Parenteral Nutrition in Infants and Children

20 downloads 220 Views 940KB Size Report
Invited Review. Parenteral Nutrition in Infants and Children. Robert J. Shulman and Sarah Phillips. Department of Pediatrics, Baylor College of Medicine, ...
Journal of Pediatric Gastroenterology and Nutrition 36:587–607 © May 2003 Lippincott Williams & Wilkins, Inc., Philadelphia

Invited Review

Parenteral Nutrition in Infants and Children Robert J. Shulman and Sarah Phillips Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Texas Children’s Hospital, Houston, Texas, U.S.A.

JPGN 36:587–607, 2003. Key Words: Parenteral nutrition— Intravenous—Feeding—Neonates—Energy—Protein—Amino

acids—Lipid emulsion. © 2003 Lippincott Williams & Wilkins, Inc.

INTRODUCTION

Our immediate reaction was to take offense, feel hurt, and shake our heads in disbelief that anyone could be so foolish as to think such things. However, as we thought about it, we realized that the question is pertinent and perceptive. Like the clinical pathways set out for common illnesses such as diarrhea and croup, the question is not so much how to follow the roadmap but rather, when not to follow it. Thus, our review will focus on two areas: 1) Common misconceptions surrounding the use of PN; and 2) When should “standard PN” (i.e., checking the weight and the box on the order sheet) not be used. These questions have lead us to review recent developments in PN (in the past 5–7 years). Whenever possible we will take an evidenced-based approach to the data. Unless specified, the studies we will discuss were carried out in pediatric patients. The risk of any review is that it is old hat to some and very new to others. We have done our best to strike a middle ground. Because it could be a subject unto itself, we will not review PN-associated cholestasis, although when pertinent, some comments will be made.

Parenteral nutrition (PN) came of age in 1964 with the demonstration that beagle puppies could be nourished successfully from 12 weeks of age to maturity by providing all nutrients intravenously (1). The first total parenteral nutrition of an infant with extreme short bowel syndrome followed in 1967 (1). Since them many lessons have been learned as a result of complications of PN. These have included nutrient deficiencies and excesses, infections, complications of inadequate or excessive energy and protein intake, liver disease, and toxicities from product contamination. Our patients have paid the price for these lessons, but fortunately improved survival has also resulted. These incidents have reinforced to us the physicians the axiom that “good judgment comes from experience . . . and experience comes from bad judgment.” (2). Along the way we have learned much about nutrition, infection, liver pathophysiology, and child development (e.g., yes, infants really can “unlearn” how to suck and swallow). Have we learned so much that administration of PN can now be placed on autopilot? The question posed to the authors when this review was solicited was whether the art and science of PN had reached the stage where administration could be treated as a routine clinical algorithm such as diarrhea or croup. Do we only need to check the weight, calculate the administration rate and check the box next to “Standard Child Solution” on the PN pharmacy order sheet? Is there really anything new in PN?

Energy Requirements, Energy Sources, and the Consequences of Overfeeding Estimating the appropriate energy intake is a fundamental step in prescribing PN. Although we have long acknowledged the risks of underfeeding our patients, more recently we have come to reevaluate energy requirements in particular clinical scenarios and to appreciate the problems associated with excessive energy intake.

Received November 25, 2002; accepted February 13, 2003. Address correspondence to Robert J. Shulman, 1100 Bates St., Houston, Texas 77030, U.S.A. (e-mail: [email protected]).

587

588

R. J. SHULMAN AND S. PHILLIPS Energy Requirements

A few terms must be defined in order to discuss energy expenditure (3). Basal metabolic rate is the energy expenditure of a recumbent child or adult in a thermoneutral environment after a 12- to 18-hour fast just when the individual has awakened but before daily activities have commenced. Basal metabolic rate is a reflection of the energy expenditure required for vital processes. Resting energy expenditure refers to the energy expenditure of a person at rest in a thermoneutral environment. Basal metabolic rate and resting energy expenditure usually do not differ by more than 10% (3). Total energy expenditure is the sum of requirements for basal metabolism, thermic effect of ingested food, thermoregulation, and activity. In older children, activity accounts for a large proportion of total energy expenditure. Thus, the total energy expenditure of a child who is hospitalized and lying in bed is reduced. Historically, it has been thought that the surgical patient requires an energy intake proportional to the severity of the illness. By definition, the energy intake would be increased compared with that of a “non-stressed” patient. However, from a practical standpoint this concept does not seem to be the case as the increased energy expenditure is short-lived. For example, although newborns have a 20% increase in resting energy expenditure after major surgery, this elevation returns to baseline within 12-24 hours (4). Infants who remain critically ill and require PN also do not appear to require increased energy intakes. Jaksic et al. measured energy expenditure in eight non-ventilated surgical neonates (gastroschisis, atresia, volvulus) on postoperative day 16 (± 12, SD) and compared them with ten infants on extracorporeal life support studied at 7 ± 3 days of age (5). There were no differences in energy expenditure between the groups (53 ± 5 vs. 55 ± 20 kcal ⭈ kg−1 ⭈ d−1) (5). This level of energy expenditure is comparable to that of “nonstressed” infants (see below). The similarity in energy expenditure was present despite the finding that IL-6 and C-reactive protein levels were significantly greater in the extracorporeal life support group reflecting an increased degree of illness, and ultimately, mortality (30% vs. 0) (5). This same group of investigators recently has shown that preterm infants (25 weeks gestation) behave similarly (6). Lloyd recently reviewed the data regarding energy requirements in surgical newborns and children (7). The evidence reveals that the increase in energy expenditure associated with surgery only lasts for 24 hours after the procedure (7). Estimating energy expenditure during the first 24 hours after surgery will overestimate the energy requirements for the entire postoperative period, potentially resulting in excess energy intake with its potential consequences (see below). Energy requirements also have been examined in nonoperated children in the intensive care unit. In a group of

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

mechanically ventilated critically ill children (N ⳱ 33; 6 ± 5 years of age) whose mean length of stay in the intensive care unit was approximately 2 weeks, measured energy expenditure was only about 8 kcal ⭈ kg−1 ⭈ d−1 (17%) above expected based on more recent data (8,9). Joosten et al. found even smaller increases in energy expenditure in a more heterogeneous group of 36 infants and children in an intensive care unit (10). Finally, Turi et al. measured energy expenditure in 21 patients in the intensive care unit with systemic inflammatory response syndrome or sepsis and compared their energy expenditures to a group of hospitalized control children (11). No differences were noted between the groups (11). Taken together, these data support the idea that in most cases there is little need to provide post operative or critically ill infants and children with much more than their resting energy expenditure, which in most cases will be an amount similar to their basal metabolic rate. The information in Table 1 can be used to calculate basal metabolic rate in normal children (9). A recent report by Duro et al. used the Enhanced Metabolic Testing Activity Chamber (EMTAC) to predict REE in 50 normal infants up to 7 months of age with a maximum weight of 9 kg (12). Their data suggest that for this age group the values of Shofield may be an underestimation (9,12). However, part of this difference may be explained by the fact that the values for Shofield are for BMR and those of Duro et al. are for REE (9,12). The equations described by Duro et al. are: REE (kcal/d) ⳱ [84.5 x Weight (kg)] – 117.33 (R2 ⳱ 0.65, P < 0.01) The prediction is improved somewhat by including length: REE (kcal/d) ⳱ [10.12 x Length (cm)] + [61.02 x Weight (kg)] – 605.08 (R2 ⳱ 0.7, P < 0.01) It is evident in the study by Jaksic et al. that there may be significant interindividual variations in energy expenditure (5,7). This should not be surprising, as even among normal newborns energy requirements do not fall within narrow margins (13). White et al. have shown that in pediatric intensive care unit patients that there is little within-day variation in energy expenditure but day-today variation is as high as 21 ± 16% (mean ± SD) (14). Ideally, energy expenditure should be measured in critically ill patients. However, it may not be practical because of the expensive equipment and the expertise required. At the very least, one should be flexible regarding the “appropriate” energy intake and monitor the patient for weight gain (fluid vs. tissue) and evidence of overfeeding (e.g., CO2 production; see below). Two recent studies may be of value in calculating resting energy expenditure for surgical infants and intensive care patients.

PARENTERAL NUTRITION IN INFANTS AND CHILDREN TABLE 1. Equations for estimating basal metabolic rate (MJ/day)* Age (years)

Male

0.4% of the total fat contains particles > 5 microns in size (162,163). What patients are candidates for PN admixtures? It may be more important to ask whether there are PN

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

mixtures that are inappropriate for certain patients. Because of the limitations in the amount of calcium and phosphorus that can be used in PN admixtures (even more severe than in non- admixture PN), their use may be inappropriate in young infants. One report has suggested that PN admixtures can be used in former preterm infants and newborns, however, in this study it was not clear that the PN admixtures had been validated as stable nor was there stability testing reported after the admixtures were prepared (169). Because the emulsion is opaque (as a result of fat emulsion) a precipitate may be invisible to the naked eye. The calcium and phosphorus requirements for neonates is 3–4 mEq ⭈ kg−1 ⭈ d−1 and 1–2 mmol ⭈ kg−1 ⭈ d−1, respectively (162). Based upon studies using a commonly employed pediatric amino acid solution (TrophAmine, B. Braun Medical, Inc., Melsungen, Germany) the PN admixture would provide inadequate amounts of these minerals (170). As an infant approached 4–6 months of age it is conceivable that their calcium and phosphorus needs could be met using a PN admixture (161,162,170). However, it must be stressed that this is dependent upon the amino acid, glucose, intravenous fat emulsion, calcium, and phosphorus products (as well as the other components) used as well as the mixing protocol. If a PN admixture is to be used in an infant less than a year of age, it should be done with particularly careful consideration regarding mineral requirements. PN admixtures only should be used in patients who are clinically stable. Changes in the formulation of PN admixtures are more costly than changes in traditional (two-in-one) PN because of the wastage of both the dextrose/amino acid and its components, and the intravenous fat emulsion. A number of questions remain about PN admixtures. These include the appropriate dosing of some vitamins, the true risks related to particulate matter and fat droplet size, and drug compatibilities. For example, there is less loss of fat-soluble vitamins such as vitamin A than in traditional (two-in-one) PN because the intravenous fat emulsion is protective (see below) (171). Some drugs are compatible with PN admixtures but not with traditional PN and visa versa (172).

PARENTERAL NUTRITION IN INFANTS AND CHILDREN TABLE 6. Adult and children (>11 years of age) parenteral multivitamin preparations Vitamin

Infuvite™* (Sabex)

M.V.I.-12™ (aaiPharma)

A (IU) D IU (␮g) E (IU) K (␮g) C (mg) Thiamin (mg) Riboflavin (mg) Niacin (mg) Pyridoxine (mg) Folate (␮g) B12 (␮g) Pantothenic Acid (mg) Biotin (␮g)

3300 IU 200 (5) 10 150 200 6 3.6 40 6 600 5 15 60

3300 200 (5) 10 — 200 3 3.6 40 4 400 5 15 60

* Conforms to the FDA amended formula for adult multivitamin preparations (165).

VITAMINS Guidelines for pediatric parenteral vitamin and mineral supplementation have been previously reviewed, albeit quite some time ago (173). Despite subsequent publications that have provided additional support for these recommendations, there has not been a recent significant evaluation or reformulation of parenteral vitamin products for premature infants, infants, or children (less than 11 years of age) (174,175). Recommendations from the 1998 National Advisory Group on Standards and Practice Guidelines emphasized the need for establishing optimal trace element and vitamin formulations for both adult and pediatric patients

599

(162). In April 2000, the Food and Drug Administration amended the adult multivitamin formulation to bring it into accordance with the 1988 recommendations of the American Medical Association-FDA Public Workshop Committee (Table 6) (176). To date, the optimal parenteral vitamin and mineral requirements for children and neonates have not been determined and a parenteral multivitamin formulation specifically for preterm infants has not been developed. While there are several parenteral vitamin preparations available for older children (> 11 years) and adults (Table 6), few are available for neonates and children. InFuvite Pediatric™ (Sabex, Inc., Boucherville, Canada) and M.V.I. Pediatric™ (aaiPharma, Inc., Wilmington, NC) are approved for use in prematures, infants, and children < 11 years (Table 7). The adult formulations are not recommended for use in low birth weight infants less than 1500 grams because of concerns about the toxicity of the propylene glycol and polysorbate additives (177,178). Limitations in the availability of the pediatric products have made children vulnerable to shortages (179). Table 7 gives the doses of vitamins recommended for infants by the American Society of Clinical Nutrition Subcommittee on Pediatric Parenteral Nutrient Requirements (). This recommendation differs from the package inserts (180). Clearly, there are patients whose needs do not fit the current vitamin formulations. For example, preterm infants, children with liver or renal disease or short bowel syndrome, or who are severely malnourished require close attention to vitamin nutriture. Some adult parenteral vitamin products that are used for children >11 years of age may put younger patients on long term PN at risk for excessive vitamin intakes (see Table 6).

TABLE 7. Infant and child parenteral multivitamin requirements and commercial preparations*

Vitamin (amount/d)

Best estimate# preterm infant

Pediatric parenteral multivitamins# 2.5 kg–11 yrs 100% of the vial (5 ml) 700 80 400 7 200 1.2 1.4 17 1 140 1 5 20

* Infuvite Pediatric™/MVI-Pediatric™. # See reference (180). ** 500 ␮g ⳱ 1643 IU. ## 1 mg ⳱ 1 IU.

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

600

R. J. SHULMAN AND S. PHILLIPS Vitamin K

One potential problem with the introduction of vitamin K into the new adult formulations is the possible interference with anticoagulants such as warfarin. Additionally, intravenous fat emulsions contain vitamin K in varying amounts (approximately 13–70 ␮g/dL) that can make titrating anticoagulant therapy even more problematic (181,182). Vitamin A A recent Cochrane review suggests that an increased vitamin A intake (via the intramuscular route) is beneficial in the preterm infant (183). Whether dosing of vitamin A in PN will provide a similar benefit is unclear. Dosing of vitamin A is complicated by its adherence to the bags and tubing and from photodegradation (184). Evidence suggests that when administered in the glucose/amino acid solution the current recommendation for vitamin A is too low (162,185,186). Loss of several lipid soluble (and water-soluble) vitamins occurs during their delivery in PN (187,188). Evidence suggests that using dark tubing and placing fatsoluble vitamins into the intravenous fat emulsion (see PN Admixtures, above) mitigates the losses of these vitamins (189–192,230). However, multivitamin manufacturers in the US do not recommend mixing vitamins in lipid emulsions (see package inserts). Obviously it is commonly done in the case of PN admixtures (see above). More research in this area is vital. Clinical Implications Our knowledge regarding the appropriateness of current vitamin preparations and intake levels is less than optimal. Prudence would suggest checking vitamin levels (especially vitamin A and possibly riboflavin) in patients on long term PN because of potential losses in the administration set, particularly when enteral intake and/or absorption is poor (191). Consideration also should be given to monitoring fat-soluble vitamin levels in patients whose vitamins are placed into the lipid emulsion. Carnitine The use of carnitine in PN for preterm infants recently has been the subject of a Cochrane Database review (193). Using weight gain as a primary outcome, there was no apparent effect of carnitine supplementation (193). However, the studies available did not assess the use of carnitine in long term PN, a situation that is more likely to result in carnitine deficiency. Tissue carnitine levels decline in infants receiving carnitine-free PN (194). An adult patient who received PN for over a year

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

has been described with symptomatic carnitine deficiency although the level of enteral intake was not described (195). It is possible that the effects of carnitine deficiency in patients on long term PN may be too subtle to be identified easily but are still metabolically relevant given the central role of carnitine in metabolism (196). Addition of carnitine does enhance intravenous fat emulsion oxidation although it has been argued that the increase may not be clinically relevant (193). In long term PN patients it may be reasonable to follow the suggestion that carnitine (as pure L-carnitine) be added at a dose of 2 to 5 mg ⭈ kg−1 ⭈ d−1 and that plasma and red blood cell concentrations be measured at baseline and at 4-month intervals until levels have stabilized, then yearly thereafter (196). Too high a carnitine dose may be detrimental (196).

Iron Parenteral nutrition patients at risk for iron deficiency are premature infants, long-term PN patients with little or no enteral intake, and patients with significant malabsorption or fluid losses. Iron may be administered orally, intramuscularly, or parenterally. Parenteral iron can be infused as an intermittent intravenous infusion or as a diluted total dose infusion. Iron dextran has been used in children (197–200). Despite its fairly widespread use, total dose infusion is not approved by the Food and Drug Administration. The use of iron dextran can be complicated by adverse reactions including a small but real risk of anaphylaxis. A test dose is recommended prior to administration of the required amount (see package insert). Iron dextran in PN can be efficacious (201,202). Although often employed in PN, the stability of iron dextran and its potential interaction with other nutrients remain as concerns (202). Its use in PN admixtures should be undertaken with extreme caution if at all (163). Until recently, iron dextran was the only formulation available for parenteral iron therapy. Two new iron products free of dextran have recently been approved for use in the United States. Sodium ferric gluconate complex in sucrose (Ferrlecit, Watson, Inc., Morristown, NJ) has been used extensively in Europe and became available for use in the United States in 1999. Iron sucrose (Venofer, Luitpold Pharmaceuticals, Inc., Shirley, NY) also previously available in Europe was introduced in late 2000. These products appear to be safe and have fewer side effects than those of iron dextran, although like iron dextran, they can cause hypotension (203,204). While the safety and efficacy of these products has not been extensively studied nor are they approved by the FDA for use in children, there is evidence that they are safe and effective in children (205).

PARENTERAL NUTRITION IN INFANTS AND CHILDREN Trace Minerals Trace mineral requirements and metabolism in general, including concerns regarding aluminum contamination and toxicity, recently have been reviewed elsewhere (180,206–209). We briefly will touch on current issues regarding specific trace minerals in PN. First, it should be remembered that PN solutions usually contain some of the various trace minerals due to contamination of the components (e.g., amino acids, calcium gluconate, multivitamins) with trace metals (210). In one report Zn, Cu, Mn, and Se were found in greatest concentration but contamination will vary depending on the commercial products used to prepare the PN (210). Heat and storage time can modestly reduce the levels of some trace elements as well (210). The importance of Se as an antioxidant is well known. A recent report suggests that the recommended intake of Se for preterm infants (N ⳱ 29, gestational age 26 weeks) may not be adequate for all infants (211). In contrast, a study of children and adults on long term PN suggested Se status is well maintained even with low or no Se intake (212). However, in this study there was great variability in Se serum levels as well as glutathione peroxidase activity with some patients actually showing deficiency (212). Given the reports of heart failure because of Se deficiency and our limited knowledge regarding adequacy of intake, it is prudent to check serum Se levels in the short term (particularly in preterm infants) and serum Se levels and glutathione peroxidase activity in older infants and adolescents (207,211–213). Some studies have suggested that chromium toxicity may be a concern. Mouser et al. noted in infants and children (up to 12 years of age) on long term PN that serum Cr levels were elevated despite following the recommended intake (180,214). These data are consistent with other studies and imply that the addition of Cr to PN at recommended levels in combination with Cr contamination in the PN fluids raises serum Cr levels above that desired (210,215–217). Again, patients on long term PN should be closely observed. Whether the impaired glomerular filtration rate noted in children on long term PN is related to Cr excess requires further investigation (217). Zn has the widest range of functions of all the trace minerals and is particularly important in wound healing and immune function. Despite its intentional and nonintentional addition to PN, many patients are at risk for Zn deficiency because of the number of predisposing clinical situations (206,207). For example, patients with gastrointestinal losses due to diarrhea, ileostomies, or even nasogastric suction are at risk for Zn deficiency because of the high Zn content of gastrointestinal fluids (218). Serum Zn levels, although not entirely reliable, are easily obtained and Zn intake can be titrated appropriately.

601

Of great concern are the numerous observations regarding Mn retention. This essential trace mineral normally is excreted in bile and has a special affinity for the extrapyramidal system (206,207). Mn often contaminates PN solution components in amounts sufficient for daily requirements (206,207,219). Manganese intoxication causes parkinsonian-like symptoms with muscular weakness, stiffness, tremors, ataxia, abnormal gait, asthenia, and difficulty with speech (220). Psychological changes have been reported including mental irritability, headaches, nervousness, compulsive actions, and hallucinations (220). Manganese accumulation can be detected in the basal ganglia as symmetric, increased signal intensity on T-1 weighted magnetic resonance images (220,221). These changes are reversible when Mn is removed from the PN (222,223). Clinical symptoms are usually but not always reversible (224). It is not surprising that Mn accumulates in individuals with cholestatic liver disease given its hepatobiliary secretion (220). Of particular concern for pediatric patients is the widely held suspicion that Mn accumulation in patients on PN may cause cholestasis. A recent study randomized 244 children on PN to receive either 1.0 (Group 1) or 0.0182 ␮mol ⭈ kg−1 ⭈ d−1 (Group 2) of Mn (224). When all patients were considered, those in Group 1 showed a trend towards higher peak manganese and direct bilirubin levels. The two groups did not differ in the occurrence of cholestasis but Group 1 patients showed a trend towards increased incidence and severity of hyperbilirubinemia (225). Of the 160 children who received >75% of their daily fluid intake from PN for >14 days, peak whole blood manganese and peak serum direct bilirubin concentrations were significantly higher in Group 1. Significantly more infants in Group 1 developed a more severe degree of direct hyperbilirubinemia (225). These data implicate Mn as a contributor to the development and/or the severity of PN-associated cholestasis (225). Based on these and other data, it is suggested that patients with any cholestasis (some would say any liver disease) should not receive Mn in their PN (220,225,226). Others would go so far as to say that Mn should not be given to individuals on PN for 30 days (207,226). There is disagreement regarding the best measure of Mn status (220). Whole blood Mn appears to correlate (in adults) with MRI-documented Mn deposition (226). Most investigators use whole blood Mn, red blood cell Mn, or Mn superoxide dismutase as measures of tissue deposition (220). SUMMARY Our knowledge regarding PN reflects the same pattern we find in all of medicine: what we do is based part upon science and part upon best judgment. It turns out that

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

602

R. J. SHULMAN AND S. PHILLIPS

practices that seem clear (i.e., based on science), often turn out to be wrong or not so clear. We now realize that aggressive nutritional support carries risks. The risk of infection may be related more to hyperglycemia (or hypertriglyceridemia) than to PN per se, and essential fatty acid requirements may not be as well defined as previously thought. When it seems as if knowledge is for naught, the following quote comes to mind: “I reserve the right to be smarter tomorrow than I am today” (227,228). In a way, we are victims of our own success. PN can provide effective nutritional support but, as is evident from this review, there are large holes in our knowledge and little likelihood of abundant future research to fill them. For the most part, these deficiencies in knowledge are more problematic for pediatric patients than for adults. Unfortunately, because pediatric PN makes up such a small share of the market, manufacturers are reluctant to spend the resources to carry out the studies needed to define the child-specific impact of new lipid emulsions, reformulated vitamins, etc. Money spent on these questions would not be recouped quickly. The responsibility for improving PN safety and efficacy lies with all concerned: industry, funding agencies, physicians, pharmacists, nurses, and nutritionists. PN is still the life-saving, far-from-perfect therapy it has been for over 40 years. Like all therapies it must be used and monitored appropriately. Using it means more than just checking the patient’s weight, and monitoring now means more than checking the serum electrolytes. Checking the box on the order sheet will never be enough. Acknowledgments: The authors thank Dr. W.C. Heird for his very helpful comments. This study was supported by the National Institute of Child Health and Human Development, Grant No. RO1 NR05337-01A2, the Daffy’s and Henriksen Foundations, and the USDA/ARS under Cooperative Agreement No. 58-6250-1-003. This work is a publication of the USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX. The contents of this publication do not necessarily reflect the views or policies of the USDA, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government.

6.

7. 8.

9. 10.

11.

12.

13. 14.

15.

16. 17.

18.

19.

20. 21. 22.

23.

24.

REFERENCES 25. 1. Spencer CT, Compher C. The development of TPN: an interview with pioneer surgical nutritionist Jonathan E. Rhoads, MD. J Am Diet Assoc 2001:101;747–50. 2. Barry Lepatner 1983 in testimony before Congress; personal communication 2002. 3. Forchelli ML, Gura KM, Shulman RJ. Metabolism estimation, recommended energy, protein and fluid needs by age in pediatric nutrition in your pocket. Corkins MR, Shulman RJ, ed., American Society for Parenteral and Enteral Nutrition (in press). 4. Pierro A, Jones MO, Donnell SC. Total parenteral nutrition in surgical infants. Biochem Soc Trans 1998 May;26:131–6. 5. Jaksic T, Shew SB, Keshen TH, Dzakovic A, Jahoor F. Do criti-

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

26.

27.

28.

cally ill surgical neonates have increased energy expenditure? J Pediatr Surg 2001;36:63–7. Garza JJ, Shew SB, Keshen TH, Dzakovic A, Jahoor F, Jaksic T. Energy expenditure in ill premature neonates. J Pediatr Surg 2002;37:289–93. Lloyd DA. Energy requirements of surgical newborn infants receiving parenteral nutrition. Nutrition 1998;14:101–04. Coss-Bu JA, Klish WJ, Walding D, Stein F, Smith EO, and Jefferson LS. Energy metabolism, nitrogen balance, and substrate utilization in critically ill children 1-3. Am J Clin Nutr 2001;74: 664–69. Schofield W. Predicting basal metabolic rate, new standards and review of previous work. Hum Nutr Clin Nutr 1985;39C S1;5–41. Koen FM, Joosten KFM, Verhoeven JJ, and Hazelzet JA. Energy Expenditure and Substrate Utilization in Mechanically Ventilated Children. Nutrition 1999;15:444–48. Turi RA, Petros AJ, Eaton S, Fasoli L, Powis M, Basu R, Spitz L, Pierro A. Energy metabolism of infants and children with systemic inflammatory response syndrome and sepsis. Annals of Surgery 2001; 233: 581–87. Duro D, Rising R, Cole C, Valois S, Cedillo M, Lifshitz F. New equations for calculating the components of energy expenditure in infants. J Pediatr 2002;140:534–9. http://www.nal.usda.gov/fnic/dga/rda.pdf White MS, Shepherd RW, McEniery JA. Energy expenditure measurements in ventilated critically ill children: within- and between-day variability. J Parenter Enteral Nutr 1999;23:300–4. Pierro A, Jpnes MO, Hammond P, Donnell SC, Lloyd DA. A new equation to predict the resting energy expenditure of surgical infants. J Pediatr Surg 1994;29:1103–8. Pierro A, Jones MO, Donnell SC. Total parenteral nutrition in surgical infants. Biochem Soc Trans 1998;26:131–6. White MS, Shepherd RW, McEniery JA. Energy expenditure in 100 ventilated, critically ill children: Improving the accuracy of predictive equations. Crit Care Med 2000; 28: 2307-2312. Dietz WH, Bandini LG, Schoeller DA. Estimates of metabolic rate in obese and nonobese adolescents. J Pediatr 1991;118: 146–49. Mondejar EF, Duro Lombardo M, Perez de la Cruz AJ, Merida Morales A, Torres Ruiz JM, Ferron Orihuela JA. Variations in oxygen consumption and carbon dioxide production during parenteral nutrition. Intensive Care Med 1982;8:169–72. Elwyn DH, Askanazi J, Kinney JM, Gump FE. Kinetics of energy substrates. Acta Chir Scand Suppl 1981;507:209–19. Koretz RL, Lipman TO, Klein S. AGA Technical Review on Parenteral Nutrition. Gastroenterology 2001;121:970–1001. Wilson DC, Cairns P, Halliday HL, Reid M, McClure G, Dodge JA. Randomised controlled trial of an aggressive nutritional regimen in sick very low birthweight infants. Arch Dis Child Fetal Neonatal Ed 1997;77:F4–11. Kalhan SC, Kilic I. Carbohydrate as nutrient in the infant and child: range of acceptable intake. Europ J Clin Nutr 1999;53: S94–S100. Denne SC, Karn CA, Wang J, Liechty EA. Effect of intravenous glucose and lipid on proteolysis and glucose production in normal newborns. Am J Physiol 1995;269:E361–7. Sunehag AL, Haymond MW, Schanler RJ, Reeds PJ, Bier DM. Gluconeogenesis in very low birth weight infants receiving total parenteral nutrition. Diabetes 1999;48:791–800. Lafeber HN, Sulkers EJ, Chapman TE, Sauer PJ. Glucose production and oxidation in preterm infants during total parenteral nutrition. Pediatr Res 1990;28:153–7. Jones MO, Pierro A, Hammond P, Nunn A, Lloyd DA. Glucose utilization in the surgical newborn infant receiving total parenteral nutrition. J Pediatr Surg 1993;28:1121–5. Nose O, Tipton JR, Ament ME. Effect of the energy source on changes in energy expenditure, respiratory quotient, and nitrogen balance during total parenteral nutrition in children. Pediatr Res 1987;21:538–41.

PARENTERAL NUTRITION IN INFANTS AND CHILDREN 29. Bresson JL, Narcy P, Putet G, Ricour C. Energy substrate utilization in infants receiving total parenteral nutrition with different glucose to fat ratios. Pediatr Res 1989;25:645–8. 30. Sheridan RL, Yu YM, Prelack K, Young VR, Burke JF, Tompkins RG. Maximal parenteral glucose oxidation in hypermetabolic young children: a stable isotope study. J Parent Ent Nutr 1998;22:212–6. 31. Schears GJ, Deutschman CS. Common nutritional issues in pediatric and adult critical care medicine. Crit Care Clinic 1997; 13:669–90. 32. Cerra FB, Benitez MR, Blackburn GL, Irwin RS, Jeejeebhoy K, Katz DP, Pingleton SK, Pomposelli J, Rombeau JL, Shronts E, Wolfe RR, Zaloga GP. Applied nutrition in ICU patients. Chest 1997;111:769–78. 33. Chan S, McCowen KC, Blackburn GL. Nutrition Management in the ICU. Chest 1999; 115: 145–48. 34. Klein CJ, Stanek GS, Wiles CE 3rd. Overfeeding macronutrients to critically ill adults: metabolic complications. J Am Diet Assoc 1998;98:795–806. 35. Shikora SA, Benotti PN. Nutritional support of the mechanically ventilated patient. Respir Care Clin N Am 1997;3: 69–90. 36. Robin AP, Carpentier YA, Askanazi J, Nordenstrom J, Kinney JM. Metabolic consequences of hypercaloric glucose infusions. Acta Chir Belg 1981;80:133–40. 37. Elwyn DH, Askanazi J, Kinney JM, Gump FE. Kinetics of energy substrates. Acta Chir Scand Suppl 1981;507:209–19. 38. Talpers SS, Romberger DJ, Bunce SB, Pingleton SK. Nutritionally associated increased carbon dioxide production. Excess total calories vs high proportion of carbohydrate calories. Chest 1992; 102:551–5. 39. Askanazi J, Weissman C, LaSala PA, Milic-Emili J, Kinney JM. Effect of protein intake on ventilatory drive. Anesthesiology 1984;60:106–10. 40. Rodriguez JL, Askanazi J, Weissman C, Hensle TW, Rosenbaum SH, Kinney JM. Ventilatory and metabolic effects of glucose infusions. Chest 1985;88:512–8. 41. Angelillo VA, Bedi S, Durfee D, Dahl J, Patterson AJ, O’Donohue WJ Jr. Effects of low and high carbohydrate feedings in ambulatory patients with chronic obstructive pulmonary disease and chronic hypercapnia. Ann Intern Med 1985;103:883–5. 42. Askanazi J, Rosenbaum SH, Hyman AI, Silverberg PA, MilicEmili J, Kinney JM. Respiratory changes induced by the large glucose loads of total parenteral nutrition. JAMA 1980;243: 1444–7. 43. Burke JF, Wolfe RR, Mullany CJ, Mathews DE, Bier DM. Glucose requirements following burn injury. Parameters of optimal glucose infusion and possible hepatic and respiratory abnormalities following excessive glucose intake. Ann Surg 1979;190: 274–85. 44. Tulikoura I, Huikuri K. Morphological fatty changes and function of the liver, serum free fatty acids, and triglycerides during parenteral nutrition. Scand J Gastroenterol 1982;17:177–85. 45. Asle Aarsland, David Chinkes, and Robert R. Wolfe. Contributions of De Novo Synthesis of Fatty Acids to Total VLDLTriglyceride Secretion during Prolonged Hyperglycemia/Hyperinsulinemia in Normal Man. J Clin Invest 1996;98: 2008–17. 46. Perioperative total parenteral nutrition in surgical patients. The Veterans Affairs Total Parenteral Nutrition Cooperative Study group. N Engl J Med 1991;325:525–32. 47. Kudsk KA, Croce MA, Fabian TC, Minard G, Tolley EA, Poret HA, Kuhl MR, Brown RO. Enteral versus parenteral feeding. Effects on septic morbidity after blunt and penetrating abdominal trauma. Ann Surg 1992;215:503-11. 48. Moore FA, Moore EE, Jones TN, McCroskey BL, Peterson VM. TEN versus TPN following major abdominal trauma–reduced septic morbidity. J Trauma 1989;29:916–22. 49. Moore FA, Feliciano DV, Andrassy RJ, McArdle AH, Booth FV, Morgenstein-Wagner TB, Kellum JM Jr, Welling RE, Moore EE.

50. 51.

52.

53.

54.

55.

56.

57.

58. 59. 60. 61.

62.

63.

64. 65.

66.

67.

68.

69.

70.

603

Early enteral feeding, compared with parenteral, reduces postoperative septic complications. The results of a meta-analysis. Ann Surg 1992;216:172–83. Bistrian BR. Hyperglycemia and infection: which is the chicken and which is the egg? J Parenter Enteral Nutr 2001;25:180–1. Engelich G, Wright DG, Hartshorn KL. Acquired disorders of phagocyte function complicating medical and surgical illnesses. Clin Infect Dis 2001;33:2040–8. Khaodhiar L, McCowen K, Bistrian B. Perioperative hyperglycemia, infection or risk? Curr Opin Clin Nutr Metab Care 1999; 2:79–82. Gore DC, Chinkes D, Heggers J. Association of hyperglycemia with increased mortality after severe burn injury. J Trauma 2001; 51:540–44. McCowen KC, Friel C, Sternberg J, Chan S, Forse RA, Burke PA, Bistrian BR. Hypocaloric total parenteral nutrition: effectiveness in prevention of hyperglycemia and infectious complications–a randomized clinical trial. Crit Care Med 2000;28: 3606–11. Cerra FB et al. Applied Nutrition in ICU Patients: A Consensus Statement of the American College of Chest Physicians. Chest 1997;111:769–78. Gutcher GR, Farrell PM. Intravenous infusion of lipid for the prevention of essential fatty acid deficiency in premature infants. Am J Clin Nutr 1991;54:1024–8. Foote KD, MacKinnon MJ, Innis SM. Effect of early introduction of formula vs fat-free parenteral nutrition on essential fatty acid status of preterm infants. Am J Clin Nutr 1991;54:93–7. O’Neill JA Jr, Caldwell MD, Meng HC. Essential fatty acid deficiency in surgical patients. Ann Surg 1977;185:535–42. Farrell PM, Gutcher GR, Palta M, DeMets D. Essential fatty acid deficiency in premature infants. Am J Clin Nutr 1988;48:220–9. Friedman Z. Essential fatty acids revisited. Am J Dis Child 1980; 134:397–408. Mascioli EA, Lopes SM, Champagne C, Driscoll DF. Essential fatty acid deficiency and home total parenteral nutrition patients. Nutrition 1996;12:245–9. Jeppesen PB, Hoy CE, Mortensen PB. Differences in essential fatty acid requirements by enteral and parenteral routes of administration in patients with fat malabsorption. Am J Clin Nutr 1999; 70:78–84. Jeejeebhoy KN, Langer B, Tsallas G, Chu RC, Kuksis A, Anderson GH. Total parenteral nutrition at home: studies in patients surviving 4 months to 5 years. Gastroenterology 1976;71: 943–53. Barr LH, Dunn GD, Brennan MF. Essential fatty acid deficiency during total parenteral nutrition. Ann Surg 1981;193:304–11. Wolfram G, Eckart J, Walther B, Zollner N. Factors influencing essential fatty acid requirement in total parenteral nutrition (TPN). J Parenter Enteral Nutr 1978;2:634–9. Anderson TL, Muttart CR, Bieber MA, Nicholson JF, Heird WC. A controlled trial of glucose versus glucose and amino acids in premature infants. J Pediatr 1979;94:947–51. Nube M, Bos LP, vd Boomgaard DM, Hekkens WT. Energy intake and the appearance of 5,8,11-eicosatrienoic acid in serum lipids during parenteral nutrition without fat. J Parenter Enteral Nutr 1982;6:134–9. Spear ML, Stahl GE, Hamosh M, McNelis WG, Richardson LL, Spence V, Polin RA, Pereira GR, Hamosh P. Effect of heparin dose and infusion rate on lipid clearance and bilirubin binding in premature infants receiving intravenous fat emulsions. J Pediatr 1988;112:94–8. Kao LC, Cheng MH, Warburton D. Triglycerides, free fatty acids, free fatty acids/albumin molar ratio, and cholesterol levels in serum of neonates receiving long-term lipid infusions: controlled trial of continuous and intermittent regimens. J Pediatr 1984;104: 429–35. Haumont D, Deckelbaum RJ, Richelle M, Dahlan W, Coussaert E, Bihain BE, Carpentier YA. Plasma lipid and plasma lipopro-

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

604

71.

72. 73.

74.

75. 76.

77.

78.

79.

80.

81.

82. 83.

84.

85. 86.

87.

88.

89. 90. 91.

92.

R. J. SHULMAN AND S. PHILLIPS tein concentrations in low birth weight infants given parenteral nutrition with twenty or ten percent lipid emulsion. J Pediatr 1989;115:787–93. Haumont D, Richelle M, Deckelbaum RJ, Coussaert E, Carpentier YA. Effect of liposomal content of lipid emulsions on plasma lipid concentrations in low birth weight infants receiving parenteral nutrition. J Pediatr 1992;121:759–63. Putet G. Lipid metabolism of the micropremie. Clin Perinatol 2000;27:57–69. Rossner S, Eklund B, Freyschuss U, Hallberg D, Kaijser L, Olsson A. Elimination of parenterally administered fat. Studies on removal sites for intralipid in normo-and hyperlipidaemic subjects. Acta Chir Scand Suppl 1976;466:56–7. Amin SB, Sinkin RA, McDermott MP, Kendig JW. Lipid intolerance in neonates receiving dexamethasone for bronchopulmonary dysplasia. Arch Pediatr Adolesc Med 1999;153:795–800. Hasselgren PO. Catabolic response to stress and injury: implications for regulation. World J Surg 2000;24:1452–9. Plank LD, Hill GL. Sequential metabolic changes following induction of systemic inflammatory response in patients with severe sepsis or major blunt trauma. World J Surg 2000;24:630–8. Dahn MS, Kirkpatrick JR, Blasier R. Alterations in the metabolism of exogenous lipid associated with sepsis. J Parenter Enteral Nutr 1984;8:169–73. Druml W, Fischer M, Ratheiser K. Use of intravenous lipids in critically ill patients with sepsis without and with hepatic failure. J Parenter Ent Nutr 1998;22:217–23. Nordenstrom J, Carpentier YA, Askanazi J, Robin AP, Elwyn DH, Hensle TW, Kinney JM. Metabolic utilization of intravenous fat emulsion during total parenteral nutrition. Ann Surg 1982;196: 221–31. Lundell K-H, Sabel K-G, Eriksson BO. Plasma metabolites after a lipid load in infants with congenital heart disease. Acta Paediatr 1999;88:718–23. Zaidan H, Dhanireddy R, Hamosh M, Pramanik AK, Chowdhry P, Hamosh P. Effect of continuous heparin administration on Intralipid clearing in very low-birth-weight infants. J Pediatr 1982;101:599–602. Griffin EA, Bryan MH, Angel A. Variations in intralipid tolerance in newborn infants. Pediatr Res 1983;17:478–81. Benderly A, Rosenthal E, Levi J, Brook G. Effect of heparin on lipoprotein profile during parenteral fat infusions. J Parenter Enteral Nutr 1983;7:37–9. Forget PP, Fernandes J, Begemann PH. Utilization of fat emulsion during total parenteral nutrition in children. Acta Paediatr Scand 1975;64:377–84. Adamkin DH, Gelke KN, Andrews BF. Fat emulsions and hypertriglyceridemia. J Parenter Enteral Nutr 1984;8:563-7. D’Harlingue A, Hopper AO, Stevenson DK, Shahin SM, Kerner JA Jr. Limited value of nephelometry in monitoring the administration of intravenous fat in neonates. J Parenter Enteral Nutr 1983;7:55–8. Zaidan H, Dhanireddy R, Hamosh M, Bengtsson-Olivecrona G, Hamosh P. Lipid clearing in premature infants during continuous heparin infusion: role of circulating lipases. Pediatr Res 1985;19: 23–5. Paust H, Park W, Brosicke H, Knoblach G, Helge H. Fat utilization in newborn infants with and without heparin administration. Comparative study with the 13C-triolein breath test. Infusionsther Klin Ernahr 1985;12:85–7. Skeie B, Askanazi J, Rothkopf MM. Intravenous fat emulsions and lung function: a review. Crit Care Med 1988;16:183–94. Hageman JR, Hunt CE. Fat emulsions and lung function. Clin Chest Med 1986;7:69-77. Newball HH, Friedewald WT, Roberts B, Levy RI, Lenfant CJ. Effect of elevated triglycerides on the diffusing capacity of man. Am Rev Respir Dis 1975;112:83–8. Wesson DE, Rich RH, Zlotkin SH, Pencharz PB. Fat overload

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

93. 94.

95.

96.

97.

98.

99.

100.

101.

102.

103. 104. 105. 106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

syndrome causing respiratory insufficiency. J Pediatr Surg 1984; 19:777–8. Hwang TL, Huang SL, Chen MF. Effects of intravenous fat emulsion on respiratory failure. Chest 1990;97:934–8. Helbock HJ, Motchnik PA, Ames BN. Toxic hydroperoxides in intravenous lipid emulsions used in preterm infants. Pediatrics 1993;91:83–7. Lavoie JC, Chessex P. The increase in vasomotor tone induced by a parenteral lipid emulsion is linked to an inhibition of prostacyclin production. Free Radic Biol Med 1994;16:795–9. Periera GR, Fox WW, Stanley CA, Baker L, Schwartz JG. Decreased oxygenation and hyperlipemia during intravenous fat infusions in premature infants. Pediatrics 1980;66:26–30. Marks KH, Turner MJ, Rothberg AD. Effect of Intralipid infusion on transcutaneous oxygen and carbon dioxide tension in sick neonates. S Afr Med J 1987;72:389–91. Sun SC, Ventura C, Verasestakul S. Effect of Intralipid-induced lipaemia on the arterial oxygen tension in preterm infants. Resuscitation 1978;6:265–70. Pia Lundman P, Eriksson M, Schenck-Gustafsson K. Transient triglyceridemia decreases vascular reactivity in young, healthy men without risk factors for coronary heart disease. Circulation 1997;96:3266–68. Alwaidh MH, Bowden L, Shaw B, Ryan SW. Randomised trial of effect of delayed intravenous lipid administration on chronic lung disease in preterm neonates. J Pediatr Gastroenterol Nutr 1996; 22:303–6. Waitzberg DL, Lotierzo PH, Logullo AF, Torrinhas RS, Pereira CC, Meier R. Parenteral lipid emulsions and phagocytic systems. Br J Nutr 2002;87:S49–57. Shulman RJ. The effect of Intralipid on the cytotoxicity of leukocytes for herpes simplex virus-infected cells: a clinical concern? J Infect Dis 1983;148:181–2. Adolph M. Lipid emulsions in parenteral nutrition. Ann Nutr Metab 1999;43:1–13. Guillou PJ. The effects of lipids on some aspects of the cellular immune response. Proc Nutr Soc 1993;52:91–100. Pomposelli JJ, Bistrian BR. Is total parenteral nutrition immunosuppressive? New Horiz 1994;2:224–9. Strunk RC, Murrow BW, Thilo E, Kunke KS, Johnson EG. Normal macrophage function in infants receiving Intralipid by lowdose intermittent administration. J Pediatr 1985;106:640–5. Wheeler JG, Boyle RJ, Abramson JS. Intralipid infusion in neonates: effects on polymorphonuclear leukocyte function. J Pediatr Gastroenterol Nutr 1985;4:453–6. Usmani SS, Harper RG, Usmani SF. Effect of a lipid emulsion (Intralipid) on polymorphonuclear leukocyte functions in the neonate. J Pediatr 1988;113:132–6. Dahlstrom KA, Goulet OJ, Roberts RL, Ricour C, Ament ME. Lipid tolerance in children receiving long-term parenteral nutrition: a biochemical and immunologic study. J Pediatr 1988;113: 985–90. Sirota L, Straussberg R, Notti I, Bessler H. Effect of lipid emulsion on IL-2 production by mononuclear cells of newborn infants and adults. Acta Paediatr 1997;86:410–3. Usmani SS, Harper RG, Sia CG, Suntharalingam K, Robeson WR. In vitro effect of Intralipid on polymorphonuclear leukocyte function in the neonate. J Pediatr 1986;109:710–2. English D, Roloff JS, Lukens JN, Parker P, Greene HL, Ghishan FK. Intravenous lipid emulsions and human neutrophil function. J Pediatr 1981;99:913–6. Puri P, Reen DJ, Browne O, Guiney EJ. Immune status of the neonate maintained on total parenteral nutrition. Arch Dis Child 1981;56:283–6. Lenssen P, Bruemmer BA, Bowden RA, Gooley T, Aker SN, Mattson D. Intravenous lipid dose and incidence of bacteremia and fungemia in patients undergoing bone marrow transplantation. Am J Clin Nutr 1998;67:927–33. Avila-Figueroa C, Goldmann DA, Richardson DK, Gray JE, Fer-

PARENTERAL NUTRITION IN INFANTS AND CHILDREN

116. 117.

118.

119.

120.

121. 122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132. 133.

134.

135. 136.

rari A, Freeman J. Intravenous lipid emulsions are the major determinant of coagulase-negative staphylococcal bacteremia in very low birth weight newborns. Pediatr Infect Dis J 1998;17: 10–7. Carpentier YA, Dupont IE. Advances in intravenous lipid emulsions. World J Surg 2000;24:1493–7. Spear ML, Spear M, Cohen AR, Pereira GR. Effect of fat infusions on platelet concentration in premature infants. J Parenter Enteral Nutr 1990;14:165–8. Dahlstrom KA, Goulet OJ, Roberts RL, Ricour C, Ament ME. Lipid tolerance in children receiving long-term parenteral nutrition: a biochemical and immunologic study. J Pediatr 1988;113: 985–90. Bistrian BR, Bothe A Jr, Blackburn GL, DeFriez AI. Low plasma cortisol and hematologic abnormalities associated with essential fatty acid deficiency in man. J Parenter Enteral Nutr 1981;5: 141–4. Shulman RJ, Langston C, Schanler RJ. Pulmonary vascular lipid deposition after administration of intravenous fat to infants. Pediatrics 1987;79:99–102. Heird WC. Amino acids in pediatric and neonatal nutrition. Curr Opin Clin Nutr Metab Care 1998;1:73–8. Brunton JA, Ball RO, Pencharz PB. Current total parenteral nutrition solutions for the neonate are inadequate. Curr Opin Clin Nutr Metab Care 2000;3:299–304. Brown MR, Thunberg BJ, Golub L, Maniscalco WM, Cox C, Shapiro DL. Decreased cholestasis with enteral instead of intravenous protein in the very low-birth-weight infant. J Pediatr Gastroenterol Nutr 1989;9:21–7. National Advisory Group on Standards and Practice Guidelines for Parenteral Nutrition. Safe practices for parenteral nutrition formulations. J Parent Ent Nutr 1998;22:49–66. Brown MR, Thunberg BJ, Golub L, Maniscalco WM. Decreased cholestasis with enteral instead of intravenous protein in the very low-birth-weight infant. J Pediatr Gastroenterol Nutr 1989;9: 21–7. Moss RL, Amii LA. New approaches to understanding the etiology and treatment of total parenteral nutrition-associated cholestasis. Semin Pediatr Surg 1999;8:140–7. Senger H, Boehm G, Beyreiss K, Braun W, Raiha N. Evidence for amino acid induced cholestasis in very-low-birth-weight infants with increasing enteral protein intake. Acta Paediatr Scand 1986;75:724–8. Woolf GM, Miller C, Kurian R, Jeejeebhoy KN. Nutritional absorption in short bowel syndrome. Evaluation of fluid, calorie, and divalent cation requirements. Dig Dis Sci 1987;32:8–15. Chin SE, Shepherd RW, Thomas BJ, Cleghorn GJ, Patrick MK, Wilcox JA, Ong TH, Lynch SV, Strong R. Nutritional support in children with end-stage liver disease: a randomized crossover trial of a branched-chain amino acid supplement. Am J Clin Nutr 1992;56:158–63. Fabbri A, Magrini N, Bianchi G, Zoli M, Marchesini G. Overview of randomized clinical trials of oral branched-chain amino acid treatment in chronic hepatic encephalopathy. J Parenter Enteral Nutr 1996;20:159–64. Marchesini G, Bianchi G, Rossi B, Brizi M, Melchionda N. Nutritional treatment with branched-chain amino acids in advanced liver cirrhosis. J Gastroenterol 2000;35 Suppl 12:7–12. Heyman MB. General and specialized parenteral amino acid formulations for nutrition support. J Am Diet Assoc 1990;90:401–8. Koretz RL. Does nutritional intervention in protein-energy malnutrition improve morbidity or mortality? J Ren Nutr 1999;9: 119–21. Brewer ED. Pediatric experience with intradialytic parenteral nutrition and supplemental tube feeding. Am J Kidney Dis 1999;33: 205–7. MacFie J. Cyclic parenteral nutrition. Nutrition 1997;13:46–55. Friel C, Bistrian B. Cycled total parenteral nutrition: is it more effective? Am J Clin Nutr 1997;65:1078–9.

605

137. Takehara H, Hino M, Kameoka K. A new method of total parenteral nutrition for surgical neonates: it is possible that cyclic TPN prevents intrahepatic cholestasis. Tokushima J Exp Med 1990;37:97–102. 138. Hwang TL, Lue MC, Chen LL. Early use of cyclic TPN prevents further deterioration of liver functions for the TPN patients with impaired liver function. Hepatogastroenterology 2000;47: 1347–50. 139. Teitelbaum DH. Parenteral nutrition-associated cholestasis. Curr Opin Pediatr 1997;9:270–5. 140. Sokol RJ. Total parenteral nutrition-related liver disease. Acta Paed Sin 1997;38:418–28. 141. Kaufman SS. Prevention of parenteral nutrition-associated liver disease in children. Pediatr Transplant 2002;6:37–42. 142. Crook MA, Hally V, Panteli JV. The importance of the refeeding syndrome. Nutrition 2001;17:632–7. 143. Brooks MJ, Melnik G. The refeeding syndrome: an approach to understanding its complications and preventing its occurrence. Pharmacotherapy 1995;15:713–26. 144. Kimuru S, Nose O, Seino Y. Effects of alternate and simultaneous administrations of calcium and phosphorous on calcium metabolism in children receiving total parenteral nutrition. J Parent Ent Nutr 1986;10:513–16. 145. Fitzgerald KA, MacKay MW. Calcium and phosphate solubility in neonatal parenteral nutrient solutions containing TrophAmine. Am J Hosp Pharm 1986;43:88–93. 146. Lenz GT, Mikrut BA. Calcium and phosphate solubility in neonatal parenteral nutrient solutions containing Aminosyn-PF or TrophAmine. Am J Hosp Pharm 1988;45:2367–71. 147. Dunham B, Marcuard S, Khazanie PG, Meade G, Craft T, Nichols K. The solubility of calcium and phosphorus in neonatal total parenteral nutrition solutions. J Parenter Enteral Nutr 1991; 15:608–11. 148. Porcelli PJ, Block SM. Increased parenteral nutrition calcium and phosphorus for very-low-birth-weight infants using computer software assisted ordering. J Am Coll Nutr 1997;16:283–7. 149. Laine L, Shulman RJ, Pitre D, Lifschitz CH, Adams J. Cysteine usage increases the need for acetate in neonates who receive total parenteral nutrition. Am J Clin Nutr 1991;54:565–67. 150. Schanler RJ, Shulman RJ, Prestridge LL. Parenteral nutrient needs of very low birth weight infants. J Pediatr 1994;125:961–8. 151. Prestridge LL, Schanler RJ, Shulman RJ, Burns PA, Laine LL. Effect of parenteral calcium and phosphorus therapy on mineral retention and bone mineral content in very low birth weight infants. J Pediatr 1993;122:761–8. 152. Dickerson RN. Parenteral nutrition. http://dcp.utmem. edu/lectures/TPNsolutionslecture.pdf 153. Rollins CJ, Elsberry VA, Pollack KA, Pollack PF, Udall JN Jr. Three-in-one parenteral nutrition: a safe and economical method of nutritional support for infants. J Parenter Enteral Nutr 1990; 14:290–4. 154. Didier ME, Fischer S, Maki DG. Total nutrient admixtures appear safer than lipid emulsion alone as regards microbial contamination: growth properties of microbial pathogens at room temperature. J Parenter Enteral Nutr 1998;22:291–6. 155. D’Angio RG, Riechers KC, Gilsdorf RB, Constantino JM. Effect of the mode of lipid administration on parenteral nutrition-related infections. Ann Pharmacother 1992;26:14–7. 156. http://www.fda.gov/cder/fdama/difconc.htm 157. United States Pharmacopeia, Quality Assureance for Sterile Drug Compounding, An Educational Source, 1998. 158. McKinnon BT. FDA Safety Alert: Hazards of Precipitation Associated With Parenteral Nutrition. Nutr Clin Prac 1996;11: 59–65. 159. Hardy G, Ball P, McElroy B. Basic principles for compounding all-in-one parenteral nutrition admixtures. Curr Opin Clin Nutr Metab Care 1998;1:291–6. 160. Driscoll DF, Baptista RJ, Bistrian BR, Blackburn GL. Practical

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

606

161. 162.

163.

164.

165.

166.

167.

168.

169.

170.

171.

172.

173.

174.

175.

176.

177. 178.

179.

180.

R. J. SHULMAN AND S. PHILLIPS considerations regarding the use of total nutrient admixtures. Am J Hosp Pharm 1986;43:416–9. Brown R, Quercia RA, Sigman R. Total nutrien admixture: a review. 1986;10–650–58. National Advisory Group on Standards and Practice Guidelines for Parenteral Nutrition. Safe practices for parenteral nutrition formulations. J Parent Ent Nutr 1998;22:49–66. Driscoll DF, Bhargava HN, Li L, Zaim RH, Babayan VK, Bistrian BR. Physicochemical stability of total nutrient admixtures. Am J Health Syst Pharm 1995;52:623-34. Shay DK, Fann LM, Jarvis WR. Respiratory distress and sudden death associated with receipt of a peripheral parenteral nutrition admixture. Infect Control Hosp Epidemiol 1997;18:814–17. Department of Health and Human Services, Public Health Service; Food and Drug Administration, FDA Safety Alert: Hazards of Precipitation Associated with Parenteral Nutrition, April 18, 1994, http://www.fda.gov/cdrh/parnutr.pdf Parry VA, Harrie KR, McIntosh-Lowe NL. Effect of various nutrient ratios on the emulsion stability of total nutrient admixtures. Am J Hosp Pharm 1986;43:3017–22. Bethune K, Allwood M, Grainger C, Wormleighton C. Use of filters during the preparation and administration of parenteral nutrition: position paper and guidelines prepared by a British pharmaceutical nutrition group working party. Nutrition 2001;17: 403-8. Driscoll DF, Bacon MN, Bistrian BR. Effects of in-line filtration on lipid particle size distribution in total nutrient admixtures. J Parenter Enteral Nutr 1996;20:296–301. Rollins CJ, Elsberry VA, Pollack KA, Pollack PF, Udall JN Jr. Three-in-one parenteral nutrition: a safe and economical method of nutritional support for infants. J Parenter Enteral Nutr 1990; 14:290–4. Bullock L, Fitzgerald JF, Walter WV. Emulsion stability in total nutrient admixtures containing a pediatric amino acid formulation. J Parenter Enteral Nutr 1992;16:64–8. Smith JL, Canham JE, Kirkland WD, Wells PA. Effect of Intralipid, amino acids, container, temperature, and duration of storage on vitamin stability in total parenteral nutrition admixtures. J Parenter Enteral Nutr 1988;12:478–83. Trissel LA, Gilbert DL, Martinez JF, Baker MB, Walter WV, Mirtallo JM. Compatibility of medications with 3-in-1 parenteral nutrition admixtures. J Parenter Enteral Nutr 1999;23:67–74. American Medical Association Department of Foods and Nutrition. Multivitamin preparations for parenteral use. A statement by the Nutrition Advisory Group. J Parent Ent Nutr 1979;3: 258–62. Green HL, Moore ME, Phillips B. Evaluation of a pediatric multiple vitamin preparation for total parenteral nutrition. II. Blood levels of vitamins A,D, and E. Pediatrics 1986;77:539–47. Moore MC, Greene HL, Phillips B, Franck L, Shulman RJ, Murrell JE, Ament ME. Evaluation of a pediatric multiple vitamin preparation for total parenteral nutrition in infants and children. I. Blood levels of water-soluble vitamins. Pediatrics 1986;77: 530–8. Department of Health and Human Services Food and Drug Administration. Parenteral multivitamin products; Drugs for Human Use; Drug efficacy study implementation; Amendment. Federal Register. April 20, 2000;56:21200–21201. Alade SL, Brown RE, Paquet A Jr. Polysorbate 80 and E-Ferol toxicity. Pediatrics 1986;77:593–7. MacDonald MG, Getson PR, Glasgow AM, Miller MK, Boeckx RL, Johnson EL. Propylene glycol: increased incidence of seizures in low birth weight infants. Pediatrics 1987;79:622–5. Shulman RJ, Phillips SM, Maciejewski S. An update on a global shortage of intravenous multivitamins. J Pediatr Gastroenterol Nutr 1999;28:361. Greene HL, Hambidge KM, Schanler R, Tsang RC. Guidelines for the use of vitamins, trace elements, calcium, magnesium, and phosphorus in infants and children receiving total parenteral nutrition: report of the Subcommittee on Pediatric Parenteral Nutri-

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193. 194.

195.

196. 197.

198.

199.

ent Requirements from the Committee on Clinical Practice Issues of the American Society for Clinical Nutrition. Am J Clin Nutr 1988;48:1324–42. Drittij-Reijnders MJ, Sels JP, Rouflart M, Thijssen HH. Vitamin K status and parenteral nutrition; the effect of Intralipid on plasma vitamin K1 levels. Eur J Clin Nutr 1994;48:525–7. Lennon C, Davidson KW, Sadowski JA, Mason JB. The vitamin K content of intravenous lipid emulsions. J Parenter Enteral Nutr 1993;17:142–4. Darlow BA, Graham PJ. Vitamin A supplementation for preventing morbidity and mortality in very low birthweight infants. Cochrane Database Syst Rev 2000;(2):CD000501. Henton DH, Merritt RJ. Vitamin A sorption to polyvinyl and polyolefin intravenous tubing. J Parenter Enteral Nutr 1990;14: 79–81. Pediatric Parenteral Multivitamin Products; Drug Efficacy Study Implementation; Announcement of Marketing Conditions; Federal Register: January 26, 2000 (Volume 65, Number 17)] [Notices] [Page 4253–4256]. Porcelli PJ, Greene HL, Adcock EW. Retinol (vitamin A) and riboflavin (vitamin B2) administration and metabolism in very low birth weight infants. Semin Perinatol 1992;16:170–80. Smith JL, Canham JE, Kirkland WD, Wells PA. Effect of Intralipid, amino acids, container, temperature, and duration of storage on vitamin stability in total parenteral nutrition admixtures. J Parenter Enteral Nutr 1988;12:478–83. Smith JL, Canham JE, Wells PA. Effect of phototherapy light, sodium bisulfite, and pH on vitamin stability in total parenteral nutrition admixtures. J Parenter Enteral Nutr 1988;12:394–402. Greene HL, Phillips BL, Franck L, Fillmore CM, Said HM, Murrell JE, Moore ME, Briggs R. Persistently low blood retinol levels during and after parenteral feeding of very low birth weight infants: examination of losses into intravenous administration sets and a method of prevention by addition to a lipid emulsion. Pediatrics 1987;79:894–900. Dahl GB, Svensson L, Kinnander NJ, Zander M, Bergstrom UK. Stability of vitamins in soybean oil fat emulsion under conditions simulating intravenous feeding of neonates and children. J Parenter Enteral Nutr 1994;18:234–9. Silvers KM, Sluis KB, Darlow BA, McGill F, Stocker R, Winterbourn CC. Limiting light-induced lipid peroxidation and vitamin loss in infant parenteral nutrition by adding multivitamin preparations to Intralipid. Acta Paediatr 2001;90:242–9. Baeckert PA, Greene HL, Fritz I, Oelberg DG, Adcock EW. Vitamin concentrations in very low birth weight infants given vitamins intravenously in a lipid emulsion: measurement of vitamins A, D, and E and riboflavin. J Pediatr 1988;113:1057–65. Cairns PA, Stalker DJ. Carnitine supplementation of parenterally fed neonates. Cochrane Database Syst Rev 2000;(4):CD000950. Penn D, Schmidt-Sommerfeld E, Pascu F. Decreased tissue carnitine concentrations in newborn infants receiving total parenteral nutrition. J Pediatr 1981;98:976–8. Worthley LI, Fishlock RC, Snoswell AM. Carnitine deficiency with hyperbilirubinemia, generalized skeletal muscle weakness and reactive hypoglycemia in a patient on long-term total parenteral nutrition: treatment with intravenous L-carnitine. J Parenter Enteral Nutr 1983;7:176–80. Borum PR. Should carnitine be added to parenteral nutrition solutions? Nutr Clin Prac 2000;15:153–4. Reed MD, Bertino JS Jr, Halpin TC Jr. Use of intravenous iron dextran injection in children receiving total parenteral nutrition. Am J Dis Child 1981;135:829–31. Halpin TC Jr, Bertino JS, Rothstein FC, Kurczynski EM, Reed MD. Iron-deficiency anemia in childhood inflammatory bowel disease: treatment with intravenous iron-dextran. J Parenter Enteral Nutr 1982;6:9–11. Surico G, Muggeo P, Muggeo V, Lucarelli A, Martucci T, Daniele M, Rigillo N. Parenteral iron supplementation for the

PARENTERAL NUTRITION IN INFANTS AND CHILDREN

200.

201. 202. 203.

204.

205.

206. 207. 208. 209. 210.

211.

212.

213.

214.

treatment of iron deficiency anemia in children. Ann Hematol 2002;81:154–7. Mamula P, Piccoli DA, Peck SN, Markowitz JE, Baldassano RN. Total dose intravenous infusion of iron dextran for irondeficiency anemia in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2002;34:286–90. Burns DL, Mascioli EA, Bistrian BR. Parenteral iron dextran therapy: a review. Nutrition 1995;11:163–8. Kumpf VJ. Parenteral iron supplementation. Nutr Clin Pract 1996;11:139–46. Charytan C, Levin N, Al-Saloum M, Hafeez T, Gagnon S, Van Wyck DB. Efficacy and safety of iron sucrose for iron deficiency in patients with dialysis-associated anemia: North American clinical trial. Am J Kidney Dis 2001;37:300–7. Michael B, Coyne DW, Fishbane S, Folkert V, Lynn R, Nissenson AR, Agarwal R, Eschbach JW, Fadem SZ, Trout JR, Strobos J, Warnock DG. Sodium ferric gluconate complex in hemodialysis patients: adverse reactions compared to placebo and iron dextran. Kidney Int 2002;61:1830–9. Pollak A, Hayde M, Hayn M, Herkner K, Lombard KA, Lubec G, Weninger M, Widness JA. Effect of intravenous iron supplementation on erythropoiesis in erythropoietin-treated premature infants. Pediatrics 2001;107:78–85. Hardy G, Reilly C. Technical aspects of trace element supplementation. Curr Opin Clin Nutr Metab Care 1999;2:277–85. Van Gossum A, Neve J. Trace element deficiency and toxicity. Curr Opin Clin Nutr Metab Care 1998;1:499–507. Allwood MC. Aluminium in parenteral nutrition admixtures: an unnecessary risk? Nutrition 1999;15:958–9. Klein GL. Aluminum in parenteral solutions revisited–again. Am J Clin Nutr 1995;61:449–56. Pluhator-Murton MM, Fedorak RN, Audette RJ, Marriage BJ, Yatscoff RW, Gramlich LM. Trace element contamination of total parenteral nutrition. 1. Contribution of component solutions. J Parenter Enteral Nutr 1999;23:222–7. Klinger G, Shamir R, Singer P, Diamond EM, Josefsberg Z, Sirota L. Parenteral selenium supplementation in extremely low birth weight infants: inadequate dosage but no correlation with hypothyroidism. J Perinatol 1999;19:568–72. Hatanaka N, Nakaden H, Yamamoto Y, Matsuo S, Fujikawa T, Matsusue S. Selenium kinetics and changes in glutathione peroxidase activities in patients receiving long-term parenteral nutrition and effects of supplementation with selenite. Nutrition 2000;16:22–6. Yusuf SW, Rehman Q, Casscells W. Cardiomyopathy in association with selenium deficiency: a case report. J Parenter Enteral Nutr 2002;26:63–6. Mouser JF, Hak EB, Helms RA, Christensen ML, Storm MC. Chromium and zinc concentrations in pediatric patients receiving long-term parenteral nutrition. Am J Health Syst Pharm 1999;56: 1950–6.

607

215. Bougle D, Bureau F, Deschrevel G, Hecquard C, Neuville D, Drosdowsky M, Duhamel JF. Chromium and parenteral nutrition in children. J Pediatr Gastroenterol Nutr 1993;17:72–4. 216. Moukarzel AA, Song MK, Buchman AL, Vargas J, Guss W, McDiarmid S, Reyen L, Ament ME. Excessive chromium intake in children receiving total parenteral nutrition. Lancet 1992;339: 385–8. 217. Hak EB, Storm MC, Helms RA. Chromium and zinc contamination of parenteral nutrient solution components commonly used in infants and children. Am J Health Syst Pharm 1998;55:150–4. 218. Moukarzel AA, Ament ME, Buchman A, Dahlstrom KA, Vargas J. Renal function of children receiving long-term parenteral nutrition. J Pediatr 1991;119:864–8. 219. Shulman RJ. Zinc and copper balance studies in infants receiving total parenteral nutrition. Am J Clin Nutr 1989;49:879–83. 220. Dickerson RN. Manganese intoxication and parenteral nutrition. Nutrition 2001;17:689–693. 221. Quaghebeur G, Taylor WJ, Kingsley DP, Fell JM, Reynolds AP, Milla PJ. MRI in children receiving total parenteral nutrition. Neuroradiology 1996;38:680–3. 222. Fell JM, Reynolds, AP, Meadows N. Manganese toxicity in children receiving long-term parenteral nutrition. Lancet 1996;347: 1218–21. 223. Bertinet DB, Tinivella M, Balzola FA, de Francesco A, Davini O, Rizzo L, Massarenti P, Leonardi MA, Balzola F. Brain manganese deposition and blood levels in patients undergoing home parenteral nutrition. J Parenter Enteral Nutr 2000;24:223–7. 224. Masumoto K, Suita S, Taguchi T, Yamanouchi T, Nagano M, Ogita K, Nakamura M, Mihara F. Manganese intoxication during intermittent parenteral nutrition: report of two cases. J Parenter Enteral Nutr 2001;25:95–9. 225. Fok TF, Chui KK, Cheung R, Ng PC, Cheung KL, Hjelm M. Manganese intake and cholestatic jaundice in neonates receiving parenteral nutrition: a randomized controlled study. Acta Paediatr 2001;90:1009–15. 226. Fitzgerald K, Mikalunas V, Rubin H, McCarthey R, Vanagunas A, Craig RM. Hypermanganesemia in patients receiving total parenteral nutrition. J Parenter Enteral Nutr 1999;23:333–6. 227. Takagi Y, Okada A, Sando K, Wasa M, Yoshida H, Hirabuki N. On-off study of manganese administration to adult patients undergoing home parenteral nutrition: new indices of in vivo manganese level. J Parenter Enteral Nutr 2001;25:87–92. 228. C. Garza, personal communication, 1980. 229. Pupim LB, Flakoll PJ, Brovillette JR. Intradialytic parenteral nutrition improves protein and energy homeostasis in chronic hemodialysis patients. J Clin Invest 2002;110:483–92. 230. Haas C, Genzel-Boroviczeny O, Koletzko B. Losses of Vitamin A and E in parenteral nutrition suitable for premature infants. Eur J Clin Nutr 2002;56:96–12.

J Pediatr Gastroenterol Nutr, Vol. 36, No. 5, May 2003