loading. Free Radic Biol Med 1996; 20: 139–143.
12. Berger MM. Vitamin C requirements in parenteral nutrition. Gastroenterology 2009; 137: S70–S78.
13. Wilson JX. Evaluation of vitamin C for adjuvant sepsis therapy. Antioxid Redox Signal 2013; 19: 2129–2140.
14. Bell C, Carson JM, Motte NW, et al. Ascorbic acid does not affect the age-associated reduction in maximal cardiac output and oxygen consumption in healthy adults. J Appl Physiol 2005; 98: 845–849.
15. Fisher BJ, Seropian IM, Kraskauskas D, et al. Ascorbic acid attenuates lipopolysaccharide-induced acute lung injury. Crit Care Med 2011; 39: 1454–1460.
16. Secor D, Li F, Ellis CG, et al. Impaired microvascular perfusion in sepsis requires activated coagulation and P-selectin-mediated platelet adhesion in capillaries. Intensive Care Med 2010; 36: 1928–1934.
17. Tyml K, Li F, Wilson JX. Septic impairment of capillary blood flow requires nicotinamide adenine dinucleotide phosphate oxidase but not nitric oxide synthase and is rapidly reversed by ascorbate through an endothelial nitric oxide synthase-dependent mechanism. Crit Care Med 2008; 36: 2355–2362.
18. McKinnon RL, Lidington D, Tyml K. Ascorbate inhibits reduced arteriolar conducted vasoconstriction in septic mouse cremaster muscle. Microcirculation 2007; 14: 697–707.
19. Tyml K, Li F, Wilson JX. Delayed ascorbate bolus protects against maldistribution of microvascular blood flow in septic rat skeletal muscle. Crit Care Med 2005; 33: 1823–1828.
20. Wu F, Wilson JX, Tyml K. Ascorbate inhibits iNOS expression and preserves vasoconstrictor responsiveness in skeletal muscle of septic mice. Am J Physiol Regul Integr Comp Physiol 2003; 285: R50–R56.
21. Zhou G, Kamenos G, Pendem S, et al. Ascorbate protects against vascular leakage in cecal ligation and puncture-induced septic peritonitis. Am J Physiol Regul Integr Comp Physiol 2012; 302: R409–R416.
22. Tanaka H, Matsuda T, Miyagantani Y, et al. Reduction of resuscitation fluid volumes in severely burned patients using ascorbic acid administration: a randomized, prospective study. Arch Surg 2000; 135: 326–331.
23. Ferrón-Celma I, Mansilla A, Hassan L, et al. Effect of vitamin C administration on neutrophil apoptosis in septic patients after abdominal surgery. J Surg Res 2009; 153: 224–230.
24. Kahn SA, Beers RJ, Lentz CW. Resuscitation after severe burn injury using high-dose ascorbic acid: a retrospective review. J Burn Care Res 2011; 32: 110–117.
25. Fowler AA III, Syed AA, Knowlson S, et al. Phase I safety trial of intravenous ascorbic acid in patients with severe sepsis. J Transl Med 2014; 12: 32.
26. Zabet MH, Mohammadi M, Ramezani M, et al. Effect of high-dose ascorbic acid on vasopressor’s requirement in septic shock. J Res Pharm Pract 2016; 5: 94–100.
27. Nathens AB, Neff MJ, Jurkovich GJ, et al. Randomized, prospective trial of antioxidant supplementation in critically ill surgical patients. Ann Surg 2002; 236: 814–822.
28. Crimi E, Liguori A, Condorelli M, et al. The beneficial effects of antioxidant supplementation in enteral feeding in critically ill patients: a prospective, randomized, double-blind, placebo-controlled trial. Anesth Analg 2004; 99: 857–863, table of contents.
29. Collier BR, Giladi A, Dossett LA, et al. Impact of high-dose antioxidants on outcomes in acutely injured patients. JPEN J Parenter Enteral Nutr 2008; 32: 384–388.
30. Berger MM, Soguel L, Shenkin A, et al. Influence of early antioxidant supplements on clinical evolution and organ function in critically ill cardiac surgery, major trauma, and subarachnoid hemorrhage patients. Crit Care 2008; 12: R101.
31. Marik PE, Khangoora V, Rivera R, et al. Hydrocortisone, vitamin C, and thiamine for the treatment of severe sepsis and septic shock: a retrospective before-after study. Chest 2017; 151: 1229–1238.
32. Oudemans-van Straaten HM, Spoelstra-de Man AM, de Waard MC. Vitamin C revisited. Crit Care 2014; 18: 460.
33. Blythe R, Cook D, Graves N. Scepticaemia: the impact on the health system and patients of delaying new treatments with uncertain evidence: a case study of the sepsis bundle. F1000Res 2018; 7: 500.
34. PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses (PRISMA), http://prisma-statement.org (accessed 8 September 2017).
35. Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med Res Methodol 2005; 5: 13.
36. Higgins J, Green S (eds). Cochrane handbook for systematic reviews of interventions version 5.1.0, March 2011, https://handbook-5-1.cochrane.org/
37. Higgins JPT, Altman DG, Gotzsche PCet al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. Br Med J 2011; 343: d5928.
38. Sterne JA, Hernan MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. Br Med J 2016; 355: i4919.
39. Higgins JPT, Thompson SG, Deeks JJ, et al. Measuring inconsistency in meta-analyses. Br Med J 2003; 327: 557–560.
40. Ebade A, Taha WS, Saleh RH, et al. Ascorbic acid versus magnesium for the prevention of atrial fibrillation after coronary artery bypass grafting surgery. Egypt J Cardiothorac Anesth 2014; 8: 59–65.
41. Sadeghpour A, Alizadehasl A, Kyavar M, et al. Impact of vitamin C supplementation on post-cardiac surgery ICU and hospital length of stay. Anesthesiol Pain Med 2015; 5: e25337.
42. Oktar GL, Sinci V, Kalaycioglu S, et al. Biochemical and hemodynamic effects of ascorbic acid and alpha-tocopherol in coronary artery surgery. Scand J Clin Lab Invest 2001; 61: 621–629.
43. Antonic M, Lipovec R, Gregorcic F, et al. Perioperative ascorbic acid supplementation does not reduce the incidence of postoperative atrial fibrillation in on-pump coronary artery bypass graft patients. J Cardiol 2017; 69: 98–102.
44. Barta E, Pechan I, Cornak V, et al. Protective effect of alpha-tocopherol and L-ascorbic acid against the ischemic-reperfusion injury in patients during open-heart surgery. Bratisl Lek Listy 1991; 92: 174–183.
45. Das D, Sen C, Goswami A. Effect of vitamin C on adrenal suppression by etomidate induction in patients undergoing cardiac surgery: a randomized controlled trial. Ann Card Anaesth 2016; 19: 410–417.
46. Safaei N, Babaei H, Azarfarin R, et al. Comparative effect of grape seed extract (Vitis vinifera) and ascorbic acid in oxidative stress induced by on-pump coronary artery bypass surgery. Ann Card Anaesth 2017; 20: 45–51.
47. Dehghani MR, Majidi N, Rahmani A, et al. Effect of oral vitamin C on atrial fibrillation development after isolated coronary artery bypass grafting surgery: a prospective randomized clinical trial. Cardiol J 2014; 21: 492–499.
48. Samadikhah J, Golzari SE, Sabermarouf B, et al. Efficacy of combination therapy of statin and vitamin C in comparison with statin in the prevention of post-CABG atrial fibrillation. Adv Pharm Bull 2014; 4: 97–100.
49. Bjordahl PM, Helmer SD, Gosnell DJ, et al. Perioperative supplementation with ascorbic acid does not prevent atrial fibrillation in coronary artery bypass graft patients. Am J Surg 2012; 204: 862–867; discussion 867.
50. Carnes CA, Chung MK, Nakayama T, et al. Ascorbate attenuates atrial pacing-induced peroxynitrite formation and electrical remodeling and decreases the incidence of postoperative atrial fibrillation. Circ Res 2001; 89: E32–E38.
51. Colby JA, Chen WT, Baker WL, et al. Effect of ascorbic acid on inflammatory markers after cardiothoracic surgery. Am J Health Syst Pharm 2011; 68: 1632–1639.
52. Eslami M, Badkoubeh RS, Mousavi M, et al. Oral ascorbic acid in combination with beta-blockers is more effective than beta-blockers alone in the prevention of atrial fibrillation after coronary artery bypass grafting. Texas Hear Inst J 2007; 34: 268–274.
53. Papoulidis P, Ananiadou O, Chalvatzoulis E, et al. The role of ascorbic acid in the prevention of atrial fibrillation after elective on-pump myocardial revascularization surgery: a single-center experience—a pilot study. Interact Cardiovasc Thorac Surg 2011; 12: 121–124.
54. Yamazaki E, Horikawa M, Fukushima R. Vitamin C supplementation in patients receiving peripheral parenteral nutrition after gastrointestinal surgery. Nutrition 2011; 27: 435–439.
55. Duffy MJO, Kane CM, Stevenson M, et al. A randomized clinical trial of ascorbic acid in open abdominal aortic aneurysm repair. Intensive Care Med Exp 2015; 3: 50.
56. Pirbudak L, Balat O, Cekmen M, et al. Effect of ascorbic acid on surgical stress response in gynecologic surgery. Int J Clin Pract 2004; 58: 928–931.
57. Preiser JC, Van Gossum A, Berre J, et al. Enteral feeding with a solution enriched with antioxidant vitamins A, C, and E enhances the resistance to oxidative stress. Crit Care Med 2000; 28: 3828–3832.
58. Barbosa E, Faintuch J, Machado Moreira EA, et al. Supplementation of vitamin E, vitamin C, and zinc attenuates oxidative stress in burned children: a randomized, double-blind, placebo-controlled pilot study. J Burn Care Res 2009; 30: 859–866.
59. Sahib AS, Al-Jawad FH, Alkaisy AA. Effect of antioxidants on the incidence of wound infection in burn patients. Ann Burns Fire Disasters 2010; 23: 199–205.
60. Bedreag OH, Rogobete AF, Sarandan M, et al. Influence of antioxidant therapy on the clinical status of multiple trauma patients. A retrospective single center study. Rom J Anaesth Intensive Care 2015; 22: 89–96.
61. Rock CL, Dechert RE, Khilnani R, et al. Carotenoids and antioxidant vitamins in patients after burn injury. J Burn Care Rehabil 1997; 18: 269–278; discussion 268.
62. Gadek JE, DeMichele SJ, Karlstad MD, et al. Effect of enteral feeding with eicosapentaenoic acid, gamma-linolenic acid, and antioxidants in patients with acute respiratory distress syndrome. Enteral nutrition in ARDS study group. Crit Care Med 1999; 27: 1409–1420.
63. Singer P, Theilla M, Fisher H, et al. Benefit of an enteral diet enriched with eicosapentaenoic acid and gamma-linolenic acid in ventilated patients with acute lung injury. Crit Care Med 2006; 34: 1033–1038.
64. Pontes-Arruda A, Martins LF, de Lima SM, et al. Enteral nutrition with eicosapentaenoic acid, gamma-linolenic acid and antioxidants in the early treatment of sepsis: results from a multicenter, prospective, randomized, double-blinded, controlled study: the INTERSEPT study. Crit Care 2011; 15: R144.
65. Pontes-Arruda A, Aragao AMA, Albuquerque JD. Effects of enteral feeding with eicosapentaenoic acid, gamma-linolenic acid, and antioxidants in mechanically ventilated patients with severe sepsis and septic shock. Crit Care Med 2006; 34: 2325–2333.
66. Grau-Carmona T, Moran-Garcia V, Garcia-de-Lorenzo A, et al. Effect of an enteral diet enriched with eicosapentaenoic acid, gamma-linolenic acid and anti-oxidants on the outcome of mechanically ventilated, critically ill, septic patients. Clin Nutr 2011; 30: 578–584.
67. Rice TW, Wheeler AP, Thompson BT, et al. Enteral omega-3 fatty acid, gamma-linolenic acid, and antioxidant supplementation in acute lung injury. JAMA 2011; 306: 1574–1581.
68. Galley HF, Howdle PD, Walker BE, et al. The effects of intravenous antioxidants in patients with septic shock. Free Radic Biol Med 1997; 23: 768–774.
69. Sawyer M, Mike J, Cahvin K. Antioxidant therapy and survival in ARDS. Crit Care Med 1989; 17: 33–37.
70. Shirai K, Yoshida S, Matsumaru N, et al. Effect of enteral diet enriched with eicosapentaenoic acid, gamma-linolenic acid, and antioxidants in patients with sepsis-induced acute respiratory distress syndrome. J Intensive Care 2015; 3: 24.
71. Howe KP, Clochesy JM, Goldstein LS, et al. Mechanical ventilation antioxidant trial. Am J Crit Care 2015; 24: 440–445.
72. Rehou S, Shahrokhi S, Natanson R, et al. Antioxidant and trace element supplementation reduce the inflammatory response in critically ill burn patients. J Burn Care Res 2018; 39: 1–9.
73. Kagan I, Cohen J, Stein M, et al. Preemptive enteral nutrition enriched with eicosapentaenoic acid, gamma-linolenic acid and antioxidants in severe multiple trauma: a prospective, randomized, double-blind study. Intensive Care Med 2015; 41: 460–469.
74. Heyland D, Muscedere J, Wischmeyer PE, et al. A randomized trial of glutamine and antioxidants in critically ill patients. N Engl J Med 2013; 368: 1489–1497.
75. van Zanten ARH, Sztark F, Kaisers UX, et al. High-protein enteral nutrition enriched with immune-modulating nutrients vs standard high-protein enteral nutrition and nosocomial infections in the ICU: a randomized clinical trial. JAMA 2014; 312: 514–524.
76. Siriwardena AK, Mason JM, Balachandra S, et al. Randomised, double blind, placebo controlled trial of intravenous antioxidant (n-acetylcysteine, selenium, vitamin C) therapy in severe acute pancreatitis. Gut 2007; 56: 1439–1444.
77. Maderazo EG, Woronick CL, Hickingbotham N, et al. A randomized trial of replacement antioxidant vitamin therapy for neutrophil locomotory dysfunction in blunt trauma. J Trauma 1991; 31: 1142–1150.
78. Porter JM, Ivatury RR, Azimuddin K, et al. Antioxidant therapy in the prevention of organ dysfunction syndrome and infectious complications after trauma: early results of a prospective randomized study. Am Surg 1999; 65: 478–483.
79. Fowler AA Iii, Kim C, Lepler L, et al. Intravenous vitamin C as adjunctive therapy for enterovirus/rhinovirus induced acute respiratory distress syndrome. World J Crit Care Med 2017; 6: 85–90.
80. Bharara A, Grossman C, Grinnan D, et al. Intravenous vitamin C administered as adjunctive therapy for recurrent acute respiratory distress syndrome. Case Reports Crit Care 2016; 2016: 8560871.
81. Kim C, Debesa O, Nicolato P, et al. Vitamin C infusion for gastric acid aspiration-induced acute respiratory distress syndrome (ARDS). Pulm Res Respir Med Open J 2017; 4: 33–37.
82. Rumelin A, Jaehde U, Kerz T, et al. Early postoperative substitution procedure of the antioxidant ascorbic acid. J Nutr Biochem 2005; 16: 104–108.
83. Patak P, Willenberg HS, Bornstein SR. Vitamin C is an important cofactor for both adrenal cortex and adrenal medulla. Endocr Res 2004; 30: 871–875.
84. May JM, Qu Z, Meredith ME. Mechanisms of ascorbic acid stimulation of norepinephrine synthesis in neuronal cells. Biochem Biophys Res Commun 2012; 426: 148–152.
85. Seitz G, Gebhardt S, Beck JF, et al. Ascorbic acid stimulates DOPA synthesis and tyrosine hydroxylase gene expression in the human neuroblastoma cell line SK-N-SH. Neurosci Lett 1998; 244: 33–36.
86. Rahman MK, Rahman F, Rahman T, et al. Dopamine-β-hydroxylase (DBH), its cofactors and other biochemical parameters in the serum of neurological patients in Bangladesh. Int J Biomed Sci 2009; 5: 395–401.
87. Ul-Hasan S, Burgess DM, Gajewiak J, et al. Characterization of the peptidylglycine α-amidating monooxygenase (PAM) from the venom ducts of neogastropods, Conus bullatus and Conus geographus. Toxicon 2013; 74: 215–224.
88. Grino M, Guillaume V, Boudouresque F, et al. Glucocorticoids regulate peptidyl-glycine alpha-amidating monooxygenase gene expression in the rat hypothalamic paraventricular nucleus. Mol Endocrinol 1990; 4: 1613–1619.
89. Carr AC, Shaw GM, Fowler AA, et al. Ascorbate-dependent vasopressor synthesis: a rationale for vitamin C administration in severe sepsis and septic shock? Crit Care 2015; 19: 418.
90. Russell JA, Walley KR, Singer J, et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N Engl J Med 2008; 358: 877–887.
91. Baltalarli A, Ozcan V, Bir F, et al. Ascorbic acid (vitamin C) and iloprost attenuate the lung injury caused by ischemia/reperfusion of the lower extremities of rats. Ann Vasc Surg 2006; 20: 49–55.
92. Feng N-H, Chu S-J, Wang D, et al. Effects of various antioxidants on endotoxin-induced lung injury and gene expression: mRNA expressions of MnSOD, interleukin-1beta and iNOS. Chin J Physiol 2004; 47: 111–120.
93. Dwenger A, Pape HC, Bantel C, et al. Ascorbic acid reduces the endotoxin-induced lung injury in awake sheep. Eur J Clin Invest 1994; 24: 229–235.
94. Fisher BJ, Kraskauskas D, Martin EJ, et al. Mechanisms of attenuation of abdominal sepsis induced acute lung injury by ascorbic acid. Am J Physiol Lung Cell Mol Physiol 2012; 303: L20–L32.
95. US National Library of Medicine. Vitamin C infusion for treatment in Sepsis induced acute lung injury (CITRIS-ALI). Identifier NCT02106975, 2014, https://clinicaltrials.gov/ct2/show/NCT02106975
96. German Nutrition Society (DGE). New reference values for vitamin C intake. Ann Nutr Metab 2015; 67: 13–20.
97. Padayatty SJ, Sun H, Wang Y, et al. Vitamin C pharmacokinetics: implications for oral and intravenous use. Ann Intern Med 2004; 140: 533–537.
98. Burzle M, Hediger MA. Functional and physiological role of vitamin C transporters. Curr Top Membr 2012; 70: 357–375.
99. Padayatty SJ, Doppman JL, Chang R, et al. Human adrenal glands secrete vitamin C in response to adrenocorticotrophic hormone. Am J Clin Nutr 2007; 86: 145–149.
100. Boonen E, Vervenne H, Meersseman P, et al. Reduced cortisol metabolism during critical illness. N Engl J Med 2013; 368: 1477–1488.
101. Annane D. The role of ACTH and corticosteroids for sepsis and septic shock: an update. Front Endocrinol 2016; 7: 70.
102. Hornig D. Distribution of ascorbic acid, metabolites and analogues in man and animals. Ann N Y Acad Sci 1975; 258: 103–118.
103. Rom S, Zuluaga-Ramirez V, Reichenbach NL, et al. PARP inhibition in leukocytes diminishes inflammation via effects on integrins/cytoskeleton and protects the blood-brain barrier. J Neuroinflammation 2016; 13: 254.
104. Lorente L, Martin MM, Abreu-Gonzalez P, et al. Sustained high serum malondialdehyde levels are associated with severity and mortality in septic patients. Crit Care 2013; 17: R290.
105. Padayatty SJ, Sun AY, Chen Q, et al. Vitamin C: intravenous use by complementary and alternative medicine practitioners and adverse effects. PLoS ONE 2010; 5: e11414.
106. Padayatty SJ, Riordan HD, Hewitt SM, et al. Intravenously administered vitamin C as cancer therapy: three cases. CMAJ 2006; 174: 937–942.
107. Buehner M, Pamplin J, Studer L, et al. Oxalate nephropathy after continuous infusion of high-dose vitamin C as an adjunct to burn resuscitation. J Burn Care Res 2016; 37: e374–e379.
108. Chen Q, Espey MG, Krishna MC, et al. Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues. Proc Natl Acad Sci USA 2005; 102: 13604–13609.
109. Ried K, Travica N, Sali A. The acute effect of high-dose intravenous vitamin C and other nutrients on blood pressure: a cohort study. Blood Press Monit 2016; 21: 160–167.
110. Grossmann M, Dobrev D, Himmel HM, et al. Ascorbic acid-induced modulation of venous tone in humans. Hypertens 2001; 37: 949–954
111. Ayoglu H, Sezer U, Akin M, et al. Selenium, copper, zinc, iron levels and mortality in patients with sepsis and systemic inflammatory response syndrome in Western Black Sea Region, Turkey. J Pak Med Assoc 2016; 66: 447–452.
112. Zafar MSH, Wani JI, Karim R, et al. Significance of serum magnesium levels in critically ill-patients. Int J Appl Basic Med Res 2014; 4: 34–37.
113. Donnino MW, Carney E, Cocchi MN, et al. Thiamine deficiency in critically ill patients with sepsis. J Crit Care 2010; 25: 576–581.
114. de Haan K, Groeneveld ABJ, de Geus HRH, et al. Vitamin D deficiency as a risk factor for infection, sepsis and mortality in the critically ill: systematic review and meta-analysis. Crit Care 2014; 18: 660.
115. Australian New Zealand Clinical Trials Registry. Vitamin C administration in severe sepsis. Trial ID ACTRN12617001184369, https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373436
116. US National Library of Medicine. High dose intravenous ascorbic acid in severe sepsis. Identifier NCT02734147, 2016, https://clinicaltrials.gov/ct2/show/NCT02734147
117. Australian New Zealand Clinical Trials Registry. Vasopressor dependent shock: intravenous vitamin C versus placebo on vasopressor use. The VALENCIA study. Trial ID ACTRN12617001392358, https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373728
118. US National Library of ClinicalTrials.gov. Vitamin C and septic shock. Identifier NCT03338569, 2017, https://clinicaltrials.gov/ct2/show/NCT03338569
119. US National Library of Medicine. The vitamin C, hydrocortisone and thiamine in patients with septic shock trial (VITAMINS). Identifier NCT03333278, 2017, https://clinicaltrials.gov/ct2/show/NCT03333278
120. US National Library of Medicine. The effect of vitamin C, thiamine and hydrocortisone on clinical course and outcome in patients with severe sepsis and septic shock. Identifier NCT03335124, 2017, https://clinicaltrials.gov/ct2/show/NCT03335124
121. US National Library of Medicine. Evaluation of hydrocortisone, vitamin C and thiamine for the treatment of septic shock (HYVITS). Identifier NCT03380507, 2017, https://clinicaltrials.gov/ct2/show/NCT03380507
122. US National Library of Medicine. Ascorbic acid, corticosteroids, and thiamine in sepsis (ACTS) trial. Identifier NCT03389555, 2018, https://clinicaltrials.gov/ct2/show/NCT03389555
123. US National Library of Medicine. Metabolic resuscitation using ascorbic acid, thiamine, and glucocorticoids in sepsis (ORANGES). Identifier NCT03422159, 2018, https://clinicaltrials.gov/ct2/show/NCT03422159

Supplementary Material

Supplemental Material

Supplemental material files:

File (supplement_prisma_checklist_.docx)

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: October 19, 2018
Issue published: January-December 2018

Keywords

  1. Critical care
  2. emergency medicine
  3. respiratory medicine
  4. sepsis
  5. nutrition
  6. respiratory distress
  7. shock
  8. vitamin C
  9. acute respiratory distress syndrome

Rights and permissions

© The Author(s) 2018.
Creative Commons License (CC BY-NC 4.0)
This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Request permissions for this article.
Request Permissions
PubMed: 30364374

Authors

Affiliations

Michael Zhang
Department of Medicine, VA Medical Center, Cleveland, OH, USA
David F Jativa
Department of Medicine, Aventura Hospital & Medical Center, Aventura, FL, USA

Notes

Michael Zhang, Department of Medicine, VA Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA. Email: [email protected]

Metrics and citations

Metrics

Journals metrics

This article was published in SAGE Open Medicine.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 7366

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 39 view articles Opens in new tab

Crossref: 40

  1. Investigational pharmacological agents for the treatment of ARDS
    Go to citation Crossref Google Scholar
  2. Challenges in Septic Shock: From New Hemodynamics to Blood Purificatio...
    Go to citation Crossref Google Scholar
  3. Rubi idaei fructus as a Source of Bioactive Chemical Compounds with an...
    Go to citation Crossref Google Scholar
  4. Health Benefits, Characterization and Biochemical Analysis of Beet Roo...
    Go to citation Crossref Google Scholar
  5. Effect of intravenous vitamin C on adult septic patients: a systematic...
    Go to citation Crossref Google Scholar
  6. Low Levels of Serum and Intracellular Vitamin C in Hospitalized COVID-...
    Go to citation Crossref Google Scholar
  7. The Role of Diet and Specific Nutrients during the COVID-19 Pandemic: ...
    Go to citation Crossref Google Scholar
  8. Association of Vitamin C Treatment with Clinical Outcomes for COVID-19...
    Go to citation Crossref Google Scholar
  9. Clinical nutrition approach in medical management of COVID-19 hospital...
    Go to citation Crossref Google ScholarPub Med
  10. Parenteral Vitamin C in Patients with Severe Infection: A Systematic R...
    Go to citation Crossref Google Scholar
  11. Vitamin C Deficiency in Critically Ill Children: Prospective Observati...
    Go to citation Crossref Google Scholar
  12. IV Vitamin C in Critically Ill Patients: A Systematic Review and Meta-...
    Go to citation Crossref Google Scholar
  13. Plasma Ascorbic Acid Levels in Critically Ill Pediatric Patients
    Go to citation Crossref Google Scholar
  14. Interplay of Nutrition and Psychoneuroendocrineimmune Modulation: Rele...
    Go to citation Crossref Google Scholar
  15. Safety and effectiveness of high-dose vitamin C in patients with COVID...
    Go to citation Crossref Google Scholar
  16. Any Role of High-Dose Vitamin C for Septic Shock in 2021?
    Go to citation Crossref Google Scholar
  17. Vitamin C in Critically Ill Patients: An Updated Systematic Review and...
    Go to citation Crossref Google Scholar
  18. Therapeutic Approaches in Modulating the Inflammatory and Immunologica...
    Go to citation Crossref Google Scholar
  19. Vitamin C and Sepsis
    Go to citation Crossref Google Scholar
  20. The efficacy of intravenous vitamin C in critically ill patients: A me...
    Go to citation Crossref Google Scholar
  21. Micronutrients in Sepsis and COVID-19: A Narrative Review on What We H...
    Go to citation Crossref Google Scholar
  22. Clinical characteristics of hospitalised patients with COVID-19 and th...
    Go to citation Crossref Google Scholar
  23. Vitamin C in the Treatment of COVID-19
    Go to citation Crossref Google Scholar
  24. Витамин С при критических состояниях: от эксперимента к клинике (часть...
    Go to citation Crossref Google Scholar
  25. RETRACTED: Clinical and Scientific Rationale for the “MATH+” Hospital ...
    Go to citation Crossref Google ScholarPub Med
  26. Response
    Go to citation Crossref Google Scholar
  27. Obesity, malnutrition, and trace element deficiency in the coronavirus...
    Go to citation Crossref Google Scholar
  28. Vitamin C—An Adjunctive Therapy for Respiratory Infection, Sepsis and ...
    Go to citation Crossref Google Scholar
  29. The Long History of Vitamin C: From Prevention of the Common Cold to P...
    Go to citation Crossref Google Scholar
  30. Adjunctive therapy with vitamin c and thiamine in patients treated wit...
    Go to citation Crossref Google Scholar
  31. Could Vitamins Help in the Fight Against COVID-19?
    Go to citation Crossref Google Scholar
  32. Vitamin C and thiamine are associated with lower mortality in sepsis
    Go to citation Crossref Google Scholar
  33. Treating sepsis with vitamin C, thiamine, and hydrocortisone: Explorin...
    Go to citation Crossref Google Scholar
  34. Nutrition in Sepsis: A Bench-to-Bedside Review
    Go to citation Crossref Google Scholar
  35. Vitamin C biochemistry: From scurvy to COVID-19 treatment
    Go to citation Crossref Google Scholar
  36. Vitamin C administration in the critically ill: a summary of recent me...
    Go to citation Crossref Google Scholar
  37. Effects of different ascorbic acid doses on the mortality of criticall...
    Go to citation Crossref Google Scholar
  38. Vitamin C in surgical sepsis
    Go to citation Crossref Google Scholar
  39. Therapeutic Perspective of Vitamin C and Its Derivatives
    Go to citation Crossref Google Scholar
  40. Pharmacological therapies for acute respiratory distress syndrome
    Go to citation Crossref Google Scholar

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:


Alternatively, view purchase options below:

Access journal content via a DeepDyve subscription or find out more about this option.