You are on page 1of 14

662996

research-article2016
NCPXXX10.1177/0884533616662996Nutrition in Clinical PracticeAnez-Bustillos et al

Invited Review
Nutrition in Clinical Practice
Volume 31 Number 5
Intravenous Fat Emulsion Formulations for the Adult and October 2016 596609
2016 American Society
Pediatric Patient: Understanding the Differences for Parenteral and Enteral Nutrition
DOI: 10.1177/0884533616662996
ncp.sagepub.com
hosted at
online.sagepub.com
Lorenzo Anez-Bustillos, MD1; Duy T. Dao, MD1; Meredith A. Baker, MD1;
Gillian L. Fell, MD, PhD1; Mark Puder, MD, PhD1; and
Kathleen M. Gura, PharmD2

Abstract
Intravenous fat emulsions (IVFEs) provide essential fatty acids (EFAs) and are a dense source of energy in parenteral nutrition (PN).
Parenterally administered lipid was introduced in the 17th century but plagued with side effects. The formulation of IVFEs later on made
it a relatively safe component for administration to patients. Many ingredients are common to all IVFEs, yet the oil source(s) and its (their)
percentage(s) makes them different from each other. The oil used dictates how IVFEs are metabolized and cleared from the body. The
fatty acids (FAs) present in each type of oil provide unique beneficial and detrimental properties. This review provides an overview of
IVFEs and discusses factors that would help clinicians choose the optimal product for their patients. (Nutr Clin Pract. 2016;31:596-609)

Keywords
lipids; intravenous fat emulsions; parenteral nutrition; fatty acids

Elucidating the characteristics of each oil source over time has investigators attempted to compound such an emulsion by
resulted in an evolution of the different formulations currently using castor oil as the main ingredient. Numerous attempts
available. Emulsions have gone from being solely made with were made between 1920 and 1960 to create a safe emulsion
soybean oil (SO) to being combined with medium-chain tri- using a variety of oils and surfactants. A cottonseed oilbased
glycerides (MCT; ie, coconut oil), olive oil (OO), and, more emulsion (ie, Lipomul; 15% cottonseed oil, 4% soy phospho-
recently, fish oil (FO). Unfortunately, the lipid, among other lipids, 0.3% poloxamer 188) became the first IVFE approved
constituents in parenteral nutrition (PN) formulations, has been in the United States in 1957.2 This was later withdrawn from
associated with the development of liver disease. Lipid-sparing the market due to severe adverse reactions, including fat over-
or lipid reduction strategies have therefore been proposed to load syndrome.24 A soybean oilbased fat emulsion (SOFE)
avoid these complications. has been the predominant IVLE available to American practi-
The ideal intravenous fat emulsion (IVFE) would reverse or tioners since its introduction in Europe in 1961 and its
prevent essential fatty acid deficiency (EFAD) without leading subsequent approval in the United States in 1972. Other oil
to complications, while simultaneously providing energy to sources have been used outside the United States to create
facilitate normal growth and development. Modifications in
their ingredients, formulation, and dosing have made IVFEs a From the 1Vascular Biology Program and the Department of Surgery. Boston
relatively safe component alone or when added to PN formula- Childrens Hospital, Boston, Massachusetts, USA; and the 2Department of
tions. The ideal emulsion, however, has yet to be developed. Pharmacy, Boston Childrens Hospital, Boston, Massachusetts, USA.
Financial disclosure: Research funding is provided by the Boston
Childrens Hospital Surgical Foundation, Boston Childrens Hospital, the
Overview of Fat Emulsions Corkin and Maher Family Fund, National Institutes of Health (NIH) grant
5T32HL007734-22 (MAB), and NIH grant 1F32DK104525-01 (GLF).
IVFEs are a source of essential fatty acids (EFAs) and serve as
Conflicts of interest: A license agreement for the use of Omegaven has
a complement to carbohydrates by providing a dense source of been signed by Boston Childrens Hospital and Fresenius Kabi, and a
nonprotein energy in PN. Fatty acids (FAs) are important patent has been submitted by Boston Childrens Hospital on behalf of Mark
sources of energy and structural components of cell mem- Puder and Kathleen M. Gura, who also serve on the Scientific Advisory
branes. They are also precursors to key modulators involved in Boards for Pronova-BASF and Sancilio and Company. Kathleen M. Gura
also serves on the Pharmaceutical Advisory Board for B. Braun USA.
cellular pathways of the immune response.1 Parenterally
administered fat was first attempted in the 17th century when This article originally appeared online on August 16, 2016.
the English naturalist William Courten tried to infuse OO intra-
Corresponding Author:
venously in dogs, which resulted in pulmonary emboli.2 Over Kathleen M. Gura, PharmD, Department of Pharmacy, Boston Childrens
time, it was recognized that fat could be given intravenously Hospital, 300 Longwood Avenue, Boston, MA 02115 USA.
only in the form of an emulsion. In the 1920s, Japanese Email: Kathleen.Gura@childrens.harvard.edu

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 597

developed the first safe IVFE for clinical use by using egg yolk
Common abbreviations (in alphabetical order) phospholipid as an emulsifying agent.2 All currently available
ALA: -linolenic acid IVFEs contain this emulsifying agent. Individuals with an egg
ARA: arachidonic acid allergy may be unable to receive them, although the egg leci-
DHA: docosahexaenoic acid thin currently used to manufacture IVFEs is highly purified
EFA: essential fatty acid and unlikely to contain allergens thought to trigger hypersensi-
EFAD: essential fatty acid deficiency tivity reactions. Given that such reactions have been described
EN: enteral nutrition in the literature and noted in the package, this potential risk
EPA: eicosapentaenoic acid should be taken into consideration when prescribing IVFEs to
FA: fatty acid patients with egg allergy.6 IVFEs also contain sterile water for
FO: fish oil injection, sodium hydroxide to maintain a pH range between 6
FOFE: fish oilbased fat emulsion and 9, and glycerol, which makes the emulsion isotonic and
HDL: high-density lipoprotein adds additional calories.
IFALD: intestinal failureassociated liver disease Manufacturing IVFEs is difficult and requires very specific
IVFE: intravenous fat emulsion pH and temperatures under a nitrogen rich-environment.
LA: linoleic acid Pharmacopeial specifications of IVFEs help determine embolic
LCT: long-chain triglyceride risks, plasma clearance, and beyond-use dating of the formula-
LDL: low-density lipoprotein tion. The United States Pharmacopeia (USP) chapter <729> has
LPL: lipoprotein lipase established value standards for globule size limits, including a
MA: Mead acid
mean droplet diameter below 500 nm and a large globule con-
MCT: medium-chain triglyceride
tent (percentage of fat globules >5 m) no greater than 0.05%
MUFA: monounsaturated fatty acid
(PFAT5).7,8 Other specifications include a pH between 6 and 9
O3FA: -3 fatty acid
and a free FA content no greater than 0.07 mEq/g.7,8 Currently,
O6FA: -6 fatty acid
all commercially available IVFEs meet the USP standards.
O9FA: -9 fatty acid
OO: olive oil
PN: parenteral nutrition FAs: An Overview
PUFA: polyunsaturated fatty acid
RES: reticuloendothelial system IVFEs provide FAs, which are normally present as cholesterol
SFA: saturated fatty acid esters in circulating lipoproteins, as phospholipids in cell
SO: soybean oil membranes, and as triglycerides in adipose tissue, blood, liver,
SOFE: soybean oilbased fat emulsion and muscle. FAs are the monomers of lipids and consist of
TNA: total nutrient admixture hydrocarbon chains that vary in length. They can be short (24
VLDL: very low-density lipoprotein carbons), medium (612 carbons), long (1421 carbons), or
very long (22 carbons) chains. Depending on the number of
double bonds, they can also be classified as saturated (no dou-
IVFEs and may soon be available. This review aims to provide ble bonds), monounsaturated (1 double bond), or polyunsatu-
an overview of lipid emulsions and discuss factors that clini- rated (>1 double bond). Saturated fatty acids (SFAs) are often
cians should consider when choosing the optimal product for solid at room temperature and structurally have a straight car-
their patients. bon chain. A classic example of this type of FA is butyric acid.
Monounsaturated FAs (MUFAs) are often liquid at room tem-
perature and solid when cool. Palmitoleic and oleic acids are
What Is Common to All IVFEs?
examples of MUFAs abundant in OO. Polyunsaturated FAs
All IVFEs share many components. IVFEs are suspensions of (PUFAs) are liquid at room temperature and include -linolenic
oil in an aqueous medium manufactured to possess properties acid (ALA) and linoleic acid (LA). Last, highly unsaturated
similar to natural chylomicrons. These are the spherical forms FAs have >3 double bonds. Examples include the very long-
of approximately 200500 nm in diameter that the human body chain FAs docosahexaenoic and eicosapentaenoic acids (DHA
uses to transport hydrophobic fat in hydrophilic blood without and EPA, respectively).
causing embolic phenomena.5
Emulsions are unstable systems and undergo physical
changes over time (eg, aggregation, creaming, and increased
EFAs
droplet size). Manufacturing an emulsion requires an emulsify- One of the original indications for the use of IVFEs in patients
ing agent to disperse the oil phase into the aqueous phase, requiring PN was the provision of EFAs. These are FAs that can-
resulting in a stable product. Numerous agents had been tried not be synthesized in the body but rather need to be obtained
to serve as emulsifiers without success. Arvid Wretlind from the diet.9,10 In contrast, FAs from the -5, -7, and -9

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
598 Nutrition in Clinical Practice 31(5)

Table 1. Attributes of Widely Available Intravenous Fat Emulsions.

Component Intralipid Omegaven ClinoLipid/ClinOleic SMOFlipid Lipidem/Lipoplus


Soybean oil, % 100 20 30 40
Medium-chain triglycerides, % 30 50
Olive oil, % 80 25
Fish oil, % 100 15 10
Glycerol, g/100 mL 2.25 2.5 2.25 2.5 2.5
Egg phospholipid, g/100 mL 1.2 1.2 1.2 1.2 1.2
Phytosterols, mg/L 439 5.7 3.66 274 2.6 207 NA125
Vitamin E, mg/100 mL 3.8 1530 3.2 1623 24.2
LA, % 50 4.4 18.5 21.4 24.5
ALA, % 9 1.8 2 2.5 3.5
EPA, % 0 19.2 0 3 3.5
DHA, % 0 12.1 0 2 2.5
ARA, % 0 14 0 0.150.6 083

ALA, -linolenic acid; ARA, arachidonic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; LA, linoleic acid; NA, not available.

families are nonessential. EFAs serve as an energy source, pro- has high levels of -tocopherol but low amounts of -tocopherol
vide structural support to cell membranes, and are precursors to (bioactive form of vitamin E).11 Evidence from ex vivo, ani-
important cellular metabolites. The -3 FAs (O3FAs) and -6 mal, and clinical studies suggests that the type of IVFE used
FAs (O6FAs) are the 2 families of EFAs. They both compete for may influence immune functions. For instance, emulsions with
space within the cell membrane and are processed by the same a high content of O6FAs have been linked with immunosup-
enzymes (ie, elongases and desaturases) to generate their more pressive effects.1214 One study evaluated the effect of lipid
active downstream products. FAs from these families differ struc- intake on the postoperative stress response and cell-mediated
turally depending on where the first double bond from the termi- immune function of patients subjected to gastric or colorectal
nal methyl group is located. The first double bond thus occurs at surgery.15 Higher postoperative concentrations of interleukin-6
the third carbon in O3FAs and at the sixth carbon in O6FAs. and C-reactive protein were seen in patients receiving a SOFE
Historically, ALA and LA have been considered the main mem- (20% of the energy intake in extremely high nonprotein calorie
bers of the O3FA and O6FA families, respectively. Recently, the intake of 40 kcal/kg/d) compared with those receiving lipid-
true essentiality of ALA and LA has been questioned. Provision free PN. This is the reason why many centers do not administer
of their main downstream products (ie, DHA and arachidonic 100% SOFE to critically ill patients, which is discussed in
acid [ARA], respectively) is just as effective at preventing the greater detail below.
development of biochemical EFAD.10 The phytosterols present in SO are plant sterols thought to
contribute to the development of intestinal failureassociated
What Is Different Between IVFEs? liver disease (IFALD).16 Phytosterols, including campesterol,
sitosterol, and stigmasterol, are typically absorbed only in small
Despite sharing several common properties, the oil source(s) amounts in the gastrointestinal (GI) tract.17 These plant sterols
used and its (their) percentage(s) dictate the key differences decrease blood cholesterol levels by interfering with their GI
between IVFEs. These differences account for their addi- absorption. This makes them beneficial agents when taken orally
tional benefits or detrimental effects, especially when used in patients with hypercholesterolemia and atherosclerosis.18,19
for prolonged periods (Table 1). Typically, IVFEs are manu- However, phytosterols accumulate in the liver when given intra-
factured with 1 of 5 types of oil: soybean, safflower, coco- venously, where they inhibit the enzyme 7-hydroxylase, the
nut, olive, or fish. Each has unique inflammatory properties rate-limiting enzyme in bile acid synthesis.16,20,21 Unlike SOFEs,
and may even confer different pharmaceutical and therapeu- FO-based fat emulsions (FOFEs) contain very little phytoster-
tic benefits. ols. This difference may partially explain the potential benefit of
FOFEs as a lipid source in PN-dependent patients with IFALD.
Levels of plasma phytosterols were measured in preterm infants
Soybean Oil receiving different IVFEs in a randomized controlled trial.22
SOFEs have been widely used for decades. SO contains high Phytosterol concentrations were higher in infants receiving
concentrations of PUFAs with a ratio of LA (O6FA) to ALA 100% SOFE. Cholestasis did not develop in this group and is not
(O3FA) of approximately 7:1.11 In addition, 25% of the FAs in surprising given the short duration of the study. The role of phy-
SO come from the nonessential family of -9 FAs (O9FAs) in tosterols in hepatocyte damage has been demonstrated by their
the form of oleic acid. SO is naturally rich in phytosterols and antagonizing effect on the farsenoid X nuclear receptor, which is

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 599

critical in regulating the level of intrahepatic bile acids.23 In (range, 27.644.0 mg/mL), and ALA is 4.7 mg/mL (range,
addition, the incorporation of phytosterols in erythrocyte mem- 1.08.4 mg/mL).31 This product provides 30% of the PUFA
branes accelerates the breakdown of these cells and increases content of standard SOFEs. In comparison to SO, OO is rich in
the bilirubin load to the liver.21 Phytosterols can also increase MUFAs that possess less proinflammatory properties and are
the risk of sepsis by altering the migratory and phagocytic func- more resistant to oxidative stress injuries from free radicals.
tion of neutrophils as shown in animal models.21,24,25 This type of mixed oil emulsion has replaced SOFE as the stan-
dard IVFE in many countries.32 In 1 randomized controlled
trial, an OO-containing IVFE (4:1 OO/SO) was compared with
Safflower Oil SOFE in preterm infants younger than 28 weeks gestational
Safflower oil had been used in IVFEs alone or in combination age.33 OO-containing IVFE was found to be safe and well tol-
with SO in the United States since 1980. It was developed as erated. Long-chain PUFA concentrations were similar in
an alternative to SO and was hypothesized to decrease the infants receiving OO IVFE in comparison to the SOFE group,
risk of fat overload syndrome.26 In comparison to SOFEs, despite the significantly lower amount of PUFAs in the OO/SO
safflower-based IVFE had higher concentrations of LA (77% IVFE. Moreover, no difference in lipid peroxidation was
vs 54%), although less ALA (0.5% vs 8%). The use of saf- observed. Similar results were reported in several other ran-
flower oil predisposed patients to develop O3FA deficiency domized controlled trials involving neonates receiving OO/SO
when used as a sole source of fat in IVFEs.27 For this reason, IVFE, with none showing any significant difference in oxida-
it was later reformulated as a 50/50 blend with SO. Safflower tive stress in comparison to SOFE monotherapy.34,35 In another
oilbased emulsions are not currently available due to raw investigation, triglyceride, cholesterol, and high-density and
material issues. low-density lipoprotein (HDL and LDL, respectively) concen-
trations and liver function tests were similar between the 2
groups. Very low-density lipoprotein (VLDL) concentrations
Coconut Oil (MCTs) were statistically lower in neonates receiving SOFE in com-
After the introduction of SOFEs, a second-generation emul- parison to those receiving the OO blend.36 Another potential
sion was introduced in Europe that contained a 50/50 mixture benefit of using OO-containing IVFE is improved glucose tol-
of SO and MCTs derived from coconut oil.11 These emulsions erance. SO appears to enhance glucose production by means of
reduce the O6FA content by 50%. MCTs are SFAs that are glycogenolysis and gluconeogenesis. Infants receiving
612 carbons long and include caprylic and capric acids.28 OO-containing IVFEs have been demonstrated to tolerate
They are easily metabolized and lack proinflammatory proper- higher carbohydrate intake without developing hyperglycemia.37
ties, both characteristics unique to this fat source. MCTs are Unlike SO and FO, OO is considered immune neutral.38 The
resistant to peroxidation and have protein-sparing effects not high content of O6FAs in SOFEs serves as a precursor to cyto-
inferior to the ones seen with long-chain triglycerides (LCTs). kines and proinflammatory prostaglandin E2 and leukotriene
In addition, MCTs do not accumulate in the liver and conse- B4.38 A study comparing the inflammatory effects between
quently do not impair hepatic function.28,29 However, MCT oils SOFE and OO-rich IVFE in preterm neonates demonstrated
are devoid of EFAs and thus cannot be used as a sole source of that proinflammatory cytokine concentrations were signifi-
fat. They may have a unique pharmaceutical benefit in that cantly higher in those receiving SOFE.39 One potential down-
MCTs added to certain total nutrient admixtures (TNAs) results side for the use of an OO-containing IVFE is the greater risk of
in a more stable lipid emulsion than those containing an abun- extraction compared with other IVFEs of the plasticizer di(2-
dance of LCTs. The shorter chain length (612 carbons vs 18 ethylhexyl)phthalate (DEHP) if it is infused using a polyvinyl-
carbons in SO) exerts less stress on the emulsifying agent and chloride (PVC) administration set. DEHP exposure could
allows the lipid phase to remain miscible with the aqueous potentially increase the risk of developing IFALD.40 For these
phase of the emulsions for longer periods of time.30 reasons, regardless of oil source, when compounding admix-
tures containing IVFE, PVC- or DEHP-coated containers
should not be used.
Olive Oil
OO is rich in O9FAs (ie, oleic acid), a type of MUFA that is not
considered essential, as it is not a precursor of eicosanoids.
Fish Oil
OO-based emulsions were introduced in Europe in the 1990s. Like OO, FO-containing IVFEs are less proinflammatory than
The relatively small amount of LA (approximately 5%) conventional SOFEs.38 The eicosanoids produced from O3FAs
explains why this oil source requires blending with an oil con- in FO are generally less inflammatory in comparison to those
taining EFA. OO has a lower content of phytosterols and abun- originating from O6FAs contained in SO. FO is rich in the anti-
dant -tocopherol. One OO-based IVFE (eg, Clinolipid; Baxter oxidant -tocopherol, which is added to prevent the oxidation
Healthcare Corporation, Deerfield, IL) is composed of 4 parts of its FAs. -Tocopherol is the form of vitamin E that is prefer-
OO/1 part SO. The mean concentration of LA is 35.8 mg/mL entially absorbed and accumulated in humans. In comparison,

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
600 Nutrition in Clinical Practice 31(5)

Figure 1. Evolution of fat sources in intravenous fat emulsions and their corresponding changes in the inflammatory profile. MCT,
medium-chain triglyceride.

SO is rich in -tocopherol, which must be methylated to the 2-prostanoids and series 4-leukotrienes, molecules known to
preferred bioactive -tocopherol. FOFEs were originally be generally more proinflammatory.50 ARA has also been
developed to supplement conventional SOFE and not intended shown to have anti-inflammatory and pro-resolving properties
for use as monotherapy.11 Unlike SOFE, FOFE has little LA by serving as the precursor to lipoxins and prostaglandin-medi-
and ALA but contains their downstream metabolites, ARA, ated lipoxins.49 Thus, the fat source in IVFEs and their result-
EPA, and DHA.11 The paucity of O6FAs has raised concerns ing inflammatory profile has dictated the evolution of the
about the development of EFAD when FOFE is used as mono- different formulations over time. Emulsions have evolved
therapy. However, in humans, 100% FOFE has been shown to from being solely made with SO in the 1960s to being com-
effectively reverse preexisting EFAD and to prevent its devel- bined with MCTs in the 1980s, OO in the 1990s, and, more
opment when dosed at 1 g/kg/d.41,42 Subsequent work in ani- recently, with FO51 (Figure 1).
mal models has confirmed this concept. In studies involving a
murine model, the provision of only the downstream metabo-
lites of LA and ALA (ie, ARA and DHA, respectively) and no Oil Sources: Impact on Clearance
other EFA was shown to prevent EFAD and hepatic steatosis Triglycerides in IVFEs are hydrolyzed by lipoprotein lipase
without negatively affecting growth or fertility.43 O3FAs pres- (LPL). This enzyme is located on the endothelial surface of
ent in FO are also natural ligands to some receptors of the extrahepatic tissues, which end up using the released FAs.52,53
G-protein-coupled receptor (GPR) family. Recent evidence The remaining lipid particles are either hydrolyzed by hepatic
demonstrates that the interaction with these receptors mediates lipases or cleared intact by other tissues.54,55 The oil source in
some of the therapeutic benefits of O3FAs in tissues such as IVFEs strongly determines how the body clears them from
the liver, brain, and bones.4447 the circulation. Brouwer etal56 compared the elimination of
intravenous (IV) OO and SO fat emulsions. Although SOFE
Do Oil Sources Determine the had a higher rate of elimination than OO-containing IVFE,
the hepatic lipase activity was more important in the elimina-
Inflammatory Characteristics of IVFEs? tion of OO. These findings suggested the presence of addi-
Each emulsion has its own inflammatory characteristics based tional clearance pathways beyond the enzymatic ones that
on the different oil source(s) and their predominant FA. The have been mentioned. The reticuloendothelial system (RES)
EPA present in FO is a precursor to the series 5-leukotrienes and other tissues also play a role in the metabolism and clear-
and series 3-prostanoids, molecules with anti-inflammatory ance pathways of IVFEs. The phospholipid concentration
properties. O3FAs also affect eicosanoid metabolism and the also seems to have an effect on clearance of IVFEs. Depending
epoxygenation pathways by decreasing the hepatic levels of on their concentration (ie, 10% and 20%), IVLEs differ in
the enzyme-soluble epoxide hydrolase.48 By doing this, O3FAs their phospholipid/triglyceride ratio (0.12 and 0.06, respec-
decrease the degradation of anti-inflammatory molecules. In tively). A study comparing the effects on plasma lipids and
addition, O3FAs serve as precursors to pro-resolving mole- lipoproteins in 10% and 20% SOFE in low-birth-weight neo-
cules that aid in the resolution of the inflammatory response.48,49 nates demonstrated that 10% emulsions led to higher concen-
In contrast, ARA present in SO leads to the formation of series trations of plasma triglycerides, accumulation of cholesterol

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 601

and phospholipids in LDL, and the appearance of lipoprotein Clinical Considerations


Xlike particles.57 These particles decrease the rate of hydro-
lysis by competing for LPL with other lipoproteins rich in Fat emulsions were initially brought to the market based on their
triglycerides. Altogether, these changes affect the hepatic ability to provide energy and EFAs (ie, a nutrition indication).
clearance of LDL and chylomicron remnants and delay the Studies used to support the approval process were statistically
hydrolysis of circulating triglycerides.57,58 For this reason, underpowered to show meaningful differences in clinical out-
20% SOFE is preferred over 10% SOFE as an IVFE source in comes. As a result, postmarketing studies have described clinical
low-birth-weight infants. Similar differences were seen in a differences in the various commercially available IVFEs. The
study comparing different concentrations of SOFE in criti- addition of IVFEs to TNAs is also a common practice in certain
cally ill adults.59 Patients who received a 30% emulsion had settings. The use of these admixtures is controversial, however,
lower plasma levels of triglycerides, phospholipids, and free given concern for the potentially unstable emulsions, incompat-
cholesterol than those who received the 10% SOFE. The 10% ibility of medications with the IVFEs, microbial growth, occlu-
group also showed an increase in levels of lipoprotein Xlike sion, and a reduced life span of IV catheters used for infusion.72,73
particles. MCT oils bind poorly to serum albumin and are Due to their high requirements for calcium and phosphorus that
cleared more rapidly from the plasma. Several properties in can affect the stability of the admixture, TNAs are not recom-
the metabolism and clearance of MCTs make them poten- mended to be used in neonates and children.73
tially a more beneficial source of fat in septic or critically ill
patients. First, they do not accumulate in tissues, including
Prevention of EFAD
the RES. Accumulation of lipids in macrophages from the
RES has proven to negatively affect their immunologic func- The main beneficiaries of IVFEs are those patients who are not
tion in both human and animal models following administra- able to tolerate or adequately absorb nutrients administered
tion of LCT emulsions.60,61 Second, they undergo rapid through the oral and/or enteral route. IVFEs serve as a dense
oxidation, and their entry into the mitochondria is mediated source of nonprotein energy and, when dosed properly, provide
by a carnitine-independent FA transport (although carnitine the EFAs that avoid the development of EFAD. This condition
may still be required for their oxidation).28,62,63 Unfortunately, occurs when less than 1%2% of the total energy consumed
the blood-brain barrier is permeable to highly soluble MCTs. comes from the EFAs ALA and LA. EFAD is rare in the general
This makes MCTs potentially toxic to the central nervous population and more commonly develops in patients with mal-
system when present in high concentrations.64 Moreover, as absorption syndromes and in those who are PN dependent.74
previously discussed, MCT oils cannot be used as a sole Clinically, EFAD most typically leads to growth retardation,
source of fat in IVFEs as they are devoid of EFAs; they must eczematous dermatitis, and alopecia.7476 As the content of
be administered in combination with an LCT. ARA, a tetraenoic acid, decreases in tissues, the content of non-
In vivo animal studies by Qi etal54 demonstrated that FOFEs essential FAs (ie, Mead acid [MA], a trienoic acid synthesized
undergo faster clearance from the blood than SOFEs. In vitro from oleic acid) increases.77 MA is produced in states of EFAD
studies have shown that -3 triglycerides are relatively resistant and is created from the elongation and desaturation of oleic acid
to the hepatic and lipoprotein lipases, with much lower lipolytic when there is insufficient O6FAs and O3FAs. The Holman
activities than those seen with O6FA-rich emulsions.65 For this Index is used to diagnose EFAD. It comprises the triene (ie,
reason, the faster clearance of FOFEs relative to SOFEs seems MA) to tetraene (ie, ARA) ratio and can be easily calculated.
to be mediated by their tissue uptake rather than by the activity Values >0.2 are indicative of EFAD.77 The biochemical signs of
of the aforementioned lipases. FOFEs also accelerate triglycer- EFAD usually precede those seen clinically and appear in as
ide clearance and inhibit the synthesis of endogenous triglycer- little as 710 days following EFA restriction. The isolated defi-
ides and VLDL. These effects are not mediated by enzymatic ciency of O3FAs and/or O6FAs is even more rare. Animals fed
actions but rather by clearance enhancement.54,66,67 O3FA-deficient diets showed impaired vision and visual evoked
The presence of heparin affects the clearance of IVFEs. potentials, polydipsia, and changes in stereotyped behavior.7880
Heparin stimulates intravascular lipolysis by promoting the Some of these findings have also been shown in humans,
release of LPL and hepatic lipases into the circulation.68 including visual impairment in preterm infants, yet more vari-
Addition of heparin is helpful when clearance of IVFEs is able results in those at term.81 Clinical manifestations in humans
compromised, such as in premature neonates.69,70 are seen especially when O3FAs are not adequately supplied in
The combination of oil sources in the newer IVFEs aims to critical periods of brain and retinal development.81 O3FA defi-
take advantage of the beneficial properties of each of the avail- ciency also leads to similar skin changes seen in patients with
able fat options. For example, the combination of MCTs with EFAD, plus hair loss, impaired growth, and depressive
FO allows for MCTs to release their readily oxidizable FAs symptoms.82 Similarly, O6FA deficiency leads to the aforemen-
while preventing O3FAs from entering oxidative pathways. tioned dermatologic and hair changes in addition to dry eyes,
Bypassing oxidative pathways allows O3FAs to exert their dysrhythmias, and impaired growth.9 Table 2 summarizes the
beneficial effects on different targets.71 differences between O3FA and O6FA deficiencies.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
602 Nutrition in Clinical Practice 31(5)

Table 2. Clinical Differences Most Commonly Seen in -3 and -6 Fatty Acid Deficiency (Data From Animal and Human Studies).

Manifestation -3 Fatty Acids -6 Fatty Acids


Constitutional symptoms Impaired growth. Dermatologic and hair changes Impaired growth. Dermatologic and hair changes
Visual symptoms Impaired vision and visual evoked potentials Dry eyes
Neurologic symptoms Changes in stereotyped behavior, depression None described
Other Polydipsia Dysrhythmias

IVFE as Therapeutic Modalities better outcomes for critically ill, PN-dependent patients.
Manzanares etal90,91 summarized the findings from different
In critically ill adults, fat-containing PN may be given to indi- trials in a recent systematic review and meta-analysis that com-
viduals with prolonged suppression of GI activity following pared different alternatives to SOFE monotherapy in this set-
severe trauma or surgery and in conditions associated with ting (Table 3). There is a trend toward significance of
malabsorption such as prolonged ileus, gastric outlet obstruc- alternatives to SOFE alone being associated with important
tion, diarrhea, and vomiting.83 This is different from circum- reductions in mortality, duration of mechanical ventilation, and
stances that lead to PN dependency in premature infants and intensive care unit (ICU) length of stay. Edmunds etal86
children that include intestinal motility disorders and condi- reviewed data from a prospective international multicenter
tions resulting from major intestinal resections such as necro- study to determine the effects of different IVFEs on clinical
tizing enterocolitis, malrotation, and/or intestinal atresia.84 outcomes in 451 critically ill patients. Inclusion criteria
included adult patients in the ICU for >72 hours, mechanically
Adult considerations.The initiation of IVFE therapy in the ventilated within 48 hours, and receiving PN without prior/
critically ill adult should not be undertaken lightly. The Cana- concurrent EN for 5 or more days. Patients who did not receive
dian Clinical Practice Guidelines recommend withholding the any form of IVFE had the longest duration of mechanical ven-
addition of pure SOFE for the first 10 days of PN therapy in tilation. Those who received either OO-containing or
patients who are otherwise not malnourished, individuals who FO-containing IVFEs were extubated and discharged sooner
are able to tolerate (at least partially) enterally administered from the ICU alive than those receiving SOFE. SOFE mono-
nutrients, or those in whom a short-term course of PN is therapy was associated with a longer ICU stay. In summary,
expected.85 The basis of this recommendation lies in the detri- although SOFE-sparing alternatives are recommended, evi-
mental inflammatory properties of emulsions rich in O6FAs dence is still lacking regarding which optimal blend of oil
and in SO being an important component in most commercially sources is the most suitable in critically ill patients.
available IVFEs.86 Similarly, recent recommendations from the
American Society for Parenteral and Enteral Nutrition (ASPEN)
and the Society of Critical Care Medicine (SCCM)87 suggest Pediatric considerations.In children and preterm infants,
withholding or limiting the use of SOFE during the first week the search for alternatives to SOFE has failed to find an ideal
of PN administration to a maximum of 100 g/wk if there is con- option for its replacement. Results from 15 studies were
cern for EFAD. A meta-analysis of 2 randomized studies evalu- pooled in a recent systematic review from the Cochrane Neo-
ating the early addition of fat emulsions in PN found no natal Group and included the use of MCT/LCT, MCT/OO/
difference in mortality rates but a large reduction in infectious FO, MCT/FO, OO/SO, and borage/SO IVFEs (either in PN or
complications when SOFE was held.85 In this population, there partial PN) in preterm neonates.92 None of the measured pri-
are minimal concerns regarding withholding fat emulsions (ie, mary outcomes, mortality, growth rate, or days to regain birth
hypocaloric nutrition and EFAD) if the patient is tolerating weight, reached statistically significant differences. Second-
some enteral nutrition (EN) and requires a short course of PN ary outcomes (ie, sepsis, IFALD, duration of ventilation,
(<10 days). The availability of alternative IVFEs containing necrotizing enterocolitis [ stage 2], intraventricular hemor-
different oils in addition to SOFE may explain why practice rhage [grades III and IV], periventricular leukomalacia,
guidelines are different in other countries, where such products patent ductus arteriosus, hyperglycemia, and hypertriglyceri-
are considered an integral part of PN for energy and to ensure demia) also failed to show any significant differences.
fatty acid provision.88 Although current Canadian guidelines Neither EFA status nor neurodevelopmental outcomes were
recommend the use of SO-sparing strategies when the addition considered. The guidelines for the management of pediatric
of IVFE to PN is indicated, ASPEN and SCCM refrain from patients with intestinal failure from ASPEN are similar to the
making recommendations about the use of alternatives to SOFE adult recommendations, also suggesting the limited use of
as they are not currently available in the United States.87,89 SOFEs, especially in infants at risk of developing IFALD.93
Given these recommendations, many studies have attempted The use of lipid reduction strategies or finding alternatives to
to determine which combination of oil sources translates into SOFEs is also recommended.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Table 3. Summary of Findings From Randomized Trials Evaluating Strategies and Alternatives to Soybean OilBased Emulsions in Adult, Critically Ill Patients (Data From
Manzanares etal91).
Authors (Year) Inclusion Criteria (n) Oil Sources Compared Measured Outcomes Conclusions
126
Nijveldt etal Patients in the surgical ICU with at LCT (Intralipid; Fresenius Kabi, Metabolic and biochemical differences with No differences in energy expenditure, nitrogen balance,
(1998) least 5 days of ventilator support, Uppsala, Sweden) vs LCT/MCT special focus on liver enzymes LFTs, carnitine, transferrin, prealbumin, serum albumin,
on PN, and septic (n = 20) (50/50, Lipofundin; B. Braun, cholesterol, TGs, and FFAs. Trend toward higher total
Melsungen, Germany) bilirubin levels in the LCT group
Lindgren Patients in ICU anticipated to LCT (Intralipid) vs LCT/MCT Nitrogen balance, energy metabolism, and Better 3-day cumulative nitrogen in the LCT/MCT group.
etal127 (2001) require PN for at least 5 days (n (36/64, Structolipid; Fresenius safety No differences in tolerance, energy expenditure, glucose
= 30) Kabi, Uppsala, Sweden) or TG levels during infusion
Garnacho- Patients in ICU likely to require at LCT (Intralipid) vs LCT/MCT Metabolic and clinical effects No differences in ICU LOS, ICU mortality, or in-hospital
Montero least 10 days of PN (n = 72) (50/50, Lipofundin) mortality. The rise in RBP and recovery of nitrogen
etal128 (2002) balance was greater in those receiving MCT/LCT
Grecu etal129 Patients in ICU with abdominal FO (Omegaven; Fresenius Kabi, Daily serum lipids, CRP levels, reoperation Decreased reoperation rate, ICU and hospital LOS in those
(2003) sepsis (n = 54) Uppsala, Sweden) + LCT vs LCT rate, ICU and hospital LOS, and mortality receiving parenteral FO. No differences in mortality
Huschak Patients in ICU following major LCT/OO (20/80, ClinOleic; Baxter Energy expenditure, energy intake/energy No difference in energy expenditure or energy intake/
etal130 (2005) trauma requiring mechanical SA, Lessines, Belgium) vs LCT/ expenditure ratio, duration of mechanical energy expenditure ratio between groups. OO group
ventilation (n = 33) MCT (50/50, Lipofundin) ventilation, and ICU LOS showed shorter duration of mechanical ventilation
and ICU LOS (of note, lipid/glucose ratio in PN was
different in experimental groups: 75/25 in LCT/OO vs
37/63 in LCT/MCT)
Iovinelli etal131 Patients in ICU with COPD LCT (Intralipid) vs LCT/MCT Nutrition status, metabolic rate, time of No difference in duration of mechanical ventilation, but
(2007) requiring mechanical ventilation (50/50, Lipofundin) ventilatory support and weaning weaning was shorter in those receiving LCT/MCT
(n = 24)
Garca-de- Patients in ICU with severe burns LCT/OO (20/80, ClinOleic) vs Adverse events, clinical outcomes, and No difference in mortality and infection rates between
Lorenzo requiring PN for 57 days (n = 22) LCT/MCT (50/50, Lipofundin) biochemical parameters groups. Patients in LCT/MCT group showed a higher
etal132 (2005) number of liver function abnormalities
Friesecke Patients in ICU expected to require LCT/MCT (50/50, Lipofundin) Changes in IL-6 and HLA-DR expression. No difference seen in all of the measured outcomes
etal133 (2008) PN for at least 6 days, with <25% alone vs LCT/MCT (50/50, Secondary outcomes included nosocomial between groups
calories provided by enteral Lipofundin) + FO (Omegaven) infections, duration of mechanical
nutrition (n = 166) ventilation, ICU LOS, and 28-day mortality
Wang etal134 Patients in ICU with severe acute LCT (Lipovenos; Fresenius Kabi, Changes in IL-10, HLA-DR, CD4+/CD8+ The group treated with FO showed a significant increase
(2009) pancreatitis (n = 56) Uppsala, Sweden) alone vs LCT ratio, infection, and need for surgery in IL-10 and HLA-DR. No differences seen in CD4+/
(Lipoveno) + FO (Omegaven) CD8+ ratio, infection rate, and need for surgery
Barbosa etal135 Patients in medical ICU with SIRS LCT/MCT (5050, NuTRIflex Plasma levels of eicosanoids and cytokines The FO group showed a greater decrease in levels of
(2010) or sepsis (n = 25) Lipid special; B. Braun, and in LPS-stimulated whole-blood culture IL-6 and a smaller decrease in levels of IL-10. There
Melsungen, Germany) vs MCT/ supernatants. Secondary measures included was no difference in days of ventilation, ICU LOS, and
LCT/FO (50/40/10, Lipoplus; B. days of ventilation, ICU LOS, hospital LOS, mortality. Surviving patients had a shorter hospital LOS
Braun, Melsungen, Germany) and mortality in the FO group

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Umpierrez Patients in medical and surgical ICU LCT (Intralipid) vs LCT/OO Clinical outcomes, hospital LOS, glycemic No differences noted in any of the measured outcomes
136
etal (2012) requiring PN for >5 days (n = 100) (20/80, ClinOleic) control, inflammatory and oxidative stress between groups
markers, and granulocyte and monocyte
functions
Pontes-Arruda Patients in ICU requiring PN LCT/OO (20/80, ClinOleic) vs The main objective of the study was No differences in secondary outcomes between groups
etal137 (2012) (n = 406) LCT/MCT (not specified) to compare PN delivery systems
(multichamber bag vs compounded) in
incidence of BSI. Secondary outcomes
included development of sepsis/shock, ICU
LOS, and 28-day mortality

BSI, bloodstream infection; COPD, chronic obstructive pulmonary disease; CRP, C-reactive protein; FFA, free fatty acid; FO, fish oil; HLA-DR, human leukocyte antigenDR; ICU, intensive care unit; IL-6, interleukin-6; IL-10,

603
interleukin-10; LCT, long-chain triglyceride; LFT, liver function test; LOS, length of stay; LPS, lipopolysaccharide; MCT, medium-chain triglyceride; OO, olive oil; PN, parenteral nutrition; RBP, retinol-binding protein; SIRS,
systemic inflammatory response syndrome; TG, triglyceride.
604 Nutrition in Clinical Practice 31(5)

Complications Associated With IVFEs IVFE for 72 hours and reintroducing the IVFE at a lower dose.
On the basis of these findings, the authors recommended that
The IVFE component in PN can cause several metabolic and IVFE doses should not exceed 4 g/kg/d, considerably higher
physiological adverse effects (AEs). Hypertriglyceridemia is than what is currently used in clinical practice. Guidelines from
one of the most common AEs and can predispose patients to the European Society of Parenteral and Enteral Nutrition
elevations in hepatic enzymes, hemolysis, and respiratory dis- (ESPEN) recommend that IVFEs can be safely administered at
tress. Other potential consequences of prolonged administration a rate of 0.71.5 g/kg over 1224 hours.88
of IVFEs include hematologic abnormalities with recurrent FOFE does not appear to exhibit similar complications
thrombocytopenia and hyperactivation of the monocyte-macro- when infused rapidly. In 1 case series, 6 children received
phage system.94 Worsening of respiratory function may be due FOFE at an infusion rate that exceeded 0.17 g/kg/h.105 Infusion
to increased pulmonary vascular constriction.95 Changes in the rates as high as 5 g/kg/h were accidentally administered (range,
alveolar-arteriolar oxygen gradient can lead to decreased gas 0.25 g/kg/h) without evidence of fat overload syndrome. In
exchange and may be due to diminished bioavailability of endo- these patients, vital signs remained stable and none showed
thelial-derived vascular relaxants.96 In 1 report, IVFE was manifestations of respiratory distress, fever, or hemorrhage.
shown to worsen bronchopulmonary dysplasia in premature Transient elevations in serum triglyceride levels were observed
infants.97 The use of IVFE has also been known to cause vaso- but promptly returned to acceptable levels. The authors sug-
constriction, leading to hypertension.98 The rapid infusion of gested that the reason for the apparent absence of fat overload
SOFE may negatively affect pulmonary gas diffusion and alter syndrome in patients receiving rapid infusions of FOFE may
hemodynamic stability in adults with respiratory distress syn- be related to their clearance and also the small number of
drome. Rapid infusion rates (ie, 100 g over 5 hours) were linked patients in this case series. Unlike SOFE, FOFE appears to be
with vasoconstriction, while significant vasodilation was cleared more rapidly from the intravascular space.106 Lipid
observed with slower infusion rates (ie, 100 g over 10 hours).99 clearance follows a biphasic pattern, with an initial rapid clear-
ance occurring within 10 minutes followed by a slower clear-
ance phase of 1025 minutes. Most IVFE clearance from the
Fat Overload Syndrome
blood occurs within the first 2 minutes of an infusion in animal
Fat overload syndrome is a well-known complication of IVFE models. IVFEs containing SO tend to clear more slowly than
therapy. It is characterized by headaches, jaundice, hepato- those containing MCTs or FO.106 Despite being cleared more
splenomegaly, respiratory distress, and spontaneous hemor- efficiently, FOFEs undergo less catabolism than SOFEs. The
rhage.100 It has been described in several case reports in the mechanisms involved in the hydrolysis of FOFEs and SOFEs
presence of rapid infusion and/or high doses of IVFE. AEs are are very different. LPL, apolipoprotein E, LDL receptor, and
due to elevations in plasma triglyceride levels that occur when lactoferrin-sensitive pathways modulate the removal of chylo-
the infusion rate exceeds the rate of hydrolysis. Whenever the micron-sized O6FA-rich IVFEs. In contrast, clearance of chy-
rate of hydrolysis exceeds the rate of uptake and oxidation of lomicron-sized FOFE is independent of these pathways.54 In 1
free FAs, plasma concentrations of FAs increase.101 Other study, FO was shown to accelerate triglyceride clearance by
symptoms seen with fat overload syndrome include anemia, facilitating LPL-mediated lipolysis. DHA and EPA in particu-
leukopenia, thrombocytopenia, low fibrinogen levels, and lar have significantly reduced chylomicron triglyceride half-
depressed levels of coagulation factor V. Oftentimes, these will lives and particle sizes, and increased the activity of preheparin
simply reverse by stopping the IVFE infusion.96 In addition to LPL.107 Taken together, these findings suggest that FO does not
discontinuing the IVFE, supportive care is the mainstay of ther- reduce production of triglycerides but rather enhances the
apy. In one instance, plasma exchange was used.102 Most pub- clearance of emulsion particles. This suggests that FOFEs do
lished case reports of rapid infusions of IVFEs involve SOFEs not remain in the systemic circulation long enough and may
or safflower IVFEs. In 1 case report, a 10-month-old infant not predispose patients to the complications associated with
developed fat overload syndrome while receiving a SOFE at a rapid infusion of SOFEs. Interestingly, the presence of FOFEs
dose of 5 g/kg/d for 5 weeks. The patient had many of the symp- in combination with SOFEs did not prevent the development
toms of fat overload syndrome, including fever, jaundice, and of fat overload syndrome. In 1 case report, a 2-year-old girl
bruising.103 In a study evaluating a safflower IVFE, 15 neonates developed fat overload syndrome as a result of accidental, very
were given a 1 g/kg/d dose and were evaluated for triglyceride rapid infusion of a SO/MCT/OO/FO IVFE that showed the
and free FA clearance.104 The dose was infused over 4 hours same complications seen with SOFE monotherapy.108 The
(0.25 g/kg/h), which exceeded the recommended 0.17 g/kg/h child was successfully treated with supportive care combining
limit. Peak serum triglyceride levels averaged 592 mg/dL for fluid infusion, transfusion of platelets, and substitution of
appropriate gestational age newborns and slightly higher for serum albumin (0.5 g/kg/d) and fresh-frozen plasma (10 mL/
those small for gestational age (606 mg/dL). Hyperbilirubinemia, kg). In addition, she received extra platelets, erythrocyte trans-
coagulopathies, and elevations in transaminases were also fusion, and filgrastim due to a very low leukocyte count. These
observed. Treatment included discontinuing both the PN and complications perhaps were due to the relatively low FOFE

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 605

content of the SO/MCT/OO/FO IVFE (15%) in comparison to IFALD is yet to be fully elucidated, but the combination of low
its SOFE content (30%). To date, there have been no published levels of phytosterols, high levels of -tocopherol, and a rela-
reports of fat overload syndrome involving SO/OO or SO/ tive abundance of O3FA to O6FA in FO may play an important
MCT IVFEs. Regardless of the oil source, triglyceride levels role.112 More recently, Nandivada etal116 reported the long-
should be monitored for any IVFE being rapidly infused. term effects of FOFE therapy and showed no increases in the
risk of mortality, need for intestinal transplantation, or devel-
opment of EFAD. In this cohort, biochemical markers of liver
Hepatic Complications
disease returned to normal levels within the first year of FOFE
The hepatic abnormalities induced by PN administration mani- therapy.
fest differently depending on whether they occur in adults or Many factors other than IVFE have also been implicated in
children. In adults, fat accumulation more often leads to the development of hepatic complications in PN-dependent
benign, asymptomatic steatosis, with mild to moderate (and patients. For example, recent findings from a retrospective
reversible) transaminitis and hyperbilirubinemia.109,110 In con- review in preterm infants showed that PN-associated cholesta-
trast, children more commonly develop intrahepatic cholesta- sis was more common in those receiving higher daily doses of
sis with resulting direct hyperbilirubinemia and hepatocellular dextrose after adjusting for daily lipid and protein intake.117
injury, both of which can lead to potentially irreversible Other etiologic factors in PN that may contribute to the devel-
changes such as fibrosis and cirrhosis if prolonged. Risk fac- opment of IFALD include excessive administration of calories
tors for the development of IFALD include prematurity in from overfeeding, aluminum contamination, and toxicity/defi-
infants and prolonged PN administration, sepsis, frequent sur- ciency of amino acids, among others.110 The details of how
gical procedures, and lack of enteral intake in both children these cause liver disease go beyond the scope of this review
and adults.111,112 article.
Until recently, the discontinuation of PN therapy and rein-
stitution of EN were the only known measures that prevented Restriction of SOFE. In addition to FOFE monotherapy, modi-
and/or treated IFALD. Unfortunately, this alternative is not fications in the amount and combinations of oil sources have
feasible in those patients who are unable to be weaned from PN been proposed for the treatment and prevention of IFALD in
and achieve full enteral autonomy. The potential role of IVFEs children. In a prospective study, Cober etal118 showed that
in the pathogenesis of IFALD has been recognized. For this reducing the dose of SOFE to neonates with IFALD from 3 g/
reason, current strategies focus on changing the fat source and kg/d to 1 g/kg/d led to a decline in bilirubin levels compared
reducing the IVFE dose. The use of FOFE to prevent and with a historical control cohort group. Rollins etal119 tested the
reverse the steatosis developed in a murine model of nonalco- same strategy in preventing the development of IFALD in high-
holic fatty liver disease was first described by Alwayn etal.113 risk neonates. Findings showed a slower rate of rise of markers
Based on findings from these experiments, Gura etal114 of cholestasis (ie, direct bilirubin and total bile acids) in the
reported the reversal of cholestasis in 2 infants who had devel- SOFE-restricted patients. Close monitoring of EFA status is
oped IFALD. Subsequently, Gura etal84 proved the safety and encouraged when adopting lipid reduction strategies given the
efficacy of FOFE for the treatment of IFALD in a larger cohort marked reduction in FAs being administered. In the Cober
of patients compared with a historical cohort that had been study, provisions were made to increase SOFE intake if trends
treated with SOFE. A recent prospective double-blind random- toward EFAD developed. Similarly, Sanchez etal120 reported
ized controlled trial by Lam etal115 assigned infants with the results of implementing a lipid reduction strategy in high-
IFALD to receive either SOFE or FOFE. The study was termi- risk surgical infants and compared them with a historical con-
nated prematurely due to enrollment challenges, but prelimi- trol cohort. The incidence of IFALD dropped significantly
nary results showed that infants in the FOFE group had a following the adoption of this approach. Patients from the his-
significantly slower increase in levels of direct hyperbilirubi- torical cohort were almost twice as likely to develop IFALD in
nemia and alanine aminotransferase (ALT) in comparison to comparison to those who were lipid restricted. The efficacy of
those in the SOFE group. In addition, infants receiving FOFE lipid reduction strategies in preventing IFALD did not hold true
were able to increase their intake of EN. This was associated in a more recent multicenter randomized controlled trial in pre-
with a significant clinical improvement, although it may have term neonates.121 Neonates younger than 2 days of life received
also served as a confounder contributing to this effect. Results SOFE at rates of 1 or 3 g/kg/d. No differences in the develop-
from this study continue to suggest the role of FOFEs as a ment of cholestasis or growth were observed.
potentially safe and effective alternative for the treatment of
IFALD. Although readily available in Europe and Asia, FOFE Other approaches. Combining other oil sources with FOFE has
has yet to be approved by the U.S. Food and Drug Administration been shown to be somewhat effective in comparison to SOFE
(FDA). Until then, use of FOFE in children with IFALD in the alone for the treatment of IFALD. Diamond etal122 administered
United States may only be done as part of a compassionate use both SOFE and FOFE (1 g/kg/d each) to 12 patients with short
protocol. The mechanism of action by which FOFE reverses bowel syndrome who developed IFALD. Overall, complete

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
606 Nutrition in Clinical Practice 31(5)

resolution of IFALD occurred in 9 patients. Five of these patients 8. Driscoll DF. Lipid injectable emulsions: pharmacopeial and safety
showed resolution of IFALD only once SOFE therapy was dis- issues. Pharm Res. 2006;23(9):1959-1969.
9. Das UN. Essential fatty acids: biochemistry, physiology and pathology.
continued and FOFE monotherapy was used. Biotechnol J. 2006;1(4):420-439.
Newer preparations such as SMOFlipid (Fresenius-Kabi, 10. Le HD, Meisel JA, de Meijer VE, Gura KM, Puder M. The essentiality
Uppsala, Sweden) combine multiple oil sources (MCTs, FO, of arachidonic acid and docosahexaenoic acid. Prostaglandins Leukot
OO, and SO) and have proven to be of benefit in patients with Essent Fatty Acids. 2009;81(2-3):165-170.
IFALD. Muhammed etal123 reported results from a case series 11. Waitzberg DL, Torrinhas RS, Jacintho TM. New parenteral lipid emul-
sions for clinical use. JPEN J Parenter Enteral Nutr. 2006;30(4):351-367.
of children with IFALD treated with SMOFlipid compared 12. Calder PC, Yaqoob P, Thies F, Wallace FA, Miles EA. Fatty acids and
with those who remained on SOFE. After 6 months of treat- lymphocyte functions. Br J Nutr. 2002;87(suppl 1):S31-S48.
ment, 5 of 8 patients on SMOFlipid showed resolution of jaun- 13. Mayer K, Grimm H, Grimminger F, Seeger W. Parenteral nutrition with
dice, compared with only 2 of 9 patients in the SOFE group. n-3 lipids in sepsis. Br J Nutr. 2002;87(suppl 1):S69-S75.
Interestingly, apart from the 2 patients who failed treatment (1 14. Waitzberg DL, Bellinati-Pires R, Salgado MM, etal. Effect of total par-
enteral nutrition with different lipid emulsions of human monocyte and
died, 1 listed for transplant), the remaining 6 patients in the neutrophil functions. Nutrition. 1997;13(2):128-132.
SMOFlipid group showed a sudden and sustained fall in biliru- 15. Furukawa K, Yamamori H, Takagi K, etal. Influences of soybean oil
bin levels as early as 1 month after switching from SOFE. emulsion on stress response and cell-mediated immune function in mod-
Another group in Australia has shown similar results when erately or severely stressed patients. Nutrition. 2002;18(3):235-240.
comparing SMOFlipid with SOFE in preterm infants with 16. Iyer KR, Spitz L, Clayton P. BAPS prize lecture: new insight into
mechanisms of parenteral nutritionassociated cholestasis: role of plant
IFALD, with significant reduction in the rate of increase, and sterols. British Association of Paediatric Surgeons. J Pediatr Surg.
then reversal, of direct bilirubin levels.124 1998;33(1):1-6.
17. Salen G, Ahrens EH, Grundy SM. Metabolism of -sitosterol in man. J
Clin Invest. 1970;49(5):952-967.
The Ideal IVFE 18. Lees AM, Mok HY, Lees RS, McCluskey MA, Grundy SM. Plant sterols as
cholesterol-lowering agents: clinical trials in patients with hypercholester-
The ideal emulsion would be one that reversed or prevented
olemia and studies of sterol balance. Atherosclerosis. 1977;28(3):325-338.
EFAD without leading to hepatic steatosis, hypertriglyceride- 19. Kakis G, Kuksis A. Effect of intravenous infusion of Intralipid, choles-
mia, or the elevation of liver enzymes, while simultaneously terol, and plant sterols on hepatic cholesterogenesis. Can J Biochem Cell
facilitating normal growth and development. Modifications in Biol. 1984;62(1):1-10.
20. Shefer S, Salen G, Nguyen L, etal. Competitive inhibition of bile acid
their ingredients, formulation, and dosing have made IVFEs a
synthesis by endogenous cholestanol and sitosterol in sitosterolemia with
relatively safe component alone or added to PN formulations. xanthomatosis: effect on cholesterol 7 alpha-hydroxylase. J Clin Invest.
The ideal IVFE, however, has yet to be developed. 1988;82(6):1833-1839.
21. Clayton PT, Whitfield P, Iyer K. The role of phytosterols in the patho-
genesis of liver complications of pediatric parenteral nutrition. Nutrition.
Statement of Authorship
1998;14(1):158-164.
L. Anez-Bustillos, D. T. Dao, M. A. Baker, G. L. Fell, M. Puder, 22. Savini S, DAscenzo R, Biagetti C, etal. The effect of 5 intravenous
and K. M. Gura contributed to the conception of the manuscript; lipid emulsions on plasma phytosterols in preterm infants receiving par-
and L. Anez-Bustillos and K. M. Gura drafted the manuscript. All enteral nutrition: a randomized clinical trial. Am J Clin Nutr. 2013;98(2):
of the authors critically revised the manuscript, gave final 312-318.
23. Carter BA, Taylor OA, Prendergast DR, etal. Stigmasterol, a soy lipid
approval, and agree to be accountable for all aspects of work
derived phytosterol, is an antagonist of the bile acid nuclear receptor
ensuring integrity and accuracy of the content of the manuscript. FXR. Pediatr Res. 2007;62(3):301-306.
24. Liz R, Pereira DF, Horst H, etal. Protected effect of Esenbeckia
References leiocarpa upon the inflammatory response induced by carrageenan
in a murine air pouch model. Int Immunopharmacol. 2011;11(12):
1. Carpentier YA, Dupont IE. Advances in intravenous lipid emulsions. 1991-1999.
World J Surg. 2000;24(12):1493-1497. 25. Liz R, Zanatta L, dos Reis GO, etal. Acute effect of -sitosterol on cal-
2. Vinnars E, Hammarqvist F. 25th Arvid Wretlinds Lecturesilver cium uptake mediates anti-inflammatory effect in murine activated neu-
anniversary, 25 years with ESPEN, the history of nutrition. Clin Nutr. trophils. J Pharm Pharmacol. 2013;65(1):115-122.
2004;23(5):955-962. 26. Byrne WJ. Intralipid or Liposyncomparable products? J Pediatr
3. Geyer RP. Parenteral nutrition. Physiol Rev. 1960;40:150-186. Gastroenterol Nutr. 1982;1(1):7-8.
4. Wretlind A. Development of fat emulsions. JPEN J Parenter Enteral 27. Holman RT, Johnson SB, Hatch TF. A case of human linolenic acid
Nutr. 1981;5(3):230-235. deficiency involving neurological abnormalities. Am J Clin Nutr.
5. Ferezou J, Bach AC. Structure and metabolic fate of triacylglycerol- 1982;35(3):617-623.
and phospholipid-rich particles of commercial parenteral fat emulsions. 28. Ulrich H, Pastores SM, Katz DP, Kvetan V. Parenteral use of medium-
Nutrition. 1999;15(1):44-50. chain triglycerides: a reappraisal. Nutrition. 1996;12(4):231-238.
6. Lunn M, Fausnight T. Hypersensitivity to total parenteral nutri- 29. Mok KT, Maiz A, Yamazaki K, etal. Structured medium-chain and long-
tion fat-emulsion component in an egg-allergic child. Pediatrics. chain triglyceride emulsions are superior to physical mixtures in sparing
2011;128(4):1025-1028. body protein in the burned rat. Metabolism. 1984;33(10):910-915.
7. Chapter 729: Globule size distribution in lipid injectable emulsions. In: 30. Driscoll DF, Nehne J, Peterss H, Franke R, Bistrian BR, Niemann W.
United States Pharmacopeia and National Formulary (USP 36-NF31). The influence of medium-chain triglycerides on the stability of all-in-one
Rockville, MD: U.S. Pharmacopeia; 2012:321-323. formulations. Int J Pharm. 2002;240(1-2):1-10.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 607

31. Clinolipid [package insert]. Deerfield, IL: Baxter Healthcare Corporation; 53. Corpeleijn WE, Vermeulen MJ, van den Akker CH, van Goudoever JB.
2014. Feeding very-low-birth-weight infants: our aspirations versus the reality
32. Sala-Vila A, Barbosa VM, Calder PC. Olive oil in parenteral nutrition. in practice. Ann Nutr Metab. 2011;58(suppl 1):20-29.
Curr Opin Clin Nutr Metab Care. 2007;10(2):165-174. 54. Qi K, Seo T, Al-Haideri M, etal. Omega-3 triglycerides modify blood
33. Deshpande GC, Simmer K, Mori T, Croft K. Parenteral lipid emulsions clearance and tissue targeting pathways of lipid emulsions. Biochemistry.
based on olive oil compared with soybean oil in preterm (<28 weeks ges- 2002;41(9):3119-3127.
tation) neonates: a randomised controlled trial. J Pediatr Gastroenterol 55. Barness L. Pediatric Nutrition Handbook. Elk Grove Village, IL:
Nutr. 2009;49(5):619-625. American Academy of Pediatrics; 2009.
34. Webb AN, Hardy P, Peterkin M, etal. Tolerability and safety of olive 56. Brouwer CB, de Bruin TW, Jansen H, Erkelens DW. Different clearance
oilbased lipid emulsion in critically ill neonates: a blinded randomized of intravenously administered olive oil and soybean-oil emulsions: role
trial. Nutrition. 2008;24(11-12):1057-1064. of hepatic lipase. Am J Clin Nutr. 1993;57(4):533-539.
35. Roggero P, Mosca F, Giann ML, etal. F2-isoprostanes and total radical- 57. Haumont D, Richelle M, Deckelbaum RJ, Coussaert E, Carpentier YA.
trapping antioxidant potential in preterm infants receiving parenteral Effect of liposomal content of lipid emulsions on plasma lipid concentra-
lipid emulsions. Nutrition. 2010;26(5):551-555. tions in low birth weight infants receiving parenteral nutrition. J Pediatr.
36. Demirel G, Oguz SS, Celik IH, Erdeve O, Uras N, Dilmen U. The meta- 1992;121(5, pt 1):759-763.
bolic effects of two different lipid emulsions used in parenterally fed 58. Haumont D, Deckelbaum RJ, Richelle M, etal. Plasma lipid and
premature infantsa randomized comparative study. Early Hum Dev. plasma lipoprotein concentrations in low birth weight infants given par-
2012;88(7):499-501. enteral nutrition with twenty or ten percent lipid emulsion. J Pediatr.
37. van Kempen AAMW, van der Crabben SN, Ackermans MT, Endert E, 1989;115(5, pt 1):787-793.
Kok JH, Sauerwein HP. Stimulation of gluconeogenesis by intravenous 59. Kalfarentzos F, Kokkinis K, Leukaditi K, Maroulis J, Onoufriou A,
lipids in preterm infants: response depends on fatty acid profile. Am J Alexopoulos K. Comparison between two fat emulsions: Intralipid 30 cent
Physiol Endocrinol Metab. 2006;290(4):E723-E730. vs intralipid 10 cent in critically ill patients. Clin Nutr. 1998;17(1):31-34.
38. Kalish BT, Fallon EM, Puder M. A tutorial on fatty acid biology. JPEN
60. Seidner DL, Mascioli EA, Istfan NW, etal. Effects of long-chain triglyc-
J Parenter Enteral Nutr. 2012;36(4):380-388.
eride emulsions on reticuloendothelial system function in humans. JPEN
39. Gawecka A, Michalkiewicz J, Kornacka MK, Luckiewicz B,
J Parenter Enteral Nutr. 1989;13(6):614-619.
Kubiszewska I. Immunologic properties differ in preterm infants fed
61. Nugent KM. Intralipid effects on reticuloendothelial function. J Leukoc
olive oil vs soy-based lipid emulsions during parenteral nutrition. JPEN
Biol. 1984;36(2):123-132.
J Parenter Enteral Nutr. 2008;32(4):448-453.
62. Rssle C, Carpentier YA, Richelle M, etal. Medium-chain triglycerides
40. Bagel S, Dessaigne B, Bourdeaux D, etal. Influence of lipid type on bis
induce alterations in carnitine metabolism. Am J Physiol. 1990;258(6, pt
(2-ethylhexyl)phthalate (DEHP) leaching from infusion line sets in par-
1):E944-E947.
enteral nutrition. JPEN J Parenter Enteral Nutr. 2011;35(6):770-775.
63. Rebouche CJ, Panagides DD, Nelson SE. Role of carnitine in utilization
41. de Meijer VE, Le HD, Meisel JA, Gura KM, Puder M. Parenteral fish
of dietary medium-chain triglycerides by term infants. Am J Clin Nutr.
oil as monotherapy prevents essential fatty acid deficiency in paren-
1990;52(5):820-824.
teral nutritiondependent patients. J Pediatr Gastroenterol Nutr.
64. Johnson RC, Young SK, Cotter R, Lin L, Rowe WB. Medium-chain-
2010;50(2):212-218.
triglyceride lipid emulsion: metabolism and tissue distribution. Am J
42. Gura KM, Parsons SK, Bechard LJ, etal. Use of a fish oilbased lipid
Clin Nutr. 1990;52(3):502-508.
emulsion to treat essential fatty acid deficiency in a soy allergic patient
65. Oliveira FL, Rumsey SC, Schlotzer E, Hansen I, Carpentier YA,
receiving parenteral nutrition. Clin Nutr. 2005;24(5):839-847.
Deckelbaum RJ. Triglyceride hydrolysis of soy oil vs fish oil emulsions.
43. Le HD, Meisel JA, de Meijer VE, etal. Docosahexaenoic acid and ara-
JPEN J Parenter Enteral Nutr. 1997;21(4):224-229.
chidonic acid prevent essential fatty acid deficiency and hepatic steato-
66. Nestel PJ. Effects of n-3 fatty acids on lipid metabolism. Annu Rev Nutr.
sis. JPEN J Parenter Enteral Nutr. 2012;36(4):431-441.
1990;10:149-167.
44. Raptis DA, Limani P, Jang JH, etal. GPR120 on Kupffer cells mediates
67. Roche HM, Gibney MJ. Long-chain n-3 polyunsaturated fatty acids
hepatoprotective effects of 3-fatty acids. J Hepatol. 2014;60(3):625-632.
and triacylglycerol metabolism in the postprandial state. Lipids.
45. Khan MZ, He L. The role of polyunsaturated fatty acids and GPR40
1999;34(suppl):S259-S265.
receptor in brain [published online May 22, 2015]. Neuropharmacology.
68. Spear ML, Stahl GE, Hamosh M, etal. Effect of heparin dose
46. Williams-Bey Y, Boularan C, Vural A, etal. Omega-3 free fatty acids
and infusion rate on lipid clearance and bilirubin binding in pre-
suppress macrophage inflammasome activation by inhibiting NF-B
mature infants receiving intravenous fat emulsions. J Pediatr.
activation and enhancing autophagy. PLoS One. 2014;9(6):e97957. 1988;112(1):94-98.
47. Cornish J, MacGibbon A, Lin J-M, etal. Modulation of osteoclastogen- 69. Berkow SE, Spear ML, Stahl GE, etal. Total parenteral nutrition with
esis by fatty acids. Endocrinology. 2008;149(11):5688-5695. Intralipid in premature infants receiving TPN with heparin: effect on
48. de Roos B, Mavrommatis Y, Brouwer IA. Long-chain n-3 polyunsatu- plasma lipolytic enzymes, lipids, and glucose. J Pediatr Gastroenterol
rated fatty acids: new insights into mechanisms relating to inflammation Nutr. 1987;6(4):581-588.
and coronary heart disease. Br J Pharmacol. 2009;158(2):413-428. 70. Kao LC, Cheng MH, Warburton D. Triglycerides, free fatty acids, free
49. Serhan CN, Chiang N. Endogenous pro-resolving and anti-inflam- fatty acids/albumin molar ratio, and cholesterol levels in serum of neo-
matory lipid mediators: a new pharmacologic genus. Br J Pharmacol. nates receiving long-term lipid infusions: controlled trial of continuous
2008;153(suppl):S200-S215. and intermittent regimens. J Pediatr. 1984;104(3):429-435.
50. Lee S, Gura KM, Kim S, Arsenault DA, Bistrian BR, Puder M. Current 71. Carpentier YA, Hacquebard M, Portois L, Dupont IE, Deckelbaum RJ,
clinical applications of omega-6 and omega-3 fatty acids. Nutr Clin Malaisse WJ. Rapid cellular enrichment of eicosapentaenoate after a sin-
Pract. 2006;21(4):323-341. gle intravenous injection of a novel medium-chain triacylglycerol:fish-
51. Vanek VW, Seidner DL, Allen P, etal. A.S.P.E.N. position paper: oil emulsion in humans. Am J Clin Nutr. 2010;91(4):875-882.
clinical role for alternative intravenous fat emulsions. Nutr Clin Pract. 72. Gervasio J. Total nutrient admixtures (3-in-1): pros vs cons for adults.
2012;27(2):150-192. Nutr Clin Pract. 2015;30(3):331-335.
52. Siderova VS, Carpentier YA, Dahlan W, Richelle M. Intravascular 73. Boullata JI, Gilbert K, Sacks G, etal. A.S.P.E.N. clinical guidelines:
metabolism of different fatty acids during lipid infusion in man. Clin parenteral nutrition ordering, order review, compounding, labeling, and
Nutr. 1993;12(6):329-336. dispensing. JPEN J Parenter Enteral Nutr. 2014;38(3):334-377.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
608 Nutrition in Clinical Practice 31(5)

74. Le HD, Fallon EM, Kalish BT, etal. The effect of varying ratios of 95. Kessler U, Zachariou Z, Raz D, Poeschl J, Linderkamp O. Effects of
docosahexaenoic acid and arachidonic acid in the prevention and rever- Intralipid infusion on hemorheology and peripheral resistance in neo-
sal of biochemical essential fatty acid deficiency in a murine model. nates and children. Pediatr Surg Int. 2005;21(3):197-202.
Metabolism. 2013;62(4):499-508. 96. Brans YW, Dutton EB, Andrew DS, Menchaca EM, West DL.
75. Aaes-Jorgensen E, Leppik EE, Hayes HW, Holman RT. Essential fatty Fat emulsion tolerance in very low birth weight neonates: effect
acid deficiency, II: in adult rats. J Nutr. 1958;66(2):245-259. on diffusion of oxygen in the lungs and on blood pH. Pediatrics.
76. Fleming CR, Smith LM, Hodges RE. Essential fatty acid defi- 1986;78(1):79-84.
ciency in adults receiving total parenteral nutrition. Am J Clin Nutr. 97. Sosenko IR, Rodriguez-Pierce M, Bancalari E. Effect of early initia-
1976;29(9):976-983. tion of intravenous lipid administration on the incidence and severity
77. Holman RT. The ratio of trienoic: tetraenoic acids in tissue lipids as a of chronic lung disease in premature infants. J Pediatr. 1993;123(6):
measure of essential fatty acid requirement. J Nutr. 1960;70:405-410. 975-982.
78. Innis SM. Essential fatty acids in infant nutrition: lessons and limitations 98. Paolisso G, Manzella D, Rizzo MR, etal. Elevated plasma fatty acid con-
from animal studies in relation to studies on infant fatty acid require- centrations stimulate the cardiac autonomic nervous system in healthy
ments. Am J Clin Nutr. 2000;71(1)(suppl):238S-244S. subjects. Am J Clin Nutr. 2000;72(3):723-730.
79. Reisbick S, Neuringer M, Hasnain R, Connor WE. Polydipsia in 99. Mathru M, Dries DJ, Zecca A, Fareed J, Rooney MW, Rao TL.
rhesus monkeys deficient in omega-3 fatty acids. Physiol Behav. Effect of fast vs slow Intralipid infusion on gas exchange, pulmonary
1990;47(2):315-323. hemodynamics, and prostaglandin metabolism. Chest. 1991;99(2):
80. Reisbick S, Neuringer M, Hasnain R, Connor WE. Home cage behavior 426-429.
of rhesus monkeys with long-term deficiency of omega-3 fatty acids. 100. Hayes BD, Gosselin S, Calello DP, etal. Systematic review of clinical
Physiol Behav. 1994;55(2):231-239. adverse events reported after acute intravenous lipid emulsion adminis-
81. Connor WE. Importance of n-3 fatty acids in health and disease. Am J tration. Clin Toxicol (Phila). 2016;54(5):1-40.
Clin Nutr. 2000;71(1)(suppl):171S-175S. 101. Koletzko B, Goulet O, Hunt J, Krohn K, Shamir R. 1. Guidelines on
82. Peet M, Murphy B, Shay J, Horrobin D. Depletion of omega-3 fatty paediatric parenteral nutrition of the European Society of Paediatric
acid levels in red blood cell membranes of depressive patients. Biol Gastroenterology, Hepatology and Nutrition (ESPGHAN) and the
Psychiatry. 1998;43(5):315-319. European Society for Clinical Nutrition and Metabolism (ESPEN), sup-
83. Calder PC, Jensen GL, Koletzko BV, Singer P, Wanten GJA. Lipid ported by the European Society of Paediatric Research (ESPR). J Pediatr
emulsions in parenteral nutrition of intensive care patients: cur- Gastroenterol Nutr. 2005;41(suppl 2):S1-S87.
rent thinking and future directions. Intensive Care Med. 2010;36(5): 102. Kollef MH, McCormack MT, Caras WE, Reddy VV, Bacon D. The fat
735-749. overload syndrome: successful treatment with plasma exchange. Ann
84. Gura KM, Lee S, Valim C, etal. Safety and efficacy of a fish-oil-based Intern Med. 1990;112(7):545-546.
fat emulsion in the treatment of parenteral nutritionassociated liver dis- 103. Heyman MB, Storch S, Ament ME. The fat overload syndrome: report of
ease. Pediatrics. 2008;121(3):e678-e686. a case and literature review. Am J Dis Child. 1981;135(7):628-630.
85. Critical Care Nutrition. Canadian Clinical Practice Guidelines 10.2: 104. Burckart GJ, Whitington PF, Halbrehder DK, Helms RA. Triglyceride
Strategies to Optimize Parenteral Nutrition and Minimize Risks: Use of and fatty acid clearance in neonates following safflower oil emulsion
Lipids. 2013. www.criticalcarenutrition.com. Accessed April 14, 2016. infusion. JPEN J Parenter Enteral Nutr. 1983;7(3):251-253.
86. Edmunds CE, Brody RA, Parrott JS, Stankorb SM, Heyland DK. The 105. Gura KM, Puder M. Rapid infusion of fish oilbased emulsion in infants
effects of different IV fat emulsions on clinical outcomes in critically ill does not appear to be associated with fat overload syndrome. Nutr Clin
patients. Crit Care Med. 2014;42(5):1168-1177. Pract. 2010;25(4):399-402.
87. McClave SA, Taylor BE, Martindale RG, etal. Guidelines for the provi- 106. Ton MN, Chang C, Carpentier YA, Deckelbaum RJ. In vivo and in vitro
sion and assessment of nutrition support therapy in the adult critically properties of an intravenous lipid emulsion containing only medium
ill patient: Society of Critical Care Medicine (SCCM) and American chain and fish oil triglycerides. Clin Nutr. 2005;24(4):492-501.
Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J 107. Park Y, Harris WS. Omega-3 fatty acid supplementation accelerates chy-
Parenter Enteral Nutr. 2016;40(2):159-211. lomicron triglyceride clearance. J Lipid Res. 2003;44(3):455-463.
88. Singer P, Berger MM, Van den Berghe G, etal. ESPEN guidelines on 108. Hojsak I, Kolacek S. Fat overload syndrome after the rapid infusion
parenteral nutrition: intensive care. Clin Nutr. 2009;28(4):387-400. of SMOFlipid emulsion. JPEN J Parenter Enteral Nutr. 2014;38(1):
89. Critical Care Nutrition. Canadian Clinical Practice Guidelines 9.2 119-121.
Composition of Parenteral Nutrition: Type of Lipids. 2013. www.criti- 109. Angelico M, Della Guardia P. Review article: hepatobiliary complica-
calcarenutrition.com. Accessed April 14, 2016. tions associated with total parenteral nutrition. Aliment Pharmacol Ther.
90. Manzanares W, Langlois PL, Heyland DK. The role of alternative lipid 2000;14(suppl 2):54-57.
emulsions in critically ill patients: what the evidence shows. JPEN J 110. Kumpf VJ. Parenteral nutritionassociated liver disease in adult and
Parenter Enteral Nutr. 2014;38(6):653-654. pediatric patients. Nutr Clin Pract. 2006;21(3):279-290.
91. Manzanares W, Langlois PL, Dhaliwal R, Lemieux M, Heyland DK. 111. Beath S V, Davies P, Papadopoulou A, etal. Parenteral nutritionrelated
Intravenous fish oil lipid emulsions in critically ill patients: an updated cholestasis in postsurgical neonates: multivariate analysis of risk factors.
systematic review and meta-analysis. Crit Care. 2015;19:167. J Pediatr Surg. 1996;31(4):604-606.
92. Kapoor V, Glover R, Malviya MN. Alternative lipid emulsions versus 112. Nandivada P, Cowan E, Carlson SJ, Chang M, Gura KM, Puder M.
pure soy oil based lipid emulsions for parenterally fed preterm infants. Mechanisms for the effects of fish oil lipid emulsions in the management
Cochrane Database Syst Rev. 2015;(12):CD009172. of parenteral nutritionassociated liver disease. Prostaglandins Leukot
93. Wales PW, Allen N, Worthington P, etal. A.S.P.E.N. clinical guide- Essent Fat Acids. 2013;89(4):153-158.
lines: support of pediatric patients with intestinal failure at risk of paren- 113. Alwayn IPJ, Gura K, Nos V, etal. Omega-3 fatty acid supplementation
teral nutritionassociated liver disease. JPEN J Parenter Enteral Nutr. prevents hepatic steatosis in a murine model of nonalcoholic fatty liver
2014;38(5):538-557. disease. Pediatr Res. 2005;57(3):445-452.
94. Goulet O, Girot R, Maier-Redelsperger M, Bougle D, Virelizier J, 114. Gura KM, Duggan CP, Collier SB, etal. Reversal of parenteral nutri-
Ricour C. Hematologic disorders following prolonged use of intra- tionassociated liver disease in two infants with short bowel syndrome
venous fat emulsions in children. JPEN J Parenter Enteral Nutr. using parenteral fish oil: implications for future management. Pediatrics.
1986;10(3):284-288. 2006;118(1):e197-e201.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016
Anez-Bustillos et al 609

115. Lam HS, Tam YH, Poon TCW, etal. A double-blind randomised con- 127. Lindgren BF, Ruokonen E, Magnusson-Borg K, Takala J. Nitrogen
trolled trial of fish oilbased versus soy-based lipid preparations in the sparing effect of structured triglycerides containing both medium-and
treatment of infants with parenteral nutritionassociated cholestasis. long-chain fatty acids in critically ill patients: a double blind randomized
Neonatology. 2014;105(4):290-296. controlled trial. Clin Nutr. 2001;20(1):43-48.
116. Nandivada P, Fell GL, Mitchell PD, etal. Long-term fish oil lipid emul- 128. Garnacho-Montero J, Ortiz-Leyba C, Jimnez-Jimnez FJ, etal. Clinical
sion use in children with intestinal failure-associated liver disease [pub- and metabolic effects of two lipid emulsions on the parenteral nutrition
lished online March 9, 2016]. JPEN J Parenter Enteral Nutr. of septic patients. Nutrition. 2002;18(2):134-138.
117. Gupta K, Wang H, Amin SB. Parenteral nutritionassociated cholestasis 129. Grecu I, Mirea L, Grintescu I. Parenteral fish oil supplementation in
in premature infants: role of macronutrients. JPEN J Parenter Enteral patients with abdominal sepsis. Clin Nutr. 2003;22:S23.
Nutr. 2016;40(3):335-341. 130. Huschak G, Zur Nieden K, Hoell T, Riemann D, Mast H, Stuttmann
118. Cober MP, Killu G, Brattain A, Welch KB, Kunisaki SM, Teitelbaum R. Olive oil based nutrition in multiple trauma patients: a pilot study.
DH. Intravenous fat emulsions reduction for patients with parenteral Intensive Care Med. 2005;31(9):1202-1208.
nutritionassociated liver disease. J Pediatr. 2012;160(3):421-427. 131. Iovinelli G, Marinangeli F, Ciccone A, etal. Parenteral nutrition in
119. Rollins MD, Ward RM, Jackson WD, etal. Effect of decreased paren- ventilated patients with chronic obstructive pulmonary disease: long
teral soybean lipid emulsion on hepatic function in infants at risk for par- chain vs medium chain triglycerides. Minerva Anestesiol. 2007;73
enteral nutritionassociated liver disease: a pilot study. J Pediatr Surg. (1-2):65-76.
2013;48(6):1348-1356. 132. Garca-de-Lorenzo A, Denia R, Atlan P, etal. Parenteral nutrition pro-
120. Sanchez SE, Braun LP, Mercer LD, Sherrill M, Stevens J, Javid PJ. viding a restricted amount of linoleic acid in severely burned patients: a
The effect of lipid restriction on the prevention of parenteral nutrition randomised double-blind study of an olive oilbased lipid emulsion v.
associated cholestasis in surgical infants. J Pediatr Surg. 2013;48(3): medium/long-chain triacylglycerols. Br J Nutr. 2005;94(2):221-230.
573-578. 133. Friesecke S, Lotze C, Khler J, Heinrich A, Felix SB, Abel P. Fish
121. Levit OL, Calkins KL, Gibson LC, etal. Low-dose intravenous soybean oil supplementation in the parenteral nutrition of critically ill medi-
oil emulsion for prevention of cholestasis in preterm neonates. JPEN J cal patients: a randomised controlled trial. Intensive Care Med.
Parenter Enteral Nutr. 2016;40(3):374-382. 2008;34(8):1411-1420.
122. Diamond IR, Sterescu A, Pencharz PB, Kim JH, Wales PW. Changing 134. Wang X, Li W, Zhang F, Pan L, Li N, Li J. Fish oilsupplemented paren-
the paradigm: Omegaven for the treatment of liver failure in pediat- teral nutrition in severe acute pancreatitis patients and effects on immune
ric short bowel syndrome. J Pediatr Gastroenterol Nutr. 2009;48(2): function and infectious risk: a randomized controlled trial. Inflammation.
209-215. 2009;32(5):304-309.
123. Muhammed R, Bremner R, Protheroe S, Johnson T, Holden C, Murphy 135. Barbosa VM, Miles EA, Calhau C, Lafuente E, Calder PC. Effects of a
MS. Resolution of parenteral nutritionassociated jaundice on chang- fish oil containing lipid emulsion on plasma phospholipid fatty acids,
ing from a soybean oil emulsion to a complex mixed-lipid emulsion. J inflammatory markers, and clinical outcomes in septic patients: a ran-
Pediatr Gastroenterol Nutr. 2012;54(6):797-802. domized, controlled clinical trial. Crit Care. 2010;14(1):R5.
124. Attard M, Patel N, Simpson J. Change from Intralipid to SMOF lipid is 1 36. Umpierrez GE, Spiegelman R, Zhao V, etal. A double-blind,

associated with improved liver function in infants with PN associated randomized clinical trial comparing soybean oilbased versus
liver disease. Arch Dis Child. 2012;97(suppl 1):A54-A55. olive oilbased lipid emulsions in adult medical-surgical intensive
125. Vanek VW, Seidner DL, Allen P, etal. A.S.P.E.N. position paper: care unit patients requiring parenteral nutrition. Crit Care Med.
clinical role for alternative intravenous fat emulsions. Nutr Clin Pract. 2012;40(6):1792-1798.
2012;27(2):150-192. 137. Pontes-Arruda A, Dos Santos MCFC, Martins LF, etal. Influence of par-
126. Nijveldt RJ, Tan AM, Prins HA, etal. Use of a mixture of medium-chain enteral nutrition delivery system on the development of bloodstream infec-
triglycerides and long-chain triglycerides versus long-chain triglycerides tions in critically ill patients: an international, multicenter, prospective,
in critically ill surgical patients: a randomized prospective double-blind open-label, controlled studyEPICOS study. JPEN J Parenter Enteral
study. Clin Nutr. 1998;17(1):23-29. Nutr. 2012;36(5):574-586.

Downloaded from ncp.sagepub.com at American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) on October 31, 2016

You might also like