• Open access
  • Published: 08 August 2014

Development and current use of parenteral nutrition in critical care – an opinion paper

  • Mette M Berger 1 &
  • Claude Pichard 2  

Critical Care volume  18 , Article number:  478 ( 2014 ) Cite this article

12k Accesses

22 Citations

16 Altmetric

Metrics details

Critically ill patients depend on artificial nutrition for the maintenance of their metabolic functions and lean body mass, as well as for limiting underfeeding-related complications. Current guidelines recommend enteral nutrition (EN), possibly within the first 48 hours, as the best way to provide the nutrients and prevent infections. EN may be difficult to realize or may be contraindicated in some patients, such as those presenting anatomic intestinal continuity problems or splanchnic ischemia. A series of contradictory trials regarding the best route and timing for feeding have left the medical community with great uncertainty regarding the place of parenteral nutrition (PN) in critically ill patients. Many of the deleterious effects attributed to PN result from inadequate indications, or from overfeeding. The latter is due firstly to the easier delivery of nutrients by PN compared with EN increasing the risk of overfeeding, and secondly to the use of approximate energy targets, generally based on predictive equations: these equations are static and inaccurate in about 70% of patients. Such high uncertainty about requirements compromises attempts at conducting nutrition trials without indirect calorimetry support because the results cannot be trusted; indeed, both underfeeding and overfeeding are equally deleterious. An individualized therapy is required. A pragmatic approach to feeding is proposed: at first to attempt EN whenever and as early as possible, then to use indirect calorimetry if available, and to monitor delivery and response to feeding, and finally to consider the option of combining EN with PN in case of insufficient EN from day 4 onwards.

Introduction

Critical illness requiring vital organ support is generally associated with an intense inflammatory response and requires bed rest, both factors favoring lean body mass catabolism. These alterations promote the risk of malnutrition, or aggravate a pre-existing malnutrition, and cause a related increased morbidity and mortality [ 1 ]. Critically ill patients depend on artificial nutrition for the maintenance of their metabolic functions and limitation of the underfeeding related to complications. Delivering adequate amounts of nutrients and energy should therefore be a basal preoccupation of the intensivist, like hydration and pain control. Giving your patient a FAST HUG (feeding, analgesia, sedation, thromboembolic prophylaxis, head-of-bed elevation, stress ulcer prophylaxis and glycemic control) is exactly this provision of basal care [ 2 ].

Current guidelines recommend early enteral nutrition (EN) as the best way to provide the nutrients [ 3 ],[ 4 ]. Nevertheless, EN is a technique associated with practical problems worldwide, resulting in frequent insufficient feed delivery [ 5 ]. Further, EN may be contraindicated in some patients, such as those presenting anatomic intestinal discontinuity or splanchnic ischemia; hard contraindications are not very frequent in our experience, varying between 5 and 7% in the Geneva and Lausanne ICUs. Intravenous administration of macronutrients and micronutrients, called parenteral nutrition (PN), becomes recommended under certain circumstances (Table  1 ) [ 6 ] and is technically easier to deliver than EN. PN has therefore been overused in many ICUs, and has been associated with both metabolic and infectious complications [ 7 ]. In 2013 the medical community is left with great uncertainty regarding the place of PN in critically ill patients.

This review aims at summarizing some critical metabolic changes in critically ill patients, and at explaining the historical background of PN development that impacts on its actual strengths and weaknesses.

Impact of disease on lean body mass

Critical illness elicits a cascade of inflammatory, immune, endocrine and metabolic responses [ 8 ]. Critically ill patients are generally considered to be hypermetabolic, with exacerbated lipolysis, proteolysis and extracellular water gain associated with fluid resuscitation [ 9 ]. In response to inflammatory mediators and oxidative stress, and particularly in patients remaining acutely ill beyond the first 72 hours, proteolysis increases massively in excess of protein synthesis, a condition called catabolism that causes a rapid loss of lean body mass, mostly controlled by the ubiquitin–proteasome pathway [ 10 ].

In the 1990s, using stable isotope techniques, it was shown that energy and protein requirements were higher in critically ill patients than in healthy subjects. Isocaloric PN was also shown to be well tolerated without generating undue hyperglycemia, but exceeding 1.3 g protein/kg/day intakes did not further increase protein accretion [ 11 ] (Figure  1 ). In critically ill trauma patients, PN providing 120% of the measured energy expenditure (EE) did not have any positive effect on protein metabolism, but only generated deleterious hypermetabolism [ 12 ]. Further, bed rest studies conducted by the European Space Agency demonstrated an intense protein catabolism related to physical immobilization [ 13 ], the latter being increased by hypocaloric feeding.

figure 1

Effect of nutrition support on total body protein in patients with an acute flare of ulcerative colitis. Conversely to patients with standard care (broken line; mean age 47.6 ± 12.1 years, mean weight 59.4 ± 12.5 kg), ulcerative colitis patients with total parenteral nutrition (TPN; solid black line; mean age 42.6 ± 10.2 years, mean weight 60.8 ± 10.6 kg) received TPN from hospital admission until day 14 after surgery. Patients without TPN showed a rapid body protein loss that fell under the critical threshold before hospital admission, and worsened during the perioperative period. On the contrary, TPN prevented the worsening of protein body loss during the perioperative period and was associated with an earlier restoration (18 weeks earlier) of normal protein stores (mean ± standard deviation). Reproduced with permission from [ 14 ].

Critically ill patients combine both intense stress and physical immobilization [ 13 ] that may cause a rapid decrease of lean tissues, which in turn has an impact on respiratory and peripheral muscle function. After an acute disease, recovery of the muscle alterations is associated with an improved global functioning and quality of life [ 15 ]. The preservation of lean body mass appears particularly important for the final outcome. Optimal nutrition support during critical illness has been postulated to limit protein catabolism and promote faster recovery, although this concept has been challenged recently in a substudy of patients from the large Early Parenteral Nutrition Completing Enteral Nutrition in Adult Critically Ill Patients (EPaNIC) trial: PN administered from admission to the ICU as glucose followed by PN did not prevent lean body mass loss [ 16 ]. Contrasting with these results, an Australian study enrolling 1,372 patients with a short-term contraindication for EN showed clinical benefits in patients randomized to early PN with shorter mechanical ventilation and a better quality of life at 2-month follow-up; the parenteral intervention of course resulted in a better protein coverage compared with standard care [ 17 ]. More data on long-term outcome are needed.

Historical development

Considering the development of PN is helpful in understanding the actual controversies. Figure  2 summarizes the major steps in the history of PN. Although attempts at feeding intravenously can be found as early as the 16th century, complete PN in its modern form was invented by Arvid Wretlind and colleagues in 1961 in Sweden [ 18 ] and in the USA [ 19 ]. From the start, this feeding technique saved many lives compromised by gastrointestinal failure, previously doomed to rapid death. Since its enthusiastic start in the 1960s, PN has evolved tremendously and has generated numerous contradictory publications.

figure 2

Milestones in the development of artificial nutrition. Development steps in energy requirements and lipid emulsions (left) and amino acid and glucose steps (right). Adapted with permission from [ 20 ].

Today, the production of metabolically balanced amino acid (AA) combinations remains a challenge. The development of the crystalline AAs [ 21 ] led to a better tolerance, but more prolonged PN treatments unmasked trace element deficiencies, requiring the development of balanced micronutrient solutions to compensate deficits resulting from the purification of the AA solutions.

The lipid emulsions remained the most difficult issue during decades. Severe complications of the cottonseed-derived lipid emulsions were observed in the USA, where their withdrawal led to incomplete PN (that is, without lipids) for decades. In Europe, however, thanks to the existence of the soybean solution using egg yolk phospholipids as an emulsifying agent, PN was complete (total PN) from the start. PN was widely used, often in patients without gastrointestinal failure, and despite the central venous access-associated complications. It is indeed much easier to deliver the prescribed amount of energy by the intravenous route than by the enteral route [ 22 ].

In the 1970s and 1980s the observation of devastating losses of lean body mass was the rationale for the delivery of hyperalimentation, as feeding was first called, using large glucose loads, erroneously supposed to suppress endogenous glucose production and thereby to prevent AA loss. The glucose–protein sparing strategy and the unavailability of lipid emulsions in the USA led to the prescription of high doses of dextrose exceeding the oxidizing capacity. Delivering >3,000 kcal/day was very common in the 1980s; hyperglycemia in the range 10 to 15 mmol/l was considered adaptive, and was generally not treated. The high glucose loads generated complications (excessive carbon dioxide production, respiratory failure, fever, additional metabolic stress, liver steatosis) [ 23 ]. It became obvious that parenteral hyperalimentation caused increased infectious complications compared with EN, which was frequently hypocaloric [ 24 ]. Only after the 2001 Leuven trial did the medical community become aware of the importance of controlling blood glucose levels by means of continuous insulin therapy [ 25 ], although the NICE SUGAR and Glucontrol trials subsequently demonstrated that intensive insulin therapy was potentially dangerous [ 26 ],[ 27 ] and that less strict glucose targets compared with the initial normoglycemia (4.1 to 6 mmol/l) used by the Leuven team were safer.

Fong and colleagues showed that 5 days of PN in healthy subjects compared with EN exacerbated the inflammatory response to endotoxin [ 28 ]. In 1991 the Veterans study showed an absence of benefits and even deleterious effects of perioperative PN in patients without malnutrition [ 24 ]. These trials nearly destroyed the concept of PN, given that clinicians became very suspicious about potential deleterious consequences.

In the 1990s, technical advances in plastics enabled the development of double-compartmental and tri-compartmental bags in Europe, with separation of the macrosubstrates during storage, being easy to handle, and providing the necessary guaranties regarding stability and sterility of the solutions, reducing contamination, errors in prescription, and costs [ 29 ].

Energy requirements

Determination of the patients’ energy and substrate requirements has proven much more difficult than expected. Evolving from the early phase of systematic hyperalimentation, the promotion of EN as the only correct way to feed patients has generated a second wave of ICU hypoalimentation from the 1990s that still persists [ 5 ],[ 30 ],[ 31 ]. A large proportion of severely ill patients leave the ICU with a cumulated deficit of up to 10,000 to 20,000 kcal [ 5 ],[ 30 ],[ 31 ], which roughly corresponds to about 5 to 10 kg lean tissues and 0.5 to 1 kg fat reserve. The guidelines have tried to promote a reasonable approach recommending 20 to 25 kcal/kg body weight in the early acute phase, to be increased to 25 to 30 kcal/kg in stabilized patients [ 6 ]. The aim was to initiate feeding early (that is, during the first 48 hours), preferentially by EN, to prevent the addition of an energy deficit to a previously malnourished patient or to avoid worsening the catabolic response of stress patients.

The problem with all energy predictive equations is that they fail in nearly 70% of patients [ 32 ],[ 33 ], being totally unreliable in the obese but also at the other end of the spectrum in patients with low body mass index <18. This unreliability occurs because EE in critically ill patients is highly variable depending on the initial injury, severity of the disease, nutritional status, time after admission and treatments, and an unpredictable variation of the body weight to EE ratio [ 34 ]. Many proposed equations are based on static variables (sex, age, weight, height) while others include dynamic variables (fever, minute ventilation, heart rate) [ 35 ]. Indirect calorimetry is considered the gold standard for determining EE, enabling feeding to be adapted to the measured EE. Two equations have been derived from such measurements and perform better than others: the Faisy–Fagon equation for mechanically ventilated patients [ 36 ], and the Toronto equation for major burns [ 37 ]. Figure  3 shows two static equations that failed to provide adequate orientation on the real energy target in the Swiss supplemental parenteral nutrition (SPN) trial compared with indirect calorimetry [ 38 ].

figure 3

Relationship between two commonly used equations and the value of energy expenditure. Indirect calorimetry study on day 3 shows that both equations overestimated and underestimated energy expenditure in an unpredictable manner. (A) Pre-enrollment target 25 to 30 kcal/kg actual body weight (BW): arrows show the relation between the calculated energy target used for enrollment (25 or 30 kcal/kg/day) and the measured energy expenditure that became the target used from day 4. (B) Target of the supplemental parenteral nutrition patients recalculated using an equation based on a corrected ideal body weight, age and gender [ 39 ], which was used in the Early Parenteral Nutrition Completing Enteral Nutrition in Adult Critically Ill Patients (EPaNIC) trial [ 40 ] (corrected ideal body weight, age and gender [ 34 ] + absolute maximal target of 2,880 kcal).

The tight calorie control study (TICACOS) is among the rare ICU studies where EE has been systematically and repeatedly measured, being determined every second day [ 41 ]. Unfortunately, the prescription of energy did not integrate non-nutritional calories (that is, associated with glucose for drug administration or lipid from propofol) despite supervision by the research team, and resulted in systematic overfeeding in the intervention group, which is likely to explain the observed increasing incidence of pneumonia and prolonged ventilation. Despite these immediate ICU complications, the TICACOS also showed that a better energy coverage in the calorimetry group with a combined nutrition support resulted in a reduced length of hospital stay and hospital mortality, but the numbers were too low to allow for definitive conclusions, needing repetition and extension of the study. Recently, the SPN trial showed that this individualized energy supply obtained by EE measurement in 65% of patients was rewarding in terms of reduction of nosocomial infections in severely ill ICU patients requiring prolonged ICU treatment [ 38 ]. Grau and colleagues have shown that liver alterations (defined as cytolysis, cholestasis or a combination of both) during enteral or parenteral feeding occur frequently if energy delivery exceeds 27 kcal/kg/day, and add to those liver alterations caused by sepsis and multiple organ failure [ 42 ]; the difference becomes significant after day 11 in patients on PN, ED producing less alteration related to lower energy intakes.

Hypocaloric feeding: a real option?

Overfeeding is a threat, so some authors have proposed prescribing hypocaloric feeding; that is, 70 to 80% of the calculated target [ 43 ]. This strategy has been mainly developed for obese patients, in whom all of the equations invariably fail; the Penn State equation adapted for obese patients being among the closest to measured EE [ 44 ]. By contrast in hemodynamic management, an underprescription of norepinephrine would not be a treatment option in case of an arterial hypotension. Similarly, we need to prescribe what is considered adequate and optimal requirement for the patient, especially because the use of industrial formula implies that delivering low amounts of calories further lowers the protein delivery – a minimum energy is required to be able to benefit from proteins [ 9 ]. Moreover the international surveys show that underfeeding remains a serious threat [ 5 ], while patients with body mass index <18 experience a maximal variability of energy delivery.

Recent research has focused on autophagy, which belongs to the healing process as removal of mitochondria damaged during the acute phase of sepsis. This mechanism is a two-edged sword, however, which may cause cell death [ 45 ]. Insulin is a well-documented inhibitor of autophagy [ 46 ]. Overfeeding leads to increased insulin requirements to achieve blood glucose control. In the EPaNIC trial [ 16 ], the patients in the early PN group needed nearly double the amount of insulin for this purpose, probably reflecting overfeeding that was associated with depressed autophagy. Figure  3 B shows that the “Leuven” equation results in frequent excessive targets when EE is verified by indirect calorimetry. By contrast, feeding in the SPN trial was guided by calorimetry, and no such increase was observed with identical insulin requirements in both arms [ 38 ].

From substrate provision to pharmaconutrition

Amino acids.

The first crystalline AAs were unbalanced, containing about 50% glycine of low biological value. Introducing tyrosine, cysteine–cystine and glutamine was technically difficult due to stability and solubility problems. Glutamine was finally solubilized under the dipeptide form glutamine–alanine by Fürst and Stehle [ 47 ]. The concept of conditionally essential AAs emerged in this period; these are AAs that are supplied by food and synthesized under normal conditions but which during critical illness become deficient because of insufficient supply and increased consumption. Isolation of various AAs led to the possibility of using them separately, potentially as drugs.

Glutamine, the most abundant free AA in the body, which constitutes over 60% of the muscle free AA pool [ 47 ], is one of these conditionally essential AAs; depletion has repeatedly been shown to occur in critically ill patients and is associated with poor prognosis [ 48 ]. Well-conducted repletion studies have shown that glutamine administration is beneficial if administered along with an optimized nutrition therapy, particularly by the parenteral route, improving glucose control and achieving reduction of both infectious complications and mortality [ 49 ]-[ 52 ]. But glutamine cannot be provided in standard PN for stability reasons, which increases the risk of deficiency.

Two recent large prospective randomized clinical trials brought confusion, the doses of glutamine and their timing largely explaining the negative results. The Scottish SIGNET trial enrolled 502 patients with gastrointestinal failure [ 53 ]. The patients were randomized to receive daily 20.2 g glutamine or 500 μg selenium, or both, versus placebo for up to 7 days. There was no overall effect of glutamine on new infections or on mortality, while selenium was associated with less infectious complications if delivered for longer than 5 days. Several shortcomings of the study, including a very short administration time and a one-size-fits-all prescription of the ready-to-use PN bags, resulted in the delivery of a very low glutamine dose (0.1 g/kg/day) for a very short period, far below international recommendations [ 6 ]. On the non-effect side, a small American prospective randomized clinical trial in 44 patients randomized to three groups receiving either isonitrogenous enteral nutrition or 0.5 g/kg/day glutamine by the intravenous or enteral route for 8 days showed no difference in antioxidant status or other markers of oxidative stress [ 54 ]; importantly, about one-third of the patients had normal baseline glutamine levels.

The REDOXS trial reports data for 1,223 patients receiving the highest doses so far used of glutamine (0.78 g/kg/day supplied as 0.35 g/kg intravenously + 30 g/day enterally), about twice the recommended doses, in patients with severe organ failure (93% of patients in shock state and 33% with renal failure) starting within the first 24 hours of admission independently of nutrition [ 55 ]; that is, earlier than any previous trial. In addition, the glumatine group suffered more organ failure which might explain on its own the higher mortality. Surprisingly, in a subset of patients with plasma determination, only 31% of patients presented with a low baseline glutamine level (<420 μmol/l) whereas 15% of these patient had supranormal plasma glutamine values at baseline. The latter finding has been shown to be associated with increased mortality [ 56 ]. Pharmacological doses of glutamine in unstable patients are therefore to be avoided.

Many of the early problems as well as the progress of PN were associated with the development of lipid emulsions [ 23 ]. Prolonged use of the glucose and AA-based formulations with no fat was associated with essential fatty acid deficiency [ 23 ]. As fatty acids are more energy dense (about 9 kcal/g) than both AAs (4 kcal/g) and glucose (3.7 kcal/g), they enable reducing both the fluid load (important in the ICU patient) and the osmolarity of the solutions, permitting peripheral administration in some patients.

Since the development of the long-chain triglyceride solution by Wretlind and colleagues, major developments have occurred (Figure  2 ). Pulmonary and hematological side effects were observed, as well as an enhanced oxidative stress causing peroxidation of the unsaturated fatty acids; the latter may cause cell death and worsening of organ failures [ 21 ]. One of the first options aiming at minimizing oxidative stress was to partially replace polyunsaturated fatty acids (PUFAs) with oils rich in medium-chain triglycerides derived from coconut oil, which are less prone to peroxidation. Another advantage of medium-chain triglycerides is that they require less carnitine for mitochondrial penetration than long-chain triglyceride solution and are metabolized more rapidly, but they increase in a reversible manner the production of ketone bodies [ 57 ].

Progressively, it became obvious that the various PUFAs have proper modulating effects on the inflammatory response [ 58 ]. Medium-chain triglycerides and monounsaturated fatty acids derived from olive oil are considered the most neutral, while the more recently developed n-3 PUFAs derived from fish oil exhibit anti-inflammatory properties [ 58 ]. The intravenous administration of n-3 PUFAs seems associated with clinical benefits [ 59 ] and quick physiological effects [ 60 ]; development is ongoing and has been reviewed recently [ 61 ].

Is parenteral pharmaconutrition worth it?

These variable results have lead to questioning the cost-efficiency resulting from glutamine and n-3 PUFA administration. The most convincing evidence comes from recent large-scale medico-economic studies. A large recent Italian study in 60,000 patients from 200 Italian ICUs strongly supports the use of glutamine containing PN [ 62 ]; the costs of treatment were completely offset by savings made by shortening the ICU stay and lower antibiotic costs. Similarly regarding n-3 PUFAs, an analysis including 23 trials in 1,502 patients showed that n-3 PUFA inclusion in PN resulted in a reduction of infection complications (relative risk = 0.61) and of lengths of stay [ 63 ], both in the ICU (nearly 2 days) and in the hospital overall (3.3 days), reducing the costs. Finally, an economic analysis of the Australian early PN study showed that the strategy of cautious early PN reduces hospital costs [ 64 ]. Well-conducted nutrition therapy, despite including a modest investment, is cost-saving in the end.

What is the optimal timing for parenteral nutrition introduction?

While the indications for total PN (delivery of glucose, proteins, fat, and micronutrients) are unchanged, much controversy has arisen regarding its timing. Guidelines have been understood in divergent ways: the European Society of Clinical Nutrition PN guidelines have been interpreted as ‘deliver PN after 2 days’ [ 6 ], while the American Society of Parenteral and Enteral Nutrition guidelines have been understood as ‘no feed before day 7’ [ 65 ].

The large, prospective, EPaNIC trial randomized 4,640 patients to early PN (2 days with glucose from day 0) followed by full PN versus late PN (day 8) after ICU admission, and concluded that early PN was harmful, with later ICU discharge and more complications (including infections), despite the application of a tight glycemic protocol [ 40 ]. The EPaNIC trial used PN in unselected patients [ 38 ]. This last characteristic is precisely one of the major pitfalls of the study; early PN in low-risk patients may be harmful [ 66 ], confirming the Veterans trial results [ 24 ]. McClave and colleagues pinpointed various limitations of the study, such as the hypercaloric and low nitrogen intakes and the over-representation of cardiovascular surgery patients, which reduce its external validity [ 66 ].

The Swiss SPN prospective randomized controlled trial enrolled 305 patients on day 3 of admission to the ICU: enrollment criteria were that patients received ≤60% of the calculated energy target from EN, were expected to stay for >5 days, and were expected to survive for >7 days [ 38 ]. All patients were given a chance to be fed enterally, only those failing to achieve 60% of target were randomized to receive, or not, supplemental PN on top of EN in order to cover their measured energy needs after day 3. Energy targets were set by indirect calorimetry after day 3 (Figure  3 A) or, if not technically possible, continued as 25 and 30 kcal/kg actual body weight/day for women and men, respectively. The difference between the indirect calorimetry value and the equation target varied between −1,000 and +1,000 kcal/day. Figure  3 shows the relation between these measurements and two commonly used equations: the simple 25 to 30 kcal/kg body weight equation [ 6 ], and a more sophisticated equation based on the corrected ideal body weight, age and gender [ 39 ] used in the EPaNIC trial. Patients were randomly assigned to continue exclusive EN or to SPN (EN + PN). The primary outcome was the occurrence of nosocomial infection from the end of intervention (day 8) until the end of the follow-up (day 28). The intervention group had less nosocomial infections ( P  = 0.03), and a lower mean number of nosocomial infections and of antibiotic days per patient, resulting in more antibiotic-free days. The study concluded that individually optimized energy supplementation with SPN starting 4 days after ICU admission could reduce nosocomial infections. The study has some shortcomings that are summarized in a series of letters [ 67 ]a-d], particularly the fact that infections during the intervention were not considered. But indeed the primary endpoint was infections after the intervention (5 days of full feeding). The difference became only significant after a few days, a time delay that appears to be normal for a metabolic intervention that requires 3 to 5 days to become measurable, distinguishing nutritional therapy from pharmacological interventions. Under conditions of insufficient EN, SPN might improve clinical outcome, if it adequately covers basal energy requirements, enabling a normal metabolic response.

Doig and colleagues enrolled 1,372 Australian patients with a temporary contraindication to EN in an early PN trial [ 68 ]. The patients were randomized within 24 hours of ICU admission to receive either standard care or early PN. Their conclusion is that ‘the provision of early PN to critically ill adults with relative contraindications to early EN, compared with standard care, did not result in a difference in day-60 mortality. The early PN strategy resulted in significantly fewer days of invasive ventilation but not significantly shorter ICU or hospital stays’ [ 68 ]. The deleterious effects observed in the EPaNIC trial were not observed in the early PN group, which supports the findings in the SPN trial [ 38 ]; indeed, the energy targets used in the Australian trial were modest, based on the Harris and Benedict equation, thereby reducing the risk of overfeeding.

Finally, 14% of patients are plagued by diarrhea as shown by a recent trial including 1,595 ICU patient-days. EN may contribute to development of diarrhea because the delivery of more than 60% of the energy expenditure almost doubled the risk of diarrhea. This suggests that in some patients the combination of EN and PN may be helpful, reducing the burden and the cost of managing diarrhea (manpower, investigations, treatment) [ 69 ].

How to monitor artificial feeding and parenteral nutrition

The clinical follow-up should integrate a close monitoring of the metabolic and gastrointestinal tolerance with a bundle combining clinical observations, a daily glycemic profile and insulin requirements to watch for potential overload. Monitoring should also include a weekly laboratory workout and observations of the changes over time of these variables (Table  2 ).

Towards a pragmatic approach

These trials are showing us the way to a reasonable nutritional therapy. Our aim is still to counteract a massive catabolism, but all patients are not equally exposed to this risk. The critically ill patient is an individual to whom mean values apply poorly. Energy equations are simple to calculate, but constitute an inadequate tool; measuring the individual requirement is essential to avoid both underfeeding and overfeeding. Starting EN within the first 48 hours at a slow progressive rate (20 ml/hour even in the sickest) is beneficial, including for non-nutritional reasons such as keeping bowel motility and IgA secretion [ 4 ]. If the gut is not functional with a transient or permanent contraindication to EN, a precise PN with low targets covering only the basal resting EE requirements, measured or based on the crude Harris and Benedict equation, may be initiated after 2 to 3 days in those patients presenting malnutrition on admission [ 68 ]. On the other hand, early overfeeding is deleterious, and is a real risk with PN [ 40 ]. After day 3, if the EN does not cover the measured expenditure, then a combined intravenous and enteral approach may prevent larger energy deficits [ 38 ]; the tolerance level to energy deficit seems to be somewhere around a cumulated balance of −50 to −80 kcal/kg body weight from admission before development of complications related to underfeeding. The concept implies a monitoring of feeding delivery to closely guide nutrition therapy.

An unequivocal approach – that is, one opposing enteral and parenteral feeding – is inadequate in most ICU patients [ 70 ]. However, a pragmatic and reasonable attitude seems the better deal for the individual patient; while PN is simpler to deliver than EN, its metabolic consequences are more complicated to handle. The authors’ experience is that PN requires tight monitoring and dedicated resources such as dieticians in the ICU [ 71 ]. PN saves lives, but easily causes overfeeding, with its deleterious side effects. We have to work at identifying and measuring needs more precisely, but also observing the individual tolerance to enteral or parenteral feeding, in order to optimize the global care of patients.

Abbreviations

Energy expenditure

Enteral nutrition

Early Parenteral Nutrition Completing Enteral Nutrition in Adult Critically Ill Patients

Parenteral nutrition

Polyunsaturated fatty acid

Supplemental parenteral nutrition

Tight Calorie Control Study

Stapleton RD, Jones N, Heyland DK: Feeding critically ill patients: what is the optimal amount of energy?. Crit Care Med. 2007, 35 (9 Suppl): S535-S540. 10.1097/01.CCM.0000279204.24648.44.

Article   PubMed   Google Scholar  

Vincent JL: Give your patient a fast hug (at least) once a day. Crit Care Med. 2005, 33: 1225-1229. 10.1097/01.CCM.0000165962.16682.46.

Kreymann KG, Berger MM, Deutz NE, Hiesmayr M, Jolliet P, Kazandjiev G, Nitenberg G, van den Berghe G, Wernerman J: DGEM (German Society for Nutritional Medicine), Ebner C, Hartl W, Heymann C, Spies C, ESPEN (European Society for Parenteral and Enteral Nutrition): ESPEN guidelines on enteral nutrition: intensive care. Clin Nutr. 2006, 25: 210-223. 10.1016/j.clnu.2006.01.021.

Article   CAS   PubMed   Google Scholar  

Artinian V, Krayem H, DiGiovine B: Effects of early enteral feeding on the outcome of critically ill mechanically ventilated medical patients. Chest. 2006, 129: 960-967. 10.1378/chest.129.4.960.

Alberda C, Gramlich L, Jones N, Jeejeebhoy K, Day AG, Dhaliwal R, Heyland DK: The relationship between nutritional intake and clinical outcomes in critically ill patients: results of an international multicenter observational study. Intensive Care Med. 2009, 35: 1728-1737. 10.1007/s00134-009-1567-4.

Singer P, Berger MM, Van den Berghe G, Biolo G, Calder P, Forbes A, Griffiths R, Kreyman G, Leverve X, Pichard C: ESPEN guidelines on parenteral nutrition: intensive care. Clin Nutr. 2009, 28: 387-400. 10.1016/j.clnu.2009.04.024.

Anderson CF, MacBurney MM: Application of A.S.P.E.N. clinical guidelines: parenteral nutrition use at a university hospital and development of a practice guideline algorithm. Nutr Clin Pract. 1996, 11: 53-58. 10.1177/011542659601100253.

Wolfe RR: Regulation of skeletal muscle protein metabolism in catabolic states. Curr Opin Clin Nutr Metab Care. 2005, 8: 61-65. 10.1097/00075197-200501000-00009.

Plank LD, Hill GL: Energy balance in critical illness. Proc Nutr Soc. 2003, 62: 545-552. 10.1079/PNS2003259.

Burckart K, Beca S, Urban RJ, Sheffield-Moore M: Pathogenesis of muscle wasting in cancer cachexia: targeted anabolic and anticatabolic therapies. Curr Opin Clin Nutr Metab Care. 2010, 13: 410-416. 10.1097/MCO.0b013e328339fdd2.

Article   PubMed Central   CAS   PubMed   Google Scholar  

Hill GL: Implications of critical illness, injury, and sepsis on lean body mass and nutritional needs. Nutrition. 1998, 14: 557-558. 10.1016/S0899-9007(98)00045-8.

Iapichino G, Gattinoni L, Solca M, Radrizzani D, Zucchetti M, Langer M, Vesconi S: Protein sparing and protein replacement in acutely injured patients during TPN with and without amino acid supply. Intensive Care Med. 1982, 8: 25-31. 10.1007/BF01686850.

Biolo G, Ciocchi B, Stulle M, Bosutti A, Barazzoni R, Zanetti M, Antonione R, Lebenstedt M, Platen P, Heer M, Guarnieri G: Calorie restriction accelerates the catabolism of lean body mass during 2 wk of bed rest. Am J Clin Nutr. 2007, 86: 366-372.

CAS   PubMed   Google Scholar  

Hill GL: Jonathan E. Rhoads Lecture. Body composition research: implications for the practice of clinical nutrition. J Parenter Enteral Nutr. 1992, 16: 197-218. 10.1177/0148607192016003197.

Article   CAS   Google Scholar  

Allison SP: The uses and limitations of nutritional support. Clin Nutr. 1992, 11: 319-330. 10.1016/0261-5614(92)90082-2.

Casaer MP, Langouche L, Coudyzer W, Vanbeckevoort D, De Dobbelaer B, Guiza FG, Wouters PJ, Mesotten D, Van den Berghe G: Impact of early parenteral nutrition on muscle and adipose tissue compartments during critical illness. Crit Care Med. 2013, 41: 2298-2309. 10.1097/CCM.0b013e31828cef02.

Doig GS: Parenteral versus enteral nutrition in the critically ill patient: additional sensitivity analysis supports benefit of early parenteral compared to delayed enteral nutrition. Intensive Care Med. 2013, 39: 981-982. 10.1007/s00134-013-2856-5.

Vinnars E: Arvid Wretlind. Clin Nutr. 2003, 22: 1-5. 10.1054/clnu.2002.0628.

Dudrick SJ, Wilmore DW, Vars HM, Rhoads JE: Long-term total parenteral nutrition with growth, development, and positive nitrogen balance. Surgery. 1968, 64: 134-142.

Berger MM: The 2013 Arvid Wretlind lecture: Evolving concepts in parenteral nutrition. Clin Nutr. 2014, ᅟ: ᅟ-[Epub ahead of print]

Google Scholar  

Vinnars E, Hammarqvist F: 25th Arvid Wretlind's Lecture – Silver Anniversary, 25 years with ESPEN, the history of nutrition. Clin Nutr. 2004, 23: 955-962. 10.1016/j.clnu.2004.06.001.

Berger MM, Chioléro RL, Pannatier A, Cayeux C, Tappy L: A 10-year survey of nutritional support in a surgical ICU: 1986–1995. Nutrition. 1997, 13: 870-877. 10.1016/S0899-9007(97)00270-0.

Calder PC, Jensen GL, Koletzko BV, Singer P, Wanten GJ: Lipid emulsions in parenteral nutrition of intensive care patients: current thinking and future directions. Intensive Care Med. 2010, 36: 735-749. 10.1007/s00134-009-1744-5.

Perioperative total parenteral nutrition in surgical patients. N Engl J Med. 1991, 325: 525-532. 10.1056/NEJM199108223250801.

Van den Berghe G, Wouters P, Weekers F, Vervaest C, Bruynickx F, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R: Intensive insulin therapy in critically ill patients. N Engl J Med. 2001, 345: 1359-1367. 10.1056/NEJMoa011300.

Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Henderson WR, Hébert PC, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ: Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009, 360: 1283-1297. 10.1056/NEJMoa0810625.

Preiser JC, Devos P, Ruiz-Santana S, Melot C, Annane D, Groeneveld J, Iapichino G, Leverve X, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chioléro R: A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Med. 2009, 35: 1738-1748. 10.1007/s00134-009-1585-2.

Fong YM, Marano MA, Braber A, He W, Moldawer LL, Bushman ED, Coyle SM, Shires GT, Lowry SF: Total parenteral nutrition and bowel rest modify the metabolic response to endotoxin in humans. Ann Surg. 1989, 210: 449-457. 10.1097/00000658-198910000-00005.

Pichard C, Schwarz G, Frei A, Kyle U, Jolliet P, Morel P, Romand JA, Sierro C: Economic investigation of the use of three-compartment total parenteral nutrition bag: prospective randomized unblinded controlled study. Clin Nutr. 2000, 19: 245-251. 10.1054/clnu.2000.0106.

Dvir D, Cohen J, Singer P: Computerized energy balance and complications in critically ill patients: an observational study. Clin Nutr. 2005, 25: 37-44. 10.1016/j.clnu.2005.10.010.

Article   Google Scholar  

Villet S, Chioléro RL, Bollmann MD, Revelly JP, Cayeux MC, Delarue J, Berger MM: Negative impact of hypocaloric feeding and energy balance on clinical outcome in ICU patients. Clin Nutr. 2005, 24: 502-509. 10.1016/j.clnu.2005.03.006.

Frankenfield DC, Ashcraft CM: Estimating energy needs in nutrition support patients. J Parenter Enteral Nutr. 2011, 35: 563-570. 10.1177/0148607111415859.

McClave SA, Lowen CC, Kleber MJ, Nicholson JF, Jimmerson SC, McConnel JW, Jung LY: Are patients fed appropriately according to their caloric requirements?. J Parenter Enteral Nutr. 1998, 22: 375-381. 10.1177/0148607198022006375.

McClave SA, Martindale RG, Kiraly L: The use of indirect calorimetry in the intensive care unit. Curr Opin Clin Nutr Metab Care. 2013, 16: 202-208. 10.1097/MCO.0b013e32835dbc54.

Cooney RN, Frankenfield DC: Determining energy needs in critically ill patients: equations or indirect calorimeters. Curr Opin Crit Care. 2012, 18: 174-177. 10.1097/MCC.0b013e3283514bbc.

Faisy C, Guerot E, Diehl JL, Labrousse J, Fagon JY: Assessment of resting energy expenditure in mechanically ventilated patients. Am J Clin Nutr. 2003, 78: 241-249.

Allard JP, Pichard C, Hoshino E, Stechison S, Fareholm L, Peters WJ, Jeejheebhoy KN: Validation of a new formula for calculating energy requirements of burn patients. J Parenter Enteral Nutr. 1990, 14: 115-118. 10.1177/0148607190014002115.

Heidegger CP, Berger MM, Graf S, Zingg W, Darmon P, Costanza MC, Thibault R, Pichard C: Optimisation of energy provision with supplemental parenteral nutrition in critically ill patients: a randomised controlled clinical trial. Lancet. 2013, 381: 385-393. 10.1016/S0140-6736(12)61351-8.

Jolliet P, Pichard C, Biolo G, Chiolero R, Grimble G, Leverve X, Nitenberg G, Novak I, Planas M, Preiser JC, Roth E, Schols AM, Wernerman J: Enteral nutrition in intensive care patients: a practical approach. Working Group on Nutrition and Metabolism, ESICM. European Society of Intensive Care Medicine. Intensive Care Med. 1998, 24: 848-859. 10.1007/s001340050677.

Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G, Van Cromphaut S, Ingels C, Meersseman P, Muller J, Vlasselaers D, Debaveye Y, Desmet L, Dubois J, Van Assche A, Vanderheyden S, Wilmer A, Van den Berghe G: Early versus late parenteral nutrition in critically ill adults. N Engl J Med. 2011, 365: 506-517. 10.1056/NEJMoa1102662.

Singer P, Anbar R, Cohen J, Shapiro H, Shalita-Chesner M, Lev S, Grozovski E, Theilla M, Frishman S, Madar Z: The tight calorie control study (TICACOS): a prospective, randomized, controlled pilot study of nutritional support in critically ill patients. Intensive Care Med. 2011, 37: 601-609. 10.1007/s00134-011-2146-z.

Grau T, Bonet A, Rubio M, Mateo D, Farre M, Acosta JA, Blesa A, Montejo JC, de Lorenzo AG, Mesejo A: Liver dysfunction associated with artificial nutrition in critically ill patients. Crit Care. 2007, 11: R10-10.1186/cc5670.

Article   PubMed Central   PubMed   Google Scholar  

Kim H, Stotts NA, Froelicher ES, Engler MM, Porter C: Why patients in critical care do not receive adequate enteral nutrition? A review of the literature. J Crit Care. 2012, 27: 702-713. 10.1016/j.jcrc.2012.07.019.

Frankenfield D: Validation of an equation for resting metabolic rate in older obese, critically ill patients. J Parenter Enteral Nutr. 2011, 35: 264-269. 10.1177/0148607110377903.

Crouser ED: Autophagy, the first step towards recovery from critical illness. Crit Care Med. 2013, 41: 358-359. 10.1097/CCM.0b013e31826bf37c.

Strappazzon F, Campello S, Cecconi F: Non-apoptotic roles for death-related molecules: when mitochondria chose cell fate. Exp Cell Res. 2012, 318: 1309-1315. 10.1016/j.yexcr.2012.01.027.

Fürst P, Stehle P: Are intravenous amino acid solutions unbalanced?. New Horiz. 1994, 2: 215-223.

PubMed   Google Scholar  

Wischmeyer PE: Glutamine: role in critical illness and ongoing clinical trials. Curr Opin Gastroenterol. 2008, 24: 190-197. 10.1097/MOG.0b013e3282f4db94.

Dechelotte P, Hasselmann M, Cynober L, Allaouchiche B, Coeffier M, Hecketsweiler B, Merle V, Mazerolles M, Samba D, Guillou YM, Petit J, Mansoor O, Colas G, Cohendy R, Barnoud D, Czernichow P, Bleichner G: l -alanyl- l -glutamine dipeptide-supplemented total parenteral nutrition reduces infectious complications and glucose intolerance in critically ill patients: the French controlled, randomized, double-blind, multicenter study. Crit Care Med. 2006, 34: 598-604. 10.1097/01.CCM.0000201004.30750.D1.

Goeters C, Wenn A, Mertes N, Wempe C, Van Aken H, Stehle P, Bone HG: Parenteral l -alanyl- l -glutamine improves 6-month outcome in critically ill patients. Crit Care Med. 2002, 30: 2032-2037. 10.1097/00003246-200209000-00013.

Stehle P, Zander J, Mertes N, Albers S, Puchstein C, Lawin P, Fürst P: Effect of parenteral glutamine peptide supplements on muscle glutamine loss and nitrogen balance after major surgery. Lancet. 1989, 1: 231-233. 10.1016/S0140-6736(89)91254-3.

Grau T, Bonet A, Miñambres E, Piñeiro L, Irles JA, Robles A, Acosta J, Herrero I, Palacios V, Lopez J, Blesa A, Martínez P: The effect of l -alanyl- l -glutamine dipeptide supplemented total parenteral nutrition on infectious morbidity and insulin sensitivity in critically ill patients. Crit Care Med. 2011, 39: 1263-1268. 10.1097/CCM.0b013e31820eb774.

Andrews PJ, Avenell A, Noble DW, Campbell MK, Croal BL, Simpson WG, Vale LD, Battison CG, Jenkinson DJ, Cook JA: Randomised trial of glutamine, selenium, or both, to supplement parenteral nutrition for critically ill patients. BMJ. 2011, 342: d1542-10.1136/bmj.d1542.

Luo M, Bazargan N, Griffith DP, Estivariz CF, Leader LM, Easley KA, Daignault NM, Hao L, Meddings JB, Galloway JR, Blumberg JB, Jones DP, Ziegler TR: Metabolic effects of enteral versus parenteral alanyl-glutamine dipeptide administration in critically ill patients receiving enteral feeding: a pilot study. Clin Nutr. 2008, 27: 297-306. 10.1016/j.clnu.2007.12.003.

Heyland DK, Muscedere J, Wischmeyer PE, Cook D, Jones G, Albert M, Elke G, Berger MM, Day AG: A randomized trial of glutamine and antioxidants in critically ill patients. N Engl J Med. 2013, 368: 1489-1497. 10.1056/NEJMoa1212722.

Rodas PC, Rooyackers O, Hebert C, Norberg A, Wernerman J: Glutamine and glutathione at ICU admission in relation to outcome. Clin Sci (Lond). 2012, 122: 591-597. 10.1042/CS20110520.

Weissman C, Chiolero R, Askanazi J, Gil KM, Elwyn D, Kinney JM: Intravenous infusion of a medium-chain triglyceride-enriched lipid emulsion. Crit Care Med. 1988, 16: 1183-1190. 10.1097/00003246-198812000-00003.

Calder PC: N-3 fatty acids, inflammation, and immunity – relevance to postsurgical and critically ill patients. Lipids. 2004, 39: 1147-1161. 10.1007/s11745-004-1342-z.

Heller AR, Rossler S, Litz RJ, Stehr SN, Heller SC, Koch R, Koch T: Omega-3 fatty acids improve the diagnosis-related clinical outcome. Crit Care Med. 2006, 34: 972-979. 10.1097/01.CCM.0000206309.83570.45.

Berger MM, Delodder F, Liaudet L, Tozzi P, Schlaepfer J, Chiolero RL, Tappy L: Three short perioperative infusions of n-3 PUFAs reduce systemic inflammation induced by cardiopulmonary bypass surgery: a randomized controlled trial. Am J Clin Nutr. 2013, 97: 246-254. 10.3945/ajcn.112.046573.

Calder PC: Lipids for intravenous nutrition in hospitalised adult patients: a multiple choice of options. Proc Nutr Soc. 2013, 72: 263-276. 10.1017/S0029665113001250.

Pradelli L, Iannazzo S, Zaniolo O, Muscaritoli M, Eandi M: Effectiveness and cost-effectiveness of supplemental glutamine dipeptide in total parenteral nutrition therapy for critically ill patients: a discrete event simulation model based on Italian data. Int J Technol Assess Health Care. 2012, 28: 22-28. 10.1017/S0266462311000705.

Pradelli L, Mayer K, Muscaritoli M, Heller AR: n-3 fatty acid-enriched parenteral nutrition regimens in elective surgical and ICU patients: a meta-analysis. Crit Care. 2012, 16: R184-10.1186/cc11668.

Doig GS, Simpson F, Early PN: Trial Investigators Group: Early parenteral nutrition in critically ill patients with short-term relative contraindications to early enteral nutrition: a randomized controlled trial. Clinicoecon Outcomes Res. 2013, 5: 369-379. 10.2147/CEOR.S48821.

Guidelines for the use of parenteral and enteral nutrition in adult and pediatric patients. J Parenter Enteral Nutr. 2002, 26: 1SA-138SA. 10.1177/014860710202600501.

McClave SA, Heyland DK, Martindale RG: Adding supplemental parenteral nutrition to hypocaloric enteral nutrition: lessons learned from the Casaer Van den Berghe study. J Parenter Enteral Nutr. 2012, 36: 15-17. 10.1177/0148607111424160.

Wilmer A, Van den Berghe G: Supplemental parenteral nutrition in critically ill patients. The Lancet. 2013, 381: 1715-10.1016/S0140-6736(13)61069-7. b) Marik P, Hooper M: Supplemental parenteral nutrition in critically ill patients. The Lancet 2013, 381:1716. c) Lopes A, Lloret Linares C, Bergmann J-F: Supplemental parenteral nutrition in critically ill patients. The Lancet 2013, 381:1716. d) Shime N, Maclaren G: Supplemental parenteral nutrition in critically ill patients. The Lancet 2013, 381:1715–1716.

Doig GS, Simpson F, Sweetman EA, Finfer SR, Cooper DJ, Heighes PT, Davies AR, O'Leary M, Solano T, Peake S: Early parenteral nutrition in critically ill patients with short-term relative contraindications to early enteral nutrition: a randomized controlled trial. JAMA. 2013, 309: 2130-2138. 10.1001/jama.2013.5124.

Thibault R, Graf S, Clerc A, Delieuvin N, Heidegger CP, Pichard C: Diarrhoea in the intensive care unit: respective contribution of feeding and antibiotics. Crit Care. 2013, 17: R153-10.1186/cc12832.

Pichard C, Attaix D, Baracos VE: A pragmatic and reasonable approach seems the best option to adapt nutrition support to the specific individual patients needs and thus optimize the global care. Curr Opin Clin Nutr Metab Care. 2013, 16: 241-242. 10.1097/MCO.0b013e328360520e.

Soguel L, Revelly JP, Schaller MD, Longchamp C, Berger MM: Impact of a stepwise introduction of multidisciplinary nutrition protocol with an ICU dietician on the quality of nutritional support and on energy balance. Crit Care Med. 2012, 40: 412-419. 10.1097/CCM.0b013e31822f0ad7.

Download references

Author information

Authors and affiliations.

Service de Médecine Intensive Adulte et Brûlés, , Lausanne University Hospital (CHUV), Lausanne, 1011, Switzerland

Mette M Berger

Clinical Nutrition, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4, 1211, Geneva 14, Switzerland

Claude Pichard

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Mette M Berger .

Additional information

Competing interests.

MMB received financial support from research grants and unrestricted academic research grants from public institutions (Fonds National Suisse de la Recherche Scientifique) and from industry: Aguettant, Baxter, B. Braun, Fresenius-Kabi AG, Nestle Medical Nutrition, and Novartis. CP received financial support from research grants and unrestricted academic research grants from the public institutions, as well as no-restrictive research grants and consulting fees from the Foundation Nutrition 2000plus, Abbott, Baxter, B. Braun, Cosmed, Fresenius-Kabi, Nestle Medical Nutrition, Novartis, Nutricia – Numico, Pfizer, and Solvay.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2, authors’ original file for figure 3, rights and permissions.

Reprints and permissions

About this article

Cite this article.

Berger, M.M., Pichard, C. Development and current use of parenteral nutrition in critical care – an opinion paper. Crit Care 18 , 478 (2014). https://doi.org/10.1186/s13054-014-0478-0

Download citation

Published : 08 August 2014

DOI : https://doi.org/10.1186/s13054-014-0478-0

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Critical Care

ISSN: 1364-8535

total parenteral nutrition research articles

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Total parenteral nutrition

Affiliation.

  • 1 Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Parel, Mumbai 400012, Maharashtra, India. [email protected]
  • PMID: 12715955

Parenteral nutrition is a form of therapy in which elemental nutrients (sugars, lipids, amino acids, vitamins and minerals) are given as an intravenous infusion. It is complementary and not competitive to enteral nutrition. While a vast majority of patients can be managed by enteral nutrition, a few patients need parenteral nutrition for survival. Very few patients may need both enteral and parenteral nutrition for short periods. The indications, delivery methods and formulations of parenteral nutrition have been refined in the past 30 years. It is now possible to give parenteral nutrition for prolonged periods at home. Three-in-one parenteral nutrition mixture bags are presently available in India. This article discusses the practical aspects of using parenteral nutrition in everyday practice.

PubMed Disclaimer

Similar articles

  • [Approaches in parenteral nutrition]. Löhlein D. Löhlein D. Infusionstherapie. 1991 Aug;18(4):182-7. Infusionstherapie. 1991. PMID: 1937841 Review. German.
  • Practical aspects and complications of total parenteral nutrition. Benotti PN, Bistrian BR. Benotti PN, et al. Crit Care Clin. 1987 Jan;3(1):115-31. Crit Care Clin. 1987. PMID: 3145104 Review.
  • [Parenteral hyperalimentation (author's transl)]. Göschke H, Leutenegger A. Göschke H, et al. Wien Klin Wochenschr. 1977 Mar 4;89(5):141-6. Wien Klin Wochenschr. 1977. PMID: 402748 German.
  • Clinical issues in the therapeutic monitoring of total parenteral nutrition. Driscoll DF, Bistrian BR. Driscoll DF, et al. Clin Lab Med. 1987 Sep;7(3):699-714. Clin Lab Med. 1987. PMID: 3115669 Review.
  • Peripheral versus central intravenous nutrition: comparison of two delivery systems. Everitt NJ. Everitt NJ. Br J Surg. 1994 Jun;81(6):920; author reply 921. Br J Surg. 1994. PMID: 8044624 No abstract available.

Publication types

  • Search in MeSH

Related information

  • Cited in Books
  • PubChem Compound
  • PubChem Compound (MeSH Keyword)
  • PubChem Substance

LinkOut - more resources

Other literature sources.

  • The Lens - Patent Citations

Miscellaneous

  • NCI CPTAC Assay Portal
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

  • Download PDF
  • Share X Facebook Email LinkedIn
  • Permissions

Guidelines for Total Parenteral Nutrition

From the departments of medicine, Memorial Hospital for Cancer and Allied Diseases and Cornell University Medical College, and the Division of Medical Research, Sloan-Kettering Institute for Cancer Research, New York.

Total parenteral nutrition is a procedure capable of maintaining patients in good condition for prolonged periods while underlying disease is being treated. Achievement of this goal presents problems in formulation, in sterile preparation, and in safe administration of the nutrient solution. A practical program is outlined for simple, inexpensive, and sterile preparation of standard solutions in closed systems. Indications and precautions for this potentially life-saving treatment are discussed together with consideration of routes of administration, ancillary equipment, and nutrient requirements for total parenteral nutrition.

Shils ME. Guidelines for Total Parenteral Nutrition. JAMA. 1972;220(13):1721–1729. doi:10.1001/jama.1972.03200130051010

Manage citations:

© 2024

Artificial Intelligence Resource Center

Cardiology in JAMA : Read the Latest

Browse and subscribe to JAMA Network podcasts!

Others Also Liked

Select your interests.

Customize your JAMA Network experience by selecting one or more topics from the list below.

  • Academic Medicine
  • Acid Base, Electrolytes, Fluids
  • Allergy and Clinical Immunology
  • American Indian or Alaska Natives
  • Anesthesiology
  • Anticoagulation
  • Art and Images in Psychiatry
  • Artificial Intelligence
  • Assisted Reproduction
  • Bleeding and Transfusion
  • Caring for the Critically Ill Patient
  • Challenges in Clinical Electrocardiography
  • Climate and Health
  • Climate Change
  • Clinical Challenge
  • Clinical Decision Support
  • Clinical Implications of Basic Neuroscience
  • Clinical Pharmacy and Pharmacology
  • Complementary and Alternative Medicine
  • Consensus Statements
  • Coronavirus (COVID-19)
  • Critical Care Medicine
  • Cultural Competency
  • Dental Medicine
  • Dermatology
  • Diabetes and Endocrinology
  • Diagnostic Test Interpretation
  • Drug Development
  • Electronic Health Records
  • Emergency Medicine
  • End of Life, Hospice, Palliative Care
  • Environmental Health
  • Equity, Diversity, and Inclusion
  • Facial Plastic Surgery
  • Gastroenterology and Hepatology
  • Genetics and Genomics
  • Genomics and Precision Health
  • Global Health
  • Guide to Statistics and Methods
  • Hair Disorders
  • Health Care Delivery Models
  • Health Care Economics, Insurance, Payment
  • Health Care Quality
  • Health Care Reform
  • Health Care Safety
  • Health Care Workforce
  • Health Disparities
  • Health Inequities
  • Health Policy
  • Health Systems Science
  • History of Medicine
  • Hypertension
  • Images in Neurology
  • Implementation Science
  • Infectious Diseases
  • Innovations in Health Care Delivery
  • JAMA Infographic
  • Law and Medicine
  • Leading Change
  • Less is More
  • LGBTQIA Medicine
  • Lifestyle Behaviors
  • Medical Coding
  • Medical Devices and Equipment
  • Medical Education
  • Medical Education and Training
  • Medical Journals and Publishing
  • Mobile Health and Telemedicine
  • Narrative Medicine
  • Neuroscience and Psychiatry
  • Notable Notes
  • Nutrition, Obesity, Exercise
  • Obstetrics and Gynecology
  • Occupational Health
  • Ophthalmology
  • Orthopedics
  • Otolaryngology
  • Pain Medicine
  • Palliative Care
  • Pathology and Laboratory Medicine
  • Patient Care
  • Patient Information
  • Performance Improvement
  • Performance Measures
  • Perioperative Care and Consultation
  • Pharmacoeconomics
  • Pharmacoepidemiology
  • Pharmacogenetics
  • Pharmacy and Clinical Pharmacology
  • Physical Medicine and Rehabilitation
  • Physical Therapy
  • Physician Leadership
  • Population Health
  • Primary Care
  • Professional Well-being
  • Professionalism
  • Psychiatry and Behavioral Health
  • Public Health
  • Pulmonary Medicine
  • Regulatory Agencies
  • Reproductive Health
  • Research, Methods, Statistics
  • Resuscitation
  • Rheumatology
  • Risk Management
  • Scientific Discovery and the Future of Medicine
  • Shared Decision Making and Communication
  • Sleep Medicine
  • Sports Medicine
  • Stem Cell Transplantation
  • Substance Use and Addiction Medicine
  • Surgical Innovation
  • Surgical Pearls
  • Teachable Moment
  • Technology and Finance
  • The Art of JAMA
  • The Arts and Medicine
  • The Rational Clinical Examination
  • Tobacco and e-Cigarettes
  • Translational Medicine
  • Trauma and Injury
  • Treatment Adherence
  • Ultrasonography
  • Users' Guide to the Medical Literature
  • Vaccination
  • Venous Thromboembolism
  • Veterans Health
  • Women's Health
  • Workflow and Process
  • Wound Care, Infection, Healing
  • Register for email alerts with links to free full-text articles
  • Access PDFs of free articles
  • Manage your interests
  • Save searches and receive search alerts

Search

Parenteral Nutrition (PN)

(total parenteral nutrition).

  • Indications for Parenteral Nutrition |
  • Nutritional Requirements and Formula Content |
  • Solutions for Parenteral Nutrition |
  • Administration of Parenteral Nutrition |
  • Monitoring of Parenteral Nutrition |
  • Complications of Parenteral Nutrition |
  • Key Points |

Parenteral nutrition (PN) is by definition given IV. PN may be infused through a peripheral or central venous access device, depending on the osmolarity of the solution.

Peripheral PN may be infused through a peripheral venous access device. The osmolarity of the solution should be ≤ 900 mOsm/L. A solution with a higher osmolarity may cause thrombophlebitis.

Central PN requires a central venous access device for safe infusion because solutions formulated for central PN have an osmolarity > 900 mOsm/L.

PN should not be used routinely in patients with an intact gastrointestinal (GI) tract. Compared with enteral nutrition , PN has the following disadvantages:

More complications

Poorer preservation of the structure and function of the GI tract

Higher cost

Indications for Parenteral Nutrition

PN may be the only feasible option for patients who do not have a functioning GI tract or who have disorders requiring complete bowel rest, such as the following ( 1 ):

Some stages of ulcerative colitis

Bowel obstruction

Certain pediatric GI disorders (eg, congenital GI anomalies , prolonged diarrhea regardless of its cause)

Short bowel syndrome due to surgery

Indications reference

1. Worthington P, Balint J, Bechtold M, et al . When Is Parenteral Nutrition Appropriate?.  JPEN J Parenter Enteral Nutr . 2017;41(3):324-377. doi:10.1177/0148607117695251

Nutritional Requirements and Formula Content

PN requires that patients receive water, usually 25 to 40 mL/kg body weight/day. Less water is needed for patients requiring fluid restriction, energy , amino acids, lipids, vitamins, and minerals (see table Basic Adult Daily Requirements for Parenteral Nutrition ).

Children who need PN may have different fluid requirements and need more energy (up to 120 kcal/kg/day) and amino acids (up to 2.5 or 3.5 g/kg/day).

Basic Adult Daily Requirements for Parenteral Nutrition

Water (/kg body weight/day)

25–40 mL

Medical patient

25–35 kcal

Postoperative patient

25–35 kcal

Hypercatabolic patient

30–45 kcal

Critically ill patient with BMI 30–50 kg/m

11–14 kcal/kg actual body weight

Critically ill patient with BMI > 50 kg/m

22–25 kcal/kg ideal body weight

Medical patient

1.0–2.0 g

Postoperative patient

1.5–2.0 g

Critically ill/hypercatabolic patient

1.2–2.0 g

Continuous renal replacement therapy

1.5–2.5 g

Critically ill patient with class I or class II obesity

≥ 2 g/kg ideal body weight

Critically ill patient with class III obesity

≥ 2.5 g/kg ideal body weight

Acetate

As needed to maintain acid-base balance

Calcium

10–15 mEq

Chloride

As needed to maintain acid-base balance

Chromium

≤ 10 mcg

Copper

0.3–0.5 mg

Magnesium

8–20 mEq

Manganese

55 mcg

Phosphorus

20–40 mmol

Potassium

1–2 mEq/kg

Selenium

60–100 mcg

Sodium

1–2 mEq/kg

Zinc

3–5 mg

Ascorbic acid

200 mg

Biotin

60 mcg

Cobalamin

5 mcg

Folate (folic acid)

600 mcg

Niacin

40 mg

Pantothenic acid

15 mg

Pyridoxine

6 mg

Riboflavin

3.6 mg

Thiamin

6 mg

Vitamin A

990 mcg

Vitamin D

5 mcg

Vitamin E

10 mg

Vitamin K

150 mcg

° C of fever.

. Appropriate Dosing for Parenteral Nutrition (ASPEN Recommendations). 2019. Accessed August 26, 2024.

Solutions for Parenteral Nutrition

Individualized PN solutions may be prepared using sterile techniques. For individualized PN solutions, electrolytes may be adjusted based on laboratory results and changes in clinical status.

Standardized, commercially available PN solutions are available for institutions that do not have the resources to make individualized solutions.

Standard vitamin and trace element preparations also are available to add to PN to meet micronutrient requirements. Stability of the PN solution depends on many factors. Clinicians should avoid adding electrolytes to standardized PN solutions to avoid the risk of electrolyte precipitation. Because stability can be reduced by modifications to PN solutions, modifications (eg, adding electrolytes or other substances) should be reviewed by an experienced pharmacist.

PN macronutrient composition varies depending on solution type:

Peripheral PN: Lower amino acid and dextrose concentrations to keep the osmolarity low, with most calories from fat

Central PN: Higher amino acid and dextrose concentrations to provide more energy in less volume

Amino acid dose is based on the patient's calculated protein requirements.

Availability of mixed-oil lipid emulsions has changed the approach to lipid dosing in PN, intending to balance provision of nutrients with potential adverse effects. Soybean oil is rich in omega-6 polyunsaturated fatty acids, which have been associated with proinflammatory and immunosuppressive effects. Thus, traditional lipid emulsions of 100% soybean oil should be restricted to fish oil , which have lower omega-6 polyunsaturated fatty acids and allow for higher total fat delivery. For adults (both stable and critically ill), 1 to 1.5 g/kg/day of the lipid emulsion blend of olive oil and soybean oil can be given, whereas 1 to 2 g/kg/day of the lipid emulsion blend of soybean oil, MCTs, olive oil, and fish oil can be given. In the United States, 100% fish oil lipid emulsion is not approved for use in adults, but it is used as a therapy for children with intestinal-failure–associated liver disease. Additional dosing considerations (eg, medication additives; calcium, phosphorus, and magnesium content) to ensure solution stability should be reviewed by an experienced pharmacist. The American Society for Parenteral and Enteral Nutrition has published recommendations for appropriate dosing of lipid emulsions ( 1 , 2 ).

PN solution volumes, types, and concentrations vary depending on patient factors, such as concomitant disorders and age:

For patients with heart, liver, or kidney failure: Limited volume (liquid) intake

For patients with diabetes: Mixed-oil lipid emulsion, providing the upper end of the recommended dosing range to allow for reduction of dextrose delivery

For neonates: Lower dextrose concentrations (17 to 18%)

Solutions references

1. Mirtallo JM, Ayers P, Boullata J, et al . ASPEN Lipid Injectable Emulsion Safety Recommendations, Part 1: Background and Adult Considerations [published correction appears in Nutr Clin Pract . 2022 Apr;37(2):482].  Nutr Clin Pract . 2020;35(5):769-782. doi:10.1002/ncp.10496

2. Cober MP, Gura KM, Mirtallo JM, et al . ASPEN lipid injectable emulsion safety recommendations part 2: Neonate and pediatric considerations.  Nutr Clin Pract . 2021;36(6):1106-1125. doi:10.1002/ncp.10778

Administration of Parenteral Nutrition

As with any central venous catheter, strict sterile technique must be used during insertion and maintenance of the catheter. The PN lumen of the central line should not be used for any other purpose. External tubing should be changed every 24 hours with the first PN solution bag of the day. The American Society for Parenteral and Enteral Nutrition recommends using a 1.2-micron in-line filter with PN to reduce exposure to particulate matter ( 1 ). Dressings should be kept sterile and are usually changed every 48 hours using strict sterile techniques.

If PN is given outside the hospital, patients and their care partners must be taught appropriate line care, PN administration, and identification of complications, including symptoms of infection. Qualified home nursing must be arranged.

PN should be advanced based on electrolyte and clinical stability. If a patient is hyperglycemic, blood glucose concentrations should be monitored and regular insulin should be provided. No other insulin type is compatible with PN solutions. The target blood glucose level for patients receiving PN is insulin insulin . Collaboration with an endocrinologist can help optimize management of blood glucose.

Administration reference

1. Worthington P, Gura KM, Kraft MD, et al . Update on the Use of Filters for Parenteral Nutrition: An ASPEN Position Paper.  Nutr Clin Pract . 2021;36(1):29-39. doi:10.1002/ncp.10587

Monitoring of Parenteral Nutrition

An interdisciplinary nutrition team composed of a physician, dietitian, pharmacist, and nurse, if available, should monitor patients.

Complete blood count should be obtained. Weight, electrolytes, magnesium, phosphorus, blood urea nitrogen, and creatinine should be monitored often (eg, daily for inpatients, weekly for outpatients). Plasma glucose should be monitored every 6 hours until patients and their glucose levels become stable. Liver tests should be monitored at least weekly, but more frequent monitoring may be required if test results are abnormal. Triglyceride levels should be monitored at least twice/week in inpatients receiving lipid emulsions. Fluid intake and output should be monitored continuously.

When patients become stable, blood tests can be done less often. Blood tests should not be done while PN is being infused.

Full nutritional assessment (including BMI calculation , anthropometric measurements , and body composition analysis ) should be repeated as often as dictated by a patient's clinical status. Adjustments to the nutritional assessment may be needed more frequently in patients who are critically ill and less frequently in outpatients who are stable and receiving PN.

total parenteral nutrition research articles

Complications of Parenteral Nutrition

About 5 to 10% of patients with a PN line have complications related to central venous access .

Catheter-related sepsis rates have decreased since the introduction of CDC guidelines that emphasize sterile techniques for catheter insertion and skin care around the insertion site ( 1 ). The increasing use of dedicated teams of physicians and nurses who specialize in various procedures including catheter insertion also has contributed to a decrease in catheter-related infection rates.

Infectious complications associated with PN have been reduced by changes in PN management such as avoiding overfeeding, maintaining optimal blood glucose concentrations, and using mixed-oil lipid emulsions.

Glucose abnormalities are common. Hyperglycemia can be avoided by monitoring plasma glucose often, adjusting the insulin dose in the PN solution, and giving subcutaneous insulin as needed. Hypoglycemia can be precipitated by suddenly stopping constant concentrated dextrose

Hepatic complications include liver dysfunction, painful hepatomegaly, and hyperammonemia. These complications can develop at any age but are most common among infants, particularly premature ones (whose liver is immature).

Liver dysfunction may be transient, evidenced by increased transaminases, bilirubin, and alkaline phosphatase; it commonly occurs when PN is started. Delayed or persistent elevations may result from excess amino acids. Pathogenesis is unknown, but cholestasis and inflammation may contribute. Progressive fibrosis occasionally develops. Fish oil lipid emulsions may be beneficial; in children, 100% fish oil lipid emulsion is a rescue therapy for this complication.

If infants develop any hepatic complication, limiting amino acids to 1.0 g/kg/day may be necessary.

Abnormalities of serum electrolytes and minerals should be corrected by modifying subsequent infusions or, if correction is urgently required, by beginning appropriate peripheral vein infusions. Vitamin and mineral deficiencies are rare when solutions are given correctly. Elevated blood urea

Volume overload (suggested by > 1 kg/day weight gain) may occur when patients have high daily energy requirements and thus require large fluid volumes. A pharmacist with expertise managing PN should review the PN solution to determine whether the volume of the solution can be reduced.

Metabolic bone disease, or bone demineralization (osteoporosis or osteomalacia), develops in some patients given PN for > 3 months. The mechanism is unknown. Advanced disease can cause severe periarticular, lower-extremity, and back pain.

Adverse reactions to lipid emulsions (eg, dyspnea, cutaneous allergic reactions, nausea, headache, back pain, sweating, dizziness) are uncommon but may occur early, particularly if lipids are given at > 1.0 kcal/kg/hour. Temporary hyperlipidemia may occur, particularly in patients with kidney or liver failure; treatment is usually not required. Delayed adverse reactions to lipid emulsions include hepatomegaly, mild elevation of liver enzymes, splenomegaly, thrombocytopenia, leukopenia, and, especially in premature infants with respiratory distress syndrome, pulmonary function abnormalities. Temporarily or permanently slowing or stopping lipid emulsion infusion may prevent or minimize these adverse reactions.

Gallbladder complications

Complications reference

1. Centers for Disease Control and Prevention (CDC) : Guidelines for the Prevention of Intravascular Catheter-Related Infections (2011). Accessed May 9, 2024.

Consider parenteral nutrition for patients who do not have a functioning gastrointestinal tract or who have disorders requiring complete bowel rest.

Calculate requirements for water, energy, amino acids, lipids, vitamins, and minerals.

Choose a solution based on patient age and organ function status; different solutions are required for neonates and for patients who have compromised heart, kidney, or liver function.

Use a central venous catheter, with strict sterile technique for insertion and maintenance.

Monitor patients closely for complications (eg, related to central venous access; abnormal glucose, electrolyte, mineral levels; hepatic or gallbladder effects; reactions to lipid emulsions; and volume overload or dehydration).

quizzes_lightbulb_red

Copyright © 2024 Merck & Co., Inc., Rahway, NJ, USA and its affiliates. All rights reserved.

  • Cookie Preferences

This icon serves as a link to download the eSSENTIAL Accessibility assistive technology app for individuals with physical disabilities. It is featured as part of our commitment to diversity and inclusion. M

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

The PMC website is updating on October 15, 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • v.153(6); 1990 Dec

Total parenteral nutrition.

Full text is available as a scanned copy of the original print version. Get a printable copy (PDF file) of the complete article (246K), or click on a page image below to browse page by page. Links to PubMed are also available for Selected References .

icon of scanned page 662

Selected References

These references are in PubMed. This may not be the complete list of references from this article.

  • Katz SJ, Oye RK. Parenteral nutrition use at a university hospital. Factors associated with inappropriate use. West J Med. 1990 Jun; 152 (6):683–686. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Mullen JL, Buzby GP, Matthews DC, Smale BF, Rosato EF. Reduction of operative morbidity and mortality by combined preoperative and postoperative nutritional support. Ann Surg. 1980 Nov; 192 (5):604–613. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Williams RH, Heatley RV, Lewis MH. Proceedings: A randomized controlled trial of preoperative intravenous nutrition in patients with stomach cancer. Br J Surg. 1976 Aug; 63 (8):667–667. [ PubMed ] [ Google Scholar ]
  • Starker PM, LaSala PA, Askanazi J, Todd G, Hensle TW, Kinney JM. The influence of preoperative total parenteral nutrition upon morbidity and mortality. Surg Gynecol Obstet. 1986 Jun; 162 (6):569–574. [ PubMed ] [ Google Scholar ]
  • Askanazi J, Hensle TW, Starker PM, Lockhart SH, LaSala PA, Olsson C, Kinney JM. Effect of immediate postoperative nutritional support on length of hospitalization. Ann Surg. 1986 Mar; 203 (3):236–239. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Young GA, Zeiderman MR, Thompson M, McMahon MJ. Influence of preoperative intravenous nutrition upon hepatic protein synthesis and plasma proteins and amino acids. JPEN J Parenter Enteral Nutr. 1989 Nov-Dec; 13 (6):596–602. [ PubMed ] [ Google Scholar ]

IMAGES

  1. Feeling the Impact of Long-Term Total Parenteral Nutrition

    total parenteral nutrition research articles

  2. Composition of total parenteral nutrition solution Ingredient

    total parenteral nutrition research articles

  3. (PDF) Total parenteral nutrition in critically ill and injured patients

    total parenteral nutrition research articles

  4. PPT

    total parenteral nutrition research articles

  5. Total Parenteral Nutrition (TPN): Uses, Administration, Contents, Side

    total parenteral nutrition research articles

  6. SOLUTION: Total parenteral nutrition

    total parenteral nutrition research articles

VIDEO

  1. Protein in Parenteral Nutrition

  2. Parenteral nutrition and Enteral nutrition #nursingnotes

  3. Total Parenteral Nutrition clinimix clinoleic pasien icu #intensivecareunit #shorts

  4. TPN ( Total parenteral nutrition) التغذية الوريدية الكاملة

  5. ENTERAL nutrition and it's TYPES. intro to parenteral nutrition. MODES OF FEEDING

  6. How I Hook up to TPN! (Total Parenteral Nutrition)

COMMENTS

  1. Advances in Medical Nutrition Therapy: Parenteral Nutrition

    Abstract. Parenteral nutrition has evolved tremendously, with parenteral formulas now safer and more accessible than ever. "All-in-one" admixtures are now available, which simplify parenteral nutrition usage and decrease line infection rates alongside other methods of infectious control. Recently published data on the benefits of parenteral ...

  2. Total Parenteral Nutrition

    Total parenteral nutrition is a medication used to manage and treat malnourishment. It is in the nutrition class of drugs. Total parenteral nutrition is indicated when there is impaired gastrointestinal function and contraindications to enteral nutrition. Total parenteral nutrition (TPN) is when IV-administered nutrition is the only source of nutrition the patient is receiving. This activity ...

  3. Parenteral nutrition in clinical practice: International challenges and

    Parenteral nutrition (PN), used in a variety of settings in order to provide nutritional needs when oral/enteral feeding is not sufficient or is contraindicated, is a complex, high-alert medication requiring training and ongoing competency assessment to ensure patient safety. 1 The PN use process (Figure 1) includes a number of key steps: patient assessment, prescribing and ordering, order ...

  4. Total Parenteral Nutrition

    Total parenteral nutrition is a medication used to manage and treat malnourishment. It is in the nutrition class of drugs. Total parenteral nutrition is indicated when there is impaired gastrointestinal function and contraindications to enteral nutrition. Total parenteral nutrition (TPN) is when IV-administered nutrition is the only source of ...

  5. Parenteral Nutrition

    Research, Methods, Statistics; Resuscitation; Rheumatology; Risk Management; Scientific Discovery and the Future of Medicine; ... If patients are unable to tolerate enteral nutrition for longer than a few days, total parenteral nutrition (TPN) can provide all the types of nutrients that a patient needs. Although TPN provides complete ...

  6. Early versus Late Parenteral Nutrition in Critically Ill Adults

    Methods. In this randomized, multicenter trial, we compared early initiation of parenteral nutrition (European guidelines) with late initiation (American and Canadian guidelines) in adults in the ...

  7. Total parenteral nutrition: A boon to non-functional gastrointestinal

    1. Introduction. Total parenteral nutrition (TPN) is a medication used in the management and treatment of malnourishment. 1 TPN is the intravenous administration of nutrition outside of the gastrointestinal tract. TPN is given when the I.V administered nutrition is the only source of nutrition. 2 It is indicated when there is an inadequate gastrointestinal function and contraindications to ...

  8. Total Parenteral Nutrition

    Total parenteral nutrition (TPN) is defined as parenteral (intravenous [IV]) nutritional support that includes calories, amino acids, electrolytes, vitamins, minerals, and trace elements. TPN is indicated in patients for a variety of conditions revolving around intestinal insufficiency due to decreased intestine length or functionality.

  9. (PDF) Total Parenteral Nutrition

    T otal parenteral nutrition is a medication used in the management and treatment of malnourishment. It is in the. nutrition class of drugs. This activity describes the indications, action, and ...

  10. Total parenteral nutrition in the management of acute renal failure

    Total parenteral nutrition has been reported to stabilize or reduce serum urea nitrogen, potassium and phosphorus levels, improve wound healing, enhance survival from acute renal failure, and possibly increase the rate of recovery of renal function. ... Original Research Article. The effects of chicory inulin-type fructans supplementation on ...

  11. (PDF) Parenteral nutrition and nursing care: A review of the recent

    Abstract. Parenteral nutrition is a life-saving intervention for patients where enteral nutrition cannot be achieved. In recent years, parenteral nutrition has been widely used thanks to advances ...

  12. Research paper Stability and compatibility of parenteral nutrition

    Standardized PN products contain fixed sets of constituents, usually named standardized total parenteral nutrition (STPN), which are designed to meet the average patients' nutritional requirements with consideration to different requirements in different age group. ... This research was carried out as part of a larger project that aimed to ...

  13. Parenteral Nutrition: Current Use, Complications, and Nutrition

    The present research was conducted to provide further analysis of the use of PN in adult critically ill patients, especially when used early and as the initial route. ... Martínez de Lagrán I., Campins L. Transition from total parenteral nutrition to enteral nutrition in critically ill patients in Spain: A national survey. Med. Intensiva ...

  14. Development and current use of parenteral nutrition in critical care

    Critically ill patients depend on artificial nutrition for the maintenance of their metabolic functions and lean body mass, as well as for limiting underfeeding-related complications. Current guidelines recommend enteral nutrition (EN), possibly within the first 48 hours, as the best way to provide the nutrients and prevent infections. EN may be difficult to realize or may be contraindicated ...

  15. Clinical Nutrition: Parenteral Nutrition

    The introduction of total parenteral nutrition in the 1960s helped to establish nutrition support as a specific therapeutic entity. The field of nutrition support is based on the tenets that nutrient depletion causes morbidity and mortality and that the prevention or correction of nutrient deficiencies can minimize or eliminate the adverse effects of malnourishment.

  16. Mortality and overall and specific infection complication rates in

    Original Research Article. Folate and cobalamin status, indicators, modulators, interactions and reference ranges from early pregnancy until birth: The Reus-Tarragona Birth Cohort Study. ... Early enteral nutrition and total parenteral nutrition on the nutritional status and blood glucose in patients with gastric cancer complicated with ...

  17. Total parenteral nutrition

    Parenteral nutrition is a form of therapy in which elemental nutrients (sugars, lipids, amino acids, vitamins and minerals) are given as an intravenous infusion. ... While a vast majority of patients can be managed by enteral nutrition, a few … Total parenteral nutrition Natl Med J India. 2003 Jan-Feb;16(1):29-33. Authors ... Research Support ...

  18. Total parenteral nutrition. A brief review

    Abstract. With increasingly greater use of total parenteral feeding in hospitals, knowledge of this mode of providing nutrition for the patient and the problems it presents looms as a "must" for the dietitian. This paper is only available as a PDF.

  19. Total Parenteral Nutrition in the Critically Ill Patient

    Total parenteral nutrition is used as a supplement or as the sole source of nutrition in these patients 16,17; however, previous evidence supporting this practice seems to be lacking. 18,19 Since these studies were reviewed in 1987, 19 additional randomized trials have been published. The purpose of this article is to review systematically ...

  20. Parenteral nutrition: Revisited

    Total Parenteral Nutrition became widely accepted after 1967, when Dudrick et al. from the University of Pennsylvania demonstrated that normal growth and development could occur solely with the administration of TPN. Seibert was the first to report that fever following TPN was due to pyrogens from the bacterial contamination of TPN fluids.

  21. Guidelines for Total Parenteral Nutrition

    June 26, 1972. Guidelines for Total Parenteral Nutrition. Maurice E. Shils, MD, ScD. Author Affiliations. From the departments of medicine, Memorial Hospital for Cancer and Allied Diseases and Cornell University Medical College, and the Division of Medical Research, Sloan-Kettering Institute for Cancer Research, New York. JAMA. 1972;220 (13 ...

  22. PDF Carol Rees Parrish, R.D., M.S., Series Editor The Hitchhiker's Guide to

    The guiding principle of nutrition support is to use the least invasive and most physiologic method of feeding. Infusing chemicals directly into the bloodstream is the least preferred method of providing nutrition support (1). Yet, for a select subset of the population, intra-venous infusion of central parenteral nutrition (PN) or

  23. Parenteral Nutrition (PN)

    PN requires that patients receive water, usually 25 to 40 mL/kg body weight/day. Less water is needed for patients requiring fluid restriction, energy, amino acids, lipids, vitamins, and minerals (see table Basic Adult Daily Requirements for Parenteral Nutrition). Children who need PN may have different fluid requirements and need more energy (up to 120 kcal/kg/day) and amino acids (up to 2.5 ...

  24. Total parenteral nutrition.

    Parenteral nutrition use at a university hospital. Factors associated with inappropriate use. West J Med. 1990 Jun; 152 (6):683-686. ... Starker PM, LaSala PA, Askanazi J, Todd G, Hensle TW, Kinney JM. The influence of preoperative total parenteral nutrition upon morbidity and mortality. Surg Gynecol Obstet. 1986 Jun; 162 (6):569-574.