You are on page 1of 27

DOI: 10.1002/jpen.

2306

REVIEW

Parenteral nutrition compatibility and stability: A


comprehensive review

Joseph I. Boullata PharmD, RPh1 Jay M. Mirtallo MS, RPh2,3


Gordon S. Sacks PharmD4 Genene Salman PharmD5 Kathleen Gura PharmD6,7
Todd Canada PharmD8 Angela Maguire PharmD9 the ASPEN Parenteral Nutrition
Safety Committee2
1
Clinical Nutrition Support Services, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
2
American Society for Parenteral and Enteral Nutrition, Silver Spring, Maryland, USA
3
College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
4
Medical Affairs for PN Market Unit, Fresenius Kabi USA, LLC, Lake Zurich, Illinois, USA
5
Department of Pharmacy Practice, College of Pharmacy, Marshall B. Ketchum University, Fullerton, California, USA
6
Pharmacy Clinical Research Program, Boston Children’s Hospital, Boston, Massachusetts, USA
7
Harvard Medical School, Boston, Massachusetts, USA
8
University of Texas MD Anderson Cancer Center, Houston, Texas, USA
9
BJC HomeCare Infusions, Overland, Missouri, USA

Correspondence
Jay M. Mirtallo, 2921 Braumiller Rd, Delaware, Abstract
OH 43015, USA.
Email: jaym@nutritioncare.org
Several guidance documents support best practices across the stages of the parenteral
nutrition (PN) use process to optimize patient safety. The critical step of PN order ver-
ification and review by the pharmacist requires a contextual assessment of the com-
patibility and stability implications of the ordered PN prescription. This article will
provide working definitions, describe PN component characteristics, and present a
wide-ranging representation of compatibility and stability concerns that need to be
considered prior to preparing a PN admixture. This paper has been approved by the
American Society for Parenteral and Enteral Nutrition (ASPEN) Board of Directors.

KEYWORDS
compatibility, parenteral nutrition, stability

INTRODUCTION the first use of PN, pharmacists were called upon to design and pre-
pare safe formulations taking into account sterility, compatibility, and
Parenteral nutrition (PN) therapy continues to be a valued clinical stability.1 Numerous national guidance documents promote the safety
intervention for many patients across care settings. It is considered and efficacy of PN therapy.2–5 Among the guidance documents are
a high-alert medication, meaning that its use requires safety-focused those that specifically address the review of the PN prescription.3 In
policies, procedures, practices, and systems to limit patient risk. From this critical step of the PN use process, a pharmacist reviews each PN

© 2022 American Society for Parenteral and Enteral Nutrition

JPEN J Parenter
J Parenter Enteral
Enteral Nutr. 2022;46:273–299.
Nutr. 2022;1–27. wileyonlinelibrary.com/journal/jpen
wileyonlinelibrary.com/journal/jpen 273
1
274
2 BOULLATA ET AL .

prescription for completeness, clinical appropriateness, and formula- This requires the pharmacist reviewing their patient’s PN formulation
tion safety, communicating any concerns with the prescriber.6,7 This is to be knowledgeable in pharmaceutics, a responsibility already empha-
inherently understood to require the pharmacist to apply their knowl- sized in the literature.9–12
edge of pharmaceutical science and ensure the likelihood of a safe PN Chemical compounds (eg, drugs, nutrients) are formulated into
formulation. dosage forms by manufacturers to create a product to be administered
This comprehensive review fills an identified void on the topic with to patients. For intravenous (IV) administration, this requires sterile,
a deeper look behind isolated tables or lists of PN compatibility and low-particulate (within compendial limits) dosage forms formulated as
stability. The article aims to reinforce the pharmacist’s appreciation of solutions—usually aqueous, but they may often include some nonaque-
the complexity of PN formulation review by defining and then describ- ous solvents. The exception are those products formulated as oil-in-
ing the major compatibility and stability pitfalls that need to be rec- water emulsions (eg, lipid injectable emulsions [ILEs]). Physicochemi-
ognized to limit the risk of infusing a potentially unsafe PN. A phar- cal properties (eg, solubility, ionization constants) are accounted for in
macist specializing in nutrition support uses their knowledge, skill, and the formulation of each manufactured IV product. Each product may
experiences to verify the safety of PN regimens recommended or pre- include substances beyond the active ingredient(s), including excipi-
scribed by others. This article also serves as a resource for other clini- ents intended for a specific role (eg, to maintain sterility, solubility, sta-
cians involved in recommending or prescribing a specific PN regimen to bility). Combining individual IV drug products (ie, components) into a
better appreciate the pharmacist’s foundation and rationale. A follow- single preparation is referred to as an IV admixture. The pharmacy asep-
up article will provide practical steps to help clinicians recommend or tically prepares these IV admixtures (also known as compounded ster-
prescribe safe PN formulations with compatibility and stability in mind, ile preparations). The most complex such admixture, containing over 50
and a third article will describe methodology for evaluating PN compat- chemical components, is PN. Throughout this paper, the compounded
ibility and stability. sterile PN, as a preparation available for administration, is referred to
These recommendations do not constitute medical or other pro- as an admixture (ie, PN admixture) regardless of whether it contains
fessional advice and should not be taken as such. To the extent that ILEs (ie, 3-in-1, total nutrient admixture [TNA]) or not (ie, 2-in-1).
the information published herein may be used to assist in the care Inevitably, the admixture is less stable than its individual compo-
of patients, this is the result of the sole professional judgment of the nent products, with risk for incompatibilities. Risk for incompatibility
attending healthcare professional whose judgment is the primary com- and instability is ever-present and an issue long recognized with PN but
ponent of quality medical care. The information presented is not a sub- less well appreciated in recent years.4,13 Compatibility and stability are
stitute for the judgment by the healthcare professional. Circumstances at least as important as sterility in determining a beyond-use date for
in clinical settings and patient indications may require actions differ- PN admixtures, whether preparing the admixtures for immediate use
ent from those recommended in this document, and in those cases, in acute care or for a 7-day supply in the home care setting.
the judgment of the treating professional should prevail. The American
Society for Parenteral and Enteral Nutrition (ASPEN) does not endorse
any particular brand of products mentioned herein. This paper has been Compatibility
approved by the ASPEN Board of Directors.
Compatibility refers to the uneventful physical and chemical coexis-
tence of two or more components over time after being combined.14–16
Pharmacy science Combining substances at any point prior to or during administration
holds the potential for physical and chemical reactivity. The term incom-
The pharmaceutical sciences often refer to the broad, interdisciplinary patibility therefore indicates an interaction between two or more sub-
foundational sciences and the unique knowledge underpinning phar- stances (active ingredients, excipients, and/or materials [eg, packaging,
macy practice.8 This is not just a collection of science for its own end, containers, infusion sets]) over time in a specific environment. These
but one that ultimately supports the safe and effective use of medica- may be physical incompatibilities (eg, complexation or leaching or des-
tions. This knowledge allows both qualitative and quantitative appli- orption) or chemical incompatibilities. The latter involve reactants of
cation of scientific principles to patient care—for example, applying intermolecular and interionic forces as might be seen with the Mail-
known data on the physical properties and chemical properties of sub- lard reaction and calcium and phosphate insolubility. The manifestation
stances to best predict solubility, compatibility, stability, formulation of an incompatibility may be a change in color, pH, osmolality or vis-
design, and physiologic action, among others. Pharmaceutical science cosity, formation of gas, or yielding of a precipitate (whether visible or
disciplines include pharmaceutics, pharmacokinetics, and pharmaco- not) altering the admixture. The result may ultimately limit therapeutic
dynamics, which are each incorporated into the clinical care of the effect of one or more active components or increase the risk for toxicity
patient.8 It is the unique responsibility of the pharmacist in general to including that from the infusion of precipitates.
ensure that prepared medications dispensed for patient use are safe Incompatibility is generally preventable, often dependent on pro-
and effective. This obligation includes ensuring the integrity of any pre- portional concentrations of ingredients, requiring some knowledge
pared parenteral dosage form, not least being the compatibility and of chemical structures, solubilities, pKa values for ionizable ingre-
stability of PN—arguably the most complex drug in routine clinical use. dients, and the pH of each component to anticipate and avoid
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 275
3

incompatibilities. The potential incompatibilities may or may not Application to practice


already be defined in the literature but are less likely described for
multicomponent admixtures such as PN. If an incompatibility is already Whether a PN formulation is completely customized to a patient’s
reported, the pharmacist will need to be able to decipher the cause to macronutrient and micronutrient needs or begins with a standard-
prevent similar recurrence in formulating a PN admixture. Multiple and ized, commercially available PN product, variation of just one compo-
overlapping forces may affect a PN admixture. For example, the pH of nent has the potential to influence the final admixture. The complex-
the final admixture may influence the solubility as well as the stability ity of a PN admixture is quickly recognized when acknowledging the
of ingredients. Consider that a pH difference of 2 units represents a many ingredients that are eventually in the patient’s infusion container.
100-fold change, since the scale is logarithmic not linear. Each PN com- Newer products and updated compatibility and stability study meth-
ponent product is manufactured at a pH at which the active ingredient ods and data may even supplant older information. Furthermore, with
is most soluble and stable. The pH of each component can vary con- each new product introduced to the market and included as a PN com-
siderably from other component products and is a critical characteris- ponent, or as clinical practice evolves to better customize the PN to
tic that needs to be considered when combining components in a PN meet patient needs, recalibration of our understanding of compatibil-
admixture to avoid incompatibility and instability related to the overall ity and stability is needed. This has happened over time, whether it
pH of the final PN admixture. was the introduction of TNAs into practice,19,20 including the newer
ILE products,21 or evaluating new drugs administered by Y-site with
PN.22,23 The importance of compatibility and stability is heightened
Stability for the patients who will receive a 7-day supply of PN admixtures at
home.24 Incompatibility or instability of an admixture is seldom obvi-
The term stability refers to the maintenance of the chemical integrity ous to the unaided eye but can be determined experimentally using val-
of each active ingredient or the physical integrity of the dosage idated methods or can be predicted based on known physicochemical
form/system over time in a given environment.14–16 Instability there- properties of all the components. Unfortunately, significant harm and
fore indicates the irreversible decomposition/degradation of active fatality can occur when available data are not taken into account or
ingredients or the dosage form/system as a result of pH, tempera- pharmaceutics is not considered.25,26 Pharmacists without the requi-
ture, light, oxygen, solvents, and/or reactants. These may be physical site knowledge or not applying principles do so at their own peril and
(eg, crystallization, adsorption, broken emulsion) or chemical (eg, place their patients at risk.
hydrolysis, oxidation). Oxidation reactions may occur in the presence
of oxygen (auto-oxidation) or additionally in the presence of light
(photo-oxidation). The environment in which PN preparation—as well PN COMPONENTS AND RELEVANT PROPERTIES
as storage and administration—takes place involves exposure to air
and light. Instability may manifest as increased particulate matter, a Numerous product components are used to prepare a PN admix-
visible haze, visible oil, or other color change but, in most cases, may ture. As each component has unique physicochemical properties, a
only be identified using appropriate methodology. The rates of degra- summary of these will be presented. Several aspects relevant to
dation or reaction vary with the reactants and the environment. The compatibility and stability will be noted below prior to the discus-
ultimate result is that instability can take away from biological activity sion of compatibility and stability in PN admixtures in subsequent
of the degraded component or generate risk from toxic by-products or sections.
particulate matter, including large lipid droplets.
The physical instability of ILE refers to increases in lipid droplet
size. However, chemical instability of ILE may include peroxide forma- Macronutrients
tion and pH change without necessarily affecting physical stability.17
Combining substances at any point prior to intravascular infusion holds Amino acids
the potential to alter chemical structure of a substance or the phys-
ical state of the admixture. For example, ILE physical instability may Commercial amino acid products available in the United States are
occur not only in a TNA but also with Y-site administration.18 Any listed in Tables 1 and 2.27–36 Most are supplied in plastic containers,
change beyond acceptable parameters (eg, maintaining at least 90% with an overwrap to avoid water loss and subsequent altered concen-
of the labeled amount or initial concentration of an active ingredient tration. Plastic is now used in preference to glass bottles for ease of
throughout infusion) indicates instability with the risk of altering thera- transport and storage, and very little plasticizer is expected to leach
peutic effect. Although individual minerals—including electrolytes and into the amino acid solution from the product’s plastic container dur-
trace elements—do not undergo decomposition, they do possess prop- ing storage.
erties (ie, ionization, valence states) that invite compatibility concerns The products vary in the number and concentration of each crys-
or stability concerns for the organic components in the PN admixture talline amino acid, total amino acid, and nitrogen content, as well as
or the emulsion itself. Instability is not reversible; the degradation can electrolyte content and pH.27–36 As a result, the amino acid prod-
be slowed but not stopped. ucts and associated compatibility and stability data are not necessarily
276
4 BOULLATA ET AL .

TA B L E 1 Content and characteristics of AA injection products (pediatric)

Product Aminosyn-PF 10%27 Premasol 10%33 TrophAmine 10%36


Manufacturer ICU Medical Baxter B. Braun
Essential AAs, g/100 ml
Cysteinea 0 <0.016 <0.016
Histidine 0.312 0.48 0.48
Isoleucine 0.76 0.82 0.82
Leucine 1.2 1.4 1.4
Lysine 0.677 0.82 0.82
Methionine 0.18 0.34 0.34
Phenylalanine 0.427 0.48 0.48
Threonine 0.512 0.42 0.42
Tryptophan 0.18 0.2 0.2
a
Tyrosine 0.044 0.24 0.24
Valine 0.673 0.78 0.78
Nonessential AAs, g/100 ml
Alanine 0.698 0.54 0.54
Arginine 1.227 1.2 1.2
Aspartic acid 0.527 0.32 0.32
Glutamic acid 0.82 0.5 0.5
Glycine 0.385 0.36 0.36
Proline 0.812 0.68 0.68
Serine 0.495 0.38 0.38
Taurine 0.07 0.025 0.025
Electrolytes, mmol/L
Acetate 46 94 96.2
Phosphate None NR NR
Sodium None NR 5
Chloride None <3 <3
Other
Na Metabisulfite NF, mg/L 0 0 <50b
Water for injection, USP qs qs qs
pH (range) 5.5 (5.0–6.5) 5.5 (5.0–6.0) 5.5 (5.0–6.0)
Osmolarity, mOsm/L 788 865 875
Total AAs, g/L 100 100 100
Nitrogen, g/L 15.2 15.5 15.5

Abbreviations: AA, amino acid; NR, not reported; USP, United States Pharmacopeia.
a
Conditionally essential.
b
The product packaged in plastic is sulfite-free, compared with the product packaged in glass.
qs: the amount which is enough or sufficient to result in an accurate final volume of the product.

interchangeable. Therefore, a cautious approach to determining com- from the manufacturer and are expected to remain stable in their orig-
patibility and stability data is needed to ensure the evidence is specific inal container during storage. Storage of amino acids should avoid light
to the product in question. Historically, these solutions contained chlo- exposure to maintain stability up to the expiration date. Adult prod-
ride salts of some of the amino acids, but they currently contain some ucts typically do not contain cysteine or taurine or glutamine. Products
acetate salts or acetic acid buffering depending on the amino acid con- intended for pediatric patients contain lower concentrations of alanine,
tent, with consequent pH differences between products. The different arginine, and lysine. Electrolytes may also be found in amino acid solu-
products contain both essential (indispensable) and nonessential (dis- tions, again varying with the product, and need to be considered for
pensable) amino acids at varying proportions in compatible mixtures compatibility and total clinical dose.
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 277
5

TA B L E 2 Content and characteristics of AA injection products (adult)

Product Aminosyn II 15%28 Clinisol 15%29 Plenamine 15%32 ProSol 20%34 Travasol 10%35
Manufacturer ICU Medical Baxter B. Braun Baxter Baxter
Essential AAs, g/100 ml
Cysteinea None None None None None
Histidine 0.45 0.894 0.894 1.18 0.48
Isoleucine 0.99 0.749 0.749 1.08 0.60
Leucine 1.5 1.04 1.04 1.08 0.73
Lysine 1.575 1.18 1.18 1.35 0.58
Methionine 0.258 0.749 0.749 0.76 0.40
Phenylalanine 0.447 1.04 1.04 1 0.56
Threonine 0.6 0.749 0.749 0.98 0.42
Tryptophan 0.3 0.25 0.25 0.32 0.18
a
Tyrosine 0.405 0.039 0.039 0.05 None
Valine 0.75 0.96 0.96 1.44 0.58
Nonessential AAs, g/100 ml
Alanine 1.49 2.17 2.17 2.76 2.07
Arginine 1.527 1.47 1.47 1.96 1.15
Aspartic acid 1.05 0.434 0.434 0.6 None
Glutamic acid 1.107 0.749 0.749 1.02 None
Glycine 0.75 1.04 1.04 2.06 1.03
Proline 1.083 0.894 0.894 1.34 0.68
Serine 0.795 0.592 0.592 1.02 0.50
Taurine None None None None None
Electrolytes, mmol/L
Acetate 107.6 127 151 140 88
Phosphate None None None None None
Sodium 50 None None None None
Chloride None None None None 40
Other
Na Metabisulfite NF, mg/L 0 0 300 NR
Water for injection, USP qs qs qs qs qs
pH (range) 5.8 (5.0–6.5) 6.0 (5.0–7.0) 5.6 (5.2–6.0) 6.0 (5.5–6.5) 6.0 (5.0–7.0)
Osmolarity, mOsm/L 1270 1357 1383 1835 998
Total AAs, g/L 150 150 150 200 100
Nitrogen, g/L 23 23.7 23.7 32.1 16.5

Abbreviations: AA, amino acid; NR, not reported; USP, United States Pharmacopeia.
a
Conditionally essential.
qs: the amount that is enough or sufficient to result in an accurate final volume of the product.

Each crystalline amino acid in a commercial product is considered nine, histidine, and lysine. This buffering capacity will be important to
an active pharmaceutical ingredient and meets standards for purity. the overall PN admixture and is more robust at a higher final concen-
Amino acids are amphoteric in nature; therefore, depending on solu- tration of amino acids in the PN. Additionally, the ratio of basic to acidic
tion pH, they can react as acids and bases, and they are each least sol- amino acids may be important to optimal emulsion stability.37
uble at their isoelectric point (at which ionic charges neutralize each Most products no longer include a sulfite (SO3 2– ) antioxidant
other), thereby reducing any ionic attraction to water. For example, preservative, with its potential to cause hypersensitivity reactions.
tyrosine has low solubility and precipitates at a more acidic pH. The To help avoid oxidation, most products are filled under nitrogen
buffering capacity (titratable acidity) of amino acid solutions is deter- atmosphere or under vacuum. However, unlabeled contaminants are
mined in large part by the proportion of the formulation that is argi- present in amino acid solutions and include aluminum, cadmium,
278
6 BOULLATA ET AL .

chromium, lead, zinc, and possibly manganese.38,39 This contamination freezing is also to be avoided.46–48 Exposure to heat during the ster-
reflects the ability of amino acids to easily form complexes with metals. ilization phase of manufacturing results in the degradation products
The affinity for metals varies with the amino acid, with different kinet- 5-hydroxymethyl-furaldehyde, levulinic acid, and formic acid, which
ics for each complex formation.38 Nonnutrient metals (eg, cadmium, drive down the pH of sterile dextrose solutions over time.20 The small
lead) are most likely to complex with alanine, aspartate, glutamate, amount of the reactive open-chain form of glucose in solution varies
glycine, histidine, methionine, phenylalanine, serine, and threonine.40 depending on that pH.49
Although this likelihood is based on individual amino acid stability con- Dextrose has a molecular weight (MW) of 198.17 Da and, at a con-
stants, the amino acid concentration in a commercial amino acid mix- centration of 5%, is considered isotonic (252 mOsm/L), whereas 70%
ture remains the major determining factor. is hypertonic with an osmolarity of 3532 mOsm/L (∼50 mOsm/L con-
Amino acids can complex with macrominerals such as calcium (with tributed for every 1% dextrose in solution). Isotonic solutions of dex-
lysine especially) and trace minerals such as copper (with cysteine trose 5% have been shown to facilitate growth of Burkholderia cepa-
especially). In most cases, the chelates of trace metals with amino acids cia and Serratia marcescens after contamination at 25◦ C.50 Most PN
are expected to maintain the bioavailability of both components.41 formulations contain dextrose at a final concentration of 10%–25% in
However, copper-cysteine complexes are likely to precipitate. The dilu- present-day clinical practice.
tion of copper (elemental) to a concentration <160 mcg/L is expected The commercially available concentrations of dextrose contain no
to avoid this complexation.42,43 Precipitates (yellow-to-brown) were more than 25 mcg/L of aluminum or 5 mg/L of di-2-ethylhexyl phtha-
noted in pediatric PN admixtures.43 The precipitate was attributed to late (DEHP) (in polyvinyl chloride [PVC] bags) within their expiration
an incompatibility between copper (as cupric sulfate [SO4 2– ]) and a periods, which may be of most importance in neonates with chronic
cysteine-containing, lower-pH amino acid solution, which did not recur exposure.51 Patients with a known allergy to corn or corn products
when using a different amino acid solution.43 No evaluation was made should avoid use of dextrose.47,48
to determine whether the precipitate was copper cysteinate, copper
sulfide (S2– ), or both.
Two amino acids are each available as a separate injectable prod- Lipid emulsions
uct in glass containers: cysteine and arginine. But only cysteine is
intended for PN admixtures when indicated. Cysteine hydrochloride ILEs are an essential component in PN as a source of energy and
is available as a 50-mg/ml (34.5 mg/ml cysteine) solution at a pH essential fatty acids and as a therapeutic modality. During the man-
of 1.0–2.5. This amino acid may form the insoluble dimeric form ufacturing process, an egg lecithin–based phospholipid emulsifier is
cystine over time. As mentioned above, the complex of cysteine used to disperse the oil phase into the aqueous phase to ensure a
with copper may precipitate out of solution, decreasing amino acid stable emulsion. ILE products are oil-in-water emulsions consisting
concentration.44 of one or more oils containing triglycerides, a phospholipid emulsifier,
Although not an amino acid, L-carnitine is synthesized endogenously and glycerol and are manufactured to mimic properties of natural
from lysine and methionine. This nutrient is available for inclusion in PN chylomicrons (Table 3).52–58 Sodium hydroxide is added to maintain
admixtures when indicated. The product (200 mg/ml) does not contain a pH range between 6 and 9, and the glycerol is added to improve
a preservative. Hydrochloric acid and sodium hydroxide may be used to tonicity and provide additional energy.59 Nearly 1 million lipid droplets
adjust the product pH to 6.0–6.5. As a zwitterion (ie, contains both pos- can be found per milliliter, and the emulsion remains most stable
itively charged and negatively charged functional groups) with a pKa of at pH 8 with a surface potential of at least −35 mV in repulsive
3.8, this very water-soluble primary amine can behave as a basic amino forces.60
acid. The United States Pharmacopeia (USP) Chapter <729> (“Globule Size
Distribution in Lipid Injectable Emulsions”) provides explicit pharma-
copeial specifications to ensure manufacturer product integrity, includ-
Dextrose ing a mean droplet diameter below 500 nm (0.5 μm) and a large globule
content (percentage of fat globules >5 μm or ‘‘microns’’ [ie, PFAT5 ]) no
Dextrose, or α-D-glucose monohydrate (C6 H12 O6 ⋅H2 O), is a natural greater than 0.05%.61 Additionally, ILEs must possess a free fatty acid
monosaccharide resulting from the hydrolysis of corn starch and is the content no greater than 0.07 mEq/g.
predominant source of carbohydrate in PN.45 Each gram of dextrose These emulsions, however, are inherently unstable systems and,
monohydrate provides 3.4 kcal or 14 kilojoules (anhydrous dextrose over time, may undergo various stages of destabilization such as aggre-
provides 4 kcal/g). It is a sterile, nonpyrogenic IV solution commercially gation, creaming, coalescence, and increased particle size. In aggre-
available in concentrations ranging from 5% to 70% and contains no gation, dispersed lipid droplets come together but do not fuse. These
antimicrobial agent. IV dextrose products have a pH range between aggregates may be redispersed with gentle agitation and safely admin-
3.2 and 6.5, with most solutions in the range of 4–4.5, since the solu- istered to patients. Creaming is the initial stage of emulsion destabiliza-
tion contains no added buffer. Dextrose may convert to anhydrous tion and is one of only two stages that may be detectable by the naked
glucose in temperatures >40◦ C, so excessive heat exposure is to be eye, the other being free oil as the final stage of a broken emulsion.
avoided and temperatures of 20–25◦ C are recommended for storage; The lipid particles present in a cream layer are destabilized, but their
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 279
7

TA B L E 3 Composition and properties of lipid injectable emulsion products

Category Component OO,SO-ILE53 SO-ILE54 SO-ILE55 FO-ILE56 SO,MCT,OO,FO-ILE57


Commercial product concentration 20 20,30 20 10 20
Source oil SO, % 20 100 100 0 30
FO, % 0 0 0 100 15
MCT, % 0 0 0 0 30
OO, % 80 0 0 0 25
Additives Egg phospholipid, g/100 ml 1.2 1.2 1.2 1.2 1.2
Glycerin, g/100 ml 2.25 2.25 2.5 2.5 2.5
α-Tocopherol, mg/100 ml 3.2 0 0 15–30 16.3–22.5
Sodium oleate, g/100 ml 0.03 0 0.03 0.03 0.03
ω-3 Linolenic acid, % 0.5–4.2 4–11 4–11 1.1 (mean) 1.5–3.5
EPA, % 0 0 0 13–26 1–3.5
DHA, % 0 0 0 14–27 1–3.5
ω-6 Linoleic acid, % 13.8–22 44–62 48–58 1.5 (mean) 14–25
Arachidonic acid, % 0 0 0 0.2–2 NR
ω-7 Palmitoleic acid, % 0 0 0 4–10 NR
ω-9 Oleic acid, % 44.3–79.5 19–30 17–30 4–11 23–35
Saturated fatty acids Caprylic acid, % 0 0 0 0 13–24
Capric acid, % 0 0 0 0 5–15
Palmitic acid, % 7.6–19.3 7–14 9–13 4–12 7–12
Stearic acid, % 0.7–5 1.4–5.5 2.5–5 0 1.5–4
Myristic acid, % 0 0 0 2–7 NR
ω-6:ω-3 ratio 9:1 7:1 7:1 1:8 2.5:1
Phytosterols, mg/L 208.8 ± 39.4 422.4 ± 130.5 NR 0 142.2 ± 15.3
Phosphate, mmol/L 15 15 15 15 15
Energy kcal/ml 2 2 2 1.12 2

Abbreviations: DHA, docosahexaenoic acid; EPA, eicosapentanoic acid; FO-ILE, fish-oil lipid injectable emulsion (Omegaven, Fresenius Kabi USA); OO,SO-ILE,
olive-oil, soybean-oil lipid injectable emulsion (Clinolipid, Baxter Healthcare Corporation); NR, none reported; SO-ILE, soybean-oil lipid injectable emulsion
(Intralipid, Baxter Healthcare Corporation; Nutrilipid, B. Braun Medical); SO,MCT,OO,FO-ILE, soybean-oil, medium-chain triglycerides, olive-oil, fish-oil lipid
injectable emulsion (Smoflipid, Fresenius Kabi USA).

individual droplet identities are generally preserved. A cream layer is ing the stability of the emulsifier. Compatibility and stability of an ILE
visible at the surface of the emulsion as a translucent band separate are dependent on the emulsifier’s strength and the presence of con-
from the remaining dispersion. With gentle agitation, a creamed emul- founders that may counteract the emulsifier’s repulsive properties to
sion may be redispersed and safely administered to a patient. Coales- keep oil droplets apart.63 The zeta potential can be negated with the
cence occurs when there is a fusion of lipid droplets, leading to a fewer addition of divalent cations (eg, calcium, magnesium), trivalent cations
number but an increase in droplet size. This type of emulsion cannot be (eg, iron), or high concentrations of monovalent cations (eg, potassium,
redispersed and is unsafe to administer to patients or include in a PN sodium) or by decreasing the emulsion’s pH. As the pH decreases to
admixture.62 <5, the zeta potential becomes less negative and more neutral, thus
The stability of ILE is dictated by its zeta potential and the effective- increasing the chance of droplets coalescing and creating larger, unsafe
ness of its emulsifying agent. The emulsifier forms a mechanical bar- particles. Optimal ILE stability is achieved at a pH range of 7–8 and
rier around each fat droplet and simultaneously forms an electrostatic when the zeta potential is high (ie, more negative), at least −35 mV,
(ionic) barrier with their ionized phosphate groups to separate droplets as the repulsive forces will exceed the attractive forces. Conversely, all
from each other and the surrounding water. Emulsifying agents cre- repulsive forces disappear by a pH of 2.5 and the lipid particles will coa-
ate a barrier that prevents droplets from coalescing to form unsafe, lesce, resulting in an unsafe emulsion. Other factors that can disrupt
larger globules. The zeta potential is a negative charge created by the an emulsion’s integrity include exposure to oxygen, as it will oxidize the
emulsifier that generates repulsive forces that act to separate parti- fats within the emulsion. Package design and use of an oxygen absorber
cles and work against attractive (van der Waal) forces, thus improv- help lessen this risk.64 Even with these protective measures, ILEs will
280
8 BOULLATA ET AL .

destabilize over time as the emulsion’s pH decreases and particle size Multielectrolyte additives
increases. Products that contain multiple electrolytes may offer some conve-
Until recently, only soybean-oil ILEs were available in the United nience as a component in preparing PN admixtures. Each product
States. In the past decade, ILEs containing olive oil, fish oil, and medium- will vary in the concentration and specific electrolyte salts, often low
chain triglycerides (MCTs) have become available (Table 3).52 Because in potassium and without phosphorus. As a result, the pH may vary
of the differences in chain length associated with these sources of between products in a range of ∼4–8, often requiring hydrochloric acid
triglycerides, the same phospholipid emulsifying agent may behave or acetic acid for pH adjustment, and are hypertonic (Hyperlyte CR [B.
differently, and emulsion stability may be different depending on the Braun] [pH 5.0–5.4], 5500 mOsm/L; TPN Electrolytes [Hospira, Inc] [pH
blend of oils used in an ILE.65 For example, MCTs can improve the sta- 6.0–7.5], 6200 mOsm/L). These sterile solutions may contain one or
bility of an ILE by displacing long-chain triglycerides (eg, from soybean, more different sodium salts (acetate, chloride, gluconate), potassium
fish, or olive oil) at the droplet surface, thus reducing stress on the chloride, magnesium chloride, and often calcium chloride. Incidentally,
emulsifier because of its shorter chain length.66 magnesium chloride and calcium chloride may be included as part of
For these reasons, compounding a TNA (ie, 3-in-1 PN admixture) standardized, commercially available PN products. When these prod-
or coadministering parenteral medications with an ILE remains a chal- ucts are used as a component for preparing a final PN admixture for
lenge. Any time an ILE has another medication or IV fluid mixed with it, a patient, the difference in any included electrolyte salts is important
there is a potential for a disruption of the emulsion or coalescence that to note, as chloride salts tend to dissociate to a greater degree and are
creates larger fat globules that may be unsafe to infuse. Some literature available for interaction, with an influence on pH.
on ILE stability may not utilize the methods specified by USP Chapter Bicarbonate, citrate, and lactate salts should be avoided in PN
<729> (ie, dynamic light scattering, light obscuration) for determining admixtures because of incompatibilities. For example, with a pH of
the stability of an emulsion. Importantly, both methods must be used nearly 8, sodium bicarbonate drives precipitation of several salts,
in a complementary fashion to assess the quality of ILEs. Dynamic light including calcium and magnesium. Cardioplegic solutions, intraperi-
scattering is only a sizing method and provides only semiquantitative toneal irrigation solutions, or dialysate solutions—which usually con-
information on the relative droplet size distribution; it does not count tain sodium, magnesium, and calcium as chloride salts, as well as con-
the number of particles or droplets. In contrast, light obscuration both taining potassium chloride in cardioplegic solutions, or other sodium
counts the fat globules present and identifies the size of lipid droplets salts (eg, acetate, bicarbonate, gluconate, or lactate) in irrigation and
(1.3–400 μm), providing “reliable” count values at a size threshold dialysate solutions—are not indicated as components to be used for PN
of 5 μm. admixtures.

Sodium and potassium chloride or acetate


Micronutrients The chloride salts (sodium chloride, potassium chloride) generally form
neutral aqueous solutions available for use in preparing PN admixtures.
The micronutrients include electrolytes, trace elements, and vitamins. As a weaker conjugate acid anion, the acetate salts (sodium acetate,
Each is a required component in a PN admixture to support a patient’s potassium acetate) form more alkaline solutions. Despite significant
metabolic needs. A parenteral micronutrient product may include mul- dissociation of the ions from these salts in solution, the monovalent
tiple electrolytes, multiple trace elements, or multiple vitamins, but minerals are less likely to interact with other components at typical
several of these nutrients are also available as single-entity products. final admixture concentrations.
The physicochemical properties of each active ingredient and accom-
panying excipients are important to the issue of PN admixture compat- Magnesium sulfate or chloride
ibility and stability. Although some of the mineral salts are organic, the Aqueous solutions of magnesium are expected to have a pH of 5.5–
remainder are inorganic. 7.0; this includes magnesium sulfate products adjusted with sulfu-
ric acid and sodium hydroxide. Magnesium chloride is not typically
available as a separate additive but has been included in combi-
Electrolytes nation products, including the standardized, commercially available
PN products.
The electrolytes, found as salts in injectable products, that serve as
components for PN admixtures include sodium, potassium, magne- Calcium gluconate or chloride
sium, calcium, and phosphorus. Generally, electrolyte solutions are Although calcium gluconate is the preferred salt for use in compound-
mildly acidic with polarity similar to water. This allows the salts to dis- ing PN because of its lower risk of precipitation, calcium chloride has
sociate in aqueous PN admixtures to varying degrees depending on also been used. The pH for calcium gluconate injections ranges from
the salt and concentration. The monovalent electrolytes (sodium and 6.0 to 8.2 and is adjusted with hydrochloric acid and/or sodium hydrox-
potassium in their representative salts) are much less likely than the ide. Based upon this pH range, this salt is more likely to be mildly alka-
multivalent electrolytes (calcium, magnesium) to pose compatibility line than acidic. The pH of calcium chloride injections is 5.5–7.5 when
concerns. diluted with water for injection, and it may contain hydrochloric acid
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 281
9

and/or sodium hydroxide for pH adjustment. Additionally, calcium chlo- Multielement additives
ride dissociates more extensively in water than does calcium gluconate, Historically, products have contained up to seven trace elements, but
rendering more calcium ions free to complex with phosphate and to multi–trace element products in the United States currently remain at
produce calcium phosphate precipitates. Even if the extent of dissoci- four elements in fixed-concentration proportions. The available prod-
ation was similar, calcium gluconate dissociates into a weak acid (glu- ucts vary in concentration of each trace element, the excipients, and
conic acid) and a strong base (calcium hydroxide), whereas calcium pH. Sulfuric acid and sodium hydroxide may be used to adjust product
chloride dissociates into a strong acid (hydrochloric acid) and a strong pH to 1.5–3.5. Products with excessive copper and manganese content
base (calcium hydroxide). have been transitioned to products with lower concentrations of these
minerals in keeping with expert recommendations.69,70 Products con-
Sodium or potassium phosphate taining fluoride, iodine, iron, and/or molybdenum may still be available
Mixtures of inorganic monobasic (dihydrogen) and dibasic (monohy- outside the United States.
drogen) phosphate salts of sodium and potassium are available for
use in compounding PN in the United States. Although the pH of Chromium
each individual phosphate salt is not identified in USP monographs, Chromium can exist in multiple oxidation states, including the toxic
the pH of sodium phosphates injection has been reported in the hexavalent chromate form. The chromic (Cr3+ ) ion is the most physio-
range of 5.0–6.0, whereas the pH of potassium phosphates injection logically valuable and the one used in parenteral products, compared
has been reported between 6.2 and 7.8. The degree of dissociation with the chromous (Cr2+ ) form. Chromic chloride is a hydrated salt
of inorganic phosphate salts into free phosphate anions is largely (MW = 266.5 Da) containing just under 20% chromium. Hydrochloric
dictated by pH. In addition, phosphate may occur in three differ- acid and sodium hydroxide may be used to adjust product pH to 1.5–
ent anionic forms, including H2 PO4 − anion (dihydrogen or monoba- 2.5. Given the amounts of chromium present as a contaminant in other
sic phosphate), HPO4 2− (monohydrogen or dibasic phosphate), and PN components (eg, amino acid solutions), the requirement for this
PO4 3− (tribasic phosphate), all of which may generate insoluble cal- trace element as an additive in short-term treatment has been recon-
cium phosphate. The content of aluminum contamination is much sidered. Although chromium concentrations can be measured using
greater in potassium phosphates injection than in sodium phosphates neutron activation analysis, atomic absorption spectrophotometry, or
injection. inductively coupled plasma with mass spectrometry (ICP-MS), these
reflect total chromium and may not adequately distinguish oxidation
state of the mineral, which is important to compatibility as well as phys-
Trace elements iologic activity.

In the time since the initial recommendations for including copper, Copper
chromium, manganese, and zinc in PN admixtures,67 and after hospi- The oxidized cupric (Cu2+ ) form is more soluble and stable in solution
tals had to prepare their own formulations,68 there have been a num- than the cuprous (Cu1+ ) state. Copper (cupric) sulfate is a hydrated salt
ber of lessons learned in IV trace element requirements as well as an (MW = 249.69 Da) containing just over 25% copper. Sulfuric acid and
evolution in the available products on the market.69–72 Currently, there sodium hydroxide may be used to adjust product pH to 2.0–3.5. Copper
are multi–trace element products as well as single-entity trace ele- (cupric) chloride is also available and may contain hydrochloric acid and
ment products available to include in PN admixtures or other carrier sodium hydroxide to adjust product pH to 1.5–2.5. Copper can form
fluids (Table 4).73–76 The multi-element products are commonly used complexes with organic anions, including some amino acids, depending
because of convenience, whereas the single-entity products become on the degree of other competing cations available in the admixture.
more valuable for long-term use in patients with varying requirements Some of these copper complexes may have limited solubility. Although
or for individual-element repletion independent of the PN admixture. copper concentrations can be measured using atomic absorption spec-
Although rarely observed, certain trace element solutions may exhibit trophotometry or ICP-MS, these reflect total copper content and can-
a slight colored hue. not distinguish oxidation state or the free form from the complexed
Products formulated for neonatal and pediatric patients do not con- mineral, including insoluble microprecipitates. For example, a PN com-
tain a preservative, as the commonly used benzyl alcohol is associ- patibility study that noted no visually apparent copper precipitate
ated with adverse effects. Most multidose vial products contain no and reported unchanged copper concentration over time using atomic
preservative/bacteriostatic agent. Without a preservative, the vials of absorption did not necessarily rule out copper microprecipitate.77
any trace element product have a limited beyond-use date after initial
entry. Of note, the concentration of each trace element is relatively low Manganese
in the products, made even lower when added to a PN admixture, cre- Of the several oxidation states of manganese, the divalent (Mn2+ ) form
ating a challenge in analysis when close to the limits of detection. Fur- is most stable in aqueous solutions. This form has been available in both
thermore, most methods of analysis determine mineral concentration the chloride and sulfate salts. Manganese sulfate is a hydrated salt (MW
without distinguishing between oxidation states or complexation and = 169 Da) containing 32.5% manganese. Sulfuric acid may be used
precipitation. to adjust product pH to 2.0–3.5. Although manganese concentrations
10
282

TA B L E 4 Trace element products

Multiple-entity product Single-entity product

Pediatric Adult

Multitrace-4 Multitrace-4
Neonatal73 Multrys Pediatric73a Tralement74b Chromium Copper Manganese Selenium Zinc Zinc
(1 ml) (1 ml) (1 ml) (1 ml) (1 ml) (1 ml) (1 ml) (1 ml) (1 ml) (1 ml)
Trace elements
Chromium (as chloride), mcg 0.85 None 1 None 4 None None None None None
Copper (as sulfate), mcg 100 60 100 300 None None None None None None
Copper (as chloride), mcg None None None None None 400 None None None None
Manganese (as sulfate), mcg 25 3 25 55 None None 100 None None None
Selenium (as selenious acid), mcg None 6 0 60 None None None 60 None None
Zinc (as sulfate), mg 1.5 1 1 3 None None None None 3 5
Other
Water for injection, USP qs qs qs qs qs qs qs qs qs qs
Preservative None None None None None None None None None None
pH 2.3–2.7 1.5–3.5 1.5–3.5 1.5–3.5 1.5–2.5 2.0–3.5 2.0–3.5 1.8–2.4 2.0–4.0 2.0–4.0
Osmolarity, mOsm/L 50 NR 34.7 114 308 327 3.64 16 96.5 157.2

Abbreviations: NR, not reported; USP, United States Pharmacopeia.


a
Manufacturer has voluntarily ceased production to develop US Food and Drug Administration (FDA)–approved products that align with American Society for Parenteral and Enteral Nutrition (ASPEN) recom-
mendations.
b
Tralement is indicated for pediatric patients ≥10 kg as well as adults.
qs: the amount that is enough or sufficient to result in an accurate final volume of the product.
BOULLATA ET AL .
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 283
11

can be measured using neutron activation analysis, atomic absorption TA B L E 5 Pediatric parenteral multivitamin ingredients
spectrophotometry, or ICP-MS, these reflect total manganese and can-
Vitamin Source Amount84,86
not distinguish between the free and the complexed mineral.
A Retinol palmitate 2300 IU (0.7 mg)
B1 Thiamin hydrochloride 1.2 mg
Selenium
B2 Riboflavin 5-phosphate sodium 1.4 mg
Selenium can exist in a number of oxidation states. Selenious acid
(H2 SeO3 ) incorporating the selenite ion (SeO3 2– ) and containing B3 Niacinamide 17 mg

61% selenium has been more commonly used than sodium selenate B5 Dexpanthenol 5 mg
(SeO4 2– ) for parenteral preparations. Nitric acid may be used to adjust B6 Pyridoxine 1 mg
product pH to 1.8–2.4. Although selenium concentrations can be mea- C Ascorbic acid 80 mg
sured using atomic absorption spectrophotometry or ICP-MS, these D3 Cholecalciferola 400 IU (10 mcg)
reflect total selenium and cannot distinguish oxidation state, includ-
D2 Ergocalciferola 400 IU (10 mcg)
ing the poorly soluble elemental (Se0 ) form, or distinguish between the
E DL -α-Tocopheryl acetate 7 IU (7 mg)
free and complexed mineral.
K1 Phytonadione 200 mcg

Zinc B9 Folic acid 140 mcg

Fortunately, zinc is very soluble across a wide range of pH in its ionic Biotin Biotin 20 mcg

and stable divalent (Zn2+ ) form. So there has been no concern about B12 Cyanocobalamin 1 mcg
reduction to the less soluble elemental form. Zinc sulfate is a hydrated a 86
Pediatric Infuvite Multiple Vitamins contains cholecalciferol, whereas
salt (MW = 287.56 Da) containing 22.7% zinc. Sulfuric acid may be used M.V.I. Pediatric84 lyophilized powder contains ergocalciferol.
to adjust product pH to 2.0–4.0. Zinc can form complexes with organic
anions, including some amino acids with multifaceted coordination
structure. Zinc chloride may form a flocculent precipitate in aqueous fulness. Unlike enterally administered iron, parenteral iron bypasses
solution, owing to cationic hydrolysis to the oxychloride, which is less the complex homeostatic control of iron bioavailability at the gas-
evident when adjusted to low pH with hydrochloric acid.68 Although trointestinal tract. Routine monitoring of iron status should be con-
zinc concentrations can be measured using atomic absorption spec- sidered, as iron overload can increase oxidative stress and the risk of
trophotometry or ICP-MS, these reflect total zinc and cannot distin- infection.82 In addition to the clinical issues associated with parenteral
guish the free from the complexed mineral. iron (eg, impaired immune function, anaphylaxis risk, support bacte-
rial growth during acute infection), there are significant compatibility
Iron concerns.81,83
For several reasons, iron salts are not typically added to PN admix-
tures, nor are they a component of currently available multi–trace ele-
ment products in the United States. However, iron salt is present as Vitamins
ferric chloride in one product, Addamel N (Fresenius Kabi), that has
been imported in times of shortage.78 Iron dextran injection, USP is Multivitamins
a dark brown, slightly viscous sterile liquid complex of ferric hydrox- Commercially available adult and pediatric IV multivitamin formu-
ide and dextran containing 50 mg/ml iron. In its undiluted state, the lations comprise water-soluble and lipid-soluble vitamins (Tables 5
pH of the solution is between 5.2 and 6.5 with added sodium hydrox- and 6).84–87 The M.V.I.-12 formulation for adults, which did not con-
ide and hydrochloric acid for pH adjustment.79 Although iron dex- tain vitamin K, is no longer available. Various excipients are added to
tran has been the traditional iron product used in PN, iron sucrose the multivitamin products to maintain the integrity of the product. To
is being used more frequently in clinical practice for iron replace- maintain stability, the adult and selected pediatric IV multivitamin for-
ment therapy because of its lower incidence of adverse effects. Iron mulations are separated into two vials.85–87 Vial 1 of the Infuvite Adult
sucrose injection is a brown, sterile, aqueous complex of polynuclear and Pediatric formulations has a pH of 5.0–6.0, whereas the pH of
iron (III)–hydroxide in sucrose (MW = 34,000–60,000 Da). Iron sucrose vial 2 is 5.0–7.0 (Sandrine Banzundama Bazuta Feza, Medical Infor-
injection (20 mg/ml) contains ∼30% sucrose wt/vol (300 mg/ml) and mation Coordinator, Sandoz Canada Inc, written communication, May
has a pH of 10.5–11.1. Undiluted, the injection has an osmolarity of 20, 2020). After the contents of vials are combined, the mixture is
1250 mOsm/L.80 only stable for 4 h under refrigeration.85–87 Furthermore, once con-
Iron is rarely indicated for inclusion in PN admixtures for short- tents of both vials are introduced into a PN admixture, the infusion
term use. Typically, patients receiving long-term PN (ie, >3 weeks) will should be completed within 24 h. A pediatric multivitamin formulation
require some form of iron supplementation to meet ongoing needs. is also available as a lyophilized powder for reconstitution.84 The adult
Microcytic, hypochromic anemia resulting from iron deficiency can multivitamin products should be further diluted in compatible solu-
occur in patients receiving iron-free PN in as little as 2 months.81 When tions of volumes ≥500 ml,85–87 whereas pediatric multivitamin prod-
administered parenterally, concern for iron overload limits its use- ucts require diluent volumes ≥100 ml.84,86
284
12 BOULLATA ET AL .

TA B L E 6 Adult parenteral multivitamin ingredients given to the patient to determine the allergenicity.93 If identified, the
allergenic component should be avoided. The patient should be closely
Vitamin Source Amount85,87
monitored in an inpatient setting when the test dose or modified PN
Vial 1 (5 ml) ingredients
formulation is initiated.93
A Retinol palmitate 3300 IU (1 mg)
B1 Thiamin hydrochloride 6 mg
Ascorbic acid
B2 Riboflavin 5-phosphate sodium 3.6 mg Besides being a component of IV multivitamin formulations, ascorbic
B3 Niacinamide 40 mg acid is available as a single-entity product with a concentration of 500
B5 Dexpanthenol 15 mg mg/ml.94,95 Excipients include disodium edetate, a chelating agent to
B6 Pyridoxine 6 mg protect against oxidation,89 and sodium hydroxide and sodium bicar-

C Ascorbic acid 200 mg bonate to buffer the solution to a pH range between 5.5 and 7.0.94,95
The solution should be stored under refrigeration and protected from
D3 Cholecalciferola 200 IU (5 mcg)
light.94,95 A hypertonic product (5900 mOsm/L), the ascorbic acid solu-
D2 Ergocalciferola 200 IU (5 mcg)
tion must be further diluted with a compatible diluent (eg, dextrose 5%
E DL -α-Tocopheryl acetate 10 IU (10 mg)
in water) to a final concentration of 1–25 mg/ml.95 Ascorbic acid is used
K1 Phytonadione 150 mcg at lower concentrations when included as an additive to PN admixtures
Vial 2 (5 ml) ingredients to avoid compatibility and stability concerns.
B9 Folic acid 600 mcg
Biotin Biotin 60 mcg Thiamin
B12 Cyanocobalamin 5 mcg Thiamin hydrochloride is available as a 100-mg/ml solution as well
as an ingredient in IV multivitamin products. The single-entity vial
a
Adult Infuvite Multiple Vitamins87 contains cholecalciferol, whereas M.V.I.
may contain chlorobutanol anhydrous and/or monothioglycerol as
Adult85 contains ergocalciferol.
preservatives.89,96,97 Sodium hydroxide is added to maintain a pH
range of 2.5–4.5.96,97 The thiamin solution should be stored at room
For both adult and pediatric multivitamin products, the lipid-soluble temperature and protected from light.96,97
vitamins are solubilized with the aqueous vitamins through the addi-
tion of the surfactant polysorbate 80.84–87 Both M.V.I. Adult and Pedi- Pyridoxine
atric products contain an additional surfactant, polysorbate 20.84,85 Pyridoxine is available as a 100-mg/ml solution in addition to being
Infusion of large amounts of polysorbate 80 has been associated with included in IV multivitamin products. The excipients of the single-
serious adverse events, including fatality in low-birth-weight infants.88 entity product include chlorobutanol anhydrous, a preservative,89
In the 1980s, an injectable vitamin E formulation (E-Ferol), contain- and sodium hydroxide to maintain the pH between 2.0 and 3.8.98
ing polysorbate 80 (9%) and polysorbate 20 (1%), administered intra- The solution should be protected from light and stored at room
venously led to the development of a multiorgan toxicity referred to temperature.98
as E-Ferol syndrome.88 E-Ferol syndrome is a life-threatening con-
dition characterized by vasculopathic hepatotoxicity, ascites, throm- Folic acid
bocytopenia, and renal failure in low-birth-weight infants.88 Follow- Folic acid is available as a sodium folate 5-mg/ml multiple-dose solu-
ing reports of this lethal reaction, E-Ferol was withdrawn from the tion as well as being a component of IV multivitamin solutions. The
market.88 Commercially available pediatric multivitamin products con- single-entity folic acid solution is yellow and contains edetate disodium
tain a small quantity (50 mg) of polysorbate 80 in 5 ml of solution or 1% and benzyl alcohol 15 mg as a preservative.89,99 Benzyl alcohol has
(wt/vol).84,86 been associated with hypersensitivity reactions and gasping syndrome
Propylene glycol facilitates the dissolution of vitamins in the adult in neonates, characterized by respiratory depression, metabolic acido-
multivitamin products.89 Because of the risk of hyperosmolality in low- sis, encephalopathy, and intracranial hemorrhage.89 The pH of the folic
birth-weight infants, propylene glycol is not included in pediatric mul- acid solution is maintained between 8.0 and 11.0 with hydrochloric acid
tivitamin products.90 Antioxidants such as gentisic acid ethanolamide, and sodium hydroxide, because of the risk for precipitation out of solu-
butylated hydroxytoluene, and butylated hydroxyanisole are added tion at lower pH.99 The folic acid vial should be protected from light and
to selected adult multivitamin products.89,91 In addition to butylated stored at room temperature.99
hydroxytoluene and butylated hydroxyanisole, mannitol serves as an Vitamins can be highly reactive molecules, setting up potential com-
antioxidant in pediatric multivitamin products.92 Buffer agents such patibility issues with each other or with other components of a PN
as sodium hydroxide/hydrochloric acid or citric acid/sodium citrate admixture as well as stability issues on their own or for other com-
optimize the pH of the multivitamin products.84–87 Multivitamin vials ponents of a PN admixture. Incompatibilities depend on the rela-
of current US products should be refrigerated and protected from tive concentrations of ingredients; pH; exposure to temperature, oxy-
light.84–87 Hypersensitivity reactions to multivitamin products (active gen (air), and light; and the duration of time in reaction. As a result
ingredients and excipients) have been reported.93 A test dose can be of compatibility and stability concerns, the vitamins are added as
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 285
13

the last component and as close to the time of administration as pH (∼7.5).107 At lower molar ratios of glucose to amines, the activation
practical. energy for the reaction may increase compared with those at higher
ratios.105
The addition of ILE to the admixture of amino acids-dextrose not
COMPATIBILITY only will increase the complexity by combining active ingredients and
excipients and by shifting pH character but will alter the physical
Admixture compatibility—Focus on macronutrients system as the entire PN admixture becomes an oil-in-water emul-
sion. An early study of a PN admixture (Veinamine AA3.2%-D20%-
Preparing a PN admixture will influence the properties and safety of L4% Intralipid) noted no visually apparent incompatibility, and fatty
the individual macronutrient components. The acidic pH of IV dextrose acid analysis revealed no significant changes in composition at 24
is well buffered when combined with an amino acid solution. The vis- or 48 h at 4◦ C.20 An evaluation of several adult amino acid prod-
cosity of the concentrated dextrose (eg, 70% product) is also tempered ucts (Travasol, Aminosyn, FreAmine III, FreAmine HBC, HepatAmine,
in an admixture. One of the first compatibility studies evaluated the and NephrAmine) in TNAs using a 100% soybean-oil ILE (Soyacal
combination of amino acids at a final concentration of 4.25% (ie, 4.25 g 20%) and dextrose 70% in a 1:1:1 volume ratio plus micronutri-
of amino acids per 100 ml of PN admixture) and dextrose at a final con- ents was conducted over 14 days at 4◦ C.49 Although the PN admix-
centration of 25% (AA4.25%-D25%), stored at 4◦ C, 25◦ C, or 37◦ C.100 ture pH and osmolality differed between preparations, they did not
The mixture showed a slight decrease in pH over time as well as color change significantly over the study period. An evaluation was per-
formation, especially at higher temperature. Some amino acid degrada- formed of a pediatric amino acid product (TrophAmine 10%) in a
tion occurred (see Stability section). In another study with an AA4.25%- PN admixture using ILE 20% (Intralipid, Liposyn II, or Nutrilipid)
D25% solution without any additives, the pH only changed from 6.5 to and dextrose 70% with micronutrients at several final macronutri-
6.4 initially but then remained constant for up to 2 weeks.101 In these ent concentrations for 24 h at 4◦ C and an additional 24 h at room
glass containers, the average counts of particulate matter in the PN temperature.108 No visible precipitates or color changes were noted,
preparation over time were reported, using the microscopic method, as and pH remained 5.5–6.6, with the lowest value associated with the
2.7 per milliliter (particle size 10–25 μm) and 0.13 per milliliter (particle highest dextrose concentrations.108 In addition to the buffering capac-
size 25–50 μm), which is well below the USP threshold.101 That thresh- ity provided by higher amino acid concentration, the higher final dex-
old criteria (USP method 2) for IV admixtures with a volume >100 ml trose concentration may provide some emulsion protection based on
state there should be ≤12 particles/ml of size ≥10 μm and ≤2 parti- its viscosity.109,110 Stability of the final admixture emulsions will be
cles/ml of size ≥25 μm.102 described later (see Stability section). This section on admixture com-
Owing to the reducing properties of dextrose, a chemical inter- patibility reinforces that PN admixtures should be stored at 4◦ C for
action referred to as the Maillard reaction may occur between glu- reasons above and beyond sterility concerns (ie, the focus of USP Chap-
cose and free (nonionized) amino acids. As a result, no commercial ter <797>). A broader point is that setting a beyond-use date for
products with amino acids and dextrose premixed can be manufac- PN admixtures will depend on sterility as well as compatibility and
tured because this glycation reaction would occur during the prod- stability.
uct’s heat sterilization. The Maillard reaction is the same nonenzy-
matic series of reactions that causes browning in food and that also
accounts for HbA1c formation. Glucosylamines form between glucose Admixture compatibility—Focus on electrolytes
carbonyl groups and amino groups of the amino acids, creating unsta-
ble Schiff bases that rearrange chemically to form Amadori compounds. Adding electrolytes directly to an amino acids solution resulted in visi-
The initial steps can be reversible, but subsequent reaction steps are ble precipitates in a large proportion of over 80 combinations that dif-
not and lead to loss of amino acid bioavailability. A solution color fered in proportional ratios of the minerals and in mixing sequences.111
change may be noted from yellow to a darker amber and brown in The majority of the precipitates evaluated were revealed to contain
the presence of the pigmented compounds.103 Unrecovered urinary calcium and phosphate. Monovalent ions can associate with the zwit-
losses of Maillard products may be accompanied by increased loss terionic form of amino acids but are not expected to form precipitate.
of trace elements (eg, zinc).104 The reaction is more likely to occur When varying the concentration of calcium and magnesium with
with time and at 25–30◦ C compared with 4◦ C but is minimized at high phosphorus content (65 mmol/L), higher levels of magnesium
pH 3.5–4.0.103,105 For example, at AA4.25%-D25%, the average loss protected against precipitation of calcium phosphate.112 This is likely
of amino acids to the Maillard products was ≤2% between 1 and 30 because magnesium forms a more stable and soluble salt with
days when stored at 4◦ C.106 This increased to a ∼6% loss at room phosphate.112 Methods for predicting calcium phosphate precipitation
temperature and will be even greater in the presence of electrolytes must be validated by experimentation and consider the entire formula-
and increasing pH values.106 Amino acid losses may be significant over tion, preparation technique, and chemical kinetics.
time, depending on storage conditions.105 The rates of reaction will In the United States, we still use inorganic phosphate salts (Na or
vary with the amino acid involved, and lysine may be the most reac- K), which pose the risk for insoluble complexes with cations—especially
tive in the Maillard reaction. A nearly 20% loss of lysine may occur by calcium. It is important to note that precipitates can occur in PN but are
1 month at 30◦ C, which tends to be more reactive, especially at higher not always visible, will not appear immediately after compounding, and
286
14 BOULLATA ET AL .

can occur later in the administration set/catheter. These precipitates admixtures, especially for neonates, will require additional study using
develop because the three ionic phosphate species exist in an equilib- different amino acids products, as well as varying the salt form and con-
rium in aqueous solution—monobasic (H2 PO4 – ), dibasic (HPO4 2– ), and centrations of calcium and phosphate.116
trivalent (PO4 3– ). Based on the pKa values for each equilibrium, the pH

of solution will determine which phosphate species predominates. The


risk of insolubility depends on much more than just the combination Phosphate salts
of calcium and phosphate salt forms and concentrations. This includes
final pH, amino acid product, amino acid final concentration, dextrose Inorganic phosphates injections available in the US market contain a
final concentration, the calcium salt, the phosphate salt, temperature, mixture of monobasic and dibasic salts of either sodium or potassium.
sequence of calcium and phosphate addition, time that the PN is stored The sodium phosphates injection contains monobasic sodium phos-
prior to administration, and other factors.112–114 phate and monohydrate and dibasic sodium phosphate, anhydrous.
Similarly, the potassium phosphates injection contains a mixture of
monobasic and dibasic potassium phosphate, anhydrous. All sources of
pH potassium or sodium phosphate (ie, amino acids) must be taken into
consideration, as high concentrations of phosphate increase the risk of
The final pH of the PN admixture is the most important factor in precipitate formation with calcium. Although the use of a single calcium
determining calcium and phosphate compatibility. Inorganic salts (such phosphate product has been advocated for use in predicting compati-
as calcium chloride, monobasic/dibasic phosphates) are more likely bility, this approach is fraught with error as the results from multiply-
to dissociate into free ions compared with organic salts (eg, cal- ing the PN calcium concentration (in mEq/L) with PN phosphorus con-
cium gluconate, sodium glycerophosphate). The extent of this dis- centration (in mmol/L) vary inconsistently as the calcium concentration
sociation is largely governed by the pH of the final formulation.112 decreases and phosphorus concentration increases.117
In aqueous solutions, phosphate may exist in three anionic forms— Sodium glycerophosphate is an organic form of phosphorus avail-
monobasic (H2 PO4 – ), dibasic (HPO4 2– ), and trivalent (PO4 3– )—and all able in Europe and has been temporarily available for use in the
three forms can produce insoluble calcium phosphate complexes.115 United States during phosphorus drug shortages. In contrast to inor-
Because extreme alkalinity is generally required to ionize the tribasic ganic phosphate injections, the phosphate component of sodium glyc-
phosphate species, it is not normally found in PN admixtures. Although erophosphate is covalently bound to a glycerol backbone, which hin-
the trivalent phosphate is not expected in PN, the dibasic species read- ders the formation of a precipitate with divalent calcium ions. The
ily forms an insoluble complex with calcium that crystallizes and precip- availability of a monobasic potassium phosphate product in Canada
itates out of solution. The concentrations of the monobasic and dibasic enhanced calcium and phosphate solubility to allow for a greater con-
are pH dependent, and a small change of 0.05 in pH can increase the centration in a PN admixture, although the aluminum content was not
dibasic phosphate salt concentration by >10% and contribute to the described.118
precipitation of dibasic calcium phosphate (CaHPO4 ).115 Essentially,
as the pH of the final admixture increases, there is more dibasic phos-
phate present to bind with free calcium. Conversely, the lower the final Calcium salts
admixture pH, the greater the percentage of the monobasic (H2 PO4 – )
salt at which it can form more soluble calcium dihydrogen phosphate Calcium gluconate salts are recommended over calcium chloride salts
salt (Ca[H2 PO4 ]2 ). when compounding PN because of superior compatibility with inor-
ganic phosphates. Because the gluconate salt of calcium dissociates
much less extensively in water compared with calcium chloride, it is the
Amino acids preferred salt for use in PN admixtures. Calcium chloride almost com-
pletely dissociates in aqueous solutions, so precipitation with phos-
Amino acid products from different manufacturers can vary in pH, phate occurs at much lower chloride salt concentrations vs gluconate
ranging from 5 to 7. Amino acid products also have an intrinsic buffer- salt concentrations. One drawback associated with calcium gluconate
ing system, largely determined by the concentrations of arginine, his- injections in glass vials is that it is heavily contaminated with alu-
tidine, and lysine. Higher final concentrations of amino acids have a minum, so the higher aluminum load must be considered, especially
greater buffering capacity, and therefore, there is less of an increase in when used in neonatal PN admixtures.112 Of note, calcium chloride is
pH when phosphate is added to the final formulation. The more moder- the salt included in commercial, standardized, multichamber PN prod-
ate pH will support calcium and phosphate solubility. High concentra- ucts with electrolytes. Compatibility testing has demonstrated that cal-
tions of amino acids have also been reported to sequester calcium and cium chloride is physically compatible with sodium glycerophosphate
decrease its reaction with phosphates. Lysine has been identified as the in pediatric PN formulations.119 Thus, compounding with sodium glyc-
amino acid most likely to complex with calcium, and the degree of that erophosphate and calcium chloride provides a mechanism for reducing
complexation is also pH dependent. Better determination of safe PN the aluminum load in PN.
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 287
15

Dextrose concentration take up to 24–48 h.112,113 The variable rates in precipitate forma-
tion have been attributed to slow crystallization from supersaturated
The USP monograph for dextrose injections indicates that these prod- mixtures.117
ucts are generally acidic, with a pH range between 3.2 and 6.5. Thus,
higher concentrations of dextrose will lower the final admixture pH,
providing a greater percentage of monobasic phosphate (H2 PO4 – ), Admixture compatibility—Focus on trace elements
which can form more soluble calcium dihydrogen phosphate salt and vitamins
(Ca[H2 PO4 ]2 ).117
Shortly after the initial American Medical Association (AMA) published
recommendations on parenteral trace elements, a study evaluated the
Temperature compatibility of individual trace elements (chromium chloride, cop-
per sulfate, manganese sulfate, zinc sulfate), as well as the combina-
Temperature may influence the dissociation of organic calcium salts, tion of the four trace elements, while accounting for metal contami-
such as calcium gluconate. As the temperature rises, there is a greater nants in all components used.121 The solutions tested included amino
dissociation, rendering more free calcium ions available to complex acids alone (Travasol 8.5% [pH 5.7]) and a PN formulation (Travasol
with phosphate salts. This phenomenon is not associated with inor- AA4.25%-D25% [pH 5.75]) in glass with electrolytes and multivitamins.
ganic calcium chloride salts, as they are almost fully ionized in aqueous Over the 48-h study period, the initial concentration of each trace ele-
solution. A temperature change from 5◦ C (41◦ F) to body temperature ment was unchanged, based on atomic absorption spectrophotometry,
at 37◦ C (98.6◦ F) has been shown to promote formation of calcium and infusion through a 0.22-μm filter did not reveal any visible precip-
phosphate crystals.120 Precipitation from temperature increases itate. Of note, each individual trace element led to some slight color
may also be related to the shift in the phosphate equilibrium from change when admixed with amino acids alone. The incompatibility of
monobasic to dibasic salts.112 copper with some amino acids, resulting in precipitates, was described
in an earlier section.112
In a study of pediatric PN (Vaminolac AA2.75%-D5.64%) that
Sequence of calcium and phosphate additions included calcium chloride and glycerophosphate, the addition of multi–
trace elements (Pediatrace) led to an unexpected ongoing precipitate
The order of mixing calcium and phosphate into PN admixtures within 4 h of admixture, which amounted to ∼14,000 particles/ml,
can affect the final solubility profile. Inorganic phosphate injections mostly smaller than 1 micron.122 The fine subvisual particles were
should be added early in the mixing sequence, and calcium glu- noted using Tyndall light, light obscuration, and turbidimetry and pro-
conate injections should be added nearly last to what should be posed to be the result of incompatibility between copper and cysteine.
the most dilute phosphate concentration possible. The official pH This was not analyzed further, and additional factors such as pH, con-
range of calcium gluconate injection is 6.0–8.2, and the formulation taminant minerals, redox conditions, mixing sequence, temperature,
is more likely to be mildly alkaline than acidic. Thus, it is reason- and container type would need to have been taken into account.
able to add calcium gluconate at the end of the sequence, so it is Another study evaluated the compatibility of copper in two
most dilute and least likely to influence the overall pH of the for- cysteine-containing pediatric PN admixtures (TrophAmine AA2.4%-
mulation. Increasing hypertonicity or osmolarity of the PN formula- D25% plus cysteine 960 mg/L and copper 0.3 mg/L, and AA3.7%-D16%
tions, such as those infused into central veins, may also improve cal- plus cysteine 1480 mg/L and copper 0.45 mg/L) over a 24-h simulated
cium and phosphate solubility. High concentrations of amino acids infusion through infusion sets with 0.22-μm inline filters.123 Aside from
may sequester calcium and decrease its complexation with phosphate visual signs of incompatibility, including precipitation, samples were
salts, whereas higher concentrations of dextrose can reduce the final obtained at several time points throughout the infusion and assayed
admixture pH.115 for both cysteine and copper. The first solution exhibited >10% reduc-
tion in cysteine and in copper, whereas the second solution had an
8% reduction in cysteine concentration and 11% reduction in cop-
Duration of storage per concentration over the study period. However, no visible partic-
ulate matter or discoloration was observed on the filters. Previous
The duration of time lapsed after compounding has been shown to work had noted cupric sulfide precipitates that discolored inline fil-
impact the process of precipitation. Although precipitates may form ters when using an adult bisulfite-free amino acid product (Novamine
during the compounding process because of poor mixing procedures, it AA4.25%) plus cysteine (200 mg/L) and copper (0.88 mg/L).124 Switch-
is more typical for dibasic calcium phosphate crystallization to progress ing to a bisulfite-containing amino acid product resulted in no pre-
through a time-related induction period. Precipitation of calcium phos- cipitate formation.124 A study to examine mechanisms compared a
phate during PN infusion may not be observed until 12 or more hours bisulfite-containing but cysteine-free amino acid product (Travasol)
after preparation, and a delayed onset of precipitate formation may with a cysteine solution (400 mg/L), following the addition of copper
288
16 BOULLATA ET AL .

(1.2 mg/L), and noted significant (98%) reduction of copper concentra- bind to plastic containers and infusion sets, but over 65% of the content
tion only in the cysteine solution.125 is expected to remain available.131 It remains unclear whether there is
Early studies suggested that insoluble iron phosphate precipitation a significant difference between ergocalciferol and cholecalciferol.
can occur in stored PN solutions devoid of multivitamins.42 The mech-
anism is unclear but appears to be inversely related to the amino acid
concentration and the duration of storage prior to the addition of the STABILITY
multivitamins. Amino acid brand may also play a role with precipitates
seen more with Synthamin (identical to Travasol at the time).42 No sim- Focus on macronutrients (alone or combined)
ilar reports have been noted using other amino acid products available
in the United States. Amino acids
Others have reported incompatibility of iron dextran with lower-
volume neonatal PN solutions. Incompatibility may be related to the Amino acid degradation may be general or specific to an amino acid side
final amino acid concentration, although it is uncertain whether the chain. General degradation can generate ammonia and carbon dioxide
incompatibility is due to the pH or related to the nature of the amino through deamidation/deamination in the presence of elevated temper-
acid solution used in the PN. In their investigation, Mayhew et al evalu- ature. The amino acid L-glutamine is excluded from products because
ated 20 neonatal PN admixtures that varied in their amino acid concen- of rapid degradation to toxic by-products upon heat sterilization. The
trations (TrophAmine 0.5%–2.5%) and otherwise contained dextrose presence of oxygen and/or light will also influence amino acid stabil-
10% as well as micronutrients, heparin and cysteine.126 Iron dextran ity. Higher degradation occurs in plastic than in glass containers, likely
was included at 10 mg/L and control solutions omitted either the iron, associated with greater oxygen permeability.
trace elements, or multivitamin component and all were stored at 19◦ C Nonionizing radiation (eg, ultraviolet or visible light) may cause
for up to 48 h and observed for precipitate, color change, turbidity, and some minimal decomposition but, in the presence of a chromophore
phase separation. Iron precipitate (rust colored) formed within 12–24 (functional group or substance), may generate oxidizing species (eg,
h in those PN admixtures with ≤1.5% amino acids. The authors con- hydrogen peroxide), which can further degrade amino acids. Most sus-
cluded that lipid-free neonatal PN solutions with a final amino acid con- ceptible to photo-oxidation are cysteine, histidine, methionine, pheny-
centration of 2% or greater were visually compatible with iron dextran lalanine, tryptophan, and tyrosine. Reactive side chains of cysteine,
10 mg/L for 48 h. These findings suggest that adding iron dextran to a tryptophan, and tyrosine make them the most susceptible to oxidation.
peripheral PN solution, which typically has a lower final concentration Photo-oxidation of amino acids (eg, tryptophan, tyrosine) is enhanced
of amino acids, would be less stable and not recommended.126 in the presence of riboflavin (a chromophore). A study of a 1% concen-
In a more recent retrospective study, Lee et al evaluated the safety tration of pediatric amino acids product in the presence or absence
of mixing low-MW (LMW) iron dextran in a cohort of 89 pediatric of riboflavin and metabisulfite (an antioxidant preservative) exposed
patients (0–21 years of age) who received PN containing a mainte- to phototherapy light (425–475 nm wavelength) generated hydrogen
nance dose of LMW iron dextran with a total of 2774 days of exposure peroxide.132 Histidine, methionine, and tryptophan undergo photo-
(1–196 days per patient).127 The mean dose of iron dextran in children oxidation in ambient light in the presence of riboflavin with some
decreased with increased weight from <5 kg (0.21 ± 0.05 mg/kg/day) protective effect in the presence of adequate ascorbic acid.133,134
to ≥40 kg (1.9 ± 0.5 mg/day). The authors concluded that PN contain- Tryptophan is an interesting case because multiple oxidation reac-
ing a maintenance dose of LMW iron dextran could be safely admin- tions influence this amino acid with several different degradation
istered to hospitalized children with respect to anaphylaxis risk, and products. The indole moiety of tryptophan is prone to ring opening
further studies would be needed to evaluate the potential to prevent from oxidation with electrophilic substitution. The degradation of
iron deficiency anemia and the need for additional IV iron infusions.127 tryptophan over time may be associated with yellowing of the amino
Unfortunately, an evaluation of risks due to potential physicochemical acid solution.135 An AA4.25%-D25% solution stored for 2–4 weeks
interactions was not performed. did not experience significant (>10%) degradation of amino acids,
The multivitamin products are compatible in dextrose 5% in including tryptophan, the most labile component, especially under
water, 0.9% sodium chloride, and PN admixtures according to the refrigeration (4◦ C).100,101
manufacturers.84–87 The compatibility is contingent on the individ-
ual vitamins. The vitamins may react with each other, macronutrients,
excipients, or the PN bag material.112 For example, vitamin A has long Lipid emulsions
been known to adsorb to plastic materials, making it less available for
the patient.112 Sorption to the bag (ie, adsorption) is more likely to When the ILE is incorporated into the PN admixture, the entire prepa-
occur with retinol acetate than with retinol palmitate because of its ration becomes an oil-in-water emulsion in character. As a result, any
greater affinity to plastics.128 This has become less of a concern with evaluation of PN admixture stability needs to consider the physical
the use of PVC-free bags and administration sets and the switch from integrity of the emulsion. In general terms, the final macronutrient con-
the acetate ester to the less reactive palmitate ester.129,130 As a dif- centrations that are expected to provide the best likelihood of a sta-
ficult vitamin to incorporate into parenteral products, vitamin D may ble emulsion in the PN admixture, for up to 30 h (at 25◦ C) or 9 days
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 289
17

(at 5◦ C), are amino acids ≥4%, dextrose ≥10%, and ILE ≥2%.4 This L1.6% Intralipid), which included electrolytes, multi–trace elements,
recommendation was based on data using 100% soybean-oil ILE prod- and multivitamins, was performed (pH of 5.5 with osmolality ∼1472
ucts and varies with ILE products containing other source oils. mOsm/kg) over 14 days at 4◦ C plus 2 days at 22◦ C using visual obser-
Extemporaneously prepared TNAs (ie, 3-in-1 PN admixture) are vation, light microscopy, electron microscopy, and Coulter counter
expected to be less stable than the ILE product. The compendia PFAT5 techniques.138 These revealed an increase in particle size over time,
limit of 0.05% might be too strict; hence, the use of PFAT5 <0.4% including some exceeding 2 and 5 μm in diameter. The amino acid and
(one log higher) has been proposed as the acceptance criterion, even dextrose content of a TNA alone is unlikely to be adequate to overcome
though studies do indicate that it is possible to compound a PN formu- the influence of high divalent ion concentrations in a PN admixture.139
lation that fulfills the PFAT5 limit of USP. Driscoll et al demonstrated The multivalent cations such as calcium and magnesium, as well as
that when the PFAT5 level exceeded 0.4%, obvious phase separation or high concentrations of the multivalent copper, iron, and zinc, have the
“cracking” occurred, reflecting the irreversible terminal stage of emul- potential to alter the stability of ILE by reducing the negatively charged
sion instability.114 repulsive forces that keep lipid droplets separated. A recent study eval-
An evaluation was performed of a pediatric amino acid product uated the influence of trace elements (copper 200 mcg, fluorine 570
(TrophAmine 10%) in a TNA using ILE 20% (Intralipid, Liposyn II, mcg, iodine 10 mcg, manganese 10 mcg, selenium 60–90 mcg, zinc 3.4–
or Nutrilipid) and dextrose 70% with micronutrients at several final 3.5 mg) as well as carnitine (100 mg) on the stability of six different 1-L
macronutrient concentrations for 24 h at 4◦ C and an additional 24 h PN admixtures (TrophAmine AA2.3-3%–D10-14%–L1.9-2.3%; various
at room temperature.108 Although evidence of creaming was observed ILE products used) for pediatric patients that included electrolytes and
in all sample PN admixtures over time, no coalescence or phase sepa- multivitamins in EVA bags.140 These admixtures were stored at 4◦ C
ration was visible. Particle size analysis revealed that mean diameters or room temperature for up to 96 h with 450 samples analyzed. Lipid
remained <0.4 μm; however, greater numbers of large lipid particles droplet diameters were evaluated by laser diffraction. The diameter
(>5 μm) were present with higher concentrations of electrolytes, rep- of droplets did not change significantly throughout the duration of the
resenting as much as 0.6% of oil droplets.108 This value is well above study and remained between 0.4 and 1 μm in most of the tested admix-
the current compendia PFAT5 limit of 0.05% or the proposed limit of tures. An interesting exception occurred in those admixtures using a
0.4%, indicating an unstable emulsion. fish-oil component with calcium concentration of 4.5 mmol/L (9 mEq/L)
PN admixture stability is determined by how well lipid droplets or higher in which 2% of droplets exceeded 5 μm and 12% exceeded
remain dispersed in the external (aqueous) phase, as cations and lower 1 micron immediately after compounding.140 Whether the inclusion of
pH values decrease the electric charge of the emulsifier tasked with the trace elements increased this risk is not known because no controls
preventing aggregation. With the advent of TNAs, studies emerged without trace elements (or carnitine) were studied.
that described the influence of trace elements on emulsion stabil- Compounded PN admixtures for home infusion may rely on a dual-
ity. One study compared organic (gluconate) with inorganic (chlo- chamber EVA bag in which the ILE portion is stored in one chamber
ride) salt forms of trace elements (copper 240 mcg/L, iron 0.5 mg/L, while the remainder of the PN ingredients are in the other for a 7-
zinc 2 mg/L) with a PN admixture (Azonutril-25 AA2%-D17.5%-L5% day period. In this way, any concern for the final emulsion’s stability
Intralipid) stored at 4◦ C or 25◦ C for up to 1 week.136 Lipid particle mea- over time is deferred. Just prior to administration, the contents of both
sures relied on Coulter counter analysis capable of including diameters chambers are combined with the addition of multivitamins at that time.
from 1 to 25 μm. The gluconate salts did not significantly alter admix- However, the emulsion stability of the final PN admixture must still be
ture pH, whereas chloride salts decreased pH slightly over the course ensured. Varying contents and component concentrations will deter-
of the study. Although the numbers and sizes of lipid droplets were not mine which PN admixtures for use at home remain stable and safe for
significantly different between them, those in the PN admixture with administration and which do not.141,142
the chloride salts had more broadly dispersed values over time risking In the smaller volumes of neonatal PN admixtures, the influence
tendency towards emulsion destabilization. of trace elements and vitamins were evaluated on formulation sta-
In an early study of emulsion stability of a PN admixture, the influ- bility in a practice setting where trace elements and vitamins were
ence of two different multimineral solution additives was evaluated.137 routinely not combined in the same bag.143,144 Three PN admixtures
The PN admixture (Travasol AA3.34%-D12.5%-L3.34% Intralipid) in were evaluated—one with multivitamins but without multi–trace ele-
large ethylene–vinyl acetate (EVA) bags included either a commercial ments, another with the trace elements but not multivitamins, and the
additive (Addamel) or a compounded eight-component additive, but third containing all components.143 The formulations were otherwise
no vitamins, stored for up to 2 weeks at 4◦ C. Of note, Addamel is not identical (Primene AA3%-D8%-L3% Lipofundin) in EVA bags with elec-
currently approved for use in the United States and is only marketed trolytes that included organic phosphate. Samples were taken at pre-
elsewhere. The lipid droplet size, determined by Coulter counter (able determined points for up to 7 days, with storage conditions at 5◦ C,
to assess particle diameters between 0.6 and 20.2 μm), did not vary 25◦ C, and 40◦ C, for evaluation of emulsion stability using zeta poten-
significantly in distribution during storage, nor did the proportion of tial, optical microscopy, dynamic light scattering, and PFAT5 . The osmo-
large (>5 μm) to small (<2 μm), with mean droplet size of ∼0.674– lality of the PN admixtures was slightly higher than 1000 mOsm/kg.
0.684 μm, all of which was no different than the control of the ILE Slight creaming and a darkening color on visual inspection were appar-
alone.137 A study of a 3.1-L PN admixture (Vamin-N AA3.4%-D16%- ent after 48 h in admixtures stored at 25◦ C and 40◦ C.143 Zeta potential
290
18 BOULLATA ET AL .

values and pH did not change significantly throughout the study period HPLC method, ascorbic acid concentrations had diminished to 67% of
for any formulation, nor did lipid droplet diameter.143 Most impor- baseline when no copper was present but decreased to 33% of baseline
tantly, none of the formulations exceeded a PFAT5 of 0.05%.143 in the presence of copper by 24 h. The generation of the ascorbic acid
Iron, in either the bound ferric (Fe3+ ) or free ferrous form (Fe2+ ), can degradation product oxalic acid was not quantified in this study.
generate free oxygen radicals, which has raised concerns regarding its Both ascorbic acid and copper have the potential ability to reduce
safety with IV administration. This is especially true in the presence of selenite to the poorly soluble elemental selenium.151 This may be more
ILE, which serves as an excellent substrate for iron-induced peroxida- likely at ascorbic acid concentrations >100 mg/L in PN admixtures with
tion because of the large number of double bonds present in the unsat- pH of 5 or lower or at ascorbic acid doses above 500 mg, and it may be
urated fatty acids.145 Despite its potential benefits, iron, especially the less likely in well-buffered admixtures with pH >5.151–157
dextran form, should not be added to ILE or TNAs, as it can result in a After selenium was recognized as a critical trace mineral to include
destabilization of the emulsion.146 Similar to divalent cations (ie, cal- in PN admixtures, several studies evaluated its stability, in particular
cium, magnesium), trivalent cations such as iron may cause a decrease given the potential risk for reduction to insoluble elemental selenium
in the surface potential of the lipid droplets, destabilizing the nega- by ascorbic acid. When only ascorbic acid (100 mg/L) and selenious
tive surface charge between lipid particles, resulting in aggregation and acid (100 mcg/L) were combined in solution, a 90% loss of selenium
coalescence of the smaller lipid particles to form larger ones.114,147 occurred by 24 h, with higher concentrations (500 mg/L) of ascorbic
These larger lipid globules can be potentially dangerous if infused, as acid responsible for near-complete loss of selenium within 1 h using a
they can result in fat emboli, especially in neonates requiring the iron. In thin-layer electrophoresis method.155 The incorporation of selenious
a study involving the necropsy findings of nearly 500 live-born infants acid into a PN admixture (Travasol AA4.25%-D25%) that included elec-
(including 30 of 41 PN patients who had received ILE), Puntis and Rush- trolytes, a product with four trace elements, and multivitamins with
ton found intravascular lipids in the small pulmonary capillaries of 15 100 or 500 mg of ascorbic acid revealed minimal selenium loss at the
infants receiving ILE that were not found in the necropsy of enterally lower amount but over 10% loss with ascorbic acid 500 mg at 24 h.
fed infants.148 This group had received significantly more ILE during The lower losses in the PN admixture were attributed to the amino acid
their PN course (in total amount [grams per kilogram] and duration content of the admixture and its influence on overall pH. Of note, other
[number of days]) compared with another group of 15 infants receiv- studies either did not determine the chemical form of selenium or did
ing ILE who did not have lipid staining.148 not evaluate the interaction using a PN admixture.152–154
When added to a 1.85-L PN admixture (Travasol AA5%-D14.3%-
L5.7% Intralipid) with electrolytes, multi–trace elements, and multi-
Focus on micronutrients (alone or combined) vitamins in an EVA bag, the inclusion of iron dextran (100 mg) was
associated with repeated filter obstruction during infusion accompa-
Trace elements nied by a yellow-brown fluid layer floating on top of the mix.146 That
supernatant contained much higher lipid concentrations than expected
Trace elements may interact with other micronutrients. For example, from the homogeneous mix. This was not repeated with an identical PN
copper and ascorbic acid interact with each other. Copper can accel- admixture, which excluded the iron dextran. When much lower concen-
erate the degradation of ascorbic acid in solution, especially at pH >4. trations of iron dextran (2 mg/L) were incorporated into simulated 0.5-
Conversely, higher concentrations of ascorbic acid can reduce copper L PN admixture (Travasol AA0.55-2.2%–D0.4-10%–L2-4% Intralipid or
to the cuprous form, which is why ascorbic acid content of adult multi- Liposyn II) in glass bottles without added electrolytes, trace elements,
vitamins was reduced to 100–200 mg from 500 mg. or vitamins and stored at 4◦ C or 25◦ C protected from light for 48 h,
A study evaluated trace element stability in a 1000-ml PN admix- no visually apparent precipitation or phase separation was noted com-
ture (Travasol AA3%-D18%) in a DEHP-free but nevertheless semiper- pared with controls.158 Photon correlation spectroscopy suggested
meable bag, using an additive of six multi–trace elements and multi- that 90% of lipid droplets were smaller than 1.3 μm, although PFAT5
vitamin with high ascorbic acid (1000 mg) content.149 Using ICP-MS, was not performed.
trace element concentrations were reported to decline over time (36 h In one investigation evaluating the stability of iron sucrose added to
or 30 days) with storage (at 4◦ C or 20◦ C protected from light). Of the lipid-free PN containing pediatric amino acids (TrophAmine AA2.6%-
trace elements formulated into the PN admixture, three (Cu, Mn, Zn) D20% [pH 5.7]; TrophAmine AA3.5%-D20% [pH 5.5]) with electrolytes
decreased significantly in concentration over the storage time, even and trace elements, it was shown that the physical stability of iron
when refrigerated. sucrose in PN is time and concentration dependent, based on visual and
The influence of copper on ascorbic acid stability was studied in 1-L microscopic inspection.159 Iron sucrose concentrations up to 2.5 mg/L
adult PN admixtures (AA5%-D25%) containing electrolytes and mul- in lipid-free PN compounded with neonatal-range amino acid concen-
tivitamins (100 mg ascorbic acid), with or without trace elements (1.2 trations and cysteine were physically stable. Iron sucrose concentra-
mg copper) in PVC bags at room temperature.150 Samples were taken tions of ≥10 mg/L were physically unstable, with particulate formation
at time points from baseline up to 36 h to assay ascorbic acid con- within 12 h and evidence of gross physical incompatibility within 24
tent (using both spectrophotometry and high-performance liquid chro- h.159 In the PN admixture containing a lower final amino acid concen-
matography [HPLC] with ultraviolet detection). Based on the preferred tration, visible particulates manifest by 8 h at the highest iron concen-
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 291
19

trations and within 24 h for all lower iron concentrations, with crystals manner.173 It should be noted that combining the amino acid and dex-
determined to be iron complexes including calcium and phosphate.159 trose solutions results in a decreased sulfite concentration, minimizing
the risk of thiamin degradation.174
A study conducted by Scheiner et al found that 93% of the initial thi-
Vitamins amin amount remained after 24 h in a PVC bag containing a dextrose-
electrolyte solution (Normosol M-D5%) and multivitamins, with a pH of
The stability of multivitamin solutions depends on the individual com- ∼5 (pH range, 4.0–6.5; 363 mOsm/L) and storage temperature of 23◦ C
ponents. The instability of vitamins can be due to oxidation, pho- in the presence of fluorescent light.173 The labeling of the Normosol M-
todegradation, or exposure to container materials.160 The main fac- D5% solution indicated bisulfites as an ingredient, but they were not
tors affecting the stability of the individual vitamins and the excipients detected. The authors concluded that the increased oxygen permeabil-
include light exposure, pH, temperature, and PN bag material (eg, plas- ity of the PVC bag (as compared with the glass container) accelerated
tic or glass).112 Light-induced peroxidation reactions may occur with the loss of sulfites, thereby decreasing the potential for destruction of
polysorbate 80 and 20.92 This reaction is enhanced at higher tempera- thiamin.173 When Normosol M-D5% with multivitamins was stored in
tures and upon exposure to air.161 a glass container (under the same room temperature, pH, and light con-
When exposed to ultraviolet light, riboflavin has been shown to cat- ditions), the amount of thiamin decreased to 51% relative to the origi-
alyze oxidation reactions.162,163 Riboflavin can act as a photosensitizer nal amount after 24 h.173 Only 3% of the initial thiamin remained in an
initiating production of reactive oxygen species or directly reacting undiluted FreAmine III 8.5% glass-bottle solution consisting of a high
with other components (eg, amino acids, fatty acids).164–166 Ultravio- concentration of bisulfites and with a pH of 6.5.173 A later study by
let light activates riboflavin-catalyzed oxidation reactions with some Smith et al revealed that thiamin degradation was the greatest in 2-in-1
amino acids.133 Exposure to 24 h of ultraviolet light was associated and 3-in-1 PN admixtures (containing electrolytes, trace elements, and
with a statistically significant decrease in the methionine, proline, tryp- multivitamins without vitamin K) with a final bisulfite concentration of
tophan, and tyrosine concentrations in a 2-in-1 PN admixture con- 3 mEq/L and pH of 6.4 at a temperature of 25◦ C compared with other
taining a riboflavin concentration of 1 mg/dl.133 Riboflavin itself can solutions at various pH and bisulfite concentrations.175 Bowman et al
be degraded to luminoflavin, luminochromo, and other compounds found that no thiamin degradation was detected for 22 h in a 1-L PN
in the presence of oxygen and light, including sunlight and artificial admixture (AA4.25%-D25% with sulfite 0.05%) contained in a plastic
light.167–169 The concentration of riboflavin in a 2-in-1 PN admixture bag.174 The PN bags were stored at a temperature of 30–31◦ C, and the
decreased by 47% and 100% upon exposure to 8 h of indirect and direct average pH of the PN solution was 5.8.174
sunlight, respectively.170 The main determinants of thiamin degradation are the presence of
PN admixtures with pH values above 5.0 reduce the risk for pre- sulfites and direct sunlight. Refrigeration has been shown to slow thi-
cipitation of folic acid.171 Results from stability studies are conflict- amin loss.173 When exposed to direct sunlight, the thiamin concentra-
ing with regards to the effects of storage container, light exposure, tion in a 2-in-1 PN admixture decreased by 26% after 8 h.170 But in
and temperature.112 In an early study evaluating the safety of vita- a sulfite-free 2-in-1 PN admixture remaining refrigerated until use, at
min addition to PN, Chen et al (using a microbiological assay, which least 75% of a large thiamin dose (50 mg) was maintained at 28 days.176
was standard at the time for several B vitamins) found that folic acid Patients should be monitored for signs and symptoms of thiamin defi-
was stable in a 1-L PN admixture (Travasol AA4.25%-D25%) contain- ciency if there is a concern for thiamin loss from the PN admixture.
ing electrolytes, trace elements, and a nine-vitamin formula plus 1 mg Ascorbic acid is the vitamin most susceptible to degradation, under-
folic acid when exposed to fluorescent light, indirect sunlight, or direct going reversible oxidation to dehydroascorbic acid catalyzed in part by
sunlight at room temperature for 8 h.170 When the PN was stored trace elements (ie, copper, manganese, zinc), which can then undergo
frozen (−40◦ C) for 5 weeks and then refrigerated (4◦ C) for another 2 hydrolysis to diketogulonic acid.112,177,178 The latter undergoes irre-
weeks, folic acid maintained stability.170 This study also revealed that versible oxidation to several metabolites, including oxalic acid.112 Of
riboflavin, pyridoxine, and thiamin were unstable in sunlight (direct or note, oxalic acid can interact with calcium, forming a poorly solu-
indirect) over that 8-h period.170 Precipitation forms at a folic acid con- ble complex with risk for precipitation. This decomposition of ascor-
centration of 0.2 mg/L in the low pH, at a high concentration of dextrose bic acid is dependent on the content of oxygen available in the PN
40% at room temperature, and under refrigeration.172 Direct sunlight admixture from bag permeability, dissolved air in solution, and other
was found to degrade pyridoxine by 86% in a 2-in-1 PN admixture after additives.177 The degradation is more likely at pH >4 and at higher
8 h.170 temperatures (22–35◦ C).179,180 The oxidation reactions are primarily
Thiamin degradation is dependent on several factors such as the sul- due to the introduction of oxygen into the PN admixture during the
fite concentration, pH, temperature, exposure to direct sunlight, and compounding process, through permeation of the PN container, or dur-
presence of oxygen.112 The stability of thiamin is less of an issue now ing infusion.112,162,181 Smith et al found that ascorbic acid degrada-
that bisulfites are rarely included as an antioxidant/preservative in tion was more pronounced in plastic containers compared with glass
other PN components (eg, amino acid solutions).112 The irreversible containers.175 The use of multilayered bags to reduce oxygen perme-
degradation of thiamin in solution into a thiazole and pyrimidine by ability has been recommended to reduce ascorbic acid degradation,
sodium metabisulfite proceeds in a concentration- and pH-dependent with the caveat that they may trap oxygen transferred into the bag
292
20 BOULLATA ET AL .

during filling. The monolayered EVA bag is more permeable to oxy- photo-oxidation that is dependent on wavelength and light intensity
gen than a multilayered bag, which allows for a greater potential for as well as the oxygen content.188 Bags that prevent oxygen perme-
reactivity with ascorbic acid.181 Inclusion of cysteine, by complexing ability or light-protective covers may each improve the stability of
with copper, may reduce the degradation of ascorbic acid.182 However, vitamin E.128,187,189 The potential degradation of retinol and vitamin
cysteine has a reducing capacity able to irreversibly degrade ascorbic E during lipid peroxidation in a PN admixture may be less when MCT
acid/dehydroascorbic acid.183 oil accompanies soybean oil, compared with ILEs of a soybean-olive oil
Vitamins may also interact with other micronutrients. A specific combination or soybean-oil ILE alone.190 More has yet to be learned
evaluation of several B vitamins using nuclear magnetic resonance about the stability of different vitamin E isomers. Vitamin K content
spectroscopy evaluated the interaction within this group and with of the PN admixture is susceptible to photodegradation regardless
other micronutrients.144 Interactions between the vitamins, especially of the presence of lipids.128,191 Exposure to daylight decreased the
pyridoxine, reflect proton exchange phenomena as opposed to inter- phytonadione concentration in PN admixtures (2-in-1 and 3-in-1) by
molecular reactions. Pyridoxine and riboflavin are sensitive to the pres- 50% after 3 h.128
ence of electrolytes—especially pyridoxine in the presence of calcium Given the above, multivitamin solutions are the least stable compo-
gluconate and phosphate—and, together with magnesium sulfate, may nent in PN admixtures, especially when exposed to oxygen and light.112
suggest some self-aggregation of pyridoxine. Divalent cations (calcium, For this reason, dispensing a 7-day supply of PN admixtures at a time
magnesium, and copper and zinc as chloride salts) may also lead to precludes multivitamin content and requires the patient/caregiver to
some self-aggregating precipitate of riboflavin, although the presence add the dose each day just prior to administration. Once the multivita-
of nicotinamide seems to inhibit riboflavin aggregation. Notwithstand- mins are added to the compatible diluents, the final admixture should
ing these observations, hydrophilic vitamins and minerals in the same ideally be administered immediately in the absence of photo-oxidative
PN admixture could be stable for up to 48 h.144 protection and completed within 24 h.84–87 The vitamin-containing PN
Studies investigating the effect of riboflavin on ascorbic acid oxi- admixture should be refrigerated when administration is delayed.84–87
dation and in combination with ILE are conflicting. The manufacturers To prevent degradation due to light exposure during administration, PN
recommend against the direct injection of multivitamins into ILE.84–87 bags should be wrapped with an opaque protective covering.192 Also,
In the presence of ultraviolet light, riboflavin catalysis of ascorbate orange or yellow infusion tubing may be used to prevent light pene-
to dehydroascorbate, generating hydrogen peroxide and free radicals, tration through the tubing during administration.192 The practice of
has been described in the literature.162,163 The hydrogen peroxide may photoprotection in the United States remains dependent on product
be the result of lipid and/or ascorbic acid oxidation.163 Laborie et al availability.193
found that ILE containing riboflavin (0.11 mmol/L) and ascorbic acid
(13.6 mmol/L) had higher amounts of peroxide production compared
with ILE alone or with riboflavin only.163 Contrary to the Laborie et al OTHER
results, Silvers et al found that the hydrogen peroxide concentration
decreased by 40% in ILE with multivitamins compared with dextrose Aside from the common nutrient components of the PN admixture,
5% in water–0.45% sodium chloride with multivitamins in an ambient additional compatibility and stability issues arise with other com-
light environment.162 The authors noted that oxidation of ascorbic acid ponents. These include medications added to or co-infused with a
still occurred but that ascorbic acid protected against lipid peroxida- PN admixture, as well as the materials found in automated com-
tion. The major source of light and oxygen exposure occurred during pounding device circuits, the PN bag, administration sets, and inline
the infusion of the admixture through the catheter. Furthermore, the filters.
opaque color of the ILE likely attenuated light-induced oxidation of
ascorbic acid.162 Silvers et al suggested that the findings by Laborie et
al were based on unreliable assay methods. Considering the beneficial Medication
effect of ascorbic acid on ILE, the precaution against direct injection
of multivitamin solutions into ILE may be unnecessary.162 Conflicting As is already apparent, PN admixtures are complex from a compat-
with previous findings, Dupertuis et al found that multivitamins con- ibility and stability perspective when accounting for all the nutrient
taining riboflavin did not influence the degradation rate of ascorbic acid components—active ingredients and excipients. The consideration to
in 3-in-1 PN admixtures exposed to daylight.181 To prevent the possi- include a nonnutrient medication in a PN admixture is to be undertaken
bility of ascorbic acid loss induced by riboflavin, PN admixtures should carefully with serious attention to compatibility and stability concerns.
be protected from light when practical. This is true whether preparing a PN completely from individual com-
Retinol is the most light-sensitive vitamin, with extensive pho- ponents or starting with a commercial, standardized, multichamber PN
todegradation depending on the wavelength and intensity of product. Similar attention needs to be given when considering medica-
light.169,184,185 The presence of ILE in the PN admixture is thought tion administration by Y-site infusion along with a concurrently infus-
to reduce the degree of vitamin A instability, but this remains ing PN admixture. An evidence-based guidance document discussed
controversial.128,175,185,186 The use of light-protective covers has the safety of using the PN admixture as a vehicle for nonnutrient medi-
been suggested.185,187 Less reactive than retinol, vitamin E undergoes cation delivery along with a summary of published data.4 Although that
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 293
21

can be referred to for further detail, the following provides a concise Co-infused with the PN admixture
summary.
When a medication needs to co-infuse with a PN admixture via Y-site
owing to limited patient access, clinicians must consider the drug prod-
Added to the PN admixture uct excipients and contents of the carrier fluid as well as the medica-
tion when evaluating compatibility and stability with PN admixtures.
The PN admixture itself will have been determined to be compati- A number of IV drugs have been studied for Y-site compatibility with
ble and stable before introducing a medication. There could be an compounded PN admixtures as well as with commercial, standardized,
advantage to including a drug within the PN admixture if it can con- multichamber PN products.22,197–205
solidate drug dosing and volume and decrease manipulations of vas- Studies are often performed in vitro, with small sample volumes,
cular access. In order to be considered, the IV drug required by a using various dilutions of a PN admixture and a drug to simulate the
patient should be a stable regimen that requires no dose titration concentrations combined during an actual Y-site infusion. Given that
and is therapeutically effective by continuous or cyclic infusion. How- the contact time during infusion is often <4 h, drug stability is rarely
ever, this still requires reviewing the available data on compatibil- evaluated. Drug compatibility via Y-site administration has most fre-
ity and stability. Unfortunately, there are a limited number of IV quently been determined by visual inspection. Within these limita-
drugs that have been evaluated so far, and few of them rely on tions, 80% (82 of 102) of medication tested with four different 2-in-
currently accepted methods of study design and analysis.4 Specific 1 PN admixtures and 78% (83 of 106) of the drugs tested with nine
criteria for evaluating compatibility and stability studies of medica- different TNAs were revealed to be visually compatible.22,197 Others
tion in PN admixtures should be followed.113,194,195 Unfortunately, revealed 80% (20 of 25) of drugs evaluated were compatible in a PN
many published studies have relied exclusively on visual compati- admixture.198 Further, 7 of 10 medications used in pediatrics were
bility, whereas some newer publications have applied some of the compatible by Y-site administration with two commercial, standard-
criteria. ized, multichamber PN admixtures.200 Also, drug Y-site compatibility
Visual physical compatibility of a medication in a PN admixture by with neonatal PN admixtures was found for 16 of 21 medications.201
itself is not indicative of chemical compatibility or drug stability. At the When no restrictions for infusion fluid, concentration, or time were
very least, drug pKa and product pH along with the presumed pH of placed, as may occur for neonates in a pragmatic study, only 5 of 131
the PN admixture are important to consider. Once the physical com- drugs were considered compatible with two PN admixtures, based on a
patibility and chemical stability are both demonstrated, verification of wide-ranging literature search.202 Y-site administration of medication
pharmacologic/therapeutic effectiveness in patient care is also neces- may rarely need to be considered for patients receiving home PN and
sary. In other words, the drug needs to be soluble, compatible, and sta- should also be evaluated for compatibility under conditions of typical
ble in the PN admixture as well as therapeutically effective—without use.203
altering the compatibility or stability of the initial PN admixture. A The results of compatibility studies with a drug can differ with a
close examination of published data is valuable, when available, but change of a single component of the PN admixture, including ILE or
extrapolation beyond study parameters (eg, component products, con- the oil source of the ILE.204,205 It is important to appreciate the details
centrations of each ingredient including the drug) is discouraged. Ide- (concentrations of each ingredient), assumptions (contact time of all
ally, a drug would only be considered if supported by pharmaceuti- active ingredients and excipients), and limitations of these studies. In
cal data describing physicochemical compatibility and stability of the the infrequent case that ILE is administered through a separate vas-
added drug and of the final PN admixture under conditions of typical cular access device from the 2-in-1 PN admixture, data on Y-site drug
use, plus clinical data confirming expected therapeutic actions of the compatibility and stability with ILE infusion have been measured.206
drug.3 As indicated earlier, all details need to be provided, including the pH
A recent example is a study that purported to show that 1 g of of each component, dilutions used, and contact time of exposure with
ampicillin (an anion) was stable in two TNAs for 7 days under refrig- performance of PFAT5 . The contact time, including across the filter sur-
eration and then for 24 h at room temperature following addition of face area, can be prolonged during low flow rates, as may be seen with
vitamins and trace elements.196 Although methods went well beyond infants.
visual compatibility, there remained some shortcomings. For example,
the drug assay was not shown to indicate stability (but drug concen-
tration was below 90% by 48 h in storage at 4◦ C), there was no PFAT5 Other components
reported, and no clinical rationale was provided as to how long the
ampicillin concentration would be expected to remain above the min- Compatibility needs to account for components of the automated com-
imum inhibitory concentration of a given bacteria during PN infusion. pounding device, the PN bag, administration set, and inline filter. Plas-
There was an increase of admixture pH, no zeta potential lower than ticizers incorporated into plastic containers, bags, and infusion sets are
−23 mV, for this low ampicillin dose studied in a 250-ml PN, with lim- not chemically bonded to the plastic and have the potential to leach
ited sampling points.196 out. DEHP is the predominant phthalate plasticizer found in PVC con-
294
22 BOULLATA ET AL .

tainers, bags, and administration sets and is known to specifically leach FINANCIAL DISCLOSURE
into lipid-containing admixtures.51,207–210 This poses a risk for toxic- None declared.
ity, so the use of EVA or multilayer EVA/ethylene–vinyl alcohol bags is
recommended for lipid-containing admixtures to flow through DEHP- ORCID
free infusion sets. The EVA bags depend on vinyl acetate content to Joseph I. Boullata PharmD, RPh https://orcid.org/0000-0003-4768-
make the plastic supple, and polyolefins do not require a plasticizer for 7703
their flexibility. Beyond the direct clinical risks from the toxicant DEHP Jay M. Mirtallo MS, RPh https://orcid.org/0000-0003-0736-6171
leaching into lipid-containing admixtures, this compound may also dis- Kathleen Gura PharmD https://orcid.org/0000-0002-0431-896X
rupt emulsion stability, likely through the release of phthalic acid (pKa Todd Canada PharmD https://orcid.org/0000-0002-2704-5749
= 2.89).114
Plasticizers are not the only component of concern. For example, REFERENCES
flexible plastic containers, bags, and administration sets (PVC, EVA, 1. Flack HL, Gans JA, Serlick SE, Dudrick SJ. The current status of par-
polypropylene) contain several metals (eg, Ba, Cd, Pb, Sn, Zn) to sta- enteral hyperalimentation. Am J Hosp Pharm. 1971;28(5):326-335.
2. Mirtallo J, Canada T, Johnson D, et al. Safe practices for parenteral
bilize the plastic polymers.211 These metals also have the potential to
nutrition. JPEN J Parenter Enteral Nutr. 2004;28(6_suppl):S39-S70.
leach into the infusate. For example, a study of trace element stabil- [Erratum published in JPEN J Parenter Enteral Nutr. 2006;30(2):177].
ity in PN noted that cobalt may have leached from the administration 3. Ayers P, Adams S, Boullata J, et al. American Society for Parenteral
set.149 When tested using a dextrose (10%), sodium chloride (0.9%), and Enteral Nutrition. A.S.P.E.N. parenteral nutrition safety consen-
sus recommendations. JPEN J Parenter Enteral Nutr. 2014;38(3):296-
and Tween-80 (5%) solution, in the three bag composition types, the
333.
primary metal appearing in the solution was zinc (at concentrations 4. Boullata JI, Gilbert K, Sacks G, et al. American Society for Parenteral
in the micrograms per liter range) from both PVC and EVA.211 The and Enteral Nutrition. A.S.P.E.N. Clinical guidelines: parenteral nutri-
findings were similar when commercial sodium chloride or dextrose tion ordering, order review, compounding, labeling, and dispensing.
JPEN J Parenter Enteral Nutr. 2014;38(3):334-377.
bags were infused through PVC administration sets, with zinc content
5. Worthington P, Balint J, Bechtold M, et al. When is parenteral nutri-
increasing significantly (remaining at the micrograms per liter range) tion appropriate? JPEN J Parenter Enteral Nutr. 2017;41(3):324-377.
despite brief infusion. Although not evaluated with an amino acid solu- 6. Boullata JI, Holcombe B, Sacks G, et al. Standardized competencies
tion or a complete PN admixture, given the propensity of amino acids for parenteral nutrition order review and parenteral nutrition prepa-
to complex with cadmium and lead, there may be a risk for those met- ration, including compounding: the ASPEN model. Nutr Clin Pract.
2016;31(4):548-555.
als leaching into the PN admixture as well.40
7. Ayers P, Boullata J, Sacks G. Parenteral nutrition safety: the story
continues. Nutr Clin Pract. 2018;33(1):46-52.
8. Allen LV, ed. Remington: The Science and Practice of Pharmacy. 22nd ed.
SUMMARY Pharmaceutical Press; 2013.
9. Barnett MI, Cosslett AG, Duffield JR, et al. Parenteral nutrition:
pharmaceutical problems of compatibility and stability. Drug Saf.
Maintaining the safety of patients requiring PN therapy requires a 1990;5(suppl 1):101-106.
recognition of the available data and the application of pharmaceu- 10. Trissel LA. Compounding our problems. Am J Hosp Pharm.
tical science to ensure the compatibility and stability of the ordered 1994;51(12):1534.
11. Allwood MC. Reflections on current issues concerning the stability of
PN prescription. This pursuit requires continued vigilance by the phar-
parenteral nutrition mixtures. Nutrition. 2002;18(7-8):691-692.
macist reviewing PN orders within the use process. The pharmacist 12. Salman G, Boullata JI, Sacks GS, Tellez-Corrales E. Call to Action:
with the requisite knowledge, skills, and experiences ensures the safety the need for enhanced nutrition support education and training in
of PN regimens as they fulfill their professional responsibility. The pharmacy schools and postgraduate programs. J Am Coll Clin Pharm.
2021;4(5):612-615.
effort occurs in collaboration with those recommending, prescribing,
13. Allwood MC. Compatibility and stability of TPN mixtures in big bags.
and preparing PN admixtures, as well as those involved in procuring all J Clin Hosp Pharm. 1984;9(3):181-198.
the relevant components. 14. Aulton ME, Taylor KMG, eds. Aulton’s Pharmaceutics. 4th ed. Churchill
Livingstone-Elsevier; 2013.
15. Florence AT, Attwood D. Physicochemical Principles of Pharmacy. 5th
CONFLICT OF INTEREST
ed. Pharmaceutical Press; 2011.
Joseph I. Boullata is a consultant and speaker for American Regent, a 16. Mirtallo JM. Parenteral formulas. In: Rombeau JL, Rolandelli RH,
consultant and speaker for B. Braun, and a speaker for Fresenius Kabi. eds. Clinical Nutrition: Parenteral Nutrition. 3rd ed. WB Saunders;
Jay M. Mirtallo is a consultant and speaker for Fresenius Kabi. Gordon 2001:118-139.
S. Sacks is an employee of Fresenius Kabi. Genene Salman has no con- 17. Tovsen ML, Smistad G, Bjerke TM, Tønnesen HH, Kristensen S.
Physicochemical stability of emulsions and admixtures for parenteral
flict of interest. Kathleen Gura is a consultant for Fresenius Kabi; on
nutrition during irradiation by glass-filtered daylight at standardized
the advisory board for B. Braun, Baxter, Fresenius Kabi, and Pfizer; and conditions. PDA J Pharm Sci Technol. 2015;69(3):346-354.
receives royalties from Fresenius Kabi. Genene Salman, Todd Canada, 18. Murphy S, Craig DQ, Murphy A. An investigation into the physical
and Angela Maguire have no conflict of interest. stability of a neonatal parenteral nutrition formulation. Acta Paediatr.
1996;85(12):1483-1486.
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 295
23

19. Jeppsson RI, Sjöberg B. Compatibility of parenteral nutrition 44. Foinard A, Perez M, Barthélémy C, et al. In vitro assessment of inter-
solutions when mixed in a plastic bag. Clin Nutr. 1984;2(3-4): action between amino acids and copper in neonatal parenteral nutri-
149-158. tion. JPEN J Parenter Enteral Nutr. 2016;40(6):827-834.
20. Ang SD, Canham JE, Daly JM. Parenteral infusion with an admixture 45. Sweetman SC. Martindale: The Complete Drug Reference. 34th ed. Phar-
of amino acids, dextrose, and fat emulsion solution: compatibility and maceutical Press; 2005.
clinical safety. JPEN J Parenter Enteral Nutr. 1987;11(1):23-27. [Erra- 46. Gennaro AR. Remington: The Science and Practice of Pharmacy. 20th ed.
tum published in JPEN 1987;11(3):274]. Lippincott Williams & Wilkins; 2000.
21. Hardy G, Puzovic M. Formulation, stability, and administration 47. Dextrose injection. Package insert. Baxter Healthcare; 2014.
of parenteral nutrition with new lipid emulsions. Nutr Clin Pract. 48. Dextrose–dextrose monohydrate injection, solution. Package insert. Fre-
2009;24(5):616-625. senius Kabi; 2018.
22. Trissel LA, Gilbert DL, Martinez JF, Baker MB, Walter WV, Mirtallo 49. Barat AC, Harrie K, Jacob M, Diamantidis TG, McIntosh NL. Effect of
JM. Compatibility of medications with 3-in-1 parenteral nutrition amino acid solutions on total nutrient admixture stability. JPEN J Par-
admixtures. JPEN J Parenter Enteral Nutr. 1999;23(2):67-74. enter Enteral Nutr. 1987;11(4):384-388.
23. Staven V, Wang S, Grønlie I, Tho I. Development and evaluation of a 50. Didier ME, Fischer S, Maki DG. Total nutrient admixtures appear
test program for Y-site compatibility testing of total parenteral nutri- safer than lipid emulsion alone as regards microbial contamination:
tion and IV drugs. Nutr J. 2016;15(1):29. growth properties of microbial pathogens at room temperature. JPEN
24. Wormleighton CV, Catling TB. Stability issues in home parenteral J Parenter Enteral Nutr. 1998;22(5):291-296.
nutrition. Clin Nutr. 1998;17(5):199-203. 51. National Toxicology Program, Center for the Evaluation of Risks to
25. Lumpkin MM. Safety alert: hazards of precipitation associated with Human Reproduction. NTP-CERHR Monograph on the Potential Human
parenteral nutrition. Am J Health-Syst Pharm. 1994;51(11):1427- Reproductive and Developmental Effects of Di(2-Ethylhexyl) Phthalate
1428. (DEHP). National Institutes of Health, US Department of Health
26. Shay DK, Fann LM, Jarvis WR. Respiratory distress and sudden death and Human Services; 2006. Accessed January 29, 2020. NIH Pub-
associated with receipt of a peripheral parenteral nutrition admix- lication No. 06–4476. https://ntp.niehs.nih.gov/ntp/ohat/phthalates/
ture. Infect Control Hosp Epidemiol. 1997;18(12):814-817. dehp/dehp-monograph.pdf
27. Aminosyn-PF 7% and Aminosyn-PF 10% amino acid injection. Package 52. Mirtallo JM, Ayers P, Boullata JI, et al. ASPEN lipid injectable emul-
insert. ICU Medical; 2018. sion safety recommendations, Part 1: background and adult consid-
28. Aminosyn II 10% and Aminosyn II 15% amino acid injection. Package erations. Nutr Clin Pract. 2020;35(5):769-782.
insert. Hospira; 2010. 53. Clinolipid. Package insert. Baxter Healthcare; 2020.
29. Clinisol 15% amino acid injection. Package insert. Baxter Healthcare; 54. Intralipid. Package insert. Baxter Healthcare; 2020.
2018. 55. Nutrilipid injection. Package insert. B. Braun Medical; 2020.
30. FreAmine III 10% amino acid injection. Package insert. B. Braun Medi- 56. Omegaven injection. Package insert. Fresenius Kabi; 2020.
cal; 2020. 57. SMOFLipid. Package insert. Fresenius Kabi; 2020.
31. HepatAmine 8% amino acid injection. Package insert. B. Braun Medical; 58. Llop Talaverón JM, Novak A, Suñé Negre JM, Tahull MB, Badosa EL,
2020. Ticó Grau JR. Phytosterol determination in lipid emulsions for par-
32. Plenamine 15% amino acids injection Package insert. B. Braun Medical; enteral nutrition. Farm Hosp. 2018;42(3):116-119.
2020. 59. Anez-Bustillos L, Dao DT, Baker MA, Fell GL, Puder M, Gura
33. Premasol 6% and Premasol 10% amino acid injection. Package insert. KM. Intravenous fat emulsion formulations for the adult and
Baxter Healthcare; 2014. pediatric patient: understanding the differences. Nutr Clin Pract.
34. ProSol 20% amino acid injection. Package insert. Baxter Healthcare; 2016;31(5):596-609.
2014. 60. Driscoll DF. Physicochemical assessment of total nutrient admixture
35. Travasol amino acid injection. Package insert. Baxter Healthcare; 2020. stability and safety: quantifying the risk. Nutrition. 1997;13(2):166-
36. TrophAmine 6% and TrophAmine 10% amino acid injection. Package 167.
insert. B. Braun Medical; 2020. 61. Klang MG. PFAT5 and the evolution of lipid admixture stability. JPEN
37. Hardy G, Ball P, McElroy B. Basic principles for compounding all-in- J Parenter Enteral Nutr. 2015;39(1_suppl):67S-71S.
one parenteral nutrition admixtures. Curr Opin Clin Nutr Metab Care. 62. Brown R, Quercia RA, Sigman R. Total nutrient admixture: a review.
1998;1(3):291-296. JPEN J Parenter Enteral Nutr. 1986;10(6):650-658.
38. do Nascimento PC, da Marques MS, Bohrer D, de Carvalho LM, Car- 63. Sayeed FA, Tripp MG, Sukumaran KB, Mikrut BA, Stelmach HA, Raihle
valho CW. Chromatography and atomic absorption spectrometry for JA. Stability of total nutrient admixtures using various intravenous
the assessment of heavy metal distribution among amino acids used fat emulsions. Am J Hosp Pharm. 1987;44(10):2271-2280.
in parenteral nutrition formulations: studies with cadmium and lead. 64. Watrobska-Swietlikowska D. Stability of commercial parenteral
Biol Trace Elem Res. 2011;141(1-3):76-85. lipid emulsions repacking to polypropylene syringes. PLoS One.
39. Olson LM, Wieruszewski PM, Jannetto PJ, et al. Quantitative assess- 2019;14(4):e0214451.
ment of trace-element contamination in parenteral nutrition compo- 65. Driscoll DF, Parikh M, Silvestri AP, Klütsch K, Bistrian BR, Nehne
nents. JPEN J Parent Enteral Nutr. 2019;43(8):970-976. J. Establishing a stability window for medium- and long-chain-
40. Unger N, Holzgrabe U. Stability and assessment of amino acids in par- triglyceride lipid-based total nutrient admixtures using USP stan-
enteral nutrition solutions. J Pharm Biomed Anal. 2018;147:125-139. dards. Am J Health-Syst Pharm. 2006;63(21):2135-2143.
41. Harraki B, Guiraud P, Rochat M, Alary J, Favier A. Interactions 66. Floyd AG. Top ten considerations in the development of parenteral
related to trace elements in parenteral nutrition. Pharm Acta Helv. emulsions. Pharm Sci Technlo Today. 1999;4(4):134-143.
1995;70(4):269-278. 67. Guidelines for essential trace element preparations for parenteral
42. Allwood MC, Martin H, Greenwood M, Maunder M. Precipita- use: a statement by an expert panel. AMA Department of Foods and
tion of trace elements in parenteral nutrition mixtures. Clin Nutr. Nutrition. JAMA. 1979;241(19):2051-2054.
1998;17(5):223-226. 68. Weinstein MM, Siegel FP, Blake MI. Formulation of a stable trace ele-
43. Thibault M. Possible incompatibility between amino acids and cop- ment solution for TPN. Am J Hosp Pharm. 1980;37(12):1620.
per in solutions for pediatric parenteral nutrition. Can J Hosp Pharm. 69. Vanek VW, Borum P, Buchman A, et al. Novel Nutrient Task Force,
2014;67(2):160-164. Parenteral Multi-Vitamin and Multi–Trace Element Working Group;
296
24 BOULLATA ET AL .

American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) 97. Thiamine hydrochloride injection. Package insert. Mylan; 2017.
Board of Directors. A.S.P.E.N. position paper: recommendations for 98. Pyroxidine hydrochloride injection. Package insert. Fresenius Kabi USA;
change in commercially available parenteral multivitamin and multi- 2019.
trace element products. Nutr Clin Pract. 2012;27(4):440-491. 99. Folic acid injection. Package insert. Fresenius Kabi USA; 2019.
70. Vanek VW, Borum P, Buchman A, et al. A call to action to bring safer 100. Laegeler WL, Tio JM, Blake MI. Stability of certain amino acids
parenteral micronutrient products to the U.S. market. Nutr Clin Pract. in a parenteral nutrition solution. Am J Hosp Pharm. 1974;31(8):
2015;30(4):559-569. 776-779.
71. Stehle P, Stoffel-Wagner B, Kuhn KS. Parenteral trace element provi- 101. Jurgens RW, Henry RS, Welco A. Amino acid stability in a mixed
sion: recent clinical research and practical conclusions. Eur J Clin Nutr. parenteral nutrition solution. Am J Hosp Pharm. 1981;38(9):1358-
2016;70(8):886-893. 1359.
72. Jin J, Mulesa L, Rouillet MC. Trace elements in parenteral nutrition: 102. USP Chapter <788> Particulate matter injections. USP 42–NF 37. The
considerations for the prescribing clinician. Nutrients. 2017;9(5):440. United States Pharmacopeial Convention; 2019.
73. MultiTrace-4 Pediatric and MultiTrace-4 Neonatal. Package insert. 103. Newton DW. Maillard reactions in pharmaceutical formulations and
American Regent; 2018. human health. Int J Pharm Compound. 2011;15(1):32-40.
74. Tralement trace elements 4 injection, USP. Package insert. American 104. Stegink LD, Freeman JB, Den Besten L, Filer LJ. Maillard reaction
Regent; 2020. products in parenteral nutrition. Prog Food Nutr Sci. 1981;5(1-6):265-
75. Selenious acid injection. Package insert. American Regent; 2020. 278.
76. Zinc sulfate injection. Package insert. American Regent; 2020. 105. Labuza TP, Massaro SA. Browning and amino acid loss in model total
77. Mitrano FP, Baptista RJ. Stability of cimetidine HCl and copper sul- parenteral nutrition solutions. J Food Sci. 1990;55(3):821-826.
fate in a TPN solutions. DICP. 1989;23(5):429-430. 106. Fry LK, Stegink LD. Formation of Maillard reaction products in par-
78. Power-Burns M. Adult multi-trace element availability. May 2013. enteral alimentation solutions. J Nutr. 1982;112(8):1631-1637.
https://www.fda.gov/media/86363/download. Accessed October 21, 107. Novaes MR, Lima LA, Sousa MV. Maillard’s reaction in par-
2020 enteral solutions supplemented with arginine. Arch Latinoam Nutr.
79. INFED. Package insert. Allergan Sales; 2020. 2001;51(3):265-268.
80. Venofer. Package insert. American Regent; 2020. 108. Bullock L, Fitzgerald JF, Walter WV. Emulsion stability in total nutri-
81. Khaodhiar L, Keane-Ellison M, Tawa NE, et al. Iron deficiency anemia ent admixtures containing a pediatric amino acid formulation. JPEN J
in patients receiving home total parenteral nutrition. JPEN J Parenter Parenter Enteral Nutr. 1992;16(1):64-68.
Enteral Nutr. 2002;26(2):114-119. 109. Washington C, Athersuch A, Kynoch DJ. The electrokinetic proper-
82. Domellöf M, Szitanyi P, Simchowitz V, Franz A, Mimouni F; ties of phospholipid-stabilized fat emulsions. IV: the effect of glucose
ESPGHAN/ESPEN/ESPR/CSPEN Working Group on Pediatric Par- and pH. Int J Pharm. 1990;64(2-3):217-222.
enteral Nutrition. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on 110. Washington C, Connoly MA, Manning R. The electrokinetic proper-
pediatric parenteral nutrition: iron and trace minerals. Clin Nutr. ties of phospholipid-stabilized fat emulsions. V: the effect of amino
2018;37(6):2354-2359. acids on stability. Int J Pharm. 1991;65(1):57-63.
83. Barry DM, Reeve AW. Increased incidence of gram-negative 111. Kaminski MV, Harris DF, Collin CF, Sommers GA. Electrolyte com-
neonatal sepsis with intramuscular iron administration. Pediatrics. patibility in a synthetic amino acid hyperalimentation solution. Am J
1977;60(6):908-912. Hosp Pharm. 1974;31(3):244-246.
84. M.V.I. pediatric. Package insert. Hospira; 2019. 112. Allwood MC, Kearney MCJ. Compatibility and stability of additives in
85. M.V.I. adult. Package insert. Hospira; 2020. parenteral nutrition admixtures. Nutrition. 1998;14(9):697-706.
86. Pediatric Infuvite multiple vitamins. Package insert. Sandoz Canada; 113. Niemiec PW, Vanderveen TW. Compatibility considerations in par-
2016. enteral nutrient solutions. Am J Hosp Pharm. 1984;41(5):893-911.
87. Adult Infuvite multiple vitamins. Package insert. Sandoz Canada; 2012. 114. Driscoll DF, Bhargava HN, Zaim RH, Babayan VK, Bistrian BR. Physic-
88. Kriegel C, Festag M, Kishore RSK, Roethlisberger D, Schmitt G. ochemical stability of total nutrient admixtures. Am J Health- Syst
Pediatric safety of polysorbates in drug formulations. Children. Pharm. 1995;52(6):623-634.
2020;7(1):1. https://doi.org/10.3390/children7010001 115. Newton DW, Driscoll DF. Chemistry and safety of phosphates injec-
89. Brayfield A. Martindale: The Complete Drug Reference. 39th ed. Phar- tions. Am J Health-Syst Pharm. 2008;65(18):1761-1766.
maceutical Press; 2019. 116. Watrobska-Swietlikowska D. Compatibility of maximum inorganic
90. MacDonald MG, Fletcher AB, Johnson EL, Boeckx RL, Getson PR, and organic calcium and phosphate content in neonatal parenteral
Miller MK. The potential toxicity to neonates of multivitamin prepa- solutions. Sci Report. 2019;9(1):10525.
rations used in parenteral nutrition. JPEN J Parenter Enteral Nutr. 117. Newton DW, Driscoll DF. Calcium and phosphate compatibility:
1987;11(2):169-171. revisited again. Am J Health-Syst Pharm. 2008;65(1):73-80.
91. Abedi F, Razavi BM, Hosseinzadeh H. A review on gentisic acid as a 118. Wong JC, McDougal AR, Tofan M, et al. Doubling calcium and phos-
plant derived phenolic acid and metabolite of aspirin: comprehensive phate concentrations in neonatal parenteral nutrition solutions using
pharmacology, toxicology, and some pharmaceutical aspects. Phy- monobasic potassium phosphate. J Am Coll Nutr. 2006;25(1):70-77.
tother Res. 2020;34(4):729-741. 119. Anderson C, MacKay M. Physical compatibility of calcium chloride
92. Lavoie JC, Bélanger S, Spalinger M, Chessex P. Admixture of a and sodium glycerophosphate in pediatric parenteral nutrition solu-
multivitamin preparation to parenteral nutrition: the major con- tions. JPEN J Parenter Enteral Nutr. 2016;40(8):1166-1169.
tributor to in vitro generation of peroxides. Pediatrics. 1997;99(3): 120. Robinson LA, Wright BT. Central venous catheter occlusion caused
E6. by body heat mediated calcium phosphate precipitation. Am J Hosp
93. Christian VJ, Tallar M, Walia CLS, Sieracki R, Goday PS. Systematic Pharm. 1982;39(1):120-121.
review of hypersensitivity to parenteral nutrition. JPEN J Parenter 121. Boddapati S, Yang K, Murty R. Intravenous solution compatibility and
Enteral Nutr. 2018;42(8):1222-1229. filter-retention characteristics of trace-element preparations. Am J
94. ASCOR. Package insert. McGuff Pharmaceuticals; 2019. Hosp Pharm. 1981;38(11):1731-1736.
95. Ascorbic acid injection. Package insert. Mylan Institutional; 2016. 122. Staven V, Wang S, Grønlie I, Tho I. Physical stability of an all-in-one
96. Thiamine hydrochloride injection. Package insert. Physicians Total parenteral nutrition admixture for preterm infants upon mixing with
Care; 2010. micronutrients and drugs. Eur J Hosp Pharm. 2020;27(1):36-42.
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 297
25

123. Cochran EB, Boehm KA. Prefilter and postfilter cysteine/cysteine 141. Tripp MG, Menon SK, Mikrut BA. Stability of total nutrient admix-
and copper concentrations in pediatric parenteral nutrition solutions. tures in a dual-chamber flexible container. Am J Hosp Pharm.
JPEN J Parenter Enteral Nutr. 1992;16(5):460-463. 1990;47(11):2496-2503.
124. Bates CG, Greiner G. Gegenheimer A. Precipitate in admixtures of 142. Watrobska-Swietlikowska D, Szlagatys-Sidorkiewicz A, Katarzyna Ł.
new amino acid injection. Am J Hosp Pharm. 1984;41(7):1312. Evaluation of physical stability of all in one parenteral admixtures for
125. Chatterji DC, Kapoor J. Precipitate in admixtures of new amino acid pediatric home care with high electrolytes concentrations. Nutr Hosp.
injection (reply). Am J Hosp Pharm. 1984;41(7):1312,1316. 2015;31(1):236-243.
126. Mayhew SL, Quick MW. Compatibility of iron dextran with 143. Lobo BW, da Veiga VF, Cabral LM, et al. Influence of the relative com-
neonatal parenteral nutrient solutions. Am J Health-Syst Pharm. position of trace elements and vitamins in physicochemical stability
1997;54(5):570-571. of total parenteral nutrition formulations for neonatal use. Nutr J.
127. Lee D, Barsky D, Hughes R, Driggers J, Poupard N, Mascarenhas M. 2012;11(1):26.
Evaluation of the safety of iron dextran with parenteral nutrition in 144. Uccello-Barretta G, Balzano F, Aiello F, Falugiani N, Desideri I. Sta-
the paediatric inpatient setting. Nutr Diet. 2017;74(5):471-475. bility of hydrophilic vitamins mixtures in the presence of electrolytes
128. Billion-Rey F, Guillaumont M, Frederich A, Aulagner G. Stability of and trace elements for parenteral nutrition: a nuclear magnetic reso-
fat-soluble vitamins A (retinol palmitate), E (tocopherol acetate), and nance spectroscopy investigation. J Pharm Biomed Anal. 2015;107:7-
K1 (phylloquinone) in total parenteral nutrition at home. JPEN J Par- 10.
enter Enteral Nutr. 1993;17(1):56-60. 145. Lavoie JC, Chessex P. Bound iron admixture prevents the sponta-
129. Henton DH, Merritt RJ. Vitamin A sorption to polyvinyl and neous generation of peroxides in total parenteral nutrition solutions.
polyolefin intravenous tubing. JPEN J Parenter Enteral Nutr. J Pediatr Gastroenterol Nutr. 1997;25(3):307-311.
1990;14(1):79-81. 146. Vaughan LM, Small C, Plunkett V. Incompatibility of iron dextran and
130. Koletzko B, Goulet O, Hunt J, Krohn K, Shamir R, Parenteral a total nutrient admixture. Am J Hosp Pharm. 1990;47(8):1745-1746.
Nutrition Guidelines Working Group; European Society for Clinical 147. Kumpf VJ. Update on parenteral iron therapy. Nutr Clin Pract.
Nutrition and Metabolism; European Society of Paediatric Gastroen- 2003;18(4):318-326.
terology, Hepatology and Nutrition (ESPGHAN); European Society 148. Puntis JW, Rushton DI. Pulmonary intravascular lipid in neonatal
of Paediatric Research (ESPR). Guidelines on pediatric parenteral necropsy specimens. Arch Dis Child. 1991;66(1 Spec No):26-28.
nutrition of the European Society of Pediatric Gastroenterology, 149. Pluhator-Murton MM, Fedorak RN, Audette RJ, et al. Trace element
Hepatology and Nutrition (ESPGHAN) and the European Society for contamination of total parenteral nutrition. 2: effect of storage dura-
Clinical Nutrition and Metabolism (ESPEN), supported by the Euro- tion and temperature. JPEN J Parenter Enteral Nutr. 1999;23(4):228-
pean Society of Pediatric Research (ESPR). J Pediatr Gastroenterol 232.
Nutr. 2005;41Suppl2:S1-S87. 150. Burge JC, Flancbaum L, Holcombe B. Copper decreases ascorbic
131. Gillis J, Jones G, Pencharz P. Delivery of vitamins A, D, and E in acid stability in total parenteral nutrition solutions. J Am Diet Assoc.
total parenteral nutrition solutions. JPEN J Parenter Enteral Nutr. 1994;94(7):777.
1983;7(1):11-14. 151. Allwood MC, Greenwood M. Assessment of trace element com-
132. Brawley V, Bhatia J, Karp WB. Effect of sodium metabisulfite on patibility in total parenteral nutrition infusions. Pharm Weekbl (Sci).
hydrogen peroxide production in light exposed pediatric parenteral 1992;14(5):321-324.
amino acid solutions. Am J Health-Syst Pharm. 1998;55(12):1288- 152. Shils ME, Levander OA. Selenium stability in TPN solutions. Am J Clin
1292. Nutr. 1982;35(4):829.
133. Bhatia J, Stegink LD, Ziegler EE. Riboflavin enhances photo-oxidation 153. Levander OA. Considerations in the design of selenium bioavailability
of amino acids under simulated clinical conditions. JPEN J Parenter studies. Fed Proc. 1983;42(6):1721.
Enteral Nutr. 1983;7(3):277-279. 154. McGee CD, Mascarenhas MG, Ostro MJ, Tsallas G, Jeejeebhoy K.
134. García J, Silva E. Flavin-sensitized photooxidation of amino acids Selenium and vitamin E stability in parenteral solutions. JPEN J Par-
present in a parenteral nutrition infusate: protection by ascorbic acid. enter Enteral Nutr. 1985;5(5):568-570.
J Nutr Biochem. 1997;8(6):341-345. 155. Ganther HE, Kraus RJ. Chemical stability of selenious acid in total
135. Unger N, Ferraro A, Holzgrabe U. Investigation of tryptophan- parenteral nutrition solutions containing ascorbic acid. JPEN J Par-
related yellowing in parenteral amino acid solution: development enter Enteral Nutr. 1989;13(2):185-188.
of a stability-indicating method and assessment of degradation 156. Sill-Steffens R, Hardy G. Sodium selenite stability in intravenous solu-
products in pharmaceutical formulations. J Pharm Biomed Anal. tions. Clin Nutr. 2004;23(4):827-828.
2020;177:112839. 157. Hardy G, Hardy I, Martin H. Selenium stability in vitamin and glu-
136. Harraki B, Guiraud P, Rochat MH, Fussellier M, Favier A. Influence of tamine mixtures. Clin Nutr. 2005;24(4):596.
copper, iron, and zinc on the physicochemical properties of parenteral 158. Tu Y-H, Knox NL, Bringer JM, et al. Compatibility of iron dextran with
admixture. J Parenter Sci Technol. 1993;47(5):199-204. total nutrient admixtures. Am J Hosp Pharm. 1992;49(9):2233-2235.
137. Iliano L, Delanghe M, van Den Baviere H, Lauwers A. Effect of elec- 159. MacKay M, Rusho W, Jackson D, McMillin G, Winther B. Physical
trolytes in the presence of some trace elements on the stability of and chemical stability of iron sucrose in parenteral nutrition. Nutr Clin
all-in-one emulsion mixtures for total parenteral nutrition. J Clin Hosp Pract. 2009;24(6):733-737.
Pharm. 1984;9(2):87-93. 160. Ferguson TI, Emery S, Price-Davies R, Cosslett AG. A review of sta-
138. Deitel M, Smith LC, Friedman KL, et al. Physical stability of a bility issues associated with vitamins in parenteral nutrition. eSPEN J.
total nutrient admixture for total parenteral nutrition. Can J Surg. 2014;9(2):e49-e53.
1989;32(4):240-243. 161. Ha E, Wang W, Wang YJ. Peroxide formation in polysorbate 80 and
139. Télessy IG, Balogh J, Turmezei J, Dredán J, Zelkó R. Stability assess- protein stability. J Pharm Sci. 2002;91(10):2252-2264.
ment of o/w parenteral nutrition emulsions in the presence of 162. Silvers KM, Darlow BA, Winterbourn CC. Lipid peroxide and hydro-
high glucose and calcium concentrations. J Pharm Biomed Anal. gen peroxide formation in parenteral nutrition solutions containing
2011;56(2):159-164. multivitamins. JPEN J Parenter Enteral Nutr. 2001;25(1):14-17.
140. Forchielli ML, Bonoli A, Stancari A, et al. Do carnitine and extra trace 163. Laborie S, Lavoie JC, Chessex P. Paradoxical role of ascorbic acid and
elements change stability of paediatric parenteral nutrition admix- riboflavin in solutions of total parenteral nutrition: implication in pho-
tures?. Clin Nutr. 2019;38(5):2369-2374. toinduced peroxide generation. Pediatr Res. 1998;43(5):601-606.
298
26 BOULLATA ET AL .

164. Silva E, Ugarte R, Andrade A, Edwards AM. Riboflavin-sensitized pho- 186. Haas C, Genzel-Boroviczény O, Koletzko B. Losses of vitamin A and E
toprocesses of tryptophan. J Photochem Photobiol B. 1994;23(1):43- in parenteral nutrition suitable for premature infants. Eur J Clin Nutr.
48. 2002;56(9):906-912.
165. Huvaere K, Cardoso DR, Homem-de-Mello P, Westermann S, Skib- 187. Allwood MC, Ball PA, Driscoll DF, Sizer T. Light protection dur-
sted LH. Light-induced oxidation of unsaturated lipids as sensitized ing parenteral nutrition infusion: is it really necessary?. Nutrition.
by flavins. J Phys Chem B. 2010;114(16):5583-5593. 2000;16(3):234-235.
166. Natera J, Massad WA, García NA. Vitamin B1 as a scavenger of reac- 188. Drott P, Meurling S, Meurling L. Clinical adsorption and photodegra-
tive oxygen species photogenerated by vitamin B2. Photochem Photo- dation of the fat soluble vitamins A and E. Clin Nutr. 1991;10(6):348-
biol. 2011;87(2):317-323. 351.
167. Ribeiro DO, Pinto DC, Lima LMTR, Volpato NM, Cabral LM, de Sousa 189. Lavoie J, Chessex P, Rouleau T, Tsopmo A, Friel J. Shielding parenteral
VP. Chemical stability study of vitamins thiamine, riboflavin, pyridox- multivitamins from light increases vitamin A and E concentration in
ine and ascorbic acid in parenteral nutrition for neonatal use. Nutr J. lung of newborn guinea pigs. Clin Nutr. 2007;26(3):341-347.
2011;10(1):47-56. 190. Guidetti M, Sforzini A, Bersani G, et al. Vitamin A and vitamin E iso-
168. Mirkovic DC, Basic Z, Roganovic B. PP185-Sun influence of type of forms stability and peroxidation potential of all-in-one admixtures
bag, sort of admixtures and exposure to daylight upon stability of for parenteral nutrition. Int J Vitam Nutr Res. 2008;78(3):156-166.
vitamins C and B2 in parenteral nutrition admixtures. Clin Nutr Suppl. 191. Schmutz CW, Martinelli E, Mühlebach S. Stability of vitamin K1
2011;6(1):93-94. assessed by HPLC in total parenteral nutrition (TPN) admixtures. Clin
169. Ferguson TI, Price-Davies R, Cosslett A. Vitamins—an unknown Nutr. 1992;12(169):110-111.
quantity (PP259). Clin Nutr. 2013;32:S219. 192. Laborie S, Lavoie JC, Pineault M, Chessex P. Protecting solutions of
170. Chen MF, Boyce HW Jr, Triplett L. Stability of the B vitamins in parenteral nutrition from peroxidation. JPEN J Parenter Enteral Nutr.
mixed parenteral nutrition solution. JPEN J Parenter Enteral Nutr. 1999;23(2):104-108.
1983;7(5):462-464. 193. Robinson DT, Ayers P, Fleming B, et al. Recommendations for pho-
171. Barker A, Hebron BS, Beck PR, Ellis B. Folic acid and total parenteral toprotection of parenteral nutrition for neonates and premature
nutrition. JPEN J Parenter Enteral Nutr. 1984;8(1):3-8. infants: an ASPEN position paper. Nutr Clin Pract. 2021;36(5):927-
172. McEvoy GK. Handbook on Injectable Drugs. 20th ed. American Society 941.
of Health-System Pharmacists; 2018. 194. Driscoll DF, Baptista RJ, Mitrano FP, et al. Parenteral nutrient admix-
173. Scheiner JM, Araujo MM, DeRitter E. Thiamin destruction by sodium tures as drug vehicles: theory and practice in the critical care setting.
bisulfite in infusion solutions. Am J Hosp Pharm. 1981;38(12):1911- DICP. 1991;25(3):276-283.
1913. 195. Newton DW. Y-site compatibility of intravenous drugs with par-
174. Bowman BB, Nguyen P. Stability of thiamin in parenteral nutrition enteral nutrition. JPEN J Parenter Enteral Nutr. 2013;37(3):297-299.
solutions. JPEN J Parenter Enteral Nutr. 1983;7(6):567-568. 196. Stawny M, Gostyńska A, Dettlaff, et al. Effect of lipid emulsion
175. Smith JL, Canham JE, Kirkland WD, Wells PA. Effect of intralipid, on stability of ampicillin in total parenteral nutrition. Nutrients.
amino acids, container, temperature, and duration of storage on vita- 2019;11(3):559.
min stability in total parenteral nutrition admixtures. JPEN J Parenter 197. Trissel LA, Gilbert DL, Martinez JF, Baker MB, Walter WV, Mir-
Enteral Nutr. 1988;12(5):478-483. [Erratum published in JPEN J Par- tallo JM. Compatibility of parenteral nutrient solutions with selected
enter Enteral Nutr 1989;13(2):167.] drugs during simulated Y-site administration. Am J Health-Syst Pharm.
176. Allwood MC. Stability of vitamins in total parenteral nutrition solu- 1997;54(11):1295-1300.
tions stored in three litter bags. Brit J Int Ther. 1982;1(1):22. 198. Bouchoud L, Fonzo-Christe C, Klingmüller M, Bonnabry P. Compat-
177. Rockwell GF, Campfield T, Nelson BC, Uden PC. Oxalogenesis in ibility of intravenous medications with parenteral nutrition: in vitro
parenteral nutrition solution components. Nutrition. 1998;14(11- evaluation. JPEN J Parenter Enteral Nutr. 2013;37(3):416-424.
12):836-839. 199. Nezvalova-Henriksen K, Nilsson N, Østerberg CT, Berge VS, Tho I.
178. Ferreyra ME, Ocaña MC, Vega R. Trace elements influence on ascor- Y-site physical compatibility of Numeta G13E with drugs frequently
bic acid availability (PP260). Clin Nutr. 2013;32:S121. used at neonatal intensive care. Pharmaceutics. 2020;12(7):677.
179. Proot P, De Pourcq L, Raymakers AA. Stability of ascorbic 200. Staven V, Iqbal H, Wang S, Grønlie I, Tho I. Physical compatibil-
acid in a standard total parenteral nutrition mixture. Clin Nutr. ity of total parenteral nutrition and drugs in Y-site administra-
1994;13(5):273-279. tion to children from neonates to adolescents. J Pharm Pharmacol.
180. Gibbons E, Allwood MC, Neal T, Hardy G. Degradation of dehy- 2017;69(4):448-462.
droascorbic acid in parenteral nutrition mixtures. J Pharm Biomed 201. Veltri M, Lee CKK. Compatibility of neonatal parenteral nutrient
Anal. 2001;25(3-4):605-611. solutions with selected intravenous drugs. Am J Health-Syst Pharm.
181. Dupertuis YM, Morch A, Fathi M. Physical characteristics of total par- 1996;53(21):2611-2613.
enteral nutrition bags significantly affect the stability of vitamins C 202. Kalikstad B, Skjerdal A, Hansen TW. Compatibility of drug infusions
and B1: a controlled prospective study. JPEN J Parenter Enteral Nutr. in the NICU. Arch Dis Child. 2010;95(9):745-748.
2002;26(5):310-316. 203. Watrobska-Swietlikowska D, Pietka M, Klek S. Evaluation of Y-site
182. Allwood MC. Factors influencing the stability of ascorbic acid in total compatibility of home total parenteral nutrition and intravenous loop
parenteral nutrition infusions. J Clin Hosp Pharm. 1984;9(2):75-85. diuretics. Medicine. 2019;98(21):(e15747).
183. Kearney MC, Allwood MC, Martin H, Neal T, Hardy G. The influence 204. Stawny M, Gostyńska A, Nadolna M, Jelińska A. Safe practice of Y-
of amino acid source on the stability of ascorbic acid in TPN admix- site drug administration: the case of colistin and parenteral nutrition.
tures. Nutrition. 1998;14(2):173-178. Pharmaceutics. 2020;12(3):292.
184. Allwood MC, Plane JH. The wavelength-dependent degradation of 205. Stawny M, Nadolna M, Jelinska A. In vitro compatibility studies of
vitamin A exposed to ultraviolet light. Int J Pharmaceutics. 1986;31(1- vancomycin with ready-to-use parenteral nutrition admixtures for
2):1-7. safer clinical practice. Clin Nutr. 2020;39(8):2539-2546.
185. Allwood MC, Martin HJ. The photodegradation of vitamins A 206. Ross EL, Salinas A, Petty K, Her C, Carpenter JF. Compatibility of
and E in parenteral nutrition mixtures during infusion. Clin Nutr. medications with intravenous lipid emulsions: effects of simulated Y-
2000;19(5):339-342. site mixing. Am J Health-Syst Pharm. 2020;77(23):1980-1985.
JOURNAL OF PARENTERAL AND ENTERAL NUTRITION 299
27

207. Agency for Toxic Substances and Disease Registry. Toxicological Profile polypropylene bags and infusion sets into infusion solutions. PDA J
for di(2-ethylhexyl)phthalate (DEHP). Agency for Toxic Substances and Pharm Sci and Tech. 2019;73(1):60-69.
Disease Registry; 2002.
208. Loff S, Kabs F, Witt K, Sartoris J, Mandl B, Niessen KH, et al.
SUPPORTING INFORMATION
Polyvinylchloride infusion lines expose infants to large amounts of
toxic plasticizers. J Pediatr Surg. 2000;35(12):1775-1781. Additional supporting information may be found in the online version
209. Green R, Hauser R, Calafat AM, et al. Use of di(2-ethylhexyl) of the article at the publisher’s website.
phthalate-containing medical products and urinary levels of mono(2-
ethylhexyl) phthalate in neonatal intensive care unit infants. Environ
Health Perspect. 2005;113(9):1222-1225.
210. Weuve J, Sanchez BN, Calafat AM, et al. Exposure to phtha- How to cite this article: Boullata JI, Mirtallo JM, Sacks GS,
lates in neonatal intensive care unit infants: urinary concentrations
et al. Parenteral nutrition compatibility and stability: A
of monoesters and oxidative metabolites. Environ Health Perspect.
2006;114(9):1424-1431. 2022;46:
comprehensive review. J Parenter Enteral Nutr. 2022;1-27.
211. Mattiazzi P, Bohrer D, Viana C, et al. Extraction/leaching of metal- 273–299. https://doi.org/10.1002/jpen.2306
https://doi.org/10.1002/jpen.2306
containing additives from polyvinyl chloride, ethyl vinyl acetate, and

You might also like