SlideShare a Scribd company logo
1 of 106
Enteral Nutrition in
Preterm Neonates
   Dr Varsha Atul Shah
Introduction
   Proper nutrition in infancy is essential for
     – normal growth
     – immunity to infection
     – optimal neurologic and cognitive development.
   Providing adequate nutrition to preterm infants is
    challenging because of several problems, some of them
    unique to these small infants. These problems include:
     – immaturity of bowel function
     – inability to suck and swallow
     – high risk of necrotizing enterocolitis (NEC)
     – illnesses that may interfere with adequate enteral
       feeding (e.g. RDS, patent ductus arteriosus)
     – medical interventions that preclude feeding (e.g.,
       umbilical vessel catheters, exchange transfusion,
       indomethacin therapy)
Long term outcome and Barker’s
              Hypothesis
   There is evidence that adaptations in the metabolic and hormonal
    milieu in the fetal and immediate neonatal period can result in
    immediate benefit but adverse long-term outcome.
   Nutritional insults at a vulnerable period of brain development, for
    example, have been shown to be associated with effects on brain
    size, cell number, behavior and learning memory.
   The emergence of cardiovascular disease, hypertension, insulin
    resistance and obesity in low birth weight infants is another concern
    raising the issue of long-term ‘‘programming.’’
   In addition, there may be transgenerational effects as evidenced by
    the association of low maternal birth weight and higher offspring
    hypertension in adulthood.
   These changes may be exacerbated by postnatal malnutrition and
    poor growth that preterm infants experience.
PHYSIOLOGY AND
            PATHOPHYSIOLOGY
   The gut has formed and has completed its rotation back
    into the abdominal cavity by 10 weeks of gestation.
   By 16 weeks, the fetus can swallow amniotic fluid.
   GI motor activity is present before 24 weeks, but
    organized peristalsis is not established until 29-30
    weeks and is facilitated by antenatal corticosteroid
    treatment.
   Coordinated sucking and swallowing develops at 32-34
    weeks.
   By term, the fetus swallows about 150 cc/kg/day of
    amniotic fluid, which has 275 mOsm/L, contains
    carbohydrates, protein, fat, electrolytes,
    immunoglobulins and growth factors, and plays an
    important role in development of GI function. Preterm
    birth interrupts this development.
Importance of Enteral Feeding

   Even if nutrients are provided parenterally, lack of
    enteric intake leads to
    –   decreased circulating gut peptides
    –   slower enterocyte turnover and nutrient transport
    –   decreased bile acid secretion
    –   increased susceptibility to infection due to impaired barrier
        function by intestinal epithelium, lack of colonization by normal
        commensal flora and colonization by pathogenic organisms.
   For fat digestion, the newborn depends on lingual lipase,
    which is stimulated by sucking and swallowing and by
    nutrients in the stomach but not the small bowel.
How should we feed?
Trophic feedings for parenterally fed
              infants (Review)
     2008 The Cochrane Collaboration
   Trophic feeding defined as dilute or full strength feedings providing < = 25 ml/kg/d
    for > = 5d (5-10days)
   Trophic feedings vs. no feedings (10 trials): Among infants given trophic
    feedings, there was an overall reduction in days to full feeding (weighted mean
    difference [WMD] = -2.6 [95% confidence limits = -4.1, -1.0]), total days that
    feedings were held (WMD = -3.1 [-4.6, -1.6]), and total hospital stay (WMD = -11.4
    [-17.2, -5.7] compared to infants given no enteral nutrient intake.
   Tests for heterogeneity were significant in analyses of days to full enteral feedings,
    days to regain birth weight, days of phototherapy, and hospital stay.
   There was no significant difference in necrotizing enterocolitis, although the findings
    do not exclude an important effect (relative risk = 1.16 [0.75, 1.79]; risk difference =
    0.02 [-0.03, 0.06].
   Trophic feedings vs. advancing feedings (one trial): Infants given trophic
    feedings required more days to reach full enteral feeding (13.4 [8.2, 18.6]) and
    tended to have a longer hospital stay (11.0 [-1.4, 23.4]) than did infants given
    advancing feedings. With only eight total cases of necrotizing enterocolitis, trophic
    feedings were associated with a marginally significant reduction in necrotizing
    enterocolitis (relative risk =0.14 [0.02, 1.07]; risk difference = -0.09 [-0.16, -0.01].
Authors’ conclusions

   In both comparisons, the group with the greater enteral intake (trophic feedings in
    the first comparison and advancing feedings in the second comparison) required
    significantly less time to reach full feedings and had a significant or near significant
    reduction in hospital stay.
   In both comparisons, the group with the greater intake also had a higher incidence of
    NEC although the difference was not statistically significant. The concern is greatest
    for the advancing feeding regimen. Even when trophic feedings were compared to no
    feedings, the relative risk for NEC was 1.16 (0.75 - 1.79), a finding consistent with a
    16% increase in NEC and a NNH of 50. A true increase of this magnitude might
    outweigh any shorter or long-term benefits of trophic feedings. Moreover, the 95% CI
    does not exclude the possibility that trophic feedings increase NEC by as much as
    79% with a NNH of 17.
   Whether no feedings, trophic feedings, or advancing feedings should initially be used
    is difficult to discern for a variety of reasons—the inherent difficulty of assessing
    enteral feedings in high-risk infants, the limited sample size and methodologic
    limitations of most studies to date, unexplained heterogeneity with respect to a
    number of outcomes, the potential for bias to affect the findings in unblinded studies,
    and the large number of infants who must be studied to assess the effect on
    necrotizing enterocolitis.
   One or more large, well designed, multi-center trials are needed to compare these
    approaches to early feeding with respect to important clinical outcomes. A conclusive
    evaluation would assess effects on not only the survival rate without necrotizing
    enterocolitis prior to discharge from the neonatal unit but also on the survival rate
    without severe gastrointestinal or neurodevelopmental disability at >= 18 months
    age.
Nasal versus oral route for placing feeding tubes in
preterm or low birth weight infants. 2008 Cochrane
                   Collaboration
Main results
 Two small randomised trials were identified.
 Only one trial reported data on the pre-specified primary outcomes
  for this review. This trial found no evidence of effect on the time
  taken to establish enteral feeding nor the time taken to regain birth
  weight. However, the trial was underpowered (N= 46) to exclude
  modest effect sizes.

Authors’ conclusions
 There are insufficient data available to inform practice. A large
  randomised controlled trial is required to determine if the use of
  naso vs oro-enteric feeding tubes affects feeding, growth and
  development, and the incidence of adverse consequences in
  preterm or low birth weight infants.
Continuous nasogastric milk feeding versus
      intermittent bolus milk feeding for
   premature infants less than 1500 grams,
         2008 Cochrane collaboration
Main results
 Overall, the seven included trials, involving 511 infants, found no
  differences in time to achieve full enteral feeds between feeding methods
  (weighted mean difference (WMD 2 days; 95%CI -0.3, 3.9) .
 In the subgroup analysis of those studies comparing continuous nasogastric
  vs. intermittent bolus nasogastric milk feedings the findings remained
  unchanged (WMD 2 days, 95% CI -0.4, 4.1).
 There was no significant difference in somatic growth and incidence of NEC
  between feeding methods irrespective of tube placement.
 One study noted a trend toward more apneas during the study period in
  infants fed by the continuous tube feeding method compared to those fed
  by intermittent feedings delivered predominantly by orogastric tube
  placements [mean difference (MD) 14.0 apneas during study period; 95%
  CI -0.2, 28.2].
 In subgroup analysis based on weight groups, one study suggested that
  infants less than 1000 grams and 1000 - 1250 grams birth weight gained
  weight faster when fed by the continuous nasogastric tube feeding method
  compared to intermittent nasogastric tube feeding method (MD 2.0 g/day;
  95% CI 0.5, 3.5; MD 2.0 g/day; 95% CI 0.2, 3.8, respectively).
   A trend toward earlier discharge for infants less than 1000 grams birth
    weight fed by the continuous tube feeding method compared to
    intermittent nasogastric tube feeding method (MD -11 days; 95% CI -21.8,
    -0.2).

Authors’ conclusions
 Small sample sizes, methodologic limitations, inconsistencies in controlling
  variables that may affect outcomes, and conflicting results of the studies to
  date make it difficult to make universal recommendations regarding the
  best tube feeding method for premature infants less than 1500 grams.
 The clinical benefits and risks of continuous vs. intermittent nasogastric
  tube milk feeding cannot be reliably discerned from the limited information
  available from randomized trials to date.
Transpyloric versus gastric tube feeding for preterm
      infants. 2008 Cochrane Collaboration

   Data from nine trials were available.
   No evidence of an effect on short term growth rates was found:
    weight:WMD -0.7 g/week (95% confidence interval (CI) -25.2, 23.8); crown
    heel length: WMD -0.7 mm/week (95% CI -2.4, 1.0); head circumference:
    WMD 0.6 mm/week (95% CI -0.9, 2.1).
   Longer term growth was reported in one study. There were not any
    statistically significant differences between the groups in the mean body
    weight or occipitofrontal head circumference at three months or at six
    months corrected age.
   None of the included studies provided data on neurodevelopmental
    outcomes.
   Transpyloric feeding was associated with a greater incidence of gastro-
    intestinal disturbance (RR 1.45, 95% CI 1.05, 2.09). There was some
    evidence that feeding via the transpyloric route increased mortality (RR
    2.46, 95% CI 1.36, 4.46). However, the outcomes of the study that
    contributed most to this finding were likely to have been affected by
    selective allocation of the less mature and sicker infants to transpyloric
    feeding.
 No statistically significant differences in the incidence of other adverse
  events, including necrotising enterocolitis, intestinal perforation, and
  aspiration pneumonia was found.
Authors’ conclusions
 No evidence of any beneficial effect of transpyloric feeding in preterm
  infants was found. However, evidence of adverse effects was noted.
  Feeding via the transpyloric route cannot be recommended for preterm
  infants
Ad libitum or demand/semi-demand feeding versus
  scheduled interval feeding for preterm infants,
           2008 Cochrane collaboration
   Seven randomised controlled trials that compared ad libitum or demand/semi-
    demand regimes with scheduled interval regimes in preterm infants in the transition
    phase from intragastric tube to oral feeding were found.
   The trials were generally small and of variable methodological quality. The duration of
    the intervention and the duration of data collection and follow up in most of the trials
    is not likely to have allowed detection of measurable effects on growth.
   The single trial that assessed growth for longer than one week found that the rate of
    weight gain was lower in the ad libitum fed infants [mean difference -3.30 (95%
    confidence interval -6.2 to -0.4) grams per kilogram per day].
   Two trials reported that feeding preterm infants using an ad libitum or demand/semi-
    demand feeding regime allowed earlier discharge from hospital, but the other trials
    did not confirm this finding.
   Not able to undertake meta-analyses because of differences in study design and in
    the way the findings were reported.

Authors’ conclusions
 There are insufficient data at present to guide clinical practice. A large randomised
   controlled trial is needed to determine if ad libitum of demand/semi-demand feeding
   of preterm infants affects clinically important outcomes. This trial should focus on
   infants in the transition phase from intragastric tube to oral feeding and should be of
   sufficient duration to assess effects on growth and time to oral feeding and hospital
   discharge.
What should we feed?
Breast milk lah, no need
       to ask…
         Really?
Formula milk versus maternal breast milk for
     feeding preterm or low birth weight infants
            2008 Cochrane Collaboration.
Main results
 No eligible trials were identified.
Authors’ conclusions
 There are no data from randomised trials of formula milk versus
  maternal breast milk for feeding preterm or low birth weight infants.
 This may relate to a perceived difficulty of allocating an alternative
  feed to an infant whose mother wishes to feed with her own breast
  milk.
 Maternal breast milk remains the default choice of enteral nutrition
  because observational studies, and meta-analyses of trials
  comparing feeding with formula milk versus donor breast milk,
  suggest that feeding with breastmilk has major non-nutrient
  advantages for preterm or low birth weight infants.
Donor breast milk versus infant formula for preterm
infants: systematic review and meta-analysis
Cochrane collaboration 2006

Results:
•Seven studies (including five randomised controlled trials), all from the 1970s
and 1980s, fulfilled the inclusion criteria. All studies compared the effect of sole
donor breast milk with formula (combined n = 471). One of these also compared
the effect of donor breast milk with formula given as a supplement to mother’s
own milk (n = 343). No studies examined fortified donor breast milk.
•A meta-analysis based on three studies found a lower risk of NEC in infants
receiving donor breast milk compared with formula (combined RR 0.21, 95% CI
0.06 to 0.76).
•Donor breast milk was associated with slower growth in the early postnatal
period, but its long-term effect is unclear.

Conclusion:
•Donor breast milk is associated with a lower risk of NEC and slower growth in
the early postnatal period, but the quality of the evidence is limited. Further
research is needed to confirm these findings and measure the effect of fortified
or supplemented donor breast milk.
Protein supplementation of human milk for
        promoting growth in preterm infants
                            Cochrane collaboration 2000

MAIN RESULTS:
•Protein supplementation of human milk results in increases in short term
weight gain (WMD 3.6 g/kg/day, 95% CI 2.4 to 4.8 g/kg/day), linear growth
(WMD 0.28 cm/week, 95% CI 0.18 to 0.38 cm/week) and head growth (WMD
0.15 cm/week, 95% CI 0.06 to 0.23 cm/week).
•There are insufficient data to evaluate long term neurodevelopmental and
growth outcomes. There are too few infants studied to be certain that adverse
effects of protein supplementation are not increased. Blood urea levels are
increased (WMD 1.0 mmol/l, 95% CI 0.8 to 1.2 mmol/l).

REVIEWER'S CONCLUSIONS:
•Protein supplementation of human milk in relatively well preterm infants results
in increases in short term weight gain, linear and head growth. Urea levels are
increased, which may reflect adequate rather than excessive dietary protein
intake.
•Further research should be directed towards the evaluation of specific levels
of protein intake in preterm infants and the clinical effects of supplementation
with protein, including long term growth and neurodevelopmental outcomes.
This may best be done in the context of refinement of available multicomponent
fortifier preparations.
Multicomponent fortified human milk for promoting growth in
preterm infants.
Cochrane collaboration 2004
MAIN RESULTS
• Supplementation of human milk with multicomponent fortifiers (in the form of protein,
calcium, phosphate, and carbohydrate, as well as vitamins and trace minerals) is
associated with short term increases in weight gain, linear and head growth. There is no
effect on serum alkaline phosphatase levels; it is not clear if there is an effect on bone
mineral content. Nitrogen retention and blood urea levels appear to be increased.
•There are insufficient data to evaluate long term neurodevelopmental and growth
outcomes, although there appears to be no effect on growth beyond one year of life.
•Use of multicomponent fortifiers does not appear to be associated with adverse effects,
although the total number of infants studied and the large amount of missing data
reduces confidence in this conclusion. Blood urea levels are increased and blood pH
levels minimally decreased, but the clinical significance of this is uncertain.

REVIEWER'S CONCLUSIONS:
•Multicomponent fortification of human milk is associated with short-term improvements in
weight gain, linear and head growth. Despite the absence of evidence of long-term
benefit and insufficient evidence to be reassured that there are no deleterious effects, it is
unlikely that further studies evaluating fortification of human milk versus no
supplementation will be performed.
•Further research should be directed toward comparisons between different proprietary
preparations and evaluating both short-term and long-term outcomes in search of the
"optimal" composition of fortifiers.
Fortification of Preterm Human Milk for Feeding
       Preterm Infants – Yale Pediatric Protocol
   Preterm human milk (PTHM) is milk expressed by a mother for her infant following a preterm
    delivery.
     –   may be stored in the Breast Milk Refrigerator for up to 48 hours.
     –   If the milk will not be fed to the infant within that time, it should be frozen as early as possible after
         expression in appropriate-sized volumes and then gently thawed prior to use. Once thawed, it should be
         used within about 4 hours.
   Infants should be fed full strength human milk as their initial enteral feeding.
   When to fortify:
     –   After an infant tolerates full enteral feeding of full-strength human milk for 5-7 days
     –   Or, after he/she tolerates about 75% his/her total daily volume (IV + NG) as full strength human milk
   How to fortify
     –   adding a powdered Human Milk Fortifier
     –   by mixing human milk with an equal volume of Premature Formula
              Mixing equal volumes of human milk and Premature Formula or Natural Care results in a nutrient concentration that equals
               an average between human milk and formula
     –   Or, by mixing human milk with an equal volume of Natural Care(similac) Human Milk Fortifier.
   The decision on which method of fortification to use will depend upon the mother's milk
    production with discussion with Lactation Consultant.
   Why fortify
     –   Increases the content of nutrients in the infant's diet, esp calcium and phosphorus. (See table next page)
     –   The composition of PTHM varies with the duration of lactation; an approximate composition of "mature"
         PTHM is also shown
Special Formula Use
                            Recommendations
Nutrition Practice Care Guidelines for Preterm Infants In the Community. Revised August 2006.
Developed by Child Development and Rehabilitation Center, Nutrition Services, Oregon Department of Human Services, Nutrition & Health
Screening – WIC Program. Oregon Pediatric Nutrition Practice Group



    Preterm formula and Human Milk Fortifier: generally for infants
     weighing less than 1,850 to 2,000 gm (about 4 to 4 ½ lbs). It is
     inappropriate for most infants who weigh more than 2.5 kg (5 ½ lbs) or are
     taking in over 500 ml daily to be fed premature formulas (Enfamil
     Premature Lipil or Similac Special Care) or Human Milk Fortifiers (Similac
     HMF, Similac Natural Care or Enfamil HMF) for all their feeds because of the
     higher vitamin A and vitamin D content of these formulas and the possible
     risk of hypervitaminosis.
   Transitional Formulas: (also called “post-discharge premature formulas”)
     Formulas such as Enfamil EnfaCare Lipil and Similac Neosure Advance
     provide 22 kcal/oz and have higher levels of protein, calcium, phosphorus,
     vitamins and other minerals than standard infant formulas. Research has
     shown that premature infants fed these formulas have improved growth
     and bone mineralization compared to those fed standard infant formulas.
  *See recommendation below for length of time an infant needs to remain on a
     transitional formula.
 Specialized formulas
   – These include Pregestimil, Alimentum, Nutramigen, Neocate and
      Elecare, and may be indicated based on feeding intolerance.
   – Enfamil AR is not indicated for preterm infants due to the risk of the
      formation of lactobezoars (hard clumps of undigested milk curds).
      These are “term” formulas and thus have less calcium, phosphorus, and
      protein than transitional formulas. If preterm infants are given these
      formulas, they should be followed more closely by an RD and the
      appropriate labs should be checked.
 Soy-based formulas are not recommended for preterm infants. Preterm
  infants receiving soy formula have suboptimal carbohydrate and mineral
  absorption and utilization than cow’s milk-based formula. AAP doesn’t
  recommend soy formula for infants born < 1800 g since preterm infants
  showed significantly less weight gain, less linear growth, and lower serum
  albumin levels than those infants receiving cow’s milk-based formulas.
  Studies also have shown lower levels of bone marker formation in the
  premature population which can lead to osteopenia.
 Goat’s milk is not recommended for preterm infants. Goat’s milk is
  deficient in folic acid and vitamin B6. It is also higher in protein than human
  milk and infant formula which puts the premature infant at risk for
  dehydration due to the higher renal solute load.
Dorset County Hospital Neonatal Unit
  Guidelines for enteral feeding (Mar05)
   Milk of choice, in order of preference, is:
    –   1. Breast milk – suckled by baby from the breast
    –   2. Mother’s expressed breast milk (EBM), fresh
    –   3. Mother’s EBM, frozen
    –   4. Donor EBM*
    –   5. Formula feed appropriate to gestational age and birth weight
 * consider using when:
        baby deemed at high risk of developing necrotising
          enterocolitis (NEC) – see section 3
        if it is available, to intensive care babies
        poor tolerance of formula feeds
 NB parental consent should be sought prior to a baby being given
  donor EBM
 This hospital actively promotes the benefits of breast feeding for all
  infants. There are particular benefits of breast milk for the preterm
  infant and sick neonate (eg reduced incidence of necrotising
  enterocolitis - ref 1).
Dorset County (cont’d)
                Early nutrition in the preterm group

 All mothers should be encouraged to produce milk for their baby as soon
  as possible after delivery if their baby is not able to feed naturally. Even if
  the mother is not planning to breast feed in the longer term or unwilling to
  breast feed, they should be advised why EBM is the best nutritional support
  for their baby.
 For premature infants whose mother’s wish to breast feed in the longer
  term, skin to skin contact should be strongly encouraged from an early
  stage. Cup feeding is often possible from 30 weeks gestation. Bottle feeding
  should be avoided if at all possible given the different technique a baby
  uses to bottle feed compared to feeding at the breast.
 If formula feeds are being used, the baby should not be started on a
  preterm formula until they are tolerating 150 ml/kg/day of a standard
  formula feed.
Dorset County (cont’d)
                  Early nutrition in the preterm group
a) Under 2kg
 For babies with a birth weight of less than 1.2 kg (and occasionally larger
   babies) parenteral nutrition will normally be necessary during the first 1-2
   weeks of life. This should be written up as soon as possible after delivery
   so that it can be commenced within 1-2 days of delivery (see TPN
   guidelines).

   i) Non-nutritive feeding (minimal enteral nutrition):
     – Small amounts of breast milk started in the first few days of life (day 1 if
       possible)
     – encourage gut maturity (ref 2)
     – Start at 1ml every 4-6 hours if <1kg (ref 5) and 1ml/hr if >1kg (ref 3). Maternal
       EBM is preferable to donor breast milk.
     – Please note section on management of infants at high risk of NEC
       (section 3)

   ii) Rate of increase of feed:
     – Once tolerating minimal enteral nutrition, increase the volume given at each
       feed. The rate of increase should be decided for each individual baby but would
       normally be less than 35ml/kg per 24 hours for babies less than 1200gm (ref 4).
       Larger babies will possibly tolerate faster increments in feed volume.
Dorset County (cont’d)
                Early nutrition in the preterm group
  iii) Final feed volume:
     – Increase feed volumes up to a maximum of:
     – 200-220 ml/kg/day for EBM
     – 180 ml/kg for term formula (or Nutriprem 2)
     – 160 ml/kg/day for preterm formula*
 Once an infant is tolerating >50% of their total fluid volume enterally, lipid
   can be discontinued in the TPN (see TPN guidelines).
 * If formula feeds are being used, the baby should not be started on a
   preterm formula until they are tolerating 150 ml/kg/day of a standard
   formula feed.
b) greater than 2kg
 Where possible allow to demand feed.
 For well infants below 37 weeks gestation, consider starting feeds at
   60ml/kg/day on day 1 of feeding aiming to reach 150ml/kg/day within 4
   days depending on progress.
Dorset County (cont’d)
                  Early nutrition in the preterm group
3. High risk groups for NEC
 Particular high risk groups include:
    –   Preterm infants < 1kg
    –   Severe intrauterine growth retardation
    –   Absent/reversed end diastolic flow on umbilical artery Doppler
    –   Sepsis
 For infants with absent or reversed end diastolic flow do not feed enterally
  for 3 days.
 For at risk groups consider waiting for maternal EBM before starting feeds.
  In these circumstances feeding human milk at a rate 1 ml every 4-6 hours
  is recommended (ref 5). If maternal EBM is not available for these groups
  consider the use of donor EBM.
 For babies at high risk of necrotising enterocolitis (NEC) there is some
  evidence that a rate of increase not exceeding 20ml/kg per 24 hours
  reduces the incidence of NEC (ref 6).
 * After a suspected episode of NEC feeds should be reintroduced using EBM
  (maternal if possible but donor if not available). If maternal EBM not
  available and mother refused permission to use donor EBM, consider using
  Pregestimil (see section 9 for advise on increasing calorie intake if
  inadequate growth on this formula).
References:
 1. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis.
  Lancet 1990; 336: 1519-1523.
 2. McClure RJ and Newell SJ. Randomised controlled trial of trophic
  feedingand gut motility. Arch Dis Child Neonatal Ed 1999; 80: F54-58.
 3. Child Nutrition Panel “Feeding the preterm infant”. Z740127 May 2002
 4. Kennedy KA et al. Rapid versus slow rate of advancement of feeding in
  parenterally fed low birth weight infants. Cochrane Review. 2001.
 5. King C. Neonatal Unit Enteral Feeding Policy. Hammersmith Hospital NHS
  Trust 2000.
 6. Anderson DM and Kliegman RM. The relationship of neonatal
  alimentation practice to the occurrence of endemic necrotising enterocolitis.
  Am J Perinatol 1991; 8: 62-67.
 7. Cooke RJ and Embleton ND. Feeding issues in preterm infants. J. Pediat.
  Neonatal Ed 2000; 83: F215-218.
 8. Nutrition in low birth weight infants. Clinical Paediatric Dietetics 2001.
Enteral Feeding
guidelines for the
preterm infant
within the neonatal
service of
the Leeds
Teaching Hospitals
NHS Trust
GROUP 1. 34-36 weeks gestational age and birthweight > 1.5Kg who is on the
  neonatal unit due to a lack of transitional care capacity.
 Timing of enteral: Immediately
 Mode of enteral: NG or oral
 Type of milk: EBM or Nutriprem (as per policy)
 Rate of increase: Full feeds as soon as able
GROUP 2. 34-36 weeks gestational age and birthweight > 1.5Kg admitted to
  Neonatal Unit for whatever reason
 Feeding will depend on primary pathology. If considered ready for feeding, then feed
  as Feeding Group 1.
GROUP 3. 34-36 weeks gestational age and birthweight < 1.5Kg
 These infants are under the 2nd centile.
 Timing of enteral: Delay for up to 24 hours in marked IUGR, or make decision to feed
  immediately
 Mode of enteral: NG or oral
 Type of milk: EBM or Nutriprem 1 (as per policy)
 Rate of increase: 60mls/kg increasing at a maximum of 1ml/Kg per day
 (0.5ml 12 hourly) up to total of 150/ml/kg/day
GROUP 4. 31-33 weeks, NO IUGR or A/REDF on antenatal Doppler
 Timing of enteral: Immediately
 Mode of enteral: NG
 Type of milk: EBM or Nutriprem 1(as per policy)
 Rate of increase: 60mls/kg, max increase per day < 25ml/kg
 (0.5ml/h, 8 hourly) as tolerated*
GROUP 5. <= 30 weeks, No IUGR (> 2nd centile) or A/REDF on antenatal
  Doppler
 Timing of enteral: Delay for 24 hours after birth
 Mode of enteral: NG
 Type of milk: EBM or Nutriprem 1(as per policy)
 Rate of increase: 1ml/kg, increasing maximum 8 hourly as tolerated* for first 72
  hours. Increase to 0.5ml/kg 6 hourly maximum as tolerated* after 72 hours of age.
 Note: Slow feeding to 12 hourly if evidence of failure to tolerate*.
 Maximum daily increase not to exceed 25 mls/kg per day.
GROUP 6. <= 33 weeks, IUGR (<= 2nd centile) or A/REDF on antenatal
  Doppler
 Timing of enteral: Delay at least 24 hours
 Mode of enteral: NG
 Type of Milk: EBM or Donor EBM (see below)
 Rate of increase:
    –   <= 1kg: 0.5ml/hr initially increasing by 0.5 ml/24 hours as tolerated
    –   > 1kg 1ml/hr initially increasing by 1ml/24 hours as tolerated
  When full feeds established continue, if using Donor EBM, continue on full feeds
   (maximum 165ml/Kg per day) for at least 5 days. Switch to Nutriprem 1 after this
   time.
 Parenteral Nutrition (PN) will be required for this group (see PN protocol)
*Tolerating enteral feeds
 Babies are considered to be tolerating enteral feeds if
 • 4 hourly NG aspirates are < 25% of total infused in the preceding 4 hours
 • No significant abdominal distension
 • No significant vomiting
 • No bile-stained aspirates
What about
supplements/adjuncts?
Longchain polyunsaturated fatty acid supplementation in preterm infants
 Cochrane collaboration 2003

Main results
•Of the eleven randomised trials included in the review, two of these were not
classified as of high quality despite blinded assessment and complete follow-up, due to
problems with assessment methodology.

Visual acuity
Visual acuity over the first year was measured by Teller acuity cards in six studies, by
VEP in four studies and by ERG in two studies. Most studies found no significant
differences in any visual assessment between supplemented and control infants.

Development
Most of the trials have used Bayley Scales of Infant Development (BSID) at 12 to 24
months postterm and shown no significant effect following supplementation. Meta-
analysis of BSID of three studies (Fewtrell 2002, O'Connor 2001, van Wezel 2002)
shows no significant effect of supplementation on development. Carlson 1993 and
Carlson 1996 demonstrated lower novelty preferences (possibly predictive of lower
intelligence) in the supplemented compared with the control group. The investigators
however concluded that supplemented infants may have more rapid visual information
processing given that they had more looks and each look was of shorter duration.
Growth
Most trials have reported no significant effect of LCPUFA supplementation on growth
of preterm infants. Two trials (Carlson 1993, Carlson 1996) suggest that LCPUFA
supplemented infants grow less well than controls, possibly due to a reduction in AA
levels which occurs when n-3 supplements are used without n-6 supplements. Recent
trials with addition of AA to the supplement have reported no significant effect on
growth. Fewtrell 2002 reported mild reductions in length and weight z scores at 18
months.
Contrary to these results, the meta-analysis of five studies (Uauy 1992, Carlson 1996,
Hansen 1997, Vanderhoof 1999, Innis 2002) showed increased weight and length at
two months post-term in supplemented infants.

Side effects
Uauy 1992 reported no significant effect of LCPUFA supplementation on bleeding time
and red cell membrane fragility.

Reviewers' conclusions
Infants enrolled in the trials were relatively mature and healthy preterm infants.
Assessment schedule and methodology, dose and source of supplementation and fatty
acid composition of the control formula varied between trials. No long-term benefits
were demonstrated for infants receiving formula supplemented with LCPUFA. There
was no evidence that supplementation of formula with n-3 and n-6 LCPUFA impaired
the growth of preterm infants.
Is supplementary iron useful when preterm infants
     are treated with erythropoietin? BestBets 2006
   In [a preterm infant who is receiving rHuEPO therapy] does [iron supplementation]
    reduce the requirement for [blood transfusion]? If so, what method of administration
    and dose reduces it most successfully
Comment(s)

•Anaemia in premature infants is a common problem. Although erythropoietin is
not used widely in neonatal practice, there is evidence of its efficacy in reducing
the need for transfusion in preterm infants,(Shannon) especially if they are not
extremely small or sick.(Soubasi). It is regularly used in situations where blood
transfusion is unacceptable.

•Iron supplementation has been a standard in neonatal care for preterm infants
for many years and helps to reduce late anaemia (Franz) if given with vitamins,
especially vitamin E.(Jansson). However, when stimulating erythropoiesis with
rHuEpo to reduce the need for transfusion, iron availability becomes critical.

• Several studies have investigated rHuEpo efficacy in preterm infants and most
of them have used supplementary iron in either the oral or parenteral routes. The
literature on the use of rHuEpo and iron mostly consists of studies on dose
variation of rHuEpo rather than variation in the iron supplementation.

•Our search yielded seven studies, but one(Pollak) was excluded owing to poor
methodological quality. Two studies by Carnielli et al and Fujiu et al compared
rHuEpo and oral iron supplementation with rHuEpo alone; however, the
interpretation of the data is difficult because Carnielli et al reported their results
only as mean values and logarithms, making statistical analysis difficult. Fujiu et
al found that no infants in either arm of their study required a blood transfusion.
•Although this may suggest that there was no difference between the groups, the
sample size was small, with only 24 infants in total, and the clinical equivalence could
not be shown. In addition, the authors state that losses due to phlebotomy in their
study were lower than those in other similar studies. This may be relevant, as one of
the most common causes of the anaemia of prematurity is iatrogenic blood loss.

•Kivivuori et al and Meyer et al compared rHuEPO treatment and parenteral iron
supplementation with rHuEPO treatment and oral iron supplementation. Combined
data from the two studies showed that there was no significant difference between the
groups for the number of blood transfusions given (odds ratio (OR) 1.65, 95%
confidence interval (CI) 0.41 to 6.64). This seems to suggest that oral iron
supplementation is at least sufficient. However, one study used intravenous iron
supplementation whereas the other used intramuscular iron supplementation.
Differences in absorption of these two different routes may be relevant, but no study
has been carried out comparing intravenous with intramuscular iron supplementation.

•Bader et al and Nazir et al compared infants receiving rHuEPO treatment and high-
dose oral iron supplementation with those receiving rHuEPO treatment and low-dose
oral iron supplementation. There was no significant difference between the two groups,
when combining data for the two studies, regarding the number of blood transfusions
(OR 0.46, 95% CI 0.04 to 5.75).
   Preterm infants on special-care baby units frequently become anaemic and require
    top-up blood transfusions. Preterm infants often have low iron stores; this becomes
    more evident if they do not receive transfusions or iron supplementation.
    Erythropoietin is used occasionally to stimulate red cell production and prevent
    anaemia, and the increased erythropoiesis that occurs as a result of rHuEPO
    treatment will deplete iron stores.

   The studies currently available do not give an adequate answer to the question as to
    which is the best mode and dose of iron supplementation with rHuEpo treatment. On
    the basis of the principle of using the lowest effective dose and the least invasive
    mode of administration, at present, low-dose oral iron would seem appropriate for
    supplementation when rHuEpo is used in preterm infants.

Clinical Bottom Line
 Evidence available to strongly support any specific recommendation for iron
    supplementation with recombinant erythropoietin treatment in premature infants
    (grade D) is insufficient. Low-dose oral iron supplementation is not inferior to other
    treatment regimens (grade B).
Glutamine supplementation to prevent morbidity
        and mortality in preterm infants
                          Cochrane collaboration 2008
Main results
•2365 preterm infants have participated in seven randomised controlled trials. All of
the participating infants were of very low birth weight.
•Three trials assessed enteral glutamine supplementation and four trials assessed
parenteral glutamine supplementation.
•The trials were generally of good methodological quality with adequate allocation
concealment, blinding of caregivers and assessors to the intervention, and complete
or near-complete follow-up of recruited infants.
•Glutamine supplementation does not have a statistically significant effect on
mortality: typical relative risk 0.98 (95% confidence interval 0.80 to 1.20); typical risk
difference 0.00 (95% confidence interval -0.03 to 0.02).
•The only trial that assessed long-term outcomes did not find any statistically
significant differences in various assessments of neurodevelopment at 18 months
corrected age. Glutamine supplementation does not have a statistically significant
effect on other neonatal morbidities including invasive infection, necrotising
enterocolitis, time to achieve full enteral nutrition, or duration of hospital stay.
Authors' conclusions
•The available data from good quality randomised controlled trials indicate that
glutamine supplementation does not confer benefits for preterm infants. The narrow
confidence intervals for the effect size estimates suggest that a further trial of this
intervention is not a research priority.
Iodine supplementation for the prevention
           of mortality and adverse
  neurodevelopmental outcomes in preterm
               infants (Review)
Main results Copyright © 2008 The Cochrane Collaboration
 We found only one randomised controlled trial (N = 121) that fulfilled the review
   eligibility criteria (Rogahn 2000).
 The participants were infants born before 33 weeks’ gestation (but most were of birth
   weight greater than 1000 grams). The primary aim of this trial was to assess the
   effect of iodine supplementation on thyroid function. The investigators did not detect
   any statistically significant effects on the plasma levels of thyroxine (free and total),
   triiodothyronine, or thyrotrophin in preterm infants (measured up to 40 weeks’ post-
   conceptional age). Only one infant died and the trial was therefore underpowered to
   detect an effect on mortality. The trial did not assess the effect of the intervention on
   neurodevelopmental morbidity. There was not a statistically significant difference in
   the incidence of chronic lung disease.
Authors’ conclusions
 There are insufficient data at present to determine whether providing preterminfants
   with supplemental iodine (to match fetal accretion rates) prevents morbidity and
   mortality in preterm infants.
 Future randomised controlled trials of iodine supplementation should focus on
   extremely preterm and extremely low birth weight infants, the group at greatest risk
   of transient hypothyroxinaemia. These trials should aim to assess the effect of iodine
   supplementation on clinically important outcomes including respiratory morbidity and
   longer term neurodevelopment.
Lactase treated feeds to promote growth and
      feeding tolerance in preterm infants (Review)
                   Copyright © 2008 The Cochrane Collaboration

Main results
 One study enrolling 130 infants of 26 - 34 weeks postconceptual age (mean postnatal
   age at entry 11 days) was identified and no identified study was excluded.
 The study was a double blind randomized controlled trial of high quality. Lactase
   treated feeds were initiated when enteral feedings provided > 75% of daily intake.
   None of the primary outcomes outlined in the protocol for this review and only one of
   the secondary outcomes, necrotizing enterocolitis (NEC), were reported on. The RR
   for NEC was 0.32 (95% CI
 0.32 (0.01, 7.79); the RD was -0.02 (95% CI -0.06, 0.03) (a reduction which was not
   statistically significant). There was a statistically significant increase in weight gain at
   study day 10 in the lactase treated feeds group but not at any other time points.
   Overall, there was not a statistically significant effect on weight gain. No adverse
   effects were noted.
Authors’ conclusions
 The only randomized trial to date provides no evidence of significant benefit to
   preterm infants from adding lactase to their feeds.
 Further research regarding effectiveness and safety are required before practice
   recommendations can be made. Randomized controlled trials comparing lactase vs
   placebo treated feeds and enrolling infants when enteral feeds are introduced are
   recommended. The primary and secondary outcomes for effectiveness and safety
   should include those identified in this review.
Effect of taurine supplementation on growth and development
        in preterm or low birth weight infants (Review)
                   Copyright © 2008 The Cochrane Collaboration
Main results
 Nine small trials were identified.
 In total, 189 infants participated. Most participants were greater than 30 weeks
   gestational age at birth and were clinically stable. In eight of the studies, taurine was
   given enterally with formula milk. Only one small trial assessed parenteral taurine
   supplementation.
 Taurine supplementation increased intestinal fat absorption [weighted mean
   difference 4.0 (95% confidence interval 1.4, 6.6) percent of intake]. However, meta-
   analyses did not reveal any statistically significant effects on growth parameters
   assessed during the neonatal period or until three to four months chronological age
   [rate of weight gain: weighted mean difference - 0.25 (95% confidence interval
   -1.16, 0.66) grams/kilogram/day; change in length: weighted mean difference 0.37
   (95% confidence interval -0.23, 0.98) millimetres/week; change in head
   circumference: weighted mean difference 0.15 (95% confidence interval -0.19, 0.50)
   millimeters/week].
 There are very limited data on the effect on neonatal mortality or morbidities, and no
   data on long-term growth or neurological outcomes.
Authors’ conclusions
 Despite that lack of evidence of benefit from randomised controlled trials, it is likely
   that taurine will continue to be added to formula milks and parenteral nutrition
   solutions used for feeding preterm and low birth weight infants given the putative
   association of taurine deficiency with various adverse outcomes.
 Further randomised controlled trials of taurine supplementation versus no
   supplementation in preterm or low birth weight infants are unlikely to be viewed as a
   research priority, but there may be issues related to dose or duration of
   supplementation in specific subgroups of infants that merit further research.
Probiotics for prevention of necrotizing
      enterocolitis in preterm infants
                        Cochrane review 2007

Main results
 Nine eligible trials randomizing 1425 infants were included.
 Included trials were highly variable with regard to enrollment criteria (i.e. birth weight
   and gestational age), baseline risk of NEC in the control groups, timing, dose,
   formulation of the probiotics, and feeding regimens. Data regarding extremely low
   birth weight infants (ELBW) could not be extrapolated.
 In a meta-analysis of trial data, enteral probiotics supplementation significantly
   reduced the incidence of severe NEC (stage II or more) [typical RR 0.32 (95% CI
   0.17, 0.60)] and mortality [typical RR 0.43 (95% CI 0.25, 0.75]. There was no
   evidence of significant reduction of nosocomial sepsis [typical RR 0.93 (95% CI 0.73,
   1.19)] or days on total parenteral nutrition (TPN) [WMD -1.9 (95% CI -4.6, 0.77)].
 The included trials reported no systemic infection with the probiotics supplemental
   organism. The statistical test of heterogeneity for NEC, mortality and sepsis was
   insignificant.
Reviewers' conclusions
 Enteral supplementation of probiotics reduced the risk of severe NEC and mortality in
   preterm infants. This analysis supports a change in practice in premature infants >
   1000 g at birth. Data regarding outcome of ELBW infants could not be extracted from
   the available studies; therefore, a reliable estimate of the safety and efficacy of
   administration of probiotic supplements cannot be made in this high risk group. A
   large randomized controlled trial is required to investigate the potential benefits and
   safety profile of probiotics supplementation in ELBW infants.
Erythromycin for the prevention and treatment of
   feeding intolerance in preterm infants (Review)
                  Copyright © 2008 The Cochrane Collaboration

Main results
 Ten randomized controlled studies (three prevention and seven treatment studies)
   were included.
 Studies varied greatly in the definition of feeding intolerance and how outcomes were
   measured, analyzed and reported, so meta-analysis of most outcomes was
   impossible. It was observed, however, that the studies using erythromycin at higher
   treatment doses (40 to 50 mg/kg/day) or in infants > 32 weeks’ GA reported more
   positive effects in improving feeding intolerance.
 Meta-analysis of high dose prevention studies showed no significant difference in NEC
   (typical RR 0.59, 95% CI 0.11, 3.01; typical RD-0.021, 95%CI -0.087, 0.045). Meta-
   analysis of high dose treatment studies showed no significant difference in septicemia
   (typical RR 0.83, 95% CI 0.47, 1.45; typical RD -0.04, 95% CI -0.17, 0.08).
Authors’ conclusions
 There is insufficient evidence to recommend the use of erythromycin in low or high
   doses for preterm infants with or at risk of feeding intolerance.
 Future research is needed to determine if there is a more precise dose range where
   erythromycin might be effective as a prokinetic agent in preterm infants > 32 weeks’
   GA.
The Role of Nutrition in
  the Prevention and
    Management of
  BPDSeminars in Perinatology
          30:200-208
Nutritional Intake
   Preterm infants with BPD are at risk for undernutrition due to difficulties maintaining appropriate
    nutritional intake
     – as a result of co-morbidities (Carlson, 2004) eg swallowing dysfunction, fatigue during
          feeding, GERD, dysfunction of other organ system eg NEC, prolonged TPN
     – medically indicated fluid restriction (Carlson, 2004; Oh et al., 2005),
     – and elevated energy expenditure (Bauer et al., 2003; Leitch & Denne, 2000).
     – Use of dexamethasone
     – Anemia of prematurity
     – Medications eg methyl xanthines and beta sympthomimetics can increase energy
          consumption
   BPD patients can have increased energy expenditure up to 25% above total caloric needs
     – Increased work of breathing
     – Higher resting metabolic rates
Enteral Nutrition
Calories
 Breast milk requires HMF, multivitamin and iron supplements
 Alternatively use prem formula
     – Provides additional calories, proteins, calcium, phosphorus and vitamins
   Additional fat may be desirable
     – Increases calories
     – Less CO2 production than carbohydrates
     – However, additional fat slows gastric emptying and may worsen GERD
   Glucose polymers
     – Provide additional 4kcal/g
     – May cause hyperosmolar diarrhoea
   Protein supplementation can be considered or provided by using a
    high caloric formula eg 30 kcal/oz
Calcium, Phosphorus and Vit D
   Osteopenia of prem more common in BPD
    – Limited solubility of Ca and phosphorus in
      TPN
    – Low enteral intake
    – Loop diuretics/urinary loss
   Provide above via prem formula or
    fortified BM
Nutrients
   Vit A
    – Deficiency associated with impaired clearance of lung
      secretions, impaired water homeostasis across
      tracheobronchial epithelium, loss of cilia, diminished
      lung injury repair and lack of airway distensability
    – Well documented to result in BPD/death in Vit A
      deficiency (NICHD study).
    – Dose of IM 5000 IU per dose 3x/week (12 doses) for
      ELBW. Oral does not work.
   Under study: inositol, Vit E
Discharge in BPD
   67% continue to have growth failure post
    discharge
   Individualised nutrition plan
   Continue 22kcal/oz prem formula for 6-8 months
   Low salt/low volume/high calorie diet may be
    necessary
   Achieve solid intake slower than chronological
    age, often tolerate spoon feeding (thicker
    consistency) better than nipple (liquid). This may
    begin at 3-4 mths post gestation
Discharge – planning
     and post
Non-nutritive sucking for promoting physiologic
   stability andnutrition in preterm infants (Review)
                    Copyright © 2008 The Cochrane Collaboration
Types of intervention
 Non-nutritive sucking involving the use of a pacifier. The intervention occurred
   before, during or after feeding by a naso/orogastric tube; before or after bottle
   feeding; or outside of feeding times.

Main results
 This review consisted of 21 studies, 15 of which were randomized controlled trials.
 NNS was found to decrease significantly the length of hospital stay in preterm infants.
   The review did not reveal a consistent benefit of NNS with respect to other major
   clinical variables (weight gain, energy intake, heart rate, oxygen saturation, intestinal
   transit time, age at full oral feeds and behavioral state).
 The review identified other positive clinical outcomes of NNS: transition from tube to
   bottle feeds and better bottle feeding performance.
 No negative outcomes were reported in any of the studies.


Authors’ conclusions
 There were also a number of limitations of the presently available evidence related to
   the design of the studies, outcome variability, and lack of long-term data.
 Based on the available evidence, NNS in preterm infants would appear to have some
   clinical benefit. It does not appear to have any short-term negative effects.
 In view of the fact that there are no long-term data, further investigations are
   recommended. In order to facilitate meta-analysis of these data, future research in
   this area should involve outcome measures consistent with those used in previous
   studies. In addition, published reports should include all relevant data.
Early discharge with home support of gavage
   feeding for stable preterm infants who have not
       established full oral feeds 2008 Cochrane
                     Collaboration
Main results
 Data from one quasi-randomised trial with 88 infants from 75 families were included
   in the review.
 Infants in the early discharge program with home gavage feeding had a mean
   hospital stay that was 9.3 days shorter [MD -9.3 (-18.49 to -0.11)] than infants in the
   control group. Infants in the early discharge program also had a lower risk of clinical
   infection during the home gavage period compared with the corresponding time in
   hospital for the control group [relative risk 0.35 (0.17 to 0.69)].
 There were no significant differences between groups in duration and extent of
   breast feeding, weight gain, re-admission within the first 12 months post discharge
   from the home gavage program or from hospital, scores reflecting parental
   satisfaction, or health service use.
Authors’ conclusions
 Experimental evidence to evaluate the benefits and risks in preterm infants of early
   discharge from hospital with home gavage feeding compared with later discharge
   upon attainment of full sucking feeds is limited to the results of one small quasi-
   randomised controlled trial.
 High quality trials with concealed allocation, complete follow-up of all randomised
   infants and adequate sample size are needed before practice recommendations can
   be made.
Use of transitional formula: the
                evidence
         Post discharge Nutrition of Preterm Infants, Journal of Perinatology 2005; 25:S15–S16
   feeding of specially designed post discharge formulas has
    demonstrated better weight (at 9 but not at 18 months) and length
    (at 9 and 18 months) as well as higher bone mineral content when
    compared to term infant formula
     –    Lucas A, King F, Bishop NB. Postdischarge formula consumption in infants born preterm. Arch Dis Child 1992;67:691–2.
     –    Lucas A, Fewtrell MS, Morley R, Singhal A, Abbott RA, Isaacs E. Randomized trial of nutrient-enriched formula versus standard
          formula for post discharge preterm infants. Pediatrics 2001;107:683–9.
   the benefit appears restricted to males, a finding similar to that of
    Cooke et al.
     –    Cooke RJ, Griffin IJ, McCormick K, Wells JC, Smith JS, Robinson SJ. Feeding preterm infants after hospital discharge: effect of dietary
          manipulation on nutrient intake and growth. Pediatr Res 1998;43:355–60

 From the NICHD network, data demonstrate that in virtually all
  infants less than 1500 g, appropriate-for-gestational-age infants are
  ‘‘converted’’ to small for-gestational-age infants by 36 weeks
  postmenstrual age.
 Some recovery in these parameters has been reported
 Chronic lung disease adds an additional burden as demonstrated by
  73% of infants in one study experiencing a decrease in weight z
  score between hospital discharge and 7 months
Use of transitional formula:
                  Guidelines
     Post discharge Nutrition of Preterm Infants, Journal of Perinatology 2005; 25:S15–S16


Until more optimal strategies are developed, striving
  toward achieving the best possible gain without
  adverse effects may be appropriate.
Guideline:
 <1800 g: 24 kcal/oz preterm infant formula
 Transition to 22 kcal/oz at >1800 g if all growth
  parameters are 25th percentile or greater and infant
  is gaining 15 to 40 g/day
 Transition from 22 kcal/oz to 20 kcal/oz term formula
  at 4–6 months corrected gestational age if all growth
  parameters are above 25th percentile
Nutrition Practice Care
Guidelines for Preterm Infants
 In the Community (Revised
        August 2006)
      Developed by: Child Development and
     Rehabilitation Center, Nutrition Services
  Oregon Department of Human Services, Nutrition
  & Health Screening – WIC Program Oregon
     Pediatric Nutrition Practice Group
Referral Criteria


These “red flags” should alert the community nutritionist of the need for
further assessment, referral and follow-up

Anthropometric “Red Flags”:
• Weight loss or significant decline in percentile ranking (“falling away”
from expected growth curve percentile)
• Poor rate of weight gain for corrected age as listed below:

Age                Weight Gain
term – 3 mos       < 20 gm/day (< 5 oz/wk)
3 – 6 mos          < 15 gm/day (< 3½ oz/wk)
6 – 9 mos          < 10 gm/day (< 2 oz/wk)
9 – 12 mos         < 6 gm/day (< 1½ oz/wk)
1 – 2 yrs          < 1 kg or < 2 lbs in 6 mos
2 – 5 yrs          < 0.7 kg or < 1½ lbs in 6 mos
VITAMIN-MINERAL SUPPLEMENTATION


 Vitamins: Supplementation with a standard infant multivitamin (with
  vitamins A, D, B1, B2, B3, B6, B12, C, and iron) is generally needed initially
  after NICU discharge to meet the preterm infant’s vitamin needs, until the
  infant is consuming larger volumes of feeds.
 Vitamin D: Per the American Academy of Pediatrics (AAP), all infants fed
  unfortified breast milk should continue to receive a supplement of 200 IU of
  Vitamin D for the first year.
     – This 200 IU of Vitamin D can be provided by continuing the 0.5 ml daily of the
       standard infant multivitamin, or by changing to 0.5 ml daily of a tri-vitamin
       supplement (vitamins A, C, and D). Infants receiving 17 oz. (500 ml) or more of
       a vitamin D-fortified infant formula do not need any additional Vitamin D
       supplementation.
   Iron: Preterm infants have lower iron stores than term infants. By 2
    months post birth (not 2 months corrected age), preterm infants should
    have an intake of 2-4 mg iron/kg/day (up to a maximum of 40 mg/day)
    from an iron-fortified infant formula and/or supplement. This iron dose
    should be continued for the first year of life.
Osteopenia of prematurity is most commonly seen in:
    • Very-low-birth-weight infants (BW < 1500g).
    • Any IUGR infant with a birthweight < 1800 g regardless of gestational age.
    • Infants with chronic lung disease/bronchopulmonary dysplasia.
    • Infants requiring long-term parenteral nutrition at birth.
    • Infants on certain medications, including diuretics & corticosteroids, that affect
    mineral absorption.
    • Infants starting feedings of unfortified breastmilk or standard formula too early, or
    soy formula.

Recommendation for checking labs:
    • 1-month post-discharge for the infants born < 1500g and IUGR with birthweight <
    1800 g.
    • 1-month post-discharge if any of the labs at discharge (if known) are outside the
    reference range.
    • If the premie is transitioning to the breast or a term formula < 3-6 months
    corrected age.
    • If the premie has had marginal intake and slower growth.

Bone health can be assessed using the following labs.
•In the absence of other disease conditions, alkaline phosphatase provides an indirect
indicator of bone cell activity.
•Higher alkaline phosphatase along with lower calcium and phosphorus levels may
indicate a need for further assessment and supplementation. These labs are usually
done in the hospital setting, and may be done in the community up until 6-9 months
corrected age.
One last example to put it all
            together …
the ADHB (Auckland District Health
        Board) guidelines
More questions than
     answers?
     Sighhh…
More questions than
     answers?
     Sighhh…
Nutrition Practice Care Guidelines for Preterm Infants In the Community. Revised August 2006.
  Developed by Child Development and Rehabilitation Center, Nutrition Services, Oregon
  Department of Human Services, Nutrition & Health Screening – WIC Program. Oregon Pediatric
  Nutrition Practice Group
APPENDIX E: SELECTION OF FEEDING AT DISCHARGE
Feeding strategies for the preterm infant need to be evaluated on an individual basis. These guidelines are designed to help
the practitioner in making feeding selections to promote optimal nutrition. Birth weight, weight at discharge, and NICU course
are usually better predictors of risk than gestational age at birth. Breastfeeding after premature birth is recommended and
encouraged whenever possible. Lactation consultation is encouraged to promote successful breastfeeding and use of a breast
pump if needed.

1. High Risk – Very Low Birth Weight Infant

Category Definition:
• Birth weight < 1500gm (3.3 lbs)
• History of TPN and diuretics
• Demonstrates poor growth
• Poor intake (< 150 ml/kg/day)
• Elevated alkaline phosphatase (> 500 U/L) and/or low phosphorus (< 4)


Formula Feeding Recommendations:
• In majority of cases, these infants will need transitional formulas (EnfaCare or Neosure) until 9 months corrected age. If
change to a term formula, check labs and monitor growth.
• Continue on transitional formula unless:
o Infant cannot tolerate formula
o Excessive rate of weight gain
o Calcium and Phosphorus exceed normal limits


Breastfeeding Recommendations:
• Supplement breastfeeding with a transitional formula (Similac Neosure or Enfamil EnfaCare) until infant able to sustain
growth, ad lib milk intake, and lab values are within normal limits
• Discuss family’s breastfeeding goals in order to support breastfeeding while still maintaining infant’s growth and lab values.
Moderate Risk – Low Birth Weight Infant

Category Definition:
     • Infant had BW > 1500 gm (3.3 lbs), has good growth, use of TPN and diuretics was minimal and alkaline
     phosphatase level WNL
     • Osteopenia evidenced by serum phosphorous <4mg/dl, alkaline phosphatase > 500 U/L
     • Weight for corrected age is less than 5th percentile on CDC 2000 growth grids
     • Infant has bronchopulmonary dysplasia/chronic lung disease with steroid use


Formula Feeding Recommendations:
     • Provide transitional formulas (EnfaCare or Neosure) to 9 months corrected age.
     • Continue on transitional formula unless:
            o Infant cannot tolerate formula
            o Excessive rate of weight gain
            o Calcium and Phosphorus exceed normal limits
            o If change to a term formula, check labs and monitor growth


Breastfeeding Recommendations:
      • Supplement breastfeeding with a transitional formula (Similac Neosure or Enfamil EnfaCare) until infant able to
      sustain growth, ad lib milk intake, and lab values are within normal limits
      • Discuss family’s breastfeeding goals in order to support breastfeeding while still maintaining infant’s growth and
      lab values.
Low Risk – Low Birth Weight Infant

Breastfeeding Recommendations:
      • Breastfeed on demand.
      • If milk intake or supply is insufficient, as evidenced by slow growth on CDC growth grid, assess the
      need for lactation support or supplementation with standard formula such as Enfamil Lipil or Similac
      Advance.


Formula Feeding Recommendations:
     • Offer standard 20 kcal/oz iron fortified infant formula such as Enfamil Lipil or Similac Advance until 1
     year corrected age.


Category Definition:
     • Birth weight > 2000 gm (4.4 lbs)

More Related Content

What's hot

Respiratory Distress in New born
Respiratory Distress in New bornRespiratory Distress in New born
Respiratory Distress in New bornAnkit Agarwal
 
Feeding of low birth weight neonates
Feeding of low birth weight neonatesFeeding of low birth weight neonates
Feeding of low birth weight neonatesDr. Mahesh Yadav
 
Parenteral nutrition in neonat
Parenteral nutrition in neonatParenteral nutrition in neonat
Parenteral nutrition in neonatReyad Al_Faky
 
Growth charts in Neonates- Preterm and term
Growth charts in Neonates- Preterm and termGrowth charts in Neonates- Preterm and term
Growth charts in Neonates- Preterm and termSujit Shrestha
 
Fluid & electrolytes management in neonates
Fluid & electrolytes management in neonatesFluid & electrolytes management in neonates
Fluid & electrolytes management in neonatesSaurav Upadhyay
 
Treatment of neonatal hypoglycemia
Treatment of neonatal hypoglycemia  Treatment of neonatal hypoglycemia
Treatment of neonatal hypoglycemia mandar haval
 
Necrotising Enterocolitis(NEC)
Necrotising Enterocolitis(NEC)Necrotising Enterocolitis(NEC)
Necrotising Enterocolitis(NEC)Sid Kaithakkoden
 
Developmental care for neonates 2016
Developmental care for neonates 2016Developmental care for neonates 2016
Developmental care for neonates 2016gotolamy
 
Neonatal Hypoglycemia
Neonatal HypoglycemiaNeonatal Hypoglycemia
Neonatal HypoglycemiaDavid Mendez
 
Neonatal nutrition and feeding
Neonatal nutrition and  feedingNeonatal nutrition and  feeding
Neonatal nutrition and feedingPrak Viroth, RN
 
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemia
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemiaPresentation on neonatal hypocalcemia hypoglycemia hypomagnesaemia
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemiaGnana Jyothi
 
Approach to Hypoglycemia in childhood
Approach to Hypoglycemia in childhoodApproach to Hypoglycemia in childhood
Approach to Hypoglycemia in childhoodRavi Kumar
 
Neonatal hypoglycemia
Neonatal hypoglycemia Neonatal hypoglycemia
Neonatal hypoglycemia Azad Haleem
 
Management of a neonate with respiratory distress
Management of a neonate with respiratory distressManagement of a neonate with respiratory distress
Management of a neonate with respiratory distressSoumya Ranjan Parida
 

What's hot (20)

Respiratory Distress in New born
Respiratory Distress in New bornRespiratory Distress in New born
Respiratory Distress in New born
 
Feeding of low birth weight neonates
Feeding of low birth weight neonatesFeeding of low birth weight neonates
Feeding of low birth weight neonates
 
Parenteral nutrition in neonat
Parenteral nutrition in neonatParenteral nutrition in neonat
Parenteral nutrition in neonat
 
Growth charts in Neonates- Preterm and term
Growth charts in Neonates- Preterm and termGrowth charts in Neonates- Preterm and term
Growth charts in Neonates- Preterm and term
 
Fluid & electrolytes management in neonates
Fluid & electrolytes management in neonatesFluid & electrolytes management in neonates
Fluid & electrolytes management in neonates
 
Treatment of neonatal hypoglycemia
Treatment of neonatal hypoglycemia  Treatment of neonatal hypoglycemia
Treatment of neonatal hypoglycemia
 
Respiratory distress in newborn
Respiratory distress in newbornRespiratory distress in newborn
Respiratory distress in newborn
 
Feeding of lbw infants
Feeding of lbw infantsFeeding of lbw infants
Feeding of lbw infants
 
Necrotising Enterocolitis(NEC)
Necrotising Enterocolitis(NEC)Necrotising Enterocolitis(NEC)
Necrotising Enterocolitis(NEC)
 
Developmental care for neonates 2016
Developmental care for neonates 2016Developmental care for neonates 2016
Developmental care for neonates 2016
 
Neonatal Hypoglycemia
Neonatal HypoglycemiaNeonatal Hypoglycemia
Neonatal Hypoglycemia
 
Neonatal nutrition and feeding
Neonatal nutrition and  feedingNeonatal nutrition and  feeding
Neonatal nutrition and feeding
 
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemia
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemiaPresentation on neonatal hypocalcemia hypoglycemia hypomagnesaemia
Presentation on neonatal hypocalcemia hypoglycemia hypomagnesaemia
 
Feeding in the low birth weight infant
Feeding in the low birth weight infantFeeding in the low birth weight infant
Feeding in the low birth weight infant
 
FLUID MANAGEMENT IN NEONATES NICU
FLUID MANAGEMENT IN NEONATES NICU FLUID MANAGEMENT IN NEONATES NICU
FLUID MANAGEMENT IN NEONATES NICU
 
Neonatal Pain
Neonatal PainNeonatal Pain
Neonatal Pain
 
Approach to Hypoglycemia in childhood
Approach to Hypoglycemia in childhoodApproach to Hypoglycemia in childhood
Approach to Hypoglycemia in childhood
 
Neonatal hypoglycemia
Neonatal hypoglycemia Neonatal hypoglycemia
Neonatal hypoglycemia
 
Pediatric Obesity
Pediatric ObesityPediatric Obesity
Pediatric Obesity
 
Management of a neonate with respiratory distress
Management of a neonate with respiratory distressManagement of a neonate with respiratory distress
Management of a neonate with respiratory distress
 

Similar to Enteral nutrition in preterm neonates

Comparison of prolonged low volume milk and routine volume milk on
Comparison of prolonged low volume milk and routine volume milk onComparison of prolonged low volume milk and routine volume milk on
Comparison of prolonged low volume milk and routine volume milk onamir mohammad Armanian
 
Protective Gut And Nutritional Stratigies
Protective Gut And Nutritional StratigiesProtective Gut And Nutritional Stratigies
Protective Gut And Nutritional StratigiesPerwin Waly
 
Teduglutida en el síndrome de intestino corto. Experiencia en Pediatría
Teduglutida en el síndrome de intestino corto. Experiencia en PediatríaTeduglutida en el síndrome de intestino corto. Experiencia en Pediatría
Teduglutida en el síndrome de intestino corto. Experiencia en PediatríaJavier González de Dios
 
Enteral nutrition in infants and children
Enteral nutrition in infants and childrenEnteral nutrition in infants and children
Enteral nutrition in infants and childrenNgọc Thái Trương
 
HPD study published article
HPD study published articleHPD study published article
HPD study published articleMeghana Gudala
 
Oral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingOral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingShirlye Cahuaya
 
Oral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingOral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingmarlonluisf
 
Breastfeeding And The Use Of Human Milk
Breastfeeding And The Use Of Human MilkBreastfeeding And The Use Of Human Milk
Breastfeeding And The Use Of Human MilkBiblioteca Virtual
 
Nutrition in vlbw infants
Nutrition in vlbw infantsNutrition in vlbw infants
Nutrition in vlbw infantsVarsha Shah
 
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...ijtsrd
 
Recomend a strategy for mu2 2013
Recomend a strategy for mu2 2013Recomend a strategy for mu2 2013
Recomend a strategy for mu2 2013MAHINOJOSA45
 
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding And
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding AndRandomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding And
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding AndBiblioteca Virtual
 
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...Avianita3
 
Reproductive Health Program Planning in Public Health: What's the Evidence?
Reproductive Health Program Planning in Public Health: What's the Evidence?Reproductive Health Program Planning in Public Health: What's the Evidence?
Reproductive Health Program Planning in Public Health: What's the Evidence?Health Evidence™
 
Prevalence of Early Childhood Caries and its Association with Body Mass Index...
Prevalence of Early Childhood Caries and its Association with Body Mass Index...Prevalence of Early Childhood Caries and its Association with Body Mass Index...
Prevalence of Early Childhood Caries and its Association with Body Mass Index...iosrjce
 

Similar to Enteral nutrition in preterm neonates (20)

Probiotics 3 ecn
Probiotics 3 ecnProbiotics 3 ecn
Probiotics 3 ecn
 
Comparison of prolonged low volume milk and routine volume milk on
Comparison of prolonged low volume milk and routine volume milk onComparison of prolonged low volume milk and routine volume milk on
Comparison of prolonged low volume milk and routine volume milk on
 
Protective Gut And Nutritional Stratigies
Protective Gut And Nutritional StratigiesProtective Gut And Nutritional Stratigies
Protective Gut And Nutritional Stratigies
 
Breast feeding
Breast feedingBreast feeding
Breast feeding
 
DavisEarly_Infant_Temperament2013TICN
DavisEarly_Infant_Temperament2013TICNDavisEarly_Infant_Temperament2013TICN
DavisEarly_Infant_Temperament2013TICN
 
Teduglutida en el síndrome de intestino corto. Experiencia en Pediatría
Teduglutida en el síndrome de intestino corto. Experiencia en PediatríaTeduglutida en el síndrome de intestino corto. Experiencia en Pediatría
Teduglutida en el síndrome de intestino corto. Experiencia en Pediatría
 
Enteral nutrition in infants and children
Enteral nutrition in infants and childrenEnteral nutrition in infants and children
Enteral nutrition in infants and children
 
HPD study published article
HPD study published articleHPD study published article
HPD study published article
 
Oral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingOral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizing
 
Oral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizingOral probiotics reduce the incidence and severity of necrotizing
Oral probiotics reduce the incidence and severity of necrotizing
 
Breastfeeding And The Use Of Human Milk
Breastfeeding And The Use Of Human MilkBreastfeeding And The Use Of Human Milk
Breastfeeding And The Use Of Human Milk
 
Nutrition in vlbw infants
Nutrition in vlbw infantsNutrition in vlbw infants
Nutrition in vlbw infants
 
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...
A Quasi Experimental Study to Evaluate the Effect of Prefeeding Oral Stimulat...
 
Recomend a strategy for mu2 2013
Recomend a strategy for mu2 2013Recomend a strategy for mu2 2013
Recomend a strategy for mu2 2013
 
Breastfeeding
BreastfeedingBreastfeeding
Breastfeeding
 
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding And
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding AndRandomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding And
Randomized Clinical Trial Of Pacifier Use And Bottle Feeding Or Cupfeeding And
 
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...
NEW Dinner ANU 2023_Nutritional Interventions to Promote Brain Development an...
 
Reproductive Health Program Planning in Public Health: What's the Evidence?
Reproductive Health Program Planning in Public Health: What's the Evidence?Reproductive Health Program Planning in Public Health: What's the Evidence?
Reproductive Health Program Planning in Public Health: What's the Evidence?
 
Faltring growth
Faltring growthFaltring growth
Faltring growth
 
Prevalence of Early Childhood Caries and its Association with Body Mass Index...
Prevalence of Early Childhood Caries and its Association with Body Mass Index...Prevalence of Early Childhood Caries and its Association with Body Mass Index...
Prevalence of Early Childhood Caries and its Association with Body Mass Index...
 

More from Varsha Shah

pediatric emergencies
pediatric emergenciespediatric emergencies
pediatric emergenciesVarsha Shah
 
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docx
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docxPediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docx
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docxVarsha Shah
 
Examination in paediatric Medicine for medical students.pptx
Examination in paediatric Medicine for medical students.pptxExamination in paediatric Medicine for medical students.pptx
Examination in paediatric Medicine for medical students.pptxVarsha Shah
 
Neonatal presentations to Emergency department.pptx
Neonatal presentations to Emergency department.pptxNeonatal presentations to Emergency department.pptx
Neonatal presentations to Emergency department.pptxVarsha Shah
 
Approach to thalassemia with abdominal distension in children
Approach to thalassemia  with abdominal distension in childrenApproach to thalassemia  with abdominal distension in children
Approach to thalassemia with abdominal distension in childrenVarsha Shah
 
Jaundice in infant
Jaundice in infantJaundice in infant
Jaundice in infantVarsha Shah
 
Approach to Cafe au lait spots in children
Approach to Cafe au lait spots in childrenApproach to Cafe au lait spots in children
Approach to Cafe au lait spots in childrenVarsha Shah
 
Mcq in neonatology for medical students
Mcq in neonatology for medical studentsMcq in neonatology for medical students
Mcq in neonatology for medical studentsVarsha Shah
 
Blood in stool in neonates
Blood in stool in neonatesBlood in stool in neonates
Blood in stool in neonatesVarsha Shah
 
Approach to cardiac murmurs and cardiac examination in children
Approach to cardiac murmurs and cardiac examination in childrenApproach to cardiac murmurs and cardiac examination in children
Approach to cardiac murmurs and cardiac examination in childrenVarsha Shah
 
Developmental assessment for medical students
Developmental assessment for medical studentsDevelopmental assessment for medical students
Developmental assessment for medical studentsVarsha Shah
 
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1][Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]Varsha Shah
 
7 breastfeeding the premature and the sick term baby
7 breastfeeding the premature and the sick term baby7 breastfeeding the premature and the sick term baby
7 breastfeeding the premature and the sick term babyVarsha Shah
 
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...Varsha Shah
 
5 breastfeeding for working mums
5 breastfeeding for working mums5 breastfeeding for working mums
5 breastfeeding for working mumsVarsha Shah
 
4 The rationale for skin to-skin contact at birth and rooming in
4 The rationale for skin to-skin contact at birth and rooming in4 The rationale for skin to-skin contact at birth and rooming in
4 The rationale for skin to-skin contact at birth and rooming inVarsha Shah
 
4 rooming in and breast feeding
4 rooming in and breast feeding4 rooming in and breast feeding
4 rooming in and breast feedingVarsha Shah
 
3 common breastfeeding challenges and its management
3 common breastfeeding challenges and its management3 common breastfeeding challenges and its management
3 common breastfeeding challenges and its managementVarsha Shah
 
2 physiology and benefits of bf, risk of artificial feeding230113
2 physiology and benefits of bf, risk of artificial feeding2301132 physiology and benefits of bf, risk of artificial feeding230113
2 physiology and benefits of bf, risk of artificial feeding230113Varsha Shah
 
1 introduction to bfhi and 10 steps of breastfeeding
1 introduction to bfhi and 10 steps of breastfeeding1 introduction to bfhi and 10 steps of breastfeeding
1 introduction to bfhi and 10 steps of breastfeedingVarsha Shah
 

More from Varsha Shah (20)

pediatric emergencies
pediatric emergenciespediatric emergencies
pediatric emergencies
 
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docx
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docxPediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docx
Pediatrics Examinations Made Easy for Medical Students by Dr Varsha 2023.docx
 
Examination in paediatric Medicine for medical students.pptx
Examination in paediatric Medicine for medical students.pptxExamination in paediatric Medicine for medical students.pptx
Examination in paediatric Medicine for medical students.pptx
 
Neonatal presentations to Emergency department.pptx
Neonatal presentations to Emergency department.pptxNeonatal presentations to Emergency department.pptx
Neonatal presentations to Emergency department.pptx
 
Approach to thalassemia with abdominal distension in children
Approach to thalassemia  with abdominal distension in childrenApproach to thalassemia  with abdominal distension in children
Approach to thalassemia with abdominal distension in children
 
Jaundice in infant
Jaundice in infantJaundice in infant
Jaundice in infant
 
Approach to Cafe au lait spots in children
Approach to Cafe au lait spots in childrenApproach to Cafe au lait spots in children
Approach to Cafe au lait spots in children
 
Mcq in neonatology for medical students
Mcq in neonatology for medical studentsMcq in neonatology for medical students
Mcq in neonatology for medical students
 
Blood in stool in neonates
Blood in stool in neonatesBlood in stool in neonates
Blood in stool in neonates
 
Approach to cardiac murmurs and cardiac examination in children
Approach to cardiac murmurs and cardiac examination in childrenApproach to cardiac murmurs and cardiac examination in children
Approach to cardiac murmurs and cardiac examination in children
 
Developmental assessment for medical students
Developmental assessment for medical studentsDevelopmental assessment for medical students
Developmental assessment for medical students
 
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1][Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]
[Mary sheridan] from_birth_to_five_years_children(bookos.org)[1]
 
7 breastfeeding the premature and the sick term baby
7 breastfeeding the premature and the sick term baby7 breastfeeding the premature and the sick term baby
7 breastfeeding the premature and the sick term baby
 
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...
6 breastfeeding and drugs and acceptable medical reasons for artificial feedi...
 
5 breastfeeding for working mums
5 breastfeeding for working mums5 breastfeeding for working mums
5 breastfeeding for working mums
 
4 The rationale for skin to-skin contact at birth and rooming in
4 The rationale for skin to-skin contact at birth and rooming in4 The rationale for skin to-skin contact at birth and rooming in
4 The rationale for skin to-skin contact at birth and rooming in
 
4 rooming in and breast feeding
4 rooming in and breast feeding4 rooming in and breast feeding
4 rooming in and breast feeding
 
3 common breastfeeding challenges and its management
3 common breastfeeding challenges and its management3 common breastfeeding challenges and its management
3 common breastfeeding challenges and its management
 
2 physiology and benefits of bf, risk of artificial feeding230113
2 physiology and benefits of bf, risk of artificial feeding2301132 physiology and benefits of bf, risk of artificial feeding230113
2 physiology and benefits of bf, risk of artificial feeding230113
 
1 introduction to bfhi and 10 steps of breastfeeding
1 introduction to bfhi and 10 steps of breastfeeding1 introduction to bfhi and 10 steps of breastfeeding
1 introduction to bfhi and 10 steps of breastfeeding
 

Recently uploaded

Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Educationpboyjonauth
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsanshu789521
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docxPoojaSen20
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Krashi Coaching
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxmanuelaromero2013
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactdawncurless
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docxPoojaSen20
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Sapana Sha
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Interactive Powerpoint_How to Master effective communication
Interactive Powerpoint_How to Master effective communicationInteractive Powerpoint_How to Master effective communication
Interactive Powerpoint_How to Master effective communicationnomboosow
 
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting DataJhengPantaleon
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 

Recently uploaded (20)

Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Education
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha elections
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docx
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptx
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impact
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docx
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
 
Interactive Powerpoint_How to Master effective communication
Interactive Powerpoint_How to Master effective communicationInteractive Powerpoint_How to Master effective communication
Interactive Powerpoint_How to Master effective communication
 
Staff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSDStaff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSD
 
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 

Enteral nutrition in preterm neonates

  • 1.
  • 2. Enteral Nutrition in Preterm Neonates Dr Varsha Atul Shah
  • 3. Introduction  Proper nutrition in infancy is essential for – normal growth – immunity to infection – optimal neurologic and cognitive development.  Providing adequate nutrition to preterm infants is challenging because of several problems, some of them unique to these small infants. These problems include: – immaturity of bowel function – inability to suck and swallow – high risk of necrotizing enterocolitis (NEC) – illnesses that may interfere with adequate enteral feeding (e.g. RDS, patent ductus arteriosus) – medical interventions that preclude feeding (e.g., umbilical vessel catheters, exchange transfusion, indomethacin therapy)
  • 4. Long term outcome and Barker’s Hypothesis  There is evidence that adaptations in the metabolic and hormonal milieu in the fetal and immediate neonatal period can result in immediate benefit but adverse long-term outcome.  Nutritional insults at a vulnerable period of brain development, for example, have been shown to be associated with effects on brain size, cell number, behavior and learning memory.  The emergence of cardiovascular disease, hypertension, insulin resistance and obesity in low birth weight infants is another concern raising the issue of long-term ‘‘programming.’’  In addition, there may be transgenerational effects as evidenced by the association of low maternal birth weight and higher offspring hypertension in adulthood.  These changes may be exacerbated by postnatal malnutrition and poor growth that preterm infants experience.
  • 5. PHYSIOLOGY AND PATHOPHYSIOLOGY  The gut has formed and has completed its rotation back into the abdominal cavity by 10 weeks of gestation.  By 16 weeks, the fetus can swallow amniotic fluid.  GI motor activity is present before 24 weeks, but organized peristalsis is not established until 29-30 weeks and is facilitated by antenatal corticosteroid treatment.  Coordinated sucking and swallowing develops at 32-34 weeks.  By term, the fetus swallows about 150 cc/kg/day of amniotic fluid, which has 275 mOsm/L, contains carbohydrates, protein, fat, electrolytes, immunoglobulins and growth factors, and plays an important role in development of GI function. Preterm birth interrupts this development.
  • 6. Importance of Enteral Feeding  Even if nutrients are provided parenterally, lack of enteric intake leads to – decreased circulating gut peptides – slower enterocyte turnover and nutrient transport – decreased bile acid secretion – increased susceptibility to infection due to impaired barrier function by intestinal epithelium, lack of colonization by normal commensal flora and colonization by pathogenic organisms.  For fat digestion, the newborn depends on lingual lipase, which is stimulated by sucking and swallowing and by nutrients in the stomach but not the small bowel.
  • 8.
  • 9. Trophic feedings for parenterally fed infants (Review) 2008 The Cochrane Collaboration  Trophic feeding defined as dilute or full strength feedings providing < = 25 ml/kg/d for > = 5d (5-10days)  Trophic feedings vs. no feedings (10 trials): Among infants given trophic feedings, there was an overall reduction in days to full feeding (weighted mean difference [WMD] = -2.6 [95% confidence limits = -4.1, -1.0]), total days that feedings were held (WMD = -3.1 [-4.6, -1.6]), and total hospital stay (WMD = -11.4 [-17.2, -5.7] compared to infants given no enteral nutrient intake.  Tests for heterogeneity were significant in analyses of days to full enteral feedings, days to regain birth weight, days of phototherapy, and hospital stay.  There was no significant difference in necrotizing enterocolitis, although the findings do not exclude an important effect (relative risk = 1.16 [0.75, 1.79]; risk difference = 0.02 [-0.03, 0.06].  Trophic feedings vs. advancing feedings (one trial): Infants given trophic feedings required more days to reach full enteral feeding (13.4 [8.2, 18.6]) and tended to have a longer hospital stay (11.0 [-1.4, 23.4]) than did infants given advancing feedings. With only eight total cases of necrotizing enterocolitis, trophic feedings were associated with a marginally significant reduction in necrotizing enterocolitis (relative risk =0.14 [0.02, 1.07]; risk difference = -0.09 [-0.16, -0.01].
  • 10. Authors’ conclusions  In both comparisons, the group with the greater enteral intake (trophic feedings in the first comparison and advancing feedings in the second comparison) required significantly less time to reach full feedings and had a significant or near significant reduction in hospital stay.  In both comparisons, the group with the greater intake also had a higher incidence of NEC although the difference was not statistically significant. The concern is greatest for the advancing feeding regimen. Even when trophic feedings were compared to no feedings, the relative risk for NEC was 1.16 (0.75 - 1.79), a finding consistent with a 16% increase in NEC and a NNH of 50. A true increase of this magnitude might outweigh any shorter or long-term benefits of trophic feedings. Moreover, the 95% CI does not exclude the possibility that trophic feedings increase NEC by as much as 79% with a NNH of 17.  Whether no feedings, trophic feedings, or advancing feedings should initially be used is difficult to discern for a variety of reasons—the inherent difficulty of assessing enteral feedings in high-risk infants, the limited sample size and methodologic limitations of most studies to date, unexplained heterogeneity with respect to a number of outcomes, the potential for bias to affect the findings in unblinded studies, and the large number of infants who must be studied to assess the effect on necrotizing enterocolitis.  One or more large, well designed, multi-center trials are needed to compare these approaches to early feeding with respect to important clinical outcomes. A conclusive evaluation would assess effects on not only the survival rate without necrotizing enterocolitis prior to discharge from the neonatal unit but also on the survival rate without severe gastrointestinal or neurodevelopmental disability at >= 18 months age.
  • 11. Nasal versus oral route for placing feeding tubes in preterm or low birth weight infants. 2008 Cochrane Collaboration Main results  Two small randomised trials were identified.  Only one trial reported data on the pre-specified primary outcomes for this review. This trial found no evidence of effect on the time taken to establish enteral feeding nor the time taken to regain birth weight. However, the trial was underpowered (N= 46) to exclude modest effect sizes. Authors’ conclusions  There are insufficient data available to inform practice. A large randomised controlled trial is required to determine if the use of naso vs oro-enteric feeding tubes affects feeding, growth and development, and the incidence of adverse consequences in preterm or low birth weight infants.
  • 12. Continuous nasogastric milk feeding versus intermittent bolus milk feeding for premature infants less than 1500 grams, 2008 Cochrane collaboration Main results  Overall, the seven included trials, involving 511 infants, found no differences in time to achieve full enteral feeds between feeding methods (weighted mean difference (WMD 2 days; 95%CI -0.3, 3.9) .  In the subgroup analysis of those studies comparing continuous nasogastric vs. intermittent bolus nasogastric milk feedings the findings remained unchanged (WMD 2 days, 95% CI -0.4, 4.1).  There was no significant difference in somatic growth and incidence of NEC between feeding methods irrespective of tube placement.  One study noted a trend toward more apneas during the study period in infants fed by the continuous tube feeding method compared to those fed by intermittent feedings delivered predominantly by orogastric tube placements [mean difference (MD) 14.0 apneas during study period; 95% CI -0.2, 28.2].  In subgroup analysis based on weight groups, one study suggested that infants less than 1000 grams and 1000 - 1250 grams birth weight gained weight faster when fed by the continuous nasogastric tube feeding method compared to intermittent nasogastric tube feeding method (MD 2.0 g/day; 95% CI 0.5, 3.5; MD 2.0 g/day; 95% CI 0.2, 3.8, respectively).
  • 13. A trend toward earlier discharge for infants less than 1000 grams birth weight fed by the continuous tube feeding method compared to intermittent nasogastric tube feeding method (MD -11 days; 95% CI -21.8, -0.2). Authors’ conclusions  Small sample sizes, methodologic limitations, inconsistencies in controlling variables that may affect outcomes, and conflicting results of the studies to date make it difficult to make universal recommendations regarding the best tube feeding method for premature infants less than 1500 grams.  The clinical benefits and risks of continuous vs. intermittent nasogastric tube milk feeding cannot be reliably discerned from the limited information available from randomized trials to date.
  • 14. Transpyloric versus gastric tube feeding for preterm infants. 2008 Cochrane Collaboration  Data from nine trials were available.  No evidence of an effect on short term growth rates was found: weight:WMD -0.7 g/week (95% confidence interval (CI) -25.2, 23.8); crown heel length: WMD -0.7 mm/week (95% CI -2.4, 1.0); head circumference: WMD 0.6 mm/week (95% CI -0.9, 2.1).  Longer term growth was reported in one study. There were not any statistically significant differences between the groups in the mean body weight or occipitofrontal head circumference at three months or at six months corrected age.  None of the included studies provided data on neurodevelopmental outcomes.  Transpyloric feeding was associated with a greater incidence of gastro- intestinal disturbance (RR 1.45, 95% CI 1.05, 2.09). There was some evidence that feeding via the transpyloric route increased mortality (RR 2.46, 95% CI 1.36, 4.46). However, the outcomes of the study that contributed most to this finding were likely to have been affected by selective allocation of the less mature and sicker infants to transpyloric feeding.
  • 15.  No statistically significant differences in the incidence of other adverse events, including necrotising enterocolitis, intestinal perforation, and aspiration pneumonia was found. Authors’ conclusions  No evidence of any beneficial effect of transpyloric feeding in preterm infants was found. However, evidence of adverse effects was noted. Feeding via the transpyloric route cannot be recommended for preterm infants
  • 16. Ad libitum or demand/semi-demand feeding versus scheduled interval feeding for preterm infants, 2008 Cochrane collaboration  Seven randomised controlled trials that compared ad libitum or demand/semi- demand regimes with scheduled interval regimes in preterm infants in the transition phase from intragastric tube to oral feeding were found.  The trials were generally small and of variable methodological quality. The duration of the intervention and the duration of data collection and follow up in most of the trials is not likely to have allowed detection of measurable effects on growth.  The single trial that assessed growth for longer than one week found that the rate of weight gain was lower in the ad libitum fed infants [mean difference -3.30 (95% confidence interval -6.2 to -0.4) grams per kilogram per day].  Two trials reported that feeding preterm infants using an ad libitum or demand/semi- demand feeding regime allowed earlier discharge from hospital, but the other trials did not confirm this finding.  Not able to undertake meta-analyses because of differences in study design and in the way the findings were reported. Authors’ conclusions  There are insufficient data at present to guide clinical practice. A large randomised controlled trial is needed to determine if ad libitum of demand/semi-demand feeding of preterm infants affects clinically important outcomes. This trial should focus on infants in the transition phase from intragastric tube to oral feeding and should be of sufficient duration to assess effects on growth and time to oral feeding and hospital discharge.
  • 17.
  • 18. What should we feed?
  • 19. Breast milk lah, no need to ask… Really?
  • 20. Formula milk versus maternal breast milk for feeding preterm or low birth weight infants 2008 Cochrane Collaboration. Main results  No eligible trials were identified. Authors’ conclusions  There are no data from randomised trials of formula milk versus maternal breast milk for feeding preterm or low birth weight infants.  This may relate to a perceived difficulty of allocating an alternative feed to an infant whose mother wishes to feed with her own breast milk.  Maternal breast milk remains the default choice of enteral nutrition because observational studies, and meta-analyses of trials comparing feeding with formula milk versus donor breast milk, suggest that feeding with breastmilk has major non-nutrient advantages for preterm or low birth weight infants.
  • 21.
  • 22. Donor breast milk versus infant formula for preterm infants: systematic review and meta-analysis Cochrane collaboration 2006 Results: •Seven studies (including five randomised controlled trials), all from the 1970s and 1980s, fulfilled the inclusion criteria. All studies compared the effect of sole donor breast milk with formula (combined n = 471). One of these also compared the effect of donor breast milk with formula given as a supplement to mother’s own milk (n = 343). No studies examined fortified donor breast milk. •A meta-analysis based on three studies found a lower risk of NEC in infants receiving donor breast milk compared with formula (combined RR 0.21, 95% CI 0.06 to 0.76). •Donor breast milk was associated with slower growth in the early postnatal period, but its long-term effect is unclear. Conclusion: •Donor breast milk is associated with a lower risk of NEC and slower growth in the early postnatal period, but the quality of the evidence is limited. Further research is needed to confirm these findings and measure the effect of fortified or supplemented donor breast milk.
  • 23. Protein supplementation of human milk for promoting growth in preterm infants Cochrane collaboration 2000 MAIN RESULTS: •Protein supplementation of human milk results in increases in short term weight gain (WMD 3.6 g/kg/day, 95% CI 2.4 to 4.8 g/kg/day), linear growth (WMD 0.28 cm/week, 95% CI 0.18 to 0.38 cm/week) and head growth (WMD 0.15 cm/week, 95% CI 0.06 to 0.23 cm/week). •There are insufficient data to evaluate long term neurodevelopmental and growth outcomes. There are too few infants studied to be certain that adverse effects of protein supplementation are not increased. Blood urea levels are increased (WMD 1.0 mmol/l, 95% CI 0.8 to 1.2 mmol/l). REVIEWER'S CONCLUSIONS: •Protein supplementation of human milk in relatively well preterm infants results in increases in short term weight gain, linear and head growth. Urea levels are increased, which may reflect adequate rather than excessive dietary protein intake. •Further research should be directed towards the evaluation of specific levels of protein intake in preterm infants and the clinical effects of supplementation with protein, including long term growth and neurodevelopmental outcomes. This may best be done in the context of refinement of available multicomponent fortifier preparations.
  • 24. Multicomponent fortified human milk for promoting growth in preterm infants. Cochrane collaboration 2004 MAIN RESULTS • Supplementation of human milk with multicomponent fortifiers (in the form of protein, calcium, phosphate, and carbohydrate, as well as vitamins and trace minerals) is associated with short term increases in weight gain, linear and head growth. There is no effect on serum alkaline phosphatase levels; it is not clear if there is an effect on bone mineral content. Nitrogen retention and blood urea levels appear to be increased. •There are insufficient data to evaluate long term neurodevelopmental and growth outcomes, although there appears to be no effect on growth beyond one year of life. •Use of multicomponent fortifiers does not appear to be associated with adverse effects, although the total number of infants studied and the large amount of missing data reduces confidence in this conclusion. Blood urea levels are increased and blood pH levels minimally decreased, but the clinical significance of this is uncertain. REVIEWER'S CONCLUSIONS: •Multicomponent fortification of human milk is associated with short-term improvements in weight gain, linear and head growth. Despite the absence of evidence of long-term benefit and insufficient evidence to be reassured that there are no deleterious effects, it is unlikely that further studies evaluating fortification of human milk versus no supplementation will be performed. •Further research should be directed toward comparisons between different proprietary preparations and evaluating both short-term and long-term outcomes in search of the "optimal" composition of fortifiers.
  • 25. Fortification of Preterm Human Milk for Feeding Preterm Infants – Yale Pediatric Protocol  Preterm human milk (PTHM) is milk expressed by a mother for her infant following a preterm delivery. – may be stored in the Breast Milk Refrigerator for up to 48 hours. – If the milk will not be fed to the infant within that time, it should be frozen as early as possible after expression in appropriate-sized volumes and then gently thawed prior to use. Once thawed, it should be used within about 4 hours.  Infants should be fed full strength human milk as their initial enteral feeding.  When to fortify: – After an infant tolerates full enteral feeding of full-strength human milk for 5-7 days – Or, after he/she tolerates about 75% his/her total daily volume (IV + NG) as full strength human milk  How to fortify – adding a powdered Human Milk Fortifier – by mixing human milk with an equal volume of Premature Formula  Mixing equal volumes of human milk and Premature Formula or Natural Care results in a nutrient concentration that equals an average between human milk and formula – Or, by mixing human milk with an equal volume of Natural Care(similac) Human Milk Fortifier.  The decision on which method of fortification to use will depend upon the mother's milk production with discussion with Lactation Consultant.  Why fortify – Increases the content of nutrients in the infant's diet, esp calcium and phosphorus. (See table next page) – The composition of PTHM varies with the duration of lactation; an approximate composition of "mature" PTHM is also shown
  • 26.
  • 27.
  • 28. Special Formula Use Recommendations Nutrition Practice Care Guidelines for Preterm Infants In the Community. Revised August 2006. Developed by Child Development and Rehabilitation Center, Nutrition Services, Oregon Department of Human Services, Nutrition & Health Screening – WIC Program. Oregon Pediatric Nutrition Practice Group  Preterm formula and Human Milk Fortifier: generally for infants weighing less than 1,850 to 2,000 gm (about 4 to 4 ½ lbs). It is inappropriate for most infants who weigh more than 2.5 kg (5 ½ lbs) or are taking in over 500 ml daily to be fed premature formulas (Enfamil Premature Lipil or Similac Special Care) or Human Milk Fortifiers (Similac HMF, Similac Natural Care or Enfamil HMF) for all their feeds because of the higher vitamin A and vitamin D content of these formulas and the possible risk of hypervitaminosis.  Transitional Formulas: (also called “post-discharge premature formulas”) Formulas such as Enfamil EnfaCare Lipil and Similac Neosure Advance provide 22 kcal/oz and have higher levels of protein, calcium, phosphorus, vitamins and other minerals than standard infant formulas. Research has shown that premature infants fed these formulas have improved growth and bone mineralization compared to those fed standard infant formulas. *See recommendation below for length of time an infant needs to remain on a transitional formula.
  • 29.  Specialized formulas – These include Pregestimil, Alimentum, Nutramigen, Neocate and Elecare, and may be indicated based on feeding intolerance. – Enfamil AR is not indicated for preterm infants due to the risk of the formation of lactobezoars (hard clumps of undigested milk curds). These are “term” formulas and thus have less calcium, phosphorus, and protein than transitional formulas. If preterm infants are given these formulas, they should be followed more closely by an RD and the appropriate labs should be checked.  Soy-based formulas are not recommended for preterm infants. Preterm infants receiving soy formula have suboptimal carbohydrate and mineral absorption and utilization than cow’s milk-based formula. AAP doesn’t recommend soy formula for infants born < 1800 g since preterm infants showed significantly less weight gain, less linear growth, and lower serum albumin levels than those infants receiving cow’s milk-based formulas. Studies also have shown lower levels of bone marker formation in the premature population which can lead to osteopenia.  Goat’s milk is not recommended for preterm infants. Goat’s milk is deficient in folic acid and vitamin B6. It is also higher in protein than human milk and infant formula which puts the premature infant at risk for dehydration due to the higher renal solute load.
  • 30. Dorset County Hospital Neonatal Unit Guidelines for enteral feeding (Mar05)  Milk of choice, in order of preference, is: – 1. Breast milk – suckled by baby from the breast – 2. Mother’s expressed breast milk (EBM), fresh – 3. Mother’s EBM, frozen – 4. Donor EBM* – 5. Formula feed appropriate to gestational age and birth weight  * consider using when:  baby deemed at high risk of developing necrotising enterocolitis (NEC) – see section 3  if it is available, to intensive care babies  poor tolerance of formula feeds  NB parental consent should be sought prior to a baby being given donor EBM  This hospital actively promotes the benefits of breast feeding for all infants. There are particular benefits of breast milk for the preterm infant and sick neonate (eg reduced incidence of necrotising enterocolitis - ref 1).
  • 31. Dorset County (cont’d) Early nutrition in the preterm group  All mothers should be encouraged to produce milk for their baby as soon as possible after delivery if their baby is not able to feed naturally. Even if the mother is not planning to breast feed in the longer term or unwilling to breast feed, they should be advised why EBM is the best nutritional support for their baby.  For premature infants whose mother’s wish to breast feed in the longer term, skin to skin contact should be strongly encouraged from an early stage. Cup feeding is often possible from 30 weeks gestation. Bottle feeding should be avoided if at all possible given the different technique a baby uses to bottle feed compared to feeding at the breast.  If formula feeds are being used, the baby should not be started on a preterm formula until they are tolerating 150 ml/kg/day of a standard formula feed.
  • 32. Dorset County (cont’d) Early nutrition in the preterm group a) Under 2kg  For babies with a birth weight of less than 1.2 kg (and occasionally larger babies) parenteral nutrition will normally be necessary during the first 1-2 weeks of life. This should be written up as soon as possible after delivery so that it can be commenced within 1-2 days of delivery (see TPN guidelines).  i) Non-nutritive feeding (minimal enteral nutrition): – Small amounts of breast milk started in the first few days of life (day 1 if possible) – encourage gut maturity (ref 2) – Start at 1ml every 4-6 hours if <1kg (ref 5) and 1ml/hr if >1kg (ref 3). Maternal EBM is preferable to donor breast milk. – Please note section on management of infants at high risk of NEC (section 3)  ii) Rate of increase of feed: – Once tolerating minimal enteral nutrition, increase the volume given at each feed. The rate of increase should be decided for each individual baby but would normally be less than 35ml/kg per 24 hours for babies less than 1200gm (ref 4). Larger babies will possibly tolerate faster increments in feed volume.
  • 33. Dorset County (cont’d) Early nutrition in the preterm group  iii) Final feed volume: – Increase feed volumes up to a maximum of: – 200-220 ml/kg/day for EBM – 180 ml/kg for term formula (or Nutriprem 2) – 160 ml/kg/day for preterm formula*  Once an infant is tolerating >50% of their total fluid volume enterally, lipid can be discontinued in the TPN (see TPN guidelines).  * If formula feeds are being used, the baby should not be started on a preterm formula until they are tolerating 150 ml/kg/day of a standard formula feed. b) greater than 2kg  Where possible allow to demand feed.  For well infants below 37 weeks gestation, consider starting feeds at 60ml/kg/day on day 1 of feeding aiming to reach 150ml/kg/day within 4 days depending on progress.
  • 34. Dorset County (cont’d) Early nutrition in the preterm group 3. High risk groups for NEC  Particular high risk groups include: – Preterm infants < 1kg – Severe intrauterine growth retardation – Absent/reversed end diastolic flow on umbilical artery Doppler – Sepsis  For infants with absent or reversed end diastolic flow do not feed enterally for 3 days.  For at risk groups consider waiting for maternal EBM before starting feeds. In these circumstances feeding human milk at a rate 1 ml every 4-6 hours is recommended (ref 5). If maternal EBM is not available for these groups consider the use of donor EBM.  For babies at high risk of necrotising enterocolitis (NEC) there is some evidence that a rate of increase not exceeding 20ml/kg per 24 hours reduces the incidence of NEC (ref 6).  * After a suspected episode of NEC feeds should be reintroduced using EBM (maternal if possible but donor if not available). If maternal EBM not available and mother refused permission to use donor EBM, consider using Pregestimil (see section 9 for advise on increasing calorie intake if inadequate growth on this formula).
  • 35. References:  1. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis. Lancet 1990; 336: 1519-1523.  2. McClure RJ and Newell SJ. Randomised controlled trial of trophic feedingand gut motility. Arch Dis Child Neonatal Ed 1999; 80: F54-58.  3. Child Nutrition Panel “Feeding the preterm infant”. Z740127 May 2002  4. Kennedy KA et al. Rapid versus slow rate of advancement of feeding in parenterally fed low birth weight infants. Cochrane Review. 2001.  5. King C. Neonatal Unit Enteral Feeding Policy. Hammersmith Hospital NHS Trust 2000.  6. Anderson DM and Kliegman RM. The relationship of neonatal alimentation practice to the occurrence of endemic necrotising enterocolitis. Am J Perinatol 1991; 8: 62-67.  7. Cooke RJ and Embleton ND. Feeding issues in preterm infants. J. Pediat. Neonatal Ed 2000; 83: F215-218.  8. Nutrition in low birth weight infants. Clinical Paediatric Dietetics 2001.
  • 36. Enteral Feeding guidelines for the preterm infant within the neonatal service of the Leeds Teaching Hospitals NHS Trust
  • 37. GROUP 1. 34-36 weeks gestational age and birthweight > 1.5Kg who is on the neonatal unit due to a lack of transitional care capacity.  Timing of enteral: Immediately  Mode of enteral: NG or oral  Type of milk: EBM or Nutriprem (as per policy)  Rate of increase: Full feeds as soon as able GROUP 2. 34-36 weeks gestational age and birthweight > 1.5Kg admitted to Neonatal Unit for whatever reason  Feeding will depend on primary pathology. If considered ready for feeding, then feed as Feeding Group 1. GROUP 3. 34-36 weeks gestational age and birthweight < 1.5Kg  These infants are under the 2nd centile.  Timing of enteral: Delay for up to 24 hours in marked IUGR, or make decision to feed immediately  Mode of enteral: NG or oral  Type of milk: EBM or Nutriprem 1 (as per policy)  Rate of increase: 60mls/kg increasing at a maximum of 1ml/Kg per day  (0.5ml 12 hourly) up to total of 150/ml/kg/day GROUP 4. 31-33 weeks, NO IUGR or A/REDF on antenatal Doppler  Timing of enteral: Immediately  Mode of enteral: NG  Type of milk: EBM or Nutriprem 1(as per policy)  Rate of increase: 60mls/kg, max increase per day < 25ml/kg  (0.5ml/h, 8 hourly) as tolerated*
  • 38. GROUP 5. <= 30 weeks, No IUGR (> 2nd centile) or A/REDF on antenatal Doppler  Timing of enteral: Delay for 24 hours after birth  Mode of enteral: NG  Type of milk: EBM or Nutriprem 1(as per policy)  Rate of increase: 1ml/kg, increasing maximum 8 hourly as tolerated* for first 72 hours. Increase to 0.5ml/kg 6 hourly maximum as tolerated* after 72 hours of age.  Note: Slow feeding to 12 hourly if evidence of failure to tolerate*.  Maximum daily increase not to exceed 25 mls/kg per day. GROUP 6. <= 33 weeks, IUGR (<= 2nd centile) or A/REDF on antenatal Doppler  Timing of enteral: Delay at least 24 hours  Mode of enteral: NG  Type of Milk: EBM or Donor EBM (see below)  Rate of increase: – <= 1kg: 0.5ml/hr initially increasing by 0.5 ml/24 hours as tolerated – > 1kg 1ml/hr initially increasing by 1ml/24 hours as tolerated  When full feeds established continue, if using Donor EBM, continue on full feeds (maximum 165ml/Kg per day) for at least 5 days. Switch to Nutriprem 1 after this time.  Parenteral Nutrition (PN) will be required for this group (see PN protocol) *Tolerating enteral feeds  Babies are considered to be tolerating enteral feeds if  • 4 hourly NG aspirates are < 25% of total infused in the preceding 4 hours  • No significant abdominal distension  • No significant vomiting  • No bile-stained aspirates
  • 40.
  • 41. Longchain polyunsaturated fatty acid supplementation in preterm infants Cochrane collaboration 2003 Main results •Of the eleven randomised trials included in the review, two of these were not classified as of high quality despite blinded assessment and complete follow-up, due to problems with assessment methodology. Visual acuity Visual acuity over the first year was measured by Teller acuity cards in six studies, by VEP in four studies and by ERG in two studies. Most studies found no significant differences in any visual assessment between supplemented and control infants. Development Most of the trials have used Bayley Scales of Infant Development (BSID) at 12 to 24 months postterm and shown no significant effect following supplementation. Meta- analysis of BSID of three studies (Fewtrell 2002, O'Connor 2001, van Wezel 2002) shows no significant effect of supplementation on development. Carlson 1993 and Carlson 1996 demonstrated lower novelty preferences (possibly predictive of lower intelligence) in the supplemented compared with the control group. The investigators however concluded that supplemented infants may have more rapid visual information processing given that they had more looks and each look was of shorter duration.
  • 42. Growth Most trials have reported no significant effect of LCPUFA supplementation on growth of preterm infants. Two trials (Carlson 1993, Carlson 1996) suggest that LCPUFA supplemented infants grow less well than controls, possibly due to a reduction in AA levels which occurs when n-3 supplements are used without n-6 supplements. Recent trials with addition of AA to the supplement have reported no significant effect on growth. Fewtrell 2002 reported mild reductions in length and weight z scores at 18 months. Contrary to these results, the meta-analysis of five studies (Uauy 1992, Carlson 1996, Hansen 1997, Vanderhoof 1999, Innis 2002) showed increased weight and length at two months post-term in supplemented infants. Side effects Uauy 1992 reported no significant effect of LCPUFA supplementation on bleeding time and red cell membrane fragility. Reviewers' conclusions Infants enrolled in the trials were relatively mature and healthy preterm infants. Assessment schedule and methodology, dose and source of supplementation and fatty acid composition of the control formula varied between trials. No long-term benefits were demonstrated for infants receiving formula supplemented with LCPUFA. There was no evidence that supplementation of formula with n-3 and n-6 LCPUFA impaired the growth of preterm infants.
  • 43. Is supplementary iron useful when preterm infants are treated with erythropoietin? BestBets 2006  In [a preterm infant who is receiving rHuEPO therapy] does [iron supplementation] reduce the requirement for [blood transfusion]? If so, what method of administration and dose reduces it most successfully
  • 44. Comment(s) •Anaemia in premature infants is a common problem. Although erythropoietin is not used widely in neonatal practice, there is evidence of its efficacy in reducing the need for transfusion in preterm infants,(Shannon) especially if they are not extremely small or sick.(Soubasi). It is regularly used in situations where blood transfusion is unacceptable. •Iron supplementation has been a standard in neonatal care for preterm infants for many years and helps to reduce late anaemia (Franz) if given with vitamins, especially vitamin E.(Jansson). However, when stimulating erythropoiesis with rHuEpo to reduce the need for transfusion, iron availability becomes critical. • Several studies have investigated rHuEpo efficacy in preterm infants and most of them have used supplementary iron in either the oral or parenteral routes. The literature on the use of rHuEpo and iron mostly consists of studies on dose variation of rHuEpo rather than variation in the iron supplementation. •Our search yielded seven studies, but one(Pollak) was excluded owing to poor methodological quality. Two studies by Carnielli et al and Fujiu et al compared rHuEpo and oral iron supplementation with rHuEpo alone; however, the interpretation of the data is difficult because Carnielli et al reported their results only as mean values and logarithms, making statistical analysis difficult. Fujiu et al found that no infants in either arm of their study required a blood transfusion.
  • 45. •Although this may suggest that there was no difference between the groups, the sample size was small, with only 24 infants in total, and the clinical equivalence could not be shown. In addition, the authors state that losses due to phlebotomy in their study were lower than those in other similar studies. This may be relevant, as one of the most common causes of the anaemia of prematurity is iatrogenic blood loss. •Kivivuori et al and Meyer et al compared rHuEPO treatment and parenteral iron supplementation with rHuEPO treatment and oral iron supplementation. Combined data from the two studies showed that there was no significant difference between the groups for the number of blood transfusions given (odds ratio (OR) 1.65, 95% confidence interval (CI) 0.41 to 6.64). This seems to suggest that oral iron supplementation is at least sufficient. However, one study used intravenous iron supplementation whereas the other used intramuscular iron supplementation. Differences in absorption of these two different routes may be relevant, but no study has been carried out comparing intravenous with intramuscular iron supplementation. •Bader et al and Nazir et al compared infants receiving rHuEPO treatment and high- dose oral iron supplementation with those receiving rHuEPO treatment and low-dose oral iron supplementation. There was no significant difference between the two groups, when combining data for the two studies, regarding the number of blood transfusions (OR 0.46, 95% CI 0.04 to 5.75).
  • 46. Preterm infants on special-care baby units frequently become anaemic and require top-up blood transfusions. Preterm infants often have low iron stores; this becomes more evident if they do not receive transfusions or iron supplementation. Erythropoietin is used occasionally to stimulate red cell production and prevent anaemia, and the increased erythropoiesis that occurs as a result of rHuEPO treatment will deplete iron stores.  The studies currently available do not give an adequate answer to the question as to which is the best mode and dose of iron supplementation with rHuEpo treatment. On the basis of the principle of using the lowest effective dose and the least invasive mode of administration, at present, low-dose oral iron would seem appropriate for supplementation when rHuEpo is used in preterm infants. Clinical Bottom Line  Evidence available to strongly support any specific recommendation for iron supplementation with recombinant erythropoietin treatment in premature infants (grade D) is insufficient. Low-dose oral iron supplementation is not inferior to other treatment regimens (grade B).
  • 47. Glutamine supplementation to prevent morbidity and mortality in preterm infants Cochrane collaboration 2008 Main results •2365 preterm infants have participated in seven randomised controlled trials. All of the participating infants were of very low birth weight. •Three trials assessed enteral glutamine supplementation and four trials assessed parenteral glutamine supplementation. •The trials were generally of good methodological quality with adequate allocation concealment, blinding of caregivers and assessors to the intervention, and complete or near-complete follow-up of recruited infants. •Glutamine supplementation does not have a statistically significant effect on mortality: typical relative risk 0.98 (95% confidence interval 0.80 to 1.20); typical risk difference 0.00 (95% confidence interval -0.03 to 0.02). •The only trial that assessed long-term outcomes did not find any statistically significant differences in various assessments of neurodevelopment at 18 months corrected age. Glutamine supplementation does not have a statistically significant effect on other neonatal morbidities including invasive infection, necrotising enterocolitis, time to achieve full enteral nutrition, or duration of hospital stay. Authors' conclusions •The available data from good quality randomised controlled trials indicate that glutamine supplementation does not confer benefits for preterm infants. The narrow confidence intervals for the effect size estimates suggest that a further trial of this intervention is not a research priority.
  • 48.
  • 49. Iodine supplementation for the prevention of mortality and adverse neurodevelopmental outcomes in preterm infants (Review) Main results Copyright © 2008 The Cochrane Collaboration  We found only one randomised controlled trial (N = 121) that fulfilled the review eligibility criteria (Rogahn 2000).  The participants were infants born before 33 weeks’ gestation (but most were of birth weight greater than 1000 grams). The primary aim of this trial was to assess the effect of iodine supplementation on thyroid function. The investigators did not detect any statistically significant effects on the plasma levels of thyroxine (free and total), triiodothyronine, or thyrotrophin in preterm infants (measured up to 40 weeks’ post- conceptional age). Only one infant died and the trial was therefore underpowered to detect an effect on mortality. The trial did not assess the effect of the intervention on neurodevelopmental morbidity. There was not a statistically significant difference in the incidence of chronic lung disease. Authors’ conclusions  There are insufficient data at present to determine whether providing preterminfants with supplemental iodine (to match fetal accretion rates) prevents morbidity and mortality in preterm infants.  Future randomised controlled trials of iodine supplementation should focus on extremely preterm and extremely low birth weight infants, the group at greatest risk of transient hypothyroxinaemia. These trials should aim to assess the effect of iodine supplementation on clinically important outcomes including respiratory morbidity and longer term neurodevelopment.
  • 50. Lactase treated feeds to promote growth and feeding tolerance in preterm infants (Review) Copyright © 2008 The Cochrane Collaboration Main results  One study enrolling 130 infants of 26 - 34 weeks postconceptual age (mean postnatal age at entry 11 days) was identified and no identified study was excluded.  The study was a double blind randomized controlled trial of high quality. Lactase treated feeds were initiated when enteral feedings provided > 75% of daily intake. None of the primary outcomes outlined in the protocol for this review and only one of the secondary outcomes, necrotizing enterocolitis (NEC), were reported on. The RR for NEC was 0.32 (95% CI  0.32 (0.01, 7.79); the RD was -0.02 (95% CI -0.06, 0.03) (a reduction which was not statistically significant). There was a statistically significant increase in weight gain at study day 10 in the lactase treated feeds group but not at any other time points. Overall, there was not a statistically significant effect on weight gain. No adverse effects were noted. Authors’ conclusions  The only randomized trial to date provides no evidence of significant benefit to preterm infants from adding lactase to their feeds.  Further research regarding effectiveness and safety are required before practice recommendations can be made. Randomized controlled trials comparing lactase vs placebo treated feeds and enrolling infants when enteral feeds are introduced are recommended. The primary and secondary outcomes for effectiveness and safety should include those identified in this review.
  • 51. Effect of taurine supplementation on growth and development in preterm or low birth weight infants (Review) Copyright © 2008 The Cochrane Collaboration Main results  Nine small trials were identified.  In total, 189 infants participated. Most participants were greater than 30 weeks gestational age at birth and were clinically stable. In eight of the studies, taurine was given enterally with formula milk. Only one small trial assessed parenteral taurine supplementation.  Taurine supplementation increased intestinal fat absorption [weighted mean difference 4.0 (95% confidence interval 1.4, 6.6) percent of intake]. However, meta- analyses did not reveal any statistically significant effects on growth parameters assessed during the neonatal period or until three to four months chronological age [rate of weight gain: weighted mean difference - 0.25 (95% confidence interval -1.16, 0.66) grams/kilogram/day; change in length: weighted mean difference 0.37 (95% confidence interval -0.23, 0.98) millimetres/week; change in head circumference: weighted mean difference 0.15 (95% confidence interval -0.19, 0.50) millimeters/week].  There are very limited data on the effect on neonatal mortality or morbidities, and no data on long-term growth or neurological outcomes. Authors’ conclusions  Despite that lack of evidence of benefit from randomised controlled trials, it is likely that taurine will continue to be added to formula milks and parenteral nutrition solutions used for feeding preterm and low birth weight infants given the putative association of taurine deficiency with various adverse outcomes.  Further randomised controlled trials of taurine supplementation versus no supplementation in preterm or low birth weight infants are unlikely to be viewed as a research priority, but there may be issues related to dose or duration of supplementation in specific subgroups of infants that merit further research.
  • 52. Probiotics for prevention of necrotizing enterocolitis in preterm infants Cochrane review 2007 Main results  Nine eligible trials randomizing 1425 infants were included.  Included trials were highly variable with regard to enrollment criteria (i.e. birth weight and gestational age), baseline risk of NEC in the control groups, timing, dose, formulation of the probiotics, and feeding regimens. Data regarding extremely low birth weight infants (ELBW) could not be extrapolated.  In a meta-analysis of trial data, enteral probiotics supplementation significantly reduced the incidence of severe NEC (stage II or more) [typical RR 0.32 (95% CI 0.17, 0.60)] and mortality [typical RR 0.43 (95% CI 0.25, 0.75]. There was no evidence of significant reduction of nosocomial sepsis [typical RR 0.93 (95% CI 0.73, 1.19)] or days on total parenteral nutrition (TPN) [WMD -1.9 (95% CI -4.6, 0.77)].  The included trials reported no systemic infection with the probiotics supplemental organism. The statistical test of heterogeneity for NEC, mortality and sepsis was insignificant. Reviewers' conclusions  Enteral supplementation of probiotics reduced the risk of severe NEC and mortality in preterm infants. This analysis supports a change in practice in premature infants > 1000 g at birth. Data regarding outcome of ELBW infants could not be extracted from the available studies; therefore, a reliable estimate of the safety and efficacy of administration of probiotic supplements cannot be made in this high risk group. A large randomized controlled trial is required to investigate the potential benefits and safety profile of probiotics supplementation in ELBW infants.
  • 53. Erythromycin for the prevention and treatment of feeding intolerance in preterm infants (Review) Copyright © 2008 The Cochrane Collaboration Main results  Ten randomized controlled studies (three prevention and seven treatment studies) were included.  Studies varied greatly in the definition of feeding intolerance and how outcomes were measured, analyzed and reported, so meta-analysis of most outcomes was impossible. It was observed, however, that the studies using erythromycin at higher treatment doses (40 to 50 mg/kg/day) or in infants > 32 weeks’ GA reported more positive effects in improving feeding intolerance.  Meta-analysis of high dose prevention studies showed no significant difference in NEC (typical RR 0.59, 95% CI 0.11, 3.01; typical RD-0.021, 95%CI -0.087, 0.045). Meta- analysis of high dose treatment studies showed no significant difference in septicemia (typical RR 0.83, 95% CI 0.47, 1.45; typical RD -0.04, 95% CI -0.17, 0.08). Authors’ conclusions  There is insufficient evidence to recommend the use of erythromycin in low or high doses for preterm infants with or at risk of feeding intolerance.  Future research is needed to determine if there is a more precise dose range where erythromycin might be effective as a prokinetic agent in preterm infants > 32 weeks’ GA.
  • 54.
  • 55.
  • 56.
  • 57.
  • 58. The Role of Nutrition in the Prevention and Management of BPDSeminars in Perinatology 30:200-208
  • 59. Nutritional Intake  Preterm infants with BPD are at risk for undernutrition due to difficulties maintaining appropriate nutritional intake – as a result of co-morbidities (Carlson, 2004) eg swallowing dysfunction, fatigue during feeding, GERD, dysfunction of other organ system eg NEC, prolonged TPN – medically indicated fluid restriction (Carlson, 2004; Oh et al., 2005), – and elevated energy expenditure (Bauer et al., 2003; Leitch & Denne, 2000). – Use of dexamethasone – Anemia of prematurity – Medications eg methyl xanthines and beta sympthomimetics can increase energy consumption  BPD patients can have increased energy expenditure up to 25% above total caloric needs – Increased work of breathing – Higher resting metabolic rates
  • 61. Calories  Breast milk requires HMF, multivitamin and iron supplements  Alternatively use prem formula – Provides additional calories, proteins, calcium, phosphorus and vitamins  Additional fat may be desirable – Increases calories – Less CO2 production than carbohydrates – However, additional fat slows gastric emptying and may worsen GERD  Glucose polymers – Provide additional 4kcal/g – May cause hyperosmolar diarrhoea  Protein supplementation can be considered or provided by using a high caloric formula eg 30 kcal/oz
  • 62. Calcium, Phosphorus and Vit D  Osteopenia of prem more common in BPD – Limited solubility of Ca and phosphorus in TPN – Low enteral intake – Loop diuretics/urinary loss  Provide above via prem formula or fortified BM
  • 63. Nutrients  Vit A – Deficiency associated with impaired clearance of lung secretions, impaired water homeostasis across tracheobronchial epithelium, loss of cilia, diminished lung injury repair and lack of airway distensability – Well documented to result in BPD/death in Vit A deficiency (NICHD study). – Dose of IM 5000 IU per dose 3x/week (12 doses) for ELBW. Oral does not work.  Under study: inositol, Vit E
  • 64. Discharge in BPD  67% continue to have growth failure post discharge  Individualised nutrition plan  Continue 22kcal/oz prem formula for 6-8 months  Low salt/low volume/high calorie diet may be necessary  Achieve solid intake slower than chronological age, often tolerate spoon feeding (thicker consistency) better than nipple (liquid). This may begin at 3-4 mths post gestation
  • 66.
  • 67. Non-nutritive sucking for promoting physiologic stability andnutrition in preterm infants (Review) Copyright © 2008 The Cochrane Collaboration Types of intervention  Non-nutritive sucking involving the use of a pacifier. The intervention occurred before, during or after feeding by a naso/orogastric tube; before or after bottle feeding; or outside of feeding times. Main results  This review consisted of 21 studies, 15 of which were randomized controlled trials.  NNS was found to decrease significantly the length of hospital stay in preterm infants. The review did not reveal a consistent benefit of NNS with respect to other major clinical variables (weight gain, energy intake, heart rate, oxygen saturation, intestinal transit time, age at full oral feeds and behavioral state).  The review identified other positive clinical outcomes of NNS: transition from tube to bottle feeds and better bottle feeding performance.  No negative outcomes were reported in any of the studies. Authors’ conclusions  There were also a number of limitations of the presently available evidence related to the design of the studies, outcome variability, and lack of long-term data.  Based on the available evidence, NNS in preterm infants would appear to have some clinical benefit. It does not appear to have any short-term negative effects.  In view of the fact that there are no long-term data, further investigations are recommended. In order to facilitate meta-analysis of these data, future research in this area should involve outcome measures consistent with those used in previous studies. In addition, published reports should include all relevant data.
  • 68. Early discharge with home support of gavage feeding for stable preterm infants who have not established full oral feeds 2008 Cochrane Collaboration Main results  Data from one quasi-randomised trial with 88 infants from 75 families were included in the review.  Infants in the early discharge program with home gavage feeding had a mean hospital stay that was 9.3 days shorter [MD -9.3 (-18.49 to -0.11)] than infants in the control group. Infants in the early discharge program also had a lower risk of clinical infection during the home gavage period compared with the corresponding time in hospital for the control group [relative risk 0.35 (0.17 to 0.69)].  There were no significant differences between groups in duration and extent of breast feeding, weight gain, re-admission within the first 12 months post discharge from the home gavage program or from hospital, scores reflecting parental satisfaction, or health service use. Authors’ conclusions  Experimental evidence to evaluate the benefits and risks in preterm infants of early discharge from hospital with home gavage feeding compared with later discharge upon attainment of full sucking feeds is limited to the results of one small quasi- randomised controlled trial.  High quality trials with concealed allocation, complete follow-up of all randomised infants and adequate sample size are needed before practice recommendations can be made.
  • 69. Use of transitional formula: the evidence Post discharge Nutrition of Preterm Infants, Journal of Perinatology 2005; 25:S15–S16  feeding of specially designed post discharge formulas has demonstrated better weight (at 9 but not at 18 months) and length (at 9 and 18 months) as well as higher bone mineral content when compared to term infant formula – Lucas A, King F, Bishop NB. Postdischarge formula consumption in infants born preterm. Arch Dis Child 1992;67:691–2. – Lucas A, Fewtrell MS, Morley R, Singhal A, Abbott RA, Isaacs E. Randomized trial of nutrient-enriched formula versus standard formula for post discharge preterm infants. Pediatrics 2001;107:683–9.  the benefit appears restricted to males, a finding similar to that of Cooke et al. – Cooke RJ, Griffin IJ, McCormick K, Wells JC, Smith JS, Robinson SJ. Feeding preterm infants after hospital discharge: effect of dietary manipulation on nutrient intake and growth. Pediatr Res 1998;43:355–60  From the NICHD network, data demonstrate that in virtually all infants less than 1500 g, appropriate-for-gestational-age infants are ‘‘converted’’ to small for-gestational-age infants by 36 weeks postmenstrual age.  Some recovery in these parameters has been reported  Chronic lung disease adds an additional burden as demonstrated by 73% of infants in one study experiencing a decrease in weight z score between hospital discharge and 7 months
  • 70. Use of transitional formula: Guidelines Post discharge Nutrition of Preterm Infants, Journal of Perinatology 2005; 25:S15–S16 Until more optimal strategies are developed, striving toward achieving the best possible gain without adverse effects may be appropriate. Guideline:  <1800 g: 24 kcal/oz preterm infant formula  Transition to 22 kcal/oz at >1800 g if all growth parameters are 25th percentile or greater and infant is gaining 15 to 40 g/day  Transition from 22 kcal/oz to 20 kcal/oz term formula at 4–6 months corrected gestational age if all growth parameters are above 25th percentile
  • 71. Nutrition Practice Care Guidelines for Preterm Infants In the Community (Revised August 2006) Developed by: Child Development and Rehabilitation Center, Nutrition Services Oregon Department of Human Services, Nutrition & Health Screening – WIC Program Oregon Pediatric Nutrition Practice Group
  • 72. Referral Criteria These “red flags” should alert the community nutritionist of the need for further assessment, referral and follow-up Anthropometric “Red Flags”: • Weight loss or significant decline in percentile ranking (“falling away” from expected growth curve percentile) • Poor rate of weight gain for corrected age as listed below: Age Weight Gain term – 3 mos < 20 gm/day (< 5 oz/wk) 3 – 6 mos < 15 gm/day (< 3½ oz/wk) 6 – 9 mos < 10 gm/day (< 2 oz/wk) 9 – 12 mos < 6 gm/day (< 1½ oz/wk) 1 – 2 yrs < 1 kg or < 2 lbs in 6 mos 2 – 5 yrs < 0.7 kg or < 1½ lbs in 6 mos
  • 73.
  • 74.
  • 75.
  • 76.
  • 77. VITAMIN-MINERAL SUPPLEMENTATION  Vitamins: Supplementation with a standard infant multivitamin (with vitamins A, D, B1, B2, B3, B6, B12, C, and iron) is generally needed initially after NICU discharge to meet the preterm infant’s vitamin needs, until the infant is consuming larger volumes of feeds.  Vitamin D: Per the American Academy of Pediatrics (AAP), all infants fed unfortified breast milk should continue to receive a supplement of 200 IU of Vitamin D for the first year. – This 200 IU of Vitamin D can be provided by continuing the 0.5 ml daily of the standard infant multivitamin, or by changing to 0.5 ml daily of a tri-vitamin supplement (vitamins A, C, and D). Infants receiving 17 oz. (500 ml) or more of a vitamin D-fortified infant formula do not need any additional Vitamin D supplementation.  Iron: Preterm infants have lower iron stores than term infants. By 2 months post birth (not 2 months corrected age), preterm infants should have an intake of 2-4 mg iron/kg/day (up to a maximum of 40 mg/day) from an iron-fortified infant formula and/or supplement. This iron dose should be continued for the first year of life.
  • 78.
  • 79.
  • 80.
  • 81.
  • 82.
  • 83.
  • 84.
  • 85.
  • 86.
  • 87. Osteopenia of prematurity is most commonly seen in: • Very-low-birth-weight infants (BW < 1500g). • Any IUGR infant with a birthweight < 1800 g regardless of gestational age. • Infants with chronic lung disease/bronchopulmonary dysplasia. • Infants requiring long-term parenteral nutrition at birth. • Infants on certain medications, including diuretics & corticosteroids, that affect mineral absorption. • Infants starting feedings of unfortified breastmilk or standard formula too early, or soy formula. Recommendation for checking labs: • 1-month post-discharge for the infants born < 1500g and IUGR with birthweight < 1800 g. • 1-month post-discharge if any of the labs at discharge (if known) are outside the reference range. • If the premie is transitioning to the breast or a term formula < 3-6 months corrected age. • If the premie has had marginal intake and slower growth. Bone health can be assessed using the following labs. •In the absence of other disease conditions, alkaline phosphatase provides an indirect indicator of bone cell activity. •Higher alkaline phosphatase along with lower calcium and phosphorus levels may indicate a need for further assessment and supplementation. These labs are usually done in the hospital setting, and may be done in the community up until 6-9 months corrected age.
  • 88.
  • 89.
  • 90.
  • 91.
  • 92.
  • 93.
  • 94.
  • 95.
  • 96.
  • 97. One last example to put it all together … the ADHB (Auckland District Health Board) guidelines
  • 98.
  • 99.
  • 100.
  • 101. More questions than answers? Sighhh…
  • 102. More questions than answers? Sighhh…
  • 103.
  • 104. Nutrition Practice Care Guidelines for Preterm Infants In the Community. Revised August 2006. Developed by Child Development and Rehabilitation Center, Nutrition Services, Oregon Department of Human Services, Nutrition & Health Screening – WIC Program. Oregon Pediatric Nutrition Practice Group APPENDIX E: SELECTION OF FEEDING AT DISCHARGE Feeding strategies for the preterm infant need to be evaluated on an individual basis. These guidelines are designed to help the practitioner in making feeding selections to promote optimal nutrition. Birth weight, weight at discharge, and NICU course are usually better predictors of risk than gestational age at birth. Breastfeeding after premature birth is recommended and encouraged whenever possible. Lactation consultation is encouraged to promote successful breastfeeding and use of a breast pump if needed. 1. High Risk – Very Low Birth Weight Infant Category Definition: • Birth weight < 1500gm (3.3 lbs) • History of TPN and diuretics • Demonstrates poor growth • Poor intake (< 150 ml/kg/day) • Elevated alkaline phosphatase (> 500 U/L) and/or low phosphorus (< 4) Formula Feeding Recommendations: • In majority of cases, these infants will need transitional formulas (EnfaCare or Neosure) until 9 months corrected age. If change to a term formula, check labs and monitor growth. • Continue on transitional formula unless: o Infant cannot tolerate formula o Excessive rate of weight gain o Calcium and Phosphorus exceed normal limits Breastfeeding Recommendations: • Supplement breastfeeding with a transitional formula (Similac Neosure or Enfamil EnfaCare) until infant able to sustain growth, ad lib milk intake, and lab values are within normal limits • Discuss family’s breastfeeding goals in order to support breastfeeding while still maintaining infant’s growth and lab values.
  • 105. Moderate Risk – Low Birth Weight Infant Category Definition: • Infant had BW > 1500 gm (3.3 lbs), has good growth, use of TPN and diuretics was minimal and alkaline phosphatase level WNL • Osteopenia evidenced by serum phosphorous <4mg/dl, alkaline phosphatase > 500 U/L • Weight for corrected age is less than 5th percentile on CDC 2000 growth grids • Infant has bronchopulmonary dysplasia/chronic lung disease with steroid use Formula Feeding Recommendations: • Provide transitional formulas (EnfaCare or Neosure) to 9 months corrected age. • Continue on transitional formula unless: o Infant cannot tolerate formula o Excessive rate of weight gain o Calcium and Phosphorus exceed normal limits o If change to a term formula, check labs and monitor growth Breastfeeding Recommendations: • Supplement breastfeeding with a transitional formula (Similac Neosure or Enfamil EnfaCare) until infant able to sustain growth, ad lib milk intake, and lab values are within normal limits • Discuss family’s breastfeeding goals in order to support breastfeeding while still maintaining infant’s growth and lab values.
  • 106. Low Risk – Low Birth Weight Infant Breastfeeding Recommendations: • Breastfeed on demand. • If milk intake or supply is insufficient, as evidenced by slow growth on CDC growth grid, assess the need for lactation support or supplementation with standard formula such as Enfamil Lipil or Similac Advance. Formula Feeding Recommendations: • Offer standard 20 kcal/oz iron fortified infant formula such as Enfamil Lipil or Similac Advance until 1 year corrected age. Category Definition: • Birth weight > 2000 gm (4.4 lbs)