You are on page 1of 9

SURGICAL INFECTIONS

Volume 19, Number X, 2018 Review Article


ª Mary Ann Liebert, Inc.
DOI: 10.1089/sur.2018.111

Patterns of Death in Patients with Sepsis


and the Use of Hydrocortisone, Ascorbic Acid,
and Thiamine to Prevent these Deaths

Paul E. Marik
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

Abstract

Background: In general, patients with sepsis die from the host response to the infecting pathogen rather than
from the infecting pathogen itself. Four patterns of death have been identified in sepsis, namely vasoplegic
shock, single-organ respiratory failure (acute respiratory distress syndrome [ARDS]), multi-system organ
failure (MSOF), and persistent MSOF with ongoing inflammation and immunosuppression with recurrent
infections (persistent inflammation-immunosuppression and catabolism syndrome [PICS]). To improve the
outcome of sepsis adjunctive therapies that modulate the immune system have been tested; these therapies that
have targeted specific molecules or pathways have universally failed.
Conclusion: We propose that the combination of hydrocortisone, intravenous ascorbic acid, and thiamine (HAT
therapy), which synergistically targets multiple pathways, restores the dysregulated immune system and organ
injury, and reduces the risk of death and organ failure following sepsis.

Keywords: hydrocortisone; multi-system organ failure; sepsis; thiamine; vitamin C

T he global burden of sepsis is substantial with an es-


timated 32 million cases and 5.3 million deaths per year
with most of these cases occurring in low income countries
The first pattern is that of refractory vasoplegic septic
shock caused by a cytokine storm, leading to death within
96 hours [10,11]. The second pattern is that of death caused
[1]. Data from the United States and Australia demonstrate by single-organ respiratory failure (acute respiratory distress
that over the last two ecades the annual incidence of sepsis syndrome [ARDS]) resulting from the combined effects of an
has increased by approximately 13% with a decrease in in- exuberant proinflammatory response combined with volume
hospital mortality from approximately 35% to 20% [2–4]. In overload; death usually occurs between three and seven days
2013 more than 1.3 million patients were hospitalized in the of onset [9,12,13]. The third pattern of death is from multi-
United States with a diagnosis of sepsis, of whom more than system organ failure (MSOF) caused by cellular bioenergetic
300,000 died [5]. Despite the vast number of patients who die failure with cell hibernation, with death typically occurring
from sepsis and our increasing understanding of the patho- between four days and two weeks [9,14]. The final pattern is
physiology of this syndrome the patterns of death in patients one of prolonged and persistent MSOF with ongoing inflam-
with sepsis have not been well defined [6]. To improve the mation and immunosuppression with recurrent and unresolved
outcome of this deadly disease it is important to understand infection (persistent inflammation-immunosuppression and
why patients with sepsis die. In this article, we propose four catabolism syndrome [PICS]) [15,16]. It is likely that the
patterns of death related to different but inter-related patho- characteristics of the pathogen (type of pathogen and mi-
physiologic mechanisms and suggest that all four patho- crobial load) together with host factors (site of infection, age,
physiologic processes are influenced positively by treatment immune status, comorbidities, and genetic makeup) as well
with intravenous vitamin C (ascorbic acid), corticosteroids, as treatment factors (delay in initiating treatment, inappro-
and thiamine (HAT therapy; Figs. 1 and 2). These four priate antibiotic agents, fluid overload, adjunctive treatment
phenotypic patterns are supported by clinical, immunologic, modalities) interact to determine the outcome and pattern of
and metabolomic data [7–9]. death.

Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, Virginia.

1
2 MARIK

Synergistic Anti-Inflammatory Properties


of HAT Therapy

Sepsis is fundamentally an inflammatory disease medi-


ated by the activation of the innate immune system by
both pathogen-associated molecular patterns (PAMPs) and
damage-associated molecular patterns (DAMPs). Calvano
et al. [19] demonstrated that exposure of blood leukocytes to
endotoxin altered the expression of 3,714 genes. These in-
clude genes for pro- and anti-inflammatory cytokines, che-
mokines, adhesion molecules, transcription factors, enzymes,
clotting factors, stress proteins, and anti-apoptotic molecules
[20]. The excess production of proinflammatory mediators
plays an important role in four pathogenetic mechanism
leading to death in patients with sepsis. We believe that HAT
therapy has potent anti-inflammatory properties that under-
lies the benefit of this treatment in patients with sepsis.
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

FIG. 1. The four patterns of sepsis related deaths. SIRS = Vitamin C suppresses activation of nuclear factor kappa-B
systemic inflammatory response syndrome; CARS = com- (NF-kB) by inhibiting tumor necrosis factor-a (TNF-a)–
pensatory anti-inflammatory response syndrome; MSOF = induced phosphorylation of inhibitory kappa-B kinase (IkB
multi-system organ failure; PICS = persistent inflammation- kinase) [21]. The primary anti-inflammatory action of gluco-
immunosuppression and catabolism syndrome. corticoids is to repress the transcriptional activity of NF-kB and
activator protein-1 (AP-1). Nuclear factor kappa-B and AP-1
increase the transcription of multiple proinflammatory media-
A limited number of studies have evaluated the causes of tors that contribute to the generation of reactive oxygen species
death in patients with sepsis. Vincent et al. [7] analyzed the (ROS), cellular injury, coagulation activation, and the endo-
cause of death in 4,459 patients with severe sepsis. In this thelial and microvascular dysfunction characteristic of sepsis.
study, 27% of patients died during the 28-day study period. Ascorbic acid decreases high mobility group box 1 (HMGB1)
Twenty-two percent of patients died of refractory septic secretion [22]; HMGB1 is an important late proinflammatory
shock, 13% from respiratory failure, and 43% died from cytokine. Histamine has been shown to play an important role
MSOF. Frencken et al. [17] studied the patterns of death in sepsis [23]. Vitamin C has been demonstrated to decrease the
among 708 patients with severe sepsis and septic shock ad- synthesis and to inactivate histamine [24].
mitted to two Dutch intensive care units (ICUs). The one-year Sepsis is characterized by the excessive production of ROS
mortality was 53.8% with 12% of patients dying within four by the induction of enzymes such as nicotinamide adenine
days of admission, 20% between days four and 28, and 22% dinucleotide phosphate-oxidase (NOX) and the uncoupling
between day 29 and one year. Weiss et al. [18] evaluated the of mitochondrial oxidative phosphorylation and iNOS [25].
patterns of death in pediatric sepsis. The median time to death In addition, ROS are produced by xanthine oxidase, lipox-
was eight days. The most common cause of death was re- ygenase, and cyclo-oxygenase. When the antioxidant de-
fractory shock (34%), followed by MSOF (27%) and single- fenses are overwhelmed, ROS can induce injury to lipids,
organ respiratory failure (9%). Early deaths (£3 d) were proteins, and nucleic acids thereby resulting in widespread
mostly caused by refractory septic shock whereas MSOF endothelial dysfunction, mitochondrial dysfunction, cellular
predominated after three days. injury, and multiple organ dysfunction. Unopposed ROS
oxidizes tetrahydrobiopterin (BH4), the cofactor of endo-
thelial nitric oxide synthase (eNOS), and thereby reduces
eNOS activity, the enzyme producing endothelial nitric oxide
[25]. Endothelial nitric oxide (eNO) maintains microcircu-
latory flow and inhibits platelet aggregation and adhesion of
activated platelets and leukocytes. In the absence of BH4,
eNOS becomes uncoupled, producing superoxide (O2-) ra-
ther than nitric oxide (NO). Superoxide (O2-) and NO yield
peroxynitrite, a more damaging ROS. Vitamin C is a key
cellular antioxidant, detoxifying exogenous oxidant radical
species that have entered cells or that have arisen within cells
because of excess O2-generation by mitochondrial metabolism,
by NOX, xanthine oxidase, or by uncoupled nitric oxide syn-
thase (NOS) [25,26]. In addition, vitamin C recycles other an-
tioxidants including a-tocopherol (vitamin E) and BH4.
Tetrahydrobiopterin plays a critical role in the function of eNOS.
Vitamin C deficiency results in the incomplete regeneration of
FIG. 2. Treatment with hydrocortisone, ascorbic acid, and BH4 resulting in the uncoupling of eNOS and the generation of
thiamine (HAT therapy) attenuates both the pro- and anti- superoxide and peroxynitrite.[25] Thiamine may act together
inflammatory response in patients with sepsis. with vitamin C to decrease oxidative injury [27,28].
HAT THERAPY AND SEPSIS 3

We propose that HAT therapy may have synergistic anti- Prevention of vasoplegic shock with HAT therapy
inflammatory and anti-oxidative properties thereby modu- Vitamin C and hydrocortisone act via multiple mechanism
lating the proinflammatory response of sepsis. This postulate to reverse vasoplegic shock and restore organ perfusion. Both
is supported by experimental studies. Barabutis et al. [29]. molecules suppress activation of NF-kB; NF-kB increase
have demonstrated that hydrocortisone together with vitamin C the transcription of multiple proinflammatory mediators
protects the vascular endothelium from damage by endotoxin responsible for the cytokine storm [21]. Vitamin C is an
while neither agent alone had this effect. Azari et al. [30] essential cofactor for the synthesis of norepinephrine,
compared the protective effects of vitamin C, vitamin E, and epinephrine, and vasopressin; in addition, vitamin C in-
hydrocortisone alone and in combination, in a renal and in- creases adrenergic transmission [43]. In a murine cecal li-
testinal ischemia-reperfusion model [31]. In these studies gation and puncture (CLP) model, prophylactic bolus
both vitamin C and hydrocortisone reduced the ischemia- injection of vitamin C attenuated the loss of arteriolar and
reperfusion injury with the combination having synergistic blood pressure responsiveness to the vasoconstrictors nor-
protective effects. Many interventions in critically ill patients epinephrine and angiotensin [44–46]. Glucocorticoids are
are time sensitive; the use of antibiotic agents and source potent anti-inflammatory agents that suppress inflammation
control in patients with sepsis are examples in which a delay by multiple mechanisms that involve both the innate and
in the initiation of therapy may adversely affect outcomes adaptive immune responses. In addition to attenuating the
[32,33]. Similarly, it is likely the benefits of HAT therapy cytokine storm, low-dose glucocorticoids have immune-
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

may diminish with delays in administration. It is important to stimulating effects that may limit the anti-inflammatory im-
emphasize that early initiation of HAT therapy is critical to a munosuppressive state. In addition, glucocorticoids have
good outcome as the pathologic processes leading to organ additional beneficial effects including increasing adrenergic
failure become less reversible with time. Furthermore, it is responsiveness [47] and preserving the endothelial glycoca-
likely that fluid overload with an aggressive approach to fluid lyx [48]. Based on the overlapping beneficial effects of
resuscitation attenuates the benefit of HAT treatment [34–36]. vitamin C and glucocorticoids in experimental models of
sepsis [49,50], we have postulated that these two agents may
Refractory Vasoplegic Shock and the Cytokine Storm act synergistically to reverse vasoplegic shock [51,52]. In a
retrospective before and after study we have demonstrated
The early phase of sepsis is generally believed to result that HAT therapy rapidly reserves vasoplegic shock with a
from the uncontrolled production of proinflammatory medi- reduction in the dose of vasopressors within two hours of the
ators, the so-called cytokine storm, which may lead to re- initiation of treatment with vasopressor independence within
fractory vasoplegic shock [37,38]. The early proinflammatory 24 hours [52].
response to bacteremic shock can be reproduced by admin-
istration of proinflammatory cytokines [39–41]. Using ad-
vanced informatics techniques from sepsis transcriptomic Acute Respiratory Distress Syndrome
datasets, Sweeney et al. [8] identified three functional tran- According to the Berlin Definition, ARDS is defined as an
scriptomic subtypes that they termed ‘‘inflammopathic, ‘‘acute diffuse, inflammatory lung injury, leading to in-
adaptive, and coagulopathic.’’ The inflammopathic subtype creased pulmonary vascular permeability, increased lung
was characterized by overactivation of the innate immune weight, and loss of aerated lung tissue with hypoxemia and
system with increased expression of proinflammatory sig- bilateral radiographic opacities’’ [53]. Sepsis is the most
naling pathways, pattern recognition receptor activity, and common cause of ARDS [54] and results from intra-alveolar
complement activation. Clinical features of this subtype in- macrophage activation with elaboration of a diverse array of
cluded a high disease severity with shock, high bandemia, and bioactive mediators including proteases, ROS, cytokines, and
high mortality. Although the early systemic inflammatory chemokines [13]. Because ARDS is caused by the excess
response has been considered the hallmark of sepsis, immu- production of proinflammatory mediators, HAT therapy
nosuppression occurs both early and late in the host sepsis rapidly reverses the pathogenic mechanism of ARDS [52,55].
response.
Refractory vasodilatory shock develops from uncontrolled
Multi-System Organ Failure
vasodilation and vascular hyporesponsiveness to endogenous
vasoconstrictors, causing failure of physiologic vasor- Multi-system organ failure is the most common cause of
egulatory mechanisms [11]. Vasoplegic shock results from death in patients with sepsis. These patients usually develop
failure of the vascular smooth muscle to constrict with pro- progressive and irreversible organ failure requiring the in-
found arterial and venodilatation [10]. Vasoplegic shock is creasing use of advanced life support measures. Because of
believed to be caused by the massive production of proin- the high burden of ongoing treatment with the extremely
flammatory cytokines with increased expression of inducible grave prognosis families will often elected to limit care al-
nitric oxide synthetase with increased production of nitric lowing for a more peaceful death.
oxide (NO), activation of KATP channels resulting in hyper- What is the mechanism of MSOF in sepsis? The prevailing
polarization of the muscle cell membrane, increased pro- theory suggests that MSOF results from impaired oxygen
duction of natriuretic peptides (which act synergistically with delivery at the cellular level because of altered microcircu-
NO), and a relative vasopressin deficiency [10]. These factors latory blood flow [56,57]. Although global oxygen delivery is
lead to profound vasodilation with hypotension and vaso- typically increased in sepsis, proponents of this theory sug-
pressor resistance. This hemodynamic pattern is complicated gest that many tissue capillary beds do not receive adequate
by decreased contractility with decreased myocardial per- oxygen supplies because of microvascular endothelial dys-
formance and diastolic dysfunction [42]. function. However, there are few data that tissue hypoxia
4 MARIK

occurs in patients with sepsis. Multiple experimental models likely that the unbalanced production of mitochondrial re-
have failed to demonstrate cellular hypoxia in sepsis [58–61]. active oxygen species (mtROS) impairs mitochondrial
Increasing oxygen delivery in patients with sepsis does not structure, enzymatic function and biogenesis and play a role
increase oxygen consumption [62–64]. A number of ran- in the mitochondrial dysfunction of sepsis. These data sug-
domized controlled trials have failed to demonstrate an im- gest that the bioenergetic failure in sepsis leads to organ
provement in outcome in patients with sepsis who received failure particularly in those organs with a high metabolic
therapy that aimed to increase oxygen delivery compared with demand and high mitochondrial concentration [79–84,86].
those who received conventional therapy [65–67]. These The bioenergetic failure does not lead to widespread cell
studies suggest that microcirculatory impairment with shunting necrosis or apoptosis but rather a state of cellular hibernation
and maldistribution of blood flow are unlikely to be central to [72]. Furthermore, the mitochondrial oxidative injury re-
the pathophysiology of sepsis-induced organ failure. leases mitochondrial DNA (mtDNA), which amplifies the
inflammatory injury leading to further immune paresis and
PICS syndrome [87].
Metabolic failure as a cause of sepsis-related MOSF
In patients with sepsis who die of MSOF histopathologic
Prevention of MSOF with HAT treatment
examination of the heart, liver, and kidneys have failed to
demonstrate necrotic or apoptotic cell death [68,69]. Takasu Critically ill patients with sepsis typically have low or
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

et al. [69] performed rapid postmortem cardiac and renal undetectable serum levels of vitamin C resulting in an acute
harvest in 44 patients with sepsis and compared the immu- scorbutic condition [52,88–90]. Recently Carr et al. [91]
nohistochemistry features with those of control organs. Im- demonstrated that 100% of patients with sepsis had low
munohistochemistry demonstrated low levels of apoptotic vitamin C levels. Low plasma concentrations are associated
cardiomyocytes in septic and control subjects, however, with the severity of organ failure and mortality [89]. Fur-
electron microscopy showed hydropic mitochondria in the thermore, low vitamin C levels have been demonstrated to be
septic specimens. It is noteworthy that per milligram of tis- predictive of the development of MSOF [90].
sue, the heart and kidney consume the most oxygen and The overlapping anti-inflammatory properties of gluco-
possess the greatest abundance of mitochondria [70]. The corticoids and vitamin C reduce the production of proin-
authors of these studies have concluded that the degree of cell flammatory mediators and ROS, which are linked to the
injury and death does not account for severity of sepsis- development of MSOF. In experimental models, parenteral
induced organ dysfunction. Similarly, animal models have vitamin C attenuates organ injury and improves survival in
failed to reveal widespread cell death in sepsis despite pro- mice with sepsis [92–94]. Dehydroascorbic acid is trans-
gressive organ failure [71]. To explain these findings ported via the GLUT1 transporter into mitochondria, where it
Hotchkiss et al. [72] have suggested that these features are converted to ascorbic acid and acts as a potent antioxidant
consistent with cell stunning or hibernation and that these limiting mitochondrial oxidant injury [95,96]. In an experi-
changes may represent a short-term adaptive response. mental model, Dhar-Mascareno et al. [97] demonstrated that
Multiple defects in mitochondrial function have been de- oxidant-induced mitochondrial damage and apoptosis in
scribed in animal models and patients with sepsis. These human endothelial cells were inhibited by vitamin C. Studies
include abnormal autophagy, decreased biogenesis (mito- using N-acetylcysteine have proven to be ineffective and
chondrial numbers), decreased activity of the components of potentially harmful in patients with sepsis possibly because
the electron transport chain (particularly complex I), and of the limited ability of this drug to enter into the mito-
uncoupling of oxidation phosphorylation [73–80]. Altera- chondria and its inability to regenerate BH4 [25,98,99]. As-
tions in fatty acid metabolism with decreased b-oxidation and corbic acid is required for the synthesis of carnitine, which is
abnormalities of the citric acid cycle appear to be a charac- required for the transport of fatty acids into the mitochondrial
teristic feature of the mitochondrial dysfunction in sepsis matrix and for b-oxidation [81,95].
[81,82]. These changes lead to decreased adenosine tri- Thiamine is the precursor of thiamine pyrophosphate, the
phosphate (ATP) production. Furthermore, damaged mito- essential coenzyme of several decarboxylases required for
chondria undergo mitophagy, i.e., autophagy directed at the glucose metabolism, the Krebs cycle, the generation of ATP,
removal of damaged mitochondria [83,84]. Matkovich et al. the pentose phosphate pathway, and the production of
[85] measured messenger RNA (mRNA) alterations in hearts NADPH. Thiamine pyrophosphate (TPP) is a critical coen-
from patients who died from sepsis and compared these with zyme for the pyruvate dehydrogenase complex, the rate-
mRNA expression in non-failing and failing human hearts. In limiting step in the citric acid cycle. Thiamine deficiency is
this study, mRNA expression levels for 198 mitochondrially common among patients with sepsis, with a range in preva-
localized energy production components, including Krebs lence between 20% and 70%, depending on measurement
cycle and electron transport genes, were reduced on average techniques and inclusion criteria [100–105]. A deficiency in
by 43%. Except for mitochondrial isocitrate dehydrogenase, thiamine leads to decreased activity of thiamine-dependent
all of the other 19 genes involved in the tricarboxylic acid enzymes that triggers a sequence of metabolic events leading
(TCA) cycle were decreased. In a sepsis model, Arulkumaran to energy compromise and decreased ATP production. It is
et al. [61] demonstrated that sepsis-induced renal dysfunction likely that thiamine deficiency compounds the mitochondrial
was associated with reduced nicotinamide adenine dinucle- injury and bioenergetic failure caused by vitamin C deple-
otide (NADH) and mitochondrial membrane potential tion. In a pilot randomized controlled trial, Donnino et al.
(MMP) with increased renal ROS production. In this study, [105] randomly assigned 88 patients with septic shock to
4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl, a free rad- receive 200 mg thiamine twice daily for seven days. In the
ical scavenger was protective against renal tubule injury. It is pre-defined subgroup of patients with thiamine deficiency,
HAT THERAPY AND SEPSIS 5

those in the thiamine treatment group had statistically sig- both chronic immune suppression and inflammation. This
nificantly lower lactate levels at 24 hours and a lower mor- syndrome has been termed the persistent inflammation/
tality at 30 days. Furthermore, in a secondary analysis of this immunosuppression and catabolism syndrome (PICS) [15,108].
study the need for renal replacement therapy and the serum This persistent inflammation is characterized by increased
creatinine were greater in the placebo group [106]. The ob- concentration of plasma interleukin (IL)-6, a persistent acute
servation that thiamine was reno-protective is an important phase response, neutrophilia, with increased immature
observation considering the high metabolic demand of the granulocyte count, anemia, lymphopenia, and, often, tachy-
kidney and the postulated role of thiamine in improving en- cardia [108]. Although these patients are profoundly im-
ergy metabolism. This finding is all the more important be- munosuppressed, inflammation is ongoing. While the exact
cause correction of mean arterial pressure and macrovascular pathophysiology of PICS syndrome is unclear, PICS is prob-
blood flow is not restorative in septic acute kidney injury ably driven by DAMPs and alarmins that are produced by
(AKI), and supports of the concept that mitochondrial dys- injured organs and tissues, such as mtDNA, nucleosides, his-
function and injury may be a critical feature of septic AKI [84]. tones, HMGB1, protein S100A, ATP, adenosine and/or hya-
Several studies suggest that vitamin C alone and when luronan products. The paradoxical immunosuppression and
combined with glucocorticoids and thiamine reduce the risk infectious complications in patients with sepsis compound as
of MSOF. Nathens et al. [107] randomly assigned 595 sur- sepsis progresses, with a shift to infection by opportunistic
gical intensive care (ICU) patients to receive intravenous organisms. Torgersen et al. [16] studied both clinical and
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

vitamin C and vitamin E for up to 28 days. The vitamin postmortem records of patients (n = 235) who died from sepsis
combination was associated with a significant reduction in the or septic shock in their institution over a period of 10 years.
incidence of MSOF (p = 0.04). The Sepsis-Related Organ Multiple organ dysfunction syndrome (MODS) was consid-
Failure Assessment (SOFA) score has been used to quantify ered the main cause of death in 52% of patients. An unresolved
the severity of organ dysfunction in patients with sepsis [14]. In infectious focus was present at autopsy in 77% of patients.
a pilot study of 24 patients with severe sepsis and septic shock, Moreover, 90% of the patients with prolonged intensive care
Fowler et al. [88] demonstrated a significant decrease in the exceeding seven days had persistent infectious foci.
SOFA score in patients randomly assigned to receive intrave-
nous vitamin C. Similarly in our observational study in which
Prevention of PICS with HAT therapy
we compared treatment with HAT therapy versus standard
treatment, we demonstrated a rapid decrease in the SOFA score The use of HAT therapy reduces the proinflammatory re-
in patients who received HAT therapy (Fig. 3) [52]. sponse and the development of MSOF and as such limits
progression to PICS syndrome. Moreover, HAT therapy
Persistent Inflammation-Immunosuppression specifically decreases the immunosuppression associated
and Catabolism Syndrome with sepsis. It has been known for more than 60 years that
vitamin C has immune-enhancing properties. In 1949. Fred
Patients with MSOF who survive the first two weeks of Klenner from Reidsville, North Carolina, reported on the use
sepsis often have a protracted clinical trajectory and exhibit of intravenous vitamin C in the treatment of polio and other
viral illnesses [109]. It was initially assumed that vitamin C
was directly viricidal (in vivo) and this mistaken belief un-
derlies the recommendations of Linus Pauling who promoted
the use of large doses of oral vitamin C (up to 18 g/d) for the
prevention and treatment of the common cold [110]. How-
ever, a number of randomized controlled trials have reported
that vitamin C supplementation had no effect on the inci-
dence of the common cold [111]. Whereas high-dose vitamin C
has in vitro viricidal properties [112,113], there are no data or
physiologic rationale to suggest that this occurs in vivo.
Rather, the anti-viral effects of vitamin C are likely because
vitamin C has specific immune-enhancing effects. Vitamin C
is concentrated in leucocytes, lymphocytes, and macro-
phages, reaching high concentrations in these cells [114].
Vitamin C improves chemotaxis, enhances neutrophil
phagocytic capacity and oxidative killing, stimulates inter-
feron production, and supports lymphocyte proliferation
[115–117]. In addition, vitamin C has been demonstrated to
attenuate the formation of neutrophil-extracellular-traps
FIG. 3. Time course of the Sepsis-Related Organ Failure (NETs) [94,118]. Vitamin C stimulates dendritic cells to
Assessment (SOFA) score over the four-day treatment pe- secrete IL-12 and thereby drive naı̈ve CD4+ T cells to dif-
riod in the treatment group and in the control group survi-
ferentiate into TH1 cells [118]. Vitamin C improves the
vors and non-survivors. *p < 0.001 for comparison of
treatment group vs control group. (Reproduced with per- generation of NK-cell progenitors from T/NK-cell progeni-
mission from Marik PE, Khangoora V, Rivera R, et al. tors [119]. Vitamin C promotes T-cell maturation, possibly
Hydrocortisone, vitamin C and thiamine for the treatment of by epigenetic mechanisms [120]. In a CLP sepsis model, Gao
severe sepsis and septic shock: A retrospective before-after et al. [121] demonstrated that parenteral vitamin C improved
study. Chest 2017;151:1229–1238). the outcome of sepsis and sepsis-induced MODS in wild-type
6 MARIK

as well as Gulo-/- mice. In this study negative immuno- 3. Stevenson EK, Rubenstein AR, Radin GT, et al. Two dec-
regulation by CD4+CD25+ regulatory T cells (Tregs) and the adesof mortality trends among patients with severe sepsis: A
secretion of inhibitory cytokines (TGF-b and IL-10) were comparative meta-analysis. Crit Care Med 2014;42:625–631.
inhibited and T-cell–mediated immunosuppression was im- 4. Kaukonen KM, Bailey M, Suzuki S, et al. Mortality re-
proved in those mice treated with vitamin C. lated to severe sepsis and septic shock among critically ill
Many physicians are reluctant to prescribe a short course patients in Australia and New Zealand, 2000–2012.
of low-dose glucocorticoid on the basis that these drugs in- JAMA 2014;311:1308–1316.
crease the risk of infections. The prolonged (>10 d) use of 5. Torio CM, Moore B1J. National Inpatient Hospital Costs:
moderate- to high-dose glucocorticoids (>400 mg hydrocor- The Most Expensive Conditions by Payer, 2013: Statis-
tisone per day) is well known to increase the risk of oppor- tical Brief #240. Healthcare Costs and Utilization Project
(HCUP) Statistical Briefs. Rockville, MD: Agency for
tunistic infections. In a landmark study, Keh et al. [122]
Healthcare Research and Quality. 2016.
performed a randomized, cross-over study that evaluated the
6. Tenhunen JJ. In death, truth lies: Why do patients with
clinical and immunologic effects of low-dose glucocorticoids sepsis die? Anesth Analg 2009;108:1731–1733.
in patients with septic shock. These investigators demon- 7. Vincent JL, Nelson DR, Williams MD. Is worsening
strated that hydrocortisone simultaneously decreased circu- multiple organ fialure the cuase of death in patients with
lating levels of both pro- and anti-inflammatory cytokines. severe sepsis? Crit Care Med 2011;39:1050–1055.
Hydrocortisone downregulated the proinflammatory immune 8. Sweeney TE, Azad TJ, Donato M, et al. Unsupervised
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

response but did not inhibit the TH1-related response and analysis of transcriptomics in bacterial sepsis across
concentrations of IL-12 and interferon-g were not sup- multiple datasets reveals three robust clusters. Crit Care
pressed. Paradoxically, by enhancing macrophage function Med 2018;46:915–992.
and the innate immune system and blunting the anti- 9. Villar J, Martinez D, Mosteiro F, et al. Is overall mortality
inflammatory response short-term glucocorticoids may re- the right composite endpoint in clinical trials of acute
duce the risk of infection and the development of PICS respiratory distress syndrome. Crit Care Med 2018; 46:
syndrome (Fig. 2). Roquilly et al. [123] randomly assigned 892–-899.
149 multi-trauma patients to receive a continuous infusion of 10. Landry DW, Oliver JA. Pathogenesis of vasodilatory
hydrocortisone (200 mg/d for 5 d) or placebo (the HYPO- shock. N Engl J Med 2001;345:588–595.
LYTE Study). The primary end point of this study was 11. Jentzer JC, Vallabhajosyula S, Khanna AK, et al. Man-
hospital-acquired pneumonia within 28 days. In the intention agement of refractory vasodilatory shock. Chest 2018;
to treat analysis, 35% of the patients treated with hydrocor- S0012-3692:30072-2.
tisone and 51% of those treated with placebo developed 12. Ware LB, Matthay MA. The acute respiratory distress
pneumonia (p = 0.007). In the three recent large randomized syndrome. N Engl J Med 2000;342:1334–1349.
13. Mac Sweeney R, McAuley DF. Acute respiratory distress
controlled trials, low-dose short-duration treatment with
syndrome. Lancet 2016;388:2416–2430.
glucocorticoids was not associated with an increased risk of
14. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-
secondary infections [124–126]. These data suggest that the related Organ Failure Assessment) score to describe organ
combination of vitamin C and low-dose glucocorticoids may dysfunction/failure. On behalf of the Working Group on
simultaneously and synergistically blunt both the excessive Sepsis-Related Problems of the European Society of Intensive
proinflammatory and immunosuppression response charac- Care Medicine. Intensive Care Med 1996;22:707–710.
teristic of sepsis and thereby prevent PICS syndrome. 15. Mira JC, Gentile LF, Mathias BJ, et al. Sepsis pathophysi-
ology, chronic critical illness, and persistent inflammation-
Conclusions immunosuppression and catabolism syndrome. Crit Care
The major pathogenetic mechanisms whereby patients with Med 2017;45:253–262.
sepsis die include refractory vasodilatory shock, ARDS, 16. Torgersen C, Moser P, Luckner G, et al. Macroscopic
MSOF, and PICS syndrome. Because of their overlapping and postmortem findings in 235 surgical intensive care pa-
tients with sepsis. Anesth Analg 2009;108:1841–1847.
synergistic immunomodulating effects, HAT therapy may re-
17. Frencken JF, van Vught LA, Peelen LM, et al. An un-
store the dysregulated immune system and thereby prevent or
balanced inflammatory cytokine response is not associated
limit vasoplegic shock, ARDS, and the bioenergetic failure that with mortality following sepsis: A prospective cohort
characterizes sepsis and reduce the risk of death. study. Crit Care Med 2017;45:e493–e499.
18. Weiss SL, Balamuth F, Hensley J, et al. The epidemiology
Author Disclosure Statement of hospital death following severe sepsis: When, why, and
The author has no real or potential conflicts of interest how children with sepsis die. Pediatr Crit Care Med 2017;
concerning this manuscript. 18:823–830.
19. Calvano SE, Xiao W, Richards DR, et al. A network-
References
based analysis of systemic inflammation in humans. Nat-
ure 2005;437:1032–1037.
1. Fleischmann C, Scherag A, Adhikari NK, et al. Assess- 20. Tang BM, Huang SJ, McLean AS. Genome-wide tran-
ment of global incidence and mortality of hospital-treated scription profiling of human sepsis: A systematic review.
sepsis. Current estimates and limitations. Am J Respir Crit Crit Care 2010;14:R237.
Care Med 2016;193:259–272. 21. Carcamo JM, Pedraza A, Borquez-Ojeda O, et al. Vitamin
2. Gaieski DF, Edwards JM, Kallan MJ, et al. Benchmarking C suppresses TNF-alpha induced NFkB activation by in-
the incidence and mortality of severe sepsis in the United hibiting IkB-alpha phosphorylation. Biochem 2002;41:
States. Crit Care Med 2013;41:1167–1174. 12995–3002.
HAT THERAPY AND SEPSIS 7

22. Kim SR, Kim YM, Park EJ, et al. Ascorbic acid reduces 40. Beutler B, Greenwald D, Hulmes JD, et al. Identity of
HMGB1 secretion in lipopolysaccharide-activated RAW tumour necrosis factor and the macrophage-secreted factor
264.7 cells and improves survival rate in septic mice by cachectin. Nature 1985;316:552–554.
activation of Nrf2/HO-1 signals. Biochem Pharmacol 41. Fischer E, Marano MA, van Zee KJ, et al. Interleukin-1
2015;95:279–289. receptor blockade improves survival and hemodynamic
23. Hattori M, Yamazaki M, Ohashi W, et al. Critical role of performance in Escherichia coli septic shock, but fails to
endogenous histamine in promoting end-organ tissue in- alter host responses to sublethal endotoxemia. J Clin In-
jury in sepsis. Intensive Care Med Exp 2016;4:36. vest 1992;89:1551–1557.
24. Hagel AF, Layritz CM, Hagel WH, et al. Intravenous 42. Sanfilippo F, Corredor C, Fletcher N, et al. Diastolic dys-
infusion of ascorbic acid decreases serum histamine con- function and mortality in septic patients: A systematic review
centrations in patients with allergic and non-allergic dis- and meta-analysis. Intensive Care Med 2015;41:1004–1013.
eases. Naunyn-Schmiedebergs Arch Pharmacol 2013;386: 43. Carr AC, Shaw G, Fowler AA, et al. Ascorbate-dependent
789–793. vasopressor synthesis—A rationale for vitamin C admin-
25. May JM, Harrison FE. Role of vitamin C in the function istration in severe sepsis and septic shock? Crit Care 2015;
of the vascular endothelium. Antioxid Redox Signal 2013; 19:418.
19:2068–2083. 44. Armour J. Ascorbate prevents microvascular dysfunction
26. Oudemans-van Straaten HM, Spoelstra-de Man AM, de in the skeletal muscle of the septic rat. J Appl Physiol
Waard MC. Vitamin C revisited. Crit Care 2014;18:460. 2001;90:795–803.
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

27. Beltramo E, Berrone E, Buttiglieri S, et al. Thiamine and 45. Wu F, Wilson JX, Tyml K. Ascorbate inhibits iNOS ex-
benfotiamine prevent increased apoptosis in endothelial pression and preserves vasoconstrictor responsiveness in
cells and pericytes cultured in high glucose. Diabetes skeletal muscle of septic mice. Am J Physiol Regul Integr
Metab Res Rev 2004;20:330–336. Comp Physiol 2003;285:R50–R56.
28. de Andrade JA, Gayer CR, Nogueira NP, et al. The effect 46. Wu F, Wilosn JX, Tymi K. Ascorbate protects against
of thiamine deficiency on inflammation, oxidative stress impaired arteriolar constriction in sepsis by inhibiting
and cellular migration in an experimental model of sepsis. inducible nitric oxide synthase expression. Free Radic
J Inflamm 2014;11:11. Biol Med 2004;37:1282–1289.
29. Barabutis N, Khangoora V, Marik PE, et al. Hydro- 47. Annane D, Bellisant E, Sebille V, et al. Impaired pressor
cortisone and ascorbic acid synergistically protect and sensitivity to noradrenaline in septic shock patients with
repair lipopolysaccharide-induced pulmonary endothelial and without impaired adrenal function reserve. Br J Clin
barrier dysfunction. Chest 2017;152:954–962. Pharmacol 1998;46:589–597.
30. Azari O, Kheirandish R, Azizi S, et al. Protective effects 48. Chappell D, Jacob M, Hofmann-Kiefer K, et al. Hydro-
of hydrocortisone, vitamin C and E alone or in combi- cortisone preserves the vascular barrier by protecting the en-
nation against renal-ischemia-reperfusion injury rat. Iran J dothelial glycocalyx. Anesthesiology 2007;107:776–784.
Pathol 2015;10:272–280. 49. Marik PE, Pastores SM, Annane D, et al. Recommenda-
31. Tavasoli M, Azari O, Kheirandish R. Evaluation of tions for the diagnosis and management of corticosteroid
combination therapy with hydrocortisone, vitamin C and insufficiency in critically ill adult patients: Consensus
vitamin E in a rat model of intestine ischemia-reperfusion statements from an international task force by the Amer-
injury. Comp Clin Pathol 2018;27:439–443. ican College of Critical Care Medicine. Crit Care Med
32. Kumar A, Roberts D, Wood KE, et al. Duration of hy- 2008;36:1937–1949.
potension before initiation of effective antimicrobial 50. Wilson JX. Evaluation of vitamin C for adjuvant sepsis
therapy is the critical determinant of survival in human therapy. Antioxid Redox Signal 2013;19:2129–2140.
septic shock. Crit Care Med 2006;34:1589–1596. 51. Marik PE. ‘‘Vitamin S’’ (steroids) and vitamin C for the
33. Boyer A, Vargas F, Coste F, et al. Influence of surgical treatment of severe sepsis and septic shock! Crit Care Med
treatment timing on mortality from necrotizing soft tissue 2016;44:1228–1229.
infections requiring intensive care management. Intensive 52. Marik PE, Khangoora V, Rivera R, et al. Hydrocortisone,
Care Med 2009;35:847–853. vitamin C and thiamine for the treatment of severe sepsis
34. Marik P, Bellomo R. A rational apprach to fluid therapy in and septic shock: A retrospective before-after study. Chest
sepsis. Br J Anaesth 2016;116:339–349. 2017;151:1229–1238.
35. Marik PE, Linde-Zwirble WT, Bittner EA, et al. Fluid 53. ARDS Definition Task Force; Ranieri VM, Rubenfeld
administration in severe sepsis and septic shock, patterns GD, et al. Acute respiratory distress syndrome: The Berlin
and outcomes. An analysis of a large national database. definition. JAMA 2012;307:2526–33.
Intensive Care Med 2017;43:625–632. 54. Bellani G, Laffey JG, Pham T, et al. Epidemiology, pat-
36. Marik PE. Iatrogenic salt water drowning and the hazards terns of care, and mortality for patients with acute respi-
of a high central venous pressure. Ann Intensive Care ratory distress syndrome in intensive care units in 50
2014;4:21. countries. JAMA 2016;315:788–800.
37. Rittirsch D, Flierl MA, Ward PA. Harmful molecular 55. Marik PE, Long A. ARDS complicating pustular psoria-
mechanisms in sepsis. Nat Rev Immunol 2008;8:776–787. sis: Treatment with low-dose corticosteorids, vitamin C
38. Parrish WR, Gallowitsch-Puerta M, Czura CJ, et al. Ex- and thiamine. BMJ Case Rep 2018. [In press; DOI:
perimental therapeutic strategies for severe sepsis: Med- 10.1136/bcr-2017-223475].
iators and mechanisms. Ann NY Acad Sci 2008;1144: 56. Russell JA. Management of sepsis. N Engl J Med 2006;
210–236. 355:1699–1713.
39. Tracey KJ, Beutler B, Lowry SF, et al. Shock and tissue 57. Hawiger J, Veach RA, Zienkiewicz J. New paradigms in
injury induced by recombinant human cachectin. Science sepsis: From prevention to protection of failing micro-
1986;234:470–474. circulation. J Thromb Haemost 2015;13:1743–1756.
8 MARIK

58. Hotchkiss RS, Rust RS, Dence CS, et al. Evaluation of the 78. Bolanos JP, Heales SJ, Peuchen S, et al. Nitric oxide-
role of cellular hypoxia in sepsis by the hypoxic marker mediated mitochondrial damage: A potential neuropro-
[18F]fluoromisonidazole. Am J Physiol 1991;261:R965– tective role for glutathione. Free Radic Biol Med 1996;21:
R972. 995–1001.
59. Boekstegers P, Weidenhofer S, Pilz G, et al. Peripheral 79. Singer M. Mitochondrial function in sepsis: Acute phase
oxygen availability within skeletal muscle in sepsis and versus multiple organ failure. Crit Care Med 2007;35:
septic shock: Comparison to limited infection and car- S441–S448.
diogenic shock. Infection 1991;19:317–323. 80. Garrabou G, Moren C, Lopez S, et al. The effects of sepsis
60. Tran M, Tam D, Bardia A, et al. PGC-1alpha promotes on mitochondria. J Infect Dis 2012;205:392–400.
recovery after acute kidney injury during systemic in- 81. Langley RJ, Tsalik EL, van Velkinburgh JC, et al. An
flammation in mice. J Clin Invest 2011;121:4003–4014. integrated clinico-metabolomic model improves predic-
61. Arulkumaran N, Pollen S, Greco E, et al. Renal tubular tion of death in sepsis. Sci Transl Med 2013;5:195ra195.
cell mitochondrial dysfunction occurs despite preserved 82. Langley RJ, Tipper JL, Bruse S, et al. Integrative ‘‘omic’’
renal oxygen delivery in experimental septic acute kidney analysis of experimental bacteremia identifies a metabolic
injury. Crit Care Med 2018;46:e318–e325. signature that distinguishes human sepsis from systemic
62. Marik PE, Sibbald WJ. Effect of stored-blood transfusion inflammatory response syndromes. Am J Respir Crit Care
on oxygen delivery in patients with sepsis. JAMA 1993; Med 2014;190:445–455.
269:3024–3029. 83. Zhang X. Calcium/calmodulin-dependent protein kinase
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

63. Marik PE, Bellomo R. Lactate clearance as a target of regulates the PINK1/Parkin and DJ-1 pathways of mito-
therapy in sepsis: A flawed paradigm. OA Critical Care phagy during sepsis. FASEB J 2017;31:4382–4395.
2013;1:3. 84. Parikh SM, Yang Y, He L, et al. Mitochondrial function
64. Garcia-Alvarez M, Marik PE, Bellomo R. Stress hy- and disturbances in the septic kidney. Semin Nephrol
perlactemia. Lancet Endo Diabetes 2014;2:339–347. 2015;35:108–119.
65. Hayes MA, Timmins AC, Yau E, et al. Elevation of 85. Matkovich SJ, Al Khiami BA, Efimov IR, et al. Wide-
systemic oxygen delivery in the treatment of critically ill spread down-regulation of cardiac mitochondrial and
patients. N Engl J Med 1994;330:1717–1722. sarcomeric genes in patients with sepsis. Crit Care Med
66. Gattinoni L, Brazzi L, Pelosi P, et al. A trial of goal- 2017;45:407–414.
oriented hemodynamic therapy in critically ill patients. N 86. Singer M. The role of mitochondrial dysfunction in sepsis-
Engl J Med 1995;333:1025–1032. induced multi-organ failure. Virulence 2014;5:66–72.
67. Early, Goal-Directed Therapy for Septic shock—A Patient- 87. Schafer ST. Mitochondrial DNA: An endogenous trigger
Level Meta-Analysis. N Engl J Med 2017;376:2223–2234. for immune paralysis. Anesthesiol 2016;124:923–933.
68. Hotchkiss RS, Swanson PE, Freeman BD, et al. Apoptotic 88. Fowler AA, Syed AA, Knowlson S, et al. Phase 1 safety
cell death in patients with sepsis, shock and multiple organ trial of intravenous ascorbic acid in patients with severe
dysfunction. Crit Care Med 1999;27:1230–1251. sepsis. J Transl Med 2014;12:32.
69. Takasu O, Gaut JP, Watanabe E, et al. Mechanisms of 89. De Grooth HM, Spoeistra-de Man AM, Oudermans-van
cardiac and renal dysfunction in patients dying of sepsis. Straaten HM. Early plasma vitamin C concentration, or-
Am J Respir Crit Care Med 2013;187:509–517. gan dysfunction and ICU mortality [Abstract]. Intensive
70. Pagliarini DJ, Calvo SE, Chang B, et al. A mitochondrial Care Med 2014;40:S199.
protein compendium elucidates complex I disease bi- 90. Borrelli E, Roux-Lombard P, Grau GE, et al. Plasma
ology. Cell 2008;134:112–123. concentrations of cytokines, their soluble receptors, and
71. Ishikawa K, May CN, Gobe G, et al. Pathophysiology of antioxidant vitamins can predict the development of
septic acute kidney injury: A different view of tubular multiple organ failure in patients at risk. Crit Care Med
injury. Contrib Nephrol 2010;165:18–27. 1996;24:392–397.
72. Graetz TJ, Hotchkiss RS. Preventing organ failure in sepsis: 91. Carr AC, Rosengrave PC, Bayer S, et al. Hypovitaminosis
The search continues. Nat Rev Nephrol 2017;13:5–6. C and vitamin C deficiency in critically ill patients despite
73. Brealey D, Brand M, Hargreaves I, et al. Association recommended enteral and parenteral intakes. Crit Care
between mitochondrial dysfunction and severity and out- 2017;21:300.
come of septic shock. Lancet 2002;360:219–223. 92. Fisher BJ, Seropian IM, Masanori Y, et al. Ascorbic acid
74. Watts JA, Kline JA, Thornton LR, et al. Metabolic dys- attenuates lipopolysaccharide-induced acute lung injury.
function and depletion of mitochondria in hearts of septic Crit Care Med 2011;39:1454–1460.
rats. J Mol Cell Cardiol 2004;36:141–150. 93. Fisher BJ, Kraskauskas D, Martin EJ, et al. Mechanisms
75. Gunst J, Derese I, Aertgeerts A, et al. Insufficient autop- of attenuation of abdominal sepsis induced acute lung
hagy contributes to mitochondrial dysfunction, organ injury by ascorbic acid. Am J Physiol Lung Cell Mol
failure, and adverse outcome in an animal model of crit- Physiol 2012;303:L20–L32.
ical illness. Crit Care Med 2013;41:177–189. 94. Fisher BJ, Kraskauskas D, Martin EJ, et al. Attenuation of
76. Azevedo LC. Mitochondrial dysfunction during sepsis. sepsis-induced organ injury in mice by vitamin C. JPEN
Endocr Metab Immune Disord Drug Targets 2010;10: 2014;38:825–839.
214–23. 95. Sagun KC, Carcamo JM, Golde DN. Vitamin C enters
77. Clementi E, Brown GC, Feelisch M, et al. Persistent in- mitochondria via facilitative glucose transporter 1 (Glut1)
hibition of cell respiration by nitric oxide: Crucial role of and confers mitochondrial protection against oxidative
S-nitrosylation of mitochondrial complex I and protective injury. FASEB J 2005;19:1657–1667.
action of glutathione. Proc Natl Acad Sci USA 1998;95: 96. Lowes DA, Webster NR, Galley HF. Dehydroascorbic
7631–7636. acid as pre-conditioner: Protection from lipopolysaccha-
HAT THERAPY AND SEPSIS 9

ride induced mitochondrial damage. Free Radical Re- tion of glyceraldehyde-3-phosphate dehydrogenase and
search 2010;44:283–292. ATP generation in human leukocytes as a possible
97. Dhar-Mascareno M, Carcamo JM, Golde DW. Hypoxia- mechanism of ascorbate-mediated immunostimulation.
reoxygenation-induced mitochondrial damage and apo- Ann NY Acad Sci 1990;587:34–48.
tosis in human endothelial cells inhibited by vitamin C. 115. Manzella JP, Roberts NJ. Human macrophage and lym-
Free Radic Biol Med 2005;38:1311–1322. phocyte responses to mitogen stimulation after exposure
98. Szakmany T. N-acetylcysteine for sepsis and systemic to influenza virus, ascorbic acid, and hyperthermia. J
inflammatory response in adults. Cochrane Database Syst Immunol 1979;123:1940–1944.
Rev 2012;9:CD006616. 116. Siegel BV. Enhancement of interferon production by
99. Molnar Z. N-acetylcysteine as the magic bullet: Too good poly(rI)-poly(rC) in mouse cell cultures by ascorbic acid.
to be true. Crit Care Med 2008;36:645–646. Nature 1975;254:531–532.
100. Cruickshank AM, Telfer AB, Shenkin A. Thiamine defi- 117. Jeong YJ, Kim JH, Hong JM, et al. Vitamin C treatment of
ciency in the critically ill. Intensive Care Med 1988;14: mouse bone marrow-derived dendritic cells enhanced
384–387. CD8(+) memory T cell production capacity of these cells
101. Corcoran T, O’Neil M, Webb SA. Prevalence of vitamin in vivo. Immunobiology 2014;219:554–564.
deficiencies on admission: relationship to hospital mor- 118. Mohammed BM, Fisher BJ, Kraskauskas D, et al. Vitamin
tality in critically ill patients. Anaesthesia & Intensive C: a novel regulator of neutrophil extracellular trap for-
Care 2009;37:254–260. mation. Nutrients 2013;5:3131–3151.
Downloaded by Tufts University package NERL from www.liebertpub.com at 07/25/18. For personal use only.

102. Holmberg MJ, Moskowitz A, Patel PV, et al. Thiamine in 119. Huijskens MJ, Walczak M, Sarkar S, et al. Ascorbic acid
septic shock patients with alcohol use disorders: An ob- propmotes proliferation of natural killer cell populations
servational pilot study. J Crit Care 2018;43:61–64. in culture systems applicable for natural killer cell ther-
103. Donnino MW, Carney E, Cocchi MN, et al. Thiamine apy. Cytotherapy 2015;17:613–620.
deficiency in critically ill patients with sepsis. J Crit Care 120. Manning J, Mitchell B, Appadurai DA, et al. Vitamin C
2010;25:567–581. promotes maturation of T-cells. Antioxid Redox Signal
104. Costa NA, Gut AL, de Souza Dorna M, et al. Serum thi- 2013;19:2054–2067.
amine concentration and oxidative stress as predictors of 121. Gao YL, Lu B, Zhai JH, et al. The parenteral vitamin C
mortality in patients with septic shock. J Crit Care 2014; improves sepsis and sepsis-induced multiple organ dys-
29:249–252. function syndrome via preventing cellular immunosup-
105. Donnino MW, Andersen LW, Chase M, et al. Rando- pression. Mediators Inflamm 2017;2017:4024672.
mized, double-blind, placebo-controlled trial of thiamine 122. Keh D, Boehnke T, Weber-Cartens S, et al. Immunologic
as a metabolic resuscitator in septic shock: A pilot study. and hemodynamic effects of ‘‘low-dose’’ hydrocortisone
Crit Care Med 2016;44:360–367. in septic shock: A double-blind, randomized, placebo-
106. Moskowitz A, Anderson LW, Cocchi MN, et al. Thiamine controlled, crossover study. Am J Respir Crit Care Med
as a renal protective agent in septic shock: A secondary 2003;167:512–520.
analysis of a randomized, double-blind, placebo- 123. Roquilly A, Mahe PJ, Seguin P, et al. Hydrocortisone
controlled trial. Ann Am Thorac Soc 2017;14:737–741. therapy for corticosteroid insufficiency related to trauma.
107. Nathens AB, Neff MJ, Jurkovich GJ, et al. Randomized, The HYPOLYT study. JAMA 2011;305:1201–1209.
prospective trial of antioxidant supplementation in criti- 124. Venkatesh B, Finfer S, Cohen J, et al. Adjunctive gluco-
cally ill surgical patients. Ann Surg 2002;236:814–822. corticoid therapy in patients with septic shock. N Engl J
108. Gentile LF, Cuenca AG, Effron PA, et al. Persistent in- Med 2018;378:797–808.
flammation and immunosuppression: A common syn- 125. Annane D, Renault A, Brub-Buisson C, et al. Hydro-
drome and new horizon for surgical intensive care. J cortisone plus fludrocortisone for adults with septic shock.
Trauma Acute Care Surg 2012;72:1491–1501. N Engl J Med 2018;378:809–818.
109. Klenner FR. The treatment of poliomyelitis and other 126. Keh D, Trips E, Marx G, et al. Effect of hydrocortisone on
virus diseases with vitamin C. South Med Surg 1949;111: development of shock among patients with severe sepsis.
209–214. The HYPRESS Randomized Clinical Trial. JAMA 2016;
110. Pauling L. Ascorbic acid and the common cold. Am J Clin 316:1755–1785.
Nutr 1971;24:1294.
111. Hemila H, Chalker E. Vitamin C for preventing and
treating the common cold. Cochrane Database Syst Rev
2013;1:CD000980. Address for correspondence:
112. Furuya A, Uozaki M, Yamasaki H, et al. Antiviral effects Dr. Paul E. Marik
of ascorbic and dehydroascorbic acids in vitro. Int J Mol Division of Pulmonary and Critical Care Medicine
Med 2008;22:541–45. Eastern Virginia Medical School
113. Madhusudana SN. In vitro inactivation of the rabies virus 825 Fairfax Avenue, Suite 410
by ascorbic acid. Int J Infect Dis 2004;8:21–25. Norfolk, VA 23517
114. Anderson R. Vitamin C and cellular immune functions.
Protection against hypochlorous acid-mediated inactiva- E-mail: marikpe@evms.edu

You might also like