You are on page 1of 143

Editor-in-Chief Editor, Emeritus International Editors

EDWARD E LAWSON, MD GILBERT I MARTIN, MD GARY DARMSTADT, MD, MS


Johns Hopkins Hospital West Covina, California Baltimore, Maryland
Baltimore, Maryland
ARTHUR I EIDELMAN, MD
Jerusalem, Israel

Book Review Editor Imaging Section Editor Maternal-Fetal Medicine Editor


JAYANT P SHENAI, MD MARILYN SIEGEL, MD AARON B CAUGHEY, MD, MPP, MPH, PhD
Nashville, Tennessee St. Louis, Missouri San Francisco, California

Editorial Board
DAVID H ADAMKIN, MD JEFFREY S GERDES, MD ARUN PRAMANIK, MD
Louisville, Kentucky Philadelphia, Pennsylvania Shreveport, Louisiana
H FRANK ANDERSEN, MD JAY P GOLDSMITH, MD JOANN PRAUSE, PhD
Dallas, Texas New Orleans, Louisiana Laguna Hills, California
RONALD ARIAGNO, MD JOSEPH HAGEMAN, MD
Evanston, Illinois KARI RAIVIO, MD, PhD
Stanford, California
Helsinki, Finland
KEITH BARRINGTON, MD JOHN HARTLINE, MD
Montreal, QC, Canada Kalamazoo, Michigan WILLIAM E ROBERTS, MD
Jackson, Mississippi
MORAYE BEAR, MA, MS DIANE HOLDITCH-DAVIS, PhD
Woodland Hills, California Chapel Hill, North Carolina WARREN ROSENFELD, MD
Mineola, New York
JATINDER BHATIA, MD THOMAS C HULSEY, MSPH, ScD
Augusta, Georgia Charleston, South Carolina OLA DIDRIK SAUGSTAD, MD, PhD
Oslo, Norway
LUC BRION, MD MICHAEL T HYNAN, PhD
Bronx, New York Milwaukee, Wisconsin MICHAEL E SPEER, MD
ROBERT KOPOTIC, MSN, RN, RRT Houston, Texas
ANITA J CATLIN, DNSc, FNP, FAAN
Rohnert Park, California Jamul, California JOSEPH SPINNATO, MD
HUGO LAGERCRANTZ, MD Cincinnati, Ohio
IRA CHASNOFF, MD
Chicago, Illinois Stockholm, Sweden ANN R STARK, MD
MALCOLM LEVENE, MD Houston, Texas
DOUGLAS CUNNINGHAM, MD
Orange, California Leeds, United Kingdom DAVID K STEVENSON, MD
KAREN H MORIN, DSN, RN Palo Alto, California
WILLIAM A ENGLE, MD
Indianapolis, Indiana Newark, Delaware
DHARMAPURI VIDYASAGAR, MD
GABRIEL ESCOBAR, MD HIROSHI NISHIDA, MD Chicago, Illinois
Oakland, California Tokyo, Japan
MICHELE C. WALSH, MD
AVROY A FANAROFF, MD JOHN PARIS, SJ Cleveland, Ohio
Cleveland, Ohio Chestnut Hill, Massachusetts
DAVID D WIRTSCHAFTER, MD
GILBERT I FURMAN, MD VINOD K PAUL, MD, PhD
Los Angeles, California
West Covina, California New Delhi, India
JANUSZ GADZINOWSKI, MD, PhD JEFFREY J POMERANCE, MD, MPH THOMAS E WISWELL, MD
Poznan, Poland Pasadena, California Orlando, Florida
THE OFFICIAL JOURNAL OF THE SECTION ON PERINATAL PEDIATRICS
AMERICAN ACADEMY OF PEDIATRICS

Volume 28 Supplement 3 December 2008

Proceedings of the First International


Conference for Meconium Aspiration Syndrome
and Meconium-induced Lung Injury

GUEST EDITORS

Gilbert I Martin, MD
Dharmapuri Vidyasagar, MD

This supplement is sponsored by the Division of Neonatology, Department of Pediatrics


at the University of Illinois, NIH, INO Therapeutics, Thrasher Research Fund,
and Section on Perinatal Pediatrics, American Academy of Pediatrics
Contact information

SITE LICENCES
Journal of Perinatology is published by Nature Publishing Group, a division of Macmillan
Publishers Ltd. The Journal is the official journal of the Section on Perinatal Pediatrics, The Americas:
American Academy of Pediatrics. It is also the official publication of the National Perinatal Tel: +1 800 221 2123. Fax: +1 212 689 9711. E-mail: institutions@natureny.com
Association. Asia Pacific:
Tel: +81 3 3267 8769. Fax: +81 3 3267 8746. E-mail: institutions@naturejpn.com
Scope. Journal of Perinatology provides all members of the perinatal/neonatal healthcare team
with original information pertinent to improving maternal, fetal and neonatal care. The scope of Australia and New Zealand:
the Journal reflects the multidisplinary nature of the subject; its coverage includes maternal and Tel: +61 3 9825 1160. Fax: +61 3 9825 1010. E-mail: nature@macmillan.com.au
fetal medicine, neonatal medicine, and follow-up of the high-risk infant. Journal of Perinatology India:
publishes peer-reviewed clinical research articles, state-of-the-art reviews relevant to clinical Tel: +91 124 288 1054. Fax: +91 124 288 1053. E-mail: harpal@nature.com
management, commentaries, letters to the editor, and brief communications including case Rest of the World:
reports and educational vignettes. These articles embrace the full scope of the specialty, including Nature Publishing Group, The Macmillan Building, 4 Crinan Street, London, N1 9XW, UK
clinical, professional, political, administrative and educational aspects. The Journal also explores Tel: +44 (0) 20 7843 4759. Fax: +44 (0) 20 7843 4998. E-mail: institutions@nature.com
legal and ethical issues, neonatal technology and product development. All manuscripts are
original and initially published exclusively in the Journal. The audience includes all those who PRINT SUBSCRIPTIONS
participate in perinatal/neonatal care, including: neonatologists, perinatologists, pediatricians, All customers (excluding Japan, Korea and China):
obstetricians/gynecologists, neonatal and perinatal nurses, respiratory therapists, pharmacists, Customer Service Department, Nature Publishing Group, Houndmills, Basingstoke, Hants, RG21 6XS,
social workers and nutritionists. UK. Tel: +44 (0) 1256 329242. Fax: +44 (0) 1256 812358. E-mail: subscriptions@nature.com
Japan, Korea and China:
This journal is covered by Index Medicus, MEDLARS, Cambridge Scientific Abstracts, Elsevier Nature Publishing Group, Nature Japan, Chiyoda Building, 2-37 Ichigayatamachi, Shinjuku-ku,
BIOBASE/Current Awareness in Biological Sciences, Combined Cumulative Index to Tokyo 162-0843, Japan. Tel: +81 3 3267 8751. Fax: +81 3 3267 8746.
Pediatrics, and CINAHL Information System. E-mail: institutions@naturejpn.com
Editorial. Manuscripts should be submitted online at http://www.mts-jper.nature.com. Detailed Prices are applicable in the following regions: US dollars ($) for North, Central, South America
‘Instructions to Authors’ are available at this website, or in print in the first issue of the volume. and Canada; Euros (h) for all European countries (excluding the UK); Sterling (d) for UK and
rest of world. Please ensure you use the appropriate currency. All prices, specifications and details
Publisher. All business correspondence including enquiries about supplement publication and are subject to change without prior notification. Single issues are available. For information,
sponsorship opportunities should be sent to Journal of Perinatology, Nature Publishing Group, please contact: Subscriptions Dept, Nature Publishing Group, Brunel Road, Houndmills,
75 Varick Street, 9th Floor, New York, NY 10013-1917, US. Tel: +1 212 726 9296. Basingstoke, Hampshire RG21 9XS, UK. Tel: +44 (0) 1256 329242.
Fax: +1 646 563 7057. Fax: +44 (0) 1256 812358. E-mail: subscriptions@nature.com
Executive Editor: Joyce-Rachel John Journal of Perinatology. (ISSN 0743-8346) is published monthly by Nature Publishing Group,
Production Editor: Gene Wu 75 Varick Street, 9th Floor, New York, NY 10013-1917. Periodicals postage is paid at New York, NY,
US and additional offices. POSTMASTER: please send address changes to Journal of
Visit the Journal’s home pages for details of the aims and scope, readership, instructions to
Perinatology, Nature Publishing Group, 342 Broadway PMB 301, New York, NY
authors and how to contact the Editor and publishing staff. Use the website to order a
10013-3910.
subscription, reprints, a sample copy or individual articles.
Reprints and Permissions. For detailed information on reprints and permission requests,
Free to all readers. The complete text of all articles in the back archive 4 years after including instructions for obtaining these online, please visit http://www.nature.com/reprints/
publication; table of contents and abstracts for all articles published within the last 4 years index.html. Alternatively, for reprints, please e-mail: reprints@nature.com. For North America:
and the complete text of the January 2008 issue. Register to receive the table of contents by e-mail Tel: +1 212 726 9631. Fax: +1 212 679 0843. Outside North America: Tel: +44 (0) 20 7843 4967.
as each issue is published. Fax: +44 (0) 20 7843 4791. For permissions, please e-mail: ajpermissions@nature.com: Tel: +44
(0) 20 7843 4613.
2008 Subscriptions
Advertisements. Enquiries concerning advertisements and supplements should be addressed
Institutional Subscriptions to: Alf Anderson, Advertising Manager, Nature Publishing Group, 75 Varick Street, 9th Floor,
New institutional policy New York, NY 10013-1917, US. Tel: +1 617 475 9231, Fax: +1 617 494 4960.
NPG has moved to a site license policy for institutional online access. Institutions may also E-mail: aanderson@boston.nature.com
purchase a separate print subscription. Supplements. Enquiries concerning supplements should be addressed to: Michelle Libby, Commercial
Subscribing to a site license Projects Executive. Tel: +1 617 475 9230. Fax: +1 617 494 4960. E-mail: m.libby@boston.nature.com
Contact your local sales representative for a tailored price quote for your institution. You will be Copyright r 2008 Nature Publishing Group
required to complete a NPG site license agreement. More information and contact details are ISSN 0743-8346
available at the http://www.nature.com/libraries. EISSN 1476-5543
Institutional print subscriptions All rights of reproduction are reserved in respect of all papers, articles, illustrations, etc, published
Orders can be placed with your regular subscription agent or through NPG – either online at in this journal in all countries of the world. All material published in this journal is protected by
www.nature.com/jp or by contacting our customer services department. Prices are as follows: copyright, which covers exclusive rights to reproduce and distribute the material. No material
published in this journal may be reproduced or stored on microfilm or in electronic, optical or
The Americas $423.00 Europe h336.00 UK/Rest of World d217.00 magnetic form without the written authorization of the publisher.
Authorization to photocopy material for internal or personal use, or internal or personal use of
Personal Subscriptions
specific clients, is granted by Nature Publishing Group to libraries and others registered with the
Personal customers who pay by personal check or credit card can either purchase a combined Copyright Clearance Center (CCC) Transactional Reporting Service, provided the relevant
print plus online subscription or an online-only subscription. Prices are as follows: copyright fee is paid direct to CCC, 222 Rosewood Drive, Danvers, MA 01923, US. Identification
code for Journal of Perinatology: 0743-8346/08.
Combined (print plus online)
Apart from any fair dealing for the purposes of research or private study, or criticism or review, as
The Americas $177.00 Europe h141.00 UK/Rest of World d91.00
permitted by Sections 107 or 108 of the US Copyright Law, this publication may be reproduced, stored or
Online-only transmitted in any form or by any means, only with the prior permission in writing of the publishers.
The Americas $160.00 Europe h127.00 UK/Rest of World d82.00 Typeset by Macmillan India Ltd, Bangalore, India

Whilst every effort is made by the publishers and editorial board to see that no inaccurate or misleading data, opinion or statement appears in this publication, they wish to make it clear that the data and
opinions appearing in the articles and advertisements herein are the responsibility of the contributor or advertiser concerned. Accordingly, the Publisher, the editorial committee and their respective
employees, officers and agents accept no liability whatsoever for the consequences of any such inaccurate or misleading data, opinion or statement. Whilst every effort is made to ensure that drug doses
and other quantities are presented accurately, readers are advised that new methods and techniques involving drug usage, and described within this publication, should only be followed in conjunction
with the drug manufacturer’s own published literature, and a medically qualified and licensed physician.
THE OFFICIAL JOURNAL OF THE SECTION ON PERINATAL PEDIATRICS
AMERICAN ACADEMY OF PEDIATRICS

Volume 28 Supplement 3 December 2008

Contents

REVIEWS

S1 Introduction: Proceedings of the First International Conference for Meconium Aspiration Syndrome
and Meconium-induced Lung Injury
GI Martin and D Vidyasagar

S3 Meconium aspiration syndrome: historical aspects


AA Fanaroff

S8 Mechanism(s) of in utero meconium passage


J Lakshmanan and MG Ross

S14 Obstetric approaches to the prevention of meconium aspiration syndrome


H Xu, S Wei and WD Fraser

S19 Delivery room management of the meconium-stained newborn


TE Wiswell

S27 Neonatal resuscitation guidelines for ILCOR and NRP: evaluating the evidence and developing a
consensus
J Kattwinkel

S30 Developing a systems approach to prevent meconium aspiration syndrome: lessons learned
from multinational studies
VK Bhutani

S36 Meconium aspiration syndrome requiring assisted ventilation: perspective in a setting with limited resources
S Velaphi and A Van Kwawegen

S43 Meconium aspiration syndrome: experiences in Taiwan


HC Lin, SY Wu, JM Wu and TF Yeh

S49 Continuous positive airway pressure and conventional mechanical ventilation in the treatment of meconium
aspiration syndrome
JP Goldsmith

Contents continuedy
S56 Treatment of MAS with PPHN using combined therapy: SLL, bolus surfactant and iNO
J Gadzinowski, K Kowalska and D Vidyasagar

S67 Nitric oxide and beyond: new insights and therapies for pulmonary hypertension
RH Steinhorn

S72 Pharmacotherapy for meconium aspiration


A Asad and R Bhat

S79 Extracorporeal membrane oxygenation: use in meconium aspiration syndrome


BL Short

S84 Role of iNO in the modulation of pulmonary vascular resistance


A Bin-Nun and MD Schreiber

S93 Neurodevelopmental outcome of infants with meconium aspiration syndrome: report of a study
and literature review
N Beligere and R Rao

S102 Studies of meconium-induced lung injury: inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya

S108 Angiotensin II in apoptotic lung injury: potential role in meconium aspiration syndrome
BD Uhal and A Abdul-Hafez

S113 Toxic effects of different meconium fractions on lung function: new therapeutic strategies for
meconium aspiration syndrome?
OD Saugstad, PA Tølløfsrud, P Lindenskov and CA Drevon

S116 The role of complement in meconium aspiration syndrome


TE Mollnes, A Castellheim, PHH Lindenskov, B Salvesen and OD Saugstad

S120 Phospholipase A2 in meconium-induced lung injury


P Kääpä and H Soukka

S123 Meconium-induced release of nitric oxide in rabbit alveolar cells


R Fontanilla, A Zagariya and D Vidyasagar

S127 Intracellular and extracellular serpins modulate lung disease


DJ Askew and GA Silverman

Copyright r 2008 Nature Publishing Group


Subscribing organisations are encouraged to copy and distribute
this table of contents for internal non-commercial purposes
This issue is now available at:
www.nature.com/jp
Journal of Perinatology (2008) 28, S1–S2
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Introduction: Proceedings of the First International Conference
for Meconium Aspiration Syndrome and Meconium-induced
Lung Injury
GI Martin and D Vidyasagar

Journal of Perinatology (2008) 28, S1–S2; doi:10.1038/jp.2008.176 forum for discussion of new findings and also identified knowledge
gaps in management and pathophysiology of MILI. Clinical and
The history of the word ‘meconium’ holds interest for all of us laboratory investigators around the world were invited to discuss
involved in perinatal care. It is believed that Aristotle coined this their experiences and research in this field. The Editorial Board of
term, which has been anglicized to mean ‘opium like.’ As opium is the Journal of Perinatology kindly agreed to publish the articles
derived from poppy seeds and when processed the resulting poppy from the speakers at the symposium in this special supplement.
seed sludge is black and tarry the word was an appropriate A summary of conclusions from the presentations at the
description. conference is presented below.
Meconium-stained amniotic fluid occurs in approximately 13% The conference was divided into four sessions. The first session
of live births. Meconium aspiration syndrome (MAS) occurs in was devoted to discussions regarding the history of MAS and the
5 to 10% of infants born through meconium-stained amniotic global burden of MAS. Developed countries have noticed a steady
fluid. When MAS occurs, there is an increase in neonatal mortality decrease in mortality and morbidity from MAS, but developing
and morbidity. Great progress has been made in the improvement countries such as India and Africa share a major burden of birth
of survival of infants with MAS. Great progress in management has asphyxia and MAS. To reduce unacceptably high infant mortality rate
been made since first description of the pathophysiology and poor in developing countries, it was concluded that more research is
outcome of infants with MAS in 1975.1 These include improved needed both in clinical management and in understanding of
intrapartum and post-delivery management of MAS. Although there MAS and MILI.
is a significant decrease in the occurrence of MAS and associated The second session was devoted to understand the mechanisms
mortality in developed countries MAS remains a major problem in of in utero passage of meconium in the fetus and potential
developing countries. pathways that can be blocked to reduce MAS. Mechanisms of
The physiological effects of MAS owing to airway obstruction vascular remodeling in the fetus that may contribute to severe
and associated increased pulmonary vascular resistance are well persistent pulmonary hypertension of newborn was described in
studied. However, the mechanism of meconium-induced infants with severe MAS. Speakers identified several potential areas
inflammation and subsequent meconium-induced lung injury of research in the future.
(MILI) are just being investigated. In recent years, there is an The third session was devoted to review the current practices of
accumulation of evidence from investigators from different perinatal, intrapartum and post-neonatal management of MAS.
institutions that MAS also causes inflammatory reaction and It was concluded that the existing evidence from the literature
cellular injury. These are new findings; the impact of these indicates that amnioinfusion in the presence of meconium-stained
findings on management is yet to be determined. amniotic fluid or oropharyngeal suction of fetus being delivered
‘The First International Meeting for MILI’ was held on 23 and and routine post-delivery endotracheal suctioning have no
24 March 2007 in Chicago. The organizing committee included: beneficial effects in preventing or reducing MAS. There were
Vidyasagar, Bhat, Bhutani, Wiswell, Gadzinowski, Saugsted and questions in developing countries, where resources are limited for
Zagariya. The conference was partially funded by a grant (to DV) monitoring the fetus to the above therapies. Therefore, there is a
from NHLBI (Grant no. HL090484-01) to promote the international need for studies in developing countries to evaluate the effectiveness
collaboration of researchers in the field. The symposium provided a of these practices.
The investigators from India and South Africa stated that
Correspondence: Dr D Vidyasagar, Division of Neonatology, Department of Pediatrics, The
University of Illinois at Chicago, 840 South Wood Street, Chicago, IL 60517, USA.
developing countries with limited resources of fetal monitoring and
E-mail: dsagar@uic.edu delivery room care should develop strategies appropriate for their
Introduction
S2

circumstance. They expressed that there is a need for collaborative the globe. The conference allowed free exchange of ideas among
studies to improve the efficacy of clinical management of MAS the researchers. In this supplement of the Journal of Perinatology
babies. we are publishing the articles submitted by the speakers. The
In the fourth session, research data regarding MILI and articles are arranged into two groups: the first 15 articles deal with
apoptosis was presented. It was clear that meconium is a strong history, epidemiology and clinical aspects of MAS. The next seven
inducer of inflammation and cellular apoptosis. Several pathways articles deal with the cellular mechanisms of MILI. We hope the
were identified for the induction of apoptosis. This session laid readers of the Journal of Perinatology will find the information
a firm foundation for future studies and potential collaboration timely and useful.
among interested investigators. The last session was devoted to
reviewing the newer treatment modalities (iNO therapy, ECMO) Acknowledgments
and long-term outcomes. It was stressed that iNO therapy
We thank all the authors for their patience and for their cooperation.
successfully decreases PPHV.
We also thank the reviewers for their diligent efforts. And special thanks to the
ECMO was ultimate therapy for non-responding patients. It was editorial staff Ms Sandy Skelley for her valuable efforts in making this project
noted that in developed countries the need for ECMO in MAS has a success. We gratefully acknowledge the grant support from, iNO therapeutics,
decreased. Other discussions included the use of room air and the NIH for enabling us to conduct the symposium and The Thrasher
resuscitation of MAS (particularly in developing countries), use of Foundation and the Perinatal Section of AAP for their support to publish this
antibiotics, steroids and other supportive pharmacologic Supplement.
treatments. These modalities are of significance in developing
countries and need to be studied.
The long-term outcome of infants with MAS was reviewed. Disclosure
Existing reports consist of diverse population and mixed therapies. D Vidyasagar has received grant support from INO Therapeutics and NIH.
It was noted that there was an increased incidence of GI Martin has declared no financial interests.
neurodevelopmental delay in babies with MSAF and MAS. These
observations indicate the need for larger follow-up studies of
infants with MSAF and MAS. Reference
In summary, the conference, which was the first of its kind on 1 Vidyasagar D, Harris V, Pildes RS. Assisted ventilation in infants with meconium
MAS, brought together established investigators in the field around aspiration syndrome Pediatrics 1975; 56(2): 208–213.

Journal of Perinatology
Journal of Perinatology (2008) 28, S3–S7
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Meconium aspiration syndrome: historical aspects
AA Fanaroff
Department of Pediatrics, Case Western Reserve University, Rainbow Babies & Children’s Hospital/University Hospitals
Case Medical Center, Cleveland, OH, USA

protein, bile acids and salts as well as drug metabolites.


The meconium aspiration syndrome (MAS) is a common problem that The black-green color results from bile pigments. When
continues to concern perinatologists and neonatologists. MAS is defined as aspirated into the lung, either in the fetus or newly born infant,
respiratory distress in an infant born through meconium-stained amniotic meconium may stimulate the release of cytokines and other
fluid (MSAF) whose symptoms cannot be otherwise explained. This disorder vasoactive substances that lead to cardiovascular and inflammatory
may be life threatening, complicated by respiratory failure, pulmonary air responses.
leaks and persistent pulmonary hypertension. Approaches to the prevention In the fetus, passage of meconium occurs physiologically early
of MAS have changed over time with collaboration between obstetricians in gestation, when it contributes to alkaline phosphatase in
and pediatricians forming the foundations for care. This report details the amniotic fluid. Abramovich1 noted that fetal defecation diminishes
management of babies delivered with associated MSAF before the after 16 weeks and ceases by 20 weeks, concurrent with innervation
accumulation of evidence for best practice through appropriately powered, of the anal sphincter. At that time, the rectum appears to be filled
prospective randomized controlled trials. with meconium. From approximately 20 to 34 weeks, fetal passage
Journal of Perinatology (2008) 28, S3–S7; doi:10.1038/jp.2008.162 of meconium was infrequent.2 Most newborn infants who pass
meconium are mature (term). Many are postmature and the
babies may exhibit peeling skin, long fingernails and decreased
vernix. The vernix, umbilical cord and nails may be meconium
stained, depending upon how long the infant has been exposed
in utero. In general, nails will become stained after 6 h and vernix
Background after 12 to 14 h of exposure.
Meconium, the fecal material that accumulates in the fetal The passage of meconium normally occurs within the first 24 to
colon throughout gestation, is a term derived from the Greek 48 h after birth. However, the passage of fetal meconium, resulting
mekoni, meaning poppy juice or opium. Commencing with in MSAF, occurs in approximately 8 to 25% of all deliveries,
Aristotle’s observation of the association between meconium primarily in situations of advanced fetal maturity or fetal stress.
staining of the amniotic fluid and a sleepy state or neonatal Most infants who are delivered with MSAF are beyond the 37 weeks
depression, obstetricians have been concerned about fetal well of gestation and meconium rarely appears in amniotic fluid before
being in the presence of meconium-stained amniotic fluid (MSAF). 32 weeks of gestation. The premature infant lacks ‘motilin,’ while
Fetal hypoxic stress may stimulate colonic activity, resulting in the assists in moving meconium through the lower gastrointestinal
passage of meconium, and also may stimulate fetal gasping track. The meconium aspiration syndrome (MAS), associated with
movements that result in meconium aspiration. aspiration or perhaps diffusion of meconium into the fetal airways,
Meconium is a sterile, thick, black-green, odorless material first occurs in about 5% of these infants.3,4
observed in the fetal intestine during the third month of gestation. Meconium aspiration syndrome is defined as respiratory distress
Meconium results from the accumulation of debris, including in an infant born through MSAF whose symptoms cannot be
desquamated cells from the intestine and skin, gastrointestinal otherwise explained. Aspirated meconium can interfere with
mucin, lanugo hair, fatty material from the vernix caseosa, normal breathing by several mechanisms. They include airway
amniotic fluid and intestinal secretions. It comprises 72 to 80% obstruction, chemical irritation, infection and surfactant
water and contains cholesterol and its precursors, lipids, enzymes inactivation (see Figure 1). When aspirated into fetal lungs,
including pancreatic phospholipase A2, mucopolysaccharides, meconium particles mechanically obstruct the small airways.
Meconium or the chemical pneumonitis it causes inhibits
Correspondence: Dr AA Fanaroff, Department of Pediatrics, MS RBC 6040, Rainbow Babies &
Children’s Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
surfactant function, and inflammation of lung tissue
E-mail: aaf2@case.edu contributes further to small airway obstruction. The chest
Meconium aspiration syndrome
AA Fanaroff
S4

Rationale for routine endotracheal intubation to


prevent MAS
The foundations for routine endotracheal intubation of infants for
MSAF was based on what today would be considered entirely
anecdotal evidence. Gregory et al.7 wished to document the
prevalence of MSAF, the nature of the MSAF (thick or watery, thick
but still liquid, particulate) and the incidence of respiratory distress
when there was meconium present. In a prospective collection of
data they reported MSAF in 8.8% of 1000 consecutive newborn
infants over a 6 month time period. They aspirated the upper and
lower airways as soon as possible ‘sometimes before the first breath’
Figure 1 Pathophysiology of meconium aspiration syndrome. From
in 80 of the 88 infants with MSAF.7 If the trachea contained
Fanaroff and Martin’s Neonatal Perinatal Medicine, Diseases of the Fetus meconium, intubation and suctioning were repeated until the
and Infant 8th edition, edited by R Martin, A Fanaroff and M Walsh, Mosby airway had been cleared. Fifty-seven infants had meconium in
Elsevier, 2006. their mouth, 62 had meconium in the stomach and 8 had
meconium in the trachea but none in the mouth or larynx. Half of
the infants with meconium in the airway (23 of 46) had abnormal
typically appears barrel-shaped, with an increased anterior– chest X-rays and 16 (70%) of these were symptomatic. They
posterior diameter caused by overinflation. Auscultation reveals subsequently compared incidence of air leaks in 35 hospitalized
rales and rhonchi. These signs usually are seen immediately infants with MAS from their institution (2 of 35; 6%) with
after birth. 15 outborn infants with MAS including 6 of the 15 (40%) with air
The respiratory manifestations include respiratory distress, leaks. They concluded that ‘all infants born through thick,
tachypnea, cyanosis, and air trapping together with reduced particulate or ‘pea soup’ meconium should have their trachea
pulmonary compliance. Cleary and Wiswell3 have proposed severity aspirated immediately after birth by the most experienced person in
criteria to define MAS: (1) mild MAS is disease that requires less the delivery room and receive thoracic physical therapy and
than 40% oxygen for less than 48 h, (2) moderate MAS is disease postural drainage for the first 8 h of life, if the meconium is
that requires more than 40% oxygen for more than 48 h with no recovered from the trachea and/or the chest X-ray is consistent with
air leak and (3) severe MAS is disease that requires assisted aspiration.’ Ting and Brady8 a year later following a retrospective
ventilation for more than 48 h and is often associated with chart review noted that of 97 infants with thick meconium who
persistent pulmonary hypertension. Of infants in whom the MAS received endotracheal suctioning at birth, 27 became symptomatic
develops, more than 4% die, accounting for 2% of all perinatal and only one died. On the other hand, 28 infants with thick
deaths. meconium who were not suctioned, 16 became symptomatic and
An estimated 25 000 to 30 000 cases and 1000 deaths related to 7 died. Soon thereafter Carson et al.9 published their combined
MAS occur annually in the United States with many more obstetric and pediatric approach to prevent MAS. In a study
cases in developing countries. However Yoder et al.5 documented originally intended to examine the effectiveness of pulmonary
a decline in the incidence of MAS in the United States lavage for MAS, they performed routine intrapartum pharyngeal
(from 5.8 to 1.5%Falmost a fourfold reduction) during the suctioning with a DeLee catheter with the infants head on
period 1990 to 1997, which they attributed to a 33% reduction in the perineum, before the onset of respirations, thereafter
the incidence of births at more than 41 weeks’ gestation. This inspecting the larynx when the baby delivered and intubating
probably can be explained by reductions in the number of infants and suctioning if meconium was present. Their treatment group
passing meconium (in association with advanced maturity) and in was compared to historical controls, MAS occurred in 1 of 273
the rate of intrauterine hypoxia (for which the risk is increased (0.4%) with routine suctioning and 18 of 947 (1.9%) (P ¼ 0.07) in
in post-term pregnancies). the non-suctioned group. They noted that routine suctioning
Acute intrapulmonary meconium contamination induces a of the trachea under direct vision after delivery is rarely necessary
concentration-dependent pulmonary hypertensive response, with but should be performed if meconium is visualized at the vocal
15 to 20% of infants with the MAS showing persistent pulmonary cords, and tracheobronchial lavage with saline may add to the
hypertension. However, evidence of a long-term process of respiratory morbidity. They indicated that ‘No deaths or severe
muscularization of distal pulmonary arterioles in infants with the cases of MAS have occurred since institution of the obstetric
MAS who died suggests that factors other than meconium suctioning procedure’ and concluded that ‘routine intrapartum
aspiration (for example, chronic intrauterine hypoxemia) may pharyngeal suctioning with a DeLee catheter of infants with
contribute to the pulmonary symptoms.6 meconium staining has significantly reduced the incidence and

Journal of Perinatology
Meconium aspiration syndrome
AA Fanaroff
S5

severity of MAS’ despite a P-value not supporting such a During the study period when the infants were hyperventilated, as
conclusion. the PaCO2 decreased from 48.9 to 28.3 mm Hg (P<0.02), the mean
On the basis of these studies by Gregory,7 Ting8 and Carson,9 PAP decreased by 36 mm Hg (P<0.001) to sub-systemic pressure
the commonest approach to MSAF, whether thick, watery or levels, and the mean alveolar/arterial oxygen difference decreased
particulate, was for the obstetrician to immediately suction the by 146 mm Hg (P<0.001). Following the decrease in PAP, patients
nasopharynx as soon as the head appeared on the perineum. were mechanically ventilated to maintain PaCO2 in the range of
Thereafter, visualization of the cords by direct laryngoscopy and 25 to 30 mm Hg until pulmonary hypertension gradually resolved
suctioning of the trachea through the endotracheal tube if in the six survivors. On the basis of this small uncontrolled study,
meconium was detected was the routine adopted at most centers. hyperventilation to lower pCO2 levels became the standard of care,
This was performed before stimulation of the infant or and in light of present knowledge probably contributed to a
administration of positive pressure ventilation. This approach number of deaf survivors.
remained the standard of care until challenged by a series of
prospective randomized trials.
Extracorporeal membrane oxygenation and MAS
The history of the introduction of extracorporeal membrane
Corticosteroids for MAS oxygenation (ECMO) into neonatal perinatal medicine is closely
Yeh et al.10 evaluated the efficacy of glucocorticoids in the aligned with the management of MAS. Bartlett et al.12 documented
treatment of infants with MAS. Using a ‘double blind’ technique 16 moribund newborn infants with respiratory failure who were
they randomized 35 infants with MAS to hydrocortisone or a lactose treated with ECMO for 1 to 8 days. Their diagnoses and outcomes
placebo. No significant differences in arterial PO2, PCO2, pH, included respiratory distress syndrome, four patients (two improved,
A-aDO2 gradients, requirement for assisted ventilation, or in one survived); MAS, eight patients (four improved, three survived);
survival were observed between the groups. In the placebo group, a persistent fetal circulation (some with diaphragmatic hernia), four
significant decrease in respiratory distress score occurred at 48 to patients (three improved, two survived). Intracranial bleeding
72 h of age (P<0.01); in hydrocortisone-treated infants, it was seen occurred in 43% accounting for most of the deaths. In a parallel
only after 72 h. The infants in the steroid group took a significantly series of 21 infants treated with conventional ventilator therapy, the
longer period of time to wean to room air than those in the control mortality rate was 90% and intracranial bleeding occurred in 57%.
group (68.9±9.6 vs 36.6±6.9 h) (P<0.01). They concluded that They modestly concluded that ECMO provided life support and
hydrocortisone is not recommended for the treatment of MAS. gains time in newborn respiratory failure. In high mortality risk
infants, the rate of survival is higher and that of intracranial
bleeding is lower with ECMO than with optimal ventilator
MAS and pulmonary hypertension management. So dawned the ECMO era, and ultimately the
Pulmonary hypertension was a significant comorbidity in infants survival of many moribund infants with pulmonary hypertension
with MAS and frequently the cause of death. The management of and respiratory failure, including those with MAS. Indeed, in the
pulmonary hypertension was haphazard and the outcomes ECMO registry, the highest survival rates (>90%) were seen
unpredictable. Fox et al.11 reported on 10 patients clinically in the patients with MAS who qualified for ECMO. With the
diagnosed as having perinatal aspiration syndromes together with evidence-based approach to prevention of MAS and the initiation of
pulmonary hypertension. These infants were either term or better less-invasive therapies, including inhaled nitric oxide,
postmature babies and at catheterization had the following surfactant therapy, surfactant lavage and various modes of
characteristics: (1) systemic or supra-systemic levels of pulmonary mechanical ventilation, including high-frequency ventilation, it is
artery pressure (PAP) (range, 50 to 117 mm Hg); (2) a degree of rare for an infant to require ECMO for MAS.13–16
pulmonary hypertension not related to the degree of aspiration
evident on chest roentgenograms; (3) evidence of right-to-left
shunting at the ductal or foramen ovale level and (4) sustained Evidence-based practice to prevent MAS
severe hypoxemia despite 100% inspired oxygen concentration. On the basis of the evidence from non-randomized studies,6–8 to
Indwelling pulmonary artery catheters were used for continuous reduce the incidence and severity of MAS the recommendation was
monitoring of PAP in these 10 infants with severe persistent to intubate and suction all babies born through thick meconium.
pulmonary hypertension of the newborn. The labile nature of PAP, However, for term babies who are vigorous at birth endotracheal
with changes up to 50 mm Hg, was documented. PAP in the eight intubation may be both difficult and unnecessary. This concept of
infants with supra-systemic pulmonary hypertension was analyzed routine intubation was challenged first by Faciglia using a
at the time of maximum decrease in pressure (mean 36.1 mm Hg) concurrent observational study.17,18 He showed no differences in
and physiologic measurements were compared over an 8-h period. outcome with and without routine intubation. Linder et al.19 in a

Journal of Perinatology
Meconium aspiration syndrome
AA Fanaroff
S6

prospective controlled trial, perhaps inadequately randomized, Long-term consequences


assessed the need for intubation of vigorous term meconium- The initial follow-up report on follow-up of MAS by Marshall
stained infants. Of 572 infants, meconium-stained infants, et al.27 in 1978 included a 1-year cohort of 17 patients,
delivered vaginally, with 1-min Apgar score of 9 or 10, none of 264 representing 3.7% of all admissions to their unit. Four patients
expectantly managed infants developed MAS and 2 of 308 intubated (23.5%) died of acute respiratory failure and two patients with MAS,
infants developed MAS. They concluded that intubation may be and persistence of the fetal circulation required cardiac
superfluous in a vigorous infant born through MSAF and practice catheterization to exclude cyanotic congenital heart disease. None
started to change. of the survivors had persistent chronic lung disease but two of three
A decade later in international prospective, randomized, patients with MAS and seizures had significant psychomotor
controlled trial assessing need for intubation of apparently vigorous retardation at follow-up examination. The current data indicate
meconium-stained infants, Wiswell et al.20 enrolled 2094 babies that there is an increased prevalence of asthmatic symptoms and
born with MSAF to either intubation and suctioning or to expectant abnormal bronchial reactivity among survivors of the MAS.
management. They found no significant differences in either MAS
or any respiratory disorders supporting the concept that it was not
necessary to intubate vigorous term infants irrespective of the Conclusion
nature of the MSAF (thick or thin). Halliday21 accumulated four
Meconium aspiration syndrome remains a challenging condition
randomized controlled trials of endotracheal intubation at birth in
most notably in developing countries. In the USA avoidance of
vigorous term meconium-stained babies, which were identified.
post-term pregnancies, improved intrapartum monitoring and
Meta-analysis of these trials does not support routine use of
amnioinfusion have dramatically reduced the incidence of MAS.
endotracheal intubation at birth in vigorous meconium-stained
Whereas there is inconclusive evidence as to the optimal ventilator
babies to reduce mortality, MAS, other respiratory symptoms or
strategy for MAS, surfactant replacement therapy has been
disorders, pneumothorax, oxygen need, stridor, HIE and
beneficial and surfactant lavage is still being explored. Inhaled
convulsions. However, the event rates of many of these outcomes is
nitric oxide has significantly enhanced the treatment of pulmonary
low in the reported trials making reliable estimates of treatment
hypertension and substantially reduced the need for ECMO. Despite
effect impossible.
optimal perinatal management, infants born through MSAF may
Vain et al.22 finally shattered all the anecdotal data when they
still develop MAS and have morbid or even fatal outcomes.
reported on their prospective randomized trial that routine
intrapartum oropharyngeal and nasopharyngeal suctioning of
term-gestation infants born through MSAF does not prevent MAS.
Disclosure
Avroy Fanaroff has received consulting fees from NATUS.

Amnioninfusion References
Amnioinfusion has been advocated as a technique to reduce the 1 Abramovich DR, Gray ES. Physiologic fetal defecation in midpregnancy. Obstet
incidence of meconium aspiration and to improve neonatal Gynecol 1982; 60: 294.
2 Ramon Y, Cajal CL, Martinez RO. Defecation in utero: a physiologic fetal function. Am
outcome. However, meconium passage may predate labor so that a
J Obstet Gynecol 2003; 188: 153.
large proportion of women with MSAF have infants with meconium 3 Cleary GM, Wiswell TE. Meconium-stained amniotic fluid and the meconium
in the trachea or bronchioles before meconium passage has been aspiration syndrome: an update. Pediatr Clin North Am 1998; 45: 511–529.
noted clinically and before amnioinfusion can be performed.23 4 Dargaville PA, Copnell B. The epidemiology of meconium aspiration syndrome:
Fraser et al.24 reported that for women in labor who have thick incidence, risk factors, therapies, and outcome. Pediatrics 2006; 117: 1712.
meconium staining of the amniotic fluid, amnioinfusion did not 5 Yoder BA, Kirsch EA, Barth WH, Gordon MC. Changing obstetric practice associated
with decreasing incidence of meconium aspiration syndrome. Obstet Gynecol 2002;
reduce the risk of moderate or severe MAS, perinatal death, or other 99: 731–739.
major maternal or neonatal disorders.25 Xu et al.26 summarized 6 Murphy JD, Vawter GF, Reid LM. Pulmonary vascular disease in fatal meconium
the evidence for the use of amnioinfusion to prevent MAS. They aspiration. J Pediatr 1984; 104: 758–762.
concluded that in clinical settings with standard peripartum 7 Gregory GA, Gooding CA, Phibbs RH, Tooley WH. Meconium aspiration in infantsFa
surveillance, the evidence does not support the use of prospective study. J Pediatr 1974; 85: 848–852.
8 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obstet Gynecol
amnioinfusion for MSAF. In settings with limited peripartum
1975; 122: 767–771.
surveillance, where complications of MSAF are common, 9 Carson B, Losey R, Bowes Jr WA, Simmons MA. Combined obstetric and pediatric
amnioinfusion appears to reduce the risk of MAS. As always they approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976;
noted that further studies were needed. 126: 712.

Journal of Perinatology
Meconium aspiration syndrome
AA Fanaroff
S7

10 Yeh TF, Srinivasan G, Harris V, Pildes RS. Hydrocortisone therapy in meconium 19 Linder N, Aranda JV, Tsur M, Matoth I, Yatsiv I, Mandelberg H et al. Need for
aspiration syndrome: a controlled study. J Pediatr 1977; 90: 140–143. endotracheal intubation and suction in meconium-stained neonates. J Pediatr 1988;
11 Fox WW, Gewitz MH, Dinwiddie R, Drummond WH, Peckham GJ. Pulmonary 112: 613–615.
hypertension in the perinatal aspiration syndromes. Pediatrics 1977; 59: 205–211. 20 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al.
12 Bartlett RH, Gazzaniga AB, Huxtable RF, Schippers HC, O’Connor MJ, Jefferies MR. Delivery room management of the apparently vigorous meconium-stained neonate:
Extracorporeal circulation (ECMO) in neonatal respiratory failure. J Thorac results of the multicenter, international collaborative trial. Pediatrics 2000; 105
Cardiovasc Surg 1977; 74: 826–833. (1 Part 1): 1–7.
13 El Shahed AI, Dargaville P, Ohlsson A, Soll RF. Surfactant for meconium aspiration 21 Halliday HL. Endotracheal intubation at birth for preventing morbidity and mortality in
syndrome in full term/near term infants. Cochrane Database Syst Rev 2007: vigorous, meconium-stained infants born at term. Cochrane Database Syst Rev 2001:
CD002054. CD000500.
14 Greenough A, Pulikot A, Dimitriou G. Prevention and management of meconium 22 Vain NE, Szyld EG, Prudent LM, Wiswell TE, Aguilar AM, Vivas NI. Oropharyngeal and
aspiration syndromeFassessment of evidence based practice. Eur J Pediatr 2005; nasopharyngeal suctioning of meconium-stained neonates before delivery of their
164: 329–330. shoulders: multicentre, randomised controlled trial. Lancet 2004; 364: 597–602.
15 Wiswell TE, Knight GR, Finer NN, Donn SM, Desai H, Walsh WF et al. A multicenter, 23 ACOG Committee Obstetric Practice. ACOG Committee Opinion Number 346, October
randomized, controlled trial comparing surfaxin (lucinactant) lavage with standard 2006: amnioninfusion does not prevent meconium aspiration syndrome. Obstet
care for treatment of meconium aspiration syndrome. Pediatrics 2002; 109: Gynecol 2006; 108: 1053.
1081–1087. 24 Fraser WD, Hofmeyr J, Lede R, Faron G, Alexander S, Goffinet F et al., Amnioinfusion
16 Nakamura T, Matsuzawa S, Sugiura M, Tamura M. A randomised control study of Trial Group. Amnioinfusion for the prevention of the meconium aspiration syndrome.
partial liquid ventilation after airway lavage with exogenous surfactant in a meconium N Engl J Med 2005; 353: 909–917.
aspiration syndrome animal model. Arch Dis Child Fetal Neonatal Ed 2000; 82: 25 Ross MG. Meconium aspiration syndromeFmore than intrapartum meconium.
F160–F162. N Engl J Med 2005; 353: 946–948.
17 Falciglia H. Failure to prevent meconium aspiration syndrome. Obstet Gynecol 1988; 26 Xu H, Hofmeyr J, Roy C, Fraser WD. Intrapartum amnioinfusion for meconium-stained
71: 349–353. amniotic fluid: a systematicreview of randomised controlled trials. British J Obstet
18 Falciglia HS, Henderschott C, Potter P, Helmchen R. Does DeLee suction at the Gynecol 2007; 114: 383–390.
perineum prevent meconium aspiration syndrome? Am J Obstet Gynecol 1992; 167: 27 Marshall R, Tyrala E, McAlister W, Sheehan M. Meconium aspiration syndrome.
1243–1249. Neonatal and follow up study. Am J Obstet Gynecol 1978; 131: 672–676.

Journal of Perinatology
Journal of Perinatology (2008) 28, S8–S13
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Mechanism(s) of in utero meconium passage
J Lakshmanan and MG Ross
Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA, USA

have been attributed as risk factors responsible for meconium


To use sheep and rat models and demonstrate that stressors activate fetal passage as well as the potential meconium aspiration in utero.
glucocorticoid (GC) system, corticotrophin-releasing factor (CRF) system Despite these risk factors, presently very little is known of the
and cholinergic neurotransmitter system (ChNS) leading to propulsive cascade of events that leads to meconium passage in the
colonic motility and in utero meconium passage. Immunohistochemical newborn immediately after birth or to the mechanisms
studies (IHS) were performed to localize GC-Receptors, CRF-receptors and contributing ‘premature’ meconium passage in utero. There
key molecules of ChNS in sheep fetal distal colon. CRF expression in are no small or large animal models that could be extrapolated
placenta and enteric endocrine cells in fetal rat system were examined and to humans. We review here recent studies gathered in our
the effects of acute hypoxia on in utero meconium passage was tested. IHS laboratory in rat and sheep models directed toward understanding
confirmed localization and gestation dependent changes in GC-Rs, CRF-Rs the cellular and molecular basis of stress-induced in utero
and cholinergic markers in sheep fetal colon. Rat placenta and enteric meconium passage.
endocrine cells express CRF and gastrointestinal tract express CRF-Rs.
Hypoxia is a potent inducer of meconium passage in term fetal rats. Stress is
a risk factor for in utero meconium passage and laboratory animal models
can be used to develop pharmacotherapy to prevent stress-induced in utero
meconium passage.
Stress and gut motility: lessons learned from
Journal of Perinatology (2008) 28, S8–S13; doi:10.1038/jp.2008.144
laboratory animals
Although impact of emotional stress on gastric and colonic motor
activity has long been known in humans only during the past two
decades, much attention has been paid to understand the neural
circuitry and hormonal effectors that mediate effects of stress on
Introduction gut functions in animal models (see review7,8). The rat is a widely
Newborn meconium passage, a developmentally programmed used animal model for stress-related alteration on gut motor
event, normally occurs within the first 24–48 h after birth, functions, although limited investigation has also been performed
although little is known of the regulatory process. Clinically, a in mouse models.9 Inhibition of gastric emptying, gastric secretion,
delay in newborn meconium passage has been observed in infants gastric contraction and stimulation of colonic motility and
born with Hirschprung disease, a defect involving the absence of defecation have now been well established in adult rats as the
intrinsic ganglia in colon.1 In contrast, we have no similar hallmarks of neurovisceral motor responses to stressors of
anatomical or neurochemical explanations for the meconium psychological, physical, chemical and immunological origin.10–13
staining observed in 7–22% of all term deliveries.2 Greater than Members of stress hormone family, more specifically
90% of cases of clinically observed meconium-stained amniotic corticotrophin-releasing factor (CRF) and urocortin, have been
fluid are noted in fetuses at or following 37 weeks gestation, being shown to mimic multifaceted acute responses to stress.8 Both
uncommon in preterm deliveries.3 Passage of meconium occurs hormones mediate their actions through CRF-receptor subtype 1
most often in deliveries beyond 40 weeks of gestation.4 An increased (CRF-R1) and CRF-receptor subtype 2 (CRF-R2). Both receptors
incidence of meconium passage in amniotic fluid is noted in the are encoded by distinct genes and both are G protein-coupled
presence of fetomaternal stress factors such as hypoxia and receptors linked to multiple intracellular signaling pathways.14
infection, independent of fetal maturation.5,6 On the basis of the Of the two CRF receptor subtypes, CRF-R1 stimulates colonic
clinical observations, fetal stress and gastrointestinal maturation motility and defecation, whereas CRF-R2 inhibits gastric emptying
and gastric contraction at times of stress.15 Anatomical,
Correspondence: Dr MG Ross, Department of Obstetrics and Gynecology, Harbor-UCLA
Medical Center, 1000 W. Carson Street, Box 3, Torrance, CA 90502, USA. physiological, pharmacological and biochemical studies have
E-mail: mikeross@ucla.edu provided unambiguous evidence that the CRF system
Mechanism of in utero meconium passage
J Lakshmanan and MG Ross
S9

(CRF, urocortins, CRF-R1 and CRF-R2) is the key player of novel hypoxic stress paradigm to demonstrate that hypoxic stress at
gastrointestinal motility disturbance during times of stress.7,16 term will provoke in utero meconium passage.21 The experimental
design includes exposure of term pregnant rats (term ¼ 22 days)
Hypothesis: in utero meconium passage is a fetal at 22 days of gestation to decreasing concentration of oxygen in a
neurovisceral motor response to stress stepwise manner as illustrated in Figure 2a. Fetuses removed from
uterine horn at the end of hypoxic exposure were alive and
We hypothesize that in utero meconium passage is a neurovisceral exhibited thick meconium staining as shown in Figure 2b, whereas
motor response to fetal stress and that the CRF system plays a control fetuses did not exhibit meconium.
critical role in the stimulation of cholinergically mediated fetal Examination of fetal and maternal plasma revealed a marked
colonic propulsive motor functions that trigger meconium passage. elevation in CRF levels,21 suggesting that the peripheral CRF
An integrated model proposed for fetal in utero meconium passage pathway may mediate the stimulation of fetal colonic motility and
is shown in Figure 1. The salient points of our hypothesis are as meconium passage. As acute hypoxia-induced meconium passage
follows: fetal–maternal stress leads to increased fetal plasma has a rapid onset, the mechanisms underlying the peripheral
glucocorticoids, which stimulate the synthesis of placental CRF.17 pathway most likely involve the activation (directly and neurally)
Stress-induced increase in glucocorticoids are also speculated to of fetal adrenal epinephrine and norepinephrine secretion as well
upregulate the CRF-R1 density18 on the myenteric neurons as well as sympathetic nervous system norepinephrine release, which in
as the expression of peripheral choline acetyltransferase,19 the turn stimulate placental CRF exoctyosis. Recent studies in our
enzyme that catalyzes the synthesis of acetylcholine, the principle laboratory indicate that rat placenta, similar to human placenta, is
neurotransmitter that regulates the motor activity of a site of CRF synthesis.22,23 In addition, enteric endocrine cells
gastrointestinal system. In conditions of stress, increased maternal expressing CRF in term rat fetal gut could also function as a
and/or fetal sympathetic tone will lead to increased circulating potential local source, as speculated in humans.24,25 Increased
levels of catecholamines, which are known stimulators of placental peripheral CRF in the fetus stimulates colonic motility through
CRF release.20 Thus, stress-induced activation of the CRF–CRF-R1 myenteric CRF-R1 and rapid meconium passage (Figure 3a).
system at the level of myenteric neurons will evoke release of The central CRF pathway involves activation of CRF neurons
acetylcholine, increased colonic smooth muscle motility and in paraventricular brain nuclei directly innervating colonic
in utero meconium passage. segments, as has been demonstrated in adult rats (Figure 3b).
Thus, acute hypoxic stress-induced in utero meconium passage
involves release of stored CRF protein pool from peripheral and/or
Stress is a potent stimulator of in utero meconium
central tissues. Ongoing studies in our laboratory also support the
passage in term fetal rats
expression of CRF-R1 in smooth muscle and enteric neuronal
Although clinical signs of stress due to hypoxia are frequently circuitry in term rat fetal gastrointestinal tract,26 lending strong
observed in human neonates born with meconium staining, there anatomical support for our hypothesis that intrauterine stress may
is no direct evidence to support the fact that hypoxic stress is a risk impact gut motility through CRF-R1 in a manner analogous to
factor for in utero meconium passage. We recently developed a stress-induced stimulation of colonic motility and defecation in
adult rats.15

Lessons learned from sheep model in support of


stress-induced in utero meconium passage
Sheep are used in our laboratory as a large animal model for
understanding the molecular mechanism of in utero meconium
passage.27–29 The larger size of the gut and longer
gestational age permit in vivo studies exploring gestation- and
hormone-dependent changes in gastrointestinal motility, and
in vitro organ bath studies examining smooth muscle contractility.
The large fetal size permits withdrawal of blood in sufficient
quantities for evaluating circulating hormone and cytokine levels.
In the following section, we summarize some of our investigations
Figure 1 An integrated model for stress-induced in utero meconium passage.
An interconnected pathway involving glucocorticoids, CRF and cholinergic
relevant to hormonal regulation of sheep fetal distal colonic
neurocircuitry systems stimulate colonic motility leading to in utero meconium motility as well as recent progress made on the ontogeny of
passage (detailed in the text). glucocorticoid receptor system, CRF system and cholinergic

Journal of Perinatology
Mechanism of in utero meconium passage
J Lakshmanan and MG Ross
S10

Oxygen content in chamber


25

20
% Oxygen
15

10

21
24
27

33
36
39
30
12
15
18

42
6
8
3
0

Time (min)

Figure 2 Hypoxic stress paradigm in term fetal rat with meconium passage. Oxygen content in hypoxic chamber is reduced in a stepwise manner from 21 to 8%
as indicated in the figure (left panel). A photograph of a fetal rat with meconium passage (arrows; right panel).

a b

Figure 3 Hypothesized mechanisms for hypoxia-induced in utero meconium passage. Acute hypoxia-induced in utero meconium passage could be mediated either by
(a) placental CRF release or (b) central hypothalamic paraventricular nucleus CRF. In both circumstances, CRF directly stimulates CRF-R1 localized on
cholinergic myenteric neurons.

synaptic circuitry system in fetal distal colonic segments. In suggesting that glucocorticoids regulate cholinergic contractile
addition, we discuss our recent advances in support of knowledge of maturation through ‘genomic’ actions. The findings obtained
CRF signaling system in the prevention of in utero meconium following glucocorticoid administration has provided partial
passage before term. support for our hypothesis that stress-mediated glucocorticoid
release may stimulate colonic motility and meconium passage.
Sheep fetal colon is a glucocorticoid target organ We also examined the in vivo effect of bethanechol on colonic
As the incidence of meconium-stained amniotic fluid increases muscular contractile and electromyogram activity in the sheep
with increase in gestational ages and fetal stress, we hypothesized fetus.28 We noticed increased strain gauze activity in response to
that fetal colon could be a glucocorticoid target organ with fetal bethanechol infusion in transverse but not in distal colon at 0.9
colonic motility function regulated by glucocorticoids.27 We gestation (135 days gestation, term ¼ 150 days). This finding
examined in vivo effects of betamethasone (a synthetic suggested the presence of a local inhibitor system for
glucocorticoid) in the presence and absence of thyroxine on cholinergic-dependent contractility in the distal colon before term,
in vitro colonic smooth muscle cholinergic contractility.27 Muscle perhaps serving as an intrinsic mechanism to prevent premature
strips prepared from sheep fetuses that received intramuscular meconium passage.
injection of betamethasone alone or in combination with thyroxine
exhibited greater contractile responsiveness to bethanechol Localization and gestation-dependent changes in glucocorticoid
(a cholinergic agonist) than muscle strips prepared from receptor expression in sheep fetal distal colon
vehicle-treated fetuses. These glucocorticoid-specific changes were To validate our hypothesis that sheep fetal colon is glucocorticoid
noticed 48 h after intramuscular injections of betamethasone, target organ, we undertook studies to evaluate the cellular

Journal of Perinatology
Mechanism of in utero meconium passage
J Lakshmanan and MG Ross
S11

localization and gestation-dependent changes in glucocorticoid endogenous CRF leading to increased motor activity and
receptors in sheep fetuses at very preterm (118–120 days), preterm meconium passage.
(130–132 days), near term (140–142 days) and term (146–147
days) gestation.30 Glucocorticoid receptor antibody elicited positive Cellular localization of CRF-binding protein in sheep
staining in smooth muscle layers as well as in myenteric and distal colon
submucosal neurons at all stages of development with maximal Corticotrophin-releasing factor-binding protein is a protein known
expression noticed at near term, suggesting the profound changes to compete with CRF-R1 and CRF-R2 for CRF and urocortin.33 Our
in nuclear transcription machineries begin to occur at near term. immunohistochemical investigation revealed expression of
We also noticed a robust change in fetal plasma cortisol levels at CRF-binding protein in the smooth muscle-enteric unit.34 We
term, suggesting that the expression of glucocorticoid receptors observed a marked decrease in smooth muscle expression of
could be turned ‘off’ by glucocorticoids at term, or immediately CRF-binding protein and a simultaneous increase in myenteric
after birth, as circulating levels of glucocorticoids reach basal levels neuronal somas at term, indicating that this protein is a critical
within 24 h after birth in many species including humans. regulator of CRF and urocortin functions at the level of smooth
muscle-enteric unit in sheep fetal distal colon.
Evidence for prereceptor metabolism of glucocorticoids
in fetal sheep distal colon CRF signaling system may play a role in preventing
Glucocorticoid actions at the cellular level are controlled by 11-beta in utero meconium passage before term
hydroxysteroid hydrogenase (11bHSD) type 1 and type 2, enzymes We performed in vitro study of sheep fetal colonic contractility to
that regulate the interconversion of active glucocorticoid and their assess the significance of CRF receptor system.29 In these studies,
inactive 11-keto metabolites. We recently examined the presence of colonic smooth muscle strips prepared from preterm sheep fetal
11 bHSD-1 and 11 bHSD-2 in sheep fetal colonic enteric nervous distal colonic segments were subjected to cholinergically dependent
system as evidence that these enzymes provide a novel means for contractility in an organ bath system following pre-incubation with
regulation of glucocorticoid-mediated ‘genomic’ activities.31 The CRF or urocortin. Both neuropeptides significantly decreased the
antibodies to 11bHSD-1 and 11 bHSD-2 elicited positive cholinergically stimulated contractility. Immunohistochemical
immunostaining of both myenteric and submucosal ganglia at all studies confirmed the expression of muscarinic receptor subtype
gestation ages. The percentage of myenteric ganglia expressing 3 and CRF-R2 in preterm sheep fetal colon. Additional studies
11bHSD-1 and 11bHSD-2 were maximal at term. The percentage of revealed positive immunostaining for CRF and urocortin in smooth
submucosal ganglia expressing 11bHSD-1 was maximal at term, muscle and enteric neurons, indicating the local availability of
whereas the expression of 11bHSD-2 remained constant throughout neuropeptides to inhibit smooth muscle motility during the time of
gestation. We speculate that increasing levels of 11bHSD-1, and thus stress.29 All in all, stress hormones (CRF and urocortin) and stress
increased active glucocorticoid levels, at term could contribute to hormone receptors (CRF-R1 and CRF-R2) appear to function in
glucocorticoid-dependent colonic contractile maturation. parallel or series pathways, and CRF-R2 plays a central role in
preventing stress-induced in utero meconium passage before term.

CRF system Cholinergic circuitry system


Identification and gestation-dependent changes in CRF Maturational changes in sheep fetal colonic cholinergic circuitry
system in sheep fetal distal colon system parallels plasma glucocorticoid surge
We recently undertook a systematic investigation to obtain We utilized antibodies specific to vesicular acetylcholine
anatomical support that CRF system is an integral component of transporter, a high-affinity choline transporter, and peripheral
distal colonic contractility apparatus.32 Our findings can be choline acetyl transferase as markers for cholinergic synaptic
summarized as follows: CRF-R1- and CRF-R2-specific antibodies circuitry system to examine the maturation of cholinergic system
strongly immunostained longitudinal and circular smooth muscles in distal colon of sheep fetuses from very preterm to term
in addition to muscularis mucosa in colonic segments at all gestation.35 The percentage of myenteric ganglia expressing
gestational ages. A marked decrease in CRF-R2 immunostaining cholinergic markers in term fetuses were significantly higher than
occurred in smooth muscle layers close to term. CRF-R1 antibody those observed in preterm. The marked increase observed in
immunostained neuronal somas both in myenteric and cholinergic markers in myenteric ganglia in fetal distal colon at
submucosal ganglia, and the percentage of ganglia expressing term is possibly mediated by effects of endogenous glucocorticoid
CRF-R1 were maximal at term gestation. On the basis of the levels, which peak at term. We speculate that glucocorticoid is the
reciprocal changes in CRF-R1 and CRF-R2 expression in the physiological hormone that regulates enteric cholinergic neural
smooth muscle-enteric unit, we speculate that CRF-R1 could be synaptic circuitry system similar to their actions in central nervous
hyper-responsive to stressors that stimulate the release of system regions.

Journal of Perinatology
Mechanism of in utero meconium passage
J Lakshmanan and MG Ross
S12

Localization of inhibitory and stimulatory muscarinic Disclosure


receptor subtypes in sheep fetal distal colon: implication Jayaraman Lakshmanan has received grant funding from NIH R03. Michael Ross
for meconium passage has received grant funding from the March of Dimes.
Acetylcholine mediates gastrointestinal motility through muscarinic
receptors. To date, five muscarinic receptors (M1–M5) have been
cloned, with M3 confirmed as the prominent mediator of smooth
muscle contraction. Of the other subtypes, M1, M2 and M4 have References
been shown to function as potent autoinhibitor (that is, inhibitory 1 Momoh JT. Hirschsprung’s disease: problems of diagnosis and treatment. Ann Trop
presynaptic muscarinic receptor) of Ach release from myenteric Paediatr 1982; 2: 31–35.
plexus. We examined the maturation and topographical 2 Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a
difference? [see comments]. Pediatrics 1990; 85: 715–721.
distribution of muscarinic receptor subtypes in distal colon from 3 Ahanya SN, Lakshmanan J, Morgan BL, Ross MG. Meconium passage in utero:
very preterm to term.36 M1–M5 antibodies elicited positive staining mechanisms, consequences, and management. Obstet Gynecol Surv 2005; 60:
in colonic sections at all gestational ages. M4 is expressed most 45–56.
abundantly in circular and longitudinal smooth muscle layers, 4 Usher RH, Boyd ME, McLean FH, Kramer MS. Assessment of fetal risk in postdate
whereas M3 is expressed in rather low levels. Positive pregnancies. Am J Obstet Gynecol 1988; 158: 259–264.
5 Miller FC, Read JA. Intrapartum assessment of the postdate fetus. Am J Obstet Gynecol
immunostaining for both M5 and M1 in smooth muscle layers are
1981; 141: 516–520.
similar, but their immunostaining intensity was significantly lower 6 Richey SD, Ramin SM, Bawdon RE, Roberts SW, Dax J, Roberts J et al. Markers of
compared with M3, M4 and M2. The percentage of neurons acute and chronic asphyxia in infants with meconium-stained amniotic fluid.
expressing inhibitory autoreceptors (M1, M2 and M4) in the enteric Am J Obstet Gynecol 1995; 172: 1212–1215.
ganglia was markedly higher in distal colonic sections of preterm 7 Tache Y, Bonaz B. Corticotropin-releasing factor receptors and stress-related alterations
and near-term fetuses, with expression declining rapidly and of gut motor function. J Clin Invest 2007; 117: 33–40.
8 Tache Y, Martinez V, Million M, Rivier J. Corticotropin-releasing factor and the brain-
markedly at term. Our results indicate that before term, muscarinic gut motor response to stress. Can J Gastroenterol 1999; 13(Suppl A): 18A–25A.
autoreceptors may inhibit presynaptic release of Ach and 9 Martinez V, Wang L, Rivier J, Grigoriadis D, Tache Y. Central CRF, urocortins and
consequently limit the cholinergically mediated smooth muscle stress increase colonic transit via CRF1 receptors while activation of CRF2 receptors
contractility. This could represent a physiological mechanism delays gastric transit in mice. J Physiol 2004; 556: 221–234.
analogous to CRF-R2 functions in preventing stress-induced 10 Million M, Wang L, Martinez V, Tache Y. Differential Fos expression in the
paraventricular nucleus of the hypothalamus, sacral parasympathetic nucleus and
in utero meconium passage before term. The rapid decline of
colonic motor response to water avoidance stress in Fischer and Lewis rats. Brain Res
muscarinic inhibitory autoreceptors noticed at term most 2000; 877: 345–353.
likely relieves the colonic contractile machinery from 11 Williams CL, Villar RG, Peterson JM, Burks TF. Stress-induced changes in intestinal
auto-inhibition and will increase efficacy of cholinergic synaptic transit in the rat: a model for irritable bowel syndrome. Gastroenterology 1988; 94:
transmission, facilitating ready responsiveness of colonic contractile 611–621.
machinery to Ach. 12 Miampamba M, Sharkey KA. c-Fos expression in the myenteric plexus, spinal cord and
brainstem following injection of formalin in the rat colonic wall. J Auton Nerv Syst
In summary, we have established for the first time a rat model 1999; 77: 140–151.
for stress-induced in utero meconium passage. We have further 13 la Fleur SE, Wick EC, Idumalla PS, Grady EF, Bhargava A. Role of peripheral
obtained anatomical support for the expression of key molecules of corticotropin-releasing factor and urocortin II in intestinal inflammation and motility
glucocorticoid system, CRF system and cholinergic circuitry system in terminal ileum. Proc Natl Acad Sci USA 2005; 102: 7647–7652.
in sheep fetal distal colon. All three systems seem positioned 14 Hillhouse EW, Grammatopoulos DK. The molecular mechanisms underlying the
regulation of the biological activity of corticotropin-releasing hormone receptors:
in distal colon readily to respond to stress. Our studies unexpectedly
implications for physiology and pathophysiology. Endocr Rev 2006; 27: 260–286.
revealed the presence of potential mechanisms to prevent the 15 Maillot C, Million M, Wei JY, Gauthier A, Tache Y. Peripheral corticotropin-releasing
in utero meconium passage before term, including the factor and stress-stimulated colonic motor activity involve type 1 receptor in rats.
expression of CRF-R2. Future studies should improve our Gastroenterology 2000; 119: 1569–1579.
understanding of the mechanisms of in utero meconium passage 16 Grundy D, Al-Chaer ED, Aziz Q, Collins SM, Ke M, Tache Y et al. Fundamentals of
and possibly to develop pharmacotherapy to prevent stress-induced neurogastroenterology: basic science. Gastroenterology 2006; 130: 1391–1411.
17 Jones SA, Brooks AN, Challis JR. Steroids modulate corticotropin-releasing hormone
in utero meconium passage and consequences of meconium production in human fetal membranes and placenta. J Clin Endocrinol Metab 1989;
aspiration. 68: 825–830.
18 Bruder ED, Jacobson L, Raff H. Plasma leptin and ghrelin in the neonatal rat:
interaction of dexamethasone and hypoxia. J Endocrinol 2005; 185: 477–484.
19 Takahashi LK, Goh CS. Glucocorticoid facilitation of cholinergic development in the rat
Acknowledgments hippocampus. Neurosci 1998; 83: 1145–1153.
20 Coukos G, Monzani A, Saletti C, Petraglia F. [Noradrenaline and interleukin-1
This research was supported by grants from the March of Dimes Foundation stimulate CRF secretion from human placental cells in culture]. Medicina (Firenze)
(MGR) and the National Institutes of Health (JL). 1988; 8: 441–442.

Journal of Perinatology
Mechanism of in utero meconium passage
J Lakshmanan and MG Ross
S13

21 Lakshmanan J, Ahanya SN, Rehan V, Oyachi N, Ross MG. Elevated plasma 29 Lakshmanan J, Oyachi N, Ahanya SA, Liu G, Mazdak M, Ross MG.
corticotrophin release factor levels and in utero meconium passage. Pediatr Res 2007; Corticotropin-releasing factor inhibition of sheep fetal colonic contractility: mechanisms
61: 176–179. to prevent meconium passage in utero. Am J Obstet Gynecol 2007; 196: 357.
22 Lakshmanan J, Salido E, Amidi F, Amidi E, Raj R, Ross MG. Rat placenta expresses 30 Lakshmanan J, Oyachi N, Liu GL, Choi GY, Ross MG. Fetal colonic enteric nervous
corticotrophin releasing factor protein and mRNA. Reproductive Sciences 2007; system is a site of glucocorticoid-induced gastrointestinal maturation. Reprod Sci 2007;
14(1 Suppl): 175A. 14(1 Suppl): 287A.
23 Frim DM, Emanuel RL, Robinson BG, Smas CM, Adler GK, Majzoub JA. 31 Lakshmanan J, Liu GL, Oyachi N, Ross MG. Evidence for pre-receptor metabolism of
Characterization and gestational regulation of corticotropin-releasing hormone glucocorticoids in ovinee fetal distal colonic enteric nervous system. Reprod Sci 2007;
messenger RNA in human placenta. J Clin Invest 1988; 82: 287–292. 14(1 Suppl): 288A.
24 Lakshmanan J, Richard JD, Liu GL, Ross MG. Corticotrophin releasing factor is a fetal 32 Lakshmanan J, Liu GL, Oyachi N, Ross MG. Localization and gestation-dependent
gut hormone. Reprod Sci 2007; 14(1 Suppl): 251A. pattern of CRF-receptos (R1, R2) expression in ovine fetal distal colon. Reprod Sci
25 Kawahito Y, Sano H, Kawata M, Yuri K, Mukai S, Yamamura Y et al. Local secretion of 2007; 14(1 Suppl): 258A.
corticotropin-releasing hormone by enterochromaffin cells in human colon. 33 Seasholtz AF, Valverde RA, Denver RJ. Corticotropin-releasing hormone-binding
Gastroenterology 1994; 106: 859–865. protein: biochemistry and function from fishes to mammals. J Endocrinol 2002; 175:
26 Richard JD, Lakshmanan J, John TA, Ross MG. Rat fetal gastrointestinal tract is a target 89–97.
organ for corticotrophin-releasing factor family neuropeptides. Am J Obstet Gyn 2006; 34 Lakshmanan J, Liu GL, Oyachi N, Ross MG. Cellular localization of corticotrophin
195(6 Suppl): S214. releasing factor binding protein (CRF-BP) in fetal ovine distal colon: a possible local
27 Ross B, Bradley K, Nijland MJ, Polk DH, Ross MG. Increased fetal colonic muscle inhibitor of stress-induced colonic motility. Reprod Sci 2007; 14(1 Suppl): 288A.
contractility following glucocorticoid and thyroxine therapy: implications for 35 Lakshmanan J, Lips KS, Liu GL, Ross MG. Maturational changes in ovine fetal colonic
meconium passage. J Matern Fetal Med 1997; 6: 129–133. cholinergic circuitry parallels plasma glucocorticoid surge. Reprod Sci 2007;
28 Acosta R, Oyachi N, Lee JJ, Lakshmanan J, Atkinson JB, Ross MG. 14(1 Suppl): 249A.
Mechanisms of meconium passage: cholinergic stimulation of 36 Lakshmanan J, Liu GL, Ross MG. Localization of inhibitory and stimulatory muscarinic
electromechanical coordination in the fetal colon. J Soc Gynecol Investig receptor subtypes in ovine fetal distal colon: implications for meconium passage.
2005; 12: 169–173. Reprod Sci 2007; 14(1 Suppl): 168A.

Journal of Perinatology
Journal of Perinatology (2008) 28, S14–S18
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Obstetric approaches to the prevention of meconium aspiration
syndrome
H Xu, S Wei and WD Fraser
Department of Obstetrics and Gynecology, Hoˆpital Sainte-Justine, Universite´ de Montre´al, Montreal, QC, Canada

Definition of meconium aspiration syndrome


Meconium aspiration syndrome (MAS) is associated with increased
Rossi et al.20 has defined MAS as respiratory distress in the first 4 h
risk for perinatal mortality and morbidities. To provide an overview of the
after birth with oxygen requirement and chest roentgenogram
advances in our knowledge concerning the obstetric approaches to the
showing characteristic features of MAS. Recently, Cleary and
prevention of MAS. The evidence of the effectiveness of intrapartum
Wiswell have defined MAS as respiratory distress in an infant born
surveillance, amnioinfusion, and delivery room management in the
through MSAF, which cannot be otherwise explained. Mild MAS has
prevention of MAS are reviewed in the present paper. Meconium aspiration
been defined as requiring <40% oxygen for <48 h, moderate MAS
syndrome remains one of the most common but challenging conditions
as requiring >40% concentration of oxygen therapy for at least
for obstetricians and pediatricians. The available evidence did not
48 h, and severe MAS if requiring assisted mechanical ventilation,
demonstrate a beneficial effect of either of obstetric strategies in the
that is, often associated with persistent pulmonary hypertension.1,20
prevention of MAS.
They further pointed out that the severity of MAS does not
Journal of Perinatology (2008) 28, S14–S18; doi:10.1038/jp.2008.145
necessarily correspond to the degree of chest radiographic
abnormality. MAS is associated with a range of radiographic
features including coarse, patchy infiltrates, consolidation,
atelectasis, pleural effusions, air leaks, hyperinflation, a wet-lung
picture and hypovascularity.1 In some cases, the chest film may be
Introduction
interpreted as normal.1
Meconium staining of the amniotic fluid (MSAF) occurs in <5% of
preterm, 7 to 22% of term deliveries, increasing to between 23 and
52% of births at >42 weeks.1–7 The mechanisms influencing
meconium passage are complex, involving hormonal and Pathophysiology and risk factors
neuroregulatory functions, chronic hypoxia or reflecting The pathophysiology of MAS is complex and remains controversial.
maturation of the fetal gastrointestinal system.8–11 MSAF is Many factors, such as airway obstruction, alveolar or parenchymal
associated with an increased risk of neonatal morbidity and inflammation, impaired surfactant production and function and
mortality. Meconium aspiration syndrome (MAS) is the most direct toxicity of meconium constituents could be involved in the
serious neonatal pathology associated with MSAF. It has been pathophysiology of the MAS.1,21,22 It has been suggested that the
reported to occur in 1.7 to 35.8% of cases complicated with MSAF, extent of lung destruction is not closely correlated to the quantity of
and 1 to 3% of liveborn infants.1,12–14 The case fatality rate of MAS meconium in lung tissue but rather to the degree of hypoxia and
has been reported to be high, ranging from 5 to 40%.1,13–17 acidosis present at delivery.23 Ghidini and Spong22 postulated that
Approximately one-third of babies with MAS require intubation and the pathologic events leading to mild, moderate or severe cases of
mechanical ventilation, and other new therapies such as high MAS may be different. Severe MAS may not be in fact causally
frequency ventilation, inhaled nitric oxide and surfactant related to the aspiration of meconium but rather may be caused by
administration, although the effectiveness of certain of these other pathologic processes occurring in utero, such as chronic
technologies remain controversial.2,17–19 Serious complications asphyxia, infection or persistent pulmonary hypertension.22 The
resulting from MAS include pneumothorax, convulsion hypothesis is in fact supported by the lack of evidence that the
and death. severity of MAS directly correlates with the amount of meconium
aspirated, the consistency of meconium and the duration of
Correspondence: Dr WD Fraser, Department of Obstetrics and Gynecology, Hôpital Sainte-
exposure to meconium.1,15,20,22,23 It is still unclear whether
Justine, Université de Montréal, 3175 Chemin de la Côte Sainte-Catherine, Montreal, QC,
Canada H3T 1C5. obstruction of airways because of aspiration of meconium has a
E-mail: william.fraser@umontreal.ca pivotal role in the progress of MAS. MAS can occur before delivery,
Obstetric management of MAS
H Xu et al
S15

even in the absence of labor, being reported in infants delivered by AI significantly reduces the risk of FHR decelerations and cesarean
elective cesarean section.24 section.45–52
Although the presence of meconium during labor is known to AI has been also proposed as a method to reduce MAS. Potential
be associated with an increased risk of perinatal morbidity and mechanisms through which AI could act include mechanical
mortality, most babies have favorable outcomes. Early recognition cushioning of the umbilical cord, which could correct or prevent
of infants at the highest risk for the development of MAS could be recurrent umbilical compressions that lead to fetal acidemia, a
essential for optimizing the clinical preventive strategies. A vast condition predisposing to MAS; and dilution of meconium that
array of risk factors for the occurrence of MAS have been identified could reduce its mechanical and inflammatory effects in the
either using unselected obstetrical populations or infants born pathogenesis of MAS.
through MSAF. Those factors are heavy MSAF, nulliparity, postterm To date, more than 15 randomized or quasi-randomized trials
delivery, fetal heart rate (FHR) abnormalities during labor, of AI for MSAF have been reported, with conflicting results.53,54 The
presence of meconium below the vocal cords, cesarean delivery and methodological quality varied across studies. The largest trial (over
the low Apgar scores.13,14,20,25–30 It has been reported that there is 1998 participants) was an international trial performed in 56
an apparent relationship between maternal ethnicity and risk of centers where EFM and neonatal intubation and suctioning for
MSAF, and the risk of MAS having been observed to be increased in babies with respiratory difficulty were routinely available.55 Analysis
black Americans, Africans and Pacific Islanders.30–32 was by intention to treat. The primary outcome was a composite
indicator that included the occurrence of perinatal death and/or
moderate or severe MAS. The results indicated that AI showed no
Intrapartum fetal monitoring effect on the primary outcome (relative risk; RR 1.26, 95%
The goal of continuous electronic fetal heart rate monitoring confidence interval; CI 0.82 to 1.95). Furthermore, the frequencies
(EFM) is to detect fetal hypoxemia and therefore reduce the risk of of oropharyngeal suctioning, laryngoscopy or intubation in the
adverse neonatal outcomes. However, the effectiveness of this delivery room were similar between groups, as were the proportion
approach to care has been questioned. Randomized trials of EFM, of babies with meconium visualized below the vocal cords. There
with or without fetal blood gas and acid–base assessment, which were no differences between groups in the occurrence of the
were conducted in the unselected obstetrical population, have combined outcome of perinatal mortality and/or serious morbidity
found no evidence that this approach to care reduces the risk of (RR 1.13, 95% CI 0.88 to 1.47). In addition, an analysis that
fetal or neonatal mortality or morbidity.33–39 stratified for the presence or absence of variable FHR decelerations
Intrapartum monitoring has been recommended to screen for before randomization found no effect on the primary outcome
early signs of fetal hypoxia, a risk factor for MAS. Several authors either, although the study was underpowered to detect such effects
have noted an increase in the frequency of FHR abnormalities in within strata.
association with MSAF.15,20,40,41 It has been reported that, in the We recently conducted a systematic review, integrating the
presence of MSAF, fetal tachycardia, variable and late decelerations results of the largest trial.54 Studies were included if they were
and decreased long-term variability are risk factors for MAS. randomized controlled trials that evaluated the effect of
Certain authors investigated the relationships among abnormal prophylactic AI during labor with MSAF; treatment was randomly
cardiotocograms in labor, MSAF and adverse neonatal outcomes allocated (AI versus controls). All included studies were further
such as low arterial cord blood pH, and low Apgar scores. The subjected to a score-based quality assessment for randomized
authors did not find that the presence of abnormal FHR patterns studies that was adapted from the Jadad score. The main analysis
increased the overall correlation between MSAF and adverse was based on the studies that were considered to be of high quality.
outcome.7,42 In contrast, Umstad et al.43 investigated the predictive The meta-analysis included a total of 4030 women, 1999 allocated
value of abnormal FHR patterns in early labor and found that the to AI and 2031 allocated to control. Of these, 3178 women were
presence of meconium in the amniotic fluid improved the recruited in clinical settings with standard peripartum surveillance
predictive properties of the test. and 852 women were randomized in centers with limited
peripartum surveillance, defined as the nonavailability of EFM
during labor. The methodological quality varied across studies.
Amnioinfusion Heterogeneity was noted across studies with respect to the AI
Amnioinfusion (AI), or transcervical infusion of saline into the protocols and the end points evaluated. In the setting of standard
amniotic cavity, was used first to relieve persistent variable FHR peripartum surveillance, the results failed to demonstrate a
decelerations during labor or to prevent the occurrence of reduction in the risk of MAS (RR 0.59, 95% CI 0.28 to 1.25),
decelerations in presence of oligohydramnios.44 Results of Apgar-5 <7 (RR 0.90, 95% CI 0.58 to 1.41) or caesarean delivery
randomized controlled trials, including a meta-analysis indicate (RR 0.89, 95% CI 0.73 to 1.10). However, in clinical settings with
that, in the presence of oligohydramnios, prophylactic intrapartum limited peripartum surveillance, AI appeared to reduce the risk of

Journal of Perinatology
Obstetric management of MAS
H Xu et al
S16

MAS (RR 0.25, 95% CI 0.13 to 0.47). Several findings from this severity of MAS and the risk of respiratory distress.61–63 They
meta-analysis are worthy of comment. Firstly, the observed suggested that combined approach of intrapartum oropharyngeal
heterogeneity in the stratum of studies conducted in centers with suctioning and endotracheal suctioning was effective in the
standard surveillance is largely attributable to our recent large reduction of MAS.
trial. The source of this heterogeneity remains largely unexplained. Vain et al. conducted a multicenter international trial to assess
The most significant finding of the meta-analysis is the apparent the effectiveness of oropharyngeal and nasopharyngeal suctioning
discordance in the observed effect of AI on MAS between centers before delivery of the shoulders for the prevention of MAS. They
with standard and limited peripartum surveillance. Continuous found that the incidence of MAS, need for mechanical ventilation,
EFM is a key component in the prevention of asphyxia in patients and neonatal mortality was similar between groups (suction versus
with MSAF. Therefore, the application of this technology may no suction). In addition, they found no evidence of a benefit of
reduce the contribution of severe asphyxia to the risk of occurrence intrapartum suctioning on the occurrence of MAS, MAS requiring
of MAS. It would appear that in settings where this technology is mechanical ventilation, or mortality.64
routinely used, AI confers no additional benefit over EFM in terms Regarding the postdelivery management such as routine
of prevention of MAS. However, in settings where continuous EFM endotracheal suctioning and intubation, reports from observational
is not routinely available, AI may be beneficial for the reduction of studies suggested that intratracheal suctioning could prevent the
MAS. Further studies in such settings are warranted to confirm this occurrence of MAS for meconium-stained neonates and significantly
hypothesis. decreased the mortality subsequent to that disorder.65–67 Intubation
AI may not be without risk. The use of AI has been reported to is not without risk and has been associated with hypoxia,
be associated with adverse events. Complications including uterine bradycardia and laryngeal stridor.67–70 Should endotracheal
overdistension and hypertonia, uterine rupture in association with suctioning and intubation be applied universally in infants born
previous uterine scar, FHR abnormality, umbilical cord prolapse through MSAF or selectively reserved for those who are depressed
and chorioamnionitis have been reported.56–59 Four cases of after birth is another topic of controversy. Some investigators
maternal deaths have been reported associated with the AI.58,59 suggested that a selective approach may be useful and
Several authors have reported the occurrence of excessive uterine justified,6,8,71–73 whereas other suggested that universal intubation
contractions or unusually rapid labor progress related to AI. In the and suctioning was the best strategy to prevent potential morbidity
AI group of Fraser et al.’s55 trial, 10 women (1.1%) experienced and mortality related to meconium staining.67
bleeding, and in 63 (6.9%) women, hypertonicity, hydramnios or Linder et al.68 suggested that nondepressed meconium-stained
uterine overdistension was diagnosed during the procedure whereas infants did not benefit from immediate intratracheal suctioning
the incidence of other maternal complications were comparable and such intervention could be harmful. Liu and Harrington
between AI and control groups. conducted a randomized trial to assess if intubation of the low-risk
American College of Obstetricians and Gynecologists has newborn with thin meconium affects the incidence of respiratory
recently published a Committee Opinion that concludes that symptoms. They were unable to demonstrate the beneficial effect of
routine prophylactic AI for the dilution of MSAF should be carried intubation and intratracheal suctioning in the infant with thin
out only in the setting of additional clinical trials. However, they meconium and an otherwise low-risk pregnancy.74 To address this
state that AI remains a reasonable approach to the treatment of question, Wiswell et al.69 conducted a multicenter randomized
repetitive variable decelerations, regardless of amniotic fluid trial, involving a total of 2094 neonates, to investigate
meconium status.60 whether intubation and suctioning of apparently vigorous,
meconium-stained neonates would reduce the risk of MAS. There
were no significant differences between intubation and expectant
Delivery room management management groups in the rates of MAS or other respiratory
Routine oropharyngeal suctioning before delivery of the infants’ disorders. Moreover, intratracheal suctioning showed no benefit
shoulders has long been involved in preventing MAS. The findings over expectant management even for infants born through the
of observational studies remain conflicting.20,26,61 –63 Falciglia thickest consistency MSAF. They further identified the independent
et al.26 compared infants with meconium-stained fluid who risk factors for the development of MAS using stepwise logistic
underwent ‘early’ oronasopharyngeal DeLee suctioning with a regression. The results indicated that the use of oropharyngeal
similar group of infants whose airways were suctioned ‘late’ (after suctioning lead to a decreased risk of MAS (8.5% in infants who did
chest delivery). They found no evidence of benefit of oropharyngeal not have intrapartum suction versus 2.7% in infants with
suctioning in the prevention of MAS. Rossi et al.20 also reported the intrapartum suctioning). The authors concluded that endotracheal
similar rates of meconium visualized in the vocal cords despite intubation and suctioning still be performed in infants born
early oropharyngeal suctioning. In contrast, several authors through MSAF, if they are not vigorous, if they need positive
reported that intrapartum pharyngeal suctioning reduced the pressure ventilation or they develop symptoms of respiratory

Journal of Perinatology
Obstetric management of MAS
H Xu et al
S17

distress. Furthermore, a recently published meta-analysis of four 13 Davis RO, Phillips III JB, Harris Jr BA, Wilson ER, Huddleston JF. Fatal meconium
randomized trials demonstrated no significant benefit of routine aspiration syndrome occurring despite airway management considered appropriate.
Am J Obstet Gynecol 1985; 151: 731–736.
endotracheal intubation and suctioning at birth over routine
14 Urbaniak KJ, McCowan LM, Townend KM. Risk factors for meconium aspiration
resuscitation including oropharyngeal suction of vigorous, syndrome. Aust N Z J Obstet Gynaecol 1996; 36: 401–406.
meconium-stained infants born at term.75 The authors 15 Hernandez C, Little BB, Dax JS, Gilstrap III LC, Rosenfeld CR. Prediction of the severity
recommended that intubation and suctioning be restricted to of meconium aspiration syndrome. Am J Obstet Gynecol 1993; 169: 61–70.
depressed newborns, that is, those with a heart rate of <100 beats 16 Falciglia HS. Failure to prevent meconium aspiration syndrome. Obstet Gynecol 1988;
per min, poor respiratory effort and poor tone. 71: 349–353.
17 Coltart TM, Byrne DL, Bates SA. Meconium aspiration syndrome: a 6-year retrospective
study. Br J Obstet Gynaecol 1989; 96: 411–414.
Conclusion 18 Bhutani VK, Chima R, Sivieri EM. Innovative neonatal ventilation and meconium
aspiration syndrome. Indian J Pediatr 2003; 70: 421–427.
MAS remains a challenging condition for obstetricians and 19 Wiswell TE. Advances in the treatment of the meconium aspiration syndrome. Acta
neonatologists. Despite the decreased risk of MAS and related Paediatr Suppl 2001; 90: 28–30.
mortality and morbidity, the available evidence did not 20 Rossi EM, Philipson EH, Williams TG, Kalhan SC. Meconium aspiration
demonstrate a beneficial effect of either of obstetric strategies in the syndrome: intrapartum and neonatal attributes. Am J Obstet Gynecol 1989; 161:
1106–1110.
prevention of MAS. The suggested apparent disparity in the effect of
21 Katz VL, Bowes WA. Meconium aspiration syndrome: reflections on a murky subject.
AI on MAS between centers with standard and limited peripartum Am J Obstet Gynecol 1992; 166: 171–183.
surveillance is worthy of attentions for clinicians. Additional 22 Ghidini A, Spong CY. Severe meconium aspiration syndrome is not caused by
well-designed randomized controlled trials in settings of limited aspiration of meconium. Am J Obsted Gynecol 2001; 185: 931–938.
peripartum surveillance are required to elucidate the optimal 23 Jovanovic R, Nguyen HT. Experimental meconium aspiration in guinea pigs. Obstet
management of MAS in this context. Gynecol 1989; 73: 652–656.
24 Greenough A. Meconium aspiration syndromeFprevention and treatment. Early
Hum Dev 1995; 41: 183–192.
Disclosure 25 Meis PJ, Hall III M, Marshall JR, Hobel CJ. Meconium passage: a new classification for
risk assessment during labor. Am J Obstet Gynecol 1987; 131: 509–513.
WD Fraser has received grant support from Utah Medical Corporation. The 26 Falciglia HS, Henderschott C, Potter P, Helmchen R. Does DeLee suction at the
remaining authors have declared no financial interests. perineum prevent meconium aspiration syndrome? Am J Obstet Gynecol 1992; 167:
1243–1249.
27 Alexander GR, Hulsey TC, Robillard PY, De Caunes F, Papiernik E. Determinants of
References meconium stained amniotic fluid in term pregnancies. J Perinatol 1994; 14: 259–263.
1 Cleary GM, Wiswell TE. Meconium-stained amniotic fluid and the meconium 28 Meydanli MM, Dilbaz B, Caliskan E, Dilbaz S, Haberal A. Risk factors for meconium
aspiration syndrome: an update. Pediatr Clin North Am 1998; 45: 511–529. aspiration syndrome in infants born through thick meconium. Int J Gynaecol Obstet
2 Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a 2001; 72: 9–15.
difference? Pediatrics 1990; 85: 715–721. 29 Paz Y, Solt I, Zimmer EZ. Variables associated with meconium aspiration syndrome in
3 Scott H, Walker M, Gruslin A. Significance of meconium-stained amniotic fluid in the labours with thick meconium. Eur J Obstet Gynecol Reprod Biol 2001; 94:
preterm population. J Perinatol 2001; 21: 174–177. 27–30.
4 Usher RH, Boyd ME, McLean FH, Kramer MS. Assessment of fetal risk in postdate 30 Dargaville PA, Copnell B, Australian and New Zealand Neonatal Network. The
pregnancies. Am J Obstet Gynecol 1988; 158: 259–264. epidemiology of meconium aspiration syndrome: incidence, risk factors, therapies, and
5 Ostrea EM, Naqvi M. The influence of gestational age on the ability of the fetus to pass outcome. Pediatrics 2006; 117: 1712–1721.
meconium in utero. Acta Obstet Gynecol Scand 1982; 61: 275–277. 31 Sriram S, Wall SN, Khoshnood B, Singh JK, Hsieh HL, Lee KS. Racial disparity in
6 Eden RD, Seifert LS, Winegar A, Spellacy W. Perinatal characteristics of uncomplicated meconium stained amniotic fluid and meconium aspiration syndrome in the United
postdate pregnancies. Obstet Gynecol 1987; 69: 296–299. States, 1989 to 2000. Obstet Gynecol 2003; 102: 1262–1268.
7 Steer PJ, Eigbe F, Lissauer TJ, Beard RW. Interrelationships among abnormal 32 Sedaghatian MR, Othman L, Hossain MM, Vidyasagar D. Risk of meconium stained
cardiotocograms in labor, meconium staining of the amniotic fluid, arterial cord blood amniotic fluid in different ethnic groups. J Perinatol 2000; 20: 257–261.
pH, and Apgar scores. Obstet Gynecol 1989; 74: 715–721. 33 Shy KK, Larson EB, Luthy DA. Evaluating a new technology: the effectiveness of
8 Lucas A, Adrian TE, Christofides N, Bloom SR, Aynsley-Green A. Plasma motilin, electronic fetal heart rate monitoring. Annu Rev Public Health 1987; 8: 165–190.
gastrin and enteroglucagon and feeding in the human newborn. Arch Dis Child 1980; 34 Lumley J. Does continuous intrapartum fetal monitoring predict long-term
55: 673–677. neurological disorders? Paediatr Perinat Epidemiol 1988; 2: 299–307.
9 Miller FC, Sacks DA, Yeh SY, Paul RH, Schifrin BS, Martin Jr CB et al. Significance of 35 Paneth N, Bommarito M, Stricker J. Electronic fetal monitoring and later outcome.
meconium during labor. Am J Obstet Gynecol 1975; 122: 573–580. Clin Invest Med 1993; 16: 159–165.
10 Bochner CJ, Medearis AL, Ross MG, Oakes GK, Jones P, Hobel CJ et al. The role of 36 Neilson JP. Electronic fetal heart rate monitoring during labor: information from
antepartum testing in the management of postterm pregnancies with heavy meconium randomized trials. Birth 1994; 21: 101–104.
in early labor. Obstet Gynecol 1987; 69: 903–907. 37 Graham EM, Petersen SM, Christo DK, Fox HE. Intrapartum electronic fetal heart rate
11 Ahanya SN, Lakshmanan J, Morgan BL, Ross MG. Meconium passage in utero: monitoring and the prevention of perinatal brain injury. Obstet Gynecol 2006; 108:
mechanisms, consequences, and management. Obstet Gynecol Surv 2005; 60: 45–56. 656–666.
12 Brown BL, Gleicher N. Intrauterine meconium aspiration. Obstet Gynecol 1981; 57: 38 Thacker SB, Stroup DF, Peterson HB. Efficacy and safety of intrapartum electronic fetal
26–29. monitoring: an update. Obstet Gynecol 1995; 86: 613–620.

Journal of Perinatology
Obstetric management of MAS
H Xu et al
S18

39 Alfirevic Z, Devane D, Gyte GM. Continuous cardiotocography (CTG) as a form of 57 Dragich DA, Ross AF, Chestnut DH, Wenstrom K. Respiratory failure associated with
electronic fetal monitoring (EFM) for fetal assessment during labour. Cochrane amnioinfusion during labor. Anesth Analg 1991; 72: 549–551.
Database Syst Rev 2006; 3: CD006066. 58 Maher JE, Wenstrom KD, Hauth JC, Meis PJ. Amniotic fluid embolism after saline
40 Starks GC. Correlation of meconium-stained amniotic fluid, early intrapartum pH, amnioinfusion: two cases and review of the literature. Obstet Gynecol 1994; 83:
and Apgar scores as predictors of perinatal outcome. Obstet Gynecol 1980; 56: 851–854.
604–609. 59 Dorairajan G, Soundararaghavan S. Maternal death after intrapartum
41 Hageman JR. Meconium staining of the amniotic fluid: the need for reassessment of saline amnioinfusion-report of two cases. Br J Obstet Gynaecol 2005; 112:
management by obstetricians and pediatricians. Curr Probl Pediatr 1993; 23: 1331–1333.
396–401. 60 ACOG Committee Obstetric Practice. ACOG Committee Opinion Number 346, October
42 Krebs HB, Petres RE, Dunn LT, Jordaan HVF, Segreti A. Intrapartum Fetal Heart Rate 2006: amnioninfusion does not prevent meconium aspiration syndrome. Obstet
Monitoring. III. Association of meconium with abnormal fetal heart rate patterns. Am J Gynecol 2006; 108: 1053.
Obstet Gynecol 1980; 137: 936–943. 61 Carson BS, Losey RW, Bowes Jr WA, Simmons MA. Combined obstetric and pediatric
43 Umstad MP. The predictive value of abnormal fetal heart rate patterns in early labour. approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126:
Aust NZJ Obstet Gynaecol 1993; 33: 145–149. 712–715.
44 Miyazaki FS, Nevarez F. Saline amnioinfusion for relief of repetitive variable 62 Yoder BA, Kirsch EA, Barth WH, Gordon MC. Changing obstetric practices associated
decelerations. A prospective randomized study. Am J Obstet Gynecol 1985; 153: with decreasing incidence of meconium aspiration syndrome. Obstet Gynecol 2002;
301–306. 99: 731–739.
45 Wang CC, Rogers MS. Lipid peroxidation in cord blood: a randomised sequential airs 63 Chishty AL, Alvi Y, Iftikhar M, Bhutta TI. Meconium aspiration in neonates: combined
study of prophylactic saline amnioinfusion for intrapartum oligohydramnios. Br J obstetric and paediatric intervention improves outcome. J Pak Med Assoc 1996; 46:
Obstet Gynaecol 1997; 104: 1145–1151. 104–108.
46 Nageotte MP, Freeman RK, Garite TJ, Dorchester W. Prophylactic intrapartum 64 Vain NE, Szyld EG, Prudent LM, Wiswell TE, Aguilar AM, Vivas NI. Oropharyngeal
amnioinfusion in patients with premature rupture of membranes. Am J Obstet Gynecol and nasopharyngeal suctioning of meconium-stained neonates before delivery
1985; 153: 557–562. of their shoulders: multicentre, randomised controlled trial. Lancet 2004; 364:
47 Nageotte MP, Bertucci L, Towers CV, Lagrew DL, Modaniou H. Prophylactic 597–602.
amnioinfusion in pregnancies complicated by oligohydramnios: a prospective study. 65 Gregory GA, Gooding CA, Phibbs RH, Tooley WH. Meconium aspiration in
Obstet Gynecol 1991; 77: 677–680. infantsFa prospective study. J Pediatr 1974; 85: 848–852.
48 Owen J, Henson BV, Hauth JC. A prospective randomized study of saline solution 66 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obstet Gynecol
amnioinfusion. Am J Obstet Gynecol 1990; 162: 1146–1149. 1975; 122: 767–771.
49 MacGregor SM, Banzhaf WC, Silver RK, Depp R. A prospective randomized evaluation 67 Wiswell T, Henley MA. Intratracheal suctioning, systematic infection, and the
of intrapartum amnioinfusion. Fetal acid–base status and cesarean delivery. J Reprod meconium aspiration syndrome. Pediatrics 1992; 89: 203–206.
Med 1991; 36: 69–73. 68 Linder N, Aranda JV, Tsur M, Matoth I, Yatsiv I, Mandelberg H et al. Need for
50 Schrimmer DB, Macri CJ, Paul RH. Prophylactic amnioinfusion as a treatment for endotracheal intubation and suction in meconium-stained neonates. J Pediatr 1988;
oligohydramnios in laboring patients: a prospective, randomized trial. Am J Obstet 112: 613–615.
Gynecol 1991; 165: 972–975. 69 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery room
51 Chauhan SP, Rutherford SE, Hess LW, Morrison JC. Prophylactic intrapartum management of the apparently vigorous meconium-stained neonate: results of the
amnioinfusion for patients with oligohydramnios. J Reprod Med 1992; 37: multicenter, international collaborative trial. Pediatrics 2000; 105: 1–7.
817–820. 70 Kresh MJ, Brion LP, Fleishman AR. Delivery room management of meconium stained
52 Pitt C, Sanchez-Ramos L, Kaunitz AM, Gaudier F. Prophylactic amnioinfusion for neonates. J Perinatol 1991; 11: 46–48.
intrapartum oligohydramnios: a meta-analysis of randomized controlled trials. Obstet 71 Bent RC, Wiswell TE, Chang A. Removing meconium from infant tracheae. Am J Dis
Gynecol 2000; 96: 861–866. Child 1992; 146: 1085–1089.
53 Hofmeyr GJ. Amnioinfusion for meconium-stained liquor in labor. Cochrane Database 72 Peng TCC, Gutcher GR, Van Dorsten JP. A selective aggressive approach to the neonate
Syst Rev 2002; 1: CD000014. exposed to meconium-stained amniotic fluid. Am J Obstet Gynecol 1996; 175:
54 Xu H, Hofmeyr J, Roy C, Fraser W. Intrapartum amnioinfusion for meconium stained 296–303.
amniotic fluid: a systematic review of randomised controlled trials. BJOG 2007; 114: 73 Yoder BA. Meconium-stained amniotic fluid and respiratory complications: impact of
383–390. selective tracheal suction. Obstet Gynecol 1994; 83: 77–84.
55 Fraser WD, Hofmeyr GJ, Lede R, Faron G, Alexander S, Goffinet F et al. Amnioinfusion 74 Liu WF, Harrington T. The need for delivery room intubation of thin meconium in the
for the prevention of the meconium aspiration syndrome. N Engl J Med 2005; 353: low-risk newborn: a clinical trial. Am J Perinatol 1998; 15: 675–682.
909–917. 75 Halliday HL, Sweet D. Endotracheal intubation at birth for preventing morbidity and
56 Wenstrom K, Andrews WW, Maher JE. Amnioinfusion survey: prevalence, protocols, and mortality in vigorous, meconium-stained infants born at term. Cochrane Database
complications. Obstet Gynecol 1995; 86: 572–576. Syst Rev 2001; 1: CD000500.

Journal of Perinatology
Journal of Perinatology (2008) 28, S19–S26
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Delivery room management of the meconium-stained newborn
TE Wiswell
Center for Neonatal Care, Orlando, FL, USA

the baby upward inverting them by 1801 and then swinging them
Review of all medical literature dealing with delivery room management of back down to their original position. This maneuver would be
meconium-stained infants. Additionally, the author contacted multiple repeated as many as 10 to 12 times. This untested therapy was
individuals involved historically or clinically with the published studies or widely practiced through the 1920s.
the persons who developed treatment guidelines. Although many therapies
have been suggested as being effective, none have been definitively proven
efficacious by the gold standard: a large, randomized, controlled trial Intratracheal suctioning
(RCT). Further adequate investigations (RCTs) need to be performed to
This author has not found any written descriptions concerning
assess whether proposed management schemes are of benefit in the care of
intubation and suctioning of meconium-stained infants before the
meconium-stained newborn infants.
year 1960. In a textbook of neonatal resuscitation published that
Journal of Perinatology (2008) 28, S19–S26; doi:10.1038/jp.2008.143
year,4 Dr L Stanley James (Figure 1) stated that if meconium had
been aspirated into the trachea, it should be suctioned out. He then
suggested using an endotracheal tube as a suction device. Dr Jack
Introduction Sinclair (personal communication) states that when he arrived as
Meconium-stained amniotic fluid (MSAF) is noted in 10 to 15% of a resident in Pediatrics at Columbia Hospital for Children in New
all deliveries (B500 000 annually in the United States).1,2 York City in 1961, both Dr James and Dr Virginia Apgar were
Approximately 3 to 4% of meconium-stained infants will develop teaching and advocating intubation and suctioning of
the meconium aspiration syndrome (MAS). Neonates that are meconium-stained babies. The latter individuals even made a
depressed and are born through thick-consistency MSAF are at the teaching movie of this procedure. Dr Lillian Blackmon (personal
highest risk for developing MAS. A considerable proportion of communication), while training at Columbia Hospital for
infants with MAS require mechanical ventilation, whereas many Children, did an elective with Dr William Tooley at the University
develop pulmonary air leaks. In addition, MAS is frequently of California at San Francisco (UCSF) in 1966. At that time,
associated with persistent pulmonary hypertension of the newborn. physicians at UCSF were not suctioning the airways of meconium-
The mortality rate among infants with the disorder may be as high stained neonates. Dr Blackmon subsequently returned to UCSF in
as 5 to 10%. Humans are not the only species affected by MAS; it is 1968. At virtually the same time, Dr William Silverman (Figure 2),
seen frequently and is a leading cause of death in domestic farm the long-time director of Neonatology at Columbia Hospital for
animals (pigs, cattle, and so on). Children, also moved west and became chief of neonatology of San
In an effort to prevent or mitigate the course of MAS, many Francisco Children’s Hospital. Dr Blackmon states that the two of
methods have been suggested to remove aspirated meconium from them began training residents and clinicians in San Francisco to
the airways. Historically, farmers would grasp meconium-stained intubate and suction meconium-stained neonates.
animals by their hindquarters and then swing them around their Dr George Gregory (Figure 3) and colleagues5 are given credit
heads. Centrifugal forces would move the meconium-stained fluid for the wide dissemination of the intubation and suctioning
toward the head of the rotating animal and the material would technique in their seminal 1974 publication. These individuals
then be manually extracted. In 1871, Dr Bernard Schultze described their prospective management of meconium-stained
described a resuscitation maneuver, one of the benefits of which infants at UCSF over a 6-month period. Eighty such infants were
purportedly included the removal of meconium from the airways3. intubated and had their tracheas suctioned. In addition, the infants
Caregivers would grasp affected infants by the shoulders vertically subsequently were treated with aerosolized distilled water, chest
at the level of the adult’s knees. The caregiver would then sweep physiotherapy (CPT) and postural drainage (positional therapy
alternating between head up and head down, as well as side to
Correspondence: Dr TE Wiswell, Center for Neonatal Care, 2718 North Orange Avenue,
Suite B, Orlando, FL 32804, USA. side). Sixteen (20%) of the infants were deemed to be ‘sick’, six
E-mail: Thomas_Wiswell@yahoo.com (38%) of whom had thin-consistency meconium suctioned from
DR management of meconium-stained newborns
TE Wiswell
S20

their airways. Of the 80 infants, 2 required mechanical ventilation, through thick, particulate meconium (despite almost 40% of their
but none were treated with continous positive airway pressure. ‘sick’ population having only thin-consistency meconium
Seven neonates developed pneumothorax and/or retrieved). They also recommended subsequent CPT and postural
pneumomediastinum. The authors stated that none of the 35 drainage for those infants from whom meconium-stained material
meconium-stained babies managed in this manner subsequent to is retrieved from the trachea, as well as for those with abnormal
the study period and admitted to their Newborn Intensive Care Unit chest roentgenograms.
required ventilation. The authors also presented data from 15 Although Gregory and colleagues are frequently cited as the
infants with MAS transferred to their unit subsequent to the study original investigators who showed benefits of intubation and
period. Of the 15 infants, 5 (33%) required either mechanical suctioning, Burke-Strickland and Edwards6 (Figure 4) published
ventilation or continous positive airway pressure, whereas 6 (40%) the results of their prospective trial of the technique a full year
developed a pneumothorax. On the basis of their experience, the earlier. The latter authors intubated and suctioned 84 meconium-
authors recommended intratracheal suctioning of all infants born stained infants over a 2-year period and compared their outcomes
with 17 such infants who did not receive tracheal cleansing.
Overall 70% of intubated infants also had saline lavage. The non-
intubated infants had more prolonged respiratory distress and
longer hospital stays compared with their intubated counterparts.
Burke-Strickland and Edwards recommended that all infants born
through MSAF of any consistency should be intubated and
suctioned.
Interestingly, 1 year after the Gregory publication, the results of
another study also based out of San Francisco were published.
Ting (Figure 5) and Brady7 did a retrospective review of outcomes
of 125 meconium-stained infants born over a 3-year period; of
these 28 babies were not intubated. Of these, 16 (57%)
developed MAS and 7 (25%) died. The remaining 97 infants were
intubated and suctioned; of these 27 (28%) developed MAS and
1 (1%) died. These investigators concluded that all infants born
through MSAF of any consistency should be intubated and
suctioned.
On the basis of their anecdotal experience, Carson et al.8
Figure 1 Dr L Stanley James (photograph courtesy of Dr Richard Polin). (Figure 6) suggested a different approach. They proposed that

Figure 2 Drs Lillian Blackmon and William Silverman (photograph courtesy of Dr Blackmon).

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S21

Figure 5 Dr Pauline Ting (photograph courtesy of Dr Ting).

Figure 3 Dr George A Gregory (photograph courtesy of Dr George Gregory).

Figure 4 Dr Nancy Edwards Dow (photograph courtesy of Dr Dow). Figure 6 Dr Bonita Carson (photograph courtesy of Dr Carson).

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S22

Figure 7 Clinician suctioning endotracheal tube directly with his mouth.

Figure 8 Dr Rebecca Bent (photograph courtesy of Dr Bent).

clinicians should visualize the hypopharynx and vocal cords of recommended in numerous textbooks and journals. The most
meconium-stained infants after oropharyngeal suctioning. These common technique (‘straw method’) consisted of providing
authors recommended that tracheal suctioning should be negative pressure at the hub of the endotracheal tube using one’s
performed only if meconium could be visualized at the vocal cords. mouth (Figure 7). Essentially, this practice was how residents in
Subsequent to these publications, there was widespread pediatrics gained and improved their intubation skills. Tasting
acceptance of the practice of intubation and suctioning of the meconium became a rite of passage for pediatric trainees.
trachea of all meconium-stained infants. This approach was Fortunately, most clinicians ultimately realized that a face mask

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S23

could be interposed between one’s mouth and the endotracheal


tube and obviated this culinary delight. In the decade following the
aforementioned publications, the incidence of MAS and deaths
attributable to the disorder appeared to decline significantly.9
It was recognized that there was a wide variation in the amount
of negative pressure produced by different individuals when directly
suctioning the endotracheal tube. Moreover, the infectious risks of
this practice were obvious. Some chose to directly place a standard
suction catheter or a de Lee suction catheter into the larynx. Many
devices (at least 20) were commercially developed as alternatives to
the straw method. Bent (Figure 8) et al.10 assessed the various
methods and found one to be consistently the best.
In 1988, Linder (Figure 9) and colleagues11 reported that a
selective approach might be more appropriate than a universal
one. They performed a trial in which apparently vigorous
meconium-stained infants were quasi-randomized to either
intratracheal suctioning or expectant management. Their results
were questioned because of major design flaws in the trial.
Although some suggested that a selective approach could be
deleterious, others found it to result in good outcomes. In the late
1990s, Wiswell et al.12 (Figure 10) spearheaded a large
Figure 10 Dr Thomas Wiswell (photography courtesy of Dr Wiswell).
international prospective, randomized controlled trial to assess a
selective approach. Almost 2100 meconium-stained neonates were
enrolled. During the 10 to 15 s after delivery, these babies had to be recommendations and advised that apparently vigorous
apparently vigorous (defined as having a heart rate >100 beats per meconium-stained infants do not need intubation and
min, having spontaneous respirations and having reasonable intratracheal suctioning.
tone). They were randomized either to be intubated and suctioned A future question that needs to be answered is whether or not
or to expectant management. The results of this approach are depressed meconium-stained infants actually benefit from having
given in the Table 1. There appeared to be no benefit to tracheal their airways intubated and suctioned. As many clinicians believe
cleansing of this population. Subsequent to this publication, the that most cases of MAS are due to in utero aspiration of MSAF, the
Neonatal Resuscitation Program (NRP) changed its potential efficacy of tracheal cleansing needs to be assessed in a
well-conducted randomized, controlled trial. Another issue that
needs to be addressed is the training of pediatric residents in
intubation skills. Historically, most residents honed their skills
during training by intubating virtually all meconium-stained
infants. However, by eliminating the need to intubate the majority
of the latter population, many believe that residents are no longer
gaining or maintaining these skills.

Intrapartum suctioning of the nasopharynx and oropharynx


Intrapartum suctioning consists of using either a suction catheter
or a bulb syringe to remove material from a meconium-stained
infant’s oropharynx and nasopharynx after the delivery of baby’s
head, but prior to delivery of the thorax. In the mid-1970s, Carson
et al.8 performed the previously mentioned study, the original goal
of which was to assess the value of tracheobronchial saline lavage
(Dr Carson, personal communication). They compared
meconium-stained infants managed differently during three
separate time periods. Group 1 infants (n ¼ 947) had no
Figure 9 Dr Nehama Linder (photograph available from the following website: intrapartum suctioning, no saline lavage and inconsistent
http://www.nacion.com/ln_ee/2007/octubre/16/_Img/1762242_0.jpg). intratracheal suctioning. Group 2 infants (n ¼ 381) had

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S24

Table 1 Incidence of MAS among 2094 apparently vigorous meconium-stained infants randomized to either intubation and suctioning or expectant management

Consistency of MSAF Intubation and suction group Expectant management group Significance
(n ¼ 1051) (%) (n ¼ 1043) (%)

Thin 5/447 (1.1) 2/453 (0.4) Not significant


Moderately thick 7/301 (2.3) 6/307 (2.0) Not significant
Thick 22/303 (7.3) 20/283 (7.1) Not significant
Overall 34/1051 (3.2) 28/1043 (2.7) Not significant
Abbreviations: MAS, meconium aspiration syndrome; MSAF, meconium-stained amniotic fluid.

were not statistically significant! Nonetheless, there was a


widespread assumption that the lower incidence of MAS in group 3
babies was due to the intrapartum suctioning. There was worldwide
acceptance of this hypothesis and it became the standard of care
for almost three decades.
Subsequent studies were unable to replicate Carson group’s
remarkably low incidence of MAS after intrapartum suctioning.
Most notable were the reports of Falciglia (Figure 11) and
colleagues. Initially, he performed a historical comparison
evaluation.13 He assessed the outcomes of meconium-stained
infants born in two different years (1975 and 1983, respectively).
During the earlier year, no intrapartum suctioning was customarily
performed, in contrast with the latter year when it was performed
routinely. There was no difference in the incidence of MAS in 1975
(2.0% of 742) compared with 1983 (2.1% of 755). Subsequently,
Falciglia et al.14 performed a concurrent observational study
during which an independent observer (unknown to the
obstetrician) documented whether or not obstetricians performed
‘early’ oronasopharyngeal suctioning while the baby’s head was at
the perineum (prior to delivery of the child’s thorax) compared
with those that received ‘later’ suctioning subsequent to delivery.
MAS was actually more common in the group that underwent
‘early’ suctioning (23/221, 10.4%) compared with those
undergoing ‘later’ suctioning (15/227, 6.9%).
To definitively assess whether or not this procedure was of
benefit to neonates, Vain (Figure 12) and colleagues15 performed
an international prospective, randomized controlled trial. The
Figure 11 Dr Horatio Falciglia (photograph available from the following population consisted of 2514 infants born through MSAF of any
website: http://www.cincinnatichildrens.org/research/div/neonatology/fs/fac/ consistency, gestational age X37 weeks and vertex (cephalic)
horacio-falciglia.htm?pres ¼ Hrs).
presentation. Subjects were randomized either to (1) suctioning of
the oropharynx and nasopharynx prior to delivery of the thorax or
intrapartum suctioning, intubation and tracheal suctioning, and to (2) no suctioning prior to delivery. No differences were found in
saline lavage. Group 3 infants (n ¼ 273) had intrapartum the incidence of MAS between suctioned (4.1%) and non-suctioned
suctioning and selective tracheal intubation/suctioning (if (3.8%) infants. Moreover, there were no differences in other key
meconium was present at the vocal cords). The authors intended to outcome variables including mortality, the need for mechanical
randomize those babies in group 3 who required intubation to ventilation, duration of mechanical ventilation and duration of
saline lavage or no lavage. However, only 2 of 273 group 3 infants supplemental oxygen use. Subsequent to this publication, both the
had meconium noted at the cords and were eligible to be lavaged. NRP and the American College of Obstetricians and Gynecologists
Hence, the authors were unable to assess whether saline lavage was (ACOG) changed their stances and no longer recommend
of benefit. The incidence of MAS in these populations was 1.9% intrapartum oropharyngeal and nasopharyngeal suctioning prior
(group 1), 1.8% (group 2) and 0.4% (group 3). These differences to delivery when MSAF is present.

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S25

Figure 12 Drs Nestor Vain and Luis Prudent (photograph courtesy of Dr Vain).

Other proposed delivery room therapies effort to prevent the child from inhaling deeply and aspirating fluid.
CPT None of the aforementioned maneuvers have ever been assessed
Chest physiotherapy is performed in an effort to prevent in clinical trials. All are potentially dangerous and none are
accumulation of debris and to improve mobilization of airway recommended.
secretions. CPT consists of techniques such as percussion,
vibration, postural drainage, saline administration and suctioning. Gastric suctioning
Theoretically, CPT should help remove meconium from the Karlowicz17 suggested suctioning of the stomachs of all
airways, prevent its consequences and improve gas exchange. It is meconium-stained neonates soon after delivery. He hypothesized
widely performed on infants born through MSAF, as well as those that some cases of MAS could potentially be caused by the postnatal
with MAS. Nevertheless, there are virtually no data to support this reflux of gastric contents into the oropharynx. The material could
therapy in meconium-stained neonates, either in the delivery room then potentially be aspirated into the airways. Suctioning of the
or thereafter. One must remember that there are potential stomach within minutes of delivery would obviate this from
complications of CPT (pneumothorax, hypoxemia, arrhythmia, occurring, although an intriguing hypothesis, gastric suctioning,
airway perforation and tissue damage). has never been evaluated for efficacy in preventing MAS.
Cricoid pressure, epiglottal blockage and thorax compression
Several other maneuvers have been suggested as being of benefit in Summary
preventing MAS.16 Cricoid pressure involves the application of Multiple delivery room therapies have been proposed as being
pressure to the neonate’s airway in an effort to compress it and effective in preventing or ameliorating MAS. However, to date, none
prevent intratracheal meconium from migrating distally. Epiglottal of the therapies assessed by randomized, controlled trials have been
blockage entails the placement of one or more fingers into the definitively proven to be effective. An important question remains:
infant’s hypopharynx in an attempt to apply pressure on the do depressed, meconium-stained neonates benefit by being
epiglottis and block the meconium passage downward. The latter two intubated and suctioned?
therapies have the potential to traumatize the airway. Both are likely
to produce a vagal response in a potentially compromised infant.
Thorax compression consists of manually encircling a Disclosure
meconium-stained infant’s chest and applying pressure in an TE Wiswell has declared no financial interests.

Journal of Perinatology
DR management of meconium-stained newborns
TE Wiswell
S26

References 10 Bent RC, Wiswell TE, Chang A. Removing meconium from infant trachea. What works
best? Am J Dis Child 1992; 146: 1085–1089.
1 Wiswell TE, Bent RC. Meconium staining and the meconium aspiration syndrome.
11 Linder NJ, Aranda V, Tsur M, Matoth I, Yatsiv I, Mandelberg H et al. Need for
Pediatr Clin North Am 1993; 40: 955–981.
endotracheal intubation and suction in meconium stained neonates. J Pediatr 1998; 112:
2 Wiswell TE, Fuloria M. Management of meconium stained amniotic fluid. Clin
Perinatol 1999; 26: 659–668. 613–615.
3 O’Donnell CPF, Gibson AT, Davis PG. Pinching, electrocution, ravens’ beaks, and 12 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery room
positive pressure ventilation: a brief history of neonatal resuscitation. Arch Dis Child management of the apparently vigorous meconium-stained neonate: results of the
2006; 91: F369–F373. multicenter, international collaborative trial. Pediatrics 2000; 105: 1–7.
4 James LS. Resuscitation procedures in the delivery room. In: Abramson H (ed). 13 Falciglia HS. Failure to prevent meconium aspiration syndrome. Obstet Gynecol 1988;
Resuscitation of the Newborn Infant. CV Mosby Co: St Louis, 1960, pp 141–161. 71: 349–353.
5 Gregory GA, Gooding CA, Phibbs RH, Tooley WH. Meconium aspiration in infants: a 14 Falciglia HS, Henderschott C, Potter P, Helmchen R. Does DeLee suction at the
prospective study. J Pediatr 1974; 85: 848–852. perineum prevent meconium aspiration syndrome? Am J Obstet Gynecol 1992; 167:
6 Burke-Strickland M, Edwards NB. Meconium aspiration in the newborn. Minn Med 1243–1249.
1973; 57: 1031–1035. 15 Vain N, Szyld E, Prudent L, Wiswell TE, Aguilar AM, Vivas NI. Oro- and nasopharyngeal
7 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obstet Gynecol suctioning of meconium-stained neonates before delivery of their shoulders: results
1975; 122: 767–771. of the international, multicenter, randomized, controlled trial. Lancet 2004; 364:
8 Carson B, Losey RW, Bowes Jr WA, Simmons MA. Combined obstetric and pediatric approach 597–602.
to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126: 712–715. 16 Cleary GM, Wiswell TE. Meconium-stained amniotic fluid and the meconium
9 Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a aspiration syndrome. An update. Pediatr Clin North Am 1998; 45: 511–529.
difference? Pediatrics 1990; 85: 715–721. 17 Karlowicz MG. More on meconium aspiration. Pediatrics 1990; 86: 1007–1008.

Journal of Perinatology
Journal of Perinatology (2008) 28, S27–S29
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Neonatal resuscitation guidelines for ILCOR and NRP: evaluating
the evidence and developing a consensus
J Kattwinkel
Division of Neonatology, Department of Pediatrics, University of Virginia Health System, Charlottesville, VA, USA

Journal of Perinatology (2008) 28, S27–S29; doi:10.1038/jp.2008.146 versus room air, various issues related to temperature
management, ventilation strategies during resuscitation,
I have been asked to describe the process used by the American components and timing associated with volume expansion, glucose
Academy of Pediatrics (AAP) and American Heart Association (AHA) management following intensive resuscitation, use of CO2 detectors
to evaluate the evidence for the purpose of developing for confirmation of tube placement, epinephrine dosing and routes
recommendations for the Neonatal Resuscitation Program. This of administration, ethical considerations regarding initiating and
symposium is primarily interested in the recommendations discontinuing resuscitation, as well as various aspects of managing
regarding management of meconium during neonatal meconium prior to, during and following delivery.
resuscitation, but I will first describe the neonatal program and the After the issues are defined, ILCOR representatives and
process used in development of all AHA and AAP resuscitation consultants are asked to begin the evidence evaluation process by
recommendations. preparing worksheets on each topic. Worksheet development begins
The Neonatal Resuscitation Program (NRP) is a standardized with an intensive review of the literature. Over 30 000 abstracted
instruction in neonatal resuscitation. It includes a and critiqued references are cataloged in an Endnotes database
self-instructional textbook and a set of skills taught by instructors. maintained at the AHA. Pertinent studies are then classified
The instructors first involved national faculty who were given the according to their level of evidence, ranging from level 1
responsibility of training regional instructors in their respective (randomized controlled trial), through levels 4 (historic,
states, who then trained hospital-based instructors, who in turn nonrandomized cohort study) and 6 (animal or mechanical model
trained participants. The program was developed by collaboration study), to level 8 (rational conjecture or common practice). Each
of the AHA and AAP and is managed and revised by the NRP study is also ranked as to the quality of evidence (fair, good or
Steering Committee administered by the AAP. There have been over excellent) and whether the study is supportive, neutral or opposes
2.2 million participants in the United States since inception of the the evidence. The neonatal resuscitation worksheets are available
NRP nearly 20 years ago, and the program has also been for review by accessing the NRP website (http://www.aap.org/nrp/
implemented in 106 other countries. Revisions are made nrpmain), clicking on ‘Science’ and ‘Evidence-Based Guidelines’,
approximately every 5 years. and then accessing the link to the worksheets maintained on the
The revision process involves six steps: define the issues, review AHA website. There are three worksheets addressing issues about
the literature, debate the evidence, publish guidelines, produce meconium.
educational materials, and deliver the program.1 The first three The worksheets serve as the focus for a series of ILCOR debates
steps are conducted in collaboration with the International Liaison that take place over approximately 3 years, which culminates in
Committee on Resuscitation (ILCOR), which then publishes a the Evidence Evaluation (E-2) conference during which consensus
document entitled Consensus on Science and Treatment is reached on the science. This conference forms the basis for the
Recommendations (COSTR). Each of the resuscitation councils COSTR document, the most recent version of which was jointly
represented on ILCOR then develops separate resuscitation published in Circulation2 and Resuscitation3 and the pediatric
guidelines appropriate for their respective countries. and neonatal portions republished in Pediatrics.4 The NRP
The neonatal resuscitation issues considered during the most Steering Committee then considered the practical implications of
recent review included topics such as the use of 100% oxygen COSTR in drafting the NRP Resuscitation Guidelines that were also
published in Circulation5 and Pediatrics.6
Correspondence: Dr J Kattwinkel, Division of Neonatology, Department of Pediatrics,
University of Virginia Health System, PO Box 800386, Charlottesville, VA 22908, USA.
Three resuscitation-associated meconium issues were examined
E-mail: jk3f@virginia.edu during the most recent evidence evaluation process.
Neonatal resuscitation guidelines for ILCOR and NRP
J Kattwinkel
S28

Does amnioinfusion reduce the incidence or severity Table 1 Experience with meconium births, as reported by Carson et al.9
of meconium aspiration syndrome? 7/1970–4/1974 5/1974–3/1975 4/1975–11/1975
It has been hypothesized that infusion of saline into
meconium-contaminated amniotic fluid will dilute the meconium Births 7585 2320 1681
and decrease the potential for developing obstruction of airways Meconium in 12.5% 16.4% 16.2%
amniotic fluid
and development of meconium aspiration syndrome. Review of the
MAS 18* 7** 1*,**
literature revealed 23 reports supporting the hypothesis:
MAS deaths 5 0 0
amnioinfusion significantly reduced the frequency of meconium
aspiration syndrome, of meconium below the cords and neonatal Abbreviation: MAS, meconium aspiration syndrome.
Period 1: endotracheal suctioning under direct vision. Period 2: intrapartum
acidemia, and was associated with a lower overall cesarean section suction+tracheobronchial lavage. Period 3: intrapartum suction+endotracheal
rate. Nine studies showed no significant benefit of amnioinfusion. suction (2)±lavage.
Three reports suggested that any demonstrated benefit of *P ¼ 0.18; **P ¼ 0.25.
amnioinfusion may have been a reflection of reversing fetal heart
rate decelerations rather than due to dilution of meconium. There
were also case reports of uterine rupture when amnioinfusion was suction the trachea immediately following birth, and then
administered during a trial of labor. In view of the conflicting data, performed a tracheobronchial lavage with normal saline. During
insufficient input from the obstetrics profession, and knowledge of the third epoch of 8 months (period 3), the intrapartum suctioning
an ongoing randomized control trial, the Committee decided to continued to be performed, but neonatal suction and lavage were
take no position on amnioinfusion during this evidence evaluation less common. Even though there were no significant differences in
cycle. Although the Guidelines were in press, the Fraser et al.7 trial the incidence of meconium aspiration syndrome among any of the
was published (as also reported at this conference), and the epochs, the practice of intrapartum suctioning was recommended
American College of Obstetrics and Gynecology released a and adopted by the vast majority of clinicians for the next 30 years,
Committee Opinion that ‘routine prophylactic amnioinfusion for probably because the practice appeared to be noninvasive and
the dilution of meconium-stained amniotic fluid is not seemed logical.
recommended.’8 However, the practice of intrapartum suctioning continued
to be questioned by various obstetricians and neonatologists, and in
2000 to 2001 Vain et al.11 conducted a randomized, multicenter,
control trial of 2514 meconium-stained deliveries occurring in
Should the current recommendation, to always 10 centers in Argentina and 1 in the United States.
perform intrapartum suctioning, be continued? Meconium-stained births were randomized to receive either
Since the Carson et al.9 study in 1976, there has been a generally pharyngeal suctioning of the fetus before delivery of the shoulders
accepted recommendation that babies born with meconium-stained or no intrapartum suctioning. Although there were 5% protocol
amniotic fluid should have their nose, mouth and pharynx deviations, an intent-to-treat analysis revealed no significant
suctioned after delivery of the head, but before delivery of the difference in the incidence of meconium aspiration syndrome
shoulders (intrapartum suctioning). The recommendation was between the two groups. The relative risk was 0.9, with 95%
based on a probably erroneous assumption that most meconium confidence limits of 0.6 to 1.3. Primarily as a result of the Vain
aspiration syndrome (MAS) aspiration occurred during the study, ILCOR and the latest edition of NRP have stated the
establishment of air breathing and that removal of the meconium following: ‘Current recommendations no longer advise routine
while the chest was still compressed in the birth canal could intrapartum oropharyngeal and nasopharyngeal suctioning for
prevent MAS. The Carson et al. study reviewed retrospectively all infants born to mothers with meconium staining of amniotic fluid
births that occurred at the University of Colorado during three time (class I).’ It should be noted that, although there was some
periods (Table 1). disagreement during development of the recommendation, the
In total, 12 to 16% of the babies born in 1970 to 1975 had NRP Steering Committee is on record as emphasizing that the new
meconium in their amniotic fluids. For the first 46 months recommendation should not be interpreted to mean that
(period 1), the standard had been not to perform intrapartum intrapartum suctioning of meconium-stained babies is
suctioning, but to perform direct endotracheal suctioning of all contraindicated, but merely that the previously recommended
meconium-stained babies after delivery. This post-delivery practice practice of routinely performing suctioning in such babies is no
had been initiated following the observation of Gregory et al.10 in longer recommended. Several members felt that clearing of the
1974 that 56% of meconium-stained neonates had meconium oropharynx to facilitate the possible need for subsequent
recovered from below the cords. For the next 10 months (period 2), visualization of the trachea for direct suctioning, still seems
Carson et al. performed intrapartum suctioning, continued to reasonable.

Journal of Perinatology
Neonatal resuscitation guidelines for ILCOR and NRP
J Kattwinkel
S29

Is the current recommendation, to limit endotracheal procedure was uncontrolled and was published over 30 years ago,
suctioning only to the nonvigorous newborn, still valid? and as there is increasing evidence that much of meconium
Traditional teaching has recommended that meconium-stained aspiration occurs well before birth and as essentially every report of
infants have endotracheal intubation immediately following birth endotracheal suctioning since the report of Gregory et al. reports
and that suction be applied to the endotracheal tube as it is some morbidity associated with the procedure, it would seem well
withdrawn. As noted above, this recommendation stemmed from justified for there to be a modern-day large, randomized,
the observation of Gregory et al.10 who reported on experience with multicenter trial comparing endotracheal suctioning versus no
1000 births, of whom 8.8% were meconium stained and 91% of endotracheal suctioning of the nonvigorous meconium-stained
those 88 had received direct endotracheal suction. Meconium newborn immediately following birth. Until such a study is
was recovered from the trachea in 56% (46 babies) and 17% performed, it is likely that subsequent NRP guidelines will continue
(8 patients) of those had had none in ‘mouth or larynx’ during to recommend that the procedure be performed.
laryngoscopy. Although this was not a control trial, from these
observations, these clinician investigators recommended that, ‘all
infants born through thick or ‘pea soup’ meconium should have Disclosure
their trachea aspirated immediately after birthy’. Again, the J Kattwinkel has received grant support from the American Academy of Pediatrics
recommendation was universally accepted and became the and the National Institutes of Health.
standard until Wiswell et al.12 conducted a randomized multicenter
control trial comparing the practice of performing endotracheal
suctioning on all babies, versus only suctioning those babies who References
were nonvigorous. Vigorous was defined as strong respiratory effort, 1 Perlman JM, Kattwinkel J. Delivery room resuscitation: past, present, and future. Clin
good muscle tone, and a heartrate greater than 100 beats per Perinatol 2006; 33: 1–9.
minute. Over 26 months, at 12 centers, 2094 meconium-stained 2 International Liaison Committee on Resuscitation. 2005 International Consensus on
Cardiopulmonary resuscitation and emergency cardiovascular care science with
neonates who were classified as ‘vigorous’ at birth, were
treatment recommendations. Circulation 2005; 112: III-1–III-136.
randomized to receive endotracheal suctioning versus conservative 3 International Liaison Committee on Resuscitation. 2005 International Consensus on
therapy. There was no significant difference in the incidence of Cardiopulmonary resuscitation and emergency cardiovascular care science with
meconium aspiration syndrome or other lung disease between treatment recommendations. Part 7. Neonatal resuscitation. Resuscitation 2005; 67:
those suctioned versus those not suctioned. As a result, ILCOR and 293–303.
4 The International Liaison Committee on Resuscitation (ILCOR) consensus on science
the NRP revised their recommendation in 2000 to limit
with treatment recommendations for pediatric and neonatal patients: neonatal
endotracheal suctioning only to those babies who were meconium resuscitation. Pediatrics 2006; 117: e978–e988.
stained and not vigorous at birth. We finally stopped the practice of 5 American Heart Association (AHA) Guidelines for Cardiopulmonary Resuscitation
chasing the healthy baby around the delivery room with a (CPR) and Emergency Cardiovascular Care (ECC) of pediatric and neonatal patients:
laryngoscope. neonatal resuscitation guidelines. Circulation 2005; 112: IV-188–IV-195.
There are still several unanswered questions regarding the 6 American Heart Association (AHA) Guidelines for Cardiopulmonary Resuscitation
(CPR) and Emergency Cardiovascular Care (ECC) of pediatric and neonatal patients:
appropriate management of the meconium-stained baby at birth
neonatal resuscitation guidelines. Pediatrics 2006; 117: e1029–e1038.
that have not received definitive answers in the most recent COSTR 7 Fraser WD, Hofmeyr J, Lede R, Faron G, Alexander S, Goffinet F et al. Amnioinfusion
document and the 2005 NRP Guidelines. First, is amnioinfusion an for the prevention of the meconium aspiration syndrome. N Engl J Med 2005; 353:
effective procedure for diluting meconium and reducing meconium 909–917.
aspiration syndrome? The above-mentioned Fraser et al.7 trial and 8 American College of Obstetrics and Gynecology (ACOG). Committee Opinion no. 346;
American College of Obstetrics and Gynecology statement8 appear October, 2006.
9 Carson BS, Losey RW, Bowes WA, Simmons MA. Combined obstetric and pediatric
to have answered that question and will likely influence the next approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126:
evidence evaluation documents. Second, is intrapartum suctioning 712–715.
contraindicated, rather than simply not recommended as a routine 10 Gregory GA, Gooding CA, Phibbs RH, Tooley WH. Meconium aspiration in
procedure? It seems unlikely that another larger control trial will infantsFa prospective study. J Pediatr 1974; 85: 848–852.
be conducted to answer this question and there appears to be little 11 Vain NE, Szyld EG, Prudent LM, Wiswell TE, Aguilar AM, Vivas NI. Oropharyngeal and
nasopharyngeal suctioning of meconium-stained neonates before delivery of their
evidence suggesting that intrapartum oropharyngeal suctioning is
shoulders: multicentre, randomised controlled trial. Lancet 2004; 364: 597–602.
particularly hazardous. And finally, is direct endotracheal 12 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery room
suctioning of the newborn of any description indicated following management of the apparently vigorous meconium-stained neonate: results of the
birth? As the original observation that formed the basis for the multicenter, international collaborative trial. Pediatrics 2000; 105: 1–7.

Journal of Perinatology
Journal of Perinatology (2008) 28, S30–S35
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Developing a systems approach to prevent meconium aspiration
syndrome: lessons learned from multinational studies
VK Bhutani
Division of Neonatal and Developmental Medicine, Department of Pediatrics, Lucile Salter Packard Children’s Hospital, Stanford
University School of Medicine, Stanford, CA, USA

(8/40 or 0.035% of live births) needed innovative ventilatory support. None


Passage of fetal bowel movement (meconium) is common (in about one
died or needed ECMO. These data describe the components for a systems
out of six births), and in some the staining of the amniotic fluid is a sign of
approach to prevent and manage adverse outcomes related to MSAF at the
fetal distress. Inhalation of meconium (aspiration syndrome, in upto one
regional level II or III perinatal center. Replication of a similar
out of five to eight such births) just before or at birth may be preventable by
strategy may be more relevant to cost containment and be a safer
a coordinated approach by well-trained and informed birth attendants.
approach for neonates at risk for MAS-related respiratory failure. This paper
Respiratory failure secondary to meconium aspiration syndrome (MAS)
assess the evidence for pivotal steps needed to prevent MAS and
remains a major cause of morbidity and mortality in the neonatal
ensuing neonatal death and disease in the context of diverse perinatal
population. Infants with hypoxemic respiratory failure because of MAS,
health services.
persistent pulmonary hypertension of the newborn and pneumonia/sepsis
have an increased survival with extracorporeal membrane oxygenation
Journal of Perinatology (2008) 28, S30–S35; doi:10.1038/jp.2008.159
(ECMO). Other treatment options earlier limited to inotropic support,
continuous airway pressure (CPAP), conventional ventilatory management,
respiratory alkalosis, paralysis and intravenous vasodilators have been Introduction
replaced by synchronized intermittent mandatory ventilation (SIMV), Optimizing the management of infants born with meconium
high-frequency oscillatory ventilation (HFOV), surfactant and inhaled nitric staining, relevant to the Indian macro- and micro-health
oxide (iNO). HFOV has been advocated for use to improve lung inflation environment, may prevent adverse outcomes owing to perinatal
while potentially decreasing lung injury through volutrauma. Other reports asphyxia, meconium aspiration syndrome (MAS), and ensuing
describe the enhanced efficacy of HFOV when combined with iNO. respiratory failure. The predictive risk factors for MAS in infants
Subsequent to studies reporting that surfactant deficiency or inactivation delivered through meconium-stained amniotic fluid (MSAF) have
may contribute to neonatal respiratory failure, exogenous surfactant therapy not been ascertained prospectively. On the basis of a retrospective
has been implemented with apparent success. Recent studies have shown study reported by Usta et al.,1 odds ratio >3 was identified when
that iNO therapy in the neonate with hypoxemic respiratory failure can an infant’s delivery was associated with the (a) induction for
result in improved oxygenation and decreased need for ECMO. However, non-reassuring fetal heart rate (FHR) pattern, (b) need for
these innovative interventions are costly, require a sophisticated endotracheal intubation (ET), (c) Apgar p3 at 1 min or (d) need
infrastructure and are not universally accessible. In this paper, a context of for a Cesarean section (Table 1). This study informed and
systems-approach for prenatal, natal and postnatal management of babies influenced clinical delivery room practice for several years. In
delivered through meconium stained amniotic fluid (MSAF) so that adverse addition, the management of meconium at the time of birthing
outcomes are minimized and the least number of babies require innovative has been examined from two perspectives: (i) suctioning of the
ventilatory support is described. Previously reported data from a single meconium from an infant’s upper airway after delivery of the head
urban perinatal center (Philadelphia, PA, USA), over a 6-year period but before delivery of the shoulders (intrapartum suctioning,
(1995–2000), demonstrated that 14.5% (3370/23 175 of live births babies oronasopharyngeal suctioning) and (ii) suctioning of an infant’s
were delivered with MSAF. These data also showed that 4.6% of babies (155/ trachea immediately after birth with an ET. Recommendations for
3370) with MSAF sustained MAS. Overall, 26% of babies (40/155) with MAS intrapartum suctioning for meconium had been based on
needed ventilatory support (or 0.17% of all live births); of these, only 20% consensus of studies that have yielded conflicting results about the
value of intrapartum oronasopharyngeal suctioning of infants born
Correspondence: Dr VK Bhutani, Division of Neonatal and Developmental Medicine, Lucile
with MSAF. A more recent large multicenter randomized trial found
Salter Packard Children’s Hospital at Stanford University School of Medicine, 750 Welch Ave,
#315, Palo Alto, CA 94305, USA. that intrapartum suctioning of meconium does not reduce the
E-mail: bhutani@stanford.edu incidence of MAS. The latter study has led the International Liaison
Developing a systems approach to prevent MAS
VK Bhutani
S31

Table 1 Clinical risk factors for MAS in infants born through MSAF MAS, occurs in 2 to 9% of neonates born through MASF and has a
Clinical risk factors Odds ratio
high mortality rate of 40%.5,7,10–13 At delivery, meconium is found
below the vocal cords in 20 to 45% of neonates born through
1 Induction for non-reassuring fetal heart rate 6.9 MSAF.4,10,13,14 If it has not been aspirated into the lungs, the
2 Need for intubation 4.9 removal of meconium from the airways before the first breath can
3 Apgar p3 at age 1 min 3.1 reduce the incidence of MAS. This preemptive removal of
4 Cesarean section 3.0 meconium from the airways can be performed at the time of
5 Previous cesarean section 2.5 delivery (intrapartum suctioning) or immediately after delivery
Abbreviations: MAS, meconium aspiration syndrome; MSAF, meconium-stained amniotic (postpartum suctioning). Intrapartum suctioning consists of
fluid. clearing the mouth, pharynx and nose with either a large-bore
Based on live births from 1990 to 1993 (Memphis, TN, USA) with MASF n ¼ 937 and
MAS in n ¼ 39 (4.2%). suction catheter (12 to 14 mm caliber) or a bulb syringe as soon as
Adapted from Usta et al.1 the head is delivered but before delivery of the shoulders.
Postpartum suctioning consists of intubating and suctioning the
trachea before performing the other steps of resuscitation.
Committee on Resuscitation2 or ILCOR to recommend against Respiratory failure is a major cause of morbidity and mortality in
routine intrapartum oronasopharyngeal suctioning for infants born the neonatal population. Infants with hypoxemia develop
with MASF. On the other hand, the current recommendation for ET respiratory failure because of MAS, persistent pulmonary
suctioning is based on a pivotal randomized, controlled trial that hypertension of the newborn and pneumonia/sepsis. Recently,
showed that ET intubation and suctioning for vigorous infants at Bhutani et al.15 suggested a system-based strategy comprising the
birth offers no benefit. It is also known that the benefit of ET prenatal, natal and postnatal management of babies delivered
suctioning in meconium-stained, depressed infants has not been through MASF, so that the adverse outcomes are minimized and
systematically studied. However, the current ILCOR treatment the least number of babies require innovative ventilatory support.
recommendation is that meconium-stained, depressed infants At an urban perinatal center in Pennsylvania, over a 6-year period
should receive ET suctioning immediately after birth and before (1995 to 2000), 14.5% (3370/23 175 of live-birth babies) were
stimulation, presuming that the equipment and expertise is delivered with MSAF (Figure 1). These data show that 4.6% of
available and that ET suctioning is not necessary for infants with babies (155/3370) with MSAF sustained MAS (Figure 2). Overall,
MASF who are vigorous. The relevance for application of ILCOR 26% (40/155) of babies with MAS needed ventilatory support
recommendations to a population with diverse access to perinatal (0.17% of all live births) (Figure 3); of these, only 20% (8/40 or
health care has been questioned. The studies and 0.035% of live births) needed innovative ventilatory support. Infants
recommendations cited by ILCOR are reported from those with low Apgar scores were more likely to need ventilatory support
conducted in developed nations and nations with sophisticated (Figure 4). None died or needed extracorporeal life support.
medicalized health-care systems. These recommendations may not
be applicable in areas with low income resources, births conducted
at home or limited perinatal access to evidence-based medical Components of a systems approach to prevent MAS
care.3 Dramatic improvements in the overall birthing conditions Interdisciplinary health-care perspectives
and the continuing advances in perinatal-neonatal practices Depending on the likelihood of MAS as a potential but significant
during the last decade have yet to universally affect the neonatal morbidity, the development of a ‘rapid response team’ was
unacceptably high risk of morbidity and mortality in the neonatal envisioned as an institutional policy (prevalent in most US birthing
population. Earlier experiences have led us to believe that building facilities) with the goal of modulating an intensive care nursery
an interdisciplinary leadership approach that lends itself to a census and also the risk of adverse neonatal outcomes. The
systems approach would provide innovative strategies to bridge the potential reduction in neonatal mortality and cost containment
existing access barriers in micro- and macro-health environments would be the anticipated advantages of team-based interventions.
of diverse social and cultural backgrounds. The team would constitute the obstetrician, pediatrician (and/or
neonatologist), the perinatal nurses (obstetrical and neonatal) and
the family, supported by the maternal child health administrators.
Global perspective of births with MASF Although the obstetrician has a task-oriented responsibility to
Meconium-stained amniotic fluid is found in 7 to 20% of take care of the mother and the family through the performance
pregnancies at the time of delivery.3–7 MSAF is associated with fetal and delivery of quality of care services in the delivery room, a
acidosis, abnormalities in FHRs and low Apgar scores, suggesting personal agenda is to strive for a ‘HAPPY’ family experience. This is
hypoxia as the stimulant of passage of meconium in utero.8,9 The achieved through minimally atraumatic birthing with the
most severe condition associated with meconium passage in utero, prevention of fetal hypoxemia and acidosis, and an effective

Journal of Perinatology
Developing a systems approach to prevent MAS
VK Bhutani
S32

4500 90
83
4000 80
3500
70
Live births

3000

Number of babies
60
2500
2000 50
1500 40
1000 30 26
24
500
20
0 7 8 8
1995 1996 1997 1998 1999 2000 10
0
Figure 1 Annual incidence of meconium staining of the amniotic fluid (MSAF) Room air Oxygen CPAP SIMV SIMV+Surf HFV+iNO
(1995 to 2000) at a single US urban perinatal center: n ¼ 3370 infants with
MSAF/23 175 live births (15.5%). Of these, 945 infants (4.4%) were intubated Figure 3 Modes of respiratory support in newborns with meconium aspiration
(based on prevailing Neonatal Resuscitation Program recommendations). Open syndrome (MAS) (n ¼ 40 needed support/155 live births with MAS, 25.8%).
bars are annual live births; speckled bars are live births with MSAF.
120

100

Number of babies
Number of babies with MSAF

700
80
600
500 60
Ventilatory
400 Ventilatory
40 support (n =24/132)
support (n =16/23)
300
20
200 None
None
0
100 Low apgar score Normal apgar score

0
1995 1996 1997 1998 1999 2000 Figure 4 Effect of low Apgar score (<3 at age 1 min or <6 at age 5 min) on
need for ventilatory support in infants with meconium aspiration syndrome
Figure 2 Annual occurrence of meconium aspiration syndrome (MAS) (1995 to (MAS). Over a 6-year period, 1995 to 2000, n ¼ 24/132 (18.2%) infants with
2000) in infants delivered through meconium staining of the amniotic fluid normal scores need ventilatory support as compared with n ¼ 16/23 (69.5%) with
(MSAF) (n ¼ 155 infants with MAS/3370 infants with MSAF ((4.6%). Speckled low scores (P<0.001). Solid bars are infants with MAS who needed ventilatory
bars are infants with MSAF and solid bars are infants with MAS. support, and the open bars are infants who were observed for need of oxygen/
ventilatory support.

management of maternal and personnel stress. The perinatal resuscitation, performance standards and intubation skills of the
birthing nurse provides support, educates and cares for the family team are now routine at all birthing facilities. The team needs to
during the changing birthing environment. A seemingly benign acquire the prenatal history and the evolving perinatal events,
process has the potential of disintegration as erstwhile birthing directly observe the natal events and render as well as execute
plans are shelved. Participation in clinical management includes postnatal management decisions. The relevant prenatal history
fetal monitoring and appropriate availability and documentation of acquired through the attending obstetrician and nurse would
instrumentation. A pediatrician needs to be accessible in a timely include the intensity of meconium staining, status of fetal
manner for unpredictable and ‘stat’ calls and have the skills and well-being and FHR patterns and fetal pH (if measured) data on
ability to perform (without anxiety) in an acute stressful response. the use of amnioinfusion, and risk factors for perinatal infections
In addition, the pediatrician needs to clarify any confusion on the and for any maternal-placental complications. The latter include a
amount of meconium staining and the status of fetal well-being, prolonged second stage of labor, abruption of the placenta,
and to make a decision of the likelihood of an elective neonatal placenta previa or cord accidents (nuchal cords, true knots, cord
intubation. The family perspectives are overwhelmed by the sudden compression and cord prolapse), abnormal fetal presentations,
medicalization of the birthing process and are aggravated by maternal hypertension, pre-eclampsia, intrauterine growth
anxiety, stress, interruption of bonding and loss of privacy with retardation, post-maturity or placental calcifications. FHR
unanticipated instrumentations and participation of often monitoring is a predominant method to assess fetal oxygenation in
unselected and unsolicited providers. labor; however, it is a nonspecific, indirect and inaccurate measure
of fetal hypoxia and acidosis. Electronic fetal monitoring is
Performance and judgment of the ‘rapid response team’ associated with increased Cesarean section because of
Besides being awake and alert, the team needs to have a ‘non-reassuring’ FHR patterns. These include FHR between 100
physiologic understanding and risk assessment ability of the and 120 with no accelerations, FHR <100 beats/min with
clinical progression of MAS. Credentialing for neonatal accelerations, increased heart rate variability of >25 beats/min for

Journal of Perinatology
Developing a systems approach to prevent MAS
VK Bhutani
S33

>30 min, late decelerations (>1 per 30 min), persistent late present in the trachea in 37% (n ¼ 238) of neonates born through
decelerations (>50% of contraction), fetal tachycardia MSAF, and MAS developed in 9.2% of cases.
(>160 beats/min) or a sinusoidal pattern. Natal observations
provide an assessment of fetal handling during instrumentation Studies that do not recommend intrapartum suctioning.
(such as vacuum extractions, forceps delivery and neck A large multicenter, randomized controlled clinical trial enrolled
manipulation as well as the handling of the baby’s head by the 2514 patients with MSAF with the desired goal of assessing the
obstetrician, and the presence of nuchal cords. Once the infant’s effectiveness of intrapartum suctioning for the prevention of MAS.18
head is delivered, the activity and vigor status and the resuscitation This study also examined the effect of intrapartum suctioning in
process are initiated, as indicated. Once the infant is stabilized, the high-risk subgroup infants (those with thick MSAF) with abnormal
umbilical cord status is ascertained meconium staining, presence FHR patterns, infants with delivery by Caesarian section and infants
of venous clots and insertion at the placenta. needing resuscitation in the delivery room. This study showed that
intrapartum suctioning does not reduce the incidence of MAS
Postnatal management decisions (relative risk (RR): 0.9, 95% confidence interval (CI): 0.6 to 1.3),
When meconium is present in the amniotic fluid, the current need for mechanical ventilation (RR: 0.8, 95% CI: 0.4 to 1.4) and
ILCOR recommendations are to defer suctioning of the mortality (RR: 0.4, 95% CI: 0.1 to 1.5).
hypopharynx on delivery of the head. If the meconium-stained
newly born infant has absent or depressed respirations, heart rate Current ILCOR recommendations. On the basis of these large
or muscle tone, residual meconium should be suctioned from the studies, the current guidelines of the American Academy of
trachea. Ventilation is of primary concern. The role of intrapartum Pediatrics and the American Heart Association through the
suctioning has been a subject of controversy and recent research. Neonatal Resuscitation Program and Pediatric Working Group of
the ILCOR1 no longer recommend routine intrapartum
Studies in favor of intrapartum suctioning. Intrapartum oronasopharyngeal suctioning by an obstetrician before delivery of
suctioning has been considered standard practice for more than the shoulder. Discretionary intrapartum suction also remains a
25 years on the basis of a study by Carson et al.16 In this standard of care. This decision is not based on a meta-analysis,
before-and-after trial, they reported an incidence of 1.9% but, on the basis of consensus of opinions that are most relevant to
(n ¼ 947) for MAS with a mortality rate of 28% during a period the care of infants already affected, improved the dating of
when only postpartum suctioning was performed, as compared pregnancy by a lowering of the gestational age at birth (fewer
with MAS incidence of 0.4% (n ¼ 273, P ¼ 0.07) and no deaths deliveries for pregnancies >41 weeks), earlier recognition of fetal
when intrapartum suctioning was also performed in addition distress and improved access to sophisticated birthing facilities in
to postpartum suctioning. The effectiveness of intrapartum developed nations. The other currently recommended approach to
suctioning was confirmed in a subset of patients enrolled in a prevent MAS is immediate postnatal ET suctioning in non-vigorous
multicenter randomized controlled trial looking into the efficacy (depressed) babies born through MSAF. In nations and
of postpartum suctioning in vigorous newborns.17 In this study by communities with improved perinatal health-care access, an
Wiswell et al., the incidence of MAS was 8.5% in infants who observational and non-intervention strategy is indicative of
did not have intrapartum suctioning (n ¼ 94), as compared eminent and cautious practice. Mandatory intrapartum suctioning
with 2.7% in infants who had intrapartum suctioning (n ¼ 54; in babies born through MSAF is being discontinued, largely on the
P ¼ 0.013). basis of a single large randomized controlled study conducted in a
setting with facilities for intensive fetal monitoring, prompt
Equivocal studies. Two prospective and non-randomized clinical response to fetal distress and 24 h availability of personnel trained
trials by Falciglia et al.10,14 compared early suctioning (suctioning in neonatal resuscitation, including postnatal intubation and ET
by the obstetrician before delivery of the thorax) and late meconium suctioning.
suctioning (suctioning by the obstetrician after delivery of the
thorax) and showed no difference in the incidence of MAS. In the Impact of different clinical settings. Most of the deliveries in
first study, no differences in the rate of meconium below the cords these countries take place either at home or at ill-equipped and
(36 vs 37%) or the incidence of MAS (20% in each group) between understaffed hospitals. The profile of women and their neonates
early and late suctioning were noted.8 The second study reported a born in a developing country setting contrasts sharply with that of
higher rate of meconium below the cords (53%) among the early developed countries. Even in the study by Vain et al.,18 which
suctioning group compared with the late suctioning group, which included 11 sites in Argentina, a middle-income country, the
had a rate of 36% (P<0.001), but there was no difference in the clinico-epidemiologic profile was not comparable with the
incidence of MAS between the two groups (P>0.05).13 Rossi et al.13 diverse environment and communities encountered in India
reported that despite intrapartum suctioning, meconium was (see Table 2). The Indian experiences document high rates of

Journal of Perinatology
Developing a systems approach to prevent MAS
VK Bhutani
S34

Table 2 Comparison of data from an Indian Database3 reported by Vain et al.18

Variable NNPD3 Vain et al.18


18 sites in India 11 sites in Argentina and 1 in the United States

Babies with MSAF (n ¼ 12 156) Intrapartum suction+ (n ¼ 1263) Intrapartum suction (n ¼ 1251)

Birth weight (g, mean±s.d.) 2646±552 3413±483 3400±496


Small for gestation 1566 (12.9%) 39 (3%) 48 (4%)
Cesarean section 5208 (42.8%) 401 (32%) 398 (32%)
Pregnancy-induced hypertension 961 (7.9%) 65 (5%) 65 (5%)
Eclampsia 177 (1.5%)
Oligohydramnios 360 (3.0%) 17 (1%) 20 (2%)
Fetal bradycardia (<120 b.p.m.) 1523 (12.5%) 145 (11%) 130 (10%)
Fetal tachycardia (>160 b.p.m.) 369 (3.0%)
Meconium aspiration syndrome 1896 (15.6%) 52 (4%) 47 (4%)
Abbreviations: MSAF, meconium-stained amniotic fluid; NNPD, National Neonatal Perinatal Database.

pregnancy-related complications such as pregnancy-induced compromised. In such conditions, the measurements of arterial
hypertension, oligohydramnios, intrauterine growth retardation blood gases or chest radiographs may not be necessary. If
and post-maturity. In addition, in the Indian settings, postnatal metabolic acidosis is documented for persistent tachypnea with
intubation and ET suctioning of non-vigorous infants could be hypoxemia, base correction with bicarbonate is not essential and
jeopardized if discretionary recommendations are made without crystalloid infusion is likely to be sufficient. Persistent tachypnea-
specific, practical and easily implemented algorithms. associated hypoxemia and hypercapnia would be indications for a
Low- and middle-income nations account for the majority of chest radiograph evaluation. When an infant is stabilized, postnatal
the 130 million live births and 4 million neonatal deaths occurring gastric suction, presumably to prevent postnatal aspiration, is not
each year.19 Therefore, we speculate that the relatively simpler and evidence-based and, similarly, gastric lavage is not necessary.
easier techniques to perform intrapartum suctioning and the Infants do need to be monitored for airway stability and subsequent
availability of appropriate biotechnologies, such as large-bore, choking/bradycardia.
portable, disposable and easily manipulated suction devices for the
mouth, nose and/or trachea that are non-traumatic, sterile and Subsequent postnatal management
independent of wall suction, may still have a role in the prevention Infants are likely to need close glucose monitoring for hypoglycemia
of MAS, whereas improvements of perinatal health-care access are because of concomitant risk factors. Intravenous dextrose infusions
initiated and implemented. and delayed enteral nutrition need to be considered for persistent
tachypnea (owing to hypoxemia and/or hypercapnia), low Apgar
Understanding immediate postnatal tachypnea and clinical scores and hypoglycemia. Screening for sepsis would be based on the
signs presence of known maternal risk factors or an abnormal chest
The most frequent benign etiology for tachypnea is respiratory radiograph. The need for continued cardiorespiratory monitoring,
compensation for metabolic acidosis as evidenced by hypocapnia parenteral nutrition and concerns for sepsis are the most common
and normoxemia. On the other hand, onset of respiratory disease reasons for continued neonatal observation and management. Within
would be evident by hypercapnia and hypoxemia. Worrisome about 30 min, age, and color changes should have stabilized.
tachypnea could be due to air leaks (pneumothoraces and Similarly, within about 6 h, age, any transient grunting, flaring and
pneumomediastinum), atelectasis, aspiration, pneumonia and, retractions should stabilize. Deviations from these time frames should
most of all, concern for incipient persistent pulmonary initiate a process of a more intense evaluation. The absence of any
hypertension. Peripheral cyanosis (manifested by blue toes) would signs of sepsis or cardiorespiratory distress beyond 24 h of age is likely
require continuous pulse oximetry to document normoxemia and to exclude the MAS.
observation for expectant care. Central cyanosis (as manifested by
blue lips) would require an evaluation for hemodynamic changes
with handling, evaluation of response to inspired oxygen and Conclusions
continued observation in a nursery environment for continued A systems approach that provides for a well-coordinated and trained
cardiorespiratory environment. Crystalloid infusion may be helpful ‘rapid response team’ for meconium-stained births is likely to
for infants with ‘benign tachypnea’ if the capillary perfusion is implement an evidence-based intervention and effectively manage

Journal of Perinatology
Developing a systems approach to prevent MAS
VK Bhutani
S35

a potential catastrophic outcome for the infant, the family and the 3 National Neonatal-Perinatal Database (NNPD). Morbidity and mortality among
staff. Classic practices of maintaining equipoise in the delivery outborn neonates at 10 tertiary care institutions in India during the year 2000. J Trop
Pediatr 2004; 50: 170–174.
room require working (and communicating) with the attending
4 Dooley SL, Pesavento DJ, Depp R, Socol ML, Tamura RK, Wiringa KS. Meconium below
obstetrician and nursing staff and supporting the education of the the vocal cords at delivery: correlation with intrapartum events. Am J Obstet Gynecol
family. The team’s approach is to be expectant but prepared to 1985; 153: 767–770.
intubate the infant. The team’s role is to monitor the progress of 5 Davis RO, Philips III JB, Harris Jr BA, Wilson ER, Huddleston JF. Fatal meconium
labor, fetal status, fetal data and family status and to be prepared to aspiration syndrome occurring despite airway management considered appropriate. Am
interact, interpret, intervene and initiate support as needed. These J Obstet Gynecol 1985; 151: 731–736.
6 Wiswell TE, Henley MA. Intratracheal suctioning, systemic infection, and the
data from a single US center also illustrate the complex
meconium aspiration syndrome. Pediatrics 1992; 89: 203–206.
interactions of technologies used during a birthing practice that 7 Chaturvedi P, Yadav B, Bharambe MS. Delivery room management of neonates born
could have both potentially useful (airway clearance) and/or through meconium stained amniotic fluid. Indian Pediatr 2000; 37: 1251–1255.
deleterious effects because of the combinations of trauma, 8 Mahomed K, Nyoni R, Masona D. Meconium staining of the liquor in a low-risk
introduction or aggravation of infection and/or interference of gas population. Paediatr Perinat Epidemiol 1994; 8: 292–300.
9 Ziadeh SM, Sunna E. Obstetric and perinatal outcome of pregnancies with term labour
exchange. Several of the components of clinical practice are based
and meconium-stained amniotic fluid. Arch Gynecol Obstet 2000; 264: 84–87.
on evidence, and their implementation in practice may need 10 Falciglia HS. Failure to prevent meconium aspiration syndrome. Obstet Gynecol 1988;
further validation or resolution by consensus. There is a health and 71: 349–353.
societal need to establish guidelines for practice at birthing 11 Yoder BA. Meconium-stained amniotic fluid and respiratory complications: impact of
facilities in the United States. Consistency in practice may lead to selective tracheal suction. Obstet Gynecol 1994; 83: 77–84.
cost containment, even though the cost-effectiveness of such an 12 Vidyasagar D, Harris V, Pildes RS. Assisted ventilation in infants with meconium
aspiration syndrome. Pediatrics 1975; 56: 208–213.
approach has not been validated. Methodological inquiry,
13 Rossi EM, Philipson EH, Williams TG, Kalhan SC. Meconium aspiration syndrome:
evidentiary analysis and reassessment of both clinical and intrapartum and neonatal attributes. Am J Obstet Gynecol 1989; 161: 1106–1110.
biotechnological options would better inform the development of a 14 Falciglia HS, Henderschott C, Potter P, Helmchen R. Does DeLee suction at the
practical stepwise algorithm to optimize bedside care. perineum prevent meconium aspiration syndrome? Am J Obstet Gynecol 1992; 167:
1243–1249.
15 Bhutani VK, Chima R, Sivieri EM. Innovative neonatal ventilation and meconium
aspiration syndrome. Indian J Pediatr 2003; 70: 421–427.
Disclosure
16 Carson BS, Losey RW, Bowes Jr WA, Simmons MA. Combined obstetric and pediatric
The author has declared no financial interests. approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126:
712–715.
17 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery room
management of the apparently vigorous meconium-stained neonate: results of the
References multicenter, international collaborative trial. Pediatrics 2000; 105: 1–7.
1 Usta IM, Mercer BM, Sibai BM. Predictive risk factors for meconium aspiration 18 Vain NE, Szyld EG, Prudent LM, Wiswell TE, Aguilar AM, Vivas NI. Oropharyngeal and
syndrome. Obstet Gynecol 1996; 86: 230–234. nasopharyngeal suctioning of meconium-stained neonates before delivery of their
2 International Liaison Committee on Resuscitation (ILCOR). Consensus on science with shoulders: multicentre, randomised controlled trial. Lancet 2004; 364: 597–602.
treatment recommendations for pediatric and neonatal patients: part 7Fneonatal 19 Lawn JE, Cousens S, Zupan J. 4 million neonatal deaths: when? Where? Why? Lancet
resuscitation. Pediatrics 2006; 117: e978–e988. 2005; 365: 891–900.

Journal of Perinatology
Journal of Perinatology (2008) 28, S36–S42
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Meconium aspiration syndrome requiring assisted ventilation:
perspective in a setting with limited resources
S Velaphi and A Van Kwawegen
Department of Paediatrics, Chris Hani Baragwanath Hospital, University of the Witwatersrand, Johannesburg, South Africa

et al.6 reported that MAS decreased nearly fourfold between 1990


To determine characteristics, management, complications and outcome of to1992 and 1997 to 1998, and this was associated with 33%
neonates with meconium aspiration syndrome (MAS) requiring mechanical reduction in births more than 41 weeks gestation, more frequent
ventilation (MV). A retrospective review of clinical data of neonates with diagnosis of nonreassuring fetal heart rate patterns and greater use
MAS who were admitted to a public hospital for MV between January 2004 of amnioinfusion. Approximately 50% of patients with MAS will have
and December 2006. Eighty-eight neonates were ventilated for MAS. Thirty- severe MAS defined as the need for mechanical ventilation (MV).11
one percent were postdates and 51% had no electronic fetal monitoring. The reports on the outcome of patients with MAS, especially those
Postnatal suctioning of meconium was not performed according to protocol who require MV, originate mainly from developed countries. Factors
in 47% of nonvigorous infants. High-frequency ventilation and surfactant such as poor monitoring during labor and low Apgar scores that are
were used in 32 and 14% of cases, respectively. Persistent pulmonary associated with the need for MV among infants with MAS11 occur
hypertension of the newborn (PPHN) and pneumothorax occurred in 57 commonly in developing countries; therefore, patients with MAS in
and 24% of cases, respectively. Overall mortality rate was 33%. Neonates developing countries might have worse outcomes compared with
suffering from MAS with PPHN had higher mortality rate of 48% compared those from developed countries. Secondly, the adjunct respiratory
with 13% in those suffering from MAS without PPHN. Factors associated therapies that have been associated with reduction in mortality are
with mortality were peak inspiratory pressure (P<0.001), pneumothorax not easily available in developing countries. The limitations of
(P<0.001) and PPHN (P ¼ 0.001). Postdates, inadequate intrapartum assessing the outcomes of MAS in developing countries are related to
monitoring and limited use of adjunct respiratory therapies were common. limited resources resulting in difficulties in making a diagnosis of
Severe MAS is associated with adverse outcome. MAS, which requires the presence of X-ray changes. In developing
Journal of Perinatology (2008) 28, S36–S42; doi:10.1038/jp.2008.155 countries, patients with possible MAS might die before a chest X-ray
(CXR) can be taken or might improve and be discharged without
having had a CXR unless they are admitted to the neonatal
intensive care unit (NICU) for MV. Therefore, patients who are
Introduction
admitted to NICU are more likely to have CXR. The aim of this
Meconium aspiration syndrome (MAS) is associated with significant study was to describe characteristics, management and outcome of
morbidity and mortality. Common complications associated with neonates with MAS in a setting where resources are limited. Owing
MAS include interstitial emphysema, pneumothorax, pneumonia to difficulties in reliably diagnosing MAS in patients who do not
and persistent pulmonary hypertension of the newborn (PPHN). require MV, this study was limited to those who required MV.
Pulmonary air leaks have been observed to occur in 15 to 33% of
cases of MAS.1,2 Although MAS is still associated with significant
mortality, mortality rates seem to have decreased over the years. Methods
Studies from 1970s and 1980s reported high mortality rates ranging Infants with severe MAS were identified by review of discharge
from 28 to 40%.3,4 Recent literature has reported mortality rates less diagnoses and hospital records of all patients who weighed
than 15%.1,2,5 This reduction in mortality rates appears to be related X2000 g and were admitted to the NICU of Chris Hani
(1) to reduction in incidence of MAS, which is related to changes in Baragwanath Hospital, Johannesburg, South Africa, between
obstetric practices6,7 and (2) to use of adjunct respiratory therapies, January 2004 and December 2006. Maternal and neonatal hospital
such as exogenous surfactant, nitric oxide and high-frequency charts were reviewed for confirmation of the diagnosis and
ventilation in the management of neonates with MAS.8–10 Yoder pertinent clinical data.
Severe MAS was diagnosed if infants met the three following
Correspondence: Dr S Velaphi, Department of Paediatrics, Chris Hani Baragwanath Hospital,
University of the Witwatersrand, PO Bertsham, Johannesburg 2013, South Africa. criteria: meconium-stained amniotic fluid (MSAF), need for MV
E-mail: velaphisc@medicine.wits.ac.za and radiographic abnormalities consistent with MAS (Figure 1).
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S37

Figure 1 Chest X-ray representative of X-rays that were selected toward making a diagnosis of meconium aspiration syndrome.

Patients were excluded from the study if there was no 60 min. PPHN was diagnosed either with a preductal versus
documentation of the presence of MSAF, if records were incomplete, postductal pulse-oximeter oxygen saturation gradient of >10% or
if CXR was not available from records and no comment was on echocardiography.
present on CXR findings in doctor’s notes or if the baby had Demographic characteristics, clinical data and neonatal
congenital abnormalities. outcomes to NICU discharge were recorded. Comparison between
During the study period, the protocol on postnatal management survivors and nonsurvivors was performed using w2 or Fisher exact
of infants born through MSAF was according to the guidelines of test for categorical variables and Student’s t-test for continuous
the International Liaison Committee on Resuscitation.12 The variables. Permission to conduct the study was received from the
guidelines stated that for all infants born through MSAF with University of the Witwatersrand Human Research Ethics
absent or depressed respiration, direct laryngoscopy should be Committee.
conducted immediately after birth for suctioning of residual
meconium from the hypopharynx and for intubation and
suctioning of the trachea. All patients who were admitted with Results
respiratory distress were started on antibiotics; patients who Among the 2157 infants who required MV from January 2004 to
required MV were started on synchronized intermittent mandatory December 2006, 800 weighed X2000 g. Of these 800 infants, 143
ventilation using a peak inspiratory pressure of 20 to 25 cm H2O, had a diagnosis of MAS or had an abnormal CXR with changes
positive end expiratory pressure of 4 to 5 cm H2O, inspiratory time suggestive of MAS and were admitted within 48 h of life. Among the
of 0.4 s and flow of 5 to 10 l min1. There were two models of 143 patients, 55 were excluded. The reasons for exclusion were
conventional ventilators used in the unit: the Bear Cub 750PSV absence of a history of MSAF, incomplete hospital records, and
infant ventilator and Newport E100M ventilator. The mode of therefore inability to confirm or exclude the presence of MSAF, and
control for the conventional ventilators was pressure limited and CXR not found or no comment in patient’s file regarding CXR
time cycled. There were two ventilators available for high-frequency findings (Figure 2). There were 88 patients who had complete
ventilation, both of them were Sensor Medics 3100A oscillatory records, with history of MSAF in mother’s or infant’s hospital file,
ventilators. All patients had CXRs ordered immediately changes on CXR and were ventilated; these were included in the
postintubation or after insertion of umbilical arterial/venous study. These 88 patients accounted for 4% of all admissions
catheters or when patient deteriorated. Sometimes, there were requiring MV and 11% of NICU admissions weighing X2000 g.
delays in having X-rays taken especially at night or over the There were 61 399 live births over this 3-year period. The incidence
weekends because of staff (radiographers) shortages. Availability of of severe MAS or of infants with MAS requiring MV was 1.43 per
surfactant was limited to 5 vials per month in 2004, and this was 1000 live births.
increased to 10 vials per month in 2005. Nitric oxide was made Characteristics of mothers to infants with MAS are shown in
available to the unit at the end of 2004. We are limited to use two Table 1. Ninety-eight percent of patients were born inside a
tanks of nitric oxide per year, with each tank of 10 l containing 900 healthcare facility and only 2% were born at home. The maternal
parts per million of nitric oxide. Both the gas and the delivery and HIV status was positive in 31% of cases, which was similar to the
monitoring system were bought from Sidewinder Medical, Cape institutional statistics from antenatal clinic. Thirty-one percent
Town, South Africa. It was only used in patients who had hypoxia were born at X41 weeks gestation. Electronic monitoring was
thought to be secondary to persistent pulmonary hypertension of documented in 49% of cases, of which 78% had abnormal tracing
the newborn, and it was stopped if there was no response in and the common abnormality was late decelerations. Forty-four

Journal of Perinatology
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S38

Total number of live births from January 2004 to December 2006 Table 1 Maternal characteristics of infants with MAS requiring ventilation
(n= 61399)
Number (%)

Maternal age
<18 years 4 (5)
Total neonatal intensive care unit (NICU) admissions
(n = 2157) 18–35 years 68 (77)
>35 years 13 (15)
Not recorded 3 (3)

NICU admissions with birth weight ≥2kg Parity


(n = 800) 0 40 (45)
1–3 37 (42)
>3 6 (7)
Not recorded 5 (6)
Birth weight .2kg + Diagnosis of MAS and/ or chest X-ray (CXR)
suggestive of meconium aspiration syndrome (MAS) Human immunodeficiency virus
(n = 143)
Positive 27 (31)
Negative 50 (57)
Unknown 11 (12)
MSAF could not be MSAF MSAF
excluded or confirmed present absent
(n=12) (n=101) (n =30) Antenatal care
Yes 74 (84)
No 14 (16)

Monitoring with CTG


CXR available and No CXR or no record of
Yes, with abnormalities 33 (38)
abnormal X-ray findings
(n=88) (n =13) Yes, with no abnormalities 10 (11)
No 45 (51)
Figure 2 Number of live births, patients requiring mechanical ventilation and
those excluded or included in the study. Gestational age by dates
<35 weeks 2 (2)
percent were born by cesarian section, and the indication for the 35–37 weeks 6 (7)
cesarian section was fetal distress in 90% of cases. Although 38–40 weeks 39 (44)
consistency of MSAF was not recorded in 44% of patients, the X41 weeks 27 (31)
majority of them were born through thick meconium. Not recorded 14 (16)
Infant details are shown in Table 2. There were 38 (43%)
Place of birth
patients who required resuscitation, with almost all of them
Inborn 75 (85)
responding to bag mask ventilation. The number of patients who
Referrals from other hospitals 5 (6)
required resuscitation was greater than those with low Apgar score Clinic 6 (7)
(score p5) at 1 min, suggesting over-reading of Apgar scores or Home 2 (2)
reflecting our practice of withholding assisted ventilation from
infants with the lowest Apgar scores because of concerns about poor Mode of delivery
neurodevelopmental outcome. Among those who required Normal vaginal delivery 43 (49)
resuscitation, only 53% were recorded as having been intubated for Cesarian section 39 (44)
suctioning of meconium. Severity of the lung disease was moderate Vacuum 5 (6)
to severe in 64% of patients as defined by an oxygen index of more Breech 1 (1)
than 10. Twenty-three patients out of the 28 who did not have
Consistency of MSAF
C-reactive protein (CRP) results are those who died on day 1 of life
Thick 47 (53)
before CRP level could be done. Among those who had CRP results,
Thin 6 (7)
92% had CRP levels more that 10 mg l1.
Unrecorded 35 (40)
Morbidity and mortality of infants with MAS are shown in
Table 3. Common complications among patients with MAS were
PPHN (57%) and pneumothorax (24%). Of the 50 patients who

Journal of Perinatology
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S39

Table 2 Characteristics of infants with MAS requiring mechanical infants Table 3 Complications, duration of stay in NICU and outcome of patients with
MAS requiring mechanical ventilation (n ¼ 88)
Number (%)
Number (%)
Birth weight (g) 3080 (2100–4330)a
Complications
Gender Pneumothorax 21 (24)
Female 45 (51) PPHN 50 (57)
Male 43 (49)
Median duration of stay in NICU (days) 4 (1–31)a
1 min Apgar score 6 (1–9) a p3 days 33 (38)
<5 19 (21) 4–7 days 35 (40)
X5 64 (73) 8–14 days 10 (11)
Not recorded 5 (6) >14 days 10 (11)

5 min Apgar score 8 (3–10)a Outcome among all patients (n ¼ 88)


<7 21 (24) Died 29 (33)
X7 54 (61) Survived 59 (67)
Not recorded 13 (15)
Outcome among those without PPHN (n ¼ 38)
Required resuscitation Died 5 (13)
No 50 (57) Survived 33 (87)
Yes 38 (43)
Bag mask ventilation 38 (43) Outcome among those with PPHN (n ¼ 50)
Bag mask ventilation+chest compression 1 (1) Died 24 (48)
Above+adrenalin 0 Survived 26 (52)

Intubated and suctioned for meconium 20/38 (53) Abbreviations: MAS, meconium aspiration syndrome; NICU, neonatal intensive care unit;
PPHN, persistent pulmonary hypertension of the newborn.
a
Median (range).
Median oxygen index (OI) (range) 15.17 (0.97–71.16)a
Number of patients with OI <10 32 (36)
Number of patients with OI between 10 and 20 42 (48)
Number of patients with OI >20 14 (16)
The use of exogenous surfactant, number of patients who were
C-reactive protein (CRP) (range) 52.9 (1–567) a put on high-frequency oscillatory ventilation and nitric oxide were
Number of patients with CRP<10 mg l1 5 (6) low during the study period, and it did not change significantly
Number of patients with CRP between 10 and 20 mg l1 8 (9) over the years (P-values all more than 0.100) (Table 5). Incidence
Number of patients with CRP >20 mg l1 47 (53) of pneumothorax, PPHN and mortality rate did not change
Number of patients with unrecorded CRP 28 (32) significantly over the years.
Abbreviations: CRP, C-reactive protein; MAS, meconium aspiration syndrome; OI, oxygen index.
a
Median (range).
Discussion
Severe MAS is defined as those cases that require assisted MV.13 It
has been reported that between 40 and 50% of infants with MAS
had PPHN, 47 were diagnosed by differential pulse oximetry will need MV.11,14,15 In public hospitals of developing countries,
saturations and only 17 (36%) had echocardiography, which offering MV is often restricted to those who are expected to have
confirmed the diagnosis of PPHN, and the other 30 did not have favorable outcomes, and among those who qualify, it is often
echocardiography because of limited human resources. Three were delayed due to limited bed availability in NICUs. Thus, patients
diagnosed only on echocardiography. Median duration of stay in who are ventilated in countries with limited resources are more
NICU was 4 days, with 22% of them staying for more than 7 days. likely to have poor outcome because of delays in instituting
One-third of patients with MAS did not survive. Factors associated treatment, limited adjunct respiratory therapies and high incidence
with mortality were female gender (P ¼ 0.046), maximum peak of having associated conditions such as infections. In this study, we
inspiratory pressure (P<0.001), development of pneumothorax sought to determine characteristics and outcome of infants who
(P<0.001) and PPHN (P ¼ 0.001) (Table 4). Most of the deaths were ventilated with MAS, in a setting where there are limited
occurred early during their NICU stay. resources.

Journal of Perinatology
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S40

Table 4 Comparison between survivors and nonsurvivors

Nonsurvivors (n ¼ 29) Survivors (n ¼ 59) P-value

Characteristics
Mean birth weight 2976±546 3128±429 0.157
Gestational age 40 (32–43) 40 (35–43) 0.843
Apgar score at 1 min 6 (1–9) 7 (2–9) 0.582
Apgar score at 5 min 8 (3–10) 8 (4–10) 0.360
Male:female 1:2.1 1:0.74 0.046
Mother human immunodeficiency virus positive 21% 36% 0.238
Requiring resuscitation at birth 38% 46% 0.640
Intubated for suctioning at birth 10/11 (91%) 16/27 (59%) 0.121
Peak pressure required on admissiona,b 26 (25–29) 25 (25–29) 0.070
OI on admission 15.6 (8.1–21.7) 12.2 (7.6–18.5) 0.183

Patients at different levels of OIs at admission 0.413


Number of patients with OI <10 8 (28%) 24 (41%)
Number of patients with OI 10–20 11 (38%) 21 (35%)
Number of patients with OI >20 10 (34%) 14 (24%)

Median maximum peak pressurea,b 32 (30–35) 28 (25–32) <0.001

Complications
Pneumothorax 15 (52%) 6 (10%) <0.001
PPHN 24 (83%) 26 (44%) 0.001
Duration of stay in NICU 2 (1–12) 10 (2–25) <0.001
Abbreviations: NICU, neonatal intensive care unit; OI, oxygen index; PPHN, persistent pulmonary hypertension of the newborn.
a
Peak pressure is in cm H2O.
b
Median (25th to 75th percentile).

Table 5 Use of respiratory adjunct therapies, complications and mortality rate over the study period

Year 2004 (n ¼ 34) Year 2005 (n ¼ 22) Year 2006 (n ¼ 32) All

Number of patients who received surfactant 2 (6%) 4 (18%) 6 (19%) 12 (14%)


Number of patients who were put on high-frequency oscillation ventilator 7 (21%) 10 (45%) 11 (34%) 28 (32%)
Number of patients who were put on nitric oxide 2 (6%) 1 (3%) 2 (6%) 5 (6%)
Number of patients with PPHN 22 (64%) 15 (68%) 13 (41%) 50 (57%)
Number of patients with pneumothorax 10 (29%) 4 (18%) 7 (22%) 21 (24%)
Number of deaths 14 (41%) 5 (23%) 10 (31%) 29 (33%)
Abbreviation: PPHN, persistent pulmonary hypertension of the newborn.

About one-third of patients with severe MAS were born at 41 monitoring during pregnancy and labor on MAS is supported by
weeks gestation or more; and 51% of patients were born to mothers reduction of MAS in institutions where postmaturity has been
who did not have electronic monitoring during labor despite reduced and monitoring is intensive.6
having a history of MSAF. Postmaturity may have contributed to The protocol of intubating and suctioning infants suffering
these patients developing MAS. The risk of MAS has been shown to from absent or depressed respiration was not followed consistently,
be higher in post-term than term infants with relative risk of 3.1 as only 53% of infants who should have been intubated according
with 95% confidence interval of 2.6 to 3.7.16 Inadequate to the protocol were actually intubated. The possible reasons for
monitoring might have led to delays in expediting delivery of this low rate of suctioning could be poor documentation of what
infants who were compromised, and this allowed for ongoing happens during resuscitation, or that junior doctors who
hypoxia resulting in severe disease. The effect of postmaturity and commonly conduct neonatal resuscitation in our hospital were not

Journal of Perinatology
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S41

confident enough to perform intubations and therefore not monitoring during labor and offering appropriate neonatal care
adhering to protocol. Inadequate suctioning of meconium could including exogenous surfactant will improve incidence of MAS and
have contributed to the development of severe MAS, as it has been those requiring ventilation, and therefore less strain on already
reported that meconium in the trachea is one of the major risk limited resources.
factors associated with diagnosis of MAS.11,17 –20 It is also possible
that this did not affect the development of severe MAS, as some
studies have suggested that aspiration of meconium occurs Disclosure
in utero and that damage in the lungs has already taken place by The authors have declared no financial interests.
the time the baby is born.1,19,21–23
The high mortality rate of 33% in this study could be related to
associated conditions, such as infections and PPHN, and low usage References
of adjunct respiratory therapies. Among the patients who had their 1 Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a
CRP level assessed, 92% had CRP >10 mg l1 and 78% had CRP difference? Pediatrics 1990; 85: 715–721.
>20 mg l1, suggesting the presence of infection or inflammation. 2 Chinese collaborative study group for neonatal respiratory diseases. Treatment of severe
meconium aspiration syndrome with porcine surfactant: a multicentre, randomized,
In vitro studies have suggested that meconium may activate
controlled trial. Acta Paediatr 2005; 94: 896–902.
alveolar macrophages with subsequent release of inflammatory 3 Carson BS, Losey RW, Bowes WA, Simmons MA. Combined obstetric and pediatric
cytokines resulting in an increase in CRP.24 Although only one approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126:
patient had positive culture, it is difficult to exclude infection when 712–715.
there is abnormal CXR and high CRP. Some studies have reported 4 Davis RO, Philips JB, Harris BA, Wilson ER, Huddelston JF. Fetal meconium aspiration
that clinical chorioamnionitis and neonatal sepsis are more syndrome occurring despite airway management considered appropriate. Am J Obstet
Gynecol 1985; 151: 731–736.
common in neonates with severe MAS.11,25,26 More than 50% of 5 Dargaville PA, Copnell B, for the Australian and New Zealand Neonatal Network. The
patients in this study had PPHN. Should one exclude or include epidemiology of meconium aspiration syndrome: incidence, risk factors, therapies and
infants with PPHN when one is looking at outcome of infants with outcome. Pediatrics 2006; 117: 1712–1721.
MAS? The literature suggests that these patients should be excluded. 6 Yoder BA, Kirsch EA, Barth Jr WH, Gordon MC. Changing obstetric practices associated
The reason for this is that the cause of PPHN in patients with MAS with decreasing incidence of meconium aspiration syndrome. Obstet Gynecol 2002;
99: 731–739.
might have preceded the aspiration of meconium. This is supported
7 Mahomed K, Mulambo T, Woelk G, Hofmeyr J, Gulmezoglu AM. The collaborative
by findings in autopsies from cases dying from severe MAS within randomized amnioinfusion for meconium project (CRAMP): 2. Zimbabwe. Br J Obstet
48 h of birth, which revealed muscularization of distal pulmonary Gynaecol 1998; 105: 309–313.
arterioles, which requires 3 to 8 days to develop, making it unlikely 8 Greenough A, Pulikot A, Dimitriou G. Prevention and management of meconium
that PPHN was a complication of MAS.23,27,28 As PPHN on its own aspiration syndromeFassessment of evidence based practice. Eur J Pediatr 2005;
is associated with high mortality rate, one would expect less 164: 329–330.
9 Finer NN, Barrington KJ. Nitric oxide for respiratory failure in infants born at or near
mortality rate from MAS if infants with PPHN are excluded. In this term. Cochrane Database Syst Rev 2006; 4: CD000399.
study, the mortality rate was 73% lower in those with PPHN 10 El Shahed AI, Dargaville P, Ohlsson A, Soll RF. Surfactant for meconium aspiration
compared to those without PPHN. The low mortality rate in syndrome in full term/near term infants. Cochrane Database Syst Rev 2007; 3:
developed countries has been associated with an increase in the use CD002054.
of exogenous surfactant, high-frequency ventilation and inhaled 11 Hernandez C, Little BB, Dax JS, Gilstrap LC, Rosenfeld CR. Prediction of the severity of
meconium aspiration syndrome. Am J Obstet Gynecol 1993; 169: 61–70.
nitric oxide, with >50% of infants receiving one or more of these
12 Kattwinkel J, Niermeyer S, Nadkarni V, Tibballs J, Phillips B, Zideman D et al. An
therapies.5 A postal survey of 227 neonatal units in the United Advisory Statement from the Pediatric Working Group of the International Liaison
Kingdom revealed that 96, 42 and 29% of units were using Committee on Resuscitation. Pediatrics 1999; 103: e56.
exogenous surfactant, high-frequency ventilation and inhaled 13 Wiswell TE, Bent RC. Meconium staining and the meconium aspiration syndrome.
nitric oxide for MAS, respectively.8 In this study, use of exogenous Pediatr Clin North Am 1993; 40: 955–981.
surfactant, high-frequency ventilation and inhaled nitric oxide 14 Liu WF, Harrington T. Delivery room risk factors for meconium aspiration syndrome.
Am J Perinatol 2002; 19: 367–378.
ranged from 6 to 19, 21 to 45 and 3 to 6%, respectively, over 15 Adhikari M, Gouws E, Velaphi SC, Gwamanda P. Meconium aspiration syndrome:
the 3 years. importance of the monitoring of labor. J Perinatol 1998; 18: 55–60.
In conclusion, patients who were ventilated for MAS were more 16 Clausson B, Cnattingius S, Axelsson O. outcomes of post-term births: the
likely to have been born postdates, to have not been monitored role of fetal growth restriction and malformations. Obstet Gynecol 1999; 94:
electronically during labor, to have been born by cesarean section 758–762.
17 Yoder B. Meconium-stained amniotic fluid and respiratory complications: impact of
and to have not been intubated for suctioning of meconium at
selective tracheal suction. Obstet Gynecol 1994; 83: 77–84.
birth. MAS requiring MV is still associated with significant 18 Dooley S, Pesavanto D, Depp R, Socol M, Tamura R, Wiringa K. Meconium below the
morbidity and mortality in the setting where there are limited vocal cords at delivery: correlation with intrapartum events. Am J Obstet Gynecol 1985;
resources. Reducing patients who deliver post-term, intensive 153: 767–770.

Journal of Perinatology
Meconium aspiration syndrome and ventilation
S Velaphi and A Van Kwawegen
S42

19 Rossi E, Philipson E, Williams T, Kalhan S. Meconium aspiration syndrome: 24 Kojima T, Hattori K, Fujiwara T, Sasai-Takedatsu M, Kobayashi Y. Meconium-induced
intrapartum neonatal attributes. Am J Obstet Gynecol 1989; 74: 715–721. lung injury mediated by activation of alveolar macrophages. Life Sci 1994; 54:
20 Usta I, Mercer B, Sibai B. Risk factors for meconium aspiration syndrome. Obstet 1559–1562.
Gynecol 1995; 86: 230–240. 25 Coughtrey H, Jeffery HE, Henderson Smart DJ, Storey B, Poulos V. Possible causes
21 Flaciglia HS. Failure to prevent meconium aspiration syndrome. Obstet Gynecol 1988; linking asphyxia, thick meconium, and respiratory distress. Aust N Z J Obstet Gynaecol
71: 349–353. 1991; 31: 97–102.
22 Cornish JD, Dreyer GL, Snyder GE, Kuehl TJ, Gerstmann DR, Null DM et al. Failure of 26 Wiswell TE, Henley MA. Intratracheal suctioning, systemic infection, and the
acute perinatal asphyxia or meconium aspiration to produce persistent pulmonary meconium aspiration syndrome. Pediatrics 1992; 89: 203–206.
hypertension in a neonatal model. Am J Obstet Gynecol 1994; 171: 43–49. 27 Perlman EJ, Moore GW, Hutchins GM. The pulmonary vasculature in meconium
23 Thureen P, Hall DM, Hoffenberg A, Tyson RM. Fatal meconium aspiration in spite of vasculature in meconium aspiration. Hum Pathol 1989; 20: 701–706.
appropriate perinatal airway management: pulmonary and placental evidence of 28 Murphy ID, Vawter GF, Reid LM. Pulmonary vascular disease in fatal meconium
prenatal disease. Am J Obstet Gynecol 1997; 176: 967–975. aspiration. J Pediatr 1984; 104: 158–162.

Journal of Perinatology
Journal of Perinatology (2008) 28, S43–S48
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Meconium aspiration syndrome: experiences in Taiwan
HC Lin1,2, SY Wu3, JM Wu4 and TF Yeh1,5
1
Department of Pediatrics, China Medical University Hospital, Taichung, Taiwan, ROC; 2School of Chinese Medicine, China Medical
University, Taichung, Taiwan; 3Department of Pediatrics, College of Medicine, National Cheng Kung University, Tainan, Taiwan;
4
Department of Pediatrics, John Stroger Hospital of Cook County, Chicago, IL, USA and 5Department of Pediatrics, College of Medicine,
China Medical University Hospital, Taichung, Taiwan, ROC

Incidence of MAS
Meconium aspiration syndrome (MAS) is still one of the most challenged
Table 2 compares the incidence and mortality of infants
diseases for the neonatologists. We reviewed our earlier studies of MAS in an
suffering from MAS between the United States and one of the
attempt to provide some idea for more understanding of MAS. This study is
tertiary centers in Taiwan (China medical university hospital,
a retrospective review and summarization of our earlier studies in MAS at
Taichung, Taiwan).1–3 It is interesting to note that data of the
two tertiary neonatal centers in Taiwan. Incidence of MAS was decreased
incidences of meconium staining of amniotic fluid, MAS
sharply in Taiwan. MAS infants who required resuscitation in the birth
and mortality are almost comparable with those from the
room being out-born, birth asphyxia and infants who developed persistent
United States. However, these data are obtained from one
pulmonary hypertension (PPHN) and pneumothrax were associated with
tertiary center with many high-risk maternal transfers; the
increasing mortality. In MAS infants who neither required mechanical
actual overall incidence in Taiwan could be much lower. The
ventilation nor had a history suggestive of perinatal infection, antibiotic
outborn infants have higher mortality rates (7.3%) than inborn
treatments would not affect the outcome of MAS. Dexamethasone did reduce
infants (0.81%) (Table 3).
inflammation response and improve cardiopulmonary perfusion. However,
steroids did not prevent the development of PPHN. Our review provided the
risk factors of mortality for MAS. Antibiotic treatments should not be a
Perinatal and postnatal factors and mortality in MAS
routine for every infant with MAS. Although steroids reduce pulmonary
inflammation, their role in the prevention of PPHN remains to be A review of 314 cases of MAS between 1995 and 20011,5 indicated
further studied. that infants who required resuscitation in the birth room, who
Journal of Perinatology (2008) 28, S43–S48. doi:10.1038/jp.2008.157 were first out-born or with birth asphyxia, and infants who
developed persistent pulmonary hypertension (PPHN) and
pneumothorax are important factors associated with increased
mortality in MAS (Table 4).

The incidence of meconium aspiration syndrome (MAS) varies


from population to population and from countries to countries.1–4 Management of meconium-stained amniotic fluid in
With the recent improvement in socioeconomic status and the delivery room
advances in prenatal care in Taiwan, the incidence of MAS in Since 1995, the management of meconium-stained amniotic
Taiwan sharply decreased in the past decade.1 The adoption of fluid has followed a guideline as shown in Figure 1.6 This
National Healthcare Insurance System is probably the most guideline is not much different from the recent recommendations
important factor leading to better health access and care. Table 1 of American Academy of Pediatrics.7,8 We do endotracheal
shows some of the factors that characterized health care in Taiwan. suction in infants who have thick meconium staining,
At present, nearly 100% of the pregnant women have prenatal care who have low Apgar score or who have respiratory distress shortly
and majority of the deliveries are carried out in large hospitals. after birth.9 We perform intrapartum oro- and nasopharyngeal
However, the mean age of the mother at the time of first childbirth suction in all infants if there is meconium staining of amniotic
is about 26 years, older than that in most of the developing and fluid.6,10 We feel that this procedure is simple and does not
developed countries. carry significant adverse effects. Using this guideline, about
50% of meconium-stained infants required endotracheal suction.
Correspondence: Dr TF Yeh, Department of Pediatrics, College of Medicine, China Medical
University, 91 Hsueh Shih Rd., Taichung, Taiwan, ROC. We do amnioinfusion only if there is oligohydramnio and
E-mail: tfyeh@mail.ncku.edu.tw thick meconium staining.11–15
Meconium aspiration syndrome
HC Lin et al
S44

The role of antibiotics in MAS one received antibiotics and the other did not. Both groups were
Meconium has been shown to enhance bacterial growth comparable in their baseline data. There was no significant
in vitro, particularly Gram-negative organisms. It is a difference between the groups in clinical course and mortality
common practice to place these infants on antibiotics. We have (Table 5).
conducted a study on 259 MAS infants who did not require Thus, antibiotic treatments would not affect the clinical course
mechanical ventilation and who did not have a history suggesting and outcome in MAS without perinatal risk factors for infection
perinatal infection.5 The infants were divided into two groups; and without ventilator use.

The role of pulmonary inflammation in the development


Table 1
of PPHN in MAS
Health care in Taiwan Pulmonary hypertension or PPHN of the newborn occurs
NHIS coverage in 10 to 15% of infants with MAS. This condition usually
Prenatal care 98–100% presents as persistent hypoxemia occurred at 6 to 24 h after
Average age of mother for first childF26 years birth. A spontaneous recovery usually occurs within 3 to
Uniform population and easy access for medical care 4 days if the patient survives, suggesting that a functional
Establishment of regionalization of perinatal care vascular constriction is probably involved in the
Minimal years of maternal education 9–12 years pathogenesis.
Average number of children per familyF1.2 Pulmonary hypertension or PPHN in infants with MAS
Abbreviation: NHIS, National Healthcare Insurance System. could be due to (1) hypertrophy or neo-muscularization of

Table 2 Incidences of MAS and mortality in the United States and Taiwan Table 4 Risk factors of mortality in MAS by logistic regression model
2 1
USA (Wiswell et al. ) Taiwan (Lin et al. ) Variable P-value Odds ratio CI lower CI upper
1997 1995–2001
Asphyxia 0.0032 0.026 0.002 0.293
Incidence of meconium staining 10–15% 13% Outborn 0.0869 0.124 0.011 1.353
MAS/meconium staining 5% 5.9% PPHN 0.0145 0.065 0.007 0.581
IMV/MAS 33% (inborn 0.81%) Shock 0.4246 2.50 0.263 23.771
Incidence of MAS/liveborn 0.5% 0.7% Pneumothorax 0.0143 0.158 0.036 0.692
Mortality/MAS 5% 4.8%
Abbreviations: MAS, meconium aspiration syndrome; PPHN, persistent pulmonary
Abbreviations: IMV, intermittent mandatory ventilation; MAS, meconium aspiration hypertension of the newborn.
syndrome. Adapted from Lin et al.1

Table 3 Incidence and mortality rate of MAS from 1995 to 2001

1995 1996 1997 1998 1999 2000 2001 Total

Inborn babies (n) 14 22 14 12 16 21 24 123


Outborn babies (n) 26 18 19 19 31 43 35 191
Survival (n) 36 39 32 29 43 63 57 299
Expired (n) 4 1 1 2 4 1 2 15
Mortality rate (%) 4/40 1/40 1/33 2/31 4/47 1/64 2/59 15/314
(10.0) (2.5) (3.0) (6.5) (8.5) (1.6) (3.4) (4.8)
Incidence of MAS in inborn (%) 14/154 22/24 14/23 12/216 16/24 21/26 24/23 123/15
9 59 72 3 06 58 60 967
(0.90) (0.89) (0.59) (0.55) (0.66) (0.79) (1.01) (0.77)
Incidence of mortality in inborn (%) 1/14 0/22 0/14 0/12 0/16 0/21 0/24 1/123
(7.14) (0) (0) (0) (0) (0) (0) (0.81)
Abbreviation: MAS, meconium aspiration syndrome.
Adapted from Lin et al.1

Journal of Perinatology
Meconium aspiration syndrome
HC Lin et al
S45

postacinar pulmonary capillaries as a result of chronic intrauterine of this study, we made the following conclusions:
hypoxia, (2) functional pulmonary vasoconstriction as a result (1) Following meconium aspiration, elevation of pulmonary
of hypoxemia, hypercarbia or acidosis or (3) functional pulmonary arterial pressure appears to be biphasic, the early phase starting
vasoconstriction as a result of lung inflammation. We
hypothesized that lung inflammation may play an important
role in pulmonary vascular constriction and hypertension. We 35 Newborn piglets
therefore conducted a MAS study on newborn piglet models.16 (1–7 days)

The purposes of the study were to investigate the following:


(1) Does MAS in newborn piglets produce pulmonary hypertension? 7
15
13

(2) Does pulmonary inflammation play a role in the development Saline control MAS MAS +dexamethasone
of pulmonary hypertension. (3) Can anti-inflammatory agents, IMV, FIO2 0.3, RR 20/min, PIP/PEEP 15-20/4-5 cm H2O
Pulmonary artery catheter-PAP, PF
such as dexamethasone, prevent pulmonary hypertension. Figure 2 Femoral artery catheter–BP
3 ml kg -1 20% meconium mixed with saline
shows the method of the study on 35 newborn piglets Dexamethasone 0.5 mg kg -1 q 12h for 2 doses
(aged 1 to 7 days).16 We also administered dexamethasone early Tracheal aspirate daily for LTB4, LTC4, LTD4, thromboxane B2, 6-keto PGF-1α

in several infants.17 The results of piglet and newborn studies Figure 2 Method for Newborn Piglet MAS study. Adapted Wu et al.16
are shown in Tables 6 and 7. On the basis of the results

Table 6 Cardiac SV

MECONIUM STAINED A. F. Before 0–12 h 12–24 h d-2 d-3 d-4


SUCTION of Oro and NASOPHARYNX
S.V. (mg kg1)
PRIOR TO DELIVERY OF THORAX
C 1.5±0.4 1.4±0.5 1.5±0.4 1.6±0.5 1.6±0.4 1.9±0.8
MAS 1.4 0.4 1.2 0.3 1.1 0.2** 1.3±0.4* 1.6±0.3 1.8±0.4
± ± ±
MAS+D 1.4±0.5 1.3±0.4 1.3±0.3 1.4±0.3 1.5±0.5 1.9±0.3
THICK THIN
MECONIUM MECONIUM
mBP (mm Hg)
C 118±16 107±11 99±7 96±8 87±9 92±10
MAS 120±11 118±11 110±16 106 14** 103 8* 102±7
± ±
E. T. SUCTION GOOD APGAR LOW APGAR
MAS+D 119±17 120±12 124±15* 120±10** 118±8* 101±6
7-10

E. T. SUCTION
mPAP (cm H2O)
C 19±6 19±5 20±5 20±8 21±6 22±9
MAS 20±3 24±5* 23±5* 25±4* 24±7 22±6
NO RESP. DIST IN RESP. DIST MAS+D 20±6 27±10* 24±6* 24±8* 26±10 23±4
Abbreviations: MAS, meconium aspiration syndrome; MAS+D, meconium aspiration
OBSERVE E. T. syndrome+dexamethasone; mBP, mean blood pressure; mPAP, mean pulmonary arterial
SUCTION pressure; SV, stroke volume.
*P<0.05, **P<0.01 (as compared with saline control).
Figure 1 A guideline for the management of meconium staining of amniotic C: saline control, n ¼ 7; MAS, n ¼ 15; MAS+D, n ¼ 13.
fluid in the delivery room. Adapted from Wu et al.16

Table 5 MAS treated with or without antibiotics

No antibiotics (n ¼ 127) Antibiotics group (n ¼ 132) P-value

Tachypnea (days) 5.9±2.7 6.2±3.5 0.91


Duration of O2 (days) 4.6±1.7 3.6±1.3 0.46
Duration of CPAP (days) 2.2±1.8 3.0±2.2 0.65
Nasal CPAP (case) 29/127 33/132 0.68
Pulmonary air leaks (case) 4/127 7/132 0.39
Abbreviations: CPAP, continuous positive airway pressure; MAS, meconium aspiration syndrome.
Mean values±s.d.
Adapted from Lin et al.5

Journal of Perinatology
Meconium aspiration syndrome
HC Lin et al
S46

Table 7 Respiratory status in MAS infants with and without dexamethasone therapy

Before d-1 d-2 d-3 d-4 d-5 d-7

IMV
C 9 6 5 5 4 3 2
D 12 6 4 1 0 0 0

FIO2
C 0.54±0.31 0.35±0.16 0.26±0.16 0.29±0.16 0.29±0.08 0.29±0.09 0.28±0.18
D 0.52±0.24 0.40±0.20 0.30±0.18 0.32±0.29 0.30±0.22 0.31±0.24 0.30±0.21

PO2 (mm Hg)


C 86±58 84±33 74±22 75±30 70±26 80±18 75±25
D 91±80 86±42 70±42 78±28 67±23 76±14 74±14

PCO2 (mm Hg)


C 43±12 36±9 36±8 39±5 43±8 37±8 41±2
D 42±12 34±7 32±5* 33±9* 30±7** 35±7 39±5

pH
C 7.28±0.12 7.41±0.07 7.40±0.05 7.40±0.07 7.39±0.09 7.39±0.06 7.45±0.17
D 7.30±0.70 7.39±0.05 7.40±0.05 7.45±0.03* 7.43±0.04* 7.40±0.06 7.44±0.05
Abbreviations: IMV, intermittent mandatory ventilation; MAS, meconium aspiration syndrome.
*P<0.05, **P<0.01.
C: control infants, n ¼ 23; D: dexamethasone-treated infants, n ¼ 27. Adapted from Yeh et al.17

Figure 3 Mean blood pressure during the study. MAS piglets that received dexamethasone had significantly higher mean blood pressure at 4, 16, 36, 72 and 82 h than
saline control piglets. *P<0.05. Adapted from Wu et al.16

from 2 to 6 h and the later phase starting from 48 h. volume, increased pulmonary blood flow and improved
(2) A good correlation was seen between tracheal aspirate TXB2, ventilation/perfusion match.
leukotriene D4 and mean pulmonary anterial pressure. (3) The The role of pulmonary inflammation in the development of
use of dexamethasone reduced tracheal aspirate TXB2 and pulmonary hypertension or PPHN in MAS and the possible role of steroids
6-keto PGF1a. (4) Dexamethasone increased cardiac stroke in the prevention of PPHN remain to be further studied (Figures 3 to 7).

Journal of Perinatology
Meconium aspiration syndrome
HC Lin et al
S47

Figure 4 Mean pulmonary arterial pressure (PAP) changes during the study. Piglets with MAS and dexamethasone had significantly higher mean PAP than saline
control piglets at 4, 24, 36, 48 and 60 h after study. There were simultaneous increases in cardiac stroke volume, suggesting that the increases of mean PAP is
a result of increased pulmonary flow. *P<0.05. Adapted from Wu et al.16

Figure 5 Changes of 6-keto prostaglandin (PGF)1a in tracheal aspirate. MAS piglets


had significantly higher 6-keto PGF1a than saline controls. The use of
dexamethasone significantly decreased 6-keto PGF1a. Adapted from Wu et al.16 Figure 7 Changes of leukotriene (LTE4) in tracheal aspirate during the study.
Piglets with MAS had significantly higher LTE4 than saline controls at 24 h. The
use of dexamethasone significantly decreased LTE4 at 48, 72 and 92 h. Adapted
from Wu et al.16

Disclosure
The authors have declared no financial interests.

References
1 Lin HC, Su BH, Lin TW, Peng CT, Tsai CH. Risk factors of mortality in meconium
aspiration syndrome. Acta Pediatr TW 2004; 45: 30–34.
2 Wiswell T, Henley MA. Intratracheal suctioning, systemic infection, and the meconium
aspiration syndrome. Pediatrics 1992; 89: 203–206.
3 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery
Figure 6 Changes of leukotriene (LTC4) in tracheal aspirate during the study. Piglets room management of the apparently vigorous meconium-stained neonate:
with MAS had significantly higher LTC4 than saline controls at 24 h. The use of results of the multicenter, international collaborative trial. Pediatrics 2000; 105:
dexamethasone significantly decreased LTC4 at 48 and 72 h. Adapted from Wu et al.16 1–7.

Journal of Perinatology
Meconium aspiration syndrome
HC Lin et al
S48

4 Gregory GA, Gooding CA, Phibbs RH, Tooley WH. Meconium aspiration in infantsFa 12 Spong CY, Ogundipe OA, Ross MG. Prophylactic amnioinfusion for meconium stained
prospective study. J Pediatr 1974; 85: 848–852. amniotic fluid. Am J Obstet Gynecol 1994; 171: 931–935.
5 Lin HC, Su BH, Tsai CH, Lin TW, Yeh TF. Role of antibiotics in management of non- 13 Sadovsky Y, Amon E, Bade ME, Petrie RH. Prophylactic amnioinfusion during labor
ventilated cases of meconium aspiration syndrome without risk factors for infection. complicated by meconium: a preliminary report. Am J Obstet Gynecol 1989; 161:
Biol Neonate 2005; 87: 51–55. 613–617.
6 Yeh TF. Resuscitation in ‘Neonatal Therapeutics’, Yeh TF (ed), 2nd edn, Mosby-Year 14 Hourdequin P, Kauffmann E, Gabriel R, Jotterand AD, Chatelet-Cheront C, Quéreux C
Book: St Louis, 1992, pp 170–179. et al. Amnioinfusion during labour: experience and review of the literature. Contracept
7 Kattwinkel J, Niermeyer S, Nadkarni V, Tibballs J, Phillips B, Zideman D et al. An Fertil Sex 1999; 27: 222–230.
advisory statement from the Pediatric Working Group of the International Liasion 15 Fraser WD, Hofmeyr J, Lede R, Faron G, Alexander S, Goffinet F, et al., Amnioinfusion
Committee on Resuscitation. Pediatrics 1999; 103: e56–e69. Trial Group. Amnioinfusion for the prevention of the meconium aspiration syndrome.
8 Kattwinkel J. Textbook of Neonatal Resuscitation, 4th edn. American Academy of N Engl J Med 2005; 353: 909–917.
Pediatrics/American Heart Association: Elk Grove Village (IL)7, 2000. 16 Wu JM, Yeh TF, Wang JY, Wang JN, Lin YJ, Hsieh WS et al. The role of pulmonary
9 Gooding CA, Gregory GA. Roentgenographic analysis of meconium aspiration of the inflammation in the development of pulmunary hypertension in newborn with
newborn. Radiology 1971; 100: 131–140. Meconium Aspiration Syndrome (MAS). Pediatr Pulmonol Suppl 1999; 18:
10 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obstet Gynecol 205–208.
1975; 122: 767–771. 17 Yeh TF, Lin YJ, Lin HC, Wu JM, Lin CH. Early postnatal dexamethasone therapy in
11 Wenstrom KD, Parsons MT. The prevention of meconium aspiration in labor using infants with Meconium Aspiration Syndrome (MAS)Fa randomized clinical trial.
amnioinfusion. Obstet Gynecol 1989; 73: 647–651. Ped Res 1998; 43: 204A.

Journal of Perinatology
Journal of Perinatology (2008) 28, S49–S55
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Continuous positive airway pressure and conventional
mechanical ventilation in the treatment of meconium aspiration
syndrome
JP Goldsmith
Department of Pediatrics, Tulane University, New Orleans, LA, USA

extracorporeal membrane oxygenation.2 It appears that when used


Meconium aspiration syndrome (MAS) is a complex syndrome that ranges optimally, conventional therapies are adequate to treat the majority
in severity from mild respiratory distress to severe respiratory failure, of infants with this disease. Neonatal MAS challenges the practicing
persistent pulmonary hypertension of the newborn and sometimes death. clinician to make the correct diagnosis, understand the changing
Understanding of the syndrome’s complicated pathophysiology will help and complex pathophysiology of the disease, and tailor the
determine the appropriate treatment strategy, including the use of respiratory support to the specific alterations in pulmonary
continuous positive airway pressure (CPAP), conventional mechanical mechanics.
ventilation (CMV) and other therapies. Approximately 30 to 50% of infants
diagnosed with MAS will require CPAP or mechanical ventilation. The
optimum modes of ventilation for MAS are not known. Very few studies have Making the diagnosis of MAS
been conducted to determine ‘best’ ventilatory strategies. Despite the
Approximately 8 to 19% of all term deliveries occur through
introduction, over the last two decades, of innovative ventilatory treatments
meconium-stained amniotic fluid (MSAF) and MAS develops in
for this disease (for example, surfactant, high-frequency ventilation, inhaled
5 to 33% of these infants.3 Respiratory complications of MAS are
nitric oxide, extracorporeal membrane oxygenation), the majority of infants
often the consequence of hypoxia in utero with significant acidosis
can be successfully managed with CPAP or mechanical ventilation alone.
and depressed respirations at birth. There also appears to be a
Journal of Perinatology (2008) 28, S49–S55; doi:10.1038/jp.2008.156
greater risk of developing MAS as well as all respiratory
complications (that is, tachypnea, pneumonia, pulmonary air
leaks) if the meconium is ‘thick’ as opposed to ‘thin’.4 However, a
recent meta-analysis of the use of amnioinfusion showed no
neonatal benefit when meconium was thinned through this process
Introduction except in settings with limited peripartal surveillance.5 Infants who
Approximately 30 to 50% of infants diagnosed with meconium are vigorous at birth have a very low risk of developing MAS.6
aspiration syndrome (MAS) will require continuous positive airway Those who develop MAS are often greater than 41 weeks gestation
pressure (CPAP) or mechanical ventilation. The optimum modes of and have clinical signs of postmaturity (decreased subcutaneous
ventilation for MAS are not known. Very few studies have been tissue, peeling skin, long nails and so on). The respiratory signs of
conducted to determine ‘best’ ventilatory strategies. Retrospective MAS are similar to other neonatal respiratory diseases. These
reviews of the application of conventional mechanical ventilation include tachypnea, retractions, grunting, nasal flaring and
(CMV) to infants with this malady span three decades, during cyanosis. If the meconium has been present in utero for greater
which time a host of adjunctive therapies have become available.1,2 than 3 h, the infant may have meconium staining of the skin,
Despite multiple therapies for this syndrome, there is a disturbingly nails and umbilical cord. The anterior–posterior diameter of the
high-mortality rate (4 to 19%) historically. However, in the most chest may be increased if there is significant air trapping.
recent of these reviews, only 20% of 40 infants with MAS requiring The classic radiographic picture of MAS includes diffuse patchy
positive pressure assistance needed ‘innovative ventilatory support’ infiltrates with areas of atelectasis mixed with areas of
(defined as the use of high-frequency ventilation (HFV), inhaled hyperinflation throughout the lung fields. However, initially the
nitric oxide or surfactant) and no infants died or required chest X-ray may not be very diagnostic, as it may take many hours
for the chemical pneumonitis secondary to MAS to develop. Other
Correspondence: Dr JP Goldsmith, Department of Pediatrics, Tulane University, 1625 Joseph
Street, New Orleans, LA 70115-5034, USA. findings on X-ray include possible air leaks and cardiomegaly, if
E-mail: goldsmith.jay@gmail.com significant perinatal asphyxia has resulted in cardiomyopathy.
CPAP and CMV in the treatment of MAS
JP Goldsmith
S50

Later in the course of the disease, the MAS picture may be what is often implied when MAS is used as the primary diagnosis.’
complicated by the radiographic signs of surfactant deficiency Importantly, the treatment of the baby’s respiratory disease will
(atelectasis, air bronchograms, decreased lung volume) as the depend on the presence and severity of pulmonary hypertension.
surfactant system may be negatively impacted by the primary Thus the diagnosis of MAS may be assumed when three criteria
disease. are met: (1) a history of meconium in the trachea; (2) clinical
Despite the classic clinical and radiological appearance of MAS, evidence of significant respiratory distress; and (3) X-ray evidence
it may be difficult in certain cases to distinguish MAS from other of aspiration pneumonia.1 The association of transient tachypnea
neonatal respiratory diseases. Table 1 lists the differential and MSAF without meconium in the airway or X-ray evidence of
diagnosis.7 It is important diagnostically and physiologically to pneumonitis is often misdiagnosed as MAS and may lead to
determine if persistent pulmonary hypertension of the newborn inappropriate treatment, including early positive pressure, which
(PPHN) is present with significant right-to-left shunting of blood then may result in inadvertent pulmonary air leaks.
through the foramen ovale and/or the ductus arteriosus resulting
in exaggerated hypoxemia. PPHN may result secondary to MAS or
may be present by itself (‘primary PPHN’) with concurrent MSAF Pulmonary pathophysiology of MAS
but no true aspiration. As noted by Goetzman8 in these cases of Multiple mechanisms are involved in the pulmonary
primary PPHN, ‘when meconium is present, it is a fellow traveler pathophysiology of MAS. Meconium has deleterious effects on the
and not the cause of the PHN (PPHN) and hypoxemia, contrary to airways, injures the pulmonary parenchyma and alveoli, inhibits
surfactant function and may cause hypoxic pulmonary
Table 1 Differential diagnosis of MAS (when MSAF is present at delivery) vasoconstriction resulting in PPHN. The complex pulmonary effects
Transient tachypnea of the newborn
of MAS are seen in Figure 1.9 Individual infants may manifest any
Aspiration of amniotic fluid or blood one or more of these effects and the degree of respiratory distress
Respiratory distress syndrome may wax and wane according to the severity of the
Sepsis with pulmonary edema pathophysiology. Often there is a vicious cycle of air trapping,
Pneumonia ventilation–perfusion mismatch, hypoxemia, right-to-left
Congenital heart disease shunting, acidosis and increased pulmonary vascular resistance,
Intrapartum asphyxia with cardiomyopathy which may be difficult to treat successfully with standard therapies.
Persistent pulmonary hypertension of the newborn Meconium may easily obstruct both the large and small airways
Delayed transition in the newborn lung. When the obstruction is partial, a ‘ball-valve’
Abbreviation: MSAF, meconium-stained amniotic fluid. mechanism may lead to air trapping and air leaks (Figure 2).10 On
Adapted from Fuloria M, Wiswell TE.7 inspiration the airway dilates allowing gas to enter, but during

Alveolar and Effects of


parenchymal mediators
Altered lung elastic
inflammation (cytokines,
forces (increased
and edema eicosanoids)
resistance, decreased Surfactant
compliance) dysfunction

Protein leak
Airway into the
obstruction airways
Meconium
aspiration
syndrome
Effects of in utero
Direct toxicity
hypoxemia (pulmonary
by meconium
vascular remodeling,
constituents
lung parenchymal
changes)
Pulmonary
vasoconstriction due
Altered to components
pulmonary of meconium
vasoreactivity

Figure 1 The complex pulmonary pathophysiology of meconium aspiration syndrome. From reference 9 with permission.

Journal of Perinatology
CPAP and CMV in the treatment of MAS
JP Goldsmith
S51

a b Table 2 Theoretical classification of neonatal pulmonary disorders

Atelectatic Obstructive

Example Respiratory distress syndrome Meconium aspiration syndrome

Physiology kLung volume mLung volume


kCompliance kCompliance
kFunctional residual capacity mFunctional residual capacity
Normal airway resistance mAirway resistance
Normal time constant mTime constant
Key: m, increased; k, decreased.
Adapted from Goldsmith JP, Karotkin EH.17
Figure 2 Air trapping behind particulate matter (i.e. meconium) in an airway,
which leads to alveolar overexpansion and rupture. Tidal gas passes the
meconium on inspiration when the airway dilates (a), but does not exit on pulmonary vasodilators and an inflammatory response resulting
expiration when the airways constrict (b). From reference 10 with permission. from MAS, which releases vasoconstrictive substances and causes
platelet aggregation thus increasing pulmonary vascular resistance.
expiration the airway constricts around the obstruction, thus Hypoxemia and acidosis also result in increased pulmonary artery
trapping air inside the lung. This may result in overexpansion of pressure. Thus in both the acute and chronic hypoxic states, with
the lung, ventilation–perfusion mismatch, decreased compliance normal or increased pulmonary vessel muscularization, the
and possibly air leak. Complete obstruction of the airway by inflammatory response to MAS may trigger the vicious cycle of
meconium may result in atelectasis and resultant hypoxemia and postnatal hypoxia, pulmonary vasoconstriction, right-to-left
hypercapnia. shunting and further hypoxia. In the presence of chronic vascular
Meconium also causes direct pulmonary parenchymal and remodeling, the combination of MAS and PPHN is much more
alveolar injury resulting in a profound inflammatory response. The difficult to treat.
release and migration of inflammatory cells and mediators, such
as cytokines and phospholipase A2, to the lung may cause direct
pulmonary injury such as epithelial cell necrosis, cell death and Altered pulmonary mechanics of MAS
apoptosis.11,12 These mediators may also result in constriction of In the theoretical classification of neonatal pulmonary disorders,
pulmonary vessels, alteration and impairment of neutrophil MAS is considered an obstructive disease, although in reality the
function and cause capillary leakage and edema similar to condition is both obstructive and atelectatic, and changes with time
respiratory distress syndrome.9 and treatment. Table 2 shows the physiologic differences between
Multiple investigations in both animals and humans have ‘pure’ atelectatic and obstructive lung disease.17 MAS is
shown the deleterious effects of meconium on endogenous characterized in this simplified chart as an obstructive disease with
surfactant.12–14 Meconium may inhibit surfactant production, increased lung volume, decreased compliance, increased functional
displace surfactant from the alveolar surface and decrease its residual capacity and increased airway resistance and time
surface tension lowering function. Surfactant inhibition is constants. The pathophysiology of MAS discussed in the previous
concentration dependent and results from a variety of toxic section alters the respiratory mechanics of the lung in four major
biochemical effects of the meconium constituents. The influx of ways as seen in Table 3. Understanding the principles of normal
neutrophils into the airways subsequent to these toxic effects ventilation and the alteration in respiratory mechanics secondary
produces proteases, which further degrade surfactant proteins and to MAS pathology is necessary to devise ventilation strategies to
frequently results in hemorrhagic edema of the lung. treat affected infants with CPAP and/or CMV.
Fox et al.15 initially reported the association of pulmonary Meconium in the pulmonary airways leads to increased airway
hypertension in association with perinatal aspiration syndromes. resistance and prolonged time constants. The time constant is a
The majority of cases of significant PPHN are associated with MAS. measure of how quickly the lung can inflate or deflate, or how
The relationship of PPHN with MAS is complex and multifactorial. long it takes for alveolar and proximal airway pressures to
Vasoconstriction of pulmonary vessels may be associated with equilibrate. The expiratory time constant (Kt) is directly related to
normal or increased muscularization of the vessel walls.16 both compliance (CL) and airway resistance (Raw) by the
Prolonged in utero hypoxia may result in vascular remodeling equation: Kt ¼ CL  Raw. It takes three time constants to discharge
secondary to hypertrophy of the medial musculature. Acute 95% of the tidal volume of one breath. If the time constant is
maladaptation secondary to intrapartum asphyxia with normal prolonged secondary to increased airway resistance (without a
vessel anatomy is caused by the inhibition of endogenous concomitant reduction in lung compliance), the patient is at risk

Journal of Perinatology
CPAP and CMV in the treatment of MAS
JP Goldsmith
S52

Table 3 Ventilation strategies to match pathophysiology of MAS TLC

Pathophysiology Pulmonary Ventilation strategy


dynamics C

Airway obstruction; mLung volume Avoid ventilation on ‘C’ portion of high FRC
(overexpansion)
ball-valve mAirway resistance compliance curve; Adequate

Volume
phenomenon mTime constants expiratory time; Avoid inverse I:E
B
ratios
Parenchymal/ kCompliance Adequate CPAP or PEEP
alveolar injury Ventilation– Avoid high inflating pressures
perfusion mismatch normal FRC
Surfactant kCompliance; Adequate CPAP or PEEP; A
dysfunction Ventilation– Volume recruitment strategy; low FRC (atelectasis)
perfusion mismatch Avoid overdistension to k incidence
of PAL Pressure
FRC = functional residual capacity
Pulmonary Exaggerated Generous oxygenation
TLC = total lung capacity
hypertension hypoxia; (80 to 120 mm Hg);
Ventilation– Avoid significant acidosis Figure 3 Lung expansion or compliance curve with flattened areas (A and C) at
perfusion mismatch both ends. Area (C) represents the situation in an overexpanded lung as occurs
with air trapping. FRC, functional residual capacity; TLC, total lung capacity.
Abbreviations: m, increased; k, decreased; CPAP, continuous positive airway pressure; I:E, From reference 19 with permission.
inspiratory:expiratory; MAS, meconium aspiration syndrome; PAL, pulmonary air leak;
PEEP, Positive end expiratory pressure.

The deactivation of surfactant from MAS complicates this


for incomplete emptying of the previously inspired breath. This picture. It makes the use of PEEP mandatory during CMV despite
condition may be exaggerated when the patient is on assisted the high functional residual capacity, increased time constant and
ventilation and there is insufficient ventilator time for exhalation air trapping that often occurs. Fox et al.20showed in a small
because of an abnormally long time constant. The result of this number of infants (n ¼ 14) that moderate levels of PEEP
‘inadvertent PEEP’will be gas trapping accompanied by an increase (4 to 7 cm H2O) resulted in the greatest increase in oxygenation
in lung volume, a build up of pressure in the alveoli and distal in babies ventilated for MAS (Figure 4).
airways, and decreased compliance.18 The addition of PPHN to the MAS pathophysiology further
Significant air trapping may result in hyperinflation and complicates the pulmonary mechanics and makes ventilatory
flattening of the diaphragms on chest X-ray, increased anterior– strategies very difficult. Increased pulmonary vascular resistance
posterior diameter of the chest and even cardiovascular effects such may result from hypoxia, acidosis and reduced cardiac output.
as decreased cardiac output, diminished blood pressure and signs Ventilatory strategies must recognize these triggers. Allowing mild
of poor perfusion. Babies with increased functional residual hyperoxia in these infants may reduce the periods of hypoxemia
capacity and high lung volumes have decreased compliance as that occur with interventions or agitation. Pulmonary toilet must
seen in the flattened portion or section ‘C’ of the standard be done judiciously with careful attention to the pulse oximeter.
pressure–volume curve (Figure 3).19 Attempting to assist Appropriate sedation and limiting interventions while nursing the
ventilation at the upper end of the compliance curve (that is, at infant in a dimly lit and quiet area of the neonatal intensive care
high lung volumes) results in a further decrease in compliance unit are all helpful adjuncts to care. Although ‘gentle ventilation’
and may lead to pulmonary air leaks. is recommended by some,21 permissive hypercapnia and the
As noted above, meconium has a direct injurious effect on the resultant respiratory acidosis may worsen the pulmonary
pulmonary airways, parenchyma and alveoli. This often results in hypertension. At the other extreme, very aggressive assisted
capillary leak and edema. The additional injury of baro- ventilation with hyperventilation in an attempt to improve
volutrauma from assisted ventilation through an endotracheal tube oxygenation may contribute to overdistension of the lung, air leaks
should be avoided if possible. Thus the use of oxygen by hood or and decrease cardiac output, further aggravating the PPHN.
cannula and nasal CPAP is advocated to avoid intubation and the
bellows action of CMV. However, term and near-term infants may
not tolerate nasal CPAP despite adequate sedation. Moreover, the Conventional ventilation strategies
use of a ‘permissive hypercapnia’ technique in these infants is As there is little evidence from clinical trials on ventilator treatment
problematic because of the sensitivity of the pulmonary vasculature of MAS, conventional management is primarily based on
to acidosis. pulmonary pathophysiology. To avoid additional air trapping and

Journal of Perinatology
CPAP and CMV in the treatment of MAS
JP Goldsmith
S53

25 The complicated pulmonary pathophysiology resulting from


areas of atelectasis and areas of hyperinflation, in association with
20
ventilation–perfusion mismatch and airway compromise, makes
ventilator management of MAS one of neonatology’s greatest
challenges. The ventilator strategy used will depend on the stage
∆ PaO2 (torr/cm H2O EEP)

15 and severity of the disease, especially the presence or absence of


significant pulmonary hypertension. Clinicians vary in their desire
to keep blood gas values in various ranges. Prior to the use of
10
inhaled nitric oxide and HFV, some clinicians employed
hyperventilation to achieve alkalosis by hypocapnia in an attempt
5 to decrease pulmonary vascular resistance.24 However, follow-up
studies on infants managed with this strategy have shown an
increased incidence of sensorineural hearing loss, pulmonary
0 barotrauma and adverse neurologic outcomes.25,26 Thus
hyperventilation has become a less-used strategy, as clinicians feel
that the newer modalities such as HFV and/or inhaled nitric oxide
Low Medium High are less dangerous than sustained hypocapnia. Contemporary
(0-3) (4-7) (8-14)
management employs the acceptance of standard blood gas ranges
Intial EEP (cm H2O)
(mean ± SEM)
or ‘gentle ventilation,’ a strategy in which higher PaCO2 levels and
lower PaO2 levels are targeted in the belief that ventilator-induced
Figure 4 Incremental improvement in oxygenation with different levels of lung injury will be reduced.21 Other than the fear of intermittent
positive end expiratory pressure in the treatment of meconium aspiration
syndrome. From reference 20 with permission. hypoxemia worsening PPHN, there is probably little reason to keep
babies hyperoxic. Moreover, catheterization data in older infants
with pulmonary hypertension secondary to bronchopulmonary
dysplasia suggest that oxygen tensions that exceed 70 to 80 mm Hg
possibly air leaks, many clinicians will treat initially with oxygen do little to decrease pulmonary artery pressure.27 No trials have
alone without positive pressure of any type. An inspired oxygen compared any of these strategies in the treatment of infants with
fraction (FiO2) of up to 1.0 has been advocated in the past to treat MAS.
hypoxemia. However, exposure to 100% oxygen for even brief Infants with MAS without associated PPHN may be managed in
periods of time in the newborn lamb increased the contractile a relatively standard fashion with slightly higher oxygen targets
response of small pulmonary arteries, presumably because of the than used for premature infants with respiratory distress syndrome.
production of reactive oxygen species.22 Human and animal studies The pH should be maintained above 7.3, PaCO2 in the 40 to
have shown significant biochemical and clinical negative effects 50 mm Hg range and the PaO2 targeted at 70 to 80 mm Hg. This
after the use of 100% oxygen during resuscitation. Therefore, it may be achieved with a moderate peak inflating pressure preferably
may be prudent to avoid oxygen concentrations greater than 80% not exceeding 25 cm H2O, a relatively rapid ventilator rate (40 to
and switch to some form of positive pressure if concentrations 60 breaths per minute), a moderate PEEP (4 to 6 cm H2O) and an
higher than this are necessary. An oxyhood is preferred to nasal adequate expiratory time (0.5 to 0.7 s) to prevent air trapping.
cannula as term and near-term infants may be less tolerant of a This strategy requires a relatively short inspiratory time of 0.3 to
cannula, and the percentage of inspired oxygen can be greater in a 0.4 s. If the diaphragms on chest X-ray are flat and gas trapping is
hood. Sufficient oxygen should be given to maintain oxygen suspected, the expiratory time should be increased to 0.7 to 0.9 s,
saturations 90 to 95%. The partial pressure of oxygen (PaO2) target PEEP decreased to 3 to 4 cm H2O and the rate decreased to allow at
may be as high as 90 mm Hg as the risk of eye and lung toxicity is least a 0.25 inspiratory time. Borderline hypoxemia in this
less in term infants, and the dangers of intermittent hypoxemia situation may be acceptable as increased ventilator settings may
may result in the development or worsening of PPHN. cause lung injury or the patient may be converted to HFV to
Although CPAP or CMV may be necessary to treat hypoxemia, improve oxygenation.
hypercapnia and acidosis, there is no general agreement on blood In infants whose MAS is complicated by significant PPHN, the
gas parameters to initiate these therapies. Ambalavanan et al.23 ventilatory strategy is somewhat different. Intermittent hypoxemia
advise that infants with MAS who have PaO2 <50 mm Hg, partial can be mitigated by allowing the PaO2 to be as high as 80 to
pressure of carbon dioxide (PaCO2) >60 mm Hg, or acidosis (pH 100 mm Hg without great concern for oxygen toxicity. As oxygen
<7.25) in an oxygen-enriched environment with FiO2 >0.80 are saturations have never been studied in this context and are
candidates for mechanical ventilation. surrogates for oxygen tensions, which do not correlate well when

Journal of Perinatology
CPAP and CMV in the treatment of MAS
JP Goldsmith
S54

saturations are greater than 95%, monitoring by saturation alone is from the disease. In a time when the newest and most costly
not recommended. Some clinicians also prefer to target mild therapy seems to find a use in every patient, we can only admire
hypocapnia (PaCO2 range of 30 to 35 mm Hg), which results in and aspire to the benchmark that Dr Bhutani et al. have set.
mild alkalosis as a buffer to acidosis that may trigger a downward
PPHN spiral. In both clinical situations of MAS with and without Disclosure
PPHN, a volume-recruitment strategy is preferred to recruit
JP Goldsmith has received consulting fees from Discovery Labs.
atelectatic areas of the lung and treat ventilation–perfusion
mismatch. Such an approach resulted in improved oxygenation References
and less ventilator-induced lung injury in an experimental piglet
1 Vidyasagar D, Yeh TF, Harris V, Pildes RD. Assisted ventilation in infants with
model.28 This strategy has to be tempered with the knowledge that meconium aspiration syndrome. Pediatrics 1975; 56: 208–213.
such an approach may worsen air trapping in the face of 2 Bhutani VK, Chima R, Sivieri EM. Innovative neonatal ventilation and meconium
mechanical airway obstruction. aspiration syndrome. Indian J Pediatr 2003; 70(5): 421–427.
All types of conventional methods of ventilation have been used 3 Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a
in these cases. Most standard ventilators now allow the operator to difference? Pediatrics 1990; 85: 848–852.
4 Wiswell TE, Gannon CM, Jacob J, Goldsmith L, Szyld E, Weiss K et al. Delivery room
choose among intermittent mandatory ventilation, patient-
management of the apparently vigorous meconium-stained neonate: results of the
triggered ventilation, pressure support ventilation and/or volume multicenter, international collaborative trial. Pediatrics 2000; 105(1 Part 1): 1–7.
controlled ventilation modes. None of these modalities have been 5 Fraser WD, Hofmeyr GJ, Lede R, Faron G, Alexander S, Goffinet F et al. An
compared with any other in a rigorous way, although each mode international trial for the prevention of meconium aspiration syndrome. New Engl J
of ventilation has its supporters. As non-asphyxiated term infants Med 2005; 353: 909–917.
may vigorously fight assisted ventilation, one approach has been to 6 Halliday HL, Sweet D. Endotracheal intubation at birth for preventing morbidity and
mortality in vigorous, meconium-stained infants born at term (Cochrane Review). The
use a patient-triggered mode (synchronized intermittent mandatory Cochrane Library, Issue 2, 2003. Oxford: Update Software.
ventilation or assist/control) in conjunction with adequate 7 Fuloria M, Wiswell TE. Managing meconium aspiration. Contemp Ob/Gyn 2000; July:
sedation. Muscle relaxation is rarely necessary and has significant 113–125.
drawbacks when this step is considered to achieve acceptable blood 8 Goetzman BW. Meconium aspiration. Am J Dis Child 1992; 146: 1282–1283.
gases and avoid high ventilatory pressures and possible 9 Wiswell TE, Srinivasan P, Roberton NRC. Aspiration syndromes. In: Greenough A,
Milner AD (eds). Neonatal Respiratory Disorders, 2nd edn, Arnold: London, UK, 2003,
barotrauma, it may be time to move to alternative therapies (that
pp 334–354.
is, inhaled nitric oxide, HFV). Careful attention must be paid to the 10 Harris TR, Herrick BR. Pneumothorax in the Newborn. Biomedical Communications,
environment in which these babies are nursed and ventilated. Arizona Health Sciences Center: Tucson, AZ, 1978.
Although it seems appropriate to provide chest physiotherapy and 11 Davey AM, Becker JD, Davis JM. Meconium aspiration syndrome: physiologic and
pulmonary toilet, these interventions must be done with caution inflammatory changes in a newborn piglet model. Pediatr Pulmonol 1993; 16:
(or not at all) as they may trigger hypoxemia, agitation and an 101–108.
12 Zagariya A, Bhat R, Uhal B, Navale S, Freidine M, Vidyasagar D. Cell death and lung
increase in pulmonary vascular resistance. Other nursing cell histology in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; 159:
interventions must be considered in the same context with 819–826.
particular attention paid to minimize environmental stimuli such 13 Dargaville PA, South M, McDougall PN. Surfactant and surfactant inhibitors in
as light and noise. meconium aspiration syndrome. J Pediatr 2001; 138: 113–115.
14 Cleary GM, Wiswell TE. Meconium stained amniotic fluid and the meconium
aspiration syndrome: an update. Pediatr Clin North Am 1998; 45: 511–529.
15 Fox WW, Gewitz MH, Dinwiddie R, Drummond WH, Peckham GJ. Pulmonary
Conclusions
hypertension in the perinatal aspiration syndromes. Pediatrics 1977; 59: 205–211.
MAS is a complex syndrome that ranges in severity from mild 16 Walsh-Sukys M. Persistent pulmonary hypertension of the newborn: the black box
respiratory distress to severe respiratory failure, PPHN and revisited. Clin Perinatol 1993; 20: 127–143.
sometimes death. Understanding of the syndrome’s-complicated 17 Goldsmith JP, Karotkin EH. Introduction. In: Goldsmith JP, Karotkin EH (eds). Assisted
pathophysiology will help determine the appropriate treatment Ventilation of the Neonate, 4th edn, WB Saunders: Philadelphia, 2003, pp 1–14.
18 Simbruner G. Inadvertent positive end-expiratory pressure in mechanically ventilated
strategy, including the use of CPAP, CMV and other therapies. The newborn infants: detection and effect on lung mechanics and gas exchange. J Pediatr
use of echocardiography, pulmonary function studies and graphics 1986; 108: 589–595.
will help guide the clinician in the choice of therapies. As shown by 19 Wood BR. Physiologic principles. In: Goldsmith JP, Karotkin EH (eds). Assisted
Bhutani et al.2 at Pennsylvania Hospital over a period of 6 years, Ventilation of the Neonate. 4th edn, WB Saunders: Philadelphia, 2003, pp 15–40.
the use of a systems-based strategy for babies delivered through 20 Fox WW, Berman LS, Downes Jr JJ, Peckham GJ. The therapeutic application of
end-expiratory pressure in the meconium aspiration syndrome. Pediatrics 1975; 56:
MSAF can minimize adverse outcomes. This review showed that
214–217.
conventional respiratory therapies such as oxygen, CPAP and CMV 21 Gupta A, Rastogi S, Sahni R, Bhutada A, Bateman D, Rastogi D et al. Inhaled nitric
were adequate treatment for 80% of MAS-affected infants with no oxide and gentle ventilation in the treatment of pulmonary hypertension of the
infant requiring extracorporeal membrane oxygenation or dying newbornFa single-center, 5 year experience. J Perinatol 2002; 22: 435–441.

Journal of Perinatology
CPAP and CMV in the treatment of MAS
JP Goldsmith
S55

22 Lakshminrusimha S, Ruddell JA, Steinhorn RH, Ryan RM, Gugino SF, Morin FC et al. 26 Walsh-Sukys MC, Tyson JE, Wright LL, Bauer CR, Korones SB, Stevenson DK et al.
Pulmonary arterial contractility in neonatal lambs increases with 100% oxygen PPHN in the era before NO: practice variation and outcomes. Pediatrics 2000;
resuscitation. Pediatr Res 2006; 59(1): 137–141. 105(1 Part 1): 14–20.
23 Ambalavanan N, Schelonka R, Carlo W. Ventilatory strategies. In: Goldsmith JP, 27 Mourani PM, Ivy DD, Gao D, Abman SH. Pulmonary vascular effects of inhaled nitric
Karotkin EH (eds). Assisted Ventilation of the Neonate, 4th edn, WB Saunders: oxide and oxygen tension in bronchopulmonary dysplasia. Am J Respir Crit Care Med
Philadelphia, 2003, pp 249–259. 2004; 170(9): 1006–1013.
24 Drummond WH, Peckham GJ, Fox WW. The clinical spectrum of the newborn with 28 van Kaam AH, Haitsma JJ, De Jaegere A, van Aalderen WM, Kok JH, Lachmann B.
persistent pulmonary hypertension. Clin Pediatr 1977; 16: 335–341. Open lung ventilation improves gas exchange and attenuates secondary lung
25 Bifano EM, Pfannenstiel A. Duration of hyperventilation and outcome in infants with injury in a piglet model of meconium aspiration. Crit Care Med 2004; 32:
persistent pulmonary hypertension. Pediatrics 1988; 81: 657–666. 443–449.

Journal of Perinatology
Journal of Perinatology (2008) 28, S56–S66
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Treatment of MAS with PPHN using combined therapy:
SLL, bolus surfactant and iNO
J Gadzinowski1, K Kowalska1 and D Vidyasagar2
1
Department of Neonatology, University of Medical Sciences, Poznan˜, Poland and 2Neonatal Division, Department of Pediatrics,
UIC, Chicago, USA

of treatment may thus lead to a thorough removal of meconium


The objective of the study was to compare the effectiveness of surfactant from the respiratory tracts and minimize the damaging effect of
treatment either by bolus or surfactant lung lavage followed by inhaled meconium on the endogenous surfactant.1 Another major
nitric oxide (iNO) therapy in infants with meconium aspiration syndrome complication of MAS is the development of persistent pulmonary
(MAS) complicated by persistent pulmonary hypertension (PPHN). In this hypertension (PPHN). Inhaled nitric oxide (iNO) has been used
study, thirteen infants with diagnosis of MAS and PPHN were first treated successfully to manage infants with PPHN. The effectiveness of iNO
with conventional respiratory support. Then between 2 and 22 h of life they would depend on unimpeded delivery of the gases to the
were randomized either to bolus surfactant treatment (n=6) or surfactant well-ventilated parts of the lungs. It is possible that iNO does not
lung lavage (SLL, n=7) treatment. Then all infants were treated with iNO reach the alveoli, which are covered with meconium. Therefore, we
therapy. The groups were compared with regard to their clinical course: hypothesize that SLL will clear the meconium, stabilize the
changes in PaO2, FiO2, MAP, OI, A-a oxygen gradient, duration of iNO alveoli and increase the diffusion of iNO resulting in better
therapy, length of ventilation and hospitalization. Complications and oxygenation.
mortality were also compared. The results showed that infants treated with
SLL had significant improvements in oxygenation, decreases in MAP and
A-a gradients. But there were no significant differences in duration of The objective of the trial
ventilation, iNO treatment, length of hospitalization or complications. In The objective of our trial was to study the effectiveness of the
conclusion these data show no advantage of SLL therapy over bolus combined therapies of bolus surfactant treatment or SLL followed
surfactant treatment in infants with MAS complicated by PPHN. by iNO in infants with meconium aspiration syndrome complicated
Journal of Perinatology (2008) 28, S56–S66; doi:10.1038/jp.2008.163 by pulmonary hypertension.

Materials
This study was conducted in the neonatal intensive care unit
Introduction
(NICU) of the Neonatology Department of the Poznan University of
Improved perinatal management of meconium aspiration has Medical Sciences, Poznan, Poland. We enrolled 13 neonates into
contributed substantially to a decrease in incidence and mortality the study during the period between 1998 and 2004. The diagnosis
because of meconium aspiration syndrome (MAS). In infants not of MAS was based on clinical and radiological criteria. PPHN was
responding to conventional treatment of MAS, other methods such diagnosed on the basis of echocardiogram results.
as bolus surfactant, surfactant lung lavage (SLL) as well as
extracorporeal membrane oxygenation (ECMO) can be used.
Surfactant treatment by bolus may be associated with Methodology of the trial
inactivation by meconium in the airways, which may explain Randomization is shown in the diagram.
non-responsiveness in earlier studies.1 It has been suggested that
the administration of surfactant followed by SLL may lead to a Group A1
washout of residual meconium both mechanically and because of Group A1 included seven newborns who were treated with a
the improved mucocilliary transport of meconium. This approach combined therapy (SLL þ bolus surfactant þ iNO). Besides the
conventional treatment, SLL with a surfactant solution Survanta
Correspondence: Professor J Gadzinowski, Department of Neonatology, Katedra Neonatologii
UM w Poznaniu, Ul. Polna 33, 60-535 Poznañ, Poland. was carried out between the 2nd and 22nd hour of life (average
E-mail: jgadzin@gpsk.am.poznan.pl time of treatment initiation 9.7±7.4 h). After the lavage
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S57

treatment, one dose of bolus Survanta at 100 mg of phospholipids (A) Preparation of surfactant solution for lavage2
per kg of body weight was given.
Subsequently, echocardiogram assessment was carried out 1. A natural surfactant Survanta (Ross Abbott Laboratories,
for the diagnosis of PPHN; iNO was administered when Zwolle, The Netherlands) was used.
PPHN was diagnosed. The initial dose of nitric oxide was 20 p.p.m. 2. The dose of phospholipids for the lavage solution was 5 mg in
(Figure 1). 1 ml of physiological salt.
3. The volume of the solution was 15 ml per kg of the patient’s
Group A2 body weight.
Group A2 included six newborns treated conventionally. Between 4. The calculated volume was divided into four parts.
the 2nd and 22nd hour of life (average time of treatment initiation 5. A single lavage volume was 2 ml kg1.
11±6.4 h), the newborns were given one dose of Survanta as a
Example of calculation: If the patient’s body weight was 3000 g,
bolus in the amount of 100 mg per kg of body weight. Infants did
45 ml of surfactant solution was used as a lavage: 9 ml of Survanta
not undergo SLL. After giving one dose of surfactant as well as
supplemented with normal saline up to 45 ml.
conducting an echocardiographic assessment and diagnosing with
PPHN, iNO was administered with an initial dose of NO at
(B) Lavage technique2
20 p.p.m.
Installation of a closed circuit system was used for the
administration of lavage. This was possible using a Bodai valve in
Lavage with a surfactant solution. Bronchoalveolar lavage with the connector in the endotracheal tube. The surfactant solution
saline natural surfactant solution was in newborns fulfilling the was instilled through the connector, whereas the lung fluid was
inclusion criteria, which was carried out after sedation and initial suctioned through the Bodai valve (Figure 2). This closed lavage
cardiopulmonary stabilization. The steps in surfactant lavage are and suctioning system helped maintain both mechanical
described below. ventilation and positive end-expiratory pressure (PEEP) at constant
levels. The prepared volume of the solution was divided into four
parts. Lavage and suctioning were conducted in four body
positions: on the right and left sides, and in the Trendelenburg and
MAS +PPHN anti-Trendelenburg positions, similar to the administration of the
Survanta dose given as treatment (in line with the producer’s
recommendations).
A1- SLL+ BOLUS A2- BOLUS The surfactant solution was administered through the
endotracheal tube. After 2 ml of the solution was instilled, the
mechanical ventilation was continued. After 3 to 5 respiratory
cycles, the secretions were suctioned (Figure 2).
iNO iNO

Figure 1 Scheme of the trial.

Treatment with bolus surfactant. Bolus surfactant was


administered through the endotracheal tube after sedation and
Surfactant stabilization.
solution (A) Surfactant preparation
A natural surfactant Survanta (Ross Abbott Laboratories) was
used.
(B) The technique of surfactant administration
The surfactant was administered endotracheally with a catheter
through an intubation tube and through the Bodai valve placed in
Bodai valve
the connector by the intubation tube. During the application, the
mechanical ventilation was continued. The dose of surfactant was
100 mg per kg of body weight. The surfactant was administered in
four positions: on the right and left sides, and in the Trendelenburg
and anti-Trendelenburg positions. Bolus surfactant was
administered once after SLL (group A1) and also once but without
Figure 2 The surfactant lung lavage (SLL) technique. SLL prior to it (group A2).

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S58

Safety of the procedures. During the whole procedure, saturation condition in the first minutes of life. No statistically significant
and heart rate were monitored. Saturation was considered safe if, differences were observed in the frequency of Cesarean section
during lavage, it was within the range of 87 to X93%. If as well as the time for inclusion in the trial.
saturation fell below 87%, the parameters of mechanical ventilation In group A1, the maturity of the newborns was 38.1 (±2.3)
were adjusted: peak pressure, fraction of inspired oxygen (FiO2) weeks of gestation. The average birth weight was 3267 g (±822.4).
and frequency of breaths. Lavage or surfactant was not given until The overall condition of the newborns in the 1st and 5th minutes
saturation reached a safe level. Once this was achieved, the of life was assessed according to the Apgar scale. The median in the
procedure was continued. first minute of life was 5 points and it was 7 points in the fifth
minute of life.
In group A2, the maturity of the newborns was 37.8 (±2.1)
Parameters analyzed in the trial weeks of gestation. The average birth weight was 3128 g (±1155.4).
The following parameters were analyzed in both groups: The overall condition of the newborns in the first and fifth minutes
of life was assessed on the basis of Apgar scale. The median in the
(1) partial arterial oxygen pressure (PaO2); first minute of life was 4 points and it was 6 points in the fifth
(2) Fraction of inspired oxygen (FiO2); minute of life.
(3) oxygenation index (OI);
(4) alveolar–arterial oxygen gradient (AaDO2).
The parameters listed above were analyzed at six time intervals: Inclusion criteria. The trial was prospective and randomized
and included newborns who fulfilled the following criteria:
 At 15 min before treatment initiation (0 h);
 after 1 h of combined therapy (1 h); (1) Gestational ages X35 weeks of gestation;
 after 2 h of combined therapy (2 h); (2) postnatal age p24 h;
 after 4 h of combined therapy (4 h); (3) MAS was diagnosed on the following basis:
 after 24 h of combined therapy (24 h);  presence of meconium-stained amniotic fluid below the
 after 48 h of combined therapy (48 h). vocal cords;
 clinical symptoms: respiratory failure that required
The following were also analyzed: treatment with mechanical ventilation (FiO2X40%) to
1. length of time on mechanical ventilation; maintain PaO2 at 50 to 80 mm Hg and saturation of 87 to
2. duration of iNO treatment; 93%;
3. length of hospital stay;  radiological findings consistent with MAS.
4. complications; (4) Parental consent to enter the baby in the study.
5. number of deaths.
Echocardiogram assessment was conducted for the diagnosis of
PPHN in all infants diagnosed as MAS. PPHN was diagnosed on the
Equipment. Intermittent mandatory ventilation (IMV) was basis of the following echocardiographic parameters:3–6
used as treatment for respiratory failure. Echocardiographic
examination was conducted by using HDI 3500 ATL and a  mean pulmonary arterial pressure in the pulmonary trunk
trans-receiving head with a frequency of 7 to 4 MHz. iNO was X40 mm Hg;
administered by using SLE 3600 INOSYS. The initial and, at the  ductus arteriosus and foramen ovale;
same time, maximum dose of NO was 20 p.p.m. Saturation was  tricuspid valve insufficiency (TI);
measured by using pulse oximeters, and maintained in the range  out of the wave A in M-mode pulmonary valve presentation.
between 87 and 93%. Arterial blood PO2 and PCO2 were measured. The blood flow in the pulmonary artery was recorded by the
Biochemical analysis was conducted by using Rapidlab. Doppler method using a pulse-wave technique and a trans-
receiving head with a high frequency of ultrasonic beams (7 to
Clinical classification and characteristics of recruited 4 MHz). The pressure in the pulmonary trunk was calculated on
newborns. After the diagnosis of MAS, the newborns were divided the basis of the time from the initial blood flow wave until it
into two groups: A1 and A2 (Figure 1) by randomization. In group reached the maximum speed (acceleration time (AcT)). The
A1, infants received bolus surfactant treatment followed by iNO. outflow wave was registered on the way from the right ventricle
Group A2 received SLL followed by iNO therapy. There were no next to the ring of the pulmonary valve or in the pulmonary trunk.
statistically significant differences in the following parameters: The flow acceleration time is shortened in the pulmonary artery.3
birth weight, gestational age as well as assessment of clinical The mean pressure was calculated on the basis of a mathematical

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S59

formula:4 80(0.5AcT). Mean pulmonary artery pressure (MPAP) the minimum and maximum values. Apgar score between the groups
<40 mm Hg was defined as the norm. was compared using the non-parametric Mann–Whitney test.
The parameters expressed in the nominal scale, such as the
Randomization. Infants were randomized into groups A1 and number of deaths and complications, were described with numbers
A2. Each group of infants was managed by a different group of and corresponding percentages. Dependence was tested with
neonatologists. Throughout the study, the infants stayed in the Fisher’s exact test. Statistical significance of a ¼ 0.05 was assumed
hospital. for verification of statistical tests.
Statistical calculations were performed using a statistical set
Statistical analysis. Parameters such as PaO2, FiO2, mean StatSoft Inc. (2005), STATISTICA, Tulusa, AZ, USA (data analysis
airway pressure (MAP), OI, AaDO2 as well as duration of hospital software system), version 7.1.
stay and the length of time on IMV and iNO were expressed in the
interval scale and described with the arithmetic mean value, Results of the trial
standard deviation as well as the minimum and maximum values PaO2
and confidential interval. The mean initial (0 h) PaO2 before treatment initiation was
The normal distribution of these parameters was checked with 48.4 mm Hg (±15.0) in group A1. After 1 h of treatment, an
Shapiro–Wilk’s test. For the parameters that showed normal increase to 114.4 mm Hg (±49.8) was observed. After 2 h, the
distribution, the Student’s t-test for independent variables was used value was 68.1 mm Hg (±15.3), after 4 h 69.6 mm Hg (±19.0),
for comparison of groups A1 and A2; otherwise, non-parametric after 24 h 78.7 mm Hg (±18.9) and after 48 h 67.9 mm Hg
Mann–Whitney’s test was used. (±12.3) (Table 1).
For comparisons of values obtained at 0, 1, 2, 4, 24 and 48 h, In group A2, the initial (0 h) oxygen pressure value before
once the normal distribution was confirmed, the analysis of treatment was 45.1 mm Hg (±14.3). No increase in this
variances for repeatable variables was carried out using the post parameter was observed, but after 1 h, its value was 46.7 mm Hg
hoc Newman–Keuls test; if normal distribution was not confirmed, (±14.9). An increase up to 51.1 mm Hg (±19.6) was observed
the non-parametric Friedman test was used with Dunn’s test for after 2 h, which further increased to 65.5 mm Hg (±38.6) after
repeated comparisons. The Apgar scale as a parameter, expressed in 4 h and a similar level of 61.9 mm Hg (±18.9) after 24 h and
the ordinal number scale, was described with the median as well as 64.0 mm Hg (±14.0) after 48 h (Table 2).

Table 1 Partial arterial oxygen pressure in group A1

PaO2 (mm Hg) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 7 48.4 34.6 62.2 15.0 32.6 70.0 43.0


1h 7 114.4 68.3 160.4 49.8 36.4 203.0 116.0
2h 7 68.1 54.0 82.3 15.3 53.0 86.0 59.0
4h 7 69.6 52.0 87.2 19.0 52.0 108.0 67.0
24 h 7 78.7 61.2 96.2 18.9 58.0 112.0 74.0
48 h 7 67.9 56.5 79.3 12.3 51.0 83.0 62.5
A statistically significant increase of PaO2 in group A1 was observed between 0 and 4 h of treatment (P ¼ 0.0311).

Table 2 Partial arterial oxygen pressure in group A2

PaO2 (mm Hg) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 6 45.1 30.0 60.1 14.3 36.6 74.0 40.2


1h 6 46.7 31.0 62.4 14.9 28.6 61.5 50.4
2h 6 51.1 30.7 71.6 19.6 31.0 80.0 46.7
4h 6 65.5 25.0 105.8 38.6 23.0 134.0 54.5
24 h 6 61.9 38.4 85.5 18.9 41.9 88.0 52.9
48 h 6 64.0 46.6 81.4 14.0 51.1 88.0 60.0
Increase of PaO2 in group A2, during the 48 h treatment was not statistically significant (P>0.05).

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S60

140
P =0.0105
120
mmHg 100

80 PaO2 A1
60 PaO2 A2

40

20

0
0h 1h 2h 4h 24h 48h

Figure 3 Partial arterial oxygen pressure in groups A1 and A2.

Table 3 Fraction of inspired oxygen in group A1

FiO2 (%) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 7 95.7 85.2 106.2 11.3 70.0 100.0 100.0


1h 7 79.4 57.9 100.7 23.2 50.0 100.0 90.0
2h 7 67.1 48.7 85.6 20.0 40.0 100.0 60.0
4h 7 57.1 41.8 72.4 16.6 35.0 85.0 60.0
24 h 7 53.7 31.6 75.8 25.0 21.0 90.0 60.0
48 h 7 40.1 22.0 58.3 19.6 21.0 70.0 30.0
A statistically significant drop of FiO2 in group A1 was observed between zero and first hour of treatment (P ¼ 0.0323).

Table 4 Fraction of inspired oxygen in group A2

FiO2 (%) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 6 95.0 82.1 107.9 12.2 70.0 100.0 100.0


1h 6 93.3 82.5 104.2 10.3 80.0 100.0 100.0
2h 6 96.8 88.1 105.2 8.3 80.0 100.0 100.0
4h 6 89.2 72.8 105.6 15.6 60.0 100.0 95.0
24 h 6 64.0 36.5 91.6 22.2 40.0 100.0 60.0
48 h 6 46.0 5.9 86.1 32.3 21.0 100.0 35.0
After 48 h of treatment, the drop in FiO2 that was observed was not statistically significant (P>0.05).

The initial values of PaO2 in groups A1 and A2 did not differ maintained at a similar level of 53.7% (±23.2). After 48 h, a drop
statistically. A comparative analysis of both groups demonstrated of FiO2 to 40.1% (±19.6) was observed (Table 3).
that in group A1 the increase in PaO2 was statistically significantly In group A2, before treatment initiation (0 h) the value of FiO2
higher in comparison with group A2 after 1 h of treatment was 95.0% (±12.2), and after 1 h of treatment a drop in this
(P ¼ 0.0105). After several hours, the difference was still noted; parameter was achieved (to 93.3% (±10.3)), and after 2 h the
however, the value was not statistically significant. After 2, 4, 24 level was 96.8% (±8.3%). A drop to 89.2% (±15.6) was observed
and 48 h, there was a smaller difference in the value of PaO2 in after 4 h, and to 64.0% (±22.2) after 24 h and to 46.0% (±32.3)
groups A1 and A2 (Figure 3). after 48 h (Table 4).
The difference in the initial values of FiO2 in groups A1 and A2
Fraction of inspired oxygen was not statistically significant. When comparing both groups, it
The mean value of the initial (0 h) fraction of inspired oxygen was observed that after the first hour of treatment, the drop of FiO2
before treatment initiation was 95.7% (±11.3) in group A1 and in group A1 was faster than it was in group A2. After several hours,
95.0% (±12.2) in group A2. In group A1, a drop in this parameter a difference that was statistically insignificant was still noted. In
to 79.4% (±23.2) was achieved after 1 h, to 67.1% (±20.0) after group A1, a drop of FiO2 was statistically significantly higher than
2 h, to 57.1% (±16.6) after 4 h, and after 24 h this parameter was it was in comparison with group A2 after 2 h (P ¼ 0.0130) and

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S61

120
P =0.0130
P =0.0113
100

80
FiO2 A1

%
60
FiO2 A2
40

20

0
0h 1h 2h 4h 24h 48h

Figure 4 Fraction of inspired oxygen in groups A1 and A2.

Table 5 Mean airway pressure in group A1

MAP (cm H2O) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 7 14.3 10.5 18.2 4.2 8.1 20.6 13.6


1h 7 11.5 8.1 14.9 3.7 8.2 17.4 9.2
2h 7 9.8 6.0 13.4 4.0 4.6 15.6 10.8
4h 7 6.6 1.8 11.5 5.2 1.4 17.5 4.8
24 h 7 4.1 0.8 7.4 3.6 1.4 11.7 3.1
48 h 7 5.0 0.7 10.7 6.1 1.4 18.6 2.6
In this group of patients, a drop in MAP that was statistically significant took place between 0 and 24 h of treatment (P ¼ 0.0114).

Table 6 Mean airway pressure in group A2

MAP (cm H2O) N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 6 13.8 5.1 22.5 8.3 6.6 29.3 13.0


1h 6 13.2 8.0 18.4 5.0 7.8 21.7 12.5
2h 6 12.0 6.6 17.4 5.2 6.2 20.7 12.2
4h 6 10.9 6.8 14.9 3.9 7.2 17.3 10.5
24 h 6 8.0 3.4 12.6 3.7 4.3 13.9 7.2
48 h 6 4.2 0.8 7.6 2.8 2.2 8.9 3.5
In this group, the drop in MAP during 48 h of treatment was statistically significant between the 1st and 48th hour of treatment (P ¼ 0.0008).

after 4 h of treatment (P ¼ 0.0113). After 24 and 48 h, a smaller parameter was observed and it was 13.2 cm H2O (±5.0). After 2 h
difference in the value of FiO2 was observed in groups A1 and A2 of treatment, a drop to 12.0 cm H2O (±5.2) was observed. A
(Figure 4). further drop was observed after 4 h to 10.9 cm H2O (±3.9) and
after 24 h to 8.0 cm H2O (±3.7). This value decreased to 4.2 cm
MAP H2O (±2.8) after 48 h (Table 6).
The initial (0 h) MAP before treatment initiation was 14.3 cm H2O The difference between the initial value of MAP in
(±4.2) in group A1, and 13.8 cm H2O (±8.3) in group A2. groups A1 and A2 was not statistically significant. When
A drop in this parameter was observed after 1 h of treatment in comparing both groups, it was noticed that after the first hour
group A1 to 11.5 cm H2O (±3.7) with a further drop in this group of treatment, the drop in MAP was more rapid in group A1 than in
after 2 h to 9.8 cm H2O (±4.0) and after 4 h to 6.6 cm H2O group A2. After 1 and 2 h, the difference was still noted; however,
(±5.2). A further drop was observed after 24 h to 4.1 cm H2O it was statistically insignificant. In group A1, a drop in
(±3.6). After 48 h, a further but small increase to 5.0 cm H2O MAP was statistically significantly higher in comparison with
(±6.1) was observed (Table 5). group A2 after 24 h of treatment (P ¼ 0.0423) (Figure 5).
In group A2, before treatment initiation (0 h), the value of MAP After 48 h, the values of MAP in groups A1 and A2 were
was 13.8 cm H2O (±8.28). After 1 h of treatment, no drop in this similar.

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S62

16
14
12
10 P =0.0423
cmH2O

MAP A1
8 MAP A2
6
4
2
0
0h 1h 2h 4h 24h 48h

Figure 5 Mean airway pressure in group A.

Table 7 Oxygenation index in group A1

OI N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 7 29.8 18.3 41.4 12.5 21.1 56.6 25.0


1h 7 11.2 0.3 22.7 12.5 3.8 39.0 6.9
2h 7 10.0 4.9 15.1 5.5 3.8 18.1 10.0
4h 7 6.3 0.4 12.3 6.4 1.2 19.6 3.,9
24 h 7 2.7 0.7 4.8 2.2 0.4 6.7 2.8
48 h 7 5.0 3.4 13.5 9.1 0.4 25.5 1.7
A statistically significant drop took place between 0 and 1 h (P ¼ 0.0003), 0 and 4 h (P ¼ 0.0398) and 0 and 48 h of treatment (P ¼ 0.0001).

Table 8 Oxygenation index in group A2

OI N Mean value 95% confidence interval +95% confidence interval Standard deviation Minimum value Maximum value Median

0h 6 32.4 6.1 58.6 25.0 11.0 80.0 24.3


1h 6 28.8 16.1 41.4 12.0 11.0 41.2 33.9
2h 6 26.8 6.8 46.7 19.0 8.9 59.5 18.0
4h 6 22.2 2.9 41.5 18.4 4.8 53.0 18.2
24 h 6 10.4 0.4 20.5 8.1 1.9 22.1 9.0
48 h 6 5.7 5.6 17.0 9.1 0.7 22.0 1.8
In group A2, a drop in OI between 0 and 48 h of treatment was statistically significant (P ¼ 0.0011).

Oxygenation index was noted that after 1 h of treatment a drop in OI in group A1


The mean initial (0 h) value of the oxygenation index before was statistically significantly faster than it was in group A2
treatment initiation was 29.8 (±12.5) in group A1 and 32.4 (P ¼ 0.0222). In group A1, a drop in OI was also statistically
(±25.0) in group A2. In group A1, this value decreased after significantly higher when compared with group A2 after 2 h
several hours and was 11.2 (±12.5) after 1 h, 10.0 (±5.5) after (P ¼ 0.0455), 4 h (P ¼ 0.0455) and 24 h of treatment
2 h and 6.3 (±6.4) after 4 h. After 24 h, this value decreased to (P ¼ 0.0423). After 48 h, the values of OI in groups A1 and A2
2.7 (±2.2), and after 48 h to 5.0 (±9.1) (Table 7). were similar (Figure 6).
In group A2, a drop in this parameter was observed after 1 h to
28.8 (±12.1), after 2 h to 26.8 (±19.0), after 4 h to 22.2 Alveolar–arterial oxygen gradient
(±18.4), after 24 h to 10.4 (±8.1), and after 48 h a drop to 5.7 The initial values of AaDO2 before treatment initiation (0 h) were
(±9.1) (Table 8). 575.6 mm Hg (±91.0) for group A1 and 589.5 mm Hg (±95.8)
The difference in the initial value of OI in groups A1 and A2 was for group A2. In group A1, a drop in these values was observed after
not statistically significant. When comparing both groups, it 1 h of treatment to 423.7 mm Hg (±184.3), after 2 h to

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S63

35
P =0.0222
30 P =0.0455
25
P =0.0455
20 OI A1
P =0.0423
15 OI A2

10
5
0
0h 1h 2h 4h 24h 48h

Figure 6 Oxygenation index in group A.

P = 0.0036
700 P = 0.0106
600
500
mmHg

400 AaDO2 A1
AaDO2 A2
300
200
100
0
0h 1h 2h 4h 24h 48h

Figure 7 Alveolar–arterial oxygen gradient in group A.

364.2 mm Hg (±128.8), after 4 h to 305.3 mm Hg (±122.9), Duration of iNO treatment


after 24 h to 261.0 mm Hg (±160.9), and after 48 h to The mean duration of iNO treatment was 2.9 days (±1.5) in
178.3 (±144.0). group A1. In group A2, this value was higher and the duration was
A drop in AaDO2 during treatment was statistically significant 4 days (±1). This difference was not statistically significant
between 0 and 24 h (P ¼ 0.0024) and between 0 and 48 h of (P>0.05).
treatment (P ¼ 0.0021).
In group A2, after 1 h the value was 575.3 mm Hg (±88.2). Length of hospitalization
After 2 h, an increase to 581.2 mm Hg (±71.2) in this parameter In group A1, the length of hospitalization was 16.4 days (±5.4)
was observed. A drop was noted after 12 h to 506 mm Hg and in group A2 it was 19.8 days (±2.9). This difference was not
(±112.6), with a further drop after 24 h to 352.3 mm Hg statistically significant (P>0.05).
(±177.5) and after 48 h to 226.0 mm Hg (±239.8).
A drop in AaDO2 during the time of the treatment was statistically Complications
significant between 0 and the 48th hour of treatment (P ¼ 0.0017). Air leaks. The incidence of pneumothorax was analyzed. In
The difference between the initial values of AaDO2 in groups A1 group A1, no cases of pneumothorax were noted. This complication
and A2 was not statistically significant. When comparing both groups, was observed in two newborns from group A2. This value was not
it was noted that after 1 h of treatment a drop in AaDO2 was more statistically significant (P>0.05).
rapid in group A1 in than in group A2. Subsequently, the difference
was statistically significant (P ¼ 0.0036). After 4 h, a drop of AaDO2 Deaths. There were no deaths in group A1. There were two cases
in group A1 was also statistically significantly higher in comparison of deaths in group A2. This difference was not statistically
with group A2 (P ¼ 0.0106). After 24 and 48 h, a smaller difference significant (P>0.05).
was observed between the value of AaDO2 in groups A1 and A2. These
differences were statistically insignificant (Figure 7).
Discussion
Duration of mechanical ventilation Meconium in the airways and alveoli leads to inactivation of
In group A1, the mean time on mechanical ventilation was endogenously and exogenously replaced surfactant. Therefore, we
6.6 days (±2.6), and in group A2 it was 7.3 days (±1.7). hypothesized that SLL would improve the clearance of meconium
This difference was not statistically significant (P>0.05). from the airways while replacing the surfactant.7 Earlier studies on

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S64

the use of SLL in newborn piglets were published by Paranka et al.8 surfactant in SLL. Studies on these subjects were also published by
A solution of Survanta was used for lung lavage. Following this Wiswell et al.20 Their trial was focused on the use of Surfaxin, a
treatment, a rapid improvement in oxygenation was observed. new generation synthetic surfactant solution. The trial group
Similar experimental studies on the use of SLL in the treatment of comprised 22 newborns and the control group included 15
MAS were conducted at different centers by Dargaville et al.9 and newborns. The inclusion criteria were similar to the ones quoted
Ogawa and co-workers.10–12 above. A statistically significantly faster drop of OI after SLL was
The first clinical studies on SLL were conducted by observed in comparison with the control group.
Ogawa10 and Lam and Yeung.13 Ogawa administered as a lavage a The dose of surfactant used for lavage is still a subject for
surfactant solution with a concentration of 12 mg of phospholipids discussion. The smallest dose (8 ml per kg of body weight) of
in 1 ml of physiological salt. Earlier, Ohama et al.11 attempted to lavage solution was proposed by Wiswell et al.21 Others have
prove that a higher concentration is more effective. The volume of proposed a dose of 10 ml solution per kg of body weight.9,14 The
the solution was 5 ml per kg of body weight. A significant drop in most frequently suggested dose is a higher doseFfrom 15 15 to
OI and AaDO2 was achieved after 4 h of treatment. 20 ml kg1.10,17 In his earlier experimental trials, Ogawa10
Another attempt to define the most optimal dose of surfactant attempted to define the most optimal dose of the lavage solution
for lavage was undertaken by Chang et al.14 In group I, the lungs and he came to the conclusion that a dose of 10 ml kg1 is most
were washed with a surfactant solution with a concentration of effective.
5 mg ml1 of physiological salt. In group II, lavage was conducted Similarly, the concentration used in the solution is also a
with a surfactant solution with a concentration of 10 mg ml1. A subject of trials. Wiswell et al.21 proposed the lowest concentration
significant increase in PaO2 was observed within 24 h. It was noted of the drugF2.5 mg ml1 of physiological salt. The majority of
that both the first and the second doses of surfactant were effective. researchers propose a dose from 4 23 to 5 7,8,15,16 or 6 mg ml1.12
The next phase of study was a multi-center study by Ogawa10 Only Ogawa in their subsequent clinical studies proposed a higher
conducted in 11 centers. A comparative analysis of the group with dose of 12 mg ml1. On the other hand, Chang et al.14 proved that
SLL against the group without SLL demonstrated that a drop in the a dose of 5 mg ml1 is as effective as a dose of 10 mg ml1. In our
oxygenation parameter and an increase in the oxygen pressure trial, a concentration of 5 mg ml1 at a dose of 15 ml per kg of
after 180 min were significantly higher in the SLL group than in body weight was used.
the control group. All of the aforementioned clinical trials on the use of SLL in
Interesting results of clinical trials were presented by Lam MAS used saline as control groups. None compared bolus
and Yeung.13,15 Survanta, a natural surfactant solution with a treatments with SLL. Meister et al.19 conducted experimental trials
concentration of 5 mg phospholipids per ml of saline and a on a rabbit model in which they compared the results of surfactant
quantity of 15 ml per kg of body weight, was administered as a with bolus surfactant treatment. They also studied the differences
lavage. The results of the trial demonstrated that the average values in natural and synthetic surfactant treatments. A significant
of OI, MAP, AaDO2 and FiO2 were significantly and statistically increase in PaO2 was observed in both natural and synthetic
lower after 48 h of treatment. surfactants, SLL, in comparison with groups treated with bolus
Similar trial results were published in 2003 by Maroszyńska surfactant and a control group. The increase was observed after
et al.16,17 4 h of treatment.
The inclusion criteria used in our study did not differ much The group of patients with the most severe clinical course of
from the ones proposed by other researchers. Some of the MAS includes cases complicated by PPHN. In the majority of cases,
researchers quoted above considered the level of OI X20 as the studies on the use of iNO in the treatment of PPHN in term
only inclusion criterion.18 In our study, the criteria used were neonates with MAS confirm its effectiveness in oxygenation
similar to those used by Lam and Yeung.13,15 The only difference improvement.2,24 It was observed that, 30 min after the
was in the time for enrollment in the trial, which was extended to administration of NO, there was a visible oxygenation improvement
24 h (average time to include newborns transferred from a level II manifested by an increase in PaO2 and a drop in OI and AaDO2.
unit). We did not use OI and MAP as the inclusion criteria; only When conducting independent randomized trials, Chen and Kao
the value of FiO2X40% was used. observed a significant drop in OI after iNO treatment of PPHN in
A more extended time for the inclusion in the studyFup to MAS. Chen et al.1 noted improvement in 30 min of iNO, whereas
120 h was proposed only by Maroszyńska et al.16,17 The majority of Kao et al. observed a statistically significant drop in OI after 60 min
the quoted experimental and clinical trials as well as our own of iNO.25 The effectiveness of iNO in the treatment of PPHN in MAS
study studied the use of natural surfactant solutions in SLL. Earlier, was also proved by Gupta et al.26 A clinical trial on the use of iNO
Meister et al.19 compared the natural and the synthetic surfactants. in PPHN in MAS was also conducted by Hwang et al.27 After 1 h of
They proved that there are no statistically significant differences in iNO treatment, they observed a statistically significant drop of OI
the results achieved after administrations of either type of and MAP.

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S65

The neonates presented in our study who were diagnosed by complications and deaths when in comparison with the group that
an echocardiographic examination with PPHN in the course of a was not treated with iNO. Similarly, in our trial no statistically
severe MAS, were treated with surfactant prior to the administration significant differences were observed in the length of mechanical
of iNO. In this group of patients, the intention was to assess the ventilation and hospitalization. In the group with SLL, bolus
combined therapy as well as to answer the question whether it was surfactant and iNO, as well as no air leaks or deaths, were
more effective to use SLL followed by bolus surfactant or surfactant encountered. In the group with bolus surfactant and iNO, there
only prior to the administration of iNO. The initial dose of NO was were 2/6 cases of pneumothorax and 2/6 deaths. However, this
20 p.p.m. The treatment was conducted in the first 24 h of the difference was not statistically significant. In the first case,
newborn’s life. The average time of treatment initiation was pneumothorax was one of the causes of death. In the second case
9.7±7.4 h in group A1 and 11±6.4 h in group A2. Oxygenation, which was complicated by pneumothorax, the patient survived. The
ventilation and other selected clinical parameters were assessed. other death was due to a sudden stop in heart activity in the course
Rais-Bachrami et al.28 attained a faster improvement in of a severe circulatory failure.
oxygenation after surfactant administration followed by iNO. After During our study, there was no attempt to answer the important
20 min, they lowered the dose of NO and after 15 min they stopped question whether the use of bolus surfactant along with SLL will
the dose of iNO. prevent the development of PPHN and whether it may decrease the
The administration of bolus surfactant followed by iNO (group need for iNO use. Chang et al.14 attempted to address this question.
A2) had a positive influence on the improvement in oxygenation. They conducted clinical studies on neonates with MAS in whom OI
However, in a majority of cases, these results were not statistically was X20. Prior to iNO use, the airways were washed with a
significant. An increase in PaO2 was observed only after 2 h of surfactant solution with a concentration of 5 mg ml1 or
treatment, and a further increase was noted within 48 h; however, 10 mg ml1 of physiological salt. A statistically significant increase
this was not statistically significant. A similar tendency was in PaO2 was observed within 24 h; 5 of 12 neonates required iNO
observed in the values of FiO2 and OI. Within 48 h, a gradual treatment.
decrease in these parameters was observed; however, this drop was Therefore, the results of the trials conducted by researchers quoted
not statistically significant either. AaDO2 declined as well. A in this paper as well as by our own trial indicate that, in severe cases of
statistically significant drop was noted after 24 h and then after MAS complicated by PPHN, the administration of bolus surfactant or
48 h of treatment as well as in the time range between the SLL followed by bolus surfactant prior to iNO treatment improves
12th and 24th hour of treatment. A drop in MAP was observed oxygenation of the patient and may lead to a decrease both in the
only after 4 h of treatment and a further drop was noted until the severity of PPHN and in the duration of iNO treatment. However, no
48th hour of treatment; however, this drop was not statistically significant decrease in the duration of IMV, hospitalization or in the
significant. Substantially better results were observed in cases when, number of complications and deaths was observed.
prior to iNO, SLL was used followed by one dose of bolus surfactant.
There is a lack of information regarding the effectiveness of this
therapy. Investigators have used solely iNO treatment in severe Conclusions
cases of MAS complicated by PPHN. A drop in OI and an increase (1) The combined therapy: SLL followed by bolus surfactant, in
in PaO2 were observed after 30 min,24 after 60 min8 and after 24 h combination with iNO in the treatment of meconium
of iNO treatment29 in different studies. aspiration syndrome accompanied by PPHN, showed
In our studies, a significant increase in PaO2 was observed in significant improvement in oxygenation as well as a decrease
the group with SLL after 1 h. In the SLL group, a drop in OI and in mechanical ventilation parameters.
FiO2 was statistically significant after 24 and 48 h. When we (2) However, SLL followed by bolus surfactant as well as combined
compared SLL with the group without SLL, we noticed statistically therapy: SLL followed by bolus surfactant combined with
significant differences much sooner, that is, after the first, second iNO did not exert significant influence over the duration of
and fourth hours. mechanical ventilation, iNO treatment as well as the length
The drop in AaDO2 in the SLL group was substantial after 2 and of hospitalization and the number of complications.
4 h, with a further drop after 24 and 48 h. MAP also declined (3) On account of small number of the patients, these studies do
significantly in the SLL group after 24 h. However, when not have enough power to choose a particular combination of
comparing the two groups we observed a significant difference only therapies. There is a need for larger series to answer these
after 48 h. These results are similar to others who did not use SLL questions.
but were treated with only iNO. Therefore, we cannot conclude with
certainty whether the use of SLL increases the effectiveness of iNO. Disclosure
Similar to our findings, others did not find statistically J Gadzinowski has received consulting fees from Abbott. The remaining authors
significant differences in the length of time on IMV, number of have declared no financial interests.

Journal of Perinatology
Treatment of MAS with PPHN using combined therapy
J Gadzinowski et al
S66

References 16 Maroszyńska I, Stoiñska B, Czop J, Dygas T, Gadzinowski J. Surfactant lavage as an


effective treatment in newborn with meconium aspiration syndrome. Arch Perinat Med
1 Park KH, Bae CW, Chung SJ. In vitro effect of meconium on the physical surface
2003; 2: 15–21.
properties and morphology of oxygenous pulmonary surfactant. J Korean Med Sci
17 Maroszyńska I, Wilkowski J, Czop J, Dygas T. Bronchoalveolar lavage with saline
1996; 11: 429–436.
surfactant solution in the treatment of neonates with severe respiratory failure due to
2 Gadzinowski J, Moœcicka A, Kowalska K. Zespół aspiracji smółkiFwspółczesne
MAS, PIE or BPD. Med Sci Monit 2003; 9(Suppl 5): 22–28.
metody leczenia. Kl Perinat i Ginekol 1998; 1: 153–160.
18 Davey AM, Becker JC, Davis JC. Meconium aspiration syndrome. physiological
3 Alekszewicz-Baranowska J, Nadciœnienie płucne W. Echokardiografia wad
and inflammatory changes in a newborn piglet model. Pediatr Pulmonol 1993;
wrodzonych serca. In: Alekszewicz-Baranowska J (Pod red). Wyd Med MAKmed:
16: 300.
Gdañsk 2000; 165–169.
19 Meister J, Balaraman V, Ramirez M, Uyehara CF, Killeen J, Ku T et al. Lavage
4 Kawalec W. Diagnostyka nieinwazyjna ukadu kra żenia. W: Kardiologia okresu
‘ administration of dilute surfactant in a piglet model of meconium aspiration. Lung
noworodkowego. Kawalec W (Pod red). PZWL Biblioteka Pediatry: Warszawa 1998;
2004; 182: 227–240.
38–50.
20 Wiswell TE, Peabody SS, Davis JM, Slayter MV, Bent RC, Merritt TA. Surfactant therapy
5 Kawalec W, Wołoszczuk-Ge˛bicka B (eds). kra żenia przejœciowe i przetrwałe and high frequency jet ventilation in management of a piglet model of meconium

nadciœnienie płucne. PZWL Biblioteka Pediatry: Warszawa 1998; 73–84. aspiration syndrome. Pediatr Res 1994; 36: 494–500.
6 Rydlewska-Sadowska W. Echokardiografia kliniczna. BIK Warszawa 1991; 11: 135–136. 21 Wiswell TE, Rebecca C, Bent M. Meconium staining and the meconium aspiration
7 Kurtis PS. Meconium aspiration. Pediatrics 2000; 106: 867. syndrome. Pediatr Clin North Am 1993; 40: 955–977.
8 Paranka MS, Walsh WF, Stancombe BB. Surfactant lavage in a piglet model of 22 Castellheim A, Lindenskov PH, Pharo A, Aamodt G, Saugstad OD, Mollnes TE.
meconium aspiration syndrome. Pediatr Res 1992; 31: 625–628. Meconium aspiration syndrome induces complement-associated systemic inflammatory
9 Dargaville PA, Mills JF, Headley BM, Chan Y, Coleman L, Loughnan PM et al. response in newborn piglets. Scand J Immunol 2005; 61: 217–225.
Therapeutic lung lavage in the piglet model of meconium aspiration syndrome. 23 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obstet Gynecol
Am J Respir Crit Care Med 2003; 168: 456–463. 1975; 122: 767.
10 Ogawa Y. Bronchial lavage with surfactant solution for the treatment of meconium 24 Marini G, Berefield ES, Carlo W. The role of nitric oxide in the treatment of neonatal
aspiration syndrome. In: Lucey J (ed) Conference Paper at Ross Special Conference pulmonary hypertension. Curr Opin Pediatr 1996; 8: 118.
Hot Topics in Neonatology 1997; 259–264. 25 Ko SY, Chang YS, Park WS. Clinical response to inhaled nitric oxide in persistent
11 Ohama Y, Itakura Y, Koyama N, Eguchi H, Ogawa Y. Effect of surfactant lavage pulmonary hypertension of the newborn. J Korean Med Sci 1998; 13: 500–506.
in a rabbit model of meconium aspiration syndrome. Acta Paediatr Jpn 1994; 26 Gupta V, Bhatia BD, Mishra OP. Meconium stained amniotic fluid antenatal
36: 236. intrapartum and neonatal attributes. Indian Pediatr 1996; 33: 293.
12 Ohama Y, Ogawa Y. Treatment of meconium aspiration syndrome with surfactant 27 Hwang SJ, Lee KH, Hwang JH. Factors affecting the response to inhaled nitric oxide
lavage in an experimental rabbit model. Pediatr Pulmonol 1999; 28: 18–23. therapy in persistent pulmonary hypertension of the newborn infants. Yonsei Med J
13 Lam BC, Yeung CY. Surfactant lavage for meconium aspiration syndrome. In: Lucey J 2004; 45: 49–55.
(ed) Conference paper at Ross Special Conference Hot Topics in Neonatology, 28 Rais-Bachrami K, Rivera O, Seale WR, Short BL. Effect of nitric oxide and high-
Washington 1997; 252–256. frequency oscillatory ventilation in meconium aspiration syndrome. Pediatr Crit Care
14 Chang HY, Hsu CH, Kao HA, Hung HY, Chang JH, Peng CC et al. Treatment of severe Med 2000; 1: 66–160.
meconium aspiration syndrome with dilute surfactant lavage. J Formos Med Assoc 29 Gupta A, Rastogi S, Sahni R, Bhutada A, Bateman D, Rastogi D et al. Inhaled
2003; 102: 326–330. nitric oxide and gentle ventilation in the treatment of pulmonary hypertension
15 Lam BC, Yeung CY. Surfactant lavage for meconium aspiration syndrome: a pilot of the newbornFa single-center, 5-year experience. J Perinatol 2002; 22:
study. Pediatrics 1999; 103(5 Part 1): 1014–1018. 435–441.

Journal of Perinatology
Journal of Perinatology (2008) 28, S67–S71
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Nitric oxide and beyond: new insights and therapies for
pulmonary hypertension
RH Steinhorn
Department of Pediatrics, Division of Neonatology, Children’s Memorial Hospital, Northwestern University’s Feinberg School of
Medicine, Chicago, IL, USA

of PPHN, and when present, it can contribute significantly to its


Persistent pulmonary hypertension of the newborn (PPHN) contributes morbidity and mortality. Newborns with hypoxemic respiratory
significantly to the morbidity and mortality associated with meconium failure and/or PPHN are at risk for numerous complications
aspiration syndrome. This review article discusses new insights into the including death, neurological injury and other morbidities.
vascular abnormalities that are associated with PPHN, including the recent Persistent pulmonary hypertension of the newborn can
recognition of the importance of oxidant stress in its pathogenesis. Recent generally be characterized as one of three types: (1) the abnormally
data are presented showing that treatment with high oxygen concentrations constricted pulmonary vasculature due to lung parenchymal
may increase production of oxygen free radicals. The rationale for the use of diseases, such as MAS, respiratory distress syndrome or pneumonia;
inhaled nitric oxide, and strategies for enhancing nitric oxide signaling are (2) the lung with normal parenchyma and remodeled pulmonary
discussed. Finally, the rationale for new treatment approaches is reviewed, vasculature, also known as idiopathic PPHN; or (3) the hypoplastic
including inhibition of cyclic guanosine monophosphate-specific vasculature as seen in congenital diaphragmatic hernia. The most
phosphodiesterases and scavengers of reactive oxygen species. common cause of PPHN is MAS. Infants with MAS will typically fall
Journal of Perinatology (2008) 28, S67–S71; doi:10.1038/jp.2008.158 into the first or second categories, and the most severe cases are
probably affected by both parenchymal and vascular disease.
Understanding the pathophysiology of the abnormally
Introduction
remodeled pulmonary vasculature is of utmost importance in
Neonatal respiratory failure affects 2% of all live births and is directing therapy. As it is not feasible to study the remodeling
responsible for a substantial proportion of neonatal mortality. process in the human infant, much of our current understanding
Although preterm infants are at higher risk of respiratory failure, is derived from animal models. One cause of idiopathic PPHN is
term and near-term infants account for one-third of the cases.1 constriction of the fetal ductus arteriosus in utero because of
A better understanding of the pathophysiology of hypoxemic exposure to nonsteroidal anti-inflammatory drugs during the third
respiratory failure is needed to develop more specific and effective trimester. Ductal constriction or ligation can be surgically
therapies. This review will focus on recent progress in our performed in utero in lambs, leading to rapid antenatal
understanding of pulmonary hypertension, which is commonly remodeling of the pulmonary vasculature. Findings in PPHN lambs
associated with the severe respiratory failure that accompanies are similar to those observed in human infants, including
meconium aspiration syndrome (MAS). increased fetal pulmonary artery pressure, pulmonary vascular
remodeling and profound hypoxemia after birth.
Pathophysiology of PPHN On the basis of work from animal models, there is strong
Shortly after birth, the fetus normally undergoes a rapid evidence that disruptions of the nitric oxide (NO)-cyclic guanosine
cardiopulmonary transition to meet the new demands of monophosphate (cGMP), prostacyclin-cyclic adenosine
extrauterine life. However, if pulmonary vascular resistance does monophosphate (cAMP) and endothelin signaling pathways play
not fall, pulmonary blood flow cannot increase, and the result is an important role in the vascular abnormalities associated with
hypoxemic respiratory failure or persistent pulmonary hypertension PPHN.2 The NO-cGMP pathway has been a topic of particularly
of the newborn (PPHN). The incidence of severe PPHN is estimated intense investigation over the past decade. Decreased expression
at 0.2% of live-born term infants. MAS is the most common cause and activity of endothelial NO synthase have been documented in
the PPHN lamb model,3 and decreased endothelial NO synthase
Correspondence: Dr RH Steinhorn, Department of Pediatrics, Division of Neonatology, expression has also been reported in umbilical venous endothelial
Children’s Memorial Hospital, Northwestern University’s Feinberg School of Medicine, 2300
Children’s Plaza, Neonatology, Box 45, Chicago, IL 60614, USA. cell cultures from human infants with meconium staining who
E-mail: r-steinhorn@northwestern.edu develop PPHN.4 These important findings were rapidly followed by
Nitric oxide and beyond
RH Steinhorn
S68

clinical testing of inhaled NO (iNO) as a therapy for hypoxemic AA


L-Arginine COX-1
respiratory failure and PPHN.
PGH2 Endothelial
NOS
PGIS Cell
NO PGI2
Inhaled nitric oxide
The primary aim of PPHN therapy is selective pulmonary
vasodilatation. Inhaled NO appears to be well suited for this effect: Guanylate Cyclase Adenylate Cyclase

it is a rapid and potent vasodilator, and because NO is a small gas GTP ATP Smooth
cGMP cAMP Muscle
molecule, it can be delivered through a ventilator directly to 5´-GMP
AMP
Cell
airspaces approximating the pulmonary vascular bed. Once in the PDE5 PDE3
VASODILATION
blood stream, NO binds avidly to hemoglobin, limiting its systemic
vascular activity and increasing its selectivity for the pulmonary
circulation. Sildenafil Milrinone
Large placebo-controlled trials provided clear evidence that iNO Figure 1 Nitric oxide (NO) and prostacyclin signaling pathways in regulation of
significantly decreases the need for extracorporeal membrane pulmonary vascular tone. NO is synthesized by NO synthase (NOS) from the
oxygenation (ECMO) support in newborns with PPHN.5,6 However, terminal nitrogen of L-arginine. NO stimulates soluble guanylate cyclase (sGC) to
it is important to note that up to 40% of infants will not improve increase intracellular cGMP. PGH2 is an arachidonic acid (AA) metabolite formed
by cyclooxygenase (COX-1) and prostacyclin synthase (PGIS) in the vascular
oxygenation or maintain a response to iNO, and iNO did not
endothelium. Prostacyclin (PGI2) stimulates adenylate cyclase in vascular smooth
reduce mortality or length of hospitalization. In addition, follow-up muscle cells, which increases intracellular cAMP. Both cGMP and cAMP indirectly
studies for 12 to 24 months indicate that iNO does not significantly decrease free cytosolic calcium, resulting in smooth muscle relaxation. Specific
alter the incidence of chronic lung disease or neurodevelopmental phosphodiesterases hydrolyze cGMP and cAMP, thus regulating the intensity and
impairment.7,8 This is an interesting and important observation duration of their vascular effects. Inhibition of these phosphodiesterases with
agents such as sildenafil and milrinone may enhance pulmonary vasodilation.
that may indicate that the underlying disease is associated with
early neurological injury. Finally, Konduri et al.9,10 determined
that starting iNO earlier in the disease course (for an oxygenation New insights into PPHN pathophysiology
index of 15 to 25) did not decrease the incidence of ECMO and/or As NO is not universally effective, there has been considerable
death or improve other patient outcomes, including the incidence interest in better understanding the biochemical pathways that
of neurodevelopmental impairment. regulate pulmonary vasoconstriction and remodeling in PPHN. For
Following the introduction of high-frequency ventilation (HFV), instance, NO mediates vasodilatation by stimulating soluble
surfactant and iNO in the early 1990s, the patient demographic of guanylate cyclase in vascular smooth muscle cells, which then
neonatal support with ECMO has changed. Data from the large converts guanosine triphosphate to cGMP (Figure 1). cGMP is the
registry maintained by the Extracorporeal Life Support central and critical second messenger that regulates contractility of
Organization indicate that the use of these therapies has increased the smooth muscle cell by modulating the activity of
steadily over the past 10 years, accompanied by a greater than 40% cGMP-dependent kinases, phosphodiesterases and ion channels. The
reduction in the number of neonates cannulated for ECMO. cGMP-dependent or type 5 phosphodiesterase is potentially
However, as overall ECMO survival has diminished over the same important, as it can downregulate cGMP concentrations by
time period, some physicians have speculated that these new degrading cGMP to the inactive 50 -GMP. Therefore, there are critical
treatment modalities may delay ECMO cannulation and have a points in the pathway downstream of NO production that serve as
negative effect on mortality and morbidity in those infants that attractive targets for manipulating cellular cGMP concentrations.
continue to require extracorporeal support. Therefore, we recently For example, expression and activity of soluble guanylate cyclase are
examined data from the Extracorporeal Life Support Organization decreased in the abnormally remodeled pulmonary vessels of the
registry between 1996 and 2003. We found that NO, HFV and PPHN lamb model, which could potentially diminish responses to
surfactant use were not associated with any adverse outcomes both endogenous and exogenous NO. This finding would indicate
during ECMO, including increased hours on ECMO or that new compounds that directly stimulate sGC at an NO-
increased time to extubation.11 Furthermore, both surfactant and independent but heme-dependent site may be helpful, a hypothesis
NO use were associated with lower ECMO mortality, and that appears to be promising in preclinical testing.12 Another
NO use was associated with a decreased risk of cardiac potential cause for low cGMP concentrations would be increased
arrest before cannulation. As ECMO is a proven therapy for expression and/or activity of cGMP-specific phosphodiesterases,
severe respiratory failure, it is reassuring that these new which could then be manipulated through use of specific inhibitors.
therapies have not had a negative impact on the most severely Several new lines of evidence now indicate that oxidant stress is
affected infants. important in the pathogenesis of PPHN. An increase in reactive

Journal of Perinatology
Nitric oxide and beyond
RH Steinhorn
S69

SOD H2O2
O2
Arachidonic Acid
O2.- and PUFAs

Isoprostanes

Contractility

Peroxynitrite

NO

Pulmonary Artery

Alveolus

Figure 2 Hypothesized role of reactive oxygen species (ROS) and their metabolites in mediating increased pulmonary arterial contractility following exposure to 100%
oxygen. Exposure to high oxygen concentrations results in the formation of superoxide anions (O 2 ). Superoxide anions combine with nitric oxide (NO) to form
peroxynitrite, a potent pulmonary vasoconstrictor. Superoxide dismutase (SOD) enzyme converts superoxide anions to hydrogen peroxide (H2O2), also a pulmonary
vasoconstrictor. When membrane lipids (arachidonic acid and polyunsaturated fatty acids (PUFA)) are exposed to ROS, such as superoxide anions and hydrogen peroxide
or peroxynitrite, a variety of isoprostanes are formed. Isoprostanes are known constrictors of pulmonary arteries. Adapted from Lakshminrusimha et al.16 with permission.

oxygen species (ROS) such as superoxide and hydrogen peroxide in with the potential to produce vasoconstriction, cytotoxicity and
the smooth muscle and adventitia of pulmonary arteries has been damage to surfactant proteins and lipids (Figure 2). New data
demonstrated in the PPHN lamb model.13,14 Possible sources of indicate that even brief periods of ventilation with 100% O2
elevated concentrations of ROS include increased expression and (30 min) are sufficient to increase reactivity of pulmonary vessels
activity of NADPH oxidase and a reduction in superoxide dismutase in normal lambs16 and to diminish the response of the lung to
(SOD) activity. PPHN lambs also demonstrate diminished binding endogenous and exogenous NO.17
of the chaperone protein, heat shock protein 90, to endothelial NO
synthase.15 Decreased heat shock protein 90–endothelial NO
synthase interactions lead to an ‘uncoupling’ of NOS activity, Alternative and emerging pulmonary vasodilators
which results in decreased synthesis of NO and increased An improved knowledge of the biochemical abnormalities
superoxide production. Once present in the lung, elevated responsible for refractory PPHN is leading to a growing list of
concentrations of ROS are believed to play a role in vascular promising new therapeutic strategies. Many investigators are
smooth muscle cell proliferation in PPHN, as well as abnormal seeking to enhance cGMP-mediated vasodilation through the use of
vascular reactivity. cGMP-specific phosphodiesterase inhibitors, direct soluble
Finally, current therapeutic practices may have an effect on guanylate cyclase activators, scavengers of ROS as well as
pulmonary vascular reactivity and remodeling. In particular, the manipulation of the cAMP pathway.
use of oxygen has recently become controversial in numerous Similar to cGMP, cAMP also stimulates vasodilatation
settings. While oxygen is a pulmonary vasodilator, the extreme (Figure 1). One potential approach that might take advantage of
hyperoxia routinely used in PPHN management may be toxic to this mechanism is using milrinone to inhibit PDE3, the
the developing lung by the formation of ROS. Although hyperoxic phosphodiesterase that metabolizes cAMP. Milrinone has been
ventilation is a mainstay of the treatment of PPHN, we know shown to decrease pulmonary artery pressure and resistance and to
surprisingly little about what oxygen concentrations will maximize act additively with iNO in animal studies. Recent reports indicate
benefits and minimize risk. Superoxide may react with arachidonic that it may decrease rebound pulmonary hypertension after
acid to increase concentrations of isoprostanes and may also discontinuation of iNO and may enhance pulmonary vasodilation
combine with NO to form peroxynitrite. Both are potent oxidants of infants refractory to iNO.18 Prostacyclin (PGI2) stimulates

Journal of Perinatology
Nitric oxide and beyond
RH Steinhorn
S70

Figure 3 Superoxide dismutase (SOD) and nitric oxide (NO) improve the arterial-to-alveolar oxygen (a/A) ratio to a similar degree. The a/A ratio of four groups of
lambs with persistent pulmonary hypertension of the newborn (PPHN) ventilated with 100% oxygen alone (100% O2), 100% oxygen with rhSOD 5 mg kg1 administered
at birth (100% O2 SOD), 100% oxygen with rhSOD 5 mg kg1 administered at 4 h of life (100% O2 4 h delay SOD) and 100% oxygen and 20 p.p.m. of inhaled NO
(iNO; 100%O2 NO). PPHN lambs ventilated with 100% oxygen were critically ill, and two of these lambs died at approximately 10 and 16 h of age (shown by arrows).
The dashed line beyond these points represents mean±s.e.m. for the remaining lambs. *P<0.05 compared with 100% oxygen-alone group. Adapted from
Lakshminrusimha et al.25 with permission.

membrane bound adenylate cyclase, increases cAMP and inhibits with pulmonary hypertension, and data analysis is nearing
pulmonary artery smooth muscle cell proliferation in vitro. completion.
Although the use of systemic infusions of PGI2 may be limited by New laboratory studies indicate that scavengers of ROS such as
systemic hypotension, inhaled PGI2 has been shown to have SOD may augment responsiveness to iNO. As described above,
vasodilator effects limited to the pulmonary circulation. Reports in increased production of superoxide is noted in experimental models
children have been positive, but to date there have been few reports of PPHN. As iNO is usually delivered with high concentrations of
of inhaled PGI2 use in neonates with PPHN.19 It is most likely that oxygen, there is the potential for enhanced production of additional
further investigations will focus on preparations specifically oxidants such as peroxynitrite. Superoxide dismutase scavenges
designed for inhalation, such as iloprost. and converts superoxide radical to hydrogen peroxide, which is
There has been particular interest in the inhibition of subsequently converted to water by the enzyme catalase.
cGMP-metabolizing phosphodiesterase activity, which would Administration of recombinant human SOD (rhSOD) has been
theoretically increase cGMP concentrations and result in tested in preterm infants without adverse effects and with trends
pulmonary vasodilation and/or increased efficacy of iNO toward decreased pulmonary morbidity. In lambs with pulmonary
(Figure 1). On the basis of clinical trials, sildenafil, a potent and hypertension, rhSOD was found to dilate the pulmonary circulation
highly specific phosphodiesterase inhibitor, has recently been and enhance responsiveness to inhaled NO.24 A recent study
approved by the Food and Drugs Administration for the treatment examined the effects of rhSOD on oxygenation over a 24-h period
of pulmonary hypertension in adults. In lambs with experimental in ventilated PPHN lambs.25 The results showed that a single dose
pulmonary hypertension, both enteric and aerosolized sildenafil of rhSOD by itself improved oxygenation to a degree that was
dilate the pulmonary vasculature and augment the pulmonary similar to iNO (Figure 3). Furthermore, rhSOD treatment appeared
vascular response to iNO. Intravenous sildenafil was found to be a to block formation of oxidants such as peroxynitrite and
selective pulmonary vasodilator with efficacy equivalent to inhaled isoprostanes. Thus, an antioxidant therapeutic approach may
NO in a piglet model of meconium aspiration, although have multiple beneficial effects: scavenging superoxide may
hypotension and worsening oxygenation resulted when it was used make both endogenous and inhaled NO more available to
in combination with iNO.20,21 Sildenafil may attenuate rebound stimulate vasodilatation and may also reduce oxidative
pulmonary hypertension after withdrawal of iNO in newborn and stress and limit lung injury. It is hoped that human trials will
pediatric patients.22 Use of sildenafil in PPHN has been limited by begin soon.
its availability only as an enteric form, although a recent report
indicates that it improved oxygenation and survival in Disclosure
human infants with PPHN compared with placebo.23 A pilot RH Steinhorn has received consulting fees from Ikaria. Grant support from the
trial of intravenous sildenafil was recently conducted in newborns NIH (HL 54705) supported the research presented in the paper.

Journal of Perinatology
Nitric oxide and beyond
RH Steinhorn
S71

References 13 Brennan LA, Steinhorn RH, Wedgwood S, Mata-Greenwood E, Roark EA, Russell JA
1 Angus DC, Linde-Swirble WT, Clermont G, Griffin MF, Clark RH. Epidemiology of et al. Increased superoxide generation is associated with pulmonary hypertension in
fetal lambs. A role for NADPH oxidase. Circ Res 2003; 92: 683–691.
neonatal respiratory failure in the United States. Am J Resp Crit Care Med 2001; 164:
1154–1160. 14 Wedgwood S, Steinhorn RH, Bunderson M, Wilham J, Lakshminrusimha S, Brennan LA
et al. Increased hydrogen peroxide downregulates soluble guanylate cyclase in the
2 Farrow KN, Fliman P, Steinhorn RH. The diseases treated with ECMO: focus on PPHN.
lungs of lambs with persistent pulmonary hypertension of the newborn. Am J Physiol
Semin Perinatol 2005; 29: 8–14.
Lung Cell Mol Physiol 2005; 289: L660–L666.
3 Shaul PW, Yuhanna IS, German Z, Chen Z, Steinhorn RH, Morin III FC.
15 Konduri GG, Ou J, Shi Y, Pritchard Jr KA. Decreased association of HSP90 impairs
Pulmonary endothelial NO synthase gene expression is decreased in fetal lambs
endothelial nitric oxide synthase in fetal lambs with persistent pulmonary
with pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 1997; 272:
hypertension. Am J Physiol Heart Circ Physiol 2003; 285: H204–H211.
L1005–L1012.
16 Lakshminrusimha S, Russell JA, Steinhorn RH, Ryan RM, Gugino SF, Morin III FC
4 Villaneuva ME, Zaher FM, Svinarich DM, Konduri GG. Decreased gene expression of
et al. Pulmonary arterial contractility in neonatal lambs increases with 100% oxygen
endothelial nitric oxide synthase in newborns with persistent pulmonary hypertension.
resuscitation. Pediatr Res 2006; 59: 137–141.
Pediatr Res 1998; 44: 338–343.
17 Lakshminrusimha S, Russell JA, Steinhorn RH, Swartz DD, Ryan RM, Gugino SF et al.
5 Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in full-term and
Pulmonary hemodynamics in neonatal lambs resuscitated with 21, 50, and 100%
nearly full-term infants with hypoxic respiratory failure. New Engl J Med 1997; 336:
oxygen. Pediatr Res 2007; 62(3): 313–318.
597–604.
18 McNamara PJ, Laique F, Muang-In S, Whyte HE. Milrinone improves oxygenation in
6 Clark RH, Kueser TJ, Walker MW, Southgate WM, Huckaby JL, Perez JA et al. Low dose neonates with severe persistent pulmonary hypertension of the newborn. J Crit Care
nitric oxide therapy for persistent pulmonary hypertension of the newborn. N Engl J 2006; 21: 217–222.
Med 2000; 342: 469–474. 19 Kelly LK, Porta NF, Goodman DM, Carroll CL, Steinhorn RH. Inhaled prostacyclin for
7 Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in term and near- term infants with persistent pulmonary hypertension refractory to inhaled nitric oxide.
term infants: neurodevelopmental follow-up of the neonatal inhaled nitric oxide study J Pediatr 2002; 141: 830–832.
group (NINOS). J Pediatr 2000; 136: 611–617. 20 Shekerdemian L, Ravn H, Penny D. Intravenous sildenafil lowers pulmonary vascular
8 Clark RH, Huckaby JL, Kueser TJ, Walker MW, Southgate WM, Perez JA et al. Low-dose resistance in a model of neonatal pulmonary hypertension. Am J Resp Crit Care Med
nitric oxide therapy for persistent pulmonary hypertension: 1-year follow-up. 2002; 165: 1098–2002.
J Perinatol 2003; 23: 300–303. 21 Shekerdemian LS, Ravn HB, Penny DJ. Interaction between inhaled nitric oxide and
9 Konduri GG, Solimani A, Sokol GM, Singer J, Ehrenkranz RA, Singhal N et al. A intravenous sildenafil in a porcine model of meconium aspiration syndrome. Pediatr
randomized trial of early versus standard inhaled nitric oxide therapy in term and Res 2004; 55: 413–418.
near-term newborn infants with hypoxic respiratory failure. Pediatrics 2004; 113: 22 Atz AM, Wessel DL. Sildenafil ameliorates effects of inhaled nitric oxide withdrawal.
559–564. Anesthesiology 1999; 91: 307–310.
10 Konduri GG, Vohr B, Robertson C, Sokol GM, Solimano A, Singer J et al. Early inhaled 23 Baquero H, Soliz A, Neira F, Venegas ME, Sola A. Oral sildenafil in infants with
nitric oxide therapy for term and near-term newborn infants with hypoxic respiratory persistent pulmonary hypertension of the newborn: a pilot randomized blinded study.
failure: neurodevelopmental follow-up. J Pediatr 2007; 150: 235–240. Pediatrics 2006; 117: 1077–1083.
11 Fliman PJ, deRegnier RA, Kinsella JP, Reynolds M, Rankin LL, Steinhorn RH. Neonatal 24 Steinhorn RH, Albert G, Swartz DD, Russell JA, Levine CR, Davis JM. Recombinant
extracorporeal life support: impact of new therapies on survival. J Pediatr 2006; 148: human superoxide dismutase enhances the effect of inhaled nitric oxide in persistent
595–599. pulmonary hypertension. Am Rev Resp Crit Care Med 2001; 164: 834–839.
12 Deruelle P, Grover TR, Abman SH. Pulmonary vascular effects of nitric oxide-cGMP 25 Lakshminrusimha S, Russell JA, Wedgwood S, Gugino SF, Kazzaz JA, Davis JM et al.
augmentation in a model of chronic pulmonary hypertension in fetal and neonatal Superoxide dismutase improves oxygenation and reduces oxidation in neonatal
sheep. Am J Physiol Lung Cell Mol Physiol 2005; 289: L798–L806. pulmonary hypertension. Am J Respir Crit Care Med 2006; 174: 1370–1377.

Journal of Perinatology
Journal of Perinatology (2008) 28, S72–S78
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Pharmacotherapy for meconium aspiration
A Asad and R Bhat
Division of Neonatology, Department of Pediatrics, University of Illinois at Medical Center, Chicago, IL, USA

with moderate-to-severe MAS. Figure 1 shows the schema of our


In this article we have attempted to review the current pharmacological presentation.
treatment options for infants with meconium aspiration syndrome with or The treatment options can be divided into general or specific.
without persistent pulmonary hypertension. These treatments include The general treatment options are the optimization of ventilation,
ventilatory support, surfactant treatment and inhaled nitric oxide (INO), in sedation and alkalanization. The more specific pharmacological
addition to older and newer pharmacological treatments. These include therapies are aimed at reducing hypoxia with the use of pulmonary
sedatives, muscle relaxants, alkali infusion, antibiotics and the newer vasodilators (inhaled nitric oxide (INO), oral sildenafil and
vasodilators. Many aspects of treatment, including ventilatory care, bosentan), and the lung injury by anti-inflammatory agents,
surfactant treatment and the use of INO, are reviewed in great detail in this surfactant lavage or replacement.
issue. On the other hand, many newer pharmacological modalities of
treatment described here have not been evaluated with randomized control
trials. We have given an overview of these emerging therapies.
Journal of Perinatology (2008) 28, S72–S78; doi:10.1038/jp.2008.160 General management
In all newborns with evidence of severe respiratory distress
handling is minimized to prevent worsening of gas exchange due
to agitation. Sedation and analgesia are used frequently in infants
with MAS and persistent pulmonary hypertension to alleviate pain
and discomfort that may lead to hypoxia and right-to-left shunting.
Introduction Opioids particularly morphine or fentanyl with midazolam are
Meconium aspiration syndrome (MAS) is defined as the presence of frequently used across the world (77 to 100%). Major side effects of
respiratory distress and hypoxemia associated with the presence of these medications include hypotension, urinary retention,
meconium during or just before delivery. Meconium-stained development of tolerance and withdrawal symptoms. Opioids can
amniotic fluid is a relatively common occurrence seen in be given either intermittently or as a continuous drip.
approximately 10 to 15% of deliveries and approximately 5% of Pharmacokinetics and metabolism of these drugs are well
these develop true MAS.1 The most severe cases require assisted studied4,5 but their impact on long-term outcome has not been
ventilation for greater than 48 h and are often associated with reported in term infants. Infact, no RCTs are available in term
persistent pulmonary hypertension. Several recent publications and infants to support its routine use.
other reviews in this issue have described in detail the
pathophysiology and different aspects of the management. Our goal Muscle relaxants
is to review the pharmacological management of MAS. Prior to 2000, depolarizing muscle relaxants were widely used
along with opioids to decrease agitation and subsequent hypoxic
Treatment strategies: the past, present and future episodes in ventilated infants. Pancuronium and Vecuronium
Various treatment modalities including intrapartum (perineal) and (0.1 mg kg1 h1) are the two most commonly used muscle
post-delivery oral suctioning were the routine until recently but two relaxants in neonatal intensive care unit. The benefits of
recent randomized controlled trials (RCTs) have shown that none neuromuscular blockade include improved oxygenation, decreased
of these techniques have decreased the incidence of MAS.2,3 These oxygen consumption and decreased accidental extubations.
approaches are well covered by different authors in this special However, a prospective multicenter observational study by
issue. We will focus primarily on different treatments in the Walsh-Sukys et al.6 showed considerable variations in muscle
management of infants admitted to neonatal intensive care unit relaxant use between centers (33 to 98%). Use of muscle relaxants
was also associated with increased mortality (Odds ratio: 1.95,
Correspondence: Dr R Bhat, Department of Pediatrics, M/C 856, University of Illinois at
Chicago Medical Center, 840 S Wood Street, Chicago, IL 60612, USA. confidence interval: 0.74, 5.18). In their series, 41% (total
E-mail: ramabhat@uic.edu N ¼ 385) of the cases had MAS as the primary diagnosis. The side
Pharmacotherapy for MAS
A Asad and R Bhat
S73

General
• Sedation Analgesia
• Muscle Relaxants
• Alkalinization

Infection & Inflammation Pharmacotherapy Pulmonary hypertension


of
• Antibiotics MAS • Vasodilators
• Anti-inflammatory agents • Hemodynamic stabilizers

Pulmonary care
• Surfactant treatment
• Surfactant lavage
• Ventilator support

Figure 1 Management of MAS.

effects of nondepolarizing agents include cardiovascular effects oxygenation. It is noted that these drugs were not studied
from histamine release, though this is less common in neonates independently of each other. Hypotension is frequently secondary to
and also tachycardia (vagolytic effect). Neuromuscular blockade heavy sedation and the use of high mean airway pressure.
can also mask seizures especially in infants depressed at birth. No Dopamine and dobutamine are the two most frequently used
randomized control studies are available at this time. inotropes in neonates. High doses of dopamine
Neonatologists are urged to consider the risks and benefits of (>10 mg kg1 min1) on its own can also contribute to
neuromuscular blockade prior to their use. pulmonary vasoconstriction.12

Alkalinization Pulmonary vasodilators


Increasing the pH from 7.45 to 7.5 either by hyperventilation or by Regulation of pulmonary vascular tone is a dynamic process and
sodium bicarbonate infusion has been shown to produce depends on the balance between various endogenous constrictors
pulmonary vasodilatation both in animal models and human (endothelin and thrombaxane) and dilators (nitric oxide (NO) and
newborns.7,8 However, both hyperventilation and sodium prostaglandins). Majority of these substances are produced by the
bicarbonate infusion should be used with caution in newborns due pulmonary endothelium. In addition, arterial oxygen and carbon
to their adverse effects on cerebral and coronary circulation.9,10 dioxide tension, and lung volume play a major role in the
Sodium bicarbonate infusion can increase intracellular acidosis regulation of pulmonary vascular tone. Pulmonary hypertension in
and worsen myocardial perfusion and cardiac output. patients with MAS is secondary to hypoxia, acidosis, release of the
Hyperventilation may also increase barotrauma and volutrauma, inflammatory mediators and alveolar atelectasis. Thus the
and increase the risk for chronic lung disease.11 Hypocarbia below pulmonary hypertension of MAS should ideally be treated with
25 mm Hg can also result in hearing loss secondary to injury to adequate ventilation and pulmonary vasodilators. The problem
hair cells.10 Walsh-Sukys et al.6 reported an increased need for until recently was the inability to find such a truly selective
extracoporeal membrane oxygenation (ECMO) and oxygen at pulmonary vasodilator. We will briefly review the past and present
28 days in patients treated with sodium bicarbonate. Neither pulmonary vasodilators used in MAS. Table 1 shows dosages of
hyperventilation nor alkali infusion have been vigorously tested in various pulmonary vasodilators used in pulmonary hypertension.
a RCT. Fortunately their use seems to be on the decline since the
availability of INO. Tolazoline
Initial management has also traditionally included inotropic Tolazoline is a nonspecific vasodilator, which had been used for
support to assist cardiac function against suprasystemic pulmonary the treatment of pulmonary hypertension at least two decades
pressures. Once again, there is no randomized study to support this before the introduction of INO. Lee and Hox31 showed that
practice. Drummond et al.8 were able to show in a case series that tolazoline led to pulmonary vasodilatation even in the absence of
dopamine infusion along with tolazoline and hyperventilation endothelium. Whereas Curtis et al.32 showed in the animal model
helped reduce pulmonary pressure. The combination of tolazoline that the vasodilatation produced was independent of NO
and dopamine had a variable and unpredictable effect on production. Its mechanism of action is directly on the vascular

Journal of Perinatology
Pharmacotherapy for MAS
A Asad and R Bhat
S74

Table 1 Pulmonary vasodilators Several of these agents are being used in adults with severe
Tolazoline IV 0.5–2 mg kg1 13,14
pulmonary arterial hypertension. Milrinone, dipyridamole,
(Not available in USA) NEB 1–2.5 mg kg1 15 zaprinast and sildenafil (Viagra) have all been used in newborns
but the number of patients treated so far are few and no large
Magnesium sulfate IV 200 mg kg1 bolus control studies are available to date.
20–150 mg kg1 h1 16
Diltiazem IV 1–2 q12 to q6 h17 Milrinone. Milrinone is a PDE-3-specific inhibitor and is one of
the earliest drugs studied in newborns. It acts synergistically with
PDE inhibitors INO inducing pulmonary vasodilatation, but in addition it is also a
Dipyridamole IV 0.3–0.6 mg kg1 18– 21 systemic vasodilator and has positive myocardial inotropic activity.
Esoprostenol/ IV 2–5 ng kg1 min1 increments of It is frequently used in newborns, pediatric and adult patients
prostacyclin 2–5 ng kg1 q15 min22–24
following cardiac surgery. Inhaled milrinone has been tried in
ET 50 ng kg1 can be repeated 2  25
adults with pulmonary hypertension undergoing cardiac surgery.
Milrinone IV Term
Loading 75 mcg kg1  60 min
In a recent case report, milrinone was successfully used in four
Maintenance 0.5–0.75 mcg kg1 min1 infants who failed to respond to INO.35 Pharmacokinetics of
<30 GA milrinone have not been studied in newborns. Milrinone may have
Loading 0.75 mcg kg1 min1 over 3 h limited role with the advent of more specific PDE-5 inhibitors, for
Maintenance 0.2 mcg kg1 min1 26 example, sildenafil.
Sildenafil PO 0.25–1 mg kg1 27,28
Tezosentan IV 5 mg h1 for 30 min, 1 mg h1 for 24–72 h29 Dipyridamole. Dipyridamole is a selective PDE-5 inhibitor. In
Adenosine UVC 25–50 mg kg1 min1 30 experimental models it has been shown to augment and prolong
Abbreviations: ET, endotracheal; IV, intravenous; GA, gestational age; NEB, nebulized; the response of NO in post-cardiac surgery patients.36 However,
PO, per oral; UVC, through umbilical vein catheter. augmentation response is not consistent and in some pediatric
See the text for details on each drug. studies decrease in pulmonary vascular resistance of >20% is noted
in only half the treated cases. Dipyridamole has been shown to help
smooth muscle through direct a-adrenergic antagonistic action. in attenuating the rebound phenomenon seen after discontinuation
Tolazoline can be given intravenously as well as by intratracheal of INO therapy in pediatric patients.18,19 Dosage of dipyridamole
route.33,34 There is still no consensus on the dose of tolazoline at ranged from 0.3 to 0.6 mg kg1.18–21
which pulmonary vasodilatation can be maximized while eliminating
the severe systemic hypotension. Severe systemic hypotension is the Zaprinast. Zaprinast is a more selective inhibitor of PDE-5 and
most common and severe adverse effect. The drug is currently not was one of the original inhibitors studied along with dipyridamole.
available in the United States and is not recommended for the Intravenous administration showed, in a fetal lamb model, to
treatment of MAS with pulmonary hypertension. decrease pulmonary arterial resistance as well as a potentiation of
INO inhaled NO-mediated pulmonary vasodilatation.36–38 The concern
An ideal pulmonary vasodilator should (a) decrease pulmonary with intravenous administration was that at moderate-to-high
vascular resistance but not systemic vascular resistance, (b) rapidly doses systemic vasodilatation was noted. Aerosolized drug therapy
relieve hypoxia, acidosis and vasoconstriction, (c) have no or has been shown to have less systemic side effects, preferential
minimal systemic effects and (d) be rapidly metabolized or cleared delivery to better ventilated areas of the lungs and a potentiation of
from the body. INO is a good example of such a drug. Indications the INO effect.39,40
and dose of INO are discussed elsewhere in this issue.
Sildenafil. The most specific PDE-5 inhibitor currently available
Phosphodiesterase inhibitors is sildenafil. It is becoming increasingly popular in the treatment
Phosphodiesterases (PDEs) are a family of enzymes that hydrolyze of pulmonary hypertension, especially in post-cardiac surgery
cyclic nucleotides and regulate their intracellular level. Both cyclic cases.41 Initial success has been noted in some pediatric cases in
adenosine monophosphate and cyclic guanosine monophosphate attenuation of INO-rebound pulmonary hypertension.42 Baquero
play a crucial role in cell signaling. They are classified based on et al.,27 in a proof-of-concept, randomized masked study showed
their substrate specificities, pharmacological properties and an improvement in oxygenation index and pulse oxygen saturation
distribution in the body. PDE inhibitors block the hydrolysis of within 6 to 30 h, and significant increase in survival rate
cyclic nucleotides and thus increase intracellular accumulation of (85%, 6/7 infants) in those randomized to receive oral sildenafil.
cyclic adenosine monophosphate or cyclic guanosine monophos- In the placebo group only 1/6 (16%) survived. The dose of
phate. A brief review of the PDEs’ inhibitors is given below. sildenafil was 1 mg kg1 every 6 h till oxygenation index decreased

Journal of Perinatology
Pharmacotherapy for MAS
A Asad and R Bhat
S75

to <20. Pharmacokinetics and bioavailability of orally pulmonary hypertension in adults. Side effects of the drug include
administered sildenafil has not been studied so far in neonates. abnormal liver enzymes. Second-generation ET-A blockers such as
A recent report by Martell et al.43 have shown a significant drop in tezosentan and sitaxsentan are being studied both in animal
pulmonary pressures within 2 min following intratracheal models and in adults with pulmonary hypertension and/or heart
administration of 0.75 or 1.5 mg kg1 dose of sildenafil in piglets failure at this time.29,52
with meconium aspiration. Inotropic support was needed to
maintain systemic blood pressure. A multicenter study with a large Adenosine
sample size is needed to assess the safety and efficacy of ATP is a purine nucleotide and well-known dilator of systemic and
intravenous, oral or intratracheal sildenafil with or without INO. pulmonary vasculature in both fetal and neonatal vessels.53 ATP
Oral sildenafil can be a lifesaving drug in the developing causes vasodilatation by acting on endothelial A2 adenosine
countries to treat pulmonary hypertension and also for infants with receptors and subsequent release of NO. Its use in a meconium
severe hypoxia and respiratory failure in community hospitals prior lung was studied by Kappa et al.53 in the porcine model and he
to their transfer to a tertiary care center. Both dipyridamole and showed that at low doses it was able to cause an amelioration of
sildenafil have been approved for clinical use in adults with the pulmonary hypertension. However, withdrawal of ATP led to
pulmonary hypertension. significant rebound and even an overshoot phenomenon.
From a randomized placebo-controlled trial of 18 infants,
Prostaglandins and prostacyclins Konduri et al.30 showed an improvement in oxygenation only in
It has been known that both the fetal and neonatal pulmonary 4/9 (45%) term infants with persistent pulmonary hypertension.
vascular bed is sensitive to the arachidonic acid metabolites. The dosage used was 25 to 50 mg kg1 min1 and no systemic side
Prostaglandin E1 (PGE1) has been in clinical use for more than effects were observed. It did not decrease the need for ECMO,
three decades for maintaining ductal patency, but it is a weak bronchopulmonary dysplasia or mortality. Adenosine does have a
pulmonary vasodilator when compared with prostacyclin (PGI2).44 short half-life and rapid metabolism, therefore it should be given
PGI2 has been in use to treat adult pulmonary hypertension for through the umbilical venous line directly to right atrium to reach
several years and is the first drug to be approved for clinical use. It the pulmonary artery.
is a potent pulmonary and systemic vasodilator, and its action is
mediated through increased cyclic adenosine monophosphate. It Magnesium sulfate
has antiproliferative and antiplatelet adhesion effects. The major Magnesium at high doses is a smooth muscle relaxant and
drawback of PGI2 is the need for long-term intravenous infusion vasodilator. It acts by antagonizing calcium entry in to smooth
and its very short half-life. Various synthetic analogs of PGI2, muscle cells. Wu et al. have shown that it also suppresses
namely treprostinil, iloprost and beraprost, are in clinical use in catecholamine release and alters metabolism of prostaglandins and
adults because of their longer half-life and ease of administration, responsiveness of smooth muscles to vasopressors.54 Owing to its
either orally or as aerosol and subcutaneous drip. Aerosolized PGI2 varying modes and site of action, it is a relatively nonspecific
has been tried in newborns who failed to respond to INO.45 Another vasodilator with a significant risk for systemic hypotension.55,56
prostaglandin, PGD2 has been evaluated in newborns with However, in clinical trials in children who have failed conventional
pulmonary hypertension but is yet to show any clinical benefit.46 therapies, significant improvement has been shown in both
oxygenation and oxygenation index without significant
Endothelin antagonists hypotension.16,56 Magnesium sulfate also has sedative and
Endothelins are vasoconstrictors derived from the endothelial cells. antithrombotic activities. In third world countries where INO is not
Of the two major types of endothelins, ET-1 mediates easily available, this multifaceted drug can be used to alleviate
vasoconstriction and smooth muscle cell proliferation through hypoxia. RCTs have not been done so far.
ET-A receptors. ET-A receptors have been found on smooth muscle
cells in pulmonary arteries.47 Endothelins are involved in
maintaining the pulmonary vascular tone in the neonatal period. Future pulmonary vasodilators
Recent studies have reported elevated ET-1 level48 and an Table 2 shows some of the newer pulmonary vasodilators currently
upregulation of ET-1 gene expression in the pulmonary undergoing investigation.
vasculature following MAS.22,49 In adults with pulmonary Table 2 Emerging pulmonary vasodilators
hypertension, the combined ET-A and ET-B receptor antagonist
bosentan has been used successfully.50,51 In pediatric and neonatal 1 Superoxide dismutase
2 Vasoactive intestinal peptide
populations information is limited to case reports. The drug is
3 Adrenomedulline
currently available only in oral formulation, which may hinder its
4 Arginine
use in critically ill newborns. Bosentan is approved by the FDA for

Journal of Perinatology
Pharmacotherapy for MAS
A Asad and R Bhat
S76

Arginine antibiotic treatment did not alter duration of tachypnea,


L-arginine infusion is being studied as an adjunct for NO as well as oxygen support and the need for nasal continuous positive airway
for its potential use in the weaning process. L-arginine is a required pressure between the treated and untreated infants. Of the
substrate for NO synthesis and under oxidative stress conditions it 306 infants, 2-month follow-up data were available only for
increases the activity of nitric oxide synthase.57 It also helps to 259 infants. None of these infants required ventilator support and
preserve endogenous nitric oxide synthase activity thus has none had perinatal risk factors. The use of antibiotics gave no
potential for helping in the weaning of NO. RCTs of L-arginine in advantage in terms of oxygen supplementation, severity of
persistent pulmonary hypertension are still lacking. respiratory distress or mortality. Two other prospective studies from
Shankar et al.68 and Krishnan et al.69 from India also concluded
Superoxide dismutase that routine post-natal antibiotic therapy is of no benefit. These
Superoxide dismuatse is an endogenous scavenger and potent studies do support the current view that routine antibiotic therapy
anti-inflammatory and anti-oxidative enzyme. There is now a in MAS does not alter clinical course. Currently in our unit only
recombinant version (rhSOD) that is commercially available. The infants admitted with MAS requiring ventilatory support or infants
rhSOD is being studied as an adjunct with INO therapy. admitted with risk factors (prolonged rupture of membranes,
Steinhorn et al.58 initially showed how single dose administration chorioamnionitis or positive antenatal group B beta streptococcus
of rhSOD in a lamb model with and without INO led to selective screen) receive antibiotics after obtaining initial cultures.
pulmonary vasodilatation. In view of these anti-inflammatory
and pulmonary vasodilator effects, rhSOD appears to be an exciting Anti-inflammatory agents
new addition to treat pulmonary hypertension in MAS. Steroids. Although the initial phase of deterioration in the lung
mechanics is because of the mechanical obstruction and surfactant
Other drugs inactivation, the persistence of the dysfunction is thought to be
Surfactant. Meconium contents are highly viscous and consist of because of activation of the inflammatory cascade. Chemical
desquamated epithelial cells and inspissated intestinal secretions. pneumonitis is the second cardinal finding in MAS. Khan et al.70
The large glycoproteins in meconium cause increased in 2002 showed histological and biochemical evidence of
adhesiveness.59 Rubin et al.60 in 1996 reported that the mucociliary meconium-induced airway dysfunction in a murine model. Our
transportability of meconium was lower than that of sputum own animal studies in newborn rabbits following tracheal
clearance in cystic fibrosis patients. Meconium constituents instillation of meconium showed significant increase in several
especially the fatty acids and other soluble proteins and bilirubin cytokines namely TNFa, IL-8 and IL-1b in tracheal aspirate.71
are direct inhibitors of surfactant.61,62 Two small randomized This was associated with significant apoptosis.72 As inflammation is
control studies have reported significant improvement in one of the major findings in MAS; several investigators have
oxygenation and decrease in barotrauma with surfactant attempted to treat MAS with steroids. Steroids are potent anti-
treatment.63,64 Readers are referred to excellent reviews on inflammatory medications. Glucocorticoids, particularly
surfactant therapy and lavage in meconium aspiration in this dexamethasone, has been shown to improve oxygenation and lung
issue. function in animal models of MAS73 and in at least one
uncontrolled study of human newborns with MAS.74 A recent
Antibiotics Cochrane review75 found two small RCTs (a 3rd study was
The use of antibiotics in MAS has been controversial. Initial unpublished) and the meta-analysis of the data showed no
use was advocated as it was believed that stress caused the passage differences in mortality, chronic lung disease and length of stay.
of meconium and the most likely reason for perinatal stress The major weaknesses in these studies were small sample size and
was an infection, hence the need to treat with antibiotics. This lack of long-term follow-up. Owing to the lack of consistent
was substantiated in 1967 when Bryan65 showed that sterile short-term benefit as well as recent concerns about its long-term
meconium was never fatal on its own, but when given outcome, dexamethasone cannot be recommended at this time as
intratracheally along with Escherichia coli it reduced the number an anti-inflammatory agent in MAS.
of organisms needed to cause death. A more recent in vitro study In summary the recent advances in pharmacological treatment
by Eidelman et al.66 showed once again that clear amniotic fluid namely INO and surfactant has certainly decreased the need for
was inhibitory for bacterial growth but in the presence of ECMO in MAS infants. However there is a definite need for further
meconium b-Streptococci grew at much faster rate. It is still multicenter studies to evaluate some of the specific and nonspecific
controversial whether every infant with meconium aspiration therapies discussed in this review. As MAS is a major cause of
needs antibiotic therapy. Few prospective RCTs are available from mortality and morbidity in the developing countries, studies
outside the United States for comparison of antibiotic use. In a focusing on prevention and early treatment should be continued to
prospective RCT of non-ventilated infants Lin et al.67 showed that alleviate this tragedy.

Journal of Perinatology
Pharmacotherapy for MAS
A Asad and R Bhat
S77

Acknowledgments 19 Ivy DD, Kinsella JP, Ziegler JW, Abman SH. Dipyridamole attenuates rebound
pulmonary hypertension after inhaled nitric oxide withdrawl in postoperative
We thank Dr Dharmapuri Vidyasagar for his many contributions on the
congenital heart disease. J Thorac Cardiovasc Surg 1998; 115: 875–882.
pathophysiology and treatment of MAS during the last 30 years. 20 Kinsella JP, Toricelli F, Ziegler JW, Ivy DD, Abman SH. Dipyridamole augmentation of
response to nitric oxide. Lancet 1995; 346: 647–648.
Disclosure 21 Travadi JN, Patole SK. Phosphodiesterase inhibitors for persistent pulmonary
hypertension of the newborn: a review. Pediatr Pulmonol 2003; 36(6): 529–535.
R Bhat has lectured while on a speakers bureau. A Asad has declared no 22 Shekerdamien LS, Penny DJ, Ryhammer PK, Reader JA, Ravn HB. EndothelinFa
financial interests. receptor blockade and inhaled nitric oxide in a porcine model of meconium aspiration
syndrome. Pediatr Res 2004; 56: 353–358.
23 Schranz D, Zepp F, Iversen S, Wippermann C, Huth R, Zimmer B et al. Effects of
tolazoline and prostacyclin on pulmonary hypertension in infants after cardiac surgery.
Crit Care Med 1992; 20(9): 1243–1249.
References 24 Bush A, Busst CM, Knight WB, Shinebourne EA. Comparison of the haemodynamic
1 Wiswell TM. Advances in treatment of the meconium aspiration syndrome. Acta effects of epoprostenol (prostacyclin) and tolazoline. Br Heart J 1988; 60(2): 141.
Paediatr Suppl 2001; 436: 28–30. 25 De Jaeger AP, den Anker JN. Endotracheal instillation of prostacyclin in preterm infants
2 Wiswell TE. Meconium in the Delivery Room Trial Group: delivery room management with persistent pulmonary hypertension. Eur Respir J 1998; 12: 932–934.
of the apparently vigorous meconium-stained neonate: results of the multicenter 26 Paradisis M, Jiang X, Mclachlan AJ, Evans N, Kluckow M, Osborn D. Population
collaborative trial. Pediatrics 2000; 105: 1–7. pharmacokinetics and dosing regimen of milrinone in preterm infants. Arch Dis Child
3 Linder N, Aranda JV, Tsur M. Need for endotracheal intubation and suction in Fetal Neonatal Ed 2007; 92: F204–F209.
meconium-stained neonates. J Pediatr 1988; 112: 613–615. 27 Baquero H, Soliz A, Neira F, Venegas ME, Sola A. Oral sildenafil in infants with
4 Bhat R, Chari G, Gulati A, Aldana O, Velamati R, Bhargava H. Pharmacokinetics of persistent pulmonary hypertension of the newborn: a pilot randomized blinded study.
Morphine in preterm and term neonates. J Pediatr 1990; 117: 477–481. Pediatrics 2006; 117: 1077–1083.
5 Bhat R, Abu-Harb M, Chari G, Gulati A. Morphine metabolism in acutely ill preterm 28 Erickson S, Reyes J, Bohn D, Adatia I. Sildenafil (Viagra) in childhood and neonatal
newborn infants. J Pediatr 1992; 120: 795–799. pulmonary hypertension. J Am Coll Cardiol 2002; 39(Suppl 2): 402–402(1).
6 Walsh-Sukys MC, Tyson JE, Wright LL, Bauer CR, Korones SB, Stevenson DK et al. 29 McMurray JJV, Teerlink JR, Cotter G, Bourge RC, Cleland JG, Jondeau G et al. Effects of
Persistent pulmonary hypertension of the newborn in the era before nitric oxide: Tezosentan on symptoms and clinical outcomes in patients with acute heart failure.
practice variation and outcomes. Pediatrics 2000; 105: 14–20. JAMA 2007; 298(17): 2009–2019.
7 Lyrene RK, Welch KA, Godoy G, Philips III JB. Alkalosis attenuates hypoxic pulmonary 30 Konduri GG, Garcia DC, Kazzi NJ, Shankaran S. Adenosine infusion improves
vasoconstriction in neonatal lambs. Pediatr Res 1985; 19: 1268–1271. oxygenation in term infants with respiratory failure. Pediatrics 1996; 97: 295–300.
8 Drummond WH, Gregory GA, Heymann MA, Phibbs RA. The independent effects of 31 Lee TS, Hou X. Dual vasoactive effects of tolazoline on rabbit pulmonary arteries.
hyperventilation, tolazoline, and dopamine in infants with persistent pulmonary J Cardiothoc Vasc Anesth 1996; 10: 364–367.
hypertension. J Pediatr 1981; 98: 608–611. 32 Curtis J, Palacino JJ, O’Neill JT. Production of pulmonary vasodilation by tolazoline,
9 Hansen NB, Brubakk AM, Bratlid D, Oh W, Stonestreet BS. The effects of variations in independent of nitric oxide production in neonatal lambs. J Pediatr 1996; 128:
PaCO2 on brain blood flow and cardiac output in the newborn piglet. Pediatr Res 118–124.
1984; 18(11): 1132–1136. 33 Paret G, Eyal O, Mayan H, Ben-Abraham R, Vardi A, Manisterski Y et al.
10 Ambalavanan N, Carlo WA. Hypocapnia and hypercapnia in respiratory management of Pharmacokinetics of endobronchial tolazoline administration in dogs. Am J Perinatol
newborn infants. Clin Perinatol 2001; 28(3): 517–531. Review. 1999; 16: 1–6.
11 Greenough A, Khetriwal B. Pulmonary hypertension in the newborn. Paediatr Respir 34 Welch JC, Bridosn JM, Gibbs JL. Endotracheal tolazoline for severe persistent pulmonary
Rev 2005; 6(2): 111–116. hypertension of the newborn. Br Heart J 1995; 73: 99–100.
12 Booker PD, Evans C, Franks R. Comparison of the haemodynamic effects of dopamine 35 Bassler D, Choong K, McNamara P, Kirpalani H. Neonatal persistent pulmonary
and dobutamine in young children undergoing cardiac surgery. Br J Anaesth 1995; hypertension treated with milrinone: four case reports. Biol Neonate 2006; 89: 1–5.
74(4): 419–423. 36 Ziegler JW, Ivy DD, Fox JJ, Kinsella JP, Clarke WR, Abman SH et al Dipyridamole, a
13 Stevenson DK, Kasting DS, Darnall Jr RA, Ariagno RL, Johnson JD, Malachowski N et al. cGMP phosphodiesterase inhibitor, causes pulmonary vasodilation in the ovine fetus.
Refractory hypoxemia associated with neonatal pulmonary diseases: the use and Am J Physiol 1995; 269: 473–479.
limitations of tolazoline. J Pediatr 1979; 95: 595–599. 37 Thusu KG, Morin III FC, Russell JA, Steinhorn RH. The cGMP phosphodiesterase
14 Nuntnarumit P, Korones SB, Yang W, Bada HS. Efficacy and safety of tolazoline for inhibitor zaprinast enhances the effect of nitric oxide. Am J Respir Crit Care Med 1995;
treatment of severe hypoxemia in extremely preterm infants. Pediatrics 2002; 109(5): 152: 1605–1610.
852–856. 38 Ichinose F, Adrie C, Hurford WE, Zapol WM. Prolonged pulmonary vasodilator
15 Parida SK, Baker S, Kuhn R, Desai N, Pauly TH. Endotracheal tolazoline action of inhaled nitric oxide by Zaprinast in awake lambs. J Appl Physiol 1995; 78:
administration in neonates with persistent pulmonary hypertension. J Perinatol 1997; 1288–1295.
17: 461–464. 39 Ichinose F, Adrie C, Hurford WE, Bloch KD, Zapol WM. Selective pulmonary
16 Tolsa JF, Cotting J, Sekarski N, Payot M, Micheli JL, Calame A. Magensium Sulphate as vasodilation induced by aerosolized zaprinast. Anesthesiology 1998; 88: 410–416.
an alternative and safe treatment for persistent pulmonary hypertension of the 40 Newman SP. Aerosol deposition considerations in inhalation therapy. Chest 1985;
newborn. Arch Dis Child Fetal Neonatal Ed 1995; 72: F184–F187. 88(2 Suppl): 152S–160S. Review.
17 Islam S, Masiakos P, Schnitzer JJ, Doody DP, Ryan DP. Diltiazem reduces pulmonary 41 Stocker C, Penny DJ, Brizard CP, Cochrane AD, Soto R, Shekerdemian LS. Intravenous
arterial pressures in recurrent pulmonary hypertension associated with pulmonary sildenafil and inhaled nitric oxide: a randomised trial in infants after cardiac surgery.
hypoplasia. J Pediatr Surg 1999; 34: 712–714. Intensive Care Med 2003; 29(11): 1996–2003.
18 Al-Alaiyan S, Al-Omran A, Dyer D. The use of phoshodiestersae inhibitor( dipyridamole) 42 Atz AM, Wessel DL. Sildenafil ameliorates effects of inhaled nitric oxide withdrawl.
to wean from inhaled nitric oxide. Intensive care med 1996; 22: 1093–1095. Anesthesiology 1999; 91: 307–310.

Journal of Perinatology
Pharmacotherapy for MAS
A Asad and R Bhat
S78

43 Martell M, Blasina F, Silvera F, Tellechea S, Godoy C, Vaamonde L. Intratracheal sildenafil 59 Lam BCC, Yeung CY. Surfactant lavage for meconium aspiration syndrome: a pilot
in the newborn with pulmonary hypertension. Pediatrics 2007; 119: 215–216. study. Pediatrics 1999; 103: 1014–1018.
44 Tamura M, Kurumatani H, Matsushita T. Comparative effects of beraprost, a stable 60 Rubin BK, Tomkiewicz RP, Patrinos ME, Easa D. The surface and transport
analogue of prostacyclin, with PGE(1), nitroglycerin and nifedipine on canine model properties of meconium and reconstituted meconium solutions. Pediatr Res 1996;
of vasoconstrictive pulmonary hypertension. Prostaglandins Leukot Essent Fatty Acids 40: 834–838.
2001; 64(3): 197–202. 61 Sun B, Curstedt T, Robertson B. Surfactant inhibition in experimental meconium
45 Bindl L, Fahnenstich H, Peukert U. Aerosolised prostacyclin for pulmonary hypertension aspiration syndrome. Acta Pediatr 1993; 82: 182–189.
in neonates. Arch Dis Child Fetal Neonatal Ed 1994; 71(3): F214–F216. 62 al-Mateen KB, Dailey K, Grimes MM, Gutcher GR. Improved oxygenation with
46 Soifer SJ, Clyman RI, Heymann MA. Effects of prostaglandin D2 on pulmonary arterial exogenous surfactant administration in experimental meconium aspiration syndrome.
pressure and oxygenation in newborn infants with persistent pulmonary hypertension. Pediatr Pulmonol 1994; 17(2): 75–80.
J Pediatr 1998; 112: 774–777. 63 Dargaville PA, South M, McDougall PN. Surfactant and surfactant inhibitors in
47 Buchan KW, Magnusson KW, Rabe KF, Summer MJ, Watts IS. Characterization of the meconium aspiration syndrome. J Pediatr 2001; 138: 113–115.
endothelin receptor mediating contraction of human pulmonary artery using BQ123 64 Findlay RD, Taeusch HW, Walther FJ. Surfactant replacement therapy for meconium
and Ro46-2005. Eur J Pharmacol 1994; 260: 221–226. aspiration syndrome. Pediatrics 1996; 97: 48–52.
48 Kuo C, Chen J. Effect of meconium aspiration on plasma endothelin-1 level and 65 Bryan CS. Enhancement of bacterial infection by meconium. Johns Hopkins Med J
pulmonary hemodynamics in a piglet model. Biol Neonate 1999; 76: 228–234. 1967; 121: 9–13.
49 Kyo CY. EndothelinFa receptor antagonist prevents neonatal pulmonary hypertension 66 Eidelman AI, Nevet A, Rudensky B, Rabinowitz R, Hammerman C, Raveh D et al. The
in meconium aspiration in piglets. J Formos Med Assoc 2001; 100: 420–423. effect of meconium staining of amniotic fluid on the growth of Escherichia coli and
50 Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM, Keogh A et al. Bosentan therapy for group B Streptococcus. J Perinatol 2002; 22: 467–471.
pulmonary arterial hypertension. N Eng J Med 2002; 346: 896–903. 67 Lin HC, Su BH, Tsai CH, Lin TW, Yeh TF. Role of antibiotics in management of
51 Sitbon O, Badesch DB, Channick RN, Frost A, Robbins IM, Simonneau G et al. Effects non-ventilated cases of meconium aspiration syndrome without risk factors for
of dual endothelin receptor antagonist bosentan in patients with pulmonary arterial infection. Biol Neonate 2005; 87: 51–55.
hypertension: a 1-year follow-up study. Chest 2003; 124: 247–254. 68 Shankar V, Paul VK, Deorari AK, Singh M. Do neonates with meconium aspiration
52 Geiger R, Pajk W, Neu N, Maier S, Kleinsasser A, Fratz S et al. Tezosentan decreases syndrome require antibiotics? Indian J Pediatr 1995; 62: 327–331.
pulmonary artery pressure and improves survival rate in an animal model of 69 Krishnan L, Nasruddin, Prabhakar P, Bhaskaranand N. Routine antibiotic cover for
meconium aspiration. Pediatr Res 2006; 59: 147–150. newborns intubated for aspirating meconium: is it necessary? Indian Pediatr 1995;
53 Kappa P, Jahnukainen T, Grinlund J, Rautanen M, Halkola L, Välimäki I. Adenosine 32(5): 529–531.
triphosphate treatment for meconium aspiration-induced pulmonary hypertension in 70 Khan AM, Elidemir O, Epstein CE, Lally KP, Xue H, Blackburn M et al. Meconium
pigs. Acta Physiol Scand 1997; 160(3): 283–289. aspiration produces airway hyperresponsiveness and eosinophilic inflammation in a
54 Wu TJ, Teng RJ, Tsou KI. Persistent pulmonary hypertension of the newborn treated murine model. Am J Physiol Lung Cell Moll Physiol 2002; 283: L785–L790.
with magnesium sulfate in premature neonates. Pediatrics 1995; 96: 472–474. 71 Zagariya A, Bhat R, Navale S, Vidyasagar D. Cytokine expression in meconium-induced
55 Patole SK, Finer NN. Experimental and clinical effects of magnesium infusion in the lungs. Indian J Pediatr 2004; 71(3): 195–201.
treatment of neonatal pulmonary hypertension. Magnes Res 1995; 8(4): 373–388. 72 Zagariya A, Bhat R, Uhal B, Navale S, Freidine M, Vidyasagar D. Cell death and lung
Review. cell histology in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; 159:
56 Abu-Osaba YK, Galal O, Mansara K, Rejjal A. Treatment of severe persistent 819–826.
pulmonary hypertension of the newborn with magnesium sulphate. Arch Dis Child 1992; 73 Khan AM, Shabarek FM, Kutchback JW, Lally KP. Effects of dexamethasone on
67: 31–35. meconium aspiration syndrome in newborn piglets. Pediatr Res 1999; 46(2):
57 Palmer RM, Rees DD, Ashton DS, Moncada S. L-arginine is the physiological precursor 179–183.
for the formation of nitric oxide in endothelium-dependent relaxation. Biochem 74 da Costa DE, Nair AK, Pai MG, Al Khusaiby SM. Steroids in full term infants with
Biophys Res Commun 1988; 153(3): 1251–1256. respiratory failure and pulmonary hypertension due to meconium aspiration syndrome.
58 Steinhorn RH, Albert G, Swartz DD, Russell JA, Levine CR, Davis JM. Recombinant Eur J Pediatr 2001; 160: 150–153.
human superoxide dismutase enhances the effect of inhaled nitric oxide in persistent 75 Ward M, Sinn J. Steroid therapy for meconium aspiration syndrome in newborn
pulmonary hypertension. Am J Respir Crit Care Med 2001; 164(5): 834–839. infants. Cochrane Database Syste Rev 2003; (4): CD003485. Review.

Journal of Perinatology
Journal of Perinatology (2008) 28, S79–S83
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Extracorporeal membrane oxygenation: use in meconium
aspiration syndrome
BL Short
Division of Neonatology, Department of Pediatrics, Children’s National Medical Center, The George Washington School of Medicine,
Washington, DC, USA

therapy as a ‘time-buying’ procedure. If you give extracorporeally


Extracorporeal membrane oxygenation (ECMO) has been successful as a respiratory and cardiac support to the level, the patient can be
rescue therapy for infants with respiratory failure with some diagnoses such placed on minimal ventilatory and pressor therapy over time, and a
as meconium aspiration syndrome (MAS) having a survival rate of more percentage of the patients will have recovery of lung and/or cardiac
than 94%. New therapies have allowed many infants who would have function. As all patients have to be systemically heparinized to be
required ECMO to be kept off ECMO, but at what cost. The survival rate for on an ECMO circuit, the known vasodilatory effect of heparin on
the neonatal ECMO patient has dropped over the years, whereas the time of the pulmonary vessels may assist in the reduction of pulmonary
ECMO has increased, indicating that the new therapies are keeping the less vascular resistance in patients placed on ECMO.2
ill infants off ECMO. The major cause of non-survival in this population In the mid-1960s, ECMO use was directed at the premature
remains intraventricular hemorrhage. The primary risk factors related to infant with the hopes that ECMO would provide the ‘artificial’
this are thought to be pre-ECMO events, such as hypoxia and/or ischemia placenta for these infants.3,4 Although the procedure provided
either prenatally or post-delivery. ECMO events that may complicate this are sufficient cardiorespiratory support for these infants, all infants
heparinization that is required while on ECMO and concern for the effect of died secondary to the high intracranial hemorrhage (ICH) rate in
shear stress and blood flow pattern changes created by the ECMO pump with the premature population. This was thought to be related to the
venoarterial ECMO, although these changes are not seen in venovenous immature brain of these infants with an already high rate of ICH
ECMO, the more common form of ECMO. Newer low-resistant microporous and to the heparinization required for the procedure. The first
artificial lungs and miniaturized pumping systems may allow ECMO to be newborn survivor in 1976 was an infant with meconium aspiration
performed using less blood and safer equipment. The smaller low-resistant syndrome (MAS), turning the focus of treatment to the term
artificial lungs provide the ability to consider giving extracorporeal infant.5 Why would MAS be a good disease to treat with a therapy
life support using only this membrane with flow provided by an such as ECMO? Figure 1 depicts the complex pathophysiology
arterial–venous shunt, thus eliminating the pumping system all together. associated with MAS, with a combination of obstructive lung
Trials are ongoing in adults and, if effective, may direct further research disease, parenchymal disease caused by inflammation and
into using this technique in newborns where the umbilical artery and vein persistent pulmonary hypertension. Ventilation alone, either
could be used as the arterial–venous shunt. conventional or high-frequency ventilation, of the severe cases may
Journal of Perinatology (2008) 28, S79–S83; doi:10.1038/jp.2008.152 not be able to break the cycle associated with severe hypoxia and
persistent pulmonary hypertension, thus making ECMO an ideal
therapy for this complex disease. While on ECMO, the ventilator
Introduction can be placed at lung rest, allowing gentle ventilation, time for the
Extracorporeal membrane oxygenation (ECMO) is the use of a body to absorb the meconium and time for the pulmonary
modified cardiopulmonary bypass circuit to supply respiratory hypertension to resolve. Figure 2 from the Extracorporeal Life
and/or cardiac support for patients in cardiorespiratory failure.1 Support Organization database shows the changing patterns of use
The ECMO circuit supplies an artificial lung and an artificial in ECMO, with the MAS infant ranging from 45 to now 35% of the
pump, which allow the patient to be supported for both respiratory neonatal patients treated with ECMO. The survival rate for MAS
and cardiac needs. On account of this, many describe ECMO infants compared with infants with other disease states is shown in
Figure 3. Infants with MAS, who do not survive ECMO, do not do so
Correspondence: Dr BL Short, Division of Neonatology, Department of Pediatrics, Children’s because of a major intracranial complication, not because of
National Medical Center, The George Washington School of Medicine, 111 Michigan Ave.,
NW, Washington, DC 20010, USA.
irreversibility of the lung disease. These findings indicate that
E-mail: bshort@cnmc.org infants with severe MAS need to be referred to an ECMO center
ECMO use in MAS
BL Short
S80

prior to meeting conventional ECMO criteria to reduce the risk for inhaled nitric oxide, the need for ECMO has reduced.1,12 –15
pre-ECMO cardiovascular instability making transfer difficult. Figure 4 shows the reduction in the number of infants being placed
Most intracranial bleeds are thought to be secondary to hypoxic/ on ECMO over time. Also depicted in this figure is the reduction in
ischemic insults prior to ECMO that would, in a non-heparinized the survival rate of the ECMO patient over time. Although the cause
patient, result in an infarction, but in the heparinized ECMO of this reduction is not clear, it may be assumed that as increasing
population results in a hemorrhage and/or hemorrhagic numbers are rescued by other therapies, the severity and acuity of
infarction.6,7 Studies are now indicating that ECMO conducted the patients going on to ECMO has increased. This finding differs
using venoarterial (VA) ECMO, in which non-pulsatile flow is from those of Fliman et al.,15 who only looked at cases from 1996
created with the ECMO pump, may also place the brain at risk.7–11 to 2003. This difference in survival data is probably related to the
This risk is minimal in venovenous (VV) ECMO, the more common time period studied, with the late 1980s, as depicted in Figure 4,
form of ECMO for the infant with MAS. representing the highest survival rate prior to most new
As new therapies have become available, such as the use of interventions. Figure 5 may support the theory of sicker patients
high-frequency ventilation, surfactant use in term infants, and now being placed on ECMO. This figure shows that as the number
of ECMO patients has decreased, the time on ECMO has increased,
indicating that the severity of lung disease in infants going on
Meconium Aspiration
ECMO is greater, that is, the less severe are being rescued by other
therapies. Table 1 compares the pre-ECMO ventilator and blood gas
Mechanical Chemical
Obstruction Inflammation
100
94
90
Air Trapping 84
Atelectasis 80 78
75
Percent Survival

70
63
Uneven Intrapulmonary 60
Ventilation Shunting 52
50
40
Air Leaks Hypoxia 30
PPHN 20
10
0
MAS PPHN CDH SEPSIS HMD Other

Figure 1 Mechanical obstruction and chemical inflammation are the hallmark Figure 3 Survival data by diagnoses for extracorporeal membrane oxygenation
for meconium aspiration syndrome (MAS), leading to respiratory failure and the (ECMO) patients (data from the Extracorporeal Life Support (ECLS) database,
development of pulmonary hypertension in the MAS patient. January 2007 report, n ¼ 21 258 patients).

100% Others
Sepsis
80% RDS
PPHN/PFC
60% MAS
CDH

40%

20%

0%
19 7
88

19 9
19 0
91

19 2
19 3
94

19 5
96

19 7
19 8
99

20 0
20 1
20 2
20 3
20 4
05
8

8
9

9
9

9
9

0
0
0
0
0
19

19

19

19

20
<=

Figure 2 This figure shows the major diagnoses treated with extracorporeal membrane oxygenation (ECMO). Although the numbers are reduced, the meconium
aspiration syndrome (MAS) population continues to be the largest group of patients treated with ECMO (data from the Extracorporeal Life Support Organization database
from 1987 to 2005).

Journal of Perinatology
ECMO use in MAS
BL Short
S81

1600 100
HFOV
Surfactant 90
1400 Cases % Survival

80
1200
70
1000 iNO
60

Survival (%)
# Cases

800 50

40
600
30
400
20
200
10

0 0
86 87 88 89 90 91 92 93 94 95 96 97 98 99 '00 '01 '02 '03 '04 '05 '06
Year

Figure 4 There has been a reduction in patients requiring extracorporeal membrane oxygenation (ECMO) (black bar) related to new therapies as depicted in the
graph. Survival (line) has decreased over this same time period (data from the Extracorporeal Life Support Organization database January summary report, n ¼ 21 258
patients; iNO, inhaled nitric oxide).

250 100
HFOV Pump Time % Survival
Surfactant 90
iNO
200 80

70
Pump Time (hours)

Survival (%)
150 60

50

100 40

30

50 20

10

0 0
86 87 88 89 90 91 92 93 94 95 96 97 98 99 '00 '01 '02 '03 '04 '05 '06
Year

Figure 5 As survival (line) has decreased, time on extracorporeal membrane oxygenation (ECMO) has increased over the years (black bar) (data from the
Extracorporeal Life Support Organization database January summary report, n ¼ 21 258 patients). iNO, inhaled nitric oxide.

data for the top three diagnoses going on to ECMO. It is interesting from animal models simulating clinical ECMO have shown an
that the MAS infant has an average mean airway pressure of 16 altered cerebral autoregulation pattern in animals exposed to VA
compared with the congenital diaphragmatic infant with a mean ECMO both with and without prior exposure to hypoxia.7–11
airway pressure of 15 mm Hg, indicating the severity of the lung Figure 6 depicts the changes in cerebral blood flow with lower
disease in these infants. cerebral perfusion pressure (lower end of the autoregulatory curve).
The risk for cerebral injury in the ECMO population ranges In the newborn lamb model, 2 h of exposure to severe hypoxia
from 5 to 12%, with severe hypoxia, birth weight and sepsis altered cerebral autoregulation, but when the animals were
representing the highest risk factors.6 Although the pre-ECMO recovered by placing them on ECMO, the alteration in
events are thought to be the primary factors leading to ICH, data autoregulation was markedly worse. Further studies using isolated

Journal of Perinatology
ECMO use in MAS
BL Short
S82

Table 1 Pre-ECMO ventilator and blood gas data (data from the ECLS 20
Control
organization neonatal database, summary from 1997–2007) VV ECMO
VA ECMO
MAS PPHN CDH 10

Age on ECMO (days) 2.00 2.86 2.27


0
Time on ECMO (hours) 138 159 266

% Change
BW (kg) 3.0 3.15 2.89
GA 38 37 37 -10
Apgar (1/5 min) 4/6 6/7 4/6
FIO2 0.96 0.95 0.95 -20
PIP/PEEP 34/3 32/3 32/3
*
MAP 16 14 15 *
-30
Pre-pH 7.00 7.18 7.08 *
Pre-pCO2 44 44 58
Pre-pO2 45 43 39 -40
Pre-sats (%) 65 61 56 1µM 10µM 100µM
Ach Concentration
Abbreviations: BW, birth weight; CDH, congenital diaphragmatic hernia;
ECLS, extracorporeal life support; ECMO, extracorporeal membrane oxygenation; Figure 7 Isolated vessel chamber studies on lamb middle cerebral arteries taken
FIO2, fractional inspired oxygenation; GA, gestational age; MAP, mean airway pressure; from animals exposed to no extracorporeal membrane oxygenation (ECMO)
MAS, meconium aspiration syndrome; PEEP, peep end expiratory pressure; PIP, peep (controls), venoarterial (VA) or venovenous (VV) ECMO are shown in this figure.
inspiratory pressure; PPHN, persistent pulmonary hypertension; Pre-sat, pre-oxygen Vessels were exposed to acetylcholine (Ach) and monitored for dilation. Percent
saturation.
change in vessel diameter is noted on the Y axis.11,12
70
60 VA ECMO may have an alteration of cerebral vascular reactivity
CBF (ml / 100 g / min)

50 resulting in altered autoregulation of the cerebral circulation. The


40
cerebral circulation becomes pressure-passive under these
conditions, making hypertension and hypotension risks for either
30
cerebral ischemia (infarction) or cerebral overperfusion (cerebral
20 hemorrhage). There has been no randomized comparison of VA
10 versus VV ECMO patients to determine whether the incidence of ICH
0 is higher in the VA ECMO population. Many centers still use VA
CPP<25 CPP 25–39 CPP 40–55 Baseline ECMO primarily, but when they use VV ECMO, it is in the less
Cerebral perfusion pressure (mm Hg)
severe infants, making a comparison of the rate of ICH in these
Control Hypoxia Hypoxia-ECMO
populations difficult. A recent study by Radhakrishnan et al.,16
Figure 6 Lamb studies of cerebral autoregulation after prolonged hypoxia and reviewing data from the Extracorporeal Life Support Organization’s
recovery using extracorporeal membrane oxygenation (ECMO) therapy. Control registry on the outcome and complication rate in the MAS
animals (circle) autoregulated down to cerebral perfusion pressures of population treated with ECMO, found that infants with MAS were
<25 mm Hg, whereas animals exposed to hypoxia (square) and recovered with
increasing fractional inspired oxygen (FIO2) could only autoregulate down to treated with VV ECMO 55% of the time compared with 29% for the
cerebral perfusion pressure (CPP) of 25 to 39. If animals were exposed to hypoxia non-MAS patient. They also found that the MAS population had a
and then recovered with ECMO, the alteration would be much worse with loss of significantly higher survival rate and lower number of
autoregulation at a CPP of 40 to 55 mm Hg.7 complications per patient versus the non-MAS patient. They
concluded that, with the lower mortality and morbidity, it was time
arterial vascular chamber techniques have shown that cerebral to consider relaxing the criteria for the MAS patient and ECMO use.
vessels taken from animals exposed to VA ECMO have a marked This may be a consideration when using VV ECMO, in which
alteration in reactivity and that this reactivity is associated with an potential cerebral alterations do not exist and ligation of the carotid
altered production of nitric oxide (see Figure 7).10–12 This artery is not a concern. It may be time to consider a randomized
alteration is thought to be secondary to the shear stress changes trial looking at morbidity instead of mortality in this population.
caused by the non-pulsatile VA ECMO circuit, because this Although MAS is the most common neonatal respiratory failure
alteration is seen only in the VA ECMO-exposed animals and not to diagnoses going on in ECMO, the increasing use of ECMO is now
animals exposed to VV ECMO, in which the pumping chamber is in neonatal and pediatric patients with heart failure post-surgery
the native heart with normal pulsatile blood flow patterns for congenital heart disease.17 With this increase, there has been
(see Figure 7).10–12 These findings indicate that patients placed on the introduction of newer devices for cardiovascular support

Journal of Perinatology
ECMO use in MAS
BL Short
S83

including various types of ventricular assist devices. Artificial heart 5 Bartlett RH, Gazzaniga AB, Jefferies R, Huxtable RF, Haiduc NJ, Fong SW et al.
support for bridge to transplant available for the adult population Extracorporeal membrane oxygenation (ECMO) cardiopulmonary support in infancy.
Trans Am Soc Artif Intern Organs 1976; 22: 80–88.
is now available for the newborn and pediatric population in
6 Bulas D, Glass P. Neonatal ECMO: neuroimaging and neurodevelopmental outcome.
Europe. Research and development of similar devices is ongoing in Semin Perinatol 2005; 29: 58–65.
the United States. The field of conventional ECMO is also 7 Short BL. The effect of extracorporeal life support on the brain: a focus on ECMO.
changing, with newer equipment, including low-resistant Semin Perinatol 2005; 29: 45–50.
microporous oxygenators and centrifugal pumps that will allow the 8 Short BL, Walker LK, Gleason CA, Bender KS, Traystman RJ. Effect of extracorporeal
procedure to be performed with smaller and safer systems.18 Newer membrane oxygenation on cerebral blood flow and cerebral oxygen metabolism in
newborn seep. Pediatr Res 1990; 28: 50–53.
low-resistant microporous membrane lungs have the ability of
9 Short BL, Walker LK, Bender KS, Traystman RJ. Impairment of cerebral autoregulation
allowing ECMO to be conducted without a pump.19–21 On account during extracorporeal membrane oxygenation in newborn lambs. Pediatri Res 1993;
of the low resistance of these membranes, gas transfer can be 33: 289–294.
achieved through an arterial–venous shunt through these 10 Ingyinn M, Lee J, Short BL, Viswanathan M. Venoarterial extracorporeal membrane
membranes. These techniques are presently used in clinical trials oxygenation (VA ECMO) impairs basal nitric oxide production in cerebral arteries of
newborn lambs. Pediatr Crit Care Med 2000; 1: 161–165.
in the adult population in Europe and have shown good success
11 Ingyinn I, Short BL, Rais-Bahrami K, Viswanathan M. Altered cerebrovascular
with survival rates as high as 70% is some studies.19–21 If the responses after exposure to venoarterial extracorporeal membrane oxygenation: role of
results of these studies show improvement in outcome, the use of a the nitric oxide pathway. Pediatr Crit Care Med 2006; 7(4): 368–373.
non-pump-driven circuit with potentially less complications and 12 The Neonatal Inhaled Nitric Oxide Group. Inhaled nitric oxide in full-term and
lower blood prime may allow for the consideration of studies using near-term infants in respiratory failure. N Engl J of Med 1997; 336: 597–604.
extracorporeal support for early intervention instead as a rescue 13 Clark RH, Kueser TJ, Walker MW, Southgate WM, Huckaby JL, Perez JA et al. Low-dose
nitric oxide therapy for persistent pulmonary hypertension of the newborn. N Engl J
therapy. These trials would be designed to look at reduction in the
Med 2000; 342: 469–474.
lung injury, time of ventilation and reduction in hospital length of 14 Walker LK, Short BL, Traystman RH. Impairment of cerebral autoregulation during
stay. To date, this technique has not been used in newborns, but the venovenous extracorporeal membrane oxygenation in the newborn lamb. Crit Care
umbilical vessels make this an attractive approach. The MAS Med 1996; 24: 2001–2006.
population would be perfect candidates to consider for such a study. 15 Fliman PJ, DeRegnier RA, Kinsella JP, Reynolds M, Rankin LL, Steinhorn RH. Neonatal
extracorporeal life support: impact of new therapies on survival. J of Pediatr 2006;
148: 595–599.
Disclosure 16 Radhakrishnan RS, Lally PA, Cox CS. ECMO for meconium aspiration syndrome:
support for relaxed entry criteria. ASAIO J 2007; 53: 489–491.
The author has declared no financial interests.
17 Hines MH. ECMO and congenital heart disease. Semin Perinatol 2005; 29: 34–39.
18 Bartlett RH. Extracorporeal life support: history and new directions. Semin Perinatol
References 2005; 29: 2–7.
1 Rais-Bahrami K, VanMeurs K. ECMO for neonatal respiratory failure. Semin Perinatol 19 Wales T. Clinical experience with the iLA membrane ventilator pumpless
2005; 29: 15–23. extracorporeal lung-assist device (review). Expert Rev Med Devices 2997; 4(3):
2 Thompson BT, Spence CR, Janssens SP, Joseph PM, Hales CA. Inhibition of hypoxic 297–305.
pulmonary hypertension by heparins of differing in vitro antiproliferative potency. Am 20 Bein T, Weber F, Philipp A, Prasser C, Pfeifer M, Schmid FX et al. A new pumpless
J Respir Crit Care Med 1994; 149(6): 1512–1517. extracorporeal interventional lung assist in critical hypoxemia/hypercapnia. Crit Care
3 White JJ, Risemberg H, Andrews HG, Mazur D, Haller Jr JA. Prolonged respiratory support Med 2006; 34(5): 1371–1377.
in newborn infants with a membrane oxygenator. Surgery 1971; 70: 288–296. 21 Liebold A, Reng CM, Philipp A, Pfeifer M, Birnbaum DE. Pumplesss extracorporeal
4 Dorson WJ, Baker E, Cohen MI, Meyer B, Molthan M, Trump A et al. A perfusion lung assist-experience with the first 20 cases. Eur J Cardiothorac Surg 2000; 17(5):
system for infants. ASAIO Trans 1969; 15: 155–160. 608–613.

Journal of Perinatology
Journal of Perinatology (2008) 28, S84–S92
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Role of iNO in the modulation of pulmonary vascular resistance
A Bin-Nun and MD Schreiber
Department of Pediatrics, Section of Neonatology, The University of Chicago, Chicago, IL, USA

endothelial cells that relaxed blood vessels, endothelium-derived


Inhaled nitric oxide (iNO) has quickly become a standard therapy for term relaxing factor. Ignarro et al.,2 in 1987, help solve the riddle with
and near-term infants with hypoxic respiratory failure and persistent compelling evidence supporting NO as the active signaling
pulmonary hypertension. Its effect on the lung is believed to be through the component of endothelium-derived relaxing factor. With
stimulation of soluble guanylyl cyclase and the increased production of Moncada’s work in 1987,3 also showing that NO is the active
cyclic guanosine 30 ,50 -monophosphate (cGMP). However, in addition to component of endothelium-derived relaxing factor, NO has also
pulmonary vasodilation and a decrease in pulmonary vascular resistance, become appreciated as an important neurotransmitter. In 1996, by
nitric oxide (NO) shows several additional potential beneficial effects on the allowing NO gas to bubble through a tissue preparation containing
lung. This article reviews NO mechanisms of action, early clinical trial of guanylylcyclase, Ferid Murad4 showed an increase in the
iNO and clinical aspects for the use of iNO in acute respiratory failure of the production of cyclic guanosine 30 ,50 -monophosphate (cGMP), thus
term and near-tem neonates. identifying the mechanism of action of this important molecule.
Journal of Perinatology (2008) 28, S84–S92; doi:10.1038/jp.2008.161 For their pioneering work, Drs Furchgott, Ignarro and Murad
received the 1998 Nobel Prize for Medicine.

Introduction: nitric oxide


Nitric oxide (NO) is a colorless, odorless gas. Earlier, NO was Pulmonary vasodilatation: mechanism of action
considered only as the toxic product of internal combustion Nitric oxide activates soluble guanylyl cyclase, leading to the
engines, burning cigarettes and lightening storms. The activation of cGMP-dependent protein kinase. In turn,
atmospheric concentration of NO is 10 to 500 p.p.b. (parts per cGMP-dependent protein kinase decreases the sensitivity of myosin
billion) in general, 1.5 p.p.m. (parts per million) in heavy traffic to calcium-induced contraction and lowers the intracellular
and 1000 p.p.m. in tobacco smoke. Human airways produce NO; calcium concentration by activating calcium-sensitive potassium
within the nasal mucosa, NO concentrations reach 25 to 64 p.p.b. channels and inhibiting the release of calcium from the
NO concentrations are considerably lower more distal, reaching sarcoplasmic reticulum. The degradation of cGMP back to
1 to 6 p.p.b. in the mouth, trachea and distal airways. guanosine triphosphate is regulated under the control of a specific
In spite of its Food and Drug Administration (FDA) approval in
December 1999, NO continues to be confused with other
compounds. Some are listed in Table 1 along with their chemical Table 1 NO impersonators
formula and bioactivity. The ability of NO to exist and exert Name Symbol Bioactivity
biological activity in several states strengthens its position as a
Nitrous oxide N2O Anesthetic (‘laughing gas’)
perfect second messenger (Table 2). .
Nitrogen dioxide NO2 Toxic free radical
Nitrate NO
3 Oxidation end product (70%)
Abbreviation: NO, nitric oxide.
Historical perspective
In 1980, Furchgott and Zawadzki1 described how intact blood Table 2 NO equivalents
vessels, possessing intact endothelium and smooth muscle, dilated Name Symbol
to stimuli that induced vasoconstriction when the endothelium was
disturbed. He termed this substance, produced in vascular Nitric oxide NO
.
Nitroxyl anion NO
Correspondence: Dr MD Schreiber, The University of Chicago Comer Children’s Hospital, Nitrosonium NO+
MC8000, 5721 S Maryland Avenue, Chicago, IL 60637, USA.
E-mail: mschreiber@peds.bsd.uchciago.edu Abbreviation: NO, nitric oxide.
iNO and PVR
A Bin-Nun and MD Schreiber
S85

Blood vessel

Endothelial cells

Vascular smooth muscle cells

Phosphodiesterase Inhibition by sildenafil


type 5 and zaprinast Inositol 1,4,5,
Soluble guanylyl -triphosphate
Nitric oxide cyclase

GTP
Activation Ca2+
cGMP
cGMP-dependent Inhibition
protein kinase of calcium release
Activation
Vascular
smooth muscle Decreased Sarcoplasmic
cell sensitivity of myosin reticulum
Inhibition

Phosphory- Myosin light-chain L-type calcium


lated myosin phosphatases Myosin channel
(contraction) (relaxation) Activation
Ca2+
K+

Calcium-sensitive
potassium channel

Figure 1 Regulation of the relaxation of vascular smooth muscle by nitric oxide (NO).5

type 5 phosphodiesterase.5 Inhaled nitric oxide (iNO) is the first as a substrate diffuses into smooth muscle to activate soluble
virtually selective pulmonary vasodilator in contrast to other guanylyl cyclase (sGC) to form cGMP and thus maintain basal
medications. Tolazoline, nitroprusside or prostaglandins E1 and D2, vascular tone. Intravascular destruction of NO by intraerythrocytic
which also decrease pulmonary vascular resistance, but as non- hemoglobin is limited by diffusional barriers, including the
selective vasodilating compounds, often has profound systemic red-cell-free layer adjacent to the endothelium and factors
effects (Figures 1 and 2). in and around the erythrocyte membrane that slow NO transit
The biochemical fates of iNO at the alveolar-capillary (Figure 3). However, sufficient NO is consumed to limit the
membrane: small amounts of nitrogen dioxide (NO2) may be activity of NO to the local environment. When NO is administered
formed if iNO mixes with high concentrations of oxygen (O2) in by inhalation, high intravascular NO concentrations promote
the air space. Depending on the milieu of the lung parenchyma, NO reactions, with erythrocyte hemoglobin and plasma proteins
NO may react with reactive oxygen species to form reactive nitrogen that can either protect it, thereby promoting systemic
species such as peroxynitrite. In the vascular space, dissolved NO is vasodilatation, or destroy it. During hemolysis or the infusion of
scavenged by oxyhemoglobin (forming methemoglobin and hemoglobin-based blood substitutes, cell-free ferrous hemoglobin
nitrate) and, to a lesser extent, plasma proteins (for example, in the plasma rapidly destroys NO by being oxidized to
forming nitrosothiols, which are stable intravascular sources of NO methemoglobin and nitrate ions. Cell-free hemoglobin may
activity).5 diffuse into extravascular spaces. Limited NO bioavailability under
The fate of intravascular NO is dose-dependent: Under normal these conditions promotes systemic vasoconstriction and organ
conditions, NO produced by endothelial NO synthase with arginine dysfunctions.6

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S86

Air space Release of


O2 NO2 reactive
Type II
oxygen species
alveolar cell
Nitric oxide
Type I
alveolar cell Formation of
reactive nitrogen
species

Inactivation by
hemoglobin
Red cell Formation of
S-nitrosothiols

Plasma proteins
Leukocyte

Vascular space Endothelial cell

Figure 2 Biochemical fates of inhaled nitric oxide (iNO) at the alveolar–capillary membrane.5

Pharmacology Physiology Pathology

Blood vessel NO concentration

-cys93 S NO
  NO+ Erythrocytes Diffusion
  barrier (plasma)
II NO2–
  Fe NO
 

  Fe II – O2 + NO•   Fe III + NO3
   
pO2 pO2
pH pH
H+
NO•

Plasma SNO NO• Xanthine NO


oxidoreductase NO• synthase
Arginine NO•
NO Citrulline
synthase
NO•
Smooth muscle cells
sGC Endothelial cells

Blood vessel

Figure 3 A model of the interactions of iNO (NO) with erythrocytes and cell-free hemoglobin in an arterial blood vessel.6

Early clinical trials vasodilation. Pulmonary hypertension resumed within minutes of


ceasing NO inhalation. While inhaling NO, there was an improved
In 1991, Frostell et al.7 showed in a lamb model that iNO (5 to oxygenation and cardiac output and decreased pulmonary artery
80 p.p.m.) can reverse pulmonary hypertension, without systemic pressure and pulmonary vascular resistance.

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S87

Figure 5 Serial changes in arterial/alveolar oxygen tension ratio (a/AO2) for


six patients over 24 h of NO inhalation.10

Figure 4 Change of preductal (J) and postductal (&) oxygen saturation 5 p.p.m. for up to 4 days) in 248 infants who were randomized to
(SpO2) in infants with persistent pulmonary hypertension of the newborn (PPHN) iNO or placebo: ECMO treatment was again significantly lower in
before and during inhalation of 80 p.p.m. NO at FiO2 0.9 wP<0.05. the iNO group (38 vs 64%). In addition, this was the first study to
show in term infants a reduction in the risk for chronic lung
disease (7 vs 20%).13 Because ECMO was used as a rescue therapy,
The first human study with iNO was conducted in eight adults
none of these studies showed a change in mortality.
with pulmonary hypertension.8 Pulmonary vascular resistance fell
Endogenously released and exogenously iNO may influence
with iNO in all patients. This was not associated with a decrease in
many facets of the developing perinatal lung, including the lung
systemic vascular resistance, in contrast to treatment with
parenchyma, bronchi and vascular structures.14 In a fetal or
intravenous prostacyclin.
preterm lung during the transition from a saccular to an alveolar
Roberts et al.9 and Kinsella et al.10 described in 1992 the first
stage (25 to 28 weeks of gestation), endogenous NO is released
use of iNO for the treatment of persistent pulmonary hypertension
primarily from epithelial and endothelial cells that contain NO
of the newborn (PPHN). Roberts treated six newborn infants with
synthase, and it is implicated in the structural and functional
PPHN with up to 80 p.p.m. of iNO. In all six patients, preductal
aspects of the development of pulmonary vasculature and airway
oxygen saturation increased; in five infants, postductal oxygen
smooth muscle. NO also contributes to growth of the lung
saturation and oxygen tensions also increased (Figure 4).
parenchyma and to extracellular matrix deposition and may
Inhalation of NO did not cause systemic hypotension.
modulate surfactant and inflammation in the developing lung.
Kinsella used a lower dose (10 to 20 p.p.m.) in nine newborn
The implications of supplementation with iNO in preterm infants
infants with PPHN. All showed a rapid improvement in
are15 beyond the scope of this review.
oxygenation without a reduction in systemic blood pressure
(Figure 5). This study was the first to show the sustained effect of
low-dose iNO in maintaining adequate oxygenation. Potential beneficial effects of iNO
Larger randomized controlled studies examining different
Thus, given the multiple mechanisms of action exerted by NO on
strategies of treatment with iNO in acute severe PPHN soon
the lung, its benefit may be mediated in several ways. Several are
followed these pilot studies. The NINOS trial enrolled 235 term and
listed here:
near-term newborn infants with hypoxic respiratory failure,
randomized to receive either iNO (20 p.p.m.) or a standard medical (1) Improved V/Q matching: Hypoxic pulmonary
treatment (FiO2 100%). Infants receiving iNO were significantly less vasoconstriction minimizes ventilation–perfusion
likely to be treated with extracorporeal membrane oxygenation mismatching in the presence of abnormal ventilation. iNO
(ECMO).11 improves oxygenation by increasing blood flow to ventilated
In another study,12 Roberts randomized 58 newborn infants lung units.5
with PPHN to either iNO (80 p.p.m.) or placebo: in 53% of infants (2) Improved PVR/SVR ratio: This results in a decrease in R to L
treated with iNO, oxygenation improved as compared with 7% in shunting at the ductal and foramen level and an improvement
the control group. The use of ECMO was again significantly in oxygenation.
decreased in the iNO group (40 vs 71%). In the final randomized, (3) Enhancement of pulmonary surfactant activity: Neonatal
controlled trial, which led to the FDA approval, Clark et al. studied animal models showed that low-dose iNO decreased or
a low-dose iNO therapy (20 p.p.m. in the first 24 h and then prevented the O2-induced detrimental effects on alveolar

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S88

surfactant, improved the apparent ability of hydrophobic segments are hyperinflated, increasing the dead space. In a
surfactant proteins to promote low-surface tension and improved heterogeneous, patchy, a form of lung disease, iNO may improve
lung volume by a process unrelated to surfactant function.15 the V/Q matching by dilating the vasculature bed around the
(4) Prevention of neomuscularization in patients with PPHN: ventilated segments. In a homogenous parenchymal lung
In a model of injured pup lungs, iNO protected against disease, the response to iNO may be low: treatment of the
pulmonary remodeling induced by lung injury by mechanisms underlying lung disease and better recruitment of the atelectatic
that are independent of pulmonary tone, inflammation or lung may improve the response to iNO.
thrombosis.16  Left ventricular dysfunction.
(5) Decreased reperfusion injury: A neonatal animal model of
It is important to evaluate and treat each of these factors when
lung ischemia–reperfusion showed that iNO can prevent
treating the hypoxemic infants.
microvascular injury, endothelial dysfunction and pulmonary
neutrophil accumulation.16 Echocardiography. Echocardiography is crucial in the
(6) NO replacement: NO, produced by endothelial NO synthase, is evaluation and management of the hypoxemic (blue) baby. The
critically involved in the cardiopulmonary transition from fetal study will measure evidence for PPHN: existence of patent ductus
to neonatal life. Endothelial NO synthase protein expression arteriosus and patent foramen ovale in the direction of the shunt,
and mRNA were decreased by a half and two-thirds, and the pressure at the right ventricle. PPHN can be also shown
respectively, in an animal model of pulmonary hypertension by the evidence of right-to left-extrapulmonary shunting, by a
(Figure 6).17 difference between the preductal and the postductal O2 saturation.
Currently, the best way to distinguish between a congenital
Clinical considerations
heart lesion, which is dependent on right-to-left shunting (for
Hypoxic respiratory failure of the term near-term infant:
example, interrupted aortic arch, total anomalous pulmonary
Hypoxemia of the near-term infant is caused by complex
venous return and hypoplastic left heart syndrome) in which
pathopysiological pathways that have a common end point of
treatment with iNO may not be indicated, other anatomical
reduced perfusion of the lungs. In some patients, one of these
variations or a normal heart with maladaptation to the
mechanisms dominates but in others several mechanisms play
extrauterine circulation is with echocardiography.
along, contributing to the severe clinical picture.
Echocardiography is also important to evaluate the cardiac
 Patent ductus arteriosus and/or patent foramen ovale with right- performance and assess ventricular function.
to-left extrapulmonary shunting.
 V/Q mismatching in the lungs: as in meconium aspiration Timing of treatment. Large-scale randomized studies support
syndrome. In some segments of the lungs, there is reduced the efficacy of iNO in term and near-term infants (>34 weeks of
ventilation and right-to-left intrapulmonary shunting, and other gestation) in the first 2 weeks of life.18 iNO has also been used
post-ECMO. iNO has also been used in the treatment of late
pulmonary hypertension complicating chronic lung disease with
some reported success,19,20 although much of the data are
anecdotal. The treatment of the premature infant is still under
investigation and is beyond the scope of this review.

Indication for treatment. Most of the studies that tested the


efficacy of iNO in acute respiratory failure used enrolled infants
with oxygenation index (OI) of 25 or above.18 Most evidence
supports the use of iNO to reduce the need for ECMO and/or death.
Some studies that used less-stringent entry criteria showed
improved oxygenation but did not reduce ECMO/death when
compared with the more ‘traditional’ criteria.21,22

Dose. The first clinical trials used higher initial doses of


80 p.p.m. Subsequent studies changed the practice to an initial
dose of 20 p.p.m. A review of the iNO treatment of 476 neonates
revealed that starting at higher doses does not improve the major
Figure 6 Pulmonary endothelial NO synthase gene expression is decreased in outcomes (ECMO and death), and there was no evidence that
fetal lambs with pulmonary hypertension.17 *P<0.05. using higher dose leads to a more rapid improvement of the

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S89

hypoxemia. Using higher does of iNO (>22 p.p.m.) led to an A possible explanation for this phenomenon might be through
increase in methemoglobin levels. This may decrease the oxygen- an increase in the concentration of superoxide and peroxynitrite
carrying capacity by hemoglobin. Using lower doses of iNO when treating with iNO. In an animal model, reactive oxygen
(<18 p.p.m.) achieved the same response of PaO2 as the higher species scavenging ameliorated alterations in endogenous NO
doses, and there were no differences in the adverse events.23 signaling during iNO therapy (Figure 9).27

Risks and complications Direct toxicity. At high inspired levels, NO is directly toxic to
Methemoglobinemia. The incidence of methemoglobinemia has tissues and can cause methemoglobinemia, formation of NO2,
been low in clinical trials, occurring more commonly in neonates pulmonary edema, alveolar hemorrhage and hypoxemia. At low
and with high inhaled doses, but usually is tolerated well. inspired levels of iNO (20 p.p.m.) NO2 formation is minimal.28

Left ventricular dysfunction Bleeding tendency. iNO is associated with increased bleeding
Inhaled NO may have adverse hemodynamic effects in patients time and inhibition of platelet aggregation in adults.29,30 A
with significant preexisting left ventricular dysfunction. Studies Cochrane review of randomized studies of iNO in the
with iNO in adults with preexisting, severe left ventricular near-term infants did not find an association between the use
dysfunction showed an increase in the left ventricular end-diastolic of iNO and bleeding events.18
pressure.24,25 This increase in end-diastolic volume resulting from
an iNO-induced increased pulmonary perfusion may not be New information
tolerated by the poorly compliant left ventricle resulting in The new clinical data emerging in the literature concentrate on the
worsened, rather than improved, oxygenation. safety of iNO: A study in premature infants dropped the concerns
that iNO might have a deleterious effect on surfactant proteins and
function. This study showed a transient improvement of surfactant
Withdrawal. A randomized control study of iNO also examined function after iNO treatment,31 and that administration of iNO does
the safety of its withdrawal in 155 term infants with pulmonary not increase inflammatory markers in the alveolar fluid.32
hypertension. For infants responding to the treatment, a
dose–response relationship between the iNO dose and decrease in Neurodevelopmental outcome. Concerns raised regarding the
PaO2 after discontinuing iNO was found (Figure 7). A reduction in long-term complications of long-term use of iNO. In a study that
iNO to 1 p.p.m. before discontinuation of the drug seems to examined the effects of iNO in premature infants, the infants in the
minimize the observed decrease in PaO2. For infants failing study group had improved neurodevelopmental outcomes at 2 years
treatment, discontinuation of iNO could pose a life-threatening of age as compared with the placebo group.33 A follow-up study at
reduction in oxygenation (Figure 8).26 18 to 24 months of age of the infants participating in the NINOS
study showed that early iNO therapy (OI: 15 to 25) as compared

Figure 7 Changes in PaO2 (no ventilator changes), 30 min after cessation of Figure 8 Percent change in oxygenation on withdrawal of placebo or NO study
inhaled NO treatment gas (masked) in patients with persistent pulmonary gas (5, 20, or 80 p.p.m.) in persistent pulmonary hypertension of the newborn
hypertension of the newborn (PPHN) treated successfully.26 (PPHN) patients declared treatment failures.26

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S90

Figure 9 Reactive oxygen species scavenging. Lung tissue nitric oxide synthase (NOS) activity and endothelial NOS (eNOS) protein levels after inhaled NO (NO) therapy
in control lambs. (a) Relative NOS activity is decreased after 24 h of inhaled NO. (b) Lung tissue eNOS protein levels, as determined by western blot analysis, are not
changed after 24 h of inhaled NO.27 *P<0.05.

with late therapy (OI>25) for respiratory failure in term and 70


Placebo
Change in 6-min walking distance (m)

near-term infants was not associated with an increase in 60 20 mg of sildenafil


40 mg of sildenafil
neurodevelopmental impairment or hearing loss.21 This new data 50 80 mg of sildenafil
suggest that iNO might be beneficial for the developing brain. 40

30
Congenital diaphragmatic hernia. The pulmonary artery 20
hypertension in congenital diaphragmatic hernia is, in part, 10
secondary to underdevelopment and anatomical changes of the
0
pulmonary vasculature (that is, maladaptation). In addition, left
–10
ventricular dysfunction and surfactant dysfunction may contribute
–20
to hypoxemia. These may lead to a different approach in treating 0 4 8 12
the hypoxemic respiratory failure in these patients. Two studies that Week
randomized infants with congenital diaphragmatic hernia to iNO Figure 10 Mean changes from baseline, with 95% confidence intervals, in the 6-
or placebo13,34 failed to show improved outcome in terms of min walking distance at week 12 in the placebo and sildenafil groups.38
reduced use of ECMO or death.
Non-invasive low-dose iNO, delivered via nasal canula at the
time of tracheal extubation, may benefit a subset of infants with cGMP-specific phosphodiesterase inhibitors (sildenafil)
congenital diaphragmatic hernia who cannot be weaned from iNO The secondary messenger of NO, cGMP, is inactivated
after surgical correction.35 In this subset of patients, pulmonary predominantly by PDE5 (phosphodiesterase type 5). Sildenafil, an
vascular hyperactivity persisted in spite of parenchymal inhibitor of PDE5, is a selective pulmonary vasodilator, partially
improvement. The role of iNO is likely far more complicated than because PDE5 is highly expressed in the lung.5
simply vasodilation alone, as the NO/cGMP pathway is also
involved in alveolar growth and lung angiogenesis.36
Amelioration effects of iNO withdrawal. Rebound pulmonary
hypertension caused by withdrawal of iNO can be markedly
Future directions and research consideration attenuated by increasing cGMP with sildenafil.37
Unfortunately, not all infants with PPHN improve after the
initiation of iNO therapy and refractory PPHN is common. Other Therapy for pulmonary hypertension. A randomized
pharmacologic modalities are important. On the basis of the placebo-controlled study in 278 adult patients with symptomatic
regulation pathways that control the production of the NO–cGMP pulmonary arterial hypertension, treatment with oral sildenafil
cascade, and other physiologic mechanisms that induce pulmonary showed an improvement in exercise capacity, WHO functional class
vasoconstriction, the following pharmacological strategies might be and hemodynamics (Figure 10).38
useful in the prevention or amelioration of PPHN. Some of these A model for meconium aspiration in piglets tested the efficacy of
have been studied in humans (adults or pediatric population) and the intravenous form of sildenafil. Sildenafil completely reversed
some are still experimental, shown to be effective in animal studies. the increase in pulmonary vascular resistance, and was shown to

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S91

be a highly effective pulmonary vasodilator, which is at least as human superoxide dismutase, with or without iNO, increased
effective as iNO.39 oxygenation and reduced pulmonary vasoconstriction.48
The effectiveness of the aerosolized form of sildenafil was tested Expanding the pharmacologic options and the combination of
in an animal model of PPHN. Sildenafil selectively decreased the several of these might improve the response to iNO and the clinical
pulmonary artery pressure. When Sildenafil was inhaled while outcome of this devastating disease.
simultaneously breathing NO, the pulmonary artery pressure
decreased even further.40 Summary
Human studies followed the animal studies for PPHN. Six
Since its discovery two decades ago, NO found its way to the arsenal
patients with PPHN were randomized to enteral sildenafil and
of therapeutic approaches available to the neonatologist in the
compared with seven patients who received placebo. Oral sildenafil
treatment of PPHN, a disease with high morbidity and mortality.
improved OI in infants with severe PPHN, which suggests that oral
Vast research, both in animal models and human, increased our
sildenafil may be effective in the treatment of PPHN.41
understanding of the features of this gas and its benefits and
Soluble guanylate cyclase activators downsides. iNO was found to be the first potent ‘fairly’ selective
BAY 41-2272 is a novel direct activator of soluble guanylate cyclase. pulmonary vasodilator. NO has multiple mechanisms of actions
This compound was tested in an animal model for PPHN. The that may benefit newborn infants with diseases associated with
effectiveness of BAY 41-2272-induced pulmonary vasodilation meconium aspiration and persistent pulmonary hypertension.
increased during the development of pulmonary hypertension in a Understanding the mechanism of action of iNO through the cGMP
response that was greater than sildenafil. It also augmented the pathway led to the introduction of other medications in this
pulmonary vasodilation induced by iNO.42 pathway: in addition to increasing cGMP concentration through
NO-induced stimulation, inhibiting cGMP metabolism through
PGI2 analogs phosphodiesterase inhibition, by sildenafil, for example, may
Iloprost is a stable analog of prostacyclin. Its mechanism of action provide additional benefit. The combination of drugs affecting
is through a different signaling pathway, involving cAMP. The use through this pathway and other pathways might have a synergistic
of the inhaled formulation has been approved by the FDA for the effect, increasing the rate of success in the treatment of PPHN.
use in adults. When given as the inhaled formulation, iloprost is a
selective pulmonary vasodilator. A first study of the use of inhaled Disclosure
iloprost for children with pulmonary artery hypertension has been
MD Schreiber has received lecture fees, consulting fees, and grant support from
recently published, reporting sustained functional improvement in Ikaria. A Bin-Nun has declared no financial interests.
some children, although occasionally induced
bronchoconstriction.43 There are several anecdotal reports for the Duality of interest
use of inhaled iloprost for refractory cases of PPHN,44 most Dr Schreiber has received grant support and honoraria from iNO Therapeutics/
reporting improved oxygenation after the initiation of therapy. No IKARIA.
adverse events were reported.
References
Endothelin-1 receptor antagonists (bosentan, sitaxsentan)
1 Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of
Endothelin-1 is a potent vasoconstrictor, and has been shown to arterial smooth muscle by acetylcholine. Nature 1980; 288(5789): 373–376.
participate in the regulation of the pulmonary vascular tone.45 A 2 Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endothelium-derived relaxing
Cochrane review of this class of drugs in conjunction with factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci
conventional therapy has shown that it can improve the clinical USA 1987; 84(24): 9265–9269.
outcomes in adults with idiopathic pulmonary artery 3 Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological
activity of endothelium-derived relaxing factor. Nature 1987; 327(6122): 524–526.
hypertension.46 Almost no experience with ERAs exists in the
4 Murad F. The 1996 Albert Lasker Medical Research Awards. Signal transduction using
neonatal population. nitric oxide and cyclic guanosine monophosphate. JAMA 1996; 276(14): 1189–1192.
5 Griffiths MJ, Evans TW. Inhaled nitric oxide therapy in adults. N Engl J Med 2005;
Rho-kinase inhibitors (fasudil, Y-27632) 353(25): 2683–2695.
Rho-kinase inactivates myosin light-chain phosphatase and 6 Schechter AN, Gladwin MT. Hemoglobin and the paracrine and endocrine functions of
promotes vasoconstriction. The inhalation of rho-kinase inhibitor nitric oxide. N Engl J Med 2003; 348(15): 1483–1485.
in an animal model achieved pulmonary vasodilation.47 7 Frostell C, Fratacci MD, Wain JC, Jones R, Zapol WM. Inhaled nitric oxide. A selective
pulmonary vasodilator reversing hypoxic pulmonary vasoconstriction. Circulation
1991; 83(6): 2038–2047.
Superoxide dismutase 8 Pepke-Zaba J, Higenbottam TW, Dinh-Xuan AT, Stone D, Wallwork J. Inhaled nitric
Hyperventilation and the formation of reactive oxygen species can oxide as a cause of selective pulmonary vasodilatation in pulmonary hypertension.
inactivate iNO. In an animal model, intratracheal recombinant Lancet 1991; 338(8776): 1173–1174.

Journal of Perinatology
iNO and PVR
A Bin-Nun and MD Schreiber
S92

9 Roberts JD, Polaner DM, Lang P, Zapol WM. Inhaled nitric oxide in persistent 29 Hogman M, Frostell C, Arnberg H, Hedenstierna G. Bleeding time prolongation and NO
pulmonary hypertension of the newborn. Lancet 1992; 340(8823): 818–819. inhalation. Lancet 1993; 341(8861): 1664–1665.
10 Kinsella JP, Neish SR, Shaffer E, Abman SH. Low-dose inhalation nitric oxide in 30 Samama CM, Diaby M, Fellahi JL, Mdhafar A, Eyraud D, Arock M et al. Inhibition of
persistent pulmonary hypertension of the newborn. Lancet 1992; 340(8823): 819–820. platelet aggregation by inhaled nitric oxide in patients with acute respiratory distress
11 The Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in full-term and syndrome. Anesthesiology 1995; 83(1): 56–65.
nearly full-term infants with hypoxic respiratory failure.. N Engl J Med 1997; 336(9): 31 Ballard PL, Merrill JD, Truog WE, Godinez RI, Godinez MH, McDevitt TM et al.
597–604. Surfactant function and composition in premature infants treated with inhaled nitric
12 Roberts Jr JD, Fineman JR, Morin III FC, Shaul PW, Rimar S, Schreiber MD et al. oxide. Pediatrics 2007; 120(2): 346–353.
Inhaled nitric oxide and persistent pulmonary hypertension of the newborn. The 32 Truog WE, Ballard PL, Norberg M, Golombek S, Savani RC, Merrill JD et al.
Inhaled Nitric Oxide Study Group. N Engl J Med 1997; 336(9): 605–610. Inflammatory markers and mediators in tracheal fluid of premature infants treated
13 Clark RH, Kueser TJ, Walker MW, Southgate WM, Huckaby JL, Perez JA et al. Low-dose with inhaled nitric oxide. Pediatrics 2007; 119(4): 670–678.
nitric oxide therapy for persistent pulmonary hypertension of the newborn. Clinical 33 Mestan KK, Marks JD, Hecox K, Huo D, Schreiber MD. Neurodevelopmental
Inhaled Nitric Oxide Research Group. N Engl J Med 2000; 342(7): 469–474. outcomes of premature infants treated with inhaled nitric oxide. N Engl J Med 2005;
14 Martin RJ, Walsh MC. Inhaled nitric oxide for preterm infantsFwho benefits? N Engl 353(1): 23–32.
J Med 2005; 353(1): 82–84. 34 The Neonatal Inhaled Nitric Oxide Study Group (NINOS). Inhaled nitric oxide and
15 Roberts Jr JD, Chiche JD, Weimann J, Steudel W, Zapol WM, Bloch KD. Nitric oxide hypoxic respiratory failure in infants with congenital diaphragmatic hernia.. Pediatrics
inhalation decreases pulmonary artery remodeling in the injured lungs of rat pups. 1997; 99(6): 838–845.
Circ Res 2000; 87(2): 140–145. 35 Kinsella JP, Parker TA, Ivy DD, Abman SH. Noninvasive delivery of inhaled nitric oxide
16 Barbotin-Larrieu F, Mazmanian M, Baudet B, Detruit H, Chapelier A, Libert JM et al. therapy for late pulmonary hypertension in newborn infants with congenital
Prevention of ischemia–reperfusion lung injury by inhaled nitric oxide in neonatal diaphragmatic hernia. J Pediatr 2003; 142(4): 397–401.
piglets. J Appl Physiol 1996; 80(3): 782–788. 36 Ladha F, Bonnet S, Eaton F, Hashimoto K, Korbutt G, Thebaud B. Sildenafil improves
17 Shaul PW, Yuhanna IS, German Z, Chen Z, Steinhorn RH, Morin III FC. Pulmonary alveolar growth and pulmonary hypertension in hyperoxia-induced lung injury. Am J
endothelial NO synthase gene expression is decreased in fetal lambs with pulmonary Respir Crit Care Med 2005; 172(6): 750–756.
hypertension. Am J Physiol 1997; 272(5 Part 1): L1005–L1012. 37 Atz AM, Wessel DL. Sildenafil ameliorates effects of inhaled nitric oxide withdrawal.
18 Finer NN, Barrington KJ. Nitric oxide for respiratory failure in infants born at or near Anesthesiology 1999; 91(1): 307–310.
term. Cochrane Database Syst Rev 2006; (4): CD000399. 38 Galie N, Ghofrani HA, Torbicki A, Barst RJ, Rubin LJ, Badesch D et al. Sildenafil
19 Clark PL, Ekekezie II, Kaftan HA, Castor CA, Truog WE. Safety and efficacy of citrate therapy for pulmonary arterial hypertension. N Engl J Med 2005; 353(20):
nitric oxide in chronic lung disease. Arch Dis Child Fetal Neonatal Ed 2002; 86(1): 2148–2157.
F41–F45. 39 Shekerdemian LS, Ravn HB, Penny DJ. Intravenous sildenafil lowers pulmonary
20 Banks BA, Seri I, Ischiropoulos H, Merrill J, Rychik J, Ballard RA. Changes in vascular resistance in a model of neonatal pulmonary hypertension. Am J Respir Crit
oxygenation with inhaled nitric oxide in severe bronchopulmonary dysplasia. Care Med 2002; 165(8): 1098–1102.
Pediatrics 1999; 103(3): 610–618. 40 Ichinose F, Erana-Garcia J, Hromi J, Raveh Y, Jones R, Krim L et al. Nebulized
21 Konduri GG, Vohr B, Robertson C, Sokol GM, Solimano A, Singer J et al. Early inhaled sildenafil is a selective pulmonary vasodilator in lambs with acute pulmonary
nitric oxide therapy for term and near-term newborn infants with hypoxic respiratory hypertension. Crit Care Med 2001; 29(5): 1000–1005.
failure: neurodevelopmental follow-up. J Pediatr 2007; 150(3): 235–240, 240 e1. 41 Baquero H, Soliz A, Neira F, Venegas ME, Sola A. Oral sildenafil in infants with
22 Davidson D, Barefield ES, Kattwinkel J, Dudell G, Damask M, Straube R et al. Inhaled persistent pulmonary hypertension of the newborn: a pilot randomized blinded study.
nitric oxide for the early treatment of persistent pulmonary hypertension of the term Pediatrics 2006; 117(4): 1077–1083.
newborn: a randomized, double-masked, placebo-controlled, dose–response, multi- 42 Deruelle P, Grover TR, Abman SH. Pulmonary vascular effects of nitric oxide–cGMP
center study. The I-NO/PPHN Study Group. Pediatrics 1998; 101(3 Part 1): 325–334. augmentation in a model of chronic pulmonary hypertension in fetal and neonatal
23 Guthrie SO, Walsh WF, Auten K, Clark RH. Initial dosing of inhaled nitric oxide in sheep. Am J Physiol Lung Cell Mol Physiol 2005; 289(5): L798–L806.
infants with hypoxic respiratory failure. J Perinatol 2004; 24(5): 290–294. 43 Ivy DD, Doran AK, Smith KJ, Mallory Jr GB, Beghetti M, Barst RJ et al. Short- and long-
24 Loh E, Stamler JS, Hare JM, Loscalzo J, Colucci WS. Cardiovascular effects of inhaled term effects of inhaled iloprost therapy in children with pulmonary arterial
nitric oxide in patients with left ventricular dysfunction. Circulation 1994; 90(6): hypertension. J Am Coll Cardiol 2008; 51(2): 161–169.
2780–2785. 44 Kelly LK, Porta NF, Goodman DM, Carroll CL, Steinhorn RH. Inhaled prostacyclin for
25 Semigran MJ, Cockrill BA, Kacmarek R, Thompson BT, Zapol WM, Dec GW et al. term infants with persistent pulmonary hypertension refractory to inhaled nitric oxide.
Hemodynamic effects of inhaled nitric oxide in heart failure. J Am Coll Cardiol 1994; J Pediatr 2002; 141(6): 830–832.
24(4): 982–988. 45 Ivy DD, Parker TA, Ziegler JW, Galan HL, Kinsella JP, Tuder RM et al. Prolonged
26 Davidson D, Barefield ES, Kattwinkel J, Dudell G, Damask M, Straube R et al. Safety of endothelin A receptor blockade attenuates chronic pulmonary hypertension in the ovine
withdrawing inhaled nitric oxide therapy in persistent pulmonary hypertension of the fetus. J Clin Invest 1997; 99(6): 1179–1186.
newborn. Pediatrics 1999; 104(2 Part 1): 231–236. 46 Liu C, Chen J. Endothelin receptor antagonists for pulmonary arterial hypertension.
27 Oishi P, Grobe A, Benavidez E, Ovadia B, Harmon C, Ross GA et al. Inhaled nitric oxide Cochrane Database Syst Rev 2006; 3: CD004434.
induced NOS inhibition and rebound pulmonary hypertension: a role for superoxide 47 Nagaoka T, Fagan KA, Gebb SA, Morris KG, Suzuki T, Shimokawa H et al. Inhaled Rho
and peroxynitrite in the intact lamb. Am J Physiol Lung Cell Mol Physiol 2006; kinase inhibitors are potent and selective vasodilators in rat pulmonary hypertension.
290(2): L359–L366. Am J Respir Crit Care Med 2005; 171(5): 494–499.
28 Kirmse M, Hess D, Fujino Y, Kacmarek RM, Hurford WE. Delivery of inhaled 48 Lakshminrusimha S, Russell JA, Wedgwood S, Gugino SF, Kazzaz JA, Davis JM et al.
nitric oxide using the Ohmeda INOvent Delivery System. Chest 1998; 113(6): Superoxide dismutase improves oxygenation and reduces oxidation in neonatal
1650–1657. pulmonary hypertension. Am J Respir Crit Care Med 2006; 174(12): 1370–1377.

Journal of Perinatology
Journal of Perinatology (2008) 28, S93–S101
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Neurodevelopmental outcome of infants with meconium
aspiration syndrome: report of a study and literature review
N Beligere and R Rao
Department of Pediatrics, University of Illinois at Chicago Medical Center, Chicago, IL, USA

when MDI and PDI scores were >85 to 110; mildly delayed when scores
There is a paucity of information on long-term outcome of infants who
were >70 to 84; and severely delayed if the scores were <69. In addition,
have suffered from meconium aspiration syndrome (MAS) in the neonatal
neurological evaluation also confirmed the disability. The report is based on
period. We analyzed long-term developmental outcome data of 35 infants
the final analysis of 29 infants. Data of six infants were not included in the
who were admitted to the neonatal intensive care unit (NICU) at the
final analysis because of incomplete information. The mean BW of the
University of Illinois Hospital at Chicago (UICMC) with a diagnosis of MAS,
infants was 3269±671 g; mean gestational age was 39.5±3.1 weeks. The
and we reviewed the literature pertinent to the subject. The objective of the
median APGAR score at 10 was 4, and at 50 was 6. Out of 29, 11 (38%)
study was to assess the neurodevelopment status of MAS infants and
infants were normal. Out of 29, 2 infants (7%) had cerebral palsy (CP) and
compare the possible effects of different variables that are known to affect
4 (14%) had severe delay at 12 months of age. Out of 29, 2 who were
the later developmental outcome. The variables included mode of delivery,
neurologically disabled had PDI <69. Out of 29, 12 (41%) had mild delay
APGAR score, cord pH, mode of treatment, and neurological findings during
in speech. No statistical difference in neurodevelopment was found in
the course of NICU. The infants were enrolled in the developmental follow-
infants born vaginally or by C-section. Our findings show poor outcome (CP
up program (DFUP) after discharge from the nursery for assessment of
and global delay) in 21% of infants who suffered MAS, even though the
long-term developmental status and neurodevelopmental outcome. In order
majority of the infants (26/29) responded to conventional ventilator support
to assess the impact of the treatment on long-term outcome and compare
alone. No difference was found in the outcome of infants between NSVD vs
our findings with previously published reports, we also reviewed the
C-section delivery. These findings suggest that infants with the diagnosis of
previously published literature on neurodevelopment outcome of infants
MAS manifest later neurodevelopmental delays, even if they respond well to
treated for MAS (with different modalities) during the last three decades.
conventional treatment. This abstract was presented at the Society for
Total of 35 infants with a diagnosis of MAS admitted to the NICU at UICMC
Pediatric Research Annual Meeting, 2000.
were followed in the DFUP clinic for 3 years during January 1999 to
September 2001. The medical records of these infants were reviewed for the
Journal of Perinatology (2008) 28, S93–S101; doi:10.1038/jp.2008.154
mode of delivery, APGAR score, birth weight (BW), gestational age, mode of
treatment during the neonatal period, and neurodevelopment status. 19/35
(54%) infants were delivered vaginally, 16/35 (46%) by cesarean section
(C-section). All were treated in the delivery room using the standard Introduction
resuscitation protocol. Following initial resuscitation, all except three Meconium aspiration syndrome (MAS) is a clinical entity that
required intubation and ventilation for varying duration. One infant contributes to the high incidence of mortality and morbidity among
required inhaled nitric oxide therapy, and two required extracorporeal term pregnancies.1 The prevalence and mortality in MAS is noted to
membrane oxygenation treatment. Subsequent to discharge, the infants be steadily decreasing with newer and more efficient modalities of
were evaluated in the clinic at 2 months of age, and then every 4 months treatment.1,2 With steady improvement in survival, the amount of
up to 3 years. The developmental assessment of mental development index morbidity among the survivors has not changed. There is a great
(MDI), psychomotor development index (PDI), and behavior rating scale need for follow-up data on the neurodevelopmental outcome of this
(BRS) were obtained using the Bayley II infant motor scale, and group of infants. The purpose of this study was to report our findings
neurodevelopment evaluation was performed using the Amiel–Tison of neurodevelopmental outcome of infants with the diagnosis of MAS
technique. Speech evaluation was performed in infants >18 months using in the neonatal period and review the literature on this supplement.
the Rossetti Infant–Toddler language scale. Infants were considered normal

Correspondence: Dr N Beligere, Division of Neonatology, Department of Pediatrics, University Methods


of Illinois at Chicago Medical Center, 840 South Wood Street, M/C 808, Chicago, IL 60612,
USA. We reviewed the medical records of 35 infants with the primary
E-mail: beligere@uic.edu diagnosis of MAS followed in the developmental follow-up program
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S94

(DFUP) clinic during January 1999 to September 2001 at the anthropometric measurements were recorded. Infants were
University of Illinois Hospital at Chicago (UICMC). Information independently evaluated using the Bayley II infant motor
was collected using the specific diagnostic code for MAS ICD-907 developmental scale4 by a certified pediatric occupational
criteria recorded in the obstetric and neonatal intensive care units therapist, physical therapist, and the developmental specialist.
(NICUs). The criteria for the diagnosis of MAS (ICD-907) include: Infants were scored based on their performance on the Bayley II
history of meconium-stained amniotic fluid before delivery, the infant motor scale. Scores of mental development index
presence of meconium below the vocal cords at the time of birth in (MDI), psychomotor development index (PDI), and behavior
infants >37 weeks of gestation, evidence of respiratory distress, and rating scale (BRS) were also used as criteria for eligibility to enroll
radiological evidence of aspiration pneumonitis. Many of these in the early intervention program (EIP). Acquisition of
infants also developed persistent pulmonary hypertension (PPHN). language was evaluated by the certified pediatric speech pathologist
The medical records of the infants who met the above criteria were after the age of 18 months using the Rossetti infant–Toddler
selected for the retrospective analysis of perinatal and neonatal language scale.5
events. The study was approved by the internal review board at the The degree of disability was further classified using one or more
University of Illinois at Chicago. of the following criteria. When the PDI <2 s.d. below the mean of
Obstetrical and NICU registry and medical records were reviewed p69, the baby’s condition was classified as cerebral palsy (CP)
for maternal complications of pregnancy, such as diabetes, with neurological evidence of spasticity. When the MDI <2 s.d.
hypertension, toxemia, and chorioamnionitis. The presence of below the mean of p69 for evidence of mental disability, the baby
meconium-stained amniotic fluid, consistency of meconium, and was considered as cognitive delay. Mild delay was considered when
presence of amnioinfusion were also ascertained. The presence of the scores of PDI or MDI were X70–84, with clinical evidence of
fetal distress and the mode of delivery were also noted. The infant’s hypotonia (gross motor delay or fine motor) and/or cognitive
condition in the delivery room and any interventions were delay. Speech delay was considered when expressive or receptive
reviewed. The following data were retrieved from the infants chart: function of communication was delayed according to the Rossetti
APGAR score at 1 and 5 min, presence of meconium below the infant–Toddler language scale.5 Diagnoses of speech delay or
vocal cords, method of resuscitation, presence of respiratory distress hearing impairment were assigned according to the disability. The
at birth, the requirement of the use of supplemental oxygen, and children with mild and severe delays were enrolled in the state-
ventilator support. The data of cord blood gases and blood gas supported EIP between 4 and 6 months of age. The EIP includes
analysis at the time of admission to the NICU were also noted. The speech therapy, physical or occupational therapy, and
gestational age was determined using fetal ultrasound assessment developmental therapy as needed.
or Ballard score. Birth weight (BW), length, and head
circumference on admission to the NICU were recorded.
Analysis and results
Developmental assessment Maternal data
During the period of 1999 to 2001, 75 infants were diagnosed as Complete maternal data were available in all 29 infants who had
having meconium-stained amniotic fluid at the time of delivery. Of complete follow-up data. Total of 16 of these infants were delivered
these, only 40 infants who developed respiratory distress and met by C-section for fetal distress. 13 infants were delivered by normal
the criteria for MAS were admitted to the NICU. Parents of all spontaneous vaginal delivery. Pregnancy complications included
infants who were admitted to NICU with a diagnosis of MAS were diabetes in 4 patients who were on insulin. Pregnancy induced
counseled by the clinic nurse and enrolled for developmental hypertension in 3, fetal decelaration in 4, chorioamnionitis
follow-up subsequent to discharge. Only 35 infants attended the in 3, premature rupture of membranes in 3 maternal drug use
follow-up clinic for varying periods of time during the 3-year study in 6, sickle-cell disease in 4, fever of unknown origin in 3, and
period. Among this group, medical records of six infants’ did not presence of prolapsed cord in 3. Umbilical cord pH was available
have complete information; hence, they were not included in the in 16/18 inborn infants and in 2/11 outborn infants. It was noted
final analysis of this study. The final report includes information 8/18 infants had cord pH>7.2 and in two infants cord pH was
on 29 infants. 6.75 and 7.10.
Developmental evaluation was conducted at 2, 4, 8, 12, 18, 24,
30, and 36 months. Each infant had at least five visits during this Neonatal data
period. These evaluations consisted of neurodevelopment Table 1 shows the perinatal data of infants in the study. Total of 11
assessment by an experienced developmental pediatrician (NB) outborn infants were transferred to UICMC within 12 h after birth.
using the Amiel–Tison neurodevelopment scale and neurological Method of resuscitation was not available. Mean admission age was
examination.3 During these visits, complete interim history, 6 h after birth APGAR score ranged from 0 to 7 at 1 min with mean
immunization status, and physical exams including of 4, and at 5 min APGAR score ranged from 4 to 9 with mean of 6.

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S95

Table 1 Demographics and birth weight gestational age Table 2 Developmental outcome

Inborn 18 Normal 11 38%


Outborn 11
Gestation age (weeks) 39.5±3.1 Abnormal findings
Birth weight (g) 3269±671 Mild defectsa 12 41%
Cerebral palsy 2 7%
APGAR score Severe global delay 4 14%
10 Range 0–7 median of 4
50 Range 0–9 median of 6 Entry in early intervention program (EIP) 19 65%
a
Includes mild speech delay or mild hypotonicity without any motor or cognitive abnormality.

We were unable to get the fetal heart rate pattern in many of the Neurodevelopmental outcome did not differ whether they were born
infants therefore not included in our data. Thick meconium fluid vaginally or by C-section.
at the time of delivery in 14 infants and meconium was present
below the vocal cord in four infants. Mean gestational age was
39.5±3.1 weeks. All infants were resuscitated in the delivery room, Discussion
in accordance with the standard NRP protocol.6 All infants were MAS is a distinct clinical syndrome that occurs in term infants.
admitted to the NICU, all required assisted ventilation. Duration of Severe meconium aspiration occurs in one in 500 deliveries: that
the ventilatory support varied between 2 and 12 days with a mean is, in only 0.2% of all births.1,7 Intrauterine fetal distress and fetal
of 6.4 days. Only one infant was treated with inhaled nitric oxide asphyxia in utero before birth have been recognized as stimulating
(INO) and high frequency ventilaton who had severe metabolic and enhancing the intestinal peristalsis of the infant, resulting in
acidosis and pulmonary hypertension. Two infants required relaxation of the anal sphincter and passage of meconium in
extracorporeal membrane oxygenation (ECMO) for 12 to 24 days utero.8,9 Previous reports8,10 have described the pathophysiology of
who did not respond to mechanical ventilation. MAS. The hypoxic distress of the fetus and gasping in utero have
The neurological findings in the neonatal period showed: been speculated as the cause of the meconium aspiration during
fourteen infants developed seizure activity during the first labor and at birth. Aspiration of thick meconium may occur during
week of life. All had electroencephalography (EEG) recordings. the respiratory effort of the first breath. This leads to obstruction of
Six of the EEGs demonstrated abnormal foci in the temporal airways, resulting in profound hypoxia. In addition, meconium
and parietal areas. Thirteen infants had magnetic resonance may lead to chemical pneumonitis, parenchymal lung damage,
imaging (MRI) scans during the neonatal course. Eleven infants and inactivation of surfactant. Other complications such as
had normal MRI, and two infants demonstrated cerebral pulmonary hypertension or pulmonary atelectasis with or without
hemorrhage. pneumothorax have been reported. Severe hypoxia may cause
brain injury and hypoxic ischemic encephalopathy.
Developmental outcome data Since the recognition of these issues, both obstetricians and
Table 2 shows developmental outcome at 36 months. In 38% neonatologists have intensified their efforts to decrease the
(11/29) of infants, the developmental status at 3 years of age was incidence of MAS using new interventions. The obstetric preventive
normal. 41% (12/29) had mild deficits, which consisted of mild interventions include amnioinfusion in the presence of thick or
hypotonia or mild speech delay. 7% (2/29) had CP. 14% (4/29) thin meconium-stained amniotic fluid11 before birth of the infant,
had severe global delay. None required oral or gastric tube feeding. and suctioning of the oropharynx at the perineum before the
All had normal nutritional status. 19/29 infants (65%) were delivery of the shoulders.12,13 The neonatal interventions include
enrolled in the EIP. Other 10 required anticipatory development tracheal suctioning at birth in the presence of meconium-stained
counseling. amniotic fluid12 to prevent meconium aspiration.
The growth and development of these infants at 3 years Although the survival rate of MAS has greatly improved over the
were appropriate for age, with the exception of one infant who last three decades,2 the long-term morbidities among survivors
had suffered from birth asphyxia resulting in CP. Two infants have been a major concern. We undertook a study to analyze the
required phenobarbital for control of seizure even at 2 years long-term outcome of infants treated for MAS in our NICU and
of age. compare the same with published reports. However, comparisons of
Of the 13 infants delivered by C-section, only 5 were normal, our findings with the published studies are constrained for several
4 had mild delay, and 4 had severe delay. 16 infants were reasons. First, the treatment modalities differed. Second, the
delivered by NSVD: 6/16 (37.5%) were normal, 8/16 (50%) primary objective of most reports was to study ‘the treatment effect’
had mild delay, and 2/16 (12.5%) had severe delay. rather than the ‘effect of MAS’ on long-term outcome. The third

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S96

issue was that the population included in follow-up studies was the eight studies18–25 reported following outcome with ECMO
sum total of varied diagnoses (MAS, CDH, PPHN, sepsis, and treatment. Four of them18–20,25 showed adverse outcome. Three
pneumonia), making it difficult to assess the independent effect of studies21–23 showed that, although MAS infants had higher
MAS alone on the outcome. Finally, the methods of developmental disabilities compared with the normal infants, they were better
assessment were not uniform and comparable. We attempt here to than infants with diaphragmatic hernia (CDH) and sepsis. Ikle
summarize the uniformity of evaluation and follow-up methods et al.24 showed in their study no significant difference in the
and findings of the published reports of long-term studies. intelligence quotient (IQ) of children at school age, in the ECMO-
We try to provide some perspective on our follow-up study, its treated group, except the children with diagnosis of MAS had lower
importance, and its relation to the current literature. Review of the IQ. These studies indicate that infants with the primary diagnosis
literature during the past 30 years has shown that there were of MAS have poor neurodevelopmental outcome. Within the ECMO-
three major strategies of treatment and outcome reports. treated group, the MAS group had a lower incidence of disability
They included the use of mechanical ventilation in the 1970s, than the CDH and sepsis group, suggesting that the presence of
additional ECMO treatment in the 1980s, and INO treatment sepsis will increase the neurological insult in comparison to MAS.
in the 1990s. The outcome results of the infants treated with The ECMO trials reported long-term outcome results.16,26–29
these different modalities are summarized in Table 3 and These studies included both MAS and PPHN infants. However,
discussed below. separation of the MAS group was not possible. Overall, the studies
showed improved survival following ECMO without concomitant
Studies in which the primary treatment of MAS was increase in morbidities. There were a few differences among the
conventional ventilation alone studies. Vaucher et al.26 on the other hand reported higher
In the 1970s, the treatment of MAS infants who developed acute morbidity in the conventional group compared with the ECMO
respiratory failure consisted of providing early ventilatory support.10 group (25 vs 6% of CP). They also reported that abnormalities
The survival was low. The clinical course10 during the immediate found in neuroimaging predicted later clinical neurodevelopmental
neonatal period and complications of MAS included hypoxic injury abnormalities. The UK Collaborative27 study showed no
of brain, pulmonary atelectasis, air leak syndrome, and other concomitant increase in morbidities in the ECMO group. Rais-
systemic insults.1 During 1978 to 2000, there were 10 other Bahrami et al.28 similarly showed that the neurodevelopmental
studies14–16 published in this category, and they reported long- outcome in the ‘near-miss ECMO’ group (who were sick enough to
term outcome of infants with the primary diagnosis of MAS. In two be on ECMO but were not) had higher handicaps (25 vs 16%) at
of these studies,14,15 MAS infants were treated with conventional follow-up than those treated with ECMO.
ventilation alone. Marshall et al.14 in a retrospective study in 1978 Glass et al.29 is the only report that compared the long-term
identified 13 of the 17 infants (78%) with MAS who survived outcome of MAS infants treated with ECMO with healthy controls.
following conventional ventilator therapy. Follow-up of 11 of the These data showed that MAS infants had significantly higher
survivors showed that 2/11 had developmental delay (18%). Both neurodevelopmental delays compared with controls. Most infants
had seizures. Brett et al.15 in 1981 followed 9 of their 11 survivors undergoing ECMO were in the MAS group. It follows that MAS is
of hyperventilation using conventional mechanical ventilation associated with higher adverse neurodevelopmental sequelae than
therapy. Seven of the survivors had MAS. The range of MDI was 89 the normal population and that these disabilities did not decrease
to 130. They concluded that hyperventilation was not associated with different treatment modalities, as demonstrated in most of the
with adverse outcome. Here, the emphasis was on the efficacy of studies discussed above.
treatment rather than the MAS. Walsh and Suky16 compared the Blackwell et al.30 showed that there were no differences in the
infants treated with conventional ventilation with the ECMO occurrence of seizures between infants with MAS and cord pH<7.20
survivors. 90% of the survivors of the ECMO-treated group had and those with cord pH>7.20, suggesting that there was a
primary diagnosis of MAS. The reported morbidity in the pre-existing injury or that a nonhypoxic mechanism is
ECMO-treated group was twice that of conventional ventilation involved in infants with severe MAS. Casey et al.31 showed that pH
treated group (55 vs 28%). The reported high morbidity among the change from birth to immediate neonatal period predicted
long-time survivors, Macrocephaly developed in 24% of the ECMO neonatal morbidity. Infants whose both cord pH and subsequent
group. They also found 4/17(28%) of the conventionally treated 2 h arterial pH were <7.20 had a higher incidence of seizures.
group had severe neurodevelopmental disabilities, compared with Total of 55% of infants in this group had associated meconium
the 20/38 (26%) of the ECMO-treated group. aspiration, compared with only 36% in infants who had cord
pH>7.20. In our study, only 2/29 (6.9%) had severe
Outcome of infants following ECMO treatment in the 1980s neurodevelopment impairment; neither required ECMO treatment.
Bartlett et al.17 used ECMO in the management of infants with These infants experienced severe intrauterine hypoxia, as noted
MAS who failed to respond to conventional therapy. The rest of the by their cord pH.

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S97

Table 3 Outcome of infants with MAS treated with conventional ventilation

Reference Author(s) Number of cases Outcome Comments


no.

14 Marshall R et al., Study period, 1974; records Outcome at 1 year: four (23.5%) died of Significant psychomotor retardation and
1978 of 1 year respiratory failure cognitive delay among the survivors
Follow-up MAS: 11 infants Two had PFC by cardiac catheterization,
two had significant psychomotor
retardation at 8 and 13 months. CP 11.9%,
both had seizures and one had
microcephaly <5%

15 Brett C et al., Study period March 1977–1979, Outcome at 16 months: seven infants were Although short-term follow-up reported,
1981 number of cases followed 9, MAS: 7, normal neurodevelopment, one had severe preliminary observation assures normal
RDS: 1, sepsis: 1, age at follow-up delay, six had normal neurological exam, development outcome with prolonged
16 months one infant had growth retardation hyperventilation
Hyper ventilated 64.4±18.6

Outcome of ECMO trials


16 Walsh-Sukys MC Study period 1987–1998 Outcome at 20 months Disability among CV was 4 (24%) and in
et al., 1994 Cases followed: 61 ECMO: 2 CP, 2 sesures, disability 20 ECMO 20 (26%)
For 20 months of both CV group, and and 1 deaf. CV: 2 disability, No difference between the two groups
ECMO group 1 CP for blind followed. 24% ECMO group had
macrocephaly at 20 months
18 Towne BH et al., Study period 1973–1980 Outcome at 12 months: 9/11 MAS infants ECMOFlife saving without increasing
1985 Cases followed: 18 were normal; 1 had developmental delay, morbidity
MAS: 14/18 seizures; and 1 had hemiperisis
11 of MAS cases follow-up

19 Andrews AF et al., Study period March Outcome mental function: normal EEG in Majority of survivors of neonates with
1986 1981–September 1983 71%, abnormal in 14, one long-time high risk of respiratory failure can be
Cases followed: 14 with PPHN treatment expected to have near normal growth and
MAS: 5, PPHN: 13, CDH: 1, RDS: 3, Normal outcome in seven (50%), five with development
others: 4 delay and failure to thrive, three with CP,
three had microcephally, and dilated
ventricles
20 Hofkosh D et al., Study period 1990–1993 in 67 cases At 6–30 months outcome by MDI/PDI: Out of entire cohort, normal in 43 (64%),
1991 from 6 months to 10 years. 6–30 normal: 27, abnormal: 4, suspect: 16. abnormal 7 (11%), suspect: 17 (25%).
months: 47 cases; 2.5–4.11 years: 10 2.5–4.11 years, IQ test normal: 7, ECMO did not increase the morbidity;
cases; 5–10 yearsFschool age: abnormal: 2, CP: 2. School age 5–10 years. comparison of control conventional
10 cases IQ normal: 9, abnormal CP: 1 (10%), ventilation group is absent
Diagnosis MAS: 33, CDH: 13, RDS: 8, sensory neural hearing loss: 21%, seizures:
PPHN: 10, others: 3 4, with abnormal outcome
21 Wilden SR et al., Study period January 1990–1992 At 24 months ECMO: GP MDI <85, PDI Overall ECMO group 23% significant
1994 Cases followed: 22 <73, abnormal expressive and receptive impairment
Diagnosis language in 48%. Hearing loss in four
MAS: 10, RDS: 7, PPHN: 3, (15%), visual impairment in one, and
sepsis: 6, CDH: 8 abnormal MRI in nine infants
22 Bernbaum J et al., Study period 1990–1993 Follow-up 6–12 months BPD and feeding difficulties, hypotonicity
1995 Cases followed: 60 Neurologic morbidity in four MAS, and in were greater in CDH, compared to MAS
MAS: 21, CDH: 19, sepsis: 7 five CDH. Hearing loss in two MAS,
hypotonicity in 8% of MAS, and low
cognitive scores in CDH. Total of 68% of
CDH had the worst outcome, with BPD and
feeding difficulty at 12 months

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S98

Table 3 Continued
Reference Author(s) Number of cases Outcome Comments
no.

23 Robertson C et al., Study period 1989–1992 In ECMO: GP disability: 6, hearing loss: 3, The infants with sepsis and respiratory
1995 Outcome at 2 years of age severe cognitive disability: 2, seizures: 1, distress had worse outcome compared to
Cases followed: 40 visual loss: 3, expressive language delay: 7, MAS alone
ECMO: 30, CDH: 8, MAS: 19, PPHN: 3, moderate cognitive delay: 2. MDI were lower
sepsis: 4 in sepsis group, with mean MDI 91.8 (9.5)

24 Ikle L et al., Study period 1988–1991 Outcome of 17 MAS: full-scale IQ varied High percentage of ECMO survivors had
1999 Cases followed: 17 for 5–8 years; from 90 to 115, performance IQ, 81–121, discrepancies between the PIQ and VIQ.
studied IQ at school age verbal IQ, 89–118; 15 children in MAS with PIQ had higher correlation to the number
Diagnosis PPHN with ECMO, MAS: 9, low functioning had low average IQ of hours the infants were on ECMO
sepsis: 3, HMD: 2, others: 3

25 Nield TA et al., Study period 1987–January 1993 Outcome MAS: major neurologic sequelae in No significant difference was found in
2000 Cases followed: 130 for 3.5 years 20%, including MR and CP, functioning functional or neurologic status between
MAS: 63, CDH: 39, PPHN: 4, sepsis: 29, disability in 17%, 24% at risk for disability, the different diagnostic groups
RDS: 8, cardiac: 5 normal function in 59%

26 Vaucher Y et al., Study period 1987–1993 Neuromotor outcome at age CLD had lower ECMO survivors had less CLD than non-
1996 Comparative study, conventional HFV MDI, PDI scores <84, 27% had CP, ECMO survivors
ventilation to ECMO, CV: 53, ECMO: neuroimaging abnormalities. Severe in 11%,
138, MAS: 97, PPHN: 19, RDS: 46, moderate in 9%, mild in 24%
sepsis: 28, studied CLD, and MRI

27 Benett CE et al., Neurodevelopmental outcome at 1. Death (1) Mortality 30/93 (32%) in ECMO and
2001 1 year of age in ECMO 2. Severe disability DQ <50 using Griffiths 54/92 (59%) in non-ECMO
Infants compared with those treated mental developmental scale (2) 10/62 (16%) in ECMO and 5/37
with conventional therapy (99 (13.5%) in non-ECMO had tone changes
survivors) (3) ECMO improves survival with no
increase in morbidity

28 Rais-Bahrami K Study period for 11 years Outcome tested for IQ at 5 years of age. Major handicap was cognitive deficits at 5
et al., 2000 Cases followed: ECMO: 76, near miss FSIQ <0.70 as cognitive disability found in years, and risk for academic school
ECMO: 20 12% of ECMO group, 11% in near miss failures
ECMO GP; MAS: 47, PPHN: 16, ECMO, half of them had MR CP. Sensory
RDS: 13 neural hearing loss academic achievement
Near miss ECMO group; MAS: 11, test, using V IQ, PIQ, FSIQ, academic school
PPHN: 6, RDS: 3 failure, ECMO (38%), near miss ECMO (37%)
29 Glass P et al., Study period, 1984–1988: 103 post-ECMO Parent interview at 5 years, major disability ECMO patient had lower threshold for
1995 compared with 37 healthy controls in 17%, 13 had MR, 4 seizures, 5 severe seizure, even as adults. Learning disability
Diagnosis motor delay, 3 with SNHL and poor academic achievement, high
MAS: 48 FSIQ (96 vs 115), VIQ (96 vs 115), rate of behavioral problems, and high risk
PPHN: 12 PIQ (96 vs 110) of school failures
CDH: 6, number of cases
followed ¼ 42
Outcome of INO trials
35 Rosenberg A et al., Study period 1992–1995 At 1 year, 6 (11.8%) had major handicap, Neurodevelopmental outcomes are similar
1997 Cases followed: 51 with PPHN, 61% MDI, PDI <69, mild disability in 7 (13.7%). to initial INO-treated group
followed up to 1 year, 33% followed to At 2 years, 4 (12.1%) had
2 years, MAS: 23, CDH: 13, PPHN: all neurodevelopmental disability, MDI, PDI
sepsis: 22, RDS: 8, other: 2 <69, 4 infants had MDI, PDI <84, 3 had
sensory neural hearing loss

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S99

Table 3 Continued
Reference Author(s) Number of cases Outcome Comments
no.

36 NINOS, 2000 Study period Outcome at 24 months, cerebral palsy in There was difference between the control
Cases followed: 173 for 18–24 months 30%, with MDI, PDI <70, bilateral group and INO-treated group
blindness in 12%, 6 had CP in INO group,
SNHL: 10 (12%) in INO 34.5% of the INO
group had at least one disability

37 Ellington M et al., Study period September 1992–1997 Outcome INO and CV group: any morbidity: ECMO-treated group had slightly higher
2001 Satisfaction survey of families of 34% (35%) disability CP: 9% (20%), morbidity compared with controls
children treated with INO between 1 hearing, speech: 11% (28%), visual
and 4 years. Cases followed: 60, INO impaired: 12% (3%), DQ: 70 0% (16%)
treated: 42, CV: 40
38 Lipkin PH et al., Study period 1994–1996 INO group: 13% had major disability, No significant difference between the INO
2002 Cases followed: 129, INO treated: 94, hearing loss in 14%. Cognitive delay: 30%, and control. Moderately severe PPHN
control: 35, diagnosis: PPHN only rehospitalization: 22%; INO had 8% CP, and within 24 h after birth had higher rate of
CD in 8%, s.d. in 10%, compared with disability
control had 6% CP, and 6% CD, 6% s.d.

39 Ichiba H et al., Study period 1996–1998 total 15 of Outcome at 3 years of age, one infant with 2/18 (9%) had neurodevelopmental
2003 INO treated follow-up to 3 years CP, one with mid disability, five with disability
MAS: 11, CDH: 1, pneumonia: 1 RDS: reactive airway disease, eight with early
1, hypoplastic lung: 1 response to INO treatment had normal
Only diagnosis PPHN, response to INO development compared with two late
treatment classified as early, late responders
and poor
Abbreviations: BPB, broncho pulmonary dysplasia, CLD, chronic lung disease; CP, cerebral palsy; CV, conventional ventilation; ECMO, extracorporeal membrane oxygenation;
FSIQ, full scale intelligent quotient; HMD, hyaline membrane disease; INO, inhaled nitric oxide; IQ, intelligence quotient; MAS, meconium aspiration syndrome; MDI, mental
development index; PFC, persistant fetal circulation; PIQ, performant intelligent quotient; PPHN, persistent pulmonary hypertension; RDS, respiratory distress syndrome;
VIQ, verbal intelligent quotient.

Outcome of infants treated with INO fundamental to the pathogenesis of neurodevelopment


In 1990, INO was introduced to treat severe PPHN associated handicap among the survivors. The NINOS study36 reported no
with MAS.32,33 Recently, Rosenberg critically reviewed the long- difference in the outcome between INO-treated infants of the larger
term outcomes of MAS and PPHN infants treated with INO group and the control group of infants at 2 years of age. 6/84 INO-
therapy.34 He concluded that, overall, INO was effective in treating treated infants and 7/87 of the controls had CP. 29.9% of the
PPHN in MAS. He observed that all the four studies under review35– controls and 34.5% of the INO-treated infants had at least one
38
reported significant medical and neurodevelopmental sequelae. disability, which was defined as CP. Sensory neural hearing loss
Ichiba et al.39 later reported that the outcome of INO-treated was observed in 12% of infants. Lipkin et al.38 reported higher
infants depended on their response to treatment. Infants who incidence of neurodevelopment disability (CP): 21 and 19% with
responded early had zero sequelae, compared with those who sensory neural hearing loss. This largest group of survivors from
responded late, who had 11% disabilities. Rosenberg et al. the INO-treated group was also found to have 20%
reported35 one-year survival of INO-treated infants with severe neurodevelopmental disability in the follow-up. However, the
morbidities. He also reported the outcome of infants who survived resulting morbidity was much higher than noted in our
the randomized trial of INO treatment with confirmed diagnosis of current group.
PPHN. These infants were followed for 1 to 2 years. At 1 year of age, All the four reports35–38 following the treatment with INO
6/51 (11.8%) were found to have severe neurological disability as claimed improvement in the survival but identified significant
measured by a Bayley II MDI or PDI score of <69, abnormal neurodevelopment morbidity among the survivors. The NICHD
neurological findings, and poor weight gain. He concluded that, collaborative reports36,40 on infants treated with INO who were
regardless of the various postnatal treatment strategies, the followed up at 24 months showed no difference in disabilities
predisposing factors and underlying factors leading to MAS are between the control and the INO-treated groups.

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S100

Summary 3 Amiel-Tison C, Grenier A. Neurological Assessment During First Year of Life. Oxford
Press: New York, 1979.
We report follow-up of 29 MAS infants, almost all of whom were
4 Bayley NM. Bayley Scales of Infant Development. New York: 2nd edn, Psychological
treated only with conventional ventilation. Only one infant was Corporation: San Antonio, 1963.
treated with INO, and two other infants required ECMO. The 5 Rossetti L. Infant Toddler Language Scale. East Moline Illinois Linguisystems. Rosetti
infants were followed up to 3 years of age. The findings of this Louis: Illinois, 1990.
study showed that, in spite of a milder clinical course of illness, 6 Bloom RS, Cropley C. Textbook of Neonatal Resuscitation, 4th edn, Elk. Grove
severe delay was noted in 21% of the infants. Our study shows that Village, IL: American Heart Association, American Academy of Pediatrics, 2000.
infants with MAS, in spite of adequate respiratory support, have the 7 Berkus MD, Langen O, Samueloff A, Xenakis EM, Field NT, Ridgway LE. Meconium
stained amniotic fluid: increased risk for adverse neonatal outcome. Obstet Gynecol
potential to develop later neurodevelopment deficits. In our study, 1994; 84: 115.
we analyzed the importance of cord pH to determine the factors 8 Wiswell TE, Bent RC. Meconium staining and meconium aspiration syndrome. Ped
that predispose to eventual morbidity. We were able to identify cord Clin North Am 1993; 40: 957.
pH<6.9 in two infants who also developed CP. There were no 9 Bascik RD. Meconium aspiration syndrome. Pediatr Clin North Am 1977; 24: 467.
differences in the outcome by mode of delivery. As reviewed above, 10 Vidyasagar D, Yeh T, Harris V, Pildes R. Assisted ventilation in infants with meconium
previous follow-up studies show that although different treatment aspiration syndrome. Pediatrics 1975; 56: 208.
11 Pierce J, Gaudier FL, Sanchez R. Intrapartum amnioinfusion for meconium
modalities, improved the survival, did not improve the outcome
stained fluid: meta analysis of prospective clinical trials. Obstet Gynecol 2000; 95:
among MAS infants. 1051–1056.
12 Ting P, Brady JP. Tracheal suction in meconium aspiration. Am J Obst Gynecol 1975;
122: 767–771.
Conclusion 13 Carson BS, Loosey RW, Bowes Jr WA, Simmons MA. Combined obstetric and pediatric
Although various degrees of neurologic abnormalities are found in approach to prevent meconium aspiration syndrome. Am J Obstet Gynecol 1976; 126:
infants who survived MAS in the neonatal period, establishing the 712–715.
14 Marshall R, Tyrala E, McAlister W, Sheehan M. MAS neonatal and follow-up study. Am
causation of neurological insult has been a difficult problem. The
J Obstet Gynecol 1978; 131(6): 672–676.
above findings suggest poor outcome in MAS, regardless of the 15 Brett C, Dekle M, Leonard CM, Clark C, Snederman S, Roth R et al. Developmental
mode of delivery (NSVD vs C-section) or mode of treatment (ECMO follow-up of hyperventilated neonates: preliminary observations. Pediatrics 1981;
vs INO). Most infants are asymptomatic at the time of discharge 68(4): 588–591.
from the nursery. However, on later follow-up, 10–20% of infants 16 Walsh-Sukys MC, Bauer RE, Cornell DJ, Friedman HG, Stork EK, Hack M. Severe
demonstrated developmental disabilities. The findings above respiratory failure in neonates: mortality and morbidity rates and neurodevelopmental
outcomes. J Pediatr 1994; 125(1): 104–110.
suggest that there are yet other unidentified events that occur
17 Bartlett RH, Roloff DW, Connell RG, Andrews AT, Dillon PW, Zwischenberger JB.
antecedent to delivery of the infant. The presence of meconium at Extracorporeal circulation in neonatal respiratory failure: a prospective randomized
birth may be simply an associated event.41 The retrospective study. Pediatrics 1985; 76: 479–487.
descriptive study has limitations because of the limited number of 18 Towne BH, Lott IT, Hicks DA, Healey T. Long term follow-up of infants
patients available. A multicenter trial involving a large number of and children treated with ECMO: a preliminary report. J Pediatr Surg 1985; 20(4):
cohorts of infants with meconium-stained amniotic fluid as well as 410–414.
MAS is very much needed to better understand the mechanisms of 19 Andrews AF, Nixon CA, Cilley RE, Roloff DW, Bentlett DH. One to three year outcomes
for 14 neonatal survivors of ECMO. Pediatrics 1986; 78(4): 692–698.
development of neurodevelopmental disabilities in these infants.
20 Hofkosh D, Thompson AE, Nozza RJ, Kemp SS, Bowen A, Feldman HM. Ten years of
extracorporeal membrane oxygenation: neurodevelopmental outcome. Pediatrics 1991;
87(4): 549–555.
Acknowledgments 21 Wilden SR, Landry SH, Zwischenberger JB. Prospective, controlled study of
This study was partially supported by Perinatal Program Grant, IDHS #53789004. developmental outcome in survivors of extracorporeal membrane oxygenation: the first
24 months. Pediatrics 1994; 93(3): 404–408.
22 Bernbaum J, Schwartz IP, Gerdes M, D’Agostino JA, Coburn CE, Polin RA. Survivors of
extracorporeal membrane oxygenation at 1 year of age: the relationship of primary
Disclosure diagnosis with health and neurodevelopmental sequelae. Pediatrics 1995; 96
The authors have declared no financial interests. (5 Part 1): 907–913.
23 Robertson C, Finer N, Sauve RS, Whitfield MF, Belgaumkar TK, Synnes AR et al.
Neurodevelopmental outcome after neonatal extracorporeal membrane oxygenation.
CMAJ-JAMC 1995; 152(12): 1981–1988.
References 24 Ikle L, Ikle DN, Moreland SG, Fashaw LM, Waas N, Rosenberg AR. Survivors of
1 Cleary GM, Wiswell TE. Meconium stained amniotic fluid and the MAS: an update. Ped neonatal extracorporeal membrane oxygenation at school age: unusual findings on
Clin North Am 1998; 45(3): 511–525. intelligence testing. Dev Med Child Neurol 1999; 41: 307–310.
2 Sriram W, Khoshnood B, Singh JK, Msieh HL, Lee KS. Racial disparity in meconium 25 Nield TA, Langenbacher D, Poulsen MK, Platzker AC. Neurodevelopmental outcome at
stained amniotic fluid and MAS in the US, 1989–2000. Obstet Gynecol 2003; 102(6): 3.5 years of age in children treated with extracorporeal life support: relationship to
1262–1267. primary diagnosis. J Pediatr 2000; 136(3): 338–344.

Journal of Perinatology
Neurodevelopmental outcome of infants with MAS
N Beligere and R Rao
S101

26 Vaucher Y, Dudell GG, Bejar R, Gist K. Predictors of early childhood outcome 34 Rosenberg A. Outcome in term infants treated with inhaled nitric oxide. J Pediatr 2002;
in candidates for extracorporeal membrane oxygenation. J Pediatr 1996; 128(1): 140(3): 284–287.
109–117. 35 Rosenberg A, Kennaugh J, Moreland SG. Longitudinal follow-up of a cohort of
27 Bennett CE, Johnson A, Field DJ, Elbourne D. UK Collaboratie ECMO Trial Group. UK newborn infants treated with inhaled nitric oxide for persistent pulmonary
collaborative randomized trial of neonatal ECMO, follow-up at 4 years of age. Lancet hypertension. J Pediatr 1997; 131(1): 70–75.
2001; 357(9262): 1094–1096. 36 The Neonatal Inhaled Nitric Oxide Study Group Journal of Pediatrics. Neonatal inhale.
28 Rais-Bahrami K, Wayner AF, Coffman C, Glass P, Short BL. Neurodevelopmental Group inhaled nitric oxide. Neurodevelopmental follow-up of NINOS. Pediatrics 2000;
outcome in ECMO v/s Near ECMO patients at 5 years of age. Clin Pediatr 2000; 39(3): 136(5): 611–617.
145–152. 37 Ellington M, O’Reilly D, Allred EN, McCormick MC, Wessel DL, Kourembanas S. Child
29 Glass P, Wagner AE, Papero PH, Rajasingham SR, Civitello LA, Kjaer MS et al. health status, neurodevelopmental outcome, and parental satisfaction in a randomized,
Neurodevelopmental status at age five years of neonates treated with extracorporeal controlled trial of nitric oxide for persistent pulmonary hypertension of the newborn.
membrane oxygenation. J Pediatr 1995; 127(3): 447–457. Am Acad Pediatr 2001; 107(6): 1351–1356.
30 Blackwell SC, Modenhauer J, Hassan SS, Redman ME, Refuerzo JS, 38 Lipkin PH, Davidson D, Spivak L, Straube R, Rhines J, Chang CT. Neurodevelopmental
Berry SM et al. Meconium aspiration syndrome in term neonates with normal and medical outcomes of persistent pulmonary hypertension in term newborns treated
acid base status at delivery: is it different? Am J Obstet Gynecol 2001; 184(7): with nitric oxide. J Pediatr 2002; 140(3): 306–310.
1422–1425. 39 Ichiba H, Matsunami S, Itoh F, Ueda T, Ohsasa Y, Yamano T. Three-year follow-
31 Casey BM, Goldaber KG, McIntire DD, Leveno KJ. Outcomes among term infants when up of term and near term infants with inhaled nitric oxide. Pediatr Int 2003; 45(3):
two-hour postnatal pH is compared with pH at delivery. Am J Obstet Gynecol 2001; 290–293.
184(3): 447–450. 40 Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in full-term
32 Roberts JD, Polaner DM, Lang P, Zapor WM. Inhaled nitric oxide in PPHN of the and nearly term infants with hypoxic respiratory failure. N Engl J Med 1997; 336:
newborn. Lancet 1992; 340: 818–819. 597–604.
33 Kinsella JP, Neish SR, Shaffer F, Abman SH. Low dose inhalational inhaled INO in 41 Ghidini A, Spong CY. Severe meconium aspiration syndrome is not caused by
PPHN of the newborn. Lancet 1992; 340: 819–820. aspiration of meconium. Am J Obstet Gynecol 2001; 185(4): 931–938.

Journal of Perinatology
Journal of Perinatology (2008) 28, S102–S107
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Studies of meconium-induced lung injury: inflammatory
cytokine expression and apoptosis
D Vidyasagar and A Zagariya
Division of Neonatology, Department of Pediatrics, The University of Illinois at Chicago, Chicago, IL, USA

inflammatory reaction may be central to the initial progression of


To review current literature related to cellular mechanisms of meconium- MAS and associated severe pulmonary hypertension.3–5
induced lung injury (MILI). Review of published experimental in vitro and Investigators using animal models have shown the presence of
in vivo MAS studies using human and animal lung cells. We found that inflammatory-induced cell death through apoptotic process in
meconium induces expression of cytokines and angiotensin II (ANG II)- meconium-instilled lungs.6,7 There is increased interest to better
induced apoptotic process in the lung cells. We postulate that inflammatory understand complex mechanisms of inducing meconium-induced
cytokines induce ANG II expression, which causes apoptotic cell death after lung cell apoptosis. The purpose of this paper is to review the
binding to its AT1 receptors. We also demonstrated expression of serpins literature and better understands the mechanisms of
associated with meconium instillation into the lungs. Serpins are proteins meconium-induced lung injury (MILI).
that inhibit cellular proteases and elastases. Expression of serpins may be an The evidence for meconium-induced inflammation and
attempt to recover lung from these injurious effects. In summary our apoptosis comes from different investigators. These studies can be
studies show that whereas meconium induces inflammatory cytokines and classified into two categories: one from human studies and the
subsequent cell apoptosis, the lung cells also try to protect themselves by other from experimental animal models. Table 1 shows selected
inducing serpins. The balance of these interactions will determine the papers in this regard. In general, these findings showed that
residual damage. We believe these new findings are very important in meconium in both human and animal models produces
understanding of MILI. inflammatory reaction, which is followed by cascade of events
Journal of Perinatology (2008) 28, S102–S107; doi:10.1038/jp.2008.153 leading to lung epithelial cell apoptosis. Findings of these studies
are summarized below.
Introduction
Human studies
Meconium aspiration syndrome (MAS) is one of the major clinical
problems encountered in the neonatal period. It is an There are very few human studies (Table 1). De Beaufort et al.8
inflammatory newborn lung disease that frequently leads to severe were the earliest investigators who studied in vitro response of
respiratory failure with a potential fatal outcome in term and human neutrophils to MAS. They showed that there was
post-term infants.1,2 Aspiration of meconium is known to cause overexpression of interleukin 8 (IL-8) in neutrophils exposed to
airway obstruction, damage of airways and surfactant inactivation. meconium. They also showed anti-IL-8 antibodies inhibited
Meconium also causes chemical pneumonitis.1–3 In the newborn, neutrophils migration. These studies suggested that MAS in infants
the major clinical manifestations include problems of oxygenation may induce neutrophils migration and overexpression of IL-8 lead
and respiratory failure associated with increased pulmonary to inflammation.
vascular reactivity. It has been shown that persistent pulmonary Uotila et al.9 studied meconium exposure to monocytes in
hypotension is a hallmark of severe MAS. Number of investigators culture and showed increased expression of syclogexynase 2
has extensively studied these aspects of MAS. expression. Jones et al.4 demonstrated increased production of
In addition to the above-described derangements meconium inflammatory cytokines in preterm infants with MAS. Castelheim10
also induces intense inflammatory reaction in the airways and showed compliment activation in human monocytes culture of
alveoli. However not much has been explored in this field. There is MAS model.
emerging evidence from different animal models that intense
Animal studies
Correspondence: Dr D Vidyasagar, Division of Neonatology, Department of Pediatrics, The
University of Illinois at Chicago, 840 South Wood Street, Chicago, IL 60517, USA. Investigators have studied meconium-induced inflammation using
E-mail: dsagar@uic.edu different animal models (Table 1). Davey et al.,11 in piglets, showed
Inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya
S103

Table 1 Human and animals models of MAS

Human models of MAS

Srinivasan and Vidyasagar2 Newborn infants Increase in bile acids, lipids, polysaccharides, in vivo and in vitro
Jones et al.4 Preterm and term newborns Production of TNFa, IL1b, and IL8 is regulated by IL-10; IL-10 not detected in preterm infants with
or without MAS
de Beaufort et al.8 Newborn infants Overexpression of IL-8 in MAS in vitro; increased migration of neotrophils; anti-IL-8 antibodies
inhibit neutrophil migration
Uotila and Kaapa9 Human monocytes In culture increase of cycloxygenase-2 expression in vitro
Castellheim et al.10 Human monocytes in culture Complement activation in MAS

Animal models of MAS


Tyler et al.3 Animal models Surfactant inactivation, chemical pneumonitis, airway obstruction, direct toxic damage of animal
lung tissues
Zagariya et al.6 Rabbit pups Intense inflammatory reactions involved in meconium-induced injury in vivo; expression of
endothelin 1 in MAS
Davey et al.11 Piglets Acute decrease in gas exchange and dynamic lung compliance; increase of total lung protein;
inhibition of surfactant
Holopainen et al.12 Piglets Inflammation, airway epithelium targeted, necrotic changes, phospholipase 2 activity increased.
Mollnes et al.13 Animal models Complement activation in MAS
Khan et al.14 Mice Increase of IL-13 expression in MAS in vivo; lymphocytic and eosinophilic inflammation
Zagariya et al.15 Rabbit pups Phospholipase A2 activates apoptosis, influx of T-lymphocytes and macrophages in vivo; increase of
TNFa, IL-1b, IL-6 and IL-8 in MAS in vivo
Zagariya et al.16 Rabbit pups Cell death by apoptosis, influx of neutrophils and macrophages in vivo
Tessler et al17 Newborn infants Reactive oxygen species in vitro
Abbreviations: IL, interleukin; MAS, meconium aspiration syndrome; TNF, tumor-necrosis factor.

acute decrease in gas exchange and dynamic lung compliance, 0.9% NaCl (pH 7.2) to a final concentration of 10% (weight/
increase of total lung protein and inhibition of surfactant. volume) and was spun down at 1000 g for 20 min (at 4 1C) to
Holopainen12 made extensive observations in piglets exposed to separate supernatant and meconium debris. Supernatant was
meconium. They demonstrated severe inflammation and airway filtered using 0.45 mm filter paper (Millipore Co., Bedford, MA,
epithelial damage, necrotic changes associated with increased USA). The cell-free filtered solution instilled into the lungs.
phospholipase 2 activities. Mollnes also showed an increased Following instillation, rabbits were allowed to breathe
compliment activation following experimental MAS in piglets.13 spontaneously in room air with no additional oxygen. No
Khan14 showed that MAS increases IL-13 expression along with respiratory support was provided to pups following instillation of
lymphocytic and iosinophilic inflammation. Tyler3 in animal model meconium. Animals were sacrificed at 0, 2, 4, 8 and 24 h after
showed that MAS leads to surfactant inactivation and pneumonitis. meconium instillation. The findings of other studies including ours
In our own laboratory, using rabbit pups,6,15,16 we have shown are described below.
that lung exposure to meconium leads to intense inflammatory
reaction with increased expression of lymphocytes, macrophages
and neutrophils. Experimental studies showed that inflammatory
response and apoptotic epithelial cell death is important feature of Inflammatory responses to meconium
meconium-induced newborn lung injury. Our studies differ from The first response to meconium injury is the release of active
previously published studies because of the methodology used. In inflammatory cells, the most important of which are neutrophils
our studies we used rabbit pups with no previous hypoxia. Another and macrophages.8,9,16 Especially, neutrophils are dramatically
major distinction from other studies is that we did not use whole increased in meconium-instilled lungs compared to saline-instilled
meconium, because whole meconium contains many components lungs. Macrophages are also increased, but not as dramatically as
that can cause lung injury. Instead, we used debris-free meconium neutrophils. Tessler recently suggested that meconium also induced
to avoid mechanical obstruction of airways. One gram of fresh free oxygen radicals.17 Now we describe the pathways of induction
newborn meconium (1 g) was homogenized on ice with 9 ml of of inflammatory response following cell influx.

Journal of Perinatology
Inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya
S104

Neutrophil influx lungs,4,16 the resulting cytokine imbalance may regulate the
Aspirated meconium produces a vigorous but transient leukocytic susceptibility of the newborn lung to meconium-induced injury.
inflammatory reaction in the newborn lungs, but the mechanisms
of the neutrophil influx are still unclear.10 Meconium itself may Complement activation
stimulate neutrophil chemotaxis and it supposedly induces the Various investigators10,13,24 using animal and human cells showed
lung inflammatory cells to produce proinflammatory chemotactic that human meconium is a potent activator of complement, an
cytokines.4,8,10,18 Ex vivo studies using a piglet model show that important mediator of inflammatory tissue damage. Meconium
moderate and high concentrations of aspirated meconium rapidly activated the alternate complement pathway by increase of
activate circulating neutrophils.19 Experimental study in piglet alternative convertase C3bBbp.10 Meconium can also induce
model suggests that meconium-induced inflammation is localized systemic complement activation, paralleling the increase in lung
in the meconium-contaminated areas of the lungs with no dysfunctions.
generalized inflammatory injury.20 Correspondingly, local
accumulation of neutrophils and macrophages and production of Phospholipase activation
IL-1b and tumor-necrosis factor-a (TNFa) cytokines have been Local release of phospholipid-degrading enzyme, phospholipase A2,
observed.2,20 Current experimental data suggests that may further play an exacerbating role in the pathogenesis of acute
intrapulmonary accumulation of activated neutrophils, similar to inflammatory lung injury after meconium aspiration in a human
various forms of acute injury in adult lungs21 could aggravate the model. As showed by Kaapa,12 experimental studies in animal
hypoxic respiratory failure. Myeloperoxidase activity is usually used models indicate that this proinflammatory activity is also contained
as an important indicator of neutrophil accumulation.22 The in human meconium itself. The existing data on human and
chemotaxis is related to the presence of powerful chemotactic role animal models thus indicate that constituents of aspirated
of IL-8. meconium, including cytokines, phospholipases, bile acids, lipids
In a study by Ruiz et al.23 addition of polyclonal and polysaccharides, may trigger a local inflammatory reaction
immunoglobulin G (IgG) activated the mononuclear cells to supposedly through stimulation of lung inflammatory cells to
express IL-8 cytokine production, which could promote produce proinflammatory cytokines and prostaglandins.8,25
intrapulmonary accumulation and infiltration of neutrophils. Moreover, recent data demonstrate that activation of lung tissue
Other studies have shown that intravenous IgG may increase of phospholipase A2, in meconium injury, may mediate cellular
neutrophil migration to the sites of inflammation via unknown apoptosis, as a noninflammatory mechanism of programmed cell
mechanisms.21 Sterile meconium increased migration of death, in porcine meconium aspiration.26
neutrophils obtained from neonates in comparison with random
migration in vitro studies. These studies show that meconium
induces chemotaxis of inflammatory cells especially neutrophils.8 Meconium-induced apoptosis
These above studies showed the neutrophil response to meconium. Apoptosis of cells exposed to meconium was one of the interesting
findings of our experimental studies. Apoptosis is a programmed
Cytokine release and their effect cell death, which does not result in inflammatory tissue injury, but
In experimental models of MAS, an intense pulmonary rather is a cell clearance mechanism promoting resolution of
inflammatory reaction with influx of polymorphonuclear inflammation.27 Normal rate of apoptosis is beneficial to the host
leukocytes, T-lymphocytes, monocytes and macrophages is organ. However, increased apoptosis may clearly cause harm for
demonstrated within a few hours.10,12,16 It is further apparent that the tissue. Recent literature including data from our laboratory
intrapulmonary meconium stimulates the lung inflammatory cells demonstrate that apoptosis may play an important role in acute
to express inflammatory cytokines, such as TNF-a, IL-1b, IL-6 and lung injury of newborn.12,16 As was demonstrated by several
IL-8, and may thereby propagate development of parenchyma cell techniques, meconium-induced apoptosis is localized preferably in
injury in the exposed animal and human lungs.8,10,16 A novel lung and its airway epithelial cells.12,16 These data demonstrate
signaling pathway involved in MAS is the role of IL-13. This that meconium-induced lung cell apoptosis leads to damage and
molecule has been shown to be markedly elevated in a murine detachment of lung airway or epithelial cells. It is a very important
model of MAS and could be a mediator of persistent airway process as airway epithelium plays a very important role in
dysfunction and remodeling in survivors of MAS.14 In fact, maintenance of cell viability and normal lung functions. In spite a
dramatic initiation of the pulmonary inflammatory response to several existing theories, exact mechanism of meconium-induced
meconium aspiration is highlighted by immediate appearance of lung cell apoptosis is presently unknown.
the lung pro-inflammatory cytokine expression in rabbit models, Usually apoptosis is seen immediately after instillation of
with a maximum at 8 to 14 h after the meconium insult.16 As low meconium into the lungs. Apoptosis occurs under the influence of
levels of anti-inflammatory IL-10 is simultaneously found in these different substances present in meconium. Apoptotic cells are

Journal of Perinatology
Inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya
S105

140 smaller amount (1 p.p.m.) of NO inhalation was similar but did


120 not reach statistical significance.30 Apoptotic cell death was induced
by exogenous NO in rat type II pneumocytes in vitro32 but not in
% of Caspase Activity

100
cultured human cells of pulmonary epithelial orendothelial
80 origin.33 Although the inhibitory effect of NO is still unclear,
60 interaction of NO with oxygen-reactive species may eliminate the
adverse affects of these toxic radicals or, alternatively, upregulate
40
protective genes that attenuate apoptosis.32 Furthermore, increased
20 cycloxygenase-2 expression, shown to be stimulated in monocytes
0 by human meconium in vitro,9 may modulate NO-mediated
0 2 4 8 24 apoptosis in macrophages.32
Figure 1 Expression of caspase 3 at 0, 2, 4, 8 and 24 h after meconium
instillation into 2-week-old rabbit lungs.
Role of ANG II and its receptors in apoptosis
smaller in size; they appear shrunk, their cytoplasm and nuclei are Pulmonary angiotensin II (ANG II) has been recently proposed to
condensed. Examination of the lung lavage cells demonstrated that contribute to receptor-mediated lung epithelial apoptosis in vivo
apoptosis usually is accompanied with inflammatory reactions, and in vitro, but in neonatal lung injuries in vivo this mechanism
extensive cell death. The apoptotic cells are identified by measuring is still unclear.34 We demonstrated using a rabbit model of MAS
caspase 3 expression.28 Maximal expression of caspase 3 was also that apoptosis occurs via cytokine-induced angiotensinogen gene
seen at 8 h after meconium instillation (Figure 1). expression, conversion of ANG I to ANG II, and binding of ANG II
ISEL labeling is another method of identifying the apoptotic to its AT1 receptors.34–36 This conversion requires the presence of
cells.16 Using ISEL labeling, we found a fragmented DNA in ANG-converting enzyme, ACE.34,36 In animal models captopril may
numerous airways and alveolar epithelial nuclei.16 ISEL-positive also decrease the apoptosis process in alveolar epithelium
cells were seen specifically in meconium-instilled lungs but not in induced by FAS, bleomycin, plant alkaloid monocrotaline, or
saline-instilled lungs. ISEL-positive sells surround the bronchioles g-irradiation.34–37 Consequently, it was hypothesized that
and gradually expanded to the entire lung field. meconium-induced apoptosis could be prevented by suppression of
Apoptosis destroys many important cells in the lung airway or ANG I conversion to ANG II by captopril pretreatment or by
alveolar epithelium. In worst cases, such destruction may reach up blocking of its AT1 receptors. Recent data from our laboratory
to 50% of airway epithelial cells. The mechanism of indicate that captopril and inhibition of AT receptor action may
meconium-induced epithelial cell apoptosis and their detachment diminish pulmonary epithelial apoptosis in meconium lungs.28,37
presently remains unknown. Also unknown are mediating Additional data from our laboratory show that cytokine-treated
mechanisms of apoptotic cell death, which we will describe below. rabbit lung and human lung airway epithelial cell line A549
demonstrate a similar rate of lung apoptosis, compared to
meconium-instilled lungs.35,38 From the above discussion it may be
Oxygen radicals in apoptosis postulated that intrapulmonary meconium induces influx of
Increased release of reactive oxygen species from the animal inflammatory cells in the lungs. These cells express inflammatory
pulmonary inflammatory cells may be associated with apoptotic cytokines, involved in expression of ANG I, which is immediately
cell death in the lung.29 Neutrophil influx following meconium processed to ANG II by ACE. Lung cell damage and apoptosis occur
exposure may also activate alveolar macrophages to produce when ANG II binds to its AT1 but not AT2 receptors (Figure 1). It
oxygen radicals, which may, at least in part, explain the is, however, not clear how the above-discussed mechanisms are
proapoptotic effect of meconium in animal and human lungs.17,19 initiated.

Nitric oxide and apoptosis Protective role of serpin in apoptosis


Although the bulk of evidence indicates that nitric oxide (NO) may During our studies to explain the mechanisms of apoptosis, we
induce apoptosis in different cell lines, including macrophages, it isolated a protein from tracheal aspirates. We found that this
may have a dual effect. It is interesting to note that inhaled smaller protein to be a novel serine proteinase inhibitor, with an apparent
quantities of NO may also inhibit apoptosis.30,31,32 Inhalation of molecular mass of 50 kDa, which was identified to be e
NO (10 p.p.m.), in addition to having a beneficial effect on a1-antitripsin.39 Serpins are known inhibitors of proteases and
hemodynamics and oxygenation in newborn lungs, also apoptosis. We hypothesize that serpin may attenuate pulmonary
significantly inhibited cell death by apoptosis. The effect of a inflammation and improve surfactant function after meconium

Journal of Perinatology
Inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya
S106

MECONIUM-INDUCED LUNG INJURY Conceptual model of MILI mechanisms

Meconium Meconium

LUNG Lung
Epithelial Cells
Alveolar & Airway
Responce Inflammatory Responce
Neutrophils and
Neutrophil Endothelial Macrophages
Macrophages
Influx Cells

Cytokines Endothelin-l Cytokines


+ –
Cytokines APOPTOSIS Serpin
Lung Injury
Inflammation Apoptosis
Figure 3 Cytokines and serpin in meconium-induced apoptosis.
Histological
Changes

Figure 2 Meconium-induced lung injury.


Summary
In summary, meconium aspiration induces a rapid and intensive
aspiration. a1-antitripsin is a member of the proteinase inhibitor inflammatory reaction and lung injury within the contaminated
(serpin) superfamily and inhibitor of neutrophil elastase. areas of the newborn lungs and produces simultaneously lung cell
a1-Antitripsin is synthesized by epithelial cells, macrophages, death by apoptosis. In addition, based on our work, we believe that
monocytes and neutrophils. Deficiency in a1-antitripsin leads to meconium-induced lung cell apoptosis is also induced by
exposure of lungs to uncontrolled proteolytic attack from accumulation of ANG II and binding to its AT1 receptors. TNFa or
neutrophil elastase or other damaging factors culminating in lung IL-8 cytokines may be involved in processing ANG II and thus is
destruction and cell apoptosis. We hypothesize that accumulation also involved in increasing meconium-induced cell apoptosis.
of a1-antitripsin in the lungs serves as a predisposed protection We also believe that degree of apoptosis is dependent on the
against MILI. production of proteases and serpins in the cells. Serpins inactivate
meconium-induced proteases thus prevent lung damage. In
summary, we propose that meconium induces cytokine expression
Unified concept of MILI
and lung cell apoptosis. At the same time expression of serpins in
Taking into consideration of all the above findings we propose a response to meconium may negate the effect of proteases induced
unified concept of the mechanism of MILI. The construct of the by meconium (Figure 3).
hypothesis is shown in Figure 2. It is proposed that meconium The significance and contribution of these lung injury processes
aspiration by the newborn besides causing airway obstruction to the clinical manifestation of MAS, however, needs to be further
causes local inflammation. The inflammatory reaction leads to investigated. Better understanding of meconium-induced
cellular influx of neutrophils and macrophages. Both release inflammatory injury and apoptosis may lead to a development of
inflammatory cytokines, which in turn cause inflammation and set new therapeutic interventions of MAS.
the process of apoptosis. The end result depends on the extent of
inflammation and degree of apoptosis. Moderate apoptosis may
leave no permanent damage. Severe apoptosis initiates fibrotic
reaction. These results may become perceptible on histological Acknowledgments
examination of the tissues. We acknowledge the grant support of Thrasher Foundation (ID no. 02823-6) for
The sequence of events in the epithelial cells is different. these studies. We also thank staff of Michael Reese Animal Laboratory,
Epithelial cells express ANG II and their AT1 and AT2 receptors. Dr Shankar Rao Navale, Dr Rama Bhat, Dr Gopal Chari, Dr Bruce Uhal, Dr Pekka
Both participate in the process of cell apoptotic cell death. Kaapa, Dr Charles Rosenfeld and summer students for everyday assistance
It is also to be noted that meconium can induce serpin in experimental work, collecting data and other support of this work.
expression, which plays a protective role against injury and
apoptosis which outcomes depends from the ration of proteases and
serpins. If this ration is large a large damage is observed; if small, Disclosure
it is minimal. The authors have declared no financial interests.

Journal of Perinatology
Inflammatory cytokine expression and apoptosis
D Vidyasagar and A Zagariya
S107

References 21 Fijishima S, Aikawa N. Neutrophil-mediated tissue injury and its modulation. Intensive
Care Med 1995; 21: 277–285.
1 Vidyasagar D, Yeh TF, Harris MD, Pildes RS. Assisted ventilation in infants with
22 Holopainen R, Soukka H, Halkola L, Kaapa P. Intravenous immunoglobulin G
meconium aspiration syndrome. Pediatrics 1975; 56(2): 208–213.
attenuates pulmonary hypertension but induces local neutrophil influx in meconium
2 Shrinivasan HB, Vidyasagar D. Meconium aspiration syndrome: current concepts and
aspiration in piglets. Biol Neonate 2005; 87: 221–228.
management. Compr Ther 1999; 25(2): 82–89.
23 Ruiz de Souza V, Carreno MP, Kaveri SV, Ledur A, Sadeghi H, Cavaillon JM et al.
3 Tyler DC, Murphy J, Cheney FW. Mechanical and chemical damage to lung tissue
Selective induction of interleukin-1 receptor antagonist and interleukin-8 in human
caused by meconium aspiration. Pediatrics 1978; 62: 454–459.
monocytes by normal polyspecific IgG (intravenous immunoglobulin). Eur J
4 Jones CA, Cayabyab RG, Kwong KY, Stotts C, Wong B, Hamdan H et al. Undetectable
Immunol 1995; 25: 1267–1273.
interleukin (IL)-10 and persistent IL-8 expression early in hyaline membrane disease:
24 Baggiolini M, Loetcher P. Chemokines in inflammation and immunity. Immunol
a possible development basis for the predisposition to chronic lung inflammation in
preterm infants. Pediatr Res 1996; 39: 966–975. Today 2000; 21: 418–420.
5 Holopainen R, Soukka H, Kaapa P. Meconium aspiration induced a 25 Lopez A, Bildfell R. Pulmonary inflammation associated with aspirated meconium and
concentration-dependent pulmonary hypertension in newborn piglets. epithelial cells in calves. Vet Pathol 1992; 29: 104–111.
Pediatr Pulmonol 1998; 25: 107–113. 26 Kaapa P. Meconium aspiration syndrome: a role for phospholipase A2 in the
6 Zagariya A, Doherty J, Bhat R, Navale S, McMillan M, Uhal B et al. Elevated pathogenesis? Acta Paediatr 2001; 90: 365–367.
immunoreactive level of ET-1 levels in newborn rabbit lungs after meconium 27 Bardales RH, Xie SS, Schaefer RF, Hsu SM. Apoptosis is a major pathway responsible for
aspiration. Pediatr Crit Care Med 2002; 3(3): 297–302. the resolution of type II pneumocytes in acute lung injury. Am J Pathol 1996; 149:
7 Soukka H, Viinikka L, Kaapa P. Involvement of thromboxane A2 prostacyclyn in the 845–852.
early pulmonary hypertension after porcine meconium aspiration. Pediatr Res 1998; 28 Zagariya A, Bhat R, Chari G, Uhal B, Navale S, Vidyasagar D. Apoptosis
44(6): 838–842. of airway epithelial cells in response to meconium. Life Sci 2005; 76(16):
8 de Beaufort AJ, Pelikan DM, Elferink JG, Berger HM. Effect of interleukin 8 in 1849–1858.
meconium on in-vitro neutrophil chemotaxis. Lancet 1998; 352(9122): 102–105. 29 Kazzas JA, Xu J, Palaia TA, Mantel L, Fein AM, Horowitz S. Cellular
9 Uotila PJ, Kaapa PO. Cyclooxygenase-2 expression in human monocytes stimulated by oxygenotoxicity. Oxidant injury without apoptosis. J Biol Chem 1996; 271:
meconium. Lancet 1998; 351: 978. 15182–15186.
10 Castellheim A, Lindenskov PH, Pharo A, Fung M, Saugstad OD, Mollnes TE. Meconium 30 Gaboury J, Woodman RC, Granger DN, Reinhardt P, Kubes P. Nitric oxide prevents
is a potent activator of complement in human serum and in piglets. Pediatr Res 2004; leukocyte adherence: roleof superoxyde. Am J Physiol 1993; 265(3 pt 2): H862–H867.
55(2): 310–318. 31 Brune B, von Knethen A, Sanday KB. Nitric oxide and its role in apoptosis. Eur J
11 Davey AM, Becker JD, Davies JM. Meconium aspiration syndrome: physiological and Pharmacol 1998; 351: 261–272.
inflammatory changes in the newborn piglet model. Pediatr Pulmonol 1993; 16: 32 Janssen YMW, Matalon S, Mossman BT. Differential induction of c-fos, c-jun, and
101–108. apoptosis in lung epithelial cells exposed to ROS or RNS. Am J Physiol 1997;
12 Holopainen R, Aho H, Laine J, Peuravuori H, Soukka H, Kaapa P. Human meconium 273(4 Pt 1): L789–L796.
has high phospholipase A2 activity and induced cellular injury and apoptosis in piglets 33 Narula P, Xu J, Kazzaz A, Robbins CG, Davis JM, Horowitz S. Synergistic cytotoxicity
lungs. Pediatr Res 1999; 46: 626–632. from nitric oxide and hyperoxia in cultured lung cells. Am J Physiol 1998; 274:
13 Mollnes TE, Song WC, Lambris JD. Complement in inflammatory tissue damage and L411–L416.
disease. Trends Immunol 2002; 23(2): 61–64. 34 Wang R, Zagariya A, Ang E, Ibarra-Sunga O, Uhal B. FAS-induced apoptosis of alveolar
14 Khan AM, Elidemir O, Epstein CE, Lally KP, Xue H, Blackburn M et al. Meconium epithelial cells require angiotensinogen II generation and receptor interaction. Am J
aspiration produces airway hyperresponsiveness and eosinophilic inflammation in a Physiol Lung Cell Mol Physiol 1999; 277: L1245–L1250.
murine model. Am J Physiol Lung Cell Mol Physiol 2002; 283(4): L785–L790. 35 Wang R, Alam G, Zagariya A, Gidea C, Pinillos H, Lalude O et al. Apoptosis of lung
15 Zagariya A, Bhat R, Navale S, Vidyasagar D. Cytokine expression in meconium-induced epithelial cells in response to TNFa requires Angiotensin II generation de novo. J Cell
lungs. Indian J Pediatr 2004; 71(3): 195–201. Physiol 2000; 185: 253–259.
16 Zagariya A, Bhat R, Uhal B, Navale S, Freidine M, Vidyasagar D. Cell death and lung 36 Wang R, Zagariya A, Ibarra-Sunga O, Gidea C, Ang E, Deshmukh S et al.
cell histology in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; Angiotensin II induced apoptosis in human and rat alveolar epithelial cells. Am J
159(11): 819–826. Physiol 1999; 276: L885–L889.
17 Tessler R, Pan J, Fiori HH, Belik J. Human meconium has a pulmonary vascular and 37 Zagariya A, Uhal B, Navale S, Vidyasagar D. Inhibition of Meconium Induced Lung
airway smooth muscle relaxant effect. Pediatr. Res 2008; 64(1): 24–28. Cell Apoptosis by Captopril. Pediatric Academic Societies and American Academy of
18 Kojima T, Hattori K, Fujuwara T, Takedatsu-Sasai M, Kobayashi Y. Meconium-induced lung Pediatrics Joint Meeting, Boston, MA 2000, Abstract.
injury mediated by activation of alveolar macrophages. Life Sci 1994; 54: 1559–1562. 38 Zagariya A, Bhat R, Uhal B, Navale S, Vidyasagar D. Captopril Inhibits Meconium
19 Soukka HR, Ahotupa M, Ruutu M, Kaapa PO. Meconium stimulates neutrophil Induced Cytokines Expression and Lung Cell Apoptosis, Pediatric Academic Societies
oxidative burst. Am J Perinatol 2002; 19(5): 279–284. Meeting, Philadelphia, PA 2001, Abstract.
20 Korhonen K, Soukka H, Halkola L, Peuravuori H, Aho H, Pulkki K et al. Meconium 39 Zagariya AM, Bhat R, Zhabotynsky E, Chari G, Navale S, Xu Q et al. Characterization of
induces only localized inflammatory lung injury in piglets. Pediatr Res 2003; 54(2): serine/cysteine protease inhibitor alpha1-antitripsin from meconium-instilled rabbit
192–197. lungs. J Cell Biochem 2005; 96(1): 137–144.

Journal of Perinatology
Journal of Perinatology (2008), S108–S112
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Angiotensin II in apoptotic lung injury: potential role in
meconium aspiration syndrome
BD Uhal and A Abdul-Hafez
Department of Physiology, Michigan State University, East Lansing, MI, USA

apoptogenic mitochondrial proteins such as cytochrome c, or an


Meconium aspiration injures a number of cell types in the lung, most extrinsic pathway that follows activation of a death receptor. The
notably airway and alveolar epithelial lining cells. Recent data show that at receptor is usually activated by its extracellular ligand, such as the
least some of the cell death induced by meconium occurs by apoptosis, and binding of Fas ligand (FasL) to Fas (APO-1 or CD95) or tumor
therefore has the potential for pharmacologic inhibition through the use of necrosis factor-a (TNF-a) binding to TNF-a receptor. On activation,
apoptosis blockers or other strategies. Related work in adult animal initiator caspases (caspase-2, -8, -9, and -10) propagate death signals
models of lung injury has shown that apoptosis of lung epithelial cells by activating downstream effector caspases (caspase-3, -6, and -7) in
induces a local (that is, entirely lung tissue specific) renin-angiotensin a cascade-like manner. Activated effector caspases mediate the release
system (RASL). Furthermore, this inducible RASL is required for the apoptotic of caspase-activated DNase (CAD) from the complex with its
response and affects other adjacent cell types through the release of angiotensin endogenous inhibitor (ICAD) through a proteolytic cleavage. The
II and related peptides. This manuscript reviews the published data supporting activated CAD then cleaves genomic DNA between nucleosomes to
this viewpoint as well as more recent works that suggest the involvement of a generate DNA fragments, which are multiples of 180 to 200 bp in
RASL in the perinatal lung damage associated with meconium aspiration length. These fragments can form the ‘DNA ladder’ pattern seen on
syndrome (MAS). The implications of these findings regarding their potential elecrophoresis, a frequently used marker of apoptosis.
for the clinical management of MAS are also discussed. Caspase-independent mechanisms of apoptosis are mediated by
Journal of Perinatology (2008) 28, S108–S112; doi:10.1038/jp.2008.149 mitochondrial effectors; in this pathway, poly(ADP-ribose)
polymerase-1-mediated DNA damage is signaled to mitochondria.
This leads to release and translocation of apoptosis-inducing factor
(AIF) from the mitochondria to the nucleus. AIF enhances
endonuclease activity through its cooperation with endonuclease G
or cyclophilin A, which leads to large-scale DNA fragmentation and
General mechanisms in apoptosis chromatin condensation.
Apoptosis, sometimes referred to as programmed cell death (PCD) In contrast to apoptosis, which requires energy in the form of
or ‘cell suicide’, is a metabolically active process of cellular ATP, necrosis is the end result of a bioenergetic catastrophe
dismantling. Apoptosis avoids the inflammation often associated resulting from ATP depletion to a level incompatible with cell
with necrotic cell death because cellular contents are often not survival. Cells that die by necrosis frequently exhibit changes in
released from apoptotic cells as they are from necrotic cells. plasma membrane and nuclear morphology but not the organized
A variety of cellular signaling pathways regulates apoptosis, which chromatin condensation or DNA laddering that are characteristic of
is characterized morphologically by nuclear fragmentation, apoptosis.1 Necrosis-like PCD, or autophagy, is also considered as
cleavage of chromosomal DNA into internucleosomal fragments, alternative PCD that is caspase independent. Autophagy features
packaging of cellular debris into ‘apoptotic bodies’ without plasma degradation of cellular components within the intact dying cell in
membrane breakdown and exposure of surface molecules targeting autophagic vacuoles that undergoes phagocytosis. Autophagic cells
the cell remnants for phagocytosis.1,2 exhibit vacuolization, degradation of cytoplasmic contents and
Major signaling pathways leading to apoptosis involve the slight chromatin condensation; the molecular mechanisms
activation of cystein-dependent, aspartate-directed proteases termed regulating autophagy are now being elucidated.2,3
caspases. Depending on the type of stimuli and/or cell type, the
apoptotic cascade can follow either an intrinsic pathway, involving
Mechanisms of apoptosis specific to the lungs
Correspondence: Dr BD Uhal, Department of Physiology, Michigan State University,
3185 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA. The topic of apoptosis and its regulation in the lungs has been
E-mail: uhal@msu.edu reviewed recently;4 this section will briefly summarize apoptosis
Apoptosis and angiotensin II in meconium aspiration
BD Uhal and A Abdul-Hafez
S109

mechanisms specific to acute respiratory distress syndrome (ARDS), studies strongly suggest a role of epithelial apoptosis as a key
chronic obstructive pulmonary disease (COPD) and pulmonary profibrotic event in lung fibrogenesis. Apoptosis of alveolar
fibrosis (PF) that may be relevant to meconium aspiration epithelial cells (AECs) is found in patients with idiopathic PF (IPF)
syndrome (MAS). and in animal models of the disease.34–36 The epithelial apoptosis
Apoptosis of epithelial cells, neutrophils and endothelial cells are colocalizes with myofibroblasts where collagen deposition is
all thought to play roles in ARDS. The Fas-FasL system is thought severe.37 Blockade of apoptosis by either the angiotensin-converting
to play a role in epithelial apoptosis in ARDS on the basis that enzyme (ACE) inhibitor captopril, caspase inhibition or the forced
bronchoalveolar lavage (BAL) fluids obtained from ARDS patients expression of p21 in lung epithelial cells exerted antifibrotic effects
contain soluble Fas and FasL and induce apoptosis in lung in animal models.36,38,39 Together, these data suggested that the
epithelial cell line.5–8 Involvement of BAX, a homolog of Bcl-2 and fibrotic response is secondary to the apoptotic death of epithelial
its likely proapoptotic effect on type II pneumocytes was suggested cells in the lung. The death receptor Fas was found to be expressed
in studies of diffuse alveolar damage.9,10 Neutrophils were found to in AECs within the lungs of IPF patients and circulating levels of its
accumulate at inflammatory sites in the lung, in pulmonary ligand FasL correlated with disease activity.40–42 Similar
edema fluid and BAL fluid from patients with ARDS.11 This observations were observed in animal models.43,44
accumulation is due in part to delayed neutrophil apoptosis,; in Activation of Fas-induced apoptosis of bronchial and AECs is
addition, apoptosis of epithelial cells is enhanced at the same sufficient to initiate a fibrotic response in animal models.43
site.12–14 BAL fluids from patients with ARDS had elevated levels of However, development of bronchiolar and alveolar epithelial
granulocyte/macrophage colony-stimulating factor that inhibits apoptosis and fibrosis in the lungs of Fas-null mice was similar to
neutophil apoptosis through phosphoinositide 3-kinase and ERK wild-type mice,45,46 which suggests that other pathways different
pathway15,16 and stabilizes the factor Mcl-1 that is crucial for the from Fas can initiate epithelial apoptosis and facilitate fibrogenesis.
delay of apoptosis initiated by antiapoptotic factors.17,18 BAL fluids Other works have suggested the involvement of c-Jun N-terminal
from patients with ARDS was also cytotoxic to lung microvascular kinase and p38, members of the mitogen-activated protein kinases
endothelial cells due to the presence of TNF-a and angiostatin, an family pathways in the signaling of epithelial apoptosis.46,47
inhibitor of angiogenesis in vivo.19,20 Furthermore, apoptosis Finally, several lines of evidence have shown that the angiotensin
signal-regulating kinase (ASK)-1 was involved in apoptosis of system plays a critical role in AECs apoptosis and in lung
pulmonary vascular endothelial cells challenged with H2O2.21 fibrogenesis, and these will be discussed next.
Given the known roles of H2O2 generated by activated neutrophils
and other phagocytes, this suggests the possibility that inhibitors of
ASK-1 and its signaling pathway might have potential benefit in The endocrine RAS versus local tissue RASes
the management of ARDS. In the classical or ‘endocrine’ RAS described many years ago, the
Cell death by apoptosis is likely an important contributor to the 58 kDa precursor protein angiotensinogen (AGT) is synthesized
pathogenesis of COPD. Apoptosis of both inflammatory and alveolar primarily by the liver and is secreted into the circulation, where it
wall cells in emphysema was reported by several research is proteolytically cleaved by the kidney-derived aspartyl protease
groups.22–25 Induction of apoptosis by active caspase-3 instillation renin to yield the 10 amino-acid peptide angiotensin I (ANGI).
in mice caused architectural changes in the lungs similar to those ANGI travels through the vasculature and on entry into the
observed in human emphysema.26 Oxidant stress, such as that pulmonary vascular bed, is cleaved by the dipeptidyl
resulting from cigarette smoke, results in the killing of lung cells carboxypeptidase ACE to yield the eight amino-acid peptide
by both apoptosis and necrosis. Glutathione S-transferase was angiotensin II (ANGII). ANGII then travels through the blood and
shown to have a protective effect on tobacco smoke-induced elicits a number of endocrine functions including the release of
apoptosis.27,28 aldosterone from the adrenals and contraction of vascular smooth
Other studies have implicated endogenous growth factors and muscle throughout a variety of vascular beds.48
cytokines in COPD-associated apoptosis. One example is vascular More recently described local RASes, which occur in virtually
endothelial growth factor (VGEF); decreased VGEF expression or ever organ and tissue in which their presence has been evaluated,
blockade of VGEF receptors caused emphysematous changes in are classified as ‘extrinsic’ or ‘intrinsic’ depending on the origin of
lung architecture.29,30 Another group of endogenous mediators RAS system components. In extrinsic local RASes (RASL[ext]), such
believed to play role in apoptosis of parenchymal cells of the lung as is believed to occur in the heart,49 one or more of the RAS
in COPD is TNF-a, FasL and Fas. However, it is unclear at present components are synthesized locally but others are derived from the
whether these molecules are elevated or contribute to the severity of endocrine RAS. In intrinsic local RASes (RASL[int]), all of the
COPD31–33 and more work is needed to clarify these issues. components of a functional RAS are synthesized locally. In many
Recent studies support the notion that apoptosis contributes to local organ systems the term ‘RAS’ is a misnomer because often
the pathogenesis of lung fibrosis as well as to its resolution. Many renin is not expressed locally but other aspartyl proteases such as

Journal of Perinatology
Apoptosis and angiotensin II in meconium aspiration
BD Uhal and A Abdul-Hafez
S110

cathepsin D can accomplish the same proteolytic conversion of AGT by antisense oligonucleotides against AGT mRNA.54,55,62 Therefore,
to ANGI; moreover, other dipeptidyl carboxypeptidases such as not only do AECs produce ANGII in response to apoptosis inducers,
chymase can cleave ANGI to ANGII. For this reason, many tissues but the ANGII production is an essential component of the
have ‘ACE-independent’ pathways of ANGII formation that may not apoptotic response and blockade of ANGII production or receptor
be affected by specific ACE inhibitors. Regardless, most tissues interaction can block the cell death. Recent studies of human lung
exhibit some degree of expression of the major ANGII receptor biopsies from patients with IPF have found double labeling of ANG
subtypes AT1 and AT2, either at baseline or after injury, which peptides and markers of apoptosis in AECs;65 these data support the
often upregulates one or both of these receptors.50 notion that ANG peptide synthesis occurs in apoptotic AECs in the
intact injured human lung as well as in animal models and
cultured cells.
The role of RASes in lung injury and fibrogenesis
A wealth of literature supports the ability of ACE inhibitors or ANG
receptor blockers to attentuate experimental lung cell death and The role of RAS in MAS and potential for
fibrogenesis. The prototype ACE inhibitors captopril or lisinopril pharmacologic control
attenuated experimental lung fibrosis induced by monocrotaline,51 Apoptotic AECs have been observed in several models of
bleomycin52 or paraquat;53 the ACE inhibitors also were found to experimental MAS.66,67 Interest therefore was turned to the
be potent inhibitors of bleomycin- or TNF-a-induced apoptosis of possibility that AECs dying in response to meconium might also
lung epithelial cells.54,55 The ANG receptor AT1 antagonists require the local intrinsic RAS for their death and might be
losartan or candesartan have been shown to prevent lung fibrosis prevented from dying by ACE inhibitors of ANG receptor blockers.
in response to radiation56 or bleomycin,57,58 and also blocked Several lines of investigation support the theory that meconium
apoptosis of lung alveolar epithelial cells in response to does indeed induce expression of the local RAS that might be
bleomycin57 or the antiarrhythmic agent amiodarone.59 Given the amenable to pharmacologic blockade. Pretreatment of 2-week-old
central role of epithelial cells death in lung injury in a variety of rabbit pups with the ACE inhibitor captopril before
disease states, attention as been focused on defining the role of intratracheal instillation of human meconium prevented both
ANGII in apoptosis of lung alveolar epithelials (AECs). meconium-induced upregulation of interleukin (IL)-6 and IL-8
In cell culture studies, ANGII itself was found to be a potent and also reduced lung epithelial cell apoptosis detected by DNA
inducer of apoptosis in both the human AEC cell line A549 and in fragmentation assay.68 The captopril also appeared to reduced
primary AECs isolated from adult rats.60 The EC50s for meconium-induced upregulation of AGT mRNA.
ANGII-induced apoptosis in these cells (50 and 10 nM, respectively) In a similar study of rats, Lukkarinen et al.69 showed that
are far above the concentrations of ANGII found in the circulation pretreatement of rats with the nonselective ANG receptor blocker
(B1 to 10 pM), even in severe hypertension,48 which provides an saralasin, given intraperitoneally before meconium instillation,
explanation of why circulating ANGII does not kill AECs as it passes prevented AEC apoptosis detected by both DNA fragmentation assay
through the lungs. Although the cultured AECs expressed both the and electron microscopy. The meconium increase AGT mRNA in
AT1 and AT2 ANG receptor subtypes, only AT1-selective antagonists the lungs, suggesting apoptosis-induced upregulation of AGT in
could prevent ANGII-induced apoptosis.61 AECs. The saralasin also reduced meconium-induced upregulation
Subsequent studies found that when cultured AECs are exposed of endothelial monocyte-activating polypeptide (EMAP-II) and total
to other inducers of apoptosis, they begin to synthesize their own lung neutrophil accumulation, but had little effect on the
ANGII de novo, that is, from the precursor AGT. The apoptosis increased wet/dry weight ratio induced by the meconium. The
inducers Fas ligand,62 TNF-a,55 bleomycin54 and amiodarone63 all saralasin also had a small but statistically insignificant ability to
induce expression of the gene for AGT in AECs at the level of AGT reverse altered arterial blood gases.
transcription, and cause a significant increase in the steady-state Although not statistically significant, this latter result suggests
level of AGT mRNA. Moreover, AECs express all the enzymes that further refinement of dosing or route of administration might
necessary for conversion of AGT to the mature peptide ANGII, offer improvement of lung function, especially if more potent and
including ACE62 and the aspartyl protease cathepsin D.64 Thus, the selective ANGII antagonists (AT1 receptor-selective) or intratracheal
alveolar epithelial cell expresses its own intrinsic local RAS in administrations are explored. Regardless, all the data taken
response to a variety of apoptosis inducers. together strongly suggest that meconium, like other inducers of
Further work showed that the expression of this local RAS and AEC apoptosis and lung injury, elicits the expression of the local
the generation of ANGII by AECs are required for AEC apoptosis in RAS in AECs and that the RAS is intimately involved in their
response to FAS ligand, TNF-a, bleomycin or amiodarone. In all demise. Given the many agents already in clinical use for the
those cases, AEC apoptosis in response to the inducer could be manipulation of the RAS, it is proposed that further exploration of
blocked by either ANG receptor blockers,55,62 ACE inhibitors54,55 or antagonists of the RAS and creative approaches to their application

Journal of Perinatology
Apoptosis and angiotensin II in meconium aspiration
BD Uhal and A Abdul-Hafez
S111

(for example, aerosols) may offer clinically useful adjuncts to the apoptosis through phosphoinositide 3-kinase and extracellular signal-regulated kinase
current pharmacotherapy of MAS in the future. pathways. J Immunol 2000; 164: 4286–4291.
17 Leuenroth SJ, Grutkoski PS, Ayala A, Simms HH. The loss of Mcl-1 expression in
human polymorphonuclear leukocytes promotes apoptosis. J Leukoc Biol 2000; 68:
158–166.
18 Derouet M, Thomas L, Cross A, Moots RJ, Edwards SW. GM-CSF signalling and
Disclosure
proteasome inhibition delay neutrophil apoptosis by increasing the stability of Mcl-1.
BD Uhal has received consulting fees from Actelion Pharmaceuticals. A Abdul-Hafez has
J Biol Chem 2004; 279(26): 26915–26921.
declared no financial interests.
19 Hamacher J, Lucas R, Lijnen HR, Buschke S, Dunant Y, Wendel A et al. Tumor necrosis
factor-alpha and angiostatin are mediators of endothelial cytotoxicity in
bronchoalveolar lavages of patients with acute respiratory distress syndrome. Am J
Respir Crit Care Med 2002; 166: 651–656.
References 20 Luca R, Lijnen HR, Suffredini AF, Pepper MS, Steinberg KP, Martin TR et al. Increased
1 Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Curr angiostatin levels in bronchoalveolar lavage fluids from ARDS patients and from
Opin Cell Biol 2004; 16(6): 663–669. human volunteers after lung instillation of endotoxin. Thromb Haemost 2002; 87:
2 Fink SL, Cookson BT. Apoptosis, pyroptosis, and necrosis: mechanistic description of 966–971.
dead and dying eukaryotic cells. Infect Immun 2005; 73(4): 1907–1916. 21 Machino T, Hashimoto S, Maruoka S, Gon Y, Hayashi S, Mizumura K et al. Apoptosis
3 Krantic S, Mechawar N, Reix S, Quirion R. Apoptosis-inducing factor: a matter of signal-regulating kinase 1-mediated signaling pathway regulates hydrogen
neuron life and death. Prog Neurobiol 2007; 81(3): 179–196. peroxide-induced apoptosis in human pulmonary vascular endothelial cells.
4 Li X, Shu R, Filippatos G, Uhal BD. Apoptosis in lung injury and remodeling. J Applied Crit Care Med 2003; 31: 2776–2781.
Physiol. Invited Review, 2004; 97(4): 1535–1542. 22 Kasahara Y, Tuder RM, Cool CD, Voelkel NF. Expression of 15-lipoxygenase and
5 Albertine KH, Soulier MF, Wang Z, Ishizaka A, Hashimoto S, Zimmerman GA et al. Fas evidence for apoptosis in the lungs from patients with COPD. Chest 2000;
and fas ligand are up-regulated in pulmonary edema fluid and lung tissue of 117(5 Suppl 1): 260S.
patients with acute lung injury and the acute respiratory distress syndrome. Am 23 Kelly MG, Elborn JS, Kelly MG, Brown V, Ennis M. Measuring granulocyte apoptosis in
J Pathol 2002; 161: 1783–1796. airway inflammation. Thorax 2002; 57(4): 376; author reply 376. 30: Imai K,
6 Hodge SJ, Hodge GL, Reynolds PN, Scicchitano R, Holmes M. Increased production of D’Armiento J. Differential gene expression of sFRP-1 and apoptosis in pulmonary
TGF-beta and apoptosis of T lymphocytes isolated from peripheral blood in COPD. Am emphysema. Chest. 2002;121(3 Suppl):7S.
J Physiol Lung Cell Mol Physiol 2003; 285(2): L492–L499. 24 Majo J, Ghezzo H, Cosio MG. Lymphocyte population and apoptosis in the
7 Matute-Bello G, Liles WC, Steinberg KP, Kiener PA, Mongovin S, Chi EY et al. Soluble lungs of smokers and their relation to emphysema. Eur Respir J 2001; 17(5):
Fas ligand induces epithelial cell apoptosis in humans with acute lung injury (ARDS). 946–953.
J Immunol 1999; 163: 2217–2225. 25 Yokohori N, Aoshiba K, Nagai A, Respiratory Failure Research Group in Japan.
8 Ware LB, Matthay MA. Alveolar fluid clearance is impaired in the majority of patients Increased levels of cell death and proliferation in alveolar wall cells in patients with
with acute lung injury and the acute respiratory distress syndrome. Am J Respir Crit pulmonary emphysema. Chest 2004; 125(2): 626–632.
Care Med 2001; 163: 1376–1383. 26 Aoshiba K, Yokohori N, Nagai A. Alveolar wall apoptosis causes lung destruction and
9 Guinee D, Brambilla E, Fleming M, Hayashi T, Rahn M, Koss M et al. The potential emphysematous changes. Am J Respir Cell Mol Biol 2003; 28: 555–562.
role of BAX and BCL-2 expression in diffuse alveolar damage. Am J Pathol 1997; 151: 27 Ishii T, Fujishiro M, Masuda M, Nakajima J, Teramoto S, Ouchi Y et al. Depletion of
999–1007. glutathione S-transferase P1 induces apoptosis in human lung fibroblasts. Exp Lung
10 Husain KD, Stromberg PE, Javadi P, Buchman TG, Karl IE, Hotchkiss RS et al. Bcl-2 Res 2003; 29(7): 523–536.
inhibits gut epithelial apoptosis induced by acute lung injury in mice but has no effect 28 Ishii T, Matsuse T, Igarashi H, Masuda M, Teramoto S, Ouchi Y. Tobacco smoke
on survival. Shock 2003; 20: 437–443. reduces viability in human lung fibroblasts: protective effect of glutathione
11 Pittet JF, Mackersie RC, Martin TR, Matthay MA. Biological markers of acute lung S-transferase P1. Am J Physiol Lung Cell Mol Physiol 2001; 280(6):
injury: prognostic and pathogenetic significance. Am J Respir Crit Care Med 1997; L1189–L1195.
155: 1187–1205. 29 Tsao PN, Su YN, Li H, Huang PH, Chien CT, Lai YL et al. Overexpression of placenta
12 Goodman ER, Stricker P, Velavicius M, Fonseca R, Kleinstein E, Lavery R et al. Role of growth factor contributes to the pathogenesis of pulmonary emphysema. Am J Respir
granulocyte-macrophage colony-stimulating factor and its receptor in the genesis of Crit Care Med 2004; 169(4): 505–511.
acute respiratory distress syndrome through an effect on neutrophil apoptosis. Arch 30 Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, Hirth PK et al.
Surg 1999; 134: 1049–1054. Inhibition of VEGF receptors causes lung cell apoptosis and emphysema. J Clin Invest
13 Matute-Bello G, Liles WC, Radella II F, Steinberg KP, Ruzinski JT, Hudson LD et al. 2000; 106(11): 1311–1319.
Modulation of neutrophil apoptosis by granulocyte colony-stimulating factor and 31 Lucey EC, Keane J, Kuang PP, Snider GL, Goldstein RH. Severity of elastase-induced
granulocyte/macrophage colony-stimulating factor during the course of acute emphysema is decreased in tumor necrosis factor-alpha and interleukin-1beta
respiratory distress syndrome. Crit Care Med 2000; 28: 1–7. receptor-deficient mice. Lab Invest 2002; 82(1): 79–85.
14 Parsey MV, Kaneko D, Shenkar R, Abraham E. Neutrophil apoptosis in the lung after 32 Takabatake N, Nakamura H, Inoue S, Terashita K, Yuki H, Kato S et al. Circulating
hemorrhage or endotoxemia: apoptosis and migration are independent of IL-1beta. levels of soluble Fas ligand and soluble Fas in patients with chronic obstructive
Clin Immunol 1999; 91: 219–225. pulmonary disease. Respir Med 2000; 94(12): 1215–1220.
15 Matute-Bello G, Liles WC, Radella II F, Steinberg KP, Ruzinski JT, Hudson LD et al. 33 Yasuda N, Gotoh K, Minatoguchi S, Asano K, Nishigaki K, Nomura M et al. An increase
Modulation of neutrophil apoptosis by granulocyte colony-stimulating factor and of soluble Fas, an inhibitor of apoptosis, associated with progression of COPD. Respir
granulocyte/macrophage colony-stimulating factor during the course of acute Med 1998; 92(8): 993–999.
respiratory distress syndrome. Crit Care Med 2000; 28: 1–7. 34 Kuwano K, Kunitake R, Kawasaki M. p21 and p53 expression in association with DNA
16 Klein JB, Rane MJ, Scherzer JA, Coxon PY, Kettritz R, Mathiesen JM et al. strand breaks in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 1996; 154:
Granulocyte-macrophage colony-stimulating factor delays neutrophil constitutive 477–483.

Journal of Perinatology
Apoptosis and angiotensin II in meconium aspiration
BD Uhal and A Abdul-Hafez
S112

35 Hagimoto N, Kuwano K, Nomoto Y, Kunitake R, Hara N. Apoptosis and expression of 52 Wang R, Ibarra-Sunga O, Pick R, Uhal BD. Abrogation of bleomycin-induced epithelial
Fas/Fas ligand mRNA in bleomycin-induced pulmonary fibrosis in mice. Am J Respir apoptosis and lung fibrosis by captopril or by a caspase inhibitor. Am J Physiol Lung
Cell Mol Biol 1997; 16: 91–101. Cell Mol Physiol 2000; 279: L143–L151.
36 Wang R, Ibarra-Sunga O, Verlinski L, Pick R, Uhal BD. Abrogation of 53 Mohammadi-Karakani A. Lisinopril ameliorates paraquat-induced lung fibrosis. Clin
bleomycin-induced epithelial apoptosis and lung fibrosis by captopril or by a caspase Chim Acta 2006; 367(1–2): 170–174.
inhibitor. Am J Physiol Lung Cell Mol Physiol 2000; 279: L143–L151. 54 Li X, Zhang H, Soledad-Conrad V, Zhuang J, Uhal BD. Bleomycin-induced apoptosis of
37 Uhal BD, Joshi I, Hughes WF, Ramos C, Pardo A, Selman M. AEC death adjacent to alveolar epithelial cells requires angiotensin synthesis de novo. Am J Physiol 2003;
underlying myofibroblasts in advanced fibrotic human lung. Am J Physiol 1998; 275: 284(3): L501–L507.
L1192–L1199. 55 Wang R, Alam G, Zagariya A, Gidea G, Pinillos H, Lalude O et al. Apoptosis of lung
38 Kuwano K, Kunitake R, Maeyama T, Hagimoto N, Kawasaki M, Matsuba T et al. epithelial cells in response to TNF-alpha requires angiotensin II generation de novo.
Attenuation of bleomycin-induced pneumopathy in mice by a caspase inhibitor. Am J J Cell Physiol 2000; 185(2): 253–259.
Physiol Lung Cell Mol Physiol 2001; 280: L316–L325. 56 Molteni A, Moulder JE, Cohen EF, Ward WF, Fish BL, Taylor JM et al. Control of
39 Inoshima I, Kuwano K, Hamada N, Hagimoto N, Yoshimi M, Maeyama T et al. radiation-induced pneumopathy and lung fibrosis by angiotensin-converting enzyme
Anti-monocyte chemoattractant protein-1 gene therapy attenuates pulmonary inhibitors and an ANG II type 1 receptor blocker. Int J Radiat Biol 2000; 76: 523–532.
fibrosis in mice. Am J Physiol Lung Cell Mol Physiol 2004; 286: 57 Li X, Rayford H, Uhal BD. Essential roles for angiotensin receptor AT1a in bleomycin-
L1038–L1044. induced apoptosis and lung fibrosis in mice. Am J Pathol 2003; 163(6): 2523–2530.
40 Domagala-Kulawik J, Droszcz P, Kraszewska I, Chazan R. Expression of Fas antigen in 58 Otsuka M, Takahashi H, Shiratori M, Chiba H, Abe S. Reduction of bleomycin induced
the cells from bronchoalveolar lavage fluid (BALF). Folia Histochem Cytobiol 2000; lung fibrosis by candesartan cilexetil, an angiotensin II type 1 receptor antagonist.
38: 185–188. Thorax 2004; 59(1): 31–38.
41 Kazufumi M, Sonoko N, Masanori K, Takateru I, Akira O. Expression of bcl-2 protein 59 Uhal BD, Wang R, Laukka J, Zhuang J, Soledad-Conrad V, Filippatos G. Inhibition of
and APO-1 (Fas antigen) in the lung tissue from patients with idiopathic pulmonary amiodarone-induced lung fibrosis but not alveolitis by angiotensin system antagonists.
fibrosis. Microsc Res Tech 1997; 38: 480–487. Pharmacol Toxicol 2003; 92: 81–87.
42 Kuwano K, Maeyama T, Inoshima I, Ninomiya K, Hagimoto N, Yoshimi M et al. 60 Wang R, Zagariya A, Ibarra-Sunga O, Gidea C, Ang E, Deshmukh S et al. Angiotensin
Increased circulating levels of soluble Fas ligand are correlated with disease activity in II induces apoptosis in human and rat alveolar epithelial cells. Am J Physiol 1999;
patients with fibrosing lung diseases. Respirology 2002; 7: 15–21. 276: L885–L889.
43 Hagimoto N, Kuwano K, Miyazaki H. Induction of apoptosis and pulmonary fibrosis 61 Papp M, Li X, Zhuang J, Wang R, Uhal BD. Angiotensin receptor subtype AT1
in mice in response to ligation of FAS antigen. Am J Respir Cell Mol Biol 1997; 17: mediates alveolar epithelial cell apoptosis in response to ANGII. Am J Physiol 2002;
272–278. 282: L713–L718.
44 Hagimoto N, Kuwano K, Nomoto Y, Kunitake R, Hara N. Apoptosis and expression of 62 Wang R, Zagariya A, Ang E, Ibarra-Sunga E, Uhal BD. Fas-induced apoptosis of
Fas/Fas ligand mRNA in bleomycin-induced pulmonary fibrosis in mice. Am J Respir alveolar epithelial cells requires angiotensin II generation and receptor interaction. Am
Cell Mol Biol 1997; 16: 91–101. J Physiol 1999; 277: L1245–L1250.
45 Aoshiba K, Yasui S, Tamaoki J, Nagai A. The Fas/Fas-ligand system is not required for 63 Uhal B, Zhang H, Abdul-Hafez A, Shu R, Uhal BD. Amiodarone induces
bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med 2000; 162: angiotensinogen gene expression in lung alveolar epithelial cells through AP-1. Basic
695–700. Clin Pharmacol Toxicol 2007; 100: 59–66.
46 Matsuoka H, Arai T, Mori M, Goya S, Kida H, Morishita H et al. p38 MAPK inhibitor, 64 Li X, Rayford H, Shu R, Zhuang J, Uhal BD. Essential role for cathepsin D in
FR-167653, ameliorates murine bleomycin-induced pulmonary fibrosis. Am J Physiol bleomycin-induced apoptosis of alveolar epithelial cells. Am J Physiol 2004; 287(1):
Lung Cell Mol Physiol 2022; 283: L103–L112. L46–L51.
47 Yoshida K, Kuwano K, Hagimoto N, Watanabe K, Matsuba T, Fujita M et al. MAP 65 Li X, Molina-Molina M, Abdul-Hafez A, Ramirez J, Serrano-Mollar A, Xaubet A et al.
kinase activation and apoptosis in lung tissues from patients with idiopathic Extravascular sources of lung angiotensin peptide synthesis in idiopathic pulmonary
pulmonary fibrosis. J Pathol 2002; 198: 388–396. fibrosis. Am J Physiol Lung Cell Mol Physiol 2006; 291(5): L887–L895.
48 Filippatos G, Tilak M, Pinillos H, Uhal BD. Regulation of apoptosis by 66 Lu MP, Du LZ, Gu WZ, Chen XX. Nitric oxide inhalation inhibits inducible nitric oxide
angiotensin II in the heart and lungs (Invited Review). Int J Mol Med 2001; 7(3): synthase but not nitrotyrosine formation and cell apoptosis in rat lungs with
273–280. meconium-induced injury. Acta Pharmacol Sin 2005; 26(9): 1123–1129.
49 Von Lutterotti N, Catanzaro DF, Sealey JE, Laragh JH. Renin is not synthesized by 67 Zagariya A, Bhat R, Chari G, Uhal B, Navale S, Vidyasagar D. Apoptosis of airway
cardiac and extrarenal vascular tissues. A review of experimental evidence. Circulation epithelial cells in response to meconium. Life Sci 2005; 76(16): 1849–1858.
1994; 89: 458–470. 68 Zagariya A, Bhat R, Navale S, Chari G, Vidyasagar D. Inhibition of meconium-induced
50 Atlas SS. The renin angiotensin system revisited: classical and nonclassical pathways of cytokine expression and cell apoptosis by pretreatment with captopril. Pediatrics 2006;
angiotensin formation. Mount Sinai J Med 1998; 65: 87–96. 117(5): 1722–1727.
51 Molteni A, Ward W, Ts’ao C, Solliday N, Dunne M. Monocrotaline-induced pulmonary 69 Lukkarinen H, Laine J, Lehtonen J, Zagariya A, Vidyasagar D, Aho H et al.
fibrosis in rats: amelioration by captopril and penicillamine. Proc Soc Exp Biol Med Angiotensin II receptor blockade inhibits pneumocyte apoptosis in experimental
1985; 180: 112–120. meconium aspiration. Pediatr Res 2004; 55(2): 326–333.

Journal of Perinatology
Journal of Perinatology (2008) 28, S113–S115
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Toxic effects of different meconium fractions on lung function:
new therapeutic strategies for meconium aspiration syndrome?
OD Saugstad1,2, PA Tølløfsrud1,2, P Lindenskov1,2 and CA Drevon1,2
1
Department of Pediatric Research and Pediatrics, Rikshospitalet Medical Center, University of Oslo, Oslo, Norway and
2
Department of Nutrition, Institute of Basic Medical Sciences, Medical Faculty, University of Oslo, Oslo, Norway

bile acids of meconium are cholic, hydroxycholic and


To review and summarize experimental data examining the effects of chenodeoxycholic acids.1–3
different fractions of meconium, and to test the effect of albumin on These different subfractions of meconium are leading to reduced
meconium aspiration both as prophylactic and rescue treatment. Newborn activity of surfactant and inflammation therefore promoting
piglets 2 to 5 days of age were made hypoxic and then instilled meconium atelectasis and impaired gas exchange. Both hydrophilic and
or fractions of meconium intratracheally. Meconium-added albumin and hydrophobic components of meconium reduce surfactant function.
albumin instilled after meconium were also tested. Lung function and Furthermore, the lipid fraction has a stronger inhibitory effect on
inflammatory cytokines were measured. Both the lipid- and water-soluble surfactant function than the water-soluble subfraction.4–7 It is well
fractions induce inflammation in the lungs with elevation of inflammatory known that bile salts may cause inflammation8,9 and that FFA,
cytokines. When meconium was mixed with albumin, the inflammatory such as oleic acid, induce severe lung failure when administered
effects of meconium were significantly ameliorated. Rescue therapy with intravenously.10
intratracheal albumin 5 min after the meconium aspiration syndrome was We have tested the effects of different meconium subfractions on
induced also improved lung function. These results indicate that at least pulmonary function in an animal model of newborn piglets.
part of the symptoms seen in the meconium aspiration syndrome could be Because albumin is able to bind FFA, we also examined whether
prevented by blocking the active substances of meconium such as bile acids albumin added to meconium alleviates the clinical symptoms of
and free fatty acids. meconium aspiration. In case such an inhibitory effect was found,
Journal of Perinatology (2008) 28, S113–S115; doi:10.1038/jp.2008.151 perhaps this also could be used as rescue therapy. We therefore
developed a rescue model instilling albumin intratracheally
following meconium aspiration. In this article we summarize and
review some of our previous results.
Introduction
Aspiration of meconium in the airways of the newborn infant leads
to plugging and subsequently to inflammatory changes, surfactant Methods
inhibition, respiratory failure and pulmonary hypertension. There We used newborn piglets undergoing hypoxia and meconium
have been three approaches to moderate these effects: (1) dilute aspiration followed by resuscitation.11 Human meconium was
meconium (amnioinfusion, lavage), (2) improve/‘strengthen’ collected from healthy term newborns and diluted in distilled
surfactant by adding for instance polymers and (3) inhibit harmful water, and rotavaporated dry. It was sterilized by g-irradiation and
effects of meconium per se. diluted with sterile water to 110 mg dry weight per ml. Hypoxia
Meconium is a complex mixture containing 75 to 80% water. with FiO2 of 0.08 was induced in newborn piglets 2 to 5 days of
The other chemical components can be divided into lipid- and age until they were close to collapse. The piglets were then
water-soluble material. The lipid includes free fatty acids (FFA), hand-ventilated for 30 s and put back on ventilator with rate
triglycerides and cholesterol. The FFA include mainly palmitic, increased from 30 to 60 per min, and peak inspiratory pressure
stearic and oleic acids. The water-soluble components include bile increased to 5 Cm H2O. Tidal volume was kept constant at 10 to
acids, bilirubin, dietary fibers and inorganic molecules. The main 15 ml kg 1 and paCO2 4.5 to 6 kPa (34 to 45 mm Hg).
In the first study 3 ml kg 1 body weight of meconium was
Correspondence: Dr OD Saugstad, Pediatrisk Forskningsinstitutt, Rikshospitalet, Oslo 0027,
Norway. instilled intratracheally giving an oxygenation index (OI) of 6 to 7
E-mail: o.d.saugstad@medisin.uio.no indicating a mild meconium aspiration syndrome (MAS). In this
Toxic effects of meconium fractions on lung
OD Saugstad et al
S114

study meconium was also mixed with albumin (30% albumin in but does not seem to have significant clinical effects in the doses
stoichiometric relation to FFA estimated to be 1.4 ml kg 1) before and administration tested out.
intratracheal in administration of the mixture.12,13 Both the lipid- and water-soluble fractions of meconium induce
In the second study 3 ml kg 1 body weight of either meconium, pulmonary inflammation and injury, however, less than
lipid- or water-soluble meconium fractions was instilled meconium alone. The lipid fraction was responsible for most of
intratracheally in separate groups.14 the cytokine activation assessed by IL-8 in tracheal aspirate.
In the third study a moderate and severe model was developed. A However, the water-soluble fraction also had some inflammatory
moderate MAS was developed by instilling meconium of 4 ml kg 1 properties.
body weight, giving an OI of approximately 12. A severe MAS was We chose albumin as a protein with high capacity to bind FFA
induced by instilling 5 ml meconium mixture per kg leading to a in spite of the fact it is known that albumin also inhibits surfactant
typical OI of 25. Five min after meconium had been instilled, 3 ml function. However, in our animal model, and probably in patients
of 30% albumin was instilled intratracheally.15 with MAS as well, surfactant function is already abolished.
The animals in all studies were resuscitated for 8 h with room Albumin added to meconium before intratracheal instillation,
air or supplemental O2 to achieve a SaO2 of 85%. almost completely blocked the negative effects of meconium.
Albumin seems to prevent development of MAS when added to
meconium. However, when albumin is given as rescue therapy the
Results effect is less clear. In another of our studies rescue therapy by
Effect of albumin lavaging the lungs with excess albumin gave worse outcome than
Animals receiving albumin mixed with meconium compared with meconium alone (submitted). Rescue therapy with albumin in low
meconium alone had a significantly reduced OI as well as doses and low volume therefore had a small but significant and
ventilation index. OI was approximately 50% in animals given positive effect on compliance, however, larger doses and volumes
meconium mixed with albumin compared with those given seem to have a detrimental effect on pulmonary function.
meconium. The need for oxygen and mean airway pressure on the The optimal albumin dose, timing and mode of administration
ventilator was also significantly reduced and compliance increased therefore are unknown. Other inhibitors of both the lipid- and
in animals given the combination of meconium and albumin.12 water-soluble fractions of meconium should be searched for and
The inflammatory cytokine interleukin (IL)-8 in tracheal tested aiming at developing an efficient rescue therapy. Several
aspirate, 8 h after reoxygenation was started, was in mean fivefold such inhibitors as polymers and anti-inflammatory agents16,17
higher (P<0.05) in animals given meconium alone (93 pg ml 1) have been described already. Furthermore, meconium-induced
as compared to those given meconium mixed with albumin apoptosis may be inhibited by angiotensin-converting enzyme
(18 pg ml 1) before intratracheal instillation.13 inhibitors, which may reduce injury.18,19
Meconium is a potent complement activator and complement
Effect of meconium fractions inhibition may represent a future therapeutic principle in MAS.20
Both the lipid- and water-soluble fractions of meconium induced In the future all the principles discussed above may be combined to
inflammatory changes, however, more in the lipid extract than the develop a powerful therapy for MAS. Given the available presented
water extract group. At 8 h after instillation of meconium, IL-8 in data, there does not appear to be a robust argument for the use of
tracheal aspirate was threefold higher in the lipid extract group than albumin in the human infant with meconium aspiration, let alone
in the animals given the water-soluble fractions of meconium. Intact suggest that there may be ‘an optimum dose’, timing and mode of
meconium had more severe effects on the lungs than the lipid- and administration.
water-soluble fractions of meconium separately.14

Effect of rescue therapy with albumin Acknowledgments


Rescue therapy with a low-dose intratracheal albumin These studies have been supported by the Norwegian Council for Research.
(stoichiometric binding of FFA) did not affect OI or IL-8 in tracheal
aspirate when measured at 8 h. However, a significantly attenuated
decrease in lung compliance was found.15 Disclosure
The authors have declared no financial interests.

Discussion
In these studies we found that albumin inhibits clinical effects of References
meconium and that the lipid fraction is more harmful than the 1 Terasaka D, Clark DA, Singh BN, Rokahr J. Free fatty acids of human meconium.
water fraction. Rescue therapy with albumin increases compliance Biol Neonate 1986; 50: 16–20.

Journal of Perinatology
Toxic effects of meconium fractions on lung
OD Saugstad et al
S115

2 Harries JT. Meconium in health and disease. Br Med Bull 1978; 34: 75–78. 13 Tllfsrud PA, Medb S, Solås AB, Robertson B, Speer CP, Seidenspinner S et al.
3 Moses D, Holm BA, Spitale P, Liu MY, Enhorning G. Inhibition of pulmonary Intratracheal albumin reduces interleukin-8 in tracheobronchial aspirates in piglets
surfactant function by meconium. Am J Obstet Gynecol 1991; 164: 477–481. after meconium aspiration. J Perinat Med 2004; 32: 78–83.
4 Sun B, Curstedt T, Robertson B. Surfactant inhibition in experimental meconium 14 Tllfsrud PA, Lindenskov PH, Drevon CA, Speer CP, Seidenspinner S,
aspiration. Acta Paediatr 1993; 82: 182–189. Saugstad OD. Comparison of pulmonary and inflammatory effects of lipid- and water-
5 Schrama AJ, de Beaufort AJ, Sukul YR, Jansen SM, Poorthuis BJ, Berger HM. soluble components in meconium in newborn piglets. Biol Neonate 2003; 84:
Phospholipase A2 is present in meconium and inhibits the activity of pulmonary 330–337.
surfactant: an in vitro study. Acta Paediatr 2001; 90: 412–416. 15 Lindenskov PH, Castellheim A, Aamodt G, Saugstad OD. Meconium induced IL-8
6 Hall SB, Lu RZ, Venkitaraman AR, Hyde RW, Notter RH. Inhibition of pulmonary production and intratracheal albumin alleviated lung injury in newborn pigs. Pediatr
surfactant by oleic acid: mechanisms and characteristics. J Appl Physiol 1992; 72: Res 2005; 57: 371–377.
1708–1716. 16 Lu KW, Goerke J, Clements JA, Taeusch HW. Hyaluronan reduces surfactant
7 Wang Z, Notter RH. Additivity of protein and nonprotein inhibitors of lung surfactant inhibition and improves rat lung function after meconium injury. Pediatr Res 2005;
activity. Am J Respir Crit Care Med 1998; 158: 28–35. 58: 206–210.
8 Henderson RD, Fung K, Cullen JB, Milne EN, Marryatt G. Bile aspiration: an 17 Korhonen K, Kiuru A, Svedstrom E, Kaapa P. Pentoxifylline reduces regional
experimental study in rabbits. Can J Surg 1975; 18: 64–69. inflammatory and ventilatory disturbances in meconium-exposed piglet lungs. Pediatr
9 Zecca E, Costa S, Lauriola V, Vento G, Papacci P, Romagnoli C. Bile acid pneumonia: a Res 2004; 56: 901–906.
‘new’ form of neonatal respiratory distress syndrome? Pediatrics 2004; 114: 269–272. 18 Zagariya A, Bhat R, Chari G, Uhal B, Navale S, Vidyasagar D. Apoptosis of airway
10 Trawoger R, Cereda M, Kolobow T. A standardized method of oleic acid infusion in epithelial cells in response to meconium. Life Sci 2005; 76: 1849–1858.
experimental acute respiratory failure. Scand J Clin Lab Invest 2001; 61: 75–81. 19 Zagariya A, Bhat R, Navale S, Chari G, Vidyasagar D. Inhibition of meconium-induced
11 Tllfsrud PA, Solås AB, Saugstad OD. Newborn piglets with meconium aspiration cytokine expression and cell apoptosis by pretreatment with captopril. Pediatrics 2006;
resuscitated with room air or 100% oxygen. Pediatr Res 2001; 50: 423–429. 117: 1722–1727.
12 Tllfsrud PA, Medb S, Solås AB, Drevon CA, Saugstad OD. Albumin mixed with 20 Castellheim A, Lindenskov PH, Pharo A, Fung M, Saugstad OD, Mollnes TE. Meconium
meconium attenuates pulmonary dysfunction in a newborn piglet model with is a potent activator of complement in human serum and in piglets. Pediatr Res 2004;
meconium aspiration. Pediatr Res 2002; 52: 545–553. 55: 310–318.

Journal of Perinatology
Journal of Perinatology (2008) 28, S116–S119
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
The role of complement in meconium aspiration syndrome
TE Mollnes1, A Castellheim1,2, PHH Lindenskov1,2, B Salvesen1,2 and OD Saugstad2
1
Institute of Immunology, Rikshospitalet, Oslo, Norway and 2Department of Pediatric Research, Rikshospitalet, Medical Faculty,
University of Oslo, Oslo, Norway

branches that are induced or activated by meconium: cytokines,


The complement system is part of the host defense with a number of arachidonic acid metabolites and reactive oxygen species.
biological effects, most of which contribute to the inflammatory reaction by In vitro meconium has been shown to trigger peritoneal
activation of cells like leukocytes and endothelial cells. An intact macrophages to produce a proinflammatory tumor necrosis
complement system is required for protection against infection and for factor-a (TNF-a) response13 and in animals instilled with human
maintaining internal inflammatory homeostasis. However, the system is a meconium interleukin (IL)-6, IL-8, TNF-a and IL-1b have been
double-edged sword as improperly or uncontrolled activation is detected in cells harvested from lung lavage within 8 h after
disadvantageous and potentially harmful for the host. Meconium aspiration meconium instillation.14 In another animal studies with the same
syndrome (MAS) is associated with a local inflammatory reaction in the observation time, TNF-a15 and IL-8 have been detected in
lungs, frequently described as a chemical pneumonitis. Cytokines, bronchoalveolar lavage fluid16,17 whereas IL-5 and IL-13 were
arachidonic acid metabolites and reactive oxygen species are involved in elevated at day 7 in mice.18 Furthermore, meconium may
this reaction. We have recently documented that meconium is a potent stimulate chemotaxis, the latter effect claimed to be because of IL-8
activator of complement in vitro and in an experimental piglet model of present in meconium.19 Finally, the levels of IL-1b, IL-6, IL-8 and
MAS, the latter presenting with an inflammatory profile closely resembling TNF-a have been compared in sterile meconium vs meconium
systemic inflammatory response syndrome. We postulate that complement contaminated with bacteria and in spite of variation among
activation may contribute to the pathogenesis of MAS. samples the median concentrations did not differ.20 The
Journal of Perinatology (2008) 28, S116–S119; doi:10.1038/jp.2008.148 transcription factor NF-kB, known to induce a number of
proinflammatory cytokines, was shown to be activated by
meconium in rat alveolar macrophages.21
Human meconium is shown to contain phospholipase A2,
thereby capable of directly damaging alveolar cells.22 Meconium
was found to increase the production of arachidonic acid
Inflammatory mediators in MAS metabolites by phospholipase A223 and elevated catalytic activity of
The pathophysiology of meconium aspiration syndrome (MAS) is phospholipase A2 was found both in tissue and lavage fluid.24
complex. Several mechanisms may be operative in the development In addition, meconium has been shown capable of upregulating
of lung injury in MAS, including mechanical obstruction,1 cyclooxygenase-2 mRNA expression in rat lungs, a process
inactivation of surfactant,2 pulmonary hypertension3–6 and lung that is inhibited by dexamethasone, but not indomethacin25
inflammation.7 Although the controversies still remain in the and enhance the release of thromboxane-A2 in human airway
literature as to what extent meconium per se leads to MAS,8,9 epithelial cells.26 Finally, apoptosis associated with
increasing evidence indicates that the inflammation induced by meconium22,27,28 could be inhibited by angiotensin II receptor
meconium is an essential part of the pathophysiology.7,10 The blockade.27
assumed chemical pneumonitis caused by meconium was first The effect of meconium on oxidative burst (release of reactive
documented in rabbits by infiltration of polymorphonuclear oxygen species) has been difficult to interpret. Although one study
leukocytes in alveolar septa within 6 h after instillation of showed that meconium stimulated alveolar macrophages to
meconium,11 and a later study documented increased counts and produce superoxide anion,29 another study showed that light and
chemotactic activity of neutrophils in lung lavage fluid from pigs.12 very light meconium inhibited neutrophil oxidative burst.30
Leukocytes are important sources for three main inflammatory Recently, it has been showed that meconium has an inhibitory
effect on neutrophils in low concentrations (0.2 mg ml 1) but in
Correspondence: Dr TE Mollnes, Institute of Immunology, Rikshospitalet, Oslo NO-0027,
Norway.
higher concentrations (1 and 2 mg ml 1) stimulates neutrophil
E-mail: t.e.mollnes@medisin.uio.no oxygen radical production progressively.31 A possible role for
Complement and meconium
TE Mollnes et al
S117

complement in meconium-induced oxidative burst has been cleaved by factor D and the alternative pathway C3 convertase,
recently postulated (see below). C3bBb, is formed leading to further cleavage of C3. C5 is then
activated in the same manner as for the classical and lectin
pathway C5 convertase.
The complement system Thus, all three pathways converge at C3 and the terminal
Complement is one of the plasma cascade systems and represents a pathway (lower part of Figure 1) proceeds in the same way
main part of fluid-phase innate immunity.32 It consists of more irrespective of the initial pathway activation, by assembly of C7 to
than 30 proteins acting together in a specific manner with the C5b-6, forming an amphiphilic complex able to insert into a lipid
principal function to protect the host against invading organisms. membrane. One C5b-7 moiety binds one C8 and one or several C9
A simplified cartoon of the complement system is shown in molecules, creating a physical pore penetrating the membrane
Figure 1. There are three initial pathways of activation. (C5b-9(m) or membrane attack complex, leading to
The classical pathway (upper left) is activated either by transmembrane leakage and subsequent cell activation, or more
natural or elicited antibodies binding to antigen, or by direct, infrequently to lysis (lower right). If the activation occurs in the
antibody-independent activation, for example, by C-reactive fluid phase and there is no membrane present, the C5b-7 complex
protein. C1q triggers the serine proteases C1r and C1s, the latter binds to vitronectin and clusterin (fluid-phase regulators of the
cleaving C4 to C4b, which exposes a specific binding site for C2. terminal pathway) and thus retains hydrophilic properties. Final
C1s then cleaves C2 and the resulting C3 convertase C4b2a cleaves assembly of a soluble C5b-9 (SC5b-9), the second form of the
C3 to C3b to form the C5 convertase C4b2a3b. Splitting of C5 to the terminal complement complex (TCC), occurs by binding of C8 and
highly potent anaphylatoxin C5a and the C6-binding fragment C5b C9. Soluble TCC is a particularly useful indicator of complement
is the last enzymatic step in the cascade. Activation of the lectin activation and an assay for use in humans, baboons and pigs has
pathway (upper middle) is initiated by mannose-binding lectin been developed.33
(MBL) recognizing mannose on bacteria, by immunoglobulin A Complement activation is strictly regulated by inhibitory
and probably by structures exposed by damaged endothelium. MBL proteins of which some are illustrated in Figure 1 (boxes with
is homologous to C1q and triggers the MBL-associated serine broken lines). The biological potency of this system is substantial
proteases (MASPs), of which three forms (MASP1, MASP2 and and when activated out of control it may break down essential
MASP3) have been described. Further lectin pathway activation is homeostatic mechanisms, for example, as seen in irreversible septic
virtually identical to classical pathway activation forming the same shock.34 Thus, it is crucially important to keep the system under
C3 and C5 convertases. The alternative pathway (upper right) control by the regulatory proteins. C1 inhibitor regulates the initial
activation differs from the classical and lectin pathways by a classical and lectin pathways. Carboxypeptidase N inactivates the
physiological, low-graded spontaneous hydrolysis of the internal anaphylatoxins C5a, C3a and C4a, of which C5a is particularly
thiol ester of the C3 molecule thereby binding factor B, which is potent. Factor I cleaves and inactivates C4b and C3b and uses
then cleaved by factor D. The C3 convertase then formed cleaves C3 C4b-binding protein (C4BP) as cofactor in the classical/lectin
to C3a and C3b, the latter binds factor B forming C3bB, which is pathway and factor H in the alternative pathway. The membrane
regulators complement receptor 1 (CR1; CD35), membrane
cofactor protein (MCP; CD46) and decay-accelerating factor
(DAF; CD55) regulate complement activation by either acting as
cofactors for factor I-mediated cleavage of C4b and C3b (CR1 and
MCP), or accelerating the decay of the biomolecular C3 and C5
convertases (CR1 and DAF). CD59, also a membrane regulator,
prevents the binding of C9 to the C5b-8 complex in the terminal
pathway.
The main biological effect of complement activation is
induction of an inflammatory reaction. Many of the activation
products are involved in this process, but C5a is particularly
important. Innumerable inflammatory mediators are induced by
Figure 1 Schematic illustration of the complement system. Activation may C5a, when it reacts with its receptor (C5aR). Most of the
occur through three initial routes: the classical, the lectin and the alterative inflammatory branches can be triggered by C5a (Figure 2),
pathways. All three pathways converge at the level of C3 and proceed through the illustrating that complement has a key function as an upstream
terminal pathway. Biologic active fragments are released when native components
are activated, like the potent anaphylatoxin C5a when C5 is cleaved. To keep the
mediator of a broad spectrum of inflammatory reactions. Thus,
system under strict control, a number of inhibitor and regulatory proteins are activated complement is a double-edged sword with undesired
present, as illustrated with dotted boxes and lines. effects in many conditions. Various reagents with potential

Journal of Perinatology
Complement and meconium
TE Mollnes et al
S118

we demonstrated that complement is activated systemically, as


detected by a substantial increase in plasma TCC. The increase in
TCC correlated positively with oxygenation and ventilatory indexes
and negatively with lung compliance. Finally, the mortality of the
piglets correlated with the increase in TCC, indicating that the
breakdown in homeostatic mechanisms may be associated with
uncontrolled complement activation. These observations led us to
further investigate the role of complement in experimental MAS.40
In this study we found that experimental MAS piglets reflected an
inflammatory response, measured by cytokines and complement,
closely resembling the systemic inflammatory response syndrome
Figure 2 Biological effects of C5a. The potent anaphylatoxin C5a released (SIRS). We therefore propose that MAS is associated with a general
during complement activation induces a number inflammatory mediators of whole-body SIRS-like inflammatory reaction. It remains, however,
which several are released to the fluid phase whereas others are expressed on the to be shown whether complement activation actually contributes to
surface of cells. The fluid-phase mediators may then act directly as inflammatory
agents or may trigger secondary effects through receptor binding. The surface
the pathophysiology of clinical MAS and if inhibition of
proteins induced by C5a include adhesion molecules, leading to trafficking of cells complement may attenuate inflammation and severity in MAS.
through interaction between endothelial cells and leukocytes. Lately, we have focused our research on the two main branches
of innate immunity: the fluid-phase complement system and the
cellular Toll-like receptor (TLR) system, the latter with focus on
therapeutic applications have been recently developed to target the CD14/MD2/TLR4 pathway. Thus, we studied the effect of
complement activation and function.35 meconium on these two branches as well as the effect of specific
inhibition of both complement and CD14. Interestingly, we found
that inhibition of both pathways efficiently attenuated the
MAS and the complement system inflammatory reaction induced by meconium (B Salvesen et al.,
In the first study of a possible interaction of complement and MAS manuscript submitted).
it was documented that meconium in vitro is a highly potent
activator of the complement system.36 Adding meconium to human
umbilical serum induced a substantial increase in TCC formation, Addendum
which was found to be mediated mainly through the alternative To the best of our knowledge we are the first and so far the only
activation pathway. As judged by their relative amounts in whole group that has studied the interaction between complement and
meconium, the lipid and water fractions contributed equally to this MAS. How comes that these studies were initiated? The answer is
activation. However, corrected for the lower amount of the lipid simple: an incidental meeting in a PhD dissertation a few years ago
fractions compared with the water fractions in normal meconium, by two scientists (first and last authors of this paper), one an expert
the lipid fraction per weight was more potent. on complement and the other on MASFboth of them at that time
In a whole-blood model established for studying the interaction virtually devoid of knowledge on the other’s research fieldFled to
of the complement system with the inflammatory network,37 we an interesting discussion. First author asked: ‘what is at present
studied the role of complement in meconium-induced leukocyte your main research’? After getting the answer he replied ‘what is
activation as measured by oxidative burst and expression of the really MAS’? Having got a brief introduction to MAS, the last author
surface activation markers CD11b and L-selectin.38 Meconium asked: ‘but what is your main focus’? After answering, last author
induced a marked oxidative burst that was reduced by 60 to 70% replied: ‘but what is actually complement’? Two completely
when adding the alternative complement pathway inhibitor different scientific worlds met and recognized that we had to
anti-factor D. Furthermore, meconium induced a substantial proceed on a novel trackFdoes meconium as such activate the
increase in CD11b and decrease in L-selectin, typical for leukocyte complement system and is complement activated in experimental
activation, which was virtually abolished (95 to 100%) by adding MAS? As there were no reports of this topic in the literature we
anti-factor D. Importantly, inhibition of complement using a initiated studies by mutually utilizing well-established models to
specific C5aR antagonist gave the same inhibition of both oxidative study complement activation and MAS, respectively. Lessons to
burst and expression of CD11b and L-selectin as anti-factor D, learn are that scientific groups to a greater extent should discuss
implying that C5a was solely responsible for the possible interacting mechanisms of pathophysiology when they
meconium-induced complement-dependent leukocyte activation. meet, and we should all accept that homeostasis in human biology
On the basis of these in vitro experiments we proceeded with is more complex than seen just from our own window,
in vivo studies using a well-established piglet model of MAS.39 Here consequently leading to collaboration to obtain scientific progress.

Journal of Perinatology
Complement and meconium
TE Mollnes et al
S119

Disclosure 21 Li YH, Yan ZQ, Brauner A, Tullus K. Meconium induces expression of inducible NO
The authors have declared no financial interests. synthase and activation of NF-kappaB in rat alveolar macrophages. Pediatr Res 2001;
49: 820–825.
22 Holopainen R, Aho H, Laine J, Peuravuori H, Soukka H, Kaapa P. Human meconium
has high phospholipase A2 activity and induces cellular injury and apoptosis in piglet
References lungs. Pediatr Res 1999; 46: 626–632.
1 Tran N, Lowe C, Sivieri EM, Shaffer TH. Sequential effects of acute meconium 23 Schrama AJ, de Beaufort AJ, Sukul YR, Jansen SM, Poorthuis BJ, Berger HM.
obstruction on pulmonary function. Pediatr Res 1980; 14: 34–38. Phospholipase A2 is present in meconium and inhibits the activity of pulmonary
2 Clark DA, Nieman GF, Thompson JE, Paskanik AM, Rokhar JE, Bredenberg CE. surfactant: an in vitro study. Acta Paediatr 2001; 90: 412–416.
Surfactant displacement by meconium free fatty acids: an alternative explanation for 24 Korhonen K, Soukka H, Halkola L, Peuravuori H, Aho H, Pulkki K et al. Meconium
atelectasis in meconium aspiration syndrome. J Pediatr 1987; 110: 765–770. induces only localized inflammatory lung injury in piglets. Pediatr Res 2003; 54:
3 Holopainen R, Soukka H, Halkola L, Kaapa P. Meconium aspiration induces a 192–197.
concentration-dependent pulmonary hypertensive response in newborn piglets. Pediatr 25 Kytola J, Kaapa P, Uotila P. Meconium aspiration stimulates cyclooxygenase-2
Pulmonol 1998; 25: 107–113. and nitric oxide synthase-2 expression in rat lungs. Pediatr Res 2003; 53:
4 Murphy JD, Vawter GF, Reid LM. Pulmonary vascular disease in fatal meconium 731–736.
aspiration. J Pediatr 1984; 104: 758–762. 26 Khan AM, Lally KP, Larsen GL, Colasurdo GN. Enhanced release of thromboxane A(2)
5 Perlman EJ, Moore GW, Hutchins GM. The pulmonary vasculature in meconium after exposure of human airway epithelial cells to meconium. Pediatr Pulmonol 2002;
aspiration. Hum Pathol 1989; 20: 701–706. 33: 111–116.
6 Aaltonen M, Soukka H, Halkola L, Jalonen J, Holopainen IE, Kero P et al. Asphyxia 27 Lukkarinen H, Laine J, Lehtonen J, Zagariya A, Vidyasagar D, Aho H et al. Angiotensin
aggravates systemic hypotension but not pulmonary hypertension in piglets with II receptor blockade inhibits pneumocyte apoptosis in experimental meconium
meconium aspiration. Pediatr Res 2003; 53: 473–478. aspiration. Pediatr Res 2004; 55: 326–333.
7 Soukka H, Rautanen M, Halkola L, Kero P, Kaapa P. Meconium aspiration induces 28 Zagariya A, Bhat R, Uhal B, Navale S, Freidine M, Vidyasagar D. Cell death and lung
ARDS-like pulmonary response in lungs of ten-week-old pigs. Pediatr Pulmonol 1997; cell histology in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; 159:
23: 205–211. 819–826.
8 Ghidini A, Spong CY. Severe meconium aspiration syndrome is not caused by 29 Kojima T, Hattori K, Fujiwara T, Sasai-Takedatsu M, Kobayashi Y. Meconium-induced
aspiration of meconium. Am J Obstet Gynecol 2001; 185: 931–938. lung injury mediated by activation of alveolar macrophages. Life Sci 1994; 54:
9 Katz VL, Bowes Jr WA. Meconium aspiration syndrome: reflections on a murky subject. 1559–1562.
Am J Obstet Gynecol 1992; 166: 171–183. 30 Clark P, Duff P. Inhibition of neutrophil oxidative burst and phagocytosis by
10 Cleary GM, Wiswell TE. Meconium-stained amniotic fluid and the meconium meconium. Am J Obstet Gynecol 1995; 173: 1301–1305.
aspiration syndrome. An update. Pediatr Clin North Am 1998; 45: 511–529. 31 Soukka HR, Ahotupa M, Ruutu M, Kaapa PO. Meconium stimulates neutrophil
11 Tyler DC, Murphy J, Cheney FW. Mechanical and chemical damage to lung tissue oxidative burst. Am J Perinatol 2002; 19: 279–284.
caused by meconium aspiration. Pediatrics 1978; 62: 454–459. 32 Mollnes TE, Song WC, Lambris JD. Complement in inflammatory tissue damage and
12 Davey AM, Becker JD, Davis JM. Meconium aspiration syndrome: physiological and disease. Trends Immunol 2002; 23: 61–64.
inflammatory changes in a newborn piglet model. Pediatr Pulmonol 1993; 16: 101–108. 33 Mollnes TE, Redl H, Hogasen K, Bengtsson A, Garred P, Speilberg L et al. Complement
13 Lally KP, Mehall JR, Xue H, Thompson J. Meconium stimulates a pro-inflammatory activation in septic baboons detected by neoepitope-specific assays for C3b/iC3b/C3c,
response in peritoneal macrophages: implications for meconium peritonitis. J Pediatr C5a and the terminal C5b-9 complement complex (TCC). Clin Exp Immunol 1993;
Surg 1999; 34: 214–217. 91: 295–300.
14 Zagariya A, Bhat R, Navale S, Vidyasagar D. Cytokine expression in meconium-induced 34 Brandtzaeg P, Mollnes TE, Kierulf P. Complement activation and endotoxin levels in
lungs. Indian J Pediatr 2004; 71: 195–201. systemic meningococcal disease. J Infect Dis 1989; 160: 58–65.
15 Korhonen K, Kiuru A, Svedstrom E, Kaapa P. Pentoxifylline reduces regional 35 Mollnes TE, Kirschfink M. Strategies of therapeutic complement inhibition. Mol
inflammatory and ventilatory disturbances in meconium-exposed piglet lungs. Immunol 2006; 43: 107–121.
Pediatr Res 2004; 56: 901–906. 36 Castellheim A, Lindenskov PH, Pharo A, Fung M, Saugstad OD, Mollnes TE. Meconium
16 Tollofsrud PA, Medbo S, Solas AB, Robertson B, Speer CP, Seidenspinner S et al. is a potent activator of complement in human serum and in piglets. Pediatr Res 2004;
Intratracheal albumin reduces IL-8 in tracheobronchial aspirates in piglets after 55: 310–318.
meconium aspiration. J Perinat Med 2004; 32: 78–83. 37 Mollnes TE, Brekke OL, Fung M, Fure H, Christiansen D, Bergseth G et al. Essential
17 Lindenskov PH, Castellheim A, Aamodt G, Saugstad OD. Meconium induced IL-8 role of the C5a receptor in E coli-induced oxidative burst and phagocytosis revealed by
production and intratracheal albumin alleviated lung injury in newborn pigs. Pediatr a novel lepirudin-based human whole blood model of inflammation. Blood 2002; 100:
Res 2005; 57: 371–377. 1869–1877.
18 Khan AM, Elidemir O, Epstein CE, Lally KP, Xue H, Blackburn M et al. Meconium 38 Castellheim A, Pharo A, Fung M, Saugstad OD, Mollnes TE. Complement C5a is
aspiration produces airway hyperresponsiveness and eosinophilic inflammation in a a key mediator of meconium-induced neutrophil activation. Pediatr Res 2004; 57:
murine model. Am J Physiol Lung Cell Mol Physiol 2002; 283: L785–L790. 242–247.
19 de Beaufort AJ, Pelikan DM, Elferink JG, Berger HM. Effect of interleukin 8 in 39 Lindenskov PH, Castellheim A, Aamodt G, Saugstad OD, Mollnes TE. Complement
meconium on in-vitro neutrophil chemotaxis. Lancet 1998; 352: 102–105. activation reflects severity of meconium aspiration syndrome in newborn pigs. Pediatr
20 de Beaufort AJ, Bakker AC, van Tol MJ, Poorthuis BJ, Schrama AJ, Berger HM. Res 2004; 56: 810–817.
Meconium is a source of pro-inflammatory substances and can induce 40 Castellheim A, Lindenskov PH, Pharo A, Aamodt G, Saugstad OD, Mollnes TE.
cytokine production in cultured A549 epithelial cells. Pediatr Res 2003; 54: Meconium aspiration syndrome induces complement-associated systemic inflammatory
491–495. response in newborn piglets. Scand J Immunol 2005; 61: 217–225.

Journal of Perinatology
Journal of Perinatology (2008) 28, S120–S122
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Phospholipase A2 in meconium-induced lung injury
P Kääpä and H Soukka
Department of Pediatrics, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland

PLA2 may occur as cytosolic (cPLA2) and secretory (sPLA2)


Although the triggering mechanisms of tissue inflammation and injury in types, increasing attention has been paid to the role of secretory
meconium-contaminated lungs are still unclear, there is increasing PLA2 as an acute phase protein in inflammatory conditions.7
evidence to suggest a central role for phospholipase A2’s (PLA2). In fact, In this paper, the present understanding of the role of secretory
elevated PLA2 activities together with high enzyme concentrations, especially PLA2 in acute lung injury, specifically when induced by meconium
the amount of pancreatic (group I) secretory PLA2 (PLA2-I), have been aspiration, is delineated. A new line of basic and clinical
detected in human meconium and in meconium-contaminated lungs. research of MAS, namely the possible contribution of meconium-
Recent data from our laboratory further indicate that human pancreatic introduced secretory PLA2 to systemic inflammation, is
PLA2, introduced in high amounts within aspirated particulate meconium, also highlighted.
is a potent inducer of lung tissue inflammatory injury. Our finding of
elevated human PLA2-I concentrations in plasma during the first hours
after intratracheal meconium administration in newborn piglets further
suggests that intrapulmonary aspiration of meconium could also have
PLA2 and acute lung injury
systemic inflammatory and injurious effects. This, however, remains to be
studied in further detail. Several lines of clinical and experimental evidence implicate that
Journal of Perinatology (2008) 28, S120–S122; doi:10.1038/jp.2008.147 excess pulmonary PLA2 activity, through generation of potent
vasoactive and proinflammatory lysophospholipids and
eicosanoids,5,6 may contribute to the progression of various
inflammatory lung disorders.5–9 In agreement, high blood and
pulmonary enzyme activity, correlating with the disease severity,
has been found in acute respiratory distress in adults.10 Although
Aspiration of meconium is known to initiate a complex cascade of high PLA2 activity in inflammatory lung injuries is commonly
pulmonary processes, resulting in progressively increasing connected with increased release of group II secretory PLA2
pulmonary vascular tone and a rapidly developing inflammatory (PLA2-II) from macrophages and platelets,6,7,11 group I PLA2
tissue injury with concomitant surfactant dysfunction in the (PLA2-I), secreted by the pancreas, is also known to be expressed
affected lungs.1,2 Although the critical role of pulmonary and to modulate cellular function in human lungs.12 Mammalian
inflammation in the course of neonatal meconium aspiration PLA2-I is in fact able to inactivate pulmonary surfactant concentra-
syndrome (MAS) has been emphasized, the triggering tion-dependently through hydrolysis of phosphatidylcholine.13
mechanisms and the manifold interplay of the injurious mediators PLA2-I may additionally, independent of the enzyme catalytic
in MAS are still poorly identified. There is yet evidence that activity, stimulate through specific membrane receptor action
human meconium may induce production of proinflammatory neutrophil function and cytokine and eicosanoid production from
agents within the lungs, and that some proinflammatory pulmonary cells.7,14 Moreover, experimental investigations have
components of meconium itself, such as cytokines and shown that exogenously administered PLA2-I may induce, most
phospholipase A2 (PLA2), may additionally contribute to intense likely through increased pulmonary thromboxane A2 synthesis,
inflammation and injury in the meconium-contaminated lung receptor-mediated contractile responses in the airways.15 It is
tissue.3,4 PLA2 represents a family of ubiquitous enzymes that therefore apparent that PLA2 activity associated with upregulation
through cleavage of membrane phospholipids release arachidonic of PLA2-I may be important in the pathogenesis of acute
acid, the precursor of proinflammatory eicosanoid.5,6 Although inflammatory lung injury. Still, variations in the tissue expressions
of different PLA2 enzyme types in the insulted lungs may also have
Correspondence: Dr P Kääpä, Department of Pediatrics and Research Centre of Applied and
consequences in the clinical course and therapeutic approaches
Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, Turku,
FIN 20520, Finland. (for example, use of specific PLA2 inhibitors) of acute lung injuries
E-mail: pekka.kaapa@utu.fi from various origin.6,7
PLA2 in meconium-injured lungs
P Kääpä and H Soukka
S121

The role of PLA2 in meconium-induced lung injury


Earlier studies in our laboratory have demonstrated that human
meconium has very high catalytic PLA2 activity, mainly (>90%)
due to high concentration of human pancreatic PLA2-I.4 In line
with this finding, tissue PLA2 activity and concentration of human
pancreatic PLA2 in the meconium-contaminated lungs are
concentration-dependently elevated and correlate directly with the
severity of the meconium-induced lung injury.4,16,17 In these
studies, promotion of lung edema formation and neutrophil influx
through insufflated human PLA2-I, either in soluble form or within
meconium, further substantiates the central role of pulmonary
PLA2-I in the development of lung injury after meconium Figure 1 Serum human pancreatic phospholipase A2 (PLA2) concentrations at
baseline (BL) and during 6 h after meconium aspiration in newborn piglets
aspiration.16,17 High PLA2 activity in meconium, mainly due to
(n ¼ 5). Mean (s.d.); *P<0.05 vs BL.
secretory PLA2-I, is additionally shown to be associated with
decreased surfactant biophysical activity in the affected lungs.18
This inactivation could be explained by enzymatic hydrolysis of
dipalmitoyl-phosphatidylcholine, the major lipid constituent of in extrapulmonary organs.23 Intrapulmonary meconium exposure
surfactant, but also the produced lyso-phosphatidylcholine may is indeed demonstrated to result in systemic complement
have an inhibitory role.19 Although some experimental data activation, cytokine release, and activation of circulating
suggest that type I secretory PLA2 may induce type II PLA2 neutrophils with production of a variety of biologically active
expression,20 challenge of the lungs with high pancreatic PLA2 mediators, including reactive oxygen radicals.1,23 In line with these
activity within aspirated meconium does not seem to influence findings, our recent studies indicate that pancreatic PLA2,
pulmonary PLA2-II production.16,17 It is thus conceivable that contained in high amount in meconium and thereby introduced
exposure of the lungs meconium with high pancreatic PLA2 into the lungs, may be absorbed, at least to some extent, into the
activity, specifically through aspiration of thick particulate pulmonary circulation.4,17 In fact, we were able to demonstrate
meconium, may markedly participate in propagation of the that human pancreatic PLA2 concentrations in plasma are elevated
ensuing pulmonary failure.17 Yet, the possible involvement of other during the first hours after intratracheal human meconium
phospholipases in the development of meconium-induced lung administration in newborn piglets (Figure 1). Similar PLA2
injury remains to be investigated. activation and circulatory release have been recognized in clinical
Although the above considerations may potentially offer new disorders that promote systemic inflammation.24 Alike, markedly
ways to treat infants with MAS, most of the current therapies of elevated lung and blood PLA2 activity, correlating with the disease
MAS are symptomatic, and their clinical effects have been severity, has been found in adults with acute respiratory distress.10
unsatisfactory and often conflicting.1 Accordingly, our earlier data The systemic inflammatory reaction with systemic release of
indicate that early administration of dexamethasone, known to mediators, such as PLA2 and cytokines, is supposed to ultimately
inhibit stimulated PLA2 synthesis,21 does not reduce lung PLA2 lead to extrapulmonary organ dysfunction and injury.24 Recent
activity or inflammation in experimental meconium aspiration.22 experimental data from our laboratory in fact indicate that severe
Alike, some of our preliminary data show that mepacrine, an meconium aspiration itself, without any complicating incidents,
unspecific PLA2 inhibitor, does not decrease PLA2 activity or prevent may result in brain tissue injury, specifically in the hippocampus.25
inflammatory injury, but rather tends to elevate tissue enzyme Nevertheless, it still remains undetermined how often and at what
activity in the meconium-contaminated lungs (P. Kääpä, intensity systemic inflammation occurs in connection with
unpublished observation). Clearly, more investigations are needed meconium aspiration and what is its significance for the outcome
before the pathophysiological relevance of PLA2 in neonatal MAS is of infants with severe MAS.
revealed.
Conclusions
Taken together, there is a bulk of evidence indicating that
The role of PLA2 in systemic inflammatory reaction intrapulmonary aspirated meconium, specifically in thick
after meconium aspiration particulate form, challenges the lungs with high human pancreatic
Recent investigations have suggested that the inflammatory injury PLA2 concentration and activity, and may thereby contribute to
processes found in MAS may not be restricted to the lungs, but may pulmonary inflammatory perturbations, systemic inflammatory
also result in systemic inflammatory reaction and possible sequelae reactions and eventually distant organ damage. These findings

Journal of Perinatology
PLA2 in meconium-injured lungs
P Kääpä and H Soukka
S122

may be amenable to development of new modes of more specific phospholipase A2 and inhibited by a direct surfactant protein A-phospholipase A2
therapeutic approaches. protein interaction. J Clin Invest 1998; 102: 1152–1160.
12 Seilhamer JJ, Randall TL, Yamanaka M, Johnson LK. Pancreatic phospholipase A2:
isolation of the human gene and cDNA from porcine pancreas and human lung. DNA
1986; 5: 519–527.
Acknowledgments
13 Hite RD, Seeds MC, Jacinto RB, Balasubramanian R, Waite M, Bass D. Hydrolysis of
This study was supported by the Foundation for Pediatric Research, the Sigrid surfactant-associated phosphatidylcholine by mammalian secretory phospholipase A2.
Juselius Foundation, and the Paulo Foundation, Finland. Am J Physiol Lung Cell Mol Physiol 1998; 275: L740–L747.
14 Granata F, Petraroli A, Boilard E, Bezzine S, Bollinger J, Del Vecchio L et al. Activation
of cytokine production by secreted phospholipase A2 in human lung macrophages
Disclosure expressing the M-type receptor. J Immunol 2005; 174: 464–474.
The authors have declared no financial interests. 15 Sommers CD, Bobbitt JL, Bemis KG, Snyder DW. Porcine pancreatic phospholipase
A2-induced contractions of guinea pig lung pleural strips. Eur J Pharmacol 1992;
216: 87–96.
16 Korhonen K, Soukka H, Halkola L, Peuravuori H, Aho H, Pulkki K et al. Meconium
References induces only localized inflammatory lung injury in piglets. Pediatr Res 2003; 54(2):
1 Wiswell TE. Meconium staining and the meconium aspiration syndrome. In: 192–197.
Stevenson D, Benitz WE, Sunshine P (eds). Fetal and Neonatal Brain Injury. 17 Sippola T, Aho H, Peuravuori H, Lukkarinen H, Gunn J, Kaapa P. Pancreatic
Mechanisms, Management and the Risks of Practice. Cambridge University Press: phospholipase A2 contributes to lung injury in experimental meconium aspiration.
Cambridge, UK, 2003, pp 612–635. Pediatr Res 2006; 59(5): 641–645.
2 Davey AM, Becker JD, Davis JM. Meconium asiration syndrome: physiological and 18 Schrama AJJ, de Beaufort AJ, Sukul YRM, Jansen SM, Poorthuis BJHM, Berger HM.
inflammatory changes in a newborn piglet model. Pediatr Pulmonol 1993; 16: Phospholipase A2 is present in meconium and inhibits the activity of pulmonary
101–108. surfactant: an in vitro study. Acta Paediatr 2001; 90: 412–416.
3 de Beaufort AJ, Pelikan DMV, Elferink JGR, Berger HM. Effect of interleukin 8 in 19 Holm BA, Keicher L, Liu M, Sokolowski J, Enhorning G. Inhibition of pulmonary
meconium on in-vitro neutrophil chemotaxis. Lancet 1998; 352: 102–105. surfactant function by phospholipases. J Appl Physiol 1991; 71: 317–321.
4 Holopainen R, Aho H, Laine J, Peuravuori H, Soukka H, Kääpä P. Human meconium 20 Kishino J, Ohara O, Nomura K, Kramer RM, Arita H. Pancreatic-type phospholipase A2
has high phospholipase A2 activity and induces cellular injury and apoptosis in piglet induces Group II phospholipase A2 expression and prostaglandin biosynthesis in rat
lungs. Pediatr Res 1999; 46: 626–632. mesangial cells. J Biol Chem 1994; 269: 5092–5098.
5 Dennis EA. Diversity of group types, regulation, and function of phospholipase A2. 21 Hoeck WG, Ramesha CS, Chang DJ, Fan N, Heller RA. Cytoplasmic
J Biol Chem 1994; 269: 13057–13060. phopholipase A2 activity and gene expression are simulated by tumor necrosis
6 Valentin E, Ghomashchi F, Gelb MH, Lazdunski M, Lambeau G. On the diversity of factor: dexamethasone blocks the induced synthesis. Proc Natl Acad Sci USA 1993; 90:
secreted phospholipases A2. J Biol Chem 1999; 274: 31195–31202. 4475–4479.
7 Triggiani M, Granata F, Frattini A, Marone G. Activation of human inflammatory cells 22 Holopainen R, Laine J, Halkola L, Aho H, Kääpä P. Dexamethasone treatment
by secreted phospholipases A2. Biochim Biophys Acta 2006; 1761(11): 1289–1300. attenuates pulmonary injury in piglet meconium aspiration. Pediatr Res 2001;
8 Niewoehner DE, Rice K, Duane P, Sinha AA, Gebhard R, Wangensteen D. Induction of 49: 1–7.
alveolar epithelial injury by phospholipase A2. J Appl Physiol 1989; 66: 261–267. 23 Lindenskov PH, Castellheim A, Aamodt G, Saugstad OD, Mollnes TE. Complement
9 Vadas P, Pruzanski W. Role of secretory phospholipases A2 in the pathobiology of activation reflects severity of meconium aspiration syndrome in newborn pigs. Pediatr
disease. Lab Invest 1986; 55: 391–404. Res 2004; 56: 810–817.
10 Kim DK, Fukuda T, Thompson BT, Cockrill B, Hales C, Bonventre JV. Bronchoalveolar 24 Anderson BO, Moore EE, Banerjee A. Phospholipase A2 regulates critical inflammatory
lavage fluid phospholipase A2 activities are increased in human adult respiratory mediators of multiple organ failure. J Surg Res 1994; 56: 199–205.
distress syndrome. Am J Physiol 1995; 269: L109–L118. 25 Aaltonen M, Soukka H, Halkola L, Jalonen J, Holopainen IE, Kääpä PO. Meconium
11 Arbibe L, Koumanov K, Vial D, Rougeot C, Faure G, Havet N et al. Generation of aspiration induces oxidative injury in the hippocampus of newborn piglets. Early Hum
lyso-phospholipids from surfactant in acute lung injury is mediated by type-II Dev 2005; 81(5): 439–447.

Journal of Perinatology
Journal of Perinatology (2008) 28, S123–S126
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Meconium-induced release of nitric oxide in rabbit alveolar cells
R Fontanilla, A Zagariya and D Vidyasagar
Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago Medical Center, Chicago, IL, USA

third is inducible, Ca-independent NO (iNOS). It has been


Previous studies have shown meconium-induced lung injury occurs shown in the lungs of septic murine to have high levels of NO
throughout release of inflammatory cytokines. The exact mechanism of produced by iNOS.3–6 NO is also a known regulator of apoptosis
cytokine-induced apoptosis is not known. In this study we hypothesized that and has been shown to inhibit the activity of many caspases both
meconium-induced apoptosis in the lungs is mediated through the in vivo and in vitro conditions in endotoxin-induced lung injury.7
production of inducible nitric oxide (NO). We studied two groups of High levels of NO can react with superoxide to produce a highly
newborn rabbit pups: one group was instilled with meconium and other oxidative peroxynitrate. We have reported in an earlier study
with normal saline. We measured precursors of NO in lung lavage from apoptosis and necrosis occurring after meconium instillation in rabbit
both groups of rabbits and NO levels were calculated accordingly. The levels lungs using DNA staining and detection of DNA fragments by in situ
of NO and NO-derivatives increased significantly in both groups. However end labeling.8
NO expression in meconium group 2 h after meconium instillation was Nitric oxide is an important inflammatory mediator. In MAS, an
significantly higher than in saline-instilled group suggesting NO production intense inflammatory reaction occurs and Li et al. (2001) reported
plays a role in meconium-induced inflammation. an increase in rat alveolar macrophage cell line (ATCC8383) when
Journal of Perinatology (2008) 28, S123–S126; doi:10.1038/jp.2008.170 exposed to meconium. They also found iNOS expression leading to
increased production of NO.9,10 The purpose of the study is to
measure the NO production in meconium-induced injury in rabbit
Introduction alveolar cells.
Meconium aspiration syndrome (MAS) is a major cause of
morbidity and mortality in neonates. It is characterized by initial
obstruction of the airway resulting in ventilation–perfusion Hypothesis
mismatch, hypoxemia and increased vascular resistance. MAS Meconium-induced apoptosis in the lungs is mediated through the
effects 1 to 3% of both term and post-term infants. Earlier studies production of inducible NO. NO production is a major intermediary
have shown that MAS includes accumulation of inflammatory step in meconium-induced lung cell apoptosis.
cells, release of inflammatory cytokines and massive cell death in
experimental models. The pathogenesis is still unclear, although
the role of bioactive mediators such as nitric oxide (NO), Methodology
pro-inflammatory cytokines, prostaglandins, thromboxane, reactive Animal model (in vivo)
oxygen species has been proposed in sepsis-induced lung injury.1 The care and handling of the animals were in accordance with
Nitric oxide is a biological signaling and effector molecule National Institutes of Health guidelines. The Animal Care and USE
involved in many biologic functions including maintaining blood Committee of Michael Reese Hospital (Chicago, IL, USA) approved
pressure, modulating neural transmissions and host defense the experimental protocol. Pregnant rabbits were housed for 3 days
mechanisms.2 It is catalyzed by enzymes of the NO synthase (NOS) before the experiments with the mother in stainless steel rabbit
family. It is generated through the reaction of L-arginine, cages. Mothers were given regular Purina rabbit chow (Scientific
nicotinamide adenine dinucleotide phosphate oxidase and O2 to Animal Feed Co., Arlington Heights, IL, USA). After delivery, pups
NO and citrulline. Reactions of NO with oxygen in aqueous were used in the study.
solution produce a relatively unreactive nitrate and nitrite ion as Two groups, 2-week-old New Zealand white rabbit pups, were
an end product. Two of the three isoforms are constitutive and the used in the study. Ten rabbits per group: group 1, saline-instilled
rabbits and group 2, meconium-instilled rabbits. Tracheotomy was
Correspondence: Dr D Vidyasagar, Professor of Pediatrics, University of Illinois at Chicago
Medical Center, 840 South Wood Street, M/C 856, Chicago, IL 60612, USA. performed under sedation with intraperitoneal ketamine
E-mail: dsagar@uic.edu (10 mg kg 1) and xylazine (1 mg kg 1). Tracheal instillation of
Meconium-induced release of NO in alveolar cells
R Fontanilla et al
S124

10% meconium solution (1.2 ml kg 1) or 0.9% NaCl (1.2 ml kg 1) Result and analysis
was made. Skin incision was closed with a 4-0 nylon suture, and NO derivative (nitrite) production in meconium-instilled lungs was
rabbits were allowed to breathe spontaneously in the room air significantly high (P<0.05) at 2 h after meconium instillation
without assisted ventilation or supplemental oxygen. At set point compared with saline instillation. The levels of nitrite also
(0, 2, 4 and 8 h), the animals were euthanized by intraperitoneal increased at 4 and 8 h after meconium instillation, but these values
injection of Nembutal (100 mg kg 1) for studies. were not significantly different compared with saline-instilled
lungs. Nitrites in saline-instilled lungs show low levels at 2 h and
Meconium preparation increased levels at 4 and 8 h comparable with meconium-instilled
A total of 15 first-pass human meconium samples were obtained pups. This shows that meconium initiates lung injury faster than
from term, healthy neonates. Fresh newborn infant meconium of saline and that both meconium and saline induced NO production.
1 g was homogenized on ice in a blender with 9 ml of 0.9% NaCl to The NO is released in response to the external injury factors such
a 10% (w/v) final concentration and was spun down at 5000 r.p.m. as meconium or saline (Table 1 and Figure 1)
for 20 min (4 1C) to separate the supernatant and pellet. The NO is a biological signaling and effector molecule involved in
supernatant was filtered by a glass filter followed by sterilization by many biologic functions including maintaining blood pressure,
0.2 mm filter (both filters from Millipore Co., Bedford, MA, USA) modulating neural transmissions and host defense mechanisms. It
and was used for instillation in the lungs of 2-week-old newborn is a free radical, which makes it react very readily with superoxide
rabbit pups. to form peroxynitrite. Peroxynitrite is a highly oxidative species that
is capable of nitrating tyrosine residues of numerous proteins lead
Reagents to the production of nitrotyrosine.11 High levels of nitrotyrosine
ELISA kits were obtained from R&D System Co. (Minneapolis, MI, formation have been shown to be involved in acute lung injury by
USA). All other materials are obtained from different sources and way of increasing vascular permeability of endothelial cells,
were of reagent grade. inhibiting leukocyte adhesion, degrading carbohydrates, inhibiting
lipid peroxidation and cleaving DNA through nitration and
In vivo animal model experiments oxidation.11,12 The excess amount of NO required to form
The lungs were isolated and homogenized in saline and peroxynitrite to produce apoptosis is not known. What has been
centrifuged at 5000 r.p.m. for 5 min. The supernatant was filtered shown in this research is that within 2 h after instillation,
before measuring NO. Filtration eliminates nucleophiles, meconium or saline enhances the production of NO. Khan et al.10
antioxidants and compounds containing sulfhydryl groups such as
cysteine and glutathione, which may interfere with color formation
in Griess reaction. Table 1 NO production 2, 4 and 8 h after meconium instillation.

Studies of NO using ELISA Levels on NO after meconium of saline instillation


NO has a very short half-life (<10 s), which makes it difficult to 2 h* 4h 8h
measure directly. However, NO is metabolized to nitrate and nitrite.
Quantification of these anions can be used indirectly to determine Meconium 43.5±12.3 mmol l 1 52.5±5 mmol l 1 55.59±26.3 mmol l 1

the amount of NO originally present. There is a direct correlation Saline 28.24±9.8 mmol l 1 52.28±26.3 mmol l 1
48.9±12.7 mmol l 1

between the nitrate and nitrite levels and NO. Nitrates and nitrites *P<0.05.
were measured using a commercially available ELISA kit
(R&D System Co.). The assay is based on colorimetric detection
Meconium
of nitrite as an azo dye product of Griess reaction. The Griess
60 Saline
reaction is a two-step diazotization reaction in which acidified NO2
produces a nitrosating agent, which reacts with sulfanilic 50

acid to produce a diazonium ion. This ion is then coupled to 40


N-1-napthylethylenediamine to form the chromophoric 30
azo-derivative, which absorbs light between 540 and 570 nm
20
and is quantitated by spectrophotometer.
10

Data analysis 0
Data from the experiments were reported as the mean±s.d. Data 2h 4h 8h
were analyzed using analysis of variance and unpaired Student’s Figure 1 NO production at 2, 4, 8 h after meconium and saline instillation in
t-test. A P<0.05 was considered to be significant. rabbit lungs. Measured using ELISA technique.

Journal of Perinatology
Meconium-induced release of NO in alveolar cells
R Fontanilla et al
S125

Figure 2 Mechanism of meconium-induced lung injury.

showed the production of NO from monolayers of A549 cells meconium-induced apoptosis. Apoptosis is induced by two main
immediately after incubating the cells with meconium. pathway: (1) mitochondrial pathway, which involves cytochrome c
The instillation of meconium or saline itself does not possess release from the mitochondria, and the activation of caspase-9,
any significant neutrophil chemotaxis13 but could stimulate the which then cleaves and activates caspases 3, 6 and 7; and (2) the
production of the pro-inflammatory cytokine and upregulate the death domain receptor pathway that involves the activation of Fas
production of the inducible form of iNOS, which produces large or tumor necrosis factor receptors followed by the activation of
amount of NO.4,14 –16 iNOS is produced in many cells including caspase-8 and subsequent activation of other caspases.5,7 The
alveolar cells in response to infection, inflammation and other long-lasting overproduction of NO acts as a modulator of apoptosis,
circumstances when the immune system is activated. The other activating the caspase family through the release of cytochrome c
form is neuronal NOS from nerve cells and endothelial cells.2 The into the cytosol.20,21 Once in the cytosol, cytochrome c interacts
inflammatory damage in this research marked by increased levels with Apaf-1 (apoptotic protease-activating factor 1), leading to the
of NO lasted for 8 h. A study carried out Holopainen et al.,17 in the activation of caspase-9 and subsequent caspases, thereby leading to
ultrastructural analysis of porcine lungs, 6 h after instillation of apoptosis. The activated caspases lead to cleavage of the nuclear
meconium, showed a significant amount of alveolar exudates and lamin and breakdown of the nucleus through caspase-3.22 In our
edematous alveolar epithelium and endothelium. The increase in study, the meconium-induced NO production may represent a key
the production of NO at 4 and 8 h could have been due to the mechanism for the apoptosis associated with meconium aspiration
worsening inflammatory process and increasing apoptosis in the syndrome.
alveolar cells. This inflammatory reaction causes an increased Despite the significant advances in management and diagnosis,
pulmonary vascular permeability producing the exudates and MAS still causes an important morbidity and mortality in neonates.
eventually inactivation of the surfactant and decreased pulmonary Earlier studies by Holopainen et al.17 showed that NO inhalation
compliance.16,18 Earlier studies also showed in vitro, those controls the rise in the pulmonary artery pressure, improves
neutrophils and plasma proteins in the alveoli inactivate arterial oxygenation and prevents further increase in epithelial
surfactant.13 This may also explain why meconium can inactivate apoptosis, but does not protect against early inflammatory damage
a surfactant. Meconium enhancing the production of NO and the caused by meconium. Steroids have been used to inhibit the
inflammatory reactions may play an important part in the inflammatory response and can downregulate the
pathogenesis of MAS. pro-inflammatory cytokine production in vitro. Li et al.9 have
Meconium aspiration may also cause apoptosis in alveolar cells. shown that steroids also inhibits meconium-stimulated NO
Apoptosis is characterized by loss of cell function and decrease in production and suggested that steroids could be used in the
cell size and its morphology.19 Zagariya et al.8 showed apoptosis in treatment of MAS.
the airway epithelium 8 h after meconium instillation using In conclusion, our findings show that meconium is a potent
ISEL-DNA end labeling. The apoptosis reported from an earlier inducer of NO production in the alveolar and airway cells
study coincide with an increase in the production of NO (Figure 2), resulting in inflammation and apoptosis, which could
in this study up to 8 h. This might play a crucial role in result in MAS.

Journal of Perinatology
Meconium-induced release of NO in alveolar cells
R Fontanilla et al
S126

Significance of the research 9 Li YH, Yan ZQ, Braunner A, Tullus K. Meconium induces expression of inducible NO
synthase and activation of NF-(kappa)B in rat alveolar macrophages. Pediatr Res
This study might be essential in understanding the role of
2001; 49: 820–825.
endogenous NO in the pathophysiology of MAS and might represent 10 Khan AM, Lally KP, Elidemir O, Colosurdo GN. Meconium enhances the release of
a key point in future therapeutic application. nitric oxide in human epithelial cells. Biol Neonate 2002; 81: 99–104.
11 Kooy NW, Royall JA, Ye YZ, Kelly DR, Beckmann JS. Evidence for in vivo peroxynitrate
production in human lung injury. Am J Respir Crit Care Med 1995; 151:
Disclosure 1250–1254.
The authors have declared no financial interests. 12 Haddad IY, Pataki G, Hu P, Galliani C, Beckman JS, Matalon S. Quantitation of
nitrotyrosine levels in lung sections of patients and animals with acute lung injury.
J Clin Invest 1994; 94: 2407–2413.
13 Davey AM, Becker JO, Davis JM. Meconium aspiration syndrome: physiological and
References inflammatory change in newborn piglet model. Pediat Pulmonol 1993; 16: 101–108.
1 Hibbs JB, Taintor RR, Vavrin Z. Nitric oxide: a cytotoxic activated macrophage effector 14 Speyer CL, Neff TA, Warner RL, Guo RF, Sarna JV, Riedemann NC et al. Regulatory
molecule. Biochem Biophys Res Commun 1987; 157: 87–94. effects of iNOS an acute lung inflammatory response in mice. AM J Pathol 2003; 163:
2 Grisham MB, Jour d’Heuil D, Wink OA. Nitric oxide I. Physiological chemistry of nitric 2319–2328.
oxide and its metabolites: implication in inflammation. Am J Physiol 1999; 276: 15 Zagariya A, Bhat R, Uhal B, Chari G, Vidyasagar D. Apoptosis of airway epithelial cells
G315–G321. in response to meconium. Life Sci 2005; 76: 1849–1858.
3 Kristoff AS, Goldrberg P, Laubauch V, Hussan SN. Role of inducible nitric oxide 16 Wiswell TE, Bent RC. Meconium staining and MAS. Pediatr Clin North Am 1993; 40:
synthase in endotoxin acute lung injury. Am J Respir Crit Care Med 1998; 158: 955–981.
1883–1889. 17 Holopainen R, Aho H, Laine J, Halkola J, Kaapa P. Nitric oxide inhalation inhibits
4 Mehta S. The effects of nitric oxide in acute lung injury. Vasc Pharmacol 2005; 43: pulmonary apoptosis but not inflammatory injury in porcine meconium aspiration.
390–403. Acta Paediatr 1999; 88: 1147–1155.
5 Rudkowski J, Barreiro E, Harfouche R, Goldberg P, Kishta O, D’orleans-Juste P et al. 18 Tyler DC, Murphy J, Cheng FW. Mechanical and chemical damage to lung tissue
Role of iNOS and nNOS in sepsis induced pulmonary apoptosis. Am J Physiol Lung caused by meconium aspiration. Pediatrics 1978; 62: 454–457.
Cell Mol Physiol 2004; 286: L793–L800. 19 Arends MJ, Weyllie AN. Apoptosis: mechanism and roles in pathology. Int Rev Exp
6 Farley KS, Wang LF, Rasavi HM, Law C, Rohan M, McCormack DG et al. Effects of Pathol 1991; 68: 251–306.
macrophage inducible nitric oxide synthase in murine septic lung injury. Am J Physiol 20 Choi BM, Pae HO, Jan SI, Kim YM, Chung HT. Nitric oxide as pro-apoptotic as well as
Lung Cell Mol Physiol 2006; 290: L1164–L1172. anti-apoptotic modulator. J Biochem Mol Biol 2002; 35: 116–126.
7 Chung HT, Pae HO, Choi BM, Billiar TR, Keim YM. Nitric oxide as a bioregulator of 21 Boyd CS, Cadenas E. Nitric oxide and cell signaling pathways in
apoptosis. Biochem Biophys Res Commun 2001; 282: 1075–1079. mitochondrial-dependent apoptosis. Biol Chem 2002; 383: 411–423.
8 Zagariya A, Bhat R, Uhal B, Navale S, Vidyasagar D. Cell death and lung cell histology 22 Pandya N, Jain S, Santani D. Apoptosis: a friend or a foe? Internet J Pharmacol ISSN
in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; 159: 819–826. 1993; 2: 1531–2976.

Journal of Perinatology
Journal of Perinatology (2008) 28, S127–S135
r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30
www.nature.com/jp

REVIEW
Intracellular and extracellular serpins modulate lung disease
DJ Askew and GA Silverman
UPMC Newborn Medicine Program, Children’s Hospital of Pittsburgh and Magee-Womens Research Institute, University of Pittsburgh
School of Medicine, Pittsburgh, PA, USA

inhibition occurs at a 1:1 stoichiometry. Importantly, peptidase


An imbalance between peptidases and their inhibitors leads to pulmonary inhibition by serpins is irreversible. The purpose of this review
disease. Imbalances occur in the adult and the neonate at risk for a specific is to describe the general mechanism and regulation of serpin
set of lung pathologies. Serpins (serine peptidase inhibitors) make up the function in the lung. Critical roles in the normal and
major source of antipeptidase activity in the lung. The purpose of this pathological lung, and serpins as potential therapeutic agents are
review is to describe the serpin mechanism of inhibition, their roles in the examined.
normal and pathological lung and their potential as therapeutic agents. Serpins are found in animals, plants, prokaryotes and viruses.
Journal of Perinatology (2008) 28, S127–S135; doi:10.1038/jp.2008.150 They are distinguished from other peptidase inhibitors because
their unique suicide substrate-like mechanism of inhibition
(reviewed in Gettins1,2). A total of 37 serpin genes in humans are
distributed within 9 clades (A–I). Clade A has 13 members
including SERPINA1 (a1-antitrypsin, a1AT) and SERPINA5
(protein C inhibitor, PCI) that are secreted into the circulation.
Introduction Clade B is made up of 13 serpins that are primarily intracellular.
The lung functions in the face of many physical challenges: The remaining eleven serpins are dispersed across clades C–I and
exposure to oxygen and environmental toxins, airborn pathogens, are secreted into the fluid phase. The tertiary structure of serpins is
continuous expansion and compression while breathing and highly conserved and consists of three b-sheets (A–B), 7 to 9
maintenance of a delicate interface enabling gas exchange with the a-helices (A–I) and extended reactive site loop (RSL) that acts as
body’s vascular system. As a result, tissue damage, inflammation, the bait for peptidase targets. The structure is critical to the unique
repair and remodeling are constant. These processes, from the suicide-substrate-like mechanism of peptidase inhibition.3
induction of apoptosis and necrosis in acute injury to the defense Inhibitory serpins exist in a metastable state resembling a loaded
mechanisms of inflammatory cells, coagulation and fibrinolysis, to mousetrap. Upon peptidase binding to the RSL, hydrolysis of the
extracellular matrix degradation and cell migration, are all P1–P10 peptide bond releases the RSL and allows the serpin to
peptidase driven. Peptidase inhibitors are required to regulate these undergo a rapid conformational transition. With the peptidase
processes and neutralize peptidases upon completion of their covalently bound to the P1 residue, the RSL is inserted into b-sheet
intended roles. Most pulmonary diseases are associated with an A as strand 4 and the peptidase is trapped in an inactive complex
imbalance between peptidase and peptidase inhibitor activity with the serpin.
(Table 1). This holds true in the neonate at risk for a specific set of This mechanism of inhibition has a side effect resulting in
dysfunctional lung pathologies, including meconium aspiration clinical diseases collectively referred to as serpinopathies.4,5
syndrome (MAS), respiratory distress syndrome (RDS) and Mutations that cause structural instability, specifically the opening
bronchopulmonary dysplasia (BPD). The balance between of b-sheet A, can lead to the formation of serpin polymers. Under
peptidase and antipeptidase activities appears critical in the conditions of high serpin concentration, such as the endoplasmic
development and progression of these diseases of premature and reticulum of cells in the liver, the RSL of one molecule is inserted
full-term newborns. Serpins (serine peptidase inhibitors) make up into b-sheet A of another. The prototype of this disease mechanism
the major source of peptidase inhibitors in the lung. Others is the genetic deficiency of a1AT.6 The two most common alleles,
peptidase inhibitors include Kunitz, Kazal and Bowman-Birk a1AT*S and a1AT*Z, result in formation of a1AT polymers in the
protein families. Unlike the other peptidase inhibitors, serpin liver and significantly diminished a1AT plasma levels. The
consequence of this is twofold for the individual: hepatocyte cell
Correspondence: Dr DJ Askew, Magee-Womens Research Institute, 204 Craft Avenue,
Room 210, Pittsburgh, PA 15213, USA.
death leading to cirrhosis,7 and predisposition to emphysema,
E-mail: rsidja@mwri.magee.edu asthma and additional respiratory diseases.8 Other serpins with
Serpins in pulmonary disease
DJ Askew and GA Silverman
S128

naturally occurring alleles shown to develop polymers and Infiltrating neutrophils express catG on the cell surface and release
subsequent serpinopathies are C1 inhibitor, antithrombin, peptidases including NE into the airway space. The primary role of
a1-antichymotrypsin (ACT), heparin cofactor II and neuroserpin. a1AT is to maintain a local balance between peptidase activities
required for inflammatory cell function and to protect the lung
against peptidase-mediated tissue damage.10
a1AT is the major source of protection against a1AT deficiency is one of the most common inherited defects in
proteolytic damage in the lung Caucasians. Mutant alleles a1AT*S (Glu264Val) and a1AT*Z
a1AT (SERPINA1) is the major peptidase inhibitor in plasma and (Glu342Lys) account for most a1AT deficiencies, with a1AT*Z the
provides the main source of antipeptidase activity in lung. a1AT is more deleterious. Approximately 4% of northern Europeans carry
a 52 kDa glycoprotein produced in and secreted from the liver, as the Z allele and approximately 1 in 2000 is homozygous
well as bronchial epithelial cells (BECs). Amino-acid residues (a1AT*ZZ) whereas 1 in 1000 is heterozygous for the two mutant
Met-Ser at the P1–P10 position within the RSL make a1AT alleles (a1AT*SZ). Individuals homozygous for the more common
a potent inhibitor of neutrophil elastase (NE), cathepsin G (catG) S allele (a1AT*SS) exhibit an B40% decrease in a1AT plasma
and proteinase-3 (Table 1).9 In the lung, NE is capable of causing levels. The a1AT*ZZ homozygous genotype results in a deficit of
extensive damage because of its proteolytic activity against B85%. Individuals with this phenotype, or heterozygous for the
structural components collagen and elastin. During inflammation two mutant alleles, are at risk for developing diseases associated
large amounts of this peptidase are delivered to the lung. with excess elastase activity such as emphysema.

Table 1 Serpins and their proposed functions in the lung

Serpin Peptidase targets Function Pulmonary diseases

SERPINA1 Neutrophil elastase, cathepsin G, proteinase-3 Protect the lung against elastase activity Empysema
a1-antitrypsin Chronic pulmonary obstructive
disease
SERPINA5 Activated protein C, thrombin-thrombomodulin Activate coagulation
Peptidase C
inhibitor
Thrombin, factors Xa, XIa Suppress coagulation
Urokinse-type plasminogen activator, tissue-type Suppress fibrinolysis
plasminogen activator

SERPINC1 Thrombin, factors IXa, Xa, XIa, XIIa, kallikrein Suppress coagulation Sepsis/ALI
Antithrombin III

SERPINE1 Urokinse-type plasminogen activator, tissue-type Suppress fibrinolysis ALI


Plasminogen plasminogen activator Idiopathic pulmonary fibrosis
activator ARDS
inhibitor-1 Asthma

SERPINB1 Neutrophil elastase, cathepsin G, proteinase-3 Protect the lung against elastase activity

SERPINB2 Urokinse-type plasminogen activator, tissue-type Protect against cell death


Plasminogen plasminogen activator
activator
inhibitor-2

SERPINB3 Cathepsins K, L, S, V Protection of cells against cytosolic lysosomal peptidases,


Inhibit cell death
SERPINB4 Cathepsin G, mast cell proteinase Protect against cell death
SERPINB6 Cathepsin G Protect cells from granule peptidases
SERPINB9 Granzyme B Protect cytotoxic lymphocytes/maintain granzyme B granules
SERPINB10 Trypsin, thrombin Protect against cell death
SERPINB12 Trypsin
SERPINB13 Cathepsin K, L Protect against cell death

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S129

a1AT deficiency: COPD and emphysema recruits an excess of migrating and activated neutrophils that
The proposed mechanism in the development of chronic release their serine peptidases to the cell surface or directly into the
obstructive pulmonary disease (COPD), including emphysema, is airway space. The elevated peptidase levels overwhelm the available
an imbalance of elastase and anti-elastase activity in the lung neutralizing activity of a1AT. CF progression is associated with
(reviewed in Elias et al.11). a1AT*ZZ and a1AT*SZ individuals are exacerbations, frequently caused by increased bacterial load or viral
at a greater risk of developing COPD. For a1AT*ZZ individuals, infection (reviewed in Goss and Burns23). Inflammation is central
plasma a1AT*Z levels of 15% translate into elastase-inhibitory to this event, including increases in interleukin (IL)-8, IL-6, IL-1b,
activities in the lung far below this. First, protein encoded by the tumor necrosis factor-a (TNFa), leukotriene B4 (LKTB4) and free
a1AT*Z allele exhibits an association rate with NE approximately NE. Exacerbations lead to airway remodeling and decreased lung
fivefold slower than normal a1AT. Second, a1AT*Z protein function.
maintains its inherent instability and continues to form polymers a1AT replacement therapy to combat NE activity in CF lungs
as it diffuses into the lung, leading to the detection of polymers in has been available for over two decades, however, there is little
lungs of a1AT*ZZ individuals, and further loss of anti-elastase statistical data derived from clinical trials relating to its
activity.12,13 Finally, it was observed that a1AT*ZZ polymers served effectiveness.24,25 Two recent papers described promising results
as a chemoattractant for human neutrophils.14 This translates into with inhaled a1AT in CF patients. Both studies found decreased
increased inflammation and NE peptidase in conjunction with inflammation associated with lowered cytokines and neutrophil cell
decreased a1AT function. numbers.26,27 Griese et al. compared peripheral lung versus the
COPD development is largely influenced by environmental bronchial deposition of aerosolized prolastin (a1AT purified from
factors, of which the most common is tobacco smoke. It has been plasma; Bayer Corporation, Clayton, NC, USA). Although no
proposed that the P1-Met residue in the RSL renders a1AT sensitive difference was observed between the two sites of treatment
to inactivation by oxidation from tobacco smoke exposure and the deposition, both groups receiving 4 weeks of daily prolastin
reactive oxygen burst released by neutrophils (reviewed in exhibited significant decreases in IL-8, TNFa, IL-1b protein and
Carrell10). Accordingly, a1AT*ZZ individuals who smoke exhibit a LKTB4 concentrations in sputum. In a second study, 4 weeks of
rapid onset of emphysema, often by 30 years of age, and death by treatment with recombinant a1AT (ra1AT) made in sheep
the age of 50.15 However, most a1AT*ZZ individuals who avoid produced significant decreases in neutrophil infiltration, and
smoking live normal length lives with minimal complications. reduced complexes between NE and endogenous a1AT, suggesting
Replacement therapy for a1AT deficiency, with intravenous a1AT that ra1AT was effective in neutralizing endogenous NE.27
from pooled human plasma, results in longer survival. Data Favorable results have also been collected from nebulized a1AT
compiled by the National Heart, Lung and Blood Institute indicate therapy studies in animal models.28 Together these results
that a1AT serum levels and lung function are improved with a1AT suggested that a1AT aerosol treatment would benefit CF patients.
replacement therapy.16 However, plasma is limited and therapy However, it will be important to examine in detail the mechanism
expensive. Gene therapy is an alternative toward which much by which a1AT inhibited inflammation and cytokine production.
progress been made in animal models.17,18 Synthetic small
molecule peptidase inhibitors are also being developed as a tool to
a1AT and lung disease in the newborn
counteract imbalances in peptidase activity (reviewed in Chughtai
and O’Riordan19). On the basis of nature of a1AT deficit caused by The premature infants lacking surfactant synthesis develop RDS.
polymerization, another therapeutic strategy is to prevent A direct result of underdeveloped lungs and respiratory system,
polymerization and facilitate a1AT secretion from the liver. This therapy includes oxygen and mechanical ventilation, and
may be accomplished using small molecules interacting with a1AT surfactant replacement. Even with improved oxygen saturation
directly or by enhanced chaperone function.20–22 monitoring and ventilation, unavoidable acute lung injury (ALI)
adds to the severity of RDS. Infants born prematurely frequently go
on to develop BPD. Prematurity is the primary risk factor for BPD,
followed by oxygen toxicity and ventilation induced lung damage
Cystic fibrosis and a1AT (reviewed in Jobe and Ikegami29 and Chess et al.30). The ‘new’
Cystic fibrosis (CF) presents another scenario where lung pathology BPD pathology consists of arrested alveolar development, resulting
and disease progression is associated with increased elastase in decreased total alveoli, and abnormal vasculature localization
activity. However, unlike a1AT deficiency, CF patients express observed in the lung periphery. Inflammatory cytokines have been
normal amounts of a1AT. Decreased fluidity of mucus in the CF associated with accelerated lung maturation.29 Serine peptidases,
airway impairs mucociliary clearance, obstructs normal diffusion released from inflammatory cells, caused epithelial cell injury and
of innate immune components, as well as a1AT, and creates lung remodeling that is associated with RDS and BPD.31 In the
localized environments bacteria may colonize. Chronic infection neonate the balance between elastase and elastase inhibitor activity

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S130

has been associated with lung injury and progression to BPD.32 In macrophages from patients with ARDS46 and IPF.45 An
addition to a1AT, low molecular mass inhibitor secretory leukocyte investigation of PAI-1, uPA and tPA levels in preterm infants with
peptidase inhibitor (SLPI) was found to be important in RDS also identified elevated ratios of PAI-1 to uPA protein in
neutralizing NE in tracheal aspirates of infants born prematurely.33 tracheal aspirate fluid.47 Elevated PAI-1 in BAL fluid occurred in
Watterberg et al.34 found that SLPI increases in the tracheal lavage bacterial pneumonia.48,49 Excessive PAI-1 levels were observed in
of RDS, whereas neonates going on to develop BPD had a ARDS and severe pneumonia cases requiring mechanical
significantly higher elastase to elastase-inhibitor ratio. Sveger ventilation.50 These findings supported the hypothesis that PAI-1 is
et al.35 also reported significant correlations with BPD development a critical regulator of fibrin deposition in the alveolar space in
and low levels of inhibitors a1AT and ACT (SERPINA3) in response to ALI. Further support for this theory was gained from
tracheobronchial aspirate of preterm infants at 3 to 4 days of age, animal models of lung injury, including bacterial endotoxin,51 and
and low SLPI at 7 to 8 days of age. Two trials assessed the effect of bleomycin52 models of ALI. These studies established PAI-1, its
a1AT therapy in preterm infants on recovery from RDS and activity and its regulation, as a critical target for therapies treating
development of BPD.36,37 A meta-analysis of the two trials found the family of diseases displaying poor lung function associated with
that although a trend for reduced risk of oxygen dependency after fibrin deposition including ALI, ARDS and COPD.
28 days existed, no significant difference was observed between PAI-1 has been identified as a potential mediator of host
groups receiving a1AT therapy and placebo for risk of BPD or defense. In mouse lung infection models, PAI-1 deficiency did not
long-term neurodevelopmental abnormalities.38 affect the outcome of infection by Gram-positive Streptococcus
Similar to endotoxin exposure, meconium aspiration leads to a pneumonia,53 but was found to be critical in defense against
rapid induction of cytokines, inflammation, decreased surfactant Gram-negative Klebsialla pneumonia.54 PAI-1-deficient animals
function, hypoxemia, pulmonary hypertension and excessive cell exhibited increased mortality at 24 and 48 h, coinciding with
death in the airway of the newborn. In animal models, all of these increased bacterial dissemination, and elevated fibrinolysis. These
events take place within 2 to 4 h of exposure to dilute human results may be attributed to the ascribed function of PAI-1 in
meconium.39–42 Following the initial inflammatory response, MAS regulating fibrinolysis or a potential role of PAI-1 in mediating
patients frequently undergo further pulmonary distress associated neutrophil recruitment signals, as was demonstrated in vitro.55 In
with oxygen toxicity upon intubation and mechanical ventilation. either case, these results suggest that PAI-1 function may prove to
Zagariya et al.43 hypothesized that a1AT in the lungs of neonates be a useful alternative target in the battle against bacterial
may attenuate meconium aspiration-induced lung injury. As has pathogens.
been observed in a1AT deficiency and CF patients, the absence of
adequate anti-elastase activity associated with a1AT resulted in
extensive pulmonary damage. Treatment with supplemental a1AT
activity would seem to be a promising approach to arrest Antithrombin III
elastase-dependent lung damage following meconium aspiration. Antithrombin III (SERPINC1, ATIII) regulates coagulation by
inhibiting activated serine peptidases. ATIII can inhibit serine
peptidases thrombin, factors IXa, Xa, XIa, XIIa and kallikrein
Plasminogen activator inhibitor-1 (Table 1).56 The peptidase inhibitor activity of ATIII is positively
Plasminogen activator inhibitor-1 (PAI-1, SERPINE1) is an regulated by its cofactor heparin binding to the D helix. This
inhibitor of the two plasminogen activators (PAs), urokinase-type induces a conformational switch putting the RSL in a more
plasminogen activator (uPA) and tissue-type plasminogen activator favorable position for serine peptidase binding.57 Heparin
(tPA). uPA is expressed in specific tissues, whereas tPA functions as molecules longer than 26 residues further accelerate peptidase
a soluble protein in the vascular system. The balance between PA inhibition through interactions with the peptidase. Defects in ATIII
and PAI-1 activities determines local fibrinolysis (Table 1). Diseases function primarily result in complications relating to thrombosis.
of acute inflammation, such as ALI and acute respiratory distress However, a protective role of ATIII has been experimentally
(ARDS) (reviewed in Ware and Matthay44), and fibrotic diseases demonstrated in animal models of sepsis58 and ischemia
like idiopathic pulmonary fibrosis (IPF)45 share a common reperfusion of grafted lungs.59 Recombinant ATIII (rATIII) at
pathology of fibrin deposition in the alveolar compartment. To elevated levels resulted in reduced to complete inhibition of
determine the best approach for treatment and prevention of these pulmonary vascular permeability associated with endotoxin
related diseases, it is important to identify the source of imbalanced treatment.58 In a lung transplant model, lungs were stored for 28 h
fibrinolysis resulting in fibrin deposition. As the main inhibitor of with normal saline, then transplanted into dogs receiving ATIII or
fibrinolysis, PAI-1 is expressed by several cell types found in the vehicle alone. Transplant animals treated with ATIII exhibited no
lung and is modulated by inflammatory cytokines and tissue change in O2 partial pressure, alveolar–arterial O2 difference or
damage. Elevated levels of PAI-1 were observed in alveolar pulmonary vascular resistance for up to 3 h.59 These encouraging

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S131

results suggested that ATIII might be used to treat sepsis-related human APC in vivo. Nishi et al.74 used the hPCI Tg mouse to
lung dysfunction, ARDS and ALI resulting from ischemia. identify a role for PCI in pulmonary hypertension. Monocrotaline
In humans, plasma ATIII levels normally decline with sepsis treatment was used to specifically induce pulmonary hypertension.
severity, and correlate with high mortality rate.60 Small clinical A significant increase in right ventricular pressure was observed in
trials suggested improved survival rates in patients receiving treated wild-type (WT) control mice compared to hPCI Tg animals.
rATIII.61–64 Eisele et al.62 reported that rATIII therapy was BAL fluid levels of thrombin–antithrombin complex, monocyte
associated with decreased lung dysfunction. However, Waydas chemoattractant protein-1, platelet-derived growth factor and IL-13,
et al.63 found no difference in the duration of organ failure. and the plasma level of TNFa were significantly increased in
A single large trial of 2314 patients with severe sepsis produced treated WT mice compared to hPCI Tg animals.74 This study
inconsistent results.65 One group in this trial, receiving ATIII but established that PCI in the lung is protective against
not heparin, exhibited an increased 90-day survival rate compared monocrotaline-induce hypertension. Furthermore, it suggested that
with placebo. It was also observed that new pulmonary dysfunction PCI fulfills both anti-inflammatory and anticoagulant activities in
was decreased in the group receiving rATIII therapy. Unfortunately, the lung.74,75 To take advantage of PCI therapeutically it will be
no critical trials have been performed to date specifically with ALI important to determine which activities as a serine peptidase
or ARDS patients to test ATIII or heparin therapy. Overall, inhibitor are protective in specific physiological conditions.
improvement in fibrin deposition and lung function observed with Clinical studies on the role of PCI are limited. Examination of
ATIII in animal models of sepsis and ALI is promising but is 58 patients with interstitial lung disease (ILD) associated with
unconfirmed in humans. diverse underlying pathologies discovered elevated PCI in the BAL
fluid of each of them.76 Groups with cryptogenic-organizing
pneumonia, collagen vascular disease (CVD-ILD) and sarcoidosis
Peptidase C inhibitor exhibited elevated levels of PCI and thrombin-activatable
Peptidase C inhibitor (PCI, SERPINA5) has a broad peptidase fibrinolysis inhibitor (TAFI), supporting the hypothesis that PCI
inhibitor profile through which it modulates both the coagulation inhibition of APC results in elevated TAFI levels.77
and fibrinolysis systems (Table 1) (reviewed in Church et al.56 and
Geiger66). PCI in plasma is the major inhibitor of the
anticoagulant peptidase activated protein C (APC). PCI also inhibits The clade B serpins protect cells with an intracellular
the thrombin–thrombomodulin complex,67 kallikrein, factors Xa, antipeptidase shield
XIa and thrombin. PCI may downregulate fibrinolysis by inhibiting In humans there are 13 clade B genes that encode serine and
plasmin activator peptidases uPA and tPA. Like ATIII, the cysteine peptidase inhibitors. The intracellular serpins are expressed
interaction between PCI and many of its target peptidases is in a wide range of tissues including lung, and target a wide range
modulated by heparin and other glycosaminoglycans.68–70 The of peptidases (Table 1). In the face of environmental insults such
interaction between PCI and target peptidase kallikrein is inhibited as bacterial and viral infection, and excessive peptidase levels
by glycosaminoglycans. In humans, PCI is a plasma protein, associated with inflammation, the induction of cell death is a
present at a concentration of B100 nM and a half-life of B23 h.71 common yet critical step in ALI. There is increasing evidence that
PCI is found in many other body fluids and secretions, and in a intracellular serpins function as a cytoprotective antipeptidase
wide range of tissues. The broad peptidase-inhibitory profile and shield, limiting damage by the misdirected peptidases as well as the
widely distributed expression has made it difficult to assign specific induction of necrosis and apoptosis (reviewed in Silverman et al.78
biological functions to this serpin. and Scott79). SERPINB1 inhibits elastases expressed by the
To determine the role of this serpin, PCI function was studied in neutrophil.80 SERPINB2, B3, B4, B10 and B13 have been
the mouse. Expression of mouse PCI was limited to the male and implicated in blocking proapoptotic signals.81,82 SERPINB6 and B9
female reproductive systems.72 Homozygous-null PCI-knockout inhibit peptidases stored in the cytolytic granules including catG
mouse appeared normal except male sterility was observed.72 and granzyme B (GzmB), respectively.83,84 SERPINB12 inhibits
Limited endogenous PCI in the mouse allowed a unique approach trypsin and is expressed in the lung.85 Together, the clade B serpin
to model human PCI pulmonary function. Hayashi et al.73 genes encode proteins with diverse antipeptidase activity, with
expressed hPCI in mice from a transgene consisting of the human critical intracellular roles in protecting cells from damage, and
PCI gene contained within 25 kb of human genomic DNA. hPCI maintaining normal function in the lung.
protein in these transgenic (Tg) animals was found to be an active
inhibitor of APC, and expressed in a pattern similar to humans.
This hPCI Tg animal system may be used as a tool to further SERPINB1 inhibits elastases in CF and BPD
explore PCI function in the lung under physiological and SERPINB1 (monocyte/neutrophil elastase inhibitor) is an inhibitor
pathological conditions, as well as to test the therapeutic effect of of NE, catG and neutrophil peptidase-3 (pr-3) (Table 1).80,86

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S132

SERPINB1 protein was shown to exist at elevated levels and in a succumb to necrosis or apoptosis following lysosomal damage and
complex with NE in the lavage of CF patients compared to normal release of its cysteine peptidases into the cytosol (reviewed in
individuals.87 Recombinant human SERPINB1 (rSERPINB1) was Lockshin and Zakeri101 and Guicciardi et al.102). Cysteine
able to protect rat lungs against injury, including hemorrhage and peptidases are likely to be involved in development of BPD, as catK,
epithelial permeability, from the instillation of NE or CF patient catL, and catS were shown to be elevated in a baboon model of
sputum preparations.88 rSERPINB1 was shown to inhibit BPD.103 SERPINB3 is predicted to provide protection to cells against
Surfactant-A degradation by the peptidase(s) in BAL fluid from CF cysteine peptidases. Maintenance of BEC by SERPINB3 and B4
patients.89 Yasumatsu et al. probed SERPINB1 function in the would limit ALI and prevent epithelial permeability during
established baboon BPD model.90,91 As in humans, SERPINB1 was infection.
localized to bronchial and glandular epithelial cells, as well as
mast cells, neutrophils and macrophages in the baboon lung.92
SERPINB1 protein in baboon lung tissue was found in high SERPINB9 inhibits granzyme B
molecular weight complexes with both NE and catG in BPD models SERPINB9 is the only known inhibitor of GzmB in humans
but not gestational controls.90 These results suggested that (Table 1).84 SERPINB9 is expressed in cytotoxic lymphocytes
SERPINB1 function in the newborn lung is critical as an (CTLs), dendritic cells (DCs) and NK cells of the monocyte and
antipeptidase shield against elastases associated with inflammation lymphocyte lineages.104,105 Using a Serpinb9-deficient mouse
and responsible for lung injury. model, Zhang et al.106 demonstrated that Serpinb9 is required to
protect CTLs from ‘accidental death’ induced by residual GzmB in
the cytosol. Serpinb9 was found to be required for the maintenance
Plasminogen activator inhibitor 2 blocks apoptosis of GzmB-containing granule integrity in CTLs. Finally, in the
Plasminogen activator inhibitor 2 (SERPINB2, PAI-2) inhibits uPA absence of Serpinb9, animals exhibited impaired clearance of
and tPA, and is expressed in macrophages and monocytes lymphocytic choriomeningitis virus.106 On the basis of these
(Table 1). Its primarily intracellular and nuclear localization findings and the localization of SERPINB9-positive monocytes and
suggests, however, that PAI-2 has functions in addition to DCs in lung tissue,107 it is proposed that this serpin is required for
regulating fibrinolysis (reviewed in Medcalf and Stasinopoulos93). normal host defense in the lung, particularly against bacterial and
Several studies have demonstrated the ability of intracellular PAI-2 viral pathogens requiring cell contact-mediated killing by the
to inhibit apoptosis. TNFa-induced apoptosis was inhibited by immune system.
ectopic PAI-2 expression in HeLa and fibrosarcoma cells.94,95
Recently, PAI-2 was found to be required for macrophage survival
following pathogen activation of the toll-like receptor-4 apoptosis Summary and future directions
pathway.96 On the basis of these studies, PAI-2 appears to be a Lung function is dependent on several peptidase driven systems
critical regulator of cell survival in cells of the host defense system including the innate and adaptive immune systems, coagulation,
where it is expressed. fibrinolysis and tissue remodeling. As a result, pulmonary diseases
are commonly linked to excessive peptidase activity. Serpins are the
major regulators of peptidase activity in the lung. It follows that
SERPINB3 and SERPINB4 are serine and cysteine understanding the biological function and regulation of serpins
peptidase inhibitors will be critical in the development of therapies against pulmonary
SERPINB3 and B4 are co-expressed in lung epithelium.97 disease.
SERPINB3 inhibits the lysosomal cysteine peptidases, cathepsin L Serpins occupy two niches in the lung, regulation of
(catL), catK and catS.98 SERPINB4 inhibits the serine peptidases extracellular peptidases and intracellular peptidases. The blood
catG and mast cell chymase (Table 1).99 Ectopic SERPINB3 plasma serpins a1AT, PAI-1, ATIII and PCI, function
expression was cytoprotective against TNFa and natural killer (NK) extracellularly. Their localization within the lung makes them
cell-induced apoptosis,81 and both SERPINB3 and B4 have been primary targets for the development of therapeutic agents, from
shown independently to protect cells against radiation.82 The simple replacement by inhalation, to recombinant gene transfer
peptidase inhibitor activity of SERPINB3 and B4 may provide and development of small molecule inhibitors that mimic their
cellular protection in the environment of the lung during ALI and activity. In the case of a1AT, this serpin inhibits a limited number
inflammation. SERPINB3 and B4 expression was induced in BECs of elastolytic peptidases. In diseases associated with excess elastase
in asthmatics and by IL-4 and IL-13.100 SERPINB3 and B4 may activity (empysema, CF), replacement therapy appears very
provide protection against neutrophil and mast cell peptidases as promising. In contrast, ATIII and PCI are multi-peptidase
well as localized bursts of reactive oxygen generated by neutrophils inhibitors and their local active concentration in vivo is likely
used to destroy pathogens. Cells exposed to reactive oxygen critical to properly balancing the function of specific peptidase

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S133

targets. To modulate the activity of these serpins therapeutically, we 10 Carrell RW. alpha 1-Antitrypsin: molecular pathology, leukocytes, and tissue damage.
may take advantage of the endogenous mechanisms regulating J Clin Invest 1986; 78: 1427–1431.
11 Elias JA, Kang MJ, Crouthers K, Homer R, Lee CG. State of the art. mechanistic
their activity, interactions with glycosaminoglycans and
heterogeneity in chronic obstructive pulmonary disease: insights from transgenic mice.
conformational changes in structure. Alternatively, extensive Proc Am Thorac Soc 2006; 3: 494–498.
success has been met using recombinant APC, a PCI target 12 Elliott PR, Bilton D, Lomas DA. Lung polymers in Z alpha1-antitrypsin deficiency-
peptidase, in the treatment of sepsis and ALI. related emphysema. Am J Respir Cell Mol Biol 1998; 18: 670–675.
The clade B serpins are mainly intracellular. They are predicted 13 Mahadeva R, Atkinson C, Li Z, Stewart S, Janciauskiene S, Kelley DG et al. Polymers of
to form an anti-peptidase shield, protecting cells against exogenous Z alpha1-antitrypsin co-localize with neutrophils in emphysematous alveoli and are
chemotactic in vivo. Am J Pathol 2005; 166: 377–386.
and endogenous peptidase activity. This hypothesis is based upon
14 Parmar JS, Mahadeva R, Reed BJ, Farahi N, Cadwallader KA, Keogan MT et al.
their intracellular localization and broad tissue distribution, Polymers of alpha(1)-antitrypsin are chemotactic for human neutrophils: a new
combined with the varied peptidase-inhibitory specificities of the paradigm for the pathogenesis of emphysema. Am J Respir Cell Mol Biol 2002; 26:
clade B members. In their absence, cellular injury would lead to 723–730.
cellular stress and death. SERPINB9, the only inhibitor of GzmB in 15 Hodges JR, Millward-Sadler GH, Barbatis C, Wright R. Heterozygous MZ alpha 1-
antitrypsin deficiency in adults with chronic active hepatitis and cryptogenic cirrhosis.
humans, is required for CTL survival, and therefore host defense
N Engl J Med 1981; 304: 557–560.
against some viral infections. SERPINB3 and SERPINB4, expressed 16 National Heart, LaBI. A registry of patients with severe deficiency of alpha1-antitrypsin.
in BEC, are poised to protect the bronchial airways against both Chest 1994; 1223–1232.
exogenous and endogenous serine and lysosomal cysteine 17 Lu Y, Choi YK, Campbell-Thompson M, Li C, Tang Q, Crawford JM et al. Therapeutic
peptidases. In lung epithelium, this antipeptidase shield is very level of functional human alpha 1 antitrypsin (hAAT) secreted from murine
important due to repeated exposure to neutrophil-derived serine muscle transduced by adeno-associated virus (rAAV1) vector. J Gene Med 2006; 8:
730–735.
peptidases and oxidative stress accompanying inflammation in
18 Flotte TR, Conlon TJ, Poirier A, Campbell-Thompson M, Byrne BJ. Preclinical
response to pulmonary injury or infection. On the basis of the characterization of a recombinant adeno-associated virus type 1-pseudotyped vector
ability to block cellular damage, including that caused by demonstrates dose-dependent injection site inflammation and dissemination of vector
lysosomal cysteine peptidases, and cell death, the intracellular genomes to distant sites. Hum Gene Ther 2007; 18: 245–256.
clade B serpins are powerful agents to target in the development of 19 Chughtai B, O’Riordan TG. Potential role of inhibitors of neutrophil elastase in treating
diseases of the airway. J Aerosol Med 2004; 17: 289–298.
therapeutics.
20 Schmidt BZ, Perlmutter DH. Grp78, Grp94, and Grp170 interact with alpha1-antitrypsin
mutants that are retained in the endoplasmic reticulum. Am J Physiol Gastrointest
Liver Physiol 2005; 289: G444–G455.
21 Burrows JA, Willis LK, Perlmutter DH. Chemical chaperones mediate increased secretion
Disclosure
of mutant alpha 1-antitrypsin (alpha 1-AT) Z: a potential pharmacological strategy for
The authors have declared no financial interests.
prevention of liver injury and emphysema in alpha 1-AT deficiency. Proc Natl Acad Sci
USA 2000; 97: 1796–1801.
22 Teckman JH. Lack of effect of oral 4-phenylbutyrate on serum alpha-1-antitrypsin in
patients with alpha-1-antitrypsin deficiency: a preliminary study. J Pediatr
References Gastroenterol Nutr 2004; 39: 34–37.
1 Gettins PGW. Serpin structure, mechanism, and function. Chem Rev. 102; 2002; 23 Goss CH, Burns JL. Exacerbations in cystic fibrosis. 1: epidemiology and pathogenesis.
4751–4804. [see comment]. Thorax 2007; 62: 360–367.
2 Gettins PGW. Mechanisms of serpin inhibition. In: Silverman GA, Lomas DA (eds). 24 Brennan S. Revisiting alpha1-antitrypsin therapy in cystic fibrosis: can it still offer
Molecular and Cellular Aspects of the Serpinopathies and Disorders in Serpin promise? [comment]. Eur Respir J 2007; 29: 229–230.
Activity. World Scientific Publishing: Tuck Link, Singapore, 2007, pp 67–100. 25 McElvaney NG, Hubbard RC, Birrer P, Chernick MS, Caplan DB, Frank MM et al.
3 Huntington JA, Read RJ, Carrell RW. Structure of a serpin–protease complex shows Aerosol alpha 1-antitrypsin treatment for cystic fibrosis. Lancet 1991; 337: 392–394.
inhibition by deformation. Nature 2000; 407: 923–926. 26 Griese M, Latzin P, Kappler M, Weckerle K, Heinzlmaier T, Bernhardt T et al.
4 Lomas DA, Evans DL, Finch JT, Carrell RW. The mechanism of Z alpha1-antitrypsan alpha1-Antitrypsin inhalation reduces airway inflammation in cystic fibrosis patients.
accumulation in the liver. Nature 1992; 357: 605–607. [see comment]. Eur Respir J 2007; 29: 240–250.
5 Lomas D. The serpinopathies and respiratory disease. In: Silverman GA, Lomas DA 27 Martin SL, Downey D, Bilton D, Keogan MT, Edgar J, Elborn JS et al. Safety and
(eds). Molecular and Cellular Aspects of the Serpinopathies and Disorders in Serpin efficacy of recombinant alpha(1)-antitrypsin therapy in cystic fibrosis. Pediatr
Activity. World Scientific Publishing: Tuck Link, Singapore, 2007, p 639. Pulmonol 2006; 41: 177–183.
6 Carrell RW, Lomas DA. Alpha1-antitrypsin deficiencyFa model for conformational 28 Cantin AM, Woods DE. Aerosolized prolastin suppresses bacterial proliferation in a
diseases. N Engl J Med 2002; 346: 45–53. model of chronic Pseudomonas aeruginosa lung infection. Am J Respir Crit Care
7 Eriksson S, Carlson J, Velez R. Risk of cirrhosis and primary liver cancer in Med 1999; 160: 1130–1135.
alpha 1-antitrypsin deficiency. N Engl J Med 1986; 324: 736–739. 29 Jobe AH, Ikegami M. Prevention of bronchopulmonary dysplasia. Curr Opin Pediatr
8 Eden E, Mitchell D, Mehlman B, Khouli H, Nejat M, Grieco MH et al. Atopy, asthma, 2001; 13: 124–129.
and emphysema in patients with severe alpha-1-antitrypysin deficiency. Am J Respir 30 Chess PR, D’Angio CT, Pryhuber GS, Maniscalco WM. Pathogenesis of broncho-
Crit Care Med 1997; 156: 68–74. pulmonary dysplasia. Semin Perinatol 2006; 30: 171–178.
9 Travis J, Salvesen GS. Human plasma proteinase inhibitors. Annu Rev Biochem 1983; 31 Merritt TA, Cochrane CG, Holcomb K, Bohl B, Hallman M, Strayer D et al. Elastase and
52: 655–709. alpha 1-proteinase inhibitor activity in tracheal aspirates during respiratory distress

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S134

syndrome. Role of inflammation in the pathogenesis of bronchopulmonary dysplasia. 52 Wygrecka M, Markart P, Ruppert C, Petri K, Preissner KT, Seeger W et al. Cellular
J Clin Invest 1983; 72: 656–666. origin of procoagulant and (anti)-fibrinolytic factors in bleomycin-injured lungs. Eur
32 Speer CP. Inflammation and bronchopulmonary dysplasia. Semin Neonatol 2003; Respir J 2007; 29(6): 1105–1114.
8: 29–38. 53 Rijneveld AW, Florquin S, Bresser P, Levi M, De Waard V, Lijnen R et al. Plasminogen
33 Sluis KB, Darlow BA, Vissers MC, Winterbourn CC. Proteinase–antiproteinase balance activator inhibitor type-1 deficiency does not influence the outcome of murine
in tracheal aspirates from neonates. Eur Respir J 1994; 7: 251–259. pneumococcal pneumonia. Blood 2003; 102: 934–939.
34 Watterberg KL, Carmichael DF, Gerdes JS, Werner S, Backstrom C, Murphy S. Secretory 54 Renckens R, Roelofs JJ, Bonta PI, Florquin S, de Vries CJ, Levi M et al. Plasminogen
leukocyte protease inhibitor and lung inflammation in developing bronchopulmonary activator inhibitor type 1 is protective during severe Gram-negative pneumonia. Blood
dysplasia. J Pediatr 1994; 125: 264–269. 2007; 109: 1593–1601.
35 Sveger T, Ohlsson K, Polberger S, Noack G, Morse H, Laurin S. Tracheobronchial 55 Kwak SH, Wang XQ, He Q, Fang WF, Mitra S, Bdeir K et al. Plasminogen activator
aspirate fluid neutrophil lipocalin, elastase- and neutrophil protease-4-alpha1- inhibitor-1 potentiates LPS-induced neutrophil activation through a JNK-mediated
antitrypsin complexes, protease inhibitors and free proteolytic activity in respiratory pathway. Thromb Haemost 2006; 95: 829–835.
distress syndrome. Acta Paediatr 2002; 91: 934–937. 56 Church F, Pike RN, Tollefsen DM, Buckle AM, Ciaccia AV, Olson ST. Regulation of
36 Dunn MS, Stiskal JA, O’Brien KK, Ito S, Cox DW, Kelly EN. alpha1-Proteinase inhibitor hemostasis by heparin-binding serpins. In: Silverman GA, Lomas DA (eds). Molecular
(A1PI) therapy for the prevention of chronic lung disease (CLD) of prematurityFa and Cellular Aspects of the Serpinopathies and Disorders in Serpin Activity. World
dose ranging study and meta-analysis with previous randomized clinical trial (RCT) Scientific Publishing: Tuck Link, Singapore, 2007, pp 509–554.
(Abstract). Pediatr Res 2000; 47: 397A. 57 Quinsey NS, Greedy AL, Bottomley SP, Whisstock JC, Pike RN. Antithrombin: in control
37 Stiskal JA, Dunn MS, Shennan AT, O’Brien KK, Kelly EN, Koppel RI et al. of coagulation. Int J Biochem Cell Biol 2004; 36: 386–389.
alpha1-Proteinase inhibitor therapy for the prevention of chronic lung disease of 58 Uchiba M, Okajima K, Murakami K. Effects of various doses of antithrombin III on
prematurity: a randomized, controlled trial. Pediatrics 1998; 101: 89–94. endotoxin-induced endothelial cell injury and coagulation abnormalities in rats.
38 Shah P, Ohlsson A. Alpha-1 proteinase inhibitor (a1PI) for preventing chronic lung Thromb Res 1998; 89: 233–241.
disease in preterm infants. Cochrane Database Syst Rev 2001 CD002775. 59 Salvatierra A, Guerrero R, Rodriguez M, Alvarez A, Soriano F, Lopez-Pedrera R et al.
39 Zagariya A, Bhat R, Uhal B, Navale S, Freidine M, Vidyasagar D. Cell death and lung cell Antithrombin III prevents early pulmonary dysfunction after lung transplantation in
histology in meconium aspirated newborn rabbit lung. Eur J Pediatr 2000; 159: 819–826. the dog. Circulation 2001; 104: 2975–2980.
40 Zagariya A, Bhat R, Chari G, Uhal B, Navale S, Vidyasagar D. Apoptosis of airway 60 Fourrier F, Chopin C, Goudemand J, Hendrycx S, Caron C, Rime A et al. Septic shock,
epithelial cells in response to meconium. Life Sci 2005; 76: 1849–1858. multiple organ failure, and disseminated intravascular coagulation. Compared patterns
41 Holopainen R, Aho H, Laine J, Peuravuori H, Soukka H, Kaapa P. Human meconium of antithrombin III, protein C, and protein S deficiencies. [see comment]. Chest 1992;
has high phospholipase A2 activity and induces cellular injury and apoptosis in piglet 101: 816–823.
lungs. Pediatr Res 1999; 46: 626–632. 61 Fourrier F, Jallot A, Leclerc L, Jourdain M, Racadot A, Chagnon JL et al. Sex steroid
42 Holopainen R, Soukka H, Halkola L, Kaapa P. Meconium aspiration induces a hormones in circulatory shock, sepsis syndrome, and septic shock. Circ Shock 1994;
concentration-dependent pulmonary hypertensive response in newborn piglets. Pediatr 43: 171–178.
Pulmonol 1998; 25: 107–113. 62 Eisele B, Lamy M, Thijs LG, Keinecke HO, Schuster HP, Matthias FR et al.
43 Zagariya AM, Bhat R, Zhabotynsky E, Chari G, Navale S, Xu Q et al. Characterization of Antithrombin III in patients with severe sepsis. A randomized, placebo-controlled,
serine/cysteine protease inhibitor alpha1-antitrypsin from meconium-instilled rabbit double-blind multicenter trial plus a meta-analysis on all randomized, placebo-
lungs. J Cell Biochem 2005; 96: 137–144. controlled, double-blind trials with antithrombin III in severe sepsis. [see comment].
44 Ware LB, Matthay MA. Clinical practice. Acute pulmonary edema. N Engl J Med 2005; Intensive Care Med 1998; 24: 663–672.
353: 2788–2796. 63 Waydhas C, Nast-Kolb D, Gippner-Steppert C, Trupka A, Pfundstein C, Schweiberer L
45 Kotani I, Sato A, Hayakawa H, Urano T, Takada Y, Takada A. Increased procoagulant et al. High-dose antithrombin III treatment of severely injured patients: results of a
and antifibrinolytic activities in the lungs with idiopathic pulmonary fibrosis. Thromb prospective study. J Trauma 1998; 45: 931–940.
Res 1995; 77: 493–504. 64 Baudo F, Caimi TM, de Cataldo F, Ravizza A, Arlati S, Casella G et al. Antithrombin III
46 Chapman HA, Yang XL, Sailor LZ, Sugarbaker DJ. Developmental expression of (ATIII) replacement therapy in patients with sepsis and/or postsurgical complications:
plasminogen activator inhibitor type 1 by human alveolar macrophages. Possible role a controlled double-blind, randomized, multicenter study. [see comment]. Intensive
in lung injury. J Immunol 1990; 145: 3398–3405. Care Med 1998; 24: 336–342.
47 Cederqvist K, Siren V, Petaja J, Vaheri A, Haglund C, Andersson S. High concentrations 65 Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I et al. Caring for the critically ill
of plasminogen activator inhibitor-1 in lungs of preterm infants with respiratory patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial.
distress syndrome. Pediatrics 2006; 117: 1226–1234. [see comment][erratum appears in JAMA 2002 Jan 9;287(2):192]. JAMA 2001; 286:
48 Choi G, Schultz MJ, van Till JW, Bresser P, van der Zee JS, Boermeester MA et al. 1869–1878.
Disturbed alveolar fibrin turnover during pneumonia is restricted to the site of 66 Geiger M. Protein C inhibitor, a serpin with functions in- and outside vascular biology.
infection. Eur Respir J 2004; 24: 786–789. Thromb Haemost 2007; 97: 343–347.
49 El-Solh AA, Okada M, Pietrantoni C, Aquilina A, Berbary E. Procoagulant and 67 Rezaie AR, Cooper ST, Church FC, Esmon CT. Protein C inhibitor is a potent inhibitor
fibrinolytic activity in ventilator-associated pneumonia: impact of inadequate of the thrombin–thrombomodulin complex. J Biol Chem 1995; 270: 25336–25339.
antimicrobial therapy. Intensive Care Med 2004; 30: 1914–1920. 68 Pratt CW, Church FC. Heparin binding to protein C inhibitor. J Biol Chem 1992; 267:
50 Gunther A, Mosavi P, Heinemann S, Ruppert C, Muth H, Markart P et al. Alveolar 8789–8794.
fibrin formation caused by enhanced procoagulant and depressed fibrinolytic capacities 69 Ecke S, Geiger M, Binder BR. Glycosaminoglycans regulate the enzyme specificity of
in severe pneumonia. Comparison with the acute respiratory distress syndrome. Am J protein C inhibitor. Ann NY Acad Sci 1992; 667: 84–86.
Respir Crit Care Med 2000; 161: 454–462. 70 Pike RN, Buckle AM, le Bonniec BF, Church FC. Control of the coagulation system by
51 Wygrecka M, Markart P, Ruppert C, Kuchenbuch T, Fink L, Bohle RM et al. serpins. Getting by with a little help from glycosaminoglycans. FEBS J 2005; 272:
Compartment- and cell-specific expression of coagulation and fibrinolysis factors in the 4842–4851.
murine lung undergoing inhalational versus intravenous endotoxin application. 71 Laurell M, Stenflo J, Carlson TH. Turnover of *I-protein C inhibitor and *I-alpha 1-
[see comment]. Thromb Haemost 2004; 92: 529–540. antitrypsin and their complexes with activated protein C. Blood 1990; 76: 2290–2295.

Journal of Perinatology
Serpins in pulmonary disease
DJ Askew and GA Silverman
S135

72 Uhrin P, Dewerchin M, Hilpert M, Chrenek P, Schofer C, Zechmeister-Machhart M 89 Rubio F, Cooley J, Accurso FJ, Remold-O’Donnell E. Linkage of neutrophil serine
et al. Disruption of the protein C inhibitor gene results in impaired spermatogenesis proteases and decreased surfactant protein-A (SP-A) levels in inflammatory lung
and male infertility. J Clin Invest 2000; 106: 1531–1539. disease. Thorax 2004; 59: 318–323.
73 Hayashi T, Nishioka J, Kamada H, Asanuma K, Kondo H, Gabazza EC et al. 90 Yasumatsu R, Altiok O, Benarafa C, Yasumatsu C, Bingol-Karakoc G, Remold-
Characterization of a novel human protein C inhibitor (PCI) gene transgenic mouse O’Donnell E et al. SERPINB1 upregulation is associated with in vivo complex
useful for studying the role of PCI in physiological and pathological conditions. formation with neutrophil elastase and cathepsin G in a baboon model of
J Thromb Haemost 2004; 2: 949–961. bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2006; 291:
74 Nishii Y, Gabazza EC, Fujimoto H, Nakahara H, Takagi T, Bruno N et al. Protective L619–L627.
role of protein C inhibitor in monocrotaline-induced pulmonary hypertension. [see 91 Coalson JJ, Winter VT, Siler-Khodr T, Yoder BA. Neonatal chronic lung disease in
comment]. J Thromb Haemost 2006; 4: 2331–2339. extremely immature baboons. Am J Respir Crit Care Med 1999; 160: 1333–1346.
75 Beaulieu LM, Church FC. Is protein C inhibitor antithrombotic and protective in 92 Zeng W, Remold-O’Donnell E. Human monocyte/neutrophil elastase inhibitor (MNEI)
pulmonary hypertension? [comment]. J Thromb Haemost 2006; 4: 2327–2330. is regulated by PU.1/Spi-1, Sp1, and NF-kappaB. J Cell Biochem 2000; 78: 519–532.
76 Fujimoto H, Gabazza EC, Hataji O, Yuda H, D’Alessandro-Gabazza CN, Nakano M et al. 93 Medcalf RL, Stasinopoulos SJ. The undecided serpin. The ins and outs of plasminogen
Thrombin-activatable fibrinolysis inhibitor and protein C inhibitor in interstitial lung activator inhibitor type 2. FEBS J 2005; 272: 4858–4867.
disease. [see comment]. Am J Respir Crit Care Med 2003; 167: 1687–1694. 94 Kumar S, Baglioni C. Protection from tumor necrosis factor-mediated cytolysis by
77 Mosnier LO, Elisen MG, Bouma BN, Meijers JC. Protein C inhibitor regulates the overexpression of plasminogen activator inhibitor type-2. J Biol Chem 1991; 266:
thrombin–thrombomodulin complex in the up- and down regulation of TAFI 20960–20964.
activation. Thromb Haemost 2001; 86: 1057–1064. 95 Dickinson JL, Bates EJ, Ferrante A, Antalis TM. Plasminogen activator inhibitor type 2
78 Silverman GA, Whisstock JC, Askew DJ, Pak SC, Luke CJ, Cataltepe S et al. Human clade inhibits tumor necrosis factor alpha-induced apoptosis. Evidence for an alternate
B serpins (ov-serpins) belong to a cohort of evolutionarily dispersed intracellular biological function. J Biol Chem 1995; 270: 27894–27904.
proteinase inhibitor clades that protect cells from promiscuous proteolysis. Cell Mol Life 96 Park JM, Greten FR, Wong A, Westrick RJ, Arthur JS, Otsu K et al. Signaling pathways
Sci 2004; 61: 301–325. and genes that inhibit pathogen-induced macrophage apoptosisFCREB and
79 Scott FL. Serpins, apoptosis and other aspects of cell death. In: Silverman, GA, NF-kappaB as key regulators. Immunity 2005; 23: 319–329.
Lomas DA (eds). Molecular and Cellular Aspects of the Serpinopathies and 97 Cataltepe S, Gornstein ER, Schick C, Kamachi Y, Chatson K, Fries J et al. Co-expression
Disorders in Serpin Activity. World Scientific Publishing: Tuck Link, Singapore, 2007, of the squamous cell carcinoma antigens 1 and 2 in normal adult human tissues and
pp 301–336. squamous cell carcinomas. J Histochem Cytochem 2000; 48: 113–122.
80 Remold-O’Donnell E. A fast-acting elastase inhibitor in human monocytes. J Exp Med 98 Schick C, Pemberton PA, Shi GP, Kamachi Y, Cataltepe S, Bartuski AJ et al. Cross-class
1985; 162: 2142–2155. inhibition of the cysteine proteinases cathepsins K, L, and S by the serpin squamous
81 Suminami Y, Nagashima S, Vujanovic NL, Hirabayashi K, Kato H, Whiteside TL. cell carcinoma antigen 1: a kinetic analysis. Biochemistry 1998; 37: 5258–5266.
Inhibition of apoptosis in human tumour cells by the tumour-associated serpin, SCC 99 Schick C, Kamachi Y, Bartuski AJ, Cataltepe S, Schechter NM, Pemberton PA et al.
antigen-1. Br J Cancer 2000; 82: 981–989. Squamous cell carcinoma antigen 2 is a novel serpin that inhibits the chymotrypsin-
82 Murakami A, Suminami Y, Hirakawa H, Nawata S, Numa F, Kato H. Squamous cell like proteinases cathepsin G and mast cell chymase. J Biol Chem 1997; 272: 1849–
carcinoma antigen suppresses radiation-induced cell death. Br J Cancer 2001; 84: 1855.
851–858. 100 Yuyama N, Davies DE, Akaiwa M, Matsui K, Hamasaki Y, Suminami Y et al. Analysis
83 Scott FL, Hirst CE, Sun J, Bird CH, Bottomley SP, Bird PI. The intracellular serpin of novel disease-related genes in bronchial asthma. Cytokine 2002; 19: 287–296.
proteinase inhibitor 6 is expressed in monocytes and granulocytes and is a potent 101 Lockshin RA, Zakeri Z. Caspase-independent cell deaths. Curr Opin Cell Biol 2002;
inhibitor of the azurophilic granule protease, cathepsin G. Blood 1999; 93: 14: 727–733.
2089–2097. 102 Guicciardi ME, Leist M, Gores GJ. Lysosomes in cell death. Oncogene 2004; 23:
84 Sun J, Bird CH, Sutton V, McDonald L, Coughlin PB, De Jong TA et al. A cytosolic 2881–2890.
granzyme B inhibitor related to the viral apoptotic regulator cytokine response 103 Altiok O, Yasumatsu R, Bingol-Karakoc G, Riese RJ, Stahlman MT, Dwyer W et al.
modifier A is present in cytotoxic lymphocytes. J Biol Chem 1996; 271: Imbalance between cysteine proteases and inhibitors in a baboon model of
27802–27809. bronchopulmonary dysplasia. Am J Respir Crit Care Med 2006; 173: 318–326.
85 Askew YS, Pak SC, Luke CJ, Askew DJ, Cataltepe S, Mills DR et al. SERPINB12 is a novel 104 Phillips T, Opferman JT, Shah R, Liu N, Froelich CJ, Ashton-Rickardt PG. A role for
member of the human ov-serpin family that is widely expressed and inhibits the granzyme B inhibitor serine protease inhibitor 6 in CD8+ memory cell
trypsin-like serine proteinases. J Biol Chem 2001; 276: 49320–49330. homeostasis. J Immunol 2004; 173: 3801–3809.
86 Cooley J, Takayama TK, Shapiro SD, Schechter NM, Remold-O’Donnell E. The serpin 105 Medema JP, Schuurhuis DH, Rea D, van Tongeren J, de Jong J, Bres SA et al.
MNEI inhibits elastase-like and chymotrypsin-like serine proteases through efficient Expression of the serpin serine protease inhibitor 6 protects dendritic cells from
reactions at two active sites. Biochemistry 2001; 40: 15762–15770. cytotoxic T lymphocyte-induced apoptosis: differential modulation by T helper type 1
87 Cooley JRF, Sontag MK, Osberg I, Accurso FJ, Remold-O’Donnell E. MNEI (monocyte/ and type 2 cells. [see comment]. J Exp Med 2001; 194: 657–667.
neutrophil elastase inhibitor) is found at increased levels in cystic fibrosis lavage fluid 106 Zhang M, Park SM, Wang Y, Shah R, Liu N, Murmann AE et al. Serine protease
(Abstract). Pediatr Pulmonol 2001; 227. inhibitor 6 protects cytotoxic T cells from self-inflicted injury by ensuring the integrity
88 Rees DD, Rogers RA, Cooley J, Mandle RJ, Kenney DM, Remold-O’Donnell E. of cytotoxic granules. Immunity 2006; 24: 451–461.
Recombinant human monocyte/neutrophil elastase inhibitor protects rat lungs against 107 Bladergroen BA, Strik MC, Bovenschen N, van Berkum O, Scheffer GL, Meijer CJ et al.
injury from cystic fibrosis airway secretions. Am J Respir Cell Mol Biol 1999; 20: The granzyme B inhibitor, protease inhibitor 9, is mainly expressed by dendritic cells
69–78. and at immune-privileged sites. J Immunol 2001; 166: 3218–3225.

Journal of Perinatology
‘Substance abuse
treatment linked with
prenatal visits improves
prenatal outcomes:
a new standard’
Journal of Perinatology presents
this retrospective cohort study
online FREE of charge. This article
evaluates the impact of Early Start,
an obstetric clinic-based prenatal
substance abuse treatment
program, for pregnant women and
their newborns.

An ‘Early Start’ may mean a healthy


beginning for both mother and child.

www.nature.com/jp

20518-04 JP hot article AJFP.indd 1 6/8/08 13:45:19


Having trouble reaching
your target audience?
Take advantage of the highly targeted advertising opportunities VOLUME 16 | NO. 12 | JANUARY 2008
www.obesityjournal.org

available in this journal. a research journal

For more information, contact:

Alf Anderson | Advertising Manager


Nature Publishing Group
Complexity of inter-organ

25 First Street, Suite 104, Cambridge, MA 02141 communication

T 617.475.9231 F 617.494.4960 E media@nature.com OBESITY_COVER.indd 1


nature publishing group

7/31/07 3:11:57 PM

www.nature.com/obesity

13400-05OBYAdvertise_RJFP4C.indd 1 29/11/07 18:21:50


Cutting-edge · High-impact · Ground-breaking

The official journal of The Obesity Society

Submit your research to Obesity today.


Obesity offers high-quality original research and presents new information in the areas of pharmacology,
nutrition medicine, genetics, adipogenesis, behavioral epidemiology, biophysics and lipid metabolism,
exercise and human physiology, nutritional epidemiology, phenotyping, fat cell physiology, aging,
neuroscience, transgenic models, metabolic syndrome, nutrition behavior, pediatric obesity, adipocyte cell
biology, and surgical interventions.

Publishing your paper in Obesity provides a wide range of benefits, including:

• Online submission – Fast and secure submission with Manuscript Central


• Timely review of articles
• Advance Online Publication (AOP) – Articles are available online in final format before they appear
in print and are immediately citable with their digital online identifier number (DOI number).
• Supplementary online material – Include additional supporting material, essential to your published
paper, online (e.g. multi-media presentations, tables).
• NPG’s ‘license to publish’ – NPG does not require authors to release copyright of their
articles. Instead, authors provide an exclusive license; you are free to reuse your paper VOLUME 16 | NO. 12 | JANUARY 2008
www.obesityjournal.org

in any of your future published work and to post a pdf version on your own website. a research journal

• Wide Exposure – Wide online dissemination of content via institutional access


and worldwide distribution of content, including free access, to institutions in
developing nations through HINARI and SciDevNet.

…and much more.


Complexity of inter-organ
communication

Visit the journal’s website today at www.nature.com/obesity for more information. OBESITY_COVER.indd 1
nature publishing group

7/31/07 3:11:57 PM

www.nature.com/obesity

13399-05OBYSubmit_RJFP4C.indd 1 6/12/07 17:32:10

You might also like