Anda di halaman 1dari 238

UJIAN TENGAH SEMESTER (UTS)

KEPERAWATAN ANAK KRITIS

Dosen Koordinnator : Ibu Fauziah Rudhiati M.Kep.,Ns.Sp.Kep.An

Disusun Oleh
Nur Hafni Hasim (215121233)

UNIVERSITAS JENDERAL ACHMAD YANI CIMAHI


FAKULTAS ILMU DAN TEKNOLOGI KESEHATAN
PROGRAM STUDI MAGISTER KEPERAWATAN
CIMAHI
2022
1. Anak J, laki-laki berusia 12 bulan yang sebelumnya sehat. Dia memerlukan
perawatan di ruang intensif karena ibu mengeluh bahwa anaknya mengalami
kesulitan bernapas. 3 hari sebelum masuk rumah sakit, ibu mengatakan bahwa
anaknya menderita flu biasa, anak rewel, hidungnya meler, demam dan batuk tetapi
ibu menilai bahwa anak J masih dapat bernapas dengan baik. Semakin hari, batuk
anak J semakin memburuk terutama di malam hari. Ibu menilai anak menjadi
bernapas lebih cepat dari biasanya. Dari hasil pengkajian bahwa HR adalah 190
x/menit, RR 65 x/menit, suhu 39.50C, TD 100/54 mmHg dan saturasi oksigen
85%. Terdapat retraksi intercostal dan penarikan di area trakea saat bernapas,
terdengar suara wheezing dan sumbatan jalan nafas bagian atas. Pemberian Oksigen
sebesar 2L melalui nasal kanul hanya meningkatkan saturasi oksigen menjadi 90%.
Usaha bernafas yang berat terus berlanjut. Dokter menegakkan diagnose medis anak J
RSV (respiratory syncytial Virus) yang menyebabkan bronchiolitis. Anak J
membutuhkan alat bantu pernapasan.
a. Jenis alat bantu pernapasan apa yang bisa diberikan pada anak J adalah
b. Jelaskan indikasi, hal yang harus kita pantau/monitoring, dan komplikasi
selama anak J memakai alat bantu napas tersebut!
c. Jelaskan kriteria keberhasilan dan kegagalan pemakaian alat bantu napas
tersebut!
a. Jenis alat bantu yang digunakan pada An J adalah Ventilator dengan mode SIM V.
dengan melihat kriteria HR adalah 190 x/menit, RR 65 x/menit, suhu 39.5 0C, TD 100/54
mmHg dan saturasi oksigen 85%. Terdapat retraksi intercostal dan penarikan di area
trakea saat bernapas, terdengar suara wheezing dan sumbatan jalan nafas bagian atas.
Pemberian Oksigen sebesar 2L melalui nasal kanul hanya meningkatkan saturasi oksigen
menjadi 90%.
Dari data diatas pasien mengalami distress pernafasan sehingga perlu dilakukan
intubasi untuk pemasangan ventilator mekanik dengan mode SIM V. Mode SIM V
untuk ventilasi pada pasien yang mempunyai beberapa kali nafas spontan tetapi belum
adekuat. Mesin ventilator secara Intermiten (Kadang-kadang) akan memberikan nafas
control ke pasien agar mencapai target minute volume yang telah diatur
b. Indikasi pemasangan ventilator adalah : (William Owens, 2012)

1. Pasien dengan gagal nafas.


Pasien dengan distres pernafasan gagal nafas, henti nafas (apnu) maupun hipoksemia
yang tidak teratasi dengan pemberian oksigen merupakan indikasi ventilasi mekanik.
Idealnya pasien telah mendapat intubasi dan pemasangan ventilasi mekanik sebelum
terjadi gagal nafas yang sebenarnya. Distres pernafasan disebabkan ketidakadekuatan
ventilasi dan atau oksigenasi. Prosesnya dapat berupa kerusakan paru (seperti pada
pneumonia) maupun karena kelemahan otot pernafasan dada (kegagalan memompa
udara karena distrofi otot).
RR >35 atau < 5x/m, SaO2 < 90% atau PaO2 <60 mmHg, pCO2 >55mmHg,
peurunan kesadaran GCS , 5ml/kg
2. Insufisiensi jantung.
Tidak semua pasien dengan ventilasi mekanik memiliki kelainan pernafasan primer.
Pada pasien dengan syok kardiogenik dan CHF, peningkatan kebutuhan aliran darah
pada sistem pernafasan (sebagai akibat peningkatan kerja nafas dan konsumsi
oksigen) dapat mengakibatkan jantung kolaps. Pemberian ventilasi mekanik untuk
mengurangi beban kerja sistem pernafasan sehingga beban kerja jantung juga
berkurang.
3. Disfungsi neurologis
Pasien dengan GCS 8 atau kurang yang beresiko mengalami apnu berulang juga
mendapatkan ventilasi mekanik. Selain itu ventilasi mekanik juga berfungsi untuk
menjaga jalan nafas pasien serta memungkinkan pemberian hiperventilasi pada klien
dengan peningkatan tekanan intra cranial.
4. Tindakan operasi
Tindakan operasi yang membutuhkan penggunaan anestesi dan sedative sangat
terbantu dengan keberadaan alat ini. Resiko terjadinya gagal napas selama operasi
akibat pengaruh obat sedative sudah bisa tertangani dengan keberadaan ventilasi
mekanik.
5. Membantu eliminasi carbondioksida
6. Membantu kerja otot pernapasan
Hal yang perlu dipantau yang saat pasien yang di pasang ventilator adalah : (Andre C.
Kalil, 2022)
a. Penilaian ventilasi dan oksigenasi
b. Alarm (pplateu<30 cmH2O). RR < 6x/mnt - > 30x/mnt, MV , 25ml/kg –
125ml/kg, PEEP , 30 cmH2O
c. Klinis : Inspeksi, Palpasi, Perkusi, Auskultasi
 Anallis gas Darah : pCO2, pO2, SaO2
 Fhoto thoraks : posisi ujung ETT, patu pleura
d. Monitor RR, SPO2
e. Ventilator : Tidal Volume respiratory Rat, p Inspriasi, grafik Flow,pressure
dan Volume

Komplikasi pemasangan ventilasi mekanik menurut Bersten dan Soni (2009):


1. Komplikasi akibat peralatan. Terkait malfungsi atau pemutusan alat,
kesalahan tempat dan kontaminasi .
2. Komplikasi terkait dengan paru-paru, seperti intubasi Airway misalnya
kerusakan gigi, pita suara dan trakea, VentilatorAcquired Pneumonia (VAP),
gangguan terkait cedera paru-paru misalnya difusi cedera paru-paru,
barotrauma misalnya pneumothorax dan keracunan O2.
3. Komplikasi yang terkait dengan kardiovaskuler, seperti penurunan preload
ventrikel kanan yang menyebabkan penurunan curah jantung, peningkatan
afterload ventrikel kanan, retensi cairan karena penurunan jantung yang
mengakibatkan penurunan aliran darah di ginjal.
4. Komplikasi lainnya seperti : luka atau perdarahan pada jaringan mukosa,
kelemahan oto-otot pernapasan dan peripheral, gangguan tidur, kecemasan,
ketakutan akibat lamanya waktu setelah masa penyembuhan, distensi akibat
menelan, imobilisasai dan masalah pencernaan
Kriteria keberhasilan pemasangan Ventilator :
1. Pengkajian subjektif :Batuk adekuat, Tidak menggunakan agent
neuromuscular blocking, Tidak ada produksi mucus yang berlebih pada
trakheobronkhial, Core problem pada pasien sudah teratasi dan Tidak
mendapatkan sedasi yang berkelanjutan
2. Pengukuran objektif : Status kardiovaskuler stabil, HR < 140 x/menit, Tidak
ada iskemik miokard , Tidak anemia (Hb > 8 g/dl), Tekanan darah sistolik 90
– 160 mmHg, GCS > 8, Tidak demam (rentang 36 < suhu < 38℃) 8)
Penggunaan vasopressor dan inotropik pada dosis minimal ( < 5
ug/kgBB/menit untuk dopamin atau dobutamin)
3. Parameter oksigenasi yang adekuat : Nilai tidal volume > 5 cc/kgBB, Nilai
vital capacity > 10 cc/kgB, RR < 35 x/menit 4) SpO2 > 95 % 5) PaO2 > 60
mmHg, dan nilai PCO2 < 60 mmHg

2. Berdasarkan data Kemenkes (2021), 84 % kematian bayi di Indonesia akibat


kelahiran perematur. Bayi prematur memiliki keterbatasan dalam penyimpanan
nutrisi sehingga berpotensi mengalami defisit nutrisi. Sistem gastrointestinal yang
belum berkembang juga berpotensi meningkatkan risiko infeksi pada bayi tersebut.
Silahkan anda analisis bagaimana optimalisasi pemberian nutrisi pada bayi
prematur, agar terhindar dari masalah sistemik. Analisis secara teori dan hasil
penelitian (min.3 penelitian dan lampirkan artikel ilmiah tersebut).

Bayi prematur rentan terhadap penyakit serius atau kematian selama periode
neonatal. Tanpa pengobatan yang tepat, mereka yang dapat bertahan hidup berada
pada situasi peningkatan risiko cacat seumur hidup dan memiliki kualitas hidup yang
buruk.
Asupan nutrisi yang kurang sejak sebelum dan pada masa kehamilan
merupakan pemicu kelahiran bayi dengan BBLR. Kegagalan pertumbuhan bayi pasca
persalinan akibat nutrisi yang tidak memadai akan berhubungan dengan
perkembangan neurokognitif bayi prematur dimasa mendatang. Oleh karena itu,
strategi pemberian nutrisi yang cukup untuk menjamin tumbuh kembang yang
optimal pada bayi prematur perlu ditekankan. Rekomendasi terapi nutrisi bayi
prematur saat ini didasarkan pada tujuan mencapai tingkat pertumbuhan. Namun,
tujuan ini seringkali sulit dicapai karena keterbatasan fisiologis yang terkait dengan
prematuritas.

1. Menurut The American Academy of Pediatrics (AAP) dan European Society of


Pediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) Committee on
Nutrition, asupan energi yang harus diberikan pada bayi prematur adalah 105-130
kkal/kg/hari dan 110-135 kkal/kg/hari). Saat terapi ini berlangsung, bayi terkadang
membutuhkan peningkatan asupan kalori karena kebutuhan energi yang lebih tinggi
selama sakit (ESPGHAN Committee on Nutrition et al., 2013).
Saat bayi prematur, bukan saja kebutuhan energi yang diutamakan, namun
dikombinasikan dengan kebutuhan protein. Kebutuhan protein yang
direkomendasikan ESPGHAN adalah 4,0-4,5 dan 3,5-4,0 g/kg/hari pada bayi
prematur dengan berat masing-masing hingga 1.000 g dan 1.000-1.800 g (Wiechers et
al., 2021).
Menurut hasil penelitian yang berjudul Optimizing Nutrition in Preterm Low
Birth Weight Infants—Consensus Summary. Frontiers in Nutrition. (Kumar et al.,
2017). Mendapatkan hasil Asupan protein melalui makanan enteral adalah 150
mL/kg/hari (volume enteral rutin) tidak cukup untuk memenuhi kebutuhan bayi yaitu
1,8-2,1 g/kg/hari untuk ASI tanpa fortifikasi, 1,5 g/kg/hari donor ASI dari manusia
yang tidak diperkaya susu, atau 3,6 g/kg/hari formula khusus untuk bayi prematur.
Penelitian mengatakan Untuk menyediakan 4 g/kg/hari protein, kita perlu
memberi makan bayi 290–340 mL/kg/hari ASI tanpa fortifikasi, 210 mL/kg/hari ASI
yang difortifikasi atau 170 (16% konsentrasi) hingga 190 mL/kg/hari (14%
konsentrasi) formula bayi prematur Penting untuk dicatat bahwa osmolaritas dan
beban zat terlarut ginjal mempengaruhi volume cairan yang diperlukan untuk nutrisi
enteral. Oleh karena itu, fortifikasi untuk bayi yang diberi ASI dan penyesuaian
konsentrasi susu formula untuk bayi prematur yang diberi susu formula diperlukan.
Untuk pemberian makanan enteral rutin, formula prematur atau ASI yang diberikan
dengan kecepatan 150-180 mL/kg/hari sudah cukup untuk memenuhi kebutuhan
nutrisi yang direkomendasikan
Osmolaritas dan beban zat terlarut ginjal mempengaruhi volume cairan yang
diperlukan untuk nutrisi enteral. Oleh karena itu, fortifikasi untuk bayi yang diberi
ASI dan penyesuaian konsentrasi susu formula untuk bayi prematur yang diberi susu
formula diperlukan. Untuk pemberian makanan enteral rutin, formula prematur atau
ASI yang diberikan dengan kecepatan 150-180 mL/kg/hari sudah cukup untuk
memenuhi kebutuhan nutrisi yang direkomendasikan

2. Terdapat beberapa hal yang perlu diperhatikan dalam pemberian protein


adalah setiap penambahan gram asupan protein untuk bayi prematur disesuaikan
dengan penambahan berat badan 6,5 g/hari dan peningkatan lingkar kepala 0,4
cm/minggu). Saat memantau penambahan berat badan bayi prematur saat perawatan
di rumah sakit, hal-hal kritis yang harus diperhatikan adalah seperti bayi di kuartil
terendah dari kenaikan berat badan di rumah sakit yaitu ≤12 g/kg/hari kemungkinan
menderita cerebral palsy selain itu dikaitkan dengan hasil pemeriksaan
perkembangan menggunakan skala Bayley dan menemukan hasil <70, serta gangguan
perkembangan saraf pada 18-22 bulan
Hal tersebut sesuai dengan judul penelitian yang berjudul Optimizing nutrition of the
preterm infant. Chinese Journal of Contemporary Pediatrics. (William, 2017)
Hasil dari penelitian mengatakan hal yang diharapkan dari terapi nutrisi ini
adalah perkembangan saraf dan pertumbuhan yang lebih baik pada bayi prematur,
penambahan berat badan >18 g/kg/hari dan peningkatan lingkar kepala >0,9
cm/minggu selama masa rawat inap dianggap optimal. Jika tingkat ini tidak dapat
dipenuhi, nutrisi enteral bayi harus ditinjau dan dimodifikasi untuk mencapai tujuan
optimal ini. Tindakan dapat difokuskan pada kandungan protein pada makanan yang
diberikan, evaluasi rasio Protein dan energi serta pemanfaatan sumber makanan yang
kaya nutrisi untuk mengurangi defisit nutrisi.
asupan protein yang memadai dapat meningkatkan pertumbuhan bayi
prematur. Jika Tindakan ini dilakukan dengan baik di NICU, maka resiko kecacatan
saraf akan jauh lebih rendah. Perawat wajib untuk memastikan rasio protein/energi
untuk mencapai pertumbuhan yang berkualitas selama berada di NICU. Tanda-tanda
pertumbuhan yang baik selama dirawat di NICU adalah peningkatan massa tubuh
tanpa lemak dan timbunan massa lemak yang terbatas. Jika hasil penilaian tumbuh
kembang menunjukan hasil yang optimal maka asupan protein dapat dikurangi
sebagai proses untuk pulang dari rumah sakit.

Implementasi pemberian nutrisi enteral pada bayi prematur

a. Pemberian makanan enteral minimal


Sebaiknya pemberian makanan enteral ditunda dalam beberapa waktu, karena
beresiko terjadinya enterokolitis nekrotikans (NEC) walaupun pada dasarnya
secara teori pemberian makanan enteral dengan volume rendah (biasanya 24
mL/kg/hari) dapat mempercepat maturitas gastrointestinal, fisiologis, endokrin,
dan metabolic. Selain itu, makanan tersebut tidak mengandung kalori yang
cukup untuk mempertahankan pertumbuhan.
Menurut hasil penelitian yang berjudul Early trophic feeding versus enteral
fasting for very preterm or very low birth weight infants. (Morgan et al., 2013)
Penelitian mengevaluasi efek pemberian makanan trofik dini versus puasa
enteral pada bayi berat lahir sangat rendah (BBLSR) dan tidak menemukan
perbedaan dalam toleransi makan, waktu untuk mencapai pemberian makanan
enteral penuh, dan kejadian NEC). Dengan demikian, dapat disimpulkan bahwa
nutrisi enteral dengan volume yang minimal tidak berkaitan dengan efek
menguntungkan atau berbahaya pada bayi prematur sehingga makanan trofik
dapat dimulai sesegera mungkin.

b. Pengenalan makanan enteral progresif


Pengenalan makanan enteral untuk bayi prematur sering tertunda selama
beberapa hari karena kekhawatiran dapat meningkatkan risiko NEC.
Hal tersebut berbeda dengan penelitian yang berjudul Delayed introduction of
progressive enteral feeds to prevent necrotising enterocolitis in very low birth
weight infants. (Morgan et al., 2014b)
Penelitian mengevaluasi efek pengenalan pemberian makan dini (<4 hari)
dengan pengenalan pemberian makan yang tertunda (4-7 hari) hasil penelitian
tidak menemukan perbedaan yang signifikan dalam hal kejadian NEC
atau penyebab kematian. Namun, bayi dengan pengenalan makanan yang
tertunda membutuhkan waktu yang lebih lama untuk beradaptasi hingga dapat
mengkonsumsi pemberian makanan enteral penuh.

c. Tingkat kemajuan pemberian makanan enteral


Salah satu pendekatan yang dapat dilakukan adalah pemberian makanan
enteral dini sebanyak 20-30 mL/kg/hari pada VLBWI dan 15-25 mL/kg/hari
pada bayi prematur karena dalam beberapa penelitian, tidak terdapat risiko NEC.
Penelitian oleh (Morgan et al., 2014a) mengevaluasi efek dari tingkat kemajuan
pemberian makanan enteral pada kejadian NEC, semua penyebab kematian, dan
morbiditas lain pada bayi prematur dan tidak menemukan perbedaan yang
signifikan. Pada kelompok yang lambat memberikan makanan enteral sejak dini
menyebabkan perkembangan lambat sehingga bayi membutuhkan waktu yang
lebih lama untuk mecapai berat badan lahir yang ideal serta lambat untuk
beradaptasi dengan makanan enteral yang diberikan sesuai panduan.

Pilihan nutrisi enteral


1. Air Susu Ibu
ASI adalah sumber nutrisi utama yang optimal untuk bayi prematur karena
memberikan perlindungan yang kuat terhadap sepsis dan infeksi lainnya, mencegah
terjadinya NEC. Efek jangka Panjang dari pemberian ASI adalah perkembangan saraf
yang baik. Oleh karena itu, AAP sangat merekomendasikan penggunaan ASI untuk
bayi prematur. Tetapi kandungan ASI tidak bisa memenuhi kebutuhan nutrisi bayi
prematur.
Hal tersebut sesuai dengan Peneltian yang berjudul Enteral nutrition for optimal
growth in preterm infants. (Kim, 2016). Strategi laktasi yang dapat dilakukan adalah
pemompaan ASI sesegera mungkin pasca persalinan. jika ibu tidak memiliki ASI
maka harus dilakukan donor asi namun harus melalui proses pasteurisasi. Kelemahan
dari ASI untuk bayi prematur adalah tidak dapat memenuhi kebutuhan nutrisi bayi
prematur, karena kalori dan protein yang tidak mencukupi, konsentrasi kalsium dan
fosfor dalam ASI juga jauh lebih rendah dari yang diperlukan untuk mendukung
pertumbuhan yang optimal
Terdapat produk dari korea yaitu suplementasi (fortifikasi) dengan multinutrient
human milk fortifiers (HMFs) yang diindikasikan pada bayi prematur berusia kurang
dari 31 minggu kehamilan dan/atau dengan berat lahir kurang dari 1.500g.
Umumnya, fortifikasi dimulai ketika volume makanan enteral mencapai 100
mL/kg/hari dan secara bertahap ditingkatkan. HMF yang dijual
mengandung kandungan protein rata-rata ASI 1,4-1,6 g/dL.
Menurut penelitian yang berjudul Novel multinutrient human milk-based human
milk fortifier promotes growth and tolerance in premature infants. (Gates et al., 2021;
Koo & Tice, 2018). Untuk pemberian suplementasi (fortifikasi) dengan multinutrient
human milk fortifiers (HMFs) harus dilakukan pemeriksaan BUN (blood Ureum
Nitrogen) secara berkala
Teori mengataakan Sebagian besar bayi prematur biasanya dipulangkan dari
NICU pada usia 35 hingga 36 minggu, dan/atau setelah berat badan mencapai 1.800-
2.100gr
Penelitian yang berjudul rowth and Bone Mineralization of Very Preterm Infants
at Term Corrected Age in Relation to Different Nutritional Intakes in the Early
Postnatal Period. (Körnmann et al., 2017). Mendapatkan hasil ibu yang berencana
untuk menyusui bayinya secara langsung setelah keluar, laju pertumbuhan harus
dipantau dengan cermat dalam waktu 48 jam setelah keluar dan dievaluasi kembali
setelah 1 minggu. Bagi bayi yang dipulangkan saat masih menerima ASI perah atau
ASI donor, asi fortifikasi adalah pilihan yang lebih praktis dan berkelanjutan
2. Susu Formula untuk bayi prematur
Susu Formula untuk bayi prematur diberikan jika bayi tidak dapat menerima ASI
dari ibunya sendiri maupun ASI donor.
Menurut penelitian Susu formula harus mengandung protein whey, polimer glukosa,
trigliserida rantai menengah, kalsium, dan fosfor, dan diperkaya dengan mineral,
vitamin, dan elemen untuk mendukung tingkat pertambahan nutrisi intrauterin. Dalam
praktiknya, dosis inisiasi pemberian susu formula pada bayi prematur adalah 70-72
kkal/100 mL. Jika ditoleransi oleh bayi, maka dapat ditingkatkan menjadi formula
preterm pekat 16% (80-82 kkal/100 mL).
Susu formula untuk bayi prematur umumnya tidak direkomendasikan untuk bayi yang
telah diijinkan pulang, karena sasaran utama telah tercapai. Sehingga susu formula
bayi prematur hanya setelah bayi keluar dari NICU 2. susu formula saat pulang
Saat pulang, bayi prematur yang telah mencapai berat badan optimal dapat
mengkonsumsi susu formula yang mengandung energi pada 75 kkal/100 mL, kaya
protein 2,1 g dan mengandung mineral, vitamin, dan elemen – elemen lain (Aggett et
al., 2006)

3. Jelaskan peran perawat intensif anak pada kasus anak post operasi VSD usia 4
tahun dan identifikasi kebutuhan perawatan perkembangan serta FCC nya!!
Peran perawat Intensive secara Umum
a. Seorang perawat yang bertugas di ruang intensif melaksanakan tiga tugas utama
yaitu, life support, memonitor keadaan pasien dan perubahan keadaan akibat
pengobatan dan mencegah komplikasi yang mungkin terjadi.
b. Di Australia diklasifikasikan empat kriteria perawat intensif yaitu, perawat Intensif
yang telah mendapat pelatihan lebih dari duabelas bulan ditambah dengan
pengalaman, perawat yang telah mendapat latihan sampai duabelas bulan, perawat
yang telah mendapat sertifikat pengobatan kritis (critical care certificate),
dan perawat sebagai pelatih (trainer).
c. Di Indonesia, ketenagaan perawat di ruang intensif diatur dalam Keputusan
Menteri Kesehatan Republik Indonesia Nomor
1778/MENKES/SK/XII/2010 tentang Pedoman Penyelenggaraan Pelayanan Intensif
di Rumah Sakit yaitu, untuk ICU level I maka perawatnya adalah
perawat terlatih yang bersertifikat bantuan hidup dasar dan bantuan lanjut, untuk ICU
level II diperlukan minimal 50% dari jumlah seluruh perawat di ICU merupakan
perawat terlatih dan bersertifikat ICU, dan untuk ICU level III diperlukan minimal
75% dari jumlah seluruh perawat di ICU merupakan perawat terlatih dan bersertifikat
ICU
3. Jelaskan peran perawat intensif anak pada kasus anak post operasi VSD usia 4
tahun dan identifikasi kebutuhan perawatan perkembangan serta FCC nya!!

A. Peran perawat Intensive.


1. Seorang perawat yang bertugas di ruang intensif melaksanakan tiga tugas utama
yaitu, life support, memonitor keadaan pasien dan perubahan keadaan
akibat pengobatan dan mencegah komplikasi yang mungkin terjadi.
2. Di Australia diklasifikasikan empat kriteria perawat intensif yaitu, perawat
Intensif yang telah mendapat pelatihan lebih dari duabelas bulan ditambah
dengan pengalaman, perawat yang telah mendapat latihan sampai duabelas bulan,
perawat yang telah mendapat sertifikat pengobatan kritis (critical care
certificate), dan perawat sebagai pelatih (trainer).
3. Di Indonesia, ketenagaan perawat di ruang intensif diatur dalam
Keputusan Menteri Kesehatan Republik Indonesia Nomor 1778/ME
NKES/SK/XII/2010 tentang Pedoman Penyelenggaraan Pelayanan
Intensif di Rumah Sakit yaitu, untuk ICU level I maka perawatnya
adalah perawat terlatih yang bersertifikat bantuan hidup dasar dan bantuan lanjut,
untuk ICU level II diperlukan minimal 50% dari jumlah seluruh perawat di ICU
merupakan perawat terlatih dan bersertifikat ICU, dan untuk ICU level III
diperlukan minimal 75% dari jumlah seluruh perawat di ICU merupakan perawat
terlatih dan bersertifikat ICU

B. Peran perawat Anak.


Perawat merupakan anggota dari tim pemberi asuhan keperawatan anak dan
orang tuanya. Perawat dapat berperan dalam berbagai aspek dalam memberikan
pelayanan kesehatan dan bekerjasama dengan anggota tim lain, dengan keluarga
terutama dalam membantu memecahkan masalah yang berkaitan dengan
perawatan anak.
Peran perawat dalam merawat pasien post Op VSD yang berusia 4 tahun adalah
sebagai berikut :
1) Sebagai pendidik.
Perawat berperan sebagai pendidik, baik secara langsung dengan memberi
penyuluhan/pendidikan kesehatan pada orang tua maupun secara tidak langsung
dengan menolong orang tua/anak memahami pengobatan dan perawatan anaknya.
Kebutuhan orang tua terhadap pendidikan kesehatan dapat mencakup pengertian
dasar penyakit anaknya, perawatan anak selama dirawat di rumah sakit, serta
perawatan lanjut untuk persiapan pulang ke rumah. Tiga domain yang dapat
dirubah oleh perawat melalui pendidikan kesehatan adalah pengetahuan,
keterampilan serta sikap keluarga dalam hal kesehatan khususnya perawatan anak
sakit.
2) Sebagai konselor
Suatu waktu anak dan keluarganya mempunyai kebutuhan psikologis berupa
dukungan/dorongan mental. Sebagai konselor, perawat dapat memberikan
konseling keperawatan ketika anak dan keluarganya membutuhkan. Hal inilah
yang membedakan layanan konseling dengan pendidikan kesehatan. Dengan cara
mendengarkan segala keluhan, melakukan sentuhan dan hadir secara fisik maka
perawat dapat saling bertukar pikiran dan pendapat dengan orang tua tentang
masalah anak dan keluarganya dan membantu mencarikan alternatif
pemecahannya.
3) Melakukan koordinasi atau kolaborasi
a. Pasien post op VSD akan mendapat obat anastesi yang mengakibatkan
kelemahan otot-otot pernafasan dan memasifkan silia. Sehingga banyak
akumulasi sekret yang berada di saluran pernafasan. Sehingga hal ini
perawat sebagai salah satu tenaga kesehatan yang berada 24 jam di
samping pasien perlu melakukan kolaborasi dengan fisioterapi untuk
dillakuakn fisioterapi untuk Pencegahan akumulasi sekret, karena pasien
dengan post op VSD aktivitasnya dibatasi. Kemudian pasien.
b. Kolaborasi dan Koordinasi dengan adalam ahli gizi terkait pemberian
Nutrisi pada anak Post Op VSD terkait diet tinggi protein dan rendah
natrium .
c. Melakukan koordinasi atau kolabosi dengan dokter dalam pemantuaun
efek sedasi dan analgesik pasien setelah post Op dan selalu melaporkan
kondisi pasien. Dengan pendekatan interdisiplin, perawat melakukan
koordinasi dan kolaborasi dengan anggota tim kesehatan lain dengan
tujuan terlaksananya asuhan yang holistik dan komprehensif. Perawat
berada pada posisi kunci untuk menjadi koordinator pelayanan kesehatan
karena 24 jam berada di samping pasien.

C. Sebagai advokat pasien,


Dalam perawatan anak dengan post VSD, perawat membantu proses
komunikasi dan permberian informasi yang layak antara pasien , keluarga dan
tenaga kesehatanlainnya dan perawat membantu membantu pasien dan keluarga
mendapatkan hak-haknya. Contohnya perawat menajadi perantara jika kelurga
ingin menanyakan tentang keadaan pasien post Op VSD kepada dokter hal tersebut
juga termasuk dalam hak pasien. Contoh pemenuhan hak yang lain adalah pasien
berhak mendampingi pasien, mendapatkan informasi terkait perawaan anaknya,
serta juga membantu dalam pasien dan keluraga untuk melakuak pemenuhan
spritual ibadah sesuai dengan agama / kepercayaan yang dianutnya selama hal itu
tidak mengganggu pasien lain. Dalam hal perawat dapat menerpakan family
Centered care.
D. Sebagai pemberi asuhan keperawatan,
Dimana perawat dalam hal ini berperan dalam menyesuaikan pemberian
asuhan keperawatan pada pasien dengan post OP VSD tetap memperhatikan
kebutuhan dasar manusia.  merawat membantu pasien VSD untuk pemenuhan
Kebutuhan Fisik-Biomedik (asuh). hiygine, nutrisi, pemenuhan cinta kasih dan
kenyamanan, untuk pemenuhan cinta da kenyaman, pasien bisa melibatkan
keluarga untuk pemenuhannya dengan menganjurakn orang tua untuk selalu berada
di samping anak, karena anak akan merasakan nyaman jika berada dekat dengan
kelurganya, hal tersebut juga dapat mencegah Autraumatik care pada anak.
Bermain merupakan salah satu kebutuhan anak, peran perawat dalam hal ini bisa
membantu pasien bermain di rumah sakit dengam tetap memperhatikan prinsip
bermain pada anak post op, bermain yang bisa dilakukan adalah dengan story
telling, mendengarkan musik, untuk story telling pasien bisa meminta orang tua
terlibat di dalam hal tersebut.
Salah satu penelitian terkait peran perawat pada pasien post op VSD. Hal ini sesuai
dengan penelitian A nurse‐driven analgesia and sedation protocol reduces length of
PICU stay and cumulative dose of benzodiazepines after corrective surgery for tetralogy
of Fallot.(Hanser et al., 2020). Hasil dalam penelitian ini adalah

Framework Data
item
Patient 1. Jenis pasien yang menjadi responden penelitian ini adalah pasien anak
Problem, (or yang mengalami TOF jenis VSD pasca operasi yang sedang dirawat di
Population PICU
2. Jumlah sampel yang menjadi responden dalam penelitian ini adalah 65
responden yang dibagi menjadi 2 kelompok yaitu kelompok pra-
implementasi (33) dan kelompok post-implementasi (32).
3. Kriteria inklusi dalam penelitian ini adalah pasien TOF VSD usia 2-6
bulan.
4. Kriteria eksklusi dalam penelitian ini adalah pasienyang memiliki
penyakit komorbid seperti kelainan kromosom (misalnya, sindrom
mikrodelesi 22q11, Trisomi 21) serta anomali ekstrakardiak yang
memerlukan perawatan khusus seperti atresia esofagus dan atresia anal.
Intervention, 1. Penelitian ini dilaksanakan dalam 2 tahap yaitu Pre-implementasi (saat
operasi) dan post implementasi (setelah operasi)
2. Pemberian obat pada fase Pre-implementasi (saat operasi) dan post
implementasi (setelah operasi) dilakukan oleh dokter. Namun, pada fase
pre-implementasi tidak dilakukan pengawasan oleh perawat karena
dilakukan di kamar operasi. Perawat hanya mengobservasi gejala klinis
pasien di fase post-implementasi
3. Obat yang diberikan dalam penelitan ini adalah;
a. Kelompok pra-implementasi
1) i.v. infus opioid (morfin: 5–100 µg·kg−1·hr−1 dengan dosis
awal: 30 g·kg−1/hr−1) dan
2) i.v. infus benzodiazepin (midazolam: 0,05-0,42
mg·kg−1·hr−1; dosis awal: 0,05 mg·kg−1·hr−1).
3) Sevofluran
b. Kelompok post-implementasi
1) i.v. infus opioid (morfin: 5–100 µg·kg−1·hr−1 dengan dosis
awal: 30 g·kg−1/hr−1) dan
2) i.v. infus benzodiazepin (midazolam: 0,05-0,42
mg·kg−1·hr−1; dosis awal: 0,05 mg·kg−1·hr−1).
3) Clonidine diberikan 24 jam setelah operasi dengan
mengurangi dosis opioid dan sedasi. Clonidine di berikan via
intravena/infus dengan dosis : 0,2–2 g·kg−1·hr−1. dosis
awal: 0,04 g·kg−1/jam−1).
4) Melatonin oral (3–5 mg/hari) diberikan setelah 3 hari tinggal
di PICU.
5) Khloral hidrat (sampai 25 mg/kg empat kali sehari)
diberikan tambahan setelah hari kedua pasca operasi
4. Alat ukur yang digunakan adalah COMFORT B untuk mengetahui efek
sedasi dari obat yang diberikan
5. Penggunaan skala NISS untuk mengetahui efek analgesic.
6. Evaluasi dilakukan setiap 8 jam.
Comparison or 1. Penelitian ini dilakukan pada 2 kelompok yaitu kelompok pre-
Control, implementasi (saat operasi) dan kelompok post-implmentasi (setelah
operasi
2. Perbandingan pemberian obat pada kedua kelompok adalah sebagi
berikut;
a. Kelompok pra-implementasi
1) i.v. infus opioid (morfin: 5–100 µg·kg−1·hr−1 dengan dosis
awal: 30 g·kg−1/hr−1) dan
2) i.v. infus benzodiazepin (midazolam: 0,05-0,42
mg·kg−1·hr−1; dosis awal: 0,05 mg·kg−1·hr−1).
3) Sevofluran
b. Kelompok post-implementasi
1) i.v. infus opioid (morfin: 5–100 µg·kg−1·hr−1 dengan dosis
awal: 30 g·kg−1/hr−1) dan
2) i.v. infus benzodiazepin (midazolam: 0,05-0,42
mg·kg−1·hr−1; dosis awal: 0,05 mg·kg−1·hr−1).
3) Clonidine diberikan 24 jam setelah operasi/berada di PICU
dengan mengurangi dosis opioid dan sedasi. Clonidine di
berikan via intravena/infus dengan dosis : 0,2–2
g·kg−1·hr−1. dosis awal: 0,04 g·kg−1/jam−1).
4) Melatonin oral (3–5 mg/hari) diberikan setelah 3 hari tinggal
di PICU.
5) Khloral hidrat (sampai 25 mg/kg empat kali sehari)
diberikan tambahan setelah hari kedua pasca operasi
Outcome 1. Hasil yang didapatkan dalam penelitian ini adalah
a. Penerapan protokol sedasi yang dipantau oleh perawat dapat
membantu mengurangi dosis opioid dan benzodiazepin, serta
lama rawat inap di PICU.
b. Setelah penerapan protokol analgesia dan sedasi, waktu rata-rata
penggunaan ventilator menjadi lebih pendek (72 [24-141] jam
menjadi 49 jam [24-98]
c. Lama perawatan di PICU berkurang secara signifikan (7 hari
[5–14] menjadi 5 hari [4–7]
d. pasien dari kedua kelompok mendapat dosis kumulatif morfin
yang sama namun mengalami penurunan (1.961 g/kg [1.113–
3.162] menjadi 1.920 g/kg [904–2.687]).
e. Terjadi peningkatan pemberian clonidine setelah 24 jam pada
pasien pasca operasi yaitu (0,53 mg/kg [0,24–0,92] menjadi
6,05 mg/kg [0,81–22,53] disertai dengan penurunan pemberian
dosis kumulatif benzodiazepin secara signifikan yaitu (7,37
mg/kg [4,70-17,65] Menjadi 5,0 mg/kg [2,70–9,12]
f. Selain itu, dosis morfin dikurangi pasca pemberian clonidine
yaitu (50,0 g·kg−1·jam−1 [39,7–79,9] menjadi. 42,5
g·kg−1·jam−1 [29,7–51,8]) dan midazolam (0,22
mg·kg−1·jam−1 [0,20–0,33] menjadi 0,15 mg·kg−1·jam−1
[0,13–0,20]).
g. Penggunaan klonidin meningkat secara signifikan (0,014
g·kg−1·jam−1 [0,009–0,021] menjadi 0,674 g·kg−1·jam−1
[0.02–1.095]
h. Kelompok pasca implementasi tidak menunjukkan peningkatan
komplikasi pasca operasi dan kejadian dan adverse event.
Time/Type of 1. Tujuan dari penelitian ini adalah untuk mengevaluasi efek dari
Study or implementasi protokol sedasi yang dilakukan oleh perawat dengan
question melihat pada durasi penggunaan ventilasi mekanis, lama perawatan,
dosis puncak dan komulatif dan efek samping dibandingkan dengan
periode praimplementasi yang dilakukan oleh dokter
2. Penelitian ini merupakan penelitian retrospektif dengan studi
observasional
3. Penelitian ini dilakukan dengan 2 tahap yaitu kelompok pra-
implementasi pada Januari 2005 dan Oktober 2010 sebanyak 33
responden dan kelompok post-implementasi pada periode Januari 2012
hingga Juli 2017 yaitu sebanyak 32 responden

E. Sebagai peneliti/pembaharu.
Melakukan penelitiain tentang penanganaan pasien post VSD untuk meningkatkan
kualitas praktik keperawatan anak.
Perawatan dengan memperhatikan perkembangan anak. Anak usia 4 tahun termasuk
dalam anak usia pra sekolah. Anak usia pra sekolah kurang dapat membedakan
antara diri sendiri dan orang lain. Mereka memiliki pemahaman bahasa yang terbatas
dan hanya dapat melihat satu aspek dari suatu objek atau situasi pada satu waktu.
a. Anak usia pra sekolah merasa fenomena nyata yang tidak berhubungan sebagai
penyebab penyakit.
b. Cara berpikir magis menyebabkan anak usia pra sekolah memandang penyakit
sebagai suatu hukuman. Selain itu anak usia pra sekolah mengalami konflik
psikososial dan takut terhadap mutilasi, menyebabkan anak terutama takut
terhadap tindakan tindakan medis
Menurut erikon
Tahap inisiatif vs rasa bersalah (4-6 tahun/pra sekolah) Pada tahap ini anak
mulai berinisiatif dalam belajar mencari pengalaman baru secara aktif melalui
aktivitasnya. Apabila anak dilarang atau dicegah maka akan tumbuh perasaan
bersalah pada dirinya. Anak post VSD akan merasa ingin tau tentang perawatann
yang dilakukannya. Perawat bisa melalukan perannya sebagai pendidik. Anak
usia 4 tahun telah paham dengan pembicaraan orang dewasa, dengan
memberikan pemahaman tentang perawat yang dilakukan yang dilakuan
menjelaskan tentang manfaat alat medis yang digunakan. Perawat bisa
menerapkan terapi bermain dramatic play untuk menjelaskan tetntang alat alat
kesehatan pada anak sehingga mengurangi kecemasan dan autraumatic care
Perkembangan Psikoseksual Menurut Sigmud Freud
Tahap oedipal/phalik (3-5 tahun). Pada tahap ini kepuasan anak terletak pada
rangsangan autoerotic yaitu merabaraba, merasakan kenikmatan dari beberapa
daerah erogennya dan mulai suka pada lawan jenis. Anak laki-laki cenderung
suka pada ibunya daripada ayahnya demikian juga sebaliknya anak perempuan
suka sama ayahnya. Pada kasus pasien post VSD diatas pada tahap ini perawat
Ibu menerapkan family centered dengan melibatkan Ibu pasien untuk memenuhi
kebutuhan psikoseksual
Perkembangan Kognitif Menurut Piaget
Tahap pra operasional (2-7 tahun) Anak mampu mengoperasionalisasikan apa
yang dipikirkan melalui tindakan sesuai dengan pikirannya. Pada saat ini anak
masih bersifat egosentris. Pikirannya masih transduktif, artinya menganggap
semua sama. Pada tahap ini perawat bisa berperan sebagai pendidik ataupun
sebagai konselor untuk memenuhi perkembangam kognitif. (Yuliastati, 2016)
DAFTAR PUSTAKA

P. J., Agostoni, C., Axelsson, I., De Curtis, M., Goulet, O., Hernell, O., Koletzko, B., Lafeber, H.
N., Michaelsen, K. F., Puntis, J. W. L., Rigo, J., Shamir, R., Szajewska, H., Turck, D.,
Weaver, L. T., & Nutrition, E. C. on. (2006). Feeding Preterm Infants After Hospital
Discharge: A Commentary by the ESPGHAN Committee on Nutrition. Journal of
Pediatric Gastroenterology and Nutrition, 42(5), 596–603.
https://doi.org/10.1097/01.mpg.0000221915.73264.c7
Cooke, R. J. (2016). Post-discharge Nutrition in Preterm Infants. In G. Buonocore, R. Bracci, &
M. Weindling (Eds.), Neonatology: A Practical Approach to Neonatal Diseases (pp. 1–
18). Springer International Publishing. https://doi.org/10.1007/978-3-319-18159-2_189-1
ESPGHAN Committee on Nutrition, Arslanoglu, S., Corpeleijn, W., Moro, G., Braegger, C.,
Campoy, C., Colomb, V., Decsi, T., Domellöf, M., Fewtrell, M., Hojsak, I., Mihatsch,
W., Mølgaard, C., Shamir, R., Turck, D., & van Goudoever, J. (2013). Donor human milk
for preterm infants: Current evidence and research directions. Journal of Pediatric
Gastroenterology and Nutrition, 57(4), 535–542.
https://doi.org/10.1097/MPG.0b013e3182a3af0a
Gates, A., Thompson, A. B., Marin, T., Waller, J. L., Patel, J., & Stansfield, B. K. (2021). Novel
multinutrient human milk-based human milk fortifier promotes growth and tolerance in
premature infants. JPEN. Journal of Parenteral and Enteral Nutrition.
https://doi.org/10.1002/jpen.2249
Hanser, A., Neunhoeffer, F., Hayer, T., Hofbeck, M., Schlensak, C., Mustafi, M., Kumpf, M., &
Michel, J. (2020). A nurse-driven analgesia and sedation protocol reduces length of PICU
stay and cumulative dose of benzodiazepines after corrective surgery for tetralogy of
Fallot. Journal for Specialists in Pediatric Nursing: JSPN, 25(3), e12291.
https://doi.org/10.1111/jspn.12291
Kim, M.-J. (2016). Enteral nutrition for optimal growth in preterm infants. Korean Journal of
Pediatrics, 59(12), 466–470. https://doi.org/10.3345/kjp.2016.59.12.466
Koo, W., & Tice, H. (2018). Human Milk Fortifiers Do Not Meet the Current Recommendation
for Nutrients in Very Low Birth Weight Infants. JPEN. Journal of Parenteral and
Enteral Nutrition, 42(4), 813–820. https://doi.org/10.1177/0148607117713202
Körnmann, M. N., Christmann, V., Gradussen, C. J. W., Rodwell, L., Gotthardt, M., Van
Goudoever, J. B., & Van Heijst, A. F. J. (2017). G. Nutrients, 9(12), 1318.
https://doi.org/10.3390/nu9121318
Kumar, R. K., Singhal, A., Vaidya, U., Banerjee, S., Anwar, F., & Rao, S. (2017). Optimizing
Nutrition in Preterm Low Birth Weight Infants—Consensus Summary. Frontiers in
Nutrition, 4, 20. https://doi.org/10.3389/fnut.2017.00020
Morgan, J., Bombell, S., & McGuire, W. (2013). Early trophic feeding versus enteral fasting for
very preterm or very low birth weight infants. The Cochrane Database of Systematic
Reviews, 3, CD000504. https://doi.org/10.1002/14651858.CD000504.pub4
Morgan, J., Young, L., & McGuire, W. (2014a). Slow advancement of enteral feed volumes to
prevent necrotising enterocolitis in very low birth weight infants. The Cochrane
Database of Systematic Reviews, 12, CD001241.
https://doi.org/10.1002/14651858.CD001241.pub5
Morgan, J., Young, L., & McGuire, W. (2014b). Delayed introduction of progressive enteral
feeds to prevent necrotising enterocolitis in very low birth weight infants. The Cochrane
Database of Systematic Reviews, 2014(12), CD001970.
https://doi.org/10.1002/14651858.CD001970.pub5
Wiechers, C., Bernhard, W., Goelz, R., Poets, C. F., & Franz, A. R. (2021). Optimizing Early
Neonatal Nutrition and Dietary Pattern in Premature Infants. International Journal of
Environmental Research and Public Health, 18(14), 7544.
https://doi.org/10.3390/ijerph18147544
William, W. H., Jr. (2017). Optimizing nutrition of the preterm infant. Chinese Journal of
Contemporary Pediatrics, 19(1), 1–21. https://doi.org/10.7499/j.issn.1008-
8830.2017.01.001
Andre C. Kalil, M. L. M. (2022). Management of VentilatorAssociated Pneumonia Guidelines.
Division of Pulmonary, Critical Care and Sleep Medicine, University of Connecticut
School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030-
1321, USA. https://doi.org/10.1016/j.ccm.2018.08.002
Hanser, A., Neunhoeffer, F., Hayer, T., Hofbeck, M., Schlensak, C., Mustafi, M., Kumpf, M., &
Michel, J. (2020). A nurse-driven analgesia and sedation protocol reduces length of PICU
stay and cumulative dose of benzodiazepines after corrective surgery for tetralogy of
Fallot. Journal for Specialists in Pediatric Nursing: JSPN, 25(3), e12291.
https://doi.org/10.1111/jspn.12291
William Owens. (2012). THE VENTILATOR BOOK. First Draught Press.
Yuliastati, N. (2016). Keperawatan Anak. Pusdik SDM Kesehatan.
Jurnal Anestesiologi Indonesia

LAPORAN KASUS

Manajemen Perioperatif Operasi Arterial Switch pada


Transposition of The Great Arteries with Intact Ventricular
Septum

Perioperative Management in Arterial Switch Operation of Transposition


of The Great Arteries with Intact Ventricular Septum

Dian Kesumarini, Herdono Poernomo

SMF Anestesiologi dan Terapi Pascabedah, Rumah Sakit Jantung dan Pembuluh Darah Harapan
Kita, Jakarta, Indonesia

Korespondensi: diankesumarini@gmail.com

ABSTRACT
Background: Congenital heart disease (CHD) occurs in one-third of all major
congenital anomalies. Transposition of the great arteries (TGA) is one of the most
complicated congenital heart anomalies. Arterial switch operation (ASO) is a procedure
to repair TGA. This procedure has high mortality rate and morbidity.
Case: A 42-day old infant with body weight of 3100 gram was referred to Rumah Sakit
Jantung dan Pembuluh darah (RSJPD) Harapan Kita due to cardiac anomalies.
echocardiography finding was TGA with intact ventricular septum (TGA-IVS), atrium
septal defect (ASD) secundum L-R shunt, and patent ductus arteriosus (PDA). Procedure
of repair included ASO using Le Compte maneuver, PDA ligation, partial closure of ASD
with 3mm to spare. Duration of cardiopulmonary bypass (CPB) was 136 minutes with
cross clamp of 85 minutes. Transfusion of PRC, FFP, and TC were given and the patient
was transported to intensive care unit (ICU) with haemodynamic support of adrenaline
0.05 mcg/kgBW/minute and milrinone 0.375 mcg/kgBW/minute. Extubation was done 72
hours after the surgery.
Discussion: ASO is a high-risk procedure, with high mortality and morbidity.
Perioperative anesthesia consideration for TGA patients include preanesthesia
management, hemodynamic during anesthesia, inotropic support, arrythmia due to
coronary problem and how to assess epicardial echocardiography after the surgery. In
postoperative period it is important to anticipate the adverse effects of CPB to
myocardium, low cardiac output syndrome ,risk of infection, and other complications
often seen in infants after this procedure.
Conclusion: Preoperative management by recognizing risk factors, intraoperative
anesthesia consideration, myocardial protection, and comprehensive postoperative care
in ICU are important to improve the outcome for patients undergoing this procedure.

Keywords: anesthesia consideration; arterial switch operation; cardiopulmonary


bypass; congenital heart disease; d-TGA

Volume 12, Nomor 2, Tahun 2020 11


Jurnal Anestesiologi Indonesia

ABSTRAK
Latar belakang: Penyakit jantung bawaan (PJB) berkontribusi terhadap hampir sepertiga
dari kelainan kongenital secara keseluruhan. Transposition of the great arteries (d-TGA)
adalah satu kelainan jantung bawaan (PJB) yang kompleks. Tindakan arterial switch
operation (ASO) menjadi pilihan koreksi pada kasus TGA. Tindakan ini mempunyai
risiko morbiditas dan mortalitas yang cukup tinggi.
Kasus: Bayi berusia 42 hari dengan berat badan 3100 gram dirujuk ke Rumah Sakit
Jantung dan Pembuluh Darah (RSJPD) Harapan Kita karena kelainan jantung. Pasien
dilakukan diagnosik ekokardiografi dan didapatkan TGA dengan septum ventrikular yang
intak (TGA-IVS), atrium septal defect (ASD) sekundum L-R shunt, dan patent ductus
arteriosus (PDA). Prosedur pembedahan meliputi ASO menggunakan manuver Le
Compte, pemotongan PDA, ASD ditutup sebagian dan disisakan 3mm. Durasi
cardiopulmonary bypass (CPB) 136 menit dengan cross clamp 85 menit, diberikan
tranfusi PRC, FFP, dan TC, lalu dipindahkan ke intensive care unit (ICU) dengan support
adrenalin 0.05 mcg/kg/menit dan milrinone 0.375 mcg/kg/menit. Ekstubasi dilakukan 72
jam pascaoperasi.
Pembahasan: Operasi arterial switch merupakan tindakan berisiko tinggi, dengan angka
kematian dan morbiditas yang tinggi. Konsiderasi perianestesia pada pasien TGA ini di
antaranya tatalaksana preanestesi, manajemen selama operasi, topangan hemodinamik,
aritmia yang diakibatkan masalah pembuluh darah koroner, dan penilaian ekokardiografi
epikardial pascaoperasi. Manajemen pascaoperasi penting untuk mengantisipasi efek dari
CPB yang berpengaruh pada miokardium, sindroma curah jantung rendah, risiko infeksi,
dan komplikasi lain yang sering terjadi pada infant setelah pembedahan ini.
Kesimpulan: Manajemen preoperatif dengan mengenali faktor risiko, tatalaksana
anestesia intraoperatif, myocardial protection, serta perawatan komprehensif
pascaoperasi di ICU sangat menentukan outcome pasien yang menjalani prosedur ini.

Kata Kunci: arterial switch operation; cardiopulmonary bypass; d-TGA; penyakit


jantung bawaan; tatalaksana anestesi

Volume 12, Nomor 2, Tahun 2020 12


Jurnal Anestesiologi Indonesia

PENDAHULUAN ekokardiografi dengan riwayat


Transposition of the great arteries (d- sebelumnya dirawat di intensive care
TGA) merupakan salah satu kelainan unit (ICU) selama dua hari dengan
jantung bawaan yang kompleks di mana keluhan sesak akibat pneumonia aspirasi.
pembuluh darah arteri pulmonal keluar Riwayat kelahiran spontan, usia
dari ventrikel kiri dan aorta keluar dari kehamilan ibu 36 minggu dan berat lahir
ventrikel kanan. Kelainan ini 2400 gram. Riwayat kebiruan disangkal.
mempunyai angka kejadian 5 % dari Riwayat antenatal care (ANC) ibu
seluruh pasien penyakit jantung bawaan teratur.
(PJB). Rumah Sakit Jantung dan
Pembuluh Darah (RSJPD) Harapan Kita Dari pemeriksaan fisik didapatkan berat
Jakarta merupakan salah satu center badan bayi 3100 gram dan panjang badan
kelainan jantung kongenital yang cukup 49 cm. Tekanan darah 77/40 mmHg, nadi
banyak melakukan arterial switch 154 kali per menit, respirasi 36 kali per
operation (ASO), jumlah kasus pada menit, dan saturasi oksigen 87%.
periode Maret 2017-Maret 2018 Pemeriksaan penunjang laboratorium
sebanyak 91 pasien dengan angka didapatkan Hb 14.9, leukosit 9060, CRP
mortalitas 11%, yang terdiri dari 1, ureum 18.5 mg/dl, dan kreatinin
transposition of the great arteries with 0.36mg/dl. Pada foto thorax ditemukan
ventricular septal defect (TGA-VSD) cardiothoracic ratio (CTR) 55%,
sebanyak 61 kasus dengan angka segmen pulmonal tidak menonjol,
kematian 8 pasien (13%) dan segmen aorta normal, tidak ada infiltrat
transposition of the great arteries with maupun kongesti paru. Pasien ini dirawat
intact ventricular septum (TGA-IVS) di ruang intermediate care unit RSJPD
sebanyak 30 kasus dengan angka Harapan Kita dengan menggunakan
kematian 2 pasien (6%). Angka ini prostaglandin 7 unit/kg/menit.
belum mencapai angka mortalitas
internasional pada prosedur serupa, Hasil ekokardiografi didapatkan situs
namun di Indonesia, angka ini adalah solitus, AV concordance, VA
angka yang terbaik. Beberapa penyebab discordance, semua PV to LA, ASD
utama kematian pada prosedur ASO sekundum L-R shunt diameter 3-4mm,
pada TGA-IVS ini di antaranya VSD (-), PDA continuous flow diameter
kegagalan fungsi ventrikel, masalah 1.5-2 mm, LVEF 70% (Teich), TAPSE
pembuluh darah koroner, dan infeksi, 0.9 cm, katup-katup dalam batas normal,
sedangkan pada TGA-VSD hipertensi arcus aorta di kiri, coartacio (-). Left
pulmonal menjadi salah satu ventricle (LV) mass index 44g/m2,
komorbiditas yang meningkatkan angka posterior wall diameter (PWD) 30 mm.
mortalitas pascaoperasi arterial switch. Dari hasil tersebut, disimpulkan
Pengenalan faktor risiko perioperatif, diagnosis pasien adalah TGA-IVS, ASD
tatalaksana intraoperatif dan sekundum L-R shunt, dan patent ductus
pascaoperasi sangat berperan penting arteriosus (PDA).
dalam menentukan outcome dari
prosedur ini.1 Persiapan anestesia meliputi induksi
inhalasi dengan sevoflurane. Pasien
KASUS sudah terpasang infus perifer di vena
Seorang bayi berusia 42 hari dirujuk oleh dorsum manus sinistra. Dilakukan
rumah sakit lain karena ditemukan pemasangan arterial line di arteri
kelainan jantung saat pemeriksaan brachialis sinistra, diberikan titrasi

Volume 12, Nomor 2, Tahun 2020 13


Jurnal Anestesiologi Indonesia

dengan total dosis fentanyl 10 mcg, blood pressure sebelum dipindahkan


vecuronium 1 mg, midazolam 1 mg. 71/38 mmHg, HR 150 x/menit, CVP 10
mmHg, SpO2 99 % dan inotropik
Dilakukan pemasangan endotracheal adrenalin 0.05 mcg/kg/menit dan
tube (ETT nomor 3,5 non cuff). milrinone 0.375 mcg/kg/menit.
Selanjutnya dilakukan pemasangan
kateter vena sentral di vena jugularis Selesai operasi pasien ditransfer ke ICU
internal dextra. Rumatan anestesi dengan dengan hemodinamik saat admisi ICU
campuran gas oksigen dan udara dengan tekanan darah 82/57 (52) mmHg, heart
fraksi 50%. Gas sevoflurane 1 vol %, rate 156x/m, CVP 9, SpO2 97 % dengan
fentanyl, dan vecuronium. support adrenalin 0.05 mcg/kg/menit,
milrinone 0.375 mcg/kg/menit, diberikan
Analisis gas darah (AGD) sebelum kalsium glukonas drip 10 mg/kg/jam,
masuk mesin pintas jantung dan ventilasi dikontrol dengan drip fentanyl 5
paru/cardiopulmonary bypass (CPB): mcg/kg/jam, midazolam 0.05
pH 7.28, pCO2 54,9, pO2 59,9, SO2 mg/kg/jam, dan vecuronium 0,1
84,6, BE -0,5, HCO3 26,3, laktat 0,8. mg/kg/jam, mode ventilator dengan
Tekanan darah selama pembedahan pressure control, rate 45, P above 13,
sistolik 45-70 mmHg dan diastolik PEEP 5 FiO2 50 %. Diberikan lanjutan
diantara 25-40 mmHg. Selama CPB, antibiotik profilaksis cefuroxime 3 x 30
didapatkan rentang mean arterial mg/kg sebanyak 6 dosis, dan pertoneal
pressure (MAP) sekitar 32-40 mmHg, dialysis siklus diaktifkan karena
AGD pasca-CPB pH 7.32, pCO2 45.6, olioguria. Pada hari ke-0 pascaoperasi
pO2 115,9, SO2 97.1, HCO3 24.4, laktat (POD-0), dilakukan pemeriksaan AGD
2,7. arteri dan vena pada jam I dan jam IV,
serta pemeriksaan laboratorium lengkap.
Temuan intraoperatif didapatkan jantung Dari pemeriksaan AGD arteri ditemukan
ukuran normal, vena inominata (+). pH: 7.22, pO2: 90.2, pCO2: 58.1, HCO3:
Aorta di anterior main pulmonary artery 24.4, BE: -3.0, SpO2: 95.1, laktat: 1.6.
(MPA), koroner 1 LCx, 2 R ini Dari pemeriksaan AGD vena ditemukan
merupakan varian normal dari koroner, pH 7.18, pO2 90.2, PCO2: 63.6, HCO3:
LPA-RPA konfluen diameter 4 mm, 24.4, BE: -3.1, SaO2: 55.8, laktat 1.8.
ASD sekundum 6 mm. Pemeriksaan laboratorium Hb 9.4 g/dl,
Ht 28%, leukosit 7140 /L, trombosit
Pasien dilakukan arterial switch 108.000 /L, PT : 15,7 detik, aPTT : 95
operation (ASO) menggunakan manuver detik, INR : 1,42, Albumin 3,5 g/dL,
Le Compte, pemotongan PDA, ASD detik, Ureum : 18,3 mg/dL, Cr : 0,35
ditutup sebagian dan disisakan 3mm. mg/dL. AGD POD-0, jam ke 4, arteri:
Durasi CPB adalah 136 menit, dengan pH: 7.42, PO2: 84.5, pCO2: 40.8, HCO3:
cross clamp selama 85 menit. Pasien 21.8, BE: -2.8, SaO2: 97.2%, laktat: 2.6
menerima transfusi packed red cell 60 mmol/L. Pada AGD vena ditemukan pH:
ml, fresh frozen plasma 60 ml, dan 7.32, pO2: 35.6, pCO2: 40.8, HCO3:
thrombocyte concentrate 51 ml. Total 24.1, BE: -1.3, SaO2: 67.1%, laktat: 2.8
urine output 10 ml dengan perdarahan mmol/L.
200 ml. Durasi operasi adalah 324 menit
dan durasi anestesi adalah 417 menit. Pada hari ke-0 pascaoperasi (POD-0)
Selesai operasi pasien dipindahkan ke diberikan transfusi PRC 10 ml/kg, dan
ruang ICU dengan hemodinamik arterial

Volume 12, Nomor 2, Tahun 2020 14


Jurnal Anestesiologi Indonesia

FFP 10 ml/kg, dengan total balans cairan inotropik, aritmia, akses perifer dan
-170 m/24 jam (-34 ml/kg/24 jam). sentral, laboratorium, x-ray,
electrocardiograpgy (EKG), mode
24 jam pascaoperasi (POD-1) dilakukan ventilator yang digunakan, jalan napas
evaluasi ekokardiografi, LVEF 45 %, dan status organ nonkardiak.1,2
TAPSE 0.5 cm, tidak ada regional wall
motion abnormality (RMWA), gradien Faktor risiko yang mempengaruhi
neoaorta 4 mmHg, neopulmonal 5 mortalitas antara lain patent foramen
mmHg, katup-katup baik. Pasien ovale (PFO) atau ASD restriktif,
hemodinamik stabil dengan tekanan hipertensi pulmonal persisten, berat
darah 71/40 (57) mmHg, HR: 140 badan lahir rendah (BBLR),
x/menit, support adrenalin 0.05 prematuritas, dan usia saat diketahui
mcg/kg/menit, dan milrinone 0.75 terdapat TGA.2,3
mcg/kg/menit, Ca glukonas 10
mg/kg/jam. Hasil AGD arteri Pada kelainan TGA-IVS terjadi fisiologi
menunjukkan pH 7,46, pO2 175, pCO2: paralel sirkulasi di mana tidak ada
33.9, HCO3: 24.7, BE: 0.8, SpO2: hubungan antara darah teroksigenasi
99.9%, laktat: 1.1mmol/L. Sementara dengan darah tidak teroksigenasi, maka
itu, hasil AGD vena menunjukkan pH: dari itu pada kelainan ini sangat penting
7.43, PCO2: 40.2, pO2: 40.7, BE: 0.8, untuk menjaga kecukupan/keadekuatan
SpO2: 76.8%, laktat: 1 mmol/L. Pasien mixing antara darah yang teroksigenasi
diberikan terapi tambahan furosemide 3 dengan darah yang tidak teroksigenasi
x 1 mg/kg intravena dan aldactone 1 x antara jantung kanan dengan jantung kiri.
6,25 mg. 12 jam pascaoperasi pasien Percampuran (mixing) ini dapat terjadi
diberikan gut feeding dan dilanjutkan pada tingkat atrium ataupun ductus
pemberian nutrisi enteral. arteriosus (PDA). Pada pasien dengan
ASD yang kecil (restriktif) perlu
24 jam pascaoperasi pasien dilakukan ballon atrial septectomy
dibangunkan, weaning ventilator (BAS), untuk menjaga percampuran
perlahan. 72 jam pascaoperasi pasien darah teroksigenasi dengan darah yang
diekstubasi, dengan total balans cairan tidak teroksigenasi pada tingkat atrial,
selama 3 hari. Pascaekstubasi dosis karena aliran ke paru berasal dari
milrinone diturunkan secara titrasi, ventrikel kiri, sedangkan darah yang
diganti dengan captopril 3 x 0.3 mg/kg tidak teroksigenasi berasal dari atrium
per oral. Setelah 12 jam pascaoperasi kanan, maka penting untuk menjaga
dilakukan down titrasi adrenalin sampai percampuran darah teroksigenasi pada
dengan stop, diganti dengan dobutamin 5 atrium kiri dengan darah tidak
mcg/kg/menit. Selama target nutrisi per teroksigenasi pada atrium kanan untuk
oral belum tercapai, diberikan menjaga aliran ke paru adekuat sehingga
maintenance cairan D40 % NS 3:1 dan pasien dapat mempertahankan saturasi.
protein parenteral 1 gr/kg/hari. Percampuran ini juga terjadi pada tingkat
PDA di mana terjadi percampuran darah
PEMBAHASAN antara aorta dengan arteri pulmonalis,
Manajemen Perioperatif untuk itu pada periode preoperatif
Beberapa hal yang harus diperhatikan penting untuk menjaga patensi PDA
pada perioperatif pasien transposition of dengan pemberian infus prostaglandine
the great arteries (TGA) antara lain (PGE1) dengan tujuan mempertahankan
status hemodinamik, level support

Volume 12, Nomor 2, Tahun 2020 15


Jurnal Anestesiologi Indonesia

kecukupan aliran ke paru dan saturasi opioid yang tinggi dikombinasi dengan
untuk perfusi organ. berkurangnya preload dan kontraktilitas
kedua ventrikel, menyebabkan aliran
Penilaian perioperatif pada pasien TGA- oksigen yang lebih rendah karena mixing
IVS antara lain evaluasi percampuran pada tingkat atrial. Penilaian kecukupan
(mixing) pada tingkat atrium setelah diameter BAS dapat bias karena mixing
prosedur BAS, evaluasi efek samping pada PDA, PDA dapat menutup bila
pemakaian PGE1 seperti apneu, prostaglandin dihentikan. Manajemen
hipotensi, demam, eksitasi sistem saraf pasien TGA termasuk memastikan
pusat, dan penurunan volume oksigenasi dan ventilasi, meningkatkan
intravaskular pada lesi ductus dependent. kedalaman anestesi tanpa menurunkan
Adanya patent ductus arteriosus (PDA) kontraktilitas jantung.1-3
menurunkan aliran darah ke sistem
gastrointestinal sehingga berpotensi Pada penilaian transesophageal
menjadi NEC. Sedangkan, pada pasien echocardiography (TEE) hal yang harus
TGA-VSD, evaluasi yang harus dinilai di dievaluasi setelah CPB di antaranya lesi
antaranya tanda kelebihan sirkulasi ke residual, regional dan global ventricular
paru akibat adanya shunt melalui septum function untuk memastikan tidak terjadi
ventrikel (VSD). Kejadian ini pada insufisiensi koroner akibat pemindahan
jangka panjang dapat menyebabkan pembuluh darah koroner yang tidak
gagal jantung atau congestive heart sempurna, fungsi katup-katup, evaluasi
failure (CHF). Tanda gagal jantung ini regurgitasi, dan mendeteksi gradien
antara lain peningkatan heart rate, anastomosis supravalvar pada neoaorta
kardiomegali, penurunan temperatur, dan neopulmonal, juga regional wall
diaforesis, berat badan rendah, takipneu, motion.3,4
retraksi subkosta dan substernal, prolong
capillary refill.2 Setelah weaning CPB, TEE dan
monitoring left arterial pressure (LAP)
Manajemen intraoperatif dilakukan untuk menilai global dan
Induksi dengan inhalasi dilakukan pada regional LV function. LAP ini juga dapat
pasien yang tidak ada akses intravena. menilai adanya perfusi ke miokardium
Teknik anestesi opioid base juga yang tidak adekuat. Sebagian pasien
dianjurkan pada pasien ini. Prinsip memerlukan inotropik dan beberapa ahli
manajemen anestesi pada TGA-VSD merekomendasikan pemberian
yang disertai hipertensi pulmonal antara nitrogliserin 1-2 mcg/kg/menit untuk
lain oksigenasi dan ventilasi yang dilatasi arteri koroner dan menurunkan
adekuat, meningkatkan kedalaman preload. Walaupun tidak umum terjadi,
anestesi tanpa menurunkan kontraktilitas obstruksi dan translokasi arteri koroner
jantung.1,3 dapat menjadi masalah serius pada
pasien setelah operasi ASO. Tanda
Pada pasien TGA-IVS dengan mixing utama dari kejadian ini adalah disfungsi
yang adekuat pada tingkat atrial maupun miokardia global, yang dapat dilihat dari
PDA, seringkali terjadi desaturasi pada ekokardiografi. Regional wall motion
saat induksi dan ventilasi positif. abnormalities (RWMA) tidak
Penyebab penurunan mixing ini bermanifestasi pada pasien neonatus.
multifaktorial. Ventilasi tekanan positif Target hemodinamik saat off bypass pada
dan paralisis yang disertai hilangnya pasien TGA adalah cardiac output yang
sekresi katekolamin endogen karena adekuat, tekanan LA serendah mungkin,

Volume 12, Nomor 2, Tahun 2020 16


Jurnal Anestesiologi Indonesia

tekanan darah sistolik berkisar 50-75 Pada jantung normal, 3 pembuluh darah
mmHg. Bila LV overdistended (ditandai arteri utama yang berhubungan dengan
dengan LAP yang tinggi dan penurunan great arteries antara lain: (1) arteri
tekanan sistemik dan cardiac output), koroner kanan, merupakan arteri koroner
diuretik dapat diberikan bila yang melalui atrioventricular groove;
hemodinamik sangat terganggu akibat (2) circumflex coronary artery,
peningkatan LAP, untuk mengeluarkan merupakan arteri coroner yang melintasi
volume darah melalui CVP sampai LAP atrioventricular groove; (3) anterior
menurun. Pengambilan darah untuk descending artery, arteri koroner yang
sampel AGD juga dapat mempengaruhi paralel dengan interventricular septum
volume intravaskular. Pada saat pada permukaan anterior jantung.4,6 Hal
transport pasien ke ICU harus ini diilustrasikan dalam Gambar 1.
diperhatikan airway, ventilasi, patensi
inotropik dan suhu. 3-5

Gambar 1. Distribusi arteri koroner pada TGA. Sconamiglio, G6

Volume 12, Nomor 2, Tahun 2020 17


Jurnal Anestesiologi Indonesia

Bagian atas proyeksi diagnostik diastolik lebih dari 30 mmHg, MAP


merupakan gambaran ekokardiografi dua lebih dari 40 mmHg, support
dimensi, sedangkan bagian bawah hemodinamik dapat diberikan inotropik
frontal view, distribusi koroner dari sisi dan inodilator seperti dobutamin atau
anterior (surgeon view).6 adrenalin, dan milrinone. Target perfusi
pada hari pertama yaitu urine output
Operasi arterial switch melibatkan lebih dari 1 ml/kg/jam. Analisa gas
pembuluh darah besar (great artery) dan darah target PH 7,35-7,45, base excess
translokasi pembuluh darah dan asal yang rendah. Bila terjadi sindrom low
pembuluh darah lainnya. Dengan cardiac output, harus dilakukan evaluasi
demikian, membentuk ekokardiografi untuk mengetahui
ventriculoarterial concordance (aorta adanya disfungsi LV, RV, RWMA,
ke ventrikel kiri). Translokasi aorta ini outflow tract obstruction, stenosis
juga termasuk mobilisasi dan pulmonal dan aorta supravalvar.
reimplantasi arteri koroner. Pada operasi Sedangkan pada pasien TGA-VSD
arterial switch ini, dibuat “button” harus dievaluasi adanya VSD, ASD
diantara koroner dan dimplantasikan ke residual, fungsi katup atrioventrikular,
ujung neoaorta. Pada prosedur ini tanda-tanda hipertensi pulmonal
dealinasi anatomi arteri koroner paskaoperasi, irama jantung, dan tanda-
sangatlah penting, karena seringkali tanda insufisiensi koroner. Hb
terdapat anomali posisi arteri koroner dipertahankan 10, fibrinogen di atas
saat sebelum dikoreksi.3,4,6 160, trombosit lebih dari 100.000, PT
kurang dari 15, PTT kurang dari 60, dan
Lesi residual yang sering pada operasi normotermia.7,8
arterial switch di antaranya branch
pulmonary artery stenosis. Stenosis Pada delayed stenal closure, penutupan
arteri pulmonalis merupakan salah satu dinding dada dilakukan bila status
tanda iskemia koroner dan left arterial hemodinamik pasien stabil, termasuk
hypertension. Lesi obstruksi residual tekanan darah, heart rate, dan analisa
pada left and right outflow dapat gas darah arteri.7,8
menjadi problem paskaoperasi.
Ventrikel kiri harus memompa melawan Hal-hal yang menghambat ekstubasi
tahanan di SVR, sehingga memerlukan antara lain perdarahan, hemodinamik
support inotropik dan afterload yang tidak stabil. Hb dipertahankan 12
reduction. Pemberian volume harus sampai dengan 13 g/dl, dan faal
hati-hati, karena compliance jantung koagulasi yang normal, dan keadaan
kurang baik. Sebelum pasien pasien yang normotermia. Untuk
dipindahkan dari kamar operasi harus pemberian nutrisi paskaoperasi jantung
diperhatikan tanda-tanda insufisensi harus memperhatikan status
koroner yang merupakan tanda iskemik hemodinamik. Initial feeding dapat
miokard, termasuk evaluasi global diberikan per NGT, dengan jumlah total
fungsi dari ventrikel kiri.1-5 enteral per hari 20 kkal/kg/hari. Bila
toleransi baik, dapat ditingkatkan
Pascaoperasi bertahap hingga mencapai target
Pada TGA, target hemodinamik hari volume enteral 120-140 ml/kg/hari,
pertama adalah heart rate kurang dari dengan catatan peningkatan volume
160 x/menit, tekanan darah sistolik dapat diberikan 20 ml/kg/hari.
lebih dari 60 mmHg, tekanan darah Sedangkan target kalori yang dapat

Volume 12, Nomor 2, Tahun 2020 18


Jurnal Anestesiologi Indonesia

dicapai 120-150 kkal/kg/hari, ketika seperti pada pasien dalam terapi


target volume tercapai maka diberikan imunosupresi, kanker, dan pada pasien
nutrisi padat kalori.7-9 yang menjalani operasi. Lain halnya
dengan antibiotik empirik yang
Antibiotik profilaksis merupakan diberikan pada pasien yang terbukti
antibiotik yang diberikan untuk infeksi atau diduga infeksi. Peralihan
mencegah infeksi yang belum terjadi. dari antibiotik profilaksis menjadi
Antibiotik profilaksis hanya digunakan empirik dapat dilihat pada Gambar 2.10
pada pasien dengan risiko tinggi infeksi,

Gambar 2. Proses peralihan dari antibiotik profilaksis menjadi empiric

Disfungsi kardiak seringkali terjadi infant memiliki protein kontraktil yang


pada pascaoperasi jantung kongenital, lebih sedikit, dan penyimpanan kalsium
yang disebabkan oleh inflamasi dari dan sarkoplasmik yang lebih sedikit
CPB, yang menyebabkan pelepasan dibandingkan otot miokardium yang
TNF- yang mempengaruhi fungsi sudah matur. Sehingga otot miokardium
kontraksi kardiomiosit. Inflamasi juga infant mempunyai compliance yang
menyebabkan perubahan struktur dan lebih rendah dan respon cairan yang
fungsi endotel, yang menyebabkan lebih rendah dibandingkan dewasa.
edema interstitial sehingga menurunkan Dengan demikian, sangat mudah terjadi
compliance otot miokardium. peningkatan afterload pada infant
Mekanisme utama disfungsi miokardial dibandingkan anak-anak dan dewasa.
adalah adanya periode iskemia Kebutuhan oksigen pada infant lebih
(cardioplegic arrest) dan reperfusion tinggi daripada anak-anak dan dewasa
injury, juga disfungsi miokardial pada sehingga memerlukan cardiac output
periode preoperatif. Efek mesin CPB dan minute ventilasi yang lebih tinggi.
terhadap miokardia pada infant lebih Pada periode pascaoperasi ventilasi
besar daripada pasien anak-anak mekanik digunakan untuk
maupun dewasa, hal ini disebabkan mengoptimalisasi delivery oxygen dan

Volume 12, Nomor 2, Tahun 2020 19


Jurnal Anestesiologi Indonesia

mengurangi konsumsi oksigen yang jaringan. Syok itu sendiri didefinisikan


dapat meningkat karena napas spontan. sebagai delivery oxygen yang tidak
Ventilasi positif mempunyai efek adekuat atau ketidakseimbangan antara
menurunkan preload dan afterload demand dan supply oksigen.
ventrikel kiri. Waktu inspirasi dapat Peningkatan demand oksigen
mencegah menurunan filling pressure pascaoperasi dapat disebabkan oleh
ventrikel kiri sehingga cardiac output inflamasi sistemik yang merupakan
lebih optimal. Beberapa literatur respon mesin CPB, demam, pelepasan
menyatakan bahwa rerata lama periode katekolamin endogen atau eksogen,
ventilasi mekanik pascaoperasi arterial stress respon surgery, respon bangun,
switch sekitar 3 hari dan proses weaning dan pernapasan spontan.
ventilator mencapai 12-24 jam.10-12 Ketidakseimbangan demand-supply
oksigen merupakan hasil dari kurang
Pada periode pasca-CPB seringkali adekuatnya delivery oxygen yang
terjadi disfungsi vaskuler yang disebabkan anemia dan low cardiac
menyebabkan peningkatan resistensi output. Penyebab low cardiac output itu
sistemik dan pulmonal. Peningkatan sendiri di antaranya venous return yang
resistensi paru ini dapat menyebabkan tidak adekuat, disfungsi ventrikel,
terjadinya disfungsi ventrikel kanan. afterload yang tinggi, atau aritmia yang
Pada beberapa kasus, pascaoperasi mempengaruhi fungsi miokardium,
arterial swtich dilakukan delayed sehingga mengurangi cardiac output.
sternal closure terutama pada pasien Inflamasi sistemik menyebabkan
dengan perdarahan, ukuran jantung leakage pembuluh darah sehingga
lebih besar secara signifikan karena menurunkan volume intravaskuler, dan
edema, dan pada keadaan di mana preload di ventrikel. Pada kasus di mana
fungsi ventrikel sangat menurun.11-13 preload berkurang karena disfungsi
Pada delayed stenal closure, penutupan diastolik maka diperlukan peningkatan
dinding dada dilakukan bila status filling pressure ventricle untuk
hemodinamik pasien stabil, termasuk mempertahankan stroke volume yang
tekanan darah, heart rate, dan analisis adekuat.8,11
gas darah arteri.7,8
Terapi vasoaktif pada pascabedah
Tamponade jantung merupakan salah jantung kongenital untuk memperbaiki
satu komplikasi serius yang dapat disfungsi kardiovaskular harus
menyebabkan hipotensi hingga cardiac memperhatikan fungsi sistolik dan
arrest. Tamponade didefinisikan sebagai diastolik ventrikel kanan dan kiri.
akumulasi cepat dari cairan, darah, dan Vasoaktif yang menyebabkan
udara pada ruang perikardium yang venodilatasi dapat menurunkan venous
meningkatkan tekanan perikardial dan return sehingga menurunkan volume
menurunkan venous return karena dan tekanan diastolik ventrikel,
penekanan pada atrium dan ventrikel sehingga tidak terjadi peningkatan
kanan. Tanda-tanda impending cardiac output. Sebaliknya vasoaktif
tamponade antara lain takikardia, perfusi yang menyebabkan dilatasi arteri dapat
perifer yang menurun, dan peningkatan meningkatkan ejeksi ventrikel, dan tidak
tekanan vena sentral.11 terjadi dilatasi vena, sehingga tidak
terjadi penurunan venous return karena
Pada periode pascaoperasi sangat tidak mempengaruhi kapasitansi
8,11
penting mempertahankan oksigenasi vena.

Volume 12, Nomor 2, Tahun 2020 20


Jurnal Anestesiologi Indonesia

Adrenalin merupakan katekolamin yang DAFTAR PUSTAKA


terbentuk dari norepinefrin yang disertai 1. Davies LK, Husain A, Weitzel NS.
gugus N-methylation. Pada dosis kurang Anesthetic management for patient
dari 0.1 mcg/kg/menit adrenalin with congenital heart disease: The
menurunkan resistensi vaskuler dan pediatric Population. In: Hensley,
pulmonal melalui stimulasi reseptor  -2. FA, Martin DE, Gravlee GP,
Pada dosis tinggi adrenalin editors. A Practical Approach to
menstimulasi reseptor -1 yang Cardiac Anesthesia. 5th Ed.
menyebabkan vasokonstriksi sehingga Philadelphia: Lippincott Williams
meningkatkan kapasitansi arteri dan and Wilkins; 2013. Chapter 14;
vena. Studi yang dilakukan Mekkaw, p.389-429
dkk menyatakan bahwa ada hubungan 2. Butts RJ, Ellis AR, Bradley SM,
antara lama ischemic time dengan Hulsey TC, Atz AM. Effect of
peningkatan skor inotropik.11,14 prostaglandin duration on outcomes
in transposition of the great arteries
Milrinone bekerja dengan menginhibisi with intact ventricular septum.
enzim phosphodiesterase inhibitor III Congenital Heart Disease. 2012
sehingga mengurangi degradasi cAMP Sept 23;7(4): 387-91.
sehingga menyebabkan penurunan 3. Gupta S, Saiyed A, Meena R, Dogra
kapasitansi vena dan arteri serta N. Anesthetic management during
mempunyai efek inotropik ringan.8,11,15 transposition of great arteries
(TGA) correction: point to be focus.
KESIMPULAN British Journal of Medicine
(1) Penilaian status perioperatif &Medical Research. 2017; 19(8):
TGA-IVS terdiri dari kecukupan mixing, 1-5
support hemodinamik, efek samping 4. Nield LE, Dragulescu A, MacColl
PGE1, dan status organ nonkardiak; (2) C, Brun H, McCrindle BW, Kuiper
Evaluasi intraoperatif harus B, et al. Coronary artery doppler
mempertimbangkan balans anestesia patterns are associated with clinical
dengan kedalaman yang cukup tanpa outcomes post-arterial switch
mengganggu hemodinamik dan operation for transposition of the
kontraktilitas. Pada penilaian TEE great arteries. European Heart
pascaoperasi harus diperhatikan lesi Journal-Cardiovascular Imaging.
residual, regional dan global ventricular 2017; 19(4): 461-68.
function, fungsi katup-katup, evaluasi 5. Villafane J, Hermoso RL, Bhatt
regurgitasi, dan mendeteksi gradien AB, Tweddell JS, Geva T, Nathan
anastomosis. Setelah weaning CPB, TEE M, et al. D-Transposition of the
dan monitoring left arterial pressure Great Arteries. The Current Era of
(LAP) untuk menilai global dan regional the Arterial Switch Operation.
LV function, dan LAP ini juga dapat Journal Of American College Of
menilai adanya perfusi ke miokardium Cardiology. Elsevier. 2014; 64
yang tidak adekuat; (3) Hal-hal yang (5):498-511
harus diperhatikan pada pascaoperasi 6. Sconamiglio, G. Arterial switch
pasien TGA-IVS antara lain target operation for transposition of great
perfusi, echo evaluasi, irama jantung, arteries: late results in adult
fungsi koroner, dan hemodinamik. patients. International
Cardiovascular Forum Journal.
2013;1: 8-15

Volume 12, Nomor 2, Tahun 2020 21


Jurnal Anestesiologi Indonesia

7. Rome J. Inpatient Pathway for Rogers’ Textbook of Pediatric


Evaluation/Treatment of the Intensive Care. 5th ed. Philadelphia:
Newborn with TGA. Children’s Wolters Kluwer; 2015. Chapter 79,
Hospital of Philadelphia. 2016; 1-3. p. 1252-60
Available from 12. Lu BY, Wu HD, Wang CC, Wu ET,
https://www.chop.edu/clinical- Huang SC, Ko WJ, et al. The Impact
pathway/newborn-tga-clinical- of Postoperative Ventilator Support
pathway-immediate-post-operative- on Outcome of the Arterial Switch
cicu-care-hand-or-team. Operation-Report from a Single
8. Sarris GE, Balmer C, Bonou P, Institute. Acta Cardiol Sin.
Comas JV, Cruz ED, Chiara LD, et 2010;26:173-8
al. Clinical guideline for 13. Gocen U. Delayed sternal closure
management of patients with after arterial switch operations: a
transposition of the great arteries single center experience. 2017;
with intact ventricular septum. Cukurova Med J.2017;42(3):540-5
European Journal of Cardio-thoracic 14. Mekkawy A, Ghoneim, El-Haddad
Surgery.2017;51:1-32 O, & Elmishaway A. Predictors of
9. Justice L, Buckley JR, Floh A, early outcome of arterial switch
Horsley M, Alten J, Anand V, et al. operation on patient with D-TGA.
Nutrition consideration in the Journal of the Egyptian Society of
pediatric cardiac intensive care unit Cardio-Thoracic Surgery. 2017;
patient. World Journal for Pediatric 25(1), 52-7
and Congenital Heart Surgery. 15. Burkhardt BEU, Rücker G, Stiller B.
2018; 9(3): 333-43. Prophylactic milrinone for the
10. Gallagher JC, MacDougall C. prevention of low cardiac output
Antibiotics Simplified. 4th ed. syndrome and mortality in children
Philadelphia: Jones &Bartlett Inc: undergoing surgery for congenital
2017:26-8 heart disease. Cochrane Database of
11. Bronicki RA, Costello JM, Brown Systematic Reviews 2015, Issue 3.
KL. Postoperative care of the Art. No.: CD009515.
pediatric cardiac surgical patient. In DOI:10.1002/14651858.CD009515.
Shaffner DH, Nichols DG editors. pub2

Volume 12, Nomor 2, Tahun 2020 22


International Journal of
Environmental Research
and Public Health

Review
Optimizing Early Neonatal Nutrition and Dietary Pattern in
Premature Infants
Cornelia Wiechers 1, * , Wolfgang Bernhard 1 , Rangmar Goelz 1 , Christian F. Poets 1 and Axel R. Franz 1,2

1 Department of Neonatology, University Children0 s Hospital, Eberhard Karls University, Calwerstr. 7,


72076 Tübingen, Germany; wolfgang.bernhard@med.uni-tuebingen.de (W.B.);
rangmar.goelz@med.uni-tuebingen.de (R.G.); christian-f.poets@med.uni-tuebingen.de (C.F.P.);
Axel.Franz@med.uni-tuebingen.de (A.R.F.)
2 Center for Pediatric Clinical Studies, University Children0 s Hospital, Eberhard Karls University,
72076 Tübingen, Germany
* Correspondence: Cornelia.wiechers@med.uni-tuebingen.de; Tel.: +49-7071-29-62310

Abstract: Providing adequate amounts of all essential macro- and micronutrients to preterm infants
during the period of extraordinarily rapid growth from 24 to 34 weeks’ postmenstrual age to achieve
growth as in utero is challenging yet important, since early growth restriction and suboptimal
neonatal nutrition have been identified as risk factors for adverse long-term development. Along
with now well-established early parenteral nutrition, this review emphasizes enteral nutrition, which
should be started early and rapidly increased. To minimize the side effects of parenteral nutrition
and improve outcomes, early full enteral nutrition based on expressed mothers’ own milk is an
 important goal. Although neonatal nutrition has improved in recent decades, existing knowledge

about, for example, the optimal composition and duration of parenteral nutrition, practical aspects of
Citation: Wiechers, C.; Bernhard, W.;
the transition to full enteral nutrition or the need for breast milk fortification is limited and intensively
Goelz, R.; Poets, C.F.; Franz, A.R.
discussed. Therefore, further prospective studies on various aspects of preterm infant feeding are
Optimizing Early Neonatal Nutrition
needed, especially with regard to the effects on long-term outcomes. This narrative review will
and Dietary Pattern in Premature
summarize currently available and still missing evidence regarding optimal preterm infant nutrition,
Infants. Int. J. Environ. Res. Public
Health 2021, 18, 7544. https://
with emphasis on enteral nutrition and early postnatal growth, and deduce a practical approach.
doi.org/10.3390/ijerph18147544
Keywords: preterm infant; nutrition; enteral feeding advancements; growth
Academic Editors:
Luís Pereira-da-Silva, Gustavo Rocha
and Susana Pissarra
1. Background
Received: 28 May 2021 Feeding preterm infants is challenging because their nutritional needs are higher
Accepted: 12 July 2021
than those of term infants. This is due to their 4-fold higher physiological growth rate
Published: 15 July 2021
during this developmental period—the last trimester of fetal development—which occurs
in the neonatal intensive care unit (NICU) rather than in utero. This phase is characterized
Publisher’s Note: MDPI stays neutral
by extraordinarily rapid, exponential growth from 24 to 34 weeks’ postmenstrual age
with regard to jurisdictional claims in
(PMA), and the acquirement of notable amounts of lean body mass, fat and reserves of
published maps and institutional affil-
micronutrients. Remarkably, between 24 and 40 weeks PMA, adipose tissue increases
iations.
80 fold, water 4 fold and lean body mass solid matter 11 fold [1].
Achieving growth rates of all body compartments and organs that are similar to
those in utero should be the primary goal of preterm infant nutrition [2]. Despite more
intensive feeding strategies for preterm infants in recent years, growth failure remains a
Copyright: © 2021 by the authors.
common problem in very preterm infants during their postnatal hospitalization [3,4] and is
Licensee MDPI, Basel, Switzerland.
associated with impaired neurocognitive outcomes [5–8]. Furthermore, premature infants
This article is an open access article
are at an increased risk of cardiovascular disease and insulin resistance in adulthood [9–11].
distributed under the terms and
The underlying link between preterm birth and later metabolic alterations is still poorly
conditions of the Creative Commons
understood, and early-life growth restriction, as well as excessive catch-up growth after
Attribution (CC BY) license (https://
initial growth failure, have been reported [8,12]. Conceivably, the inadequate postnatal
creativecommons.org/licenses/by/
4.0/).
growth of preterm infants may have deleterious “programming” effects on metabolic

Int. J. Environ. Res. Public Health 2021, 18, 7544. https://doi.org/10.3390/ijerph18147544 https://www.mdpi.com/journal/ijerph
Int. J. Environ. Res. Public Health 2021, 18, 7544 2 of 13

health similar to those of intrauterine growth restriction in term infants [13,14], occurring
during the same period of development.
Postnatal growth of preterm infants in the NICU is the result of a complex interaction
of various factors, such as neonatal morbidity and inflammation preventing anabolism,
increased respiratory work causing increased energy requirements and, of course, nutrition
and the immaturity of the gastrointestinal tract [15]. Therefore, providing adequate macro-
and micronutrients to preterm infants and achieving growth similar to that in utero is
challenging [2].
In the last decades, an increasing number of studies has been carried out regarding
the nutrition of premature infants [16–20], but knowledge remains limited, e.g., concerning
the optimal macro- and micronutrient intake through parenteral nutrition (both in the first
week after birth and thereafter), the best way to achieve full enteral feeding, or indications
for and the optimal composition of a breast milk fortifier.
Furthermore, long-term outcome data in infants following various nutritional inter-
ventions are often lacking. This review summarizes the perspective of a level 3 NICU with
a focus on very early enteral nutrition and its repercussions on other aspects of neonatal
nutrition (Tables 1 and 2). Therefore, we searched PubMed and the Cochrane library for
each of the topics addressed above to provide a narrative review of current evidence, open
questions, as well as controversial aspects.

Table 1. Summary of recommendations for postnatal parenteral nutrition in preterm infants according to the ESPGHAN
guideline 2018.

ESPGHAN Recommendations for Parenteral Nutrition


- Start: Day 1 with at least 1.5 g/kg/d, day 2 and onwards 2.5 g/kg/d and 3.5 g/kg/d, accompanied by
Amino acids [21]
non-protein intakes >65 kcal/kg/d
- Start: (a) immediately or no later than 2 days after birth (b) at discontinuation of enteral feeding at the
time of onset of PN
- Intake: parenteral lipid intake should not exceed 4 g/kg/day
- Essential fatty acids: providing a minimum linoleic acid intake of 0.25 g/kg/day
Lipids [22]
- Administration: continuously over 24 h
- Pure soybean oil: May provide a less balanced nutrition than composite intravenous lipid emulsions.
PN lasting longer than a few days, pure SO ILEs should no longer be used
- continuous over 24 h
- Parenteral glucose supply in mg/kg per min (g/kg per day):
Carbohydrates [23] Start: day 1: 4–8 (5.8–11.5), day 2 onwards: target 8–10 (11.5–14.4), min 4 (5.8); max 12 (17.3)
- Glucose blood glucose levels: Avoid Hyperglycemia (>8 mmol/L/145 mg/dL) or Hypoglycemia
(≤2.5 mmol/L/45 mg/dL)
Intake in mmol
Calcium Phosphorus Magnesium
(mg)/kg/d
Calcium,
0.8–2.0 1.0–2.0 0.1–0.2
phosphorus and First days
(32–80) (31–62) (2.5–5.0)
magnesium [24]
Growing 1.6–3.5 1.6–3.5 0.2–0.3
premature (100–140) (77–108) (5.0–7.5)
- Infants receiving PN should receive parenteral vitamins
- Administration of water- and fat-soluble vitamins in fat emulsion to increase vitamin stability
Vitamins [25]
- Recommendations for doses of individual vitamins are provided in the guideline [25], but optimal
doses and infusion conditions for vitamins in infants are not known
- Following trace minerals shoud be provided in preterm infants with PN:
- Zinc: 400–500 µg/kg/day
- Copper: 40 µg/kg/day
Trace minerals [26] - Iodine: 1–10 µg/kg daily
- Selenium: 7 µg/kg/day
- Manganese: in long term PN max. 1 µg /kg/day
- Molybdenum: in long term PN max. 1 µg /kg/day
Int. J. Environ. Res. Public Health 2021, 18, 7544 3 of 13

Table 1. Cont.

ESPGHAN Recommendations for Parenteral Nutrition


- Prefer enteral rather than parenteral administration
- NO administration in short term PN (<3 weeks)
Iron [26] - Monitoring of iron status
- If necessary: 200–250 µg/kg/day
- CAVE: no intravenous iron preparation is approved for pediatric use in Europe

Table 2. Practice Points.

- Bridge the period until full enteral feeding is established [21–23,27]


Parenteral Nutrition - Start immediately after birth with glucose, amino acids and fat [21–23]
- Use standardized parenteral nutrition solutions for most preterm patients [28]
- Start enteral nutrition on day 1 [21]
Transition from parenteral
- Accelerate enteral feeding volume in daily increments of 25–30 mL/kg [29]
to enteral nutrition
- Ignore gastric residuals as long as abdominal findings are normal [17,30]
- Promote and support lactation [31]
- Bridge the period until sufficient mother’s own milk production with DHM [2,20,31–33]
Enteral Nutrition - Fortify breastmilk to improve postnatal growth [2,34], consider adapted or individualized
fortification to meet nutritional needs [35]
- Consider short-term pasteurization in ELBW/VLBW to prevent pCMV infection [36,37]

2. Parenteral Nutrition
During the first postnatal days, complementary to enteral feeding, parenteral nutrition
is an integral part of preterm infant care, bridging the period until full enteral feeding is
established. Although neonatal parenteral nutrition has been established since the late
1960s [38], and has improved considerably since, evidence on the optimal composition
of macro- and micronutrients is limited, yet intensively discussed. Several studies and
systematic reviews have shown improved short-term growth and a shortened time to
regain birth weight using neonatal parenteral nutrition. However, according to randomized
controlled trials, its long-term benefit for metabolism and neurological development is still
unclear [39–42]. Nevertheless, data on associations between higher nutrient intake and
improved growth suggest that parenteral nutrition in the first postnatal weeks is likely to
improve cognitive outcomes [5,42,43].
Therefore, initiating parenteral nutrition immediately after delivery is recommended
for preterm infants (Table 1) [27]. Since 2018, the guidelines of the European Society for
Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) recommend an amino
acid supply of at least 1.5 g/kg/d for the first postnatal day, increasing to 2.5–3.5 g/kg/d
from postnatal day 2 onwards [21]. To provide a rich source of energy at low volume,
intravenous lipid emulsions are an indispensable component of neonatal parenteral nu-
trition. These can be started shortly after birth but should not exceed 4 g/kg/d [22]. In a
systematic review including 29 studies with >2000 infants, no benefit of new lipid emul-
sions including fish oil, compared to conventional soybean oil-based lipid emulsions, was
found for the prevention of cholestasis, growth, mortality, retinopathy of prematurity and
bronchopulmonary dysplasia (BPD) [41]. However, according to the current ESPGHAN
recommendation, the latter emulsions should not be used for more than a few days in
term and preterm neonates, as pure soybean oil may provide a less balanced nutrition than
compound fat emulsions (e.g., soybean/olive with or without fish oil) [22].
A parenteral glucose supply should start at 4–8 mg/kg/min, and avoid overfeeding
or excessive glucose load by regular blood glucose measurements [23]. The maximum
endogenous glucose production, as well as the glucose oxidation rate, which both are
approximately 7–8 mg/kg/min (10–11.5 g/kg/day) in preterm infants, should not be
exceeded, at least not initially. In addition, though effects on growth have mostly been
studied for macronutrient intakes, an adequate micronutrient intake is also necessary for
Int. J. Environ. Res. Public Health 2021, 18, 7544 4 of 13

tissue, particularly parenchyma accretion [24,25]. In line with this, early and enhanced post-
natal parenteral nutrition is associated with increased electrolyte requirements, particularly
concerning phosphate and potassium, to meet the increased anabolism of parenchymal
tissues [24,44].
Standardized parenteral nutrition solutions and computerized prescriptions are rec-
ommended to improve patient safety [28]. Therefore, the authors practice nutrition with
standardized in-house “parenteral starter solutions” starting in the first postnatal hour and
containing 3.5 g/kg amino acids and 4.2 mg/kg/min glucose, as well as small amounts of
calcium, phosphate, and sodium. With an additional standardized fat emulsion including
fat- and water-soluble vitamins, a fat intake of 2.5 g/kg/d is achieved from the first day of
life onwards.

3. Transition from Intravenous to Enteral Sources


Prolonged parenteral nutrition is associated with cholestasis, thrombosis, infectious
and metabolic risks [29], partly due to an inadequate composition of existing products.
Hence, meeting nutritional needs through full enteral nutrition is a general goal. Re-
markably, the intestine of an extremely premature infant can already digest, tolerate and
metabolize human milk. In utero, amniotic fluid with its bioactive peptides not only plays
an important role in fetal gastrointestinal development [45], but animal data show that up
to 14% of intrauterine nutrient requirements are supplied and absorbed prenatally through
the intestine [46,47].
Nevertheless, evidence-based recommendations for the transition from intravenous to
enteral nutrient supply are lacking, and the optimal rate of enteral feeding advancements
in preterm infants is unclear [16,29,48,49]. Frequently, there are concerns that rapid enteral
feeding advancements may cause necrotizing enterocolitis (NEC). In a systematic review
of 9 studies including 1106 preterm infants, however, no increased risk of NEC was found
with early enteral feeding starting within 3 days after birth compared to a more delayed
onset [50]. Current evidence suggests that accelerating enteral feeding volume in daily
increments of 30 mL/kg does not increase the risk of NEC, death, or neurodevelopmental
disability at 24 months in preterm infants [16,29,48,51]. Instead, growth rates similar to
intrauterine trajectories can be achieved by rapid increases in enteral feeding volume and by
achieving full enteral nutrition within 5–7 days of birth even in extremely low gestational
age neonates [48,49]. However, even if weight gain along intra-uterine trajectories is
achieved, very preterm infants still show insufficient lean body mass growth and an
increased fraction of body fat at term-equivalent age [52,53]. This can be interpreted as a
lack of essential nutrients required to achieve parenchymal and muscle mass growth as in
utero [54].
Particularly during the transition from intravenous to enteral nutrition, “increased”
gastric residues are detected during routine evaluation. The belief that increased gastric
residuals may be predictive of NEC frequently results in withholding or delaying enteral
feeding advancements [30]. Physiologically, however, 2–4 mL/kg of gastric residual fluid
is regularly aspirated just prior to any scheduled feeding [55]. Similarly, gastric residual
volume varies depending on patient position and feeding tube size and position, further
limiting the clinical usefulness of this practice [56]. A recent systematic review showed
insufficient evidence to support routine surveillance of gastric residuals with the intention
to prevent NEC [30]. In a subsequent randomized controlled trial involving 143 preterm
infants below 1250 g birth weight, no benefit was found for this practice [17]. Obviously, this
small study was under-powered to assess the effect of such a practice on NEC rates, but the
group that had no gastric residues determined showed an advanced enteral feeding pattern
and higher feeding volumes by week 5 [17]. Furthermore, discarding gastric residuals (e.g.,
those showing a dark green color) results in a loss of bile acids and phosphatidylcholine
from bile and digestive enzymes from the pancreas and enterocytes, all playing important
roles in intestinal homeostasis, regulation and digestion [57]. Thus, based on current
evidence, restricting monitoring of gastric residues to infants with symptoms of severe
Int. J. Environ. Res. Public Health 2021, 18, 7544 5 of 13

gastrointestinal dysfunction, such as emesis, abdominal tenderness, absent bowel sounds or


bloody stools likely improves enteral nutrient supply. However, the safety of this approach
remains to be proven.

4. Enteral Nutrition
Unquestionably, mothers’ own milk is the preferred source of nutrition for preterm
infants because of its numerous short- and longer-term health benefits, such as protection
against NEC, late-onset sepsis, and bronchopulmonary dysplasia (BPD), as well as im-
proved neurodevelopment [32,58]. Additionally, early oral administration of colostrum pro-
vides immunological components, probably stimulating the immune system and protecting
from inadequate bacterial colonization by lactoferrin, sIgA and other compounds [59,60].
Moreover, expressed breast milk contains high numbers of myeloid-derived suppressor
cells, which may prevent excessive inflammatory reactions and enhance tolerance to food
antigens [61,62].
However, mothers of preterm infants face a variety of barriers against breastfeeding.
Expressing breast milk approximately eight times/day during the first two weeks after
birth is the only proven way to increase the likelihood of achieving an adequate milk
supply of more than 500 mL per day, necessary for subsequent exclusive or predominant
breastfeeding. To achieve this, mothers must be encouraged to start manual and/or
mechanical breast milk expression shortly after birth [63,64]. In addition to the emotional
stress and concerns about the health of their baby [65], expressing breast milk 2–3 hourly,
as recommended, is time consuming and needs to be included in a tight daily routine with
kangarooing and, if present, caring for older siblings [66]. Appropriate prenatal counselling
and postnatal support for a variety of systems (e.g., double electric, bedside and free home
breast pumps for milk expression) by clinical staff in the NICU, peer support, skin-to-skin
care, staff education and a lactation consultant should therefore be common practice [67].
When mothers’ own milk is available in insufficient quantity or is contraindicated
(e.g., acquired immunodeficiency syndrome, chemotherapy), donor human milk (DHM)
is the adequate substitute for preterm infant feeding, as recommended by all relevant
societies [2,20,31–33]. In 2019, a systematic review of 12 studies, including 1879 infants
<2500 g birth weight, showed that formula feeding, compared to predominantly non-
supplemented DHM, resulted in improved weight gain, linear length and head growth,
indicating an inadequately low nutrient supply through non-supplemented DHM [33].
However, formula feeding also resulted in a higher risk of NEC (typical risk ratio (RR) 1.87,
95% CI 1.23 to 2.85) [33] and was associated with a lower quantity of breast- compared to
formula feeding at discharge [20]. DHM is not available in all NICUs and is considerably
more expensive than formula (e.g., $15/100 mL from a US not-for-profit Human Milk Bank
compared to $3/100 mL for preterm formula) [68]. By contrast, from a societal perspective,
the total cost of providing DHM to preterm infants is equal to formula feeding, due to
a reduced NEC rate [69]. In essence, it is important to reiterate that fresh mothers’ own
milk is the first choice for feeding preterm infants, and that great efforts should be made to
promote lactation, bridging the time to sufficient breast milk supply with (supplemented)
DHM [31].

5. Fortification
Human breast milk is optimally designed for term newborns and infants, who double
their weight within 4–6 months after birth. However, in line with the physiological
intrauterine growth rate during the 3rd trimester, very preterm infants double their weight
within 4–6 weeks. Thus, an increased supply of macro- and micronutrients is necessary for
adequate growth. Requirements for energy, protein, (essential) fatty acids, minerals such as
calcium and phosphate, as well as micronutrients like iron and vitamin D, to name a few,
are higher than in healthy newborns. For additional constitutive components, like choline,
increased requirements compared to current recommendations are debated as well [54].
All these nutrients are principally present in multi-nutrient human milk fortifiers (HMF),
Int. J. Environ. Res. Public Health 2021, 18, 7544 6 of 13

although their quantities are still controversial [54,70]. Hereby, intakes recommended
by ESPGHAN in 2010 (energy: 110–135 kcal/kg/d, protein: 4.0–4.5 g/kg/d (<1 kg) and
3.5–4.0 g/kg/d (1–1.8 kg), carbohydrate: 11.6–13.2 g/kg/d, fat: 4.8–6.6 g/kg/d) should
be achieved [2]. However, several recent studies indicate a ceiling effect for the beneficial
effect of protein intake on growth at approximately 4.5 g/kg/d [18,71].
Although breastmilk fortification is practiced in most NICUs [34], evidence for its
impact on long-term outcomes is remarkably sparse [72]. In a systematic review, multi-
nutrient fortification of human milk vs. non-fortified human milk showed increased
in-hospital growth rates for weight, head circumference and length without increasing
the risk of NEC [72]. However, the limited follow-up data for post-discharge growth and
neurodevelopment in later childhood show no benefit from fortification [72]. A recent
meta-analysis showed that early fortification starting at 20–40 mL/kg/d of enteral feeds
vs. late fortification (starting at 100 mL/kg/d) had little or no effect on short-term growth
outcomes [73].
However, as breast milk has no uniform nutrient content and marked inter- and
intra-individual variability exists [74], there are strategies for individualizing fortifica-
tion to match the nutritional needs of preterm infants [34]. Individual fortification is
performed by measuring the infant’s blood urea nitrogen (‘adjustable’ fortification) or the
macronutrient content of breast milk using a milk analyzer (‘targeted’ fortification). In a
recent review including 7 RCTs with a total of 521 participants, increased growth rates
for weight, length and head circumference were found with moderate to low evidence
following individualized compared to standard non-individualized fortification [35]. In
2021, a double-blind, randomized controlled trial was conducted in 103 preterm infants
<30 weeks comparing standard versus targeted fortification with modular proteins, lipids,
and carbohydrates [52]. The targeted fortification group had higher macronutrient intakes
and higher average growth velocity across the first 21 days of intervention (21.2 ± 2.5 vs.
19.3 ± 2.4 g/kg/d). Not surprisingly, infants born to mothers with a low breast milk protein
content showed the greatest benefit from targeted fortification regarding their weight at
36 weeks, length, head circumference, fat and fat-free mass [52]. Likewise, donor milk often
contains low levels of protein, suggesting targeted fortification to improve growth [75,76].
However, data on the clinical benefit of individual fortification by adjusting breast milk
macronutrients beyond short-term growth are sparse and inconclusive. A secondary analy-
sis of a randomized controlled trial indicated that ‘adapted’ protein supplementation, by
calculating breast milk protein content based on the duration of lactation, may be an easy
and inexpensive alternative to ‘targeted’ protein supplementation for achieving protein
supply on target in >95% of analyzed breast milk samples [77].
In recent years, discussions have addressed the question of whether multi-nutrient
fortifiers (HMF) derived from human rather than bovine milk may further reduce the
risk of NEC. However, the potential benefits of HMF derived from human milk have
been insufficiently investigated, especially in comparison with feeding regimens without
supplementary formula feeding, the latter already known to increase NEC rates. In 127
preterm infants <1250 g, an RCT using human vs. bovine milk-derived HMF in infants
fed human milk failed to improve feeding tolerance [78], but was underpowered to assess
effects on mortality and morbidity such as NEC. Moreover, concerns exist against the
commercialization of human milk, as the milk used to produce the fortifier is no longer
available as donor milk for very preterm infants. Other disadvantages are high cost,
unequal access to these products in different countries, and the fact that the large volume
of such liquid human milk-based HMF reduces the volume of expressed breast milk
administered to the infants by up to 1/3. Therefore, the use of human milk-based HMF is
currently not recommended by most committees and experts on pediatric nutrition [34,79].

6. Breast Milk–Acquired Cytomegalovirus Infection


Cytomegalovirus (CMV) reactivates in the lactating breast of up to 96% of CMV-
seropositive mothers, i.e., in approximately 50–80% of all mothers of preterm infants [19,80].
Int. J. Environ. Res. Public Health 2021, 18, 7544 7 of 13

It can cause severe sepsis-like symptoms with highly variable organ manifestations [80,81].
Postnatal transmission occurs in approximately 40% of infants <32 weeks’ gestation, with
higher transmission rates the lower gestational age is [19,80]. In most cases, at least one
of the following clinical signs is found: apnea and bradycardia, hepatosplenomegaly,
hepatitis, pneumonitis, intestinal distention and altered laboratory parameters (includ-
ing lymphocytopenia, neutropenia, thrombocytopenia, and elevated liver enzymes) [19].
Although most of these symptoms are self-limiting, CMV-related deaths have also been
reported [81]. In addition, an increased risk of developing BPD has been described in
cohorts of 385 extremely [82] and 2132 very low birth weight (VLBW) infants [83], as
well as in a recent study involving >100 000 VLBW infants [84]. NEC also seems to be
associated with postnatal CMV infection (pCMV). In numerous case reports and case series,
pCMV could be identified in gut specimens [85,86]. A recent study enrolling 596 VLBW
infants found an almost 3-times higher risk of NEC in CMV-positive than in CMV-negative
infants [87]. An actual multicenter study comprising 304 VLBW infants with postnatal
CMV observed significant associations with hearing and growth impairment, as well as
a prolonged hospitalization (by 12 days), but not with NEC [88]. Principally, all organ
systems, including the brain, can be involved in pCMV disease, as shown by autopsy
findings [89].
Some controversy exists whether early pCMV has negative consequences for neurocog-
nitive development [81,90,91]. In a recent cohort study involving 356 infants <32 weeks’
gestation [91], no negative impact on neurodevelopment until age six years was found
in a subgroup of 49 CMV-positive infants (14%). In a case-control study of 42 former
VLBW infants, pCMV positive infants had significantly lower test results at age six years
in the simultaneous processing scale of the Kaufman Assessment Battery for Children. In
a further follow-up study on 11–16 year old adolescents born at <32 wk GA (19 with, 23
without early pCMV infection), there was evidence of adverse effects of pCMV infection on
cognitive function [92]. This was supported by their functional magnetic resonance imag-
ing (MRI) results (n = 15) showing different activations in two brain regions for language
performance and differences in grey matter volume compared to children without pCMV
infection (n = 19) [93]. Intellectual deficits resulting from pCMV might be more obvious
in older children with more complex reasoning. These human data are supported by a
neonatal guinea pig model, where postnatally infected pups showed significant cognitive
deficits and brain anomalies compared to controls [94].
Notwithstanding the increasing knowledge about short- and long-term consequences
of pCMV infection, there is currently no consensus about preventive measures, but further
efforts seem justified. The Red Book Committee of the American Academy of Pediatrics
recommends serologic screening of mothers of infants born at <32 weeks and to consider
short-term breast milk pasteurization in those tested CMV-seropositive [36,37]. Others
recommend the pasteurization of breast milk from CMV-positive women in infants born
at <28 0/7 wk GA or with a birth weight <1000 g starting on day four until reaching 32
0/7 weeks post-menstrual age [95]. Further prospective studies are urgently needed.
For effectively eliminating CMV from breast milk, heat inactivation is required,
whereas freeze thawing is not sufficient. Holder pasteurization (63 ◦ C for 30 min) is
safe, but it reduces most of the nutritionally and immunologically relevant components in
human milk, such as immune cells, antibodies, enzymes, growth factors and hormones [96].
The authors’ institution therefore practices short-term heat inactivation (heating to 62 ◦ C
for 5 s) in their patients born at <32 weeks, as this sufficiently prevents CMV transmission
while preserving most benefits of breast milk [81,97].

7. Post-Discharge Nutrition
Achieving percentile-parallel growth using mothers’ own milk is the goal of post-
discharge nutrition [2]. Breastfeeding of preterm infants, starting with skin-to-skin contact
and non-nutritive sucking, should enable predominant breastfeeding at the time of dis-
charge. However, if weaning from the nasogastric tube is impossible, hospital discharge
Int. J. Environ. Res. Public Health 2021, 18, 7544 8 of 13

with tube feeding at home and adequate follow-up is the only feasible perspective [98]. In
Europe, this applies to approximately 40% of infants <32 weeks GA [99]. A recent meta-
analysis of 1251 preterm infants demonstrated that post-discharge formula feeding with
74 kcal/mL does not improve weight or head circumference growth compared to standard
term infant formula (≈67 kcal/100 mL) [100]. Limited evidence suggests that feeding
preterm infant formula (80 kcal/100 mL, usually in-house available only) compared to
standard term formula increases growth rates up to 18 months after birth (mean differences:
500 g weight, 10 mm length, 5 mm head circumference). No convincing evidence exists to
support discharging preterm infants with nutrient-fortified mothers’ milk [101]. Therefore,
the ESPGHAN guideline recommends individualized post-discharge nutrition adapted
to postnatal growth; i.e., for preterm infants with adequate weight gain until discharge,
fortified mothers’ milk or a special discharge formula is not required after discharge [2],
whereas infants who have grown less well initially should receive fortified breast milk
or a special post-discharge formula until at least three months corrected age [2]. This is
because of the common observation that former very preterm infants discharged home
shortly after discontinuation of naso-gastric tube top-up feeding experience a period of
inadequate weight gain [102], which is of unknown clinical importance but at least a major
burden for their parents.

8. Research Perspectives
Based on the concept that postnatal growth and body composition of preterm infants
should ideally mimic intra-uterine growth, and hence that postnatal nutrition should be
oriented at placental supply rather than breast milk composition (which is tailored to term
infants), we postulate that micronutrients that are actively transported to the fetus against a
concentration gradient must be of importance. As an example, fetal plasma concentrations
of choline, an essential nutrient for all age groups, are 3–4 times those of the parturient
throughout gestation, and very rapidly decrease by 50% or more after preterm birth [54,70].
It is also remarkable that the placenta enriches the fetus with docosahexaenoic acid (DHA)
and arachidonic acid (ARA), whereas linoleic acid (LA) is actively held back in the maternal
circulation. Based on current feeding regimens for preterm infants, the resulting fatty acid
profile of fetal lipoprotein phospholipids (high ARA, low LA, increasing DHA towards
term birth) is transformed to adult values (high LA, low ARA, low DHA) within one
week. Consequently, the preterm infant’s lipidome at term-corrected age is dramatically
different from that of term born infants in all compartments yet investigated, indicating
ARA and DHA deficiency and LA-overnutrition [103]. Addressing these and other nutrient
deficiencies and imbalances may help further to improve lean body mass growth and
long-term outcomes.

9. Conclusions
Preterm infants should be provided with all the macro- and micronutrients required to
achieve growth as in utero. To minimize side effects of parenteral nutrition, enteral feeding
should be started in the first days after birth, preferably based on supplemented mother’s
own milk or DHM. Further prospective studies are needed for many aspects of preterm
infant feeding.

Author Contributions: C.W. conceptualized, drafted the initial manuscript, and reviewed and
revised the manuscript. C.F.P., W.B., R.G., A.R.F. revising the article critically for important intellectual
content. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not Applicable.
Informed Consent Statement: Not Applicable.
Data Availability Statement: Not Applicable.
Int. J. Environ. Res. Public Health 2021, 18, 7544 9 of 13

Conflicts of Interest: The authors have no financial relationship relevant to this article to disclose.
The authors declare that they have no competing interests.

Abbreviations

ARA Arachidonic acid


BPD Bronchopulmonary dysplasia
CMV Cytomegalovirus
DHA Docosahexaenoic acid
DHM Donor human milk
ELBW Extremely low birth weight (<1000 g)
GA Gestational Age
HMF Human milk fortifiers
LA Linoleic acid
NEC Necrotizing enterocolitis
NICU Neonatal intensive care unit
pCMV Postnatal cytomegalovirus infection
PMA Postmenstrual age
VLBW Very low birth weight (<1500 g)
wk Week

References
1. Stocker, J.T.; Dehner, L.P.; Husain, A.N. Means and standard deviations of weights and measurements of lifeborn infants by body
weight (Appendix 28–29). In Stocker & Dehner’s Pediatric Pathology, 2nd ed.; Stocker, J.T., Dehner, L.P., Eds.; Lippinkott Williams &
Wilkins: Philadelphia, PA, USA, 2002.
2. Agostoni, C.; Buonocore, G.; Carnielli, V.P.; De Curtis, M.; Darmaun, D.; Decsi, T.; Domellöf, M.; Embleton, N.D.; Fusch, C.;
Genzel-Boroviczeny, O.; et al. Enteral Nutrient Supply for Preterm Infants: Commentary from the European Society of Paediatric
Gastroenterology, Hepatology and Nutrition Committee on Nutrition. J. Pediatr. Gastroenterol. Nutr. 2010, 50, 85–91. [CrossRef]
3. Johnson, M.J.; Wootton, S.A.; Leaf, A.A.; Jackson, A.A. Preterm Birth and Body Composition at Term Equivalent Age: A Systematic
Review and Meta-analysis. Pediatrics 2012, 130, e640–e649. [CrossRef]
4. Cerasani, J.; Ceroni, F.; De Cosmi, V.; Mazzocchi, A.; Morniroli, D.; Roggero, P.; Mosca, F.; Agostoni, C.; Giannì, M.L. Human Milk
Feeding and Preterm Infants’ Growth and Body Composition: A Literature Review. Nutrients 2020, 12, 1155. [CrossRef] [PubMed]
5. Stephens, B.E.; Walden, R.V.; Gargus, R.A.; Tucker, R.; McKinley, L.; Mance, M.; Nye, J.; Vohr, B.R. First-Week Protein and Energy
Intakes Are Associated With 18-Month Developmental Outcomes in Extremely Low Birth Weight Infants. Pediatrics 2009, 123,
1337–1343. [CrossRef] [PubMed]
6. Franz, A.R.; Pohlandt, F.; Bode, H.; Mihatsch, W.A.; Sander, S.; Kron, M.; Steinmacher, J. Intrauterine, Early Neonatal, and
Postdischarge Growth and Neurodevelopmental Outcome at 5.4 Years in Extremely Preterm Infants After Intensive Neonatal
Nutritional Support. Pediatrics 2009, 123, e101–e109. [CrossRef]
7. Ehrenkranz, R.A.; Dusick, A.M.; Vohr, B.R.; Wright, L.L.; Wrage, L.A.; Poole, W.K. Growth in the Neonatal Intensive Care Unit
Influences Neurodevelopmental and Growth Outcomes of Extremely Low Birth Weight Infants. Pediatrics 2006, 117, 1253–1261.
[CrossRef]
8. Martínez-Jiménez, M.D.; Gómez-García, F.J.; Gil-Campos, M.; Pérez-Navero, J.L. Comorbidities in childhood associated with
extrauterine growth restriction in preterm infants: A scoping review. Eur. J. Pediatr. 2020, 179, 1255–1265. [CrossRef] [PubMed]
9. Hovi, P.; Andersson, S.; Eriksson, J.; Järvenpää, A.-L.; Strang-Karlsson, S.; Mäkitie, O.; Kajantie, E. Glucose Regulation in Young
Adults with Very Low Birth Weight. N. Engl. J. Med. 2007, 356, 2053–2063. [CrossRef]
10. Belfort, M.B.; Martin, C.R.; Smith, V.C.; Gillman, M.W.; McCormick, M.C. Infant weight gain and school-age blood pressure and
cognition in former preterm infants. Pediatrics 2010, 125, e1419–e1426. [CrossRef]
11. Singhal, A.; Cole, T.J.; Lucas, A. Early nutrition in preterm infants and later blood pressure: Two cohorts after randomised trials.
Lancet 2001, 357, 413–419. [CrossRef]
12. Kerkhof, G.F.; Willemsen, R.H.; Leunissen, R.W.; Breukhoven, P.E.; Hokken-Koelega, A.C. Health Profile of Young Adults Born
Preterm: Negative Effects of Rapid Weight Gain in Early Life. J. Clin. Endocrinol. Metab. 2012, 97, 4498–4506. [CrossRef]
13. Valdez, R.; Athens, M.A.; Thompson, G.H.; Bradshaw, B.S.; Stern, M.P. Birthweight and adult health outcomes in a biethnic
population in the USA. Diabetologia 1994, 37, 624–631. [CrossRef] [PubMed]
14. Painter, R.C.; Osmond, C.; Gluckman, P.; Hanson, M.; Phillips, D.I.W.; Roseboom, T.J. Transgenerational effects of prenatal
exposure to the Dutch famine on neonatal adiposity and health in later life. BJOG Int. J. Obstet. Gynaecol. 2008, 115, 1243–1249.
[CrossRef] [PubMed]
15. Simon, L.; Frondas-Chauty, A.; Senterre, T.; Flamant, C.; Darmaun, D.; Rozé, J.-C. Determinants of body composition in preterm
infants at the time of hospital discharge. Am. J. Clin. Nutr. 2014, 100, 98–104. [CrossRef]
Int. J. Environ. Res. Public Health 2021, 18, 7544 10 of 13

16. Dorling, J.; Abbott, J.; Berrington, J.; Bosiak, B.; Bowler, U.; Boyle, E.; Embleton, N.; Hewer, O.; Johnson, S.; Juszczak, E.; et al.
Controlled Trial of Two Incremental Milk-Feeding Rates in Preterm Infants. N. Engl. J. Med. 2019, 381, 1434–1443. [CrossRef]
17. Parker, L.A.; Weaver, M.; Torrazza, R.J.M.; Shuster, J.; Li, N.; Krueger, C.; Neu, J. Effect of Gastric Residual Evaluation on Enteral
Intake in Extremely Preterm Infants. JAMA Pediatr. 2019, 173, 534–543. [CrossRef]
18. Maas, C.; Mathes, M.; Bleeker, C.; Vek, J.; Bernhard, W.; Wiechers, C.; Peter, A.; Poets, C.F.; Franz, A.R. Effect of Increased Enteral
Protein Intake on Growth in Human Milk–Fed Preterm Infants: A Randomized Clinical Trial. JAMA Pediatr. 2017, 171, 16–22.
[CrossRef] [PubMed]
19. Hamprecht, K.; Maschmann, J.; Vochem, M.; Dietz, K.; Speer, C.P.; Jahn, G. Epidemiology of transmission of cytomegalovirus
from mother to preterm infant by breastfeeding. Lancet 2001, 357, 513–518. [CrossRef]
20. Kantorowska, A.; Wei, J.C.; Cohen, R.S.; Lawrence, R.A.; Gould, J.B.; Lee, H.C. Impact of Donor Milk Availability on Breast Milk
Use and Necrotizing Enterocolitis Rates. Pediatr. 2016, 137, e20153123. [CrossRef] [PubMed]
21. Van Goudoever, J.; Carnielli, V.; Darmaun, D.; De Pipaon, M.S.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; Decsi, T.; Domellöf,
M.; et al. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Amino acids. Clin. Nutr. 2018, 37,
2315–2323. [CrossRef] [PubMed]
22. Lapillonne, A.; Mis, N.F.; Goulet, O.; van den Akker, C.H.V.D.; Wu, J.; Koletzko, B.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.;
et al. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Lipids. Clin. Nutr. 2018, 37, 2324–2336.
[CrossRef] [PubMed]
23. Mesotten, D.; Joosten, K.; van Kempen, A.; Verbruggen, S.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; Carnielli, V.; Darmaun,
D.; et al. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Carbohydrates. Clin. Nutr. 2018, 37,
2337–2343. [CrossRef] [PubMed]
24. Mihatsch, W.; Fewtrell, M.; Goulet, O.; Molgaard, C.; Picaud, J.-C.; Senterre, T.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; et al.
ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Calcium, phosphorus and magnesium. Clin.
Nutr. 2018, 37, 2360–2365. [CrossRef]
25. Bronsky, J.; Campoy, C.; Braegger, C.; Cai, W.; Carnielli, V.; Darmaun, D.; Decsi, T.; Domellöf, M.; Embleton, N.; Fewtrell, M.;
et al. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Vitamins. Clin. Nutr. 2018, 37, 2366–2378.
[CrossRef] [PubMed]
26. Domellöf, M.; Szitanyi, P.; Simchowitz, V.; Franz, A.; Mimouni, F.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; Carnielli, V.; et al.
ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Iron and trace minerals. Clin. Nutr. 2018, 37,
2354–2359. [CrossRef]
27. Joosten, K.; Embleton, N.; Yan, W.; Senterre, T.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; Carnielli, V.; Darmaun, D.;
et al. ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Energy. Clin. Nutr. 2018, 37, 2309–2314.
[CrossRef] [PubMed]
28. Riskin, A.; Picaud, J.-C.; Shamir, R.; Braegger, C.; Bronsky, J.; Cai, W.; Campoy, C.; Carnielli, V.; Darmaun, D.; Decsi, T.; et al.
ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition: Standard versus individualized parenteral
nutrition. Clin. Nutr. 2018, 37, 2409–2417. [CrossRef] [PubMed]
29. Oddie, S.J.; Young, L.; McGuire, W. Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low
birth weight infants. Cochrane Database Syst. Rev. 2017, 8, 001241. [CrossRef]
30. Abiramalatha, T.; Thanigainathan, S.; Ninan, B. Routine monitoring of gastric residual for prevention of necrotising enterocolitis
in preterm infants. Cochrane Database Syst. Rev. 2019, 7, CD012937. [CrossRef]
31. Arslanoglu, S.; Corpeleijn, W.; Moro, G.; Braegger, C.; Campoy, C.; Colomb, V.; Decsi, T.; Domellöf, M.; Fewtrell, M.; Hojsak, I.;
et al. Donor Human Milk for Preterm Infants. J. Pediatr. Gastroenterol. Nutr. 2013, 57, 535–542. [CrossRef]
32. Beidelman, A.I.; Schanler, R.J. Breastfeeding and the Use of Human Milk: Section on breastfeeding. Pediatrics 2012, 129, e827–e841.
[CrossRef]
33. Quigley, M.; Embleton, N.; McGuire, W. Formula versus donor breast milk for feeding preterm or low birth weight infants.
Cochrane Database Syst. Rev. 2019, 7, CD002971. [CrossRef] [PubMed]
34. Arslanoglu, S.; Boquien, C.-Y.; King, C.; Lamireau, D.; Tonetto, P.; Barnett, D.; Bertino, E.; Gaya, A.; Gebauer, C.; Grovslien, A.;
et al. Fortification of Human Milk for Preterm Infants: Update and Recommendations of the European Milk Bank Association
(EMBA) Working Group on Human Milk Fortification. Front. Pediatr. 2019, 7, 76. [CrossRef] [PubMed]
35. Fabrizio, V.; Trzaski, J.M.; Brownell, E.A.; Esposito, P.; Lainwala, S.; Lussier, M.M.; I. Hagadorn, J. Individualized versus standard
diet fortification for growth and development in preterm infants receiving human milk. Cochrane Database Syst. Rev. 2020, 11,
CD013465. [CrossRef] [PubMed]
36. Osterholm, E.A.; Schleiss, M.R. Impact of breast milk-acquired cytomegalovirus infection in premature infants: Pathogenesis,
prevention, and clinical consequences? Rev. Med. Virol. 2020, 30, 1–11. [CrossRef]
37. Kimberlin, D.W.B.M.; Jackson, M.A.; Long, S.S. American Academy of Pediatrics Human Milk. In Red Book: 2018 Report of the
Committee on Infectious Diseases, 31st ed.; American Academy of Pediatrics: Itasca, IL, USA, 2018; pp. 113–121.
38. Wilmore, D.W.; Dudrick, S.J. Growth and development of an infant receiving all nutrients exclusively by vein. JAMA 1968, 203,
860–864. [CrossRef]
39. Morgan, C.; McGowan, P.; Herwitker, S.; Hart, A.E.; Turner, M. Postnatal Head Growth in Preterm Infants: A Randomized
Controlled Parenteral Nutrition Study. Pediatrics 2013, 133, e120–e128. [CrossRef]
Int. J. Environ. Res. Public Health 2021, 18, 7544 11 of 13

40. Moon, K.; Athalye-Jape, G.K.; Rao, U.; Rao, S.C. Early versus late parenteral nutrition for critically ill term and late preterm
infants. Cochrane Database Syst. Rev. 2020, 2020, CD013141. [CrossRef]
41. Kapoor, V.; Malviya, M.N.; Soll, R. Lipid emulsions for parenterally fed term and late preterm infants. Cochrane Database Syst. Rev.
2019, 6, CD013171. [CrossRef]
42. Isaacs, E.B.; Gadian, D.G.; Sabatini, S.; Chong, W.K.; Quinn, B.T.; Fischl, B.R.; Lucas, A. The Effect of Early Human Diet on
Caudate Volumes and IQ. Pediatr. Res. 2008, 63, 308–314. [CrossRef]
43. Duerden, E.G.; PIANO study group; Thompson, B.; Poppe, T.; Alsweiler, J.; Gamble, G.; Jiang, Y.; Leung, M.; Tottman, A.C.;
Wouldes, T.; et al. Early protein intake predicts functional connectivity and neurocognition in preterm born children. Sci. Rep.
2021, 11, 4085. [CrossRef]
44. Bonsante, F.; Iacobelli, S.; Latorre, G.; Rigo, J.; De Felice, C.; Robillard, P.Y.; Gouyon, J.B. Initial Amino Acid Intake Influences
Phosphorus and Calcium Homeostasis in Preterm Infants—It Is Time to Change the Composition of the Early Parenteral Nutrition.
PLoS ONE 2013, 8, e72880. [CrossRef]
45. Siggers, J.; Østergaard, M.V.; Siggers, R.H.; Skovgaard, K.; Mølbak, L.; Thymann, T.; Schmidt, M.; Møller, H.K.; Purup, S.; Fink,
L.N.; et al. Postnatal amniotic fluid intake reduces gut inflammatory responses and necrotizing enterocolitis in preterm neonates.
Am. J. Physiol. Liver Physiol. 2013, 304, G864–G875. [CrossRef] [PubMed]
46. Mulvihill, S.; Stone, M.; Debas, H.; Fonkalsrud, E. The role of amniotic fluid in fetal nutrition. J. Pediatr. Surg. 1985, 20, 668–672.
[CrossRef]
47. Østergaard, M.V.; Bering, S.B.; Jensen, M.L.; Thymann, T.; Purup, S.; Diness, M.; Schmidt, M.; Sangild, P.T. Modulation of
Intestinal Inflammation by Minimal Enteral Nutrition With Amniotic Fluid in Preterm Pigs. J. Parenter. Enter. Nutr. 2013, 38,
576–586. [CrossRef]
48. Maas, C.; Mitt, S.; Full, A.; Arand, J.; Bernhard, W.; Poets, C.; Franz, A. A Historic Cohort Study on Accelerated Advancement of
Enteral Feeding Volumes in Very Premature Infants. Neonatology 2013, 103, 67–73. [CrossRef] [PubMed]
49. Maas, C.; Franz, A.; Von Krogh, S.; Arand, J.; Poets, C.F. Growth and morbidity of extremely preterm infants after early full
enteral nutrition. Arch. Dis. Child. Fetal Neonatal Ed. 2017, 103, F79–F81. [CrossRef]
50. Morgan, J.; Young, L.; McGuire, W. Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very
low birth weight infants. Cochrane Database Syst. Rev. 2014, 2014, CD001970. [CrossRef] [PubMed]
51. Maas, C.; Wiechers, C.; Bernhard, W.; Poets, C.F.; Franz, A.R. Early feeding of fortified breast milk and in-hospital-growth in very
premature infants: A retrospective cohort analysis. BMC Pediatr. 2013, 13, 178. [CrossRef]
52. Rochow, N.; Fusch, G.; Ali, A.; Bhatia, A.; So, H.Y.; Iskander, R.; Chessell, L.; el Helou, S.; Fusch, C. Individualized target
fortification of breast milk with protein, carbohydrates, and fat for preterm infants: A double-blind randomized controlled trial.
Clin. Nutr. 2021, 40, 54–63. [CrossRef]
53. Wiechers, C.; Doll, J.-N.; Maas, C.; Gründler, K.; Büchner, K.; Bevot, A.; Poets, C.F.; Franz, A.R. Körperfettanteil bei Frühgeborenen
zum Zeitpunkt der Entlassung und im Alter von korrigiert 4 Monaten. Monat.r Kinderh. 2019, 167, S53–S196.
54. Bernhard, W.; Poets, C.F.; Franz, A.R. Choline and choline-related nutrients in regular and preterm infant growth. Eur. J. Nutr.
2019, 58, 931–945. [CrossRef] [PubMed]
55. Mihatsch, W.A.; Von Schoenaich, P.; Fahnenstich, H.; Dehne, N.; Ebbecke, H.; Plath, C.; Von Stockhausen, H.-B.; Muche, R.; Franz,
A.; Pohlandt, F. The Significance of Gastric Residuals in the Early Enteral Feeding Advancement of Extremely Low Birth Weight
Infants. Pediatrics 2002, 109, 457–459. [CrossRef] [PubMed]
56. Chen, S.-S.; Tzeng, Y.-L.; Gau, B.-S.; Kuo, P.-C.; Chen, J.-Y. Effects of prone and supine positioning on gastric residuals in preterm
infants: A time series with cross-over study. Int. J. Nurs. Stud. 2013, 50, 1459–1467. [CrossRef]
57. Gayatri, A.-J.; Megan, N.; Ching-Tat, L.; Elizabeth, N.; Donna, G.; Karen, S.; Sanjay, P. Composition of Coloured Gastric Residuals
in Extremely Preterm Infants-A Nested Prospective Observational Study. Nutrients 2020, 12, 2585. [CrossRef]
58. Victora, C.G.; Bahl, R.; Barros, A.J.D.; França, G.V.A.; Horton, S.; Krasevec, J.; Murch, S.; Sankar, M.J.; Walker, N.; Rollins, N.C.;
et al. Breastfeeding in the 21st century: Epidemiology, mechanisms, and lifelong effect. Lancet 2016, 387, 475–490. [CrossRef]
59. Maffei, D.; Brewer, M.; Codipilly, C.; Weinberger, B.; Schanler, R.J. Early oral colostrum administration in preterm infants. J.
Perinatol. 2019, 40, 284–287. [CrossRef]
60. Nasuf, A.W.A.; Ojha, S.; Dorling, J. Oropharyngeal colostrum in preventing mortality and morbidity in preterm infants. Cochrane
Database Syst. Rev. 2018, 9, CD011921. [CrossRef]
61. Köstlin-Gille, N.; Flaig, L.-A.; Ginzel, M.; Arand, J.; Poets, C.F.; Gille, C. Granulocytic Myeloid-Derived Suppressor Cells in Breast
Milk (BM-MDSC) Correlate with Gestational Age and Postnatal Age and Are Influenced by Infant’s Sex. Nutrients 2020, 12, 2571.
[CrossRef]
62. Köstlin-Gille, N.; Gille, C. Myeloid-Derived Suppressor Cells in Pregnancy and the Neonatal Period. Front. Immunol. 2020, 11,
584712. [CrossRef]
63. Parker, L.A.; Sullivan, S.; Krueger, C.; Kelechi, T.; Mueller, M. Effect of early breast milk expression on milk volume and timing of
lactogenesis stage II among mothers of very low birth weight infants: A pilot study. J. Perinatol. 2011, 32, 205–209. [CrossRef]
64. Parker, L.A.; Sullivan, S.; Kruger, C.; Mueller, M. Timing of milk expression following delivery in mothers delivering preterm
very low birth weight infants: A randomized trial. J. Perinatol. 2020, 40, 1–10. [CrossRef] [PubMed]
65. Sisk, P.M.; Lovelady, C.A.; Dillard, R.G.; Gruber, K. Lactation Counseling for Mothers of Very Low Birth Weight Infants: Effect on
Maternal Anxiety and Infant Intake of Human Milk. Pediatrics 2006, 117, e67–e75. [CrossRef] [PubMed]
Int. J. Environ. Res. Public Health 2021, 18, 7544 12 of 13

66. Hurst, N.; Engebretson, J.; Mahoney, J.S. Providing Mother’s Own Milk in the Context of the NICU. J. Hum. Lact. 2013, 29,
366–373. [CrossRef]
67. Renfrew, M.J.; Dyson, L.; McCormick, F.; Misso, K.; Stenhouse, E.; King, S.E.; Williams, A.F. Breastfeeding promotion for infants
in neonatal units: A systematic review. Child Care Health Dev. 2010, 36, 165–178. [CrossRef]
68. Jegier, B.J.; Johnson, T.J.; Engstrom, J.L.; Patel, A.; Loera, F.; Meier, P. The Institutional Cost of Acquiring 100 mL of Human Milk
for Very Low Birth Weight Infants in the Neonatal Intensive Care Unit. J. Hum. Lact. 2013, 29, 390–399. [CrossRef]
69. Trang, S.; Zupancic, J.A.; Unger, S.; Kiss, A.; Bando, N.; Wong, S.; Gibbins, S.; O’Connor, D. Cost-Effectiveness of Supplemental
Donor Milk Versus Formula for Very Low Birth Weight Infants. Pediatrics 2018, 141, e20170737. [CrossRef] [PubMed]
70. Shunova, A.; Böckmann, K.A.; Minarski, M.; Franz, A.R.; Wiechers, C.; Poets, C.F.; Bernhard, W. Choline Content of Term and
Preterm Infant Formulae Compared to Expressed Breast Milk—How Do We Justify the Discrepancies? Nutrients 2020, 12, 3815.
[CrossRef] [PubMed]
71. Reid, J.; Makrides, M.; McPhee, A.J.; Stark, M.J.; Miller, J.; Collins, C.T. The Effect of Increasing the Protein Content of Human
Milk Fortifier to 1.8 g/100 mL on Growth in Preterm Infants: A Randomised Controlled Trial. Nutrients 2018, 10, 634. [CrossRef]
72. Brown, J.V.E.; Embleton, N.; Harding, J.E.; McGuire, W. Multi-nutrient fortification of human milk for preterm infants. Cochrane
Database Syst. Rev. 2016, CD000343. [CrossRef]
73. Thanigainathan, S.; Abiramalatha, T. Early fortification of human milk versus late fortification to promote growth in preterm
infants. Cochrane Database Syst. Rev. 2020, 7, CD013392. [CrossRef]
74. Weber, A.; Loui, A.; Jochum, F.; Buhrer, C.; Obladen, M. Breast milk from mothers of very low birthweight infants: Variability in
fat and protein content. Acta Paediatr. 2001, 90, 772–775. [CrossRef]
75. Colaizy, T.T.; Carlson, S.; Saftlas, A.F.; Morriss, F.H., Jr. Growth in VLBW infants fed predominantly fortified maternal and donor
human milk diets: A retrospective cohort study. BMC Pediatr. 2012, 12, 124. [CrossRef]
76. Colaizy, T.T. Donor human milk for very low birth weights. Curr. Opin. Pediatr. 2015, 27, 172–176. [CrossRef] [PubMed]
77. Minarski, M.; Maas, C.; Engel, C.; Heinrich, C.; Böckmann, K.; Bernhard, W.; Poets, C.F.; Franz, A.R. Calculating Protein Content of
Expressed Breast Milk to Optimize Protein Supplementation in Very Low Birth Weight Infants with Minimal Effort—A Secondary
Analysis. Nutrients 2020, 12, 1231. [CrossRef] [PubMed]
78. O’Connor, D.L.; Kiss, A.; Tomlinson, C.; Bando, N.; Bayliss, A.; Campbell, D.M.; Daneman, A.; Francis, J.; Kotsopoulos, K.; Shah,
P.S.; et al. Nutrient enrichment of human milk with human and bovine milk–based fortifiers for infants born weighing. Curr. Dev.
Nutr. 2019, 3, nz129.
79. Ernährungskommissionen der Deutschen Gesellschaft für Kinder- und Jugendmedizin e. V. (DGKJ); Österreichischen Gesellschaft
für Kinder- und Jugendheilkunde e. V. (ÖGKJ). Kommerzielle Muttermilchverstärker aus humaner Milch: Unzureichend belegter
Nutzen und hohe Kosten. Monatsschrift Kinderheilkd. 2018, 167, 145–148. [CrossRef]
80. Martins-Celini, F.P.; Yamamoto, A.Y.; Passos, D.M.; Nascimento, S.D.D.; Lima, E.V.; Di Giovanni, C.M.; Quadrado, E.R.S.; Barta,
R.; Aragon, D.C.; Prado, S.I.D.; et al. Incidence, Risk Factors, and Morbidity of Acquired Postnatal Cytomegalovirus Infection
Among Preterm Infants Fed Maternal Milk in a Highly Seropositive Population. Clin. Infect. Dis. 2016, 63, 929–936. [CrossRef]
81. Hamprecht, K.; Goelz, R. Postnatal Cytomegalovirus Infection Through Human Milk in Preterm Infants. Clin. Perinatol. 2017, 44,
121–130. [CrossRef]
82. Yoo, H.S.; Sung, S.I.; Jung, Y.J.; Lee, M.S.; Han, Y.M.; Ahn, S.Y.; Chang, Y.S.; Park, W.S. Prevention of Cytomegalovirus Transmission
via Breast Milk in Extremely Low Birth Weight Infants. Yonsei Med. J. 2015, 56, 998–1006. [CrossRef]
83. Mukhopadhyay, S.; Meyer, S.A.; Permar, S.R.; Puopolo, K.M. Symptomatic Postnatal Cytomegalovirus Testing among Very
Low-Birth-Weight Infants: Indications and Outcomes. Am. J. Perinatol. 2016, 33, 894–902. [CrossRef]
84. Kelly, M.S.; Benjamin, D.K.; Puopolo, K.M.; Laughon, M.M.; Clark, R.; Mukhopadhyay, S.; Smith, P.B.; Permar, S.R. Postnatal
Cytomegalovirus Infection and the Risk for Bronchopulmonary Dysplasia. JAMA Pediatr. 2015, 169, e153785. [CrossRef]
85. Goelz, R.; Hamprecht, K.; Klingel, K.; Poets, C.F. Intestinal manifestations of postnatal and congenital cytomegalovirus infection
in term and preterm infants. J. Clin. Virol. 2016, 83, 29–36. [CrossRef]
86. Omarsdottir, S.; Agnarsdottir, M.; Casper, C.; Orrego, A.; Vanpée, M.; Rahbar, A.; Söderberg-Nauclér, C. High prevalence of
cytomegalovirus infection in surgical intestinal specimens from infants with necrotizing enterocolitis and spontaneous intestinal
perforation: A retrospective observational study. J. Clin. Virol. 2017, 93, 57–64. [CrossRef]
87. Patel, R.M.; Shenvi, N.; Knezevic, A.; Hinkes, M.; Bugg, G.W.; Stowell, S.R.; Roback, J.D.; Easley, K.; Josephson, C. Observational
study of cytomegalovirus from breast milk and necrotising enterocolitis. Arch. Dis. Child. Fetal Neonatal Ed. 2019, 105, 259–265.
[CrossRef]
88. Weimer, K.E.D.; Kelly, M.S.; Permar, S.R.; Clark, R.; Greenberg, R.G. Association of Adverse Hearing, Growth, and Discharge Age
Outcomes With Postnatal Cytomegalovirus Infection in Infants With Very Low Birth Weight. JAMA Pediatr. 2020, 174, 133–140.
[CrossRef]
89. Lopes, A.-A.; Champion, V.; Mitanchez, D. Nutrition of Preterm Infants and Raw Breast Milk-Acquired Cytomegalovirus
Infection: French National Audit of Clinical Practices and Diagnostic Approach. Nutrients 2018, 10, 1119. [CrossRef]
90. Lanzieri, T.M.; Dollard, S.C.; Josephson, C.D.; Schmid, D.S.; Bialek, S.R. Breast Milk-Acquired Cytomegalovirus Infection and
Disease in VLBW and Premature Infants. Pediatrics 2013, 131, e1937–e1945. [CrossRef]
Int. J. Environ. Res. Public Health 2021, 18, 7544 13 of 13

91. Gunkel, J.; De Vries, L.S.; Jongmans, M.; Koopman-Esseboom, C.; Van Haastert, I.C.; Eijsermans, M.C.J.; Van Stam, C.; Van Zanten,
B.G.A.; Wolfs, T.F.W.; Nijman, J. Outcome of Preterm Infants With Postnatal Cytomegalovirus Infection. Pediatrics 2018, 141,
e20170635. [CrossRef]
92. Brecht, K.F.; Goelz, R.; Bevot, A.; Krägeloh-Mann, I.; Wilke, M.; Lidzba, K. Postnatal Human Cytomegalovirus Infection in
Preterm Infants Has Long-Term Neuropsychological Sequelae. J. Pediatr. 2015, 166, 834–839.e1. [CrossRef]
93. Dorn, M.; Lidzba, K.; Bevot, A.; Goelz, R.; Wilke, M.; Hauser, T.-K. Long-term neurobiological consequences of early postnatal
hCMV-infection in former preterms. Hum. Brain Mapp. 2013, 35, 2594–2606. [CrossRef]
94. Fernández-Alarcón, C.; Meyer, L.E.; McVoy, M.A.; Lokensgard, J.R.; Hu, S.; Benneyworth, M.A.; Anderholm, K.M.; Janus, B.C.;
Schleiss, M.R. Impairment in neurocognitive function following experimental neonatal guinea pig cytomegalovirus infection.
Pediatr. Res. 2021, 89, 838–845. [CrossRef]
95. Ernährungskommission der Österreichischen Gesellschaft für Kinder- und Jugendheilkunde; Haiden, N.; Wald, M.; Berger, A.
Prävention von CMV-Infektionen bei Frühgeborenen. Monatsschrift Kinderheilknd. 2019, 167, 323–328. [CrossRef]
96. Goelz, R.; Hihn, E.; Hamprecht, K.; Dietz, K.; Jahn, G.; Poets, C.; Elmlinger, M. Effects of Different CMV-Heat-Inactivation-Methods
on Growth Factors in Human Breast Milk. Pediatr. Res. 2009, 65, 458–461. [CrossRef]
97. Bapistella, S.; Hamprecht, K.; Thomas, W.; Speer, C.P.; Dietz, K.; Maschmann, J.; Poets, C.F.; Goelz, R. Short-term Pasteurization
of Breast Milk to Prevent Postnatal Cytomegalovirus Transmission in Very Preterm Infants. Clin. Infect. Dis. 2018, 69, 438–444.
[CrossRef]
98. Ahnfeldt, A.M.; Stanchev, H.; Jørgensen, H.L.; Greisen, G. Age and weight at final discharge from an early discharge programme
for stable but tube-fed preterm infants. Acta Paediatr. 2015, 104, 377–383. [CrossRef]
99. Wilson, E.; Bonamy, A.-K.E.; Bonet, M.; Toome, L.; Rodrigues, C.E.D.; Howell, E.A.; Cuttini, M.; Zeitlin, J.; The EPICE Research
Group. Room for improvement in breast milk feeding after very preterm birth in Europe: Results from the EPICE cohort. Matern.
Child Nutr. 2017, 14, e12485. [CrossRef]
100. Young, L.; Embleton, N.D.; McGuire, W. Nutrient-enriched formula versus standard formula for preterm infants following
hospital discharge. Cochrane Database Syst. Rev. 2016, 2016, CD004696. [CrossRef]
101. Young, L.; Embleton, N.; McCormick, F.M.; McGuire, W. Multinutrient fortification of human breast milk for preterm infants
following hospital discharge. Cochrane Database Syst. Rev. 2013, CD004866. [CrossRef]
102. Wiechers, C.; Doll, J.-N.; Maas, C.; Gründler, K.; Büchner, K.; Poets, C.F.; Franz, A.R. Enteral feeding advancement and extremely
preterm infants’ growth until 5 years. BMC Pediatr. 2021, in press.
103. Böckmann, K.A.; von Stumpff, A.; Bernhard, W.; Shunova, A.; Minarski, M.; Frische, B.; Warmann, S.; Schleicher, E.; Poets, C.F.;
Franz, A.R. Fatty acid composition of adipose tissue at term indicates deficiency of arachidonic and docosahexaenoic acid and
excessive linoleic acid supply in preterm infants. Eur. J. Nutr. 2021, 60, 861–872. [CrossRef] [PubMed]
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

doi: 10.7499/j.issn.1008-8830.2017.01.001

专家讲座

Optimizing nutrition of the preterm infant


William W. Hay, Jr.
(Perinatal Research Center, Child Maternal Health Program, Colorado Clinical and Translational Sciences Institute
University of Colorado School of Medicine, Aurora, Colorado, USA)

Abstract: Objective The goal of nutrition of the preterm infant is to meet the growth rate of the healthy fetus
of the same gestational age and to produce the same body composition of the healthy fetus in terms of organ growth,
tissue components, and cell number and structure. Nutritional quantity and quality are fundamental for normal growth
and development of preterm infants, including neurodevelopmental outcomes. Failure to provide the necessary amounts
of all of the essential nutrients has produced not only growth failure, but also increased morbidity and less than optimal
neurodevelopment. Growth velocities during the NICU hospitalization period for preterm infants exert a significant effect
on neurodevelopmental and anthropometric outcomes. Despite the obvious need for optimal nutrition, growth failure is
almost universal among preterm infants. There is every reason, therefore, to optimize nutrition of the preterm infant, in
terms of total energy and protein, but also in terms of individual components such as amino acids, specific carbohydrates
and lipids, and even oxygen. This review presents scientific rationale for nutrient requirements and practical guidelines
and approaches to intravenous and enteral feeding for preterm infants. Intravenous feeding, including amino acids,
should be started right after birth at rates that are appropriate for the gestational age of the infant. Enteral feeding should
be started as soon as possible after birth, using mother’s colostrum and milk as first choices. Enteral feeding should
begin with trophic amounts and advanced as rapidly as tolerated, decreasing IV nutrition accordingly, while maintaining
nutrient intakes at recommended rates. Feeding protocols are valuable for improving nutrition and related outcomes.
Further research is needed to determine the optimal nutrition and rate of growth in preterm infants that will achieve
optimal neurocognitive benefits while minimizing the longer-term risk of chronic diseases.
[Chin J Contemp Pediatr, 2017, 19(1): 1-21]
Key words: Intravenous feeding; Parenteral feeding; Enteral feeding; Amino acid; Glucose; Lipid; Necrotizing
enterocolitis; Preterm infant

Introduction repair in response to injuries (such as oxygen toxicity


and barotrauma in the lung, inflammation in the gut,
The goal of nutrition of the preterm infant is to and wound healing), and general underdevelopment of
meet the growth rate of the healthy fetus of the same all other organs such as intestine[5-6], skeletal muscle[7],
gestational age[1]. It also is important to produce the and brain[8-9].
same body composition of the healthy fetus in terms of As documented by many studies over many
organ growth, tissue components, and cell number and years, neuronal underdevelopment may be the most
structure. Failure to provide the necessary amounts serious adverse consequence of under nutrition of
of all of the essential nutrients has produced not only preterm infants, which clearly leads to later life
[2]
growth failure , but also increased vulnerability to cognitive deficiencies[10-13]. The most severe adverse
infectious diseases arising from suboptimal immune neurodevelopmental outcomes occur in preterm
[3]
defenses , increased respiratory distress from lung infants who had IUGR and also suffered postnatal
[4]
injury due to muscle weakness , impaired tissue under nutrition. While it remains uncertain whether

[Received] August 26, 2016;[Accepted] October 15, 2016


[Biography] William W. Hay, Jr., MD, Professor of Pediatrics (Neonatology)/Scientific Director, Perinatal Research Center/Director, Child
Maternal Health Program, Colorado Clinical and Translational Sciences Institute University of Colorado School of Medicine (Email: bill.
hay@ucdenver.edu).

1
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

improved neuronal development, brain growth, outcomes. Growth velocities during the NICU
and cognitive outcomes are the direct result of hospitalization period for preterm infants exert a
improved nutrition or whether healthier infants can significant, and possibly independent, effect on
[14]
be fed more , studies clearly are documenting the neurodevelopmental and anthropometric outcomes[13].
association of improved neurodevelopmental outcome As shown by Lucas and colleagues [32], 4 weeks of
with improved nutrition and growth of preterm enriched formula with greater amounts of energy as
[15-17]
infants . well as protein fed to preterm infants improved growth
Despite such needs, it remains a challenge to and neurodevelopmental outcomes at 18 months
consistently provide adequate nutrition to preterm corrected gestational age when compared to preterm
infants, particularly right after birth when nutritional infants fed lower amounts of energy and protein
support is largely dependent on intravenous nutrition in standard term formulas. At 7.5 years of age, the
and often is complicated by glucose and lipid infants fed the enriched preterm formula demonstrated
[18-19]
intolerance . During this period, large nutritional higher cognitive functions[33], and even at age 16,
deficits, in particular protein deficits, can rapidly these infants had larger brain size and larger volumes
accumulate in preterm infants. In such infants, protein of the caudate nucleus measured using MRI F:\
stores can decline by as much as 1.5%/day, in contrast Documents\Neonatal Nutrition, for Bo Sun, Chinese
to the normally growing fetus that has a positive Journal of Comtemporary Pediatrics\Nutritional_
[20]
net protein balance of ~2%/day . Many studies support_for_extremely_low-birth_weight_infants_
from around the world have provided consistent abandoning_cat.html - 134 and higher intelligence
evidence that early amino acid intake, at least quotient (IQ) scores [16,34]. Regression analysis of
3 g/kg/day, before 5 days of life, can reduce protein infants from several studies have shown a significant
breakdown and deficits and improve protein balance positive correlation between both cumulative energy
and growth outcomes in preterm, very-low-birth- and protein intakes and greater head circumference
[21-27]
weight infants . This is one of the most consistent at 36 weeks postmenstrual age [35], F:\Documents\
observations among controlled research trials and Neonatal Nutrition, for Bo Sun, Chinese Journal of
[19]
observational studies in neonatal medicine . Comtemporary Pediatrics\Nutritional_support_for_
Nevertheless, growth failure is almost universal extremely_low-birth_weight_infants_abandoning_
[28]
among preterm infants . 80% of very low-birth cat.html - 136and better mental and psychomotor
weight preterm infants involved in studies conducted developmental index (MDI and PDI) scores at 3
by the NIH Neonatal Research Network from 2008 months' corrected age [36] . Furthermore, a recent
to 2010 had documented growth failure [29-30], and retrospective study showed that the MDI of ELBW
among infants within the Vermont Oxford Hospital infants at age 1.5 years increased by 8.2 and
Network, at least 50% were growth-restricted at the 4.6 points, respectively, for every extra gram of
[2]
time of discharge from the NICU . Such growth protein/kg per day or every extra 10 kcal/kg per day
failure is, however, preventable, and so is the they received during the first week of life[37]. This early
neurodevelopmental impairment that comes from postnatal period appeared to be critical, as nutritional
insufficient nutrition. Protein deficiency is critical, in intakes during the next few weeks of life did not show
this regard, as protein is the nutritional component correlations with neurocognitive outcome. While
most essential for development of neurological continued nutritional support during the NICU period,
[31]
function , but overall nutritional quantity and as well as after discharge, is fundamental, clearly,
quality also are fundamental for normal growth the first few days after birth are most important for
and development, including neurodevelopmental achieving positive protein and energy balance and for

2
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

promoting improved growth and development. Most studies also show, however, that most
There is every reason, therefore, to optimize preterm infants fail to grow well after birth, usually for
nutrition of the preterm infant, in terms of total energy many days. They also don’t keep up with intrauterine
and protein, but also in terms of individual components growth and thus end up growth restricted by term[41-42]
such as amino acids, specific carbohydrates and lipids, (Figure 2). This problem has not improved over the past
and even oxygen. If this is the case, how successful almost 70 years[43].
is current nutritional practice for preterm infants in
terms of producing optimal nutrition and growth 2 000

and development of preterm infants? The answer is,


universally, not as well as it should. It is imperative,
1 500

Weight (g)
therefore, to reassess the rate of growth that should be
achieved in preterm infants and the basic nutritional
1 000
requirements needed to achieve this growth rate. Intrauterine growth
24-25 weeks
26-27 weeks
28-29 weeks
How fast should a preterm infant grow? 500

24 28 32 36

Postmenstrual age (weeks)


Direct measurement of normal human fetal
growth is not possible. Most commonly, therefore, Figure 2 Postnal growth of VLBW infants vs.
fetal growth rates have been estimated from expected intrauterine growth[42]

anthropometric measurements at the time of birth


among “healthy”, normal appearing preterm infants While there are many possible reasons why
of viable gestational age through to term. Using preterm infants do not grow after birth as well as the
this approach, multiple studies have shown that the normal fetus of the same gestational age, including
average fractional fetal weight gain from 24 to 38 stress responses (e.g., increased secretion and
weeks gestation is 17 g/kg/d[38-40] (Figure 1). plasma concentrations of catabolic hormones such
catecholamines, glucagon, and cortisol, intermittent
4 000 hypoxia, respiratory distress, mechanical ventilation,
sepsis, inadequate body temperature maintenance,
3 500
Olsen, 2010, Mean boys
and girls
etc.), a principal cause is insufficient nutrition, as
3 000 Fenton (boys, 2013)
evidenced by cumulative deficits in energy and
Weight (g)

Fenton (girls, 2013)


2 500 Denver
protein, even with more “aggressive” nutrition that
Baltimore
2 000
Montreal provides closer to estimated nutrient requirements[44]
1 500 Portland
(Figure 3) .
Chapel Hill
1 000 12 U.S. Cities

25 30 35 40 45

Gestational age (weeks)

Figure 1 Birth weights by gestaional age[38-40]


Regardless of the growth curve, normal human fetal growth rate is
~17 g/kg/day from 28-40 weeks.

3
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

Intake (Kcal/K) 200 10

Intake (G/K)
100 5
0 0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 1 3 5 7 9 11 13 15 17 19 21 23 25 27
-100 -5
Aggressive group
-200 -10
Conventional group Aggressive group
-300
-15 Conventional group
Deficit (Kcal/K)

-400
-20

Deficit (G/K)
-500
-25
-600
-30
-700
-35
Postnatal days Postnatal days

A principal reason-Inadequate Nutrition as evidenced by cumulative deficits in energy and protein, even with more "aggressive" nutrition.

Figure 3 Energy and protein intake and cumulative deficits in the first 28 postnatal days[44]

Why are preterm VLBW infants not fed enough that of the normally nourished and normally growing
to grow as well as the fetus? There are several human fetus of the same gestational age.
practical and common reasons. First, most preterm
infants have a delayed start to receiving nutrients, Nutrient requirements for preterm infants
e.g., low rates of or even no IV amino acids on the
first day of life (to sometimes several days after If we are going to feed preterm infants to grow
birth). Second, enteral feedings often are withheld, as well as the normal, healthy human fetus, we should
sometimes for days. Third, most infants receive very consider the normal fetal nutrient and co-factor
slow advances of nutrient supply, e.g., IV amino acid requirements and at least provide those. There may
infusion rates of <3 g/kg/d. Fourth, there are usually be additional adjustments for conditions that arise
rather slow advances of IV amino acids and lipids after birth, but it is unreasonable to consider nutrition
after starting, and slow advances of enteral feeds. sufficient if it provides less than what the normal fetus
Fifth, most infants receive dilute nutritional mixes, receives. What nutrients and co-factors, therefore,
e.g., unfortified breast milk (mother’s own or banked), should we provide to the preterm infant at the same
and insufficient amounts of essential amino acids in gestational age of the fetus in utero?
TPN mixes. There also are many excuses offered as Oxygen
“reasons” for withholding, slowing down, or stopping   First, while not actually a nutrient, oxygen is
feedings, practically none of which has a rational basis necessary for the metabolism of all nutrients and
from controlled trials. For the most part, it is simply particularly for producing growth. Numerous studies
assumed that infants with any kind of physiological have shown that hypoxia and oxygen consumption
instability cannot tolerate normal amounts of nutrition, rates less than normal restrict growth. The normal
either parenteral or enteral (Table 1). As a result, fetus exposed to hypoxia responds by increasing
preterm infants commonly are under nourished from erythropoietin, producing more red blood cells,
birth and often for days to sometimes weeks, and all thereby increasing its blood oxygen content to
of the practical reasons and excuses, justified or not, compensate for the reduction in molecular oxygen
reduce nutrient intake, which leads to growth failure supply[45]. Also, many studies have shown that preterm
and impaired neurodevelopment. It is not unexpected, infants who receive blood transfusions for severe
therefore, that growth, which for the fetal period of anemia not only increase their blood oxygen content
human development requires constant supplies of but also improve their growth rate. In one study, for
nutrients, does not occur or even decreases relative to example, preterm infants did not gain weight as well

4
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

Table 1 “Excuses” for withholding, slowing down, or stopping feeding of preterm infants, with comments about why this
    might be done and why it might not be rational
Item Excuse
1. abdominal distension fear of NEC
2. green gastric aspirates fear of NEC
3. UA and/or UV catheters fear of gut ischemia, and thus NEC
4. GER fear of apnea(actually, it probably is the other way around)
5. tachypnea fear of aspiration
6. heart murmurs fear of PDA and gut ischemia—and NEC
7. PDAs fear of gut ischemia—and NEC
8. indomethacin fear of gut ischemia—and NEC
9. high BUN fear of urea poisoning and amino acid toxicity
10. high bilirubin fear of FFA displacing bilirubin from albumin
11. high WBC/CRP fear of decreased metabolism, proteolysis
12. skin rashes fear of allergies
fear of poor metabolism (though stopping IV lipids and reducing
13. hyperglycemia
dextrose infusion rates at least have a rational basis)
14. hypothermia fear of sepsis
15. hyperthermia fear of sepsis
16. hypo- or hyperkalemia poor gut function, bad IV nutrient mix
17. hypo- or hypernatremia bad IV nutrient mix, dehydrated
18. thrombotic episodes need to use heparin, reduce IV rate
19. polycythemia risk of clots and gut ischemia and NEC
20. SpO2 values are low can’t metabolize nutrients
21. On catecholamines fear of gut ischemia, NEC, hyperglycemia
22. Anemia fear of gut ischemia
23. Transfusions risks of NEC, transfusion related immunomodulation
how does this reduce digestion and nutrient absorption and
24. on a ventilator
anabolism?
this one baffles me; of course it’s low if the baby is under-
25. low energy expenditure
nourished!
26. might need surgery so they should be starved first?
27. “Just doesn’t look good”— I have no response to this one!
28. Mother couldn’t be here so we held the feeding for her to give good for the mother, but bad for the baby!
29. Intermittent hypoxic episodes these get better with starvation??
30. We wanted “fasting” electrolyte values in the morning What does fasting have to do with electrolytes?
31. We wanted to be sure the baby was stable well, sure, and starved, too
always a scapegoat around—the ubiquitous, infamous “other
32. “The other attending” doesn’t like to advance feeds very fast
attending”
And more!

when their Hb concentration was ≤ 8.5 g/dL (Hct lower 20 s) to limit transfusion risks (transfusion-
≤ 25%), but their weight gain improved after related inflammatory disorders, e.g., NEC, strokes,
transfusion with red blood cells to a blood Hb lung injury) and to be maintained at lower PaO 2
concentration of ≥ 11.4 g/dL (Hct ≥ 34%). The more values to prevent oxygen toxicity (e.g., ROP,
severe the anemia (lower the blood oxygen content), BPD). Furthermore, preterm infants lose, by adult
therefore, the more likely that growth failure will standards, enormous amounts of blood for all sorts
[46]
develop . Commonly, however, current practice of biochemical and hematological measurements.
usually reduces blood oxygen content by allowing Unfortunately, we have little capacity to measure
preterm infants to be more anemic (hematocrits in the clinical effect of low O 2 supply on nutrient

5
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

metabolism (energy + protein), protein accretion, and the gastrointestinal tract, that do not use glucose
and neuronal growth and development, limiting our as much as the brain and heart. Regardless of the
ability to assess the impact, in any given infant, of utilization or production rates, preterm infant plasma
different low blood oxygen contents on producing glucose concentrations should be in the same range
growth restriction. Our goal, therefore, should be to as normal human fetuses of the same gestational age,
determine optimal nutrient and anabolic hormone i.e., between 3.0 mmol/L and 6 mmol/L[52]. This is the
supplies to promote growth, especially of neurons/ same range achieved by normal, healthy term newborn
dendrites, at lower blood O 2 levels. We also might infants after the first 12-24 hours from birth following
improve our administration of erythropoietin (give the normal nadir in glucose concentration from 1-3
more, or develop more effective protocols), provide hours of age[53].
safer transfusions, use delayed cord clamping and cord Lipids
stripping routinely, reduce blood sampling, microsize    Normal human fetal development involves
blood sample assays, and provide more and better considerable fat deposition in adipose tissue, primarily
noninvasive monitoring. peripherally in subcutaneous regions of the body.
Glucose Normal human fetuses produce about 15 g/kg body
   Adequate glucose also must be provided to weight as fat by term. Early in the third trimester
ensure appropriate growth rates. Glucose is essential (24-28 weeks), however, there is little lipid uptake,
for normal metabolism in all cells, but as the principal oxidation, or accretion as fat by the fetus. Lipid supply
energy substrate of the normal fetus and thus the and fat accretion increase progressively over the
preterm infant, glucose is necessary to support growth, third trimester. It remains uncertain how much lipid
particularly protein synthesis from amino acids and net is oxidized in the human fetus, but likely not very
protein balance. In fetal studies, for example, reduction much, as the concentrations of carnitine palmitoyl
in glucose supply and plasma glucose concentrations transferase, the enzyme that transports long chain fatty
[47]
lead to growth failure . This is a relatively uncommon acids into the mitochondria for lipid oxidation, are
problem in preterm infants, however, as most of the quite low, and glucose and amino acid supplies are
time, they are infused with more glucose than they more than sufficient to meet most energy requirements.
can use, resulting in hyperglycemia. Normal neonatal After birth, however, even in very preterm infants and
glucose production and utilization rates have been even quite soon after birth, supplemental carnitine
[48]
estimated from animal studies and measured in (e.g., in mother’s milk) promotes lipid oxidation.
[49-50]
infants with stable isotope techniques . Preterm    Extremely preterm infants often are slow to
infants at ~28 weeks gestation maintain hepatic clear plasma of lipid due to maturational deficiency
glucose production rates of 2-3 mg/min/kg which are of lipases (inversely related to GA), low levels of
not easily suppressed, either by glucose or insulin, as carnitine palmitoyltransferase (CPT), and chronically
[51]
they are in normal children . Glucose utilization rates reduced carnitine intakes >2 weeks after birth with
can be as high as 7-9 mg/min/kg, however, largely prolonged IV nutrition[54]. There also is competition
for the brain, but also for the heart. Term infants of free fatty acids for oxidation due to early postnatal
also produce ~2-3 mg/min/kg of glucose, but their hyperglycemia. As a result of such conditions, many
glucose utilization rates are lower at ~3-5 mg/min/kg recommend limiting lipid intake to 0.5-1 g/kg/day
(still largely for the brain). The decrease in weight- in presence of other disorders that limit fatty acid
specific glucose utilization rates from very preterm to oxidation, such as sepsis, severe lung disease, surgical
term gestational age is the result of increasing body stress, steroid use, persistent hyperglycemia, and
proportions of organs, such as bone, muscle, lung, cholestasis. This practice is poorly justified, however,

6
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

with little evidence for mechanisms involved and at least this one essential fatty acid, but the long term
how to manipulate them, other than added carnitine to significance of this deficiency is not known or how
promote better fatty acid oxidation. these infants would develop if fed to sufficiency.
   The quality of fatty acids and triglycerides Energy
[55]
and other lipid products also is important . Little    In terms of total energy (primarily from
information exists, however, about the actual mix of carbohydrates and lipids), there is a persistent but
such substances in fetal plasma that might provide unsubstantiated concern that higher intake rates of
guidance for the optimal mix of plasma lipid substances amino acids and protein won’t produce more growth
after birth. A major concern, however, is how to unless higher calorie intakes also are provided. This
provide sufficient long chain polyunsaturated essential concern is based on the unjustified fixation on a
fatty acids that are fundamental components of the continuous 22 kcal/g protein ratio. Actually, excessive
[56]
developing nervous system . Normal fetal white caloric intake simply increases body fat content
adipose tissue accumulation in the 3rd trimester of the relative to lean mass. Above 80-90 kcal/kg/day non-
essential omega 3 long chain polyunsaturated fatty acids protein caloric intake, there is no further increase in
(PUFAs), particularly docosahexaenoic acid (DHA, net protein balance for any further increase in energy
22:6n-3) ranges from ~45 to ~70 mg/day. According to intake. Protein gain primarily depends on protein
current practice, at 3.7g fat/dL human milk with 0.2- intake, regardless of the energy intake [60]. This has
0.4% fatty acids as 22:6n-3, a 1 kg preterm infant fed been corroborated by elegant nutritional balance
at full enteral feeds of 180 mL/day would get only 13- studies by Kashyap and collegues[61] (Figure 4), who
25 mg 22:6n-3/day, clearly far below normal in utero showed that while greater protein intakes would
accretion rates. As shown by Lapillone and colleagues, increase weight, length, and head circumference,
therefore, postnatal DHA deficiency is an inevitable additional lipid with the greater protein intakes only
consequence of current recommendations and practices increased weight and subcutaneous fat.
for feeding milk, milk supplements, and formulas in
preterm infants[57]. In terms of potential benefits of 30 1.6 1.6 1.2 group 1
25 1.4 1.4 1.0 group 2
providing greater amounts of DHA, one recent study 20
1.2 1.2
0.8
group 3
g/kg/day

1.0 1.0
cm/wk

cm/wk

cm/wk

has shown that higher DHA and lower linoleic acid 15 0.8 0.8 0.6
0.6 0.6
levels in the first few weeks of life are associated with 10
0.4 0.4
0.4
5 0.2 0.2 0.2
decreased intraventricular hemorrhage, improved 0 0.0 0.0 0.0
weight length head triceps
microstructural brain development, and improved circumference skinfold

outcomes in preterm infants[58]. Several other studies


Figure 4 Growth rates with varying protein and
also have shown that preterm infants fed increased energy intakes [61] Preterm infants, birth weight 900-1 750 g:
DHA have higher visual acuity, particularly at 2 and 4 Group 1, 2.24 g/kg/day and 115 kcal/kg/day; Group 2, 3.6 g/kg/day and
months and improved Bayley mental development and 115 kcal/kg/day; Group 3, 3.5 g/kg/day and 149 kcal/kg/day.
*
P<0.05: weight in group 1 less than in group 2 and group 3;
MacArthur Communicative inventories at 12 months[59],
head circumference in group 1 less than in group 2 and group 3; triceps
but longer term studies do not show a clear benefit. skinfold greater in group 3 than in group 1 and group 2.
Thus, the current diet for preterm infants is deficient in

7
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

  Despite such evidence, recent observations of Amino acids and protein


actual intake of nutrients in clinical neonatal practice   The most important requirement for growth is
has been weighted to energy, both carbohydrates protein intake, as amino acids intravenously and as
and lipids, and not to protein, leading to problems intact to partially or fully hydrolyzed protein enterally.
of carbohydrate and lipid excess and protein Lean body mass, which consists mostly of protein,
[62]
insufficiency . Such a diet mix leads to fatter, accounts for 90% of growth in the 3rd trimester
shorter, less muscular infants, and perhaps to longer of gestation. Without sufficient protein, cellular
term neurological deficits. Unfortunately, this problem replication, hypertrophy, and development cannot take
is most common among the smallest, most preterm place as optimally as they should, and insufficient
infants who are fed far more energy and much less protein intake will adversely affect proliferation and
protein than needed to meet appropriate growth rate growth of all cells in all organs (Table 2) [28].
and body composition[63].

Table 2 Feeding guidelines: intake amounts (mL/kg/day), by day of feeding (DOF)

Gestational age
DOF 1 DOF 2 DOF 3 DOF 4 DOF 5 DOF 6 DOF 7 DOF 8 DOF 9 DOF 10
(weeks)
< 26 ≤ 20 ≤ 20 ≤ 20 ≤ 20 ≤ 50 ≤ 80 ≤ 110 ≤ 140 ≤ 160 160-180
(fortify
26-29 ≤ 20 ≤ 20 ≤ 20 ≤ 50 ≤ 80 ≤ 110 ≤ 140 160-180
(fortify
29-32 ≤ 20 ≤ 20 ≤ 50 ≤ 80 ≤ 110 ≤ 140 160-180
(fortify
32-34 ≤ 20 ≤ 50 ≤ 80 ≤ 110 ≤ 140 160-180
(fortify
34-37 ≤ 40 ≤ 80 ≤ 120 ≤ 140 160-180
(fortify
Caveats
• Withhold feeds in infants with defined intestinal obstruction or ileus.
• Asphyxia, respiratory distress, sepsis, hypotension, glucose disturbances, ventilation, and umbilical catheters are not contraindications for
trophic feeds.
• Infants at higher risk (two or more risk factors) may have feeding amounts reduced or initiated at GI priming volumes with advancement as
medically appropriate.
• Infants may nipple bottle feeds up to the maximum feeding volume, or breastfeed as able. Infants should not initially be placed on ad lib
nippling or advanced to higher volumes, due to increased risk for feeding intolerance.
• Breast milk is preferred. Donor milk also is acceptable, but will need fortification even more than mother’s own breast milk.
• Human Milk Fortifiers (HMFs) should contain hydrolyzed protein if based on cow milk protein, or use “human” HMFs.
Risk factors for “more conservative” (but not stopping) advancement of feeding
1) Non-reassuring fetal heart rate tracing with need for resuscitation of the infant after delivery and evidence of asphyxia (severe and prolonged
metabolic acidosis from persistent hypotension).
2) More severe and asymmetric IUGR or IUGR with reversed or absent end-diastolic flow on fetal Doppler ultrasound.
3) Monochorionic twin gestation with twin-twin transfusion syndrome.
4) Marked polycythemia (Hct >65).
5) Significant cardiovascular instability: need for chest compressions, use of vasoactive agents, or need for aggressive volume expansion to
achieve sufficient circulation.
6) Marked and prolonged and recurrent apnea.
7) Symptomatic patent ductus arteriosus with reduced urine flow rate and progressive metabolic acidosis.
8) Clinically significant (cardiovascular compromise) sepsis.
9) Prolonged (> 7 days) absence of enteral feedings.

8
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

  The reason for this need for protein is that protein nutrition appears to be a beneficial clinical strategy[67],
synthesis and accretion rates are very high in the fetus the optimal amount of that support is not fully
(net protein balance of ~2%/day) and therefore the known[19], including the time after birth to start, the
preterm infant of the same gestational age, requiring doses to use, and the rate of advancement[68]. The
large amino acid uptake rates. Fetal animal growth Current recommendations are that IV nutrition should
data, when scaled to human fetal growth rate, predict be initiated within a few hours of birth[28].
fetal amino acid requirements ≈ 3.6-4.8 g/kg/day Amino acids
at ~24-28 weeks of human gestation. This animal   At least 2.0 g/kg/day of amino acids should be
data has been corroborated for human fetuses by started within 2-3 hours of birth, and increased to 3.5-
[64]
the Factorial Method , which predicts that human 4.0 g/kg/day over the first 24-48 h for infants <30
fetal amino acid requirements ≈ 4 g/kg/day at 24-28 weeks gestation (up to 4.0 g/kg/day for extremely low
weeks of gestation. It is important to note, however, birth weight infants <27 weeks gestation).
that fractional protein synthesis rates decrease with    Even when solutions are infused at these or
gestational age and development. Thus, between 30 yet higher rates, intakes of certain essential amino
and 37 weeks, the protein requirement for growth acids, particularly the branched-chain amino acids
decreases to ~ 2-3 g/kg/day and by term, protein such as leucine and isoleucine[69], but also threonine
requirements decrease to those of the normal breast and lysine[70], may not produce high enough plasma
fed infant, or ~ 1.5-2 g/kg/day. Among many studies concentrations to promote the rate of cellular essential
in preterm infants, net protein balance as measured by amino acid uptake needed to produce appropriate
net nitrogen retention is directly and linearly related protein accretion. There has been some concern for
[19,65]
to protein intake . Specific studies demonstrate the excessively high plasma amino acid concentrations
same linear relationship between IV amino acid intake at infusion rates >3.0-4.0 g/kg/day, particularly in
[22,66]
and protein balance through 3 to 4 g/kg/day . sick and physiologically unstable preterm infants[71].
Also, infants who experienced severe and chronic
Practical feeding guidelines: intravenous intrauterine growth restriction may have developed
feeding adaptations that actually limit amino acid synthesis
into protein [72]. There is little evidence, however,
   The goal of early intravenous feeding is to that most preterm infants will have significant
maintain normal cellular energy and amino acid complications from the amino acid infusion rates
supplies. Metabolic and thus nutritional requirements noted as necessary to produce in utero protein
do not stop with birth, and the smaller the infant, the accretion and growth rates. In fact, most studies have
less body stores of nutrients are available to provide noted that current IV amino acid mixtures and infusion
nutrients for metabolic needs. Intravenous feeding, rates produce lower plasma concentrations than are
therefore, is always indicated when normal metabolic needed for optimal amino acid metabolism and protein
and nutritional needs are not met by normal enteral balance[22,69-70]. The overall impact of such temporarily
feeding. Furthermore, the metabolic and nutrient insufficient amounts of essential and total amino acids
requirements of the newborn preterm infant are on growth and neurodevelopment of preterm VLBW
equal to or greater than those of the fetus of the same infants is uncertain, but it is quite likely that even
gestational age. It is reasonable, therefore, to provide temporary shortfalls limit growth.
the preterm infant with at least the same nutrient Glucose
supplies that the fetus of the same gestational age    Glucose (as 10% Dextrose in water) should
receives for nutrition. Although early intravenous be infused as soon as possible after birth to prevent

9
· ·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

hypoglycemia and at the highest rate tolerated without during IV nutritional support. In addition to the most
causing hyperglycemia. common cause of hyperglycemia, which is excessive
  Hypoglycemia, defined arbitrarily as a plasma dextrose infusion, hyperglycemia also is the result
glucose concentration <45 mg/dL in the 1st 2 days of persistent hepatic glucose production[50] that is not
after birth and <50-60 mg/dL after 2 days of life, down-regulated by increasing plasma concentrations
should be avoided as much as possible. While low of glucose or insulin[51]. Hyperglycemia also is caused
glucose concentrations do occur even among preterm by catecholamine treatment of low blood pressure,
infants receiving IV dextrose infusions or TPN, there hydrocortisone treatment of low blood pressure and
is no evidence that occasional transiently low values evolving chronic lung disease, and high IV lipid
in the 40-50 mg/dL range are a problem. infusion rates.
  Hyperglycemia, defined arbitrarily as a plasma    Mixed hyper- and hypoglycemia occur quite
glucose concentration >120 mg/dL at any time commonly in very preterm, IUGR infants, but should
after birth, is a more common glucose metabolic be considered in any very preterm infant, as many
[73]
disorder than hypoglycemia in preterm infants , of these infants experienced IUGR before their
largely the result of excessive dextrose infusion rates preterm birth. Hyperglycemia is common in such
(>6-8 mg/kg/min) in the first hours to days after birth infants due to reduced pancreatic β-cell number and
when stress reactive hormones that independently insulin production, hepatic insulin resistance and
contribute to glucose production, such as increased glucose production, decreased glucose
[74]
norepinephrine and cortisol, are commonly present . disposal capacity because of their smaller brains and
Hyperglycemia increases energy expenditure reduced muscle, and higher secretion rates and plasma
(glucose-to-fat synthesis is energy expensive), oxygen concentrations of catecholamines and cortisol that
consumption (leading to hypoxia), carbon dioxide limit insulin production and action. Hypoglycemia
production (leading to tachypnea), fat deposition in is common in such infants due to their greater head/
excess of lean mass, and fatty infiltration of heart brain to body/liver ratio and thus greater body weight-
and liver. Hyperglycemia in preterm infants also specific glucose utilization rate, increased peripheral
is a potentially serious cause of cellular and even tissue glucose uptake capacity from increased or at
systemic inflammation from excess reactive oxygen least maintained tissue glucose transporters, increased
[75-76]
species production . Such effects may underlie fractional insulin secretion and susceptibility to
the increased risk of retinopathy of prematurity metabolic stimulation of insulin secretion, and good
[77-79]
noted among infants with hyperglycemia . As health and care that reduces catecholamine and cortisol
noted in one recent study, at every time point in the secretion and concentrations and their negative impact
first 12 hours after birth, hyperglycemia produces on insulin secretion and action[82].
more unfavorable outcomes in infants with hypoxia-   Prevention and treatment of hyperglycemia is
[80]
ischemia . There also is the potential for sustained important and is best accomplished by using lower
hyperglycemia in preterm infants to diminish longer IV dextrose infusion rates in the first place and
term growth and even cognitive development, while lowering them, along with reducing lipid infusion
also more positively diminishing body fat content. rates and catecholamine and cortisol treatments and
Down-regulation of the growth IGF-1/growth preventing hypoxic episodes. Use of insulin should
[81]
hormone axis may be responsible . be reserved for the most serious hyperglycemic
   While hyperglycemia usually occurs in the conditions (plasma glucose concentrations >200-
first few days after birth, particularly among the 250 mg/dL and unresponsive to other approaches).
most immature infants, it can be found at any time There are many complications of using insulin to

10·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

prevent or treat hyperglycemia. Insulin actually and liver failure in infants who experience prolonged
makes the baby fatter (including fatty infiltration Intravenous nutrition [88]. ELBW infants often are
of heart and liver). Insulin increases complications slow to clear plasma of lipid due to maturational
of excess allostatic load (reactive oxygen species deficiency of lipases (inversely related to GA), low
production and inflammation). Insulin increases the levels of carnitine palmitoyltransferase (CPT) and
[83]
risk of hypoglycemia . Furthermore, although not chronically reduced carnitine intakes >2 weeks after
studied yet in preterm infants, in children with cardiac birth, and competition of FFA oxidation with carbon
disorders in intensive care, tight glucose control did from hyperglycemia. In response, many recommend
not improve outcomes, but did result in more cases limiting lipid infusion rates to 0.5-1 g/kg/day in
of hypoglycemia, and those children had poorer the first few days of life, particularly in presence of
[84]
neurodevelopmental outcomes . Infused insulin does infection, severe lung disease, surgical stress, and
not promote glucose uptake or utilization by the brain steroid use. There is little rational justification for this
or enhance neuronal growth or dendritic development. practice, except when hypertriglyceridemia occurs
Not the least, negative feedback mechanisms limit in the presence of hyperglycemia, and to prevent
the effect of insulin to promote protein synthesis, cholestasis. Some have added carnitine to promote
net protein balance, and growth; insulin just does better fatty acid oxidation, but possible benefits only
not work as a growth hormone to augment growth appear after total IV nutrition of more than 2 weeks[54].
when given in excess. When insulin infusion was   It is clear, however, that postnatal deficiencies
compared to reduced IV glucose infusion rate to treat of essential fatty acids, particularly the long
[85-86]
hyperglycemia in 500-750 g infants , there were c h a i n p o l y u n s a t u r a t e d f a t t y a c i d ( L C P U FA )
no differences in all age/weight groups for rates of docosahexaenoic acid or DHA is an inevitable
death, sepsis, ROP, NEC, intracranial hemorrhage, consequence of current recommendations and
chronic lung disease, days in the NICU, or rates of practices of intravenous lipid feeding, as well as for
growth. It makes more sense to just limit the glucose feeding milk, milk supplements, and formulas in
infusion rate, and perhaps to increase IV amino acid preterm infants, leading to increasing cumulative
infusion rates to 4 g/kg/day, which has been shown to DHA deficits. Unfortunately, to date, long term
reduce the number of episodes of hyperglycemia and neurodevelopmental benefits of DHA or omega-3
[87]
the time-averaged plasma glucose concentration . LCPUFA supplementation are equivocal and shorter
Lipids term improvements in BPD, ROP, NEC, sepsis, and
   Intravenous lipid infusion should be started death are uncertain.
within 24 hours of birth and advanced from 2.0 Total energy: Intravenous nutrition should
to 3.0-3.5 g/kg/day as tolerated, usually over 2-3 achieve a caloric intake of 50 kcal/kg/day on day 1
days. Intravenous lipids provide essential carbon for and 80 kcal/kg/day by day 3.
oxidative metabolism and also provide essential fatty Amino acids: Intravenous amino acid infusion
acids that promote membrane formation, particularly should be started right after birth, at least at 2.0 g/kg per
in the brain, but also in all cells in the body. Excess day, and advanced to gestational age specific protein
lipid infusion rates, however, commonly produce needs by day 2 to 3.
hypertriglyceridemia (arbitrarily defined as plasma   Between 24-30 weeks, initial intravenous amino
triglyceride concentrations >150 mg/dL), which has acid infusion rates should range from 3.5 to 4.0 g/kg/
been associated with adverse lipid deposition and day. Between 30 and 36 weeks, as fractional protein
[62,76]
subsequent inflammation in hepatocytes , that may synthesis rates decline, intravenous amino acid
underlie the common problem of cholestatic jaundice infusion rates of 3.0 to 3.5 g/kg/day are appropriate.

·11·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

At term, amino acid requirements decrease to those for longer periods, and advancing enteral nutrition
of the normal breast fed infant, or 1.5-2.0 g/kg/day. and milk fortification sooner and at faster rates[94].
While some have questioned whether the early, high Fortification also should be started before full enteral
amino acid infusion rate in very preterm infants feeding rates are achieved, perhaps at 50 mL/kg/day[92].
might lead to potential adverse outcomes, previous A recent study in 100 VLBW infants randomized
observational and randomized controlled trials have to early fortification of enteral feeding at a feeding
not found smaller head circumferences in response volume of 20 mL/kg/day or delayed fortification at a
[23-27,89]
to higher amino acid intakes , and one recent feeding volume of 100 mL/kg/day (all infants were
randomized controlled trial showed that higher amino fed according to a standardized enteral and parenteral
[27]
acid intakes resulted in greater head circumferences . feeding protocol; fortification was with Mead Johnson
Even small differences in head circumference are Nutrition acidified human milk fortifier) reported that
unlikely to demonstrate developmentally important the infants randomized to early fortification had a
differences in brain volumes when early, higher higher median daily protein intake at 1, 2 and 3 weeks
amino acid infusion rates are used, and the studies of age as well as a higher cumulative protein intake
addressing this issue actually have shown that the head in the first 4 weeks of life. There was no difference
circumferences measured are just above or just below in days to reach full feeding volumes, episodes of
the 50th percentile for head circumference at 40 weeks feeding intolerance or necrotizing enterocolitis[95].
corrected gestational age[90], representing improved
head circumference percentiles from those measured at Practical feeding guidelines: enteral feeding
birth[19]. Furthermore, even at 3.5 g/kg/day (if actually
achieved), there may be inadequate essential amino    There are many adverse consequences of
acids to produce plasma concentrations sufficient to intravenous feeding only with no enteral nutrition.
achieve appropriate cellular amino acid uptake rates Problems of exclusive intravenous feeding include
[69-70,89,91]
and, thus, rates of protein synthesis . increased rates of sepsis (both due to suppressed
immunity, reduced milk-associated immune function,
Practical feeding guidelines: transition from and abnormal gut microbiome, as well as central
intravenous to enteral feeding line associated bacterial infections or CLABSI),
gut mucosal atrophy and lack of production of gut-
  During the transition from parenteral to enteral derived trophic hormones, increased adipose tissue
nutrition, a shortfall of nutrient intakes may lead to and fatty infiltration of the heart and liver, systemic
[92]
insufficient protein and energy intakes . This problem inflammatory response syndrome, reduced mucosal
arises when parenteral nutrition is diminished or IgA from unstimulated and under developed Payer’s
terminated early in order to limit the use of indwelling patches, and increased adhesion molecules and
catheters because of the associated risk of infection polymorphonuclear cell attraction. Animal models
(central line associated blood stream infection) and such as the neonatal piglet have shown reduced
to limit total water intake and its potential to produce accretion of lean mass and bone mineral content as
metabolic (dilutional acidosis, hyponatremia) and well as increased fat mass when neonatal piglets
[93]
cardiopulmonary (PDAs) disorders . As noted by were fed TPN exclusively vs. enteral nutrition over
[28]
Hay and Ziegler , practical solutions to this problem a 16-day period[74]. Studies in adults[96-98] and older
include temporarily increasing the total and especially infants and children including term infants[99] have
the essential amino acid concentrations in parenteral shown greater rates of various morbidities and even
nutrition solutions, continuing parenteral nutrition mortality when enteral nutrition is withheld and

12·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

intravenous nutrition is used exclusively. Some of Z-score changes (birth to hospital discharge) by
these problems most likely are the result of excessive year of study as human milk (mother’s and donor)
glucose infusion with hyperglycemia, even when feeding increased [108] . For infants <28-30 weeks
insulin is added as part of “tight glucose control” to gestation, to provide about 4.0 g/kg/day of protein,
[100]
maintain normal glycemia . For older infants, as the amount needed to duplicate fetal growth, a protein
in children and adults, delaying intravenous nutrition concentration of ≥2.8 g/100 mL of milk must be
while optimizing very early enteral nutrition appears reached. While mother’s milk primarily, but also
to be safer, with fewer cases of infection and fewer donor milk (secondarily), are the recommended enteral
days to discharge.99 Starting enteral nutrition as feeding products for newborn infants, their nutrient
soon as possible, therefore, is beneficial, particularly concentrations generally are too low, particularly of
when combined with appropriate early intravenous protein, to meet the growth requirements of preterm
[101-102]
nutrition . infants. This cannot be reached with a powder fortifier;
   Of particular note is the improvement in gut thus, protein intakes remain below adequate intake
development and growth using “trophic” or “gut levels with use of powder fortifiers. Liquid fortifiers
priming” enteral feeds. Benefits include improved add greater amounts of protein (around 1.7 g of protein
mucosal growth and development that clearly depend per 100 mL of milk) and satisfactory protein intakes are
on enteral (not parenteral) nutrients, improved feeding achieved most of the time. The customary formulation
tolerance and growth, less need for phototherapy, to do this involves adding 1 mL of liquid protein
decreased cholestasis, decreased osteopenia, improved fortifier per 25 mL of milk, which will increase protein
gastrointestinal trophic hormone surges, and improved intake by 1 g/kg/day at 150 mL/kg/day of enteral
GI motility, all with no increase in complications (e.g., feeds[109]. This approach increases protein gain and
NEC, particularly with colostrum and milk)[103-104]. It growth, as nearly all milk but particularly donor milk
also is important to note that enteral feeding increases contains insufficient amounts of protein, especially
[105]
superior mesenteric artery blood flow , which may as lactation matures[110-111]. While a systematic review
account for the lack of a causal role for PDAs or early did not find significant increases in the rate of NEC
[106]
indomethacin treatment to lead to early NEC .A infants whose mother’s milk was supplemented with
recent retrospective cohort study from a single NICU cow milk derived protein fortifiers[112], recent studies
evaluated 415 infants over a 5-year period who were have documented lower rates of NEC when human
treated with indomethacin for PDA closure. The milk derived protein fortifiers were used[113].
infants were divided into three groups based on enteral
feed volume during treatment: those whose feeds were Summary and recommendation
held (n=229), those whose feeds were ≤60 mL/kg/day
(n=142), and those whose feeds were >60 mL/kg/day    Growth outcomes of ELBW infants remain
(n=44). This study found no significant difference in suboptimal, because they are not fed enough,
incidence of NEC (≥ Bell stage IIa) and noted that a especially of protein. Early protein losses are
preemptive reduction in enteral feeds was associated minimized by providing 3-4 g/kg/day of amino acids;
[107]
with a significantly longer time to reach full feeds . less amino acids and protein could lead to neurological
  As with intravenous nutrition, enteral nutrient deficits. In general, preterm infants have been fed
regimens also must provide sufficient protein to excessive energy, which only makes them fatter;
meet the growth rates of the normally growing fetus but they still lack essential fatty acids, particularly
of the same gestational age. Several studies have the LC PUFAs, particularly DHA. Providing at
noted declining growth and head circumference least ~70 (intravenous) to 90 (enteral) non-protein

13·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

kcal/kg/day and 3-4 g/kg/day of amino acids or protein of nutrition during NICU hospitalization and after
may approximate fetal protein accretion and growth discharge on neurodevelopment or the risk of
in reasonably healthy ELBW infants. Late preterm or developing later life disorders such as hypertension,
moderately preterm infants also receive suboptimal obesity, or insulin resistance[115]. Further research is
nutrition and do not grow optimally for the same needed to determine the optimal nutrition and rate of
reasons—insufficient protein and suboptimal amounts growth in preterm infants that will achieve optimal
of mother’s milk with insufficient fortification. neurocognitive benefits while minimizing the longer-
Research also is needed to determine optimal amino term risk of chronic diseases.
acid and energy supplies in sick infants and those who
have experienced significant intrauterine and postnatal Practical recommendations
growth restriction.
  Only a few pre-discharge intervention studies   Normal fetal nutrition is a reasonable guide to the
to promote growth in preterm infants during their nutrient requirements for preterm infants of the same
NICU stay have information on later neurocognitive, gestational age. Several aspects of fetal nutrition are
adiposity, and insulin resistance/cardiovascular risk important to recognize. First, amino acids are pumped
factor outcomes. Abundant and consistent evidence into the fetus by active transport in the placenta at
from observational studies link faster postnatal rates that are higher than the fetus can use for protein
growth to better neurocognitive outcomes in preterm synthesis and net protein accretion. The excess amino
infants, but these studies show no obvious windows of acid load is oxidized for energy. Secondly, glucose
association and there is a high risk of confounding by is taken up and used to meet energy needs according
other factors/disease processes that affect both growth to the metabolic rate of the fetus, which does not
and cognition. Where associations are reported in increase significantly with excess glucose. Excess
observational studies linking faster postnatal growth to glucose not only causes hypoxia and acidosis, but
adverse cardiovascular risk markers in preterm infants, also leads to various forms of toxicity that impair
the findings have often not been adjusted for body size insulin secretion and induce cellular and systemic
at the time of the outcome measurements. Moreover, inflammation. Fetal nutrition, therefore, is designed
comparisons were often not made to the normal for growth and to maintain energy balance. In contrast
ranges of these outcome parameters in unselected “customary” ELBW/VLBW Nutrition is aimed at an
populations. Present evidence suggests that even brief excess of energy and insufficient amounts of protein.
periods of relative undernutrition during a sensitive Glucose is pumped into the infant at rates that are
period of development have significant adverse effects higher than the infant can use. The excess glucose load
[114]
on later development . produces hyperglycemia, which leads to inflammation
  It also is worth noting that there still is much yet and cellular toxicity. Furthermore, amino acids are
to learn about how to optimally feed preterm infants. provided at rates that are less than needed for normal
The balance of studies, for example, have shown that growth rates.
growth between birth and expected term and 12-18    To more appropriately support growth of the
months post-term has little or no significant effect on preterm infant to meet the growth rates and body
later blood pressure and metabolic syndrome, whereas compositions of the normal fetus of the same
reduced growth during the NICU hospitalization very gestational age, preterm neonatal nutrition should be
clearly adversely impacts later neurodevelopment. aimed at supporting growth and maintaining energy.
There is, however, a paucity of well-designed, IV nutrition should begin right after birth, and amino
controlled studies in preterm infants of the effects acids should be infused at rates just higher than

14·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

the infant needs (3-4 g/kg/day). The excess amino   There also is little evidence to support delayed
acid load will be oxidized, providing useful energy enteral nutrition, both in terms of starting time after
and diminishing hyperglycemia. Glucose should be birth and rate of advancement, in infants who had
infused to meet glucose needs, adjusting the infusion IUGR with abnormal antenatal Doppler values,
rate to maintain normal glucose concentrations (50- particularly absent or reversed end diastolic flow
100 mg/dL). Maximum intravenous glucose infusion (AREDF) in the umbilical artery, in whom elective
rates should not exceed 6-10 mg/kg/min = 27- preterm delivery is common[123]. Such infants do have
42 kcal/kg/day. Lipids should be provided to meet increased rates of NEC and feeding intolerance, which
additional energy (and EFA) needs: 2-3 g/kg/day = 18- indicates that in addition to using fresh mother’s
36 kcal/kg/day. Intravenous feeding should be started milk, feedings should not be started until the infant
right after birth. is physiologically stable, and should be advanced
  Enteral feeding should be started as soon as the cautiously, though there is no reason to restrict early
[116-117]
infant is stable , but for most preterm infants, oral colostrum administration both to the buccal
mother’s colostrum should be provided within the mucosa and into the stomach[124] .
first hour or two after birth, and buccal swabbing    Table 2 below is one example of a feeding
with colostrum should be done immediately to help advancement protocol that adjusts for gestational age
establish an appropriate oral and gastrointestinal and prior growth restriction.
[118]
microflora in the infant . Subsequent feeding should
preferably use fresh mother’s milk, with donor milk an [References]
acceptable alternative. Protein (and minerals) should
[1] American Academy of Pediatrics, Committee on Nutrition:
be used to supplement mother’s milk and especially
Nutritional needs of low-birth-weight infants[J]. Pediatrics,
donor milk to meet the gestational age-specific protein 1985, 75(5): 976-985.
requirements necessary for normal fetal growth rates. [2] Hobar JD, Ehrenkranz RA, Badger GJ, et al. Weight growth
velocity and postnatal growth failure in infants 501 to 1500
Liquid fortifiers provide more protein than powder
grams: 2000-2013[J]. Pediatrics, 2015, 136(1): e84-e92.
fortifiers and they do not lead to infections since they [3] Lafeber HN, Westerbeek EA, van den Berg A, et al. Nutritional
can be sterilized in contrast to the powder fortifiers. factors influencing infections in preterm infants[J]. J Nutr, 2008,
138(9): 1813S-1817S.
   Trophic or gut priming enteral feeding with
[4] Bhatia J, Parish A. Nutrition and the lung[J]. Neonatology, 2009,
milk should be advanced progressively according 95(4): 362-367.
to standard protocols to avoid too many delays or [5] Commare CE, Tappenden KA. Development of the infant
intestine: implications for nutrition support[J]. Nutr Clin Pract,
stopping feedings for events, such as gastric residuals,
2007, 22(2): 159-173.
that have no bearing on adverse clinical conditions, [6] Berni Canani R, Passariello A, Buccigrossi V, et al. The
particularly necrotizing enterocolitis [119]. An every nutritional modulation of the evolving intestine[J]. J Clin
Gastroenterol, 2008, 42(Suppl 3 Pt 2): S197-S200.
3-hour feeding schedule works for most preterm
[7] Yau KI, Chang MH. Growth and body composition of preterm,
infants, though there might be an advantage to every small-for-gestational-age infants at a postmenstrual age of 37-40
2 hour feeds in the smallest infants. Many studies weeks[J]. Early Hum Dev, 1993, 33(2): 117-131.
[8] Smart JL. Vulnerability of developing brain to undernutrition[J].
and meta-analyses have documented that there is no
Ups J Med Sci, 1990, 48(Suppl): 21-41.
increase in the rate of NEC or other major morbidities [9] Smart JL. Critical periods in brain development[M]//Bock GR,
when feedings were advanced at 30-35 mL/kg/day vs. Whelan J. The Childhood Environment and Adult Disease. Ciba

slower rates of advancement(15-20 mL/kg/day[120-121]). Found Symp, Chichester. Wiley, 1991: 109-128.
[10] Georgieff MK, Hoffman JS, Pereira GR, et al. Effect of neonatal
Achieving full enteral feedings sooner reduces the caloric deprivation on head growth and 1-year developmental
risk from TPN and infections associated with invasive status in preterm infants[J]. J Pediatr, 1985, 107(4): 581-587.
catheters [122]
. [11] Hack M, Breslau N, Weissman B, et al. Effect of very low birth

·15·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

weight and subnormal head size on cognitive abilities at school [26] Wilson DC, Cairns P, Halliday HL, et al. Randomised controlled
age[J]. N Engl J Med, 1991, 325(4): 231-237. trial of an aggressive nutritional regimen in sick very low
[12] Ghods E, Kreissl A, Brandstetter S, et al. Head circumference birthweight infants[J]. Arch Dis Child Fetal Neonatal Ed, 1997,
catch-up growth among preterm very low birth weight infants: 77(1): F4-F11.
effect on neurodevelopmental outcome[J]. J Perinat Med, 2011, [27] Morgan C, McGowan P, Herwitker S, et al. Postnatal head
39 (5): 579-586. growth in preterm infants: a randomized controlled parenteral
[13] Ehrenkranz RA, Dusick AM, Vohr BR, et al. Growth in the nutrition study[J]. Pediatrics, 2014, 133(1): e120-e128.
neonatal intensive care unit influences neurodevelopmental [28] Hay WW Jr, Ziegler EE. Growth failure among preterm infants
and growth outcomes of extremely low birth weight infants[J]. is not innocuous and must be prevented. Commentary[J]. J
Pediatrics, 2006, 117(4): 1253-1261. Perinatol, 2016, 36(7): 500-502.
[14] Belfort MB, Kuban KC, O'Shea TM, et al; Extremely Low [29] Poindexter B. Approaches to growth faltering[M]//Koletzko
Gestational Age Newborn (ELGAN) Study Investigators; B, Poindexter B, Uauy R. Nutritional Care of Preterm Infants.
Extremely Low Gestational Age Newborn ELGAN Study Karger: Basel, 2014: 228-238.
Investigators. Weight status in the first 2 years of life and [30] Stoll BJ, Hansen NI, Bell EF, et al; Eunice Kennedy Shriver
neurodevelopmental impairment in extremely low gestational National Institute of Child Health and Human Development
age newborns[J]. J Pediatr, 2016,168(e2): 30-35. Neonatal Research Network. Neonatal outcomes of extremely
[15] Lodygensky GA, Seghier ML, Warfield SK, et al. Intrauterine preterm infants from the NICHD Neonatal Research Network[J].
growth restriction affects the preterm infant’s hippocampus[J]. Pediatrics, 2010, 126(3): 443-456.
Pediatr Res, 2008, 63(4): 438-443. [31] Morgane PJ, Mokler DJ, Galler JR. Effects of prenatal protein
[16] Isaacs EB, Gadian DG, Sabatini S, et al. The effect of early malnutrition on the hippocampal formation[J]. Neurosci
human diet on caudate volumes and IQ[J]. Pediatr Res, 2008, Biobehav Rev, 2002, 26(4): 471-483.
63(4): 229-231. [32] Lucas A, Morley R, Cole TJ, et al. Early diet in preterm babies
[17] Belfort MB, Rifas-Shiman SL, Sullivan T, et al. Infant growth and developmental status at 18 months[J]. Lancet, 1990,
before and after term: effects on neurodevelopment in preterm 335(8704): 1477-1481.
infants[J]. Pediatrics, 2011, 128(4): e899-e906. [33] Lucas A, Morley R, Cole TJ. Randomised trial of early diet in
[18] Vasu V, Modi N. Assessing the impact of preterm nutrition[J]. preterm babies and later intelligence quotient[J]. BMJ, 1998,
Early Hum Dev, 2007, 83 (12): 813-818. 317(7171): 1481-1487.
[19] Denne SC. Early nutritional support for extremely premature [34] Isaacs EB, Morley R, Lucas A. Early diet and general cognitive
infants: what amino acid amount should be given? [J]. Am J outcome at adolescence in children born at or below 30 weeks
Clin Nutr, 2016, 103(6): 1383-1384. gestation[J]. J Pediatr, 2009, 155(2): 229-234.
[20] Denne SC, Poindexter BB. Evidence supporting early nutritional [35] Tan MJ, Cooke RW. Improving head growth in very preterm
support with parenteral amino acid infusion[J]. Semin Perinatol, infants–a randomised controlled trial I: neonatal outcomes[J].
2007, 31(2): 56-60. Arch Dis Child Fetal Neonatal Ed, 2008, 93(5): F337-F341.
[21] Van Goudoever JB, Colen T, Wattimena JL, et al. Immediate [36] Tan M, Abernethy L, Cooke R. Improving head growth in
commencement of amino acid supplementation in preterm preterm infants – a randomised controlled trial II: MRI and
infants: effect on serum amino acid concentrations and protein developmental outcomes in the first year[J]. Arch Dis Child
kinetics on the first day of life[J]. J Pediatr, 1995, 127(3): 458- Fetal Neonatal Ed, 2008, 93 (5): F342-F346.
465. [37] Stephens BE, Walden RV, Gargus RA, et al. First-week protein
[22] Thureen PJ, Melara D, Fennessey PV, et al. Effect of low versus and energy intakes are associated with 18-month developmental
high intravenous amino acid intake on very low birth weight outcomes in extremely low birth weight infants[J]. Pediatrics,
infants in the early neonatal period[J]. Pediatr Res, 2003, 53(1): 2009, 123(5): 1337-1343.
24-32. [38] Naeye R, Dixon J. Distortions in fetal growth standards[J].
[23] Poindexter BB, Langer JC, Dusick AM, et al; National Pediatr Res, 1978, 12 (10): 987-991.
Institute of Child Health and Human Development Neonatal [39] Fenton TR, Kim JH. A systematic review and meta-analysis
Research Network. Early provision of parenteral amino acids to revise the Fenton growth chart for preterm infants[J]. BMC
in extremely low birth weight infants: relation to growth and Pediatr, 2003, 13: 59.
neurodevelopmental outcome[J]. J Pediatr, 2006, 148(3): 300- [40] Olsen IE, Groveman SA, Lawson ML, et al. New intrauterine
305. growth curves based on United States data[J]. Pediatrics, 2010,
[24] Cormack BE, Bloomfield FH. Increased protein intake decreases 125(2): e214-e224.
postnatal growth faltering in ELBW babies[J]. Arch Dis Child [41] Carlson SJ, Ziegler EE. Nutrient intakes and growth of very low
Fetal Neonatal Ed, 2013, 98(5): F399-F404. birth weight infants[J]. J Perinatol, 1998, 18(4): 252-258.
[25] Bolisetty S, Pharande P, Nirthanakumaran L, et al. Improved [42] Ehrenkranz RA, Younes N, Lemons JA, et al. Longitudinal
nutrient intake following implementation of the consensus growth of hospitalized very low birth weight infants[J].
standardised parenteral nutrition formulations in preterm Pediatrics, 1999, 104(2 Pt 1): 280-289.
neonates: a before-after intervention study[J]. BMC Pediatr, [43] Dancis J, O'connell Jr, Holt LE Jr. A grid for recording the
2014, 14: 309-315. weight of premature infants[J]. J Pediatrics, 1948, 33(1): 570-

16·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

572. Needs of the Preterm Infant. Pawling, NY: Caduceus Medical


[44] Dinerstein A, Solana CL, Perez GP, et al. Early and aggressive Publishers, 1993: 29-46.
nutritional strategy (parenteral and enteral) decreases postnatal [61] Kashyap S, Forsyth M, Zucker C, et al. Effects of varying
growth failure in very low birth weight infants[J]. J Perinatol, protein and energy intakes on growth and metabolic response in
2006, 26(7): 436-442. low birth weight infants[J]. J Pediatr, 1986, 108(6): 955-963.
[45] Kitanaka T, Alonso JG, Gilbert RD, et al. Fetal responses to [62] Vasu V , Thomas EL, Durighel G, et al. Early nutritional
long-term hypoxemia in sheep[J]. Am J Physiol, 1989, 256(6 Pt determinants of intrahepatocellular lipid deposition in preterm
2): R1348-R1354. infants at term age[J]. Int J Obes (Lond), 2013, 37(4): 500-504.
[46] Stockman JA, Clark DA. Weight gain: a response to transfusion [63] Olsen IE, Lawson ML, Ferguson AN, et al. BMI curves for
in selected preterm infants[J]. Am J Dis Child, 1984, 138(9): preterm infants[J]. Pediatrics, 2015, 135(3): e572-e581.
828-830. [64] Ziegler EE, O'Donnell AM, Nelson SE, et al. Body composition
[47] Carver TD, Anderson SM, Aldoretta PA, et al. Glucose of the reference fetus[J]. Growth, 1976, 40(4): 329-341.
suppression of insulin secretion in chronically hyperglycemic [65] Embleton ND. Optimal protein and energy intakes in preterm
fetal sheep[J]. Pediatr Res, 1995, 38(5): 754-762. infants[J]. Early Human Dev, 2007, 83(12): 831-837.
[48] Hay WW Jr, Sparks JW, Quissell BJ, et al. Simultaneous [66] Thureen PJ, Anderson AH, Baron KA, et al. Protein balance in
measurements of umbilical uptake, fetal utilization rate, and the first week of life in ventilated neonates receiving parenteral
fetal turnover rate of glucose [J]. Am J Physiol, 1981, 240(6): nutrition[J]. Am J Clin Nutr, 1998, 68(5): 1128-1135.
E662-E668,. [67] Morgan C. Early amino acid administration in very preterm
[49] Zarlengo KM, Battaglia FC, Fennessey P, et al. Relationship infants: Too little, too late, or too much, too soon? [J]. Semin
between glucose utilization rate and glucose concentration in Fetal Neonatal Med, 2013, 18: 160-165.
preterm infants[J]. Biol Neonate, 1986, 49(4): 181-189. [68] Uthaya S, Modi N. Practical preterm parenteral nutrition:
[50] Sunehag A, Ewald U, Larsson A, et al. Glucose production rate systematic literature review and recommendations for
in extremely immature neonates (< 28 weeks) studied by use of practice[J]. Early Human Dev, 2014, 90(11): 747-753.
deuterated glucose[J]. Pediatr Res, 1993, 33(2): 97-100. [69] Clark RH, Chace DH, Spitzer AR; Pediatrix Amino Acid
[51] Marconi AM, Paolini C, Buscaglia M, et al.The impact of Study Group. Effects of two different doses of amino acid
gestational age and fetal growth on the maternal-fetal glucose supplementation on growth and blood amino acid levels in
concentration difference[J]. Obstet Gynecol, 1996, 87(6): 937- premature neonates admitted to the neonatal intensive care unit:
942. a randomized, controlled trial[J]. Pediatrics, 2007,120(6): 1286-
[52] Srinivasan G, Pildes RS, Cattamanchi G, et al. Plasma glucose 1296.
values in normal neonates: a new look[J]. J Pediatr, 1986, [70] Thureen PJ, Hay WW. Early aggressive nutrition in preterm
109(1): 114-117. infants[J]. Semin Neonatol, 2001, 6(5): 403-415.
[53] Helms RA, Mauer EC, Hay WW Jr, et al. Effect of intravenous [71] Embleton ND, Morgan C, King C. Balancing the risks and
L-carnitine on growth and fat metabolism parameters during benefits of parenteral nutrition for preterm infants: can we define
parenteral nutrition in neonates[J]. JPEN J Parenter Enteral Nutr, the optimal composition[J]. Arch Dis Child Fetal Neonatal Ed,
1990,14(5): 448-453. 2015, 100(1): F72-F75.
[54] Sellmayer A, Koletzko B. Long-chain polyunsaturated [72] Brown LD, Rozance PJ, Thorn SR, et al. Acute supplementation
fatty acids and eicosanoids in infants – Physiological and of amino acids increases net protein accretion in IUGR fetal
pathophysiological aspects and open questions[J]. Lipids, 1999, sheep[J]. Am J Physiol Endocrinol Metab, 2012, 303(3):
34(2): 199-205. E352-E364.
[55] Carlson SE. Docosahexaenoic acid and arachidonic acid in [73] Sunehag AL. Hyperglycemia is a risk factor for early death and
infant development[J]. Semin Neonatol, 2001, 6(5): 437-449. morbidity in extremely low birth-weight infants[J]. Pediatrics,
[56] Chacko SK, Ordonez J, Sauer PJ, et al. Gluconeogenesis is not 2006, 118(5): 1811-1818.
regulated by either glucose or insulin in extremely low birth [74] Stensvold HJ, Strommen K, Lang AM, et al. Early enhanced
weight infants receiving total parenteral nutrition[J]. J Pediatr, parenteral nutrition, hyperglycemia, and death among extremely
2011, 158 (6): 891-896. low-birth-weight infants[J]. JAMA Pediatr, 2015, 169(11):
[57] Lapillonne A, Eleni dit Trolli SE, Kermorvant-Duchemin E. 1003-1010.
Postnatal DHA deficiency is an inevitable consequence of [75] Picard M, Juster RP, McEwen BS. Mitochondrial allostatic load
current recommendations and practices in preterm infants[J]. puts the 'gluc' back in glucocorticoids[J]. Nat Rev Endocrinol,
Neonatology, 2010, 98(4): 397-403. 2014, 10(5): 303-310.
[58] Tam EWY, Chau V, Barkovich AJ, et al. Early postnatal [76] Stoll B, Horst DA, Cui L, et al. Chronic parenteral nutrition
docosahexaenoic acid levels and improved preterm brain induces hepatic inflammation, steatosis, and insulin resistance in
development[J]. Pediatr Res, 2016, 79(5): 723-730. neonatal pigs[J]. J Nutr, 2010, 140(12): 2193-2200.
[59] Carlson SE, Werkman SH, Rhodes PG, et al. Visual acuity [77] Chavez-Valdez R, McGowan J, Cannon E, et al. Contribution of
development in healthy preterm infants: effect of marine oil early glycemic status in the development of severe retinopathy
supplementation[J]. Am J Clin Nutr, 1993, 58(1): 35-42. of prematurity in a cohort of ELBW infants[J]. J Perinatology,
[60] Micheli JL, Schutz Y. Protein[M]//Tsang RC. Nutritional 2011, 31(12): 749-756.

17·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

[78] Mohsen L, Abou-Alam M, El-Dib M, et al. A prospective recent recommendations in VLBW infants and postnatal growth
study on hyperglycemia and retinopathy of prematurity[J]. J restriction[J]. J Pediatr Gastroenterol Nutr, 2011, 53(5): 536-
Perinatology, 2014, 34(6): 453-457. 542.
[79] Garg R, Agthe AG, Donohue PK, et al. Hyperglycemia and [93] Miller M, Vaidya R, Rastogi D, et al. From parenteral to enteral
Retinopathy of Prematurity in Very Low Birth Weight Infants[J]. nutrition: a nutrition-based approach for evaluation of postnatal
J Perinatology, 2003, 23(3): 186-194. growth failure in preterm infants[J]. J Parenter Enteral Nutr,
[80] Basu SK, Kaiser JR, Guffey D, et al; CoolCap Study Group. 2014, 38(4): 489-497.
Hypoglycaemia and hyperglycaemia are associated with [94] The SIFT Investigators Group. Early enteral feeding strategies
unfavourable outcome in infants with hypoxic ischaemic for very preterm infants: current evidence from Cochrane
encephalopathy: a post hoc analysis of the CoolCap Study[J]. reviews[J]. Arch Dis Child Fetal Neonatal Ed, 2013, 98(6):
Arch Dis Child Fetal Neonatal Ed, 2016, 101(2): F149-F155. F470-F472.
[81] Scheurer JM, Gray HL, Demerath EW, et al. Diminished growth [95] Shah SD, Dereddy N, Jones TL, et al. Early versus delayed
and lower adiposity in hyperglycemic very low birth weight human milk fortification in very low birth weight infants-a
neonates at 4 months corrected age[J]. J Perinatol, 2016, 36(2): randomized controlled trial[J]. J Pediatr, 2016, 174: 126-131.
145-150. [96] Casaer MP, Mesotten D, Hermans G, et al. Early versus late
[82] Hay WW Jr, Brown LD, Rozance PJ, et al. Challenges in parenteral nutrition in critically ill adults[J]. N Engl J Med,
nourishing the intrauterine growth-restricted foetus - Lessons 2011, 365(6): 506-517.
learned from studies in the intrauterine growth-restricted foetal [97] Gramlich L, Kichian K, Pinilla J, et al. Does enteral nutrition
sheep[J]. Acta Paediatr, 2016, 105(8): 881-889. compared to parenteral nutrition result in better outcomes
[83] Beardsall K, Vanhaesebrouck S, Ogilvy-Stuart AL, et al. A in critically ill adult patients? A systematic review of the
randomised controlled trial of early insulin therapy in very low literature[J]. Nutrition, 2004, 20(10): 843-848.
birth weight infants, "NIRTURE" (neonatal insulin replacement [98] Seres DS, Valcarcel M, Guillaume M. Advantages of enteral
therapy in Europe) [J]. BMC Pediatr, 2007, 7: 29. nutrition over parenteral nutrition[J]. Therap Adv Gastroenterol,
[84] Sadhwani A, Asaro LA, Goldberg C, et al. Impact of tight 2013, 6(2): 157-167.
glycemic control on neurodevelopmental outcomes at 1 year [99] Fivez T, Kerklaan D, Mesotten D, et al. Early versus late
of age for children with congenital heart disease: a randomized parenteral nutrition in critically ill children[J]. N Engl J Med,
controlled trial[J]. J Pediatrics, 2016, 174: 193-198. 2016, 374(12): 1111-1122.
[85] Bottino M, Cowett RM, Sinclair JC. Interventions for treatment [100] Srinivasan V, Agus MS. Tight glucose control in critically ill
of neonatal hyperglycemia in very low birth weight infants[J]. children—a systematic review and metab-analysis[J]. Pediatr
Cochrane Database Syst Rev, 2009, (1): CD007453. Diabetes, 2014, 15(2): 75-83.
[86] Meetze W, Bowsher R, Compton J, et al. Hyperglycemia in [101] Ohnishi S, Ichiba H, Tanaka Y, et al. Early and intensive
extremely-low-birth-weight infants[J]. Biol Neonate, 1998, nutritional strategy combining parenteral and enteral feeding
74(3): 214-21. promotes neurodevelopment and growth at 18months of
[87] Burattini I, Bellagamba MP, Spagnoli C, et al; Marche Neonatal corrected age and 3years of age in extremely low birth weight
Network. Targeting 2.5 versus 4 g/kg/day of amino acids for infants[J]. Early Hum Dev, 2016, 100: 35-41.
extremely low birth weight infants: a randomized clinical [102] Liu J, Kong K, Tao Y, et al. Optimal timing for introducing
trial[J]. J Pediatr, 2013, 163(5): 1278-1282. enteral nutrition in the neonatal intensive care unit[J]. Asia Pac J
[88] Wang H, Khaoustov VI, Krishnan B, et al. Total parenteral Clin Nutr, 2015, 24(2): 219-226.
nutrition induces liver steatosis and apoptosis in neonatal [103] Owens L, Burrin DG, Berseth CL. Minimal enteral feeding
piglets[J]. J Nutr, 2006, 136(10): 2547-2552. induces maturation of intestinal motor function but not mucosal
[89] Clark RH, Chace DH, Spitzer AR. Effects of two different doses growth in neonatal dogs[J]. J Nutr, 2002, 132(9): 2717-2722.
of amino acid supplementation on growth and blood amino acid [104] Morgan J, Young L, McGuire W. Delayed introduction of
levels in premature neonates admitted to the neonatal intensive progressive enteral feeds to prevent necrotizing enterocolitis in
care unit: a randomized, controlled trial[J]. Pediatrics, 2007, very low birth weight infants[J]. Cochrane Database Syst Rev,
120(6): 1286-1296. 2011, 16(3): CD001970.
[90] Uthaya S, Liu X, Babalis D, et al. Nutritional evaluation and [105] Niinikoski H, Stoll B, Guan X, et al. Onset of small intestinal
optimisation in neonates: a randomized, double-blind controlled atrophy is associated with reduced intestinal blood flow in TPN-
trial of amino acid regimen and intravenous lipid composition fed neonatal piglets[J]. J Nutr, 2004, 134(6): 1467-1474.
in preterm parenteral nutrition[J]. Am J Clin Nutr, 2016, 103(6): [106] Clyman R, Wickremasinghe A, Jhaveri N, et al. Ductus
1443-1452. Arteriosus Feed or Fast with Indomethacin or Ibuprofen
[91] Morgan C, Burgess L. High protein intake does not prevent (DAFFII) Investigators. Enteral feeding during indomethacin
low plasma levels of conditionally essential amino acids in and ibuprofen treatment of a patent ductus arteriosus[J]. J
very preterm infants receiving parenteral nutrition[J]. JPEN Pediatr, 2013, 163(2): 406-411.
J Parenter Enteral Nutr, 2015, Jul 6. pii: 0148607115594009, [107] Louis D, Torgalkar R, Shah J, et al. Enteral feeding during
Epub ahead of print. indomethacin treatment for patent ductus arteriosus: association
[92] Senterre T, Rigo J. Optimizing early nutritional support based on with gastrointestinal outcomes[J]. J Perinatol, 2016, 36(7): 544-

18·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

548. infant[J]. Semin Fetal Neonatal Med, 2013, 18: 150-154.


[108] Chowning R, Radmacher P, Lewis S, et al. A retrospective [117] Dutta S, Singh B, Chessell L, et al. Guidelines for feeding very
analysis of the effect of human milk on prevention of necrotizing low birth weight infants[J]. Nutrients, 2015, 7(1): 423-442.
enterocolitis and postnatal growth[J]. J Perinatol, 2016, 36(3): [118] Rodriguez NA, Meier PP, Groer MW, et al. A pilot study
221-224. to determine the safety and feasibility of oropharyngeal
[109] Ziegler EE. Human milk and human milk fortifiers[J]. World administration of own mother’s colostrum to extremely low-
Rev Nutr Diet, 2014, 110: 215-227. birth-weight infants[J]. Adv Neonatal Care, 2010, 10(4): 206-
[110] Arslanoglu S, Moro GE, Ziegler EE; The Wapm Working 212.
Group On Nutrition. Optimization of human milk fortification [119] Rochow N, Fusch G, Muhlinghaus A, et al. A nutritional
for preterm infants: new concepts and recommendations[J]. J program to improve outcome of very low birth weight infants[J].
Perinat Med, 2010, 38(3): 233-238. Clin Nutr, 2012, 31(1): 124-131.
[111] Rochow N, Fusch G, Choi A, et al. Target fortification of breast [120] Morgan J, Young L, McGuire W. Slow advancement of enteral
milk with fat, protein, and carbohydrates for preterm infants[J]. feed volumes to prevent necrotizing enterocolitis in very low
J Pediatrics, 2013, 163(4): 1001-1007. birth weight infants[J]. Cochrane Database Syst Rev, 2011,
[112] Kuschel CA, Harding JE. Multicomponent fortified human milk 16(3): CD001241.
for promoting growth in preterm infants[J]. Cochrane Database [121] Dorling J. How quickly should we aim for full milk feeds? [J].
Syst Rev, 2004, (1): CD000343. Infant, 2012, 8: 167168.
[113] Sullivan S, Schanler RJ, Kim JH, et al. An exclusively human [122] Härtel C, Haase B, Browning-Carmo K, et al. Does the enteral
milk-based diet is associated with a lower rate of necrotizing feeding advancement affect short-term outcomes in very low
enterocolitis than a diet of human milk and bovine milk-based birth weight infants? [J]. J Pediatr Gastroenterol Nutr, 2009,
products[J]. J Pediatr, 2010, 156(4): 562.e1-567.e1. 48(4): 464-470.
[114] Ong KK, Kennedy K, Castañeda-Gutiérrez E, et al. Postnatal [123] Mihatsch WA, Pohlandt F, Franz AR, et al. Early feeding
growth in preterm infants and later health outcomes: a advancement in very low-birth-weight infants with intrauterine
systematic review[J]. Acta Paediatr, 2015, 104(10): 974-986. growth retardation and increased umbilical artery resistance[J]. J
[115] Lapillone A, Griffin IJ. Feeding preterm infants today for later Pediatr Gastroenterol Nutr, 2002, 35(2): 144-148.
metabolic and cardiovascular outcomes[J]. J Pediatr, 2013, [124] Dorling J, Kempley S, Leaf A. Feeding growth restricted
162(3 Suppl): S7-S16. preterm infants with abnormal antenatal Doppler results[J]. Arch
[116] Leaf A. Introducing enteral feeds in the high-risk preterm Dis Child Fetal Neonatal Ed, 2005, 90(5): F359-F363.

附 "Optimizing nutrition of the preterm infant" 一文的中文摘译


早产儿营养的最优化 (黑明燕 摘译)

[摘要] 早产儿营养补给的目标是使早产儿能够达到同胎龄健康胎儿的正常生长速率,并且在器官生长、
组织成分、以及细胞数量和结构方面也能达到健康胎儿水平。营养物质的数量与质量对于早产儿的正常生长发
育、包括神经系统发育是至关重要的。基本营养素的供给不足不仅会造成生长受限,而且会增加病死率或影响
神经发育结局。早产儿在 NICU 住院期间的生长速率在神经发育及整个人体发育的结局中发挥了重要作用。尽
管对于最佳营养的需求非常明显,但早产儿生长落后的例子比比皆是。因此,根据总的能量及蛋白质需求以及
个体成分如氨基酸、碳水化合物及脂肪,甚至细化到氧气的需求来优化早产儿的营养是非常必要的。该综述阐
述了早产儿科学合理的营养需求、具有实用性的营养指南以及早产儿静脉营养、肠内营养的方法步骤。包括氨
基酸在内的静脉营养,应该从一出生便按照相应胎龄所适合的速率开始补充。肠内营养则应在出生后尽早开始,
首选母亲的初乳和牛奶。肠内营养应根据热能需要开始建立并在能耐受的范围内快速增加,保持营养摄入在推
荐速率的同时相应地减少静脉营养摄入量。制定一个合理的喂养方案对于改善营养状况及相关的转归是重要的。
什么是早产儿的最佳营养和生长速率尚有待于进一步的研究来确定,这种最佳营养不仅能使早产儿神经认知得
到最充分的发育,同时还要最大程度地限制其远期发生慢性疾病的风险。
[关键词] 静脉营养;胃肠外营养;肠内营养;氨基酸;葡萄糖;脂肪;坏死性小肠结肠炎;早产儿

1 概述 疾病的易感性,增加呼吸窘迫的发生率,降低损伤组织
的修复能力以及影响大脑发育。神经元发育障碍可能是
早产儿营养补给的目标是达到同胎龄健康胎儿的 早产儿营养缺失所造成的最严重的后遗症,可导致认知
正常生长速率。基本营养素的供给不足不仅会造成生长 缺陷,神经发育结局的改善与早产儿营养状况的提高相
受限,同时会导致免疫防御不足而增加早产儿对感染性 关。
19·
·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

根据总的能量及蛋白质需求以及个体成分如氨基 加至 3.5~4 g/kg/d(极低出生体重儿 <27 周妊娠增加至


酸、碳水化合物及脂肪,甚至细化到氧气的需求来优化 4 g/kg/d)。
早产儿的营养是非常必要的。临床上给早产儿提供充 葡萄糖:出生后葡萄糖(10% 葡萄糖液)的输注
足的营养一直面临着挑战,特别是早产儿刚出生时依 速度应以新生儿能耐受且不引起高血糖的最大速率予以
赖静脉营养的情况下,此时蛋白质储存的降低每天可 输注,以防低血糖的发生。高血糖的定义为出生后任意
达 1.5%。在出生后最初 5 d 保证每天至少 3 g/kg 氨基酸 血糖浓度 >120 mg/dL。出生后 1 h 到几天,各种应激反
摄入量可以减少极低出生体重早产儿的蛋白质分解和缺 应导致体内去甲肾上腺素和皮质醇的水平较高,因此葡
失,并可提高蛋白质平衡和生长结局。营养物质的数量 萄糖输注过快时(> 6~8 mg/kg/min)会造成高血糖。早
与质量对于早产儿的正常生长发育(包括神经系统发育 产儿高血糖是加重全身炎症反应的潜在因素,且高血糖
的结局)至关重要,但临床上早产儿生长发育受限的现 的早产儿患视网膜病变的风险增加。
象较普遍。早产儿在 NICU 住院期间的生长速率在早产 脂质:静脉脂质输注应在出生后 24 h 内开始,并
儿神经系统发育结局和体型发育方面发挥了关键的、不 提高到可耐受的 2.0 至 3.0~3.5 g/kg/d,通常持续 2~3 d。
可被取代的作用。Lucas 等的研究显示,给予 4 周充足 静脉中的脂质为氧化代谢提供了碳,并且提供了有利于
剂量的蛋白质及能量的早产儿,在纠正胎龄 18 个月时 全身细胞特别是脑组织细胞膜的合成所需的脂肪酸。但
生长发育及神经发育得到明显改善,7.5 岁时的认知能 是,过多的脂质输注会导致高甘油三酯血症(定义为任
力高于对照组,16 岁时头颅 MRI 影像学检查显示其头 意血甘油三酯浓度大于 150 mg/dL),会导致脂质的不
围和尾状核体积高于对照组且智商(IQ)评分较高。 良沉积及肝细胞发生炎症,这可能也是长期静脉营养的
然而目前的营养方案仍很难实现早产儿最佳营养 患儿常发生胆管性黄疸和肝衰竭的原因。极低出生体重
管理从而达到最佳生长发育状态,因此,有必要重新评 儿清除血浆脂质的能力降低,建议出生后头几天的脂质
估早产儿应该达到的生长率以及实现该生长率的基本营 输注率为 0.5~1.0 g/kg/d,特别是合并感染、严重肺疾病、
养需求。 外科应激、使用激素时。
热 卡: 第 1 天 为 50 kcal/kg/d, 至 第 3 天 应 达 到
2 关于早产儿生长速度 80 kcal/kg/d。

直接测量正常的胎儿生长是不可能的。目前根 4 关于静脉营养向肠内营养的过渡
据貌似“正常”的存活早产儿间接估算的数据,妊娠
24~38 周胎儿体重平均增加 17 g/kg/d(图 1),但实际 在肠外营养向肠内营养过渡期间,由于营养摄入
上大多数早产儿出生后达不到宫内生长的速度(图 2), 量的暂时性下降,蛋白质和能量的摄入可能会下降。解
主要的原因就是营养不足(图 3)。首先,大多数早产 决这一问题的办法是尽快增加肠内营养和应用母乳强化
儿出生后第一日(甚至数日)的静脉氨基酸量很低甚至 剂。母乳强化剂可在肠内喂养量达到 50 mL/kg/d 时就开
没有;其次,开始肠内喂养的时间滞后;第三,大多数 始。一项对 100 个极低出生体重儿的随机研究显示,在
婴儿只能耐受有限的营养支持,如静脉氨基酸的输注率 肠内喂养量仅 20 mL/kg/d 的时候就开始早期添加母乳强
<3 g/kg/d;第四,在开始营养支持后,静脉氨基酸、脂 化剂,与肠内喂养达到 100 mL/kg/d 后才开始添加母乳
类和肠内营养的增加都很缓慢;第五,大部分婴儿应用 强化剂相比,早期开始添加母乳强化剂的婴儿在第 1、2、
的是未强化母乳或必需氨基酸含量不足的完全胃肠外营 3 周龄的每日蛋白摄入量更高,在第 4 周龄的累计蛋白
养。多数情况下人们只是假设生理上尚不稳定的早产儿 摄入量也更高,但两组在达到完全喂养量所需的天数、
不能承受正常量肠外或肠内营养。 喂养耐受和坏死性小肠结肠炎发生率方面没有差异。

3 关于全静脉注射 5 关于肠内营养

氨基酸:应在生后 2~3 h 内开始注射至少 2 g/kg/d 长时间肠外营养可导致感染率增加(由于免疫抑


的氨基酸,<30 周孕龄的早产儿应在第一个 24 ~ 48 h 增 制、肠道菌群失调以及中心静脉置管相关的细菌感染几

· 20·
第 19 卷 第 1 期 中国当代儿科杂志 Vol.19 No.1
2017 年 1 月 Chin J Contemp Pediatr Jan. 2017

率增加)、肠黏膜萎缩、肠道分泌的消化酶减少、心肝 地维持早产儿的生长以达到相同胎龄儿的生长速率和身
脂肪浸润增加、全身炎症反应综合征、IgA 减少等。在 体成分,早产儿的营养应旨在支持生长和维持能量。静
成人和年长儿、足月儿的研究中,当肠内营养受限而静 脉营养应在出生后立即开始,并且氨基酸的输注率应稍
脉营养过多时,多种疾病的发生率或病死率均增加。这 高于婴儿所需(3~4 g/kg/d),这样有助于减少高血糖的
些问题很可能是过多输注葡萄糖导致高血糖的结果。尽 发生。葡萄糖的供给应满足机体需要、维持正常的血糖
早开始肠内营养,同时适度给予静脉营养才是有益的。 浓度(50~100 mg/dL),葡萄糖静脉输注的最大速度应
与静脉营养一样,肠内营养应提供可以满足相 不超过 10 mg/kg/min(27~42 kcal/kg/d)。脂质供给应满
同 胎 龄 胎 儿 正 常 生 长 发 育 所 需 的 蛋 白 质。 对 于 胎 龄 足额外的能量的需要:2~3 g/kg/d(18~36 kcal/kg/d)。
<28~30 周的婴儿,必须提供≥ 2.9 g/100 mL 蛋白浓度的 肠内喂养应在婴儿稳定后尽早开始。对于大多数
奶,以满足胎儿生长所需的 4.0 g/kg/d 的蛋白质。尽管 早产儿,在出生后的 1~2 h 内可予以口服初乳,并且立
母亲的母乳(首选)或供者的母乳(次选)都是新生儿 即再在口颊部位用初乳进行涂抹,这样做将有助于早产
肠内营养所推荐的食物,但它们的营养素浓度特别是蛋 儿建立正常的口腔和肠道菌群。后续的喂养则最好使用
白质的浓度太低,不能满足早产儿的生长需求。液体母 新鲜的母乳,在亲母母乳不足时,也可选用捐赠母乳。
乳强化剂与粉状母乳强化剂相比,液体强化剂可以添加 可在母乳中添加母乳强化剂以增加蛋白质和矿物质的含
的蛋白质较多(每 100 mL 牛奶大约 1.7 g 蛋白质),通 量,液体母乳强化剂比粉末母乳强化剂提供的蛋白质更
常可达到满意的蛋白摄入量。 多且不会导致感染,因为液体母乳强化剂可被消毒而粉
液态母乳强化剂的添加量为每 25 mL 奶加 1 mL 母 末状母乳强化剂不易消毒。
乳强化剂,肠内喂养 150 mL/kg/d 添加了强化剂的母乳, 肠内喂养量应按照喂养指南的要求逐步增加,要
可提高 1 g/kg/d 的蛋白摄入量。系统回顾研究并未发现 尽量避免因婴儿存在胃残留 , 或担心坏死性小肠结肠炎
牛奶来源的母乳强化剂可导致婴儿坏死性小肠结肠炎的 的发生而延迟或停止肠内营养。每 3 h 喂一次的方法适
发生率提高。最新研究发现,使用人乳来源的母乳强化 用于大部分早产儿。关于加奶速度,临床研究和 Meta
剂时婴儿坏死性小肠结肠炎的发生率更低。 分析结果显示,以 30~35 mL/kg/d 的速度增加肠内喂养
量与以 15~20 mL/kg/d 的速度增加肠内喂养量相比,坏
6 其他建议 死性小肠结肠炎的发生率并没有增加。对于产前脐血彩
超不正常(特别是脐动脉血液断流或者母胎输血)的宫
胎儿营养是专为生长和维持能量平衡设计的,应 内生长受限患儿,尽管坏死性小肠结肠炎或喂养不耐受
注意到胎儿营养的以下几个方面的问题:首先,氨基酸 的风险会增加,但目前没有证据显示对这部分早产儿需
是在胎盘以主动转运的方式摄入到胎儿体内的,其速率 延迟开奶或减慢肠内喂养添加速度。所有早产儿都可以
高于胎儿蛋白质合成及净蛋白累积所需;其次,葡萄糖 尽早开始应用初乳进行口腔颊黏膜的擦拭。
的摄入和利用依据的是胎儿代谢所需的热卡。为了更好
(本文编辑:邓芳明)

· 21·
713202
research-article2017
PENXXX10.1177/0148607117713202Journal of Parenteral and Enteral NutritionKoo and Tice

Original Communication
Journal of Parenteral and Enteral
Nutrition
Human Milk Fortifiers Do Not Meet the Current Volume XX Number X
Month 201X 1­–8
Recommendation for Nutrients in Very Low © 2017 American Society

Birth Weight Infants for Parenteral and Enteral Nutrition


DOI: 10.1177/0148607117713202
https://doi.org/10.1177/0148607117713202
journals.sagepub.com/home/pen

Winston Koo, MBBS1,2; and Hilary Tice, PharmD1,3

Abstract
Background: Use of multinutrient fortifiers is standard of care for small preterm infants fed exclusively human milk. However, adequacy
of human milk fortifiers (HMFs) to meet the recommended intake for macronutrients and micronutrients is now known. Materials and
Methods: Nutrient content of human milk fortified according to manufacturer’s recommendations was compared at isocaloric levels for 1
human milk–based (HMF-A), 2 bovine milk protein–based (HMF-B, HMF-C), and 2 preterm infant formulas (PTF-B, PTF-C). In addition,
4 multivitamin supplements were compared. Results: At 130 kcal/kg, intake of macronutrients was similar to the recommendation,
although deficient and excess intake of micronutrient occurred with all fortifiers. Four to 9 micronutrients were absent in HMF or PTF
(biotin, choline, inositol, carnitine, taurine, molybdenum, iodine, selenium, or chromium). For the remainder, HMF resulted in deficient
intake for 1–13 micronutrients, occurring most frequently with HMF-A. Excess micronutrients (3–15 at <50% and 1–3 at 109%–437%)
occurred with all HMF and most frequently with HMF-B and HMF-C. At 150 kcal/kg, deficient intake improved but generally remained
below recommendation, while excess intake became exaggerated. PTF and multivitamin formulations do not fully compensate for the
deficiencies and can result in extremely high micronutrient intake. Conclusions: At the recommended energy intake for very low birth
weight infants, many micronutrients are absent or are present in grossly inadequate amounts, and several micronutrients are in excess.
Reformulation of HMF is urgently needed since PTF or multivitamin supplement only partially corrects some deficiencies while providing
some nutrients in excess. (JPEN J Parenter Enteral Nutr. XXXX;xx:xx-xx)

Keywords
prematurity; human milk; human milk fortifier; vitamin supplement; preterm infant formula; micronutrients; macronutrients; milk
fortification; very low birth weight

Clinical Relevancy Statement nutrition support of all VLBW infants fed own mother’s milk
or donor human milk.3-5
Nutrition support of small preterm infants receiving human For VLBW infants, recommended ranges of intake to mini-
milk includes fortification with multiple nutrients. Knowledge mize the risk of nutrition deficiency and excess are
of the limitations of various commercial human milk fortifiers available.1,2,6,7 Multiple commercial human milk–based or
to meet the recommended intake for macronutrients and micro- bovine milk protein–based human milk fortifiers (HMFs) as liq-
nutrients allows the industry to make necessary formulation uid or powder preparations and nutrient-enriched preterm infant
changes to improve the nutrition support of infants most vul- formulas (PTFs) are used as fortification of human milk.
nerable to the ill effects from deficient or excess intake of However, there is a lack of comparative data on the adequacy to
nutrients.

From the 1Department of Pediatrics, Louisiana State University Health


Background Sciences Center, Shreveport, Louisiana, USA; 2Department of Nutrition
and Food Science, Wayne State University, Detroit, Michigan, USA; and
Nutrition support for the small preterm infant is complicated at
the 3Department of Clinical Sciences, University of Louisiana, Monroe,
birth by the low or absent nutrient reserve and after birth by Louisiana, USA
issues related to delivery, tolerance, and utilization of the nutri-
Financial disclosure: None declared.
ents and by greater demands of nutrients for catch-up growth
after recovery from illnesses. Deficiencies in energy, macronu- Conflicts of interest: None declared.
trients, and multiple micronutrients are well documented to Received for publication March 23, 2017; accepted for publication May
have adverse effects on physiologic function and postnatal 11, 2017.
growth of very low birth weight (VLBW) infants (<1500 g).1,2
Corresponding Author:
Small preterm infants fed exclusively human milk are at risk Winston Koo, MBBS, Department of Nutrition and Food Science, Wayne
for deficiency of multiple nutrients, and the use of fortifiers State University, Detroit, MI 48202, USA.
containing multiple nutrients is now the standard of care in the Email: wkoo@lsuhsc.edu
2 Journal of Parenteral and Enteral Nutrition XX(X)

provide the recommended quantity of macronutrients and micro- <1 kg: energy (130–150 kcal), protein (3.8–4.4 g), fat (6.2–8.4
nutrients from the use of different HMFs or PTFs as fortification g), and carbohydrate (9–20 g).2 The recommended ranges of
of human milk for VLBW infants. The goal of this study is to intake of micronutrients are the same for all VLBW infants.2
determine whether the nutrient contents of human milk after for- However, there is a range of recommended intakes for indi-
tification with different commercial HMFs or PTFs can meet the vidual micronutrients, as indicated by the difference between
current recommended range of intake.2 the upper and lower recommended intakes for fat-soluble vita-
mins (A = 114%, D = 167%, E = 100%, K = 25%), water-solu-
ble vitamins (B1 = 33%, B2 = 44%, B3 = 33%, B5 = 42%, B6 =
Materials and Methods 40%, B7 = 67%, B9 = 100%, B12 = 0%, C = 33%), other organic
Data for the nutrients from HMF were based on the final con- compounds (choline = 94%, inositol = 153%, carnitine = 0,
tent in the human milk fortified according to the HMF manu- taurine = 100%), minerals (Ca = 120%, P = 133%, Mg = 90%),
facturer’s recommendation. These data were obtained from trace minerals (Fe = 100%, Mo = 0%, Zn = 200%, Cu = 25%,
product information at the manufacturer’s website, and nutri- I = 500%, Se = 246%, Mn = 1007%, Cr = 125%), and electro-
ent contents were calculated and compared according to the lytes (Na = 67%, K = 50%, Cl = 133%).
energy intake recommended by the American Academy of There are 2 commercial liquid fortifiers from 1 manufac-
Pediatrics Committee of Nutrition.2 Data for liquid and powder turer (Ross Products; HMF hydrolyzed protein–concentrated
(where available) HMF were obtained. In addition, the nutrient liquid and HMF-concentrated liquid). While these products
content of human milk fortified by PTF with the highest energy have similar energy content, the former provides slightly
content (≈1 kcal/mL) as recommended by manufacturers at the higher protein and lower fat and carbohydrate content and
volume ratio of 1:1 with human milk was obtained via the some minor variability in micronutrient content. For the pur-
same procedure. The macronutrient and micronutrient data pose of comparison, only the former product is used in this
were compared with the recommended enteral intake2 at isoca- report. Furthermore, comparison of HMFs from various manu-
loric levels. facturers is based on the use of liquid products.
Data on commercial infant multivitamins preparations were Based on the standard fortifications recommended by manu-
obtained from Lexicomp Pediatric and Neonatal Lexi-Drugs facturers, the percentage differences between the macronutri-
(http://online.lexi.com). All websites were accessed multiple ents and micronutrients in human milk after fortification versus
times, and data were confirmed by both investigators and an the recommended intake2 are shown in Table 2. When human
assistant. milk was fortified to the recommended caloric intake of 130
kcal/kg for VLBW infants, HMF-A provided lower than the
recommended minimum quantity of protein, all fat-soluble
Results vitamins, all water-soluble vitamins, and Fe but higher than the
There are 3 manufacturers of HMF in the United States. One maximum recommended quantity of K, Cu, and Mn. However,
HMF is human milk based (HMF-A: Prolacta Bioscience, HMF-B provided lower than the recommended quantity of ino-
Monrovia, CA; www.prolacta.com) and is available only as a sitol, Na, and Fe but higher than the recommended quantity of
liquid preparation. Two HMFs are bovine milk protein based 2 fat-soluble vitamins, 8 water-soluble vitamins, Mg, P, Cu, and
(HMF-B and HMF-C) and are available as liquid and powder Mn. HMF-C provided lower than the recommended quantity of
preparations. HMF-B is from Ross Products (Abbott Lab, Mg but higher than the recommended quantity of protein, 2 fat-
Columbus, OH; www.abbottnutrition.com), and HMF-C is soluble vitamins, 5 water-soluble vitamins, K, and Mn. At the
from Mead Johnson Nutritional (Evansville, IN; www.enfamil. upper recommended caloric intake of 150 kcal/kg, HMF-C was
com). PTFs (PTF-B, PTF-C) from the aforementioned manu- the only HMF that achieved greater than the recommended
facturers also are recommended by the manufacturers as forti- minimum micronutrient intake, whereas excess intake of micro-
fiers for human milk. nutrients from all HMF became exaggerated (Table 2).
In addition to the recommended intake for enteral protein, Based on the maximum amount of HMF or the use of PTF
fat, and carbohydrate, there are recommendations for the to fortify human milk as recommended by manufacturers, the
enteral intake of 31 micronutrients,2 including 4 fat-soluble percentage differences between the fortified human milk and
vitamins, 9 water-soluble vitamins, 4 other organic com- the recommended intake2 for various macronutrients and
pounds, 8 trace minerals, and 3 electrolytes. However, differ- micronutrients are shown in Table 3. At the caloric intake of
ent manufacturers have 4–9 micronutrients absent from HMFs 130 kcal/kg, HMF-A provides protein content at slightly higher
or PTFs (Table 1). than the maximum recommended quantity, although the pat-
For infants with weights 1–1.5 kg, the recommended ranges tern of micronutrient deficiencies and excesses remains similar
of enteral intake/kg/d for energy (110–130 kcal), protein (3.4– to the use of standard volumes of HMF-A. However, the use of
4.2 g), fat (5.3–7.2 g), and carbohydrate (7–17 g) are slightly PTF-B provided lower than the recommended quantity of
lower than the recommended ranges for infants with weights protein, vitamins D and E, Na, and Fe but higher than the
Koo and Tice 3

Table 1.  AAPCON Recommended Intake of Micronutrients as Compared With the Composition of Commercial Human Milk
Fortifiers.

Commercial Liquid Human Milk Fortifiers


a
AAPCON Recommendation HMF-A HMF-B HMF-C PTF-B PTF-C
Fat-soluble vitamins A, D, E, K All All All All All
Water-soluble vitamins B1 (thiamin), B2 No biotin All All All All
(riboflavin), B3 (niacin), B5 (pantothenic
acid), B6, B7 (biotin), B9 (folate), B12, and C
Choline, inositol, carnitine, taurine None No carnitine or None No carnitine No carnitine or
taurine or taurine taurine
Calcium, magnesium, phosphorus All All All All All
Sodium, potassium, chloride All All All All All
Iron, zinc, copper, manganese, iodine Mo, Cr, I, Mo, Cr: absent Mo, Cr, I, Mo, Cr: absent Mo, Cr: absent
(I), selenium (Se), molybdenum (Mo), Se: absent Se: absent
chromium (Cr)

AAPCON, American Academy of Pediatrics Committee of Nutrition; HMF-A, human milk–based fortifier from Prolacta Bioscience (Monrovia, CA);
HMF-B, bovine milk protein–based fortifier from Ross Products (Abbott Lab, Columbus, OH); HMF-C, bovine milk protein–based fortifier from Mead
Johnson Nutritional (Evansville, IN); PTF-B and PTF-C, preterm infant formula at energy density of ≈1 kcal/mL from same manufacturers as HMF-B
and HMF-C, respectively.
a
Recommended enteral intake for very low birth weight infants.

recommended quantity of 5 water-soluble vitamins, K, Cu and Discussion


Mn. PTF-C provided lower than the recommended quantity of
protein, vitamins E and K, and 6 water-soluble vitamins. At the There are numerous potential benefits to exclusive human milk
upper recommended caloric intake of 150 kcal/kg, HMF-A feeding for VLBW infants,3 but its use alone, even at very high
provided higher than the maximum range of protein and fat but volumes, is associated with deficits in the intake of energy and
a similar pattern of micronutrient deficiencies and excesses multiple nutrients, which results in significant growth restric-
when compared with the lower energy intake of 130 kcal/kg. tion during an infant’s stay in the neonatal intensive care unit.
PTF fortification of human milk provided macronutrients at Much progress has been made to minimize the energy and
the recommended range or higher, while the extent of micronu- macronutrient deficit by starting parenteral nutrition within
trient deficiencies improved but the micronutrient excesses hours of birth and with early transition to enteral feeding. In
became exaggerated (Table 3). contrast, data to define the needs of many micronutrients are
The compositions of powder and liquid formulation of relatively limited, but sufficient data, albeit incomplete, are
HMF from the same manufacturer were the same. However, available to generate consensus on recommended intake for 31
the quantity of most nutrients for the powder HMF is some- micronutrients.1,2
what higher than the liquid HMF, and the pattern of differences The commercial availability of HMF provided a means to
of fortified milk from the recommended intake2 was similar minimize the deficit in energy, macronutrients, and micronutri-
regardless of the liquid or powder formulation used. ents for human milk–fed VLBW infants and is the standard of
There are 4 commercial infant multivitamin preparations care in neonative intensive care unit.3-5 However, postnatal
available: Poly-Vi-Sol with iron (Mead Johnson & Co, LLC, growth deficit continues despite the routine use of HMF and
Evansville, IN), Quflora (CarWin Pharmaceutical Associates, early transition to enteral feeding.8,9 Compared with the use of
LLC, Slidell, LA), Floriva Plus (BonGeo Pharmaceuticals, Inc, PTFs, the extent of growth deficit increases with increased vol-
Roseland, NJ), and AquADEKs (Actavis Pharma, Inc, ume of fortified human milk consumed by VLBW infants.8,9
Parsippany, NJ). When supplemented at 1 mL daily to the To our knowledge, this is the first systematic review of
human milk fortified with standard volume of HMF-A at 130 commercially available HMFs in the United States to deter-
kcal/kg, none of the preparations can correct all micronutrient mine whether their use in recommended quantities can meet
deficiencies, while 2 multivitamin products could not provide the recommended intake for specific nutrients.2 The liquid-
the minimum recommended intake for ≥1 water-soluble vita- based HMF was used as the principal source for comparison
mins. All preparations resulted in higher than the maximum among manufacturers because of its superiority in maintaining
recommended intake of multiple fat-soluble vitamins by 6%– sterility compared with powder HMF, although it limits the
3900%, water-soluble vitamins by 72%–610%, and mineral volume of human milk to about 80% of desired fluid goal due
and trace minerals by 43%–688% (Table 4). to the volume of displacement by liquid HMF. Comparison at
4
Table 2.  Percentage Difference in Macronutrients and Micronutrients in Fortified Human Milk Versus the Recommended Enteral Intake at 2 Isocaloric Levels (130 vs 150 kcal/
kg) Based on the Use of Standard Volume of Human Milk Fortifiers Recommended by the Manufacturers.a

Energy, 130 kcal/kg Energy, 150 kcal/kg

Nutrients HMF-A HMF-B HMF-C HMF-A HMF-B HMF-C


Fortifier:human milk, mL 5:20 5:25 5:25 5:20 5:25 5:25
b
Macronutrients
Protein −4 6 18 WR 22 36
Fat WR WR WR 7 WR 7
Carbohydrate WR WR WR WR WR WR
Micronutrientsc
Fat-soluble vitamins A, D, E, K A, –16; D, –71; E, A, 7; K, 34 A, 8; K, 3 A, –3; D, –66; E, A, 24; K, 55 A, 25; K, 19
–65; K, –95 –60; K, –94
Water-soluble vitamins B1, B2, B3, B5, B6, B7, absent; B1, –82; B1, 21; B2, 31; B3, B1, 8; B2, 8; B3, 8; B7, absent; B1, B1, 40; B2, 51; B3, B1, 25; B2, 25; B3,
B7, B9, B12, C B2, –66; B3, –92; 16; B5, 14; B6, B12, 195, C, 14 –79; B2, –61; B3, 34; B5, 32; B6, 61; 25; B5, 5; B6, 8;
B5, –71; B6, –83; 40; B7, 437; B12, –91; B5, –67; B6, B7, 520; B12, 200, B9, 5; B12, 240,
B9, –49; B12, –57, 160, C, 137 –81; B9, –41; B12, C, 173 C, 31
C, –23, –50, C, –12
Choline, inositol (Ino), carnitine (Car), Absent Car, Tau: absent; Absent Absent Car, Tau: absent Absent
taurine (Tau) Ino, –8
Calcium (Ca), magnesium (Mg), WR P, 18; Mg, 5 Mg, –13 Ca, 2 Ca, 4; P, 36; Mg, 21 WR
phosphorus (P)
Sodium (Na), potassium (K), chloride (Cl) K, 30 Na, –11; K, 64 K, 9 K, 50 K, 90 K, 26
Iron (Fe), zinc, copper (Cu), manganese Mo, Cr, I, Se: Mo, Cr: absent; Mo, Cr, I, Se: Mo, Cr, I, Se: Mo, Cr: absent; Mo, Cr, I, Se:
(Mn), iodine (I), selenium (Se), absent; Fe, –84; Fe, –62; Cu, 14; absent; Mn, 79 absent; Fe, –82; Fe, –56; Cu, 31; absent; Cu, 1;
molybdenum (Mo), chromium (Cr) Cu, 21; Mn, 109 Mn, 66 Cu, 40; Mn, 152 Mn, 92 Mn, 107

HMF-A, human milk–based fortifier from Prolacta Bioscience (Monrovia, CA); HMF-B, bovine milk protein–based fortifier from Ross Products (Abbott Lab, Columbus, OH); HMF-C, bovine milk
protein–based fortifier from Mead Johnson Nutritional (Evansville, IN); WR, within range of recommended intake.
a
Content of each nutrient is within the range recommended by American Academy of Pediatrics Committee of Nutrition2 unless there is a trailing number which indicates the percent above the maximum
or below the minimum (as indicated by negative symbol) recommended.
b
Intake of macronutrients versus the recommended intake2 for infants with weights <1 kg.
c
Intake of micronutrients versus the recommended intake2 for all infants with weights ≤1.5 kg.
Table 3.  Percentage Difference in Macronutrients and Micronutrients in Fortified Human Milk Versus the Recommended Enteral Intake at 2 Isocaloric Levels (130 vs 150 kcal/
kg) Based on the Use of Maximum Volume of Human Milk Fortifier or Preterm Infant Formula Recommended by the Manufacturers.a

Energy, 130 kcal/kg Energy, 150 kcal/kg

Nutrients HMF-A PTF-B PTF-C HMF-A PTF-B PTF-C


Fortifier:human milk, mL 50:50 50:50 50:50 50:50 50:50 50:50
Macronutrientsb
Protein 5 −9 −4 21 WR WR
Fat WR WR WR 12 13 WR
Carbohydrate WR WR WR WR WR WR
Micronutrientsc
Fat-soluble vitamins A, D, E, K A, –38; D, –45; E, D, –20; E, –32 E, –4; K, –10 A, –28; D, –37; E, D, –7; E, –22; K, 8 A, 4
–68; K, –95 –63; K, –94
Water-soluble vitamins B1, B2, B3, B5, B6, B7, B9, B7: absent; B1, –86; B2, 49; B6, 4; B7, B1, –4; B3, –10; B5, B7: absent; B1, B1, 2; B2, 72; B5, 6; B6, –1; B7, –1; B12,
B12, C B2, –69; B3, –93; 388; B12, 54; –10; B6, –14; B7, –84; B2, –64; B3, B6, 20; B7, 463; –10; C, 15
B5, –71; B6, –85; C, 55 –14; B12, –22 –92; B5, –67; B6, B12, 78; C, 78
B9, –24; B12, –44; –83; B9, –12; B12,
C, –61 –35; C, –55
Choline, inositol, carnitine (Car), taurine (Tau) Absent Car, Tau: absent Car, Tau: absent Absent Car, Tau: absent Car, Tau: absent
Calcium, magnesium, phosphorus WR WR WR WR WR WR
Sodium (Na), potassium (K), chloride (Cl) Cl, –14; K, 2 Na, –24; K, 24 WR Cl, –1; K, 18 Na, –12; K, 43 K, 13
Iron (Fe), zinc, copper (Cu), manganese (Mn), Fe, –81; Mn, 481; Fe, –25; Cu, 64; Fe, –25; Mo, Cr: Fe, –78; Cu, 11; Fe, –13; Cu, 89; Fe, –14; Cu, 10;
iodine (I), selenium (Se), molybdenum (Mo), Mo, Cr, I, Se: Mn, 29; Mo, Cr: absent Mn, 481; Mo, Cr, Mn, 49; Mo, Cr: Mn, 9; Mo, Cr:
chromium (Cr) absent absent I, Se: absent absent absent

HMF-A, human milk–based fortifier from Prolacta Bioscience (Monrovia, CA); PTF-B and PTF-C, preterm infant formula at energy density of ≈1 kcal/mL from Ross Products (Abbott Lab, Columbus,
OH) and Mead Johnson Nutritional (Evansville, IN), respectively; WR, within range of recommended intake.
a
Content of each nutrient is within the range recommended by American Academy of Pediatrics Committee of Nutrition2 unless there is a trailing number which indicates the percent above the maximum
or below the minimum (as indicated by negative symbol) recommended.
b
Intake of macronutrients versus the recommended intake2 for infants with weights <1 kg.
c
Intake of micronutrients versus the recommended intake2 for all infants with weights ≤1.5 kg.

5
6 Journal of Parenteral and Enteral Nutrition XX(X)

Table 4.  Micronutrient Content of Human Milk Fortified With Standard Volume of HMF-A to 130 and 150 kcal/kg and Supplemented
With 1 mL of Commercial Multivitamins.a

Compared With Recommendedb Energy, 130 kcal/kg Energy, 150 kcal/kg


Poly-Vi-Sol With Iron Contains Vitamins A, D, E, B1, B2, B3, B6, C, Iron
Lower None None
Higher B1 (122), B2 (90), B3 (72), B6 (102), C (103), D B1 (124), B2 (94), B3 (73), B6 (104), C
(11), iron (158) (112), D (13), iron (159)
Quflora Contains Vitamins A, D, E, B1, B2, B3, B6, B9, B12, C; Magnesium (Mg), Fluoride (F), Copper (Cu)
Lower B3 (70) B3 (69)
Higher A (6), B1 (122), B2 (90), B6 (102), B12 (610), C A (12), B1 (124), B2 (94), B6 (104), B12
(103), D (11), Mg (43), Cu (688), Fc (617), C (112), D (13), Mg (55), Cu
(707), Fc
Floriva Plus Contains Vitamins A, D, E, B1, B2, B3, B5, B6, B7, B9, B12, C; Fluoride (F)
Lower B3 (37), B7 (17) B3 (36), B7 (17)
Higher A (16), B1 (122), B2 (90), B6 (1817), B12 (610), C A (22), B1 (124), B2 (94), B6 (1819), B12
(91), D (11), Fc (617), C (100), D (13), Fc
AquADEKs Contains Vitamins A, D, E, K, B1, B2, B3, B5, B6, B7, C; Zinc (Zn), Selenium (Se)
Lower None None
Higher A (323), B1 (164), B2 (90), B3 (90), B5 (97), B6 A (329), B1 (166), B2 (94), B3 (32), B5
(198), B7 (150), C (145), D (61), E (334), K (100), B6 (200), B7 (150), C (154), D (63),
(3904), Zn (118), Se (222) E (337), K (3905), Zn (126), Se (222)

HMF-A, human milk-based fortifier from Prolacta Bioscience, Monrovia, CA.


a
Poly-Vi-Sol with iron from MeadJohnson & Co, LLC (Evansville, IN). Quflora from CarWin Pharmaceutical Associates, LLC (Slidell, LA). Floriva
Plus from BonGeo Pharmaceuticals, Inc (Roseland, NJ). AquADEKs from Actavis Pharma, Inc (Parsippany, NJ).
b
Number in parentheses indicates the percentage lower or higher than the respective minimum or maximum intake recommended by the American
Academy of Pediatrics Committee of Nutrition.2
c
Fluoride—no recommendation by the American Academy of Pediatrics Committee of Nutrition.

isocaloric levels provided a valid comparison of the quantita- apparently no regulatory mandate on the composition of HMF
tive difference in nutrient contents provided by different means or PTF. However, the extent of work needed to ensure nutrient
to fortify human milk. compatibility and bioavailability and to satisfy good manufac-
Our data indicate that at the isocaloric level of 130 kcal/kg, turing practices (https://www.fda.gov/food/guidanceregula-
all HMFs generally provide macronutrients in the recom- tion/cgmp/; accessed April 30, 2017) is likely dependent on the
mended range. However, when used to deliver lower total number and type of ingredients added to HMF and PTF. These
energy intake, there is a risk of delivering less than the mini- issues presumably contributed to the varied composition of
mum recommended protein intake, especially with the use of HMFs and PTFs among manufacturers.
human milk–based fortifier. The major deficiency with all The recommended range of intake for micronutrients is
HMFs is the inadequate provision of multiple micronutrients. much more varied when compared with that for macronutrient
None of the commercial HMFs provide all recommended intake, since there are limited data on the development of defi-
micronutrients, and human milk–based fortifier contains the ciency and toxicity at specific intakes. However, the recom-
least number of micronutrients, which results in the highest mended intake is consistent with the concept of reasonable
number of micronutrients delivered below the minimum rec- range of intake (RRI), defined as the range of average intakes
ommended intake. derived from observations or evaluated under controlled con-
The lack of multiple micronutrients (biotin, choline, inosi- ditions that appear to sustain adequate nutrition without clini-
tol, carnitine, taurine, molybdenum, iodine, selenium, or chro- cal, functional, or biochemical abnormalities.7 The lower RRI
mium) from HMF is of concern, since these nutrients are is generally the estimated average requirement, while the upper
essential or conditionally essential nutrients for VLBW RRI, if the upper tolerable nutrient intake is not available
infants.1,2,6,7 Unlike the regulatory guidelines for the manufac- (https://ods.od.nih.gov/pubs/conferences/tolerable_upper_
ture of infant formulas for term infants (https://www.fda.gov/ intake.pdf; accessed March 21, 2017), is based on an intake
food/guidanceregulation/guidancedocumentsregulatoryinfor- that is considered to be safe for medically stable VLBW
mation/ucm384451.htm; accessed April 30, 2017), there is infants. Thus, it is prudent to maintain the nutrient intake
Koo and Tice 7

within the range recommended by the American Academy of impairment. Thus, prolonged intake of any nutrient above
Pediatrics Committee of Nutrition.2 the maximum recommended range is to be avoided.
Clinical and biochemical adverse effects have been When higher volumes of human milk–based fortifier are
reported from deficiency in energy, macronutrients, and many used in a 1:1 ratio by volume with human milk as recom-
micronutrients including all fat-soluble vitamins, some water- mended by the manufacturer, it is possible to meet the desired
soluble vitamins, Fe, Zn, and Cu.1,7,10-13 Our data indicate that increase in energy, macronutrients, minerals, and electrolytes,
VLBW infants receiving less than the minimum recommended but its use cannot compensate for the lack of multiple other
intake are at risk of developing deficiency in multiple nutri- micronutrients. Additionally, this further lowers the volume of
ents, and it is unlikely that endogenous synthesis of some human milk with its associated decrease in potential benefits
micronutrients, such as vitamin D, or renal conservation of from bioactive and other factors4,17 present in human milk,
sodium can compensate for the extent of deficient intake although the extent of these benefits in human milk, especially
regardless of the HMF used. Because of the limited number of frozen mother’s milk and processed donor human milk,5,18
nutrients in HMFs, especially the human milk–based fortifier, remain ill defined. The potential for excess intake of Mn from
their chronic use might have contributed to the lower growth human milk–based fortifier could raise the risk for toxicity.
for VLBW infants fed “fortified” human milk8,9 and poten- The use of concentrated PTF up to 50% of volume of intake
tially other clinical or subclinical ill effects. lessens the extent of deficiency for micronutrients when com-
Our data demonstrate that excessive intake of macronu- pared with the use of the same volume of HMF. However, at
trients and micronutrients can occur with the use of any the usual caloric intake, the intake of protein, some fat-soluble
HMF, although it is more likely with the use of bovine milk and water-soluble vitamins, and iron remains lower than the
protein–based fortifiers. Most of the excess intake is within minimum quantity recommended. The number and extent of
50% of the maximum recommended intake. However, deficiency or excess of micronutrients vary between the 2
HMF-A provided Mn at up to 109%; HMF-B provided vita- PTFs with the risk for deficiency increasing at energy intakes
mins B7, B12, and C at 137%–437%; and HMF-C provided <130 kcal/kg/d. Our data also indicate that the use of any type
vitamin B12 at 195% above the maximum recommended of fortifier in quantities greater than the manufacturer’s recom-
intake.2 With an increase in energy intake to 150 kcal/kg, mendation should be avoided, since it may satisfy the energy
the excessive intake of nutrients is exaggerated—for exam- and macronutrient intake but runs the risk of greater imbal-
ple, the Mn intake from HMF-A, HMF-B, and HMF-C ances in micronutrient intake.
could increase up to 152%, 92%, and 107% above the maxi- The use of multivitamin supplements is a common practice
mum recommended intake. Although the tolerable upper in nutrition support for VLBW infants, and their use with iron
limits of intake for infants are not well defined, chronic is recommended for preterm infants fed human milk.3 However,
excess intake of certain nutrients, such as Mn and Cu, our data show that none of the commercial multivitamin for-
increases the risk for toxicity.7,10-13 In addition, excessive mulations provide all recommended vitamins and that they
intake of some nutrients may exacerbate the deficiency state lack multiple other micronutrients. Their use improves but
of another nutrient—for example, excessive Fe and polyun- does not eliminate the deficiency of micronutrients for those
saturated fatty acid intake can exacerbate vitamin E defi- receiving human milk–based fortifiers. For infants receiving
ciency, resulting in hemolytic anemia in VLBW infants.14 bovine milk protein–based fortifier, multivitamin supplements
Additionally, a modest increase in folate intake at twice the are of little use, since there is already an excess of multiple
recommended dietary allowance may adversely affect vita- vitamins. One of the bovine milk protein–based fortifiers con-
min B12-related function in elderly adults.15 tains less than the minimum recommended iron; therefore, iron
Nutrient content of infant formulas is typically higher supplementation is warranted. One multivitamin formulation
than the average level in human milk. This provides most designed for infants with hepatobiliary dysfunction contains
infants with at least as much as they would receive from large quantities of micronutrients, which should preclude its
human milk, and when coupled with the manufacturing pro- long-term use in otherwise stable and growing preterm infants.
cess of providing extra nutrients (overage) to maintain the One limitation of this study is the inability to take into
labeled content of nutrients throughout the shelf life,16 it is account all of the extensive biological variabilities of human
possible that the excessive intake of nutrients from the use of milk composition.1,3,5,18 However, the nutrient needs of VLBW
HMF and PTF is underestimated. It is possible that one may infants are much higher than the nutrient content present in
compensate for the excessive intake of any nutrient by unfortified human milk. To achieve appropriate nutrition sup-
decreasing its bioavailability from decreased intestinal port, it is probably more important to consider overall nutrient
absorption, increased hepatobiliary or renal excretion, or profile rather than individual nutrients. Chronic excessive or
increased metabolism before the storage capacity is over- deficient intake in ≥1 nutrients has the potential to adversely
whelmed with subsequent adverse effects. However, the influence metabolism with unintended consequences on
extent of these compensatory mechanisms in small preterm growth and development. Thus, the final nutrient content of
infants is limited, especially for those with hepatic or renal human milk fortified with various HMFs or PTFs provides a
8 Journal of Parenteral and Enteral Nutrition XX(X)

sound basis to reformulate the HMF to meet the VLBW infant’s safety, and usage options in the United States. Policy statement, American
need. The incomplete provision of all recommended nutrients Academy of Pediatrics. Pediatrics. 2017;139(1):e20163044.
6. Klein CJ. Nutrient requirements for preterm infant formulas. J Nutr.
in PTF (and HMF) presumably is due to the lack of informa- 2002;132(6)(suppl 1):1395s-577s.
tion on many of the currently recommended micronutrients 7. Uauy R, Koletzko B, Zlotkin SH. Concepts, definitions and approaches
when these products were initially formulated.6 to define the nutritional needs of LBW infants. In: Tsang RC, Uauy R,
We conclude that at the recommended energy intake for Tsang RC, Koletzko B, Zlotkin SH, eds. Nutrition of the Preterm Infant:
VLBW infants, commercial HMFs—when used according to Scientific Basis and Practice Guidelines. 2nd ed. Cincinnati, OH: Digital
Education Publishing Inc; 2005:1:21.
manufacturer’s recommendations—generally provide ade- 8. Maas C, Wiechers C, Bernhard W, Poets CF, Franz AR. Early feeding of
quate quantities of protein, fat, and carbohydrate. However, fortified breast milk and in-hospital-growth in very premature infants: a
many essential micronutrients are not present or are present in retrospective cohort analysis. BMC Pediatr. 2013;13:178.
grossly inadequate amounts, especially with human milk– 9. O’Connor DL, Gibbins S, Kiss A, et al. Effect of supplemental donor
based fortifier, while several micronutrients are present in human milk compared with preterm formula on neurodevelopment of very
low-birth-weight infants at 18 months: a randomized clinical trial. JAMA.
extremely high doses. The use of PTF and multivitamin formu- 2016;316:1897-1905.
lations do not fully compensate for the deficiencies and can 10. Koo W, Lulic-Botica M, Saba M, Warren L. Minerals. In: Corkins MR,
result in extremely high intake of some micronutrients. Since ed. The ASPEN Pediatric Nutrition Support Core Curriculum. 2nd ed.
supplement of individual nutrients is impractical, reformula- Silver Spring, MD: American Society for Parenteral and Enteral Nutrition;
tion of HMF to provide an improved nutrient profile is urgently 2015:57-68.
11. Koo W, Saba M, Lulic-Botica M, Christie J. Fat soluble vitamins.
needed. In: Corkins MR, ed. The ASPEN Pediatric Nutrition Support Core
Curriculum. 2nd ed. Silver Spring, MD: American Society for Parenteral
Statement of Authorship and Enteral Nutrition; 2015:91-106.
12. Koo W, Christie J, Saba M, Lulic-Botica M, Warren L. Water soluble
W. Koo contributed to the conception and design of the research;
essentials micronutrients. In: Corkins MR, ed. The ASPEN Pediatric
both authors equally contributed to the acquisition, analysis, and Nutrition Support Core Curriculum. 2nd ed. Silver Spring, MD: American
interpretation of data; and W. Koo drafted the manuscript. Both Society for Parenteral and Enteral Nutrition; 2015:69-90.
authors critically revised the manuscript, agree to be fully account- 13. Vanek VW, Borum P, Buchman A, et al. A call to action to bring safer
able for ensuring the integrity and accuracy of the work, and read parenteral micronutrient products to the US market. Nutr Clin Pract.
and approved the final manuscript. 2015;30:559-569.
14. Williams ML, Shoot RJ, O’Neal PL, Oski FA. Role of dietary iron and
fat on vitamin E deficiency anemia of infancy. N Engl J Med. 1975;
References 292:887-889.
1. Tsang RC, Uauy R, Tsang RC, Koletzko B, Zlotkin SH, eds. Nutrition 15. Brito A, Verdugo R, Hertrampf E, et al. Vitamin B-12 treatment of asymp-
of the Preterm Infant: Scientific Basis and Practice Guidelines. 2nd ed. tomatic, deficient, elderly Chileans improves conductivity in myelinated
Cincinnati, OH: Digital Education Publishing Inc; 2005. peripheral nerves, but high serum folate impairs vitamin B-12 status
2. American Academy of Pediatrics. Nutritional needs of the preterm infants. response assessed by the combined indicator of vitamin B-12 status. Am J
In: Kleinman RE, Greer FR, eds. Pediatric Nutrition. 7th ed. Elk Grove Clin Nutr. 2016;103:250-257.
Village, IL: American Academy of Pediatrics; 2014:83-121. 16. Hansen JW, Cook DA, Cordano A, Miguel SG. Human milk substitutes.
3. Section on Breastfeeding, American Academy of Pediatrics. Breastfeeding In: Tsang RC, Nichols BL, eds. Nutrition During Infancy. Philadelphia,
and the use of human milk. Pediatr. 2012;129:e827-e841. PA: Hanley & Belfus Inc; 1988:378-398.
4. American Academy of Pediatrics. Breast feeding. In: Kleinman RE, Greer 17. Ballard O, Morrow AL. Human milk composition: nutrients and bioactive
FR, eds. Pediatric Nutrition. 7th ed. Elk Grove Village, IL: American factors. Ped Clin North Amer. 2013;60:49-74.
Academy of Pediatrics; 2014:41-60. 18. Meier P, Patel A, Esquerra-Zwiers A. Donor human milk update: evi-
5. Committee on Nutrition, Section on Breastfeeding, Committee on Fetus dence, mechanisms, and priorities for research and practice. J Pediatr.
and Newborn. Donor human milk for the high risk infant: preparation, 2017;180:15-21.
Early trophic feeding versus enteral fasting for very preterm
or very low birth weight infants (Review)

Morgan J, Bombell S, McGuire W

This is a reprint of a Cochrane review, prepared and maintained by The Cochrane Collaboration and published in The Cochrane Library
2013, Issue 3
http://www.thecochranelibrary.com

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review)
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
TABLE OF CONTENTS
HEADER . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
ABSTRACT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
PLAIN LANGUAGE SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
BACKGROUND . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
OBJECTIVES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
METHODS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
RESULTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
DISCUSSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Figure 1. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
AUTHORS’ CONCLUSIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
ACKNOWLEDGEMENTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
CHARACTERISTICS OF STUDIES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
DATA AND ANALYSES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Analysis 1.1. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 1 Days to reach full enteral feeding. 21
Analysis 1.2. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 2 Incidence of necrotising
enterocolitis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Analysis 1.3. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 3 Mortality. . . . . . . . 23
Analysis 1.4. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 4 Days to regain birth weight. 24
Analysis 1.5. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 5 Incidence of invasive infection. 25
Analysis 1.6. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 6 Duration of phototherapy (days). 26
Analysis 1.7. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 7 Days of hospital stay. . . . 27
WHAT’S NEW . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
HISTORY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
CONTRIBUTIONS OF AUTHORS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
DECLARATIONS OF INTEREST . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
SOURCES OF SUPPORT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
INDEX TERMS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) i
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
[Intervention Review]

Early trophic feeding versus enteral fasting for very preterm


or very low birth weight infants

Jessie Morgan1 , Sarah Bombell2 , William McGuire1

1 HullYork Medical School & Centre for Reviews and Dissemination, University of York, York, UK. 2 Centre for Newborn Care,
Australian National University, Canberra, Australia

Contact address: William McGuire, Hull York Medical School & Centre for Reviews and Dissemination, University of York, York,
Y010 5DD, UK. William.McGuire@hyms.ac.uk.

Editorial group: Cochrane Neonatal Group.


Publication status and date: New search for studies and content updated (conclusions changed), published in Issue 3, 2013.
Review content assessed as up-to-date: 27 December 2012.

Citation: Morgan J, Bombell S, McGuire W. Early trophic feeding versus enteral fasting for very preterm or very low birth weight
infants. Cochrane Database of Systematic Reviews 2013, Issue 3. Art. No.: CD000504. DOI: 10.1002/14651858.CD000504.pub4.

Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

ABSTRACT

Background

The introduction of enteral feeds for very preterm (< 32 weeks) or very low birth weight (< 1500 grams) infants is often delayed due to
concern that early introduction may not be tolerated and may increase the risk of necrotising enterocolitis. However, prolonged enteral
fasting may diminish the functional adaptation of the immature gastrointestinal tract and extend the need for parenteral nutrition with
its attendant infectious and metabolic risks. Trophic feeding, giving infants very small volumes of milk to promote intestinal maturation,
may enhance feeding tolerance and decrease the time taken to reach full enteral feeding independently of parenteral nutrition.

Objectives

To determine the effect of early trophic feeding versus enteral fasting on feed tolerance, growth and development, and the incidence of
neonatal morbidity (including necrotising enterocolitis and invasive infection) and mortality in very preterm or VLBW infants.

Search methods

We used the standard search strategy of the Cochrane Neonatal Review Group. This included electronic searches of the Cochrane
Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2012, Issue 12), MEDLINE, EMBASE and CINAHL (1980
until December 2012), conference proceedings and previous reviews.

Selection criteria

Randomised or quasi-randomised controlled trials that assessed the effects of early trophic feeding (milk volumes up to 24 ml/kg/day
introduced before 96 hours postnatal age and continued until at least one week after birth) versus a comparable period of enteral fasting
in very preterm or very low birth weight infants.

Data collection and analysis

We extracted data using the standard methods of the Cochrane Neonatal Review Group with separate evaluation of trial quality and
data extraction by two authors and synthesis of data using risk ratio, risk difference and mean difference.
Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 1
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Main results

Nine trials in which a total of 754 very preterm or very low birth weight infants participated were eligible for inclusion. Few participants
were extremely preterm (< 28 weeks) or extremely low birth weight (< 1000 grams) or growth restricted. These trials did not provide
any evidence that early trophic feeding affected feed tolerance or growth rates. Meta-analysis did not detect a statistically significant
effect on the incidence of necrotising enterocolitis: typical risk ratio 1.07 (95% confidence interval 0.67 to 1.70); risk difference 0.01
(-0.03 to 0.05).

Authors’ conclusions

The available trial data do not provide evidence of important beneficial or harmful effects of early trophic feeding for very preterm or
very low birth weight infants. The applicability of these findings to extremely preterm, extremely low birth weight or growth restricted
infants is limited. Further randomised controlled trials would be needed to determine how trophic feeding compared with enteral
fasting affects important outcomes in this population.

PLAIN LANGUAGE SUMMARY

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

There is insufficient evidence to determine whether feeding very preterm or very low birth weight infants small quantities of milk during
the first week after birth (early trophic feeding) compared with fasting helps bowel development and improves subsequent feeding,
growth and development. Analysis of nine trials does not suggest that this practice increases the risk of a severe bowel disorder called
’necrotising enterocolitis’. Further trials could provide more robust evidence to inform this key area of care.

BACKGROUND
necrotising enterocolitis in very preterm or VLBW infants (Patole
2005). However, enteral fasting during the early neonatal period
also has potential disadvantages. Because gastrointestinal hormone
Description of the condition secretion and motility are stimulated by enteral milk, delayed en-
Necrotising enterocolitis is an important cause of morbidity and teral feeding could diminish the functional adaptation of the im-
mortality in very preterm (< 32 weeks) or very low birth weight mature gastrointestinal tract (Johnson 1976; Aynsley-Green 1983;
(VLBW: < 1500 grams) infants. Extremely low birth weight Berseth 1990). Consequent intestinal dysmotility may exacerbate
(ELBW: < 1000 grams) and extremely preterm (< 28 weeks) infants feed intolerance leading to a delay in establishing enteral feeding
are at highest risk (Rees 2007). Intrauterine growth restriction may independently of parenteral nutrition. Enteral fasting might also
be an additional specific risk factor, especially if associated with cause hyperbilirubinaemia by increasing enterohepatic recircula-
circulatory redistribution demonstrated by absent or reversed end- tion of bilirubin and delaying hepatic enzyme maturation. Pro-
diastolic flow velocities (AREDFV) in antenatal Doppler studies longing the duration of use of parenteral nutrition may be asso-
of the fetal aorta or umbilical artery (Bernstein 2000; Dorling ciated with infectious and metabolic complications that have ad-
2005). verse consequences for survival, duration of hospital stay, growth
Most very preterm or VLBW infants who develop necrotising en- and development (Flidel-Rimon 2004; Flidel-Rimon 2006).
terocolitis have received enteral milk feeds. Evidence exists that
feeding with formula milk rather than breast milk increases the risk
(Lucas 1990; Quigley 2007; Meinzen-Derr 2009). The timing of
the introduction of enteral feeding may also be an important mod-
Description of the intervention
ifiable risk factor for the development of necrotising enterocolitis Trophic feeding (also referred to as minimal enteral nutrition, gut
(Henderson 2009). Observational data suggest that feeding strate- priming and hypocaloric feeding) was developed and adopted into
gies that include delaying the introduction of progressive enteral clinical practice as an alternative to complete enteral fasting for
feeds until after five to seven days postnatally reduces the risk of very preterm or VLBW infants during the early neonatal period
Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 2
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
(Klingenberg 2012). Early trophic feeding is conventionally de- Types of interventions
fined as giving small volumes of milk (typically 12 to 24 ml/kg/ Early trophic feeding: enteral feeding with milk volumes up to 24
day) intragastrically starting within the first few days after birth, ml/kg/day (1 ml/kg/hour) beginning within four days after birth
without advancing the feed volumes during the first week postna- and continued for at least five days or until at least one week after
tally (McClure 2001). The primary aim of trophic feeding is to birth versus enteral fasting for the same period.
accelerate gastrointestinal physiological, endocrine and metabolic Once progressive enteral feeding has started, infants should have
maturity and so allow infants to transition to full enteral feeding received the same type of milk (breast milk or formula), the same
independent of parenteral nutrition more quickly. However, any route and mode of feeding (intragastric or transpyloric, bolus gav-
beneficial effects may be negated if early trophic feeding increases age or continuous) and the same rate of feed volume advancement
the risk of necrotising enterocolitis in very preterm or VLBW in- in both groups.
fants.

Types of outcome measures


Why it is important to do this review
This review focuses on the question of whether early trophic feed-
ing compared with a similar period of enteral fasting improves Primary outcomes
feed tolerance without increasing the risk of necrotising entero- 1. Feed intolerance: days to establish full enteral feeding
colitis in very preterm or VLBW infants. Other Cochrane reviews independently of parenteral nutrition.
address the questions of whether introducing progressive enteral 2. Necrotising enterocolitis confirmed by at least two of the
milk feeds (beyond trophic volumes) later or slowing the rate of following features:
advancement of feed volumes affects the risk of necrotising entero- • abdominal radiograph showing pneumatosis intestinalis or
colitis, mortality and other morbidities (Morgan 2011a; Morgan gas in the portal venous system or free air in the abdomen;
2011b). • abdominal distension with abdominal radiograph with
gaseous distension or frothy appearance of bowel lumen (or
both);
• blood in stool;
• lethargy, hypotonia or apnoea (or combination of these);
OBJECTIVES
or a diagnosis confirmed at surgery or autopsy (Walsh 1986).
To determine the effect of early trophic feeding versus enteral fast-
ing on feed tolerance, growth and development, and the incidence
of neonatal morbidity (including necrotising enterocolitis and in- Secondary outcomes
vasive infection) and mortality in very preterm or VLBW infants.
1. All-cause mortality prior to hospital discharge.
2. Growth: (i) Time to regain birth weight and rates of weight
gain, linear growth, head growth or skinfold thickness growth up
METHODS to six months of age corrected for preterm birth; (ii) Long-term
growth: weight, height or head circumference and/or proportion
of infants who remain below the 10th percentile for the index
population’s distribution assessed at intervals from six months of
Criteria for considering studies for this review age.
3. Neurodevelopment: death or severe neurodevelopmental
disability defined as any one or combination of the following:
Types of studies non-ambulant cerebral palsy, developmental delay
(developmental quotient less than 70), auditory and visual
Randomised or quasi-randomised controlled trials including clus- impairment. Each component will be analysed individually as
ter-randomised trials. well as part of the composite outcome.
4. Incidence of invasive infection as determined by culture of
bacteria or fungus from blood, cerebrospinal fluid, urine or from
Types of participants a normally sterile body space.
VLBW (< 1500 grams) or very preterm (< 32 weeks) newborn 5. Duration of phototherapy for hyperbilirubinaemia (days).
infants. 6. Duration of hospital stay (days).

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 3
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Search methods for identification of studies Data extraction and management
We used the standard search strategy of the Cochrane Neonatal We used a data collection form to extract relevant information
Group (http://neonatal.cochrane.org/). from each included study. Two review authors extracted the data
separately. We discussed any disagreements with the third author
until we reached consensus.
Electronic searches
We searched the Cochrane Central Register of Controlled Trials
(CENTRAL, The Cochrane Library 2012, Issue 12), MEDLINE Assessment of risk of bias in included studies
(1980 to December 2012), EMBASE (1980 to December 2012) We used the criteria and standard methods of the Cochrane
and CINAHL (1982 to December 2012) using the following text Neonatal Review Group to assess the methodological quality of
words and MeSH terms: [Infan*, OR Infant/, OR Preterm, OR any included trials. We requested additional information from the
Prem*, OR Infant premature/, OR Neonat*, OR New ADJ born, trial authors to clarify methodology and results as necessary. We
OR New?born, Infant newborn/, OR Very Low Birth Weight, OR evaluated and reported the following issues in the ’Risk of bias’
VLBW, OR Extremely Low Birth Weight, OR ELBW, OR Infant tables:
Very Low Birth Weight/ OR Infant Extremely Low Birth Weight/ 1. Sequence generation: We categorised the method used to
] AND [Breast feeding, OR Breast feeding/, OR human milk, OR generate the allocation sequence as:
human milk/, OR formula, Infant formula/, OR Trophic feeding, i) low risk: any random process e.g. random number
OR minimal enteral nutrition, OR MEN, OR minimal enteral table; computer random number generator;
feeding, OR MEF, OR gut priming, OR enteral feed*, OR enteral ii) high risk: any non random process e.g. odd or even
nutrition/]. date of birth; patient case-record number;
The search outputs were limited with the relevant search filters for iii) unclear.
clinical trials. We did not apply any language restriction. 2. Allocation concealment: We categorised the method used
We searched ClinicalTrials.gov and Current Controlled Trials for to conceal the allocation sequence as:
completed or ongoing trials. i) low risk: e.g. telephone or central randomisation;
consecutively numbered sealed opaque envelopes;
ii) high risk: open random allocation; unsealed or non-
Searching other resources opaque envelopes, alternation; date of birth;
We examined reference lists in previous reviews and studies. iii) unclear.
We examined the references in studies identified as potentially rel- 3. Blinding: We assessed blinding of participants, clinicians
evant. We also searched the abstracts from the annual meetings and care givers, and outcome assessors separately for different
of the Pediatric Academic Societies (1993 to 2012), the European outcomes and categorised the methods as:
Society for Pediatric Research (1995 to 2012), the UK Royal Col- i) low risk;
lege of Paediatrics and Child Health (2000 to 2012) and the Peri- ii) high risk;
natal Society of Australia and New Zealand (2000 to 2012). We iii) unclear.
considered trials reported only as abstracts to be eligible if suffi- 4. Incomplete outcome data: We described the completeness
cient information was available from the report, or from contact of data including attrition and exclusions from the analysis for
with the authors, to fulfil the inclusion criteria. each outcome and any reasons for attrition or exclusion where
reported. We assessed whether missing data were balanced across
groups or were related to outcomes. Where sufficient information
was reported or supplied by the trial authors, we re-included
Data collection and analysis missing data in the analyses. We categorised completeness as:
We used the standard methods of the Cochrane Neonatal Review i) low risk: < 20% missing data;
Group (http://neonatal.cochrane.org/). ii) high risk: > 20% missing data;
iii) unclear.

Selection of studies
Two review authors screened the title and abstract of all studies Measures of treatment effect
identified by the above search strategy. We reassessed the full text We calculated risk ratio (RR) and risk difference (RD) for dichoto-
of any potentially eligible reports and excluded those studies that mous data and mean difference (MD) for continuous data, with
did not meet all of the inclusion criteria. Review authors discussed respective 95% confidence intervals (CI). We used a fixed-effect
any disagreements until consensus was achieved. model for meta-analysis.

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 4
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Assessment of heterogeneity VLBW. Most of the other trials specifically excluded infants who
We examined the treatment effects of individual trials and hetero- were small for gestational age at birth and infants with congenital
geneity between trial results by inspecting the forest plots if more anomalies, gastrointestinal problems or neurological problems.
than one trial was included in a meta-analysis. We calculated the
I² statistic for statistical heterogeneity. If substantial (I² > 50%)
heterogeneity was detected, we explored the possible causes (for Interventions
example, differences in study design, participants, interventions Trophic feeding was generally started within the first three days
or completeness of outcome assessments) in sensitivity analyses. after birth and continued for varying durations; either until in-
fants were judged to be clinically stable (for example following
endotracheal extubation or removal of umbilical catheters) or for
Subgroup analysis and investigation of heterogeneity pre-defined intervals, generally 7 to 10 days after birth. Feeding
We planned the following subgroup analyses: volumes ranged from about 12 to 24 ml/kg/day. One trial admin-
1. trials in which most infants were exclusively formula-fed; istered milk at a rate of 25 ml/kg/day with no intention to increase
2. trials in which most infants were at least partially fed with this volume for six to eight days (Becerra 1996). Although this
human milk (maternal or donor); rate exceeded our definition of minimal enteral nutrition by 1 ml/
3. trials in which most participants were of ELBW (< 1000 kg/day, we made a consensus decision to include the trial.
grams) or extremely preterm (< 28 weeks); In most trials, infants received either expressed breast milk or for-
4. trials in which participants were infants with intrauterine mula milk (diluted or full-strength) or a mixture of breast milk and
growth restriction, or infants with absent or reversed end- formula. In two trials, infants received only formula milk (Dunn
diastolic flow velocities detected on antenatal Doppler studies of 1988; Meetze 1992). Control infants received no enteral nutrition
the fetal aorta or umbilical artery. for at least one week after birth. Infants in both comparison groups
received standard parenteral nutrition during the trial period.
In most trials, milk was administered by intermittent bolus gavage
feeds via oro or nasogastric tube. In Schanler 1999, participating
infants were also allocated to either bolus or continuous feeding
RESULTS
using a factorial design. In Troche 1995, infants weighing < 800
grams at birth received feeds via a continuous infusion whereas
those weighing > 800 grams at birth received intermittent bolus
Description of studies feeds.
See: Characteristics of included studies; Characteristics of excluded
studies.
We identified 17 articles using the above search strategy. Outcomes
Most trials assessed feed intolerance (variously defined) and inci-
dence of necrotising enterocolitis. Short-term growth parameters
Included studies were reported in a variety of ways, most commonly time to regain
Nine trials fulfilled the inclusion criteria (Dunn 1988; Meetze birth weight and weight gain during the neonatal period (either
1992; Troche 1995; Becerra 1996; Schanler 1999; McClure 2000; as median and range or as mean and standard deviation). Most
Sáenz de Pipaón 2003; van Elburg 2004; Mosqueda 2008; see reports also gave information on adverse outcomes including mor-
table ’Characteristics of included studies’). tality. None of the trials reported long-term growth and neurode-
velopmental outcomes for surviving infants.

Participants
The included studies were all undertaken since the late 1980s Excluded studies
by investigators attached to neonatal units in Europe and North We excluded eight studies (LaGamma 1985; Ostertag 1986; Slagle
America. Most were small single-centre studies. 754 infants par- 1988; Berseth 1992; Berseth 1993; Berseth 2003; Weiler 2006;
ticipated in total (range 29 to 190). Most participants were ap- Said 2008; see table ’Characteristics of excluded studies’).
propriate-for-gestational age VLBW or very preterm infants re-
ceiving standard intensive care interventions such as mechanical
ventilation and parenteral nutrition. In van Elburg 2004, partici-
pants were infants of birth weight less than 2000 grams who were
Risk of bias in included studies
small for gestational age (< 10th percentile for birth weight). We Most of the trials had some methodological weaknesses. In four
included this study because > 80% of participating infants were trials it was unclear whether allocation was concealed. Care givers

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 5
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
were not blinded to treatment group in any trial. Few trials un- • Weight: reported MD 130 (95% CI 1 to 250) grams/week.
dertook blinded assessments for any of the outcomes, and several • Head circumference: reported MD 0.7 (95% CI 0.1 to 1.3)
of the trials did not include results for all infants randomised (see cm/week
table ’Characteristics of included studies’).
Mosqueda 2008 reported no statistically significant difference in
rates of weight gain during the trial period: MD -7.3 (95% CI -
19.2 to 4.6) grams/week.
Effects of interventions
Sáenz de Pipaón 2003 reported that the weight above birth weight
attained by day 21 was not statistically significantly different (188
Primary outcomes grams versus 190 grams).
Troche 1995 reported that infants in the trophic feeding group
had a higher increase in weight over birth weight to day 30 (223
Feed intolerance: time to establish full enteral feeding (SD 125) versus 95 (SD 161) grams).
(outcome 1.1; eight trials) Meetze 1992 reported no statistically significant difference in
Meta-analysis of data from six trials that reported mean and stan- weight gain between the groups at day 30: 264 (SD 126) grams ver-
dard deviation (SD) did not detect a statistically significant effect: sus 213 (SD 142) grams. Increases in head circumference, length
mean difference (MD) -1.05 (95% confidence interval (CI) -2.61 and mid-arm circumference were reported to be similar for both
to 0.51) days. The meta-analysis contained significant statistical groups.
heterogeneity in (I² = 73%) (Analysis 1.1). Dunn 1988 measured growth throughout the study period up
Two trials reported median and range data. Neither detected a sta- until 60 days of life and did not detect any significant differences
tistically significant difference: 32 days versus 32 days (Mosqueda between the two groups.
2008); 13 days versus 13 days (van Elburg 2004). Long-term growth parameters were not reported by any of the
trials.

Necrotising enterocolitis (outcome 1.2; nine trials)


Meta-analysis did not detect a statistically significant effect: typical Neurodevelopment
risk ratio (RR) 1.07 (95% CI 0.67 to 1.70); typical risk difference None of the trials assessed neurodevelopmental outcomes.
(RD) 0.01 (95% CI -0.03 to 0.05). There was no evidence of
heterogeneity (I² = 0%) (Analysis 1.2).
Incidence of invasive infection (outcome 1.5; four trials)
Meta-analysis of three trials did not detect a statistically significant
Secondary outcomes
difference: typical RR 1.06 (95% CI 0.72 to 1.56); typical RD 0.02
(95% CI -0.10 to 0.13). There was no evidence of heterogeneity
Mortality (outcome 1.3; eight trials) (I² = 25%) (Analysis 1.5).
Meta-analysis did not detect a statistically significant effect: typical McClure 2000 reported that infants in the minimal enteral nutri-
RR 0.66 (95% CI 0.41 to 1.07); typical RD -0.04 (95% CI - tion group had a statistically significantly lower mean number of
0.10 to 0.01). There was no evidence of heterogeneity (I² = 0%) episodes of “culture-confirmed sepsis” (0.5 versus 1.2 in control
(Analysis 1.3). group). These data could not be included in the meta-analysis.

Growth (outcome 1.4; eight trials) Duration of phototherapy (days) (outcome 1.6; three trials)
None of the trials reported a statistically significant difference in Meta-analysis did not detect a statistically significant effect: MD
the time to regain birth weight. Meta-analysis of five trials with data 0.35 (95% CI -0.29 to 0.99) days (Analysis 1.6).
as mean and SD: MD -0.01 (95% CI -0.96 to 0.95) days. There
was no evidence of statistical heterogeneity (I² = 23%) (Analysis
Duration of hospital stay (outcome 1.7; five trials)
1.4).
Two trials reported median and range data. Neither detected a sta- Meta-analysis of four trials that reported data as mean and SD
tistically significant difference: 13 days versus 12 days (Mosqueda did not detect a statistically significant effect: MD -3.9 (95% CI -
2008); 11 days versus 10 days (van Elburg 2004). 11.5 to 3.8) days (Analysis 1.7). There was evidence of borderline
McClure 2000 reported that the average rate of weight gain and statistical heterogeneity (I² = 48%).
head circumference gain during the six weeks after birth was bor- One trial that reported median and range data did not find a statis-
derline significantly higher in infants who had received trophic tically significant difference: 81 days versus 79.5 days (Mosqueda
feeds: 2008).

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 6
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Subgroup analyses substantial statistical heterogeneity in the meta-analysis limits the
1. Exclusively formula milk-fed infants: In two trials, infants validity of this finding. The heterogeneity was not explained by
received only formula milk as trophic feeds (Dunn 1988; Meetze differences between trials in methodological quality or the type of
1992). In the other trials, infants received either breast milk or intervention or participants. It may be that variations in enteral
formula milk or a mixture. Subgroup data were not available. feeding protocols and practices contributed to heterogeneity.
2. Infants at least partially fed with breast milk: Subgroup data These findings may not be applicable to some infants at high-
were not available. est risk of developing feed intolerance or necrotising enterocol-
3. Extremely low birth weight (ELBW) or extremely preterm itis. Only a minority of participants in the included trials were
infants: One trial restricted participation to ELBW infants extremely low birth weight (ELBW) or extremely preterm infants
(Mosqueda 2008). In the other trials, it is likely that less than or had evidence of intrauterine growth restriction. None of the
one-third of all participants were ELBW or extremely preterm trials specifically recruited infants with absent or reversed end-
but subgroup data were not available. diastolic flow velocities on Doppler ultrasound of the umbilical
4. Infants with intrauterine growth restriction or infants with arteries. Furthermore, the risk-benefit balance of enteral feeding
absent or reversed end-diastolic flow velocities (AREDFV): In strategies may differ between breast milk-fed and formula-fed very
those trials where birth weight < 10th percentile was not an preterm or VLBW infants. One study reported that mothers who
exclusion criterion, subgroup data were not available. One trial expressed breast milk for early trophic feeding were more likely to
restricted participation to infants who were small for gestational continue to provide breast milk as the ongoing principal form of
age (birth weight < 10th percentile for reference population) nutrition for their infants (Schanler 1999). Further study to con-
(van Elburg 2004). firm and define the mechanism of this association is merited given
that feeding with breast milk compared to formula reduces the
risk of necrotising enterocolitis in very preterm or VLBW infants
(Quigley 2007).
It is also unclear whether the findings can be applied to infants
DISCUSSION who receive continuous infusion of milk feeds as all of the infants
in the included trials received enteral feeds as interval boluses. A
recently described issue is that bolus administration of volumes
Summary of main results up to 0.5 ml results in substantial retention of milk within stan-
dard gastric feeding tubes (which will then be aspirated prior to
The available data from randomised controlled trials do not pro- the next feed). Consequently, infants will not actually receive any
vide evidence that early trophic feeding compared to enteral fast- milk intragastrically unless trophic feeding is delivered continu-
ing confers any substantial benefits for very preterm or very low ously (McHale 2010). Randomised controlled trials have reported
birth weight (VLBW) infants. Although some trials reported that conflicting findings about the effect on continuous enteral infu-
minimal enteral nutrition reduced the time taken to establish full sion on feed tolerance in very preterm and VLBW infants (Premji
enteral feeds, meta-analysis of all of the available data did not de- 2011).
tect a statistically significant effect.
The trial data do not suggest that minimal enteral nutrition is
associated with important harms. Meta-analyses did not detect
Quality of the evidence
statistically significant effects on the incidence of necrotising en-
terocolitis, invasive infection or all-cause mortality. Only limited The included trials were generally of good methodological qual-
data on growth outcomes were found. Trials found inconsistent ity but in common with other trials of feeding interventions in
effects on short-term growth and meta-analysis did not reveal a this population it was not possible to mask care givers and clin-
significant difference in the time taken to regain birth weight. The ical assessors to the nature of the intervention (Figure 1). This
clinical importance of any short-term effects is unclear as no long- may be an important source of bias particularly in trials that did
term growth or developmental outcomes were assessed. not use prespecified definitions of feed intolerance that mandated
interrupting or ceasing feed volume advancement. Care givers or
clinicians who were aware of the treatment group may have de-
fined feed intolerance subjectively and differentially. Any surveil-
Overall completeness and applicability of lance and ascertainment biases secondary to the lack of blinding
evidence are more likely to have caused an over-estimation of the incidence
These findings should be applied with caution. Although we did of feed intolerance or necrotising enterocolitis in infants who re-
not find evidence of an effect on feed intolerance, the existence of ceived minimal enteral nutrition.

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 7
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Figure 1. ’Risk of bias’ graph: review authors’ judgements about each risk of bias item presented as
percentages across all included studies.

so that findings are applicable to these infants at highest risk of


necrotising enterocolitis. Undertaking trials of feeding interven-
tions in this population is problematic (Tyson 2007). It is diffi-
AUTHORS’ CONCLUSIONS
cult to perform a pragmatic trial that will ensure that care givers
Implications for practice
and investigators are unaware of the allocated feeding regimen. A
The available trial data do not provide strong evidence that early priori agreements on objective definitions of feed intolerance and
trophic feeding has important effects on feed intolerance, growth indications for interruption of enteral feeding and for investiga-
or development. There is no evidence that trophic feeding has ad- tion of necrotising enterocolitis may help minimise the impact of
verse effects. For necrotising enterocolitis, the lower and bounds this source of bias. Trials should also aim to assess more objective
of the 95% CI of the number needed to treat for an additional outcomes, principally mortality and long-term growth and devel-
harmful outcome (NNTH) estimate are consistent with either five opment.
more cases or three fewer cases in every 100 infants who receive
early trophic feeding. For mortality, the NNTH 95% CI is con-
sistent with one more case or 10 fewer cases in every 100 infants
who receive early trophic feeding.
ACKNOWLEDGEMENTS
Implications for research We thank Dr Schanler for providing further data from Schanler
1999 and Dr Sáenz de Pipaón for clarification on data from Sáenz
Any new randomised controlled trials of early trophic feeding ver-
de Pipaón 2003.
sus enteral fasting should aim to ensure the participation of ex-
tremely low birth weight (ELBW) and extremely preterm infants as We are grateful to Ms Bethan Carter for developing and running
well as infants with evidence of compromised intrauterine growth the electronic search.

REFERENCES

References to studies included in this review gastrointestinal function: preliminary report of a


randomized trial. Journal of Pediatrics 1988;112(4):622–9.
Becerra 1996 {published and unpublished data}
Becerra M, Ambiado S, Kuntsman G, Figueroa A, Balboa McClure 2000 {published and unpublished data}
P, Fernandez P, et al.Feeding VLBW infants; Effect of early ∗
McClure RJ, Newell SJ. Randomised controlled trial of
enteral stimulation (EES) [abstract]. Pediatric Research clinical outcome following trophic feeding. Archives of
1996;39:304A. Disease in Childhood. Fetal and Neonatal Edition 2000;82
Dunn 1988 {published data only} (1):F29–F33.
Dunn L, Hulman S, Weiner J, Kleigman R. Beneficial McClure RJ, Newell SJ. Randomised controlled trial of
effects of early hypocaloric enteral feeding on neonatal trophic feeding and gut motility. Archives of Disease in
Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 8
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Childhood. Fetal and Neonatal Edition 1999;80(1):F54–8. Berseth 2003 {published and unpublished data}
McClure RJ, Newell SJ. Randomized controlled study of Berseth CL, Bisquera JA, Paje VU. Prolonging small feeding
digestive enzyme activity following trophic feeding. Acta volumes early in life decreases the incidence of necrotizing
Paediatrica 2002;91(3):292–6. enterocolitis in very low birth weight infants. Pediatrics
Meetze 1992 {published data only} 2003;111(3):529–34.
Meetze WH, Valentine C, McGuigan JE, Conlon M, LaGamma 1985 {published data only}
Sacks N, Neu J. Gastrointestinal priming prior to full LaGamma EF, Ostertag S, Birenbaum H. Failure of delayed
enteral nutrition in very low birth weight infants. Journal oral feedings to prevent necrotizing enterocolitis. American
of Pediatric Gastroenterology and Nutrition 1992;15(2): Journal of Diseases of Children 1985;139(4):385–9.
163–70. Ostertag 1986 {published data only}
Mosqueda 2008 {published data only} Ostertag SG, LaGamma EF, Reisen CE, Ferrentino FL.
Mosqueda E, Sapiegiene L, Glynn L, Wilson-Costello Early enteral feeding does not affect the incidence of
D, Weiss M. The early use of minimal enteral nutrition necrotizing enterocolitis. Pediatrics 1986;77(3):275–80.
in extremely low birth weight newborns. Journal of
Said 2008 {published data only}
Perinatology 2008;28(4):264–9.
Said H, Elmetwally D, Said D. Randomised controlled trial
Sáenz de Pipaón 2003 {published and unpublished data} of early versus late enteral feeding of prematurely born
Sáenz de Pipaón M, VanBeek RH, Quero J, Perez J, infants with birth weight <1200 grams. Kasr El Aini Medical
Wattimena DJ, Sauer PJ. Effect of minimal enteral feeding Journal 2008;14:1–10.
on splanchnic uptake of leucine in the postabsorptive state
Slagle 1988 {published data only}
in preterm infants. Pediatric Research 2003;53(2):281–7.
Slagle TA, Gross SJ. Effect of early low-volume enteral
Schanler 1999 {published and unpublished data} substrate on subsequent feeding tolerance in very low birth
Schanler RJ, Shulman RJ, Lau C, Smith EO, Heitkemper weight infants. Journal of Pediatrics 1988;113(3):526–31.
MM. Feeding strategies for premature infants: randomized
Weiler 2006 {published data only}
trial of gastrointestinal priming and tube-feeding method.
Weiler HA, Fitzpatrick-Wong SC, Schellenberg JM, Fair
Pediatrics 1999;103(2):434–9.
DE, McCloy UR, Veitch RR, et al.Minimal enteral feeding
Shulman RJ, Schanler RJ, Lau C, Heitkemper M, Ou C,
within 3 d of birth in prematurely born infants with birth
Smith EO. Early feeding, antenatal glucocorticoids, and
weight < or = 1200 g improves bone mass by term age.
human milk decrease intestinal permeability in preterm
American Journal of Human Nutrition 2006;83(1):155–62.
infants. Pediatric Research 1998;44(4):519–23.
Shulman RJ, Schanler RJ, Lau C, Heitkemper M, Ou C, Additional references
Smith EO. Early feeding, feeding tolerance, and lactase
activity in preterm infants. Journal of Pediatrics 1998;133 Aynsley-Green 1983
(5):645–9. Aynsley-Green A. Hormones and postnatal adaptation to
Troche 1995 {published data only} enteral nutrition. Journal of Pediatric Gastroenterology and
Troche B, Harvey-Wilkes K, Engle WD, Nielsen HC, Nutrition 1983;2(3):418–27.
Frantz ID, Mitchell ML, et al.Early minimal feedings Bernstein 2000
promote growth in critically ill premature infants. Biology of Bernstein IM, Horbar JD, Badger GJ, Ohlsson A, Golan
the Neonate 1995;67(3):172–81. A. Morbidity and mortality among very-low-birth-weight
van Elburg 2004 {published data only} neonates with intrauterine growth restriction. The Vermont
van Elburg RM, van den Berg A, Bunkers CM, van Oxford Network. American Journal of Obstetrics and
Lingen RA, Smink EW, van Eyck J, et al.Minimal enteral Gynecology 2000;182(1 Pt 1):198–206.
feeding, fetal blood flow pulsatility, and postnatal intestinal Berseth 1990
permeability in preterm infants with intrauterine growth Berseth CL. Neonatal small intestinal motility: the motor
retardation. Archives of Disease in Childhood, Fetal and responses to feeding in term and preterm infants. Journal of
Neonatal Edition 2004;89(4):F293–6. Pediatrics 1990;117(5):777–82.
Dorling 2005
References to studies excluded from this review
Dorling J, Kempley S, Leaf A. Feeding growth restricted
Berseth 1992 {published data only} preterm infants with abnormal antenatal Doppler results.
Berseth CL. Effect of early feeding on maturation of the Archives of Disease in Childhood. Fetal and Neonatal Edition
preterm infant’s small intestine. Journal of Pediatrics 1992; 2005;90(5):F359–63.
120(6):947–53. Flidel-Rimon 2004
Berseth 1993 {published data only} Flidel-Rimon O, Friedman S, Lev E, Juster-Reicher
Berseth CL, Nordyke C. Enteral nutrients promote A, Amitay M, Shinwell ES. Early enteral feeding and
postnatal maturation of intestinal motor activity in preterm nosocomial sepsis in very low birthweight infants. Archives
infants. American Journal of Physiology 1993;264(6 Pt 1): of Disease in Childhood. Fetal and Neonatal Edition 2004;89
G1046–51. (4):289–92.

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 9
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Flidel-Rimon 2006 Patole 2005
Flidel-Rimon O, Branski D, Shinwell ES. The fear of Patole SK, de Klerk N. Impact of standardised feeding
necrotizing enterocolitis versus achieving optimal growth in regimens on incidence of neonatal necrotising enterocolitis:
preterm infants--an opinion. Acta Paediatrica 2006;95(11): a systematic review and meta-analysis of observational
1341–4. studies. Archives of Disease in Childhood. Fetal and Neonatal
Edition 2005;90(2):147–51.
Henderson 2009
Henderson G, Craig S, Brocklehurst P, McGuire W. Enteral Premji 2011
feeding regimens and necrotising enterocolitis in preterm Premji SS, Chessell L. Continuous nasogastric milk feeding
infants: a multicentre case-control study. Archives of Disease versus intermittent bolus milk feeding for premature
in Childhood. Fetal and Neonatal Edition 2009;94(2): infants less than 1500 grams. Cochrane Database of
F120–3. Systematic Reviews 2011, Issue 11. [DOI: 10.1002/
14651858.CD001819.pub2]
Johnson 1976
Quigley 2007
Johnson CR. The trophic action of gastrointestinal
Quigley MA, Henderson G, Anthony MY, McGuire
hormones. Gastroenterology 1976;70(2):278–88.
W. Formula milk versus donor breast milk for feeding
Klingenberg 2012 preterm or low birth weight infants. Cochrane Database
Klingenberg C, Embleton ND, Jacobs SE, O’Connell LAF, of Systematic Reviews 2007, Issue 4. [DOI: 10.1002/
Kuschel CA. Enteral feeding practices in very preterm 14651858.CD002971.pub2]
infants: an international survey. Archives of Disease in Rees 2007
Childhood. Fetal and Neonatal Edition 2012;97(1):F56–61. Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes
of neonates with medically and surgically treated necrotizing
Lucas 1990
enterocolitis. Archives of Disease in Childhood. Fetal and
Lucas A, Cole TJ. Breast milk and neonatal necrotising
Neonatal Edition 2007;92(3):193–8.
enterocolitis. Lancet 1990;336(8730):1519–23.
Tyson 2007
McClure 2001 Tyson JE, Kennedy KA, Lucke JF, Pedroza C. Dilemmas
McClure RJ. Trophic feeding of the preterm infant. Acta initiating enteral feedings in high risk infants: how can they
Paediatrica. Supplement 2001;90(436):19–21. be resolved?. Seminars in Perinatology 2007;31(2):61–73.
McHale 2010 Walsh 1986
McHale SM, McCarthy R, O’Donnel CPF. How minimal is Walsh MC, Kliegman RM. Necrotizing enterocolitis:
“minimal” enteral feeding?. Archives of Disease in Childhood. treatment based on staging criteria. Pediatric Clinics of
Fetal and Neonatal Edition 2010;95(2):F149–50. North America 1986;33(1):179–201.

Meinzen-Derr 2009 References to other published versions of this review


Meinzen-Derr J, Poindexter B, Wrage L, Morrow AL, Stoll
B, Donovan EF. Role of human milk in extremely low birth Bombell 2009
weight infants’ risk of necrotizing enterocolitis or death. Bombell S, McGuire W. Early trophic feeding for
Journal of Perinatology 2009;29(1):57–62. very low birth weight infants. Cochrane Database of
Systematic Reviews 2009, Issue 3. [DOI: 10.1002/
Morgan 2011a 14651858.CD000504.pub3]
Morgan J, Young L, McGuire W. Delayed introduction of
Tyson 1997
progressive enteral feeds to prevent necrotising enterocolitis
Tyson JE, Kennedy KA. Minimal enteral nutrition for
in very low birth weight infants. Cochrane Database
promoting feeding tolerance and preventing morbidity
of Systematic Reviews 2011, Issue 3. [DOI: 10.1002/
in parenterally fed infants. Cochrane Database of
14651858.CD001970.pub3]
Systematic Reviews 1997, Issue 4. [DOI: 10.1002/
Morgan 2011b 14651858.CD000504]
Morgan J, Young L, McGuire W. Slow advancement of Tyson 2005
enteral feed volumes to prevent necrotising enterocolitis Tyson JE, Kennedy KA. Trophic feedings for parenterally
in very low birth weight infants. Cochrane Database fed infants. Cochrane Database of Systematic Reviews 2005,
of Systematic Reviews 2011, Issue 3. [DOI: 10.1002/ Issue 3. [DOI: 10.1002/14651858.CD000504.pub2]
14651858.CD001241.pub3] ∗
Indicates the major publication for the study

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 10
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
CHARACTERISTICS OF STUDIES

Characteristics of included studies [ordered by study ID]

Becerra 1996

Methods Randomised controlled trial

Participants VLBW infants with asphyxia, respiratory distress syndrome, suspected or documented
sepsis, hypotension, hypo- or hyperglycaemia, or anaemia or polycythaemia. The pro-
portion who received mechanical ventilation was not stated. Exclusions included immi-
nently expected death, major congenital anomalies or metabolic conditions

Interventions Minimal enteral nutrition (N = 96) vs. enteral fasting (N = 94) until 7 days after birth.
Intervention group received minimal enteral feeds of breast milk or preterm formula
milk at 25 ml/kg/day for 1 week. Control infants were not fed until 6 to 8 days after
birth

Outcomes Time to establish full enteral feeds


Incidence of necrotising enterocolitis
Time to regain birth weight

Notes Data as reported in abstract or in correspondence with the principal investigator


The method of administration of feeds was not described

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Not described - reported in abstract form only
bias)

Allocation concealment (selection bias) Unclear risk Unclear

Blinding (performance bias and detection High risk Not reported by likely that care givers and investigators
bias) were aware of allocation groups
All outcomes

Blinding of outcome assessment (detection Unclear risk Not reported


bias)
All outcomes

Incomplete outcome data (attrition bias) Low risk All data were accounted for
All outcomes

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 11
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Dunn 1988

Methods Randomised controlled trial

Participants VLBW infants with respiratory distress syndrome treated with mechanical ventilation
and with an umbilical artery catheter in situ.
Setting: Rainbow Babies and Children’s Hospital, Cleveland, USA

Interventions Minimal enteral nutrition (N = 19) vs. enteral fasting (N = 20) until 9 days after birth.
Intervention group infants received minimal enteral feeds from 48 hours at 15 to 20 ml/
kg/day of diluted preterm formula milk

Outcomes Time to establish full enteral feeds


Incidence of necrotising enterocolitis
Growth: time to regain birth weight and growth throughout study period
Duration of phototherapy
Mortality
Incidence of sepsis
Duration of hospital stay

Notes All infants received formula milk. Feeds were given by intermittent gavage nasogastric
technique
Data enabling calculation of SD relating to duration of hospital stay were not provided.
We have imputed this information from standard deviations provided by Meetze 1992,
a trial with similar sample size, as recommended by the Cochrane Handbook

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Stratified into groups according to birth weight then ran-
bias) domised using cards in paired envelopes

Allocation concealment (selection bias) High risk Unclear if envelopes were sealed - possibility that alloca-
tion groups could have been predicted

Blinding (performance bias and detection Unclear risk No blinding of care givers or investigators after allocation
bias)
All outcomes

Blinding of outcome assessment (detection Unclear risk No reference to whether interpretation of radiographs
bias) was blind
All outcomes

Incomplete outcome data (attrition bias) Unclear risk 9 infants were excluded from some of the outcome data:
All outcomes 5 deaths in the control group, 1 death in the interven-
tion group and 3 infants removed from the minimal en-
teral nutrition group due to severe unrecognised aortic
coarctation, systemic candidiasis and ileus precluding the
introduction of feeds. These infants have been included
in intention-to-treat analysis. Uncertainty exists about

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 12
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Dunn 1988 (Continued)

whether these infants went on to develop necrotising en-


terocolitis as this is not formally reported. We have as-
sumed they did not

McClure 2000

Methods Randomised controlled trial

Participants Infants weighing < 1750 grams at birth with respiratory distress syndrome who required
mechanical ventilation beyond 48 hours
Setting: Leeds General Infirmary, UK

Interventions Minimal enteral nutrition (N = 48) vs. enteral fasting (N = 52). Minimal enteral nutrition
(0.5 to 1 ml/hour of expressed maternal breast milk or preterm formula) was given from
day 3 until mechanical ventilation was discontinued. The control group received no
enteral feeding while mechanical ventilation was provided

Outcomes Feeding tolerance; days to full enteral feeding


Incidence of necrotising enterocolitis
Time to regain birth weight and growth parameters during hospital admission
Days to full oral intake, duration of parenteral nutrition
Incidence of invasive infection

Notes Both groups received parenteral nutrition. Following discontinuation of mechanical


ventilation, “nutritive” enteral feedings were initiated at 1 ml/kg/hour and increased by
1 ml/kg/hour every 8 to 12 hours as tolerated
All feeds were given by intermittent gavage

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Computer-generated sequence


bias)

Allocation concealment (selection bias) Low risk Blinding of investigators at the time of randomisation

Blinding (performance bias and detection High risk Care givers and investigators were not blinded to alloca-
bias) tion groups after randomisation had occurred
All outcomes

Blinding of outcome assessment (detection Unclear risk No description of whether radiological assessment was
bias) blind. Laboratory staff were blinded to allocation groups
All outcomes

Incomplete outcome data (attrition bias) Low risk All data were accounted for
All outcomes

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 13
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Meetze 1992

Methods Randomised controlled trial

Participants Infants of birth weight 501 to 1250 grams and gestational age at birth 25 to 32 weeks
Proportion of infants receiving mechanical ventilation not stated
Setting: neonatal unit, Gainesville, USA

Interventions Minimal enteral nutrition (N = 22) vs. enteral fasting (N = 25). The minimal enteral
nutrition group received preterm formula beginning at 2.5 ml/kg/day on day 3 advancing
to 22 ml/kg/day on day 14. During this time controls were not fed. Both groups received
progressive enteral feeds from day 15

Outcomes Incidence of necrotising enterocolitis


Growth at day 30
Mortality
Duration of phototherapy
Duration of hospital stay

Notes Infants receiving breast milk were excluded


All feeds were given by intermittent bolus orogastric administration

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Stratified randomisation based on birth weight (method
bias) of randomisation not described)

Allocation concealment (selection bias) Unclear risk Unclear

Blinding (performance bias and detection Unclear risk Care givers and investigators not blinded to intervention
bias) group
All outcomes

Blinding of outcome assessment (detection Unclear risk Not described


bias)
All outcomes

Incomplete outcome data (attrition bias) Low risk Not all data were accounted for. 7 infants were not in-
All outcomes cluded in all components of the final analyses: 1 infant
in the minimal enteral nutrition group developed necro-
tising enterocolitis on day 7 and was subsequently ex-
cluded from further analyses, 2 infants died and 4 parents
withdrew consent. This accounts for 15% of all infants
participating at time of randomisation. 6 other infants
developed necrotising enterocolitis after day 20 and were
included in all components of the analysis

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 14
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Mosqueda 2008

Methods Randomised controlled trial

Participants ELBW infants < 24 hours old


Infants with congenital anomalies, infants receiving inotrope support or exchange trans-
fusion and infants with severe acidaemia were ineligible
Setting: Neonatal Intensive Care Unit of Loyola University Medical Center, Maywood,
USA

Interventions Minimal enteral nutrition (N = 41) vs. enteral fasting (N = 43). Minimal enteral nutrition
(12 ml/kg/day) with expressed breast milk or standard formula milk was given from
day 2 until day 7. The control group received no enteral feeding. Both groups received
standard parenteral nutrition. Both groups received progressive enteral feeds (increasing
by 10 ml/kg/day) from day 8

Outcomes Feeding tolerance; days to full enteral feeding


Incidence of necrotising enterocolitis
Time to regain birth weight and growth parameters during hospital admission
Duration of hospital admission

Notes Feeds were given intermittently as boluses of nasogastric or orogastric feeds

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Not stated


bias)

Allocation concealment (selection bias) Low risk Sealed envelopes

Blinding (performance bias and detection High risk Not stated but unlikely that care givers and investigators
bias) were blinded to allocation groups
All outcomes

Blinding of outcome assessment (detection Unclear risk Unclear if interpretation of abdominal X-rays was blind
bias)
All outcomes

Incomplete outcome data (attrition bias) High risk Overall 23 out of 84 infants were not included in all
All outcomes components of the analysis due to protocol violation,
withdrawal of consent or death (8 in the minimal enteral
feeding group, 15 in the control group). This equates to
27% of the initial infants at randomisation

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 15
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Schanler 1999

Methods Randomised controlled trial

Participants Infants 26 to 30 weeks’ gestation whose birth weight was appropriate for gestational age,
who had no major congenital anomalies
Setting: Texas Children’s Hospital, Texas, USA

Interventions Minimal enteral nutrition (N = 82) vs. enteral fasting (N = 89). The minimal enteral
feeding group received 20 ml/kg/day of expressed breast milk or half-strength preterm
formula from day 4 to 14 after birth

Outcomes Feeding tolerance; days to full enteral feeding


Incidence of necrotising enterocolitis
Time to regain birth weight and growth parameters during hospital admission
Incidence of invasive infection
Mortality

Notes This study used a factorial design in which infants were randomised to 4 groups (con-
tinuous minimal enteral feeds, intermittent bolus minimal enteral feeds, enteral fast-
ing followed by continuous feeding, enteral fasting followed by bolus feeding) to allow
simultaneous assessment of the use of both minimal enteral nutrition and continuous
feedings vs. bolus. In this review, Schanler 1999 refers to outcomes reported for all infants
in trophic feedings group vs. all control infants
[February 2009: mortality data received from Dr Schanler.] [June 2012: incidence of
infection data received from Dr Schanler]

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Stratification by gestational age and type of milk followed
bias) by randomisation using sealed opaque envelopes

Allocation concealment (selection bias) Low risk Adequate given the use of sealed envelopes

Blinding (performance bias and detection Unclear risk Care givers and investigators not blinded following ran-
bias) domisation
All outcomes

Blinding of outcome assessment (detection Unclear risk Not described


bias)
All outcomes

Incomplete outcome data (attrition bias) Unclear risk Intention-to-treat analysis


All outcomes

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 16
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Sáenz de Pipaón 2003

Methods Randomised controlled trial

Participants Infants weighing < 1600 grams at birth. Exclusions included infants of diabetic mothers,
major congenital anomalies and proven sepsis
Setting: La Paz University Hospital, Madrid, Spain

Interventions Minimal enteral nutrition (N = 24) vs. enteral fasting (N = 12). On day 1, infants were
randomly allocated to either minimal enteral nutrition (10 ml/kg/day on day 1, then 20
ml/kg/day through until day 7) or enteral fasting for 7 days

Outcomes This was primarily a metabolic study examining whether enteral leucine uptake was
affected by trophic feeding
Authors also reported time to establish full feeds
Communication with authors revealed data were collected on the incidence of necrotising
enterocolitis and mortality

Notes March 2009: clarification of methods and outcome data received from Dr Saenz de
Pipaon (principal investigator):
“If the mother wished to give breast milk and the baby was allocated to the minimal
enteral nutrition group, he or she started on day one to receive breast milk. If the mother
was not able or did not wish to give breast milk the infant received formula. If the baby
was allocated to the enteral fasting group, breast milk or formula was given from day
seven.”

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Correspondence with principal investigator revealed ran-
bias) domisation involved sealed opaque envelopes with 2:1
allocation ratio

Allocation concealment (selection bias) Low risk Satisfactory

Blinding (performance bias and detection High risk No blinding of care givers or investigators
bias)
All outcomes

Blinding of outcome assessment (detection Unclear risk No statement about blinding of radiological assessment
bias)
All outcomes

Incomplete outcome data (attrition bias) Low risk All data were accounted for
All outcomes

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 17
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Troche 1995

Methods Randomised controlled trial

Participants Infants born at 25 to 30 weeks’ gestation with respiratory distress, an umbilical artery
catheter in situ, and an anticipated need for mechanical ventilation for at least 3 days.
Infants with asphyxia or respiratory failure despite ventilatory support were excluded
Setting: University of Boston, USA

Interventions Minimal enteral nutrition (N = 16) vs. enteral fasting (N = 13)


Infants in the minimal enteral nutrition group received maternal breast milk or standard
formula beginning within 24 hours after birth at a rate of 0.5 to 1.0 ml/hour until the
umbilical artery catheter was removed. Controls were fasted until the umbilical arterial
catheter was removed. Both groups received parenteral nutrition beginning on day 3

Outcomes Feeding tolerance; days to full enteral feeding


Incidence of necrotising enterocolitis
Time to regain birth weight
Mortality

Notes In infants < 800g at birth, feeds were given by continuous infusion, for those > 800 g
feeds were given as boluses

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Random numbers table


bias)

Allocation concealment (selection bias) Unclear risk Unclear

Blinding (performance bias and detection High risk Not stated but likely that care givers and investigators
bias) were aware of intervention group after allocation
All outcomes

Blinding of outcome assessment (detection Unclear risk Not stated


bias)
All outcomes

Incomplete outcome data (attrition bias) Low risk 2 infants developed necrotising enterocolitis but were
All outcomes then subsequently excluded from growth data

van Elburg 2004

Methods Randomised controlled trial

Participants Infants of birth weight < 2000 grams who were small for gestational age (< 10th percentile
for birth weight)

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 18
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
van Elburg 2004 (Continued)

Interventions Minimal enteral nutrition (N = 28) vs. enteral fasting (N = 28)


Minimal enteral nutrition (0.5 ml every 2 hours for infants < 1000 grams, 1 ml every
2 hours for infants > 1000 grams) with expressed breast milk or preterm formula milk
was given from day 2 for 5 days. The control group received no enteral feeding. Both
groups received standard parenteral nutrition. Both groups received progressive enteral
feeds (increasing by 10 ml/kg/day) from day 8

Outcomes Feeding tolerance; days to full enteral feeding


Incidence of necrotising enterocolitis
Time to regain birth weight and growth parameters during hospital admission
Duration of intensive care admission

Notes The primary aim of this study was to assess the effect of minimal enteral nutrition on
intestinal permeability in preterm infants with intra-uterine growth restriction
The method of administration of feeds was not described

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Selection of cards designating the allocation group in
bias) sealed envelopes

Allocation concealment (selection bias) Low risk Sealed opaque envelopes

Blinding (performance bias and detection Unclear risk Care givers and investigators were not blinded to alloca-
bias) tion groups
All outcomes

Blinding of outcome assessment (detection Unclear risk Unclear if interpretation of radiological images was blind
bias)
All outcomes

Incomplete outcome data (attrition bias) High risk Not all data were accounted for - 25% lost to follow-up
All outcomes due to incomplete data collection, death and one case of
congenital CMV infection

ELBW: extremely low birth weight


SD: standard deviation
VLBW: very low birth weight

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 19
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion

Berseth 1992 This trial compared 2 minimal enteral nutrition regimens. Infants were randomly assigned to receive minimal
enteral nutrition on postnatal days 3 to 5 (early feeding) or on days 10 to 14 (late feeding). The trial was excluded
because infants did not have the same feeding regimen after completion of the early trophic feeding versus enteral
fasting phase

Berseth 1993 This trial did not assess the effect of minimal enteral nutrition. Both groups were fasted enterally during the first
week after birth. In the intervention group, minimal enteral feeding was introduced 8 days after birth and controls
were given the same volume of water enterally

Berseth 2003 This randomised controlled trial compared minimal enteral nutrition with progressive enteral feed volume ad-
vancement (at daily increments of 20 ml/kg)

LaGamma 1985 Although not clearly stated in the title or abstract, this was not a randomised controlled trial

Ostertag 1986 This trial compared delayed versus early introduction of progressive enteral feeds (advanced by 10 ml/kg/day).
This trial has been included in the Cochrane review of ’Delayed enteral feeding to prevent necrotising enterocolitis
in very low birth weight infants’ (Morgan 2011a)

Said 2008 This trial compared delayed versus early introduction of enteral nutrition and may be eligible for inclusion in an
update of the Cochrane review of ’Delayed enteral feeding to prevent necrotising enterocolitis in very low birth
weight infants’ (Morgan 2011a)

Slagle 1988 This trial did not assess the effect of early minimal enteral nutrition. Both groups were fasted enterally during the
first week after birth. Minimal enteral nutrition was introduced after 8 days in the intervention group

Weiler 2006 Infants were randomly allocated to minimal enteral nutrition starting on either day 2 or day 4 after birth, that is
both groups received ’minimal enteral nutrition’

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 20
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
DATA AND ANALYSES

Comparison 1. Effects of trophic feeding versus enteral fasting

No. of No. of
Outcome or subgroup title studies participants Statistical method Effect size

1 Days to reach full enteral feeding 6 556 Mean Difference (IV, Fixed, 95% CI) -1.05 [-2.61, 0.51]
2 Incidence of necrotising 9 748 Risk Ratio (M-H, Fixed, 95% CI) 1.07 [0.67, 1.70]
enterocolitis
3 Mortality 8 558 Risk Ratio (M-H, Fixed, 95% CI) 0.66 [0.41, 1.07]
4 Days to regain birth weight 5 518 Mean Difference (IV, Fixed, 95% CI) -0.01 [-0.96, 0.95]
5 Incidence of invasive infection 3 237 Risk Ratio (M-H, Fixed, 95% CI) 1.06 [0.72, 1.56]
6 Duration of phototherapy (days) 3 170 Mean Difference (IV, Fixed, 95% CI) 0.35 [-0.29, 0.99]
7 Days of hospital stay 4 341 Mean Difference (IV, Fixed, 95% CI) -3.85 [-11.54, 3.84]

Analysis 1.1. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 1 Days to reach full
enteral feeding.

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 1 Days to reach full enteral feeding

Mean Mean
Study or subgroup Trophic feeding Enteral fasting Difference Weight Difference
N Mean(SD) N Mean(SD) IV,Fixed,95% CI IV,Fixed,95% CI

Becerra 1996 96 18.2 (10.3) 94 16.8 (7.7) 36.5 % 1.40 [ -1.18, 3.98 ]

Dunn 1988 15 31.2 (9.4) 15 47.3 (26.7) 1.2 % -16.10 [ -30.42, -1.78 ]

McClure 2000 48 24.8 (11.9) 52 36.1 (23.2) 4.8 % -11.30 [ -18.45, -4.15 ]

Schanler 1999 82 35 (32) 89 32 (20) 3.7 % 3.00 [ -5.08, 11.08 ]

S enz de Pipa n 2003 24 17 (5) 14 17 (5) 22.4 % 0.0 [ -3.30, 3.30 ]

Troche 1995 16 10 (3) 11 13 (4) 31.4 % -3.00 [ -5.78, -0.22 ]

Total (95% CI) 281 275 100.0 % -1.05 [ -2.61, 0.51 ]


Heterogeneity: Chi2 = 18.84, df = 5 (P = 0.002); I2 =73%
Test for overall effect: Z = 1.32 (P = 0.19)
Test for subgroup differences: Not applicable

-20 -10 0 10 20
Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 21
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.2. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 2 Incidence of
necrotising enterocolitis.

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 2 Incidence of necrotising enterocolitis

Study or subgroup Trophic feeding Enteral fasting Risk Ratio Risk Ratio
n/N n/N M-H,Fixed,95% CI M-H,Fixed,95% CI
Becerra 1996 8/96 6/94 1.31 [ 0.47, 3.62 ]

Dunn 1988 3/19 1/20 3.16 [ 0.36, 27.78 ]

McClure 2000 1/48 2/52 0.54 [ 0.05, 5.78 ]

Meetze 1992 3/20 4/21 0.79 [ 0.20, 3.09 ]

Mosqueda 2008 3/41 4/43 0.79 [ 0.19, 3.30 ]

Schanler 1999 13/82 10/89 1.41 [ 0.65, 3.04 ]

S enz de Pipa n 2003 0/24 0/14 0.0 [ 0.0, 0.0 ]

Troche 1995 0/16 2/13 0.16 [ 0.01, 3.16 ]

van Elburg 2004 0/28 1/28 0.33 [ 0.01, 7.85 ]

Total (95% CI) 374 374 1.07 [ 0.67, 1.70 ]


Total events: 31 (Trophic feeding), 30 (Enteral fasting)
Heterogeneity: Chi2 = 4.35, df = 7 (P = 0.74); I2 =0.0%
Test for overall effect: Z = 0.27 (P = 0.78)
Test for subgroup differences: Not applicable

0.01 0.1 1 10 100


Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 22
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.3. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 3 Mortality.

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 3 Mortality

Study or subgroup Trophic feeding Enteral fasting Risk Ratio Risk Ratio
n/N n/N M-H,Fixed,95% CI M-H,Fixed,95% CI
Dunn 1988 1/19 5/20 0.21 [ 0.03, 1.64 ]

McClure 2000 6/48 11/52 0.59 [ 0.24, 1.47 ]

Meetze 1992 0/20 2/23 0.23 [ 0.01, 4.50 ]

Mosqueda 2008 7/41 11/43 0.67 [ 0.29, 1.55 ]

Schanler 1999 6/82 6/89 1.09 [ 0.36, 3.23 ]

S enz de Pipa n 2003 0/24 0/12 0.0 [ 0.0, 0.0 ]

Troche 1995 1/16 1/13 0.81 [ 0.06, 11.77 ]

van Elburg 2004 2/28 1/28 2.00 [ 0.19, 20.82 ]

Total (95% CI) 278 280 0.66 [ 0.41, 1.07 ]


Total events: 23 (Trophic feeding), 37 (Enteral fasting)
Heterogeneity: Chi2 = 3.41, df = 6 (P = 0.76); I2 =0.0%
Test for overall effect: Z = 1.67 (P = 0.094)
Test for subgroup differences: Not applicable

0.01 0.1 1 10 100


Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 23
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.4. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 4 Days to regain birth
weight.

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 4 Days to regain birth weight

Mean Mean
Study or subgroup Trophic feeding Enteral fasting Difference Weight Difference
N Mean(SD) N Mean(SD) IV,Fixed,95% CI IV,Fixed,95% CI

Becerra 1996 96 14.3 (5.5) 94 13.5 (5.2) 39.4 % 0.80 [ -0.72, 2.32 ]

Dunn 1988 15 19.9 (6.2) 15 24.4 (8.5) 3.2 % -4.50 [ -9.82, 0.82 ]

McClure 2000 48 16.4 (6) 52 18.2 (9.2) 10.0 % -1.80 [ -4.82, 1.22 ]

Schanler 1999 82 12.5 (5) 89 12.5 (6) 33.5 % 0.0 [ -1.65, 1.65 ]

Troche 1995 16 19 (2) 11 19 (4) 13.9 % 0.0 [ -2.56, 2.56 ]

Total (95% CI) 257 261 100.0 % -0.01 [ -0.96, 0.95 ]


Heterogeneity: Chi2 = 5.17, df = 4 (P = 0.27); I2 =23%
Test for overall effect: Z = 0.02 (P = 0.98)
Test for subgroup differences: Not applicable

-10 -5 0 5 10
Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 24
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.5. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 5 Incidence of
invasive infection.
Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 5 Incidence of invasive infection

Study or subgroup Trophic feeding Enteral fasting Risk Ratio Weight Risk Ratio
n/N n/N M-H,Fixed,95% CI M-H,Fixed,95% CI
Dunn 1988 6/19 4/20 11.2 % 1.58 [ 0.53, 4.74 ]

Mosqueda 2008 13/41 9/43 25.2 % 1.51 [ 0.73, 3.16 ]

Schanler 1999 17/55 23/59 63.6 % 0.79 [ 0.48, 1.32 ]

Total (95% CI) 115 122 100.0 % 1.06 [ 0.72, 1.56 ]


Total events: 36 (Trophic feeding), 36 (Enteral fasting)
Heterogeneity: Chi2 = 2.67, df = 2 (P = 0.26); I2 =25%
Test for overall effect: Z = 0.31 (P = 0.76)
Test for subgroup differences: Not applicable

0.5 0.7 1 1.5 2


Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 25
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.6. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 6 Duration of
phototherapy (days).

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 6 Duration of phototherapy (days)

Mean Mean
Study or subgroup Trophic feeding Enteral fasting Difference Weight Difference
N Mean(SD) N Mean(SD) IV,Fixed,95% CI IV,Fixed,95% CI

Dunn 1988 15 6.8 (2.4) 15 9.5 (4) 7.4 % -2.70 [ -5.06, -0.34 ]

McClure 2000 48 2.3 (1.7) 52 1.8 (1.8) 87.1 % 0.50 [ -0.19, 1.19 ]

Meetze 1992 19 6.3 (5.2) 21 4.3 (3.2) 5.6 % 2.00 [ -0.71, 4.71 ]

Total (95% CI) 82 88 100.0 % 0.35 [ -0.29, 0.99 ]


Heterogeneity: Chi2 = 8.02, df = 2 (P = 0.02); I2 =75%
Test for overall effect: Z = 1.07 (P = 0.29)
Test for subgroup differences: Not applicable

-4 -2 0 2 4
Favours trophic Favours fasting

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 26
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Analysis 1.7. Comparison 1 Effects of trophic feeding versus enteral fasting, Outcome 7 Days of hospital stay.

Review: Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants

Comparison: 1 Effects of trophic feeding versus enteral fasting

Outcome: 7 Days of hospital stay

Mean Mean
Study or subgroup Trophic feeding Enteral fasting Difference Weight Difference
N Mean(SD) N Mean(SD) IV,Fixed,95% CI IV,Fixed,95% CI

Dunn 1988 15 98 (20.9) 15 102 (33) 15.1 % -4.00 [ -23.77, 15.77 ]

McClure 2000 48 70.3 (27.2) 52 92.4 (58.3) 19.1 % -22.10 [ -39.72, -4.48 ]

Meetze 1992 19 73 (20.9) 21 76 (33) 20.6 % -3.00 [ -19.96, 13.96 ]

Schanler 1999 82 84 (43) 89 80.5 (32) 45.2 % 3.50 [ -7.94, 14.94 ]

Total (95% CI) 164 177 100.0 % -3.85 [ -11.54, 3.84 ]


Heterogeneity: Chi2 = 5.72, df = 3 (P = 0.13); I2 =48%
Test for overall effect: Z = 0.98 (P = 0.33)
Test for subgroup differences: Not applicable

-20 -10 0 10 20
Favours trophic Favours fasting

WHAT’S NEW
Last assessed as up-to-date: 27 December 2012.

Date Event Description

6 February 2013 New citation required and conclusions have changed The title has been amended to ’Early trophic feeding
versus enteral fasting for very preterm or very low birth
weight infants’ to emphasise the comparison with fast-
ing rather than progressive feeding
The search strategy was updated in December 2012.
One new study was assessed for eligibility but was ex-
cluded based on the definition of the interventions
Further (unpublished) data were obtained from cur-
rent included trials and added to the meta-analyses

27 December 2012 New search has been performed This updates the review ’Early trophic feeding for very
low birth weight infants’ (Bombell 2009).

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 27
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
HISTORY
Protocol first published: Issue 4, 1997
Review first published: Issue 4, 1997

Date Event Description

7 March 2009 New citation required and conclusions have changed New authorship: Sarah Bombell, William McGuire.

7 March 2009 New search has been performed This updates the review ’Trophic feedings for parenter-
ally fed infants’ by Tyson JE, Kennedy KA, Cochrane
Database of Systematic Reviews 2005, Issue 3 (Tyson
2005).
The title has been modified to ’Early trophic feeding for
very low birth weight infants’ and has a new authorship
of Sarah Bombell and William McGuire. Changes made
to the original protocol are outlined below:
1. The population has been restricted to very low birth
weight and very preterm infants
2. Early trophic feeding is defined as enteral feeding up
to 24 ml/kg/day (1 ml/kg/hour) beginning within four
days after birth and continued until at least one week
after birth versus enteral fasting for at least one week af-
ter birth. On the subsequent introduction of progressive
enteral feeding, infants should have received the same
type of milk (breast milk or formula), the same route
and mode of feeding (intragastric or transpyloric, bolus
gavage or continuous), and the same rate of feed volume
advancement in both groups
3. Subgroup analyses of extremely low birth weight and
extremely preterm infants and infants with evidence of
intrauterine growth restriction or absent or reversed end-
diastolic flow velocities in Doppler studies of the fetal
aorta or umbilical artery were prespecified.

Search updated February 2009. Three new trials were


included (Sáenz de Pipaón 2003; van Elburg 2004;
Mosqueda 2008).
Five trials included in the previous version of this review
have been excluded because they did not fulfil the stricter
definition of the intervention and comparison (Ostertag
1986; Slagle 1988; Berseth 1992; Berseth 1993; Berseth
2003).
The main change to the findings and implications for
practice is that the typical estimate for feed tolerance
(time to full enteral feeding) is no longer statistically
significant

28 October 2008 Amended Converted to new review format.

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 28
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
(Continued)

31 March 2005 New search has been performed This review updates the existing review of ’Minimal
enteral nutrition in parenterally fed neonates’ that was
published in The Cochrane Library, Disk Issue 4, 1997.
Three new eligible trials (Berseth 2003; McClure 2000;
Schanler 1999) have been found.

31 March 2005 New citation required and conclusions have changed Substantive amendment.

CONTRIBUTIONS OF AUTHORS
The review authors developed the protocol, undertook the literature search, appraised the articles, extracted and entered the data, and
completed the review jointly.

DECLARATIONS OF INTEREST
None.

SOURCES OF SUPPORT
Internal sources
• Centre for Reviews and Dissemination, University of York, UK.

External sources
• NIHR, UK.
• Eunice Kennedy Shriver National Institute of Child Health and Human Development National Institutes of Health,
Department of Health and Human Services, USA.
The Cochrane Neonatal Review Group has been funded in part with Federal funds from the Eunice Kennedy Shriver National
Institute of Child Health and Human Development National Institutes of Health, Department of Health and Human Services, USA,
under Contract No. HHSN267200603418C

INDEX TERMS

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 29
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Medical Subject Headings (MeSH)
∗ Milk;Adaptation, Physiological; Child Development [∗ physiology]; Enteral Nutrition [adverse effects; ∗ methods]; Enterocolitis,
Necrotizing [prevention & control]; Infant Formula; Infant, Newborn; Infant, Premature; Infant, Very Low Birth Weight [growth &
development; ∗ physiology]; Milk, Human; Parenteral Nutrition [adverse effects; methods]; Randomized Controlled Trials as Topic

MeSH check words


Animals; Humans

Early trophic feeding versus enteral fasting for very preterm or very low birth weight infants (Review) 30
Copyright © 2013 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Slow advancement of enteral feed volumes to prevent
necrotising enterocolitis in very low birth weight infants
(Review)

Morgan J, Young L, McGuire W

This is a reprint of a Cochrane review, prepared and maintained by The Cochrane Collaboration and published in The Cochrane Library
2014, Issue 12
http://www.thecochranelibrary.com

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review)
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
TABLE OF CONTENTS
HEADER . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
ABSTRACT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
PLAIN LANGUAGE SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
BACKGROUND . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
OBJECTIVES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
METHODS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
RESULTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Figure 1. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Figure 2. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Figure 3. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Figure 4. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
DISCUSSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Figure 5. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
AUTHORS’ CONCLUSIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
ACKNOWLEDGEMENTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
CHARACTERISTICS OF STUDIES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
DATA AND ANALYSES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
WHAT’S NEW . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
HISTORY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
CONTRIBUTIONS OF AUTHORS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
DECLARATIONS OF INTEREST . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
SOURCES OF SUPPORT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
INDEX TERMS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) i
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
[Intervention Review]

Slow advancement of enteral feed volumes to prevent


necrotising enterocolitis in very low birth weight infants

Jessie Morgan1 , Lauren Young2 , William McGuire1

1 Hull
York Medical School & Centre for Reviews and Dissemination, University of York, York, UK. 2 Neonatal Unit, Mercy Hospital
for Women, Heidelberg, Australia

Contact address: William McGuire, Hull York Medical School & Centre for Reviews and Dissemination, University of York, York,
Y010 5DD, UK. William.McGuire@hyms.ac.uk.

Editorial group: Cochrane Neonatal Group.


Publication status and date: New search for studies and content updated (no change to conclusions), published in Issue 12, 2014.
Review content assessed as up-to-date: 3 October 2014.

Citation: Morgan J, Young L, McGuire W. Slow advancement of enteral feed volumes to prevent necrotising enterocoli-
tis in very low birth weight infants. Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No.: CD001241. DOI:
10.1002/14651858.CD001241.pub5.

Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

ABSTRACT

Background

Early enteral feeding practices are potentially modifiable risk factors for necrotising enterocolitis in very preterm or very low birth weight
(VLBW) infants. Observational studies suggest that conservative feeding regimens, including slowly advancing enteral feed volumes,
reduce the risk of necrotising enterocolitis. However, slow feed advancement may delay establishment of full enteral feeding and be
associated with metabolic and infectious morbidities secondary to prolonged exposure to parenteral nutrition.

Objectives

To determine the effect of slow rates of enteral feed advancement on the incidence of necrotising enterocolitis, mortality, and other
morbidities in very preterm or VLBW infants.

Search methods

We used the standard search strategy of the Cochrane Neonatal Review Group Specialised Register. We searched the Cochrane Central
Register of Controlled Trials (CENTRAL 2014, Issue 8), MEDLINE, EMBASE, and CINAHL (to September 2014), conference
proceedings, and previous reviews.

Selection criteria

Randomised or quasi-randomised controlled trials that assessed the effect of slow (up to 24 ml/kg per day) versus faster rates of
advancement of enteral feed volumes upon the incidence of necrotising enterocolitis in very preterm or VLBW infants.

Data collection and analysis

Two review authors independently assessed trial eligibility and risk of bias and undertook data extraction. We analysed the treatment
effects in the individual trials and reported the risk ratio and risk difference for dichotomous data and mean difference for continuous
data, with respective 95% confidence intervals. We used a fixed-effect model in meta-analyses and explored the potential causes of
heterogeneity in sensitivity analyses.
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 1
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Main results

We identified six randomised controlled trials in which a total of 618 infants participated. Most participants were stable preterm infants
of birth weight between 1000 g and 1500 g. Few participants were extremely preterm, extremely low birth weight, or growth-restricted.
The trials typically defined slow advancement as daily increments of 15 ml/kg to 20 ml/kg and faster advancement as 30 ml/kg to
35 ml/kg. Meta-analyses did not detect statistically significant effects on the risk of necrotising enterocolitis (typical risk ratio (RR)
0.96, 95% confidence interval (CI) 0.55 to 1.70) or all-cause mortality (typical RR 1.57, 95% CI 0.92 to 2.70). Infants who had slow
advancement took significantly longer to regain birth weight (reported median differences 2 to 6 days) and to establish full enteral
feeding (1 to 5 days).

Authors’ conclusions

The available trial data suggest that advancing enteral feed volumes at daily increments of 30 ml/kg to 35 ml/kg does not increase
the risk of necrotising enterocolitis in very preterm or VLBW infants. Advancing the volume of enteral feeds at slow rates resulted in
several days delay in regaining birth weight and establishing full enteral feeds. The applicability of these findings to extremely preterm,
extremely low birth weight, or growth-restricted infants is limited. Further randomised controlled trials in these populations may be
warranted to resolve this uncertainty.

PLAIN LANGUAGE SUMMARY

Slowly advancing milk feeds does not reduce the risk of necrotising enterocolitis in very low birth weight infants

Very preterm (born before 32 weeks’ gestation) or very low birth weight (less than 1500 g) infants are at risk of developing a severe
bowel disorder known as necrotising enterocolitis. It is thought that one possible way to prevent this condition is to limit the amount
of milk feeds that infants receive each day for the first few weeks after birth. Six randomised controlled trials have assessed the effect of
slowly (rather than more quickly) increasing the volume of milk feeds given to very preterm or very low birth weight infants. Combined
analysis of these trials did not reveal any effect of the intervention on the risk of necrotising enterocolitis. Infants fed more slowly
regained birth weight and attained full enteral feeding several days later than infants fed more quickly.

BACKGROUND Description of the intervention


Short gestational age at birth is the major clinical risk factor for
developing NEC (Beeby 1992; Luig 2005). The other putative
major risk factor is intrauterine growth restriction, especially if
Description of the condition
associated with absent or reversed end-diastolic flow velocities in
Necrotising enterocolitis (NEC), a syndrome of acute intestinal Doppler studies of the fetal aorta or umbilical artery (Bernstein
necrosis of unknown aetiology, affects about 5% of very preterm 2000; Garite 2004; Dorling 2005). Most very preterm or VLBW
(less than 32 weeks) or very low birth weight (VLBW) (less than infants who develop NEC have received enteral milk feeds. Evi-
1500 g) infants (Holman 1997). Infants who develop NEC expe- dence exists that feeding with artificial formula rather than human
rience more nosocomial infections, have lower levels of nutrient milk increases the risk of developing NEC (Quigley 2014). Other
intake, grow more slowly, and have longer durations of intensive differences in enteral feeding regimens, such as the timing of intro-
care and hospital stay than gestation-comparable infants who do duction of feeds and the size of the daily volume increments, may
not develop NEC (Bisquera 2002; Guthrie 2003). The associ- also contribute to inter-unit variation in the incidence of NEC.
ated mortality rate is more than 20%. Compared with their peers, Multicentre benchmarking studies have found that those neona-
infants who develop NEC have a higher incidence of long-term tal centres where enteral feeding is introduced earlier and feeding
neurological disability, which may be a consequence of infection volumes advanced more quickly tend to have higher incidences
and under-nutrition during a critical period of brain development of NEC (Uauy 1991). Observational studies have suggested that
(Stoll 2004; Soraisham 2006; Rees 2007; Pike 2012).
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 2
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
delaying the introduction of enteral feeds beyond the first few days Types of interventions
after birth, or increasing the volume of feeds by less than about Advancement of enteral feeds at no more than 24 ml/kg (birth
20 ml/kg to 24 ml/kg body weight each day, is associated with a weight or current body weight) per day versus faster rates of feeds
lower risk of developing NEC in very preterm or VLBW infants advancement. Infants should have received the same type of milk
(Brown 1978; McKeown 1992; Patole 2005; Henderson 2009). and in both groups the advancement of feed volume should have
commenced within five days of introduction of enteral feeds.

Why it is important to do this review


Types of outcome measures
Potential disadvantages associated with slowing the advancement
of enteral feed volumes include delaying the establishment of full
enteral nutrition and extending the duration of receipt of par- Primary outcomes
enteral nutrition (Flidel-Rimon 2004). Prolonged use of parenteral 1. NEC confirmed by at least two of the following features:
nutrition is associated with infectious and metabolic risks that may • abdominal radiograph showing pneumatosis intestinalis or
have adverse consequences for survival, growth, and development gas in the portal venous system or free air in the abdomen;
(Stoll 2004). It has been argued that the risk of NEC should not be • abdominal distension with abdominal radiograph with
considered in isolation of these other potential clinical outcomes gaseous distension or frothy appearance of bowel lumen (or
when determining feeding policies and practice for very preterm both);
or VLBW infants (Flidel-Rimon 2006; Chauhan 2008; Härtel • blood in stool;
2009). • lethargy, hypotonia, or apnoea (or a combination of these);
Other Cochrane reviews have addressed the questions of whether
delaying the introduction of any enteral milk feeding or restricting or a diagnosis confirmed at surgery or autopsy (Walsh 1986).
feed volumes to trophic levels (minimal enteral nutrition) affect 2. All-cause mortality during the neonatal period and prior to
the risk of NEC in very preterm or VLBW infants (Bombell hospital discharge.
2009; Morgan 2011). This review focussed on the question of
whether advancing feed volumes at slow rates compared to faster
Secondary outcomes
rates affects the risk of NEC, mortality, and other morbidities.
1. Growth
i) Time to regain birth weight and subsequent rates of
weight gain, linear growth, head growth, or skinfold thickness
growth up to six months (corrected for preterm birth).
OBJECTIVES ii) Long-term growth: weight, height, or head
To determine the effect of slow rates of enteral feed advancement circumference (or proportion of infants who remained below the
10th percentile for the index population’s distribution) assessed
on the incidence of NEC, mortality, and other morbidities in very
at intervals from six months of age.
preterm or VLBW infants.
2. Neurodevelopment
i) Death or severe neurodevelopmental disability defined
as any one or a combination of the following: nonambulant
METHODS cerebral palsy, developmental delay (developmental quotient less
than 70), auditory and visual impairment. Each component was
to be analysed individually as well as part of the composite
Criteria for considering studies for this review outcome.
ii) Neurodevelopmental scores in survivors aged greater
than or equal to 12 months measured using validated assessment
tools.
Types of studies
iii) Cognitive and educational outcomes in survivors aged
Controlled trials utilising either random or quasi-random partic- more than five years.
ipant allocation. 3. Time to establish full enteral feeding (independently of
parenteral nutrition).
4. Time to establish oral feeding (independently of parenteral
Types of participants nutrition or enteral tube feeding, or both).
Enterally fed very preterm (less than 32 weeks) or VLBW (less 5. Feed intolerance (defined as a requirement to cease enteral
than 1500 g) newborn infants. feeds).

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 3
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
6. Incidence of invasive infection as determined by culture of Data extraction and management
bacteria or fungus from blood, cerebrospinal fluid, urine, or from We used a data collection form to aid extraction of relevant infor-
a normally sterile body space. mation from each included study. Two review authors extracted
7. Duration of hospital stay (days). the data separately. Any disagreements were discussed until con-
sensus was achieved. We contacted the investigators for further
information if data from the trial reports were insufficient.
Search methods for identification of studies
We used the standard search strategy of the Cochrane Neonatal Assessment of risk of bias in included studies
Review Group (http://neonatal.cochrane.org/). We used the criteria and standard methods of The Cochrane
Collaboration and the Cochrane Neonatal Group to assess the
methodological quality of any included trials (Higgins 2011). We
Electronic searches requested additional information from the trial authors to clarify
We searched the Cochrane Central Register of Controlled Tri- methodology and results as necessary. We evaluated and reported
als (CENTRAL 2014, Issue 8), MEDLINE (1966 to September the following issues in the ’Risk of bias’ tables:
2014), EMBASE (1980 to September 2014), and CINAHL (1982 Sequence generation (the method used to generate the allocation
to September 2014) using a combination of the following text sequence):
words and MeSH terms: [Infant, Newborn OR Infant, Premature • low risk: any truly random process, e.g. random number
OR Infant, Low Birth Weight OR Infant, Very Low Birth Weight/ table, computer random number generator;
OR infan* OR neonat* OR preterm OR prem*] AND “Infant- • high risk: any nonrandom process, e.g. odd or even date of
Nutrition”/all subheadings OR Infant Formula OR milk OR for- birth, hospital or clinic record number;
mula OR trophic feeding OR minimal enteral nutrition OR gut • unclear risk: no or unclear information provided.
priming]. We limited the search outputs with the relevant search
Allocation concealment (the method used to conceal the allocation
filters for clinical trials. We did not apply any language restrictions.
sequence):
We searched ClinicalTrials.gov for completed or ongoing trials.
• low risk: e.g. telephone or central randomisation;
consecutively numbered, sealed, opaque envelopes;
• high risk: open random allocation, e.g. unsealed or
Searching other resources
nonopaque envelopes, alternation, date of birth;
We examined the reference lists of all studies identified as poten- • unclear: no or unclear information provided.
tially relevant.
We searched the abstracts from the annual meetings of the Pedi- Blinding (the methods used to ensure blinding of participants,
atric Academic Societies (1993 to 2014), the European Society clinicians and caregivers, and outcome assessors):
for Pediatric Research (1995 to 2013), the UK Royal College of • low risk;
Paediatrics and Child Health (2000 to 2014), and the Perinatal • high risk;
Society of Australia and New Zealand (2000 to 2014). Trials re- • unclear.
ported only as abstracts were eligible if sufficient information was Incomplete outcome data (completeness of data including attri-
available from the report or from contact with the authors to fulfill tion and exclusions from the analysis for each outcome and any
the inclusion criteria. reasons for attrition or exclusion where reported): We will assess
whether missing data are balanced across groups or are related to
outcomes. Where sufficient information is reported or supplied
Data collection and analysis by the trial authors, we will reinstate missing data in the analyses.
We will categorise completeness as:
We used the standard methods of the Cochrane Neonatal Review • low risk: adequate (< 10% missing data);
Group (http://neonatal.cochrane.org/). • high risk: inadequate (> 10% missing data);
• unclear risk: no or unclear information provided.

Selection of studies
Two review authors screened the titles and abstracts of all studies Measures of treatment effect
identified by the above search strategy. We assessed the full texts We calculated risk ratio (RR) and risk difference (RD) for dichoto-
of any potentially eligible reports, and those studies that did not mous data and mean difference (MD) for continuous data, with
meet all of the inclusion criteria were excluded. We discussed any respective 95% confidence intervals (CI). When it was deemed
disagreements until consensus was achieved. appropriate to combine two or more study arms, we obtained the

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 4
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
treatment effects from the combined data using the methods de- 2010; Karagol 2012; Mukhopadhyay 2014 (see Characteristics of
scribed in the Cochrane Handbook for Systematic Reviews of Inter- included studies).
ventions (Higgins 2011). We determined the number needed to
treat to benefit (NNTB) or harm (NNTH) for a statistically sig-
nificant difference in the RD. Included studies

Unit of analysis issues Population


The unit of analysis was the participating infant in individually A total of 618 infants participated in the six included trials. The
randomised trials and the neonatal unit (or subunit) for cluster trials were undertaken in neonatal care centres in North America
randomised trials. (Rayyis 1999; Caple 2004), India (Salhotra 2004; Krishnamurthy
2010; Mukhopadhyay 2014), and Turkey (Karagol 2012) within
the past 10 to 15 years.
Assessment of heterogeneity All of the trials specified participant birth weight eligibility criteria:
If more than one trial was included in a meta-analysis, we exam- • Rayyis 1999 < 1500 g
ined the treatment effects of individual trials and the heterogeneity • Caple 2004 1000 g to 2000 g
between trial results by inspecting the forest plots. We calculated • Salhotra 2004 < 1250 g
the I² statistic for each analysis to quantify inconsistency across • Krishnamurthy 2010 1000 g to 1500 g
studies and to describe the percentage of variability in effect esti- • Karagol 2012 750 g to 1250 g
mates that may be due to heterogeneity rather than sampling er- • Mukhopadhyay 2014 1000 g to 1250 g
ror. If we detected substantial (I² greater than 50%) heterogeneity, Since most participants in Caple 2004 were of birth weight less
we explored the possible causes (for example, differences in study than 1500 g or gestational age less than 32 weeks, we made a
design, participants, interventions, or completeness of outcome consensus decision to include the trial. Infants born ’small for
assessments) in sensitivity analyses. gestational age’ (birth weight less than 10th percentile of the index
population’s distribution) were not eligible to participate in Caple
Data synthesis 2004 but were included in the other trials. More than 95% of
the participants in Salhotra 2004 were ’small for gestational age’.
We used the fixed-effect model in RevMan 5 (RevMan 2011) for
One-third of participants in Karagol 2012 were ELBW infants.
meta-analysis.
All participants in Mukhopadhyay 2014 had antenatal evidence
of absent or reversed end-diastolic flow.
Subgroup analysis and investigation of heterogeneity
We planned the following subgroup analyses: Interventions and comparisons
1. trials in which most infants were exclusively formula fed; All trials commenced interval bolus intragastric feeding within one
2. trials in which most infants were at least partially fed with to five days after birth. Infants were randomly allocated to one of
human milk (maternal or donor); two rates of daily increments in enteral feed volume:
3. trials in which most participants were of extremely low • Rayyis 1999 15 versus 35 ml/kg/day
birth weight (ELBW) (< 1000 g) or extremely preterm • Caple 2004 20 versus 35 ml/kg/day
gestational age (< 28 weeks); • Salhotra 2004 15 versus 30 ml/kg/day
4. trials in which participants were infants with intrauterine • Krishnamurthy 2010 20 versus 30 ml/kg/day
growth restriction or infants with absent or reversed end-diastolic • Karagol 2012 20 versus 30 ml/kg/day
flow velocities detected on antenatal Doppler studies of the fetal • Mukhopadhyay 2014 20 versus 30 ml/kg/day
aorta or umbilical artery.
In one trial, only formula-fed infants were eligible to participate
(Rayyis 1999). In Caple 2004, Krishnamurthy 2010, Karagol
2012, and Mukhopadhyay 2014, infants received either expressed
breast milk or formula, or a combination of the two. In Salhotra
RESULTS
2004, all participating infants were fed exclusively expressed breast
milk. All of the trial protocols specified indications for interrupt-
ing or ceasing enteral feeding, such as residual gastric contents of
Description of studies more than about one-third of the previous feed volume, frequent
Six randomised controlled trials fulfilled the review eligibility cri- vomiting, abdominal distention, or detection of blood in the stools
teria: Rayyis 1999; Caple 2004; Salhotra 2004; Krishnamurthy (including occult blood).

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 5
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Outcomes tion concealment and reported complete or near-complete assess-
All of the trials reported the incidence of NEC confirmed radi- ments of the primary outcomes. None of the trials were able to
ologically or at surgery or autopsy. The other reported outcomes conceal the feeding strategies from parents, caregivers, or clinical
included time to regain birth weight, time to establish full enteral investigators. Three studies clearly masked the assessment of ab-
feeding, duration of hospital stay, and rates of invasive infection. dominal radiographs (for diagnosis of NEC). In Salhotra 2004 and
Karagol 2012, it was unclear whether precautions had been taken
to ensure that radiological assessors were blinded to the allocation
Excluded studies group.

Two trials were excluded (Book 1976; Berseth 2003) (see


Characteristics of excluded studies). In Book 1976, enteral feeding
volumes were advanced at 10 ml/kg/day versus 20 ml/kg/day, that Effects of interventions
is both groups received ’slow’ advancement of feed volumes. In
Berseth 2003, infants were randomly allocated to either a stable
(not progressively increased) trophic feeding volume or to feed Primary outcomes
volume advancement at 20 ml/kg/day.

Incidence of necrotising enterocolitis (Outcome 1.1)


Meta-analysis did not detect a statistically significant effect: typical
Risk of bias in included studies RR 0.96 (95% CI 0.55 to 1.63); typical RD -0.00 (95% CI -
The methodological quality of the included trials was generally 0.04 to 0.04) (6 trials, 618 infants) (Figure 1). There was not any
good. All six trials employed methods to ensure adequate alloca- statistical evidence of heterogeneity (I² = 0%).

Figure 1. Forest plot of comparison: 1 Slow versus faster rates of feed advancement, outcome: 1.1
Incidence of necrotising enterocolitis.

ical RR 1.57 (95% CI 0.92 to 2.70); typical RD 0.04 (95% CI -


Mortality (Outcome 1.2)
0.01 to 0.09) (5 trials, 460 infants) (Figure 2). There was not any
Meta-analysis did not find a statistically significant difference: typ-
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 6
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
statistical evidence of heterogeneity (I² = 0%).

Figure 2. Forest plot of comparison: 1 Slow versus faster rates of feed advancement, outcome: 1.2 Mortality.

Time to establish full enteral feeding

Secondary outcomes All of the trials reported that it took statistically significantly longer
to establish full enteral feeds in infants in the slow-rate-of-advance-
ment group:
• Rayyis 1999 median difference 4 days
Growth • Caple 2004 mean difference 3 (95% CI 2 to 3) days
Five trials reported that infants in the slow-rate-of-advancement • Salhotra 2004 mean difference 4.8 (95% CI not given) days
group took a statistically significantly longer time to regain birth • Krishnamurthy 2010 median difference 2 days
weight: • Karagol 2012 mean difference 3.2 (95% CI not given) days
• Rayyis 1999 median difference 3 days • Mukhopadhyay 2014 mean difference 0.6 (95% CI not
• Caple 2004 mean difference 2 (95% CI 1 to 3) days given) days
• Salhotra 2004 median difference 5 days
• Krishnamurthy 2010 median difference 6 days Time to establish full oral feeding
• Karagol 2012 mean difference 3.8 (95% CI not given) days
Not reported by any of the included trials.
Mukhopadhyay 2014 did not report this outcome.
None of the trials reported longer-term growth parameters.
Feeds intolerance (causing interruption of enteral feeding)
(Outcome 1.3)
Meta-analysis of data from four trials (275 infants) did not find a
Neurodevelopment statistically significant difference: typical RR 1.23 (95% CI 0.87
None of the trials assessed neurodevelopmental outcomes. to 1.73); typical RD 0.06 (95% CI -0.04 to 0.17) (Figure 3).

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 7
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Figure 3. Forest plot of comparison: 1 Slow versus faster rates of feed advancement, outcome: 1.3 Feeds
intolerance (causing interruption of enteral feeding).

Incidence of invasive infection (Outcome 1.4)


Meta-analysis of data from three trials (222 infants) did not detect
a statistically significant effect: typical RR 1.58 (95% CI 0.81 to
3.09); typical RD 0.06 (95% CI -0.03 to 0.15) (Figure 4).

Figure 4. Forest plot of comparison: 1 Slow versus faster rates of feed advancement, outcome: 1.4
Incidence of invasive infection.

1. Exclusively formula-fed infants (Rayyis 1999). No statistically


Duration of hospital stay
significant differences:
Two trials did not detect a statistically significant difference: • NEC: RR 1.44 (95% CI 0.63 to 3.32); RD 0.04 (95% CI -
• Rayyis 1999 median difference 4 days 0.05 to 0.13)
• Caple 2004 mean difference 5 (95% CI -1 to 8) days • Mortality: RR 0.59 (95% CI 0.10 to 3.46); RD -0.01 (95%
CI -0.06 to 0.03)
Two trials reported that the duration of hospital stay was statis-
tically significantly longer in infants in the slow-rate-of-advance- 2. Infants at least partially fed with human milk. Subgroup data
ment group: not available.
• Krishnamurthy 2010 median difference 1.5 days 3. ELBW or extremely preterm infants. None of the trials recruited
• Karagol 2012 mean difference 6 (95% CI not given) days predominantly ELBW or extremely preterm infants.
4. Infants with intrauterine growth restriction or absent or reversed
end-diastolic flow velocities (Salhotra 2004; Mukhopadhyay
2014). No statistically significant difference in NEC, but border-
Subgroup analyses
line significant increase in mortality in slow group:
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 8
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
• NEC: typical RR 0.34 (95% CI 0.06 to 2.04); typical RD - 1999; Caple 2004). Infants who had severe respiratory distress
0.07 (95% CI -0.18 to 0.04) (Figure 1) requiring oxygen supplementation or ventilatory support were
• Mortality: typical RR 2.13 (95% CI 1.02 to 4.47); typical not eligible to participate in three of the trials (Salhotra 2004;
RD 0.02 (95% CI 0.02 to 0.38); number needed to treat to Krishnamurthy 2010; Karagol 2012). The findings may not be
harm 5 (95% CI 3 to 50), 2 trials, 83 infants (Figure 2) applicable to these populations, which are at highest risk of devel-
oping feed intolerance or NEC (Luig 2005).
Fewer than half of all of the participating infants were fed with
breast milk. Evidence exists that artificial-formula feeding increases
DISCUSSION the risk of feed intolerance and NEC (Quigley 2014). The risk-
benefit balance of enteral feeding strategies may differ between
human milk-fed and formula-fed very preterm or VLBW infants.
Summary of main results
It is also unclear whether the findings can be applied to infants
The currently available trial data do not suggest that advancing who receive continuous infusion of intragastric feeds, as all of
enteral feed volumes at slow rates (typically 15 ml/kg/day to 20 ml/ the infants in the included trials received enteral feeds as interval
kg/day) compared to faster rates (30 ml/kg/day to 35 ml/kg/day) boluses. Randomised controlled trials have reported conflicting
reduces the risk of NEC in very preterm or VLBW infants. The findings about the effect of continuous enteral infusion on feed
boundaries of the 95% CI for the estimate of effect are consistent tolerance in very preterm or VLBW infants (Premji 2011).
with either four extra or four fewer cases of NEC in every 100 Although the finding that slow enteral feed volume advancement
infants who have slow rates of feed advancement. Similarly, a meta- delays the establishment of full enteral feeds may seem intuitive,
analysis of data from these trials does not indicate an effect on it is also plausible that advancing feed volumes faster could have
all-cause mortality, with the 95% CI boundaries being consistent resulted in more feed intolerance and therefore a delay in the estab-
with either nine extra or one fewer death in every 100 infants who lishment of full enteral feeding. The included trials pre-specified
have slow rates of feed advancement. A pre-specified subgroup definitions of feed intolerance that mandated interrupting or ceas-
analysis showed a borderline statistically significant increase in risk ing feed volume advancement, principally the detection of ’gastric
of death with slow feeding in infants with growth restriction or residuals’ (the gastric content aspirated prior to a planned gastric
antenatal absent end-diastolic flow velocities. However, given that tube feed) and abdominal distension. However, the trial reports
this finding was based on data from only two small trials with a presented only limited data on the frequency of these outcomes.
total of 83 participants, it should be interpreted cautiously. Furthermore, there is only limited evidence that the volume or
Infants who had slow advancement of feed volumes regained their colour of gastric residuals is predictive of the risk of NEC for in-
birth weight several days later than infants who had faster rates fants whose feed volumes are advanced conservatively (Mihatsch
of advancement of feed volumes. The clinical importance of this 2002; Cobb 2004; Bertino 2009). Similarly, the clinical impor-
effect is unclear as long-term growth or developmental outcomes tance of abdominal distension or bowel loops visible through the
have not been assessed. Similarly, infants who had slow advance- abdominal wall (without other features of intra-abdominal pathol-
ment of feed volumes established full enteral feeding one to five ogy) is unclear, especially in the modern era, when early and pro-
days later than infants who had faster rates of advancement of feed longed use of continuous positive airway pressure results in in-
volumes. Whether this is associated with such important adverse testinal gaseous distension.
clinical consequences as a higher rate of nosocomial infection sec-
ondary to prolonged use of parenteral nutrition is not yet known,
as few studies have reported this outcome. Despite the effect on Quality of the evidence
the establishment of enteral feeding, the included trials did not
find consistent evidence of an important effect on the duration of The included trials were generally of good methodological qual-
hospital admission. ity, but, in common with other trials of feeding interventions in
this population, it was not possible to mask caregivers and clinical
assessors to the nature of the intervention (Figure 5). Although
the lack of blinding may have resulted in surveillance and ascer-
Overall completeness and applicability of tainment biases, it is more likely to have caused an overestimation
evidence of the incidence of feed intolerance and NEC in infants whose
These findings should be applied with caution for several reasons. feed volumes were advanced faster. The assessment of abdominal
Few of the trial participants were ELBW or extremely preterm radiographs for signs of NEC was masked in most trials to en-
infants. One-third of the participants in Karagol 2012 were of sure that the diagnosis of severe NEC (confirmed by the radio-
ELBW, but only a minority of infants in the two larger trials logical detection of gas in the bowel wall or portal tract) was not
weighed less than 1000 g or were less than 28 weeks gestation at prone to bias. However, since the microbial generation of gas in
birth, or had evidence of intrauterine growth restriction (Rayyis the bowel wall is substrate dependent, infants who received more

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 9
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
enteral milk (substrate) may have been more likely to demonstrate
this radiological sign than infants with equally severe bowel disease
who had less intraluminal substrate. This ’substrate effect’ is also
more likely to cause overascertainment of NEC in the infants who
had faster rates of feed volume advancement (Tyson 2007).

Figure 5. Risk of bias graph: review authors’ judgements about each risk of bias item presented as
percentages across all included studies.

Agreements and disagreements with other volume of enteral feeds at slow rates (less than 24 ml/kg/day)
studies or reviews results in several days delay in the time taken to regain birth weight
This review focused specifically on the comparison of slow ver- and establish full enteral feeds. Only limited data are available on
sus faster rates of feed volume advancement and did not compare the effect of this intervention on outcomes for extremely preterm
progressive advancement with enteral fasting or trophic feeding or ELBW infants or infants who are growth-restricted. Although
(minimal enteral nutrition). Only one randomised controlled trial current practice tends to favour a conservative approach to enteral
has compared trophic feeding with progressive enteral feed volume feeding in these populations, clinicians should consider that there
advancement (at daily increments of 20 ml/kg) (Berseth 2003). are other possible consequences of advancing feed volumes slowly,
Although the trial found the risk of NEC to be statistically sig- such as prolonging the use of parenteral nutrition, that may be
nificantly higher in the infants whose feed volumes were progres- associated with important adverse clinical outcomes.
sively advanced, this finding should be interpreted cautiously. The
trial was stopped early following an interim analysis, therefore the
Implications for research
finding of an effect on the incidence of NEC may be spurious
(Montori 2005). Caregivers and assessors were not blind to the Further randomised controlled trials could provide more precise
intervention. As discussed above, this may have resulted in several estimates of the effects of different rates of daily increases in en-
sources of bias that are likely to cause an overestimation of the inci- teral feed volumes on important outcomes for extremely preterm
dence of NEC in infants whose feed volumes are being advanced. or ELBW infants. Trials should aim to ensure the participation
of infants with evidence of compromised intrauterine growth, so
that subgroup analyses can be planned for this population at high
risk of NEC. Masking caregivers and investigators to the nature
of this intervention is likely not possible. Since the unblinded
AUTHORS’ CONCLUSIONS
evaluation of feed intolerance and NEC is subject to surveillance
and ascertainment biases, trials could aim to assess more objective
Implications for practice outcomes, principally mortality and long-term growth and devel-
These data suggest that advancing enteral feed volumes at daily opment. Furthermore, since conservative feeding strategies may
increments of up to 30 ml/kg to 35 ml/kg does not increase the result in such ’competing outcomes’ as invasive infection that may
risk of NEC in very preterm or VLBW infants. Increasing the affect long-term survival and neurodisability rates, it is essential
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 10
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
that trials are powered and structured to assess these outcomes.

ACKNOWLEDGEMENTS
We are grateful to Drs Namasivayam Ambalavanan and Kanya
Mukhopadhyay for providing further details and data (Rayyis
1999; Mukhopadhyay 2014).
This report is independent research funded by a UK National In-
stitute for Health Research Grant (NIHR) Cochrane Programme
Grant (13/89/12). The views expressed in this publication are
those of the authors and not necessarily those of the National
Health Service, the NIHR, or the UK Department of Health.

REFERENCES

References to studies included in this review References to studies excluded from this review
Caple 2004 {published data only} Berseth 2003 {published data only}
Caple J, Armentrout D, Huseby V, Halbardier B, Garcia J, Berseth CL, Bisquera JA, Paje VU. Prolonging small feeding
Sparks JW, et al.Randomized, controlled trial of slow versus volumes early in life decreases the incidence of necrotizing
rapid feeding volume advancement in preterm infants. enterocolitis in very low birth weight infants. Pediatrics
Pediatrics 2004;114(6):1597–600. [PUBMED: 15574620] 2003;111(3):529–34. [PUBMED: 12612232]
Karagol 2012 {published data only} Book 1976 {published data only}
Karagol BS, Zenciroglu A, Okumus N, Polin RA. Book LS, Herbst JJ, Jung AL. Comparison of fast- and slow-
Randomized controlled trial of slow versus rapid enteral feeding rate schedules to the development of necrotizing
feeding advancements on the clinical outcomes of preterm enterocolitis. Journal of Pediatrics 1976;89(3):462–6.
infants with 750-1250g. Journal of Parenteral and Enteral [PUBMED: 989057]
Nutrition 2013;37(2):223–8. [PUBMED: 22664861]
Additional references
Krishnamurthy 2010 {published data only}
Krishnamurthy S, Gupta P, Debnath S, Gomber S. Slow Beeby 1992
versus rapid enteral feeding advancement in preterm Beeby PJ, Jeffery H. Risk factors for necrotising enterocolitis:
newborn infants 1000-1499 g: a randomized controlled the influence of gestational age. Archives of Disease in
trial. Acta Paediatrica 2010;99(1):42–6. [PUBMED: Childhood 1992;67(4 Spec No):423–5. [PUBMED:
20002013] 1586186]
Mukhopadhyay 2014 {published data only} Bernstein 2000
Mukhopadhyay K, Jain S. Feed intolerance and necrotising Bernstein IM, Horbar JD, Badger GJ, Ohlsson A, Golan
enterocolitis in rapid vs slow feeding in preterm neonates A. Morbidity and mortality among very-low-birth-weight
with absent end diastolic flow: a randomised controlled neonates with intrauterine growth restriction. The Vermont
trial. Pediatric Academic Societies and Asian Society for Oxford Network. American Journal of Obstetrics and
Pediatric Research Joint Meeting. Vancouver, Canada: Gynecology 2000;182(1 Pt 1):198–206. [PUBMED:
Pediatric Academic Societies, 2014. 10649179]
Rayyis 1999 {published data only} Bertino 2009
Rayyis SF, Ambalavanan N, Wright L, Carlo WA. Bertino E, Giuliani F, Prandi G, Coscia A, Martano C,
Randomized trial of “slow” versus “fast” feed advancements Fabris C. Necrotizing enterocolitis: risk factor analysis and
on the incidence of necrotizing enterocolitis in very low role of gastric residuals in very low birth weight infants.
birth weight infants. Journal of Pediatrics 1999;134(3): Journal of Pediatric Gastroenterology and Nutrition 2009;48
293–7. [PUBMED: 10064664] (4):437–42. [PUBMED: 19330932]
Salhotra 2004 {published data only} Bisquera 2002
Salhotra A, Ramji S. Slow versus fast enteral feed Bisquera JA, Cooper TR, Berseth CL. Impact of necrotizing
advancement in very low birth weight infants: a randomized enterocolitis on length of stay and hospital charges in very
control trial. Indian Pediatrics 2004;41(5):435–41. low birth weight infants. Pediatrics 2002;109(3):423–8.
[PUBMED: 15181294] [PUBMED: 11875136]
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 11
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Bombell 2009 Holman 1997
Bombell S, McGuire W. Early trophic feeding for Holman RC, Stoll BJ, Clarke MJ, Glass RI. The
very low birth weight infants. Cochrane Database of epidemiology of necrotizing enterocolitis infant mortality in
Systematic Reviews 2009, Issue 3. [DOI: 10.1002/ the United States. American Journal of Public Health 1997;
14651858.CD000504.pub3; PUBMED: 19588318] 87(12):2026–31. [PUBMED: 9431297]
Brown 1978 Härtel 2009
Brown EG, Sweet AY. Preventing necrotizing enterocolitis Härtel C, Haase B, Browning-Carmo K, Gebauer C, Kattner
in neonates. JAMA 1978;240(22):2452–4. E, Kribs A, et al.Does the enteral feeding advancement
Chauhan 2008 affect short-term outcomes in very low birth weight infants?
Chauhan M, Henderson G, McGuire W. Enteral . Journal of Pediatric Gastroenterology and Nutrition 2009;48
feeding for very low birth weight infants: reducing the (4):464–70. [PUBMED: 19322056]
risk of necrotising enterocolitis. Archives of Disease in Luig 2005
Childhood. Fetal and Neonatal Edition 2008;93(2):F162–6. Luig M, Lui K, NSW & ACT NICUS Group.
[PUBMED: 18006565] Epidemiology of necrotizing enterocolitis--Part II: Risks
Cobb 2004 and susceptibility of premature infants during the surfactant
Cobb BA, Carlo WA, Ambalavanan N. Gastric residuals era: a regional study. Journal of Paediatrics and Child Health
and their relationship to necrotizing enterocolitis in very 2005;41(4):174–9. [PUBMED: 15813870]
low birth weight infants. Pediatrics 2004;113(1 Pt 1):50–3. McKeown 1992
[PUBMED: 14702446] McKeown RE, Marsh TD, Amarnath U, Garrison CZ,
Dorling 2005 Addy CL, Thompson SJ, et al.Role of delayed feeding and
Dorling J, Kempley S, Leaf A. Feeding growth restricted of feeding increments in necrotizing enterocolitis. Journal of
preterm infants with abnormal antenatal Doppler results. Pediatrics 1992;121(5 Pt 1):764–70. [PUBMED: 1432431]
Archives of Disease in Childhood. Fetal and Neonatal Edition
Mihatsch 2002
2005;90(5):F359–63. [PUBMED: 16113150]
Mihatsch WA, von Schoenaich P, Fahnenstich H, Dehne
Flidel-Rimon 2004 N, Ebbecke H, Plath C, et al. The significance of
Flidel-Rimon O, Friedman S, Lev E, Juster-Reicher gastric residuals in the early enteral feeding advancement of
A, Amitay M, Shinwell ES. Early enteral feeding and extremely low birth weight infants. Pediatrics 2002;109(3):
nosocomial sepsis in very low birthweight infants. Archives 457–9. [PUBMED: 11875141]
of Disease in Childhood. Fetal and Neonatal Edition 2004;89
Montori 2005
(4):F289–92. [PUBMED: 15210657]
Montori VM, Devereaux PJ, Adhikari NK, Burns KE,
Flidel-Rimon 2006
Eggert CH, Briel M, et al.Randomized trials stopped early
Flidel-Rimon O, Branski D, Shinwell ES. The fear of
for benefit: a systematic review. JAMA 2005;294(17):
necrotizing enterocolitis versus achieving optimal growth in
2203–9. [PUBMED: 16264162]
preterm infants--an opinion. Acta Paediatrica 2006;95(11):
1341–4. [PUBMED: 17062457] Morgan 2011
Morgan J, Young L, McGuire W. Delayed introduction of
Garite 2004
progressive enteral feeds to prevent necrotising enterocolitis
Garite TJ, Clark R, Thorp JA. Intrauterine growth
in very low birth weight infants. Cochrane Database
restriction increases morbidity and mortality among
of Systematic Reviews 2011, Issue 3. [DOI: 10.1002/
premature neonates. American Journal of Obstetrics and
14651858.CD001970.pub3; PUBMED: 21412877]
Gynecology 2004;191(2):481–7. [PUBMED: 15343225]
Guthrie 2003 Patole 2005
Guthrie SO, Gordon PV, Thomas V, Thorp JA, Peabody J, Patole SK, de Klerk N. Impact of standardised feeding
Clark RH. Necrotizing enterocolitis among neonates in the regimens on incidence of neonatal necrotising enterocolitis:
United States. Journal of Perinatology 2003;23(4):278–85. a systematic review and meta-analysis of observational
[PUBMED: 12774133] studies. Archives of Disease in Childhood. Fetal and Neonatal
Edition 2005;90(2):F147–51. [PUBMED: 15724039]
Henderson 2009
Henderson G, Craig S, Brocklehurst P, McGuire W. Enteral Pike 2012
feeding regimens and necrotising enterocolitis in preterm Pike K, Brocklehurst P, Jones D, Kenyon S, Salt A, Taylor
infants: a multicentre case-control study. Archives of Disease D, et al.Outcomes at 7 years for babies who developed
in Childhood. Fetal and Neonatal Edition 2009;94(2): neonatal necrotising enterocolitis: the ORACLE Children
F120–3. [PUBMED: 17768154] Study. Archives of Disease in Childhood. Fetal and Neonatal
Higgins 2011 Edition 2012;97(5):F318–22. [PUBMED: 22933088]
Higgins JPT, Green S (editors). Cochrane Handbook Premji 2011
for Systematic Reviews of Interventions Version 5.1.0 Premji SS, Chessell L. Continuous nasogastric milk feeding
[updated March 2011]. The Cochrane Collaboration, versus intermittent bolus milk feeding for premature
2011. Available from www.cochrane-handbook.org. infants less than 1500 grams. Cochrane Database of
Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 12
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Systematic Reviews 2011, Issue 11. [DOI: 10.1002/ low birth weight infants: biodemographic and clinical
14651858.CD001819.pub2; PUBMED: 22071802] correlates. National Institute of Child Health and Human
Quigley 2014 Development Neonatal Research Network. Journal of
Quigley MA, McGuire W. Formula versus donor breast Pediatrics 1991;119(4):630–8. [PUBMED: 1919897]
milk for feeding preterm or low birth weight infants. Walsh 1986
Cochrane Database of Systematic Reviews 2014, Issue 4. Walsh MC, Kliegman RM. Necrotizing enterocolitis:
[DOI: 10.1002/14651858.CD002971.pub2; PUBMED: treatment based on staging criteria. Pediatric Clinics of North
24752468] America 1986;33(1):179–201. [PUBMED: 3081865]
Rees 2007
Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes References to other published versions of this review
of neonates with medically and surgically treated necrotizing
enterocolitis. Archives of Disease in Childhood. Fetal
and Neonatal Edition 2007;92(3):F193–8. [PUBMED: Kennedy 2005
16984980] Kennedy KA, Tyson JE, Chamnanvanakij S. Rapid versus
RevMan 2011 slow rate of advancement of feedings for promoting growth
The Nordic Cochrane Centre, The Cochrane Collaboration. and preventing necrotizing enterocolitis in parenterally
Review Manager (RevMan). 5.1. Copenhagen: The Nordic fed low-birth-weight infants. Cochrane Database of
Cochrane Centre, The Cochrane Collaboration, 2011. Systematic Reviews 2005, Issue 2. [DOI: 10.1002/
14651858.CD001241.pub2]
Soraisham 2006
Soraisham AS, Amin HJ, Al-Hindi MY, Singhal N, McGuire 2008
Sauve RS. Does necrotising enterocolitis impact the McGuire W, Bombell S. Slow advancement of enteral
neurodevelopmental and growth outcomes in preterm feed volumes to prevent necrotising enterocolitis in
infants with birthweight < or =1250 g?. Journal of Paediatrics very low birth weight infants. Cochrane Database of
and Child Health 2006;42(9):499–504. [PUBMED: Systematic Reviews 2008, Issue 2. [DOI: 10.1002/
16925534] 14651858.CD001241.pub2; PUBMED: 18425870]
Stoll 2004
Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Morgan 2011a
Hintz SR, Vohr B, et al.Neurodevelopmental and growth Morgan J, Young L, McGuire W. Slow advancement of
impairment among extremely low-birth-weight infants enteral feed volumes to prevent necrotising enterocolitis
with neonatal infection. JAMA 2004;292(19):2357–65. in very low birth weight infants. Cochrane Database
[PUBMED: 15547163] of Systematic Reviews 2011, Issue 3. [DOI: 10.1002/
14651858.CD001241.pub3; PUBMED: 21412870]
Tyson 2007
Tyson JE, Kennedy KA, Lucke JF, Pedroza C. Dilemmas Morgan 2013
initiating enteral feedings in high risk infants: how can they Morgan J, Young L, McGuire W. Slow advancement of
be resolved?. Seminars in Perinatology 2007;31(2):61–73. enteral feed volumes to prevent necrotising enterocolitis
[PUBMED: 17462490] in very low birth weight infants. Cochrane Database
Uauy 1991 of Systematic Reviews 2013, Issue 3. [DOI: 10.1002/
Uauy RD, Fanaroff AA, Korones SB, Phillips EA, 14651858.CD001241.pub4; PUBMED: 23543511]
Phillips JB, Wright LL. Necrotizing enterocolitis in very ∗
Indicates the major publication for the study

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 13
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
CHARACTERISTICS OF STUDIES

Characteristics of included studies [ordered by study ID]

Caple 2004

Methods Randomised controlled trial

Participants Preterm infants of birth weight 1000 g to 2000 g (appropriate birth weight for gestational
age and of gestational age < 35 weeks at birth), who were starting formula feeds
Setting: Neonatal Unit, Department of Pediatrics, University of Texas Medical School,
Houston, Texas, USA

Interventions Feeds advancement at 20 ml/kg/day (N = 74) versus 30 ml/kg/day (N = 84)

Outcomes NEC (Bell stage II/III)


Time to regain birth weight, time to achieve full enteral feeds, and time to hospital
discharge

Notes Feeds were ceased if the residual gastric aspirate was more than one-third of the previous
feed volume, or if there was frequent vomiting, abdominal distention, or bloody stools
(including occult blood)
We have not been able to obtain data on all-cause mortality from the principal investi-
gators

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Random number sequence


bias)

Allocation concealment (selection bias) Low risk Blinded draw from envelope by caregivers not involved
in study

Blinding (performance bias and detection High risk Caregivers and clinical investigators were not blinded
bias) once allocation to intervention groups had occurred
Clinical assessments

Blinding (performance bias and detection Low risk Radiologists interpreting X-rays were blinded to the in-
bias) tervention group
Radiological assessments

Incomplete outcome data (attrition bias) Low risk Three infants excluded after enrolment because of proto-
All outcomes col violations have been included in this review and meta-
analysis. Two infants (one in each group) were excluded
because they were determined not eligible for enrolment
because of an in utero gastrointestinal perforation and
fetal alcohol syndrome; these infants were not included
in the meta-analysis

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 14
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Karagol 2012

Methods Randomised controlled trial

Participants Preterm infants < 32 weeks gestation with birth weights of 750 g to 1250 g. 32% of
participants weighed < 1000 g. Exclusion criteria included major congenital malforma-
tions, severe respiratory distress, presence of umbilical vessel catheters, contraindications
to enteral feeding, perinatal asphyxia, or cardiovascular compromise
Setting: Division of Neonatology, Dr Sami Ulus Maternity, Children’s Education and
Research Hospital, Ankara, Turkey

Interventions Slow advancement at 20 ml/kg/day (N = 46) versus rapid advancement at 30 ml/kg/day


(N = 46)

Outcomes NEC (stage II/III), all-cause mortality, time to regain birth weight, time to reach full
enteral feeds, feed intolerance, duration of hospital stay, rates of invasive infection
Subgroup analysis for ELBW infants

Notes Feeds were ceased if any of the following occurred: gastric residuals > 5 ml/kg or > 50%
of feed volume, vomiting > 3 times in 24 hours, increase in abdominal girth > 2 cm
between feeds, abdominal tenderness or erythema, reduced bowel sounds, blood in the
stools, or recurrent apnoea

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Computer-generated sequence


bias)

Allocation concealment (selection bias) Low risk Opaque, sealed envelopes

Blinding (performance bias and detection High risk Caregivers and study investigators were not blinded.
bias)
Clinical assessments

Blinding (performance bias and detection Unclear risk No reference to whether staff interpreting radiological
bias) images were blinded to the study groups
Radiological assessments

Incomplete outcome data (attrition bias) Low risk No participants lost to follow-up
All outcomes

Krishnamurthy 2010

Methods Randomised controlled trial

Participants Preterm infants (birth weight 1000 g to 1499 g) and gestational age < 34 weeks at birth.
Exclusion criteria included respiratory distress, mechanical ventilation, inotrope support,
and umbilical arterial or venous catheterisation

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 15
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Krishnamurthy 2010 (Continued)

Setting: Department of Paediatrics, University College of Medical Sciences, Dehli, India

Interventions Feeds advancement at 20 ml/kg/day (N = 50) versus 30 ml/kg/day (N = 50)

Outcomes NEC (stage II/III)


Incidence of nosocomial infection, in-hospital mortality, time to regain birth weight,
time to achieve full enteral feeds, and time to hospital discharge

Notes All feeds were delivered by gavage via nasogastric tubes at 2-hour intervals
Feeds were ceased if any of the following occurred: residual gastric contents more than
50% of the previous feed volume (delayed if volume was 25% to 50%); more than 3
episodes of apnoea in the preceding hour; abdominal distention or tenderness; or bloody
stools (including occult blood)
Parenteral nutrition was not available.

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Computer-generated sequence


bias)

Allocation concealment (selection bias) Low risk Opaque, sealed envelopes

Blinding (performance bias and detection High risk Caregivers and investigators were not blinded to the in-
bias) terventions
Clinical assessments

Blinding (performance bias and detection Low risk Radiologist interpreting X-rays was blinded to the inter-
bias) vention group
Radiological assessments

Incomplete outcome data (attrition bias) Low risk No loss to follow-up


All outcomes

Mukhopadhyay 2014

Methods Randomised controlled trial

Participants Preterm infants (birth weight 1000 g to 1249 g) and gestational age > 30 weeks at
birth who have antenatal evidence of absent end diastolic flow velocities (presumed in
umbilical artery)
Setting: Department of Paediatrics, Postgraduate Institute of Medical Education & Re-
search, Chandigarh, India

Interventions Feeds advancement at 20 ml/kg/day (N = 15) versus 30 ml/kg/day (N = 15)

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 16
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Mukhopadhyay 2014 (Continued)

Outcomes NEC (all stages)


Late-onset bloodstream (culture-positive) infection, in-hospital mortality, time to achieve
full enteral feeds

Notes Pre-specified subgroup of larger trial that enrolled infants with birth weight > 1250 g
and compared feed advancement at 30 ml/kg/day versus 40 ml/kg/day
Published as conference abstract; further data courtesy of Dr Mukhopadhyay (September
2014)

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Computer-generated


bias)

Allocation concealment (selection bias) Low risk Sealed, opaque envelopes

Blinding (performance bias and detection High risk Caregivers and investigators were not blinded to the in-
bias) terventions
Clinical assessments

Blinding (performance bias and detection Unclear risk Not stated


bias)
Radiological assessments

Incomplete outcome data (attrition bias) Low risk Complete follow-up for primary outcomes
All outcomes

Rayyis 1999

Methods Randomised controlled trial

Participants Very low birth weight infants of gestational age < 34 weeks at birth
Setting: Neonatal Unit, Department of Pediatrics, University of Alabama, Birmingham,
Alabama, USA

Interventions Feeds advancement at 15 ml/kg/day (N = 98) versus 35 ml/kg/day (N = 87)

Outcomes NEC (stage II/III)


Time to regain birth weight, time to achieve full enteral feeds, and time to hospital
discharge

Notes Infants for whom full or partial feeding with expressed breast milk was planned were not
eligible to participate. Feeding was commenced using standard ’term’ artificial formula,
then switched to nutrient-enriched ’preterm’ formula when full enteral feeding had been
achieved.Feeds were ceased if any of the following occurred: residual gastric contents
more than 30% of the previous feed volume, abdominal distention or tenderness, or

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 17
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Rayyis 1999 (Continued)

bloody stools (including occult blood)

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Unclear risk Not stated


bias)

Allocation concealment (selection bias) Low risk Opaque, sealed envelopes

Blinding (performance bias and detection High risk Caregivers and investigators not blinded to the interven-
bias) tion groups
Clinical assessments

Blinding (performance bias and detection Low risk Radiologist interpreting the X-rays was blinded to the
bias) study group
Radiological assessments

Incomplete outcome data (attrition bias) Low risk 7 protocol violations occurred after enrolment, but all
All outcomes infants were included the final data analysis

Salhotra 2004

Methods Randomised controlled trial

Participants Preterm infants of birth weight < 1250 g. More than 95% of the participants were ’small
for gestational age’. Exclusion criteria included recurrent apnoea, respiratory distress
requiring supplemental oxygen, and receipt of inotrope support
Setting: Neonatal Unit , Maulana Azad Medical College (tertiary-level teaching hospital)
, New Delhi, India

Interventions Feeds advancement at 15 ml/kg/day (N = 26) versus 30 ml/kg/day (N = 27)

Outcomes NEC (stage II/III)


Neonatal mortality, time to regain birth weight, time to achieve full enteral feeds, and
time to hospital discharge

Notes Feeds were ceased if the residual gastric content was more than 30% of the previous feed
volume or if there was abdominal distention
Mortality data courtesy of Dr Namasivayam Ambalavanan

Risk of bias

Bias Authors’ judgement Support for judgement

Random sequence generation (selection Low risk Computer-generated sequence


bias)

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 18
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Salhotra 2004 (Continued)

Allocation concealment (selection bias) Low risk Opaque, sealed envelopes

Blinding (performance bias and detection High risk Investigators were blinded at allocation stage, but it is
bias) unclear if they remained blinded thereafter. Caregivers
Clinical assessments were not blinded to intervention group

Blinding (performance bias and detection Unclear risk No statement about blinding of radiological assessors to
bias) intervention group
Radiological assessments

Incomplete outcome data (attrition bias) Low risk No losses to follow-up


All outcomes

Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion

Berseth 2003 Infants were randomly allocated to either a stable (not progressively increased) trophic feeding volume or to feed
volume advancement at 20 ml/kg/day

Book 1976 Enteral feeding volumes were advanced at 10 ml/kg/day versus 20 ml/kg/day, that is both groups received ’slow’
advancement of feed volumes

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 19
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
DATA AND ANALYSES

Comparison 1. Slow versus faster rates of feed advancement

No. of No. of
Outcome or subgroup title studies participants Statistical method Effect size

1 Incidence of necrotising 6 Risk Ratio (M-H, Fixed, 95% CI) Subtotals only
enterocolitis
1.1 All trials 6 618 Risk Ratio (M-H, Fixed, 95% CI) 0.96 [0.55, 1.70]
1.2 Trials in which most 2 83 Risk Ratio (M-H, Fixed, 95% CI) 0.34 [0.06, 2.04]
infants were small for
gestational age or growth
restricted
2 Mortality 5 Risk Ratio (M-H, Fixed, 95% CI) Subtotals only
2.1 All trials 5 460 Risk Ratio (M-H, Fixed, 95% CI) 1.57 [0.92, 2.70]
2.2 Trials in which most 2 83 Risk Ratio (M-H, Fixed, 95% CI) 2.13 [1.02, 4.47]
infants were small for
gestational age or growth
restricted
3 Feeds intolerance (causing 4 275 Risk Ratio (M-H, Fixed, 95% CI) 1.23 [0.87, 1.73]
interruption of enteral feeding)
4 Incidence of invasive infection 3 222 Risk Ratio (M-H, Fixed, 95% CI) 1.58 [0.81, 3.09]

WHAT’S NEW
Last assessed as up-to-date: 3 October 2014.

Date Event Description

3 October 2014 New search has been performed This updates the review ’Slow advancement of enteral
feed volumes to prevent necrotising enterocolitis in very
low birth weight infants’ (Morgan 2013).

3 October 2014 New citation required but conclusions have not changed Updated search in September 2014 identified one new
trial for inclusion (Mukhopadhyay 2014), increasing the
total number of participating infants to 618 (from 588)
and narrowing confidence intervals for the estimates of
effect

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 20
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
HISTORY
Protocol first published: Issue 4, 1998
Review first published: Issue 4, 1998

Date Event Description

11 January 2011 New citation required and conclusions have changed Addition of new data, and increasing the total number
of participating infants to 496, narrowed the confi-
dence intervals for the estimates of effect and modified
the implications for practice and research

15 December 2010 New search has been performed This updates the review “Slow advancement of en-
teral feed volumes to prevent necrotising enterocoli-
tis in very low birth weight infants” published in the
Cochrane Database of Systematic Reviews, Issue 2,
2008 (McGuire 2008).
Search updated in December 2010. One new trial in-
cluded (Krishnamurthy 2010).
New co-authors: Jessie Morgan and Lauren Young.

13 February 2008 New citation required but conclusions have not New authorship: Bombell S, McGuire W
changed

2 February 2008 New search has been performed This updates the review “Rapid versus slow rate of
advancement of feedings for promoting growth and
preventing necrotizing enterocolitis in parenterally fed
low-birth-weight infants” by Kennedy and Tyson,
published in the Cochrane Database of Systematic Re-
views, Issue 2, 2000 (Kennedy 2000)
The title has been modified to read “Slow advancement
of enteral feed volumes to prevent necrotising entero-
colitis in very low birth weight infants” and has a new
authorship of Sarah Bombell and William McGuire.
Changes made to the original protocol are outlined
below:

1. “Slow” rate of feed advancement has been defined


as daily increments up to 24 ml/kg (body weight).
2. The population has been restricted to very low birth
weight and very preterm infants.
3. Mortality, adverse neurodevelopment, growth pa-
rameters, and infection rates have been included as
outcomes of interest.

Search updated December 2007. One new trial has


been included (Salhotra 2004). One trial previously
included has now been excluded (Book 1976).

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 21
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
(Continued)

The findings and implications for practice and research


of the review have not changed overall

11 January 2008 Amended Converted to new review format.

CONTRIBUTIONS OF AUTHORS
Drs Young and McGuire updated the search, independently determined the eligibility of identified studies, assessed the methodological
quality of the included trials, and extracted the relevant information and data.

DECLARATIONS OF INTEREST
None

SOURCES OF SUPPORT
Internal sources
• Centre for Reviews and Dissemination, Hull York Medical School, UK.

External sources
• National Institute for Health Research (NIHR), UK.
NIHR Cochrane Programme Grant (13/89/12)
• Eunice Kennedy Shriver National Institute of Child Health and Human Development National Institutes of Health,
Department of Health and Human Services, USA.
Editorial support of the Cochrane Neonatal Review Group has been funded with Federal funds from the Eunice Kennedy Shriver
National Institute of Child Health and Human Development National Institutes of Health, Department of Health and Human
Services, USA, under Contract No. HHSN267200603418C.

INDEX TERMS

Medical Subject Headings (MeSH)


∗ Infant,
Very Low Birth Weight; Enteral Nutrition [∗ methods]; Enterocolitis, Necrotizing [∗ prevention & control]; Infant, Low Birth
Weight; Infant, Newborn; Infant, Premature; Infant, Premature, Diseases [∗ prevention & control]; Parenteral Nutrition [adverse effects];
Randomized Controlled Trials as Topic

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 22
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
MeSH check words
Humans

Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants (Review) 23
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Review article
Kim Enteral2016;59(12):466-470
MJ J•Pediatr
Korean nutrition
https://doi.org/10.3345/kjp.2016.59.12.466
pISSN 1738-1061•eISSN 2092-7258
Korean J Pediatr

Enteral nutrition for optimal growth in preterm


infants
Myo-Jing Kim, MD
Department of Pediatrics, Dong-A University College of Medicine, Busan, Korea

Early, aggressive nutrition is an important contributing factor of long-term neurodevelopmental Corresponding author: Myo-Jing Kim, MD
outcomes. To ensure optimal growth in premature infants, adequate protein intake and optimal protein/ Department of Pediatrics, Dong-A University Col-
lege of Medicine, 32 Daesingongwon-ro, Seo-gu,
energy ratio should be emphasized rather than the overall energy intake. Minimal enteral nutrition should Busan 49201, Korea
be initiated as soon as possible in the first days of life, and feeding advancement should be individualized Tel: +82-51-240-2589
according to the clinical course of the infant. During hospitalization, enteral nutrition with preterm formula Fax: +82-51-242-2765
and fortified human milk represent the best feeding practices for facilitating growth. After discharge, the E-mail: myojing@dau.ac.kr
enteral nutrition strategy should be individualized according to the infant’s weight at discharge. Infants Received: 9 September, 2015
with suboptimal weight for their postconceptional age at discharge should receive supplementation with Revised: 13 October, 2015
human milk fortifiers or nutrient-enriched feeding, and the enteral nutrition strategy should be reviewed Accepted: 16 November, 2015
and modified continuously to achieve the target growth parameters.

Key words: Enteral nutrition, Infant, Premature

Introduction
The current recommendations for nutrition of preterm infants are based on the goal of
achieving the growth rate and body composition of a normal fetus at the same postmens­
trual age1). However, this goal is often difficult to achieve because of the physiologic limita­
tions associated with prematurity. Postnatal growth restrictions remain a clinical complica­
tion of prematurity, especially in extremely low birth weight infants (ELBWI)2). Inadequate
nutrition has long been considered a cause of growth retardation. The association between
postnatal growth failure due to inadequate nutrition and impaired long-term neurocogni­
tive development is of particular concern. Therefore, a strategy for providing adequate
nutrition to ensure optimal growth in premature infants needs to be emphasized.

Principles of enteral nutrition


The American Academy of Pediatrics (AAP) and the European Society of Paediatric
Gastroenterology, Hepatology, and Nutrition (ESPGHAN) Committee on Nutrition recom­
mended an energy intake of 105–130 kcal/kg/day3) and 110–135 kcal/kg/day4) for preterm
infants, respectively. Infants sometimes require increased caloric intake owing to a higher Copyright © 2016 by The Korean Pediatric Society
energy demand during an illness. Thus, simply meeting these recommended energy re­
quirements may not be sufficient. Failure to provide adequate protein can result in adverse This is an open-access article distributed under the
terms of the Creative Commons Attribution Non-
long-term outcomes in premature infants. Each additional gram of protein intake for Commercial License (http://creativecommons.org/
licenses/by-nc/4.0/) which permits unrestricted non-
ELBWI is associated with additional weight gains of 6.5 g/day and head circumference commercial use, distribution, and reproduction in any
increase of 0.4 cm/wk5). The National Institute of Child Health and Human Development’s medium, provided the original work is properly cited.

466 https://doi.org/10.3345/kjp.2016.59.12.466
Korean J Pediatr 2016;59(12):466-470

Neonatal Research Network evaluated the association between increase >0.9 cm/wk during the hospitalization period are con­
weight gain in the neonatal intensive care unit (NICU) and long- sidered optimal6). If these rates cannot be met, the infant’s enteral
term neurodevelopmental outcomes in ELBWI. Infants in the nutrition should be reviewed and modified to achieve this optimal
lowest quartile of in-hospital weight gain (≤12 g/kg/day) were as­ goal. Focus should be placed on the protein content, P/E ratio,
sociated with higher odds of cerebral palsy, Bayley scales of In­ and utilization of nutrient-enriched feeding sources to reduce
fant Development II mental developmental index <70, and nutritional deficits.
neurodevelopmental impairment at 18–22 months of corrected
age, as compared with infants in the highest quartile of in-
hospital weight gain (≥21 g/kg/day). Similar findings have also Practice of enteral nutrition
been reported for in-hospital head circumference growth6).
These findings suggest that adequate protein intake can im­ 1. Minimal enteral feeding
prove the growth of ELBWI and that adequate growth in the Although trophic or minimal enteral feedings involve hypo­
NICU is associated with a lower risk of long-term neurodevelop­ caloric, low-volume (typically ≤24 mL/kg/day) nutrition to ac­
mental outcomes. Moreover, an increased protein/energy (P/E) celerate gastrointestinal, physiological, endocrine, and meta­bolic
ratio is mandatory to ensure quality growth, presenting as in­ maturity, they do not contain sufficient calories to sustain soma­
creased lean body mass and limited fat mass deposits. The tic growth8). The most common reason for delays in enteral feed­
ESPGHAN recommends protein intakes of 4.0–4.5 and 3.5–4.0 g/ ing is the clinicians’ concern for the risk of necrotizing entero­
kg/day in preterm infants weighing up to 1,000 g and 1,000– colitis (NEC). A recent meta-analysis evaluated the effects of early
1,800 g, respectively4). With optimal infant growth status, the trophic feeding versus enteral fasting in very low birth weight
protein intake can be reduced in anticipation of hospital dis­ infants (VLBWI), and found no differences in the feeding tole­
charge. However, achieving the recommended protein intake by rance, time to reach full enteral feeding, and incidence of NEC9).
enteral nutrition remains a difficult task. The protein intake by Thus, the authors concluded that minimal enteral nutrition was
enteral feeding at 150 mL/kg/day (routine enteral volume) is not associated with important beneficial or harmful effects in
insufficient for meeting the infant’s requirements of 1.8–2.1 g/kg/ VLBWI and confirmed the absence of reasons for not starting tro­
day of unfortified preterm human milk, 1.5 g/kg/day of unforti­ phic feeding as soon as possible.
fied donor human milk, or 3.6 g/kg/day of preterm formula7).
Hence, the question remains as to how much we need to feed 2. Introduction of progressive enteral feedings
ELBWI in order to achieve the recommended daily protein intake. The introduction of enteral feeds for VLBWI is often delayed
To provide 4 g/kg/day of protein, we need to feed the infant for several days or longer after birth because of concerns that
290–340 mL/kg/day of unfortified preterm human milk, 210 mL/ early introduction may not be tolerated and/or may increase the
kg/day of fortified human milk (1 pack/60 mL, Maeil Babywell risk of NEC. A systemic review evaluated the effects of early feed­
Human Milk Fortifier, Maeil Co. Ltd., Seoul, Korea), or 170 (16% ing introduction (<4 days) versus delayed feeding introduc­tion
concentration) to 190 mL/kg/day (14% concentration) of preterm (4–7 days) and found no significant differences in terms of the
formula (Maeil Babywell Preemie, Maeil Co. Ltd.; Namyang incidence of NEC or all-cause mortality. However, infants with
Premie Formula, Namyang Dairy Products Co., Ltd., Seoul, delayed feeding introduction required longer periods before
Korea). It is important to note that osmolarity and the renal solute achieving full enteral feedings and delays in hospitaldischarge10).
load influence the fluid volumes that are necessary for enteral
nutrition. The ESPGHAN reported that 135 and 2004) mL/kg/day 3. Rate of advancement of enteral feedings
should be considered the minimum and maximum fluid volume, The appropriate rate of enteral feeding advancement has also
respectively. Hence, fortification for human milk-fed infants and been the subject of much debate, and an association between
adjustment of the formula concentration for preterm formula-fed rapidly advanced enteral feeding and the risk of NEC has been
infants are needed. For routine enteral feeding, preterm formula suggested. However, a recent Cochrane review evaluated the ef­
or fortified human milk that is fed at rates of 150–180 mL/kg/day fects of the rate of enteral feeding advancement on the incidence
is sufficient for attaining the recommended nutrient require­ of NEC, all-cause mortality, and other morbidities in VLBWI and
ments4). found no significant differences between the rates. In the slow
Moreover, as growth in the NICU is associated with long-term advancement group, the infants required a longer period to regain
neurodevelopmental outcomes, the goal of growth in the NICU the birth weight and to reach full enteral feeding11). One reason­
may be questioned. able approach is to advance enteral feeds by 20–30 mL/kg/day in
For better neurodevelopmental and growth outcomes in VLBWI and by 15–25 mL/kg/day in ELBWI7).
ELBWI, weight gains of >18 g/kg/day and head circumference

https://doi.org/10.3345/kjp.2016.59.12.466 467
Kim MJ • Enteral nutrition

Choice of enteral nutrition to be the best solution for inadequate protein intake with standard
fortification14). Currently, two methods have been proposed for
1. Human milk individualization. Evaluated through periodic determinations of
Human milk is the optimal primary nutritional source for pre­ the blood urea nitrogen, targeted and adjustable fortifications are
term infants because it offers strong protection against sepsis and designed based on the analyses of the mother’s own milk and
other infections and offers short-term protection against NEC. de5pend on the metabolic response of each infant, respectively.
Regarding the long-term effects, human milk improves neurode­ The use of individualized fortification is recommended to allow
velopmental outcomes, and has a dose-dependent effect until 30 adequate protein intake and growth15). The compositions of the
months of corrected age12). Accordingly, the AAP strongly recom­ commercially available HMFs16) in Korea are shown in Table 1.
mends the use of human milk for preterm infants3). Lactation
strategies that increase the mother’s own milk production should 2. Preterm formulas
be emphasized in order to enable pumping immediately after Preterm formula is available for preterm infants who are un­
delivery1). In the absence of maternal human milk, pasteurized able to receive their mother’s own milk and donor human milk.
donor human milk is a good option for initiating feeding13). How­ In general, these formulas predominantly contain whey protein,
ever, despite these benefits, human milk does not completely meet glucose polymers, medium-chain triglycerides, calcium, and
the nutritional needs of preterm infants, owing to insuffici­ent phosphorus, and they are variably enriched with minerals,
calories and proteins necessary to support the optimal growth vitamins, and trace elements to support intrauterine nutrient
and lean body mass accretion. In addition, the concentrations of accretion rates. The compositions of the commercially available
calcium and phosphorus in human milk are also significantly formulas for preterm infants in Korea are shown in Table 2. In
lower than the required levels7). practice, a preterm formula of 70–72 kcal/100 mL is initially used.
For these reasons, supplementation (fortification) with multinu­ If tolerated by the infants, a higher energy formula consisting of
trient human milk fortifiers (HMFs) is needed in order to meet the 16% concentrated preterm formula (80–82 kcal/100 mL) may be
nutritional requirements and improve postnatal growth. HMFs used to meet the energy demand.
are indicated in preterm infants aged less than 31 gestational
weeks and/or with a birth weight less than 1,500 g. No definite
guidelines regarding the timing for starting HMFs currently exist.
Postdischarge nutrition
However, fortification of human milk does not need to be delayed
until full enteral feedings achieved. Generally, fortification com­ At the time of birth, approximately 20% of VLBWI are small
mences when the enteral feeding volume reaches 100 mL/kg/day for gestational age. After NICU care, however, 79% of these
and is gradually increased from half to full strength. Whereas the infants experience poor growth and weigh less than the 10th per­
protein content of human milk declines over the first few weeks centile at 36 weeks’ postconceptional age (PCA)17). Achieving
from 1.7 g/dL at 7 days to 1.2 g/dL at 42 days, commercial HMFs postdischarge catch-up growth, especially during the vital period
assume an average protein content of human milk of 1.4–1.6 g/ between 40 and 48 weeks PCA, is critical for optimal neurode­
dL7). Therefore, protein content from standard fortification does velopment in premature infants. The rate of weight gain is depen­
not reach the amount present in preterm formula, thereby re­ dent on the absolute intakes of protein and energy. A high P/E
sulting in slower growth compared to preterm formula-fed in­ ratio is required to increase lean body mass and protein accretion
fants. Consequently, individualized fortification is now believed and to limit fat mass deposition1). Major strategies to reach this
goal include continuing preterm formula, human milk alone/
Table 1. Comparison between the nutrient composition of preterm
human milk and Korean human milk fortifier
Table 2. Comparison between the nutrient composition of Korean
Maeil Babywell Human Milk Fortifier premature formulas (per 100 mL)
Preterm human milk
Nutrients 1.6 pack HMF+PHM (1 pack/60 mL) Maeil Babywell Preemie Namyang Premie Formula
/100 kcal /100 mL /100 kcal /100 mL Nutrients
14% 16% 14% 16%
Calory (kcal) 100 67 14 100 71 Calory (kcal) 70 80 72 82
Protein (g) 2.1 1.4 0.8 2.8 1.9 Protein (g) 2.1 2.4 2.1 2.4
P/E ratio 2.1 2.1 - 2.8 2.7 P/E ratio 3.0 3.0 2.9 2.9
Fat (g) 5.8 3.9 0.32 5.2 3.7 Fat (g) 3.6 4.1 3.8 4.3
Carbohydrate (g) 9.9 6.6 1.9 10.6 7.5 Carbohydrate (g) 7.4 8.5 7.3 8.3
Volume (mL) 149 100 15.5 142 100 Modified from Korean Society of Neonatology. Manual of neonatal care. 3rd
HMF, human milk fortifier; PHM, preterm human milk; P/E, protein/energy. ed., with permission of The Korean Society of Neonatology16).

468 https://doi.org/10.3345/kjp.2016.59.12.466
Korean J Pediatr 2016;59(12):466-470

partially, full nutrient-fortified human milk, nutrient-enriched should be breastfed when possible. When formula-fed, such in­
postdischarge formula (PDF), and term formula. fants should be fed standard formula with long-chain polyunsa­
turated fatty acids. Human milk-fed infants with subnormal
1. Preterm formula weight for their PCA after discharge should receive supplemen­
The high amount of protein, vitamins, minerals, and electro­ tation with HMFs to provide adequate nutrients. If formula-fed
lytes present in preterm formulas are required in order to meet the with suboptimal weight for their PCA, such infants should receive
increased needs associated with rapid growth, decreased intestinal special PDF to decrease the risk of long-term growth failure4).
absorption, and limited fluid tolerance. However, preterm formula Nevertheless, if the infants in Korea are discharged with a sub­
is not generally recommended for postdischarge nutrition and is optimal weight for PCA, preterm formula is used until a weight of
only used until discharge or before switching to PDF. 3.5–4.0 kg is achieved after discharge, enhanced nutrient intake
may be achieved by the replacement of one third, half, or two-
2. Postdischarge formula thirds of daily energy intake with a preterm formula, restricted
PDF is specially designed for preterm infants after hospital fortification with HMFs (half strength), or human milk fortified
discharge. PDF is less nutrient-dense than preterm formula, but with a preterm formula (80, 90, or 100 kcal/100 mL).
provides energy at 75 kcal/100 mL (vs. 68 kcal/dL for term for­
mula), is protein-enriched (2.1 g vs. 1.4–1.5 g/dL), and contains a
variable amount of mineral, vitamins, and trace elements. Al­ Conclusions
though many studies have reported the benefits of PDF, commer­
cial PDF products are currently unavailable in Korea. The use of To reach the goals of nutrition for preterm infants, “aggressive”
PDF has been shown to result in more complete catch-up and and adequate enteral nutrition are needed. In recent years, focus
greater linear growth, weight gain, and bone mineralization than has shifted from the overall energy intake to the optimal protein
term formula18). Moreover, some studies have reported that after intake and P/E ratio. A high overall energy intake may result in
term, PDF cannot change the quantity of growth but rather im­ excess accretion of adipose tissue, whereas high protein intake
proves the quality of growth and lowers the fat mass compared to may have beneficial effects on the quality of growth and long-
term formula19). Generally, PDF use is recommended until 9- term neurodevelopmental outcomes. Minimal enteral nutrition
month PCA2). However, a recent meta-analysis concluded that the should begin as soon as possible during the 1st day of life, and
use of PDF is not supported by the available evidence, because feeding advancement should be conducted based on the clinical
there were no consistent effects on growth at 12–18 months’ course of each infant. During hospitalization, enteral nutrition
corrected age20). Further research on the effects of PDF on later with preterm formula and human milk with HMFs are the best
growth and neurodevelopmental outcomes are needed. feeding practices to improve growth. After discharge, the enteral
nutrition strategy should be individualized according to the
3. Breast feeding weight at discharge and should be reviewed and modified conti­
Most preterm infants are usually discharged by 35- to 36-week nuously to achieve the target growth parameters.
PCA, and/or once a weight of 1,800–2,100 g is achieved. How­
ever, maximal oral feeding performance occurs by 35- to 37-
week PCA. Therefore, preterm infants are unable to manage suffi­ Conflict of interest
cient volumes to meet their nutritional requirements. For this
reason, if the mother plans to breastfeed the infant directly fol­ No potential conflict of interest relevant to this article was
low­ing discharge, the growth rate should be monitored carefully reported.
within 48 hours after discharge and reevaluated after 1 week21).
For those infants who are discharged while still receiving ex­
pressed or donor human milk, fortification is a more practical, References
continuous option22).
The AAP reported that, by the time of discharge, many VLBWI 1. Su BH. Optimizing nutrition in preterm infants. Pediatr Neonatol
2014;55:5-13.
will have achieved growth at an intrauterine rate but remain
2. American Academy of Pediatrics Committee on Nutrition: Nutri­
below the 10th percentile for their PCA. The AAP further stated tional needs of low-birth-weight infants. Pediatrics 1985;75:976-
that more information is needed on what to feed breastfed pre­ 86.
term infants after discharge, and close growth monitoring is 3. American Academy of Pediatrics. Nutritional needs of preterm
infants. Pediatric nutrition handbook. Elk Grove Village (IL):
indicated3). On the other hand, the ESPGHAN recommends that
American Academy of Pediatrics, 2009.
infants with an appropriate weight for their PCA at discharge

https://doi.org/10.3345/kjp.2016.59.12.466 469
Kim MJ • Enteral nutrition

4. Agostoni C, Buonocore G, Carnielli VP, De Curtis M, Darmaun D, 13. ESPGHAN Committee on Nutrition, Arslanoglu S, Corpeleijn W,
Decsi T, et al. Enteral nutrient supply for preterm infants: com­ Moro G, Braegger C, Campoy C, et al. Donor human milk for pre­
mentary from the European Society of Paediatric Gastroen­ term infants: current evidence and research directions. J Pediatr
terology, Hepatology and Nutrition Committee on Nutrition. J Gastroenterol Nutr 2013;57:535-42.
Pediatr Gastroenterol Nutr 2010;50:85-91. 14. Arslanoglu S, Moro GE, Ziegler EE, The Wapm Working Group On
5. Ernst KD, Radmacher PG, Rafail ST, Adamkin DH. Postnatal Nutrition. Optimization of human milk fortification for preterm
malnutrition of extremely low birth-weight infants with catch-up infants: new concepts and recommendations. J Perinat Med 2010;
growth postdischarge. J Perinatol 2003;23:477-82. 38:233-8.
6. Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage LA, Poole 15. Polberger S, Räihä NC, Juvonen P, Moro GE, Minoli I, Warm A. In­
WK. Growth in the neonatal intensive care unit influences neuro­ dividualized protein fortification of human milk for preterm in­
developmental and growth outcomes of extremely low birth fants: comparison of ultrafiltrated human milk protein and a bo­
weight infants. Pediatrics 2006;117:1253-61. vine whey fortifier. J Pediatr Gastroenterol Nutr 1999;29:332-8.
7. Poindexter BB, Ehrenkranz RA. Nutrient requirements and pro­ 16. The Korean Society of Neonatology. Manual of neonatal care. 3rd
vision of nutritional support in the premature neonate. In: Martin ed. Seoul: EUIHAK Publishing Corp., 2014;96-112.
RJ, Fanaroff AA, Walsh MC, editors, Fanaroff and Martin's neo­ 17. Stoll BJ, Hansen NI, Bell EF, Shankaran S, Laptook AR, Walsh MC,
natal-perinatal medicine: diseases of the fetus and infant. 10th ed. et al. Neonatal outcomes of extremely preterm infants from the
Philadelphia (PA): Elsevier Saunders, 2015:592-612. NICHD Neonatal Research Network. Pediatrics 2010;126:443-56.
8. Cho SJ. Enteral nutrition of the premature infant. Korean J Pediatr 18. Griffin IJ, Cooke RJ. Nutrition of preterm infants after hospital
2010;53:7-13. discharge. J Pediatr Gastroenterol Nutr 2007;45 Suppl 3:S195-203.
9. Morgan J, Bombell S, McGuire W. Early trophic feeding versus 19. Amesz EM, Schaafsma A, Cranendonk A, Lafeber HN. Optimal
enteral fasting for very preterm or very low birth weight infants. growth and lower fat mass in preterm infants fed a protein-enri­
Cochrane Database Syst Rev 2013;(3):CD000504. ched postdischarge formula. J Pediatr Gastroenterol Nutr 2010;50:
10. Morgan J, Young L, McGuire W. Delayed introduction of progres­ 200-7.
sive enteral feeds to prevent necrotising enterocolitis in very low 20. Young L, Morgan J, McCormick FM, McGuire W. Nutrient-enriched
birth weight infants. Cochrane Database Syst Rev 2014;(12): formula versus standard term formula for preterm infants follow­
CD001970. ing hospital discharge. Cochrane Database Syst Rev 2012;(3):
11. Morgan J, Young L, McGuire W. Slow advancement of enteral feed CD004696.
volumes to prevent necrotising enterocolitis in very low birth 21. O'Connor DL, Unger S. Post-discharge nutrition of the breastfed
weight infants. Cochrane Database Syst Rev 2014;(12):CD001241. preterm infant. Semin Fetal Neonatal Med 2013 May 21 [Epub].
12. Vohr BR, Poindexter BB, Dusick AM, McKinley LT, Higgins RD, http://doi.org/10.1016/j.siny.2013.05.001.
Langer JC, et al. Persistent beneficial effects of breast milk ingested 22. Picaud JC, Decullier E, Plan O, Pidoux O, Bin-Dorel S, van Egroo
in the neonatal intensive care unit on outcomes of extremely low LD, et al. Growth and bone mineralization in preterm infants fed
birth weight infants at 30 months of age. Pediatrics 2007;120: preterm formula or standard term formula after discharge. J
e953-9. Pediatr 2008;153:616-21, 621.e1-2.

470 https://doi.org/10.3345/kjp.2016.59.12.466
A Novel Multi-Nutrient Human Milk Based Human Milk Fortifier Promotes Growth and Tolerance in
Premature Infants.

Amy Gates PhD, RDN, CSP, LD1, Amy B. Thompson MD1, Terri Marin PhD NNP-BC,
FAAN, CAANP, MBA2, Jennifer L. Waller PhD3, Jenny Patel MD1, Brian K. Stansfield MD1

Affiliations:
1. Department of Pediatrics, Division of Neonatology, Medical College of Georgia, Augusta
University, Augusta, GA, United States
2. College of Nursing, Augusta University, Augusta, GA, United States
3. Department of Population Health Sciences, Division of Biostatistics and Data Science,
Medical College of Georgia, Augusta University, Augusta, GA, United States

Address correspondence to:


Brian K. Stansfield, MD
Department of Pediatrics
Medical College of Georgia at Augusta University
1120 15th Street, BI 6033
Augusta, GA, 30912
bstansfield@augusta.edu
P: 706.721.2331

Funding Source: Medolac Laboratories, A Public Benefit Company (Medolac) provided


Fortify MVP™ (Novel HMF) in kind for the trial.

Role of Funder: Medolac designed and produced the human milk fortifier and provided the
fortifier for the study. Medolac did not participate or assist with study design, study conduct,
data analysis, or study reports.

Financial Disclosures:
Amy Gates
Mead Johnson Nutrition – Medical Science Liaison
Mead Johnson Nutrition – Speaker’s Bureau, Advisory Board, Grant Recipient
Medolac Laboratories – In-Kind Grant Recipient

Terri Marin
Mead Johnson Nutrition – Speaker’s Bureau

Brian Stansfield
Medolac – Medical Consultant for FDA review of clinical data

Jennifer Waller
Medolac – Statistical Consultant for FDA review of clinical data

Amy Thompson and Jenny Patel have nothing to disclose

This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process, which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
10.1111/jpen.2249.

This article is protected by copyright. All rights reserved.


2

Abstract

Background: The objective of this study was to determine whether human milk

supplemented with a novel human milk-based HMF (Novel HMF), compared with a bovine

milk-based HMF (Bovine HMF), supports preterm infant growth through 36 weeks post-

menstrual age (PMA).

Methods: This single center, prospective trial compared growth and nutritional outcomes of

preterm infants provided a human milk-based diet (mother’s own milk or donor milk)

supplemented with a Novel HMF to historic controls provided Bovine HMF. Preterm infants

with an estimated gestational age (EGA) between 23 and 33 weeks PMA and birth weight

between 750 and 1,800 grams provided an exclusive human milk diet were eligible for study

inclusion. Weight, length, head circumference (HC) were monitored weekly. The occurrence

of late-onset sepsis, days of nil per os (NPO), necrotizing enterocolitis (NEC), metabolic

acidosis, and serious adverse events were monitored.

Results: Birth weight, length, HC, and EGA were similar between the Novel HMF (n = 37)

and Bovine HMF (n = 49) groups. The days to regain birth weight was shorter in the Novel

HMF group (9.4  4.0 vs. 11.4  4.8, p=0.0343) with similar weight gain (g/day) from birth

to 36 weeks PMA. Adjusted weight growth velocity (g/kg/day) was significantly higher in the

Novel HMF group at 14 and 21 days, but similar at 36 weeks PMA. The Novel HMF group

experienced fewer NPO days with similar total number of feeding days.

Conclusions: A novel, multi-nutrient human milk-based HMF is well tolerated and meets the

nutritional needs of preterm infants.

This article is protected by copyright. All rights reserved.


3

Clinical Relevancy Statement: Fortification of human milk is necessary to support

physiologic preterm infant growth and human milk-based human milk fortifiers (HMF)

provide an option for a truly exclusive human milk diet. However, the only commercially

available human milk-derived HMF is unsterile and lacks supplemental vitamins, which are

critical for physiologic development. We provide evidence that a sterile, multi-nutrient,

protein enriched human milk-based HMF is well-tolerated and supports physiologic preterm

infant growth.

This article is protected by copyright. All rights reserved.


4

Introduction

A growing body of evidence suggests that human milk for preterm infants is associated with

both short and long-term benefits including lower risk for necrotizing enterocolitis (NEC) and

bronchopulmonary dysplasia (BPD) as well as better cognitive outcomes that may persist into

adolescence1-10. The advantages for mother’s milk for the preterm infant are clear 11
;

however, several obstacles must be overcome to provide sufficient energy and nutrients to

support appropriate linear growth in the immediate post-natal period. The content of human

milk evolves over time and clear differences between preterm and term human milk have
12-14
been described . For example, preterm human milk is protein rich as compared to term

human milk, which influences postnatal growth for preterm infants who receive supplemental

or exclusive donor human milk derived from full term pregnancies. More importantly, the

nutrient requirements of the preterm infant must be supplied in a weight-appropriate volume

of human milk, which is often insufficient to provide adequate concentrations of micro- and

macronutrients and often requires supplementation in the form of human milk fortifiers
15, 16
(HMF) . The addition of HMF to human milk is well-tolerated and improves short term

gains in weight, length, and head circumference and may have benefits for bone

mineralization and neurocognitive outcomes 17.

Until recently, HMFs were bovine-derived, and offering high concentrations of

protein, vitamins, and minerals in an enteral supplement. Sterile infant formulas, calorie and

protein modulars, and HMF are preferred because of the risk of Enterobacter sakazakii

contamination. However, bovine-derived HMFs are associated with lower feeding tolerance
18
and higher risk for metabolic acidosis, when compared to human milk derived HMF. . The

recent development of a human-milk based HMF provided an option for a truly exclusive

This article is protected by copyright. All rights reserved.


5

human milk diet (mother’s own milk or donor milk plus a human milk derived HMF) for
19
preterm infants and may have some clinical benefit in reducing NEC . Although a recent

randomized trial failed to show benefit for human-milk based HMF in the prevention of

NEC, the study did show similar postnatal growth between the two groups, which has been a

concern related to the use of human milk-based HMFs 20, 21. However, the only commercially

available human milk-based HMF lacks important characteristics available in bovine milk-

based HMFs including supplemental vitamins and sterilization. In spite of the limited data on

human milk-based HMFs and their relatively high acquisition costs, approximately 20% of

NICUs in the United States use a human-milk based HMF 22.

Given the potential benefit of sterile human milk-based HMFs and limited availability

of such products to meet current demands, we prospectively assigned preterm infants to a

novel, multi-nutrient human milk-based HMF and compared growth metrics and biochemical

markers of metabolism between the prospective cohort and a historical cohort provided a

Bovine HMF to supplement human milk feedings.

Methods

Study Population

The prospective arm of this study was reviewed and approved by the Augusta University

Internal Review Board (IRB) and registered with ClinicalTrials.gov (NCT03839173). A

parent or guardian was approached for study participation and written informed consent was

obtained prior to study participation. Eligibility criteria for the prospective and historical

cohorts included 1) birth weight between 750 and 1,800 grams, 2) estimated gestational age

(EGA) between 23 and 33 completed weeks, 3) admission to the Children’s Hospital of

Georgia NICU prior to 24 hours of life, 4) commitment to a human milk diet (mother’s own

milk or Medolac Benefit-20™ donor milk, Medolac Laboratories, Boulder City, NV)), and 5)

This article is protected by copyright. All rights reserved.


6

initiation of enteral feedings by day of life 7 (DOL). Eligible subjects from both the

prospective and historical cohorts were excluded for: 1) intrauterine growth restriction

(IUGR) with birth weight less than 3rd centile, 2) known congenital or acquired renal

condition affecting electrolyte metabolism and/or excretion prior to DOL 7, 3) known or

acquired gastrointestinal condition affecting nutrient absorption prior to DOL 7, 4) five

minute APGAR score ≤ 3, 5) Grade III or IV intraventricular hemorrhage (IVH), 6) major

congenital anomalies, or 7) use of bovine-based infant formula or fortifier prior to DOL 7.

Figure 1 describes the number of infants excluded for each exclusion criteria.

Manufacture of the Novel Human Milk Fortifier

During manufacturing, a fractionated, concentrated product is created by separating whole

human donor milk into cream and skim. Skim milk is filtered at refrigeration temperatures

with ultrafiltration (used commonly in the dairy industry for the same purpose), then

blended with human milk cream and whole human milk to meet the macronutrient (fat,

protein, and carbohydrate) specifications of Fortify MVP™. After homogenization, selected

minerals and vitamins are added to the protein and fat enriched milk concentrate to reach

the finished product specification. The vitamins and minerals are blended with a high sheer

mixer until completely incorporated, then packaged in 50mL increments into standard 4oz

spouted retort pouches.

Study Design

This was a single-center, prospective observational cohort study with historical cohort

comparison of preterm infants who received human milk fortified with a human milk-based

HMF (termed Novel HMF) with a historical control of preterm infants who received human

milk fortified with a bovine-based HMF (termed Bovine HMF) from birth through 36 weeks

This article is protected by copyright. All rights reserved.


7

PMA. Participants in the prospective arm received a sterile, multi-nutrient, protein enriched

human milk based HMF (Fortify MVP™ Medolac, Boulder City, Nevada), while infants in

the historical control received a commercially sterile, multi-nutrient, acidified protein

enriched bovine based HMF (Enfamil Human Milk Fortifier, Mead Johnson Nutrition,

Evansville, IN). During both study periods, a standard institution-specific feeding protocol

based on birth weight was used. Feeding protocols were specific for birth weights <750

grams, 750-1000 grams, 1000-1250 grams, 1250-1500 grams, and 1500-1800 grams. Initial

feeding volumes and frequency of volume advancement differed between the feeding

protocols. The complete feeding protocols can be found in Supplemental file XX. Enteral

feeds were initiated prior to DOL 7 in both cohorts and advanced by 10-20 mL/kg/day per the

feeding protocol to an enteral feeding volume goal of 150 mL/kg/day. For both groups, HMF

was added to human milk when infants achieved an enteral feeding volume of 80 mL/kg/day

for at least 24 hours with a fortification target of 24 kcal/ounce assuming a caloric density for

human milk of 20 kcal/ounce. If mother’s own milk was not available Medolac Benefit-20™

donor milk was used (20 calories/ounce and 1.5 grams protein/100 calories). Infants in both

cohorts who experienced extrauterine growth failure, defined as crossing two percentile

curves or deviation away from the 3rd percentile on the gender-specific Fenton Growth

Curve23, could be transitioned to a bovine-based premature infant formula prior to 36 weeks.

A description of the estimated nutrient composition for the Novel and Bovine HMF when

added to human milk (24kcal/ounce) (Supplementary Table I). The reporting of this study

conforms to the STROBE statement.

Outcomes

The primary objective was to determine whether human milk supplemented with a novel

human milk-based fortifier (HMF) with increased protein, as compared to a bovine milk-

This article is protected by copyright. All rights reserved.


8

based HMF, supports optimal preterm infant weight, length, and head circumference growth

through 36 weeks post-menstrual age (PMA). Differences in feeding-relevant outcomes

including feeding tolerance, rates of necrotizing enterocolitis24 (NEC), and biochemical

markers of metabolism were assessed as secondary outcomes.

Anthropometric Measures

Each week from birth to 36 weeks PMA, infants in both cohorts were weighed unclothed and

without a diaper on the same electronically calibrated scale. For the prospective arm, length

was measured weekly by the lead clinical investigator (A.G.) and one clinician using the

Tanis Fenton method with a Premie Stadiometer infant length board. Similarly, head

circumference was measured each week by the lead investigator using the Tanis Fenton

method with a flexible, non-stretchable vinyl tape25 (Seca, Hamburg, Germany). For infants

in the historical cohort, length and head circumference measurements were performed weekly

by the bedside nurse using an infant length board and a flexible, non-stretchable vinyl tape,

respectively. Weight gain was calculated as grams per day (g/day) from birth to 36 weeks

PMA. Weight growth velocity (GV) was calculated from birth to days 14, 21, and 36 weeks

PMA as:

( )
( )
( )
( )

where

= weight at the start of feeds

= weight at day of calculation

= date on day N where N = date corresponding to DOL 14, 21 and 36 weeks

PMA

= date of birth

This article is protected by copyright. All rights reserved.


9

Head circumference and length growth velocities were calculated as centimeters per week

(cm/week) from birth to 36 weeks PMA.

Clinical Outcomes

Metabolic acidosis was defined as serum CO2 value of ≤ 17 mEq/L accompanied by

treatment with a sodium or potassium citrate salt. Suspected and/or confirmed NEC was

determined using Bell’s staging criteria. Late-onset sepsis (LOS) was defined as any positive

blood culture between DOL seven and 36 weeks PMA associated with antimicrobial therapy

for at least 72 hours. All cases of potential LOS, suggested by a positive blood culture, were

reviewed by two clinicians to limit bias.

Sample Size

Based on a sample of recent patients that met inclusion and exclusion criteria, the study was

powered to show a 20% difference in weight growth velocity at 36 weeks PMA between the

Novel and Bovine HMF groups. The sample size was also examined for other variables

including serum magnesium and CO2. Assuming an alpha level of 0.05 and power of 0.80,

and a mixed model for weight velocity, the sample size was determined for interaction

between HMF group and time under a compound symmetric correlation structure with a

correlation between measurement times of 0.5. A sample size of 33 infants per group was

adequate to show differences in weight gain, and serum magnesium and CO2.

Statistical Methods

All statistical analysis was performed using SAS 9.4 and statistical significance was assessed

using an alpha level of 0.05. Descriptive statistics by HMF group (Bovine or Novel) were

determined on all variables and included the frequency and percent for categorical variables,

medians and interquartile ranges for ordinal variables, or the mean and standard deviation for

This article is protected by copyright. All rights reserved.


10

continuous variables. chi-square tests for categorical variables, Kruskal-Wallis tests for

ordinal variables, and two-sample t-tests for continuous variables were used to examine

differences between HMF groups for demographic and baseline measurements, feeding and

clinical outcomes, markers of protein metabolism, electrolytes, and bone health.

Analysis of covariance (ANCOVA) was used to examine differences between HMF

types for return-to-birth-weight day (RTBW), average weight-gain-per-day, head

circumference growth velocity, and length growth velocity at 36-weeks PMA. Each

ANCOVA model controlled for EGA, race, sex, multiple birth, birth weight percentile, birth

head circumference percentile, and birth length percentile. Due to the proximity to birth and

lack of measurements on the day of feeding initiation, birth head circumference and birth

length were controlled for in the average weight gain per day analysis.

For 36-week growth velocity for length and head circumference and for average

weight gain per day, an ANCOVA model was used to examine differences between HMF

groups controlling for EGA and the measure at start of feed. A repeated measures mixed

model was used to examine differences over time (14d, 21d, and 36wk PMA) between HMF

groups for weight growth velocity. The model included main effects and the two-factor

interaction between HMF group and time controlling for EGA and the measure at start of

feed. The subject nested within group was considered a random effect. A Kenward-Rogers

adjustment to the degrees of freedom for testing and assumed an unstructured correlation

structure between measurement times was used. A Bonferroni adjustment to the overall

alpha level (adjusted alpha=0.05/9=0.0056) was used to examine post hoc pairwise

differences between HMF groups within measurement time and between measurement times

within HMF group.

This article is protected by copyright. All rights reserved.


11

Results

Participant characteristics in the Novel and Bovine HMF groups and unadjusted measures are

described in Table 1. No differences in birth weight or gestational age were identified;

however, participants in the Novel HMF group had statistically lower weight- and length-for-

gestational age at birth and at the start of feeds when compared with the Bovine HMF group.

The number of days to return-to-birth-weight (RTBW) was also lower in the Novel HMF

group when compared with the Bovine HMF group. Based on the unadjusted difference

between groups in RTBW days, we examined the difference in RTBW controlling for

demographic and birth characteristics. The Novel HMF group had a lower (8.92  1.09d vs.

10.96  0.93d, p = 0.0601), but not statistically significant, adjusted mean number of days to

return-to-birth-weight when compared to the Bovine HMF group after controlling for

gestational age, race, sex, multiple births, and percentiles for birth weight, length, and head

circumference (Supplementary Table II).

Next, we compared weight gain (grams/day), length growth velocity, and head

circumference growth velocity between the Novel and Bovine HMF. While controlling for

weight, length, and head circumference at the start of feeds, no statistically significant

differences between the groups in weight gain, length growth velocity, or head circumference

growth velocity were identified (Supplementary Table III). Similar results were observed

after controlling for gestational age, race, sex, multiple births, and for weight, length, and

head circumference at birth (Supplementary Table IV).

In order to examine differences between groups in weight growth velocity

(grams/kg/day) over time, we used repeated measures mixed models to adjust for birth weight

in both models, and gestational age, race, sex, multiple births, and birth length and head

circumference in the final model. In both adjusted models, the interaction between HMF type

This article is protected by copyright. All rights reserved.


12

and time was statistically significant indicating that the pattern of mean weight growth

velocity across time was different between the Novel and Bovine HMF groups (Table 2).

Weight growth velocity from 14 days to 21 days was greater in the Bovine HMF group when

compared with the Novel HMF group, but similar between groups from 21 days to 36 weeks.

Post hoc pairwise comparisons between HMF groups indicated that the Novel HMF group

had significantly greater growth velocity at days 14 (p<0.0001) and 21 (p=0.0020) but not at

36 weeks (p=0.3727). Within each HMF group, post hoc pairwise comparisons between

measurement times showed that there were statistically significant differences between

measurement times (14 days vs 21 days p=0.0005 for Novel HMF and p<0.0001 for Bovine

HMF; 14 days vs 36 weeks p<0.0001 for Novel and Bovine HMF; and 21 days vs 36 weeks

p<0.0001 for Novel and Bovine HMF).

Next, we identified and compared feeding relevant outcomes between HMF groups.

The timing of feeding initiation, total HMF days, and total feeding days did not differ

between groups, but HMF was introduced at an earlier DOL in the Novel HMF group when

compared with the Bovine HMF group (Table 3). The Novel HMF group also had

significantly greater total estimated energy intake and significantly less days without any

feedings (i.e. nil per os). Importantly, no differences in necrotizing enterocolitis were

observed between HMF groups, but the percentage of study participants requiring oxygen at

36 weeks PMA was higher in the Novel HMF group when compared with the Bovine HMF

group (Supplementary Table V).

Finally, we measured serum and urine electrolytes, vitamin D concentration, and

hemoglobin and hematocrit at 21 days of life. The Bovine HMF group had significantly

higher blood urea nitrogen (BUN) (16.7  10.1 vs. 11.0  9.7, p = 0.0112) and serum calcium

(10.0  0.4 vs. 9.7  0.5, p = 0.0128) concentrations at DOL 21 when compared with the

This article is protected by copyright. All rights reserved.


13

Novel HMF group, although mean values for BUN and calcium were within traditional

normal ranges (Supplementary Table VI).

Discussion

A substantial body of literature supports the routine use of human milk in the diet of preterm

infants and this practice is emerging as a standard of care after policy statements by the

American Academy of Pediatrics among other professional organizations7, 26, 27. Audacious

goals to provide all preterm infants with an exclusive human milk diet have been proposed

with present recommendations focused on providing human milk to the highest risk preterm

population. Regardless of the source of human milk, fortification of both mother’s own milk

or donor human milk is necessary to support proper extrauterine growth and improved

neurodevelopmental outcomes. The present study provides promising evidence that a novel

human milk-based HMF is well tolerated and supports the nutritional needs of preterm

infants.

Early postnatal weight loss is a physiological response that is exaggerated in preterm

infants and reaches a nadir towards the end of the first week, which is followed by a

downward growth trajectory that continues for at least 2 more weeks28. Thus, a shorter

duration to return to birth weight likely reflects earlier nutritional adequacy that meets the

high metabolic demands of the preterm infant. The mean number of postnatal days required

to regain birth weight was two days lower in the Novel HMF group when compared with the

Bovine HMF group in unadjusted analysis, but failed to reach statistical significance (p=0.06)

when accounting for covariates associated with preterm infant growth and outcomes.

Reflecting an earlier return to birth weight, mean growth velocity (g/kg/day) at postnatal days

14 and 21 was higher in the Novel HMF group when compared with the Bovine HMF group.

This article is protected by copyright. All rights reserved.


14

Relevant to the interpretation of these data, birth weight and weight at the start of feedings

did not differ between groups, but HMF was introduced at an earlier postnatal day in the

Novel HMF group when compared with the Bovine HMF group. Earlier fortification in the

Novel HMF cohort is likely to underlie the enhanced growth velocity at these early time

points. However, it must also be acknowledged that mean total HMF days did not differ

between groups.

Weight gain (g/day), length and head circumference growth velocity at 36 weeks

postmenstrual age (PMA) were similar between groups and were largely influenced by the

particular measurement at the start of feedings and the estimated gestational age. Monitoring

weight gain across time does not clearly represent the patterns of growth for preterm infants

of different gestational ages since weight gain is relatively flat for extremely preterm infants

(i.e. < 28 weeks) on the lower percentile curves while preterm infants on the upper percentile

curves experience positive linear growth beginning soon after birth. This trend is unique to

weight-for-age percentile charts and the dispersion between upper and lower percentiles

continues to diverge through term corrected age. On the other hand, growth velocity

(g/kg/day) provides a more consistent expression of growth over time irrespective of

gestational age 25, 28. Weight growth velocity was nearly two-fold higher throughout the study

period and was consistently higher at 14 days, 21 days, and 36 weeks PMA in the Novel

HMF group when compared with the Bovine HMF group. Taken together, the early return to

birth weight coupled with the higher growth velocity suggests that human milk fortified with

the Novel HMF meets the energy needs of preterm infants.

In order to more accurately assess growth characteristics in the Novel HMF group, we

compared feeding-relevant outcomes between cohorts. Several key feeding outcomes were

similar between groups including DOL for start of feedings, total feeding days, and days of

This article is protected by copyright. All rights reserved.


15

parenteral nutrition. Together, these data suggest that early feeding patterns were largely

similar between the study periods with the exception of earlier introduction of fortifier in the

Novel HMF group. While it is certainly reasonable to conclude that earlier fortification

enhances growth velocity, particularly in the first weeks of life, the total number of HMF

days did not differ between groups. Further, the differences in weight growth velocity

persisted to 36 weeks PMA suggesting that the Novel HMF group continued to demonstrate

an advantage in growth velocity through term corrected age. Whether the early growth

advantages demonstrated in the Novel HMF group reflects earlier fortification and place

these groups on different growth trajectories and are maintained throughout the first months

of life is an important and clinically relevant question that remains.

Mean total feeding days with an exclusive human milk-based diet was marginally, but

not statistically higher (p=0.0759) in the Bovine HMF group when compared with the Novel

HMF group. Historically, a concern for preterm infants provided an exclusive human milk

diet was the poor growth observed in these infants when compared with preterm infants

provided a bovine-based preterm formula. This relationship appears to be particularly true

with donor human milk. Donor human milk appears to exert a linear and negative effect on

weight and head circumference growth velocity at 36 weeks PMA in preterm infants in
29
comparison to mother’s own milk and preterm formula . Others have shown similar

impaired growth in preterm infants provided a high percentage of donor human milk and has

been attributed to reduced fat, protein, and energy in donor human milk as compared to
30-32 33
mother’s own milk , but this relationship is not universally observed . In our study

population, differences in total volume or percent of mother’s own milk did not differ

between HMF groups. The emergence of fortifiers and improved growth tracking has largely

removed the barriers to growth that were previously observed and impeded the widespread
25
adoption of exclusive human milk diets . Fortification of human milk in particular has

This article is protected by copyright. All rights reserved.


16

greatly improved anthropometric outcomes in preterm infants provided a human milk diet 34.

In our own study, both fortification strategies achieved a minimum target > 10 g/kg/day for

weight growth velocity with higher, but not statistically significant, growth velocity observed

in the Novel HMF group when compared with the Bovine HMF group at 36 weeks PMA.

This growth advantage was likely achieved through greater total estimated energy intake in

the Novel HMF group while also maintaining similar protein delivery when compared with

the Bovine HMF group. The higher energy intake and growth velocity in the Novel HMF

cohort is an important finding that opposes a recent systematic review and meta-analysis

demonstrating improved growth in preterm infants provided different Bovine HMFs when

compared with a human milk-derived HMF35. Direct comparisons between bovine and

human milk-derived HMFs are limited and our findings directly contribute to this growing
36, 37
body of evidence . The improved growth velocity may also reflect the lower number of

NPO days in the Novel HMF group. Total days of parenteral nutrition did not differ between

groups suggesting that prolonged NPO was uncommon in both groups, but the lower mean

number of NPO days may reflect better feeding tolerance in the Novel HMF group.

The frequency of clinically important outcomes including death, medical or surgical

NEC, late onset sepsis, and retinopathy of prematurity (ROP) did not differ between groups.

ROP is often identified in preterm infants who experience extrauterine growth failure and

improving growth with a variety of dietary interventions is efficacious in reducing ROP38-42.

Similarly, BPD is a multifactorial outcome that is closely tied to postnatal growth. Poor

postnatal growth due to higher oxygen consumption or demand or inadequate nutrient supply
43, 44
are linked to higher rates of BPD . We did observe a statistically significant increase in

infants requiring oxygen at 36 weeks PMA, the clinical definition of BPD, in the Novel HMF

group when compared with the Bovine HMF group. As both groups experienced acceptable

growth velocity with the Novel HMF group achieving an even higher growth velocity than

This article is protected by copyright. All rights reserved.


17

the Bovine HMF group, it is unlikely that nutritional inadequacy or insufficient energy supply

contributed to the observed difference in outcome. Rather, the Novel HMF group was

statistically smaller-for-gestational age and includes more infants who experienced slow

intrauterine growth. Further, it is plausible that an earlier return to birth weight reflects a

shorter diuretic phase and potentially greater fluid retention in the immediate post-partum

period. Both, small-for-gestational age and higher fluid intake/retention in the first week of

life are important risk factor for BPD 45-49. While these potential explanations are reasonable,

it must be noted that the mean percent weight loss between birth and start of feeds was

similar between the cohorts (5.9%  6.7 vs. 6.1%  6.1, p=0.93), and the Novel HMF group

continued to be smaller-for-age at feeding initiation when compared with the Bovine HMF

group. A limitation to our study was that the weight, length and head circumference

measurements for the retrospective cohort group were not taken by one trained individual but

were collected from the electronic medical record. For the prospective group, weight, length

and head circumference measurements were taken by the principal investigator plus one

trained neonatal nurse practitioner.

Finally, we examined serum electrolyte concentrations along with measures of bone

mineralization and blood volume at the end of the first month of life in both cohorts. Both

groups displayed similar values within the expected normal range for all electrolytes with a

statistically higher serum calcium concentration observed in the Bovine HMF group when

compared with the Novel HMF group. Serum alkaline phosphatase and phosphorus were

within normal limits for both groups. Vitamin D status and hemoglobin values were similar

between groups.

Several limitations for this study are acknowledged. First, the use of a historical

control to compare nutritional outcomes and growth does not provide a true comparison

This article is protected by copyright. All rights reserved.


18

between the novel and Bovine HMF. Our goal in this cohort study was to identify whether

appropriate growth outcomes for preterm infants could be achieved with the Novel HMF;

thus, we feel that a historical control provided a sufficient comparison group and recognize

that superiority of the Novel HMF is not truly evaluated. Relevant to this point, feeding

protocols were unchanged during both study periods and the prospective and historical

cohorts did not differ for important demographics such as EGA and birth weight or total

feeding days and exposure to mother’s own milk. We also recognize that the lack of

randomization and blinding in the prospective cohort does introduce potential bias. All

measurements were made by a single member of the investigative team during both cohorts

(AG) and outcomes in the prospective group were confirmed by the investigative team (AG

and BKS) as well as a member of the healthcare team that is not involved in the study.

Finally, we acknowledge that limited data for some measurements in the historical control

precludes true comparisons between the groups.

The present study provides evidence that a novel human milk-based HMF is well-

tolerated and supports the nutritional needs of preterm infants. When added to human milk,

maternal or donor, the novel human milk-based HMF meets the recommendations for protein

and micro-nutrients. Furthermore, the novel fortifier provides healthcare providers with a

potential alternative to bovine based HMFs and supports a complete human milk diet for

premature infants.

Statement of Authorship

This article is protected by copyright. All rights reserved.


19

A. Gates and B.K. Stansfield equally contributed to the conception and design of the

research; A. Gates, A. B. Thompson, and J. Patel contributed to the acquisition of the data; J.

L. Waller contributed to the analysis of the data; A. Gates, A. B. Thompson, T. Marrin, J.L.

Waller, and B.K. Stansfield contributed to the interpretation of the data; and A. Gates and

B.K. Stansfield drafted the manuscript. All authors critically revised the manuscript, agree to

be fully accountable for ensuring the integrity and accuracy of the work, and read and

approved the final manuscript.

Supplementary Material

Supplementary table 1-6 are online at

This article is protected by copyright. All rights reserved.


20

References

1. Sullivan. An Exlusively Human Milk-Based Diet is Associated with a Lower Rate of Necrotizing
Enterocolitis than a Diet of Human Milk and Bovine Milk-Based Products. The Journal of
Pediatrics. 2010:562-567.
2. Cristofalo EA, Schanler, Richard, Blanco, Cynthia, Sullivan, Sandra, et. al. Randomized Trial of
Exclusive Human Milk versus Preterm Formula Diets in Extremely Premature Infants. The
Journal of Pediatrics. 2013:1592-1595.
3. Ghandehari Heli LML, Rechtman David. An exclusive human milk-based diet in extremely
premature infants reduces the probability of remaining on total parenteral nutrition: a
reanalysis of the data. BMC Research Notes. 2012:1-5.
4. Stoll Barbara HN, Bell Edward, Shankaran Seetha, Laptook Abbot, Walsh Michele, et. al.
Neonatal Outcomes of Extremely Preterm Infants from the NICHD Neonatal Research
Network. Pediatrics. 2010;3(126):443-456.
5. Madore LS, Bora S, Erdei C, Jumani T, Dengos AR, Sen S. Effects of Donor Breastmilk Feeding
on Growth and Early Neurodevelopmental Outcomes in Preterm Infants: An Observational
Study. Clinical Therapies. 2017:1210-1120.
6. Feldman R EA. Direct and indirect effects of breast milk on the neurobehavioral and
cognitive development of premature infants. Developmental Psychobiology.
2003:43(42):109-119.
7. Quigley M, Embleton ND, McGuire W. Formula versus donor breast milk for feeding preterm
or low birth weight infants. Cochrane Database Syst Rev. Jul 19 2019;7:CD002971.
8. Meinzen-Derr J, Poindexter B, Wrage L, Morrow AL, Stoll B, Donovan EF. Role of human milk
in extremely low birth weight infants' risk of necrotizing enterocolitis or death. J Perinatol.
Jan 2009;29(1):57-62.
9. Ghandehari H, Lee ML, Rechtman DJ. An exclusive human milk-based diet in extremely
premature infants reduces the probability of remaining on total parenteral nutrition: a
reanalysis of the data. BMC Res Notes. Apr 25 2012;5:188.
10. Victoria CG BR, Barros AJD, Franca GVA, Horton S, Krasevec J. Breastfeeding in the 21st
century: epidemiology, mechanisms, and lifelong effect. The Lancet. January 30
2016;387(10017):475-490.
11. Breastfeeding and the Use of Human Milk. Pediatrics. 2012;123(3):e827-e841.
12. Gates A, Marin T, Leo G, Stansfield BK. Review of Preterm Human-Milk Nutrient
Composition. Nutr Clin Pract. Aug 30 2020.
13. Bhatia J. Human milk and the premature infant. Ann Nutr Metab. 2013;62 Suppl 3:8-14.
14. Underwood MA. Human milk for the premature infant. Pediatr Clin North Am. Feb
2013;60(1):189-207.
15. Koletzko B PB, Uauy R. Recommended Nutrient Intake Levels for Stable, Fully Enterally Fed
Very Low Birth Weight Infants. Nutritional Care of Preterm Infants. Scientific Basis and
Practical Guidelines. . Basel, Switzerland: Karger; 2010:297-299.
16. Casavale KO, Ahuja JKC, Wu X, et al. NIH workshop on human milk composition: summary
and visions. Am J Clin Nutr. Sep 1 2019;110(3):769-779.
17. Arslanoglu S, Boquien CY, King C, et al. Fortification of Human Milk for Preterm Infants:
Update and Recommendations of the European Milk Bank Association (EMBA) Working
Group on Human Milk Fortification. Front Pediatr. 2019;7:76.
18. Thoene M, Hanson C, Lyden E, Dugick L, Ruybal L, Anderson-Berry A. Comparison of the
effect of two human milk fortifiers on clinical outcomes in premature infants. Nutrients. Jan
3 2014;6(1):261-275.

This article is protected by copyright. All rights reserved.


21

19. Sullivan S, Schanler RJ, Kim JH, et al. An exclusively human milk-based diet is associated with
a lower rate of necrotizing enterocolitis than a diet of human milk and bovine milk-based
products. J Pediatr. Apr 2010;156(4):562-567 e561.
20. O'Connor DL, Kiss A, Tomlinson C, et al. Nutrient enrichment of human milk with human and
bovine milk-based fortifiers for infants born weighing <1250 g: a randomized clinical trial.
Am J Clin Nutr. Jul 1 2018;108(1):108-116.
21. Eibensteiner F, Auer-Hackenberg L, Jilma B, Thanhaeuser M, Wald M, Haiden N. Growth,
Feeding Tolerance and Metabolism in Extreme Preterm Infants under an Exclusive Human
Milk Diet. Nutrients. Jun 26 2019;11(7).
22. Perrin MT. Donor Human Milk and Fortifier Use in United States Level 2, 3, and 4 Neonatal
Care Hospitals. J Pediatr Gastroenterol Nutr. Apr 2018;66(4):664-669.
23. Fenton TR, Nasser R, Eliasziw M, Kim JH, Bilan D, Sauve R. Validating the weight gain of
preterm infants between the reference growth curve of the fetus and the term infant. BMC
Pediatr. Jun 11 2013;13:92.
24. Juhl SM, Hansen ML, Gormsen M, Skov T, Greisen G. Staging of necrotising enterocolitis by
Bell's criteria is supported by a statistical pattern analysis of clinical and radiological
variables. Acta Paediatr. May 2019;108(5):842-848.
25. Fenton TR, Chan HT, Madhu A, et al. Preterm Infant Growth Velocity Calculations: A
Systematic Review. Pediatrics. Mar 2017;139(3).
26. Committee On N, Section On B, Committee On F, Newborn. Donor Human Milk for the High-
Risk Infant: Preparation, Safety, and Usage Options in the United States. Pediatrics. Jan
2017;139(1).
27. Section on B. Breastfeeding and the use of human milk. Pediatrics. Mar 2012;129(3):e827-
841.
28. Cole TJ, Statnikov Y, Santhakumaran S, et al. Birth weight and longitudinal growth in infants
born below 32 weeks' gestation: a UK population study. Arch Dis Child Fetal Neonatal Ed. Jan
2014;99(1):F34-40.
29. Brownell EA, Matson AP, Smith KC, et al. Dose-response Relationship Between Donor Human
Milk, Mother's Own Milk, Preterm Formula, and Neonatal Growth Outcomes. J Pediatr
Gastroenterol Nutr. Jul 2018;67(1):90-96.
30. Soldateli B, Parker M, Melvin P, Gupta M, Belfort M. Human milk feeding and physical
growth in very low-birth-weight infants: a multicenter study. J Perinatol. Aug
2020;40(8):1246-1252.
31. Castellano Yanez C, Castillo Barrio B, Munoz Labian MDC, et al. Providing very preterm
infants with donor human milk led to faster breastfeeding rates but worse biometric gains.
Acta Paediatr. Apr 2019;108(4):766-767.
32. Perrin MT, Belfort MB, Hagadorn JI, et al. The Nutritional Composition and Energy Content of
Donor Human Milk: A Systematic Review. Adv Nutr. Jul 1 2020;11(4):960-970.
33. Sisk PM, Lambeth TM, Rojas MA, et al. Necrotizing Enterocolitis and Growth in Preterm
Infants Fed Predominantly Maternal Milk, Pasteurized Donor Milk, or Preterm Formula: A
Retrospective Study. Am J Perinatol. Jun 2017;34(7):676-683.
34. Kuschel CA, Harding JE. Multicomponent fortified human milk for promoting growth in
preterm infants. Cochrane Database Syst Rev. 2004(1):CD000343.
35. Ananthan A, Balasubramanian H, Rao S, Patole S. Human Milk-Derived Fortifiers Compared
with Bovine Milk-Derived Fortifiers in Preterm Infants: A Systematic Review and Meta-
Analysis. Adv Nutr. Sep 1 2020;11(5):1325-1333.
36. Ananthan A, Balasubramanian H, Rao S, Patole S. Response to comments by Prof Abrams
and Prof Lucas on "Human Milk-Derived Fortifiers Compared with Bovine Milk-Derived
Fortifiers in Preterm Infants: A Systematic Review and Meta-Analysis". Adv Nutr. Nov 16
2020;11(6):1713-1715.

This article is protected by copyright. All rights reserved.


22

37. Lucas A, Abrams SA. Comment on "Human Milk-Derived Fortifiers Compared with Bovine
Milk-Derived Fortifiers in Preterm Infants: A Systematic Review and Meta-Analysis". Adv
Nutr. Nov 16 2020;11(6):1712-1713.
38. Aydemir O, Sarikabadayi YU, Aydemir C, et al. Adjusted poor weight gain for birth weight and
gestational age as a predictor of severe ROP in VLBW infants. Eye (Lond). Jun
2011;25(6):725-729.
39. Binenbaum G, Ying GS, Quinn GE, et al. The CHOP postnatal weight gain, birth weight, and
gestational age retinopathy of prematurity risk model. Arch Ophthalmol. Dec
2012;130(12):1560-1565.
40. Porcelli PJ, Weaver RG, Jr. The influence of early postnatal nutrition on retinopathy of
prematurity in extremely low birth weight infants. Early Hum Dev. Jun 2010;86(6):391-396.
41. VanderVeen DK, Martin CR, Mehendale R, et al. Early nutrition and weight gain in preterm
newborns and the risk of retinopathy of prematurity. PLoS One. 2013;8(5):e64325.
42. Wallace DK, Kylstra JA, Phillips SJ, Hall JG. Poor postnatal weight gain: a risk factor for severe
retinopathy of prematurity. J AAPOS. Dec 2000;4(6):343-347.
43. Biniwale MA, Ehrenkranz RA. The role of nutrition in the prevention and management of
bronchopulmonary dysplasia. Semin Perinatol. Aug 2006;30(4):200-208.
44. Wemhoner A, Ortner D, Tschirch E, Strasak A, Rudiger M. Nutrition of preterm infants in
relation to bronchopulmonary dysplasia. BMC Pulm Med. Feb 3 2011;11:7.
45. Rocha G, de Lima FF, Machado AP, et al. Small for gestational age very preterm infants
present a higher risk of developing bronchopulmonary dysplasia. J Neonatal Perinatal Med.
2019;12(4):419-427.
46. Reiss I, Landmann E, Heckmann M, Misselwitz B, Gortner L. Increased risk of
bronchopulmonary dysplasia and increased mortality in very preterm infants being small for
gestational age. Arch Gynecol Obstet. Nov 2003;269(1):40-44.
47. Kewitz G, Wudel S, Hopp H, Hopfenmuller W, Vogel M, Roots I. Below median birth weight in
appropriate-for-gestational-age preterm infants as a risk factor for bronchopulmonary
dysplasia. J Perinat Med. 2008;36(4):359-364.
48. Havinga J, Williams A, Hassan N, et al. Individualized fluid management in extremely preterm
neonates to ensure adequate diuresis without increasing complications. J Perinatol. Feb
2021;41(2):240-246.
49. Oh W, Poindexter BB, Perritt R, et al. Association between fluid intake and weight loss during
the first ten days of life and risk of bronchopulmonary dysplasia in extremely low birth
weight infants. J Pediatr. Dec 2005;147(6):786-790.

This article is protected by copyright. All rights reserved.


23

Table 1: Descriptive statistics by HMF type and test for differences for demographic and other
preterm infant characteristics

Test p-
Variable Bovine HMF Novel HMF
Statistic value

Birthweight†, g 1130.9 (186.7) n=49 1146.0 (238.6) n=37 0.33 0.7435

z-score† 0.4 (0.7) n=49 -0.1 (0.8) n=37 -3.00 0.0035

Percentile†, % 63.4 (23.1) n=49 46.6 (23.3) n=37 -3.33 0.0013

Birth Class ƒ 0.20 0.6576

≤1000g 15 (30.6) 13 (35.1)

>1000g 34 (69.4) 24 (64.9)

Feeding Protocol Birthweight Ranges ƒ * 0.2390

750-999g 15 (30.6) 12 (32.4)

1000-1499g 33 (67.4) 21 (56.8)

1500g 1 (2.0) 4 (10.8)

RTBW†, d 11.4 (4.8) n=49 9.4 (4.0) n=37 -2.15 0.0343

Birth Length†, cm 36.4 (2.6) n=49 36.8 (3.0) n=37 0.54 0.5895

z-score† 0.2 (0.7) n=49 -0.2 (1.0) n=37 -1.65 0.1047

Percentile†, % 56.6 (22.2) n=49 44.9 (28.1) n=37 -2.14 0.0351

Birth Head Circumference†, cm 25.2 (1.8) n=49 25.8 (1.8) n=37 1.54 0.1263

z-score† -0.1 (0.9) n=49 -0.3 (0.9) n=37 -0.86 0.3947

Percentile†, % 47.7 (27.3) n=49 40.9 (26.9) n=37 -1.15 0.2536

Estimated Gestational Age†, weeks 27.9 (1.9) n=49 28.8 (2.2) n=37 1.91 0.0601

Apgar score at 5 minutes‡ 6.0 (5.0-8.0) n=49 6.0 (5.0-7.0) n=37 4.17 0.0411

Gender ƒ 0.23 0.6343

Female 20 (40.8) 17 (46.0)

Male 29 (59.2) 20 (54.0)

Race ƒ * 0.1426

This article is protected by copyright. All rights reserved.


24

White 14 (28.6) 8 (21.6)

Black 31 (63.3) 29 (78.4)

Hispanic 4 (8.1) 0 (0.0)

Multiples ƒ 3.04 0.0811

Yes 6 (12.2) 10 (27.0)

No 43 (87.8) 27 (73.0)

Start of Feeds

Weight at start of feeds†, g 1064.9 (183.7) n=49 1073.5 (219.6) n=37 0.20 0.8440

Percentile†, % 45.6 (22.9) n=49 31.8 (20.8) n=37 -2.89 0.0050

Length at start of feeds†, g 36.4 (2.6) n=49 36.9 (3.0) n=37 0.68 0.4983

Percentile†, % 56.6 (22.2) n=49 45.0 (28.0) n=37 -2.14 0.0353

36-week PMA Measurements

Weight†, g 2226.7 (314.9) n=46 2142.3 (297.5) n=35 -1.22 0.2245

Percentile†, % 22.0 (20.6) n=45 16.4 (18.2) n=35 -1.26 0.2122

Weight gain†, g/day 20.0 (5.1) n=45 20.3 (4.9) n=35 0.30 0.7673

Weight Growth Velocity†, g/kg/day 11.8 (2.0) n=45 12.3 (2.4) n=35

Length†, cm 44.5 (2.0) n=39 43.7 (2) n=35 -1.59 0.1160

Percentile†, % 21.3 (21.4) n=39 17.0 (17.3) n=35 -0.94 0.3486

Growth Velocity†, cm/week 1.0 (0.3) n=39 1.0 (0.2) n=35

Head Circumference†, cm 30.7 (1.3) n=47 30.9 (1.3) n=35 0.51 0.6142

Percentile†, % 19.6 (18.6) n=47 20.2 (18.9) n=35 0.15 0.8802

Growth Velocity†, cm/week 0.7 (0.2) n=47 0.7 (0.1) n=35



mean (SD) n, t-statistic presented

median (IQR) n , Kruskal-Wallis chi-square statistic presented
ƒ
n (%), chi-square statistics presented
* Fisher’s Exact test performed
HMF=Human Milk Fortifier

This article is protected by copyright. All rights reserved.


25

Table 2: Mixed model results for difference in weight growth velocity over time between
HMF types, simple and final adjusted model

Adjusted LS
Effect in Model F p-value
Mean (SE)

Simple Model

HMF Type 17.10 <0.0001

Bovine-based HMF 6.05 (0.45)

Novel 8.89 (0.52)

Time Point 204.75 <0.0001

14d 3.43 (0.53)

21d 6.97 (0.40)

36wk 12.02 (0.24)

HMF Type x Time Point 17.28 <0.0001

Bovine-based HMF

14d (N=48) 0.66 (0.070)

21d (N=46) 5.54 (0.53)

36wk (N=47) 11.84 (0.32)

Novel

14d (n=36) 6.20 (0.79)

21d (N=36) 8.28 (0.60)

36wk (N=35) 12.20 (0.36)

Birth Weight, g 0.04 0.8346

Final Adjusted Model

HMF Type 15.80 <0.0001

Bovine-based HMF 6.14 (0.59)

Novel 9.08 (0.67)

Time Point 202.65 <0.0001

14d 3.56 (0.65)

This article is protected by copyright. All rights reserved.


26

21d 7.10 (0.55)

36wk 12.16 (0.45)

HMF Type x Time Point 16.79 <0.0001

Bovine-based HMF

14d (N=48) 0.75 (0.079)

21d (N=46) 5.76 (0.65)

36wk (N=47) 11.91 (0.49)

Novel

14d (n=36) 6.37 (0.91)

21d (N=36) 8.45 (0.74)

36wk (N=35) 12.41 (0.56)

Birth Weight, g 0.02 0.9002

EGA, weeks 0.17 0.6846

Race 1.22 0.3000

Sex 1.30 0.2576

Multiple Birth 1.16 0.2841

Birth Head Circumference, cm 1.64 0.2036

Birth Length, cm 0.72 0.3985

HMF=Human Milk Fortifier, EGA=Estimated Gestational Age, LS=Least Squares, SE=Standard


Error

This article is protected by copyright. All rights reserved.


27

Table 3: Descriptive statistics [mean (SD) n] by HMF type and test for differences for feeding outcomes

t-
Variable Bovine HMF Novel HMF p-value
statistic

Days of Parenteral Feeding, d 17.4 (8.4) n=49 16.2 (7.8) n=37 -0.69 0.4931

DOL Feeding Started, d 2.4 (1.5) n=49 2.1 (1.2) n=37 -1.04 0.3001

Total Feeding Days, d 50.4 (13.8) n=49 45.4 (14.8) n=37 -1.60 0.1129

DOL HMF Started, d 16.4 (8.1) n=49 12.0 (5.3) n=36 -3.03 0.0032

Total HMF Days, d 28.7 (12.7) n=49 30.5 (11.3) n=37 0.70 0.4875

DOL Full Feeds Reached, d 20.5 (8.6) n=48 18.9 (9.8) n=36 -0.77 0.4430

Total Volume MOM, mL/kg 3355.0 (4265.3) n=49 2880.6 (3627.7) n=37 -0.54 0.5880

Total Volume Donor Milk, mL/kg 2954.4 (3173.9) n=49 3982.8 (3325.3) n=37 1.46 0.1487

Percent of Volume from MOM, % 47.4 (49.6) n=49 42.7 (42.6) n=37 -0.47 0.6408

Total estimated energy Intake,


82.2 (17.3) n=45 95.2 (17.0) n=35 3.36 0.0012
kcal/kg/day

Days NPO, d 2.1 (3.3) n=49 0.7 (1.8) n=37 -2.45 0.0166

Total Protein Intake, g/kg/day 3.1 (0.8) n=45 3.0 (0.6) n=35 -1.16 0.2480

DOL=Day of Life, HMF=Human Milk Fortifier, MOM=Mother’s Own Milk, kcal=kilocalories,


NPO=Nothing per OS

This article is protected by copyright. All rights reserved.


28

This article is protected by copyright. All rights reserved.


Review
published: 26 May 2017
doi: 10.3389/fnut.2017.00020

Optimizing Nutrition in Preterm Low


Birth weight infants—Consensus
Summary
R. Kishore Kumar1, Atul Singhal 2, Umesh Vaidya 3, Saswata Banerjee 4, Fahmina Anwar 5*
and Shashidhar Rao 5
1
 Cloudnine Hospital, Bangalore, Karnataka, India, 2 Institute of Child Health, UCL, London, United Kingdom, 3 KEM Hospital,
Pune, India, 4 Nestle Nutrition, Kolkata, India, 5 Medical and Scientific Affairs, Nestle Nutrition, South Asia Region, Gurgaon, India

Preterm birth survivors are at a higher risk of growth and developmental disabilities
compared to their term counterparts. Development of strategies to lower the com-
plications of preterm birth forms the rising need of the hour. Appropriate nutrition is
essential for the growth and development of preterm infants. Early administration of
optimal nutrition to preterm birth survivors lowers the risk of adverse health outcomes
and improves cognition in adulthood. A group of neonatologists, pediatricians, and
nutrition experts convened to discuss and frame evidence-based recommendations for
optimizing nutrition in preterm low birth weight (LBW) infants. The following were the
Edited by:
Donato F. Romagnolo,
primary recommendations of the panel: (1) enteral feeding is safe and may be preferred
University of Arizona, United States to parenteral nutrition due to the complications associated with the latter; however,
Reviewed by: parenteral nutrition may be a useful adjunct to enteral feeding in some critical cases;
Pedro Magalhães,
(2) early, fast, or continuous enteral feeding yields better outcomes compared to late,
Agostinho Neto University, Angola
Sharon Ross, slow, or intermittent feeding, respectively; (3) routine use of nasogastric tubes is not
National Cancer Institute (NIH), advisable; (4) preterm infants can be fed while on ventilator or continuous positive air-
United States
way pressure; (5) routine evaluation of gastric residuals and abdominal girth should be
*Correspondence:
Fahmina Anwar
avoided; (6) expressed breast milk (EBM) is the first choice for feeding preterm infants
fahmina.anwar@in.nestle.com due to its beneficial effects on cardiovascular, neurological, bone health, and growth
outcomes; the second choice is donor pasteurized human milk; (7) EBM or donor milk
Specialty section:
may be fortified with human milk fortifiers, without increasing the osmolality of the milk, to
This article was submitted
to Clinical Nutrition, meet the high protein requirements of preterm infants; (8) standard fortification is effective
a section of the journal and safe but does not fulfill the high protein needs; (9) use of targeted and adjustable
Frontiers in Nutrition
fortification, where possible, helps provide optimal nutrition; (10) optimizing weight gain
Received: 09 January 2017
Accepted: 29 April 2017
in preterm infants prevents long-term cardiovascular complications; (11) checking for
Published: 26 May 2017 optimal weight and sucking/swallowing ability is essential prior to discharge of preterm
Citation: infants; and (12) appropriate counseling and regular follow-up and monitoring after
Kumar RK, Singhal A, Vaidya U, discharge will help achieve better long-term health outcomes. This consensus summary
Banerjee S, Anwar F and Rao S
(2017) Optimizing Nutrition in Preterm serves as a useful guide to clinicians in addressing the challenges and providing optimal
Low Birth Weight Infants— nutrition to preterm LBW infants.
Consensus Summary.
Front. Nutr. 4:20. Keywords: optimizing nutrition, preterm low birth weight infants, enteral feeding, expressed breast milk, donor
doi: 10.3389/fnut.2017.00020 pasteurized human milk, fortification

Frontiers in Nutrition  |  www.frontiersin.org 1 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

INTRODUCTION the consensus meeting. This was followed by the selection of


a group of neonatologists, pediatricians, and nutrition experts,
About 15 million preterm births are recorded each year world- who convened in August 2016 to brainstorm and address the
wide. According to the World Health Organization, there has various challenges in providing optimal nutrition to preterm
been an increase in the incidence of preterm births over the past LBW infants. The panel discussion also focused at highlight-
20 years in 62 of the 65 countries with available trend data. Over ing the advantages and disadvantages, optimal intakes, and
one million children die from preterm birth-related complica- practical recommendations for various enteral nutrition sup-
tions annually. Preterm births are the leading cause of newborn plementation strategies.
deaths and the second leading cause of death, after pneumonia, This consensus summary paper compiles the evidence-based
in children under the age of 5 years. Furthermore, survivors of recommendations from the Expert panel and serves as a useful
preterm birth are at a higher risk of adverse developmental dis- tool to the clinicians in optimizing nutrition in LBW preterm
abilities (1). They have higher rates of adverse health outcomes in infants for beneficial health outcomes in the long term.
early adulthood compared with their term counterparts (2). The
various adverse developmental outcomes in adults born preterm
have been outlined in Table 1 (3–9). EFFECTS OF EARLY AND AGGRESSIVE
Although there has been an improvement in the overall mor- NUTRITIONAL STRATEGIES
tality in extremely premature infants in recent times, there is a
rising need to develop newer strategies for lowering the potential It has been noted that better nutrition in the early postnatal phases
complications of preterm birth (10). in preterm infants results in higher verbal intelligence quotient (IQ)
scores and improved cognitive function in the long term (18, 19).
Nutrition—An Important Factor Influencing Higher protein and energy intake during the first week after birth in
extremely LBW infants is associated with higher mental development
Developmental Outcomes in Preterm index scores and lower risk of growth retardation at 18 months after
Infants birth (20). Early and higher protein and energy intake have also been
Nutrition is essential for growth, metabolism, and immunity in correlated with faster head growth and an increase in head circumfer-
a preterm newborn low birth weight (LBW) infant (11–13). In ence in preterm infants (21, 22); increase in head circumference has
a preterm infant, poor nutrition is associated with poorer head been positively correlated with improved cognitive outcomes (23).
growth; persistent smaller head size results in poor psychomotor Therefore, the administration of early aggressive nutritional enteral
and mental skills, higher rates of cerebral palsy, and autism (14). and parenteral support may help improve growth and developmental
Impaired weight and growth in preterm infants are significantly outcomes in preterm newborn LBW infants (24).
associated with adverse neurodevelopmental outcomes in later
life (15). Barker’s hypothesis also states that infants with LBW are
at a higher risk of coronary heart disease, hypertension, and type CHALLENGES IN PROVIDING NUTRITION
2 diabetes in adulthood (16, 17). TO PRETERM INFANTS

OBJECTIVES AND METHODS Parenteral versus Enteral Feeding


Enteral feeding is preferred to parenteral feeding, as the latter may
Considering the significant contribution of nutrition to the be associated with catheter-related complications, infections,
development of preterm newborn LBW infants, a key opinion and sepsis, among others (25, 26). However, institution of early
leader who has extensive expertise in the related domain parenteral nutrition can sometimes be critical and a necessary
reached out to other experts who had proficiency in various adjunct to enteral therapy.
areas related to the topic/s proposed to be discussed during
Does Enteral Feeding Carry an Increased
Table 1 | Adverse developmental sequelae in adults born preterm. Risk of Necrotizing Enterocolitis (NEC)
Parameter Adverse sequelae in adults born preterm and Infections?
One of the most feared complications of enteral feeding of a pre-
Neurological • Significant decrease in brain volume (3). term infant is the risk of NEC; this came into limelight based on a
• Increased risk of neurological disabilities (4).
study published in The Lancet in 1990. In this study, NEC was more
Cardiovascular and metabolic • Low insulin sensitivity and high blood
common in preterm infants fed formula milk compared to infants
pressure (5).
• Increased intra-abdominal fat and higher risk fed breast milk (27). However, there have been improvements in
of metabolic complications (6). feeding regimens and methods since this publication. In studies
• Increased arterial stiffness (7). published after this paper, there has been no increase in the risk
• Reduced ventricular size and volume; of NEC with fast or early enteral feeding of expressed breast milk
impaired systolic function (8).
(EBM) or formula milk as compared to slow or delayed introduc-
Bone health • Significantly lower bone mineral density (9).
Others • Increased risk of social disabilities in adulthood
tion of enteral feeding in LBW infants (28, 29). Furthermore,
(in terms of educational level attained, income, enteral feeding has been found to aid in the development of the
and establishment of family) (4). gut and lower the risk of infections and sepsis (26, 30).

Frontiers in Nutrition  |  www.frontiersin.org 2 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

When Should Enteral Feeding Be Started: Can Preterm Infants Be Fed While on
Early or Late? Ventilator or Continuous Positive Airway
Delaying the administration of progressive enteral feeding to Pressure (CPAP)?
reduce the risk of NEC may not be an appropriate approach. Ventilator or CPAP treatment should not serve as a hindrance
Systematic reviews have reported that delaying enteral feeding to enteral feeding. Assisted ventilation does not increase the
does not lead to a reduction in the risk of NEC. On the contrary, risk of gastroesophageal reflux and is, therefore, not a con-
this approach may prolong the time to achieve full enteral traindication to enteral feeding in preterm LBW infants (43).
feeding (31). Furthermore, early versus late (<48 versus >72 h Although nasal CPAP therapy results in gaseous bowel
after birth) initiation of enteral feeding has been found to be distension or the CPAP belly syndrome in the majority of very
associated with a significantly lesser time to gain birth weight, LBW infants, this may not be attributed to NEC; the feeding
and shorter duration of parenteral nutrition and hospital stay, method has no correlation with the occurrence of the CPAP belly
without any increase in the complication rate (32). A reduced syndrome (44).
incidence of osteopenia of prematurity and jaundice has also
been noted with early versus late enteral feeding in very LBW
infants (33). Is Routine Evaluation of Gastric Aspiration
and Abdominal Girth Justified?
Gastric aspiration and evaluation of gastric residuals may delay
Advancement of Volume of Enteral Feed:
enteral tube feeding and cause damage to the gastric mucosa
Rapid versus Slow (45, 46). Increase in abdominal girth < 1.5 cm occurs normally,
Several studies have assessed the outcomes with rapid versus slow and in the absence of any clinical signs, this may not be indica-
advancement of enteral feeding in preterm infants. In a study tive of any disease (47). Therefore, it is advisable to avoid routine
conducted in neonates with LBW (<1,250 g), rapid versus slow checking of gastric residuals and abdominal girth.
advancement of enteral feeding was associated with a significantly
lesser time required to achieve full enteral feeding and regain
birth weight; a shorter duration of hospital stay; comparable feed NUTRITION TO PRETERM INFANTS—
tolerance; and no increase in the risk of NEC. EBM was used in WHAT TO FEED?
this study (28). Several other randomized studies and systematic
reviews using formula milk have also reported similar findings Growth Rate with Standard Nutritional
(31, 34, 35).
Practices
The prevailing nutritional practices for preterm infants include
Frequency of Feeding: Continuous versus minimal enteral feeds (10 mL/kg/day); use of breast milk, donor
Hourly milk (if maternal milk is not available), or fortified human milk;
Continuous nasogastric versus intermittent bolus milk feeding feed advancements of 20  mL/kg/day; and parenteral nutrition
in preterm LBW infants has long been a topic of debate. While with amino acids and lipids (48). Despite following standard
a few studies reveal comparable outcomes with the two feeding practices, the growth in preterm infants may not be optimal
methods, a few others support a significantly faster growth rate in most cases. Current nutritional strategies or practices being
and achievement of full enteral feeding with the continuous followed for intrauterine growth restriction (IUGR)/preterm
feeding technique (36, 37). A systematic review of these feeding infants are not able to prevent postnatal growth restriction (49).
methods in infants weighing less than 1,500  g reported faster Extrauterine growth restriction (EUGR) is a serious issue in
weight gain and earlier hospital discharge with the continuous preterm LBW infants, with an incidence of about 28, 34, and 16%
tube feeding method (38). Furthermore, a significant increase in for weight, length, and head circumference, respectively (50).
pulmonary resistance, airflow, and respiratory instability and a The growth rate of preterm and extremely LBW infants during
decrease in cerebral perfusion have been noted with the bolus hospitalization in the neonatal intensive care unit significantly
feeding method (39, 40) Therefore, continuous feeding seems impacts neurodevelopmental and growth outcomes in later
to be logical in preterm infants till the development of sucking– life (51).
swallowing coordination.
Current Enteral Nutritional Strategies—
How to Feed Enterally: Nasogastric versus Pros and Cons
Orogastric Route The enteral nutrition options for preterm infants include EBM,
The passage of nasogastric tube has been noted to increase air- fortified EBM, and formula milk (52).
way resistance in preterm infants by 30–50% (41). An increased
incidence of periodic breathing and central apnea has also been Expressed Breast Milk
noted with nasogastric tubes, in preterm infants (42). Hence, Breast milk should be the milk of choice for providing nutrition
the routine use of nasogastric tubes is not advisable in preterm to preterm LBW infants, due to its several inherent advantages
infants. (Table 2) (53–69).

Frontiers in Nutrition  |  www.frontiersin.org 3 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

Table 3 | Recommended enteral nutrient intakes for preterm infants.


Consensus recommendations on the use of mother’s milk for feeding
preterm infants. Nutrient Per kg per day Nutrient Per kg per day

• The first choice of human milk for feeding preterm infants is expressed Fluid, mL 135–200 Calcium, mg 120–140
breast milk from the mother; the second choice is donor pasteurized Energy, kcal 110–135 Phosphate, mg 60–90
human milk. Protein, g 3.5–4.5 Vitamin D, IU 800–1,000
• “Donor pasteurized human milk” should be the term used for donor milk. Fat, g 4.8–6.6 Vitamin A, IU 1,300–3,300
• A pasteurizer should be used for pasteurization of human milk; unpasteur- Carbohydrates, g 11.6–13.2 Iron, mg 2–3
ized milk should not be used in case of donor human milk.
• Donor pasteurized human milk should be screened for human immuno-
deficiency virus (HIV), hepatitis C virus (HCV), hepatitis B antigen (HBsAg),
venereal disease, and bacteria, using relevant tests/cultures.
• The donor mother should also be screened for HIV, HCV, HBsAg, and venereal
Given the high nutrient needs of preterm infants, human milk
disease within 6 months of donating milk. alone may not be able to comprehensively provide the preterm
• If milk banks exist locally, pooled milk may be used, provided proper infants’ requirement of proteins, energy, minerals, vitamins, and
consent has been obtained. trace elements (71). Furthermore, feeding preterm infants with
• Milk banks should liaise with agencies for pasteurization of pooled small
human milk alone may result in slower growth rates and lesser
aliquots of breast milk.
• Donor human milk may be stored at −20°C for 6 months; preterm infants increase in head circumference as compared to feeding with for-
should not be fed milk stored for more than 3 months. tified human milk (72). According to ESPGHAN, the preferred
• Although the human milk analyzer is a useful tool to analyze the nutrient nutrition for premature infants is fortified human milk, prefer-
content of human milk and enable subsequent fortification, it is currently ably from the infant’s own mother, or formula milk designed for
being used as only a research tool and not in day-to-day clinical prac-
tice. The standard fortification method is recommended in daily clinical
preterm infants (70).
settings.
Fortified Human Milk
Fortifying breast milk helps in providing additional nutrients
Table 2 | Advantages of feeding breast milk.
to LBW infants whose needs are not met by EBM alone (53).
Overall outcomes Fortification is based on the principle of increasing the concentra-
• Better feed tolerance (53, 54). tion of nutrients to such levels that the infant’s needs are met with
• Lower risk of NEC, sepsis, and late-onset sepsis (53–55). customary feeding volumes (73). Fortification may be done with
• Reduced length of hospital stay and risk of rehospitalization (56, 57).
any one nutrient (monocomponent) or multiple nutrients (mul-
Microvascular outcome ticomponent). Although there is a lack of evidence supporting
• Protective role in preventing retinopathy of prematurity (58).
the benefits of monocomponent fortification with carbohydrates
Cardiovascular outcomes
• Lower risk of hypertension and atherosclerosis later in life (59, 60).
and fats, fortification with proteins has been found to result in
• Improved left and right ventricular end-diastolic volume index and stroke an increase in weight gain, linear growth, and head growth in
volume index, and beneficial long-term cardiovascular outcomes (61). preterm infants. However, protein-alone fortification has not
Bone health been associated with long-term growth and neurodevelopmental
• Significant increase in whole-body bone area and bone mineral content (62). outcomes (74, 75). While fortifying human milk, it is essential to
Neurological outcomes balance the osmolality of the feed, as fortification increases the
• Improved neurological development in later years (53).
osmolality of the feed (53).
• Significantly higher IQ in later years, even after adjusting for maternal IQ
(63–65).
The benefits of enteral feeds rich in docosahexaenoic acid
• Better receptive language at 3 years and verbal and non-verbal IQ at (DHA) are debatable. Dietary DHA has been reported to be an
7 years. This outcome has been noted with a longer duration of feeding important nutrient affecting neurological development in pre-
breast milk; each month of feeding breast milk may increase verbal IQ by 0.35 term infants, in some reviews (76). Furthermore, studies report
points and non-verbal IQ by 0.29 points (66).
significantly improved cognitive function in preterm girls fed
• Significant improvement in white matter microstructure, which may translate
to improved cognitive, behavioral, and real-world academic performance enteral feeds high in DHA (77). However, a few other systematic
(67, 68). reviews indicate no benefit or harm with long-chain polyunsatu-
• Significantly higher brain volume and white matter volume, which in turn rated fatty acid supplementation in preterm infants (78).
correlates with significantly improved verbal IQ, especially in boys (25% Multicomponent fortification has been associated with
increase in IQ) (69).
short-term weight gain and linear and head growth in preterm
• Improved mental and psychomotor development, and behavioral scores
(57). infants. However, long-term evidences are lacking (72, 79). The
National Neonatology Forum of India recommends the use
of multicomponent fortification to infants born at <32  weeks’
gestation, or with <1,500 g birth weight who fail to gain weight
Although feeding human milk has several proven benefits, it is despite receiving full volumes of breast milk (up to 180–200 mL/
essential to determine whether it can meet the higher nutritional kg/day) (80).
needs of preterm infants. The recommended enteral macro and
micronutrient intakes for preterm infants according to the Clinical Evidences on Fortified Human Milk
European Society of Pediatric Gastroenterology, Hepatology, and Few Indian studies have reported the outcomes of feed-
Nutrition (ESPGHAN) have been summarized in Table 3 (70). ing fortified human milk to preterm infants. In a study

Frontiers in Nutrition  |  www.frontiersin.org 4 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

by Mukhopadhyay et  al., in 166 preterm infants weighing Consensus recommendations on the supplementation of protein,
≤1,500 g, infants fed fortified EBM experienced significantly calories, and calcium in daily clinical practice, for the optimum
better growth in terms of weight gain, length, and head circum- growth of preterm infants.
ference when compared to infants exclusively fed human milk. • HMF may be used only when the infant reaches a feed of 100 mL/kg/day.
This effect was more evident in infants who were small for However, based on the clinical situation, appropriate amounts of HMF may
gestational age; length and weight were significantly increased be used even before the set feed limit has been achieved.
in this group, which was fed fortified EBM (81). In a study by • In clinical practice, one sachet (1 g) of HMF may be used for 20 or 25 mL
of expressed or donor pasteurized human milk, depending on the product
Gathwala et al., fortification of EBM was not associated with
guideline.
delayed gastric emptying or feed intolerance in preterm neo- • It is advisable to monitor the calorie and protein intake of the infant to
nates (82). In a study by Agarwal et al., there was an increase evaluate the protein requirements.
in osmolality of breast milk after the addition of human milk • The recommended protein intake should be 3.5–4 g/kg/day, depending on
fortifier (HMF). However, the osmolality did not change after the birth weight and desired growth.
• It is important to monitor the growth velocity of the infant, along with
storage at 4°C for 6 h (83). These results are in contrast to those
monitoring for osteopenia of maturity.
noted in an international study by Henriksen et al. in preterm • The calorie requirement of a preterm infant is usually met with the addition
very LBW infants fed fortified milk. The EUGR of these infants of HMF, which provides about 4 g/kg/day of protein and 3.5–4 g/kg/day
increased to 58% at hospital discharge, from 33% at birth (84). of fats.
Therefore, the outcomes may not be optimal in all cases; this • A higher protein and higher calorie strategy will help optimize nutrition in
preterm infants.
may be due to the low energy and protein content of the feeds
• Calculation of protein intake while adding HMF to EBM will help in the
(84, 85). simultaneous monitoring of the requirements for carbohydrates, fats, and
Standard fortification of human milk may be associated calories.
with protein deficits, as the protein content is low during • There is no need to monitor and supplement additional calcium in preterm
fortification, assuming the availability of higher amounts of infants on total parenteral nutrition and HMF; additional supplementation of
calcium may result in nephrocalcinosis.
protein in human milk (86). However, it must be noted that • Serum calcium may be measured at 12–24 h after birth, followed by regular
the protein content of EBM is variable and may differ based monitoring, to prevent hypercalcemia.
on the duration of lactation and from sample to sample (86, • Serum phosphorus and alkaline phosphatase may be measured once a
87). Therefore, standard fortification of human milk may not week after 2 weeks of birth, to detect any osteopenia of prematurity and to
take appropriate measures.
meet the recommended protein intake in preterm infants (88).
To overcome this limitation and optimize human milk forti-
fication, the concept of individualized fortification has been
introduced. Side Effects of Fortification
• Earlier studies report significantly delayed gastric emptying
Individualized Fortification in some preterm infants who cannot tolerate fortified milk
Two methods of individualized fortification have been pro- (93). However, recent studies suggest that fortifying breast
posed—targeted and adjustable (86). milk may not result in clinically significant feeding intoler-
Targeted fortification—this method involves analyzing the ance if the recommended concentrations of fortifier are used
protein content of human milk and fortifying it to meet the (94).
infant’s nutrient requirement. A target protein intake is chosen • The use of fortifiers containing iron may decrease the antibac-
based on predefined infant requirements. This method helps in terial action of preterm milk (95).
individualized fortification of preterm infants (89). • There is no clear evidence linking human milk fortification
Adjustable fortification—the protein intake in this method with NEC risk (72). Hence, weighing the benefits of growth
is periodically adjusted based on the metabolic response of the and neurodevelopment with fortification versus possible
infant evaluated from the blood urea nitrogen tests. This method feed intolerance and NEC is at the sole discretion of the
is more suitable for stable preterm infants. Furthermore, it is a clinician.
more practical and feasible method that does not require frequent
analyses of the milk (86). In comparison to the standard fortifica-
Formula Milk
tion method, the adjustable protein fortification technique has
Formula milk contains all essential nutrients and is specifically
been found to result in a significant improvement in growth
designed to meet the requirements of LBW infants (53).
indices, weight gain, and head circumference in preterm very
LBW infants. This improvement significantly correlated with the
higher protein intake in infants fed using the adjustable regimen
(90, 91). Consensus recommendations on choosing the right preterm formula.

In this context, it may be pertinent to mention that excess • Osmolality is a key factor that needs to be taken into consideration when
protein intake to promote faster postnatal growth may not nec- choosing a preterm formula.
essarily be beneficial; it may result in increased blood pressure • Preterm formula can be used in preterm infants weighing less than
1.5 kg.
in the long term (92). Therefore, it is essential to optimize the
• Docosahexaenoic acid (DHA) is an important part of preterm formula.
nutrient intake based on the infants’ requirements.

Frontiers in Nutrition  |  www.frontiersin.org 5 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

OPTIMIZING GROWTH IN PRETERM is advised to initiate expression of breast milk soon after birth.
INFANTS TO LOWER CARDIOVASCULAR Relaxation, massaging, and warming the breasts, hand expres-
sion, and the use of low-cost pumps may be some cost-effective
RISK interventions for expressing breast milk (108). Combining pump
Faster weight gain in preterm infants may be associated with suction, breast compression, and hand expression has been found
an increased risk of overweight and obesity; higher body fat to enhance breast milk production (109). In any case, early nutri-
percentage, waist circumference, serum triglycerides, and blood tion in preterm LBW infants should be critically optimized to have
pressure; endothelial dysfunction; and adverse long-term cardio- beneficial effects, both in the short term and the long term (110).
vascular outcomes (96–99). Therefore, monitoring and ensuring
optimal weight gain is essential throughout the entire gestational KEY CONCLUSION
spectrum, to prevent long-term complications.
• Preterm birth survivors present with higher rates of adverse
developmental disabilities and health outcomes compared to
POSTDISCHARGE NUTRITION their term counterparts.
OF PRETERM INFANTS • Optimal nutrition is highly essential for growth, metabolism,
and immunity in preterm LBW infants; it lowers the risk of
Although there is no clear evidence on the added benefit of
adverse morbidities in adulthood.
administering a nutrient-enriched diet to preterm infants after
• Enteral feeding may be preferred to parenteral feeding due to
discharge, a few reviews suggest an improvement in growth
the complications associated with the latter.
parameters with no effect on neurodevelopmental outcomes
• Early and rapid initiation of enteral feeding has several advan-
(100, 101). Studies indicate that variations in dietary nutrient
tages compared to late and slow feeding; continuous feeding
intake can contribute significantly to growth deficits in preterm
may be preferred to intermittent hourly feeding.
infants, thus highlighting the choice of appropriate nutrition in
• Early enteral feeding does not carry any additional risk of NEC
this cohort (102, 103).
in preterm infants; on the contrary, it aids in the development
The numerous benefits of mother’s milk have been discussed
of the gut and reduces the risk of infections.
previously. While a few studies have reported comparable
• EBM should be the first choice for feeding preterm infants
outcomes, a few others have reported better short-term growth
due to its numerous inherent advantages; donor pasteurized
rates in preterm infants fed human milk compared to formula
human milk is the second choice.
milk (104, 105). A meta-analysis of 14 randomized clinical trials
• Given the high nutrient requirements of preterm infants, espe-
reveals increased short-term growth rates with fortified human
cially the requirements of proteins, EBM or donor milk should
milk compared to unfortified breast milk, with no increase in the
be fortified with HMF without increasing the osmolality of the
risk of NEC (72). Furthermore, fortifying human milk using a
milk.
human milk-based fortifier has been found to have a significantly
• In routine clinical practice, standard fortification may be fol-
lower risk of NEC and lower morbidity and mortality compared
lowed. However, the use of targeted and adjustable fortification,
to bovine milk-based fortifier (106, 107).
where possible, may help in optimal nutrient supplementation
Owing to the beneficial effects of breast milk and the para-
to preterm infants.
doxical inability of preterm LBW infants to feed at the breast, it
• Monitoring the growth velocity is essential, as rapid weight
gain in preterm infants may be associated with future cardio-
Consensus recommendations on postdischarge nutrition. vascular risk. Hence, optimal weight gain should be the target.
• Counseling and regular follow-up after discharge and moni-
• The ideal discharge weight of preterm infants may be dependent on several
factors, including the ability of local services to take care of the infant after
toring of the preterm infant, preferably until adolescence, are
discharge. advisable.
• The sucking/swallowing ability of infants should be good at discharge.
• The following signs should be monitored and immediate medical attention
should be sought for: inability to feed, lethargy, temperature alterations, and
AUTHOR CONTRIBUTIONS
mottling/cyanosis.
• Counseling should be provided for resuscitation prior to discharge.
All the authors contributed equally to the Conceptualization,
• The first follow-up should be done within 3–7 days, followed by a weekly review, and finalization of the manuscript.
follow-up.
• Preterm infants should be monitored for up to a minimum of 2 years and
preferably till adolescence.
ACKNOWLEDGMENTS
• In preterm infants who are on formula milk, switching to a standard formula
is recommended after they have reached their birth centile (i.e., after
We would like to acknowledge BioQuest Solutions for their sup-
catch-up has been completed). There is not much evidence on the benefits port in editorial services.
of switching after an increase in weight beyond this.
• Complementary feeding may be initiated at the corrected age of
4 months. No special attention is required while initiating complementary
FUNDING
feeding.
The program is funded by Nestle Nutrition Institute, South Asia.

Frontiers in Nutrition  |  www.frontiersin.org 6 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

REFERENCES development to adulthood. J Pediatr (2003) 142:463–8. doi:10.1067/mpd.


2003.149
1. March of Dimes, Partnership for Maternal Newborn and Child Health, Save 22. Morgan C, McGowan P, Herwitker S, Hart AE, Turner MA. Postnatal head
the Children, World Health Organization. Howson CP, Kinney MV, Lawn growth in preterm infants: a randomized controlled parenteral nutrition
JE, editors. Born Too Soon: the Global Action Report on Preterm Birth (2012). study. Pediatrics (2014) 133:e120–8. doi:10.1542/peds.2013-2207
Available from: http://www.who.int/pmnch/media/news/2012/201204_born- 23. Leppänen M, Lapinleimu H, Lind A, Matomäki J, Lehtonen L, Haataja L, et al.
toosoon-report.pdf Antenatal and postnatal growth and 5-year cognitive outcome in very preterm
2. Doyle LW, Anderson PJ. Adult outcome of extremely preterm infants. Pediatrics infants. Pediatrics (2014) 133(1):63–70. doi:10.1542/peds.2013-1187
(2010) 126:342–51. doi:10.1542/peds.2010-0710 24. Ehrenkranz RA. Early, aggressive nutritional management for very low
3. Nosarti C, Al-Asady MH, Frangou S, Stewart AL, Rifkin L, Murray RM. birth weight infants: what is the evidence? Semin Perinatol (2007) 31:48–55.
Adolescents who were born very preterm have decreased brain volumes. Brain doi:10.1053/j.semperi.2007.02.001
(2002) 125(Pt 7):1616–23. doi:10.1093/brain/awf157 25. Maroulis J, Kalfarentzos F. Complications of parenteral nutrition at the end of
4. Moster D, Lie RT, Markestad T. Long-term medical and social conse- the century. Clin Nutr (2000) 19:295–304. doi:10.1054/clnu.1999.0089
quences of preterm birth. N Engl J Med (2008) 359:262–73. doi:10.1056/ 26. Leaf A. Early enteral feeding in high-risk preterm infants. Nutrition (2007)
NEJMoa0706475 3(1):27–30.
5. Rotteveel J, van Weissenbruch MM, Twisk JW, Delemarre-Van de Waal HA. 27. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis. Lancet
Infant and childhood growth patterns, insulin sensitivity, and blood pressure (1990) 336:1519–23. doi:10.1016/0140-6736(90)93304-8
in prematurely born young adults. Pediatrics (2008) 122:313–21. doi:10.1542/ 28. Salhotra A, Ramji S. Slow versus fast enteral feed advancement in very low
peds.2007-2012 birth weight infants: a randomized control trial. Indian Pediatr (2004)
6. Uthaya S, Thomas EL, Hamilton G, Doré CJ, Bell J, Modi N. Altered adiposity 41(5):435–41.
after extremely preterm birth. Pediatr Res (2005) 57:211–5. doi:10.1203/01. 29. Morgan J, Young L, McGuire W. Slow advancement of enteral feed volumes
PDR.0000148284.58934.1C to prevent necrotising enterocolitis in very low birth weight infants. Cochrane
7. Rossi P, Tauzin L, Marchand E, Boussuges A, Gaudart J, Frances Y. Respective Database Syst Rev (2014) (12):CD001241. doi:10.1002/14651858.CD001241.
roles of preterm birth and fetal growth restriction in blood pressure and arterial pub5
stiffness in adolescence. J Adolesc Health (2011) 48:520–2. doi:10.1016/ 30. Flidel-Rimon O, Friedman S, Lev E, Juster-Reicher A, Amitay M, Shinwell ES.
j.jadohealth.2010.08.004 Early enteral feeding and nosocomial sepsis in very low birthweight infants.
8. Lewandowski AJ, Bradlow WM, Augustine D, Davis EF, Francis J, Singhal A, Arch Dis Child Fetal Neonatal Ed (2004) 89:F289–92. doi:10.1136/adc.
et  al. Right ventricular systolic dysfunction in young adults born preterm. 2002.021923
Circulation (2013) 128:713–20. doi:10.1161/CIRCULATIONAHA.113.002583 31. Morgan J, Young L, McGuire W. Delayed introduction of progressive enteral
9. Hovi P, Andersson S, Järvenpää AL, Eriksson JG, Strang-Karlsson S, Kajantie E, feeds to prevent necrotising enterocolitis in very low birth weight infants.
et  al. Decreased bone mineral density in adults born with very low birth Cochrane Database Syst Rev (2014) (12):CD001970. doi:10.1002/14651858.
weight: a cohort study. PLoS Med (2009) 6:e1000135. doi:10.1371/journal. CD001970.pub5
pmed.1000135 32. Sallakh-Niknezhad A, Bashar-Hashemi F, Satarzadeh N, Ghojazadeh M,
10. Patel RM, Kandefer S, Walsh MC, Bell EF, Carlo WA, Laptook AR, et al. Causes Sahnazarli G. Early versus late trophic feeding in very low birth weight
and timing of death in extremely premature infants from 2000 through 2011. preterm infants. Iran J Pediatr (2012) 22(2):171–6.
N Engl J Med (2015) 372:331–40. doi:10.1056/NEJMoa1403489 33. Dunn L, Hulman S, Weiner J, Kliegman R. Beneficial effects of early hypo-
11. Hanson C, Sundermeier J, Dugick L, Lyden E, Anderson-Berry AL. Imple­ caloric enteral feeding on neonatal gastrointestinal function: preliminary
mentation, process, and outcomes of nutrition best practices for infants report of a randomized trial. J Pediatr (1988) 112:622–9. doi:10.1016/
<1500 g. Nutr Clin Pract (2011) 26:614–24. doi:10.1177/0884533611418984 S0022-3476(88)80185-9
12. Donovan R, Puppala B, Angst D, Coyle BW. Outcomes of early nutrition 34. Rayyis SF, Ambalavanan N, Wright L, Carlo WA. Randomized trial of “slow”
support in extremely low-birth-weight infants. Nutr Clin Pract (2006) versus “fast” feed advancements on the incidence of necrotizing enterocolitis
21:395–400. doi:10.1177/0115426506021004395 in very low birth weight infants. J Pediatr (1999) 134:293–7. doi:10.1016/
13. Ganapathy S. Long chain polyunsaturated fatty acids and immunity in infants. S0022-3476(99)70452-X
Indian Pediatr (2009) 46(9):785–90. 35. Karagol BS, Zenciroglu A, Okumus N, Polin RA. Randomized controlled trial
14. Lee KA, Hayes BC. Head size and growth in the very preterm infant: a litera- of slow vs rapid enteral feeding advancements on the clinical outcomes of
ture review. Res Rep Neonatol (2015) 5:1–7. doi:10.2147/RRN.S74449 preterm infants with birth weight 750–1250 g. J Parenter Enteral Nutr (2013)
15. Vinall J, Grunau RE, Brant R, Chau V, Poskitt KJ, Synnes AR, et al. Slower post- 37:223–8. doi:10.1177/0148607112449482
natal growth is associated with delayed cerebral cortical maturation in preterm 36. Silvestre MA, Morbach CA, Brans YW, Shankaran S. A prospective random-
newborns. Sci Transl Med (2013) 5:168ra8. doi:10.1126/scitranslmed.3004666 ized trial comparing continuous versus intermittent feeding methods in very
16. Barker DJ, Winter PD, Osmond C, Margetts B, Simmonds SJ. Weight in low birth weight neonates. J Pediatr (1996) 128:748–52. doi:10.1016/S0022-
infancy and death from ischaemic heart disease. Lancet (1989) 2:577–80. 3476(96)70324-4
doi:10.1016/S0140-6736(89)90710-1 37. Dsilna A, Christensson K, Alfredsson L, Lagercrantz H, Blennow M.
17. Barker DJ, Eriksson JG, Forsén T, Osmond C. Fetal origins of adult disease: Continuous feeding promotes gastrointestinal tolerance and growth in very
strength of effects and biological basis. Int J Epidemiol (2002) 31:1235–9. low birth weight infants. J Pediatr (2005) 147:43–9. doi:10.1016/j.jpeds.
doi:10.1093/ije/31.6.1235 2005.03.003
18. Isaacs EB, Gadian DG, Sabatini S, Chong WK, Quinn BT, Fischl BR, et  al. 38. Premji SS, Chessell L. Continuous nasogastric milk feeding versus intermit-
The effect of early human diet on caudate volumes and IQ. Pediatr Res (2008) tent bolus milk feeding for premature infants less than 1500 grams. Cochrane
63:308–14. doi:10.1203/PDR.0b013e318163a271 Database Syst Rev (2011) (11):CD001819. doi:10.1002/14651858.CD001819.
19. Franz AR, Pohlandt F, Bode H, Mihatsch WA, Sander S, Kron M, et  al. pub2
Intrauterine, early neonatal, and postdischarge growth and neurodevel- 39. Blondheim O, Abbasi S, Fox WW, Bhutani VK. Effect of enteral gavage feeding
opmental outcome at 5.4 years in extremely preterm infants after intensive rate on pulmonary functions of very low birth weight infants. J Pediatr (1993)
neonatal nutritional support. Pediatrics (2009) 123:e101–9. doi:10.1542/ 122:751–5. doi:10.1016/S0022-3476(06)80021-1
peds.2008-1352 40. Nelle M, Hoecker C, Linderkamp O. Effects of bolus tube feeding on cerebral
20. Stephens BE, Walden RV, Gargus RA, Tucker R, McKinley L, Mance M, et al. blood flow velocity in neonates. Arch Dis Child Fetal Neonatal Ed (1997)
First-week protein and energy intakes are associated with 18-month devel- 76:F54–6. doi:10.1136/fn.76.1.F54
opmental outcomes in extremely lowbirth weight infants. Pediatrics (2009) 41. Stocks J. Effect of nasogastric tubes on nasal resistance during infancy. Arch
123:1337–43. doi:10.1542/peds.2008-0211 Dis Child (1980) 55:17–21. doi:10.1136/adc.55.1.17
21. Brandt I, Sticker EJ, Lentze MJ. Catch-up growth of head circumference of 42. van Someren V, Linnett SJ, Stothers JK, Sullivan PG. An investigation into
very low birth weight, small for gestational age preterm infants and mental the benefits of resiting nasoenteric feeding tubes. Pediatrics (1984) 74:379–83.

Frontiers in Nutrition  |  www.frontiersin.org 7 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

43. Newell SJ, Morgan ME, Durbin GM, Booth IW, McNeish AS. Does mechan- 66. Belfort MB, Rifas-Shiman SL, Kleinman KP, Guthrie LB, Bellinger DC,
ical ventilation precipitate gastro-oesophageal reflux during enteral feeding? Taveras EM, et  al. Infant feeding and childhood cognition at ages 3 and 7
Arch Dis Child (1989) 64:1352–5. doi:10.1136/adc.64.10_Spec_No.1352 years: effects of breastfeeding duration and exclusivity. JAMA Pediatr (2013)
44. Jaile JC, Levin T, Wung JT, Abramson SJ, Ruzal-Shapiro C, Berdon WE. Benign 167:836–44. doi:10.1001/jamapediatrics.2013.455
gaseous distension of the bowel in premature infants treated with nasal con- 67. Deoni SC, Dean DC III, Piryatinsky I, O’Muircheartaigh J, Waskiewicz N,
tinuous airway pressure: a study of contributing factors. AJR Am J Roentgenol Lehman K, et  al. Breastfeeding and early white matter development: a
(1992) 158:125–7. doi:10.2214/ajr.158.1.1727337 cross-sectional study. Neuroimage (2013) 82:77–86. doi:10.1016/j.neuroimage.
45. McClave SA, Snider HL. Clinical use of gastric residual volumes as a monitor 2013.05.090
for patients on enteral tube feeding. JPEN J Parenter Enteral Nutr (2002) 68. Northam GB, Liégeois F, Chong WK, Wyatt JS, Baldeweg T. Total brain white
26:S43–8. doi:10.1177/014860710202600607 matter is a major determinant of IQ in adolescents born preterm. Ann Neurol
46. Parker L, Torrazza RM, Li Y, Talaga E, Shuster J, Neu J. Aspiration and evalu- (2011) 69:702–11. doi:10.1002/ana.22263
ation of gastric residuals in the neonatal intensive care unit: state of the science. 69. Isaacs EB, Fischl BR, Quinn BT, Chong WK, Gadian DG, Lucas A. Impact of
J Perinat Neonatal Nurs (2015) 29:51–9. doi:10.1097/JPN.0000000000000080 breast milk on intelligence quotient, brain size, and white matter development.
47. Bhatia P, Johnson KJ, Bell EF. Variability of abdominal circumference Pediatr Res (2010) 67:357–62. doi:10.1203/PDR.0b013e3181d026da
of premature infants. J Pediatr Surg (1990) 25:543–4. doi:10.1016/0022- 70. ESPGHAN Committee on Nutrition, Agostoni C, Braegger C, Decsi T,
3468(90)90569-U Kolacek S, Koletzko B, et al. Role of dietary factors and food habits in the devel-
48. Ziegler EE, Carlson SJ.  Early nutrition of very low birth weight infants. J Matern opment of childhood obesity: a commentary by the ESPGHAN Committee
Fetal Neonatal Med (2009) 22:191–7. doi:10.1080/14767050802630169 on Nutrition. J Pediatr Gastroenterol Nutr (2011) 52:662–9. doi:10.1097/MPG.
49. Puntis JW. Nutritional support in the premature newborn. Postgrad Med J 0b013e3182169253
(2006) 82:192–8. doi:10.1136/pgmj.2005.038109 71. Kumar P, Sundaram V. Protein Supplementation of Human Milk for Promoting
50. Clark RH, Thomas P, Peabody J. Extrauterine growth restriction remains a Growth in Preterm Infants. (2011). Available from: https://extranet.who.int/
serious problem in prematurely born neonates. Pediatrics (2003) 111:986–90. rhl/topics/newborn-health/newborn-nutrition-and-feeding/protein-
doi:10.1542/peds.111.5.986 supplementation-human-milk-promoting-growth-preterm-infants
51. Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage LA, Poole WK. 72. Brown JV, Embleton ND, Harding JE, McGuire W. Multi-nutrient fortifica-
Growth in the neonatal intensive care unit influences neurodevelopmental tion of human milk for preterm infants. Cochrane Database Syst Rev (2016)
and growth outcomes of extremely low birth weight infants. Pediatrics (2006) (5):CD000343. doi:10.1002/14651858.CD000343.pub3
117:1253–61. doi:10.1542/peds.2005-1368 73. Di Natale C, Coclite E, Di Ventura L, Di Fabio S. Fortification of maternal milk
52. Su BH. Optimising nutrition in preterm infants. Pediatr Neonatol (2014) for preterm infants. J Matern Fetal Neonatal Med (2011) 24(Suppl 1):41–3.
55:5–13. doi:10.1016/j.pedneo.2013.07.003 doi:10.3109/14767058.2011.607569
53. Preterm and low birth weight babies. In: Bentley D, Aubrey S, Bentley M, 74. Kuschel CA, Harding JE. Protein supplementation of human milk for
editors. Infant Feeding and Nutrition for Primary Care. Abingdon: Radcliffe promoting growth in preterm infants. Cochrane Database Syst Rev (2000)
Medical Press Ltd (2004). p. 47–51. (2):CD000433. doi:10.1002/14651858.CD000433
54. Section on Breastfeeding. Breastfeeding and the use of human milk. Pediatrics 75. Fenton TR, Premji SS, Al-Wassia H, Sauve RS. Higher versus lower protein
(2012) 129:e827–41. doi:10.1542/peds.2011-3552 intake in formula-fed low birth weight infants. Cochrane Database Syst Rev
55. Schanler RJ, Shulman RJ, Lau C. Feeding strategies for premature infants: (2014) (4):CD003959. doi:10.1002/14651858.CD003959.pub3
beneficial outcomes of feeding fortified human milk versus preterm formula. 76. Szajewska H, Makrides M. Is early nutrition related to short-term health
Pediatrics (1999) 103:1150–7. doi:10.1542/peds.103.6.1150 and long-term outcome? Ann Nutr Metab (2011) 58(Suppl 1):38–48.
56. Ganapathy V, Hay JW, Kim JH. Costs of necrotizing enterocolitis and doi:10.1159/000323465
cost-effectiveness of exclusively human milk-based products in feeding 77. Makrides M, Gibson RA, McPhee AJ, Collins CT, Davis PG, Doyle LW, et al.
extremely premature infants. Breastfeed Med (2012) 7:29–37. doi:10.1089/ Neurodevelopmental outcomes of preterm infants fed high-dose docosa-
bfm.2011.0002 hexaenoic acid: a randomized controlled trial. JAMA (2009) 301:175–82.
57. Vohr BR, Poindexter BB, Dusick AM, McKinley LT, Higgins RD, Langer JC, doi:10.1001/jama.2008.945
et al. Persistent beneficial effects of breast milk ingested in the neonatal inten- 78. Schulzke SM, Patole SK, Simmer K. Long-chain polyunsaturated fatty acid
sive care unit on outcomes of extremely low birth weight infants at 30 months supplementation in preterm infants. Cochrane Database Syst Rev (2011)
of age. Pediatrics (2007) 120:e953–9. doi:10.1542/peds.2006-3227 (2):CD000375. doi:10.1002/14651858.CD000375.pub4
58. Zhou J, Shukla VV, John D, Chen C. Human milk feeding as a protective 79. Kuschel CA, Harding JE. Multicomponent fortified human milk for promoting
factor for retinopathy of prematurity: a meta-analysis. Pediatrics (2015) growth in preterm infants. Cochrane Database Syst Rev (2004) (1):CD000343.
136:e1576–86. doi:10.1542/peds.2015-2372 doi:10.1002/14651858.CD000343.pub2
59. Singhal A, Cole TJ, Lucas A. Early nutrition in preterm infants and later 80. National Neonatology Forum, India. Bhakoo ON, Kumar P, Jain N, Thakre R,
blood pressure: two cohorts after randomised trials. Lancet (2001) 357:413–9. Murki S, Venkataseshan S, editors. Evidence Based Clinical Practice Guidelines.
doi:10.1016/S0140-6736(00)04004-6 (2010). Available from: http://www.nnfi.org/images/pdf/nnf_cpg_consoli-
60. Singhal A, Cole TJ, Fewtrell M, Lucas A. Breastmilk feeding and lipoprotein dated_file-january102011.pdf
profile in adolescents born preterm: follow-up of a prospective randomised 81. Mukhopadhyay K, Narnag A, Mahajan R. Effect of human milk fortification in
study. Lancet (2004) 363:1571–8. doi:10.1016/S0140-6736(04)16198-9 appropriate for gestation and small for gestation preterm babies: a randomized
61. Lewandowski AJ, Lamata P, Francis JM, Piechnik SK, Ferreira VM, Boardman H, controlled trial. Indian Pediatr (2007) 44(4):286–90.
et  al. Breast milk consumption in preterm neonates and cardiac shape in 82. Gathwala G, Shaw C, Shaw P, Yadav S, Sen J. Human milk fortification and
adulthood. Pediatrics (2016) 138(1):e20160050. doi:10.1542/peds.2016-0050 gastric emptying in the preterm neonate. Int J Clin Pract (2008) 62:1039–43.
62. Fewtrell MS, Williams JE, Singhal A, Murgatroyd PR, Fuller N, Lucas A. doi:10.1111/j.1742-1241.2006.01201.x
Early diet and peak bone mass: 20 year follow-up of a randomized trial of early 83. Agarwal R, Singal A, Aggarwal R, Deorari AK, Paul VK. Effect of fortification
diet in infants born preterm. Bone (2009) 45:142–9. doi:10.1016/j.bone.2009. with human milk fortifier (HMF) and other fortifying agents on the osmolality
03.657 of pretermbreast milk. Indian Pediatr (2004) 41(1):63–7.
63. Lucas A, Morley R, Cole TJ, Lister G, Leeson-Payne C. Breast milk and 84. Henriksen C, Westerberg AC, Rønnestad A, Nakstad B, Veierød MB, Drevon CA,
subsequent intelligence quotient in children born preterm. Lancet (1992) et  al. Growth and nutrient intake among very-low-birth-weight infants fed
339:261–4. doi:10.1016/0140-6736(92)91329-7 fortified human milk during hospitalisation. Br J Nutr (2009) 102:1179–86.
64. Anderson JW, Johnstone BM, Remley DT. Breast-feeding and cognitive doi:10.1017/S0007114509371755
development: a meta-analysis. Am J Clin Nutr (1999) 70:525–35. 85. Carlson SJ, Ziegler EE. Nutrient intakes and growth of very low birth weight
65. Horta BL, Loret de Mola C, Victora CG. Breastfeeding and intelligence: a sys- infants. J Perinatol (1998) 18:252–8.
tematic review and meta-analysis. Acta Paediatr (2015) 104:14–9. doi:10.1111/ 86. Arslanoglu S, Moro GE, Ziegler EE; The Wapm Working Group On Nutrition.
apa.13139 Optimization of human milk fortification for preterm infants: new concepts

Frontiers in Nutrition  |  www.frontiersin.org 8 May 2017 | Volume 4 | Article 20


Kumar et al. Nutrition in Preterm LBW Infants

and recommendations. J Perinat Med (2010) 38:233–8. doi:10.1515/JPM. Database Syst Rev (2007) (4):CD004862. doi:10.1002/14651858.CD004862.
2010.073 pub2
87. Lemons JA, Moye L, Hall D, Simmons M. Differences in the composition 101. Teller IC, Embleton ND, Griffin IJ, van Elburg RM. Post-discharge formula
of preterm and term human milk during early lactation. Pediatr Res (1982) feeding in preterm infants: a systematic review mapping evidence about
16:113–7. doi:10.1203/00006450-198202000-00007 the role of macronutrient enrichment. Clin Nutr (2016) 35:791–801.
88. Corvaglia L, Aceti A, Paoletti V, Mariani E, Patrono D, Ancora G, et  al. doi:10.1016/j.clnu.2015.08.006
Standard fortification of preterm human milk fails to meet recommended 102. Embleton NE, Pang N, Cooke RJ. Postnatal malnutrition and growth
protein intake: bedside evaluation by near-infrared-reflectance-analysis. Early retardation: an inevitable consequence of current recommendations in
Hum Dev (2010) 86:237–40. doi:10.1016/j.earlhumdev.2010.04.001 preterm infants? Pediatrics (2001) 107:270–3. doi:10.1542/peds.107.2.270
89. Polberger S, Räihä NC, Juvonen P, Moro GE, Minoli I, Warm A. Individualized 103. Olsen IE, Richardson DK, Schmid CH, Ausman LM, Dwyer JT. Intersite dif-
protein fortification of human milk for preterm infants: comparison of ferences in weight growth velocity of extremely premature infants. Pediatrics
ultrafiltrated human milk protein and a bovine whey fortifier. J Pediatr (2002) 110:1125–32. doi:10.1542/peds.110.6.1125
Gastroenterol Nutr (1999) 29:332–8. doi:10.1097/00005176-199909000-00017 104. Corpeleijn WE, de Waard M, Christmann V, van Goudoever JB, Jansen-van
90. Arslanoglu S, Moro GE, Ziegler EE. Adjustable fortification of human milk der Weide MC, Kooi EM, et  al. Effect of donor milk on severe infections
fed to preterm infants: does it make a difference? J Perinatol (2006) 26:614–21. and mortality in very low-birth-weight infants: the early nutrition study
doi:10.1038/sj.jp.7211571 randomized clinical trial. JAMA Pediatr (2016) 170:654–61. doi:10.1001/
91. Alan S, Atasay B, Cakir U, Yildiz D, Kilic A, Kahvecioglu D, et al. An intention jamapediatrics.2016.0183
to achieve better postnatal in-hospital-growth for preterm infants: adjustable 105. Quigley M, McGuire W. Formula versus donor breast milk for feeding
protein fortification of human milk. Early Hum Dev (2013) 89:1017–23. preterm or low birth weight infants. Cochrane Database Syst Rev (2014)
doi:10.1016/j.earlhumdev.2013.08.015 (4):CD002971. doi:10.1002/14651858.CD002971.pub3
92. Singhal A, Cole TJ, Fewtrell M, Kennedy K, Stephenson T, Elias-Jones A, et al. 106. Sullivan S, Schanler RJ, Kim JH, Patel AL, Trawöger R, Kiechl-
Promotion of faster weight gain in infants born small for gestational age: is Kohlendorfer U, et al. An exclusively human milk-based diet is associated
there an adverse effect on later blood pressure? Circulation (2007) 115:213–20. with a lower rate of necrotizing enterocolitis than a diet of human milk
doi:10.1161/CIRCULATIONAHA.106.617811 and bovine milk-based products. J Pediatr (2010) 156:562–7. doi:10.1016/j.
93. Ewer AK, Yu VY. Gastric emptying in pre-term infants: the effect of breast jpeds.2009.10.040
milk fortifier. Acta Paediatr (1996) 85:1112–5. doi:10.1111/j.1651-2227.1996. 107. Abrams SA, Schanler RJ, Lee ML, Rechtman DJ. Greater mortality and mor-
tb14227.x bidity in extremely preterm infants fed a diet containing cow milk protein
94. Yigit S, Akgoz A, Memisoglu A, Akata D, Ziegler EE. Breast milk fortification: products. Breastfeed Med (2014) 9:281–5. doi:10.1089/bfm.2014.0024
effect on gastric emptying. J Matern Fetal Neonatal Med (2008) 21:843–6. 108. Becker GE, Smith HA, Cooney F. Methods of milk expression for lactating
doi:10.1080/14767050802287176 women. Cochrane Database Syst Rev (2016) 9:CD006170. doi:10.1002/
95. Chan GM. Effects of powdered human milk fortifiers on the antibac- 14651858.CD006170.pub5
terial actions of human milk. J Perinatol (2003) 23:620–3. doi:10.1038/ 109. Morton J, Hall JY, Wong RJ, Thairu L, Benitz WE, Rhine WD. Combining
sj.jp.7211003 hand techniques with electric pumping increases milk production in
96. Wang G, Johnson S, Gong Y, Polk S, Divall S, Radovick S, et al. Weight gain mothers of preterm infants. J Perinatol (2009) 29:757–64. doi:10.1038/
in infancy and overweight or obesity in childhood across the gestational jp.2009.87
spectrum: a prospective birth cohort study. Sci Rep (2016) 6:29867. doi:10.1038/ 110. van den Akker CH, Vlaardingerbroek H, van Goudoever JB. Nutritional
srep29867 support for extremely low-birth weight infants: abandoning catabolism in
97. Singhal A, Cole TJ, Fewtrell M, Deanfield J, Lucas A. Is slower early growth the neonatal intensive care unit. Curr Opin Clin Nutr Metab Care (2010)
beneficial for long-term cardiovascular health? Circulation (2004) 109: 13:327–35. doi:10.1097/MCO.0b013e328337d925
1108–13. doi:10.1161/01.CIR.0000118500.23649.DF
98. Kerkhof GF, Willemsen RH, Leunissen RW, Breukhoven PE, Hokken- Conflict of Interest Statement: RK, AS, and UV have none to declare. SB, FA, and
Koelega ACS. Health profile of young adults born preterm: negative effects SR are employees of Nestle Nutrition.
of rapid weight gain in early life. J Clin Endocrinol Metab (2012) 97:4498–506.
doi:10.1210/jc.2012-1716 Copyright © 2017 Kumar, Singhal, Vaidya, Banerjee, Anwar and Rao. This is an
99. Belfort MB, Martin CR, Smith VC, Gillman MW, McCormick MC. Infant open-access article distributed under the terms of the Creative Commons Attribution
weight gain and school-age blood pressure and cognition in former preterm License (CC BY). The use, distribution or reproduction in other forums is permitted,
infants. Pediatrics (2010) 125:e1419–26. doi:10.1542/peds.2009-2746 provided the original author(s) or licensor are credited and that the original publica-
100. Henderson G, Fahey T, McGuire W. Nutrient-enriched formula milk versus tion in this journal is cited, in accordance with accepted academic practice. No use,
human breast milk for preterm infants following hospital discharge. Cochrane distribution or reproduction is permitted which does not comply with these terms.

Frontiers in Nutrition  |  www.frontiersin.org 9 May 2017 | Volume 4 | Article 20


Journal of Pediatric Gastroenterology and Nutrition
42:596 Y 603 Ó May 2006 Lippincott Williams & Wilkins, Philadelphia

Medical Position Paper

Feeding Preterm Infants After Hospital Discharge


A Commentary by the ESPGHAN Committee on Nutrition
ESPGHAN Committee on Nutrition: *Peter J. Aggett2, †Carlo Agostoni, ‡Irene Axelsson,
§Mario De Curtis4, kOlivier Goulet, ¶Olle Hernell, LBerthold Koletzko1, **Harry N. Lafeber,
††Kim F. Michaelsen, ‡‡John W.L. Puntis, §§Jacques Rigo, kkRaanan Shamir,
Downloaded from http://journals.lww.com/jpgn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 04/16/2022

¶¶Hania Szajewska3, ##Dominique Turck, and ***Lawrence T. Weaver

*University of Central Lancashire, Preston, UK; †University of Milan, Milan, Italy; ‡University of Lund, Lund, Sweden;
§University of Rome and INRAN, Rome, Italy; ¬Hôpital Necker Enfants-Malades, Paris, France; ¶Umea University, Umea, Sweden;
#Ludwig-Maximilians-University, Munich, Germany; **Free University of Amsterdam, Amsterdam, The Netherlands; ††The Royal
Veterinary and Agricultural University, Frederiksberg, Denmark; ‡‡The General Infirmary, Leeds, UK; §§University of Liege,
Liege, Belgium; ¬¬Meyer Children’s Hospital of Haifa, Haifa, Israel; ¶¶The Medical University of Warsaw, Warsaw, Poland;
##University of Lille, Lille, France; and ***University of Glasgow, Glasgow, UK
1
Committee Chair; 2Past Committee Chair; 3Committee Secretary; 4Guest

ABSTRACT provision of long-chain polyunsaturated fatty acids. Infants


Survival of small premature infants has markedly improved discharged with a subnormal weight for postconceptional age
during the last few decades. These infants are discharged from are at increased risk of long-term growth failure, and the human
hospital care with body weight below the usual birth weight of milk they consume should be supplemented, for example, with a
healthy term infants. Early nutrition support of preterm infants human milk fortifier to provide an adequate nutrient supply. If
influences long-term health outcomes. Therefore, the ESPGHAN formula-fed, such infants should receive special postdischarge
Committee on Nutrition has reviewed available evidence on formula with high contents of protein, minerals and trace
feeding preterm infants after hospital discharge. Close monitor- elements as well as a long-chain polyunsaturated fatty acid
ing of growth during hospital stay and after discharge is supply, at least until a postconceptional age of 40 weeks, but
recommended to enable the provision of adequate nutrition possibly until about 52 weeks postconceptional age. Continued
support. Measurements of length and head circumference, in growth monitoring is required to adapt feeding choices to the
addition to weight, must be used to identify those preterm infants needs of individual infants and to avoid underfeeding or
with poor growth that may need additional nutrition support. overfeeding. JPGN 42:596 Y 603, 2006. Key Words: Infant
Infants with an appropriate weight for postconceptional age at nutritionVPrematureVVLBWVGrowth and develop-
discharge should be breast-fed when possible. When formula- mentVESPGHAN Committee on NutritionVNutrition assess-
fed, such infants should be fed regular infant formula with ment. Ó 2006 by Lippincott Williams & Wilkins

The survival of small premature infants has markedly In this article, the ESPGHAN Committee on Nutrition
improved during the last few decades because of refine- reviews the available evidence on feeding human milk
ments in obstetric and neonatal care. These changes have and milk formulae postdischarge in preterm infants,
raised further questions with regard to immediate and based on an electronic literature search (PubMed) of
long-term effects of nutritional care in premature infants. randomised controlled trials performed until September
In most parts of Europe, preterm infants tend to be 2004 and offers recommendations for practice and for
discharged from hospital care earlier than before for further research.
economic and other reasons, with body weights far below
typical birth weights of healthy term infants. The
GROWTH OF PREMATURE INFANTS
question has arisen whether such infants might require
IN THE HOSPITAL
special nutritional regimens or special discharge formulae.
Numerous studies underline the importance of early
Received February 28, 2006; accepted March 7, 2006.
Reprints: Jacques Rigo, MD, PhD, Department of Neonatology and
feeding of very low birth weight (VLBW; G1500 g) and
Nutrition, University of Liege, CHR Citadelle, Boulevard du XII de extremely low birth weight (ELBW; G1000 g) infants
Ligne 1, 4000 Liège, Belgium. (e-mail: j.rigo@ulg.ac.be). for their short- and long-term development (1Y 8). The

596

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
ESPGHAN COMMITTEE ON NUTRITION 597

incidence of intrauterine growth retardation (IUGR) is high T 2 SD), small for GA (SGA; birth weight, G10th
(10% Y 30%) in VLBW infants and postnatal weight and percentile or Gmean j 2 SD) or large for GA (LGA;
length gain often does not match intrauterine growth rates, birth weight, 990th percentile or 9mean + 2 SD). The
because postnatal parenteral and enteral nutrition usually comparison of weight, length and head circumference
do not achieve quantitative and qualitative nutrient provi- measurements provides information as to whether the
sion that would allow to approximate normal intrauterine anthropometric parameters are proportionate or dispro-
growth (9 Y 11). Increasing immaturity and morbidity with portionate. Even if AGA, SGA and LGA are defined by
decreasing gestational age (GA) are major factors limiting statistical criteria rather than clinical end points, they
nutrient intakes achieved during the first weeks of life. allow identification of subjects with growth abnormal-
Moreover, in practice, the recommended intakes are often ities (27).
not prescribed, and even the prescribed intakes are At the time of hospital discharge, the growth of
frequently not achieved, for example, because of concerns preterm infants can be classified into 4 different patterns:
of intolerance or the occurrence of adverse events (10 Y 12).
In many neonatal units throughout Europe, stable 1. infants with a birth weight and a body weight at
VLBW and ELBW infants are discharged from hospital discharge appropriate for postconceptional age (appro-
care at postconceptional ages of about 35 to 36 weeks priate growth);
and with body weights of about 1800 to 2100 g, 2. infants born AGA but with a discharge weight below the
respectively. For the feeding of such infants after dis- reference growth chart (postnatal growth restriction);
charge, human milk with or without added nutrient sup- 3. infants born SGA with a discharge weight still below
plements, standard infant formulae designed for healthy the reference growth chart (IUGR);
term infants, preterm formulae designed for feeding im- 4. infants born SGA with a discharge weight appro-
mature babies and special postdischarge formulae de- priate for postconceptional age (early postnatal
signed for low birth weight infants with increased catch-up growth).
density of nutrients are used. In making feeding choices,
considerations may include the aim of achieving early The relative proportion of these 4 main categories of
catch-up growth (an increased growth velocity after a preterm infants discharged from neonatal units differs
temporary growth retardation) as well as reducing long- according to GAs of the patients’ population and the
term growth deficits and risk of potential adverse effects nutritional policy during the first weeks of life. How-
in adulthood (13 Y 15). ever, during the early neonatal period full catch-up
Nutritional recommendations of various expert groups growth (pattern 4) is exceptional in clinical practice.
(16 Y 19) are based primarily on the studies of stable
preterm infants with few complications and aimed at
providing amounts of nutrients that may approximate
intrauterine growth. These recommended intakes often GROWTH OF PREMATURE INFANTS AFTER
do not cover added nutrient requirements that may be HOSPITAL DISCHARGE
needed to compensate for nutritional deficits accumu-
lated during the early postnatal period (3,11,20,21). Numerous studies have evaluated longitudinal growth
Because of these cumulative nutritional deficits during in VLBW infants (2,13,21,28 Y 30). Some degree of
the first weeks or even months of life, some postnatal catch-up growth is observed in most VLBW premature
growth retardation is regularly observed in VLBW and infants during the first months of life, although the rate
ELBW infants (3,9 Y 11,20,21). During hospital stay, the differs between studies partly related to sex and period
feeding of both human milk with an added human milk of growth restriction (prenatal or postnatal). Although
fortifier and of preterm formulae often generates a infants appropriate for corrected age at discharge usually
slower growth rate and different body composition than maintain normal growth subsequently (pattern 1), about
that achieved by the fetus in utero, with a relative low 80% of VLBW infants with postnatal growth retardation
lean body mass and a lower bone mineral content (pattern 2) and SGA populations (patterns 3 and 4) show
associated with osteopenia and a risk of fractures in catch-up growth by 2 to 3 years old (21). Catch-up
early life (22 Y 26). growth of VLBW preterm infants appears to be faster
Nutritional evaluation of a newborn begins at birth and more complete in girls than in boys, although data
with anthropometric assessment that provides informa- are controversial (11,28), and in growth restricted AGA
tion on intrauterine growth and continues up to (pattern 2) than in SGA (patterns 3 and 4) VLBW
discharge to evaluate postnatal growth. Anthropometric preterm infants (2,31). These studies suggest that
measurements most commonly used for assessment of subgroups of preterm infants with a higher risk for
nutritional status are weight, length and head circum- long-term growth restriction can be defined (patterns 2
ference plotted against GA. Plotting birth weight against and 3). In contrast to body weight, reduced bone
GA helps determine whether an infant is appropriate for mineralisation generally improves rapidly during the
GA (AGA; birth weight, 10th Y 90th percentile or mean first months of life. Between 6 and 12 months of age,

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
598 POSTDISCHARGE NUTRITION IN PRETERM INFANTS

bone mineralisation of infants born preterm reaches trial, enriched postdischarge formula was evaluated with
values, adjusted for anthropometric parameters, similar regard to growth (59 Y 62) and neurodevelopmental out-
to healthy term infants (32,33) and appears appropriate come in SGA term infants. Infants receiving enriched
for the skeletal and body size achieved. formula showed greater gains in length and head cir-
cumference than those fed term formula, not only
during the 9 month intervention period but also up to
POTENTIAL CONSEQUENCES OF PRENATAL 18 months, and the differences were larger in girls than
AND POSTNATAL NUTRITION in boys (62). Girls fed the enriched formula had sig-
nificantly lower scores in all subscales and in overall
Adequate nutrition during early infancy is essential developmental quotient compared with girls fed term
for the overall well-being of the child and can have a infant formula at 9 months, although no difference
major impact on long-term development. Preterm and between the groups was present at 18 months of age
term infants born SGA carry a high risk for continued (63). A breast-fed reference group was also evaluated as
growth deficits, neurodevelopmental abnormalities and nonrandomised control group and demonstrated a sig-
behavioural problems (34 Y 38). Increasing evidence nificant growth and neurodevelopmental advantage
suggests that either low birth weight or rapid postnatal compared with formula-fed infants (62,63). The ques-
weight gain or the combination of both may predispose tions raised here deserve careful evaluation in future
to adverse long-term effects, such as increased risk for studies with long-term follow-up that includes a detailed
hypertension, cardiovascular diseases, type 2 diabetes description of early growth patterns, dietary intakes and
and osteoporosis in adulthood (39 Y 47). Those preterm potential confounders.
infants who fail to achieve their growth potential during
the first weeks of postnatal life have a less favourable
outcome with respect to growth and neurodevelopment STUDIES ON THE USE OF NUTRIENT
(48 Y 50) and could therefore be subject to the same ENRICHED FORMULAE IN PRETERM
influences as infants born growth retarded at term INFANTS AFTER DISCHARGE
(51,52). The window for catch-up in growth-retarded
babies appears to be narrow. If catch-up growth does At the time of discharge, many VLBW and ELBW
not take place in early life, the chances that it will occur infants have cumulative deficits in the accretion of
later are limited (6,28,31,53). In human infants, this energy, protein, minerals and other nutrients, resulting
critical period may approximate to the first year with in higher nutrient requirements per kilogram of body
respect to development of head circumference and the weight than healthy AGA term infants. Therefore, the
first 3 years with respect to final height (54,55). potential benefits of diets providing a higher concen-
For VLBW infants, the consequences of postnatal tration of energy, protein and specific nutrients, such as
nutrition deficits for neurodevelopmental outcomes long-chain polyunsaturated fatty acids (LCPUFAs),
have been difficult to document because the neuro- minerals, electrolytes and trace elements, on growth,
development of preterm infants is a composite of bone mineralisation and developmental outcomes have
multiple factors including genetics, morbidity such as been investigated in several studies since 1992 (Tables 1
intraventricular hemorrhage, periventricular leukoma- and 2) (15,64 Y 72).
lacia and chronic lung disease, nutritional intake and All these studies had methodological limitations, for
other factors. In a recent study on VLBW infants, example, related to inclusion criteria, the selection of
postnatal growth pattern rather than SGA status was major end points or completeness of follow-up (Table 2).
significantly associated with neurodevelopmental out- The inclusion of premature infants with a birth weight
come at 2 years (49). A better neurodevelopmental
outcome was observed in AGA preterm infants main-
taining favourable growth velocity or in SGA demon-
strating early catch-up growth after the term equivalent TABLE 1. Macronutrient composition of standard,
(1,49). Similar results were observed among SGA postdischarge and preterm formula
preterm infants with catch-up growth from birth to term
Standard Postdischarge Preterm
as well as from term equivalent up to 8 months of age or formula formula formula
older (30,56 Y 58). On the basis of these studies,
neonatologists were encouraged to promote early Protein 1.4 Y 1.5 1.8 Y 1.9 2.2 Y 2.3
(g/100 mL)
Baggressive[ nutrition (ie, provision of high energy Energy 67 72 Y 74 80 Y 90
and nutrient intakes) to promote early catch-up growth (kcal/100 mL)
in VLBW infants (59 Y 61). However, potential unto- Calcium 35 Y 54 70 Y 80 100 Y 108
ward effects of prolonged use of formulae with high (mg/100 mL)
protein and mineral density during the first year of life Protein T 2.2 T 2.5 T 2.8
(g/100 kcal)
need to be considered. In a recent randomised controlled

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
ESPGHAN COMMITTEE ON NUTRITION 599

below 1650 to 1800 g or a GA below 34 to 37 weeks milk affected growth and development. Because of
selects a high-risk population for long-term growth differences in the way individual trials measured and
impairment, such as preterm infants with weight or presented outcomes, data synthesis was limited. Meta-
length below the reference curves at the time of analysis of data from 2 trials (62,66 Y 68) found a
discharge. Analysis of growth indices during an inter- statistically significant effect on crown-to-heel length at
mediate period (2 Y 18 months) as the end point may not 18 months postterm (weighted mean difference, 9.7 mm).
allow a clear evaluation of the potential for catch-up Meta-analysis of data from the same 2 trials (62,66 Y 68)
growth resulting from a change in the protein and that assessed neurodevelopment at 18 months postterm
mineral density of the formula. Because approximately did not reveal a statistically significant difference in
80% of preterm infants whose size is not appropriate for either Bayley Mental Development Index or Psychomo-
postconceptional age at the time of discharge show tor Development Index. At the end of their evaluation,
catch-up growth by the age of 2 to 3 years, the effects the authors conclude that the limited available data do
of dietary interventions should be evaluated at this age not provide strong evidence that feeding preterm or
in a high-risk population. However, such studies are low birth weight infants after hospital discharge with
difficult to perform. Considering a 20% risk of long- energy- and protein-enriched formula compared with
term growth restriction in a high-risk population and a standard term formula affects growth rates and devel-
target reduction of 50%, more than 200 infants would opment up to 18 months postterm.
need to be included into such a study. In addition, it is The prolonged use of postdischarge or preterm
also desirable to study neurodevelopment to inves- formulae (Table 2) instead of standard formulae in
tigate the potential effects of dietary interventions and preterm infants may considerably modify the intake of
early catch-up growth. Similarly, the relative contribu- nutrients such as protein, LCPUFA, minerals and trace
tion of a preexisting intrauterine growth restriction elements. The role of these individual substrates de-
needs to be considered as an additional confounding serves further attention. The supply of lipids providing
factor that could influence long-term growth and LCPUFAs may benefit visual acuity and cognitive
development. development in VLBW infants (74), but some studies
Although the available studies differed in design and also reported a significant advantage on weight and
reported conflicting results (Table 2), they provide length growth independently of the protein supply
valuable information. Comparing the various studies, (74,75). Similarly, an increased zinc intake could play
the use of preterm formula does not seem to induce an independent significant role on growth and motor
more positive effects than that observed with the use of development (76,77).
postdischarge formula. Compared with controls, for- Two of the cited studies (62,64) evaluated the feeding
mulae with an increased energy density tended to be of human milk after discharge, but human milk could
consumed at lower volume intakes and thus did not not be allocated by randomisation and depended on
increase total energy supply (15,64,66,72). In contrast, maternal choice. These studies suggest that the use of
increased protein density led to higher protein intake nutrient-rich formulae increases weight gain after dis-
influencing nitrogen metabolism and blood urea ni- charge without any benefit on developmental score as
trogen concentration (64,66). A positive effect on compared with feeding of human milk. This issue was
growth parameters was not observed in all studies recently evaluated by O’Connor et al. (78) who
(Table 2), but when seen occurred mainly during the compared growth and development in 4 groups of
early postdischarge period approximately until the first premature infants fed fortified human milk (980%),
weeks after the equivalent of term birth, particularly in preterm formula (980%) or a combination of human
VLBW infants and in boys (62,68,69). In studies milk and preterm formula (950% and G50%, respec-
evaluating body composition, an observed enhanced tively) from birth to corrected age at term followed by
weight gain did not seem to affect preferentially the lean human milk or postdischarge formula up to 12 months
body mass deposition and may be limited to preterm of corrected age. Human milk provision was based on
infants who are AGA at the time of discharge without maternal choice. This study showed that the growth of
any positive effect in those with growth restriction (70). VLBW infants was inversely related to human milk
In addition, evaluating the neurodevelopment outcomes at consumption up to corrected term, but despite the
18 months, Cooke et al. (68) reported that the use of slower early growth, human milkYfed low birth weight
preterm formula as postdischarge formula had no impact infants had development at least comparable to that of
on neurodevelopmental outcome. infants fed enriched formula from birth to 12 months of
Recently, a Cochrane review on the effect of energy- corrected age. It is important to stress that in this study
and protein-enriched formula for improving growth and an early catch-up growth for length (G9 months) and
development in preterm or low birth weight infants after head circumference (G4 months) was reported in the
hospital discharge has been published (73). Evaluating group fed predominantly with human milk for more
much the same studies, the authors found little evidence than 6 months. A positive relationship between duration
that feeding with energy- and protein-enriched formula of human milk feeding and the later Bayley Mental

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
600 POSTDISCHARGE NUTRITION IN PRETERM INFANTS

TABLE 2. Summary of the study design and main results observed on the prolonged use of enriched formula in preterm infants
after discharge

Author (year) Study Allocation Completeness of


and ref number design concealment* Blinding ITT† follow-up‡

Lucas et al RCT Unclear Reported as double-blinded; No Yes


(1992Y1993)(15,62) not stated who was blinded
Wheeler and Hall RCT Unclear Reported as double-blinded; No Yes
(1996)(64) not stated who was blinded
Brunton et al (1998)(65) RCT Unclear Reported as blinded; No Yes
not stated who was blinded
Cooke et al (2001)(66 Y 68) RCT Unclear Reported as blinded; No Yes
not stated who was blinded
Carver et al (2001)(69) RCT Unclear Reported as double-blind; No 6 mo, 74/125
not stated who was blinded (59%)
Lucas et al (2001)(62) RCT Adequate Yes (investigators, caregivers, Yes Yes
outcome assessors
and data analysis)
De Curtis et al (2002)(70) RCT Unclear Reported as blinded; No Yes
not stated who was blinded
HM, human milk; PDF, postdischarge formula; PTF, preterm formula; SF, standard formula; BDP, bronchopulmonary dysplasia; RTC,
randomised controlled trial; ITT, intention to treat; HC, head circumference; BMC , bone mineral content.
*Allocation concealment: adequate, randomisation method described that would not allow investigator/caregivers to know or influence
intervention group before eligible participant entered in the study; unclear, randomisation stated but no information on method used is available;
inadequate, method of randomisation used such as alternate medical record numbers or unsealed envelopes; any information in the study that
indicated that investigators or participants could influence intervention group.
†ITT, intention-to-treat analysis: yes, specifically reported by authors that intention-to-treat analysis was undertaken, and this was confirmed on
study assessment; yes, not stated but confirmed upon study assessment; no, not reported, and lack of intention-to-treat analysis was confirmed on
study assessment. Patients who were randomised were not included in the analysis because they did not receive the study intervention, withdrew
from the study or were not included because of protocol violation); no, stated but not confirmed upon study assessment.
‡Completeness of follow-up: trials with more than 80% follow-up of participants.

Index was observed, with a more marked benefit in in- feeding. Early nutritional support can reduce the degree
fants with chronic lung disease. In addition, human milk of growth failure and may limit the need for high
feeding was also associated with a reduction in serious nutrient supplies for preterm infants after discharge.
adverse events, specifically in the number of hospital- Infants with an appropriate weight for postconceptional
isation days after discharge. However, confounding age at discharge should be breast-fed when possible.
effects such as socioeconomic factors related to maternal When formula-fed, such infants may be given standard
choice to provide breast milk cannot be excluded. infant formula with provision of LCPUFA. Infants
The literature review revealed only limited informa- discharged with a subnormal weight for postconceptional
tion on the possible effects of the type and time of age and thus with an increased risk of long-term growth
introduction of complementary feeding in infants born failure, if fed on human milk should be supplemented to
preterm (63,76,78 Y 81). Although not the focus of this provide an adequate nutrient intake, for example with a
article, the committee recommends that these questions human milk fortifier. If such infants are fed formula, they
should be studied in further well designed trials. should receive special postdischarge formula with high
contents of protein, minerals and trace elements as well
as LCPUFA, at least until a postconceptional age of
CONCLUSIONS 40 weeks, but possibly until about 52 weeks. Further
research is required to determine the specific nutritional
Early nutritional support of preterm infants is impor- needs of infants born preterm with prenatal and postnatal
tant because it influences long-term health and develop- growth restriction during and particularly after hospital
ment. Growth monitoring up to discharge and thereafter stay and to evaluate the effects of nutritional interven-
must be based on regular measurements of weight, tions on long-term growth, neurodevelopment and other
length and head circumference to identify those preterm health outcomes. More research is also needed on the
infants with poor growth that may need additional effects of different types of complementary feeding and
nutritional support. Continued growth monitoring is their time of introduction. Such studies are expected to
required to adapt feeding choices to the needs of contribute to development of better nutritional support in
individual infants and to avoid underfeeding and over- these groups of infants.

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
ESPGHAN COMMITTEE ON NUTRITION 601

TABLE 2. (continued)
Duration of
Follow-up intervention
n Population (mo) Diet (mo) Main outcomes Effects on outcome measures
31 Preterm; G1850 g 9 SF, PDF 9 Weight, length, HC, SF G PDF
BMC SF = PDF
59 G35 wk; G1800 g 3 HM, SF, PTF 2 Weight, HM G SF, PTF
Length, HC HM, SF G PTF
60 VLBW G1500 g 3 SF, PTF 3 Weight, HC, ST = PTF
+ BPD length, BMC ST G PTF
Body composition BMC, LBM ST G PTF mainly for boys
113 e34 wk, e1750 g 18 SF, PTF 6 Weight, length, HC, BMC SF G PTF in boys
Developmental Score SF = PTF
125 G37 wk; G1800 g 12 SF, PDF 12 Weight, length, HC SF 9 PDF mainly for BW
G1250 g and boys
229 G37 wk; G1750 g 18 HM, SF, PDF 9 Weight, length, HM = FS G PDF mainly for boys
HC, Developmental Score FS = PDF
33 G34 wk; G1800 g 2 SF, PDF 2 Weight, length, HC SF = PDF
Weight gain composition SF = PDF

REFERENCES 11. Embleton NE, Pang N, Cooke RJ. Postnatal malnutrition and
growth retardation: an inevitable consequence of current recom-
1. Lucas A, Morley R, Cole TJ. Randomised trial of early diet in mendations in preterm infants? Pediatrics 2001;107:270 Y 3.
preterm babies and later intelligence quotient. BMJ 1998;317: 12. Rigo J. Protein, amino acid and other nitrogen compounds. In:
1481Y7. Tsang RCUR, Koletzko B, Zlotkin SH, eds. Nutrition of the
2. Hack M, Schluchter M, Cartar L, Rahman M, Cuttler L, Borawski Preterm Infant: Scientific Basis and Practice, 2nd ed. Cincinnati,
E. Growth of very low birth weight infants to age 20 years. OH: Digital Educational Publishing, 2005:45 Y 80.
Pediatrics 2003;112:e30 Y 8. 13. Fewtrell MS. Growth and nutrition after discharge. Semin
3. Clark RH, Thomas P, Peabody J. Extrauterine growth restriction Neonatol 2003;8:169 Y 76.
remains a serious problem in prematurely born neonates. 14. Griffin IJ. Postdischarge nutrition for high risk neonates. Clin
Pediatrics 2003;111:986 Y 90. Perinatol 2002;29:327 Y 44.
4. Singhal A, Wells J, Cole TJ, Fewtrell M, Lucas A. Programming 15. Lucas A, Bishop NJ, King FJ, Cole TJ. Randomised trial of nutri-
of lean body mass: a link between birth weight, obesity, and tion for preterm infants after discharge. Arch Dis Child 1992;
cardiovascular disease? Am J Clin Nutr 2003;77:726 Y 30. 67:324 Y 7.
5. Dobbing J. Nutrition and the developing brain. Lancet 1973;1:48. 16. American Academy of Pediatrics Committee on Nutrition. Nutri-
6. Dobbing J, Sands J. Quantitative growth and development of tional needs of low-birth weight infant. Pediatrics 1985;75:
human brain. Arch Dis Child 1973;48:757Y767. 976 Y 86.
7. Lemons JA, Bauer CR, Oh W, et al. Very low birth weight 17. Wharton B. Protein. In: Osney Mead O, ed. Nutrition and
outcomes of the National Institute of Child health and human Feeding of Preterm infants. Blackwell Scientific Publications,
development neonatal research network, January 1995 through 1987:37 Y 46.
December 1996. NICHD Neonatal Research Network. Pediatrics 18. Tsang RCLA, Uauy R, Zlotkin S, ed. Nutritional Needs of the
2001;107:E1. Preterm Infant: Scientific Basis and Practical Guidelines. Williams &
8. Mortaz M, Fewtrell MS, Cole TJ, Lucas A. Cholesterol metabo- Wilkins, Baltimore, ed. Baltimore, MD: Williams & Wilkins, 1993.
lism in 8 to 12-year-old children born preterm or at term. Acta 19. Nutrients needs and feeding of premature infants. Can Med Assoc
Paediatr 2003;92:525 Y 30. J 1995;152:1765 Y 85.
9. Bloom BT, Mulligan J, Arnold C, et al. Improving growth of very 20. Rigo J, Boboli H, Franckart G, Pieltain C, De Curtis M.
low birth weight infants in the first 28 days. Pediatrics 2003; Surveillance of the very-low birthweight infant: growth and nu-
112:8 Y 14. trition. Arch Pe´diatr 1998;5:449 Y 53.
10. Dusick AM, Poindexter BB, Ehrenkranz RA, Lemons JA. Growth 21. Rigo J, de Curtis M, Pieltain C. Nutritional assessment in preterm
failure in the preterm infant: can we catch up? Semin Perinatol infants with special reference to body composition. Semin
2003;27:302 Y 10. Neonatol 2001;6:383 Y 91.

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
602 POSTDISCHARGE NUTRITION IN PRETERM INFANTS

22. Pieltain C, De Curtis M, Gerard P, Rigo J. Weight gain com- 43. Hales CN, Ozanne SE. The dangerous road of catch-up growth.
position in preterm infants with dual energy X-ray absorptiometry. J Physiol 2003;547:5 Y 10.
Pediatr Res 2001;49:120 Y 4. 44. Metcalfe N, Monaghan P. Compensation for a bad start: grow
23. Lucas A, Gore SM, Cole TJ, Bamford MF, Dossetor JF, Barr I. now, pay later? Trends Ecol Evol 2001;16:254 Y 60.
Multicentre trial on feeding low birthweight infants: effects of diet 45. Hales CN, Ozanne SE. Reply to Comment on: Hales CN, Ozanne
on early growth. Arch Dis Child 1984;59:722 Y 30. SE (2003). For debate: fetal and early postnatal growth restriction
24. Rigo J, De Curtis M, Pieltain C, Picaud JC, Salle BL, Senterre J. lead to diabetes, the metabolic syndrome and renal failure.
Bone mineral metabolism in the micropremie. Clin Perinatol Diabetologia 2004;47:1336.
2000;27:147 Y 70. 46. Fagerberg B, Bondjers L, Nilsson P. Low birth weight in
25. Reichman B, Chessex P, Verellen G, et al. Dietary composition combination with catch-up growth predicts the occurrence of
and macronutrient storage in preterm infants. Pediatrics 1983; the metabolic syndrome in men at late middle age: the Athero-
72:322 Y 8. sclerosis and Insulin Resistance study. J Intern Med 2004;256:
26. Kashyap S, Ohira-Kist K, Abildskov K, et al. Effects of quality of 254 Y 9.
energy intake on growth and metabolic response of enterally fed 47. Cole T. Modeling postnatal exposures and their interactions with
low-birth-weight infants. Pediatr Res 2001;50:390 Y 7. birth size. J Nutr 2004;134:201 Y 4.
27. Lee PACS, Hokken-Koelega ACS, Czernichow P. International 48. Brandt I, Sticker EJ, Lentze MJ. Catch-up growth of head
small for gestational age advisory board consensus development circumference of very low birth weight, small for gestational age
conference statement: management of short children born small preterm infants and mental development to adulthood. J Pediatr
for gestational age. Pediatrics 2001;111:1253 Y 61. 2003;142:463 Y 8.
28. Guo SS, Roche AF, Chumlea WC, Casey PH, Moore WM. 49. Latal-Hajnal B, von Siebenthal K, Kovari H, Bucher HU, Largo
Growth in weight, recumbent length, and head circumference for RH. Postnatal growth in VLBW infants: significant association
preterm low-birthweight infants during the first three years of life with neurodevelopmental outcome. J Pediatr 2003;143:163 Y 70.
using gestation-adjusted ages. Early Hum Dev 1997;47:305 Y 25. 50. Luo ZC, Albertsson-Wikland K, Karlberg J. Length and body
29. Hack M, Merkatz IR, McGrath SK, Jones PK, Fanaroff AA. mass index at birth and target height influences on patterns of
Catch-up growth in very-low-birth-weight infants. Clinical corre- postnatal growth in children born small for gestational age.
lates. Am J Dis Child 1984;138:370 Y 5. Pediatrics 1998;102:E72.
30. Hack M, Weissman B, Borawski-Clark E. Catch-up growth during 51. Hofman P, Regan F, Harris M, Robinson E, Jackson W, Cutfield
childhood among very low-birth-weight children. Arch Pediatr W. The metabolic consequences of prematurity. Growth Horm
Adolesc Med 1996;150:1122 Y 9. IGF Res 2004;14:S136 Y 9.
31. Harding JE, McCowan LM. Perinatal predictors of growth 52. Hofman P, Regan F, Jackson W, et al. Premature birth and later
patterns to 18 months in children born small for gestational age. insulin resistance. N Engl J Med 2004;351:2179 Y 2186.
Early Hum Dev 2003;74:13 Y 26. 53. Gale CR, O’Callaghan FJ, Godfrey KM, Law CM, Martyn CN.
32. Fewtrell MS, Prentice A, Jones SC, et al. Bone mineralization and Critical periods of brain growth and cognitive function in children.
turnover in preterm infants at 8-12 years of age: the effect of early Brain 2004;127:321 Y 9.
diet. J Bone Miner Res 1999;14:810 Y 20. 54. Eriksson J. Commentary: early Fcatch-up_ growth is good for later
33. Salle BL, Putet G. Calcium, phosphorus and vitamin D require- health. Int J Epidemiol 2001;30:1330 Y 31.
ments in premature infants. In: Salle BL, ed. Nutrition of the Low 55. Victora CG, Barros FC, Horta BL, Martorell R. Short-term
Birth Weight. Nestle´ Nutrition Workshop Series, 1993:125 Y 35. benefits of catch-up growth for small-for-gestational-age infants.
34. Westwood M, Kramer MS, Munz D, Lovett JM, Watters GV. Int J Epidemiol 2001;30:1325 Y 30.
Growth and development of full-term nonasphyxiated small- 56. Lucas A, Fewtrell MS, Davies PS, Bishop NJ, Clough H, Cole TJ.
for-gestational-age newborns: follow-up through adolescence. Breastfeeding and catch-up growth in infants born small for
Pediatrics 1983;71:376 Y 82. gestational age. Acta Paediatr 1997;86:564 Y 9.
35. Grantham-McGregor SM. Small for gestational age, term babies, 57. Georgieff MK, Hoffman JS, Pereira GR, Bernbaum J, Hoffman-
in the first six years of life. Eur J Clin Nutr 1998;52(Suppl 1): Williamson M. Effect of neonatal caloric deprivation on head
S59 Y 64. growth and 1-year developmental status in preterm infants. J Pediatr
36. Richards M, Hardy R, Kuh D, Wadsworth ME. Birth weight and 1985;107:581 Y 7.
cognitive function in the British 1946 birth cohort: longitudinal 58. Lundgren EM, Cnattingius S, Jonsson B, Tuvemo T. Intellectual
population based study. BMJ 2001;322:199 Y 203. and psychological performance in males born small for gesta-
37. Strauss RS. Adult functional outcome of those born small for tional age with and without catch-up growth. Pediatr Res 2001;50:
gestational age: twenty-six-year follow-up of the 1970 British 91 Y 6.
Birth Cohort. JAMA 2000;283:625 Y 32. 59. Wilson DC, Cairns P, Halliday HL, Reid M, McClure G, Dodge
38. Dörner G. Perinatal hormone levels and brain organization. In: JA. Randomised controlled trial of an aggressive nutritional
Stumpf WEGL, ed. Anatomical Neuroendocrinology. Basel: regimen in sick very low birthweight infants. Arch Dis Child
Karger, 1975:142 Y 252. Fetal Neonatal Ed 1997;77:F4 Y 11.
39. Tu Y, West R, Ellison G, Gilthorpe M. Why evidence for the fetal 60. Ziegler EE, Thureen PJ, Carlson SJ. Aggressive nutrition of the
origins of adult disease might be a statistical artifact: the Breversal very low birthweight infant. Clin Perinatol 2002;29:225 Y 44.
paradox[ for the relation between birth weight and blood pressure 61. Thureen PJ, Hay WW Jr. Early aggressive nutrition in preterm
in later life. Am J Epidemiol 2005;161(1):27 Y 32. infants. Semin Neonatol 2001;6:403 Y 15.
40. Barker DJ, Eriksson JG, Forsen T, Osmond C. Fetal origins of 62. Lucas A, Fewtrell MS, Morley R, et al. Randomized trial of
adult disease: strength of effects and biological basis. Int J nutrient-enriched formula versus standard formula for post-
Epidemiol 2002;31:1235 Y 9. discharge preterm infants. Pediatrics 2001;108:703 Y 11.
41. Toschke A, Grote V, Koletzko B, von Kries R. Identifying 63. Morley R, Fewtrell MS, Abbott R, Stephenson T, MacFayden U,
children at high risk for overweight at school entry by weight gain Lucas A. Neurodevelopment in children born small for gestational
during the first 2 years. Arch Pediatr Adolesc Med 2004;158: age: a randomized trial of nutrient-enriched versus standard
449 Y 52. formula and comparison with a reference breastfed group.
42. Eriksson JG, Forsen T, Tuomilehto J, Jaddoe VW, Osmond C, Pediatrics 2004;113:515 Y 21.
Barker DJ. Effects of size at birth and childhood growth on the 64. Wheeler RE, Hall RT. Feeding of premature infant formula after
insulin resistance syndrome in elderly individuals. Diabetologia hospital discharge of infants weighing less than 1800 grams at
2002;45:342 Y 8. birth. J Perinatol 1996;16:111 Y 6.

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
ESPGHAN COMMITTEE ON NUTRITION 603

65. Brunton JA, Saigal S, Atkinson SA. Growth and body composition 74. O’Conner D, Hall R, Adamkin D, et al. Growth and development
in infants with bronchopulmonary dysplasia up to 3 months in preterm infants fed long-chain polyunsatured fatty acids: a
corrected age: a randomized trial of a high-energy nutrient-enriched prospective, randomized controlled trial. Pediatrics 2001;108:
formula fed after hospital discharge. J Pediatr 1998;133:340 Y5. 359 Y 71.
66. Cooke RJ, Griffin IJ, McCormick K, et al. Feeding preterm infants 75. Fewtrell MS, Abbott R, Kennedy K, et al. Randomized, double-
after hospital discharge: effect of dietary manipulation on nutrient blind trial of long chain polyunsaturated fatty acid supplementation
intake and growth. Pediatr Res 1998;43:355 Y 60. with fish oil and borage oil in preterm infants. J Pediatr 2004;
67. Cooke RJ, McCormick K, Griffin IJ, et al. Feeding preterm infants 144:471 Y 9.
after hospital discharge: effect of diet on body composition. 76. Friel J, Andrews W, Matthew J, et al. Zinc supplementation in
Pediatr Res 1999;46:461 Y 4. very-low-birth-weight infants. J Pediatr Gastroenterol Nutr 1993;
68. Cooke RJ, Embleton ND, Griffin IJ, Wells JC, McCormick KP. 17:97 Y 104.
Feeding preterm infants after hospital discharge: growth and 77. Castillo-Duran C, Weisstaub G. Zinc supplementation and growth
development at 18 months of age. Pediatr Res 2001;49:719 Y 22. of the fetus and low birth weight infant. J Nutr 2003;133:
69. Carver J, Wu P, Hall R, et al. Growth of preterm infants fed 1494S Y 7S.
nutrient-enriched or term formula after hospital discharge. 78. O’Connor D, Jacobs J, Hall RT, et al. Growth and development of
Pediatrics 2001;107:683 Y 9. premature infants fed predominantly human milk, predominantly
70. De Curtis M, Pieltain C, Rigo J. Body composition in preterm premature infant formula, or a combination of human milk and
infants fed standard term or enriched formula after hospital premature formula. J Pediatr Gastroenterol Nutr 2003;37:
discharge. Eur J Nutr 2002;41:177 Y 82. 437 Y 46.
71. Bishop NJ, King FJ, Lucas A. Increased bone mineral content of 79. Marriott L, Foote K, Bishop NJ, Kimber A, Morgan J. Weaning
preterm infants fed with a nutrient enriched formula after preterm infants: a randomised controlled trial. Arch Dis Child
discharge from hospital. Arch Dis Child 1993;68:573 Y 8. Fetal Neonatal Ed 2003;88:F302 Y 07.
72. Lucas A, Kirkwood CD, Bishop NJ. Postdischarge formula con- 80. Fewtrell M, Lucas A, Morgan J. Factors associated with weaning
sumption in infants born preterm. Arch Dis Child 1992;67:691 Y 2. in full term and preterm infants. Arch Dis Child Fetal Neonatal Ed
73. Henderson GFT, McGuire W. Caloric and protein-enriched formula 2003;88:F296 Y 301.
versus standard term formula for improving growth and development 81. Norris F, Larkin M, Williams C, Hampton S, Morgan J. Factors
in preterm or low birth weight infants following hospital discharge. affecting the introduction of complementary foods in the preterm
Cochrane Database Syst Rev 2005;18:CD00 4696. infants. Eur J Clin Nutr 2002;56:448 Y 54.

J Pediatr Gastroenterol Nutr, Vol. 42, No. 5, May 2006

Copyr ight © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Post-discharge Nutrition in Preterm
Infants

Richard J. Cooke

Contents Abstract
The fundamental principle underlying nutri-
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
tional support is that intake meets needs,
Postnatal Malnutrition and Growth Failure in thereby ensuring the best outcome, in the case
Preterm VLBWI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
of the preterm infant, optimal growth, and
Importance of Postnatal Malnutrition and development. Achieving this goal is problem-
Growth Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
atic and most, if not all, very-low-birth-weight
Catch-Up Growth, Insulin Resistance, and infants (VLBWI) are undernourished and
Visceral Obesity in Preterm Infants . . . . . . . . . . . . 7 undergrown when first discharged home. This
Post-discharge Nutritional Support in Preterm has important implications for nutritional care
Infant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 of these high-risk infants after hospital
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
discharge.
Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Breastfeeding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Introduction
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
The fundamental principle underlying nutritional
support is that intake meets needs, thereby ensur-
ing the best outcome, in the case of the preterm
infant, optimal growth, and development. Achiev-
ing this goal is problematic and most, if not all,
\very-low-birth-weight infants (VLBWI) are
undernourished and undergrown when first
discharged home. This has important implications
for nutritional care of these high-risk infants after
hospital discharge.
In this chapter the following issues will be
addressed:

• Postnatal malnutrition and growth failure


R.J. Cooke (*) (PGF) between birth and hospital discharge in
Department of Pediatrics, University of Tennessee Health
preterm infants
Science Center, Memphis, TN, USA
e-mail: richardjcooke@mac.com

# Springer International Publishing Switzerland 2016 1


G. Buonocore et al. (eds.), Neonatology,
DOI 10.1007/978-3-319-18159-2_189-1
2 R.J. Cooke

Fig. 1 Growth curve for a Postnatal Growth In A 24 w 690 AGA Infant*


24-week AGA preterm 3500
infant who regains birth Fetus 3058 g
weight by 7, 14, and 21 days 3000
and then grows at a rate of 1 w – 2792 g
17 g/kg/day 2500 2 w – 2527 g

Weight (g)
3 w – 2285 g
2000

1500

1000

500

0
23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38
Gestational Age w

• Regaining birth weight at 1,2 and 3 w and growing at 17 g/kg/d

• Catch-up growth, insulin resistance, and vis- intercurrent “illness” not only reduces intake but
ceral obesity in preterm infants also may alter nutrient assimilation and increase
• Nutritional care of VLBWI after hospital requirements (Mehta et al. 2013). However, other
discharge factors underpin the development of growth fail-
ure in these infants.
Current recommendations are that the rate of
Postnatal Malnutrition and Growth weight gain parallels that of the fetus of the same
Failure in Preterm VLBWI gestational age once birth weight has been
regained (AAPCON 1998; Klein 2002). Yet, a
Several studies have examined postnatal growth 24-week appropriately grown (AGA) infant
in preterm infants during initial hospital stay and weighing ~690 g at birth who regains birth weight
noted that most preterm, if not all very-low-birth- by 2 weeks of age and grows at the recommended
weight infants (VLBWI), are growth retarded at rate, i.e., 17 g/kg/day, will weigh 2,527 g, ~531 g
hospital discharge (Wilson et al. 1997; Carlson less than the fetus, and be growth retarded at
and Ziegler 1998; Embleton et al. 2001; Clark 37 weeks (Fig. 1). If the same infant takes
et al. 2003; Olsen et al. 2002; Ehrenkranz 3 weeks to regain birth weight and grows at the
et al. 1999; Cooke et al. 2004). In studies where same rate, then she/he will weigh 2,285 g, 773 g
nutritional intake was measured, recommended less than the fetus at 37 weeks.
dietary intakes (RDIs) took time to establish, and Up to 40 % of infants born prematurely are
infants accrued a significant nutrient deficit that undergrown or small for gestational age (SGA)
was directly related to the growth deficit (Wilson at birth (Larsen et al. 1990). A 24-week 550 g
et al. 1997; Carlson and Ziegler 1998; Embleton SGA infant who regains birth weight at 2 weeks
et al. 2001; Clark et al. 2003; Olsen et al. 2002). and then gains at the recommended rate of 17 g/
In studies where intake was not measured, poor kg/day will weigh 1,727 g at 37 weeks, 800 g less
growth was related to non-nutritional factors such than the 690 AGA infant at the same gestational
as “illness” (Ehrenkranz et al. 1999; Cooke age and ~1,300 g less than the fetus at the same
et al. 2004). In the recent study of Griffin gestational age (Fig. 2). Recommendations related
et al. (2016), necrotizing enterocolitis was noted to suboptimal weight will systematically underes-
to have a significant negative on growth (Griffin timate needs and growth potential in these high-
et al. 2016). This is not surprising because risk infants.
Post-discharge Nutrition in Preterm Infants 3

Fig. 2 Growth curve for a Postnatal Growth in 24 w AGA and SGA Infant*
24-week AGA and SGA 3000
preterm infants who regain
birth weight by 14 and then AGA – 2527 g
2500
grow at a rate of 17 g/kg/day
SGA – 1727 g
2000

Weight (g)
1500

1000

500

0
23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38
Gestational Age w
• Regaining birth weight at 2 w and growing at 17 g/kg/d

Table 1 Protein and energy needs in preterm infants


Weight g 500–700 700–900 900–1,200 1,200–1,500
Protein g/kg/day 4.0 4.0 4.0 3.9
Energy kcal/kg/day 105 108 119 127
P:E g/100 kcal 3.8 3.7 3.4 3.1

At the same time, current recommendations infants (n = 149) at hospital discharge (Cooke
assume that nutritional requirements are consis- and Griffin 2009). Weight, length, and head cir-
tent throughout gestation for all preterm VLBWI cumference were measured using standard meth-
(AAPCON 1998; Klein 2002). However, Ziegler odology (Cooke et al. 1998). Body composition
et al. have noted that protein and energy require- was measured using dual-emission x-ray absorp-
ments change with advancing gestation (Ziegler tiometry (Cooke et al. 1999).
et al. 2002; Table 1). In effect, a formula The post-menstrual age at discharge was
containing a protein-energy ratio of 3.0 g/ 37  1.2 weeks. However, Z-score for head cir-
100 kcal, as is currently fed to preterm infants, cumference ( 0.1  0.6) was significantly
does not meet the protein needs of the infants greater than that for weight ( 1.4  0.6) which
weighing 1,500 g at birth, i.e., the VLBWI. in turn was greater than for crown-heel length
Recommended dietary intakes are based on ( 1.9  0.6; p < 0.0001) (Cooke and Griffin
needs for maintenance and normal growth 2009). Body composition results are compared
(AAPCON 1998; Klein 2002), and no allowance to the reference infant at the same weight and
is made for recovery or “catch-up” growth (Heird same gestation in Table 2.
2001). In the study of Embleton et al., the accrued Global fat-free mass was less in study infants
protein deficit at hospital discharge varied from than the reference infant at the same weight
15 to 25 g/kg (Embleton et al. 2001). An addi- (2,062 < 2,252 g; p < 0.0001) or gestation
tional protein intake of 0.5–1.0 g/kg/day would (2,062 < 2,667 g; p < 0.0001). Global fat mass
have been required to recoup this deficit before was greater in study infants than the reference
hospital discharge, further compounding the infant at the same weight (307 > 198 g,
problem. 13 > 8 %) or gestation (307 > 273 g;
To further examine this issue, body size and 13 > 9 %; p < 0.0001) (Cooke and Griffin
body composition were measured in preterm 2009).
4 R.J. Cooke

Table 2 Body size and composition of study infants com- dietary protein and energy intakes in these
pared to the reference infant at the same weight and same infants.
gestation (Cooke and Griffin 2009)
Reference Reference
Study at at
infantsa 35 weeksa 37 weeksa Importance of Postnatal Malnutrition
Body 2,369  305 2,450 2,940 and Growth Failure
weight
g The importance of malnutrition cannot be
Fat-free 2,062  277a 2,252a 2,667a underestimated. Nutrients play a critical role on
mass g the promotion of normal health and prevention of
Fat-free 87  3.4 91.9 90.7
disease (Shils et al. 2006). It is not surprising,
mass %
Fat 307  130b 198b 273b
therefore, that it can be directly related to alter-
mass g ations in organ structure and function which, in
Fat 13  3.4b 8.1b 9.3b turn, are paralleled by an increased morbidity and
mass % mortality in adults and children (Shils et al. 2006).
a
Study infants < reference at 35 week or 37 weeks However, the effects of malnutrition are greater
( p < 0.0001) during infancy than later in life. There are several
b
Study infants > reference at 35 week or 37 weeks
( p < 0.001)
reasons for this.
Requirements are a function of growth rate; the
greater the rate, the greater the requirement and the
A reduced body size that is paralleled by more likely that deficiency will occur. Growth rates
reduced linear growth and a reduced fat-free are greater during infancy than later in life; i.e., the
mass indicate that dietary protein needs were term infant will double birth weight by 4–5 m,
not met in these infants before hospital dis- triple birth weight by 12 m, and approximately
charge, as has been previously suggested quadruple it by 24 m.
(Ziegler et al. 2002). A reduced fat-free mass Growth is also thought to be “preprogrammed”
coupled with an increased global and central fat to occur at a certain time or “critical” epoch which
mass echoes concerns about visceral obesity and if missed may not be recoverable (Dobbing 1981).
the development of insulin resistance in these Thus, even short periods of nutritional deprivation
high-risk infants (Uthaya et al. 2005; Yeung may not only affect somatic but also brain growth
2006). and development (Dobbing 1981), the area of the
Although many factors contribute to the devel- brain that is “programmed” to grow fastest being
opment of PGF and the degree of which will vary, the most affected (Bedi 1987).
depending upon the level of maturity/size of the In term infants, malnutrition during infancy is
infant (Embleton et al. 2001), it is clear that: associated with permanent alterations in brain
growth and function. Brain size is reduced (Galler
1. It is not preventable given current recommen- et al. 1996; Stoch et al. 1982; Dobbing 1987;
dations and the way infants are fed; i.e., target Winick and Rosso 1969a, b), the brain cortex is
growth rates related to a suboptimal body thinner (Dobbing and Sands 1971), neuronal
ensure that most, if not all, infants will be numbers are decreased (Dobbing et al. 1971),
moderately/severely growth retarded at hospi- myelination is reduced (Krigman and Hogan
tal discharge. 1976), and dendritic morphology is altered
2. Close attention must be paid to nutritional (Benitez-Bribiesca et al. 1999; Cordero
support and growth of these infants after et al. 1993), all of which can be related to poorer
hospital discharge. A reduced body size neurodevelopmental outcome (Pryor et al. 1995;
that is paralleled by a reduction in fat-free Ounsted et al. 1988; Gross et al. 1983; Hack and
mass but a relative increase in fat mass indi- Breslau 1986; Hack et al. 1991; Kitchen
cates that particular attention be paid to
Post-discharge Nutrition in Preterm Infants 5

et al. 1992a; Cooke and Foulder-Hughes 2003; 2001; Dinerstein et al. 2006; Ibrahim et al. 2004;
Stathis et al. 1999; Peterson et al. 2006). Donovan et al. 2006).
These concerns are greater in preterm infants. While a clear relationship exists between
Growth rates are greater. A preterm infant is “catch-up” growth and development in preterm
“programmed” to quadruple brain weight infants, the time frame within which it needs to
between 24 and 40 weeks gestation or 16 weeks occur is not well delineated. In most studies,
(Alexander et al. 1996), almost six times faster infants who “catch up” or “catch back” by
than the term infant. Preterm infants are also more 6–9 m corrected age have better neurodeve-
vulnerable to the effects of perinatal ischemia and lopmental outcome (Morley 1999; Scott and
inflammation, therefore the development of Usher 1966; Hack et al. 1982; Hack and Fanaroff
periventricular intraventricular hemorrhage and 1984; Latal-Hajnal et al. 2003). This is illustrated
periventricular leukomalacia (Volpe 2008). in the study of Latal-Hajnal et al. (2003).
Compared to term, preterm infants are also In this study, infants were stratified at birth and
more likely to be fetally and/or postnatally mal- into those who were appropriate size for gesta-
nourished. Up to 40 % of preterm infants are tional age (AGA weight 10th percentile) and
growth retarded or small for gestational age SGA (weight <10th percentile). This process
(SGA) at birth (Greisen 1992). At the same time, was repeated at 2 years of age. Four groups
up to 100 % of preterm VLBWI undergo signifi- emerged: (a) AGA at birth and 2 years
cant postnatal growth failure and are SGA at hos- (AGA-AGA), (b) AGA at birth and SGA at
pital discharge (Ehrenkranz et al. 1999). 2 years (AGA-SGA), (c) SGA at birth and AGA
Poor fetal growth is paralleled by reduced at 2 years (SGA-AGA), and (d) SGA at birth and
organ growth as well as altered structure and 2 years (SGA-SGA).
function (McCance and Widdowson 1974) but No differences were noted in development
not all organ systems are affected equally. This is between the AGA-AGA and SGA-SGA infants.
well illustrated in the study of Myers After adjusting for co-variables, infants who
et al. (1971). In this study, 30 % reduction in “caught up,” i.e., SGA-AGA, had greater Bayley
body weight in SGA monkeys was associated PDIs than those who did not (SGA-SGA). Infants
with an 8 % reduction in brain weight but who “faltered” (AGA-SGA) had lower PDIs and
35 % reduction in lung, liver, pancreatic, and MDIs than those who continued to thrive
spleen weights when compared to their AGA (AGA-AGA). These authors concluded that the
counterparts (Myers et al. 1971). Thus, the course of postnatal growth primarily determined
brain is “spared” at the expense of other organs later development.
which, e.g., through the development of chronic To more closely examine this issue,
lung disease, sepsis, etc., may amplify undernu- Dharmaraj et al. prospectively followed growth
trition by reducing intake and/or increasing between birth and18 months corrected age (mca)
requirements. and developmental outcome at 18 mca
In the late 1980s and early 1990s, studies indi- (Dharmaraj et al. 2005). It was hypothesized
cated that poor growth between birth-hospital dis- that the greater the degree of early growth failure
charge was associated with poorer (birth to 28 days), the poorer the development at
neurodevelopment (Morley 1999; Ehrenkranz 18 mca. Infants were stratified at 28 days into
et al. 2006) and that better growth, as achieved those with mild (fall in weight Z-score % 1.0;
by feeding a nutrient-enriched formula, was asso- MGF) and severe (fall in weight Z-score >1.0;
ciated with better developmental outcomes (Lucas SGF) growth failure. This process was repeated
et al. 1998, 1990). More recently, early parenteral at 18 mca.
nutrition coupled with the early introduction and The characteristics of the study infants are
advancement of enteral feeds has been associated presented in Table 3. No differences were noted
with better growth but many infants continue to be with the exception that gestational age was less
SGA at hospital discharge (Evans and Thureen (SGR-SGR < MGR-MGR, MGR-SGR,
6 R.J. Cooke

Table 3 Characteristics of study infants (Dharmaraj et al. 2005)


Group (N) MGR-MGR (50) MGR-SGR (18) SGR-SGR (16) SGR-MGR (24)
Birth weight (g) 1,320  339 1,348  387 1,312  559 1,271  408
Gestation (w) 30  1.6a 30  1.8a 28  3.0a 29  2.3
BPD 13 (26 %) 3 (17 %) 6 (38 %) 10 (42 %)
Abnormal CUS 10 (20 %) 5 (28 %) 3 (19 %) 6 (25 %)
PVL 3 (6 %) 2 (11 %) 1 (6 %) 3 (13 %)
Cerebral palsy 4 (8 %) 3 (17 %) 5 (30 %)b 2 (8 %)
a
SGR-SGR < MGR-MGR, MGR-SGR, p < 0.05
b
SGR-SGR > MGR-MGR, SGR-MGR, p < 0.10

Fig. 3 Growth, i.e., change Postnatal Growth in Study Infants*


in weight Z-score between 1.0
Change In Weight Z-score From Birth

birth and 28 days, term and


one through 18 month 0.5 MGF-MGF
corrected age in infants in
the study of Dharmaraj et al. 0
SGF-MGF
−0.5

−1.0
−1.5 MGF-SGF

−2.0 SGF-SGF

Birth 28d Disc EDD +1m +2m +3m +6m + 9m + 12m +18m

* Dharmaraj et al60

Table 4 MDI and PDI in study infants (Dharmaraj et al. 2005)


Group (N) MGR-MGR MGR-SGR SGR-MGR SGR-SGR
MDIa 93  18 91  18 95  1 77  19
PDIa 90  17a 83  19 87  19b 77  22a
MDIb 96  15 93  20 98  16 88  11
PDIb 94  13 89  15 91  14 89  16
MDI ~ MGR-MGR, SGR-MGR > SGR-SGR. p < 0.05
a
PDI ~ MGR-MGR ( p < 0.05), SGR-MGR ( p < 0.10) > SGR-SGR
b
Infants with cerebral palsy excluded. MDI ~ MGR-MGR ( p < 0.05), SGR-MGR ( p < 0.10) > SGR-SGR

p < 0.05) and the frequency of cerebral palsy 18 mca (MGF-MGF) and SGF at 28 days but
tended to be greater (SGR-SGR > MGR-MGR, mild growth failure (SGF-MGF) at 18 mca. How-
SGR-MGR, p < 0.10) in infants with severe ever, growth faltered in the other groups, that is,
growth failure at 28 days and 18 m corrected age. MGF-SGF and SGF-SGF.
Growth of these infants is presented in Fig. 3. The developmental outcome data are
Between birth and 28 days, Z-scores for weight presented in Table 4. MDI and PDI were less
decreased and then recovered to some degree in in SGF-SGF than MGF-MGF infants,
all groups. After 28 days, growth continued to supporting the primary hypothesis that poor
improve in infants with MGF at 28 days and growth between birth and 28 days is a marker
Post-discharge Nutrition in Preterm Infants 7

for poor development. MDI and PDI were also programmed within a specific time frame or crit-
greater in infants who recovered after 28 days ical epoch which if missed may not be recoverable
(SGF-MGF) than those who did not (McCance and Widdowson 1974; Widdowson
(SGF-SGF), indicating that “catch-up” growth and McCance 1975). It also depends upon the
between 28 days and 18 mca is paralleled by severity and duration of the insult; the more severe
markedly improved development. and prolonged the insult, the greater the accrued
Collectively, these data support the conclu- nutritional deficit. Intake, therefore, must not only
sions of Latal-Hajnal et al.; i.e., it is the course replace the deficit but also meet needs for mainte-
of postnatal growth that determines later develop- nance and normal growth.
mental outcome in preterm infants. The data of With acute illness, infants develop a
Dharmaraj et al. also indicate that a “critical win- hypermetabolic state, and there is an acute deple-
dow of opportunity” exists between 28 days and tion of body protein to meet energy needs (Lowry
1–2 mca during which “recovery” or “catch-up” and Perez 2006). With chronic protein-energy
growth, to birth-weight percentile, is paralleled by malnutrition during infancy, visceral tissue is con-
better neurodevelopmental outcome. If infants are served at the expense of protein and fat (Ashworth
to “recover,” this is the time frame within which to and Millward 1986). Nutritional rehabilitation in
do it. these infants with high-energy intakes largely
results in increased weight gain (MacLean and
Graham 1980) and body fat (MacLean and Gra-
Catch-Up Growth, Insulin Resistance, ham 1980; Jackson 1990); i.e., the nature of the
and Visceral Obesity in Preterm Infants gain is dependent upon the compositional nature
of intake.
Despite the prevalence/severity of PGF that is At hospital discharge, all preterm VLBWI
paralleled by an increased morbidity/mortality have accrued a significant protein and energy
before/after hospital discharge, concern has been deficit (Embleton et al. 2001) and are lighter and
expressed about measures taken to improve shorter and have a reduced lean but greater fat
growth after hospital discharge. Concern largely mass than the reference infant at the same body
relates to “catch-up” growth and the subsequent weight or gestation (Uthaya et al. 2005). Thereaf-
development of insulin resistance (Singhal ter, a diet that is relatively high in energy will
et al. 2001, 2003) and metabolic syndrome X promote “catch-up” that is paralleled by increased
(Ong and Loos 2006) leading to the idea that fat deposition. A diet that better meets protein
“bigger might not be better,” even in preterm needs will be paralleled by increased lean mass
VLBWI (Singhal et al. 2003). accretion.
However, a certain amount of confusion exists Energy density is the prime determinant of
when the terms “rapid” and “catch-up” growth are volume of intake in infants (Fomon 1993a;
used; i.e., they are sometimes used interchange- Cooke et al. 1998). Yet at borderline low protein
ably (Ong and Loos 2006) but are not the same. intakes, volume/energy intake is increased to
“Rapid” growth is “an abnormal growth pattern compensate and is associated with increased fat
that moves upward beyond the original growth accretion (Fomon 1993b). The energy cost of lean
percentile on a distance growth chart” mass accretion is ~13.4 kcal/g and that of fat
(Hermanussen 2013a). “Catch-up” is a physio- accretion is 10.6 kcal/g (Roberts and Young
logic condition of temporary overgrowth that 1988). At equivalent energy intakes, therefore,
occurs after illness and/or starvation, and in the the greater the rate of lean mass accretion, the
process, the original growth percentile is lesser the tendency to fat accretion. A little more
reestablished (Hermanussen 2013b). rather than less protein, therefore, might be con-
The extent to which “catch-up” occurs depends sidered appropriate.
on many factors, including timing of the insult. The relative roles of genetic, intrauterine, and
Growth/development is thought to be environmental factors in the pathogenesis of
8 R.J. Cooke

Postnatal Growth in Study Infants* formula (protein content 1.8 g/100 kcal, discharge
0 6 m) (B), a preterm formula to term and then a
-.5 term formula to 6 m (C), or unfortified breast milk
(D) (Cooke et al. 1998, 1999). The results are
-.1
presented in Fig. 4.
-1.5 “Rapid” growth was not noted in either treat-
-2 * ment group; i.e., Z-scores for weight did not
* * exceed birth percentile. Between discharge and
-2.5
term, “catch-up” growth in weight and length
-3
Weight Z-score occurred in the formula but not the breastfed
-3.5 infants. Thereafter, “catch-up” occurred in all
0 groups and was faster and more complete in
-.5 infants fed the preterm formula. Infants with the
-1 poorest growth were those fed the preterm for-
-1.5 mula between discharge and term and the term
-2
* * * formula thereafter (Fig. 4).
-2.5 * * More complete “catch-up” growth was
-3 * paralleled by an increase in fat-free mass and
-3.5 total fat mass but not % fat mass (Table 5). No
-4 Length Z-score differences were detected in central fat mass
-4.5 between the groups, but leg fat mass was greater
Group Preterm Term Cross-Over Breast in infants fed the preterm formula (Table 6). In
* Cooke et al 16,
effect, “catch-up” growth was paralleled by an
Discharge Term 12 w 6m 12 m increase in linear growth, lean, and fat mass accre-
tion, the latter reflecting increased peripheral not
Fig. 4 Growth between discharge and 12 months in central fat mass accretion.
infants fed a preterm infant formula, a term formula, or a
preterm formula (discharge-term)/term formula (after term,
Collectively, these data:
crossover) or breastfed
(a) Do not support concerns about altered adipos-
ity in preterm infants fed a nutrient-enriched
insulin resistance are unclear, but diet also plays a formula after hospital discharge
key role (Cornier et al. 2008). A high energy, i.e., (b) Indicate that rate and composition of growth
glycemic or fat content, increases insulin needs, are dependent upon the composition of the
fat synthesis, storage, etc. (Bremer et al. 2012). diet
Whether a “normal” infant diet, i.e., that appro- (c) Do not support the recommendation that a
priate for a term infant, is likely to do so in preterm formula be fed to term and
VLBWI with PGF is unclear. What is clear is discontinued thereafter (Aggett et al. 2006)
that growth and nutritional need to be closely
monitored during the first 1–2 m after hospital
discharge, with particular attention paid to ade- Post-discharge Nutritional Support
quacy of protein intake and protein-energy status, in Preterm Infant
i.e., monitoring linear growth and biochemical
measures of status if needed, i.e., blood urea nitro- Overview
gen, prealbumin, and albumin levels.
To more closely examine this issue, body size Before hospital discharge, it is important that
and composition were measured in preterm the method of feeding and growth patterns are
infants fed either a preterm formula (protein con- well established. Nutritional status of each
tent = 2.7 g/100 kcal, discharge 6 m) (A), a term infant should be closely evaluated, with the
Post-discharge Nutrition in Preterm Infants 9

Table 5 Body composition of preterm infants fed either preterm formula, term formula, preterm formula (to term)/term
formula (after term, crossover), or breastfed (Cooke et al. 1999)
Term 12 weeks 6 months 12 months
Group (n = 148) (n = 141) (n = 145) (n = 138)
Fat-free Preterm 2,745  445 4,270  452 5,208  638 6,872  806 A>
mass (g) (A)
Term (B) 2,393  276 4,022  411 5,139  515 6,592  738 B
( p < 0.05)
Crossover 2,507  244 3,948  431 4,978  541 6,399  881 C
(C) ( p < 0.001)
Breastfed 2,171  296 3,762  1,051 5,063  568 6,451  746 D
(D) ( p < 0.005)
Fat mass (g) Preterm 570  256 1,455  461 2,033  686 2,332  679 A>
(A)
Term (B) 511  222 1,367  419 1,940  586 2,058  477 B
( p < 0.05)
Crossover 566  204 1,188  366 1,815  632 2,077  623 C
(C) ( p < 0.005)
Breastfed 331  128 1,365  527 1,934  658 2,153  645
(D)
Fat mass % Preterm 17  5.5 25  4.7 28  5.9 25  5.3
(A)
Term (B) 17  6.0 25  5.5 27  5.1 24  4.4
Crossover 18  4.7 23  4.1 26  5.8 24  4.5
(C)
Breastfed 13  3.2 25  8.2 27  6.8 25  4.8
(D)

Table 6 Regional body composition of preterm infants fed either preterm formula, term formula, preterm formula
(to term)/term formula (after term, crossover), or breastfed
G Group Term 12 weeks 6 months 12 months
Trunk Preterm (A) 172  65 532  168 651  218 699  249
Term (B) 182  71 485  154 635  170 613  160
Crossover (C) 186  67 394  136 579  221 573  162
Breastfed (D) 147  54 503  183 688  229 657  230
Legs Preterm (A) 159  58 500  155 736  270 975  354 A>
Term (B) 133  73 451  168 652  300 830  271 B ( p < 0.01)
Crossover (C) 129  75 458  133 602  247 822  409 C ( p < 0.001)
Breastfed (D) 98  58 454  183 654  289 897  254 D ( p < 0.10)

information communicated to the primary care 2–3 m of life (Rawlings et al. 1999). They are also
physician and arrangements for follow-up more susceptible to even marginal levels of intake
clearly defined. and benefit more when fed nutrient-enriched for-
Intake after discharge must not only meet mulas before (Lucas et al. 1989a, 1990, 1994;
needs for maintenance and normal growth but Morley and Lucas 1997) and after hospital dis-
also “catch-up” growth. It is assumed that needs charge (Cooke et al. 1999; Lucas et al. 2001; Car-
for normal growth are similar in preterm boys and ver et al. 2001; Agosti et al. 2003).
girls. Yet, preterm boys grow faster in utero and It is also assumed that needs for “recovery” are
accrete more lean mass than girls during the first similar for all preterm infants. This is not the case.
10 R.J. Cooke

Up to 40 % of infants prematurely are small for consuming, is also important to ensure that the
gestational age (SGA) at birth. Needs are likely to composition of growth is also appropriate
differ between the AGA and SGA infants. At the (Singhal et al. 2003).
same time, the severity of PGF not only varies
widely between infants discharged from a given
NICU (Embleton et al. 2001); i.e., the smaller and Studies
more immature the infant, the greater the accrued
nutrient deficit, but also between NICUs (Olsen Formula Feeding
et al. 2002; Cooke et al. 2004; Griffin et al. 2016;
Horbar et al. 2015). In effect, needs for “recovery” Growth
are also highly variable. Many studies have examined post-discharge
Collectively, these data emphasize the growth in preterm infants (Kitchen et al. 1992a, b;
importance of individualized assessment before Ernst et al. 1990; Casey et al. 1990; Fitzhardinge
discharge and, irrespective of what an infant is and Inwood 1989; Fenton et al. 1990; Ross
fed, close nutritional monitoring after dis- et al. 1990). Although some “catch-up” growth
charge. Not surprisingly, many questions has been observed, preterm infants do not grow as
remain unanswered. How often should growth well as their term counterparts and are smaller at
be monitored? 3 years (Casey et al. 1991), 8 years (Hack
In the follow-up study of Dharmaraj et al. 1993), and adulthood (Hack et al. 2003).
et al. (2005), growth velocity was greatest There are several possible reasons for this.
between discharge and term, continuing to Current in-hospital feeding practices ensure
1–2 mca, a time frame within which brain growth that most, if not all, VLBWI are undernourished
velocity is also greatest and likely to be function- and growth retarded at initial hospital discharge
ally significant (Fig. 3). Growth monitoring dur- (Embleton et al. 2001; Cooke et al. 2004). A
ing this time frame should be done at least weekly, “critical epoch” of growth may, therefore, have
if not more often in high-risk infants; i.e., the been missed. Preterm infants also have greater
greater the degree of PGF, the greater the level morbidity than term infants during the first year
of concern, until an acceptable growth pattern is of life (McCormick et al. 1980; Hack et al. 1983;
achieved and maintained. Navas et al. 1992; Thomas et al. 2000; Wang
In the study of Cooke et al. (1998), significant et al. 1995a), and intercurrent illness will affect
growth faltering was noted: growth, irrespective of whether infants are admit-
ted to hospital or not.
(a) In breastfed but not formula-fed infants Until relatively recently, little attention had
between discharge and term (Fig. 4) been paid to nutritional factors in the pathogen-
(b) When a dietary change was made, i.e., infants esis of this problem. For most early studies,
fed a preterm formula between discharge and infants were fed either unfortified human milk
term and then the term formula thereafter or a term infant formula after hospital discharge
(Fig. 4) (Kitchen et al. 1992a, b; Ernst et al. 1990; Casey
et al. 1990; Fitzhardinge and Inwood 1989;
More frequent growth monitoring would also Fenton et al. 1990; Ross et al. 1990). Both feed-
appear prudent under these circumstances. ing regimens were designed to meet nutritional
Which growth parameters should be monitored needs of the term rather than the rapidly growing
and evaluated? Accurate measurement of body preterm infant. Infants, therefore, may have been
weight and head circumference, with subsequent partly underfed during the first 6–12 months
conversion to Z-scores, is necessary to determine of life.
whether or not an infant is “tracking” toward More recently, studies have examined the
original birth-weight centile. Accurate measure- effects of feeding nutrient-enriched formulas,i.e.,
ment of body length, while difficult and time- formulas with a greater nutrient density than a
Post-discharge Nutrition in Preterm Infants 11

Weight Gain in Study Infants* representative. The conclusions drawn by Hen-


55
Weight Gain (g/d) derson et al. on growth, therefore, must be
50
questioned.
45
40
The more recent and more comprehensive
35 meta-analysis of Teller et al. is very informative
30 (Teller et al. 2016). Thirty-one eligible studies
25 were identified which were noted to differ signif-
20 icantly in study design, i.e., (a) inclusion criteria,
15
Preterm ( ) > Term ( ), (b) point of enrolment, (c) nutrient content of
10 Cross-Over (x) (p <.001) formula fed, (d) duration of intervention, and
5 (e) outcomes evaluated.
Disc - 38w - Term - 4w - 8w - 12w - 4m - 5m - 6m
In the main, no consistent differences were
* Cooke et al 16, detected in body weight, length, or head circum-
ference between treatment groups. What was
Fig. 5 Growth velocity in study infants fed either a pre- noted was a consistent dose-response effect, i.e.,
term infant formula (closed circle), a term formula (open protein-energy ratios of 2.5–3.0 g/100 ml were
circle) or a preterm infant formula (discharge-term), or
term infant formula (X)
associated with increased linear growth as well as
increased weight and head circumference noted at
3, 6, 9, and 12 mca (Teller et al. 2016).
term infant formula, to preterm infants after hos- Where body composition was measured using
pital discharge (Cooke et al. 1998, 1999, 2001; dual-emission x-ray absorptiometry (DEXA),
Lucas et al. 1992, 2001; Carver et al. 2001; Agosti increased linear growth was associated with
et al. 2003; Bishop et al. 1993; Chan et al. 1994; increased lean mass accretion, which was
Wheeler and Hall 1996; Lapillonne et al. 2004; paralleled by a reduced or increased fat mass but
Koo and Hockman 2006; De Curtis et al. 2002). In not increased adiposity (Cooke et al. 1999;
this respect, two meta-analyses are relevant. Lapillonne et al. 2004; Roggero et al. 2012). An
Reviewing the results of studies published before exception to these observations was the study of
2005 Henderson et al. concluded that feeding with Koo et al. where reduced weight gain and linear
nutrient-enriched formulas had little effect on growth were associated with reduced lean and fat
growth (Henderson et al. 2005). As such, their mass accretion (Koo and Hockman 2006).
conclusions on growth merit examination.
The first outcome variable evaluated was Development
“growth during the trial period.” The results of In the meta-analyses of Henderson et al. (2005)
one study were used, i.e., de Curtis et al. (2002) and of Teller et al. (2016), no consistent effects
wherein the sample size was 33 and the end point were detected on neurodevelopment. This is not
was gain in weight and crown-heel length and entirely surprising. In the meta-analysis of Teller
head circumference between 36 wca and 2 mca. et al., it was only in infants who were fed the
Furthermore, growth velocity changes rapidly formula with the highest protein content
during this time frame (Fig. 5) and that which is (2.5–3.0 g/100 ml) that consistent effects were
averaged over a 12-week period may not reflect noted on growth, an effect that was more pro-
early but significant differences between the treat- nounced in boys than girls. Yet, these studies
ment groups (Cooke et al. 1998). were not “powered” to control for the
The second end point was “longer-term confounding effect of sex on head growth and
growth,” i.e., weight, length, and head circumfer- development.
ence at 6 m, 9 m, and 18 m corrected age. Data In the study of Lucas et al. (1989a, 1990), sex
from only one study was used at 6 mca was a confounding variable but sample size
(Litmanovitz et al. 2004) and one at 9 mca (n = 219) was large enough, therefore adequately
(Lucas et al. 2001), again not entirely “powered,” to detect differences in development
12 R.J. Cooke

between treatment groups. Such was not the case Breastfeeding


for the studies of Agosti et al. (2003) (n = 121)
and Jeon et al. (2011) (n = 90) and Cooke Human milk has been recommended as the pre-
et al. (2001) (n = 113) where sample sizes are ferred method of feeding for preterm infants after
much smaller. In the latter study, boys fed the hospital discharge (Canada 2004; AAPCON
term formula had the poorest head growth, 2009; Agostoni et al. 2009; Lapillonne
which in turn was paralleled by ten-point reduc- et al. 2013). In a nutritional sense this, at first
tion in MDI when compared to girls fed the term glance, is puzzling. Before hospital discharge,
formula, suggesting that boys are at a disadvan- preterm infants fed human milk do not grow as
tage when fed a term infant formula. well as infants fed nutrient-enriched formulas
An additional consideration is as follows. (Tyson et al. 1983; Gross 1983). It is, therefore,
Brain growth is rapid during the last trimester recommended that human milk be fortified with
and first 2 years of life, a time when even minor additional nutrients (Klein 2002). Growth
insults may have significant effects (Dobbing improves but it is still not as good as in infants
1981). But not all parts of the brain are developing fed a preterm infant formula (Carlson and Ziegler
at the same rate, therefore not equally vulnerable 1998). The reasons for this are not entirely clear.
(Dobbing 1981). In effect global measures of Fortifiers differ in nutrient composition and it
function, such as Bayley’s assessment, may not is unclear which, if any, really meets require-
be sensitive enough to detect minor or focal dif- ments. The composition of human milk varies
ferences in developmental outcome (Singer widely (Atkinson et al. 1995), and because it is
et al. 1994). not consistently measured, there is no way of
Dabydeen et al. prospectively randomized knowing what the infant is really receiving. In
term and preterm infants with perinatal brain effect, intake less adequately meets requirements
injury to either a control or high-energy and and growth is poorer. Nonetheless, because of
high-protein diet after perinatal brain injury dur- lower rates of morbidity, e.g., sepsis and necrotiz-
ing the first year of life (Dabydeen et al. 2008). ing enterocolitis (Schanler et al. 1999), and
Infants fed the high-energy and high-protein diet improved developmental outcome (Morley
had a greater head growth and axonal diameters et al. 1988a), it is recommended that fortified
when compared to the control group. It was con- human milk be fed whenever possible before hos-
cluded that infants with significant perinatal brain pital discharge (Klein 2002).
injury had increased nutritional requirements and After hospital discharge, breastfed infants also
that inadequate intake, as is commonly noted in grow more poorly than those fed nutrient-
neurologically impaired infants, may compromise enriched formulas (Lucas et al. 2001; Chan
subsequent brain growth and development et al. 1994; Wauben et al. 1998). This again is
(Dabydeen et al. 2008). not surprising. Before discharge, intake less ade-
Collectively, these studies (Lucas et al. 2001; quately meets requirements, the accrued nutri-
Agosti et al. 2003; Cooke et al. 2001; Dabydeen tional deficit, and, therefore, needs for
et al. 2008) suggest that the feeding with a formula “recovery” are greater. Mature human milk is
with a protein-energy ratio of 2.5–3.0 g/100 ml designed to meet the needs of the term infant
is associated with better head growth and trends and not the preterm infant, and there are signifi-
toward better post-developmental outcome. In the cant differences in nutrient content between
case of the otherwise “normal” preterm infant, mature human milk and formulas used to feed
male infants are most likely to benefit. In infants infants before and after discharge (Table 7).
with perinatal brain injury, both term and preterm Poorer growth after discharge is paralleled by
infants may benefit. Further more appropriately alterations in bone mineral metabolism and body
“powered” studies are needed to address these composition. Bone mineral accretion is less in
issues. infants fed un-supplemented human milk after
discharge (Lucas et al. 2001; Chan et al. 1994;
Post-discharge Nutrition in Preterm Infants 13

Table 7 A comparison of nutrient content of human milk with preterm, nutrient-enriched, and term infant formulas
Per liter Human milk Preterm Post-discharge Term
Energy kcal 670 810 730 67
Protein g 10 23 20 14
Fat g 35 42 40 36
Carbohydrate g 70 88 78 73
Calcium mg 260 1,400 850 530
Phosphorus mg 140 700 480 330
Sodium mmol 9 15 11 8
Chloride mmol 16 19 17 12
Zinc mg 3.2 12.1 9 6
Vitamin A mg 0.7 3 1 0.6

Wauben et al. 1998; Kurl et al. 2003; Schanler II developmental scores were greater in infants fed
et al. 1992). This, in turn, can be related to out- fortified human milk (Aimone et al. 2009).
come on a short-term, e.g., osteopenia (Lucas In a larger more recent randomized controlled
et al. 1989b) and fractures (Koo et al. 1988), and trial between hospital discharge and 4 m corrected
long-term, e.g., poorer linear growth (Fewtrell age, Zacharisson et al. fed preterm VLBWI
et al. 2000), basis. Interestingly, infants fed (24–32 weeks; 535–2,255 g) either unfortified
human milk also have increased fat accretion (n = 102; group A) or fortified (n = 105; group
when compared to those fed a nutrient-enriched B) human milk and compared their growth to a
formula (Wauben et al. 1998). group of infants fed a preterm formula (n = 113;
Thus, infants grow as they are fed. When min- group C, preterm formula) (Zachariassen
eral intake is inadequate, bone mineral accretion is et al. 2011). No differences were detected in
decreased. Whether the latter in some way alters growth between groups A and B at 12 mca, and
“programming” and, therefore, health in adult life it was concluded that while fortification did not
is unclear. What is clear is that the assessment of affect the durations of breastfeeding, it also did
bone mineral status, i.e., serum phosphorus and not influence growth at 1 year of age.
alkaline phosphatase, is also an important consid- Yet, important and consistent differences were
eration during this critical time frame (Schanler noted in breastfed and the formula-fed infants.
2005). Infants in C had a greater increase in weight
Several studies have examined the effect of Z-score between discharge and term and in length
nutrient fortification of human milk after hospital Z-score until 6 months CA. At 12 mca, boys in C
discharge. In a randomized controlled pilot trial, were longer and heavier than in groups A and B,
preterm infants (750–1,800 g) were fed unfortified while girls in group C were longer and heavier
(n = 20) or fortified (n = 19) human milk after than in group A only. Greater protein intakes in C
hospital discharge-12 week (O’Connor were also related to increased serum urea nitrogen
et al. 2008). A multi-nutrient fortifier, estimated levels.
to ensure an energy and protein density of 80 kcal The results of this much larger scale study are
and 2.2 g/100 ml, was added to 50 % of feeds. puzzling in that no significant differences were
Weight tended to be greater (all infants), while detected in growth between infants fed unfortified
length (all infants) and head circumference, and fortified milk. One possibility relates to the
infants 1,250 g only, were greater in the fortified small sample size in the original study (O’Connor
group. In a follow-up of the same infants, weight, et al. 2008), n ~ 20/treatment group; i.e., the
linear growth, bone mineral content, and head results are not generalizable. Alternatively, the
growth (infants <1,250 g only) but not Bayley’s level of fortification used in the latter study may
have been inadequate to consistently improve
14 R.J. Cooke

growth in such a heterogeneous population. It is information relayed to the primary care physi-
notable that the timing of introduction of comple- cian. Infants with the greatest degree of growth
mentary foods was later with nutrient enrichment, failure at discharge are at greatest risk for fur-
perhaps suggesting that energy needs were more ther “growth faltering.”
adequately met and infants satiated longer. 2. Irrespective of what is fed, growth should be
The results of these studies raise more ques- closely monitored and intake adjusted with the
tions than answers: goal that birth centile, at least for weight, is
reestablished between discharge and 1–2 mca.
(a) Should all preterm VLBWI be fortified human 3. Continuing and close communication between
milk after hospital discharge? Since all designated personnel in the neonatal intensive
VLBWI accrue a significant nutrient deficit care unit and the infant’s family as well as the
between birth and hospital discharge and primary care providers is essential during this
mature human milk is designed for the needs critical “epoch” of growth and development.
of “normal” infants, i.e., needs for mainte-
nance and normal growth, it seems prudent
to fortify in all breastfed infants after References
discharge.
(b) What is the best way to fortify human milk? AAPCON (1998) Nutritional needs of preterm infants. In:
Kleinman RE (ed) Pediatric nutrition handbook. Amer-
The nutritional content of fortifiers differs. Is
ican Academy of Pediatrics, Elk Grove Village, pp
one better than another? Further studies are 55–88
needed to examine this issue. In the mean- AAPCON (2009) Nutritional needs of the preterm infant.
time, the level of fortification should be In: Kleinman RE (ed) Pediatric nutrition handbook, 6th
edn. American Academy of Pediatrics, Elk Grove Vil-
adjusted to ensure that growth, in terms of
lage, pp 79–104
weight and length gain, is “tracking” in the Aggett PJ, Agostoni C, Axelsson I et al (2006) Feeding
right direction (Arslanoglu et al. 2006; Cooke preterm infants after hospital discharge: a commentary
2006). by the ESPGHAN committee on nutrition. J Pediatr
Gastroenterol Nutr 42:596–603
(c) How long should fortification be continued?
Agosti M, Vegni C, Calciolari G, Marini A (2003) Post-
There are no data in this area. Data in formula- discharge nutrition of the very low-birthweight infant:
fed infants indicate that discontinuation of interim results of the multicentric GAMMA study. Acta
preterm formula and then feeding a term for- Paediatr Suppl 91:39–43
Agostoni C, Braegger C, Decsi T et al (2009) Breast-
mula at term is paralleled by a rapid deceler-
feeding: a commentary by the ESPGHAN committee
ation in growth that is not entirely recovered on nutrition. J Pediatr Gastroenterol Nutr 49:112–125
by 6 or 12 m corrected age (Fig. 4; Cooke Aimone A, Rovet J, Ward W et al (2009) Growth and body
et al. 1998). At least, 1–2 mca, therefore, composition of human milk-fed premature infants pro-
vided with extra energy and nutrients early after hospi-
seems advisable since this is also the period
tal discharge: 1-year follow-up. J Pediatr Gastroenterol
of greatest brain growth velocity and appears Nutr 49:456–466
to be functionally significant (Dharmaraj Alexander GR, Himes JH, Kaufman RB, Mor J, Kogan M
et al. 2005). (1996) A United States national reference for fetal
growth. Obstet Gynecol 87:163–168
Arslanoglu S, Moro GE, Ziegler EE (2006) Adjustable
fortification of human milk fed to preterm infants:
Summary does it make a difference? J Perinatol 26:614–621
Ashworth A, Millward DJ (1986) Catch-up growth in
children. Nutr Rev 44:157–163
Many questions remain unanswered about what to
Atkinson SA, Alston-Mills B, Lonnerdal B, Neville MC
feed preterm, particularly VLBWI, infants after (1995) Major minerals and ionic constituents of human
hospital discharge but several things are clear: and bovine milks. In: Jensen RG (ed) Handbook of
milk composition. Academic, San Diego, pp 593–622
Bedi KS (1987) Lasting neuroanatomical changes follow-
1. Nutritional status of all infants should be
ing undernutrition during early life. In: Dobbing J
closely evaluated before discharge and the
Post-discharge Nutrition in Preterm Infants 15

(ed) Early nutrition and later achievement. Academic, Cordero ME, D’Acuna E, Benveniste S, Prado R, Nunez
London, pp 1–49 JA, Colombo M (1993) Dendritic development in neo-
Benitez-Bribiesca L, De la Rosa-Alvarez I, Mansilla- cortex of infants with early postnatal life undernutri-
Olivares A (1999) Dendritic spine pathology in infants tion. Pediatr Neurol 9:457–464
with severe protein-calorie malnutrition. Pediatrics Cornier MA, Dabelea D, Hernandez TL et al (2008) The
104:e21 metabolic syndrome. Endocr Rev 29:777–822
Bishop NJ, King FJ, Lucas A (1993) Increased bone min- Dabydeen L, Thomas JE, Aston TJ, Hartley H, Sinha SK,
eral content of preterm infants fed with a nutrient Eyre JA (2008) High-energy and -protein diet increases
enriched formula after discharge from hospital. Arch brain and corticospinal tract growth in term and preterm
Dis Child 68:573–578 infants after perinatal brain injury. Pediatrics
Bremer AA, Mietus-Snyder M, Lustig RH (2012) Toward 121:148–156
a unifying hypothesis of metabolic syndrome. Pediat- De Curtis M, Pieltain C, Rigo J (2002) Body composition
rics 129:557–570 in preterm infants fed standard term or enriched for-
Carlson SJ, Ziegler EE (1998) Nutrient intakes and growth mula after hospital discharge. Eur J Nutr 41:177–182
of very low birth weight infants. Perinatology Dharmaraj ST, Henderson M, Embleton ND, Fenton AC,
18:252–258 Cooke RJ (2005) Postnatal growth retardation, catch-
Carver JD, Wu PY, Hall RT et al (2001) Growth of preterm up growth and developmental outcome in preterm
infants fed nutrient-enriched or term formula after hos- infants. Arch Dis Child 90:11A
pital discharge. Pediatrics 107:683–689 Dinerstein A, Nieto RM, Solana CL, Perez GP, Otheguy
Casey PH, Kraemer HC, Bernbaum J et al (1990) Growth LE, Larguia AM (2006) Early and aggressive nutri-
patterns of low birth weight preterm infants: a longitu- tional strategy (parenteral and enteral) decreases post-
dinal analysis of a large, varied sample. J Pediatr natal growth failure in very low birth weight infants. J
117:298–307 Perinatol 26:436–442
Casey PH, Kraemer HC, Bernbaum J, Yogman MW, Sells Dobbing J (1981) The later development of the brain and
JC (1991) Growth status and growth rates of a varied its vulnerability. In: Davis JA, Dobbing J (eds) Scien-
sample of low birth weight, preterm infants: a longitu- tific foundations of pediatrics. University Park Press,
dinal cohort from birth to three years of age. J Pediatr Baltimore, pp 744–758
119:599–605 Dobbing J (1987) Early nutrition and later achievement.
Chan GM, Borschel MW, Jacobs JR (1994) Effects of Academic, London
human milk or formula feeding on the growth, behav- Dobbing J, Sands J (1971) Vulnerability of developing
ior, and protein status of preterm infants discharged brain. IX. The effect of nutritional growth retardation
from the newborn intensive care unit. Am J Clin Nutr on the timing of the brain growth-spurt. Biol Neonate
60:710–716 19:363–378
Clark RH, Thomas P, Peabody J (2003) Extrauterine Dobbing J, Hopewell JW, Lynch A (1971) Vulnerability of
growth restriction remains a serious problem in prema- developing brain. VII. Permanent deficit of neurons in
turely born neonates. Pediatrics 111:986–990 cerebral and cerebellar cortex following early mild
Cooke RJ (2006) Adjustable fortification of human milk undernutrition. Exp Neurol 32:439–447
fed to preterm infants. J Perinatol 26:591–592 Donovan R, Puppala B, Angst D, Coyle BW (2006) Out-
Cooke RW, Foulder-Hughes L (2003) Growth impairment comes of early nutrition support in extremely low-
in the very preterm and cognitive and motor perfor- birth-weight infants. Nutr Clin Pract 21:395–400
mance at 7 years. Arch Dis Child 88:482–487 Ehrenkranz RA, Younes N, Lemons JA et al (1999) Lon-
Cooke RJ, Griffin I (2009) Altered body composition in gitudinal growth of hospitalized very low birth weight
preterm infants at hospital discharge. Acta Paediatr infants. Pediatrics 104:280–289
98:1269–1273 Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage
Cooke RJ, Griffin IJ, McCormick K et al (1998) Feeding LA, Poole WK (2006) Growth in the neonatal intensive
preterm infants after hospital discharge: effect of die- care unit influences neurodevelopmental and growth
tary manipulation on nutrient intake and growth. outcomes of extremely low birth weight infants. Pedi-
Pediatr Res 43:355–360 atrics 117:1253–1261
Cooke RJ, McCormick K, Griffin IJ et al (1999) Feeding Embleton NE, Pang N, Cooke RJ (2001) Postnatal malnu-
preterm infants after hospital discharge: effect of diet trition and growth retardation: an inevitable conse-
on body composition. Pediatr Res 46:461–464 quence of current recommendations in preterm
Cooke RJ, Embleton ND, Griffin IJ, Wells JC, McCormick infants? Pediatrics 107:270–273
KP (2001) Feeding preterm infants after hospital dis- Ernst JA, Bull MJ, Rickard KA, Brady MS, Lemons JA
charge: growth and development at 18 months of age. (1990) Growth outcome and feeding practices of the
Pediatr Res 49:719–722 very low birth weight infant (less than 1500 grams)
Cooke RJ, Ainsworth SB, Fenton AC (2004) Postnatal within the first year of life. J Pediatr 117:S156–S166
growth retardation: a universal problem in preterm Evans RA, Thureen P (2001) Early feeding strategies in
infants. Arch Dis Child Fetal Neonatal Ed 89: preterm and critically ill neonates. Neonatal Netw
F428–F430 20:7–18
16 R.J. Cooke

Fenton TR, McMillan DD, Sauve RS (1990) Nutrition and Heird WC (2001) Determination of nutritional require-
growth analysis of very low birth weight infants. Pedi- ments in preterm infants, with special reference to
atrics 86:378–383 ‘catch-up’ growth. Semin Neonatol 6:365–375
Fewtrell PA, Cole TJ, Lucas A (2000) Effects of growth Henderson G, Fahey T, McGuire W (2005) Calorie and
during infancy and childhood on bone mineralization protein-enriched formula versus standard term for-
and turnover in preterm children aged 8–12 years. Acta mula for improving growth and development in
Paediatr 89:148–153 preterm or low birth weight infants following hos-
Fitzhardinge PM, Inwood S (1989) Long-term growth in pital discharge. Cochrane Database Syst Rev
small-for-date children. Acta Paediatr Scand – Suppl CD004696
349:27–33, discussion 4 Hermanussen M (2013a) Basics. Rapid growth. In:
Fomon S (1993a) Energy. In: Fomon SJ, Bell EF (eds) Hermanussen M (ed) Auxology studying human
Nutrition of normal infants, 2nd edn. Mosby, St growth and development. Schweizerbart’sche,
Louis, pp 103–120 Stuttgaart, pp 18–19
Fomon S (1993b) Protein. In: Fomon SJ (ed) Nutrition Hermanussen M (2013b) Basics. In: Hermanussen M
of normal infants, 2nd edn. Mosby, St Louis, pp (ed) Auxology studying human growth and develop-
121–139 ment. Schweizerbart’sche, Stuttgart
Galler J, Shumsky J, Morgane PJ (1996) Malnutrition and Horbar JD, Ehrenkranz RA, Badger GJ et al (2015) Weight
brain development. In: Walker AW, Watkins J (eds) growth velocity and postnatal growth failure in infants
Paediatric nutrition. Decker, New York, pp 196–212 501 to 1500 grams: 2000–2013. Pediatrics 136:
Greisen G (1992) Estimation of fetal weight by ultrasound. e84–e92
Horm Res 38:208–210 Ibrahim HM, Jeroudi MA, Baier RJ, Dhanireddy R,
Griffin IJ, Tancredi DJ, Bertino E, Lee HC, Profit J (2016) Krouskop RW (2004) Aggressive early total parental
Postnatal growth failure in very low birthweight infants nutrition in low-birth-weight infants. J Perinatol
born between 2005 and 2012. Arch Dis Child Fetal 24:482–486
Neonatal Ed 101:50–55 Jackson AA (1990) Protein requirements for catch-up
Gross SJ (1983) Growth and biochemical response of growth. Proc Nutr Soc 49:507–516
preterm infants fed human milk or modified infant Jeon GW, Jung YJ, Koh SY et al (2011) Preterm infants fed
formula. N Engl J Med 308:237–241 nutrient-enriched formula until 6 months show
Gross SJ, Oehler JM, Eckerman CO (1983) Head growth improved growth and development. Pediatr Int: Off J
and developmental outcome in very low-birth-weight Jpn Pediatr Soc 53:683–688
infants. Pediatrics 71:70–75 Kitchen WH, Doyle LW, Ford GW, Callanan C, Rickards
Hack M, Breslau N (1986) Very low birth weight infants: AL, Kelly E (1992a) Very low birth weight and growth
effects of brain growth during infancy on intelligence to age 8 years. II: head dimensions and intelligence.
quotient at 3 years of age. Pediatrics 77:196–202 Am J Dis Child 146:46–50
Hack M, Fanaroff AA (1984) The outcome of growth Kitchen WH, Doyle LW, Ford GW, Callanan C (1992b)
failure associated with preterm birth. Clin Obstet Very low birth weight and growth to age 8 years. I:
Gynecol 27:647–663 weight and height. Am J Dis Child 146:40–45
Hack M, Merkatz IR, Gordon D, Jones PK, Fanaroff AA Klein CJ (2002) Nutrient requirements for preterm infant
(1982) The prognostic significance of postnatal growth formulas. J Nutr 132:1395S–1577S
in very low – birth weight infants. Am J Obstet Gynecol Koo WW, Hockman EM (2006) Posthospital discharge
143:693–699 feeding for preterm infants: effects of standard com-
Hack M, Caron B, Rivers A, Fanaroff AA (1983) The very pared with enriched milk formula on growth, bone
low birth weight infant: the broader spectrum of mor- mass, and body composition. Am J Clin Nutr
bidity during infancy and early childhood. J Dev Behav 84:1357–1364
Pediatr 4:243–249 Koo WW, Sherman R, Succop P et al (1988) Sequential
Hack M, Breslau N, Weissman B, Aram D, Klein N, bone mineral content in small preterm infants with and
Borawski E (1991) Effect of very low birth weight without fractures and rickets. J Bone Miner Res
and subnormal head size on cognitive abilities at school 3:193–197
age [see comments]. N Engl J Med 325:231–237 Krigman MR, Hogan EL (1976) Undernutrition in the
Hack M, Weissman B, Breslau N, Klein N, Borawski- developing rat: effect upon myelination. Brain Res
Clark E, Fanaroff AA (1993) Health of very low birth 107:239–255
weight children during their first eight years. J Pediatr Kurl S, Heinonen K, Lansimies E (2003) Pre- and post-
122:887–892 discharge feeding of very preterm infants: impact on
Hack M, Schluchter M, Cartar L, Rahman M, Cuttler L, growth and bone mineralization. Clin Physiol Funct
Borawski E (2003) Growth of very low birth weight Imaging 23:182–189
infants to age 20 years. Pediatrics 112:e30–e38 Lapillonne A, Salle BL, Glorieux FH, Claris O (2004)
Health Canada (2004) Breast feeding duration Bone mineralization and growth are enhanced in pre-
recommendations term infants fed an isocaloric, nutrient-enriched
Post-discharge Nutrition in Preterm Infants 17

preterm formula through term. Am J Clin Nutr Morley R (1999) Early growth and later development. In:
80:1595–1603 Ziegler EE, Lucas A, Moro GE (eds) Nutrition of the
Lapillonne A, O’Connor DL, Wang D, Rigo J (2013) very low birth weight infant. Lippincott Williams and
Nutritional recommendations for the late-preterm Wilkins, Philadelphia, pp 19–32
infant and the preterm infant after hospital discharge. Morley R, Lucas A (1997) Nutrition and cognitive devel-
J Pediatr 162:S90–S100 opment. Br Med Bull 53:123–134
Larsen T, Petersen S, Greisen G, Larsen JF (1990) Normal Morley R, Cole TJ, Powell R, Lucas A (1988) Mother’s
fetal growth evaluated by longitudinal ultrasound choice to provide breast milk and developmental out-
examinations. Early Hum Dev 24:37–45 come. Arch Dis Child 63:1382–1385
Latal-Hajnal B, von Siebenthal K, Kovari H, Bucher HU, Myers RE, Hill DE, Holt AB, Scott RE, Mellits ED, Cheek
Largo RH (2003) Postnatal growth in VLBW infants: DB (1971) Fetal growth retardation produced by exper-
significant association with neurodevelopmental out- imental placental insufficiency in the rhesus monkey.
come. J Pediatr 143:163–170 I. Body weight, organ size. Biol Neonate 18:379–394
Litmanovitz I, Dolfin T, Arnon S et al (2004) Bone strength Navas L, Wang E, de Carvalho V, Robinson J, PICNIC
and growth of preterm infants fed nutrient-enriched or (1992) Improved outcome of respiratory syncytial virus
term formula after hospital discharge. Pediatr Res infection in a high-risk hospitalized population of
55:274A Canadian children. J Pediatr 121:348–354
Lowry S, Perez JM (2006) The hypercatabolic state. In: O’Connor DL, Khan S, Weishuhn K et al (2008) Growth
Shils M, Shike M, Ross CA, Caballero B, Cousins R and nutrient intakes of human milk-fed preterm infants
(eds) Modern nutrition in health and disease, 10th edn. provided with extra energy and nutrients after hospital
Lippincott, Williams & Wilkins, Philadelphia, pp discharge. Pediatrics 121:766–776
1381–1400 Olsen IE, Richardson DK, Schmid CH, Ausman LM,
Lucas A, Morley R, Cole TJ et al (1989a) Early diet in Dwyer JT (2002) Intersite differences in weight growth
preterm babies and developmental status in infancy velocity of extremely premature infants. Pediatrics
[see comments]. Arch Dis Child 64:1570–1578 110:1125–1132
Lucas A, Brooke OG, Baker BA, Bishop N, Morley R Ong KK, Loos RJ (2006) Rapid infancy weight gain and
(1989b) High alkaline phosphatase activity and growth subsequent obesity: systematic reviews and hopeful
in preterm neonates. Arch Dis Child 64:902–909 suggestions. Acta Paediatr 95:904–908
Lucas A, Morley R, Cole TJ et al (1990) Early diet in Ounsted M, Moar VA, Scott A (1988) Head circumference
preterm babies and developmental status at 18 months. and developmental ability at the age of seven years.
Lancet 335:1477–1481 Acta Paediatr Scand 77:374–379
Lucas A, Bishop NJ, King FJ, Cole TJ (1992) Randomised Peterson J, Taylor HG, Minich N, Klein N, Hack M (2006)
trial of nutrition for preterm infants after discharge [see Subnormal head circumference in very low birth
comments]. Arch Dis Child 67:324–327 weight children: neonatal correlates and school-age
Lucas A, Morley R, Cole TJ, Gore SM (1994) A consequences. Early Hum Dev 82:325–334
randomised multicentre study of human milk versus Pryor J, Silva PA, Brooke M (1995) Growth, development
formula and later development in preterm infants. and behaviour in adolescents born small-for-gesta-
Arch Dis Child Fetal Neonatal Ed 70:F141–F146 tional-age. J Paediatr Child Health 31:403–407
Lucas A, Morley R, Cole TJ (1998) Randomised trial of Rawlings DJ, Cooke RJ, McCormick K et al (1999) Body
early diet in preterm babies and later intelligence quo- composition of preterm infants during infancy. Arch
tient. BMJ 317:1481–1487 Dis Child Fetal Neonatal Ed 80:F188–F191
Lucas A, Fewtrell MS, Morley R et al (2001) Randomized Roberts SB, Young VR (1988) Energy costs of fat and
trial of nutrient-enriched formula versus standard for- protein deposition in the human infant. Am J Clin
mula for postdischarge preterm infants. Pediatrics Nutr 48:951–955
108:703–711 Roggero P, Gianni ML, Piemontese P, Amato O, Agosti M,
MacLean WC Jr, Graham GG (1980) The effect of energy Mosca F (2012) Effect of nutrition on growth and body
intake on nitrogen content of weight gained by recov- composition in infants born preterm. J Matern Fetal
ering malnourished infants. Am J Clin Nutr Neonatal Med 25(Suppl 3):49–52
33:903–909 Ross G, Lipper EG, Auld PA (1990) Growth achievement
McCance RA, Widdowson EM (1974) The determinants of of very low birth weight premature children at school
growth and form. Proc R Soc Lond B Biol Sci age. J Pediatr 117:307–309
185:1–17 Schanler RJ (2005) Post-discharge nutrition for the preterm
McCormick MC, Shapiro S, Starfield BH (1980) infant. Acta Paediatr Suppl 94:68–73
Rehospitalization in the first year of life for high-risk Schanler RJ, Burns PA, Abrams SA, Garza C (1992) Bone
survivors. Pediatrics 66:991–999 mineralization outcomes in human milk-fed preterm
Mehta NM, Corkins MR, Lyman B et al (2013) Defining infants. Pediatr Res 31:583–586
pediatric malnutrition: a paradigm shift toward Schanler RJ, Shulman RJ, Lau C (1999) Feeding strategies
etiology-related definitions. JPEN J Parenter Enteral for premature infants: beneficial outcomes of feeding
Nutr 37:460–481
18 R.J. Cooke

fortified human milk versus preterm formula [com- Uthaya S, Thomas EL, Hamilton G, Dore CJ, Bell J, Modi
ment]. Pediatrics 103:1150–1157 N (2005) Altered adiposity after extremely preterm
Scott KE, Usher R (1966) Fetal malnutrition: its incidence, birth. Pediatr Res 57:211–215
causes, and effects. Am J Obstet Gynecol 94:951–963 Volpe JJ (2008) Neurology of the newborn, 5th edn.
Shils ME, Shike M, Ross AC, Caballero B, Cousins RJ Saunders Elsevier, Philadelphia
(2006) Modern nutrition in health and disease. Wang E, Law B, Stephens D, PICNIC (1995) Prospective
Lippincott Williams and Wilkins, Philadelphia study of risk factors and outcomes in patients
Singer L, Arendt R, Song LY, Warshawsky E, Kliegman R hospitalised with respiratory syncytial viral lower
(1994) Direct and indirect interactions of cocaine with respiratory tract infection. J Pediatr 126:212–219
childbirth outcomes. Arch Pediatr Adolesc Med Wauben IP, Atkinson SA, Shah JK, Paes B (1998) Growth
148:959–964 and body composition of preterm infants: influence of
Singhal A, Cole TJ, Lucas A (2001) Early nutrition in nutrient fortification of mother’s milk in hospital and
preterm infants and later blood pressure: two cohorts breastfeeding post- hospital discharge (in process cita-
after randomised trials. Lancet 357:413–419 tion). Acta Paediatr 87:780–785
Singhal A, Fewtrell M, Cole TJ, Lucas A (2003) Low Wheeler RE, Hall RT (1996) Feeding of premature infant
nutrient intake and early growth for later insulin resis- formula after hospital discharge of infants weighing
tance in adolescents born preterm. Lancet less than 1800 grams at birth. J Perinatol 16:111–116
361:1089–1097 Widdowson EM, McCance RA (1975) A review: new
Stathis SL, O’Callaghan M, Harvey J, Rogers Y (1999) thoughts on growth. Pediatr Res 9:154–156
Head circumference in ELBW babies is associated with Wilson DC, Cairns P, Halliday HL, Reid M, McClure G,
learning difficulties and cognition but not ADHD in the Dodge JA (1997) Randomised controlled trial of an
school-aged child. Dev Med Child Neurol 41:375–380 aggressive nutritional regimen in sick very low
Stoch MB, Smythe PM, Moodie AD, Bradshaw D (1982) birthweight infants. Arch Dis Child Fetal Neonatal Ed
Psychosocial outcome and CT findings after gross 77:F4–F11
undernourishment during infancy: a 20-year develop- Winick M, Rosso P (1969a) The effect of severe early
mental study. Dev Med Child Neurol 24:419–436 malnutrition on cellular growth of human brain. Pediatr
Teller IC, Embleton ND, Griffin IJ, van Elburg RM (2016) Res 3:181–184
Post-discharge formula feeding in preterm infants: a Winick M, Rosso P (1969b) Head circumference and cel-
systematic review mapping evidence about the role of lular growth of the brain in normal and marasmic chil-
macronutrient enrichment. Clin Nutr 35(4):791–801 dren. J Pediatr 74:774–778
Thomas M, Bedford-Russel A, Sharland M (2000) Yeung MY (2006) Postnatal growth, neurodevelopment
Hospitalisation of RSV infection in ex-preterm infants and altered adiposity after preterm birth – from a clin-
– implications for RSV immune globulin. Arch Dis ical nutrition perspective. Acta Paediatr 95:909–917
Child 183:122–127 Zachariassen G, Faerk J, Grytter C et al (2011) Nutrient
Tyson JE, Lasky RE, Mize CE et al (1983) Growth, met- enrichment of mother’s milk and growth of very pre-
abolic response, and development in very-low-birth- term infants after hospital discharge. Pediatrics 127:
weight infants fed banked human milk or enriched e995–e1003
formula. I. Neonatal findings. J Pediatr 103:95–104 Ziegler EE, Thureen PJ, Carlson SJ (2002) Aggressive
nutrition of the very low birthweight infant. Clin
Perinatol 29:225–244
 
Cochrane
Library
Cochrane Database of Systematic Reviews

   
Delayed introduction of progressive enteral feeds to prevent
necrotising enterocolitis in very low birth weight infants (Review)

  Morgan J, Young L, McGuire W  

  Morgan J, Young L, McGuire W.  


Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants.
Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No.: CD001970.
DOI: 10.1002/14651858.CD001970.pub5.

  www.cochranelibrary.com  

 
Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight
infants (Review)
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

TABLE OF CONTENTS
HEADER......................................................................................................................................................................................................... 1
ABSTRACT..................................................................................................................................................................................................... 1
PLAIN LANGUAGE SUMMARY....................................................................................................................................................................... 2
BACKGROUND.............................................................................................................................................................................................. 3
OBJECTIVES.................................................................................................................................................................................................. 3
METHODS..................................................................................................................................................................................................... 3
RESULTS........................................................................................................................................................................................................ 5
Figure 1.................................................................................................................................................................................................. 6
Figure 2.................................................................................................................................................................................................. 7
Figure 3.................................................................................................................................................................................................. 7
Figure 4.................................................................................................................................................................................................. 8
Figure 5.................................................................................................................................................................................................. 9
DISCUSSION.................................................................................................................................................................................................. 9
AUTHORS' CONCLUSIONS........................................................................................................................................................................... 10
ACKNOWLEDGEMENTS................................................................................................................................................................................ 10
REFERENCES................................................................................................................................................................................................ 11
CHARACTERISTICS OF STUDIES.................................................................................................................................................................. 14
DATA AND ANALYSES.................................................................................................................................................................................... 22
Analysis 1.1. Comparison 1 Delayed versus early introduction of progressive enteral feeding, Outcome 1 Necrotising 22
enterocolitis...........................................................................................................................................................................................
Analysis 1.2. Comparison 1 Delayed versus early introduction of progressive enteral feeding, Outcome 2 Mortality prior to 23
discharge................................................................................................................................................................................................
Analysis 1.3. Comparison 1 Delayed versus early introduction of progressive enteral feeding, Outcome 3 Feed intolerance........ 24
Analysis 1.4. Comparison 1 Delayed versus early introduction of progressive enteral feeding, Outcome 4 Incidence of invasive 24
infection.................................................................................................................................................................................................
Analysis 1.5. Comparison 1 Delayed versus early introduction of progressive enteral feeding, Outcome 5 Duration of hospital 24
admission (days)...................................................................................................................................................................................
WHAT'S NEW................................................................................................................................................................................................. 24
HISTORY........................................................................................................................................................................................................ 25
CONTRIBUTIONS OF AUTHORS................................................................................................................................................................... 26
DECLARATIONS OF INTEREST..................................................................................................................................................................... 26
SOURCES OF SUPPORT............................................................................................................................................................................... 26
INDEX TERMS............................................................................................................................................................................................... 26

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) i
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

[Intervention Review]

Delayed introduction of progressive enteral feeds to prevent necrotising


enterocolitis in very low birth weight infants

Jessie Morgan1, Lauren Young2, William McGuire1

1Hull York Medical School & Centre for Reviews and Dissemination, University of York, York, UK. 2Neonatal Unit, Mercy Hospital for
Women, Heidelberg, Australia

Contact address: William McGuire, Hull York Medical School & Centre for Reviews and Dissemination, University of York, York, Y010 5DD,
UK. William.McGuire@hyms.ac.uk.

Editorial group: Cochrane Neonatal Group


Publication status and date: New search for studies and content updated (no change to conclusions), published in Issue 12, 2014.

Citation: Morgan J, Young L, McGuire W. Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis
in very low birth weight infants. Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No.: CD001970. DOI:
10.1002/14651858.CD001970.pub5.

Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

ABSTRACT

Background
The introduction of enteral feeds for very preterm (less than 32 weeks' gestation) or very low birth weight (VLBW; less than 1500 g) infants
is often delayed for several days or longer after birth due to concern that early introduction may not be tolerated and may increase the risk
of necrotising enterocolitis (NEC). However, delaying enteral feeding could diminish the functional adaptation of the gastrointestinal tract
and prolong the need for parenteral nutrition with its attendant infectious and metabolic risks.

Objectives
To determine the effect of delayed introduction of progressive enteral feeds on the incidence of NEC, mortality and other morbidities in
very preterm or VLBW infants.

Search methods
We searched the Cochrane Central Register of Controlled Trials (CENTRAL, 2014, Issue 8), MEDLINE (1966 to September 2014), EMBASE
(1980 to September 2014), CINAHL (1982 to September 2014), conference proceedings and previous reviews.

Selection criteria
We included randomised or quasi-randomised controlled trials that assessed the effect of delayed (more than four days after birth) versus
earlier introduction of progressive enteral feeds on the incidence of NEC, mortality and other morbidities in very preterm or VLBW infants.

Data collection and analysis


Two review authors independently assessed trial eligibility and risk of bias and undertook data extraction. We analysed the treatment
effects in the individual trials and reported the risk ratio (RR) and risk difference for dichotomous data and mean difference for continuous
data, with respective 95% confidence intervals (CI). We used a fixed-effect model in meta-analyses and explored the potential causes of
heterogeneity in sensitivity analyses.

Main results
We identified nine randomised controlled trials in which 1106 infants participated. Few participants were extremely preterm (less 28 weeks'
gestation) or extremely low birth weight (less than 1000 g). The trials defined delayed introduction of progressive enteral feeds as later than
four to seven days after birth and early introduction as four days or less after birth. Meta-analyses did not detect statistically significant
effects on the risk of NEC (typical RR 0.93, 95% CI 0.64 to 1.34; 8 trials; 1092 infants) or all-cause mortality (typical RR 1.18, 95% CI 0.75

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 1
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

to 1.88; 7 trials; 967 infants). Four of the trials restricted participation to growth-restricted infants with Doppler ultrasound evidence of
abnormal fetal circulatory distribution or flow. Planned subgroup analyses of these trials found no statistically significant effects on the risk
of NEC or all-cause mortality. Infants who had delayed introduction of enteral feeds took longer to establish full enteral feeding (reported
median differences two to four days).

Authors' conclusions
The evidence available from randomised controlled trials suggested that delaying the introduction of progressive enteral feeds beyond
four days after birth did not reduce the risk of developing NEC in very preterm or VLBW infants, including growth-restricted infants. Delaying
the introduction of progressive enteral feeds resulted in a few days' delay in establishing full enteral feeds but the clinical importance
of this effect was unclear. The applicability of these findings to extremely preterm or extremely low birth weight was uncertain. Further
randomised controlled trials in this population may be warranted.

PLAIN LANGUAGE SUMMARY

No evidence that delayed introduction of progressive enteral feeds prevents necrotising enterocolitis in very low birth weight
infants

Background

Very preterm (less than 32 weeks' gestation) or very low birth weight (less than 1500 g) infants are at risk of developing a severe bowel
disorder called necrotising enterocolitis, where parts of the bowel become inflamed and start to die. One possible way to prevent this
condition is to delay the introduction of milk feeds until several days (or longer) after birth.

Study characteristics

We search scientific databases for clinical trials assessing the effect of delayed (more than four days after birth) versus earlier introduction
of progressive enteral feeds (where breast or formula milk is fed directly by a tube into the stomach) on the incidence of necrotising
enterocolitis, death and general health in very low birth weight infants. The evidence is current to September 2014.

Key results

We found nine trials with 1106 infants that assessed the effect of delayed rather than early introduction of milk feeds for very preterm or very
low birth weight infants. Data from these trials did not provide any evidence that delaying enteral feeding reduces the risk of necrotising
enterocolitis.

Quality of the evidence

The included trials were generally of reasonable methodological quality but, in common with other trials of feeding interventions in infants,
it was not possible to mask carers and clinical assessors to the given treatment.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 2
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

BACKGROUND METHODS

Description of the condition Criteria for considering studies for this review
Necrotising enterocolitis (NEC) is an important cause of morbidity, Types of studies
mortality and neuro-disability in very preterm (less than 32 weeks'
Randomised or quasi-randomised controlled trials or cluster-
gestation) or very low birth weight (VLBW: less than 1500 g)
randomised trials.
infants. Extremely low birth weight (ELBW: less than 1000 g) and
extremely preterm (less than 28 weeks' gestation) infants are at Types of participants
greatest risk (Bisquera 2002; Holman 2006; Rees 2007; Berrington
2012). Intrauterine growth restriction may be an additional specific VLBW (less than 1500 g) or very preterm (less than 32 weeks'
risk factor, especially if associated with circulatory redistribution gestation) newborn infants.
demonstrated by absent or reversed end-diastolic flow velocities
in antenatal Doppler studies of the fetal aorta or umbilical artery Types of interventions
(Bernstein 2000; Garite 2004; Dorling 2005; Kamoji 2008). Delayed introduction (four or more days after birth) of progressive
enteral feeds versus earlier introduction of enteral feeds. We
Description of the intervention defined progressive enteral feeding as the intention to advance
Most very preterm or VLBW infants who develop NEC have received feed volumes in excess of minimal enteral nutrition levels (24 mL/
enteral milk feeds. Evidence exists that feeding with artificial kg/day) within five days of commencement or by one week after
formula rather than human milk increases the risk of developing birth.
NEC (Quigley 2014). The timing of the introduction and the rate
Infants in each group should have received the same type of milk
of progression of enteral feed volumes may also be modifiable
(breast milk or formula), the same route and mode of feeding
risk factors for the development of NEC (Brown 1978; Uauy 1991;
(intragastric or transpyloric, bolus gavage or continuous) and the
Henderson 2009). Data from observational studies suggest that
same rate of feed volume advancement in both groups.
using feeding regimens that include delaying the introduction of
progressive enteral feeds until beyond about four to seven days Types of outcome measures
after birth reduces the risk of NEC (Patole 2005; Hay 2008).
Primary outcomes
Why it is important to do this review
1. NEC confirmed by at least two of the following features:
In current clinical practice, the introduction of progressive enteral
feeds for very preterm or VLBW infants is often preceded by a period a. abdominal radiograph showing pneumatosis intestinalis or gas
of enteral fasting or 'minimal enteral nutrition' (Boyle 2004; Patole in the portal venous system or free air in the abdomen;
2004; Hay 2008; Klingenberg 2012). However, there may also be
b. abdominal distension with abdominal radiograph with gaseous
potential disadvantages associated with delaying the introduction
distension or frothy appearance of bowel lumen (or both);
of progressive enteral feeds. Because gastrointestinal hormone
secretion and motility are stimulated by enteral milk, delayed c. blood in stool;
enteral feeding could diminish the functional adaptation of the
gastrointestinal tract (Berseth 1990; Burrin 2002). Prolonging the d. lethargy, hypotonia or apnoea (or combination of these).
duration of use of parenteral nutrition may be associated with
infectious and metabolic complications that increase mortality and Or by a diagnosis confirmed at surgery or autopsy (Walsh 1986).
morbidity, prolong hospital stay, and adversely affect growth and
development (Flidel-Rimon 2004; Stoll 2004). It has been argued 2. All-cause mortality during the neonatal period and prior to
that the risk of NEC should not be considered in isolation of hospital discharge.
these other potential clinical outcomes when determining feeding
Secondary outcomes
policies and practice for very preterm or VLBW infants (Flidel-Rimon
2006; Hay 2008; Hartel 2009). 3. Growth:

This review focused on the comparison of delayed versus earlier a. time to regain birth weight and subsequent rates of weight gain,
introduction of progressive enteral feeding; that is, advancing the linear growth, head growth or skinfold thickness growth up to six
volume of milk feeds beyond minimal enteral nutrition levels. months (corrected for preterm birth);
We addressed the effect of minimal enteral nutrition, the early
introduction of small volume enteral feeds (up to 24 mL/kg/day) b. long-term growth: weight, height or head circumference (with or
without advancing the feed volumes for at least five days versus without proportion of infants who remain below the 10th percentile
enteral fasting in another Cochrane review (Morgan 2013a). for the index population's distribution) assessed at intervals from
six months of age.
OBJECTIVES
4. Neurodevelopment:
To determine the effect of delayed introduction of progressive
enteral feeds on the incidence of NEC, mortality and other a. death or severe neurodevelopmental disability defined as any
morbidities in very preterm or VLBW infants. one or combination of the following: non-ambulant cerebral
palsy, developmental delay (developmental quotient less than 70),

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 3
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

auditory and visual impairment. Each component was analysed Selection of studies
individually as well as part of the composite outcome;
Two review authors screened the title and abstract of all studies
b. neurodevelopmental scores in survivors aged 12 months or identified by the above search strategy. We assessed the full text
greater measured using validated assessment tools; of any potentially eligible reports and excluded those studies
that did not meet all of the inclusion criteria. We discussed any
c. cognitive and educational outcomes in survivors aged more than disagreements until we achieved consensus.
five years old.
Data extraction and management
5. Time to establish full enteral feeding (independently of
We used a data collection form to aid extraction of relevant
parenteral nutrition).
information from each included study. Two review authors
6. Time to establish oral feeding (independently of parenteral extracted the data separately. We discussed any disagreements
nutrition or enteral tube feeding, or both). until we reached consensus. We contacted the investigators for
further information if data from the trial reports were insufficient.
7. Feed intolerance (defined as a requirement to cease enteral
feeds). Assessment of risk of bias in included studies
We used the criteria and standard methods of the Cochrane
8. Incidence of invasive infection as determined by culture of
Neonatal Review Group to assess the methodological quality of any
bacteria or fungus from blood, cerebrospinal fluid, urine or from a
included trials (neonatal.cochrane.org/). We requested additional
normally sterile body space.
information from the trial authors to clarify methodology and
9. Duration of hospital stay (days). results as necessary. We evaluated and reported the following
issues in the 'Risk of bias' tables:
Search methods for identification of studies
1. Sequence generation: we categorised the method used to
We used the standard search strategy of the Cochrane Neonatal generate the allocation sequence as:
Review Group (neonatal.cochrane.org/). a. low risk: any random process (e.g. random number table,
computer random number generator);
Electronic searches b. high risk: any non-random process (e.g. odd or even date of
We searched the Cochrane Central Register of Controlled Trials birth, participant case-record number);
(CENTRAL, 2014, Issue 8), MEDLINE (1966 to September 2014), c. unclear risk.
EMBASE (1980 to September 2014) and CINAHL (1982 to September 2. Allocation concealment: we categorised the method used to
2014) using a combination of the following text words and MeSH conceal the allocation sequence as:
terms: [Infant, Newborn OR Infant, Premature OR Infant, Low Birth a. low risk (e.g. telephone or central randomisation,
Weight OR Infant, Very Low Birth Weight/ OR infan* OR neonat* consecutively numbered sealed opaque envelopes);
OR preterm OR prem*] AND "Infant-Nutrition"/ all subheadings OR
b. high risk (e.g. open random allocation; unsealed or non-
Infant Formula OR milk OR formula OR trophic feeding OR minimal
opaque envelopes, alternation; date of birth);
enteral nutrition OR gut priming]. We limited the search outputs
with the relevant search filters for clinical trials as recommended c. unclear risk.
in the Cochrane Handbook for Systematic Reviews of Interventions 3. Blinding: we assessed blinding of participants, clinicians and
(Higgins 2011). We applied no language restrictions. carers, and outcome assessors separately for different outcomes
and categorised the methods as:
We searched ClinicalTrials.gov (clinicaltrials.gov) and Current a. low risk;
Controlled Trials (www.controlled-trials.com/) for completed or b. high risk;
ongoing trials. c. unclear risk.
Searching other resources 4. Incomplete outcome data: we described the completeness of
data including attrition and exclusions from the analysis for
We examined the references in all studies identified as potentially each outcome and any reasons for attrition or exclusion where
relevant. reported. We assessed whether missing data were balanced
across groups or were related to outcomes. Where sufficient
We searched the abstracts from the annual meetings of the
information was reported or supplied by the trial authors,
Pediatric Academic Societies (1993 to 2014), the European Society
we re-included missing data in the analyses. We categorised
for Pediatric Research (1995 to 2014), the UK Royal College of
completeness as:
Paediatrics and Child Health (2000 to 2014) and the Perinatal
a. low risk: less than 20% missing data;
Society of Australia and New Zealand (2000 to 2013). Trials
reported only as abstracts were eligible if sufficient information was b. high risk: 20% or greater missing data;
available from the report, or from contact with the authors, to fulfil c. unclear risk.
the inclusion criteria.
Measures of treatment effect
Data collection and analysis We calculated risk ratio (RR) and risk difference (RD) for
We used the standard methods of the Cochrane Neonatal Review dichotomous data and mean difference (MD) for continuous data,
Group (neonatal.cochrane.org/). with respective 95% confidence intervals (CI). We planned to

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 4
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

determine the number needed to treat for an additional beneficial 3. Davey 1994: 62 clinically stable preterm infants of birth weight
outcome (NNTB) or additional harmful outcome (NNTH) for any less than 2000 g who had a low umbilical artery catheter in situ.
statistically significant differences in the RD. Since most participants were of birth weight less than 1500 g
or gestational age less than 32 weeks, we made a consensus
Unit of analysis issues decision to include the trial.
The unit on analysis was the participating infant in individually
The six more recent trials were performed in the 2000s to 2010s.
randomised trials and the neonatal unit (or sub-unit) for cluster-
randomised trials. 1. Karagianni 2010: single-centre study in Greece, 84 infants less
than 35 weeks' gestation with a birth weight less than 10th
Assessment of heterogeneity
percentile and evidence of abnormal fetal blood flow patterns
If we included more than one trial in a meta-analysis, we examined on Doppler ultrasound of the umbilical artery.
the treatment effects of individual trials and heterogeneity between 2. Pérez 2011: single-centre study in Columbia, 239 very preterm or
trial results by inspecting the forest plots. We calculated the I2 VLBW infants.
statistic for each analysis to quantify inconsistency across studies 3. Leaf 2012: 54-centre trial in the UK and Ireland, 404 infants
and describe the percentage of variability in effect estimates (a) less than 35 weeks' gestation, (b) birth weight less than
that may be due to heterogeneity rather than sampling error. 10th percentile and (c) evidence of abnormal fetal blood flow
If we detected substantial (I2 greater than 50%) heterogeneity, patterns on Doppler ultrasound studies. Since most participants
we explored the possible causes (e.g. differences in study were of birth weight less than 1500 g, we made a consensus
design, participants, interventions or completeness of outcome decision to include the trial.
assessments) in sensitivity analyses. 4. Abdelmaaboud 2012: single-centre study in Qatar, 125 preterm
infants with intrauterine growth restriction and abnormal
Data synthesis
Doppler flow patterns on ultrasound of the umbilical artery.
We used the fixed-effect model in Review Manager 5 for meta- Since most participants were of birth weight less than 1500 g, we
analysis (RevMan 2011). made a consensus decision to include the trial.
5. Armanian 2013: single-centre study in Iran, 82 VLBW infants.
Subgroup analysis and investigation of heterogeneity
6. Arnon 2013: single-centre study in Israel, 60 small for gestation
We planned the following subgroup analyses: age preterm infants. Since most participants were of birth
weight less than 1500 g, we made a consensus decision to
1. trials in which most infants were exclusively formula-fed; include the trial.
2. trials in which most infants were at least partially fed with
human milk (maternal or donor); Interventions/comparisons
3. trials in which most participants were of ELBW (less than 1000 g) 1. Eight trials defined 'delayed' introduction of enteral feeds as
or extremely preterm (less than 28 weeks' gestation); later than day four to seven after birth (Ostertag 1986; Davey
4. trials in which participants were infants with intrauterine growth 1994; Karagianni 2010; Pérez 2011; Abdelmaaboud 2012; Leaf
restriction, or infants with absent or reversed end-diastolic flow 2012; Armanian 2013; Arnon 2013) and one trial defined it as day
velocities detected on antenatal Doppler studies of the fetal 10 after birth (Khayata 1987).
aorta or umbilical artery. 2. 'Early' feeding varied from day one to four after birth.
RESULTS In seven trials, infants received breast milk, artificial formula or
a combination of the two (Davey 1994; Karagianni 2010; Pérez
Description of studies 2011; Abdelmaaboud 2012; Leaf 2012; Armanian 2013; Arnon 2013).
We identified 13 reports for screening. In two trials, infants received only formula-feed (Ostertag 1986;
Khayata 1987). Infants received enteral feeds by gavage at one-
Included studies or two-hourly intervals in all of the trials except Ostertag 1986,
where infants received feeds by continuous intragastric infusion. In
Nine trials fulfilled the review eligibility criteria: Ostertag Ostertag 1986, infants were initially fed with a sterile water infusion
1986; Khayata 1987; Davey 1994; Karagianni 2010; Pérez 2011; slowly progressing to a 2.5% dextrose solution followed by half-
Abdelmaaboud 2012; Leaf 2012; Armanian 2013; Arnon 2013 (see strength formula. They reached full-strength formula milk seven
Characteristics of included studies table). days after initiating enteral feeds.
Population All of the trial protocols, except that of the smallest trial (Khayata
A total of 1106 infants participated in the included trials. 1987), specified criteria and indications for advancing (daily
increments of 15 to 30 mL/kg) or interrupting enteral feed (e.g.
The three smallest trials were undertaken in neonatal care centres residual gastric contents not greater than 3 to 5 mL or one-
in North America during the 1980s and early 1990s. third to one-half of the previous feed volume, frequent vomiting,
abdominal distention or detection of blood in the stools).
1. Ostertag 1986: 38 VLBW infants assessed to be at high risk of
developing NEC. Outcomes
2. Khayata 1987: 12 VLBW infants. Eight of the trials reported the incidence of NEC (confirmed
radiologically, or at surgery or autopsy) (Ostertag 1986; Davey
Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 5
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

1994; Karagianni 2010; Pérez 2011; Abdelmaaboud 2012; Leaf Sanghvi 2013; Chetry 2014) (see Characteristics of excluded studies
2012; Armanian 2013; Arnon 2013). The other reported outcomes table).
included mortality, time to establish full enteral feeding, growth
and duration of hospital stay. Only two trials reported the incidence Risk of bias in included studies
of invasive infection (Leaf 2012; Arnon 2013).
Quality assessments are described in the Characteristics of
Excluded studies included studies table and summarised in Figure 1.

We excluded eight studies after full-text screening (Higgs 1974;


Glass 1984; LaGamma 1985; Wilson 1997; Weiler 2006; Said 2008;
 
Figure 1.   'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages
across all included studies.

 
The smallest trial (12 infants) was reported in abstract form only Effects of interventions
and methodological details were not described (Khayata 1987).
Primary outcomes
The other trials had various methodological weaknesses. In five Necrotising enterocolitis (Outcome 1.1)
trials, methods to ensure adequate allocation concealment were
not described. None of the trials concealed the feeding strategies Meta-analysis did not detect a statistically significant effect (typical
from parents, carers or clinical investigators. Complete or near- RR 0.93, 95% CI 0.64 to 1.34; typical RD -0.01, 95% CI -0.04 to
complete assessments of the primary outcomes were reported and 0.03; 8 trials; 1092 infants). There was no statistical evidence of
data were available to undertake intention-to-treat analyses as heterogeneity (Figure 2).
required.
 

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 6
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Figure 2.   Forest plot of comparison: 1 Delayed versus early introduction of progressive enteral feeding, outcome:
1.1 Necrotising enterocolitis.

 
Mortality prior to discharge (Outcome 1.2) 0.04; 7 trials, 967 infants). There was no statistical evidence of
heterogeneity (Figure 3).
Meta-analysis did not detect a statistically significant effect (typical
RR 1.18, 95% CI 0.75 to 1.88; typical RD 0.01, 95% CI -0.02 to
 
Figure 3.   Forest plot of comparison: 1 Delayed versus early introduction of progressive enteral feeding, outcome:
1.2 Mortality prior to discharge.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 7
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Secondary outcomes Time to establish full enteral feeding


Growth The median time to establish full enteral feeding was longer in the
delayed introduction group but the reports did not provide data to
Two trials did not detect a statistically significant difference in the
allow quantitative synthesis:
median time to regain birth weight:
1. Ostertag 1986: not reported;
1. Davey 1994: 13 days for both groups (range not reported; 62
infants); 2. Khayata 1987: not reported;
2. Abdelmaaboud 2012: 13 days in the delayed group compared 3. Davey 1994: three days;
with 14 days in the early introduction group (range not reported; 4. Karagianni 2010: three days;
125 infants). 5. Pérez 2011: four days;
6. Abdelmaaboud 2012: two days;
Two trials did not detect statistically significant differences in the
rate of weight gain but the reports did not provide data to allow 7. Leaf 2012: three days;
quantitative synthesis (Khayata 1987; Pérez 2011). 8. Armanian 2013: five days;
9. Arnon 2013: three days.
None of the other trials reported hospital growth parameters.
Time to establish full oral feeding
None of the trials assessed any long-term (post-hospital discharge)
growth parameters. None of the trials assessed time to establish full oral feeding.

Neurodevelopment Feed intolerance (Outcome 1.3)

None of the trials assessed neurodevelopmental outcomes. Meta-analysis of data from three trials did not detect a statistically
significant difference in feed intolerance (typical RR 0.84, 95% CI
0.62 to 1.15; typical RD -0.06, 95% CI -0.17 to 0.05; 288 infants; Figure
4) (Karagianni 2010; Abdelmaaboud 2012; Armanian 2013).
 
Figure 4.   Forest plot of comparison: 1 Delayed versus early introduction of progressive enteral feeding, outcome:
1.3 Feed intolerance.

 
One trial did not detect a statistically significant difference but the Duration of hospital stay (Outcome 1.5)
report did not provide data to allow quantitative synthesis (Davey
Meta-analysis from three trials showed a statistically significant
1994).
longer duration in the delayed feeding group (MD 2.11 days, 95%
None of the other trials reported the incidence of feed intolerance. CI 0.31 to 3.90; 346 infants; Figure 5). The meta-analysis contained
substantial heterogeneity (Chi2 = 7.66; degrees of freedom (df) = 2;
Incidence of invasive infection (Outcome 1.4) P value = 0.02; I2 = 74%).
Meta-analysis of data from two trials did not detect a statistically
significant difference (typical RR 1.27, 95% CI 0.95 to 1.70; typical
RD 0.07, 95% CI -0.01 to 0.15; 457 infants) (Leaf 2012; Arnon 2013).
 

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 8
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Figure 5.   Forest plot of comparison: 1 Delayed versus early introduction of progressive enteral feeding, outcome:
1.5 Duration of hospital admission (days).

 
Another two trials did not detect a statistically significant effect but Overall completeness and applicability of evidence
the reports did not provide data to allow quantitative synthesis
(Abdelmaaboud 2012; Leaf 2012). These data are relevant to current practice since most of
the included trials were conducted in the 2000s with infants
Subgroup analyses receiving 'modern' antenatal care including exposure to antenatal
corticosteroids and exogenous surfactant, interventions that
1. Two trials only recruited exclusively formula-fed infants reduce the risk of NEC or death in this population (Roberts 2006;
(Ostertag 1986; Khayata 1987). Only Ostertag 1986 reported the Seger 2009; Soll 2009; Soll 2010). Four of these trials specifically
effect on NEC (RR 1.08, 95% CI 0.40 to 2.94; RD 0.02, 95% CI -0.27 recruited infants thought to be at higher risk of developing NEC due
to 0.31; 38 infants) or mortality (RR 1.44, 95% CI 0.57 to 3.61; RD to intrauterine growth restriction and abnormal fetal circulatory
0.12, 95% CI -0.18 to 0.42; 38 infants). distribution or flow. This widens the applicability of the findings
2. Trials in which most infants were at least partially fed with since this is the population for which most clinical uncertainty
human milk (maternal or donor): subgroup data not available. and variation in practice with regard to early feeding strategies
3. ELBW or extremely preterm infants: none of the trials recruited exists (Boyle 2004). Previously, this population of infants has been
predominantly ELBW or extremely preterm infants. specifically excluded from participating in many trials of early
4. Four trials recruited only infants with intrauterine growth enteral feeding practices (Tyson 2007).
restriction and abnormal flow velocities detected on antenatal
Evidence exists that artificial formula feeding increases the risk
Doppler studies (Karagianni 2010; Abdelmaaboud 2012; Leaf
NEC (Quigley 2014). The risk-benefit balance of enteral feeding
2012; Arnon 2013). Meta-analysis did not detect any statistically
strategies may differ between human milk-fed and formula-fed very
significant differences in the incidence of NEC (typical RR 0.87,
preterm or VLBW infants. Currently there are insufficient data to
95% CI 0.54 to 1.41; typical RD -0.01, 95% CI -0.06 to 0.03; 673
comment on whether there is a differential effect of the timing
infants; Figure 2) or mortality (typical RR 1.06, 95% CI 0.55 to
of the introduction of enteral feeds depending on whether infants
2.05; typical RD 0.00, 95% CI -0.04 to 0.05; 548 infants; Figure 3).
received human breast milk versus formula.
DISCUSSION
It is also unclear whether the findings can be applied to infants
who receive continuous infusion of intragastric feeds, as most
Summary of main results
of the infants in the included trials received enteral feeds
Analyses of data from nine randomised controlled trials with 1106 as interval gastric boluses. Randomised controlled trials have
infants did not provide evidence that delayed introduction of reported conflicting findings about the effect on continuous enteral
progressive enteral feeds reduced the risk of NEC. The boundaries infusion on feed tolerance in VLBW (and especially ELBW) infants
of the 95% CI for the estimate of effect are consistent with either (Premji 2011).
four fewer or three extra cases of NEC in every 100 infants who had
delayed introduction of progressive enteral feeds. Meta-analysis Most of the included trials were undertaken in neonatal care
of data from these trials did not indicate an effect on all-cause centres in middle- or high-income countries. It is less clear how
mortality with the 95% CI boundaries being consistent with either applicable this evidence is to neonatal care practices in low-income
two fewer or four more deaths in every 100 infants who had countries. Conservative strategies, such as delayed introduction of
delayed introduction of progressive enteral feeds. Similarly, pre- enteral feeds, may confer substantial nutritional disadvantage in
specified subgroup meta-analyses showed no effect of delayed settings with less technologically developed healthcare provision
introduction of enteral feeds on NEC or mortality in infants with where adjunctive parenteral nutrition is not readily and safely
growth restriction or antenatal evidence of absent end-diastolic available. In some low- or middle-income countries where severe
flow velocities. infection (diarrhoea, pneumonia, septicaemia) is a much more
important cause of mortality and morbidity, the nutritional and
Infants who had delayed introduction of feeds achieved full enteral immunological advantages of early feeding, particularly with breast
feeding several days later than infants who had earlier introduction. milk, may outweigh any risks associated with enteral feeding for
Whether this was associated with important clinical adverse very preterm or VLBW infants (Narayanan 1981; de Silva 2004). We
consequences, such as a higher rate of nosocomial infection identified two feasibility trials undertaken in India in the late 2000s/
secondary to prolonged use of parenteral nutrition or a longer early 2010s that compared exclusive enteral feeding (no parenteral
duration of hospital admission, remains unclear. fluid) from birth with gradual introduction of enteral feeds over
several days in VLBW infants with birth weight greater than 1000 g
Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 9
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

(Sanghvi 2013; Chetry 2014). While these trials were not eligible for AUTHORS' CONCLUSIONS
inclusion in this review, neither found evidence of an effect on NEC
or other adverse outcomes. Implications for practice

Quality of the evidence The available data from randomised controlled trials do not
provide evidence that delaying the introduction of progressive
The included trials were generally of reasonable methodological enteral feeds beyond four days after birth reduces the risk of
quality but, in common with other trials of feeding interventions necrotising enterocolitis (NEC), mortality, and other morbidities
in this population, it was not possible to mask carers and clinical in very preterm or very low birth weight (VLBW) infants. Delaying
assessors to the nature of the intervention. Although the lack the introduction of progressive enteral feeds may result in several
of blinding may have resulted in surveillance and ascertainment days' delay in establishing full enteral feeds but the long-term
biases, this is more likely to have caused an underestimation of the clinical importance of these effects is unclear. Subgroup analyses
incidence of NEC in infants whose enteral feeding was delayed. The of trials in which participating infants had evidence of intrauterine
assessment of abdominal radiographs was masked in three studies growth restriction or abnormal circulatory distribution or flow did
to ensure that the diagnosis of stage II/III NEC (confirmed by the not find any statistically significant effects. However, only limited
radiological detection of gas in the bowel wall or portal tract) was data are available on the effect of this intervention on outcomes for
not prone to bias. However, since the microbial generation of gas in extremely preterm or extremely low birth weight (ELBW) infants.
the bowel wall is substrate dependent, infants who received more
enteral milk (substrate) may have been more likely to demonstrate Implications for research
this radiological sign than infants with equally severe bowel disease
Further randomised controlled trials of delayed versus early
who had less intraluminal substrate. This 'substrate effect' is also
introduction of progressive enteral feeds could provide more
more likely to cause under-ascertainment of NEC in the infants
precise estimates of the effects on important outcomes for
whose enteral feeding was delayed (Tyson 2007).
extremely preterm or ELBW infants. With regard to stable VLBW
Potential biases in the review process infants with birth weight greater than 1000 g (or very preterm
infants with gestational age 28 to 31 weeks), the key research
The definition of delayed introduction of progressive feeds may question is now whether exclusive enteral feeding from birth is
vary between different subpopulations of very preterm or VLBW better than gradual introduction.
infants who have different empiric risks for developing feed
intolerance and NEC. The effects of enteral feeding are likely to be Masking carers and investigators to the nature of this intervention
very different for a mechanical ventilator-dependent or inotrope- is unlikely to be possible. Since the unblinded evaluation of feed
dependent infant of birth weight less than 700 g compared with intolerance and NEC is subject to surveillance and ascertainment
a clinically stable infant of birth weight greater than 1400 g. For biases, trials could aim to assess more objective outcomes,
this Cochrane review, we defined delayed introduction as later principally mortality and long-term growth and development.
than four days after birth since some observational studies have Furthermore, since conservative feeding strategies may result in
found the risk of NEC to be lower when feeds are introduced five to other 'competing outcomes', such as invasive infection that may
seven days after birth (Patole 2005). For ELBW or extremely preterm affect long-term survival and neuro-disability rates, it is essential
infants, it may be more appropriate to define delayed introduction that trials are powered and structured to assess these outcomes.
as more than seven days after birth (or even later). Small-intestinal
motility is poorly organised before about 28 weeks' gestation ACKNOWLEDGEMENTS
resulting in a higher risk of feed intolerance. In addition, enteral
feeds are often delayed in this population because of respiratory This report is independent research funded by a UK National
or metabolic instability or because of other putative risk factors Institute of Health Research Grant (NIHR) Cochrane Programme
for NEC, such as the existence of a patent ductus arteriosus, the Grant (13/89/12). The views expressed in this publication are those
use of non-steroidal anti-inflammatory drugs or the presence of an of the authors and not necessarily those of the National Health
umbilical arterial catheter (Boyle 2004). Service (NHS), the NIHR or the UK Department of Health.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 10
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

REFERENCES
 
References to studies included in this review * Leaf A, Dorling J, Kempley S, McCormick K, Mannix P, Linsell L,
et al. on behalf of ADEPT Clinical Investigators Group. Early or
Abdelmaaboud 2012 {published data only}
delayed enteral feeding for preterm growth-restricted infants:
Abdelmaaboud M, Mohammed A. A randomized controlled trial a randomized trial. Pediatrics 2012;129(5):1-9. [DOI: 10.1542/
on early versus late minimal enteral feeding in preterm growth- peds.2011-2379; PUBMED: 22492770]
restricted neonates with abnormal antenatal Doppler studies.
Journal of Neonatal-Perinatal Medicine 2012;5(2):1-8. Leaf A,  Dorling J,  Kempley S,  McCormick K,  Mannix P,
Brocklehurst P. Abnormal Doppler enteral prescription
Armanian 2013 {published and unpublished data} trial study: the results of a trial of feeding in a high risk
Armanian AM, Mirbod SM, Kazemipour S, Hassanzade A. group of premature babies. Archives of Disease in Childhood.
Comparison of prolonged low volume milk and routine volume Fetal and Neonatal Edition 2010;95:Fa9. [DOI: 10.1136/
milk on incidence of necrotizing enterocolitis in very low birth adc.2010.192310.4.1]
weight neonates. Pakistan Journal of Medical Sciences 2013;29(1
Suppl):312-6. Ostertag 1986 {published and unpublished data}
Ostertag SG, LaGamma EF, Reisen CE, Ferrentino FL. Early
Arnon 2013 {published data only} enteral feeding does not affect the incidence of necrotizing
Arnon S, Sulam D, Konikoff F, Regev RH, Litmanovitz I, Naftali T. enterocolitis. Pediatrics 1986;77(3):275-80. [PUBMED: 3081868]
Very early feeding in stable small for gestational age preterm
infants: a randomized clinical trial. Jornal de Pediatria Pérez 2011 {published data only}
2013;89(4):388-93. Pérez LA, Pradilla GL, Díaz G, Bayter SM. Necrotising
enterocolitis among preterm newborns with early feeding
Davey 1994 {published data only (unpublished sought but not [Incidencia de enterocolitis necrosante en niños prematuros
used)} alimentados precozmente]. Biomédica 2011;31(4):485-91. [DOI:
Davey AM, Wagner CL, Cox C, Kendig JW. Feeding premature 10.1590/S0120-41572011000400003; PUBMED: 22674359]
infants while low umbilical artery catheters are in place: a
prospective, randomized trial. Journal of Pediatrics 1994;124(5  
Pt 1):795-9. [PUBMED: 8176571] References to studies excluded from this review
Chetry 2014 {published data only}
Karagianni 2010 {published data only}
Chetry S, Kler N, Saluja S, Garg P, Soni A, Thakur A, et al. A
Karagianni P, Briana DD, Mitsiakos G, Elias A, Theodoridis T,
randomised control trial comparing initiation of total enteral
Chatziioannidis E, et al. Early versus delayed minimal enteral
feeds on 1st day of life with standard feeding regimen in stable
feeding and risk for necrotizing enterocolitis in preterm growth-
very low birth weight infants born between > 30-34 weeks
restricted infants with abnormal antenatal Doppler results.
gestation and 1000-1500 gms. Pediatric Academic Societies
American Journal of Perinatology 2010;27(5):367-73. [DOI:
[2930.442]. 2014.
10.1055/s-0029-1243310; PUBMED: 20013579]
Glass 1984 {published data only}
Khayata 1987 {published data only (unpublished sought but not
used)} Glass EJ, Hume R, Lang MA, Forfar JO. Parenteral nutrition
compared with transpyloric feeding. Archives of Disease in
Khayata S, Gutcher G, Bamberger J, Heimler R. Early versus late
Childhood 1984;59(2):131-5. [PUBMED: 6422864]
feeding of low birth weight (LBW) infants: effect on growth and
hyperbilirubinemia. Pediatric Research 1987;21:431A. Higgs 1974 {published data only}
Leaf 2012 {published data only} Higgs SC, Malan AF, De Heese HV. A comparison of oral feeding
and total parenteral nutrition in infants of very low birthweight.
Kempley S, Gupta N, Linsell L, Dorling J, McCormick K,
South African Medical Journal 1974;48(52):2169-73. [PUBMED:
Mannix P. Feeding infants below 29 weeks' gestation with
4215158]
abnormal antenatal Doppler: analysis from a randomised trial.
Archives of Disease in Childhood. Fetal and Neonatal Edition LaGamma 1985 {published data only}
2014;99(1):F6-11. [PUBMED: 23973795]
LaGamma EF, Ostertag S, Birenbaum H. Failure of delayed oral
Leaf A, Dorling J, Kempley S, McCormick K, Mannix P, feedings to prevent necrotizing enterocolitis. Results of study in
Brocklehurst P. ADEPT - Abnormal Doppler Enteral Prescription very-low-birth-weight neonates. American Journal of Diseases of
Trial. BMC Pediatrics 2009;9:63. [DOI: 10.1186/1471-2431-9-63; Children 1985;139(4):385-9. [PUBMED: 3919570]
PUBMED: 19799788]
Said 2008 {published data only}
Leaf A, Dorling J, Kempley S, McCormick K, Mannix P, Said H, Elmetwally D, Said N. Randomized controlled trial of
Brocklehurst P. When should feeds be started in the high risk early versus late enteral feeding in prematurely born infants
preterm infant? The Abnormal Doppler Enteral Prescription Trial with birth weight ≤ 1200 grams. Kasr El Aini Medical Journal
(ADEPT). E-PAS20101670.7. 2010. 2008;14:1-10.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 11
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Sanghvi 2013 {published data only} de Silva 2004


Sanghvi KP, Joshi P, Nabi F, Kabra N. Feasibility of exclusive de Silva A, Jones PW, Spencer SA. Does human milk reduce
enteral feeds from birth in VLBW infants >1200 g - an RCT. Acta infection rates in preterm infants? A systematic review.
Paediatrica 2013;102(7):e299-304. [DOI: 10.1111/apa.12254; Archives of Disease in Childhood. Fetal and Neonatal Edition
PUBMED: 23621289] 2004;89(6):F509-13. [PUBMED: 15499143]

Weiler 2006 {published data only} Dorling 2005


Weiler HA, Fitzpatrick-Wong SC, Chellenberg JM, Fair DE, Dorling J, Kempley S, Leaf A. Feeding growth restricted
McCloy UR, Veitch RR, et al. Minimal enteral feeding within 3 d of preterm infants with abnormal antenatal Doppler results.
birth in prematurely born infants with birth weight < or = 1200g Archives of Disease in Childhood. Fetal and Neonatal Edition
improves bone mass by term age. American Journal of Clinical 2005;90(5):F359-63. [PUBMED: 16113150]
Nutrition 2006;83(1):155-62. [PUBMED: 16403735]
Flidel-Rimon 2004
Wilson 1997 {published data only} Flidel-Rimon O, Friedman S, Lev E, Juster-Reicher A, Amitay M,
Wilson DC, Cairns P, Halliday HL, Reid M, McClure G, Dodge JA. Shinwell ES. Early enteral feeding and nosocomial sepsis in very
Randomised controlled trial of an aggressive nutritional low birthweight infants. Archives of Disease in Childhood. Fetal
regimen in sick very low birthweight infants. Archives of Disease and Neonatal Edition 2004;89(4):F289-92. [PUBMED: 15210657]
in Childhood. Fetal and Neonatal Edition 1997;77(1):F4-11.
[PUBMED: 9279175] Flidel-Rimon 2006
Flidel-Rimon O, Branski D, Shinwell ES. The fear of necrotizing
  enterocolitis versus achieving optimal growth in preterm infants
Additional references - an opinion. Acta Paediatrica 2006;95(11):1341-4. [PUBMED:
Bernstein 2000 17062457]
Bernstein IM, Horbar JD, Badger GJ, Ohlsson A, Golan A. Garite 2004
Morbidity and mortality among very-low-birth-weight neonates
Garite TJ, Clark R, Thorp JA. Intrauterine growth restriction
with intrauterine growth restriction. The Vermont Oxford
increases morbidity and mortality among premature
Network. American Journal of Obstetrics and Gynecology
neonates. American Journal of Obstetrics and Gynecology
2000;182(1 Pt 1):198-206. [PUBMED: 10649179]
2004;191(2):481-7. [PUBMED: 15343225]
Berrington 2012
Hartel 2009
Berrington JE, Hearn RI, Bythell M, Wright C, Embleton ND.
Hartel C, Haase B, Browning-Carmo K, Gebauer C, Kattner E,
Deaths in preterm infants: changing pathology over 2 decades.
Kribs A, et al. Does the enteral feeding advancement affect
Journal of Pediatrics 2012;160(1):49-53. [DOI: 10.1016/
short-term outcomes in very low birth weight infants?. Journal
j.jpeds.2011.06.046; PUBMED: 21868028]
of Pediatric Gastroenterology and Nutrition 2009;48(4):464-70.
Berseth 1990 [DOI: 10.1097/MPG.0b013e31818c5fc3; PUBMED: 19322056]
Berseth CL. Neonatal small intestinal motility: motor responses Hay 2008
to feeding in term and preterm infants. Journal of Pediatrics
Hay WW Jr. Strategies for feeding the preterm infant.
1990;117(5):777-82. [PUBMED: 2121949]
Neonatology 2008;94(4):245-54. [DOI: 10.1159/000151643;
Bisquera 2002 PUBMED: 18836284]
Bisquera JA, Cooper TR, Berseth CL. Impact of necrotizing Henderson 2009
enterocolitis on length of stay and hospital charges in very low
Henderson G, Craig S, Brocklehurst P, McGuire W. Enteral
birth weight infants. Pediatrics 2002;109(3):423-8. [PUBMED:
feeding regimens and necrotising enterocolitis in preterm
11875136]
infants: a multicentre case-control study. Archives of Disease
Boyle 2004 in Childhood. Fetal and Neonatal Edition 2009;94(2):F120-3.
[PUBMED: 17768154]
Boyle EM, Menon G, Elton R, McIntosh N. Variation in feeding
practice in preterm and low birth weight infants in Scotland. Hershkovitz 2000
Early Human Development 2004;77:125-6.
Hershkovitz R, Kingdom JC, Geary M, Rodeck CH. Fetal cerebral
Brown 1978 blood flow redistribution in late gestation: identification
of compromise in small fetuses with normal umbilical
Brown EG, Sweet AY. Preventing necrotizing enterocolitis in
artery Doppler. Ultrasound in Obstetrics & Gynecology
neonates. JAMA 1978;240(22):2452-4. [PUBMED: 101680]
2000;15(3):209-12. [PUBMED: 10846776]
Burrin 2002
Higgins 2011
Burrin DG, Stoll B. Key nutrients and growth factors for
Higgins JPT, Green S, editors. 2011. Cochrane Handbook for
the neonatal gastrointestinal tract. Clinics in Perinatology
Systematic Reviews of Interventions Version 5.1.0 [updated
2002;29(1):65-96. [PUBMED: 11917740]
March 2011]. The Cochrane Collaboration, 2011. Available from
www.cochrane-handbook.org.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 12
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Holman 2006 RevMan 2011 [Computer program]


Holman RC, Stoll BJ, Curns AT, Yorita KL, Steiner CA, Nordic Cochrane Centre. The Cochrane Collaboration. Review
Schonberger LB. Necrotising enterocolitis hospitalisations Manager (RevMan). Version 5.1. Copenhagen: Nordic Cochrane
among neonates in the United States. Paediatric and Perinatal Centre. The Cochrane Collaboration, 2011.
Epidemiology 2006;20(6):498-506. [PUBMED: 17052286]
Roberts 2006
Kamoji 2008 Roberts D, Dalziel S. Antenatal corticosteroids for accelerating
Kamoji VM, Dorling JS, Manktelow B, Draper ES, Field DJ. fetal lung maturation for women at risk of preterm birth.
Antenatal umbilical Doppler abnormalities: an independent Cochrane Database of Systematic Reviews 2006, Issue 3. [DOI:
risk factor for early onset neonatal necrotizing enterocolitis 10.1002/14651858.CD004454.pub2]
in premature infants. Acta Paediatrica 2008;97(3):327-31.
[10.1111/j.1651-2227.2008.00671.x; PUBMED: 18298781] Seger 2009
Seger N, Soll R. Animal derived surfactant extract for
Klingenberg 2012 treatment of respiratory distress syndrome. Cochrane
Klingenberg C, Embleton ND, Jacobs SE, O'Connell LA, Database of Systematic Reviews 2009, Issue 2. [DOI:
Kuschel CA. Enteral feeding practices in very preterm infants: 10.1002/14651858.CD007836]
an international survey. Archives of Disease in Childhood.
Fetal and Neonatal Edition 2012;97(1):F56-61. [DOI: 10.1136/ Soll 2009
adc.2010.204123; PUBMED: 21856644] Soll R, Ozek E. Multiple versus single doses of exogenous
surfactant for the prevention or treatment of neonatal
Morgan 2013a respiratory distress syndrome. Cochrane Database of Systematic
Morgan J, Bombell S, McGuire W. Early trophic feeding versus Reviews 2009, Issue 1. [DOI: 10.1002/14651858.CD000141.pub2]
enteral fasting for very preterm or very low birth weight infants.
Cochrane Database of Systematic Reviews 2013, Issue 3. [DOI: Soll 2010
10.1002/14651858.CD000504.pub3] Soll R, Ozek E. Prophylactic protein free synthetic surfactant
for preventing morbidity and mortality in preterm infants.
Narayanan 1981 Cochrane Database of Systematic Reviews 2010, Issue 1. [DOI:
Narayanan I, Prakash K, Gujral VV. The value of human milk in 10.1002/14651858.CD001079.pub2]
the prevention of infection in the high-risk low-birth-weight
infant. Journal of Pediatrics 1981;99(3):496-8. [PUBMED: Stoll 2004
6790690] Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz SR,
Vohr B, et al. Neurodevelopmental and growth impairment
Patole 2004 among extremely low-birth-weight infants with neonatal
Patole S, Muller R. Enteral feeding of preterm neonates: a infection. JAMA 2004;292(19):2357-65. [PUBMED: 15547163]
survey of Australian neonatologists. Journal of Maternal-Fetal &
Neonatal Medicine 2004;16(5):309-14. [PUBMED: 15621549] Tyson 2007
Tyson JE, Kennedy KA, Lucke JF, Pedroza C. Dilemmas initiating
Patole 2005 enteral feedings in high risk infants: how can they be resolved?.
Patole SK, de Klerk N. Impact of standardised feeding Seminars in Perinatology 2007;31(2):61-73. [PUBMED: 17462490]
regimens on incidence of neonatal necrotising enterocolitis: a
systematic review and meta-analysis of observational studies. Uauy 1991
Archives of Disease in Childhood. Fetal and Neonatal Edition Uauy RD, Fanaroff AA, Korones SB, Phillips EA, Phillips JB,
2005;90(2):F147-51. [PUBMED: 15724039] Wright LL. Necrotizing enterocolitis in very low birth weight
infants: biodemographic and clinical correlates. National
Premji 2011 Institute of Child Health and Human Development Neonatal
Premji SS, Chessell L. Continuous nasogastric milk feeding Research Network. Journal of Pediatrics 1991;119(4):630-8.
versus intermittent bolus milk feeding for premature infants [PUBMED: 1919897]
less than 1500 grams. Cochrane Database of Systematic Reviews
2011, Issue 11. [DOI: 10.1002/14651858.CD001819.pub2] Walsh 1986
Walsh MC, Kliegman RM. Necrotizing enterocolitis: treatment
Quigley 2014 based on staging criteria. Pediatric Clinics of North America
Quigley MA, McGuire W. Formula versus donor breast milk 1986;33(1):179-201. [PUBMED: 3081865]
for feeding preterm or low birth weight infants. Cochrane
Database of Systematic Reviews 2014, Issue 4. [DOI:  
10.1002/14651858.CD002971.pub2] References to other published versions of this review
Bombell 2008
Rees 2007
Bombell S, McGuire W. Delayed introduction of progressive
Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes
enteral feeds to prevent necrotising enterocolitis in very low
of neonates with medically and surgically treated necrotizing
birth weight infants. Cochrane Database of Systematic Reviews
enterocolitis. Archives of Disease in Childhood. Fetal and
2008, Issue 2. [DOI: 10.1002/14651858.CD001970.pub2]
Neonatal Edition 2007;92(3):F193-8. [PUBMED: 16984980]

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 13
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Kennedy 2005 very low birth weight infants. Cochrane Database of Systematic
Kennedy KA, Tyson JE, Chamnanvanikij S. Early versus Reviews 2011, Issue 3. [DOI: 10.1002/14651858.CD001970.pub3]
delayed initiation of progressive enteral feedings for
Morgan 2013b
parenterally fed low birth weight or preterm infants.
Cochrane Database of Systematic Reviews 2005, Issue 1. [DOI: Morgan J, Young L, McGuire W. Delayed introduction of
10.1002/14651858.CD001970.pub2] progressive enteral feeds to prevent necrotising enterocolitis in
very low birth weight infants. Cochrane Database of Systematic
Morgan 2011 Reviews 2013, Issue 5. [DOI: 10.1002/14651858.CD001970.pub4]
Morgan J, Young L, McGuire W. Delayed introduction of
 
progressive enteral feeds to prevent necrotising enterocolitis in
* Indicates the major publication for the study
 
CHARACTERISTICS OF STUDIES

Characteristics of included studies [ordered by study ID]


 
Abdelmaaboud 2012 
Methods Randomised controlled trial

Participants Preterm infants, 28-36 weeks' gestation with birth weight < 10th centile, antenatal ultrasound showing
intrauterine growth restriction, absent or reversed end diastolic flow on Doppler waveforms of the um-
bilical artery with evidence of cerebral redistribution, arterial cord pH ≥ 7.0 and base deficit ≥ -12 and 5-
minute Apgar score of > 5.

Infants were excluded if there was any major congenital abnormality, twin-twin transfusion, intrauter-
ine transfusion, exchange transfusion, rhesus iso-immunisation, significant multi-organ failure, in-
otropic drug support or minimal enteral feeding had already started

Setting: single centre: Women's Hospital, Hamad Medical Corporation, Qatar

Interventions Early introduction of progressive enteral feeds on day 3 (62 infants) versus late introduction of enteral
feeds on day 6 (63 infants)

Outcomes Incidence of NEC (stage II/III), time to reach full enteral feeds (sustained for 72 hours), rates of feed in-
tolerance, mortality and duration of hospital stay

Notes > 90% of participants were VLBW

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Low risk Computer-generated tables


tion (selection bias)

Allocation concealment Unclear risk Not stated


(selection bias)

Blinding (performance High risk Carers not blinded to intervention group


bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk No information on blinding of radiological assessors to intervention groups
bias and detection bias)
Radiological assessments

Incomplete outcome data Low risk Complete follow-up assessment


(attrition bias)
All outcomes

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 14
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

 
 
Armanian 2013 
Methods Randomised controlled trial

Participants VLBW infants

Infants were excluded if there was a congenital abnormality

Setting: Isfahan Faculty of Medicine, Iran

Interventions Delayed introduction of progressive enteral feeds (only minimal volumes until day 7 (47 infants) versus
early introduction on day 3 (35 infants)

Infants received either unfortified breast milk or formula (no subgroup data available). Volumes and
rates of advancement or progressive feeds were the same in both groups (20 mL/kg/day)

Outcomes Incidence of NEC, mortality, days to full enteral feeds, duration of hospital stay

Notes  

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Unclear risk Not stated


tion (selection bias)

Allocation concealment Unclear risk Not stated


(selection bias)

Blinding (performance High risk Carers not blinded to intervention group


bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk No information on blinding of radiological assessors to intervention groups
bias and detection bias)
Radiological assessments

Incomplete outcome data Low risk Complete follow-up assessment


(attrition bias)
All outcomes

 
 
Arnon 2013 
Methods Randomised controlled trial

Participants Preterm infants, birth weight < 10th centile*, and antenatal evidence of absent or reversed end dias-
tolic flow on Doppler waveforms of the umbilical artery

Infants were excluded if there was a major congenital abnormality, receipt of mechanical ventilation or
enteral feeding had already started

Setting: single centre: Meir Medical Centre, Kfar Saba, Tel Aviv, Israel

Interventions Delayed progressive enteral feeding (day 4-5 after birth, 30 infants) versus earlier enteral feeding (day 2
after birth, 30 infants)

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 15
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Arnon 2013  (Continued)
Infants received expressed breast or formula or both

Outcomes Incidence of NEC, mortality, nosocomial infection, days to reach full enteral feeds, duration of hospital
stay

Notes *Most participants were VLBW (range 963-1683 g)

Original study published in Portuguese

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Unclear risk Not stated


tion (selection bias)

Allocation concealment Unclear risk Not stated


(selection bias)

Blinding (performance High risk Carers not blinded to intervention group


bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk No information on blinding of radiological assessors to intervention groups
bias and detection bias)
Radiological assessments

Incomplete outcome data Low risk Complete follow-up assessment


(attrition bias)
All outcomes

 
 
Davey 1994 
Methods Randomised controlled trial

Participants 62 preterm infants with birth weight < 2000 g who were clinically stable and who had an umbilical
artery catheter in place

Infants were excluded if they had a lethal condition or had received a double-volume exchange transfu-
sion

Setting: Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, US

Interventions Delayed introduction of enteral feeds (median 5 days; 31 infants) versus earlier introduction (median 2
days; 31 infants)

Infants received either breast milk or diluted formula (no subgroup data available). Volumes and rates
of advancement were the same in both groups

Outcomes Days to regain birth weight, days to full enteral feeding, duration of hospital stay, incidence of NEC and
mortality

Notes The trial inclusion criterion for birth weight was < 2000 g. Since > 80% of infants were VLBW or very
preterm, we decided to include the trial

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 16
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Davey 1994  (Continued)
Infants in the delayed introduction group commenced enteral feeds when the umbilical artery catheter
had been removed for 24 hours and the infant was clinically stable. Infants in the earlier introduction
group commenced feeds with the umbilical artery catheter in situ

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Unclear risk Sequence generation method not reported
tion (selection bias)

Allocation concealment Low risk Sealed opaque envelopes


(selection bias)

Blinding (performance High risk Not stated but unlikely that carers were blinded to intervention groups
bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk Not described


bias and detection bias)
Radiological assessments

Incomplete outcome data Low risk The trial excluded 2 infants in the early introduction group post-randomisation
(attrition bias) due to protocol violation. All other participants were accounted for
All outcomes

 
 
Karagianni 2010 
Methods Randomised controlled trial

Participants 84 singleton newborn infants of gestational age 27- 34 weeks' and birth weight < 10th percentile who
also had antenatal Doppler ultrasound evidence within 7 days before birth of 'pathological fetal perfu-
sion', defined as uterine or umbilical arterial pulsatility index > 90th percentile and middle cerebral ar-
terial pulsatility index < 10th percentile for gestational age

Infants were excluded with a major congenital anomaly or infection or had received exchange transfu-
sion or inotrope support

Setting: Neonatology Department, Aristotle University, Thessaloniki, Greece

Interventions Delayed (> 5 days after birth; 42 infants) versus early (≤ 5 days; 42 infants) introduction of enteral feeds
(expressed breast milk or preterm formula milk)

Minimal enteral feeding was continued until day 7 after birth and then feed volumes were advanced at
daily targeted increments of 15 mL/kg

Outcomes Incidence of NEC, mortality*, days to full enteral feeds*, duration of hospital stay*

Notes *Unpublished data courtesy of Dr Karagianni

Of the 84 infants enrolled, 81 completed the study. 3 infants died before 5 days after birth. We have in-
cluded these infants in the intention-to-treat analysis of mortality > 90% of infants were VLBW

Risk of bias

Bias Authors' judgement Support for judgement

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 17
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Karagianni 2010  (Continued)
Random sequence genera- Low risk Computer-generated sequence
tion (selection bias)

Allocation concealment Low risk Opaque sealed envelopes


(selection bias)

Blinding (performance High risk Carers and clinical assessors not blinded to allocation groups
bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk No information available about blinding of radiological assessors
bias and detection bias)
Radiological assessments

Incomplete outcome data Low risk All data were included in the analyses
(attrition bias)
All outcomes

 
 
Khayata 1987 
Methods Randomised controlled trial

Participants 12 VLBW infants

Interventions Delayed introduction of enteral feeds (day 10 after birth; 7 infants) versus earlier introduction (< 4 days;
5 infants)

All infants received standard calorie formula. Volumes and rates of advancement were the same in both
groups

Outcomes Growth during the first 6 weeks after birth

Notes This trial was reported as an abstract only. Further (unpublished) methodological or outcome data
were not available

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Unclear risk Not described


tion (selection bias)

Allocation concealment Unclear risk Not described


(selection bias)

Blinding (performance High risk Not described but unlikely to be blinded


bias and detection bias)
Clinical assessments

Blinding (performance Unclear risk Not described


bias and detection bias)
Radiological assessments

Incomplete outcome data Unclear risk Not described


(attrition bias)

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 18
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Khayata 1987  (Continued)
All outcomes

 
 
Leaf 2012 
Methods Randomised controlled trial

Participants 404 preterm infants < 35 weeks' gestation and birth weight < 10th percentile and antenatal Doppler ul-
trasound evidence of:

1. absent or reversed end diastolic flow velocities on at least 50% of the Doppler waveforms from the
umbilical artery on at least 1 occasion during pregnancy

or

2. 'cerebral redistribution', defined as occurring when both the umbilical artery pulsatility index > 95th
percentile and the middle cerebral artery pulsatility index < 5th percentile for gestational age (Her-
shkovitz 2000)

Infants were excluded with a major congenital anomaly, receipt of in-utero transfusion, multi-organ
failure or need for inotrope support

Setting: 54 neonatal care centres in UK and Ireland

Interventions Delayed (day 5 after birth; 202 infants) versus early (day 2 after birth; 202 infants) introduction of milk
feeds

Protocol for advancing feed volumes was the same in both groups

Outcomes Days to full feeds (150 mL/kg/day) sustained for 3 days, incidence of NEC (all stages, and stage II/III),
mortality, invasive infection, time to regain birth weight, duration of hospital stay

Notes > 90% of infants were VLBW

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Low risk Computer-generated sequence


tion (selection bias)

Allocation concealment Low risk Central telephone randomisation


(selection bias)

Blinding (performance High risk Carers were not blinded to the allocation groups
bias and detection bias)
Clinical assessments

Blinding (performance Low risk All cases of NEC were reviewed independently by a committee that were blind-
bias and detection bias) ed to the study groups
Radiological assessments

Incomplete outcome data Unclear risk 2 infants (1 from each group) were excluded from the trial after randomisation
(attrition bias) occurred (error in recruitment and consent withdrawal)
All outcomes

 
 

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 19
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Ostertag 1986 
Methods Randomised controlled trial

Participants 38 VLBW infants assessed to be at 'high risk' of developing NEC based on a risk assessment score

Setting: Perinatology Center, New York Hospital-Cornell Medical Center, New York, USA

Interventions Delayed introduction of enteral feeds (day 7 after birth; 20 infants) versus earlier introduction (day 1; 18
infants)

Infants received feeds by continuous intragastric infusion starting initially with sterile water, then pro-
gressing to 2.5% dextrose, diluted formula, then full-strength standard calorie formula milk. Volumes
and rates of advancement were the same in both groups: constant infusion at 1 mL/hour for 7 days
then daily increments of 10 mL/kg/day

Outcomes Incidence of NEC and mortality

Notes Further details about exclusions after randomisation kindly provided by Dr La Gamma (March 2009)

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Low risk Random number table


tion (selection bias)

Allocation concealment Unclear risk Not described


(selection bias)

Blinding (performance High risk Carers not blinded to allocation groups


bias and detection bias)
Clinical assessments

Blinding (performance Low risk Radiologists reviewing abdominal films were blinded to the group assign-
bias and detection bias) ments
Radiological assessments

Incomplete outcome data Low risk 3 infants died before 7 days after birth. The investigators excluded 1 infant be-
(attrition bias) fore day 14 because of a feeding protocol violation. We have included all of
All outcomes these infants in the relevant intention-to-treat analyses

 
 
Pérez 2011 
Methods Randomised controlled trial

Participants 239 very preterm or VLBW infants. Included infants had not received any previous enteral feeds

Infants were excluded with congenital anomalies of the gastrointestinal tract, intrauterine growth re-
striction and respiratory or haemodynamic instability

Setting: Ramón González Valencia de Bucaramanga University Hospital, Columbia

Interventions Delayed enteral feeding (day 5 after birth, 104 infants) versus earlier enteral feeding (day 1-2 after birth,
135 infants)

All infants received a combination of breast and formula milk. Feed volumes exceeded trophic volumes
by the third day of enteral feeding

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 20
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Pérez 2011  (Continued)
Outcomes Incidence of NEC, mortality, duration of hospital stay, growth, days to reach full feeds (150 mL/kg/day)

Notes Original study published in Spanish

Risk of bias

Bias Authors' judgement Support for judgement

Random sequence genera- Unclear risk Not described (states "randomly assigned", and "controlled clinical trial")
tion (selection bias)

Allocation concealment Low risk Not described


(selection bias)

Blinding (performance High risk Carers or investigators not blinded to allocation groups
bias and detection bias)
Clinical assessments

Blinding (performance Low risk Abdominal radiographs interpreted by radiologist who was independent from
bias and detection bias) the study and blind to the allocation groups
Radiological assessments

Incomplete outcome data Low risk Complete follow-up assessment for primary outcomes
(attrition bias)
All outcomes

NEC: necrotising enterocolitis; VLBW: very low birth weight.


 
Characteristics of excluded studies [ordered by study ID]
 
Study Reason for exclusion

Chetry 2014 Both groups received early enteral feeds

Glass 1984 Infants were allocated alternately to either early (first day after birth) or delayed transpyloric enter-
al feeding. The delayed feeding group commenced enteral nutrition when assessed to be "clinically
stable" but this included initiation within 4 days after birth

Higgs 1974 Infants in the delayed progressive enteral feeds group received total parenteral nutrition as a co-in-
tervention

LaGamma 1985 This was not a randomised controlled trial

Said 2008 Infants in the delayed progressive enteral feeding group received minimal enteral nutrition prior to
feed advancement as a co-intervention

Sanghvi 2013 Both groups received early enteral feeds

Weiler 2006 Infants in both groups received some enteral feeds before 4 days after birth

Wilson 1997 Infants in the delayed progressive enteral feeds group also received delayed advancement of par-
enteral nutrition as a co-intervention

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 21
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

 
DATA AND ANALYSES
 
Comparison 1.   Delayed versus early introduction of progressive enteral feeding

Outcome or subgroup title No. of No. of Statistical method Effect size


studies partici-
pants

1 Necrotising enterocolitis 8   Risk Ratio (M-H, Fixed, 95% CI) Subtotals only

1.1 All trials 8 1092 Risk Ratio (M-H, Fixed, 95% CI) 0.93 [0.64, 1.34]

1.2 Trials of infants with intrauterine 4 673 Risk Ratio (M-H, Fixed, 95% CI) 0.87 [0.54, 1.41]
growth restriction or abnormal ante-
natal Doppler flow velocities

2 Mortality prior to discharge 7   Risk Ratio (M-H, Fixed, 95% CI) Subtotals only

2.1 All trials 7 967 Risk Ratio (M-H, Fixed, 95% CI) 1.18 [0.75, 1.88]

2.2 Trials of infants with intrauterine 3 548 Risk Ratio (M-H, Fixed, 95% CI) 1.06 [0.55, 2.05]
growth restriction or abnormal ante-
natal Doppler flow velocities

3 Feed intolerance 3 288 Risk Ratio (M-H, Fixed, 95% CI) 0.84 [0.62, 1.15]

4 Incidence of invasive infection 2 457 Risk Ratio (M-H, Fixed, 95% CI) 1.27 [0.95, 1.70]

5 Duration of hospital admission (days) 3 346 Mean Difference (IV, Fixed, 95% CI) 2.11 [0.31, 3.90]

 
 
Analysis 1.1.   Comparison 1 Delayed versus early introduction of
progressive enteral feeding, Outcome 1 Necrotising enterocolitis.
Study or subgroup Favours Early Risk Ratio Weight Risk Ratio
Delayed
  n/N n/N M-H, Fixed, 95% CI   M-H, Fixed, 95% CI
1.1.1 All trials  
Abdelmaaboud 2012 6/63 8/62 15.38% 0.74[0.27,2]
Armanian 2013 1/35 1/47 1.63% 1.34[0.09,20.74]
Arnon 2013 0/30 0/30   Not estimable
Davey 1994 4/31 2/29 3.94% 1.87[0.37,9.46]
Karagianni 2010 4/42 6/42 11.44% 0.67[0.2,2.19]
Leaf 2012 18/202 18/202 34.33% 1[0.54,1.87]
Ostertag 1986 6/20 5/18 10.04% 1.08[0.4,2.94]
Pérez 2011 9/104 14/135 23.24% 0.83[0.38,1.85]
Subtotal (95% CI) 527 565 100% 0.93[0.64,1.34]
Total events: 48 (Favours Delayed), 54 (Early)  
Heterogeneity: Tau2=0; Chi2=1.5, df=6(P=0.96); I2=0%  
Test for overall effect: Z=0.38(P=0.7)  
   
1.1.2 Trials of infants with intrauterine growth restriction or abnormal  
antenatal Doppler flow velocities
Favours Delayed 0.05 0.2 1 5 20 Favours Early

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 22
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Study or subgroup Favours Early Risk Ratio Weight Risk Ratio


Delayed
  n/N n/N M-H, Fixed, 95% CI   M-H, Fixed, 95% CI
Abdelmaaboud 2012 6/63 8/62 25.15% 0.74[0.27,2]
Arnon 2013 0/30 0/30   Not estimable
Karagianni 2010 4/42 6/42 18.71% 0.67[0.2,2.19]
Leaf 2012 18/202 18/202 56.14% 1[0.54,1.87]
Subtotal (95% CI) 337 336 100% 0.87[0.54,1.41]
Total events: 28 (Favours Delayed), 32 (Early)  
Heterogeneity: Tau2=0; Chi2=0.49, df=2(P=0.78); I2=0%  
Test for overall effect: Z=0.56(P=0.58)  
Test for subgroup differences: Chi2=0.05, df=1 (P=0.83), I2=0%  

Favours Delayed 0.05 0.2 1 5 20 Favours Early

 
 
Analysis 1.2.   Comparison 1 Delayed versus early introduction of
progressive enteral feeding, Outcome 2 Mortality prior to discharge.
Study or subgroup Favours Early Risk Ratio Weight Risk Ratio
Delayed
  n/N n/N M-H, Fixed, 95% CI   M-H, Fixed, 95% CI
1.2.1 All trials  
Armanian 2013 0/35 2/47 7.31% 0.27[0.01,5.39]
Arnon 2013 0/30 0/30   Not estimable
Davey 1994 0/31 1/29 5.28% 0.31[0.01,7.38]
Karagianni 2010 4/42 4/42 13.65% 1[0.27,3.74]
Leaf 2012 13/202 12/202 40.95% 1.08[0.51,2.32]
Ostertag 1986 8/20 5/18 17.96% 1.44[0.57,3.61]
Pérez 2011 8/104 5/135 14.85% 2.08[0.7,6.16]
Subtotal (95% CI) 464 503 100% 1.18[0.75,1.88]
Total events: 33 (Favours Delayed), 29 (Early)  
Heterogeneity: Tau2=0; Chi2=2.94, df=5(P=0.71); I2=0%  
Test for overall effect: Z=0.71(P=0.48)  
   
1.2.2 Trials of infants with intrauterine growth restriction or abnormal  
antenatal Doppler flow velocities
Arnon 2013 0/30 0/30   Not estimable
Karagianni 2010 4/42 4/42 25% 1[0.27,3.74]
Leaf 2012 13/202 12/202 75% 1.08[0.51,2.32]
Subtotal (95% CI) 274 274 100% 1.06[0.55,2.05]
Total events: 17 (Favours Delayed), 16 (Early)  
Heterogeneity: Tau2=0; Chi2=0.01, df=1(P=0.92); I2=0%  
Test for overall effect: Z=0.18(P=0.86)  
Test for subgroup differences: Chi2=0.07, df=1 (P=0.79), I2=0%  

Favours Delayed 0.02 0.1 1 10 50 Favours Early

 
 

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 23
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Analysis 1.3.   Comparison 1 Delayed versus early introduction


of progressive enteral feeding, Outcome 3 Feed intolerance.
Study or subgroup Delayed Early Risk Ratio Weight Risk Ratio
  n/N n/N M-H, Fixed, 95% CI   M-H, Fixed, 95% CI
Abdelmaaboud 2012 23/63 22/62 39.02% 1.03[0.64,1.64]
Armanian 2013 9/35 24/47 36.05% 0.5[0.27,0.94]
Karagianni 2010 15/41 14/40 24.94% 1.05[0.58,1.87]
   
Total (95% CI) 139 149 100% 0.84[0.62,1.15]
Total events: 47 (Delayed), 60 (Early)  
Heterogeneity: Tau2=0; Chi2=3.8, df=2(P=0.15); I2=47.32%  
Test for overall effect: Z=1.07(P=0.29)  

Favours delayed 0.5 0.7 1 1.5 2 Favours early

 
 
Analysis 1.4.   Comparison 1 Delayed versus early introduction of
progressive enteral feeding, Outcome 4 Incidence of invasive infection.
Study or subgroup Delayed Early Risk Ratio Weight Risk Ratio
  n/N n/N M-H, Fixed, 95% CI   M-H, Fixed, 95% CI
Arnon 2013 4/30 2/30 3.5% 2[0.4,10.11]
Leaf 2012 69/199 55/198 96.5% 1.25[0.93,1.68]
   
Total (95% CI) 229 228 100% 1.27[0.95,1.7]
Total events: 73 (Delayed), 57 (Early)  
Heterogeneity: Tau2=0; Chi2=0.32, df=1(P=0.57); I2=0%  
Test for overall effect: Z=1.64(P=0.1)  

Favours delayed 0.1 0.2 0.5 1 2 5 10 Favours early

 
 
Analysis 1.5.   Comparison 1 Delayed versus early introduction of progressive
enteral feeding, Outcome 5 Duration of hospital admission (days).
Study or subgroup Delayed Early Mean Difference Weight Mean Difference
  N Mean(SD) N Mean(SD) Fixed, 95% CI   Fixed, 95% CI
Arnon 2013 30 46 (12) 30 37 (9) 11.21% 9[3.63,14.37]
Davey 1994 31 37.5 (3.8) 29 36.6 (4.5) 72.24% 0.9[-1.21,3.01]
Pérez 2011 96 30.1 (18) 130 27.4 (14.9) 16.55% 2.7[-1.72,7.12]
   
Total *** 157   189   100% 2.11[0.31,3.9]
Heterogeneity: Tau2=0; Chi2=7.66, df=2(P=0.02); I2=73.88%  
Test for overall effect: Z=2.3(P=0.02)  

Favours Delayed -10 -5 0 5 10 Favours Early

 
WHAT'S NEW
 

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 24
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

Date Event Description

3 October 2014 New citation required but conclusions Updated search in September 2014 identified two new trials for
have not changed inclusion in this review update (Armanian 2013; Arnon 2013).

3 October 2014 New search has been performed This updates the review "Delayed introduction of progressive en-
teral feeds to prevent necrotising enterocolitis in very low birth
weight infants" (Morgan 2013b).

 
HISTORY
Protocol first published: Issue 4, 1998
Review first published: Issue 4, 1998

 
Date Event Description

13 January 2011 New search has been performed This updates the review "Delayed introduction of progressive en-
teral feeds to prevent necrotising enterocolitis in very low birth
weight infants" published in the Cochrane Database of Systemat-
ic Reviews, Issue 2, 2008 (Bombell 2008).

Updated search includes three new trials (Ostertag 1986; Kara-


gianni 2010; Leaf 2012).

New authorship for this review update.

13 January 2011 New citation required and conclusions The addition of new trial data has increased the total number
have changed of participating infants to 600 and modified the implications for
practice and research.

2 February 2008 New search has been performed This updates the review "Early versus delayed initiation of pro-
gressive enteral feedings for parenterally fed low birth weight or
preterm infants" published in the Cochrane Database of System-
atic Reviews, Issue 1, 2000 (Kennedy 2000).

The title has been changed to "Delayed introduction of progres-


sive enteral feeds to prevent necrotising enterocolitis in very low
birth weight infants" and has a new authorship of Sarah Bombell
and William McGuire. Changes made to the original protocol are
outlined below:

1. Introduction of progressive enteral feeds is defined as feed


volumes more than 24 ml/kg/day (1 ml/kg/hour).
2. The population has been restricted to very low birth weight
and very preterm infants
3. Subgroup analyses of extremely low birth weight and extreme-
ly preterm infants, and infants with evidence of intrauterine
growth restriction or absent or reversed end-diastolic flow veloc-
ities in Doppler studies of the fetal aorta or umbilical artery were
prespecified.

Search updated December 2007. No new trials were included,


but one on-going trial was identified.

The findings and implications for practice and research of the re-
view have not changed overall.

11 January 2008 Amended Converted to new review format.

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 25
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Cochrane Trusted evidence.
Informed decisions.
 
 
Library Better health. Cochrane Database of Systematic Reviews

 
CONTRIBUTIONS OF AUTHORS
Lauren Young and William McGuire updated the search, independently determined the eligibility of identified studies, assessed the
methodological quality of the included trials, and extracted the relevant information and data.

All authors completed the final review.

DECLARATIONS OF INTEREST
None known.

SOURCES OF SUPPORT

Internal sources
• Centre for Reviews and Dissemination, Hull York Medical School, UK.

External sources
• National Institute for Health Research, UK.
• Eunice Kennedy Shriver National Institute of Child Health and Human Development National Institutes of Health, Department of Health
and Human Services, USA.

Editorial support of the Cochrane Neonatal Review Group was funded with Federal funds from the Eunice Kennedy Shriver National
Institute of Child Health and Human Development National Institutes of Health, Department of Health and Human Services, USA, under
Contract No. HHSN267200603418C

INDEX TERMS

Medical Subject Headings (MeSH)


*Infant, Low Birth Weight;  *Infant, Premature;  Cause of Death;  Enteral Nutrition  [*methods];  Enterocolitis, Necrotizing  [*prevention
& control];  Infant, Extremely Low Birth Weight;  Infant, Extremely Premature;  Infant, Very Low Birth Weight;  Parenteral Nutrition; 
Randomized Controlled Trials as Topic;  Time Factors

MeSH check words


Humans; Infant, Newborn

Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants (Review) 26
Copyright © 2014 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
SOCIETY COMMENTARY

Donor Human Milk for Preterm Infants: Current Evidence


and Research Directions
y
Sertac Arslanoglu, zWillemijn Corpeleijn, Guido Moro, §Christian Braegger,
jj
Cristina Campoy, ôVirginie Colomb, #Tamas Decsi, Magnus Domellöf, yyMary Fewtrell,
zz
Iva Hojsak, §§Walter Mihatsch, jjjjChristian Mølgaard, ôôRaanan Shamir, ##Dominique Turck, and
z
Johannes van Goudoever, ESPGHAN Committee on Nutrition

ABSTRACT
guidelines. Storage and processing of human milk reduces some biological
The Committee on Nutrition of the European Society for Pediatric Gastro-
components, which may diminish its health benefits. From a nutritional point
enterology, Hepatology, and Nutrition aims to document the existing
of view, DHM, like HM, does not meet the requirements of preterm infants,
evidence of the benefits and common concerns deriving from the use of
necessitating a specific fortification regimen to optimize growth. Future
donor human milk (DHM) in preterm infants. The comment also outlines
research should focus on the improvement of milk processing in HMB,
gaps in knowledge and gives recommendations for practice and suggestions particularly of heat treatment; on the optimization of HM fortification; and
for future research directions. Protection against necrotizing enterocolitis is on further evaluation of the potential clinical benefits of processed and
the major clinical benefit deriving from the use of DHM when compared fortified DHM.
with formula. Limited data also suggest unfortified DHM to be associated
with improved feeding tolerance and with reduced cardiovascular risk Key Words: donor milk, human milk, human milk banking, pasteurization,
factors during adolescence. Presence of a human milk bank (HMB) does preterm infant
not decrease breast-feeding rates at discharge, but decreases the use of
formula during the first weeks of life. This commentary emphasizes that
fresh own mother’s milk (OMM) is the first choice in preterm infant feeding (JPGN 2013;57: 535–542)
and strong efforts should be made to promote lactation. When OMM is not
available, DHM is the recommended alternative. When neither OMM nor
DHM is available, preterm formula should be used. DHM should be
provided from an established HMB, which follows specific safety I n a recent position paper by the European Society for Pediatric
Gastroenterology, Hepatology, and Nutrition Committee on
Nutrition, it was concluded that breast-feeding is the natural and
advisable way of supporting the growth and development of healthy
Received June 11, 2013; accepted June 11, 2013. term infants (1). Human milk (HM) also offers benefits to preterm
From the Italian Association of Human Milk Banks, Milan, Italy, the infants (2–4); however, in preterm infants, breast-feeding may not
yDivision of Neonatology, İzmir Dr Behçet Uz Children’s Hospital,
be possible, and own mother’s milk (OMM) may not be available.
İzmir, Turkey, the zPaediatrics, VU University Medical Center, Amster-
In this situation, donor HM (DHM) and preterm infant formula are
dam, the Netherlands, and Paediatrics, Emma Children’s Hospital-AMC,
Amsterdam, The Netherlands, the §University Children’s Hospital, the alternatives.
Zurich, Switzerland, the jjDepartment of Paediatrics, University of Official bodies such as the World Health Organization (5)
Granada, Granada, Spain, the ôHospital Necker Paris, Paris, France, and the American Academy of Pediatrics (6) recommend the use
the #Department of Paediatrics, University of Pecs, Pecs, Hungary, the of donated breast milk as the first alternative, when maternal milk
Department of Clinical Sciences, Paediatrics, Umeå University, is not available. American Academy of Pediatrics states that in
Umeå, Sweden, the yyMRC Childhood Nutrition Research Centre, such a situation, pasteurized DHM should be the first choice for
UCL Institute of Child Health, London, UK, the zzReferral Center preterm infants. To offer this opportunity to preterm infants, HM
for Pediatric Gastroenterology and Nutrition, Children’s Hospital should be obtained from a HM bank (HMB). The number of
Zagreb, Zagreb, Croatia, the §§Department of Paediatrics, Harlaching,
Munich Municipal Hospitals, Munich, Germany, the jjjjDepartment of
HMBs is rapidly increasing worldwide. At present, in Europe,
Nutrition, Exercise and Sports, Faculty of Science, University of Copen- there are 186 HMBs, and new banks will be established with the
hagen, Denmark, the ôôSchneider Children’s Medical Center of Israel, support of the European Milk Bank Association (www.european-
Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel, and milkbanking.com); however, DHM is not available to all preterm
the ##Jeanne de Flandre Children’s Hospital, Lille University Faculty of infants.
Medicine, Lille, France. In many countries, national policies to improve infant health
Address correspondence and reprint requests to Sertac Arslanoglu, MD, outcomes consider DHM obtained from an HMB to be a reasonable
Associate Professor of Neonatology, Division of Neonatology, İzmir and effective tool in the delivery of health care to infants and
Dr. Behçet Uz Children’s Hospital, İzmir 35210, Turkey (e-mail: children (7). Some countries have developed national guidelines
asertac@tiscali.it).
that are published in English (8–11).
Drs Arslanoglu, Corpeleijn, and Moro are the guests; Dr Mihatsch is a
Committee secretary; and Dr Goudoever is a Committee chairman. This review aims to document the published evidence
The authors report no conflicts of interest. regarding the benefits deriving from the use of DHM for preterm
Copyright # 2013 by European Society for Pediatric Gastroenterology, infants, and to address the main concerns limiting its widespread
Hepatology, and Nutrition and North American Society for Pediatric adoption as a standard care. It also outlines the gaps in knowledge,
Gastroenterology, Hepatology, and Nutrition and gives recommendations for practice and suggestions for
DOI: 10.1097/MPG.0b013e3182a3af0a future research.

JPGN  Volume 57, Number 4, October 2013 535


Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
Arslanoglu et al JPGN  Volume 57, Number 4, October 2013

METHODS effect of native cow’s-milk protein on the developing human


The literature review included electronic searches of intestine. A recent multicenter RCT compared the outcomes of
MEDLINE (1966 –October 2011), EMBASE (1980–October very-low-birth-weight (VLBW) infants fortified by 2 different
2011), CINAHL (1981–October 2011), the Cochrane Library, kinds of HM fortifier (HMF) (20). One group received HM-based
and conference proceedings. The electronic search used the HMF (HM concentrate with minerals and vitamins), whereas the
following text words and MeSH terms: donor milk, human milk, other received bovine milk–based HMF and PF. The HM-based
breast milk, banked milk, milk bank, milk banking, (human fortifier group had a significantly lower incidence of overall and
milk OR breast milk) AND outcomes, (human milk OR breast surgical NEC than the other group.
milk) AND necrotizing enterocolitis, (human milk OR breast milk)
AND infection, (human milk OR breast milk) AND neurodevelop-
ment, (human milk OR breast milk) AND bronchopulmonary Conclusion and Comments on NEC
dysplasia, (human milk OR breast milk) AND (pasteurization OR
 Feeding preterm infants with DHM is associated with
heat).
Reference lists of the previous reviews and relevant studies a decreased risk of NEC when compared with formula
were examined. Trials that had been reported only as abstracts were feeding.
 There are limited data on the comparison of feeding with
eligible for inclusion if sufficient information was available from
the report. fortified DHM versus PF. Because fortification of HM is the
present practice for preterm and particularly for VLBW
infants, future studies should compare the effect of feeding
CLINICAL BENEFITS DERIVING FROM with fortified DHM versus formula on the NEC incidence.
THE USE OF DHM  An exclusive HM diet (HM þ HM-based fortifier) may
Randomized controlled trials (RCTs) focusing specifically reduce the NEC incidence even further, but this needs to
on pasteurized DHM as a sole diet are sparse because it is no longer be confirmed.
considered acceptable to randomize infants to any other diet if
OMM is available. In most of the studies randomly assigning
infants to HM or formula, the HM group includes both OMM
and DHM. Feeding Tolerance
Concerns regarding feeding intolerance and the perceived
Necrotizing Enterocolitis (NEC) risk of NEC are the main obstacles for initiation and advancement
of enteral feeds in VLBW infants. Three intervention trials
Three systematic reviews (2,12,13) addressed specifically (14,15,21,22) conducted in the 1980s and included in the recent
the effect of DHM versus formula on clinical outcomes. All of these 2 systematic reviews (2,13) reported significantly fewer episodes
reviews suggest that the use of DHM has a protective effect against of feeding intolerance (14,15,21), withdrawals because of
NEC in premature infants. intolerance (14), and diarrhoea (22) in the unfortified DHM
The Cochrane review in 2007 (12) considered 5 RCTs group compared with the formula group. In the large multicenter
conducted in preterm and low-birth-weight infants: Gross et al English trial (15,21), infants fed exclusively unfortified DHM
1983, Lucas et al 1984 (trials 1 and 2), Schanler et al 2005, and (trial 1) and as a supplement to OMM (trial 2) were found to
Tyson et al 1983 (14–18). In these studies, Gross et al (14) establish full enteral feeds earlier and had fewer vomits and signs
compared term formula with unfortified DHM, whereas Lucas of gastric stasis compared with those who received infant
et al (15,16) and Tyson et al (17) compared preterm formula formula; however, the data have not been published as a full
(PF) with unfortified DHM. Lucas trial 1 provides NEC incidence article. All of the studies have been performed using native
comparing an exclusively DHM diet with PF in 159 infants, protein formula in contrast to hydrolyzed protein preterm infant
whereas Lucas trial 2 compares DHM with PF as a supplement formula, which has been shown to significantly improve feeding
to OMM (15,16). The RCT conducted by Schanler et al (18) is the tolerance.
only one comparing PF with fortified DHM. DHM was pasteurized
in all of the studies except the Tyson RCT (17). A meta-analysis of
data from 5 trials demonstrated a significantly higher incidence of Conclusion and Comments on Feeding Tolerance
NEC in formula-fed infants (typical relative risk 2.5, with 95%
confidence interval [CI] 1.2–5.1). The observed effect sizes were  Limited available data from the1980s support the hypothesis
similar across 5 studies, and there was no statistical evidence of that unfortified DHM results in improved feeding tolerance
heterogeneity. The pooled estimate suggests that 1 extra case of compared with formula.
NEC will occur in every 33 infants who receive formula milk. The  Studies comparing the effect of fortified DHM versus formula
systematic review and meta-analysis of Boyd et al in 2007 (2) and on feeding tolerance are lacking.
an earlier systematic review and meta-analysis by McGuire et al in
2003 (13) came to similar conclusions. The paucity of data on
comparison of formula milk with nutrient-fortified HM (only 1
study) is the limitation in these reviews and highlights the need for Bronchopulmonary Dysplasia
new RCTs comparing the effect of fortified donor milk versus PF on
NEC occurrence. One RCT (18) designed to compare the incidence of infec-
An intriguing point is the mechanism through which DHM tion-related events in extremely premature infants (<30 weeks of
may be offering protection against NEC. This protection may be gestation) observed a reduction in the incidence of bronchopul-
through the supply of immunoprotective factors to the immature monary dysplasia (BPD) (oxygen need at postmenstrual age of
mucosa; however, the absence of harmful antigens may also be a 36 weeks) in the fortified DHM-fed infants compared with those fed
contributing factor (19). NEC may be caused by the detrimental PF (15% vs 28%; P ¼ 0.048).

536 www.jpgn.org

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
JPGN  Volume 57, Number 4, October 2013 Donor Human Milk for Preterm Infants

Conclusion and Comment on BPD Conclusion and Comments on Neurodevelopment

DHM may be protective against BPD. This needs to be deter-  No beneficial effect on neurocognitive outcome has been
mined by further RCTs. shown in the only available RCT.
 The comparison was made between PF and unfortified
DHM, which was frequently drip milk having low energy
content. This practice does not reflect the current feeding
Long-Term Cardiovascular Risk Factors strategies in NICUs.
 Studies comparing fortified DHM and PF groups with regard
Data on cardiovascular risk factors during adolescence are to neurodevelopment are needed.
available from follow-up of a single randomized trial conducted in
5 neonatal units in the UK in the early 1980s. In 1 limb of the
original study (15), preterm infants were randomized to receive
either unfortified DHM or a PF; randomization was stratified Allergy
according to whether or not the mother provided her own milk.
This is the only trial in which infants have been randomized to HM The neonatal period is a critical window of opportunity for
versus formula without confounding by the mother’s decision to immunological adaptation. HM plays an important role in the
breast-feed (23). development of the immune system through its immunoactive
Adolescents (ages 13 to 16 years) who had been random- factors. Among these factors, HM oligosaccharides and long-chain
ized to receive DHM, either as sole diet or as a supplement to polyunsaturated fatty acids are well-known key immunomodulating
maternal breast milk during the neonatal period, had significantly components (27,28). Recently, HM transforming growth factor-b
lower mean blood pressure (BP) (mean differences 4.1); how- has been indicated as an immunoregulatory cytokine, particularly
ever, follow-up was 26% only (24). Adolescents who had been for allergy prevention (29,30). The English multicenter trial eval-
randomized to DHM also had a more favorable plasma lipid uating the effect of feeding in the early postnatal period on allergic
profile, with a lower ratio of low-density to high-density lipo- manifestations at 18 months after term found no difference in the
protein cholesterol than those fed PF (25). Owing to the low incidence of allergic reactions between the DHM- and formula-fed
percentage of follow-up (26%), the significance of these findings groups (31); however, in a subgroup of preterm infants with high
is uncertain. risk for allergy, cow’s-milk–based formula increased the risk of
developing 1 allergic manifestations (particularly eczema) (odds
ratio 3.6; 95% CI 1.4–9.1). High risk was defined as having a first-
degree relative with a history of atopic disease (eczema, asthma, hay
Conclusion and Comments on Cardiovascular Risk Factors fever, drug reactions, or confirmed food allergy). No studies are

available examining the influence of HM as compared with formula
DHM in early life may have beneficial effects on cardiovas- in infants with a high risk for developing allergy.
cular risk factors measured during adolescence; the signifi-
cance of these findings for the development of cardiovascular
disease is uncertain. Conclusion and Comment on Allergy
 A limitation in the evaluation of these findings is that the
comparison was made between PF and unfortified DHM.  The only available RCT shows that DHM does not have a
This practice does not reflect the present feeding strategies protective effect against the development of allergy in pre-
in neonatal intensive care units (NICUs). If the underlying term infants; however, the same RCT reports a protective
mechanism for these effects relates to slower early growth, effect of DHM against eczema in preterm infants at high risk
it is important to consider whether these effects would persist if for allergy.
fortified DHM is used and early growth rates are faster.
 Further studies should compare the long-term outcomes
between fortified DHM versus PF fed infants.
CONCERNS AND UNCERTAINTIES
Safety
Long-Term Neurodevelopment Microbiological Safety
DHM should be obtained from established HMBs that follow
The only RCT reporting impact of DHM on neurocognitive specific guidelines for screening, storage, and handling procedures to
outcomes is the English 3-center study (26). In this study, 502 optimize its composition while ensuring its safety for the recipient
preterm infants were assigned to receive either unfortified mature (32). Many countries now have their own HMB guidelines (8–11,33).
DHM or PF as sole enteral feeds or as supplements to OMM. PF was The first HMB was established as early as 1909 in Vienna, Austria.
associated with an improved neurocognitive outcome at 1 year and Many banks have been established since then, and some closed
no difference in neurocognitive outcomes (Bayley scores) was seen following the early years of the HIV pandemic in the 1980s.
between the 2 diet groups at 18 months, but it must be noted that the Pasteurization of the milk minimizes the risk of disease
DHM collected in the United Kingdom in the early 1980 s had an transmission via HM, inactivating most of the viral and bacterial
energy content of 50 kcal/100 mL. The low energy content was the contaminants. In addition, donors are screened in a similar way as
result of the fact that collected DHM was frequently drip milk which for blood donation. No report has been published showing transfer
had a lower fat content (26). Despite the importance of this outcome of diseases through pasteurized DHM, although milk may contain
parameter, no further follow-up results have been published by the microorganisms (34). Nevertheless, HMBs, like blood banks,
authors with regard to long-term neurodevelopment, whereas other should be aware of the threat of emerging (milk transmissible)
parameters such as cardiovascular biomarkers in adolescence have pathogens that are not included in contemporary screening proto-
been published. cols. There is concern that growth of Bacillus sp during the heating

www.jpgn.org 537

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
Arslanoglu et al JPGN  Volume 57, Number 4, October 2013

process may be increased (35); however, although spore-forming used method. It results in the loss of the quantity and/or activity of
Bacillus sp may survive pasteurization, this is thought to be a rare some biologically functional milk components to varying degrees:
contaminant of human breast milk in contrast to cow’s milk (36).
Regardless, this type of contamination can be controlled by proper 1. Mild to moderate decrease in IgA and secretory IgA
storage and handling after pasteurization, which should prevent any concentrations (20%–30%, range 0%–60%) and activity
Bacillus sp from growing. HMBs should have policies for micro- (33%–39%) (44–53).
biological quality control. 2. Considerable loss in concentration/activity of lactoferrin
(50%–75%) (46,47,49–51,54,55), lysozyme (24%–74%)
(44–48,50–52,54), IgG (34%–76%) (45,47), some cytokines
Chemical Pollutants, Including Drugs of Abuse (interleukin-10, tumor necrosis factor-a) (56,57), growth factors,
and hormones (insulin-like growth factor 1, adinopectin, insulin,
Environmental pollutants such as mercury, dioxins, and and leptin) (58–60), and antioxidant capacity of HM (61).
polychlorinated biphenyls (PCBs) are taken up via food and stored
3. Almost complete loss of lipase activity (44,49), IgM
in fatty tissue. There are no specific studies conducted with DHM. (concentrations) (45,46), and white blood cells (62,63).
Some of the pollutants can act as endocrine disruptors involving
thyroid, hypothalamus, and gonads (37,38). Prenatal exposure to an Other nutritional and biological components, such as oligo-
organochlorine compound has been reported to result in impaired saccharides (64), lactose, glucose (65,66), long-chain polyunsatu-
neurodevelopment at 4 years (39), whereas perinatal exposure to rated fatty acids, gangliosides (57,67,68), vitamins A, D, E, B12, folic
high PCB levels has been associated with neurotoxicity (40), and acid (44,69), some cytokines (interleukin-2, -4, -5, -8, -13) (57), and
perinatal dioxin exposure has been associated with persistent some growth factors (EGF and TGF-b1), are preserved (56,58).
hematologic and immunologic disturbances (41). Theoretically, Holder pasteurization maintains the bactericidal activity of
these substances can be excreted in breast milk. The concentration the milk against Escherichia coli better than high-temperature
of PCBs and dioxins in breast milk of European women has short-term pasteurization (70). It has been also shown that despite
decreased during the last decade as a consequence of measures the reduction in IgA concentrations, remaining molecules in the
against environmental pollution. Furthermore, as suggested in the Holder-pasteurized HM effectively inhibit bacterial (enteropatho-
study, monitoring the effect of PCBs in colostral milk on the visual genic E coli) adhesion (71). Similarly, in an earlier study, although
function in infants (42), HM may be offsetting potential deleterious Holder pasteurization decreased the activities of specific antibody
effects of these pollutants through its various biofactors. Future to E coli and lactoferrin, pasteurized milk remained effective at
studies should address the presence of these pollutants in DHM and inhibiting in vitro growth of E coli (54).
their possible effects on infant health. New methods to improve the biological quality and safety of
Besides environmental pollutants, other unwanted substances DHM are under investigation (72). High-temperature short-term
such as medication, alcohol, nicotine, and drugs of abuse are also pasteurization (flash pasteurization, 728C for 5–15 seconds)
excreted into the milk. Presently, no internationally accepted list of (44,49,55,58,70) and its homemade low-tech variant for developing
medicines that can safely be used by milk donors exists. HMBs are countries (flash-heat treatment) (73–75), thermoultrasonic treat-
therefore expected to compile their own list based on available ment (50), high-pressure processing (76,77), and Ohmic heat
literature and pharmacological properties. Because DHM is generally treatment (72) are the alternative methods on which present studies
intended for sick and premature infants, and infants are often exposed are focused.
to milk from >1 donor, guidelines for medication use in HM donors
should be more strict than those for women who are solely feeding Conclusion and Comments on Nutritional and
their own healthy infants. The safety of DHM relies heavily on the Biological Quality of DHM
accurate reporting of nicotine, alcohol, or drug abuse of potential
donors because it is not feasible to routinely test all milk for a wide  Holder pasteurization, the most commonly used procedure, is
range of harmful substances. Special attention should also be paid to safe but reduces the nutritional/biological quality of DHM.
the use of herbal remedies and herbal teas because some contain  Pasteurization should be optimized to maintain microbiolo-
harmful substances, for example, fennel tea can contain substantial gical safety while preserving the highest amount and activity
amounts of estragole (43). of the bioactive milk components.

Conclusion and Comments on Safety

 DHM should be pasteurized. Slow Growth


 Donors should be screened in a similar way as for blood Slow Growth Because of Inadequate Nutrient
donation, and should be asked about their use of alcohol,
nicotine, and drugs.
Content of DHM
 HM does not meet the high nutrient requirements of the
Studies are needed to address the presence and possible health
VLBW infant. Standard multicomponent fortification of HM
consequences of pollutants in DHM.
designed to optimize nutritional intakes (78) often falls short of this
goal with regard to protein (79,80). This problem may be amplified
with DHM, which is most often provided by the mothers of term
infants beyond 1 month postpartum and which is likely to have lower
Alterations in Nutritional and Biological Quality protein content than preterm mothers’ milk (66,81–83). A recent
of DHM observational study indicates that using standard fortification, weight
gain is faster in preterm infants fed OMM than in those fed DHM,
Some significant concerns are related to the possible altera- whereas there is no difference in terms of linear growth (84).
tions in the nutritional and biological quality of DHM because of its The systematic reviews of Quigley (12) and Boyd (2)
handling and storage, but particularly because of the heat treatment. reported that preterm or low-birth-weight infants who received
Holder pasteurization (62.58C, 30 minutes) is the most commonly formula regained birth weight earlier and had higher short-term

538 www.jpgn.org

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
JPGN  Volume 57, Number 4, October 2013 Donor Human Milk for Preterm Infants

rates of weight gain, linear growth, and head growth than infants OMM come first. When OMM is not available or is insufficient,
who received DHM; however, of 8 trials included, only 1 (18) DHM is used along with the ongoing efforts to promote lactation. In
compared fortified DHM with PF. In this trial, infants fed DHM had fact, the European Milk Bank Association, in its constitution, states
a slower rate of weight gain compared with PF (17.1 vs 20.1 the first 3 objectives of the Association as follows:
g  kg1  day1; P ¼ 0.001). Length and head circumference incre-
ments were similar in the 2 groups. 1. To promote breast-feeding
The fat and protein content of HM is extremely variable, and 2. To promote the donation of HM to HMBs
protein decreases with lactation duration. In recent years, it has 3. To promote the use of DHM for premature infants and
become evident that preterm infants fed fortified HM (OMM or other infants with specific needs who do not have access
DHM) receive less protein than assumed (85) and continue to grow to OMM
more slowly in the short term, even with standard HM fortification,
Some concerns have been raised that the presence of an HMB
compared with PF-fed infants. Although there is some uncertainty
and the use of DHM may attenuate the efforts to promote lactation
about the optimal growth, postnatal growth failure has not been
resulting in decreased breast-feeding rates.
solved with HM fortification in standard fashion (79). Thus, HM
No RCT could be identified addressing this concern, but
fortification should be optimized to achieve better short-term
recently some reports showed the opposite: A report from Australia
growth, which is associated with improved neurocognitive out-
(93) cites the breast-feeding rates in the 3 years following the
come. Individualized fortification has been shown to be effective in
establishment of an HMB. Despite a marked increase in DHM use in
improving protein intake, weight gain, and head circumference gain
the NICU, opening an HMB did not reduce the rate of milk
(86,87). There are 2 ways to individualize HM fortification:
expression, and breast-feeding rates at discharge increased.
‘‘adjustable fortification’’ (individualization based on blood urea
In an attempt to improve HM availability for preterm infants,
nitrogen measurements) (86) and ‘‘targeted fortification’’ (indivi-
health care providers of a NICU in UT designed an integrated
dualization based on milk analysis) (87). Improvement of the
approach: ‘‘Breast Milk Early Saves Trouble Program’’ (94). This
quality of HMF is a further issue, and HM-based fortifier may
program consisted of using exclusively HM (OMM and/or DHM) in
offer benefits compared with cow’s-milk–based fortifiers as shown
the NICU. Its implementation for 12 months increased HM and
in the multicentric study using Prolact þ H2MF (HM concentrate
DHM use in NICU, and breast-feeding rates at discharge tended to
with minerals and vitamins) (20). Earlier studies showed that infants
increase compared with the situation before the implementation
fed exclusively HM proteins (HM þ HM protein supplement) have
period (53% vs 44%; P ¼ 0.09)
plasma amino acid concentrations that differ significantly from
A Spanish study conducted in Madrid (95) directly
those fed either whey-predominant or casein-predominant formulas
addressed this concern and evaluated the effect of opening an
at similar protein intakes. The amino acid pattern of low-birth-
HMB in a NICU on the rates of exclusive breast-feeding at
weight infants fed exclusively HM proteins is similar to the pattern
discharge and formula use in the NICU. The researchers concluded
found in growing breast-fed term infants (88,89).
that presence of an HMB in a neonatal unit did not reduce the rate of
exclusive breast-feeding at discharge, but did reduce the use of
Potential Slow Growth Because of Alterations in infant formula during the first 4 weeks of life (37% vs 60%;
the Nutritional Quality of DHM P ¼ 0.01). The availability of having DHM also enabled earlier
initiation of enteral feeding.
As mentioned before, lipase activity is almost completely
lost following Holder pasteurization. It has also been shown that
heat induces alterations of the milk fat globule surface removing the
glycoprotein filaments (90). These heat-induced changes can Conclusion and Comment on the Relation of HMBs
explain the reduced milk fat absorption reported in pasteurized and Rates of Breast-feeding
HM-fed preterm infants (91,92). Optimizing the heat processing
and determining the best method of pasteurization for maintaining The existing data show that the presence of an HMB and use of
the nutritional and biological quality of DHM are essential. DHM in the NICU do not decrease the breast-feeding rates at
discharge, but decrease formula use during the first weeks
of life.
Conclusion and Comments on Growth

 HM- and DHM-fed preterm infants have slower early growth


than PF-fed infants. CONCLUSIONS, RECOMMENDATIONS,
 Inadequacy of standard HM fortification, particularly with FUTURE RESEARCH DIRECTIONS
regard to protein, and decreased fat absorption owing to the
loss of lipase activity following pasteurization and loss of fat Conclusions
during handling are the main factors explaining the slower Based on the evidence presented in this commentary, the
growth seen in infants who receive DHM. European Society for Pediatric Gastroenterology, Hepatology, and
 Individualized fortification (adjustable or targeted) may help Nutrition Committee on Nutrition concludes the following:
to ensure adequate nutrient intakes.

1. DHM is associated with reduced NEC rates compared with
Studies on the quality of fortifiers and different heat treatment cow’s-milk–based formula.
strategies are needed.
2. Unfortified DHM, like HM, is associated with slower neonatal
growth when compared with PF.
Does the Presence of an HMB Compete With 3. Appropriately handled and pasteurized DHM is microbio-
logically safe.
Breast-feeding? 4. Presence of an HMB does not decrease the breast-feeding rates
The purpose of HM banking is to provide a HM supply for at discharge, but may decrease formula use during the first
infants (mainly preterm). Promotion of breast-feeding and use of weeks of life.

www.jpgn.org 539

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
Arslanoglu et al JPGN  Volume 57, Number 4, October 2013

Recommendations 10. HMBANA. Guidelines for the establishment and operation of a donor
human milk bank. https://www.hmbana.org/products/publications. Pub-
lished 2011. Accessed September 5, 2013.
1. OMM is the first choice in preterm infant feeding, and strong 11. National Institute for Health and Clinical Excellence (NICE). Donor
efforts should be made to promote lactation. Breast Milk Banks: The Operation of Donor Milk Bank Services. http://
2. When mother’s milk is not available, DHM is the preferred www.ncbi.nlm.nih.gov/pubmed/22319806. Accessed September 5,
choice. When mother’s milk and DHM are not available, PF 2013.
12. Quigley MA, Henderson G, Anthony NY, et al. Formula milk versus
should be used.
donor breast milk for feeding preterm or low birth weight infants.
3. No DHM should be provided outside the organization of an Cochrane Database Syst Rev 2007:CD002971.
established HMB. 13. McGuire W, Anthony MY. Donor human milk versus formula for
4. Adequate screening of donors and pasteurization of the donor preventing necrotising enterocolitis in preterm infants: systematic re-
milk should be performed. view. Arch Dis Child Fetal Neonatal Ed 2003;88:F11–4.
5. DHM should be fortified to meet early nutrient requirements 14. Gross SJ. Growth and biochemical response of preterm infants fed
and achieve better short-term growth, which is associated with human milk or modified infant formula. N Engl J Med 1983;308:237–
improved neurocognitive outcome. Individualized fortification 41.
is advised. 15. Lucas A, Gore SM, Cole TJ, et al. Multicentre trial on feeding low
birthweight infants: effects of diet on early growth. Arch Dis Child
1984;59:722–30.
Research Directions 16. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis.
Lancet 1990;336:1519–23.
17. Tyson JE, Lasky RE, Mize CE, et al. Growth, metabolic response, and
1. Randomized clinical trials comparing development in very-low-birth-weight infants fed banked human milk or
a. The impact of feeding with PF versus fortified DHM on enriched formula. I. Neonatal findings. J Pediatr 1983;103:95–104.
short-term clinical outcomes: growth, NEC, sepsis and other 18. Schanler RJ, Lau C, Hurst NM, et al. Randomized trial of donor human
infections, retinopathy of prematurity, BPD, feeding milk versus preterm formula as substitutes for mothers’ own milk in the
tolerance, and mortality feeding of extremely premature infants. Pediatrics 2005;116:400–6.
b. The impact of feeding with PF versus fortified DHM on 19. Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med 2011;
long-term clinical outcomes: allergy, neurodevelopmental 364:255–64.
20. Sullivan S, Schanler RJ, Kim JH, et al. An exclusively human milk-
outcomes, obesity, metabolic syndrome, and other cardio-
based diet is associated with a lower rate of necrotizing enterocolitis
vascular risk factors than a diet of human milk and bovine milk-based products. J Pediatr
c. The impact of feeding with DHM with bovine fortifier 2010;156:562–7e1.
versus HM diet (OMM/DHM þ HM-based fortifier) on 21. Lucas A. AIDS and human milk bank closures. Lancet 1987;1:1092–3.
short-term and long-term clinical outcomes 22. Schultz K, Soltesz G, Mestyan J. The metabolic consequences of human
2. Development and evaluation of different pasteurization milk and formula feeding in premature infants. Acta Paediatr Scand
techniques to optimize microbiological safety, and to maintain 1980;69:647–52.
the biological and nutritional quality of HM 23. Fewtrell MS. Breast-feeding and later risk of CVD and obesity:
3. Development of systems to ensure the lowest possible level of evidence from randomised trials. Proc Nutr Soc 2011;70:472–7.
toxic products and pollutants in the donor HM 24. Singhal A, Cole TJ, Lucas A. Early nutrition in preterm infants and later
blood pressure: two cohorts after randomised trials. Lancet 2001;
357:413–9.
25. Singhal A, Cole TJ, Fewtrell M, et al. Breastmilk feeding and lipopro-
tein profile in adolescents born preterm: follow-up of a prospective
REFERENCES randomised study. Lancet 2004;363:1571–8.
1. Agostoni C, Braegger C, Decsi T, et al. Breast-feeding: a commentary 26. Lucas A, Morley R, Cole TJ, et al. A randomised multicentre study of
by the ESPGHAN Committee on Nutrition. J Pediatr Gastroenterol human milk versus formula and later development in preterm infants.
Nutr 2009;49:112–25. Arch Dis Child Fetal Neonatal Ed 1994;70:F141–6.
2. Boyd CA, Quigley MA, Brocklehurst P. Donor breast milk versus infant 27. Bode L. Human milk oligosaccharides: prebiotics and beyond. Nutr Rev
formula for preterm infants: systematic review and meta-analysis. Arch 2009;67(suppl 2):S183–91.
Dis Child Fetal Neonatal Ed 2007;92:F169–75. 28. Gottrand F. Long-chain polyunsaturated fatty acids influence the im-
3. Sisk PM, Lovelady CA, Dillard RG, et al. Early human milk feeding is mune system of infants. J Nutr 2008;138:S1807–12.
associated with a lower risk of necrotizing enterocolitis in very low birth 29. Penttila IA. Milk-derived transforming growth factor-beta and the infant
weight infants. J Perinatol 2007;27:428–33. immune response. J Pediatr 2010;156:S21–5.
4. Meinzen-Derr J, Poindexter B, Wrage L, et al. Role of human milk in 30. Verhasselt V, Milcent V, Cazareth J, et al. Breast milk-mediated transfer
extremely low birth weight infants’ risk of necrotizing enterocolitis or of an antigen induces tolerance and protection from allergic asthma. Nat
death. J Perinatol 2009;29:57–62. Med 2008;14:170–5.
5. WHO/UNICEF. Global strategy for infant and young child 31. Lucas A, Brooke OG, Morley R, et al. Early diet of preterm infants and
feeding. http://whqlibdoc.who.int/publications/2003/9241562218.pdf. development of allergic or atopic disease: a randomised prospective
Published 2003. Accessed September 5, 2013. study. BMJ 1990;300:837–40.
32. Arslanoglu S, Ziegler EE, Moro GE. World Association of Perinatal
6. American Academy of Pediatrics. Section on Breastfeeding. Breast-
Medicine Working Group On Nutrition. Donor human milk in preterm
feeding and the use of human milk. Pediatrics 2012;129:e827–41.
infant feeding: evidence and recommendations. J Perinat Med 2010;
7. Arnold LD. Global health policies that support the use of banked donor 38:347–51.
human milk: a human rights issue. Int Breastfeed J 2006;1:26. 33. Décret No 2010-805 du 13 juillet 2010 relatif aux missions, à l’auto-
8. Hartmann BT, Pang WW, Keil AD, et al. Best practice guidelines for the risation et aux conditions techniques d’organisation et de fonctionne-
operation of a donor human milk bank in an Australian NICU. Early ment des lactariums [in French]. http://sdp.perinat-france.org/ADLF/
Hum Dev 2007;83:667–73. textes_legislation_adlf.php. Accessed September 24, 2012.
9. Arslanoglu S, Bertino E, Tonetto P, et al. Guidelines for the establish- 34. Landers S, Updegrove K. Bacteriological screening of donor human
ment and operation of a donor human milk bank. J Matern Fetal milk before and after Holder pasteurization. Breastfeed Med 2010;
Neonatal Med 2010;23(suppl 2):1–20. 5:117–21.

540 www.jpgn.org

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
JPGN  Volume 57, Number 4, October 2013 Donor Human Milk for Preterm Infants

35. Hanson ML, Wendorff WL, Houck KB. Effect of heat treatment of milk 61. Silvestre D, Miranda M, Muriach M, et al. Antioxidant capacity of
on activation of Bacillus spores. J Food Prot 2005;68:1484–6. human milk: effect of thermal conditions for the pasteurization. Acta
36. Crielly EM, Logan NA, Anderton A. Studies on the Bacillus flora of Paediatr 2008;97:1070–4.
milk and milk products. J Appl Bacteriol 1994;77:256–63. 62. Tully DB, Jones F, Tully MR. Donor milk: what’s in it and what’s not.
37. Robins JC, Marsit CJ, Padbury JF, et al. Endocrine disruptors, environ- J Hum Lact 2001;17:152–5.
mental oxygen, epigenetics and pregnancy. Front Biosci (Elite Ed) 63. Björkstén B, Burman LG, De Château P, et al. Collecting and banking
2011;3:690–700Review. human milk: to heat or not to heat? BMJ 1980;281:765–9.
38. Crofton KM. Thyroid disrupting chemicals: mechanisms and mixtures. 64. Bertino E, Coppa GV, Giuliani F, et al. Effects of Holder pasteurization
Int J Androl 2008;31:209–23. on human milk oligosaccharides. Int J Immunopathol Pharmacol
39. Puertas R, Lopez-Espinosa MJ, Cruz F, et al. Prenatal exposure to mirex 2008;21:381–5.
impairs neurodevelopment at age of 4 years. Neurotoxicology 2010; 65. de Segura AG, Escuder D, Montilla A, et al. Heating-induced bacter-
31:154–60. iological and biochemical modifications in human donor milk after
40. Huisman M, Koopman-Esseboom C, Fidler V, et al. Perinatal exposure holder pasteurisation. J Pediatr Gastroenterol Nutr 2012;54:197–
to polychlorinated biphenyls and dioxins and its effect on neonatal 203.
neurological development. Early Hum Dev 1995;41:111–27. 66. Vieira AA, Soares FV, Pimenta HP, et al. Analysis of the influence of
41. ten Tusscher GW, Steerenberg PA, van Loveren H, et al. Persistent pasteurization, freezing/thawing, and offer processes on human milk’s
hematologic and immunologic disturbances in 8-year-old Dutch chil- macronutrient concentrations. Early Hum Dev 2011;87:577–80.
dren associated with perinatal dioxin exposure. Environ Health Perspect 67. Henderson TR, Fay TN, Hamosh M. Effect of pasteurization on long
2003;111:1519–23. chain polyunsaturated fatty acid levels and enzyme activities of human
42. Riva E, Grandi F, Massetto N, et al. Polychlorinated biphenyls in milk. J Pediatr 1998;132:876–8.
colostral milk and visual function at 12 months of life. Acta Paediatr 68. Fidler N, Sauerwald TU, Koletzko B, et al. Effects of human milk
2004;93:1103–7. pasteurization and sterilization on available fat content and fatty acid
43. Raffo A, Nicoli S, Leclercq C, et al. Quantification of estragole in fennel composition. J Pediatr Gastroenterol Nutr 1998;27:317–22.
herbal teas: implications on the assessment of dietary exposure to 69. Van Zoeren-Grobben D, Schrijver J, Van den Berg H, et al. Human milk
estragole. Food Chem Toxicol 2011;49:370–5. vitamin content after pasteurisation, storage, or tube feeding. Arch Dis
44. Hamprecht K, Maschmann J, Muller D, et al. Cytomegalovirus (CMV) Child 1987;62:161–5.
inactivation in breast milk: reassessment of pasteurization and freeze- 70. Silvestre D, Ruiz P, Martinez-Costa C, et al. Effect of pasteurization
thawing. Pediatr Res 2004;56:529–35. on the bactericidal capacity of human milk. J Hum Lact 2008;24:
45. Koenig A, de Albuquerque Diniz EM, Barbosa SF, et al. Immunologic 371–6.
factors in human milk: the effects of gestational age and pasteurization. 71. Carbonare SB, Palmeira P, Silva ML, et al. Effect of microwave
J Hum Lact 2005;21:439–43. radiation, pasteurization and lyophilization on the ability of human
46. Ford JE, Law BA, Marshall VM, et al. Influence of the heat treatment of milk to inhibit Escherichia coli adherence to HEp-2 cells. J Diarrhoeal
human milk on some of its protective constituents. J Pediatr 1977; Dis Res 1996;14:90–4.
90:29–35. 72. Moro GE, Arslanoglu S. Heat treatment of human milk. J Pediatr
47. Evans TJ, Ryley HC, Neale LM, et al. Effect of storage and heat on Gastroenterol Nutr 2012;4:165–6.
antimicrobial proteins in human milk. Arch Dis Child 1978;53:239–41. 73. Chantry CJ, Wiedeman J, Buehring G, et al. Effect of flash-heat
48. Gibbs JH, Fisher C, Bhattacharya S, et al. Drip breast milk: it’s treatment on antimicrobial activity of breastmilk. Breastfeed Med
composition, collection and pasteurization. Early Hum Dev 1977;1: 2011;6:111–6.
227–45. 74. Volk ML, Hanson CV, Israel-Ballard K, et al. Inactivation of cell-
49. Baro C, Giribaldi M, Arslanoglu S, et al. Effect of two pasteurization associated and cell-free HIV-1 by flash-heat treatment of breast milk.
methods on the protein content of human milk. Front Biosci (Elite Ed) J Acquir Immune Defic Syndr 2010;53:665–6.
2011;3:818–29. 75. Israel-Ballard K, Chantry C, Dewey K, et al. Viral, nutritional, and
50. Czank C, Simmer K, Hartmann PE. Simultaneous pasteurization and bacterial safety of flash-heated and pretoria-pasteurized breast milk to
homogenization of human milk by combining heat and ultrasound: prevent mother-to-child transmission of HIV in resource-poor countries:
effect on milk quality. J Dairy Res 2010;77:183–9. a pilot study. J Acquir Immune Defic Syndr 2005;40:175–81.
51. Akinbi H, Meinzen-Derr J, Auer C, et al. Alterations in the host defense 76. Viazis S, Farkas BE, Jaykus LA. Inactivation of bacterial pathogens in
properties of human milk following prolonged storage or pasteurization. human milk by high-pressure processing. J Food Prot 2008;71:109–18.
J Pediatr Gastroenterol Nutr 2010;51:347–52. 77. Permanyer M, Castellote C, Ramirez-Santana C, et al. Maintenance of
52. Wills ME, Han VE, Harris DA, et al. Short-time low-temperature breast milk immunoglobulin A after high-pressure processing. J Dairy
pasteurisation of human milk. Early Hum Dev 1982;7:71–80. Sci 2010;93:877–83.
53. Chen HY, Allen JC. Human milk antibacterial factors: the effect of 78. Kuschel CA, Harding JE. Multicomponent fortified human milk for
temperature on defense systems. Adv Exp Med Biol 2001;501:341–8. promoting growth in preterm infants. Cochrane Database Syst Rev
54. Eyres R, Elliott RB, Howie RN, et al. Low-temperature pasteurisation of 2004:CD000343.
human milk. N Z Med J 1978;87:134–5. 79. Arslanoglu S, Moro GE, Ziegler EE. The World Association of Perinatal
55. Conesa C, Rota C, Castillo E, et al. Antibacterial activity of recombinant Medicine Working Group On Nutrition. Optimization of human milk
human lactoferrin from rice: effect of heat treatment. Biosci Biotechnol fortification for preterm infants: new concepts and recommendations.
Biochem 2009;73:1301–7. J Perinat Med 2010;38:233–8.
56. Untalan PB, Keeney SE, Palkowetz KH, et al. Heat susceptibility of 80. Ziegler EE. Meeting the nutritional needs of the low-birth-weight infant.
interleukin-10 and other cytokines in donor human milk. Breastfeed Ann Nutr Metab 2011;58(suppl 1):8–18.
Med 2009;4:137–44. 81. Wojcik KY, Rechtman DJ, Lee ML, et al. Macronutrient analysis of a
57. Ewaschuk JB, Unger S, O’Connor DL, et al. Effect of pasteurization on nationwide sample of donor breast milk. J Am Diet Assoc 2009;
selected immune components of donated human breast milk. J Perinatol 109:137–40.
2011;31:593–8. 82. Casadio YS, Williams TM, Lai CT, et al. Evaluation of a mid-infrared
58. Goelz R, Hihn E, Hamprecht K, et al. Effects of different CMV-heat- analyzer for the determination of the macronutrient composition of
inactivation-methods on growth factors in human breast milk. Pediatr human milk. J Hum Lact 2010;26:376–83.
Res 2009;65:458–61. 83. Michaelsen KF, Skafte L, Badsberg JH, et al. Variation in macronu-
59. Resto M, O’Connor D, Leef K, et al. Leptin levels in preterm human trients in human bank milk: influencing factors and implications for
breast milk and infant formula. Pediatrics 2001;108:E15. human milk banking. J Pediatr Gastroenterol Nutr 1990;11:229–39.
60. Ley SH, Hanley AJ, Stone D, et al. Effects of pasteurization on 84. Montjaux-Regis N, Cristini C, Arnaud C, et al. Improved growth of
adiponectin and insulin concentrations in donor human milk. Pediatr preterm infants receiving mother’s own raw milk compared with
Res 2011;70:278–81. pasteurized donor milk. Acta Paediatr 2011;100:1548–54.

www.jpgn.org 541

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.
Arslanoglu et al JPGN  Volume 57, Number 4, October 2013

85. Arslanoglu S, Moro GE, Ziegler EE. Preterm infants fed fortified human 90. Buchheim W, Welsch U, Huston GE, et al. Glycoprotein filament
milk receive less protein than they need. J Perinatol 2009;29:489–92. removal from human milk fat globules by heat treatment. Pediatrics
86. Arslanoglu S, Moro GE, Ziegler EE. Adjustable fortification of human 1988;81:141–6.
milk fed to preterm infants: does it make a difference? J Perinatol 91. Andersson Y, Savman K, Blackberg L, et al. Pasteurization of mother’s
2006;26:614–21. own milk reduces fat absorption and growth in preterm infants. Acta
87. Polberger S, Räihä NC, Juvonen P, et al. Individualized protein for- Paediatr 2007;96:1445–9.
tification of human milk for preterm infants: comparison of ultrafiltrated 92. Williamson S, Finucane E, Ellis H, et al. Effect of heat treatment of
human milk protein and a bovine whey fortifier. J Pediatr Gastroenterol human milk on absorption of nitrogen, fat, sodium, calcium, and
Nutr 1999;29:332–8. phosphorus by preterm infants. Arch Dis Child 1978;53:555–63.
88. Moro G, Fulconis F, Minoli I, et al. Growth and plasma amino acid 93. Simmer K, Hartmann B. The knowns and unknowns of human milk
concentrations in very low birthweight infants fed either human milk banking. Early Hum Dev 2009;85:701–4.
protein fortified human milk or a whey predominant formula. Acta 94. Montgomery D, Schmutz N, Baer VL, et al. Effects of instituting the
Paediatr Scand 1989;78:18–22. ‘‘BEST Program’’ (Breast Milk Early Saves Trouble) in a level III
89. Moro GE, Minoli I, Fulconis F, et al. Growth and metabolic responses in NICU. J Hum Lact 2008;24:248–51.
low-birth-weight Infants fed human milk fortified with human milk 95. Utrera Torres MI, Medina Lopez C, et al. Does opening a milk bank in a
protein or with a bovine milk preparation. J Pediatr Gastroenterol Nutr neonatal unit change infant feeding practices? A before and after study.
1991;13:150–4. Int Breastfeed J 2010;5:4.

542 www.jpgn.org

Copyright 2013 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Anda mungkin juga menyukai