Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Review

Journal of Parenteral and Enteral


Nutrition
Essential Fatty Acid Requirements and Intravenous Volume 00 Number 0
xxx 2019 1–11
Lipid Emulsions 
C 2019 American Society for

Parenteral and Enteral Nutrition


DOI: 10.1002/jpen.1537
wileyonlinelibrary.com

Leah Gramlich, MD1 ; Carol Ireton-Jones, PhD2 ; John M. Miles, MD3 ;


Maya Morrison, MSRD4 ; and Alessandro Pontes-Arruda, MD4

Abstract
Linoleic acid (LA) and α-linolenic acid (ALA) must be supplied to the human body and are therefore considered essential fatty
acids. This narrative review discusses the signs, symptoms, diagnosis, prevention, and treatment of essential fatty acid deficiency
(EFAD). EFAD may occur in patients with conditions that severely limit the intake, digestion, absorption, and/or metabolism of
fat. EFAD may be prevented in patients requiring parenteral nutrition by inclusion of an intravenous lipid emulsion (ILE) as a
source of LA and ALA. Early ILEs consisted solely of soybean oil (SO), a good source of LA and ALA, but being rich in LA
may promote the production of proinflammatory fatty acids. Subsequent ILE formulations replaced part of the SO with other fat
sources to decrease the amount of proinflammatory fatty acids. Although rare, EFAD is diagnosed by an elevated triene:tetraene
(T:T) ratio, which reflects increased metabolism of oleic acid to Mead acid in the absence of adequate LA and ALA. Assays for
measuring fatty acids have improved over the years, and therefore it is necessary to take into account the particular assay used
and its reference range when determining if the T:T ratio indicates EFAD. In patients with a high degree of suspicion for EFAD,
obtaining a fatty acid profile may provide additional useful information for making a diagnosis of EFAD. In patients receiving an
ILE, the T:T ratio and fatty acid profile should be interpreted in light of the fatty acid composition of the ILE to ensure accurate
diagnosis of EFAD. (JPEN J Parenter Enteral Nutr. 2019;00:1–11)

Keywords
essential fatty acids; essential fatty acid deficiency; essential fatty acid requirements; intravenous fat emulsion; intravenous lipid
emulsion; parenteral nutrition

Introduction bonds in the acyl chain) or saturated (ie, no double bonds


in the acyl chain).1 The human body is able to synthesize
Fatty acids are hydrocarbon chains of variable length, with most fatty acids or obtain them from the diet.1,2 However,
a methyl group at one end and a carboxyl group at the other 2 polyunsaturated fatty acids, linoleic acid (LA; 18:2n-6, an
end.1 Fatty acids can be unsaturated (ie, 1 or more double n-6 fatty acid) and α-linolenic acid (ALA; 18:3n-3, an n-3

From the 1 Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada; 2 Good Nutrition for Good Living, Dallas, Texas,
USA; 3 Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA; and 4 Baxter Healthcare
Corporation, Deerfield, Illinois, USA.
Financial disclosure: This review was sponsored by Baxter Healthcare Corporation. Medical writing assistance was provided by Justine Southby,
PhD, CMPP, and Tania Dickson, PhD, CMPP, of ProScribe Envision Pharma Group and was funded by Baxter Healthcare Corporation.
ProScribe’s services complied with international guidelines for Good Publication Practice (GPP3).
Conflicts of interest: L. Gramlich has been on advisory boards for Shire, has received research funding from Baxter Healthcare Corporation,
Abbott Laboratories, and Fresenius Kabi, and has participated in Speaker’s Bureaus for Baxter Healthcare Corporation, Abbott Laboratories,
Fresenius Kabi, and Shire. C. Ireton-Jones has participated in a Speaker’s Bureau for Fresenius Kabi and is an employee of Nutrishare Inc (Elk
Grove, CA, USA). J. M. Miles has served as a consultant for Baxter Healthcare Corporation. M. Morrison and A. Pontes-Arruda are employees
of and own shares in Baxter International Inc, the corporate parent of Baxter Healthcare Corporation. Baxter Healthcare Corporation was
involved in the preparation of the manuscript.
Received for publication August 30, 2018; accepted for publication February 28, 2019.
This article originally appeared online on xxxx 0, 2019.
Corresponding author:
Maya Morrison, MSRD, Baxter Healthcare Corporation, 1 Baxter Parkway, Deerfield, IL 60015, USA.
Email: maya_dally@baxter.com
2 Journal of Parenteral and Enteral Nutrition 00(0)

Figure 1. Fatty acid structures and biosynthetic pathways. Adapted from Lee S, et al. Current clinical applications of Ω-6 and
Ω-3 fatty acids. Nutr Clin Pract. 2006;21:323-341. Copyright 2006 American Society for Parenteral and Enteral Nutrition, with
permission from John Wiley and Sons.

fatty acid), cannot be synthesized by humans because they anti-inflammatory.5 As fatty acids are involved in so many
lack the desaturase enzymes capable of catalyzing double physiological processes, changes in fatty acid intake can have
bond formation at the n-6 or n-3 position of the hydrocar- major effects.1,2
bon chain (counting from the methyl carbon), respectively.2 Essential fatty acid deficiency (EFAD) is prevented
Thus, these 2 fatty acids can only be obtained from the diet if 2%–4% of total energy comes from LA and 0.25%–
and are considered essential fatty acids (EFAs).1,2 0.5% from ALA, and thus it is not difficult to meet these
The EFAs LA and ALA serve as parent n-6 and n- requirements under usual circumstances.6,7 As a result,
3 fatty acids, respectively, giving rise to longer-chain fatty EFAD is rare in healthy children and adults who have a
acids through desaturation and elongation (Figure 1). For varied diet with an adequate intake of EFAs.5,8,9 In addition,
example, LA is metabolized to arachidonic acid (AA; because adipose tissue contains LA, mobilization of EFAs
20:4n-6), and ALA is metabolized to docosahexaenoic acid from adipose tissue may prevent EFAD when the supply of
(DHA; 22:6n-3) and eicosapentaenoic acid (EPA; 20:5n- exogenous fat is insufficient, provided that the individual has
3) (Figure 1).1,3 These longer-chain fatty acids give rise adequate fat stores.8,10 However, patients with conditions
to bioactive lipid mediators in the form of eicosanoids that severely limit the intake, digestion, absorption,
(prostaglandins, thromboxanes, and leukotrienes), which and/or metabolism of fat are at risk of developing EFAD,
are growth factors and inflammatory mediators, and influ- especially if fat stores are depleted. This includes patients
ence gene expression.1,4 The n-6-derived eicosanoids (eg, with chronic fat malabsorption because of pancreatic
from AA) are considered proinflammatory, whereas the insufficiency, massive bowel resection or mucosal
n-3-derived eicosanoids (eg, from EPA) are considered disease,9,11,12 patients with cystic fibrosis,13 children
Gramlich et al 3

receiving parenteral nutrition (PN) who are subjected to the Diagnostic Testing section).6,9 Other nonspecific bio-
severe soybean oil (SO) lipid restriction,14 and preterm chemical manifestations of EFAD include elevated liver
infants.15 In preterm infants, the risk of EFAD is related enzymes, hyperlipidemia, thrombocytopenia, and altered
not only to their limited fat stores but also to their reduced platelet aggregation.9 Thus, in patients at risk of EFAD
ability to synthesize AA and DHA, which are in high and exhibiting these biochemical manifestations, assess-
demand during brain and retinal development, and are ment of EFAD should be considered. It should be noted
therefore considered “conditionally essential.”16-19 that elevated liver enzymes, hyperlipidemia, and altered
Although EFAD is generally rare, it became a significant platelet aggregation can also be a consequence of ILE
concern for patients receiving the early formulations of administration.35 Clinical manifestations of EFAD include
lipid-free PN.5,20 In the 1970s, there were many reports of hair loss, impaired wound healing, increased susceptibility
EFAD in infant and adult patients receiving PN.21-27 To to infection, reduced growth rate in infants and children,
provide an adequate amount of energy, early PN formula- and a dry scaly rash arising from increased transepidermal
tions consisted primarily of high concentrations of glucose water loss due to increased water permeability of the skin
and amino acids and did not provide EFAs because lipid (see Fleming et al25 for a photograph of the type of rash
emulsions for intravenous administration did not become associated with EFAD).6,9,21,25,36,37
available in the United States until the 1970s.3,28 In addition,
in patients receiving PN with dextrose but no lipid, dextrose-
mediated insulin secretion has been found to suppress the
ILEs and EFAD
mobilization of EFAs from adipose tissue (depending on SO-Based ILEs
the amount of dextrose given).6,10 After the risk of EFAD
in patients receiving prolonged fat-free PN was recognized, Most of the early ILEs included in PN to provide a source of
intravenous lipid emulsions (ILEs; also referred to as IVLEs EFAs consisted of 100% SO, for example, Intralipid (Baxter
or IVFEs [intravenous fat emulsions]) were added to PN Healthcare Corporation, Deerfield, IL, USA; Table 1), Nu-
formulations to provide a source of LA and ALA.28-30 trilipid (B. Braun, Bethlehem, PA, USA), and Lipovenoes
ILEs also provided a nonglucose source of energy, thereby (Fresenius Kabi, Bad Homburg, Germany).1,30,33,38 SO-
reducing the need for high glucose infusion rates.28 ILE- based ILEs contain a large amount of LA and a moderate
containing formulations have been found to treat21,24,26,27 amount of ALA (Table 2).1 Although SO ILE has been
and prevent27,31,32 EFAD in adult, infant, and pediatric used successfully to deliver lipid via PN, concerns have been
patients receiving PN. Over the years, ILEs have become raised that this type of ILE, being rich in the n-6 fatty
a crucial component of PN, and they have evolved with acid LA, promotes the production of proinflammatory n-
respect to lipid type and content.3,30,33 6-derived eicosanoids, resulting in increased oxidative stress
The advent of novel ILEs, which have variable concentra- and systemic inflammation.1,3,5,28 In addition, SO-based
tions of EFAs, provides an opportunity to review the con- ILEs have been associated with the development of intesti-
dition of EFAD, including highlighting the circumstances nal failure–associated liver disease (IFALD), particularly in
in which it might occur in patients receiving PN despite pediatric patients.19 However, multiple components of the
the availability of ILEs and discussing the considerations SO ILE have been implicated, including not only the high
associated with the choice of ILE. The aim of this narrative ratio of n-6 to n-3 fatty acids but also the high phytosterol
review (based on a search of the literature between 1974 and levels and low vitamin E content compared with other types
September 2017) is to discuss the laboratory and clinical of ILE.19 As a result, in subsequent ILE formulations,
signs and symptoms, diagnosis, and treatment of EFAD. the SO has been partially replaced with lipid sources that
In addition, this review will discuss the role of ILEs in the provide less bioactive fatty acids, thereby decreasing the
treatment, prevention, and diagnosis of EFAD in patients amount of proinflammatory EFAs.1 These ILEs are widely
receiving PN. With the introduction of ILEs with different available in Europe and Canada but less so in the United
compositions, it is critical to understand the dosing of ILEs States.3,30,35
and how their use affects EFA status.
Medium-Chain Triglyceride–Based Intravenous
Laboratory and Clinical Signs and Symptoms Emulsions
of EFAD Medium-chain triglyceride (MCT)–based ILEs (for exam-
Biochemical evidence of EFAD appears earlier than the ple, Lipofundin-MCT [B. Braun, Melsungen, Germany]
clinical signs and symptoms.9,26,34 Diagnostic biochemi- and Lipovenoes MCT [Fresenius Kabi]) consist of mixtures
cal manifestations of EFAD include decreased plasma of SO and MCTs derived from coconut oils and other
levels of LA and ALA, and an elevated triene:tetraene tropical nut oils.1,30,33 MCTs are saturated fatty acids with
(T:T) ratio (the significance of this ratio is described in hydrocarbon chains of 6–10 carbon atoms esterified to
4 Journal of Parenteral and Enteral Nutrition 00(0)

Table 1. Indications of Commercially Available ILEs in the United States and/or Europe.a

ILE Indication References

Intralipid Source of energy and EFAs for patients requiring Intralipid [prescribing information]: Deerfield, IL,
PN for extended periods of time (usually >5 USA: Baxter Healthcare Corporation, 2015.
days) and a source of EFAs for prevention of http://ecatalog.baxter.com/ecatalog/loadResour
EFAD ce.blob?bid=20000307
For use as part of a balanced intravenous feeding Intralipid [summary of product characteristics]:
regimen in patients who are unable to receive Runcorn, Cheshire, UK: Fresenius Kabi
sufficient amounts of nutrients enterally Limited, 2018. http://www.mhra.gov.uk/home/
groups/spcpil/documents/spcpil/con1539317317
991.pdf
Lipofundin-MCTb Source of energy and EFAs for patients requiring Lipofundin-MCT [summary of product
PN characteristics]: Melsungen, Germany: B. Braun
Melsungen AG, 2018. http://www.mhra.gov.uk/
home/groups/spcpil/documents/spcpil/con15405
26906305.pdf
ClinOleic/ClinoLipid Source of energy and EFAs for PN in adults when Clinolipid [prescribing information]: Deerfield, IL,
oral or enteral nutrition is not possible, USA: Baxter Healthcare Corporation, 2013.
insufficient, or contraindicated https://www.accessdata.fda.gov/drugsatfda_docs
/label/2013/204508s000lbl.pdf
Source of energy and EFAs for patients requiring ClinOleic [summary of product characteristics]:
PN Thetford, Norfolk, UK: Baxter Healthcare Ltd,
2015. https://ecatalog.baxter.com/ecatalog/
loadResource.blob?bid=64438
Omegaven Source of energy and fatty acids in pediatric Omegaven [prescribing information]: Fresenius
patients with PNAC with direct or conjugated Kabi USA, 2018. https://www.accessdata.fda.
bilirubin levels ࣙ2 mg/dL gov/drugsatfda_docs/label/2018/0210589s000lbl
edt.pdf
PN supplementation with long-chain n-3-fatty Omegaven [summary of product characteristics]:
acids, especially EPA and DHA, when oral or Bad Homburg, Germany: Fresenius AG, 2015:
enteral nutrition is impossible, insufficient, or http://www.mhra.gov.uk/home/groups/spcpil/do
contraindicatedc cuments/spcpil/con1535688413229.pdf
SMOFlipid Source of energy and EFAs for PN in adults when SMOFlipid [prescribing information]: Fresenius
oral or enteral nutrition is not possible, Kabi, 2016. https://www.accessdata.fda.gov/
insufficient, or contraindicated drugsatfda_docs/label/2016/207648lbl.pdf
Source of energy and EFAs and n-3 fatty acids for SMOFlipid [summary of product characteristics]:
PN in patients when oral or enteral nutrition is Runcorn, Cheshire, UK: Fresenius Kabi
impossible, insufficient, or contraindicated Limited, 2019. http://www.mhra.gov.uk/home/
groups/spcpil/documents/spcpil/con1548997873
854.pdf
Lipidem/Lipoplusb Source of energy and essential n-6 fatty acids and Lipidem/Lipoplus [prescribing information]:
n-3 fatty acids as a component of a PN regimen B. Braun, Melsungen, Germany. https://www.
for adults bbraun.com.vn/content/dam/catalog/bb
raun/bbraunProductCatalog/S/AEM2015/en-vn
/b/lipoplus.pdf.bb-.41648313/lipoplus.pdf

DHA, docosahexaenoic acid; EFA, essential fatty acid; EFAD, essential fatty acid deficiency; EPA, eicosapentaenoic acid; ILE, intravenous lipid
emulsion; MCT, medium-chain triglyceride; PN, parenteral nutrition; PNAC, parenteral nutrition–associated cholestasis.
a Note that this list is not exhaustive, and individuals should consult the prescribing information in their country for the appropriate use of these

products.
b Not approved/available in the United States.
c Note that the European Summary of Product Characteristics states that Omegaven should be administered simultaneously with other fat

emulsions.

the glycerol backbone.6 MCTs do not contribute to the Olive Oil–Based ILEs
synthesis of eicosanoids and thus provide a source of fat
energy without additional systemic effects.3 However, as Olive oil (OO) is rich in oleic acid, an n-9 monounsaturated
MCTs do not provide EFAs, they are unsuitable for use as a fatty acid that is considered immune neutral.30,32,33 This
sole source of fat.30 type of ILE consists of 20% SO and 80% OO and has
Gramlich et al 5

Table 2. Oil Sources and Fatty Acid Composition of Commercially Available ILEs.a

Component Lipofundin- ClinOleic/ Lipidem/


(% by weight) Intralipid MCT ClinoLipid Omegaven SMOFlipid Lipoplus

Oil source
Soybean oil 100 50 20 0 30 40
Coconut oil 0 50 0 0 30 50
Olive oil 0 0 80 0 25 0
Fish oil 0 0 0 100 15 10
Fatty acid
LA 44–62 24–29 14–22 1.5c 14–25 24–29
ALA 4–11 2.5–5.5 0.5–4.2 1.1c 1.5–3.5 2.5–5.5
Oleic acid 19–30 11b 44–80 4–11 23–35 8b
AA 0 0 0 0.2–2 0 0
DHA 0 0 0 14–27 1–3.5 4.3–8.6d
EPA 0 0 0 13–26 1–3.5

AA, arachidonic acid; ALA, α-linolenic acid; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; ILE, intravenous lipid emulsion; LA,
linoleic acid; MCT, medium-chain triglyceride.
a From the current prescribing information or summary of product characteristics for each product (Table 1), except where otherwise indicated.

Note that the percentages of fatty acids in these ILEs are usually reported as a range rather than an absolute value.
b From Calder et al.28
c Mean value.
d Range for DHA and EPA combined.

approximately one-third of the LA of ILEs containing based ILE Omegaven (Fresenius Kabi; Table 1) contains
100% SO (eg, 19% vs 53% for ClinOleic [Baxter Healthcare much less LA (4.5% vs 53%) and ALA (1.8% vs 8%)
Corporation; Table 1] vs Intralipid; Table 2).30,32 Investiga- than the 100% SO-based ILE Intralipid, but it contains
tion of the anti-inflammatory effects of 3 types of ILE using their downstream metabolites, namely AA, DHA, and EPA
a number of in vivo (murine models of inflammation) and (Table 2).35 Following administration of a 100% FO ILE
in vitro (endothelial adhesion assays) techniques showed to a patient who had developed EFAD (elevated T:T ratio,
the anti-inflammatory superiority of ClinOleic to that of skin rash) because a severe soy allergy precluded the use
an SO/MCT (50:50) ILE and a mixed ILE containing SO, of SO-based ILE, the patient’s T:T ratio improved, and
MCT, OO, and fish oil (FO).39 the rash disappeared.40 Other reports support the notion
The development of ILEs containing lower levels of LA that a 100% FO-based ILE may be an effective alternative
and ALA raised concerns about their ability to prevent ILE for the treatment and prevention of EFAD.41-43 Murine
EFAD.34 However, although the LA content of OO-based studies suggest that, although the parent fatty acids LA and
ILE is lower than that of SO-based ILE, studies have shown ALA have historically been considered EFAs, provision of
that this does not reduce the potential of OO-based ILE their metabolites AA and DHA may also be sufficient to
to prevent EFAD.31,32 Vahedi et al31 found no biochemical prevent EFAD.34 However, the manufacturer of the 100%
evidence (based on T:T ratio >0.2) of EFAD in 13 patients FO-based ILE does not recommend its use as the sole source
with intestinal failure receiving OO-based ILE or SO-based of fat energy because of concerns that the low levels of
ILE in a 3-month, double-blind, randomized study. Olthof LA and ALA could lead to EFAD43 (see Table 1 for its
et al32 found no biochemical (T:T ratio >0.2) or clinical indications). As a result, FO ILE may be administered in
(skin rash) evidence of EFAD in 30 patients with intestinal conjunction with an SO ILE or as a component of a mixed
failure receiving long-term (>3 months) home PN (HPN) ILE formulation. For example, SMOFlipid (Fresenius Kabi;
with OO-based ILE. Appropriate dosing of the ILE to meet Table 1) contains 30% SO, 30% MCT, 25% OO, and 15%
LA and ALA requirements will avoid EFAD in patients FO (Table 2), and Lipidem/Lipoplus (B. Braun; Table 1)
receiving an OO-based ILE. contains 50% MCT, 40% SO, and 10% FO (Table 2).3,28,30

FO-Based ILEs Comparison of ILEs


FO is considered anti-inflammatory because it contains Available evidence suggests that mixed ILEs, which combine
a higher proportion of n-3 than n-6 fatty acids, with n- MCT, OO, and/or FO, offer some advantages over the use
3-derived eicosanoids being considered anti-inflammatory of SO alone. A recent systematic review assessed the effects
compared with n-6-derived eicosanoids.5 The 100% FO- of currently available ILEs in adult patients receiving HPN
6 Journal of Parenteral and Enteral Nutrition 00(0)

Table 3. Doses of Intravenous Lipid Emulsion Required to Meet Essential Fatty Acid Requirements in Adults.

Parameter Intralipid 20% Lipofundin-MCT 20% ClinOleic 20% SMOFlipid 20%

kcal/L 2000 1908 2000 2000


kcal/mL 2 1.9 2 2
% LAa 53 29 19 20
kcal from LA, kcal/mL 1.06 0.55 0.38 0.40
Volume required to obtain 75.5 144.6 210.5 200.0
80 kcal from LA, mL

LA, linoleic acid; MCT, medium-chain triglyceride.


a From Calder et al.28

and found that mixed ILEs may exert long-term clinical of dosing of 4 different types of ILE to provide sufficient
benefit as indicated by improved fatty acid profiles and LA to a term infant weighing 3 kg (0.1 g/kg/d)48 and a
antioxidant status.44 However, only 3 studies met the criteria preterm infant weighing 1.5 kg (0.25 g/kg/d).48 Tables 3
for the systematic review, and no randomized controlled and 4 show the minimum volumes of ILE to prevent
trials comparing mixed ILEs were identified. The authors EFAD, rather than total lipid dosing. The ideal ILE dose
therefore recommend prospective studies over a number of in relation to energy needs is not known. For example,
years to give a better indication of the long-term effects the amount of ILE required to provide minimal amounts
of the different ILEs.44 A systematic review conducted to of EFAs in infants and children may not be sufficient to
assess the safety and efficacy of mixed ILEs compared provide the recommended lipid doses (up to 4 g/kg/d in
with 100% SO-based ILEs in preterm infants found no term and preterm infants and up to 3 g/kg/d in children).48
statistically significant differences in clinically important Lipid provision in children varies depending on the clinical
safety outcomes and insufficient evidence to recommend situation and is generally somewhere in the range of 25%–
any mixed ILE over a 100% SO-based ILE or vice versa 50% of nonprotein energy, depending on age, growth, and
in preterm infants.45 The authors of this analysis also developmental stage.48
recommended larger randomized studies to evaluate the
effectiveness of mixed ILEs compared with 100% SO-
based ILEs in preterm infants.45 Further investigation is
Diagnosis of EFAD
important because both SO-based and FO-containing ILEs Diagnostic Testing
have been found to affect DHA levels in preterm infants.
Within the first postnatal week, there is a decline in DHA An elevated T:T ratio is considered biochemical evidence
and AA and an excess of LA in preterm infants receiving the of EFAD, in which a triene is a hydrocarbon with 3
100% SO-based ILE Intralipid; prolonged use contributes double bonds between carbon atoms and a tetraene is a
to a prolonged lower DHA status, even after establishment hydrocarbon with 4 double bonds between carbon atoms.
of full enteral feedings.46 Although FO-containing ILEs Under normal circumstances, desaturases (which insert
raise DHA and EPA levels, the early postnatal deficit in double bonds between adjacent carbon atoms) and elon-
DHA is not eliminated and there is a decline in AA, even gases (which lengthen hydrocarbon chains) act on LA to
greater than that observed with Intralipid.46 Thus, there is produce AA and on ALA to produce DHA and EPA
concern that current ILEs do not provide adequate amounts (Figure 1).1,3,8 In the absence of adequate LA and ALA,
of DHA essential for neurodevelopment in preterm infants. these enzymes instead act on the nonessential n-9 fatty acid
oleic acid (18:1n-9), resulting in increased production of
Mead acid (20:3n-9, a triene; Figure 1), which, in combina-
Dosing of ILEs tion with the reduced production of AA (a tetraene) from
An LA intake of 2%–4% of total energy, and an ALA LA, leads to an elevated T:T ratio.3,8,9
intake of 0.25%–0.5% of total energy, are recommended to The use of different methods to extract and quantify
prevent EFAD.6,7,47 As it is the SO component of the ILE fatty acids has led to a number of different threshold
that provides clinically significant amounts of LA and ALA, values for the T:T ratio being proposed to indicate EFAD
the amount of ILE required to provide sufficient energy (Table 5).42,49-53 Based on the polyunsaturated fatty acid
from LA and ALA depends on its SO content, which varies patterns of heart tissue, erythrocytes, and plasma from
between the different types of ILE (Table 2). Table 3 shows rats that developed severe symptoms of EFAD after being
examples of dosing of 4 different types of ILE to provide fed a fat-free diet, Holman49 suggested that a plasma T:T
4% of total energy as LA to an adult weighing 80 kg, based ratio of <0.4 indicated that the minimum dietary require-
on the LA content of the ILE. Table 4 shows examples ment for LA has been met. Subsequently, Holman et al,50
Gramlich et al 7

Table 4. Doses of ILE Required to Meet Essential Fatty Acid Requirements in Term and Preterm Infants.

LA per Day
Total Energy Required to Lipofundin-MCT SMOFlipid
Weight per Day Prevent EFAD Intralipid 20% 20% ClinOleic 20% 20%

Term infant
3 kg 300 kcal 0.3 g 2.83 mL 5.17 mL 7.89 mL 7.50 mL
(100 kcal/kg) (0.1 g/kga )
Preterm infant
1.5 kg 180 kcal 0.375 g 3.54 mL 6.47 mL 9.87 mL 9.38 mL
(120 kcal/kg) (0.25 g/kgb )

EFAD, essential fatty acid deficiency; ESPGHAN, European Society of Paediatric Gastroenterology, Hepatology and Nutrition; ILE,
intravenous lipid emulsion; LA, linoleic acid; MCT, medium-chain triglyceride.
a ESPGHAN guidelines recommend that term infants receive 0.1 g/kg/d of LA to prevent EFAD.48 Note that the ESPGHAN guidelines also

recommend that children receive 0.1 g/kg/d of LA to prevent EFAD. Therefore, the ILE dosing for children can be determined in a similar manner
as for term infants, based on the weight of the child.
b ESPGHAN guidelines recommend that preterm infants receive 0.25 g/kg/d of LA to prevent EFAD.48

using packed-column gas-liquid chromatography to mea- packed columns. The improved measurement of 1 compo-
sure fatty acids in the plasma from individuals unlikely nent of the T:T ratio (but not both) led to the lower T:T
to have derangements in EFA metabolism, suggested that ratios observed with the newer methods. It should also be
a plasma T:T ratio of 0.2 is the upper limit of normal noted that the more recent assays provide reference ranges
with respect to EFA status. Siguel et al,51 using capillary- for the T:T ratio rather than a threshold value indicative
column gas-liquid chromatography to measure fatty acids in of EFAD. Reference ranges represent values typically seen
plasma from reference subjects and patients with intestinal in a given population and depend on the fatty acid dietary
fat malabsorption and suspected EFAD, suggested that intake of that population. Thus, when evaluating whether
a plasma T:T ratio of >0.025 is indicative of EFAD. a T:T ratio is indicative of EFAD, the method used to
Lagerstadt et al,52 using a capillary gas chromatography- measure fatty acids and the reference range established for
electron-capture negative-ion mass spectrometry method to that method need to be taken into account.
measure fatty acids in plasma from healthy adult donors
or pediatric patients with no apparent metabolic/nutrition
abnormalities, established reference ranges for the T:T ratio
Interpreting Fatty Acid Profiles
for 4 age groups (Table 5), with a reference range of 0.010– Although the T:T ratio is diagnostic of EFAD, it does not
0.038 established for adults. These reference ranges for the take into account the n-3 fatty acid status19 and may be
T:T ratio are currently used by the Mayo Medical Labora- affected by the fatty acid profile of the ILE the patient
tories at the Mayo Clinic (Mayo Clinic Fatty Acid Profile, is receiving. Vahedi et al31 found a significant increase in
Essential, Serum).54 More recently, Kish-Trier et al,53 using oleic acid content in plasma phospholipids in patients re-
a gas chromatography-negative chemical ionization-mass ceiving OO-based ILE, which is rich in oleic acid, compared
spectrometry method modified from that of Lagerstadt et with patients receiving SO-based ILE after 3 months of
al52 to measure fatty acids in plasma/serum samples that treatment. Importantly, oleic acid is metabolized into Mead
were normal on routine biochemical genetic tests or col- acid (Figure 1); thus, the T:T ratio would be expected to
lected from healthy children and adults, reported reference be higher in these patients and may potentially exceed the
ranges for the T:T ratio for 3 age groups (Table 5), with a normal range, leading to an erroneous diagnosis of EFAD.
reference range of 0.004–0.051 established for adults. Similarly, de Meijer et al43 found that patients who were
It should be noted that the newer methods of mea- receiving SO-based ILE at baseline had fatty acid profiles
suring fatty acids described above51-53 established thresh- reflective of the 100% SO-based ILE, that is, high levels
olds/reference ranges for the T:T ratio that are an order of of LA and AA and low levels of EPA and DHA. When
magnitude lower than the previously reported threshold of these patients were switched to 100% FO ILE, their fatty
0.2.50 Siguel et al51 ascribed the differences between a T:T acid profiles gradually changed over the course of 6 weeks,
ratio of >0.025 indicating EFAD as determined by their with decreasing levels of LA and AA and increasing levels
assay (capillary-column gas-liquid chromatography) and a of EPA and DHA. In this situation, the T:T ratio would also
T:T ratio of >0.2 indicating EFAD as determined by Hol- be expected to be higher as the tetraene levels are reduced.
man et al50 (packed-column gas-liquid chromatography) to Gramlich et al8 evaluated the fatty acid profiles (obtained
capillary columns being able to separate and quantify small from the Mayo Clinic) of 3 patients receiving PN with
quantities of Mead acid (a triene) more accurately than ILE and reported the patients’ T:T ratios, LA, ALA, and
8 Journal of Parenteral and Enteral Nutrition 00(0)

Table 5. Fatty Acid Assays and Their Associated T:T Ratio Threshold/Reference Range.

Study Subjects/Diagnosis Assay Threshold/Reference Range

Holman 196049 Rats with severe symptoms of Fatty acids measured in heart A plasma T:T ratio <0.4
EFAD after being fed a tissue, plasma, and indicates that the minimum
fat-free diet erythrocytes by alkaline dietary requirement for LA
isomerization of the has been met
ethanol-ether extract
Holman et al 197950 Hospital patients without a Fatty acids measured in plasma A plasma T:T ratio of 0.2 is the
diagnosis of a disease in by packed-column gas-liquid upper limit of normalcy with
which EFA metabolism may chromatography respect to EFA status
be deranged plus 14 healthy
adult donors
Approximately 10
individuals of each sex and
aged from each decade
between 0 and 90 years
Siguel et al 198751 56 reference subjects and Fatty acids measured in plasma A plasma T:T ratio of >0.025
10 patients with intestinal fat by capillary-column is indicative of EFAD
malabsorption and gas-liquid chromatography
suspected EFAD
Lagerstadt et al 196 plasma specimens from Fatty acids measured in plasma Reference ranges established
200152 healthy adult donors or by capillary gas for the T:T ratio:
pediatric patients with no chromatography-electron- <1 month: 0.017–0.083
apparent metabolic or capture negative-ion mass 1–12 months: 0.013–0.050
nutrition abnormalities spectrometry 1–17 years: 0.013–0.050
>17 years: 0.010–0.038
Kish-Trier et al 306 plasma/serum samples Fatty acids measured in Reference ranges established
201653 found to be normal on plasma/serum by capillary for the T:T ratio:
routine biochemical genetic gas <1 month: 0.006–0.052
tests or collected from chromatography-negative 1 month to ࣘ12 months:
healthy children and adults chemical ionization-mass 0.002–0.046
spectrometry (modified from >1 year: 0.004–0.051
the method of Lagerstadt et
al 200152 )

EFA, essential fatty acid; EFAD, essential fatty acid deficiency; LA, linoleic acid; T:T, triene:tetraene.

Mead acid levels plus the oleic acid and AA levels. Each in conjunction with reduced AA levels.5,9,24,55 A correct
of the patients was receiving OO-based ILE at the time of diagnosis of EFAD requires that the T:T ratio is elevated
obtaining the fatty acid profile, with patients 1 and 2 having because LA levels are low enough to result in an increased
switched from SO-based ILE. Patient 1 had low LA levels production of Mead acid as well as a decreased production
and a T:T ratio (0.032) within the Mayo Clinic reference of AA, that is, both the numerator (triene/Mead acid) is high
range (0.010–0.038); patient 2 had LA levels within the and the denominator (tetraene/AA) is low. Changes in the
reference range, elevated Mead acid and oleic acid levels, numerator only or the denominator only are most likely to
and an elevated T:T ratio (0.042); and patient 3 had low reflect dietary changes.
LA levels, elevated Mead acid and oleic acid levels, and It has therefore been suggested that the fatty acid profile
an elevated T:T ratio (0.051).8 Based on the elevated T:T as a whole should be considered when assessing whether a
ratios according to the assay reference range, it might be patient has EFAD, especially in patients receiving PN with
concluded that patients 2 and 3 had biochemical evidence one of the mixed ILEs.8,34,53 Kish-Trier et al53 recommended
of EFAD. However, the authors suggested that the low LA that the levels of individual n-6 and n-3 fatty acids be taken
levels and/or elevated Mead acid levels observed in these into account as well as the T:T ratio when assessing EFAD,
2 patients may be reflective of the composition of the OO- with a pattern of increased T:T ratio, decreased levels of
based ILE (ie, lower LA and significantly more oleic acid LA and ALA, and increased levels of Mead acid being
than SO-based ILE).8 In support of this interpretation, indicative of EFAD.
all 3 patients had AA levels within the reference range, Although the fatty acid profile is valuable in assessing
whereas in EFAD, elevated Mead acid levels are observed whether a patient has EFAD, we recommend that patients
Gramlich et al 9

are not routinely tested for fatty acid status when receiving EFAD who were receiving fat-free PN alleviated their scaly
an enteral or oral diet or adequate ILE. The risk of clinically skin rash but did not correct their abnormal T:T ratios.26
significant EFAD is extremely remote in patients receiving One study reported that topical application of sunflower oil
PN who have adequate fat stores and are receiving PN for a partly corrected EFAD in 1 infant and prevented EFAD
relatively brief period of time. In addition, the risk of EFAD in another infant.27 However, other studies have found
is very low in patients receiving PN who are on an enteral no effect of topical applications of oils or LA on EFAD
or oral diet. On the other hand, we recommend screening symptoms in infant or adult patients.9
for EFAD in selected patients who have depleted fat stores
or who are receiving fat-free PN for >4 weeks, including Ongoing Knowledge Gaps
HPN patients. We also recommend screening for EFAD
Several knowledge gaps exist, and numerous research ques-
in patients (particularly children) receiving fat-free PN or
tions need to be addressed. There is a need to determine
inadequate ILE, intravenous SO lipid–restricted diets (<1
the contemporary incidence of EFAD in the setting of the
g/kg/d of SO), or doses of alternative ILEs less than the
newer ILEs. Additionally, clarity regarding whether at-risk
recommended dose for age (eg, a preterm infant receiving
patients should be screened for EFAD, as well as the cri-
1.5 g/kg/d of SMOFlipid or a child with IFALD receiving
teria for determining the appropriate timing for when such
0.5 g/kg/d of FO monotherapy). On the other hand, we note
screening should occur, is needed. Currently unanswered
that, for example, a preterm infant receiving 1.5 g/kg/d of
questions in the setting of the newer ILEs include how
SMOFlipid is not necessarily at risk of EFAD if enteral
quickly EFAD occurs and for how long does the body need
feeding is also provided or this dosage is used in the short
to be depleted of EFAs? Is there a safe period for how
term only. Ultimately, there are no set criteria for screening
long enteral feeding can be withheld before patients develop
for EFAD. Preterm infants receiving limited ILE dosing
EFAD, and does short-term EFA depletion have short-
may be at a higher risk of EFAD and need more frequent
term or long-term consequences? This last question may
monitoring until enteral nutrition is established. Adults,
be very important in the preterm and infant population,
however, may need less frequent monitoring as they have
as long-term consequences of depletion of AA and DHA
greater fat stores, unless they are malnourished, in which
(“conditionally essential” EFAs) have been reported,17,57
case the risk of EFAD is increased. These considerations
raising concerns about the potential impact of depletion of
are negated by enteral or oral nutrition, and therefore the
LA or ALA. Lastly, the key question yet to be addressed is
decision of when to screen for EFAD requires clinical
whether the current cutoffs for the T:T ratio remain relevant
judgment.
in the setting of the newer ILEs.

Treatment of EFAD Conclusions


As described above, patients with EFAD may be treated Although EFAD is uncommon in the general population,
with PN formulations containing LA and ALA. For PN- certain populations of patients are at risk of developing
dependent patients who cannot receive ILE (for example, EFAD, including patients receiving PN. The inclusion of
patients with severe hypertriglyceridemia or an allergy to ILEs in PN formulations, as a source of EFAs, was found
ILE), oral and topical preparations containing EFAs may be to prevent EFAD in infant and adult patients. An elevated
treatment options. There is some evidence that oral prepa- T:T ratio has traditionally been considered biochemical
rations containing EFAs may treat EFAD. Richardson and evidence of EFAD. As the assays for measuring fatty acids
Sgoutas23 reported EFAD in 4 patients receiving fat-free have improved over the years, the particular assay used and
PN; 2 patients received supplementation with oral LA in the its reference range need to be kept in mind when assessing
form of safflower oil, which reversed the EFAD. Unless a whether a patient has EFAD, as well as when evaluating the
patient’s intestinal absorption is completely impaired, oral T:T ratios published in the literature. The fatty acid profile
EFA preparations would be expected to be of some benefit as a whole may provide additional information, especially
in treating EFAD. However, many HPN patients with short in patients receiving newer ILE formulations, as the fatty
bowel syndrome have some intestinal absorptive capacity acid composition, including the proportion of EFAs, varies
but cannot sustain themselves solely with enteral energy and between the different types of ILE. The optimal balance
hence require delivery of nutrients, including lipids, via PN. of fatty acids in ILEs that delivers clinical benefit while
Studies of topical application of oils containing EFAs reducing adverse effects remains to be determined.
have reported mixed results. One study reported that topical Diagnosis of EFAD is multifactorial, and abnormal lab-
application of sunflower seed oil in 2 infants receiving fat- oratory values can occur in the absence of clinical symptoms
free PN resulted in rapid reversal of both the biochemical and physical findings. Therefore, in patients receiving ILE,
and clinical manifestation of EFAD.56 One study reported the following factors need to be considered when interpret-
that topical application of safflower oil in 3 infants with ing the fatty acid profile (T:T) and suggesting the diagnosis
10 Journal of Parenteral and Enteral Nutrition 00(0)

of EFAD: (1) the fatty acid composition of the ILE, (2) the 15. Salama GS, Kaabneh MA, Almasaeed MN, Alquran M. Intra-
fatty acid profile levels of the downstream metabolites of venous lipids for preterm infants: a review. Clin Med Insights Pediatr.
2015;9:25-36.
LA and ALA (AA, DHA, and EPA) as well as the levels
16. Larque E, Demmelmair H, Koletzko B. Perinatal supply and
of LA and ALA themselves; (3) the patient’s oral intake metabolism of long-chain polyunsaturated fatty acids: importance for
and absorption of fatty acids, and (4) the patient’s fatty acid the early development of the nervous system. Ann N Y Acad Sci.
stores. 2002;967:299-310.
17. Carlson SE, Colombo J. Docosahexaenoic acid and arachidonic acid
nutrition in early development. Adv Pediatr. 2016;63(1):453-471.
Statement of Authorship 18. Hadley KB, Ryan AS, Forsyth S, Gautier S, Salem N, Jr. The essential-
L. Gramlich, C. Ireton-Jones, J. M. Miles, M. Morrison, and ity of arachidonic acid in infant development. Nutrients. 2016;8(4):216.
A. Pontes-Arruda contributed to the conception and design 19. Gura KM, Crowley M. A detailed guide to lipid therapy in intestinal
failure. Semin Pediatr Surg. 2018;27(4):242-255.
of the research; M. Morrison contributed to the acquisition
20. Worthington P, Balint J, Bechtold M, et al. When is parenteral nutrition
and analysis of the data; L. Gramlich, C. Ireton-Jones, J. M.
appropriate? JPEN J Parenter Enteral Nutr. 2017;41(3):324-377.
Miles, M. Morrison, and A. Pontes-Arruda contributed to the 21. Caldwell MD, Jonsson HT, Othersen HB, Jr. Essential fatty acid
interpretation of the data; and L. Gramlich, C. Ireton-Jones, deficiency in an infant receiving prolonged parenteral alimentation.
J. M. Miles, M. Morrison, and A. Pontes-Arruda contributed J Pediatr. 1972;81(5):894-898.
to drafting the manuscript. All authors critically revised the 22. Paulsrud JR, Pensler L, Whitten CF, Stewart S, Holman RT. Essential
manuscript, agree to be fully accountable for ensuring the fatty acid deficiency in infants induced by fat-free intravenous feeding.
integrity and accuracy of the work, and read and approved the Am J Clin Nutr. 1972;25(9):897-904.
final manuscript. 23. Richardson TJ, Sgoutas D. Essential fatty acid deficiency in four adult
patients during total parenteral nutrition. Am J Clin Nutr. 1975;28(3):
258-263.
References 24. Riella MC, Broviac JW, Wells M, Scribner BH. Essential fatty acid
1. Wanten GJ, Calder PC. Immune modulation by parenteral lipid deficiency in human adults during total parenteral nutrition. Ann Intern
emulsions. Am J Clin Nutr. 2007;85(5):1171-1184. Med. 1975;83(6):786-789.
2. Calder PC, Grimble RF. Polyunsaturated fatty acids, inflammation and 25. Fleming CR, Smith LM, Hodges RE. Essential fatty acid defi-
immunity. Eur J Clin Nutr. 2002;56(Suppl 3):S14-S19. ciency in adults receiving total parenteral nutrition. Am J Clin Nutr.
3. Fell GL, Nandivada P, Gura KM, Puder M. Intravenous lipid emul- 1976;29(9):976-983.
sions in parenteral nutrition. Adv Nutr. 2015;6(5):600-610. 26. O’Neill JA, Jr., Caldwell MD, Meng HC. Essential fatty acid deficiency
4. Calder PC. Omega-3 polyunsaturated fatty acids and inflamma- in surgical patients. Ann Surg. 1977;185(5):535-542.
tory processes: nutrition or pharmacology? Br J Clin Pharmacol. 27. Postuma R, Pease PW, Watts R, Taylor S, McEvoy FA. Essential fatty
2013;75(3):645-662. acid deficiency in infants receiving parenteral nutrition. J Pediatr Surg.
5. Le HD, Meisel JA, de Meijer VE, Gura KM, Puder M. The essentiality 1978;13(4):393-398.
of arachidonic acid and docosahexaenoic acid. Prostaglandins Leukot 28. Calder PC, Jensen GL, Koletzko BV, Singer P, Wanten GJ. Lipid
Essent Fatty Acids. 2009;81(2-3):165-170. emulsions in parenteral nutrition of intensive care patients: current
6. Hise ME, Brown JC. Lipids. In: Gottschlich MM, ed. The A.S.P.E.N. thinking and future directions. Intensive Care Med. 2010;36(5):735-749.
Nutrition Support Core Curriculum: A Case-Based Approach – The 29. Pironi L, Arends J, Bozzetti F, et al. ESPEN guidelines on chronic
Adult Patient. Silver Spring, MD: A.S.P.E.N.; 2007:48-70. intestinal failure in adults. Clin Nutr. 2016;35(2):247-307.
7. Fats and fatty acids in human nutrition. Report of an expert consulta- 30. Raman M, Almutairdi A, Mulesa L, et al. Parenteral nutrition and
tion. FAO Food Nutr Pap. 2010;91:1-166. lipids. Nutrients. 2017;9(4):388.
8. Gramlich L, Meddings L, Alberda C, et al. Essential fatty acid 31. Vahedi K, Atlan P, Joly F, et al. A 3-month double-blind randomised
deficiency in 2015: the impact of novel intravenous lipid emulsions. study comparing an olive oil- with a soyabean oil-based intravenous
JPEN J Parenter Enteral Nutr. 2015;39(Suppl 1):61S-66S. lipid emulsion in home parenteral nutrition patients. Br J Nutr.
9. Mogensen KM. Nutrition Issues in Gastroenterology, series #164. 2005;94(6):909-916.
Essential fatty acid deficiency. Pract Gastroenterol. 2017;41(6):37-44. 32. Olthof ED, Roelofs HM, Fisk HL, Calder PC, Wanten GJ. No clinical
10. Mascioli EA, Smith MF, Trerice MS, Meng HC, Blackburn GL. or biochemical evidence for essential fatty acid deficiency in home pa-
Effect of total parenteral nutrition with cycling on essential fatty acid tients who depend on long-term mixed olive oil- and soybean oil-based
deficiency. JPEN J Parenter Enteral Nutr. 1979;3(3):171-173. parenteral nutrition. JPEN J Parenter Enteral Nutr. 2016;40(7):982-
11. Jeppesen PB, Christensen MS, Hoy CE, Mortensen PB. Essential fatty 988.
acid deficiency in patients with severe fat malabsorption. Am J Clin 33. Mundi MS, Salonen BR, Bonnes SL, Hurt RT. Nutrition Issues in
Nutr. 1997;65(3):837-843. Gastroenterology, series #166. Parenteral nutrition–lipid emulsions
12. Jeppesen PB, Hoy CE, Mortensen PB. Deficiencies of essential fatty and potential complications. Pract Gastroenterol. 2017;41(8):32-37.
acids, vitamin A and E and changes in plasma lipoproteins in patients 34. Anez-Bustillos L, Dao DT, Fell GL, et al. Redefining essential fatty
with reduced fat absorption or intestinal failure. Eur J Clin Nutr. acids in the era of novel intravenous lipid emulsions. Clin Nutr.
2000;54(8):632-642. 2018;37(3):784-789.
13. Maqbool A, Schall JI, Garcia-Espana JF, et al. Serum linoleic acid 35. Anez-Bustillos L, Dao DT, Baker MA, et al. Intravenous fat emulsion
status as a clinical indicator of essential fatty acid status in children formulations for the adult and pediatric patient: understanding the
with cystic fibrosis. J Pediatr Gastroenterol Nutr. 2008;47(5):635-644. differences. Nutr Clin Pract. 2016;31(5):596-609.
14. Cober MP, Killu G, Brattain A, et al. Intravenous fat emulsions 36. Jeppesen PB, Hoy CE, Mortensen PB. Essential fatty acid deficiency
reduction for patients with parenteral nutrition-associated liver disease. in patients receiving home parenteral nutrition. Am J Clin Nutr.
J Pediatr. 2012;160(3):421-427. 1998;68(1):126-133.
Gramlich et al 11

37. Prottey C, Hartop PJ, Press M. Correction of the cutaneous man- 47. Holman RT, Johnson SB, Hatch TF. A case of human linolenic
ifestations of essential fatty acid deficiency in man by application acid deficiency involving neurological abnormalities. Am J Clin Nutr.
of sunflower-seed oil to the skin. J Invest Dermatol. 1975;64(4): 1982;35(3):617-623.
228-234. 48. Lapillonne A, Fidler Mis N, Goulet O, et al. ESPGHAN/
38. Nutrilipid I.V. fat emulsion [prescribing information]. Bethlehem, ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition:
PA: B. Braun; 2014. Available at: https://www.bbraunusa.com/en/ lipids. Clin Nutr. 2018;37(6 Pt B):2324-2336.
products/b2/nutrilipid-20-soybeanoilivfatemulsion.html. Accessed July 49. Holman RT. The ratio of trienoic:tetraenoic acids in tissue lipids as a
26, 2018. measure of essential fatty acid requirement. J Nutr. 1960;70(3):405-410.
39. Buschmann K, Poeschl J, Braach N, et al. The olive oil-based lipid 50. Holman RT, Smythe L, Johnson S. Effect of sex and age on fatty acid
clinoleic blocks leukocyte recruitment and improves survival during composition of human serum lipids. Am J Clin Nutr. 1979;32(12):2390-
systemic inflammation: a comparative in vivo study of different par- 2399.
enteral lipid emulsions. Mediators Inflamm. 2015;757059. 51. Siguel EN, Chee KM, Gong JX, Schaefer EJ. Criteria for essential fatty
40. Gura KM, Parsons SK, Bechard LJ, et al. Use of a fish oil-based acid deficiency in plasma as assessed by capillary column gas-liquid
lipid emulsion to treat essential fatty acid deficiency in a soy al- chromatography. Clin Chem. 1987;33(10):1869-1873.
lergic patient receiving parenteral nutrition. Clin Nutr. 2005;24(5): 52. Lagerstedt SA, Hinrichs DR, Batt SM, et al. Quantitative determina-
839-847. tion of plasma c8-c26 total fatty acids for the biochemical diagnosis of
41. Gura K, Strijbosch R, Arnold S, McPherson C, Puder M. The role nutritional and metabolic disorders. Mol Genet Metab. 2001;73(1):38-
of an intravenous fat emulsion composed of fish oil in a parenteral 45.
nutrition-dependent patient with hypertriglyceridemia. Nutr Clin Pract. 53. Kish-Trier E, Schwarz EL, Pasquali M, Yuzyuk T. Quantitation of
2007;22(6):664-672. total fatty acids in plasma and serum by GC-NCI-MS. Clin Mass
42. Ernst KD. Essential fatty acid deficiency during parenteral soybean oil Spectrom. 2016;2:11-17.
lipid minimization. J Perinatol. 2017;37(6):695-697. 54. Mayo Clinic, Mayo Medical Laboratories. Fatty Acid Profile, Essen-
43. de Meijer VE, Le HD, Meisel JA, Gura KM, Puder M. Parenteral tial, Serum. Available at: https://www.mayomedicallaboratories.com/
fish oil as monotherapy prevents essential fatty acid deficiency in test-catalog/Clinical+and+Interpretive/82426. Accessed October 18,
parenteral nutrition-dependent patients. J Pediatr Gastroenterol Nutr. 2017.
2010;50(2):212-218. 55. Hamilton C, Austin T, Seidner DL. Essential fatty acid deficiency in
44. Jones CJ, Calder PC. Influence of different intravenous lipid emulsions human adults during parenteral nutrition. Nutr Clin Pract. 2006;21(4):
on fatty acid status and laboratory and clinical outcomes in adult 387-394.
patients receiving home parenteral nutrition: a systematic review. Clin 56. Friedman Z, Shochat SJ, Maisels MJ, Marks KH, Lamberth EL, Jr.
Nutr. 2018;37(1):285-291. Correction of essential fatty acid deficiency in newborn infants by
45. Kapoor V, Glover R, Malviya MN. Alternative lipid emulsions versus cutaneous application of sunflower-seed oil. Pediatrics. 1976;58(5):650-
pure soy oil based lipid emulsions for parenterally fed preterm infants. 654.
Cochrane Database Syst Rev. 2015;(12):CD009172. 57. Colombo J, Jill Shaddy D, Kerling EH, Gustafson KM, Carlson SE.
46. Robinson DT, Martin CR. Fatty acid requirements for the preterm Docosahexaenoic acid (DHA) and arachidonic acid (ARA) balance
infant. Semin Fetal Neonatal Med. 2017;22(1):8-14. in developmental outcomes. Prostaglandins Leukot Essent Fatty Acids.
2017;121:52-56.

You might also like