You are on page 1of 245

1

AIIMS PROTOCOL

NAME: _________________________________________________________________________
2

CONTENT
3 1

Symposium on AIIMS Protocols in Neonatology IV

Acute Renal Failure in Neonates


Sreeram Subramanian, Ramesh Agarwal, Ashok K. Deorari, Vinod K. Paul and Arvind Bagga1

Department of Pediatrics, Divisions of Neonatology, 1Nephrology, All India Institute of Medical Sciences, Ansari
Nagar, New Delhi, India

ABSTRACT
Acute renal failure (ARF) is a common condition seen in neonatal intensive care units. It is broadly classified into prerenal,
intrinsic renal and post renal failure. There is no consensus on the definition of neonatal ARF. Of utmost importance is to
differentiate prerenal from intrinsic renal failure. The most common causes of neonatal ARF are hypovolemia, hypotension
and, hypoxia. Among several indices that are available for differentiating prerenal failure from intrinsic renal failure, fractional
excretion of sodium is the preferred index. Diagnostic fluid challenge with or without frusemide is a bed side method for
differentiating prerenal failure from intrinsic renal failure. Babies with ARF have to be monitored for several metabolic
derangements like hyponatremia, hyperkalemia, hypocalcemia, and acidosis and have to be managed accordingly. Fluid
balance should be precise in order to avoid fluid overload. It is difficult to provide adequate calories due to fluid restriction.
Dialysis has to be instituted to preempt complications. Peritoneal dialysis is the easiest and safest modality. These babies need
long term follow up as they are prone for long term complications. [Indian J Pediatr 2008; 75 (4) : 385-391] E-mail :
arvindbagga@hotmail.com

Key words : Acute renal failure; Neonate, Hyperkalemia; Dialysis

Acute renal failure (ARF) is a frequent clinical condition in DIAGNOSIS OF ARF


neonatal intensive care unit. There is wide variation in the
incidence of ARF across studies. It affects approximately Plasma creatinine
1-24% of newborns in the NICU. 1,2 ARF is an acute
reduction in glomerular filtration rate (GFR) with both ARF is suspected if
failure to remove solutes and water leading to concurrent Plasma creatinine is more than 1.5 mg/dL for at least 24
net solute and water retention oligoanuric renal to 48 hrs, if mothers renal function is normal.2
failure. 2
Serum creatinine is increasing by 0.3 mg/dL/day
Classification of ARF
Serum creatinine fails to fall below maternal plasma
Based on the urine output, it can be of two types: creatinine within 5-7 days
(i) Oligoanuric (ii) Non oliguric The above definitions can be used with a reasonable
Based on the site of origin of insult it can be of 3 types: 3 degree of accuracy in term neonates. In preterm neonates,
the physiological decline in plasma creatinine can extend
(i) Prerenal (75- 80%) (ii) Intrinsic renal (10-15%) (iii) over 2-3 weeks. In fact the plasma creatinine can rise
Post renal (5%). transiently and then decline. The plasma creatinine
remains elevated due to reabsorption of creatinine across
Persistence of insult can convert pre renal or post renal
permeable tubules.
failure to intrinsic renal failure.
Urine output
Oliguria: It has been defined as urine output less than 1
mL/kg/h after first day of life for both term and preterm
neonates. One should also be aware of the fact that some
term neonates may void for the first time at around 24 h
Correspondence and Reprint requests : Prof Arvind Bagga,
Division of Nephrology, Department of Pediatrics, All India of life.
Institute of Medical Sciences, Ansari Nagar, New Delhi -110029, ARF can also present with normal renal output in one
India.
third of the cases. This can happen especially in
[Received March 28, 2008; Accepted March 28, 2008]

Indian Journal of Pediatrics, Volume 75April, 2008 385


4 2

S. Subramanian et al

asphyxiated neonates. Hence it is essential to monitor proposed to differentiate them. Most important among
plasma creatinine apart from urine output. The common them would be urine sodium, renal failure index (RFI)
clinical scenario that leads to suspicion of renal failure is and fractional excretion of sodium (FENa) (Table 1). The
oliguria. In face of such an event it becomes extremely important prerequisite is that the urine sample for
important to differentiate prerenal and intrinsic renal measuring indices must be obtained prior to fluid and
failure as in the former the damage to the kidneys is yet to diuretic challenge. This is difficult to obtain in many
begin where as in the later it already has. babies as the babies are oliguric and results are not
available immediately and hence practically they are of
Concept of acute kidney injury (AKI)4 limited utility. Among the various indices available FENa
is the preferred index. FENa more than 2.5 to 3.0% is
Several definitions have been proposed for defining ARF
found to be associated with intrinsic ARF. Preterm babies
and there is no consensus. An attempt has been made to
lose sodium in the urine due to the tubular immaturity,
define parameters and to bring uniformity across age
hence higher cutoffs must be used. An FENa of more than
groups and various clinical situations. The product of
6% can be used to define intrinsic ARF in babies born
such an attempt is the concept of acute kidney injury.
between 29- 32 weeks of gestation.5 Urine sodium more
An abrupt (within 48 h) reduction in kidney function than 50 mEq/L is suggestive of intrinsic ARF whereas
currently defined as an absolute increase in serum urine sodium less than 20 mEq/L is seen in pre renal
creatinine of more than or equal to 0.3 mg/dL, a failure.
percentage increase in serum creatinine of more than or
equal to 50% (1.5-fold from baseline), or a reduction in TABLE 1. Parameters to Differentiate Prerenal from Intrinsic
urine output (documented oliguria of less than 0.5 mL/ Renal Failure1
Kg/h for more than six hours).
Parameters Prerenal Intrinsic renal
Pre renal vs intrinsic renal failure UNa 20 mEq/L >50
Renal failure index* Low < 1 High > 4
Several methods have been developed to differentiate Fractional excretion of Na$ 1 >3
them; the sheer number reflects the importance. When a
baby has not passed urine in the past 12 hrs, the first and
the foremost thing is to look for distended bladder. urine Na plasma creatinine
Palpation of the abdomen, ultrasound of the abdomen (if *Renal failure index = 100
urine creatinine
available at bed side) can be employed to look for
distended bladder. It is better to avoid catheterization of urine Na plasma creatinine
the bladder in order to prevent infection but it may be $Fractional excretion = 100
necessary in sick babies. In such situations it has to be of sodium plasma sodium urine creatinine
done under strict asepsis. Compression of the bladder
(suprapubic pressure) should be avoided especially in
preterm infants for the fear of VUR and rarely bladder The renal failure index (RFI) can also be used. RFI
and renal rupture. 1 more than 4 in term and more than 8 in preterm babies
less than 32 weeks is suggestive of intrinsic ARF.
After confirming the absence of urine in the bladder,
fluid challenge should be given (Fig. 1). The common Urianalysis: The presence of granular casts, hyaline
causes of prerenal azotemia are hypovolemia, systemic casts, RBC, proteins and tubular cells suggests an
hypotension and hypoxia (in more than 80% of cases). 2 It intrinsic cause.
is essential to look for signs of fluid excess and fluid Ultrasonography and Doppler: Useful in ruling out
deficit. In the absence of obvious sign of fluid overload or congenital anomalies like polycystic kidneys, dysplasia of
congestive cardiac failure, a normal saline bolus of 10 kidneys and obstructive causes of renal failure like
mL/kg should be given over 20 min (some experts advise posterior urethral valves. Renal doppler studies are
20 mL/kg over 2 h). If baby fails to pass urine with in one helpful in diagnosing vascular thrombosis.
hour the fluid bolus should be repeated. In spite of two
fluid bolus if urine output fails to ensue, frusemide Voiding cystourethrography : Can identify lesions of
should be given in a single dose of l mg/kg (in a non the lower urinary tract that cause obstruction, such as
dehydrated patient). Urine output ensues in 2-3 hrs in posterior urethral valves.
prerenal failure. If this fails it is intrinsic renal failure.
Etiology of renal failure
Role of indices
Having differentiated prerenal from intrinsic renal failure,
Differentiation of pre renal and intrinsic renal can be done evaluate for the exact etiology of renal failure. There are
basing on urinary indices. Several indices have been several causes of ARF (Table 2)

386 Indian Journal of Pediatrics, Volume 75April, 2008


5 3

Acute Renal Failure in Neonates

TABLE 2. Etiology of Neonatal Renal Failure the kidneys is useful in evaluating congenital lesions and
obstructions. Doppler can define the vascular supply of
I. Congenital malformations
 Renal agenesis the kidney.
 Renal hypoplasia/dysplasia
 Cystic diseases of kidney e.g., autosomal recessive poly- MANAGEMENT OF RENAL FAILURE
cystic kidney
II. Acquired renal disorders Fluid management
Acute tubular necrosis
 Perinatal asphyxia Fluids must be restricted to insensible water loss (IWL)
 Perinatal hypoxia due to respiratory distress syndrome, along with urinary loss. The urinary loss must be replaced
traumatic delivery volume for volume. The insensible water loss in a term
 Sepsis neonate is 25 mL/kg/day. In preterm neonates this can
 Hypovolemia due to dehydration, severe patent ductus
vary widely depending on gestation, postnatal age, use of
arteriousus
Vascular radiant warmers, phototherapy. It can vary from 40-100
 Arterial thrombosis or embolism or stenosis mL/kg/day. IWL can be assumed to be 40 mL/kg/day in
 Venous thrombosis preterm infants for calculating fluids in neonates
Drugs: maternal use of ACE inhibitors, indomethacin (adequate care must be taken to reduce IWL by using
Baby: indomethacin, tolazoline, aminoglycosides
caps, socks, cling wrap, oil especially for preterm babies
III. Urinary tract obstruction under radiant warmer). 6 It is advisable to revise fluid
Posterior urethral valves requirement every eight hourly basing on urine output.
Pelviureteric obstruction, ureterovesical obstruction The fluid should be electrolyte free, 10% dextrose water.
ACE: angiotensin converting enzyme.
Hyponatremia
Babies with ARF must be investigated not only for
evaluating the cause and but also for complications. Blood Babies can have hyponatremia in oliguric renal failure.
levels of creatinine urea, electrolytes, pH bicarbonate and Hyponatremia is due to dilution secondary to water
urinary levels of sodium and creatinine must be done. retention, hence has to be corrected with fluid
Microscopic examination of urine must be done for RBC, restriction. In most of the cases, there is no sodium
granular or hyaline casts. Urine culture must be done deficit.
especially in cases of obstructive lesions where babies are
prone for urinary tract infection. Ultrasound imaging of If serum sodium is between 120-135 mEq/L,

TABLE 3. Management of Hyperkalemia

Medication Level of K+ at which it Dose Mechanism Onset of action


is instituted

Calcium ECG changes suggestive of 0.5 to 1 mL/kg over 5-10 min Modifies myocardial 5-10 min
gluconate hyperkalemia excitability
Sodium K+ - 6.0-6.5 mEq/L 1 mEq/kg over 10-30 min Intracellular uptake 30 min
bicarbonate of K+
Glucose and K+ - 6.5-7.5 mEq/L 0.5 g/kg/h of glucose and 0.2 U of Intracellular uptake 30 min.
insulin regular insulin per g of glucose of K+
over 2 h
Salbutamol IV K+ - 6.5-7.5 mEq/L 4 g/kg over 20 min Intracellular uptake of Min
infusion* K+
Cation exchange K+ more than 6.0 mEq/L 1 g/kg intrarectally q 6 h Exchange of K for Na 1-2 h
resin (Na/Ca or Ca2+ .
polystyrene
sulfonate)**
Exchange K+ more than 7.5 mEq/L Washed RBC reconstituted with 5% Uptake of K + by RBC. Minutes
transfusion albumin
Peritoneal K+ more than 7.5 mEq/L Use a dialysate with low K + Dialysis Minutes
dialysis concentration

*Administration of salbutamol can cause a transient increase in serum K concentration, so it should not be used as the first line
medication. Salbutamol aerosol is not very effective in neonates.
**Oral administration of polystyrene resin should be avoided in VLBW infants and those with poor peristalsis (gastric bezoars after
oral administration and cecal perforation after enema, other complications like hypernatremia, fluid retention can occur)

Indian Journal of Pediatrics, Volume 75April, 2008 387


6 4

S. Subramanian et al

restriction of fluids will suffice. serum sodium must indomethacin.9 At present low dose dopamine does not
be monitored at least 12 hourly. seem to have any role in the prevention or treatment of
ARF except as on intrope in patients with hypotension or
If hyponatremia is associated with symptoms like
congestive cardiac failure.
seizures, or if hyponatremia is less than 120 mEq/L
it requires prompt correction with 3% hypertonic Role of theophylline
saline at a dose of 5 mL/kg over 4-5 h.
Adenosine antagonists are able to reverse the intra-renal
Hyponatremia unresponsive to above therapy is an
vasoconstrictor state of ARF. Low dose theophylline (0.5-
indication for dialysis.
1mg/kg) has been shown to prevent hypoxia induced
Babies with non-oliguric ARF may have very large renal insufficiency in newborn rabbits.10 The mechanism
urinary sodium losses of up to 10 mEq/kg/day, and is adenosine antagonism and not by cyclic AMP
these must be replaced. phosphodiesterase antagonism. In vasomotor
Hyperkalemia nephropathy of very preterm infants with respiratory
distress syndrome, early theophylline administration
Hyperkalemia (K+ more than 6.0 mEq/L) is one of the improves renal function during the first two days of life.11
most dangerous complications that develops in babies Prophylactic theophylline, given early after birth, has
with ARF. Extremely low birth weight (ELBW) babies are beneficial effects on reducing the renal dysfunction in
at higher risk of hyperkalemia. The reasons are asphyxiated full-term infants. 12 Thus theophylline may
multifactorial. Reduction in glomerular filtration rate, have role in the management of renal dysfunction but
urinary potassium secretion, acidosis, immature tubular data are limited, further studies are needed. Presently it
response to aldosterone all contribute to the development has no role in the management of ARF.
of hyperkalemia.
Nutrition
The first step in the management of hyperkalemia is to
stop administration of potassium in the fluids; various The goal is to provide 100 kcal/kg/day. Proteins or
medications are available to reverse dangerous amino acids are provided in a dose of 1-2 g/kg/day.13
hyperkalemia. ECG will help in diagnosing cardiac effects Total parenteral nutrition can be provided if enteral
of hyperkalemia (Table 3). If ECG changes are evident nutrition cannot be established. If enteral feeding is
calcium gluconate 10% is given. This will decrease the possible, breast milk should be used. Caloric density can
myocardial excitability but will not lower the potassium be increased by adding corn oil, medium chain
levels. This should immediately be followed by methods triglycerides or maltodextrins. If breast milk cannot be
to decrease the potassium levels. Hyperkalemia which is given low phosphate formula milk with low renal solute
unresponsive to medications is one of the most common load can be given.
indications for instituting dialysis.
Acidosis
Hypocalcemia Mild metabolic acidosis is common in babies with ARF. If
pH is less than 7.2 sodium bicarbonate can be used for
Hypocalcemia can develop in babies with ARF. It may
correction of acidosis. It is given in a dose of 1-2 mEq/kg
result from hyperphosphatemia and skeletal resistance to
over 3-4 hrs. But this should be done carefully as it can
parathyroid hormone. Symptomatic hypocalcemia should
cause fluid overload, hypernatremia, intracranial
be corrected by infusing 10% calcium gluconate at a dose
hemorrhage and intracellular acidosis. Babies with
of 0.5-1 mL/kg over 5-10 min under cardiac monitoring.
persistent acidosis require dialysis.
Role of dopamine Hypertension
Renal blood flow increases with low dose of dopamine; Fluid overload in neonatal ARF can result in mild
action is via DA1 and DA2 receptors. There is a definite role hypertension, which can be controlled with fluid
of dopamine in babies who are hypotensive, who are in restriction and antihypertensive agents. The development
congestive cardiac failure, as these babies will need of severe hypertension in the setting of neonatal ARF
inotropic and vasoactive support. Preterm infants are should raise the suspicion for renal artery or venous
hypersensitive to alpha receptors and hence even low thrombosis.
doses of dopamine can cause vasoconstriction and raise
renal vascular resistance.7 This may explain the difficulty Drug dosage modification
in dosing of dopamine for improving renal function. Dosage of the drugs has to be modified in babies with
Dopamine when combined with frusemide has been ARF as the major pathway of excretion of several drugs is
shown to cause natiruresis and diuresis in preterm infants kidneys. In ARF such drugs can accumulate and can
RDS and oliguria.8 Cochrane review concluded that cause toxicity. For modifying drug dosage, the creatinine
dopamine has no role in the management of ARF due to clearance has to be calculated.

388 Indian Journal of Pediatrics, Volume 75April, 2008


7 5

Acute Renal Failure in Neonates

Oliguria : urine output < 1 ml/kg/h for the past 12 hrs in a baby more than 24 hrs of age

Assess urinary bladder size by clinical or


bedside USG if available
Assess and correct hydration
Check for any underlying condition
predisposing to ARF like hypotension,
hypoxia, and hypovolemia.
Estimate creatinine and sodium in blood and
urine
No evidence of CCF

Normal saline bolus 20 ml/kg over 2 h

Urine output present < 1 mL/kg/h


Urine output present >1 mL/kg/h

IV Frusemide
1 mg/kg stat

UOP UOP
< /mL/kg/h > 1 mL/kg/h

INTRINSIC RENAL FAILURE PRE RENAL FAILURE

Fig. 1. Evaluation of Baby with Oliguria. UOP : Urine output CCF : congestive cardiac failure

k length in cm for renal replacement therapy are fluid overload,


Creatinine clearance = hyperkalemia, hyponatremia and severe metabolic
Serum creatinine
acidosis which are unresponsive to medical management.
where k in Dialysis has to be instituted to preempt complications in
Term babies is 0.44 renal failure. A newborn who is anuric and is having
Preterm babies is 0.33 metabolic complications will ultimately require dialysis
(e.g., hyperkalemia in anuric baby is unlikely to respond
Basing on creatinine clearance the dosage of the drugs
to medical management alone and require dialysis).
have to be modified.
Dialysis and filtration techniques are the available
Renal replacement therapy
modalities. Dialysis is a process of removal of plasma
Before instituting dialysis, it is always better to consider solutes by diffusion down their concentration gradients
the prognosis of the condition. The common indications across a semi permeable membrane. The membrane may

Indian Journal of Pediatrics, Volume 75April, 2008 389


8 6

S. Subramanian et al

be a synthetic one (hemodialysis) or peritoneum Hyperglycemia can occur due to absorption of dextrose
separating the splanchnic blood from fluid instilled into from PD fluid especially in cases where higher
the peritoneal space (peritoneal dialysis).14 Filtration concentrations of dextrose are used. Bleeding, perforation
involves removal of protein free plasma water across a of abdominal viscera, peritonitis, adhesion of catheter tip
membrane by convection. The filtered water contains to omentum (Be careful while removing catheter or else
other plasma solutes at a concentration similar to plasma will be delighted to see omentum!) PD cannot be done in
and can be thought of as glomerular filtrate equivalent. babies with necrotizing enterocolitis, babies who
Hemodiafiltration involves both dialysis and filtration. underwent abdominal surgery and in those with severe
respiratory compromise as it may worsen with abdominal
PD has major advantages as the access is relatively
distension.
easy and is technically simple. Peritoneal dialysis has to
be done only under strict aseptic conditions. Hemofiltration and hemodiafiltration are effective in
neonates with ARF in whom PD is contraindicated. The
Peritoneal dialysis catheters: 15 PD catheters are made up of
complication rates are less. Hemofiltration is particularly
soft silastic, which is smooth silicone polymer of methyl-
useful in the presence of fluid overload.
silicate, either in curled or straight configurations. Most of
Hemodiafiltration is more useful in the presence of fluid
the catheters have side holes that allow for easy ingress
overload and azotemia with electrolyte disturbances. 2
and egress of fluid regardless of the catheter position in
the peritoneum. Permanent catheters have cuffs. Pig-tail Outcome
catheters and straight catheters without cuffs have been
used in neonates who are anticipated to need PD access Non oliguric renal failure has a better prognosis when
for a brief period of time. Straight Tenckhoff and coiled compared to oliguric renal failure. Mortality ranges from
Tenckhoff catheters are available. Coiled Tenckhoff 25 to 78% in oligoanuric renal failure. 16 Long term
catheters are useful for chronic dialysis. abnormalities in GFR and tubular function are common in
babies who survive ARF and is secondary to hyper-
Procedure
filtration in the surviving nephrons. The long term
The catheter is inserted into the peritoneal cavity and consequence of such an acute insult is not known.
connected to a three way cannula. The common sites of
Follow up
insertion are in the midline below the umbilicus, right or
left lower quadrant of the abdomen. Urinary bladder All babies who develop ARF need follow up. Adequacy
must be emptied before insertion of the catheter. The of growth and nutrition, blood pressure, and renal
dialysate fluid is connected to a pediatric burette set and function status has to be monitored. Newborns who have
its terminal end is connected to one of the ports of three ARF are predisposed to the development of chronic renal
way cannula. The remaining port of the three way is failure in the future. Long-term follow-up of extremely
connected to a intravenous (IV) set, the end of which is let low birth weight infants who had neonatal ARF has
into a sterile container (empty IV fluid bottle). The shown that prominent risk factors for progression of renal
abdomen is distended with 20 mL/kg of peritoneal disease at 1 year of age included a spot urinary protein/
dialysis fluid. 20-30 mL/kg of dialysis fluid is infused creatinine ratio of greater than 0.6 and serum creatinine
over 10 min. A dwell time of 20-30 min is used before greater than 0.6 mg/dL.17
draining the fluid over 10 min. The dwell time can be
reduced in case of respiratory compromise. A total of 20-
40 cycles can be used or it can be continued till the desired REFERENCES
effect is obtained. Blood sugar, serum electrolytes have to
be monitored every 6 hourly and serum creatinine every 1. Suhas M Nafday et al. In MG MacDonald, eds. Renal Disease
24 hourly. Averys Neonatology pathophysiology and management of
newborn, 6th ed. Lippincott Williams and Wilkins. 2005; 981-
The common dialysate fluid contains 1.7% dextrose 1065.
with lactate. If higher gradient is required as in case of 2. Gouyon J B, Guignard J P, Management of acute renal failure
in newborns. Pediatr Nephrol 2000; 14 : 1037-1044.
fluid overload 3% solution can be used. This can be 3. Hentschel R, Lodige B, Bulla M. Renal insufficiency in the
prepared by adding 25 mL of 50% dextrose to one liter of neonatal period. Clin Nephrol 1996; 46 : 54-58.
1.7% PD fluid. In case of liver failure lactate free 4. Ravindra LM, John AK, Sudhir VS, Bruce AM, Claudio R,
bicarbonate containing fluid has to be used. If baby David GW et al. Acute Kidney Injury Network: report of an
becomes hypokalemic during the procedure, add 1.5 mL initiative to improve outcomes in acute kidney injury. Critical
Care 2007; 11 : R31.
of KCl to one liter of dialysate fluid. At the end of the
5. Ishizaki Y et al. Evaluation of diagnostic criteria of acute renal
procedure the catheter can be removed and the tip and failure in premature infants. Acta Paediatr Jpn 1983; 35: 311-
the fluid are sent for culture. 315.
6. Chawla D, Agarwal R, Deorari AK, Paul VK. Fluid and
PD is invasive procedure and complications can occur. electrolyte management in term and preterm neonates.

390 Indian Journal of Pediatrics, Volume 75April, 2008


9 7

Acute Renal Failure in Neonates

AIIMS-NICU protocols 2008, www.newbornwhocc.org. syndrome. Intensive Care Med 1995; 21 : 511-514.
7. Seri I et al. Effects of low dose dopamine infusion on 12. Jenik AG, Ceriani Cernadas JM, Gorenstein A, Ramirez JA,
cardiovascular and renal functions cerebral blood flow and Vain N, Armadans M et al. A randomized, double-blind,
plasma cathecolamines levels in sick preterm neonates. placebo-controlled trial of the effects of prophylactic
Pediatric Res 1993; 34 : 742-749. theophylline on renal function in term neonates with perinatal
8. Tulassy T, Seri I. Acute oliguria in preterm infants with asphyxia. Pediatrics 2000; 105 : E45.
hyaline membrane disease; interaction of dopamine and 13. Philippe SF Jacquelyyn RE, Tivadar T, Seri I. In William T,
frusemide. Acta Pediatr Scand 1986; 75 : 420-424. Roberta B, Christine AG, eds. Acute and chronic renal failure,
9. Barrington K, Brion LP. Dopamine versus no treatment to Averys diseases of newborn, 8th ed. Saunders, 2005; 1298-1306.
prevent renal dysfunction in indomethacin-treated preterm 14. Coulthard M G, Brayan V, Managing acute renal failure in
newborn infants. Cochrane Database of Systematic Reviews very low birthweight infants. Arch Dis Child 1995; 73: F187-
2002, Issue 3. Art. No.:CD003213. F192.
10. Toth-Heyn P, Drukker A, Guignard J P, The stressed neonatal 15. Marsha ML, Annabelle NC, Peter DY. Neonatal peritoneal
kidney; from pathophysiology to clinical management of dialysis. Neo Reviews 2005; 6 : e384-e391.
neonatal vasomotor nephropathy. Pediatr Nephrol 2000, 14 : 16. Chevalier R. Prognostic factors in neonatal acute real failure.
227-239. Pediatrics 1984; 74: 165-272.
11. Huet F, Semama D, Guignard JP et al. Effect pf theophylline on 17. Annabelle NC, Minnie MS. Acute renal failure management in
renal insufficiency in neonates with respiratory distress the neonate. Neo Reviews 2005, 6: e369-e376.

Indian Journal of Pediatrics, Volume 75April, 2008 391


10
11
12
13
14 8

Symposium on AIIMS Protocols in Neonatology 1

Apnea in the Newborn


Satish Mishra, Ramesh Agarwal, M. Jeevasankar, Rajiv Aggarwal1, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, 1Narayana Hridalaya, Bangalore, Karnataka, India

ABSTRACT
Apnea, defined as cessation of breathing resulting in pathological changes in heart rate and oxygen saturation, is a common
occurrence especially in preterm neonates. It is due to immaturity of the central nervous system (apnea of prematurity) or
secondary to other causes such as metabolic disturbances etc. Secondary causes of apnea should be excluded before a
diagnosis of apnea of prematurity is made. Methylxanthines and continuous positive airway pressure form the mainstay of
treatment. Mechanical ventilation is reserved for apnea resistant to the above therapy. An approach to the management of apnea
in neonates is described. [Indian J Pediatr 2008; 75 (1) : 57-61] E-mail: vinodkpaul@hotmail.com

Key words : Apnea; Methylxanthines

About 30-45% of preterm babies exhibit a periodic usually presents after 1-2 days of life (the detection may
breathing pattern characterized by 3 or more respiratory be delayed by the presence of ventilatory support in the
pauses of greater than 3 seconds duration. Periodic initial days of life) and within the first 7 days.
breathing is a normal event, reflective of immaturity of
Secondary causes: Secondary causes of apnea
respiratory control system in these infants and does not
include: (a) Temperature instability: hypothermia and
merit any treatment. In contrast, apnea is a pathological
hyperthermia, (b) Neurological: birth trauma, drugs,
cessation of breathing that results in hemodynamic
intracranial infections, intracranial hemorrhage, seizures,
disturbances and hence merits treatment.
perinatal asphyxia, congenital myopathies or
Definition neuropathies, placental transfer of narcotics, magnesium
sulphate, or general anesthetics, central nervous system
Apnea is defined as cessation of respiration for longer
malformations, (c) Pulmonary: respiratory distress
than 20 sec, or shorter duration in presence of cyanosis or
syndrome (RDS), pneumonia, pulmonary hemorrhage,
bradycardia.1
obstructive airway lesion, pneumothorax, hypoxemia,
Incidence hypercarbia, tracheal occlusion by neck flexion, (d)
Cardiac: congenital cyanotic heart disease, hypo/
Apnea in preterm infants is usually related to immaturity
hypertension, congestive heart failure, patent ductus
of the central nervous system and is called apnea of
arteriosus, increased vagal tone (e) Gastro-intestinal:
prematurity (AOP). It can also occur secondary to other
gastro esophageal reflux, abdominal distension, (f)
causes and is a common manifestation of most neonatal
Hematological: anemia, (g) Infections: sepsis, pneumonia,
diseases. Incidence of AOP is inversely proportional to
meningitis, necrotizing enterocolitis, (h) Metabolic:
gestational age. It varies from 10% in infants born at
acidosis, hypoglycemia, hypocalcemia, hyponatremia,
gestation of 34 weeks or more to more than 60% in infants
hypernatremia and, (i) Inborn errors of metabolism.
born at less than 28 weeks of gestation.1
AOP is a diagnosis of exclusion and should be
ETIOLOGY OF APNEA2,3 considered only after secondary causes of apnea have
been excluded. Common causes of secondary apnea
Apnea of prematurity(AOP). It is probably related to include sepsis, pneumonia, asphyxia, temperature
immaturity of the central nervous system. This condition instability and anemia.

TYPES OF APNEA4
Correspondence and Reprint requests : Dr. Vinod K Paul, Professor,
Department of Pediatrics, All India Institute of Medical Sciences, Central apnea: (40%) Central apnea is characterized by
Ansari Nagar, New Delhi-110029, India
total cessation of inspiratory efforts with no evidence of
[Received November 3, 2007; Accepted December 3, 2007] obstruction.

Indian Journal of Pediatrics, Volume 75January, 2008 57


15 9

S. Mishra et al

Obstructive apnea. (10%) In obstructed apnea, the of heart rate or color.


infant tries to breathe against an obstructed upper airway,
Subtle seizures: Apnea is an uncommon presentation of
resulting in chest wall motion without airflow throughout
a neonatal seizure in preterm infants. Sudden alteration
the entire apneic episode
in muscle tone, twitching movements, vacant stare and
Mixed apnea. (50%) Mixed apnea consists of up rolling of eyes suggests a seizure. Also tachycardia
obstructed respiratory efforts usually following central preceding/ accompanying an apneic attack usually
pauses. Purely obstructive apnea in the absence of a suggests seizure activity.
positional problem is probably uncommon.
EVALUATION OF A CHILD WITH APNEA
MONITORING
Emergency treatment
The neonate should be checked for bradycardia, cyanosis
All babies less than 34 weeks gestation should be
and airway obstruction. The neck should be positioned in
monitored for at least in the first week of life or till
slight extension; oro-pharynx gently suctioned, if
absence of apneic episodes for at least 7 days. Babies 34
required and tactile stimulation should be given. Most
weeks gestation should be monitored if they are sick.3
apneic spells respond to tactile stimulation. Oxygen by
Apnea Monitors5 head box or nasal cannula is provided if the infant is
hypoxic (maintain saturation between 90-93%). If the
Pulse oximeters: These are commonly used for
neonate continues to remain apneic and does not respond
monitoring of apnea. These monitors detect changes in
to tactile stimulation, ventilation with bag and mask
heart rate and/or saturation due to apneic episodes.
(BMV) using 100% oxygen should be initiated. If BMV
Facility for detecting chest wall movement is absent in
fails to initiate spontaneous respiration in the newborn,
these monitors.
then the infant should be managed with positive pressure
Other apnea monitors ventilation.
Movement sensors Clinical examination
(i) Ripple type mattress
After stabilization, the infant should be evaluated for a
(ii) Mattress with sensory pad
possible underlying cause. History should be reviewed
(iii) Pressure sensitive capsule
for possible causes of secondary apnea including
These monitors interpret chest/abdominal movements perinatal asphyxia, maternal drugs, neonatal sepsis and
as respiration. In general, these monitors will fail to feed intolerance. The infant should be examined for
diagnose obstructive apnea and may not distinguish temperature instability, hypotension, jaundice, pallor,
body movements from breathing. cardiac murmur for PDA and poor perfusion. Onset of
apnea within the first 7 days in a premature infant
Thoracic impedence based monitors: These monitors (gestation < 34 weeks) would be suggestive of apnea of
translate changes in thoracic impedence that occur with prematurity (AOP).
breathing as respiratory activity. Like the movement
sensors, these monitors also fail to diagnose obstructive Investigations
apnea.
Neonates with apnea should be investigated to exclude
Respiratory inductive plethysmography: It uses common causes of secondary apnea. Investigations that
abdominal and thoracic movements during respiration. should be considered include blood glucose, hematocrit,
Abdominal thoracic bands or the Graseby capsule are electrolytes, sepsis8 screen, blood culture, arterial blood
used, the inductance of which changes with breathing. gas, chest X-ray, abdominal X-ray, ultrasound head and
other investigations depending on the history and
Magnetometer: Electrical signal produced by chest or physical examination.
abdominal movement then can be detected by the sensor.
Apnea monitors based on chest wall movement are
TREATMENT
likely to miss obstructive apnea. Monitors with facilities
for measuring heart rate and oxygen saturation would be
more useful in the monitoring of significant apnea in General measures
preterm infants.
Maintain airway, breathing and circulation.
Differential Diagnosis Avoid vigorous suctioning of oro-pharynx
Periodic breathing: It consists of breathing for 10-15 Avoid oral feeds repeated episodes of apnea
seconds, followed by apnea for 5-10 sec without change requiring BMV.

58 Indian Journal of Pediatrics, Volume 75January, 2008


16 10

Apnea in the Newborn

Decrease environmental temperature to lower end Continuous positive airway pressure (CPAP)
of thermo-neutral range. Avoid swings in Mechanical ventilation
environmental temperature.
Kinesthetic stimulation
Treatment of the underlying cause: sepsis, anemia,
Pharmacotherapy for AOP
polycythemia, hypoglycemia, hypocalcemia,
respiratory distress syndrome (RDS). Aminophylline, caffeine and doxapram have been used
in the treatment of AOP. The indications for starting
Transfuse packed cells if hematocrit <30%.
drugs are:
Specific measures for AOP Mechanical ventilation
These include For treatment for apnea of prematurity.6
Drugs including aminophylline, caffeine, doxapram Post extubation to reduce the incidence of apnea.7

Algorithm for management of neonatal apnea

Neonate with apnea

Emergency treatment
Maintain temperature, ABC

Investigations
BS,PCV,ABG
Evaluation to exclude secondary Sepsis screen
causes of apnea CXR, AXR
Na, K, Ca
US Head

Apnea of prematurity

Start specific
Treatment
Start aminophylline

Apnea responds No response

Continue till CPAP


34 weeks
Trial of doxapram Responds
No response

Stop drugs if Fails


no apnea for
last 7 days IMV/sNIPPV Continue x 48 hours

(ABC: airway; breathing; circulation; BS: blood sugar; PCV: packed cell volume; ABG: arterial blood gas; Na: sodium;
K: potassium; Ca: calcium; US: ultrasound; CPAP: continuous positive airway pressure; IMV: intermittent mandatory
ventilation); SNIPPV: Synchronised nasal intermittet positive preserve ventilation)

Indian Journal of Pediatrics, Volume 75January, 2008 59


17 11

S. Mishra et al

Methylxanthines caution or preferably avoided.


Methylxanthines have been the mainstay of At present, indications for doxapram include failure
pharmacologic treatment of AOP. Xanthine therapy to respond to both methylxanthine and CPAP therapy.
increases minute ventilation, improves CO2 sensitivity, The loading dose is preferably avoided. Doxapram
decreases hypoxic depression of breathing, enhances infusion is started at 0.5 mg/Kg/hour and increased
diaphragmatic contractility, and decreases periodic gradually to a maximum of 2-2.5 mg/Kg/hr. Doxapram
breathing. The major mechanism of action is likely may be tried for a period of 48 hours before weaning the
through competitive antagonism of adenosine receptors. drug. Methylxanthine therapy should be continued
Adenosine acts as an inhibitory neuro-regulator in the during doxapram infusion. Adverse effects include
central nervous system and is released during hypoxia. seizures, hypertension, hyperactivity, hyperglycemia and
Neonates exhibit hypoxic respiratory depression, and the abdominal distension. It should be avoided in the first
ability of methylxanthines to block this response may week of the life or if the serum bilirubin is high, because
contribute to their effect on apnea. A recent Cochrane an association with intraventricular hemorrhage and
review of the use of methylxanthines concluded that kernicterus has been reported.13 Injection doxapram has
methylxanthines are effective in reducing the number of 0.9% benzyl alcohol as a preservative. The recommended
apneic attacks and the use of mechanical ventilation. dose of 2-2.5 mg/Kg/hr would deliver 21.6-32.4 mg/Kg/
day of benzyl alcohol. Although this dose is below the
The loading dose of intravenous aminophylline is 5 to
toxic dose of alcohol (45 mg/Kg/day), there have been
6 mg/Kg, followed by 1.5 to 3 mg/Kg every 8 to 12 hours.
case reports of gasping syndrome with this lower dose
Oral theosphylline can be administered once the infant
in literature.
becomes stable in the same dose. Caffeine, both oral and
intravenous use, has some advantages over theophylline. Which drug to use?
Because it has a higher therapeutic index, toxicity is less
Present evidence shows that aminophylline, caffeine and
of a concern. Also, once-daily dosing is possible due to its
doxapram are equally effective in the treatment of AOP.
longer half-life. A typical loading dose of 20 mg/Kg
However, their clinical use is dependent upon adverse
caffeine citrate is followed in 24 hours by 5 to 8 mg/Kg
effects. The drug of choice would be caffeine, which is not
per dose, administered once every 24 hours.
available in India. Hence, we prefer to use aminophylline
Recommended therapeutic levels are 5 to 10 g/ml for
as the drug of choice in the management of AOP.
aminophylline and 8 to 20 g/ml for caffeine. 8
Aminophylline should be continued till 34 weeks Continuous positive airway pressure
corrected gestational age and stopped thereafter if no
Continuous positive airway pressure (CPAP), usually
episodes of apnea have occurred in the last 7 days.
administered using nasal prongs (NCPAP), is also an
Aminophylline initiated in order to facilitate extubation
effective treatment of AOP and is typically used when
may be stopped after 7 days.9
clinically significant episodes persist despite optimal
Elimination of methylxanthines is prolonged in infants methylxanthine therapy.15 At CPAP level of 5 cm water,
especially in preterm neonates. Xanthine therapy should infants with AOP will have fewer episodes. This
be discontinued at least 1 to 2 weeks prior to discharge, a reduction is primarily related to significant reduction in
guideline that is especially relevant for caffeine because episodes of obstructive and mixed apneas and has been
of its longer half-life. Do not discharge the patient until attributed to splinting open of the upper airways by the
methylxanthines have been stopped. There is no role for positive airway pressure. If NCPAP in conjunction with
prophylactic use of methylxanthines for prevention of a methylxanthine, is not sufficient to prevent recurring
apnea, bradycardia, or desaturation in premature episodes associated with bradycardia and intermittent
infants.10 Adverse effects include tachycardia, jitteriness, hypoxemia, some investigators have suggested use of
irritability, feed intolerance, vomiting and nasal intermittent positive pressure ventilation (NIPPV).
hyperglycemia. The drug is available as: NIPPV may most likely be needed when episodes are
related predominantly to central apneas and are not
Injection: Aminophylline ampoule 250 mg per 10-ml
effectively eliminated by the NCPAP.3 CPAP may also be
ampoule.
used to reduce post-extubation apnea in preterm
Oral: Theophylline 50 mg/5 ml in Theoped syrup
infants.16
Caffeine: The drug is not available in India at present.
A CPAP of 5 cm H2O is usually used. CPAP may be
Doxapram11, 12
delivered by nasal prongs or nasopharyngeal tube.
Intravenous Doxapram might reduce AOP in the short Endotracheal CPAP is not used in the treatment of apneic
term. The possible effects of treatment are usually not spells. CPAP has no role in prophylaxis against apnea of
sustained after 48 hours of commencement of doxapram prematurity. Adverse effects of CPAP include
treatment. More over, doxapram is associated with barotrauma, abdominal distension, feeding intolerance
serious side effects and hence should be used with and local nasal irritation

60 Indian Journal of Pediatrics, Volume 75January, 2008


18 12

Apnea in the Newborn

Mechanical Ventilation lower developmental indices at two years.3

The infant should be ventilated if both pharmacotherapy


REFERENCES
and CPAP have been tried and significant apneas
continue to occur. If the lungs are normal, the infant
should be ventilated at minimum pressures (peak 1. Hunt CE. Apnea and sudden infant death syndrome. In
Kligman RM, Neider ML, Super DM, eds. Practical strategiesin
inspiratory pressure of 10-12 cm of water and positive
pediatric diagnosis and therapy. Philadelphia; WB Saunders,
end expiratory pressure of 3-5 cm of water), low rate (20- 1996: 135-147.
25 per minute), short Ti (0.35-0.40 seconds) and low FiO2 2. Bhatia J. Current options in the management of apnea of
(0.3-0.5). This method is effective in all forms of apnea. prematurity. Clin Pediatr 2000; 39 : 327-336.
3. Thompson MW, Hunt CE. In MacDonald MG, Mullett MD,
Kinesthetic stimulation Seshia MMK, eds. Averys Neonatology Pathophysiology &
Management of the Newborn 6 th ed. Philadelphia; Lippincott
Water bed, oscillating bed mattress. Present evidence Williams and Wilkins, 1998; 539-45.
does not support any role for this mode of therapy either 4. Martin RJ, Abu-Shaweesh, Baird TM. Pathophysiologic
in the prevention or treatment of apnea.17 Mechanisms Underlying Apnea of Prematurity. Neo Reviews
2002; 3 : e59.
5. Deorari AK. In Deorari AK, Paul VK, eds. Neonatal Equipment
PERSISTENT APNEA Everything that you like to know 3rd ed. New Delhi; Sagar
Publication, 2006; 60-61.
6. Henderson-Smart DJ, Steer P. Methylxanthine treatment for
Apneic episodes may persist beyond 37-40 weeks in some apnea in preterm infants. Cochrane Database Syst Rev 2000.
infants, especially those born before 28 weeks of 7. Henderson-Smart DJ, Steer P. Prophylactic methylxanthine for
extubation in preterm infants. Cochrane Database Syst Rev 2004.
gestation. Methylxanthine therapy should be continued if 8. Martin RJ, Abu-Shaweesh , Baird TM. Clinical Associations,
apneic episodes continue to occur beyond 34 weeks of Treatment, and Outcome of Apnea of Prematurity. Neo
corrected gestational age. The neonate should be re- Reviews 2002; 3: e66.
evaluated for secondary causes of apnea especially 9. Miller MJ, Martin RJ. Apnea of prematurity. Clin Perinatol
neurological problems and gastro-esophageal reflux. 1992; 19 : 789-808.
10. Henderson-Smart DJ, Steer PA. Prophylactic methylxanthine
Home monitoring is not possible in our country and these
for prevention of apnea in preterm infants. Cochrane Database
infants would require NICU care until drugs can be Syst Rev 2006.
weaned and stopped. 11. Henderson-Smart DJ, Steer PA. Doxapram treatment for
apnea in preterm infants. Cochrane Database Syst Rev 2004.
12. Hansen T, Corbet A. Control of breathing. In Taeusch HW,
SUDDEN INFANT DEATH SYNDROME Ballard RA, eds. Averys diseases of the newborn, 7 th ed.
(SIDS) AND APNEA Philadelphia, WB Saunders, 1998; 552-561.
13. Jardine DS, Rogers K. Relationship of benzyl alcohol to
kernicterus, intraventricular hemorrhage, and mortality in
AOP is not found to be an independent risk factor for premature infants. Pediatrics; 83 : 721.
SIDS. Only 2-4% of patients with SIDS have a history of 14. Kumar M, Kabra NS, Paes B. Carnitine supplementation for
preterm infants with recurrent apnea. Cochrane Database Syst
AOP. Rev 2004.
15. Henderson-Smart DJ, Subramanian P, Davis PG. Continuous
positive airway pressure versus theophylline for apnea in
NEURODEVELOPMENT OUTCOME preterm infants. Cochrane Database Syst Rev 2005.
16. Davis PG, Henderson-Smart DJ. Nasal continuous positive
airway pressure immediately after extubation for preventing
Most reports have found little evidence of any morbidity in preterm infants. Cochrane Database Syst Rev 2003.
neurodevelopment risk directly attributed to a history of 17. Osborn DA, Henderson-Smart DJ. Kinesthetic stimulation for
AOP. Precisely measured predischarge apnea related to treating apnea in preterm infants. Cochrane Database Syst Rev
AOP, however, has been reported to be predictive of 2003.

Indian Journal of Pediatrics, Volume 75January, 2008 61


19
20
21
22
23 13

Symposium on AIIMS Protocols in Neonatology III

Approach to Inborn Errors of Metabolism Presenting in


the Neonate
Suvasini Sharma, Pradeep Kumar, Ramesh Agarwal, Madhulika Kabra, Ashok K. Deorari and
Vinod K. Paul

Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India

ABSTRACT
Inborn errors of metabolism (IEM) are an important cause of acute illness in newborns. Presentation may mimic common
neonatal conditions such as sepsis. Prompt detection requires a high index of suspicion and the early measurement of
biochemical markers such as blood ammonia. Diagnosis is important not only for treatment but also for genetic counselling.
Guidelines for diagnosis and early management of IEM presenting in the neonatal period are described. [Indian J Pediatr
2008; 75 (3) : 271-276] E-mail: ashokdeorari_56@hotmail.com

Key words : Inborn errors of metabolism; Encephalopathy; Hyperammonemia

Inborn errors of metabolism (IEM) are disorders in which Family history of neonatal deaths
there is a block at some point in the normal metabolic Rapidly progressive encephalopathy and seizures of
pathway caused by a genetic defect of a specific enzyme. unexplained cause
The number of diseases in humans known to be Severe metabolic acidosis
attributable to inherited point defects in metabolism now Persistent vomiting
exceeds 500.1 While the diseases individually are rare, Peculiar odor
they collectively account for a significant proportion of Acute fatty liver or HELLP (hemolysis, elevated liver
neonatal and childhood morbidity and mortality. enzymes and low platelet counts) during pregnancy:
Diagnosis is important not only for treatment and seen in women carrying fetuses with long-chain-3-
prognostication but also for genetic counselling and hydroxyacyl-coenzyme dehydrogenase deficiency
antenatal diagnosis in subsequent pregnancies. (LCHADD).
Table 1 describes examination findings that may
provide a clue to the underlying IEM.
CLINICAL PRESENTATION
TABLE 1. Clinical Pointers Towards Specific IEMs
Severe illness in the newborn, regardless of the Clinical finding Disorder
underlying cause, tends to manifest with non-specific
findings, such as poor feeding, drowsiness, lethargy, Coarse facies Lysosomal disorders
Cataract Galactosemia, Zellweger syndrome
hypotonia and failure to thrive. IEM should be considered Retinitis pigmentosa Mitochondrial disorders
in the differential diagnosis of any sick neonate along with Cherry red spot Lipidosis
common acquired causes such as sepsis, hypoxic-ischemic Hepatomegaly Storage disorders, urea cycle defects
encephalopathy, duct-dependant cardiac lesions, Renal enlargement Zellweger syndrome
congenital adrenal hyperplasia and congenital infections. Eczema/alopecia Biotinidase deficiency
Abnormal kinky hair Menke disease
Clinical pointers towards an underlying IEM include.2
Decreased pigmentation Phenylketonuria
Deterioration after a period of apparent normalcy
Parental consanguinity Patterns of presentation include.2,3
(i) Encephalopathy with or without metabolic acidosis:
Encephalopathy, seizures, and tone abnormalities are
Correspondence and Reprint requests : Dr Ashok K Deorari, predominant presenting features of organic acidemias,
Professor, Department of Pediatrics, All India Institute of Medical urea cycle defects and congenital lactic acidosis.
Sciences, Ansari Nagar, New Delhi 110029, India Intractable seizures are prominent in pyridoxine
[Received February 7, 2008; Accepted February 7, 2008] dependency, non-ketotic hyperglycinemia, molybdenum

Indian Journal of Pediatrics, Volume 75March, 2008 271


24 14

S. Sharma et al

co-factor defect and folinic-acid responsive seizures. Fig 1 gives the algorithmic approach to a newborn
with suspected IEM. Disease category can be diagnosed
(ii) Acute liver disease: This could manifest as-
based on blood ammonia, blood gas analysis and urine
Jaundice alone- Gilbert syndrome, Criggler-Najjar ketone testing. Hyperammonemia without acidosis is
syndrome caused by urea cycle defects. Metabolic acidosis with or
Hepatic failure (jaundice, ascites, hypoglycemia, without hyperammonemia is a feature of organic
coagulopathy)- Tyrosinemia, galactosemia, neonatal acidemias and fatty acid oxidation defects. Fig. 2 explains
hemochromatosis, glycogen storage disease type IV. the algorithmic approach to neonate with persistent
hypoglycemia and suspected underlying IEM. Table 2
Neonatal cholestasis: alpha-1 antitrypsin deficiency,
explains the categorization of IEM based on simple
Niemann-Pick disease type C.
metabolic screening tests.
Hypoglycemia: persistent and severe hypoglycemia
TABLE 2. Categorization of Neonatal IEM Using Metabolic
may be an indicator of an underlying IEM.
Screening Tests
Hypoglycemia is a feature of galactosemia, fatty acid
oxidation defects, organic acidemias, glycogen storage Acidosis Ketosis -Lactate -Ammonia Diagnosis
disorders and disorders of gluconeogenesis.
- + - - Maple syrup urine
(iii) Dysmorphic features: Seen in peroxisomal disorders, disease
pyruvate dehydrogenase deficiency, congenital disorders + +/- - +/- Organic aciduria
+ +/- + - Lactic acidosis
of glycosylation (CDG), and lysosomal storage diseases. - - - + Urea cycle
Some IEMs may present with non-immune hydrops - - - - Non-ketotic
fetalis; these include lysosomal storage disorders and hyperglycinemia
CDG. sulfite oxidase
deficiency,
(iv) Cardiac disease: Cardiomyopathy is a prominent peroxisomal,
feature in some IEM including fatty acid oxidation Phenylketonuria,
defects, glycogen storage disease type II and galactosemia
mitochondrial electron transport chain defects.
Second line investigations (ancillary and confirmatory
INVESTIGATIONS tests)
These tests need to be performed in a targeted manner,
Metabolic investigations should be initiated as soon as the based on presumptive diagnosis reached after first line
possibility is considered. The outcome of treatment of investigations:
many IEM especially those associated with Gas chromatography mass spectrometry (GCMS) of
hyperammonemia is directly related to the rapidity with urine- for diagnosis of organic acidemias.
which problems are detected and appropriate
management instituted. Plasma amino acids and acyl carnitine profile: by
tandem mass spectrometry (TMS)- for diagnosis of
First line investigations (metabolic screen) organic acidemias, urea cycle defects,
The following tests should be obtained in all babies with aminoacidopathies and fatty acid oxidation defects.
suspected IEM. High performance liquid chromatography (HPLC):
Complete blood count: (neutropenia and for quantitative analysis of amino acids in blood and
thrombocytopenia seen in propionic and urine; required for diagnosis of organic acidemias
methylmalonic academia) and aminoacidopathies.

Arterial blood gases and electrolytes Lactate/pyruvate ratio- in cases with elevated
Blood glucose lactate.
Plasma ammonia (Normal values in newborn: 90- Urinary orotic acid- in cases with hyperammonemia
150 g/dl or 64-107 mol/L) for classification of urea cycle defect.
Arterial blood lactate (Normal values: 0.5-1.6 mol/ Enzyme assay: This is required for definitive
L) diagnosis, but not available for most IEMs.
Liver function tests Available enzyme assays include: biotinidase assay-
Urine ketones in cases with suspected biotinidase deficiency
Urine reducing substances. (intractable seizures, seborrheic rash, alopecia); and
Serum uric acid (low in molybdenum cofactor . GALT (galactose 1-phosphate uridyl transferase )
deficiency). assay- in cases with suspected galactosemia

272 Indian Journal of Pediatrics, Volume 75March, 2008


25 15

Approach to Inborn Error of Metabolism Presenting in the Neonate

Fig 1. Approach to newborn with suspected metabolic disorder Suspected Metabolic Disorder

(hypoglycemia, cataracts, reducing sugars in urine). Magnetic resonance spectroscopy (MRS): may be
helpful in selected disorders e.g. lactate peak
Neuroimaging: MRI may provide helpful pointers
elevated in mitochondrial disorders, leucine peak
towards etiology while results of definitive
elevated in MSUD.
investigations are pending. Some IEM may be
Electroencephalography (EEG): some EEG
associated with structural malformations e.g.,
abnormalities may be suggestive of particular IEM;
Zellweger syndrome has diffuse cortical migration
e.g., comb-like rhythm in MSUD, burst suppression
and sulcation abnormalities. Agenesis of corpus
in NKH and holocarboxylase synthetase deficiency.5
callosum has been reported in Menkes disease,
Plasma very long chain fatty acid (VLCFA) levels:
pyruvate decarboxylase deficiency and nonketotic
elevated in peroxisomal disorders.
hyperglycinemia.4 Examples of other neuroimaging
findings in IEM include: Mutation analysis when available.
Maple syrup urine disease (MSUD): brainstem and CSF aminoacid analysis: CSF Glycine levels elevated in
cerebellar edema NKH.
Propionic and methylmalonic acidemia: basal Precautions to be observed while collecting samples
ganglia signal change
Should be collected before specific treatment is started or
Glutaric aciduria: frontotemporal atrophy, subdural feeds are stopped, as may be falsely normal if the child is
hematomas off feeds.

Indian Journal of Pediatrics, Volume 75March, 2008 273


26 16

S. Sharma et al

Fig. 2. Approach to newborn with persistent hypoglycemia and suspected IEM

Samples for blood ammonia and lactate should be broadly categorize the patients IEM (e.g., urea cycle
transported in ice and immediately tested. Lactate defect, organic academia, congenital lactic acidosis etc), on
sample should be arterial and should be collected after 2 the basis of which, empirical treatment can be instituted.
hrs fasting in a preheparinized syringe. Ammonia sample
Aims of treatment
is to be collected approximately after 2 hours of fasting in
EDTA vacutainer. Avoid air mixing. Sample should be 1. To reduce the formation of toxic metabolites by
free flowing. decreasing substrate availability (by stopping feeds
and preventing endogenous catabolism)
Detailed history including drug details should be
provided to the lab. (sodium valproate therapy may 2. To provide adequate calories
increase ammonia levels). 3. To enhance the excretion of toxic metabolites.
Samples to be obtained in infant with suspected IEM 4. To institute co-factor therapy for specific disease and
when diagnosis is uncertain and death seems inevitable also empirically if diagnosis not established.
(Metabolic autopsy)6 5. Supportive care- treatment of seizures (avoid
sodium valproate may increase ammonia levels),
Blood: 5-10 ml; frozen at -200C; both heparinized (for
maintain euglycemia and normothermia, fluid,
chromosomal studies) and EDTA (for DNA studies)
electrolyte and acid-base balance, treatment of
samples to be taken
infection, mechanical ventilation if required.
Urine: frozen at 20oC
CSF: store at 20oC Management of hyperammonemia7,8
Skin biopsy: including dermis in culture medium or Discontinue all feeds. Provide adequate calories by
saline with glucose. Store at 4-80C. Do not freeze. intravenous glucose and lipids. Maintain glucose infusion
Liver, muscle, kidney and heart biopsy: as indicated. rate 8-10mg/kg/min. Start intravenous lipid 0.5g/kg/
Clinical photograph (in cases with dysmorphism) day (up to 3g/kg/day). After stabilization gradually add
protein 0.25 g/kg till 1.5 g/kg/day.
Infantogram (in cases with skeletal abnormalities)
Dialysis is the only means for rapid removal of
TREATMENT ammonia, and hemodialysis is more effective and faster
than peritoneal dialysis, however peritoneal dialysis may
In most cases, treatment needs to be instituted empirically be more widely available and feasible. Exchange
without a specific diagnosis. The metabolic screen helps to transfusion is not useful.

274 Indian Journal of Pediatrics, Volume 75March, 2008


27 17

Approach to Inborn Error of Metabolism Presenting in the Neonate

Alternative pathways for nitrogen excretion: and blood glucose. In glucose transporter defect,
CSF glucose level is equal to or less than 1/3rd of the
Sodium benzoate (IV or oral)- loading dose 250 mg/kg
blood glucose level. This disorder responds to the
then 250-400 mg/kg/day in 4 divided doses. (Intravenous
ketogenic diet.
preparation not available in India).
Management of asymptomatic newborn with a history
Sodium phenylbutyrate (not available in India)-
of sibling death with suspected IEM:
loading dose 250 mg/kg followed by 250-500 mg/kg/
day. After baseline metabolic screen, start oral dextrose
feeds (10% dextrose).
L-arginine (oral or IV)- 300 mg/kg/day (Intravenous
preparation not available in India) After 24 hours, repeat screen. If normal, start breast
feeds. Monitor sugar, blood gases and urine
L-carnitine (oral or IV)- 200 mg/kg/day ketones, blood ammonia 6 hourly.
Supportive care: treatment of sepsis, seizures, Some authorities recommend starting medium
ventilation. Avoid sodium valproate. chain triglycerides (MCT oil) before starting breast
feeds,3 however, this is not being followed in our
Acute management of newborn with suspected organic
center (because of unpalatibility of MCT oil).
acidemia9
After 48 hours, repeat metabolic screen. Obtain
The patient is kept nil per orally and intravenous samples for TMS and urine organic acid tests.
glucose is provided.
The infant will need careful observation and follow-
Supportive care: hydration, treatment of sepsis, up for the first few months, as IEM may present in
seizures, ventilation. different age groups in members of the same family.
Carnitine: 100 mg/kg/day IV or oral.
Treat acidosis: Sodium bicarbonate 0.35-0.5mEq/ Long term treatment of IEM
kg/hr (max 1-2mEq/kg/hr) The following modalities are available
Start Biotin 10 mg/day orally.
Dietary treatment: This is the mainstay of treatment in
Start Vitamin B12 1-2 mg/day I/M (useful in B12
phenylketonuria, maple syrup urine disease,
responsive forms of methylmalonic acidemias)
homocystinuria, galactosemia, and glycogen storage
Start Thiamine 300 mg/day (useful in Thiamine-
disease Type I and III. Special diets for PKU and MSUD
responsive variants of MSUD).
are commercially available in the west. These are not
If hyperammonemia is present, treat as explained available in India, but can be imported. These special diets
above. are however very expensive, and cannot be afforded by
Management of congenital lactic acidosis most Indian patients. Based on the amino acid content of
some common food products available in India, dietary
Supportive care: hydration, treatment of sepsis, exchanges are calculated and a low phenylalanine diet for
seizures, ventilation. Avoid sodium valproate. PKU and diet low in branched chain amino acids for
Treat acidosis: sodium bicarbonate 0.35-0.5mEq/ MSUD are being used in our center. However, there are
kg/hr (max 1-2mEq/kg/hr) no studies to document the efficacy of these indigenous
Thiamine: up to 300 mg/day in 4 divided doses. diets. Some disorders like urea cycle disorders and
organic acidurias require dietary modification (protein
Riboflavin: 100 mg/day in 4 divided doses.
restriction) in addition to other modalities.11
Add co-enzyme Q: 5-15 mg/kg/day
Enzyme replacement therapy (ERT): ERT is now
L-carnitine: 50-100 mg/kg orally.
commercially available for some lysosomal storage
Treatment of newborn with refractory seizures with no disorders.12 However, these disorders do not manifest in
obvious etiology (suspected metabolic etiology)10 the newborn period, an exception being Pompes disease
(Glycogen storage disorder Type II)which may present in
If patient persists to have seizures despite 2 or 3
the newborn period and for which ERT is now available.
antiepileptic drugs in adequate doses, consider trial
of pyridoxine 100 mg intravenously. If intravenous Cofactor replacement therapy: The catalytic properties of
preparation not available, oral pyridoxine can be many enzymes depend on the participation of non
given (15 mg/Kg/day). protein prosthetic groups, such as vitamins and minerals,
as obligatory cofactors. The following co-factors may be
If seizures persist despite pyridoxine, give trial of
beneficial in certain IEM:13
biotin 10 mg/day and folinic acid 15 mg/day
(folinic acid responsive seizures). Thiamine: mitochondrial disorders, thiamine
responsive variants of MSUD, PDH deficiency &
Rule out glucose transporter defect: measure CSF complex I deficiency)

Indian Journal of Pediatrics, Volume 75March, 2008 275


28 18

S. Sharma et al

Appendix: Commercially Available Formulations Used in IEM

Co-factor Trade name, formulation

Pyridoxine Tab Benadon (40mg) (Nicholas Piramal), Inj Vitneurin (1 ampoule contains
50 mg pyridoxine)
Hydroxycobalamin (Vitamin B12) Inj Trineurosol (1000mcg/ml) (Tridoss Laboratories)
Thiamine Tab Benalgis (75 mg) (Franco India)
Riboflavin Tab Riboflavin (5 mg) (Shreya)
Biotin Tab Essvit (5mg, 10mg) (Ecopharma)
Carnitine Syrup L-Carnitor (5ml=500 mg), Tab L-Carnitor (500 mg), Inj carnitor (1g/
5ml) (Elder)
Folinic acid Tab Leukorin (15 mg) (Samarth)
Sodium Benzoate Satchet 20g (Hesh Co.)
Arginine ARG-9 Satchet (3g) (Noveau Medicament)
Coenzyme Q Tab CoQ 30 mg, 50 mg. (Universal Medicare)

Riboflavin: Glutaric aciduria Type I, Type II, mild phenylketonuria, peroxisomal defects.
variants of ETF, ETF-DH, complex I deficiency Enzyme assay: useful in lysosomal storage
Pyridoxine: 50% of cases of homocystinuria due to disorders like Niemann-Pick disease, Gaucher
cystathionine -synthetase deficiency, pyridoxine disease.
dependency with seizures, xanthurenic aciduria, DNA based (molecular) diagnosis: Detection of
primary hyperoxaluria type I, Hyperornithemia mutation in proband/ carrier parents is a
with gyrate atrophy prerequisite.
Cobalamin: Methylmalonic academia (cblA, cblB),
Homocystinuria and methylmalonic academia REFERENCES
(cblC, cblD, cblF)
Folinic acid: Hereditary orotic aciduria, Methionine 1. Childs B, Valle D, Jimenez-Sanchez. The Inborn error and
synthase deficiency, Cerebral folate transporter biochemical variability. In Scriver CR, Beaudet AL, Sly WS,
deficiency, hereditary folate malabsorption, Kearns- Valle D, eds. The metabolic and molecular basis of inherited
disease, 8th ed, New York; McGraw-Hill, 2001: 155-166.
Sayre syndrome 2. Clarke, JTR. A Clinical guide to inherited metabolic diseases. 3rd
Biotin: Biotinidase deficiency, holocarboxylase Ed. Cambridge University Press, Cambridge; 2006.
synthetase deficiency 3. Cataltepe SU, Levy HL. Inborn errors of metabolism. In
Cloherty JP, Eichenwald EC, Stark AR, eds. Manual of neonatal
care. 6th ed. Philadelphia; Lippincott Williams and Wilkins,
PREVENTION 2008; 558-573.
4. Blaser S, Feigenbaum A. A neuroimaging approach to inborn
Neonatal screening: Tandem mass spectrometry is used in errors of metabolism. Neuroimag Clin N Am 2004; 14: 307-329.
some countries for neonatal screening for IEM. Disorders 5. Nordli DR, De Vivo DC. Classification of infantile seizures:
which can be detected by TMS include aminoacidopathies Implications for identification and treatment of inborn errors
of metabolism. J Child Neurol 2002; 17 (Suppl 3): 3S3-3S8.
(phenylketonuria, MSUD, Homocystinuria, Citrullinemia, 6. Leonard JV, Morris AAM. Diagnosis and early management
Argininosuccinic aciduria, hepatorenal tyrosinemia), fatty of inborn errors of metabolism presenting around the time of
acid oxidation defects, organic acidemias (glutaric birth. Acta Pediatrica 2006; 95: 6-14.
aciduria, propionic acidemia, methylmalonic acidemia, 7. Summar M. Current strategies for the management of
isovaleric acidemia). The cost of this procedure is high, a neonatal urea cycle disorders. J Pediatr 2001; 38: S30-S39.
8. Leonard JV, Morris AAM. Urea cycle disorders. Semin
potent dis-inventive for resource poor countries like India. Neonatol 2002; 7: 27-35.
Also, the though the test is highly sensitive, the specificity 9. de Baulny HO, Saudubray JM. Branched-chain organic
is relatively low; and there are difficulties in interpretation acidurias. Semin Neonatol 2002; 7: 65-74.
of abnormal test results in apparently healthy infants. 10. Wolf NI, Bast T, Surtees S. Epilepsy in inborn errors of
metabolism. Epileptic Disord 2005; 7 : 67-81.
Genetic counselling and prenatal diagnosis: Most of the IEM 11. Kabra M. Dietary management of Inborn errors of
are single gene defects, inherited in an autosomal metabolism. Indian J Pediatr 2002; 69: 421-426.
recessive manner, with a 25% recurrence risk. Therefore, 12. Brady RO, Schiffmann R. Enzyme-replacement therapy for
metabolic storage disorders. Lancet Neurol 2004; 3: 752-756.
when the diagnosis is known and confirmed in the index
13. Saudubray JM, Sedel F, Walter JH. Clinical approach to
case, prenatal diagnosis can be offered, wherever treatable inborn metabolic diseases: an introduction. J Inherit
available for the subsequent pregnancies. The samples Metab Dis 2006; 29: 261-274.
required are chorionic villus tissue or amniotic fluid. 14. Elias S, Simpson JL, Shulman LP. Techniques for prenatal
Modalities available are:14 diagnosis. In Rimoin DL, Connor JH, Pyeritz RE, Korf BR, eds.
Emery and Rimoins Principles and practice of medical genetics.
Substrate or metabolite detection: useful in London; Churchill-Livingstone, 2002: 802-825.

276 Indian Journal of Pediatrics, Volume 75March, 2008


29
30
31
32
33 19

Symposium on AIIMS Protocols in Neonatology - V

Blood and Blood Component Therapy in Neonates


Richa Jain, Bipin Jose, Poonam Coshic1, Ramesh Agarwal and Ashok K. Deorari

Division of Neonatology, Department of Pediatrics, 1Department of Transfusion Medicine, All India Institute of
Medical Sciences, New Delhi, India

ABSTRACT
Blood component therapy is a very common intervention practiced in newborns; nearly 85% of extremely low birth weight
(ELBW) babies get transfusions during their hospital stay. However, there are no set guidelines for transfusion of blood
component therapy in newborns. This protocol includes available types of blood components , their methods of preparation,
indications and side effects of transfusion, in relation to newborns. [Indian J Pediatr 2008; 75 (5) : 489-495] Email:
aranag@rediffmail.com

Key words: Transfusion; Packed red cells; Platelets; Newborn

Blood components used in modern day practice Indications for PRBC transfusion in neonatal practice
include, apart from whole blood, a variety of other
PRBCs are the most commonly used blood product in
products, like red blood cell components, platelet
neonatal transfusions. 3 Indications for transfusion of
concentrates, and plasma. Blood component
PRBCs are mainly for resolution of symptomatic
transfusion has been considered to be a safe and low
anemia and improvement of tissue oxygenation. Tissue
risk procedure. In the last few decades there has been
oxygenation depends on cardiac output, oxygen
recognition of hazards of transfusion of blood and its
saturation and hemoglobin concentration. Once cardiac
products. It is no longer considered to be a low or no
output and oxygen saturation are optimal, tissue
risk procedure, and consequently an increasing need for
oxygenation can only be improved by increasing the
stricter guidelines for transfusing blood products has
hemoglobin level. The guidelines for transfusion of
been recognized, not just to check infections, but also to
PRBC vary according to age, level of sickness and
minimize other side effects of transfusion. Preterm
hematocrit (Table 1).3
neonates comprise the most heavily transfused group of
patients, and about 85% of extremely low birth weight Packed Red Blood Cells (PRBCs)
newborns receive a transfusion by the end of their
Most RBC components available today are derived from
hospital stay.1,2
the collection of 350-450 mL of whole blood into sterile
plastic bags containing citrate-phosphate-dextrose
(CPD) anticoagulant. The whole blood is spun to
RED BLOOD CELL PRODUCTS
sediment out the RBCs, and most of the plasma is
removed by pushing it into a pre-attached satellite bag.
Red cells and their products include packed red blood Generally, 100 to 110 mL of a nutrient additive solution
cells (PRBCs) and modified blood products used for is added back to the packed RBCs, creating an
specific situations including: additive RBC product that has a final hematocrit of
55% to 60%. A variety of additive solutions are in use
Leukocyte reduced RBCs today, each of which contains a particular mix of
Irradiated RBCs glucose, adenine, and mannitol. These solutions
Washed RBCs prolong the shelf life of the RBC product from 21 days
RBCs with low CMV risk (packed RBCs in CPD) to 42 days (additive RBCs). A
transfusion of 10 mL/kg of additive RBCs would be
Correspondence and Reprint requests : Dr Ramesh Agarwal, expected to raise the newborns hematocrit by 7% to 8%.
Assistant Professor, Division of Neonatology, Department of Red cells collected in CPDA-1 are kept as packed RBCs,
Pediatrics, All India Institute of Medical Sciences, New Delhi, that is, additive solution is not added. CPDA-1 packed
India. RBCs have a hematocrit of approximately 75% and a
[Received April 24, 2008; Accepted April 24, 2008] shelf life of 35 days. An infusion of 10 mL/kg of CPDA-

Indian Journal of Pediatrics, Volume 75May, 2008 489


34 20

Richa Jain et al

TABLE 1. Guidelines for Packed Red Blood Cells (PRBCs) the RBCs. It may also be done to reduce potassium in
Transfusion Thresholds for Preterm Neonates 3 stored RBCs in large volume transfusions. Isotonic
Less than 28 days of age and saline is added to blood components, and
Assisted ventilation with FiO 2 more than 0.3: Hb 12.0 gm/ centrifugation is done followed by removal of
dL or PCV less than 40% supernatant, and resuspension of the cells in saline.
Assisted ventilation with FiO2 less than 0.3: Hb 11.0 g/dL Washed products must be used within 4 hours of
or PCV less than 35%
processing, if stored at room temperature or within 24
CPAP: Hb less than 10 gm/dL or PCV less than 30%
More than 28 days of age and hours, if stored in the refrigerator.
Assisted ventilation: Hb less than 10 gm/dL or PCV less
CMV reduced RBCs
than 30%
CPAP: Hb less than 8 gm/dL or PCV less than 25% CMV reduced RBCs are used to reduce the risk of
Any age, breathing spontaneously and
transmitting CMV, which may be a cause of
On FiO2 more than 0.21: Hb less than 8 gm/dL or PCV less
than 25% considerable concern in newborns. CMV reduction can
On Room Air: Hb less than 7 gm/dL or PCV less than 20% be achieved by either leukoreduciton of blood
components, or by pre-selecting donors who are CMV
negative. Providing CMV reduced blood is important in
1 packed RBCs would be expected to raise the patients
preterm infants, who have a more severe form of CMV
hematocrit by 9% to 10%.
infection than term newborns.
Modified RBC products
Whole blood versus PRBC
Leukocyte reduced RBCs
Overriding indication for whole blood transfusion is
Leukocyte depletion or reduction has been defined as when there is need for concurrent replacement of
achieving a concentration of less than 5 x 10 6 volume and coagulation factors (only fresh blood will
leukocytes per unit of RBCs. Leukocyte reduction is supply coagulation factors).
important in neonatal transfusion. It helps in
Reconstituted whole blood
preventing non-hemolytic febrile transfusion reactions
(NHFTR), HLA alloimmunization, transmission of It is obtained by resuspension of PRBCs, which is
leukotropic viruses (CMV, EBV and HTLV-1), frozen and deglycerolized. Red cells are frozen with
transfusion related GVHD, and transfusion related glycerol and stored at -80 oC in vapor phase of liquid
acute lung injury (TRALI).4 nitrogen. RBCs can be suspended in compatible but not
necessarily group-specific plasma. Reconstituted blood
Methods of leuko-reduction include the following: 4
can be used in case of rare blood groups or when
a. Centrifugation and removal of buffy coat (pre- neonate has multiple antibodies from previous
storage leukoreduction) transfusions so that compatible blood is difficult to
b. Use of leukocyte filters (pre or post storage) procure.
c. Washing of RBCs with saline
Practical Issues
d. Freezing and thawing of red cells
Amount of transfusion to be given: It has been seen that
Gamma irradiation
transfusion with PRBC at a dose of 20 mL/kg is well
Gamma irradiation of blood components including tolerated and results in an overall decrease in
RBCs, platelets and white blood cell products is done to number of transfusions compared to transfusions
inactivate donor T cells, and the associated risk of done at 10 mL/kg. There is also a higher rise in
transfusion associated graft versus host disease (TA- hemoglobin with a higher dose of PRBCs.8
GVHD), which may occur in immunosupressed
Properties of RBC products used in neonatal transfusion:
patients, very small babies, in large volume transfusions
and during intrauterine transfusions.5 a. RBCs should be freshly prepared and should not
be more than 7 days old. This translates into a
Irradiation causes an increase in the rate of leakage
high 2, 3-DPG concentration and higher tissue
of potassium out of RBCs during storage, and irradiated
extraction of oxygen. Other concerns with old
RBCs have a shortened shelf life of only 28 days.
RBCs are hyperkalemia, and a reduced RBC life
Irradiation does not adversely affect the function or
span.
viability of platelets.6 While most blood banks use a
dose of 1,500 cGy, the selection of an appropriate dose b. In small and sick neonates, where it is anticipated
of gamma irradiation remains an issue.7 that blood component therapy may be needed
more than once, it may help to have aliquots from
Washed RBCs
a single donor given as sequential transfusions.9
Saline washing is mainly done to remove plasma from This is done practically by reserving a bag of fresh

490 Indian Journal of Pediatrics, Volume 75May, 2008


35 21

Blood and Blood Component Therapy in Neonates

PRBC for up to 7 days for a newborn and platelets in the presence of normal platelet counts may
withdrawing small aliquots required repeatedly also cause bleeding tendency. Thrombocytopenia has
from that bag under laminar flow using a sterile been observed in 15% of newborns at birth.11-13 Severe
connecting device, into a fresh blood bag. The thrombocytopenia defined as platelet count of less than
PRBC bag is immediately resealed under the 50,000/cubic mm may occur in 0.10.5% of newborns.13-
14
laminar flow, and can be reused for withdrawing In NICU, there is a higher incidence; with thrombo-
similar small quantities of blood for up to 7 days. cytopenia being observed in up to 2235% of all babies
admitted to NICUs and in up to 50% of those admitted
Choosing the blood group for neonatal transfusions:4
to NICUs who require intensive care. Significant
a. It is preferable to take samples from both, mother proportions (20%) of these episodes of
and the newborn, for initial testing prior to thrombocytopenia are severe.15-16 Thus a large number of
transfusion. Mothers sample should be tested for neonates are at risk for bleeding disorders in NICU.
blood group and for any atypical red cell
Immune thrombocytopenia
antibodies.
a. Neonatal alloimmune thrombocytopenia (NAIT)
b. ABO compatibility is essential while transfusing
PRBCs. Though ABO antigens may be expressed The best choice of platelet transfusion is human platelet
only weakly on neonatal erythrocytes, neonates antigen (HPA) compatible platelets, which are generally
serum may contain transplacentally acquired maternal platelets, meticulously washed and
maternal IgG anti-A and/or anti-B. irradiated. The aim is to maintain the platelet count
above 30,000/ cubic mm.17 However; HPA compatible
c. Blood should be of newborns ABO and Rh group.
platelets are not easily available. In the absence of
It should be compatible with any ABO or atypical
immunologically compatible platelets, random donor
red cell antibody present in the maternal serum.
platelet transfusions may be an acceptable alternative,
d. In exchange transfusions for hemolytic disease of and has been shown to increase platelet counts above
newborn, blood transfused should be compatible 40,000/cubic mm in most of the transfused patients.18
with mothers serum. If the mothers and the
An alternative approach is the use of intravenous
babys blood groups are the same, use Rh negative
immunoglobulin (IVIG) (1 g/kg/day on two
blood of babys ABO group. In case mothers and
consecutive days or 0.5 g/kg/day for four days), alone
babys blood group is not compatible, use group O
or in combination with random donor platelet
and Rh negative blood for exchange transfusion.
transfusion. 19
Volume and rate of transfusion
b. Neonatal autoimmune thrombocytopenia
a. Volume of packed RBC = Blood volume (mL/kg)
The goal is to keep the count above 30,000/cubic mm.
x (desired minus actual hematocrit)/ hematocrit
IVIG is given if counts are less than the acceptable
of transfused RBC
minimum at a dose of 1 g/kg/day on two consecutive
b. Rate of infusion should be less than 10 mL/kg/ days. 20
hour in the absence of cardiac failure.
Nonimmunologically mediated thrombocytopenia
c. Rate should not be more than 2 mL/kg/hour in
Low platelet count occurring at less than 72 hours of
the presence of cardiac failure.
age is caused most commonly by placental
d. If more volume is to be transfused, it should be insufficiency, maternal PIH, early onset sepsis (EOS),
done in smaller aliquots. and perinatal asphyxia. EOS and asphyxia may, in
particular, lead to severe thrombocytopenia.
Expected response
Thrombocytopenia occurring beyond the initial 72
Each transfusion of 9 mL/kg of body weight should hours is most commonly caused by sepsis and
increase hemoglobin level by 3 g/dL. Meticulous necrotising enterocolitis. Other infrequent causes
monitoring of input, output and vital signs are include intrauterine infections, metabolic errors and
mandatory during blood transfusion. congenital defects in platelet production.10, 16 Indications
for platelet transfusion in nonimmune thrombo-
cytopenia depend on the level of sickness of newborn
PLATELET TRANSFUSION (Table 2).3
Types of platelets available
Thrombocytopenia is defined as platelet count less than
1.5 lakh/cubic mm. 10 Presence of thrombocytopenia Random donor platelet (RDP)
leads to an increase in risk of bleeding. Dysfunctional Each unit of a random donor pool is obtained from a

Indian Journal of Pediatrics, Volume 75May, 2008 491


36 22

Richa Jain et al

TABLE 2. Indications for Platelet Transfusion in Nonimmune platelet count from a 5-mL/kg dose would be 20 to
Thrombocytopenia in Newborn 60,000/cubic mm.15 Doses of up to 10-20 ml/kg may
Platelet count less than 30,000/cubic mm: transfuse all be used in case of severe thrombocytopenia.9
neonates, even if asymptomatic
Platelet count 30,000 to 50,000/cubic mm: consider
transfusion in PLASMA DERIVATIVES
a. Sick or bleeding newborns
b. Newborns less than 1000 gm or less than 1 week of age
c. Previous major bleeding tendency (IVH grade 3-4) Plasma contains about 1 unit/mL of each of the
d. Newborns with concurrent coagulopathy coagulation factors as well as normal concentrations of
e. Requiring surgery or exchange transfusion other plasma proteins. Labile coagulation factors, like
Platelet count more than 50,000 to 99,000/cubic mm: factors V and VIII, are not stable in plasma stored for
transfuse only if actively bleeding
prolonged periods at 16 C; therefore plasma is
usually stored frozen at 18 C or lower. Fresh frozen
single whole blood unit. Multiple such units from many plasma (FFP) is stored within 8 hours of collection. It
donors can be pooled together or each such unit can be contains about 87% of factor VIII present at the time of
given separately. PRBC is separated from the platelet collection and must contain at least 0.70 IU/mL of factor
rich plasma, which is then respun at a higher speed to VIII.22
make a concentrated platelet button
Fresh frozen plasma
Single donor platelet (SDP)
FFP has traditionally been used for a variety of reasons,
SDP units are obtained by a process called including volume replacement, treatment of
plateletpheresis. Here, from a single donor itself disseminated intravascular coagulopathy (DIC), during
multiple platelet units are separated. This is achieved the treatment of a bleeding neonate, for prevention of
by returning RBCs and platelet poor plasma to donors intraventricular hemorrhage, and in sepsis.3 It has not
circulation after plateletpheresis. The procedure is been shown to have any survival benefits in most of
repeated 4 to 6 times, yielding 4 to 6 units of platelets these conditions and currently the only valid
from one individual. It is especially useful to prevent indications for transfusing FFP in a newborn include
alloimmunization in multiply transfused patients. Both
SDPs and RDPs are irradiated. It is more cost effective to Disseminated intravascular coagulopathy
screen the larger SDP units for bacterial contamination, Vitamin K deficiency bleeding
than to screen individual random donor units.21 The Inherited deficiencies of coagulation factors
concentration of platelets is more in SDP than in RDP, Other rare indications include patients with
with SDP having a platelet concentration of 3x1011/unit afibrinogenemia, von Willebrand factor deficiency,
and RDP having a concentration of 0.5x1010/unit. In congenital antithrombin III deficiency, protein C
neonatal transfusion practice, RDP is generally deficiency and protein S deficiency when specific factor
adequate to treat thrombocytopenia. SDP is required replacement is not available. It is also used for
only if prolonged and severe thrombocytopenia is reconstitution of blood for exchange transfusion.
anticipated, requiring multiple platelet transfusions.
Cryoprecipitate
Platelet storage: Platelets are stored at 20C to 24C
using continuous gentle horizontal agitation in storage It is prepared from FFP by thawing at 2 4 o C.
bags specifically designed to permit O 2 and CO 2 Undissolved cryoprecipitate is collected by
exchange to optimize platelet quality. The storage time centrifugation and supernatant plasma is aseptically
from collection to transfusion of platelets (RDPs) is 5 expressed into a satellite bag.
days. SDPs can be stored for up to 7 days. Cryoprecipitate contains about 80 to 100 U of factor
Practical Issues VIII in 10-25 mL of plasma, 300 mg of fibrinogen and
varying amounts of factor XIII. It is stored at a
Platelets should never be filtered through a temperature of -20o C or below.
micropore blood filter before transfusion, as it will
considerably decrease the number of platelets. Indications for use of cryoprecipitate

Female Rh-negative infants should receive platelets Congenital factor VIII deficiency
from Rh-negative donors to prevent Rh sensitization Congenital factor XIII deficiency
from the contaminating red blood cells. Afibrinogenemia and dysfibrinogenemia
von Willebrand disease
The usual recommended dose of platelets for
neonates is 1 unit of platelets per 10 kg body weight, Practical Issues:9
which amounts to 5 mL/kg. The predicted rise in FFP should be group AB, or compatible with

492 Indian Journal of Pediatrics, Volume 75May, 2008


37 23

Blood and Blood Component Therapy in Neonates

recipients ABO red cell antigens derived from either asymptomatic bacteraemia in the
Volume of FFP to be transfused is usually 1020 mL/ donor, or from inadequate skin sterilization leading
kg to bacterial contamination of the blood. Platelets are
Volume of cryoprecipitate to be transfused is usually at a higher risk of causing bacterial infection than
5 mL/kg. other blood components, as they are stored at room
temperature, leading to rapid multiplication of
infectious organisms. The highest fatality is seen
TRANSFUSION ASSOCIATED RISKS
when the contaminating organism is a gram-
negative bacteria. In case of a febrile non-hemolytic
Blood transfusion reactions may be broadly classified reaction post transfusion, bacterial contamination
as always remains a possibility. It generally causes a
higher rise in temperature than other febrile
Infectious transfusion reactions.
Non-infectious
a. Acute Parasites: Plasmodium, trypanosome, and several
i. Immunologic other parasites may be transmitted through blood,
ii. Non-immunologic depending on the endemicity of the area.
b. Delayed Transfusion transmitted malaria is not uncommon
in India, and may occur in spite of blood bag testing,
Infectious complications as the screening tests for malaria are insensitive.25
In India, it is mandatory to test every unit of blood Prions : Variant Cruetzfold Jacob Disease ( v CJD) is
collected for hepatitis B, hepatitis C, HIV/AIDS, an established complication of blood transfusion
syphilis and malaria. 23 However, transfusion and has been reported since 2004. It is thought to
transmitted infections are still a considerable risk, have an incubation period of approximately 6.5
because of the relative insensitivity of screening tests, years. There is no easy test as yet to detect the
and several other organisms besides those tested for, presence of prions. It is not very clear whether leuko-
which may be transmitted through blood. reduction prevents transmission of CJD.24 Restricted
Viral infections: Transmissible diseases can be caused transfusions and avoidance of transfusions unless
by viruses like human immunodeficiency virus (HIV), essential, are the only ways currently to prevent
hepatitis B and C viruses (HBV & HCV), and transmission.
cytomegalovirus (CMV). Other uncommon viruses Noninfectious complications
like hepatitis G virus, human herpes virus-8 and
transfusion-transmitted virus have also been These can be further sub classified as immune mediated
detected. Viral infections contaminate platelet and nonimmune mediated reactions, and as acute and
products more commonly than RBC products due to delayed complications.
a higher temperature used for storage of platelet Acute immune mediated reactions
products.24 Though screening for HIV, HBV and HCV
is mandatory in blood banks, other viruses still Immune mediated hemolysis : Acute hemolytic
present an unaddressed problem. Insensitivity of transfusion reactions are a common cause of
pathogen testing is also an issue, and risk of viral transfusion related fatality in adult patients, but these
infections with blood transfusions remains real. Risk are rare in neonates. Newborns do not form red blood
of post transfusion hepatitis B/C in India is about cell (RBC) antibodies; all antibodies present are
10% in adults despite routine testing because of low maternal in origin.
viraemia and mutant strain undetectable by routine
(i) Newborns must be screened for maternal RBC
ELISA. 25 HIV prevalence among blood donors is
antibodies, including ABO antibodies if non-O
different in various parts of the country.
RBCs are to be given as the first transfusion.
CMV: Transfusion related CMV infections in
(ii) If the initial results are negative, no further testing is
newborns were initially identified in the year 1969,
needed for the initial 4 postnatal months.
and since then transfusion associated CMV
transmission is a well known entity. It has been Infants are at a higher risk of passive immune
reported that there is a seroconversion rate of 10-30% hemolysis from infusion of ABO-incompatible plasma
in preterm newborns transfused with CMV positive present in PRBC or platelet concentrates. Smaller
blood. Leukodepletion and selection of CMV quantities of ABO-incompatible plasma (less than 5
negative donors decreases the risk of transfusion mL/kg) are generally well tolerated. Newborns do not
transmitted CMV.26 manifest the usual symptoms of hemolysis that are
observed in older patients, such as fever, hypotension,
Bacterial infections: Bacteria in donor blood are
Indian Journal of Pediatrics, Volume 75May, 2008 493
38 24

Richa Jain et al

and flank pain. An acute hemolytic event may be There is no role for routine use of furosemide while
present as increased pallor, presence of plasma free transfusing newborns.
hemoglobin, hemoglobinuria, increased serum
Metabolic complications:29 These complications occur
potassium levels, and acidosis. Results of the direct
with large volume of transfusions like exchange
antiglobulin (Coombs) test may confirm the presence of
transfusions.
an antibody on the RBC surface. Treatment is mainly
supportive and involves maintenance of blood pressure a) Hyperkalemia: In stored blood, potassium levels
and kidney perfusion with intravenous saline bolus of tend to be high. It has been seen that after storage
10 to 20 mL/kg along with forced diuresis with for around 42 days, potassium levels may reach
furosemide. Enforcing strict guidelines for patient 50 meq/L in a RBC unit.30 Though small volume
identification and issue of blood; and minimizing transfusions o not have much risk of metabolic
human error is essential in preventing immune disturbances, large volume transfusions may lead
mediated hemolysis. to hyperkalemia. Washing PRBCs before
reconstituting with FFP before exchange
TRALI (Transfusion related acute lung injury): It refers to
transfusion helps in preventing this complication.
noncardiogenic pulmonary edema complicating
transfusion therapy. It is a common and under-reported b) Hypoglycemia: Blood stored in CPD blood has a
complication occurring after therapy with blood high content of glucose leading to a rebound rise
components. It has been associated with all plasma- in insulin release 1-2 hours after transfusion. This
containing blood products, most commonly whole may lead to hypoglycemia and routine
blood, packed RBCs, fresh-frozen plasma, and platelets. monitoring is necessary, particularly after
It has also been reported after the transfusion of exchange transfusion, after 2 and 6 hours, to
cryoprecipitate and IVIG. The most common symptoms ensure that this complication does not occur.
associated with TRALI are dyspnea, cough, and fever,
c) Acid- base derangements: Metabolism of citrate in
associated with hypo- or hypertension. It occurs most
CPD leads to late metabolic alkalosis. Metabolic
commonly with in the initial 6 hours after transfusion.
acidosis is an immediate complication occurring
The presence of anti-HLA and/or anti-granulocyte
in sick babies who cannot metabolize citrate.
antibodies in the plasma of donors is implicated in the
pathogenesis of TRALI. Diagnosis requires a high d) Hypocalcemia and hypomagnesemia are caused by
index of suspicion, and confirmation of donor serum binding of these ions by citrate present in CPD
cross-reacting antibodies against the recipient. blood.
Treatment is mainly supportive in this self-limiting
condition. 27, 28 Delayed complications

Febrile nonhemolytic transfusion reactions (FNHTR) are Alloimmunization: Alloimmunization is an


suspected in the absence of hemolysis with an increase uncommon occurrence before the age of 4 months,
in body temperature of less than 2C. For reactions and is caused by transfusion of blood products with
associated with a temperature rise of greater than 2C or are mismatched for highly immunogenic antigens
with hypotension, bacterial contamination also should like Rh. 31
be suspected and a Gram stain and microbial culture Transfusion associated graft versus host disease (TA-
performed on the remaining blood product. GVHD): Newborns are at risk for TA-GVHD if they
Allergic reactions : Allergic reactions are caused by have received intrauterine transfusions, exchange
presence of preformed immunoglobulin E antibody transfusions, or are very small, or immuno-
against an allergen in the transfused plasma, and are a compromised. Unchecked donor T cell proliferation
rare occurrence in newborns. In some cases, release of is the cause of TA-GVHD, and it can be effectively
residual cytokines or chemokines (e.g., RANTES) from prevented by leukoreduction of the transfused blood
stored platelets also may cause allergic reactions. These products in at risk patients.
reactions are generally mild, and respond to
antihistaminics. Severe anaphylactic reactions are rare. REFERENCES
Acute non immune reactions
1. Bell EF, Strauss RG, Widness JA, Mahoney LT, Mock DM et
Fluid overload: Neonates are at increased risk of fluid al. Randomized Trial of Liberal Versus Restrictive
overload from transfusion because the volume of the Guidelines for Red Blood Cell Transfusion in Preterm
Infants. Pediatrics 2005; 115 : 1685-1691.
blood component issued may exceed the volume that
2. Ohls R J. Transfusions in the Preterm Neonates. NeoReviews
may be transfused safely into neonates. Care should 2007;8 :377-386.
be taken to ensure that, in the absence of blood loss, 3. Murray NA, Roberts IAG. Neonatal transfusion practice.
volumes infused do not exceed 10 to 20 mL/kg. Arch Dis Child FN 2004; 89 : 101-107.

494 Indian Journal of Pediatrics, Volume 75May, 2008


39 25

Blood and Blood Component Therapy in Neonates

4. Chatterjee K, Sen A. Step by Step Blood Transfusion Services. 1st positive platelet transfusion in neonatal alloimmune
ed. New Delhi. Jaypee Publishers; 2006; 238-300. thrombocytopenia (NAIT). Blood 2006; 107 : 3761-3763.
5. Schroeder ML. Transfusion-associated graft-versus-host 19. Blanchette VS, Johnson J, Rand M. The management of
disease. Br J Haematol 2002; 117 : 275-287. alloimmune neonatal thrombocytopenia. Baillieres Clin
6. Moroff G, Holme S, AuBuchon JP, Heaton WA, Sweeney Haematol 2000; 13 : 365-390.
JD, et al. Viability and in vitro properties of AS-1 red cells 20. Kelton JG. Idiopathic thrombocytopenic purpura
after gamma irradiation. Transfusion 1999; 39: 128-134. complicating pregnancy. Blood Rev 2002; 16 : 43-46.
7. Pelszynsky MM, Moroff G, Luban NLC, Taylor BJ, Quinones 21. Galel SA Therapeutic techniques: Selection of Blood
RR. Effect of y Irradiation of Red Blood Cell Units on T-cell Components for Neonatal Transfusion. NeoReviews 2005; 6:
Inactivation as Assessed by Limiting Dilution Analysis: e351-e355.
Implications for Preventing Transfusion-Associated Graft- 22. Tucci M. Goal-directed blood transfusion therapies Current
Versus-Host Disease. Blood 1994; 83 : 1683-1689. Concepts in Pediatric Critical Care Refresher Course.
8. Paul DA, Leef KH, Locke RG, Stefano JL. Transfusion available at : http://sccmcms.sccm.org.accessed on april
volume in infants with very low birth weight: a randomized 15, 2008.
trial of 10 versus 20 ml/kg. J Pediatr Hematol Oncol 2002; 23. Choudhury LP, Tetali S. Ethical challenges in voluntary
24 : 43-46. blood donation in Kerala, India. J Med Ethics 2007; 33 : 140-
9. British Committee for Standards in Haematology. Available 142.
at: www.bcshguidelines.com. Accessed on April 20, 2008. 24. Madjdpour C, Heindl V, Spahn DR. Risks, benefits,
10. Roberts I,Murray NA. Neonatal thrombocytopenia: causes alternatives and indications of allogenic blood transfusion.
and management. Arch Dis Child FN 2003; 88 : F359-F364. Minerva Anestesiol 2006; 72 : 283-298.
11. Hohlfeld P, Forestier F, Kaplan C, Tissot JD, Daffos F. Fetal 25. Choudhury N, Phadke S. Transfusion transmitted diseases.
thrombocytopenia: a retrospective survey of 5,194 fetal Indian J Pediatr 2001; 68 : 951-958.
blood samplings. Blood 1994; 84 : 1851-1856. 26. Bowden RA, Slichter SJ, Sayers M, Weisdorf D, Cays M et al.
12. Burrows RF, Kelton JG. Incidentally detected A Comparison of Filtered Leukocyte-Reduced and
thrombocytopenia in healthy mothers and their infants. N Cytomegalovirus (CMV) Seronegative Blood Products for
Engl J Med 1988; 319 : 142-145. the Prevention of Transfusion-Associated CMV Infection
13. Sainio S, Jarvenpaa A-S, Renlund M, Riikonen S, Teramo K After Marrow Transplant. Blood 1995; 86 : 3598-3603.
et al. Thrombocytopenia in term infants: a population- 27. Yang X, Ahmed S, Chandrasekaran V. Transfusion-related
based study. Obstet Gynecol 2000; 95 : 441-446. acute lung injury resulting from designated blood
14. Uhrynowska M, Niznikowska-M M, Zupanska B. Neonatal transfusion between mother and child: a report of two cases.
and maternal thrombocytopenia: incidence and immune Am J Clin Pathol 2004; 121 : 590-592.
background. Eur J Haematol 2000; 64 : 42-46. 28. Looney MR, Gropper MA, Manhay MA. Transfusion-
15. Castle V, Andrew M, Kelton J, Girm D, Johston M et al. Related Acute Lung Injury. A Review. Chest 2004; 126: 249-
Frequency and mechanism of neonatal thrombocytopenia. J 258.
Pediatr 1986; 108 : 749-755. 29. Martin CR, Cloherty JP. Neonatal hyperbilirubinemia. In
16. Murray NA, Howarth LJ, McCloy MP, Letsky EA, Roberts Cloherty JP, Eichenwald ER, Stark AR, eds. Manual of
IAG. Platelet transfusion in the management of severe Neonatal Care. 5th ed. Philadelphia, Lippincott Willams and
thrombocytopenia in neonatal intensive care unit (NICU) Wilkins, 2004; 185-221.
patients. Transfus Med 2002; 12 : 35-41. 30. Strauss RG. Transfusion approach to neonatal anemia.
17. Rothenberger S. Neonatal alloimmune thrombocytopenia. NeoReviews 2000;1:e74-e80.
Ther Apher 2002; 6 : 32-35. 31. Galel S A, Fontaine MJ. Hazards of Neonatal Blood
18. Kiefel V, Bassler D, Kroll H, Paes B, Giers G et al. Antigen- Transfusion. NeoReviews 2006;7:e 69-e75.

Indian Journal of Pediatrics, Volume 75May, 2008 495


40
41
42
43
44 26

Symposium on AIIMS Protocols in Neonatology IV

Chronic Lung Disease in Newborns


M. Jeeva Sankar, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar,
New Delhi, India

ABSTRACT
Chronic lung disease (CLD) or bronchopulmonary dysplasia (BPD) occurs in preterm infants who require respiratory support
in the first few days of birth. Apart from prematurity, oxygen therapy and assisted ventilation, factors like intrauterine/postnatal
infections, patent ductus arteriosus, and genetic polymorphisms also contribute to its pathogenesis. The severe form of BPD
with extensive inflammatory changes is rarely seen nowadays; instead, a milder form characterized by decreased alveolar
septation due to arrest in lung development is more common. A multitude of strategies, mainly pharmacological and
ventilatory, have been employed for prevention and treatment of BPD. Unfortunately, most of them have not been proved to
be beneficial. A comprehensive protocol for management of BPD based on the current evidence is discussed here. [Indian
J Pediatr 2008; 75 (4) : 369-376] Email: ashokdeorari_56@hotmail.com

Key words : Bronchopulmonary dysplasia; Prematurity; Prevention; Treatment

Chronic lung disease (CLD) or bronchopulmonary supplemental oxygen at 36 weeks postmenstrual age
dysplasia (BPD) occurs in preterm infants who require (PMA) as the diagnostic criterion especially in preterm
mechanical ventilation and/or oxygen therapy for a very low birth weight (VLBW) infants. 4 The later
primary lung disorder in the initial few days after birth. definition, used widely in clinical trials even now, has the
Though the incidence of CLD has largely remained limitation of spuriously labeling more mature infants (e.g.,
unchanged over the years, the improved survival of more those born at 34-35 weeks) as having CLD.
immature infants has led to increased numbers of infants
To address the inconsistencies in the diagnostic criteria,
with this disorder.1 These infants are more likely to have
the US National Institute of Health (NIH) organized a
persistent respiratory symptoms requiring frequent
consensus conference in the year 2000 which suggested a
hospital admissions especially in the first two years after
new definition by incorporating many elements of the
birth.
previous definitions of BPD. The suggestion was to use
oxygen need for > 28 days and at 36 weeks PMA to
DEFINITION AND INCIDENCE identify different severity of BPD (Table 1).5
Recently, Ehrenkranz et al validated the consensus
The lack of uniformity in the diagnostic criteria for CLD definition in a cohort of preterm (<32 weeks) extremely
partly explains the wide variation in the reported low birth weight (ELBW) infants and reported an
incidence among different centers.2 The initial diagnostic incidence of 77% by the new criteria.6 Few reports are
criteria mandated continuing oxygen dependency during available from the centers in India; one study from
the first 28 days of life with compatible clinical and Chandigarh found the incidence of CLD (defined as need
radiographic findings to label an infant as having BPD.3 for oxygen at or beyond 28 days of life) to be 50% and 9%
The fact that many infants would have intervals in the in ELBW and VLBW infants respectively.7
first few weeks during which they do not require any
supplemental oxygen signified the major drawback of this
PATHOGENESIS
definition. Later, it was proposed to use the need for

CLD has a multifactorial etiology; the major risk factors


include prematurity, oxygen therapy, mechanical
Correspondence and Reprint requests : Prof. Ashok K. Deorari, ventilation, infection, patent ductus arteriosus (PDA), and
Professor, Department of Pediatrics, All India Institute of Medical genetic predisposition.8 By far the most important factor
Sciences, Ansari Nagar, New Delhi 110029, India.
in the pathogenesis of CLD is prematurity. Exposure of
[Received March 24, 2008; Accepted March 24, 2008]

Indian Journal of Pediatrics, Volume 75April, 2008 369


45 27

M.J. Sankar et al

TABLE 1. Definition of BPD5

Gestational age
< 32 weeks > 32 weeks

Time point of assessment 36 weeks PMA or discharge* > 28 days but < 56 days postnatal age or discharge*
Treatment with oxygen > 21% for at least 28 days > 21% for at least 28 days
BPD
Mild Breathing room air at 36 weeks PMA or Breathing room air at 56 days postnatal age or discharge*
discharge*
Moderate Need* for <30% oxygen at 36 weeks PMA Need* for <30% oxygen at 56 days postnatal age or
or discharge* discharge*
Severe Need for > 30% oxygen and/or positive Need for > 30% oxygen and/or positive pressure
pressure (IMV/CPAP) at 36 weeks PMA (IMV/CPAP) at 56 days postnatal age or discharge*
or discharge*

*- whichever comes first


(PMA, Postmenstrual age; BPD, bronchopulmonary dysplasia; IMV, Intermittent mandatory ventilation; CPAP, continuous positive
airway pressure)

immature lungs to high O2 concentrations and positive Radiographic features of old and new BPD are quite
pressure ventilation results in oxidative stress and different, not surprising given the vastly different
ventilator induced lung injury (barotrauma/volutruma). pathologic findings. Old BPD, as originally described by
The resulting injury and inflammation lead to abnormal Northway, had four distinct stages: stage 1, consistent
reparative processes in the lung. This is compounded by with hyaline membrane disease; stage 2, opaque lung
inflammation resulting from infections (intra-uterine/ fields with air bronchograms due to areas of atelectasis
postnatal) that occur commonly in these infants. PDA alternating with emphysema; stage 3, small radiolucent
contributes further to this process by inducing pulmonary cysts; and stage 4, hyperinflated lungs with generalized
edema and vascular endothelial injury. Recently, genetic cystic areas and dense fibrotic strands. 10 In contrast,
polymorphisms are also thought to play a role in the infants with new BPD show only haziness reflecting
causation of BPD.9 diffuse loss of lung volume or increased lung fluid.
Occasionally they have dense areas of segmental or lobar
atelectasis or pneumonic infiltrates, but they do not show
PATHOLOGY: OLD VS NEW BPD
severe over inflation.

The severe form of BPD (old BPD) seen in infants who


MANAGEMENT
received aggressive ventilation and were exposed to high
inspired oxygen concentration from birth is rare
nowadays. This form was characterized by severe Given the multitude of factors that contribute to the
morphological changes including emphysema, atelectasis, pathogenesis of bronchopulmonary dysplasia, it is not
and fibrosis and marked epithelial metaplasia and smooth surprising that there is no magic bullet for its prevention
muscle hypertrophy in the airways and in the pulmonary and/or treatment. Indeed, the best bet for prevention of
vasculature.10 BPD would be to prevent preterm births itself, an
implausible option as of now.
In contrast, the milder forms of BPD (new BPD) seen
today occurs in infants who had only mild respiratory
PREVENTION
failure requiring shorter duration of ventilation and/or
oxygen therapy immediately after birth. Pathologically,
Prevention requires a multidisciplinary approach starting
this form is characterized by a striking decrease in
right from the antenatal period.
alveolar septation and impaired vascular development,
changes more compatible with an arrest in lung
development than with mechanical injury.11 ANTENATAL PERIOD

CLINICAL AND RADIOLOGICAL FEATURES Use of antenatal steroids in mothers at risk for delivering
a premature infant reduces the incidence of neonatal
deaths and RDS but does not reduce the incidence of CLD.
Respiratory signs in infants with CLD include fast but This could arguably be due to increased survival of very
shallow breathing, retractions, and paradoxical breathing. immature infants who are at high risk of BPD or because
Rales and coarse rhonchi are usually heard on of the to inability to detect a real protective effect.12
auscultation. Antenatal thyrotropin-releasing hormone (TRH) has not

370 Indian Journal of Pediatrics, Volume 75April, 2008


46 28

Chronic Lung Disease in Newborns

been effective in prevention of BPD.13 HFV with conventional ventilation in preterm infants
have yielded conflicting results. A recent meta-analysis
that included 17 RCTs of conventional vs high frequency
AFTER BIRTH
ventilation found no significant difference in the incidence
of BPD. Therefore, elective use of HFV cannot be
A. Ventilatory strategies recommended for preterm infants with RDS at present.18

Given that no ideal pharmacological agents are available Volume targeted ventilation: The observation that
for prevention of BPD, attention has now shifted to volutrauma and not barotrauma is the primary
optimal ventilatory strategies that would prevent/ determinant of VILI has enthused neonatologists to use
reduce lung injury and permit adequate lung volume controlled/targeted modes of ventilation in place
development. of conventional pressure controlled modes. Only a few
randomized trials are available in this regard till date. The
Continuous positive airway pressure (CPAP): Early Cochrane review that included four RCTs found
initiation of nasal CPAP has been shown to reduce the significant reduction in the duration of ventilation and
need for intubation and mechanical ventilation. Since, pneumothorax rates but only a borderline reduction in the
need for mechanical ventilation is one of the major risk incidence of BPD (typical RR 0.34; 95% CI: 0.11, 1.05).19
factors for BPD, use of early CPAP should logically reduce The trials did not report the combined outcome of BPD
its incidence. Numerous studies, mostly non-randomized, and death as well as long term respiratory/
have reported the benefits of early CPAP in minimizing neurodevelopmental outcomes.
the need for mechanical ventilation and thes the incidence
of chronic lung disease.14 Surprisingly few randomized Permissive hypercapnia: Retrospective studies have
controlled trials are available till date in this regard. suggested that hypocapnia that occurs during assisted
Recently, a multi-centric study on CPAP vs intubation and ventilation is an independent risk factor for BPD.
ventilation in infants born at 25-28 weeks gestation found Subsequently, minimal ventilation using smaller tidal
significant reduction in the need for oxygen at 28 days of volumes / less peak inflation pressures while accepting
life but not at 36 weeks PMA.15 Similarly, extubation to mild hypercapnia (PaCO2 45-55 mm Hg) was studied in
CPAP following early surfactant administration preterm infants. One such study in preterm ELBW infants
(INtubateSURfactantExtubate) has been shown to reduce (target PaCO2 >52 mm Hg in the study group) reported
the need for mechanical ventilation but it is still uncertain less need for mechanical ventilation but no reduction in
if BPD is reduced by this approach. Clearly, more the need for supplemental oxygen at 36 weeks PMA.20
evidence is needed in this regard before coming to any Clearly, more studies are needed to prove the intended
meaningful conclusion. benefits of this promising strategy.

Nasal intermittent positive pressure ventilation (NIPPV): Permissive hypoxemia: Exposure to high oxygen
NIPPV is a method of augmenting NCPAP by delivering concentration has long been recognized as an important
ventilator breaths via the nasal prongs. It is thought to factor in the pathogenesis of BPD. Preterm infants are
improve the tidal and minute volumes and decrease the more vulnerable to the harmful effects of free oxygen
inspiratory effort required by neonates as compared to radicals. Surprisingly, there are few data to suggest either
nCPAP. This should reduce the need for reintubation thus the optimal oxygen level required or the optimum target
avoiding ventilator induced lung injury (VILI). The range for oxygen saturations (SpO 2) in these infants.
Cochrane review that included three RCTs found a trend Observational studies suggest that in comparison with the
towards reduction in the rates of chronic lung disease more liberal oxygen therapy, the restrictive approach of
with NIPPV (typical RR 0.73; 95% CI: 0.49, 1.07). 16 accepting lower oxygen saturation values is associated
However, more trials are required to document the safety with decreased incidences of CLD and ROP. Two RCTs
and effectiveness of this relatively new modality. have been conducted to evaluate whether it is better to
aim for high oxygen saturation in infants who are more
Patient-triggered ventilation (PTV): Patient triggered than a few weeks old: BOOST-trial and STOP-ROP trial.21,
modes (SIMV, assist-control, and pressure support 22
Both these studies indicate that maintaining higher
ventilation) improve the infant-ventilator asynchrony; this oxygen saturation (>95%) is associated with increased
should theoretically reduce the risk of VILI. The Cochrane need for oxygen at 36 weeks PMA and greater use of
review concluded that though PTV is associated with postnatal steroids and diuretics in premature infants
shorter duration of ventilation, it does not reduce the when compared to maintaining lower oxygen saturation
incidence of BPD.17 of 89-94%. Still, the uncertainty about optimal
High-frequency ventilation (HFV): Animal studies oxygenation has led to wide variation of policies on
indicate that HFV could lead to less lung injury when oxygen-monitoring and therapy in neonatal nurseries. In
compared to conventional ventilation. However, response to the growing demand to resolve this
randomized controlled trials comparing elective use of uncertainty, an international collaborative effort has been

Indian Journal of Pediatrics, Volume 75April, 2008 371


47 29

M.J. Sankar et al

mounted to conduct large multicentre RCTs, the results of vitamin A (in the above said dose) for ELBW infants with
which may help determine the optimal oxygen saturation respiratory distress requiring supplemental oxygen or
targets in very premature infants. mechanical ventilation at 24 hours of age.
B. Fluids and Nutrition (iii) Methylxanthines: Xanthines such as caffeine and
aminophylline have been routinely used for prevention/
Fluid restriction: Anecdotal data indicate that relative fluid
treatment of apnea and for facilitation of extubation in
restriction reduces the incidence of BPD in preterm
premature infants. Recently, a large RCT that used
infants. However, systematic review of the studies on
caffeine for these indications in infants with birth weight
fluid restriction has not found any significant reduction.23
of 500-1250g has shown a significant reduction in the
Moreover, what represents fluid restriction in VLBW
incidence of BPD. The authors attributed this rather
infants is not definitely known. Hence, no definite
unexpected finding to reduced duration of mechanical
recommendation can be made regarding fluid restriction
ventilation in the caffeine treated group. 28 However,
as a strategy for reducing the incidence of BPD.
caffeine is not available in India and one should be careful
Nutrition: Nutrition plays an important role in lung before extrapolating the beneficial effects to
development and maturation. Aggressive parenteral aminophylline. We use aminophylline after extubation
nutrition and early enteral feeding may help decrease the and for treatment of apnea of prematurity in preterm
incidence of BPD in VLBW infants.24 Ideally, nutritional VLBW infants.
management should begin from day one of life to
(iv) Indomethacin / Ibuprofen therapy for PDA: Patent
minimize the respiratory morbidities. The initial
ductus arteriosus is one of the major risk factors for BPD.
management should meet the estimated fluid, protein,
Hence, prevention or treatment of PDA should ideally
and energy needs. Since enteral feeding is often delayed
reduce its risk. However, prophylactic use of
in these infants due to gastrointestinal immaturity,
indomethacin in very low birth weight infants has failed
parenteral nutrition with proteins and lipids should be
to show any reduction in the incidence of BPD despite a
initiated as soon as possible after birth. It should be
significant reduction in the incidence of PDA.29 Similar
continued until daily oral intake reaches at least 130 mL/
results are obtained with ibuprofen, another drug used
kg. Only expressed breast milk is to be used for enteral
for closure of PDA. 30 In contrast, treatment of
feeding. Fortifying breast milk with human milk fortifier
symptomatic PDA could possibly reduce the incidence of
(HMF) will make up for deficiencies of protein and
BPD.31
minerals like calcium and phosphorus. If fluids need to be
restricted, addition of fat such as medium chain (v) Postnatal steroids: Given that inflammation plays a
triglyceride (MCT) oil or glucose polymers will help in central role in the pathogenesis of BPD, steroids were
achieving the adequate growth. thought to be a natural choice for its management.
However, this therapy has turned out to be the most
The role of specific nutrients (e.g., inositol, vitamin E,
controversial area of care following reports of adverse
selenium, glutamine etc. except for vitamin A) however,
neurodevelopmental outcomes. Conventionally, steroid
remains speculative till now.
therapy for BPD is categorized into 3 broad groups based
C. Pharmacological strategies on the timing of initiation: early (during the first 96 hrs
after birth), moderately early (between postnatal days 7
(i) Exogenous surfactant: Prophylactic surfactant therapy in
and 14), and delayed (after 3 weeks of age). Meta-analyses
infants born before 30 weeks of gestation has not been
of RCTs of the first two regimes have shown a significant
shown to reduce the incidence of BPD. However,
reduction in the incidence of BPD at 36 weeks PMA.32, 33
surfactant treatment for established RDS (rescue therapy)
However, there are important concerns regarding both
in infants born at or after 30 weeks gestation is associated
short-term (hypertension, gastrointestinal perforation,
with significant reduction in the incidence of BPD.25 The
poor somatic growth) as well as long-term adverse effects
apparent lack of effect in the first group could probably be
(neurodevelopmental outcomes including cerebral palsy).
due to the increased survival of more immature infants
One systematic review in this regard concluded that
(similar to antenatal steroids).
infants who received steroids were twice as likely to
(ii) Vitamin A: Vitamin A is essential for maintaining develop cerebral palsy as the control infants.34 In view of
the integrity of respiratory tract epithelial cells. Very these findings, routine early use of high-dose steroids is
preterm infants are relatively deficient in vitamin A which not recommended at present. Considering the fact that no
has been shown to be associated with CLD. A large RCT other treatment options have proved to be consistently
of 807 infants with a birth weight of less than 1000 g has beneficial in preventing BPD, some centers still
shown that intramuscular vitamin A (5000 units three recommend moderately early use of steroids at lower
times a week for 4 weeks from birth) decreases the risk of doses and for shorter duration in ventilator-dependent
CLD (OR 0.89; 95%CI 0.8-0.99).26 A meta-analysis of seven infants. We use steroids occasionally in ELBW infants
RCTs has confirmed this finding.27 We use intramuscular who continue to be on mechanical ventilation even after

372 Indian Journal of Pediatrics, Volume 75April, 2008


48 30

Chronic Lung Disease in Newborns

10-14 days of life (3-day course using low dose prevention and treatment of BPD have not shown any
dexamethasone). 35 benefit.41
Inhaled steroids thought to reduce the adverse effects (ix) Bronchodilators: They have not been found to be
associated with systemic administration have not been useful for prevention of BPD. 42
shown to be beneficial either for prevention or for
(x) Emerging therapies: Preterm infants are susceptible to
treatment.36
oxidant injury because they are deficient in antioxidant
(vi) iNO: Animal studies have shown that iNO therapy, enzymes. Hence, antioxidants such as superoxide
in addition to causing pulmonary vasodilatation also dismuatase (SOD) promise to be an exciting strategy for
reduces lung inflammation and promotes lung growth. prevention of BPD. A randomized trial that enrolled
Unfortunately, most clinical studies in preterm infants around 300 infants proved the safe nature of the drug
with severe respiratory failure have not demonstrated any CuZnSOD, but did not find any difference in the primary
reduction in the risk of death or CLD with iNO. 37 outcome of BPD at 36 weeks PMA. Interestingly, SOD
Recently, two large RCTs conducted in this regard treated infants had fewer episodes of respiratory illness at
indicate that iNO therapy might be beneficial in a select I year of age suggesting that the drug could prevent long-
group of preterm infants. 38, 39 However, one should term lung injury caused by reactive oxygen species.43
remember that the appropriate dose, timing, duration, Further studies are needed to define its exact role in the
and more importantly, the subgroup of infants who are management of BPD. Other antioxidants/free radical
likely to benefit with this mode of therapy have not yet scavengers like vitamins C and E, allopurinol, N-acetyl-
been clearly defined. Moreover, the prohibitive cost of Cysteine have not been proved to be useful till now.
iNO therapy precludes its use on a routine basis.
The options available for prevention and their current
(vii) Diuretic therapy: Diuretics help by increasing the status are summarized in table 2.
reabsorption of fluid from the lungs. Though studies
have shown beneficial effects in lung physiology, no such
TREATMENT OF EVOLVING/ESTABLISHED BPD
effects were observed in mortality or the incidence of
BPD. Coupled with the potential risks involved with long
term therapy, chronic use of furosemide or any other There are extremely limited data from clinical trials on
diuretics cannot be recommended now. However, which to base optimal ventilatory management in
diuretics can be used sparingly if there are clinical/ established BPD. The major goal is to maintain adequate
radiographic features of pulmonary edema in an infant gas exchange with as minimal support as possible. CPAP
with evolving or established BPD.40 We use furosemide and NIPPV should be attempted as much as possible. For
0.5-1 mg/kg/day in infants with features suggestive of infants on ventilator, the settings should be titrated
excess lung fluid; we stop it after 24-48 hours if no keeping in mind the rapidly changing pulmonary
improvement is noted in the clinical condition. mechanics (increasing airway resistance as well as
(viii) Mast cell stabilizers: Cromolyn sodium has been improving compliance). Often, slow rates with long Ti are
shown to decrease neutrophil migration and activation needed as the disease progresses. Some neonates with
thus minimizing inflammation in the lungs. Two trials marked variability in compliance and resistance may
that have studied the possible role of cromolyn for benefit from volume targeted ventilation. Similarly, PTV

TABLE 2. BPD - Preventive Strategies and Their Current Status

Strategies Proven benefit Promising Probably beneficial No benefit


(needs more studies) (effects + )

Ventilatory - NIPPV Early CPAP High frequency


Volume targeted ventilation Patient triggered modes ventilation
Permissive hypercapnia
Permissive hypoxemia
Fluids and nutrition - Aggressive early enteral and Fluid restriction
parenteral nutrition
Pharmacological Vitamin A Superoxide dismutase Antenatal steroids Antenatal TRH
Postnatal steroids Exogenous surfactant Prophylactic
(but harmful as well) Methylxanthines indomethacin/ibuprofen
iNO therapy Inhaled steroids
Diuretics Bronchodilators
Mast cell stabilizers

(NIPPV, Nasal intermittent positive pressure ventilation; CPAP, continuous positive airway pressure; iNO, Inhaled nitric oxide; TRH,
Thyrotropin releasing hormone)

Indian Journal of Pediatrics, Volume 75April, 2008 373


49 31

M.J. Sankar et al

may be useful in infants who fight the ventilator.44 agitation.


Accepting relatively high PaCO2 (45-55 mm Hg provided
Infants developing BPD require 20 to 40% more
pH >7.25) and slightly low saturations (88-93%) would
calories than their age-matched healthy controls. Their
help in minimizing the ventilator settings and thus help in
caloric requirement varies from 120 to 150 Kcal/kg/day.
early extubation (Table 3).
This can be achieved by fortifying breast milk with
The role of drugs in evolving/established BPD is human milk fortifier (HMF) or infant formula. For infants
minimal except in select group of infants (Table 3). Most who require more calories, fat supplementation (e.g. MCT
of the drugs used in this regard have already been oil) is preferable to adding carbohydrates because of the
discussed under prevention of BPD. Diuretics and less pronounced effects on CO2 levels (Table 3).24
bronchodilators can be used if the clinical condition
Fig. 1 summarizes the steps of prevention and
warrants but should be stopped if no response occurs
treatment of BPD (starting from the antenatal period until
within 24-48 hours of initiation of therapy. This is
the time of discharge) in a preterm VLBW infant.
especially important in case of diuretic therapy. Steroids
initiated after 3 weeks of life (late regime) does not reduce
the need for oxygen at 36 weeks PMA but definitely REFERENCES
reduces the need for home oxygen therapy. However, it is
associated with increased incidence of growth failure and 1. Smith VC, Zupancic JA, McCormick MC, Croen LA, Greene J,
hypertension.45 One should weigh the risk-benefit ratio Escobar GJ, Richardson DK. Trends in severe BPD rates
before initiating steroids for an infant with established between 1994 and 2002. J Pediatr 2005; 146 : 469-473.
2. Bancalari E, Claure N. Definitions and diagnostic criteria for
BPD. Infants with BPD spells (sudden episodes of
bronchopulmonary dysplasia. Semin Perinatol 2006; 30 : 164-
deterioration due to marked expiratory airflow limitation) 170.
may require sedation and muscle relaxation to reduce 3. Bancalari E, Abdenour GE, Feller R, Gannon J.

TABLE 3. Management of Evolving or Established BPD#

Evolving BPD Established BPD


(2-4 weeks age) (>4 weeks age)

Ventilatory strategies Minimizing ventilatory support Minimizing ventilatory support


(e.g. using nCPAP whenever possible) Tolerating higher PaCO2 (55-60 mm Hg provided
Tolerating slightly higher PaCO2 (45-55 mm Hg pH >7.25)
provided pH >7.25) Target SpO2 : 89-94%
Target SpO2 : 88-93% If on IMV:
If on IMV: o Use PTV if possible
o Use PTV if possible o Slow rates (20-40/min)
o Slow rates (25-40/min) o Moderate PEEP (4-8 cm H2O)
o Moderate PEEP (4-5 cm H2O) o Longer Ti (0.4-0.7 sec)
o Moderate Ti (0.35-0.45 sec) o Larger tidal volume (5-8 mL/kg)
o Low tidal volume (3-6 mL/kg)
o Early extubation to CPAP

Pharmacological strategies Methylxanthines to facilitate extubation Steroids:* Individualize based on the clinical
Steroids:* Consider in ELBW infants on ventilator condition
support even after 10-14 days of age
Specific management: Specific management:
o Diuretics for features of pulmonary edema o Bronchodilators for bronchospasm
o Bronchodilators for bronchospasm o Sedation and muscle relaxation for BPD
spells

Others Nutrition:
o Increase daily calorie intake to 120 to 150 Kcal/ Same as for evolving BPD
kg/d
o Give expressed breast milk fortified with HMF
o Use fat supplementation (e.g. MCT oil) for
providing additional calories
o Give multivitamin supplements to meet RDA
#
Modified from Reference 44
* Could result in potentially harmful effects including adverse neurodevelopmental outcomes; counsel parents before initiation of therapy
(nCPAP, nasal continuous positive airway pressure ;IMV, intermittent mandatory ventilation; PTV, patient triggered ventilation; PEEP,
positive end expiratory pressure; Ti, Inspiratory time; HMF, human milk fortifier; MCT, medium chain triglycerides; RDA, recommended
dietary allowance)

374 Indian Journal of Pediatrics, Volume 75April, 2008


50 32

Chronic Lung Disease in Newborns

Antenatal period Antenatal steroids (2 doses of betamethasone 24 hrs apart)

At birth Avoid excessive pressure during resuscitation (use appropriate size bag for BMV)

Birth to 24 hrs
Fluids: 60-80 mL/kg/d
Nutrition: oral feeds - breast milk (MEN/full feeds) to be initiated in stable infants
Early CPAP
If on ventilator:
o Early rescue surfactant as indicated
o Settings: fast rates (50-60/min), moderate PEEP (4-5 cm H2O), short Ti (0.25-0.4 s)
o Target values- SpO2: 87-92%; PaCO 2 45-55 mm Hg; pH: 7.25-7.35
o Early extubation to CPAP
o Methylxanthines to facilitate extubation

Fluids: daily increment of 15-20 mL/kg/d to reach a maximum of 150 mL/kg/d by


24 hrs to 1 week
day7
Nutrition:
o Parenteral: TPN for ELBW infants till full enteral feeds are achieved
o Enteral: Gradually increase feed volume by 20-30 mL/kg/d if accepting well; give
only breast milk; fortify with HMF after reaching 100 mL/kg/d
If on ventilator:
o Settings and target values as above
o Extubate to CPAP if possible
o Methylxanthines to facilitate extubation
For ELBW infants on oxygen or ventilator support at 24 hrs: Inj. vitamin A 5000 units
IM thrice weekly for 4 weeks

1 to 4 weeks Fluids: 150 to 160 mL/kg/d


Nutrition: Fortify breast milk with HMF; add more calories if needed
If on ventilator:
o Settings and target values as in Table 3
o Extubate to CPAP as early as possible
Diuretics/steroids/bronchodilators as indicated (see Table 3)

> 4 weeks Fluids: 150 to 160 mL/kg/d


Nutrition: Fortify breast milk with HMF; add more calories if needed
If on ventilator:
o Settings and target values as in Table 3
o Extubate to CPAP as early as possible
Sedatives/steroids/bronchodilators as indicated (see Table 3)

Fig. 1. Flow-chart for management of BPD


(BMV, bag and mask ventilation; CPAP, continuous positive airway pressure; PEEP, positive end expiratory pressure; MEN, Minimal
enteral nutrition; ELBW, extremely low birth weight infants; TPN, total parenteral nutrition; HMF, human milk fortifier; PTV, patient
triggered ventilation; Ti, Inspiratory time)

Bronchopulmonary dysplasia: clinical presentation. J Pediatr Pediatrics 2005; 116 : 1353-1360.


1979; 95 : 819-823. 7. Narang A, Kumar P, Kumar R. Chronic Lung Disease in
4. Shennan AT, Dunn MS, Ohlsson A, Lennox K, Hoskins EM. Neonates: Emerging problem in India. Indian Pediatr 2002; 39:
Abnormal pulmonary outcomes in premature infants: 158-162.
prediction from oxygen requirement in the neonatal period. 8. Kinsella JP, Greenough A, Abman SH. Bronchopulmonary
Pediatrics 1988; 82 : 527-532. dysplasia. Lancet 2006; 29; 367 : 1421-1431.
5. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J 9. Makri V, Hospes B, Stoll-Becker S, Borkhardt A, Gortner
Respir Crit Care Med 2001; 163 : 1723-1729. L.Polymorphisms of surfactant protein B encoding gene:
6. Ehrenkranz RA, Walsh MC, Vohr BR, Jobe AH, Wright LL, modifi ers of the course of neonatal respiratory distress. Eur J
Fanaroff AA, Wrage LA, Poole K; National Institutes of Child Pediatr 2002; 161 : 604-608.
Health and Human Development Neonatal Research 10. Northway WH Jr, Rosan RC, Porter DY: Pulmonary disease
Network. Validation of the National Institutes of Health following respiratory therapy of hyaline-membrane disease.
consensus definition of bronchopulmonary dysplasia. Bronchopulmonary dysplasia. N Engl J Med 1967; 276 : 357.

Indian Journal of Pediatrics, Volume 75April, 2008 375


51 33

M.J. Sankar et al

11. Jobe AH. The New BPD: an arrest of lung development. infants. Cochrane Database Syst Rev 2002; 4: CD000501.
Pediatr Res 1999; 46 : 641-643. 28. Schmidt B, Roberts RS, Davis P, Doyle LW, Barrington KJ,
12. Crowley PA. Antenatal corticosteroid therapy: a meta-analysis Ohlsson A et al. Caffeine for Apnea of Prematurity Trial
of the randomized trials, 1972 to 1994. Am J Obstet Gynecol Group. Caffeine therapy for apnoea of prematurity. N Engl J
1995; 173 : 322-335. Med 2006; 354 : 2112-2121.
13. Crowther CA, Alfi revic Z, Haslam RR. Thyrotrophin 29. Schmidt B, Davis P, Moddemann D, Ohlsson A, Roberts RS,
releasing hormone added to corticosteroids for women at risk Saigal S et al. Trial of Indomethacin Prophylaxis in Preterms
of preterm birth for preventing neonatal respiratory disease. (TIPP) Investigators. Long-term effects of indomethacin
Cochrane Database Syst Rev 2004; 2: CD000019. prophylaxis in extremely-low-birth-weight infants. N Engl J
14. Avery ME, Tooley WH, Keller JB, Hurd SS, Bryan MH, Cotton Med 2001; 344 : 1966-1972.
RB et al. Is chronic lung disease in low birth weight infants 30. Shah SS, Ohlsson A. Ibuprofen for the prevention of patent
preventable? A survey of eight centers. Pediatrics 1987; 79 : 26- ductus arteriosus in preterm and/or low birth weight infants.
30. Cochrane Database Syst Rev 2006;(1):CD004213.
15. Morley CJ, Davis PG, Doyle LW, Brion LP, Hascoet JM, Carlin 31. Clyman RI. Recommendations for the postnatal use of
JB; COIN Trial Investigators. Nasal CPAP or intubation at indomethacin: an analysis of four separate treatment
birth for very preterm infants. N Engl J Med 2008;14; 358 : 700- strategies. J Pediatr 1996; 128 : 601-607.
708. 32. Halliday HL, Ehrenkranz RA. Early postnatal (< 96 hours)
16. Davis PG, Lemyre B, De Paoli AG. Nasal intermittent positive corticosteroids for preventing chronic lung disease in preterm
pressure ventilation (NIPPV) versus nasal continuous positive infants. Cochrane Database Syst Rev 2003; (1): CD001146.
airway pressure (NCPAP) for preterm neonates after 33. Halliday HL, Ehrenkranz RA. Moderately early (714 days)
extubation. Cochrane Database of Systematic Reviews 2001; 3 : postnatal corticosteroids for preventing chronic lung disease
CD003212. in preterm infants. Cochrane Database Syst Rev 2003; (1):
17. Greenough A, Dimitriou G, Prendergast M, Milner AD. CD001144.
Synchronized mechanical ventilation for respiratory support 34. Barrington KJ. The adverse neuro-developmental effects of
in newborn infants. Cochrane Database of Systematic Reviews postnatal steroids in the preterm infant: a systematic review of
2008; 3 : CD000456. RCTs. BMC Pediatrics 2001; 1: 114.
18. Thome UH, Carlo WA, Pohlandt F. Ventilation strategies and 35. Sankar MJ, Deorari AK. Postnatal corticosteroids for chronic
outcome in Randomised Trials of High Frequency Ventilation. lung disease (CLD). Indian Pediatr 2007; 44 : 531-539.
Arch Dis Child 2005; 90 : F466-F473. 36. Shah V, Ohlsson A, Halliday HL, Dunn MS. Early
19. McCallionN, Davis PG, MorleyCJ. Volume-targeted versus administration of inhaled corticosteroids for preventing
pressure-limited ventilation in the neonate. Cochrane Database chronic lung disease in ventilated very low birth weight
of Systematic Reviews 2005; 3: CD003666. preterm neonates (Cochrane Review). Cochrane Database Syst
20. Carlo WA, Stark AR, Wright LL, Tyson JE, Papile LA, Rev 2000; 2: CD001969.
Shankaran S et al. Minimal ventilation to prevent 37. Barrington KJ, Finer NN. Inhaled nitric oxide for respiratory
bronchopulmonary dysplasia in extremely-low-birth-weight failure in preterm infants. Cochrane Database Syst Rev 2006:
infants. J Pediatr 2002; 141 : 370-374. CD000509.
21. Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM. 38. Kinsella JP, Cutter GR, Walsh WF, Gerstmann DR, Bose CL et
Oxygen-saturation targets and outcomes in extremely preterm al. Early inhaled nitric oxide therapy in premature newborns
infants. N Engl J Med 2003; 4; 349 : 959-967. with respiratory failure. N Engl J Med 2006; 355 : 354-364.
22. Supplemental Therapeutic Oxygen for Prethreshold 39. Ballard RA, Truog WE, Cnaan A, Martin RJ, Ballard PJ et al.
Retinopathy of Prematurity (STOP-ROP), a randomized, Inhaled nitric oxide in preterm infants undergoing mechanical
controlled trial. I: primary outcomes. Pediatrics 2000; 105 : 295- ventilation. N Engl J Med 2006; 355 : 343-353.
310. 40. Baveja R, Christou H. Pharmacological strategies in the
23. Barrington KJ, Al-Hazzani FN. Fluid restriction for treatment prevention and management of bronchopulmonary dysplasia.
of preterm babies with chronic lung disease. (Protocol) Semin Perinatol 2006; 30 : 209-218.
Cochrane Database of Systematic Reviews 2005; (3) : CD005389. 41. Ng GY, Ohlsson A. Cromolyn sodium for the prevention of
24. Biniwale MA, Ehrenkranz RA. The role of nutrition in the chronic lung disease in preterm infants. Cochrane Database Syst
prevention and management of bronchopulmonary dysplasia. Rev 2001; (2) : CD003059.
Semin Perinatol 2006; 30 : 200-208. 42. Ng GY, da S, Ohlsson A. Bronchodilators for the prevention
25. Engle WA; American Academy of Pediatrics Committee on and treatment of chronic lung disease in preterm infants.
Fetus and Newborn. Surfactant-replacement therapy for Cochrane Database Syst Rev 2001; (3) : CD003214.A
respiratory distress in the preterm and term neonate. Pediatrics 43. Davis JM, Parad RB, Michele T et al. Pulmonary outcome at
2008; 121 : 419-432. one year corrected age in premature infants treated at birth
26. Tyson JE, Wright LL, Oh W, Kennedy KA, Mele L, with recombinant CuZn superoxide dismutase. Pediatrics
Ehrenkranz RA, Stoll BJ, Lemons JA, Stevenson DK, Bauer 2003; 111 : 469-476.
CR, Korones SB, Fanaroff AA. Vitamin A supplementation 44. Ambalavanan N, Carlo WA. Ventilatory strategies in the
for extremely-low-birth-weight infants. National Institute of prevention and management of bronchopulmonary dysplasia.
Child Health and Human Development Neonatal Research Semin Perinatol 2006; 30 : 192-199.
Network. N Engl J Med 1999; 340 : 1962-198. 45. Halliday HL, Ehrenkranz RA. Delayed (3 weeks) postnatal
27. Darlow BA, Graham PJ. Vitamin A supplementation for corticosteroids for chronic lung disease in preterm infants.
preventing morbidity and mortality in very low birth weight Cochrane Database Syst Rev 2003; (1): CD001145.

376 Indian Journal of Pediatrics, Volume 75April, 2008


52
53
54
55
56 34

Symposium on AIIMS Protocols in Neonatology IV

Congenital Hypothyroidism
Vandana Jain1, Ramesh Agarwal2, Ashok K. Deorari2 and Vinod K. Paul2
1
Division of Pediatric Endocrinology, 2Division of Neonatology, Department of Pediatrics, All India Institute of
Medical Sciences Ansari Nagar, New Delhi, India

ABSTRACT
Congenital Hypothyroidism (CH) is one of the most common preventable causes of mental retardation with a worldwide
incidence of 1:4000 live births. Ideally universal screening at 3-4 days of age should be done for detecting CH. Abnormal
values on screening (T4 < 6.5 ug/dL, TSH > 20mu/L) should be confirmed by a venous sample (using age appropriate cut-
offs) before initiating treatment. Term as well as preterm infants with low T4 and elevated TSH should be started on L-thyroxine
at a dose of 10-15g/ kg/ day as soon as the diagnosis is made. Regular monitoring should be done to ensure that T4 is in
the upper half of normal range. The outcome of CH depends on the time of initiation of therapy and the dose of L-thyroxine
used with the best outcome in infants started on treatment before 2 weeks of age with a dose > 9.5g/ kg/ day. [Indian J
Pediatr 2008; 75 (4) : 363-367] Email: aranag@rediffmail.com

Key words : Congenital hypothyroidism; L-thyroxine; Newborn

Congenital Hypothyroidism (CH) is one of the most increased local availability of the physiologically more
common preventable causes of mental retardation. The important T3. Near normal cognitive outcome is possible
worldwide incidence is 1:4000 live births and the in even the most severely affected infants with CH as long
estimated incidence in India is 1:2500-2800 live births.1 as postnatal therapy is initiated early in optimum doses
Thyroid dysgenesis is the commonest cause accounting and maternal thyroid function is normal. In contrast,
for 75-80% of all cases of CH. when both maternal and fetal hypothyroidism are present
as in severe iodine deficiency, there is a significant
Embryology and physiology of the thyroid in the fetus: The
impairment in neuro-intellectual development despite
thyroid gland originates as a proliferation of endodermal
adequate therapy soon after birth.2
epithelial cells at 3 to 4 weeks of gestation. Synthesis and
secretion of thyroxine (T4) and triiodothyronine (T3) starts Neonatal physiology: After birth, the term baby
from 12 weeks of gestation. Hypothalamic-pituitary- experiences a surge of TSH as a physiological response to
thyroid (HPT) axis begins to develop in the first trimester cold environment. The TSH concentration can rise to 80
and thyrotropin-releasing hormone (TRH) and thyroid mU/L and falls quickly in the first 24 hours, followed by
stimulating hormone (TSH) are also detectable by the end a slower decrease to below 10 mU/L after the first
of first trimester. However, the activity of HPT axis is low postnatal week. The rise in TSH initiates increase of T4
with insufficient production of thyroid hormones by the and free T4(fT4) to 17 g/dL and 3.5 ng/dL, respectively
fetus until about 18-20 weeks of gestation. In the second at 24 to 36 hours after birth with a slow decline to adult
half of gestation, the T4 and TSH levels increase values over 4-5 weeks. Preterm infants demonstrate a
progressively. In the first trimester, the fetus is dependent similar but blunted response due to HPT axis immaturity.
on transplacental passage of thyroid hormones.
In the hypothyroid fetus, this transplacental passage of
maternal thyroid hormones is critical for neuroprotection ETIOLOGY OF CH
throughout the intra-uterine life. The cord blood T 4
concentration at birth in infants who are unable to CH can be permanent or transient. Thyroid dysgenesis is the
synthesize T4 is 30% of normal. In addition, there is commonest cause of permanent CH affecting 1 in 4000
increased intracerebral conversion of T4 to T3, resulting in live births. It is usually sporadic with a 2:1 female to male
preponderance. The cause is largely unknown but
maternal cytotoxic antibodies and genetic mutations
Correspondence and Reprint requests : Dr. Ramesh Agarwal, causing inactivation of thyroid receptor are sometimes
Assistant Professor, Department of Pediatrics, All India Institute of found (Table 1).
Medical Sciences, Ansari Nagar, New Delhi 110029, India.
[Received March 17, 2008; Accepted March 17, 2008] Thyroid hormone synthetic defects account for 10% of all

Indian Journal of Pediatrics, Volume 75April, 2008 363


57 35

V. Jain et al

cases. These are inherited as autosomal recessive TSH concentrations in the context of low T3 and in the
disorders. The defect can lie in iodide trapping or more severe cases, low T4 concentrations.
organification, iodotyrosine coupling or deiodination and
thyroglobulin synthesis or secretion. The commonest of
DIAGNOSIS
these is a defect in the thyroid peroxidase (TPO) activity
leading to impaired oxidation and organification of iodide
to iodine. These disorders usually result in goitrous Newborn screening: Ideally universal screening at 3-4 days
hypothyroidism. Iodine deficiency is responsible for of age should be done for detecting CH. Alternatively
endemic cretinism and hypothyroidism in some regions cord blood can also be used if screening is being done
of India. only for CH and not other inborn errors of metabolism.
TABLE 1. Etiology of CH Universal newborn screening is currently being done in
many parts of the world including Western Europe,
1. Permanent hypothyroidism. North America, Japan, Australia, and parts of Eastern
b. Thyroid dysgenesis (aplasia, hypoplasia or ectopia). Europe, Asia, South America, and Central America. Three
c. Thyroid hormone biosynthetic defects.
d. Iodine deficiency (endemic cretinism).
approaches are being used for screening:
e. Hypothalamic-pituitary hypothyroidism. Primary TSH, back upT4
2. Transient hypothyroidism.
a. TSH binding inhibitory immunoglobulins Primary T4, back up TSH
b. Exposure to goitrogens (iodides or antithyroid drugs). Concomitant T4 and TSH
c. Transient hypothyroxinemia of prematurity
d. Sick euthyroid syndrome In the first approach, TSH is measured first. T4 is
measured only if TSH is > 20mu/L. This approach is
Hypothalamic- pituitary hypothyroidism has an incidence likely to miss central hypothyroidism, thyroid binding
of 1 in 100,000. It may be isolated or associated with globulin deficiency and hypothyroxinema with delayed
concomitant deficiency of other pituitary hormones and elevation of TSH. In the second approach, T4 is checked
present with hypoglycemia and microphallus. Transient first and if low TSH is also checked. This is likely to miss
hypothyroidism due to transplacental transfer of TSH milder/ subclinical cases of CH in which T4 is initially
binding inhibitory immunoglobulins (TBII) from mothers normal with elevated TSH. Concomitant measurement of
with autoimmune thyroid disease is seen in 1: 50,000 T4 and TSH is the most sensitive approach but incurs a
births. Their effect wanes off by 3-6 months in the higher cost.5
majority but may last up to 9 months. Exposure to iodine in
Abnormal values on screening (T4 < 6.5 g/dL, TSH>
sick preterm infants (e.g., application of povidone iodine
20mU/L) should always be confirmed by a venous
for skin disinfection) or intake of iodine containing
sample (using age appropriate cut-offs given in
expectorants by pregnant mothers can lead to transient
Table 3)6, 7 before initiating treatment. Among infants with
hypothyroidism.
proven CH, TSH is >50 mU/L in 90% and T4 is < 6.5 g/
Transient hypothyroxinema of prematurity refers to low dL in 75% of cases.
serum concentration of thyroid hormones in up to 85% of
In the absence of universal screening, the newborns
preterm infants in early postnatal life as compared to term
with the following indications should be screened:
infants. The normal levels of fT4 and TSH in preterm
infants are presented in Table 2. 3 This reflects the Family history of CH
underdevelopment of the HPT axis, which cannot History of thyroid disease or antithyroid medicine
compensate for the loss of maternal thyroid hormone in intake in mother
preterm infants. There has been a concern that transient
Presence of other conditions like Downs syndrome,
hypothyroxinemia is associated with adverse
trisomy 18, neural tube defects, congenital heart
neurodevelopmental outcomes and decreased survival in
disease, metabolic disorders, familial autoimmune
affected infants.4
disorders and Pierre- Robbins syndrome which are
Sick euthyroid syndrome reflects suppression of the associated with higher prevalence of CH
pituitarys response to TRH, with inappropriately low
Thyroid functions should be tested in any infant with
TABLE 2. Reference Ranges for Serum Free T4 (fT4) and TSH in signs/ symptoms of hypothyroidism such as prolonged
Preterm Infants jaundice, constipation, poor feeding, umbilical hernia,
Age in weeks Free T4 (ng/dl) TSH (mu/l) macroglossia, wide open posterior fontanel and
edematous and dry skin. The test results should be
25-27 06-2.2 0.2-30.3 compared to the age related norms as presented in
28-30 0.6-3.4 0.2-20.6
table 3.
31-33 1.0-3.8 0.7-20.9
34-36 1.2-4.4 1.2-21.6 Once the diagnosis is established, further

364 Indian Journal of Pediatrics, Volume 75April, 2008


58 36

Congenital Hypothyroidism

TABLE 3. Reference Ranges for T4, fT4 and TSH in Term Infants newborns) TBG deficiency. TBG levels should be
According to Age 6, 7 evaluated to confirm this but this test is not available
Age T4 (g/dl) fT4 (pg/ml) TSH (U/ml) routinely. If free T4 is also low along with low T4
mean (range) mean (SD)/ range mean (range) with normal TSH, central hypothyroidism should be
suspected.
Cord blood 10.8 (6.6-15) 13.8 (3.5) 10.0 (1-20)
1-3 days 16.5 (11-21.5) * 5.6 (1-10) During monitoring for adequacy of treatment, we
4-7 days * 22.3 (3.9) * usually monitor with T4 (total) level. This assumes a
1-2 weeks 12.7 (8.2-17.2) * 2.3 (0.5-6.5) normal TBG level. This can be confirmed by
2-6 weeks 6.5-16.3** 0.9-2.2 1.7-9.1**
6 weeks to 11.1 (5.9-1.3) * 2.3 (0.5-6.5) measuring free T4 or TBG levels once at the time of
12 months the first post-treatment T4 measurement.

*No data available ** data for median/mean not available


TREATMENT OF CH
investigations to determine the etiology may be done.
These are considered optional because the initial Term as well as preterm infants with low T4 and elevated TSH
treatment is not altered by these. These investigations are should be started on L-thyroxine as soon as the diagnosis
useful in infants with borderline thyroid function test is made. The initial dose of L-thyroxine should be 10-
results and in determining whether CH is likely to be 15g/ kg/ day with the aim to normalize the T4 level at
transient or permanent. 8 A list of these diagnostic studies the earliest. Those infants with severe hypothyroidism
is presented in table 4 and an algorithmic approach to (very low T4, very high TSH and absence of distal femoral
investigation in Fig. 1. If it is not possible to get the and proximal tibial epiphyses on radiograph of knee)
investigations performed immediately, the therapy should be started with the highest dose of 15g/ kg/
should be started without any delay.9 day.11
TABLE 4. Diagnostic Studies for Evaluation of CH
Monitoring of Therapy : T4 should be kept in the upper half
1. Imaging Studies (will determine location and size) of normal range (10-16 g/dL) or free T4 in the 1.4 - 2.3
a. Scintigraphy (99mTc or 123 I) ng/dl range with the TSH suppressed in the normal
b. Sonography range. T4 and TSH levels should be checked according to
2. Function Studies
the following schedule:
a. 123 I uptake
b. Serum thyroglobulin 0-6 months: every 6 weeks
3. Suspected inborn error of T4 synthesis
6 months-3 years: every 3 months
a. 123I uptake and perchlorate discharge
4. Suspected autoimmune thyroid disease > 3 years: 6 monthly
a. Maternal and neonatal serum TBII measurement (not
T4 and TSH should also be checked 6-8 weeks after any
routinely available)
5. Suspected iodine exposure or deficiency dosage change.
a. Urinary iodine measurement
It is equally important to avoid over treatment.
6.Ancillary test to determine severity of fetal hypothyroidism
a. Radiograph of knee for skeletal maturation Adverse effects of over treatment include premature
fusion of cranial sutures, acceleration of skeletal
maturation and problems with temperament and
When should we ask for free T4 levels?
behavior.
In most situations, T4 (total) levels are sufficient for
Asymptomatic hyperthyrotropinemia: Elevated TSH with
diagnosis of hypothyroidism and monitoring treatment.
normal T4 values are seen commonly. This
Free T4 estimation is three times costlier and availability
hyperthyrotropinemia can be transient or permanent.
of the test is limited. There are certain specific situations
Perinatal iodine exposure is the commonest cause of
when estimation of free T4 should be done6, 10:
transient elevation of TSH. Other causes of
In premature newborns, T4 (total) values may be low hyperthyrotropinemia include defects in biological
because of abnormal protein binding or low levels of activity of TSH or TSH receptor, a mild thyroid hormone
thyroxine binding globulin (TBG) due to immaturity biosynthesis defect, subtle developmental defects or a
of liver function or undernutrition. Therefore, free T4 disturbance of the TSH feedback control system.
values provide a better estimate of true thyroid Hyperthyrotropinemia in newborns is usually treated but
function in premature or sick newborns. in the presence of free T4 levels in upper half of normal
range, expectant management can be followed. In case of
Free T4 should be asked for in case of finding a low
starting treatment, a 6 week trial of putting the child off
T4 with normal TSH. If free T4 is normal, it can be a
therapy followed by measuring TSH and T4 levels should
case of congenital partial (prevalence 1: 4000-12000
be done at 3 years of age.
newborns) or complete (prevalence 1: 15000

Indian Journal of Pediatrics, Volume 75April, 2008 365


59 37

V. Jain et al

Fig. 1. Approach to a newborn infant with positive screening test for CH

Preterm infants with low T4 and normal TSH levels can be continued by the hyperthyroid mothers during
(Transient hypothyroxinemia of prematurity): Use of breast feeding because concentration of these drugs is
levothyroxine in an attempt to normalize levels remains very low in breast milk. If we have been able to document
controversial because there is insufficient evidence that the presence of TBII in an infant and are attributing
early treatment with thyroid hormone leads to improved hypothyroidism to maternal autoimmune thyroiditis,
outcomes. Larger studies, especially in the extremely treatment should be started if T4 is low and continued for
preterm infants are needed to resolve this issue.12 3-6 months 6. However, when TBII estimation is not
available it is best to continue treatment till the age of 3
Transient Hypothyroidism: Infants with presumed
years and then give a trial off therapy for 6 weeks
transient hypothyroidism due to maternal goitrogenic
followed by retesting of T4 and TSH to determine the
drugs need not be treated unless low T4 and elevated
need for continuation of therapy.
TSH values persist beyond 2 weeks. Therapy can be
discontinued after 8-12 weeks. Intake of antithyroid drugs The management has been summarized in Panel 1.

366 Indian Journal of Pediatrics, Volume 75April, 2008


60 38

Congenital Hypothyroidism

PANEL 1. Management of Congenital Hypothyroidism et al. Neonatal screening for congenital hypothyroidism using
the filter paper thyroxine technique. Indian J Med Res 1994;
Abnormal values on screening (T4 < 6.5 ug/dL, TSH > 20mu/ 100: 36-42.
L) should always be confirmed by a venous sample using age 2. Fisher DA, Klein AH. Thyroid development and disorders of
appropriate cut-offs. thyroid function in the newborn. N Engl J Med 1981; 304 : 702
Investigations to determine the etiology such as scintigraphy 712.
should be done as soon as the diagnosis is made. If it is not 3. Adams LM, Emery JR, Clark SJ, Carlton EI, Nelson JC.
possible, the therapy should be started without delay. Reference ranges for newer thyroid function tests in
The initial dose of L-thyroxine should be 10-15g/ kg/ day premature infants. J Pediatr 1995; 126 : 122-127.
with the aim to normalize the T4 level at the earliest. T4 should 4. Nikta Forghani, Tandy Aye. Hypothyroxinemia and
be kept in the upper half of normal range (10-16 g/dL) with prematurity. Neoreviews 2008; 9 : e66-e71.
TSH level suppressed in the normal range. 5. American Academy of Pediatrics, Rose SR; Section on
Asymptomatic hyperthyrotropinemia should be treated unless Endocrinology and Committee on Genetics, American
the free T4 levels are in upper half of normal range. Thyroid Association, Brown RS; Public Health Committee,
When treatment has been started in an infant with suspected Lawson Wilkins Pediatric Endocrine Society, Foley T,
transient hypothyroidism or isolated increase in TSH or Kaplowitz PB, Kaye CI, Sundararajan S, Varma SK. Update of
borderline values of T4 and TSH, a 6 week trial of putting the newborn screening and therapy for congenital
child off therapy followed by measuring TSH and T4 levels hypothyroidism. Pediatrics 2006; 117 : 2290-303.
should be done at 3 years of age. 6. Fisher DA. Disorders of the thyroid in newborns and infants.
In Sperling MA, ed. Pediatric Endocrinology, 2 nd ed.
Philadelphia; Saunders, 2002; 161-186.
OUTCOME
7. Fisher DA. Disorders of the thyroid in childhood and
The best outcome occurs with L-thyroxine therapy started adolescence. In Sperling MA, ed. Pediatric Endocrinology, 2nd
ed. Philadelphia; Saunders, 2002; 187-210.
by 2 weeks of age at 9.5g/kg or more per day, compared
8. LaFranchi S. Congenital hypothyroidism: etiologies, diagnosis
with lower doses or later start of therapy. Residual defects and management. Thyroid 1999; 9 : 735-740.
can include impaired visuospatial processing and 9. Fisher DA. Management of congenital hypothyroidism. J Clin
selective memory and sensorimotor defects. More than Endocrinol Metab 1991; 72 : 525-528.
80% of infants given replacement therapy before three 10. Brown RS. The thyroid gland. In Brook CGD, Hindmarsh PC
eds. Clinical Pediatric Endocrinology, 4th ed. London; Blackwell
months of age have an IQ greater than 85 but show signs
Science, 2001; 288-320.
of minimal brain damage, including impairment of 11. LaFranchi SH, Austin J. How should we be treating children
arithmetic ability, speech, or fine motor coordination in with congenital hypothyroidism? J Pediatr Endocrinol Metab
later life.5 When treatment is started between 3-6 months, 2007; 20 : 559-578.
the mean IQ is 71 and when delayed to beyond 6 months, 12. Postnatal thyroid hormones for preterm infants with transient
the mean IQ drops to 54.13 hypothyroxinaemia. Cochrane Database Syst Rev 2007:
CD005945.
13. Klien AH, Meltzer S, Kenny FM. Improved prognosis in
REFERENCES congenital hypothyroidism treated before age three months. J
Pediatr 1972; 81 : 912-915.
1. Desai MP, Upadhye P, Colaco MP, Mehre M, Naik SP, Vaz FE

Indian Journal of Pediatrics, Volume 75April, 2008 367


61
62
63
64
65 39

Symposium on AIIMS Protocols in Neonatology - V

Protocol for Administering Continuous Positive Airway


Pressure in Neonates
M. Jeeva Sankar, Jhuma Sankar, Ramesh Agarwal, Vinod K. Paul and Ashok K. Deorari

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi,
India

ABSTRACT

Continuous positive airway pressure (CPAP) is a simple, inexpensive and gentle mode of respiratory support in preterm
very low birth weight (VLBW) infants. It helps by preventing the alveolar collapse and increasing the functional residual
capacity of the lungs. Since it results in less ventilator induced lung injury than mechanical ventilation, it should theoretically
reduce the incidence of chronic lung disease in VLBW infants. Various devices have been used for CPAP generation and
delivery. The relative merits and demerits of these devices and the guidelines for CPAP therapy in neonates are discussed
in this protocol. [Indian J Pediatr 2008; 75 (5) : 471-478] Email: ashokdeorari_56@hotmail.com

Key words: Continuous positive airway pressure; Devices, Fixation; Treatment; Protocol

Continuous positive airway pressure (CPAP), often successful use of nasal prongs to provide CPAP in these
thought to be the missing link between supplemental infants. 4 After the initial enthusiasm, it gradually fell
oxygen and mechanical ventilation, is gaining immense out of favor in 1980s because of the advent of newer
popularity in neonatal intensive care units. Being modes of ventilation (such as high frequency
technically simple, inexpensive and effective, it has ventilation) and the perceived complications of CPAP
become the primary mode of respiratory support in (such as air leak). However, reports of significantly
preterm very low birth weight (VLBW) infants. The lower incidence of chronic lung disease (CLD) from
evidence, clinical studies, and the controversies Columbia University unit that used more CPAP
regarding its use have been extensively reviewed and (Hudson prongs) as compared to other North American
are not discussed here. 1, 2 This protocol deals mainly Centers have led to a resurgence of interest in CPAP
with the practical aspects of CPAP administration in over the past 15 years.5
neonates.
CPAP: HOW DOES IT WORK?
DEFINITION AND BACKGROUND
CPAP predominantly helps by preventing collapse of
CPAP refers to the application of positive pressure to the the alveoli with marginal stability. This results in better
airway of a spontaneously breathing infant through out recruitment of alveoli thus increasing the functional
the respiratory cycle. residual capacity (FRC). The physiologic effects of CPAP
are represented in fig. 1.
The first clinical use of CPAP was reported by
Gregory et al in a landmark report in 1971. They Components of CPAP system
described the use of CPAP via endotracheal tube or a
The components of a CPAP system are:
head box in preterm infants with respiratory distress
syndrome (RDS).3 Shortly after this, Kattwinkel reported Gas source: To provide continuous supply of warm
humidified and blended gases i.e., air and oxygen
Pressure generator: To create the positive pressure
Correspondence and Reprint requests : Prof Ashok K Deorari, in the circuit
Professor, Department of Pediatrics, All India Institute of Medical Patient interface/delivery system: To connect the
Sciences, Ansari Nagar, New Delhi 110029, India. CPAP circuit to the infants airway.
[Received April 24, 2008; Accepted April 24, 2008]

Indian Journal of Pediatrics, Volume 75May, 2008 471


66 40

M.J. Sankar et al

CPAP

Fig. 1. Effects of CPAP


(FRC, functional residual capacity; V/Q, ventilation-perfusion ratio; PVR, pulmonary vascular resistance; PaCO 2 & PaO 2 ,
partial pressure of carbon-di-oxide and oxygen respectively in the arterial blood)

Devices used for pressure generation Bubble CPAP: A typical bubble CPAP setup is shown
in Panel 2. One has to remember that though classified
The pressure sources of CPAP can be broadly grouped
as a continuous flow device, flow may still need to be
into: 6
adjusted to maintain continuous bubbling in the water
Continuous flow devices
chamber and thus the required level of CPAP.
Variable flow devices (Fig. 2)

Fig. 2. Types of CPAP Devices

472 Indian Journal of Pediatrics, Volume 75May, 2008


67 41

Protocol for Administering Continuous Positive Airway Pressure in Neonates

Variable flow CPAP: A typical example is the Infant flow Devices used for CPAP delivery (Patient interface)
driver (IFD). It uses the Bernoulli Effect via dual injector
Various devices used for CPAP delivery include:
jets directed towards each nasal prong to maintain a
constant pressure. If the infant requires more Nasal prongs (single/double or binasal)
inspiratory flow, the Venturi action of the injector jets
Long (or) nasopharyngeal prongs
entrains additional flow. When the infant makes a
spontaneous expiratory effort, there is a fluidic flip Nasal cannulae (Fig. 3)
that causes the flow to flip around and to leave the
generator chamber via the expiratory limb (Coanda Nasal masks
effect). So, unlike in the other methods of CPAP where Face mask, endotracheal, and head box are no longer
the infant has to exhale against the incoming gas flow, used for delivering CPAP in neonates.
the fluidic flip of the variable flow devices assist his
exhalation thus reducing the work of breathing. Endotracheal CPAP is not recommended because it
has been found to increase the work of breathing (infant
We use continuous flow CPAP by both conventional has to breathe through a straw).
ventilators and bubble CPAP device in our unit.
The advantages and disadvantages of each of these
The advantages and disadvantages of each of these methods have been summarized in table 2.
methods are given in table 1.
TABLE 1. A Comparison of CPAP Devices Used for Pressure Generation7

Device Examples Approximate Advantages Disadvantages Remarks


Cost (INR)

Conventional Bear Cub, Bird- 5 10 lakhs No need of Expensive Of practical utility in


ventilator VIP, Draeger separate Standard flow of units having ventilators
derived CPAP Baby log, equipment 5-8L/min may be but not so in a small
Newport, Can be easily insufficient in the hospital/nursing home
Sechrist, Siemens, switched over presence of high without a neonatal
SLE, etc. to mechanical leak ventilator.
ventilation, if Difficult to know
CPAP fails if the set flow is
sufficient or not
(insufficient flow
can lead to increased
WOB)

Stand-alone Lectromedik, 25,000 to Economical Most of them do Though inexpensive,


CPAP machines Meditrin, 80,000 Useful for small not have proper they have not been
(Indigenous Phoenix, hospitals blenders and/or tested adequately;
CPAP) Shreeyash, Zeal Can have bubble pressure niggling issues observed
CPAP option manometer during daily use

Bubble CPAP Indian: 50,000 to 80,000 Simple and Flow has to be It seems unlikely that
Mediserve, inexpensive altered to ensure oscillations delivered at
Meditrin Oscillations proper bubbling the nares are
Imported: 1,60,000 produced by It is difficult to transmitted up to the
Fisher & Paykel continuous detect high flow alveoli;
bubbling might which can lead Still, the stand-alone
contribute to gas to over distension option makes it an easy
exchange (akin to of the lungs and cost effective
HFV) proposition in
Can identify large developing countries
leaks at the nares
(bubbling stops)

Variable flow Arabella, IFD, 3 lakhs Maintains more Expensive On theoretical grounds,
devices Viasys SiPAP uniform pressure Requires more this device scores more
Might decrease the technical expertise than the other two;
WOB However the prohibitive
Recruits lung cost and the lack of
volume more evidence regarding its
effectively superiority preclude its
widespread use

(WOB, work of breathing; HFV, high frequency ventilation; IFD, infant flow driver)

Indian Journal of Pediatrics, Volume 75May, 2008 473


68 42

M.J. Sankar et al

listed in Panel 1.
Practically, CPAP is very useful in preterm (<35 weeks)
infants with respiratory distress/failure of any etiology.
Some of these indications have been briefly described
below:
RDS: The most common indication for CPAP is mild
to moderate RDS. It helps in this condition by
preventing collapse of alveoli with marginal stability.
The recruitment of more alveoli helps to increase the
FRC thus helping in better oxygenation (Fig. 1).
Numerous studies have proved its efficacy in
reducing the need for mechanical ventilation and
probably the incidence of chronic lung disease in
infants with RDS.10, 11
CPAP and surfactant: The beneficial effect of CPAP in
preterm infants (<29 to 30 weeks) could probably be
enhanced by administering surfactant. In this
approach, if respiratory distress progresses even
Fig. 3. CPAP Delivery Systems after initiating CPAP, the baby is intubated, given

T ABLE 2. Advantages and Disadvantages of Common CPAP delivery Systems

Delivery system Advantages Disadvantages Remarks

Nasal prongs Simple device Relatively difficult to fix Studies have shown that they
(single/binasal) Lower resistance leads to Risk of trauma to nasal are more effective than
Example: greater transmission septum and turbinates nasopharyngeal prongs (in
Argyle of pressure Leak through mouth means post-extubation setting) 8
Hudson Mouth leak acts like a end expiration pressure
IFD prongs pop-off mechanism is variable

Nasopharyngeal prongs Easy availability More easily blocked Though more economical and
(e.g., using a cut Economical by secretions easily available, they are found
endotracheal tube) More secure fixation Likely to get kinked to be inferior to short binasal
Length is estimated by prongs8
measuring the distance from
the earlobe to the tip of the
chin or nose;
Tube placement is confirmed
by direct visualization of the
tip behind the uvula

Nasal cannulae
(with an outer diameter of Ease of application Unreliable pressure delivery Mainly tried in apnea of
3mm and flows up to May need high flows to prematurity paucity of
2 L/min) generate pressure data in other conditions 9
if only oxygen is used Still experimental
(without air blender)
FiO2 delivered may be high
Large leaks around the
cannulae

Nasal masks Minimal nasal trauma Difficulty in obtaining an New generation masks are yet
adequate seal to be studied in detail

(IFD, Infant flow driver)

In our unit, we use short bi-nasal prongs for delivering surfactant, and then extubated and put back on
CPAP (both ventilator and bubble CPAP). CPAP again. Known as INSURE (Intubation-
Surfactant-Extubation), this approach might further
Indications for CPAP reduce the need for subsequent ventilation and
The common clinical indications of CPAP have been improve the outcome in extreme preterm infants.12

474 Indian Journal of Pediatrics, Volume 75May, 2008


69 43

Protocol for Administering Continuous Positive Airway Pressure in Neonates

Panel 1. Indications for CPAP CONTRAINDICATIONS OF CPAP


Common indications
Respiratory distress syndrome (RDS) The important contraindications for CPAP include:
Apnea of prematurity (especially obstructive apnea) Progressive respiratory failure with PaCO2 levels
Post-extubation in preterm VLBW infants >60 mmHg and/or inability to maintain
Transient tachypnea of newborn (TTNB)/delayed
oxygenation (PaO2 <50 mmHg)
adaptation Certain congenital malformations of the airway
Other indications (choanal atresia, cleft palate, tracheo esophageal
Pneumonia fistula, congenital diaphragmatic hernia, etc.)
Meconium aspiration/ other aspiration syndromes Severe cardiovascular instability (hypotension)
Pulmonary edema/pulmonary hemorrhage Poor respiratory drive (frequent apnea and
Laryngomalacia/ tracheomalacia/ bronchomalacia bradycardia) that is not improved by CPAP.

GUIDELINES FOR INITIATION OF CPAP THERAPY


However, clinical trials have not shown any
reduction in the incidence of CLD so far. More
When to initiate CPAP?
studies are needed to confirm or refute its possible
beneficial effects. We do not routinely employ INSURE The timing of initiation of CPAP in preterm infants with
technique at present. respiratory distress needs further elaboration.
Apnea of prematurity: The mechanism by which Early CPAP: It is important to note that CPAP helps
CPAP helps in apnea of prematurity has been mainly by preventing the alveolar collapse in infants
explained before (Fig. 1). It is typically used when with surfactant deficiency. Once atelectasis and
clinically significant episodes persist despite optimal collapse have occurred, CPAP might not help much.
methylxanthine therapy. Therefore, all preterm infants (<35 weeks) with any
Post-extubation in VLBW infants: CPAP reduces the sign of respiratory distress (tachypnea/chest in
incidence of apnea, respiratory acidosis, and drawing/grunting) should be started immediately on
increased oxygen requirement in VLBW infants CPAP.
extubated after a brief period of mechanical Prophylactic CPAP: Extending this logic, some have
ventilation. advocated use of prophylactic CPAP (before the onset
Delayed adaptation/TTNB: In these conditions of respiratory distress) in preterm VLBW infants as
associated with excess lung fluid, CPAP helps by majority of them would eventually develop respiratory
maintaining the lung expansion. Though useful in distress. However, there is no evidence for any
premature infants, term and near-term neonates with additional benefit with this approach; indeed, there are
TTNB often do not tolerate this mode of respiratory concerns regarding increased adverse effects such as
support. intraventricular hemorrhage. Hence, prophylactic
CPAP is NOT recommended at present.13
Pneumonia: CPAP can be tried in stable infants with
mild to moderate respiratory distress due to How to set up a CPAP apparatus?
pneumonia. It helps in this condition by maintaining
The steps in setting-up a bubble CPAP are summarized
the lung expansion preventing any collapse due to
in Panel 2.
fluids and secretions.
How to fix the CPAP delivery system (nasal cannula)
Meconium aspiration syndrome: Use of CPAP is a
contentious issue in this condition as most of the The most difficult aspect of using nasal CPAP is the
infants would already have hyperexpanded lung positioning and fixation of the patient interface. The
fields and CPAP might further aggravate it. optimal technique of fixation depends on the type of
Moreover, these term infants are unlikely to tolerate delivery system used; the exact technique used does not
CPAP well. It is only indicated in a rare infant with matter as long as the device is secure and not
predominant collapse/atelectasis (preferably proven traumatizing.
by chest X-ray).
Short binasal prongs: It is important to choose the
We use CPAP predominantly in preterm infants (<35 appropriate sized prong that snugly fits in the nasal
weeks and birth weight <1800g) with respiratory distress, cavity to avoid a significant leak. However, to avoid
apnea of prematurity, delayed adaptation, and pneumonia; causing any injury, it should be fixed straight and not
also we extubate VLBW infants to CPAP routinely. We pressed hard against the nasal septum.
occasionally use CPAP in near-term and term infants with
We use a modified cap (made from adult cotton socks) and
transient tachypnea and pneumonia.

Indian Journal of Pediatrics, Volume 75May, 2008 475


70 44

M.J. Sankar et al

Panel 2. Steps of Initiation and Nursing Care during CPAP7

A. How to set-up a bubble CPAP?


1. Connect the air and oxygen tubing (pressurized gases from either central manifold or from compressor and oxygen cylinder respectively)
2. Attach both to the air-oxygen blender
3. Set the flow using flow meter (usually at 5-8 L/min)
4. Set up the inspiratory limb:
a. From the flow meter to the humidifier and
b. From the humidifier to the patient end (e.g., nasal cannula); fill water in the humidifier and humidify the gases to 34-370C.
5. Set up the expiratory limb - from the patient end to a chamber filled with sterile water. Immerse it under the water up to the required depth
(which is determined by the intended pressure - e.g., to deliver 5 cm H20, immerse up to 5 cm mark in the tube).
6. Attach a pressure manometer at the patient end

7. Set required pressure and FiO2, low pressure alarm and apnea alarm
8. Occlude the patient end of the ventilator circuit with your palm and observe if:
a. Bubbling occurs in the water chamber - If there are no bubbles, look for any leak in the circuit; if no leak is found, increase the
flow by 1 L/min and recheck.
b. The set pressure is delivered (see the manometer reading) - If it is less than the set pressure, look for any leaks in the circuit/
around the cannula. If no leak is found, increase the flow and recheck.

B. Initiation of CPAP
1. Place a roll under infants shoulder to slightly extend the neck
2. Application of prongs:
a. Choose the correct size prong (the prongs should fill the nasal opening without stretching the skin)
b. Apply a thin strip of Tegaderm on overlying skin of septum
c. Place the prongs with the curve downwards and fix as shown in Fig. 4.
3. Attach the patient end of the ventilator circuit to the cannula
4. Attach a pulse-oximeter to the infant

C. Nursing Care
1. Monitor the infant frequently (see text); observe if the baby is comfortable
2. Pass an orogastric tube. Keep the proximal end of tube open. If the infant is being fed while on CPAP, close the tube for half an hour after
giving feeds and keep it open for the next 90 minutes (if fed 2 hourly).
3. Do regular but gentle nasal suction to clear the mucus 4 hourly or as and when required.
4. Clean the nasal cannula and check its patency once per shift.
5. Change the infants position regularly every 2-4 hours and check the skin condition frequently for redness and sores.

476 Indian Journal of Pediatrics, Volume 75May, 2008


71 45

Protocol for Administering Continuous Positive Airway Pressure in Neonates

TABLE 3. Protocol for CPAP Therapy in Three Common Neonatal Conditions

Indications
RDS Apnea of prematurity Post extubation

How to initiate ?
Pressure Start at 5cm H 2 O Start at 4cm H 2 O Start at 4-5cm H 2O
FiO 2 0.5 (titrate based on SpO2 ) 0.21-0.4 (as decided by SpO 2 ) 0.05 to 0.1 above the
pre-extubation FiO2
What to do if there
is no improvement?
Pressure Increase in steps of 1-2cm H 2O to Increase up to 5cm H 2 O Increased in steps of 1-2cm
reach a maximum of 7-8 cm H2 O (further increase is not H 2 O to reach a maximum of
warranted usually in this 7-8cm H 2 O
condition - may lead to
hyperinflation)
FiO 2 Increase in steps of 0.05 (if FiO2 increase does not Increase in steps of 0.05 to a
oxygenation is still compromised) help much maximum of 0.8
up to a maximum of 0.8
Failure of CPAP Worsening respiratory distress (as Recurrent episodes of apnea
indicated by Silverman scoring) and/ requiring PPV
or hypoxemia (PaO 2 <50mmHg) /
hypercarbia (PaCO 2 >60mmHg)
despite CPAP pressure of 7-8 cm
H2 O and FiO 2 of 0.8
(Likely to occur in infants with severe (Likely to occur in infants (Likely to occur in ELBW infants
RDS, associated sepsis, and in ELBW with central apneas sepsis ) and in sepsis / pneumonia, PDA,
infants who have not received ANS) metabolic acidosis, and collapse)
Same as for RDS
Weaning from CPAP
When to wean When there is no respiratory distress No episodes of apnea/ Same as for RDS
and SpO 2 / blood gases are normal desaturation / bradycardia
for atleast 12-24 has hrs
How to wean Reduce FiO 2 in steps of 0.05 to 0.4, Same as for RDS
then decrease pressure in steps of
1-2cm H2O until 3-4 cm H 2 O
(infants clinical condition will guide
the speed of weaning)

tapes to secure the binasal prongs (Fig. 4). d. Abdominal girth


Steps of initiation and nursing care before and during Hazards/Complications of CPAP
CPAP
The steps of initiation and nursing care during CPAP CPAP though less invasive and generally safer than
therapy are given in Panel 2. IMV, is not free of side effects.
Pulmonary air leaks are probably the most
PROTOCOL FOR CPAP THERAPY important clinically significant adverse effect of
CPAP. 14 It occurs following over distension of the
lungs caused by inappropriately high pressures.
Protocol for CPAP therapy in the three most common They tend to occur when the lung compliance starts
clinical indications is given in table 3. improving and the oxygen requirements also show a
Monitoring while on CPAP reduction. One has to note that the two recent trials
on CPAP for RDS have shown either a trend or a
The following parameters need to be monitored while definite increase in the incidence of pneumothorax.15,
the infant is on CPAP: 16
Therefore, extra vigilance is required during CPAP
Continuous monitoring of respiratory rate, heart rate, therapy.
SpO2 Decreased cardiac output due to reduction in the
Serial monitoring of venous return, decreased right ventricular stroke
a. Severity of respiratory distress by using Downes or volume, and altered dispensability of left ventricle.17
Silverman score This effect can be minimized by using optimal CPAP
b. Arterial blood gases (ABGs) and achieving adequate intravascular volume.
c. Perfusion - CFT, BP, peripheral pulses, urine output Impedance of pulmonary blood flow with increased

Indian Journal of Pediatrics, Volume 75May, 2008 477


72 46

M.J. Sankar et al

REFERENCES

1. Sankar MJ, Deorari AK. CPAP A gentler mode of


ventilation. J Neonatol 2007; 21 : 160-165.
2. Upadhyay A, Deorari AK. Continuous positive airway
pressure - a gentler approach to ventilation. Indian Pediatr
2004; 41 : 459-469.
3. Gregory GA, Kitterman JA, Phibbs RH et al. Treatment of
idiopathic respiratory distress syndrome with continuous
positive airway pressure. N Engl J Med 1971; 284 : 333-340.
4. Kattwinkel J, Nearman HS, Fanaroff AA, Katona PG, Klaus
MH. Apnea of prematurity. Comparative therapeutic
effects of cutaneous stimulation and nasal continuous
positive airway pressure. J Pediatr 1975; 86 : 588-592.
5. Avery ME, Tooley WH, Keller JP et al. Is chronic lung
disease in low birth weight infants preventable? A survey of
eight centers. Pediatrics 1987; 79 : 26-30.
6. Courtney SE, Barrington KJ.Continuous positive airway
pressure and noninvasive ventilation. Clin Perinatol 2007; 34:
73-92.
7. Anonymous. Continuous Positive Airway Pressure Machines.
In Deorari AK, Paul VK, eds. Neonatal Equipment. 3rd ed.
New Delhi, Sagar Publications; 2006, 129-137.
8. De Paoli AG, Davis PG, Faber B, Morley CJ. Devices and
pressure sources for administration of nasal continuous
positive airway pressure (NCPAP) in preterm neonates.
Cochrane Database of Syst Rev 2002: CD002977.
9. Sreenan C, Lemke RP, Hudson-Mason A et al. High-flow
nasal cannulae in the management of apnea of prematurity:
a comparison with conventional nasal continuous positive
Fig. 4. Steps in Fixation of CPAP Nasal Cannula airway pressure. Pediatrics 2001; 107 : 1081-1083.
10. Gittermann MK, Fusch C, Gittermann AR, Regazzoni BM,
Moessinger AC. Early nasal continuous positive airway
pulmonary vascular resistance (with inappro- pressure treatment reduces need for intubation in very low
birth infants. Eur J Pediatr 1997; 156: 384-388.
priately high CPAP pressure)
11. Poets CF, Sens B. Changes in intubation rates and outcome
Gastric distension and CPAP belly syndrome. of VLBW -a population based study. Pediatrics 1996; 98:
24-27.
These complications are rarely seen nowadays. The
12. Verder H, Robertson B, Greisen G, Ebbesen F, Albertsen P,
risk is further minimized by routine use of orogastric Lundstron JT. Surfactant therapy and nasal continuous
tube. positive airway pressure for newborns with respiratory
distress syndrome. Danish-Swedish Multicentre Study
Nasal irritation, damage to the septal mucosa, or Group. N Engl J Med 1994; 331: 1051-1055.
skin damage and necrosis from the fixing devices. 13. Subramaniam P, Henderson-Smart DJ, Davis PG.
Prophylactic nasal continuous positive airways pressure for
preventing morbidity and mortality in very preterm infants.
Cochrane Database of SystRev 2005: CD001243.
CONCLUSION 14. Hall RT, Rhodes PG. Pneumothorax and pneumome-
diastinum in infants with idiopathic respiratory distress
syndrome receiving CPAP. Pediatrics 1975; 55: 493.
CPAP has been well established as the first line therapy 15. Buckmaster AG, Arnolda G, Wright IM, Foster JP,
in the management of respiratory distress in preterm Henderson-Smart DJ. Continuous positive airway pressure
VLBW infants. It helps by preventing alveolar collapse, therapy for infants with respiratory distress in non tertiary
maintaining airway stability and stabilizing the chest care centers: a randomized, controlled trial. Pediatrics 2007;
wall. Various devices, both for pressure generation and 120 : 509-518.
16. Morley CJ, Davis PG, Doyle LW, Brion LP, Hascoet JM,
for delivery of CPAP, are available for use in neonates. Carlin JB; COIN Trial Investigators.Nasal CPAP or
The advantages and disadvantages of each device, intubation at birth for very preterm infants. N Engl J Med
method of fixation of short binasal prongs, and a 2008; 358 : 700-708.
protocol for initiation of CPAP have been discussed in 17. Tittley JG, Fremes SE, Weisel RD, Christakis GT, Evans PJ,
this protocol. Madonik MM et al. Hemo-dynamic and myocardial
metabolic conse-quences of PEEP. Chest 1985; 88: 496-502.

478 Indian Journal of Pediatrics, Volume 75May, 2008


73
74
75
76
77 47

Symposium on AIIMS Protocols in Neonatology V

Feeding of Low Birth Weight Infants


M. Jeeva Sankar, Ramesh Agarwal, Satish Mishra, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar,
New Delhi, India

ABSTRACT
Optimal feeding of low birth weight (LBW) infants improves their immediate survival and subsequent growth and development.
Being a heterogeneous group comprising term and preterm neonates, their feeding abilities, fluid and nutritional requirements
are quite different from normal birth weight infants. A practical approach to feeding a LBW infant including choice of initial
feeding method, progression of oral feeds, and nutritional supplementation based on her oral feeding skills and nutritional
requirements is being discussed in this protocol. Growth monitoring, management of feed intolerance, and the essential skills
involved in feeding them have also been described in detail. [Indian J Pediatr 2008; 75 (5) : 459-469] Email: vinodpaul@
neonatalhealth.com

Key words : Low birth weight; Feeding; Expression of breast milk; Fortification; Growth monitoring

Globally, about 18 million infants are born with a birth contrast, feeding of LBW infants is relatively difficult
weight of <2500g every year.1 Though these low birth because of the following limitations:
weight (LBW) infants constitute only about 14% of the
Though majority of them are born at term, a
total live births, they account for 60-80% of total neonatal
significant proportion are born premature with
deaths.2 Most of these deaths can be prevented with extra
inadequate feeding skills. They might not be able to
attention to warmth, prevention of infections and more
breastfeed and would require other methods of
importantly, optimal feeding.
feeding such as spoon or gastric tube feeding.
These infants are prone to have significant illnesses in
WHY IS IT IMPORTANT?
the first few weeks of life; the underlying condition
often precludes enteral feeding.
Nutritional management influences immediate survival
Preterm very low birth infants (VLBW) infants have
as well as subsequent growth and development of LBW
higher fluid requirements in the first few days of life
infants. Even simple interventions such as early initiation
due to excessive insensible water loss.
of breastfeeding and avoidance of pre-lacteal feeding have
been shown to improve their survival in resource Since intrauterine accretion of nutrients occurs mainly
restricted settings.3 Early nutrition could also influence the in the later part of the third trimester, VLBW infants
long term neurodevelopmental outcomes; malnutrition at (usually born before 32 weeks gestation) have low
a vulnerable period of brain development has been shown body stores at birth. Hence, they require
to have deleterious effects in experimental animals.4 supplementation of various nutrients. Even term LBW
infants who are likely to be growth restricted need
higher calories for catch-up growth.
HOW IS IT DIFFERENT?
Because of the gut immaturity, they are more likely to
experience feed intolerance necessitating adequate
Term infants with normal birth weight require minimal monitoring and treatment.
assistance for feeding in the immediate postnatal period -
they are able to feed directly from mothers breast. In PROTOCOL FOR FEEDING LBW INFANTS

In this protocol, we address the following issues:


Correspondence and Reprint requests : Prof Vinod K. Paul,
Professor & Head, Department of Pediatrics, All India Institute of How to decide the initial method of feeding in a given
Medical Sciences, Ansari Nagar, New Delhi-110029, India LBW infant?
[Received April 23, 2008; Accepted April 23, 2008]

Indian Journal of Pediatrics, Volume 75May, 2008 459


78 48

M.J. Sankar et al

For infants initiated on modes other than birth in healthy LBW infants with the appropriate feeding
breastfeeding: method determined by their gestation and oral feeding
a. How to progress to breastfeeding? skills.
b. What milk to be given?
Maturation of oral feeding skills: Breastfeeding requires
c. How much milk to be given?
effective sucking, swallowing and a proper coordination
What supplements are required? between suck/swallow and breathing. These complex
skills mature with increasing gestation (Table 1).
How to assess the feeding adequacy and monitor the
growth? The fetus is able to swallow amniotic fluid by as early
as 11 to 12 weeks gestation. Mouthing can be observed at
How to identify and manage feed intolerance?
15 weeks but the coordinated sucking movements are not
usually present until about 28 weeks gestation. Single
DECIDING THE INITIAL METHOD OF FEEDING sucks can be recorded manometrically at 28 weeks and
sucking bursts by 31 weeks gestation. A mature sucking
pattern that can adequately express milk from the breast
It is essential to categorize LBW infants into two major is not present until 32-34 weeks gestation.5 However, the
groups sick and healthy - before deciding the method of coordination between suck/swallow and breathing is not
feeding. fully achieved until 37 weeks of gestation.
Sick infants The maturation of oral feeding skills and the choice of
This group constitutes infants with significant respiratory initial feeding method at different gestational ages are
distress requiring assisted ventilation, shock requiring summarized in table 1.
inotropic support, seizures, symptomatic hypoglycemia/ THE INITIAL FEEDING METHOD
hypocalcemia, electrolyte abnormalities, renal/cardiac
failure, surgical conditions of gastrointestinal tract, Traditionally, the initial feeding method in a LBW infant
necrotizing enterocolitis (NEC), hydrops, etc. These is decided based on her birth weight. This is not an ideal
infants are usually started on intravenous (IV) fluids. way because the feeding ability depends largely on
Enteral feeds should be initiated as soon as they are gestation rather than the birth weight.
hemodynamically stable with the choice of feeding However, it is important to remember that not all infants
method based on the infants gestation and clinical born at a particular gestation would have same feeding
condition (see below). skills. Hence the ideal way in a given infant would be to
It is important to realize that enteral feeding is evaluate if the feeding skills expected for his/her
important even in sick neonates. Oral feeds should not be gestation are present and then decide accordingly (Fig. 1).
delayed in them without any valid reason. Even infants All stable LBW infants, irrespective of their initial
with respiratory distress and/or on assisted ventilation feeding method should be put on their mothers breast.
can be started on enteral feeds once the initial acute phase The immature sucking observed in preterm infants born
is over and the infants color, saturation and perfusion before 34 weeks might not meet their daily fluid and
have improved. Similarly, sepsis (unless associated with nutritional requirements but helps in rapid maturation of
shock/sclerema/NEC) is not a contraindication for their feeding skills and also improves the milk secretion in
enteral feeding. their mothers (Non-nutritive sucking).
Healthy LBW infants Spoon/paladai feeding
Enteral feeding should be initiated immediately after In our unit, we use paladai feeding in LBW infants who
TABLE 1. Maturation of Oral Feeding Skills and the Choice of Initial Feeding Method in LBW Infants5

Gestational age Maturation of feeding skills Initial feeding method

< 28 weeks No proper sucking efforts Intravenous fluids


No propulsive motility in the gut
28-31 weeks Sucking bursts develop
No coordination between suck/swallow Oro-gastric (or naso-gastric) tube feeding with occasional spoon/
and breathing paladai feeding
32-34 weeks Slightly mature sucking pattern
Coordination between breathing and Feeding by spoon/paladai/cup
swallowing begins
>34 weeks Mature sucking pattern Breastfeeding
More coordination between breathing and
swallowing

460 Indian Journal of Pediatrics, Volume 75May, 2008


79 49

Feeding of Low Birth Weight Infants

are not able to feed directly from the breast. The steps of Panel 1. Steps of Paladai Feeding6
paladai feeding are described in Panel 1.6
Place the infant in up-right posture on mothers lap
Intra-gastric tube feeding
Keep a cotton napkin around the neck to mop the spillage.
The steps of intra-gastric tube feeding are given in Panel
Take the required amount of expressed breast milk by using
2. Some of the controversial issues in gastric tube feeding a clean syringe
are discussed below:
Fill the paladai with milk little short of the brim;
Naso-gastric vs. oro-gastric feeding: Physiological studies
Hold the paladai from the sides; DO NOT put your finger
have shown that naso-gastric (NG) tube increases the
airway impedance and the work of breathing in very Place it at the lips of the baby in the corner of the mouth
preterm infants.7 Hence, oro-gastric tube feeding might be Tip the paladai to pour a small amount of milk into the
preferable in these infants. We employ only oro-gastric tube infants mouth
feeding in our unit. Feed the infant slowly; he/she will actively swallow the
Intermittent bolus vs. continuous intra-gastric feeding: There milk
are no differences in the time to reach full enteral feeding/ Repeat the process until the required amount has been fed
somatic growth/incidence of NEC between infants fed by If the infant does not actively accept and swallow, try to
intermittent bolus or continuous intra-gastric feeding.8 arouse him/her with gentle stimulation
Studies have shown that gastric emptying and duodenal
While estimating the milk intake, deduct the amount of milk
motor responses are enhanced in infants given continuous
left in the cup and the amount of estimated spillage
intra-gastric feeding.9 But a major disadvantage of this
method is that the lipids in the milk tend to separate and Wash the paladai with soap and water and then put in boiling
stick to the syringe and tubes during continuous infusion water for 20 minutes to sterilize before next feed

Fig. 1. Deciding the Initial Feeding Method in LBW infants

Indian Journal of Pediatrics, Volume 75May, 2008 461


80 50

M.J. Sankar et al

resulting in significant loss of energy and fat content. We


Panel 2. Steps of Intra-gastric Tube Feeding6
use intermittent bolus feeding in our unit.
Before starting a feed, check the position of the tube
Remove the plunger the syringe (ideally a sterile syringe
Progression of Oral Feeds
should be used) All LBW infants, irrespective of their gestation and birth
Connect the barrel of the syringe to the end of the gastric tube weight, should ultimately be able to feed directly from the
Pinch the tube and fill the barrel of the syringe with the mothers breast. For preterm LBW infants started on IV
required volume of milk fluids/OG tube/paladai feeding, the steps of progression
Hold the tube with one hand, release the pinch and elevate the to direct and exclusive breastfeeding are summarized in
syringe barrel Fig. 2.
Let the milk run from the syringe through the gastric tube by
gravity; DO NOT force milk through the gastric tube by using Term LBW infants started on IV fluids (because of their
the plunger of the syringe sickness) can be put on the breast once they are
Control the flow by altering the height of the syringe. hemodynamically stable.
Lowering the syringe slows the milk flow, raising the syringe
makes the milk flow faster Special situations
It should take about 10-15 minutes for the milk to flow into the Extremely low birth weight infants: They are usually
infants stomach
started on parenteral nutrition from day 1. Enteral feeds
Observe the infant during the entire gastric tube feed. Do not in the form of trophic feeding or minimal enteral nutrition
leave the infant unattended. Stop the tube feed if the infant
(MEN) are initiated once the infant is hemodynamically
shows any of the following signs: breathing difficulty, change
in colour/ looks blue, becomes floppy, and vomiting stable. Further advancement is based on the infants
Cap the end of the gastric tube between feeds; if the infant is on
ability to tolerate the feeds (See AIIMS protocol on
CPAP, the tube is preferably left open after about half an hour Minimal enteral nutrition).10
Avoid flushing the tube with water or saline after giving feeds. Severe IUGR with antenatally detected Doppler flow
abnormalities: Fetuses with abnormal Doppler flow such

Fig. 2. Progression of Oral Feeding in Preterm LBW Infants

462 Indian Journal of Pediatrics, Volume 75May, 2008


81 51

Feeding of Low Birth Weight Infants

as absent/reversed end diastolic flow (A/REDF) in the


Panel 3. Steps of expression of breast milk6
umbilical artery are likely to have had mesenteric
ischemia in utero. After birth, they have a significant risk The mother should wash her hands thoroughly.
of developing feed intolerance and NEC.11 The timing of She should hold a clean wide mouthed container near her
initiation of oral feeds in these infants is controversial. We breast.
usually delay feeding up to 48-72 hours in preterm (<35
Ask her to gently massage the breast for 5-10 minutes
weeks) infants with A/REDF. before expressing the milk (using the pulp of two fingers
Infants on CPAP/ventilation: These infants can be started or with knuckles of the fist in a circular motion towards
the nipple as if kneading dough). Massage should not
on OG tube feeds once they are hemodynamically stable.
hurt her.
But it is important to leave the tube open intermittently to
reduce gastric distension. Ask her to put her thumb ABOVE the nipple and areola,
and her first finger BELOW the areola opposite the thumb.
She should support the breast with her other fingers.
CHOICE OF MILK FOR LBW INFANTS Ask her to press her thumb and first finger slightly inward
towards the chest wall.
All LBW infants, irrespective of their initial feeding She should press her breast behind the nipple and areola
method should receive ONLY breast milk. This can be between her fingers and thumb. She must press on the
ensured even in those infants who are fed by paladai or lactiferous sinuses beneath the areola.
gastric tube by giving expressed breast milk (mothers Press and release, press and release. This should not hurt-
own milk or human donor milk). if it hurts, the technique is wrong. It may take some time
before milk starts coming.
Expressed breast milk (EBM): All preterm infants
mothers should be counseled and supported in expressing Ask her to press the areola in the same way from the
their own milk for feeding their infants. Expression SIDES, to make sure that milk is expressed from all
segments of the breast.
should ideally be initiated within hours of delivery so that
the infant gets the benefits of feeding colostrum. She should express one breast first till the milk flow slows;
Thereafter, it should be done 2-3 hourly - this would then express the other side; and then repeat both sides.
ensure that the infant is exclusively breastfed and also Avoid rubbing or sliding her fingers along the skin.
helps in maintaining the lactation in the mother.
Avoid squeezing the nipple itself. Pressing or pulling the
The steps of breast milk expression are given in Panel 3. nipple cannot express the milk.
We counsel mothers for expression of breast milk soon
after delivery by demonstration and by using poster
(Annexure 1) & videos (available on our website: HOW MUCH MILK IS TO BE GIVEN?
www.newbornwhocc. org)
Expressed breast milk can be stored for about 6 hours at It is essential to calculate the fluid requirements and feed
room temperature and for 24 hours in refrigerator. volumes for infants on paladai/gastric tube feeding.

Donor human milk: In centers where optimal milk Fluid requirement: The daily fluid requirement is
banking facilities are available, donor human milk can be determined based on the estimated insensible water loss,
used for feeding a LBW infant. At present, only a few other losses, and urine output. Extreme preterm infants
centers in India have standardized human milk banking need more fluids in the initial weeks of life because of the
facilities. Hence, it is not a practical option in our country. high insensible water loss.

Special situations We usually start fluids at 80 mL and 60 mL/kg/day for


infants birth weights of <1500g and 1500-2500g
Sick mothers/ contraindication to breastfeeding: In these rare respectively. Further requirements are calculated by daily
circumstances, the options available are estimation of weight loss/gain, serum sodium, urine
Formula feeds: output and specific gravity. The usual daily increment
a. Preterm formula in VLBW infants and would be about 15-20 mL/kg/day so that by the end of
b. Term formula in infants weighing >1500g at first week 150 mL/kg/day is reached in both the
birth categories. We usually reach a maximum of 180mL/kg/
day by day 14 (Refer to AIIMS protocol on Fluids and
Animal milk: e.g., undiluted cows milk electrolyte management in term and preterm neonates).12
Once the mothers condition becomes stable (or the Feed volume: After estimating the fluid requirements, the
contraindication to breastfeeding no longer exists), these individual feed volume to be given by OG tube or paladai
infants should be started on exclusive breastfeeding. (2-hrly/3-hrly) should be determined.

Indian Journal of Pediatrics, Volume 75May, 2008 463


82 52

M.J. Sankar et al

multi-nutrient supplementation or fortification of breast


milk (cf. VLBW infants). However, vitamin D and iron
might still have to be supplemented in them. While iron
supplementation is mandatory for all infants, vitamin D is
contentious because of the paucity of the data regarding
its levels and deficiency status in different populations.
Some would argue that the daily requirement of vitamin
D is met usually by de novo synthesis in the skin
(following exposure to sun light) and hence no
supplementation is required. However, because LBW
infants are more at risk of orteopenia than healthy term
infants, most neonatal units tend to supplement vitamin
D in the dose of 200-400 IU/day.13, 14
One has to assess the mothers nutritional status, their
exposure to sun, and the infants exposure to sun before
adopting a policy for their respective unit(s).
We supplement both vitamin D and iron in infants with
birth weights of 1500-2499 grams; vitamin D (200 IU) is
started at 2 weeks and iron (2 mg/kg/day) at 2 months of
life; both are continued till 1 year of age (Table 2).
VLBW infants
These infants who are usually born before 32-34 weeks
gestation have inadequate body stores of most of the
nutrients. Expressed breast milk has inadequate amounts
of protein, energy, calcium, phosphorus, trace elements
(iron, zinc) and vitamins (D, E & K) that are unable to
meet their daily recommended intakes (Table 3). Hence,
these infants need multi-nutrient supplementation till
they reach term gestation (40 weeks postmenstrual age).
After this period, their requirements are similar to those
infants with birth weights of 1500-2499 grams.
Multi-nutrient supplementation can be ensured by one of
the following methods:

Annexure 1. Poster on the procedure of Expression of Breast Milk Supplementing individual nutrients e.g., calcium,
phosphorus, vitamins, etc.
NUTRITIONAL SUPPLEMENTATION By fortification of expressed breast milk:

LBW infants, especially those who are born preterm Fortification with human milk fortifiers (HMF)
require supplementation of various nutrients to meet their Fortification with preterm formula
high demands. Since the requirements of VLBW infants
differ significantly from those with birth weights of 1500- Supplementing breast milk with minerals and vitamins:
2499 grams, they have been discussed separately. The following nutrients have to be added to the expressed
breast milk to meet the VLBW infants high requirements:
Infants with birth weights of 1500-2500g
Calcium1 and phosphorusa (140-160 mg/kg/d & 70-
These infants are more likely to be born at term or near 80 mg/kg/d respectively for infants on EBM)
term gestation (>34 weeks); hence, they do not require

TABLE 2. Nutritional Supplements for Infants with Birth Weights of 1500-2499 g

Nutrients Method of supplementation Dose When to start Till when?

Vitamin D* Multivitamin drops/syrup 200-400 IU/day 2 weeks of life Till 1 year of age
Iron Iron drops/syrup 2 mg/kg/day (maximum 15mg/day) 6 8 weeks of age Till 1 year of age

*See text

464 Indian Journal of Pediatrics, Volume 75May, 2008


83 53

Feeding of Low Birth Weight Infants

TABLE 3. Recommended Dietary Allowance in Preterm VLBW infants and the Estimated Intakes with Fortified/unfortified Human
Milk

At daily intake of 180 mL/kg


RDA* Only expressed EBM fortified with EBM fortified with Preterm
(Units/kg/day) breast milk# Lactodex-HMF formula (4g/100mL)
(4g/100mL)

Energy (kcal) 105-130 117 144 153


Protein (g) 3.5-4.0 2.46 3.2 3.4
Carbohydrates (g) 10-14 11.6 16.84 15.58
Fat (g) 5.4-7.2 6.8 7.1 9.06
Calcium (mg) 210 43.2 223 103
Phosphorus (mg) 110 22.2 112 52
Vitamin A (IU) 90-270 680 3308 980
Vitamin D (IU/day) 400 3.5 903 40
Vitamin E (IU) >1.3 1.9 6.3 3.6
Vitamin B1 (mcg) > 48 36.2 79.4 231
Vitamin B2 (mcg) > 72 84.2 156.2 564.2
Vitamin B6 (mcg) > 42 25.7 115.7 221
Folic acid (mcg) 39.6 6 150 36
Zinc (mg) >0.6 0.6 0.96 0.96
Remarks Deficient in protein, calcium, Deficient in protein Deficient in calcium,
phosphorus, and vitamins phosphorus, vitamin D,
B1, B6 and D; Zinc content and folic acid; protein is
is slightly less than the RDA slightly less.

* AAPCON 200415
#
Based on preterm mature milk (after 2 weeks of delivery)
(RDA, recommended dietary allowance; EBM, expressed breast milk)

Vitamin D 2 (400 IU/day), vitamin B complex and osmolality, clinical studies have not shown any significant
zinc b (about 0.5mg/day) usually in the form of adverse effects following fortification of human milk. The
multivitamin drops Cochrane review on fortification found short term
improvement in weight gain, linear and head growth
Folate (about 50 mcg/kg/day) 3
with out any increase in adverse effects such as NEC.17
Iron (2 mg/kg/day)4
The standard preparations of human milk fortifiers
a. Eg. Syr. Ostocalcium (Glaxo SmithKline Co.). Syr.
(HMF) used in developed countries are not available in
Ossopan-D (TTK Healthcare)
India. The only preparation available (Lactodex-HMF,
b. E.g. Dexvita drops (Tridoss Co.), Visyneral-zinc
Raptakos, Brett & Co. Ltd; Rs. 10/- per sachet) has some
drops (Lifeon Co.), Dexvita drops (Tridoss Co.)
limitations: inappropriately high vitamin A, no iron, etc.
c. E.g. Folium (Speciality Meditech Co.) Folvite
Short of other options, it may still have to be used in
(Wyeth Lederie Co.)
VLBW infants. One study from Chandigarh has reported
d. E.g. Ferrochelate (Albert David Co.) Tonoferon
better growth with its use.18
(East India Co.)
As seen from table 3, preterm VLBW infants on
However, one has to remember that supplementation of
expressed breast milk fortified with HMF do not require
minerals and vitamins would not meet the high protein
any supplementation (except for iron).
requirements of these infants (Table 3). Hence, this
method is usually not preferred. Fortification with preterm formula: The other option
available for fortification is preterm formula (e.g., Dexolac
To avoid abnormal increase in the osmolality, these
Special Care [Wockhardt Co.], Pre-Lactogen [Nestle Co.]).
supplements should be added at different times in the
The recommended concentration is 0.4g per 10mL of
day.
breast milk. Though more economical than fortification by
Fortification with HMF. Fortification of expressed breast HMF, this method has two major drawbacks - it is
milk with HMF increases the nutrient content of the milk difficult to measure such small amounts of formula
with out compromising its other beneficial effects (such as powder and the RDA of some minerals and vitamins (e.g.,
reduction of NEC, infections, etc.). Experimental studies calcium, phosphorus, vitamin D, folic acid) are not met
have shown that the use of fortified human milk results in even after fortification. While the former problem can be
net nutrient retention that approaches or is greater than managed to a certain extent by using a small scoop of 1g
expected intrauterine rates of accretion in preterm size for 25mL of milk, the later is circumvented by
infants.16 Though there are concerns about the increase in additional supplementation (Table 3).

Indian Journal of Pediatrics, Volume 75May, 2008 465


84 54

M.J. Sankar et al

The recommended dietary allowances (RDA) and the GROWTH MONITORING OF LBW INFANTS
estimated intakes with fortified human milk are given in
table 3.
Regular growth monitoring helps in assessing the
Fortification/supplementation in VLBW infants nutritional status and adequacy of feeding; it also
Summary: identifies those infants with inadequate weight gain.
The protocol for nutritional supplementation in VLBW All LBW infants should be weighed daily till the time
infants until 40 weeks PMA and beyond is described in of discharge from the hospital. Other anthropometric
tables 4 and 5. parameters such as length and head circumference should
be recorded weekly.
We use HMF fortification for all preterm (<32 weeks) VLBW
infants. It is started once they reach 150 mL/kg/day of enteral Both term and preterm LBW infants tend to lose
feeds in the dose recommended by the manufacturer (4g [2 weight (about 10% and 15% respectively) in the first 7
sachets] /100mL of expressed breast milk). We start iron at 4-6 days of life; they regain their birth weight by 10-14 days.
weeks in the dose of 2mg/kg/day. Thereafter, the weight gain should be at least 15-20g/kg/
day till a weight of 2-2.5 kg is reached. After this, a gain of
If HMF is unavailable or parents could not afford it, we
20 to 30 g/day is considered appropriate.19
fortify EBM with preterm formula (0.4g/10 mL). Since
calcium, phosphorus, and vitamin D intakes are low even Growth charts: Using a growth chart is a simple and
after fortification with formula, we supplement these effective way to monitor the growth. Serial plotting of
nutrients additionally (Table 4). We also add zinc and weight and other anthropometric indicators in the growth
iron as mentioned before. chart allows the individual infants growth to be
compared with a reference standard. It helps in early
We continue fortification till the infant reaches 40 weeks
identification of growth faltering in these infants.
PMA or attains 2 kg (whichever is later).

TABLE 4. Nutritional Supplementation in VLBW Infants Till 40 Weeks PMA

Type of feeding

Nutrients Only expressed EBM fortified with EBM fortified with Preterm formula
breast milk* Lactodex-HMF*

Calcium Start calcium supplements Not needed Start calcium supplements to meet the RDA
(140-160 mg/kg/day) once the infant is once the infant is on full enteral feeds
on full enteral feeds (e.g., Syr. Ostocalcium at 5-6mL/kg/d)
(e.g. Syr. Ostocalcium at 8-10mL/kg/d)
Phosphorus Start supplements (70-80 mg/kg/day) Not needed Start supplements to meet the RDA once the infant
once the infant is on full enteral feeds is on full enteral feeds (e.g., Syr. Ostocalcium at
(e.g., Syr. Ostocalcium at 8-10mL/kg/d) 5-6mL/kg/d)
Zinc and Start multivitamin supplements once the Not needed Not needed
vitamins infant is on full feeds
B1, B6 (e.g., ViSyneral zinc / Dexvita drops at
0.5mL/day)
Vitamin D (Usually obtained from multivitamin drops Not needed (Usually obtained from multivitamin drops and
and calcium supplements that contain calcium supplements that contain vitamin D)
vitamin D)
Folic acid Start supplements once the infant is on Not needed Start supplements once the infant is on full feeds
full feeds (e.g., Folvite/folium at 0.1 mL/day)
(e.g., Folvite/folium drops at 0.3 mL/day)
Iron Start iron (2 mg/kg/d) at 4-6 weeks of life Start iron (2 mg/kg/d) Start iron (2 mg/kg/d) at 4-6 weeks of life
(e.g., Tonoferon drops at 2 drops/kg/day) at 4-6 weeks (e.g., Tonoferon drops at 2 drops/kg/day)
(e.g., Tonoferon drops
at 2 drops/kg/day)

(PMA, postmenstrual age; EBM, expressed breast milk; HMF, human milk fortifier)
Note: The examples quoted are only indicative; Readers are encouraged to use similar products of their choice.

TABLE 5. Nutritional Supplementation in VLBW Infants After 40 Weeks PMA

Nutrients Method of Dose Till when?


supplementation

Vitamin D Multivitamin drops/syrup 200-400 IU/ day Till 1 year of age


Iron Iron drops/syrup 2 mg/kg/day (maximum 15mg/day) Till 1 year of age

466 Indian Journal of Pediatrics, Volume 75May, 2008


85 55

Feeding of Low Birth Weight Infants

Two types of growth charts are commonly used for anemia, feed intolerance,etc.
growth monitoring in preterm infants: intrauterine and If these measures are not successful, increase either
postnatal growth charts. Of these, the later chart is the
preferred because it is a more realistic representation of
the true postnatal growth (than an intrauterine growth Energy (calorie) content of milk by adding MCT oil,
chart) and also shows the initial weight loss that occurs in corn starch, etc. Infants on formula feeds can be
the first two weeks of life. The two postnatal charts that given concentrated feeds (by reconstituting 1 scoop
are most commonly used for growth monitoring of in 25 mL of water) OR
preterm VLBW infants are: Wrights and Ehrenkranz Feed volume to 200 mL/kg/day.
charts.20, 21 We use either of these in our unit.
Panel 4. Causes of inadequate weight gain
Once the preterm LBW infants reach 40 weeks PMA, 1. Inadequate intake
WHO growth charts should be used for growth Breastfed infants:
monitoring. Incorrect feeding method (improper positioning/
attachment)*
When to Discharge? Less frequent breastfeeding, not feeding in the night hours*
Prematurely removing the baby from the breast (before the
After they reach 34 weeks gestation and are above infant completes feeds)
1400g Infants on spoon /paladai feeds:
Incorrect method of feeding* (e.g., excess spilling)
AND Show consistent weight gain for at least 3 Incorrect measurement/calculation
consecutive days. Infrequent feeding
Not fortifying the milk in VLBW infants
Energy expenditure in infants who have difficulty in
MANAGEMENT OF INADEQUATE WEIGHT GAIN accepting spoon feeds
2. Increased demands
Illnesses such as hypothermia/cold stress*, bronchopulmonary
Inadequate weight gain is a common and pertinent dysplasia
problem in LBW infants. It starts at the time of initial Medications such as corticosteroids
admission and continues after discharge resulting in Underlying disease/pathological conditions
failure to thrive and wasting in the first year of life. The Anemia*, hyponatremia, late metabolic acidosis
common causes are summarized in Panel 4. Late onset sepsis
Feed intolerance and/or GER
Management of inadequate weight gain consists of the
* Common conditions
following steps: (EBM, expressed breast milk; GER, gastroesophageal reflux)
Proper counseling of mothers and ensuring adequate
support for breastfeeding their infants; includes FEED INTOLERANCE
assessment of positioning/attachment, managing
The inability to tolerate enteral feedings in extremely
sore/flat nipple etc.
premature infants is a major concern for the pediatrician
Explaining the frequency and timing of both /neonatologist caring for such infants. Often, feed
breastfeeding and spoon/paladai feeds: Infrequent intolerance is the predominant factor affecting the
feeding is one of the commonest causes of inadequate duration of hospitalization in these infants.
weight gain. Mothers should be properly counseled
There are no universally agreed-upon criteria to define
regarding the frequency and the importance of night
feed intolerance in preterm infants. 17 Various clinical
feeds. A time-table where mother can fill the timing
features that are usually considered to be the indicator(s)
and amount of feeding is very helpful in ensuring
of feed intolerance are summarized below (Panel 5):
frequent feeding.
Giving EBM by spoon/paladai feeds after Panel 5. Indicator(s) of feed intolerance17
breastfeeding also helps in preterm infants who tire Symptoms
out easily while sucking from the breast. 1. Vomiting (altered milk/bile or blood-stained)*
2. Systemic features: lethargy, apnea
Proper demonstration of the correct method of Signs
expression of milk and paladai feeding: It is important 1. Abdominal distension (with or without visible bowel
to observe how the mother gives paladai feeds; the loops)*
2. Increased gastric residuals: >2mL/kg or any change from
technique and amount of spillage should be noted.
previous pattern
This should be followed by a practical demonstration 3. Abdominal tenderness
of the proper procedure. 4. Reduced or absent bowel sounds
Initiating fortification of breast milk when indicated 5. Systemic signs: cyanosis, bradycardia, etc.

Management of the underlying condition(s) such as * Common signs

Indian Journal of Pediatrics, Volume 75May, 2008 467


86 56

M.J. Sankar et al

Fig. 3. Approach to feed intolerance in LBW infants

Of these, vomiting, abdominal distension, and increased gastric emptying. Since several factors (both systemic and
gastric residual volume form the triad for defining feed local) influence the gastric emptying, the residual volume
intolerance. is a poor and non-specific indicator of fed intolerance.
Measures to enhance the specificity - by quantifying the
Vomiting: The characteristic of vomitus is important in
volume and by using different cut-offs for defining feed
assessing the cause: while altered milk is usually
intolerance - have not been found to be much useful.
innocuous, bile- or blood-stained aspirate should be
Moreover, repeated gastric aspiration to look for residuals
thoroughly investigated.
could injure the delicate mucosa aggravating the local
Abdominal distension: It is essential to serially monitor pathology.
the abdominal girth in all preterm LBW infants admitted
We monitor the abdominal girth every 2 hours in all preterm
in neonatal nursery. This helps in early identification of
LBW infants admitted in the nursery. We do not routinely
feed intolerance and eliminates the need for routine
aspirate the gastric contents before giving next feed. It is done
gastric aspirate.
only if there is an increase in abdominal girth by >2 cm from
Gastric residual volume: It indicates the rapidity of the baseline.

468 Indian Journal of Pediatrics, Volume 75May, 2008


87 57

Feeding of Low Birth Weight Infants

Management of feed intolerance McMillan DD, Singhal N eds. Practical Procedures for the
Newborn Nursery, 2nd ed. New Delhi, Sagar Publishers, 2003:
The common factors attributed to feed intolerance in 71-78.
preterm infants are: immature intestinal motility, 7. Stocks J. Effect of nasogastric tubes on nasal resistance during
immaturity of digestive enzymes, underlying medical infancy. Arch Dis Child 1980; 55 : 17-21.
8. Premji SS, Chessell L. Continuous nasogastric milk feeding
conditions such as sepsis, inappropriate feed volume, and
versus intermittent bolus milk feeding for premature infants
giving hyperosmolar medications/feedings, and less than 1500 grams. Cochrane Database of Systematic Reviews
importantly, necrotizing enterocolitis (NEC). 2001, Issue 1. Art. No.: CD001819.
9. DeVille KT, Shulman RJ, Berseth CL. Slow infusion feeding
While issues such as feed volume and osmolality can enhances gastric emptying in preterm infants compared to
be controlled to an extent, feed intolerance due to bolus feeding. Clin Res 1993; 41 : 787A.
immaturity is rarely amenable to any intervention; 10. Mishra S, Agarwal R, Jeevasankar M, Deorari AK, Paul VK.
conservative management till the gut attains full maturity Minimal enteral nutrition. Indian J Pediatr 2008; 75 : 267-269.
11. Dorling J, Kempley S, Leaf A. Feeding growth restricted
is often the only option left.
preterm infants with abnormal antenatal Doppler results. Arch
The steps in evaluation and management of an infant Dis Child Fetal Neonatal Ed 2005; 90 : F359-F363.
12. Chawla D, Agarwal R, Deorari AK, Paul VK. Fluid and
with feed intolerance are given in fig. 3.
electrolyte management in term and preterm neonates. Indian
J Pediatr 2008; 75 : 255-259.
CONCLUSION 13. Annonymous. Nutrition. In Edmond K, Bahl R, eds. Optimal
feeding of law-birth weights infnats Technical Review. World
Optimal feeding of LBW infants is important for the Health Organization 2006; 42.
14. Abrams SA. Abnormalities of serum calcium and magnesium.
immediate survival as well as for subsequent growth.
In Cloherty JP, Eichenwald EC, Stark AR eds. Manual of
Unlike their normal birth weight counterparts, these Neonatal Care. 6th ed. Philadelphia: Lippincott Williams &
infants have vastly different feeding abilities and Wilkins 2008; p558.
nutritional requirements. They are also prone to develop 15. American Academy of Pediatrics Committee on Nutrition:
feed intolerance in the immediate postnatal period. It is Nutritional needs of preterm infants. In Kleinman RE ed.
Pediatric Nutrition Handbook American Academy of Pediatrics. Elk
important for all health care providers caring for such
Grove Village, IL, American Academy of Pediatrics, 2004; 23-
infants to be well versant with the necessary skills 54.
required for feeding them. It is equally important to have 16. Schanler RJ, Garza C. Improved mineral balance in very low
a protocol based approach to manage various issues that birth weight infants fed fortified human milk. J Pediatr 1987;
occur while feeding them. 112 : 452-456.
17. Kuschel CA, Harding JE. Multicomponent fortified human
milk for promoting growth in preterm infants. Cochrane
REFERENCES Database of Systematic Reviews 1998, Issue 4. Art. No.:
CD000343.
1. UNICEF. State of the Worlds Children 2005. New York: 18. Mukhopadhyay K, Narnag A, Mahajan R. Effect of human
UNICEF, 2004. milk fortification in appropriate for gestation and small for
2. Bang A, Reddy MH, Deshmukh MD. Child mortality in gestation preterm babies: a randomized controlled trial. Indian
Maharashtra. Economic Political weekly 2002; 37 : 4947-4965. Pediatr 2007; 44 : 286-290.
3. Edmond KM, Kirkwood BR, Tawiah CA, Agyei SO. Impact of 19. Schanler RJ. Enteral Nutrition for the High-Risk Neonate. In
early infant feeding practices on mortality in low birth weight Taeusch HW, Ballard RA, Gleason CA eds. Averys Diseases of
infants from rural Ghana. J Perinatol 2008; [Epub ahead of the Newborn, 8th ed. Philadelphia, Saunders, 2005; 1043-60.
print] 20. Wright K, Dawson JP, Fallis D, Vogt E, Lorch V. New
4. Levitsky DA, Strupp BJ. Malnutrition and the brain: changing postnatal growth grids for very low birth weight infants.
concepts, changing concerns. J Nutr 1995; 125 : 2212S2220S. Pediatrics 1993; 91 : 922-926.
5. Omari TI, Rudolph CD. Gastrointestinal Motility. In: Polin RA 21. Ehrenkranz RA, Younes N, Lemons JA, Fanaroff AA,
and Fox WW eds. Fetal and Neonatal Physiology, 2 nd ed. Donovan EF, Wright LL et al. Longitudinal growth of
Philadelphia, WB Saunders Co, 1998; 1125-1138. hospitalized very low birth weight infants. Pediatrics 1999; 104
6. Anonymous. Feeding. In Deorari AK, Paul VK, Scotland J, : 280-289.

Indian Journal of Pediatrics, Volume 75May, 2008 469


88
89
90
91
92 58

Symposium on AIIMS Protocols in Neonatology - V

Follow-up of High Risk Neonates


Pradeep Kumar, M. Jeeva Sankar, Savita Sapra, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, India

ABSTRACT
The improvement in perinatal care has led to increase in survival as well as reduction of morbidity in sick newborns. These
babies need to be followed up regularly to assess growth and neurodevelopmental outcome and for early stimulation and
rehabilitation. We present a protocol describing the various components of a follow up program, and services. [Indian J
Pediatr 2008; 75 (5) : 479-487] E-mail: aranag@rediffmail.com

Key words: Follow up; Neurodevelopmental; Outcome; Early Stimulation

Improving perinatal and neonatal care has led to be placed on these.


increased survival of infants who are at-risk for long-
However, a rigorous follow-up of all the neonates
term morbidities such as developmental delay and
discharged from a particular health facility would
visual/hearing problems 1, 2 Moreover, many of these
neither be practical nor feasible. Therefore, it is
neonates (e.g., extremely low birth weight infants) tend
important to select a cohort of neonates who are at a
to have higher incidence of growth failure and ongoing
higher risk of developing these adverse outcomes at-
medical illnesses A proper and appropriate follow-up
risk infants. Surprisingly, there are no standardized
program would help in early detection of these
guidelines for follow up of high risk infants even in
problems thus paving way for early intervention.
tertiary care centers. 4 We have devised a follow up
protocol which identifies the subset of neonates to be
IMPORTANCE OF FOLLOW-UP CARE followed up and outlines the optimal time for follow-up
visits and the appropriate assessment measures to be
adopted.
Numerous studies have shown that despite substantial
improvements in the neonatal mortality, the incidence
of chronic morbidities and adverse outcomes among SETTING UP OF FOLLOW-UP SERVICES
survivors has not declined much.3 This highlights the
need for a follow-up care service that would ensure
High risk infants follow-up requires a multidisci-
systematic monitoring of the general health and
plinary approach involving a team of pediatricians,
neurodevelopmental outcomes after discharge from the
child psychologist, pediatric neurologist, ophthalmo-
hospital. The monitoring would help the infants and
logist, otorhinolaryngologist, physiotherapist, occupa-
their families (early identification of problems and
tional therapist, medical social worker, and a dietician.
hence early rehabilitation services) as well as the
The respective role of each team member is summarized
physicians involved in their care (to improve the quality
in table 1.
of care provided and for research purposes). There is a
common perception that high risk follow-up mainly Ideally, all the required personnel should be
concerns with detection and management of available under one roof at a place earmarked for
neurosensory disability. Infact growth failure and follow-up care. If this is not feasible, at least the services
ongoing illnesses are equally, if not more important of pediatrician, clinical psychologist, dietician, medical
issues in high risk follow-up. Adequate emphasis must social worker, and physiotherapist should be ensured
in the follow-up clinic. Medical social worker is an
important member of the team liasoning with the
Correspondence and Reprint requests : Dr Ramesh Agarwal, family and helps them to keep follow up visits. Infants
Assistant Professor, Department of Pediatrics, All India Institute who need hearing/visual assessment or speech therapy
of Medical Sciences, Ansari Nagar, New Delhi 110029, India. can be referred to the concerned specialist on fixed
[Received April 24, 2008; Accepted April 24, 2008] days.

Indian Journal of Pediatrics, Volume 75May, 2008 479


93 59

Pradeep Kumar et al

TABLE 1. Personnel Required for Follow-up Program and their Individual Roles

S. No Team member Role(s)

1. Pediatrician(s) / neonatologist(s) Serves as the nodal person of the team


To assess growth and screen for developmental delay
To manage intercurrent illnesses
2. Child psychologist(s) For formal neurodevelopmental assessment

Screening for behavioral problems and their management
3. Pediatric neurologist Long-term management of neurological illnesses such as seizures
4. Ophthalmologist Follow-up of ROP screening/treatment

Assessment of visual acuity and screening for problems such as
strabismus, nystagmus, refractory errors, etc.
5. Otorhinolaryngologist Hearing assessment (BERA, OAE, etc.)

Management of hearing impairment, if any
6. Dietician Dietary advice regarding complementary feeding

Management of infants with failure to thrive and those with special
needs (e.g. galactosemia)
7. Medical social worker To take care of the social issues to help improve follow up rates

8. Physiotherapist Assessment and grading of muscle tone and power


Plan an appropriate training program for each infant with tone
abnormalities
To teach the parents for continuing the prescribed exercises at home
9. Speech / occupational therapist Rehabilitation of infants with impairment/disability

WHO NEEDS FOLLOW-UP CARE? The developing brain of premature babies is


extremely vulnerable to injury; the risk for
Selection of high-risk infants should be based on the neurodevelopmental deficit increases with decreasing
gestational age, birth weight, occurrence and severity of gestational age and birth weight resulting in relatively
perinatal/neonatal illnesses, interventions received in high risk of cerebral palsy, developmental delay,
the neonatal intensive care unit (NICU), presence of hearing and vision impairment and subnormal
malformations, etc. It can further be modified for each academic achievement 5 . Similarly, small for date
unit based on their admission and outcome profiles. infants (birth weight < 3rd centile) are also at significant
risk of poor long term outcomes. Those who required
Panel 1 lists the cohort of high risk infants whom we
mechanical ventilation for more than 24hours, babies
follow-up in our unit.
with metabolic problems symptomatic hypoglycemia
Panel 1. High Risk Neonates Who Need Follow-up Care as half of them have abnormal neurodevelopmental
(customize as per local policy) outcome, symptomatic hypocalcemia, birth asphyxia
Babies with <1800g birth weight and/or gestation <35 Apgar score 3 or less at 5 min, abnormal neurological
weeks examination at discharge, seizures, hyperbilirubinemia
Small for date (<3 rd centile) and large for date (>97th > 20mg/dL or requirement of exchange transfusion, Rh
centile) hemolytic disease of newborn as they have anemia
Perinatal asphyxia - Apgar score 3 or less at 5 min and/or
presenting till three to six months age, infections
hypoxic ischemic encephalopathy
Mechanical ventilation for more than 24 hours
culture positive sepsis or meningitis, babies born to HIV
Metabolic problems Symptomatic hypoglycemia and infected mothers, twin with intrauterine death of co-
hypocalcemia twin due to increased incidence of cerebral venous
Seizures thromboembolic phenomenon, twin to twin transfusion
Infections meningitis and/or culture positive sepsis or major malformation. All infants cared for in the
Shock requiring inotropic/vasopressor support NICU should have periodic preventive assessment by
Major morbidities such as chronic lung disease, their primary care physicians which should include
intraventricular hemorrhage, and periventricular
leucomalacia regular assessment of growth, sensory function,
Infants born to HIV-positive mothers behavior and neurodevelopment. Infants with suspect
Twin with intrauterine death of co-twin findings should be referred for more comprehensive
Twin to twin transfusion evaluation to a center with experience in follow up of
Hyperbilirubinemia > 20mg/dL or requirement of high risk neonates.
exchange transfusion
Rh hemolytic disease of newborn
PRE-REQUISITES FOR FOLLOW-UP
Major malformations
Inborn errors of metabolism / other genetic disorders
Abnormal neurological examination at discharge
To ensure proper follow-up of the high risk infants,

480 Indian Journal of Pediatrics, Volume 75May, 2008


94 60

Follow-up of High Risk Neonates

parents (especially mother) and other family members PROCEDURE FOR FOLLOW-UP
should be counseled even before discharge from the
hospital. Discharge should be planned well in advance Venue: A specified site should be earmarked for follow
so that the mother can be counseled adequately. up services. The parents should be properly
communicated about the venue and it should also be
Discharge planning: Discharge planning should ideally
mentioned in the discharge summary. Registration
begin as soon as the baby is admitted in the nursery.
procedure at the follow-up clinic should be simplified
This gives adequate time for the caretakers to ask
to avoid any undue delay. Ongoing illness is common
questions and practice skills. The following criteria
problem among these infants. If the infant develops any
should be fulfilled before discharging a high risk infant:
illness requiring admission, priority should be given for
Hemodynamically stable; able to maintain body the same.
temperature in open crib
Record maintenance: There should be a separate but
On full enteral feeds (either breast feeding or by uniform file for each high risk infant . We have separate
paladai/spoon) files for male and female babies. Male babies get blue
and female babies get pink files. Addresses and
Parents confident enough to take care of the baby at
telephone numbers should be entered clearly in the file.
home
If possible, an alternate address and telephone number
Has crossed birth weight and showing a stable should also be recorded. It may be good idea to enquire
weight gain for at least three consecutive days; in an important landmark for locating the house in case
case of very low birth weight infants, weight should one needs to make a home visit. The family should also
be at least 1400 grams before considering for be given a booklet containing follow-up information.
discharge and should have crossed birth weight.
Schedule: The follow up schedule should be explained
Not on any medications (except for vitamins and to the parents (see below). Timings should be fixed and
iron supplementation). Ideally preterm babies on adhoc visits should be discouraged.
theophylline therapy for apnea of prematurity should
Corrected age: Age of the child since the expected date
be off therapy for at least five days to make sure that
of delivery. The correction for gestational immaturity at
there is no recurrence.
birth should be done till 24 months age. All
Received vaccination as per schedule (based on developmental milestones are assessed according to
postnatal age). corrected age to compensate for the prematurity. The
addition of complementary feeds is also according to
These criteria can be individualized to meet the infant
corrected age.
and family needs.
Postnatal age: Age of the child since birth.
Counseling prior to discharge: Counseling plays an
Immunization is done according to postnatal age.
important role in the care of these babies at home;
regular counseling sessions should be done before
discharge. Parents should be given advice regarding: WHEN TO FOLLOW-UP
Temperature regulation proper clothing, cap, socks,
Kangaroo mother care etc. For the purpose of follow-up visits, at-risk infants can
Feeding type and amount of milk, method of be grouped under two major categories: (1) preterm/
administration, and nutritional supplementation, if LBW infants and (2) infants with other conditions. The
any. follow-up schedule for both these categories has been
summarized in table 2. This schedule represents
Prevention of infections hand washing, avoidance minimum number of visits of high risk neonates. If the
of visitors, etc. baby has ongoing issues or illness, more frequent visits
Follow-up visits where and when (Table 2) are recommended. Please note that first contact of the
Danger signs recognition and where to report if infant with the health providers after discharge is
signs are present important and helps in identification of adjustment
problems at home. Ideally this contact should be
Vaccination schedule, next visit, etc. achieved by the home visit.
Special needs e.g., next visits for ROP screening.
The selection of age of assessment depends on
If possible the family should be provided with the developmental acquisitions available at a given age,
telephone number of the health care provider e.g., on- availability and applicability of appropriate test
duty doctor in case the family needs to consult for instruments at specific ages and the cost and feasibility
infants illness. of long-term tracking in the population in question. The

Indian Journal of Pediatrics, Volume 75May, 2008 481


95 61

Pradeep Kumar et al

TABLE 2. Follow-up Schedule of At-risk Infants

Cohort Schedule for follow-up

Infants with <1800g birth weight and/or After 3-7 days of discharge to check if the baby has been adjusted well in
gestation <35 weeks the home environment. Every 2 weeks until a weight of 3 kg
(immunization schedule until 10-14 weeks to be covered in these visits)
At 3, 6, 9, 12 and 18months of corrected age and then every 6 months
until age of 8years
All other conditions 2 weeks after discharge
At 6, 10, 14 weeks of postnatal age
At 3, 6, 9, 12 and 18months of corrected age and then every 6 months
until age of 8years

Note: If a preterm infant (< 35 weeks) develop any other morbidity covered in other conditions, he should be followed up as per
the schedule outlined for the first group of cohort

long term follow up of complete cohorts is optimal for amount, dilution and mode of feeding should be noted
determining the outcome of high risk neonates and the if supplemental feeding is given. It is a good idea to
safety of antenatal and perinatal interventions. Very enquire about source of milk as milk supplied by local
low birth weight babies or those born at less than 33 vendors is often diluted (dilution has the same impact
weeks gestation should be followed up for eye check up on the infant whether done by the family or the
for retinopathy of prematurity till the postnatal age of vendor!). It is also important to record the duration of
44 weeks. exclusive breast feeding. If a baby is not gaining
adequate weight on exclusive breast feeding take care
Some neurological abnormalities that are identified
of any illness, maternal problems which may interfere
in the first year of life are transient or improve whereas
with feeding and milk output. If poor weight gain
findings in other children may worsen over time.6 By 12
persists despite all measures to improve breast milk
months corrected age the cognitive and language
output, supplementation can be considered.
assessment can be done. By 18-24 months corrected age
there is improved prediction to early school age Complementary feeding should be started at 6
performance.7, 8, 9, 10 The importance of long term follow months corrected age. Initially, semisolids should be
up lies in the fact that minor neurological disabilities advised in accordance with the local cultural practices.
may not be detected early and become apparent only Spend adequate time on explaining what to give and
with increasing age. Standard follow-up for many how to give. The common practice of giving too little or
multicenter networks is currently at 18-24 months too dilute complementary food such as rice-water, dal-
corrected age. water, too much of juice, etc should be discouraged. The
recommended meal frequencies assuming a diet with
energy density of 0.8 kcal per gram or above and low
WHAT SHOULD BE DONE AT FOLLOW-UP?
breast milk intake are: 2-3 meals per day for infants
aged 6-8 months; 3-4 meals per day for infants aged 9-
Table 3 summarizes the plan for follow-up. 11 months and children 12-24 months; additional
nutritious snacks may be offered 1-2 times a day, as
Assessment of feeding and dietary counseling desired. Complementary foods should be varied and
Parents should be asked about the infants diet and include adequate quantities of meat, poultry, fish or
offered dietary counseling at each visit. Breast feeding eggs, as well as vitamin A-rich fruits and vegetables
frequency and adequacy should be assessed. The every day. Where this is not possible, the use of fortified

TABLE 3. Follow-up Plan for High Risk Infants

Assessment Age in months

Assessment of feeding and dietary counseling 1 2 3 6 9 12 15 18 24 ......8years


Growth monitoring All visits
Immunization As per schedule (based on postnatal age)
Neurological examination
Developmental assessment and DQ
Hearing (BERA)
Ophthalmic evaluation
USG/CT brain As indicated
if previous test abnormal

482 Indian Journal of Pediatrics, Volume 75May, 2008


96 62

Follow-up of High Risk Neonates

complementary foods and vitamin mineral and DMeQ = MeA/CA x 100). The composite DQ is
supplements may be necessary to ensure adequacy of derived as an average of DMoQ and DMeQ.
particular nutrient intakes. As infants grow, the
The Vineland Social Maturity Scale measures social
consistency of complementary foods should change
competence, self-help skills, and adaptive behavior
from semisolid to solid foods and the variety of foods
from infancy to adulthood. The Vineland scale consists
offered should increase. By eight months, infants can
of a 117-item interview with a parent or other primary
eat finger foods and by 12 months, most children can
caregiver.
eat the same types of food as the rest of the family. The
major problem with the family food is that it is not It is emphasized here that developmental
nutrient-rich.10 stimulation of the child should not be delayed if the
above mentioned tests are not available. Age
Growth monitoring
appropriate stimulation should be provided to these
Growth (including weight, head circumference, mid- babies. Mental development index and Psychomotor
arm circumference and length) should be monitored development index at 3, 12,18 and 24 months and
and plotted on an appropriate growth chart at each every 3 months if abnormal.
visit. We use Wrights charts (till 40 weeks PMA) and
Immunization
WHO growth charts (for preterm infants after 40 weeks
PMA and for term infants) for growth monitoring . The Immunization should be ensured according to
infants growth pattern (slope of the curve) is compared chronological age . Parents should be offered the option
with the standard curve; any deviation should be noted of using additional vaccines such as Hemophilus
and appropriate remedial action taken. Weight should influenzae B, typhoid and MMR.
be taken on an electronic weighing scale. Length should
Ongoing problems
be measured with an infantometer. The infant should be
held supine and legs fully extended. The feet should be They should be mentioned in the follow up notes . The
pressed against the movable foot piece with the ankles management of ongoing illnesses is an integral part of
fixed to 90. Head circumference should be measured any high risk follow up program. The hospital
with nonstretchable fiberglass tape.11 admission of the child should be prioritized, if
required.
Developmental assessment
Neurological assessment
Assessment of developmental milestones should be
done according to the corrected age. The milestones Evaluation of muscle tone is an integral part of the
should be assessed in four domains- gross motor, fine neurological examination. A waxing and waning
motor, language, and personal-social. The date of pattern of neuromotor development from 28 weeks of
assessment and the infants corrected age should be gestation to the end of first year of life was reported by
mentioned against each milestone. Based on the date of Amiel-Tison. From 28 to 40 weeks gestation, the
achievement of milestones in a particular domain and acquisition of muscle tone and motor function spreads
the expected age of achieving them, the developmental from lower extremities towards the head. After full term,
age can be calculated. the process is reversed so that relaxation and the motor
control proceed downwards for the next 12 to 18
Infants who lag behind in any domain should
months. So the upper limbs begin to relax and acquire
undergo a formal developmental evaluation by a
skills before the lower limbs. The axial tone follows a
clinical psychologist using tests such as Developmental
similar pattern. Head control appears first followed by
assessment of Indian Infant II (DASII II).12 This scale
the ability to sit, stand and walk. Hypertonia or
consists of 67 items for assessment of motor
hypotonia should be looked for by measuring the
development and 163 items for assessment of mental
following angles: adductor angle, popliteal angle, ankle
development. Motor scale assesses control of gross and
dorsiflexion, and scarf sign; any asymmetry between
fine motor muscle groups. Mental scale assesses
the extremities should also be recorded. Any history of
cognitive, personal and social skills development. Both
seizures or involuntary movements should also be
mental development index and psychomotor
recorded.
development index can be calculated by DASII. The age
placement of the item at the total score rank of the scale Hypertonia in lower limbs is defined as when either
is noted as the child developmental age. This converts adductor angle is restricted to less than the age specific
the child total scores to his motor age (MoA) and norms as per Amiel-Tison or if there is scissoring or
mental age(MeA). The respective ages are used to tight tendo-achilles or restriction of ankle dorsiflexion
calculate his motor and mental development quotients on extension of knee. Hypertonia in upper limbs is
respectively by comparing them with his chronological defined as when scarf sign does not cross midline at
age and multiplying it by 100. (DMoQ = MoA/CA x 100 one year corrected age. Hypertonia of the neck

Indian Journal of Pediatrics, Volume 75May, 2008 483


97 63

Pradeep Kumar et al

TABLE 4. Muscle Tone Norms

Age Adductor Popliteal Dorsiflexion Scarf sign


(months) angle angle angle

0-3 40 -80 80 -100 60 -70 Elbow does not cross midline


4-6 70 -110 90 -120 60 -70 Elbow crosses midline
7-9 110 -140 110 -160 60 -70 Elbow goes beyond axillary line
10-12 140 -160 150 -170 60 -70

Fig. 1. Amiel-Tison Method oa assessment of tone in infants

484 Indian Journal of Pediatrics, Volume 75May, 2008


98 64

Follow-up of High Risk Neonates

extensors can be inferred by an increased gap between 24 and so on. There is a low vision card containing 25.5
the nape of the neck and examination table with the 23 cm patch of 0.23 cm cycle/cm( 2.2 cm wide black or
infant lying in supine position. white stripes). The seventeenth card is a blank grey card
with no grating pattern. The gratings have 82 84%
The following angles should be measured to assess
contrast and are matched to the surrounding grey card
tone as shown in fig 1, table 4.
to within 1% in space average luminance. This
Truncal extensor hypertonia: there is a tendency of body minimizes the chance of a patient fixating because of
to go into hyperextension or opisthotonus. brightness difference. Detection of pattern alone
determines the fixating preference. Proper illumination
Cerebral palsy: Definitely abnormal neurological
without any shadows should be ensured (10 candelas /
examination with upper motor neuron signs with
sqm). Testing distance from patients eyes to the cards
motor developmental delay.
should be maintained constant as it determines the
Spastic hypertonia syndromes visual acuity. Children from 7m to 3y should be tested at
55 cm and later at 84 cm.
Hemiplegia- only one half of body involved
Diplegia- paresis of lower limbs more than upper limbs Rehabilitation for visual impairment should be early
Quadriplegia- Paresis of all four limbs with upper limb so that the child gets appropriate stimulation. If delayed
involvement equal to or more than lower limbs. the restoration of the vision may not be possible
Abnormal neurological examination should be defined because of continuous sensory deprivation of the optic
as definite abnormalities In the form of: nerve. The child should be provided with glasses or
a) Brisk reflexes with hypertonia or corrective surgery as appropriate. It should be
b) Brisk reflexes with hypotonia or emphasized that a good high risk follow up program
c) Definitely and consistently elicited asymmetrical does not only pick up handicaps early but also ensures
signs or early corrective measures and rehabilitation. This
d) Persistent abnormal posturing or abnormal emphasizes the multidisciplinary and well coordinated
movements approach to such babies
The tone abnormalities should be taken care by regular Hearing evaluation
physiotherapy. This improves mobility of joints and
High risk infants have higher incidence of moderate to
locomotion of the child. The child should be provided
profound hearing loss (2.5-5% vs 1%). Since clinical
with special shoes if required. Orthopedic evaluation
screening is often unreliable, brainstem auditory
should be done and corrective surgery for contractures
evoked responses (BAER/BERA) should be performed
should be done as required. All possible efforts should
between 40 weeks PMA and 3 months postnatal age. A
be made to improve mobility of these children and make
screening BERA is usually done initially. If this is
them functionally less dependent and independent if
abnormal, a diagnostic BERA should be done within 2
possible.
weeks of the initial test. Infants with unilateral
Eye evaluation abnormal results should have follow-up testing within
three months. The test should be carried out in a sound-
The check-up for retinopathy of prematurity starts in proof room and the infant should be sedated with oral
the NICU and continues till 44 weeks postconceptional triclofos 50mg/kg 30 min before the procedure. To
age or till the retinal vessels have matured. Refer to measure the electrical pulses, small monitoring
protocol on Retinopathy of prematurity.13 electrodes are placed on the scalp. Earphones provide a
At 9 months corrected age the ophthalmologist clicking noise to the ear and the response from the
should evaluate the baby for vision, squint, cataract and brainstem is measured time-locked to the clicks. The
optic atrophy. Subjective visual assessment can be clicks may become louder or softer, faster or slower, to
made from clinical clues as inability to fixate eyes, see how the auditory responds to these different
roving eye movements and nystagmus. Objective visual stimulus parameters. The other method of assessment
assessment should be done with the Teller Acuity Card. for hearing is oto-acoustic emission (OAE). This
It has seventeen 25.5 51 cm cards. Fifteen of these records acoustic feedback from the cochlea through the
contain 12.5 12.5 cm patches of square-wave gratings( ossicles to the tympanic membrane and ear canal
vertical black and white strips) ranging in spatial following a click stimulus. It is quicker to perform than
frequency from 38.0 cycles/cm to 0.32 cycles/cm. The BERA but is more likely to be affected by debris or fluid
range is in half octave steps. A cycle consists of one in the external and middle ear. It is unable to detect
black and one white stripe and an octave is a halving or some form of sensorineural hearing loss including
doubling of spatial frequency. In Snellens terms it is an auditory dyssynchrony.
halving or doubling of the denominator e.g. 6/6, 612, 6/ The severity of hearing loss is profound (70 dB or
24. Half octave steps would be 6/6, 6/9, 6/12, 6/18, 6/ more of hearing loss), severe (50 dB - 70 dB), moderate

Indian Journal of Pediatrics, Volume 75May, 2008 485


99 65

Pradeep Kumar et al

(30 dB - 50 dB) and mild (15 dB - 30 dB). 2-4 months


The audiological testing should be done at 3 months General stimulation
of age. Infants with true hearing loss should be referred
Hold the baby at the shoulder
for early intervention to enhance the childs acquisition
Place things just out of the reach of the baby.
of developmentally appropriate language skills. The
Stimulate him to reach out and grasp the object
child should be provided with hearing aids and if
severe to profound hearing loss cochlear implants Auditory
should be considered by 12 months age. Fitting of
Give sound producing toys
hearing aids by the age of 6 months has been associated
Talk to the child more frequently
with improved speech outcome. Initiation of early
Point out the names of objects shown to the child
intervention services before three months age has been
associated with improved cognitive development at 3 Visual
years age.14
Hang bright objects about 30cm above the crib
Maintain eye contact while talking to the child
EARLY STIMULATION
Tactile
The high risk baby requires more attention of the family Give the child paper to crumble and things to bite
members. Parents and family members need to aid the and suck
development process in an age appropriate way Place the child on a rubber mat on the ground
spending quality time with children. Such interactions allowing him to move freely
improve parent child relationship and bring about
4-6 months:
positive parental attitudinal change. Effective parents
supervise their children in an age appropriate way, use General activities:
consistent positive discipline, communicate clearly and
Sit the baby in the mothers lap and ask her to gently
supportively, and show warmth, affection,
bounce her knees singing songs.
encouragement, and approval. The actions of the child
Place the child flat on the back on the ground over a
should be appreciated. This makes him happy and
soft surface. Show him a colorful toy. Slowly turn him
encourages doing more activities.
by flexing the far away leg. Assist him to turn over
0-2 months: the tummy.
Show an attractive toy and encourage the child to
Activities
reach out to it.
Maintain eye to eye contact Put your hands under the childs feet and move his
Talk and sing to the baby while bathing, dressing legs up and down like pedaling a cycle.
and feeding
Auditory
Help the baby to turn his head to sound and light
Shake a bell or a squeaky toy over the head of the
Auditory
baby. Encourage him to turn his head and locate the
Provide different sounds to the child like rattle, bell, sound
squeezing a toy. Make the child listen to music, high
pitched and low pitched human sounds 6-8 months
Humming in a soft low voice
Call the child by his name
Visual Make the child sit as long as possible. Give support
to his pelvis.
Keep the baby in a well lighted room
Give him pieces of paper to tear
Shine mobile, color balls and hang bright clothes
Encourage him to roll over his tummy by showing
Tactile him colorful toys on one side.
Put the baby on different surfaces like soft clothes, 8-10 months
mattresses, rubber mat and mothers lap
Change the childs position frequently like putting Make the child stand by holding onto the furniture
on his back, sides and tummy Encourage the child to clap hands
Give him a small container and ask to drop small
Kinesthetic thing into it.
Support the head and gently rock the child avoiding Encourage him to produce monosyllables.
sudden jerky movements Show him picture books and assist to turn the pages.

486 Indian Journal of Pediatrics, Volume 75May, 2008


100 66

Follow-up of High Risk Neonates

10-12 months: M, Paul VK. Survival and morbidity in Extremely Low Birth
Weight (ELBW) infants. Indian Pediatr 2003; 40: 130-135.
Let the child play with other children 2. Costello D, Friedman H, Minich N, Siner B, Taylor G,
Name the body parts while bathing him Schuchlter M, Hack M. Improved neurodevelopmental
Take the child on a walk and show him different outcomes for extremely low birth weight infants in 2000-
2002. Pediatrics 2007; 119: 37-45.
animals and birds
3. Escobar G, Littenberg B, Petitti DB Outcome among
Do simple actions like clapping, bye-bye and surviving very low birthweight infants: a meta-analysis.
encourage copying these actions. Arch Dis Child Feb1991; 66: 204 - 211.
Encourage him to pull to stand by holding the 4. Wang CJ, McGlynn EA, Brook RH et al. Quality-of-care
furniture indicators for the neuro-developmental follow-up of very
low birth weight children: results of an expert panel
Make the child sit in front of a mirror so that he can
process. Pediatrics 2006; 117 : 2080-2092.
see himself 5. Vohr BR, Wright L, Anna M, Perritt R, Poole WK, Tyson JE
et al. Center for the Neonatal Research Network Center
12-15 months:
differences and outcomes of exteremely low birth weight
Give picture books to the child. Talk about what you infants. Pediatrics 2004; 113 : 781-789.
6. Drillien C. Abnormal neurological signs in the first year of
see and let him turn the pages
life in low birth weight infants: possible prognostic
Ask him to put cubes one over the other significance. Dev Med Child Neurol 1997; 14 : 575-84.
Ask him to put things into the container and then 7. Weisglas-Kuperus N, Baerts W, Smrkovsky M, Sauer PJ.
take out things out of the container. Effects of biological and social factors on the cognitive
Hide a small toy under a cloth. Encourage the baby to development of very low birth weight children. Pediatrics
find the hidden toy. 1993; 92 : 658-665.
8. Dezoete JA, MacArthur BA, Tuck B. Prediction of Bayley
Ask the child to scribble by drawing a few lines. and Stanford-Binet scores with a group of very low
First demonstrate what he is supposed to do. birthweight children. Child Care Health Dev 2003; 29 : 367-
372.
How to ensure a good follow up rate 9. Lee H, Barratt MS. Cognitive development of preterm low
birth weight children at 5 to 8 years old. J Dev Behav Pediatr
The importance of follow up should be emphasized 1993; 14 : 242-249.
frequently to the parents. The permanent and present 10. Report of the global consultation, and summary of guiding
addresses along with phone numbers should be kept to principles for complementary feeding of the breastfed child
Authors: World Health Organization
ensure follow up. If the parents do not turn up for follow 11. Implementation of the WHO Multicenter Growth Reference
up they should be telephoned and letters should be Study in India N. Bhandari, S. Taneja, T. Rongsen, J.
posted to ensure good follow up rates. There should be Chetia, P. Sharma, R. Bahl, D. K. Kashyap, and M. K. Bhan,
a dedicated person who can adjust the timing with the for the WHO Multicenter Growth Reference Study Group
parents. If possible home visits should be arranged for 12. Phatak B. Mental and motor growth of Indian babies (1-30
months). Final report. Department of Child Development,
those who do not turn up. There should be a
MSUB, Baroda, 1970.
comprehensive assessment of the child under one roof 13. Chawla D, Agarwal R., Deorari AK, Paul VK. Retinopathy
to minimize the hassles of roaming from one corner of of Prematurity. Indian J Pediatr 2008; 75 : 73-76.
the hospital to the other. 14. NIH Joint Committee on Infant Hearing. Year 2000
position statement: Principles and guidelines for early
hearing detection and intervention programmes. Pediatrics
REFERENCES
2000; 106 : 798-817.

1. Narayan S, Aggarwal R, Upadhyay A, Deorari AK, Singh

Indian Journal of Pediatrics, Volume 75May, 2008 487


101
102
103
104
105 67
Indian J Pediatr (2010) 77:11231128
DOI 10.1007/s12098-010-0176-0

SYMPOSIUM ON AIIMS PROTOCOLS IN NEONATOLOGY-I

Hypocalcemia in the Newborn


Ashish Jain & Ramesh Agarwal & M. Jeeva Sankar &
Ashok Deorari & Vinod K. Paul

Received: 27 July 2010 / Accepted: 2 August 2010 / Published online: 25 August 2010
# Dr. K C Chaudhuri Foundation 2010

Abstract Hypocalcemia is a frequently observed clinical reabsorbtion, skeletal calcium stores, and vitamin D status.
and laboratory abnormality in neonates. Ionic calcium is Hence, after delivery, calcium levels start decreasing (the
crucial for many biochemical processes including blood rate and extent of decrease is inversely proportional to the
coagulation, neuromuscular excitability, cell membrane gestation) and reaches a nadir of 7.5-8.5 mg/dL in healthy
integrity, and many of the cellular enzymatic activities. term babies by day 2 of life. This drop in postnatal SCa
Healthy term infants undergo a physiological nadir in may be related to hypoparathyroidism, end organ unre-
serum calcium levels by 24-48 h of age. This nadir may sponsiveness to parathyroid hormone [2], abnormalities of
drop to hypocalcemic levels in high-risk neonates including vitamin D metabolism, hyperphosphatemia, hypomagnese-
infants of diabetic mothers, preterm infants and infants with mia, and hypercalcitonemia [3] which occurs by 12-24 h of
perinatal asphyxia. The early onset hypocalcemia which age. PTH levels increase gradually in the first 48 h of life
presents within 72 h requires treatment with calcium and normal levels of SCa are regained by 3rd day of life
supplementation for at least 72 h. In contrast, late onset [4]. The efficacy of the intestinal absorption of calcium and
hypocalcemia usually presents after 7 days and requires the renal handling matures by 24 wks. This transition
longer term therapy. phase is responsible for the increased risk of early onset
hypocalcemia in high-risk neonates.
Keywords Hypocalcemia . Newborn . Therapy

Calcium homeostasis in newborn


During the last trimester, calcium is actively transferred from
mother to the fetus as demonstrated by a significantly high Body calcium exists in two major compartments: skeleton
level of total calcium concentration in cord blood compared (99%) and extracellular fluid (1%). Calcium in the
to maternal serum [1]. Parathyroid hormone (PTH) and extracellular fluid is present in three forms: (a) bound to
calcitonin (CT) do not cross the placental barrier. The PTH albumin (40%) (b) bound to anions like phosphorus, citrate,
related peptide (PTHrP) is the main regulator of the positive sulfate and lactate (10%) and (c) free ionized form (50%)
calcium balance across the placenta. Serum calcium (SCa) [5]. Ionized calcium is crucial for many biochemical
in the fetus is 10-11 mg/dL at term (1-2 mg higher as processes including blood coagulation, neuromuscular
compared to mother). excitability, cell membrane integrity and function, and
After birth the SCa levels in newborns depend on the cellular enzymatic and secretory activity.
PTH secretion, dietary calcium intake, renal calcium Measurement of the total serum Ca concentration alone
can be misleading because the relationship between total
A. Jain : R. Agarwal : M. J. Sankar : A. Deorari (*) : V. K. Paul and ionized Ca is not always linear. Correlation is poor
Division of Neonatology, Department of Pediatrics, when the serum albumin concentration is low or, to a lesser
All India Institute of Medical Sciences,
degree, with disturbances in acid-base status, both of which
Ansari Nagar,
New Delhi 110029, India occur frequently in premature or sick infants. With
e-mail: sdeorari@yahoo.com hypoalbuminemia, the total Ca concentration will be low
106 68
1124 Indian J Pediatr (2010) 77:11231128

while the ionized fraction will be normal unless some other diabetes mellitus causes hypomagnesemic state in the fetus.
factor is affecting Ca metabolism. More so, falsely low This hypomagnesemia induces functional hypoparathyroidism
ionic calcium levels may be recorded in alkalosis and with and hypocalcemia in the infant. A high incidence of birth
heparin use. asphyxia and prematurity in infants of diabetic mothers are
In general, the plasma calcium concentration falls by also contributing factors.
0.8 mg/dL (0.2 mmol/L) for every 1.0 g/dL fall in the
plasma albumin concentration. Perinatal asphyxia Delayed introduction of feeds, in-
Therefore, estimation of total calcium levels is a poor creased calcitonin production, increased endogenous
substitute for measuring the ionized levels. phosphate load, renal insufficiency, and diminished
parathyroid hormone secretion all may contribute to
hypocalcemia.
Definition
Maternal hyperparathyroidism This causes intrauterine
Hypocalcemia is defined as total serum calcium of less than hypercalcemia suppressing the parathyroid activity in the
7 mg/dL (1.75 mmol/L) or ionized calcium less than 4 mg/dL fetus resulting in impaired parathyroid responsiveness to
(1 mmol/L) in preterm infants and less than 8 mg/dL hypocalcaemia after birth. Hypocalcaemia may be severe
(2 mmol/L; total) or <1.2 mmol/L (ionic) in term neonates [6]. and prolonged.
The SCa concentration is usually reported in different ways
viz. mg/dL, meq/L and mmol/L The relationship between Intrauterine growth restriction (IUGR) Infants with IUGR
these units is related to the following equations: mmol=L may have hypocalcemia if they are born preterm and/or
mg=dL  10  molecular wt; meq=L mmol=L  valency. have had perinatal asphyxia. Small for gestational age is not
Since the molecular weight of calcium is 40 and the valence an independent risk factor for ENH.
is +2, 1 mg/dL is equivalent to 0.25 mmol/L and to 0.5 meq/L.
Thus, values in mg/dl may be converted to molar units Iatrogenic Any condition causing alkalosis increases the
(mmol/L) by dividing by 4. binding of the calcium with albumin and causes decrease in
ionic calcium levels

Early onset neonatal hypocalcemia (ENH) (Table 1)


Screening is recommended in at risk neonates (if facilities
This condition is fairly common and seen within the first exist)
3-4 days of life in following clinical settings:
1. Preterm infants born before 32 wks
Prematurity This may be related to premature termination 2. Infants of diabetic mothers on IV fluids
of trans-placental supply, exaggeration of the postnatal drop 3. Infants born after severe perinatal asphyxia defined as
to hypocalcemic levels, increased calcitonin and diminished Apgar score <4 at 1 min of age
target organ responsiveness to parathyroid hormone.

Infant of diabetic mother (gestational and insulin dependent) Time schedule for screening
This may be related to increased calcium demands of a
macrosomic baby [7]. Magnesium depletion in mothers with At 24 and 48 hrs of age in at risk babies.

Clinical presentation
Table 1 Causes of early onset hypocalcemia
1. Asymptomatic: ENH is usually asymptomatic unlike
Prematurity
the late onset variety and is incidentally detected.
Preeclampsia
2. Symptomatic: The symptoms may be of neuromuscular
Infant of Diabetic mother
irritability - myoclonic jerks, jitteriness, exaggerated startle,
Perinatal stress/ asphyxia
and seizures. They may represent the cardiac involvement
Maternal intake of anticonvulsants (phenobarbitone,
phenytoin sodium)
like- tachycardia, heart failure, prolonged QT interval,
Maternal hyperparathyroidism
decreased contractibility. More often they are non-specific
and not related to the severity of hypocalcemia. Apnea,
Iatrogenic (alkalosis, use of blood products,
diuretics, phototherapy, lipid infusions etc) cyanosis, tachypnoea, vomiting and laryngospasm are
other symptoms that are noted.
107 69
Indian J Pediatr (2010) 77:11231128 1125

Diagnosis Treatment of early onset hypocalcemia

1. Laboratory: Total or ionized serum calcium (total (1 ml of calcium gluconate (10%) gives 9 mg of elemental
<7 mg/dL or ionized <4.0 mg/dL). Ionized calcium is calcium)
the preferred mode for diagnosis of hypocalcemia.
2. ECG: QoTc >0.22 s or QTc >0.45 s 1. Patients at increased risk of hypocalcemia (prophylactic):
Preterm infants (32 wks), sick infants of diabetic
mothers and those with severe perinatal asphyxia should
QT interval in seconds receive 40 mg/kg/day of elemental calcium (4 mL/kg/day
QTc p
R  R interval in seconds of 10% calcium gluconate) for prevention of early onset
QoT interval in seconds hypocalcemia. However, there is not sufficient evidence
QoTc p for this practice. Infants tolerating oral feeds may receive
R  R interval in seconds
this calcium orally q 6 hourly. Therapy should be con-
(QT interval is measured from origin of q wave to end of tinued for 3 days. Oral calcium preparations have high
T wave on ECG; QoT is measured from origin of q wave to osmolality and should be avoided in babies at higher risk
origin of T wave). of necrotizing enterocolitis.
A diagnosis of hypocalcemia based only on ECG criteria 2. Patients diagnosed to have asymptomatic hypocalcemia
is likely to yield a high false positive rate. Although these (on screening): Infants detected to have hypocalcemia
parameters have good correlation with hypocalcemia in low on screening and who are otherwise asymptomatic
birth weight infants (sensitivity of 77% and specificity of should receive 80-mg/kg/day elemental calcium
94.7%) [8], neonates suspected to have hypocalcemia by (8 mL/kg/day of 10% calcium gluconate) for 48 hrs.
ECG criteria should have the diagnosis confirmed by This may be tapered to 50% dose for another 24 hrs
measurement of serum calcium levels. and then discontinued. Neonates tolerating oral feeds

Fig. 1 Management of early


Hypocalcemia
neonatal hypocalcemia Total serum Cal <7 mg/dl

Asymptomatic
80 mg/kg/day for 48 hrs
(8 mL/kg/day of 10% calcium gluconate )

Taper to 40 mg/kg/day for one day


Then stop

Symptomatic
Bolus of 2 mL/kg calcium gluconate 1:1 diluted with 5 % dextrose over 10 minutes
under cardiac monitoring

Followed by continuous infusion 80 mg/kg/day for 48 hrs


(8 mL/kg/day of 10% calcium gluconate )
Document normal calcium at 48 hrs

Then taper to 40 mg/kg/day for one day


Then stop

Prophylactic
Preterm< 32 wk, sick IDM,severe asphyxia
40 mg/kg/day for 3 days
(4ml/kg/day of 10% calcium gluconate )
IV or oral if can tolerate per oral

Treatment is for 72 hrs


Continuous infusion is better than bolus
Symptomatic babies treatment is 48 hrs continuous infusion
In case the hypocalcemia does not correct with the above by 72 hrs then investigate for
causes of late hypocalcemia.- Refer Table 2
108 70
1126 Indian J Pediatr (2010) 77:11231128

may be treated with oral calcium (IV preparation may Late onset neonatal hypocalcemia (LNH)
be used orally).
3. Patients diagnosed to have symptomatic hypocalcemia: This condition is rare as compared to ENH. It usually
These patients should receive a bolus dose of 2 mL/kg/dose presents at the end of the first wk of life. It is usually
diluted 1:1 with 5% dextrose over 10 min under cardiac symptomatic in the form of neonatal tetany or seizures.
monitoring. When there is severe hypocalcaemia with poor This is usually caused by high phosphate intake (iatrogenic).
cardiac function, calcium chloride 20 mg/kg may be given The causes are listed in Table 2.
through a central line over 10-30 min (as chloride in
comparison to gluconate does not require the metabolism Examination
by the liver for the release of free calcium). This should be
followed by a continuous IV infusion of 80 mg/kg/day Such babies should have an examination with special
elemental calcium for 48 h. Continuous infusion is emphasis on cataracts, hearing problems, and any evidence
preferred to IV bolus doses (1 mL/kg/dose q 6 hrly). of basal ganglia involvement (movement disorder).
Calcium infusion should be dropped to 50% of the original
dose for the next 24 h and then discontinued. The infusion Investigations
may be replaced with oral calcium therapy on the last day.
Normal calcium values should be documented at 48 h These should be considered in LNH or if the hypocalcemia
before weaning the infusion. does not respond to adequate doses of calcium. The work
All categories of hypocalcemia should be treated for at Table 2 Causes of Late Onset Hypocalcemia
least 72 h. Continuous infusion is preferred to IV bolus
doses. Symptomatic hypocalcemia should be treated with a Increased phosphate load
continuous infusion for at least 48 h (Fig. 1). Cow milk, renal insufficiency
Hypomagnesemia
Vitamin D deficiency
Precautions and side effects
Maternal vitamin D deficiency
Malabsorption
Bradycardia and arrhythmia are known side effects of bolus
Renal insufficiency
IV calcium administration. Hence, bolus doses of calcium
Hepatobiliary disease
should be diluted 1:1 with 5% dextrose and given slowly (over
PTH resistence
10 to 30 min) under cardiac monitoring. An umbilical venous
Transient neonatal pseudohypoparathyroidism
catheter (UVC) may be used for administration of calcium
Hypoparathyroidism
only after ensuring that the tip is positioned in the inferior vena
Primary
cava. Hepatic necrosis may occur if the tip of the UVC lies in a
branch of the portal vein. Umbilical artery catheter (UAC) Hypoplasia, aplasia of parathyroid glands - (Di Georges
syndrome), CATCH 22 syndrome (cardiac anomaly,
should never be used for giving calcium injections. Accidental abnormal facies, thymic aplasia, cleft palate, hypocalcaemia
injection into the UAC may result in arterial spasms and with deletion on chromosome 22)
intestinal necrosis. Skin and subcutaneous tissue necrosis may Activating mutations of the calcium sensing receptor (CSR)
occur due to extravasation. Secondary
Hence, IV sites where calcium is being infused should Maternal hyperparathyroidism
be checked at least q 2 hrly to monitor for extravasation and Metabolic Syndromes
avoid subcutaneous tissue necrosis. Kenny-caffey syndrome
Long-chain fatty acyl CoA dehydrogenase deficiency
Prolonged or resistant hypocalcemia Kearns-sayre syndrome
Iatrogenic
This condition should be considered in the following Citrated blood products
situations: Lipid infusions
Bicrbonate therepy
& Symptomatic hypocalcemia unresponsive to adequate
Diueretics (loop diuretics0
doses of calcium therapy
Glucocorticosteriods
& Infants needing calcium supplements beyond 72 h of age
Phosphate therepy
& Hypocalcemia presenting at the end of the first wk.
Alkalosis
These infants should be investigated for causes of LNH Phototherapy
(see below) Table 3.
109 71
Indian J Pediatr (2010) 77:11231128 1127

up of such a case is very important to determine the 3. Hypoparathyroidism [9] These infants tend to be
etiology. The same can be planned as per the Table 3. hyperphosphatemic and hypocalcemic with normal
If hypocalcemia is present with hyperphosphatemia and renal function. Elevated phosphate levels in the
a normal renal function, hypoparathyroidism should be absence of exogenous phosphate load (cows milk)
strongly suspected and presence of normal renal functions indicates
parathormone inefficiency. It is important to realize
Treatment of LNH that if the phosphate level is very high, then adding
calcium will lead to calcium deposition and tissue
The treatment of LNH is specific to etiology and may in damage. Thus, attempts should be made to reduce the
certain diseases be life-long. phosphate (so as to keep the calcium and the
phosphate product less than 55) [10]. These neonates
1. Hypomagnesemia: Symptomatic hypocalcemia unre- need supplementation with calcium (50 mg/kg/day in 3
sponsive to adequate doses of IV calcium therapy is divided doses) and 1,25(OH)2 Vitamin D3 (0.5-1 g/day).
usually due to hypomagnesemia. It may present either as Syrups with 125 mg and 250 mg per 5 ml of calcium are
ENH or later as LNH. The neonate should receive 2 doses available.1,25(OH)2 vitamin D3 (calcitriol) is available as
of 0.2 mL/kg of 50% MgSO4 injection, 12 h apart, deep 0.25 g capsules. Therapy may be stopped in hypocal-
IM followed by a maintenance dose of 0.2 mL/kg/day of cemia secondary to maternal hyperparathyroidism after
50% MgSO4, PO for 3 days. 6 wks.
2. High phosphate load: These infants have hyperphos- 4. Vitamin D deficiency states: These babies have
phatemia with near normal calcium levels. Exclusive hypocalcemia associated with hypophosphatemia due
breast-feeding should be encouraged and top feeding to an intact parathormone response on the kidneys.
with cows milk should be discontinued. Phosphate They benefit from Vitamin D3 supplementation in a
binding gels should be avoided. dose of 30-60 ng/kg/day

Table 3 Investigations required in infants with persistent / late onset hypocalcemia

Investigations required

First line Second line Others


Serum phosphate Serum magnesium CT brain for calcification
Serum alkaline phosphatase (SAP) Serum parathormone levels (PTH) Echocardiography
Liver function tests Urine calcium creatinine ratio Vitamin D levels (1,25 D3)
Renal function tests Maternal calcium, phosphate, and Hearing evaluation
alkaline phosphatase
X ray chest/ wrist Serum cortisol
Arterial pH Thyroid function tests
S No Disorder causing hypocalcaemia Findings
1 Hypoparathyroidism High : Phosphate
Low : SAP, PTH, 1,25 D3
2 Pseudo Hypoparathyroidim High : SAP, PTH, Phosphate
Low : 1,25 D3
3 Chronic renal failure High : phosphate, SAP, PTH, pH
(acidotic), deranged RFT
Low : 1,25 D3
4 Hypomagnesemia High : PTH
Low : Phosphate, Mg,1,25 D3
5 VDDR1 High : SAP, PTH
Low : Phosphate, 1,25 D3
6 VDDR II High : SAP, 1, 25 D3, PTH
Low : Phosphate

(VDDR; vitamin D dependent rickets)


110
1128 Indian J Pediatr (2010) 77:11231128

Monitoring 2. Linarelli LG, Bobik J, Bobik C. Newborn urinary cyclic AMP and
developmental responsiveness to parathyroid harmone. Pediatrics.
1972;50:1423.
The baby is monitored for the SCa, and phosphate, 24 h 3. Hillman L, Rajanasathit S, Slatopolsky E, Haddad JG. Serial
urinary calcium, and calcium creatinine ratio. Try to keep the measurements of serum calcium, magnesium, parathyroid hor-
calcium in the lower range as defective distal tubular mone, calcitonin, and 25-hydroxy-vitamin D in premature and
term infants during the first week of life. Pediatr Res.
absorption leads to hypercalciuria and nephrocalcinosis [11].
1977;11:789844.
4. Salle BL, Delvin EE, Lapillonne A, Bishop NJ, Glorieux FH.
Prognosis and outcome Perinatal metabolism of vitamin D. Am J Clin Nutr. 2000;71:1317S
24S.
5. Singh J, Moghal N, Pearce SH, Cheetham T. The investigation of
Most cases of early neonatal hypocalcemia resolve within hypocalcaemia and rickets. Arch Dis Child. 2003;88:4037.
48-72 h without any clinically significant sequelae. 6. Oden J, Bourgeois M. Neonatal endocrinology. Indian J Pediatr.
Late neonatal hypocalcemia secondary to exogenous 2000;67:21723.
phosphate load and magnesium deficiency also responds 7. Schwartz R, Teramo KA. Effects of diabetic pregnancy on the
fetus and newborn. Semin Perinatol. 2000;24:12035.
well to phosphate restriction and magnesium repletion.
8. Nekvasil R, Stejskal J, Tuma A. Detection of early onset neonatal
When caused by hypoparathyroidism, hypocalcemia hypocalcemia in low birth weight infants by Q-Tc and Q-oTc
requires continued therapy with vitamin D metabolites interval measurement. Acta Paediatr Acad Sci Hung.
and calcium salts. The period of therapy depends on the 1980;21:20310.
9. Marx SJ. Hyperparathyroid and hypoparathyroid disorders. N
nature of the hypoparathyroidism which can be transient,
Engl J Med. 2000;343:186375.
last several wks to months, or be permanent. 10. Sharma J, Bajpai A, Kabra M, Menon PSN. Hypocalcemia
clinical, biochemical, radiological profile and follow-up in a
tertiary hospital in India. Indian Pediatr. 2002;39:27682.
11. Rigo J, Curtis MD. Disorders of calcium, phosphorus and
References magnesium metabolism. In: Martin RJ, Fanaroff AA, Walsh
MC, editors. Neonatal Perinatal Medicine- Diseases of the
1. Schauberger CW, Pitkin RM. Maternal-perinatal calcium relation- fetus and infant. 8th ed. Pihladelphia: Elsevier; 2006. p. 1508
ships. Obstet Gynecol. 1979;53:746. 14.
111
112
113
114
115 72
Indian J Pediatr (2010) 77:11371142
DOI 10.1007/s12098-010-0175-1

SYMPOSIUM ON AIIMS PROTOCOLS IN NEONATOLOGY-I

Hypoglycemia in the Newborn


Ashish Jain & Rajiv Aggarwal & M. Jeeva Sankar &
Ramesh Agarwal & Ashok K. Deorari & Vinod K. Paul

Received: 27 July 2010 / Accepted: 2 August 2010 / Published online: 7 September 2010
# Dr. K C Chaudhuri Foundation 2010

Abstract Hypoglycemia in a neonate is defined as blood Hypoglycemia is a common disorder.in neonates [1].
sugar value below 40 mg/dL. It is commonly associated There is, however, no universal definition for this
with a variety of neonatal conditions like prematurity, condition [2]. Currently there is insufficient evidence with
intrauterine growth restriction and maternal diabetes. respect to what constitutes hypoglycemia. The issue is
Screening for hypoglycemia in high-risk situations is further complicated by the fact that low blood glucose
recommended. Supervised breast-feeding may be an initial levels (BGLs) are associated with increased cerebral blood
treatment option in asymptomatic hypoglycemia. However, flow as a compensatory mechanism. Brain is able to utilize
symptomatic hypoglycemia should always be treated with alternative energy substrates to maintain its cerebral
a continuous infusion of parenteral dextrose. Neonates energy milieu. Various investigators have empirically
needing dextrose infusion rates above 12 mg/kg/min should recommended different blood glucose levels that should
be investigated for a definite cause of hypoglycemia. be maintained in neonatal period to prevent injury to the
Hypoglycemia has been linked to poor neuro-developmental developing brain [3, 4]. The normal range of blood
outcome, and hence aggressive screening and treatment is glucose is variable and depends upon factors like birth-
recommended. weight, gestational age, body stores, feeding status,
availability of energy sources as well as the presence or
Keywords Hypoglycemia . Screening . Newborn . Therapy absence of disease [5, 6]. Thus, the definition of
hypoglycemia should be flexible and encompass all these
aspects. Further, there is no concrete evidence to show the
causation of adverse long-term outcomes by a particular
level or duration of hypoglycemia [7]. Hence, a consensus
A. Jain has been to evolve an operational threshold.
Department of Pediatrics, Hindu Rao Hospital,
New Delhi, India

R. Aggarwal
Definition
Department of Pediatrics, Narayana Hrudayalaya,
Bangalore, India The operational threshold for hypoglycemia is defined as
that concentration of plasma or whole blood glucose at
R. Aggarwal
Division of Neonatology, Department of Pediatrics, AIIMS,
which clinicians should consider intervention, based on
New Delhi, India the evidence currently available in literature [8]. This
so-called operational threshold values are useful guide-
M. Jeeva Sankar : R. Agarwal (*) : A. K. Deorari : V. K. Paul lines for clinicians to take appropriate actions. Till proper
Department of Pediatrics, All India Institute of Medical Sciences,
Ansari Nagar,
evidence is generated, this value is currently believed to
New Delhi 110029, India be a blood glucose value of less than 40 mg/dL (plasma
e-mail: aranag@rediffmail.com glucose less than 45 mg/dL) [9].
116 73
1138 Indian J Pediatr (2010) 77:11371142

Screening for Hypoglycemia When Should be Screening is Stopped?

Normal blood glucose levels are maintained by glyco- & At risk infants (Table 1): At the end of 72 h.
genolysis and by gluconeogenesis from a variety of non- & In an infant on IV fluids: Has two consecutive values
carbohydrate energy sources. Neonatal hypoglycemia >50 mg/dL on total oral feeds after stopping of the IV fluids.
often occurs in infants with impaired gluconeogenesis, & Infant whose blood sugar normalized on oral feed:
brought about by increased insulin production, altered Consider at risk and monitor for 48 h
counter-regulatory hormone production or an inadequate
substrate supply.
Screening for hypoglycemia is recommended in high
Infants in Whom Screening is not Required
risk infants (Table 1).
Screening for hypoglycemia is not recommended in term
healthy breast-fed appropriate-for-gestational age (AGA)
Time Schedule for Screening
infants. However, term infants with poor feeding, presence
of inadequate lactation or presence of cold stress may be
There is a paucity of the literature that looks into optimal
considered for screening.
timing and the intervals of glucose monitoring. Lowest blood
sugar values are seen at 2 h of life. IDMs frequently
experience asymptomatic hypoglycemia very early viz. 1 to
2 h and rarely beyond 12 h (range 0.8 to 8.5 h), supporting Method of Glucose Estimation
early screening for this population [11]. However, preterm
and SGA may be at risk up to 36 h (range 0.8 to 34.2 h) [12]. a. Bed side reagent strips (Glucose oxidase): Though
Some SGA and preterm infants may develop hypoglycemia widely used and an important point of care method,
when feeding is not established. Based on these assumptions glucose estimation by this method is unreliable, especially
and current knowledge, Table 2 elaborates the schedule and at levels where therapeutic intervention is required such as
frequency of monitoring in different situations. BGL 4050 mg/dL. They are useful for screening purpose
but low values should be always confirmed by formal
laboratory analysis. However, treatment may be initiated
Education and Counseling of Caregivers Regarding based on the results of the reagent strips. It is important to
the Screening also consider the variations between capillary and venous,
blood and plasma, and immediate and stored samples
Parents should be made aware that their infant is at-risk (whole blood sugar is 1015% less than the plasma sugar,
and therefore requires blood tests at regular intervals. the glucose levels can fall by 1418 mg/dL per hr in blood
This will ensure appropriate parental participation in samples that await the analysis [13].) Arterial samples
monitoring and allay fears if further interventions are have slightly higher value compared to venous and
required. capillary samples.

Table 1 High risk situations where screening is recommended [10]

1 Low birth weight infants (<2000 gm)


2 Preterm infants (35 wks)
3 Small for gestational age infants (SGA) : birth weight <10th percentile
4 Infant of diabetic mothers (IDM) - insulin dependent and gestational diabetes
5 Large for gestational age (LGA) infants: birth weight >90th percentilea
6 Infants with Rh-hemolytic disease
7 Infants born to mothers receiving therapy with terbutaline/propranolol/lebatolol/oral hypoglycemic agents
8 Infants with morphological IUGR. This group includes neonates with birth weight between 10th25th and
possibly up to 50th percentile with features of fetal under-nutrition such as three or more loose skin folds in gluteal region,
decreased overall subcutaneous fat, and head circumference to chest circumference difference greater than 3 cm
9 Any sick neonate such as those with perinatal asphyxia, polycythemia, sepsis, shock etc,
when they are in active phase of illness. The screening may be discontinued once their condition gets stabilized.
10 Infants on total parenteral nutrition
a
LGA infants because of constitutional reasons such as infants of constitutionally large parents may also be exempted from routine screening
117 74
Indian J Pediatr (2010) 77:11371142 1139

Table 2 Schedule of blood glucose monitoring

Symptomatology of infants Time schedule for screening

At risk neonates (S. No 1-8 in Table 1) 2, 6, 12, 24, 48, and 72 hrs
Sick infants Sepsis, asphyxia, shock in the active phase of illness Every 68 hrs (individualize as needed)
Stable VLBW infants on parenteral nutrition Initial 72 hrs: every 6 to 8 hrs; after 72 hrs in stable babies: once a day

Infants exhibiting signs compatible with hypoglycemia at any time also need to be investigated

The first generation strips focused on change in color of cyanosis, convulsions, intermittent apneic spells or
of enzyme on application of blood drop. The color can tachypnea, weak and high pitched cry, limpness and
be read by naked eye or more recently by reflectance lethargy, difficulty in feeding, and eye rolling. Episodes of
meters. The readings tend to get affected by hematocrit sweating, sudden pallor, hypothermia and cardiac arrest
values, acidosis, presence of bilirubin, presence of edema have also been reported.
etc. The newer generation glucose reagent strips generate
a current on reaction of glucose with enzymes such
glucose oxidase or glucose dehydrogenase. The amount
Diagnosis
of current is proportional to amount of sugar present in
plasma. Though these second generation glucose readers
a. Asymptomatic hypoglycemia: It is said to be present
are more accurate than the previous version, they are still
when the blood glucose level is less than 40 mg/dl (to
not entirely reliable. Any abnormal BGLs by this
be confirmed by laboratory estimation) and the infant
technique must be confirmed by standard laboratory
does not manifest any clinical features
methods.
b. Symptomatic hypoglycemia: This diagnosis should be
b. Laboratory diagnosis: This is the most accurate made if hypoglycemia coexists with clinical symp-
method. In the laboratory (lab), glucose can be tomatology. Neonates generally present with nonspe-
measured by either the glucose oxidase (calorimetric) cific signs that result from a variety of illnesses.
method or by the glucose electrode method (as used in Therefore, careful evaluation should be done to look
blood gas and electrolyte analyzer machine). Blood for all possible causes especially those that can be
samples should be analyzed quickly to avoid errone- attributed to hypoglycemia.
ously low glucose levels.
If clinical signs attributable to hypoglycemia persist
despite intravenous glucose, then other causes of persistent /
resistant hypoglycemia should be explored.
Clinical Signs Associated with Hypoglycemia

a. Asymptomatic: It is well known that low BGL may not Management of Asymptomatic Hypoglycemia
manifest clinically and be totally asymptomatic. These
infants should also be treated in view of the possible Table 3
adverse long term effects [14, 15]. However, there is
considerable controversy with regards to if asymptom- Oral FeedsIssues
atic hypoglycemia results in a neuronal damage.
b. Symptomatic: Clinical signs of hypoglycemia in order of Direct breast-feeding is the best option for trial of an oral
frequency are stupor, jitteriness, tremors, apathy, episodes feed. If the infant is unable to suck, expressed breast milk

Table 3 Management plan of infants with asymptomatic hypoglycemia on screening

Blood sugar 2040 mg/dL Trial of oral feeds (expressed breast milk or formula) and repeat blood test after 1 hr. If repeat blood
sugar is more than 50 mg/dL, two hrly feeds is ensured with 6 hrly monitoring for 48 hrs
If repeat blood sugar is <40 mg/dL, IV Dextrose is started and further management is
as for symptomatic hypoglycemia
Blood sugar levels <20 mg/dL IV Dextrose is started at 6 mg/kg/min of glucose;further management is as for symptomatic hypoglycemia
118 75
1140 Indian J Pediatr (2010) 77:11371142

may be used. Breast milk promotes ketogenesis (ketoacids Do not stop an IV infusion of glucose abruptly; severe
are important alternate sources for the brain along with less rebound hypoglycemia may occur. Avoid using >12.5%
important pyruvate, free fatty acids, glycerol, and amino dextrose infusion through a peripheral vein due to the risk
acids). If breast milk is not available, then formula feeds of thrombophlebitis.
may be given in at-risk neonates. If oral feeds are contra-
indicated, start glucose infusion.
Some of the randomized clinical trials in SGA [16] and Recurrent / Resistant Hypoglycemia
appropriate-for-gestational age [17] infants found that the
sugar or sucrose fortified milk (5 g sugar per 100 mL milk) This condition should be considered when there is a
raises blood glucose and prevents hypoglycemia. Such failure to maintain normal blood sugar levels despite a
supplementation may be tried in the asymptomatic neonates glucose infusion of 12 mg/kg/min or when stabilization
with blood sugar levels between 20 to 40 mg/dL. However, is not achieved by 7 days of therapy. High levels of
this practice carries a potential to compromise breast feeding glucose infusion may be needed in the infants to achieve
rates, and therefore one should be prudent in exercising this euglycemia.
option. Besides increasing the rate of glucose infusion, drugs
All symptomatic infants should be treated with IV fluids. may also be tried in the treatment of resistant hypoglyce-
mia. Before administration of the drugs, take the samples to
investigate the cause (Table 4). Drugs that are used include
Management of Symptomatic Hypoglycemia the following:
(1) Hydrocortisone 5 mg/kg/day IV or PO in two divided
For symptomatic hypoglycemia including seizures, a bolus
doses for 24 to 48 h
of 2 mL/kg of 10% dextrose (200 mg/kg) should be given.
(2) Diazoxide 1025 mg/kg/day in three divided doses
This mini-bolus helps to rapidly achieve the steady state
PO. Diazoxide acts by keeping the KATP channels of
levels of blood glucose [15]. Immediately after the bolus, a
the -cells of the pancreas open, thereby reducing the
glucose infusion at an initial rate of 68 mg/kg/min should
secretion of insulin. It is therefore useful in states of
be started. Check blood sugar after 30 to 60 min and then
unregulated insulin secretion like in insulinomas.
every 6 h until blood sugar is >50 mg/dL.
(3) Glucagon 100 g/kg subcutaneous or intramuscular
Repeat subsequent hypoglycemic episodes may be treated
(max 300 g)maximum of three doses. Glucagon acts
by increasing the glucose infusion rate by 2 mg/kg/min until
by mobilizing hepatic glycogen stores, enhancing
a maximum of 12 mg/kg/min. After 24 h of IV glucose
gluconeogenesis and promoting ketogenesis. These
therapy, once two or more consecutive blood glucose values
effects are not consistently seen in small-for-gestational
are >50 mg/dL, the infusion can be tapered off at the rate of
age infants. Side effects of glucagon include vomiting,
2 mg/kg/min every 6 h with BGL monitoring. Tapering has
diarrhea and hypokalemia and at high doses it may
to be accompanied by concomitant increase in oral feeds.
stimulate insulin release.
Once a rate of 4 mg/kg/min of glucose infusion is reached
(4) Octreotide (synthetic somatostatin in dose of 210 g/kg/
and oral intake is adequate and the blood sugar values are
day subcutaneously two to three times a day.
consistently >50 mg/dL, the infusion can be stopped without
further tapering. Ensure continuous glucose infusion without Do not use diazoxide and glucagon in small for
any interruption preferably using infusion pump. gestational age infants.

Table 4 Important causes of


resistant hypoglycemia and Important causes of resistant hypoglycemia Investigations to be considered
investigations
Congenital hypopitutarism Serum insulin levels
Adrenal insufficiency Serum cortisol levels
Hyperinsulinemic states Growth hormone levels
Galactosemia Blood ammonia
Glycogen storage disorders Blood lactate levels
Maple syrup urine disease Urine ketones and reducing substances
Mitochondrial disorders Urine and sugar aminoacidogram
Fatty acid oxidation defect Free fatty acid levels
Galactose 1 phosphate uridyl transferase levels
119 76
Indian J Pediatr (2010) 77:11371142 1141

Useful Formulae similar relationship of lower head circumference and


developmental scores was highlighted by Duvanel et al.
a Infusion rate % of dextrose being infused  rate mL=hr [18] Further, a systematic review of 18 studies on neuro-

mg=kg=min body weight in kg  6 development after hypoglycemia showed poor methodo-
b Infusion rate IV rate mL=kg=day  % of dextrose logical quality of all but two studies. None of the studies

mg=kg=min 144 provided a valid estimate of the effect of neonatal
c Infusion rate mg=kg=min Fluid rate mL=kg=day hypoglycemia on neurodevelopment [19]. Though these
 0:007 x % of dextrose infused studies have major limitations, it would seem prudent to
follow up all infants who had confirmed hypoglycemia in the
high-risk category, till a future optimal study is performed
Follow-up and Outcome [19]. The outcome of hypoglycemia is determined by
factors like, duration, degree of hypoglycemia, rate of
Lucas in 1988 linked hypoglycemia to long term adverse cerebral blood flow, and cerebral utilization of glucose.
outcomes in a retrospective multicentric study. Later, a Special attention should be paid to neuro-developmental

Fig. 1 Algorithm for manage-


Hypoglycemia
ment of neonatal hypoglycemia Blood sugar < 40 mg/dL

Symptomatic including
Asymptomatic seizures

Bolus of 2 mL/kg 10%


20-40 mg/dL <20 mg/dL glucose

IV glucose infusion @ 6 mg/kg/min


Monitor hourly till euglycemic and
Trial of oral / fortified feeds
then 6 hrly
(5g/100mL of sugar)

Blood sugar >50 mg/dL Blood sugar >50 mg/dL


Monitor the blood sugar after 1 hr

Stable for 24 hrs on IV fluids; 2


glucose @ 2
values of blood sugar >50 mg/dL mg/kg/min till
euglycemia
> 40 mg/dL < 40 mg/dL

Weaning at 2 mg/kg/min every


6 hrs; Increase till the glucose
Frequent feeds oral feeds; infusion rate is >12
Monitoring to continue 6 hrly mg/kg/min

Stop IV fluids when


Monitor blood sugar the rate is 4 mg/kg/min Send
and the infant is stable Serum insulin. cortisol ,
and growth hormone
Stop after 48 hrs levels
Blood ammonia
Stop monitoring when 2
Blood lactate levels
values are more than 50 on
Before discharge ensure that Free fatty acid levels
full oral feeds
there is no feeding difficulty Urine ketones and
reducing substances
Urine aminoacidogram
Before discharge ensure
that there is no feeding
difficulty Hydrocortisone
Diazoxide (not in SGA)
Glucagon (not in SGA)
Octreotide

Refer to Specialist center


Pancreatic diagnosis
PET Scan, Pancreatic Biopsy
120
1142 Indian J Pediatr (2010) 77:11371142

outcome, overall IQ, reading ability, arithmetic proficiency 7. Rozance PJ, Hay Jr WW. Hypoglycemia in newborn infants:
features associated with adverse outcomes. Biol Neonate.
and motor performance (Fig. 1).
2006;90:7486.
The infants can be assessed at 1 month corrected age 8. Cornblath M, Schwartz R. Outcome of neonatal hypoglycemia. Br
for vision / eye evaluation. At 3, 6, 9, 12 and 18 months Med J. 1999;318:194.
corrected age they can be followed up for growth, 9. Hay Jr WW, Raju TNK, Higgins RD, et al. Knowledge gaps and
research needs for understanding and treating neonatal hypogly-
neurodevelopment, vision and hearing loss. Vision can
cemia: workshop report from Eunice Kennedy Shriver National
be assessed with Teller acuity card and hearing should be Institute of Child Health and Human Development. J Pediatr.
assessed by Brainstem evoked auditory responses. Neu- 2009;155:6127.
rodevelopment will be assessed by the clinical psychol- 10. Kalhan S, Parimi P. Gluconeogenesis in the fetus and neonate.
Semin Perinatal. 2000;24:94106.
ogist using DASII 2. MRI at 46 weeks provides a good 11. Srinivasan G, Pilades RS, Cattamanchi G, et al. Plasma glucose
estimate of hypoglycemic injury and therefore should values in normal neonates: a new look. J Pediatr. 1986;109:1147.
be considered in follow up of such infants subject to 12. Holtrop PC. The frequency of hypoglycemia in full term and small
affordability. for gestational age newborns. Am J Perinatol. 1993;10:1504.
13. Cowett RM, Damico LB. Capillary (heelstick) versus venous
blood sampling for determination of glucose concentration in
neonate. Biol Neonate. 1992;62:326.
14. Lucas A, Morley R. Outcome of neonatal hypoglycemia. Br Med
References J. 1999;318:194.
15. Filan PM, Inder TE, Cameron FJ, et al. Neonatal hypoglycemia
and occipital cerebral injury. J Pediatr. 2006;148:5525.
1. Cornblath M. Neonatal hypoglycemia 30 years later: does it injure 16. Singhal PK, Singh M, Paul VK. Prevention of hypoglycemia: a
the brain? Historical summary and present challenges. Acta controlled evaluation of sugar fortified milk feeding in small- for-
Paediatr Jpn. 1997;39:S7S11. date infants. Indian Pediatr. 1992;29:13659.
2. Cornblath M, Hawdon JM, Williams AF, et al. Controversies 17. Singhal PK, Singh M, Paul VK, Malhotra AK, Deorari AK,
regarding definition of neonatal hypoglycemia: suggested opera- Ghorpade MD. A controlled study of sugar fortified milk feeding
tional thresholds. Pediatrics. 2000;105:11415. in prevention of neonatal hypoglycemia. Indian J Med Res.
3. Termote B, Verswijvel G, Gelin G, Palmers Y. Neonatal 1991;94:3425.
hypoglycemic brain injury. JBR-BTR. 2008;91:1167. 18. Duvanel CB, Fawer CL, Cotting J, Hothfield P, Matthieu JM.
4. Inder T. How low can I go? The impact of hypoglycemia on the Long term effects of neonatal hypoglycemia on brain growth and
immature brain. Pediatrics. 2008;122:4401. psychomotor development in small-for-gestational age preterm
5. Mitanchez D. Glucose regulation in preterm newborn infants. infants. J Pediatr. 1999;134:4928.
Horm Res. 2007;68:26571. 19. Boluyt N, van Kempen A. Martin offringa. Neurodevelopment
6. Cornblath M, Ichord R. Hypoglycemia in the neonate. Semin after neonatal hypoglycemia: a systematic review and design of an
Perinatol. 2000;24:13649. optimal future study. Pediatrics. 2006;117:223143.
121
122
123
124
125 77

Symposium on AIIMS Protocols in Neonatology II

Management of Infants with Intra-uterine Growth


Restriction
Ashok K. Deorari, Ramesh Agarwal and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, India

ABSTRACT
Intra-uterine growth restriction (IUGR) contributes to almost two-thirds of LBW infants born in India. Poor nutritional status and
frequent pregnancies are common pre-disposing conditions in addition to obstetric and medical problems during pregnancy.
Growth restriction may be symmetrical or asymmetrical depending on the time of insult during pregnancy. The pathological
insult in an asymmetrical IUGR occurs during the later part of the pregnancy and has a brain-sparing effect. Common
morbidities are more frequent in <3rd percentile group as compared to 3rd10th percentile group. Guidelines for management
of IUGR neonates in these two groups have been provided in the protocol. [Indian J Pediatr 2008; 75 (2) : 171-174] E-mail:
sdeorari@yahoo.com

Key words : IUGR; LBW; Postnatal management

Nearly one third of neonates born in India are low birth the terminology. SGA a statistical definition, is used for
weight (LBW), weighing less than 2500 grams at birth. A neonates whose birth weight is lower than (less than 10th
babys low birth weight is either the result of preterm percentile for that particular gestational age) population
birth (before 37 completed weeks of gestation) or due to norms. IUGR is a clinical definition and includes neonates
intrauterine growth restriction (IUGR). Later condition is with clinical evidence of malnutrition. This may be in the
akin to malnutrition and may be present in both term and form of loose skin folds on the face and in the gluteal
preterm infants. Neonates affected by IUGR are usually region, absence of subcutaneous fat and peeling of skin.
undernourished, undersized and therefore, low birth Although most IUGR infants would also be SGA, it is
weight. Two-third LBW neonates born in India fall in possible that a small minority of IUGR infants may have
this category.1 Since, IUGR neonates are more likely to birth weights above the 10 th percentile. These
suffer complications including cold stress and morphological IUGR infants would behave like SGA
hypoglycemia, it is important that these infants are infants and should be managed along the same lines as
identified and managed appropriately at birth.2 Even after SGA infants. For purposes of discussion in this paper, the
recovering from neonatal complications, they remain term IUGR would include both the groups of infants .
more prone to poor physical growth, poor
neurodevelopmental outcome, recurrent infection and
chronic diseases (hypertension, hyperlipidemia, diabetes ETIOLOGY
mellitus, coronary heart disease) later in life3.

Poor nutritional status of the mother and frequent


IUGR AND SGA (SMALL-FOR-GESTATIONAL AGE) pregnancies are the major cause of IUGR. Mothers with a
weight of less than 40 Kg and a height of less than 145 cm
often give birth to SGA infants. Insufficient nutritional
Although both the terms are used inter-changeably and
intake during pregnancy also has an adverse effect on
both denote malnutrition, there is a minor difference in
fetal weight. Maternal hypertension, pre-eclampsia, post-
maturity, frequent pregnancies, multiple pregnancy,
anemia, malaria and tobacco use are other causes of
Correspondence and Reprint requests : Dr Ashok K. Deorari, IUGR. 4-6 Chronic maternal diseases of heart, kidneys,
Professor, Department of Pediatrics, All India Institute of Medical lungs or liver may also lead to IUGR.
Sciences, Ansari Nagar, New Delhi 110029, India.
[Received January 11, 2008; Accepted January 11, 2008]

Indian Journal of Pediatrics, Volume 75February, 2008 171


126 78

Ashok K. Deorari et al

TYPES OF IUGR TABLE 1. Common Morbidities in SGA Neonates

Period: Jan 1999 to Period: Aug 04 to


Infants with IUGR are often classified as having Dec 00 (n=156) Jul 05 (n=144)
symmetrical (head circumference, length and weight
Weight <3rd Weight 3rd-10th Weight <10th
equally affected) or asymmetrical (with relative head
percentile percentile (n=109)
growth sparing) growth restriction. Infants with (n=47) percentile (n=144)
symmetric IUGR often have an earlier onset and are
associated with causes that affect total fetal cell number Birth asphyxia
including chromosomal, genetic, teratogenic, intra-uterine Total 4(8.5%) 10 (9.2%) 25 (17%)
Moderate 2 8
infections and severe hypertensive etiologies. Asymmetric Severe 2 2 -
IUGR is often of a later onset, demonstrates preservation Hypoglycemia
of blood flow to brain and is associated with poor Total 12 (25.5%) 14 (12.8%) 24 (17%)
maternal nutrition or late onset exacerbation of maternal Symptomatic 6 3 3
vascular disease (pre-eclampsia, chronic hypertension).7 Asymptomatic 6 11 21
Polycythemia
Total 14 (29.8%) 17 (15.6%) 14 (10%)
CLINICAL FEATURES Symptomatic 3 8 3
Asymptomatic 11 9 11
Hypothermia 0 4 (3.7%) (13.2%)
IUGR or SGA infants are often term or near-term in *Includes 1 baby with hypothermia and 18 babies with cold stress
gestation. Their birth weight usually falls below the 10th
percentile.8 The neonate has an emaciated look and loose and only 7% at 24 h of life. No cases of polycythemia were
skin because of lack of subcutaneous tissue. These are detected at 48 and 72 h of life.
particularly prominent over the buttocks and the thighs.
They look alert and are often plethoric. Comparison of the
head circumference with chest circumference is helpful in MANAGEMENT
the identification of a SGA infant. In infants with
appropriate growth, the head size is usually bigger than
the chest by about 2-cm. In SGA infants, the head Early delivery is indicated if there is arrest of fetal growth
circumference usually exceeds the chest circumference by and pulmonary maturity is satisfactory. Fetal hypoxia
more than 3 cm. A preterm SGA infant would have a may necessitate emergency cesarean section and one
combination of clinical features suggestive of both, should be prepared to receive an asphyxiated infant. If
prematurity and IUGR.9 liquor is meconium stained and the neonate is depressed,
endotracheal suctioning is essential.10 Infant should be
screened for any congenital malformations. Based on
PROBLEMS OF SGA INFANTS initial assessment, decision is taken to either keep the
infant in nursery or with mother (Fig. 1).
Common neonatal morbidities encountered in SGA Birth weight 3 rd 10 th percentile : In the absence of
infants born in our hospital are given in Table 1. The complications including perinatal asphyxia and
common morbidities encountered in IUGR neonates respiratory distress, these neonates may be managed with
include: (a) perinatal asphyxia, (b) hypothermia, (c) the mother (Table 2). Skin-to-skin care helps in
hypoglycemia and (d) polycythemia. These morbidities maintaining temperature and facilitates breast-feeding.
are commoner in the more severely growth restricted Early initiation of breastfeeding and/ or assisted feeding
babies (<3rd percentile) as compared to babies in the 3rd to helps in averting hypoglycemia. Term SGA infants
10th percentile category. usually do not pose any serious difficulties because they
have no problems in direct breast-feeding. To avoid
From August 2004 to July 2005, 144 SGA babies were
hypoglycemia, they should be put to breast within one
born in our hospital. 24 (17%) developed hypoglycemia
hour of birth. However, these infants are at risk of
and 14 (10%) had polycythemia requiring partial
morbidities and should be monitored regularly for
exchange transfusion. Amongst 24 babies with
hypoglycemia and polycythemia in the first 48-72 hours.
hypoglycemia 50% of the total episodes occurred at 2 h,
22% at 48 h, 11% each at 6 and 12 h and only 4% each at 24 Neonates with asymptomatic hypoglycemia should be
and 72 h of age. 12 (50%) had multiple episodes of supplemented with sugar fortified formula feeds. This
hypoglycemia. 3 babies were symptomatic and required may be given with the help of a cup and spoon/ paladai.
intravenous fluid therapy. Rest were managed with Neonates with normoglycemia on regular feeds should be
supplementary oral feeds. Of 14 babies with gradually weaned to exclusive breast-feeding within the
polycythemia, only 3 were symptomatic. Polycythemia next 3-4 days. Failure to maintain normoglycemia despite
was detected at 2 h in 50%, at 6 h in 29%, at 12 h in 14% regular oral feeds should be treated with IV fluids.

172 Indian Journal of Pediatrics, Volume 75February, 2008


127 79

Management of Infants with Intra-uterine Growth Restriction

TABLE 2. Management of SGA Infants neonatal intensive care unit.11


Criteria for admission to Nursery Birth weight <3 rd percentile, gestation <35 weeks :
All SGA infants < 2 SD (3rd percentile) Neonates with severe growth restriction (<3rd percentile)
- Infants with gestational age < 35 wks
or with presence of complications should be managed in
- Infants with birth asphyxia, respiratory distress etc.
Care of SGA infants with mothers (birth weight between 3rd and the intensive care unit (Table 2). This group would
10th percentile, gestation >35 wks) include infants with perinatal asphyxia, symptomatic
Early initiation of breast feeding (within 1 hour) hypoglycemia, symptomatic polycythemia, prematurity
Skin-to-skin care to maintain temperature, monitoring of (<35 weeks), respiratory distress and hypothermia. They
cold stress by mother and health professionals.
should be monitored for hypoglycemia, polycythemia
Monitor blood sugar, hematocrit
Prevent infections and feed intolerance in the initial few days.
Care of SGA infants in Nursery (birth weight <3rd percentile or
Infants with gestation <30 weeks (birth weight <1200
gestation <35 wks)
Nurse in thermo neutral environment grams) should be started on IV fluids initially and
It stable, early initiation of feeds ( EBM). gradually weaned to oral feeds over the next few days. In
Feed by orogastric tube or katori-spoon /paladai if gestation the absence of other complications, oro-gastric feeds
>32 wks should be started for neonates >30 weeks (>1200 grams)
Initial intravenous fluids followed by orogastric or katori- and gradually shifted to katori-spoon/ paladai feeding. An
spoon /paladai if gestation <32 wks
Monitor blood sugar, hematocrit
infant on full oral feeds with spoon-feeding may be tried
Care of SGA infants with absent or reversed en-diastolic blood on direct breast-feeding. These high-risk infants need to
flow be observed for a minimum of 72 hours for hypoglycemia.
At higher risk of development of NEC Infants on full katori-spoon feeding and/ or breast-
If preterm (gestation <32 weeks): Nil per oral or on minimal feeding may be shifted to the mother after 72 hours if she
enteral nutrition for first 48-72 h of life followed by gradual
is confident of on going-care.
advancement of feed volume
Paladai/spoon feeding : Feeding with a spoon (or a
Neonates with symptomatic hypoglycemia should be
similar device such as paladai) and katori (or any other
shifted to a special care nursery and managed
receptacle such as cup) has been found to be safe in SGA
appropriately with a glucose bolus followed by a
infants.8 This mode of feeding is a bridge between gavage
continuous glucose infusion at 6-8 mg/Kg/min.
feeding and direct breast-feeding. It is based on the
Neonates with asymptomatic polycythemia and a premise that neonates with a gestation of 30-32 weeks or
hematocrit <75 maybe managed conservatively by more are in a position to swallow the feeds satisfactorily
increasing fluid intake. The infant should receive regular even though they may not be good at sucking or
(2-3 hourly) breast feeds with extra supplementation. coordinated sucking and swallowing. A medium sized
Infants with symptomatic polycythemia or hematocrit >75 katori and a small (1-2 ml size) spoon should be used. The
should be managed by partial exchange transfusion in the spoon should be filled just short of the brim with

SGA*

Term (borderline)
<35 wk

<3rd percentile 3rd 10th percentile

Admit nursery Monitor with mother Admit nursery

Breastfeeding or katori-spoon Breastfeeding <30 wk initial on IV


30 34 wk orogastric or katori-spoon
*Blood sugar, hematocrit, temperature monitoring
Fig. 1. Algorithm for management of SGA infants

Indian Journal of Pediatrics, Volume 75February, 2008 173


128 80

Ashok K. Deorari et al

expressed milk, should be placed at the corner of mouth significant morbidities (e.g., hypoglycemia, polycythemia,
and milk should be allowed to flow into the infants birth asphyxia) during hospital stay.
mouth slowly, avoiding any spillage. The infant would
actively swallow the milk. This process should be REFERENCES
repeated till the required amount has been fed. If the
infant does not actively accept and swallow the feed, an 1. United Nations Childrens Fund and World Health
attempt should be made to wake the infant with gentle Organization, Low Birthweight: Country,regional and global
stimulation. If he is still sluggish, do not insist on this estimates. UNICEF, New York, 2004.
method. It is better to switch back to gavage feeds till the 2. Arora NK, Paul VK, Singh M. Morbidity and mortality in
term infants with intrauterine growth retardation. J Trop
infant is ready.
Pediatr 1987; 33 : 186-189.
3. Teberg AJ, Walther FJ, Pena IC. Mortality, morbidity, and
outcome of the small-for-gestational-age preterm infant. Semin
SGA/IUGR BABIES WITH ABSENT OR REVERSED
Perinatol 1988; 12 : 84-94.
END-DIASTOLIC FLOW (AREDF) IN UMBILICAL 4. Mavalankar DV, Gray RH, Trivedi CR, Parikh VC. Risk
ARTERY factors for small for gestational age births in Ahmedabad,
India. J Trop Pediatr 1994; 40 : 285-290.
5. Arora NK, Singh M, Paul VK, Bhargava VL. Etiology of fetal
In the IUGR fetus, hypoxemia produces circulatory growth retardation in hospital born infants. Indian J Med Res
redistribution towards the brain and away from the 1987; 85 : 395-400.
viscera and placenta, culminating in umbilical artery or 6. Bhatia BD, Agarwal KN, Jain NP, Bhargava V. Growth
pattern of intrauterine growth retarded (IUGR) infants in first
aortic AREDF in the most severely affected fetuses. The
nine months of life. Acta Pediatr Scand 1984; 73 : 189-196.
combination of antenatal and persisting postnatal 7. Singh M. Disorders of weight and gestation. In Singh M, ed.
disturbances of gut perfusion, interacting with the Care of the Newborn, 5th ed. New Delhi; Sagar Publications,
metabolic demands of feeding, may adversely affect 1999; 224-245.
intestinal tissue oxygenation, combining with stasis and 8. Singh M, Giri SK, Ramachandran K. Intrauterine growth
curves of live-born infants. Indian Pediatr 1974; 11: 475-479.
immunological factors to contribute to the development
9. Paul VK. Management of LBW babies. In Deorari AK, ed.
of NEC. A review by Dorling et al including 14 studies NNF Teaching Aids on Newborn Care. 2nd ed, New Delhi; Noble
and 1178 neonates found higher risk of NEC in IUGR Vision, 1998; 25-36 .
babies with AREDF (odds ratio: 2.13; 95% CI: 1.49 to 10. Niermayer S, Kattwinkel J, Van Reempts P, Nadkarni V,
3.03).12 Although evidence for feeding strategy to be Phillips B, Zideman D et al. International guidelines for
adopted in these babies is limited, it may be prudent to neonatal resuscitation. An excerpt from the guidelines 2000
for cardiopulmonary resuscitation and emergency
start and persist with minimal enteral nutrition for first cardiovascular care. International consensus on Science.
48-72 h of life. Contributors and reviewers for the neonatal resuscitation
guiidelines. Pediatrics 2000; 106 : E29.
LONG-TERM OUTCOME AND FOLLOW-UP 11. Deorari AK, Paul VK, Shrestha L, Singh M. Symptomatic
neonatal polycythemia: comparison of partial exchange
transfusion with saline versus plasma. Indian Pediatr 1995; 32:
IUGR babies are at risk for poor growth and neuro- 1167-1171.
developmental outcome.3 We routinely follow IUGR 12. Dorling J, Kempley S, Leaf A. Feeding growth restricted
babies with birthweight below <3rd percentile and those preterm infants with abnormal antenatal Doppler results. Arch
with birthweight 3-10 th percentile if they develop Dis Child Fetal Neonatal Ed 2005; 90 : F359-F363.

174 Indian Journal of Pediatrics, Volume 75February, 2008


129
130
131
132
133 81

Symposium on AIIMS Protocols in Neonatology II

Jaundice in the Newborns


Satish Mishra, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi,
India

ABSTRACT
Hyperbilirubinemia is the commonest morbidity in the neonatal period and 5-10% of all newborns require intervention for
pathological jaundice. Neonates on exclusive breast- feeding have a different pattern and degree of jaundice as compared
to artificially fed babies.. Latest guidelines from American Academy of Pediatrics (AAP) for management of jaundice in a normal
term newborn have been included in the protocol. Separate guidelines have been provided for the management of jaundice
in sick term babies, preterm and low birth weight babies, for hemolytic jaundice and prolonged hyperbilirubinemia. [Indian
J Pediatr 2008; 75 (2) : 157-163] E-mail: sdeorari@yahoo.com

Key words : Jaundice; Newborns; Guidelines; Phototherapy; Exchange

Jaundice is an important problem in the first week of life. PATHOLOGICAL JAUNDICE


It is a cause of concern for the physician and a source of
anxiety for the parents. High bilirubin levels may be toxic
to the developing central nervous system and may cause TSB concentrations have been defined as non-physiologic
neurological impairment even in term newborns. Nearly if concentration exceeds 5 mg/dl on first day of life in
60% of term newborn becomes visibly jaundiced in the term neonate, 10 mg/dL on second day, or 12-13
first week of life.1 In most cases, it is benign and no thereafter. 6 Any TSB elevation exceeding 17 mg/dL
intervention is required. Approximately 5-10 % of them should be presumed pathologic and warrants
have clinically significant hyperbilirubinemia mandating investigation for a cause and possible intervention, such
the use of phototherapy.2-3 as phototherapy. 7 Appearance of jaundice within 24
hours, peak TSB levels above the expected normal range
(Fig. 1)8, presence of clinical jaundice beyond 3 weeks and
conjugated bilirubin (dark urine staining the clothes and
PHYSIOLOGICAL JAUNDICE
light colored stool) would be categorized under
pathological jaundice.
This is attributable to physiological immaturity of
neonates to handle increased bilirubin production. Visible
jaundice usually appears between 24-72 hours of age.
Total serum bilirubin (TSB) level usually rises in full-term
Serum Bilirubin (mg/dl)

High Risk Zone


infants to a peak of 6 to 8 mg/dL by 3 days of age and Zon
e
isk
eR
then falls. A rise to 12mg/dL is in the physiologic range. med
iat
k Zo
ne
mol/L

ter
h In te Ris
In premature infants, the peak may be 10 to 12 mg/dL on Hig
In term
edia
Low
the fifth day of life, possibly rising over 15 mg/dL
without any specific abnormality of bilirubin metabolism. Low Risk Zone

Levels under 2 mg/dL may not be seen until one month


of age in both full term and premature infants.4 Safe
bilirubin levels in preterms vary according to gestational
age. 5 Postnatal Age (hours)

Fig. 1. Nomogram for designation of risk in 2840 well newborns at


36 or more weeks gestational age with birth weight of 2000
Correspondence and Reprint requests : Dr Ashok K. Deorari, g or more or 35 or more weeks gestational age and birth
Professor, Department of Pediatrics, All India Institute of Medical weight of 2500 g or more based on the hour-specific serum
Sciences, Ansari Nagar, New Delhi 110029, India. bilirubin values 8
[Received January 1, 2008; Accepted January 1, 2008]

Indian Journal of Pediatrics, Volume 75February, 2008 157


134 82

S. Mishra et al

BREAST-FEEDING AND JAUNDICE TABLE 1. Guide to Dermal Staining with Level of Bilirubin
(Modified from Kramers Original Article)14
Exclusively breast-fed infants have a different pattern of Area of body Level of bilirubin
physiological jaundice as compared to artificially fed
babies. 7,9,10 Jaundice in breast-fed babies usually appears Face 4-6 mg/dl
Chest, upper abdomen 8-10 mg/dl
between 24-72 hours of age, peaks by 5-15 days of life and
Lower abdomen, thighs 12-14 mg/dl
disappears by the third week of life. They have also been Arms, lower legs 15-18 mg/dl
reported to have higher bilirubin levels. Schneiders meta- Palms, soles 15-20 mg/dl
analysis of 25 studies has shown that 13% of breast-fed
babies had peak TSB levels of 12 mg/dL or higher as phototherapy and if the baby has dark skin.
compared to 4% of artificially fed babies.11 One third of all
breast-fed babies are detected to have mild clinical
MEASUREMENT OF TSB LEVELS
jaundice in the third week of life, which may persist into
the 2nd to 3rd month of life in a few babies.
Biochemical: Laboratory estimation of TSB based on High
Authors have stated that this increased frequency is Performance Liquid Chromatography (HPLC) remains
not related to characteristics of breast milk but rather to the gold standard for TSB estimation. However, this test
the pattern of breast-feeding. Decreased frequency of is not universally available and laboratory estimation of
breast-feeding is associated with exaggeration of TSB usually done in labs is based on Vanden Bergh
physiological jaundice. Encouraging a mother to breast- reaction. It usually have marked interlaboratory
feed her baby at least 10-12 times per day would be variability with coefficient of variation up to 10 to 12
helpful in the management of jaundice in a term healthy percent for TSB and over 20 percent for conjugated
baby. fraction.15
Micro method for bilirubin estimation: It is based on
BREAST MILK JAUNDICE spectro-photometry and estimates TSB on a micro blood
sample. It is useful in neonates, as bilirubin is
predominantly unconjugated.
Approximately 2-4% of exclusively breast-fed term babies
have jaundice in excess of 10 mg/dL in the third week of Transcutaneous bilirubinometer: This method is non
life.12,13 These babies with TSB beyond 10 mg/dL after the invasive and based based on reflectance data of multiple
third week of life should be investigated for prolonged wavelengths from the bilirubin stained skin. It averages
jaundice. A diagnosis of breast milk jaundice should be the spectra of five replicate measurements at one site to
considered if the TSB is predominantly unconjugated, give bilirubin estimation in mmol/l (or mg/dl).
other causes of prolonged jaundice have been excluded Transcutaneous bilimeter (TcB) has a linear correlation to
and the infant is in good health. Mothers should be TSB and may be useful as a screening device to detect
advised to continue breast-feeding frequently intervals clinically significant jaundice and decrease the need for
and TSB levels usually decline over a period of time. frequent TSB determinations.16
Interruption of breast-feeding is not recommended unless
TSB level exceeds 20 mg/dl.
CLINICAL APPROACH TO JAUNDICE

CLINICAL EXAMINATION Is the newborn term or preterm? : Basic pathophysiology


of jaundice is same in term and preterm neonates but at
lower gestation babies are at higher risk of developing
Originally described by Kramer,14 dermal staining of hyperbilirubinemia and require closer surveillance and
bilirubin may be used as a clinical guide to the level of monitoring. TSB values for intervention also vary in term,
jaundice. Dermal staining in newborn progresses in a near-term, and preterm neonates less than 35 weeks
cephalo-caudal direction. The newborn should be period of gestation.
examined in good daylight. The skin should be blanched
with digital pressure and the underlying color of skin and Management of hyperbilirubinemia in preterm infants
subcutaneous tissue should be noted. A rough guide for is still in grey zone for lack of enough objective evidences.
level of dermal staining with level of bilirubin is included Clinical practice guidelines from American Academy of
in Table 1. Pediatrics (AAP) applies to the newborn infants of 35 or
more weeks of gestation.17
Newborns detected to have yellow discoloration of the
skin beyond the legs should have an urgent laboratory Is there evidence of hemolysis?: Setting of Rh or less
confirmation for levels of TSB. Clinical assessment is not frequently ABO incompatibility, onset of jaundice within
very reliable if a newborn has been receiving 24 hours, presence of pallor and hydrops, presence of

158 Indian Journal of Pediatrics, Volume 75February, 2008


135 83

Jaundice in the Newborns

hepato-splenomegaly, presence of hemolysis on the


peripheral blood smear, raised reticulocyte count (>8%),
rapid rise of bilirubin (>5 mg/dl in 24 hours or >0.5 mg/

Total Serum Bilirubin (mg/dl)


dl/hr) or a suggestive family history of significant
jaundice should raise a suspicion of hemolytic jaundice.

mol/L
However, end-tidal carbon monoxide corrected for
ambient carbon monoxide (ETCOc) levels can confirm the
presence or absence of hemolysis, and measurement of 38 wk and well)
Infants at lower risk (
ETCO c is the only clinical test that provides a direct Infants at medium risk ( 38 wk+risk factors or 35-37 6/7 wk. and well
Infants at higher risk (35-37 6/7 wk. + risk factors)
measurement of the rate of heme catabolism and the rate
of bilirubin production.17 Age
Use total bilirubin. Do not subtract direct reacting or conjugated bilirubin
Risk factors isoimmune hemolytic disease GGPD deficiency, asphyxia, significant lethargy, temperature,
Does the infant have an underlying serious disease? instability, sepsis, acidosis, albumin < 3.0 g/dL (If measured).
For well infants 35-37 6/7 wk can adjust TSB levels for intervention around the medium risk line. It is an options
(sepsis, galactosemia) : Presence of lethargy, poor to intervene at lower TSB level for infant closer to 35 wk and at higher TSB levels for those closer to 37 6/7 wk
It is an option to provide conventional phtotherapy in hospital or at home at TSB levels 2-3 mg/dL (35-50 mol/
feeding, failure to thrive, hepato-splenomegaly, L) below those shown but home phototherapy should not be used in any infants with risk factor

temperature instability or apnea may be a marker of an Fig. 2. Guidelines for phototherapy in hospitalized infants of 35 or
underlying serious disease. more weeks gestation17

Does the infant have cholestatic jaundice? Presence of jaundice. Repeat TSB in 424 h depending on infants age
jaundice (>10 mg/dl) beyond 3 weeks, presence of dark and TSB level.
urine (staining the clothes) or pale colored stools would
We usually do repeat TSB within 4 to 6 hr if initial TSB
suggest cholestatic jaundice.
was in or near the exchange transfusion range. Meanwhile
blood is arranged for the exchange transfusion, so that
JAUNDICE IN A TERM HEALTHY BABY exchange can be done if there is no significant fall in the
TSB. In a healthy neonate without setting for hemolytic
Advise for physiological jaundice: The parents should be jaundice and TSB not near exchange range we repeat TSB
explained about the benign nature of jaundice. The after 12 to 24 hr. In Rh isoimmunization we do repeat TSB
mother should be encouraged to breast-feed frequently. at 8 to 12 hr interval for first 48 hr and 12 to 24 hourly
The newborn should be exclusively breast-fed with no top afterwards when probability of unexpected rise in TSB
feeds, water or dextrose water. Mother should be told to usually decrease.
bring the baby to the hospital if the baby looks too yellow
We follow guidelines of the American Academy of
or yellow discoloration of the skin beyond the legs .
Pediatrics (AAP)s for initiating phototherapy in term and
Any newborn discharged prior to 72 hours of life TABLE 2. Risk Factors for Development of Severe Hyperbiliru-
should be evaluated again in the next 48 hours for binemia in Infants of 35 or More Weeks Gestation (in
adequacy of breast-feeding and progression of jaundice. Approximate Order of Importance)17

Clinical judgment should be used in determining Major risk factors


follow-up. Earlier or more frequent follow-up should be
Predischarge TSB or TcB level in the high-risk zone (Fig. 2)
provided for those who have risk factors for
Jaundice observed in the first 24 h
hyperbilirubinemia (Table 2).17 Blood group incompatibility with positive direct antiglobulin test,
other known hemolytic disease (e.g., G6PD deficiency)
Management of pathological jaundice: Any term or near-
Gestational age 3536 wk
term newborn noted to have yellow staining of the skin Previous sibling received phototherapy
beyond the legs/estimated clinical or TcB in the high risk Cephalohematoma or significant bruising
zone of nomogram should have a confirmatory serum If breastfeeding is not going well and weight loss is excessive
bilirubin level. The American Academy of Pediatrics
Minor risk factors
(AAP)17 has laid down criteria for managing babies with Predischarge TSB or TcB level in the high intermediate-risk zone
bilirubin in the pathological range (Fig. 2). Jaundice Gestational age 3738 wk
appearing within 24 hours should be managed as Jaundice observed before discharge
hemolytic jaundice. Previous sibling with jaundice
Macrosomic infant of a diabetic mother
All infants with bilirubin levels in the phototherapy
range should have the following investigations: blood Male gender
Decreased risk (these factors are associated with decreased risk of
type and Coombs test, if not obtained with cord blood (if significant jaundice, listed in order of decreasing importance)
mother is Rh negative or O group); complete blood count TSB or TcB level in the low-risk zone (Fig. 2)
and smear for hemolysis and red blood cell morphology; Gestational age 41 wk
reticulocyte count and G6PD estimation. These
Discharge from hospital after 72 h
investigations are done to exclude any hemolytic cause of

Indian Journal of Pediatrics, Volume 75February, 2008 159


136 84

S. Mishra et al

38 wk and well)
Infants at lower risk (
age are mentioned in Table 4. Intervention for Rh
Infants at medium risk ( 38 wk+risk factors or 35-37 6/7 wk. and well hemolytic disease in preterm babies is indicated at lower
Infants at higher risk (35-37 6/7 wk. + risk factors)
values. A level greater than 0.5% and 1% birth weight
Total Serum Bilirubin (mg/dl)

(Kg) can be used as a rough guide for phototherapy and


exchange blood transfusion respectively.

mol/L
TABLE 4. Indications for Exchange Transfusion in Rh
Isoimmunization 6

An exchange transfusion soon after birth is indicated if:


Cord bilirubin is 5mg/dl
Cord Hb is 10 mg/dl, PCV <30
Age
The dashed lines for the first 24 hours indicate uncertaintity due to a wide range of clinical circumstances and Previous sibling history and positive DCT.
a range of responses to phototherapy
Immediate exchange transfusion is recommended if infant shows signs of acute bilirubin encephalopathy Subsequent exchange transfusions are indicated if:
(hypertonia, arching, retrocollis fever, high pitched cry) or if TSB is 5 mg/dl (85 mol/L) above these lines.
Risk factor- Isoimmune hemolytic disease G6PD deficiency, asphyxia, significant lethargy, temperature,
1. Bilirubin 10 mg/dl within 24 hours of age
instability, sepsis, acidosis. 2. Bilirubin 15 mg/dl between 25-48 hours of age
Measure serum albumin and calculate B/A ratio (see legend)
Use total bilirubin do not subtract direct reacting or conjugeted bilirubin. 3. Bilirubin 20 mg/dl after 48 hours of age.
Infant is well and 35-37 6/7 wk (medium risk) can individualize TSB levels for exchange based on actual
gestational age. Rate of rise of bilirubin is 0.5 mg/dl/hr.

Fig. 3. Guidelines for exchange transfusion in infants 35 or more


weeks gestation17 Intravenous immunoglobulin (IVIG) is an alternative
therapy which may be effective in treating isoimmune
near term infants (Fig. 2).17 For preterm and VLBW infants haemolytic jaundice. In isoimmune haemolysis red blood
guidelines for phototherapy are not so clear for lack of cells are probably destroyed by an antibody-dependent
data. We follow the ranges given in Table 3. cytotoxic mechanism mediated by Fc receptor bearing
cells of the neonatal reticuloendothelial system. The
TABLE 3. Management of Neonatal Hyperbilirubinemia in Low putative mechanism of IVIG action is non-specific
Birth Weight Babies Based on Bilirubin Levels (mg/dl)5
blockade of Fc receptors. IVIG may be used in a dose of
Weight (gm) Phototherapy Consider exchange 0.5g to 1g/Kg (single dose) after the first ET/as early as
transfusion possible if ET not indicated. Phenobarbitone 5 mg/Kg/
day x 5 days may be started after the first ET/with in 12
500-750 5-8 12-15
750-1000 6-10 >15
hrs if ET not indicated
1000-1250 8-10 15-18 ABO Incompatibility: Babies born to women with O
1250-1500 10-12 17-20 blood group should be closely monitored for jaundice and
1500-2500 15-18 20-25
discharged after 72 hours. If the maternal blood is group
O, Rh-positive, it is an option to test the cord blood for the
For paucity of evidence these phototherapy guidelines infants blood type and direct antibody test, but it is not
are given only for first week of life. We follows the same required provided that there is appropriate surveillance,
guidelines for neonates with hyperbilirubinemia post first risk assessment before discharge, and follow-up. Jaundice
week of life, however, these babies are probably less prone due to ABO incompatibility usually appears in the first 24
for bilirubin induced brain damage with similar TSB. hours. In the presence of significant jaundice or jaundice
appearing within 24 hours, the work up for pathological
HEMOLYTIC JAUNDICE jaundice should be done. An approach to phototherapy
and ET has been outlined in section 15.

The common causes of hemolytic jaundice include Rh Other hemolytic states: G6PD deficiency, hereditary
hemolytic disease, ABO incompatibility, G-6-PD spherocytosis, minor group incompatibilities should be
deficiency and minor blood group incompatibility. managed similar to ABO incompatibility. Investigations
for G-6-PD deficiency should be considered in all term
Rh hemolytic disease: A baby born to an Rh-negative and near-term infants with jaundice requiring
mother (and Rh-positive father) should have Rh typing phototherapy, with a family history of significant jaundice
and a Direct Coombs test (DCT) on cord blood. or a geographic origin associated with G-6-PD deficiency.
Newborns suspected to have Rh isoimmunization should
have a blood group and Rh typing, DCT, PCV and serum
bilirubin on cord blood to facilitate early treatment. A JAUNDICE IN PRETERM BABIES
reticulocyte count should be sent prior to the first
exchange transfusion (ET). Intensive phototherapy should
be started at birth and continued till two consecutive In premature infants, the term physiologic jaundice is of
readings are below phototherapy range. Indications for little value. In VLBW infants, TSB levels well within the
exchange transfusion at birth and subsequently at a later physiologic range might be hazardous and sometimes

160 Indian Journal of Pediatrics, Volume 75February, 2008


137 85

Jaundice in the Newborns

need to be treated with phototherapy.18 screen


Clinicians should ensure that all premature infants are Investigations to rule out hypothyroidism
routinely monitored for the development of jaundice. Investigations to rule out urinary tract infection.
Serum bilirubin should be measured at 24 hours of age
with follow up estimations every 12-24 hours until the
TREATMENT OPTIONS
levels stabilize. Recommendations for starting
phototherapy in VLBW infants have been mentioned in
Table 3. Bilirubin should be repeated within 24-48 hours Phototherapy
of stopping phototherapy or sooner if clinical jaundice Factors That Affect the Dose and Efficacy of
reappears. Direct bilirubin should be measured weekly in phototherapy17
infants on parentral nutrition.
(a) Spectrum of light emitted: Blue-green spectrum is most
As a rough guide in healthy low birth weight infants effective. At these wavelengths, light penetrates skin
phototherapy should be started at a bilirubin level of 1% well and is absorbed maximally by bilirubin. Use
of the birth weight (in grams) e.g., a baby weighing 1000 special blue tubes or LED light source with output in
grams should receive phototherapy if the bilirubin levels blue-green spectrum for intensive PT.
exceed 10 mg/dl. An exchange transfusion should be
considered at a value of 5 mg/dl higher than that for (b) Spectral irradiance (irradiance in certain wavelength
phototherapy (1% of birth weight + 5 mg/dl). A sick band) delivered to surface of infant: If special blue
VLBW baby requires intervention at lower levels. fluorescent tubes are used, bring tubes as close to
infant as possible to increase irradiance. Special blue
SMALL FOR GESTATIONAL AGE (SGA) BABIES tubes 1015 cm above the infant will produce an
irradiance of at least 35 W/cm2 per nm.
Bilirubin handling in newborn is related to maturity of (c) Spectral power (average spectral irradiance across surface
liver, which is dependent upon gestational age. area) For intensive PT, expose maximum surface area
Gestational age and corresponding appropriate weight of infant to PT. Double surface phototherapy may be
may be a better guide for intervention as compared to more effective than single surface phototherapy.
actual birth weight in SGA infants.
(d) Cause of jaundice: PT is likely to be less effective if
jaundice is due to hemolysis or if cholestasis is
JAUNDICE IN A SICK NEWBORN present. When hemolysis is present, start PT at
lower TSB levels. Use intensive PT. Failure of PT
At high bilirubin levels sick neonates are more prone for suggests that hemolysis is the cause of jaundice.
bilirubin induces brain damage than the healthy neonate
(e) TSB level at start of PT: The higher the TSB, the more
of similar gestation and weight. Intervention for jaundice
rapid the decline in TSB with PT. Use intensive PT
in this group should start at lower levels of TSB (at a
for higher TSB levels. Anticipate a more rapid
centile line below the expected for that gestation in Fig. 2
decrease in TSB when TSB >20 mg/dL (342 mol/L).
and 3).
Continuous phototherapy : Is better than intermittent
Additional investigations in a sick newborn include
phototherapy. Phototherapy should be interrupted in a
direct bilirubin, septic screen, blood and urine culture and
newborn only during breast-feeding and nappy changes.
urine for reducing substances
Conventional phototherapy: If jaundice is non-hemolytic
PROLONGED JAUNDICE BEYOND 3 WEEKS or rate of rise of jaundice is slow then one can use either
conventional or fibre-optic phototherapy units.

This is defined as persistence of significant jaundice (10 Intensive phototherapy: In case of hemolytic or rapidly
mg/dl) beyond three weeks in a term baby. The common rising bilirubin or when a conventional unit is not
causes include breast milk jaundice, extravasated blood effective, use of intensive phototherapy is warranted. This
(cephalhematoma), ongoing hemolytic disease, G-6PD can be achieved by placing the infant on bili-blanket and
deficiency and hypothyroidism. One should rule out using additional overhead phototherapy units with
cholestasis by noting the urine and stool color and special blue lights and lowering the phototherapy units to
checking the level of direct bilirubin. The diagnostic work within a distance of 15-20 cm. Intensive phototherapy
up in such a newborn includes: implies irradiance in the blue-green spectrum
(wavelengths of approximately 430490 nm) of at least 30
Investigations to rule out cholestasis (stool color, urine W/cm2 per nm (measured at the infants skin directly
color, direct and indirect bilirubin levels) below the center of the phototherapy unit) and delivered
Investigations to rule out ongoing hemolysis, G-6PD to as much of the infants surface area as possible.

Indian Journal of Pediatrics, Volume 75February, 2008 161


138 86

S. Mishra et al

Measurements should be made with a radiometer compatible) packed cells suspended in AB plasma or O
specified by the manufacturer of the phototherapy group whole blood (Rh compatible with baby).
system.17
Other situations: Cross-matched with babys blood
Hydration: Maintaining adequate hydration and good group.
urine output should help to improve the efficacy of
Blood volume used
phototherapy.
Partial exchange done at birth in Rh hemolytic disease
Failure of phototherapy: Failure of phototherapy has
with severe anemia / hydrops (aims at increasing the
been defined as an inability to observe a decline in
oxygen carrying capacity of blood): 50 ml/ kg of
bilirubin of 1-2 mg/dl after 4-6 hours and/ or to keep the
packed cells
bilirubin below the exchange transfusion level. Exchange
transfusion is recommended if the TSB rises to these levels Double volume exchange: 2 x (80-100 ml/kg) x birth
despite intensive phototherapy. For readmitted infants, if weight in Kg (arrange 70% PRBC and 30% FFP, so that
the TSB level is above the exchange level, repeat TSB PCV of whole arranged blood ~ 50-55)
measurement every 2 to 3 hours and consider exchange if
Pharmacological Treatment
the TSB remains above the levels indicated after intensive
phototherapy for 6 hours. The recommended levels for Phenobarbitone: It improves hepatic uptake, conjugation
exchange transfusion have been given in Fig. 3. However, and excretion of bilirubin thus helps in lowering of
an exchange transfusion (ET) may be performed at the bilirubin. However, its effect takes time. When used
slightest suspicion of bilirubin encephalopathy prophylactically in a dose of 5 mg/Kg for 3-5 days after
irrespective of the bilirubin value. birth, it has shown to effective in babies with hemolytic
disease, extravasated blood and in preterms without any
When Should Phototherapy Be Stopped?: There is no
significant side effects.
standard for discontinuing phototherapy. For infants who
are readmitted after their birth hospitalization (usually for Intravenous Immunoglobulins (IVIG): High dose
TSB levels of 18 mg/dL or higher), phototherapy may be intravenous g-globulin (IVIG) (0.5 to 1 gm/Kg) has been
discontinued when the serum bilirubin level falls below shown to reduce the need for exchange transfusions in Rh
13 to 14 mg/dL. Discharge from the hospital need not be and ABO hemolytic disease.17
delayed to observe the infant for rebound. If phototherapy
is used for infants with hemolytic diseases or is initiated Pharmacologic Therapy: There is now evidence that
early and discontinued before the infant is 3 to 4 days old, hyperbilirubinemia can be effectively prevented or treated
a follow-up bilirubin measurement within 24 hours after with tin-mesoporphyrin, a drug that inhibits the
discharge is recommended. For infants who are production of heme oxygenase. Tin-mesoporphyrin is not
readmitted with hyperbilirubinemia and then discharged, approved by the US Food and Drug Administration. If
significant rebound is rare, but a repeat TSB measurement approved, tin-mesoporphyrin could find immediate
or clinical follow-up 24 hours after discharge is a clinical application in preventing the need for exchange
option. transfusion in infants who are not responding to
phototherapy.
Sunlight Exposure: Although sunlight provides sufficient
irradiance in the 425- to 475-nm band to provide FOLLOW-UP
phototherapy, the practical difficulties involved in safely
exposing a naked newborn to the sun either inside or Babies with serum bilirubin 20 mg/dl and those who
outside (and avoiding sunburn) preclude the use of require exchange transfusion should be kept under
sunlight as a reliable therapeutic tool, and it therefore is follow-up in the high- risk clinic for neuro-developmental
not recommended. outcome. Hearing assessment (BERA) should be done at
Exchange transfusion 0-3 months of corrected age. With prompt treatment, even
very elevated serum bilirubin levels within the range of
Rh isoimmunization: Blood used for exchange 25 to 29 mg/dl are not likely to result in long-term
transfusion in neonates with Rh isoimmunization should adverse effects on neurodevelopment. 19 Provisional
always have Rh negative blood group. The best choice committee for quality improvement and subcommittee on
would be O (Rh) negative packed cells suspended in AB hyperbilirubinemia.
plasma. O (Rh) negative whole blood or cross-matched
babys blood group (Rh negative) may also be used in an REFERENCES
emergency.
1. American Academy of Pediatrics. Practice parameter:
ABO incompatibility: Only O group blood should be
management of hyperbilirubinemia in the healthy term
used for exchange transfusion in neonates with ABO newborn. Pediatrics 1994; 94 : 558-565.
incompatibility. The best choice would be O group (Rh 2. Maisels MJ, Gifford K, Antle CE, Lab GR. Jaundice in the

162 Indian Journal of Pediatrics, Volume 75February, 2008


139 87

Jaundice in the Newborns

healthy newborn infant: a new approach to an old problem. 12. Clarkson JE, Cowan JO, Herbison GP. Jaundice in full term
Pediatrics 1988; 81 : 505-511. healthy neonates: A population study. Aust Pediatr J 1984;
3. Kiviahan C, Jams EJ. The natural history of neonatal jaundice. 20 : 303-308.
Pediatrics 1984; 74 : 364-370. 13. Winfield CR, MacFaul R. Clinical study of prolonged jaundice
4. Martin CR, Cloherty JP. Neonatal hyperbilirubinemia. In in breast and bottle-fed babies. Arch Dis Child 1978; 53: 506-
Cloherty JP, Eichenwald EC, Stark AR, eds. Manual of Neonatal 507.
Care, 15th ed. Philadelphia: Lippincott Williams & Wilkins, 14. Kramer LI. Advancement of dermal icterus in jaundiced
2004; 185-221. newborn. Am J Dis Child 1969; 118 : 454-458.
5. Cashore WJ. Bilirubin and jaundice in the micropremie. Clin 15. Singh M. Jaundice. In Singh M, ed. Care of the Newborn, 6th ed.
Perinatol 2000; 27 : 171-179. New Delhi; Sagar Publications, 2004; 248.
6. Madan A, Mac Mohan JR, Stevenson DK. Neonatal 16. Stanley Ip, Mei Chung, John Kulig, Rebecca OBrien, Robert
Hyperbilirubinemia. In Taeush HW, Ballard RA, Gleason CA, Sege et al. Technical report, An evidence-based review of
eds. Averys Diseases of the Newborn, 8th ed. Philadelphia; WB important issues concerning neonatal hyperbilirubinemia.
Saunders, 1226-1256. Pediatrics 2004; 114 : e130-e150.
7. Maisels MJ, Gifford K. Normal serum bilirubin levels in new- 17. Maisels MJ, Baltz RD, Bhutani VK et al. Management of
borns and effect of breast-feeding. Pediatrics 1986; 78 : 837-843. hyperbilirubinemia in the newborn infant 35 or more weeks of
8. Bhutani VK, Johnson L, Sivieri EM. Predictive ability of a gestation. Pediatrics 2004; 114 : 297-316.
predischarge hour-specific serum bilirubin for subsequent 18. Maisels MJ. Jaundice. In MacDonald MG, Mullett MD, Seshia
significant hyperbilirubinemia in healthy term and near-term MMK, eds. Averys Neonatology Pathophysiology and
newborns. Pediatrics 1999; 103 : 6-14. Management of the Newborn, 6th ed. Philadelphia; Lippincott
9. Gartner LM, Herschel M. Jaundice and breast-feeding. Pediatr Williams & Wilkins; 2005; 775.
Clin North Am 2001; 48 : 389-399. 19. Thomas BN, Petra L, Rita JJ, Donna MF, Yvonne WW.
10. Gartner LM, Lee KS. Jaundice in the breast-fed infant. Clin Outcomes among Newborns with Total Serum Bilirubin
Perinatol 1999; 26 : 431-445. Levels of 25 mg per Deciliter or More. NEJM 2006; 354 : 1889-
11. Schneider AP. Breast milk jaundice in the newborn: A real 1900.
entity. JAMA 1986; 255 : 3270-3274.

Indian Journal of Pediatrics, Volume 75February, 2008 163


140
141
142
143
144 88

Symposium on AIIMS Protocols in Neonatology V

Kangaroo Mother Care-an Alternative to Conventional


Care
Anu Thukral, Deepak Chawla, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar,
New Delhi, India

ABSTRACT

The term kangaroo mother care ( KMC ) is derived from practical similarities to marsupial care-giving, i.e., the premature infant
is kept warm in the maternal pouch and close to the breasts for unlimited feeding. It is a gentle and effective method that avoids
agitation routinely experienced in a busy ward with preterm infants. An important main stay of kangaroo mother care is
breastfeeding encouragement. Observational studies have shown reduction in mortality after institution of KMC. Preterm
babies exposed to skin to skin contact showed a better mental development and better results in motor tests. It also improves
thermal care. All stable LBW babies are candidate for KMC. Often this is desirable, until the babys gestation reaches term
or the weight is around 2500 g. The mother and family members are encouraged to take care of the baby in KMC and should
be counseled to come for follow-up visits regularly. [Indian J Pediatr 2008; 75 (5) : 497-503] E-mail: ashokdeorari_56
@hotmail.com

Key words : Kangaroo mother care; Skin to skin contact; Thermal care low birth weight; Skin contact and breast feeding

Kangaroo mother care (KMC) is care of preterm or low and close to the breasts for unlimited feeding. The
birth weight infants carried skin-to-skin with the mother. mothers are used as incubators and as the main source
KMC was initially conceived as an alternative to the usual of food and stimulation for LBW infants while they
minimal in-hospital care for stable low birth weight mature enough to face extrauterine life in similar
infants. Low birthweight, defined as weight at birth of less conditions as those born at term. The method is applied
than 2500 g irrespective of gestational age, has an adverse only after the LBW infant has stabilized. Introduction of
effect on child survival and development. World-wide, KMC results in early hospital discharge of low birth
twenty-five million LBW infants are born each year, the weight infants.
great majority (96%) of them in developing countries.
In the last few decades, several health services have
Conventional neonatal care of LBW infants is expensive
adopted KMC, thus showing that it is possible to adapt
and needs both trained personnel and permanent logistic
this practice to different contexts of access to neonatal care
support. In developing countries, financial and human
technology. The first experiments conducted in
resources for neonatal care are limited and hospital wards
industrialized countries demonstrated that KMC was safe
for LBW infants are often overcrowded. Thus,
in terms of physiological response of newborns and that
interventions for LBW infants that reduce neonatal
the method brought benefits regarding the breast feeding
morbidity, mortality and costs would be desirable in
practice and decreased the number of hospitalizations, in
developing countries like India.
addition to reduced infant crying at six months of life.1, 2
KMC was first suggested in 1978 by Dr Edgar Rey in Experiments carried out in developing countries have
Bogot, Colombia. The term kangaroo care is derived shown that KMC is safe in terms of mortality and may
from practical similarities to marsupial care-giving, i.e., reduce severe morbidity and avoid hospital
the premature infant is kept warm in the maternal pouch readmissions.3, 4 Therefore, KMC can potentially improve
the health and survival of LBW newborns, especially in
those places where resources are scarce.5
The major components of KMC are: (1) skin-to-skin
Correspondence and Reprint requests : Dr. Ashok Deorari,
Professor, Department of Pediatrics, All India Institute of Medical contact. Babies are kept, day and night, between the
Sciences, Ansari Nagar, New Delhi 110029, India mothers breasts firmly attached to the chest in an upright
[Received April 28. 2008; Accepted April 28, 2008] position, (2) frequent and exclusive or nearly exclusive

Indian Journal of Pediatrics, Volume 75May, 2008 497


145 89

A. Thukral et al

breast feeding and (3) early discharge from hospital hence a follow-up program and access to emergency
regardless of weight or gestational age. Respiratory, services must be ensured.
thermal and feeding stabilization are crucial for the
Finally it is a gentle and effective method that avoids
success of this intervention. The definition of stabilization
agitation routinely experienced in a busy ward with
is not precise, and has been defined as independent of
preterm infants.
gestational age and weight, which are the main variables
associated with those vital functions.
BENEFITS OF KMC
COMPONENTS OF KMC
Physiological benefits
Kangaroo position Heart and respiratory rates, respiration, oxygenation,
oxygen consumption, blood glucose, sleep patterns and
The kangaroo position consists of skin-to-skin contact
behavior observed in preterm/LBW infants held skin-to-
(SSC) between the mother and the infant in a strictly
skin tend to be similar to or better than those observed in
vertical position, between the mothers breasts and under
infants separated from their mothers.2, 3
her clothes. SSC should be started as early as possible
after birth and can be of two types depending upon the
duration: continuous or intermittent.1 The continuous CLINICAL BENEFITS
modality is usually employed as an alternative to
minimal care in an incubator for infants who have already
overcome major problems while adapting to extra-uterine Effect on breastfeeding
life, are able to suck and swallow properly and are An important main-stay of kangroo mother care is
thriving in neutral thermal environment. To replace breastfeeding encouragement. Although evidence shows
incubators the kangaroo position should be maintained as countless benefits of breastfeeding for preterm babies, the
long as possible, ideally 24 hr/day. The provider must prevalence of breastfeeding in this group is quite low. 6-
sleep in a semi- reclining position to avoid the reflux in 11
Studies carried out in areas where KMC is done show
more preterm infants. The Kangaroo position is that mothers who establish skin to skin contact with their
maintained until the infant no longer tolerates it- he preterm babies have a significantly higher milk
sweats and refuses the Kangaroo position. When production than their control group. Furthermore these
continuous care is not possible, the kangaroo position can studies have also revealed that interruption of breast
be used intermittently, providing the proven emotional feeding was more frequent among mothers who were not
and breastfeeding promotion benefits. The kangaroo submitted to this method. Whitelaw et al carried out a
position must be offered for as long as possible (1-2 hrs at randomized trial among babies less than 1500 gm and
least), provided the infant tolerates it well. This 1-2 h span found higher breastfeeding rates at 6 weeks in KMC
is important as it provides the stimulation that the mother group (55% vs 28%).2
needs to increase the milk volume and facilitate milk let-
down. This is initiated in the hospital and continued at Ramanathan et al conducted a randomized control trial
home. to study the effect of Kangaroo Mother Care (KMC) on
breast feeding rates, weight gain and length of
Kangaroo nutrition hospitalization of very low birth neonates and to assess
Kangaroo nutrition is the delivery of nutrition to the acceptability of Kangaroo Mother Care by nurses and
kangarooed infants as soon as oral feeding is possible. mothers.12 Babies whose birth weight was less than 1500
It is based on exclusive breastfeeding by direct sucking, grams were included in the study once they were stable.
whenever possible. Goal is to provide exclusive or nearly Neonates in the KMC group demonstrated better weight
exclusive breastfeeding with fortification if needed. gain after the first week of life (15.9 4.5 gm/day vs 10.6
Breastfeeding is an integral component of KMC and it 4.5 gm/day in the KMC group and control group
might contribute to significant gains in neurological respectively) and earlier hospital discharge (27.2 7 vs
development and IQ. 34.6 7 days in KMC and control group respectively).
The number of mothers exclusively breastfeeding their
Kangaroo discharge and follow-up babies at 6 week follow-up was double in the KMC group
Early home discharge in the kangaroo position from the than in the control group (12/14 vs 6/14). Rao et al also
neonatal unit is one of the original components of the found higher rate of exclusive breastfeeding in KMC
KMC intervention. If not safely possible, the mother- group.13
infant dyad can room-in together in a minimal care In a meta-analysis by Neonatal Review Group of the
facility (kangaroo wards) until safe discharge is possible. Cochrane Collaboration, randomized trials comparing
Mothers at home require adequate support and follow-up KMC and conventional neonatal care in LBW infants were

498 Indian Journal of Pediatrics, Volume 75May, 2008


146 90

Kangaroo Mother Care-an Alternative to Conventional Care

analyzed.14 Three studies, involving 1362 infants, were differences were seen in weight, length, or head
included. All the trials were conducted in developing circumference at 41 weeks corrected gestational age or at
countries. KMC was found to decrease probability of not 12 months corrected age or in weight at discharge.
exclusively breastfeeding at discharge (relative risk 0.41,
Other effects
95% confidence interval 0.25 to 0.68).
Prolonged skin-to-skin contact between the mother and
Finally, studies of the effect of KMC on breast feeding
her preterm/LBW infant, as in KMC, provides effective
show that, where KMC is practiced, the duration of breast
thermal control and may be associated with a reduced risk
feeding is prolonged, breast milk production is more
of hypothermia.12,13, 17,18
stable, the number of feeds per day is increased, breast
feeding competence is increased, and more premature In 1989, Affonso et al19 in a study involving 33 mothers
infants are discharged on exclusive breast feeding. who had skin to skin contact with their babies and a
Mothers expressed a clear preference for KMC and health control group, observed a greater tendency towards
workers found it safe and convenient. KMC was cheaper emotional stability in mothers exposed to KMC. They also
in terms of salaries and other running costs. reported a more intense feeling of reliability and
competence in these mothers as compared with those
Mortality and morbidity
mothers whose babies received conventional care. In a
Observational studies have shown reduction in mortality study involving 488 mothers of preterm babies, Tessier et
after institution of KMC but interpretation of their results al 20 observed that those submitted to KMC felt more
is difficult because of remarkable difference in KMC competent and had an increased perception of babys
group from historical controls.15 competences. Moreover they felt less stressed even when
the hospital stay was longer. These subjective feelings
A systematic review conducted by Cochrane Library14,
may be seen as positive indirect signs of establishment of
concluded that although KMC does not improve survival,
parent and child bonding favored by KMC and could be
it reduces the incidence of respiratory tract infections and
positive signs of greater participation of parents in baby
decreases the rate of nosocomial infections..The authors
care and stimulation, allowing them a more
concluded that although KMC seemingly reduces infant
comprehensive and individualized care.
morbidity, evidence is still insufficient to recommend it as
a routine practice. They also pointed out that more Brazelton21 in a careful study with babies asserted that
randomized; controlled and well designed trials should be a preterm babys nervous system becomes more easily
carried out. organized while in calm environment without excessive
stimuli. By observing the physiological pattern during
Recently, a randomized control trial conducted in two
intrauterine life it is generally noted that babys
hospitals in South Africa16 revealed that babies who were
experiences occur in a cyclic rhythm of activity and are
kept skin to skin contact had better outcome as regards
determined by the possibility to rest and sleep whenever
physiological stabilization compared to those babies who
necessary, which according to Korrones22 occurred 80% of
were kept in incubators.
the time. Mann et al23 regard sleep as a positive influence
Growth on the development of the brain.
Mixed results are reported about long term growth Preterm babies exposed to skin to skin contact showed
following KMC. A RCT from India has shown higher a better mental development and better results in motor
increments in weight, length and head circumference in tests. The presence of parents facilitates skin to skin
kangaroo infants in neonatal period.12 In another RCT contact which in its turn allows tactile proprioreceptive
KMC infants showed a slightly larger daily weight gain stimulation and protects against an overload of aversive
while they were cared for in hospital, but in the overall stimuli, being an acceptable method for proper
period of study their growth did not differ from that of stimulation of the babys neurobehavioral development.
the control group. 13 In a meta-analysis by Neonatal
Review Group of the Cochrane Collaboration randomized Cost-effectiveness
trials14 comparing KMC and conventional neonatal care in
Cost-effectiveness of KMC as compared to conventional
LBW infants, KMC infants had gained more weight per
care has not been studies well and there is a need to
day by discharge than controls (weighted mean difference
conduct such studies especially in low-resource settings.
3.6 g/day, 95% confidence interval 0.8 to 6.4) and had a
larger head circumference at 6 months corrected age than Use of KMC in different settings
controls (weighted mean difference 0.34 cm, 95%
KMC may be used in three different scenarios:
confidence interval 0.11 to 0.57) although these differences
are of low clinical significance. Sloan et al in 1994 reported Settings with a very low level of development and
there were no significant differences between the groups severely restricted access to any level of neonatal care.
in growth indices during the 6-month follow-up. No Premature infants who are born at home with no

Indian Journal of Pediatrics, Volume 75May, 2008 499


147 91

A. Thukral et al

assistance or with the assistance of a traditional birth about KMC, problems faced while implementing KMC
attendant and preterm infants who are born at first programs were discussed and possible solutions were
level unit with no specialized care and no possibility suggested. The basic strategy for facilitating the
of being transferred to a healthcare unit with implementation and success of KMC programs can be
specialized care. In such cases, KMC including skin- summarized in three words: communication,
to-skin contact, breastfeeding and the best possible sensitiveness and education.
health-care follow-up represents the best means for
Nurses, physicians and other staff involved in the care
ensuring the survival of non-sick premature infants
of the mother and the baby should be trained. This
who have no significant conditions other than being
training may best be done by exposing them to units
premature. Safety, efficacy and applicability of KMC
already practicing KMC. Educational material such as
in such a setting needs to be further investigated.
information sheets, posters, video films on KMC in local
Access to appropriate resources but which are language should be available to the mothers, families and
insufficient for the number of premature births. Here community. If possible, reclining chairs in the nursery
KMC represents an effective alternative which allows and postnatal wards, and beds with adjustable back rest
better utilization of available resources. should be arranged. Mother can provide KMC sitting on
an ordinary chair or in a semi-reclining posture on a bed
Little or no restriction on access to high-technology
with the help of pillows.
neonatal care. KMC is used mainly during
hospitalization to establish healthy bonding between Criteria for eligibility of KMC
mother and infant and to increase the breastfeeding
rates. The intermittent kangaroo position in hospital Baby
is the most widely used component in such a setting.
All stable LBW babies are eligible for KMC. However,
Requirements for KMC implementation very sick babies needing special care should be cared
under radiant warmer initially. KMC should be started
KMC is feasible everywhere, because it is not based on after the baby is hemodynamically stable. Short KMC
equipment, and it presents advantages for the sessions can be initiated during recovery with ongoing
organization of health services provided the following medical treatment (IV fluids, oxygen therapy). KMC can
requirements are met: be provided while the baby is being fed via orogastric
The health facility (the hospital or the neonatal ward) tube or on oxygen therapy. Guidelines for practicing
should have an open doors policy for parents at all KMC include:
times. A room near to or at the neonatal unit,
Birth weight >1800 g: These babies are generally stable at
furnished with comfortable seats for the mothers, is
birth. Therefore, in most of them KMC can be initiated
needed for conducting the kangaroo adaptation and
soon after birth.
for teaching and training mothers. The presence of a
nurse available full time, trained in the kangaroo Birth weight 1200-1799 g: Many babies of this group have
technique, is indispensable. significant problems in neonatal period. It might take a
Breastfeeding must be the official feeding policy of the few days before KMC can be initiated. If such a baby is
hospital for all newborn infants. born in a place where neonatal care services are
inadequate, he should be transferred to a proper facility
Staff should receive adequate training on KMC, immediately after birth, along with the mother/ family
including the special breastfeeding and feeding needs member. He should be transferred to a referral hospital
of LBWI. Extra training is usually needed on after initial stabilization and appropriate management.
stimulation of breastfeeding, expression and One of the best ways of transporting small babies is by
conservation of breast milk, mode of administration of keeping them in continuous skin-to-skin contact with the
expressed breast milk, and daily monitoring of mother/family member during transport.
growth.
Birth weight <1200 g: Frequently, these babies develop
Whenever good quality follow-up (including initial
serious prematurity-related morbidity, often starting soon
daily follow up visits) cannot be warranted, early
after birth. They benefit the most from in-utero transfer to
discharge in kangaroo position should be not
the institutions with neonatal intensive care facilities. It
attempted. On the contrary, KMC should be provided
may take days to weeks before babys condition allows
as an in-hospital activity, allowing mothers and infants
initiation of KMC.
to room together for as long as needed.
Mother
Besides human and physical resources, implementing a
KMC program needs institutional, social and health care All mothers can provide KMC, irrespective of age, parity,
workers support. During the II International Workshop education, culture and religion.24 The following points

500 Indian Journal of Pediatrics, Volume 75May, 2008


148 92

Kangaroo Mother Care-an Alternative to Conventional Care

must be taken into consideration when counseling on KMC procedure


KMC:
Kangaroo positioning: The baby should be placed
Willingness: The mother must be willing to provide between the mothers breasts in an upright position. The
KMC. Healthcare providers should counsel and head should be turned to one side and in a slightly
motivate her. Once the mother realizes the benefits of extended position. This slightly extended head position
KMC for her baby, she will learn and undertake KMC. keeps the airway open and allows eye to eye contact
between the mother and her baby. The hips should be
General health and nutrition: The mother should be
flexed and abducted in a frog position; the arms should
free from serious illness to be able to provide KMC.
also be flexed. Babys abdomen should be at the level of
She should receive adequate diet and supplements
the mothers epigastrium. Mothers breathing stimulates
recommended by her physician.
the baby, thus reducing the occurrence of apnea (Fig. 1).
Hygiene: The mother should maintain good hygiene: Support the babys bottom with a sling/binder.
daily bath/sponge, change of clothes, hand washing,
Monitoring: Babies receiving KMC should be monitored
short and clean finger nails.
Supportive family: Apart from supporting the mother,
family members should also be encouraged to provide
KMC when mother wishes to take rest. Mother would
need familys cooperation to deal with her
conventional responsibilities of household chores till
the baby requires KMC.
Supportive community: Community awareness about
the benefits should be created. This is particularly
important when there are social, economic or family
constraints.
Initiation of KMC
Counseling: When baby is ready for KMC, arrange a time
that is convenient to the mother and her baby. The first
few sessions are important and require extended
interaction. Demonstrate to her the KMC procedure in a
caring, gentle manner and with patience. Answer her
queries and allay her anxieties. Encourage her to bring her Fig. 1. Kangaroo positioning
mother/mother in law, husband or any other member of carefully especially during the initial stages. Nursing staff
the family. It helps in building positive attitude of the should make sure that babys neck position is neither too
family and ensuring family support to the mother which flexed nor too extended, airway is clear, breathing is
is particularly crucial for post-discharge home-based regular, color is pink and baby is maintaining
KMC. 25 It is helpful that the mother starting KMC temperature. Mother should be involved in observing the
interacts with someone already practicing KMC for her baby during KMC so that she herself can continue
baby. monitoring at home.
Mothers clothing: KMC can be provided using any front- Feeding: The mother should be explained how to
open, light dress as per the local culture. KMC works well breastfeed while the baby is in KMC position. Holding the
with blouse and sari, gown or shawl. Suitable apparel that baby near the breast stimulates milk production. She may
can retain the baby for extended period of time can be express milk while the baby is still in KMC position. The
adapted locally. baby could be fed with paladai, spoon or tube, depending
Babys clothing: Baby is dressed with cap, socks, nappy, on the condition of the baby.
and front-open sleeveless shirt. Privacy: KMC unavoidably requires some exposure on
the part of the mother. This can make her nervous and
Time of initiation
could be de-motivating. The staff must respect mothers
KMC can be started as soon as baby is stable and sensitivities in this regard and ensure culturally
receiving oral feeds. Babies with severe illness and those acceptable privacy standards in the nursery and the
requiring special treatment must wait until recovery wards where KMC is practiced.
before KMC can be started. Short KMC sessions can be
initiated during recovery with ongoing medical treatment Duration: Skin-to-skin contact should start gradually in
(iv fluids, low concentration of additional oxygen) the nursery, with a smooth transition from conventional

Indian Journal of Pediatrics, Volume 75May, 2008 501


149 93

A. Thukral et al

care to continuous KMC. Sessions that last less than one continued for as long as possible, at the institution and
hour should be avoided because frequent handling may then at home. Often this is desirable until the babys
be stressful for the baby. The length of skin-to-skin gestation reaches term or the weight is around 2500 g. She
contacts should be gradually increased up to 24 hours a starts wriggling to show that she is uncomfortable, pulls
day, interrupted only for changing diapers. When the her limbs out, cries and fusses every time the mother tries
baby does not require intensive care, she should be to put her back skin to skin. This is the time to wean the
transferred to the post-natal ward where KMC should be baby from KMC. Mothers can provide skin-to-skin
continued. contact occasionally after giving the baby a bath and
during cold nights.
Can the mother continue KMC during sleep and
resting? Post-discharge follow-up
The mother can sleep with baby in kangroo position in Close follow up is a fundamental pre-requisite of KMC
reclined or semi recumbent position about 15 degrees practice. Baby is followed once or twice a week till 37-40
from horizontal. This can be done with an adjustable bed weeks of gestation or till the baby reaches 2.5-3 kg of
or with pillows on an ordinary bed. A comfortable chair weight. Thereafter, a follow up once in 2-4 weeks may be
with an adjustable back may be used for resting during enough till 3 months of post-conception age. Later the
the day (Fig 2). baby should be seen at an interval of 1-2 months during
first year of life. The baby should gain adequate weight
(15-20 gm/kg/day up to 40 weeks of post-conception age
and 10 gm/kg/ day subsequently). More frequent visits
should be made if the baby is not growing well or his
condition demands.

REFERENCES

1. Ludington-Hoe SM, Hadeed AJ, Anderson GC. Physiological


response to skin to skin contact in hospitalized premature
infants. J Perinatol 1991; 11: 19-24
2. Whitelaw A, Heisterkamp G, Sleath K, Acolet D, Richards M.
Skin to skin contact for very low birthweight infants and their
mothers. Arch Dis Child 1988; 63: 1377-1381.
3. Sloan NL, Camacho LW, Rojas EP, Stern C. Kangaroo mother
method: randomized controlled trial of an alternative method
Fig. 2. Kangaroo mother care being provided in postnatal ward
of care for stabilized low-birthweight infants. Maternidad
Isidro Ayora Study Team. Lancet 1994; 344: 782-785.
Discharge criteria 4. Charpak N, Ruiz Pelaez JG, Charpak Y. Rey-Martinez
Kangaroo Mother Program: an alternative way of caring for
The standard policy of the unit for discharge from the low birth weight infants? One year mortality in a two cohort
hospital should be followed. Generally the following study. Pediatrics 1994; 94: 804-810.
criteria are accepted at most centers:26 5. Cattaneo A, Davanzo R, Worku B et al. Kangaroo mother care
for low birthweight infants: a randomized controlled trial in
Babys general health is good and no evidence of different settings. Acta Paediatr 1998; 87: 976-985.
infection 6. American Academy of Pediatrics. Workgroup on
breastfeeding and use of human milk. Pediatrics 1997; 100:
Feeding well, and receiving exclusively or 1035-1039.
predominantly breast milk. 7. Amin SB, Merle KS, Orlando MS, Dalzell LE, Guillet R. Brain
stem maturation in premature infants as a function of enteral
Gaining weight (at least 15-20 gm/kg/day for at least feeding type. Pediatrics 2000; 106: 318-320.
three consecutive days) 8. Carlson SE, Cooke RJ, Rhodes PG. Long term feeding of
formulas high in linolenic acid and marine oil to very low
Maintaining body temperature satisfactorily for at birth weight infants: phospholipids fatty acids. Pediatr Res
least three consecutive days in room temperature. 1991; 30: 404-412.
9. Carlson SE, Werman SH, Cooke RJ, Rhodes PG, Peeples JM,
The mother and family members are confident to take Tolley EA. Visual acuity development in healthy preterm
care of the baby in KMC and should be asked to come infants: effect of marine oil supplementation. Am J Clin Nutr
for follow-up visits regularly. 1993; 58: 35-42.
10. Lucas A, Morley R, Cole TJ, Lister G, Leeson Payne C. Milk
When to discontinue KMC? and subsequent intelligent quotient in infants born preterm.
Lancet 1992; 339: 261-264
When the mother and baby are comfortable, KMC is 11. Hurst NM, Valentine CJ, Renfro L, Burns P, Ferlic L. Skin-to-

502 Indian Journal of Pediatrics, Volume 75May, 2008


150 94

Kangaroo Mother Care-an Alternative to Conventional Care

skin holding in the neonatal intensive care unit influences Pediatr 2005; 72 : 35-38.
maternal milk volume. J Perinatol 1997; 17 : 213-217. 19. Affonso DD, Wahelber V, Persson B. Exploration of mothers
12. Ramanathan K, Paul VK, Deorari AK, Taneja U, George G. reactions to the kangroo method of prematurity care. Neonatal
Kangaroo Mother Care in very low birth weight infants. Indian Netw 1989; 7: 43-51.
J Pediatr 2001; 68 : 1019-1023. 20. Tessier R, Cristo M, Velez S et al. Kangroo mother care and the
13. Rao PN, Udani R, Nanavati R. Kangaroo mother care for low bonding hypothesis. Pediatrics 1989; 102: 17-23.
birth weight infants: a randomized controlled trial. Indian 21. Brazelton TB. Early intervention. In: Fritzgerald HE, Lester
Pediatr 2008; 45 : 17-23. BM, Yogman MV, ed. Theory and research in behavioral
14. Conde-Agudelo A, Diaz Rossello JL, Belizan JM. Kangaroo pediatrics. New York; Plenum Press; 1982; 104-119.
mother care to reduce morbidity and mortality in low 22. Korones SB. Disturbance and infants rest. In Moore TD, ed.
birthweight infants. Cochrane Database Syst Rev 2003: Iatrogenic problems in intensive care. Report of 69 th . Ross
CD002771. conference on Pediatric research. Columbus, OH: Ross
15. Bergman NJ, Jurisoo LA. The kangaroo-method for treating laboratories; 1976; 94-97.
low birth weight babies in a developing country. Trop Doct 23. Mann NP, Haddow R, Stokes L, Goodley S, Rutter N. effect of
1994; 24 : 57-60. night and day on preterm infants in newborn nursery:
16. Bergman NJ, Linley LL, Fawcus RR. Randomized controlled randomized trial. BMJ 1986; 293: 1265-1267.
trial of skin to skin contact from birth vs. conventional 24. Udani RH, Nanavati RN. Training manual on Kangaroo mother
incubator for physiological stabilization in 1200to 2199 gram care. Published by the Department of neonatology. KEM
newborns. Acta Pediatr 2004; 93: 779-885. Hospital and Seth GS medical college Mumbai. September
17. Charpak N, Ruiz-Pelaez JG, Figueroa de CZ, Charpak Y. 2004.
Kangaroo mother versus traditional care for newborn infants 25. Website of KMC India Network. Guidelines for parents and
</=2000 grams: a randomized, controlled trial. Pediatrics 1997; health providers are available online at www.kmcindia.org
100 : 682-688. 26. World Health Organization. Kangaroo mother care: a practical
18. Kadam S, Binoy S, Kanbur W, Mondkar JA, Fernandez A. guide. Department of Reproductive Health and Research,
Feasibility of kangaroo mother care in Mumbai. Indian J WHO, Geneva.2003.

Indian Journal of Pediatrics, Volume 75May, 2008 503


151
152
153
154
155 95

Symposium on AIIMS Protocols in Neonatology III

Minimal Enteral Nutrition


Satish Mishra, Ramesh Agarwal, M. Jeevasankar, Ashok K. Deorari and Vinod K. Paul

WHO Collaborating Centre for Training & Research in Newborn Care, Division of Neonatology, Department of
Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India

ABSTRACT
Although parenteral nutrition has been used widely in the management of sick very low birth weight infants, a smooth transition
to the enteral route is most desirable. Trophic feeding is the practice of feeding small volume of enteral feeds in order to
stimulate the development of the immature gastrointestinal tract of the preterm infant. This practice has also been termed as
minimal enteral nutrition (MEN). MEN improves gastrointestinal enzyme activity, hormone release, blood flow, motility and
microbial flora. Clinical benefits include improved milk tolerance, greater postnatal growth, reduced systemic sepsis and shorter
hospital stay. There is currently no evidence of any adverse effects following MEN. MEN can be commenced in neonates on
ventilation and total parenteral nutrition. A protocol of giving MEN has been presented here. [ Indian J Pediatr 2008;
75(3) : 267-269] E-mail: ashokdeorari_56@hotmail.com

Key words : Very low birth weight infant; Minimal enternal nutrition; Expressed breast milk

Because of concern that oral feedings might increase the preterm neonates may be partly responsible for the
risk of necrotizing enterocolitis (NEC), some high-risk common problems of feed intolerance encountered in
infants have received prolonged period of total parenteral these newborns.
nutrition (TPN) without any enteral feedings. However,
Minimal enteral nutrition (MEN)
lack of enteral nutrients may diminish gastrointestinal
functional and structural integrity by diminishing This is a practice wherein some minute volumes of feeds
hormonal activity, growth of intestinal mucosa, lactase are given to the baby in order to stimulate the
activity, nutrient absorption, or motor maturation. These development of the immature gastrointestinal tract of the
problems may then compromise later feeding tolerance preterm infant. Studies have shown that neonates who
and growth, and thus prolong the hospital stay. The were fed earlier with minimal feeds had lower episodes of
practice of providing minimal enteral nutrition (MEN) or feeding intolerance and gained weight faster as compared
trophic feedings (small volume feedings that provide to neonates who were fed late.2-4 These feeds are of small
minimal calories) for some period after birth was volume ranging from 10 to 15mL/Kg/day and not
developed as a strategy to enhance the functional intended for providing adequate calories. Although MEN
maturation of the gastrointestinal tract and to facilitate a does not provide sufficient calories for growth, it is
smooth and rapid transition from parenteral to enteral beneficial as it exerts a trophic effect on the gut mucosa.
nutrition .
Benefits of MEN
Problems of delayed feeding
Animal studies have shown a 2-3 fold increase in
In several animal species, absence of enteral nutrients is intestinal mucosa mass with early feeding. The trophic
associated with diminished intestinal growth, atrophy of effect on intestinal mucosa may be mediated via various
intestinal mucosa, delayed maturation of intestinal growth factors in human milk. These include insulin,
enzymes, and increase in permeability and bacterial epidermal growth factor and other peptides known to
translocation. A lack of enteral nutrients also affects exert direct trophic effects. Premature infants receiving
intestinal motility, perfusion, and hormonal responses.1 It MEN had cumulative greater milk intake, which was
is possible that a prolonged delay in starting feeds in associated with lower serum alkaline phosphatase
activity. 5 Intestinal motility pattern matures more rapidly
in premature infants receiving early enteral feeding. 4
Correspondence and Reprint requests : Dr. Ashok K Deorari, Investigators have demonstrated that trophic feeds were
Professor, Department of Pediatrics, All India Institute of Medical associated with greater absorption of calcium and
Sciences, Ansari Nagar, New Delhi 110029, India. phosphorus, greater lactase activity, and reduces
[Received February 7, 2008; Accepted February 7, 2008] intestinal permeability.

Indian Journal of Pediatrics, Volume 75March, 2008 267


156 96

S. Mishra et al

Besides the effect on intestinal mucosa, other beneficial following MEN.9


responses to MEN include: (a) motor effects: which
Indications for MEN
include better peristaltic activity and coordinated motor
activity in response to feeding. (b) vascular effects: which All preterm infants especially 32 weeks of gestation, in
include post-prandial decrease in splanchnic vascular whom enteral feeding has not yet been started due to
resistance. This decrease would be associated with an underlying illness, should be commenced on MEN.
increase in intestinal blood flow and oxygen uptake in
Contraindications
response to feeding. (c) endocrine: effects which include
the significant increases in plasma concentrations of MEN should be avoided in infants with severe
enteroglucagon, gastrin, and gastric-inhibiting hemodynamic instability, suspected or confirmed NEC,
polypeptide in preterm infants after milk feedings of as evidence of intestinal obstruction perforation or
little as 12 mL/kg per day. Similar surges in the trophic paralytic ileus. Mechanical ventilation and or use of
hormones were not seen in intravenously nourished umbilical catheters are not contraindication to using
infants. 6 MEN
Clinical benefits of MEN Protocol for MEN
Cochrane meta-analysis on studies evaluating the clinical MEN should be commenced after ensuring hemodynamic
effects of MEN done by Tyson et al,7 has showed that, stability in preterm neonates. This is usually possible by
among infants given trophic feedings, there was an day 2-3 in sick preterm neonates. However MEN may be
overall reduction in mean days to full feeding (weighted started on the first day itself in hemodynamically stable
mean difference [WMD] = -2.6 days; number of days that neonates if no contraindications to feeding exist.
feeding was withheld (WMD = -3.1 days; and total Expressed human breast milk is the preferred milk for
hospital stay (WMD = -11.4 days) compared to infants MEN. Commercial formulas are best avoided for MEN.10
given no enteral nutrient intake. There was no significant Recommended volume is 10-15 mL/Kg/day. This should
increase in the risk of necrotizing enterocolitis (relative be divided into equal aliquots and administered by
risk = 1.16 [0.75, 1.79]). McClure et al, 8 have shown a gavage feeding in a 3-6 hourly schedule. In preterm very
reduction in episodes of culture confirmed sepsis and low birth weight infants born with antenatal diagnosis of
faster weight gain in babies receiving minimal enteral altered in umbilical arterial blood flow (reversed or
nutrition. However, conclusive evidence in favor of MEN absent end diastolic flow), MEN can possibly be delayed
is not yet available due to the small size and number of for 2 to 3 days. Advancement of feeds should also be slow
studies on this issue. Nonetheless, due to obvious and carefully monitored in these infants.
beneficial effects (Table 1), MEN has become a widely
used practice in neonatal intensive care units. Monitoring

Adverse effects of MEN The infant should be monitored for any evidence of feed
intolerance including abdominal girth, gastric residuals or
There is currently no evidence of any adverse effects clinical signs of NEC. If the abdominal girth has increased
by 2 cm, gastric residual volume (GRV) should be
TABLE 1. Advantages of Gastrointestinal Priming with MEN1
checked. Feeding should be stopped in the presence of
1. Shortens time to regain birth weight significant aspirate (>25% of feed or >3mL whichever is
2. Improves feeding tolerance more) and/or bilious or blood stained aspirates.
3. Enhances enzyme maturation
4. Improves gastrointestinal motility Progression to full feeds As baby gains clinical
5. Improves mineral absorption, mineralization stability, feeds are advanced at the rate of 20-30 mL/Kg/
6. Lowers incidence of cholestasis day. Baby is monitored as mentioned above and volume
TABLE 2. Protocol on Minimal Enteral Nutrition (MEN)* of feeds increased gradually to full enteral feeds.

For whom
All preterm infants especially those less than 32 weeks gestation, who are hemodynamically stable but cannot be given enteral feeds

What to feed
Preferably expressed breast milk (EBM)

How much to feed


EBM 8-12 mL/kg/day divided into 4-8 feeds given by gavage feeding

For <1000g 1-2ml every 4-6 hour; for 1000 g 2-3 ml every 2-4 hour;
*Can be started while baby is on ventilator and /or receiving total parenteral nutrition
*In severe birth asphyxia, MEN should be started after 48-72 hours
*VLBW infants born with antenatal diagnosis of altered umbilical arterial blood flow (reversed or absent end diastolic flow), MEN can
possibly be delayed for 2 to 3 days.

268 Indian Journal of Pediatrics, Volume 75March, 2008


157 97

Minimal Enteral Nutrition

In view of profound clinical benefits routine practice of 4. Berseth CL. Effect of early feeding on maturation of the
minimal enteral nutrition should be encouraged in preterm infants small intestine. J Pediatr 1992; 120 : 947-953.
5. Klagsbrun M. Human milk stimulates DNA synthesis and
developing countries using expressed breast milk for
cellular proliferation in cultured fibroblasts. Proc Natl Acad Sci
VLBW infants. USA 1978; 75 : 5057-5061.
6. Berseth CL. Minimal enteral feedings. Clin Perinatol 1995; 22:
REFERENCES 195-205.
7. Tyson JE, Kennedy KA. Trophic feeds for parenterally fed
infants. Cochrane Database Syst Rev 2005.
1. Schanler RJ. Enteral nutrition for high risk neonates. In
8. McClure RJ, Newell SJ. Randomized controlled study of
Ballard RA, ed. Averys Diseases of the Newborn, Philadelphia;
clinical outcome following trophic feeding. Arch Dis Child Fetal
WB Saunders, 2005: 1044.
Neonatal 2000; 82 : F29-F33.
2. Dunn L, Hulman S, Weiner J, Kliegman R. Beneficial effects of
9. McClure RJ. Trophic feeding of the preterm infant. Acta Pediatr
early hypocaloric enteral feeding on neonatal gastrointestinal
2001; 90 : 19-23.
function. Preliminary report of a randomized trial. J Pediatr
10. Troche B, Harvey-Wilkes K, Engle WD, Nielsen HC, Frantz
1988; 112 : 622-629.
ID, Mitchell ML, Hermos RJ. Early minimal feedings promote
3. Slagle Ta, Gross SJ. Effect of early low volume enteral
growth in critically ill premature infants. Biol Neonate 1995; 67:
substrate on subsequent feeding tolerance in very low birth
172-181.
weight infants. J Pediatr 1988; 113 : 526-531.

Indian Journal of Pediatrics, Volume 75March, 2008 269


158
159
160
161
162 98

Symposium on AIIMS Protocols in Neonatology III

Patent Ductus Arteriosus in Preterm Neonates


Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar,
New Delhi, India

ABSTRACT
Failure of the ductus arteriosus to close within 48-96 hours of postnatal age results in a left to right shunt across the ductus
and overloading of the pulmonary circulation. This is more likely to happen in premature neonates with respiratory distress
syndrome. Deterioration in the respiratory status on day 3-4 in a ventilated neonate and unexplained metabolic acidosis may
be the earliest indicators of a patent ductus arteriosus (PDA). Indomethacin is the main stay of medical management of PDA
in preterm neonates. Guidelines for administration of indomethacin have been described in the protocol. Restricted fluid
therapy may be beneficial in the prevention of PDA in preterm neonates. Presence of PDA in a term neonate should be
investigated to rule out an underlying congenital heart disease. [Indian J Pediatr 2008; 75 (3) : 277-280] E-mail:
ashokdeorari_56@hotmail.com

Key words : Preterm; Patent ductus arteriosus; Indomethacin; Ibuprofen

During intrauterine life, 10% of the cardiac output passes Ductus arteriosus in preterm neonates
through the lungs. The remaining 90% is shunted via the
The normal mechanisms of ductal closure fail to work in
ductus arteriosus (DA) to the aorta and systemic
preterm neonates. The various factors contributing to an
circulation. After birth, most of the right ventricular
increased incidence of PDA in preterms include: (a)
output should pass through the lungs to facilitate proper
Increased sensitivity of the ductus to prostaglandins as
gas exchange. In order to make this possible, the ductus
compared to term neonates (b) Sensitivity to
undergoes constriction and functional closure soon after
prostaglandins is sustained for a longer period (c) Higher
birth in term neonates. Eighty percent (80%) of the DA in
incidence of hypoxia and acidosis (d) Defective smooth
term infants close by 48 hours and nearly 100% by 96
muscle migration resulting in compromised anatomical
hours. Failure of this normal closure results in problems
closure.
especially in preterm neonates.
Hemodynamic consequences of PDA
PHYSIOLOGY Shunting of blood from the systemic circulation to the
pulmonary circulation results in congestive cardiac
failure, which manifests clinically with wide pulse
Oxygen and endothelin are very strong vasoconstrictors
pressure and bounding pulses. Overloading of the
and prostaglandins E2 and I2 are strong vasodilators of the
pulmonary vasculature leads to pulmonary edema/
DA. Lower oxygen concentrations in utero and high
hemorrhage which predisposes the neonate to chronic
circulating PgE2 and PgI2 levels help in keeping the ductus
lung disease. Blood flow to the kidney and
patent. Sudden elevation in circulating oxygen tension
gastrointestinal tract is compromised predisposing to
and fall in prostaglandin levels soon after delivery results
acute renal failure (ARF) and necrotizing enterocolitis
in strong vasoconstriction and functional closure of the
(NEC). Hypo-perfusion followed by reperfusion increases
DA soon after delivery. It is believed that this action of
the risk of intraventricular hemorrhage (IVH).
oxygen is mediated via the formation of the endothelin
molecule. This functional closure is followed by Risk factors for PDA in preterm neonates
anatomical closure in the next 1-3 months.
The incidence of PDA is inversely related to gestational
age and birth weight. A hemodynamically significant
shunt due to PDA has been reported in 40% of infants less
Correspondence and Reprint requests : Dr Ashok K. Deorari, than 1000 grams and 20% of infants between 1000-1500
Professor, Department of Pediatrics, All India Institute of Medical grams.1-3 Respiratory distress syndrome (RDS) in preterm
Sciences, Ansari Nagar, New Delhi 110029, India. neonates needing ventilation and surfactant is an
[Received February 7, 2008; Accepted February 7, 2008]

Indian Journal of Pediatrics, Volume 75March, 2008 277


163 99

R. Agarwal et al

additional risk factor for PDA. Prophylactic use of severity of the shunt across the PDA.
synthetic surfactant has been associated with an increased
risk of PDA. Lack of antenatal steroids, presence of sepsis,
and liberal fluid therapy are other risk factors for STRATEGIES OF MANAGEMENT5
developing PDA.

Prophylactic treatment: In this strategy, treatment is


CLINICAL FEATURES started before the appearance of PDA, usually within the
first 24 hours of birth. Indomethacin has been tried as
prophylactic treatment in very preterm neonates,
Hyperdynamic circulation : A wide pulse pressure (>25 mm
especially <1000 grams. Although it was found to
Hg), prominent precordial pulsations and bounding
decrease symptomatic PDA and IVH, it was not
pulses and an ejection systolic murmur (occasionally pan
associated with any change in long-term morbidity and
systolic and continuous murmur) heard best at the 2nd left
neuro-developmental outcome. More over, indomethacin
parasternal area 4 are usually present on clinical
has also been found to decrease cerebral and renal blood
examination.
flow and hence is not recommended as a prophylactic
Indicators of ductus opening on a ventilated baby: Metabolic agent in the prevention of PDA6. Trials with prophylactic
acidosis not attributable to hypoperfusion and sepsis, ibuprofen are still going on7.
deteriorating respiratory status on day 3-4 after a period
Early Symptomatic: In this strategy, treatment is started
of relative stability, increasing ventilatory requirements
as soon as the PDA is detected even if it is not
on day 3-4, unexplained CO2 retention, fluctuating FiO2
hemodynamically significant.
requirements and recurrent apneas in a ventilated baby
should raise clinical suspicions of a symptomatic PDA. Weight <1000 grams: Among neonates detected to
have a PDA, 80% of neonates would progress to develop
Studies have revealed that echocardiographic criteria
a hemodynamically significant shunt. Hence , it is
of a significant left to right shunt usually precede clinical
recommended to treat PDA in this group early even
symptoms by an interval of 2-3 days. However, clinical
though it may not be hemodynamically significant.
features have a better correlation with long-term
However, in view of the recent evidence, the risk and
morbidity and available evidence does not recommend
benefits must be weighed before treating asymptomatic
routine screening with echocardiography for at-risk
babies.8
neonates.3
Weight >1000 grams: Early treatment is not
Differential Diagnosis
recommended in this group as progression to
(i) AV fistula, (ii) Ruptured sinus of valsalva, (iii) Aorto- symptomatic PDA is less common and spontaneous
pulmonary window. closure are known to occur in this group 8 .
Late Symptomatic : Only hemodynamically significant
PDA is treated in this strategy and it is the recommended
INVESTIGATIONS
approach for neonates >1000 grams.

Chest X-ray: Radiographic findings are non-specific for


diagnosis of PDA. They include cardiomegaly, upturned MANAGEMENT
left bronchus due to left atrial enlargement and
pulmonary plethora.
(i) General measures
Echocardiography : Echocardiography is not
Fluid restriction: 60% of maintenance fluids
recommended routinely for all preterm neonates. A
clinical diagnosis of PDA should preferably be confirmed TABLE Dosage of Indomethacin and Ibuprofen for Closure of
by echo prior to starting medical therapy. Patent Ductus Arteriosus
Echocardiographic criteria include (a) Left atrial dilatation Indomethacin
(Left atrial: Aortic root>1.6) (b) Diastolic turbulence Initial Dose
(backflow) on doppler in the pulmonary artery and (c) 0.2 mg/kg stat followed by age adjusted doses:
Direct imaging to measure the diameter of PDA. Subsequent dose
< 2 day- 0.1 mg/kg/dose 12 hourly for 2 doses
A hemodynamically significant PDA is diagnosed in 2-7 day- 0.2 mg/kg/dose 12 hourly for 2 doses
the presence of a ductus diameter >1.5mm and absent/ > 7 day- 0.25 mg/kg/dose 12 hourly for 2 doses
retrograde diastolic flow in the post-ductal aorta. Doppler Ibuprofen
echo is more specific and sensitive for the diagnosis of 10 mg/kg stat followed by
PDA while an M mode echo is useful for assessing the 5 mg/kg/dose 24 hourly for 2 doses

278 Indian Journal of Pediatrics, Volume 75March, 2008


164 100

Patent Ductus Arteriosus in Preterm Neonates

Avoid hypoxia and acidosis enlarging or evolving intraventricular hemorrhage (IVH);


platelet count < 60,000/mm3
High peek end expiratory pressure (PEEP) and lower
inspiratory time (Ti), if otherwise appropriate9. Gastrointestinal: necrotizing enterocolitis; blood in stool
Frusemide is generally not required. Frusemide Efficacy
increases prostaglandin levels, which may interfere
The closure rate with indomethacin is 80%. The efficacy is
with the therapeutic effect of indomethacin. However
unaffected by gestation or birth weight. The full course
enough evidence for this concern is not yet available10.
should be completed even if closure is achieved before the
In intractable CHF, we occasionally use frusemide in a
third dose. Longer treatment with lower doses (0.1 mg/
dose of 1 mg/kg/dose 12 hourly. Careful evaluation of
kg/dose for 6 doses) has similar toxicity and efficacy and
hydration status is essential before and during
are not recommended.12
treatment with frusemide.
Ibuprofen is also an inhibitor of prostaglandin synthesis
Digoxin has no role in management of PDA.
and is effective in closing the ductus. It has an equal
(ii) Medical Management efficacy as compared to indomethacin with fewer side
effects.13 Its use is associated with a lower incidence of
(a) Indomethacin
oliguria and renal compromise. It has been found to have
Mechanism of action: The postulated mechanism of action a lesser effect on mesenteric and cerebral blood flow as
is an inhibition of cyclo-oxygenase (COX) enzyme in the compared to indomethacin. The dose is 10 mg/kg stat
prostaglandin pathway. Indomethacin has a greater followed by 5 mg/kg/dose x 2 doses at 24 hour intervals
affinity for COX 1 (renal) as against COX 2 (extra-renal). given orally.
Due to this greater affinity for renal COX1, the incidence
of renal complications is higher with indomethacin as
compared to other inhibitors of prostaglandin synthesis.
Indications for indomethacin use:
Early symptomatic treatment of PDA in <1000 grams
Late symptomatic treatment of PDA in > 1000 grams *
Re-treatment after failure of the first course of
Indomethacin.
Recurrence of PDA after the first course of
indomethacin.
Oral medication: Due to non-availability of the IV
formulation, we have been using oral indomethacin for
closure of PDA. Oral indomethacin is available as 25 mg
tablets and it can be broken into lower doses. We usually
mix the dose with 2-5 ml expressed breast milk and
administer it through the oro-gastric tube.
Side effects and monitoring: Adverse effects include renal
compromise due to its effect on COX 1, bleeding tendency
due to its effect on platelet function and increased risk of
necrotizing enterocolitis. Decreased cerebral blood flow
associated with the use of indomethacin may be
associated with poor neuro-developmental outcome.
Various parameters that should be monitored include (a)
Urine output: If urine output falls below 1 ml/kg/hr low
dose dopamine at 1-2 mg/kg/min for its renal effect may
be used 11 (b) Renal function (alternate days) and (c)
Platelet counts (daily)
Contraindications
Renal: Urine output< 0.6 ml/kg/h, blood urea>30 mg/dl,
creatinine>1.8 mg/dl
*Risk benefit must be weighed for asymptomatic growth
Bleeding: Bleeding from IV sites; gastrointestinal bleeding, retended baby16

Indian Journal of Pediatrics, Volume 75March, 2008 279


165 101

R. Agarwal et al

(b) Surgical ligation : The indications for surgical therapy Surgical ligation should be considered if PDA recurs
include a contraindication to medical therapy and failure after the second course or if contraindications to
of a second course of indomethacin. Surgical ligation may indomethacin/ ibuprofen exist.
be carried out by Thoracotomy or Video assisted Both ibuprofen and indomethacin may be used
thoracoscopy.14 orally.
(c) Restricted fluid intake for prevention of PDA These recommendations do not apply to PDA in term
15
Bell et al have conducted a meta-analysis on studies neonates. Term neonates with PDA should not be treated
evaluating fluid therapy and have shown that restricted with indomethacin/ ibuprofen and should have a
fluid intake in the initial 4-5 days of life is associated with detailed echo to rule out an underlying congenital heart
a lower incidence of PDA. In our unit, we usually start defect. They would require surgery for closure of an
with 60 ml/kg in babies weighing between 1000-1500 isolated patent ductus arteriosus.
grams and 80 ml/kg for babies between 750-1000 grams.
We try to maintain serum sodium of 135-145 meq/L, REFERENCES
urine output of 1-3 ml/kg/hr and a urine specific gravity
of 1.005 to 1.012. We use thin transparent plastic barriers 1. Wechsler SB, Wernovsky G. Cardiac disorders. In Cloherty JP,
Stark AR, eds. Manual on neonatal care, 6th ed. 2008; 388-435.
(clingwrap) to create a microenvironment around the
2. Archer N. Cardiac diseases. In Rennie JM, Roberton NRC, eds.
baby in order to decrease insensible water losses. We Textbook of neonatal care, 3rd ed. Churchill Livingstone, 1999;
would allow a weight loss of at least 2-3% per day and 687-689.
generally do not exceed a fluid intake of 150-160 ml/kg/ 3. Clyman RI. Patent ductus arteriosus in preterm neonates. In
day by day 5-7 day of life. Using this scheme of fluid Taeush HW, Ballard RA, eds. Averys diseases of the newborn,
8th ed. WB Saunders; 2005; 816-826.
therapy, we have achieved a low incidence of PDA in our 4. Zahaka KG, Patel CR. Congenital cardiac defects. In Fanaroff
unit. AA, Martin RJ, eds. Neonatal perinatal medicine Disorders of the
fetus and infants, 6 th ed. 1997; 1155-1157.
GUIDELINES FOR TREATMENT 5. Clyman RI. Treatment of premature neonates with patent
ductus arteriosus: analysis of four treatment strategies. J
Pediatr 1996; 128 : 601-607.
Preterm babies are at risk of developing 6. Foulie WP. Prophylactic Indomethacin - systematic review
hemodynamically significant PDA. and meta-analysis. Arch Dis Child 1996; 74 : F81-F87.
7. De Carolis MP, Romagnoli C, Polimeni V, Piersigilli F, Zecca
Prematurity, lack of antenatal steroids, RDS and E, Pappacci P et al. Prophylactic ibuprofen therapy of patent
sepsis increase the risk of PDA. ductus arteriosus in preterm infants. Eur J Pediatr 2000; 159 :
At-risk preterm neonates should be monitored for 364-368.
8. Bose CL, Laughan MM. Patent ductus arteriosus: Lack of
clinical features suggestive of PDA.
evidence for common treatments. Arch Dis Child Fetal Neonatal
Echocardiography should be used to confirm a 2007; 92 : F498-F502.
clinical suspicion of PDA. In the absence of an echo, 9. Vanhaesebrouck S, Zonnenberg I, Vandervoot P et al.
treatment may be started with a clinical diagnosis. Conservative treatment for patent ductus arteriosus in the
preterm. Arch Dis Child Fetal Neonatal Ed 2007; 92 : F244-F247.
Babies <1000 grams with a diagnosis of PDA 10. Brion LP, Campbell DE. Furosemide in indomethacin treated
(irrespective of hemodynamic effect) should be infants - systematic review and meta-analysis. Pediatr Nephrol
treated. Risk benefit must be weighed specially in 1999; 13 : 212-218.
11. Fajardo CA, Whyte RK, Stele BT. Effect of dopamine on
growth retarded babies with absent or reverse
failure of indomethacin to close patent ductus arteriosus. J
diastolic blood flow.16 Pediatr 1992; 121 : 771-775.
Babies >1000 grams with a diagnosis of PDA should 12. Tammela O, Ojala R, Livainen T, Lautamatti V, Pokela ML,
be treated only if a hemodynamically significant left Janas M et al. Short versus prolonged indomethacin therapy
for patent ductus arteriosus in preterm infants. J Pediatr 1999;
to right shunt is present.
134: 552-557.
Prophylactic treatment with indomethacin is not 13. Van Overmeire B, Smets K, Lecoutere D, Van de Broek H,
recommended. Weyler J, Degroote K et al. A comparison of ibuprofen and
indomethacin for closure of patent ductus arteriosus. N Engl
Platelet counts and renal parameters should be J Med 2000; 343 : 674-681.
checked prior to starting therapy with indomethacin 14. Burke RP, Jacobs JP, Cheng W, Trento A, Fontana GP. Video-
and repeated after 24 hours. assisted thoracoscopic surgery for patent ductus arteriosus in
low birth weight neonates and infants. Pediatrics 1999; 104:
A second course of indomethacin should be tried 227-230.
after failure of the first course or recurrence after the 15. Bell EF, Acarregui MJ. Restricted versus liberal water intake
first course. for preventing morbidity and mortality in preterm infants.
Cochrane Database Syst Rev 2000; 2 : CD000503.
Preliminary studies seem to suggest that ibuprofen is
16. Mc Namara, Sehgal A. Towards rational management of the
a safe alternative to indomethacin in the treatment of patent ductus arteriosus: the need for disease staging. Arch Dis
PDA. Child Fetal Neonatal 2007; 92: F425-F427.

280 Indian Journal of Pediatrics, Volume 75March, 2008


166
167
168
169
170 102

Symposium on AIIMS Protocols in Neonatology IV

Perinatal HIV
Sunil Saharan, Rakesh Lodha, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, India

ABSTRACT
HIV pandemic is one of the most serious health crises the world faces today. Approximately 5-10% of all cases of HIV are
children. Majority of children acquire infection through mother-to-child transmission either during pregnancy, delivery, or by
breast-feeding. MTCT can be reduced to <2% by antiretroviral prophylaxis to women during pregnancy and labour and to the
infant in the first weeks of life, obstetrical interventions including elective cesarean delivery and complete avoidance of
breastfeeding. Guidelines for postnatal diagnosis of HIV infection, feeding, immunization and administration of cotrimoxazole
prophylaxis have been described in the protocol. [Indian J Pediatr 2008; 75 (4) : 359-362] E-mail: aranag@rediffmail.com

Key words : HIV; Pregnancy; Perinatal; Mother-child transmission; Neonate

The human immunodeficiency virus (HIV) pandemic is


Abbreviations
one of the most serious health crises the world faces
ART - Antiretroviral therapy
today. AIDS has killed more than 25 million people since AZT - Zidovudine
1981 and an estimated 33.2 million people are now living Sd NVP - Single dose nevirapine
with HIV, about 2.5 million of whom are children.1 In 2007
alone, an estimated 420,000 children were newly infected
with HIV.1 In India it is estimated 2.47 million (2 - 3.1 Maternal factors: Advanced disease (low CD4 count,
million) people were living with HIV infection in year symptoms of AIDS), primary infection of mother
2006. Adult prevalence of HIV infection in our country is during pregnancy, first of twins, rupture of
0.36% (0.27-0.47%). The prevalence rate for females is 0.29 membranes more than four hours, maternal bleeding,
percent, while for males it is 0.43 percent. Prevalence of mother not on antiretroviral therapy, vaginal delivery,
HIV in pregnant women population was observed to be other sexually transmitted diseases, isolated HIV-1
0.60%.2 infection

Mode of transmission Fetoplacental factors: chorioamnionitis, placenta previa,


prematurity (increased peripartum transmission)
Most children living with HIV acquire the infection
through mother-to-child transmission (MTCT). HIV Infant factors: HLA concordance with mother
infection can be transmitted from an infected mother to Postnatal factors: breast feeding, higher breast milk
her fetus during pregnancy, during delivery, or by breast- virus load, mastitis or maternal nipple lesions,
feeding. HIV can be transmitted to the fetus as early as the maternal seroconversion during breastfeeding, infant
first and second trimester of pregnancy. However, having thrush at less than six month age (in
maternal transmission to the fetus occurs most commonly breastfeeding infant)
in the perinatal period.
Prevention of perinatal HIV
Risk factors for perinatal HIV transmission3
In the absence of any intervention, the risk of perinatal
Viral factors: High viral load, non syncytium inducing transmission is 1530% in non-breastfeeding
phenotype, HIV 1 population.
Breastfeeding by an infected mother increases the risk
by 520% to a total of 2045%.4
Correspondence and Reprint requests : Dr. Ramesh Agarwal, The risk of MTCT can be reduced to less than 2% by
Assistant Professor, Department of Pediatrics, All India Institute of interventions that include antiretroviral (ARV)
Medical Sciences, Ansari Nagar, New Delhi, India prophylaxis given to women during pregnancy and
[Received March 17, 2008; Accepted March 17, 2008]

Indian Journal of Pediatrics, Volume 75April, 2008 359


171 103

S. Saharan et al

labour and to the infant in the first weeks of life,5 medically indicated.
obstetrical interventions including elective cesarean Delivery by elective cesarean section at 38 weeks
delivery (prior to the onset of labour and rupture of before onset of labour and rupture of membranes.
membranes), 6 and complete avoidance of
Avoid procedures increasing risk of exposure of child
breastfeeding.7
to maternal blood and secretions like use of scalp
ARV regimens for treating pregnant women8 electrodes.
Refer to Fig (1A) Breast feeding
Recommended regimen for pregnant women with Breast-feeding is an important modality of
indication for antiretroviral therapy (ART) is transmission of HIV infection in developing countries.
combination of zidovudine (AZT), lamivudine (3TC)
The risk of HIV infection via breast-feeding is highest
and nevirapine (NVP) in antepartum, intrapartum
in the early months of breast-feeding.9
and postpartum period.
Factors that increase the likelihood of transmission
Recommended regimen for pregnant women who are include detectable levels of HIV in breast milk, the
not eligible for ART, but for preventing MTCT is: presence of mastitis and low maternal CD4+ T cell
Antepartum: AZT starting at 28 weeks of count.
pregnancy Exclusive breast-feeding has been reported to carry a
Intrapartum: a combination of single dose (Sd) lower risk of HIV transmission than mixed feeding.7
NVP, AZT and 3TC According to current UN recommendations (WHO,
Postpartum: a combination of AZT and 3TC for 7 2001) when replacement feeding is acceptable,
days. feasible, affordable, sustainable and safe (AFASS),
avoidance of all breastfeeding by HIV-infected moth-
Recommended regimen for pregnant women who are ers is recommended. Otherwise exclusive
in labour and have not received ART is: breastfeeding is recommended during the first
Intrapartum: a combination of Sd-NVP, AZT and months of life.
3TC WHO recommends that the transition between
Postpartum: a combination of AZT and 3TC 7 exclusive breastfeeding and early cessation of
days breastfeeding should be kept as short as possible, -
Omission of the Sd-NVP for the mother may be early and abrupt cessation - bearing in mind that
considered for women who receive at least four weeks mixed feeding during this period carries a 70% greater
of AZT before delivery. risk of MTCT.10
When Sd-NVP is used to prevent MTCT, either alone Replacement feeding should be given by katori spoon.
or in combination with AZT, the rationale of giving
Postnatal Diagnosis of HIV Infection11
AZT and 3TC intrapartum and for seven days
postpartum is to prevent resistance to NVP. Refer to Fig. 2
ARV regimens for infants born to HIV positive In children younger than 18 months, diagnosis of HIV
mothers8 infection is based on: a positive virological test at 6
weeks for HIV or its components (HIV-RNA or HIV-
Refer to Fig. 1(B)
DNA or HIV p24 antigen) confirmed by a second
The recommended regimen for infants is Sd-NVP + virological test obtained from a separate
AZT for one week. determination taken more than four weeks after the
When delivery occurs within two hours of a woman first one.12
taking Sd-NVP, the infant should receive Sd-NVP
immediately after delivery and AZT for four weeks. If an infant or child is breastfeeding, he or she remains
at risk of acquiring HIV infection throughout the
If the mother receives less than four weeks of
breastfeeding period. Virological assays for HIV
antenatal ART, then four weeks rather than one week
infection should be conducted at least six weeks or
of AZT is recommended for the infant.
more after the complete cessation of breastfeeding to
Zidovudine 4 mg/kg/dose, Nevirapine 2 mg/kg rule out HIV infection.

Intrapartum interventions Antibody testing is not recommended for definitive or


confirmatory diagnosis of HIV infection in children
Avoid artificial rupture of membrane ARM unless until 18 months of age.13

360 Indian Journal of Pediatrics, Volume 75April, 2008


172 104

Perinatal HIV

Note:
If the mother receives less than four weeks of AZT before delivary, the AZT dose for the infant should be extended to four weeks
When delivery occurs within two hours of a woman taking Sd-NVP, the infant should receive Sd-NVP immediately after delivery
and AZT for four weeks

Fig. 1. Protocol on approach to (A) HIV infected mother (B) Baby born to HIV infected mother8

Fig. 2. Diagnostic approach for HIV exposed infants11

Indian Journal of Pediatrics, Volume 75April, 2008 361


173 105

S. Saharan et al

Co-trimoxazole prophylaxis mother with HIV infection. Pediatr Clin North Am 20004; 51:
909-937.
Co-trimoxazole prophylaxis is recommended for all 4. De Cock KM, Fowler MG, Mercier E et al. Prevention of
HIV-exposed infants under age 18 months starting at mother-to-child HIV transmission in resource-poor countries:
46 weeks of age or when first seen and continued translating research into policy and practice. JAMA 2000; 283:
1175-1182.
until HIV infection can be excluded.14
5. Lallemant M, Jourdain G, Le Coeur S et al. Single-dose
Co-trimoxazole prophylaxis is also recommended for Perinatal nevirapine plus standard zidovudine to prevent
mother-to-child transmission of HIV-1 in Thailand. N Engl J
a breastfeeding child of any age, continued until HIV
Med 2004; 351 : 217-228.
infection can be excluded following cessation of 6. Thome C, Patel D, Fiore S, Peckham C, Newell ML. Mother-
breastfeeding, with testing performed six weeks or to-child transmission of HIV infection in the era of highly
more after breastfeeding was stopped. active antiretroviral therapy. Clin lnfec Dis 2005; 40:458-465.
7. Magoni M, Bassani L, Okong P et al. Mode of infant feeding
In children < 6 month dose is 2.5 ml once a day and HIV infection in children in a program for prevention of
(trimethoprim 40 mg and sulphamethoxazole 200 mother-to-child transmission in Uganda. AIDS 2005; 19 : 433-
mg/5 ml) 437.
8. WHO. Antiretroviral drugs for treating pre~nant women and
Immunization15 preventing HIV infection in infants: towards universal access. 2006.
9. Dunn DT, Tess BH, Rodrigues LC, Ades AE. Mother-to-child
HIV exposed or infected but asymptomatic children transmission of HIV: implications of variation in maternal
should receive all standard vaccines as per national infectivity. AIDS 1998; 12 : 2211-2216.
schedule. 10. WHO. New data on the prevention of Mother-to-Child
Transmission of HIV and their policy implications. Conclusions
HIV infected children with immune suppression or and recommendations, Geneva, 2001.
symptoms should receive all standard vaccines except 11. WHO. Intiretroviral therapy of HIV infection in infants and
varicella, BCG and OPV vaccines. children: towards universal access. 2006.
12. Sherman GG, Cooper PA, Coovadia AH et al. Polymerase
Consider HiB and pneumococcal vaccines in all HIV chain reaction for diagnosis of human immunodeficiency
exposed children (irrespective of symptoms or CD4 virus infection in infancy in low resource settings. Pediatr
count) Infect Dis J 2005; 24 : 993-997.
13. Chantry CJ, Cooper ER, Pelton SI, Zorilla C, Hillyer GV, Diaz
C. Seroreversion in human immunodeficiency virus-exposed
REFERENCES but un infected infants. Pediatr Infect Dis J 1995; 14 : 382-387.
14. WHO. Guidelines on co-trimoxazole prophylaxis for HIV-related
infections among children, adolescents and adults in resource-
1. UNAIDS/WHO. AIDS Epidemic Update. December, 2007.
limited settings recommendations for a public health approach.
2. National AIDS Control Organization. HIV Sentinel
2006.
Surveillance and HIV estimation 2006. Available at: http://
15. Anonymous. Immunization in special circumstances. In Shah
www.nacoonline.org/upload/NACO%PDF/Note%20on%20
RC, Shah NK, Kukreja S (Eds). IAP Guide Book on
HIV%20Sentinel%Surveillance%20and20%HIV%20
Immunization, Mumbai; IAP Committee on Immunization,
Estimation_01%Feb%2008.pdf(accessed on 1/4/2008).
2005-2006; 52-54.
3. Havens P, Waters D. Management of the infant born to a

362 Indian Journal of Pediatrics, Volume 75April, 2008


174
175
176
177
178 106
Indian J Pediatr (2010) 77:11171121
DOI 10.1007/s12098-010-0177-z

SYMPOSIUM ON AIIMS PROTOCOLS IN NEONATOLOGY-I

Management of Polycythemia in Neonates


M. Jeeva Sankar & Ramesh Agarwal & Ashok Deorari &
Vinod K. Paul

Received: 27 July 2010 / Accepted: 2 August 2010 / Published online: 21 August 2010
# Dr. K C Chaudhuri Foundation 2010

Abstract Polycythemia is defined as a venous hemato- The viscosity of blood is directly proportional to the
crit above 65%. The hematocrit in a newborn peaks at hematocrit and plasma viscosity and inversely proportional
2 h of age and decreases gradually after that. The to the deformability of red blood cells. Symptoms of
relationship between hematocrit and viscosity is almost hypoperfusion correlate better with viscosity as compared
linear till 65% and exponential thereafter. Increased to hematocrit. Viscosity is, however, difficult to measure at
viscosity of blood is associated with symptoms of the bedside. Hyperviscocity is therefore suspected in the
hypo-perfusion. Clinical features related to hyperviscos- presence of an abnormally high hematocrit with or without
ity may affect all organ systems. Neonates born small for suggestive symptoms. Relationship between viscosity and
gestational age (SGA), infants of diabetic mothers hematocrit is almost linear up to a hematocrit of 65%
(IDM), and multiple births are at risk for polycythemia. and exponential thereafter [1, 2]. The polycythemia-
They should therefore undergo screening at 2, 12, and hyperviscocity syndrome is thus usually confined to infants
24 h of age. Polycythemia may be symptomatic or with hematocrits of more than 65%; it is very rare with
asymptomatic and guidelines for the management of both hematocrits of <60%.
types based on the current evidence are provided in the
protocol.
Definition
Keywords Polycythemia . Blood viscosity . Newborn .
Therapy A diagnosis of polycythemia is made in the presence of a
venous hematocrit more than 65% or a venous hemoglobin
Polycythemia or an increased hematocrit is associated concentration in excess of 22.0 g/dl. Hyperviscosity is
with hyperviscosity of blood. As the viscosity increases, defined as a viscosity greater than 14.6 cP at a shear rate of
there is an impairment of tissue oxygenation and perfusion 11.5 per second [3].
and a tendency to form microthrombi. Significant damage
may occur if these events occur in the cerebral cortex,
kidneys and adrenal glands. Hence, this condition requires Incidence
urgent diagnosis and prompt management.
The incidence of polycythemia is 1.5%4% of all live births
[4, 5]. The incidence is higher among both small for
M. J. Sankar : R. Agarwal : A. Deorari : V. K. Paul
gestational age (SGA) and large for gestational age (LGA)
Division of Neonatology, Department of Pediatrics,
All India Institute of Medical Sciences, infants. The incidence of polycythemia is 15% among term
Ansari Nagar, New Delhi, India SGA infants as compared to 2% in term appropriate for
gestational age (AGA) infants [6]. Neonates born at high
A. Deorari (*)
altitudes also have a higher incidence of polycythemia [1].
Department of Pediatrics, All India Institute of Medical Sciences,
Ansari Nagar, New Delhi 110029, India Polycythemia is unlikely to occur in neonates born at less
e-mail: ashokdeorari_56@hotmail.com than 34 week gestation.
179 107
1118 Indian J Pediatr (2010) 77:11171121

Table 1 Clinical features


ascribed to polycythemia and Central nervous system
hyperviscosity Early: Hypotonia and sleepiness, irritability, jitteriness , seizures and infarcts
Late: Motor deficits, lower achievement and IQ scores
Metabolism
Hypoglycemia
Jaundice
Hypocalcemia
Heart and lungs
Tachycardia, tachypnea, respiratory distress
Cyanosis, plethora
Chest radiography: cardiomegaly, pulmonary plethora
Echocardiography: increased pulmonary resistance, decreased cardiac output
Gastrointestinal tract
Poor suck, vomiting
Feed intoleranceabdominal distenstion
Necrotizing enterocolitis
Kidneys
Oliguria (depending on blood volume)
Transient hypertension
Renal vein thrombosis
Hematology
Mild thrombocytopenia
Thrombosis (rare)
Miscellaneous
Peripheral gangrene
Priapism
Testicular infarction

Physiological Changes in Postnatal Life high-risk neonates at 2 h of age. A normal value at 2 h of


age (hematocrit <65%) does not merit any further
Significant changes take place in the hematocrit from birth screening unless the infant is symptomatic. Hematocrit
through the first 24 h of life. The hematocrit peaks at 2 h of values >65% at 2 h of age merit repeat screening at 12 and
age and values up to 71% may be normal at this age [7, 8] 24 h. Any infant with clinical features suggestive of
It gradually declines to 68% by 6 h and usually stabilizes polycythemia should be investigated for the same.
by 12 to 24 h. The initial rise in hematocrit is related to a
transudation of fluid out of the intravascular space. Capillary vs. Venous Hematocrit

Capillary hematocrit measurements are unreliable and


Clinical Features highly subject to variations in blood flow. Capillary
hematocrits are significantly higher than venous hematoc-
Polycythemia can result in a wide range of symptoms involving rits. This difference is even more apparent in infants
several organ systems (Table 1). About 50% of neonates with
polycythemia develop one or more symptoms. However, most Table 2 Screening for Polycythemia
of these symptoms are non-specific and may be related to the
underlying conditions rather than due to polycythemia per se. Screening is recommended for the following:

(a) Small for gestational age (SGA) infants


(b) Infants of diabetic mothers (IDM)
Screening for Polycythemia
(c) Large for gestational age (LGA) infants
(d) Monochorionic twins especially the larger twin
Screening should be done for polycythemia in certain
(e) Morphological features of growth retardation.
high-risk groups (Table 2). We recommend screening in
180 108
Indian J Pediatr (2010) 77:11171121 1119

receiving large placental transfusion [9]. Capillary samples


Capillary hematocrit >65%
may be used for screening, but all high values should be
confirmed by a venous sample for the diagnosis of
polycythemia. Confirm with venous hematocrit

Methods of Hematocrit Analysis

The two available methods are: Exclude dehydration


Automated hematology analyzer: This calculates the Check weight loss
hematocrit from a direct measurement of mean cell
volume and the hemoglobin.
Micro-centrifuge: Blood is collected in heparinized
micro-capillaries (110 mm length and 12 mm
internal diameter) and centrifuged at 10,00015,000 Symptomatic Asymptomatic
revolutions per min (rpm) for 35 min. Plasma
separates and the packed cell volume is measured
to give the hematocrit. An automated analyzer gives
lower values as compared to hematocrits measured
by the centrifugation method [10]. Most of the PET PCV 65-70 PCV 70-75 PCV >75%
reported data on polycythemia is on centrifuged
hematocrits.

Monitoring Consider
hydration PET
Management
Actively look for
Before a diagnosis of polycythemia is considered, it is symptoms
mandatory to exclude dehydration. If the birth weight is
known, re-weighing the baby and looking for excessive PET: partial exchange transfusion
weight loss would help in the diagnosis of dehydration. Fig. 1 Algorithm for management of polycythemia
If this is present, it should be corrected by increasing
fluid intake. The hematocrit should be measured again
(a) Symptomatic Polycythemia
after correction of dehydration. Once a diagnosis of
polycythemia is made, associated metabolic problems
The definitive treatment for polycythemia is to perform a
including hypoglycemia should be excluded.
partial exchange transfusion (PET). PET involves removing
Management of polycythemia is dependent upon two
some of the blood volume and replacing it with fluids so as
factors (Fig. 1):
to decrease the hematocrit to a target packed cell volume of
1. Presence of symptoms suggestive of polycythemia 55%. Following partial exchange transfusion, symptoms
and/or like jitteriness may persist for 12 days despite the
2. Absolute value of hematocrit hematocrit being lowered to physiological ranges.

Volume to be Blood volume* x (Observed hematocrit Desired hematocrit)


exchanged =
Observed hematocrit

*Blood volume is estimated to be 80-90 ml/kg in term babies and 90-100 ml/kg
in preterm babies
181 109
1120 Indian J Pediatr (2010) 77:11171121

The volume of blood to be exchanged is given by the Evidence for Management of Polycythemia
formula shown in the box.
As a rough guide, the volume of blood to be exchanged Partial exchange transfusion reverses the physiological
is usually 20 ml/kg. abnormalities associated with the polycythemia-
hyperviscocity syndrome. It improves capillary perfusion,
PET: Peripheral vs. Umbilical Route cerebral blood flow and cardiac function. However, there
is very little data to suggest that PET improves long term
Partial exchange transfusion may be carried out via the outcome in patients with polycythemia. The Cochrane
peripheral route or the central route. In the former, blood is reviewpublished this year (2010)concludes that
withdrawn from the peripheral arterial line and replaced there are no proven clinically significant short or long-
simultaneously with saline via the venous line. In the central term benefits of PET in polycythemic newborn infants
route, the umbilical venous catheter is used for withdrawing who are clinically well or who have minor symptoms
blood while the same amount of saline is replaced through a related to hyperviscosity; PET may lead to an increase in
peripheral vein. Alternatively, the umbilical venous catheter the risk of NEC [12]. However, as the review authors
may be used both for withdrawal of blood and replacement pointed out, the data regarding developmental outcomes
with saline. Two systematic reviews (including a Cochrane are extremely imprecise due to the large number of
review) have shown that the partial exchange transfusion surviving infants who were not assessed and, therefore,
through umbilical route may be associated with increased the true risks and benefits of PET are unclear. It is possible
risk of necrotizing enterocolitis [11, 12]. that the underlying etiology of polycythemia is a more
important determinant of ultimate outcome. Given the
PET: Fluids to Be Used uncertainty regarding the long term outcomes, it is
preferable to do partial exchange transfusion in symptom-
Crystalloids such as normal saline (NS) or ringers lactate atic infants with hematocrit of >65% and in asymptomatic
(RL) are preferred over colloids because they are less neonates with hematocrit of >75%.
expensive and easily available, produce a similar reduction
in hematocrit as colloids [13, 14], and do not have the risk
of transfusion associated infections. Moreover, adult plasma
has been shown to increase the blood viscosity when mixed References
with fetal erythrocytes. We use only normal saline for
partial exchange transfusion.
1. Mackintosh TF, Walkar CH. Blood viscosity in the newborn. Arch
Dis Child. 1973;48:54753.
2. Phibbs RH. Neonatal Polycythemia. In: Rudolph AB, editor.
(b) Asymptomatic polycythemia Pediatrics. 6th ed. New York: Appleton Century Crofts; 1997. p.
179.
3. Ramamurthy RS, Brans WY. Neonatal Polycythemia I. Criteria
The line of management in infants with asymptomatic for diagnosis and treatment. Pediatrics. 1981;68:16874.
polycythemia depends upon their hematocrit values. 4. Wirth FH, Goldberg KE, Lubchenco LO. Neonatal hyperviscocity
I. Incidence. Pediatrics. 1979;63:8336.
i. Hematocrit >75%: These infants are usually managed 5. Stevens K, Wirth FH. Incidence of neonatal hyperviscosity at sea
with partial exchange transfusion. level. Pediatrics. 1980;97:118.
ii. Hematocrit between 70% and 75%: Conservative 6. Bada HS, Korones SB, Pourcyrous M, et al. Asymptomatic
syndrome of polycythemic hyperviscocity: effect of partial
management with hydration may be tried in infants exchange transfusion. J Pediatr. 1992;120:57985.
with hematocrit of 70% to 75%. An extra fluid aliquot 7. Shohat M, Merlob P, Reisner SH. Neonatal Polycythemia. I. Early
of 20 ml/kg may be added to the daily fluid require- diagnosis and incidence relating to time of sampling. Pediatrics.
ments. The additional fluid may be ensured by either 1984;73:710.
8. Shohat M, Reisner SH, Mimouni F, Merlob P. Neonatal
enteral (supervised feeding) or parenteral route (IV polycythemia II. Definition related to time of sampling. Pediatrics.
fluids). The rationale for this therapy is hemodilution 1984;73:113.
and the resultant decrease in viscosity. However, liberal 9. Oh W. Neonatal polycythemia and hyperviscosity. Pediatr Clin
fluid therapy may be associated with problems espe- North Am. 1986;33:52332.
10. Goldberg K, Wirth FH, Hathaway WE, et al. Neonatal hyper-
cially in preterm neonates. viscocity II. Effect of partial exchange transfusion. Pediatrics.
iii. Hematocrit between 65% and 70%: They only need 1982;69:41925.
11. Dempsey EM, Barrington K. Short and long term outcomes
monitoring for any symptoms of polycythemia and re-
following partial exchange transfusion in the polycythaemic
estimation of hematocrit. Further management depends newborn: a systematic review. Arch Dis Child Fetal Neonatal
upon the repeat hematocrit values. Ed. 2006;91:F26.
182 110
Indian J Pediatr (2010) 77:11171121 1121

12. Ozek E, Soll R, Schimmel MS. Partial exchange transfusion to transfusion with saline versus plasma. Indian Pediatr.
prevent neurodevelopmental disability in infants with 1995;32:116771.
polycythemia. Cochrane Database Syst Rev 1. 2010: 14. de Waal KA, Baerts W, Offringa M. Systematic review of the
CD005089. optimal fluid for dilutional exchange transfusion in neonatal
13. Deorari AK, Paul VK, Shreshta L, Singh M. Symptomatic polycythaemia. Arch Dis Child Fetal Neonatal Ed. 2006;91:
neonatal polycythemia: comparison of partial exchange F710.
183
184
185
186
187 111

Symposium on AIIMS Protocols in Neonatology II

Post-resuscitation Management of Asphyxiated Neonates


Ramesh Agarwal, Ashish Jain, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, India

ABSTRACT
Inspite of major advances in monitoring technology and knowledge of fetal and perinatal medicine, perinatal asphyxia is one
of the significant causes of mortality and long term morbidity. Data from National Neonatal Perinatal Database suggests that
perinatal asphyxia contributes to almost 20% of neonatal deaths in India. Failure to initiate or sustain respiration after birth
has been defined as criteria for the diagnosis of asphyxia by WHO. Perinatal asphyxia results in hypoxic injury to various
organs including kidneys, lungs and liver but the most serious effects are seen on the central nervous system. Levenes
classification is a useful clinical tool for grading the severity of hypoxic ischemic encephalopathy. Good supportive care is
essential in the first 48 hours after asphyxia to prevent ongoing brain injury in the penumbra region. Strict monitoring and
prompt correction is needed for common problems including temperature maintenance, blood sugars, blood pressure and
oxygenation. Phenobarbitone is the drug of choice for the treatment of convulsions. [Indian J Pediatr 2008; 75 (2) : 175-180]
E-mail: vinodkpaul@hotmail.com

Key words : Asphyxia; HIE

According to latest estimates by World Health exist. It is probably better to use the term perinatal
Organization (WHO), approximately 4 million babies die asphyxia since asphyxia may occur in utero, at birth or in
each year before they reach the age of one month.1 Ninety- the postnatal period. WHO 2 has defined perinatal
eight percent of these neonatal deaths take place in the asphyxia as a failure to initiate and sustain breathing at
developing countries. Perinatal asphyxia and birth birth The National Neonatal Perinatal Database (NNPD),
injuries together contribute to almost 29% of these deaths.1 2000 used a similar definition for perinatal asphyxia.3 It
Most of the births in developing countries occur at home, defined moderate asphyxia as slow gasping breathing or
usually attended by untrained birth attendants. Failure to an Apgar score of 4-6 at 1 minute of age. Severe asphyxia
initiate and sustain breathing immediately after delivery was defined as no breathing or an Apgar score of 0-3 at 1
has been associated with hypoxic-ischemic injury to the minute of age.
central nervous system (CNS) and the clinical
As per the American Academy of Pediatrics (AAP) and
manifestations of this injury have been termed as hypoxic
American College of Obstetrics and Gynecology (ACOG),
ischemic encephalopathy (HIE). Some experts prefer to
all of the following must be present for designation of
use term neonatal encephalopathy , as it is not always
asphyxia viz. (a) profound metabolic or mixed academia
possible to document the hypoxic ischemic insult and
(pH< 7.00) in cord. (b) persistence of Apgar scores 0-3 for
there may be potential several other etiologies operating.
longer than 5 minutes. (c) neonatal neurologic sequelae
HIE is of concern in an asphyxiated neonate because it can
(e.g., seizures, coma, hypotonia). (d) multiple organ
lead to serious long-term neuro-motor sequelae among
involvement (e.g., of the kidney, lungs, liver, heart,
survivors.
intestine)
Definitions based on Apgar scores may be useful as it
DEFINITION
can be used for formulating guidelines for post-asphyxial
treatment of neonates. Apgar scores are also useful for
A gold standard definition of birth asphyxia does not predicting long term outcome in infants with perinatal
asphyxia.4,5

Correspondence and Reprint requests : Dr Vinod K. Paul, INDIAN DATA


Professor, Department of Pediatrics, All India Institute of Medical
Sciences, Ansari Nagar, New Delhi 110029, India According to NNPD 20003 data collected from 17 tertiary
[Received January 11, 2008; Accepted January 11, 2008] neonatal intensive care units in India, Apgar scores <7 at

Indian Journal of Pediatrics, Volume 75February, 2008 175


188 112

R. Agarwal et al

1 minute (includes moderate and severe asphyxia) were in term neonates was proposed by Sarnat and Sarnat7. A
documented in 9% of all intramural deliveries.2 2.5% simple and practical classification of HIE by severity of
babies continued to have Apgar scores <7 at 5 minutes of manifestations provided by Levene is recommended for
age. Bag and mask ventilation was used in 4.5% infants routine use (Table 2).8
and less than 1% infants needed cardiac compressions
and/or medications for resuscitation at birth. Perinatal
EVOLUTION OF HIE CHANGES
asphyxia was responsible for 20% of all neonatal deaths.
Manifestations of HIE were seen in approximately 1.5% of
all babies. Perinatal asphyxia was the commonest cause of Hypoxic-ischemic brain damage is a gradually evolving
still-births accounting for one-third of all such cases. process, which begins during the insult and extends
beyond the resuscitation period. Although the initial
brain injury (phase of energy failure) is related to hypoxia
SYSTEMIC CONSEQUENCES OF ASPHYXIA
and ischemia, subsequent reperfusion and generation of
free radicals contributes to ongoing injury.9 The initial
Perinatal asphyxia leads to muti-organ dysfunction. hypoxic-ischemic injury results in an area of infarction.
Virtually any organ can be effected. And care in the The immediate area surrounding this area is termed as
nursery should be oriented to determining the presence or penumbra.9 The penumbra continues to show adverse
the absence of dysfunction of the critical organ systems.. changes in the form of neuronal necrosis or apoptosis
Many of these complications are potentially fatal. In term (programmed cell death) even after the hypoxic insult is
infants with asphyxia, renal, CNS, cardiac and lung over.9 It is possible that these post-hypoxic changes in the
dysfunction occur in 50%, 28%, 25% and 25% cases, penumbra area may be amenable to therapeutic
respectively.6 The extent of organ system dysfunction interventions. The duration of this delayed phase of
determines the early outcome of an asphyxiated neonate neuronal injury is not known precisely, but appears to
(Table 1). increase over first 24 to 48 hours and than starts to resolve.
Thus good supportive therapy is essential for the first 48
TABLE 1. Organ System Dysfunction in Perinatal Asphyxia
hours of post-asphyxial period to reduce neuronal injury
CNS Hypoxic ischemic encephalopathy, intracranial in the penumbra area. The extent of penumbra and the
hemorrhage seizures, long-term neurological duration for which these adverse changes continue is
sequelae variable. Additional factors that may influence the
Cardiac Myocardial dysfunction, valvular dysfunction,
rhythm abnormalities, congestive cardiac failure outcome include the nutritional satus of the brain, severe
Renal Hematuria, acute tubular necrosis, renal vein intrauterine restriction, preexisting brain pathology, and
thrombosis the frequent siezuure disorder that anifest in the early
Metabolic Acidosis, hypoglycemia, hypocalcemia, postnatal age ( within hours of birth)This phase may also
hyponatremia provide a therapeutic window for newer modes of
Pulmonary Delayed adaptation, respiratory failure, meconium
aspiration
therapy in asphyxiated neonates.
Surfactant depletion, primary pulmonary
hypertension
GI tract Necrotizing enterocolitis, hepatic dysfunction MANAGEMENT OF A NEONATE WITH
Hematological Thrombocytopenia, coagulation abnormalities PERINATAL ASPHYXIA

Metabolic involvement may include hypocalcemia, Initial management (Fig. 1)


hyponatremia (as a result of SIADH or direct renal
injury), and alterations in glucose metabolism. There may Management of each and every baby needs to be
be hematological alterations (thrombocytopenia and DIC) individualized. Given below are broad guidelines.

Hypoxic ischemic encephalopathy (HIE) refers to the Transfer the baby to special care newborn unit: A baby
CNS dysfunction associated with perinatal asphyxia. HIE who fails to initiate and sustain respiration at birth is at
is of foremost concern in an asphyxiated neonate because risk of hypoxic brain injury and needs regular monitoring.
of its potential to cause serious long-term neuromotor All these bsbies should have a cord gas analysis
sequelae among survivors. A detailed classification of HIE performed. Infants with moderate asphyxia (Apgar score
TABLE 2. Classification of HIE (Levene)8

Feature Mild Moderate Severe

Consciousness Irritable Lethargy Comatose


Tone Hypotonia Marked hypotonia Severe hypotonia
Seizures No Yes Prolonged
Sucking/respiration Poor suck Unable to suck Unable to sustain spontaneous respiration

176 Indian Journal of Pediatrics, Volume 75February, 2008


189 113

Post-resuscitation Management of Asphyxiated Neonates

lead to acidosis, myocardial depression, hypotension,


Transfer to newborn unit
(Obtain a cord gas) bleeding tendency and pulmonary hemorrhage etc.
The therapeutic modality of hypothermia to prevent
disability is a new evolving therepy.10 The combination of
Keep baby under radiant the three main trials suggest that mild hypothermia is
warmer, maintain normal associated with significant reduction in deaths and severe
temperature (avoiod hyperthermia) disability following asphyxia.11 Therapeutic hypothermia
for encephalopathic asphyxiated newborn infants should
be only administered in well designed randomized
controlled trials as of now.12, 13, 14 as many of the aspects of
Check vital signs hypothermia need to be optimized and refined, and the
at regular intervals as needed magnitude of the risk vs the benefit when applied to the
population of the infants with HIE has yet to be
established.
Check blood gases, blood sugar,
Check vital signs: Immediate clinical assessment should
hematocrit and serum calcium
if needed/ siezures be made by recording respiration, heart rate, blood
pressure, capillary refill time, temperature and oxygen
saturation. Urine output monitoring should be done.
Insert intravenous line for Start intravenous fluids (in severely affected babies): All
dextrose and lab work babies with Apgar scores <4 at 1 minute or <7 at 5minutes
of age should be started on intravenous fluids. Current
other lines like for Invasive BP (UAC) , and UVC for CVP
recommendations to restrict fluid input are based mainly
monitoring is preferred in sick baby
on data from the treatment of adults and older children,
or from animal models of cerebral hypoxia. The rationale
is that fluid restriction may limit cerebral oedema, which
Consider use of volume may be important in the pathogenesis of brain damage
Expander after perinatal asphyxia. However, there is concern that
excessive fluid restriction may cause dehydration and
Keep the ionotropes standby if no response hypotension, resulting in decreased cerebral perfusion
and further brain damage.15
Check blood sugar, hematocrit and blood gases: Check
Vitamin K, stomach wash blood sugar (to detect hypoglycemia or hyperglycemia),
urine output monitoring hematocrit (to detect anemia and polycythemia) and
blood gases (to detect metabolic acidosis, hypoxia,
hyperoxia and respiratory failure). Hyperoxia, hypocarbia
Calcium (40mg/kg) is started in babies with severe birth asphyxia
prophylactically and hyperglycemia are equally detrimental to an injured
brain and emphasis should be given to maintaining all
Fig. 1. Summary of initial management of asphyxiated neonates
parameters in the normal range for the first 48-72 hours.

4-6 at 1 minute of age) may be transferred to the mother. Maintain the blood gases and acid-base status in the
However, these infants should also be monitored physiological ranges including partial pressure of arterial
frequently in the first 48-72 hours for features suggestive oxygen (PaO2), 80-100 mm Hg; partial pressure of arterial
of HIE. Infants with severe asphyxia (Apgar score 0-3 at 1 carbon dioxide (PaCO2), 35-40 mm Hg; and pH, 7.35-7.45.
minute or need for prolonged bag and mask ventilation Consider volume expander: If the capillary refill time is
>5 minutes) should be transferred to a special care more than 3 seconds or if there is metabolic acidosis,
newborn unit for observation and treatment. volume expansion with normal saline (or Ringers lactate)
Maintain temperature : Place the baby under the radiant 10 ml/kg over 5-10 min should be instituted. This may be
warmer after drying the baby. Maintain normal repeated, if required. One should remember that decrease
temperature of the baby. Adverse outcomes have been in vascular tone results in relative hypovolemia (preload)
associated with pyrexia in these newborns, hence it is to in babies with asphyxia. Maintain the mean BP above 35
be avoided. It is during these early minutes after birth that mm Hg (for term infants). Dopamine or dobutamine can
the chances of hypothermia are highest. Hypothermia be used to maintain adequate cardiac output. Attention to
imposes additional stress to the baby by increasing perfusion is the single most important component of therapy of
metabolic needs in the face of hypoxia-ischemia. This may asphyxiated neonates at this stage.

Indian Journal of Pediatrics, Volume 75February, 2008 177


190 114

R. Agarwal et al

Miscellaneous: Vitamin K (I mg IM) should be Hypoechoic areas are seen only in very severe cases
administered to all infants with perinatal asphyxia. A (having large areas of infraction). In preterm babies,
stomach wash may be performed if there was meconium however, cranial ultrasound is able to detect
staining. periventricular leukomalacia and intraventricular-
periventricular hemorrhage.
SUBSEQUENT MANAGEMENT Computed tomography (CT): CT scan in the acute stage of
HIE in term babies many show generalized low
Continue monitoring: Monitoring of vital parameters attenuation of brain parenchyma.
referred to above must be continued. Accurate record of
Magnetic resonance maging: MRI provides prognostic
urine output is a must. Blood gases should be checked as
information, The findings of abnormalities of thalami and
often as required. In addition, periodic evaluation of
ganglia in term, White and grey matter abnormalities at
blood chemistry should be done: blood sugar (2, 6, 12, 24,
term equivalent in preterms are strong predictors of
48 and 72 hours of age), hematocrit (once a day for the
subsequent risk of poor neurodevelopmental outcome.16
first few days) and serum sodium, potassium and
It is important to understand that MRI is preferred over
calcium (once a day). The renal function studies (serum
CT as it has a greater interobserver agreement and no
creatinine, creatinine clearance, and BUN ) may be
radiation exposure.
estimated if required. Cardiac and liver enzymes values
are an adjunct to assess the degree of hypoxic-ischemic In general, EEG, CT and US does not help a great deal
injury to these other organs. These studies also provide in the acute management of the baby. Their utility is
some insight into injuries to other organs, such as the essentially for prognostication.
bowel.
ECHO: In infants needing ionotropic support, an ECHO
A special effort needs to be made for monitoring the at times may give an insight into the contractinbility and
neurological status of the baby. Assessment of sensorium, the asphyxial injury to the heart (MR,)and guide
tone, seizures, autonomic disturbance and reflexes should appropriate management strategy.
be made every 4-6 hours. Hypotonia in HIE is typically
Treatment (Fig. 2)
differential in term babies, especially in the early stages of
HIE. It affects upper limbs more than lower limbs and (a) Maintain oxygenation and ventilation: Babies with
involves proximal musculature more than the distal respiratory failure require oxygen and assisted
musculature. Thus, scarf sign becomes readily abnormal. ventilation. Some babies would already be on oxygen and
Flexors of neck become weak and poor traction response bag and mask (or endotracheal tube) ventilation when
is seen. Seizures may be subtle in character and therefore, transferred from the labor room. Oxygenation in those
a close observation is required to document them. Based
on the findings, HIE is classified as mild, moderate and Maintain
severe using Levenes classification (Table 2). oxygenation
and ventilation
Special investigations: EEG does not help in the routine
management of most cases of HIE. Its use lies in Maintain
Maintain
prognostication to some extent. Burst suppression normal
adequate
pattern, low voltage or iso-electric EEG indicates poor perfusion
glucose
outcome.
Amplitude-integrated electroencephalography (aEEG):
Several studies have shown that a single-channel aEEG
performed within a few hours of birth can help evaluate
the severity of brain injury in the infant with HIE. While Subsequent management of
asphyxiated neonates
a normal aEEG may not necessarily mean that the brain is
normal, a severe or moderately severe aEEG abnormality
may indicate brain injury and poor outcome. The
abnormalities include wide fluctuations in the amplitude
with the baseline voltages dropping to near zero and the
peak amplitudes under 5 mV. Seizure spikes may be
seen.15 Although many centers are using aEEG, note that Maintain normal
Treat seizures Maintain normal serum calcium
considerable training is required for conducting and
properly interpreting the aEEG tracings. hematocrit

Cranial ultrasound (US): Cranial ultrasound is not good


for detecting changes of HIE in the term babies. Fig. 2. Summary of subsequent management of asphyxiated neonates

178 Indian Journal of Pediatrics, Volume 75February, 2008


191 115

Post-resuscitation Management of Asphyxiated Neonates

babies who have adequate spontaneous breathing can be 20 mg/kg, intravenously slowly over 20 minutes. If there
achieved in the oxygen hood. But if there is apnea, or is no response, two additional doses of 10 mg/kg can be
spontaneous respiration is inadequate or there is given every 15 minutes. The maximum loading lose is
continuing hypoxia or hypercarbia, assisted ventilation is thus 40 mg/kg. The rate of infusion of phenobarbitone
indicated. pH should be maintained above 7.30. The should not exceed 1 mg/kg/min and preferably an
target PaO2 is 80-100 torr in term babies and 60-80 torr in infusion pump should be used to deliver the drug. If
preterm infants. Pulse oximeter saturation values are convulsions are still uncontrolled, phenytoin sodium
maintained in the 90-95% range in term babies and 90-93% should be added in a dose of 20 mg/kg intravenously
in the preterm infants. The target PaCO2 value is 40-45 slowly over 20 minutes. Maintenance therapy of both
torr. phenobarbitone and phenytoin is started 12 hours later in
a dose of 5 mg/kg/day in a single dose. Generally, one or
(b) Maintain adequate perfusion: Ensuring normal
both of these anticonvulsants are effective. Occasional,
perfusion is of critical importance in management. The
short-lasting, mild seizures not interfering with
markers of normal perfusion are normal blood pressure,
cardiopulmonary status may be left alone. For intractable
capillary refill time of less than 3 seconds, normal urine
seizures Clonazepam, Midazolam, Paraldehyde, and
output, and absence of metabolic acidosis. The BP should
Valproate may be tried. Diazepam is generally avoided in
be maintained in the upper normal range. In sick babies,
neonates. Always look and treat for other specific etiology
arterial line is placed for guiding management of blood
of seizures (hypoglycemia, hypocalcemia,
pressure. This is achieved by judicious use of volume
hypomagnesemia, polycythemia), which may co-exist.
expanders and vasopressors:
(c) Volume expansion: Use saline, Ringers lactate and
blood to maintain intravascular volume. In sick neonates, NEWER MODES OF THERAPY
confirm central venous pressure (CVP) by placing an
umbilical venous line.
Corticosteroids have no role on the treatment of HIE.
(d) Vasopressors: Dopamine and dobutamine are the Likewise, the current evidence does not support the use of
drugs of choice. Begin with dopamine 3-5 microgram/ mannitol in the management of HIE.
kg/min. Increase the dose in a stepwise fashion. At about
10 microgram/kg/min dose, if the perfusion is still poor, Some interest has been generated in the protective role
dobutamine should be added in a dose of 5 microgram/ of prophylactic phenobarbitone in newborns with
kg/min. The usual maximum dose of each drug is 20 perinatal asphyxia. A dose of 40 mg/kg administered
microgram/kg/min. prophylactically was associated with a better neuro-
developmental outcome at 3 years of age.17 However, the
(e) Maintain normal blood glucose: Both hypoglycemia Cochrane review database systematic review by Evans et
and hyperglycemia are indesirable. Glucose is the al (2000) that included the 5 RCTs derived no difference
substrate for brain and its requirements go up in HIE. in the risk of death, neurodisability.18 Another study using
Hence, it must be made available. But hyperglycemia can 40 mg/kg within 1st hour showed a significant reduction
precipitate hyperosmolality and aggravate lactic acidosis. in HIE with no difference in complications. 19
Hence, the dictum is to maintain blood sugar above 60 Recommendation for use of prophylactic phenobarbitone
mg/dl, but not exceeding 100 mg/dl. still awaits further studies.
(f) Maintain normal calcium level: Calcium should be A large number of drugs are under investigations for
provided in a maintenance dose of 4 ml/kg/day (of 10% neuro-protection in HIE. These need to be used in the
calcium gluconate) for 1-2 days. This may be given as a early period of hypoxic ischemic injury.10 They act by
continuous infusion or as 1:1 diluted boluses, slowly causing blockade of free radical generation (allopurinol,
under cardiac monitoring. The aim is to maintain serum oxypurinol), scavenging of oxidants (superoxide
calcium concentration in the normal range. dismutase, glutathione, N-acetyl cysteine and alpha
tocopherol), calcium channel blockade (flunarizine,
(g) Maintain normal hematocrit: Anemia should be
nimodipine), blockage of NMDA receptors (magnesium,
corrected and hemotocrit should be maintained >40% in
MK801, dextromethorphan) and blockage of
ventilated neonates. It is also equally important to treat
inflammatory mediators (phospholipase A 2 ,
polycythemia. Polycythemia causes hyperviscosity with
indomethcin). One promising modality on the horizon is
adverse cardiopulmonary consequences. It is therefore
cerebral hypothermia. 12,13,14,20 Mild reductions in
recommended that if the venous hematocrit in a baby is
temperature of the body as a whole or of the head (brain)
above 65%, it should be brought down to 55% by partial
exchange transfusion using normal saline. has been shown to minimize the effects of hypoxic
ischemic encephalopathy. It may be noted that none of the
(h) Treat seizures: The anticonvulsant of choice for modalities mentioned in this section are ready for routine
controlling seizures is phenobarbitone. The initial dose is use as yet.

Indian Journal of Pediatrics, Volume 75February, 2008 179


192 116

R. Agarwal et al

FOLLOWUP Pediatr 2001; 138 : 798-803.


6. Perlman JM, Tack ED, Martin T, Shackelford G, Amon E.
Acute systemic organ injury in term infants fter asphyxia. Am
All the neonates with the moderate and severe asphyxia, J Dis Child 1989; 143 : 617-620.
especially those with stage II and III HIE staging should 7. Sarnat HB, Sarnat MS. Neonatal encephalopathy following
fetal distress: A clinical and electroencephalographic study.
be followed in the High risk clinic, they should have a Arch Neurol 1976; 33: 695-706.
complete neurological assessment and intervention if 8. Levene MI. The asphyxiated newborn infant. In Levene MI,
needed during the followup. A formal psychometric Lilford RJ, ed. Fetal and neonatal neurology and neuro-surgary.
assessment at 18 months should be performed in all these Edinburgh; Churchil Livingstone, 1995: 405-426.
babies 9. Du-Pleiss AJ, Johnston MV. Hypoxic ischemic injury in
newborn: cellular mechanism and potential strategies for
neuroprotection. Clinics in Perinatology 1997; 29: 627-654.
10. Vannucci RC. Current and potentially new management
LONG TERM OUTCOME strategies for perinatal hypoxic ischemic encephalopathy.
Pediatrics 1990; 85 : 961-968.
11. Thoresen M, Wyatt J. Keeping a cool head, post-hypoxic
Among the infants who survive severe HIE, the sequelae hypothermia an old idea revisited. Acta Paediatr 1997; 86:
include mental retardation, epilepsy, and cerebral palsy of 1029-1033.
12. Gluckman PD, Wyatt JS, Azzopardi D, Ballardi R, Edwards
varying degrees. The latter can be in the form of
AD, Ferreio DM et al. Selective head cooling with mild
hemiplegia, paraplegia, or quadriplegia. Such infants systemic hypothermia after neonatal encephalopathy :
need careful evaluation and support. They may need to Multicenter randomized trial. Lancet 2005; 365: 663-670.
be referred to specialized clinics capable of providing 13. Shankaran S, Laptook AR, Ehraenkranz RA, Tyson JE, Mc
coordinated comprehensive follow-up care. Donald SA, Donovan TF et al. Whole body hypothermia for
neonates with hypoxic ischemic encephalopathy. NEJM 2005;
The incidence of long-term complications depends on 353: 1574-1584.
the severity of HIE. Up to 80% of infants who survive 14. Eicher DJ, Wagner CL, Katikaneni LP, Hulsey TC, Bass WT,
severe HIE develop serious complications, 10-20% Kaufman DA et al. Moderate hypothermia in neonatal
encephalopathy: efficacy outcomes. Pediatr Neurol 2005; 32:
develop moderately serious disabilities, and up to 10% are 11-17.
normal. Among the infants who survive moderately 15. Kecskes Z, Healy G, Jensen A. Fluid restriction for term
severe HIE, 30-50% may suffer from serious long-term infants with hypoxicischaemic encephalopathy following
complications, and 10-20% with minor neurological perinatal asphyxia. In The Cochrane Library: Issue 3, 2005.
morbidities. Infants with mild HIE tend to be free from 16. Lianne J, Woodward, Peter J Anderson, Nicole C Austin,
Kelly Howard, Terrie E Inder. Neonatal MRI to predict
serious CNS complications
neurodevelopmental outcomes in preterm infants. NEJM
2006; 355 : 685-694.
17. Hall RT, Hall FK, Daily DK. High-dose phenobarbital therapy
REFERENCES in term newborn infants with severe perinatal asphyxia: a
randomized, prospective study with three-year follow-up. J
Pediatr 1998; 132 : 345-348.
1. Costello A, Francis V, Byrne A, Puddephatt C. State of the 18. Evans DJ, Levene MI. Anticonvulsants for preventing
worlds newborns. Kinetik Communications 2001. mortality and morbidity in full term newborns with perinatal
2. World Health Organizaton. Perinatal mortality: a listing of asphyxia. Cochrane Database Syst Rev 2000; CD001240.
available information. FRH/MSM.96.7.Geneva:WHO, 1996. 19. Vargas-Origel A, Espinosa- Garcia JO, Muniz-Quezada E,
3. Report of the National Neonatal Perinatal Database (National Vargas-Nieto MA, Aguilar-Garcia G, Prevention of hypoxic-
Neonatology Forum, India) 2000 ischemic encephalopathy with high dose, early phenobarbitol
4. Casey BM, McIntire DD, Leveno KJ. The continuing value of therepy. Gac Med Mex 2004; 140 : 147-153.
the Apgar score for the assessment of newborn infants. New 20. John S, Wyatt, Peter D, Gluckman, Ping Y, Liu, Denis
Engl J Med 2001; 344 : 467-471. Azzopardi, Roberta Ballard, A. David Edwards et al. Gunn for
5. Moster D, Lie RT, Irgens LM, Bjerkedal T, Markestad T. The the CoolCap Study Group Determinants of Outcomes After
association of Apgar score with subsequent death and Head Cooling for Neonatal Encephalopathy. Pediatrics 2007;
cerebral palsy: A population based study in term infants. J 119: 912-921.

180 Indian Journal of Pediatrics, Volume 75February, 2008


193
194
195
196
197 117
Indian J Pediatr (2010) 77:11291135
DOI 10.1007/s12098-010-0209-8

SYMPOSIUM ON AIIMS PROTOCOLS IN NEONATOLOGY-I

Management of Neonatal Seizures


Jeeva M. Sankar & Ramesh Agarwal & Ashok Deorari &
Vinod K. Paul

Received: 28 July 2010 / Accepted: 2 August 2010 / Published online: 30 September 2010
# Dr. K C Chaudhuri Foundation 2010

Abstract Seizures in the newborn period constitute a Definition


medical emergency. Subtle seizures are the commonest
type of seizures occurring in the neonatal period. Myoclon- A seizure is defined clinically as a paroxysmal alteration in
ic seizures carry the worst prognosis in terms of long-term neurologic function, i.e., motor, behavior and/or autonomic
neurodevelopmental outcome. Hypoxic-ischemic encepha- function. This definition includes [3]:
lopathy is the most common cause of neonatal seizures.
1. Epileptic seizures: phenomena associated with
Multiple etiologies often co-exist in neonates and hence it is
corresponding EEG seizure activity e.g. clonic seizures
essential to rule out common causes such as hypoglycae-
2. Non-epileptic seizures: clinical seizures without
mia, hypocalcemia, and meningitis before initiating specific
corresponding EEG correlate e.g., subtle and general-
therapy. A comprehensive evidence based approach for
ized tonic seizures
management of neonatal seizures has been described in this
3. EEG seizures: abnormal EEG activity with no clinical
protocol.
correlation.
Keywords Seizure . Newborn . Anti-epileptic therapy

Neonatal seizures (NS) are the most frequent and Classification


distinctive clinical manifestation of neurological dysfunc-
tion in the newborn infant. The incidence of NS is 2.8 per Four types of NS have been identified [1] (Table 1):
1000 in infants with birth weights of more than 2500 g; it
is higher in preterm low birth weight neonatesas high as Subtle seizures: They are called subtle because the
57.5 per 1000 in very low birth weight infants [1]. Infants clinical manifestations are mild and frequently missed.
with NS are at high risk of neonatal death or neurological They are the commonest type and constitute about 50%
impairment and epilepsy disorders in later life. Though, of all seizures. Common examples of subtle seizures
mortality due to NS has decreased over the yrs from 40% include:
to about 20%, the prevalence of long-term neurodevelop- 1. OcularTonic horizontal deviation of eyes or
ment sequelae has largely remained unchanged at around sustained eye opening with ocular fixation or
30% [2]. Improper and inadequate management of cycled fluttering
seizures could be one of the major reasons behind this 2. Oralfaciallingual movementsChewing, tongue-
phenomenon. thrusting, lip-smacking, etc.
3. Limb movementsCycling, paddling, boxing-jabs,
J. M. Sankar : R. Agarwal (*) : A. Deorari : V. K. Paul etc.
Division of Neonatology, Department of Pediatrics, All India 4. Autonomic phenomenaTachycardia or bradycardia
Institute of Medical Sciences,
5. Apnea may be a rare manifestation of seizures.
Ansari Nagar,
New Delhi 110029, India Apnea due to seizure activity has an accelerated or
e-mail: aranag@rediffmail.com a normal heart rate when evaluated 20 sec after
198 118
1130 Indian J Pediatr (2010) 77:11291135

Table 1 Investigations Required in a Neonate with Seizures

Essential investigations (required in all with few exceptionsa) Additional investigations

Blood sugar Hematocrit (if plethoric and/or at risk for polycythemia)


Serum Na, calcium, magnesium Serum bilirubin (if icteric)
Cerebrospinal fluid (CSF) examination Arterial blood gas and anion gap (lethargy, vomiting, family history, etc.)
Cranial ultrasound (US) and Imaging: CT and/or MRI (if no etiology found after essential investigations)
Electroencephalography (EEG) and/or amplitude integrated EEG TORCH screen for congenital infections
Work-up for inborn errors of metabolism
a
Given in the text

onset. Bradycardia is thus not an early manifesta- Hypoxic-Ischemic Encephalopathy (HIE) HIE secondary to
tion in convulsive apnea but may occur later due to perinatal asphyxia is the commonest cause of NS. Most
prolonged hypoxemia. seizures due to HIE (about 5065%) start within the first
Clonic seizures: They are rhythmic movements of 12 h of life while the rest manifest by 2448 h of age.
muscle groups. They have both fast and slow compo- Additional problems like hypoglycemia, hypocalcemia, and
nents, occur with a frequency of 13 jerks per sec, and intracranial hemorrhage may co-exist in neonates with
are commonly associated with EEG changes. perinatal asphyxia and these should always be excluded.
Tonic seizures: This type refers to a sustained flexion Subtle seizures are the most common type of seizures
or extension of axial or appendicular muscle groups. following HIE.
These seizures may be focal or generalized and may
resemble decerebrate (tonic extension of all limbs) or Metabolic Causes Common metabolic causes of seizures
decorticate posturing (flexion of upper limbs and include hypoglycemia, hypocalcemia, and hypomagnese-
extension of lower limbs). Usually there are no EEG mia. Rare causes include pyridoxine deficiency and inborn
changes in generalized tonic seizures. errors of metabolism (IEM).
Myoclonic seizures: These manifest as single or
multiple lightning fast jerks of the upper or lower Infections Meningitis should be excluded in all neonates
limbs and are usually distinguished from clonic move- with seizures. Meningoencephalitis secondary to intrauter-
ments because of more rapid speed of myoclonic jerks, ine infections (TORCH group, syphilis) may also present as
absence of slow return and predilection for flexor seizures in the neonatal period.
muscle groups. Common changes seen on the EEG
include burst suppression pattern, focal sharp waves Intracranial Hemorrhage Seizures due to subarachnoid,
and hypsarrhythmia. intraparenchymal or subdural hemorrhage occur more often
Myoclonic seizures carry the worst prognosis in terms in term neonates, while seizures secondary to intraventric-
of neuro-developmental outcome and seizure recur- ular hemorrhage (IVH) occur in preterm infants. Most
rence. Focal clonic seizures have the best prognosis. seizures due to intracranial hemorrhage occur between 2
and 7 days of age. Seizures occurring in a term well baby
on day 23 of life is often due to subarachnoid hemorrhage.

Common Causes of Neonatal Seizures [1, 48] Developmental Defects Cerebral dysgenesis and neuronal
migration disorders are rare causes of seizures in the
The most common causes of seizures as per the recently neonatal period.
published studies from the country are hypoxic ischemic
encephalopathy, metabolic disturbances (hypoglycemia and Miscellaneous They include polycythemia, maternal nar-
hypocalcemia), and meningitis; the incidence of intraven- cotic withdrawal, drug toxicity (e.g. theophylline, doxap-
tricular hemorrhage was low in both the studies [7, 8]. ram), local anesthetic injection into scalp, and phacomatosis
Etiology could, however, vary between different centres (e.g. tuberous sclerosis, incontinentia pigmentii). Acciden-
depending upon the patient population (term vs preterm), tal injection of local anesthetic into scalp may be suspected
level of monitoring (only clinical vs electrical and clinical in the presence of unilateral fixed and dilated pupil.
seizures), etc. Multifocal clonic seizures on the 5th day of life may be
199 119
Indian J Pediatr (2010) 77:11291135 1131

related to low zinc levels in the CSF fluid (benign neonatal deaths would be suggestive of inborn errors of
idiopathic neonatal convulsions). metabolism. History of seizures in either parent or siblings
Seizures due to SAH and late onset hypocalcemia carry in the neonatal period may suggest benign familial neonatal
a good prognosis for long term neuro-developmental convulsions (BFNC).
outcome while seizures related to hypoglycemia, cerebral
malformations, and meningitis have a high risk for adverse Examination
outcome.
Vital Signs Heart rate, respiration, blood pressure, capillary
refill time and temperature should be recorded in all infants.

Approach to an Infant with Neonatal Seizures [1, 46] General Examination Gestation, birth-weight, and weight
for age should be recorded as they may provide important
History clues to the etiologyfor e.g.,, seizures in a term well
baby may be due to subarachnoid hemorrhage while
Seizure History A complete description of the seizure seizures in a large for date baby may be secondary to
should be obtained from the parents/attendant. History of hypoglycemia. The neonate should also be examined for
associated eye movements, restraint of episode by passive the presence of any obvious malformations or dysmorphic
flexion of the affected limb, change in color of skin features.
(mottling or cyanosis), autonomic phenomena, and wheth-
er the infant was conscious or sleeping at the time of CNS Examination Presence of a bulging anterior fontanel
seizure should be elicited. The day of life on which the may be suggestive of meningitis or intracranial hemor-
seizures occurred may provide an important clue to its rhage. A detailed neurological examination should include
diagnosis. While seizures occurring on day 03 might be assessment of consciousness (alert/drowsy/comatose), tone
related to perinatal asphyxia, intracranial hemorrhage, and (hypotonia or hypertonia), and fundus examination for
metabolic causes, those occurring on day 47 may be due chorioretinitis.
to sepsis, meningitis, metabolic causes, and developmental
defects. Systemic Examination Presence of hepatosplenomegaly or
an abnormal urine odor may be suggestive of IEM. The
Antenatal History History suggestive of intrauterine infec- skin should be examined for the presence of any neuro-
tion, maternal diabetes, and narcotic addiction should be cutaneous markers. Presence of hypopigmented macules or
elicited in the antenatal history. A history of sudden ash-leaf spot would be suggestive of tuberous sclerosis.
increase in fetal movements may be suggestive of intra-
uterine convulsions.

Perinatal History Perinatal asphyxia is the commonest Investigations


cause of neonatal seizures and a detailed history including
history of fetal distress, decreased fetal movements, instru- Essential Investigations Investigations that should be con-
mental delivery, need for resuscitation in the labor room, sidered in all neonates with seizures include blood sugar,
Apgar scores, and abnormal cord pH (<7) and base deficit serum electrolytes (Na, Ca, Mg), cerebrospinal fluid (CSF)
(>10 mEq/L) should be obtained. Use of a pudendal block examination, cranial ultrasound (US), and electroencepha-
for mid-cavity forceps may be associated with accidental lography (EEG). CSF examination should be done in all
injection of the local anesthetic into the fetal scalp. cases as seizures may be the first sign of meningitis. It should
not be omitted even if another etiology such as hypoglycemia
Feeding History Appearance of clinical features including is present because meningitis can often coexist. CSF study
lethargy, poor activity, drowsiness, and vomiting after may be withheld temporarily if severe cardio-respiratory
initiation of breast-feeding may be suggestive of inborn compromise is present or even omitted in infants with severe
errors of metabolism. Late onset hypocalcemia should be birth asphyxia (documented abnormal cord pH/base excess
considered in the presence of top feeding with cows and onset within 1224 h). An arterial blood gas (ABG) may
milk. have to be performed if IEM is strongly suspected.
One should carry out all these investigations even if one
Family History History of consanguinity in parents, family or more investigations are positive, as multiple etiologies
history of seizures or mental retardation and early fetal/ may coexist, e.g. sepsis, meningitis and hypoglycemia.
200 120
1132 Indian J Pediatr (2010) 77:11291135

Additional Investigations These may be considered in Metabolic Screen This includes blood and urine ketones,
neonates who do not respond to a combination of urine reducing substances, blood ammonia, anion gap,
phenobarbitone and phenytoin or earlier in neonates with urine and plasma aminoacidogram, serum and CSF lactate/
specific features. These include neuroimaging (CT, MRI), pyruvate ratio.
screen for congenital infections (TORCH) and for inborn
errors of metabolism.
Management
Imaging Neurosonography is an excellent tool for detection
of intraventricular and parenchymal hemorrhage but is Initial Medical Management
unable to detect SAH and subdural hemorrhage. It should
be done in all infants with seizures. CT scan should be done The first step in successful management of seizures is to
in all infants where an etiology is not available after the first nurse the baby in thermoneutral environment and to ensure
line of investigations. It can be diagnostic in subarachnoid airway, breathing, and circulation (TABC). Oxygen should
hemorrhage and developmental malformations. Magnetic be started, IV access should be secured, and blood should
resonance imaging (MRI) is indicated only if investigations be collected for glucose and other investigations. A brief
do not reveal any etiology and seizures are resistant to usual relevant history should be obtained and quick clinical
anti-epileptic therapy. It can be diagnostic in cerebral examination should be performed. All this should not
dysgenesis, lissencephaly, and other neuronal migration require more than 25 min.
disorders.
Correction of Hypoglycemia and Hypocalcemia
Electroencephalogram (EEG) EEG has both diagnostic
and prognostic role in seizures. It should be done in all If glucostix shows hypoglycemia or if there is no facility to
neonates who need anticonvulsant therapy. Ictal EEG may test blood sugar immediately, 2 ml/kg of 10% dextrose
be useful for the diagnosis of suspected seizures and also should be given as a bolus injection followed by a
for diagnosis of seizures in muscle-relaxed infants. It continuous infusion of 68 mg/kg/min.
should be done as soon as the neonate is stable enough to If hypoglycemia has been treated or excluded as a cause
be transported for EEG, preferably within first wk. EEG of convulsions, the neonate should receive 2 ml/kg of 10%
should be performed for at least 1 h [9]. Inter-ictal EEG is calcium gluconate IV over 10 min under strict cardiac
useful for long-term prognosis of neonates with seizures. monitoring. If ionized calcium levels are suggestive of
A background abnormality in both term and preterm hypocalcemia, the newborn should receive calcium gluco-
neonates indicates a high risk for neurological sequelae. nate at 8 ml/kg/d for 3 days. If seizures continue despite
These changes include burst-suppression pattern, low correction of hypocalcemia, 0.25 ml/kg of 50% magnesium
voltage invariant pattern and electro-cerebral inactivity. sulfate should be given intramuscularly (IM).

Amplitude Integrated EEG This new method provides Anti-Epileptic Drug Therapy (AED) [1]
continuous monitoring of cerebral electrical activity at the
bedside in critically sick newborns. a EEG is helpful in Anti-epileptic drugs (AED) should be considered in the
evaluating the background as well in identification of presence of even a single clinical seizure since clinical
seizure activity in neonatal seizures. As with conventional observations tend to grossly underestimate electrical seiz-
EEG, background abnormalities like burst-suppression or ures (diagnosed by EEG) and facilities for continuous EEG
continuous low voltage pattern in aEEG also help in monitoring are not universally available. If aEEG is being
prognosticating the infant with seizures particularly in the used, eliminating all electrical seizure activity should be the
setting of HIE. Seizure activity on aEEG is characterized by goal of AED therapy [1]. AED should be given if seizures
a rapid rise in both the lower and upper margins of the persist even after correction of hypoglycemia/ hypocalce-
trace. Some seizures that are focal or relatively brief are, mia (Fig. 1).
however, missed by this technique [1].
Phenobarbitone (Pb)
Screen for Congenital Infections TORCH screen and
VDRL should be considered in the presence of hepatosple- It is the drug of choice in neonatal seizures. The dose is
nomegaly, thrombocytopenia, intrauterine growth restric- 20 mg/kg/IV slowly over 20 min (not faster than 1 mg/kg/
tion, small for gestational age, and presence of min). If seizures persist after completion of this loading
chorioretinitis. dose, additional doses of phenobarbitone 10 mg/kg may
201 121
Indian J Pediatr (2010) 77:11291135 1133

Fig. 1 Acute management of


neonatal seizures Neonate with seizures

. Identify and characterize the seizure


. Secure airway and optimize breathing, circulation, and temperature
. Start oxygen, if seizures are continuous
. Secure IV access and take samples for baseline investigations including sugar, calcium,
. If hypoglycemic (blood sugar <40 mg/dl): administer 2 ml/kg of 10% dextrose as bolus
magnesium, sodium, potassium, arterial blood gas, hematocrit, sepsis screen

. followed by a continuous infusion of 6-8 mg/kg/min


If blood sugar is in normal range, sample for ionized calcium should be withdrawn; if
abnormal, 2 ml/kg of calcium gluconate (10%) should be given IV under cardiac monitoring

Seizures persist

Administer phenobarbitone 20mg/kg IV stat


over 20 min
Seizures continue

Repeat phenobarbitonein 10 mg/kg/dose


aliquots until 40 mg/kg dose is reached

Seizures continue

Administer phenytoin 20 mg/kg IV slowly


over 20 min under cardiac monitoring

Seizures continue

Repeat phenytoin 10 mg/kg/dose

Seizures continue

Consider Lorazepam / midazolam bolus and


midazolam infusion if needed;
Consider ventilation
Seizures continue

Consider other antiepileptic drugs,


pyridoxine, and exchange transfusion;
Consider ventilation

Seizures controlled

Wean AED slowly to maintenance phenobarbitone

be used every 2030 min until a total dose of 40 mg/kg under cardiac monitoring. Phenytoin should be diluted in
has been given. The maintenance dose of Pb is 35 mg/kg/ normal saline as it is incompatible with dextrose solution.
day in 12 divided doses, started 12 h after the loading A repeat dose of 10 mg/kg may be tried in refractory
dose. seizures. The maintenance dose is 35 mg/kg/d (maximum
of 8 mg/kg/d) in 24 divided doses. Oral suspension has
Phenytoin very erratic absorption from gut in neonates, so it should be
avoided. Thus ,only IV route is preferred in neonates and it
Phenytoin is indicated if the maximal dose of phenobar- should preferably be discontinued before discharge.
bitone (40 mg/kg) fails to resolve seizures or earlier, if Fosphenytoin, the prodrug of phenytoin, does not cause
adverse effects like respiratory depression, hypotension or the same degree of hypotension or cardiac abnormalities,
bradycardia ensue with phenobarbitone. The dose is has high water solubility (therefore can be given IM), and is
20 mg/kg IV at a rate of not more than 1 mg/kg/min less likely to lead to soft-tissue injury when compared with
202 122
1134 Indian J Pediatr (2010) 77:11291135

phenytoin. It is dosed in phenytoin equivalents (1.5 mg/kg depression and sedation than lorazepam. However, when
of fosphenytoin is equivalent to 1 mg/kg of phenytoin). used as continuous infusion, the infant has to be
monitored for respiratory depression, apnea, and brady-
Benzodiazepines cardia (equipment for resuscitation and assisted ventila-
tion should be available at the bedside of all neonates
This group of drugs may be required in up to 1520% of given multiple doses of AED).
neonatal seizures. The commonly used benzodiazepines are The doses of These drugs are given below:
lorazepam and midazolam. Diazepam is generally avoided
Lorazepam: 0.05 mg/kg IV bolus over 25 min; may
in neonates due to its short duration of action, narrow
be repeated
therapeutic index, and because of the presence of sodium
Midazolam: 0.15 mg/kg IV bolus followed by infusion
benzoate as a preservative. Lorazepam is preferred over
of 0.1 to 0.4 mg/kg/hr.
diazepam as it has a longer duration of action and results in
less adverse effects (sedation and cardiovascular effects). According to Volpe, the expected response of neonatal
Midazolam is faster acting than lorazepam and may be clinical seizures to anticonvulsants is 40% to the initial 20-
administered as an infusion. It causes less respiratory mg/kg loading dose of phenobarbitone, 70% to a total of

Fig. 2 Weaning of anticonvul-


sant therapy Newborn on anticonvulsant therapy

Wean all antiepileptic drugs except


phenobarbitone once seizure controlled

Perform neurological examination


prior to discharge

Normal Abnormal

Stop phenobarbitone Continue


prior to discharge phenobarbitone
for 1 month

Repeat neurological
examination at 1month

Normal
examination Abnormal examination

Evaluate EEG

Taper drugs Normal EEG Abnormal EEG


over 2 wks Taper drugs Continue drug;
over 2 wks reassess at 3 months*

*Intractable seizures may need lifelong therapy; consider switching over to other drugs (phenytoin or
carbamazepine)
203 123
Indian J Pediatr (2010) 77:11291135 1135

40 mg/kg of Pb, 85% to a 20-mg/kg of phenytoin, and 95% Exchange Transfusion This is indicated in life-threatening
to 100% to 0.05 to 0.1 mg/kg lorazepam [1]. metabolic disorders, accidental injection of local anaesthe-
tic, trans-placental transfer of maternal drugs (e.g. chlor-
Antiepileptic Drugs for Seizures Refractory to Above propamide), and bilirubin encephalopathy.
Treatment

In exceptional circumstances when the seizures are refrac- Maintenance Anti-Epileptic Therapy
tory to the first-line AEDs, the following second-line drugs
might be tried. Principles of AED used in older children and adults are
applicable to neonates also. Monotherapy is the most
Lidocaine It is usually administered as a bolus dose of appropriate strategy to control seizures. Attempts should
4 mg/kg IV followed by an infusion rate of 2 mg/kg/hr. It is be made to stop all anti-epileptic drugs and wean the baby
tapered over several days. Adverse effects include arrhyth- to only phenobarbitone at 35 mg/kg/day. If seizures are
mias, hypotension, and seizures. It should not be adminis- uncontrolled or if clinical toxicity appears, a second AED
tered with phenytoin. may be added. The choice may vary from phenytoin,
carbamezepine, and valproic acid.
Paraldehyde It may be used in seizures refractory to the first
line drugs. A dose of 0.10.2 ml/kg/dose may be given IM or When to Discontinue AED
0.3 ml/kg/dose mixed with coconut oil in 3:1 may be used by
per rectal route. Additional doses may be used after 30 min This is highly individualized and no specific guidelines are
and q 46 hrly. Adverse effects include pulmonary hemor- available. We usually try to discontinue all medication at
rhage, pulmonary edema, hypotension, and liver injury. discharge if clinical examination is normal, irrespective of
etiology and EEG. If neurological examination is persistently
Sodium Valproate It can be used for maintenance therapy in abnormal at discharge, AED is continued and the baby is
neonates. Per rectal route may be used in acute condition. reassessed at one month. If the baby is normal on examination
IV preparation is now available. Dose is 2025 mg/kg/d and seizure free at 1 month, phenobarbitone is discontinued
followed by 510 mg/kg every 12 h. It should, however, be over 2 wks. If neurological assessment is not normal, an EEG
used with caution in newborns given the uncertain risk of is obtained. If EEG is not overtly paroxysmal, phenobarbitone
hepatotoxicity following its use. is tapered and stopped. If EEG is overtly abnormal, the infant
is reassessed in the same manner at 3 months and then 3
Vigabatrin It has been used in neonates for refractory monthly till 1 yr of age (Fig. 2). The goal is to discontinue
seizures, primarily for infantile spasms. The dose is phenobarbitone as early as possible.
50 mg/kg/day.

Topiramate It shows promise in neonatal seizures because References


of its potential neuroprotective effect against injury caused
by seizures. Topiramate has been used for refractory 1. Volpe JJ. Neonatal Seizures .Neurology of the newborn, 5th ed.
Philadelphia: Saunders Elsevier; 2008. p. 20344.
infantile spasms in infants. The higher volume of distribu- 2. Tekgul H, Gauvreau K, Soul J, et al. The current etiologic profile
tion compared with other drugs requires higher initial and and neurodevelopmental outcome of seizures in term newborn
maintenance doses of approximately 3 mg/kg. infants. Pediatrics. 2006;117:127080.
3. Mizrahi EM, Kellaway P. Characterization and classification. In
Diagnosis and management of neonatal seizures. Lippincott-
Raven, 1998; 1535.
Other Therapies 4. Painter MJ, Scher MS, Stein MD, et al. Phenobarbitone compared
with phenytoin for treatment of neonatal seizures. N Engl J Med.
Pyridoxine A therapeutic trial of pyridoxine is reserved as a 1999;341:485899.
5. Rennie JM. Neonatal seizures. Eur J Pediatr. 1997;156:837.
last resort in refractory seizures. Intravenous route is the 6. Laroia N. Controversies in diagnosis and management of neonatal
preferred method; however, suitable IV preparations are not seizures. Indian Pediatr. 2000;37:36772.
universally available. Hence, intramuscular (IM) route may 7. Iype M, Prasad M, Nair PM, Geetha S, Kailas L. The newborn with
have to be used (1 ml of neurobion has 50 mg pyridoxine and seizuresa follow-up study. Indian Pediatr. 2008;45:74952.
8. Kumar A, Gupta A, Talukdar B. Clinico-etiological and EEG
1 ml each may be administered both the sides in either the
profile of neonatal seizures. Indian J Pediatr. 2007;74:337.
gluteal region or anterolateral aspect of thigh). It should ideally 9. Wical BS. Neonatal seizures and electrographic analysis: evaluation
be done in the NICU as hypotension and apnea can occur. and outcomes. Pediatr Neurol. 1994;10:2715.
204
205
206
207
208 124

Symposium on AIIMS Protocols in Neonatology III

Sepsis in the Newborn


M. Jeeva Sankar, Ramesh Agarwal, Ashok K Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar,
New Delhi, India

ABSTRACT
Infections are the single largest cause of neonatal deaths globally. According to National Neonatal Perinatal Database (2002-
03), the incidence of neonatal sepsis in India was 30 per 1000 live-births; klebsiella pneumoniae and staphylococcus aureus
were the two most common organisms isolated. Based on the onset, neonatal sepsis is classified into two major categories:
early onset sepsis, which usually presents with respiratory distress and pneumonia within 72 hours of age and late onset
sepsis, that usually presents with septicemia and pneumonia after 72 hours of age. Clinical features of sepsis are non-specific
in neonates and a high index of suspicion is required for the timely diagnosis of sepsis. Although blood culture is the gold
standard for the diagnosis of sepsis, culture reports would be available only after 48-72 hours. A practical septic screen for
the diagnosis of sepsis has been described and some suggestions for antibiotic use have been included in the protocol.
[Indian J Pediatr 2008; 75 (3) : 261-266] E-mail: vinodpaul@neonatalhealth.com

Key words : Infections; Newborn; Sepsis screen; Antibiotics

Sepsis is the commonest cause of neonatal mortality; it is from National Neonatal Perinatal Database (NNPD, 2002-
responsible for about 30-50% of the total neonatal deaths 03) is 30 per 1000 live births. The database comprising 18
in developing countries1,2. It is estimated that up to 20% of tertiary care neonatal units across India found sepsis to be
neonates develop sepsis and approximately 1% die of one of the commonest causes of neonatal mortality
sepsis related causes2. Sepsis related mortality is largely contributing to 19% of all neonatal deaths3. Septicemia
preventable with rational antimicrobial therapy and was the commonest clinical category with an incidence of
aggressive supportive care. 23 per 1000 live births while the incidence of meningitis
was reported to be 3 per 1000 live births. Among
intramural births, Klebsiella pneumoniae was the most
DEFINITION
frequently isolated pathogen (32.5%), followed by
Staphylococcus aureus (13.6%). Among extramural
Neonatal sepsis is a clinical syndrome characterized by neonates (referred from community/other hospitals),
signs and symptoms of infection with or without Klebsiella pneumoniae was again the commonest
accompanying bacteremia in the first month of life. It organism (27%), followed by Staphylococcus aureus (15%)
encompasses various systemic infections of the newborn and Pseudomonas (13%).3
such as septicemia, meningitis, pneumonia, arthritis,
osteomyelitis, and urinary tract infections. Superficial
CLASSIFICATION OF NEONATAL SEPSIS
infections like conjunctivitis and oral thrush are not
usually included under neonatal sepsis.
Neonatal sepsis can be classified into two major categories
depending up on the onset of symptoms4.
EPIDEMIOLOGY: INDIAN DATA
Early onset sepsis (EOS): It presents within the first 72
hours of life. In severe cases, the neonate may be
The incidence of neonatal sepsis according to the data symptomatic at birth. Infants with EOS usually present
with respiratory distress and pneumonia. The source of
infection is generally the maternal genital tract. Some
Correspondence and Reprint requests : Dr. Vinod K. Paul, maternal / perinatal conditions have been associated with
Professor, Department of Pediatrics, All India Institute of Medical an increased risk of EOS. Knowledge about these potential
Sciences, Ansari Nagar, New Delhi 110029, India risk factors would help in early diagnosis of sepsis. Based
[Received February 7, 2008; Accepted February 7, 2008] on the studies from India, the following risk factors seem

Indian Journal of Pediatrics, Volume 75March, 2008 261


209 125

M.J. Sankar et al

to be associated with an increased risk of early onset abdominal distension, paralytic ileus, necrotizing
sepsis.4, 5 enterocolitis (NEC).
1. Low birth weight (<2500 grams) or prematurity Hepatic: Hepatomegaly, direct hyperbilirubinemia
(especially with urinary tract infections).
2. Febrile illness in the mother with evidence of bacterial
infection within 2 weeks prior to delivery. Renal: Acute renal failure.

3. Foul smelling and/or meconium stained liquor. Hematological: Bleeding, petechiae, purpura.
4. Rupture of membranes >24 hours. Skin changes: Multiple pustules, abscess, sclerema,
5. Single unclean or > 3 sterile vaginal examination(s) mottling, umbilical redness and discharge.
during labor
6. Prolonged labor (sum of 1st and 2nd stage of labor > 24
INVESTIGATIONS
hrs)
7. Perinatal asphyxia (Apgar score <4 at 1 minute)
Since treatment should be initiated in a neonate suspected
Presence of foul smelling liquor or three of the above to have sepsis without any delay, only minimal and rapid
mentioned risk factors warrant initiation of antibiotic investigations should be undertaken.8
treatment. Infants with two risk factors should be
investigated and then treated accordingly. Blood culture: It is the gold standard for diagnosis of
septicemia and should be performed in all cases of
Late onset sepsis (LOS): It usually presents after 72 suspected sepsis prior to starting antibiotics. A positive
hours of age. The source of infection in LOS is either blood culture with sensitivity of the isolated organism is
nosocomial (hospital-acquired) or community-acquired the best guide to antimicrobial therapy. Therefore, it is
and neonates usually present with septicemia, pneumonia very important to follow the proper procedure for
or meningitis.6, 7 Various factors that predispose to an collecting a blood culture. The resident doctor/staff
increased risk of nosocomial sepsis include low birth should wear sterile gloves prior to the procedure and
weight, prematurity, admission in intensive care unit, prepare a patch of skin approx. 5-cm in diameter over the
mechanical ventilation, invasive procedures, proposed veni-puncture site. This area should be cleansed
administration of parenteral fluids, and use of stock thoroughly with alcohol, followed by povidone-iodine,
solutions. Factors that might increase the risk of and followed again by alcohol. Povidone-iodine should
community-acquired LOS include poor hygiene, poor be applied in concentric circles moving outward from the
cord care, bottle-feeding, and prelacteal feeds. In contrast, centre. The skin should be allowed to dry for at least 1
breastfeeding helps in prevention of infections. minute before the sample is collected. One-mL sample of
blood should be adequate for a blood culture bottle
CLINICAL FEATURES containing 5-10 mL of culture media. Since samples
collected from indwelling lines and catheters are likely to
Non-specific features: The earliest signs of sepsis are often be contaminated, cultures should be collected only from
subtle and nonspecific; indeed, a high index of suspicion a fresh veni-puncture site. All blood cultures should be
is needed for early diagnosis. Neonates with sepsis may observed for at least 72 hours before they are reported as
present with one or more of the following symptoms and sterile. It is now possible to detect bacterial growth within
signs (a) Hypothermia or fever (former is more common 12-24 hours by using improved bacteriological techniques
in preterm low birth weight infants) (b) Lethargy, poor such as BACTEC and BACT/ALERT blood culture
cry, refusal to suck (c) Poor perfusion, prolonged capillary systems. These advanced techniques can detect bacteria at
refill time (d) Hypotonia, absent neonatal reflexes (e) a concentration of 1-2 colony-forming unit (cfu) per mL.
Brady/tachycardia (f) Respiratory distress, apnea and Septic screen.9,10 All neonates suspected to have sepsis
gasping respiration (g) Hypo/hyperglycemia (h) should have a septic screen to corroborate the diagnosis.
Metabolic acidosis. However, the decision to start antibiotics need not be
Specific features related to various systems conditional to the sepsis screen result, if there is a strong
clinical suspicion of sepsis. The various components of the
Central nervous system (CNS): Bulging anterior septic screen include total leukocyte count, absolute
fontanelle, vacant stare, high-pitched cry, excess neutrophil count, immature to total neutrophil ratio,
irritability, stupor/coma, seizures, neck retraction. micro-erythrocyte sedimentation rate and C reactive
Presence of these features should raise a clinical protein (Table 1). The absolute neutrophil count varies
suspicion of meningitis. considerably in the immediate neonatal period and
Cardiac: Hypotension, poor perfusion, shock. normal reference ranges are available from Manroes
Gastrointestinal: Feed intolerance, vomiting, diarrhea, charts.11 The lower limit for normal total neutrophil

262 Indian Journal of Pediatrics, Volume 75March, 2008


210 126

Sepsis in the Newborn

TABLE 1. A Practical Sepsis Screen cerebrospinal fluid characteristics are unique in the
newborn period and normal values are given in Table 2.13
Components Abnormal value
Radiology: Chest X-ray should be considered in the
Total leukocyte count <5000/mm 3
Absolute neutrophil count Low counts as per Manroe presence of respiratory distress or apnea. An abdominal
chart11 for term and X-ray is indicated in the presence of abdominal signs
Mouzinhos chart12 for suggestive of necrotizing enterocolitis (NEC).
VLBW infants Neurosonogram and computed tomography (CT scan)
Immature/total neutrophil >0.2 should be performed in all patients diagnosed to have
Micro-ESR >15 mm in 1st hour
C reactive protein (CRP) >1 mg/dL meningitis.

(ESR, erythrocyte sedimentation rate)


Urine culture: In early onset sepsis, urine cultures have
a low yield and are not indicated. Urine cultures obtained
counts in the newborn begins at 1800/cmm, rises to 7200/ by suprapubic puncture or bladder catheterization have
cmm at 12 hours of age and then declines and persists at been recommended in all cases of LOS. Since the
1800/cmm after 72 hours of age. For very low birth procedures are painful and the yield is often poor, we do
weight infants, the reference ranges are available from not recommend a routine urine culture in neonates with
Mouzinhos charts. 12 The ratio of immature to total sepsis. However, neonates at risk for fungal sepsis and
neutrophils (I/T ratio) is 0.16 at birth and declines to a very low birth weight infants with poor weight gain
peak value of 0.12 after 72 hours of age. Presence of two should have a urine examination done to exclude urinary
abnormal parameters in a screen is associated with a tract infection (UTI). UTI may be diagnosed in the
sensitivity of 93-100%, specificity of 83%, positive and presence of one of the following: (a) >10 WBC/mm3 in a
negative predictive values of 27% and 100% respectively 10 mL centrifuged sample (b) >104 organisms /mL in
in detecting sepsis. Hence, if two (or more) parameters are urine obtained by catheterization and (c) any organism in
abnormal, it should be considered as a positive screen and urine obtained by suprapubic aspiration
the neonate should be started on antibiotics. If the screen
is negative but clinical suspicion persists, it should be
MANAGEMENT
repeated within 12 hours. If the screen is still negative,
sepsis can be excluded with reasonable certainty. For
early onset sepsis, documentation of polymorphs in the Supportive: Adequate and proper supportive care is crucial
neonatal gastric aspirate at birth could serve as a marker in a sick neonate with sepsis. He/she should be nursed in
of chorioamnionitis and it may be taken as one parameter a thermo-neutral environment taking care to avoid hypo/
of sepsis screen. hyperthermia. Oxygen saturation should be maintained
in the normal range; mechanical ventilation may have to
Lumbar puncture (LP): The incidence of meningitis in
be initiated if necessary. If the infant is hemodynamically
neonatal sepsis has varied from 0.3-3% in various
unstable, intravenous fluids should be administered and
studies. 3,6 The clinical features of septicemia and
the infant is to be monitored for hypo/hyperglycemia.
meningitis often overlap; it is quite possible to have
Volume expansion with crystalloids/colloids and
meningitis along with septicemia without any specific
judicious use of inotropes are essential to maintain normal
symptomatology. This justifies the extra precaution of
tissue perfusion and blood pressure. Packed red cells and
performing LP in neonates suspected to have sepsis. In
fresh frozen plasma might have to be used in the event of
EOS, lumbar puncture is indicated in the presence of a
anemia or bleeding diathesis.
positive blood culture or if the clinical picture is consistent
with septicemia. It is not indicated if antibiotics have been Antimicrobial therapy: There cannot be a single
started solely due to the presence of risk factors. In recommendation for the antibiotic regimen of neonatal
situations of late onset sepsis, LP should be done in all sepsis for all settings. The choice of antibiotics depends on
infants prior to starting antibiotics. Lumbar puncture the prevailing flora in the given unit and their
could be postponed in a critically sick neonate. It should antimicrobial sensitivity. This protocol does not aim to
be performed once the clinical condition stabilizes. The provide a universal recommendation for all settings but
TABLE 2. Normal Cerebrospinal Fluid Examination in Neonates13 lays down broad guidelines for the providers to make a
rational choice of antibiotic combination. Decision to start
CSF Components Normal range antibiotics is based upon clinical features and/ or a
Cells/mm3 8 (0-30 cells)
positive septic screen. However duration of antibiotic
PMN (%) 60% therapy is dependent upon the presence of a positive
CSF protein (mg/dL) 90 (20-170) blood culture and meningitis (Table 3).
CSF glucose (mg/dL) 52 (34-119)
CSF/ blood glucose (%) 51 (44-248) Indications for Starting Antibiotics: The indications for
starting antibiotics in neonates at risk of EOS include any
(PMN, polymorphonuclear cells; CSF, cerebrospinal fluid) one of the following:

Indian Journal of Pediatrics, Volume 75March, 2008 263


211 127

M.J. Sankar et al

TABLE 3. Duration of Antibiotic Therapy in Neonatal Sepsis (a) positive septic screen and/or
Diagnosis Duration (b) strong clinical suspicion of sepsis. (Fig. 1)
Meningitis (with or without positive blood/ 21 days Prophylactic Antibiotics: We do not use prophylactic
CSF culture) antibiotics in the following circumstances: infants on IV
Blood culture positive but no meningitis 14 days
fluids/TPN, meconium aspiration syndrome, and after
Culture negative, sepsis screen positive and 7-10 days
clinical course consistent with sepsis exchange transfusion(s). An exchange transfusion
Culture and sepsis screen negative, but 5-7 days conducted under strict asepsis (single use catheter, sterile
clinical course compatible with sepsis gloves, removal of catheter after the procedure) does not
increase the risk of sepsis and hence does not merit
(a) presence of 3 risk factors for early onset sepsis (see antibiotics. However, a messy exchange transfusion could
above) be treated with prophylactic antibiotics. In our unit,
ventilated neonates are treated with prophylactic
(b) presence of foul smelling liquor antibiotics for 5-7 days.
(c) presence of 2 antenatal risk factor(s) and a positive Choice of antibiotics: Empirical antibiotic therapy should
septic screen and be unit-specific and determined by the prevalent
(d) strong clinical suspicion of sepsis. spectrum of etiological agents and their antibiotic
sensitivity pattern. Antibiotics once started should be
The indications for starting antibiotics in LOS include:

Fig. 1. Protocol for sepsis

264 Indian Journal of Pediatrics, Volume 75March, 2008


212 128

Sepsis in the Newborn

modified according to the sensitivity reports. Guidelines For infections that are acquired during hospital stay,
for empirical antibiotic therapy have been provided in resistant pathogens are likely and a combination of
Table 4. ampicillin or cloxacillin with gentamicin or amikacin may
be instituted. In nurseries where this combination is
The empirical choice of antibiotics is dependent upon
ineffective due to the presence of multiple resistant strains
the probable source of infection. For infections that are
of klebsiella and other gram-negative bacilli, a
likely to be community-acquired where resistant strains
combination of a third generation cephalosporin
are unlikely, a combination of ampicillin or penicillin with
(cefotaxime or ceftazidime) with amikacin may be
gentamicin may be a good choice as a first line therapy.
appropriate. 3 rd generation cephalosporins have very

TABLE 4. Empirical Choice of Antibiotics for Treatment of Neonatal Sepsis

Clinical situation Septicemia & Meningitis


Pneumonia

FIRST LINE Penicillin or Ampicillin and Gentamicin Add Cefotaxime


Community-acquired (Resistant strains
are unlikely)
SECOND LINE Ampicillin or Cloxacillin and Add Cefotaxime
Hospital-acquired (Some strains Gentamicin or Amikacin
are likely to be resistant)
THIRD LINE Cefotaxime or Piperacillin-Tazobactam or
Hospital-acquired sepsis (Most Ciprofloxacin Same (Avoid Ciprofloxacin)
strains are likely to be resistant) andAmikacin;

Consider Vancomycin if MRSA is suspected.

TABLE 5. Drugs, Route of Administration and Doses of Common Antibiotics Used.

Drug Route Birth Weight 2000g Birth Weight >2000g


0-7 d >7 days 0-7 days >7 days

Amikacin I/V, I/M 7.5 q12h 7.5 q8h 10 q12h 10 q8h


Ampicillin
Meningitis I/V 100 q12h 100 q8h 100 q 8h 100 q6h
Others I/V, I/M 25 q12h 25 q8h 25 q8h 25 q6h

Cefotoxime
Meningitis I/V 50 q6h 50 q6h 50 q6h 50 q6h
Others I/M, I/V 50 q12h 50 q8h 50 q12h 50 q8h

Piperacillin+ I/V 50-100 q12h 50-100 q8h 50-100 q12h 50-100 q12h

Tazobactam
Ceftriaxone I/M, I/V 50 q24h 50 q24h 50 q24h 75 q24h
Ciprofloxacin I/V, PO 10-20 q24h 10-20 q24h 10-20 q12h 10-20 q12h

Cloxacillin
Meningitis I/V 50 q12h 50 q8h 50 q8h 50 q6h
Others I/V 25 q12h 25 q8h 25 q8h 25 q6h

Gentamicin
Conventional I/V, I/M 2.5 q12h 2.5 q8h 2.5 q12h 2.5 q8h
Single dose I/M 4 q24 h 4 q24 hr 5 q24h 5 q24h

Netilmicin I/V, I/M 2.5 q12h 2.5 q8h 2.5 q12h 2.5 q8h

Penicillin G (units/kg/dose)
Meningitis I/V 75,000 q12h 75,000 q8h 75,000 q8h 75,000 q6h
-100,000 -1,00,000 -1,00,000 -1,00,000
Others I/V, I/M 25,000 q12h 25,000 q8h 25,000 q8h 25,000 q6h
Vancomycin I/V 15 q12h 15 q8h 15 q12h 15 q8h

All doses are in mg/kg/dose;


(I/V, intravenous; I/M, intramuscular; PO, per-oral; h, hourly)
Protocol for sepsis

Indian Journal of Pediatrics, Volume 75March, 2008 265


213 129

M.J. Sankar et al

good CSF penetration and are traditionally thought to Granulocyte-Macrophage colony stimulating factor (GM-
have excellent antimicrobial activity against gram CSF): This mode of treatment is still experimental.19
negative organisms. Hence they were considered to be a
good choice for the treatment of nosocomial infections
and meningitis. However, recent reports suggest that at REFERENCES
least 60-70% of the gram-ve organisms are resistant to
them.14-16 More over, routine use of these antibiotics might
1. Bang AT, Bang RA, Bactule SB, Reddy HM, Deshmukh MD.
increase the risk of infections with ESBL (extended
Effect of home-based neonatal care and management of sepsis
spectrum beta-lactamase) positive organisms. Therefore it on neonatal mortality: field trial in rural India. Lancet 1999;
is preferable to use antibiotics such as piperacillin- 354 : 1955-19861.
tazobactam or methicillin/vancomycin in units with high 2. Stoll BJ. The global impact of neonatal infection. Clin Perinatol
incidence of resistant strains. A combination of 1997; 24 : 1-21.
3. Report of the National Neonatal Perinatal Database (National
piperacillin-tazobactam with amikacin should be
Neonatology Forum) 2002-2003.
considered if pseudomonas sepsis is suspected. Penicillin 4. Singh M, Narang A, Bhakoo ON. Predictive perinatal score in
resistant staphylococcus aureus should be treated with the diagnosis of neonatal sepsis. J Trop Pediatr 1994; 40 : 365-
cloxacillin, nafcillin or methicillin. Addition of an 368.
aminoglycoside is useful in therapy against 5. Takkar VP, Bhakoo ON, Narang A. Scoring system for the
staphylococcus. Methicillin resistant staphylococcus prediction of early neonatal infections. Indian Pediatr 1974; 11
: 597-600
aureus (MRSA) should be treated with a combination of 6. Baltimore RS. Neonatal nosocomial infections. Semin Perinatol
ciprofloxacin or vancomycin with amikacin. Ciprofloxacin 1998; 22 : 25-32.
has excellent activity against gram-negative organisms 7. Wolach B. Neonatal sepsis: pathogenesis and supportive
also; however, it does not have good CSF penetration. It therapy. Semin Perinatol 1997; 21 : 28-38.
may be used for the treatment of resistant gram-negative 8. Gerdes JS, Polin R. Early diagnosis and treatment of neonatal
sepsis. Indian J Pediatr 1998; 65 : 63-78.
bacteremia after excluding meningitis. For sepsis due to
9. Polinski C. The value of white blood cell count and
enterococcus, a combination of ampicillin and gentamicin differential in the prediction of neonatal sepsis. Neonatal Netw
is a good choice for initial therapy. Vancomycin should be 1996; 15 : 13-23
used for the treatment of enterococcus resistant to the first 10. Da Silva O, Ohlsson A, Kenyon C. Accuracy of leukocyte
line of therapy. indices and C-reactive protein for diagnosis of neonatal sepsis:
a critical review. Pediatr Infect Dis J 1995; 14 : 362-366.
The dosage, route, and frequency of commonly used 11. Manroe BL, Weinberg AG, Rosenfeld CR, Browne R. The
antimicrobial agents are given in table 5. neonatal blood count in health and disease. I.Refernce values
for neutrophilic cells. J Pediatr 1979; 95 : 89-98.
Reserve antibiotics: Newer antibiotics like aztreonam, 12. Mouzinho A, Rosenfeld CR, Sanchez PJ, Risser R. Revised
meropenem and imipenem are also now available in the reference ranges for circulating neutrophils in very-low-birth-
weight neonates. Pediatrics 1994; 94 : 76-82.
market. Aztreonam has excellent activity against gram-
13. Sarff LD, Platt LH, McCracken GH Jr. Cerebrospinal fluid
negative organisms while meropenem is effective against evaluation in neonates: Comparison of high-risk neonates
most bacterial pathogens except methicillin resistant with and without meningitis. J Pediatr 1976; 88 : 473-477.
staphylococcus aureus (MRSA) and enterococcus. 14. Upadhyay A, Aggarwal R, Kapil A, Singh S, Paul VK, Deorari
Imipenem is generally avoided in neonates because of the AK. Profile of neonatal sepsis in a tertiary care neonatal unit
reported increase in the incidence of seizures following its from India: A retrospective study. J Neonatology 2006; 20 : 50-
57.
use. Empirical use of these antibiotics should be avoided; 15. Deorari Ashok K. For the Investigators of the National
they should be reserved for situations where sensitivity of Neonatal Perinatal Database (NNPD). Changing pattern of
the isolated organism warrants their use. bacteriologic profile in Neonatal Sepsis among intramural
babies. J Neonatology 2006; 20 : 8-15.
Adjunctive therapy 16. Zaidi AK, Huskins WC, Thaver D, Bhutta ZA, Abbas Z,
Goldmann DA. Hospital-acquired neonatal infections in
Exchange transfusion (ET): Sadana et al17 have evaluated the developing countries. Lancet 2005; 365 : 1175-1188.
role of double volume exchange transfusion in septic 17. Sadana S, Mathur NB, Thakur A. Exchange transfusion in
neonates with sclerema and demonstrated a 50% septic neonates with sclerema: effect on immunoglobulin and
reduction in sepsis related mortality in the treated group. complement levels. Indian Pediatr 1997; 34 : 20-25.
We perform double-volume exchange transfusion with 18. Jenson HB, Pollock HB. The role of intravenous
immunoglobulin for the prevention and treatment of neonatal
cross-matched fresh whole blood as adjunctive therapy in
sepsis. Semin Perinatol 1998; 22 : 50-63.
septic neonates with sclerema. 19. Goldman S, Ellis R, Dhar V, Cairo MS. Rationale and potential
use of cytokines in the prevention and treatment of neonatal
Intravenous Immunoglobulin (IVIG): Non-specific pooled
sepsis. Clin Perinatol 1998; 25 : 699-710.
IVIG has not been found to be useful.18

266 Indian Journal of Pediatrics, Volume 75March, 2008


214
215
216
217
218 130
Indian J Pediatr (April 2012) 79(4):501509
DOI 10.1007/s12098-010-0279-7

SYMPOSIUM ON AIIMS PROTOCOLS IN NEONATOLOGY

Retinopathy of Prematurity
Deepak Chawla & Ramesh Agarwal & Ashok Deorari &
Vinod K. Paul & Parijat Chandra & Rajvardhan V. Azad

Received: 17 August 2010 / Accepted: 13 October 2010 / Published online: 27 October 2010
# Dr. K C Chaudhuri Foundation 2010

Abstract Retinopathy of prematurity (ROP) occurs due to in the protocol. Close co-operation between the ophthal-
abnormal proliferation of retinal vessels. The most impor- mologist and neonatologist is essential for successful
tant risk factors which predispose to development of ROP management of ROP.
include oxygen therapy, anemia needing blood transfusion,
sepsis and apnea. Very low birth weight neonates, those Keywords Prematurity . Risk factors . Retinopathy of
born at 32 week of gestation and other preterm neonates prematurity
with risk factors must be screened for ROP. As a general
rule first screening should be done at 1 month of postnatal
age. If screening detects ROP not needing treatment follow Introduction
up should be planned according to location and stage of
ROP. Better visual outcomes are observed with earlier Retinopathy of prematurity (ROP) is a vasoproliferative
treatment at lower threshold. Peripheral retinal ablation disorder of the retina among premature babies. ROP begins
with diode laser under adequate analgesia and sedation is to develop between 32 and 34 week after conception,
the preferred method for treatment of severe ROP. Guide- regardless of gestational age at delivery and has two distinct
lines regarding the procedure of dilatation, ophthalmic phases [1]. During the acute first phase, the normal
examination and treatment (if required) have been provided vasculogenesis of the retina is disturbed by the relative
hyperoxia of the extrauterine environment. This causes
D. Chawla vaso-obliteration and non-vascularization of some areas of
Department of Pediatrics, Government Medical College, the anterior retina [2]. The subsequent hypoxia causes a
Chandigarh, India second chronic phase, characterized by the proliferation of
P. Chandra : R. V. Azad
vascular and glial cells, arteriovenous shunt formation,
Department of Ophthalmology, Dr. R. P. Centre for Ophthalmic occasionally leading to involution or permanent cicatricial
Sciences, All India Institute of Medical Sciences, changes and visual impairment [3, 4]. In its more severe
Ansari Nagar, forms, it results in severe visual impairment or blindness,
New Delhi 110029, India
both of which carry a high financial cost for the community
R. Agarwal : A. Deorari : V. K. Paul but also a high individual cost by affecting the normal
Division of Neonatology & WHO Collaborating Centre for motor, language, conceptual, and social development of the
Training & Research in Newborn Care, Department of Pediatrics, child.
All India Institute of Medical Sciences,
Ansari Nagar,
New Delhi 110029, India
Epidemiology
A. Deorari (*)
Department of Pediatrics, All India Institute of Medical Sciences,
Studies from India reporting incidence of ROP provide
Ansari Nagar,
New Delhi 110029, India interesting insights. Although screening criteria differ
e-mail: ashokdeorari_56@hotmail.com across different units and time-periods, overall incidence
219 131
502 Indian J Pediatr (April 2012) 79(4):501509

of ROP varies from 20% to 52% (Table 1) with more recent detachment occurs and leads to visual loss in only a few
studies reporting lower rates of ROP ranging from 20% to percent of infants with severe ROP, and in most cases, ROP
30% [514]. An important lesson is learnt from units regresses spontaneously. The most conspicuous question is
reporting ROP in different time periods [9]. Initial low why ROP in some premature infants progresses despite
incidence of ROP rises with better screening protocols, rigorous and timely intervention, while in other cases with
availability of assisted ventilation services and survival of similar clinical characteristics it regresses. Evidence has
sicker, smaller neonates. In this phase, even sick but suggested that African-American infants are less prone to
relatively mature (late preterm) neonates have been reported severe outcome ROP than white infants and Alaskan
to develop ROP. This period is followed by gradual decline natives develop threshold ROP earlier than non-natives.
in incidence of ROP especially of more severe variety. This racial variation suggests that genetic, socioeconomic,
Prematurity is the single most important risk factor or dietary factors may be involved [15]. Poor early weight
responsible for retinopathy of prematurity. Incidence of gain in postnatal period has been observed to be a risk
ROP increases with decreasing gestation and birth weight. factor for development of severe ROP [16, 17].
However, not all preterm neonates develop ROP. Important
risk factors which increase the probability of developing
ROP are oxygen therapy, anemia needing blood transfu- Diagnosis
sion, sepsis and apnea [5, 7, 9, 11]. Nevertheless, a very
preterm extremely low birth weight neonate can develop A screening program is needed for early diagnosis and
ROP even without exposure to oxygen or presence of these secondary prevention of visual loss due to ROP in at-risk
risk factors. neonates. For documentation of the deterioration or
In general, more than 50% of preterm infants weighing regression of ROP, classification of degrees of severity for
less than 1,250 g at birth show evidence of ROP and about therapeutic interventions, and consistent reporting in clinical
10% of the infants develop severe ROP. However, retinal trials the International Classification of ROP (ICROP) is used

Table 1 Incidence and risk factors of retinopathy of prematurity

First author Year Screening criteria n Incidence Risk factors

Chaudhari S 2009 [5] Gestation 32 week or birth weight 552 ROP: 22.3% (123/552) Apnea, septicemia and
<1,500 g or additional risk factors Threshold ROP: oxygen therapy
7.4% (41/552)
Charan R 1995 [6] Birth weight 1,700 g and admitted 165 ROP: 47% (78/165) Not studied
to the neonatal unit
Gupta VP 2004 [7] Gestation <35 week or birth 60 ROP: 21.7% Apnea, sepsis,
weight <1,500 g oxygen therapy
Maheshwasri R 1996 [8, 9] Gestation <35 week or birth weight 66 ROP: 20% (13/66) Not studied
<1,500 g or premature neonate Threshold ROP:
needing oxygen for more than 24 h 6/66 (9.1%)
Dutta S 2004 [10] Gestation 32 week or birth weight 108 Not reported Administration of packed
1,700 g or premature babies of any cells and double-volume
gestation who have received prolonged exchange transfusion
oxygen therapy (30 days)
Rekha S 1996 [11] Gestation <35 week or birth 100 ROP: 46% (46/100) Anemia, duration of
weight <1,500 g Threshold ROP: oxygen therapy
9/100 (9%)
Gopal L 1995 [12] Premature neonates with birth 50 ROP: 38% (19/50) No formal analysis. All
weight <2,000 g Threshold ROP: with threshold ROP had
16% (8/50) received oxygen and 6 of 8
had received blood transfusion.
Varughese S 2001 [13] Gestation <34 week or birth 79 ROP: 52% (41/79) Not studied
weight <1,500 g Threshold ROP:
6.3% (5/79)
Sharma P 2009 [14] Preterm infants with birth weight 1,500 g 704 ROP: 11.9% (84/704) Respiratory distress syndrome
or gestation 32 week. Infants with birth 33 (4.7%) infants had
weight 1,5011,800 g or gestation 3334 severe ROP requiring
week screened if additional risk laser therapy
factors present
220 132
Indian J Pediatr (April 2012) 79(4):501509 503

(Table 2) [18]. ICROP describes vascularization of the retina appears at 36 week of post-menstrual age and threshold
and characterizes ROP by its position (zone, Fig. 1), severity disease at 37 week. Vascularization is complete by 40 week
(stage), and extent (clock hours). of gestation. Thus the crucial period for detection of ROP is
Some definitions used in relation to ROP are as follows: from 32 week to 40 week of post-menstrual period. The
critical phase is from 3435 week to 3738 week age
Aggressive Posterior ROP (AP-ROP) A rapidly progressing, during which the progression of the disease takes place and
severe form of ROP. If untreated, it usually progresses to stage treatment may have to be instituted. It may also be noted
5 ROP. The characteristic features of this type of ROP are its that ROP usually does not develop before 2 week of
posterior location, prominence of plus disease, and the ill- postnatal age.
defined nature of the retinopathy. This may not have classical
ridge or extraretinal fibrovascular proliferation. This rapidly Which Babies Should be Screened?
progressing retinopathy has been referred previously as type
II ROP and Rush disease.It is observed most commonly in Selecting neonates for screening depends on incidence of
Zone I, but may also occur in posterior Zone II. ROP at different gestation ages. Gestation and birth weight
cut-off for screening shifts lower as smaller and sicker
Threshold Disease Presence of stage 3 with plus disease in neonates start surviving. Based on current incidence and
Zone I or II, extending in 5 or more contiguous or risk factors reported in Indian literature following group of
8 cumulative clock hours (30 degree sectors). neonates should be screened.

Pre-threshold Disease Presence of less than threshold & Babies with Birth Weight <1,500 g
disease in Zone 1, or stage 2 plus disease in Zone 2, or & Babies Born at 32 week of Gestation
stage 3 (without plus) disease in Zone 2, or stage 3 plus & Selected preterm infants with a birth weight between
disease with extent less than that for threshold disease. 1,500 and 2,000 g or gestational age of more than 32 week
with sickness like need of cardiorespiratory support,
prolonged oxygen therapy, apnea of prematurity, anemia
Protocol for Screening
needing blood transfusion and neonatal sepsis or believed
by their attending pediatrician or neonatologist to be at
The aim of the screening programme is to detect ROP early,
high risk. This third criterion is important as it brings in
follow it up closely during its evolution and treat if it
many more larger babies into the screening guidelines
reaches a potentially serious severity.
without raising the screening parameters [19].
What is the Screening Window
When and How Often to Screen
Progression of ROP follows a distinct time-table according
to the post-menstrual age of the baby. Hardly any ROP is First screening examination should be carried out at
detected before 32 week of gestation. The median age for 31 weeks of gestation or 4 week of age, whichever is later
detection of stage 1 ROP is 34 week. Pre-threshold ROP (Table 3) [20]. A good rule to remember is first screening at

Table 2 Classification of retinopathy of prematurity

1. Location Zone I Circle with optic nerve at centre and a radius of twice the distance from optic nerve to macula
Zone II From edge of Zone I to the nasal ora serrata nasally and equator temporally
Zone III Lateral most crescent shaped area from Zone II to ora-serrata temporally
2. Severity Stage 1 Presence of thin white demarcation line separating the vascular from avascular retina
Stage 2 The line becomes prominent because of lifting of retina to form a ridge having height and width
Stage 3 Presence of extra retinal fibro-vascular proliferation with abnormal vessels and fibrous tissue arising
from the ridge and extending into vitreous
Stage 4 Partial retinal detachment; not involving macula (4A) or involving macula (4B)
Stage 5 Complete retinal detachment
3. Plus disease Presence of dilatation and tortuosity of posterior retinal vessels. Associated with vitreous haze,
pupillary rigidity
4. Extent Extent of involvement of the retina as expressed as clock hours (30 degree sectors)
5.Pre-plus disease Vascular abnormalities of the posterior pole that are insufficient for the diagnosis of plus disease
but that demonstrate more arterial tortuosity and more venous dilatation than normal
221 133
504 Indian J Pediatr (April 2012) 79(4):501509

Fig. 1 International classifica-


tion of retinopathy of
prematurity (ICROP) zones

1 month of postnatal age in babies born at >26 week of Stage 1 or 2 ROP: Zone III
gestation age. Regressing ROP: Zone III
Follow-up examinations should be recommended by the
Findings that suggest further examinations are not
examining ophthalmologist on the basis of retinal findings.
needed include:
& 1-week or less follow-up
& Zone III retinal vascularization attained without previ-
Stage 1 or 2 ROP: Zone I ous Zone I or II ROP
Stage 3 ROP: Zone II & Full retinal vascularization
& 1- to 2-week follow-up & Postmenstrual age of 45 week and no prethreshold
disease (defined as stage 3 ROP in Zone II, any ROP in
Immature vascularization: Zone Ino ROP Zone I) or worse ROP is present
Stage 2 ROP: Zone II & Regression of ROP
Regressing ROP: Zone I
& 2-week follow-up Where to Examine the Baby?
Stage 1 ROP: Zone II
Regressing ROP: Zone II Neonates are best examined in the neonatal unit itself under
supervision of attending pediatrician. It is not wise to
& 2- to 3-week follow-up
transport small babies to ophthalmic outpatient or ward for
Immature vascularization: Zone IIno ROP examination.

Table 3 Timing of first screening


eye examination based on gesta- Gestation age at birth (weeks) Age at initial examination
tional age at birth [20]
Postmenstrual age (wks) Chronological age (wks)

22 31 9
23 31 8
24 31 7
25 31 6
26 31 5
27 31 4
28 32 4
29 33 4
30 34 4
31 35 4
32 36 4
222 134
Indian J Pediatr (April 2012) 79(4):501509 505

How to Dilate the Pupils? clarity; followed by the posterior pole to look for plus
disease; followed by sequential examination of all clock
Pupils are dilated with Phenylephrine 2.5% and Tropica- hours of the peripheral retina. A scleral depressor is often
mide 0.5%. One drop of Tropicamide is instilled every 10 used to indent the eye externally to examine areas of
15 min for 4 times starting 1 h before the scheduled time interest, rotate and stabilize the eye.
for examination. This is followed by phenylephrine, one
drop just before examination. Phenylephrine is available in How to Record Findings During Screening?
10% concentration; it should be diluted 4 times before use
in neonates. Repeated instillation of phenylephrine is Ophthalmological notes should be made after each ROP
avoided for the fear of hypertension. examination, detailing zone, stage and extent in terms of
clock hours of any ROP and the presence of any pre-plus or
What Does the Examination Entail? plus disease (Fig. 2). These notes should include a
recommendation for the timing of the next examination (if
Screening of ROP involves indirect ophthalmoscopy using any) and be kept with the babys medical record.
20 D or 28/30 D lens by an experienced ophthalmologist.
After instilling a topical anesthetic drop like Proparacaine, a What Precautions are Taken During Examination?
wire speculum is inserted to keep the eye-lids apart. First
the anterior segment of the eye is examined to look for ROP screening examinations can have short-term effects on
tunica vasculosa lentis, pupillary dilation, and lens/media blood pressure, heart rate and respiratory function in the

Fig. 2 Retinopathy of prematu-


rity screening examination
record sheet Neonatal Ophthalmology Examination Record

Gestation: Birth Weight:


Name:

CR. No:

DoB: __/__/____ Gender: M/F weeks grams

Risks:

Stage 1: Stage 2: Stage 3: Stage 4/5: Laser: AP-ROP:

Date: R L

Postmenstrual Age:

Follow-up:

Examiner: Zone: Stage: Preplus: Y / N Zone: Stage: Preplus: Y / N


Plus: Y / N Plus: Y / N
Comments:
223 135
506 Indian J Pediatr (April 2012) 79(4):501509

premature baby [21]. The examinations should be kept as of an unfavourable outcome without treatment. The results
short as possible and precautions taken to ensure that showed an overall significant benefit for the early treatment
emergency situations can be dealt with promptly and of eyes with high-risk prethreshold disease. Based on
effectively. results of ETROP, two new terminologies have been
Eye examination during screening lasts several minutes suggested:
and may cause considerable pain to the neonate. A
systematic review and meta-analysis comprising four Type 1 ROP: & Zone I, any stage ROP with plus
studies has reported that oral sucrose reduces pain during disease
eye examination [21]. Of the two studies reporting the role & Zone I, stage 3 ROP with or
of topical proparacaine drops, one has observed significant without plus disease
pain reduction. & Zone II, stage 2 or 3 ROP with
Discomfort to the baby should be minimized by plus disease
administering oral sucrose just before examination, pre-
Type 2 ROP: & Zone I, stage 1 or 2 ROP without
treatment of the eyes with a topical proparacaine and
plus disease
swaddling the baby. Baby should not have been fed just
& Zone II, stage 3 ROP without plus
before examination to avoid vomiting and aspiration. Hand
disease
washing should be done and asepsis maintained.
Peripheral retinal ablation should be carried out for all
Use of Wide-Field Digital Camera (RetCam) for Screening cases with type 1 ROP and continued serial examinations
are advised for type 2 ROP.
A wide-field digital camera (RetCam) capable of retinal
imaging in preterm infants has been evaluated as an What are Treatment Modalities Available and What
alternative to indirect ophthalmoscopy for screening. are Their Advantages and Disadvantages?
Retinal images taken by camera can be stored, transmitted
to expert, reviewed, analyzed and sequentially compared Peripheral retinal ablation of avascular retina anterior to the
over time and are useful for telemedicine purposes. Studies ridge can be done by either cryotherapy or diode laser.
comparing RetCam with the indirect ophthalmoscope have Diode laser ablation has replaced cryotherapy due to lower
reported variable sensitivity and good specificity [22]. rate of postoperative ocular and systemic complications and
However, due to high cost and due to limitations in less damage to the adjacent tissues compared with
diagnostic sensitivity, specificity, and accuracy when image cryotherapy. Other advantages are that the laser spots are
quality is poor, it is not recommended to replace bedside visible during treatment minimizing the risk of missing
ophthalmoscopic examination. Digital fundus images can areas requiring treatment, and that laser equipment is
be used as a useful adjunct to conventional bedside ROP portable allowing use outside of the operating theatre. The
screening by indirect ophthalmoscopy. procedure can be carried out under general anesthesia or
under sedation depending on the feasibility and expertise.
Treatment for ROP should include the entire avascular
Treatment retina anterior to the ridge with burn spacing of between 0.5
to 1 burn-widths apart.
What is the Indication?
Pre-anesthetic Medication
Early Treatment of Retinopathy of Prematurity (ETROP)
trial recruited neonates at 26 centres in the US and Oral feeds should be discontinued 3 h prior to the procedure
compared early treatment of high-risk prethreshold with (Table 4). Baby should be started on intravenous fluids, and
conventional threshold treatment [23]. 401 babies meeting put on cardio-respiratory monitor. Dilatation of pupil is
the criteria for high-risk of an unfavourable outcome with done by using 0.5% Tropicamide and 2.5% phenylephrine
prethreshold in at least one eye, were randomized to receive as described in the section on protocol for screening.
either early or conventional treatment. The level of risk was
determined by a risk analysis programme which used, Anesthesia/Sedation
among other factors, degree of ROP (stage, zone and
presence of plus), rate of ROP progression, birthweight, Topical anaesthesia alone provides insufficient analgesia for
gestational age and ethnicity to classify eyes as at either ROP treatment and should not be used. Babies may be
high-risk (i.e. 15% chance) or low-risk (<15% chance) treated under adequate sedation and analgesia in an
224 136
Indian J Pediatr (April 2012) 79(4):501509 507

Table 4 Preparation for laser ablative therapy & Antibiotic drops (such as chloramphenicol) should be
Take consent instilled 68 hrly for 23 days.
Ensure good pupillary dilatation
Nil by mouth 3 h prior to procedure
Start on intravenous fluids
Prevention
Put on vital sign monitor/pulse oximeter
Judicious Oxygen Therapy
Warmer for maintaining temperature
Arrange equipment and check functioning thereof
Oxygen is a drug and it should be administered in a
Intubation equipment
quantity that is absolutely necessary. Each neonatal care
Endotracheal tubes No. 2.5, 3, 3.5
unit should have a written policy outlining appropriate use
Resuscitation bag and face masks
of oxygen therapy. If a preterm neonate born at <32 weeks
Oxygen delivery system
gestation needs resuscitation at birth, inhaled oxygen
Syringes
concentration (FiO2) should be titrated to prevent hyperoxia
Infusion pumps
and achieve gradual increase in oxygen saturation (70% at
Ventilator
3 min and 80% at 5 min after birth) [24]. During acute care
Arrange drugs, fill syringes in advance with drugs in appropriate
dilution and label them: morphine, midazolam, normal saline 10%
of a sick preterm neonate, ROP is more likely to develop if
dextrose, adrenaline partial pressure of oxygen in arterial blood is more than
80 mm Hg [25]. Oxygen level in blood should be
continuously monitored using pulse oximeter. It has been
observed that if oxygen saturation in a baby on oxygen
operation theatre if this can be arranged in a timely way. If therapy is kept between 85% and 93%, in about 90%
shifting to operation theatre is not possible or is causing samples partial pressure of oxygen is in desirable range
delay in treatment, babies may be treated more rapidly in (4080 mm Hg) [26]. Various observational studies have
the neonatal unit under adequate sedation and analgesia. reported that incidence and severity of ROP is lowered if
oxygen saturation targets are kept in desirable range and if
Procedure units implement written policies regarding oxygen admin-
istration and monitoring [27, 28]. During recovery phase of
Both the eyes can be treated at the same sitting time unless respiratory illness in preterm neonates, targeting higher
contraindicated by instability of the baby. If baby is not oxygen saturations has been associated with increased
tolerating the procedure, consider abandoning the proce- incidence/severity of bronchopulmonary dysplasia without
dure for the time being. Vital signs and oxygen saturation any benefit in stopping progression of ROP or improving
should be monitored very closely. growth and development [29, 30].

Monitoring After Laser Therapy Judicious Use of Blood Transfusions

After laser therapy, first examination should take place 5 Transfusion of packed RBCs is another risk factor of ROP.
7 days after the treatment and should be continued at least Adult RBCs are rich in 2,3 DPG and adult Hb binds less
weekly for signs of decreasing activity and regression. Re- firmly to oxygen, thus releasing excess oxygen to the
treatment should be performed usually 1014 days after initial retinal tissue. Packed cell transfusions should be given
treatment when there has been a failure of the ROP to regress. when hematocrit falls below following ranges: ventilated
babies 40%, babies with cardio-pulmonary disease but not
Post-operative Care on ventilators 35%, sick neonates but not having cardio-
pulmonary manifestations 30%, symptomatic anemia 25%
& The baby should be closely monitored. If condition and asymptomatic anemia 20%.
permits, oral feeds can be started shortly after the
procedure. Vitamin E Supplementation
& Premature babies, especially those with chronic lung
disease may have increase or re-appearance of apneic Very low birth weight neonates should receive 1525 IU of
episodes or an increase in oxygen requirement. There- vitamin E daily as supplement. However, higher doses
fore, they should be carefully monitored for 4872 h given by intravenous route have been associated with
after the procedure. increased risk of neonatal sepsis [31].
225 137
508 Indian J Pediatr (April 2012) 79(4):501509

Prenatal Steroids & Completeness of screening programme: Percentage of


babies <32 week GA or <1,501 g birthweight who
Use of prenatal steroids is a well-known approach to receive at least one ROP eye examination.
prevent respiratory distress and intraventricular hemor- & Timing of first screen: Percentage of babies <27 week
rhage, two important risk factors of ROP. Although there GA receiving a first ROP screening examination by
are some concerns that prenatal steroids may induce ROP, 4 week of postnatal age.
this is not borne out by other studies. The authors believe & ROP Treatment: Percentage of babies with any zone 1
prenatal steroids prevent acute illnesses in premature babies ROP who receive treatment.
and should be administered to all mothers with preterm & Timing of treatment: Percentage of babies needing ROP
labor between 2434 weeks of gestation. The preferred treatment for their ROP who are treated within 48 h of
preparation of steroids for prenatal used is betamethasone in the decision to treat being made.
two doses of 12 mg each given intramuscularly, 24 h apart.

Bevacizumab
References
Intravitreal injection of bevacizumab, a neutralizing anti-
VEGF molecule has been demonstrated to diminish the 1. Flynn JT. The premature retina: a model for the in vivo study of
neovascular response significantly in animal models [32]. molecular genetics? Eye. 1992;6:1615.
2. Kushner BJ, Essner D, Cohen IJ, Flynn JT. Retrolental fibroplasia
However, due to uncertainties with respect to the dosing, II. Pathologic correlation. Arch Ophthalmol. 1977;95:2938.
frequency, timing, and adjunct therapies to be used and 3. Chan-Ling T, Tout S, Hollander H, Stone J. Vascular changes and
potential to cause serious systemic adverse effects, use of their mechanisms in the feline model of retinopathy of prematu-
bevacizumab is not recommended outside the scope of rity. Invest Ophthalmol Vis Sci. 1992;33:212847.
4. Chan-Ling T, Gock B, Stone J. The effect of oxygen on
clinical trial.
vasoformative cell division. Evidence that physiological hypoxia
is the stimulus for normal retinal vasculogenesis. Invest Oph-
thalmol Vis Sci. 1995;36:120114.
Quality Improvement 5. Chaudhari S, Patwardhan V, Vaidya U, Kadam S, Kamat A.
Retinopathy of prematurity in a tertiary care centerincidence,
risk factors and outcome. Indian Pediatr. 2009;46:21924.
Roles and Responsibilities 6. Charan R, Dogra MR, Gupta A, Narang A. The incidence of
retinopathy of prematurity in a neonatal care unit. Indian J
Ophthalmol. 1995;43:1236.
& All units caring for babies at risk of ROP should have a 7. Gupta VP, Dhaliwal U, Sharma R, Gupta P, Rohatgi J.
written protocol in relation to the screening for, and Retinopathy of prematurityrisk factors. Indian J Pediatr.
treatment of, ROP. This should include responsibilities 2004;71:88792.
8. Maheshwari R, Kumar H, Paul VK, Singh M, Deorari AK, Tiwari
for follow-up of babies transferred or discharged from HK. Incidence and risk factors of retinopathy of prematurity in a
the unit before screening is complete. tertiary care newborn unit in New Delhi. Natl Med J India.
& If babies are transferred either before ROP screening is 1996;9:2114.
initiated or when it has been started but not completed, 9. Aggarwal R, Deorari AK, Azad RV, et al. Changing profile of
retinopathy of prematurity. J Trop Pediatr. 2002;48:23942.
it is the responsibility of the consultant neonatologist to 10. Dutta S, Narang S, Narang A, Dogra M, Gupta A. Risk factors of
ensure that the neonatal team in the receiving unit is threshold retinopathy of prematurity. Indian Pediatr. 2004;41:665
aware of the need to start or continue ROP screening. 71.
& Whenever possible ROP screening should be completed 11. Rekha S, Battu RR. Retinopathy of prematurity: incidence and
risk factors. Indian Pediatr. 1996;33:9991003.
prior to discharge. There should be a record of all 12. Gopal L, Sharma T, Ramachandran S, Shanmugasundaram R,
babies who require review and the arrangements for Asha V. Retinopathy of prematurity: a study. Indian J Ophthalmol.
their follow-up. 1995;43:5961.
& For babies discharged home before screening is com- 13. Varughese S, Jain S, Gupta N, Singh S, Tyagi V, Puliyel JM.
Magnitude of the problem of retinopathy of prematurity. experi-
plete, the first followup out-patient appointment must ence in a large maternity unit with a medium size level-3 nursery.
be made before hospital discharge and the importance Indian J Ophthalmol. 2001;49:1878.
of attendance explained to the parents. 14. Sharma P. DM dissertation: Risk factors for severe retinopathy of
prematurity in preterm low birth weight neonates Department of
Pediatrics. New Delhi: All India Institute of Medical Sciences
Auditing 2009 (Submitted for publication to Indian Journal of Pediatrics).
15. Saunders RA, Donahue ML, Christmann LM, et al. Racial
variation in retinopathy of prematurity. The cryotherapy for
Following outcomes should be regularly audited in units retinopathy of prematurity cooperative group. Arch Ophthalmol.
with ROP screening and treatment programme. 1997;115:6048.
226 138
Indian J Pediatr (April 2012) 79(4):501509 509

16. Hellstrom A, Hard AL, Engstrom E, et al. Early weight gain 25. Flynn JT, Bancalari E, Snyder ES, et al. A cohort study of
predicts retinopathy in preterm infants: new, simple, efficient transcutaneous oxygen tension and the incidence and severity
approach to screening. Pediatrics. 2009;123:e63845. of retinopathy of prematurity. N Engl J Med. 1992;326:1050
17. Hellstrom A, Ley D, Hansen-Pupp I, et al. New insights into the 4.
development of retinopathy of prematurityimportance of early 26. Castillo A, Sola A, Baquero H, et al. Pulse oxygen saturation
weight gain. Acta Paediatr 2010;99:5028. levels and arterial oxygen tension values in newborns receiving
18. The International Classification of Retinopathy of Prematurity oxygen therapy in the neonatal intensive care unit: is 85% to 93%
revisited. Arch Ophthalmol. 2005;123:99199. an acceptable range? Pediatrics. 2008;121:8829.
19. Azad R, Chandra P, Patwardhan SD, Gupta A. Importance of 27. Tin W, Milligan DW, Pennefather P, Hey E. Pulse oximetry,
the third criterion for retinopathy of prematurity screening in severe retinopathy, and outcome at one year in babies of less than
developing countries. J Pediatr Ophthalmol Strabismus. 28 weeks gestation. Arch Dis Child Fetal Neonatal Ed. 2001;84:
2009;46:3324. F10610.
20. Screening examination of premature infants for retinopathy of 28. Chow LC, Wright KW, Sola A. Can changes in clinical practice
prematurity. Pediatrics. 2006;117:572576. decrease the incidence of severe retinopathy of prematurity in very
21. Sun X, Lemyre B, Barrowman N, OConnor M. Pain management low birth weight infants? Pediatrics. 2003;111:33945.
during eye examinations for retinopathy of prematurity in preterm 29. Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM. Oxygen-
infants: a systematic review. Acta Paediatr. 2010;99:32934. saturation targets and outcomes in extremely preterm infants. N
22. Kemper AR, Wallace DK, Quinn GE. Systematic review of digital Engl J Med. 2003;349:95967.
imaging screening strategies for retinopathy of prematurity. 30. Supplemental Therapeutic Oxygen for Prethreshold Retinopathy
Pediatrics. 2008;122:82530. Of Prematurity (STOP-ROP), a randomized, controlled trial. I:
23. Early Treatment For Retinopathy Of Prematurity Cooperative Group. primary outcomes. Pediatrics. 2000;105:295310.
Revised indications for the treatment of retinopathy of prematurity: 31. Brion LP, Bell EF, Raghuveer TS. Vitamin E supplementation for
results of the early treatment for retinopathy of prematurity prevention of morbidity and mortality in preterm infants.
randomized trial. Arch Ophthalmol. 2003;121:168494. Cochrane Database Syst Rev. 2003:CD003665.
24. Finer N, Leone T. Oxygen saturation monitoring for the preterm 32. Micieli JA, Surkont M, Smith AF. A systematic analysis of the off-
infant: the evidence basis for current practice. Pediatr Res. label use of bevacizumab for severe retinopathy of prematurity. Am J
2009;65:37580. Ophthalmol. 2009;148:53643. e532.
227
228
229
230
231 139

Symposium on AIIMS Protocols in Neonatology IV

Parenteral Nutrition
Deepak Chawla, Anu Thukral, Ramesh Agarwal, Ashok K. Deorari and Vinod K. Paul

Division of Neonatology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New
Delhi, India

ABSTRACT
Nutritional insufficiency, leading to early growth deficits has long-lasting effects, including short stature and poor
neurodevelopmental outcomes. Early enteral feeding is commonly limited by immaturity of gastrointestinal motor function in
preterm neonates. To ensure that a stressed premature infant receives an adequate but not excessive amount of glucose,
the amount of carbohydrate delivered in the form of dextrose is commonly initiated at the endogenous hepatic glucose
production and utilization rate of 4 to 6 mg/kg/min; and 8 to 10 mg/kg/min in ELBW infants. The early provision of protein is
critical to attain positive nitrogen balance and accretion as premature babies lose ~1% of their protein stores daily. Aminoacid
can be used at concentrations of 3-3.5g/kg/day and lipid at 3.5-4g/kg/day as long as the fat intake remains less than 60% of
nonprotein calories. Sodium, potassium, chloride, calcium, magnesium and phosphorus need to be provided in PN solution
as per their daily needs. Hospital-acquired infection (HAI) is a major complication of PN. All efforts should be made to avoid
it. [Indian J Pediatr 2008; 75 (4) : 377-383] E-mail: ashokdeorari_56@hotmail.com

Key words : Parentral nutrition neonates; Lipid; Glucose; Aminoacids; Non protein calories

The goal of nutrition management in neonates, especially fact that with availability of optimum nutrient sources,
very low birth weight (VLBW) infants is the achievement early administration of PN is now both safe and
of postnatal growth at a rate that approximates the efficacious.
intrauterine growth of a normal fetus at the same
postconceptional age. Although, this is best achieved Indications
with optimal enteral nutrition, early enteral feeding is
PN should be considered in neonates who are not on
commonly limited by immaturity of gastrointestinal
significant enteral feeds for more than 3-5 days or are
motor function, manifested principally as delayed
anticipated to be receiving less than 50% of total energy
stomach emptying, gastro-esophageal reflux, abdominal
requirement by day 7 of life (Table 2).
distension, and infrequent stooling. Nutritional
insufficiency, leading to early growth deficits has long- Energy
lasting effects, including short stature and poor
neurodevelopmental outcomes (Table 1) 1. Likewise, Determination of appropriate energy and nutritional
establishing an alternative source of nutrition becomes a requirements of a newborn infant is the first step in
life-sustaining intervention in surgical neonates with formulating PN. Preterm infants have very low energy
congenital or acquired disease causing gastrointestinal reserves due to low amounts of fat as well as low
failure. Parenteral nutrition (PN), first introduced in the glycogen reserves. Cessation of placental nutrient
late 1960s, has been used extensively in neonatal intensive supplies and low reserves results in metabolic shock and
care units (NICU) of developed countries. With protein catabolism if appropriate amounts of energy and
improving survival of extremely premature neonates and proteins are not provided soon after birth.2 A daily energy
increasing number of NICUs in India, need of parenteral intake of 120-130 kcal/kg is needed to meet the metabolic
nutrition is being widely recognized among health care demands of a healthy premature neonate and to allow for
providers. This recognition of need is accompanied by the growth rate comparable to intrauterine growth rate (Table
3)3. Energy requirement of term neonate is 100-120 kcal/
kg/day. Energy intake of sick neonates (e.g., acute
respiratory illness, chronic lung disease, necrotizing
Correspondence and Reprint requests : Dr. Ashok K. Deorari, enterocoliltis) is not exactly known but is likely to be near
Professor, Department of Pediatrics, All India Institute of Medical upper limits of the energy requirement of preterm infant.3
Sciences, Ansari Nagar, New Delhi 110029, India.
10% dextrose solution provides 0.34 kcal/ml. 10% lipid
[Received March 13, 2008; Accepted March 13, 2008]

Indian Journal of Pediatrics, Volume 75April, 2008 377


232 140

D. Chawla et al

solution provides 0.9 kcal/ml and 20% lipid solution receive TPN suggest that protein accretion occurs by
provides 1.1 kcal/ml. If sufficient amount of non-protein amino acid stimulation of protein synthesis rather than by
energy is not provided, amino acids are catabolised for suppression of protein breakdown. 4, 5 Protein
energy production. Adequate balance between nitrogen requirements for the neonate tend to be inversely related
and non-protein energy sources (Protein/Energy ratio: 3- to gestational age and size due to more rapid growth rates
4 gm/100 kcal) is needed to promote protein accretion.2 and greater protein losses in the smaller, more premature
Balance between carbohydrates and fat is needed to infants.6 The early provision of protein is critical to attain
prevent excessive fat deposition and excessive production positive nitrogen balance and accretion, as premature
of CO2. The ideal distribution of calories should be 50-55% babies lose ~1% of their protein stores daily.7 Benefits
carbohydrate, 10-15% proteins and 30-35% fats. include improvement in nitrogen balance, stable plasma
AA profile and better growth in neonatal period. AA
Amino acids solutions are available as 10% and 20% preparations
(appendix).
Amino acids (AA) are building blocks of the body. The
amount needed, calculated using factorial approach is Carbohydrates
3.0-3.5 gm/kg/day (0.3 gm/kg/d to mimic intrauterine
Carbohydrates are the main energy substrate for the
changes in body composition + 2.2 to 2.5 gm/kg/d for
neonates receiving PN. Although, glucose is routinely
normal growth + 1 gm/kg/d obligatory urinary and
administered to VLBW infants beginning soon after birth,
dermal protein loss). An optimal AA solution should
the main objective of this established practice is to
contain essential (valine, leucine, isoleucine, methionine,
maintain euglycemia. During PN, glucose infusion rate is
phenylalanine, threonine, lysine and histidine) and
gradually advanced and objective is the achievement of
conditionally essential (cysteine, tyrosine, glutamine,
higher energy intake. Glucose is available as 5%, 10%,
arginine, proline, glycine and taurine) AAs, should not
25% and 50% solutions.
have excess of glycine and methionine and should not
contain sorbitol. AA infusion can be started between 0 To ensure that a stressed premature infant receives an
and 36 h of birth. The amount started on day 1 of PN has adequate but not excessive amount of glucose, the
varied from 0.5 to 3.0 gm/kg/d in different studies. To amount of carbohydrate delivered in the form of dextrose
avoid negative protein balance, one should start with at is commonly initiated at the endogenous hepatic glucose
least 1 to 1.5 gm/kg/d and then increase by 1 gm/kg/d production and utilization rate of 4 to 6 mg/kg/min; and
to maximum of 3.5 gm/kg/d. With this regimen, there 8 to 10 mg/kg/min in ELBW infants. It provides 40 to 50
have been no reports of side effects like metabolic kcal/kg/d and preserves carbohydrate stores.
acidosis, hyperaminoacidemia, azotemia or
Frequently smaller, more unstable premature infants
hyperammonaemia.1 Studies in preterm babies who

TABLE 1. Consequences of Suboptimal Nutrient Intake 1

Short term Long term

Increased vulnerability to infections Poor growth


Free-radical mediated damage Poor neurodevelopment outcome
Greater need of ventilator support Susceptibility to cardiovascular diseases
Reduced cell growth in specific organ systems (heart, kidney, pancreas)

TABLE 2. Indications of Parenteral Nutrition

Indications of parenteral nutrition

Birth weight less than 1000 gm


Birth weight 1000-1500 gm and anticipated to be not on significant feeds for 3 or more days
Birth weight more than 1500 gm and anticipated to be not on significant feeds for 5 or more days
Surgical conditions in neonates: Necrotizing enterocolitis, Gastroschisis, Omphalocele, Tracheo-esophageal fistula, Intestinal atresia,
Mal-rotation, Short bowel syndrome, and Meconium ileus

TABLE 3. Daily Energy Intake Recommended for Preterm Infants3

Committee Recommended energy intake (kcal/kg/day)

American Academy of Pediatrics 105-130


Canadian Pediatric Society 105-135
European Society of Gastroenterology and Nutrition 98-128
Life Sciences Research Office 110-135

378 Indian Journal of Pediatrics, Volume 75April, 2008


233 141

Parenteral Nutrition

develop hyperglycemia due to decreased insulin kg per day by day 4, has been well tolerated without
production and insulin resistance. Glucose infusion rates noticeable adverse effects. Consider avoiding lipids for a
(GIR) for these babies may need to be limited to 4 mg/ short period in the unstable, late preterm infant who has
kg/min or less, while larger preterm infants or term evidence of increased PVR. Lipids may be restricted in
infants can often tolerate up to 8 mg/kg/min initially.8, 9 patients with hyperbilirubinemia in minimum amounts
Once the GIR supports acceptable serum glucose values, that will provide only the essential fatty acids. Because
it is advanced in a gradual, stepwise fashion (0.5 to 1 mg/ both lipids and bilirubin are transported in the blood by
kg/min) to a suggested maximum glucose oxidative rate albumin, lipids competing for binding sites on albumin
for neonates of 12 to 13 mg/kg/min to support growth may result in insufficient binding of bilirubin to facilitate
and maintained there unless serum glucose values change excretion. 16 Persistently high bilirubin values may
significantly. increase the risk of kernicterus from the deposition of
bilirubin in brain cells. A free fatty acid to albumin ratio
Excessive carbohydrate delivery above the amount
(FFA:albumin) greater than 6:1 is thought to be clinically
that can be oxidized for energy and glycogen storage will
significant.
lead to an increase in basal metabolic rate,10 fat deposition,
cholestasis,11 hepatic steatosis,12 or overfeeding. IVL emulsions are aqueous suspensions containing
neutral triglycerides derived from soybean, safflower oil,
Insulin has been used along with glucose to serve two
egg yolk to emulsify and glycerine to adjust tonicity.
distinct purposes. One is to manage hyperglycemia if
Hydrolysis of triglycerides by hepatic and lipoprotein
infant is developing high glucose levels despite glucose
lipase results in formation of free fatty acids. IVL
infusion rate of 4-6 mg/kg/minute. In this case, insulin is
emulsions are available in two strengths: 10% and 20%
stopped as soon as euglycemia is achieved. Second
(Appendix). Use of 20% lipid emulsion is preferable to a
objective is to achieve higher glucose infusion rate and
10% solution to decrease the risk of hypertriglyceridemia,
promote growth. Later approach does not result in
hypercholesterolemia, and hyperphospholipidemia
increased AA accretion, can induce lactic acidosis and is
(Fig. 1). When lipids are exposed to light, they form
therefore not recommended.13
potentially toxic lipid hydroperoxides. Hence lipid
Lipids

Lipids are essential components of parenteral nutrition for


preterm infants to provide essential fatty acids (EFAs) and
to meet high energy needs. Parenteral lipids are an
attractive source of nutrition in the first postnatal days
because of their high energy density, energy efficiency,
isotonicity with plasma, and suitability for administration
through a peripheral vein. Parenteral lipid emulsions
enable the delivery of fat-soluble vitamins. Even a short
delay of 3 to 7 days in supplying lipids to parenterally fed
preterm infants leads to biochemical EFA deficiency.14
Such deficiency increases antioxidant susceptibility in
preterm infants. EFA deficiency can be prevented with
Fig. 1. Metabolism of intravenous lipid emulsions. Lipoprotein X
introduction of as little as 0.5 to 1.0 g/kg per day of lipid
inhibits the clearance of remnant particles
infusion. Fluid-restricted, growth-compromised patients
or those limited to peripheral line access may require as Appendix
high as 3.5 to 4 grams fat/kg/d to achieve adequate Sources of Parenteral Solutions
energy for growth and protein sparing. This intake is
Component Source Concentration
appropriate as long as the fat intake remains less than 60%
of nonprotein calories.15 Proteins Aminoven 6% and 10%
Primene
The routine use of intravenous lipid (IVL) emulsions Lipids Intralipid 10%, 10% PLR
has not been universally accepted in critically ill, (phospholipids reduced), 20%
ventilated VLBW infants because of potential Glucose Dextrose 5%, 10%, 25%, 50%
complications like adverse effects on gas exchange and NaCl NaCl 0.9%, 3%
KCl KCl 15%
displacement of bilirubin from albumin. Proper use of IVL
Calcium Calcium gluconate 10%
emulsions includes slow infusion rates (<0.15 g/kg per Multivitamin Adult MVI -
hour), slow increases in dosage, and avoidance of unduly Trace elements Celcel -
high doses (>3.0 g/k per day). Studies have shown that TMA
administration of IVLs, beginning on day 1 at a dose of 1.0 Magnesium Magnesium sulfate 50%
g/kg per day and increasing in stepwise fashion to 3.0 g/ sulfate

Indian Journal of Pediatrics, Volume 75April, 2008 379


234 142

D. Chawla et al

syringes and tubing should be covered by wrapping it in TABLE 4. Daily Requirement of Minerals
aluminum foil.
Mineral Requirement
Lipids may be restricted in patients with
Sodium 0-3 meq/kg/d (1st week of life)
hyperbilirubinemia in minimum amounts that will 3-6 meq/kg/d (beyond 1 st week)
provide only the essential fatty acids. Some clinicians will Potassium 0-2 meq/kg/d (1st week of life)
reduce or withhold lipids if rising bilirubin trends 1-3 meq/kg/d (beyond 1 st week)
approach levels requiring exchange transfusions. Because Chloride 2-3 meq/kg/d
both lipids and bilirubin are transported in the blood by Calcium 150-200 mg/kg/day
Magnesium 15-25 mg/d
albumin, lipids competing for binding sites on albumin
Phosphate 20-40 mg/kg/d
may result in insufficient binding of bilirubin to facilitate
excretion. 16 Persistently high bilirubin values may
TABLE 5. Recommended Vitamin Intake
increase the risk of kernicterus from the deposition of
bilirubin in brain cells. A free fatty acid to albumin ratio Vitamin Term (daily dose) Preterm (dose/kg/day)
(FFA:albumin) greater than 6:1 is thought to be clinically
Vitamin A (IU) 2300 1640
significant.
Vitamin D (IU) 400 160
Minerals Vitamin E (IU) 7 2.8
Vitamin K (g) 200 80
Sodium, potassium, chloride, calcium, magnesium and Vitamin B6 (g) 1000 180
phosphorus need to be provided in PN solution as per Vitamin B12 (g) 1 0.3
Vitamin C (mg) 80 25
their daily needs (Table 4). Except phosphate, all these
Biotin (g) 20 6
minerals are easily available in India. Sodium, potassium, Folic acid (g) 140 56
and chloride are essential to life and requirements are Niacin (mg) 17 6.8
dependent on obligatory losses, abnormal losses, and Pantothenic acid (mg) 5 2
amounts necessary for growth. Estimated and advisable Riboflavin (g) 1400 150
intakes are based on accretion studies and urinary and Thiamin (g) 1200 350
fecal losses from balance studies completed in the late
1970s. 17 Dosage of zinc to be provided is 150-400 microgram/kg/
d even with short-term PN, but a suitable preparation is
Calcium, phosphorus, and magnesium are the most difficult to find in Indian market.
abundant minerals in the body. They are closely
interrelated to each other in metabolism, the formation of Fluids
tissue structure, and function.
Intravenous fluid is the carrying medium for PN. It is
Vitamins started at 60-80 ml/kg/d and advanced by 15-20 ml/kg/
d to maximum of 150 ml/kg/d by end of first week of
Vitamins are added in PN solution to meet the daily life. Fluid therapy is regulated by monitoring hydration
requirement (Table 5). Separate preparations of fat-soluble status of the infant (weight gain/loss, serum sodium,
and water-soluble vitamins suitable for neonates are not urinary specific gravity, urine output and osmolality of
available in India. Multivitamin injection (MVI), when plasma and urine.
added in a dose of 1.5 ml/kg to AA-glucose solution
meets the need of vitamin A and most other vitamin. Evidence-based recommendations
Furthermore, intravenous vitamin delivery may be less Evidence-based recommendations for use of PN
due to photodegradation of vitamins A, D, E, K, B2, B6, B12, constituents are summarized in Table 6.
C, and folic acid and adsorption of vitamins A, D, and E
into the vinyl delivery bags and tubing. Vitamin K needs Dispensing PN solution
to be given separately as weekly intramuscular injections. In developed countries PN solution is prepared by central
Although vitamin B12 is not present in MVI, its deficiency pharmacy and delivered ready to be used. But this facility
is not manifested unless the neonate is on long-term PN. is usually not available in most of Indian hospitals and
physicians and nurses have to chart and prepare PN.
Trace elements Steps for calculation and preparing PN are as follows (a
Trace elements like zinc, copper, manganese, selenium, PN chart is provided in appendix):
fluorine and iodine should be provided in PN solutions. Determine total fluid requirement for the day
Zinc is universally recommended from day one of TPN, Subtract amount of fluid to be used for medications
whereas the other trace minerals are generally provided (e.g., diluting and infusing antibiotics) and enteral
after two, four, or 12 weeks of TPN without any feeds
appreciable enteral feeding. Copper, selenium,
molybdenum, and iron can be delivered separately also. Plan AA, IVL and glucose to be given over 24 h

380 Indian Journal of Pediatrics, Volume 75April, 2008


235 143

Parenteral Nutrition

TABLE 6. Evidence-based Recommendations for Parenteral Nutrition

Component Recommendations

Fluids Day 1: 60-80 mL/kg/d.


Postnatal weight loss of 5% per day to a maximum of 15% is acceptable. This is achieved by progressively
increasing the fluid intake to 120-150 mL/kg/d by one week of age.
Energy An intake of 50 kcal/kg/d is sufficient to match ongoing expenditure, but it does not meet additional
requirements of growth.
The goal energy intake is 120 kcal/kg/d (higher in infants with chronic lung disease)
Protein Optimal parenteral amino acid intake is 3.5 g/kg/d.
Parenteral amino acids can begin from day 1 at 1-1.5 gm/kg/d
Carbohydrates From day 1, 6 mg/kg/min can be infused, increased by 2 mg/kg/min/d to 12-14 mg/kg/min and adjusted to
maintain euglycemia
Insulin is only used in infants who continue to have hyperglycemia associated with glycosuria and osmotic
dieresis even after the glucose intake has been reduced to 6 mg/kg/min. Insulin is given as a continuous infusion
commencing at a rate of 0.05 units/kg/h, increasing as required for persistent hyperglycemia.
Fat Intravenous fat, 1 g/kg/d can be started from day 1, at the same time as when intravenous amino acids are
started. This is increased to 2 g/kg/d and 3 g/kg/d over the next two days.
It is delivered as a continuous infusion of 20% intravenous fat via a syringe pump, separate from the infusate
containing the amino acids and glucose. The syringe and infusion line should be shielded from ambient light.
Minerals and Trace Minerals should include: sodium, chloride, potassium, calcium, phosphorus, magnesium.
Elements Trace elements should include: zinc, copper, selenium, manganese, iodine, chromium, and molybdenum.
Vitamins Vitamins must be added to the fat emulsion to minimize loss during administration due to adherence to tubing
and photo-degradation.

Take IVL suspension in one syringe and add MVI in Short-term PN can be given through peripheral venous
to it. line. Another attractive option in neonates is central line
In second syringe mix AA, dextrose, electrolytes and inserted through umbilical vein. Position of central line
trace elements should be confirmed by X-ray before starting infusion
through it. The venous access used for PN should not be
IVL+MVI suspension is infused separately from AA-
interrupted for giving antibiotics or other medications.
Glucose-Minerals solution, although they can be
For this a separate intravenous line should be established.
mixed at the site of infusion using a three-way
adapter. Peripheral access offers the advantage of a lower risk of
For calculating amount of each PN component, use infection due to the greater distance of the catheter from
following formula: the central circulation as well as a smaller risk of
mechanical complications. However, nutrition delivery is
Amount to be given per kg body weight Body weight limited with peripheral lines due to constraints created by
Amount of component =
Strength of solution a solutions osmolarity. Osmolality refers to the number of
particles per weight of water in kilograms and is typically
For example, for a baby weighing 1.5 kg to be given 3 used to describe enteral feedings and a standard in
meq/kg of sodium, amount of 3% NaCl to be used is: describing blood. 18 Osmolarity refers to the number of
millimoles of liquid or solid in a liter of solution and is the
3 meq/kg 1.5 kg
Amount of 3% NaCl = = 9 ml preferred term for TPN. The sample worksheet of TPN is
0.5 meq/ml shown in Table 7. In peripheral access concentration
greater than 900-1000 mOsm/ L are to be avoided. With
central access, solutions greater than 1,000 mOsm/L are
Route of administration
acceptable.
PN can be delivered through peripheral or central venous
Monitoring and complications
lines. The limiting factor in deciding route of delivery is
osmolarity of the AA-glucose solution which is dependent Meticulous monitoring is needed in a neonate receiving
on dextrose concentration. A dextrose concentration PN. Monitoring protocol and its rationale is summarized
greater than 12.5% has an acidic pH and can be irritating in Table 8. Monitoring should be more frequent in the
to the peripheral veins. In addition to dextrose, initial stages. Once a steady metabolic stage has been
electrolytes and minerals added to the solution increase achieved, monitoring can be reduced to once a week.
the osmolarity of the solution. Hypertonic solution need
to be administered through central venous line. Increasing Complications of PN can be nutrient-related or venous
use of peripherally inserted central catheters (PICC) has access-related. Nutrient related complications include
facilitated administration of PN while avoiding many hypoglycemia (plasma sugar < 54 mg%), hyperglycemia
potential complications of surgically inserted central lines. (plasma sugar > 150 mg%) (glucose related); azotemia,

Indian Journal of Pediatrics, Volume 75April, 2008 381


236 144

D. Chawla et al

TABLE 7. TPN Worksheet

TABLE 8. Monitoring Schedule for a Neonate on Parenteral metabolic acidosis (protein-related); hypertriglyceridemia
Nutrition
(triglyceride > 200 mg/dl) (lipid-related), cholestasis and
Parameter Frequency trace element deficiency. Most of these complications can
be avoided by proper monitoring and provision of
Blood sugar 2-3 times a day while increasing
nutrients. PN-related cholestasis is usually complication
glucose infusing rate
Once a day while on stable glucose of long-term PN and can be avoided by provision of at
infusion rate least minimal-enteral nutrition. Catheter-related
Urine sugar Each urine sample ideally complications include dislodgement and infection.
Serum electrolytes Twice a week initially, then weekly
Blood urea Twice a week initially, then weekly Prevention of infection
Calcium, magnesium Weekly
and phosphorous Hospital-acquired infection (HAI) is a major complication
Serum albumin Weekly
of PN. All efforts should be made to avoid HAI.
Packed cell volume Weekly
Liver function tests Weekly Aseptic precautions during preparation of PN
Serum triglycerides Weekly
ANTHROPOMETRY Use of laminar flow
Weight Daily at the same time No compromise on disposables
Length Weekly
Trained staff
Head circumference Weekly
FLUID ml/kg/day - Daily No reuse of the PN solutions
Nutrient Intake Calculation Energy in Kcal per kg day No interruption of the venous line carrying PN
Protein in grams per kg per day
Use of bacterial filter in AA-glucose line

382 Indian Journal of Pediatrics, Volume 75April, 2008


237 145

Parenteral Nutrition

REFERENCES 10. Kanarek K, Santeiro M, Malone J. Continuous infusion of


insulin in hyperglycemic low-birth weight infants receiving
1. te Braake FW, van den Akker CH, Riedijk MA, van parenteral nutrition with and without lipid emulsion. J
Goudoever JB. Parenteral amino acid and energy Parenter Enteral Nutr 1991; 15 : 417-420.
administration to premature infants in early life. Semin Fetal 11. Henry B. Pediatric Parenteral Nutrition Support. In Nevin-
Neonatal Med 2007; 12 : 11-18. Folino N, eds. Pediatric Manual of Clinical Dietetics, Faulhabes;
2. Ziegler EE, Thureen PJ, Carlson SJ. Aggressive nutrition of the 2003; 495-514.
very low birthweight infant. Clin Perinatol 2002; 29 : 225-44. 12. Shulman R. New developments in total parenteral nutrition
3. Hulzebos CV, Sauer PJ. Energy requirements. Semin Fetal for children. Curr Gastroenterol Rep 2000; 2 : 253-258.
Neonatal Med 2007; 12 : 2-10. 13. Poindexter BB, Karn CA, Denne SC. Exogenous insulin
4. Thureen P, Melara D, Fennessey P. Effect of low versus high reduces proteolysis and protein synthesis in extremely low
intravenous amino acid intake on very low birth weight birth weight infants. J Pediatr 1998; 132 : 948-953.
infants in the early neonatal period. Pediatr Res 2003; 53: 24-32. 14. Gutcher GR, Farrell PM. Intravenous infusion of lipid for the
5. Denne S, Karn C, Alrichs J. Proteolysis and phenylanine prevention of essential fatty acid deficiency in premature
hydroxylation in response to parenteral nutrition in extremely infants. Am J Clin Nutr 1991; 54 : 1024-1028.
premature and normal newborns. J Clin Invest 1996; 97: 746- 15. Diamond R. Parenteral nutrition in the critically ill infant and
754. child. In Baker R, Baker S, Davis A, eds. Pediatric Parenteral
6. Heird W. Amino acid and energy needs of pediatric patients Nutrition, New York, NY: Chapman and Hall; 1997; 273-300.
receiving parenteral nutrition. Pediatr Clin North Am 1995; 42: 16. Aba-Sinden A, Bollinger R. Challenges and controversies in
765-789. the nutrition support of the preterm infant. Support Line 2002;
7. Heird W, Discoll J. Total parenteral nutrition. Neo Reviews 2 : 2-15.
2003; 4 : e137-e139. 17. Ziegler E, ODonnell A, Nelson S. Body composition of the
8. Denne S, Poindexter B, Leitch C. Nutrition and metabolism in reference fetus. Growth 1976; 40 : 320-341.
the high-risk neonate. Part 2: Parenteral Nutrition. In Fanaroff 18. Fuhrman M. Management of complications of parenteral
A, Martin R, eds. Neonatal-Perinatal Medicine, St. Louis, MO: nutrition. In Matarese L, Gottschlich M, eds. Contemporary
Mosby; 2002; 598-617. Nutrition Support Practice: A Clinical Guide, Philadelphia, PA:
9. Lifshitz C. Carbohydrate Needs in Preterm and Term Infants, Vol Saunders; 1998; 236-237. 249, 252-253.
122, Philadelphia, PA: Hanley and Belus; 1988.

Indian Journal of Pediatrics, Volume 75April, 2008 383


238
239
240
241
242
243
244
245

You might also like